US20150166591A1 - Methods and compositions for raf kinase mediated diseases - Google Patents

Methods and compositions for raf kinase mediated diseases Download PDF

Info

Publication number
US20150166591A1
US20150166591A1 US14/398,893 US201314398893A US2015166591A1 US 20150166591 A1 US20150166591 A1 US 20150166591A1 US 201314398893 A US201314398893 A US 201314398893A US 2015166591 A1 US2015166591 A1 US 2015166591A1
Authority
US
United States
Prior art keywords
amino
alkyl
compound
cycloalkyl
phenyl
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
US14/398,893
Other languages
English (en)
Inventor
Xiaotian Zhu
Yihan Wang
William C. Shakespeare
Wei-Sheng Huang
David C. Dalgarno
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.)
Ariad Pharmaceuticals Inc
Original Assignee
Ariad Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ariad Pharmaceuticals Inc filed Critical Ariad Pharmaceuticals Inc
Priority to US14/398,893 priority Critical patent/US20150166591A1/en
Publication of US20150166591A1 publication Critical patent/US20150166591A1/en
Assigned to ARIAD PHARMACEUTICALS, INC. reassignment ARIAD PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DALGARNO, DAVID C., HUANG, WEI-SHENG, SHAKESPEARE, WILLIAM C., ZHU, XIAOTIAN, WANG, YIHAN
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/645Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having two nitrogen atoms as the only ring hetero atoms
    • C07F9/6509Six-membered rings
    • C07F9/6512Six-membered rings having the nitrogen atoms in positions 1 and 3
    • 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
    • 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
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6515Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having three nitrogen atoms as the only ring hetero atoms
    • C07F9/6521Six-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65583Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system each of the hetero rings containing nitrogen as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65586Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system at least one of the hetero rings does not contain nitrogen as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6581Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms
    • C07F9/6584Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms having one phosphorus atom as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6564Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms
    • C07F9/6581Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms
    • C07F9/6584Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having phosphorus atoms, with or without nitrogen, oxygen, sulfur, selenium or tellurium atoms, as ring hetero atoms having phosphorus and nitrogen atoms with or without oxygen or sulfur atoms, as ring hetero atoms having one phosphorus atom as ring hetero atom
    • C07F9/65842Cyclic amide derivatives of acids of phosphorus, in which one nitrogen atom belongs to the ring
    • C07F9/65846Cyclic amide derivatives of acids of phosphorus, in which one nitrogen atom belongs to the ring the phosphorus atom being part of a six-membered ring which may be condensed with another ring system

Definitions

  • This invention relates to pharmaceutical compositions and methods for inhibiting the proliferation of cells.
  • kinase inhibitors In human clinical studies with non-small cell lung cancer (NSCLC) patients, the kinase inhibitors, erlotinib and gefitinib have been found to be effective, but in only a subset of patients. It was later determined that the responsive patients had certain mutations in the gene for epidermal growth factor receptor (EGFR).
  • EGFR epidermal growth factor receptor
  • the mutant forms of EGFR are enzymatically active without the need for ligand stimulation. They are also particularly sensitive to kinase inhibitors like erlotinib and gefitinib, which competitively bind to the ATP binding site of the EGFR kinase domain. Those mutations have been cataloged and described at length in the scientific literature.
  • T790M mutation render drugs like erlotinib and gefitinib less effective. Those mutations are associated with resistance to the drugs and to relapse in patients with cancer cells having the T790M mutation.
  • New therapies are needed for the treatment of EGFR-driven cancers in which mutations confer resistance to front line tyrosine kinase inhibitor (“TKI”) therapies.
  • TKI front line tyrosine kinase inhibitor
  • new therapies for inhibiting cells expressing such gefitinib-resistant or erlotinib-resistant EGFR genes could be of profound benefit.
  • This invention relates to the discovery of a class of compounds that inhibit EGFR and medically significant mutant forms thereof.
  • mutants include mutant EGFR proteins that are enzymatically active in the absence of protein ligand and mutants such as the T790M EGFR mutant that are resistant to the EGFR inhibitors erlotinib and gefitinib.
  • Compounds of this invention possessing desirable selectivity for the intended biological targets and advantageous pharmaceutical properties make them of interest for treating cancers characterized by the expression of EGFR or an EGFR mutant, especially in cases that are resistant or refractory to erlotinib or gefitinib.
  • This invention provides a compound of Formula (I):
  • U 1 and U 2 are both N and U 3 is C—R e ; or U 3 is N, one of U 1 and U 2 is N, and the other is C—R d ; or U 3 is C—R e , one of U 1 and U 2 is N, and the other is C—R d ;
  • V 1 is O, S, NR V , CO, CH 2 , or CF 2 ;
  • R V is H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, or aryl;
  • R d is H, halo, CN, alkyl, cycloalkyl, alkoxy, haloalkyl, alkenyl, haloalkenyl or halocycloalkyl;
  • R e is H or NH 2 ; or, R d and R e , together with the ring atom to which each is attached, form a 5- or 6-membered ring containing one, two or three heteroatoms, independently selected from N, S and O, wherein the 5- or 6-membered ring so formed is substituted with R h which is C 1-4 alkyl or halo;
  • R g is H, F, —P(O)(R 3A )(R 3B ), —S(O)N(R 3C )(R 3D ), —S(O) 2 R 3E , —C(O)N(R 3F )(R 3G ), —OC(O)N(R 3F )(R 3G ), —NR 3H C(O)OR 3I or a 5- or 6-membered heterocyclic ring comprising 1, 2, 3 or 4 N atoms;
  • R g2 is H, F, W 1 , —P(O)(R 3A )(R 3B ), —S(O)N(R 3C )(R 3D ), —S(O) 2 R 3E , —C(O)N(R 3F )(R 3G ), —OC(O)N(R 3F )(R 3G ), —NR 3H C(O)OR 3I , C 1-6 alkoxy or C 1-4 alkyl;
  • R g1 is H, F, —OR 2 , —P(O)(R 3A )(R 3B ), —S(O)N(R 3C )(R 3D ), —S(O) 2 R 3E , —C(O)N(R 3F )(R 3G ), —OC(O)N(R 3F )(R 3G ), —NR 3 HC(O)OR 3I , or substituted or unsubstituted 5- or 6-membered heterocyclic ring;
  • Ring A is:
  • R b2 is H, F, or an optionally substituted 5- or 6-membered heterocyclic ring containing 1, 2 or 3 N or O atoms;
  • R b4 is H, F, W 1 , C 1-6 alkoxy, C 3-6 alkenyloxy, C 3-6 cycloalkoxy, —OC(O)N(R 5A )(R 5B ), —NR 5C C(O)OR 5D , or substituted or unsubstituted 5- or 6-membered heterocyclic ring comprising 1, 2 or 3 N or O atoms;
  • each R 5A , R 5B , R 5C and R 5D is independently H, alkyl, alkenyl, alkynyl or heteroalkyl, or R 5A and R 5B , together with the atom to which each is attached, form a substituted or unsubstituted 5- or 6-membered heterocyclic ring;
  • R a1 is H, halo, W 1 , —CN, —NO 2 , —R 1 , —OR 2 , —O—NR 1 R 2 , —NR 1 R 2 , —NR 1 —NR 1 R 2 , —NR 1 —OR 2 , —C(O)YR 2 , —OC(O)YR 2 , —NR 1 C(O)YR 2 , —SC(O)YR 2 , —NR 1 C( ⁇ S)YR 2 , —OC( ⁇ S)YR 2 , —C( ⁇ S)YR 2 , —YC( ⁇ NR 1 )YR 2 , —YC( ⁇ N—OR 1 )YR 2 , —YC( ⁇ N—NR 1 R 2 )YR 2 , —YP( ⁇ O)(YR 1 )(YR 2 ), —S(O) r R 2 , —SO 2 NR 1 R 2
  • X 1 and X 2 are each independently CH or N;
  • R a1 and R b4 together with the atom to which each is attached, form an optionally substituted 5- or 6-membered heterocyclic ring comprising 1, 2 or 3 heteroatoms independently selected from N and O;
  • R a2 is H, halo, C 1-6 alkyl, C 2-6 alkenyl, C 3-6 cycloalkyl, C 1-6 alkoxy, C 2-6 alkenyloxy, C 3-6 cycloalkyloxy or 4- to 7-membered heterocyclyl, wherein the alkyl, alkenyl, cycloalkyl, alkoxy, C 2-6 alkenyloxy, cycloalkyloxy and heterocyclyl are unsubstituted or substituted with one or more halo, amino, C 1-6 alkylamino, or di-Cm alkylamino groups;
  • Y is independently a bond, —O—, —S— or —NR 1 —;
  • R 1 and R 2 are independently H or R 15 ;
  • R 4 is alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, heterocyclic and heteroaryl;
  • R X is halo
  • R 7 is H, alkyl or heteroalkyl, wherein the alkyl and heteroalkyl groups are independently optionally substituted with an amino, alkylamino or dialkylamino group;
  • R 8 is C 1-6 alkyl
  • R 9 and R 10 are independently H, halo, —C(O)R 16 , alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, heterocyclyl or heteroaryl, wherein R 9 and R 10 , if not H, are optionally substituted with one or more halo, amino, alkylamino, dialkylamino, alkoxy, cycloalkyl, heterocyclyl or heteroaryl groups, wherein said group, if not halo, is optionally substituted with one or more halo, C 1-4 alkyl, alkoxyl, halo(C 1-4 )alkyl or C 3-7 cycloalkyl groups;
  • R 11 is H, halo, —C(O)—OR 12 , alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, heterocyclic, or heteroaryl, wherein R 11 , if not H, is optionally substituted with one or more halo, amino, alkoxyl, cycloalkyl, heterocyclic or heteroaryl groups, wherein said group, if not halo, is optionally substituted with one or more halo or alkyl, alkoxyl, cycloalkyl or heterocyclyl groups, wherein the alkyl, alkoxyl, cycloalkyl and heterocyclyl group is optionally substituted with one or more alkyl, halo or hydroxyl substituents;
  • R 12 is alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, heterocyclic, or heteroaryl;
  • R 13 is H or C 1-4 alkyl
  • R 14 is R T or R W ;
  • R 15 is alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, 4- to 7-membered heterocyclyl, or heteroaryl, wherein each R 15 is optionally substituted with one or more halo, cycloalkyl, heterocyclic or heteroaryl groups, wherein said cycloalkyl, heterocyclic or heteroaryl groups(s) are independently optionally substituted with one or more halo, alkyl, haloalkyl, hydroxyalkyl, amino, dialkylamino or cycloalkyl groups;
  • R 16 is OH, —O-alkyl, cycloalkyl, heterocyclyl, —NH 2 , —NH-alkyl, or —N-dialkyl wherein the alkyl, cycloalkyl or heterocyclyl moiety is optionally substituted with halo, amino, alkylamino, dialkylamino, alkyl or hydroxyl;
  • R T is H or —CH 3 ;
  • R W is halo; substituted methyl; or an optionally substituted group selected from (C 2-6 )alkyl, (C 1-6 )heteroalkyl, heterocyclyl, aryl and heteroaryl; wherein the substituents on the optionally substituted (C 2-6 )alkyl, (C 1-6 )heteroalkyl, heterocyclyl, aryl and heteroaryl groups are selected from halo, haloalkyl, alkoxy, heterocyclyl, substituted heterocyclyl, amino, alkylamino, and dialkylamino, and in the case of an optionally substituted heterocyclyl, the optional substituents may further be selected from hydroxyl, alkyl, haloalkyl, hyroxyalkyl, alkoxyalkyl, amino, alkylamino and dialkylamino;
  • the compound comprises at least one —P(O)(R 3A )(R 3B );
  • R d is H, halo, CN, alkyl, alkoxy, haloalkyl, alkenyl, haloalkenyl or halocycloalkyl;
  • W 1 is —NR 7 C(O)C(R 11 ) ⁇ CR 9 R 10 , —C(O)C(R 11 ) ⁇ CR 9 R 10 , —CH 2 P(O)C(R 11 ) ⁇ CR 9 R 10 , —OP(O)C(R 8 ) ⁇ CR 9 R 10 , —NR 7 S(O) 2 C(R 11 ) ⁇ CR 9 R 10 , —NR 7 C(O)C C ⁇ C—R 14 ,
  • R g is H, —P(O)(R 3A )(R 3B ), —S(O)N(R 3D )(R 3D ), —S(O) 2 R 3E , —C(O)N(R 3F )(R 3G ), —OC(O)N(R 3F )(R 3G ), —NR 3 HC(O)OR 3I , a 5- or 6-membered heterocyclic ring comprising 1, 2, 3 or 4 N atoms, wherein each of R 3A , R 3B , R 3D , R 3D , R 3E , R 3F , R 3G , R 3H , and R 3I is, independently, selected from H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, and heteroalkyl, or R 3A and R 3B , or R 3D and R 3D , or
  • R g1 is H, F, —P(O)(R 3A )(R 3B ), —S(O)N(R 3D )(R 3D ), —S(O) 2 R 3E , —C(O)N(R 3F )(R 3G ), —OC(O)N(R 3F )(R 3G ), —NR 3 ′′C(O)OR 3I , or an optionally substituted 5- or 6-membered heterocyclic ring; wherein the variable terms are as defined for Formula (I).
  • U 1 is N
  • U 2 is C—R d
  • U 3 is C—Re.
  • U 1 is C—R d
  • U 2 is N
  • U 3 is C—R e .
  • U 1 is N
  • U 2 is N
  • U 3 is C—R e .
  • V 1 is NH. In another V 1 is O. In certain embodiments of both classes R d is Cl. In certain embodiments of both classes R g or R g1 is —P(O)(CH 3 ) 2 or —P(O)(CH 2 CH 3 ) 2 .
  • U 3 is N, one of U 1 and U 2 is N, and the other is C—R d .
  • R a1 ; R a2 ; R b2 ; R b4 ; R g ; R g1 ; R g2 ; R d ; and R h are as defined in Formula (I).
  • R a2 ; R b2 ; R b4 ; R g ; R g1 ; R g2 ; R d ; and R h are as defined in Formula (I).
  • R a1 ; R a2 ; R b2 ; R b4 ; R g ; R g1 ; R g2 ; R d ; and R h are as defined in Formula (I).
  • R a2 ; R b2 ; R b4 ; R g ; R g1 ; R g2 ; R d ; and R h are as defined in Formula (I), and R a1 is selected from W 1 , —R 1 , —C(O)YR 2 , —C( ⁇ S)YR 2 , —C( ⁇ NR 1 )YR 2 , —C( ⁇ N—OR 1 )YR 2 , —C( ⁇ N—NR 1 R 2 )YR 2 , —S(O) r R 2 , and
  • each Y is, independently, a bond, —O—, —S— or —NR 1 —;
  • each occurrence of R 1 and R 2 is, independently, selected from H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, heterocyclic and heteroaryl; each of X 1 and X 2 is, independently, selected from CH and N;
  • W 1 is as defined in Formula (I);
  • R 4 is selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, heterocyclic and heteroaryl.
  • Ring A is cyclopentyl
  • Ring A is cyclopentyl
  • R a1 ; R a2 ; R b4 ; R g ; R g1 ; R g2 ; R d ; and R h are as defined in Formula (I).
  • R a1 is selected from W 1 , —R 1 , —C(O)YR 2 , —C( ⁇ S)YR 2 , —C( ⁇ NR 1 )YR 2 , —C( ⁇ N ⁇ OR 1 )YR 2 , —C( ⁇ N—NR 1 R 2 )YR 2 , —S(O) r R 2 , and
  • each Y is, independently, a bond, —O—, —S— or —NR 1 —;
  • W 1 is as defined in Formula (I);
  • each occurrence of R 1 and R 2 is, independently, selected from H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, heterocyclic and heteroaryl;
  • R a1 is W 1 , —R 1 , —C(O)YR 2 , —C( ⁇ S)YR 2 , —C( ⁇ NR 1 )YR 2 , —C( ⁇ N—OR 1 )YR 2 , —C( ⁇ N—NR 1 R 2 )YR 2 , —S(O) r R 2 , or
  • each Y is, independently, a bond, —O—, —S— or —NR 1 —;
  • R b2 ; R b4 ; R g ; R g1 ; R g2 ; R d ; R h and W 1 are as defined in Formula (I);
  • each occurrence of R 1 and R 2 is, independently, selected from H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, heterocyclic and heteroaryl;
  • each of X 1 and X 2 is, independently, CH or N;
  • R 4 is alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, heterocyclic or heteroaryl.
  • R a1 is selected from W 1 , —R 1 , —C(O)YR 2 , —C( ⁇ S)YR 2 , —C( ⁇ NR 1 )YR 2 , —C( ⁇ N—OR 1 )YR 2 , —C( ⁇ N—NR 1 R 2 )R 2 , —S(O) r R 2 , and
  • each Y is, independently, a bond, —O—, —S— or —NR 1 —;
  • W 1 is as defined in Formula (I);
  • each occurrence of R 1 and R 2 is, independently, selected from H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, heterocyclic and heteroaryl;
  • each of X 1 and X 2 is, independently, selected from CH and N; and R 4 is selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, heterocyclic and heteroaryl.
  • R a2 is H, F, Cl, —CH 3 , —CF 3 , —CH 2 CH 3 , —OCH 3 , —OCF 3 , —OCH 2 CH 3 or -heterocyclyl-O—, e.g., a 4-membered heterocyclyl.
  • R d is H, Cl, F, Br, I, CN, CH 3 , CF 3 , cyclopropyl or —CH 2 CH 2 ⁇ CH 2 .
  • R b2 is H.
  • R g1 is H and R g2 is H, F, C 1-6 alkyl, or C 1-6 alkoxy.
  • R g is —P(O)(R 3A )(R 3B ), wherein R 3A and R 3B , are as defined in Formula (I). In some cases R g is —P(O)(CH 3 ) 2 or —P(O)(CH 2 CH 3 ) 2 .
  • R g is —S(O) 2 R 3E ), wherein R 3E is as defined in Formula (I).
  • R g is —S(O) 2 CH(CH 3 ) 2 .
  • R a1 is a 5- or 6-membered heteroaryl ring optionally substituted with an —OH, halo, alkyl, substituted alkyl substituent.
  • R a1 is a 4-, 5-, 6- or 7-membered heterocycle which is optionally substituted with one or more groups selected from halo and R 17 .
  • R 17 is an alkyl, cycloalkyl, heteroalkyl, 4- to 7-membered heterocyclyl or heteroaryl group, and R 17 is optionally substituted with one or more halo, alkyl, cycloalkyl, heterocyclic or heteroaryl groups, of which, the cycloalkyl, heterocyclic or heteroaryl substituent is optionally substituted with one or more halo, alkyl, haloalkyl, hydroxyalkyl, amino, dialkylamino or cycloalkyl groups.
  • R a1 is selected from the following:
  • R a1 is —OR 2 , as defined in Formula (I).
  • R a1 is selected from the following:
  • R a1 is an optionally substituted alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, or heteroalkyl group, wherein optional substituents are selected from halo, cycloalkyl, heterocyclic or heteroaryl groups, wherein said cycloalkyl, heterocyclic and heteroaryl substituent(s) are independently optionally substituted with one or more halo, alkyl, haloalkyl, hydroxyalkyl, amino, dialkylamino or cycloalkyl groups.
  • R a1 is an optionally substituted alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, or heteroalkyl group, wherein optional substituents are selected from halo, cycloalkyl, heterocyclic or heteroaryl groups, wherein said cycloalkyl, heterocyclic
  • R a1 is of the Formula:
  • J 1 and J 2 are independently H, halo or R J ; or J 1 and J 2 together with the atom to which each is attached form an optionally substituted ring which is C 3-8 cycloalkyl, 3- to 7-membered heterocyclic, or heteroaryl;
  • J 3 and J 4 are independently H or R J ; or J 3 and J 4 together with the atom to which each is attached form an optionally substituted ring which is 3- to 7-membered heterocyclic or heteroaryl ring;
  • R J is C 1-6 alkyl, C 3-8 cycloalkyl, C 1-8 heteroalkyl, or 3- to 7-membered heterocyclyl, wherein each R J is independently selected from halo, haloalkyl, hydroxyl, hydroxyalkyl, amino, alkylamino, dialkylamino, cycloalkyl, alkoxy, cycloalkoxy and heterocyclic groups, wherein the alkyl, cycloalkyl, and heterocyclic groups on R J are optionally substituted with one or more halo, alkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl, amino, alkylamino, dialkylamino or cycloalkyl groups; and, z is 1-3.
  • R a1 may be selected from the following:
  • R b4 is —NR 7 C(O)C(R 11 ) ⁇ CR 9 R 10 , —NR 7 S(O) 2 C(R 9 )(R 10 )(R X ), —NR 7 C(O)C ⁇ C—R 14 , Or —NR 7 C(O)C(R 9 )(R 10 )(R X ), wherein R 7 , R 9 , R 10 , R 11 , R 14 , and R X are as defined in Formula (I).
  • R b4 may be —NHC(O)CH ⁇ CH 2 or may be selected from the following:
  • R b4 is —NHC(O)C ⁇ CH or may be selected from the following:
  • R b4 is W 1 as defined in Formula (I).
  • R a2 is H, halo, —CH 3 , —CF 3 , —CH 2 CH 3 , —OCH 3 , —OCF 3 , —OCH 2 CH 3 , —OCH 2 CH 2 N(CH 3 ) 2 or —O-heterocyclyl.
  • R g is —P(O)(R 3A )(R 3B ), wherein R 3A and R 3B , are as defined in Formula (I), e.g., R g is —P(O)(CH 3 ) 2 or —P(O)(CH 2 CH 3 ) 2 .
  • R g is —S(O) 2 R 3E ), wherein R 3E is as defined in Formula (I), e.g., R g is —S(O) 2 CH(CH 3 ) 2 .
  • R d may be CI, F, Br, I, or CH 3 .
  • R g1 is H and R g2 is H, F, C 1-6 alkyl, or C 1-6 alkoxy.
  • R g is —P(O)(R 3A )(R 3B ) or —S(O) 2 R 3E , wherein R 3A ; R 3B ; and R 3E are as defined in formula (I).
  • R g can be selected from —P(O)(CH 3 ) 2 and —S(O) 2 (CH(CH 3 ) 2 ).
  • R a1 is a 5 or 6 member heterocyclic ring including 1 or 2 N or O atoms which is unsubstituted or substituted with an alkyl group.
  • R a1 can be selected from any of the following groups:
  • R a2 is methoxy;
  • R d is CI, F, Br, I, or CH 3 ;
  • R g is —P(O)(CH 3 ) 2 or —S(O) 2 (CH(CH 3 ) 2 ).
  • U 1 is N, U 2 is C—R d , and U 3 is C—R e ; U 1 is C—R d , U 2 is N, and U 3 is C—R e ; U 1 is N, U 2 is N, and U 3 is C—R e ; or U 3 is N, one of U 1 and U 2 is N, and the other is C—R d .
  • Exemplary compounds include those in which V 1 is NH, V 1 is O, R d is Cl, R g or R g1 is —P(O)(CH 3 ) 2 or —P(O)(CH 2 CH 3 ) 2 , and/or R b4 is —NHC(O)C(R 11 ) ⁇ CH 2 .
  • R b4 is —NHC(O)C(R 11 ) ⁇ CH 2 ;
  • R 11 is —C(O)—OR 12 , —CH 2 N(CH 3 ) 2 , H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, heterocyclic, or heteroaryl;
  • R 12 is alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroalkyl, heterocyclic, or heteroaryl;
  • R d is Cl; and
  • R g or R g1 is selected from —P(O)(CH 3 ) 2 and —P(O)(CH 2 CH 3 ) 2 .
  • R b4 is —NHC(O)CH ⁇ CH 2 .
  • the invention features a method for treating an EGFR-driven cancer in a subject by administering to the subject a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof.
  • the invention features a method for treating an EGFR-driven cancer in a subject, the method including (a) providing a subject having an EGFR-driven cancer characterized by the presence of a mutation in epidermal growth factor receptor kinase (EGFR), and (b) administering to the subject a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof.
  • EGFR-driven cancer is characterized by the presence of one or more mutations selected from: (i) L858R, (ii) T790M, (iii) both L858R and T790M, (iv) delE746_A750, and (v) both delE746_A750 and T790M.
  • the EGFR-driven cancer can be a non-small cell lung cancer (NSCLS); glioblastoma; pancreatic cancer; head and neck cancer (e.g., squamous cell carcinoma); breast cancer; colorectal cancer; epithelial cancer; ovarian cancer; prostate cancer; an adenocarcinoma, or any EGFR-driven cancer described herein.
  • NSCMS non-small cell lung cancer
  • glioblastoma pancreatic cancer
  • head and neck cancer e.g., squamous cell carcinoma
  • breast cancer colorectal cancer
  • epithelial cancer ovarian cancer
  • prostate cancer an adenocarcinoma, or any EGFR-driven cancer described herein.
  • the method further includes administering to the subject a first kinase inhibitor selected from erlotinib, gefitinib, and pharmaceutically acceptable salts thereof, within 6 days (e.g., within 2 weeks, 1 week, 6 days, 5 days, 4 days, 3 days, 2 days, 1 day, or simultaneously) of administering the compound of the invention (e.g., a compound of any of Formulas (I), (Ia)-(Ic), (IIa)-(IIc), (IIIa)-(IIIe), (IVa)-(IVe), (Va)-(Ve), (VIa)-(VIe), (VIIIa)-(VIIIe), (IXa)-(IXe), and (Xa)-(Xe)), wherein each of the compound of the invention and the first kinase inhibitor are administered in an amount that together is sufficient to treat the EGFR-driven cancer.
  • a first kinase inhibitor selected from erlotinib, gefitinib
  • the invention features a method of inhibiting the proliferation of a cell expressing an EGFR mutant, the method including contacting the cell with a compound of the invention, or a pharmaceutically acceptable salt thereof, in an amount sufficient to inhibit the proliferation.
  • the EGFR mutant can be characterized by the presence of one or more mutations in epidermal growth factor receptor kinase (EGFR) selected from: (i) L858R, (ii) T790M, (iii) both L858R and T790M, (iv) delE746_A750, (v) both delE746_A750 and T790M, and any other EGFR mutations described herein.
  • EGFR epidermal growth factor receptor kinase
  • the cell is a cancer cell (e.g., a cell from a non-small cell lung cancer (NSCLS); glioblastoma; pancreatic cancer; head and neck cancer (e.g., squamous cell carcinoma); breast cancer; colorectal cancer; epithelial cancer; ovarian cancer; prostate cancer; an adenocarcinoma, or any other EGFR expressing cancer described herein).
  • NSC non-small cell lung cancer
  • glioblastoma pancreatic cancer
  • head and neck cancer e.g., squamous cell carcinoma
  • breast cancer colorectal cancer
  • epithelial cancer ovarian cancer
  • prostate cancer an adenocarcinoma, or any other EGFR expressing cancer described herein.
  • the invention further features a method of treating an EGFR-driven cancer refractory to a first kinase inhibitor selected from erlotinib, gefitinib, and pharmaceutically acceptable salts thereof, in a subject by administering to the subject a compound of the invention, or a pharmaceutically acceptable salt thereof, in an amount sufficient to treat the cancer.
  • a first kinase inhibitor selected from erlotinib, gefitinib, and pharmaceutically acceptable salts thereof
  • the compound in any of Formulas I, III(a)-(e), IV(a)-(e), V(a)-(e), VI(a)-(e), VII(a)-(e), VIII(a)-(e), IX(a)-(e), and X(a)-(e), the compound can be either in its free base form, or a pharmaceutically acceptable salt.
  • the response criteria for the methods of the invention can be graded according to the response evaluation criteria in solid tumors (RECIST) guidelines (see Eur. J. Cancer 45:228 (2009)) that define when cancer patients improve (“respond”), stay the same (“stabilize”), or worsen (“progression”) during treatments.
  • a complete response is characterized by: (i) disappearance of all target lesions; and (ii) any pathological lymph nodes (whether target or non-target) must have reduction in short axis to ⁇ 10 mm.
  • a partial response is characterized by: (i) at least a 30% decrease in the sum of diameters of target lesions, taking as reference the baseline sum diameters.
  • a progressive disease is characterized by (i) at least a 5%, 10%, or 20% increase in the sum of diameters of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study); or (ii) the appearance of one or more new lesions.
  • administering refers to a method of giving a dosage of a pharmaceutical composition to a mammal, where the method is, e.g., oral, intravenous, intraperitoneal, intraarterial, or intramuscular.
  • the preferred method of administration can vary depending on various factors, e.g., the components of the pharmaceutical composition, site of the potential or actual disease and severity of disease.
  • EGFR-driven cancer is meant a cancer characterized by inappropriately high expression of an EGFR gene or by a mutation in an EGFR gene that alters the biological activity of an EGFR nucleic acid molecule or polypeptide.
  • EGFR-driven cancers can arise in any tissue, including brain, blood, connective tissue, liver, mouth, muscle, spleen, stomach, testis, and trachea.
  • EGFR-driven cancers can include non-small cell lung cancer (NSCLS), including one or more of squamous cell carcinoma, adenocarcinoma, adenocarcinoma, bronchioloalveolar carcinoma (BAC), BAC with focal invasion, adenocarcinoma with BAC features, and large cell carcinoma; neural tumors, such as glioblastomas; pancreatic cancer; head and neck cancers (e.g., squamous cell carcinoma); breast cancer; colorectal cancer; epithelial cancer, including squamous cell carcinoma; ovarian cancer; prostate cancer; adenocarcinomas; and including cancers which are EGFR mediated.
  • NSCS non-small cell lung cancer
  • BAC bronchioloalveolar carcinoma
  • BAC bronchioloalveolar carcinoma
  • BAC BAC with focal invasion, adenocarcinoma with BAC features, and large cell carcinoma
  • neural tumors such as glioblastomas
  • pancreatic cancer head
  • an “EGFR mutant” or “mutant” includes one or more deletions, substitutions, or additions in the amino acid or nucleotide sequences of EGFR protein, or EGFR coding sequence.
  • the EGFR mutant can also include one or more deletions, substitutions, or additions, or a fragment thereof, as long as the mutant retains or increases tyrosine kinase activity, compared to wild type EGFR.
  • kinase or phosphorylation activity can be increased (e.g., by at least 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or even 100%), as compared to wild type EGFR.
  • Particular EGFR mutants are described herein, where mutations are provided relative to the position of an amino acid in human EGFR, as described in the sequence provided in NCBI GenBank Reference Sequence: NP — 005219.2.
  • the term “inhibiting the proliferation of a cell expressing an EGFR mutant” refers to measurably slowing, stopping, or reversing the growth rate of the EGFR-expressing cells in vitro or in vivo. Desirably, a slowing of the growth rate is by at least 10%, 20%, 30%, 50%, or even 70%, as determined using a suitable assay for determination of cell growth rates (e.g., a cell growth assay described herein).
  • the EGFR mutant can be any EGFR mutant described herein.
  • refractory refers to a cancer which is progressive in response to a given particular therapy.
  • the cancer can be refractory either from the initial administration of the therapy; or become refractory over time in response to the therapy.
  • sequence identity is meant the shared identity between two or more nucleic acid sequence, or two or more amino acid sequences, expressed in the terms of the identity between the sequences. Sequence identity can be measured in terms of percentage identity; the higher the percentage, the more identical the sequences are. Homologs or orthologs of nucleic acid or amino acid sequences possess a relatively high degree of sequence identity when aligned using standard methods. Methods of alignment of sequences for comparison are well known in the art. Various programs and alignment algorithms are described in: Smith and Watermann, Adv. Appl. Math. 2:482 (1981); Needleman and Wunsch, J. Mol. Biol. 48:443 (1970); Pearson and Lipman, Proc. Natl. Acad. Sci.
  • NCBI National Center for Biological Information
  • the option can be set as follows: ⁇ i is set to a file containing the first nucleic acid sequence to be compared; ⁇ j is set to a file containing the second nucleic acid sequence to be compared; ⁇ p is set to blastn; ⁇ o is set to any desired file name; ⁇ q is set to ⁇ 1; ⁇ r is set to 2; and all other options are left at their default setting.
  • the number of matches is determined by counting the number of positions where an identical nucleotide or amino acid residue is present in both sequences.
  • the percent sequence identity is determined by dividing the number of matches either by the length of the sequence set forth in the identified sequence, or by an articulated length (such as 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, or 400 consecutive nucleotides or amino acid residues from a sequence set forth in an identified sequence), followed by multiplying the resulting value by 100.
  • an articulated length such as 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, or 400 consecutive nucleotides or amino acid residues from a sequence set forth in an identified sequence
  • Nucleic acid molecules that hybridize under stringent conditions to an EGFR gene sequence typically hybridize to a probe based on either an entire EGFR gene or selected portions of the gene (e.g., the kinase domain or a segment of the gene that contains the mutated codons described herein), under conditions described above.
  • treating refers to administering a pharmaceutical composition for prophylactic and/or therapeutic purposes.
  • To “prevent disease” refers to prophylactic treatment of a subject who is not yet ill, but who is susceptible to, or otherwise at risk of, a particular disease.
  • To “treat disease” or use for “therapeutic treatment” refers to administering treatment to a subject already suffering from a disease to improve or stabilize the subject's condition.
  • treating is the administration to a subject either for therapeutic or prophylactic purposes.
  • substituents may be selected from those disclosed herein as generally appropriate in the given context, e.g. on an alkyl carbon, an aryl carbon, etc., and specifically include substitution exemplified in the examples.
  • Other parameters of functional groups are also disclosed in detail below.
  • alkyl refers to linear, branched, cyclic, and polycyclic non aromatic hydrocarbon groups, which may be substituted or unsubstituted. Unless otherwise specified, “alkyl” groups contain one to eight, and typically one to six carbon atoms.
  • alkyl examples include, without limitation, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, butyl, isobutyl, sec-butyl, tert-butyl, cyclobutyl, pentyl, isopentyl tert-pentyl, cyclopentyl, hexyl, isohexyl, cyclohexyl, and n-heptyl, among others.
  • alkyl groups include, without limitation, haloalkyl groups (e.g., fluoromethyl, difluoromethyl, trifluoromethyl, 2-fluoroethyl, 3-fluoropropyl), hydroxymethyl, 2-hydroxyethyl, 3-hydroxypropyl, benzyl, substituted benzyl, and phenethyl, among others.
  • haloalkyl groups e.g., fluoromethyl, difluoromethyl, trifluoromethyl, 2-fluoroethyl, 3-fluoropropyl
  • hydroxymethyl 2-hydroxyethyl, 3-hydroxypropyl
  • benzyl substituted benzyl
  • phenethyl phenethyl
  • alkoxy refers to a subset of alkyl in which an alkyl group as defined above with the indicated number of carbons attached through an oxygen bridge, —O-alkyl, wherein the alkyl group contains 1 to 8 carbons atoms and is substituted or unsubstituted.
  • alkoxy groups include, without limitation, methoxy, ethoxy, n-propoxy, i-propoxy, t-butoxy, n-butoxy, s-pentoxy, —OCF 3 , and —O-cyclopropyl.
  • haloalkyl refers to a subset of alkyl in which an alkyl group as defined above having one or more hydrogen atoms of the alkyl substituted with a halogen atom.
  • exemplary haloalkyl groups include, without limitation, fluoromethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl and the like.
  • alkenyl refers to a branched or unbranched hydrocarbon group containing one or more double bonds and having from 2 to 8 carbon atoms.
  • An alkenyl may optionally include monocyclic or polycyclic rings, in which each ring desirably has from three to six members.
  • the alkenyl group may be substituted or unsubstituted.
  • Alkenyl groups include, without limitation, vinyl, allyl, 2-cyclopropyl-1-ethenyl, 1-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 2-methyl-1-propenyl, and 2-methyl-2-propenyl.
  • alkynyl refers to a branched or unbranched hydrocarbon group containing one or more triple bonds and having from 2 to 8 carbon atoms.
  • the alkynyl group may be substituted or unsubstituted.
  • Alkynyls include, without limitation, ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, and 3-butynyl.
  • cycloalkyl refers to cyclic or polycyclic hydrocarbon groups of from 3 to 13 carbon atoms, any of which is saturated. Cycloalkyl groups may be substituted or unsubstituted. Exemplary cycloalkyl groups include, without limitation, cyclopropyl, norbornyl, [2.2.2]bicyclooctane, and [4.4.0]bicyclodecane, and the like, which, as in the case of other alkyl moieties, may optionally be substituted.
  • cycloalkenyl refers to cyclic or polycyclic hydrocarbon groups of from 3 to 13 carbon atoms, preferably from 5 to 8 carbon atoms, containing one or more double bonds. Cycloalkenyl groups may be substituted or unsubstituted. Exemplary cycloalkenyl groups include, without limitation, cyclopentenyl, cyclohexenyl, and cyclooctenyl.
  • cycloalkynyl refers to cyclic or polycyclic hydrocarbon groups of from 5 to 13 carbon atoms containing one or more triple bonds. Cycloalkynyl groups may be substituted or unsubstituted.
  • heteroalkyl means a branched or unbranched alkyl, alkenyl, or alkynyl group having from 1 to 14 carbon atoms in addition to 1, 2, 3 or 4 heteroatoms independently selected from the group consisting of N, O, S, and P.
  • Heteroalkyls include, without limitation, tertiary amines, secondary amines, ethers, thioethers, amides, thioamides, carbamates, thiocarbamates, hydrazones, imines, phosphodiesters, phosphoramidates, sulfonamides, and disulfides.
  • a heteroalkyl may optionally include monocyclic, bicyclic, or tricyclic rings, in which each ring desirably has three to six members.
  • the heteroalkyl group may be substituted or unsubstituted.
  • heteroalkyls include, without limitation, polyethers, such as methoxymethyl and ethoxyethyl.
  • heterocyclic ring and “heterocyclyl” refer to non-aromatic ring systems having five to fourteen ring atoms in which one or more ring carbons, preferably one to four, are each replaced by a heteroatom such as N, O, S, or P, which may be used alone or as part of a larger moiety as in “heterocyclyl-alkyl” (a heterocyclyl-substituted C 1-6 alkyl), “heterocyclyl-alkoxy” (a heterocyclyl-substituted C 1-6 alkoxy), or “heterocycloxy-alkyl” (a heterocycloxy-substituted C 1-6 alkyl), and includes aralkyl, aralkoxy, and aryloxyalkyl groups.
  • Heterocyclic rings may be substituted or unsubstituted and may include one, two, or three fused or unfused ring systems.
  • the heterocyclic ring is a 5- to 7-membered monocyclic or 7- to 14-membered bicyclic heterocyclic ring consisting of 2 to 6 carbon atoms and 1, 2, 3, or 4 heteroatoms independently selected from N, O, and S and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • heterocyclic rings include, without limitation, 3-1H-benzimidazol-2-one, (1-substituted)-2-oxo-benzimidazol-3-yl, 2-tetrahydrofuranyl, 3-tetrahydrofuranyl, 2-tetrahydrothiophenyl, 3-tetrahydrothiophenyl, 2-morpholinyl, 3-morpholinyl, 4-morpholinyl, 2-thiomorpholinyl, 3-thiomorpholinyl, 4-thiomorpholinyl, 1-pyrrolidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 1-piperazinyl, 2-piperazinyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 4-thiazolidinyl, diazolonyl, N-substituted diazolonyl, 1-phthalimidinyl, benzoxanyl, benzopyrrolidinyl, benzopiperi
  • a heterocyclyl group can include two or more of the ring systems listed above.
  • Heterocyclic rings include those in which a non-aromatic heteroatom-containing ring is fused to one or more aromatic or non-aromatic rings, such as in an indolinyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl, where the radical or point of attachment is on the non-aromatic heteroatom-containing ring.
  • aryl used alone or as part of a larger moiety as in “aralkyl” (an aryl-substituted C 1-6 alkyl), “aralkoxy” (an aryl-substituted C 1-6 alkoxy), or “aryloxyalkyl” (an aryloxy-substituted C 1-6 alkyl), refers to aromatic monocyclic or polycyclic ring groups having six to fourteen ring atoms, such as phenyl, 1-naphthyl, 2-naphthyl, 1-anthracyl, and 2-anthracyl and includes aralkyl, aralkoxy, and aryloxyalkyl groups.
  • aryl may be substituted or unsubstituted.
  • aryl includes fused polycyclic aromatic ring systems in which an aromatic ring is fused to one or more rings.
  • Non-limiting examples of aryl groups include phenyl, hydroxyphenyl, halophenyl, alkoxyphenyl, dialkoxyphenyl, trialkoxyphenyl, alkylenedioxyphenyl, naphthyl, phenanthryl, anthryl, phenanthro, 1-naphthyl, 2-naphthyl, 1-anthracyl, and 2-anthracyl.
  • aryl is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as in a indanyl, phenanthridinyl, or tetrahydronaphthyl, where the radical or point of attachment is on the aromatic ring.
  • heteroaryl refers to stable heterocyclic, and polyheterocyclic aromatic moieties having 5-14 ring atoms. Heteroaryl groups may be substituted or unsubstituted and include both monocyclic and polycyclic ring systems.
  • heteroaryl rings include 5-membered monocyclic rings, such as thienyl, pyrrolyl, imidazolyl, pyrazolyl, furyl, isothiazolyl, furazanyl, isoxazolyl, and thiazolyl; 6-membered monocyclic rings, such as pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, and triazinyl; and polycyclic heterocyclic rings, such as benzo[b]thienyl, naphtho[2,3-b]thienyl, thianthrenyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxathienyl, indolizinyl, isoindolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolyl, phthalazinyl, naphthyridinyl, quinox
  • heteroaryl rings include, without limitation, 2-furanyl, 3-furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxadiazolyl, 5-oxadiazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-pyrimidyl, 3-pyridazinyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 5-tetrazolyl, 2-triazolyl, 5-triazolyl, 2-thienyl, 3-thienyl, carbazolyl, benzimidazoly
  • Heteroaryl groups further include a group in which a heteroaromatic ring is fused to one or more aromatic or nonaromatic rings where the radical or point of attachment is on the heteroaromatic ring, such as tetrahydroquinoline, tetrahydroisoquinoline, and pyrido[3,4-d]pyrimidinyl, imidazo[1,2-a]pyrimidyl, imidazo[1,2-a]pyrazinyl, imidazo[1,2-a]pyiridinyl, imidazo[1,2-c]pyrimidyl, pyrazolo[1,5-a][1,3,5]triazinyl, pyrazolo[1,5-c]pyrimidyl, imidazo[1,2-b]pyridazinyl, imidazo[1,5-a]pyrimidyl, pyrazolo[1,5-b][1,2,4]triazine, quinolyl, isoquinolyl, quinox
  • An aryl group or heteroaryl group may contain one or more substituents.
  • substituents for aryl or heteroaryl group include halogen (F, Cl, Br or I), alkyl, alkenyl, alkynyl, heteroalkyl, —NO 2 , —CN, —R A , —OR B , —S(O) r R B , (wherein r is 0, 1 or 2), —SO 2 NR A R B , —NR A R B , —O—NR A R B , —NR A —NR A R B , —(CO)YR B , —O(CO)YR B , —NR A (CO)YR B , —S(CO)YR B , —NR A C( ⁇ S)YR B , —OC( ⁇ S)YR B , —C( ⁇ S)YR B , —YC( ⁇ NR A )YR B , —YC(
  • R C is selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, and heterocyclyl.
  • each of R A and R B is, independently, selected from hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, heteroaryl, and heterocyclyl.
  • R A , R B and R C optionally bears one or more substituents selected from amino, alkylamino, dialkylamino, aminocarbonyl, halogen, alkyl, aryl, heteroalkyl, heteroaryl, carbocycle, heterocycle, alkylaminocarbonyl, dial kylaminocarbonyl, alkylaminocarbonyloxy, dialkylaminocarbonyloxy, nitro, cyano, carboxy, alkoxycarbonyl, alkylcarbonyl, alkoxy, haloalkoxy groups, hydroxy, protected hydroxyl groups (e.g., —O—X, where X is acyl, phenyl, substituted phenyl, benzyl, substituted benzyl, phenethyl, or substituted phenethyl), -M-heteroaryl, -M-heterocycle, -M-aryl, -M-OR B , -M-SR B
  • Non-limiting illustrations of a substituted R A , R B or R C group include haloalkyl and trihaloalkyl, alkoxyalkyl, halophenyl, chloromethyl, trichloromethyl, trifluoromethyl, methoxyethyl, alkoxyphenyl, halophenyl, —CH 2 -aryl, —CH 2 -heterocycle, —CH 2 C(O)NH 2 , —C(O)CH 2 N(CH 3 ) 2 , —CH 2 CH 2 OH, —CH 2 OC(O)NH 2 , —CH 2 CH 2 NH 2 , —CH 2 CH 2 CH 2 NEt 2 , —CH 2 OCH 3 , —C(O)NH 2 , —CH 2 CH 2 -heterocycle, —C( ⁇ S)CH 3 , —C( ⁇ S)NH 2 , —C( ⁇ NH)NH 2 , —C( ⁇ NH
  • alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, heteroalkyl, cycloalkyl, cycloalkenyl, cycloalkynyl, or heterocyclic group may contain one or more substituents selected from those listed above for aryl and heteroaryl groups, in addition to ⁇ O, ⁇ S, ⁇ NH, ⁇ NNR A R B , ⁇ NNHC(O)R B , ⁇ NNHCO 2 RB, or ⁇ NNHSO 2 RB, wherein R A and R B are as defined above.
  • the EGFR-driven cancers which can be treated using the compositions and method of the invention include, for example, EGFR mutants including one or more deletions, substitutions, or additions in the amino acid or nucleotide sequences of EGFR, or fragments thereof.
  • Mutations in EGFR can occur in any part of the EGFR sequence.
  • EGFR mutants arise from mutations in the kinase domain (i.e., exons 18-24 in the EGFR sequence) or in the extracellular domain (i.e., exons 2-16 in the EGFR sequence).
  • mutations typically occur in the kinase domain, including one or more of a point mutation in exon 18 (e.g., L688P, V689M, P694L/S, N700D, L703V, E709K/Q/A/GN, 1715S, L718P, G719C/A/S/R, or S720P/F), a deletion in exon 19 that may or may not include an insertion (e.g., delG719, delE746_E749, delE746_A750, delE746_A750insRP, delE746_A750insQP, delE746_T751, delE746_T751insA/I/V, delE746_T751insVA, delE746_S752, delE746_S752insAN/D, delE746_P53insLS, delL747_E749
  • activation mutants are typical, and 90% deletion of 746-750 (ELREA) and L858R result in sustained phosphorylation of EGFR without ligand stimulation.
  • ELREA 746-750
  • L858R L858R
  • mutations typically, but not exclusively, occur in the extracellular domain, including EGFR variant I (EGFRvI) lacking the extracellular domain and resembling the v-erbB oncoprotein; EGFRvII lacking 83 amino acids from domain IV; and EGFRvIII lacking amino acids 30-297 from domains I and II, which is the most common amplification and is reported in 30-50% of glioblastomas and 5% of squamous cell carcinoma.
  • EGFRvI EGFR variant I
  • EGFRvIII lacking 83 amino acids from domain IV
  • EGFRvIII lacking amino acids 30-297 from domains I and II, which is the most common amplification and is reported in 30-50% of glioblastomas and 5% of squamous cell carcinoma.
  • Other mutations for glioblastoma include one or more of point mutations in exon 2 (e.g., D46N or G63R), exon 3 (e.g., R108K in domain I), exon 7 (e.g., T263P or A289D/TN in domain II), exon 8 (e.g., R324L or E330K), exon 15 (e.g., P596L or G598V in domain IV), or exon 21 (L861Q in the kinase domain).
  • exon 2 e.g., D46N or G63R
  • exon 3 e.g., R108K in domain I
  • exon 7 e.g., T263P or A289D/TN in domain II
  • exon 8 e.g., R324L or E330K
  • exon 15 e.g., P596L or G598V in domain IV
  • exon 21 L861Q in the kinase domain
  • EGFR mutants also include those with a combination of two or more mutations, as described herein.
  • Exemplary combinations include S7681 and G719A; S7681 and V769L; H773R and W731Stop; R776G and L858R; R776H and L861Q; T790M and L858R; T790M and delE746_A750; R803W and delE746_T751insVA; delL747_E749 and A750P; delL747_S752 and E746V; delL747_S752 and P753S; P772_H773insYNP and H773Y; P772_H773insNP and H773Y; and D770_N771insG and N771T.
  • T790M e.g., T790M and L858R or T790M and delE746_A750, with or without concomitant inhibition of single mutants L858R and delE746_A750.
  • EGFR mutants can be either activation mutants or resistant mutants.
  • Activation mutants include those with substitutions that increase drug sensitivity (e.g., G719C/S/A, delE746_A750, or L858R).
  • Resistant mutants include those with substitutions that increase drug resistance (e.g., T790M or any combination including T790M).
  • EGFR-driven cancers include those having any mutant described herein.
  • EGFRvIII is commonly found in glioblastoma and has also been reported in breast, ovarian, prostate, and lung carcinomas.
  • Exemplary EGFR-driven cancers glioblastoma, lung cancer (e.g., squamous cell carcinoma, non-small cell lung cancer, adenocarcinoma, bronchioloalveolar carcinoma (BAC), BAC with focal invasion, adenocarcinoma with BAC features, and large cell carcinoma), pancreatic cancer, head and neck cancers (e.g., squamous cell carcinoma), breast cancer, colorectal cancer, epithelial cancer (e.g., squamous cell carcinoma), ovarian cancer, and prostate cancer.
  • lung cancer e.g., squamous cell carcinoma, non-small cell lung cancer, adenocarcinoma, bronchioloalveolar carcinoma (BAC), BAC with focal invasion, adenocar
  • the invention described herein would benefit patient populations having higher risk for TKI-resistant mutations.
  • About 8,000 to 16,000 new cases per year can be estimated based on: incidence of non-small cell lung cancer (about 160,000 new cases in the U.S.), the response to erlonitinib in the general population (about 10%, resulting in a sensitive population of 16,000), the presence of activation mutations (10-20% in white and 30-40% in Asian population, resulting in a sensitive population of 16,000-32,000), acquisition of secondary resistance (most if not all patients, resulting in a sensitive population of 16,000-32,000), and percentage of patients carrying the T790M point mutations (about 50%, resulting in a sensitive population of 8,000-16,000).
  • Patients having TKI-resistant mutations include those patients having cancers resistant to one or more of erlotinib, gefitinib, CL-387,785, BIBW 2992 (CAS Reg. No. 439081-18-2), Cl-1033, neratinib (HKI-272), MP-412 (AV-412), PF-299804, AEE78, and XL64.
  • the inventions relates to treatment of EGFR-driven cancers having the T790M point mutation.
  • reversible inhibitors e.g., C1-1033, neratinib (HKI-272), and PF-299804
  • C1-1033, neratinib (HKI-272), and PF-299804 are less potent in cell lines having the T790M mutation and do not inhibit T790M at clinically achievable concentrations. Since the ATP Km of T790M and WT are similar, concentrations that inhibit the mutant will inhibit the WT and result in gastrointestinal and cutaneous events.
  • An EGFR mutant also includes other amino acid and nucleotide sequences of EGFR with one or more deletions, substitutions, or additions, such as point mutations, that retain or increase tyrosine kinase or phosphorylation activity.
  • preferable substitutions are conservative substitutions, which are substitutions between amino acids similar in properties such as structural, electric, polar, or hydrophobic properties.
  • the substitution can be conducted between basic amino acids (e.g., Lys, Arg, and His), or between acidic amino acids (e.g., Asp and Glu), or between amino acids having non-charged polar side chains (e.g., Gly, Asn, Gin, Ser, Thr, Tyr, and Cys), or between amino acids having hydrophobic side chains (e.g., Ala, Val, Leu, Ile, Pro, Phe, and Met), or between amino acids having branched side chains (e.g., Thr, Val, Leu, and Ile), or between amino acids having aromatic side chains (e.g., Tyr, Trp, Phe, and His).
  • basic amino acids e.g., Lys, Arg, and His
  • acidic amino acids e.g., Asp and Glu
  • amino acids having non-charged polar side chains e.g., Gly, Asn, Gin, Ser, Thr, Tyr, and Cys
  • amino acids having hydrophobic side chains e.
  • the DNA encoding an EGFR mutant protein may comprise a nucleotide sequence capable of hybridizing to a complement sequence of the nucleotide sequence encoding an EGFR mutant, as defined herein, under stringent conditions.
  • the stringent conditions include low, medium or high stringent conditions.
  • An example of the stringent conditions includes hybridization at approximately 42-55° C. in approximately 2-6 ⁇ SSC, followed by wash at approximately 50-65° C. in approximately 0.1-1 ⁇ SSC containing approximately 0.1-0.2% SDS, where 1 ⁇ SSC is a solution containing 0.15 M NaCl and 0.015 M Na citrate, pH 7.0. Wash can be performed once or more.
  • stringent conditions may be set at a temperature approximately 5° C. lower than a melting temperature (Tm) of a specific nucleotide sequence at defined ionic strength and pH.
  • Tm melting temperature
  • GenBank accession numbers for EGFR include MIM131550, AA128420, NM — 005228, NP — 005219.2, and GeneID: 1956.
  • compositions and methods of the invention can be used to treat subjects having an EGFR-driven cancer (i.e., cancers characterized by EGFR mutant expression or overexpression).
  • EGFR mutant expression or overexpression can be determined in a diagnostic or prognostic assay by evaluating levels of EGFR mutants in biological sample, or secreted by the cell (e.g., via an immunohistochemistry assay using anti-EGFR antibodies or anti-p-EGFR antibodies; FACS analysis, etc.).
  • FISH fluorescent in situ hybridization using a nucleic acid based probe corresponding to an EGFR mutant-encoding nucleic acid or the complement thereof
  • FISH fluorescent in situ hybridization using a nucleic acid based probe corresponding to an EGFR mutant-encoding nucleic acid or the complement thereof
  • PCR polymerase chain reaction
  • RT-PCR real time quantitative PCR
  • various in vivo assays are available to the skilled practitioner. For example, one can expose cells within the body of the mammal to an antibody which is optionally labeled with a detectable label, e.g., a radioactive isotope, and binding of the antibody to cells in the mammal can be evaluated, e.g., by external scanning for radioactivity or by analyzing a biopsy taken from a mammal previously exposed to the antibody.
  • a detectable label e.g., a radioactive isotope
  • Examples of biological properties that can be measured in isolated cells include mRNA expression, protein expression, and DNA quantification. Additionally, the DNA of cells isolated by the methods of the invention can be sequenced, or certain sequence characteristics (e.g., polymorphisms and chromosomal abnormalities) can be identified using standard techniques, e.g., FISH or PCR. The chemical components of cells, and other analytes, may also be assayed after isolation. Cells may also be assayed without lysis, e.g., using extracellular or intracellular stains or by other observation, e.g., morphology or growth characteristics in various media.
  • sequence characteristics e.g., polymorphisms and chromosomal abnormalities
  • FISH fluorescent in situ hybridization
  • FISH is a cytogenetic technique which can be used to detect and localize the presence or absence of specific DNA or RNA sequences on chromosomes.
  • FISH incorporates the use of fluorescently labeled nucleic acid probes which bind only to those parts of the chromosome with which they show a high degree of sequence similarity.
  • Fluorescence microscopy can be used to find out where the fluorescent probe bound to the chromosome.
  • the basic steps of FISH are outlined below.
  • Exemplary FISH probes include Vysis EGFR SpectrumOrangel CEP SpectrumGreen Probe (Abbott, Downers Grove, Ill.), which hybridizes to band 7p12; and ZytoLight SPEC EGFR/CEN 7 Dual Color Probe (ZytoVision), which hybridizes to the alpha-satellite sequences of the centromere of chromosome 7.
  • Probes are generally labeled with fluorophores, with targets for antibodies, with biotin, or any combination thereof. This can be done in various ways, for example using random priming, nick translation, and PCR using tagged nucleotides.
  • a sample or aliquot of a population of cells is used for FISH analysis.
  • cells are trypsinized to disperse into single cells, cytospun onto glass slides, and then fixed with paraformaldehyde before storing in 70% ethanol.
  • the chromosomes are firmly attached to a substrate, usually glass. After preparation, the probe is applied to the chromosome RNA and starts to hybridize. In several wash steps, all unhybridized or partially hybridized probes are washed away.
  • An epifluorescence microscope can be used for observation of the hybridized sequences.
  • the white light of the source lamp is filtered so that only the relevant wavelengths for excitation of the fluorescent molecules arrive onto the sample.
  • Emission of the fluorochromes happens, in general, at larger wavelengths, which allows one to distinguish between excitation and emission light by mean of another optical filter. With a more sophisticated filter set, it is possible to distinguish between several excitation and emission bands, and thus between several fluorochromes, which allows observation of many different probes on the same strand.
  • FISH can have resolution ranging from huge chromosomes or tiny ( ⁇ 100 kilobase) sequences.
  • the probes can be quantified simply by counting dots or comparing color.
  • Allele-specific quantitative real time-PCR may also be used to identify a nucleic acid encoding a mutant EGFR protein (see, for e.g., Diagnostic Innovations DxS BCR-ABL T3151 Mutation Test Kit, and Singer et al., Methods in Molec. Biol. 181:145 (2001)).
  • This technique utilizes Taq DNA polymerase, which is extremely effective at distinguishing between a match and a mismatch at the 3′-end of the primer (when the 3′-base is mismatched, no efficient amplification occurs).
  • the 3′-end of the primer may be designed to specifically hybridize to a nucleic acid sequence that corresponds to a codon that encodes a mutant amino acid in an EGFR mutant, as described herein.
  • the specific mutated sequences can be selectively amplified in a patient sample.
  • This technique further utilizes a Scorpion probe molecule, which is a bifunctional molecule containing a PCR primer, a fluorophore, and a quencher. The fluorophore in the probe interacts with a quencher, which reduces fluorescence.
  • the Scorpion probe binds to the amplicon, the fluorophore and quencher in the Scorpion probe become separated, which leads to an increase in fluorescence from the reaction tube.
  • Any of the primers described herein may be used in allele-specific quantitative real time PCR.
  • a biological sample can be analyzed to detect a mutation in an EGFR gene, or expression levels of an EGFR gene, by methods that are known in the art. For example, methods such as direct nucleic acid sequencing, altered hybridization, aberrant electrophoretic gel migration, binding or cleavage mediated by mismatch binding proteins, single-strand conformational polymorphism (SSCP) analysis, or restriction fragment length polymorphism (RFLP) analysis of PCR products derived from a patient sample can be used to detect a mutation in an EGFR gene; ELISA can be used to measure levels of EGFR polypeptide; and PCR can be used to measure the level of an EGFR nucleic acid molecule.
  • methods such as direct nucleic acid sequencing, altered hybridization, aberrant electrophoretic gel migration, binding or cleavage mediated by mismatch binding proteins, single-strand conformational polymorphism (SSCP) analysis, or restriction fragment length polymorphism (RFLP) analysis of PCR products derived from a patient sample can be used to detect
  • any of these techniques may be used to facilitate detection of a mutation in a candidate gene, and each is well known in the art; examples of particular techniques are described, without limitation, in Orita et al. (Proc. Natl. Acad. Sci. USA 86:2766 (1989)) and Sheffield et al. (Proc. Natl. Acad. Sci. USA 86:232 (1989)).
  • expression of the candidate gene in a biological sample e.g., a biopsy
  • PCR see, e.g., Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York, N.Y. (1995); PCR Technology: Principles and Applications for DNA Amplification, H. A. Ehrlich, Ed., Stockton Press, NY; Yap et al., Nucl. Acids. Res. 19:4294 (1991)).
  • nucleic acid or protein sequence may identify in a nucleic acid or protein sequence a residue (e.g., amino acid or nucleotide) or codon that corresponds to a residue or codon in wild-type EGFR or EGFR mutants using a number of sequence alignment software programs (e.g., NCBI BLAST website). Such software programs may allow for gaps in the alignment of the compared sequences. Using such software, one skilled in the art may identify a nucleotide, amino acid, or amino acid that corresponding to a specific nucleotide, amino acid, or codon in wild-type EGFR or EGFR mutants.
  • sequence alignment software programs e.g., NCBI BLAST website
  • EGFR expression in a biological sample can be determined by using any of a number of standard techniques that are well known in the art or described herein.
  • Exemplary biological samples include plasma, blood, sputum, pleural effusion, bronchoalveolar lavage, or biopsy, such as a lung biopsy and lymph node biopsy.
  • EGFR expression in a biological sample e.g., a blood or tissue sample
  • PCR Technology Principles and Applications for DNA Amplification, H. A. Ehrlich, Ed., Stockton Press, NY; Yap et al., Nucl. Acids. Res. 19:4294 (1991)).
  • Compounds of Formula (I) can be prepared using methods and materials analogous to those described in the art, e.g., as disclosed in detail in International patent applications WO 2004/080980, WO 2005/016894, WO 2006/021454, WO 2006/021457, WO 2009/143389, and WO 2009/126515.
  • compounds of Formula (I) in which R e is H and R d is H, Cl, CF 3 , or CH 3 can be synthesized from 2,4-dichloropyrimidine, 2,4,5-trichloropyrimidine, 2,4-dichloro-5-(trifluoromethyl)pyrimidine, or 2,4-dichloro-5-methylpyrimidine, respectively, as described in PCT Publication No. WO/2009/143389.
  • Compounds of Formula I can be Formulated into a pharmaceutical composition that comprises a compound of Formula I (as an active pharmaceutical ingredient) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.
  • compositions containing a compound of Formula I suitable for administration may be Formulated using conventional materials and methods, a wide variety of which are well known. Suitable dosage forms include those in solution, suspension or emulsion form, and solid oral dosage forms such as capsules, tablets, gel caps, caplets, etc. Methods well known in the art for making Formulations, including the foregoing unit dosage forms, are found, for example, in “Remington: The Science and Practice of Pharmacy” (20th ed., ed. A. R. Gennaro, 2000, Lippincott Williams & Wilkins).
  • Compounds of Formula (I) can be Formulated for any route of administration (e.g., orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by transdermal patch, powders, ointments, or drops), sublingually, bucally, as an oral or nasal spray) effective for use in the methods of the invention.
  • routes of administration e.g., orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by transdermal patch, powders, ointments, or drops), sublingually, bucally, as an oral or nasal spray
  • routes of administration e.g., orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by transdermal patch, powders, ointments, or drops), sublingually, bucally, as an oral or nasal spray
  • compounds of Formula (I) are
  • a compound of Formula (I) can be Formulated for as a capsule for oral administration containing nominally 10 mg, 50 mg, 100 mg, 150 mg, 250 mg, 500 mg, or any dosage amounts described herein as the free base or acid addition salt of the compound (e.g., the hydrochloride salt).
  • the unit dosage forms of the invention can include a compound of the invention, or a salt thereof, Formulated with excipients, fillers, flow enhancers, lubricants, and/or disintegrants as needed.
  • a unit dosage form can include colloidal silicon dioxide (a flow enhancer), lactose anhydrous (a filler), magnesium stearate (a lubricant), microcrystalline cellulose (a filler), and/or sodium starch glycolate (a disintegrant).
  • colloidal silicon dioxide a flow enhancer
  • lactose anhydrous a filler
  • magnesium stearate a lubricant
  • microcrystalline cellulose a filler
  • sodium starch glycolate a disintegrant
  • the compound of the invention and the inactive ingredients can be Formulated utilizing, for example, conventional blending, and encapsulation processes.
  • compounds of Formula (I) are Formulated as described in PCT Publication Nos. WO2009/143389 and WO2009/126515.
  • Compounds of Formula (I) can be useful for treating EGFR-driven cancers.
  • the compounds can be useful for treating EGFR-driven cancers that express EGFR mutants and for treating EGFR-driven cancers that are refractory to TKI therapies (e.g., erlotinib or gefitinib).
  • Such cancers can include, among others, non-small cell lung cancer (NSCLS), including one or more of squamous cell carcinoma, adenocarcinoma, adenocarcinoma, bronchioloalveolar carcinoma (BAC), BAC with focal invasion, adenocarcinoma with BAC features, and large cell carcinoma; neural tumors, such as glioblastomas; pancreatic cancer; head and neck cancers (e.g., squamous cell carcinoma); breast cancer; colorectal cancer; epithelial cancer, including squamous cell carcinoma; ovarian cancer; prostate cancer; adenocarcinomas; and including cancers which are EGFR mediated.
  • NSCS non-small cell lung cancer
  • BAC bronchioloalveolar carcinoma
  • BAC bronchioloalveolar carcinoma
  • BAC BAC with focal invasion, adenocarcinoma with BAC features, and large cell carcinoma
  • neural tumors such as glioblastomas
  • pancreatic cancer head
  • the present invention is based upon the discovery that compounds of Formula (I) can be used to treat EGFR-driven cancers, EGFR-driven cancers that express EGFR mutants, and for treating EGFR-driven cancers that are refractory to TKI therapy, such as erlotinib or gefitinib.
  • Compounds of Formula (I) can also be used in a maintenance role to prevent recurrence of cancer in patients in need of such a treatment.
  • the effective systemic dose of a compound of Formula (I) will typically be in the range of an average daily dose of from 10 mg to 2,000 mg of the compound per kg of patient body weight, administered in single or multiple doses.
  • a compound of the invention may be administered to patients in need of such treatment in a daily dose range of about 50 to about 2,000 mg per patient. Administration may be once or multiple times daily, weekly (or at some other multiple-day interval) or on an intermittent schedule.
  • the compound may be administered one or more times per day on a weekly basis (e.g. every Monday) indefinitely or for a period of weeks, e.g. 4-10 weeks.
  • it may be administered daily for a period of days (e.g.
  • a compound of the invention may be administered daily for 5 days, then discontinued for 9 days, then administered daily for another 5 day period, then discontinued for 9 days, and so on, repeating the cycle indefinitely, or for a total of 4-10 times.
  • each component of the combination therapy may be administered at their monotherapy dosing levels and schedules.
  • erlotinib has been administered orally for the treatment of NSCLC at 150 mg daily and of pancreatic cancer at 100 mg daily.
  • gefitinib has been administered orally for the treatment of NSCLC at 250 mg daily.
  • the effective systemic dose of a compound of the invention will typically be in the range of an average daily dose of from 10 mg to 2,000 mg of the compound per kg of patient body weight, administered in single or multiple doses.
  • a compound of the invention may be administered to patients in need of such treatment in a daily dose range of about 50 to about 2,000 mg per patient. Administration may be once or multiple times daily, weekly (or at some other multiple-day interval) or on an intermittent schedule.
  • the compound may be administered one or more times per day on a weekly basis (e.g. every Monday) indefinitely or for a period of weeks, e.g. 4-10 weeks.
  • it may be administered daily for a period of days (e.g.
  • a compound of the invention may be administered daily for 5 days, then discontinued for 9 days, then administered daily for another 5 day period, then discontinued for 9 days, and so on, repeating the cycle indefinitely, or for a total of 4-10 times.
  • a TKI e.g., erlotinib or gefitinib
  • a compound of Formula (I) with a reduced dosing level in one or both components.
  • the aniline (280 mg, 0.606 mmol) was dissolved in 8 mL of DCM and 0.3 mL of triethylamine was added. The mixture was cooled to ⁇ 35° C. and acryloyl chloride (54.8 mg, 49 ⁇ l, 0.606 mmol, 1.0 eq) was added in portions. The reaction was stirred around ⁇ 30° C. for 15 min and quenched with saturated Na 2 CO 3 . The mixture was worked up with sat. Na 2 CO 3 /DCM and purified with preparation plates to give a light brown solid, 205 mg, in 66% yield.
  • Step 1 Compound 31 was prepared according to the procedure described for the synthesis of compound 1 in Example 1, using 2-iodo-3-methylaniline instead of 2-iodoaniline as the starting material.
  • a suspension of 31 (0.53 mmol), 2,4,5-trichloropyrimidine (1.0 eq), potassium carbonate (1.2 eq), and tetrabutylammonium hydrogensulfate (0.1 eq) in DMF was stirred at 65° C. for 18 hrs. Upon cooling the reaction mixture was filtered and the filtrate was concentrated. The residue was taken up into a mixture of EtOAc and water. After extraction with EtOAc (3 ⁇ ), the combined organic phases were concentrated to give essentially pure material which was used directly in next step reaction.
  • Step 2 A solution of 32 (0.82 mmol), 2-methoxy-5-nitroaniline 33 (1 eq) and TFA (3 eq) in 2-BuOH (3 mL) was heated at 100° C. for 18 hrs. Upon cooling EtOAc and aq. NaHCO 3 were added to the reaction mixture. Extraction (3 ⁇ ) and concentration of combined extracts gave a solid which was purified on silica gel column (ISCO machine) with 10% MeOH in CH 2 Cl 2 as the eluents, furnishing 34 as a brownish solid (55%).
  • Step 3 To a suspension of 34 (0.46 mmol) and zinc powder (6 eq) in acetone (9 mL) and water (1 mL) was added ammonium chloride (10 eq) at 0° C. After the mixture was stirred at room temperature for 30 min, HPLC indicated a complete conversion. Acetone was removed on rotavap and the residue was suspended in DCM and water. Filtration was carried out and the filtrate was extracted with DCM. Concentration of combined organic layers gave crude aniline 35, which was used in the next step without purification.
  • Step 4 To a solution of aniline 35 (0.43 mmol) and N, N-diisopropylethylamine (1.1 eq) in DCM (2 mL) was added acryloyl chloride (1.05 eq) at 0° C. After the mixture was stirred at room temperature overnight, the volatile components were removed on rotavap. The residue was purified on silica gel column with 3% MeOH in DCM as eluents, furnishing amide 36 as beige solid (48 mg, 21%).
  • 2-(Dimethylaminomethyl)acrylic acid 39 was prepared according to a literature procedure ( Synth. Comm. 1995, 25, 641). To a solution of 39 (65 mg, 0.5 mmol), coupling reagent TBTU (1.2 eq) and N, N-diisopropylethylamine (3.0 eq) in DMF (5 mL) and DCM (20 mL) was added 5 (1 eq). After the mixture was stirred at room temperature overnight, the volatile components were removed on rotavap and the residue was purified by reverse phase prep-HPLC, furnishing the title compound as a tan solid (23 mg, 9%).
  • Step 1 the starting material 41 was prepared from 3-fluoro-4-chlorophenol via nitration and subsequent O-methylation, according to a published procedure (US Patent Publication No. 20080300242).
  • the suspension of 41 (1.0 g, 4.86 mmol), 1-methylpiperzine (1 eq) and K 2 CO 3 (1 eq) in DMF (20 mL) was heated at 80° C. for 4 hrs. DMF was removed and the residue was partitioned between DCM and water. Extraction and concentration followed by silica gel column chromatograph (10% MeOH in DCM as eluents) furnished 42 (1.26 g, 91%).
  • Steps 2 and 3 A degassed suspension of 42 (0.96 g, 3.4 mmol), benzophenone imine (1.5 eq), palladium acetate (0.1 eq), xantphos (0.2 eq) and cesium carbonate (1.6 eq) in DMF (20 mL) was heated at 110° C. overnight. Upon cooling the reaction mixture was filtered and the filtrate was concentrated. The solid residue was dissolved in dioxane and 2M aq. HCl (1:1, 40 mL) and then heated at 70° C. for 2 hrs. Upon removing dioxane on rotavap, the water layer was washed with DCM and then basified with aq. NaHCO 3 . Extraction and concentration followed by silica gel column chromatograph (10% MeOH in DCM as eluents) furnished 43 (0.41 g, 45%).
  • Step 4 To a solution of 43 (0.38 g, 1.42 mmol) in THF (15 mL) was added NaH (2 eq.) under N 2 at 0° C. in multiple portions. After bubbles of H 2 were no longer observed, Boc 2 O (4 eq.) was added. The resulting reaction mixture was heated at 50° C. and then refluxed for 2 hrs. The reaction was quenched with MeOH. Usual workup followed by silica gel column chromatograph (5% MeOH in DCM as eluents) furnished 44 (0.46 g, 88%).
  • Step 5 With EtOAc as the solvent 44 (0.46 g) was hydrogenated under 50 psi to afford 45 (0.42 g, 99%). Upon removing the solvent, the crude material was used directly in the next step.
  • Step 6 A degassed suspension of 45 (0.42 g, 3.4 mmol), 2 (1.5 eq), palladium acetate (0.1 eq), xantphos (0.2 eq) and cesium carbonate (1.3 eq) in DMF (10 mL) was heated at 110° C. for 48 hrs. Usual workup followed by silica gel column chromatograph (5% MeOH in DCM as eluents) furnished 46 (0.49 g, 64%).
  • Step 7 To a solution of 46 in DCM was added excessive TFA. After the mixture was stirred at room temperature for 2 hrs, the volatile components were removed on rotavap. The residue was dissolved in EtOAC and the solution was basified with aq. NaHCO 3 . Extraction and concentration gave 47 as tan solid.
  • Step 8 Crude 47 (100 mg) was converted to 48 by using the procedure described in Example 14, step 4. The final product was purified by reverse phase prep-HPLC (10.4 mg, 9%).
  • Step 1 Under N 2 , to a suspension of 41 (0.5 g, 2.43 mmol) and NaH (1.5 eq) in THF (10 mL) was added dropwise a solution of 2-(dimethylamino)ethanol (1.1 eq) in THF (2 mL) at 0° C. The resulting mixture was stirred at room temperature for 3 hrs. Usual workup followed by silica gel column chromatograph (10% MeOH in DCM as eluents) furnished 49 (0.53 g, 79%).
  • Step 2 To a degassed suspension of 49 (0.275 g, 1.0 mmol), Pd 2 (dba) 3 (0.1 eq), 2-(di-t-butylphosphino)-N, N-dimethylbiphenylamine (0.1 eq) and sodium t-butoxide (1.4 eq) in dioxane (10 mL) was added a solution of NH 3 in dioxane (0.5 M in a N 2 -sealed bottle, 10 mL). The resulting mixture was heated at 80° C. for 3 hrs. Usual workup followed by silica gel column chromatograph (15% MeOH in DCM as eluents) furnished 50 (0.15 g, 55%).
  • Steps 3 to 7 The poly-substituted aniline 50 was converted to the title compound 51 according to the procedure described in Example 18 by substituting 50 for 43.
  • 2,4-Dimethoxy-5-nitroaniline 53 was prepared from 1,5-difluoro-2,4-dinitrobenzene via double S N Ar substitution to generate 52 and subsequent mono-reduction of nitro groups, according to a published procedure ( J. Org. Chem. 2005, 70, 10660). This was converted to the title compound 54 as for Example 14 by substituting 53 for 33 and 2 for 32.
  • Step 1 A degassed suspension of 2-nitro-4-bromoanisole (2.32 g, 10 mmol), N,N-dimethyl-1,3-propanediamine (1.1 eq), Pd 2 (dba) 3 (0.02 eq), dppf (0.04 eq) and sodium t-butoxide (1.5 eq) in dioxane (20 mL) was heated at 110° C. overnight. Upon cooling the reaction was quenched with water. The volatile components were removed on rotavap and the residue was partitioned between EtOAc and water. Extraction and concentration followed by silica gel column chromatograph (15% MeOH in DCM as eluents) furnished 55 (0.66 g, 26%).
  • Steps 2 to 6 The secondary amine 55 was converted to the title compound 56 as for Example 18 by substituting 55 for 43.
  • Step 1 Acetic anhydride (7.8 mL, 82.6 mmol) was added dropwise with vigorously stirring to a suspension of 3-fluoro-4-aminophenol (10 g, 78.6 mmol) in water (20 mL). Insoluble amide started to precipitate as a white solid in a few minutes. After the reaction mixture was stirred for 10 more minutes, the white solid was collected via filtration and washed with cold water. Silica gel column chromatograph (5% MeOH in DCM as eluent) gave pure 57 (8.75 g, 66%).
  • Step 2 To a suspension of 57 (8.75 g, 51.73 mmol) and K 2 CO 3 (1.1 eq) in THF (30 mL) was added methyl iodide (1.2 eq). The mixture was heated in a sealed tube at 60° C. overnight. Filtration and concentration followed by silica gel column chromatograph (5% MeOH in DCM as eluent) furnished pure 58 (7.17 g, 89%).
  • Step 3 Nitric acid (70%, 3.83 mL) was added dropwise to a solution of 58 (7 g) in DCM (70 mL) with vigorously stirring. After stirred at room temperature for 1 hr, the reaction mixture was refluxed for 3 hrs. DCM was removed on rotavap and the residue was washed with cold water and then subjected to a silica gel column chromatograph purification (5% MeOH in DCM as eluent) to afford 59 (3.26 g, 37%).
  • Step 4 Poly-substituted nitrobenzene 59 was reduced to corresponding aniline 60 according to the procedure described in Example 14, step 3.
  • Step 5 Poly-substituted aniline 60 (200 mg, 1 mmol) was coupled with precursor 2 (1.5 eq) to afford 61 (320 mg, 67%) via the procedure described in Example 18, step 6.
  • Step 6 N-arylacetamide 61 (320 mg) was heated at reflux in 6N HCl for 30 min. After basification, extraction and concentration aryl amine 62 was obtained (290 mg, 98%).
  • Step 7 Crude 62 (150 mg) was converted to 63 by using the procedure described in Example 14, step 4. The final product was purified by reverse phase prep-HPLC (41 mg, 24%).
  • Step 1 2,4-Dchloro-7H-pyrrolo[2,3-d]pyrimidine (5.0 g, 27 mmol) was suspended in MeCN (300 mL) and AcOH (60 mL); to this was added selectfluor (1-chloromethyl-4-fluoro-1,4-diazoniabicyclo[2.2.2]octane bis(tetrafluoroborate), 1.4 eq, 13.2 g) in one portion. The reaction mixture was stirred at 60° C. overnight. HPLC monitoring indicated complete conversion. After the solvents were evaporated to the volume of ⁇ 100 mL, toluene (20 mL) was added and the suspension was filtered.
  • Step 2 A solution of 64 (1.22 g, 6 mmol) in THF (10 mL) was slowly added to a suspension of NaH (1 eq) in THF (10 mL) at 0° C. After the mixture was stirred for 10 min, a solution of tosyl chloride (1 eq) in THF (5 mL) was slowly added. Stirring was continued for 30 min at 0° C. and then at room temperature overnight. The reaction was shown to be complete via HPLC monitoring and was quenched via the addition of aq. NH 4 Cl (1-2 mL). The reaction mixture was then filtered through celite and the filtrate was evaporated. The crude product was purified via column chromatography (ISCO machine, EtOAc/Heptane, EtOAc on a 0-100% gradient) to give 65 (1.22 g, 56%).
  • Step 3 In a microwave vessel (20 mL) were placed 65 (600 mg, 1.7 mmol), 1 (282 mg, 1.7 mmol) and isopropanol (10 mL). After HCl (1.3 mL, 4 M in dioxane) was added, the resulting mixture was stirred in the microwave reactor at 150° C. for 2 hrs. The solvents were evaporated and the residue was purified via silica column chromatography (ISCO machine, EtOAc/Heptane, 0-100% EtOAc to elute impurities and then MeOH/DCM, 0-20% MeOH) to give pure 66 (900 mg, 54%).
  • Step 4 Intermediate 66 (493 mg, 1 mmol), 2-methoxy-5-nitroaniline 33 (168 mg, 1 mmol), K 2 CO 3 (1.4 mmol), Pd 2 dba 3 (5 mol %) and X-Phos (10 mol %) were weighed into a 100 mL round bottom flask and placed under N 2 .
  • the solvents toluene (10 mL) and tert-butanol (2 mL) were added as a mixture and the stirring solution was evacuated and backfilled with N 2 three times. The resulting mixture was then stirred overnight at 110° C. HPLC showed the reaction to be complete and the solvents were evaporated.
  • the residue was purified by silica column chromatography on ISCO machine (5% MeOH in DCM as eluents) to give coupling product 67 (570 mg, 91%).
  • Step 5 Poly-substituted nitrobenzene 67 was reduced to corresponding aniline 68 according to the procedure described in Example 14, step 3.
  • Step 6 Crude aniline 68 (123 mg) was converted to 69 by using the procedure described in Example 14, step 4. The product was purified by silica column chromatography on ISCO machine (5% MeOH in DCM as eluents). Yield: 69 mg, 51%.
  • Step 7 A solution of 69 (60 mg) and TBAF (1 M in THF, 0.3 mL) in THF (10 mL) was refluxed for 5 hrs. HPLC indicated a complete reaction. After the solvent was evaporated, the residue was purified by silica column chromatography on ISCO machine (5% MeOH in DCM as eluents). The product was co-eluted with TBAF; water wash furnished pure product (10 mg, 21%).
  • Step 1 To a suspension of 5-fluoro-2-nitroanisole (50 mmol, 8.5 g) and zinc powder (3.5 eq, 11.4 g) in acetone (45 mL) and water (5 mL) was added ammonium chloride (11 eq, 29.3 g) at 0° C. in multiple portions. After the mixture was stirred at r.t. overnight, HPLC indicated a complete conversion. Acetone was removed on rotavap and the residue was suspended in DCM and water. Filtration was carried out and the filtrate was extracted with DCM. Concentration of combined organic layers gave crude aniline ( ⁇ 7.0 g), which was used in the next step without purification.
  • Step 2 To a suspension of 4-fluoro-2-methoxyaniline (5.1 g, 36.1 mmol) in concentrated sulfuric acid (55 mL) was added guanidine nitrate (4.38 g, 36.1 mmol) in portion wise under ice cooling over 15 min. The mixture was stirred at the same temperature for additional 15 min. The reaction was then poured into a saturated cold NaHCO 3 solution and the precipitated solid were collected by filtration. The residue was taken up in EtOAc and dried over anhydrous Na 2 SO 4 . The solvent was stripped off to afford the B (4.72 g).
  • Step 3 The above compound B (0.1 g, 0.53 mmol) and 4-(ortho dimethyl phosphinyl anilino)-5-chloro-2-chloro pyrimidine (0.17 g, 0.53 mmol) were dissolved in a mixture of 2-butanol (1.2 mL) & trifluoroacetic acid (0.25 mL) and were heated to 100° C. in a seal tube for overnight. The reaction mixture was then cooled to rt and poured into a saturated NaHCO 3 solution while stirring to afford an orange solid which was filtered, washed with Et 2 O to remove final traces of water. The product was dried to afford C (0.19 g) which was directly used in the next step.
  • Step 4 NaH (0.039 g, 0.96 mmol, 60% dispersion in oil) was taken up in a dry capped microwave vial. To this, 1-(2-hydroxyethyl)-4-methylpiperazine (0.023 g, 0.16 mmol) dissolved in dry tetrahydrofuran (1.6 mL) was added dropwise. The mixture was stirred at rt for 20 min. Intermediate C (0.075 g, 0.16 mmol) was then added in one portion to this suspension and the mixture was heated to 67° C. in the closed seal tube for 25 min. The mixture was allowed to reach at rt and quenched with a few drops of methanol. Solvent was removed under vacuum and the resultant crude was subjected to FCC eluting with DCM-MeOH (95/5) to furnish the desired product D (0.081 g).
  • Step 5 Compound D (0.078 g, 0.13 mmol) was dissolved in a mixture of acetone (1.3 mL) and water (0.3 mL). To this, zinc nano powder (0.07 g, 1.3 mmol) was added immediately followed by addition of NH 4 Cl (0.16 g, 2.6 mmol) in small portions. The mixture was vigorously stirred at r.t for 30 min. Anhydrous Na2SO4 was then added to this stirring mixture and the resultant crude was filtered, solvents were evaporated and the residue were taken up in DCM and directly loaded on the silica gel cartridge and eluted with DCM-MeOH—NH 3 (90/10) to furnish the desired product E (0.044 g).
  • Step 6 To a solution of E (0.044 g, 0.078 mmol) in dry tetrahydrofuran (0.52 mL) was added DIPEA (0.027 mL, 0.156 mmol) at 0° C. under stirring. This was followed by the addition of acryloyl chloride (0.007 g, 0.078 mmol). The reaction was stirred at that temperature for additional 1 h. Solvent was stripped off under vacuum and the crude was purified by FCC using DCM-MeOH—NH 3 (90/10) to furnish a gum which was further triturated with Et 2 O to furnish a solid material F (0.02 g).
  • Step 1 The suspension of advanced intermediate C, (3-dimethylamino)pyrrolidine (1 eq) and K2CO3 (2 eq) in DMF was stirred at r.t. overnight. The solid components were filtered off and the filtrate was concentrated on rotavap and then on vacuum pump. The residue was essentially pure by HPLC analysis and was used directly in the next step.
  • Steps 2 and 3 these were carried out according to the procedure outlined in Scheme 24, steps 5 and 6.
  • Step 1 the starting material A underwent a S N Ar reaction with intermediate 2 according to the procedure described in step 1 of Example 14.
  • Step 2 To a solution of compound B (246 mg, 0.5 mmol), coupling reagent TBTU (1.2 eq) and N, N-diisopropylethylamine (3.0 eq) in DMSO (10 mL) was added piperazine (1 eq). After the mixture was stirred at room temperature overnight, water and DCM was added to facilitate extraction. Combined extracts were washed with water to remove DMSO. After drying over Na2SO4 and concentration on rotavap the residue was purified by silica gel column chromatography with 5% MEOH in DCM as the eluents, furnishing compound C as a tan solid (154 mg, 54%).
  • Step 3 A solution of compound C (570 mg) and BH3-Me2S (2.0 M in THF, 5 eq, 0.6 mL) in THF (5 mL) was stirred at r.t. overnight. 6N aq. HCl (15 mL) was added and the resulting solution was stirred at r.t. for 5 hr. Solid K2CO3 was carefully added to basify the reaction mixture. After extractions with DCM the combined organic phases were dried, concentrated and purified by silica gel column chromatography with 5% MeOH in DCM as the eluents, furnishing compound D as a tan solid (300 mg, 54%).
  • Steps 4 and 5 Compounds C and D were converted into final compounds E and F respectively according to the procedure described in Example 14.
  • Step-1 Synthesis of 61:
  • Step-2 Synthesis of Examples 62, 63 and 64
  • Example 75 (360 mg) in MeOH (1.5 mL) and water (0.5 mL) was treated with K 2 CO 3 (1.0 g) at 50° C. for 2 h. The reaction mixture was cooled down to room temperature. The organic solution was transferred to a new vial and diluted with DCM (3.0 mL). The pH was adjusted to 6-7 by adding hydrochloric acid. The resulting organic layer was evaporated and the residue was washed with MeOH to give product as a yellow solid (304 mg, yield 89%)
  • Step-3 The residue from Step-3 was dissolved in THF (3.0 mL) and Et 3 N (0.05 mL), and the resulting solution was treated with acroyl chloride. The reaction was monitored by HPLC until disappearance of starting material. The reaction was then quenched with aq.NaHCO 3 and extracted with DCM (2 ⁇ 5.0 mL). The organic solution was concentrated and the residue was purified by a prep-TLC plate (10% MeOH/DCM) to give product as a yellow solid (112 mg, yield 45% for two steps).
  • step-1 product 2.4 g
  • MeOH 5.0 mL
  • HCl 10 mL, 2N
  • the mixture was heated to reflux for 15 min.
  • Solvent was evaporated and the residue was dissolved in DCM (6.0 mL).
  • the organic solution was dried, filtered and evaporated to give colorless oil (1.5 g, yield 77%).
  • the nitro compound was synthesized by reacting compound B and the product of step-2 according the general procedure.
  • Step 1 To a solution of benzyl glycidyl ether (5.0 g) in MeOH (5.0 mL) was added NaOMe (5.0 mL, 25% in methanol). The mixture was warmed up to 50° C. for 1 h and then heated to reflux for 5 min. The mixture was treated with wet NaHCO 3 and filtered. Solvent was evaporated and the residue was dissolved in DCM (20 mL). The solution was dried and evaporated to give yellowish oil (4.8 g, yield 80%).
  • Step 2 A solution of step 1 product (3.0 g) in DCM (100 mL) was treated with PDC (6.0 g) and molecular sieves (4.0 g). The mixture was stirred at room temperature for 4 h and diluted with Et 2 O (100 mL). The mixture was filtered through a Celite pad and solvent evaporated to give yellow oil (1.7 g, yield 57%).
  • Step 3 The compound was synthesized according to the following procedure: To a mixture of 2,2-dimethyl-1,3-dioxan-5-one (2.6 g), dimethyl amine HCl salt (1.8 g) in DCM (50 mL) was added NaHB(OAc) 3 (6.0 g) and Et 3 N (3.0 mL). The reaction mixture was stirred at room temperature overnight and then diluted with aq.NaHCO3. The organic layer was dried and evaporated to give colorless oil (2.5 g, yield 79%).
  • Step 4 A solution of step-3 product (1.5 g) in MeOH (10 mL) was charged with Pd—C (0.5 g, 10% wet) and hydrogenated under a hydrogen balloon at room temperature overnight. The catalyst was filtered off and the solvent was evaporated to give the product (0.64 g, yield 71%) as colorless oil.
  • Step 5 The compound was synthesized according the general procedure from compound B and step-4 product as a yellow solid.
  • Example 37 was synthesized according to a similar procedure to the following: To a mixture of tert-butyl (4-(4-((5-chloro-4-((2-(dimethylphosphoryl)phenyl) amino)pyrimidin-2-yl)amino)-5-methoxy-2-nitrophenyl)-2-methylbut-3-yn-2-yl)carbamate (240 mg) in acetone (1.0 mL) and Zn dust (0.3 g), NH 4 Cl (0.15 g), was added drops of water. The mixture was stirring at room temperature for 15 min and then diluted with DCM (5 mL). After filtration, the organic solution was evaporated and the residue was used for next step.
  • Step 1 To a solution of N,N′-dimethylethylenediamine (300 mg) in DMF (2.0 mL) was added K 2 CO 3 (1.0 g) and compound B (466 mg). The mixture was heated at 80° C. for 3 h. Solvent was evaporated and the residue was extracted with DCM and then purified by a prep-TLC plate (10% MeOH/DCM with 1% NH 3 in methanol) to give product as a yellow solid (400 mg, yield 75%).
  • Step 2 A solution of step 1 product (1 eq) in DMF (3.0 mL) was treated with NaHCO 3 (0.5 g) and the respective bromide (2.5 eq) at 50° C. for 5 h. Solvent was evaporated and the products were purified by a prep-TLC plate (8% MeOH/DCM) to give product as yellow solids.
  • Step 3 Reduction of the nitro group and formation of the corresponding acrylamide was carried out according to the procedure in Scheme 32, step-3 and step-4.
  • Step 1 A mixture of N-(2-methoxyethyl)methylamine (1.8 g) and ethyl bromoacetate (3.4 g) in acetonitrile (20 mL) was treated with K 2 CO 3 (4.0 g) and NaI (20 mmol). The mixture was refluxed overnight. Solvent was evaporated and the residue was extracted with DCM and then purified on Silica gel column (0-8% MeOH/DCM) to give the product as colorless oil (3.2 g, yield 93%).
  • Step 2 To a solution of step-1 product (3.5 g) in THF (20 mL) was added LAH (800 mg) portion wise. The resulting mixture was stirred at room temperature overnight and then quenched with EtOAc and water. After filtration, the organic solution was evaporated to give colorless oil (2.0 g, yield 75%).
  • Step 3 The step 3 product compound was synthesized according the general procedure from compound B (400 mg) and step-4 product to give the title compound as a yellow solid (200 mg, yield 40%).
  • Step 4 Example 39 was synthesized according to a similar procedure to that used in Step 6 of Example 37.
  • step-1 product 230 mg
  • MeOH 1.0 mL
  • HCl in dioane 3.0 mL, 4.0M solution
  • Step-4 was carried out using the procedures of Scheme 32 step-3 and step-4 to yield the desired product (Example 68) as a yellow solid (44 mg)
  • step-1 product (1.8 g) and DPPA (2.79 g) in t-BuOH (30 mL) was added Et 3 N (2.02 g). The mixture was refluxed for 20 h. Solvent was evaporated and the residue was column purified on Silica gel (20% Et 2 O)/heptane) to give the product as a white solid (1.3 g, yield 52%).
  • step-2 product 1.2 g
  • THF THF
  • n-Bu 4 NF 6.0 ml, 1.0M solution in THF
  • the mixture was stirred at room temperature for 1.5 h and solvent was evaporated.
  • the residue was column purified on Silica gel (20% Et 2 O)/heptane) to give the product as a white solid (0.75 g, yield 87%).
  • Step-3 The product of Step-3 was then reacted with Compound A using the procedure of Scheme 32, step-2.
  • step-4 product 250 mg
  • DCM dioxane
  • HCl in dioxane 1.5 mL, 4.0M solution
  • solvent was evaporated and the residue was treated with aq.K 2 CO 3 and then purified by a prep-TLC plate (10% MeOH/DCM) to give yellow solid (150 mg).
  • the yellow solid was dissolved in DCM (3.0 mL) and treated with formyl aldehyde (5 drops, 40% aq. solution) and MgSO4 (0.5 g, anhydrous) for 30 min. To the mixture was added NaBH(OAc) 3 (2 eq). The reaction mixture was stirred for 1 h and diluted with aq. NaHCO 3 . The mixture was extracted with DCM and product was and purified by prep-TLC plates (10% MeOH/DCM) to give yellow solid (50 mg, yield 23%).
  • Step 1 To a solution of 3-methoxy-4-nitrobenzyl alcohol (5 g) in DCM (100 mL) was added PDC (1.5 eq) and molecular sieves (6.0 g). The mixture was stirred at room temperature for 2 h and diluted with Et 2 O (100 mL). The mixture was filtered through a Celite pad and solvent was evaporated. The residue was washed with a small amount MeOH to give off white solid (3.7 g, yield 74%).
  • Step 2 To a solution of step-1 product (0.91 g) in DCM (10 mL) was added (Ethoxycarbonylmethlene)triphenylphosphorane (2.0 g). The mixture was stirred at room temperature for 30 min. Solvent was evaporated and the residue was column purified on Silica gel (20% Et 2 O)/heptane) to give a yellowish solid (1.1 g, yield 87%).
  • Step 3 A solution of step-2 product (0.52 g) in MeOH (10 mL) was charged with Pd—C (0.5 g, 10% wet) and hydrogenated under a hydrogen balloon at room temperature overnight. The catalyst was filtered off and solvent was evaporated to give yellow oil (0.45 g, yield 97%).
  • Step 4 A vial was charged with H 2 SO 4 (2.0 mL) and cooled to 0° C. Step-3 product (0.4 g) was carefully introduced. Guanidine nitrate (1 eq) was added. The mixture was stirred at 0° C. for 2 h and at room temperature for 1 h. The mixture was treated with excess wet NaHCO3 and extracted with DCM (10 mL). The product was and purified by prep-TLC plates (8% MeOH/DCM) to give orange solid (0.34 g, yield 71%).
  • Step 5 A solution of compound C (320 mg), step-4 product (268 mg) and TFA (0.3 mL) in 2-BuOH (2 mL) was heated at 100° C. for 18 hrs. Upon cooling EtOAc and aq. NaHCO 3 were added to the reaction mixture. Extraction (3x) and concentration of combined extracts gave a solid which purified by prep-TLC plates (15% MeOH/DCM) to give orange solid (410 mg, yield 71%).
  • Step 6 To a suspension of step-5 product (400 mg) in MeOH (2.0 mL) was added K 2 CO 3 (1.0 g) and water (0.5 mL). The reaction vial was capped and heated at 60° C. for 15 min. The mixture was cooled down to room temperature and the top layer was transferred to a new vial and diluted with water. The pH was adjusted to 5-6 by adding aq HCl (2N) and the product was collected by filtration as a yellow solid (310 mg, yield 86%).
  • Step 7 To a mixture of step-6 product (260 mg) and Et 2 NH (1.1 mmol) in DMF (2.0 mL) was added HBTU (1.3 mmol) and Et 3 N (0.14 mL). The mixture was stirred at room temperature for 2 h and diluted with DCM (5.0 mL). The mixture was washed with aq. K 2 CO 3 and evaporated. The residue was purified by prep-TLC plates (15% MeOH/DCM) to give an orange solid (250 mg, yield 87%).
  • Step 8 To a solution of step-7 product (250 mg) in THF (1.0 mL) was added BH 3 Me 2 S (4.0 mL, 2.0M solution in THF). The mixture was stirred at 60° C. for 2 h and solvent was evaporated. The residue was dissolved in MeOH (2.0 ml) and treated with wet K 2 CO 3 in a capped vial at 70° C. for 1 h. The organic solution was evaporated and the residue was purified by prep-TLC plates (25% MeOH/DCM) to give an orange solid (170 mg, yield 70%).
  • Step 9 Reduction of the nitro group and formation of the corresponding acrylamide was carried out according to the procedure in Scheme 32, step-3 and step-4, to produce the final product.
  • Step 1 N,N-diethyl-2-(3-methoxy-4-nitrophenyl)acetamide was made in accordance with the methods disclosed herein.
  • Step 2 To a solution of N,N-diethyl-2-(3-methoxy-4-nitrophenyl)acetamide (1.0 g) in THF (5.0 mL) was added BH 3 Me 2 S (20. mL, 2.0M solution in THF). The mixture was stirred at 60C for 2 h and solvent was evaporated. The residue was dissolved in MeOH (10 ml) and treated with wet K 2 CO 3 in a capped vial at 70° C. for 1 h. The organic solution was evaporated and the residue was purified on Silica gel column (5% MeOH/DCM) to give the product as an orange oil (0.62 g, yield 65%)
  • Step 3 A solution of step-2 product (600 mg) in MeOH (10 mL) was charged with Pd—C (0.5 g) and hydrogenated under a hydrogen balloon at room temperature for 3 h. The catalyst was filtered off and the solvent was evaporated to give a yellow oil (430 mg, yield 81%)
  • Step 4 The product of step 4 was synthesized according the procedure of step 4 of Scheme 39 as an orange oil.
  • Step 5 The product of step 4 was synthesized according the procedure of step 5 of Scheme 13 to afford an orange solid.
  • Step 6 Reduction of the nitro group and formation of the corresponding acrylamide was carried out according to the procedure in Scheme 32, step-3 and step-4, to produce the final product.
  • Step 1 A mixture of glycidyl methyl ether and aq. methylamine (1.5 eq) was heated at 55° C. for 2 hr. Removal of volatile components gave intermediate A which was used directly in next step.
  • Step 2 Intermediate A (400 mg, 3.34 mmol, 1.0 eq) in 4 ml of dioxane and 4 ml of water was adjusted to pH 12 with 4M NaOH, and cooled to 0° C. O(Boc)2 (806 mg, 3.69 mmol, 1.1 eq) was added in 2 portions and the mixture was stirred at R.T. overnight. The mixture was evaporated to reduce volume by one half and then treated with sat. NaHCO3 (3 ml). This was extracted with EtOAc (3 ⁇ 25 ml), dried, evaporated, and chromatographed (EtOAc/Heptane 1:2 to 1:1) to give an oil, 703 mg, in 96% yield.
  • Steps 3-5 the chemistry was carried out following the procedures described in Scheme 24.
  • Step-1 to a solution of compound 71 (527 mg, 1.0 mmol) and tert-butyl(3-butynyl)carbamate (400 mg) in DMF (4.0 mL), was added (Ph 3 P) 2 PdCl 2 (0.1 g), CuI (0.05 g) and Et 3 N (0.2 mL). This degased mixture was stirred at 80° C. for 3 h. The reaction mixture was concentrated and the residue was purified by a prep-TLC plate (8% MeOH/DCM) to give compound 73 as a yellow solid (280 mg, yield 46%).
  • Step-2 compound 73 (280 mg, 0.46 mmol) was dissolved in DCM (2.0 mL) and treated with HCl in dioxane (1.0 mL, 4.0 M) at room temperature for 20 min. Solvent was evaporated and the residue was treated with aq. sodium bicarbonate and extracted with DCM. Combined organic layers were dried and evaporated to give compound 74 (220 mg, 93%) as yellow solid.
  • Step-4 to a suspension of compound 75 (180 mg) and Zn dust (0.3 g) in acetone (1.0 mL) was added NH 4 Cl (0.15 g) and drops of water. The mixture was stirring at room temperature for 15 min and then diluted with DCM (5 mL). After filtration, the organic solution was evaporated and the residue (140 mg) was used for next step.
  • Example 129 Title compound was prepared in a manner similar to Example 129 using S-(+)-glycidyl methyl ether as starting material.
  • Zinc cyanide 70 mg, 0.65 eq
  • intermediate 27 (490 mg, 1 eq)
  • Pd2(dba)3 46 mg, 0.05 eq
  • DPPF 69 mg, 0.13 eq
  • DMF-H 2 O 3 mL
  • This mixture was flushed with Ar and then heated at 130° C. for 1.5 hr.
  • the reaction mixture was evaporated and the residue was purified on silica gel column (0-5% MeOH in DCM) furnished intermediate A as a yellow solid (32%).
  • Example 133 Title compound was prepared in a manner similar to Example 133 by substituting 2-chloroacetyl chloride for acryloyl chloride in last step.
  • Example 137 Title compound was prepared in a manner similar to Example 137 by substituting 2,4-dichloro-5-methylpyrimidine for 2,4,5-trichloropyrimidine.
  • Steps 1-2 the chemistry was carried out according to the procedure described for steps 2 and 3 in Scheme 24.
  • Step 3 Into a Schlenk flask was loaded intermediate C (260 mg), PdCl2(PPh3)2 (5 mol %) and Cul (10 mol %). This flask was capped with a rubber septum and then degassed under vacuum for 1 min before N2 refill. Anhydrous DMF (4 mL) was added followed by the addition of DIPEA (0.1 mL) and 1-(dimethylamino)-3-butyne (0.1 mL, note this alkyne was purged with dry N2 gas immediately before use). The flask was sealed and the mixture was stirred at 80° C. for 16 h. Upon cooling, the reaction mixture was taken up in EtOAc and water. Extraction and concentration of combined organic layers afforded crude D which was used directly in the next step.
  • Steps 4-5 the chemistry was carried out according to the procedure described for steps 3 and 4 in Scheme 12.
  • Step 1 To a stirred mixture of diethylamine (10.3 ml, 99.6 mmol), 3-chloro-3-methylbut-1-yne (10.2 g, 99.6 mmol), triethylamine (16.7 mL, 119 mmol), and THF (100 mL) at 0° C. was added copper (I) chloride (0.986 g, 9.96 mmol). The resulting suspension was allowed to warm to room temperature, and stirring continued for 4 hours. The reaction mixture was partitioned between diethyl ether (250 mL) and a saturated aqueous solution of NaHCO 3 (100 mL).
  • Steps 2-4 intermediate A was converted into desired final compound in accordance with the method described in Scheme 48 by substituting A for N,N-dimethylpropargylamine.
  • Step 1 to a solution of (N-Boc)propargylamine (3.1 g) in DMF (20 mL) was slowly added NaH (1.1 eq) at 0° C. under N2. After the mixture was stirred at r.t. for 1 hr, methyl iodide (1.1 eq) was added at 0° C. Ice bath was removed and the flask was warmed up naturally to r.t. Water was added to quench the reaction. After removal of solvents the residue was taken up in EtOAc and water. Extraction and concentration of combined organic phases gave essentially pure A (95%).
  • Steps 2-4 Intermediate A was converted into compound B in accordance with the method described in steps 3-5 of Scheme 48 by substituting A for N,N-dimethylpropargylamine.
  • Step 5 to a solution of compound B (189 mg) in DCM (3 mL) was added TFA (1 mL). The mixture was stirred at rt for 1 h. The volatile components were removed under reduced pressure to give a cloudy residue. Trituration with MTBE followed by filtration and wash with MTBE gave the desired product as a off-white powder (92 mg, 57%).
  • Step 1 a solution of appropriate secondary amine and propargyl bromide (supplied as a solution in toluene) in ether was stirred at 0° C. then r.t. for 3 hr. The precipitate was filtered off and the filtrate was carefully distilled to furnish product A. Contamination of A with small amount of toluene was unavoidable in some cases but this did not affect next step reaction. Certain compounds A with relatively high boiling points were purified by silica gel column chromatography (5% MeOH in DCM) to remove residual secondary amine which was found to directly displace the bromide in the next step.
  • Steps 2-4 Crude or pure A was converted into desired final compound in accordance with the method described in Scheme 48 by substituting A for N,N-dimethylpropargylamine.
  • IM1 was prepared according to a literature procedure (WO 2009143389). A solution of IM1 (284 mg, 1.12 mmol) and dimethylamine (1.23 mmol, 1.1 mL, 40% in water) in THF-H 2 O (1:1,4 mL) was stirred at 80° C. overnight. HPLC showed full conversion. The reaction mixture was concentrated under reduced pressure followed by partition between saturated sodium bicarbonate solution and DCM. Upon extraction the combined DCM extracts were dried over anhydrous sodium sulfate. The solvents were removed to give IM2 as yellow viscous oil (315 mg). No further purification was performed.
  • IM2 (315 mg) was reduced into IM3 using standard Zn/NH 4 Cl procedure described previously; a pale yellow oil was obtained (308 mg). This crude material was used directly in the next step.
  • IM4 was prepared according to a literature procedure (Nature 2009, 462, 1070). This material (400 mg) underwent a S N Ar reaction with IM3 (308 mg) under standard condition (TFA, 2-BuOH, 100° C., overnight). Conventional workup followed by flash chromatography on silica gel (eluent: 0-15% MeOH in DCM) afforded IM5 as a white foam (89 mg, 15%).
  • Examples 33-41 were made in accordance with the methods shown in Scheme 26 by substituting secondary amines for (3-dimethylamino)pyrrolidine.
  • Example 54 was made in accordance with the methods shown in Scheme 28.
  • Example 57 was made in accordance with the methods shown in Scheme 30.
  • Example 65 was made in accordance with the methods shown in Scheme 32.
  • Example 66 was made in accordance with the methods shown in Scheme 33.
  • Example 67 was made in accordance with the methods shown in Scheme 35.
  • Example 73 was made in accordance with the methods shown in Scheme 38.
  • Example 74 was made in accordance with the methods shown in Scheme 40.
  • Example 75 was made in accordance with the methods shown in Scheme 41.
  • Example 76 was made in accordance with the methods shown in Scheme 42.
US14/398,893 2012-05-05 2013-03-15 Methods and compositions for raf kinase mediated diseases Abandoned US20150166591A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/398,893 US20150166591A1 (en) 2012-05-05 2013-03-15 Methods and compositions for raf kinase mediated diseases

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201261643228P 2012-05-05 2012-05-05
US201261723779P 2012-11-07 2012-11-07
US201261724283P 2012-11-08 2012-11-08
US201361801953P 2013-03-15 2013-03-15
PCT/US2013/032713 WO2013169401A1 (fr) 2012-05-05 2013-03-15 Composés pour inhiber la prolifération cellulaire dans les cancers induits par l'egfr
US14/398,893 US20150166591A1 (en) 2012-05-05 2013-03-15 Methods and compositions for raf kinase mediated diseases

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/032713 A-371-Of-International WO2013169401A1 (fr) 2012-05-05 2013-03-15 Composés pour inhiber la prolifération cellulaire dans les cancers induits par l'egfr

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/044,382 Continuation US9834571B2 (en) 2012-05-05 2016-02-16 Compounds for inhibiting cell proliferation in EGFR-driven cancers

Publications (1)

Publication Number Publication Date
US20150166591A1 true US20150166591A1 (en) 2015-06-18

Family

ID=49551142

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/398,893 Abandoned US20150166591A1 (en) 2012-05-05 2013-03-15 Methods and compositions for raf kinase mediated diseases
US15/044,382 Active US9834571B2 (en) 2012-05-05 2016-02-16 Compounds for inhibiting cell proliferation in EGFR-driven cancers

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/044,382 Active US9834571B2 (en) 2012-05-05 2016-02-16 Compounds for inhibiting cell proliferation in EGFR-driven cancers

Country Status (4)

Country Link
US (2) US20150166591A1 (fr)
JP (1) JP6469567B2 (fr)
AU (1) AU2013204563B2 (fr)
WO (1) WO2013169401A1 (fr)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9611283B1 (en) 2013-04-10 2017-04-04 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in ALK-driven cancers
US9834571B2 (en) 2012-05-05 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9834518B2 (en) 2011-05-04 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
EP3392245A4 (fr) * 2015-12-15 2019-08-21 Precedo Pharmaceuticals Co., Ltd. Nouveau double inhibiteur de egfr et de alk
CN111718325A (zh) * 2019-03-22 2020-09-29 烟台药物研究所 一种2,4,5-取代嘧啶类化合物及其制备方法和应用
US11034669B2 (en) 2018-11-30 2021-06-15 Nuvation Bio Inc. Pyrrole and pyrazole compounds and methods of use thereof
CN113135920A (zh) * 2016-06-30 2021-07-20 株式会社大熊制药 吡唑并嘧啶衍生物作为激酶抑制剂

Families Citing this family (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9273077B2 (en) 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
CA2723961C (fr) 2008-05-21 2017-03-21 Ariad Pharmaceuticals, Inc. Derives phosphores servant d'inhibiteurs de kinase
US9446064B2 (en) 2013-03-14 2016-09-20 Epizyme, Inc. Combination therapy for treating cancer
PT3060550T (pt) 2013-10-21 2019-08-27 Merck Patent Gmbh Compostos de heteroarilo como inibidores de btk e utilizações dos mesmos
US9879008B2 (en) * 2014-04-14 2018-01-30 Shanghai Haiyan Pharmaceutical Technology Co., Ltd. 2,3,4,6-tetra-substituted benzene-1,5-diamine derivatives, preparation method therefor and medicinal use thereof
SI3157916T1 (sl) * 2014-06-19 2019-05-31 Ariad Pharmaceuticals, Inc. Heteroarilne spojine za zaviranja kinaze
WO2016000581A1 (fr) * 2014-07-04 2016-01-07 南京明德新药研发股份有限公司 Oxyde d'aryl-phosphore et sulfure d'aryl-phosphore spirocycliques
CN105315285B (zh) * 2014-07-25 2017-12-08 上海海雁医药科技有限公司 2,4‑二取代7H‑吡咯并[2,3‑d]嘧啶衍生物、其制法与医药上的用途
WO2016025649A1 (fr) * 2014-08-13 2016-02-18 Celgene Avilomics Research, Inc. Combinaisons d'un inhibiteur d'erk et d'un inhibiteur de dot1l et procédés associés
WO2016025648A1 (fr) * 2014-08-13 2016-02-18 Celgene Avilomics Research, Inc. Combinaisons d'un inhibiteur d'erk et d'un inhibiteur de raf et procédés associés
WO2016025641A1 (fr) * 2014-08-13 2016-02-18 Celgene Avilomics Research, Inc. Combinaisons d'un inhibiteur d'erk et inhibiteur d'egfr et méthodes associées
WO2016025639A1 (fr) * 2014-08-13 2016-02-18 Celgene Avilomics Research, Inc. Combinaisons d'un inhibiteur de erk et d'un agent chimiothérapeutique, et procédés associés
CN111187221B (zh) 2014-10-11 2023-09-26 上海翰森生物医药科技有限公司 Egfr抑制剂及其制备和应用
CN105503827B (zh) * 2014-10-11 2019-09-24 上海翰森生物医药科技有限公司 Egfr抑制剂及其制备方法和用途
CN105601573B (zh) * 2014-11-24 2021-07-02 中国科学院上海药物研究所 2-氨基嘧啶类化合物及其药物组合物和应用
SG10201909060SA (en) * 2014-12-11 2019-11-28 Beta Pharma Inc Substituted 2-anilinopyrimidine derivatives as egfr modulators
WO2016187028A1 (fr) * 2015-05-15 2016-11-24 Celgene Avilomics Research, Inc. Composés hétéroaryles, leur synthèse, et intermédiaires de ceux-ci
CN106187915A (zh) * 2015-05-27 2016-12-07 上海翰森生物医药科技有限公司 具有alk与egfr双重活性的抑制剂及其制备方法和应用
CN104987324B (zh) * 2015-06-04 2018-05-04 湖北生物医药产业技术研究院有限公司 作为alk抑制剂的嘧啶衍生物
CN105085483B (zh) * 2015-06-04 2019-01-01 湖北生物医药产业技术研究院有限公司 激酶抑制剂及其应用
US9695110B2 (en) 2015-06-22 2017-07-04 Eastman Chemical Company Reductive preparation of tertiary dimethylamines from nitriles
US9469595B1 (en) * 2015-06-22 2016-10-18 Eastman Chemical Company Reductive amination of nitriles using transfer hydrogenation
CN106928150B (zh) * 2015-12-31 2020-07-31 恩瑞生物医药科技(上海)有限公司 丙烯酰胺苯胺衍生物及其药学上的应用
CN108473473A (zh) * 2016-05-06 2018-08-31 深圳市塔吉瑞生物医药有限公司 用于抑制蛋白酪氨酸激酶活性的稠合嘧啶类化合物
CN106083736A (zh) * 2016-06-21 2016-11-09 郑州泰基鸿诺医药股份有限公司 一种嘧啶类化合物、egfr抑制剂及其应用
CN107793417B (zh) * 2016-09-05 2020-06-09 复旦大学 吡咯并[2,3-d]嘧啶类化合物及其盐,及制备方法和药用用途
US10294256B2 (en) 2016-12-13 2019-05-21 Bristol-Myers Squibb Company Phosphine oxide alkyl amide substituted heteroaryl compounds as modulators of IL-12, IL-23 and/or IFN alpha responses
CN108498477A (zh) * 2017-02-27 2018-09-07 江苏奥赛康药业股份有限公司 一种2-氨基嘧啶类化合物的药用组合物及其制备方法
EP3656769B1 (fr) * 2017-07-19 2023-01-25 Chia Tai Tianqing Pharmaceutical Group Co., Ltd. Composé aryl-phosphore-oxygène utilisé en tant qu'inhibiteur de kinase egfr
WO2019034153A1 (fr) * 2017-08-18 2019-02-21 北京韩美药品有限公司 Composé chimique, composition pharmaceutique de celui-ci, utilisation et application correspondantes
CN109627263B (zh) * 2017-12-21 2022-05-20 深圳市塔吉瑞生物医药有限公司 用于抑制激酶活性的二苯氨基嘧啶类化合物
CN108017633A (zh) * 2018-01-30 2018-05-11 天津大学 N-[5-(嘧啶-2-氨基)-2,4-二取代苯基]-2-氟代丙烯酰胺衍生物及应用
WO2019177375A1 (fr) * 2018-03-13 2019-09-19 포로노이바이오 주식회사 Dérivés de pyrimidine substitués en positions 2, 4 et 5, leur procédé de préparation et composition pharmaceutique pour la prévention ou le traitement du cancer les contenant en tant que principe actif
JP7138768B2 (ja) * 2018-04-20 2022-09-16 ▲貴▼州伊▲諾▼其尼科技有限公司 ジメチルホスフィンオキシド化合物
CN110856719B (zh) * 2018-08-23 2021-08-10 江苏奥赛康药业有限公司 一种2-氨基嘧啶类化合物的药用组合物
CN113260613B (zh) * 2019-01-18 2022-12-30 正大天晴药业集团股份有限公司 一种egfr抑制剂的盐、晶型及其制备方法
CN111454218A (zh) * 2019-01-22 2020-07-28 烟台药物研究所 一种2,4,5-取代嘧啶类化合物及其制备方法和应用
SG11202110376XA (en) 2019-03-19 2021-10-28 Voronoi Inc Heteroaryl derivative, method for producing the same, and pharmaceutical composition comprising same as effective component
WO2020200191A1 (fr) * 2019-04-04 2020-10-08 Betta Pharmaceuticals Co., Ltd Inhibiteurs d'egfr, compositions et procédés associés
WO2020216371A1 (fr) * 2019-04-26 2020-10-29 江苏先声药业有限公司 Inhibiteur d'egfr et son utilisation
WO2021003417A1 (fr) 2019-07-03 2021-01-07 Sumitomo Dainippon Pharma Oncology, Inc. Inhibiteurs de tyrosine kinase non récepteur 1 (tnk1) et leurs utilisations
WO2021057882A1 (fr) * 2019-09-26 2021-04-01 贝达药业股份有限公司 Inhibiteur d'egfr, composition et son procédé de préparation
CA3163959A1 (fr) * 2020-01-07 2021-07-15 Nathanael S. Gray Inhibiteurs cyano-pyrimidines de l'egfr/her2
CA3167899A1 (fr) * 2020-02-14 2021-08-19 Xiangyong LIU Compose d'oxyde de quinolyle phosphine, et composition et application de celui-ci
WO2022002241A1 (fr) * 2020-07-03 2022-01-06 成都地奥九泓制药厂 Composés d'oxyde d'arylphosphine et leur utilisation
TW202227425A (zh) * 2020-09-18 2022-07-16 南韓商沃若諾伊生物公司 雜芳基衍生物、彼之製法、及包含彼作為活性成份之藥學組成物
CN117412981A (zh) * 2021-06-03 2024-01-16 希格生科(深圳)有限公司 氧化膦衍生物及其制备方法和应用
KR20230054567A (ko) * 2021-10-15 2023-04-25 제이투에이치바이오텍 (주) Alk 및/또는 egfr 돌연변이 키나제 억제 효과를 나타내는 화합물 및 이의 의약 용도

Family Cites Families (285)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1129797A (en) 1966-03-31 1968-10-09 Ici Ltd Phosphorus-containing pyrimidine derivatives and biologically active compositions containing them
AU3934085A (en) 1984-01-30 1985-08-09 Icrf Patents Ltd. Improvements relating to growth factors
EP0387923B1 (fr) 1986-04-17 1994-02-16 Zeneca Limited Fongicides
US5401638A (en) 1986-06-04 1995-03-28 Oncogene Science, Inc. Detection and quantification of neu related proteins in the biological fluids of humans
US4966622A (en) 1988-04-12 1990-10-30 Ciba-Geigy Corporation N-phenyl-N-pyrimidin-2-ylureas
GB8828222D0 (en) 1988-12-02 1989-01-05 Ici Plc Reactive dyes
GB8903019D0 (en) 1989-02-10 1989-03-30 Ici Plc Fungicides
DE68926248T2 (de) 1989-09-29 1996-12-19 Oncogene Science Inc p100 "neu" menschlisches Protein and Verwendung dieses Proteins zum Nachweis von preneoplasmatischen- oder neoplasmatischen beim Menschen
GB9016584D0 (en) 1990-07-27 1990-09-12 Ici Plc Fungicides
EP0472053B1 (fr) 1990-08-20 1998-06-17 Eisai Co., Ltd. Dérivés de sulfonamide
GB9220964D0 (en) 1991-11-15 1992-11-18 Ici Plc Anionic compounds
TW225528B (fr) 1992-04-03 1994-06-21 Ciba Geigy Ag
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
CA2148931A1 (fr) 1993-10-01 1995-04-13 Jurg Zimmermann Derives pyrimidineamine et leurs procedes de preparation
BR9407799A (pt) 1993-10-12 1997-05-06 Du Pont Merck Pharma Composição de matéria método de tratamento e composição farmaceutica
ATE251141T1 (de) 1995-03-10 2003-10-15 Berlex Lab Benzamidin-derivate, deren herstellung und deren verwendung als anti-koagulantien
US5728201A (en) 1995-09-14 1998-03-17 Canon Kabushiki Kaisha Ink, and ink-jet recording method and instruments using the same
GB9519197D0 (en) 1995-09-20 1995-11-22 Affinity Chromatography Ltd Novel affinity ligands and their use
GB9523675D0 (en) 1995-11-20 1996-01-24 Celltech Therapeutics Ltd Chemical compounds
AU7692896A (en) 1995-12-01 1997-06-27 Novartis Ag Quinazolin-2,4-diazirines as NPY receptor antagonist
GB9608771D0 (en) 1996-04-27 1996-07-03 Agrevo Uk Ltd Pyrimethanil salts
JP3106966B2 (ja) 1996-07-17 2000-11-06 富士ゼロックス株式会社 インクジェット記録用インク及びインクジェット記録方法
JPH1060338A (ja) 1996-08-21 1998-03-03 Fuji Xerox Co Ltd インクジェット記録用インク及びインクジェット記録方法
US6008234A (en) 1996-09-12 1999-12-28 Berlex Laboratories, Inc. Benzamidine derivatives substituted by cyclic amino acid and cyclic hydroxy acid derivatives and their use as anti-coagulants
SK29599A3 (en) 1996-09-12 1999-08-06 Schering Ag Benzamidine derivatives substituted by cyclic amino acid or cyclic hydroxy acid derivatives and their use as anti-coagulants
US6004985A (en) 1996-10-09 1999-12-21 Berlex Laboratories, Inc. Thio acid derived monocylic N-heterocyclics as anticoagulants
US5994071A (en) 1997-04-04 1999-11-30 Albany Medical College Assessment of prostate cancer
CO4940418A1 (es) 1997-07-18 2000-07-24 Novartis Ag Modificacion de cristal de un derivado de n-fenil-2- pirimidinamina, procesos para su fabricacion y su uso
US6573044B1 (en) 1997-08-07 2003-06-03 The Regents Of The University Of California Methods of using chemical libraries to search for new kinase inhibitors
JPH1160573A (ja) 1997-08-22 1999-03-02 Nippon Kayaku Co Ltd トリアジン誘導体及びテロメラーゼ阻害剤
AU1507199A (en) 1997-12-15 1999-07-05 Yamanouchi Pharmaceutical Co., Ltd. Novel pyrimidine-5-carboxamide derivatives
DE59905472D1 (de) 1998-06-10 2003-06-12 Basf Ag Verwendung von 2-(n-phenylamino)pyrimidinen als fungizide sowie neue 2-(n-phenylamino)pyrimidine
WO2000015645A1 (fr) 1998-09-11 2000-03-23 Kyorin Pharmaceutical Co., Ltd. Derives d'esters phosphoniques et leur procede de production
OA11674A (en) 1998-11-10 2005-01-12 Janssen Pharmaceutica Nv HIV Replication inhibiting pyrimidines.
DE69927516T2 (de) 1998-11-17 2006-03-16 Kumiai Chemical Industry Co., Ltd. Pyrimidinylbenzimidazol- und triazinylbenzimidazol-derivate und fungizide für landwirtschaft/gartenbau
US6262088B1 (en) 1998-11-19 2001-07-17 Berlex Laboratories, Inc. Polyhydroxylated monocyclic N-heterocyclic derivatives as anti-coagulants
US6127376A (en) 1998-12-04 2000-10-03 Berlex Laboratories, Inc. Aryl and heterocyclyl substituted pyrimidine derivatives as anti-coagulants
CA2362401A1 (fr) 1999-02-04 2000-08-10 Paul Fleming Composes de fixation au recepteur heptahelicoidal couple a la proteine g et leurs procedes d'utilisation
GB9910807D0 (en) 1999-05-10 1999-07-07 Prometic Biosciences Limited Novel detoxification agents and their use
GB9914258D0 (en) 1999-06-18 1999-08-18 Celltech Therapeutics Ltd Chemical compounds
GB9918035D0 (en) 1999-07-30 1999-09-29 Novartis Ag Organic compounds
ITMI992711A1 (it) 1999-12-27 2001-06-27 Novartis Ag Composti organici
WO2001060816A1 (fr) 2000-02-17 2001-08-23 Amgen Inc. Inhibiteurs de kinases
GB0004887D0 (en) 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
GB0004888D0 (en) 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
US7087608B2 (en) 2000-03-03 2006-08-08 Robert Charles Atkins Use of PDGF receptor tyrosine kinase inhibitors for the treatment of diabetic nephropathy
US6525051B2 (en) 2000-03-27 2003-02-25 Schering Aktiengesellschaft N-heterocyclic derivatives as NOS inhibitors
ES2542326T3 (es) 2000-05-08 2015-08-04 Janssen Pharmaceutica Nv Inhibidores de la replicación del VIH
DK1282607T3 (en) 2000-05-08 2016-02-01 Janssen Pharmaceutica Nv Prodrugs of HIV replication inhibiting pyrimidines
WO2002000024A1 (fr) 2000-06-30 2002-01-03 The Regents Of The University Of California Nouvelle technique destinee au traitement de la leucemie
GB0016877D0 (en) 2000-07-11 2000-08-30 Astrazeneca Ab Chemical compounds
GB0022438D0 (en) 2000-09-13 2000-11-01 Novartis Ag Organic Compounds
DE60112964T2 (de) 2000-12-05 2006-02-16 Clariant Finance (Bvi) Ltd., Road Town Trichromie-faerbeverfahren
ATE430742T1 (de) 2000-12-21 2009-05-15 Smithkline Beecham Corp Pyrimidinamine als angiogenesemodulatoren
US6881737B2 (en) 2001-04-11 2005-04-19 Amgen Inc. Substituted triazinyl acrylamide derivatives and methods of use
CA2450769A1 (fr) 2001-06-15 2002-12-27 Vertex Pharmaceuticals Incorporated 5-(2-aminopyrimidine-4-yl) benzisoxazoles en tant qu'inhibiteurs de proteine kinases
WO2003000186A2 (fr) 2001-06-21 2003-01-03 Ariad Pharmaceuticals, Inc. Nouvelles phenylamino-pyrimidines et leur utilisation
GB0115109D0 (en) 2001-06-21 2001-08-15 Aventis Pharma Ltd Chemical compounds
WO2003000251A1 (fr) 2001-06-21 2003-01-03 Ariad Pharmaceuticals, Inc. Nouvelles indolinones et leurs applications
WO2003023010A2 (fr) 2001-09-07 2003-03-20 Irm Llc Recepteurs neuro-sensoriels
WO2003030909A1 (fr) 2001-09-25 2003-04-17 Bayer Pharmaceuticals Corporation 2- et 4-aminopyrimidines n-substituees par un noyau bicyclique utilisees comme inhibiteurs de kinases dans le traitement du cancer
US6949644B2 (en) 2001-10-12 2005-09-27 Irm Llc Methods for the synthesis of substituted purines
AU2002342051B2 (en) 2001-10-12 2009-06-11 Irm Llc Kinase inhibitor scaffolds and methods for their preparation
US20060009642A1 (en) 2001-10-12 2006-01-12 Irm Llc, A Delaware Limited Liability Company Methods for the synthesis of substituted purines
US6770652B2 (en) 2001-10-18 2004-08-03 Duquesne University Of The Holy Ghost Multiple acting anti-angiogenic and cytotoxic compounds and methods for using the same
EP1448556A1 (fr) 2001-11-01 2004-08-25 Janssen Pharmaceutica N.V. Heteroaryl amines utiles comme inhibiteurs de glycogene synthase kinase 3beta (inhibiteurs de gsk3)
US7910586B2 (en) 2002-01-04 2011-03-22 The Rockefeller University Compositions and methods for prevention and treatment of amyloid-β peptide-related disorders
JPWO2003066677A1 (ja) 2002-02-05 2005-05-26 独立行政法人科学技術振興機構 Egf/egfr複合体
AU2003209077A1 (en) 2002-02-08 2003-09-02 Smithkline Beecham Corporation Pyrimidine compounds
GB0206215D0 (en) 2002-03-15 2002-05-01 Novartis Ag Organic compounds
CA2481449A1 (fr) 2002-04-26 2003-11-06 Gilead Sciences, Inc. Technique et compositions permettant d'identifier des composes therapeutiques anti-vih
US20050239054A1 (en) 2002-04-26 2005-10-27 Arimilli Murty N Method and compositions for identifying anti-HIV therapeutic compounds
US7704995B2 (en) 2002-05-03 2010-04-27 Exelixis, Inc. Protein kinase modulators and methods of use
AU2003231231A1 (en) 2002-05-06 2003-11-11 Bayer Pharmaceuticals Corporation Pyridinyl amino pyrimidine derivatives useful for treating hyper-proliferative disorders
AR039540A1 (es) 2002-05-13 2005-02-23 Tibotec Pharm Ltd Compuestos microbicidas con contenido de pirimidina o triazina
WO2003097855A2 (fr) 2002-05-14 2003-11-27 Baylor College Of Medicine Petites molecules inhibitrices de l'expression du gene her2
CA2486853A1 (fr) 2002-05-27 2003-12-04 Novartis Ag Alcanols bi-aromatiques
GB0217431D0 (en) 2002-07-27 2002-09-04 Astrazeneca Ab Novel compounds
US7517886B2 (en) 2002-07-29 2009-04-14 Rigel Pharmaceuticals, Inc. Methods of treating or preventing autoimmune diseases with 2,4-pyrimidinediamine compounds
AU2003255819A1 (en) 2002-08-24 2004-03-11 Astrazeneca Ab Pyrimidine derivatives as modulators of chemokine receptor activity
ATE454378T1 (de) 2002-11-01 2010-01-15 Vertex Pharma Verbindungen, die sich als inhibitoren vonjak und anderen proteinkinasen eignen
JP4505449B2 (ja) 2003-02-11 2010-07-21 アイアールエム・リミテッド・ライアビリティ・カンパニー 新規二環式化合物および組成物
CL2004000303A1 (es) 2003-02-20 2005-04-08 Tibotec Pharm Ltd Compuestos derivados de pirimidinas y triazinas; proceso de preparacion; composicion farmaceutica; y su uso para inhibir la replicacion del vih.
ES2325440T3 (es) 2003-02-20 2009-09-04 Smithkline Beecham Corporation Compuestos de pirimidina.
US7151096B2 (en) 2003-03-05 2006-12-19 Irm Llc Cyclic compounds and compositions as protein kinase inhibitors
GB0305929D0 (en) 2003-03-14 2003-04-23 Novartis Ag Organic compounds
AR043633A1 (es) 2003-03-20 2005-08-03 Schering Corp Ligandos de receptores de canabinoides
GB2400101A (en) 2003-03-28 2004-10-06 Biofocus Discovery Ltd Compounds capable of binding to the active site of protein kinases
US20050014753A1 (en) 2003-04-04 2005-01-20 Irm Llc Novel compounds and compositions as protein kinase inhibitors
WO2004093812A2 (fr) 2003-04-22 2004-11-04 Irm Llc Composes induisant une differentiation neuronale dans des cellules souche embryonnaires
US7273716B2 (en) 2003-04-25 2007-09-25 Gilead Sciences, Inc. Methods and compositions for identifying therapeutic compounds with GS-7340 ester hydrolase
WO2004098520A2 (fr) 2003-05-01 2004-11-18 Irm Llc Composes et compositions utiles en tant qu'inhibiteurs des proteines kinase
GB0311274D0 (en) 2003-05-16 2003-06-18 Astrazeneca Ab Chemical compounds
MY150088A (en) 2003-05-19 2013-11-29 Irm Llc Immunosuppressant compounds and compositions
US7317027B2 (en) 2003-05-19 2008-01-08 Sanofi-Aventis Deutschland Gmbh Azaindole-derivatives as factor Xa inhibitors
EP1651648A4 (fr) 2003-07-29 2009-09-02 Irm Llc Composes et compositions utilises en tant qu'inhibiteurs des proteines kinases
CN102358738A (zh) 2003-07-30 2012-02-22 里格尔药品股份有限公司 2,4-嘧啶二胺化合物及其预防和治疗自体免疫疾病的用途
DE602004032446D1 (de) 2003-08-07 2011-06-09 Rigel Pharmaceuticals Inc 2,4-pyrimidindiamin-verbindungen und verwendungen als antiproliferative mittel
PL1660458T3 (pl) 2003-08-15 2012-07-31 Novartis Ag 2,4-pirymidynodwuaminy stosowane w leczeniu chorób nowotworowych, chorób zapalnych i zaburzeń układu immunologicznego
MXPA06001758A (es) 2003-08-15 2006-08-11 Irm Llc Anilino purinas sustituidas en la posicion 6 utiles como inhibidores de rtk.
US7338957B2 (en) 2003-08-28 2008-03-04 Irm Llc Compounds and compositions as protein kinase inhibitors
GB0321710D0 (en) 2003-09-16 2003-10-15 Novartis Ag Organic compounds
CA2538413A1 (fr) 2003-09-18 2005-03-24 Novartis Ag 2,4-di (phenylamino) pyrimidines utiles dans le traitement de troubles proliferatifs
AU2004278413B2 (en) 2003-09-30 2008-07-31 Irm Llc Compounds and compositions as protein kinase inhibitors
AU2004275888B2 (en) 2003-10-02 2008-09-18 Irm Llc Compounds and compositions as protein kinase inhibitors
MXPA06003996A (es) 2003-10-08 2006-07-05 Irm Llc Compuestos y composiciones como inhibidores de la proteina cinasa.
DE10349587A1 (de) 2003-10-24 2005-05-25 Merck Patent Gmbh Benzimidazolylderivate
MY141255A (en) 2003-12-11 2010-03-31 Memory Pharm Corp Phosphodiesterase 4 inhibitors, including n-substituted diarylamine analogs
EP1694652A1 (fr) 2003-12-19 2006-08-30 Rigel Pharmaceuticals, Inc. Stereoisomeres et melanges stereoisomeriques d'intermediaires synthetiques de 1-(2,4-pyrimidinediamino)-2-cyclopentanecarboxamide
EP1708571A4 (fr) 2004-01-16 2009-07-08 Merck & Co Inc Nouveau sel cristallin d'un inhibiteur de dipeptidyle peptidase-iv
SG149817A1 (en) 2004-01-16 2009-02-27 Wyeth Corp Quinoline intermediates of receptor tyrosine kinase inhibitors and the synthesis thereof
EP1713806B1 (fr) 2004-02-14 2013-05-08 Irm Llc Composes et compositions tenant lieu d'inhibiteurs de la proteine kinase
CA2566531A1 (fr) 2004-05-18 2005-12-15 Rigel Pharmaceuticals, Inc. Composes pyrimidinediamines a substitution cycloalkyle et leurs utilisations
EP1598343A1 (fr) 2004-05-19 2005-11-23 Boehringer Ingelheim International GmbH Dérivés de 2-arylaminopyrimidines comme des inhibiteurs de PLK
TW200610762A (en) 2004-06-10 2006-04-01 Irm Llc Compounds and compositions as protein kinase inhibitors
RU2007102226A (ru) 2004-06-23 2008-07-27 Айрм Ллк (Bm) Соединения и композиции в качестве ингибиторов протеинкиназы
GB0512324D0 (en) 2005-06-16 2005-07-27 Novartis Ag Organic compounds
US7521457B2 (en) 2004-08-20 2009-04-21 Boehringer Ingelheim International Gmbh Pyrimidines as PLK inhibitors
GB0419160D0 (en) 2004-08-27 2004-09-29 Novartis Ag Organic compounds
GB0419161D0 (en) 2004-08-27 2004-09-29 Novartis Ag Organic compounds
WO2006038594A1 (fr) 2004-10-04 2006-04-13 Ono Pharmaceutical Co., Ltd. Inhibiteur de canal de calcium de type n
AU2005295788A1 (en) 2004-10-13 2006-04-27 Wyeth N-benzenesulfonyl substituted anilino-pyrimidine analogs
GT200500321A (es) 2004-11-09 2006-09-04 Compuestos y composiciones como inhibidores de proteina kinase.
GB2420559B (en) 2004-11-15 2008-08-06 Rigel Pharmaceuticals Inc Stereoisomerically enriched 3-aminocarbonyl bicycloheptene pyrimidinediamine compounds and their uses
ES2325035T3 (es) 2004-12-21 2009-08-24 Smithkline Beecham Corporation Inhibidores de quinasa erbb de 2-pirimidinil pirazolopiridina.
EP1841760B1 (fr) 2004-12-30 2011-08-10 Exelixis, Inc. Dérivés de pyrimidine en tant que modulateurs de kinase et méthodes d'emploi
DE602006010979D1 (de) 2005-01-19 2010-01-21 Rigel Pharmaceuticals Inc Prodrugs aus 2,4-pyrimidindiamin-verbindungen und ihre verwendungen
CN101106990B (zh) 2005-01-26 2010-12-08 Irm责任有限公司 用作蛋白激酶抑制剂的化合物和组合物
TW200639163A (en) 2005-02-04 2006-11-16 Genentech Inc RAF inhibitor compounds and methods
BRPI0608513A2 (pt) 2005-03-15 2010-01-05 Irm Llc compostos e composições como inibidores da proteìna quinase
EP1863794A2 (fr) 2005-03-16 2007-12-12 Targegen, Inc. Composes de pyrimidine et methodes d'utilisation
DE102005016634A1 (de) 2005-04-12 2006-10-19 Merck Patent Gmbh Neuartige Aza-Hetercyclen als Kinase-Inhibitoren
WO2006124874A2 (fr) 2005-05-12 2006-11-23 Kalypsys, Inc. Inhibiteurs de la b-raf kinase
EP1891066B1 (fr) 2005-05-13 2010-12-22 Irm, Llc Composes et compositions convenant comme inhibiteurs de proteine kinase
WO2006124863A2 (fr) 2005-05-16 2006-11-23 Irm Llc Composes et compositions en tant qu'inhibiteurs de proteine kinase
EP2049497A2 (fr) 2005-05-19 2009-04-22 Astex Therapeutics Limited Composes pharmaceutiques
WO2006128129A2 (fr) 2005-05-26 2006-11-30 Synta Pharmaceuticals Corp. Traitement anticancereux
WO2006128172A2 (fr) 2005-05-26 2006-11-30 Synta Pharmaceuticals Corp. Procede pour traiter des troubles auto-immuns regules par des lymphocytes b
WO2006129100A1 (fr) 2005-06-03 2006-12-07 Glaxo Group Limited Nouveaux composes
US20070203161A1 (en) 2006-02-24 2007-08-30 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the jak pathway
WO2006133426A2 (fr) 2005-06-08 2006-12-14 Rigel Pharmaceuticals, Inc. Compositions et procedes d'inhibition de la voie jak
US20070032514A1 (en) 2005-07-01 2007-02-08 Zahn Stephan K 2,4-diamino-pyrimidines as aurora inhibitors
BRPI0613501A2 (pt) 2005-07-01 2011-01-11 Irm Llc composto, composição farmacêutica e uso do mesmo
WO2007006926A2 (fr) 2005-07-11 2007-01-18 Sanofi-Aventis Nouveaux derives de 2,4-dianilinopyrimidines, leur preparation, a titre de medicaments, compositions pharmaceutiques et notamment comme inhibiteurs de ikk
KR20080034483A (ko) 2005-07-26 2008-04-21 버텍스 파마슈티칼스 인코포레이티드 단백질 키나제의 억제제로서 유용한 벤즈이미다졸
WO2007016228A2 (fr) 2005-08-02 2007-02-08 Irm Llc Composés et compositions en tant qu’inhibiteurs de kinase de protéine
CA2617359A1 (fr) 2005-08-09 2007-02-22 Irm Llc Composes et compositions utiles comme inhibiteurs de proteines kinases
EP1940843A4 (fr) 2005-08-11 2010-09-15 Ariad Pharma Inc Derives heterocycliques insatures
MX2008002165A (es) 2005-08-16 2008-04-29 Irm Llc Compuestos y composiciones como inhibidores de la proteina cinasa.
US7973060B2 (en) 2005-10-13 2011-07-05 Crystalgenomics, Inc. Fab I inhibitor and process for preparing same
EA200800664A1 (ru) 2005-10-13 2009-02-27 Глаксо Груп Лимитед Производные пирролопиримидина в качестве ингибиторов syk
US8372851B2 (en) 2005-10-21 2013-02-12 Exelixis, Inc. Pyrazolo pyrimidines as casein kinase II (CK2) modulators
ATE444294T1 (de) 2005-10-28 2009-10-15 Irm Llc Verbindungen und zusammensetzungen als proteinkinaseinhibitoren
DK1951684T3 (en) 2005-11-01 2016-10-24 Targegen Inc BIARYLMETAPYRIMIDIN kinase inhibitors
US8133900B2 (en) 2005-11-01 2012-03-13 Targegen, Inc. Use of bi-aryl meta-pyrimidine inhibitors of kinases
US8604042B2 (en) 2005-11-01 2013-12-10 Targegen, Inc. Bi-aryl meta-pyrimidine inhibitors of kinases
CN101300234A (zh) 2005-11-03 2008-11-05 Irm责任有限公司 蛋白激酶抑制剂
SI1948180T1 (sl) 2005-11-11 2013-06-28 Boehringer Ingelheim International Gmbh Kombinacijsko zdravljenje raka, ki obsega EGFR/HER2 inhibitorje
CN101326182B (zh) 2005-12-05 2011-09-28 史密丝克莱恩比彻姆公司 2-嘧啶基吡唑并吡啶ErbB激酶抑制剂
EP1968950A4 (fr) 2005-12-19 2010-04-28 Genentech Inc Inhibiteurs de pyrimidine kinase
DE102005062742A1 (de) 2005-12-22 2007-06-28 Bayer Schering Pharma Ag Sulfoximin substituierte Pyrimidine, Verfahren zu deren Herstellung und ihre Verwendung als Arzneimittel
MX2008008642A (es) * 2006-01-13 2008-09-12 Pharmacyclics Inc Inhibidores de las tirosina cinasas y usos de los mismos.
JO2660B1 (en) 2006-01-20 2012-06-17 نوفارتيس ايه جي Pi-3 inhibitors and methods of use
TW200736232A (en) 2006-01-26 2007-10-01 Astrazeneca Ab Pyrimidine derivatives
KR20080110998A (ko) 2006-01-30 2008-12-22 엑셀리시스, 인코포레이티드 Jak­2 조절자로서 4­아릴­2­아미노­피리미딘 또는 4­아릴­2­아미노알킬­피리미딘 및 이들을 포함하는 약제학적 조성물
AU2007212345A1 (en) 2006-02-06 2007-08-16 Irm Llc Compounds and compositions as protein kinase inhibitors
TW200804364A (en) 2006-02-22 2008-01-16 Boehringer Ingelheim Int New compounds
US20070208164A1 (en) 2006-02-27 2007-09-06 Wyeth Methods of synthesizing radiolabeled 3-cyano[14C]quinolines
EP2004641B1 (fr) 2006-03-30 2010-09-29 Tibotec Pharmaceuticals Pyrimidines substituées 5-(hydroxyméthylène et aminométhylène) inhibant le vih
EP2004632B1 (fr) 2006-03-30 2014-03-12 Janssen R&D Ireland Pyrimidines à substitution 5-amido inhibant le vih
GB0608386D0 (en) 2006-04-27 2006-06-07 Senexis Ltd Compounds
US7601716B2 (en) 2006-05-01 2009-10-13 Cephalon, Inc. Pyridopyrazines and derivatives thereof as ALK and c-Met inhibitors
MX2008014618A (es) 2006-05-15 2008-11-28 Irm Llc Composiciones y metodos para inhibidores de cinasas del receptor fgf.
DE102006027156A1 (de) 2006-06-08 2007-12-13 Bayer Schering Pharma Ag Sulfimide als Proteinkinaseinhibitoren
US8222256B2 (en) 2006-07-05 2012-07-17 Exelixis, Inc. Methods of using IGFIR and ABL kinase modulators
EP2046759A1 (fr) 2006-07-21 2009-04-15 Novartis AG Composés de 2,4-di(arylamino)-pyrimidine-5-carboxamide comme inhibiteurs des jak kinases
GB0614579D0 (en) 2006-07-21 2006-08-30 Black James Foundation Pyrimidine derivatives
DE102006041382A1 (de) 2006-08-29 2008-03-20 Bayer Schering Pharma Ag Carbamoyl-Sulfoximide als Proteinkinaseinhibitoren
WO2008039359A2 (fr) 2006-09-25 2008-04-03 Janssen Pharmaceutica N.V. Inhibiteurs de pyrimidine kinase bicyclique
US8097630B2 (en) 2006-10-10 2012-01-17 Rigel Pharmaceuticals, Inc. Pinane-substituted pyrimidinediamine derivatives useful as Axl inhibitors
CA2598893C (fr) 2006-10-11 2012-04-10 Astellas Pharma Inc. Gene de fusion eml4-alk
WO2008049123A2 (fr) 2006-10-19 2008-04-24 Rigel Pharmaceuticals, Inc. Compositions et procédés pour l'inhibition de la voie jak
NZ576278A (en) 2006-10-19 2011-12-22 Signal Pharm Llc Heteroaryl compounds, compositions thereof, and their use as protein kinase inhibitors
TWI432427B (zh) 2006-10-23 2014-04-01 Cephalon Inc 作為ALK及c-MET抑制劑之2,4-二胺基嘧啶之融合雙環衍生物
WO2008057280A1 (fr) 2006-10-27 2008-05-15 Amgen Inc. Composés à cycles multiples et procédés d'utilisation
AU2007333394C1 (en) 2006-12-08 2011-08-18 Novartis Ag Compounds and compositions as protein kinase inhibitors
EP3012249A1 (fr) 2006-12-08 2016-04-27 Novartis AG Composes et compostions inhibtant la proteine kinase
WO2008079719A1 (fr) 2006-12-19 2008-07-03 Genentech, Inc. Inhibiteurs de pyrimidine kinase
EP1939185A1 (fr) 2006-12-20 2008-07-02 Bayer Schering Pharma Aktiengesellschaft Nouveaux hetaryl-phénylènediamine-pyrimidines en tant qu'inhibiteurs de protéine kinase pour le traitement du cancer
EP2123654A4 (fr) 2006-12-28 2011-05-11 Taisho Pharmaceutical Co Ltd Composé pyrazolopyridimidine
CA2674178C (fr) 2006-12-29 2015-11-10 Tibotec Pharmaceuticals Ltd. Pyrimidines substituees en position 6 inhibant le vih
AU2007341228B2 (en) 2006-12-29 2013-04-18 Janssen Sciences Ireland Uc HIV inhibiting 5,6-substituted pyrimidines
FR2911138B1 (fr) 2007-01-05 2009-02-20 Sanofi Aventis Sa Nouveaux derives de n, n'-2,4-dianilinopyrimidines, leur preparation a titre de medicaments, compositions pharmaceutiques et notamment comme inhibiteurs de ikk
EA200900983A1 (ru) 2007-01-26 2010-02-26 Айрм Ллк Соединения и композиции в качестве ингибиторов киназы
TW200902010A (en) 2007-01-26 2009-01-16 Smithkline Beecham Corp Anthranilamide inhibitors of aurora kinase
ES2702362T3 (es) 2007-01-31 2019-02-28 Ym Biosciences Australia Pty Compuestos a base de tiopirimidina y usos de los mismos
DE102007010801A1 (de) 2007-03-02 2008-09-04 Bayer Cropscience Ag Diaminopyrimidine als Fungizide
MX2009010045A (es) 2007-03-20 2009-12-04 Smithkline Beecham Corp Compuestos quimicos.
AU2008229151A1 (en) 2007-03-20 2008-09-25 Smithkline Beecham Corporation Chemical compounds
US7947698B2 (en) 2007-03-23 2011-05-24 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the JAK pathway
ES2400622T3 (es) 2007-03-30 2013-04-11 Sanofi Compuestos de hidracida pirimidina como inhibidores de PGDS
CA2689989A1 (fr) 2007-06-04 2008-12-11 Avila Therapeutics, Inc. Composes heterocycliques et utilisations de ceux-ci
WO2008154249A2 (fr) 2007-06-08 2008-12-18 Genentech, Inc. Marqueurs d'expression de gène de résistance tumorale à un traitement par inhibiteur her2
CN101687821A (zh) 2007-06-21 2010-03-31 Irm责任有限公司 蛋白激酶抑制剂及其使用方法
TWI389893B (zh) 2007-07-06 2013-03-21 Astellas Pharma Inc 二(芳胺基)芳基化合物
KR20100049068A (ko) 2007-07-17 2010-05-11 리겔 파마슈티칼스, 인크. Pkc 억제제로서의 시클릭 아민 치환된 피리미딘디아민
JP5298128B2 (ja) 2007-08-08 2013-09-25 グラクソスミスクライン エルエルシー 癌の治療用のIGF−1R阻害剤としての2−[(2−{フェニルアミノ}−1H−ピロロ[2,3−d]ピリミジン−4−イル)アミノ]ベンズアミド誘導体
WO2009032668A2 (fr) 2007-08-28 2009-03-12 Irm Llc Composés et compositions en tant qu'inhibiteurs de kinase
WO2009032694A1 (fr) 2007-08-28 2009-03-12 Dana Farber Cancer Institute Pyrimidine substituée par amino, dérivés de pyrollopyridine et de pyrazolopyrimidine utilisés en tant qu'inhibiteurs de la kinase et dans le traitement de troubles prolifératifs et de maladies liées à l'angiogenèse
US7989465B2 (en) 2007-10-19 2011-08-02 Avila Therapeutics, Inc. 4,6-disubstituted pyrimidines useful as kinase inhibitors
EP2214486A4 (fr) * 2007-10-19 2011-03-09 Avila Therapeutics Inc Composés hétéroaryles et leurs utilisations
CN104327084B (zh) 2007-11-28 2017-06-06 达那-法伯癌症研究所 Bcr‑abl的小分子豆蔻酸酯抑制剂及其使用方法
WO2009071535A1 (fr) 2007-12-03 2009-06-11 Boehringer Ingelheim International Gmbh Nouveaux composés
WO2009080638A2 (fr) 2007-12-20 2009-07-02 Cellzome Limited Sulfamides en tant qu'inhibiteurs de zap-70
JP5755449B2 (ja) 2007-12-21 2015-07-29 セルジーン アビロミクス リサーチ, インコーポレイテッド Hcvプロテアーゼ阻害剤およびその使用
US8293705B2 (en) 2007-12-21 2012-10-23 Avila Therapeutics, Inc. HCV protease inhibitors and uses thereof
US8309685B2 (en) 2007-12-21 2012-11-13 Celgene Avilomics Research, Inc. HCV protease inhibitors and uses thereof
TWI487522B (zh) 2007-12-21 2015-06-11 賽基艾維洛米斯研究股份有限公司 Hcv蛋白酶抑制劑及其用途(一)
AR070127A1 (es) 2008-01-11 2010-03-17 Novartis Ag Pirrolo - pirimidinas y pirrolo -piridinas
EP2229217A1 (fr) 2008-01-14 2010-09-22 Irm Llc Compositions et méthodes de traitement de cancers
US20110110923A1 (en) 2008-02-12 2011-05-12 The Brigham And Women's Hospital, Inc. Fish assay for eml4 and alk fusion in lung cancer
AU2009221164B2 (en) 2008-03-05 2012-07-26 Novartis Ag Use of pyrimidine derivatives for the treatment of EGFR dependent diseases or diseases that have acquired resistance to agents that target EGFR family members
US20110098288A1 (en) 2008-03-11 2011-04-28 Jeremy Major Sulfonamides as zap-70 inhibitors
CL2009000600A1 (es) 2008-03-20 2010-05-07 Bayer Cropscience Ag Uso de compuestos de diaminopirimidina como agentes fitosanitarios; compuestos de diaminopirimidina; su procedimiento de preparacion; agente que los contiene; procedimiento para la preparacion de dicho agente; y procedimiento para combatir plagas de animales y/u hongos dañinos patogenos de plantas.
US8518931B2 (en) 2008-04-07 2013-08-27 Irm Llc Compounds and compositions as kinase inhibitors
MX2010010968A (es) 2008-04-07 2010-10-26 Irm Llc Compuestos y composiciones como inhibidores de cinasa de proteina.
WO2009127642A2 (fr) 2008-04-15 2009-10-22 Cellzome Limited Utilisation d’inhibiteurs de lrrk2 pour maladies neurodégénératives
JP2011520785A (ja) 2008-04-16 2011-07-21 バイオリポックス エービー 薬剤としての使用のためのビスアリール化合物
EP2271631B1 (fr) 2008-04-22 2018-07-04 Portola Pharmaceuticals, Inc. Inhibiteurs de protéines kinases
WO2009143018A2 (fr) 2008-05-19 2009-11-26 Plexxikon, Inc. Composés et procédés de modulation des kinases, et indications associées
US20130225528A1 (en) 2008-05-21 2013-08-29 Ariad Pharmaceuticals, Inc. Phosphorus Derivatives as Kinase Inhibitors
CA2723961C (fr) 2008-05-21 2017-03-21 Ariad Pharmaceuticals, Inc. Derives phosphores servant d'inhibiteurs de kinase
US20130225527A1 (en) 2008-05-21 2013-08-29 Ariad Pharmaceuticals, Inc. Phosphorus Derivatives as Kinase Inhibitors
US9273077B2 (en) 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
US20110144107A1 (en) 2008-06-11 2011-06-16 Irm Llc Compounds and compositions useful for the treatment of malaria
MX2010013876A (es) 2008-06-20 2011-03-04 Metabolex Inc Agonistas de arilo grpr119 y sus usos .
US8338439B2 (en) * 2008-06-27 2012-12-25 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
BRPI0914682B8 (pt) 2008-06-27 2021-05-25 Avila Therapeutics Inc compostos de heteroarila e composições compreendendo os referidos compostos
CN102203064A (zh) 2008-09-02 2011-09-28 贝林格尔.英格海姆国际有限公司 新的苯甲酰胺,其制备及其作为药物的用途
JP2012501983A (ja) 2008-09-03 2012-01-26 バイエル・クロップサイエンス・アーゲー 4−アルキル−置換ジアミノピリミジン類
CN102405284B (zh) * 2008-09-05 2016-01-20 新基阿维罗米克斯研究公司 设计不可逆抑制剂的算法
WO2010044885A2 (fr) 2008-10-17 2010-04-22 Whitehead Institute For Biomedical Research Complexes mtor solubles et modulateurs associés
WO2010048149A2 (fr) 2008-10-20 2010-04-29 Kalypsys, Inc. Modulateurs hétérocycliques de gpr119 pour le traitement d'une maladie
US8110578B2 (en) 2008-10-27 2012-02-07 Signal Pharmaceuticals, Llc Pyrazino[2,3-b]pyrazine mTOR kinase inhibitors for oncology indications and diseases associated with the mTOR/PI3K/Akt pathway
CN101723936B (zh) 2008-10-27 2014-01-15 上海睿星基因技术有限公司 激酶抑制剂及其在药学中的用途
US8846664B2 (en) 2008-11-12 2014-09-30 Ariad Pharmaceuticals, Inc. Pyrazinopyrazines and derivatives as kinase inhibitors
EP2381777A4 (fr) 2008-12-12 2012-08-08 Ariad Pharma Inc Dérivés d'azaindole en tant qu'inhibiteurs de kinases
US8809343B2 (en) 2008-12-26 2014-08-19 Fudan University Pyrimidine derivative, preparation method and use thereof
EP3255047B1 (fr) 2009-01-06 2021-06-30 Dana-Farber Cancer Institute, Inc. Composés d'échafaudage de kinase pyrimido-diazépinone et procédés de traitement de troubles
US9364477B2 (en) 2009-02-12 2016-06-14 Cell Signaling Technology, Inc. Mutant ROS expression in human cancer
US20100222381A1 (en) 2009-02-27 2010-09-02 Hariprasad Vankayalapati Cyclopentathiophene/cyclohexathiophene DNA methyltransferase inhibitors
US20110071158A1 (en) 2009-03-18 2011-03-24 Boehringer Ingelheim International Gmbh New compounds
WO2010111406A2 (fr) 2009-03-24 2010-09-30 Myriad Pharmaceuticals, Inc. Composés et leurs utilisations thérapeutiques
US20120270237A9 (en) 2009-04-03 2012-10-25 Cellzone AG Methods for the identification of kinase interacting molecules and for the purification of kinase proteins
CN102482277B (zh) * 2009-05-05 2017-09-19 达纳-法伯癌症研究所有限公司 表皮生长因子受体抑制剂及治疗障碍的方法
US8410126B2 (en) 2009-05-29 2013-04-02 Boehringer Ingelheim International Gmbh Pyrimidine inhibitors of PKTK2
US20120142667A1 (en) 2009-06-10 2012-06-07 Nigel Ramsden Pyrimidine derivatives as zap-70 inhibitors
WO2010146132A1 (fr) 2009-06-18 2010-12-23 Cellzome Limited Sulfonamides et sulfamides servant d'inhibiteurs de la zap-70
EP2467142B1 (fr) 2009-08-17 2016-09-21 Memorial Sloan-Kettering Cancer Center Dérivés 2-(pyrimidin-5-yl)-thiopyrimidine en tant que modulateurs Hsp70 and Hsc70 pour le traitement de maladies proliferatives
CA2773848A1 (fr) 2009-09-09 2011-03-17 Avila Therapeutics, Inc. Inhibiteurs de pi3 kinase et leurs utilisations
RU2012114902A (ru) 2009-09-16 2013-10-27 Авила Терапьютикс, Инк. Конъюгаты и ингибиторы протеинкиназы
JP2013505927A (ja) 2009-09-25 2013-02-21 バーテックス ファーマシューティカルズ インコーポレイテッド プロテインキナーゼ阻害剤として有用なピリミジン誘導体の調製方法
EP3091021B1 (fr) 2009-10-26 2019-08-28 Signal Pharmaceuticals, LLC Procédés de synthèse et de purification de composés hétéroaryles
US20110207736A1 (en) 2009-12-23 2011-08-25 Gatekeeper Pharmaceuticals, Inc. Compounds that modulate egfr activity and methods for treating or preventing conditions therewith
CA2785738A1 (fr) 2009-12-30 2011-07-07 Avila Therapeutics, Inc. Modification covalente de proteine, dirigee sur un ligand
CA2789711C (fr) 2010-02-17 2014-08-05 Amgen Inc. Derives d'arylcarboxamide en tant qu'inhibiteurs de canal sodique pour le traitement de la douleur
US8383793B2 (en) 2010-04-15 2013-02-26 St. Jude Children's Research Hospital Methods and compositions for the diagnosis and treatment of cancer resistant to anaplastic lymphoma kinase (ALK) kinase inhibitors
US20130137709A1 (en) 2010-05-05 2013-05-30 Nathanael S. Gray Compounds that modulate EGFR activity and methods for treating or preventing conditions therewith
EP2569302A1 (fr) 2010-05-13 2013-03-20 Amgen, Inc Composés d'hétéroaryloxyhétérocyclyle convenant comme inhibiteurs de la pde10
BR112012033253A2 (pt) * 2010-06-23 2016-11-22 Hanmi Science Co Ltd novos derivados de pirimidina fundidos para inibição da atividade de tirosina quinase
IT1401326B1 (it) 2010-07-29 2013-07-18 Parlux S P A Dispositivo silenziatore per asciugacapelli.
KR20130099040A (ko) 2010-08-10 2013-09-05 셀진 아빌로믹스 리서치, 인코포레이티드 Btk 억제제의 베실레이트 염
WO2012022045A1 (fr) 2010-08-20 2012-02-23 Hutchison Medipharma Limited Composés pyrrolopyrimidiniques et leurs utilisations
CA2810900A1 (fr) 2010-10-14 2012-04-19 Ariad Pharmaceuticals, Inc. Methodes d'inhibition de la proliferation cellulaire dans des cancers induits par l'egfr
JP5956999B2 (ja) 2010-11-01 2016-07-27 セルジーン アヴィロミクス リサーチ, インコーポレイテッド ヘテロアリール化合物およびその使用
BR112013010564B1 (pt) 2010-11-01 2021-09-21 Celgene Car Llc Compostos heterocíclicos e composições compreendendo os mesmos
ES2665013T3 (es) 2010-11-10 2018-04-24 Celgene Car Llc Inhibidores de EGFR selectivos de mutante y usos de los mismos
CN103501612B (zh) * 2011-05-04 2017-03-29 阿里亚德医药股份有限公司 抑制表皮生长因子受体导致的癌症中细胞增殖的化合物
CA2881993C (fr) 2011-07-27 2017-05-09 Astrazeneca Ab Composes de 4-methoxy-n3-(pyrimidine-2-yl)benzene-1,3-diamine substitue et ses sels.
WO2013042006A1 (fr) 2011-09-22 2013-03-28 Pfizer Inc. Dérivés de pyrrolopyrimidine et de purine
WO2013169401A1 (fr) 2012-05-05 2013-11-14 Ariad Pharmaceuticals, Inc. Composés pour inhiber la prolifération cellulaire dans les cancers induits par l'egfr
EP3143015B1 (fr) 2014-05-13 2019-02-20 ARIAD Pharmaceuticals, Inc. Composés hétéroaryl pour l'inhibition de kinase
SI3157916T1 (sl) 2014-06-19 2019-05-31 Ariad Pharmaceuticals, Inc. Heteroarilne spojine za zaviranja kinaze
TN2017000157A1 (en) 2014-10-21 2018-10-19 Ariad Pharma Inc Crystalline forms of 5-chloro-n4-[-2-(dimethylphosphoryl) phenyl]-n2-{2-methoxy-4-[4-(4-methylpiperazin-1-yl) piperidin-1-yl] pyrimidine-2,4-diamine
US20210323976A1 (en) 2015-05-13 2021-10-21 Ariad Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9834518B2 (en) 2011-05-04 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9834571B2 (en) 2012-05-05 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9611283B1 (en) 2013-04-10 2017-04-04 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in ALK-driven cancers
EP3392245A4 (fr) * 2015-12-15 2019-08-21 Precedo Pharmaceuticals Co., Ltd. Nouveau double inhibiteur de egfr et de alk
CN113135920A (zh) * 2016-06-30 2021-07-20 株式会社大熊制药 吡唑并嘧啶衍生物作为激酶抑制剂
US11034669B2 (en) 2018-11-30 2021-06-15 Nuvation Bio Inc. Pyrrole and pyrazole compounds and methods of use thereof
CN111718325A (zh) * 2019-03-22 2020-09-29 烟台药物研究所 一种2,4,5-取代嘧啶类化合物及其制备方法和应用

Also Published As

Publication number Publication date
AU2013204563A1 (en) 2013-11-21
JP2015518490A (ja) 2015-07-02
US9834571B2 (en) 2017-12-05
AU2013204563B2 (en) 2016-05-19
JP6469567B2 (ja) 2019-02-13
US20160244469A1 (en) 2016-08-25
WO2013169401A1 (fr) 2013-11-14

Similar Documents

Publication Publication Date Title
US9834571B2 (en) Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9834518B2 (en) Compounds for inhibiting cell proliferation in EGFR-driven cancers
AU2011315831B2 (en) Methods for inhibiting cell proliferation in EGFR-driven cancers
US10039759B2 (en) Quinolines as FGFR kinase modulators
TWI233437B (en) Substituted 3-cyanoquinolines
AU2005244104B2 (en) 4,5-disubstituted-2-aryl pyrimidines
TWI280960B (en) Quinazoline derivatives process for preparing the same and pharmaceutical compositions containing the same
WO2013030218A1 (fr) Inhibiteurs sélectifs et réversibles de la protéase 7 spécifique à l'ubiquitine
JP2013227358A (ja) チロシンキナーゼの阻害剤
JP2003528861A (ja) 4−アミノ―5−シアノ―2−アニリノ―ピリミジン誘導体及びその細胞周期キナーゼ阻害剤としての使用
JP2007520558A (ja) キナーゼ阻害剤として有用なピリミジノン化合物
EP2844642B1 (fr) Composés pour inhiber la prolifération cellulaire dans les cancers induits par l'egfr
JP2022544700A (ja) 筋萎縮性側索硬化症および関連障害の治療のためにtdp-43に結合する分子
TW378208B (en) Pharmacologically active N-phenyl-4-(4-pyridyl)-2-pyrimidineamine derivatives
AU2013203904A1 (en) Methods for inhibiting cell proliferation in EGFR-driven cancers
BR112017022243B1 (pt) Formas cristalinas do composto inibidor de fgfr4 cristalino, método para preparar uma forma cristalina e composição farmacêutica e usos terapêuticos das mesmas

Legal Events

Date Code Title Description
AS Assignment

Owner name: ARIAD PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DALGARNO, DAVID C.;HUANG, WEI-SHENG;SHAKESPEARE, WILLIAM C.;AND OTHERS;SIGNING DATES FROM 20150504 TO 20151027;REEL/FRAME:036891/0819

STCB Information on status: application discontinuation

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