WO2013091502A1 - Tri-heterocyclic derivatives, preparation process and uses thereof - Google Patents

Tri-heterocyclic derivatives, preparation process and uses thereof Download PDF

Info

Publication number
WO2013091502A1
WO2013091502A1 PCT/CN2012/086492 CN2012086492W WO2013091502A1 WO 2013091502 A1 WO2013091502 A1 WO 2013091502A1 CN 2012086492 W CN2012086492 W CN 2012086492W WO 2013091502 A1 WO2013091502 A1 WO 2013091502A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
unsubstitued
compound
alkyl
cycloalkyl
Prior art date
Application number
PCT/CN2012/086492
Other languages
French (fr)
Inventor
Daxin Gao
Heping Yang
Yajun YU
Original Assignee
Shanghai De Novo Pharmatech Co Ltd.
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 Shanghai De Novo Pharmatech Co Ltd. filed Critical Shanghai De Novo Pharmatech Co Ltd.
Priority to EP12858726.8A priority Critical patent/EP2763998B1/en
Priority to CA2855764A priority patent/CA2855764A1/en
Priority to AU2012357339A priority patent/AU2012357339B2/en
Priority to US14/356,982 priority patent/US9216997B2/en
Priority to JP2014547693A priority patent/JP6028306B2/en
Priority to KR1020147015840A priority patent/KR20140104427A/en
Publication of WO2013091502A1 publication Critical patent/WO2013091502A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/429Thiazoles condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains three hetero rings
    • C07D513/14Ortho-condensed systems

Definitions

  • the present invention belongs to pharmaceutical chemistry field, and relates to a compound as protein kinase inhibitor, preaparation process, composition containing the same and uses thereof; specifically relates to a new tri-heterocyclic derivatives, preaparation process, composition containing the same and uses thereof, particularly to their pharmaceutical use as inhibitors of kinase.
  • Protein kinases are a large family of proteins that play a pivotal role in the regulation of a wide variety of cellular processes, maintaining control over cellular function. Protein tyrosine kinases may be classified as growth factor receptor (e.g. VEGFR, EGFR, PDGFR, FGFR and erbB2) or non-receptor (e.g. c-src and bcr-abl) kinases. Receptor tyrosine kinases (RTKs) play a key role in the regulation of cell proliferation, differentiation, metabolism, migration, and survival. Upon ligand binding, they undergo tyrosine phosphorylation at specific residues in the cytoplasmic tail.
  • growth factor receptor e.g. VEGFR, EGFR, PDGFR, FGFR and erbB2
  • non-receptor e.g. c-src and bcr-abl
  • RTKs Receptor tyrosine kinases
  • Flt3 FMS-like receptor tyrosine kinase-3
  • AML acute myeloid leukemia
  • a large portion of DCs are derived from hematopoietic progenitors that express FLT3 receptor (CD135), and stimulation of the receptor via FLT3 ligand either in vivo or in vitro is known to drive expansion and differentiation of these progenitors toward a DC phenotype.
  • dendritic cells are the central antigen-presenting cells for initiation of T cell responses to mediate immune response including the autoreactive immune response. Inhibition of FLT3 signaling may thus produce an inhibition of DC-induced stimulation of T cells, thereby inhibiting autoimmune responses.
  • Flt3 inhibitor CEP-701 a drug already known to block actions of the growth-promoting FLT3 gene, to be effective in mice model engineered to mimic multiple sclerosis (MS model).
  • Multiple sclerosis is a disease that causes T-cells to destroy the myelin protein sheath around nerves in the central nervous system.
  • Flt3 inhibitor CEP-701 to be effective in reducing myelin loss in the MS mice model.
  • the study also shows that hemopoietic cytokines such as Flt3 ligand (a dendritic cell-mobilizing factor) and M-CSF are elevated significantly in the serum of patients with Langerhans cell histiocytosis and systemic lupus erythematosus. Higher level of cytokines correlated with patients having more extensive diseases.
  • kinase inhibitors for use in cancer therapy.
  • urea derivatives have been reported to be selective Flt3 inhibitors in Bioorg. Med. Chem. Lett. 10:2051-2054 (2000), WO 99/32106 published 1 Jul. 1999, PCT publication WO 99/32111 published 1 Jul. 1999 and PCT publication WO2007/109120 published in 27 Sept. 2007.
  • the present inventors have now discovered a new kind of tri-heterocyclic derivatives, preaparation process, composition containing the same and uses thereof, particularly to their pharmaceutical use as inhibitors of kinase.
  • the compounds exhibit a surprising increase in the level of inhibition of MV4-1 1 cell proliferation (MV4-11 cell is a human leukemia cell lines that express a FLT3-ITD mutation), and/or a surprising increase in the solubility of the compound (in aqueous media and/or phosphate buffer solution)-enhanced solubility may be of use in formulation the compounds, for example, for administration by an IV route, or for oral formulations (e.g. liquid and small tablet forms) for pediatric use.
  • the oral bioavailability of the compounds of the present invention may be enhanced.
  • the compounds may also be less susceptible to the action of MDR1 in cells.
  • the first aspect of the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof:
  • Ar is selected from the group consisting of optionally substituted or unsubstitued aryl, and optionally substituted or unsubstitued heteroaryl, when substituted, the substituents could be one or more groups independently selected from halogen, alkyl, haloalkyl, or hydroxyl alkyl;
  • L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyi, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyl, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyi, heterocycloalkyl,
  • Y is O, S, NR 2 R 2 ' or a direct bond (that means L directly connected with the left heteroaryl);
  • X-i , X 2 , X3 and X 4 are independently N or CR-i ,
  • R-i is H, or -Y-L;
  • R, R 2 and R 2 ' are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, haloalkoxyl, hydroxyl, amino, aminocarbonyl, sulfonamido, cyano, alkynyl, alkoxyl, aryloxyl, carboxylic acid, carboxylic ester or halogen, or R 2 , R 2 ' together with the nitrogen atom to which they are attached, form a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms.
  • the 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl.
  • R 2 Preferably R 2 ,
  • R 2 ' and the N could be formed a heterocycle such as , and R 7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-
  • R 7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-
  • One preferably subset of compound of the present invention provides formula (I) or a pharmaceutically acceptable salt thereof, wherein:
  • L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl, when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyi,
  • Y is O, S, NR 2 R 2 ' or a direct bond
  • X-i , X 2 , X 3 and X 4 are independently N or CR-i ,
  • R, R 2 and R 2 ' are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, haloalkoxyl, hydroxyl, amino, amido, aminocarbonyl, sulfonamido, cyano, alkynyl, alkoxyl, aryloxyl, carboxylic acid, carboxylic ester or halogen, or R 2 , R 2 ' together with the nitrogen atom to which they are attached, form a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms.
  • the 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl.
  • R 2 Preferably R 2 ,
  • R 2 ' and the N could be formed a heterocycle such as , and R 7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-
  • R 7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-
  • L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl, when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyi,
  • Y is O, S or NR 2 R 2 ';
  • X-i , X 2 , X3 and X 4 are independently N or CR-i ,
  • R-i is H, or -Y-L
  • R, R 2, and R 2 ' are each independently selected from the group consisting of hydrogen or alkyl, or R 2 , R 2 ' together with the nitrogen atom to which they are attached, formed a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms.
  • the 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl.
  • R 2 , R 2 ' and the N could be formed a heterocycle such as and R 7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-
  • R 7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-
  • L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyi,
  • Y is O
  • X-i , X 2 , X 3 and X 4 are independently N or CH;
  • R is hydrogen.
  • Another preferably subset of compound of the present invention provides formula (I) or a pharmaceutically acceptable salt thereof, wherein:
  • L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyi,
  • Y is NR 2 R 2 '
  • X X 2 , X3 and X 4 are independently N or CH;
  • R is hydrogen
  • R 2 , R 2 ' is hydrogen or alkyl, or R 2 , R 2 ' together with the nitrogen atom to which they are attached, formed a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms.
  • the 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl.
  • R 2 , R 2 ' and the N could be framed a heterocycle such as and R 7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-
  • R 7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-
  • L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyi,
  • Y is a direct bond or O, S or NR 2 R 2 ';
  • X X 2 , X 3 and X 4 are independently CR
  • R is H, or -Y-L; R is hydrogen;
  • R 2 , R 2 ' is hydrogen or alkyl, or R 2 , R 2 ' together with the nitrogen atom to which they are attached, formed a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms.
  • the 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl.
  • R 2 , R 2 'and the N could be formed a heterocycle such as and R 7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-
  • R 7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-
  • L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyi, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyi, heterocycloalkyi,
  • Y is a direct bond
  • X X 2 , X 3 and X 4 are independently N or CR
  • R is H, or -Y-L
  • R is independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, haloalkoxyl, hydroxyl, amino, aminocarbonyl, sulfonamido, cyano, alkynyl, alkoxyl, aryloxyl, carboxylic acid, carboxylic ester or halogen.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein the compounds are selected from, but not limited to the following compounds:
  • the second aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) as defined in the above mentioned, or a pharmaceutically acceptable salt thereof, as well as a pharmaceutically acceptable carrier, adjuvant, excipient, or diluent in a therapeutically effective dose.
  • the third aspect of the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in the above mentioned for the use in preparation of a medicament as Flt3 kinase inhibitor.
  • the fourth aspect of the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in the above mentioned for the use in preparation of a medicament by modulating FLT3-mediated diseases, especially said medicament comprising administering a therapeutically effective amount of a compound of formula (I) as defined in the above mentioned, or a pharmaceutically acceptable salt thereof, as well as an isomer, a solvate, a hydrate, or a prodrug.
  • the fifth aspect of the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in the above mentioned for the use in preparation of a medicament for treating a cellular proliferative disorderes diseases by inhibiting Flt3 kinase activities, wherein said diseases such as acute myeloid leukemia, chronic myeloid leukemia, myeloma, ovarian cancer, breast cancer, prostate cancer, pancreatic cancer, colorectal cancer, stomach cancer, non small cell lung cancer, thyroid cancer, brain cancer or lymphoma, said medicament comprising administering a therapeutically effective amount of a compound of formula (I) as defined in the above mentioned, or a pharmaceutically acceptable salt thereof.
  • diseases such as acute myeloid leukemia, chronic myeloid leukemia, myeloma, ovarian cancer, breast cancer, prostate cancer, pancreatic cancer, colorectal cancer, stomach cancer, non small cell lung cancer, thyroid cancer, brain cancer or lymphoma
  • said medicament comprising administering a therapeutic
  • the sixth aspect of the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in the above mentioned for the use in preparation of a medicament for treating inflammatory and autoimmune diseases by inhibiting Flt3 kinase activities, wherein said diseases such as asthma, lupus, systemic lupus, rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, or Crohn's disease, said medicament comprising administering a therapeutically effective amount of a compound of formula (I) as defined in the first aspect, or a pharmaceutically acceptable salt thereof.
  • alkyl refers to a saturated aliphatic hydrocarbon group including C1-C20 straight chain and branched chain groups.
  • an alkyl group is a moderate size alkyl having C1-C10 straight chain and branched chain groups, e.g., methyl, ethyl, propyl, 2-propyl, n-butyl, / ' so-butyl, ferf-butyl, pentyl, neopentyl, hexyl, isohexyl, heptyl, octyl, and the like.
  • it is a lower alkyl having C1-C6 straight chain and branched chain groups, e.g., methyl, ethyl, propyl, 2-propyl, n-butyl, / ' so-butyl, or ferf-butyl, pentyl, neopentyl, hexyl, isohexyl, and the like.
  • Said "optionally substituted alkyl” refers to an alkyl group which may be substituted at any available point of attachment and by one to 4 subtituents selected from the group consisting of halo, hydroxyl, lower alkoxy, aryl(optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), aryloxy(optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), heteroaryl(optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), heterocycloalkyl(optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), haloalkoxyl, amino, aminocarbonyl, cyano, alkynyl, carboxylic acid, or carboxylic ester.
  • cycloalkyl refers to a 3 to 8 membered all-carbon monocyclic ring, an all-carbon 5-membered/6-membered or 6-membered/6-membered fused bicyclic ring or a multicyclic fused ring (a "fused" ring system means that each ring in the system shares an adjacent pair of carbon atoms with other ring in the system) group wherein one or more rings may contain one or more double bonds, but none of the rings has a completely conjugated p/ ' -electron system.
  • cycloalkyl groups are cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, chcyclohexyl, cyclohexadienyl, adamantyl, cycloheptyl, cycloheptatrienyl, and the like.
  • the cycloalkyl group may be substituted or unsubstituted.
  • the substituent group(s) is preferably one or more independently selected from the group consisting of lower alkyl, trihaloalkyl, halo, hydroxy, hydroxylalkyl,aminoalkyl, amino, aryl (optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), aminocarbonyl, sulfonamido, ureido, aryloxy (optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), 6-membered heteroaryl (having 1 to 3 nitrogen atoms on the ring, the carbons on the ring being optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), 5-membered heteroaryl (having 1 to 3 heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, the carbon and nitrogen atoms of the group being optionally substituted
  • alkenyl refers to an alkyl group as defined above having at least 2 carbon atoms and at least one carbon-carbon double bond. Representative examples include, but are not limited to ethenyl,
  • the optionally substituted alkenyl means the the alkenyl which may be substituted with one or more groups which each independently is halo, cyano, lower alkyl or lower alkoxy groups.
  • alkynyl refers to an alkyl group as defined above having at least 2 carbon atoms and at least one carbon-carbon triple bond. Representative examples include, but are not limited to ethynyl, 1-propynyl,
  • the optionally substituted alkenyl means the alkynyl which may be substituted with one or more groups which each independently is halo, cyano, lower alkyl or lower alkoxy groups.
  • aryl (Ar) refers to groups having at least one aromatic ring, i.e., having a conjugated p/ ' -electron system, including all-carbon cyclic aryl, and biaryl group.
  • aryl include phenyl, nathathyl, and the like.
  • Said aryl group may be optionally substituted with one or more groups each independently selected from the group consisting of halo, trihalomethyl, hydroxy, hydroxylalkyl, mercapto, alkylthio, nitro, cyano, alkoxyl and alkyl.
  • Het refers to an aryl having 1 to 3 heteroatoms selected from the group consisting of N, O, and S as ring atoms, the remaining ring atoms being C.
  • Said ring is 5- or 6- membered ring.
  • heteroaryl groups include furyl, thienyl, pyridinyl, pyrrolyl, N-alkyl pyrrolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazole, pyrimidinyl, pyrazinyl, imidazolyl, triazolyl, tetrazolyl, oxatriazolyl, pyridazinyl, triazinyl, oxadiazolyl, thiadiazolyl, furazanyl, and the like.
  • Said heteroaryl group may be optionally substituted with (via a carbon ot hetero atom) one or more groups each independently selected from the group consisting of halo, , haloalkyl, hydroxy, hydroxylalkyl, mercapto, alkylthio, nitro, cyano, alkoxyl and alkyl.
  • heterocycloalkyl refers to a mono-heterocycloalkyl, bi-heterocycloalkyl, bridged-hetero ycloalkyl or spiro-heterocycloalkyl of 4 to 12 ring atoms, wherein one, or two ring heteroatoms are selected from the group consisting of N, O, and S (O)n (n is integer from 0 to 2), the remaining ring atoms are C, in addition, the ring may also have one or more double bonds, but not have a completely conjugated p/ ' -electron system.
  • Said mono heterocycloalkyl includes, but is not limited to azetidyl, pyrrolidyl, piperidyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, and the like.
  • bi-heterocycloalkyl examples include but not limited to
  • bridged- heterocycloalkyl included but not limited to
  • spiro-heterocycloalkyl examples include but not limited to
  • heterocycloalkyi may be substituted or unsubstituted.
  • the substituent group is preferably one or more, more preferably one, two, or three, further more preferably one or two groups, each independently selected from the group consisting of lower alkyi, lower hydroxylalkyl, trihaloalkyl, halo, hydroxy, haloalkyl, mercapto, nitro, lower alkoxyl, cyano, amino, aminoalkyl, alkoxyalkyl, cycloalkyi, heterocycloalkyi, methylsulfonyl, and acyl.
  • cycloalkylalkyl refers to a radical of the formula - RaRb, where Ra is an alkyi radical as defined above and Rb is a cycloalkyi radical as defined above.
  • the alkyi radical and the cycloalkyi radical may be optionally substituted as defined above.
  • arylalkyl refers to a radical of the formula - RaRc where Ra is an alkyi radical as defined above and Rc is aryl radicals as defined above, e.g., benzyl, diphenylmethyl and the like.
  • the alkyi radical and the aryl radical(s) may be optionally substituted as described above.
  • Heterocycloalkylalkyl refers to a radical of the formula - RaRd where Ra is an alkyi radical as defined above and Rd is a heterocycloalkyi radical as defined above, and if the heterocycloalkyi is a nitrogen-containing heterocycloalkyi, the heterocycloalkyi may be attached to the alkyi radical at the nitrogen atom or at carbon atom.
  • the alkyi part of the heterocycloalkyi radical may be optionally substituted as defined above for an alkyi group.
  • the heterocycloalkyi part of the heterocycloalkyi radical may be optionally substituted as defined above for a heterocycloalkyi group.
  • Heteroarylalkyi refers to a radical of the formula -RaRe where Ra is an alkyi radical as defined above and Re is a heteroaryl radical as defined above.
  • the heteroaryl part of the heteroarylalkyi radical may be optionally substituted as defined above for a heteroaryl group.
  • the alkyi part of the heteroarylalkyi radical may be optionally substituted as defined above for an alkyi group.
  • Said "hydroxy” refers to an -OH group.
  • alkoxyl refers to both an -O-(alkyl) and an -0-(unsubstituted cycloalkyi) group.
  • Representative examples include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like.
  • amino refers to a -NH 2, -NHCH 3 , -NHCH 2 CH 3 , -NH-n-propyl, -NH-isopropyl, -N(CH 3 ) 2 , -NHcyclopropyl, -NHPh, -NHpyridyl, pyrrolidinyl, piperazinyl, morpholino, piperidino, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkylalkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHheteroarylalkyl, etc.
  • Said "-NHalkylhydroxyl" refers to a-NH(alkyl)hydroxyl, wherein alkyi is as defined above.
  • Representative examples include, but are not limited to, e.g. -NHCH 2 CH 2 OH, -NHCH 2 CH(OH)CH 2 OH, etc.
  • Said "-NHalkoxylalkyl” refers to a-NH(alkyl)-0-(alkyl), wherein alkyi is as defined above.
  • -NHalkylamnio refers to -NH(alkyl)amnio, wherein alkyi and amnio is as defined above.
  • Representative examples include, but are not limited to, e.g. -NHCH 2 CHCNH 2 , -NHCH 2 CH 2 NHCH 3 ,
  • Said "-NHcycloalkyl” refers to NH-cycloalkyl, wherein cycloalkyd is as defined above.
  • -NHcycloalkylalkyl refers to a -NH-alkyl-cycloalkyl, wherein alkyi and cycloalkyi are as defined above.
  • Representative examples include, but are not limited to, e.g. NH-CH 2 -cyclopropyl-NH 2 , NH-CH 2 -
  • -NHheterocycloalkyl refers to NH-heterocycloalkyl, wherein heterocycloalkyl is as defined above.
  • Said "-NHheterocycloalkylalkyl” refers to NH-heterocycloalkyl-alkyl, wherein heterocycloalkyl and alkyl is as defined above.
  • -NHaryl refers to NH-aryl, wherein aryl is as defined above.
  • -NHarylalkyl refers to a -NH-alkyl-aryl, wherein aryl and alkyl are as defined above.
  • Representative examples include, but are not limited to, e.g. and the like.
  • -NH heteroarylalkyl refers to a -NH-alkyl-heteroaryl, wherein heteroaryl and alkyl are as defined above.
  • Representative examples include, but are not limited to, e.g. and the like.
  • haloalkoxy refers to an -O-(haloalkyl). Representative examples include, but are not limited to, trifluoromethoxy, trichloromethoxy, tribromomethoxy, and the like.
  • aryloxyl refers to both an -O-aryl and an -O-heteroaryl group, wherein the aryl and heteroaryl are as defined above.
  • Representative examples include, but are not limited to, phenoxy, pyridinyloxy, furanyloxy, thienyloxy, pyrimidinyloxy, pyrazinyloxy, and the like, and derivatives thereof.
  • Said "mercapto” refers to a -SH group.
  • alkylthio refers to a -S-(alkyl) and a -S-(unsubstituted cycloalkyl) group.
  • Representative examples include, but are not limited to, methylthio, ethylthio, propylthio, butylthio, cyclopropylthio, cyclobutylthio, cyclopentylthio, cyclohexylthio, and the like.
  • arylthio refers to a -S-aryl and a -S-heteroaryl group, wherein the aryl and heteroaryl are as defined above.
  • Representative examples include, but are not limited to, e.g., phenylthio, pyridinylthio, furanylthio, thienylthio, pyrimidinylthio, and the like, and derivatives thereof.
  • acyl refers to a -C(0)-R 3 " group, where R 3 is selected from the group consisting of hydrogen, lower alkyl, trihalomethyl, unsubstituted cycloalkyl, aryl (optionally substituted with one or more, preferably one, two, or three substituents selected from the group consisting of lower alkyl, trihalomethyl, lower alkoxy and halo groups), heteroaryl (bonded through a ring carbon) (optionally substituted with one or more, preferably one, two, or three substitutents selected from the group consisting of lower alkyl, trihaloalkyl, lower alkoxy and halo groups), and heteroalicyclic (bonded through a ring carbon) (optionally substituted with one or more, preferably one, two, or three substituents selected from the group consisting of lower alkyl, trihaloalkyl, lower alkoxy and halo groups).
  • Representative acyl groups include, but are not limited to, acetyl, tri
  • Said “thioacyl” refers to a -C(S)-R 3 group, wherein R 3 is as defined above.
  • halo refers to fluoro, chloro, bromo, or iodo, preferably fluoro or chloro.
  • haloalkyl refers to a (CH 3 ) 2 CH 2 FC-, or a CH 3 (CH 2 F) 2 C-, or a (CH 2 F) 3 C- group.
  • Said "cyano” refers to a -C ⁇ N group.
  • trihaloalkyl, or trihalomethyl refers to a CF 3 or the like, wherein the halo is define as above.
  • carboxylic acid refers to a -COOH group.
  • Said “carboxylic ester” refers to a -COOR 3 group, wherein R 3 is alkyl or cycloalkyl.
  • hydroxyalkyl refers to a -alkyl-OH group, wherein alkyl and hydroxyl as defined above.
  • the Representative hydroxyl alkyl group include but are not limited to, -CH 2 OH, -CH 2 CH 2 OH, CH 3 CH(OH)CH 2 -, (CH 3 ) 2 (CH 2 OH)C-, CH 3 (CH 2 OH) 2 C-, or (CH 2 OH) 3 C-, and the like.
  • aminoalkyl refers to a -alkyl-amnio group, wherein alkyl and amnio are as defined above.
  • the Representative amnioalkyl group include but are not limited to -CH 2 NH 2 , -CH 2 NHCH 3 , -CH 2 N(CH 3 )2,
  • R 6 refers to a sulfonamino substituents, for example, C lower alkyl, C 3 . 8 cycloalkyl, C 3 . 20 heterocycloalkyl, C 5 .
  • aryl and C 5 .
  • heteroaryl wherein alkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl are as defined above.
  • ureido refers to -NR 3 C(0)NR 4 R 5 where R 3 is hydrogen or alkyl; R 4 and R 5 are defined as above.
  • methylsulfonyl refers to CH 3 S0 2 -.
  • prodrug refers to a compound which, when metabolized in vivo, converts back to the original active compound.
  • the prodrug is inactive, or less active than the active compound, but may provide advantageous handling, administration, or metabolic properties.
  • heterocyclyl optionally substituted with an alkyl group means that the alkyl may or may not be present, and the description includes situations where the heterocyclic group is substituted with an alkyl group and situations where the heterocyclic group is not substituted with the alkyl group.
  • Said "pharmaceutical composition” refers to a mixture of one or more of the compounds of formula (I) as described in present invention herein, or hydrates, or solvates, or isomers, or physiologically/pharmaceutically acceptable salts or prodrugs thereof, with other chemical components, such as physiologically/pharmaceutically acceptable carriers and recipients.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound to warm blood animals and human.
  • Said “examplary compound” refers to the compound which given from the present invention, such as: the examplary compound 8-1 is illustrated as example 1 , the examplary compound 8-2 is illustrated as example 2, and etc.
  • ureas, or cyclic forms thereof, compounds of the present invention may be synthesized by methods well known to those of skill in the art or using methods known in the art in combination with methods described herein.
  • Chloronitropyridine 20 was used as starting material, after methoxylation, reduced nitro 21 into amine 22 by iron powder, then was converted into amniothiazole 23, demethylation to obtain phenol 24, condensed with compound 4 to obtain tricyclic nitro compound 25, after reacted with chloride 16, reduced nitro into amine 27 by iron powder, the final target urea compound was obtained by reacting 27 with isocyanate 7 as showed in scheme 3-A.
  • Nitroprimidinechloride compound 1 used as a starting material after substituted by 2,2-dimethoxyethanamine 91 , then reduced the nitro 91 by iron powder to obtain amine compound 92, the compound 92 was then condensed with compound 4, followed by the reduction of nitro group with zinc powder into amine 94, coupled with isocyanate 7 to form urea intermediate 95, and acid hydrolysis acetal into aldehyde 96 which was finally formed different 97 series compounds by reduction amnination with different amines, as showed in scheme 4-A.
  • the ureas may be converted to thioureas by treating with Lawesson's reagent in the presence of toluene.
  • protecting group Cbz refers to chemical moieties that block some or all reactive moieties and prevent such groups from participating in chemical reactions until the protective group is removed. It is preferred that each protective group be removable by a different means. Protective groups that are cleaved under totally disparate reaction conditions fulfill the requirement of differential removal. Protective groups can be removed by acid, base, and hydroge no lysis. Groups such as trityl, dimethoxytrityl, acetal and t-butyldimethylsilyl are acid labile and may be used to protect carboxy and hydroxy reactive moieties in the presence of amino groups protected with Cbz groups, which are removable by hydrogenolysis.
  • the present invention also relates to a compounds of formula (I) or a pharmaceutically acceptable salt thereof, wherein a compound of formula (I) may be present in the form of free base or in the form of salts formed by acid addition which are pharmaceutically acceptable non-toxic.
  • the pharmaceutically acceptable salts include hydrochloride, p-toluenesulfonate, benzenesulfonate, naphthalenesulfonate, tartarate, maleate, lactate, methanesulfonate, ethanesulfonate, sulfate, phosphate, citrate, acetate and trifluoroacetate, preferably p-toluenesulfonate, benzenesulfonate, methanesulfonate, hydrochloride, tartarate and trifluoroacetate.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof in an effective therapeutic dose, and a pharmaceutically acceptable carrier, and a use of the compounds of the present disclosure or pharmaceutical acceptable salts in the preparation of a medicament as a kinase inhibitor.
  • this disclosure also provides a pharmaceutical composition comprising the above mentioned compounds in an effective therapeutic dose, as well as their use in the preparation of a medicament as Flt3 inhibitor.
  • compositions may be prepared from the active ingredients in combination with pharmaceutically acceptable carriers.
  • the pharmaceutically acceptable salts of the compounds of formula (I) include conventional non-toxic salts or quarternary ammonium salts of the compounds of formula (I) formed e.g. from non- toxic inorganic or organic acids.
  • non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glyeolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, sulfanilic, 2-acetoxybenzoic, fumaric, naphthalenesulfonic, toluenesulfonic, benzenesulfonic, ethanesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like
  • the pharmaceutically acceptable salts of the present invention can be synthesized by conventional chemical methods.
  • the salts are prepared by reacting the free base or acid with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid or base, in a suitable solvent or solvent combination.
  • the compounds of the present invention may have asymmetric centers and occur as racemates, racemic mixtures, and as individual diastereomer. All such isomers including optical, enantiomeric, diasteriomeric, stereoisomeric, epimeric, and geometric isomers are included in the present invention.
  • the compounds of the present invention may be in crystalline or non- crystalline form, it may exist in a number of different polymorphic forms, and may optionally be hydrated or solvated. This invention includes within its scope stoichiometric hydrates as well as compounds containing variable amount of water.
  • the present invention described herein also includes a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of compound of formula (I) or a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable carrier in the use of preparation of a medicament as kinase inhibitors.
  • FLT3 activity includes, but is not limited to, enhanced FLT3 activity resulting from increased or de novo expression of FLT3 in cells, increased FLT3 expression or activity, and FLT3 mutations resulting in constitutive activation.
  • the existence of inappropriate or abnormal FLT3 ligand and FLT3 levels or activity can be determined using well known methods in the art. For example, abnormally high FLT3 levels can be determined using commercially available ELISA kits. FLT3 levels can be determined using flow cytometric analysis, immunohistochemical analysis, and in situ hybridization techniques.
  • An inappropriate activation of the FLT3 can be determined by an increase in one or more of the activities occurring subsequent to FLT3 binding: (1 ) phosphorylation or autophosphorylation of FLT3; (2) phosphorylation of a FLT3 substrate, e.g., Stat5, Ras; (3) activation of an related complex, e.g., PI3K; (4) activation of an adaptor molecule; and (5) cellular proliferation. These activities are readily measured by well known methods in the art.
  • a compound of formula (I) disclosed herein would be useful for, but not limited to, the prevention or treatment of proliferative diseases, conditions, or disorders in a patient by administering to the patient a compound of formula (I) or a composition comparising a compound of formula (I) disclosed herein in an effective amount.
  • diseases, conditions, or disorders include cancer, particularly hematopoietic cancer, metastatic cancer, atherosclerosis, and lung fibrosis.
  • neoplasia including cancer and metastasis, including, but not limited to: carcinoma such as cancer of the bladder, breast, colon, kidney, liver, lung (including small cell lung cancer), esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage (including leukemia, acute lymphocitic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma); hematopoietic tumors of myeloid lineage (including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia); tumors of me
  • tumors of the central and peripheral nervous system including astrocytoma, neuroblastoma, glioma and schwannomas); and other tumors (including melanoma, seminoma, teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma, thyroid follicular cancer and Kaposi's sarcoma).
  • the compounds also would be useful for treatment FLT3-mediated and/or CSF-1 R mediated diseases like immune dysfunction, autoimmune diseases, kidney diseases, tissue transplant rejection, lupus erythematosis, multiple sclerosis, inflammatory bowel disease, rheumatoid arthritis, arthritis, asthma.
  • FLT3-mediated and/or CSF-1 R mediated diseases like immune dysfunction, autoimmune diseases, kidney diseases, tissue transplant rejection, lupus erythematosis, multiple sclerosis, inflammatory bowel disease, rheumatoid arthritis, arthritis, asthma.
  • the compounds disclosed herein are also useful in the treatment of diabetic conditions such as diabetic retinopathy and microangiopathy.
  • the compounds disclosed herein are also useful in the reduction of blood flow in a tumor in a subject.
  • the compounds disclosed herein are also useful in the reduction of metastasis of a tumor in a subject.
  • these compounds are also useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like.
  • animals include horses, dogs, and cats.
  • the compounds of formula(l) disclosed herein include the pharmaceutically acceptable derivatives thereof.
  • the treatment method that includes administering a compound or composition disclosed herein can further include administering to the patient an additional therapeutic agent (combination therapy) selected from: a chemotherapeutic or anti-proliferative agent, or an anti-inflammatory agent, wherein the additional therapeutic agent is appropriate for the disease being treated and the additional therapeutic agent is administered together with a compound or composition disclosed herein as a single dosage form or separately from the compound or composition as part of a multiple dosage form.
  • the additional therapeutic agent may be administered at the same time as a compound disclosed herein or at a different time. In the latter case, administration may be staggered by, for example, 6 hours, 12 hours, 1 day, 2 days, 3 days, 1 week, 2 weeks, 3 weeks, 1 month, or 2 months.
  • the invention also features a method of inhibiting the growth of a cell that expresses VEGFR, or c-Met, that includes contacting the cell with a compound or composition disclosed herein, thereby causing inhibition of growth of the cell.
  • a cell whose growth can be inhibited include: a breast cancer cell, a colorectal cancer cell, a lung cancer cell, a papillary carcinoma cell, a prostate cancer cell, a lymphoma cell, a colon cancer cell, a pancreatic cancer cell, an ovarian cancer cell, a cervical cancer cell, a central nervous system cancer cell, an osteogenic sarcoma cell, a renal carcinoma cell, a hepatocellular carcinoma cell, a bladder cancer cell, a gastric carcinoma cell, a head and neck squamous carcinoma cell, a melanoma cell, or a leukemia cell.
  • the invention also provided herein a method of inhibiting VEGFR, and/or c-Met kinase activity in a biological sample that includes contacting the biological sample with a compound or composition disclosed herein.
  • biological sample means a sample outside a living organism and includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • Inhibition of kinase activity, particularly VEGFR or c-Met kinase activity, in a biological sample is useful for a variety of purposes known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ-transplantation, biological specimen storage, and biological assays.
  • an "effective amount” or “effective dose” of the compound or pharmaceutically acceptable composition is that amount effective for treating or lessening the severity of one or more of the aforementioned disorders.
  • the compounds and compositions, according to the method disclosed herein, may be administered using any amount and any route of administration effective for treating or lessening the severity of the disorder or disease. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like.
  • a compound or composition can also be administered with one or more other therapeutic agents, as discussed above.
  • the compounds disclosed herein or pharmaceutical compositions thereof may also be used for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters.
  • vascular stents for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury).
  • patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a compound disclosed herein.
  • the dosage used can be varied depending upon the type of cancer, the age and general condition of the patient, the particular compound administered, the presence or level of toxicity or adverse effects experienced with the drug, and other factors.
  • a representative example of a suitable dosage range is from as low as about 0.01 mg/kg to as high as about 100 mg/kg. However, the dosage administered is generally left to the discretion of the physician.
  • the methods of treatment are preferably carried out by delivering a compound of formula (I) orally or parenterally.
  • parenteral' as used herein includes intravenous, intramuscular, or intraperitoneal administration.
  • the subcutaneous and intramuscular forms of parenteral administration are generally preferred.
  • the instant invention can also be carried out by delivering the compound of formula I subcutaneously, intranasally, intrarectally, transdermal ⁇ or intravaginally.
  • the compounds of formula (I) may also be administered by inhalation.
  • inhalation' is meant intranasal and oral inhalation administration.
  • Appropriate dosage forms for such administration such as an aerosol formulation or a metered dose inhaler, may be prepared by convention techniques.
  • the invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) and a pharmaceutically acceptable carrier.
  • the compounds of formula (I) may also be included in pharmaceutical compositions in combination with a second therapeutically active compound.
  • the pharmaceutical carrier employed may be, for example, either a solid, liquid or gas.
  • solid carriers include lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid and the like.
  • liquid carriers are syrup, peanut oil, olive oil, water and the like.
  • gaseous carriers include carbon dioxide and nitrogen.
  • the carrier or diluent may include time delay material well known in the art, such as glyceryl monostearate or glycervl distearate, alone or with a wax.
  • a wide variety of pharmaceutical dosage forms can be employed. If a solid dosage is used for oral administration, the preparation can be in the form of a tablet, hard gelatin capsule, troche or lozenge. The amount of solid carrier will vary widely, but generally will be from about 0.025 mg to about 1 g. When a liquid dosage form is desired for oral administration, the preparation is typically in the form of a syrup, emulsion, soft gelatin capsule, suspension or solution. When a parenteral dosage form is to be employed, the drug may be in solid or liquid form, and may be formulated for administration directly or may be suitable for reconstitution. Topical dosage forms are also included. Examples of topical dosage forms are solids, liquids and semi-solids. Solids would include dusting powders, poultices and the like.
  • Liquids include solutions, suspensions and emulsions.
  • Semi-solids include creams, ointments, gels and the like.
  • the amount of a compound of formula (I) used topically will, of course, vary with the compound chosen, the nature and severity of the condition, and can be varied in accordance with the discretion of the physician.
  • a representative, topical, dose of a compound of formula I is from as low as about 0.01 mg to as high as about 2.0 g, administered one to four, preferably one to two times daily.
  • he active ingredient may comprise, for topical administration, from about 0.001 % to about 10% w/w.
  • Drops according to the present invention may comprise sterile or non-sterile aqueous or oil solutions or suspensions, and may be prepared by dissolving the active ingredient in a suitable aqueous solution, optionally including a bactericidal and/or fungicidal agent and/or any other suitable preservative, and optionally including a surface active agent.
  • the resulting solution may then be clarified by filtration, transferred to a suitable container which is then sealed and sterilized by autoclaving or maintaining at 98- 100°C for half an hour.
  • the solution may be sterilized by filtration and transferred to the container aseptically.
  • bactericidal and fungicidal agents suitable for inclusion in the drops are phenylmercuric nitrate or acetate (0.002%), benzalkonium chloride (0.01 %) and chlorhexidine acetate (0.01 %).
  • Suitable solvents for the preparation of an oily solution include glycerol, diluted alcohol and propylene glycol.
  • Lotions according to the present invention include those suitable for application to the skin or eye.
  • An eye lotion may comprise a sterile aqueous solution optionally containing a bactericide and may be prepared by methods similar to those for the preparation of drops.
  • Lotions or liniments for application to the skin may also include an agent to hasten drying and to cool the skin, such as an alcohol or acetone, and/or a moisturizer such as glycerol or an oil such as castor oil or arachis oil.
  • Creams, ointments or pastes according to the present invention are semi-solid formulations of the active ingredient for external application. They may be made by mixing the active ingredient in finely- divided or powdered form, alone or in solution or suspension in an aqueous or non-aqueous liquid, with a greasy or non-greasy base.
  • the base may comprise hydrocarbons such as hard, soft or liquid paraffin, glycerol, beeswax, a metallic soap; a mucilage; an oil of natural origin such as almond, com, arachis, castor or olive oil; wool fat or its derivatives, or a fatty acid such as stearic or oleic acid together with an alcohol such as propylene glycol or macrogels.
  • the formulation may incorporate any suitable surface active agent such as an anionic, cationic or non-ionic surfactant such as sorbitan esters or polyoxyethylene derivatives thereof.
  • suitable surface active agent such as an anionic, cationic or non-ionic surfactant such as sorbitan esters or polyoxyethylene derivatives thereof.
  • Suspending agents such as natural gums, cellulose derivatives or inorganic materials such as silicas, and other ingredients such as lanolin may also be included.
  • the coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof.
  • the coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysacca rides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics into the composition.
  • Implantable devices coated with a compound disclosed herein are another embodiment disclosed herein.
  • the compounds may also be coated on implantable medical devices, such as beads, or co-formulated with a polymer or other molecule, to provide a "drug depot" thus permitting the drug to be released over a longer time period than administration of an aqueous solution of the drug.
  • FIG.1 is demonstrated the antitumor effect on MV4-11 acute myeloid leukemia zenograft tumor model of the examplary compound of the present invention.
  • A solvent
  • B Quizartinib
  • C the examplary compound of the present invention
  • H NMR nuclear magnetic resonance
  • MS mass spectrometry
  • H NMR chemical shifts ( ⁇ ) were recorded in ppm (10 ⁇ 6 ).
  • NMR was performed on a Bruker AVANCE-400 spectrometer.
  • the suitable solvents were deuterated-chloroform (CDCI 3 ), deuterated-dimethyl sulfoxide (DMSO-cf 6 ) and deuterated-methanol (CD 3 OD) with tetramethylsilane (TMS) as an internal standard and chemical shifts were recorded in ppm (10 ⁇ 6 ).
  • Solvent A (10mM Ammonium hydrogen carbonate in water)
  • the average of inhibitory rate of Flt3 kinase and IC 50 was determined by a Caliper's mobility shift assay (MSA).
  • Thin-layer silica-gel was Yantai Huanghai HSGF254 or Qingdao GF254 silica-gel plate. Column chromatography generally used Yantai Huanghai 200-300 mesh silica gel as carrier.
  • CDCI 3 deuterated-chloroform
  • CD 3 OD deuterated-methanol
  • DIPEA diisopropylethylamine
  • TEA triethylamine
  • HOAc acetic acid
  • KSCN potassium thiocyanate
  • Fe iron powder
  • K 2 C0 3 potassium carbonate
  • DPPA Diphenylphosphonic azide
  • Example 26 ( Compound 35-4 was made by the method in scheme-3-B, through using (S)-(-)-1-Boc-2- pyrrolidinemethanol to replace intermediate 21 , as a light pink solid )
  • Compound 63-2 was prepared in the same method as compound 63-1 in scheme-3-F by using the intermediate 69 instead of intermediate 56. it is a light yellow solid.
  • MV4-11 cell ATCC, CRL-9591
  • MV4-11 cells a human leukemia cell line expressing constitutive active FLT3 receptor and contains internal tandem duplications (ITD) found in the AML patients.
  • MV4-11 cells were plated in 96 well plates at 15,000 cells per well in 100 ul IMDM medium (Invitrogen, 12440-053) containing 10% fetal bovine serum.
  • Test compounds were prepared in 100% DMSO and added to the cells to achieve final concentrations from 0.2 uM to 0.000001 uM (10 concentration points in 3 fold serial dilution). The culture plates were then incubated at 37C in 5% C0 2 for 72 hours.
  • Cell viability was then quantified at room temp using a CellTiter-Glo assay (Promega G7571 ) following the manufacture's instruction. Briefly, a volume of 10Oul Celltiter-Glo reagenet was added to each well to induce cell lysis. After 10 min incubation at RT, a luminescent signal was produced by measuring the amount of ATP. The signal was directly proportional to the number of viable cells present in culture.
  • FLT3 is a receptor tyrosine kinase involved in survival and proliferation of leukemic cells. Constitutively activating FLT3 mutations has been found in about 30% of all patients with acute myeloid leukemia (AML).
  • AML acute myeloid leukemia
  • the tested compounds were screened for their ability to inhibit FLT3 kinase activity using Caliper's mobility shift assay (MSA).
  • MSA Caliper's mobility shift assay
  • the assay uses a microfluidic chip to measure the conversion of a fluorescent peptide substrate to a phosphorylated product following separation by electrophoresis. The signature of the fluorescence signal over time reveals the extent of the reaction.
  • Each Compound was added into the reaction to final concentrations from 300 nM to 0.015 nM (10 concentration points in 3 fold serial dilution).
  • IC 50 values of the compounds of the present invention in inhibiting the FLT3 kinase are listed in following table 2.
  • Table 2 showed that most of the compounds of the present invention have the better FLT3 kinase inhibitory activities than Quizartinib.
  • the present invention compound(C) (3mg/kg, po, qd) was evaluated in MV4-11 acute myeloid leukemia zenograft tumor model, Quizartinib (B) (3mg/kg, po, qd)) used as a positive control.
  • NOD/SCID female mice 18-25 g were used. Each mouse was inoculated subcutaneously at the right flank with MV4-11 tumor cells (1 x 10 7 ) in 0.1 ml of PBS/Matrigel (1 :1 ) for tumor development. The treatments were started when the mean tumor size reached 188 mm 3 . The animals were separated even as test and control group randomly, and each group is consisted of 6 tumor-bearing mice. The test articles were orally administered (2.14 ml) to the tumor-bearing mice once a day. The date of tumor cell inoculation is denoted as day 0. The tumor volume was measured every four days, and the animals were sacrified at 22 days. Statistical analyses of difference in tumor volume among the groups were evaluated using a one-way ANOVA followed by individual comparisons using Games-Howell (equal variance not assumed).
  • FIG. 1 showed the antitumor effect in MV4-11 acute myeloid leukemia zenograft tumor model assay of the examplary compound of the present invention, where the mean tumor volume of the mice treated with either vehicle (A), positive control compound Quizartinib (B), or the examplary compound (C) of the present inventionis measured and plotted versus the dosing days.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Pulmonology (AREA)
  • Epidemiology (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Dermatology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

The present invention relates to a tri-heterocyclic derivatives, preparation process and uses thereof, specifically relates to a tri-heterocyclic derivatives of the formula (I) or a pharmaceutically acceptable salt thereof, preparation process, and further relates to a pharmaceutically acceptable composition comprising compounds of formula (I), or a pharmaceutically acceptable salt thereof, and their pharmaceutical use as inhibitors of kinase.

Description

DESCRIPTION
TRI-HETEROCYCLIC DERIVATIVES, PREPARATION PROCESS AND USES THEREOF FIELD OF THE INVENTION
The present invention belongs to pharmaceutical chemistry field, and relates to a compound as protein kinase inhibitor, preaparation process, composition containing the same and uses thereof; specifically relates to a new tri-heterocyclic derivatives, preaparation process, composition containing the same and uses thereof, particularly to their pharmaceutical use as inhibitors of kinase.
BACKGROUND OF THE INVENTION
Protein kinases are a large family of proteins that play a pivotal role in the regulation of a wide variety of cellular processes, maintaining control over cellular function. Protein tyrosine kinases may be classified as growth factor receptor (e.g. VEGFR, EGFR, PDGFR, FGFR and erbB2) or non-receptor (e.g. c-src and bcr-abl) kinases. Receptor tyrosine kinases (RTKs) play a key role in the regulation of cell proliferation, differentiation, metabolism, migration, and survival. Upon ligand binding, they undergo tyrosine phosphorylation at specific residues in the cytoplasmic tail. This leads to the binding of protein substrates and/or the establishment docking sites for adaptor proteins involved in RTK-mediated signal transduction. When unregulated, receptor tyrosine kinases can contribute to the rise of disease states associated with such aberrant kinase activity. Flt3 (FMS-like receptor tyrosine kinase-3), a member of calss III tyrosine kinase receptor family, is predominantly expressed in hematopoietic progenitor cells and plays an important role in the pathogenesis of acute myeloid leukemia (AML). Flt3 is expressed in blast cells of most patients with AML including wild-type and two forms of Flt3 mutations. These two mutations identified in the AML patients are internal tandem duplication (ITD) mutations in the juxtamembrane domain and point mutations (TKD) in the activation loop of the TKD. (See, Ryan J. Mattison et al Reviews in Reviews on Recent Clinical Trials, 2007, 2, 135-141 ). The relapse rates in the FU3/ITD mutation AML patients are significantly increased and the overall survival rates decreased compared with the AML patients without the Flt3 mutation. So development of a drug of inhibiting FH3/ITD mutant kinase could provide an effective way to treat AML. Currently more than a dozen known Flt3 inhibitors are being developed and some have shown promising clinical effects against AML (See Levis et al. Int J Hematol, 2005, 82: 100-107).
A large portion of DCs are derived from hematopoietic progenitors that express FLT3 receptor (CD135), and stimulation of the receptor via FLT3 ligand either in vivo or in vitro is known to drive expansion and differentiation of these progenitors toward a DC phenotype. Since dendritic cells are the central antigen-presenting cells for initiation of T cell responses to mediate immune response including the autoreactive immune response. Inhibition of FLT3 signaling may thus produce an inhibition of DC-induced stimulation of T cells, thereby inhibiting autoimmune responses. One study shows the Flt3 inhibitor CEP-701 , a drug already known to block actions of the growth-promoting FLT3 gene, to be effective in mice model engineered to mimic multiple sclerosis (MS model). Multiple sclerosis is a disease that causes T-cells to destroy the myelin protein sheath around nerves in the central nervous system. This study shows Flt3 inhibitor CEP-701 to be effective in reducing myelin loss in the MS mice model. (See Whartenby et al. PNAS (2005) 102: 16741-16746). The study also shows that hemopoietic cytokines such as Flt3 ligand (a dendritic cell-mobilizing factor) and M-CSF are elevated significantly in the serum of patients with Langerhans cell histiocytosis and systemic lupus erythematosus. Higher level of cytokines correlated with patients having more extensive diseases. The highest serum level of Flt3 and M-CSF were linked with the patients with high risk of extensive skin and /or multisystem involvement, which further implicates Flt3 signaling in the disregulation of dendritic cell progenitors in those autoimmune diseases (See Rolland et al. J Immunol., 2005, 174:3067-3071 ).
There is considerable interest in the development of kinase inhibitors for use in cancer therapy. Among them, urea derivatives have been reported to be selective Flt3 inhibitors in Bioorg. Med. Chem. Lett. 10:2051-2054 (2000), WO 99/32106 published 1 Jul. 1999, PCT publication WO 99/32111 published 1 Jul. 1999 and PCT publication WO2007/109120 published in 27 Sept. 2007.
SUMMARY OF THE INVENTION
The present inventors have now discovered a new kind of tri-heterocyclic derivatives, preaparation process, composition containing the same and uses thereof, particularly to their pharmaceutical use as inhibitors of kinase. The compounds exhibit a surprising increase in the level of inhibition of MV4-1 1 cell proliferation (MV4-11 cell is a human leukemia cell lines that express a FLT3-ITD mutation), and/or a surprising increase in the solubility of the compound (in aqueous media and/or phosphate buffer solution)-enhanced solubility may be of use in formulation the compounds, for example, for administration by an IV route, or for oral formulations (e.g. liquid and small tablet forms) for pediatric use. The oral bioavailability of the compounds of the present invention may be enhanced. The compounds may also be less susceptible to the action of MDR1 in cells.
Accordingly, the first aspect of the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof:
Figure imgf000003_0001
Wherein:
Ar is selected from the group consisting of optionally substituted or unsubstitued aryl, and optionally substituted or unsubstitued heteroaryl, when substituted, the substituents could be one or more groups independently selected from halogen, alkyl, haloalkyl, or hydroxyl alkyl;
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyi, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyl, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyi, heterocycloalkyl, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxylalkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkylalkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHheteroarylalkyl;
Y is O, S, NR2R2' or a direct bond (that means L directly connected with the left heteroaryl);
X-i , X2, X3 and X4 are independently N or CR-i ,
R-i is H, or -Y-L; R, R2 and R2' are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, haloalkoxyl, hydroxyl, amino, aminocarbonyl, sulfonamido, cyano, alkynyl, alkoxyl, aryloxyl, carboxylic acid, carboxylic ester or halogen, or R2, R2' together with the nitrogen atom to which they are attached, form a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms. The 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl. Preferably R2,
R2' and the N could be formed a heterocycle such as
Figure imgf000004_0003
, and R7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-|-C6 straight chain and branched chain alkyl, such as methyl, ethyl, propyl, 2-propyl, n-butyl, /'so-butyl, ferf-butyl, pentyl, neopentyl, hexyl, isohexyl.
One preferably subset of compound of the present invention provides formula (I) or a pharmaceutically acceptable salt thereof, wherein:
Ar is
Figure imgf000004_0002
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl, when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyi, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxyl alkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkylalkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHheteroarylalkyl;
Y is O, S, NR2R2' or a direct bond;
X-i , X2, X3 and X4 are independently N or CR-i ,
Figure imgf000004_0001
R, R2 and R2' are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, haloalkoxyl, hydroxyl, amino, amido, aminocarbonyl, sulfonamido, cyano, alkynyl, alkoxyl, aryloxyl, carboxylic acid, carboxylic ester or halogen, or R2, R2' together with the nitrogen atom to which they are attached, form a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms. The 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl. Preferably R2,
R2' and the N could be formed a heterocycle such as
Figure imgf000004_0004
, and R7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-|-C6 straight chain and branched chain alkyl, such as methyl, ethyl, propyl, 2-propyl, n-butyl, /'so-butyl, ferf-butyl, pentyl, neopentyl, hexyl, isohexyl.
Another preferably subset of compound of the present invention provides formula (I) or a pharmaceutically acceptable salt thereof, wherein:
Figure imgf000005_0001
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl, when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyi, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxyl alkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkylalkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHheteroarylalkyl;
Y is O, S or NR2R2';
X-i , X2, X3 and X4 are independently N or CR-i ,
R-i is H, or -Y-L;
R, R2, and R2' are each independently selected from the group consisting of hydrogen or alkyl, or R2, R2' together with the nitrogen atom to which they are attached, formed a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms. The 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl. Preferably R2, R2' and the N could be formed a heterocycle such as
Figure imgf000005_0002
and R7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-|-C6 straight chain and branched chain alkyl, such as methyl, ethyl, propyl, 2-propyl, n-butyl, /'so-butyl, ferf-butyl, pentyl, neopentyl, hexyl, isohexyl.
Another preferably subset of compound of the present invention provides formula (I) or a pharmaceutically acceptable salt thereof, wherein:
Ar is
Figure imgf000005_0003
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyi, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxyl alkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkylalkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHheteroarylalkyl;
Y is O;
X-i , X2, X3 and X4 are independently N or CH;
R is hydrogen. Another preferably subset of compound of the present invention provides formula (I) or a pharmaceutically acceptable salt thereof, wherein:
Ar is
Figure imgf000006_0001
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyi, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxyl alkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkylalkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHheteroarylalkyl;
Y is NR2R2';
X X2, X3 and X4 are independently N or CH;
R is hydrogen;
R2, R2' is hydrogen or alkyl, or R2, R2' together with the nitrogen atom to which they are attached, formed a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms. The 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl. Preferably R2, R2' and the N could be framed a heterocycle such as
Figure imgf000006_0002
and R7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-|-C6 straight chain and branched chain alkyl, such as methyl, ethyl, propyl, 2-propyl, n-butyl, /'so-butyl, ferf-butyl, pentyl, neopentyl, hexyl, isohexyl.
Another preferably subset of compound of the present invention provides formula (I) or a pharmaceutically acceptable salt thereof, wherein:
Ar is
Figure imgf000006_0003
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyi, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxyl alkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkylalkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHheteroarylalkyl;
Y is a direct bond or O, S or NR2R2';
X X2, X3 and X4 are independently CR
R is H, or -Y-L; R is hydrogen;
R2, R2' is hydrogen or alkyl, or R2, R2' together with the nitrogen atom to which they are attached, formed a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms. The 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl. Preferably R2, R2'and the N could be formed a heterocycle such as
Figure imgf000007_0002
and R7 could be the groups independently selected from H, methylsulfonyl, lower alkyl, that is C-|-C6 straight chain and branched chain alkyl, such as methyl, ethyl, propyl, 2-propyl, n-butyl, /'so-butyl, ferf-butyl, pentyl, neopentyl, hexyl, isohexyl.
Another preferably subset of compound of of the present invention provides formula (I) or a pharmaceutically acceptable salt thereof, wherein:
Figure imgf000007_0003
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyi, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyi, heterocycloalkyi, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxyl alkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkylalkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHheteroarylalkyl;
Y is a direct bond;
X X2, X3 and X4 are independently N or CR
R is H, or -Y-L;
R is independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, haloalkoxyl, hydroxyl, amino, aminocarbonyl, sulfonamido, cyano, alkynyl, alkoxyl, aryloxyl, carboxylic acid, carboxylic ester or halogen.
The present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein the compounds are selected from, but not limited to the following compounds:
Figure imgf000007_0001
Figure imgf000008_0001
Figure imgf000009_0001
Figure imgf000010_0001
Figure imgf000011_0001
The second aspect of the present invention provides a pharmaceutical composition comprising a compound of formula (I) as defined in the above mentioned, or a pharmaceutically acceptable salt thereof, as well as a pharmaceutically acceptable carrier, adjuvant, excipient, or diluent in a therapeutically effective dose.
The third aspect of the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in the above mentioned for the use in preparation of a medicament as Flt3 kinase inhibitor.
The fourth aspect of the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in the above mentioned for the use in preparation of a medicament by modulating FLT3-mediated diseases, especially said medicament comprising administering a therapeutically effective amount of a compound of formula (I) as defined in the above mentioned, or a pharmaceutically acceptable salt thereof, as well as an isomer, a solvate, a hydrate, or a prodrug.
The fifth aspect of the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in the above mentioned for the use in preparation of a medicament for treating a cellular proliferative disorderes diseases by inhibiting Flt3 kinase activities, wherein said diseases such as acute myeloid leukemia, chronic myeloid leukemia, myeloma, ovarian cancer, breast cancer, prostate cancer, pancreatic cancer, colorectal cancer, stomach cancer, non small cell lung cancer, thyroid cancer, brain cancer or lymphoma, said medicament comprising administering a therapeutically effective amount of a compound of formula (I) as defined in the above mentioned, or a pharmaceutically acceptable salt thereof.
the sixth aspect of the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in the above mentioned for the use in preparation of a medicament for treating inflammatory and autoimmune diseases by inhibiting Flt3 kinase activities, wherein said diseases such as asthma, lupus, systemic lupus, rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, or Crohn's disease, said medicament comprising administering a therapeutically effective amount of a compound of formula (I) as defined in the first aspect, or a pharmaceutically acceptable salt thereof. DETAILED DESCRIPTION OF THE INVENTION
Unless otherwise stated, the following terms used in the specification and claims have the meanings discussed below.
Said "alkyl" refers to a saturated aliphatic hydrocarbon group including C1-C20 straight chain and branched chain groups. Preferably an alkyl group is a moderate size alkyl having C1-C10 straight chain and branched chain groups, e.g., methyl, ethyl, propyl, 2-propyl, n-butyl, /'so-butyl, ferf-butyl, pentyl, neopentyl, hexyl, isohexyl, heptyl, octyl, and the like. More preferably, it is a lower alkyl having C1-C6 straight chain and branched chain groups, e.g., methyl, ethyl, propyl, 2-propyl, n-butyl, /'so-butyl, or ferf-butyl, pentyl, neopentyl, hexyl, isohexyl, and the like.
Said "optionally substituted alkyl" refers to an alkyl group which may be substituted at any available point of attachment and by one to 4 subtituents selected from the group consisting of halo, hydroxyl, lower alkoxy, aryl(optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), aryloxy(optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), heteroaryl(optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), heterocycloalkyl(optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), haloalkoxyl, amino, aminocarbonyl, cyano, alkynyl, carboxylic acid, or carboxylic ester.
Said "cycloalkyl" refers to a 3 to 8 membered all-carbon monocyclic ring, an all-carbon 5-membered/6-membered or 6-membered/6-membered fused bicyclic ring or a multicyclic fused ring (a "fused" ring system means that each ring in the system shares an adjacent pair of carbon atoms with other ring in the system) group wherein one or more rings may contain one or more double bonds, but none of the rings has a completely conjugated p/'-electron system. Examples of cycloalkyl groups are cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, chcyclohexyl, cyclohexadienyl, adamantyl, cycloheptyl, cycloheptatrienyl, and the like. The cycloalkyl group may be substituted or unsubstituted. When substituted, the substituent group(s) is preferably one or more independently selected from the group consisting of lower alkyl, trihaloalkyl, halo, hydroxy, hydroxylalkyl,aminoalkyl, amino, aryl (optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), aminocarbonyl, sulfonamido, ureido, aryloxy (optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), 6-membered heteroaryl (having 1 to 3 nitrogen atoms on the ring, the carbons on the ring being optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), 5-membered heteroaryl (having 1 to 3 heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, the carbon and nitrogen atoms of the group being optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), 5- or 6-membered heterocyclic alkyl (having 1 to 3 heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, the carbon and nitrogen (if present) atoms of the group being optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), mercapto, (lower alkyl) thio, arylthio (optionally substituted with one or more groups which each independently is halo, hydroxy, lower alkyl or lower alkoxy groups), cyano, acyl, thioacyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, nitro.
Said "alkenyl" refers to an alkyl group as defined above having at least 2 carbon atoms and at least one carbon-carbon double bond. Representative examples include, but are not limited to ethenyl,
1 - propenyl, 2-propenyl, 1-, 2-, 3-butenyl, and the like. The optionally substituted alkenyl means the the alkenyl which may be substituted with one or more groups which each independently is halo, cyano, lower alkyl or lower alkoxy groups.
Said "alkynyl" refers to an alkyl group as defined above having at least 2 carbon atoms and at least one carbon-carbon triple bond. Representative examples include, but are not limited to ethynyl, 1-propynyl,
2- propynyl, 1-, 2-, 3-butynyl, and the like. The optionally substituted alkenyl means the alkynyl which may be substituted with one or more groups which each independently is halo, cyano, lower alkyl or lower alkoxy groups.
Said "aryl (Ar)" refers to groups having at least one aromatic ring, i.e., having a conjugated p/'-electron system, including all-carbon cyclic aryl, and biaryl group. Examples of aryl include phenyl, nathathyl, and the like. Said aryl group may be optionally substituted with one or more groups each independently selected from the group consisting of halo, trihalomethyl, hydroxy, hydroxylalkyl, mercapto, alkylthio, nitro, cyano, alkoxyl and alkyl.
Said "heteroaryl (Het)" refers to an aryl having 1 to 3 heteroatoms selected from the group consisting of N, O, and S as ring atoms, the remaining ring atoms being C. Said ring is 5- or 6- membered ring. The examples of heteroaryl groups include furyl, thienyl, pyridinyl, pyrrolyl, N-alkyl pyrrolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazole, pyrimidinyl, pyrazinyl, imidazolyl, triazolyl, tetrazolyl, oxatriazolyl, pyridazinyl, triazinyl, oxadiazolyl, thiadiazolyl, furazanyl, and the like. Said heteroaryl group may be optionally substituted with (via a carbon ot hetero atom) one or more groups each independently selected from the group consisting of halo, , haloalkyl, hydroxy, hydroxylalkyl, mercapto, alkylthio, nitro, cyano, alkoxyl and alkyl.
Said "heterocycloalkyl" refers to a mono-heterocycloalkyl, bi-heterocycloalkyl, bridged-hetero ycloalkyl or spiro-heterocycloalkyl of 4 to 12 ring atoms, wherein one, or two ring heteroatoms are selected from the group consisting of N, O, and S (O)n (n is integer from 0 to 2), the remaining ring atoms are C, in addition, the ring may also have one or more double bonds, but not have a completely conjugated p/'-electron system.
Said mono heterocycloalkyl includes, but is not limited to azetidyl, pyrrolidyl, piperidyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, and the like.
The examples of said bi-heterocycloalkyl include but not limited to
Figure imgf000013_0001
The example of said bridged- heterocycloalkyl included but not limited to
Figure imgf000013_0002
The examples of said spiro-heterocycloalkyl include but not limited to
Figure imgf000014_0001
Said "heterocycloalkyi" may be substituted or unsubstituted. When substituted, the substituent group is preferably one or more, more preferably one, two, or three, further more preferably one or two groups, each independently selected from the group consisting of lower alkyi, lower hydroxylalkyl, trihaloalkyl, halo, hydroxy, haloalkyl, mercapto, nitro, lower alkoxyl, cyano, amino, aminoalkyl, alkoxyalkyl, cycloalkyi, heterocycloalkyi, methylsulfonyl, and acyl. Said "cycloalkylalkyl" refers to a radical of the formula - RaRb, where Ra is an alkyi radical as defined above and Rb is a cycloalkyi radical as defined above. The alkyi radical and the cycloalkyi radical may be optionally substituted as defined above.
Said "arylalkyl" refers to a radical of the formula - RaRc where Ra is an alkyi radical as defined above and Rc is aryl radicals as defined above, e.g., benzyl, diphenylmethyl and the like. The alkyi radical and the aryl radical(s) may be optionally substituted as described above.
Said "Heterocycloalkylalkyl" refers to a radical of the formula - RaRd where Ra is an alkyi radical as defined above and Rd is a heterocycloalkyi radical as defined above, and if the heterocycloalkyi is a nitrogen-containing heterocycloalkyi, the heterocycloalkyi may be attached to the alkyi radical at the nitrogen atom or at carbon atom. The alkyi part of the heterocycloalkyi radical may be optionally substituted as defined above for an alkyi group. The heterocycloalkyi part of the heterocycloalkyi radical may be optionally substituted as defined above for a heterocycloalkyi group.
Said "Heteroarylalkyi" refers to a radical of the formula -RaRe where Ra is an alkyi radical as defined above and Re is a heteroaryl radical as defined above. The heteroaryl part of the heteroarylalkyi radical may be optionally substituted as defined above for a heteroaryl group. The alkyi part of the heteroarylalkyi radical may be optionally substituted as defined above for an alkyi group.
Said "hydroxy" refers to an -OH group.
Said "alkoxyl" refers to both an -O-(alkyl) and an -0-(unsubstituted cycloalkyi) group. Representative examples include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like.
Said "amino" refers to a -NH2, -NHCH3, -NHCH2CH3, -NH-n-propyl, -NH-isopropyl, -N(CH3)2, -NHcyclopropyl, -NHPh, -NHpyridyl, pyrrolidinyl, piperazinyl, morpholino, piperidino, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkylalkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHheteroarylalkyl, etc.
Said "-NHalkylhydroxyl " refers to a-NH(alkyl)hydroxyl, wherein alkyi is as defined above. Representative examples include, but are not limited to, e.g. -NHCH2CH2OH, -NHCH2CH(OH)CH2OH, etc.
Said "-NHalkoxylalkyl" refers to a-NH(alkyl)-0-(alkyl), wherein alkyi is as defined above.
Said "-NHalkylamnio" refers to -NH(alkyl)amnio, wherein alkyi and amnio is as defined above. Representative examples include, but are not limited to, e.g. -NHCH2CHCNH2, -NHCH2CH2NHCH3,
-NHCH2CH2N(CH3)2, -NHCH2CH(OH)H2NH2,
Figure imgf000014_0002
, etc.
Said "-NHcycloalkyl" refers to NH-cycloalkyl, wherein cycloalkyd is as defined above.
Said "-NHcycloalkylalkyl" refers to a -NH-alkyl-cycloalkyl, wherein alkyi and cycloalkyi are as defined above. Representative examples include, but are not limited to, e.g. NH-CH2-cyclopropyl-NH2, NH-CH2-
Figure imgf000014_0003
Said "-NHheterocycloalkyl" refers to NH-heterocycloalkyl, wherein heterocycloalkyl is as defined above.
Said "-NHheterocycloalkylalkyl" refers to NH-heterocycloalkyl-alkyl, wherein heterocycloalkyl and alkyl is as defined above.
Said "-NHaryl" refers to NH-aryl, wherein aryl is as defined above.
Said "-NHarylalkyl" refers to a -NH-alkyl-aryl, wherein aryl and alkyl are as defined above.
Representative examples include, but are not limited to, e.g.
Figure imgf000015_0002
and the like.
Said "-NH heteroarylalkyl" refers to a -NH-alkyl-heteroaryl, wherein heteroaryl and alkyl are as defined above. Representative examples include, but are not limited to, e.g.
Figure imgf000015_0001
and the like.
Said "haloalkoxy" refers to an -O-(haloalkyl). Representative examples include, but are not limited to, trifluoromethoxy, trichloromethoxy, tribromomethoxy, and the like.
Said "aryloxyl" refers to both an -O-aryl and an -O-heteroaryl group, wherein the aryl and heteroaryl are as defined above. Representative examples include, but are not limited to, phenoxy, pyridinyloxy, furanyloxy, thienyloxy, pyrimidinyloxy, pyrazinyloxy, and the like, and derivatives thereof.
Said "mercapto" refers to a -SH group.
Said "alkylthio" refers to a -S-(alkyl) and a -S-(unsubstituted cycloalkyl) group. Representative examples include, but are not limited to, methylthio, ethylthio, propylthio, butylthio, cyclopropylthio, cyclobutylthio, cyclopentylthio, cyclohexylthio, and the like.
Said "arylthio" refers to a -S-aryl and a -S-heteroaryl group, wherein the aryl and heteroaryl are as defined above. Representative examples include, but are not limited to, e.g., phenylthio, pyridinylthio, furanylthio, thienylthio, pyrimidinylthio, and the like, and derivatives thereof.
Said "acyl" refers to a -C(0)-R3" group, where R3 is selected from the group consisting of hydrogen, lower alkyl, trihalomethyl, unsubstituted cycloalkyl, aryl (optionally substituted with one or more, preferably one, two, or three substituents selected from the group consisting of lower alkyl, trihalomethyl, lower alkoxy and halo groups), heteroaryl (bonded through a ring carbon) (optionally substituted with one or more, preferably one, two, or three substitutents selected from the group consisting of lower alkyl, trihaloalkyl, lower alkoxy and halo groups), and heteroalicyclic (bonded through a ring carbon) (optionally substituted with one or more, preferably one, two, or three substituents selected from the group consisting of lower alkyl, trihaloalkyl, lower alkoxy and halo groups). Representative acyl groups include, but are not limited to, acetyl, trifluoroacetyl, benzoyl, and the like.
Said "thioacyl" refers to a -C(S)-R3 group, wherein R3 is as defined above.
Said "acetyl" refers to a -C(=0)CH3 group.
Said "halo" refers to fluoro, chloro, bromo, or iodo, preferably fluoro or chloro.
Said "haloalkyl," refers to a (CH3)2CH2FC-, or a CH3(CH2F)2C-, or a (CH2F)3C- group.
Said "cyano" refers to a -C≡N group.
Said "trihaloalkyl, or trihalomethyl" refers to a CF3 or the like, wherein the halo is define as above. Said "carboxylic acid" refers to a -COOH group.
Said "carboxylic ester" refers to a -COOR3 group, wherein R3 is alkyl or cycloalkyl.
Said "hydroxyalkyl" refers to a -alkyl-OH group, wherein alkyl and hydroxyl as defined above. The Representative hydroxyl alkyl group include but are not limited to, -CH2OH, -CH2CH2OH, CH3CH(OH)CH2-, (CH3)2(CH2OH)C-, CH3(CH2OH)2C-, or (CH2OH)3C-, and the like.
Said "aminoalkyl" refers to a -alkyl-amnio group, wherein alkyl and amnio are as defined above. The Representative amnioalkyl group include but are not limited to -CH2NH2, -CH2NHCH3, -CH2N(CH3)2,
-CH2CH2NH2, -CH2CH2NHCH3, -CH2CH2N(CH3)2,
Figure imgf000016_0003
, and the like.
Said "amido" refers to a -C(=0)NR4R5 which R4 and R5 refer to amino substituents, R4 may be the same as (or may not be the same as ) R5, which the substituents could be hydrogen or alkyl (difined as above). Representative amido groups include, -C(=0)NH2, -C(=0)NHCH3, -C(=0)N(CH3)2, -C(=0)NCH3CH2CH3 as well as the groups in which R4 and R5, together with the nitrogen atom to which they are attached, formed a heterocyclic ring, like morpholino, piperazinyl, piperidino, and the like.
Said "aminocarbonyl" refers to -NHC(=0)CH3, -NCH3C(=0)CH3, -NHC(=0)CH2CH3, -NCH3C(=0)CH2CH3, -NHC(=0)-cyclopropyl, -NCH3C(=0)-cyclopropyl, -NHC(=0)Ph, -NCH3C(=0)Ph, and the like.
Said "sulfonamido" refers to a -NR4S(=0)2R6, or a -NR4R5S(=0)2R6, in which R4 may be the same as (or may not be the same as ) R5 and R4R5 are hydrogen or alkyl or R4 and R5, together with the nitrogen atom to which they are attached, form a heterocyclic ring, like morpholino, piperazinyl, piperidino and the like. R6 refers to a sulfonamino substituents, for example, C lower alkyl, C3.8 cycloalkyl, C3.20 heterocycloalkyl, C5.20 aryl, and C5.20 heteroaryl, wherein alkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl are as defined above. Representative sulfonamido groups include, -NHS(=0)2CH3
-NCH3S(=0)2CH3, -NHS(=0)2Ph, and -NCH3S(=0)2Ph,
Figure imgf000016_0001
Figure imgf000016_0002
Ar and Het are as described above.
Said "ureido" refers to -NR3C(0)NR4R5 where R3 is hydrogen or alkyl; R4 and R5 are defined as above.
Said "methylsulfonyl" refers to CH3S02-.
Said "prodrug" refers to a compound which, when metabolized in vivo, converts back to the original active compound. Typically, the prodrug is inactive, or less active than the active compound, but may provide advantageous handling, administration, or metabolic properties.
Said "optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance may or may not occur. For example, "heterocyclyl optionally substituted with an alkyl group" means that the alkyl may or may not be present, and the description includes situations where the heterocyclic group is substituted with an alkyl group and situations where the heterocyclic group is not substituted with the alkyl group.
Said "pharmaceutical composition" refers to a mixture of one or more of the compounds of formula (I) as described in present invention herein, or hydrates, or solvates, or isomers, or physiologically/pharmaceutically acceptable salts or prodrugs thereof, with other chemical components, such as physiologically/pharmaceutically acceptable carriers and recipients. The purpose of a pharmaceutical composition is to facilitate administration of a compound to warm blood animals and human.
Said "examplary compound" refers to the compound which given from the present invention, such as: the examplary compound 8-1 is illustrated as example 1 , the examplary compound 8-2 is illustrated as example 2, and etc. SYNTHESIS OF THE COMPOUNDS OF THE INVENTION
The ureas, or cyclic forms thereof, compounds of the present invention may be synthesized by methods well known to those of skill in the art or using methods known in the art in combination with methods described herein.
Method 1 :
Nitroprimidinechloride compound 1 used as a starting material, after substituted by morpholine, the nitro group was reduced by iron powder to obtain amine compound 3, condensed with compound 4 and then reduced by iron powder to get the key intermediate amine 6, which was coupled with isocyanate 7 to afford the target urea compound, as showed in scheme 1.
Figure imgf000017_0001
Method 2:
Compound 9 used as a starting material, the thiazole ring was opened by basic hydrolysis, then re-closed by acetyl chloride to get intermediate 12. After compound 12 was condensed with compound 4 to obtain the nitro compound 14, de-methylation by HBr to get the phenol intermediate 15, alkylated the phenol group by chloride compound 16, and the the nitro compound 17 was reduced into amine 18 by iron powder, the target urea compound was obtained by reacting 18 with isocyanate 7, as showed in scheme 2.
Figure imgf000017_0002
Method 3:
Chloronitropyridine 20 was used as starting material, after methoxylation, reduced nitro 21 into amine 22 by iron powder, then was converted into amniothiazole 23, demethylation to obtain phenol 24, condensed with compound 4 to obtain tricyclic nitro compound 25, after reacted with chloride 16, reduced nitro into amine 27 by iron powder, the final target urea compound was obtained by reacting 27 with isocyanate 7 as showed in scheme 3-A.
The other invented compounds could be made by from scheme 3-B to scheme 3-I.
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Method 4:
Nitroprimidinechloride compound 1 used as a starting material, after substituted by 2,2-dimethoxyethanamine 91 , then reduced the nitro 91 by iron powder to obtain amine compound 92, the compound 92 was then condensed with compound 4, followed by the reduction of nitro group with zinc powder into amine 94, coupled with isocyanate 7 to form urea intermediate 95, and acid hydrolysis acetal into aldehyde 96 which was finally formed different 97 series compounds by reduction amnination with different amines, as showed in scheme 4-A.
Figure imgf000021_0002
Figure imgf000022_0001
Scheme 4-A
The ureas may be converted to thioureas by treating with Lawesson's reagent in the presence of toluene.
The term "protecting group Cbz" (see scheme 3-B, 3-C, 3-F) refers to chemical moieties that block some or all reactive moieties and prevent such groups from participating in chemical reactions until the protective group is removed. It is preferred that each protective group be removable by a different means. Protective groups that are cleaved under totally disparate reaction conditions fulfill the requirement of differential removal. Protective groups can be removed by acid, base, and hydroge no lysis. Groups such as trityl, dimethoxytrityl, acetal and t-butyldimethylsilyl are acid labile and may be used to protect carboxy and hydroxy reactive moieties in the presence of amino groups protected with Cbz groups, which are removable by hydrogenolysis.
The present invention also relates to a compounds of formula (I) or a pharmaceutically acceptable salt thereof, wherein a compound of formula (I) may be present in the form of free base or in the form of salts formed by acid addition which are pharmaceutically acceptable non-toxic. The pharmaceutically acceptable salts include hydrochloride, p-toluenesulfonate, benzenesulfonate, naphthalenesulfonate, tartarate, maleate, lactate, methanesulfonate, ethanesulfonate, sulfate, phosphate, citrate, acetate and trifluoroacetate, preferably p-toluenesulfonate, benzenesulfonate, methanesulfonate, hydrochloride, tartarate and trifluoroacetate.
Furthermore, the present invention relates to a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof in an effective therapeutic dose, and a pharmaceutically acceptable carrier, and a use of the compounds of the present disclosure or pharmaceutical acceptable salts in the preparation of a medicament as a kinase inhibitor. In other words, this disclosure also provides a pharmaceutical composition comprising the above mentioned compounds in an effective therapeutic dose, as well as their use in the preparation of a medicament as Flt3 inhibitor.
The compounds of the present invention are useful in various pharmaceutically acceptable salt forms. The term "pharmaceutically acceptable salt" refers to those salt forms which would be apparent to the pharmaceutical chemist, i.e., those which are substantially non-toxic and which provide the desired pharmacokinetic properties, palatability, absorption, distribution, metabolism or excretion. Other factors, more practical in nature, which are also important in the selection, are cost of the raw materials, ease of crystallization, yield, stability, hygroscopicity and flowability of the resulting bulk drug. Conveniently, pharmaceutical compositions may be prepared from the active ingredients in combination with pharmaceutically acceptable carriers..
The pharmaceutically acceptable salts of the compounds of formula (I) include conventional non-toxic salts or quarternary ammonium salts of the compounds of formula (I) formed e.g. from non- toxic inorganic or organic acids. For example, non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glyeolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, sulfanilic, 2-acetoxybenzoic, fumaric, naphthalenesulfonic, toluenesulfonic, benzenesulfonic, ethanesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like.
The pharmaceutically acceptable salts of the present invention can be synthesized by conventional chemical methods.
Generally, the salts are prepared by reacting the free base or acid with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid or base, in a suitable solvent or solvent combination.
The compounds of the present invention may have asymmetric centers and occur as racemates, racemic mixtures, and as individual diastereomer. All such isomers including optical, enantiomeric, diasteriomeric, stereoisomeric, epimeric, and geometric isomers are included in the present invention.
The compounds of the present invention may be in crystalline or non- crystalline form, it may exist in a number of different polymorphic forms, and may optionally be hydrated or solvated. This invention includes within its scope stoichiometric hydrates as well as compounds containing variable amount of water.
The present invention described herein also includes a pharmaceutical composition comprising a therapeutically effective amount of compound of formula (I) or a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable carrier in the use of preparation of a medicament as kinase inhibitors.
Compounds provided herein are useful in treating conditions characterized by inappropriate FLT3 activity such as proliferative disorders. FLT3 activity includes, but is not limited to, enhanced FLT3 activity resulting from increased or de novo expression of FLT3 in cells, increased FLT3 expression or activity, and FLT3 mutations resulting in constitutive activation. The existence of inappropriate or abnormal FLT3 ligand and FLT3 levels or activity can be determined using well known methods in the art. For example, abnormally high FLT3 levels can be determined using commercially available ELISA kits. FLT3 levels can be determined using flow cytometric analysis, immunohistochemical analysis, and in situ hybridization techniques.
An inappropriate activation of the FLT3 can be determined by an increase in one or more of the activities occurring subsequent to FLT3 binding: (1 ) phosphorylation or autophosphorylation of FLT3; (2) phosphorylation of a FLT3 substrate, e.g., Stat5, Ras; (3) activation of an related complex, e.g., PI3K; (4) activation of an adaptor molecule; and (5) cellular proliferation. These activities are readily measured by well known methods in the art.
A compound of formula (I) disclosed herein would be useful for, but not limited to, the prevention or treatment of proliferative diseases, conditions, or disorders in a patient by administering to the patient a compound of formula (I) or a composition comparising a compound of formula (I) disclosed herein in an effective amount. Such diseases, conditions, or disorders include cancer, particularly hematopoietic cancer, metastatic cancer, atherosclerosis, and lung fibrosis.
Compounds disclosed herein would be useful for the treatment of neoplasia including cancer and metastasis, including, but not limited to: carcinoma such as cancer of the bladder, breast, colon, kidney, liver, lung (including small cell lung cancer), esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage (including leukemia, acute lymphocitic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma); hematopoietic tumors of myeloid lineage (including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia); tumors of mesenchymal origin (including fibrosarcoma and rhabdomyosarcoma, and other sarcomas, e.g. soft tissue and bone); tumors of the central and peripheral nervous system (including astrocytoma, neuroblastoma, glioma and schwannomas); and other tumors (including melanoma, seminoma, teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma, thyroid follicular cancer and Kaposi's sarcoma).
The compounds also would be useful for treatment FLT3-mediated and/or CSF-1 R mediated diseases like immune dysfunction, autoimmune diseases, kidney diseases, tissue transplant rejection, lupus erythematosis, multiple sclerosis, inflammatory bowel disease, rheumatoid arthritis, arthritis, asthma.
The compounds disclosed herein are also useful in the treatment of diabetic conditions such as diabetic retinopathy and microangiopathy.
The compounds disclosed herein are also useful in the reduction of blood flow in a tumor in a subject. The compounds disclosed herein are also useful in the reduction of metastasis of a tumor in a subject. Besides being useful for human treatment, these compounds are also useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like. In other embodiments, animals include horses, dogs, and cats. As used herein, the compounds of formula(l) disclosed herein include the pharmaceutically acceptable derivatives thereof.
Where the plural form is used for compounds, salts, and the like, this is taken to refer to also a single compound, salt, and the like.
The treatment method that includes administering a compound or composition disclosed herein can further include administering to the patient an additional therapeutic agent (combination therapy) selected from: a chemotherapeutic or anti-proliferative agent, or an anti-inflammatory agent, wherein the additional therapeutic agent is appropriate for the disease being treated and the additional therapeutic agent is administered together with a compound or composition disclosed herein as a single dosage form or separately from the compound or composition as part of a multiple dosage form. The additional therapeutic agent may be administered at the same time as a compound disclosed herein or at a different time. In the latter case, administration may be staggered by, for example, 6 hours, 12 hours, 1 day, 2 days, 3 days, 1 week, 2 weeks, 3 weeks, 1 month, or 2 months.
The invention also features a method of inhibiting the growth of a cell that expresses VEGFR, or c-Met, that includes contacting the cell with a compound or composition disclosed herein, thereby causing inhibition of growth of the cell. Examples of a cell whose growth can be inhibited include: a breast cancer cell, a colorectal cancer cell, a lung cancer cell, a papillary carcinoma cell, a prostate cancer cell, a lymphoma cell, a colon cancer cell, a pancreatic cancer cell, an ovarian cancer cell, a cervical cancer cell, a central nervous system cancer cell, an osteogenic sarcoma cell, a renal carcinoma cell, a hepatocellular carcinoma cell, a bladder cancer cell, a gastric carcinoma cell, a head and neck squamous carcinoma cell, a melanoma cell, or a leukemia cell.
The invention also provided herein a method of inhibiting VEGFR, and/or c-Met kinase activity in a biological sample that includes contacting the biological sample with a compound or composition disclosed herein. The term "biological sample" as used herein, means a sample outside a living organism and includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof. Inhibition of kinase activity, particularly VEGFR or c-Met kinase activity, in a biological sample is useful for a variety of purposes known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ-transplantation, biological specimen storage, and biological assays.
In certain embodiments disclosed herein, an "effective amount" or "effective dose" of the compound or pharmaceutically acceptable composition is that amount effective for treating or lessening the severity of one or more of the aforementioned disorders. The compounds and compositions, according to the method disclosed herein, may be administered using any amount and any route of administration effective for treating or lessening the severity of the disorder or disease. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like. A compound or composition can also be administered with one or more other therapeutic agents, as discussed above.
The compounds disclosed herein or pharmaceutical compositions thereof may also be used for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters. Vascular stents, for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury). However, patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a compound disclosed herein.
When administered to a patient for the treatment of cancer, the dosage used can be varied depending upon the type of cancer, the age and general condition of the patient, the particular compound administered, the presence or level of toxicity or adverse effects experienced with the drug, and other factors. A representative example of a suitable dosage range is from as low as about 0.01 mg/kg to as high as about 100 mg/kg. However, the dosage administered is generally left to the discretion of the physician.
The methods of treatment are preferably carried out by delivering a compound of formula (I) orally or parenterally. The term 'parenteral' as used herein includes intravenous, intramuscular, or intraperitoneal administration. The subcutaneous and intramuscular forms of parenteral administration are generally preferred. The instant invention can also be carried out by delivering the compound of formula I subcutaneously, intranasally, intrarectally, transdermal^ or intravaginally.
The compounds of formula (I) may also be administered by inhalation. By 'inhalation' is meant intranasal and oral inhalation administration. Appropriate dosage forms for such administration, such as an aerosol formulation or a metered dose inhaler, may be prepared by convention techniques.
The invention also relates to a pharmaceutical composition comprising a compound of formula (I) and a pharmaceutically acceptable carrier. The compounds of formula (I) may also be included in pharmaceutical compositions in combination with a second therapeutically active compound. The pharmaceutical carrier employed may be, for example, either a solid, liquid or gas. Exemples of solid carriers include lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid and the like. Exemples of liquid carriers are syrup, peanut oil, olive oil, water and the like. Examples of gaseous carriers include carbon dioxide and nitrogen. Similarly, the carrier or diluent may include time delay material well known in the art, such as glyceryl monostearate or glycervl distearate, alone or with a wax.
A wide variety of pharmaceutical dosage forms can be employed. If a solid dosage is used for oral administration, the preparation can be in the form of a tablet, hard gelatin capsule, troche or lozenge. The amount of solid carrier will vary widely, but generally will be from about 0.025 mg to about 1 g. When a liquid dosage form is desired for oral administration, the preparation is typically in the form of a syrup, emulsion, soft gelatin capsule, suspension or solution. When a parenteral dosage form is to be employed, the drug may be in solid or liquid form, and may be formulated for administration directly or may be suitable for reconstitution. Topical dosage forms are also included. Examples of topical dosage forms are solids, liquids and semi-solids. Solids would include dusting powders, poultices and the like. Liquids include solutions, suspensions and emulsions. Semi-solids include creams, ointments, gels and the like. The amount of a compound of formula (I) used topically will, of course, vary with the compound chosen, the nature and severity of the condition, and can be varied in accordance with the discretion of the physician. A representative, topical, dose of a compound of formula I is from as low as about 0.01 mg to as high as about 2.0 g, administered one to four, preferably one to two times daily.
he active ingredient may comprise, for topical administration, from about 0.001 % to about 10% w/w. Drops according to the present invention may comprise sterile or non-sterile aqueous or oil solutions or suspensions, and may be prepared by dissolving the active ingredient in a suitable aqueous solution, optionally including a bactericidal and/or fungicidal agent and/or any other suitable preservative, and optionally including a surface active agent. The resulting solution may then be clarified by filtration, transferred to a suitable container which is then sealed and sterilized by autoclaving or maintaining at 98- 100°C for half an hour. Alternatively, the solution may be sterilized by filtration and transferred to the container aseptically. Examples of bactericidal and fungicidal agents suitable for inclusion in the drops are phenylmercuric nitrate or acetate (0.002%), benzalkonium chloride (0.01 %) and chlorhexidine acetate (0.01 %). Suitable solvents for the preparation of an oily solution include glycerol, diluted alcohol and propylene glycol.
Lotions according to the present invention include those suitable for application to the skin or eye. An eye lotion may comprise a sterile aqueous solution optionally containing a bactericide and may be prepared by methods similar to those for the preparation of drops. Lotions or liniments for application to the skin may also include an agent to hasten drying and to cool the skin, such as an alcohol or acetone, and/or a moisturizer such as glycerol or an oil such as castor oil or arachis oil.
Creams, ointments or pastes according to the present invention are semi-solid formulations of the active ingredient for external application. They may be made by mixing the active ingredient in finely- divided or powdered form, alone or in solution or suspension in an aqueous or non-aqueous liquid, with a greasy or non-greasy base. The base may comprise hydrocarbons such as hard, soft or liquid paraffin, glycerol, beeswax, a metallic soap; a mucilage; an oil of natural origin such as almond, com, arachis, castor or olive oil; wool fat or its derivatives, or a fatty acid such as stearic or oleic acid together with an alcohol such as propylene glycol or macrogels. The formulation may incorporate any suitable surface active agent such as an anionic, cationic or non-ionic surfactant such as sorbitan esters or polyoxyethylene derivatives thereof. Suspending agents such as natural gums, cellulose derivatives or inorganic materials such as silicas, and other ingredients such as lanolin may also be included.
Suitable coatings and the general preparation of coated implantable devices are described in US
6099562, 5886026 and 5304121 , the contents of each of which are incorporated by reference herein. The coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof. The coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysacca rides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics into the composition. Implantable devices coated with a compound disclosed herein are another embodiment disclosed herein. The compounds may also be coated on implantable medical devices, such as beads, or co-formulated with a polymer or other molecule, to provide a "drug depot" thus permitting the drug to be released over a longer time period than administration of an aqueous solution of the drug.
BRIEF DESCRIPTION OF THE DRAWING
FIG.1 is demonstrated the antitumor effect on MV4-11 acute myeloid leukemia zenograft tumor model of the examplary compound of the present invention. A (solvent) is used as a negative control group, B (Quizartinib) is used as a positive control group, and C (the examplary compound of the present invention) is used in tested group. EXAMPLES
The following examples serve to illustrate the compounds in this invention and the preparation process, but the examples should not be considered as limiting the scope of the invention.
The structures of all compounds were identified by nuclear magnetic resonance ( H NMR) and mass spectrometry (MS). H NMR chemical shifts (δ) were recorded in ppm (10~6). NMR was performed on a Bruker AVANCE-400 spectrometer. The suitable solvents were deuterated-chloroform (CDCI3), deuterated-dimethyl sulfoxide (DMSO-cf6) and deuterated-methanol (CD3OD) with tetramethylsilane (TMS) as an internal standard and chemical shifts were recorded in ppm (10~6).
The analytical low-resolution mass spectra (MS) were recorded on Agilent 1200 HPLC/6120 using a
XBridge C18, 4.6 x 50 mm, 3.5 μιη, using a gradient elution method: 5%-95% B for 1.5 min followed by 95% B over 2 min.
Solvent A:(10mM Ammonium hydrogen carbonate in water)
Solvent B: Acetonitrile
The average of inhibitory rate of Flt3 kinase and IC50 was determined by a Caliper's mobility shift assay (MSA).
Thin-layer silica-gel was Yantai Huanghai HSGF254 or Qingdao GF254 silica-gel plate. Column chromatography generally used Yantai Huanghai 200-300 mesh silica gel as carrier.
The following abbreviations have been used:
DMSO-D6: deuterated-dimethyl sulfoxide;
CDCI3: deuterated-chloroform;
CD3OD: deuterated-methanol;
THF: tetrahydrofuran;
DMF: N,N-dimethylformamide;
EtOAc: ethyl acetate;
MeOH: methanol;
EtOH: ethanol;
MeCN: acetonitrile;
DIPEA: diisopropylethylamine;
TEA: triethylamine;
DCM: dichloromethane;
HOAc: acetic acid;
Cbz: benzyloxycarbonyl;
Mel: iodomethane;
HCHO: formaldehyde;
NaH: sodium hydride;
Br2: bromine;
HBr: hydrogen bromide;
KSCN: potassium thiocyanate;
t-BuOK: potassium tert-butoxide;
Fe: iron powder;
K2C03: potassium carbonate;
KOH: potassium hydroxyide;
DPPA: Diphenylphosphonic azide;
CH3SO2CI (TsCI): methyl aminosulfonyl; NaBH3CN: sodium cyanoborohydride;
NH2NH2: hydrazine.
All the following intermediates were prepared according to either the reported literatures or by the methods described in this invention.
Examples 1-6 (Compounds 8-1 to 8-6 were made by the method in scheme-1 )
Synthesis of compound 2
To a mixture of 2-chloro-5-nitro-4-thiocyanatopyrimidine (compound 1 ) (100 mg, 0.46 mmol) in DMF (50 ml) was added morpholine (60 mg, 0.69 mmol). After the mixture was stirred at rt for 1 h, water (20 ml ) was added and then the precipitate was collected by the filtration to afford the crude compound 2 (83 mg, yield 43%) as a yellow solid, used in the next step without further purification.
m/z: [M+H] + 267.9
Synthesis of compound 3
To a mixture of compound 2 (230 mg, 0.86 mmol) in HOAc (5 ml) was heated to 60 °C, and then iron powder (125 mg, 2.24 mmol) was added. After the mixture was stirred at the same temperature for 1 h, the mixture was filtered through celite. The filtrate was evaporated, and the residue was dissolved in EtOAc (50 ml). Insoluble materials was removed by filtration, the filtrate was neutralized with saturated NaHC03 solution andthen washed with brine dried over Na2S04, and concentrated to give crude compound 3 (170 mg, yield 83%), used in the next step without further purification,
m / z: [M+H] + 238.0
Synthesis of compound 5
To a mixture of compound 3 (170 mg, 0.72 mmol) in EtOH (10 ml) was added 2-bromo-1-(4- nitrophenyl) ethanone 4 (174 mg, 0.72 mmol). After the mixture was refluxed overnight, the reaction was cooled to room temperature, filtration of the resulting precipitate to afford crude compound 5 (66 mg, yield 24%) as a yellow solid, used in the next step without further purification.
m / z: [M+H] + 383.0
Synthesis of compound 6
A mixture of compound 5 (65 mg, 0.17 mmol) in HOAc (5 ml) was heated to 60 °C, and then iron powder (94 mg, 1.7 mmol) was added. After the mixture was stirred at the same temperature for 1 h, the mixture was filtered through celite. The filtrate was evaporated, the residue was neutralized with saturated NaHC03 solution, and extracted with EtOAc (20 ml x 3). The combined organic extracts were washed with brine, dried over Na2S04, filtered and concentrated to afford crude compound 6 (60 mg, yield 100%) as a red solid, used in the next step without further purification,
m / z: [M+H] + 353.1
The synthesis of compound 8-1
A mixture of compound 6 (20 mg, 0.056 mmol) and 5-tert-butyl-3-isocyanatoisoxazole 7(11 mg, 0.068 mmol) in toluene (5 ml) was stirred at 80 °C for 2h until the reaction finished (monitored by TLC). After cooled to room temperature, the reaction was then added with a mixture of DCM (10 ml), water (5 ml) and saturated NaHC03, The aqueous phase was extracted with DCM (10 ml x 2), and the combined organic extracts were dried over NaS04, filtered and concentrated to volume of about 2 ml and petroleum ether was added to formation of a solid. The precipitate was collected by filtration to afford crude compound 8-1 (7 mg, yield 24%) as a pink solid,
m / z: [M+H] + 519.0
H NMR (CDCI3): δ 9.32 (1 H, br), 8.50 (1 H, s), 7.83-7.79 (4H, m), 7.59-7.56 (2H, m), 5.92(1 H, s), 3.86 (4H, d, J = 4.8 Hz), 3.82 (4H, d, J = 4.0 Hz), 1.39 (9H, s).
Figure imgf000029_0001
Compound 8-2 was prepared according to the method of 8-1 in scheme-1 , as a pink solid, m / z: [M+H] + 477.0
Figure imgf000029_0006
Compound 8-3 was prepared according to the method of 8-1 in scheme-1 , as an off-white solid, m / z: [M+H] + 618.3
Figure imgf000029_0002
Compound 8-4 was prepared according to the method of 8-1 in scheme-1 , as an off-white solid saIt)
m / z: [M+H] + 518.2
Figure imgf000029_0003
Compound 8-5 was prepared according to the method of 8-1 in scheme-1 , as a yellow solid, m / z: [M+H] + 532.1
Figure imgf000029_0004
Compound 8-6 was prepared according to the method of 8-1 in scheme-1 , as a light yellow solid, m / z: [M+H] + 602.3
Example 7 (Compound 19 was made by the method in scheme-2)
Synthesis of compound 10 A mixture of compound 9 (3.0 g, 16.65 mmol) in aqueous KOH (5 N, 30 ml) was heated to reflux for 24h. After cooled to room temperature, the reaction solution was adjusted pH=6 by adding concentrated HCI. The precipitated solid was collected by filtration, and dried under vacuum to afford crude compound 10 (2.4 g, yield 93%) as a brown solid, used in next step without further purification.
Synthesis of compound 12
To a mixture of compound 10 (0.3 g, 1 .93 mmol) in toluene (4 ml) was added acetyl chloride (compound 11 ) (0.167 g, 2.13 mmol) dropwise in 15 min. After the mixture was heated at 80°C overnight, cooled to room temperature, the mixture was diluted with DCM (20 ml), and the adjusted pH=8 by saturated NaHC03. The organic phase was separated, washed with brine, dried over Na2S04, and concentrated to give the desired crude compound 12 (0.23 g, yield 66%) as yellow oil, used in next step without further purification.
HNMR (CDCI3): 57.82(11-1, d, J=8.8Hz), 7.28(1 H, d, J=2.4Hz), 7.04(1 H, dd,
Figure imgf000030_0001
J2=2.4Hz), 3.86(3H, s), 2.79(3H, s)
Synthesis of compound 13
A mixture of compound 12 (1.5 g, 8.37 mmol) and compound 4 (2-bromine-4'-nitrobenzene ketone)
(2.04 g, 8.37 mmol) in 5 ml of toluene was refluxed overnight. Filtration of resulted precipitate to afford the crude compound 13 (2.7 g, yield 76%) as a yellow solid, used in the next step without further purification.
Synthesis of compound 14
To a mixture of compound 13 (1.0 g, 2.36 mmol) in EtOH (10 ml), was slowly added TEA (262 mg, 2.6 mmol). After the mixture was heated to 105°C for 30 min, the mixture was cooled to room temperature. Filtrated of the resulted precipitate to afford crude compound 14 (0.76 g, yield 99%) as a yellow solid, used in the next step without further purification.
m / z: [M+H] + 325.0
Synthesis of compound 15
A mixture of compound 14 (300 mg, 0.31 mmol) in 33% HBr in HOAc (6 ml) in a sealed tube was stirred at 100 °C overnight (monitored by TLC). After the reaction was cooled to room temperature, the mixture was diluted with EtOAc (100 ml), filtered and the cake was dissolved in DCM (20 ml) and washed with saturated NaHC03 solution. After the aqueous phase was extracted with DCM (20 ml x 3), the combined organic phase was washed with brine, dried over Na2S04, filtered, concentrated to a volume about 5 ml, petroleum ether (10 ml) was added and an yellow solid was precipitated out, collected the solid by the filtration to afford crude compound 15 (220 mg, yield 76%) as a yellow solid, used in the next step without further purification,
m / z: [M+H] + 311.0
Synthesis of compound 17
To a mixture of compound 15 (50 mg, 0.16 mmol) in DMF (2 ml), was added K2C03 (33 mg, 0.24 mmol) and 4-(2-chloroethyl) morpholine 16 (48 mg, 0.32 mmol). After the mixture was heated to 60 °C for 4h, the mixture was partitioned between with DCM (20 ml) and water (10 ml), the aqueous phase was then extracted with DCM (10 ml x 2). The combined organic phase was washed with brine, dried over Na2S04, filtered and concentrated to give crude compound 17 (68 mg, yield 100%) as a yellow solid, used in the next step without further purification,
m / z: [M+H] + 424.0
Synthesis of compound 18
A mixture of compound 17 (68 mg, 0.16 mmol) in HOAc (3 ml) was heated to 60 °C, and then iron powder (89 mg, 1.61 mmol) was added. After the mixture was stirred at the same temperature for 1 h, the mixture was filtered through celite. The filtrate was evaporated and added in DCM (10 ml), then neutralized with aqueous saturated NaHC03 solution, and finally the aqueous phase was extracted with DCM (10 ml x 2). The combined organic phase was washed with brine, dried over Na2S04, filtered and concentrated to give crude compound 18 (47 mg, yield 74%), used in the next step without further purification.
m / z: [M+H] + 394.1
Synthesis of compound 19
To a mixture of compound 18 (47 mg, 0.12 mmol) in toluene (5 ml), was added compound 7 (5-tert-butyl-3-isocyanatoisoxazole) (24 mg, 0.14 mmol). After the mixture was stirred at 80 °C for 2h, the mixture was diluted with water (10 ml), and extracted with DCM (10 ml x 3). The combined organic phase was washed with saturated NaHC03 solution and brine, separately, dried over Na2S04, filtered and concentrated to afford crude product, which was purified by preparative TLC (5% MeOH in DCM), and then re crystallized from DCM and petroleum ether to give compound 19 (8 mg, yield 12%) as a yellow solid.
Figure imgf000031_0001
m / z: [M+H] + 560.2
H NMR (DMSO-d6): δ 9.52 (s, 1 H), 8.87 (s, 1 H), 8.19 (s, 1 H), 7.81-7.79 (d, J = 8.8 Hz, 1 H), 7.58-7.45(m, 5H), 7.07 (m, 1 H), 6.66 (s, 1 H), 6.52 (s, 1 H), 4.14 (t, J = 5.6 Hz, 2H), 3.59 (t, J = 4.4 Hz, 4H), 3.3 (m, 4H), 2.72 (t, J = 5.6 Hz, 2H), 1.30 (s, 9H).
Examples 8-22 (Compounds 28-1 to 28-15 were made by the method in scheme-3-A)
Synthesis of 2-methoxy-5-nitropyridine
A mixture of compound 20 (5.6 g, 35.3 mmol) in MeOH (50 ml) was added potassium tert-butoxide (5 g, 44.5 mmol). After the mixture was stirred at rt for 1 h, the mixture was poured into water (500 ml), filtration of resulted precipitate, air-dried to afford 2-methoxy-5-nitropyridine (5.04 g, yield 93%) as a white powder, used in the next step without further purification.
Synthesis of compound 22
A mixture of 2-methoxy-5-nitropyridine (5.0 g, 32.4 mmol) in HOAc (50 ml) was added iron powder (5 g, 89.53 mmol). After the mixture was heated to 60°C for 2h, the mixture was filtered through celite, and the filtered cake was washed with HOAc and then water. The filtrate was concentrate and the residue was neutralized with saturated NaHC03 solution, the aqueous phase was extracted with DCM (100 ml), and insoluble materials were removed by filtration. After the aqueous phase was extracted with DCM (100 ml x 4), the combined organic phase was washed with brine and concentrated to afford crude product, which was purified by silica gel column chromatography (2% MeOH in DCM) to afford compound 22 (3.5 g, yield 86%) as a brown oil
m / z: [M+H] +125.2
Synthesis of compound 23
A mixture of compound 22 (3.4 g, 27.39 mmol) and potassium thiocyanate (11 .7 g, 120.5 mmol) in HOAc (50 ml) at 0°C, bromine (2.35 ml in 25 ml of HOAc) was added from a dropping funnel at such a rate that the temperature never rose beyond 0°C. After all the bromine has been added (75 min), the mixture was stirred at rt overnight. The mixture was quenched by saturated Na2S03 solution, concentrated under vacuum and the residue was neutralized with saturated NaHC03 solution, and then DCM (100 ml) was added. Insoluble materials were removed by filtration, and the separated aqueous phase was extracted with DCM (100 ml x 4). The combined organic phase was washed with brine, dried over NaS04, filtered and concentrated to afford crude compound 23 (4.0 g, yield 80%) as a yellow solid, used in the next step without further purification. m / z: [M+H] + 182.2
Synthesis of compound 24
Compound 23 (0.5 g, 2.76 mmol) was dissolved in 33% HBr in HOAc (5 ml), the mixture was stirred at 130°C for 3h, and cooled to room temperature. The mixture was solidified by adding acetone (10 ml). The desired compound 24 (800 mg, yield 88%) was collected by filtration as a white solid, used in the next step without further purification,
m / z: [M+H] + 168.1
Synthesis of compound 25
A mixture of compound 24 (hydrobromide) (400 mg) in EtOH (10 ml) was added potassium carbonate (400 mg), the mixture was stirred at rt for 1 h. The solid was filtered off and the filtrate was concentrated to afford compound 24 in the form of a free base (205 mg, 1.23 mmol), which was then dissolved in EtOH (15ml). The resulted solution was added compound 4 (394 mg, 1.61 mmol). After the resulted mixture was refluxed overnight, filtration of the resulted precipitate to afford crude compound 25 (140 mg, yield 37%) as a yellow solid, used in the next step without further purification.
m / z: [M+H] + 313.1
Synthesis of compound 26
A mixture of compound 25 (50 mg, 0.16 mmol) in DMF (2 ml) was added potassium carbonate (44 mg, 0.32 mmol) and 4-(2-chloroethyl) morpholine (compound 16) (35 mg, 0.24 mmol). After the mixture was stirred at 60°C for 2h, the reaction mixture was poured into water (10 ml), the precipitated solid was collected by filtration, washed with water and methyl tert-butyl ether to give crude compound 26 (30 mg, yield 44%) as a yellow solid, used in the next step without further purification.
m / z: [M+H] + 426.2
Synthesis of compound 27
A mixture of compound 26 (30 mg, 0.07 mmol) in HOAc (2 ml) was heated to 60°C, and to this mixture was added iron powder (39 mg, 0.7mmol). After the reaction mixture was stirred at 60°C for 1 h, the mixture was filtered through celite, and the filtrate was concentrated under vacuum. The concentrated residue was neutralized with saturated NaHC03 solution, and the aqueous was extracted with 2% methanol in DCM (10 ml x 5), the combined organic extracts were washed with brine, dried over NaS04, filtered and concentrated to afford crude product, which was purified by preparative TLC (5% MeOH in DCM) to afford compound 27 (27 mg, yield 97%) as a yellow solid,
m / z: [M+H] + 396.2
Synthesis of compound 28-1
A mixture of compound 27 (27 mg, 0.068 mmol) and 5-tert-butyl-3-isocyanatoisoxazole 7 (12 mg, 0.075 mmol) in toluene (3 ml) was stirred at 80 °C for 2h until the reaction completed (monitored by TLC). After cooled to room temperature, the reaction was portioned between DCM (5 ml) and saturated NaHC03 solution (10 ml). The aqueous phase was then extracted with DCM (10 ml x 2), and the combined organic extracts were dried over NaS04, filtered and concentrated to volume of about 5 ml and then petroleum ether (5 ml) was added to the formation of a solid. The precipitate was collected by filtration to afford compound 28-1 (11 mg, yield 28%) as a yellow solid.
m / z: [M+H] + 562.2
H NMR (DMSO-d6): δ 9.72-9.65 (1 H, br), 9.25-9.19 (1 H, br), 8.63 (1 H, s), 8.32-8.30 (1 H, d, J = 8.8 Hz), 7.79-7.77(2H, d, J = 9.2 Hz ), 7.54-7.52 (2H, d, J = 8.4 Hz), 7.07-7.05 (1 H, d, J = 8.8 Hz), 6.53 (1 H, s), 4.44 (2H, t, J = 5.6 Hz), 3.57 (4H, t, J = 4.4 Hz), 3.3 (4H, m), 2.72 (2H, t, J = 6.0 Hz), 1.31 (9H, s).
Figure imgf000033_0001
Compound 28-2 was prepared according to the method of 28-1 in scheme 3-A as a yellow solid, m / z: [M+H] + 520.2
Figure imgf000033_0002
Compound 28-3 was prepared according to the method of 28-1 in scheme 3-A as a yellow solid, m / z: [M+H] + 561.2
Figure imgf000033_0003
Compound 28-4 was prepared according to the method of 28-1 in scheme 3-A as a yellow solid, m / z: [M+H] + 575.3
Figure imgf000033_0004
Compound 28-5 was prepared according to the method of 28-1 in scheme 3-A as an red solid, m / z: [M+H] + 617.1
Figure imgf000033_0005
Compound 28-6 was prepared according to the method of 28-1 in scheme 3-A as a white solid, m / z: [M+H] + 576.3
Figure imgf000033_0006
Compound 28-7 was prepared according to the method of 28-1 in scheme 3-A as a yellow solid, m / z: [M+H] + 573.3
Figure imgf000033_0007
Compound 28-8 was prepared according to the method of 28-1 in scheme 3-A as a white solid, m / z: [M+H] + 587.3
Figure imgf000034_0001
Compound 28-9 was prepared according to the method of 28-1 in scheme 3-A as a white solid, m/z: [M+H] + 546.3
Figure imgf000034_0002
Compound 28-10 was prepared according to the method of 28-1 in scheme 3-A as white solid, m/z: [M+H] + 560.3
Figure imgf000034_0003
Compound 28-11 was prepared according to the method of 28-1 in scheme 3-Aas white solid, m/z: [M+H] + 518.3
Figure imgf000034_0004
Compound 28-12 was prepared according to the method of 28-1 in scheme 3-A as white solid, m/z: [M+H] + 548.2
Figure imgf000034_0005
Compound 28-13 was prepared according to the method of 28-1 in scheme 3-A as white solid, m/z: [M+H] + 562.3
Figure imgf000034_0006
Compound 28-14 was prepared according to the method of 28-1 in scheme 3-Aas a off-white solid, m/z: [M+H] + 548.3
Figure imgf000034_0007
Compound 28-15 was prepared according to the method of 28-1 in scheme 3-Aas a off-white solid, m/z: [M+H] + 574.3
Figure imgf000035_0001
Examples 23-25 (Compounds 35-1 to 35-3 were made by the method in scheme-3-B)
Synthesis of compound 29
To a mixture of compound 20 (500 mg, 3.15 mmol) and compound 21 (907 mg, 4.10 mmol) in DMF (15 ml) was added K2C03 (871 mg, 6.31 mmol). The mixture was heated to 60°C and stirred for 48h. The mixture was diluted with DCM (150 ml), the organic phase was washed with water, brine, dried Na2S04separately filtered and concentrated to afford crude product ,which was purified by silica gel column chromatography (0-5% MeOH in DCM) to afford compound 29 (430 mg, yield 32%) as a yellow solid.
m / z: [M+H] + 344.1
Synthesis of compound 30
To a mixture of compound 29 (350 mg, 1.02 mmol) in HOAc (30 ml) was added iron powder (1.14 g, 20.39 mmol). The mixture was heated to 60°C and stired for 2h. The mixture was concentrate and the residue was neutralized with saturated NaHC03 solution, and extracted with DCM (100 ml). The organic layer was washed with brine, dried Na2S04. The solvent was removed under vacuum. The crude product was purified by preparative TLC(5% MeOH in DCM) to afford compound 30 (210 mg, yield 66%) as a white solid.
m / z: [M+H] + 314.3
Synthesis of compound 31
To a solution of compound 30 (53 mg, 0.169 mmol) in HOAc (2 ml) was added KSCN (73 mg, 0.761 mmol). After the solution was cooled to 0°C, then Br2 (40 mg, 0.253 mmol) in HOAc (1 ml) was added dropwise that the temperature never rose beyond 0°C. After all the bromine has been added (60 min), the mixture was stirred at rt overnight. The mixture was quenched by saturated Na2S03 solution, and concentrated. The residue was neutralized with saturated NaHS04 solution, and DCM (100 ml) was added. Insoluble materials were removed by filtration, and the separated aqueous phase was extracted with DCM (100 ml x 4). The combined organic phase was washed with brine, dried over NaS04, filtered and concentrated to afford crude product, which was purified by preparative TLC (5% MeOH in DCM) to afford compound 31 (34 mg, yield 54%) as a white solid.
m / z: [M+H] + 371.1
Synthesis of compound 32
To a mixture of compound 31 (34.0 mg, 0.091 mmol) in EtOH (3 ml) was added compound 4 (2-bromo -1 -(4-nitrophenyl) ethanone) (22.4 mg, 0.091 mmol). The resulted mixture was refluxed overnight. Filtrated of the resulted precipitate to afford crude compound 32 (12 mg, yeild 25%) as a yellow solid, used in the next step without further purification.
m / z: [M+H] + 516.1
Synthesis of compound 33
A mixture of compound 32 (20 mg, 0.038 mmol) and iron powder (43 mg, 0.775 mmol) in HOAc (5 ml) was heated to 60 °C and stired for 2h, mixture was then filtered through celite, the filtrate was concentrated. The residue was neutralized with saturated NaHC03 solution, extracted with DCM (10 ml x 5). The combined extracts were washed with brine, dried over NaS04, filtered and concentrated to afford crude product, which was purified by preparative TLC (5% MeOH in DCM) to afford compound 33 (8 mg, yield 42%) as a yellow solid. m / z: [M+H] + 486.2
Synthesis of compound 34
A mixture of compound 33 (8 mg, 0.016 mmol) and 5-tert-butyl-3-isocyanatoisoxazole 7 (3.2 mg, 0.019 mmol) in toluene (2 ml) was stirred at 80 °C for 2h. The reaction was added with DCM (5 ml), and the mixture was neutralized with saturated NaHC03 solution. The aqueous phase was extracted with DCM (10 ml x 2), and the combined organic extracts were dried over NaS04, filtered and concentrated to give the crude product which was purified by preparative TLC (7% MeOH in DCM) to afford compound 34 (7.0 mg, yield 65%) as a yellow solid.
m / z: [M+H] + 652.2
Synthesis of compound 35-1
To a mixture of compound 34 (4 mg, 6.1 umol) in AcOH (2 ml) was added 33% HBr in HOAc (1 ml), the mixture was stirred at rt. for 3h. The mixture was then diluted with DCM (30 ml) and water (20 ml), neutralized with saturated NaHC03, and extracted with DCM (10 ml x 2). The combined organic extracts were dried over NaS04, filtered and concentrated to give the crude product, and washed by DCM (3 ml) to afford pure compound 35-1 (1.5 mg, yield 47%) as a white solid.
m / z: [M+H] + 518.2
Figure imgf000036_0001
Synthesis of compound 35-2
The solution of compound 35-1 (15 mg, 0.029 mmol) in DCM (2 ml) was added triethyl amine (6 mg, 0.058 mmol), 4-N,N-dimethylaminopyridine (4 mg, 0.029 mmol), and acetic anhydride (6 mg, 0.058 mmol). The resulted reaction solution was stirred at RT for 2h, and the reaction was concentrated and purified by preparative TLC (7% MeOH in DCM) to afford compound 35-2 (7 mg, yield 43%), as a light yellow solid. m / z: [M+H] + 560.3
Figure imgf000036_0002
Synthesis of compound 35-3
The solution of compound 35-1 (15 mg, 0.029 mmol) in DCM (2 ml), was added triethyl amine (6 mg, 0.058 mmol), and methanesulfonyl chloride(4 mg, 0.032 mmol). The resulted reaction solution was stirred at rt for 2h, and the reaction was concentrated and purified by preparative TLC (7% MeOH in DCM) to afford compound 35-3 (1.2 mg, yield 6.9%), as a white solid.
m/z: [M+H] + 596.3
Figure imgf000036_0003
Example 26 ( Compound 35-4 was made by the method in scheme-3-B, through using (S)-(-)-1-Boc-2- pyrrolidinemethanol to replace intermediate 21 , as a light pink solid )
Figure imgf000037_0001
m / z: [M+H] + 532.3
Examples 27-30 (Compounds method in scheme-3-C)
Figure imgf000037_0002
Synthesis of compound 36-1
A mixture of compound 32 (50 mg, 0.096 mmol) in DMF (2 ml) was added 60% NaH (19.4 mg, 0.484 mmol). After stirred for 30min at rt, CH3I (68.8 mg, 0.484 mmol) was added to the mixture. The mixture was stirred at rt for 30min. The reaction mixture was diluted with DCM (30 ml), washed with water, brine, dried Na2S04, filtered and concentrated to afford crude product, which was washed by petroleum ether to afford crude compound 36-1 (50 mg, yield 97%) as a yellow solid, used in the next step without further purification.
m / z: [M+H] + 530.1
Synthesis of compound 37-1
A mixture of 36-1 (50 mg, 0.094 mmol) and iron powder (105 mg, 1 .890mmol) in HOAc (10 ml) was heated to 60 °C and stired for 2h. The mixture was filtered through celite, and the filtrate was concentrated. The residue was neutralized with aqueous saturated NaHC03 solution, and the aqueous was extracted with DCM (10 ml x 5). The combined organic extracts were washed with brine, dried over NaS04, filtered and concentrated to afford crude product, which was purified by preparative TLC (5% MeOH in DCM) to afford 37-1 (23 mg, yield 49%) as a yellow solid, used in the next step without further purification.
m / z: [M+H] + 500.2
Synthesis of compound 38-1
A mixture of compound 37-1 (23 mg, 0.046 mmol) and 5-tert-butyl-3-isocyanatoisoxazole 7 (9.1 mg, 0.055 mmol) in toluene (3 ml) and CHCI3 (1.5 ml) was stirred at 80 °C for 2h, and the reaction was diluted with DCM (5 ml), and the mixture was then neutralized with aqueous saturated NaHC03 solution. The aqueous phase was extracted with DCM (10 ml x 2), and the combined organic extracts were dried over NaS04, filtered and concentrated to give the crude product, which was purified by preparative TLC (7% MeOH in DCM) to afford compound 38-1 (19 mg, yeild 62%) as a white solid.
m / z: [M+H] + 666.2
Synthesis of compound 39-1
To a mixture of compound 38-1 (10.0 mg, 15 umol) in AcOH (2 ml) was added 33% HBr in HOAc (1 ml), the mixture was stirred at rt for 3h. The mixture was partitioned between DCM (30 ml) and water (20 ml), neutralized with saturated NaHC03, and the aqueous was extracted with DCM (10 ml x 2). The combined organic extracts were dried over NaS04, filtered and concentrated to give the crude product which was purified by preparative TLC (7% MeOH in DCM) to afford compound 39-1 (4.2 mg, yield 53%) as a white solid.
m / z: [M+H] + 532.3
HNMR(DMSO-d6): δ9.56(1 Η, s), 8.91 (1 H, s), 8.64 (1 H, s), 8.33-8.31 (1 H, d, J=8.8 Hz), 7.79-7.77(2H, d, J=8.8 Hz), 7.54-7.52 (2H, d, J=8.8 Hz), 7.09-7.07 (1 H, d, J=8.8 Hz), 6.53(1 H, s), 4.32 (2H, s), 2.35 (3H, s), 1 .31 (9H, s), 0.64 (4H, m)
Compound 39-2 was prepared according to the method of 39-1 in scheme 3-C as a off-white solid. m / z: [M+H] + 546.3
HNMR(DMSO-d6): δ9.55(1 Η, s), 8.93 (1 H, s), 8.64 (1 H, s), 8.33-8.31 (1 H, d, J=8.8 Hz), 7.79-7.77 (2H, d, J=8.8 Hz), 7.54-7.52 (2H, d, J=8.4 Hz), 7.09-7.07 (1 H, d, J=8.8 Hz), 6.52 (1 H, s), 4.32 (2H, s), 2.75-2.69(2H, q, J=7.2), 1.31 (9H, s), 1.00-0.96 (3H, t, J=7.2Hz), 0.65 (4H, s).
Compound 39-3 was prepared according to the method of 39-1 in scheme 3-C as a light yellow solid. m / z: [M+H] + 560.3
Synthesis of compound 39-4
A mixture of compound 39-1 (15 mg, 0.028 mmol) in MeOH (5 ml) was added 37% formaldehyde (0.1 ml) and one drop of TFA. After the mixture was stirred at rt. for 4h, NaBH3CN (18 mg, 0.293 mmol) was added to the reaction mixture, and stirred overnight at rt.. The solvent was removed under vacuum, and the residue was partitioned between DCM (50 ml) and water (20 ml). The separated aqueous phase was extracted with DCM (10 ml x 2), and the combined organic extracts were washed with brine, dried over NaS04, and concentrated to give the crude product which was purified by preparative TLC (7% MeOH in DCM) to afford compound 39-4 (11 mg, yield 71 %) as a white solid.
m / z: [M+H] + 546.2
Figure imgf000038_0001
Example 31 (Compound 46 was made by the method in scheme-3D)
Synthesis of compound 41
A mixture of compound 20 (5.0 g, 31.54 mmol) in DMF (50 ml), was slowly add morpholine 40 (6.87 g, 78.84 mmol). The mixture was stirred at rt for 1 h, and yellowish solid was precipitated out. The resulted suspension was poured in to water (100 ml) and the precipitate solid was collected by filtration, and washed with tret-Butyl methyl ether, air-dried to give the crude compound 41 (7.4 g, yield 100%) as a light yellow solid, used in the next step without further purification.
m / z: [M+H] + 210.1
Synthesis of compound 42
A mixture of compound 41 (4 g, 19.1 mmol) in HOAc (40 ml) was added iron powder (5.34 g, 89.53 mmol). The mixture was heated to 60°C and stirred for 2h, then the mixture was filtered through celite, the filted cake was washed with HOAc and then water. The filtrate was concentrated and the residue was neutralized with saturated NaHC03 solution, the aqueous was extracted with EtOAc (100 ml x 5) and DCM (100 ml x 2). The organic combined extracts were washed with brine, dried over NaS04, filtered and concentrated to afford crude compound 42 (2.7 g, yield 79%) as a red solid, used in the next step without further purification.
m / z: [M+H] + 180.3
Synthesis of compound 43
To a mixture of compound 42 (2.0 g, 11.2 mmol) in HOAc (20 ml) was added KSCN (4.7 g, 49.1 mmol), and the solution was cooled to 0°C. To this reaction solution was added Br2 (0.95 ml in 20 ml HOAc) by a dropping funnel at such a rate that the temperature never rose above 0°C. After all the bromine has been added (75 min), the mixture was stirred at rt overnight. The mixture was quenched by saturated Na2S03 solution, concentrated under vacuum and the residue was neutralized with saturated NaHC03 solution. The aqueous was added DCM (100 ml), and insoluble materials were removed by filtration. The separated aqueous phase was then extracted with DCM (100 ml x 2), and the combined organic extracts were washed with brine, dried over NaS04, filtered and concentrated under vacuum to afford crude product, which was purified by column chromatography on silica gel (2% -3% MeOH in DCM) to afford compound 43 (450 mg, yield 17%) as a yellow solid.
m / z: [M+H] + 237.1
Synthesis of compound 44
To a solution of compound 43 (200 mg, 0.85 mmol) in EtOH (5 ml), was added 2-bromo-1-(4- nitrophenyl) ethanone 4 (206 mg, 0.85 mmol), the reaction solution was refluxed overnight. The resulted precipitate was collected by filtration to afford crude compound 44 (144 mg, yield 45%) as a yellow solid, used in the next step without further purification.
Synthesis of compound 45
To a mixture of compound 44 (140 mg, 0.37 mmol) in HOAc (10 ml) heated to 60 °C was added iron powder (102 mg, 1 .84 mmol), and the mixture was stirred at the same temperature for 1 h. The mixture was filtered through celite, and the filtrate was evaporated and residue was neutralized with saturated NaHC03 solution, and the aqueous was extracted with DCM (20 ml x 3). The combined organic extracts were washed with brine, dried over Na2S04, filtered and concentrated to afford crude product, which was purified by silica gel column chromatography (2%-3% MeOH in DCM) to afford compound 45 (26 mg, yield 20%) as a yellow solid.
m / z: [M+H] + 352.2
Synthesis of compound 46
A mixture of compound 45 (10 mg, 0.028 mmol) and 5-tert-butyl-3-isocyanatoisoxazole 7 (9 mg, 0.056 mmol) in the mixed solvents of toluene (3 ml) and chloroform (3 ml) was stirred at 80°C for 2h until the reaction finished (monitored by TLC). The reaction was diluted with DCM (10 ml), and the mixture was neutralized with saturated NaHC03 solution. The aqueous phase was extracted with DCM (5 ml), and the combined organic extracts were dried over NaS04, filtered and concentrated to volume of about 5 ml and petroleum ether (5 ml) was added to formation of a solid. The precipitate was collected by filtration to afford crude compound 46 (10.8 mg, yield 73%) as an off-white solid.
m / z: [M+H] + 518.2
Figure imgf000039_0001
Examples 32-33 (Compounds 53-1 to 53-2 were made by the method in scheme-3-E)
Synthesis of compound 48
To a solution of compound 20 (50 mg, 0.315 mmol) and compound 47 (64 mg, 0.315 mmol) in acetonitrile (10 ml) was added DIPEA (122 mg, 0.946 mmol). The mixture was stirred at rt for 48h, and was diluted with DCM (20 ml), washed with water, brine, and dried Na2S04. The solvent was removed under vacuum, and the crude product was purified by silica gel column chromatography (100% DCM to 5% MeOH in DCM) to afford compound 48 (58 mg, yield 73%) as a yellow oil.
m / z: [M+H] + 253.2
Synthesis of compound 49
To a solution of compound 48 (200 mg, 0.792 mmol) in HOAc (10 ml) was added iron powder (442 mg, 7.93 mmol). After the mixture was heated to 60°C and stirred for 2h, the mixture was concentrate and the residue was neutralized with aqueous saturated NaHC03 solution, water extracted with DCM (100 ml). The organic phase was washed with brine, dried Na2S04, filtered and concentrated to give the crude product which was purified by preparative TLC (15% MeOH in DCM) to afford compound 49 (75 mg, yield 42%) as a yellow solid.
m / z: [M+H] + 223.3
Synthesis of compound 50
A solution of compound 49 (72 mg, 0.323 mmol) and KSCN (141 mg, 1.460 mmol) in HOAc (5 ml) was cooled to 0°C, then Br2 (77 mg, 0.485 mmol) in HOAc (1 ml) was added dropwise that the temperature never rose beyond 0°C. After all the bromine has been added (60 min), the mixture was stirred at rt overnight, and the reaction was quenched by saturated Na2S03 solution. The reaction mixture was concentrated and the residue was neutralized with aqueous saturated NaHC03 solution, and diluted with DCM (100 ml). Insoluble materials were removed by filtration, and the separated aqueous phase was extracted with DCM (50 ml x 4). The combined organic extracts were washed with brine, dried over NaS04, filtered and concentrated to afford crude product which was purified by preparative TLC (5% MeOH in DCM) to afford compound 50 (26 mg, yield 29%) as a brown solid,
m / z: [M+H] + 280.2
Synthesis of compound 51
To a mixture of compound 50 (26 mg, 0.093 mmol) in EtOH (5 ml) was added 2-bromo-1-(4- nitrophenyl) ethanone 4 (22.7 mg, 0.093 mmol). The resulted mixture was refluxed overnight, and the resulted precipitate was collected by filtration to afford crude compound 51 (23 mg, yield 58%) as a yellow solid, used in the next step without further purification.
m/z: [M+H] + 425.1
Synthesis of compound 52
A mixture of compound 51 (30 mg, 0.070 mmol) and iron powder (78 mg, 1 .410 mmol) in HOAc (5 ml) was heated to 60 °C for 2h. Then the mixture was filtered through celite, and the filtrate was concentrated. The residue was neutralized with saturated NaHC03 solution, extracted with DCM (10 ml x 5), and the combined organic extracts were washed with brine, dried over NaS04, filtered and concentrated to afford crude product, which was purified by preparative TLC (7% MeOH in DCM) to afford compound 52 (26 mg, yield 93%) as a yellow solid.
m / z: [M+H] + 395.2
Synthesis of compound 53-1
A mixture of compound 52 (26 mg, 0.065 mmol) and 5-tert-butyl-3-isocyanatoisoxazole 7 (13 mg, 0.079 mmol) in toluene (3 ml) and CCI4 (1.5 ml) was stirred at 80 °C for 1 h. The reaction was concentrated to give the crude product, which was purified by preparative TLC (7% MeOH in DCM) to afford compound 53-1 (12 mg, yield 32%) as a yellow solid.
Figure imgf000040_0001
m / z: [M+H] + 561.2
HNMR(DMSO-d6) 59.54(1 H, s), 8.87 (1 H, s), 8.50 (1 H, s), 7.98-7.96 (1 H, d, J=4.8 Hz), 7.77-7.75(2H, d, J=8.8 Hz), 7.52-7.49 (2H, d, J=8.8 Hz), 6.96(1 H, t, J=5.6Hz), 6.70-6.68 (1 H, d, J=8.8 Hz), 6.52 (1 H, s), 3.59 (4H, t, J=4.4Hz), 3.43-3.36 (4H, m), 2.43 (4H, br), 1.30 (9H, s)
Synthesis of compound 53-2 To a mixture of compound 53-1 (5 mg, 0.008 mmol) in MeOH (3 ml) was added 37% formaldehyde (0.04 ml) and one drop of TFA, and the mixture was stirred at rt for 4h, then NaBH3CN (28 mg, 0.445 mmol) was added. After stirred overnight, the solvent was removed under vacuum, and the mixture was diluted with DCM (20 ml), washed with water, brine, and dried over Na2S04. The solvent was removed under vacuum, and the crude product was purified by preparative TLC (7% MeOH in DCM) to afford compound 53-2 (3.5 mg, yield 68%) as a white solid.
Figure imgf000041_0001
m / z: [M+H] + 575.3
Examples 34-35 (Compounds 63-1 to 63-2 were made by the method in scheme-3F)
Synthesis of compound 54
To a stirring ice-cooled solution of compound 21 (500 mg, 2.26 mmol) and triethylamine (457 mg, 4.52 mmol) in DCM (10 ml), was added methanesulfonyl chloride (310 mg, 2.71 mmol) dropwise. The mixture was stirred 0.5h at 0°C, then the mixture was washed by water, and brine, the organic phase was dried over Na2S04, filtered and concentrated to give the crude compound 54 (650 mg, yield 96%) as a white solid, used in the next step without further purification.
Synthesis of compound 55
To a suspension of isoindoline-1 , 3-dione (540 mg, 3.67 mmol) and potassium carbonate (304 mg, 2.2 mmol) in DMSO (10 ml) at 100°C, was added compound 54 (550 mg, 1.84 mmol, dissolved in DMSO (2 ml). After the mixture was stirred 0.5h at 100°C, the reaction mixture was cooled down, and diluted with EtOAc (50 ml). The reaction mixture was washed by water and brine, dried over Na2S04, concentrated to give the crude compound 55 (620 mg, yield 96%) as a white solid, used in the next step without further purification.
Synthesis of compound 56
A stirring solution of compound 55 (720 mg, 2.05 mmol) and hydrazine hydrate (0.5ml in EtOH (20 ml) was refluxed for 4h. The insoluble material was filtered off and the filtrate was concentrated to give the residue which was dissolved in DCM. After insoluble material was removed by filtration, the filtrate was concentrated to give crude compound 56 (410 mg, yield 90%) as a colorless oil, used in the next step without further purification,
m / z: [M+H]+ 221.3
Synthesis of compound 57
To a solution of compound 20 (316 mg, 2.00 mmol) and compound 56 (400 mg, 1.82 mmol) in acetonitrile (20 ml), was added DIPEA (469 mg, 3.63 mmol). The resulted solution was stirred 48h at room temperature. The reaction mixture was portioned between DCM and water (20 ml, each), then the organic phase was washed by water, brine, dried over Na2S04, filtered, and concentrated to give crude product, which was purified by silica gel column chromatography (0-5% MeOH in DCM) to afford compound 57 (300 mg, yield 48%) as a yellow solid,
m / z: [M+H]+ 343.3
The synthesis of compound 58
To the solution of compound 57 (300 mg, 0.876 mmol) in HOAc (15 ml), was added iron powder (244 mg, 4.38 mmol). The resulted mixture was stirred at 60°C for 1 h. The mixture was concentrated and the residue was neutralized with saturated NaHC03 solution, the aqueous phase was extracted with DCM (100 ml), and the combined organic phase was washed with water and brine, dried over Na2S04, filtered and concentrated to afford crude compound 58 (250 mg, yield 91 %) as a brown solid, used in the next step without further purification,
m / z: [M+H]+ 313.2
Synthesis of compound 59
To an ice-cooling stirring mixture of compound 58 (250 mg, 0.80 mmol) and KSCN (350 mg, 3.60 mmol) in HOAc (10 ml), was added bromine (191 mg, 1.2 mmol, dissolved in 2 ml HOAc) dropwise over 60min. After the resulted mixture was stirred at room temperature for 60min, the reaction mixture was quenched by saturated Na2S03 solution and neutralized with NaHC03solution, then the mixture was diluted by DCM (100 ml), and insoluble impurity was filtered off. The separated aqueous phase was extracted with DCM (50 ml x 4), and the combined organic phase was washed with brine, dried over Na2S04, filtered and concentrated to afford crude product, which was purified by silica gel column chromatography (0-5% MeOH in DCM) to afford compound 59 (26 mg, yield 29%) as a brown solid,
m / z: [M+H]+ 370.3
Synthesis of compound 60
To a solution of compound 59 (75 mg, 0.093 mmol) in EtOH (5 ml), was added 2-bromo-1-(4- nitrophenyl)ethanone 4 (22.7 mg, 0.093 mmol). After the resulted reaction solution was refluxed overnight, the reaction mixture was concentrated, and the residue was purified by preparative TLC (7% MeOH in DCM) to afford compound 60 (10 mg, yield 10%) as a yellow solid.
m / z: [M+H]+ 515.2
Synthesis of compound 61
To a stirring solution of compound 60 (10 mg, 0.019 mmol) in HOAc (5 ml), was added iron powder (10 mg, 0.194 mmol). After the resulted mixture was stirred 1 h at 60°C, the reaction mixture was cooled down to room temperature, and filtered through celite. Then the filtrate was concentrated, and the residue was neutralized with saturated NaHC03 solution, the aqueous phase was extracted with DCM (10 ml x 5), and the combined organic phase was washed with brine, dried over Na2S04, filtered and concentrated to afford crude compound 61 (9.0 mg, yield 96%) as a yellow solid, used in the next step without further purification.
m / z: [M+H]+ 485.3
Synthesis of compound 62
To a stirring solution of compound 61 (9.0 mg, 0.018 mmol) in toluene (3 ml) and chloroform (1.5 ml), was added 5-tert-butyl-3-isocyanatoisoxazole (3 mg, 0.018 mmol). After the mixture was stirred 1 h at 80°C, the mixture was concentrated and the residue was purified by preparative TLC (7% MeOH in DCM) to afford compound 62 (7 mg, yield 58%) as a yellow solid.
m / z:[M+H]+ 651.3
Synthesis of compound 63-1
To a stirring solution of compound 62 (7 mg, 10 umol) in HOAc (3 ml), was added 33%HBr-HOAc solution (1 ml). After the resulted mixture was stirred 1 h at room temperature, the reaction solution was partitioned between DCM (30 ml) and water (20 ml). The aqueous phase was neutralized with saturated NaHC03 solution, and extracted with DCM (10ml x 2). The combined organic phase was washed with brine, dried over Na2S04, filtered and concentrated to afford crude product, which was purified by preparative TLC (15% MeOH in DCM) to afford compound 63-1 (1.2 mg, yield 21 %) as a white solid.
Figure imgf000043_0001
m / z: [M+H]+ 517.2
Synthesis of compound 63-2
Compound 63-2 was prepared in the same method as compound 63-1 in scheme-3-F by using the intermediate 69 instead of intermediate 56. it is a light yellow solid.
Figure imgf000043_0002
Synthesis of compound 65
To an ice-cooling stirring solution of compound 64 ( 3.0 g, 11.4 mmol ) in DMF (20 ml), was added NaH (0.68 g 60% 17.09 mmol) in small portion, then added Mel (0.85 ml, 13.6 mmol) dropwise. After the resulted mixture was stirred 1 h at room temperature (monitored by TLC), the mixture was diluted with EtOAc (150 ml), washed with water (50 ml x 4), brine and dried over Na2S04, filtered and concentrated to afford crude compound 65 (3.5 g, yield 100%) as a colorless oil, used in the next step without further purification.
Synthesis of compound 66
To a solution of compound 65 (3.0 g, 10.8 mmol) in THF (30 ml), was added NaBH4 (4.09 g, 0.18 mol). The resulted mixture was refluxed 0.5h, then methanol (20 ml) was added dropwise at reflux, and the reaction was stirred at reflux for another 3h. The reaction was concentrated to dryness under vacuum, water phase was extracted in EtOAc (50 ml), organic phase was washed with water (20 ml) and brine successively, dried over Na2S04, filtered and concentrated to afford crude compound 66 (2.5 g, yield 98%) as a colorless oil, used in the next step without further purification.
Synthesis of compound 67
To an ice-cooling solution of compound 66 (2.2 g, 9.35 mmol) and triethylamine (1.42 g, 14.03 mmol) in DCM (20 ml), was added Methanesulfonyl chloride (1.29 g, 11 .2 mmol, dissolved in 5 ml DCM)dropwise, then resulted mixture was stirred 1 h at room temperature. The reaction solution was diluted with DCM (30 ml), washed with water (15 ml x 3) and brine, dried over Na2S04, filtered and concentrated to afford crude compound 67 (2.9 g, yield 99%) as a colorless oil, used in the next step without further purification.
Synthesis of compound 68
To a suspension of lsoindoline-1 , 3-dione (4.23 g, 28.7 mmol) and potassium carbonate (4.63 g, 33.5 mmol) in DMSO (20 ml) at 100°C, was added compound 67 (3.0 g, 9.57 mmol, dissolved in DMSO (5 ml) dropwise. The reaction mixture was stirred for another 2h at this temperature, then the reaction mixture cooled to room temperature and diluted with EtOAc (100 ml), washed with water (20 ml x 4), brine, dried over Na2S04, filtered and concentrated to give crude product, which was purified by preparative TLC (petroleum ether: EtOAc = 5:1 ) to afford compound 68 (0.90 g, yield 26%) as a white solid, used in the next step without further purification.
Synthesis of compound 69
A stirring solution of compound 68 (0.9 g, 2.47 mmol) and hydrazine hydrate (0.5 ml) in EtOH (10 ml) was refluxed for 2h. The insoluble material was filtered off and the filtrate was concentrated, and then the residue was dissolved in DCM, after insoluble material was removed by filtration, the filtrate was concentrated to give crude compound 69 (550 mg, yield 95%), used in the next step without further purification.
m / z: [M+H]+ 235.3
Example 36 (Compound 80 was made by the method in scheme-3-G)
Synthesis of compound 71
To a stirring solution of compound 70 (55.8 g , 300 mmol) in 95% EtOH (200 ml), was added aqueous NaOH (12 g, 300 mmol) (50 ml) dropwise. The reaction mixture was stirred overnight at room temperature, decompression to remove EtOH, water (300 ml) was added and the stirred. After the aqueous phase was washed with EtOAc (50 ml x 2), the aqueous phase was acidified to pH 1.5 with aqueous 10% HCI and the aqueous phase was extracted with EtOAc (100 ml x 3). The combined organic phase was dried over Na2S04, filtered, and concentrated to give crude compound 71 (35 g, yield 73%) as a colorless oil, used directly in the next step without further purification.
Synthesis of compound 72
To a stirring solution of compound 71 (1 .0 g, 6.32 mmol) and triethylamine (0.97 ml, 7.59 mmol) in THF (18 ml) at -10 °C, was added isobutyl chloroformate (0.90 ml, 6.96 mmol) dropwise. Then the reaction mixture was stirred 1 h at 0°C, the insoluble material was filtered off, and the filtrate was directly used later. To an ice-cooling solution of NaBH4 (0.71 g, 18.97 mmol) in THF (10 ml) and water (2.5 ml), was added the filtrate obtained above dropwise. After the reaction mixture was allowed to stirred at 0°C for 1 h, then reaction mixture was poured into 10% of aqueous solution HOAc, the aqueous phase was extracted with EtOAc (30 ml x 3), and the combined organic phase was washed with brine, dried over Na2S04, filtered and concentrated to afford crude product, which was purified by silica gel column chromatography (Petroleum ether: ethyl acetate = 9: 1 to 2:1 ) to give compound 72 (0.65 g, yield 61 %) as a colorless oil.
HNMR (CDCI3): δ 4.15(2H, q, J=7.0Hz), 3.62(2H, s), 1.28-1.23(5H, m), 0.86(2H, q, J=4.2Hz) Synthesis of compound 73
To a stirring ice-cooling solution of compound 72 (200 mg, 1.39 mmol) and triethylamine (280 mg,
2.77 mmol) in DCM (10 ml), was added methanesulfonyl chloride (190 mg, 1 .66 mmol, dissolve in 1 ml DCM) dropwise. The reaction mixture was stirred 1 h at 0°C, then diluted with DCM (20ml), the organic phase was washed with water and brine, dried over Na2S04, filtered and concentrated to afford crude compound 73 (300 mg, yield 97%) as a colorless oil, used directly in the next step without further purification.
Synthesis of compound 74
To a stirring solution of compound 15 (5 mg, 0.016 mmol) in DMF (1 ml), was added cesium carbonate (5 mg, 0.016 mmol) and compound 73 ( 5 mg, 0.016 mmol). The resulted mixture was stirred 2h at 130°C, and the reaction mixture was cooled down to room temperature, and diluted with DCM (20 ml) and water (10 ml), the water phase was extracted with DCM (10 ml x 2)), the organic phase was washed with water, brine, dried over Na2S04, filtered and concentrated to afford crude product which was purified by preparative TLC (DCM) to afford compound 74 (4 mg, yield 56%) as a yellow solid.
m / z: [M+H] + 437.2
Synthesis of compound 75
To a stirring solution of compound 74 (10 mg, 0.022 mmol) in a mixed solvent of THF (1 ml), EtOH (0.5 ml) and water (0.25 ml), was added sodium hydroxide (18 mg, 0.22 mmol). The resulted mixture was stirred for 4h at room temperature, and then the mixture was neutralized with aqueous HCI (2 N), pH=7, the solvent was concentrated, the aqueous phase was extracted with DCM (10 ml χ 3), and the combined organic phase was washed with brine, dried over Na2S04, filtered and concentrated to afford the crude compound 75 (27 mg, yield 97%) as a yellow solid, used directly in the next step without further purification.
m / z: [M+H] + 409.3
Synthesis of compound 76
To a stirring solution of compound 75 (40 mg, 0.097 mmol), 4-methoxybenzyl alcohol (40 mg , 0.293 mmol) in toluene (3 ml), was added triethylamine (29 mg , 0.293 mmol) and DPPA (53 mg, 0.195 mmol). The resulted mixture was stirred 0.5h at room temperature, then refluxed for 3h. The reaction mixture was concentrated and the residue was purified by preparative TLC (petroleum ether: ethyl acetate = 2:1 ) to give compound 76 (35 mg, yield 65%) as a yellow solid,
m / z: [M+H] + 544.2
Synthesis of compound 77
To a solution of compound 76 (35 mg, 0.064 mmol ) in DMF (2 ml), was added sodium hydride(60%, 7 mg, 0.321 mmol). The resulted mixture was stirred 30min at room temperature, then Mel (45 mg, 0.321 mmol) was added, and stirred for a another 30min. The mixture was diluted with DCM (30 ml), washed with water, brine and dried over Na2S04, filtered and concentrated to afford crude product, which was purified by preparative TLC (petroleum ether : ethyl acetate = 2: 1 ) to afford compound 77 (30 mg, yield 83%) as a yellow solid,
m / z: [M+H] + 558.2
Synthesis of compound 78
To a stirring solution of compound 77 (30 mg, 0.053 mmol) in HOAc (2 ml), was added iron powder (60 mg, 1.08 mmol). After the reaction mixture was stirred 1 h at 60°C, the mixture was filtered through celite, and the filtrate was concentrated to dryness. The residue was neutralized with saturated sodium bicarbonate solution, and extracted with DCM (10 ml x 2). The combined organic phase was washed with brine, dried over Na2S04, filtered and concentrated to afford crude compound 78 (25 mg, yield 88%), used in the next step without further purification.
Synthesis of compound 79
To a solution of compound 78 (25 mg, 0.047 mmol) in a mixed solvent of toluene (1.5 ml) and chloroform (0.5 ml), was added 5-ferf-butyl-3-isocyanatoisoxazole (10 mg, 0.056 mmol). The resulted solution was stirred 2h at 80°C, then diluted with water (10ml), extracted with DCM (10 ml x 3). The combined organic phase was washed with brine, dried over Na2S04, filtered, concentrated to give the crude compound, which was purified by preparative TLC (5% MeOH in DCM) to afford desired compound 79 (8 mg, yield 24%) as a yellow solid,
m/z: [M+H] + 694.3
Synthesis of compound 80
A solution of compound 79 (8 mg, 0.011 mmol) and trifluoroacetic acid (0.3 ml) in DCM (3 ml) was stirred 2hr at room temperature. The reaction solution was concentrated and the residue was dissolved in DCM (10 ml), washed with saturated sodium bicarbonate solution, brine, dried over Na2S04, filtered and concentrated to afford the crude product, which was purified by preparative TLC (10 % MeOH in DCM) to afford compound 80 (2.4 mg, yield 39%) as a white solid.
Figure imgf000046_0001
m / z: [M+H] + 530.3
Example 37 (Compound 83 was made by the method in scheme-3H)
Synthesis of compound 81
To a solution of compound 76 (25 mg, 0.045 mmol) in HOAc (2 ml), was added iron powder (51 mg, 0.919 mmol). After the mixture was stirred 1 h at 60°C, then the mixture was cooled down to room temperature, the solid was filtered off through celite, and the filtrate was concentrated. The residue was neutralized by aqueous saturated sodium bicarbonate, and the aqueous was extracted with DCM (10 ml x 2). The combined organic phase was washed with brine, dried over Na2S04, filtered and concentrated to afford crude compound 81 (20 mg, yield 84%), used directly in next step without further purification.
Synthesis of compound 82
To a solution of compound 81 (20 mg, 0.038 mmol) in a mixed solvent of toluene (1.5 ml) and chloroform (0.5 ml), was added 5-ferf-butyl-3-isocyanatoisoxazole 7 (8 mg, 0.046 mmol). After the resulted mixture was stirred 2hr at 80°C, the reaction mixture was diluted by water (10 ml), and the aqueous was extracted with DCM (10 ml x 3). The combined organic phase was washed with brine, dried over Na2S04, filtered, concentrated to give the crude product, which was purified by preparative TLC (5% MeOH in DCM ) to give the desired compound 82 (9 mg, yield 34%) as a yellow solid.
m / z: [M+H] + 680.3
Synthesis of compound 83
A solution of compound 82 (9 mg, 0.013 mmol) and trifluoroacetic acid (0.2 ml) in DCM (2 ml) was stirred at room temperature for 0.5h. The mixture was concentrated and the residue was dissolved in DCM (10 ml), washed with aqueous saturated sodium bicarbonate, brine, dried over Na2S04, filtered and concentrated to afford crude product, which was purified by preparative TLC (10% MeOH in DCM) to afford compound 83 (1.2 mg, yield 17%) as a white solid.
m / z: [M+H] + 516.3
Examples 38-41 (Compounds 89-1 , 89-3, 89-4, and 89-5 were made by the method in scheme-3-l)
Figure imgf000046_0002
Figure imgf000046_0003
Synthesis of compound 84
To a stirring solution of compound 20 (5.0 g, 31.5 mmol) in acetic acid (50 ml) was added iron powder (8.8 g, 157.6 mmol) in small portion at room temperature, the reaction was exothermic and the temperature rise to 80°C, end of added and kept the temperature at 40-50°C and stirred for 2h. The reaction was filtered through celite and washed with little acetic acid, the filtrate was evaporated to dryness, adjusted pH=8 with saturated solution of sodium bicarbonate, extracted with dichloromethane (100 ml x 5). The combined organic phase was washed with brine, dried over Na2S04, filtered and concentrated to afford crude compound 84 (3.9 g, yield 96%) as a brown solid, used directly in next step without further purification.
Synthesis of compound 85
Compound 84 (2.5 g, 19.4 mmol) and potassium thiocyanate (8.5 g, 37.5 mmol) were dissolved in acetic acid (25 ml). To this solution was added dropwise a solution of bromine (1.5 ml, 29.2 mmol in acetic acid 10 ml) for about 60 min, the resulted mixture was stirred at room temperature for overnight, then water (20 ml) was added. The reaction mixture was stirred and filtered at 85°C, the filtered cake was washed by acetic acid. The obtained filtrate was concentrated to 1/3 volume, neutralized with aqueous ammonia solution to pH=6, the precipitate was collected by filtration to afford crude compound 85 (2.5 g, yield 69%) as a yellow solid, used directly in next step without further purification.
Synthesis of compound 86
To a stirring solution of compound 85 (1.4 g, 7.5 mmol) in n-butanol (15 ml) was added 2-bromo-1-(4- nitrophenyl)ethanone 4 (2.02 g, 8.3 mmol) and sodium bicarbonate (0.63 g, 7.5 mol), the resulted mixture was stirred at refluxing for 4h, then the reaction was cooled down to room temperature, filtered to afford the crude compound 86 (1.06 g, yield 42%) as a yellow solid, used directly in next step without further purification.
m / z: [M+H] + 331.1
Synthesis of compound 87
To a suspension of compound 86 (1.0 g, 3.02 mmol) in acetic acid (200 ml) was added iron powder (0.88 g, 15.8 mmol) in one portion at room temperature, the resulted mixture was stirred at 60°C for 5h. The reaction was filtered through celite. The filtrate was concentrated and neutralized with sodium bicarbonate, the water phase was extracted with DCM (100 ml x 3) which include little MeOH, The combined organic phase was washed with brine and dried over Na2S04, filtered and concentrated to afford crude compound 87 (0.26 g, yield 28%) as a brown solid, used directly in next step without further purification.
m / z: [M+H] + 301.2
Synthesis of compound 88-1
A solution of compound 87 (80 mg, 0.26 mmol) in N-i ,N-|-dimethylethane-1 ,2-diamine (5 ml) was stirred at 125°C for 3 days, then the mixture was evaporated to dryness, the residues was dissolved in ethyl acetate (100 ml) and washed sequentially with a saturated solution of ammonium chloride and brine. The organic phase was dried over Na2S04, filtered and concentrated. The residue was purified by preparative TLC (6% methanol in dichloromethane) to afford compound 88-1 (60 mg, yield 64%) as a white solid.
m / z: [M+H] + 353.3
Synthesis of compound 88-2
A mixture of compound 87 (40 mg, 0.13 mmol) in DMSO (1 ml) and DMF (0.5 ml), potassium carbonate (90 mg, 0.66 mol) and (1 S,4S)-2-oxa-5-azabicyclo[2.2.1]heptane (54 mg, 0.40 mmol) was added, stirred at 125°C for 5 days. The reaction was cooled down to room temperature, diluted with ethyl acetate, washed with water (20 ml x 3) and brine,, dried over Na2S04, filtered and concentrated, the residue was purified by preparative TLC (5% methanol in dichloromethane) to afford compound 88-2 (20 mg, yield 41 %) as an off-white solid.
m / z: [M+H] + 364.3
Synthesis of compound 89-1
A solution of compound 88-1 (60 mg, 0.17 mmol) and 5-(tert-butyl)-3-isocyanatoisoxazole 7 (31 mg, 0.18 mmol) in a mixted solvent of toluene (5 ml) and chloroform (1 ml) was stirred at 80°C for 2h. The reaction mixture was filtered, the filtered cake was recrystallized from dichloromethane/toluene to afford compound 89-1 (45 mg, yield 51 %) as a white solid.
Examples 42-44 (Compounds 97-1 to 97-3 were made by the method in scheme 4-A)
Figure imgf000048_0001
Figure imgf000048_0002
Synthesis of compound 91
To an ice-cooling solution of 2-chloro-5-nitro-4-thiocyanatopyrimidine (compound 1 ) (520 mg, 2.4 mmol) in toluene (10 ml) was added a solution of 2,2-dimethoxyethanamine (compound 90) (378 mg, 3.6 mmol) in ethanol (1 ml), the resulted mixture was stirred at room temperature for 1 h, then the mixture was filtered to afford compound 91 (650 mg, yield 95%) as a white solid, used directly in next step without further purification.
Synthesis of compound 92
To a solution of compound 91 (630 mg, 2.21 mmol) in acetic acid (10 ml) was added iron powder (616 mg, 11.04 mmol) in one portion at room temperature, the resulted mixture was stirred at 60°C for 1 h. The mixture was filtered through celite, the filtrate was evaporated to dryness, the residue was dissolved in dichloromethane contained 20% methanol (50 ml), adjusted pH=8 with saturated solution of sodium bicarbonate, the insoluble residue was filtered off. The filtrate was washed with sodium bicarbonate solution, brine, dried over Na2S04, filtered and concentrated to afford crude compound 92 (350 mg, yield 62%) as a deep brown solid, used directly in next step without further purification,
m / z: [M+H] + 256.1
Synthesis of compound 93
A mixture of compound 92 (150 mg, 0.58 mmol) and 2-bromo-1-(4-nitrophenyl) ethanone 4 (143 mg, 0.58 mmol) in ethanol (5 ml) was stirred at 60°C for overnight, refluxed at 100 °C for 3h, then the reaction mixture was cooled down to room temperature, filtered to afford crude compound 93 (120 mg, yield 51 %) as a yellow solid, used directly in next step without further purification.
m / z: [M+H] + 401.1
Synthesis of compound 94 To a stirring solution of compound 93 (500 mg, 1 .25 mmol) in acetic acid (20 ml) was added zinc powder (408 mg, 6.24 mmol) at room temperature, the resulted mixture was stirred at 60°C for 1 h. The mixture was filtered through celite, the filtrate was evaporated to dryness, neutralized with saturated solution of sodium bicarbonate, then the aqueous phase was extracted with dichloromethane contained 10% methanol (100 ml). The organic phase was washed with brine, dried over Na2S04, filtered and concentrated to afford crude compound 94 (150 mg, yield 32%) as a brown solid, used directly in next step without further purification.
Synthesis of compound 95
A mixture of compound 94 (50 mg, 0.13 mmoDand 5-(tert-butyl)-3-isocyanatoisoxazole 7 (24 mg, 0.14 mol ) in a mixed solvent of toluene (5 ml), chloroform (1 ml) and 1 ,4-dioxane (1 .5 ml) was stirred at 80°C for 2h. The mixture was concentrated, then the residue was purified by preparative TLC (7% methanol in dichloromethane) to afford compound 95 (50 mg, yield 51 %) as a yellow solid,
m / z: [M+H] + 537.3
Synthesis of compound 97-1
Compound 95 (15 mg, 0.03 mmol), dimethylamine hydrochloride (25 mg, 0.30 mmol) and zinc(ll) chloride (20 mg) were dissolved in a mixed solvent of methanol (2.0 ml) and dichloromethane (1 ml), the resulted mixture was stirred at room temperature for 2h, then sodium cyanoborohydride (20 mg, 0.30mmol) was added to the solution and stirred for overnight. The reaction was added water (5 ml), and the reaction mixture was extracted with dichloromethane contained 20% methanol (5 ml x 3). The combined organic phase was dried over Na2S04, filtered and concentrated. The residue was purified by preparative TLC (15% methanol in dichloromethane) to afford compound 97-1 (6 mg, yield 37%) as a yellow solid,
m / z: [M+H] + 520.3
Compound 97-2 and 97-3 were prepared according to the method of 97-1 in scheme 4-A and the results were listed in the above table.
The biological assays:
The following representative assays (but not limited to) were performed in assesing the biological activities disclosed herein.
1. MV4-11 cell proliferation assay
The effect of test compounds on cancer cell viability was tested in MV4-11 cell (ATCC, CRL-9591 ), a human leukemia cell line expressing constitutive active FLT3 receptor and contains internal tandem duplications (ITD) found in the AML patients. MV4-11 cells were plated in 96 well plates at 15,000 cells per well in 100 ul IMDM medium (Invitrogen, 12440-053) containing 10% fetal bovine serum. Test compounds were prepared in 100% DMSO and added to the cells to achieve final concentrations from 0.2 uM to 0.000001 uM (10 concentration points in 3 fold serial dilution). The culture plates were then incubated at 37C in 5% C02 for 72 hours.
After 72 hours, the cell morphology was observed under an inverted microscope. Cell viability was then quantified at room temp using a CellTiter-Glo assay (Promega G7571 ) following the manufacture's instruction. Briefly, a volume of 10Oul Celltiter-Glo reagenet was added to each well to induce cell lysis. After 10 min incubation at RT, a luminescent signal was produced by measuring the amount of ATP. The signal was directly proportional to the number of viable cells present in culture.
The IC50 values of compounds of the present invention in inhibiting MV4-11 cell proliferation are listed in following table 1 . Table 1
Figure imgf000050_0001
FLT3 is a receptor tyrosine kinase involved in survival and proliferation of leukemic cells. Constitutively activating FLT3 mutations has been found in about 30% of all patients with acute myeloid leukemia (AML). The tested compounds were screened for their ability to inhibit FLT3 kinase activity using Caliper's mobility shift assay (MSA). The assay uses a microfluidic chip to measure the conversion of a fluorescent peptide substrate to a phosphorylated product following separation by electrophoresis. The signature of the fluorescence signal over time reveals the extent of the reaction.
Protein tyrosine kinase assays were carried out in a final volume of 25 ul containing 0.9 nM purified FLT3 (Carna, Cat 08-154) enzyme protein, 50 mM HEPES [pH=7.5], 0.0015 % Brij-35, 10 mM MgCI2, 2 mM DTT, 2% DMSO, 97 uM ATP, 1.5 uM peptide 2. Each Compound was added into the reaction to final concentrations from 300 nM to 0.015 nM (10 concentration points in 3 fold serial dilution). The assay was carried out in 384 well plates at 28 °C for 60 min and terminated by adding 25 ul stop buffer (containing 100 mM HEPES [pH=7.5], 0.015 % Brij-35, 0.2% Coating Reagent #3, 50 mM EDTA). Data were collected on Caliper and converted into inhibition values. IC50 values were obtained using XLfit graphic program.
IC50 values of the compounds of the present invention in inhibiting the FLT3 kinase are listed in following table 2.
Table 2
Figure imgf000051_0001
Table 2 showed that most of the compounds of the present invention have the better FLT3 kinase inhibitory activities than Quizartinib.
3. The present invention compound(C) (3mg/kg, po, qd) was evaluated in MV4-11 acute myeloid leukemia zenograft tumor model, Quizartinib (B) (3mg/kg, po, qd)) used as a positive control.
Compound C (18 mg) in 22% 2-hydroxypropyl^=cyclodextrin (60ml), ultrasonic 15 min to dissolve, an average of 2.14ml divided into each of the small bottle, reserved 22 bottles for experiment.
6-8 weeks NOD/SCID female mice (18-25 g) were used. Each mouse was inoculated subcutaneously at the right flank with MV4-11 tumor cells (1 x 107) in 0.1 ml of PBS/Matrigel (1 :1 ) for tumor development. The treatments were started when the mean tumor size reached 188 mm3. The animals were separated even as test and control group randomly, and each group is consisted of 6 tumor-bearing mice. The test articles were orally administered (2.14 ml) to the tumor-bearing mice once a day. The date of tumor cell inoculation is denoted as day 0. The tumor volume was measured every four days, and the animals were sacrified at 22 days. Statistical analyses of difference in tumor volume among the groups were evaluated using a one-way ANOVA followed by individual comparisons using Games-Howell (equal variance not assumed).
All data were analyzed using SPSS 17.0. p < 0.05 was considered to be statistically significant. The antitumor activities of compounds of disclose are calculated by the formula: T C % =TRTV/CRTV x 100% = (TRTV: drug treated group RTV; CRTV: vihecle treated group RTV) . Evaluation standard: T/C (%) > 40% considered as not effective; T/C (%) < 40 % considered as effective with P < 0.05.
FIG. 1 showed the antitumor effect in MV4-11 acute myeloid leukemia zenograft tumor model assay of the examplary compound of the present invention, where the mean tumor volume of the mice treated with either vehicle (A), positive control compound Quizartinib (B), or the examplary compound (C) of the present inventionis measured and plotted versus the dosing days. It can be seen that the present invention compound (C) (3 mg/kg, po, qd, 22 days) has demonstrated T/C = 8% (P=0.011 ), compared to the positive control compound Quizartinib (B) (3 mg/kg, po, qd, 22 days) with T/C = 9% (P=0.01 ), that means the examplary compound (C) of the present invention may have the better antitumor effect in this model testing.

Claims

1. A Compound of the formula (I) or a pharmaceutically acceptable salt thereof,
Figure imgf000053_0001
Wherein:
Ar is selected from the group consisting of optionally substituted or unsubstitued aryl and heteroaryl, when substituted, the substituents independently selected from halogen, alkyl, haloalkyl, or hydroxylalkyl;
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl, or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyi, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxylalkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkylalkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHheteroarylalkyl;
Y is O, S, NR2R2' or a direct bond;
X-i , X2, X3 and X4 are independently N or CR-i ; Ri is H, or -Y-L;
R, R2, and R2' are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, haloalkoxyl, hydroxyl, amino, aminocarbonyl, sulfonamido, cyano, alkynyl, alkoxyl, aryloxyl, carboxylic acid, carboxylic ester or halogen, or R2, R2' together with the nitrogen atom to which they are attached, formed a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms, the 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl.
2. The compound of the formula (I) or a pharmaceutically acceptable salt thereof according to claim 1 , wherein: Ar is
Figure imgf000053_0002
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl, or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyi, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxylalkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalky- lalkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHhetero- arylalkyl;
Y is O, S, NR2 R2' or a direct bond;
X-i , X2, X3 and X4 are independently N or CR-i ; R-i is H, or -Y-L;
R, R2, and R2' are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, haloalkoxyl, hydroxyl, amino, amido, aminocarbonyl, sulfonamido, cyano, alkynyl, alkoxyl, aryloxyl, carboxylic acid, carboxylic ester or halogen, or R2, R2' together with the nitrogen atom to which they are attached, formed a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms, the 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl.
3. The compound of the formula (I) or a pharmaceutically acceptable salt thereof according to claim 1 , wherein: Ar is
Figure imgf000054_0001
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyi, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyi, heterocycloalkyi, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxylalkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkyl- alkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHhetero- arylalkyl;
Y is O, S or NR2 R2';
X-i , X2, X3 and X4 are independently N or CR-i ; R-i is H, or -Y-L;
R, R2 and R2' are each independently selected from the group consisting of hydrogen or alkyl, or R2, R2' together with the nitrogen atom to which they are attached, formed a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms, the 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl.
4. The compound of the formula (I) or a pharmaceutically acceptable salt thereof according to claim 1 , wherein: Ar is
Figure imgf000054_0002
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyi, optionally or unsubstitued substituted aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxylalkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkyl- alkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHhetero- arylalkyl;
Y is O;
X-i , X2, X3, X4, are independently N or CH;
R is hydrogen.
5. The compound of the formula (I) or a pharmaceutically acceptable salt thereof according to claim 1 , wherein: Ar is
Figure imgf000055_0001
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyl, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxylalkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkyl- alkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHhetero- arylalkyl;
Y is NR2 R2';
R2, R2' is hydrogen or alkyl, or R2,, R2' together with the nitrogen atom to which they are attached, formed a 3- to 7-membered heterocycloalkyl ring, and the hetero atom could be selected from at least one of O, S or N atoms, the 3- to 7-membered heterocycloalkyl ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyl, aryl, or heteroaryl;
X-i , X2, X3, X4, are independently N or CH;
R is hydrogen.
6. The compound of the formula (I) or a pharmaceutically acceptable salt thereof according to claim 1 , wherein: Ar is
Figure imgf000055_0002
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyl, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyl, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxylalkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkyl- alkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHhetero- arylalkyl;
Y is a direct bond, O, S or N R2 R2';
R2, R2' is hydrogen or alkyl, or R2,, R2', together with the nitrogen atom to which they are attached, formed a 3- to 7-membered heterocycloalkyi ring, and the hetero atom could be selected from at least one of O, S or N atoms, the 3- to 7-membered heterocycloalkyi ring could be further optionally substituted with a group independently selected from alkyl, cycloalkyl, methylsulfonyl, ureido, acyl, amido, aminocarbonyl, alkylamino, alkylhydroxyl, heterocycloalkyi, aryl, or heteroaryl;
X-i , X2, X3 and X4 are independently CR-i ; R-i is H, or -Y-L;
R is hydrogen.
7. The compound of the formula (I) or a pharmaceutically acceptable salt thereof according to claim 1 , wherein: Ar is
Figure imgf000056_0001
L is H, optionally substituted or unsubstitued alkyl, optionally substituted or unsubstitued cycloalkyl, optionally substituted or unsubstitued cycloalkylalkyi, optionally substituted or unsubstitued aryl, optionally substituted or unsubstitued arylalkyl, optionally substituted or unsubstitued sulfonamido, optionally substituted or unsubstitued heterocycloalkyi, optionally substituted or unsubstitued heterocycloalkylalkyl, optionally substituted or unsubstitued heteroaryl or optionally substituted or unsubstitued heteroarylalkyl; when substituted, the substituents can be one or more groups independently selected from the group consisting of halo, alkyl, haloalkyl, cycloalkyl, cycloalkylalkyi, heterocycloalkyi, heterocycloalkylalkyl, amino, aminoalkyl, amido, aminocarbonyl, sulfonamido, ureido, cyano, acetyl, acyl, carboxylic acid, hydroxyl, hydroxylalkyl, alkoxyl, -NHalkylhydroxyl, -NHalkoxyalkyl, -NHalkylamnio, -NHcycloalkyl, -NHcycloalkyl- alkyl, -NHheterocycloalkyl, -NHheterocycloalkylalkyl, -NHaryl, -NHarylalkyl, -NHheteroaryl, or -NHheter- oarylalkyl;
Y is a direct bond;
X-i , X2, X3 and X4 are independently N or CR-i ; R-i is H, or -Y-L;
R is independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, haloalkoxyl, hydroxyl, amino, aminocarbonyl, sulfonamido, cyano, alkynyl, alkoxyl, aryloxyl, carboxylic acid, carboxylic ester or halogen.
8. The compound according to claim 1 , which is selected from the following compounds consisting of,
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
9. A pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in any one of claims 1 to 8, as well as a pharmaceutically acceptable carrier, adjuvant, excipient, or diluent
10. A compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in any one of the claims 1 to 8 for the use in preparation of a medicament as Flt3 kinase inhibitor.
11. A compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in any one of the claims 1 to 8 for the use in preparation of a medicament for modulating FLT3-mediated diseases, wherein said medicament comprising administering a therapeutically effective amount of the compound of formula (I) or a pharmaceutically acceptable salt thereof, as well as an isomer, a solvate, a hydrate, or a prodrug.
12. A compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in any one of the claims 1 to 8 for the use in preparation of a medicament for treating a cellular proliferative disorder diseases by inhibiting Flt3 kinase activities, wherein said diseases comprising acute myeloid leukemia, chronic myeloid leukemia, myeloma, ovarian cancer, breast cancer, prostate cancer, pancreatic cancer, colorectal cancer, stomach cancer, non small cell lung cancer, thyroid cancer, brain cancer or lymphoma, said medicament comprising administering a therapeutically effective amount of the compound of formula (I) or a pharmaceutically acceptable salt thereof.
13. A compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in any one of the claims 1 to 8 for the use in preparation of a medicament for treating inflammatory and autoimmune diseases by inhibiting Flt3 kinase activities, wherein said diseases comprising asthma, lupus, systemic lupus, rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, or Crohn's disease, said medicament comprising administering a therapeutically effective amount of the compound of formula (I) or a pharmaceutically acceptable salt thereof.
PCT/CN2012/086492 2011-12-20 2012-12-13 Tri-heterocyclic derivatives, preparation process and uses thereof WO2013091502A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
EP12858726.8A EP2763998B1 (en) 2011-12-20 2012-12-13 Tri-heterocyclic derivatives, preparation process and uses thereof
CA2855764A CA2855764A1 (en) 2011-12-20 2012-12-13 Tri-heterocyclic derivatives, preparation process and uses thereof
AU2012357339A AU2012357339B2 (en) 2011-12-20 2012-12-13 Tri-heterocyclic derivatives, preparation process and uses thereof
US14/356,982 US9216997B2 (en) 2011-12-20 2012-12-13 Tri-heterocyclic derivatives, preparation process and uses thereof
JP2014547693A JP6028306B2 (en) 2011-12-20 2012-12-13 Triheterocyclic derivatives, preparation methods and uses thereof
KR1020147015840A KR20140104427A (en) 2011-12-20 2012-12-13 Tri-heterocyclic derivatives, preparation process and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201110430906.3 2011-12-20
CN201110430906 2011-12-20
CN201210337103.8A CN103172648B (en) 2011-12-20 2012-09-12 Three Hete rocyclic derivatives, preparation method and application
CN201210337103.8 2012-09-12

Publications (1)

Publication Number Publication Date
WO2013091502A1 true WO2013091502A1 (en) 2013-06-27

Family

ID=48632900

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2012/086492 WO2013091502A1 (en) 2011-12-20 2012-12-13 Tri-heterocyclic derivatives, preparation process and uses thereof

Country Status (8)

Country Link
US (1) US9216997B2 (en)
EP (1) EP2763998B1 (en)
JP (1) JP6028306B2 (en)
KR (1) KR20140104427A (en)
CN (1) CN103172648B (en)
AU (1) AU2012357339B2 (en)
CA (1) CA2855764A1 (en)
WO (1) WO2013091502A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015043492A1 (en) * 2013-09-26 2015-04-02 Sunshine Lake Pharma Co., Ltd. Substituted urea derivatives and uses thereof in medicine
CN104513257A (en) * 2013-09-26 2015-04-15 广东东阳光药业有限公司 Substituted urea derivatives and application thereof in drugs
CN105523995A (en) * 2015-12-17 2016-04-27 浙江汇能生物股份有限公司 Preparation method for malaridine intermediate 2-methoxy-5-aminopyridine
WO2017109088A1 (en) 2015-12-23 2017-06-29 Savira Pharmaceuticals Gmbh Pyrimidone derivatives and their use in the treatment, amelioration or prevention of a viral disease
IT201900015030A1 (en) * 2019-08-26 2021-02-26 Univ Degli Studi Di Palermo NEW THERAPEUTIC AGENTS FOR THE TREATMENT OF HEMATOLOGICAL DISORDERS
EP3559006A4 (en) * 2016-12-23 2021-03-03 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of fetal liver kinase polypeptides

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201311891D0 (en) 2013-07-03 2013-08-14 Glaxosmithkline Ip Dev Ltd Novel compound
GB201311888D0 (en) 2013-07-03 2013-08-14 Glaxosmithkline Ip Dev Ltd Novel compounds
EP3169671B1 (en) 2014-07-17 2019-08-21 Sunshine Lake Pharma Co., Ltd. 1-(5-(tert.-butyl)isoxazol-3-yl)-3-(4-((phenyl)ethynyl)phenyl)urea derivatives and related compounds as flt3 inhibitors for treating cancer
CN105801602B (en) * 2014-12-31 2018-05-04 四川大学 5- tertiary Ding isoxazoles -3- bases -3- (4- substituted-phenyls) urea derivative and its preparation method and application
CN105884828A (en) * 2015-02-16 2016-08-24 上海迪诺医药科技有限公司 Polycyclic compound, pharmaceutical composition and application thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007109120A2 (en) * 2006-03-17 2007-09-27 Ambit Biosciences Corporation Imidazolothiazole compounds for the treatment of disease
WO2009038757A2 (en) * 2007-09-19 2009-03-26 Ambit Biosciences Corporation Solid forms comprising n-(5-tert-butyl-isoxazol-3-yl)-n'-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,1-b][1,3]benzothiazol-2-yl]phenyl}urea, compositions thereof, and uses therewith
WO2010054058A1 (en) * 2008-11-06 2010-05-14 Ambit Bioscience Corporation Imidazolothiazole compounds as modulators of protein kinase
WO2011056764A1 (en) * 2009-11-05 2011-05-12 Ambit Biosciences Corp. Isotopically enriched or fluorinated imidazo[2,1-b][1,3]benzothiazoles
EP1041982B1 (en) * 1997-12-22 2011-10-19 Bayer HealthCare LLC INHIBITION OF p38 KINASE ACTIVITY USING SUBSTITUTED HETEROCYCLIC UREAS

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2436266T3 (en) * 2007-11-08 2013-12-30 Ambit Biosciences Corporation Methods for administering N- (5-tert-butyl-isoxazol-3-yl) -N '- {4- [7- (2-morpholin-4-yl-ethoxy) imidazo [2,1-b] [1, 3] benzothiazol-2-yl] phenyl} urea to treat proliferative diseases
CA2755976C (en) * 2009-03-23 2020-04-07 Ambit Biosciences Corporation Methods of treatment using combination therapy

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1041982B1 (en) * 1997-12-22 2011-10-19 Bayer HealthCare LLC INHIBITION OF p38 KINASE ACTIVITY USING SUBSTITUTED HETEROCYCLIC UREAS
WO2007109120A2 (en) * 2006-03-17 2007-09-27 Ambit Biosciences Corporation Imidazolothiazole compounds for the treatment of disease
WO2009038757A2 (en) * 2007-09-19 2009-03-26 Ambit Biosciences Corporation Solid forms comprising n-(5-tert-butyl-isoxazol-3-yl)-n'-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,1-b][1,3]benzothiazol-2-yl]phenyl}urea, compositions thereof, and uses therewith
WO2010054058A1 (en) * 2008-11-06 2010-05-14 Ambit Bioscience Corporation Imidazolothiazole compounds as modulators of protein kinase
WO2011056764A1 (en) * 2009-11-05 2011-05-12 Ambit Biosciences Corp. Isotopically enriched or fluorinated imidazo[2,1-b][1,3]benzothiazoles

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
QI CHAO ET AL.: "Identification of -(5-tert-Butyl-isoxazol-3-yl)-N'-}4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,1-b] [1,3 ]benzothi azol-2-yl]phenyl}ureaDihydrochloride", JOURNAL OF MEDICINAL CHEMISTRY, vol. 52, no. 23, 16 September 2009 (2009-09-16), pages 7808 - 7816, XP002615038 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015043492A1 (en) * 2013-09-26 2015-04-02 Sunshine Lake Pharma Co., Ltd. Substituted urea derivatives and uses thereof in medicine
CN104513257A (en) * 2013-09-26 2015-04-15 广东东阳光药业有限公司 Substituted urea derivatives and application thereof in drugs
CN104513252A (en) * 2013-09-26 2015-04-15 广东东阳光药业有限公司 Substituted urea derivatives and application thereof in drugs
CN105523995A (en) * 2015-12-17 2016-04-27 浙江汇能生物股份有限公司 Preparation method for malaridine intermediate 2-methoxy-5-aminopyridine
WO2017109088A1 (en) 2015-12-23 2017-06-29 Savira Pharmaceuticals Gmbh Pyrimidone derivatives and their use in the treatment, amelioration or prevention of a viral disease
EP3559006A4 (en) * 2016-12-23 2021-03-03 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of fetal liver kinase polypeptides
IT201900015030A1 (en) * 2019-08-26 2021-02-26 Univ Degli Studi Di Palermo NEW THERAPEUTIC AGENTS FOR THE TREATMENT OF HEMATOLOGICAL DISORDERS
WO2021038452A1 (en) * 2019-08-26 2021-03-04 Universita' Degli Studi Di Palermo New therapeutic agents for the treatment of haematological pathologies

Also Published As

Publication number Publication date
US20140329800A1 (en) 2014-11-06
AU2012357339A1 (en) 2014-05-22
EP2763998B1 (en) 2016-04-27
CN103172648B (en) 2016-05-25
EP2763998A1 (en) 2014-08-13
JP2015500844A (en) 2015-01-08
KR20140104427A (en) 2014-08-28
CN103172648A (en) 2013-06-26
EP2763998A4 (en) 2014-09-17
US9216997B2 (en) 2015-12-22
CA2855764A1 (en) 2013-06-27
AU2012357339B2 (en) 2016-04-21
JP6028306B2 (en) 2016-11-16

Similar Documents

Publication Publication Date Title
EP2763998B1 (en) Tri-heterocyclic derivatives, preparation process and uses thereof
EP3600270B1 (en) Compounds and compositions for treating hematological disorders
CN112062778B (en) Benzoxazepine oxazolidinone compounds and methods of use thereof
JP7476380B2 (en) Compounds and compositions for treating hematological disorders
US9573943B2 (en) Pyrazol-4-yl-heterocyclyl-carboxamide compounds and methods of use
JP6522807B2 (en) Benzoxazepine oxazolidinone compounds and methods of use
US10968203B2 (en) Pyrimidinyl-pyridyloxy-naphthyl compounds and methods of treating IRE1-related diseases and disorders
PH12015500488B1 (en) Cyclic ether pyrazol-4-yl-heterocyclyl-carboxamide compounds and methods of use
CN103896942A (en) Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
BR112012018415A2 (en) compound, composition, methods of preparing a composition and treatment, and, use of a compound.
CA3104521A1 (en) Pikfyve inhibitors
EP2828257A1 (en) Antibacterial compounds
TW202325298A (en) A nitrogen-containing tetracyclic compound, a preparation method and medical use thereof
CN113354630B (en) 5,6-dihydrobenzo [ h ] quinazoline compound and application thereof
WO2023099072A1 (en) Compounds
CN117886813A (en) SHP2 phosphatase allosteric inhibitors
CN117677614A (en) Heterocyclic compound with AKT kinase inhibition activity, preparation method and medical application thereof
BR112017023121B1 (en) COMPOUND, PHARMACEUTICAL COMPOSITION, PROCESS FOR PREPARING A PHARMACEUTICAL COMPOSITION, KIT FOR THE THERAPEUTIC TREATMENT OF CANCER, AND USE OF A COMPOUND

Legal Events

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

Ref document number: 12858726

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
ENP Entry into the national phase

Ref document number: 2014547693

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2012858726

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 14356982

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2855764

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2012357339

Country of ref document: AU

Date of ref document: 20121213

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20147015840

Country of ref document: KR

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE