WO2024249642A1 - Egfr inhibitors - Google Patents
Egfr inhibitors Download PDFInfo
- Publication number
- WO2024249642A1 WO2024249642A1 PCT/US2024/031685 US2024031685W WO2024249642A1 WO 2024249642 A1 WO2024249642 A1 WO 2024249642A1 US 2024031685 W US2024031685 W US 2024031685W WO 2024249642 A1 WO2024249642 A1 WO 2024249642A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- compound
- mmol
- cycloalkyl
- alkyl
- pyrimidin
- Prior art date
Links
- 229940121647 egfr inhibitor Drugs 0.000 title description 12
- 150000001875 compounds Chemical class 0.000 claims abstract description 179
- 238000000034 method Methods 0.000 claims abstract description 39
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 30
- 201000011510 cancer Diseases 0.000 claims abstract description 21
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 14
- 230000002401 inhibitory effect Effects 0.000 claims abstract description 13
- 150000003839 salts Chemical class 0.000 claims abstract description 8
- 102000052116 epidermal growth factor receptor activity proteins Human genes 0.000 claims abstract 2
- 108700015053 epidermal growth factor receptor activity proteins Proteins 0.000 claims abstract 2
- YOHYSYJDKVYCJI-UHFFFAOYSA-N n-[3-[[6-[3-(trifluoromethyl)anilino]pyrimidin-4-yl]amino]phenyl]cyclopropanecarboxamide Chemical compound FC(F)(F)C1=CC=CC(NC=2N=CN=C(NC=3C=C(NC(=O)C4CC4)C=CC=3)C=2)=C1 YOHYSYJDKVYCJI-UHFFFAOYSA-N 0.000 claims abstract 2
- -1 NR1aR1b Chemical group 0.000 claims description 190
- 229910052757 nitrogen Inorganic materials 0.000 claims description 47
- 125000005913 (C3-C6) cycloalkyl group Chemical group 0.000 claims description 30
- 125000000623 heterocyclic group Chemical group 0.000 claims description 22
- YZCKVEUIGOORGS-OUBTZVSYSA-N Deuterium Chemical compound [2H] YZCKVEUIGOORGS-OUBTZVSYSA-N 0.000 claims description 21
- 229910052805 deuterium Inorganic materials 0.000 claims description 21
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 20
- 125000001559 cyclopropyl group Chemical group [H]C1([H])C([H])([H])C1([H])* 0.000 claims description 19
- JNCMHMUGTWEVOZ-UHFFFAOYSA-N F[CH]F Chemical compound F[CH]F JNCMHMUGTWEVOZ-UHFFFAOYSA-N 0.000 claims description 17
- 125000005843 halogen group Chemical group 0.000 claims description 17
- WCYWZMWISLQXQU-FIBGUPNXSA-N trideuteriomethane Chemical compound [2H][C]([2H])[2H] WCYWZMWISLQXQU-FIBGUPNXSA-N 0.000 claims description 16
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 claims description 16
- 229910052731 fluorine Inorganic materials 0.000 claims description 13
- 229910052799 carbon Inorganic materials 0.000 claims description 12
- 125000004169 (C1-C6) alkyl group Chemical group 0.000 claims description 11
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 10
- 125000000217 alkyl group Chemical group 0.000 claims description 10
- 201000005202 lung cancer Diseases 0.000 claims description 10
- 208000020816 lung neoplasm Diseases 0.000 claims description 10
- 125000003118 aryl group Chemical group 0.000 claims description 9
- 125000006273 (C1-C3) alkyl group Chemical group 0.000 claims description 8
- 125000001072 heteroaryl group Chemical group 0.000 claims description 7
- 208000003174 Brain Neoplasms Diseases 0.000 claims description 6
- 125000001313 C5-C10 heteroaryl group Chemical group 0.000 claims description 6
- 125000004432 carbon atom Chemical group C* 0.000 claims description 6
- 229910052801 chlorine Inorganic materials 0.000 claims description 6
- 125000004093 cyano group Chemical group *C#N 0.000 claims description 6
- 125000000753 cycloalkyl group Chemical group 0.000 claims description 6
- 125000001153 fluoro group Chemical group F* 0.000 claims description 6
- 229910052739 hydrogen Inorganic materials 0.000 claims description 6
- 125000006570 (C5-C6) heteroaryl group Chemical group 0.000 claims description 5
- 125000000041 C6-C10 aryl group Chemical group 0.000 claims description 5
- 239000003937 drug carrier Substances 0.000 claims description 5
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 claims description 5
- 125000004178 (C1-C4) alkyl group Chemical group 0.000 claims description 4
- 125000000229 (C1-C4)alkoxy group Chemical group 0.000 claims description 4
- 125000002883 imidazolyl group Chemical group 0.000 claims description 4
- 125000000714 pyrimidinyl group Chemical group 0.000 claims description 4
- 125000002943 quinolinyl group Chemical group N1=C(C=CC2=CC=CC=C12)* 0.000 claims description 4
- 229910052794 bromium Inorganic materials 0.000 claims description 3
- 125000003406 indolizinyl group Chemical group C=1(C=CN2C=CC=CC12)* 0.000 claims description 3
- 125000003226 pyrazolyl group Chemical group 0.000 claims description 3
- MPVDXIMFBOLMNW-UHFFFAOYSA-N chembl1615565 Chemical compound OC1=CC=C2C=C(S(O)(=O)=O)C=C(S(O)(=O)=O)C2=C1N=NC1=CC=CC=C1 MPVDXIMFBOLMNW-UHFFFAOYSA-N 0.000 claims description 2
- 125000000000 cycloalkoxy group Chemical group 0.000 claims description 2
- 125000001028 difluoromethyl group Chemical group [H]C(F)(F)* 0.000 claims description 2
- 125000004433 nitrogen atom Chemical group N* 0.000 claims description 2
- 125000004076 pyridyl group Chemical group 0.000 claims description 2
- 125000000335 thiazolyl group Chemical group 0.000 claims description 2
- 125000001425 triazolyl group Chemical group 0.000 claims description 2
- 125000003373 pyrazinyl group Chemical group 0.000 claims 1
- 125000002098 pyridazinyl group Chemical group 0.000 claims 1
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 104
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 95
- 239000000203 mixture Substances 0.000 description 95
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 84
- 238000003786 synthesis reaction Methods 0.000 description 82
- 230000015572 biosynthetic process Effects 0.000 description 79
- OKKJLVBELUTLKV-MZCSYVLQSA-N Deuterated methanol Chemical compound [2H]OC([2H])([2H])[2H] OKKJLVBELUTLKV-MZCSYVLQSA-N 0.000 description 76
- 239000000243 solution Substances 0.000 description 75
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 65
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 65
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 64
- 238000005160 1H NMR spectroscopy Methods 0.000 description 59
- 230000002829 reductive effect Effects 0.000 description 58
- 210000004027 cell Anatomy 0.000 description 55
- 239000007787 solid Substances 0.000 description 55
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 54
- 239000000543 intermediate Substances 0.000 description 43
- 102000001301 EGF receptor Human genes 0.000 description 40
- 108060006698 EGF receptor Proteins 0.000 description 40
- 239000007832 Na2SO4 Substances 0.000 description 40
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 40
- 229910052938 sodium sulfate Inorganic materials 0.000 description 40
- 239000012044 organic layer Substances 0.000 description 38
- 239000000706 filtrate Substances 0.000 description 36
- 235000019439 ethyl acetate Nutrition 0.000 description 32
- 239000012267 brine Substances 0.000 description 31
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical compound O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 31
- 239000002904 solvent Substances 0.000 description 31
- 239000011541 reaction mixture Substances 0.000 description 29
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 26
- IAZDPXIOMUYVGZ-WFGJKAKNSA-N Dimethyl sulfoxide Chemical compound [2H]C([2H])([2H])S(=O)C([2H])([2H])[2H] IAZDPXIOMUYVGZ-WFGJKAKNSA-N 0.000 description 24
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 23
- 238000010898 silica gel chromatography Methods 0.000 description 23
- KDLHZDBZIXYQEI-UHFFFAOYSA-N Palladium Chemical compound [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 description 21
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 17
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 16
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 16
- 241001465754 Metazoa Species 0.000 description 16
- HEDRZPFGACZZDS-MICDWDOJSA-N Trichloro(2H)methane Chemical compound [2H]C(Cl)(Cl)Cl HEDRZPFGACZZDS-MICDWDOJSA-N 0.000 description 16
- 230000035772 mutation Effects 0.000 description 16
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 15
- 241000699666 Mus <mouse, genus> Species 0.000 description 14
- 241000699670 Mus sp. Species 0.000 description 13
- 210000004556 brain Anatomy 0.000 description 13
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 12
- BWHMMNNQKKPAPP-UHFFFAOYSA-L potassium carbonate Chemical compound [K+].[K+].[O-]C([O-])=O BWHMMNNQKKPAPP-UHFFFAOYSA-L 0.000 description 12
- 238000006243 chemical reaction Methods 0.000 description 11
- 229910000104 sodium hydride Inorganic materials 0.000 description 11
- NLXLAEXVIDQMFP-UHFFFAOYSA-N Ammonia chloride Chemical compound [NH4+].[Cl-] NLXLAEXVIDQMFP-UHFFFAOYSA-N 0.000 description 10
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 10
- 238000005415 bioluminescence Methods 0.000 description 10
- 230000029918 bioluminescence Effects 0.000 description 10
- 238000003818 flash chromatography Methods 0.000 description 9
- VLKZOEOYAKHREP-UHFFFAOYSA-N n-Hexane Chemical compound CCCCCC VLKZOEOYAKHREP-UHFFFAOYSA-N 0.000 description 9
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 9
- 239000003921 oil Substances 0.000 description 9
- 238000002953 preparative HPLC Methods 0.000 description 9
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 9
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 description 9
- FJDQFPXHSGXQBY-UHFFFAOYSA-L caesium carbonate Chemical compound [Cs+].[Cs+].[O-]C([O-])=O FJDQFPXHSGXQBY-UHFFFAOYSA-L 0.000 description 8
- 230000001404 mediated effect Effects 0.000 description 8
- 239000002953 phosphate buffered saline Substances 0.000 description 8
- 102200048928 rs121434568 Human genes 0.000 description 8
- 238000012360 testing method Methods 0.000 description 8
- 238000004809 thin layer chromatography Methods 0.000 description 8
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 7
- 101000851181 Homo sapiens Epidermal growth factor receptor Proteins 0.000 description 7
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 7
- 239000000460 chlorine Substances 0.000 description 7
- UQLDLKMNUJERMK-UHFFFAOYSA-L di(octadecanoyloxy)lead Chemical compound [Pb+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O UQLDLKMNUJERMK-UHFFFAOYSA-L 0.000 description 7
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 7
- 102000045108 human EGFR Human genes 0.000 description 7
- 238000011534 incubation Methods 0.000 description 7
- 239000012299 nitrogen atmosphere Substances 0.000 description 7
- CTSLXHKWHWQRSH-UHFFFAOYSA-N oxalyl chloride Chemical compound ClC(=O)C(Cl)=O CTSLXHKWHWQRSH-UHFFFAOYSA-N 0.000 description 7
- RIOQSEWOXXDEQQ-UHFFFAOYSA-N triphenylphosphine Chemical compound C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 RIOQSEWOXXDEQQ-UHFFFAOYSA-N 0.000 description 7
- 239000003981 vehicle Substances 0.000 description 7
- KZPYGQFFRCFCPP-UHFFFAOYSA-N 1,1'-bis(diphenylphosphino)ferrocene Chemical compound [Fe+2].C1=CC=C[C-]1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=C[C-]1P(C=1C=CC=CC=1)C1=CC=CC=C1 KZPYGQFFRCFCPP-UHFFFAOYSA-N 0.000 description 6
- KEZNMOUMHOZFRA-UHFFFAOYSA-N 1h-pyrazol-4-ylboronic acid Chemical compound OB(O)C=1C=NNC=1 KEZNMOUMHOZFRA-UHFFFAOYSA-N 0.000 description 6
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 6
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 6
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 6
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 6
- RYGOBSYXIIUFOR-FIBGUPNXSA-N [1-(trideuteriomethyl)pyrazol-4-yl]boronic acid Chemical compound [2H]C([2H])([2H])n1cc(cn1)B(O)O RYGOBSYXIIUFOR-FIBGUPNXSA-N 0.000 description 6
- 238000003556 assay Methods 0.000 description 6
- BDAGIHXWWSANSR-UHFFFAOYSA-N methanoic acid Natural products OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 6
- 238000003756 stirring Methods 0.000 description 6
- AVRPFRMDMNDIDH-UHFFFAOYSA-N 1h-quinazolin-2-one Chemical compound C1=CC=CC2=NC(O)=NC=C21 AVRPFRMDMNDIDH-UHFFFAOYSA-N 0.000 description 5
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 5
- 206010027476 Metastases Diseases 0.000 description 5
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 5
- 125000003545 alkoxy group Chemical group 0.000 description 5
- 230000004075 alteration Effects 0.000 description 5
- 230000037396 body weight Effects 0.000 description 5
- 238000001516 cell proliferation assay Methods 0.000 description 5
- 239000003153 chemical reaction reagent Substances 0.000 description 5
- 239000003795 chemical substances by application Substances 0.000 description 5
- 239000012230 colorless oil Substances 0.000 description 5
- 230000008878 coupling Effects 0.000 description 5
- 238000010168 coupling process Methods 0.000 description 5
- 238000005859 coupling reaction Methods 0.000 description 5
- 238000005516 engineering process Methods 0.000 description 5
- 239000012091 fetal bovine serum Substances 0.000 description 5
- 239000007924 injection Substances 0.000 description 5
- 238000002347 injection Methods 0.000 description 5
- 238000011081 inoculation Methods 0.000 description 5
- 230000009401 metastasis Effects 0.000 description 5
- JLPBFHATJXMBFM-UHFFFAOYSA-N n,n'-bis(3-chloro-2-fluorophenyl)methanimidamide Chemical compound FC1=C(Cl)C=CC=C1NC=NC1=CC=CC(Cl)=C1F JLPBFHATJXMBFM-UHFFFAOYSA-N 0.000 description 5
- 229910052763 palladium Inorganic materials 0.000 description 5
- 229910000027 potassium carbonate Inorganic materials 0.000 description 5
- 230000004083 survival effect Effects 0.000 description 5
- RYGOBSYXIIUFOR-UHFFFAOYSA-N (1-methylpyrazol-4-yl)boronic acid Chemical compound CN1C=C(B(O)O)C=N1 RYGOBSYXIIUFOR-UHFFFAOYSA-N 0.000 description 4
- VKOOOTNDYLOXMQ-UHFFFAOYSA-N 2-(3-methylpyridin-4-yl)propan-2-ol Chemical compound Cc1cnccc1C(C)(C)O VKOOOTNDYLOXMQ-UHFFFAOYSA-N 0.000 description 4
- TVTJUIAKQFIXCE-HUKYDQBMSA-N 2-amino-9-[(2R,3S,4S,5R)-4-fluoro-3-hydroxy-5-(hydroxymethyl)oxolan-2-yl]-7-prop-2-ynyl-1H-purine-6,8-dione Chemical compound NC=1NC(C=2N(C(N(C=2N=1)[C@@H]1O[C@@H]([C@H]([C@H]1O)F)CO)=O)CC#C)=O TVTJUIAKQFIXCE-HUKYDQBMSA-N 0.000 description 4
- 206010002091 Anaesthesia Diseases 0.000 description 4
- IGXWBGJHJZYPQS-SSDOTTSWSA-N D-Luciferin Chemical compound OC(=O)[C@H]1CSC(C=2SC3=CC=C(O)C=C3N=2)=N1 IGXWBGJHJZYPQS-SSDOTTSWSA-N 0.000 description 4
- CSNNHWWHGAXBCP-UHFFFAOYSA-L Magnesium sulfate Chemical compound [Mg+2].[O-][S+2]([O-])([O-])[O-] CSNNHWWHGAXBCP-UHFFFAOYSA-L 0.000 description 4
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 4
- 239000012980 RPMI-1640 medium Substances 0.000 description 4
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 4
- DTQVDTLACAAQTR-UHFFFAOYSA-N Trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 4
- UOXFJNMDWKLYQZ-UHFFFAOYSA-N [1-(difluoromethyl)pyrazol-4-yl]boronic acid Chemical compound OB(O)C=1C=NN(C(F)F)C=1 UOXFJNMDWKLYQZ-UHFFFAOYSA-N 0.000 description 4
- 238000004458 analytical method Methods 0.000 description 4
- 239000007864 aqueous solution Substances 0.000 description 4
- 230000008499 blood brain barrier function Effects 0.000 description 4
- 210000001218 blood-brain barrier Anatomy 0.000 description 4
- 229910000024 caesium carbonate Inorganic materials 0.000 description 4
- 150000001721 carbon Chemical group 0.000 description 4
- 229940125851 compound 27 Drugs 0.000 description 4
- 238000001816 cooling Methods 0.000 description 4
- 238000001990 intravenous administration Methods 0.000 description 4
- 239000003446 ligand Substances 0.000 description 4
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 4
- NXPHGHWWQRMDIA-UHFFFAOYSA-M magnesium;carbanide;bromide Chemical compound [CH3-].[Mg+2].[Br-] NXPHGHWWQRMDIA-UHFFFAOYSA-M 0.000 description 4
- 239000000546 pharmaceutical excipient Substances 0.000 description 4
- XHXFXVLFKHQFAL-UHFFFAOYSA-N phosphoryl trichloride Chemical compound ClP(Cl)(Cl)=O XHXFXVLFKHQFAL-UHFFFAOYSA-N 0.000 description 4
- LPNYRYFBWFDTMA-UHFFFAOYSA-N potassium tert-butoxide Chemical compound [K+].CC(C)(C)[O-] LPNYRYFBWFDTMA-UHFFFAOYSA-N 0.000 description 4
- 239000000047 product Substances 0.000 description 4
- 150000003246 quinazolines Chemical class 0.000 description 4
- 239000000725 suspension Substances 0.000 description 4
- FYSNRJHAOHDILO-UHFFFAOYSA-N thionyl chloride Chemical compound ClS(Cl)=O FYSNRJHAOHDILO-UHFFFAOYSA-N 0.000 description 4
- 125000004776 1-fluoroethyl group Chemical group [H]C([H])([H])C([H])(F)* 0.000 description 3
- JWUJQDFVADABEY-UHFFFAOYSA-N 2-methyltetrahydrofuran Chemical compound CC1CCCO1 JWUJQDFVADABEY-UHFFFAOYSA-N 0.000 description 3
- ZNZRVANGLTZTCR-UHFFFAOYSA-N 2-pyrimidin-2-ylpropan-2-ol Chemical compound CC(C)(O)C1=NC=CC=N1 ZNZRVANGLTZTCR-UHFFFAOYSA-N 0.000 description 3
- XWBTZHDDWRNOQH-UHFFFAOYSA-N 3-chloro-2-fluoroaniline Chemical compound NC1=CC=CC(Cl)=C1F XWBTZHDDWRNOQH-UHFFFAOYSA-N 0.000 description 3
- RQRIMESFWBLDAP-UHFFFAOYSA-N 7-bromo-5-fluoro-3H-quinazolin-4-one Chemical compound BrC1=CC(=C2C(NC=NC2=C1)=O)F RQRIMESFWBLDAP-UHFFFAOYSA-N 0.000 description 3
- WVDDGKGOMKODPV-UHFFFAOYSA-N Benzyl alcohol Chemical compound OCC1=CC=CC=C1 WVDDGKGOMKODPV-UHFFFAOYSA-N 0.000 description 3
- BTBUEUYNUDRHOZ-UHFFFAOYSA-N Borate Chemical class [O-]B([O-])[O-] BTBUEUYNUDRHOZ-UHFFFAOYSA-N 0.000 description 3
- CYCGRDQQIOGCKX-UHFFFAOYSA-N Dehydro-luciferin Natural products OC(=O)C1=CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 CYCGRDQQIOGCKX-UHFFFAOYSA-N 0.000 description 3
- CXFSTHGZJSFROJ-UHFFFAOYSA-N FC1=C2C(Cl)=NC=NC2=CC(Br)=C1 Chemical compound FC1=C2C(Cl)=NC=NC2=CC(Br)=C1 CXFSTHGZJSFROJ-UHFFFAOYSA-N 0.000 description 3
- BJGNCJDXODQBOB-UHFFFAOYSA-N Fivefly Luciferin Natural products OC(=O)C1CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 BJGNCJDXODQBOB-UHFFFAOYSA-N 0.000 description 3
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 3
- 239000005411 L01XE02 - Gefitinib Substances 0.000 description 3
- 239000005551 L01XE03 - Erlotinib Substances 0.000 description 3
- DDWFXDSYGUXRAY-UHFFFAOYSA-N Luciferin Natural products CCc1c(C)c(CC2NC(=O)C(=C2C=C)C)[nH]c1Cc3[nH]c4C(=C5/NC(CC(=O)O)C(C)C5CC(=O)O)CC(=O)c4c3C DDWFXDSYGUXRAY-UHFFFAOYSA-N 0.000 description 3
- ZRVUJXDFFKFLMG-UHFFFAOYSA-N Meloxicam Chemical compound OC=1C2=CC=CC=C2S(=O)(=O)N(C)C=1C(=O)NC1=NC=C(C)S1 ZRVUJXDFFKFLMG-UHFFFAOYSA-N 0.000 description 3
- 241000700159 Rattus Species 0.000 description 3
- XPOLVIIHTDKJRY-UHFFFAOYSA-N acetic acid;methanimidamide Chemical compound NC=N.CC(O)=O XPOLVIIHTDKJRY-UHFFFAOYSA-N 0.000 description 3
- 239000004480 active ingredient Substances 0.000 description 3
- 239000013543 active substance Substances 0.000 description 3
- 230000000259 anti-tumor effect Effects 0.000 description 3
- 239000002518 antifoaming agent Substances 0.000 description 3
- 125000003710 aryl alkyl group Chemical group 0.000 description 3
- 125000002619 bicyclic group Chemical group 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- ZADPBFCGQRWHPN-UHFFFAOYSA-N boronic acid Chemical class OBO ZADPBFCGQRWHPN-UHFFFAOYSA-N 0.000 description 3
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 3
- 230000004663 cell proliferation Effects 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- 125000001309 chloro group Chemical group Cl* 0.000 description 3
- 239000012043 crude product Substances 0.000 description 3
- 239000003814 drug Substances 0.000 description 3
- 229960001433 erlotinib Drugs 0.000 description 3
- AAKJLRGGTJKAMG-UHFFFAOYSA-N erlotinib Chemical compound C=12C=C(OCCOC)C(OCCOC)=CC2=NC=NC=1NC1=CC=CC(C#C)=C1 AAKJLRGGTJKAMG-UHFFFAOYSA-N 0.000 description 3
- 239000000284 extract Substances 0.000 description 3
- 235000019253 formic acid Nutrition 0.000 description 3
- 238000009472 formulation Methods 0.000 description 3
- XGALLCVXEZPNRQ-UHFFFAOYSA-N gefitinib Chemical compound C=12C=C(OCCCN3CCOCC3)C(OC)=CC2=NC=NC=1NC1=CC=C(F)C(Cl)=C1 XGALLCVXEZPNRQ-UHFFFAOYSA-N 0.000 description 3
- 229960002584 gefitinib Drugs 0.000 description 3
- 125000001188 haloalkyl group Chemical group 0.000 description 3
- RAXXELZNTBOGNW-UHFFFAOYSA-N imidazole Natural products C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 description 3
- 238000001294 liquid chromatography-tandem mass spectrometry Methods 0.000 description 3
- 229960001929 meloxicam Drugs 0.000 description 3
- 125000002950 monocyclic group Chemical group 0.000 description 3
- 125000000449 nitro group Chemical group [O-][N+](*)=O 0.000 description 3
- 231100000590 oncogenic Toxicity 0.000 description 3
- 230000002246 oncogenic effect Effects 0.000 description 3
- 238000003305 oral gavage Methods 0.000 description 3
- 229960003278 osimertinib Drugs 0.000 description 3
- DUYJMQONPNNFPI-UHFFFAOYSA-N osimertinib Chemical compound COC1=CC(N(C)CCN(C)C)=C(NC(=O)C=C)C=C1NC1=NC=CC(C=2C3=CC=CC=C3N(C)C=2)=N1 DUYJMQONPNNFPI-UHFFFAOYSA-N 0.000 description 3
- 230000002018 overexpression Effects 0.000 description 3
- 239000012071 phase Substances 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 229940002612 prodrug Drugs 0.000 description 3
- 239000000651 prodrug Substances 0.000 description 3
- 238000000746 purification Methods 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 229920006395 saturated elastomer Polymers 0.000 description 3
- 239000012279 sodium borohydride Substances 0.000 description 3
- 229910000033 sodium borohydride Inorganic materials 0.000 description 3
- JVBXVOWTABLYPX-UHFFFAOYSA-L sodium dithionite Chemical compound [Na+].[Na+].[O-]S(=O)S([O-])=O JVBXVOWTABLYPX-UHFFFAOYSA-L 0.000 description 3
- 238000012453 sprague-dawley rat model Methods 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- 229910052717 sulfur Inorganic materials 0.000 description 3
- 208000024891 symptom Diseases 0.000 description 3
- 230000001225 therapeutic effect Effects 0.000 description 3
- WTFFOOAJSDVASL-UHFFFAOYSA-N tributyl(pyrimidin-2-yl)stannane Chemical compound CCCC[Sn](CCCC)(CCCC)C1=NC=CC=N1 WTFFOOAJSDVASL-UHFFFAOYSA-N 0.000 description 3
- 230000004614 tumor growth Effects 0.000 description 3
- HACQYPYUKMKUPY-UHFFFAOYSA-N (1-cyclopropylpyrazol-4-yl)boronic acid Chemical compound C1=C(B(O)O)C=NN1C1CC1 HACQYPYUKMKUPY-UHFFFAOYSA-N 0.000 description 2
- ASGMFNBUXDJWJJ-JLCFBVMHSA-N (1R,3R)-3-[[3-bromo-1-[4-(5-methyl-1,3,4-thiadiazol-2-yl)phenyl]pyrazolo[3,4-d]pyrimidin-6-yl]amino]-N,1-dimethylcyclopentane-1-carboxamide Chemical compound BrC1=NN(C2=NC(=NC=C21)N[C@H]1C[C@@](CC1)(C(=O)NC)C)C1=CC=C(C=C1)C=1SC(=NN=1)C ASGMFNBUXDJWJJ-JLCFBVMHSA-N 0.000 description 2
- YTMHZXYXPHNWMA-YFKPBYRVSA-N (1s)-1-pyrimidin-2-ylethanol Chemical compound C[C@H](O)C1=NC=CC=N1 YTMHZXYXPHNWMA-YFKPBYRVSA-N 0.000 description 2
- GLGNXYJARSMNGJ-VKTIVEEGSA-N (1s,2s,3r,4r)-3-[[5-chloro-2-[(1-ethyl-6-methoxy-2-oxo-4,5-dihydro-3h-1-benzazepin-7-yl)amino]pyrimidin-4-yl]amino]bicyclo[2.2.1]hept-5-ene-2-carboxamide Chemical compound CCN1C(=O)CCCC2=C(OC)C(NC=3N=C(C(=CN=3)Cl)N[C@H]3[C@H]([C@@]4([H])C[C@@]3(C=C4)[H])C(N)=O)=CC=C21 GLGNXYJARSMNGJ-VKTIVEEGSA-N 0.000 description 2
- GHYOCDFICYLMRF-UTIIJYGPSA-N (2S,3R)-N-[(2S)-3-(cyclopenten-1-yl)-1-[(2R)-2-methyloxiran-2-yl]-1-oxopropan-2-yl]-3-hydroxy-3-(4-methoxyphenyl)-2-[[(2S)-2-[(2-morpholin-4-ylacetyl)amino]propanoyl]amino]propanamide Chemical compound C1(=CCCC1)C[C@@H](C(=O)[C@@]1(OC1)C)NC([C@H]([C@@H](C1=CC=C(C=C1)OC)O)NC([C@H](C)NC(CN1CCOCC1)=O)=O)=O GHYOCDFICYLMRF-UTIIJYGPSA-N 0.000 description 2
- GCTFTMWXZFLTRR-GFCCVEGCSA-N (2r)-2-amino-n-[3-(difluoromethoxy)-4-(1,3-oxazol-5-yl)phenyl]-4-methylpentanamide Chemical compound FC(F)OC1=CC(NC(=O)[C@H](N)CC(C)C)=CC=C1C1=CN=CO1 GCTFTMWXZFLTRR-GFCCVEGCSA-N 0.000 description 2
- STBLNCCBQMHSRC-BATDWUPUSA-N (2s)-n-[(3s,4s)-5-acetyl-7-cyano-4-methyl-1-[(2-methylnaphthalen-1-yl)methyl]-2-oxo-3,4-dihydro-1,5-benzodiazepin-3-yl]-2-(methylamino)propanamide Chemical compound O=C1[C@@H](NC(=O)[C@H](C)NC)[C@H](C)N(C(C)=O)C2=CC(C#N)=CC=C2N1CC1=C(C)C=CC2=CC=CC=C12 STBLNCCBQMHSRC-BATDWUPUSA-N 0.000 description 2
- UDQTXCHQKHIQMH-KYGLGHNPSA-N (3ar,5s,6s,7r,7ar)-5-(difluoromethyl)-2-(ethylamino)-5,6,7,7a-tetrahydro-3ah-pyrano[3,2-d][1,3]thiazole-6,7-diol Chemical compound S1C(NCC)=N[C@H]2[C@@H]1O[C@H](C(F)F)[C@@H](O)[C@@H]2O UDQTXCHQKHIQMH-KYGLGHNPSA-N 0.000 description 2
- KLKYHMHIZULRNF-UHFFFAOYSA-N (4-bromo-2-fluoro-5-nitrophenyl) ethyl carbonate Chemical compound CCOC(=O)OC1=CC([N+]([O-])=O)=C(Br)C=C1F KLKYHMHIZULRNF-UHFFFAOYSA-N 0.000 description 2
- QFKWURUXHYYPGR-UHFFFAOYSA-N (4-bromo-2-fluorophenyl) ethyl carbonate Chemical compound CCOC(=O)OC1=CC=C(Br)C=C1F QFKWURUXHYYPGR-UHFFFAOYSA-N 0.000 description 2
- KKHFRAFPESRGGD-UHFFFAOYSA-N 1,3-dimethyl-7-[3-(n-methylanilino)propyl]purine-2,6-dione Chemical compound C1=NC=2N(C)C(=O)N(C)C(=O)C=2N1CCCN(C)C1=CC=CC=C1 KKHFRAFPESRGGD-UHFFFAOYSA-N 0.000 description 2
- JWUGKJKBCKJFOU-UHFFFAOYSA-N 1,3-thiazol-2-ylboronic acid Chemical compound OB(O)C1=NC=CS1 JWUGKJKBCKJFOU-UHFFFAOYSA-N 0.000 description 2
- 239000004912 1,5-cyclooctadiene Substances 0.000 description 2
- RZNHHGMCDDENDY-UHFFFAOYSA-N 1-(1-methylimidazol-2-yl)ethanol Chemical compound CC(O)C1=NC=CN1C RZNHHGMCDDENDY-UHFFFAOYSA-N 0.000 description 2
- KKMMTRPLGRQILS-UHFFFAOYSA-N 1-(1-methylindol-2-yl)ethanol Chemical compound C1=CC=C2N(C)C(C(O)C)=CC2=C1 KKMMTRPLGRQILS-UHFFFAOYSA-N 0.000 description 2
- MHSLDASSAFCCDO-UHFFFAOYSA-N 1-(5-tert-butyl-2-methylpyrazol-3-yl)-3-(4-pyridin-4-yloxyphenyl)urea Chemical compound CN1N=C(C(C)(C)C)C=C1NC(=O)NC(C=C1)=CC=C1OC1=CC=NC=C1 MHSLDASSAFCCDO-UHFFFAOYSA-N 0.000 description 2
- ATWIFXNYFADQFA-UHFFFAOYSA-N 1-(difluoromethyl)pyrazole-3-carbaldehyde Chemical compound FC(F)N1C=CC(C=O)=N1 ATWIFXNYFADQFA-UHFFFAOYSA-N 0.000 description 2
- GCVIZGROEHEVRV-UHFFFAOYSA-N 1-(difluoromethyl)pyrazole-4-carbaldehyde Chemical compound FC(F)N1C=C(C=O)C=N1 GCVIZGROEHEVRV-UHFFFAOYSA-N 0.000 description 2
- ONBQEOIKXPHGMB-VBSBHUPXSA-N 1-[2-[(2s,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]oxy-4,6-dihydroxyphenyl]-3-(4-hydroxyphenyl)propan-1-one Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1OC1=CC(O)=CC(O)=C1C(=O)CCC1=CC=C(O)C=C1 ONBQEOIKXPHGMB-VBSBHUPXSA-N 0.000 description 2
- XSVXRIZSFVAIRI-UHFFFAOYSA-N 1-[4-(Difluoromethyl)phenyl]ethanone Chemical compound CC(=O)C1=CC=C(C(F)F)C=C1 XSVXRIZSFVAIRI-UHFFFAOYSA-N 0.000 description 2
- IXCFPGIBOGCRPU-UHFFFAOYSA-N 1-[4-(difluoromethyl)phenyl]ethanol Chemical compound CC(O)C1=CC=C(C(F)F)C=C1 IXCFPGIBOGCRPU-UHFFFAOYSA-N 0.000 description 2
- FCQKOPJZMPKYGR-UHFFFAOYSA-N 1-pyrimidin-4-ylethanol Chemical compound CC(O)C1=CC=NC=N1 FCQKOPJZMPKYGR-UHFFFAOYSA-N 0.000 description 2
- UZKADDSUUBRMKO-UHFFFAOYSA-N 1-pyrimidin-4-ylethanone Chemical compound CC(=O)C1=CC=NC=N1 UZKADDSUUBRMKO-UHFFFAOYSA-N 0.000 description 2
- MWGMIUFKOSHPDB-UHFFFAOYSA-N 2-(1,3-thiazol-2-yl)propan-2-ol Chemical compound CC(C)(O)C1=NC=CS1 MWGMIUFKOSHPDB-UHFFFAOYSA-N 0.000 description 2
- HZAXFHJVJLSVMW-UHFFFAOYSA-N 2-Aminoethan-1-ol Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 description 2
- VVCMGAUPZIKYTH-VGHSCWAPSA-N 2-acetyloxybenzoic acid;[(2s,3r)-4-(dimethylamino)-3-methyl-1,2-diphenylbutan-2-yl] propanoate;1,3,7-trimethylpurine-2,6-dione Chemical compound CC(=O)OC1=CC=CC=C1C(O)=O.CN1C(=O)N(C)C(=O)C2=C1N=CN2C.C([C@](OC(=O)CC)([C@H](C)CN(C)C)C=1C=CC=CC=1)C1=CC=CC=C1 VVCMGAUPZIKYTH-VGHSCWAPSA-N 0.000 description 2
- MIQCEWVIFALFRN-UHFFFAOYSA-N 2-amino-4-bromo-6-fluorobenzonitrile Chemical compound NC1=CC(Br)=CC(F)=C1C#N MIQCEWVIFALFRN-UHFFFAOYSA-N 0.000 description 2
- YSUIQYOGTINQIN-UZFYAQMZSA-N 2-amino-9-[(1S,6R,8R,9S,10R,15R,17R,18R)-8-(6-aminopurin-9-yl)-9,18-difluoro-3,12-dihydroxy-3,12-bis(sulfanylidene)-2,4,7,11,13,16-hexaoxa-3lambda5,12lambda5-diphosphatricyclo[13.2.1.06,10]octadecan-17-yl]-1H-purin-6-one Chemical compound NC1=NC2=C(N=CN2[C@@H]2O[C@@H]3COP(S)(=O)O[C@@H]4[C@@H](COP(S)(=O)O[C@@H]2[C@@H]3F)O[C@H]([C@H]4F)N2C=NC3=C2N=CN=C3N)C(=O)N1 YSUIQYOGTINQIN-UZFYAQMZSA-N 0.000 description 2
- IJFIDXAHOIPGGK-UHFFFAOYSA-N 2-chloro-5-(difluoromethyl)pyrimidine Chemical compound FC(F)C1=CN=C(Cl)N=C1 IJFIDXAHOIPGGK-UHFFFAOYSA-N 0.000 description 2
- 125000004777 2-fluoroethyl group Chemical group [H]C([H])(F)C([H])([H])* 0.000 description 2
- OSWFIVFLDKOXQC-UHFFFAOYSA-N 4-(3-methoxyphenyl)aniline Chemical compound COC1=CC=CC(C=2C=CC(N)=CC=2)=C1 OSWFIVFLDKOXQC-UHFFFAOYSA-N 0.000 description 2
- FCGBLTVXZYYMCD-UHFFFAOYSA-N 4-bromo-2-fluoro-5-nitrophenol Chemical compound OC1=CC([N+]([O-])=O)=C(Br)C=C1F FCGBLTVXZYYMCD-UHFFFAOYSA-N 0.000 description 2
- LCIMDZVLQQSHAK-UHFFFAOYSA-N 5-fluoroquinolin-6-amine Chemical compound Nc1ccc2ncccc2c1F LCIMDZVLQQSHAK-UHFFFAOYSA-N 0.000 description 2
- MHZKBGKRWWXAAN-UHFFFAOYSA-N 6-bromo-7-fluoro-1,2-benzothiazole Chemical compound Fc1c(Br)ccc2cnsc12 MHZKBGKRWWXAAN-UHFFFAOYSA-N 0.000 description 2
- XKRFYHLGVUSROY-UHFFFAOYSA-N Argon Chemical compound [Ar] XKRFYHLGVUSROY-UHFFFAOYSA-N 0.000 description 2
- 238000011729 BALB/c nude mouse Methods 0.000 description 2
- WKBOTKDWSSQWDR-UHFFFAOYSA-N Bromine atom Chemical group [Br] WKBOTKDWSSQWDR-UHFFFAOYSA-N 0.000 description 2
- JQUCWIWWWKZNCS-LESHARBVSA-N C(C1=CC=CC=C1)(=O)NC=1SC[C@H]2[C@@](N1)(CO[C@H](C2)C)C=2SC=C(N2)NC(=O)C2=NC=C(C=C2)OC(F)F Chemical compound C(C1=CC=CC=C1)(=O)NC=1SC[C@H]2[C@@](N1)(CO[C@H](C2)C)C=2SC=C(N2)NC(=O)C2=NC=C(C=C2)OC(F)F JQUCWIWWWKZNCS-LESHARBVSA-N 0.000 description 2
- SXSFCYFODZPWMN-UHFFFAOYSA-N CC(C)(C)OC(NC(C=CC1=C2SN=C1)=C2F)=O Chemical compound CC(C)(C)OC(NC(C=CC1=C2SN=C1)=C2F)=O SXSFCYFODZPWMN-UHFFFAOYSA-N 0.000 description 2
- BQXUPNKLZNSUMC-YUQWMIPFSA-N CCN(CCCCCOCC(=O)N[C@H](C(=O)N1C[C@H](O)C[C@H]1C(=O)N[C@@H](C)c1ccc(cc1)-c1scnc1C)C(C)(C)C)CCOc1ccc(cc1)C(=O)c1c(sc2cc(O)ccc12)-c1ccc(O)cc1 Chemical compound CCN(CCCCCOCC(=O)N[C@H](C(=O)N1C[C@H](O)C[C@H]1C(=O)N[C@@H](C)c1ccc(cc1)-c1scnc1C)C(C)(C)C)CCOc1ccc(cc1)C(=O)c1c(sc2cc(O)ccc12)-c1ccc(O)cc1 BQXUPNKLZNSUMC-YUQWMIPFSA-N 0.000 description 2
- WFSBUKVQYMFJDJ-MKMNVTDBSA-N CN(C)\C=N\C1=C(C#N)C(F)=CC(Br)=C1 Chemical compound CN(C)\C=N\C1=C(C#N)C(F)=CC(Br)=C1 WFSBUKVQYMFJDJ-MKMNVTDBSA-N 0.000 description 2
- BVKZGUZCCUSVTD-UHFFFAOYSA-L Carbonate Chemical compound [O-]C([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-L 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 2
- 229940127007 Compound 39 Drugs 0.000 description 2
- 229910021595 Copper(I) iodide Inorganic materials 0.000 description 2
- 229920002261 Corn starch Polymers 0.000 description 2
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 2
- 206010071975 EGFR gene mutation Diseases 0.000 description 2
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 2
- WMFOQBRAJBCJND-UHFFFAOYSA-M Lithium hydroxide Chemical compound [Li+].[OH-] WMFOQBRAJBCJND-UHFFFAOYSA-M 0.000 description 2
- LVDRREOUMKACNJ-BKMJKUGQSA-N N-[(2R,3S)-2-(4-chlorophenyl)-1-(1,4-dimethyl-2-oxoquinolin-7-yl)-6-oxopiperidin-3-yl]-2-methylpropane-1-sulfonamide Chemical compound CC(C)CS(=O)(=O)N[C@H]1CCC(=O)N([C@@H]1c1ccc(Cl)cc1)c1ccc2c(C)cc(=O)n(C)c2c1 LVDRREOUMKACNJ-BKMJKUGQSA-N 0.000 description 2
- WRIVZDGVZMCVSL-UHFFFAOYSA-N NC(C=CC1=C2SN=C1)=C2F Chemical compound NC(C=CC1=C2SN=C1)=C2F WRIVZDGVZMCVSL-UHFFFAOYSA-N 0.000 description 2
- QOVYHDHLFPKQQG-NDEPHWFRSA-N N[C@@H](CCC(=O)N1CCC(CC1)NC1=C2C=CC=CC2=NC(NCC2=CN(CCCNCCCNC3CCCCC3)N=N2)=N1)C(O)=O Chemical compound N[C@@H](CCC(=O)N1CCC(CC1)NC1=C2C=CC=CC2=NC(NCC2=CN(CCCNCCCNC3CCCCC3)N=N2)=N1)C(O)=O QOVYHDHLFPKQQG-NDEPHWFRSA-N 0.000 description 2
- 229910004878 Na2S2O4 Inorganic materials 0.000 description 2
- GRYLNZFGIOXLOG-UHFFFAOYSA-N Nitric acid Chemical compound O[N+]([O-])=O GRYLNZFGIOXLOG-UHFFFAOYSA-N 0.000 description 2
- 229910019213 POCl3 Inorganic materials 0.000 description 2
- WTKZEGDFNFYCGP-UHFFFAOYSA-N Pyrazole Chemical compound C=1C=NNC=1 WTKZEGDFNFYCGP-UHFFFAOYSA-N 0.000 description 2
- CDBYLPFSWZWCQE-UHFFFAOYSA-L Sodium Carbonate Chemical compound [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 2
- PXIPVTKHYLBLMZ-UHFFFAOYSA-N Sodium azide Chemical compound [Na+].[N-]=[N+]=[N-] PXIPVTKHYLBLMZ-UHFFFAOYSA-N 0.000 description 2
- GUGOEEXESWIERI-UHFFFAOYSA-N Terfenadine Chemical compound C1=CC(C(C)(C)C)=CC=C1C(O)CCCN1CCC(C(O)(C=2C=CC=CC=2)C=2C=CC=CC=2)CC1 GUGOEEXESWIERI-UHFFFAOYSA-N 0.000 description 2
- XTXRWKRVRITETP-UHFFFAOYSA-N Vinyl acetate Chemical compound CC(=O)OC=C XTXRWKRVRITETP-UHFFFAOYSA-N 0.000 description 2
- QHNSEQWCTMDSMV-UHFFFAOYSA-N [1-(difluoromethyl)pyrazol-3-yl]methanol Chemical compound OCC=1C=CN(C(F)F)N=1 QHNSEQWCTMDSMV-UHFFFAOYSA-N 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 230000037005 anaesthesia Effects 0.000 description 2
- 230000003698 anagen phase Effects 0.000 description 2
- 239000007900 aqueous suspension Substances 0.000 description 2
- 239000012300 argon atmosphere Substances 0.000 description 2
- XRWSZZJLZRKHHD-WVWIJVSJSA-N asunaprevir Chemical compound O=C([C@@H]1C[C@H](CN1C(=O)[C@@H](NC(=O)OC(C)(C)C)C(C)(C)C)OC1=NC=C(C2=CC=C(Cl)C=C21)OC)N[C@]1(C(=O)NS(=O)(=O)C2CC2)C[C@H]1C=C XRWSZZJLZRKHHD-WVWIJVSJSA-N 0.000 description 2
- 239000012298 atmosphere Substances 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 150000008359 benzonitriles Chemical class 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 125000001246 bromo group Chemical group Br* 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- HGXJOXHYPGNVNK-UHFFFAOYSA-N butane;ethenoxyethane;tin Chemical compound CCCC[Sn](CCCC)(CCCC)C(=C)OCC HGXJOXHYPGNVNK-UHFFFAOYSA-N 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 239000003054 catalyst Substances 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 230000005754 cellular signaling Effects 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 238000004440 column chromatography Methods 0.000 description 2
- 229940125904 compound 1 Drugs 0.000 description 2
- 229940125797 compound 12 Drugs 0.000 description 2
- 229940125758 compound 15 Drugs 0.000 description 2
- 229940126142 compound 16 Drugs 0.000 description 2
- 229940125961 compound 24 Drugs 0.000 description 2
- 229940125878 compound 36 Drugs 0.000 description 2
- 229940125936 compound 42 Drugs 0.000 description 2
- 229940125900 compound 59 Drugs 0.000 description 2
- DOBRDRYODQBAMW-UHFFFAOYSA-N copper(i) cyanide Chemical compound [Cu+].N#[C-] DOBRDRYODQBAMW-UHFFFAOYSA-N 0.000 description 2
- 239000008120 corn starch Substances 0.000 description 2
- 125000004122 cyclic group Chemical group 0.000 description 2
- NZZFYRREKKOMAT-UHFFFAOYSA-N diiodomethane Chemical compound ICI NZZFYRREKKOMAT-UHFFFAOYSA-N 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- ZUOUZKKEUPVFJK-UHFFFAOYSA-N diphenyl Chemical compound C1=CC=CC=C1C1=CC=CC=C1 ZUOUZKKEUPVFJK-UHFFFAOYSA-N 0.000 description 2
- 230000007783 downstream signaling Effects 0.000 description 2
- 239000000839 emulsion Substances 0.000 description 2
- ABCAYSKBEAULDO-UHFFFAOYSA-N ethyl 4-fluoro-5-[(2-methylpropan-2-yl)oxycarbonylamino]pyrazolo[1,5-a]pyridine-3-carboxylate Chemical compound C1=CC(NC(=O)OC(C)(C)C)=C(F)C2=C(C(=O)OCC)C=NN21 ABCAYSKBEAULDO-UHFFFAOYSA-N 0.000 description 2
- RIFGWPKJUGCATF-UHFFFAOYSA-N ethyl chloroformate Chemical compound CCOC(Cl)=O RIFGWPKJUGCATF-UHFFFAOYSA-N 0.000 description 2
- 239000012065 filter cake Substances 0.000 description 2
- 125000005842 heteroatom Chemical group 0.000 description 2
- 125000004415 heterocyclylalkyl group Chemical group 0.000 description 2
- 150000002430 hydrocarbons Chemical group 0.000 description 2
- 239000005457 ice water Substances 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 238000007917 intracranial administration Methods 0.000 description 2
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 2
- 239000008101 lactose Substances 0.000 description 2
- 239000010410 layer Substances 0.000 description 2
- KWGKDLIKAYFUFQ-UHFFFAOYSA-M lithium chloride Chemical compound [Li+].[Cl-] KWGKDLIKAYFUFQ-UHFFFAOYSA-M 0.000 description 2
- 235000019359 magnesium stearate Nutrition 0.000 description 2
- 229910052943 magnesium sulfate Inorganic materials 0.000 description 2
- NUJOXMJBOLGQSY-UHFFFAOYSA-N manganese dioxide Chemical compound O=[Mn]=O NUJOXMJBOLGQSY-UHFFFAOYSA-N 0.000 description 2
- UJCVCPISHPLHNC-UHFFFAOYSA-N methyl 1-(difluoromethyl)pyrazole-3-carboxylate Chemical compound COC(=O)C=1C=CN(C(F)F)N=1 UJCVCPISHPLHNC-UHFFFAOYSA-N 0.000 description 2
- UWOZODSMOOALPG-UHFFFAOYSA-N methyl 3-methylpyridine-4-carboxylate Chemical compound COC(=O)C1=CC=NC=C1C UWOZODSMOOALPG-UHFFFAOYSA-N 0.000 description 2
- 229910017604 nitric acid Inorganic materials 0.000 description 2
- 150000005181 nitrobenzenes Chemical class 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 125000002971 oxazolyl group Chemical group 0.000 description 2
- 229910052760 oxygen Inorganic materials 0.000 description 2
- HFPZCAJZSCWRBC-UHFFFAOYSA-N p-cymene Chemical compound CC(C)C1=CC=C(C)C=C1 HFPZCAJZSCWRBC-UHFFFAOYSA-N 0.000 description 2
- 230000036407 pain Effects 0.000 description 2
- 230000035515 penetration Effects 0.000 description 2
- 229910052698 phosphorus Inorganic materials 0.000 description 2
- 230000026731 phosphorylation Effects 0.000 description 2
- 238000006366 phosphorylation reaction Methods 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 238000000425 proton nuclear magnetic resonance spectrum Methods 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 239000012363 selectfluor Substances 0.000 description 2
- 238000000926 separation method Methods 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- MRTAVLDNYYEJHK-UHFFFAOYSA-M sodium;2-chloro-2,2-difluoroacetate Chemical compound [Na+].[O-]C(=O)C(F)(F)Cl MRTAVLDNYYEJHK-UHFFFAOYSA-M 0.000 description 2
- 239000012453 solvate Substances 0.000 description 2
- 125000001424 substituent group Chemical group 0.000 description 2
- 239000006228 supernatant Substances 0.000 description 2
- 238000001356 surgical procedure Methods 0.000 description 2
- 239000003826 tablet Substances 0.000 description 2
- 229960000351 terfenadine Drugs 0.000 description 2
- DYHSDKLCOJIUFX-UHFFFAOYSA-N tert-butoxycarbonyl anhydride Chemical compound CC(C)(C)OC(=O)OC(=O)OC(C)(C)C DYHSDKLCOJIUFX-UHFFFAOYSA-N 0.000 description 2
- YFDSDPIBEUFTMI-UHFFFAOYSA-N tribromoethanol Chemical compound OCC(Br)(Br)Br YFDSDPIBEUFTMI-UHFFFAOYSA-N 0.000 description 2
- 229950004616 tribromoethanol Drugs 0.000 description 2
- AQRLNPVMDITEJU-UHFFFAOYSA-N triethylsilane Chemical compound CC[SiH](CC)CC AQRLNPVMDITEJU-UHFFFAOYSA-N 0.000 description 2
- PYOKUURKVVELLB-UHFFFAOYSA-N trimethyl orthoformate Chemical compound COC(OC)OC PYOKUURKVVELLB-UHFFFAOYSA-N 0.000 description 2
- 210000004881 tumor cell Anatomy 0.000 description 2
- 229940121358 tyrosine kinase inhibitor Drugs 0.000 description 2
- 238000005406 washing Methods 0.000 description 2
- 238000005303 weighing Methods 0.000 description 2
- CXNIUSPIQKWYAI-UHFFFAOYSA-N xantphos Chemical compound C=12OC3=C(P(C=4C=CC=CC=4)C=4C=CC=CC=4)C=CC=C3C(C)(C)C2=CC=CC=1P(C=1C=CC=CC=1)C1=CC=CC=C1 CXNIUSPIQKWYAI-UHFFFAOYSA-N 0.000 description 2
- UAOUIVVJBYDFKD-XKCDOFEDSA-N (1R,9R,10S,11R,12R,15S,18S,21R)-10,11,21-trihydroxy-8,8-dimethyl-14-methylidene-4-(prop-2-enylamino)-20-oxa-5-thia-3-azahexacyclo[9.7.2.112,15.01,9.02,6.012,18]henicosa-2(6),3-dien-13-one Chemical compound C([C@@H]1[C@@H](O)[C@@]23C(C1=C)=O)C[C@H]2[C@]12C(N=C(NCC=C)S4)=C4CC(C)(C)[C@H]1[C@H](O)[C@]3(O)OC2 UAOUIVVJBYDFKD-XKCDOFEDSA-N 0.000 description 1
- AOSZTAHDEDLTLQ-AZKQZHLXSA-N (1S,2S,4R,8S,9S,11S,12R,13S,19S)-6-[(3-chlorophenyl)methyl]-12,19-difluoro-11-hydroxy-8-(2-hydroxyacetyl)-9,13-dimethyl-6-azapentacyclo[10.8.0.02,9.04,8.013,18]icosa-14,17-dien-16-one Chemical compound C([C@@H]1C[C@H]2[C@H]3[C@]([C@]4(C=CC(=O)C=C4[C@@H](F)C3)C)(F)[C@@H](O)C[C@@]2([C@@]1(C1)C(=O)CO)C)N1CC1=CC=CC(Cl)=C1 AOSZTAHDEDLTLQ-AZKQZHLXSA-N 0.000 description 1
- ABJSOROVZZKJGI-OCYUSGCXSA-N (1r,2r,4r)-2-(4-bromophenyl)-n-[(4-chlorophenyl)-(2-fluoropyridin-4-yl)methyl]-4-morpholin-4-ylcyclohexane-1-carboxamide Chemical compound C1=NC(F)=CC(C(NC(=O)[C@H]2[C@@H](C[C@@H](CC2)N2CCOCC2)C=2C=CC(Br)=CC=2)C=2C=CC(Cl)=CC=2)=C1 ABJSOROVZZKJGI-OCYUSGCXSA-N 0.000 description 1
- SZUVGFMDDVSKSI-WIFOCOSTSA-N (1s,2s,3s,5r)-1-(carboxymethyl)-3,5-bis[(4-phenoxyphenyl)methyl-propylcarbamoyl]cyclopentane-1,2-dicarboxylic acid Chemical compound O=C([C@@H]1[C@@H]([C@](CC(O)=O)([C@H](C(=O)N(CCC)CC=2C=CC(OC=3C=CC=CC=3)=CC=2)C1)C(O)=O)C(O)=O)N(CCC)CC(C=C1)=CC=C1OC1=CC=CC=C1 SZUVGFMDDVSKSI-WIFOCOSTSA-N 0.000 description 1
- IUSARDYWEPUTPN-OZBXUNDUSA-N (2r)-n-[(2s,3r)-4-[[(4s)-6-(2,2-dimethylpropyl)spiro[3,4-dihydropyrano[2,3-b]pyridine-2,1'-cyclobutane]-4-yl]amino]-3-hydroxy-1-[3-(1,3-thiazol-2-yl)phenyl]butan-2-yl]-2-methoxypropanamide Chemical compound C([C@H](NC(=O)[C@@H](C)OC)[C@H](O)CN[C@@H]1C2=CC(CC(C)(C)C)=CN=C2OC2(CCC2)C1)C(C=1)=CC=CC=1C1=NC=CS1 IUSARDYWEPUTPN-OZBXUNDUSA-N 0.000 description 1
- YJLIKUSWRSEPSM-WGQQHEPDSA-N (2r,3r,4s,5r)-2-[6-amino-8-[(4-phenylphenyl)methylamino]purin-9-yl]-5-(hydroxymethyl)oxolane-3,4-diol Chemical compound C=1C=C(C=2C=CC=CC=2)C=CC=1CNC1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O YJLIKUSWRSEPSM-WGQQHEPDSA-N 0.000 description 1
- VIJSPAIQWVPKQZ-BLECARSGSA-N (2s)-2-[[(2s)-2-[[(2s)-2-[[(2s)-2-[[(2s)-2-[[(2s)-2-acetamido-5-(diaminomethylideneamino)pentanoyl]amino]-4-methylpentanoyl]amino]-4,4-dimethylpentanoyl]amino]-4-methylpentanoyl]amino]propanoyl]amino]-5-(diaminomethylideneamino)pentanoic acid Chemical compound NC(=N)NCCC[C@@H](C(O)=O)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(C)(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCNC(N)=N)NC(C)=O VIJSPAIQWVPKQZ-BLECARSGSA-N 0.000 description 1
- WWTBZEKOSBFBEM-SPWPXUSOSA-N (2s)-2-[[2-benzyl-3-[hydroxy-[(1r)-2-phenyl-1-(phenylmethoxycarbonylamino)ethyl]phosphoryl]propanoyl]amino]-3-(1h-indol-3-yl)propanoic acid Chemical compound N([C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)O)C(=O)C(CP(O)(=O)[C@H](CC=1C=CC=CC=1)NC(=O)OCC=1C=CC=CC=1)CC1=CC=CC=C1 WWTBZEKOSBFBEM-SPWPXUSOSA-N 0.000 description 1
- QFLWZFQWSBQYPS-AWRAUJHKSA-N (3S)-3-[[(2S)-2-[[(2S)-2-[5-[(3aS,6aR)-2-oxo-1,3,3a,4,6,6a-hexahydrothieno[3,4-d]imidazol-4-yl]pentanoylamino]-3-methylbutanoyl]amino]-3-(4-hydroxyphenyl)propanoyl]amino]-4-[1-bis(4-chlorophenoxy)phosphorylbutylamino]-4-oxobutanoic acid Chemical compound CCCC(NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](Cc1ccc(O)cc1)NC(=O)[C@@H](NC(=O)CCCCC1SC[C@@H]2NC(=O)N[C@H]12)C(C)C)P(=O)(Oc1ccc(Cl)cc1)Oc1ccc(Cl)cc1 QFLWZFQWSBQYPS-AWRAUJHKSA-N 0.000 description 1
- IWZSHWBGHQBIML-ZGGLMWTQSA-N (3S,8S,10R,13S,14S,17S)-17-isoquinolin-7-yl-N,N,10,13-tetramethyl-2,3,4,7,8,9,11,12,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-3-amine Chemical compound CN(C)[C@H]1CC[C@]2(C)C3CC[C@@]4(C)[C@@H](CC[C@@H]4c4ccc5ccncc5c4)[C@@H]3CC=C2C1 IWZSHWBGHQBIML-ZGGLMWTQSA-N 0.000 description 1
- HUWSZNZAROKDRZ-RRLWZMAJSA-N (3r,4r)-3-azaniumyl-5-[[(2s,3r)-1-[(2s)-2,3-dicarboxypyrrolidin-1-yl]-3-methyl-1-oxopentan-2-yl]amino]-5-oxo-4-sulfanylpentane-1-sulfonate Chemical compound OS(=O)(=O)CC[C@@H](N)[C@@H](S)C(=O)N[C@@H]([C@H](C)CC)C(=O)N1CCC(C(O)=O)[C@H]1C(O)=O HUWSZNZAROKDRZ-RRLWZMAJSA-N 0.000 description 1
- MPDDTAJMJCESGV-CTUHWIOQSA-M (3r,5r)-7-[2-(4-fluorophenyl)-5-[methyl-[(1r)-1-phenylethyl]carbamoyl]-4-propan-2-ylpyrazol-3-yl]-3,5-dihydroxyheptanoate Chemical compound C1([C@@H](C)N(C)C(=O)C2=NN(C(CC[C@@H](O)C[C@@H](O)CC([O-])=O)=C2C(C)C)C=2C=CC(F)=CC=2)=CC=CC=C1 MPDDTAJMJCESGV-CTUHWIOQSA-M 0.000 description 1
- YQOLEILXOBUDMU-KRWDZBQOSA-N (4R)-5-[(6-bromo-3-methyl-2-pyrrolidin-1-ylquinoline-4-carbonyl)amino]-4-(2-chlorophenyl)pentanoic acid Chemical compound CC1=C(C2=C(C=CC(=C2)Br)N=C1N3CCCC3)C(=O)NC[C@H](CCC(=O)O)C4=CC=CC=C4Cl YQOLEILXOBUDMU-KRWDZBQOSA-N 0.000 description 1
- DYLIWHYUXAJDOJ-OWOJBTEDSA-N (e)-4-(6-aminopurin-9-yl)but-2-en-1-ol Chemical compound NC1=NC=NC2=C1N=CN2C\C=C\CO DYLIWHYUXAJDOJ-OWOJBTEDSA-N 0.000 description 1
- SKAFKZTYRHXQIT-UHFFFAOYSA-N 1,2-thiazol-5-ylboronic acid Chemical compound OB(O)C1=CC=NS1 SKAFKZTYRHXQIT-UHFFFAOYSA-N 0.000 description 1
- KQZLRWGGWXJPOS-NLFPWZOASA-N 1-[(1R)-1-(2,4-dichlorophenyl)ethyl]-6-[(4S,5R)-4-[(2S)-2-(hydroxymethyl)pyrrolidin-1-yl]-5-methylcyclohexen-1-yl]pyrazolo[3,4-b]pyrazine-3-carbonitrile Chemical compound ClC1=C(C=CC(=C1)Cl)[C@@H](C)N1N=C(C=2C1=NC(=CN=2)C1=CC[C@@H]([C@@H](C1)C)N1[C@@H](CCC1)CO)C#N KQZLRWGGWXJPOS-NLFPWZOASA-N 0.000 description 1
- WZZBNLYBHUDSHF-DHLKQENFSA-N 1-[(3s,4s)-4-[8-(2-chloro-4-pyrimidin-2-yloxyphenyl)-7-fluoro-2-methylimidazo[4,5-c]quinolin-1-yl]-3-fluoropiperidin-1-yl]-2-hydroxyethanone Chemical compound CC1=NC2=CN=C3C=C(F)C(C=4C(=CC(OC=5N=CC=CN=5)=CC=4)Cl)=CC3=C2N1[C@H]1CCN(C(=O)CO)C[C@@H]1F WZZBNLYBHUDSHF-DHLKQENFSA-N 0.000 description 1
- UNILWMWFPHPYOR-KXEYIPSPSA-M 1-[6-[2-[3-[3-[3-[2-[2-[3-[[2-[2-[[(2r)-1-[[2-[[(2r)-1-[3-[2-[2-[3-[[2-(2-amino-2-oxoethoxy)acetyl]amino]propoxy]ethoxy]ethoxy]propylamino]-3-hydroxy-1-oxopropan-2-yl]amino]-2-oxoethyl]amino]-3-[(2r)-2,3-di(hexadecanoyloxy)propyl]sulfanyl-1-oxopropan-2-yl Chemical compound O=C1C(SCCC(=O)NCCCOCCOCCOCCCNC(=O)COCC(=O)N[C@@H](CSC[C@@H](COC(=O)CCCCCCCCCCCCCCC)OC(=O)CCCCCCCCCCCCCCC)C(=O)NCC(=O)N[C@H](CO)C(=O)NCCCOCCOCCOCCCNC(=O)COCC(N)=O)CC(=O)N1CCNC(=O)CCCCCN\1C2=CC=C(S([O-])(=O)=O)C=C2CC/1=C/C=C/C=C/C1=[N+](CC)C2=CC=C(S([O-])(=O)=O)C=C2C1 UNILWMWFPHPYOR-KXEYIPSPSA-M 0.000 description 1
- QRADKVYIJIAENZ-UHFFFAOYSA-N 1-[[bromo(difluoro)methyl]-ethoxyphosphoryl]oxyethane Chemical compound CCOP(=O)(C(F)(F)Br)OCC QRADKVYIJIAENZ-UHFFFAOYSA-N 0.000 description 1
- HUSPSWKWFREKSS-UHFFFAOYSA-N 1-bromo-4-(difluoromethyl)benzene Chemical compound FC(F)C1=CC=C(Br)C=C1 HUSPSWKWFREKSS-UHFFFAOYSA-N 0.000 description 1
- BSSKDKZDQARXEH-UHFFFAOYSA-N 1-bromo-5-fluoro-4-methoxy-2-nitrobenzene Chemical compound COC1=CC([N+]([O-])=O)=C(Br)C=C1F BSSKDKZDQARXEH-UHFFFAOYSA-N 0.000 description 1
- FQASUNMXKLPOOF-UHFFFAOYSA-N 1-methylpyrazol-4-ol Chemical compound CN1C=C(O)C=N1 FQASUNMXKLPOOF-UHFFFAOYSA-N 0.000 description 1
- 125000001637 1-naphthyl group Chemical group [H]C1=C([H])C([H])=C2C(*)=C([H])C([H])=C([H])C2=C1[H] 0.000 description 1
- YBYIRNPNPLQARY-UHFFFAOYSA-N 1H-indene Natural products C1=CC=C2CC=CC2=C1 YBYIRNPNPLQARY-UHFFFAOYSA-N 0.000 description 1
- LRGBDJBDJXZTTD-UHFFFAOYSA-N 1h-pyrazole-4-carbaldehyde Chemical compound O=CC=1C=NNC=1 LRGBDJBDJXZTTD-UHFFFAOYSA-N 0.000 description 1
- 125000004206 2,2,2-trifluoroethyl group Chemical group [H]C([H])(*)C(F)(F)F 0.000 description 1
- 125000004778 2,2-difluoroethyl group Chemical group [H]C([H])(*)C([H])(F)F 0.000 description 1
- PYRKKGOKRMZEIT-UHFFFAOYSA-N 2-[6-(2-cyclopropylethoxy)-9-(2-hydroxy-2-methylpropyl)-1h-phenanthro[9,10-d]imidazol-2-yl]-5-fluorobenzene-1,3-dicarbonitrile Chemical compound C1=C2C3=CC(CC(C)(O)C)=CC=C3C=3NC(C=4C(=CC(F)=CC=4C#N)C#N)=NC=3C2=CC=C1OCCC1CC1 PYRKKGOKRMZEIT-UHFFFAOYSA-N 0.000 description 1
- FMKGJQHNYMWDFJ-CVEARBPZSA-N 2-[[4-(2,2-difluoropropoxy)pyrimidin-5-yl]methylamino]-4-[[(1R,4S)-4-hydroxy-3,3-dimethylcyclohexyl]amino]pyrimidine-5-carbonitrile Chemical compound FC(COC1=NC=NC=C1CNC1=NC=C(C(=N1)N[C@H]1CC([C@H](CC1)O)(C)C)C#N)(C)F FMKGJQHNYMWDFJ-CVEARBPZSA-N 0.000 description 1
- LJYQVOPFBNMTKJ-UHFFFAOYSA-N 2-chloropyrimidine-5-carbaldehyde Chemical compound ClC1=NC=C(C=O)C=N1 LJYQVOPFBNMTKJ-UHFFFAOYSA-N 0.000 description 1
- AXDGIPMYJALRKV-UHFFFAOYSA-N 2-iodopyrimidine Chemical compound IC1=NC=CC=N1 AXDGIPMYJALRKV-UHFFFAOYSA-N 0.000 description 1
- LFOIDLOIBZFWDO-UHFFFAOYSA-N 2-methoxy-6-[6-methoxy-4-[(3-phenylmethoxyphenyl)methoxy]-1-benzofuran-2-yl]imidazo[2,1-b][1,3,4]thiadiazole Chemical compound N1=C2SC(OC)=NN2C=C1C(OC1=CC(OC)=C2)=CC1=C2OCC(C=1)=CC=CC=1OCC1=CC=CC=C1 LFOIDLOIBZFWDO-UHFFFAOYSA-N 0.000 description 1
- 125000001622 2-naphthyl group Chemical group [H]C1=C([H])C([H])=C2C([H])=C(*)C([H])=C([H])C2=C1[H] 0.000 description 1
- NPAXPTHCUCUHPT-UHFFFAOYSA-N 3,4,7,8-tetramethyl-1,10-phenanthroline Chemical compound CC1=CN=C2C3=NC=C(C)C(C)=C3C=CC2=C1C NPAXPTHCUCUHPT-UHFFFAOYSA-N 0.000 description 1
- QBWKPGNFQQJGFY-QLFBSQMISA-N 3-[(1r)-1-[(2r,6s)-2,6-dimethylmorpholin-4-yl]ethyl]-n-[6-methyl-3-(1h-pyrazol-4-yl)imidazo[1,2-a]pyrazin-8-yl]-1,2-thiazol-5-amine Chemical compound N1([C@H](C)C2=NSC(NC=3C4=NC=C(N4C=C(C)N=3)C3=CNN=C3)=C2)C[C@H](C)O[C@H](C)C1 QBWKPGNFQQJGFY-QLFBSQMISA-N 0.000 description 1
- BGAJNPLDJJBRHK-UHFFFAOYSA-N 3-[2-[5-(3-chloro-4-propan-2-yloxyphenyl)-1,3,4-thiadiazol-2-yl]-3-methyl-6,7-dihydro-4h-pyrazolo[4,3-c]pyridin-5-yl]propanoic acid Chemical compound C1=C(Cl)C(OC(C)C)=CC=C1C1=NN=C(N2C(=C3CN(CCC(O)=O)CCC3=N2)C)S1 BGAJNPLDJJBRHK-UHFFFAOYSA-N 0.000 description 1
- UFIKBUVVVGSMGW-UHFFFAOYSA-N 3-fluoropyridin-4-amine Chemical compound NC1=CC=NC=C1F UFIKBUVVVGSMGW-UHFFFAOYSA-N 0.000 description 1
- OSMAGAVKVRGYGR-UHFFFAOYSA-N 3-methylpyridine-4-carboxylic acid Chemical compound CC1=CN=CC=C1C(O)=O OSMAGAVKVRGYGR-UHFFFAOYSA-N 0.000 description 1
- VZBZUFGPXJGXNJ-UHFFFAOYSA-N 4-(3-chloro-2-fluoroanilino)-7-methoxyquinazolin-6-ol Chemical compound C=12C=C(O)C(OC)=CC2=NC=NC=1NC1=CC=CC(Cl)=C1F VZBZUFGPXJGXNJ-UHFFFAOYSA-N 0.000 description 1
- WYFCZWSWFGJODV-MIANJLSGSA-N 4-[[(1s)-2-[(e)-3-[3-chloro-2-fluoro-6-(tetrazol-1-yl)phenyl]prop-2-enoyl]-5-(4-methyl-2-oxopiperazin-1-yl)-3,4-dihydro-1h-isoquinoline-1-carbonyl]amino]benzoic acid Chemical compound O=C1CN(C)CCN1C1=CC=CC2=C1CCN(C(=O)\C=C\C=1C(=CC=C(Cl)C=1F)N1N=NN=C1)[C@@H]2C(=O)NC1=CC=C(C(O)=O)C=C1 WYFCZWSWFGJODV-MIANJLSGSA-N 0.000 description 1
- IMOLPSNRLZLWQR-UHFFFAOYSA-N 4-bromo-2,3-difluorobenzaldehyde Chemical compound FC1=C(F)C(C=O)=CC=C1Br IMOLPSNRLZLWQR-UHFFFAOYSA-N 0.000 description 1
- RYVOZMPTISNBDB-UHFFFAOYSA-N 4-bromo-2-fluorophenol Chemical compound OC1=CC=C(Br)C=C1F RYVOZMPTISNBDB-UHFFFAOYSA-N 0.000 description 1
- XFJBGINZIMNZBW-CRAIPNDOSA-N 5-chloro-2-[4-[(1r,2s)-2-[2-(5-methylsulfonylpyridin-2-yl)oxyethyl]cyclopropyl]piperidin-1-yl]pyrimidine Chemical compound N1=CC(S(=O)(=O)C)=CC=C1OCC[C@H]1[C@@H](C2CCN(CC2)C=2N=CC(Cl)=CN=2)C1 XFJBGINZIMNZBW-CRAIPNDOSA-N 0.000 description 1
- 125000004938 5-pyridyl group Chemical group N1=CC=CC(=C1)* 0.000 description 1
- HCCNBKFJYUWLEX-UHFFFAOYSA-N 7-(6-methoxypyridin-3-yl)-1-(2-propoxyethyl)-3-(pyrazin-2-ylmethylamino)pyrido[3,4-b]pyrazin-2-one Chemical compound O=C1N(CCOCCC)C2=CC(C=3C=NC(OC)=CC=3)=NC=C2N=C1NCC1=CN=CC=N1 HCCNBKFJYUWLEX-UHFFFAOYSA-N 0.000 description 1
- 208000031261 Acute myeloid leukaemia Diseases 0.000 description 1
- ATRRKUHOCOJYRX-UHFFFAOYSA-N Ammonium bicarbonate Chemical compound [NH4+].OC([O-])=O ATRRKUHOCOJYRX-UHFFFAOYSA-N 0.000 description 1
- 229910000013 Ammonium bicarbonate Inorganic materials 0.000 description 1
- VHUUQVKOLVNVRT-UHFFFAOYSA-N Ammonium hydroxide Chemical compound [NH4+].[OH-] VHUUQVKOLVNVRT-UHFFFAOYSA-N 0.000 description 1
- 101100404726 Arabidopsis thaliana NHX7 gene Proteins 0.000 description 1
- 208000010839 B-cell chronic lymphocytic leukemia Diseases 0.000 description 1
- 206010005003 Bladder cancer Diseases 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- FERIUCNNQQJTOY-UHFFFAOYSA-N Butyric acid Natural products CCCC(O)=O FERIUCNNQQJTOY-UHFFFAOYSA-N 0.000 description 1
- 206010008342 Cervix carcinoma Diseases 0.000 description 1
- 206010009944 Colon cancer Diseases 0.000 description 1
- 208000001333 Colorectal Neoplasms Diseases 0.000 description 1
- 229940126657 Compound 17 Drugs 0.000 description 1
- 229940126639 Compound 33 Drugs 0.000 description 1
- XFXPMWWXUTWYJX-UHFFFAOYSA-N Cyanide Chemical compound N#[C-] XFXPMWWXUTWYJX-UHFFFAOYSA-N 0.000 description 1
- KDXKERNSBIXSRK-RXMQYKEDSA-N D-lysine Chemical compound NCCCC[C@@H](N)C(O)=O KDXKERNSBIXSRK-RXMQYKEDSA-N 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 206010014733 Endometrial cancer Diseases 0.000 description 1
- 206010014759 Endometrial neoplasm Diseases 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 208000000461 Esophageal Neoplasms Diseases 0.000 description 1
- SEMPRRKRMFNDHU-UHFFFAOYSA-N FC1=C2C(NC3=CC=C4N=CC=CC4=C3F)=NC=NC2=CC(Br)=C1 Chemical compound FC1=C2C(NC3=CC=C4N=CC=CC4=C3F)=NC=NC2=CC(Br)=C1 SEMPRRKRMFNDHU-UHFFFAOYSA-N 0.000 description 1
- 108091008794 FGF receptors Proteins 0.000 description 1
- KRHYYFGTRYWZRS-UHFFFAOYSA-N Fluorane Chemical compound F KRHYYFGTRYWZRS-UHFFFAOYSA-N 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 1
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 1
- 206010069755 K-ras gene mutation Diseases 0.000 description 1
- 208000008839 Kidney Neoplasms Diseases 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- 208000031671 Large B-Cell Diffuse Lymphoma Diseases 0.000 description 1
- 108060001084 Luciferase Proteins 0.000 description 1
- 239000005089 Luciferase Substances 0.000 description 1
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 description 1
- 206010064912 Malignant transformation Diseases 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 206010059282 Metastases to central nervous system Diseases 0.000 description 1
- 238000006751 Mitsunobu reaction Methods 0.000 description 1
- 208000034578 Multiple myelomas Diseases 0.000 description 1
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 description 1
- OPFJDXRVMFKJJO-ZHHKINOHSA-N N-{[3-(2-benzamido-4-methyl-1,3-thiazol-5-yl)-pyrazol-5-yl]carbonyl}-G-dR-G-dD-dD-dD-NH2 Chemical compound S1C(C=2NN=C(C=2)C(=O)NCC(=O)N[C@H](CCCN=C(N)N)C(=O)NCC(=O)N[C@H](CC(O)=O)C(=O)N[C@H](CC(O)=O)C(=O)N[C@H](CC(O)=O)C(N)=O)=C(C)N=C1NC(=O)C1=CC=CC=C1 OPFJDXRVMFKJJO-ZHHKINOHSA-N 0.000 description 1
- 238000005481 NMR spectroscopy Methods 0.000 description 1
- JCXJVPUVTGWSNB-UHFFFAOYSA-N Nitrogen dioxide Chemical compound O=[N]=O JCXJVPUVTGWSNB-UHFFFAOYSA-N 0.000 description 1
- 206010030155 Oesophageal carcinoma Diseases 0.000 description 1
- 206010033128 Ovarian cancer Diseases 0.000 description 1
- 206010061535 Ovarian neoplasm Diseases 0.000 description 1
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 1
- QGMRQYFBGABWDR-UHFFFAOYSA-M Pentobarbital sodium Chemical compound [Na+].CCCC(C)C1(CC)C(=O)NC(=O)[N-]C1=O QGMRQYFBGABWDR-UHFFFAOYSA-M 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 229920002565 Polyethylene Glycol 400 Polymers 0.000 description 1
- 102100033237 Pro-epidermal growth factor Human genes 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 1
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 1
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 1
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 description 1
- 102100029986 Receptor tyrosine-protein kinase erbB-3 Human genes 0.000 description 1
- 101710100969 Receptor tyrosine-protein kinase erbB-3 Proteins 0.000 description 1
- 102100029981 Receptor tyrosine-protein kinase erbB-4 Human genes 0.000 description 1
- 101710100963 Receptor tyrosine-protein kinase erbB-4 Proteins 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- 108700008625 Reporter Genes Proteins 0.000 description 1
- 102000057028 SOS1 Human genes 0.000 description 1
- 108700022176 SOS1 Proteins 0.000 description 1
- 101100197320 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) RPL35A gene Proteins 0.000 description 1
- PNUZDKCDAWUEGK-CYZMBNFOSA-N Sitafloxacin Chemical compound C([C@H]1N)N(C=2C(=C3C(C(C(C(O)=O)=CN3[C@H]3[C@H](C3)F)=O)=CC=2F)Cl)CC11CC1 PNUZDKCDAWUEGK-CYZMBNFOSA-N 0.000 description 1
- KEAYESYHFKHZAL-UHFFFAOYSA-N Sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 1
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 1
- 101150100839 Sos1 gene Proteins 0.000 description 1
- 208000000102 Squamous Cell Carcinoma of Head and Neck Diseases 0.000 description 1
- 208000005718 Stomach Neoplasms Diseases 0.000 description 1
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 208000024770 Thyroid neoplasm Diseases 0.000 description 1
- OKJPEAGHQZHRQV-UHFFFAOYSA-N Triiodomethane Natural products IC(I)I OKJPEAGHQZHRQV-UHFFFAOYSA-N 0.000 description 1
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 description 1
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 description 1
- 208000002495 Uterine Neoplasms Diseases 0.000 description 1
- QYKIQEUNHZKYBP-UHFFFAOYSA-N Vinyl ether Chemical compound C=COC=C QYKIQEUNHZKYBP-UHFFFAOYSA-N 0.000 description 1
- LJOOWESTVASNOG-UFJKPHDISA-N [(1s,3r,4ar,7s,8s,8as)-3-hydroxy-8-[2-[(4r)-4-hydroxy-6-oxooxan-2-yl]ethyl]-7-methyl-1,2,3,4,4a,7,8,8a-octahydronaphthalen-1-yl] (2s)-2-methylbutanoate Chemical compound C([C@H]1[C@@H](C)C=C[C@H]2C[C@@H](O)C[C@@H]([C@H]12)OC(=O)[C@@H](C)CC)CC1C[C@@H](O)CC(=O)O1 LJOOWESTVASNOG-UFJKPHDISA-N 0.000 description 1
- SPXSEZMVRJLHQG-XMMPIXPASA-N [(2R)-1-[[4-[(3-phenylmethoxyphenoxy)methyl]phenyl]methyl]pyrrolidin-2-yl]methanol Chemical compound C(C1=CC=CC=C1)OC=1C=C(OCC2=CC=C(CN3[C@H](CCC3)CO)C=C2)C=CC=1 SPXSEZMVRJLHQG-XMMPIXPASA-N 0.000 description 1
- LNUFLCYMSVYYNW-ZPJMAFJPSA-N [(2r,3r,4s,5r,6r)-2-[(2r,3r,4s,5r,6r)-6-[(2r,3r,4s,5r,6r)-6-[(2r,3r,4s,5r,6r)-6-[[(3s,5s,8r,9s,10s,13r,14s,17r)-10,13-dimethyl-17-[(2r)-6-methylheptan-2-yl]-2,3,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydro-1h-cyclopenta[a]phenanthren-3-yl]oxy]-4,5-disulfo Chemical compound O([C@@H]1[C@@H](COS(O)(=O)=O)O[C@@H]([C@@H]([C@H]1OS(O)(=O)=O)OS(O)(=O)=O)O[C@@H]1[C@@H](COS(O)(=O)=O)O[C@@H]([C@@H]([C@H]1OS(O)(=O)=O)OS(O)(=O)=O)O[C@@H]1[C@@H](COS(O)(=O)=O)O[C@H]([C@@H]([C@H]1OS(O)(=O)=O)OS(O)(=O)=O)O[C@@H]1C[C@@H]2CC[C@H]3[C@@H]4CC[C@@H]([C@]4(CC[C@@H]3[C@@]2(C)CC1)C)[C@H](C)CCCC(C)C)[C@H]1O[C@H](COS(O)(=O)=O)[C@@H](OS(O)(=O)=O)[C@H](OS(O)(=O)=O)[C@H]1OS(O)(=O)=O LNUFLCYMSVYYNW-ZPJMAFJPSA-N 0.000 description 1
- PSLUFJFHTBIXMW-WYEYVKMPSA-N [(3r,4ar,5s,6s,6as,10s,10ar,10bs)-3-ethenyl-10,10b-dihydroxy-3,4a,7,7,10a-pentamethyl-1-oxo-6-(2-pyridin-2-ylethylcarbamoyloxy)-5,6,6a,8,9,10-hexahydro-2h-benzo[f]chromen-5-yl] acetate Chemical compound O([C@@H]1[C@@H]([C@]2(O[C@](C)(CC(=O)[C@]2(O)[C@@]2(C)[C@@H](O)CCC(C)(C)[C@@H]21)C=C)C)OC(=O)C)C(=O)NCCC1=CC=CC=N1 PSLUFJFHTBIXMW-WYEYVKMPSA-N 0.000 description 1
- SMNRFWMNPDABKZ-WVALLCKVSA-N [[(2R,3S,4R,5S)-5-(2,6-dioxo-3H-pyridin-3-yl)-3,4-dihydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl] [[[(2R,3S,4S,5R,6R)-4-fluoro-3,5-dihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-hydroxyphosphoryl]oxy-hydroxyphosphoryl] hydrogen phosphate Chemical compound OC[C@H]1O[C@H](OP(O)(=O)OP(O)(=O)OP(O)(=O)OP(O)(=O)OC[C@H]2O[C@H]([C@H](O)[C@@H]2O)C2C=CC(=O)NC2=O)[C@H](O)[C@@H](F)[C@@H]1O SMNRFWMNPDABKZ-WVALLCKVSA-N 0.000 description 1
- 229940124532 absorption promoter Drugs 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 125000004442 acylamino group Chemical group 0.000 description 1
- 150000001335 aliphatic alkanes Chemical class 0.000 description 1
- 125000003342 alkenyl group Chemical group 0.000 description 1
- 125000005090 alkenylcarbonyl group Chemical group 0.000 description 1
- 125000004453 alkoxycarbonyl group Chemical group 0.000 description 1
- 125000003282 alkyl amino group Chemical group 0.000 description 1
- 125000004448 alkyl carbonyl group Chemical group 0.000 description 1
- 125000004390 alkyl sulfonyl group Chemical group 0.000 description 1
- 125000000304 alkynyl group Chemical group 0.000 description 1
- 125000003368 amide group Chemical group 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 125000004103 aminoalkyl group Chemical group 0.000 description 1
- 235000012538 ammonium bicarbonate Nutrition 0.000 description 1
- 239000001099 ammonium carbonate Substances 0.000 description 1
- 239000000908 ammonium hydroxide Substances 0.000 description 1
- 230000003444 anaesthetic effect Effects 0.000 description 1
- 230000033115 angiogenesis Effects 0.000 description 1
- 230000006909 anti-apoptosis Effects 0.000 description 1
- 230000001857 anti-mycotic effect Effects 0.000 description 1
- 239000002543 antimycotic Substances 0.000 description 1
- 229910052786 argon Inorganic materials 0.000 description 1
- 125000001769 aryl amino group Chemical group 0.000 description 1
- 125000003725 azepanyl group Chemical group 0.000 description 1
- 125000002393 azetidinyl group Chemical group 0.000 description 1
- 125000004069 aziridinyl group Chemical group 0.000 description 1
- 125000003785 benzimidazolyl group Chemical group N1=C(NC2=C1C=CC=C2)* 0.000 description 1
- 125000000499 benzofuranyl group Chemical group O1C(=CC2=C1C=CC=C2)* 0.000 description 1
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 1
- 125000001164 benzothiazolyl group Chemical group S1C(=NC2=C1C=CC=C2)* 0.000 description 1
- 125000004196 benzothienyl group Chemical group S1C(=CC2=C1C=CC=C2)* 0.000 description 1
- 125000004541 benzoxazolyl group Chemical group O1C(=NC2=C1C=CC=C2)* 0.000 description 1
- 235000019445 benzyl alcohol Nutrition 0.000 description 1
- KGNDCEVUMONOKF-UGPLYTSKSA-N benzyl n-[(2r)-1-[(2s,4r)-2-[[(2s)-6-amino-1-(1,3-benzoxazol-2-yl)-1,1-dihydroxyhexan-2-yl]carbamoyl]-4-[(4-methylphenyl)methoxy]pyrrolidin-1-yl]-1-oxo-4-phenylbutan-2-yl]carbamate Chemical compound C1=CC(C)=CC=C1CO[C@H]1CN(C(=O)[C@@H](CCC=2C=CC=CC=2)NC(=O)OCC=2C=CC=CC=2)[C@H](C(=O)N[C@@H](CCCCN)C(O)(O)C=2OC3=CC=CC=C3N=2)C1 KGNDCEVUMONOKF-UGPLYTSKSA-N 0.000 description 1
- 238000004166 bioassay Methods 0.000 description 1
- 239000004305 biphenyl Substances 0.000 description 1
- 235000010290 biphenyl Nutrition 0.000 description 1
- 238000006664 bond formation reaction Methods 0.000 description 1
- 210000000133 brain stem Anatomy 0.000 description 1
- 210000005013 brain tissue Anatomy 0.000 description 1
- 125000003917 carbamoyl group Chemical group [H]N([H])C(*)=O 0.000 description 1
- 125000000609 carbazolyl group Chemical group C1(=CC=CC=2C3=CC=CC=C3NC12)* 0.000 description 1
- 235000011089 carbon dioxide Nutrition 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 230000003197 catalytic effect Effects 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 235000010980 cellulose Nutrition 0.000 description 1
- 210000003169 central nervous system Anatomy 0.000 description 1
- 201000010881 cervical cancer Diseases 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 208000006990 cholangiocarcinoma Diseases 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- 208000032852 chronic lymphocytic leukemia Diseases 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 229940125773 compound 10 Drugs 0.000 description 1
- 229940126543 compound 14 Drugs 0.000 description 1
- 229940125782 compound 2 Drugs 0.000 description 1
- 229940125810 compound 20 Drugs 0.000 description 1
- 229940126086 compound 21 Drugs 0.000 description 1
- 229940126208 compound 22 Drugs 0.000 description 1
- 229940125833 compound 23 Drugs 0.000 description 1
- 229940125846 compound 25 Drugs 0.000 description 1
- 229940127204 compound 29 Drugs 0.000 description 1
- 229940126214 compound 3 Drugs 0.000 description 1
- 229940125877 compound 31 Drugs 0.000 description 1
- 229940125807 compound 37 Drugs 0.000 description 1
- 229940127573 compound 38 Drugs 0.000 description 1
- 229940126540 compound 41 Drugs 0.000 description 1
- 229940125844 compound 46 Drugs 0.000 description 1
- 229940127271 compound 49 Drugs 0.000 description 1
- 229940125898 compound 5 Drugs 0.000 description 1
- 229940126545 compound 53 Drugs 0.000 description 1
- 229940127113 compound 57 Drugs 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- LSXDOTMGLUJQCM-UHFFFAOYSA-M copper(i) iodide Chemical compound I[Cu] LSXDOTMGLUJQCM-UHFFFAOYSA-M 0.000 description 1
- 238000002425 crystallisation Methods 0.000 description 1
- 230000008025 crystallization Effects 0.000 description 1
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 125000000582 cycloheptyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000000596 cyclohexenyl group Chemical group C1(=CCCCC1)* 0.000 description 1
- 125000000113 cyclohexyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000000640 cyclooctyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- NXQGGXCHGDYOHB-UHFFFAOYSA-L cyclopenta-1,4-dien-1-yl(diphenyl)phosphane;dichloropalladium;iron(2+) Chemical compound [Fe+2].Cl[Pd]Cl.[CH-]1C=CC(P(C=2C=CC=CC=2)C=2C=CC=CC=2)=C1.[CH-]1C=CC(P(C=2C=CC=CC=2)C=2C=CC=CC=2)=C1 NXQGGXCHGDYOHB-UHFFFAOYSA-L 0.000 description 1
- 125000002433 cyclopentenyl group Chemical group C1(=CCCC1)* 0.000 description 1
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- JMYVMOUINOAAPA-UHFFFAOYSA-N cyclopropanecarbaldehyde Chemical compound O=CC1CC1 JMYVMOUINOAAPA-UHFFFAOYSA-N 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 239000003599 detergent Substances 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- FAMRKDQNMBBFBR-BQYQJAHWSA-N diethyl azodicarboxylate Substances CCOC(=O)\N=N\C(=O)OCC FAMRKDQNMBBFBR-BQYQJAHWSA-N 0.000 description 1
- CSJLBAMHHLJAAS-UHFFFAOYSA-N diethylaminosulfur trifluoride Substances CCN(CC)S(F)(F)F CSJLBAMHHLJAAS-UHFFFAOYSA-N 0.000 description 1
- HQWPLXHWEZZGKY-UHFFFAOYSA-N diethylzinc Chemical compound CC[Zn]CC HQWPLXHWEZZGKY-UHFFFAOYSA-N 0.000 description 1
- 206010012818 diffuse large B-cell lymphoma Diseases 0.000 description 1
- 125000006001 difluoroethyl group Chemical group 0.000 description 1
- 125000004852 dihydrofuranyl group Chemical group O1C(CC=C1)* 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- 238000006471 dimerization reaction Methods 0.000 description 1
- 125000000532 dioxanyl group Chemical group 0.000 description 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 239000006185 dispersion Substances 0.000 description 1
- 238000006073 displacement reaction Methods 0.000 description 1
- 238000004821 distillation Methods 0.000 description 1
- CVLLAKCGAFNZHJ-UHFFFAOYSA-N ditert-butyl-[6-methoxy-3-methyl-2-[2,4,6-tri(propan-2-yl)phenyl]phenyl]phosphane Chemical compound COC1=CC=C(C)C(C=2C(=CC(=CC=2C(C)C)C(C)C)C(C)C)=C1P(C(C)(C)C)C(C)(C)C CVLLAKCGAFNZHJ-UHFFFAOYSA-N 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 230000008482 dysregulation Effects 0.000 description 1
- 230000000694 effects Effects 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 238000010828 elution Methods 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 230000001804 emulsifying effect Effects 0.000 description 1
- 201000004101 esophageal cancer Diseases 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- 125000001301 ethoxy group Chemical group [H]C([H])([H])C([H])([H])O* 0.000 description 1
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 1
- FAMRKDQNMBBFBR-UHFFFAOYSA-N ethyl n-ethoxycarbonyliminocarbamate Chemical compound CCOC(=O)N=NC(=O)OCC FAMRKDQNMBBFBR-UHFFFAOYSA-N 0.000 description 1
- FMVJYQGSRWVMQV-UHFFFAOYSA-N ethyl propiolate Chemical compound CCOC(=O)C#C FMVJYQGSRWVMQV-UHFFFAOYSA-N 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 102000052178 fibroblast growth factor receptor activity proteins Human genes 0.000 description 1
- 239000012467 final product Substances 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 125000003784 fluoroethyl group Chemical group [H]C([H])(F)C([H])([H])* 0.000 description 1
- 125000004216 fluoromethyl group Chemical group [H]C([H])(F)* 0.000 description 1
- 125000002541 furyl group Chemical group 0.000 description 1
- 206010017758 gastric cancer Diseases 0.000 description 1
- 208000005017 glioblastoma Diseases 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 230000005484 gravity Effects 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- JAXFJECJQZDFJS-XHEPKHHKSA-N gtpl8555 Chemical compound OC(=O)C[C@H](N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](C(C)C)C(=O)N1CCC[C@@H]1C(=O)N[C@H](B1O[C@@]2(C)[C@H]3C[C@H](C3(C)C)C[C@H]2O1)CCC1=CC=C(F)C=C1 JAXFJECJQZDFJS-XHEPKHHKSA-N 0.000 description 1
- 150000004820 halides Chemical class 0.000 description 1
- 125000004438 haloalkoxy group Chemical group 0.000 description 1
- 125000005347 halocycloalkyl group Chemical group 0.000 description 1
- 229910052736 halogen Inorganic materials 0.000 description 1
- 201000000459 head and neck squamous cell carcinoma Diseases 0.000 description 1
- 238000010438 heat treatment Methods 0.000 description 1
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 1
- 239000001257 hydrogen Substances 0.000 description 1
- 125000004435 hydrogen atom Chemical group [H]* 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- 125000002632 imidazolidinyl group Chemical group 0.000 description 1
- 150000002466 imines Chemical class 0.000 description 1
- 238000007654 immersion Methods 0.000 description 1
- 125000003392 indanyl group Chemical group C1(CCC2=CC=CC=C12)* 0.000 description 1
- 125000003454 indenyl group Chemical group C1(C=CC2=CC=CC=C12)* 0.000 description 1
- 125000001041 indolyl group Chemical group 0.000 description 1
- 230000004968 inflammatory condition Effects 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 238000007919 intrasynovial administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 230000009545 invasion Effects 0.000 description 1
- 229910052740 iodine Inorganic materials 0.000 description 1
- INQOMBQAUSQDDS-UHFFFAOYSA-N iodomethane Chemical compound IC INQOMBQAUSQDDS-UHFFFAOYSA-N 0.000 description 1
- 125000000959 isobutyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])* 0.000 description 1
- 125000003253 isopropoxy group Chemical group [H]C([H])([H])C([H])(O*)C([H])([H])[H] 0.000 description 1
- 125000002183 isoquinolinyl group Chemical group C1(=NC=CC2=CC=CC=C12)* 0.000 description 1
- 125000000842 isoxazolyl group Chemical group 0.000 description 1
- ZLVXBBHTMQJRSX-VMGNSXQWSA-N jdtic Chemical compound C1([C@]2(C)CCN(C[C@@H]2C)C[C@H](C(C)C)NC(=O)[C@@H]2NCC3=CC(O)=CC=C3C2)=CC=CC(O)=C1 ZLVXBBHTMQJRSX-VMGNSXQWSA-N 0.000 description 1
- 201000010982 kidney cancer Diseases 0.000 description 1
- 201000000062 kidney sarcoma Diseases 0.000 description 1
- 208000014018 liver neoplasm Diseases 0.000 description 1
- RENRQMCACQEWFC-UGKGYDQZSA-N lnp023 Chemical compound C1([C@H]2N(CC=3C=4C=CNC=4C(C)=CC=3OC)CC[C@@H](C2)OCC)=CC=C(C(O)=O)C=C1 RENRQMCACQEWFC-UGKGYDQZSA-N 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 238000004020 luminiscence type Methods 0.000 description 1
- LVKCSZQWLOVUGB-UHFFFAOYSA-M magnesium;propane;bromide Chemical compound [Mg+2].[Br-].C[CH-]C LVKCSZQWLOVUGB-UHFFFAOYSA-M 0.000 description 1
- 230000036212 malign transformation Effects 0.000 description 1
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 108010082117 matrigel Proteins 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 239000002609 medium Substances 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 239000002207 metabolite Substances 0.000 description 1
- 229910001507 metal halide Inorganic materials 0.000 description 1
- 150000005309 metal halides Chemical class 0.000 description 1
- 206010061289 metastatic neoplasm Diseases 0.000 description 1
- ORUCTBNNYKZMSK-UHFFFAOYSA-N methyl 1h-pyrazole-5-carboxylate Chemical compound COC(=O)C=1C=CNN=1 ORUCTBNNYKZMSK-UHFFFAOYSA-N 0.000 description 1
- HPMNCYDJGIMYKN-UHFFFAOYSA-N methyl 4-bromo-5-fluoro-2-nitrobenzoate Chemical compound COC(=O)C1=CC(F)=C(Br)C=C1[N+]([O-])=O HPMNCYDJGIMYKN-UHFFFAOYSA-N 0.000 description 1
- JOQJEWAXHQDQAG-UHFFFAOYSA-N methyl pyrimidine-2-carboxylate Chemical compound COC(=O)C1=NC=CC=N1 JOQJEWAXHQDQAG-UHFFFAOYSA-N 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 238000004264 monolayer culture Methods 0.000 description 1
- 125000002757 morpholinyl group Chemical group 0.000 description 1
- REPVNSJSTLRQEQ-UHFFFAOYSA-N n,n-dimethylacetamide;n,n-dimethylformamide Chemical compound CN(C)C=O.CN(C)C(C)=O REPVNSJSTLRQEQ-UHFFFAOYSA-N 0.000 description 1
- OQJBFFCUFALWQL-UHFFFAOYSA-N n-(piperidine-1-carbonylimino)piperidine-1-carboxamide Chemical compound C1CCCCN1C(=O)N=NC(=O)N1CCCCC1 OQJBFFCUFALWQL-UHFFFAOYSA-N 0.000 description 1
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001280 n-hexyl group Chemical group C(CCCCC)* 0.000 description 1
- 125000000740 n-pentyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- XBXCNNQPRYLIDE-UHFFFAOYSA-M n-tert-butylcarbamate Chemical compound CC(C)(C)NC([O-])=O XBXCNNQPRYLIDE-UHFFFAOYSA-M 0.000 description 1
- 239000007922 nasal spray Substances 0.000 description 1
- IOMMMLWIABWRKL-WUTDNEBXSA-N nazartinib Chemical compound C1N(C(=O)/C=C/CN(C)C)CCCC[C@H]1N1C2=C(Cl)C=CC=C2N=C1NC(=O)C1=CC=NC(C)=C1 IOMMMLWIABWRKL-WUTDNEBXSA-N 0.000 description 1
- 238000006396 nitration reaction Methods 0.000 description 1
- 150000002828 nitro derivatives Chemical class 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 230000000269 nucleophilic effect Effects 0.000 description 1
- YLACRFYIUQZNIV-UHFFFAOYSA-N o-(2,4-dinitrophenyl)hydroxylamine Chemical compound NOC1=CC=C([N+]([O-])=O)C=C1[N+]([O-])=O YLACRFYIUQZNIV-UHFFFAOYSA-N 0.000 description 1
- PIDFDZJZLOTZTM-KHVQSSSXSA-N ombitasvir Chemical compound COC(=O)N[C@@H](C(C)C)C(=O)N1CCC[C@H]1C(=O)NC1=CC=C([C@H]2N([C@@H](CC2)C=2C=CC(NC(=O)[C@H]3N(CCC3)C(=O)[C@@H](NC(=O)OC)C(C)C)=CC=2)C=2C=CC(=CC=2)C(C)(C)C)C=C1 PIDFDZJZLOTZTM-KHVQSSSXSA-N 0.000 description 1
- 239000012074 organic phase Substances 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 208000008443 pancreatic carcinoma Diseases 0.000 description 1
- 210000003455 parietal bone Anatomy 0.000 description 1
- 229960002275 pentobarbital sodium Drugs 0.000 description 1
- 239000003208 petroleum Substances 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 150000002989 phenols Chemical class 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- 125000004193 piperazinyl group Chemical group 0.000 description 1
- 125000003386 piperidinyl group Chemical group 0.000 description 1
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 1
- 229920000136 polysorbate Polymers 0.000 description 1
- 229920000053 polysorbate 80 Polymers 0.000 description 1
- 235000015320 potassium carbonate Nutrition 0.000 description 1
- 235000011181 potassium carbonates Nutrition 0.000 description 1
- 239000012286 potassium permanganate Substances 0.000 description 1
- 238000004262 preparative liquid chromatography Methods 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- VVWRJUBEIPHGQF-MDZDMXLPSA-N propan-2-yl (ne)-n-propan-2-yloxycarbonyliminocarbamate Chemical compound CC(C)OC(=O)\N=N\C(=O)OC(C)C VVWRJUBEIPHGQF-MDZDMXLPSA-N 0.000 description 1
- 125000006239 protecting group Chemical group 0.000 description 1
- 125000000719 pyrrolidinyl group Chemical group 0.000 description 1
- DRYRBWIFRVMRPV-UHFFFAOYSA-N quinazolin-4-amine Chemical compound C1=CC=C2C(N)=NC=NC2=C1 DRYRBWIFRVMRPV-UHFFFAOYSA-N 0.000 description 1
- RJSRSRITMWVIQT-UHFFFAOYSA-N quinolin-6-amine Chemical compound N1=CC=CC2=CC(N)=CC=C21 RJSRSRITMWVIQT-UHFFFAOYSA-N 0.000 description 1
- 125000004621 quinuclidinyl group Chemical group N12C(CC(CC1)CC2)* 0.000 description 1
- 102000027426 receptor tyrosine kinases Human genes 0.000 description 1
- 108091008598 receptor tyrosine kinases Proteins 0.000 description 1
- 238000010992 reflux Methods 0.000 description 1
- 238000005057 refrigeration Methods 0.000 description 1
- 239000011369 resultant mixture Substances 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 238000007363 ring formation reaction Methods 0.000 description 1
- 102200048955 rs121434569 Human genes 0.000 description 1
- 238000013207 serial dilution Methods 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 229910002027 silica gel Inorganic materials 0.000 description 1
- 239000000741 silica gel Substances 0.000 description 1
- 239000000377 silicon dioxide Substances 0.000 description 1
- 229910052814 silicon oxide Inorganic materials 0.000 description 1
- 210000003625 skull Anatomy 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 229910000029 sodium carbonate Inorganic materials 0.000 description 1
- 239000012312 sodium hydride Substances 0.000 description 1
- 235000019333 sodium laurylsulphate Nutrition 0.000 description 1
- 230000003381 solubilizing effect Effects 0.000 description 1
- 239000011877 solvent mixture Substances 0.000 description 1
- 230000003595 spectral effect Effects 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 201000011549 stomach cancer Diseases 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 239000003765 sweetening agent Substances 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- WIZGKLCQBIOGNX-UHFFFAOYSA-N tert-butyl n-(3-fluoropyridin-4-yl)carbamate Chemical compound CC(C)(C)OC(=O)NC1=CC=NC=C1F WIZGKLCQBIOGNX-UHFFFAOYSA-N 0.000 description 1
- YLQBMQCUIZJEEH-UHFFFAOYSA-N tetrahydrofuran Natural products C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 description 1
- 125000003718 tetrahydrofuranyl group Chemical group 0.000 description 1
- 125000001412 tetrahydropyranyl group Chemical group 0.000 description 1
- 125000003507 tetrahydrothiofenyl group Chemical group 0.000 description 1
- 125000003831 tetrazolyl group Chemical group 0.000 description 1
- 125000001113 thiadiazolyl group Chemical group 0.000 description 1
- 125000001544 thienyl group Chemical group 0.000 description 1
- 201000002510 thyroid cancer Diseases 0.000 description 1
- JOXIMZWYDAKGHI-UHFFFAOYSA-N toluene-4-sulfonic acid Chemical compound CC1=CC=C(S(O)(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-N 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 238000000844 transformation Methods 0.000 description 1
- 206010044412 transitional cell carcinoma Diseases 0.000 description 1
- TUQOTMZNTHZOKS-UHFFFAOYSA-N tributylphosphine Chemical compound CCCCP(CCCC)CCCC TUQOTMZNTHZOKS-UHFFFAOYSA-N 0.000 description 1
- 125000004205 trifluoroethyl group Chemical group [H]C([H])(*)C(F)(F)F 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 239000005483 tyrosine kinase inhibitor Substances 0.000 description 1
- 201000005112 urinary bladder cancer Diseases 0.000 description 1
- 206010046766 uterine cancer Diseases 0.000 description 1
- 238000005292 vacuum distillation Methods 0.000 description 1
- 125000000391 vinyl group Chemical group [H]C([*])=C([H])[H] 0.000 description 1
- 238000006886 vinylation reaction Methods 0.000 description 1
- 229910052727 yttrium Inorganic materials 0.000 description 1
- 229910052725 zinc Inorganic materials 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D417/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
- C07D417/14—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D239/00—Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
- C07D239/70—Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
- C07D239/72—Quinazolines; Hydrogenated quinazolines
- C07D239/86—Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
- C07D239/94—Nitrogen atoms
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
- C07D401/12—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/14—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D403/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
- C07D403/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
- C07D403/12—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D403/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
- C07D403/14—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
- C07D471/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
- C07D471/04—Ortho-condensed systems
Definitions
- EGFR INHIBITORS CROSS REFERENCES TO RELATED APPLICATONS
- BACKGROUND Epidermal growth factor receptor belongs to the HER family of receptors which comprise four related proteins EGFR, HER2, HER3, and HER4.
- the HER receptors are known to be activated by binding to different ligands including EGF (see Duggirala et.al., Biomol Ther, 2022, 30, 19). After binding to a ligand, the receptor forms functionally active dimers including EGFR-EGFR, EGFR-HER2, EGFR-HER3, and EGFR-HER4. Dimerization induces the activation of the tyrosine kinase domain of EGFR, leading to a series of downstream signaling pathways for cell proliferation, survival, and anti- apoptosis (see Uribe et al., Cancers 2021, 13, 2748).
- Dysregulations of EGFR signal transduction pathways can promote malignant transformation of normal cells into tumor cells and subsequently assist tumor cell proliferation, invasion, metastasis, and angiogenesis.
- NSCLC non-small cell lung cancer
- the two most common EGFR gene mutations are del19 (i.e., a short in-frame deletion in exon 19) and L858R (i.e., a single missense mutation in exon 21). See Konduri et.al., Cancer Discov, 2016, 6, 601. These two mutations trigger ligand independent EGFR activation, driving cancerous growth and metastasis of NSCLC (see Uribe et al., Cancers 2021, 13, 2748).
- TKIs small-molecule EGFR tyrosine kinase inhibitors
- erlotinib small-molecule EGFR tyrosine kinase inhibitors
- gefitinib small-molecule EGFR tyrosine kinase inhibitors
- TKI resistance has been associated with a secondary mutation (T790M) in the EGFR’s kinase domain in 50%–60% of the cases (see Pao et al., PLoS Med.2005, 2, e73 and Leonetti et al., British Journal of Cancer, 2019, 121, 725).
- T790M secondary mutation
- osimertinib as a third-generation EGFR inhibitor was developed for treating patients carrying the EGFR T790M mutation.
- X can be an S, G, N, Y, T, or D
- Tumors harboring double mutants e.g., Del19/C797X and L858R/C797X, are no longer sensitive to osimertinib or other third-generation EGFR inhibitors. There is no approved medicine to treat patients carrying double mutants.
- EGFR inhibitors such as gefitinib and erlotinib are not effective in treating brain cancer due to their low ability to cross the blood-brain barrier. It is critical that EGFR inhibitors are capable of crossing the blood-brain barrier, as about 70% of NSCLC patients bearing mutant-EGFR develop brain metastases (see Subramaniam et al, Front Oncol.2018, 8, 208). Thus, there is an unmet medical need for effective EGFR inhibitors to penetrate blood- brain barrier and treat NSCLC patients having double EGFR mutants and brain metastasis.
- the present invention is based on an unexpected discovery that certain quinazoline compounds show promising anti-tumor activities.
- this invention relates to quinazoline compounds of Formula I: I, in which one of X and Y is H, deuterium, or halo; the other of X and Y is , in which R 1 is F, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, or 4-6 membered heterocyclyl, R 2 is H, deuterium, F, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, or 4-6 membered heterocyclyl, or R 1 and R 2 , together with the carbon atom they bond to, are C 3 -C 6 cycloalkyl or 4-6 membered heterocyclyl; R3 is 5-10 membered heteroaryl orC 6 -C 10 aryl; and n is 1, 2, or 3; R 4 is H, halo, cyano, C 3 -C 6 cycloalkyl, ORa, 4-10 membered heterocyclyl, or 5-6 membere
- the above-described compounds have one or more of the following features: (i) n is 1; (ii) R 1 is F, C 1 -C 3 alkyl, C 3 -C 6 cycloalkyl, preferably methyl, CF 3 , CHF 2 , cyclopropyl, or CD 3 , and more preferably, methyl; or R 1 and R 2 , together with the carbon atom they bond to, are C 3 -C 6 cycloalkyl, preferably cyclopropyl; (iii) R 2 is H, F, C 1 -C 3 alkyl, C 3 -C 6 cycloalkyl, preferably methyl, CF 3 , CHF 2 , cyclopropyl, or CD 3 ; (iv) R 3 is ; preferably, R 3 is pyrimidinyl, imidazolyl,
- R 7 is phenyl optionally substituted with one or more groups selected from the group consisting of F, Cl, Br and acetylenyl, preferably R 7 is . Further, R 7 can also be 9- or 10- membered bicyclic heteroaryl optionally substitutes with halo (e.g., F).
- R 6 is H, CH 3 , CD 3 , or CHF 2 , preferably H or CHF 2 ;
- R 8 is H or deuterium, preferably H.
- Subsets of the compounds of Formula I are represented by Formulas IA and IB: .
- R 1 -R 7 and n is defined above including any combinations of features (i) to (vii) as shown above.
- R 4 can be
- Preferred compounds of Formula IA have the following features: (i) n is 1; (ii) R 5 is H or deuterium; (iii) R 1 is F, C 1 -C 3 alkyl, or C 3 -C 6 cycloalkyl, preferably methyl, CF 3 , CHF 2 , cyclopropyl, or CD 3 , and R 2 is H, F, CH 3 , D, CD 3 , cyclopropyl, CF 3 , CHF 2 ; or R 1 and R 2 , together with the carbon atom they are bonded to, is cyclopropyl;
- n is 1, 2, or 3 and R 1 is F, CH 3 , CD 3 , CF 3 , CHF 2 , cyclopropyl.
- R 4 can be
- Preferred compounds of Formula IB include the following features: (i) n is 1; (ii) R 5 is H or deuterium; (iii) R 1 is F, CH 3 , CD 3 , CF 3 , CHF 2 , cyclopropyl; (iv) R 2 is H, CH 3 , D, CD 3 , F, cyclopropyl, CF 3 , or CHF 2 ; (v) R 3 is (vii) R 6 is H, CH 3 or CHF 2 ; and (viii) R 7 is .
- Another aspect of this invention is a method of treating cancer comprising administering to a subject in need thereof an effective amount of any one of the compounds described above.
- Nonlimiting examples of cancer treatable by the compounds of this invention include lung cancer (e.g., non-small cell lung cancer) and brain cancer.
- the current invention further includes use of such a compound (e.g., a pharmaceutical composition containing one of the compounds of Formula I described above) for treating cancer or for the manufacture of a medicament for treating cancer.
- a method of inhibiting EGFR by administering to a subject in need thereof an effective amount of any one of the compounds described above.
- a pharmaceutical composition comprising any of the compounds described above and a pharmaceutically acceptable carrier. As described above, the pharmaceutical composition is particularly useful in treating cancer. Table 1 below shows 65 exemplary compounds of the present invention, i.e., Compounds 1-65.
- halo herein refers to a fluoro, chloro, bromo, or iodo radical. Examples include a fluoro radical (F) and a bromo radical (Br).
- alkyl refers to a straight or branched hydrocarbon group, containing 1-20 carbon atoms (e.g., C 1-6 ) and a monovalent radical center derived by the removal of a hydrogen atom from a carbon atom of a parent alkane.
- alkyl groups are methyl, ethyl, n- propyl, isopropyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl.
- haloalkyl refers to alkyl substituted with one or more halo atoms.
- Examples include fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl (e.g., 1-fluoroetyl and 2-fluoroethyl), difluoroethyl (e.g., 1,1-, 1,2-, and 2,2-difluoroethyl), and trifluoroethyl (e.g., 2,2,2- trifluoroethyl).
- alkyl as used herein includes haloalkyl.
- alkoxy refers to an –O–alkyl group. Examples are methoxy, ethoxy, propoxy, and isopropoxy.
- Alkoxy also includes haloalkoxy, namely, alkoxy substituted with one or more halogens, e.g., –O-CH 2 Cl and –O-CHClCH 2 Cl.
- cycloalkyl refers to a nonaromatic, saturated or unsaturated monocyclic, bicyclic, tricyclic, or tetracyclic hydrocarbon group containing 3 to 12 carbons (e.g., C 3-6 and C 3-10 ). Examples are cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl.
- heterocyclyl refers to a nonaromatic, saturated or unsaturated, 3–8 membered monocyclic, 8–12 membered bicyclic, or 11–14 membered tricyclic ring system having one or more heteroatoms (e.g., O, N, P, and S).
- Examples include aziridinyl, azetidinyl, pyrrolidinyl, dihydrofuranyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiophenyl, tetrahydro-2-H-thiopyran-1,1-dioxidyl, piperazinyl, piperidinyl, morpholinyl, imidazolidinyl, azepanyl, dihydrothiadiazolyl, dioxanyl, and quinuclidinyl.
- Both “cycloalkyl” and “heterocyclyl” also include fused, bridged, and spiro ring systems.
- aryl refers a 6-carbon monocyclic, 10-carbon bicyclic, 14-carbon tricyclic aromatic ring system wherein each ring can have one or more (e.g., 1 to 10, 1 to 5, and 1 to 3) substituents. Examples include phenyl, biphenyl, 1 ⁇ or 2-naphthyl, 1,2-dihydronaphthyl, 1,2,3,4-tetrahydronaphthyl, indenyl, and indanyl.
- aralkyl refers to alkyl substituted with an aryl group.
- heteroaryl refers to an aromatic 5–8 membered monocyclic, 8–12 membered bicyclic, or 11–14 membered tricyclic ring system having one or more heteroatoms (e.g., O, N, P, and S).
- heterocyclylalkyl refers to an alkyl group substituted with a heterocyclyl or heteroaryl group.
- Alkyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, aralkyl, heterocyclylalkyl, and heteroaryl mentioned herein include both substituted and unsubstituted moieties, unless specified otherwise.
- Examples of a substituent include deuterium (D), hydroxyl (OH), halo (e.g., F and Cl), amino (NH 2 ), cyano (CN), nitro (NO 2 ), alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, acylamino, alkylamino, aminoalkyl, haloalkyl (e.g., trifluoromethyl), heterocyclyl, alkoxycarbonyl, amido, carboxy (COOH), alkanesulfonyl, alkylcarbonyl, alkenylcarbonyl, carbamido, carbamyl, carboxyl, thioureido, thiocyanato, sulfonamido, aryl, arylamino, aralkyl, and heteroaryl.
- D deuterium
- OH hydroxyl
- halo e.g., F and Cl
- amino NH 2
- the pharmaceutically acceptable salts include those listed in Handbook of Pharmaceutical Salts: Properties, Selection and Use, 2 nd Revised Edition, P. H. Stahl and C. G. Wermuth (Eds.), Wiley-VCH, New York, (2011).
- other salts are contemplated in the invention. They may serve as intermediates in the purification of compounds or in the preparation of other pharmaceutically acceptable salts, or are useful for identification, characterization or purification of compounds of the invention.
- a solvate refers to a complex formed between an active compound and a pharmaceutically acceptable solvent.
- a prodrug refers to a compound that, after administration, is metabolized into a pharmaceutically active drug.
- examples of a prodrug include esters and other pharmaceutically acceptable derivatives.
- the compounds of the present invention may contain one or more non-aromatic double bonds or asymmetric centers. Each of them occurs as a racemate or a racemic mixture, a single R enantiomer, a single S enantiomer, an individual diastereomer, a diastereometric mixture, a cis-isomer, or a trans-isomer.
- treating refers to administering one or more of the compounds to a subject with the purpose to confer a therapeutic effect, e.g., to slow, interrupt, arrest, control, or stop of the progression of an existing disorder and/or symptoms thereof, but does not necessarily indicate a total elimination of all symptoms.
- An effective amount refers to the amount of a compound that is required to confer the therapeutic effect. Effective doses will vary, as recognized by those skilled in the art, depending on the types of symptoms treated, route of administration, excipient usage, and the possibility of co-usage with other therapeutic treatment.
- Cancer treatable with a compound of this invention includes those caused by KRAS mutation, SOS1 oncogenic mutation, or oncogenic mutation/overexpression of receptor tyrosine kinases such as EGFR, FGFR, etc.
- Nonlimiting examples include pancreatic cancer, lung cancer, colorectal cancer, cholangiocarcinoma, multiple myeloma, melanoma, uterine cancer, endometrial cancer, thyroid cancer, acute myeloid leukemia, bladder cancer, urothelial cancer, gastric cancer, cervical cancer, head and neck squamous cell carcinoma, diffuse large B cell lymphoma, esophageal cancer, chronic lymphocytic leukemia, hepatocellular cancer, breast cancer, ovarian cancer, prostate cancer, glioblastoma, renal cancer and sarcoma, and brain metastasis.
- the compounds of this invention are particularly effective in treating cancer associated with EGFR such as brain cancer, lung cancer (e.g., non-small cell lung cancer, and non-small cell lung cancer with brain metastasis or leptomeningeal).
- subject refers to an animal including human or non-human, such as a mammal.
- a human is a preferred subject.
- a compound of this invention may be administered alone or in the form of a pharmaceutical composition with pharmaceutically acceptable carriers, diluents or excipients.
- Such pharmaceutical compositions and processes for making the same are known in the art (See, e.g., Remington: The Science and Practice of Pharmacy, A. Adejare, Editor, 23rd Edition., Academic Press, 2020).
- composition or a kit containing one or more of the above-described compounds can be administered alone or co-administered with at least one other pharmacologically active substance simultaneously, concurrently, sequentially, successively, alternately, or separately.
- Simultaneous administration also referring to as concomitant administration, includes administration at substantially the same time.
- Concurrent administration includes administering the active agents within the same general time period, for example on the same day(s) but not necessarily at the same time.
- Alternate administration includes administration of one agent during a time period, for example over the course of a few days or a week, followed by administration of the other agent(s) during a subsequent period of time, for example over the course of a few days or a week, and then repeating the pattern for one or more cycles.
- Sequential or successive administration includes administration of one agent during a first time period (for example over the course of a few days or a week) using one or more doses, followed by administration of the other agent(s) during a second and/or additional time period (for example over the course of a few days or a week) using one or more doses.
- An overlapping schedule may also be employed, which includes administration of the active agents on different days over the treatment period, not necessarily according to a regular sequence.
- the elements of the combinations of this invention may be administered (whether dependently or independently) by methods customary to the skilled person, e.g., by oral, enteral, parenteral, nasal, vaginal, rectal, or topical routes of administration and may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, excipients and/or vehicles appropriate for each route of administration.
- a composition for oral administration can be any orally acceptable dosage form including capsules, tablets, emulsions and aqueous suspensions, dispersions, and solutions.
- commonly used carriers include lactose and corn starch.
- Lubricating agents such as magnesium stearate, are also typically added.
- useful diluents include lactose and dried corn starch.
- a nasal aerosol or inhalation composition can be prepared according to techniques well known in the art of pharmaceutical formulation.
- such a composition can be prepared as a solution in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents.
- a composition having one or more of the above-described compounds can also be administered in the form of suppositories for rectal administration.
- the carrier in the pharmaceutical composition must be “acceptable” in the sense that it is compatible with the active ingredient of the composition (and preferably, capable of stabilizing the active ingredient) and not deleterious to the subject to be treated.
- One or more solubilizing agents can be utilized as pharmaceutical excipients for delivery of an active compound. Examples include colloidal silicon oxide, magnesium stearate, cellulose, sodium lauryl sulfate, and D&C Yellow # 10. The details of one or more embodiments of the invention are set forth in the description below. Other features, objects, and advantages of the invention will be apparent from the description and from the claims. DETAILED DESCRIPTION The present invention is based on a surprising discovery that the compounds of Formula I reproduced below are effective in inhibiting EGFR and treating cancer.
- I Variables R 1 -R 7 , X, and Y are defined above. Further, in a subset of the compounds of formula I, R 3 is Preferably, R 3 is . In another subset of the compounds of formula I, R 4 is
- R 7 is phenyl, and R 7 is optionally substituted with one or more groups selected from the groups consisting of F, Cl, Br and acetylenyl; Preferably, R 7 is . More preferably, R 7 is .
- R 7 is 9- or 10- membered bicyclic heteroaryl, preferably quinolinyl, indolizinyl, pyrazolo[1,5-a]pyridinyl, imidazo[1,2- a]pyridinyl, benzo[c]isothiazolyl or benzo[d]isothiazolyl; and R 7 is optionally substituted with halo (for example F); Preferably, R 7 is . . In a preferred subset of the compounds of formula IA:
- R 3 is
- the compounds of the invention are useful in treating mutant EGFR mediated cancer that has metastasized to brain and central nervous system. They are also suitable for treating mutant EGFR mediated local or metathesized lung cancer.
- the compound can be a compound of Formula I, IA, or IB.
- the compounds of Formula I can be prepared by synthetic methods well known in the art. See, e.g., R. Larock, Comprehensive Organic Transformations (3 rd Ed., John Wiley and Sons 2018); P. G. M. Wuts and T. W. Greene, Greene’s Protective Groups in Organic Synthesis (4 th Ed., John Wiley and Sons 2007); L. Fieser and M.
- Fieser, Fieser and Fieser are Reagents for Organic Synthesis (John Wiley and Sons 1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis (2 nd ed., John Wiley and Sons 2009) and subsequent editions thereof.
- the compounds thus prepared can be purified following conventional methods such as crystallization, distillation/vacuum distillation, flash chromatography over silica, and preparative liquid chromatography.
- Efficacy of the compounds of this invention can be initially determined using cellular pERK potency assay or 2D cell proliferation assay, all described below. The selected compounds can be further tested to verify their efficacy, e.g., by administering it to an animal.
- a compound of this invention is preferably formulated into a pharmaceutical composition containing a pharmaceutical carrier.
- the pharmaceutical composition is then given to a subject in need thereof to inhibit mutant EGFR (e.g. del19, L858R, del19/C797S, del19/L858R, amplification) thus treating cancer.
- mutant EGFR e.g. del19, L858R, del19/C797S, del19/L858R, amplification
- Schemes I to V depict five exemplary routes for preparing a compound of the invention (In the following schemes, Ar is R 7 ).
- Scheme I Scheme III The compounds of the invention can be prepared by combining the reactions in these general synthetic schemes or additional reaction schemes specifically depicted herein or reactions known in the art and not limited to the general schemes described herein.
- General Synthetic Procedures Provided below are exemplary processes for preparing compounds of Formula I (e.g., IA and IB). These processes provide further features of the invention and are illustrated below.
- compounds of formula IA and IB can be synthesized using the procedures illustrated in Schemes I to V above.
- quinazolinone IA-2 can be prepared by reacting benzonitrile derivative IA-1 with formic acid in the presence of H 2 SO 4 .
- Quinazolinone IA-2 is then converted to chloride IA-3 using oxalyl chloride, thionyl chloride, or POCl 3 in a suitable solvent such as chloroform.
- the chlorine atom in IA-3 is displaced by IA-4 (i.e., H 2 N-Ar) to yield IA-5 in a suitable solvent such as isopropanol.
- the fluorine atom in IA-5 is displaced by chiral or achiral alcohol IA-6 in the presence of a base, such as NaH or t-BuOK, in a suitable solvent such as THF to yield ether IA-7.
- R 4 is introduced using palladium mediated coupling with borate, boronic acid, or boronate ester derivatives such as IA-8a or IA-8b in a suitable solvent (e.g., dioxane and water) to provide a compound of formula IA-9.
- a suitable solvent e.g., dioxane and water
- reduction of the halide of IA-7 with Et 3 SiH in the presence of a palladium catalyst or catalytic hydrogen reduction in a suitable solvent such as dioxane can provide a compound of Formula IA-10.
- R 4 is introduced using palladium mediated coupling with borate, boronic acid or boronate ester derivatives (e.g., IA-8a and IA-8b) in a suitable solvent such as dioxane and water to provide a compound of formula IA-9.
- a suitable solvent such as dioxane and water
- nucleophilic displacement reaction with an amine or heteroaryl R 4 (e.g., pyrazole and imidazole or their derivatives) in a suitable solvent such as dioxane provides a compound of formula IA-9.
- the bromine atom in intermediate IA-13 can be displaced using metal halide (e.g., cuprous iodide) mediated coupling with heteroaryl-OH derivatives (IA-8d) such as 1-methyl-1H-pyrazol-4-ol, in the presence of a suitable ligand such as 3,4,7,8-tetramethyl-1,10-phenanthroline in a suitable solvent such as toluene to generate compound of formula 1A-9.
- the bromine atom in intermediate IA-13 can also be displaced by HO-alkyl (IA-8d) group via palladium mediated C-O bond formation using RockPhos Pd G3 (CAS# 2009020-38-4) catalyst.
- Quinazolinone derivative IB-4 is converted to chloride IB-5 using oxalyl chloride, thionyl chloride or POCl 3 in a suitable solvent such as CHCl 3 .
- the chlorine atom in IB-5 is displaced by IA-4 (i.e., H 2 N-Ar) to provide intermediate IB-6.
- R 4 is introduced using palladium mediated coupling with borate, boronic acid or boronate ester derivatives (e.g., IA-8a or IA-8b) in a suitable solvent such as dioxane and water to yield the final product of formula IB-7.
- Some of the compounds of formula IB can be synthesized following scheme IV.
- IB-8 is etherified using a Mitsunobu reaction using reagents such as diethyl azodicarboxylate/Ph 3 P or 1,1′-(azodicarbonyl)dipiperidine/n-Bu3P with chiral or achiral alcohol IA-6 to yield a compound of formula IB-9.
- reagents such as diethyl azodicarboxylate/Ph 3 P or 1,1′-(azodicarbonyl)dipiperidine/n-Bu3P with chiral or achiral alcohol IA-6 to yield a compound of formula IB-9.
- Compounds of formula IB can also be synthesized following scheme V.
- Phenol derivative IB-10 is converted to a corresponding carbonate IB-11 using ethyl carbonochloridate in a suitable solvent such as dichloromethane.
- Nitration of the carbonate IB-11 in a mixture of H 2 SO 4 and HNO 3 affords nitro derivative IB-12, which is hydrolyzed using a base such as NaHCO 3 in a suitable solvent such as methanol to from phenol IB-13.
- the phenol IB-13 is converted to vinyl ether using di- ⁇ -chlorobis[(1,2,5,6- ⁇ )-1,5-cyclooctadiene]diiridium mediated vinylation using vinyl acetate in a suitable solvent such as toluene to form intermediate IB-14.
- the vinyl group in IB-14 is converted to cyclopropyl derivative IB-15 using diiodomethane and diethyl zinc in a suitable solvent such as DCM.
- the bromo group in IB-15 is displaced with CuCN in a suitable solvent such as NMP to form the cyano intermediate IB-16.
- the fluorine atom in IB-16 is displaced with chiral or achiral alcohol IA-6 in the presence of a base (e.g., NaH and t-BuOK) in a suitable solvent such as THF to generate ether IB-17.
- a base e.g., NaH and t-BuOK
- the nitro group in IB-17 is selectively reduced with sodium dithionite in a suitable solvent such as tetrahydrofuran and water, to provide intermediate amino IB-18, which reacts with (E)-N,N'-bis(3-chloro-2-fluorophenyl)formimidamide (IB-19) in the presence of acetic acid in a suitable solvent such as 2-methyl tetrahydrofuran, to generate quinazolinone derivative of formula IB-20.
- All chemicals, reagents, and solvents were purchased from commercial sources when available and were used as received without further purification. Air- and moisture-sensitive reactions were carried out under an inert atmosphere of argon or nitrogen in oven-dried glassware.
- TLC analytical thin layer chromatography
- 1 H NMR spectra were recorded on Agilent Technologies or VARIAN, 400 MHz NMR spectrometer. 1 H NMR spectra were reported in parts per million ( ⁇ ) relative to TMS (0.0), DMSO-d 6 (2.50) or CD 3 OD (4.80) as an internal reference.
- HPLC high performance liquid chromatography
- a reverse phase column (SHIMADZU, Column, Boston Analytics, Inc, Green ODS C18, 21.2 x 250 mm) of a size appropriate to the quantity of material being separated, generally eluting with a gradient of increasing concentration of methanol or acetonitrile in water, containing 0.1% trifluoroacetic acid or 0.1% formic acid or 0.04% ammonium bicarbonate in water/MeCN at a rate of elution suitable to the column size and separation to be achieved.
- Chemical names are generated using ChemDraw Professional version 19.1.
- EXAMPLES The following intermediates may be used to prepare compounds of the present invention.
- EXAMPLE 36 Synthesis of (S)-N-(3-chloro-2-fluorophenyl)-7-cyclopropoxy-6-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 36).
- Step 1 Synthesis of 4-bromo-2-fluorophenyl ethyl carbonate
- a solution of 4-bromo-2-fluorophenol (35 g, 183 mmol, 1 eq) and TEA (22.2 g, 220 mmol, 1.2 eq) in dry DCM (350 mL) at 0 °C was added dropwise a solution of ethyl carbonochloridate (23.9 g, 220 mmol, 1.2 eq).
- the reaction mixture was stirred at 0 °C for 1 h under N 2 , then allowed to warm to room temperature, quenched with H 2 O (100 mL) and extracted with DCM (3 x 100 mL).
- EXAMPLE 39 Synthesis of N-(3-chloro-2-fluorophenyl)-7-cyclopropoxy-6- ⁇ [2-(pyrimidin- 2-yl)propan-2-yl]oxy ⁇ quinazolin-4-amine (Compound 39).
- Step 1 Synthesis of 4-cyclopropoxy-2-nitro-5- ⁇ [2-(pyrimidin-2-yl)propan-2-yl]oxy ⁇ - benzonitrile
- 2-(pyrimidin-2-yl)propan-2-ol 251 mg, 1.82 mmol, 1.5 eq
- dry THF 5 mL
- NaH 44 mg, 1.82 mmol, 1.5 eq
- the reaction mixture was heated to 80 °C and AcOH (0.5 mL) was added.
- the reaction mixture was stirred at 90 °C for 16 h, then cooled to rt and extracted with EtOAc (3 x 10 mL).
- the combined organic layers were washed with NaHCO 3 (aq) (10 mL), dried over anhydrous Na 2 SO 4 and filtered.
- the filtrate was concentrated and purified by prep-HPLC to afford the desired product (62.1 mg, 49% yield) as a white solid.
- EXAMPLE 40 Synthesis of (S)-N-(3-chloro-2-fluorophenyl)-N-methyl-7-(1-methyl-1H- pyrazol-4-yl)-6-(1-(pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 40).
- EXAMPLE 42 Synthesis of N-(3-chloro-2-fluorophenyl)-7-methoxy-6-((2-(3-methyl- pyridin-4-yl)propan-2-yl)oxy)quinazolin-4-amine (Compound 42).
- Step 1 Synthesis of 4-(2-(5-bromo-2-methoxy-4-nitrophenoxy)propan-2-yl)-3-methylpyridine
- 2-(3- methylpyridin-4-yl)propan-2-ol (0.91 g, 6 mmol) under N2 atmosphere.
- the reaction mixture was stirred at 0 °C for 1 h.
- EXAMPLE 50 Synthesis of N-(3-chloro-2,4-difluorophenyl)-7-(pyrimidin-2-yl)-5-((2- (pyrimidin-2-yl)propan-2-yl)oxy)quinazolin-4-amine (Compound 50).
- Compound 51 can be prepared from 1-(4-methyl- 4H-1,2,4-triazol-3-yl)ethan-1-ol (CAS:149762-18-5) and intermediate 1A-12-4.
- Compound 53 can be prepared from 1-(1-methyl-1H- imidazol-2-yl)ethan-1-ol (CAS:41507-36-2) and intermediate 1A-12-4.
- N-(3-chloro-2,4-difluorophenyl)-7-(isothiazol-5-yl)-5-((2-(pyrimidin-2-yl)propan-2- yl)oxy)quinazolin-4-amine (Compound 55) can be prepared from isothiazole-5-boronic acid (CAS:1162262-43-1) and intermediate 1A-13-7.
- N-(3-chloro-4-fluorophenyl)-7-(1-cyclopropyl-1H-pyrazol-4-yl)-5-((2-(pyrimidin-2- yl)propan-2-yl)oxy)quinazolin-4-amine (Compound 57) can be prepared from (1-cyclopropyl- 1H-pyrazol-4-yl)boronic acid (1678534-30-9) and intermediate 1A-13-11.
- N-(3-chloro-2,4-difluorophenyl)-7-(1-(methyl-d3)-1H-pyrazol-4-yl)-5-((2-(thiazol-2- yl)propan-2-yl)oxy)quinazolin-4-amine (Compound 58) can be prepared from 2-thiazol-2-yl- propan-2-ol (CAS:16077-78-4), (1-(methyl-d3)-1H-pyrazol-4-yl)boronic acid (CAS: 2256709- 52-9) and intermediate 1A-12-3.
- EXAMPLE 60 N-(3-chloro-2,4-difluorophenyl)-5-((2-(pyrimidin-2-yl)propan-2-yl)oxy)-7- (thiazol-2-yl)quinazolin-4-amine (Compound 60)
- Compound 60 can be prepared from 1,3-thiazole-2-boronic acid (CAS: 389630-95-9) and intermediate 1A-13-10 following the procedure of Example 1.
- EXAMPLE 62 N-(3-chloro-2,4-difluorophenyl)-7-methoxy-5-((2-(pyrimidin-2-yl)propan-2- yl)oxy)quinazolin-4-amine (Compound 62) Compound 62 can be prepared following general scheme II, Step 4, using intermediate 1A-13-10 and methanol.
- EXAMPLE 63 (S)-N-(3-chloro-2,4-difluorophenyl)-7-methoxy-5-(1-(pyrimidin-2-yl)eth- oxy)quinazolin-4-amine (Compound 63) Compound 63 can be prepared following general scheme II, Step 4, using intermediate 1A-13-9 and methanol.
- EXAMPLE 64 N-(3-chloro-4-fluorophenyl)-7-methoxy-5-((2-(pyrimidin-2-yl)propan-2-yl)- oxy)quinazolin-4-amine (Compound 64) Compound 64 can be prepared following general scheme II, Step 4, using intermediate 1A-13-11 and methanol.
- EXAMPLE 65 (S)-N-(3-chloro-4-fluorophenyl)-7-methoxy-5-(1-(pyrimidin-2-yl)ethoxy) quinazolin-4-amine (Compound 65) Compound 65 can be prepared following general scheme II, Step 4, using intermediate 1A-13-4 and methanol.
- EGFR aberrations become constitutively active and thus induces cascades of cellular signaling events that result in increased phosphorylation of ERK at Threonine 202 and Tyrosine 204 (pERK).
- the so-called In Cell ELISA procedure as described below measures the level of cellular pERK in response to EGFR specific inhibitors in EGFR aberrations of three EGFR cell lines: PC-9 (EGFR/del19), A431 (EGFR amplification), and 11-18 (EGFR/L858R).
- PC-9 cells were obtained from the American Type Culture Collection (ATCC, Manassas, Virginia) and 11-18 and A431 cells were commercially available from RIKEN Cell Bank (Japan).
- PC-9 and 11-18 cells were grown and maintained using RPMI-1640 medium supplemented with 10% heat-inactivated fetal bovine serum.
- A431 cells were grown and maintained using DMEM (low glucose) supplemented with 10% heat-inactivated fetal bovine serum.
- test compounds were prepared with 3-fold serial dilutions in DMSO, with a top concentration of 10 mM.
- 50 ⁇ L of a test compound diluted in media was added to each well of cell culture plate with the final concentration of the compound spanning from 0.00005 ⁇ M to 10 ⁇ M.
- the cells were incubated for 3 hours at 37°C with 5% CO 2 . After the culture medium was removed, the cells were fixed with 200 ⁇ L of 4% formaldehyde in phosphate- buffered saline (PBS) and incubated at RT for 20 min. The plate was then washed three times with PBST (PBS with 0.05% Tween® 20), followed by incubation with 100 ⁇ L of pre-cooled methanol at -20°C for 20 min. After incubation, methanol was removed. The plate was washed with PBST.
- PBS phosphate- buffered saline
- the cells were then permeabilized with 100 ⁇ L/well of 0.1% Triton TM X-100 in PBS at RT for 20 min and then quenched with a buffer (PBST containing 1% H 2 O 2 and 0.1% sodium azide) for 20 min at RT with gentle shaking.
- a buffer PBST containing 1% H 2 O 2 and 0.1% sodium azide
- a blocking buffer 250 ul/well, PierceTM Protein-Free-PBS Blocking Buffer, Cat# 37572
- anti-pERK antibody cell signaling, 1:1000 dilution in 5% BSA in PBST
- secondary antibody- HRP secondary antibody- HRP
- the cellular pERK level was determined using a microplate reader (Biotek® Synergy TM H1, Agilent Technologies Inc., Santa Clara, California) to detect the chemiluminescence signal.
- IC 50 values were determined by fitting a 4-parameter sigmoidal concentration-response model.
- the pERK assay results showed that each of Compounds 1-34 ,36, 37, and 39-41 has an IC 50 of no more than 10 nM and each of Compounds 35, 38, and 42 has an IC 50 between 10.1 nm and 50 nM.
- the results demonstrated that the compounds of this invention are highly effective in inhibiting EGFR.
- 2D Cell Proliferation Assay A cell proliferation assay was used to examine the potency with which compounds inhibit in vitro cell proliferation of cancer cell lines carrying EGFR aberrations or EGFR wild type. The assay demonstrated the molecular mode of action of compounds. Low IC 50 values are indicative of high potency of EGFR inhibitors. It was observed that EGFR inhibitors demonstrated potent inhibitory effects on the proliferation of human cancer cell lines or Baf/3 mouse cell lines carrying EGFR aberrations.
- the cell proliferation assay was performed in two-dimensional (2D) anchorage-dependent conditions in 96 well plates (Black/Clear Flat Bottom commercially available from Corning Inc., Corning, New York) with the following cell lines: Ba/F3 EGFR-Del19/C797S (human EGFR): a mouse Ba/F3 cell line stably expressed exogenous human EGFR with del19 and C797S mutation, Ba/F3 EGFR-L858/C797S (human EGFR): a mouse Ba/F3 cell line stably expressed exogenous human EGFR with L858R and C797S mutation, Ba/F3 EGFR-C797S (human EGFR): a mouse Ba/F3 cell line stably expressed exogenous human EGFR with C797S mutation, PC-9: a human lung cancer cell line with EGFR del19 mutation, 11-18: a human lung cancer cell line with EGFR L858R mutation, A431: a human
- Luminescence signals were determined with a Biotek® Synergy TM H1 plate reader (Agilent Technologies Inc., Santa Clara, California).
- Brain to plasma ratio was determined with 5 in 1 cassette dosing or 4 in 1 cassette dosing or 3 in 1 cassette dosing or 2 in 1 cassette dosing or discrete dosing using non-tumor bearing Sprague Dawley (SD) rats.
- SD Sprague Dawley
- a total of 3 male SD rats were selected from the animal pool of SD rats.
- the compounds were dissolved in a mixture of 5% EtOH/85%PE 400/10% H 2 O solution and the animals were dosed orally at 2 mg/kg dose with a concentration of 0.2 mg/mL.
- the plasma and brain samples were collected 2h after dosing.
- 0.3 mL blood was collected from each animal into K 2 EDTA tubes kept on ice. The samples were centrifuged within 30 minutes of collection under refrigeration at 5 °C for 5 minutes at 6000 rpm to obtain plasma samples.
- a plasma sample (20 ⁇ L) was added to 2 ⁇ L MeOH and 200 ⁇ L of 5 ng/mL Terfenadine in methanol/acetonitrile (1:1, v/v). It was agitated for 1 minute and centrifuged at 4000 rpm for 15 minutes. A supernatant was separated and diluted with 5 volumes of MeOH/H 2 O (1;1, v/v, 0.1% FA) for analysis below.
- PK Pharmacokinetic Studies in Balb/c mice: The pharmacokinetic (PK) properties of compounds of the invention were obtained following a single intravenous (IV) bolus (e.g., 1 mg/kg) and a single oral gavage (PO) (e.g., 10 mg/kg) administration to female Balb/c mice (fed, ages within 5-7 weeks).
- IV intravenous
- PO oral gavage
- the vehicle was 5% DMA/5% EtOH/40% PEG 400/50% H 2 O (solution) and the blood samples were collected at 0 (pre-dose), 0.083, 0.25, 1, 4, 8, 12, 24, 48, 72, 96, and 120 hours post-dose.
- the vehicle was 1% HEC/0.25% Tween-80/0.05% Antifoam/H 2 O (suspension) and the blood samples were collected at 0 (pre-dose), 0.25, 1, 4, 8, 12, 24, 48, 72, 96, and 120 hours post- dose for PO.
- the plasma samples were prepared by centrifugation at 4oC and 3200xg for 10 minutes, and then quickly frozen over dry ice and kept at -60oC or lower until analysis.
- the concentrations of the test article and its potential metabolites (if detectable) in the plasma samples were determined by a validated liquid chromatography-tandem mass spectrometry (LC-MS/MS).
- KC-2253-Ba/F3 EGFR Del19-C797S-luc2 situ brain Model The cell line used was Ba/F3 EGFR Del19-C797S-luc2 (Kyinno, Cat.
- cKC-2253 engineered by introducing human EGFR containing Del19 and C797 mutations and luciferase reporter gene into Ba/F3 cells.
- Ba/F3 EGFR Del19-C797S-luc2 cells were maintained and expanded in RPMI-1640 containing 10% fetal bovine serum at 37oC, 5% CO 2 incubator. The cells growing in an exponential growth phase were harvested and counted for tumor inoculation.
- BALB/c nude mice female, 6-8 weeks, weighing approximately 18-22g) from Biocytogen (Beijing, China) were used. After one week of acclimation, animals were anesthetized by ip injection of Pentobarbital sodium (30 mg/kg).
- mice were prepared with an injection point in the skull located on the right parietal bone, 3-3.5 mm lateral to the sagittal suture, and 0.5 mm posterior to the coronal suture.
- Each mouse was inoculated intracranially with the Ba/F3 EGFR Del19-C797S-luc2 (5 ⁇ 10 4 cells in 2 ⁇ L PBS) via a microinjector. The needle was inserted to a depth of 4 mm, then retracted by 0.7 mm. After the injection, the mouse was kept warm until they recovered from the anesthesia. Animals were observed post-anesthesia until all animals recovered.
- mice were dosed with meloxicam (5 mg/kg, once daily for 3 days) after the mouse recovered from the anesthesia.
- meloxicam 5 mg/kg, once daily for 3 days
- the inoculated mice were weighed and were pre-anesthetized with 1.25% tribromoethanol solution (20 ⁇ l/g).
- D-Luciferin solution intraperitoneally injected D-Luciferin solution at a concentration of 15 mg/mL and a volume of 0.2 mL. All mice were measured for bioluminescence at 9 minutes after the luciferin administration and intensity of bioluminescence for each mouse was recorded.
- mice bearing established brain tumors based on bioluminescence were selected and randomized into 7 groups according to bioluminescence intensity and body weight. Mice were dosed once daily for 31 consecutive days with indicated test articles in suspension formulation with vehicle (1% HEC (w/v)/0.25% Twen-80 (v/v)/0.05% Antifoam (v/v)/H 2 O) or vehicle control by oral gavage. During dosing, mouse body weight was measured three times a week and tumor growth based on bioluminescence intensity was measured on day 3, 7, 10, 14, 17 and 31. After dosing stopped at day 32, mice were kept alive to observe extended survival until day 51 which study was terminated. The results showed that a compound of the invention exhibited excellent brain penetration with improved antitumor activity and survival.
- PC-9_Luc EGFR/del19 mutation
- PC-9_Luc human lung cancer cells derived from RCB4455 (RIKEN BioResource Center, Tsukuba, Japan) was used for the study.
- the cells were maintained in vitro as a monolayer culture in RPMI 1640 medium supplemented with 10% fetal bovine serum and 1% Antibiotic- Antimycotic at 37 °C in an atmosphere of 5% CO 2 in air.
- the cells were subcultured twice a week by trypsin-EDTA treatment.
- mice Female, 6-8 weeks, weighing approximately 18-22g were purchased from Zhejiang Vital River Laboratory Animal Co., LTD. and used for the study. After one week of acclimation, animals were anesthetized by ip injection of 1.25% avertin solution with dosing volume of 20 ⁇ L/g. The anesthetized mice were properly positioned. The head skin of the mouse was sterilized with 70% alcohol and was draped in a sterile fashion. A ⁇ 1cm length incision was made just at the right of the midline and anterior to the interaural line.
- mice were inoculated intracranially with the PC-9_Luc cells (3 ⁇ 10 5 cells in 3 ⁇ L PBS+20% Matrigel) at a depth of 3.5 mm via the microinjector. It was kept warm and observed post-anesthesia until recovered. For pain relief from surgery and tumor inoculation, the mouse was dosed orally with 10 mg/kg of meloxicam (10 ⁇ L/g ). All mice were treated again with 5 mg/kg meloxicam (10 ⁇ L/g) in the morning of the next day post-surgery. To monitor tumor growth and mouse status, the body weight was obtained. Bioluminescence measurements were performed by intraperitoneal injection of luciferin at 150 mg/kg.
- mice After 10 minutes of the luciferin administration, the mice were anesthetized before bioluminescence determination with IVIS (Lumina II). The bioluminescence of the whole animal body, including metastatic tumors, was determined and recorded. On day 7 post-tumor inoculation, mice were randomized into 7 groups based on the intensity of bioluminescence measured and body weight and dosed daily for 28 consecutive days with indicated test articles in suspension formulation with vehicle (1% HEC (w/v)/0.25% Twen-80 (v/v)/0.05% Antifoam (v/v)/H 2 O) or vehicle control by oral gavage. During dosing, mouse body weight was measured twice a week and tumor growth based on bioluminescence intensity was measured once a week.
- vehicle 1% HEC (w/v)/0.25% Twen-80 (v/v)/0.05% Antifoam (v/v)/H 2 O
- mice were observed for extended survival.
- the results showed that a compound of the invention showed very potent anti-tumor activity with improved survival.
- All the animals in the treatment groups dosed in the range of 0.6 to 6 mg/kg/day, survived for more than 60 days, while all the animals in the controlled group died before D24 post-treatment.
- OTHER EMBODIMENTS All of the features disclosed in this specification may be combined in any combination. Each feature disclosed in this specification may be replaced by an alternative feature serving the same, equivalent, or similar purpose. Thus, unless expressly stated otherwise, each feature disclosed is only an example of a generic series of equivalent or similar features.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Compounds of Formula (I): a pharmaceutically acceptable salt, a stereoisomer, or a racemate thereof. Each of R4, R5, R6, R7, R8, X, and Y is defined herein. Also disclosed are pharmaceutical compositions containing such a compound, methods of treating cancer or inhibiting EGFR using the compound.
Description
EGFR INHIBITORS CROSS REFERENCES TO RELATED APPLICATONS This application claims the benefit of priority based on US Application Series No. 63/470,633 filed on June 2, 2023, the content and disclosure of which is incorporated herein by reference in its entirety. TECHNICAL FIELD The present invention provides quinazoline compounds, pharmaceutical compositions thereof, their preparation methods, and use as EGFR inhibitors for treating cancer. BACKGROUND Epidermal growth factor receptor (EGFR) belongs to the HER family of receptors which comprise four related proteins EGFR, HER2, HER3, and HER4. The HER receptors are known to be activated by binding to different ligands including EGF (see Duggirala et.al., Biomol Ther, 2022, 30, 19). After binding to a ligand, the receptor forms functionally active dimers including EGFR-EGFR, EGFR-HER2, EGFR-HER3, and EGFR-HER4. Dimerization induces the activation of the tyrosine kinase domain of EGFR, leading to a series of downstream signaling pathways for cell proliferation, survival, and anti- apoptosis (see Uribe et al., Cancers 2021, 13, 2748). Dysregulations of EGFR signal transduction pathways, including EGFR gene mutations, can promote malignant transformation of normal cells into tumor cells and subsequently assist tumor cell proliferation, invasion, metastasis, and angiogenesis. In non-small cell lung cancer (NSCLC) patients, the two most common EGFR gene mutations are del19 (i.e., a short in-frame deletion in exon 19) and L858R (i.e., a single missense mutation in exon 21). See Konduri et.al., Cancer Discov, 2016, 6, 601. These two mutations trigger ligand independent EGFR activation, driving cancerous growth and metastasis of NSCLC (see Uribe et al., Cancers 2021, 13, 2748). Given the prominent importance of EGFR signaling in cancer development, several small-molecule EGFR tyrosine kinase inhibitors (TKIs) such as erlotinib, gefitinib, and the like, have been developed to inhibit oncogenic EGFR activity for treating NSCLC patients harboring del19 and L858R mutations. Nevertheless, almost all cancer patients developed resistance to EGFR-TKIs (erlotinib and gefitinib) after 10–14 months of treatment. The TKI resistance has been associated with a secondary mutation (T790M) in the EGFR’s kinase
domain in 50%–60% of the cases (see Pao et al., PLoS Med.2005, 2, e73 and Leonetti et al., British Journal of Cancer, 2019, 121, 725). To overcome T790M mutation resistance, osimertinib as a third-generation EGFR inhibitor was developed for treating patients carrying the EGFR T790M mutation. Unfortunately, acquired resistance to osimertinib inevitably occurs, relating to EGFR C797X mutations (wherein "X" can be an S, G, N, Y, T, or D), which occurs in exon 20 (see Papadimitrakopoulou et al., Annals of Oncology, 2018, 29, LBA51 and Bertoli et al., Int. J. Mol. Sci. 2022, 23, 6936). Tumors harboring double mutants, e.g., Del19/C797X and L858R/C797X, are no longer sensitive to osimertinib or other third-generation EGFR inhibitors. There is no approved medicine to treat patients carrying double mutants. Further, current EGFR inhibitors such as gefitinib and erlotinib are not effective in treating brain cancer due to their low ability to cross the blood-brain barrier. It is critical that EGFR inhibitors are capable of crossing the blood-brain barrier, as about 70% of NSCLC patients bearing mutant-EGFR develop brain metastases (see Subramaniam et al, Front Oncol.2018, 8, 208). Thus, there is an unmet medical need for effective EGFR inhibitors to penetrate blood- brain barrier and treat NSCLC patients having double EGFR mutants and brain metastasis. SUMMARY The present invention is based on an unexpected discovery that certain quinazoline compounds show promising anti-tumor activities. In one aspect, this invention relates to quinazoline compounds of Formula I:
I, in which one of X and Y is H, deuterium, or halo; the other of X and Y is
, in which R1 is F, C1-C6 alkyl, C3-C6 cycloalkyl, or 4-6 membered heterocyclyl, R2 is H, deuterium, F, C1-C6 alkyl, C3-C6 cycloalkyl, or 4-6 membered heterocyclyl, or R1 and R2, together with the carbon atom they bond to, are C3-C6
cycloalkyl or 4-6 membered heterocyclyl; R3 is 5-10 membered heteroaryl orC6-C10 aryl; and n is 1, 2, or 3; R4 is H, halo, cyano, C3-C6 cycloalkyl, ORa, 4-10 membered heterocyclyl, or 5-6 membered heteroaryl, in which Ra is C1-C6 alkyl, C3-C6 cycloalkyl, 4-10 membered heterocyclyl, or 5-6 membered heteroaryl; R5 is H or deuterium; R6 is H, C1-C6 alkyl, or C3-C6 cycloalkyl; R7 is 6-10 membered aryl or 5-10 membered heteroaryl; R8 is H, deuterium, halo, C1-C6 alkyl, or C3-C6 cycloalkyl; alkyl is optionally substituted with one or more groups selected from the group consisting of deuterium, halo, cyano, C1-C4 alkoxy, NR1aR1b, C3-C6 cycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, and 4-6 membered heterocyclyl, in which R1a and R1b, independently, is H, deuterium, C1-C4 alkyl, or C3-C6 cycloalkyl, or R1a and R1b, together with the nitrogen atom they bond to, are C4-C6 heterocyclyl; and each of cycloalkyl, heterocyclyl, aryl, and heteroaryl, independently, is optionally substituted with one or more groups selected from the group consisting of deuterium, halo, cyano, acetylenyl, C1-C4 alkyl, C1-C4 alkoxy, C3-C6 cycloalkyl, C3-C6 cycloalkoxy, 4-6 membered heterocyclyl, and NR1aR1b. Preferably, the above-described compounds have one or more of the following features: (i) n is 1; (ii) R1 is F, C1-C3 alkyl, C3-C6 cycloalkyl, preferably methyl, CF3, CHF2, cyclopropyl, or CD3, and more preferably, methyl; or R1 and R2, together with the carbon atom they bond to, are C3-C6 cycloalkyl, preferably cyclopropyl; (iii) R2 is H, F, C1-C3 alkyl, C3-C6 cycloalkyl, preferably methyl, CF3, CHF2, cyclopropyl, or CD3; (iv) R3 is
; preferably, R3 is pyrimidinyl, imidazolyl,
;
(vi) R7 is phenyl optionally substituted with one or more groups selected from the group consisting of F, Cl, Br and acetylenyl, preferably R7 is
. Further, R7 can also be 9- or 10- membered bicyclic heteroaryl optionally substitutes with halo (e.g., F). Examples include quinolinyl, indolizinyl, pyrazolo[1,5-a]pyridinyl, imidazo[1,2-a]pyridinyl, benzo[c]isothiazolyl, and benzo[d]isothiazolyl such as
(vii) R6 is H, CH3, CD3, or CHF2, preferably H or CHF2; (viii) R8 is H or deuterium, preferably H. Subsets of the compounds of Formula I are represented by Formulas IA and IB:
. Each of R1-R7 and n is defined above including any combinations of features (i) to (vii) as shown above. In addition, in Formula IA, R4 can be
Preferred compounds of Formula IA have the following features: (i) n is 1; (ii) R5 is H or deuterium; (iii) R1 is F, C1-C3 alkyl, or C3-C6 cycloalkyl, preferably methyl, CF3, CHF2, cyclopropyl, or CD3, and R2 is H, F, CH3, D, CD3, cyclopropyl, CF3, CHF2; or R1 and R2, together with the carbon atom they are bonded to, is cyclopropyl;
In Formula IB above, preferably n is 1, 2, or 3 and R1 is F, CH3, CD3, CF3, CHF2, cyclopropyl. Further in Formula IB, R4 can be
. Preferred compounds of Formula IB include the following features: (i) n is 1; (ii) R5 is H or deuterium; (iii) R1 is F, CH3, CD3, CF3, CHF2, cyclopropyl; (iv) R2 is H, CH3, D, CD3, F, cyclopropyl, CF3, or CHF2; (v) R3 is
(vii) R6 is H, CH3 or CHF2; and (viii) R7 is
. Another aspect of this invention is a method of treating cancer comprising administering to a subject in need thereof an effective amount of any one of the compounds described above. Nonlimiting examples of cancer treatable by the compounds of this invention include lung cancer (e.g., non-small cell lung cancer) and brain cancer. The current invention further includes use of such a compound (e.g., a pharmaceutical composition containing one of the compounds of Formula I described above) for treating cancer or for the manufacture of a medicament for treating cancer. Also within the scope of this invention is a method of inhibiting EGFR by administering to a subject in need thereof an effective amount of any one of the compounds described above. Still within the scope of this invention is a pharmaceutical composition comprising any of the compounds described above and a pharmaceutically acceptable carrier. As described above, the pharmaceutical composition is particularly useful in treating cancer. Table 1 below shows 65 exemplary compounds of the present invention, i.e., Compounds 1-65. Table 1
Preferred compounds include Compounds 1-4, 6-8, 10, 15-17, 20-24, 27, 28, 31-33, 35, and 43-65. The term “halo” herein refers to a fluoro, chloro, bromo, or iodo radical. Examples include a fluoro radical (F) and a bromo radical (Br). The term “alkyl” refers to a straight or branched hydrocarbon group, containing 1-20 carbon atoms (e.g., C1-6) and a monovalent radical center derived by the removal of a hydrogen atom from a carbon atom of a parent alkane. Exemplary alkyl groups are methyl, ethyl, n- propyl, isopropyl, n-butyl, iso-butyl, tert-butyl, n-pentyl, and n-hexyl. The term “haloalkyl” refers to alkyl substituted with one or more halo atoms. Examples include fluoromethyl, difluoromethyl, trifluoromethyl, fluoroethyl (e.g., 1-fluoroetyl and 2-fluoroethyl), difluoroethyl (e.g., 1,1-, 1,2-, and 2,2-difluoroethyl), and trifluoroethyl (e.g., 2,2,2- trifluoroethyl). The term “alkyl” as used herein includes haloalkyl. The term “alkoxy” refers to an –O–alkyl group. Examples are methoxy, ethoxy, propoxy, and isopropoxy. Alkoxy also includes haloalkoxy, namely, alkoxy substituted with one or more halogens, e.g., –O-CH2Cl and –O-CHClCH2Cl. The term “cycloalkyl” refers to a nonaromatic, saturated or unsaturated monocyclic, bicyclic, tricyclic, or tetracyclic hydrocarbon group containing 3 to 12 carbons (e.g., C3-6 and C3-10). Examples are cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl. The term “heterocyclyl” refers to a nonaromatic, saturated or unsaturated, 3–8 membered monocyclic, 8–12 membered bicyclic, or 11–14 membered tricyclic ring system having one or more heteroatoms (e.g., O, N, P, and S). Examples include aziridinyl, azetidinyl, pyrrolidinyl, dihydrofuranyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiophenyl, tetrahydro-2-H-thiopyran-1,1-dioxidyl, piperazinyl, piperidinyl, morpholinyl, imidazolidinyl, azepanyl, dihydrothiadiazolyl, dioxanyl, and quinuclidinyl. Both “cycloalkyl” and “heterocyclyl” also include fused, bridged, and spiro ring systems. They further include substituted groups such as halocycloalkyl and haloheterocyclyl.
The term “aryl” refers a 6-carbon monocyclic, 10-carbon bicyclic, 14-carbon tricyclic aromatic ring system wherein each ring can have one or more (e.g., 1 to 10, 1 to 5, and 1 to 3) substituents. Examples include phenyl, biphenyl, 1‑ or 2-naphthyl, 1,2-dihydronaphthyl, 1,2,3,4-tetrahydronaphthyl, indenyl, and indanyl. The term “aralkyl” refers to alkyl substituted with an aryl group. The term "heteroaryl" refers to an aromatic 5–8 membered monocyclic, 8–12 membered bicyclic, or 11–14 membered tricyclic ring system having one or more heteroatoms (e.g., O, N, P, and S). Examples include pyridinyl, pyrimidinyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzoxazolyl, benzothiophenyl, benzofuranyl, pyrazolyl, triazolyl, oxazolyl, thiadiazolyl, tetrazolyl, oxazolyl, isoxazolyl, carbazolyl, furyl, imidazolyl, thienyl, thiazolyl, and benzothiazolyl. The term “heterocyclylalkyl” refers to an alkyl group substituted with a heterocyclyl or heteroaryl group. Alkyl, alkoxyl, cycloalkyl, heterocyclyl, aryl, aralkyl, heterocyclylalkyl, and heteroaryl mentioned herein include both substituted and unsubstituted moieties, unless specified otherwise. Examples of a substituent include deuterium (D), hydroxyl (OH), halo (e.g., F and Cl), amino (NH2), cyano (CN), nitro (NO2), alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, acylamino, alkylamino, aminoalkyl, haloalkyl (e.g., trifluoromethyl), heterocyclyl, alkoxycarbonyl, amido, carboxy (COOH), alkanesulfonyl, alkylcarbonyl, alkenylcarbonyl, carbamido, carbamyl, carboxyl, thioureido, thiocyanato, sulfonamido, aryl, arylamino, aralkyl, and heteroaryl. All substitutes can be further substituted. The term “compound”, when referring to a compound of this invention, also includes its salts, solvates, and prodrugs. The pharmaceutically acceptable salts include those listed in Handbook of Pharmaceutical Salts: Properties, Selection and Use, 2nd Revised Edition, P. H. Stahl and C. G. Wermuth (Eds.), Wiley-VCH, New York, (2011). In addition to pharmaceutically acceptable salts, other salts are contemplated in the invention. They may serve as intermediates in the purification of compounds or in the preparation of other pharmaceutically acceptable salts, or are useful for identification, characterization or purification of compounds of the invention. A solvate refers to a complex formed between an active compound and a pharmaceutically acceptable solvent. Examples of a pharmaceutically acceptable solvent include water, ethanol, isopropanol, ethyl acetate, acetic acid, and ethanolamine. A prodrug refers to a compound that, after administration, is metabolized into a pharmaceutically active drug. Examples of a prodrug include esters and other pharmaceutically acceptable derivatives.
The compounds of the present invention may contain one or more non-aromatic double bonds or asymmetric centers. Each of them occurs as a racemate or a racemic mixture, a single R enantiomer, a single S enantiomer, an individual diastereomer, a diastereometric mixture, a cis-isomer, or a trans-isomer. Compounds of such isomeric forms are within the scope of this invention. They can be present as a mixture or can be isolated using chiral synthesis or chiral separation technologies. The term “treating” or “treatment” refers to administering one or more of the compounds to a subject with the purpose to confer a therapeutic effect, e.g., to slow, interrupt, arrest, control, or stop of the progression of an existing disorder and/or symptoms thereof, but does not necessarily indicate a total elimination of all symptoms. “An effective amount” refers to the amount of a compound that is required to confer the therapeutic effect. Effective doses will vary, as recognized by those skilled in the art, depending on the types of symptoms treated, route of administration, excipient usage, and the possibility of co-usage with other therapeutic treatment. Cancer treatable with a compound of this invention includes those caused by KRAS mutation, SOS1 oncogenic mutation, or oncogenic mutation/overexpression of receptor tyrosine kinases such as EGFR, FGFR, etc. Nonlimiting examples include pancreatic cancer, lung cancer, colorectal cancer, cholangiocarcinoma, multiple myeloma, melanoma, uterine cancer, endometrial cancer, thyroid cancer, acute myeloid leukemia, bladder cancer, urothelial cancer, gastric cancer, cervical cancer, head and neck squamous cell carcinoma, diffuse large B cell lymphoma, esophageal cancer, chronic lymphocytic leukemia, hepatocellular cancer, breast cancer, ovarian cancer, prostate cancer, glioblastoma, renal cancer and sarcoma, and brain metastasis. The compounds of this invention are particularly effective in treating cancer associated with EGFR such as brain cancer, lung cancer (e.g., non-small cell lung cancer, and non-small cell lung cancer with brain metastasis or leptomeningeal). The term “subject” refers to an animal including human or non-human, such as a mammal. A human is a preferred subject. A compound of this invention may be administered alone or in the form of a pharmaceutical composition with pharmaceutically acceptable carriers, diluents or excipients. Such pharmaceutical compositions and processes for making the same are known in the art (See, e.g., Remington: The Science and Practice of Pharmacy, A. Adejare, Editor, 23rd Edition., Academic Press, 2020).
To practice the method of the present invention, a composition or a kit containing one or more of the above-described compounds can be administered alone or co-administered with at least one other pharmacologically active substance simultaneously, concurrently, sequentially, successively, alternately, or separately. Simultaneous administration, also referring to as concomitant administration, includes administration at substantially the same time. Concurrent administration includes administering the active agents within the same general time period, for example on the same day(s) but not necessarily at the same time. Alternate administration includes administration of one agent during a time period, for example over the course of a few days or a week, followed by administration of the other agent(s) during a subsequent period of time, for example over the course of a few days or a week, and then repeating the pattern for one or more cycles. Sequential or successive administration includes administration of one agent during a first time period (for example over the course of a few days or a week) using one or more doses, followed by administration of the other agent(s) during a second and/or additional time period (for example over the course of a few days or a week) using one or more doses. An overlapping schedule may also be employed, which includes administration of the active agents on different days over the treatment period, not necessarily according to a regular sequence. Variations on these general guidelines may also be employed, e.g., according to the agents used and the condition of the subject. The elements of the combinations of this invention may be administered (whether dependently or independently) by methods customary to the skilled person, e.g., by oral, enteral, parenteral, nasal, vaginal, rectal, or topical routes of administration and may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, excipients and/or vehicles appropriate for each route of administration. The term “parenteral” as used herein refers to subcutaneous, intracutaneous, intravenous, intraperitoneal, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional, or intracranial injection, as well as any suitable infusion technique. A composition for oral administration can be any orally acceptable dosage form including capsules, tablets, emulsions and aqueous suspensions, dispersions, and solutions. In the case of tablets, commonly used carriers include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions or emulsions
are administered orally, the active ingredient can be suspended or dissolved in an oily phase combined with emulsifying or suspending agents. If desired, certain sweetening, flavoring, or coloring agents can be added. A nasal aerosol or inhalation composition can be prepared according to techniques well known in the art of pharmaceutical formulation. For example, such a composition can be prepared as a solution in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents. A composition having one or more of the above-described compounds can also be administered in the form of suppositories for rectal administration. The carrier in the pharmaceutical composition must be “acceptable” in the sense that it is compatible with the active ingredient of the composition (and preferably, capable of stabilizing the active ingredient) and not deleterious to the subject to be treated. One or more solubilizing agents can be utilized as pharmaceutical excipients for delivery of an active compound. Examples include colloidal silicon oxide, magnesium stearate, cellulose, sodium lauryl sulfate, and D&C Yellow # 10. The details of one or more embodiments of the invention are set forth in the description below. Other features, objects, and advantages of the invention will be apparent from the description and from the claims. DETAILED DESCRIPTION The present invention is based on a surprising discovery that the compounds of Formula I reproduced below are effective in inhibiting EGFR and treating cancer.
I Variables R1-R7, X, and Y are defined above. Further, in a subset of the compounds of formula I, R3 is
Preferably, R3 is
. In another subset of the compounds of formula I, R4 is
. In a third subset of the compounds of formula I, R7 is phenyl, and R7 is optionally substituted with one or more groups selected from the groups consisting of F, Cl, Br and acetylenyl; Preferably, R7 is
. More preferably, R7 is
. In another subset of the compounds of formula I, R7 is 9- or 10- membered bicyclic heteroaryl, preferably quinolinyl, indolizinyl, pyrazolo[1,5-a]pyridinyl, imidazo[1,2- a]pyridinyl, benzo[c]isothiazolyl or benzo[d]isothiazolyl; and R7 is optionally substituted with halo (for example F); Preferably, R7 is .
. In a preferred subset of the compounds of formula IA:
. The compounds of the invention are useful in treating mutant EGFR mediated cancer that has metastasized to brain and central nervous system. They are also suitable for treating mutant EGFR mediated local or metathesized lung cancer. The compound can be a compound of Formula I, IA, or IB. The compounds of Formula I can be prepared by synthetic methods well known in the art. See, e.g., R. Larock, Comprehensive Organic Transformations (3rd Ed., John Wiley and Sons 2018); P. G. M. Wuts and T. W. Greene, Greene’s Protective Groups in Organic Synthesis (4th Ed., John Wiley and Sons 2007); L. Fieser and M. Fieser, Fieser and Fieser’s Reagents for Organic Synthesis (John Wiley and Sons 1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis (2nd ed., John Wiley and Sons 2009) and subsequent editions thereof. The compounds thus prepared can be purified following conventional methods such as crystallization, distillation/vacuum distillation, flash chromatography over silica, and preparative liquid chromatography. Efficacy of the compounds of this invention can be initially determined using cellular pERK potency assay or 2D cell proliferation assay, all described below. The selected
compounds can be further tested to verify their efficacy, e.g., by administering it to an animal. Based on the results, an appropriate dosage range and administration route can be determined. A compound of this invention is preferably formulated into a pharmaceutical composition containing a pharmaceutical carrier. The pharmaceutical composition is then given to a subject in need thereof to inhibit mutant EGFR (e.g. del19, L858R, del19/C797S, del19/L858R, amplification) thus treating cancer. Without further elaboration, it is believed that one skilled in the art can, based on the above description, utilize the present invention to its fullest extent. The following examples are to be construed as merely illustrative and not limitative in any way whatsoever. All publications cited herein are hereby incorporated by reference in their entirety. Set forth below are examples illustrating preparation and efficacy evaluation of compounds of this invention. The abbreviations as used herein are provided in Table 2 below with their definitions, which are used conventionally in the art. Table 2
Schemes I to V depict five exemplary routes for preparing a compound of the invention (In the following schemes, Ar is R7).
Scheme I
Scheme III
The compounds of the invention can be prepared by combining the reactions in these general synthetic schemes or additional reaction schemes specifically depicted herein or reactions known in the art and not limited to the general schemes described herein. General Synthetic Procedures Provided below are exemplary processes for preparing compounds of Formula I (e.g., IA and IB). These processes provide further features of the invention and are illustrated below. Generally, compounds of formula IA and IB can be synthesized using the procedures illustrated in Schemes I to V above. As shown in Scheme I, quinazolinone IA-2 can be prepared by reacting benzonitrile derivative IA-1 with formic acid in the presence of H2SO4. Quinazolinone IA-2 is then converted to chloride IA-3 using oxalyl chloride, thionyl chloride, or POCl3 in a suitable solvent such as chloroform. The chlorine atom in IA-3 is displaced by IA-4 (i.e., H2N-Ar) to yield IA-5 in a suitable solvent such as isopropanol. The fluorine atom in IA-5 is displaced by chiral or achiral alcohol IA-6 in the presence of a base, such as NaH or t-BuOK, in a suitable solvent such as THF to yield ether IA-7. Finally, R4 is introduced using palladium mediated coupling with borate, boronic acid, or boronate ester derivatives such as IA-8a or IA-8b in a suitable solvent (e.g., dioxane and water) to provide a compound of formula IA-9. Furthermore, reduction of the halide of IA-7 with Et3SiH in the presence of a palladium catalyst or catalytic hydrogen reduction in a suitable solvent such as dioxane can provide a compound of Formula IA-10. Compounds of formula IA can also be prepared following Scheme II, starting with a reaction between benzonitrile derivative IA-1 and DMF-DMA to generate imine IA-11, which undergoes ring formation in the presence of H2N-Ar (IA-4) in acetic acid to form quinazoline derivative IA-12. The fluorine atom in IA-12 is displaced with chiral or achiral alcohol IA-6 in the presence of a base (e.g., NaH and t-BuOK) in a suitable solvent such as THF to generate ether IA-13. Finally, R4 is introduced using palladium mediated coupling with borate, boronic acid or boronate ester derivatives (e.g., IA-8a and IA-8b) in a suitable solvent such as dioxane and water to provide a compound of formula IA-9. Alternatively, nucleophilic displacement
reaction with an amine or heteroaryl R4 (e.g., pyrazole and imidazole or their derivatives) in a suitable solvent such as dioxane provides a compound of formula IA-9. Furthermore, the bromine atom in intermediate IA-13 can be displaced using metal halide (e.g., cuprous iodide) mediated coupling with heteroaryl-OH derivatives (IA-8d) such as 1-methyl-1H-pyrazol-4-ol, in the presence of a suitable ligand such as 3,4,7,8-tetramethyl-1,10-phenanthroline in a suitable solvent such as toluene to generate compound of formula 1A-9. The bromine atom in intermediate IA-13 can also be displaced by HO-alkyl (IA-8d) group via palladium mediated C-O bond formation using RockPhos Pd G3 (CAS# 2009020-38-4) catalyst. Compounds of formula IB can be synthesized following Scheme III. The fluorine atom in nitrobenzene derivative IB-1 is displaced by chiral or achiral alcohol IA-6 in the presence of a base (e.g., NaH and tBuOK) in a suitable solvent such as THF and DMF to yield nitrobenzene derivative IB-2. The nitro group in IB-2 is selectively reduced with Fe or Zn and NH4Cl in a suitable solvent such as ethanol and water, to provide intermediate amino IB-3, which reacts with formamidine acetate in the presence of pTSA in a suitable solvent such as ethanol, to generate quinazolinone derivative IB-4. Quinazolinone derivative IB-4 is converted to chloride IB-5 using oxalyl chloride, thionyl chloride or POCl3 in a suitable solvent such as CHCl3. The chlorine atom in IB-5 is displaced by IA-4 (i.e., H2N-Ar) to provide intermediate IB-6. Finally, R4 is introduced using palladium mediated coupling with borate, boronic acid or boronate ester derivatives (e.g., IA-8a or IA-8b) in a suitable solvent such as dioxane and water to yield the final product of formula IB-7. Some of the compounds of formula IB can be synthesized following scheme IV. IB-8 is etherified using a Mitsunobu reaction using reagents such as diethyl azodicarboxylate/Ph3P or 1,1′-(azodicarbonyl)dipiperidine/n-Bu3P with chiral or achiral alcohol IA-6 to yield a compound of formula IB-9. Compounds of formula IB can also be synthesized following scheme V. Phenol derivative IB-10 is converted to a corresponding carbonate IB-11 using ethyl carbonochloridate in a suitable solvent such as dichloromethane. Nitration of the carbonate IB-11 in a mixture of H2SO4 and HNO3 affords nitro derivative IB-12, which is hydrolyzed using a base such as NaHCO3 in a suitable solvent such as methanol to from phenol IB-13. The phenol IB-13 is converted to vinyl ether using di-μ-chlorobis[(1,2,5,6-η)-1,5-cyclooctadiene]diiridium mediated vinylation using vinyl acetate in a suitable solvent such as toluene to form intermediate IB-14. The vinyl group in IB-14 is converted to cyclopropyl derivative IB-15 using diiodomethane and diethyl zinc in a suitable solvent such as DCM. The bromo group in
IB-15 is displaced with CuCN in a suitable solvent such as NMP to form the cyano intermediate IB-16. The fluorine atom in IB-16 is displaced with chiral or achiral alcohol IA-6 in the presence of a base (e.g., NaH and t-BuOK) in a suitable solvent such as THF to generate ether IB-17. The nitro group in IB-17 is selectively reduced with sodium dithionite in a suitable solvent such as tetrahydrofuran and water, to provide intermediate amino IB-18, which reacts with (E)-N,N'-bis(3-chloro-2-fluorophenyl)formimidamide (IB-19) in the presence of acetic acid in a suitable solvent such as 2-methyl tetrahydrofuran, to generate quinazolinone derivative of formula IB-20. All chemicals, reagents, and solvents were purchased from commercial sources when available and were used as received without further purification. Air- and moisture-sensitive reactions were carried out under an inert atmosphere of argon or nitrogen in oven-dried glassware. The reactions were monitored using analytical thin layer chromatography (TLC). The developed TLC plates were visualized by UV light (λ = 254, 365 nm) or immersion in alkaline potassium permanganate solution followed by heating on a hot plate. All reactions were stirred magnetically at ambient temperature unless otherwise indicated.1H NMR spectra were recorded on Agilent Technologies or VARIAN, 400 MHz NMR spectrometer.1H NMR spectra were reported in parts per million (δ) relative to TMS (0.0), DMSO-d6 (2.50) or CD3OD (4.80) as an internal reference. 1H NMR data are reported as follows: chemical shift in ppm; multiplicity (s = singlet, d = doublet, t = triplet, q = quartet, quint = quintet, m = multiplet/complex pattern, td = triplet of doublets, ddd = doublet of doublet of doublets, br = broad signal); coupling constants (J) in Hz, integration. Mass spectral data was collected on SHIMADZU, LCMS-2020. Column chromatography is performed with regular gravity or flash chromatography, or pre-packed silica gel cartridges using a medium pressure chromatography apparatus (SANTAI Technologies, SepaBean machine U200), eluting with the solvent or solvent mixture indicated in the respective experiment. Preparative high performance liquid chromatography (HPLC) is performed using a reverse phase column (SHIMADZU, Column, Boston Analytics, Inc, Green ODS C18, 21.2 x 250 mm) of a size appropriate to the quantity of material being separated, generally eluting with a gradient of increasing concentration of methanol or acetonitrile in water, containing 0.1% trifluoroacetic acid or 0.1% formic acid or 0.04% ammonium bicarbonate in water/MeCN at a rate of elution suitable to the column size and separation to be achieved. Chemical names are generated using ChemDraw Professional version 19.1.
EXAMPLES The following intermediates may be used to prepare compounds of the present invention. Synthesis of 5-fluoroquinolin-6-amine (1A-4-1)
To a solution of quinolin-6-amine (4 g, 27.76 mmol, 1 eq) in dioxane (120 mL) was added NaHCO3 (7 g, 83.24 mmol, 3 eq). Under stirring at 40 °C, SelectFluorTM (1-chloro- methyl-4-fluoro-1,4-diazoniabicyclo[2.2.2]octane bis(tetrafluoroborate), 12.8 g, 36 mmol, 1.3 eq) was added to the above solution in one portion. The resultant mixture was stirred at 40 °C for 4 h under nitrogen, cooled to room temperature, and filtered thought celite® with washing by EtOAc (50 mL x 3). The filtrate was concentrated under reduced pressure and purified by silica gel column chromatography, eluting with PE/DCM (1:1) to afford the title compound (1.95 g, 43% yield) as a brown solid. LCMS: m/z = 163.25 [M+1]+. Synthesis of 7-fluorobenzo[d]isothiazol-6-amine (1A-4-2)
Step 1. Synthesis of 6-bromo-7-fluorobenzo[d]isothiazole To a solution of 4-bromo-2,3-difluorobenzaldehyde (7 g, 32 mmol, 1 eq) in DMF (70 mL) was added S8 (1.12 g, 35 mmol, 1.1 eq) and ammonium hydroxide (3.5 mL). The mixture was stirred at 100 °C for 16 h. After cooling to rt, it was diluted with H2O (100 mL) and extracted with EtOAc (100 mL x 3). The combined organic layers were washed with brine (60 mL x 3), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure and purified by silica gel column chromatography (PE/EA (20:1)) to afford the title compound (2 g, 27.3% yield) as a yellow solid. LCMS: m/z = 231.85/233.85 [M+1]+. Step 2. Synthesis of tert-butyl (7-fluorobenzo[d]isothiazol-6-yl)carbamate A mixture of 6-bromo-7-fluorobenzo[d]isothiazole (1.5 g, 6.5 mmol, 1 eq), tert-butyl carbamate (838 mg, 7.1 mmol, 1.1 eq), Cs2CO3 (6.35 g, 19 mmol, 3 eq), Xantphos (564 mg,
0.97 mmol, 0.15 eq) and Pd2(dba)3 (297 mg, 0.32 mmol, 0.05 eq) in toluene (20 mL) was degassed and purged with nitrogen and the mixture was stirred at 100 °C for 14 h. After cooling to rt, the mixture was diluted with H2O (20 mL) and extracted with DCM (20 mL x 3). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure and purified by silica gel column chromatography (PE/EA (15:1)) to afford the title compound (820 mg, 47.7% yield) as a yellow solid. LCMS: m/z = 268.95 [M+1]+. Step 3. Synthesis of 7-fluorobenzo[d]isothiazol-6-amine To a solution of tert-butyl (7-fluorobenzo[d]isothiazol-6-yl)carbamate (1.1 g, 4 mmol, 1 eq) in dioxane (25 mL) was added HCl/dioxane (15 mL). The mixture was stirred at 40 °C for 5 h under nitrogen. The reaction mixture was concentrated under vacuum to afford the title compound (700 mg, 99% yield) as a yellow solid. LCMS: m/z = 169.05 [M+1]+. Synthesis of 4-fluoropyrazolo[1,5-a] pyridin-5-amine (1A-4-3)
Step 1. Synthesis of tert-(3-fluoropyridin-4-yl) carbamate To a solution of 3-fluoropyridin-4-amine (4 g, 36 mmol, 1 eq) in THF (40 mL) was added di-tert-butyl dicarbonate (9.37 g, 43 mmol, 1.2 eq). After being stirred at rt for 6 h, the reaction mixture was condensed to dryness. The residue was suspended in petroleum ether (60 ml) and then stirred for 1 h, filtered to give the title compound (7.6 g, 98 % yield) as a white solid. LCMS: m/z = 213.2 [M+1] +. Step 2. Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)-4-fluoropyrazolo[1,5-a]pyridine-3- carboxylate
To a solution of tert-butyl (3-fluoropyridin-4-yl)carbamate (1 g, 4.7 mmol, 1 eq) in MeCN (20 mL) was added O-(2,4-dinitrophenyl)hydroxylamine (938 mg, 4.7 mmol, 1 eq) at room temperature, the mixture was stirred at 40 °C for 12 h under N2. The solvent was removed under vacuum to afford the crude residue. To the crude residue was added dry DMF (20 mL), K2CO3 (1.95 g, 14.13 mmol, 3 eq) and ethyl propiolate (693.4 mg, 7 mmol, 1.5 eq). The suspension was stirred at room temperature for 12 h. The reaction was quenched with water and extracted with EtOAc (100 mL x 3). The organic phases were washed with water (50 mL x 3) and brine (50 mL), dried over Na2SO4, concentrated under reduced pressure. The residue was purified by FCC (eluted with 0%~20% EA in PE) to afford ethyl 5-((tert-butoxycarbonyl) amino)-4-fluoropyrazolo[1,5-a] pyridine-3-carboxylate (400 mg, 22 % yield) as a white solid. LCMS: m/z = 324.13 [M+1]+. Step 3. Synthesis of 5-((tert-butoxycarbonyl)amino)-4-fluoropyrazolo[1,5-a]pyridine-3- carboxylic acid Ethyl 5-((tert-butoxycarbonyl) amino)-4-fluoropyrazolo[1,5-a] pyridine-3-carboxylate (1.3 g, 4 mmol, 1 eq) and LiOH (290 mg,12 mmol, 3 eq) were dissolved in THF/H2O/MeOH (1:1:1= 8mL:8mL:8mL). The mixture was stirred at 60 °C for 3 h. The reaction mixture was acidified with 2N aqueous HCl to pH 5~6, extracted with EtOAc (50 mL x 3), dried over Na2SO4, concentrated under reduced pressure to afford 5-((tert-butoxycarbonyl)amino)-4- fluoropyrazolo[1,5-a] pyridine-3-carboxylic acid (800 mg, 67% yield) as a white solid. LCMS: m/z = 296.10 [M+1]+. Step 4. Synthesis of 4-fluoropyrazolo[1,5-a] pyridin-5-amine A solution of 5-((tert-butoxycarbonyl)amino)-4-fluoropyrazolo[1,5-a]pyridine-3- carboxylic acid (800 mg, 2.71 mmol, 1 eq) in 40% aqueous H2SO4 (10 mL) was stirred at 100 °C for 18 h. The solution was cooled to 0 °C and ice water (10 mL) was added. The reaction mixture was neutralized to pH 7 by 6M NaOH solution and extracted with DCM (100 mL x 2). The combined organic extracts were washed with brine (10 mL) and dried over Na2SO4. The solvent was evaporated under reduced pressure to afford 4-fluoropyrazolo[1,5-a] pyridin-5- amine (370 mg, 90 % yield) as a brown solid. LCMS: m/z = 152.06 [M+1]+. Synthesis of 1-(pyrimidin-4-yl)ethanol (1A-6-1)
Step 1. Synthesis of 1-(pyrimidin-4-yl)ethanol To a solution of 1-(pyrimidin-4-yl)ethanone (300 mg, 2.45 mmol, 1 eq) in MeOH (10 mL) was added NaBH4 (139 mg, 3.68 mmol, 1.5 eq) at 0 °C. The mixture was stirred at room temperature for 1 h under nitrogen, then diluted with H2O (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (PE:EtOAc = 2:1) to afford the title compound (180 mg, 59% yield) as colorless oil. Synthesis of cyclopropyl(pyrimidin-2-yl)methanol (1A-6-2)
To a solution of 2-iodopyrimidine (500 mg, 2.43 mmol, 1.1 eq) in dry THF (3 mL) was added i-PrMgBr·LiCl (2.2 mL, 2.87 mmol, 1.3 eq, 1.3 M) dropwise at 0 °C, and stirred for 0.5 h under nitrogen. The solution of cyclopropanecarbaldehyde (155 mg, 2.2 mmol, 1 eq) in dry THF (2 mL) was added and then stirred at 0 °C for 1 h. The mixture was cooled to room temperature and quenched with saturated aqueous NH4Cl (10 mL) solution, extracted with DCM (20 mL x 2) and washed with brine (10 mL). The combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (DCM : MeOH = 96 : 4) to afford the title compound (150 mg, 45% yield) as yellow oil. LCMS: m/z = 151.30 [M+1]+. Synthesis of (E)-N,N'-bis(3-chloro-2-fluorophenyl)formimidamide (1B-19)
A solution of 3-chloro-2-fluoroaniline (2 g, 14 mmol, 1 eq), trimethoxymethane (642 mg, 6 mmol, 0.44 eq) and AcOH (36 mg, 0.6 mmol, 0.044 eq) in hexane (40 mL) was stirred at 48 °C for 16 h under nitrogen. The mixture was cooled to 20 °C and stirred at 20 °C for 3 h. The mixture was filtered. The filter cake was washed with hexane (3 x 10 mL) and concentrated under reduced pressure to afford the title compound (660 mg, 16% yield) as a white solid.
1H NMR (400 MHz, DMSO-d6) δ 9.98 (s, 1H), 8.09 (s, 1H), 7.24 – 7.00 (m, 6H). Synthesis of 2-(pyrimidin-2-yl)propan-2-ol (1A-6-3)
To a solution of methyl pyrimidine-2-carboxylate (2 g, 14.5 mmol, 1 eq) in THF (20 mL) was added methyl magnesium bromide (19.3 mL, 57.9 mmol, 4 eq) at 0 °C and stirred at room temperature for 2 h. The mixture was quenched with saturated NH4Cl (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated in vacuo and purified by column chromatography (PE:EA=3:2) to afford the title compound (700 mg, 34% yield) as yellow oil. LCMS: m/z = 139.30 [M+1]+. Synthesis of (S)-1-(pyrimidin-4-yl)ethan-1-ol (1A-6-4)
To a solution of HCO2H (396 mg, 8.6 mmol, 3.5 eq) was added TEA (745 mg, 7.4 mmol, 1 eq) dropwise at 0 °C, followed by the addition of a solution of 1-(pyrimidin-4- yl)ethan-1-one (300 mg, 2.46 mmol, 1 eq) in DCM (8 ml) and RuCl(p-cymene)[(S,S)-Ts- DPEN] (47 mg, 0.074 mmol, 0.03 eq). The reaction mixture was stirred at rt for 2 h under nitrogen. The mixture was concentrated at 25 °C under reduced pressure. The residue was purified by silica gel column chromatography, eluting with (DCM:MeOH= 99:1 ) to afford the title compound (283 mg, 93% yield, 66% ee) as black oil. LCMS: m/z = 125.25 [M+1]+. Synthesis of 1-(5-(difluoromethyl)pyrimidin-2-yl)ethan-1-ol (1A-6-5)
Step 1. Synthesis of 2-chloro-5-(difluoromethyl)pyrimidine To a solution of 2-chloropyrimidine-5-carbaldehyde (3 g, 21 mmol, 1 eq) in CHCl3 (60 mL) was added DAST (3.39 g, 21 mmol, 1 eq). The mixture was stirred at 70 °C for 1 h. After cooling to rt, the resulting mixture was diluted with NaHCO3 (50 mL) and extracted with DCM (60 mL x 2). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure and purified by silica gel column chromatography, eluting with PE/EA (15:1) to afford the title compound (2.4 g, 69.4% yield) as a white solid. LCMS: m/z = 165.00 [M+1]+. Step 2. Synthesis of 1-(5-(difluoromethyl)pyrimidin-2-yl)ethan-1-one To a solution of 2-chloro-5-(difluoromethyl)pyrimidine (1.2 g, 7.29 mmol, 1 eq) and tributyl(1-ethoxyethenyl)stannane (3.42 g, 9.48 mmol, 1.3 equiv.) in 1,4-dioxane (20 mL) was added Pd(PPh3)2Cl2 (255.9 mg, 0.36 mmol, 0.05 equiv.). The reaction mixture was degassed, purged with nitrogen 3 times and the mixture was stirred at 100 °C for 16 h under nitrogen. The reaction solution was cooled to room temperature. KF aqueous solution (40 mL) was added and stirred for 1 h. The mixture was extracted with EA (40 mL x 3). To the combined organic layers was added 4N HCl aqueous solution (20 mL) and stirred for 6 h at rt. The resulting mixture was diluted with saturated NaHCO3 (20 mL) and extracted with EtOAc (20 mL x 3), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure and purified by silica gel column chromatography, eluting with PE/EA (3:1) to afford the title compound (820 mg, 65.3% yield) as a white solid. LCMS: m/z = 173.10 [M+1]+. Step 3. Synthesis of 1-(5-(difluoromethyl)pyrimidin-2-yl)ethan-1-ol To a solution of 1-(5-(difluoromethyl)pyrimidin-2-yl)ethan-1-one (620 mg, 3.6 mmol, 1 eq) in MeOH (10 mL) was added NaBH4 (164 mg, 4.325 mmol, 1.2 eq) in one portion. The mixture was stirred at 0 °C for 0.1 h under nitrogen. The resulting mixture is quenched with H2O (10 mL) and extracted with DCM (2 x 50 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure and purified by silica gel column chromatography, eluting with PE/EA (1:1) to afford the title compound (420 mg, 67 % yield) as white oil. LCMS: m/z = 175.00 [M+1]+.
Synthesis of 1-(4-(difluoromethyl)phenyl)ethan-1-ol (1A-6-6)
Step 1. Synthesis of 1-(4-(difluoromethyl)phenyl)ethan-1-one To a solution of 1-bromo-4-(difluoromethyl)benzene (1 g, 4.8 mmol, 10 equiv.) and tributyl(1-ethoxyethenyl)stannane (2.27 g, 6.28 mmol, 1.3 equiv.) in 1,4-dioxane (20 mL) was added Pd(PPh3)2Cl2 (169 mg, 0.24 mmol, 0.05 equiv.). The reaction mixture was degassed and purged with nitrogen 3 times and stirred at 100 °C for 16 h under nitrogen. The reaction solution was cooled to room temperature. A KF aqueous solution (20 mL) was added and stirred for 1 h. The solution was extracted with EA (30 mL x 3). To the combined organic layers was added 1N HCl aqueous solution (40 mL). The mixture was stirred for 1 h, diluted with saturated NaHCO3 (80 mL) and extracted with EtOAc (100 mL x 3). The combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure and purified by silica gel column chromatography, eluting with PE/EA (10:1) to afford the title compound (710 mg, 86.3% yield) as colorless oil. 1H NMR (400 MHz, CDCl3) δ 8.02 (d, J = 8.0 Hz, 2H), 7.60 (d, J = 8.0 Hz, 2H), 6.68 (t, J = 56.0 Hz, 1H), 2.62 (s, 3H) ppm. Step 2. Synthesis of 1-(4-(difluoromethyl)phenyl)ethan-1-ol To a solution of 1-(4-(difluoromethyl)phenyl)ethan-1-one (930 mg, 5.47 mmol, 1 eq) in MeOH (15 mL) was added NaBH4 (414 mg, 10.94 mmol, 2 eq) in one portion. The mixture was stirred at 0 °C for 1 h under nitrogen. The resulting mixture was quenched with H2O (10 mL) and extracted with DCM (50 mL x 2). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure and purified by silica gel column chromatography, eluting with PE/EA (3:1) to afford the title compound (850 mg, 90.4% yield) as colorless oil. 1H NMR (400 MHz, CDCl3) δ 7.59-7.38 (m, 4H), 6.62 (t, J = 56.4 Hz, 1H), 4.94 (q, J = 6.4 Hz, 1H), 1.49 (d, J = 6.4 Hz, 3H).
Synthesis of 1-(1-(difluoromethyl)-1H-pyrazol-3-yl)ethan-1-ol (1A-6-7)
Step 1. Synthesis of methyl 1-(difluoromethyl)-1H-pyrazole-3-carboxylate To a solution of methyl 1H-pyrazole-3-carboxylate (9 g, 0.07 mol, 1 eq) in DMA (100 mL) was added sodium 2-chloro-2,2-difluoroacetate (13.5 g, 0.09 mmol, 1.3 eq) and Cs2CO3 (49 g, 0.14 mol, 2 eq) at 25 °C. After stirring for 16 h at 100 °C the reaction mixture was allowed to cool to room temperature. The resulting mixture was diluted with H2O (100 mL) and extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine (3 x 100 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography, eluting with (PE : EtOAc = 2 : 1) to afford the title compound (3.8 g, 30% yield) as a white solid. LCMS: m/z = 177.25 [M+1]+ Step 2. Synthesis of (1-(difluoromethyl)-1H-pyrazol-3-yl)methanol To a solution of methyl 1-(difluoromethyl)-1H-pyrazole-3-carboxylate (3.8 g, 0.02 mol, 1 eq) in anhydrous THF (50 mL) was added dropwise DIBAL-H (50.3 mL, 50.3 mol, 2.5 eq, 1M) at 0 °C and stirred for 2 h under nitrogen. The mixture was cooled to 0 °C, THF (100 mL) was added to the reaction mixture, followed by H2O (2.01 mL). After stirring for 15 minutes, 15% NaOH (aq) (2 mL) was added, followed by H2O (5.21 mL). It was again stirred at rt for 15 min. The combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure to afford the title compound (2.9 g, 97% yield) as yellow oil. LCMS: m/z = 149.15 [M+1]+ . Step 3. Synthesis of 1-(difluoromethyl)-1H-pyrazole-3-carbaldehyde A mixture of (1-(difluoromethyl)-1H-pyrazol-3-yl)methanol (1.3 g, 8.7 mmol, 1 eq) and MnO2 (7.6 g, 0.32 mol, 1.3 eq) in DCM (13 mL) was stirred at 25 °C for 16 h under nitrogen.
The resulting mixture was dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure to afford the title compound (1.2 g, 94% yield) as yellow oil. LCMS: m/z = 147.30 [M+1]+. Step 4. Synthesis of 1-(1-(difluoromethyl)-1H-pyrazol-3-yl)ethan-1-ol To a mixture of 1-(difluoromethyl)-1H-pyrazole-3-carbaldehyde (1.2 g, 0.0087 mol, 1 eq) in dry THF (30 mL) was added dropwise MeMgBr (6.3 mL, 0.019 mol, 2.2 eq, 3 M) at -78 °C and stirred for 4 h under nitrogen. The resulting mixture was diluted with NH4Cl (50 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (1 x 50 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography, eluting with (PE : EtOAc = 10 : 1) to afford the title compound (454 mg, 32% yield) as a white solid. LCMS: m/z = 163.20 [M+1]+. Synthesis of 1-(1-(difluoromethyl)-1H-pyrazol-4-yl)ethan-1-ol (1A-6-8)
Step 1. Synthesis of 1-(difluoromethyl)-1H-pyrazole-4-carbaldehyde To a solution of 1H-pyrazole-4-carbaldehyde (2.1 g, 0.022 mol, 1 eq) in MeCN (30 mL) was added diethyl (bromodifluoromethyl)phosphonate (10 g, 0.037 mol, 1.7 eq) and KF (3.4 g, 0.06 mol, 3 eq) at 25 °C and the reaction mixture was stirred for 16 h. The resulting mixture was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography, eluting with (PE:EtOAc = 100:1) to afford the title compound (1.4 g, 43% yield) as colorless oil. LCMS: m/z = 147.30 [M+1]+. Step 2. Synthesis of 1-(1-(difluoromethyl)-1H-pyrazol-4-yl)ethan-1-ol To a solution of 1-(difluoromethyl)-1H-pyrazole-4-carbaldehyde (633 mg, 4.3 mmol, 1 eq) in THF (10 mL) was added dropwise MeMgBr (2.8 mL, 8.6 mmol, 2 eq, 3 M) at -78 °C and stirred for 4 h at rt under nitrogen. The resulting mixture was diluted with NH4Cl (10 mL) and extracted with EtOAc (3 x 510 mL). The combined organic layers were washed with brine
(1 x 10 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography, eluting with PE/EtOAc (10/1) to afford the title compound (670 mg, 96% yield) as yellow oil. LCMS: m/z = 163.20 [M+1]+ . Synthesis of 2-(3-methylpyridin-4-yl)propan-2-ol (IA-6-9)
Step 1. Synthesis of methyl 3-methylisonicotinate Thionyl chloride (2.33 mL, 32 mmol) was added dropwise over 5 minutes to a stirred solution of 3-methyl-isonicotinic acid (4 g, 29 mmol) in MeOH (60 mL) at 0 °C under Argon atmosphere. The resulting solution was stirred and heated at reflux for 2 h. The mixture was allowed to cool to rt. The solvent was evaporated under reduced pressure. The mixture was dissolved in CH2Cl2 (100 mL) and washed with saturated NaHCO3 (aq.) (30 mL). The organic layer was dried (MgSO4) and evaporated under reduced pressure to give methyl 3-methyl- isonicotinate (4 g, 91%) as colorless oil. LC-MS: m/z = 152.1 [M+1]+. Step 2. Synthesis of 2-(3-methylpyridin-4-yl)propan-2-ol MeMgBr (66.15 mL in THF, 66 mmol) was added dropwise over 10 min to a stirred solution of methyl 3-methylisonicotinate (4.0 g, 26.46 mmol) in THF (60 mL) at -78 °C under argon atmosphere. The resulting solution was stirred at 0 °C for 2 h. The mixture was quenched with saturated NH4Cl (aq.) (50 mL) at 0 °C and extracted with EA (50 mL x 3). The combined organic layers were dried (MgSO4), concentrated and purified by flash chromatography with PE/EA=3/1-1/1 to give the title product, 2-(3-methylpyridin-4-yl)propan-2-ol (2.1 g, 52.5%) as a white solid. LC-MS: m/z = 152.1 [M+1]+. EXAMPLE 1: (R)-N-(5-fluoroquinolin-6-yl)-7-(1-methyl-1H-pyrazol-4-yl)-5-(1-(pyrimidin- 2-yl)ethoxy)quinazolin-4-amine (Compound 1).
Step 1. Synthesis of (E)-N'-(5-bromo-2-cyano-3-fluorophenyl)-N,N-dimethylformimidamide. A mixture of 2-amino-4-bromo-6-fluorobenzonitrile (2 g, 9.3 mmol, 1 eq) in DMF- DMA (20 mL) was stirred at 120 °C for 2 h under nitrogen. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was triturated with ethyl ether (20 mL), filtered and dried to afford the title compound (2.2 g, 88% yield) as a white solid. LC-MS: m/z =270.15/272.15 [M+1]+. Step 2. Synthesis of 7-bromo-5-fluoro-N-(5-fluoroquinolin-6-yl)quinazolin-4-amine. A mixture of (E)-N'-(5-bromo-2-cyano-3-fluorophenyl)-N,N-dimethylformimidamide (1 g, 3.7 mmol, 1 eq) and 5-fluoroquinolin-6-amine (660 mg, 4.1 mmol, 1.1 eq) in AcOH (12 mL) was stirred at 100 °C for 16 h under nitrogen. The mixture was poured into water (30 mL). The precipitated solid was filtered, washed with water, dried under reduced pressure to afford the title compound (965 mg, 67% yield) as a brown solid. LC-MS: m/z = 387.20/389.20 [M+1]+. Following steps 1 and 2 above, intermediates 1A-12-2, 1A-12-3, 1A-12-4, 1A-12-5, and 1A-12-6 were synthesized.
Step 3. Synthesis of (R)-7-bromo-N-(5-fluoroquinolin-6-yl)-5-(1-(pyrimidin-2-yl)ethoxy)- quinazolin-4-amine To a solution of (R)-1-(pyrimidin-2-yl)ethanol (48 mg, 0.39 mmol, 3 eq) in anhydrous THF (2 mL) was added NaH (31 mg, 0.77 mmol, 6 eq, 60%) at 0 °C and stirred for 0.5 h under nitrogen. A solution of 5-fluoro-N-(5-fluoroquinolin-6-yl)quinazolin-4-amine (50 mg, 0.13 mmol, 1 eq) in THF (1 mL) was added and then stirred at 80 °C for 4 h. The mixture was cooled to room temperature and poured into water (10 mL). The precipitated solid was filtered, washed with water, and dried under reduced pressure to afford the title compound (61 mg, 96% yield) as a brown solid. LC-MS: m/z = 491.35/493.35 [M+1]+. Following steps 1-3 described above, intermediates 1A-13-2, 1A-13-3, 1A-13-4, 1A- 13-5, 1A-13-6, 1A-13-7, 1A-13-8 and 1A-13-9 below were synthesized.
Step 4. Synthesis of (R)-N-(5-fluoroquinolin-6-yl)-7-(1-methyl-1H-pyrazol-4-yl)-5-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 1). A mixture of (R)-7-bromo-N-(5-fluoroquinolin-6-yl)-5-(1-(pyrimidin-2-yl)ethoxy)- quinazolin-4-amine (61 mg, 0.12 mmol, 1 eq), (1-methyl-1H-pyrazol-4-yl)boronic acid (20 mg, 0.16 mmol, 1.3 eq), K2CO3 (51 mg, 0.372 mmol, 3 eq), and Pd(dppf)Cl2 (9.9 mg, 0.012 mmol, 0.1 eq) in dioxane (3 mL) and H2O (0.4 mL) was degassed and purged with nitrogen 3 times. The mixture was stirred at 80 °C for 16 h under nitrogen, diluted with H2O (20 mL), and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The resulting residue was purified by Prep-TLC (DCM:MeOH = 15:1) to afford the title compound (18 mg, 30% yield) as a white solid. LC-MS: m/z = 493.25 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 11.06 (s, 1H), 8.92 (dd, J = 4.1, 1.4 Hz, 1H), 8.75 (d, J = 4.9 Hz, 2H), 8.54 (t, J = 8.8 Hz, 1H), 8.48 (d, J = 8.2 Hz, 1H), 8.46 (s, 1H), 8.40 (s, 1H),
8.07 (s, 1H), 7.94 (d, J = 9.2 Hz, 1H), 7.61 (dd, J = 8.5, 4.2 Hz, 1H), 7.54 (s, 1H), 7.47 (s, 1H), 7.42 (t, J = 4.9 Hz, 1H), 6.23 (q, J = 6.0 Hz, 1H), 3.87 (s, 3H), 1.82 (d, J = 6.3 Hz, 3H). EXAMPLES 2-11 Following the procedure described in Example 1, Compounds 2-11 were synthesized. Compound 2: (R)-N-(3-chloro-2-fluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-5-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 49%. LC-MS: m/z = 476.10 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.70– 8.68 (m, 3H), 8.36 (s, 1H), 8.10 (s, 1H), 7.86 – 7.79 (m, 1H), 7.68 (d, J = 0.9 Hz, 1H), 7.60 – 7.53 (m, 1H), 7.49 (d, J = 1.2 Hz, 1H), 7.41 (t, J = 4.9 Hz, 1H), 7.35 (td, J = 8.2, 1.5 Hz, 1H), 6.23 (q, J = 6.4 Hz, 1H), 3.98 (s, 3H), 1.91 (d, J = 6.4 Hz, 3H). Compound 3: (S)-N-(3-chloro-2,4-difluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-5- (1-(pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 23%. LC-MS: m/z = 494.15 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.74 – 8.66 (m, 3H), 8.39 (s, 1H), 8.12 (s, 1H), 7.82- 7.78 (m, 1H), 7.70 (s, 1H), 7.49 (d, J = 1.0 Hz, 1H), 7.42 (t, J = 5.0 Hz, 1H), 7.34 (td, J = 9.0, 2.0 Hz, 1H), 6.23 (q, J = 6.5 Hz, 1H), 3.99 (s, 3H), 1.91 (d, J = 6.4 Hz, 3H). Compound 4: (R)-N-(3-chloro-2,4-difluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-5- [1-(pyrimidin-2-yl)ethoxy]quinazolin-4-amine. Yield 34%. LC-MS: m/z = 494.12 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.73 – 8.66 (m, 3H), 8.39 (s, 1H), 8.12 (s, 1H), 7.84- 7.78 (m, 1H), 7.70 (s, 1H), 7.49 (d, J = 1.1 Hz, 1H), 7.42 (t, J = 4.9 Hz, 1H), 7.34 (td, J = 9.0, 1.9 Hz, 1H), 6.23 (q, J = 6.3 Hz, 1H), 3.98 (s, 3H), 1.91 (d, J = 6.4 Hz, 3H). Compound 5: N-(3-chloro-2,4-difluorophenyl)-5-(cyclopropyl(pyrimidin-2-yl)meth- oxy)-7-(1-methyl-1H-pyrazol-4-yl)quinazolin-4-amine. Yield 39%. LC-MS: m/z = 520.20 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 10.82 (s, 1H), 8.76 (d, J = 4.8 Hz, 2H), 8.45 (s, 1H), 8.33 (s, 1H), 8.27-8.24 (m, 1H), 8.00 (s, 1H), 7.51 (d, J = 1.6 Hz, 1H), 7.44-7.38 (m, 3H), 5.61 (d, J = 8.4 Hz, 1H), 3.86 (s, 3H), 1.65-1.53 (m, 1H), 0.68-0.65 (m, 2H), 0.62-0.51 (m, 2H). Compound 6: (S)-N-(3-chloro-4-fluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-5-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 28.5%. LC-MS: m/z = 476.20 [M+1]+.
1H NMR (400 MHz, DMSO-d6) δ 11.07 (s, 1H), 8.87 (d, J = 4.9 Hz, 2H), 8.48 (d, J = 9.2 Hz, 2H), 8.32 – 8.26 (m, 1H), 8.14 (s, 1H), 7.86 – 7.79 (m, 1H), 7.57 – 7.46 (m, 4H), 6.26 (q, J = 5.6 Hz, 1H), 3.87 (s, 3H), 1.75 (d, J = 6.2 Hz, 3H). Compound 7: (S)-N-(3-bromo-2,4-difluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-5- (1-(pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 30%. LC-MS: m/z = 538.20/540.20 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 10.90 (s, 1H), 8.74 (d, J = 4.8 Hz, 2H), 8.42 (d, J = 6.4 Hz, 2H), 8.16-8.09 (m, 1H), 8.08 (s, 1H), 7.53 (s, 1H), 7.49-7.43 (m, 2H), 7.35 (t, J = 8.4 Hz, 1H), 6.20 (q, J = 6.8 Hz, 1H), 3.87 (s, 3H), 1.78 (d, J = 6.4 Hz, 3H). Compound 8: (S)-N-(3-bromo-4-fluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-5-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 28%. LC-MS: m/z = 520.20/522.20 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.82 (d, J = 4.9 Hz, 2H), 8.42 (s, 1H), 8.29 (dd, J = 6.3, 2.5 Hz, 1H), 8.18 (d, J = 0.6 Hz, 1H), 7.99 (s, 1H), 7.91 – 7.85 (m, 1H), 7.47 – 7.43 (m, 2H), 7.36 (s, 1H), 7.28 (t, J = 9.1 Hz, 1H), 6.08 (q, J = 6.9 Hz, 1H), 3.95 (s, 3H), 1.85 (d, J = 6.3 Hz, 3H). Compound 9: (S)-N-(3-chloro-2,4-difluorophenyl)-7-(1H-pyrazol-4-yl)-5-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 23.6%. LC-MS: m/z = 480.35 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 8.75 (d, J = 4.8 Hz, 2H), 8.41 (s, 1H), 8.26 (s, 2H), 8.08 (dd, J = 14.0, 8.8 Hz, 1H), 7.56 (s, 1H), 7.47 (s, 1H), 7.44 (t, J = 4.8 Hz, 1H), 7.38 (t, J = 8.8 Hz, 1H), 6.19 (q, J = 6.4 Hz, 1H), 1.77 (d, J = 6.4 Hz, 3H). Compound 10: (S)-N-(3-chloro-2,4-difluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)- 5-(1-(pyrimidin-4-yl)ethoxy)quinazolin-4-amine. Yield 41 %. LC-MS: m/z = 494.20 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 11.65 (s, 1H), 8.99 (d, J = 1.0 Hz, 1H), 8.86 (d, J = 5.2 Hz, 1H), 8.84 (s, 1H), 8.49 (s, 1H), 8.08 (s, 1H), 7.87 (td, J = 8.7, 5.8 Hz, 1H), 7.73 (dd, J = 5.2, 1.1 Hz, 1H), 7.68 (s, 1H), 7.63 (s, 1H), 7.51 (td, J = 9.0, 1.7 Hz, 1H), 6.40 (q, J = 6.4 Hz, 1H), 3.90 (s, 3H), 1.77 (d, J = 6.4 Hz, 3H). Compound 11: (S)-N-(3-chloro-2,4-difluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)- 5-(1-(pyridin-2-yl)ethoxy)quinazolin-4-amine. Yield 23 %. LC-MS: m/z = 493.15 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.73 (s, 1H), 8.59 (d, J = 5.1 Hz, 1H), 8.36 (s, 1H),
8.27 (t, J = 7.8 Hz, 1H), 8.06 (s, 1H), 8.05 – 7.96 (m, 2H), 7.72 – 7.66 (m, 1H), 7.59 (s, 1H), 7.51 (s, 1H), 7.31 (t, J = 8.2 Hz, 1H), 6.41 (q, J = 6.1 Hz, 1H), 3.97 (s, 3H), 1.97 (d, J = 6.4 Hz, 3H). EXAMPLE 12: (R)-N-(4-fluoropyrazolo[1,5-a] pyridin-5-yl)-7-(1-methyl-1H-pyrazol-4-yl)- 5-(1-(pyrimidin-2-yl) ethoxy) quinazolin-4-amine (Compound 12).
Step 1. Synthesis of 7-bromo-5-fluoroquinazolin-4(3H)-one A solution of 2-amino-4-bromo-6-fluorobenzonitrile (2 g, 9.3 mmol, 1 eq) in HCOOH (30 mL) and H2SO4 (0.3 mL) was stirred at 100 °C for 3 h. The reaction mixture was concentrated under reduced pressure, the crude was diluted with cold water (30 mL) and stirred for 20 min at room temperature. The residue was triturated with water (5 mL) at room temperature and filtered to afford 7-bromo-5-fluoroquinazolin-4(3H)-one (370 mg, 90 % yield) as a white solid. LC-MS: m/z = 243.10/245.10 [M+1]+. Step 2. Synthesis of 7-bromo-4-chloro-5-fluoroquinazoline To a solution of 7-bromo-5-fluoroquinazolin-4(3H)-one (500 mg, 2 mmol, 1 eq) in CHCl3 (30 mL) was added (COCl)2 (1 mL) and DMF (5 drop). The resulting mixture was stirred at 70 °C for 2 h under nitrogen. After completion, the reaction mixture was added to ice cold saturated NaHCO3 solution (10 mL), extracted with DCM/MeOH (10/1) (50 mL x 2). The combined organic layers were dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude residue was purified by silica gel column chromatography (PE/EA (5/1)) to afford 7-bromo-4-chloro-5-fluoroquinazoline (470 mg, 87.5% yield) as a white solid. LC-MS: m/z = 260.95/262.95 [M+1]+.
Step 3. Synthesis of 7-bromo-5-fluoro-N-(4-fluoropyrazolo[1,5-a]pyridin-5-yl)quinazolin-4- amine A mixture of 4-fluoropyrazolo[1,5-a]pyridin-5-amine (75.1 mg, 0.497 mmol, 1 eq.) and 7-bromo-4-chloro-5-fluoroquinazoline (130 mg, 0.497 mmol, 1 eq.) in isopropyl alcohol (5 ml) was stirred at 90°C for 1 h. The reaction mixture was concentrated under reduced pressure. The residue was purified by FCC (10% methanol in DCM) to afford 7-bromo-5-fluoro-N-{4- fluoro-pyrazolo[1,5-a]pyridin-5-yl}quinazolin-4-amine (110 mg, 0.292 mmol, 58.8%) as a yellow solid. LC-MS: m/z = 375.95/377.95 [M+1]+. Following the procedure described above, the intermediates 1A-5-2 and 1A-5-3 were prepared.
Step 4. Synthesis of (R)-7-bromo-N-(4-fluoropyrazolo[1,5-a]pyridin-5-yl)-5-(1-(pyrimidin-2- yl)ethoxy)quinazolin-4-amine To a solution of (1R)-1-(pyrimidin-2-yl)ethan-1-ol (145 mg, 1.17 mmol, 4 eq.) in THF (10 ml) at 0 °C was added sodium hydride (28 mg, 1.17 mmol, 4 eq.) and stirred at 0 °C for 20 minutes, followed by addition of 7-bromo-5-fluoro-N-(4-fluoropyrazolo[1,5-a]pyridin-5- yl)quinazolin-4-amine (110 mg, 0.292 mmol, 4 eq.). After stirring for 16 h at 80 °C, the reaction mixture was cooled to rt and concentrated under reduced pressure. The crude residue was purified by FCC (8% methanol/DCM) to afford 7-bromo-N-{4-fluoropyrazolo[1,5- a]pyridin-5-yl}-5-[(1R)-1-(pyrimidin-2-yl)ethoxy]quinazolin-4-amine (100 mg, 0.2 mmol, 71%) as a yellow solid. LC-MS: m/z = 480.10/482.10 [M+1]+.
Following the procedure described above, intermediates 1A-7-2 and 1A-7-3 were synthesized.
Step 5. Synthesis of (R)-N-(4-fluoropyrazolo[1,5-a]pyridin-5-yl)-7-(1-methyl-1H-pyrazol-4- yl)-5-(1-(pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 12) A mixture of (1-methyl-1H-pyrazol-4-yl)boronic acid (34 mg, 0.271 mmol, 1.3 equiv.), 7-bromo-N-{4-fluoropyrazolo[1,5-a]pyridin-5-yl}-5-[(1R)-1-(pyrimidin-2-yl)ethoxy]quina- zolin-4-amine (100 mg, 0.2 mmol, 1 eq.) and potassium carbonate (86 mg, 0.62 mmol, 3 eq.) in 1,4-dioxane (5 ml) and H2O (0.5 ml) was degassed with N2 three times. [1,1'-Bis(diphenyl- phosphino)ferrocene]palladium(II) chloride (15.3 mg, 0.021 mmol, 0.1 equiv.) was added. The resulting mixture was stirred at 100 °C for 5 h, warmed to rt, filtered, and concentrated under vacuum to yield crude product. The crude was purified by Prep-HPLC to afford N-{4-fluoro- pyrazolo[1,5-a]pyridin-5-yl}-7-(1-methyl-1H-pyrazol-4-yl)-5-[(1R)-1-(pyrimidin-2-yl)- ethoxy]quinazolin-4-amine HCl salt (44.9 mg, 0.093 mmol, 44.8%) as a yellow solid. LC-MS: m/z = 482.35 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.76 (s, 1H), 8.75 (s, 2H), 8.60 (d, J = 7.2 Hz, 1H), 8.46 (s, 1H), 8.21 (s, 1H), 8.10 (d, J = 2.0 Hz, 1H), 7.75 (d, J = 1.6 Hz, 1H), 7.54 (d, J = 1.6 Hz, 1H), 7.43 (t, J = 4.8 Hz, 1H), 7.38 (t, J = 7.2 Hz, 1H), 6.88 (d, J = 1.6 Hz, 1H), 6.29 (q, J = 6.4 Hz, 1H), 4.02 (s, 3H), 1.95 (d, J = 6.4 Hz, 3H). EXAMPLES 13 and 14 Following the procedure described in Example 12 above, Compounds 13 and 14 were synthesized. Compound 13: N-(5-Fluoroquinolin-6-yl)-7-(1-methyl-1H-pyrazol-4-yl)-5-(1-
(pyrimidin-4-yl)ethoxy)quinazolin-4-amine. Yield 25%. LC-MS: m/z = 493.10 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 10.64 (s, 1H), 9.11 (s, 1H), 8.90 (d, J = 2.9 Hz, 1H), 8.85-8.80 (m, 2H), 8.52 (s, 1H), 8.46 (d, J = 8.1 Hz, 1H), 8.32 (s, 1H), 8.01 (s, 1H), 7.95 (d, J = 9.3 Hz, 1H), 7.73 (d, J = 4.7 Hz, 1H), 7.60 (dd, J = 8.4, 4.2 Hz, 1H), 7.55 (s, 1H), 7.31 (s, 1H), 6.21 (q, J = 6.9 Hz, 1H), 3.86 (s, 3H), 1.84 (d, J = 6.3 Hz, 3H). Compound 14: (R)-7-Fluoro-N-(7-(1-methyl-1H-pyrazol-4-yl)-5-(1-(pyrimidin-2-yl)- ethoxy)quinazolin-4-yl)benzo[d]isothiazol-6-amine. Yield 22.7%. LC-MS: m/z = 499.15 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 9.10 (d, J = 4.2 Hz, 1H), 8.70-8.68 (m, 3H), 8.40 (s, 1H), 8.18 (d, J = 8.5 Hz, 1H), 8.14 (s, 1H), 8.00-7.96 (m, 1H), 7.73 (s, 1H), 7.51 (s, 1H), 7.39 (t, J = 4.9 Hz, 1H), 6.29-6.24 (m, 1H), 3.99 (s, 3H), 1.93 (d, J = 6.4 Hz, 3H). EXAMPLE 15: Synthesis of N-(3-chloro-2,4-difluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)- 5-((2-(pyrimidin-2-yl)propan-2-yl)oxy)quinazolin-4-amine (Compound 15).
Step 1. Synthesis of 7-bromo-N-(3-chloro-2,4-difluorophenyl)-5-((2-(pyrimidin-2-yl)- propan-2-yl)oxy)quinazolin-4-amine To a solution of 2-(pyrimidin-2-yl)propan-2-ol (204.5 mg, 1.48 mmol, 3 eq) in dry THF (8 mL) was added NaH (118.4 mg, 2.96 mmol, 6.0 eq, 60% wt) at 0 °C and stirred for 0.5 h under nitrogen atmosphere. A solution of 7-bromo-N-(3-chloro-2,4-difluorophenyl)-5-fluoro- quinazolin-4-amine (190 mg, 0.494 mmol, 1.0 eq) in dry THF (2 mL) was added and stirred at 80 °C for 24 h. The mixture was cooled to room temperature and diluted with water (5 mL), extracted with EA (50 mL x 3) and washed with brine (5 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography, eluting with (EA: PE = 30:70) to afford the title compound (100 mg, 40.5% yield) as a yellow solid. LC-MS: m/z = 506.15/508.15 [M+1]+. Following the above method, intermediate 1A-13-11 was synthesized.
Step 2. Synthesis of N-(3-chloro-2,4-difluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-5-((2- (pyrimidin-2-yl)propan-2-yl)oxy)quinazolin-4-amine (Compound 15) A mixture of 7-bromo-N-(3-chloro-2,4-difluorophenyl)-5-((2-(pyrimidin-2-yl)propan- 2-yl)oxy)quinazolin-4-amine (80 mg, 0.158 mmol, 1 eq), (1-methyl-1H-pyrazol-4-yl)boronic acid (60 mg, 0.475 mmol, 3 eq) and K2CO3 (65.6 mg, 0.475 mmol, 3 eq) in mixture of dioxane (6.4 mL) and H2O (0.8 mL) was degassed and purged with nitrogen 3 times. Pd(dppf)Cl2 (26 mg, 0.032 mmol, 0.2 eq) was added and purged with nitrogen 3 times, then the mixture was stirred at 80 °C for 16 h under nitrogen atmosphere. The resulting mixture was diluted with H2O (10 mL), cooled to rt, extracted with EtOAc (3 x 20 mL) and washed with brine (10 mL). The combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The resulting residue was purified by Prep-HPLC (0.04% NH4HCO3 in H2O/CH3CN) to afford the title compound (30.61 mg, 38% yield) as a white solid. LC-MS: m/z = 508.30 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 10.98 (s, 1H), 8.77 (d, J = 4.8 Hz, 2H), 8.42 (s, 1H), 8.21 (s, 1H), 8.12-8.06 (m, 1H), 7.80 (s, 1H), 7.53 (d, J = 1.6 Hz, 1H), 7.47-7.44 (m, 1H), 7.41- 7.36 (m, 1H), 6.88 (d, J = 1.6 Hz, 1H), 3.84 (s, 3H), 2.01 (s, 6H). EXAMPLE 16 Following the procedure described in Example 15, Compound 16 was prepared. Compound 16: N-(3-chloro-4-fluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-5-((2-(pyrimidin- 2-yl)propan-2-yl)oxy)quinazolin-4-amine. Yield 66.8%. LC-MS: m/z = 490.20 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.68 (d, J = 4.8 Hz, 2H), 8.62 (s, 1H), 8.28 (s, 1H), 7.97 (s, 1H), 7.90 (dd, J = 6.8, 2.4 Hz, 1H), 7.60-7.56 (m, 1H), 7.45-7.42 (m, 3H), 7.41-7.39 (m, 1H), 3.97 (s, 3H), 2.17 (s, 6H). EXAMPLE 17 Compound 17: Synthesis of (S)-N-(3-chloro-2,4-difluorophenyl)-7-(1H-pyrazol-1-yl)-5-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine.
A mixture of (S)-7-bromo-N-(3-chloro-2,4-difluorophenyl)-5-(1-(pyrimidin-2-yl)eth- oxy)quinazolin-4-amine (1A-13-7, 150 mg, 0.31 mmol, 1 eq), 1H-pyrazole (83 mg, 1.22 mmol, 4 eq), Cs2CO3 (299.5 mg, 0.92 mmol, 3 eq) and Ephos-Pd-G4 (30 mg, 0.1 mmol, 0.1 eq) in dioxane (5 mL) was degassed under vacuum and purged with nitrogen 3 times and then the mixture was stirred at 100 °C for 24 h under nitrogen. The resulting mixture was diluted with H2O (10 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The crude product was purified by Prep-TLC (DCM:MeOH = 25:1) to afford the title compound (21 mg, 14.4% yield) as a white solid. LC-MS: m/z = 480.20 [M+1]+. 1H NMR (400 MHz, CDCl3) δ11.00 (s, 1H), 8.67 (d, J = 4.8 Hz, 2H), 8.59 (s, 1H), 8.20- 8.14 (m, 1H), 8.10 (d, J = 2.4 Hz, 1H), 7.76 (s, 1H), 7.72 (s, 1H), 7.68 (s, 1H), 7.26-7.24 (m, 1H), 7.08 (t, J = 8.4 Hz, 1H), 6.52 (s, 1H), 6.04-5.95 (m, 1H), 1.96 (d, J = 6.4 Hz, 3H). EXAMPLES 18-22 Following the procedure described in Example 17, Compounds 18-22 were synthesized. Compound 18: (S)-N-(3-chloro-2,4-difluorophenyl)-7-(4-methyl-1H-pyrazol-1-yl)-5- (1-(pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 9.0 %. LC-MS: m/z = 494.25 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 10.88 (s, 1H), 8.74 (d, J = 4.8 Hz, 2H), 8.55 (s, 1H), 8.46 (s, 1H), 8.11-8.03 (m, 1H), 7.68 (s, 2H), 7.64 (s, 1H), 7.44 (t, J = 4.8 Hz, 1H), 7.40 (t, J = 8.8 Hz, 1H), 6.15 (q, J = 6.4 Hz, 1H), 2.08 (s, 3H), 1.79 (d, J = 6.4 Hz, 3H). Compound 19: (S)-N-(3-chloro-4-fluorophenyl)-7-(1H-pyrazol-1-yl)-5-(1-(pyrimidin- 2-yl)ethoxy)quinazolin-4-amine. Yield 19%. LC-MS: m/z = 462.20 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.77 (d, J = 5.0 Hz, 2H), 8.72 (s, 1H), 8.64 (d, J = 2.8 Hz, 1H), 8.06 (dd, J = 6.6, 2.6 Hz, 1H), 7.97 (d, J = 1.9 Hz, 1H), 7.88 (d, J = 1.7 Hz, 1H), 7.80 (d, J = 1.9 Hz, 1H), 7.78 – 7.74 (m, 1H), 7.48 – 7.42 (m, 2H), 6.68 (dd, J = 2.7, 1.7 Hz, 1H), 6.26 (q, J = 6.4 Hz, 1H), 1.92 (d, J = 6.5 Hz, 3H). Compound 20: N-(3-chloro-4-fluorophenyl)-7-(1H-pyrazol-1-yl)-5-((2-(pyrimidin-2- yl)propan-2-yl)oxy)quinazolin-4-amine. Yield 60.9%. LC-MS: m/z = 476.15 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.67-8.66 (m, 3H), 8.47 (d, J = 2.4 Hz, 1H), 7.91-7.89 (m, 2H), 7.86 (s, 1H), 7.68 (d, J = 2.0 Hz, 1H), 7.61-7.57 (m, 1H), 7.45 (d, J = 8.8 Hz, 1H),
7.41 (t, J = 4.8 Hz, 1H), 6.66 (s, 1H), 2.19 (s, 6H). Compound 21: N-(3-chloro-2,4-difluorophenyl)-7-(1H-pyrazol-1-yl)-5-((2- (pyrimidin-2-yl)propan-2-yl)oxy)quinazolin-4-amine. Yield 18.5%. LC-MS: m/z = 494.15 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.67 (d, J = 4.8 Hz, 2H), 8.64 (s, 1H), 8.47 (d, J = 2.8 Hz, 1H), 7.95-7.76 (m, 3H), 7.70 (d, J = 2.0 Hz, 1H), 7.40 (t, J = 5.0 Hz, 1H), 7.37-7.29 (m, 1H), 6.65 (s, 1H), 2.18 (s, 6H). Compound 22: (S)-N-(3-chloro-4-fluorophenyl)-7-(4-fluoro-1H-pyrazol-1-yl)-5-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 12%. LC-MS: m/z = 480.10 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 12.01 (s, 1H), 9.16 (d, J = 4.4 Hz, 1H), 8.84 (s, 1H), 8.81 (d, J = 4.9 Hz, 2H), 8.12 (dd, J = 6.8, 2.5 Hz, 1H), 8.07 (d, J = 4.0 Hz, 1H), 7.88 (s, 2H), 7.78-7.73 (m, 1H), 7.60 (t, J = 8.9 Hz, 1H), 7.56 (t, J = 4.9 Hz, 1H), 6.33 (q, J = 6.4 Hz, 1H), 1.80 (d, J = 6.4 Hz, 3H). EXAMPLE 23: Synthesis of (S)-N-(3-chloro-4-fluorophenyl)-5-(1-(pyrimidin-2-yl)ethoxy)- quinazolin-4-amine (Compound 23). To a mixture of 7-bromo-N-(3-chloro-4-fluorophenyl)-5-[(1S)-1-(pyrimidin-2-yl)- ethoxy]quinazolin-4-amine (200 mg, 0.42 mmol, 1 eq), triethylsilane (147 mg, 1.26 mmol, 3 eq) and CsF (128 mg, 0.84 mmol, 2 eq) in 1,4-dioxane (5 mL) was added Pd(dppf)Cl2 (16 mg, 0.02 mmol, 0.05 eq.). After degassed and purged with nitrogen 3 times, the mixture was stirred at 50 °C for 16 h under nitrogen, then diluted with H2O (10 mL), cooled to rt, and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The crude product was purified by Prep-TLC (DCM:MeOH = 15:1) to afford the title compound (91.7 mg, 54.6% yield) as a white solid. LC-MS: m/z = 396.15 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.75 (d, J = 5.2 Hz, 2H), 8.71 (s, 1H), 8.07 (dd, J = 6.8, 2.4 Hz, 1H), 8.01 (t, J = 8.4 Hz, 1H), 7.80 – 7.75 (m, 1H), 7.55 (d, J = 8.4 Hz, 1H), 7.47 – 7.42 (m, 2H), 7.42 – 7.38 (m, 1H), 6.11 (q, J = 6.8 Hz, 1H), 1.88 (d, J = 6.4 Hz, 3H). EXAMPLE 24 Following the procedure described in Example 23, Compound 24 was synthesized. Compound 24: N-(3-chloro-4-fluorophenyl)-5-((2-(pyrimidin-2-yl)propan-2-yl)oxy)- quinazolin-4-amine. Yield 67%. LC-MS: m/z = 410.15 [M+1]+.
1H NMR (400 MHz, CD3OD) δ 8.66-8.63 (m, 3H), 7.93 (t, J = 8.4 Hz, 1H), 7.90 (dd, J = 6.4, 2.4 Hz, 1H), 7.61-7.56 (m, 1H), 7.49 (d, J = 8.4 Hz, 1H), 7.44 (t, J = 8.8 Hz, 1H), 7.40 (t, J = 4.8 Hz, 1H), 7.37 (d, J = 8.4 Hz, 1H), 2.12 (s, 6H). EXAMPLE 25: N-(3-chloro-2-fluorophenyl)-6-(1-(1-(difluoromethyl)-1H-pyrazol-3- yl)ethoxy)-7-methoxyquinazolin-4-amine (Compound 25).
To a mixture of 4-((3-chloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-ol (105 mg, 0.32 mmol, 1 eq) in toluene (2 mL) was added 1-(1-(difluoromethyl)-1H-pyrazol-3- yl)ethan-1-ol (160 mg, 0.96 mmol, 3 eq) and PPh3 (368 mg, 1.4 mmol, 4.4 eq). A solution of DIAD (239 mg, 1.2 mmol, 3.7 eq) in THF was added at 0 °C under N2. The resulting mixture was stirred at 100 °C for 16 h. After cooling to rt, it was diluted with H2O (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (1 x 10 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The resulting residue was purified by Prep-HPLC to afford the title compound (28.7 mg, 18% yield) as a white solid. LC-MS: m/z = 464.10 [M+1]+. 1H NMR (400 MHz, CD3OD): δ 8.65 (s, 1H), 8.15 (s, 1H), 8.03 (s, 1H), 7.80 (s, 1H), 7.54-7.42 (m, 2H), 7.30-7.26 (m, 2H), 6.59 (d, J = 2.6 Hz, 1H), 5.83 (q, J = 6.2 Hz, 1H), 4.08 (s, 3H), 1.76 (d, J = 6.4 Hz, 3H). EXAMPLE 26 Following the procedure described in Example 25, Compound 26 was prepared. Compound 26: N-(3-chloro-2-fluorophenyl)-6-(1-(1-(difluoromethyl)-1H-pyrazol-4- yl)ethoxy)-7-methoxyquinazolin-4-amine. Yield 7%. LC-MS: m/z = 464.15 [M+1]+. 1H NMR (400 MHz, CD3OD): δ 8.65 (s, 1H), 8.15 (s, 1H), 8.03 (s, 1H), 7.80 (s, 1H), 7.59 – 7.42 (m, 3H), 7.31 – 7.27 (m, 1H), 7.26 (s, 1H), 5.82 (q, J = 6.3 Hz, 1H), 4.08 (s, 3H), 1.76 (d, J = 6.4 Hz, 3H). EXAMPLE 27: (S)-N-(3-chloro-2-fluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-6-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 27).
Step 1. Synthesis of (S)-methyl 4-bromo-2-nitro-5-(1-(pyrimidin-2-yl)ethoxy)benzoate To a solution of (S)-1-(pyrimidin-2-yl)ethanol (1 g, 8 mmol, 1.5 eq) in DMF (20 mL) was added NaH (322 mg, 8 mmol, 1.5 eq, 60%) at 0 °C and stirred for 0.5 h under nitrogen. A solution of methyl 4-bromo-5-fluoro-2-nitrobenzoate (1.49 g, 5.37 mmol, 1 eq) in DMF (15 mL) was added and stirred at room temperature for 1 h. The mixture was quenched with sat NH4Cl (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography, eluting with (PE : EA = 5 : 1) to afford the title compound (1.19 g, 58% yield) as a yellow solid. LC-MS: m/z = 382.25 and 384.25 [M+1]+. Following the above procedure, intermediates 1B-2-2, 1B-2-3 and 1B-2-4 were prepared.
Step 2. Synthesis of (S)-methyl 2-amino-4-bromo-5-(1-(pyrimidin-2-yl)ethoxy)benzoate A mixture of (S)-methyl 4-bromo-2-nitro-5-(1-(pyrimidin-2-yl)ethoxy)benzoate (185 mg, 0.48 mmol, 1 eq), Fe (135 mg, 2.42 mmol, 5 eq) and NH4Cl (259 mg, 4.84 mmol, 10 eq) in EtOH (5 mL) and H2O (0.5 mL) was degassed and purged with nitrogen 3 times. It was stirred at 80 °C for 2 h under nitrogen, filtered and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The resulting residue was purified by Prep-TLC (PE:EA = 5:1) to afford the title compound (120 mg, 71% yield) as a white solid. LC-MS: m/z = 352.10/354.10 [M+1]+. Following the above procedure, intermediates 1B-3-2, 1B-3-3, and 1B-3-4 were prepared.
Step 3. Synthesis of (S)-7-bromo-6-(1-(pyrimidin-2-yl)ethoxy)quinazolin-4(3H)-one A mixture (S)-methyl 2-amino-4-bromo-5-(1-(pyrimidin-2-yl)ethoxy)benzoate (120 mg, 0.34 mmol, 1 eq), formimidamide acetate (106 mg, 1.02 mmol, 3 eq) and p-TSA (6.4 mg, 0.034 mmol, 0.1 eq) in EtOH (5 mL) was stirred at 100 °C for 12 h, concentrated, and purified by FCC (DCM : MeOH = 40 : 1) to afford the title compound (105 mg, 89 % yield) as a white solid. LC-MS: m/z = 347.20/349.20 [M+1] +. Following the above procedure, intermediates 1B-4-2, 1B-4-3, and 1B-4-4 were prepared.
Step 4. Synthesis of (S)-7-bromo-4-chloro-6-(1-(pyrimidin-2-yl)ethoxy)quinazoline. To a solution of (S)-7-bromo-6-(1-(pyrimidin-2-yl)ethoxy)quinazolin-4(3H)-one (105 mg, 0.3 mmol, 1 eq) in CHCl3 (50 mL) was added (COCl)2 (0.2 mL) and DMF (2 drops). The mixture was stirred at 70 °C for 2 h under nitrogen and then concentrated to afford a crude title compound (110 mg, 100% yield) as a yellow solid. LCMS: m/z 365.40/367.40 [M+1]+. Following the above procedure, intermediates 1B-5-2 1B-5-3, and 1B-5-4 were prepared.
Step 5. Synthesis of (S)-7-bromo-N-(3-chloro-2-fluorophenyl)-6-(1-(pyrimidin-2-yl)ethoxy) quinazolin-4-amine A mixture of (S)-7-bromo-4-chloro-6-(1-(pyrimidin-2-yl)ethoxy)quinazoline (110 mg, 0.3 mmol, 1 equiv.) and 3-chloro-2-fluoroaniline (132 mg, 0.9 mmol, 3 equiv.) in ACN (5 ml) was stirred at 70 °C for 1 h, subsequently quenched with saturated NaHCO3, and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by FCC (DCM:MeOH = 30:1) to afford the title compound (110 mg, 0.29 mmol, 76%) as a white solid. LC-MS: m/z = 474.55/476.55 [M+1]+. Following the above procedure, intermediates 1B-6-2, 1B-6-3, 1B-6-4, 1B-6-5 and 1B- 6-6 were prepared.
Step 6. Synthesis of (S)-N-(3-chloro-2-fluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-6-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 27) A mixture of (S)-7-bromo-N-(3-chloro-2-fluorophenyl)-6-(1-(pyrimidin-2-yl)ethoxy)- quinazolin-4-amine (110 mg, 0.23 mmol, 1 eq), (1-methyl-1H-pyrazol-4-yl)boronic acid (44 mg, 0.348 mmol, 1.5 eq), K2CO3 (96 mg, 0.696 mmol, 3 eq) and Pd(dppf)Cl2 (18 mg, 0.023 mmol, 0.1 eq) in dioxane (5 mL) and H2O (0.6 mL) was degassed and purged with nitrogen 3 times, then stirred at 80 °C for 4 h under nitrogen, diluted with H2O (20 mL), and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The resulting residue was purified by Prep-TLC (DCM:MeOH = 15:1) to
afford the title compound (61.2 mg, 56% yield) as a white solid. LC-MS: m/z = 476.15 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.82 (d, J = 4.9 Hz, 2H), 8.53 (s, 1H), 8.34 (s, 1H), 8.22 (s, 1H), 7.94 (s, 1H), 7.75 (s, 1H), 7.58 (t, J = 7.4 Hz, 1H), 7.43 – 7.34 (m, 2H), 7.20 (t, J = 8.1 Hz, 1H), 5.94 (q, J = 6.6 Hz, 1H), 3.97 (s, 3H), 1.86 (d, J = 6.4 Hz, 3H). EXAMPLES 28-35 Following the procedure described in Example 27, Compounds 28-35 of this invention were prepared. Compound 28: (R)-N-(3-chloro-2-fluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-6-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 59%. LC-MS: m/z = 476.20 [M+1]+. 1H NMR (400 MHz, CD3OD): δ 8.82 (d, J = 4.9 Hz, 2H), 8.53 (s, 1H), 8.33 (s, 1H), 8.22 (s, 1H), 7.93 (s, 1H), 7.74 (s, 1H), 7.58 (t, J = 7.1 Hz, 1H), 7.44 – 7.34 (m, 2H), 7.20 (t, J = 8.1 Hz, 1H), 5.94 (q, J = 6.4 Hz, 1H), 3.97 (s, 3H), 1.86 (d, J = 6.4 Hz, 3H). Compound 29: (R)-N-(3-chloro-2-fluorophenyl)-7-(1-(difluoromethyl)-1H-pyrazol-4- yl)-6-(1-(pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 57%. LC-MS: m/z = 512.00 [M+1]+. 1H NMR (400 MHz, dmso-d6): δ 9.79 (s, 1H), 9.35 (s, 1H), 8.86 (d, J = 4.9 Hz, 2H), 8.66 (s, 1H), 8.40 (s, 1H), 8.22 (s, 1H), 8.05 (s, 1H), 7.91 (t, J = 59.1 Hz, 1H), 7.55 – 7.44 (m, 3H), 7.26 (t, J = 8.2 Hz, 1H), 6.06 (q, J = 6.5 Hz, 1H), 1.76 (d, J = 6.3 Hz, 3H). Compound 30: N-(4-fluoropyrazolo[1,5-a]pyridin-5-yl)-7-(1-methyl-1H-pyrazol-4- yl)-6-(1-(pyrimidin-4-yl)ethoxy)quinazolin-4-amine. Yield 29%. LC-MS: m/z = 482.25 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 9.36 (s, 1H), 8.90 (s, 1H), 8.74 (s, 1H), 8.56 (s, 1H), 8.50 (d, J = 7.3 Hz, 1H), 8.28 (s, 1H), 8.15 (s, 1H), 8.08 (s, 1H), 8.04 (d, J = 2.1 Hz, 1H), 7.82 (d, J = 5.4 Hz, 1H), 7.07 (t, J = 7.1 Hz, 1H), 6.81 (s, 1H), 6.11 (q, J = 6.4 Hz, 1H), 4.04 (s, 3H), 1.94 (d, J = 6.4 Hz, 3H). Compound 31: (S)-N-(3,4-dichloro-2-fluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-6- (1-(pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 36%. LC-MS: m/z = 510.20 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.82 (d, J = 4.9 Hz, 2H), 8.52 (s, 1H), 8.35 (s, 1H), 8.22 (s, 1H), 7.94 (s, 1H), 7.76 (s, 1H), 7.54-7.51 (m, 1H), 7.61 (t, J = 5.0 Hz, 1H), 7.41-7.37 (m, 2H), 3.97 (s, 3H), 1.86 (d, J = 6.4 Hz, 3H).
Compound 32: (S)-N-(3-chloro-2,4-difluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-6- (1-(pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 52.8%. LC-MS: m/z = 494.20 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.83 (d, J = 4.8 Hz, 2H), 8.54 (s, 1H), 8.33 (s, 1H), 8.22 (s, 1H), 7.96 (s, 1H), 7.76 (s, 1H), 7.55 (dd, J = 14.0, 8.4 Hz, 1H), 7.40 (t, J = 4.8 Hz, 1H), 7.19 (t, J = 8.0 Hz, 1H), 5.95 (q, J = 6.4 Hz, 1H), 3.98 (s, 3H), 1.87 (d, J = 6.4 Hz, 3H). Compound 33: (S)-N-(3-chloro-2,4-difluorophenyl)-7-(1-cyclopropyl-1H-pyrazol-4- yl)-5-(1-(pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 51.5%. LC-MS: m/z = 520.25 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.84 (d, J = 4.9 Hz, 2H), 8.70 (s, 1H), 8.33 (s, 1H), 8.22 (s, 1H), 7.96 (s, 1H), 7.78 (s, 1H), 7.56 (dd, J = 14.1, 8.6 Hz, 1H), 7.42 (t, J = 4.9 Hz, 1H), 7.19 (t, J = 8.2 Hz, 1H), 5.96 (q, J = 6.2 Hz, 1H), 3.76-3.70 (m, 1H), 1.85 (d, J = 6.4 Hz, 3H), 1.20 – 1.08 (m, 4H). Compound 34: (R)-N-(4-fluoropyrazolo[1,5-a]pyridin-5-yl)-7-(1-methyl-1H-pyrazol- 4-yl)-6-(1-(pyrimidin-2-yl)ethoxy)quinazolin-4-amine. Yield 33%. LC-MS: m/z = 482.15 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.85 (d, J = 4.8 Hz, 2H), 8.58 (s, 1H), 8.45 (d, J = 7.6 Hz, 1H), 8.40 (s, 1H), 8.25 (s, 1H), 8.03-7.96 (m, 2H), 7.84 (s, 1H), 7.42 (t, J = 4.8 Hz, 1H), 7.19 (t, J = 7.2 Hz, 1H), 6.76 (s, 1H), 6.04-5.94 (m, 1H), 4.00 (s, 3H), 1.89 (d, J = 6.4 Hz, 3H). Compound 35: (S)-6-(1-(4-fluorophenyl)ethoxy)-N-(4-fluoropyrazolo[1,5-a]pyridin- 5-yl)-7-(1-methyl-1H-pyrazol-4-yl)quinazolin-4-amine . Yield 29%. LC-MS: m/z = 498.30 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.70 (s, 1H), 8.51 (d, J = 7.6 Hz, 1H), 8.41 (s, 1H), 8.19 (s, 1H), 8.05 (d, J = 2.0 Hz, 1H), 8.01 (d, J = 4.4 Hz, 2H), 7.49 (dd, J = 8.8, 5.2 Hz, 2H), 7.12-7.04 (m, 3H), 6.84 (d, J = 1.6 Hz, 1H), 5.94 (q, J = 6.8 Hz, 1H), 4.02 (s, 3H), 1.85 (d, J = 6.4 Hz, 3H). EXAMPLE 36: Synthesis of (S)-N-(3-chloro-2-fluorophenyl)-7-cyclopropoxy-6-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 36).
Step 1. Synthesis of 4-bromo-2-fluorophenyl ethyl carbonate To a solution of 4-bromo-2-fluorophenol (35 g, 183 mmol, 1 eq) and TEA (22.2 g, 220 mmol, 1.2 eq) in dry DCM (350 mL) at 0 °C was added dropwise a solution of ethyl carbonochloridate (23.9 g, 220 mmol, 1.2 eq). The reaction mixture was stirred at 0 °C for 1 h under N2, then allowed to warm to room temperature, quenched with H2O (100 mL) and extracted with DCM (3 x 100 mL). The combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure to afford the title compound (48.5 g, 100% yield) as yellow oil. 1HNMR (400 MHz, CDCl3) δ 7.33 (dd, J = 9.5, 2.3 Hz, 1H), 7.26 (ddd, J = 8.6, 2.3, 1.4 Hz, 1H), 7.12 – 7.06 (m, 1H), 4.37 – 4.29 (m, 2H), 1.37 (t, J = 7.2 Hz, 3H). Step 2. Synthesis of 4-bromo-2-fluoro-5-nitrophenyl ethyl carbonate To a solution of 4-bromo-2-fluorophenyl ethyl carbonate (48.5 g, 184 mmol, 1 eq) in H2SO4 (90 mL) was added dropwise HNO3 (13.1 mL, 276 mmol, 1.5 eq) at 0 °C. After 1 h, the reaction mixture was poured into ice water and extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with water, NaHCO3 and brine, dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure to afford the title compound (53.2 g, 93% yield) as a yellow oil. 1H NMR (400 MHz, CDCl3) δ 7.93 (d, J = 7.1 Hz, 1H), 7.58 (d, J = 8.9 Hz, 1H), 4.40 – 4.32 (m, 2H), 1.39 (t, J = 7.2 Hz, 3H). Step 3. Synthesis of 4-bromo-2-fluoro-5-nitrophenol To a solution of 4-bromo-2-fluoro-5-nitrophenyl ethyl carbonate (53.2 g, 173 mmol, 1 eq) in MeOH (550 mL) was added NaHCO3 (29.1 g, 345 mmol, 2 eq). The reaction mixture was stirred at 0 °C for 3 h. MeOH was evaporated under reduced pressure. Water (200 mL) was added to the residue and the aqueous layer was acidified to pH = 5 by the addition of 4N HCl. The aqueous layer was filtered. The filter cake was washed with water (x 3) and concentrated under reduced pressure to afford the title compound (29.4 g, 72% yield) as a
yellow solid. LC-MS: m/z = 234.10/236.10 [M+1]+. Step 4. Synthesis of 1-bromo-5-fluoro-2-nitro-4-(vinyloxy)benzene A mixture of 4-bromo-2-fluoro-5-nitrophenol (18.3 g, 77.5 mmol, 1 eq), vinyl acetate (33.4 g, 387 mmol, 5 eq), [Ir(COD)Cl]2 (1.57 g, 2.33 mmol, 0.03 eq) and Na2CO3 (4.93 g, 46.5 mmol, 0.6 eq) in toluene (190 mL) was stirred at 120 °C for 16 h under nitrogen in a sealed tube. The mixture was cooled, concentrated, and purified by prep-FCC (PE: EA=100:1) to afford the title compound (12.76 g, 62% yield) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ 7.69 (d, J = 7.5 Hz, 1H), 7.52 (d, J = 9.5 Hz, 1H), 6.59 (ddd, J = 13.5, 5.9, 0.7 Hz, 1H), 4.91 (dd, J = 13.5, 2.5 Hz, 1H), 4.69 (dd, J = 6.0, 2.5 Hz, 1H). Step 5. Synthesis of 1-bromo-4-cyclopropoxy-5-fluoro-2-nitrobenzene Diethylzinc (195 mL, 1M in hexane, 4 eq) was dissolved in dry DCM (200 mL) at 0 °C, then TFA (22.2 g, 195 mmol, 4 eq) in dry DCM was added dropwise at 0 °C, 10 min later diiodomethane (52.2 g, 195 mmol, 4 eq) in dry DCM was added dropwise at 0 °C. After stirring the mixture at 0 °C for 15 min, 1-bromo-5-fluoro-2-nitro-4-(vinyloxy)benzene (12.76 g, 48.7 mmol, 1 eq) in dry DCM was added dropwise. The reaction mixture was stirred at 0 to 30 °C for 16 h under nitrogen atmosphere. The mixture was quenched with 1N HCl, extracted with DCM (3 x 100 mL). The combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated and purified by silica gel column chromatography, eluting with (PE:EA=100:1) to afford the title compound (4.27 g, 31% yield) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ 7.88 (d, J = 7.7 Hz, 1H), 7.42 (d, J = 9.9 Hz, 1H), 3.85 (dt, J = 5.7, 2.7 Hz, 1H), 0.90 – 0.86 (m, 4H). Step 6. Synthesis of 4-cyclopropoxy-5-fluoro-2-nitrobenzonitrile To a solution of 1-bromo-4-cyclopropoxy-5-fluoro-2-nitrobenzene (4.27 g, 15.5 mmol, 1 eq) in NMP (50 mL) was added CuCN (1.8 g, 20 mmol, 1.3 eq) at room temperature and stirred at 130 °C for 2 h under nitrogen. The mixture was filtered through diatomite and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (5 x 20 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure and purified by silica gel column chromatography, eluting with (PE:EA=80:1) to afford the title compound (2.35 g, 68% yield) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ 8.24 (d, J = 7.2 Hz, 1H), 7.54 (d, J = 9.6 Hz, 1H), 4.02 – 3.94 (m, 1H), 1.03 – 0.89 (m, 4H).
Step 7. Synthesis of (S)-4-cyclopropoxy-2-nitro-5-(1-(pyrimidin-2-yl)ethoxy)benzonitrile To a solution of (S)-1-(pyrimidin-2-yl)ethan-1-ol (251 mg, 2 mmol, 1.5 eq) in THF (5 mL) was added NaH (81 mg, 2 mmol, 1.5 eq) at 0 °C and stirred for 0.5 h under nitrogen. A solution of 4-cyclopropoxy-5-fluoro-2-nitrobenzonitrile (300 mg, 1.35 mmol, 1 eq) was added and then stirred at 0 °C to room temperature for 0.5 h. The mixture was quenched with an aqueous saturated NH4Cl (5 mL) solution and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography, eluting with (PE: EA = 1:1) to afford the title compound (440 mg, 98% yield) as a yellow solid. LCMS: m/z 327.15 [M+1]+. Following the above procedure, intermediates 1B-17-2 and 1B-17-3 were prepared.
Step 8. Synthesis of (S)-2-amino-4-cyclopropoxy-5-(1-(pyrimidin-2-yl)ethoxy)benzonitrile To a solution of (S)-4-cyclopropoxy-2-nitro-5-(1-(pyrimidin-2-yl)ethoxy)benzonitrile (200 mg, 0.61 mmol, 1 eq) in THF (3 mL) and H2O (3 mL) was added Na2S2O4 (320 mg, 1.84 mmol, 3 eq) at 0 °C and stirred at 0 °C for 2 minutes under nitrogen atmosphere. The mixture was quenched with aqueous NaHCO3, extracted with EtOAc (3 x 10 mL). The combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure to afford the title compound (79 mg, 43% yield) as a yellow solid.
LC-MS: m/z = 297.35 [M+1]+. Following the above procedure, intermediates 1B-18-2 and 1B-18-3 were prepared.
Step 9. Synthesis of (S)-N-(3-chloro-2-fluorophenyl)-7-cyclopropoxy-6-(1-(pyrimidin-2- yl)ethoxy)quinazolin-4-amine (Compound 36) To a solution of (S)-2-amino-4-cyclopropoxy-5-(1-(pyrimidin-2-yl)ethoxy)- benzonitrile (79 mg, 0.27 mmol, 1 eq) in 2-MeTHF (4 mL) was added (E)-N,N'-bis(3-chloro- 2-fluorophenyl)formimidamide (100 mg, 0.33 mmol, 1.25 eq) at room temperature. The reaction mixture was heated to 80 °C and AcOH (0.4 mL) was added. After stirring the reaction mixture at 90 °C for 16 h, the mixture was cooled to rt and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with aqueous NaHCO3 (10 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated and purified by prep-HPLC to afford the desired product (28 mg, 23% yield) as a white solid. LC-MS: m/z = 452.20 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.78 (d, J = 4.9 Hz, 2H), 8.31 (s, 1H), 7.61 (s, 1H), 7.56 –7.50 (m, 2H), 7.39 (t, J = 4.9 Hz, 1H), 7.36 (t, J = 7.3 Hz, 1H), 7.18 (t, J = 8.2 Hz, 1H), 5.68 (q, J = 6.5 Hz, 1H), 4.01 – 3.92 (m, 1H), 1.78 (d, J = 6.5 Hz, 3H), 0.95 – 0.88 (m, 2H), 0.86 – 0.79 (m, 2H). EXAMPLES 37 and 38 Following the procedure described in Example 36, Compounds 37 and 38 were prepared.
Compound 37: N-(3-chloro-2-fluorophenyl)-7-cyclopropoxy-6-(1-(5-(difluorometh- yl)pyrimidin-2-yl)-ethoxy)quinazolin-4-amine. Yield 33.4%. LC-MS: m/z = 502.25 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.96 (s, 2H), 8.32 (s, 1H), 7.65 (s, 1H), 7.56 (s, 1H), 7.51 (t, J = 7.2 Hz, 1H), 7.36 (t, J = 7.2 Hz, 1H), 7.18 (t, J = 8.0 Hz, 1H), 6.95 (t, J = 55.0 Hz, 1H), 5.76 (q, J = 6.8 Hz, 1H), 3.98-3.94 (m, 1H), 1.80 (d, J = 6.8 Hz, 3H), 0.94-0.89 (m, 2H), 0.82-0.78 (m, 2H). Compound 38: N-(3-chloro-2-fluorophenyl)-7-cyclopropoxy-6-(1-(4-(difluorometh- yl)phenyl)-ethoxy)quinazolin-4-amine. Yield 29.8%. LC-MS: m/z = 500.25 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.31 (s, 1H), 7.67 (s, 1H), 7.56-7.54 (m, 4H), 7.52-7.49 (m, 2H) 7.37 (t, J = 6.8 Hz, 1H), 7.19 (t, J = 7.6 Hz, 1H), 6.70 (t, J = 56.0 Hz, 1H), 5.69 (q, J = 6.0 Hz, 1H), 4.02-3.99 (m, 1H), 1.68 (d, J = 6.4 Hz, 3H), 0.97-0.93 (m, 2H), 0.87-0.82 (m, 2H). EXAMPLE 39: Synthesis of N-(3-chloro-2-fluorophenyl)-7-cyclopropoxy-6-{[2-(pyrimidin- 2-yl)propan-2-yl]oxy}quinazolin-4-amine (Compound 39).
Step 1. Synthesis of 4-cyclopropoxy-2-nitro-5-{[2-(pyrimidin-2-yl)propan-2-yl]oxy}- benzonitrile To a solution of 2-(pyrimidin-2-yl)propan-2-ol (251 mg, 1.82 mmol, 1.5 eq) in dry THF (5 mL) was added NaH (44 mg, 1.82 mmol, 1.5 eq) at 0 °C and stirred for 0.5 h under nitrogen. The solution of 4-cyclopropoxy-5-fluoro-2-nitrobenzonitrile (270 mg, 1.21 mmol, 1 eq) in dry THF (5 mL) was added and then stirred at room temperature for 0.5 h. The mixture was quenched with saturated NH4Cl (5 mL) and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography, eluting with (PE: EA=3:1) to afford the title compound (370 mg, 89% yield) as a yellow solid. LC-MS: m/z = 341.10 [M+1]+. Step 2. Synthesis of 2-amino-4-cyclopropoxy-5-{[2-(pyrimidin-2-yl)propan-2-yl]oxy}- benzonitrile To a solution of 4-cyclopropoxy-2-nitro-5-{[2-(pyrimidin-2-yl)propan-2-yl]oxy}- benzonitrile (288 mg, 0.85 mmol, 1 eq) in THF (3 mL) and H2O (3 mL) was added Na2S2O4
(442 mg, 2.54 mmol, 3 eq) at 0 °C and stirred at 0 °C for 10 min under nitrogen. The mixture was quenched with NaHCO3 (aq), extracted with EtOAc (3 x 10 mL). The combined organic layers were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure and purified by silica gel column chromatography, eluting with (DCM: MeOH=50:1) to afford the title compound (83 mg, 31% yield) as a yellow solid. LC-MS: m/z = 311.10 [M+1]+. Step 3. Synthesis of N-(3-chloro-2-fluorophenyl)-7-cyclopropoxy-6-{[2-(pyrimidin-2-yl)- propan-2-yl]oxy}quinazolin-4-amine (Compound 39) To a solution of 2-amino-4-cyclopropoxy-5-{[2-(pyrimidin-2-yl)propan-2-yl]oxy}- benzonitrile (83 mg, 0.27 mmol, 1 eq) in 2-MeTHF (5 mL) was added (E)-N,N'-bis(3-chloro- 2-fluoro-phenyl)formimidamide (100 mg, 0.33 mmol, 1.25 eq) at room temperature. The reaction mixture was heated to 80 °C and AcOH (0.5 mL) was added. The reaction mixture was stirred at 90 °C for 16 h, then cooled to rt and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with NaHCO3(aq) (10 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated and purified by prep-HPLC to afford the desired product (62.1 mg, 49% yield) as a white solid. LC-MS: m/z = 466.05 [M+1]+ 1H NMR (400 MHz, CD3OD) δ 8.79 (d, J = 4.9 Hz, 2H), 8.33 (s, 1H), 7.56 (s, 1H), 7.53 (t, J = 7.6 Hz, 1H), 7.49 (s, 1H), 7.40 (t, J = 4.9 Hz, 1H), 7.37 – 7.33 (m, 1H), 7.20 – 7.15 (m, 1H), 3.77 – 3.74 (m, 1H), 1.80 (s, 6H), 0.81 – 0.78 (m, 2H), 0.58 – 0.54 (m, 2H). EXAMPLE 40: Synthesis of (S)-N-(3-chloro-2-fluorophenyl)-N-methyl-7-(1-methyl-1H- pyrazol-4-yl)-6-(1-(pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 40). To a mixture of (S)-N-(3-chloro-2-fluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-6-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 27, 80 mg, 0.19 mmol, 1 eq.) and Cs2CO3 (109 mg, 0.34 mmol, 2 eq.) in DMF (2 mL), iodomethane (71.6 mg, 0.5 mmol, 3 eq.) was added dropwise and the reaction was stirred at rt for 1 h. The mixture was diluted with EA (20 mL), washed with brine (10 mL x3), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure, and the residue was purified by Prep-HPLC (0.04% NH4HCO3 in H2O/CH3CN) to afford the title compound (20 mg, 0.041 mmol, 24.3%) as a yellow solid. LC-MS: m/z = 490.05 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.81 (d, J = 4.8 Hz, 2H), 8.52 (s, 1H), 8.23 (s, 1H), 7.82 (s, 1H), 7.77 (s, 1H), 7.61 (s, 1H), 7.39 (t, J = 4.8 Hz, 1H), 7.05 (td, J = 8.4, 6.8 Hz, 2H), 6.92 (td, J = 7.2, 2.0 Hz, 1H), 5.80 (q, J = 6.4 Hz, 1H), 3.97 (s, 3H), 3.69 (s, 3H), 1.83 (d, J = 6.4 Hz, 3H).
EXAMPLE 41: Synthesis of (S)-N-(3-chloro-2-fluorophenyl)-N-(difluoromethyl)-7-(1- methyl-1H-pyrazol-4-yl)-6-(1-(pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 41). A mixture of (S)-N-(3-chloro-2-fluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-6-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 27, 80 mg, 0.17 mmol, 1 eq.), sodium 2-chloro-2,2-difluoroacetate (76.9 mg, 0.5 mmol, 3 eq.) and dipotassium carbonate (69.7 mg, 0.5 mmol, 3 eq.) in CH3CN (5 mL) was stirred at 60 °C for 16 h. The mixture was diluted with EA (50 mL), washed with brine (5 mL x 3), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by Prep-HPLC (0.04% NH4HCO3 in H2O/CH3CN) to afford the title compound (21.52 mg, 0.041 mmol, 24.3%) as a yellow solid. LC-MS: m/z = 526.25 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.82 (d, J = 4.8 Hz, 2H), 8.46 (s, 1H), 8.12 (s, 1H), 8.04 (s, 1H), 7.84-7.79 (m, 2H), 7.57 (t, J = 57.6 Hz, 1H), 7.41 (t, J = 4.8 Hz, 1H), 7.15-7.01 (m, 2H), 6.91 (td, J = 7.6, 2.0 Hz, 1H), 5.81 (q, J = 6.5 Hz, 1H), 3.99 (s, 3H), 1.85 (d, J = 6.4 Hz, 3H). EXAMPLE 42: Synthesis of N-(3-chloro-2-fluorophenyl)-7-methoxy-6-((2-(3-methyl- pyridin-4-yl)propan-2-yl)oxy)quinazolin-4-amine (Compound 42).
Step 1. Synthesis of 4-(2-(5-bromo-2-methoxy-4-nitrophenoxy)propan-2-yl)-3-methylpyridine To a mixture of NaH (0.48 g, 12 mmol) in DMF (25 mL) was added 2-(3- methylpyridin-4-yl)propan-2-ol (0.91 g, 6 mmol) under N2 atmosphere. The reaction mixture was stirred at 0 °C for 1 h. Then 1-bromo-5-fluoro-4-methoxy-2-nitrobenzene (1.5 g, 6 mmol) was added. The resulting mixture was stirred at rt for 1 h, then poured into water (25 ml), extracted with EA (25 mL x 3), washed with brine solution (25 mL x 3), concentrated, and purified by flash chromatography with PE/EA=5/1-2/1 to give 4-[2-(5-bromo-2-methoxy-4- nitrophenoxy)propan-2-yl]-3-methylpyridine (1.2 g, 2.833 mmol, 47%) as a green solid. LC-MS: m/z = 381.35/383.35 [M+1]+.
Step 2. Synthesis of methyl 4-methoxy-5-((2-(3-methylpyridin-4-yl)propan-2-yl)oxy)-2- nitrobenzoate To a solution of 4-{2-[(5-bromo-2-methoxy-4-nitrophenyl)oxy]prop-2-yl}-3-methyl- pyridine (1 g, 2.623 mmol) in CH3OH (25 mL) was added Pd(dppf)Cl2 (0.38 g, 0.525 mmol) under CO. The reaction mixture was stirred at 70 °C for 18 h, then poured into water (25 ml), cooled to rt and extracted with EA (25.0 ml x 3), washed with NaCl solution (25 ml x 3), concentrated, and purified by flash chromatography with PE/EA=5/1-2/1 to give methyl 4- methoxy-5-{[2-(3-methylpyridin-4-yl)prop-2-yl]oxy}-2-nitrobenzoate (700 mg, 1.9 mmol, 73%) as a green solid. LC-MS: m/z = 361.4 [M+1]+. Step 3. Synthesis of methyl 2-amino-4-methoxy-5-((2-(3-methylpyridin-4-yl)propan-2-yl)oxy) benzoate To a solution of methyl 4-methoxy-5-{[2-(3-methylpyridin-4-yl)prop-2-yl]oxy}-2- nitrobenzoate (700 mg, 1.9 mmol) in CH3OH (25 mL) was added Pd/C under H2. The reaction mixture was stirred at rt for 1 h, then poured into water (25 ml), extracted with EA (25 ml x 3), washed with brine (25 ml x 3), concentrated under reduced pressure, and purified by flash chromatography with PE/EA=5/1-2/1 to give methyl 2-amino-4-methoxy-5-{[2-(3-methyl- pyridin-4-yl)prop-2-yl]oxy}benzoate (500 mg, 1.4 mmol, 70%) as an colorless solid. LC-MS: m/z = 331.4 [M+1]+. Step 4. Synthesis of 7-methoxy-6-((2-(3-methylpyridin-4-yl)propan-2-yl)oxy)quinazolin-4-ol To a solution of methyl 2-amino-4-methoxy-5-{[2-(3-methylpyridin-4-yl)prop-2-yl]- oxy}benzoate (500 mg, 1.5 mmol) in ethanol (25 mL) was added formamidine acetate (315 mg, 3 mmol) under N2 atmosphere. The reaction mixture was stirred at 85°C for 18 h, then poured into water (25 mL), cooled to rt, and extracted with EA (25 mL x 3). The combined organic layers were washed with brine (25 ml x 3), concentrated under reduced pressure, and purified by flash chromatography with PE/EA=5/1-2/1 to give 7-methoxy-6-{[2-(3-methylpyridin-4- yl)prop-2-yl]oxy}quinazolin-4-ol (300 mg, 0.83 mmol, 55%) as a colorless solid. LC-MS: m/z = 326.4 [M+1]+. Step 5. Synthesis of 4-chloro-7-methoxy-6-((2-(3-methylpyridin-4-yl)propan-2-yl)oxy) quinazoline To a solution of 7-methoxy-6-{[2-(3-methylpyridin-4-yl)prop-2-yl]oxy}quinazolin-4- ol (300 mg, 0.92 mmol) in CHCl3 (25 mL) was added (COCl)2 (234 mg, 1.8 mmol) and DMF (3.37 mg, 0.046 mmol) under N2. The reaction mixture was stirred at 85 °C for 2 h, then poured into water (25 mL), cooled to rt, and extracted with EA (25 mL x 3). The combined organic
extract was washed with brine (25 mL x 3), concentrated under reduced pressure, and purified by flash chromatography with PE/EA=3/1-1/1 to give 4-chloro-7-methoxy-6-{[2-(3- methylpyridin-4-yl)prop-2-yl]oxy}quinazoline (200 mg, 0.52 mmol, 57%) as a colorless solid. LC-MS: m/z = 343.8 [M+1]+. Step 6. Synthesis of N-(3-chloro-2-fluorophenyl)-7-methoxy-6-((2-(3-methylpyridin-4- yl)propan-2-yl)oxy)quinazolin-4-amine (Compound 42) To a solution of 4-chloro-7-methoxy-6-{[2-(3-methylpyridin-4-yl)prop-2-yl]oxy}qui- nazoline (250 mg, 0.727 mmol) in IPA (25 mL) was added 3-chloro-2-fluoroaniline (423 mg, 2.9 mmol) under N2. The reaction mixture was stirred at 100 °C for 18 h, then poured into water (25 ml), cooled to rt, and extracted with EA (25 mL x3). The combined organic extract was washed with a saturated NaCl solution (25 mL x 3), concentrated under reduced pressure, and purified by flash chromatography with PE/EA=3/1-1/1 to give the title product N-(3-chloro-2- fluorophenyl)-7-methoxy-6-((2-(3-methylpyridin-4-yl)propan-2-yl)oxy)quinazolin-4-amine (180 mg, 0.39 mmol, 54%) as a white solid. LC-MS: m/z = 453.0 [M+1]+. 1H NMR (400 MHz, DMSO) δ 9.32 (s, 1H), 8.43 (d, J = 4.0 Hz, 1H), 8.37 (s, 1H), 8.32 (s, 1H), 7.55 – 7.43 (m, 3H), 7.39 (s, 1H), 7.30 – 7.23 (m, 2H), 3.89 (s, 3H), 2.44 (s, 3H), 1.81 (s, 6H). EXAMPLES 43-49 Compounds 43-49 of this invention were prepared following the procedure described in Example 1. N-(3-chloro-2,4-difluorophenyl)-7-(1H-pyrazol-4-yl)-5-((2-(pyrimidin-2-yl)propan-2- yl)oxy)quinazolin-4-amine (Compound 43) was prepared from (1H-pyrazol-4-yl)boronic acid (CAS:763120-58-7) and intermediate 1A-13-10. LC-MS: m/z = 494.10, [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.68 (s, 1H), 8.66 (d, J = 4.8 Hz, 2H), 8.32 (s, 2H), 7.83- 7.71 (m, 1H), 7.62 (s, 1H), 7.57 (s, 1H), 7.41 (t, J = 4.8 Hz, 1H), 7.36 (td, J = 9.0, 2.0 Hz, 1H). N-(3-chloro-4-fluorophenyl)-7-(1H-pyrazol-4-yl)-5-((2-(pyrimidin-2-yl)propan-2- yl)oxy)quinazolin-4-amine (Compound 44) was prepared from (1H-pyrazol-4-yl)boronic acid (CAS:763120-58-7) and intermediate 1A-13-11. LC-MS: m/z = 476.15, [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.68 (d, J = 4.9 Hz, 2H), 8.63 (s, 1H), 8.19 (s, 2H), 7.91 (dd, J = 6.5, 2.4 Hz, 1H), 7.62 – 7.56 (m, 1H), 7.48 (s, 2H), 7.46 – 7.39 (m, 2H), 2.17 (s, 6H).
(S)-N-(3-chloro-4-fluorophenyl)-7-(1H-pyrazol-4-yl)-5-(1-(pyrimidin-2-yl)ethoxy)- quinazolin-4-amine (Compound 45) was prepared from (1H-pyrazol-4-yl)boronic acid (CAS: 763120-58-7) and intermediate 1A-13-4. LC-MS: m/z = 462.10, [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.77 (d, J = 4.9 Hz, 2H), 8.69 (s, 1H), 8.35 (s, 2H), 8.07 (dd, J = 6.7, 2.7 Hz, 1H), 7.81-7.75 (m, 2H), 7.50 (s, 1H), 7.48 – 7.42 (m, 2H), 6.27 (q, J = 6.6 Hz, 1H), 1.90 (d, J = 6.4 Hz, 3H). N-(3-chloro-2,4-difluorophenyl)-7-(1-(methyl-d3)-1H-pyrazol-4-yl)-5-((2-(pyrimidin- 2-yl)propan-2-yl)oxy)quinazolin-4-amine (Compound 46) was prepared from (1-(methyl-d3)- 1H-pyrazol-4-yl)boronic acid (CAS: 2256709-52-9) and intermediate 1A-13-10. LC-MS: m/z = 511.30, [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.72–8.55 (m, 3H), 8.33 (s, 1H), 8.03 (s, 1H), 7.82- 7.69 (m, 1H), 7.57 (s, 1H), 7.51 (s, 1H), 7.41 (t, J = 4.8 Hz, 1H), 7.36 (t, J = 8.8 Hz, 1H), 2.19 (s, 6H). N-(3-chloro-4-fluorophenyl)-7-(1-(methyl-d3)-1H-pyrazol-4-yl)-5-((2-(pyrimidin-2- yl)propan-2-yl)oxy)quinazolin-4-amine (Compound 47) was prepared from (1-(methyl-d3)- 1H-pyrazol-4-yl)boronic acid (CAS: 2256709-52-9) and intermediate 1A-13-11. LC-MS: m/z = 493.20, [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.68 (d, J = 4.9 Hz, 2H), 8.62 (s, 1H), 8.28 (s, 1H), 7.97 (s, 1H), 7.92-7.87 (m, 1H), 7.61-7.56 (m, 1H), 7.47-7.42 (m, 3H), 7.41 (t, J = 4.8 Hz, 1H), 2.17 (s, 6H). (S)-N-(3-chloro-2,4-difluorophenyl)-7-(1-(methyl-d3)-1H-pyrazol-4-yl)-5-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 48) was prepared from (1-(methyl- d3)-1H-pyrazol-4-yl)boronic acid (CAS: 2256709-52-9) and intermediate 1A-13-7. LC-MS: m/z = 497.10, [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.71 (s, 1H), 8.69 (d, J = 2.7 Hz, 2H), 8.45 (s, 1H), 8.20 (s, 1H), 7.85-7.78 (m, 1H), 7.71 (s, 1H), 7.52 (s, 1H), 7.43 (t, J = 4.9 Hz, 1H), 7.34 (t, J = 8.8 Hz, 1H), 6.24 (q, J = 6.4 Hz, 1H), 1.91 (d, J = 6.4 Hz, 3H). (S)-N-(3-chloro-4-fluorophenyl)-7-(1-(difluoromethyl)-1H-pyrazol-4-yl)-5-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 49) was prepared from (1-(difluoro- methyl)-1H-pyrazol-4-yl)boronic acid (CAS:1312693-57-4) and intermediate 1A-13-4. LC-MS: m/z = 512.10, [M+1]+.
1H NMR (400 MHz, CD3OD) δ 8.92 (s, 1H), 8.77 (d, J = 4.9 Hz, 2H), 8.71 (s, 1H), 8.37 (s, 1H), 8.07 (dd, J = 6.6, 2.6 Hz, 1H), 7.82 (s, 1H), 7.80 – 7.75 (m, 1H), 7.60 – 7.58 (m, 2H), 7.49 – 7.43 (m, 2H), 6.28 (q, J = 6.4 Hz, 1H), 1.90 (d, J = 6.4 Hz, 3H). EXAMPLE 50: Synthesis of N-(3-chloro-2,4-difluorophenyl)-7-(pyrimidin-2-yl)-5-((2- (pyrimidin-2-yl)propan-2-yl)oxy)quinazolin-4-amine (Compound 50).
A mixture of 7-bromo-N-(3-chloro-2,4-difluorophenyl)-5-((2-(pyrimidin-2-yl)propan- 2-yl)oxy)quinazolin-4-amine (IA-13-10, 86 mg, 0.17 mmol, 1 eq.), 2-(tributylstannyl)- pyrimidine (75 mg, 0.203 mmol, 1.2 eq.), CuI (3.23 mg, 0.017 mmol, 0.1 eq.) and Pd(dppf)Cl2 (12.4 mg, 0.017 mmol, 0.1 eq.) in DMF (3 mL) was degassed and purged with nitrogen three times and then the mixture was stirred at 100 °C for 24 h under nitrogen atmosphere. The resulting mixture was diluted with H2O (10 mL), cooled to rt, and extracted with DCM/MeOH (10/1) (3 x 30 mL). The combined organic layers were washed with 10% aqueous KF (10 mL x 2), brine (10 mL), dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The resulting residue was purified by Prep-HPLC (TFA (0.1%) in H2O/CH3CN) to afford the title compound (13.4 mg, 15.6% yield) as a gray solid. LC-MS: m/z = 506.20 [M+1]+. 1H NMR (400 MHz, CD3OD) δ 8.95 (d, J = 4.8 Hz, 2H), 8.68 (d, J = 4.8 Hz, 2H), 8.67 (s, 1H), 8.49 (d, J = 1.2 Hz, 1H), 8.36 (d, J = 1.2 Hz, 1H), 7.92 (td, J = 8.8, 5.6 Hz, 1H), 7.49 (t, J = 4.8 Hz, 1H), 7.40 (t, J = 4.8 Hz, 1H), 7.37-7.29 (m, 1H), 2.19 (s, 6H). EXAMPLES 51-58 Compounds 51-58 of this invention can be readily prepared following the procedure described in Example 1. N-(3-chloro-4-fluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-5-(1-(4-methyl-4H-1,2,4- triazol-3-yl)ethoxy)quinazolin-4-amine (Compound 51) can be prepared from 1-(4-methyl- 4H-1,2,4-triazol-3-yl)ethan-1-ol (CAS:149762-18-5) and intermediate 1A-12-4.
N-(3-chloro-4-fluorophenyl)-7-(1-methyl-1H-pyrazol-4-yl)-5-(1-(pyrimidin-2-yl) cyclopropoxy)quinazolin-4-amine (Compound 52) can be prepared from 1-(pyrimidin-2- yl)cyclopropan-1-ol (CAS:2229094-28-2) and intermediate 1A-12-4.
N-(3-chloro-4-fluorophenyl)-5-(1-(1-methyl-1H-imidazol-2-yl)ethoxy)-7-(1-methyl- 1H-pyrazol-4-yl)quinazolin-4-amine (Compound 53) can be prepared from 1-(1-methyl-1H- imidazol-2-yl)ethan-1-ol (CAS:41507-36-2) and intermediate 1A-12-4.
(S)-N-(3-chloro-2,4-difluorophenyl)-7-(1-(difluoromethyl)-1H-pyrazol-4-yl)-5-(1- (pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 54) can be prepared from (1-(di- fluoromethyl)-1H-pyrazol-4-yl)boronic acid (CAS:1312693-57-4) and intermediate 1A-13-7. N-(3-chloro-2,4-difluorophenyl)-7-(isothiazol-5-yl)-5-((2-(pyrimidin-2-yl)propan-2- yl)oxy)quinazolin-4-amine (Compound 55) can be prepared from isothiazole-5-boronic acid (CAS:1162262-43-1) and intermediate 1A-13-7. N-(3-chloro-2,4-difluorophenyl)-5-(1-(1-(methyl)-1H-indol-2-yl)ethoxy)-7-(1- (methyl-d3)-1H-pyrazol-4-yl)quinazolin-4-amine (Compound 56) can be prepared from 1-(1- methylindol-2-yl)ethanol (CAS: 29124-10-5), (1-(methyl-d3)-1H-pyrazol-4-yl)boronic acid (CAS: 2256709-52-9), and intermediate 1A-12-3.
N-(3-chloro-4-fluorophenyl)-7-(1-cyclopropyl-1H-pyrazol-4-yl)-5-((2-(pyrimidin-2- yl)propan-2-yl)oxy)quinazolin-4-amine (Compound 57) can be prepared from (1-cyclopropyl- 1H-pyrazol-4-yl)boronic acid (1678534-30-9) and intermediate 1A-13-11. N-(3-chloro-2,4-difluorophenyl)-7-(1-(methyl-d3)-1H-pyrazol-4-yl)-5-((2-(thiazol-2- yl)propan-2-yl)oxy)quinazolin-4-amine (Compound 58) can be prepared from 2-thiazol-2-yl- propan-2-ol (CAS:16077-78-4), (1-(methyl-d3)-1H-pyrazol-4-yl)boronic acid (CAS: 2256709- 52-9) and intermediate 1A-12-3.
EXAMPLE 59: (S)-N-(3-chloro-2,4-difluorophenyl)-7-(1-(methyl-d3)-1H-pyrazol-4-yl)-6- (1-(pyrimidin-2-yl)ethoxy)quinazolin-4-amine (Compound 59) Compound 59 can beprepared from (1-(methyl-d3)-1H-pyrazol-4-yl)boronic acid (CAS: 2256709-52-9) and intermediate 1B-6-4 following the procedure of Example 27. EXAMPLE 60: N-(3-chloro-2,4-difluorophenyl)-5-((2-(pyrimidin-2-yl)propan-2-yl)oxy)-7- (thiazol-2-yl)quinazolin-4-amine (Compound 60) Compound 60 can be prepared from 1,3-thiazole-2-boronic acid (CAS: 389630-95-9) and intermediate 1A-13-10 following the procedure of Example 1. EXAMPLE 61: (S)-N-(3-chloro-2,4-difluorophenyl)-7-(pyrimidin-2-yl)-5-(1-(pyrimidin-2- yl) ethoxy)quinazolin-4-amine (Compound 61) Compound 61 can be prepared following the procedure of Example 50, using intermediate 1A-13-7 and 2-(tributylstannyl)pyrimidine (CAS# 153435-63-3). EXAMPLE 62: N-(3-chloro-2,4-difluorophenyl)-7-methoxy-5-((2-(pyrimidin-2-yl)propan-2- yl)oxy)quinazolin-4-amine (Compound 62)
Compound 62 can be prepared following general scheme II, Step 4, using intermediate 1A-13-10 and methanol. EXAMPLE 63: (S)-N-(3-chloro-2,4-difluorophenyl)-7-methoxy-5-(1-(pyrimidin-2-yl)eth- oxy)quinazolin-4-amine (Compound 63) Compound 63 can be prepared following general scheme II, Step 4, using intermediate 1A-13-9 and methanol. EXAMPLE 64: N-(3-chloro-4-fluorophenyl)-7-methoxy-5-((2-(pyrimidin-2-yl)propan-2-yl)- oxy)quinazolin-4-amine (Compound 64) Compound 64 can be prepared following general scheme II, Step 4, using intermediate 1A-13-11 and methanol. EXAMPLE 65: (S)-N-(3-chloro-4-fluorophenyl)-7-methoxy-5-(1-(pyrimidin-2-yl)ethoxy) quinazolin-4-amine (Compound 65) Compound 65 can be prepared following general scheme II, Step 4, using intermediate 1A-13-4 and methanol. Biological assays pERK Potency assay: EGFR mediates downstream signaling cascades, i.e., ERK phosphorylation in cells with EGFR aberrations. EGFR aberrations become constitutively active and thus induces cascades of cellular signaling events that result in increased phosphorylation of ERK at Threonine 202 and Tyrosine 204 (pERK). The so-called In Cell ELISA procedure as described below measures the level of cellular pERK in response to EGFR specific inhibitors in EGFR aberrations of three EGFR cell lines: PC-9 (EGFR/del19), A431 (EGFR amplification), and 11-18 (EGFR/L858R). PC-9 cells were obtained from the American Type Culture Collection (ATCC, Manassas, Virginia) and 11-18 and A431 cells were commercially available from RIKEN Cell Bank (Japan). PC-9 and 11-18 cells were grown and maintained using RPMI-1640 medium supplemented with 10% heat-inactivated fetal bovine serum. A431 cells were grown and maintained using DMEM (low glucose) supplemented with 10% heat-inactivated fetal bovine serum. On the day before a compound was added, cells were plated in poly-D-lysine coated 96-well cell culture plates (Corning® Biocoat® Cat#356640) at 50000 cells/well/100 μL for PC-9 and 11-18 cells and 20000 cells/well/100 μL for A431 cells. They were grown overnight in an incubator at 37°C with 5% CO2. Test compounds were prepared with 3-fold serial dilutions in DMSO, with a top concentration of 10 mM. On the day of the assay, 50 μL of a test compound diluted in media was added to each well of cell culture plate with the final
concentration of the compound spanning from 0.00005 μM to 10 μM. After adding the test compound, the cells were incubated for 3 hours at 37°C with 5% CO2. After the culture medium was removed, the cells were fixed with 200 μL of 4% formaldehyde in phosphate- buffered saline (PBS) and incubated at RT for 20 min. The plate was then washed three times with PBST (PBS with 0.05% Tween® 20), followed by incubation with 100 μL of pre-cooled methanol at -20°C for 20 min. After incubation, methanol was removed. The plate was washed with PBST. The cells were then permeabilized with 100 μL/well of 0.1% TritonTM X-100 in PBS at RT for 20 min and then quenched with a buffer (PBST containing 1% H2O2 and 0.1% sodium azide) for 20 min at RT with gentle shaking. After washing twice with PBST, a blocking buffer (250 ul/well, Pierce™ Protein-Free-PBS Blocking Buffer, Cat# 37572) was added to the cells for 1-h incubation at RT, followed by anti-pERK antibody (cell signaling, 1:1000 dilution in 5% BSA in PBST) incubation overnight at 4°C and secondary antibody- HRP (Jackson immunoresearch,1:3000 dilution in 1% BSA in PBST) incubation at RT for 1h. PBST wash was carried out after each step three times. Before adding a substrate (ELISABrightTM, commercially available from Advansta, San Jose, California), the plate was washed with PBS twice to remove detergent residues. The cellular pERK level was determined using a microplate reader (Biotek® SynergyTM H1, Agilent Technologies Inc., Santa Clara, California) to detect the chemiluminescence signal. IC50 values were determined by fitting a 4-parameter sigmoidal concentration-response model. The pERK assay results showed that each of Compounds 1-34 ,36, 37, and 39-41 has an IC50 of no more than 10 nM and each of Compounds 35, 38, and 42 has an IC50 between 10.1 nm and 50 nM. The results demonstrated that the compounds of this invention are highly effective in inhibiting EGFR. 2D Cell Proliferation Assay: A cell proliferation assay was used to examine the potency with which compounds inhibit in vitro cell proliferation of cancer cell lines carrying EGFR aberrations or EGFR wild type. The assay demonstrated the molecular mode of action of compounds. Low IC50 values are indicative of high potency of EGFR inhibitors. It was observed that EGFR inhibitors demonstrated potent inhibitory effects on the proliferation of human cancer cell lines or Baf/3 mouse cell lines carrying EGFR aberrations. The cell proliferation assay was performed in two-dimensional (2D) anchorage-dependent conditions in 96 well plates (Black/Clear Flat Bottom commercially available from Corning Inc., Corning, New York) with the following cell lines:
Ba/F3 EGFR-Del19/C797S (human EGFR): a mouse Ba/F3 cell line stably expressed exogenous human EGFR with del19 and C797S mutation, Ba/F3 EGFR-L858/C797S (human EGFR): a mouse Ba/F3 cell line stably expressed exogenous human EGFR with L858R and C797S mutation, Ba/F3 EGFR-C797S (human EGFR): a mouse Ba/F3 cell line stably expressed exogenous human EGFR with C797S mutation, PC-9: a human lung cancer cell line with EGFR del19 mutation, 11-18: a human lung cancer cell line with EGFR L858R mutation, A431: a human epidermoid cancer cell line with over expression of wild type EGFR, and H1568: a human lung cancer cell line with over expression of wild type EGFR. Cell lines were purchased from the American Type Culture Collection (ATCC, Manassas, Virginia) or RIKEN Cell Bank (Japan). All Baf/3 EGFR mutant cell lines were commercially available from Kyinn Inc (China). All cell lines were maintained in RPMI-1640 or DMEM supplemented with 10% heat-inactivated fetal bovine serum. 2D cell proliferation Assay conditions: Cells growing in the log phase were collected for Baf/3 cells and detached with Gibco™ TrypLE™ Express Enzyme for A431, PC-9, 11-18 and H1568 and plated in 96 well plate at 1500 to 3000 cells/well in 100 μL of media. After overnight incubation at 37°C, 5% CO2 incubator, the cells were treated with a test compound (50 μL/well) at a final concentration spanning from 0.000025 μM to 5 μM. They were incubated at 37°C, 5% CO2, and 95% humidity for 96 hours. At the end of incubation, a 2D CTG reagent from Promega was added to each well according to the vendor’s recommendation and mixed for 10 min in dark. Luminescence signals were determined with a Biotek® SynergyTM H1 plate reader (Agilent Technologies Inc., Santa Clara, California). The data were fitted using a sigmoidal curve analysis program (GraphPad PrismTM, Dotmatics Inc., San Diego, California) with variable hill slope and IC50 was determined. The results showed that, unexpectedly, (i) Compounds 1, 3, 4, 6-12, 15-19, 20, 21, 27- 29, 32, and 34 each have an IC50 of no more than 10 nM for inhibiting BaF3/LR/CS cells, (ii) Compounds 1, 3, 4, 6-12, 15-19, 20, 21, 27-29, and 32 each have an IC50 of no more than 10 nM for inhibiting BaF3/del19/CS cells, (iii) Compounds 3, 4, 6, 7, 9, 10, and 15-17 each have an IC50 of no more than 10 nM for inhibiting BaF3 CS cells, and Compounds 8, 11, 18, 20 and 21 each have an IC50 of 10 nM to 50 nM for inhibiting BaF3 CS cells, (iv) Compounds 3, 4, 6, 7, 15-17, 21, 27-29, and 32 each have an IC50 of no more than 10 nM for inhibiting PC-9 cells,
and Compounds 1, 10, 12, 19, 20, and 34 each have an IC50 of 10 nM to 50 nM for inhibiting PC-9 cells, and (v) Compound 32 has an IC50 of 10 nM to 50 nM for inhibiting A431 cells. Brain penetration assay conditions: Brain to plasma ratio was determined with 5 in 1 cassette dosing or 4 in 1 cassette dosing or 3 in 1 cassette dosing or 2 in 1 cassette dosing or discrete dosing using non-tumor bearing Sprague Dawley (SD) rats. For each 5 in 1 cassette dosing or 4 in 1 cassette dosing or 3 in 1 cassette dosing or 2 in 1 cassette dosing or discrete dosing studies a total of 3 male SD rats were selected from the animal pool of SD rats. The compounds were dissolved in a mixture of 5% EtOH/85%PE 400/10% H2O solution and the animals were dosed orally at 2 mg/kg dose with a concentration of 0.2 mg/mL. The plasma and brain samples were collected 2h after dosing. For plasma samples, 0.3 mL blood was collected from each animal into K2EDTA tubes kept on ice. The samples were centrifuged within 30 minutes of collection under refrigeration at 5 °C for 5 minutes at 6000 rpm to obtain plasma samples. A plasma sample (20 μL) was added to 2 μL MeOH and 200 μL of 5 ng/mL Terfenadine in methanol/acetonitrile (1:1, v/v). It was agitated for 1 minute and centrifuged at 4000 rpm for 15 minutes. A supernatant was separated and diluted with 5 volumes of MeOH/H2O (1;1, v/v, 0.1% FA) for analysis below. For brain samples, after a rat was euthanized, its brain was removed and weighed. Water (4x) was added to homogenize the brain tissue in a high-speed homogenizer. To a 50 μL of brain homogenate was added 5 µL MeOH and 200 μL of 5 ng/mL Terfenadine in Methanol/ Acetonitrile (1:1, v/v). After vortex for 1 minute and centrifugation at 4000 rpm for 15 min, a supernatant was separated and diluted with 3 volumes of MeOH/H2O (1:1, v/v, 0.1% FA) for analysis below. All samples were analysed via LC/MS/MS on AB Sciex 5500 using Kintex 1.7μ C18 100A column (50 x 2.1mm). Brain to plasma ratios (Concentrationbrain/Concentrationplasma) of each test compound was calculated. The results showed that, surprisingly, Compounds 1-8, 10, 12, 15, 17-19, 25, 26, 31-33, and 39 each have a brain to plasma ratio of 0.5 or greater. The results demonstrated that compounds of this invention effectively penetrate the blood-brain barrier. Pharmacokinetic Studies in Balb/c mice: The pharmacokinetic (PK) properties of compounds of the invention were obtained following a single intravenous (IV) bolus (e.g., 1 mg/kg) and a single oral gavage (PO) (e.g., 10 mg/kg) administration to female Balb/c mice (fed, ages within 5-7 weeks). For the IV studies (5 ml/kg), the vehicle was 5% DMA/5% EtOH/40% PEG 400/50% H2O (solution) and the blood samples were collected at 0 (pre-dose),
0.083, 0.25, 1, 4, 8, 12, 24, 48, 72, 96, and 120 hours post-dose. For the PO (10 ml/kg) studies, the vehicle was 1% HEC/0.25% Tween-80/0.05% Antifoam/H2O (suspension) and the blood samples were collected at 0 (pre-dose), 0.25, 1, 4, 8, 12, 24, 48, 72, 96, and 120 hours post- dose for PO. The plasma samples were prepared by centrifugation at 4ºC and 3200xg for 10 minutes, and then quickly frozen over dry ice and kept at -60ºC or lower until analysis. The concentrations of the test article and its potential metabolites (if detectable) in the plasma samples were determined by a validated liquid chromatography-tandem mass spectrometry (LC-MS/MS). The pharmacokinetic parameters (e.g., t1/2, C0, Cmax, Tmax, CL, AUC, etc.) were estimated by a non-compartmental analysis (NCA) using WinNonlin® (Certara, Radnor, Pennsylvania). The results showed that the compounds as tested unexpectedly exhibited great oral bioavailability (>50%) and prolonged t1/2, e.g., greater than 10 or 20 hours. KC-2253-Ba/F3 EGFR Del19-C797S-luc2 situ brain Model: The cell line used was Ba/F3 EGFR Del19-C797S-luc2 (Kyinno, Cat. Number: cKC-2253) engineered by introducing human EGFR containing Del19 and C797 mutations and luciferase reporter gene into Ba/F3 cells. Ba/F3 EGFR Del19-C797S-luc2 cells were maintained and expanded in RPMI-1640 containing 10% fetal bovine serum at 37ºC, 5% CO2 incubator. The cells growing in an exponential growth phase were harvested and counted for tumor inoculation. BALB/c nude mice (female, 6-8 weeks, weighing approximately 18-22g) from Biocytogen (Beijing, China) were used. After one week of acclimation, animals were anesthetized by ip injection of Pentobarbital sodium (30 mg/kg). The anesthetized mice were prepared with an injection point in the skull located on the right parietal bone, 3-3.5 mm lateral to the sagittal suture, and 0.5 mm posterior to the coronal suture. Each mouse was inoculated intracranially with the Ba/F3 EGFR Del19-C797S-luc2 (5 × 104 cells in 2 μL PBS) via a microinjector. The needle was inserted to a depth of 4 mm, then retracted by 0.7 mm. After the injection, the mouse was kept warm until they recovered from the anesthesia. Animals were observed post-anesthesia until all animals recovered. For pain relief, the animals were dosed with meloxicam (5 mg/kg, once daily for 3 days) after the mouse recovered from the anesthesia. On day 7 post-tumor inoculation, the inoculated mice were weighed and were pre-anesthetized with 1.25% tribromoethanol solution (20 µl/g). When the animals were in a complete anesthetic state, intraperitoneally injected D-Luciferin solution at a concentration of 15 mg/mL and a volume of 0.2 mL. All mice were measured for bioluminescence at 9 minutes after the luciferin administration and intensity of
bioluminescence for each mouse was recorded. Mice bearing established brain tumors based on bioluminescence were selected and randomized into 7 groups according to bioluminescence intensity and body weight. Mice were dosed once daily for 31 consecutive days with indicated test articles in suspension formulation with vehicle (1% HEC (w/v)/0.25% Twen-80 (v/v)/0.05% Antifoam (v/v)/H2O) or vehicle control by oral gavage. During dosing, mouse body weight was measured three times a week and tumor growth based on bioluminescence intensity was measured on day 3, 7, 10, 14, 17 and 31. After dosing stopped at day 32, mice were kept alive to observe extended survival until day 51 which study was terminated. The results showed that a compound of the invention exhibited excellent brain penetration with improved antitumor activity and survival. All the animals in the treatment groups, dosed in the range of 0.6 to 6 mg/kg/day, survived for more than 44 days, while all the animals in the controlled group died before D15 post-treatment. Intracranial PC-9_Luc human lung cancer xenograft model: PC-9_Luc (EGFR/del19 mutation) human lung cancer cells derived from RCB4455 (RIKEN BioResource Center, Tsukuba, Japan) was used for the study. The cells were maintained in vitro as a monolayer culture in RPMI 1640 medium supplemented with 10% fetal bovine serum and 1% Antibiotic- Antimycotic at 37 °C in an atmosphere of 5% CO2 in air. The cells were subcultured twice a week by trypsin-EDTA treatment. The cells growing in an exponential growth phase were harvested and counted for tumor inoculation. BALB/c nude mice (female, 6-8 weeks, weighing approximately 18-22g) were purchased from Zhejiang Vital River Laboratory Animal Co., LTD. and used for the study. After one week of acclimation, animals were anesthetized by ip injection of 1.25% avertin solution with dosing volume of 20 μL/g. The anesthetized mice were properly positioned. The head skin of the mouse was sterilized with 70% alcohol and was draped in a sterile fashion. A ~1cm length incision was made just at the right of the midline and anterior to the interaural line. Each mouse was inoculated intracranially with the PC-9_Luc cells (3 ×105 cells in 3 μL PBS+20% Matrigel) at a depth of 3.5 mm via the microinjector. It was kept warm and observed post-anesthesia until recovered. For pain relief from surgery and tumor inoculation, the mouse was dosed orally with 10 mg/kg of meloxicam (10 μL/g ). All mice were treated again with 5 mg/kg meloxicam (10 μL/g) in the morning of the next day post-surgery. To monitor tumor growth and mouse status, the body weight was obtained. Bioluminescence measurements were performed by intraperitoneal injection of luciferin at 150 mg/kg. After 10 minutes of the luciferin administration, the mice were anesthetized before bioluminescence determination with IVIS (Lumina II). The bioluminescence of the whole
animal body, including metastatic tumors, was determined and recorded. On day 7 post-tumor inoculation, mice were randomized into 7 groups based on the intensity of bioluminescence measured and body weight and dosed daily for 28 consecutive days with indicated test articles in suspension formulation with vehicle (1% HEC (w/v)/0.25% Twen-80 (v/v)/0.05% Antifoam (v/v)/H2O) or vehicle control by oral gavage. During dosing, mouse body weight was measured twice a week and tumor growth based on bioluminescence intensity was measured once a week. After dosing stopped on day 28, mice were observed for extended survival. The results showed that a compound of the invention showed very potent anti-tumor activity with improved survival. All the animals in the treatment groups, dosed in the range of 0.6 to 6 mg/kg/day, survived for more than 60 days, while all the animals in the controlled group died before D24 post-treatment. OTHER EMBODIMENTS All of the features disclosed in this specification may be combined in any combination. Each feature disclosed in this specification may be replaced by an alternative feature serving the same, equivalent, or similar purpose. Thus, unless expressly stated otherwise, each feature disclosed is only an example of a generic series of equivalent or similar features. From the above description, one skilled in the art can easily ascertain the essential characteristics of the present invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions. For example, compounds structurally analogous to the compounds of this invention also can be made, screened for their efficacy in treating an inflammatory condition. Thus, other embodiments are also within the claims.
Claims
WHAT IS CLAIMED IS: 1. A compound of Formula I:
I, or a pharmaceutically acceptable salt or stereoisomers or racemates thereof, wherein: one of X and Y is H, deuterium, or halo; the other of X and Y is
, in which each of R1 is F, C1-C6 alkyl, C3-C6 cycloalkyl, or 4-6 membered heterocyclyl, R2 is H, deuterium, F, C1-C6 alkyl, C3-C6 cycloalkyl, or 4-6 membered heterocyclyl, or R1 and R2, together with the carbon atom they bond to, are C3-C6 cycloalkyl or 4-6 membered heterocyclyl; R3 is 5-10 membered heteroaryl or C6-C10 aryl; and n is 1, 2, or 3; R4 is H, halo, cyano, C3-C6 cycloalkyl, ORa, 4-10 membered heterocyclyl, or 5-6 membered heteroaryl, in which Ra is C1-C6 alkyl, C3-C6 cycloalkyl, 4-10 membered heterocyclyl, or 5-6 membered heteroaryl; R5 is H or deuterium; R6 is H, C1-C6 alkyl, or C3-C6 cycloalkyl; R7 is 6-10 membered aryl or 5-10 membered heteroaryl; R8 is H, deuterium, halo, C1-C6 alkyl, or C3-C6 cycloalkyl; alkyl is optionally substituted with one or more groups selected from the group consisting of deuterium, halo, cyano, C1-C4 alkoxy, NR1aR1b, C3-C6 cycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, and 4-6 membered heterocyclyl, in which R1a and R1b, independently, is H, deuterium, C1-C4 alkyl, or C3-C6 cycloalkyl, or R1a and R1b, together with the nitrogen atom they bond to, are C4-C6 heterocyclyl; and each of cycloalkyl, heteroccyclyl, aryl, and heteroaryl, independently, is optionally substituted with one or more groups selected from the group consisting of deuterium, halo, cyano, acetylenyl, C1-C4 alkyl, C1-C4 alkoxy, C3-C6 cycloalkyl, C3-C6 cycloalkoxy, 4-6 membered heterocyclyl, and NR1aR1b.
2. The compound of claim 1, wherein (i) n is 1, and (ii) R1 is F, C1-C6 alkyl or C3-C6 cycloalkyl, preferably methyl, CF3, CHF2, cyclopropyl, or CD3; or R1 and R2, together with the carbon atom they bond to, are C3-C6 cycloalkyl. 3. The compound of claim 1 or 2, wherein R2 is H, F, C1-C3 alkyl or C3-C6 cycloalkyl, preferably methyl, CF3, CHF2, cyclopropyl, or CD3; or R1 and R2, together with the carbon atom they bond to, are C3-C6 cycloalkyl. 4. The compound of any one of the preceding claims, wherein R3 is imidazolyl, pyrazolyl, triazolyl, phenyl, pyrazinyl, pyridazinyl, pyridinyl, or pyrimidinyl. 5. The compound of any one of claims 1 to 3, wherein R3 is
. 7. The compound of any one of the preceding claims, wherein R4 is 5- or 6-membered heteroaryl, preferably imidazolyl, pyrazolyl, thiazolyl, or pyrimidinyl, optionally substituted with CD3, fluoro, methyl, trifluoromethyl or difluoromethyl.
. 9. The compound of any one of the preceding claims, wherein R7 is phenyl optionally substituted with one or more groups selected from the group consisting of F, Cl, Br and acetylenyl; preferably, R7 is
. 10. The compound of any one of claims 1 to 8, wherein R7 is 9- or 10- membered bicyclic heteroaryl optionally substituted with halo, preferably quinolinyl, indolizinyl, pyrazolo[1,5-a]pyridinyl, imidazo[1,2-a]pyridinyl, benzo[c]isothiazolyl, or benzo[d]isothiazolyl, more preferably, R7 is
11. The compound of any one of the preceding claims, wherein R6 is H, CH3, CD3, or CHF2. 12. The compound of any one of the preceding claims, wherein R8 is H or deuterium. 13. The compound of any one of the preceding claims, wherein n is 1. 14. The compound of any one of the preceding claims, wherein the compound is of Formula IA:
IA. 15. The compound of claim 14, wherein R4 is
. 16. The compound of any one of claims 1 to 13, wherein the compound is of Formula IB:
IB. 17. The compound of claim 16, wherein R4 is
. 18. The compound of claim 14, wherein n is 1; R5 is H or deuterium; R1 is F, C1-C3 alkyl or C3-C6 cycloalkyl, preferably methyl, CF3, CHF2, cyclopropyl, or CD3, and R2 is H, F, C1-C3 alkyl, preferably methyl, CF3, CHF2, cyclopropyl, or CD3; or R1 and R2, together with the carbon atom they are bonded to, is cyclopropyl; R3 is
,
19. The compound of claim 16, wherein n is 1; R5 is H or deuterium; R1 is F, C1-C3 alkyl or C3-C6 cycloalkyl, preferably methyl, CF3, CHF2, cyclopropyl, or CD3, and R2 is H, F, C1-C3 alkyl, preferably methyl, CF3, CHF2, cyclopropyl, or CD3; or R1 and R2, together with the carbon atom they are bonded to, is cyclopropyl; R3 is
R4 is
. 20. A compound that is one of Compounds 1-65. 21. A pharmaceutical composition comprising a compound of any one of claims 1 to 20 and a pharmaceutically acceptable carrier. 22. A method of treating cancer comprising administering to a subject in need thereof an effective amount of a compound of any one of claims 1 to 20, or a pharmaceutical composition of claim 21. 23. The method of claim 22, wherein the cancer is a lung cancer or a brain cancer. 24. A method of inhibiting EGFR comprising administering to a subject in need thereof an effective amount of a compound of any one of claims 1 to 20, or a pharmaceutical composition of claim 21.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202363470633P | 2023-06-02 | 2023-06-02 | |
US63/470,633 | 2023-06-02 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024249642A1 true WO2024249642A1 (en) | 2024-12-05 |
Family
ID=93658505
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2024/031685 WO2024249642A1 (en) | 2023-06-02 | 2024-05-30 | Egfr inhibitors |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024249642A1 (en) |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20210353627A1 (en) * | 2018-09-21 | 2021-11-18 | Spectrum Pharmaceuticals, Inc. | Novel quinazoline egfr inhibitors |
US20210386742A1 (en) * | 2018-09-18 | 2021-12-16 | Suzhou Zanrong Pharma Limited | Quinazoline derivatives as antitumor agents |
US20220324862A1 (en) * | 2021-03-31 | 2022-10-13 | Acerand Therapeutics (Usa) Limited | Pyridopyrimidinone compounds |
-
2024
- 2024-05-30 WO PCT/US2024/031685 patent/WO2024249642A1/en unknown
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20210386742A1 (en) * | 2018-09-18 | 2021-12-16 | Suzhou Zanrong Pharma Limited | Quinazoline derivatives as antitumor agents |
US20210353627A1 (en) * | 2018-09-21 | 2021-11-18 | Spectrum Pharmaceuticals, Inc. | Novel quinazoline egfr inhibitors |
US20220324862A1 (en) * | 2021-03-31 | 2022-10-13 | Acerand Therapeutics (Usa) Limited | Pyridopyrimidinone compounds |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP6457623B2 (en) | 2,4-disubstituted 7H-pyrrolo [2,3-d] pyrimidine derivatives, process for their preparation and use in medicine | |
CN117242079A (en) | KRAS G12D inhibitors and their uses | |
JP3457164B2 (en) | Aminoquinazoline derivative | |
AU2015222584B2 (en) | 2,4-disubstituted phenylene-1,5-diamine derivatives and applications thereof, and pharmaceutical compositions and pharmaceutically acceptable compositions prepared therefrom | |
CN113061132B (en) | Condensed ring lactam compound, preparation method and application | |
ES2617678T3 (en) | Compounds that act as kinase inhibitors | |
CN117157292A (en) | KRAS G12D inhibitors and uses thereof | |
TW201336835A (en) | Pharmaceutical use of 4-quinazolone amide derivatives | |
TWI814234B (en) | Preparation and Application of Mutant Protein Inhibitors | |
KR20170001694A (en) | New phosphate compounds, a process for their preparation and pharmaceutical compositions containing them | |
JP6342069B2 (en) | Azabicyclo derivatives, their production process and use in medicine | |
KR102426138B1 (en) | FUSED QUINOLINE COMPOUNDS AS PI3K, mTOR INHIBITORS | |
WO2018064852A1 (en) | Naphthyridine compound, pharmaceutical composition and use thereof | |
CN117222646A (en) | KRAS G12C inhibitors and uses thereof | |
CN116143806A (en) | Nitrogen-containing heterocyclic compound, preparation method and application | |
WO2022271630A1 (en) | Egfr inhibitors | |
CN115073450A (en) | KRAS G12C Preparation and application of mutant protein inhibitor | |
JP2024534804A (en) | SOS1 inhibitors and their uses | |
TW201315733A (en) | Imidazo quinoline derivatives and salts thereof, preparation process and pharmaceutical use thereof | |
AU2019331934A1 (en) | Multi-substituted pyridone derivatives and medical use thereof | |
CN116143805A (en) | A class of nitrogen-containing heterocyclic biaryl compounds, preparation method and use | |
CN117800976A (en) | Nitrogen-containing heterocyclic compound, preparation method and application | |
CN115836064A (en) | Triazine derivative with EGFR (epidermal growth factor receptor) inhibitory activity and preparation method and application thereof | |
WO2024249642A1 (en) | Egfr inhibitors | |
CA3090876C (en) | Dioxinoquinoline compounds, preparation method and uses thereof |
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: 24816425 Country of ref document: EP Kind code of ref document: A1 |