US20110183988A1 - Compounds and methods for kinase modulation, and indications therefor - Google Patents

Compounds and methods for kinase modulation, and indications therefor Download PDF

Info

Publication number
US20110183988A1
US20110183988A1 US12/975,239 US97523910A US2011183988A1 US 20110183988 A1 US20110183988 A1 US 20110183988A1 US 97523910 A US97523910 A US 97523910A US 2011183988 A1 US2011183988 A1 US 2011183988A1
Authority
US
United States
Prior art keywords
difluoro
phenyl
pyrrolo
pyridine
carbonyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/975,239
Other languages
English (en)
Inventor
Prabha N. Ibrahim
Guoxian Wu
Jack Lin
Wayne Spevak
Hanna Cho
Todd Ewing
Chao Zhang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Plexxikon Inc
Original Assignee
Ibrahim Prabha N
Guoxian Wu
Jack Lin
Wayne Spevak
Hanna Cho
Todd Ewing
Chao Zhang
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ibrahim Prabha N, Guoxian Wu, Jack Lin, Wayne Spevak, Hanna Cho, Todd Ewing, Chao Zhang filed Critical Ibrahim Prabha N
Priority to US12/975,239 priority Critical patent/US20110183988A1/en
Priority to TW099145373A priority patent/TW201132639A/zh
Publication of US20110183988A1 publication Critical patent/US20110183988A1/en
Assigned to PLEXXIKON INC. reassignment PLEXXIKON INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHO, HANNA, IBRAHIM, PRABHA N., EWING, TODD, SPEVAK, WAYNE, LIN, JACK, WU, GUOXIAN, ZHANG, CHAO
Priority to US14/514,232 priority patent/US9440969B2/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/4965Non-condensed pyrazines
    • A61K31/497Non-condensed pyrazines containing further 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
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • 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
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further 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
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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

Definitions

  • disclosed compounds are kinase inhibitors.
  • compounds are provided, as well as various salts thereof, formulations thereof, conjugates thereof, derivatives thereof, forms thereof and uses thereof.
  • compounds are any one of Formulae I-XX as described below.
  • the compounds inhibit one or more Raf protein kinases, including one or more of A-Raf, B-Raf, and c-Raf-1, and any mutations thereof.
  • the compounds inhibit each of c-Raf-1, B-Raf, and B-Raf V600X protein kinase (where X is an amino acid other than valine, e.g., alanine, arginine, aspartic acid, glycine, lysine or methionine).
  • the compounds inhibit a B-Raf V600E mutant protein kinase.
  • the compounds inhibit each of c-Raf-1, B-Raf, B-Raf V600X, and B-Raf V600E protein kinase.
  • the compounds inhibit each of cRaf-1, B-Raf, and B-Raf V600E protein kinase.
  • Also contemplated in accordance with the present invention are methods for the use of the compounds in treating diseases and conditions associated with regulation of the activity of one or more Raf protein kinases, including one or more of A-Raf, B-Raf, and c-Raf-1, and any mutations thereof.
  • the use of compounds for therapeutic methods involving modulation of protein kinases are provided.
  • the compounds inhibit the activity on one or more Raf kinases, including A-Raf, B-Raf and/or c-Raf-1, including any mutations thereof.
  • the compounds are used for therapeutic methods involving modulation of one or more Raf protein kinases, including treatment of a variety of indications, including, but not limited to, melanoma, glioma, glioblastoma multiforme, pilocytic astrocytoma, colorectal cancer, thyroid cancer, lung cancer, ovarian cancer, prostate cancer, liver cancer, gallbladder cancer, gastrointestinal stromal tumors, biliary tract cancer, cholangiocarcinoma, acute pain, chronic pain and polycystic kidney disease.
  • a variety of indications including, but not limited to, melanoma, glioma, glioblastoma multiforme, pilocytic astrocytoma, colorectal cancer, thyroid cancer, lung cancer, ovarian cancer, prostate cancer, liver cancer, gallbladder cancer, gastrointestinal stromal tumors, biliary tract cancer, cholangiocarcinoma, acute pain, chronic pain and polycystic
  • the compounds are used for therapeutic methods involving modulation of B-Raf V600E mutant protein kinase, including treatment of a variety of indications, including, but not limited to, melanoma, glioma, glioblastoma multiforme, pilocytic astrocytoma, colorectal cancer, thyroid cancer, lung cancer, ovarian cancer, prostate cancer, liver cancer, gallbladder cancer, gastrointestinal stromal tumors, biliary tract cancer, and cholangiocarcinoma.
  • the compounds are used for therapeutic methods involving modulation of c-Raf-1 protein kinase, including treatment of a variety of indications, including, but not limited to, acute pain, chronic pain and polycystic kidney disease.
  • Ar is bicyclic heteroaryl optionally independently substituted with one or more R 2 (wherein R 2 is one or more of R 24 , R 25 , R 26 , R 27 , R 28 , R 29 , R 30 , R 31 , R 32 , R 33 , R 34 , R 35 , R 36 , R 37 , R 38 , R 39 , R 40 , R 41 , R 42 , R 43 and R 44 , as appropriate).
  • R 2 is one or more of R 24 , R 25 , R 26 , R 27 , R 28 , R 29 , R 30 , R 31 , R 32 , R 33 , R 34 , R 35 , R 36 , R 37 , R 38 , R 39 , R 40 , R 41 , R 42 , R 43 and R 44 , as appropriate).
  • Ar is selected from Group A as follows:
  • Ar is any one of the bicyclic heteroaryl moieties set forth above in Group A, and R 1 is n-propyl.
  • Ar is any one of the bicyclic heteroaryl moieties set forth above in Group A, and R 1 is 4-trifluoromethyl-phenyl.
  • Ar is any one of the bicyclic heteroaryl moieties set forth above in Group A, and R 1 is 2-fluoro-phenyl.
  • Ar is any one of the bicyclic heteroaryl moieties set forth above in Group A, and R 1 is 3-fluoro-phenyl.
  • Ar is any one of the bicyclic heteroaryl moieties set forth above in Group A, and R 1 is 2,5-difluoro-phenyl.
  • Ar is selected from the group consisting of thiophenyl optionally independently substituted with one or more R 3 , thiazolyl optionally independently substituted with one or more R 4 , oxazolyl optionally independently substituted with one or more R 5 , pyrazolyl optionally independently substituted with one or more R 6 , pyridyl optionally independently substituted with one or more R 7 , pyrimidinyl optionally independently substituted with one or more R 8 , phenyl substituted with one or more R 9 , and pyrazinyl optionally independently substituted with one or more R 10 .
  • Ar is selected from the group consisting of:
  • R 1 is n-propyl
  • R 1 is 4-trifluoromethyl-phenyl.
  • R 1 is 2-fluoro-phenyl, 3-fluoro-phenyl or 2,5-difluoro-phenyl.
  • Ar is not 2-methoxy-pyrimidin-5-yl.
  • R 1 is 2-fluoro-phenyl
  • R 1 is 3-fluoro-phenyl.
  • R 1 is 2,5-difluoro-phenyl.
  • Ar is thiophenyl, providing compounds of Formula II as follows:
  • each R 3 is independently —CN, —C(O)—R 11 , —S(O) 2 —R 12 , fluoro, chloro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, phenyl, or heteroaryl, wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl, lower alkoxy, or fluoro substituted lower alkoxy.
  • R 1 is as defined in any of the embodiments above.
  • Ar is thiophenyl, providing compounds of Formula III as follows:
  • Ar is thiazolyl, providing compounds of Formula IV as follows:
  • each R 4 is independently —CN, —OH, —NH 2 , —C(O)—R 11 , —S(O) 2 —R 12 , fluoro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, phenyl, or heteroaryl, wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl, lower alkoxy, or fluoro substituted lower alkoxy.
  • R 1 is as defined in any of the embodiments above.
  • Ar is thiazolyl, providing compounds of Formula V as follows:
  • each R 4 is independently —C(O)—R 11 , —S(O) 2 —R 12 , fluoro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, phenyl, or heteroaryl, wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl, lower alkoxy, or fluoro substituted lower alkoxy.
  • R 1 is as defined in any of the embodiments above.
  • Ar is thiazolyl, providing compounds of Formula VI as follows:
  • each R 4 is independently —C(O)—R 11 , —S(O) 2 —R 12 , fluoro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy; fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, phenyl, or heteroaryl, wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl, lower alkoxy, or fluoro substituted lower alkoxy.
  • R 1 is as defined in any of the embodiments above.
  • Ar is oxazolyl, providing compounds of Formula VII as follows:
  • each R 5 is independently —C(O)—R 11 , —S(O) 2 —R 12 , fluoro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, phenyl, or heteroaryl, wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl, lower alkoxy, or fluoro substituted lower alkoxy.
  • R 1 is as defined in any of the embodiments above.
  • Ar is oxazolyl, providing compounds of Formula VIII as follows:
  • each R 5 is independently —C(O)—R 11 , —S(O) 2 —R 12 , fluoro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, phenyl, or heteroaryl, wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl, lower alkoxy, or fluoro substituted lower alkoxy.
  • R 1 is as defined in any of the embodiments above.
  • Ar is oxazolyl, providing compounds of Formula IX as follows:
  • each R 5 is independently —C(O)—R 11 , —S(O) 2 -R 12 , fluoro, lower alkyl, fluoro substituted lower alkyl, lower alkoxy, fluoro substituted lower alkoxy, mono-alkylamino, di-alkylamino, cycloalkyl, heterocycloalkyl, phenyl, or heteroaryl, wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl, lower alkoxy, or fluoro substituted lower alkoxy.
  • R 1 is as defined in any of the embodiments above.
  • Ar is pyrazolyl, providing compounds of Formula X as follows:
  • each R 6 is independently fluoro or lower alkyl, wherein lower alkyl is optionally independently substituted with one or more fluoro, lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino.
  • R 1 is as defined in any of the embodiments above.
  • Ar is pyrazolyl, providing compounds of Formula XI as follows:
  • R 59 , R 60 and R 61 are independently selected from hydrogen or the R 6 substituents.
  • each R 6 is independently fluoro or lower alkyl, wherein lower alkyl is optionally independently substituted with one or more fluoro, lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino.
  • R 1 is as defined in any of the embodiments above.
  • R 1 is not ethyl, n-propyl, cyclopropyl, n-butyl, isobutyl, p-trifluoromethylphenyl, 3-fluorophenyl, 3-difluoromethoxyphenyl, p-n-propylphenyl or p-isopropylphenyl.
  • Ar is pyrazolyl, providing compounds of Formula XII as follows:
  • R 59 , R 60 and R 62 are independently selected from hydrogen or the R 6 substituents.
  • each R 6 is independently fluoro or lower alkyl, wherein lower alkyl is optionally independently substituted with one or more fluoro, lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino.
  • R 1 is as defined in any of the embodiments above.
  • Ar is pyridyl, providing compounds of Formula XIII as follows:
  • each R 7 is independently —C(O)—R 11 , —S(O) 2 —R 12 , —C(O)—N(H)—O—R 16 , —O—R 13 , —N(R 14 )—R 15 , fluoro, chloro, lower alkyl, cycloalkyl, heterocycloalkyl,phenyl, or heteroaryl, wherein lower alkyl is optionally independently substituted with one or more fluoro, lower alkoxy, mono-alkylamino, di-alkylamino, cycloalkylamino, or heterocycloalkylamino, and wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl
  • Ar is pyridyl, providing compounds of Formula XIV as follows:
  • each R 7 is independently —C(O)—R 11 , —S(O) 2 —R 12 , —C(O)—N(H)—O—R 16 , —N(R 14 )—R 15 , fluoro, chloro, lower alkyl, cycloalkyl, heterocycloalkyl, phenyl, or heteroaryl, wherein lower alkyl is optionally independently substituted with one or more fluoro, lower alkoxy, mono-alkylamino, di-alkylamino, cycloalkylamino, or heterocycloalkylamino, and wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl, lower alkoxy, or
  • Ar is pyridyl, providing compounds of Formula XV as follows:
  • each R 7 is independently —C(O)—R 11 , —S(O) 2 —R 12 , —C(O)—N(H)—O—R 12 , —O—R 13 , —N(R 14 )—R 15 , fluoro, chloro, lower alkyl, cycloalkyl, heterocycloalkyl, phenyl, or heteroaryl, wherein lower alkyl is optionally independently substituted with one or more fluoro, lower alkoxy, mono-alkylamino, di-alkylamino, cycloalkylamino, or heterocycloalkylamino, and wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alky
  • Ar is pyrimidinyl, providing compounds of Formula XVI as follows:
  • each R 8 is independently —C(O)—R 11 , —S(O) 2 —R 12 , —C(O)—N(H)—O—R 16 , —N(R 14 )—R 15 , fluoro, lower alkyl, cycloalkyl, heterocycloalkyl, phenyl, or heteroaryl, wherein lower alkyl is optionally independently substituted with one or more fluoro, lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino, and wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl, lower alkoxy, or fluoro substituted lower alkoxy.
  • R 1 is as defined in
  • Ar is pyrimidinyl, providing compounds of Formula XVII as follows:
  • each R 8 is independently —C(O)—R 11 , —S(O) 2 —R 12 , —C(O)—N(H)—O—R 16 , —O—R 13 , —N(R 14 )—R 15 , fluoro, lower alkyl, cycloalkyl, heterocycloalkyl, phenyl, or heteroaryl, wherein lower alkyl is optionally independently substituted with one or more fluoro, lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino, and wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl, lower alkoxy, or fluoro substituted lower alkyl
  • Ar is pyrimidinyl
  • each R 8 is independently —C(O)—R 11 , —S(O) 2 —R 12 , —C(O)—N(H)—O—R 16 , —O—R 13 , —N(R 14 )—R 15 , fluoro, lower alkyl, cycloalkyl, heterocycloalkyl, phenyl, or heteroaryl, wherein lower alkyl is optionally independently substituted with one or more fluoro, lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino, and wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl, lower alkoxy, or fluoro substituted lower alkyl
  • Ar is phenyl, providing compounds of Formula XIX as follows:
  • each R 9 is independently —C(O)—R 11 , —S(O) 2 —R 12 , —C(O)—N(H)—O—R 16 , —O—R 13 , —N(R 14 )—R 15 , fluoro, lower alkyl, cycloalkyl, heterocycloalkyl, phenyl, heteroaryl, wherein lower alkyl is optionally independently substituted with one or more fluoro, lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino, and wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl, lower alkoxy, or fluoro substituted lower alkoxy
  • Ar is pyrazinyl, providing compounds of Formula XX as follows:
  • each R 10 is independently —C(O)—R 11 , —S(O) 2 —R 12 , —C(O)—N(H)—O—R 16 , —O—R 13 , —N(R 14 )—R 15 , fluoro, lower alkyl, cycloalkyl, heterocycloalkyl, phenyl, or heteroaryl, wherein lower alkyl is optionally independently substituted with one or more fluoro, lower alkoxy, mono-alkylamino, di-alkylamino, or cycloalkylamino, and wherein cycloalkyl, heterocycloalkyl, phenyl and heteroaryl are optionally independently substituted with one or more lower alkyl, fluoro substituted lower alkyl, lower alkoxy, or fluoro substituted lower alkoxy
  • R 24 and R 26 are each independently H, methyl, ethyl, propyl, methoxycarbonyl, ethylsulfonyl, methylcarbamoyl, acetyl or methylsufonyl.
  • X is at the 4-position of the 6-membered benzene ring. In another embodiment, X is at the 5-position of the 6-membered benzene ring. In another embodiment, X is at the 6-position of the 6-membered benzene ring. In another embodiment, X is at the 7-position of the 6-membered benzene ring. All the other variables are as defined in any of the above embodiments.
  • R 32 and R 34 are each independently H, methyl, ethyl, propyl, methoxycarbonyl, ethylsulfonyl, methylcarbamoyl, acetyl or methylsufonyl.
  • X is at the 4-position of the 6-membered benzene ring. In another embodiment, X is at the 5-position of the 6-membered benzene ring. In another embodiment, X is at the 6-position of the 6-membered benzene ring. In another embodiment, X is at the 7-position of the 6-membered benzene ring. All the other variables are as defined in any of the above embodiments.
  • the subscript p is 0.
  • X is at the 4-position of the 6-membered benzene ring. In another embodiment, X is at the 5-position of the 6-membered benzene ring. In another embodiment, X is at the 6-position of the 6-membered benzene ring. In another embodiment, X is at the 7-position of the 6-membered benzene ring. All the other variables are as defined in any of the above embodiments.
  • the subscript p is 0.
  • W is at the 5-position of the 6-membered benzene ring.
  • W is at the 6-position of the 6-membered benzene ring.
  • W is at the 7-position of the 6-membered benzene ring.
  • W is at the 8-position of the 6-membered benzene ring.
  • Ar is quinoxalinyl
  • W is at the 5 or 6-position of the 6-membered benzene ring. All the other variables are as defined in any of the above embodiments.
  • the subscript p is 0.
  • y can be at any of the positions on the bicylic ring.
  • Y is at the 3-position of the 5-membered imidazole ring.
  • X is at the 3-position of the 5-membered imidazole ring. All the other variables are as defined in any of the above embodiments.
  • the subscript p is 0.
  • R 36 is H, methyl, ethyl, propyl, methoxycarbonyl, ethylsulfonyl, methylcarbamoyl, acetyl or methylsufonyl.
  • X is at the 2-position of the 5-membered imidazole ring.
  • X is at the 4-position of the 6-membered benzene ring.
  • X is at the 5-position of the 6-membered benzene ring.
  • X is at the 6-position of the 6-membered benzene ring.
  • X is at the 7-position of the 6-membered benzene ring. All the other variables are as defined in any of the above embodiments.
  • R 70 and R 72 are each independently H, methyl, ethyl, propyl, methoxycarbonyl, ethylsulfonyl, methylcarbamoyl, acetyl or methylsufonyl.
  • X is at the 4-position of the 6-membered benzene ring. In another embodiment, X is at the 5-position of the 6-membered benzene ring. In another embodiment, X is at the 6-position of the 6-membered benzene ring. In another embodiment, X is at the 7-position of the 6-membered benzene ring. All the other variables are as defined in any of the above embodiments.
  • Ar is 2-thiazolyl, 4-thiazolyl or 5-thiazolyl, each of which is optionally substituted with from 1 to 2 R 4 .
  • Ar is 1-pyrazolyl, 3-pyrazolyl, 4-pyrazolyl or 5-pyrazolyl, each of which is optionally substituted with from 1 to 3 R 6 .
  • Ar is 2-thiophenyl or 3-thiophenyl, each of which is optionally substituted with from 1 to 3 R 3 .
  • Ar is 2-pyridyl, 3-pyridyl or 4-pyridyl, each of which is optionally substituted with from 1 to 3 R 7 .
  • Ar is 2-pyrimidinyl, 3-pyrimidinyl or 4-pyrimidinyl each of which is optionally substituted with from 1 to 3 R 8 .
  • Ar is 2-pyrazinyl, optionally substituted with from 1 to 2 R 10 .
  • Ar is 2-oxazolyl, 4-oxazolyl or 5-oxazolyl, each of which is optionally substituted with from 1 to 2 R 5 .
  • Ar is phenyl, optionally substituted with from 1 to 3 R 9 .
  • the substituent R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 or R 10 is selected from —CN, —OH, —NH 2 , —NO 2 , —C(O)—R 11 , —S(O) 2 —R 12 , optionally substituted lower alkyl, optionally substituted lower alkoxy, optionally substituted alkylamino, optionally substituted phenyl or optionally substituted heteroaryl. All the other variables are as defined in any of the above embodiments.
  • R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 or R 10 is selected from 4-piperidinyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, methyl, ethyl, propyl, isopropyl, 1,2,3,4-tetrazol-1-yl, 1,2,3,4-tetrazol-3-yl, 1,2,3,4-tetrazol-5-yl, 4-pyrazolylsulfonyl, 3-pyrazolylsulfonyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrazinyl, carbamoyl, alkylcarbamoyl, alkoxycarbonyl, phenoxycarbonyl, 1-imidazolyl, 2-imidazolyl, 4-imidazolyl, 1-pyrazolyl, 3-pyrazolyl, 4-pyrazolyl, dialkyla
  • Ar is 1H-4-indazolyl, 1H-5-indazolyl, 1H-6-indazolyl, 1H-7-indazolyl, 1,3-benzothiazol-4yl, 1,3-benzothiazol-5yl, 1,3-benzothiazol-6yl, 1,3-benzothiazol-7yl, imidazo[1,2-a]pyridine-2-yl, imidazo[1,2-a]pyridine-3-yl, imidazo[1,2-a]pyridine-5-yl, imidazo[1,2-a]pyridine-6-yl, imidazo[1,2-a]pyridine-7-yl, imidazo[1,2-a]pyridine-8-yl, imidazo[1,2-a]pyrimidin-5-yl, imidazo[1,2-a]pyrimidin-6-yl, imidazo[1,2-a]pyrimidin-7-yl, imidazo[1,2-a]pyrimidin-8-yl, 1
  • Ar is 1H-4-indazolyl, 1H-5-indazolyl, 1H-6-indazolyl, 1H-7-indazolyl, 1-methyl-4-indazolyl, 1-methyl-5-indazolyl, 1-methyl-6-indazolyl, 1-methyl-7-indazolyl, 1-acetyl-4-indazolyl, 1-acetyl-5-indazolyl, 1-acetyl-6-indazolyl, 1-acetyl-7-indazolyl, 1- methylsulfonyl-4-indazolyl, 1-methylsulfonyl-5-indazolyl, 1-methylsulfonyl-6-indazolyl, 1-methylsulfonyl-7-indazolyl, 1-methyl-3-amino-6-indazolyl, 1,3-benzothiazol-4yl, 1,3-benzothiazol-5yl, 1,3-benzothiazol-5yl, 1,3-benz
  • the compound is selected from the group consisting of:
  • the compound is selected from the group consisting of:
  • the compound is selected from the group consisting of:
  • the compound is selected from the group consisting of:
  • the compound is selected from the group consisting of:
  • the compound is selected from the group consisting of:
  • compounds are provided, wherein the compound is selected from the group consisting of:
  • the compound is selected from the group consisting of:
  • the compound is selected from the group consisting of:
  • the compound is selected from the group consisting of:
  • the compound is selected from the group consisting of:
  • the compound is selected from the group consisting of:
  • the compound is selected from the group consisting of:
  • the compound is selected from the group consisting of:
  • compounds are provided, wherein the compound is selected from the group consisting of:
  • the compound includes any prodrug thereof.
  • the compound includes any tautomer thereof.
  • the compound includes any stereoisomer thereof.
  • the compound includes any pharmaceutically acceptable formulation thereof.
  • the compound includes any conjugate thereof.
  • the compound includes any derivative thereof.
  • the compound includes any form thereof.
  • a compound or group of compounds includes salts of such compound(s) (including pharmaceutically acceptable salts), formulations of such compound(s) (including pharmaceutically acceptable formulations), conjugates thereof, derivatives thereof, forms thereof, prodrugs thereof, and all stereoisomers thereof.
  • a compound as described herein includes compounds of Formulae I-XX, including all sub-embodiments thereof, and compounds as listed in the second aspect above, including all sub-embodiments thereof.
  • methods for treating any Raf protein kinase mediated disease or condition in an animal subject in need thereof, wherein the method involves administering to the subject an effective amount of any one or more compound(s) as described herein. In certain embodiments, the method involves administering to the subject an effective amount of any one or more compound(s) as described herein in combination with one or more other therapies for the disease or condition.
  • methods for treating any B-Raf protein kinase mediated disease or condition, including any B-Raf mutant kinase mediated disease or condition in an animal subject in need thereof, wherein the method involves administering to the subject an effective amount of any one or more compound(s) as described herein. In certain embodiments, the method involves administering to the subject an effective amount of any one or more compound(s) as described herein in combination with one or more other therapies for the disease or condition.
  • methods are provided for treating any B-Raf V600E mutant protein kinase mediated disease or condition in an animal subject in need thereof, wherein the method involves administering to the subject an effective amount of any one or more compound(s) as described herein. In certain embodiments, the method involves administering to the subject an effective amount of any one or more compound(s) as described herein in combination with one or more other therapies for the disease or condition.
  • methods are provided for treating any c-Raf-1 protein kinase mediated disease or condition, including any c-Raf-1 mutant kinase mediated disease or condition in an animal subject in need thereof, wherein the method involves administering to the subject an effective amount of any one or more compound(s) as described herein. In certain embodiments, the method involves administering to the subject an effective amount of any one or more compound(s) as described herein in combination with one or more other therapies for the disease or condition.
  • a compound as described herein is a Raf kinase inhibitor and has an IC 50 of less than 500 nM, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM as determined in a generally accepted Raf kinase activity assay.
  • a compound as described herein will have an IC 50 of less than 500 nM, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM with respect to B-Raf, c-Raf-1, or B-Raf V600E mutant.
  • a compound as described herein will selectively inhibit one or more Raf kinases relative to one or more other non-Raf kinases.
  • a compound as described herein is a pan Raf inhibitor, i.e. inhibits each of B-Raf V600E mutant kinase, B-Raf kinase and c-Raf-1 kinase, with an IC 50 of less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM as determined in each of a generally accepted B-Raf V600E mutant kinase activity assay, B-Raf kinase activity assay, and c-Raf-1 kinase activity assay.
  • the compounds are approximately equipotent on each of B-Raf V600E, B-Raf and c-Raf-1, i.e. the ratio of IC 50 for any of B-Raf V600E, B-Raf and c-Raf-1 divided by the IC 50 for any other of B-Raf V600E, B-Raf and c-Raf-1 (e.g. B-Raf IC 50 divided by B-Raf V600E IC 50 ) is in the range of 10 to 0.1, also 5 to 0.2.
  • the compound is selective relative to other protein kinases, such that the ratio of IC 50 for another kinase assessed comparably, divided by the IC 50 for any of B-Raf V600E, B-Raf and c-Raf-1 is >10, also >20, also >30, also >40, also >50, also >60, also >70, also >80, also >90, also >100, wherein the other protein kinase includes, but is not limited to CSK, Insulin receptor kinase, AMPK, PDGFR or VEGFR.
  • the pan Raf inhibitor is a compound selected from the group consisting of:
  • compositions that include a therapeutically effective amount of any one or more compound(s) as described herein and at least one pharmaceutically acceptable carrier, excipient, and/or diluent, including combinations of any two or more compounds as described herein.
  • the composition can further include a plurality of different pharmacologically active compounds, which can include a plurality of compounds as described herein.
  • the composition can include any one or more compound(s) as described herein along with one or more compounds that are therapeutically effective for the same disease indication.
  • the composition includes any one or more compound(s) as described herein along with one or more compounds that are therapeutically effective for the same disease indication, wherein the compounds have a synergistic effect on the disease indication.
  • the composition includes any one or more compound(s) as described herein effective in treating a cancer and one or more other compounds that are effective in treating the same cancer, further wherein the compounds are synergistically effective in treating the cancer.
  • the invention provides methods for treating a disease or condition mediated by one or more Raf kinases (including A-Raf, B-Raf, and c-Raf-1 kinases), including mutations thereof, in a subject in need thereof by administering to the subject an effective amount of a composition including any one or more compound(s) as described herein.
  • the invention provides methods for treating a disease or condition mediated by one or more Raf kinases, including mutations thereof, by administering to the subject an effective amount of a composition including any one or more compound(s) as described herein in combination with one or more other suitable therapies for treating the disease.
  • the invention provides methods for treating a disease or condition mediated by B-Raf kinase, including any mutations thereof, in a subject in need thereof by administering to the subject an effective amount of a composition including any one or more compound(s) as described herein.
  • the invention provides methods for treating a disease or condition mediated by B-Raf kinase, including any mutations thereof, by administering to the subject an effective amount of a composition including any one or more compound(s) as described herein in combination with one or more other suitable therapies for treating the disease.
  • the invention provides methods for treating a disease or condition mediated by B-Raf V600E mutant kinase, in a subject in need thereof by administering to the subject an effective amount of a composition including any one or more compound(s) as described herein.
  • the invention provides methods for treating a disease or condition mediated by B-Raf V600E mutant kinase, by administering to the subject an effective amount of a composition including any one or more compound(s) as described herein in combination with one or more other suitable therapies for treating the disease.
  • the invention provides methods for treating a cancer mediated by B-Raf V600E mutant by administering to the subject an effective amount of a composition including any one or more compound(s) as described herein. In one embodiment, the invention provides methods for treating a cancer mediated by B-Raf V600E mutant by administering to the subject an effective amount of a composition including any one or more compound(s) as described herein in combination with one or more suitable anticancer therapies, such as one or more chemotherapeutic drugs.
  • the invention provides methods for treating a disease or condition mediated by c-Raf-1 kinase, including any mutations thereof, in a subject in need thereof by administering to the subject an effective amount of a composition including any one or more compound(s) as described herein.
  • the invention provides methods for treating a disease or condition mediated by c-Raf-1 kinase, including any mutations thereof, by administering to the subject an effective amount of a composition including any one or more compound(s) as described herein in combination with one or more other suitable therapies for treating the disease.
  • the invention provides a method of treating a cancer in a subject in need thereof by administering to the subject an effective amount of a composition including any one or more compound(s) as described herein.
  • the invention provides a method of treating a cancer in a subject in need thereof by administering to the subject an effective amount of a composition including any one or more compound(s) as described herein in combination with one or more other therapies or medical procedures effective in treating the cancer.
  • Other therapies or medical procedures include suitable anticancer therapy (e.g. drug therapy, vaccine therapy, gene therapy, photodynamic therapy) or medical procedure (e.g. surgery, radiation treatment, hyperthermia heating, bone marrow or stem cell transplant).
  • the one or more suitable anticancer therapies or medical procedures is selected from treatment with a chemotherapeutic agent (e.g. chemotherapeutic drug), radiation treatment (e.g. x-ray, ⁇ -ray, or electron, proton, neutron, or ⁇ particle beam), hyperthermia heating (e.g. microwave, ultrasound, radiofrequency ablation), Vaccine therapy (e.g. AFP gene hepatocellular carcinoma vaccine, AFP adenoviral vector vaccine, AG-858, allogeneic GM-CSF-secretion breast cancer vaccine, dendritic cell peptide vaccines), gene therapy (e.g.
  • a chemotherapeutic agent e.g. chemotherapeutic drug
  • radiation treatment e.g. x-ray, ⁇ -ray, or electron, proton, neutron, or ⁇ particle beam
  • hyperthermia heating e.g. microwave, ultrasound, radiofrequency ablation
  • Vaccine therapy e.g. AFP gene hepatocellular carcinoma vaccine,
  • Ad5CMV-p53 vector adenovector encoding MDA7, adenovirus 5-tumor necrosis factor alpha), photodynamic therapy (e.g. aminolevulinic acid, motexafin lutetium), surgery, or bone marrow and stem cell transplantation.
  • photodynamic therapy e.g. aminolevulinic acid, motexafin lutetium
  • the invention provides a method of treating a cancer in a subject in need thereof by administering to the subject an effective oxyit of a composition including any one or more compound(s) as described herein in combination with one or more suitable chemotherapeutic agents.
  • the one or more suitable chemotherapeutic agents is selected from an alkylating agent, including, but not limited to, adozelesin, altretamine, bendamustine, bizelesin, busulfan, carboplatin, carboquone, carmofur, carmustine, chlorambucil, cisplatin, cyclophosphamide, dacarbazine, estramustine, etoglucid, fotemustine, hepsulfam, ifosfamide, improsulfan, irofulven, lomustine, mannosulfan, mechlorethamine, melphalan, mitobronitol, nedaplatin, nimustine, oxaliplatin, piposulfan, prednimustine, procarbazine, ranimustine, satraplatin, semustine, streptozocin, temozolomide, thiotepa, tre
  • P13K inhibitors e.g. BEZ235, GDC-0941, XL147, XL765
  • Cdk4 inhibitors e.g. PD-332991
  • Akt inhibitors e.g. Hsp90 inhibitors
  • farnesyltransferase inhibitors e.g. tipifarnib
  • the method of treating a cancer involves administering to the subject an effective amount of a composition including any one or more compound(s) as described herein in combination with a chemotherapeutic agent selected from capecitabine, 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, vinblastine, bevacizumab, cetuximab, interferon-a, interleukin-2, or erlotinib.
  • a chemotherapeutic agent selected from capecitabine, 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, vinblastine, bevacizumab, cetuximab, interferon-a, interleukin-2, or erlotinib.
  • the invention provides a method of treating a disease or condition in a subject in need thereof, by administering to the subject a therapeutically effective amount of any one or more compound(s) as described herein, a prodrug of such compound, a pharmaceutically acceptable salt of such compound or prodrug, or a pharmaceutically acceptable formulation of such compound or prodrug.
  • the compound can be alone or can be part of a composition.
  • the invention provides a method of treating a disease or condition in a subject, by administering to the subject a therapeutically effective amount of any one or more compound(s) as described herein, a prodrug of such compound, a pharmaceutically acceptable salt of such compound or prodrug, or a pharmaceutically acceptable formulation of such compound or prodrug in combination with one or more other suitable therapies for the disease or condition.
  • kits that include a compound or composition thereof as described herein.
  • the compound or composition is packaged, e.g., in a vial, bottle, flask, which may be further packaged, e.g., within a box, envelope, or bag; the compound or composition is approved by the U.S.
  • the compound or composition is approved for administration to a mammal, e.g., a human, for a protein kinase mediated disease or condition;
  • the invention kit may include written instructions for use and/or other indication that the compound or composition is suitable or approved for administration to a mammal, e.g., a human, for a Raf protein kinase-mediated disease or condition; and the compound or composition may be packaged in unit dose or single dose form, e.g., single dose pills, capsules, or the like.
  • the invention provides methods for treating an A-Raf-mediated, B-Raf-mediated and/or c-Raf-1-mediated disease or condition in a subject in need thereof (e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats), e.g., a disease or condition characterized by abnormal A-Raf, B-Raf, and/or c-Raf-1 activity (e.g. kinase activity).
  • a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats
  • a disease or condition characterized by abnormal A-Raf, B-Raf, and/or c-Raf-1 activity e.g. kinase activity.
  • invention methods may involve administering to the subject suffering from or at risk of an A-Raf-mediated, B-Raf-mediated and/or c-Raf-1-mediated disease or condition an effective amount of any one or more Raf inhibitor(s) as described herein.
  • the A-Raf-mediated, B-Raf-mediated, and/or c-Raf-1-mediated disease is selected from the group consisting of neurologic diseases, including, but not limited to, multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease, seizures and epilepsy; neoplastic diseases including, but not limited to, melanoma, glioma, glioblastoma multiforme, pilocytic astrocytoma, sarcoma, carcinoma (e.g.
  • lymphoma e.g.
  • histiocytic lymphoma neurofibromatosis, gastrointestinal stromal tumors, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuroendocrine tumors such as medullary thyroid cancer, carcinoid, small cell lung cancer, Kaposi's sarcoma, and pheochromocytoma; pain of neuropathic or inflammatory origin, including, but not limited to, acute pain, chronic pain, cancer-related pain, and migraine; cardiovascular diseases including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g.
  • inflammation and/or proliferation including, but not limited to, psoriasis, eczema, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease (IBD); immunodeficiency diseases, including, but not limited to, organ transplant rejection, graft versus host disease, and Kaposi's sarcoma associated with HIV; renal, cystic, or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia, polycystic liver disease, tuberous sclerosis, Von Hippel Lindau disease, medullary cystic kidney disease, nephronophthisis, and cystic fibrosis; metabolic disorders, including, but not limited to, obesity
  • the disease or condition is selected from the group consisting of melanoma, glioma, glioblastoma multiforme, pilocytic astrocytoma, sarcoma, liver cancer, biliary tract cancer, cholangiocarcinoma, colorectal cancer, lung cancer, gallbladder cancer, breast cancer, pancreatic cancer, thyroid cancer, renal cancer, ovarian cancer, adrenocortical cancer, prostate cancer, histiocytic lymphoma, neurofibromatosis, gastrointestinal stromal tumors, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, medullary thyroid cancer, carcinoid, small cell lung cancer, Kaposi's sarcoma, pheochromocytoma, acute pain, chronic pain, and polycystic kidney disease.
  • the disease or condition is selected from the group consisting of melanoma, glioma, glioblastoma multiforme, pilocytic astrocytoma, colorectal cancer, thyroid cancer, lung cancer, ovarian cancer, prostate cancer, liver cancer, gallbladder cancer, gastrointestinal stromal tumors, biliary tract cancer, cholangiocarcinoma, acute pain, chronic pain, and polycystic kidney disease.
  • the invention provides methods for treating a c-Raf-1-mediated disease or condition in a subject in need thereof (e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats), e.g., a disease or condition characterized by abnormal c-Raf-1 activity (e.g. kinase activity).
  • a c-Raf-1-mediated disease is selected from the group consisting of polycystic kidney disease, acute pain, and chronic pain.
  • the invention provides methods for treating a B-Raf V600E mutant-mediated disease or condition in a subject in need thereof (e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats), e.g., a disease or condition characterized by abnormal B-Raf V600E mutant activity (e.g. kinase activity).
  • a subject in need thereof e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats
  • a disease or condition characterized by abnormal B-Raf V600E mutant activity e.g. kinase activity
  • the B-Raf V600E mutant-mediated disease is a cancer, preferably a cancer selected from the group consisting of melanoma, glioma, glioblastoma multiforme, pilocytic astrocytoma, colorectal cancer, thyroid cancer, lung cancer, ovarian cancer, prostate cancer, liver cancer, gallbladder cancer, gastrointestinal stromal tumors, biliary tract cancer, and cholangiocarcinoma.
  • the invention provides methods for treating a cancer in a subject in need thereof (e.g. a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats).
  • a mammal such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats.
  • invention methods may involve administering to the subject suffering from or at risk of a cancer an effective amount of any one or more pan Raf inhibitor(s) as described herein, wherein the cancer is selected from the group consisting of melanoma, glioma, glioblastoma, pilocytic astrocytoma, liver cancer, biliary tract cancer, cholangiocarcinoma, colorectal cancer, lung cancer, bladder cancer, gallbladder cancer, breast cancer, pancreatic cancer, thyroid cancer, kidney cancer, ovarian cancer, adrenocortical cancer, prostate cancer, gastrointestinal stromal tumors, medullary thyroid cancer, tumor angiogenesis, acute myeloid leukemia, chronic myelomonocytic leukemia, childhood acute lymphoblastic leukemia, plasma cell leukemia, and multiple myeloma.
  • the cancer is selected from the group consisting of melanoma, glioma, glioblastoma, pilocytic
  • any one or more compound(s) as described herein can be used in the preparation of a medicament for the treatment of an A-Raf-mediated, B-Raf-mediated or c-Raf-1-mediated disease or condition selected from the group consisting of neurologic diseases, including, but not limited to, multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease, seizures and epilepsy; neoplastic diseases including, but not limited to, melanoma, glioma, glioblastoma multiforme, pilocytic astrocytoma sarcoma, carcinoma (e.g.
  • lymphoma e.g.
  • histiocytic lymphoma neurofibromatosis, gastrointestinal stromal tumors, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuroendocrine tumors such as medullary thyroid cancer, carcinoid, small cell lung cancer, Kaposi's sarcoma, and pheochromocytoma; pain of neuropathic or inflammatory origin, including, but not limited to, acute pain, chronic pain, cancer-related pain, and migraine; cardiovascular diseases including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g.
  • inflammation and/or proliferation including, but not limited to, psoriasis, eczema, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease (IBD); immunodeficiency diseases, including, but not limited to, organ transplant rejection, graft versus host disease, and Kaposi's sarcoma associated with HIV; renal, cystic, or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia, polycystic liver disease, tuberous sclerosis, Von Hippel Lindau disease, medullary cystic kidney disease, nephronophthisis, and cystic fibrosis; metabolic disorders, including, but not limited to, obesity
  • the disease or condition is selected from the group consisting of melanoma, glioma, glioblastoma multiforme, pilocytic astrocytoma sarcoma, liver cancer, biliary tract cancer, cholangiocarcinoma, colorectal cancer, lung cancer, gallbladder cancer, breast cancer, pancreatic cancer, thyroid cancer, renal cancer, ovarian cancer, adrenocortical cancer, prostate cancer, histiocytic lymphoma, neurofibromatosis, gastrointestinal stromal tumors, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, medullary thyroid cancer, carcinoid, small cell lung cancer, Kaposi's sarcoma, pheochromocytoma, pain, and polycystic kidney disease.
  • the disease or condition is selected from the group consisting of melanoma, glioma, glioblastoma multiforme, pilocytic astrocytoma colorectal cancer, thyroid cancer, lung cancer, ovarian cancer, prostate cancer, liver cancer, gallbladder cancer, gastrointestinal stromal tumors, biliary tract cancer, cholangiocarcinoma, acute pain, chronic pain, and polycystic kidney disease.
  • any one or more pan Raf inhibitor(s) as described herein can be used in the preparation of a medicament for the treatment of a cancer selected from the group consisting of melanoma, glioma, glioblastoma, pilocytic astrocytoma, liver cancer, biliary tract cancer, cholangiocarcinoma, colorectal cancer, lung cancer, bladder cancer, gallbladder cancer, breast cancer, pancreatic cancer, thyroid cancer, kidney cancer, ovarian cancer, adrenocortical cancer, prostate cancer, gastrointestinal stromal tumors, medullary thyroid cancer, tumor angiogenesis, acute myeloid leukemia, chronic myelomonocytic leukemia, childhood acute lymphoblastic leukemia, plasma cell leukemia, and multiple myeloma.
  • a cancer selected from the group consisting of melanoma, glioma, glioblastoma, pilocytic astrocytoma, liver cancer, bil
  • a compound as disclosed herein (including any compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Table I, or any other compounds specifically disclosed herein) or any compounds recited in claims 1 - 27 is a pan Raf inhibitor.
  • a compound as disclosed herein (including any compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Table I, or any other compounds specifically disclosed herein) is a Ras activity inhibitor.
  • a compound as disclosed herein (including any compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Table I, or any other compounds specifically disclosed herein) is both a pan Raf inhibitor and a Ras activity inhibitor.
  • a compound as disclosed herein (including any compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Table I, or any other compounds specifically disclosed herein) is a pan Raf inhibitor having an IC 50 of less than 500 nM in activity assays for each of B-Raf, c-Raf-1 and B-Raf V600E protein kinases and is a Ras activity inhibitor that inhibits proliferation of a mutant Ras cell line with an IC 50 of less than 1 ⁇ M.
  • the invention provides a method for preparing a compound of Formula I
  • the method includes contacting a compound of Formula Ia:
  • X 1 is a halogen, such as Cl, Br or I; P 1 is a protecting group;
  • Q is a boronic acid or ester residue.
  • Ar is as defined in any of the embodiments descried herein for compounds of Formulas I-XX, Table I or any other compounds specifically disclosed herein or any compounds recited in claims 1 - 27 and in the Examples below.
  • Exemplary Q includes, but is not limited to —B(OH) 2 or
  • the reaction is performed in the presence of a palladium complex, such as 1,1′-bis(diphenylphosphino)ferroceneklichloropalladium(II).
  • a palladium complex such as 1,1′-bis(diphenylphosphino)ferroceneklichloropalladium(II).
  • protecting group P 1 is t-butoxycarbonyl, 2,6-dichlorophenylcarbonyl or phenylsulfonyl.
  • hydrogen includes for example 1 H, 2 H, 3 H; carbon includes for example 11 C, 12 C, 13 C, 14 C; oxygen includes for example 16 O, 17 O, 18 O; nitrogen includes for example 13 N, 14 N, 15 N; sulfur includes for example 32 S, 33 S, 34 S, 35 S, 36 S, 37 S, 38 S; fluoro includes for example 17 F, 18 F, 19 F; chloro includes for example 35 Cl, 36 Cl, 37 Cl, 38 Cl, 39 Cl; and the like.
  • Halogen refer to all halogens, that is, chloro (Cl), fluoro (F), bromo (Br), or iodo (I).
  • Haldroxyl or “hydroxy” refer to the group —OH.
  • Thiol refers to the group —SH.
  • “Lower alkyl” alone or in combination means an alkane-derived radical containing from 1 to 6 carbon atoms (unless specifically defined) that includes a straight chain alkyl or branched alkyl.
  • the straight chain or branched lower alkyl group is chemically feasible and attached at any available point to provide a stable compound.
  • a lower alkyl is a straight or branched alkyl group containing from 1-6, 1-4, or 1-2, carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, t-butyl, and the like.
  • a lower alkyl may be independently substituted as described herein, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, wherein the substituents are as indicated.
  • fluoro substituted lower alkyl denotes a lower alkyl group substituted with one or more fluoro atoms, such as perfluoroalkyl, where preferably the lower alkyl is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms. It is understood that any such substitutions, or substitution of lower alkyl on another moiety are chemically feasible and attached at any available atom to provide a stable compound.
  • Cycloalkyl refers to saturated or unsaturated, non-aromatic monocyclic carbon ring systems of 3-10, also 3-8, more preferably 3-6, ring members per ring, such as cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, and the like.
  • a “substituted cycloalkyl” is a cycloalkyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, wherein the substituents are as indicated. It is understood that substitutions on cycloalkyl, or substitutions of cycloalkyl on another moiety are chemically feasible and attached at any available atom to provide a stable compound.
  • Heterocycloalkyl refers to a saturated or unsaturated non-aromatic cycloalkyl group having from 5 to 10 atoms in which from 1 to 3 carbon atoms in the ring are replaced by heteroatoms of O, S or N, and are optionally fused with benzo or heteroaryl of 5-6 ring members. Heterocycloalkyl is also intended to include oxidized S or N, such as sulfinyl, sulfonyl and N-oxide of a tertiary ring nitrogen. Heterocycloalkyl is also intended to include compounds in which a ring carbon may be oxo substituted, i.e.
  • the ring carbon is a carbonyl group, such as lactones and lactams.
  • the point of attachment of the heterocycloalkyl ring is at a carbon or nitrogen atom such that a stable ring is retained.
  • heterocycloalkyl groups include, but are not limited to, morpholino, tetrahydrofuranyl, dihydropyridinyl, piperidinyl, pyrrolidinyl, pyrrolidonyl, piperazinyl, dihydrobenzofuryl, and dihydroindolyl.
  • a “substituted heterocycloalkyl” is a heterocycloalkyl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, wherein the substituents are as indicated. It is understood that substitutions on heterocycloalkyl, or substitutions of heterocycloalkyl on another moiety are chemically feasible and attached at any available atom to provide a stable compound.
  • Aryl alone or in combination refers to a monocyclic or bicyclic ring system containing aromatic hydrocarbons such as phenyl or naphthyl, which may be optionally fused with a cycloalkyl of preferably 5-7, more preferably 5-6, ring members.
  • a “substituted aryl” is an aryl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, wherein the substituents are as indicated. It is understood that substitutions on aryl, or substitutions of aryl on another moiety are chemically feasible and attached at any available atom to provide a stable compound.
  • Heteroaryl alone or in combination refers to a monocyclic aromatic ring structure containing 5 or 6 ring atoms, or a bicyclic aromatic group having 8 to 10 atoms, containing one or more, preferably 1-4, more preferably 1-3, even more preferably 1-2, heteroatoms independently selected from the group consisting of O, S, and N. Heteroaryl is also intended to include oxidized S or N, such as sulfinyl, sulfonyl and N-oxide of a tertiary ring nitrogen. A carbon or nitrogen atom is the point of attachment of the heteroaryl ring structure such that a stable compound is provided.
  • heteroaryl groups include, but are not limited to, pyridinyl, pyridazinyl, pyrazinyl, quinaoxalyl, indolizinyl, benzo[b]thienyl, quinazolinyl, purinyl, indolyl, quinolinyl, pyrimidinyl, pyrrolyl, pyrazolyl, oxazolyl, thiazolyl, thienyl, isoxazolyl, oxathiadiazolyl, isothiazolyl, tetrazolyl, imidazolyl, triazolyl, furanyl, benzofuryl, and indolyl.
  • a “substituted heteroaryl” is a heteroaryl that is independently substituted, unless indicated otherwise, with one or more, preferably 1, 2, 3, 4 or 5, also 1, 2, or 3 substituents, wherein the substituents are as indicated. It is understood that substitutions on heteroaryl, or substitutions of heteroaryl on another moiety are chemically feasible and attached at any available atom to provide a stable compound.
  • “Lower alkoxy” denotes the group —OR a , where R a is lower alkyl.
  • “Substituted lower alkoxy” denotes lower alkoxy in which R a is lower alkyl substituted with one or more substituents as indicated herein. Preferably, substitution of lower alkoxy is with 1, 2, 3, 4, or 5 substituents, also 1, 2, or 3 substituents.
  • “fluoro substituted lower alkoxy” denotes lower alkoxy in which the lower alkyl is substituted with one or more fluoro atoms, where preferably the lower alkoxy is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms. It is understood that substitutions on alkoxy, or alkoxy substitution of other moieties are chemically feasible and attached at any available atom to provide a stable compound.
  • Lower alkylthio denotes the group —SR b , where R b is lower alkyl.
  • Substituted lower alkylthio denotes lower alkylthio in which R b is lower alkyl substituted with one or more substituents as indicated herein. Preferably, substitution of lower alkylthio is with 1, 2, 3, 4, or 5 substituents, also 1, 2, or 3 substituents.
  • fluoro substituted lower alkylthio denotes lower alkylthio in which the lower alkyl is substituted with one or more fluoro atoms, where preferably the lower alkylthio is substituted with 1, 2, 3, 4 or 5 fluoro atoms, also 1, 2, or 3 fluoro atoms. It is understood that substitutions on alkylthio, or alkylthio substitution of other moieties are chemically feasible and attached at any available atom to provide a stable compound.
  • “Mono-alkylamino” denotes the group —NHR c where R c is lower alkyl. “Di-alkylamino” denotes the group —NR c R d , where R c and R d are independently lower alkyl. “Cycloalkylamino” denotes the group —NR e R f , where R e and R f combine with the nitrogen to form a 5-7 membered heterocycloalkyl, where the heterocycloalkyl may contain an additional heteroatom within the ring, such as O, N, or S, and may also be further substituted with lower alkyl.
  • Examples of 5-7 membered heterocycloalkyl include, but are not limited to, piperidine, piperazine, 4-methylpiperazine, morpholine, and thiomorpholine. It is understood that when mono-alkylamino, di-alkylamino, or cycloalkylamino are substituents on other moieties, these are chemically feasible and attached at any available atom to provide a stable compound.
  • Protecting group refers to a grouping of atoms that when attached to a reactive group in a molecule masks, reduces or prevents that reactivity. Examples of protecting groups can be found in T. W. Greene and P. G. Wuts, P ROTECTIVE G ROUPS IN O RGANIC C HEMISTRY, (Wiley, 4th ed. 2006), Beaucage and Iyer, Tetrahedron 48:2223-231 1 (1992), and Harrison and Harrison et al., C OMPENDIUM OF S YNTHETIC O RGANIC M ETHODS, Vols. 1-8 (John Wiley and Sons. 1971-1996).
  • Representative amino protecting groups include formyl, acetyl, trifluoroacetyl, benzyl, benzyloxycarbonyl (CBZ), tert-butoxycarbonyl (Boc), trimethyl silyl (TMS), 2-trimethylsilyl-ethanesulfonyl (SES), trityl and substituted trityl groups, allyloxycarbonyl, 9-fluorenylmethyloxycarbonyl (FMOC), nitro-veratryloxycarbonyl (NVOC), tri-isopropylsilyl (TIPS), phenylsulphonyl and the like (see also, Boyle, A. L. (Editor), C URRENT P ROTOCOLS IN N UCLEIC A CID C HEMISTRY, John Wiley and Sons, New York,Volume 1, 2000).
  • the terms “treat”, “treating”, “therapy”, “therapies”, and like terms refer to the administration of material, e.g., any one or more compound(s) as described herein in an amount effective to prevent, alleviate, or ameliorate one or more symptoms of a disease or condition, i.e., indication, and/or to prolong the survival of the subject being treated.
  • Raf protein kinase mediated disease or condition refers to a disease or condition in which the biological function of a Raf protein kinase (also referred to as Raf kinase, or Raf), including any of A-Raf protein kinase, B-Raf protein kinase or c-Raf-1 protein kinase, or any mutation thereof, affects the development, course, and/or symptoms of the disease or condition, and/or in which modulation of Raf alters the development, course, and/or symptoms of the disease or condition.
  • the Raf mediated disease or condition includes a disease or condition for which Raf modulation provides a therapeutic benefit, e.g. wherein treatment with Raf inhibitor(s), including one or more compound(s) described herein, provides a therapeutic benefit to the subject suffering from or at risk of the disease or condition.
  • A-Raf protein kinase mediated disease or condition refers to a disease or condition in which the biological function of an A-Raf protein kinase (also referred to as A-Raf kinase, or A-Raf), including any mutations thereof, affects the development, course, and/or symptoms of the disease or condition, and/or in which modulation of A-Raf alters the development, course, and/or symptoms of the disease or condition.
  • the A-Raf mediated disease or condition includes a disease or condition for which A-Raf inhibition provides a therapeutic benefit, e.g. wherein treatment with a compound that inhibits A-Raf, including one or more compound(s) described herein, provides a therapeutic benefit to the subject suffering from or at risk of the disease or condition.
  • B-Raf protein kinase mediated disease or condition refers to a disease or condition in which the biological function of a B-Raf protein kinase (also referred to as B-Raf kinase, or B-Raf), including any mutations thereof, affects the development, course, and/or symptoms of the disease or condition, and/or in which modulation of B-Raf alters the development, course, and/or symptoms of the disease or condition.
  • the B-Raf mediated disease or condition includes a disease or condition for which B-Raf inhibition provides a therapeutic benefit, e.g. wherein treatment with a compound that inhibits B-Raf, including one or more compound(s) described herein, provides a therapeutic benefit to the subject suffering from or at risk of the disease or condition.
  • B-Raf V600E mutant protein kinase mediated disease or condition refers to a disease or condition in which the biological function of B-Raf V600E mutant protein kinase (also referred to as B-Raf V600E kinase, or B-Raf V600E) affects the development, course, and/or symptoms of the disease or condition, and/or in which modulation of B-Raf V600E alters the development, course, and/or symptoms of the disease or condition.
  • the B-Raf V600E mediated disease or condition includes a disease or condition for which B-Raf V600E inhibition provides a therapeutic benefit, e.g. wherein treatment with a compound that inhibits B-Raf V600E, including one or more compound(s) described herein, provides a therapeutic benefit to the subject suffering from or at risk of the disease or condition.
  • c-Raf-1 protein kinase mediated disease or condition refers to a disease or condition in which the biological function of a c-Raf-1 protein kinase (also referred to as c-Raf-1 kinase, or c-Raf-1), including any mutations thereof, affects the development, course, and/or symptoms of the disease or condition, and/or in which modulation of c-Raf-1 alters the development, course, and/or symptoms of the disease or condition.
  • the c-Raf-1 mediated disease or condition includes a disease or condition for which c-Raf-1 inhibition provides a therapeutic benefit, e.g. wherein treatment with a compound that inhibits c-Raf-1, including one or more compound(s) described herein, provides a therapeutic benefit to the subject suffering from or at risk of the disease or condition.
  • Raf inhibitor refers to a compound that inhibits at least one of A-Raf, B-Raf, c-Raf-1, or any mutations thereof, i.e. a compound having an IC 50 of less than 500 nM, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM as determined in a generally accepted Raf kinase activity assay.
  • Such compounds are preferably, but not necessarily, selective with respect to other protein kinases, i.e.
  • the IC 50 for the other kinase divided by the IC 50 for the Raf kinase is >10, also >20, also >30, also >40, also >50, also >60, also >70, also >80, also >90, also >100.
  • the compounds are selective relative to other protein kinases including, but not limited to, CSK, Insulin receptor kinase, AMPK, PDGFR or VEGFR.
  • the term “pan Raf inhibitor” refers to a compound that inhibits at least each of B-Raf, c-Raf-1 and B-Raf V600E protein kinases, i.e. a compound having an IC 50 of less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM as determined in a generally accepted B-Raf kinase activity assay, and having an IC 50 of less than 500 nM, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM as determined in a comparable generally accepted c-Raf-1 kinase activity assay, and having an 10 50 of less than 500 nM, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5
  • the pan Raf inhibitor may be, but is not necessarily, approximately equipotent on each of B-Raf, c-Raf-1 and B-Raf V600E.
  • Compounds are considered approximately equipotent on each of B-Raf V600E, B-Raf and c-Raf-1 if the ratio of IC 50 for any of B-Raf V600E, B-Raf and c-Raf-1 divided by the IC 50 for any other of B-Raf V600E, B-Raf and c-Raf-1 (e.g. B-Raf IC 50 divided by B-Raf V600E IC 50 ) is in the range of 10 to 0.1, also 5 to 0.2.
  • Such compounds are preferably, but not necessarily, selective with respect to other protein kinases, i.e. when compared to another protein kinase, the IC 50 for the other kinase divided by the IC 50 for any of B-Raf, c-Raf-1 or B-Raf V600E is >10, also >20, also >30, also >40, also >50, also >60, also >70, also >80, also >90, also >100.
  • the compounds are selective relative to other protein kinases including, but not limited to, CSK, Insulin receptor kinase, AMPK, PDGFR or VEGFR.
  • a pan Raf inhibitor may be used to treat any B-Raf, c-Raf-1 or B-Raf V600E kinase mediated disease or condition
  • the inihibition of each of B-Raf, c-Raf-1 and B-Raf V600E provides beneficial effects in treating cancers, in particular cancers having a Ras pathway mutation, including, but not limited to, melanoma, glioma, glioblastoma, pilocytic astrocytoma, liver cancer, biliary tract cancer, cholangiocarcinoma, colorectal cancer, lung cancer, bladder cancer, gallbladder cancer, breast cancer, pancreatic cancer, thyroid cancer, kidney cancer, ovarian cancer, adrenocortical cancer, prostate cancer, gastrointestinal stromal tumors, medullary thyroid cancer, tumor angiogenesis, acute myeloid leukemia, chronic myelomonocytic leukemia, childhood acute lymphoblastic le
  • Ras activity inhibitor refers to a compound that inhibits proliferation of a mutant Ras cell line; i.e., a compound that inhibits proliferation of a mutant Ras cell line with an IC 50 of less than 1 ⁇ M, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line with an IC 50 of less than 1 ⁇ M. In some embodiments, a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line with an IC 50 of less than 100 nM.
  • a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line with an IC 50 of less than 50 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line with an IC 50 of less than 20 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line with an IC 50 of less than 10 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line with an IC 50 of less than 5 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line with an IC 50 of less than 1 nM.
  • the mutant Ras cell line is a N-Ras mutant cell line, a K-Ras mutant cell line, or a H-Ras mutant cell line.
  • a Ras activity inhibitor inhibits proliferation of a mutant N-Ras cell line with an IC 50 of less than 1 ⁇ M, less than 100 nM, less than 50 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 1 nM.
  • a Ras activity inhibitor inhibits proliferation of a N-Ras mutant cell line with an IC 50 of less than 1 ⁇ M.
  • a Ras activity inhibitor inhibits proliferation of a N-Ras mutant cell line with an IC 50 of less than 100 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of a N-Ras mutant cell line with an IC 50 of less than 50 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of a N-Ras mutant cell line with an IC 50 of less than 20 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of a N-Ras mutant cell line with an IC 50 of less than 10 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of a N-Ras mutant cell line with an IC 50 of less than 5 nM.
  • a Ras activity inhibitor inhibits proliferation of a N-Ras mutant cell line with an IC 50 of less than 1 nM.
  • the N-Ras mutant cell line is one or more cell lines selected from the group consisting of M244, M202, M207, SK-MEL-2, IPC-298, S′117, M296, SK-MEL-30, SK-MEL-173, and HL-60.
  • the N-Ras mutant cell line is one or more cell lines selected from the group consisting of M244, M202, M207, SK-MEL-2, SK-MEL-173, and IPC298.
  • a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line selected from the group consisting of M244, M202, M207, SK-MEL-2, SK-MEL-173, and IPC298 with an IC 50 of less than 1 ⁇ M. In some embodiments, a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line selected from the group consisting of M244, M202, M207, SK-MEL-2, SK-MEL-173, and IPC298 with an IC 50 of less than 100 nM.
  • a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line selected from the group consisting of M244, M202, M207, SK-MEL-2, SK-MEL-173, and IPC298 with an IC 50 of less than 50 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line selected from the group consisting of M244, M202, M207, SK-MEL-2, SK-MEL-173, and IPC298 with an IC 50 of less than 20 nM.
  • a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line selected from the group consisting of M244, M202, M207, SK-MEL-2, SK-MEL-173, and IPC298 with an IC 50 of less than 10 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line selected from the group consisting of M244, M202, M207, SK-MEL-2, SK-MEL-173, and IPC298 with an IC 50 of less than 5 nM.
  • a Ras activity inhibitor inhibits proliferation of a mutant Ras cell line selected from the group consisting of M244, M202, M207, SK-MEL-2, SK-MEL-173, and IPC298 with an IC 50 of less than 1 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of IPC298 cells with an IC 50 of less than 1 ⁇ M. In some embodiments, a Ras activity inhibitor inhibits proliferation of IPC298 cells with an IC 50 of less than 100 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of IPC298 cells with an IC 50 of less than 50 nM.
  • a Ras activity inhibitor inhibits proliferation of IPC298 cells with an IC 50 of less than 20 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of IPC298 cells with an IC 50 of less than 10 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of IPC298 cells with an IC 50 of less than 5 nM. In some embodiments, a Ras activity inhibitor inhibits proliferation of IPC298 cells with an IC 50 of less than 1 nM.
  • solid form refers to a solid preparation (i.e. a preparation that is neither gas nor liquid) of a pharmaceutically active compound that is suitable for administration to an intended animal subject for therapeutic purposes.
  • the solid form includes any complex, such as a salt, co-crystal or an amorphous complex, as well as any polymorph of the compound.
  • the solid form may be substantially crystalline, semi-crystalline or substantially amorphous.
  • the solid form may be administered directly or used in the preparation of a suitable composition having improved pharmaceutical properties.
  • the solid form may be used in a formulation comprising at least one pharmaceutically acceptable carrier or excipient.
  • substantially crystalline material embraces material which has greater than about 90% crystallinity; and “crystalline” material embraces material which has greater than about 98% crystallinity.
  • substantially amorphous material embraces material which has no more than about 10% crystallinity; and “amorphous” material embraces material which has no more than about 2% crystallinity.
  • the term “semi-crystalline” material embraces material which is greater than 10% crystallinity, but no greater than 90% crystallinity; preferably “semi-crystalline” material embraces material which is greater than 20% crystallinity, but no greater than 80% crystallinity.
  • a mixture of solid forms of a compound may be prepared, for example, a mixture of amorphous and crystalline solid forms, e.g. to provide a “semi-crystalline” solid form.
  • Such a “semi-crystalline” solid form may be prepared by methods known in the art, for example by mixing an amorphous solid form with a crystalline solid form in the desired ratio.
  • a compound mixed with acid or base forms an amorphous complex
  • a semi-crystalline solid can be prepared employing an amount of compound component in excess of the stoichiometry of the compound and acid or base in the amorphous complex, thereby resulting in an amount of the amorphous complex that is based on the stoichiometry thereof, with excess compound in a crystalline form.
  • the amount of excess compound used in the preparation of the complex can be adjusted to provide the desired ratio of amorphous complex to crystalline compound in the resulting mixture of solid forms.
  • preparing said complex with a 2:1 mole ratio of compound to acid or base will result in a solid form of 50% amorphous complex and 50% crystalline compound.
  • Such a mixture of solid forms may be beneficial as a drug product, for example, by providing an amorphous component having improved biopharmaceutical properties along with the crystalline component.
  • the amorphous component would be more readily bioavailable while the crystalline component would have a delayed bioavailablity.
  • Such a mixture may provide both rapid and extended exposure to the active compound.
  • the term “complex” refers to a combination of a pharmaceutically active compound and an additional molecular species that forms or produces a new chemical species in a solid form.
  • the complex may be a salt, i.e. where the additional molecular species provides an acid/base counter ion to an acid/base group of the compound resulting in an acid:base interaction that forms a typical salt. While such salt forms are typically substantially crystalline, they can also be partially crystalline, substantially amorphous, or amorphous forms.
  • the additional molecular species, in combination with the pharmaceutically active compound forms a non-salt co-crystal, i.e.
  • the compound and molecular species do not interact by way of a typical acid:base interaction, but still form a substantially crystalline structure.
  • Co-crystals may also be formed from a salt of the compound and an additional molecular species.
  • the complex is a substantially amorphous complex, which may contain salt-like acid:base interactions that do not form typical salt crystals, but instead form a substantially amorphous solid, i.e. a solid whose X-ray powder diffraction pattern exhibits no sharp peaks (e.g. exhibits an amorphous halo).
  • the term “stoichiometry” refers to the molar ratio of a combination of two or more components, for example, the molar ratio of acid or base to compound that form an amorphous complex. For example, a 1:1 mixture of acid or base with compound (i.e. I mole acid or base per mole of compound) resulting in an amorphous solid form has a 1:1 stoichiometry.
  • composition refers to a pharmaceutical preparation suitable for administration to an intended subject for therapeutic purposes that contains at least one pharmaceutically active compound, including any solid form thereof.
  • the composition may include at least one pharmaceutically acceptable component to provide an improved formulation of the compound, such as a suitable carrier or excipient.
  • the term “subject” refers to a living organism that is treated with compounds as described herein, including, but not limited to, any mammal, such as a human, other primates, sports animals, animals of commercial interest such as cattle, farm animals such as horses, or pets such as dogs and cats.
  • biopharmaceutical properties refers to the pharmacokinetic action of a compound or complex of the present invention, including the dissolution, absorption and distribution of the compound on administration to a subject.
  • certain solid forms of compounds of the invention such as amorphous complexes of compounds of the invention, are intended to provide improved dissolution and absorption of the active compound, which is typically reflected in improved C max (i.e. the maximum achieved concentration in the plasma after administration of the drug) and improved AUC (i.e. area under the curve of drug plasma concentration vs. time after administration of the drug).
  • pharmaceutically acceptable indicates that the indicated material does not have properties that would cause a reasonably prudent medical practitioner to avoid administration of the material to a patient, taking into consideration the disease or conditions to be treated and the respective route of administration. For example, it is commonly required that such a material be essentially sterile, e.g., for injectibles.
  • the term “therapeutically effective” or “effective amount” indicates that the materials or amount of material is effective to prevent, alleviate, or ameliorate one or more symptoms of a disease or medical condition, and/or to prolong the survival of the subject being treated.
  • the terms “synergistically effective” or “synergistic effect” indicate that two or more compounds that are therapeutically effective, when used in combination, provide improved therapeutic effects greater than the additive effect that would be expected based on the effect of each compound used by itself.
  • enzymes can be assayed based on their ability to act upon a detectable substrate.
  • a compound can be assayed based on its ability to bind to a particular target molecule or molecules.
  • the term “modulating” or “modulate” refers to an effect of altering a biological activity (i.e. increasing or decreasing the activity), especially a biological activity associated with a particular biomolecule such as a protein kinase.
  • a biological activity i.e. increasing or decreasing the activity
  • an inhibitor of a particular biomolecule modulates the activity of that biomolecule, e.g., an enzyme, by decreasing the activity of the biomolecule, such as an enzyme.
  • Such activity is typically indicated in terms of an inhibitory concentration (IC 50 ) of the compound for an inhibitor with respect to, for example, an enzyme.
  • the term “contacting” means that the compound(s) are caused to be in sufficient proximity to a particular molecule, complex, cell, tissue, organism, or other specified material that potential binding interactions and/or chemical reaction between the compound and other specified material can occur.
  • “Pain” or a “pain condition” can be acute and/or chronic pain, including, without limitation, arachnoiditis; arthritis (e.g. osteoarthritis, rheumatoid arthritis, ankylosing spondylitis, gout); back pain (e.g. sciatica, ruptured disc, spondylolisthesis, radiculopathy); burn pain; cancer pain; dysmenorrhea; headaches (e.g. migraine, cluster headaches, tension headaches); head and facial pain (e.g. cranial neuralgia, trigeminal neuralgia); hyperalgesia; hyperpathia; inflammatory pain (e.g.
  • irritable bowel syndrome pain associated with irritable bowel syndrome, inflammatory bowel disease, ulcerative colitis, Crohn's disease, cystitis, pain from bacterial, fungal or viral infection); keloid or scar tissue formation; labor or delivery pain; muscle pain (e.g. as a result of polymyositis, dermatomyositis, inclusion body myositis, repetitive stress injury (e.g. writer's cramp, carpal tunnel syndrome, tendonitis, tenosynovitis)); myofascial pain syndromes (e.g. fibromyalgia); neuropathic pain (e.g.
  • diabetic neuropathy causalgia, entrapment neuropathy, brachial plexus avulsion, occipital neuralgia, gout, reflex sympathetic dystrophy syndrome, phantom limb or post-amputation pain, postherpetic neuralgia, central pain syndrome, or nerve pain resulting from trauma (e.g. nerve injury), disease (e.g. diabetes, multiple sclerosis, Guillan-Barre Syndrome, myasthenia gravis, neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, or cancer treatment); pain associated with skin disorders (e.g. shingles, herpes simplex, skin tumors, cysts, neurofibromatosis); sports injuries (e.g.
  • vascular disease or injury e.g. vasculitis, coronary artery disease, reperfusion injury (e.g. following ischemia, stroke, or myocardial infarcts)
  • other specific organ or tissue pain e.g. ocular pain, corneal pain, bone pain, heart pain, visceral pain (e.g. kidney, gallbladder, gastrointestinal), joint pain, dental pain, pelvic hypersensitivity, pelvic pain, renal colic, urinary incontinence
  • other disease associated pain e.g.
  • sickle cell anemia AIDS, herpes zoster, psoriasis, endometriosis, asthma, chronic obstructive pulmonary disease (COPD), silicosis, pulmonary sarcoidosis, esophagitis, heart burn, gastroesophageal reflux disorder, stomach and duodenal ulcers, functional dyspepsia, bone resorption disease, osteoporosis, cerebral malaria, bacterial meningitis); or pain due to graft v. host rejection or allograft rejections.
  • COPD chronic obstructive pulmonary disease
  • Protein kinases play key roles in propagating biochemical signals in diverse biological pathways. More than 500 kinases have been described, and specific kinases have been implicated in a wide range of diseases or conditions (i.e., indications), including for example without limitation, cancer, cardiovascular disease, inflammatory disease, neurological disease, and other diseases. As such, kinases represent important control points for small molecule therapeutic intervention. Specific target protein kinases contemplated by the present invention are described in the art, including, without limitation, protein kinases as described in U.S. patent application Ser. No. 11/473,347 (see also, PCT publication WO2007002433), the disclosure of which is hereby incorporated by reference as it relates to such kinase targets, as well as the following:
  • A-Raf Target kinase
  • A-Raf i.e., v-raf murine sarcoma 3611 viral oncogene homolog 1
  • ARAF ARAF
  • the mature protein comprises RBD (i.e., Ras binding domain) and phorbol-ester/DAG-type zinc finger domain and is involved in the transduction of mitogenic signals from the cell membrane to the nucleus.
  • A-Raf inhibitors may be useful in treating neurologic diseases such as multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease; neoplastic diseases including, but not limited to, melanoma, glioma, sarcoma, carcinoma (e.g. colorectal, lung, breast, pancreatic, thyroid, renal, ovarian), lymphoma (e.g.
  • neurologic diseases such as multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease
  • neoplastic diseases including, but not limited to, melanoma, glioma, sarcoma, carcinoma (e.g. colorectal, lung, breast, pancreatic, thyroid, renal, ovarian), lymphoma (e.g.
  • muscle regeneration or degeneration including, but not limited to, vascular restenosis, sarcopenia, muscular dystrophies (including, but not limited to, Duchenne, Becker, Emery-Dreifuss, Limb-Girdle, Facioscapulohumeral, Myotonic, Oculopharyngeal, Distal and Congenital Muscular Dystrophies), motor neuron diseases (including, but not limited to, amyotrophic lateral sclerosis, infantile progressive spinal muscular atrophy, intermediate spinal muscular atrophy, juvenile spinal muscular atrophy, spinal bulbar muscular atrophy, and adult spinal muscular atrophy), inflammatory myopathies (including, but not limited to, dermatomyositis, polymyositis, and inclusion body myos
  • B-Raf Target kinase B-Raf (i.e., v-raf murine sarcoma viral oncogene homolog B1) is a 84.4 kDa serine/threonine kinase encoded by chromosome 7q34 (symbol: BRAF).
  • the mature protein comprises RBD (i.e., Ras binding domain), C1 (i.e., protein kinase C conserved region 1) and STK (i.e., serine/threonine kinase) domains.
  • Target kinase B-Raf is involved in the transduction of mitogenic signals from the cell membrane to the nucleus and may play a role in the postsynaptic responses of hippocampal neurons.
  • genes of the RAF family encode kinases that are regulated by Ras and mediate cellular responses to growth signals.
  • B-Raf kinase is a key component of the RAS ⁇ Raf ⁇ MEK ⁇ ERK/MAP kinase signaling pathway, which plays a fundamental role in the regulation of cell growth, division and proliferation, and, when constitutively activated, causes tumorigenesis.
  • the B-type, or B-Raf is the strongest activator of the downstream MAP kinase signaling.
  • the BRAF gene is frequently mutated in a variety of human tumors, especially in malignant melanoma and colon carcinoma.
  • the most common reported mutation was a missense thymine (T) to adenine (A) transversion at nucleotide 1796 (T1796A; amino acid change in the B-Raf protein is Val ⁇ 600> to Glu ⁇ 600>) observed in 80% of malignant melanoma tumors.
  • T1796A missense thymine
  • A adenine transversion at nucleotide 1796
  • Functional analysis reveals that this transversion is the only detected mutation that causes constitutive activation of B-Raf kinase activity, independent of RAS activation, by converting B-Raf into a dominant transforming protein.
  • Niihori et al. report that in 43 individuals with cardio-facio-cutaneous (CFC) syndrome, they identified two heterozygous KRAS mutations in three individuals and eight BRAF mutations in 16 individuals, suggesting that dysregulation of the RAS-RAF-ERK pathway is a common molecular basis for the three related disorders (Niihori et al., Nat Genet. 2006, 38(3):294-6).
  • Raf kinase inhibitors such as the Pan Raf kinase inhibitors as described herein.
  • the ability of these compounds to inhibit multiple Raf kinase targets, including c-Raf-1, B-Raf, and B-Raf V600E, provides additional benefits for inhibiting activating mutations in this pathway, with such cancers less likely to develop resistance to such inhibitors as they are targeting several points in the pathway.
  • Pan Raf kinase inhibitors as described herein may be useful in treating a variety of cancers, including, but not limited to, melanoma, glibma, glioblastoma mulitforme, pilocytic astrocytoma, carcinoma (e.g.
  • gastrointestinal, liver, biliary tract, bile duct (cholangiocarcinoma), colorectal, lung, brain, bladder, gallbladder, breast, pancreatic, thyroid, kidney, ovarian, adrenocortical, prostate), gastrointestinal stromal tumors, medullary thyroid cancer, tumor angiogenesis, acute myeloid leukemia, chronic myelomonocytic leukemia, childhood acute lymphoblastic leukemia, plasma cell leukemia, and multiple myeloma. See McDermott et al., PNAS, 2007, 104(50): 19936-19941; and Jaiswal et al., PLoS One, 2009, 4(5):e5717.
  • c-Raf-1 Target kinase c-Raf-1 (i.e., v-raf murine sarcoma viral oncogene homolog 1) is a 73.0 kDa STK encoded by chromosome 3p25 (symbol: RAF1).
  • c-Raf-1 can be targeted to the mitochondria by BCL2 (i.e., oncogene B-cell leukemia 2) which is a regulator of apoptotic cell death. Active c-Raf-1 improves BCL2-mediated resistance to apoptosis, and c-Raf-1 phosphorylates BAD (i.e., BCL2-binding protein).
  • BAD i.e., BCL2-binding protein
  • c-Raf-1 is implicated in carcinomas, including colorectal, ovarian, lung and renal cell carcinoma. c-Raf-1 is also implicated as an important mediator of tumor angiogenesis (Hood, J. D. et al., 2002, Science 296, 2404). c-Raf-1 inhibitors may also be useful for the treatment of acute myeloid leukemia and myelodysplastic syndromes (Crump, Curr Pharm Des 2002, 8(25):2243-8).
  • c-Raf-1 activators may be useful as treatment for neuroendocrine tumors, such as medullary thyroid cancer, carcinoid, small cell lung cancer and pheochromocytoma (Kunnimalaiyaan et al., Anticancer Drugs 2006, 17(2):139-42).
  • Raf inhibitors may be useful in treating A-Raf-mediated, B-Raf-mediated or c-Raf-1-mediated diseases or conditions selected from the group consisting of neurologic diseases, including, but not limited to, multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease, seizures and epilepsy; neoplastic diseases including, but not limited to, melanoma, glioma, glioblastoma multiforme, pilocytic astrocytoma, sarcoma, carcinoma (e.g.
  • neurologic diseases including, but not limited to, multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease, seizures and epilepsy
  • neoplastic diseases including, but not limited to, melanoma, glioma, glioblastoma multiforme, pilocytic astrocytoma, sarcoma, carcinoma (e.g.
  • bile duct cholangiocarcinoma
  • colorectal lung, brain, bladder, gallbladder, breast, pancreatic, thyroid, renal, ovarian, adrenocortical, prostate
  • lymphoma e.g.
  • histiocytic lymphoma neurofibromatosis, acute myeloid leukemia, myelodysplastic syndrome, leukemia, chronic myelomonocytic leukemia, childhood, acute lymphoblastic leukemia, plasma cell leukemia, multiple myeloma, tumor angiogenesis, gastrointestinal stromal tumors, neuroendocrine tumors such as medullary thyroid cancer, carcinoid, small cell lung cancer, Kaposi's sarcoma, and pheochromocytoma; pain of neuropathic or inflammatory origin, including, but not limited to, acute pain, chronic pain, cancer-related pain, and migraine; cardiovascular diseases including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g.
  • inflammation and/or proliferation including, but not limited to, psoriasis, eczema, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease (IBD); immunodeficiency diseases, including, but not limited to, organ transplant rejection, graft versus host disease, and Kaposi's sarcoma associated with HIV; renal, cystic, or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia, polycystic liver disease, tuberous sclerosis, Von Hippel Lindau disease, medullary cystic kidney disease, nephronophthisis, and cystic fibrosis; metabolic disorders, including, but not limited to, obesity
  • a number of different assays for kinase activity can be utilized for assaying for active modulators and/or determining specificity of a modulator for a particular kinase or group or kinases.
  • assays for kinase activity can be utilized for assaying for active modulators and/or determining specificity of a modulator for a particular kinase or group or kinases.
  • one of ordinary skill in the art will know of other assays that can be utilized and can modify an assay for a particular application. For example, numerous papers concerning kinases describe assays that can be used.
  • Additional alternative assays can employ binding determinations.
  • this sort of assay can be formatted either in a fluorescence resonance energy transfer (FRET) format, or using an AlphaScreen (amplified luminescent proximity homogeneous assay) format by varying the donor and acceptor reagents that are attached to streptavidin or the phosphor-specific antibody.
  • FRET fluorescence resonance energy transfer
  • AlphaScreen amplified luminescent proximity homogeneous assay
  • invention compounds may exist in a number of different forms or derivatives, all within the scope of the present invention.
  • Alternative forms or derivatives include, for example, (a) prodrugs, and active metabolites (b) tautomers, isomers (including stereoisomers and regioisomers), and racemic mixtures (c) pharmaceutically acceptable salts and (d) solid forms, including different crystal forms, polymorphic or amorphous solids, including hydrates and solvates thereof, and other forms.
  • the invention also includes prodrugs (generally pharmaceutically acceptable prodrugs), active metabolic derivatives (active metabolites), and their pharmaceutically acceptable salts.
  • Prodrugs are compounds or pharmaceutically acceptable salts thereof which, when metabolized under physiological conditions or when converted by solvolysis, yield the desired active compound.
  • Prodrugs include, without limitation, esters, amides, carbamates, carbonates, ureides, solvates, or hydrates of the active compound.
  • the prodrug is inactive, or less active than the active compound, but may provide one or more advantageous handling, administration, and/or metabolic properties.
  • some prodrugs are esters of the active compound; during metabolysis, the ester group is cleaved to yield the active drug.
  • Esters include, for example, esters of a carboxylic acid group, or S-acyl or O-acyl derivatives of thiol, alcohol, or phenol groups.
  • Prodrugs may also include variants wherein an —NH group of the compound has undergone acylation, such as the 1-position of the 1H-pyrrolo[2,3-b]pyridine ring, or the nitrogen of the sulfonamide group of compounds as described herein, where cleavage of the acyl group provides the free —NH group of the active drug.
  • Some prodrugs are activated enzymatically to yield the active compound, or a compound may undergo further chemical reaction to yield the active compound.
  • Prodrugs may proceed from prodrug form to active form in a single step or may have one or more intermediate forms which may themselves have activity or may be inactive.
  • bioprecursor prodrugs can be conceptually divided into two non-exclusive categories, bioprecursor prodrugs and carrier prodrugs.
  • bioprecursor prodrugs are compounds that are inactive or have low activity compared to the corresponding active drug compound, that contain one or more protective groups and are converted to an active form by metabolism or solvolysis. Both the active drug form and any released metabolic products should have acceptably low toxicity.
  • the formation of active drug compound involves a metabolic process or reaction that is one of the following types:
  • Oxidative reactions are exemplified without limitation by reactions such as oxidation of alcohol, carbonyl, and acid functionalities, hydroxylation of aliphatic carbons, hydroxylation of alicyclic carbon atoms, oxidation of aromatic carbon atoms, oxidation of carbon-carbon double bonds, oxidation of nitrogen-containing functional groups, oxidation of silicon, phosphorus, arsenic, and sulfur, oxidative N-dealkylation, oxidative O- and S-dealkylation, oxidative deamination, as well as other oxidative reactions.
  • Reductive reactions are exemplified without limitation by reactions such as reduction of carbonyl functionalitites, reduction of alcohol functionalities and carbon-carbon double bonds, reduction of nitrogen-containing functional groups, and other reduction reactions.
  • Reactions without change in the state of oxidation are exemplified without limitation by reactions such as hydrolysis of esters and ethers, hydrolytic cleavage of carbon-nitrogen single bonds, hydrolytic cleavage of non-aromatic heterocycles, hydration and dehydration at multiple bonds, new atomic linkages resulting from dehydration reactions, hydrolytic dehalogenation, removal of hydrogen halide molecule, and other such reactions.
  • Carrier prodrugs are drug compounds that contain a transport moiety, e.g., that improves uptake and/or localized delivery to a site(s) of action.
  • a transport moiety e.g., that improves uptake and/or localized delivery to a site(s) of action.
  • the linkage between the drug moiety and the transport moiety is a covalent bond
  • the prodrug is inactive or less active than the drug compound
  • the prodrug and any release transport moiety are acceptably non-toxic.
  • the transport moiety is intended to enhance uptake
  • the release of the transport moiety should be rapid.
  • it is desirable to utilize a moiety that provides slow release e.g., certain polymers or other moieties, such as cyclodextrins.
  • carrier prodrugs are often advantageous for orally administered drugs.
  • the transport moiety provides targeted delivery of the drug, for example the drug may be conjugated to an antibody or antibody fragment.
  • Carrier prodrugs can, for example, be used to improve one or more of the following properties: increased lipophilicity, increased duration of pharmacological effects, increased site-specificity, decreased toxicity and adverse reactions, and/or improvement in drug formulation (e.g., stability, water solubility, suppression of an undesirable organoleptic or physiochemical property).
  • lipophilicity can be increased by esterification of hydroxyl groups with lipophilic carboxylic acids, or of carboxylic acid groups with alcohols, e.g., aliphatic alcohols. Wermuth, supra.
  • Metabolites e.g., active metabolites
  • prodrugs as described above, e.g., bioprecursor prodrugs.
  • metabolites are pharmacologically active compounds or compounds that further metabolize to pharmacologically active compounds that are derivatives resulting from metabolic processes in the body of a subject.
  • active metabolites are such pharmacologically active derivative compounds.
  • the prodrug compound is generally inactive or of lower activity than the metabolic product.
  • the parent compound may be either an active compound or may be an inactive prodrug.
  • one or more alkoxy groups can be metabolized to hydroxyl groups while retaining pharmacologic activity and/or carboxyl groups can be esterified, e.g., glucuronidation.
  • carboxyl groups can be esterified, e.g., glucuronidation.
  • there can be more than one metabolite where an intermediate metabolite(s) is further metabolized to provide an active metabolite.
  • a derivative compound resulting from metabolic glucuronidation may be inactive or of low activity, and can be further metabolized to provide an active metabolite.
  • Metabolites of a compound may be identified using routine techniques known in the art, and their activities determined using tests such as those described herein. See, e.g., Bertolini et al., 1997, J. Med. Chem., 40:2011-2016; Shan et al., 1997, J Pharm Sci 86(7):756-757; Bagshawe, 1995, Drug Dev. Res., 34:220-230; Wermuth, supra.
  • some of the compounds according to the present invention may exist as stereoisomers, i.e. having the same atomic connectivity of covalently bonded atoms yet differing in the spatial orientation of the atoms.
  • compounds may be optical stereoisomers, which contain one or more chiral centers, and therefore, may exist in two or more stereoisomeric forms (e.g. enantiomers or diastereomers).
  • stereoisomers i.e., essentially free of other stereoisomers
  • racemates i.e., essentially free of other stereoisomers
  • stereoisomers include geometric isomers, such as cis- or trans- orientation of substituents on adjacent carbons of a double bond. All such single stereoisomers, racemates and mixtures thereof are intended to be within the scope of the present invention. Unless specified to the contrary, all such steroisomeric forms are included within the formulae provided herein.
  • a chiral compound of the present invention is in a form that contains at least 80% of a single isomer (60% enantiomeric excess (“e.e.”) or diastereomeric excess (“d.e.”)), or at least 85% (70% e.e. or d.e.), 90% (80% e.e. or d.e.), 95% (90% e.e. or d.e.), 97.5% (95% e.e. or d.e.), or 99% (98% e.e. or d.e.).
  • 60% enantiomeric excess (“e.e.”) or diastereomeric excess (“d.e.”) or at least 85% (70% e.e. or d.e.), 90% (80% e.e. or d.e.), 95% (90% e.e. or d.e.), 97.5% (95% e.e. or d.e.), or 99% (98% e.e. or d.e
  • an optically pure compound having one chiral center is one that consists essentially of one of the two possible enantiomers (i.e., is enantiomerically pure), and an optically pure compound having more than one chiral center is one that is both diastereomerically pure and enantiomerically pure.
  • the compound is present in optically pure form, such optically pure form being prepared and/or isolated by methods known in the art (e.g. by recrystallization techniques, chiral synthetic techniques (including synthesis from optically pure starting materials), and chromatographic separation using a chiral column.
  • a compound herein includes pharmaceutically acceptable salts of such compound.
  • compounds described herein can be in the form of pharmaceutically acceptable salts, or can be formulated as pharmaceutically acceptable salts.
  • Contemplated pharmaceutically acceptable salt forms include, without limitation, mono, bis, tris, tetrakis. and so on.
  • Pharmaceutically acceptable salts are non-toxic in the amounts and concentrations at which they are administered. The preparation of such salts can facilitate the pharmacological use by altering the physical characteristics of a compound without preventing it from exerting its physiological effect. Useful alterations in physical properties include lowering the melting point to facilitate transmucosal administration and increasing the solubility to facilitate administering higher concentrations of the drug.
  • a compound of the invention may possess a sufficiently acidic, a sufficiently basic, or both functional groups, and accordingly can react with any of a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt.
  • Pharmaceutically acceptable salts include acid addition salts such as those containing chloride, bromide, iodide, hydrochloride, acetate, phenylacetate, acrylate, ascorbate, aspartate, benzoate, 2-phenoxybenzoate, 2-acetoxybenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, methylbenzoate, bicarbonate, butyne-1,4 dioate, hexyne-1,6-dioate, caproate, caprylate, chlorobenzoate, cinnamate, citrate, decanoate, formate, fumarate, glycolate, gluconate, glucarate, glucuronate, glucose-6-phosphate, glutamate, heptanoate, hexanoate, isethionate, isobutyrate, gamma-hydroxybutyrate, phenylbutyrate, lactate, malate, maleate, hydroxymaleate, methyl
  • pharmaceutically acceptable salts also include basic addition salts such as those containing benzathine, chloroprocaine, choline, ethanolamine, diethanolamine, triethanolamine, t-butylamine, dicyclohexylamine, ethylenediamine, N,N′-dibenzylethylenediamine, meglumine, hydroxyethylpyrrolidine, piperidine, morpholine, piperazine, procaine, aluminum, calcium, copper, iron, lithium, magnesium, manganese, potassium, sodium, zinc, ammonium, and mono-, di-, or tri-alkylamines (e.g.
  • diethylamine or salts derived from amino acids such as L-histidine, L-glycine, L-lysine, and L-arginine.
  • L-histidine amino acids
  • L-glycine amino acids
  • L-lysine amino acids
  • L-arginine amino acids
  • salts can be prepared by standard techniques.
  • the free-base form of a compound can be dissolved in a suitable solvent, such as an aqueous or aqueous-alcohol solution containing the appropriate acid and then isolated by evaporating the solution.
  • a salt can be prepared by reacting the free base and acid in an organic solvent. If the particular compound is an acid, the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an appropriate inorganic or organic base.
  • the compounds and salts may exist in different crystal or polymorphic forms, or may be formulated as co-crystals, or may be in an amorphous form, or may be any combination thereof (e.g. partially crystalline, partially amorphous, or mixtures of polymorphs) all of which are intended to be within the scope of the present invention and specified formulae.
  • salts are formed by acid/base addition, i.e.
  • co-crystals are a new chemical species that is formed between neutral compounds, resulting in the compound and an additional molecular species in the same crystal structure.
  • compounds of the invention are complexed with an acid or a base, including base addition salts such as ammonium, diethylamine, ethanolamine, ethylenediamine, diethanolamine, t-butylamine, piperazine, meglumine; acid addition salts, such as acetate, acetylsalicylate, besylate, camsylate, citrate, formate, fumarate, glutarate, hydrochlorate, maleate, mesylate, nitrate, oxalate, phosphate, succinate, sulfate, tartrate, thiocyanate and tosylate; and amino acids such as alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, t
  • an amorphous complex is preferably formed rather than a crystalline material such as a typical salt or co-crystal.
  • the amorphous form of the complex is facilitated by additional processing, such as by spray-drying, mechanochemical methods such as roller compaction, or microwave irradiation of the parent compound mixed with the acid or base.
  • additional processing such as by spray-drying, mechanochemical methods such as roller compaction, or microwave irradiation of the parent compound mixed with the acid or base.
  • Such methods may also include addition of ionic and/or non-ionic polymer systems, including, but not limited to, hydroxypropyl methyl cellulose acetate succinate (HPMCAS) and methacrylic acid copolymer (e.g. Eudragit® L100-55), that further stabilize the amorphous nature of the complex.
  • HPMCAS hydroxypropyl methyl cellulose acetate succinate
  • methacrylic acid copolymer e.g. Eudragit® L100-
  • amorphous complexes provide several advantages. For example, lowering of the melting temperature relative to the free base facilitiates additional processing, such as hot melt extrusion, to further improve the biopharmaceutical properties of the compound. Also, the amorphous complex is readily friable, which provides improved compression for loading of the solid into capsule or tablet form.
  • the formulae are intended to cover hydrated or solvated as well as unhydrated or unsolvated forms of the identified structures.
  • the indicated compounds include both hydrated and non-hydrated forms.
  • Other examples of solvates include the structures in combination with a suitable solvent, such as isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, or ethanolamine.
  • the methods and compounds will typically be used in therapy for human subjects. However, they may also be used to treat similar or identical indications in other animal subjects.
  • Compounds described herein can be administered by different routes, including injection (i.e. parenteral, including intravenous, intraperitoneal, subcutaneous, and intramuscular), oral, transdermal, transmucosal, rectal, or inhalant. Such dosage forms should allow the compound to reach target cells. Other factors are well known in the art, and include considerations such as toxicity and dosage forms that retard the compound or composition from exerting its effects. Techniques and formulations generally may be found in Remington: The Science and Practice of Pharmacy, 21 st edition, Lippincott, Williams and Wilkins, Philadelphia, Pa., 2005 (hereby incorporated by reference herein).
  • compositions will comprise pharmaceutically acceptable carriers or excipients, such as fillers, binders, disintegrants, glidants, lubricants, complexing agents, solubilizers, and surfactants, which may be chosen to facilitate administration of the compound by a particular route.
  • carriers include calcium carbonate, calcium phosphate, various sugars such as lactose, glucose, or sucrose, types of starch, cellulose derivatives, gelatin, lipids, liposomes, nanoparticles, and the like.
  • Carriers also include physiologically compatible liquids as solvents or for suspensions, including, for example, sterile solutions of water for injection (WFI), saline solution, dextrose solution, Hank's solution, Ringer's solution, vegetable oils, mineral oils, animal oils, polyethylene glycols, liquid paraffin, and the like.
  • WFI water for injection
  • Excipients may also include, for example, colloidal silicon dioxide, silica gel, talc, magnesium silicate, calcium silicate, sodium aluminosilicate, magnesium trisilicate, powdered cellulose, macrocrystalline cellulose, carboxymethyl cellulose, cross-linked sodium carboxymethylcellulose, sodium benzoate, calcium carbonate, magnesium carbonate, stearic acid, aluminum stearate, calcium stearate, magnesium stearate, zinc stearate, sodium stearyl fumarate, syloid, stearowet C, magnesium oxide, starch, sodium starch glycolate, glyceryl monostearate, glyceryl dibehenate, glyceryl palmitostearate, hydrogenated vegetable oil, hydrogenated cotton seed oil, castor seed oil mineral oil, polyethylene glycol (e.g.
  • PEG 4000-8000 polyoxyethylene glycol
  • poloxamers povidone
  • crospovidone croscarmellose sodium
  • alginic acid casein
  • methacrylic acid divinylbenzene copolymer sodium docusate
  • cyclodextrins e.g. 2-hydroxypropyl-.delta.-cyclodextrin
  • polysorbates e.g.
  • polysorbate 80 cetrimide
  • TPGS d-alpha-tocopheryl polyethylene glycol 1000 succinate
  • magnesium lauryl sulfate sodium lauryl sulfate
  • polyethylene glycol ethers di-fatty acid ester of polyethylene glycols
  • a polyoxyalkylene sorbitan fatty acid ester e.g., polyoxyethylene sorbitan ester Tween®
  • polyoxyethylene sorbitan fatty acid esters sorbitan fatty acid ester, e.g.
  • a fatty acid such as oleic, stearic or palmitic acid
  • mannitol xylitol
  • sorbitol maltose
  • lactose
  • oral administration may be used.
  • Pharmaceutical preparations for oral use can be formulated into conventional oral dosage forms such as capsules, tablets, and liquid preparations such as syrups, elixirs, and concentrated drops.
  • Compounds described herein may be combined with solid excipients, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain, for example, tablets, coated tablets, hard capsules, soft capsules, solutions (e.g. aqueous, alcoholic, or oily solutions) and the like.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, glucose, sucrose, mannitol, or sorbitol; cellulose preparations, for example, corn starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose (CMC), and/or polyvinylpyrrolidone (PVP: povidone); oily excipients, including vegetable and animal oils, such as sunflower oil, olive oil, or codliver oil.
  • fillers such as sugars, including lactose, glucose, sucrose, mannitol, or sorbitol
  • cellulose preparations for example, corn starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose (CMC), and/or polyvinylpyrrolidone (PVP: povid
  • the oral dosage formulations may also contain disintegrating agents, such as the cross-linked polyvinylpyrrolidone, agar, or alginic acid, or a salt thereof such as sodium alginate; a lubricant, such as talc or magnesium stearate; a plasticizer, such as glycerol or sorbitol; a sweetening such as sucrose, fructose, lactose, or aspartame; a natural or artificial flavoring agent, such as peppermint, oil of wintergreen, or cherry flavoring; or dye-stuffs or pigments, which may be used for identification or characterization of different doses or combinations. Also provided are dragee cores with suitable coatings.
  • concentrated sugar solutions may be used, which may optionally contain, for example, gum arabic, talc, poly-vinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • compositions that can be used orally include push-fit capsules made of gelatin (“gelcaps”), as well as soft, sealed capsules made of gelatin, and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • injection parenteral administration
  • Compounds described herein for injection may be formulated in sterile liquid solutions, preferably in physiologically compatible buffers or solutions, such as saline solution, Hank's solution, or Ringer's solution.
  • Dispersions may also be prepared in non-aqueous solutions, such as glycerol, propylene glycol, ethanol, liquid polyethylene glycols, triacetin, and vegetable oils. Solutions may also contain a preservative, such as methylparaben, propylparaben, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • the compounds may be formulated in solid form, including, for example, lyophilized forms, and redissolved or suspended prior to use.
  • transmucosal, topical or transdermal administration may be used.
  • penetrants appropriate to the barrier to be permeated are used.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, bile salts and fusidic acid derivatives.
  • detergents may be used to facilitate permeation.
  • Transmucosal administration for example, may be through nasal sprays or suppositories (rectal or vaginal).
  • Compositions of compounds described herein for topical administration may be formulated as oils, creams, lotions, ointments, and the like by choice of appropriate carriers known in the art.
  • Suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohol (greater than C 12 ). In some embodiments, carriers are selected such that the active ingredient is soluble. Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired. Creams for topical application are preferably formulated from a mixture of mineral oil, self-emulsifying beeswax and water in which mixture the active ingredient, dissolved in a small amount of solvent (e.g., an oil), is admixed.
  • solvent e.g., an oil
  • administration by transdermal means may comprise a transdermal patch or dressing such as a bandage impregnated with an active ingredient and optionally one or more carriers or diluents known in the art.
  • a transdermal patch or dressing such as a bandage impregnated with an active ingredient and optionally one or more carriers or diluents known in the art.
  • the dosage administration will be continuous rather than intermittent throughout the dosage regimen.
  • compounds are administered as inhalants.
  • Compounds described herein may be formulated as dry powder or a suitable solution, suspension, or aerosol.
  • Powders and solutions may be formulated with suitable additives known in the art.
  • powders may include a suitable powder base such as lactose or starch, and solutions may comprise propylene glycol, sterile water, ethanol, sodium chloride and other additives, such as acid, alkali and buffer salts.
  • Such solutions or suspensions may be administered by inhaling via spray, pump, atomizer, or nebulizer, and the like.
  • corticosteroids such as fluticasone proprionate, beclomethasone dipropionate, triamcinolone acetonide, budesonide, and mometasone furoate
  • beta agonists such as albuterol, salmeterol, and formoterol
  • anticholinergic agents such as ipratroprium bromide or tiotropium
  • vasodilators such as treprostinal and iloprost
  • enzymes such as DNAase; therapeutic proteins; immunoglobulin antibodies
  • an oligonucleotide such as single or double stranded DNA or RNA, siRNA
  • antibiotics such as tobramycin
  • muscarinic receptor antagonists leukotriene antagonists
  • cytokine antagonists protease inhibitors
  • cromolyn sodium nedocril sodium
  • sodium cromoglycate for example corticosteroids such as fluticasone proprionate, beclomethasone dipropionat
  • the amounts of various compounds to be administered can be determined by standard procedures taking into account factors such as the compound activity (in vitro, e.g. the compound IC 50 vs. target, or in vivo activity in animal efficacy models), pharmacokinetic results in animal models (e.g. biological half-life or bioavailability), the age, size, and weight of the subject, and the disorder associated with the subject. The importance of these and other factors are well known to those of ordinary skill in the art. Generally, a dose will be in the range of about 0.01 to 50 mg/kg, also about 0.1 to 20 mg/kg of the subject being treated. Multiple doses may be used.
  • the compounds described herein may also be used in combination with other therapies for treating the same disease.
  • Such combination use includes administration of the compounds and one or more other therapeutics at different times, or co-administration of the compound and one or more other therapies.
  • dosage may be modified for one or more of the compounds of the invention or other therapeutics used in combination, e.g., reduction in the amount dosed relative to a compound or therapy used alone, by methods well known to those of ordinary skill in the art.
  • use in combination includes use with other therapies, drugs, medical procedures etc., where the other therapy or procedure may be administered at different times (e.g. within a short time, such as within hours (e.g. 1, 2, 3, 4-24 hours), or within a longer time (e.g. 1-2 days, 2-4 days, 4-7 days, 1-4 weeks)) than a compound described herein, or at the same time as a compound described herein.
  • Use in combination also includes use with a therapy or medical procedure that is administered once or infrequently, such as surgery; along with a compound described herein administered within a short time or longer time before or after the other therapy or procedure.
  • the present invention provides for delivery of a compound described herein and one or more other drug therapeutics delivered by a different route of administration or by the same route of administration.
  • the use in combination for any route of administration includes delivery of a compound described herein and one or more other drug therapeutics delivered by the same route of administration together in any formulation, including formulations where the two compounds are chemically linked in such a way that they maintain their therapeutic activity when administered.
  • the other drug therapy may be co-administered with a compound described herein.
  • Use in combination by co-administration includes administration of co-formulations or formulations of chemically joined compounds, or administration of two or more compounds in separate formulations within a short time of each other (e.g.
  • Co-administration of separate formulations includes co-administration by delivery via one device, for example the same inhalant device, the same syringe, etc., or administration from separate devices within a short time of each other.
  • Co-formulations of a compound described herein and one or more additional drug therapies delivered by the same route includes preparation of the materials together such that they can be administered by one device, including the separate compounds combined in one formulation, or compounds that are modified such that they are chemically joined, yet still maintain their biological activity.
  • Such chemically joined compounds may have a linkage that is substantially maintained in vivo, or the linkage may break down in vivo, separating the two active components.
  • composition comprising a pharmaceutically acceptable carrier; and a compound according to Embodiment 1.
  • a kit comprising a compound according to Embodiment 1 or a composition according to Embodiment 2.
  • composition comprising a pharmaceutically acceptable carrier; and a compound according to Embodiment 4.
  • a kit comprising a compound according to Embodiment 4 or a composition according to Embodiment 5.
  • composition comprising a pharmaceutically acceptable carrier; and a compound according to Embodiment 7.
  • a kit comprising a compound according to Embodiment 7 or a composition according to Embodiment 8.
  • a composition comprising a pharmaceutically acceptable carrier; and a compound according to Embodiment 10.
  • a kit comprising a compound according to Embodiment 10 or a composition according to Embodiment 11.
  • composition comprising a pharmaceutically acceptable carrier; and a compound according to Embodiment 13.
  • a kit comprising a compound according to Embodiment 13.
  • a kit comprising a composition according to Embodiment 14.
  • a compound according to any one of Embodiments I, 4, 7, 10 or 13, or a compound listed on Table I for the preparation of a medicament for treating one or more indications selected from the group consisting of melanoma, glioma, glioblastoma multiforme, pilocytic astrocytoma, colorectal cancer, thyroid cancer, lung cancer, ovarian cancer, prostate cancer, liver cancer, gallbladder cancer, gastrointestinal stromal tumors, biliary tract cancer, testicular cancer, and cholangiocarcinoma in a subject in need thereof.
  • a method for treating melanoma in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating glioma in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating glioblastoma multiforme in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating pilocytic astrocytoma comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating colorectal cancer comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating thyroid cancer in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating, lung cancer in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating ovarian cancer in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating prostate cancer in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating liver cancer in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating gallbladder cancer in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating gastrointestinal stromal tumors in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating biliary tract cancer in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating cholangiocarcinoma in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table 1.
  • a method for treating acute pain in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table 1.
  • a method for treating chronic pain in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating polycystic kindney disease in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • histiocytic lymphoma neurofibromatosis, gastrointestinal stromal tumors, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuroendocrine tumors such as medullary thyroid cancer, carcinoid, small cell lung cancer, Kaposi's sarcoma, and pheochromocytoma; pain of neuropathic or inflammatory origin, including, but not limited to, acute pain, chronic pain, cancer-related pain, and migraine; cardiovascular diseases including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g.
  • inflammation and/or proliferation including, but not limited to, psoriasis, eczema, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease (IBD); immunodeficiency diseases, including, but not limited to, organ transplant rejection, graft versus host disease, and Kaposi's sarcoma associated with HIV; renal, cystic, or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia, polycystic liver disease, tuberous sclerosis, Von Hippel Lindau disease, medullary cystic kidney disease, nephronophthisis, and cystic fibrosis; metabolic disorders, including, but not limited to, obesity
  • a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I for the preparation of a medicament for treating one or more indications selected from the group consisting of multi-infarct dementia, head injury, spinal cord injury, Alzheimer's disease (AD), Parkinson's disease, seizures and epilepsy; neoplastic diseases including, but not limited to, melanoma. glioma, glioblastoma multiforme, pilocytic astrocytoma sarcoma, carcinoma (e.g.
  • lymphoma e.g.
  • histiocytic lymphoma neurofibromatosis, gastrointestinal stromal tumors, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuroendocrine tumors such as medullary thyroid cancer, carcinoid, small cell lung cancer, Kaposi's sarcoma, and pheochromocytoma; pain of neuropathic or inflammatory origin, including, but not limited to, acute pain, chronic pain, cancer-related pain, and migraine; cardiovascular diseases including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g.
  • inflammation and/or proliferation including, but not limited to, psoriasis, eczema, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease (IBD); immunodeficiency diseases, including, but not limited to, organ transplant rejection, graft versus host disease, and Kaposi's sarcoma associated with HIV; renal, cystic, or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease.
  • nephrosclerosis glomerulonephritis, prostate hyperplasia, polycystic liver disease, tuberous sclerosis, Von Hippel Lindau disease, medullary cystic kidney disease, nephronophthisis, and cystic fibrosis; metabolic disorders, including, but not limited to, obesity; infection, including, but not limited to Helicobacter pylori, Hepatitis and Influenza viruses, fever, HIV and sepsis; pulmonary diseases including, but not limited to, chronic obstructive pulmonary disease (COPD) and acute respiratory distress syndrome (ARDS); genetic developmental diseases, including, but not limited to, Noonan's syndrome, Costello syndrome, (faciocutaneoskeletal syndrome), LEOPARD syndrome, cardio-faciocutaneous syndrome (CFC), and neural crest syndrome abnormalities causing cardiovascular, skeletal, intestinal, skin, hair and endocrine diseases; and diseases associated with muscle regeneration or degeneration, including, but not limited to, sarcopenia, muscular dystrophies (including, but
  • Distal and Congenital Muscular Dystrophies include motor neuron diseases (including, but not limited to, amyotrophic lateral sclerosis, infantile progressive spinal muscular atrophy, intermediate spinal muscular atrophy, juvenile spinal muscular atrophy, spinal bulbar muscular atrophy, and adult spinal muscular atrophy), inflammatory myopathies (including, but not limited to, dermatomyositis, polymyositis, and inclusion body myositis), diseases of the neuromuscular junction (including, but not limited to, myasthenia gravis, Lambert-Eaton syndrome, and congenital myasthenic syndrome), myopathies due to endocrine abnormalities (including, but not limited to, hyperthyroid myopathy and hypothyroid myopathy) diseases of peripheral nerve (including, but not limited to, Charcot-Marie-Tooth disease, Dejerine-Sottas disease, and Friedreich's ataxia), other myopathies (including, but not limited to, myotonia congenita, paramyotonia congenita, central core
  • lymphoma e.g.
  • histiocytic lymphoma neurofibromatosis, gastrointestinal stromal tumors, acute myeloid leukemia, myelodysplastic syndrome, leukemia, tumor angiogenesis, neuroendocrine tumors such as medullary thyroid cancer, carcinoid, small cell lung cancer, Kaposi's sarcoma, and pheochromocytoma; pain of neuropathic or inflammatory origin, including, but not limited to, acute pain, chronic pain, cancer-related pain, and migraine; cardiovascular diseases including, but not limited to, heart failure, ischemic stroke, cardiac hypertrophy, thrombosis (e.g.
  • inflammation and/or proliferation including, but not limited to, psoriasis, eczema, arthritis and autoimmune diseases and conditions, osteoarthritis, endometriosis, scarring, vascular restenosis, fibrotic disorders, rheumatoid arthritis, inflammatory bowel disease (IBD); immunodeficiency diseases, including, but not limited to, organ transplant rejection, graft versus host disease, and Kaposi's sarcoma associated with HIV; renal, cystic, or prostatic diseases, including, but not limited to, diabetic nephropathy, polycystic kidney disease, nephrosclerosis, glomerulonephritis, prostate hyperplasia, polycystic liver disease, tuberous sclerosis, Von Hippel Lindau disease, medullary cystic kidney disease, nephronophthisis, and cystic fibrosis; metabolic disorders, including, but not limited to, obesity
  • a method for treating testicular cancer in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating Noonan's syndrome in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • a method for treating cardio-faciocutaneous syndrome (CFC) in a subject in need thereof comprising administering to said subject an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, or a compound listed on Table I.
  • CFC cardio-faciocutaneous syndrome
  • CFC cardio-faciocutaneous syndrome
  • a compound or composition according to any one of the preceeding Embodiments wherein said compound inhibits proliferation of a mutant Ras cell line with an IC 50 of less than 20 nM and wherein said compound is a pan Raf inhibitor having an IC 50 of less than 500 nM in activity assays for each of B-Raf, c-Raf-1 and B-Raf V600E protein kinases.
  • a compound or composition according to any one of the preceeding Embodiments wherein said compound inhibits proliferation of a a N-Ras mutant cell line with an IC 50 of less than 1 mM and wherein said compound is a pan Raf inhibitor having an IC 50 of less than 500 nM in activity assays for each of B-Raf, c-Raf-1 and B-Raf V600E protein kinases.
  • a compound or composition according to any one of the preceeding Embodiments wherein said compound inhibits proliferation of a mutant Ras cell line selected from the group consisting of M244, M202, M207, SK-MEL-2, SK-MEL-173, and IPC298 with an IC 50 of less than 1 ⁇ M and wherein said compound is a pan Raf inhibitor having an IC 50 of less than 500 nM in activity assays for each of B-Raf, c-Raf-1 and B-Raf V600E protein kinases.
  • a compound or composition according to any one of the preceeding Embodiments wherein said compound inhibits proliferation of a mutant Ras cell line selected from the group consisting of M244, M202, M207, SK-MEL-2, SK-MEL-173, and IPC298 with an IC 50 of less than 100 nM and wherein said compound is a pan Raf inhibitor having an IC 50 of less than 500 nM in activity assays for each of B-Raf, c-Raf-1 and B-Raf V600E protein kinases.
  • a method for treating a subject suffering from or at risk of a disease or condition comprising administering to the subject in need thereof an effective amount of a compound according to any one of Embodiments 1, 4, 7, 10 or 13, wherein the disease or condition is selected from the group consisting of melanoma, glioma, glioblastoma, pilocytic astrocytoma, liver cancer, biliary tract cancer, cholangiocarcinoma, colorectal cancer, lung cancer, bladder cancer, gallbladder cancer, breast cancer, pancreatic cancer, thyroid cancer, kidney cancer, ovarian cancer, adrenocortical cancer, prostate cancer, gastrointestinal stromal tumors, medullary thyroid cancer, tumor angiogenesis, acute myeloid leukemia, chronic myelomonocytic leukemia, childhood acute lymphoblastic leukemia, plasma cell leukemia, and multiple myeloma.
  • a disease or condition selected from the group consisting of melanoma, glioma, glioblastoma, pilocytic astrocytoma, liver cancer, biliary tract cancer,
  • a disease or condition selected from the group consisting of melanoma, glioma, glioblastoma, pilocytic astrocytoma, liver cancer, biliary tract cancer, cholangiocarcinoma, colorectal cancer, lung cancer, bladder cancer, gallbladder cancer, breast cancer, pancreatic cancer, thyroid cancer, kidney cancer, ovarian cancer, adrenocortical cancer, prostate cancer, gastrointestinal stromal tumors, medullary thyroid cancer, tumor angiogenesis, acute myeloid leukemia, chronic myelomonocytic leukemia, childhood acute lymphoblastic leukemia, plasma cell leukemia, and multiple myeloma.
  • P-0001 Propane-1-sulfonic acid ⁇ 2,4-difluoro-3-[5-(1H-indazol-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl ⁇ -amide P-0001 is prepared in eight steps from 2,4-difluoro-phenylamine 1 as shown in Scheme 1.
  • diisopropylamine 24.87 g, 250 mmol
  • 160 mL of anhydrous tetrahydrofuran 160 mL
  • n-Butyllithium 154 mL, 1.6 M in hexanes, 250 mmol
  • (2,4-difluoro-phenyl)-carbamic acid methyl ester 2, 20.00 g, 110 mmol
  • 40 mL of tetrahydrofuran is added dropwise, maintaining the temperature below ⁇ 70° C.
  • the reaction is stirred at ⁇ 78° C. for 1 hour, then warmed to room temperature and stirred for 1 minute.
  • the reaction is poured into 250 mL of water, the pH is adjusted to 1 with addition of 2M hydrochloric acid, and the mixture is extracted with 3 ⁇ 250 mL of ethyl acetate. The organic layers are combined, dried over magnesium sulfate, filtered and the filtrate concentrated under vacuum. The resulting material is slurried in hexane and the resulting solid is collected by filtration and dried to provide the desired compound (3, 19.38 g, 84%).
  • Step 3 Preparation of ⁇ 3-[(5-bromo-1H-pyrrolo[2,3-b]pyridin-3-yl)-hydroxy-methyl]-2,4-difluoro-phenyl ⁇ -carbamic acid methyl ester (5)
  • the resulting material is purified by silica gel column chromatography, eluting with a gradient of ethyl acetate:hexane. Appropriate fractions are combined and concentrated under vacuum to provide the desired compound (5, 1.91 g, 91%).
  • aqueous layers are combined and back extracted with 1 ⁇ 500 mL of ethyl acetate.
  • the organic layers are combined and washed with 1 ⁇ 500 mL of saturated aqueous sodium bicarbonate, dried over magnesium sulfate, filtered and the filtrate concentrated under vacuum to provide the desired compound (7, 13.34 g).
  • the reaction is stirred at room temperature another 3 hours, then diluted with 500 mL of water and extracted 3 ⁇ 250 mL with dichloromethane. The organic layers are combined and washed with 250 mL of brine, dried over magnesium sulfate, filtered and the filtrate concentrated under vacuum. The resulting material is purified by silica gel column chromatography, eluting with a gradient of ethyl acetate:hexane. Appropriate fractions are combined and concentrated under vacuum to provide the desired compound (9, 8.41 g, 50%).
  • propane-1-sulfonic acid[3-(5-bromo-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide (9, 8.40 g, 18.3 mmol) is combined with 100 mL of toluene, 4-dimethylaminopyridine (447 mg, 3.7 mmol), diisopropylethylamine (4.74 g, 36.7 mmol) and 2,6-dichloro-benzoyl chloride (10, 4.34 g, 20.7 mmol).
  • the reaction is stirred at room temperature overnight, then diluted with 300 mL of dichloromethane and washed with 200 mL of saturated aqueous sodium bicarbonate, then 200 mL of brine.
  • the organic layer is dried over magnesium sulfate, filtered and the filtrate concentrated under vacuum.
  • the resulting material is purified by silica gel column chromatography, eluting with a gradient of ethyl acetate:hexane. Appropriate fractions are combined to provide crude material that is further purified with additional chromatography, eluting with 1:1 hexane:dichloromethane up to 100% dichloromethane. Appropriate fractions are combined and concentrated under vacuum to provide the desired compound (11, 4.15 g, 36%).
  • Step 8 Preparation of propane-1-sulfonic acid(2,4-difluoro-3-[5-(1H-indazol-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbony]-phenyl)-amide (P-0001)
  • Step 8a Preparation of propane-1-sulfonic acid ⁇ 3-[1-(2,6-dichloro-benzoyl)-5-(1H-indol-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl ⁇ -amide (P-0017)
  • Propane-1-sulfonic acid ⁇ 2,4-difluoro-3-[5-(1H-indazol-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl ⁇ -amide P-0001 is further reacted according to the following step 9 to provide propane-1-sulfonic acid ⁇ 3-[5-(1-acetyl-1H-indazol-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl ⁇ -amide P-0007.
  • Step 9 Preparation of propane-1-sulfonic acid ⁇ 3-[5-(1-acetyl-1H-indazol-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl ⁇ -amide (P-0007)
  • the resulting material is purified by silica gel column chromatography, eluting with a gradient of 20-100% ethyl acetate (with 2% acetic acid) in hexanes. Appropriate fractions are combined and concentrated under vacuum, and the resulting solid is further washed with a mixture of ethyl acetate:hexanes to provide the desired compound.
  • MS (ESI) [M+H + ] + 538.0.
  • Propane-1-sulfonic acid ⁇ 2,4-difluoro-3-[5-(1H-indazol-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl ⁇ -amide P-0001 is alternatively further reacted according to the following step 9a to provide propane-1-sulfonic acid ⁇ 2,4-difluoro-3-[5-(1-methanesulfonyl-1H-indazol-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl ⁇ -amide P-0016.
  • Step 9a Preparation of propane-1-sulfonic acid ⁇ 2,4-difluoro-3-[5-(1-methanesulfonyl-1H-indazol-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl ⁇ -amide (P-0016)
  • 5-Bromo-1H-pyrrolo[2,3-b]pyridine (4, 0.525 g, 2.66 mmol) is combined with aluminum trichloride (2.05 g, 15.4 mmol) and 10 mL of nitromethane.
  • 2-Fluoro-3-nitro-benzoyl chloride (17, 0.521 g, 2.56 mmol) in 5 mL of nitromethane is added and the reation is heated at 45° C. for 6 hours.
  • the reaction is quenched by slowly adding 5 mL of methanol.
  • the resulting precipitate is collected by filtration and combined with ethyl acetate, water and celite, then stirred for 30 minutes and filtered.
  • N-[3-(5-bromo-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2-fluoro-phenyl]-2-fluoro-benzenesulfonamide (21, 100 mg, 0.203 mmol)
  • 2-methoxy-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-pyrimidine (22, 72 mg, 0.305 mmol)
  • tetrakis-(triphenylphosphine)palladium(0) 10 mg
  • the reaction mixture is heated in a microwave at 160° C. for 15 minutes.
  • the mixture is filtered through celite, and the celite is washed with water and ethyl acetate.
  • the aqueous layer is separated from the filtrate and extracted with ethyl acetate.
  • the organic portions are combined and washed with brine, then dried over sodium sulfate, filtered and the filtrate concentrated under vacuum.
  • the resulting material is purified by silica gel column chromatography, eluting with methanol and dichloromethane. Appropriate fractions are combined and concentrated under vacuum to provide the desired compound (P-1518, 27 mg).
  • MS (ESI) [M+H + ] + 521.95.
  • Propane-1-sulfonic acid ⁇ 2,4-difluoro-3-[5-(1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl ⁇ -amide 23 is prepared according to Scheme 1a, replacing N-[3-(5-bromo-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2-fluoro-phenyl]-2-fluoro-benzenesulfonamide 21 with propane-1-sulfonic acid [3-(5-bromo-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide and replacing 2-methoxy-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-pyrimidine 22 with 1H-pyrazole-4-carboxylic acid.
  • Step 1 Preparation of 4- ⁇ 3-[2,6-difluoro-3-(propane-1-sulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl)-pyrazole-1-carboxylic acid phenyl ester (P-1016)
  • P-0005 N- ⁇ 3-[5-(1-acetyl-1H-indazol-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl ⁇ -2,5-difluoro-benzenesulfonamide
  • P-0005 is prepared in three steps from (3-amino-2,6-difluoro-phenyl)-(5-bromo-1H-pyrrolo[2,3-b]pyridin-3-yl)-methanone 7 and 2,5-difluoro-benzenesulfonyl chloride 15 as shown in Scheme 2.
  • Step 1 Preparation of N-[3-(5-bromo-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-2,5-difluoro-benzenesulfonamide (26)
  • Step 2 Preparation of N-(2,4-difluoro-3-[5-(1H-indazol-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl ⁇ -2,5-difluoro-benzenesulfonamide (P-0002)
  • Step 3 Preparation of N- ⁇ 3-[5-(1-acetyl-1H-indazol-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl ⁇ -2,5-difluoro-benzenesulfonamide (P-0005)
  • P-0002 is alternatively reacted according to the following step 3a to provide 5- ⁇ 3-[3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl ⁇ -indazole-1-carboxylic acid methylamide P-0003.
  • Step 3a Preparation of 5- ⁇ 3-[3-(2,5-difluoro-benzenesulfonylamino)-2,6-difluoro-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl ⁇ -indazole-1-carboxylic acid methylamide (P-0003)
  • methyl isocyanate (6.5 mg, 0.11 mmol) is combined with N- ⁇ 2,4-difluoro-3-[5-(1H-indazol-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl ⁇ -2,5-difluoro-benzenesulfonamide (P-0002, 22 mg, 0.031 mmol), tetrakis(triphenylphosphine)palladium(0) (2.2 mg, 0.0019 mmol), potassium carbonate (1.0 mL, 1.0 M in water) and 3 mL of acetonitrile. The reaction is heated at 140° C.
  • P-0008 is prepared in three steps from 5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-pyrrolo[2,3-b]pyridine 17 and propane-1-sulfonic acid(2,4-difluoro-3-formyl-phenyl)-amide 18 as shown in Scheme 3.
  • propane-1-sulfonic acid(2,4-difluoro-3- ⁇ hydroxy-[5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-methyl ⁇ -phenyl)-amide (29, 0.155 g, 0.306 mmol) is combined with 10 mL of tetrahydrofuran and 3 mL of dichloromethane.
  • Dess-Martin periodinane (0.648 g, 1.53 mmol) is added and the reaction is stirred at room temperature for 20 minutes.
  • Additional compounds such as propane-1-sulfonic acid ⁇ 2,4-difluoro-3-[5-(1-pyrimidin-2-yl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl ⁇ -amide P-1005, are prepared following the protocol of steps 3a and 4 of Scheme 3a (steps 1 and 2 are the same as Scheme 3).
  • Step 3 Preparation of propane-1-sulfonic acid ⁇ 3-[1-(2,6-dichloro-benzoyl)-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl ⁇ -amide (32)
  • the reaction is stirred at room temperature for 17 hours, then diluted with 100 mL of ethyl acetate and extracted.
  • the organic layer is washed sequentially with 40 mL of saturated aqueous sodium bicarbonate, 40 mL of water, and 40 mL of brine, then dried with sodium sulfate, filtered and the filtrate concentrated under vacuum.
  • the resulting material is purified by silica gel column chromatography, eluting with a gradient of ethyl acetate and hexane. Appropriate fractions are combined and concentrated under vacuum and the resulting material is triturated with diethyl ether to provide the desired compound.
  • Step 4 Preparation of propane-1-sulfonic acid ⁇ 2,4-difluoro-3-[5-(1-pyrimidin-2-yl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl ⁇ -amide (P-1005)
  • the reaction mixture is microwave irradiated at 145° C. for 15 minutes, then neutralized with acetic acid, and extracted with 2 ⁇ 1 mL of ethyl acetate.
  • the combined organic layer is concentrated under vacuum, the residue dissolved in 500 ⁇ L of dimethylsulfoxide and purified by HPLC using a C18 column, eluting with water/acetonitrile and 0.1% trifluoroacetic acid, 20-100% acetonitrile at 6 mL per minute. Appropriate fractions are combined and concentrated under vacuum to provide the desired compound.
  • MS (ESI) [M+H + [ + 524.5.
  • P-1002 is prepared in two steps from propane-1-sulfonic acid[3-(5-bromo-1H-pyrrolo[2,3-b]pyridine-3-carbonyl)-2,4-difluoro-phenyl]-amide 9 and 4-[4-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-pyrazol-1-yl]-piperidine-1-carboxylic acid tert-butyl ester 34 as shown in Scheme 4.
  • Step 1 Preparation of 4-(4- ⁇ 3-[2,6-difluoro-3-(propane-1-sulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl ⁇ -pyrazol-1-yl)-piperidine-1-carboxylic acid tert-butyl ester (P-1001)
  • Step 2 Preparation of propane-1-sulfonic acid ⁇ 2,4-difluoro-3-[5-(1-piperidin-4-yl-1H-pyrazol-4-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl ⁇ -amide (P-1002)
  • P-1004 is isolated after step 2, suspending in acetonitrile water instead of tetrahydrofuran, and the resulting precipitate is isolated and washed with water to provide the desired compound (P-1004, 121 mg).
  • MS (ESI) [M+H + ] + 631.0.
  • P-1800 is prepared in two steps from 1H-pyrrolo[2,3-b]pyridin-5-ylamine 35 and nicotinoyl chloride 36 as shown in Scheme 5.
  • Step 2 Preparation of N- ⁇ 3-[2,6-difluoro-3-(propane-1-sulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl ⁇ -nicotinamide (P-1800)
  • N-(1H-pyrrolo[2,3-b]pyridin-5-yl)-nicotinamide 37, 0.050 g, 0.21 mmol
  • Aluminum trichloride 0.1 g, 1 mmol
  • 2,6-difluoro-3-(propane-1-sulfonylamino)-benzoyl chloride 38, 0.075 g, 0.25 mmol
  • the reaction is stirred at room temperature for 36 hours, then diluted with water and extracted 3 ⁇ with ethyl acetate.
  • Step 2a Preparation of 5-methyl-2,3-dihydro-isoxazole-3-carboxylic acid ⁇ 3-[2,6-difluoro-3-(propane-1-sulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridin-5-yl)-amide (P-1802)
  • Step 1 Preparation of 3-[2,6-difluoro-3-(propane-1-sulfonylamino)-benzoyl]-1H-pyrrolo[2,3-b]pyridine-5-carboxylic acid pyridin-3-ylamide (P-1801)
  • P-1520 is prepared in five steps from 5-iodo-1H-pyrrolo[2,3-b]pyridine 43 and 2,6-difluoro-3-nitro-benzoyl chloride 44 as shown in Scheme 7.
  • Step 1 Preparation of (2,6-difluoro-3-nitro-phenyl)-(5-iodo-1H-pyrrolo[2,3-b]pyridin-3-yl)-methanone (45)
  • Step 2 Preparation of (3-amino-2,6-difluoro-phenyl)-(5-iodo-1H-pyrrolo[2,3-b]pyridin-3-yl)-methanone (46)
  • Step 3 Preparation of (3-amino-2,6-difluorophenyl)-[5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]-methanone (48)
  • Step 4 Preparation of (3-amino-2,6-difluoro-phenyl)-[5-(2-methyl-pyrimidin-5-yl)-1H-pyrrolo[2,3-b]pyridin-3-yl]methanone (50)
  • reaction is heated at 100° C. overnight. Upon cooling, the reaction mixture is partitioned between water/brine and ethyl acetate/tetrahydrofuran and then filtered through Celite. The organic layer is separated and concentrated under vacuum. The resulting residue is purified by silica gel column chromatography eluting with 0-10% methanol in dichloromethane to provide the desired compound as a tan solid (50, ⁇ 220 mg).
  • Step 5 Preparation of N- ⁇ 2,4-difluoro-3-[5-(2-methyl-pyrimidin-5-yl)-1H-pyrrolo[2,3-b]pyridine-3-carbonyl]-phenyl ⁇ -2-fluoro-benzenesulfonamide (P-1520)
  • the compounds described herein show favorable properties that provide advantages as a pharmaceutical as well.
  • compounds may show favorable solubility, favorable pharmacokinetic properties, and low Cyp inhibition. The compounds are assessed in the following assays or similar assays available to one skilled in the art.
  • biochemical activity IC 50 values are determined with respect to inhibition of B-Raf kinase activity, c-Raf-1 kinase activity, or B-Raf V600E kinase activity, where inhibition of phosphorylation of a peptide substrate is measured as a function of compound concentration.
  • Compounds to be tested are diluted in dimethyl sulfoxide to a concentration of 0.1 mM.
  • the reaction is started with addition of 10 ⁇ L of 200 ⁇ M ATP (i.e. final 100 ⁇ M ATP).
  • Stop Solution After incubation of the kinase reaction for 45 minutes at room temperature, 5 ⁇ L/well of Stop Solution is added (25 mM Hepes pH 7.5, 100 mM EDTA, 0.01% BSA with donor beads (Streptavidin coated beads, Perkin Elmer), acceptor beads (Protein A coated, Perkin Elmer), and anti phosphor MEK1/2 antibody (CellSignal), each at final concentration 10 ⁇ g/mL). The plates are incubated for 3 hours at room temperature and read on Envision reader (Perkin Elmer). Phosphorylation of Mek1 results in binding of the anti-phosphor-MEK1/2antibody and association of the donor and acceptor beads such that signal correlates with kinase activity. The signal vs. compound concentration is used to determine the IC 50 .
  • Compounds are assessed in a variety of cell based assays. For example human cell lines with B-Raf V600E mutation (A375 melanoma, SKMEL3 melanoma, and COLO205 colon adenocarcinoma), as well as tumorigenic cell lines with wild-type B-RAF (SW620 colon adenocarcinoma) or with Ras mutations (SKMEL2 melanoma and IPC298 melanoma).
  • Similar assays may be used to assess additional tumorigenic cell lines with Ras mutations, including, but not limited to, M202, M207, M243, M244, M296, S117, HCT116, HCT15, DLD1, MiaPaCa, A549, NCI-H23, NCI-H460, HOP62, MDA-MB231, Hs-578T, HL60, MOLT-4, and CCRF-CEM.
  • the cells are plated, 50 ⁇ L in each well of a 96-well dish (Corning 3610) and incubated at 37° C. in 5% CO 2 overnight, cells plated to a final concentration of cells as follows:
  • compound at a maximum concentration of 5 mM is serially diluted 1:3 (e.g. 10 ⁇ L with 30 ⁇ L dimethyl sulfoxide) for a total of 8 point titration with DMSO as a control.
  • a 1 ⁇ L aliquot of each dilution point and control is added to 249 ⁇ L growth media and 50 ⁇ L it is added to a well containing cells, providing 10 ⁇ M compound at the maximum concentration point.
  • the cells are incubated for 3 days at 37° C. in 5% CO 2 .
  • ATPlite 1 step Luminescence Assay System (Perkin Elmer #6016739) is brought to room temperature along with the cell cultures. ATPlite is added 25 ⁇ L to each well, shake for 2 minutes, and the cells are incubated at room temperature for 10 minutes, then luminescence is read on Safire reader. The measured luminescence correlates directly with cell number, such that the reading as a function of compound concentration is used to determine the IC 50 value.
  • results of these assays may vary as assay conditions are varied. Inhibition levels determined under the conditions described herein represent a relative activity for the compounds tested under the specific conditions employed.
  • the cell based assays are likely to show variability due to the complexity of the system and the sensitivity thereof to any changes in the assay conditions. As such, some level of inhibition in the cell based assays is indicative of the compounds having some inhibitory activity for those cells, whereas lack of inhibition below the threshold of the highest concentration tested does not necessarily indicate that the compound has no inhibitory activity on the cells, only that under the conditions tested, no inhibition is observed.
  • Results for compounds that are tested and show substantially no inhibition below the highest tested concentration are represented as “ ⁇ ” in the tables below. In some instances, the compounds were not tested in all of the assays, or assay results were not valid, as indicated by NA in the tables below.
  • turbidity of compounds in aqueous solutions is assessed.
  • a series of aqueous buffers with varying pH is used.
  • each compound is diluted into four different physiologically relevant buffers and solution turbidity is measured by spectrophotometry.
  • concentration of compound that demonstrates turbidity by forming enough insoluble suspension to raise the average optical density above 0.01 at three wavelengths (490, 535, and 650 nm) is used to define the limit of the compound solubility in that buffer.
  • the buffers used are Simulated Gastric Fluid (SGF-pH 1.5) 0.5M NaCl, pH 1.5; Simulated Intestinal fluid (SIF-pH 4.5 and pH 6.8) 0.05M NaH 2 PO 4 , pH 4.5 and 6.8; and Hepes Buffer (HEPES-pH 7.4) 10 mM HEPES, 150 mM NaCl, pH 7.4. Control compounds pyrene, estriol and propranolol HCl are also assessed. Plates are spun and then mixed for 1 minute, and the absorbance is read using a Tecan Satire II to read wavelengths in the visible range (490, 535, and 650 nm) at four locations per well, reflecting the degree of turbidity present.
  • SGF-pH 1.5 Simulated Gastric Fluid
  • SIF-pH 4.5 and pH 6.8 Simulated Intestinal fluid
  • HEPES-pH 7.4 Hepes Buffer
  • Control compounds pyrene, estriol and propranolol HCl
  • the average optical density for each wavelength in each well is graphed vs. compound concentration, and the concentration at which the curve crosses a threshold O.D. of 0.01 for each wavelength is reported as the endpoint turbidity assay result.
  • the average of the three wavelengths is used to compare turbidity of compounds.
  • Compounds are considered to have low solubility if the threshold concentration is ⁇ 31.3 ⁇ M, moderate solubility if the threshold concentration is 31.3 ⁇ M to 250 ⁇ M, and high solubility if the threshold concentration is >250 ⁇ M.
  • CYP (Cytochrome P450) enzymes are the major drug metabolizing enzymes present in the liver.
  • the inhibition of CYP enzyme activity (IC 50 ) for each of CYP1A2, CYP2C19, CYP2C9, CYP2D6 CYP3A4(BFC) and CYP3A4(BQ) is determined for compounds, where inhibition of metabolism of a known substrate leads to a decrease in the fluorescence of the metabolized product.
  • the fluorescence of the product is monitored as a function of compound concentration.
  • Enzyme and substrate mix (10 ⁇ L; 0.5 pmol CYP1A2/5 ⁇ M CEC; 1.0 pmol CYP2C9/75 ⁇ M MFC; 0.5 pmol CYP2C19/25 ⁇ M CEC; 1.5 pmol CYP2D6/1.5 ⁇ M AMMC; 1.0 pmol CYP3A4/50 ⁇ M BFC; or 1.0 pmol CYP3A4/40 ⁇ M BQ) is added to these assay plates. Assay plates are incubated at 37° C.
  • the signal versus compound concentration is used to determine the IC 50 .
  • the enzymes and substrates for this assay are obtained from BD Biosciences. While other factors are involved in determining CYP effects in vivo, compounds preferably have IC 50 values of >5 ⁇ M, more preferably IC 50 values of >10 ⁇ M.
  • Rats are dosed daily with compound either by IV injections via surgically implanted jugular catheters or by oral gavage (PO).
  • Each compound is prepared as a 20 mg/mL stock solution in dimethyl sulfoxide, which is further diluted to provide the dosing stock at the desired concentration for the IV or PO formulations.
  • IV dosing the dosing stock is diluted into a 1:1:8 mixture of Solutol®:ethanol:water.
  • PO dosing the dosing stock is diluted into 1% methylcellulose.
  • compounds are diluted to 0.5 mg/mL each for IV dosing and 0.4 mg/mL each for PO dosing and dosed at 1 mg/kg (2mL/kg) or 2 mg/kg (5 mL/kg), respectively.
  • IV dosed animals tail vein blood samples are collected with lithium heparin anticoagulant at 5, 15, 30, and 60 minutes and 4, 8, and 24 hours post dosing each day.
  • PO dosed animals tail vein blood samples are collected with lithium heparin anticoagulant at 30 minutes, 1, 2, 4, 8 and 24 hours post dosing each day.
  • Dogs are dosed daily by oral capsules in a suitable formulation at 50 mg/mL.
  • Cephalic vein blood samples are collected with lithium heparin anticoagulant at 30 minutes, 1, 2, 4, 8 and 24 hours post dosing each day. All samples are processed to plasma and frozen for later analysis of each compound by LC/MS/MS. Plasma levels as a function of time are plotted to assess the AUC (ng*hr/mL).
  • Compounds according to the present invention preferably show improved pharmacokinetic properties relative to previously described compounds, i.e. they have substantially higher values for one or more of AUC, Cmax and half-life relative to previously described compounds.
  • Compounds of the invention in combination with a standard chemotherapeutic agent, such as 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, or vinblastine, can be assessed for their effectiveness in killing human tumor cells.
  • a standard chemotherapeutic agent such as 5-fluorouracil, carboplatin, dacarbazine, gefitinib, oxaliplatin, paclitaxel, SN-38, temozolomide, or vinblastine.
  • Additional features of the complex can be used to demonstrate improved properties, such as comparison of the intrinsic dissolution rate of a similarly prepared substantially amorphous citrate complex or formulation thereof as compared to that of a crystalline form of the compound or similar formulation thereof in simulated gastric fluid (SGF) without enzyme and in simulated intestinal fluid (SIF).
  • SGF gastric fluid
  • SIF simulated intestinal fluid
  • a pellet of test sample is dissolved in the appropriate fluid, and the UV absorbance as a function of time is measured at 254 nm (SGF) or 310 nm (SIF) and plotted.
  • any of the terms “comprising”, “consisting essentially of” and “consisting of” may be replaced with either of the other two terms.
  • the invention also includes another embodiment wherein one of these terms is replaced with another of these terms.
  • the terms have their established meaning.
  • one embodiment may encompass a method “comprising” a series of steps, another embodiment would encompass a method “consisting essentially of” the same steps, and a third embodiment would encompass a method “consisting of” the same steps.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Plural Heterocyclic Compounds (AREA)
US12/975,239 2009-12-23 2010-12-21 Compounds and methods for kinase modulation, and indications therefor Abandoned US20110183988A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US12/975,239 US20110183988A1 (en) 2009-12-23 2010-12-21 Compounds and methods for kinase modulation, and indications therefor
TW099145373A TW201132639A (en) 2009-12-23 2010-12-22 Compounds and methods for kinase modulation, and indications therefor
US14/514,232 US9440969B2 (en) 2009-12-23 2014-10-14 Compounds and methods for kinase modulation, and indications therefor

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US28993009P 2009-12-23 2009-12-23
US38331810P 2010-09-15 2010-09-15
US12/975,239 US20110183988A1 (en) 2009-12-23 2010-12-21 Compounds and methods for kinase modulation, and indications therefor

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/514,232 Continuation US9440969B2 (en) 2009-12-23 2014-10-14 Compounds and methods for kinase modulation, and indications therefor

Publications (1)

Publication Number Publication Date
US20110183988A1 true US20110183988A1 (en) 2011-07-28

Family

ID=44196390

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/975,239 Abandoned US20110183988A1 (en) 2009-12-23 2010-12-21 Compounds and methods for kinase modulation, and indications therefor
US14/514,232 Active US9440969B2 (en) 2009-12-23 2014-10-14 Compounds and methods for kinase modulation, and indications therefor

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/514,232 Active US9440969B2 (en) 2009-12-23 2014-10-14 Compounds and methods for kinase modulation, and indications therefor

Country Status (14)

Country Link
US (2) US20110183988A1 (fr)
EP (1) EP2516438A4 (fr)
JP (1) JP5815558B2 (fr)
KR (1) KR20120112623A (fr)
CN (1) CN102753549A (fr)
AR (1) AR079690A1 (fr)
AU (1) AU2010336524B2 (fr)
BR (1) BR112012015745A2 (fr)
CA (1) CA2784393A1 (fr)
MX (1) MX2012007429A (fr)
RU (1) RU2012131373A (fr)
TW (1) TW201132639A (fr)
UY (1) UY33159A (fr)
WO (1) WO2011079133A2 (fr)

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012138809A1 (fr) * 2011-04-05 2012-10-11 Dawei Zhang Utilisation de composés hétérocycliques en tant qu'inhibiteurs des kinases
US20140094611A1 (en) * 2012-05-31 2014-04-03 Plexxikon Inc. Synthesis of heterocyclic compounds
WO2014176268A1 (fr) * 2013-04-22 2014-10-30 Abbvie Inc. Thiazoles et utilisations de ceux-ci
US9096593B2 (en) 2009-11-06 2015-08-04 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9169250B2 (en) 2006-11-22 2015-10-27 Plexxikon Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
US20160002228A1 (en) * 2013-01-18 2016-01-07 Genentech, Inc. 3-substituted pyrazoles and use as dlk inhibitors
US9260437B2 (en) 2008-05-19 2016-02-16 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9358235B2 (en) 2012-03-19 2016-06-07 Plexxikon Inc. Kinase modulation, and indications therefor
US9399639B2 (en) 2012-08-27 2016-07-26 Calitor Sciences, Llc Substituted azaindole compounds, salts, pharmaceutical compositions thereof and methods of use
US9440969B2 (en) 2009-12-23 2016-09-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9447089B2 (en) 2009-04-03 2016-09-20 Plexxikon Inc. Compositions and uses thereof
US9469640B2 (en) 2007-07-17 2016-10-18 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9550768B2 (en) 2011-05-17 2017-01-24 Plexxikon Inc. Kinase modulation and indications therefor
US9617267B2 (en) 2009-11-18 2017-04-11 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9624213B2 (en) 2011-02-07 2017-04-18 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9676748B2 (en) 2012-12-21 2017-06-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9682981B2 (en) 2010-04-21 2017-06-20 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9718847B2 (en) 2013-03-15 2017-08-01 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9771369B2 (en) 2014-03-04 2017-09-26 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9771363B2 (en) 2015-09-21 2017-09-26 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9822109B2 (en) 2013-03-15 2017-11-21 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9856259B2 (en) 2014-09-15 2018-01-02 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9873700B2 (en) 2013-05-30 2018-01-23 Plexxikon Inc. Compounds for kinase modulation, and indications therefor
US9938273B2 (en) 2015-12-07 2018-04-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10160755B2 (en) 2015-04-08 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10160747B2 (en) 2016-03-16 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10227357B2 (en) 2012-09-06 2019-03-12 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10428067B2 (en) 2017-06-07 2019-10-01 Plexxikon Inc. Compounds and methods for kinase modulation
US10435404B2 (en) 2017-07-25 2019-10-08 Plexxikon Inc. Formulations of a compound modulating kinases
US10508085B2 (en) 2016-09-22 2019-12-17 Plexxikon Inc. Compounds and methods for IDO and TDO modulation, and indications therefor
US10577366B2 (en) 2017-03-20 2020-03-03 Plexxikon Inc. Crystalline forms of a compound that inhibits bromodomain
US10703757B2 (en) 2016-12-23 2020-07-07 Plexxikon Inc. Compounds and methods for CDK8 modulation and indications therefor
US10717735B2 (en) 2017-10-13 2020-07-21 Plexxikon Inc. Solid forms of a compound for modulating kinases
US10829484B2 (en) 2015-07-28 2020-11-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US11103505B2 (en) 2017-10-27 2021-08-31 Plexxikon Inc. Formulations of a compound modulating kinases
US11149011B2 (en) 2018-03-20 2021-10-19 Plexxikon Inc. Compounds and methods for IDO and TDO modulation, and indications therefor

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012135631A1 (fr) 2011-03-30 2012-10-04 Arrien Pharmaeuticals Llc 5-(pyrazin-2-yl)-1h-pyrazolo[3,4-b]pyridine substituée et dérivés de pyrazolo[3,4-b]pyridine en tant qu'inhibiteurs de protéine kinase
CN112574196A (zh) 2015-05-06 2021-03-30 普莱希科公司 调节激酶的1H-吡咯并[2,3-b]吡啶衍生物的合成
SI3292123T1 (sl) 2015-05-06 2020-10-30 Plexxikon Inc. Trdne oblike spojine, ki modulira kinaze
BR112017025045A2 (pt) 2015-05-22 2018-08-07 Plexxikon Inc plx-8394 ou plx-7904 para uso no tratamento de doenças relacionadas a braf-v600
CN107771178B (zh) 2015-05-22 2021-04-06 普莱希科公司 杂环化合物的合成
CN110291089B (zh) * 2017-01-17 2022-05-27 海帕瑞吉尼克斯股份有限公司 用于促进肝再生或者减少或预防肝细胞死亡的蛋白激酶抑制剂
WO2019149738A1 (fr) 2018-01-31 2019-08-08 Heparegenix Gmbh Inhibiteurs de protéine kinase mkk4 pour favoriser la régénération hépatique ou pour réduire ou prévenir la mort des hépatocytes
EP3810139A1 (fr) 2018-06-21 2021-04-28 HepaRegeniX GmbH Inhibiteurs de protéine kinase tricycliques pour favoriser la régénération du foie ou réduire ou prévenir la mort des hépatocytes
EP3823968A1 (fr) * 2018-07-16 2021-05-26 HepaRegeniX GmbH Inhibiteurs de protéine kinase permettant de favoriser la régénération du foie, ou de réduire ou de prévenir la mort des hépatocytes
KR20220018475A (ko) 2019-04-09 2022-02-15 플렉시콘 인코퍼레이티드 Ep300 또는 cbp 조절 및 이의 표시를 위한 응축 아진
CN111217654A (zh) * 2020-01-02 2020-06-02 陕西科技大学 钯催化的间位芳基化反应及其在维罗非尼类似物合成中的应用
JP2023522949A (ja) 2020-04-23 2023-06-01 オプナ バイオ ソシエテ アノニム Cd73調節のための化合物及び方法並びにそれらの表示
EP4199926A1 (fr) 2020-08-21 2023-06-28 Plexxikon Inc. Thérapies anticancéreuses à base de médicaments combinatoires

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040077595A1 (en) * 2002-09-06 2004-04-22 Insert Therapeutics, Inc. Cyclodextrin-based polymers for therapeutics delivery
US20100286142A1 (en) * 2009-05-06 2010-11-11 Ibrahim Prabha N Compounds and Methods for Kinase Modulation, and Indications Therefor
US20100310659A1 (en) * 2009-04-03 2010-12-09 Plexxikon, Inc. Compositions and Uses Thereof
US7863289B2 (en) * 2006-12-21 2011-01-04 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
US7863288B2 (en) * 2005-06-22 2011-01-04 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor

Family Cites Families (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040171062A1 (en) 2002-02-28 2004-09-02 Plexxikon, Inc. Methods for the design of molecular scaffolds and ligands
EP1558751A4 (fr) 2002-09-16 2007-08-22 Plexxikon Inc Structure cristalline de la proteine kinase pim-1
US20050048573A1 (en) 2003-02-03 2005-03-03 Plexxikon, Inc. PDE5A crystal structure and uses
EP1627045A2 (fr) 2003-02-28 2006-02-22 Plexxikon, Inc. Structure cristalline de pyk2 et ses utilisations
US20050079548A1 (en) 2003-07-07 2005-04-14 Plexxikon, Inc. Ligand development using PDE4B crystal structures
JP4845730B2 (ja) 2003-07-17 2011-12-28 プレキシコン,インコーポレーテッド Ppar活性化合物
US7348338B2 (en) 2003-07-17 2008-03-25 Plexxikon, Inc. PPAR active compounds
WO2005028624A2 (fr) 2003-09-15 2005-03-31 Plexxikon, Inc. Echafaudages moleculaires pour le developpement de ligands des proteines kinases
US7517970B2 (en) 2003-12-19 2009-04-14 Plexxikon, Inc. Nucleic acids encoding kinase and phosphatase enzymes, expression vectors and cells containing same
US20070066641A1 (en) 2003-12-19 2007-03-22 Prabha Ibrahim Compounds and methods for development of RET modulators
DK1696920T3 (en) 2003-12-19 2015-01-19 Plexxikon Inc RELATIONS AND PROCEDURES FOR THE DEVELOPMENT OF LAW MODULATORS
JP2008503446A (ja) 2004-05-06 2008-02-07 プレキシコン,インコーポレーテッド Pde4b阻害剤及びその使用
JP2008503473A (ja) 2004-06-17 2008-02-07 プレキシコン,インコーポレーテッド C−kit活性を調節する化合物
US7498342B2 (en) 2004-06-17 2009-03-03 Plexxikon, Inc. Compounds modulating c-kit activity
JP2008512380A (ja) 2004-09-03 2008-04-24 プレキシコン,インコーポレーテッド Pde4b阻害剤
AU2005311826A1 (en) 2004-11-30 2006-06-08 Plexxikon, Inc. Indole derivatives for use as PPAR active compounds
US7531568B2 (en) 2004-11-30 2009-05-12 Plexxikon, Inc. PPAR active compounds
US20060160135A1 (en) 2004-12-08 2006-07-20 Weiru Wang SF-1 and LRH-1 modulator development
WO2007013896A2 (fr) 2005-05-17 2007-02-01 Plexxikon, Inc. Composes modulant l'activite de c-kit et c-fms et utilisations
US20080234349A1 (en) 2005-09-07 2008-09-25 Jack Lin PPAR active compounds
TW200800872A (en) 2005-09-07 2008-01-01 Plexxikon Inc PPAR active compounds
US20070072904A1 (en) 2005-09-07 2007-03-29 Jack Lin PPAR active compounds
WO2008063888A2 (fr) 2006-11-22 2008-05-29 Plexxikon, Inc. Composés modulant l'activité de c-fms et/ou de c-kit et utilisations associées
WO2008079909A1 (fr) 2006-12-21 2008-07-03 Plexxikon, Inc. Composés et méthodes de modulation des kinases, et indications connexes
AU2007336811A1 (en) 2006-12-21 2008-07-03 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
EP2121591A2 (fr) 2007-03-08 2009-11-25 Plexxikon, Inc. Composés actifs sur ppar
PE20090159A1 (es) 2007-03-08 2009-02-21 Plexxikon Inc COMPUESTOS DERIVADOS DE ACIDO INDOL-PROPIONICO COMO MODULADORES PPARs
WO2009012283A1 (fr) 2007-07-17 2009-01-22 Plexxikon Inc. Composés et procédés pour la modulation des kinases et leurs indications
JP2009077712A (ja) * 2007-09-11 2009-04-16 F Hoffmann La Roche Ag B−Rafキナーゼ阻害剤に対する感受性についての診断試験
US8158636B2 (en) 2008-05-19 2012-04-17 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
PE20091846A1 (es) 2008-05-19 2009-12-16 Plexxikon Inc DERIVADOS DE PIRROLO[2,3-d]-PIRIMIDINA COMO MODULADORES DE CINASAS
JP5767965B2 (ja) 2008-06-10 2015-08-26 プレキシコン インコーポレーテッドPlexxikon Inc. キナーゼを調節する5h−ピロロ[2,3−b]ピラジン誘導体、およびその適応症
WO2009152087A1 (fr) 2008-06-10 2009-12-17 Plexxikon, Inc. Composés hétéroaryles bicycliques et procédés destinés à la modulation de kinases, et des indications pour celle-ci
KR20110130463A (ko) 2009-03-11 2011-12-05 플렉시콘, 인코퍼레이티드 Raf 키나제의 억제를 위한 피롤로[2,3-b]피리딘 유도체
WO2010104945A1 (fr) * 2009-03-11 2010-09-16 Plexxikon, Inc. Dérivés de pyrrolo [2, 3-b] pyridine pour l'inhibition des kinases raf
WO2010111527A1 (fr) 2009-03-26 2010-09-30 Plexxikon, Inc. Pyrazolo [ 3, 4 -b] pyridines en tant qu'inhibiteurs de la kinase et leur utilisation médicale
SG175877A1 (en) 2009-05-04 2011-12-29 Plexxikon Inc Compounds and methods for inhibition of renin, and indications therefor
JP2013510166A (ja) 2009-11-06 2013-03-21 プレキシコン インコーポレーテッド キナーゼ調節のための化合物、方法およびその適用
JP2013511541A (ja) 2009-11-18 2013-04-04 プレキシコン インコーポレーテッド キナーゼ調節のための化合物および方法、ならびにその適応
CA2784393A1 (fr) 2009-12-23 2011-06-30 Plexxikon, Inc. Composes et methodes pour la modulation des kinases, et leurs indications
TWI619713B (zh) 2010-04-21 2018-04-01 普雷辛肯公司 用於激酶調節的化合物和方法及其適應症
US8642606B2 (en) 2010-09-29 2014-02-04 Plexxikon Inc. ZAP-70 active compounds
AU2012214762B2 (en) 2011-02-07 2015-08-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
TWI558702B (zh) 2011-02-21 2016-11-21 普雷辛肯公司 醫藥活性物質的固態形式
AU2012255275B2 (en) 2011-05-17 2016-01-28 Plexxikon Inc. Kinase modulation and indications therefor
US9358235B2 (en) 2012-03-19 2016-06-07 Plexxikon Inc. Kinase modulation, and indications therefor
US9150570B2 (en) 2012-05-31 2015-10-06 Plexxikon Inc. Synthesis of heterocyclic compounds
US10227357B2 (en) 2012-09-06 2019-03-12 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
SG11201504754QA (en) 2012-12-21 2015-07-30 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor
US20140303121A1 (en) 2013-03-15 2014-10-09 Plexxikon Inc. Heterocyclic compounds and uses thereof
PE20151997A1 (es) 2013-03-15 2016-01-13 Plexxikon Inc Compuestos heterociclicos y usos de los mismos
BR112015028845A2 (pt) 2013-05-30 2017-07-25 Plexxikon Inc compostos para a modulação da quinase e indicações da mesma

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040077595A1 (en) * 2002-09-06 2004-04-22 Insert Therapeutics, Inc. Cyclodextrin-based polymers for therapeutics delivery
US7863288B2 (en) * 2005-06-22 2011-01-04 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
US7863289B2 (en) * 2006-12-21 2011-01-04 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
US20100310659A1 (en) * 2009-04-03 2010-12-09 Plexxikon, Inc. Compositions and Uses Thereof
US20100286142A1 (en) * 2009-05-06 2010-11-11 Ibrahim Prabha N Compounds and Methods for Kinase Modulation, and Indications Therefor
US8198273B2 (en) * 2009-05-06 2012-06-12 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Pratilas et. al., Hcaplus 2008:670875 "Marker genes showing changes in levels of expression in response to antineoplastic drug therapy and their use in selection of chemotherapy", 2008 *

Cited By (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9169250B2 (en) 2006-11-22 2015-10-27 Plexxikon Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
US9487515B2 (en) 2006-11-22 2016-11-08 Plexxikon Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
US9469640B2 (en) 2007-07-17 2016-10-18 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9844539B2 (en) 2007-07-17 2017-12-19 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10426760B2 (en) 2007-07-17 2019-10-01 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9260437B2 (en) 2008-05-19 2016-02-16 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9663517B2 (en) 2009-04-03 2017-05-30 Plexxikon Inc. Compositions and uses thereof
US9447089B2 (en) 2009-04-03 2016-09-20 Plexxikon Inc. Compositions and uses thereof
US9096593B2 (en) 2009-11-06 2015-08-04 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9617267B2 (en) 2009-11-18 2017-04-11 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9440969B2 (en) 2009-12-23 2016-09-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9682981B2 (en) 2010-04-21 2017-06-20 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US11337976B2 (en) 2011-02-07 2022-05-24 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9624213B2 (en) 2011-02-07 2017-04-18 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
WO2012138809A1 (fr) * 2011-04-05 2012-10-11 Dawei Zhang Utilisation de composés hétérocycliques en tant qu'inhibiteurs des kinases
US9550768B2 (en) 2011-05-17 2017-01-24 Plexxikon Inc. Kinase modulation and indications therefor
US10123998B2 (en) 2012-03-19 2018-11-13 Plexxikon Inc. Kinase modulation, and indications therefor
US9358235B2 (en) 2012-03-19 2016-06-07 Plexxikon Inc. Kinase modulation, and indications therefor
US9730918B2 (en) 2012-03-19 2017-08-15 Plexxikon Inc. Kinase modulation, and indications therefor
US9150570B2 (en) * 2012-05-31 2015-10-06 Plexxikon Inc. Synthesis of heterocyclic compounds
US9695169B2 (en) 2012-05-31 2017-07-04 Plexxikon Inc. Synthesis of heterocyclic compounds
US20140094611A1 (en) * 2012-05-31 2014-04-03 Plexxikon Inc. Synthesis of heterocyclic compounds
US9399639B2 (en) 2012-08-27 2016-07-26 Calitor Sciences, Llc Substituted azaindole compounds, salts, pharmaceutical compositions thereof and methods of use
US10227357B2 (en) 2012-09-06 2019-03-12 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10301280B2 (en) 2012-12-21 2019-05-28 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9676748B2 (en) 2012-12-21 2017-06-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US20160002228A1 (en) * 2013-01-18 2016-01-07 Genentech, Inc. 3-substituted pyrazoles and use as dlk inhibitors
US9550777B2 (en) * 2013-01-18 2017-01-24 Genentech, Inc. 3-substituted pyrazoles and use as DLK inhibitors
US10028942B2 (en) 2013-01-18 2018-07-24 Genentech, Inc. 3-substituted pyrazoles and uses thereof
US9822109B2 (en) 2013-03-15 2017-11-21 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9718847B2 (en) 2013-03-15 2017-08-01 Plexxikon Inc. Heterocyclic compounds and uses thereof
WO2014176268A1 (fr) * 2013-04-22 2014-10-30 Abbvie Inc. Thiazoles et utilisations de ceux-ci
US10233179B2 (en) 2013-04-22 2019-03-19 Abbvie Inc. Thiazoles and uses thereof
US9873700B2 (en) 2013-05-30 2018-01-23 Plexxikon Inc. Compounds for kinase modulation, and indications therefor
US10421761B2 (en) 2013-05-30 2019-09-24 Plexxikon Inc. Compounds for kinase modulation, and indications therefor
US9771369B2 (en) 2014-03-04 2017-09-26 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US9856259B2 (en) 2014-09-15 2018-01-02 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10160755B2 (en) 2015-04-08 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10829484B2 (en) 2015-07-28 2020-11-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10370374B2 (en) 2015-09-21 2019-08-06 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10899761B2 (en) 2015-09-21 2021-01-26 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9771363B2 (en) 2015-09-21 2017-09-26 Plexxikon Inc. Heterocyclic compounds and uses thereof
US10647716B2 (en) 2015-09-21 2020-05-12 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9975894B2 (en) 2015-09-21 2018-05-22 Plexxikon Inc. Heterocyclic compounds and uses thereof
US9938273B2 (en) 2015-12-07 2018-04-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10160747B2 (en) 2016-03-16 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10508085B2 (en) 2016-09-22 2019-12-17 Plexxikon Inc. Compounds and methods for IDO and TDO modulation, and indications therefor
US10703757B2 (en) 2016-12-23 2020-07-07 Plexxikon Inc. Compounds and methods for CDK8 modulation and indications therefor
US10577366B2 (en) 2017-03-20 2020-03-03 Plexxikon Inc. Crystalline forms of a compound that inhibits bromodomain
US10428067B2 (en) 2017-06-07 2019-10-01 Plexxikon Inc. Compounds and methods for kinase modulation
US10961240B2 (en) 2017-07-25 2021-03-30 Plexxikon Inc. Formulations of a compound modulating kinases
US10941142B2 (en) 2017-07-25 2021-03-09 Plexxikon Inc. Formulations of a compound modulating kinases
US10435404B2 (en) 2017-07-25 2019-10-08 Plexxikon Inc. Formulations of a compound modulating kinases
US10717735B2 (en) 2017-10-13 2020-07-21 Plexxikon Inc. Solid forms of a compound for modulating kinases
US11103505B2 (en) 2017-10-27 2021-08-31 Plexxikon Inc. Formulations of a compound modulating kinases
US11149011B2 (en) 2018-03-20 2021-10-19 Plexxikon Inc. Compounds and methods for IDO and TDO modulation, and indications therefor

Also Published As

Publication number Publication date
EP2516438A4 (fr) 2013-06-12
AU2010336524A1 (en) 2012-07-05
RU2012131373A (ru) 2014-01-27
AR079690A1 (es) 2012-02-15
JP2013515736A (ja) 2013-05-09
KR20120112623A (ko) 2012-10-11
WO2011079133A2 (fr) 2011-06-30
AU2010336524B2 (en) 2015-10-08
JP5815558B2 (ja) 2015-11-17
MX2012007429A (es) 2012-07-23
CA2784393A1 (fr) 2011-06-30
UY33159A (es) 2011-07-29
WO2011079133A3 (fr) 2011-11-17
BR112012015745A2 (pt) 2016-05-17
CN102753549A (zh) 2012-10-24
EP2516438A2 (fr) 2012-10-31
TW201132639A (en) 2011-10-01
US20150080372A1 (en) 2015-03-19
US9440969B2 (en) 2016-09-13

Similar Documents

Publication Publication Date Title
US9440969B2 (en) Compounds and methods for kinase modulation, and indications therefor
US9617267B2 (en) Compounds and methods for kinase modulation, and indications therefor
US8198273B2 (en) Compounds and methods for kinase modulation, and indications therefor
US9469640B2 (en) Compounds and methods for kinase modulation, and indications therefor
US8901301B2 (en) Pyrrolo[2,3-]pyridine kinase inhibitors
AU2010315126B2 (en) Compounds and methods for kinase modulation, and indications therefor
EP3004060B1 (fr) Composés pour modulation de kinases, et indications correspondantes

Legal Events

Date Code Title Description
AS Assignment

Owner name: PLEXXIKON INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:IBRAHIM, PRABHA N.;WU, GUOXIAN;LIN, JACK;AND OTHERS;SIGNING DATES FROM 20120521 TO 20120523;REEL/FRAME:028287/0132

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION