CN118829640A - Heterocyclic compounds for inducing G12D mutant KRAS protein degradation - Google Patents
Heterocyclic compounds for inducing G12D mutant KRAS protein degradation Download PDFInfo
- Publication number
- CN118829640A CN118829640A CN202380025718.3A CN202380025718A CN118829640A CN 118829640 A CN118829640 A CN 118829640A CN 202380025718 A CN202380025718 A CN 202380025718A CN 118829640 A CN118829640 A CN 118829640A
- Authority
- CN
- China
- Prior art keywords
- formula
- group
- substituted
- compound
- alkyl
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 102200006539 rs121913529 Human genes 0.000 title abstract description 81
- 102100030708 GTPase KRas Human genes 0.000 title abstract description 78
- 101000584612 Homo sapiens GTPase KRas Proteins 0.000 title abstract description 78
- 150000002391 heterocyclic compounds Chemical class 0.000 title abstract description 11
- 230000001939 inductive effect Effects 0.000 title description 8
- 230000017854 proteolysis Effects 0.000 title description 6
- 150000001875 compounds Chemical class 0.000 claims abstract description 576
- 150000003839 salts Chemical class 0.000 claims abstract description 188
- 206010061902 Pancreatic neoplasm Diseases 0.000 claims abstract description 94
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 claims abstract description 93
- 201000002528 pancreatic cancer Diseases 0.000 claims abstract description 93
- 208000008443 pancreatic carcinoma Diseases 0.000 claims abstract description 93
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 40
- -1 R 7 is H Chemical group 0.000 claims description 300
- 125000000217 alkyl group Chemical group 0.000 claims description 204
- 238000004519 manufacturing process Methods 0.000 claims description 202
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical group N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 claims description 169
- 229910052799 carbon Inorganic materials 0.000 claims description 134
- 229910052739 hydrogen Inorganic materials 0.000 claims description 132
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 123
- 229910052757 nitrogen Chemical group 0.000 claims description 87
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 claims description 86
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 claims description 83
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical group [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 claims description 75
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical group [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 claims description 75
- 229910052760 oxygen Inorganic materials 0.000 claims description 75
- 239000001301 oxygen Substances 0.000 claims description 75
- 125000005842 heteroatom Chemical group 0.000 claims description 74
- 229910052717 sulfur Inorganic materials 0.000 claims description 70
- 239000011593 sulfur Chemical group 0.000 claims description 69
- 125000000623 heterocyclic group Chemical group 0.000 claims description 67
- 229910052731 fluorine Inorganic materials 0.000 claims description 65
- 125000002947 alkylene group Chemical group 0.000 claims description 62
- 125000000843 phenylene group Chemical group C1(=C(C=CC=C1)*)* 0.000 claims description 59
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 claims description 58
- 229920006395 saturated elastomer Polymers 0.000 claims description 55
- 125000004429 atom Chemical group 0.000 claims description 50
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 claims description 49
- 125000005913 (C3-C6) cycloalkyl group Chemical group 0.000 claims description 39
- 125000001559 cyclopropyl group Chemical group [H]C1([H])C([H])([H])C1([H])* 0.000 claims description 38
- 239000000470 constituent Substances 0.000 claims description 37
- 229910052736 halogen Inorganic materials 0.000 claims description 36
- 150000002367 halogens Chemical class 0.000 claims description 36
- 239000000460 chlorine Substances 0.000 claims description 34
- 125000002393 azetidinyl group Chemical group 0.000 claims description 31
- RWRDLPDLKQPQOW-UHFFFAOYSA-N Pyrrolidine Chemical group C1CCNC1 RWRDLPDLKQPQOW-UHFFFAOYSA-N 0.000 claims description 30
- 125000004433 nitrogen atom Chemical group N* 0.000 claims description 30
- 150000001721 carbon Chemical group 0.000 claims description 26
- 238000000034 method Methods 0.000 claims description 25
- 125000004193 piperazinyl group Chemical group 0.000 claims description 23
- 125000003718 tetrahydrofuranyl group Chemical group 0.000 claims description 22
- 125000006163 5-membered heteroaryl group Chemical group 0.000 claims description 21
- 229910052801 chlorine Inorganic materials 0.000 claims description 21
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 claims description 20
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 claims description 20
- 125000001412 tetrahydropyranyl group Chemical group 0.000 claims description 20
- 125000000719 pyrrolidinyl group Chemical group 0.000 claims description 19
- 125000003386 piperidinyl group Chemical group 0.000 claims description 17
- 125000006164 6-membered heteroaryl group Chemical group 0.000 claims description 15
- 125000004432 carbon atom Chemical group C* 0.000 claims description 15
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 15
- 125000003226 pyrazolyl group Chemical group 0.000 claims description 15
- 125000000391 vinyl group Chemical group [H]C([*])=C([H])[H] 0.000 claims description 15
- 229920002554 vinyl polymer Polymers 0.000 claims description 15
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical group [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 claims description 14
- 125000004076 pyridyl group Chemical group 0.000 claims description 11
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 10
- 125000000714 pyrimidinyl group Chemical group 0.000 claims description 10
- 125000001624 naphthyl group Chemical group 0.000 claims description 9
- 150000000022 5-membered heteroarenes Chemical class 0.000 claims description 8
- 150000000025 6-membered heteroarenes Chemical class 0.000 claims description 8
- 125000003277 amino group Chemical group 0.000 claims description 8
- 125000004169 (C1-C6) alkyl group Chemical group 0.000 claims description 7
- 125000002887 hydroxy group Chemical group [H]O* 0.000 claims description 7
- JNCMHMUGTWEVOZ-UHFFFAOYSA-N F[CH]F Chemical compound F[CH]F JNCMHMUGTWEVOZ-UHFFFAOYSA-N 0.000 claims description 6
- 239000005864 Sulphur Chemical group 0.000 claims description 6
- 125000003682 3-furyl group Chemical group O1C([H])=C([*])C([H])=C1[H] 0.000 claims description 4
- 229910052698 phosphorus Inorganic materials 0.000 claims 1
- 239000003814 drug Substances 0.000 abstract description 10
- 239000004480 active ingredient Substances 0.000 abstract description 8
- 230000002797 proteolythic effect Effects 0.000 abstract description 8
- 229940124597 therapeutic agent Drugs 0.000 abstract description 6
- 230000002401 inhibitory effect Effects 0.000 abstract description 5
- 239000012830 cancer therapeutic Substances 0.000 abstract 1
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 252
- 238000006243 chemical reaction Methods 0.000 description 182
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 130
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 106
- 239000000243 solution Substances 0.000 description 102
- 239000002994 raw material Substances 0.000 description 96
- 239000000203 mixture Substances 0.000 description 93
- 238000003786 synthesis reaction Methods 0.000 description 82
- 230000015572 biosynthetic process Effects 0.000 description 76
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 75
- VLKZOEOYAKHREP-UHFFFAOYSA-N n-Hexane Chemical compound CCCCCC VLKZOEOYAKHREP-UHFFFAOYSA-N 0.000 description 72
- 239000007787 solid Substances 0.000 description 70
- 230000002829 reductive effect Effects 0.000 description 63
- 238000001816 cooling Methods 0.000 description 59
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 58
- 239000002904 solvent Substances 0.000 description 56
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 53
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-Dimethylformamide Chemical compound CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 53
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 49
- 238000010898 silica gel chromatography Methods 0.000 description 49
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 38
- 239000012044 organic layer Substances 0.000 description 37
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-Diisopropylethylamine (DIPEA) Chemical compound CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 0.000 description 34
- 125000006239 protecting group Chemical group 0.000 description 34
- 238000012360 testing method Methods 0.000 description 33
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 31
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 30
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 30
- 239000011541 reaction mixture Substances 0.000 description 29
- 239000011780 sodium chloride Substances 0.000 description 29
- 238000010511 deprotection reaction Methods 0.000 description 28
- 238000006467 substitution reaction Methods 0.000 description 28
- 206010028980 Neoplasm Diseases 0.000 description 27
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 27
- KDLHZDBZIXYQEI-UHFFFAOYSA-N Palladium Chemical compound [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 description 25
- 239000000706 filtrate Substances 0.000 description 25
- 238000010438 heat treatment Methods 0.000 description 25
- SJRJJKPEHAURKC-UHFFFAOYSA-N N-Methylmorpholine Chemical compound CN1CCOCC1 SJRJJKPEHAURKC-UHFFFAOYSA-N 0.000 description 24
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 24
- 238000011065 in-situ storage Methods 0.000 description 24
- 238000003756 stirring Methods 0.000 description 24
- 239000006228 supernatant Substances 0.000 description 24
- 125000004546 quinazolin-4-yl group Chemical group N1=CN=C(C2=CC=CC=C12)* 0.000 description 23
- 239000000741 silica gel Substances 0.000 description 23
- 229910002027 silica gel Inorganic materials 0.000 description 23
- 210000004027 cell Anatomy 0.000 description 21
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 20
- 238000001914 filtration Methods 0.000 description 19
- 239000010410 layer Substances 0.000 description 18
- BWHMMNNQKKPAPP-UHFFFAOYSA-L potassium carbonate Chemical compound [K+].[K+].[O-]C([O-])=O BWHMMNNQKKPAPP-UHFFFAOYSA-L 0.000 description 18
- 238000010992 reflux Methods 0.000 description 18
- CSNNHWWHGAXBCP-UHFFFAOYSA-L Magnesium sulfate Chemical compound [Mg+2].[O-][S+2]([O-])([O-])[O-] CSNNHWWHGAXBCP-UHFFFAOYSA-L 0.000 description 17
- 238000006161 Suzuki-Miyaura coupling reaction Methods 0.000 description 16
- LPQZERIRKRYGGM-UHFFFAOYSA-N tert-butyl pyrrolidine-1-carboxylate Chemical compound CC(C)(C)OC(=O)N1CCCC1 LPQZERIRKRYGGM-UHFFFAOYSA-N 0.000 description 16
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 15
- 239000012300 argon atmosphere Substances 0.000 description 15
- FJDQFPXHSGXQBY-UHFFFAOYSA-L caesium carbonate Chemical compound [Cs+].[Cs+].[O-]C([O-])=O FJDQFPXHSGXQBY-UHFFFAOYSA-L 0.000 description 15
- 229910000024 caesium carbonate Inorganic materials 0.000 description 15
- 239000003054 catalyst Substances 0.000 description 15
- 150000002170 ethers Chemical class 0.000 description 15
- 230000005764 inhibitory process Effects 0.000 description 15
- NLXLAEXVIDQMFP-UHFFFAOYSA-N Ammonia chloride Chemical compound [NH4+].[Cl-] NLXLAEXVIDQMFP-UHFFFAOYSA-N 0.000 description 14
- CDBYLPFSWZWCQE-UHFFFAOYSA-L Sodium Carbonate Chemical compound [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 14
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 14
- 239000003795 chemical substances by application Substances 0.000 description 14
- 150000008282 halocarbons Chemical class 0.000 description 14
- 230000035772 mutation Effects 0.000 description 14
- 239000000126 substance Substances 0.000 description 14
- 125000006273 (C1-C3) alkyl group Chemical group 0.000 description 13
- DKGAVHZHDRPRBM-UHFFFAOYSA-N Tert-Butanol Chemical compound CC(C)(C)O DKGAVHZHDRPRBM-UHFFFAOYSA-N 0.000 description 13
- 125000004184 methoxymethyl group Chemical group [H]C([H])([H])OC([H])([H])* 0.000 description 13
- 239000002953 phosphate buffered saline Substances 0.000 description 13
- 125000001424 substituent group Chemical group 0.000 description 13
- 238000005406 washing Methods 0.000 description 13
- UHOVQNZJYSORNB-UHFFFAOYSA-N Benzene Chemical compound C1=CC=CC=C1 UHOVQNZJYSORNB-UHFFFAOYSA-N 0.000 description 12
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 12
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 12
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 12
- 150000004945 aromatic hydrocarbons Chemical class 0.000 description 12
- 125000005619 boric acid group Chemical group 0.000 description 12
- 108090000623 proteins and genes Proteins 0.000 description 12
- NYCVCXMSZNOGDH-UHFFFAOYSA-N pyrrolidine-1-carboxylic acid Chemical compound OC(=O)N1CCCC1 NYCVCXMSZNOGDH-UHFFFAOYSA-N 0.000 description 12
- XTHFKEDIFFGKHM-UHFFFAOYSA-N Dimethoxyethane Chemical compound COCCOC XTHFKEDIFFGKHM-UHFFFAOYSA-N 0.000 description 11
- 239000012298 atmosphere Substances 0.000 description 11
- 239000001257 hydrogen Substances 0.000 description 11
- 125000004029 hydroxymethyl group Chemical group [H]OC([H])([H])* 0.000 description 11
- 239000003921 oil Substances 0.000 description 11
- 125000003566 oxetanyl group Chemical group 0.000 description 11
- 102000004169 proteins and genes Human genes 0.000 description 11
- XKRFYHLGVUSROY-UHFFFAOYSA-N Argon Chemical group [Ar] XKRFYHLGVUSROY-UHFFFAOYSA-N 0.000 description 10
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 10
- 238000000605 extraction Methods 0.000 description 10
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 9
- 239000002253 acid Substances 0.000 description 9
- 150000001298 alcohols Chemical class 0.000 description 9
- 238000007112 amidation reaction Methods 0.000 description 9
- 239000007864 aqueous solution Substances 0.000 description 9
- 201000011510 cancer Diseases 0.000 description 9
- 239000003153 chemical reaction reagent Substances 0.000 description 9
- 230000001394 metastastic effect Effects 0.000 description 9
- 206010061289 metastatic neoplasm Diseases 0.000 description 9
- 201000008129 pancreatic ductal adenocarcinoma Diseases 0.000 description 9
- 229910000027 potassium carbonate Inorganic materials 0.000 description 9
- LPNYRYFBWFDTMA-UHFFFAOYSA-N potassium tert-butoxide Chemical compound [K+].CC(C)(C)[O-] LPNYRYFBWFDTMA-UHFFFAOYSA-N 0.000 description 9
- 102000016914 ras Proteins Human genes 0.000 description 9
- 230000000306 recurrent effect Effects 0.000 description 9
- PPASLZSBLFJQEF-RKJRWTFHSA-M sodium ascorbate Substances [Na+].OC[C@@H](O)[C@H]1OC(=O)C(O)=C1[O-] PPASLZSBLFJQEF-RKJRWTFHSA-M 0.000 description 9
- 235000010378 sodium ascorbate Nutrition 0.000 description 9
- 229960005055 sodium ascorbate Drugs 0.000 description 9
- PPASLZSBLFJQEF-RXSVEWSESA-M sodium-L-ascorbate Chemical compound [Na+].OC[C@H](O)[C@H]1OC(=O)C(O)=C1[O-] PPASLZSBLFJQEF-RXSVEWSESA-M 0.000 description 9
- ZGTMUACCHSMWAC-UHFFFAOYSA-L EDTA disodium salt (anhydrous) Chemical compound [Na+].[Na+].OC(=O)CN(CC([O-])=O)CCN(CC(O)=O)CC([O-])=O ZGTMUACCHSMWAC-UHFFFAOYSA-L 0.000 description 8
- XKMLYUALXHKNFT-UUOKFMHZSA-N Guanosine-5'-triphosphate Chemical compound C1=2NC(N)=NC(=O)C=2N=CN1[C@@H]1O[C@H](COP(O)(=O)OP(O)(=O)OP(O)(O)=O)[C@@H](O)[C@H]1O XKMLYUALXHKNFT-UUOKFMHZSA-N 0.000 description 8
- 102000006275 Ubiquitin-Protein Ligases Human genes 0.000 description 8
- 108010083111 Ubiquitin-Protein Ligases Proteins 0.000 description 8
- 230000001588 bifunctional effect Effects 0.000 description 8
- 238000004440 column chromatography Methods 0.000 description 8
- IXCSERBJSXMMFS-UHFFFAOYSA-N hydrogen chloride Substances Cl.Cl IXCSERBJSXMMFS-UHFFFAOYSA-N 0.000 description 8
- 229910000041 hydrogen chloride Inorganic materials 0.000 description 8
- 239000012299 nitrogen atmosphere Substances 0.000 description 8
- 150000007530 organic bases Chemical class 0.000 description 8
- 239000000843 powder Substances 0.000 description 8
- 235000018102 proteins Nutrition 0.000 description 8
- JWVCLYRUEFBMGU-UHFFFAOYSA-N quinazoline Chemical compound N1=CN=CC2=CC=CC=C21 JWVCLYRUEFBMGU-UHFFFAOYSA-N 0.000 description 8
- 239000000725 suspension Substances 0.000 description 8
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 7
- 239000007821 HATU Substances 0.000 description 7
- FXHOOIRPVKKKFG-UHFFFAOYSA-N N,N-Dimethylacetamide Chemical compound CN(C)C(C)=O FXHOOIRPVKKKFG-UHFFFAOYSA-N 0.000 description 7
- 230000002378 acidificating effect Effects 0.000 description 7
- 235000019270 ammonium chloride Nutrition 0.000 description 7
- 230000000903 blocking effect Effects 0.000 description 7
- ARUVKPQLZAKDPS-UHFFFAOYSA-L copper(II) sulfate Chemical compound [Cu+2].[O-][S+2]([O-])([O-])[O-] ARUVKPQLZAKDPS-UHFFFAOYSA-L 0.000 description 7
- 229910000366 copper(II) sulfate Inorganic materials 0.000 description 7
- 238000000354 decomposition reaction Methods 0.000 description 7
- 230000000694 effects Effects 0.000 description 7
- 150000007529 inorganic bases Chemical class 0.000 description 7
- 125000004123 n-propyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])* 0.000 description 7
- 229910052763 palladium Inorganic materials 0.000 description 7
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 7
- 235000017557 sodium bicarbonate Nutrition 0.000 description 7
- 229910000029 sodium carbonate Inorganic materials 0.000 description 7
- 235000017550 sodium carbonate Nutrition 0.000 description 7
- 239000007858 starting material Substances 0.000 description 7
- OKUCEQDKBKYEJY-QMMMGPOBSA-N tert-butyl (3s)-3-(methylamino)pyrrolidine-1-carboxylate Chemical compound CN[C@H]1CCN(C(=O)OC(C)(C)C)C1 OKUCEQDKBKYEJY-QMMMGPOBSA-N 0.000 description 7
- 125000005931 tert-butyloxycarbonyl group Chemical group [H]C([H])([H])C(OC(*)=O)(C([H])([H])[H])C([H])([H])[H] 0.000 description 7
- 125000002221 trityl group Chemical group [H]C1=C([H])C([H])=C([H])C([H])=C1C([*])(C1=C(C(=C(C(=C1[H])[H])[H])[H])[H])C1=C([H])C([H])=C([H])C([H])=C1[H] 0.000 description 7
- WSLDOOZREJYCGB-UHFFFAOYSA-N 1,2-Dichloroethane Chemical compound ClCCCl WSLDOOZREJYCGB-UHFFFAOYSA-N 0.000 description 6
- OISVCGZHLKNMSJ-UHFFFAOYSA-N 2,6-dimethylpyridine Chemical compound CC1=CC=CC(C)=N1 OISVCGZHLKNMSJ-UHFFFAOYSA-N 0.000 description 6
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 6
- GSNUFIFRDBKVIE-UHFFFAOYSA-N DMF Natural products CC1=CC=C(C)O1 GSNUFIFRDBKVIE-UHFFFAOYSA-N 0.000 description 6
- 229940124785 KRAS inhibitor Drugs 0.000 description 6
- SECXISVLQFMRJM-UHFFFAOYSA-N N-Methylpyrrolidone Chemical compound CN1CCCC1=O SECXISVLQFMRJM-UHFFFAOYSA-N 0.000 description 6
- 101150040459 RAS gene Proteins 0.000 description 6
- 235000001014 amino acid Nutrition 0.000 description 6
- 150000001413 amino acids Chemical class 0.000 description 6
- KGBXLFKZBHKPEV-UHFFFAOYSA-N boric acid Chemical class OB(O)O KGBXLFKZBHKPEV-UHFFFAOYSA-N 0.000 description 6
- 238000009903 catalytic hydrogenation reaction Methods 0.000 description 6
- 239000003638 chemical reducing agent Substances 0.000 description 6
- MXFYYFVVIIWKFE-UHFFFAOYSA-N dicyclohexyl-[2-[2,6-di(propan-2-yloxy)phenyl]phenyl]phosphane Chemical group CC(C)OC1=CC=CC(OC(C)C)=C1C1=CC=CC=C1P(C1CCCCC1)C1CCCCC1 MXFYYFVVIIWKFE-UHFFFAOYSA-N 0.000 description 6
- 125000006001 difluoroethyl group Chemical group 0.000 description 6
- 229910052740 iodine Inorganic materials 0.000 description 6
- 239000003446 ligand Substances 0.000 description 6
- KJIFKLIQANRMOU-UHFFFAOYSA-N oxidanium;4-methylbenzenesulfonate Chemical compound O.CC1=CC=C(S(O)(=O)=O)C=C1 KJIFKLIQANRMOU-UHFFFAOYSA-N 0.000 description 6
- YJVFFLUZDVXJQI-UHFFFAOYSA-L palladium(ii) acetate Chemical compound [Pd+2].CC([O-])=O.CC([O-])=O YJVFFLUZDVXJQI-UHFFFAOYSA-L 0.000 description 6
- XHXFXVLFKHQFAL-UHFFFAOYSA-N phosphoryl trichloride Chemical compound ClP(Cl)(Cl)=O XHXFXVLFKHQFAL-UHFFFAOYSA-N 0.000 description 6
- 238000002360 preparation method Methods 0.000 description 6
- 230000008569 process Effects 0.000 description 6
- 125000000335 thiazolyl group Chemical group 0.000 description 6
- FYSNRJHAOHDILO-UHFFFAOYSA-N thionyl chloride Chemical compound ClS(Cl)=O FYSNRJHAOHDILO-UHFFFAOYSA-N 0.000 description 6
- 125000001425 triazolyl group Chemical group 0.000 description 6
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 description 6
- LWIHDJKSTIGBAC-UHFFFAOYSA-K tripotassium phosphate Chemical compound [K+].[K+].[K+].[O-]P([O-])([O-])=O LWIHDJKSTIGBAC-UHFFFAOYSA-K 0.000 description 6
- QGWNDRXFNXRZMB-UUOKFMHZSA-N GDP Chemical compound C1=2NC(N)=NC(=O)C=2N=CN1[C@@H]1O[C@H](COP(O)(=O)OP(O)(O)=O)[C@@H](O)[C@H]1O QGWNDRXFNXRZMB-UUOKFMHZSA-N 0.000 description 5
- CTQNGGLPUBDAKN-UHFFFAOYSA-N O-Xylene Chemical compound CC1=CC=CC=C1C CTQNGGLPUBDAKN-UHFFFAOYSA-N 0.000 description 5
- KWYUFKZDYYNOTN-UHFFFAOYSA-M Potassium hydroxide Chemical compound [OH-].[K+] KWYUFKZDYYNOTN-UHFFFAOYSA-M 0.000 description 5
- 229940024606 amino acid Drugs 0.000 description 5
- 229910052786 argon Inorganic materials 0.000 description 5
- 238000005859 coupling reaction Methods 0.000 description 5
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 5
- 229940079593 drug Drugs 0.000 description 5
- 125000006125 ethylsulfonyl group Chemical group 0.000 description 5
- 238000011156 evaluation Methods 0.000 description 5
- QGWNDRXFNXRZMB-UHFFFAOYSA-N guanidine diphosphate Natural products C1=2NC(N)=NC(=O)C=2N=CN1C1OC(COP(O)(=O)OP(O)(O)=O)C(O)C1O QGWNDRXFNXRZMB-UHFFFAOYSA-N 0.000 description 5
- 125000001072 heteroaryl group Chemical group 0.000 description 5
- 239000005457 ice water Substances 0.000 description 5
- 125000002883 imidazolyl group Chemical group 0.000 description 5
- 239000000411 inducer Substances 0.000 description 5
- 239000007788 liquid Substances 0.000 description 5
- 125000002757 morpholinyl group Chemical group 0.000 description 5
- 125000002971 oxazolyl group Chemical group 0.000 description 5
- LXNAVEXFUKBNMK-UHFFFAOYSA-N palladium(II) acetate Substances [Pd].CC(O)=O.CC(O)=O LXNAVEXFUKBNMK-UHFFFAOYSA-N 0.000 description 5
- 239000006187 pill Substances 0.000 description 5
- 230000001225 therapeutic effect Effects 0.000 description 5
- 239000008096 xylene Substances 0.000 description 5
- MEKOFIRRDATTAG-UHFFFAOYSA-N 2,2,5,8-tetramethyl-3,4-dihydrochromen-6-ol Chemical compound C1CC(C)(C)OC2=C1C(C)=C(O)C=C2C MEKOFIRRDATTAG-UHFFFAOYSA-N 0.000 description 4
- VHYFNPMBLIVWCW-UHFFFAOYSA-N 4-Dimethylaminopyridine Chemical compound CN(C)C1=CC=NC=C1 VHYFNPMBLIVWCW-UHFFFAOYSA-N 0.000 description 4
- 102000007469 Actins Human genes 0.000 description 4
- 108010085238 Actins Proteins 0.000 description 4
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 4
- 102000001301 EGF receptor Human genes 0.000 description 4
- 108060006698 EGF receptor Proteins 0.000 description 4
- 101150105104 Kras gene Proteins 0.000 description 4
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 4
- PXHVJJICTQNCMI-UHFFFAOYSA-N Nickel Chemical compound [Ni] PXHVJJICTQNCMI-UHFFFAOYSA-N 0.000 description 4
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 4
- SMWDFEZZVXVKRB-UHFFFAOYSA-N Quinoline Chemical compound N1=CC=CC2=CC=CC=C21 SMWDFEZZVXVKRB-UHFFFAOYSA-N 0.000 description 4
- 239000012980 RPMI-1640 medium Substances 0.000 description 4
- 102000005588 Son of Sevenless Proteins Human genes 0.000 description 4
- 108010059447 Son of Sevenless Proteins Proteins 0.000 description 4
- DTQVDTLACAAQTR-UHFFFAOYSA-N Trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 4
- 235000010724 Wisteria floribunda Nutrition 0.000 description 4
- RQPZNWPYLFFXCP-UHFFFAOYSA-L barium dihydroxide Chemical compound [OH-].[OH-].[Ba+2] RQPZNWPYLFFXCP-UHFFFAOYSA-L 0.000 description 4
- 229910001863 barium hydroxide Inorganic materials 0.000 description 4
- YNHIGQDRGKUECZ-UHFFFAOYSA-L bis(triphenylphosphine)palladium(ii) dichloride Chemical compound [Cl-].[Cl-].[Pd+2].C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 YNHIGQDRGKUECZ-UHFFFAOYSA-L 0.000 description 4
- PFKFTWBEEFSNDU-UHFFFAOYSA-N carbonyldiimidazole Chemical compound C1=CN=CN1C(=O)N1C=CN=C1 PFKFTWBEEFSNDU-UHFFFAOYSA-N 0.000 description 4
- 150000001879 copper Chemical class 0.000 description 4
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 4
- 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 4
- 238000005695 dehalogenation reaction Methods 0.000 description 4
- 125000001028 difluoromethyl group Chemical group [H]C(F)(F)* 0.000 description 4
- 125000002147 dimethylamino group Chemical group [H]C([H])([H])N(*)C([H])([H])[H] 0.000 description 4
- DNJIEGIFACGWOD-UHFFFAOYSA-N ethanethiol Chemical compound CCS DNJIEGIFACGWOD-UHFFFAOYSA-N 0.000 description 4
- 239000012091 fetal bovine serum Substances 0.000 description 4
- 125000000524 functional group Chemical group 0.000 description 4
- LEQAOMBKQFMDFZ-UHFFFAOYSA-N glyoxal Chemical compound O=CC=O LEQAOMBKQFMDFZ-UHFFFAOYSA-N 0.000 description 4
- 230000003301 hydrolyzing effect Effects 0.000 description 4
- BDAGIHXWWSANSR-UHFFFAOYSA-N methanoic acid Natural products OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 4
- NFHFRUOZVGFOOS-UHFFFAOYSA-N palladium;triphenylphosphane Chemical compound [Pd].C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 NFHFRUOZVGFOOS-UHFFFAOYSA-N 0.000 description 4
- 230000026731 phosphorylation Effects 0.000 description 4
- 238000006366 phosphorylation reaction Methods 0.000 description 4
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 4
- SCVFZCLFOSHCOH-UHFFFAOYSA-M potassium acetate Chemical compound [K+].CC([O-])=O SCVFZCLFOSHCOH-UHFFFAOYSA-M 0.000 description 4
- 108010014186 ras Proteins Proteins 0.000 description 4
- 238000000611 regression analysis Methods 0.000 description 4
- JOXIMZWYDAKGHI-UHFFFAOYSA-N toluene-4-sulfonic acid Chemical compound CC1=CC=C(S(O)(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-N 0.000 description 4
- BODOJNQCVLBBQH-NSHDSACASA-N (2R)-2-amino-2-[4-(4-methyl-1,3-thiazol-5-yl)phenyl]ethanol Chemical compound N=1C(=C(SC=1)C1=CC=C([C@@H](N)CO)C=C1)C BODOJNQCVLBBQH-NSHDSACASA-N 0.000 description 3
- 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 3
- WAPNOHKVXSQRPX-UHFFFAOYSA-N 1-phenylethanol Chemical compound CC(O)C1=CC=CC=C1 WAPNOHKVXSQRPX-UHFFFAOYSA-N 0.000 description 3
- NHQDETIJWKXCTC-UHFFFAOYSA-N 3-chloroperbenzoic acid Chemical compound OOC(=O)C1=CC=CC(Cl)=C1 NHQDETIJWKXCTC-UHFFFAOYSA-N 0.000 description 3
- 125000004575 3-pyrrolidinyl group Chemical group [H]N1C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 3
- MSHFRERJPWKJFX-UHFFFAOYSA-N 4-Methoxybenzyl alcohol Chemical compound COC1=CC=C(CO)C=C1 MSHFRERJPWKJFX-UHFFFAOYSA-N 0.000 description 3
- QCQCHGYLTSGIGX-GHXANHINSA-N 4-[[(3ar,5ar,5br,7ar,9s,11ar,11br,13as)-5a,5b,8,8,11a-pentamethyl-3a-[(5-methylpyridine-3-carbonyl)amino]-2-oxo-1-propan-2-yl-4,5,6,7,7a,9,10,11,11b,12,13,13a-dodecahydro-3h-cyclopenta[a]chrysen-9-yl]oxy]-2,2-dimethyl-4-oxobutanoic acid Chemical compound N([C@@]12CC[C@@]3(C)[C@]4(C)CC[C@H]5C(C)(C)[C@@H](OC(=O)CC(C)(C)C(O)=O)CC[C@]5(C)[C@H]4CC[C@@H]3C1=C(C(C2)=O)C(C)C)C(=O)C1=CN=CC(C)=C1 QCQCHGYLTSGIGX-GHXANHINSA-N 0.000 description 3
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- 108020004705 Codon Proteins 0.000 description 3
- QOSSAOTZNIDXMA-UHFFFAOYSA-N Dicylcohexylcarbodiimide Chemical compound C1CCCCC1N=C=NC1CCCCC1 QOSSAOTZNIDXMA-UHFFFAOYSA-N 0.000 description 3
- 102000013446 GTP Phosphohydrolases Human genes 0.000 description 3
- 108091006109 GTPases Proteins 0.000 description 3
- OFOBLEOULBTSOW-UHFFFAOYSA-N Malonic acid Chemical compound OC(=O)CC(O)=O OFOBLEOULBTSOW-UHFFFAOYSA-N 0.000 description 3
- MUBZPKHOEPUJKR-UHFFFAOYSA-N Oxalic acid Chemical compound OC(=O)C(O)=O MUBZPKHOEPUJKR-UHFFFAOYSA-N 0.000 description 3
- 208000008900 Pancreatic Ductal Carcinoma Diseases 0.000 description 3
- 101150003085 Pdcl gene Proteins 0.000 description 3
- 229920001213 Polysorbate 20 Polymers 0.000 description 3
- 102000001788 Proto-Oncogene Proteins c-raf Human genes 0.000 description 3
- 108010029869 Proto-Oncogene Proteins c-raf Proteins 0.000 description 3
- KEAYESYHFKHZAL-UHFFFAOYSA-N Sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 3
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 3
- 239000000654 additive Substances 0.000 description 3
- XPNGNIFUDRPBFJ-UHFFFAOYSA-N alpha-methylbenzylalcohol Natural products CC1=CC=CC=C1CO XPNGNIFUDRPBFJ-UHFFFAOYSA-N 0.000 description 3
- 230000000259 anti-tumor effect Effects 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- 229910002091 carbon monoxide Inorganic materials 0.000 description 3
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 3
- 238000004113 cell culture Methods 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 230000009918 complex formation Effects 0.000 description 3
- 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 3
- 239000002274 desiccant Substances 0.000 description 3
- 125000000816 ethylene group Chemical group [H]C([H])([*:1])C([H])([H])[*:2] 0.000 description 3
- 239000012737 fresh medium Substances 0.000 description 3
- 239000007789 gas Substances 0.000 description 3
- 125000003187 heptyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 3
- 230000007062 hydrolysis Effects 0.000 description 3
- 238000006460 hydrolysis reaction Methods 0.000 description 3
- 230000001965 increasing effect Effects 0.000 description 3
- 238000002347 injection Methods 0.000 description 3
- 239000007924 injection Substances 0.000 description 3
- 239000002198 insoluble material Substances 0.000 description 3
- 239000000543 intermediate Substances 0.000 description 3
- 229910052751 metal Inorganic materials 0.000 description 3
- 239000002184 metal Substances 0.000 description 3
- 229940098779 methanesulfonic acid Drugs 0.000 description 3
- 125000001570 methylene group Chemical group [H]C([H])([*:1])[*:2] 0.000 description 3
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 3
- 239000013642 negative control Substances 0.000 description 3
- 238000007254 oxidation reaction Methods 0.000 description 3
- WVDDGKGOMKODPV-ZQBYOMGUSA-N phenyl(114C)methanol Chemical compound O[14CH2]C1=CC=CC=C1 WVDDGKGOMKODPV-ZQBYOMGUSA-N 0.000 description 3
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 3
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 3
- 229910000160 potassium phosphate Inorganic materials 0.000 description 3
- 235000011009 potassium phosphates Nutrition 0.000 description 3
- 239000003755 preservative agent Substances 0.000 description 3
- 229940002612 prodrug Drugs 0.000 description 3
- 239000000651 prodrug Substances 0.000 description 3
- 230000000750 progressive effect Effects 0.000 description 3
- 229940124823 proteolysis targeting chimeric molecule Drugs 0.000 description 3
- 238000000746 purification Methods 0.000 description 3
- 125000002914 sec-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 3
- 239000012312 sodium hydride Substances 0.000 description 3
- 229910000104 sodium hydride Inorganic materials 0.000 description 3
- 239000008247 solid mixture Substances 0.000 description 3
- 239000003381 stabilizer Substances 0.000 description 3
- 238000001356 surgical procedure Methods 0.000 description 3
- 238000010189 synthetic method Methods 0.000 description 3
- YTTFFPATQICAQN-BYPYZUCNSA-N (2s)-2-methoxypropan-1-ol Chemical compound CO[C@@H](C)CO YTTFFPATQICAQN-BYPYZUCNSA-N 0.000 description 2
- SZXBQTSZISFIAO-ZETCQYMHSA-N (2s)-3-methyl-2-[(2-methylpropan-2-yl)oxycarbonylamino]butanoic acid Chemical compound CC(C)[C@@H](C(O)=O)NC(=O)OC(C)(C)C SZXBQTSZISFIAO-ZETCQYMHSA-N 0.000 description 2
- BENKAPCDIOILGV-RQJHMYQMSA-N (2s,4r)-4-hydroxy-1-[(2-methylpropan-2-yl)oxycarbonyl]pyrrolidine-2-carboxylic acid Chemical compound CC(C)(C)OC(=O)N1C[C@H](O)C[C@H]1C(O)=O BENKAPCDIOILGV-RQJHMYQMSA-N 0.000 description 2
- QVCUKHQDEZNNOC-UHFFFAOYSA-N 1,2-diazabicyclo[2.2.2]octane Chemical compound C1CC2CCN1NC2 QVCUKHQDEZNNOC-UHFFFAOYSA-N 0.000 description 2
- CYSGHNMQYZDMIA-UHFFFAOYSA-N 1,3-Dimethyl-2-imidazolidinon Chemical compound CN1CCN(C)C1=O CYSGHNMQYZDMIA-UHFFFAOYSA-N 0.000 description 2
- ASOKPJOREAFHNY-UHFFFAOYSA-N 1-Hydroxybenzotriazole Chemical compound C1=CC=C2N(O)N=NC2=C1 ASOKPJOREAFHNY-UHFFFAOYSA-N 0.000 description 2
- HZAXFHJVJLSVMW-UHFFFAOYSA-N 2-Aminoethan-1-ol Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 description 2
- WRRUOUDEMJQAPB-UHFFFAOYSA-N 2-azido-1,3-dimethyl-2H-imidazole Chemical compound CN1C(N(C=C1)C)N=[N+]=[N-] WRRUOUDEMJQAPB-UHFFFAOYSA-N 0.000 description 2
- XWKFPIODWVPXLX-UHFFFAOYSA-N 2-methyl-5-methylpyridine Natural products CC1=CC=C(C)N=C1 XWKFPIODWVPXLX-UHFFFAOYSA-N 0.000 description 2
- TWBPWBPGNQWFSJ-UHFFFAOYSA-N 2-phenylaniline Chemical group NC1=CC=CC=C1C1=CC=CC=C1 TWBPWBPGNQWFSJ-UHFFFAOYSA-N 0.000 description 2
- BMYNFMYTOJXKLE-UHFFFAOYSA-N 3-azaniumyl-2-hydroxypropanoate Chemical compound NCC(O)C(O)=O BMYNFMYTOJXKLE-UHFFFAOYSA-N 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
- QMHIMXFNBOYPND-UHFFFAOYSA-N 4-methylthiazole Chemical compound CC1=CSC=N1 QMHIMXFNBOYPND-UHFFFAOYSA-N 0.000 description 2
- YYROPELSRYBVMQ-UHFFFAOYSA-N 4-toluenesulfonyl chloride Chemical compound CC1=CC=C(S(Cl)(=O)=O)C=C1 YYROPELSRYBVMQ-UHFFFAOYSA-N 0.000 description 2
- IEHLGMIULROOSJ-UHFFFAOYSA-N 7-bromo-2,4-dichloro-8-fluoro-6-iodoquinazoline Chemical compound BrC1=C(C=C2C(=NC(=NC2=C1F)Cl)Cl)I IEHLGMIULROOSJ-UHFFFAOYSA-N 0.000 description 2
- 125000005330 8 membered heterocyclic group Chemical group 0.000 description 2
- KUVBUQLOKUUMFT-LBPRGKRZSA-N CCN1N=CC=C1C1=CC=C([C@H](CO)N)C=C1 Chemical compound CCN1N=CC=C1C1=CC=C([C@H](CO)N)C=C1 KUVBUQLOKUUMFT-LBPRGKRZSA-N 0.000 description 2
- 102000015367 CRBN Human genes 0.000 description 2
- GAWIXWVDTYZWAW-UHFFFAOYSA-N C[CH]O Chemical group C[CH]O GAWIXWVDTYZWAW-UHFFFAOYSA-N 0.000 description 2
- 241000283707 Capra Species 0.000 description 2
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 2
- VMQMZMRVKUZKQL-UHFFFAOYSA-N Cu+ Chemical compound [Cu+] VMQMZMRVKUZKQL-UHFFFAOYSA-N 0.000 description 2
- 239000003109 Disodium ethylene diamine tetraacetate Substances 0.000 description 2
- SNRUBQQJIBEYMU-UHFFFAOYSA-N Dodecane Natural products CCCCCCCCCCCC SNRUBQQJIBEYMU-UHFFFAOYSA-N 0.000 description 2
- 238000002965 ELISA Methods 0.000 description 2
- QUSNBJAOOMFDIB-UHFFFAOYSA-N Ethylamine Chemical compound CCN QUSNBJAOOMFDIB-UHFFFAOYSA-N 0.000 description 2
- VZCYOOQTPOCHFL-OWOJBTEDSA-N Fumaric acid Chemical compound OC(=O)\C=C\C(O)=O VZCYOOQTPOCHFL-OWOJBTEDSA-N 0.000 description 2
- 102000018898 GTPase-Activating Proteins Human genes 0.000 description 2
- 108091006094 GTPase-accelerating proteins Proteins 0.000 description 2
- 206010064571 Gene mutation Diseases 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 101000941994 Homo sapiens Protein cereblon Proteins 0.000 description 2
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 2
- 206010069755 K-ras gene mutation Diseases 0.000 description 2
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- BAVYZALUXZFZLV-UHFFFAOYSA-N Methylamine Chemical compound NC BAVYZALUXZFZLV-UHFFFAOYSA-N 0.000 description 2
- 241000699666 Mus <mouse, genus> Species 0.000 description 2
- 241000699670 Mus sp. Species 0.000 description 2
- LRHPLDYGYMQRHN-UHFFFAOYSA-N N-Butanol Chemical compound CCCCO LRHPLDYGYMQRHN-UHFFFAOYSA-N 0.000 description 2
- MZRVEZGGRBJDDB-UHFFFAOYSA-N N-Butyllithium Chemical compound [Li]CCCC MZRVEZGGRBJDDB-UHFFFAOYSA-N 0.000 description 2
- AMQJEAYHLZJPGS-UHFFFAOYSA-N N-Pentanol Chemical compound CCCCCO AMQJEAYHLZJPGS-UHFFFAOYSA-N 0.000 description 2
- PCLIMKBDDGJMGD-UHFFFAOYSA-N N-bromosuccinimide Chemical compound BrN1C(=O)CCC1=O PCLIMKBDDGJMGD-UHFFFAOYSA-N 0.000 description 2
- JAUOIFJMECXRGI-UHFFFAOYSA-N Neoclaritin Chemical compound C=1C(Cl)=CC=C2C=1CCC1=CC=CN=C1C2=C1CCNCC1 JAUOIFJMECXRGI-UHFFFAOYSA-N 0.000 description 2
- RKPABCXUYPTBTF-UHFFFAOYSA-N OC(C1=CC(I)=C2Br)=NC(Cl)=NC1=C2F Chemical compound OC(C1=CC(I)=C2Br)=NC(Cl)=NC1=C2F RKPABCXUYPTBTF-UHFFFAOYSA-N 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- KFSLWBXXFJQRDL-UHFFFAOYSA-N Peracetic acid Chemical compound CC(=O)OO KFSLWBXXFJQRDL-UHFFFAOYSA-N 0.000 description 2
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 2
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical compound C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 description 2
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 2
- 239000013504 Triton X-100 Substances 0.000 description 2
- 229920004890 Triton X-100 Polymers 0.000 description 2
- 238000002441 X-ray diffraction Methods 0.000 description 2
- 230000000996 additive effect Effects 0.000 description 2
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 2
- 238000005576 amination reaction Methods 0.000 description 2
- 235000003704 aspartic acid Nutrition 0.000 description 2
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 2
- 229910052794 bromium Inorganic materials 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 238000005266 casting Methods 0.000 description 2
- 230000015556 catabolic process Effects 0.000 description 2
- 230000004663 cell proliferation Effects 0.000 description 2
- 239000012320 chlorinating reagent Substances 0.000 description 2
- 238000002648 combination therapy Methods 0.000 description 2
- 238000013329 compounding Methods 0.000 description 2
- 230000008094 contradictory effect Effects 0.000 description 2
- 238000007796 conventional method Methods 0.000 description 2
- 230000008878 coupling Effects 0.000 description 2
- 238000010168 coupling process Methods 0.000 description 2
- 238000006352 cycloaddition reaction Methods 0.000 description 2
- 230000018044 dehydration Effects 0.000 description 2
- 238000006297 dehydration reaction Methods 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 239000012954 diazonium Substances 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-O diazynium Chemical compound [NH+]#N IJGRMHOSHXDMSA-UHFFFAOYSA-O 0.000 description 2
- WMKGGPCROCCUDY-PHEQNACWSA-N dibenzylideneacetone Chemical compound C=1C=CC=CC=1\C=C\C(=O)\C=C\C1=CC=CC=C1 WMKGGPCROCCUDY-PHEQNACWSA-N 0.000 description 2
- 125000006222 dimethylaminomethyl group Chemical group [H]C([H])([H])N(C([H])([H])[H])C([H])([H])* 0.000 description 2
- XBDQKXXYIPTUBI-UHFFFAOYSA-N dimethylselenoniopropionate Natural products CCC(O)=O XBDQKXXYIPTUBI-UHFFFAOYSA-N 0.000 description 2
- 235000019301 disodium ethylene diamine tetraacetate Nutrition 0.000 description 2
- 238000004090 dissolution Methods 0.000 description 2
- 125000003438 dodecyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 239000000839 emulsion Substances 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 239000006260 foam Substances 0.000 description 2
- 235000019253 formic acid Nutrition 0.000 description 2
- 238000001640 fractional crystallisation Methods 0.000 description 2
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 2
- 229960005277 gemcitabine Drugs 0.000 description 2
- 229940015043 glyoxal Drugs 0.000 description 2
- HHLFWLYXYJOTON-UHFFFAOYSA-N glyoxylic acid Chemical compound OC(=O)C=O HHLFWLYXYJOTON-UHFFFAOYSA-N 0.000 description 2
- 239000008187 granular material Substances 0.000 description 2
- 230000002140 halogenating effect Effects 0.000 description 2
- 150000004677 hydrates Chemical class 0.000 description 2
- TUJKJAMUKRIRHC-UHFFFAOYSA-N hydroxyl Chemical compound [OH] TUJKJAMUKRIRHC-UHFFFAOYSA-N 0.000 description 2
- 229960002591 hydroxyproline Drugs 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 239000003701 inert diluent Substances 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- 208000020082 intraepithelial neoplasia Diseases 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 125000000959 isobutyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])* 0.000 description 2
- 125000000842 isoxazolyl group Chemical group 0.000 description 2
- 238000002372 labelling Methods 0.000 description 2
- JVTAAEKCZFNVCJ-UHFFFAOYSA-N lactic acid Chemical compound CC(O)C(O)=O JVTAAEKCZFNVCJ-UHFFFAOYSA-N 0.000 description 2
- 125000005647 linker group Chemical group 0.000 description 2
- 239000000314 lubricant Substances 0.000 description 2
- NUJOXMJBOLGQSY-UHFFFAOYSA-N manganese dioxide Chemical compound O=[Mn]=O NUJOXMJBOLGQSY-UHFFFAOYSA-N 0.000 description 2
- 239000002609 medium Substances 0.000 description 2
- 239000012046 mixed solvent Substances 0.000 description 2
- 125000001280 n-hexyl group Chemical group C(CCCCC)* 0.000 description 2
- 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 2
- 125000002347 octyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 2
- 230000003287 optical effect Effects 0.000 description 2
- 239000007800 oxidant agent Substances 0.000 description 2
- MUJIDPITZJWBSW-UHFFFAOYSA-N palladium(2+) Chemical compound [Pd+2] MUJIDPITZJWBSW-UHFFFAOYSA-N 0.000 description 2
- 238000007911 parenteral administration Methods 0.000 description 2
- 229910052697 platinum Inorganic materials 0.000 description 2
- 239000013641 positive control Substances 0.000 description 2
- 239000011591 potassium Substances 0.000 description 2
- 229910052700 potassium Inorganic materials 0.000 description 2
- 235000011056 potassium acetate Nutrition 0.000 description 2
- 230000002335 preservative effect Effects 0.000 description 2
- 230000002265 prevention Effects 0.000 description 2
- 239000000047 product Substances 0.000 description 2
- 238000011865 proteolysis targeting chimera technique Methods 0.000 description 2
- 125000002098 pyridazinyl group Chemical group 0.000 description 2
- LNNRQIKKDKDFRV-UHFFFAOYSA-N quinazolin-8-ol Chemical compound N1=CN=C2C(O)=CC=CC2=C1 LNNRQIKKDKDFRV-UHFFFAOYSA-N 0.000 description 2
- 230000002285 radioactive effect Effects 0.000 description 2
- 102200006538 rs121913530 Human genes 0.000 description 2
- 108010026668 snake venom protein C activator Proteins 0.000 description 2
- JQWHASGSAFIOCM-UHFFFAOYSA-M sodium periodate Chemical compound [Na+].[O-]I(=O)(=O)=O JQWHASGSAFIOCM-UHFFFAOYSA-M 0.000 description 2
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical class O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 2
- 239000012453 solvate Substances 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 239000003826 tablet Substances 0.000 description 2
- UXAWXZDXVOYLII-UHFFFAOYSA-N tert-butyl 2,5-diazabicyclo[2.2.1]heptane-2-carboxylate Chemical compound C1C2N(C(=O)OC(C)(C)C)CC1NC2 UXAWXZDXVOYLII-UHFFFAOYSA-N 0.000 description 2
- FESDUDPSRMWIDL-UHFFFAOYSA-N tert-butyl n,n'-di(propan-2-yl)carbamimidate Chemical compound CC(C)NC(OC(C)(C)C)=NC(C)C FESDUDPSRMWIDL-UHFFFAOYSA-N 0.000 description 2
- FPGGTKZVZWFYPV-UHFFFAOYSA-M tetrabutylammonium fluoride Chemical compound [F-].CCCC[N+](CCCC)(CCCC)CCCC FPGGTKZVZWFYPV-UHFFFAOYSA-M 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 125000004568 thiomorpholinyl group Chemical group 0.000 description 2
- 238000002877 time resolved fluorescence resonance energy transfer Methods 0.000 description 2
- CFOAUYCPAUGDFF-UHFFFAOYSA-N tosmic Chemical class CC1=CC=C(S(=O)(=O)C[N+]#[C-])C=C1 CFOAUYCPAUGDFF-UHFFFAOYSA-N 0.000 description 2
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- PBIMIGNDTBRRPI-UHFFFAOYSA-N trifluoro borate Chemical group FOB(OF)OF PBIMIGNDTBRRPI-UHFFFAOYSA-N 0.000 description 2
- PYOKUURKVVELLB-UHFFFAOYSA-N trimethyl orthoformate Chemical compound COC(OC)OC PYOKUURKVVELLB-UHFFFAOYSA-N 0.000 description 2
- 239000000080 wetting agent Substances 0.000 description 2
- QBYIENPQHBMVBV-HFEGYEGKSA-N (2R)-2-hydroxy-2-phenylacetic acid Chemical compound O[C@@H](C(O)=O)c1ccccc1.O[C@@H](C(O)=O)c1ccccc1 QBYIENPQHBMVBV-HFEGYEGKSA-N 0.000 description 1
- SRQPEYLZIUEVIA-QMMMGPOBSA-N (2r)-2-amino-2-(4-fluorophenyl)ethanol Chemical compound OC[C@H](N)C1=CC=C(F)C=C1 SRQPEYLZIUEVIA-QMMMGPOBSA-N 0.000 description 1
- 125000004400 (C1-C12) alkyl group Chemical group 0.000 description 1
- WAPNOHKVXSQRPX-ZETCQYMHSA-N (S)-1-phenylethanol Chemical compound C[C@H](O)C1=CC=CC=C1 WAPNOHKVXSQRPX-ZETCQYMHSA-N 0.000 description 1
- BJEPYKJPYRNKOW-REOHCLBHSA-N (S)-malic acid Chemical compound OC(=O)[C@@H](O)CC(O)=O BJEPYKJPYRNKOW-REOHCLBHSA-N 0.000 description 1
- MOWXJLUYGFNTAL-DEOSSOPVSA-N (s)-[2-chloro-4-fluoro-5-(7-morpholin-4-ylquinazolin-4-yl)phenyl]-(6-methoxypyridazin-3-yl)methanol Chemical compound N1=NC(OC)=CC=C1[C@@H](O)C1=CC(C=2C3=CC=C(C=C3N=CN=2)N2CCOCC2)=C(F)C=C1Cl MOWXJLUYGFNTAL-DEOSSOPVSA-N 0.000 description 1
- HHBCEKAWSILOOP-UHFFFAOYSA-N 1,3-dibromo-1,3,5-triazinane-2,4,6-trione Chemical compound BrN1C(=O)NC(=O)N(Br)C1=O HHBCEKAWSILOOP-UHFFFAOYSA-N 0.000 description 1
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 1
- WKGZJBVXZWCZQC-UHFFFAOYSA-N 1-(1-benzyltriazol-4-yl)-n,n-bis[(1-benzyltriazol-4-yl)methyl]methanamine Chemical compound C=1N(CC=2C=CC=CC=2)N=NC=1CN(CC=1N=NN(CC=2C=CC=CC=2)C=1)CC(N=N1)=CN1CC1=CC=CC=C1 WKGZJBVXZWCZQC-UHFFFAOYSA-N 0.000 description 1
- MBIGNFNSALGEMI-UHFFFAOYSA-N 1-(chloromethyl)-4-ethynylbenzene Chemical compound ClCC1=CC=C(C#C)C=C1 MBIGNFNSALGEMI-UHFFFAOYSA-N 0.000 description 1
- LMDZBCPBFSXMTL-UHFFFAOYSA-N 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide Chemical compound CCN=C=NCCCN(C)C LMDZBCPBFSXMTL-UHFFFAOYSA-N 0.000 description 1
- 125000004066 1-hydroxyethyl group Chemical group [H]OC([H])([*])C([H])([H])[H] 0.000 description 1
- JQCSUVJDBHJKNG-UHFFFAOYSA-N 1-methoxy-ethyl Chemical group C[CH]OC JQCSUVJDBHJKNG-UHFFFAOYSA-N 0.000 description 1
- 102000010400 1-phosphatidylinositol-3-kinase activity proteins Human genes 0.000 description 1
- QMNUDYFKZYBWQX-UHFFFAOYSA-N 1H-quinazolin-4-one Chemical compound C1=CC=C2C(=O)N=CNC2=C1 QMNUDYFKZYBWQX-UHFFFAOYSA-N 0.000 description 1
- GQHTUMJGOHRCHB-UHFFFAOYSA-N 2,3,4,6,7,8,9,10-octahydropyrimido[1,2-a]azepine Chemical compound C1CCCCN2CCCN=C21 GQHTUMJGOHRCHB-UHFFFAOYSA-N 0.000 description 1
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 1
- KMGUEILFFWDGFV-UHFFFAOYSA-N 2-benzoyl-2-benzoyloxy-3-hydroxybutanedioic acid Chemical compound C=1C=CC=CC=1C(=O)C(C(C(O)=O)O)(C(O)=O)OC(=O)C1=CC=CC=C1 KMGUEILFFWDGFV-UHFFFAOYSA-N 0.000 description 1
- MARXMDRWROUXMD-UHFFFAOYSA-N 2-bromoisoindole-1,3-dione Chemical compound C1=CC=C2C(=O)N(Br)C(=O)C2=C1 MARXMDRWROUXMD-UHFFFAOYSA-N 0.000 description 1
- 125000005449 2-fluoro-1,4-phenylene group Chemical group [H]C1=C([*:2])C([H])=C(F)C([*:1])=C1[H] 0.000 description 1
- HFIXOSLXVQGUNQ-UHFFFAOYSA-N 2-formyl-2,3-dihydroxybutanedioic acid Chemical compound OC(=O)C(O)C(O)(C=O)C(O)=O HFIXOSLXVQGUNQ-UHFFFAOYSA-N 0.000 description 1
- ZZPNDIHOQDQVNU-UHFFFAOYSA-N 2-hydroxy-4,4,5,5-tetramethyl-1,3,2-dioxaborolane Chemical group CC1(C)OB(O)OC1(C)C ZZPNDIHOQDQVNU-UHFFFAOYSA-N 0.000 description 1
- 125000000954 2-hydroxyethyl group Chemical group [H]C([*])([H])C([H])([H])O[H] 0.000 description 1
- BMIBJCFFZPYJHF-UHFFFAOYSA-N 2-methoxy-5-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine Chemical group COC1=NC=C(C)C=C1B1OC(C)(C)C(C)(C)O1 BMIBJCFFZPYJHF-UHFFFAOYSA-N 0.000 description 1
- 125000004200 2-methoxyethyl group Chemical group [H]C([H])([H])OC([H])([H])C([H])([H])* 0.000 description 1
- YOETUEMZNOLGDB-UHFFFAOYSA-N 2-methylpropyl carbonochloridate Chemical compound CC(C)COC(Cl)=O YOETUEMZNOLGDB-UHFFFAOYSA-N 0.000 description 1
- FRIBMENBGGCKPD-UHFFFAOYSA-N 3-(2,3-dimethoxyphenyl)prop-2-enal Chemical compound COC1=CC=CC(C=CC=O)=C1OC FRIBMENBGGCKPD-UHFFFAOYSA-N 0.000 description 1
- HCDMJFOHIXMBOV-UHFFFAOYSA-N 3-(2,6-difluoro-3,5-dimethoxyphenyl)-1-ethyl-8-(morpholin-4-ylmethyl)-4,7-dihydropyrrolo[4,5]pyrido[1,2-d]pyrimidin-2-one Chemical compound C=1C2=C3N(CC)C(=O)N(C=4C(=C(OC)C=C(OC)C=4F)F)CC3=CN=C2NC=1CN1CCOCC1 HCDMJFOHIXMBOV-UHFFFAOYSA-N 0.000 description 1
- BYHQTRFJOGIQAO-GOSISDBHSA-N 3-(4-bromophenyl)-8-[(2R)-2-hydroxypropyl]-1-[(3-methoxyphenyl)methyl]-1,3,8-triazaspiro[4.5]decan-2-one Chemical compound C[C@H](CN1CCC2(CC1)CN(C(=O)N2CC3=CC(=CC=C3)OC)C4=CC=C(C=C4)Br)O BYHQTRFJOGIQAO-GOSISDBHSA-N 0.000 description 1
- YNJSNEKCXVFDKW-UHFFFAOYSA-N 3-(5-amino-1h-indol-3-yl)-2-azaniumylpropanoate Chemical compound C1=C(N)C=C2C(CC(N)C(O)=O)=CNC2=C1 YNJSNEKCXVFDKW-UHFFFAOYSA-N 0.000 description 1
- PBVAJRFEEOIAGW-UHFFFAOYSA-N 3-[bis(2-carboxyethyl)phosphanyl]propanoic acid;hydrochloride Chemical compound Cl.OC(=O)CCP(CCC(O)=O)CCC(O)=O PBVAJRFEEOIAGW-UHFFFAOYSA-N 0.000 description 1
- 125000005451 3-fluoro-1,4-phenylene group Chemical group [H]C1=C([*:1])C([H])=C(F)C([*:2])=C1[H] 0.000 description 1
- KVCQTKNUUQOELD-UHFFFAOYSA-N 4-amino-n-[1-(3-chloro-2-fluoroanilino)-6-methylisoquinolin-5-yl]thieno[3,2-d]pyrimidine-7-carboxamide Chemical compound N=1C=CC2=C(NC(=O)C=3C4=NC=NC(N)=C4SC=3)C(C)=CC=C2C=1NC1=CC=CC(Cl)=C1F KVCQTKNUUQOELD-UHFFFAOYSA-N 0.000 description 1
- ZEFMBAFMCSYJOO-UHFFFAOYSA-N 4-nitro-3-(trifluoromethyl)phenol Chemical compound OC1=CC=C([N+]([O-])=O)C(C(F)(F)F)=C1 ZEFMBAFMCSYJOO-UHFFFAOYSA-N 0.000 description 1
- KCBWAFJCKVKYHO-UHFFFAOYSA-N 6-(4-cyclopropyl-6-methoxypyrimidin-5-yl)-1-[[4-[1-propan-2-yl-4-(trifluoromethyl)imidazol-2-yl]phenyl]methyl]pyrazolo[3,4-d]pyrimidine Chemical compound C1(CC1)C1=NC=NC(=C1C1=NC=C2C(=N1)N(N=C2)CC1=CC=C(C=C1)C=1N(C=C(N=1)C(F)(F)F)C(C)C)OC KCBWAFJCKVKYHO-UHFFFAOYSA-N 0.000 description 1
- ZCYVEMRRCGMTRW-UHFFFAOYSA-N 7553-56-2 Chemical compound [I] ZCYVEMRRCGMTRW-UHFFFAOYSA-N 0.000 description 1
- 229940088872 Apoptosis inhibitor Drugs 0.000 description 1
- NAKJQIQGGWAPCK-UHFFFAOYSA-N C#CC(C=CC(CCl)=C1)=C1F Chemical compound C#CC(C=CC(CCl)=C1)=C1F NAKJQIQGGWAPCK-UHFFFAOYSA-N 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- OKTJSMMVPCPJKN-NJFSPNSNSA-N Carbon-14 Chemical compound [14C] OKTJSMMVPCPJKN-NJFSPNSNSA-N 0.000 description 1
- 102000014914 Carrier Proteins Human genes 0.000 description 1
- ZAMOUSCENKQFHK-UHFFFAOYSA-N Chlorine atom Chemical compound [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 description 1
- 229910021595 Copper(I) iodide Inorganic materials 0.000 description 1
- 102000009508 Cyclin-Dependent Kinase Inhibitor p16 Human genes 0.000 description 1
- 108010009392 Cyclin-Dependent Kinase Inhibitor p16 Proteins 0.000 description 1
- 229920000858 Cyclodextrin Polymers 0.000 description 1
- YZCKVEUIGOORGS-OUBTZVSYSA-N Deuterium Chemical compound [2H] YZCKVEUIGOORGS-OUBTZVSYSA-N 0.000 description 1
- FEWJPZIEWOKRBE-JCYAYHJZSA-N Dextrotartaric acid Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-JCYAYHJZSA-N 0.000 description 1
- 206010013710 Drug interaction Diseases 0.000 description 1
- 206010058314 Dysplasia Diseases 0.000 description 1
- 102000012199 E3 ubiquitin-protein ligase Mdm2 Human genes 0.000 description 1
- 108050002772 E3 ubiquitin-protein ligase Mdm2 Proteins 0.000 description 1
- 241000283074 Equus asinus Species 0.000 description 1
- 235000014755 Eruca sativa Nutrition 0.000 description 1
- 244000024675 Eruca sativa Species 0.000 description 1
- 239000001116 FEMA 4028 Substances 0.000 description 1
- KRHYYFGTRYWZRS-UHFFFAOYSA-M Fluoride anion Chemical compound [F-] KRHYYFGTRYWZRS-UHFFFAOYSA-M 0.000 description 1
- PXGOKWXKJXAPGV-UHFFFAOYSA-N Fluorine Chemical compound FF PXGOKWXKJXAPGV-UHFFFAOYSA-N 0.000 description 1
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 1
- 102100029974 GTPase HRas Human genes 0.000 description 1
- 102100039788 GTPase NRas Human genes 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Chemical compound OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 238000007341 Heck reaction Methods 0.000 description 1
- 101000584633 Homo sapiens GTPase HRas Proteins 0.000 description 1
- 101000744505 Homo sapiens GTPase NRas Proteins 0.000 description 1
- 101150117869 Hras gene Proteins 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- VQTUBCCKSQIDNK-UHFFFAOYSA-N Isobutene Chemical group CC(C)=C VQTUBCCKSQIDNK-UHFFFAOYSA-N 0.000 description 1
- AHLPHDHHMVZTML-BYPYZUCNSA-N L-Ornithine Chemical compound NCCC[C@H](N)C(O)=O AHLPHDHHMVZTML-BYPYZUCNSA-N 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 1
- 239000005551 L01XE03 - Erlotinib Substances 0.000 description 1
- 102000003960 Ligases Human genes 0.000 description 1
- 108090000364 Ligases Proteins 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 229940124647 MEK inhibitor Drugs 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 239000012359 Methanesulfonyl chloride Substances 0.000 description 1
- 102100025725 Mothers against decapentaplegic homolog 4 Human genes 0.000 description 1
- 101710143112 Mothers against decapentaplegic homolog 4 Proteins 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- KWYHDKDOAIKMQN-UHFFFAOYSA-N N,N,N',N'-tetramethylethylenediamine Chemical compound CN(C)CCN(C)C KWYHDKDOAIKMQN-UHFFFAOYSA-N 0.000 description 1
- AYCPARAPKDAOEN-LJQANCHMSA-N N-[(1S)-2-(dimethylamino)-1-phenylethyl]-6,6-dimethyl-3-[(2-methyl-4-thieno[3,2-d]pyrimidinyl)amino]-1,4-dihydropyrrolo[3,4-c]pyrazole-5-carboxamide Chemical compound C1([C@H](NC(=O)N2C(C=3NN=C(NC=4C=5SC=CC=5N=C(C)N=4)C=3C2)(C)C)CN(C)C)=CC=CC=C1 AYCPARAPKDAOEN-LJQANCHMSA-N 0.000 description 1
- WXNXCEHXYPACJF-ZETCQYMHSA-N N-acetyl-L-leucine Chemical compound CC(C)C[C@@H](C(O)=O)NC(C)=O WXNXCEHXYPACJF-ZETCQYMHSA-N 0.000 description 1
- PHSPJQZRQAJPPF-UHFFFAOYSA-N N-alpha-Methylhistamine Chemical compound CNCCC1=CN=CN1 PHSPJQZRQAJPPF-UHFFFAOYSA-N 0.000 description 1
- 101150073096 NRAS gene Proteins 0.000 description 1
- GRYLNZFGIOXLOG-UHFFFAOYSA-N Nitric acid Chemical compound O[N+]([O-])=O GRYLNZFGIOXLOG-UHFFFAOYSA-N 0.000 description 1
- 239000012425 OXONE® Substances 0.000 description 1
- 108700020796 Oncogene Proteins 0.000 description 1
- 102000043276 Oncogene Human genes 0.000 description 1
- AHLPHDHHMVZTML-UHFFFAOYSA-N Orn-delta-NH2 Natural products NCCCC(N)C(O)=O AHLPHDHHMVZTML-UHFFFAOYSA-N 0.000 description 1
- UTJLXEIPEHZYQJ-UHFFFAOYSA-N Ornithine Natural products OC(=O)C(C)CCCN UTJLXEIPEHZYQJ-UHFFFAOYSA-N 0.000 description 1
- 108091007960 PI3Ks Proteins 0.000 description 1
- 229930040373 Paraformaldehyde Natural products 0.000 description 1
- 102000004245 Proteasome Endopeptidase Complex Human genes 0.000 description 1
- 108090000708 Proteasome Endopeptidase Complex Proteins 0.000 description 1
- IWYDHOAUDWTVEP-UHFFFAOYSA-N R-2-phenyl-2-hydroxyacetic acid Natural products OC(=O)C(O)C1=CC=CC=C1 IWYDHOAUDWTVEP-UHFFFAOYSA-N 0.000 description 1
- 102100033479 RAF proto-oncogene serine/threonine-protein kinase Human genes 0.000 description 1
- 102000034442 RING-type E3 ubiquitin transferases Human genes 0.000 description 1
- 108030001238 RING-type E3 ubiquitin transferases Proteins 0.000 description 1
- 102100023320 Ral guanine nucleotide dissociation stimulator Human genes 0.000 description 1
- 101150015043 Ralgds gene Proteins 0.000 description 1
- 239000007868 Raney catalyst Substances 0.000 description 1
- 229910000564 Raney nickel Inorganic materials 0.000 description 1
- 229940078123 Ras inhibitor Drugs 0.000 description 1
- 101000744436 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) Trans-acting factor D Proteins 0.000 description 1
- UIIMBOGNXHQVGW-DEQYMQKBSA-M Sodium bicarbonate-14C Chemical compound [Na+].O[14C]([O-])=O UIIMBOGNXHQVGW-DEQYMQKBSA-M 0.000 description 1
- 239000005708 Sodium hypochlorite Substances 0.000 description 1
- KDYFGRWQOYBRFD-UHFFFAOYSA-N Succinic acid Natural products OC(=O)CCC(O)=O KDYFGRWQOYBRFD-UHFFFAOYSA-N 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-N Tartaric acid Natural products [H+].[H+].[O-]C(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-N 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
- 239000007983 Tris buffer Substances 0.000 description 1
- YZCKVEUIGOORGS-NJFSPNSNSA-N Tritium Chemical compound [3H] YZCKVEUIGOORGS-NJFSPNSNSA-N 0.000 description 1
- 238000006887 Ullmann reaction Methods 0.000 description 1
- LXRZVMYMQHNYJB-UNXOBOICSA-N [(1R,2S,4R)-4-[[5-[4-[(1R)-7-chloro-1,2,3,4-tetrahydroisoquinolin-1-yl]-5-methylthiophene-2-carbonyl]pyrimidin-4-yl]amino]-2-hydroxycyclopentyl]methyl sulfamate Chemical compound CC1=C(C=C(S1)C(=O)C1=C(N[C@H]2C[C@H](O)[C@@H](COS(N)(=O)=O)C2)N=CN=C1)[C@@H]1NCCC2=C1C=C(Cl)C=C2 LXRZVMYMQHNYJB-UNXOBOICSA-N 0.000 description 1
- BSYVTEYKTMYBMK-RXMQYKEDSA-N [(2r)-oxolan-2-yl]methanol Chemical compound OC[C@H]1CCCO1 BSYVTEYKTMYBMK-RXMQYKEDSA-N 0.000 description 1
- BSYVTEYKTMYBMK-YFKPBYRVSA-N [(2s)-oxolan-2-yl]methanol Chemical compound OC[C@@H]1CCCO1 BSYVTEYKTMYBMK-YFKPBYRVSA-N 0.000 description 1
- TUEIURIZJQRMQE-UHFFFAOYSA-N [2-(tert-butylsulfamoyl)phenyl]boronic acid Chemical compound CC(C)(C)NS(=O)(=O)C1=CC=CC=C1B(O)O TUEIURIZJQRMQE-UHFFFAOYSA-N 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 235000011054 acetic acid Nutrition 0.000 description 1
- 125000002777 acetyl group Chemical group [H]C([H])([H])C(*)=O 0.000 description 1
- 229960000669 acetylleucine Drugs 0.000 description 1
- 150000008065 acid anhydrides Chemical class 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 238000005917 acylation reaction Methods 0.000 description 1
- 230000001464 adherent effect Effects 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- BJEPYKJPYRNKOW-UHFFFAOYSA-N alpha-hydroxysuccinic acid Natural products OC(=O)C(O)CC(O)=O BJEPYKJPYRNKOW-UHFFFAOYSA-N 0.000 description 1
- 229910052782 aluminium Inorganic materials 0.000 description 1
- XAGFODPZIPBFFR-UHFFFAOYSA-N aluminium Chemical compound [Al] XAGFODPZIPBFFR-UHFFFAOYSA-N 0.000 description 1
- 230000009435 amidation Effects 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 150000003862 amino acid derivatives Chemical class 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- 150000003863 ammonium salts Chemical class 0.000 description 1
- 230000033115 angiogenesis Effects 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 239000000158 apoptosis inhibitor Substances 0.000 description 1
- 239000003125 aqueous solvent Substances 0.000 description 1
- 239000012131 assay buffer Substances 0.000 description 1
- 239000012752 auxiliary agent Substances 0.000 description 1
- 125000003943 azolyl group Chemical group 0.000 description 1
- 239000003899 bactericide agent Substances 0.000 description 1
- SRSXLGNVWSONIS-UHFFFAOYSA-N benzenesulfonic acid Chemical compound OS(=O)(=O)C1=CC=CC=C1 SRSXLGNVWSONIS-UHFFFAOYSA-N 0.000 description 1
- 229940092714 benzenesulfonic acid Drugs 0.000 description 1
- 125000003236 benzoyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C(*)=O 0.000 description 1
- 125000001797 benzyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])* 0.000 description 1
- 125000001584 benzyloxycarbonyl group Chemical group C(=O)(OCC1=CC=CC=C1)* 0.000 description 1
- 229960004853 betadex Drugs 0.000 description 1
- 108091008324 binding proteins Proteins 0.000 description 1
- 239000004305 biphenyl Substances 0.000 description 1
- 235000010290 biphenyl Nutrition 0.000 description 1
- 238000007664 blowing Methods 0.000 description 1
- 239000004327 boric acid Substances 0.000 description 1
- 125000001246 bromo group Chemical group Br* 0.000 description 1
- KDYFGRWQOYBRFD-NUQCWPJISA-N butanedioic acid Chemical compound O[14C](=O)CC[14C](O)=O KDYFGRWQOYBRFD-NUQCWPJISA-N 0.000 description 1
- 125000000484 butyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 229910002092 carbon dioxide Inorganic materials 0.000 description 1
- 150000001732 carboxylic acid derivatives Chemical class 0.000 description 1
- 230000003197 catalytic effect Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 230000005754 cellular signaling Effects 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 238000002512 chemotherapy Methods 0.000 description 1
- 238000005660 chlorination reaction Methods 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- KRVSOGSZCMJSLX-UHFFFAOYSA-L chromic acid Substances O[Cr](O)(=O)=O KRVSOGSZCMJSLX-UHFFFAOYSA-L 0.000 description 1
- 238000004891 communication Methods 0.000 description 1
- 238000006482 condensation reaction Methods 0.000 description 1
- YNYHGRUPNQLZHB-UHFFFAOYSA-M copper(1+);trifluoromethanesulfonate Chemical compound [Cu+].[O-]S(=O)(=O)C(F)(F)F YNYHGRUPNQLZHB-UHFFFAOYSA-M 0.000 description 1
- 239000012228 culture supernatant Substances 0.000 description 1
- 238000012258 culturing Methods 0.000 description 1
- 125000000753 cycloalkyl group Chemical group 0.000 description 1
- WLVKDFJTYKELLQ-UHFFFAOYSA-N cyclopropylboronic acid Chemical compound OB(O)C1CC1 WLVKDFJTYKELLQ-UHFFFAOYSA-N 0.000 description 1
- VRLDVERQJMEPIF-UHFFFAOYSA-N dbdmh Chemical compound CC1(C)N(Br)C(=O)N(Br)C1=O VRLDVERQJMEPIF-UHFFFAOYSA-N 0.000 description 1
- 238000007872 degassing Methods 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 238000011033 desalting Methods 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 229910052805 deuterium Inorganic materials 0.000 description 1
- 125000000664 diazo group Chemical group [N-]=[N+]=[*] 0.000 description 1
- 125000002720 diazolyl group Chemical group 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 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 1
- 201000010099 disease Diseases 0.000 description 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 239000006185 dispersion Substances 0.000 description 1
- 239000012153 distilled water Substances 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- WNAHIZMDSQCWRP-UHFFFAOYSA-N dodecane-1-thiol Chemical compound CCCCCCCCCCCCS WNAHIZMDSQCWRP-UHFFFAOYSA-N 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 230000007783 downstream signaling Effects 0.000 description 1
- 239000000890 drug combination Substances 0.000 description 1
- 238000009509 drug development Methods 0.000 description 1
- 229940112141 dry powder inhaler Drugs 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 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 1
- 229960001433 erlotinib Drugs 0.000 description 1
- 230000032050 esterification Effects 0.000 description 1
- 238000005886 esterification reaction Methods 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- CCIVGXIOQKPBKL-UHFFFAOYSA-M ethanesulfonate Chemical compound CCS([O-])(=O)=O CCIVGXIOQKPBKL-UHFFFAOYSA-M 0.000 description 1
- 125000004705 ethylthio group Chemical group C(C)S* 0.000 description 1
- 230000005284 excitation Effects 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 239000011737 fluorine Substances 0.000 description 1
- 229960002949 fluorouracil Drugs 0.000 description 1
- PJZDLZXMGBOJRF-CXOZILEQSA-L folfirinox Chemical compound [Pt+4].[O-]C(=O)C([O-])=O.[NH-][C@H]1CCCC[C@@H]1[NH-].FC1=CNC(=O)NC1=O.C1NC=2NC(N)=NC(=O)C=2N(C=O)C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1.C1=C2C(CC)=C3CN(C(C4=C([C@@](C(=O)OC4)(O)CC)C=4)=O)C=4C3=NC2=CC=C1OC(=O)N(CC1)CCC1N1CCCCC1 PJZDLZXMGBOJRF-CXOZILEQSA-L 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 235000003599 food sweetener Nutrition 0.000 description 1
- 239000003205 fragrance Substances 0.000 description 1
- 239000001530 fumaric acid Substances 0.000 description 1
- AWJWCTOOIBYHON-UHFFFAOYSA-N furo[3,4-b]pyrazine-5,7-dione Chemical compound C1=CN=C2C(=O)OC(=O)C2=N1 AWJWCTOOIBYHON-UHFFFAOYSA-N 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 150000004820 halides Chemical class 0.000 description 1
- 125000005843 halogen group Chemical group 0.000 description 1
- 150000002390 heteroarenes Chemical class 0.000 description 1
- 150000002430 hydrocarbons Chemical group 0.000 description 1
- 150000002431 hydrogen Chemical class 0.000 description 1
- XMBWDFGMSWQBCA-UHFFFAOYSA-N hydrogen iodide Chemical compound I XMBWDFGMSWQBCA-UHFFFAOYSA-N 0.000 description 1
- 229940071870 hydroiodic acid Drugs 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 239000012535 impurity Substances 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 108091006086 inhibitor proteins Proteins 0.000 description 1
- 230000031146 intracellular signal transduction Effects 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 239000011630 iodine Substances 0.000 description 1
- HVTICUPFWKNHNG-UHFFFAOYSA-N iodoethane Chemical compound CCI HVTICUPFWKNHNG-UHFFFAOYSA-N 0.000 description 1
- UWKQSNNFCGGAFS-XIFFEERXSA-N irinotecan Chemical compound C1=C2C(CC)=C3CN(C(C4=C([C@@](C(=O)OC4)(O)CC)C=4)=O)C=4C3=NC2=CC=C1OC(=O)N(CC1)CCC1N1CCCCC1 UWKQSNNFCGGAFS-XIFFEERXSA-N 0.000 description 1
- 229960004768 irinotecan Drugs 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 125000001786 isothiazolyl group Chemical group 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 230000000155 isotopic effect Effects 0.000 description 1
- 239000004310 lactic acid Substances 0.000 description 1
- 235000014655 lactic acid Nutrition 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 238000004020 luminiscence type Methods 0.000 description 1
- 229960003646 lysine Drugs 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- VZCYOOQTPOCHFL-UPHRSURJSA-N maleic acid Chemical compound OC(=O)\C=C/C(O)=O VZCYOOQTPOCHFL-UPHRSURJSA-N 0.000 description 1
- 239000011976 maleic acid Substances 0.000 description 1
- 239000001630 malic acid Substances 0.000 description 1
- 235000011090 malic acid Nutrition 0.000 description 1
- 230000036210 malignancy Effects 0.000 description 1
- 230000003211 malignant effect Effects 0.000 description 1
- 229960002510 mandelic acid Drugs 0.000 description 1
- 108010082117 matrigel Proteins 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 102000006240 membrane receptors Human genes 0.000 description 1
- 108020004084 membrane receptors Proteins 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 150000002739 metals Chemical class 0.000 description 1
- QARBMVPHQWIHKH-UHFFFAOYSA-N methanesulfonyl chloride Chemical compound CS(Cl)(=O)=O QARBMVPHQWIHKH-UHFFFAOYSA-N 0.000 description 1
- 239000002480 mineral oil Substances 0.000 description 1
- 235000010446 mineral oil Nutrition 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 239000002829 mitogen activated protein kinase inhibitor Substances 0.000 description 1
- SYSQUGFVNFXIIT-UHFFFAOYSA-N n-[4-(1,3-benzoxazol-2-yl)phenyl]-4-nitrobenzenesulfonamide Chemical class C1=CC([N+](=O)[O-])=CC=C1S(=O)(=O)NC1=CC=C(C=2OC3=CC=CC=C3N=2)C=C1 SYSQUGFVNFXIIT-UHFFFAOYSA-N 0.000 description 1
- GFRDSYFROJUKBF-UHFFFAOYSA-N n-diazoimidazole-1-sulfonamide Chemical compound [N-]=[N+]=NS(=O)(=O)N1C=CN=C1 GFRDSYFROJUKBF-UHFFFAOYSA-N 0.000 description 1
- 230000035407 negative regulation of cell proliferation Effects 0.000 description 1
- 229910052759 nickel Inorganic materials 0.000 description 1
- 229910017604 nitric acid Inorganic materials 0.000 description 1
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen(.) Chemical compound [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 1
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 1
- 239000012457 nonaqueous media Substances 0.000 description 1
- 238000011580 nude mouse model Methods 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 229960003104 ornithine Drugs 0.000 description 1
- 150000002905 orthoesters Chemical class 0.000 description 1
- 235000006408 oxalic acid Nutrition 0.000 description 1
- DWAFYCQODLXJNR-BNTLRKBRSA-L oxaliplatin Chemical compound O1C(=O)C(=O)O[Pt]11N[C@@H]2CCCC[C@H]2N1 DWAFYCQODLXJNR-BNTLRKBRSA-L 0.000 description 1
- 229960001756 oxaliplatin Drugs 0.000 description 1
- LMYJGUNNJIDROI-UHFFFAOYSA-N oxan-4-ol Chemical compound OC1CCOCC1 LMYJGUNNJIDROI-UHFFFAOYSA-N 0.000 description 1
- 125000000160 oxazolidinyl group Chemical group 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- MUMZUERVLWJKNR-UHFFFAOYSA-N oxoplatinum Chemical compound [Pt]=O MUMZUERVLWJKNR-UHFFFAOYSA-N 0.000 description 1
- 239000003002 pH adjusting agent Substances 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 229920002866 paraformaldehyde Polymers 0.000 description 1
- HJKYXKSLRZKNSI-UHFFFAOYSA-I pentapotassium;hydrogen sulfate;oxido sulfate;sulfuric acid Chemical compound [K+].[K+].[K+].[K+].[K+].OS([O-])(=O)=O.[O-]S([O-])(=O)=O.OS(=O)(=O)O[O-].OS(=O)(=O)O[O-] HJKYXKSLRZKNSI-UHFFFAOYSA-I 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- ZUOUZKKEUPVFJK-UHFFFAOYSA-N phenylbenzene Natural products C1=CC=CC=C1C1=CC=CC=C1 ZUOUZKKEUPVFJK-UHFFFAOYSA-N 0.000 description 1
- UYWQUFXKFGHYNT-UHFFFAOYSA-N phenylmethyl ester of formic acid Natural products O=COCC1=CC=CC=C1 UYWQUFXKFGHYNT-UHFFFAOYSA-N 0.000 description 1
- 239000008363 phosphate buffer Substances 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 239000002504 physiological saline solution Substances 0.000 description 1
- 229910003446 platinum oxide Inorganic materials 0.000 description 1
- 238000010837 poor prognosis Methods 0.000 description 1
- 238000002600 positron emission tomography Methods 0.000 description 1
- 235000011181 potassium carbonates Nutrition 0.000 description 1
- 239000012286 potassium permanganate Substances 0.000 description 1
- 238000000634 powder X-ray diffraction Methods 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 239000003380 propellant Substances 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 235000019260 propionic acid Nutrition 0.000 description 1
- 239000003223 protective agent Substances 0.000 description 1
- 239000008213 purified water Substances 0.000 description 1
- 125000003373 pyrazinyl group Chemical group 0.000 description 1
- UMJSCPRVCHMLSP-UHFFFAOYSA-N pyridine Natural products COC1=CC=CN=C1 UMJSCPRVCHMLSP-UHFFFAOYSA-N 0.000 description 1
- 150000003246 quinazolines Chemical class 0.000 description 1
- IUVKMZGDUIUOCP-BTNSXGMBSA-N quinbolone Chemical compound O([C@H]1CC[C@H]2[C@H]3[C@@H]([C@]4(C=CC(=O)C=C4CC3)C)CC[C@@]21C)C1=CCCC1 IUVKMZGDUIUOCP-BTNSXGMBSA-N 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 239000012266 salt solution Substances 0.000 description 1
- 229930195734 saturated hydrocarbon Natural products 0.000 description 1
- XIIOFHFUYBLOLW-UHFFFAOYSA-N selpercatinib Chemical compound OC(COC=1C=C(C=2N(C=1)N=CC=2C#N)C=1C=NC(=CC=1)N1CC2N(C(C1)C2)CC=1C=NC(=CC=1)OC)(C)C XIIOFHFUYBLOLW-UHFFFAOYSA-N 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- SUKJFIGYRHOWBL-UHFFFAOYSA-N sodium hypochlorite Chemical compound [Na+].Cl[O-] SUKJFIGYRHOWBL-UHFFFAOYSA-N 0.000 description 1
- 229910052938 sodium sulfate Inorganic materials 0.000 description 1
- 235000011152 sodium sulphate Nutrition 0.000 description 1
- AKHNMLFCWUSKQB-UHFFFAOYSA-L sodium thiosulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=S AKHNMLFCWUSKQB-UHFFFAOYSA-L 0.000 description 1
- PODWXQQNRWNDGD-UHFFFAOYSA-L sodium thiosulfate pentahydrate Chemical compound O.O.O.O.O.[Na+].[Na+].[O-]S([S-])(=O)=O PODWXQQNRWNDGD-UHFFFAOYSA-L 0.000 description 1
- 235000019345 sodium thiosulphate Nutrition 0.000 description 1
- 238000003797 solvolysis reaction Methods 0.000 description 1
- 238000001179 sorption measurement Methods 0.000 description 1
- VNFWTIYUKDMAOP-UHFFFAOYSA-N sphos Chemical compound COC1=CC=CC(OC)=C1C1=CC=CC=C1P(C1CCCCC1)C1CCCCC1 VNFWTIYUKDMAOP-UHFFFAOYSA-N 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000011301 standard therapy Methods 0.000 description 1
- 239000008223 sterile water Substances 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 238000009495 sugar coating Methods 0.000 description 1
- 125000004434 sulfur atom Chemical group 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 239000003765 sweetening agent Substances 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- 239000006188 syrup Substances 0.000 description 1
- 235000020357 syrup Nutrition 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 239000011975 tartaric acid Substances 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- UQVALAFEVCIICF-SQNIBIBYSA-N tert-butyl (2S,4R)-4-hydroxy-2-[[(1R)-2-hydroxy-1-[4-(4-methyl-1,3-thiazol-5-yl)phenyl]ethyl]carbamoyl]pyrrolidine-1-carboxylate Chemical compound O[C@@H]1C[C@H](N(C1)C(=O)OC(C)(C)C)C(N[C@@H](CO)C1=CC=C(C=C1)C1=C(N=CS1)C)=O UQVALAFEVCIICF-SQNIBIBYSA-N 0.000 description 1
- OKUCEQDKBKYEJY-MRVPVSSYSA-N tert-butyl (3r)-3-(methylamino)pyrrolidine-1-carboxylate Chemical compound CN[C@@H]1CCN(C(=O)OC(C)(C)C)C1 OKUCEQDKBKYEJY-MRVPVSSYSA-N 0.000 description 1
- CMIBWIAICVBURI-ZETCQYMHSA-N tert-butyl (3s)-3-aminopyrrolidine-1-carboxylate Chemical compound CC(C)(C)OC(=O)N1CC[C@H](N)C1 CMIBWIAICVBURI-ZETCQYMHSA-N 0.000 description 1
- 125000001981 tert-butyldimethylsilyl group Chemical group [H]C([H])([H])[Si]([H])(C([H])([H])[H])[*]C(C([H])([H])[H])(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- NHGXDBSUJJNIRV-UHFFFAOYSA-M tetrabutylammonium chloride Chemical compound [Cl-].CCCC[N+](CCCC)(CCCC)CCCC NHGXDBSUJJNIRV-UHFFFAOYSA-M 0.000 description 1
- YLQBMQCUIZJEEH-UHFFFAOYSA-N tetrahydrofuran Natural products C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 description 1
- 125000003831 tetrazolyl group Chemical group 0.000 description 1
- 125000001113 thiadiazolyl group Chemical group 0.000 description 1
- 239000002562 thickening agent Substances 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 125000004306 triazinyl group Chemical group 0.000 description 1
- IMNIMPAHZVJRPE-UHFFFAOYSA-N triethylenediamine Chemical compound C1CN2CCN1CC2 IMNIMPAHZVJRPE-UHFFFAOYSA-N 0.000 description 1
- ITMCEJHCFYSIIV-UHFFFAOYSA-M triflate Chemical compound [O-]S(=O)(=O)C(F)(F)F ITMCEJHCFYSIIV-UHFFFAOYSA-M 0.000 description 1
- 125000004044 trifluoroacetyl group Chemical group FC(C(=O)*)(F)F 0.000 description 1
- 125000004205 trifluoroethyl group Chemical group [H]C([H])(*)C(F)(F)F 0.000 description 1
- NQPHMXWPDCSHTE-UHFFFAOYSA-N trifluoromethanesulfonyl azide Chemical compound FC(F)(F)S(=O)(=O)N=[N+]=[N-] NQPHMXWPDCSHTE-UHFFFAOYSA-N 0.000 description 1
- 125000003258 trimethylene group Chemical group [H]C([H])([*:2])C([H])([H])C([H])([H])[*:1] 0.000 description 1
- OJZNYUDKNVNEMV-UHFFFAOYSA-M trimethylstannanylium;hydroxide Chemical compound C[Sn](C)(C)O OJZNYUDKNVNEMV-UHFFFAOYSA-M 0.000 description 1
- 229910052722 tritium Inorganic materials 0.000 description 1
- 230000004614 tumor growth Effects 0.000 description 1
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 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
- 210000003462 vein Anatomy 0.000 description 1
Landscapes
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Plural Heterocyclic Compounds (AREA)
Abstract
The present invention provides a compound useful as an active ingredient of a pharmaceutical composition for the treatment of pancreatic cancer. The present inventors have studied on compounds useful as active ingredients of pharmaceutical compositions for the treatment of pancreatic cancer, and have found that heterocyclic compounds represented by the formula (I) and the formula (IA) have excellent G12D mutant KRAS proteolytic activity and G12D mutant KRAS inhibitory activity, and are useful as pancreatic cancer therapeutic agents, thereby completing the present invention. The heterocyclic compound or salt thereof of the present invention is useful as a therapeutic agent for pancreatic cancer.
Description
Technical Field
The present invention relates to a pharmaceutical composition and a heterocyclic compound which has an excellent effect of inducing the proteolytic degradation of G12D mutant KRAS and is useful as a G12D mutant KRAS inhibitor, and for example, is expected to be useful as an active ingredient of a pharmaceutical composition for the treatment of pancreatic cancer.
Background
Pancreatic Cancer, which is mainly pancreatic ductal adenocarcinoma, is a Cancer with a survival rate of 10% or less (CA Cancer j.clin.,2016,66, p.7-30) for 5 years and a very poor prognosis, and it is reported that about 46 ten thousand cases are newly increased every year worldwide (CA Cancer j.clin.,2018,68, p.394-424). The most effective therapy in the treatment of pancreatic cancer is surgery, but most of them have been shifted due to early finding difficulty, and most of them cannot be expected to have therapeutic effects by the surgical treatment. Chemotherapy and radiotherapy are used without surgical treatment, but survival rate is not ideal. Currently, FOLFIRINOX therapy (multi-drug combination therapy with the addition of levofolinate on the basis of 3 chemotherapeutics, 5-FU, irinotecan and oxaliplatin) is used as a standard therapy for pancreatic cancer, but due to its strong toxicity, patients prescribed are limited to ECOG physical states (ECOG Performance Status) of 1 or less, and careful patient selection is required (j.clin.oncol., 2018,36, p.2545-2556). As a molecular targeted therapeutic, although the combination therapy of erlotinib, an Epidermal Growth Factor Receptor (EGFR) inhibitor, with gemcitabine has been approved, the total lifetime is prolonged by about 2 weeks as compared with gemcitabine alone, and a satisfactory therapeutic effect is not obtained, and there is still a need for a highly effective therapeutic (j.clin.oncol., 2007,25, p.1960-1966).
RAS proteins are low molecular Guanosine Triphosphate (GTP) binding proteins of about 21kDa consisting of 188-189 amino acids, and there are 4 major proteins (KRAS (KRAS 4A and KRAS 4B), NRAS, HRAS) produced by 3 genes of KRAS gene, NRAS gene, HRAS gene. RAS proteins exist as active GTP-bound and as inactive GDP-bound. The RAS protein is activated by exchanging Guanosine Diphosphate (GDP) with GTP by ligand stimulation of cell membrane receptors such as EGFR. Active RAS binds to up to 20 effector proteins such as RAF, PI3K, RALGDS, etc. to activate downstream signaling cascades. On the other hand, active RAS converts GTP into GDP by endogenous GTP hydrolysis (GTPase) activity, thereby becoming inactive. The GTPase activity may be enhanced by GTPase Activating Proteins (GAPs). Therefore, RAS plays an important role in the growth, proliferation, angiogenesis and other processes of cells by taking on the important "molecular switch" function in the intracellular signaling pathway such as EGFR (Nature Rev.Cancer,2011,11,p.761-774、Nature Rev.Drug Discov.,2014,13,p.828-851、Nature Rev.Drug Discov.,2016,15,p.771-785).
When an amino acid substitution occurs due to mutation of the RAS gene, the function of the RAS as GTPase is reduced, and the response to GAP is reduced, thereby forming a continuous activation state, and a signal is continuously transmitted downstream. This excessive signal leads to canceration and hyperproliferation of tumors. Pancreatic ductal adenocarcinoma is said to occur as a result of pancreatic intraepithelial neoplasia/PANCREATIC INTRAEPITHELIAL neoplasia (PanIN) undergoing a stage of less dysplasia to a stage of greater, early stage PanIN having seen KRAS gene mutations. Thereafter, the oncogenes INK4A, p and SMAD4 were abnormal and malignant (Nature Rev. Cancer,2010,10, p. 683-695). Furthermore, mutations are seen in the KRAS gene in 90% or more of pancreatic ductal adenocarcinomas, with most of the point mutations at codon 12 of KRAS exon 2 (CANCER CELL 2017,32, p.185-203). Thus, KRAS plays an important role in the cancerous and progressive processes of pancreatic cancer.
As KRAS gene mutations, KRAS G12C mutations, KRAS G12D mutations, etc. are known, but G12C mutations KRAS are highly frequent in non-small cell lung cancer, but are only a few percent in pancreatic cancer (CANCER CELL 2014,25, p.272-281), therapeutic agents against other KRAS mutations are desirable. The G12D mutation KRAS was reported to be seen in about 34% of pancreatic cancers, with the highest proportion among KRAS mutations (nat. Rev. Cancer,2018,18, p. 767-777).
Patent documents 1,2 and 3 disclose RAS inhibitors, and patent documents 2 and 3 disclose compounds represented by the following formulas (a) and (B), respectively (reference is made to the publication for the meaning of symbols in the formulas). Patent documents 1,2 and 3 describe that it is useful for cancers in which there is a mutation at KRAS codon 12, including G12D mutation, but do not describe the effect on G12D mutated KRAS cancers.
In addition, patent documents 9, 10 and 11 disclose KRAS G12D inhibitors.
In recent years, as a technique for inducing target protein degradation, bifunctional compounds, collectively referred to as PROTAC (PROteolysis-TARGETING CHIMERA, protein degradation targeting chimera), SNIPER (SPECIFIC AND Nongenetic IAP-DEPENDENT PROTEIN ERASER, specific and non-genetic IAP-dependent protein eraser), and the like, have been found and expected as one of novel drug development modes (drug. The bifunctional compound promotes complex formation of the target protein and the E3 ligase in the cell, inducing target protein breakdown by utilizing the ubiquitin-proteasome system. Ubiquitin-proteasome systems are one of the proteolytic mechanisms within cells. The protein called E3 ligase recognizes and ubiquitinates the protein that should be decomposed, thereby advancing the decomposition in the proteasome.
More than 600E 3 ligases exist in organisms and can be broadly divided into 4 types, HECT-domain E3s, U-box E3s, monomeric RING E3s and multi-subunit E3 s. Currently, E3 ligases used as bifunctional decomposition inducers such as PROTAC, SNIPER are limited, and representative examples thereof include Von Hippel-Lindau (VHL), celebron (CRBN), apoptosis inhibitor protein (IAP), and mouse double-micro homolog 2 (mouse double minute homolog, MDM 2). In particular, patent document 4 reports VHL, and patent document 5 reports CRBN.
The bifunctional compound is a compound in which a ligand of a target protein and a ligand of E3 ligase are linked by a linker, and a bifunctional compound that breaks down KRAS protein has been reported previously (non-patent document 1, non-patent document 2, patent document 6, patent document 7, patent document 8, patent document 12, and patent document 13). Further, patent document 14 and patent document 15 report bifunctional compounds that reduce the level of G12D mutant KRAS protein, and patent document 16 reports quinazoline compounds for inducing the decomposition of G12D mutant KRAS protein. However, there is no report that suggests that a bifunctional compound breaks down a G12D mutant KRAS protein, except for patent documents 14 to 16.
Prior art literature
Patent literature
Patent document 1: international publication No. 2016/049565
Patent document 2: international publication No. 2016/049568
Patent document 3: international publication No. 2017/172979
Patent document 4: international publication No. 2013/106643
Patent document 5: international publication No. 2015/160845
Patent document 6: U.S. patent application publication No. 2018/0015087
Patent document 7: international publication No. 2019/195609
Patent document 8: international publication No. 2020/018788
Patent document 9: international publication No. 2021/04671
Patent document 10: international publication No. 2021/106231
Patent document 11: international publication No. 2021/107160
Patent document 12: international publication No. 2021/051034
Patent document 13: international publication No. 2021/207172
Patent document 14: international publication No. 2022/148421
Patent document 15: international publication No. 2022/148422
Patent document 16: international publication No. 2022/173032
Non-patent literature
Non-patent document 1: cell chem. Biol 2020,27, p19-31
Non-patent document 2: ACS cent.sci.,2020,6, p1367-1375
Disclosure of Invention
Problems to be solved by the invention
Provided is a heterocyclic compound which has excellent action of inducing G12D mutant KRAS protein degradation, is useful as a G12D mutant KRAS inhibitor, and is expected to be useful as an active ingredient of a pharmaceutical composition for the treatment of pancreatic cancer, particularly G12D mutant KRAS positive pancreatic cancer.
Means for solving the problems
As a result of intensive studies on compounds useful as active ingredients of pharmaceutical compositions for the treatment of pancreatic cancer, the present inventors have found that a heterocyclic compound of the formula (I) and the formula (IA), in particular, a bifunctional compound of the formula (I) and the formula (IA) characterized by linking a substituent at the 8-position of the heterocyclic compound selected from the group consisting of quinazoline and quinoline to a ligand of E3 ligase or linking a substituent at the 8-position of the heterocyclic compound selected from the group consisting of quinazoline and quinoline to a ligand of E3 ligase with a linker has excellent G12D mutant KRAS proteolytic induction effect and G12D mutant KRAS inhibitory activity, and have completed the present invention.
That is, the present invention relates to a compound of formula (I) or a salt thereof, and a pharmaceutical composition containing a compound of formula (I) or a salt thereof and one or more pharmaceutically acceptable excipients.
(In the formula (I),
A is CR A or N, and the total number of the components is N,
R A is H or C 1-3 alkyl,
R 1 is a naphthyl group which may be substituted by OH or a group selected from the group consisting of the following formula (II), formula (III) and formula (IV),
R 1a、R1b are identical or different from each other and are H, methyl, F or Cl,
R 1c is F, cl, methyl or ethyl,
R 2 is H, halogen, optionally substituted C 1-3 alkyl, cyclopropyl or vinyl,
R 3 is-P-Q or V,
P is-CH 2 -, -O-or-N (R P) -,
R P is H or C 1-3 alkyl which can be substituted,
Q is the following formula (V) or formula (VI),
V is the following formula (VII),
R Q, which are identical or different from each other, are OH or C 1-3 alkyl, and R Q is bonded only to carbon atoms as constituent atoms of rings selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
M is a number from 0 to 2,
R 4 is an optionally substituted C 1-6 alkyl group, an optionally substituted 4-to 6-membered saturated heterocyclic group containing 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen, an optionally substituted 5-membered heteroaryl group containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen or an optionally substituted 6-membered heteroaryl group containing 1 to 3 nitrogen atoms,
R 5 is C 1-6 alkyl which may be substituted, C 3-6 cycloalkyl which may be substituted or a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 hetero atom selected from the group consisting of oxygen, sulfur and nitrogen,
R 6a、R6b, identical or different from one another, is H or C 1-6 -alkyl which may be substituted, or R 6a、R6b together with the carbon to which they are bonded may form C 3-6 -cycloalkyl which may be substituted or a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 heteroatom selected from the group consisting of oxygen, sulfur and nitrogen,
R 7 is H, halogen, C 1-3 alkyl, -SO 2CH3、C3-6 cycloalkyl, an optionally substituted 4-to 6-membered saturated heterocyclic group containing 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen, an optionally substituted 5-membered heteroaryl group containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen or a 6-membered heteroaryl group containing 1 to 3 nitrogen atoms,
W is optionally substituted phenylene or optionally substituted 6-membered heteroarene diyl having 1 to 3 nitrogen atoms,
X is a bond, -CH 2 -, -O-, -S-, or-NR 4x -,
R 4x is H or C 1-3 alkyl,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is- (L 1-L2-L3-L4) -,
L 1、L2、L3、L4, which are identical or different from one another, are selected from the group consisting of bond, -O-, -NR L1 -, pyrrolidinediyl which may be substituted, piperidinyl which may be substituted, piperazinediyl which may be substituted, C 1-3 alkylene which may be substituted and C=O,
R L1 is H or C 1-3 alkyl,
Z is NH or a 5-membered heteroarene diyl containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulphur and nitrogen,
Or Y-L-Z is the following formula (VIII). )
The present invention also relates to a compound of formula (IA) or a salt thereof, and a pharmaceutical composition containing a compound of formula (IA) or a salt thereof and one or more pharmaceutically acceptable excipients.
(In the formula (I),
A is CR A or N, and the total number of the components is N,
R A is H or C 1-3 alkyl,
E is CH or N, and the R is H,
G is CR 2 or N, and the total number of the components is N,
R 1 is a naphthyl group which may be substituted by OH or a group selected from the group consisting of the following formula (II), formula (III) and formula (IV),
R 1a、R1b are identical or different from each other and are H, methyl, F or Cl,
R 1c is F, cl, methyl or ethyl,
R 2 is H, halogen, optionally substituted C 1-3 alkyl, cyclopropyl or vinyl,
R 3 is-P-Q or V,
P is-CH 2 -, -O-or-N (R P) -,
R P is H or C 1-3 alkyl which can be substituted,
Q is the following formula (V) or formula (VI),
V is the following formula (VII),
R Q, which are identical or different from each other, are OH or C 1-3 alkyl, and R Q is bonded only to carbon atoms as constituent atoms of rings selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
In addition, (i) when A is CR A, or (ii) when A is N and E or G is N,
V may be a saturated or unsaturated 7-to 8-membered bridged heterocyclic group other than the formula (VII),
M is a number from 0 to 2,
R 4 is an optionally substituted C 1-6 alkyl group, an optionally substituted 4-to 6-membered saturated heterocyclic group containing 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen, an optionally substituted 5-membered heteroaryl group containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen or an optionally substituted 6-membered heteroaryl group containing 1 to 3 nitrogen atoms,
R 5 is C 1-6 alkyl which may be substituted, C 3-6 cycloalkyl which may be substituted or a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 hetero atom selected from the group consisting of oxygen, sulfur and nitrogen,
R 6a、R6b, identical or different from one another, is H or C 1-6 -alkyl which may be substituted, or R 6a、R6b together with the carbon to which they are bonded may form C 3-6 -cycloalkyl which may be substituted or a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 heteroatom selected from the group consisting of oxygen, sulfur and nitrogen,
R 7 is H, halogen, C 1-3 alkyl, -SO 2CH3、C3-6 cycloalkyl, an optionally substituted 4-to 6-membered saturated heterocyclic group containing 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen, an optionally substituted 5-membered heteroaryl group containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen or a 6-membered heteroaryl group containing 1 to 3 nitrogen atoms,
L Y is-O- (optionally substituted C 1-3 alkylene) -, S- (C 1-3 alkylene which may be substituted) -, -SO 2 - (optionally substituted C 1-3 alkylene) -, -NR Y - (optionally substituted C 1-3 alkylene) -, and- (C 1-3 alkylene which may be substituted) -O-, C 1-3 alkylene which may be substituted) -S-, C 1-3 alkylene which may be substituted) -SO 2 -, C 1-3 alkylene which may be substituted) -NR Y -,
R Y is H or C 1-3 alkyl,
R P1 is OH or F, and the total number of the components is H,
R P2a is H or F,
R P2b is H, and the hydrogen atom,
W is optionally substituted phenylene or optionally substituted 6-membered heteroarene diyl having 1 to 3 nitrogen atoms,
X is a bond, -CH 2 -, -O-, -S-, or-NR 4x -,
R 4x is H or C 1-3 alkyl,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is- (L 1-L2-L3-L4) -,
L 1、L2、L3、L4, which are identical or different from one another, are selected from the group consisting of bond, -O-, -NR L1 -, pyrrolidinediyl which may be substituted, piperidinyl which may be substituted, piperazinediyl which may be substituted, C 1-3 alkylene which may be substituted and C=O,
R L1 is H or C 1-3 alkyl,
Z is NH or a 5-membered heteroarene diyl containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulphur and nitrogen,
Or Y-L-Z is the following formula (VIII). )
The compound of formula (IA) includes a compound of formula (I).
Unless otherwise specified, the same symbols in a given chemical formula in the present specification are used in other chemical formulas, and the same symbols are used in the same meaning.
In addition, the present invention relates to pharmaceutical compositions comprising a compound of formula (I) or a salt thereof and one or more pharmaceutically acceptable excipients; as one embodiment, a pharmaceutical composition for pancreatic cancer treatment; as one mode, a pharmaceutical composition for the treatment of G12D mutant KRAS positive pancreatic cancer; as one embodiment, a pharmaceutical composition for the treatment of metastatic pancreatic cancer; as one mode, a pharmaceutical composition for the treatment of locally advanced pancreatic cancer; as one mode, a pharmaceutical composition for the treatment of recurrent or refractory pancreatic cancer; a pharmaceutical composition for treating pancreatic cancer in an untreated and/or treatment-history patient as one mode; as one mode, a pharmaceutical composition for the treatment of metastatic G12D mutant KRAS positive pancreatic cancer; as one mode, a pharmaceutical composition for the treatment of locally progressive G12D mutant KRAS positive pancreatic cancer; as one mode, a pharmaceutical composition for the treatment of recurrent or refractory G12D mutant KRAS positive pancreatic cancer; pharmaceutical compositions for the treatment of G12D mutant KRAS positive pancreatic cancer in untreated and/or treatment-history patients as one mode. The pharmaceutical composition comprises pancreatic cancer containing a compound of formula (I) or a salt thereof, and a therapeutic agent for G12D mutant KRAS-positive pancreatic cancer as one embodiment.
In addition, the present invention relates to the use of a compound of formula (I) or a salt thereof for the manufacture of a pharmaceutical composition for the treatment of pancreatic cancer, G12D mutant KRAS positive pancreatic cancer as one mode, metastatic pancreatic cancer as one mode, locally advanced pancreatic cancer as one mode, recurrent or refractory pancreatic cancer as one mode, pancreatic cancer of an untreated and/or treated patient as one mode, metastatic G12D mutant KRAS positive pancreatic cancer as one mode, locally advanced G12D mutant KRAS positive pancreatic cancer as one mode, recurrent or refractory G12D mutant KRAS positive pancreatic cancer as one mode, G12D mutant KRAS positive pancreatic cancer as one mode of untreated and/or treated patient; use of a compound of formula (I) or a salt thereof for the treatment of pancreatic cancer, G12D mutant KRAS positive pancreatic cancer as one mode; a compound of formula (I) or a salt thereof for use in the treatment of pancreatic cancer, G12D mutant KRAS positive pancreatic cancer as one mode; and a method of treating pancreatic cancer, G12D mutant KRAS positive pancreatic cancer as one mode, comprising the step of administering to a subject an effective amount of a compound of formula (I) or a salt thereof.
In addition, the present invention relates to a compound of formula (I) or a salt thereof as a G12D mutant KRAS proteolytic inducer and/or a G12D mutant KRAS inhibitor, a G12D mutant KRAS proteolytic inducer and/or a G12D mutant KRAS inhibitor containing a compound of formula (I) or a salt thereof.
By "subject" is meant a person or other animal in need of such treatment, as one means, a person in need of such prevention or treatment.
In addition, the present invention relates to pharmaceutical compositions comprising a compound of formula (IA) or a salt thereof and one or more pharmaceutically acceptable excipients; as one embodiment, a pharmaceutical composition for pancreatic cancer treatment; as one mode, a pharmaceutical composition for the treatment of G12D mutant KRAS positive pancreatic cancer; as one embodiment, a pharmaceutical composition for the treatment of metastatic pancreatic cancer; as one mode, a pharmaceutical composition for the treatment of locally advanced pancreatic cancer; as one mode, a pharmaceutical composition for the treatment of recurrent or refractory pancreatic cancer; a pharmaceutical composition for treating pancreatic cancer in an untreated and/or treatment-history patient as one mode; as one mode, a pharmaceutical composition for the treatment of metastatic G12D mutant KRAS positive pancreatic cancer; as one mode, a pharmaceutical composition for the treatment of locally progressive G12D mutant KRAS positive pancreatic cancer; as one mode, a pharmaceutical composition for the treatment of recurrent or refractory G12D mutant KRAS positive pancreatic cancer; pharmaceutical compositions for the treatment of G12D mutant KRAS positive pancreatic cancer in untreated and/or treatment-history patients as one mode. The pharmaceutical composition comprises a pancreatic cancer comprising a compound of formula (IA) or a salt thereof, and a therapeutic agent for G12D mutant KRAS-positive pancreatic cancer as one embodiment.
In addition, the present invention relates to the use of a compound of formula (IA) or a salt thereof for the manufacture of a pharmaceutical composition for the treatment of pancreatic cancer, G12D mutant KRAS positive pancreatic cancer as one mode, metastatic pancreatic cancer as one mode, locally advanced pancreatic cancer as one mode, recurrent or refractory pancreatic cancer as one mode, pancreatic cancer of an untreated and/or treated patient as one mode, metastatic G12D mutant KRAS positive pancreatic cancer as one mode, locally advanced G12D mutant KRAS positive pancreatic cancer as one mode, recurrent or refractory G12D mutant KRAS positive pancreatic cancer as one mode, G12D mutant KRAS positive pancreatic cancer of an untreated and/or treated patient as one mode; use of a compound of formula (IA) or a salt thereof for the treatment of pancreatic cancer, G12D mutant KRAS positive pancreatic cancer as one mode; a compound of formula (IA) or a salt thereof for use in the treatment of pancreatic cancer, G12D mutant KRAS positive pancreatic cancer as one mode; and a method of treating pancreatic cancer, G12D mutant KRAS positive pancreatic cancer as one mode, comprising the step of administering to a subject an effective amount of a compound of formula (IA) or a salt thereof.
In addition, the present invention relates to a compound of formula (IA) or a salt thereof as a G12D mutant KRAS proteolytic inducer and/or a G12D mutant KRAS inhibitor, a G12D mutant KRAS proteolytic inducer and/or a G12D mutant KRAS inhibitor containing a compound of formula (IA) or a salt thereof.
By "subject" is meant a person or other animal in need of such treatment, as one means, a person in need of such prevention or treatment.
Effects of the invention
The compound of formula (I) or (IA) or a salt thereof has an effect of inducing the proteolytic decomposition of G12D mutant KRAS and G12D mutant KRAS inhibitory activity, and can be used as a therapeutic agent for pancreatic cancer, particularly G12D mutant KRAS positive pancreatic cancer.
Detailed Description
The present invention will be described in detail below.
In the present specification, "may be substituted" means unsubstituted or having 1 to 5 substituents. As one mode, it means unsubstituted or having 1 to 3 substituents. When 2 or more substituents are present, these substituents may be the same or different from each other.
"C 1-12 alkyl" means a straight or branched alkyl group having 1 to 12 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, dodecyl, etc. (hereinafter, the same applies to carbon numbers). As one embodiment, ethyl or dodecyl.
Similarly, "C 1-6 alkyl" refers to straight or branched alkyl groups having 1 to 6 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl or n-hexyl, as one mode, methyl, ethyl, n-propyl, isopropyl or sec-butyl, as one mode, methyl, ethyl, isopropyl or tert-butyl, as one mode, methyl, ethyl, n-propyl, isopropyl, n-butyl.
Similarly, "C 1-3 alkyl" refers to a straight or branched alkyl group having 1 to 3 carbon atoms, such as methyl, ethyl, n-propyl, or isopropyl, as one embodiment, methyl or ethyl, as one embodiment, n-propyl or isopropyl, as one embodiment, methyl or isopropyl, as one embodiment, ethyl or isopropyl, as one embodiment, methyl, as one embodiment, ethyl, as one embodiment, isopropyl, as one embodiment, n-propyl.
"C 3-6 cycloalkyl" means cycloalkyl having 3 to 6 carbon atoms, for example cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl. In one embodiment, the catalyst is a cyclobutyl group, a cyclopentyl group or a cyclohexyl group, in one embodiment, a cyclobutyl group or a cyclopentyl group, in one embodiment, a cyclopentyl group or a cyclohexyl group, in one embodiment, a cyclopropyl group, in one embodiment, a cyclobutyl group, in one embodiment, a cyclopentyl group, in one embodiment, a cyclohexyl group.
The "C 1-3 alkylene" refers to a divalent group obtained by removing a hydrogen atom from a C 1-3 alkyl group, and is a linear or branched C 1-3 alkylene group, for example, methylene, ethylene, trimethylene, methyl methylene, 1-dimethylmethylene, or the like. As one embodiment, the C 1-3 alkylene group is a straight-chain or branched-chain alkylene group, as one embodiment, a methylene group, an ethylene group, or a trimethylene group, as one embodiment, a methylene group or an ethylene group, as one embodiment, a methylene group, as one embodiment, an ethylene group.
"Saturated heterocyclic group" means a saturated hydrocarbon ring group containing a heteroatom selected from the group consisting of oxygen, sulfur and nitrogen as a ring constituting atom. In addition, a sulfur atom as a ring constituent atom of the saturated heterocyclic group may be oxidized.
Thus, the "4-to 6-membered saturated heterocyclic group" means a 4-to 6-membered saturated heterocyclic group containing a heteroatom selected from the group consisting of oxygen, sulfur and nitrogen as a ring constituting atom. One embodiment of the "4-to 6-membered saturated heterocyclic group" is a 4-to 6-membered saturated heterocyclic group containing 1 to 2 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen as ring constituting atoms. As one mode of a 4-to 6-membered saturated heterocyclic group containing 1 to 2 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen as ring constituent atoms, a 4-to 6-membered saturated heterocyclic group containing 1 hetero atom selected from the group consisting of oxygen, sulfur and nitrogen as ring constituent atoms, a 5-to 6-membered saturated heterocyclic group containing 1 to 2 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen as ring constituent atoms, a 4-membered saturated heterocyclic group containing 1 hetero atom selected from the group consisting of oxygen, sulfur and nitrogen as ring constituent atoms, a 5-membered saturated heterocyclic group containing 1 to 2 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen as ring constituent atoms, a 6-membered saturated heterocyclic group containing 1 to 2 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen as ring constituent atoms, a oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, pyrrolidinyl, piperidinyl, pyrrolidinyl, and the like,An oxazolyl, imidazolyl, piperazinyl, morpholinyl, thiomorpholinyl, or dioxothiomorpholinyl group as one mode, an oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, or dioxothiomorpholinyl group as one mode, an oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, pyrrolidinyl, piperidinyl, or morpholinyl group as one mode, an oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, pyrrolidinyl, or piperidinyl group as one mode, an oxetanyl, tetrahydrofuranyl, or tetrahydropyranyl as one mode, an pyrrolidinyl or piperidinyl group as one mode, an oxetanyl as one mode, a tetrahydrofuranyl as one mode, a tetrahydropyranyl as one mode, a pyrrolidinyl as one mode, a piperidinyl as one mode, a morpholinyl as one mode, a tetrahydropyranyl as one mode, a pyrrolidinyl as one mode, an oxetanyl as one mode, a tetrahydropyranyl as one modeAn oxazolidinyl group.
"Heteroaryl" refers to an unsaturated heterocyclic group containing a heteroatom selected from the group consisting of oxygen, sulfur, and nitrogen as a ring constituting atom.
Thus, "5-membered heteroaryl" refers to an unsaturated heterocyclic group containing 1 to 4 5-membered rings containing heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen as ring constituting atoms.
As one embodiment of the "5-membered heteroaryl group", there is an unsaturated heterocyclic group having a 5-membered ring containing 1 to 3 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen as ring constituting atoms, and as one embodiment, there is a pyrazolyl group, an imidazolyl group, a triazolyl group, a tetrazolyl group,Azolyl, isoOxazolyl, thiazolyl, isothiazolyl, and the like,Diazolyl or thiadiazolyl, as one mode, pyrazolyl, imidazolyl, triazolyl,An oxazolyl group or a thiazolyl group, as one mode, is pyrazolyl imidazolyl group,An oxazolyl or thiazolyl group, as one mode, a pyrazolyl, imidazolyl, triazolyl or isoxazolyl groupAzolyl, as one embodiment, is pyrazolyl,An oxazolyl or thiazolyl group, as one mode, a pyrazolyl, triazolyl or isoxazolyl groupOxazolyl, as one embodiment, pyrazolyl or thiazolyl, as one embodiment, pyrazolyl or triazolyl, as one embodiment, pyrazolyl, as one embodiment, imidazolyl, as one embodiment, isThe oxazolyl group is, as an embodiment, a thiazolyl group, and as an embodiment, a triazolyl group. The term "5-membered heteroarenediyl" refers to a divalent group formed by removing any 1 hydrogen from "5-membered heteroaryl".
"6-Membered heteroaryl" means a 6-membered ring unsaturated heterocyclic group containing 1 to 3 nitrogen atoms as ring constituting atoms. One embodiment of the "6-membered heteroaryl group" is a pyridyl group, a pyrimidinyl group, a pyrazinyl group, a pyridazinyl group or a triazinyl group, one embodiment is a pyridyl group or a pyridazinyl group, one embodiment is a pyridyl group or a pyrimidinyl group, one embodiment is a pyridyl group, one embodiment is a pyrimidinyl group. The "6-membered heteroarenediyl" refers to a divalent group formed by removing any 1 hydrogen from the "6-membered heteroaryl".
"Halogen" means F, cl, br and I. One embodiment of "halogen" is F, cl or Br, one embodiment is F or Cl, one embodiment is F or Br, one embodiment is F, one embodiment is Cl, and one embodiment is Br.
"Saturated or unsaturated 7-to 8-membered bridged heterocyclic group" means a saturated 7-to 8-membered bridged heterocyclic group containing 1 to 2 nitrogen atoms as ring constituting atoms or a 7-to 8-membered bridged heterocyclic group having an unsaturated bond containing 1 to 2 nitrogen atoms. In one embodiment, the heterocyclic group is a saturated 7-to 8-membered heterocyclic group containing 2 nitrogen atoms, and in one embodiment, the heterocyclic group is a saturated 7-to 8-membered heterocyclic group containing 2 nitrogen atoms and having 1 of the 2 nitrogen atoms bonded to 1 hydrogen atom. For example diazabicyclo [2.2.2] octyl, diazabicyclo [3.2.1] octyl diazabicyclo [3.2.1] octenyl diazabicyclo [3.1.1] heptyl, diazabicyclo [2.2.1] heptyl, and diazabicyclo [2.2.1] heptenyl. As one mode, diazabicyclo [2.2.2] octyl, diazabicyclo [3.2.1] oct-6-enyl, diazabicyclo [3.2.1] oct-2-enyl, diazabicyclo [3.1.1] heptyl, diazabicyclo [2.2.1] hept-5-enyl, as one mode, diazabicyclo [2.2.2] octyl, diazabicyclo [3.2.1] octyl, diazabicyclo [3.1.1] heptyl or diazabicyclo [2.2.1] heptyl, as one mode, 2, 5-diazabicyclo [2.2.2] octyl, 3, 8-diazabicyclo [3.2.1] octyl, 3, 6-diazabicyclo [3.1.1] heptyl or 2, 5-diazabicyclo [ 2.1.1 ] heptyl, as one mode, 2, 2.1-bicycloheptyl, as one mode, 2, 5-diazabicyclo [ 2.1.1 ] heptyl, as one mode, 2-5-diazabicyclo [ 2.2.1.1 ] heptyl.
As an acceptable substituent in the "optionally substituted C 1-6 alkyl" and "optionally substituted C 1-3 alkyl" are mentioned F, OH, OCH 3、N(CH3)2, optionally substituted C 3-6 cycloalkyl, azabicyclo [3.3.0] octyl or a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen. As one mode, F, OH, OCH 3、N(CH3)2, hydroxymethyl, methoxymethyl, difluoroethyl, optionally substituted cyclopropyl, tetrahydrofuranyl, optionally substituted tetrahydropyranyl, morpholinyl, optionally substituted pyrrolidinyl, optionally substituted piperidinyl or azabicyclo [3.3.0] octyl, as one mode, F, OH, OCH 3、N(CH3)2, Hydroxymethyl, methoxymethyl, optionally substituted cyclopropyl, tetrahydrofuranyl, optionally substituted tetrahydropyranyl, optionally substituted pyrrolidinyl, as one form F, OH, OCH 3、N(CH3)2, hydroxymethyl, methoxymethyl, cyclopropyl, (hydroxymethyl) cyclopropyl, (methoxymethyl) cyclopropyl, tetrahydrofuranyl, tetrahydropyranyl, (hydroxymethyl) tetrahydropyranyl, (methoxymethyl) tetrahydropyranyl, pyrrolidinyl or methylpyrrolidinyl, as one form F, F, OH, OCH 3, (methoxymethyl) cyclopropyl, tetrahydrofuranyl or methylpyrrolidinyl, F, OH or cyclopropyl as one mode, F, OH or OCH 3 as one mode, OH or OCH 3 as one mode, As one embodiment, F or OCH 3, as one embodiment, OH, as one embodiment, F, as one embodiment, OCH 3.
As one mode of acceptable substituents in "5-membered heteroaryl group which may be substituted", "6-membered heteroarenediyl group which may be substituted", "C 3-6 cycloalkyl group which may be substituted", "pyrazolyl group which may be substituted", "pyridinyl group which may be substituted", "pyrimidinyl group which may be substituted", "phenyl group which may be substituted" and "cyclopropyl group which may be substituted", C 1-3 alkyl group which may be substituted by a group selected from the group consisting of OH and OCH 3, -SO 2CH3, halogen, OH, OCH 3 or C 3-6 cycloalkyl group. As one embodiment, C 1-3 alkyl which may be substituted with a group selected from the group consisting of OH and OCH 3, as one embodiment, C 1-3 alkyl which may be substituted with OH, as one embodiment, C 1-3 alkyl which may be substituted with OCH 3, as one embodiment, C 1-3 alkyl or halogen, as one embodiment, methyl, ethyl, methoxymethyl or F, as one embodiment, methyl, ethyl or F.
As one mode of acceptable substituents in the "4-to 6-membered saturated heterocyclic group which may be substituted", "pyrrolidinyl group which may be substituted", "piperidinyl group which may be substituted", "oxetanyl group which may be substituted", "tetrahydrofuranyl group which may be substituted", and "tetrahydropyranyl group which may be substituted", there is C 1-3 alkyl group which may be substituted with a group selected from the group consisting of F, OH and OCH 3, F. OH, OCH 3, oxo or oxetanyl. F, OH or OCH 3, OH or methyl, and C 1-3 alkyl which may be substituted by a group selected from the group consisting of F, OH and OCH 3, F, Oxo or oxetanyl, as one form of C 1-3 alkyl or oxo which may be substituted with a group selected from the group consisting of F, OH and OCH 3, as one form of C 1-3 alkyl which may be substituted with a group selected from the group consisting of F, OH and OCH 3, As one embodiment, a C 1-3 alkyl group which may be substituted by F, as one embodiment, a C 1-3 alkyl group which may be substituted by OH, as one embodiment, a C 1-3 alkyl group which may be substituted by OCH 3, as one embodiment, C 1-3 alkyl.
As one mode of acceptable substituents in "pyrrolidinediyl which may be substituted", "piperidediyl which may be substituted", "piperazinediyl which may be substituted", "C 1-3 alkylene which may be substituted", no F, OH, OCH 3 or C 1-3 alkyl which may be substituted. As one embodiment, F, OH, OCH 3, methyl, ethyl, hydroxymethyl or methoxymethyl, as one embodiment, F, OH, OCH 3 or methyl.
As one mode of the "C 1-3 alkyl group which may be substituted by F", a methyl group which may be substituted by F or an ethyl group which may be substituted by F is mentioned. Examples are methyl, ethyl, monofluoromethyl, difluoromethyl, trifluoromethyl, monofluoroethyl, difluoroethyl, trifluoroethyl. In one embodiment, the solvent is a methyl group, an ethyl group, a monofluoromethyl group, a difluoromethyl group or a difluoroethyl group, in one embodiment, a monofluoromethyl group or a difluoroethyl group, in one embodiment, a difluoromethyl group or a difluoroethyl group, in one embodiment, a monofluoromethyl group, in one embodiment, a difluoromethyl group, in one embodiment, a difluoroethyl group, in one embodiment, a2, 2-difluoroethyl group.
As one mode of the "C 1-3 alkyl group which may be substituted with OH", a methyl group which may be substituted with 1 OH or an ethyl group which may be substituted with 1 to 2 OH is mentioned. For example, methyl, ethyl, hydroxymethyl, 1-hydroxyethyl, 2-hydroxyethyl, 1, 2-dihydroxyethyl. As one embodiment, methyl, ethyl or hydroxymethyl is used, as one embodiment, methyl or hydroxymethyl is used, as one embodiment, hydroxymethyl or hydroxyethyl is used, as one embodiment, hydroxymethyl is used, as one embodiment, hydroxyethyl is used.
As one mode of "C 1-3 alkyl group which may be substituted by OCH 3", there is methyl group which may be substituted by 1 OCH 3 or ethyl group which may be substituted by 1 to 2 OCH 3. For example, methyl, ethyl, methoxymethyl, 1-methoxyethyl, 2-methoxyethyl, 1, 2-dimethoxyethyl. In one embodiment, the catalyst is methoxymethyl or methoxyethyl, in one embodiment, methoxymethyl, and in one embodiment, methoxyethyl.
As one mode of "C 1-3 alkyl group which may be substituted with N (CH 3)2), methyl group which may be substituted with 1N (CH 3)2) or ethyl group which may be substituted with 1N (CH 3)2), methyl group, ethyl group, dimethylaminomethyl group or dimethylaminoethyl group, methyl group or dimethylaminomethyl group, and dimethylaminoethyl group, respectively.
As one embodiment of the "phenylene group which may be substituted with F", a phenylene group which may be substituted with 1 to 2F is used. In one embodiment, the phenylene group may be substituted with 1F, the phenylene group or the fluorophenylene group, in one embodiment, the phenylene group, in one embodiment, the 2-fluoro-1, 4-phenylene group, and in one embodiment, the 3-fluoro-1, 4-phenylene group.
"G12D mutation" means a mutation in the wild-type protein from glycine to aspartic acid at an amino acid residue corresponding to codon 12.
"G12D mutant KRAS" means KRAS having the "G12D mutation" described above.
"Pancreatic cancer" refers to a malignancy that forms in the pancreas. For example, pancreatic ductal carcinoma and pancreatic ductal adenocarcinoma, as one embodiment, pancreatic ductal carcinoma and, as one embodiment, pancreatic ductal adenocarcinoma. Furthermore, as one embodiment, metastatic pancreatic cancer, locally advanced pancreatic cancer, recurrent or refractory pancreatic cancer, and untreated and/or treatment-history pancreatic cancer are all other embodiments.
"G12D mutant KRAS positive pancreatic cancer" refers to G12D mutant KRAS positive pancreatic cancer. For example, pancreatic cancer in which KRAS G12D mutation has occurred is pancreatic cancer in which G12D mutation has a high KRAS positive rate. As one mode, G12D mutant KRAS positive pancreatic ductal carcinoma, and as one mode, G12D mutant KRAS positive pancreatic ductal adenocarcinoma.
One embodiment of the compound of formula (I) or a salt thereof in the present invention is shown below.
(1-1)
A compound or salt thereof wherein a is CR A or N, R A is H or C 1-3 alkyl.
(1-2)
A is a compound or salt thereof wherein CR A、RA is H or C 1-3 alkyl.
(1-3)
A is a compound of N or a salt thereof.
(1-4)
A is CH or N or a salt thereof.
(1-5)
A compound or salt thereof wherein a is CR A or N, R A is C 1-3 alkyl.
(1-6)
A is CH or a salt thereof.
(2-1)
R 1 is a naphthyl group which may be substituted by OH or a group selected from the group consisting of the following formula (II), formula (III) and formula (IV),
R 1a、R1b are identical or different from each other and are H, methyl, F or Cl,
A compound wherein R 1c is F, cl, methyl or ethyl, or a salt thereof.
(2-2)
R 1 is a naphthyl group which may be substituted by OH or a group selected from the group consisting of the following formula (II) and formula (III),
R 1a、R1b are identical or different from each other and are H, methyl, F or Cl,
A compound wherein R 1c is F, cl, methyl or ethyl, or a salt thereof.
(2-3)
R 1 is a group of formula (IIa) or formula (IIIa),
R 1a、R1b are identical or different from each other and are H, methyl, F or Cl compounds or salts thereof.
(2-4)
R 1 is of formula (IIa) or formula (IIIa),
R 1a、R1b are identical or different from each other, are H or F compounds or salts thereof.
(2-5)
A compound of formula (IIa) or formula (IIIa) wherein R 1 is formula (IIa), R 1a is F, and R 1b is H, or a salt thereof.
(2-6)
R 1 is a compound of the following formula (IIb) or a salt thereof.
(3-1)
A compound in which R 2 is H, halogen, C 1-3 alkyl which may be substituted, cyclopropyl or vinyl, or a salt thereof.
(3-2)
R 2 is halogen, C 1-3 alkyl, cyclopropyl or vinyl, and the C 1-3 alkyl may be substituted with a group selected from the group consisting of OH and OCH 3, or a salt thereof.
(3-3)
A compound wherein R 2 is halogen, C 1-3 alkyl, cyclopropyl or vinyl, or a salt thereof.
(3-4)
A compound wherein R 2 is cyclopropyl or a salt thereof.
(4-1)
R 3 is-P-Q or V,
P is-CH 2 -, -O-or-N (R P) -,
R P is H or C 1-3 alkyl which can be substituted,
Q is the following formula (V) or formula (VI),
V is the following formula (VII),
R Q, which are identical or different from each other, are OH or C 1-3 alkyl, and R Q is bonded only to carbon atoms as constituent atoms of rings selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
A compound wherein m is 0 to 2 or a salt thereof.
(4-2)
R3:
(I) When A is CR A, it is-P-Q,
P is-O-,
(Ii) A is-P-Q or V when A is N,
P is-O-or-N (R P) -,
R P is H or C 1-3 alkyl,
In either case of (i) and (ii) above, Q is a compound of the following formula (V) or formula (VI),
V is the following formula (VII),
R Q is the same or different from each other and is OH or methyl, R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
A compound wherein m is 0 to 2 or a salt thereof.
(4-3)
R3:
(I) When A is CH, it is-P-Q,
P is-O-,
(Ii) A is-P-Q or V when A is N,
P is-O-or-N (R P) -,
R P is H or C 1-3 alkyl,
In either case of (i) and (ii) above, Q is a compound of the following formula (V) or formula (VI),
V is the following formula (VII),
R Q is the same or different from each other and is OH or methyl, R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
A compound wherein m is 0 to 2 or a salt thereof.
(4-4)
R 3 is-P-Q,
P is-O-or-N (R P) -,
R P is H or C 1-3 alkyl,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
A compound wherein m is 0 to 2 or a salt thereof.
(4-5)
R 3 is-P-Q,
P is-O-,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
A compound wherein m is 0 to 2 or a salt thereof.
(4-6)
R 3 is-P-Q,
P is-O-,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
A compound wherein m is 0 to 1 or a salt thereof.
(4-7)
R 3 is V, and the R is H,
V is the following formula (VII),
R Q which are identical or different from each other are OH or methyl, R Q is bonded only to a carbon atom as a ring constituting atom of the piperazine ring represented by formula (VII),
A compound wherein m is 0 to 2 or a salt thereof.
(4-8)
R 3 is V, and the R is H,
V is the following formula (VII),
R Q which are identical or different from each other are OH or methyl, R Q is bonded only to a carbon atom as a ring constituting atom of the piperazine ring represented by formula (VII),
A compound wherein m is 0 to 1 or a salt thereof.
(4-9)
R 3 is-P-Q,
P is-O-or-N (R P) -,
R P is H or C 1-3 alkyl,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
A compound wherein m is 0 or 1, or a salt thereof.
(4-10)
R 3 is-P-Q,
P is-N (R P) -,
R P is H or C 1-3 alkyl,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
A compound wherein m is 0 or 1, or a salt thereof.
(4-11)
R 3 is-P-Q,
P is-N (R P) -,
R P is H or C 1-3 alkyl,
Q is the following formula (VI),
A compound wherein m is 0 or a salt thereof.
(4-12)
R 3 is-P-Q,
P is-N (R P) -,
R P is methyl or ethyl, and the amino group is a methyl or ethyl,
Q is (VI),
A compound wherein m is 0 or a salt thereof.
(5-1)
R 4 is C 1-6 alkyl which can be substituted, 4-to 6-membered saturated heterocyclic group which can be substituted and contains 1 to 2 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen, 5-membered heteroaryl which can be substituted and contains 1 to 4 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen, or 6-membered heteroaryl which can be substituted and contains 1 to 3 nitrogen atoms, or a salt thereof.
(5-2)
A compound in which R 4 is C 1-6 alkyl group which may be substituted, oxetanyl group which may be substituted, tetrahydrofuranyl group which may be substituted, tetrahydropyranyl group which may be substituted, pyrazolyl group which may be substituted, pyridinyl group which may be substituted, pyrimidinyl group which may be substituted, pyrrolidinyl group which may be substituted or piperidinyl group which may be substituted, or a salt thereof.
(5-3)
R 4 is C 1-6 alkyl which can be substituted, 4-to 6-membered saturated heterocyclic group which can be substituted containing 1 to 2 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen, pyrazolyl which can be substituted, pyridyl which can be substituted or pyrimidinyl which can be substituted, or a salt thereof.
(5-4)
R 4 is a compound which may be substituted with a group selected from the group consisting of F, OH, OCH 3、CF3、CHF2, cyclopropyl which may be substituted, pyrrolidinyl which may be substituted, and tetrahydrofuranyl which may be substituted, C 1-6 alkyl which may be substituted, tetrahydropyranyl which may be substituted, pyrazolyl which may be substituted, pyridinyl which may be substituted, or pyrimidinyl which may be substituted, or a salt thereof.
(5-5)
R 4 is a C 1-6 alkyl or tetrahydropyranyl compound or salt thereof that may be substituted with a group selected from the group consisting of F, OH, OCH 3、CF3、CHF2, cyclopropyl that may be substituted, pyrrolidinyl that may be substituted, and tetrahydrofuranyl that may be substituted.
(5-6)
R 4 is a compound which may be substituted with a group selected from the group consisting of F, OH, OCH 3、CF3、CHF2, cyclopropyl substituted with 1 methoxymethyl, pyrrolidinyl substituted with C 1-6 alkyl and tetrahydrofuranyl substituted with C 1-6 alkyl, tetrahydropyranyl substituted with C 1-6 alkyl, pyrazolyl substituted with C 1-6 alkyl, pyridinyl substituted with C 1-6 alkyl or pyrimidinyl substituted with C 1-6 alkyl, or a salt thereof.
(5-7)
R 4 is a compound or a salt thereof of C 1-6 alkyl or tetrahydropyranyl which may be substituted by a group selected from the group consisting of F, OH, OCH 3、CF3、CHF2, cyclopropyl substituted by 1 methoxymethyl, pyrrolidinyl which may be substituted by C 1-6 alkyl, and tetrahydrofuranyl which may be substituted by C 1-6 alkyl.
(5-8)
R 4 is a C 1-6 alkyl or tetrahydropyranyl compound or salt thereof that may be substituted with a group selected from the group consisting of OCH 3 and tetrahydrofuranyl.
(6-1)
R 5 is C 1-6 alkyl which can be substituted, C 3-6 cycloalkyl which can be substituted or a compound containing 1 hetero atom selected from the group consisting of oxygen, sulfur and nitrogen and 4-to 6-membered saturated heterocyclic group which can be substituted or a salt thereof.
(6-2)
A compound wherein R 5 is methyl, ethyl, isopropyl, tert-butyl, or C 3-6 cycloalkyl, or a salt thereof.
(6-3)
A compound wherein R 5 is ethyl, isopropyl, t-butyl, or C 3-6 cycloalkyl, or a salt thereof.
(6-4)
A compound wherein R 5 is isopropyl or C 3-6 cycloalkyl, or a salt thereof.
(6-5)
A compound wherein R 5 is isopropyl or a salt thereof.
(7-1)
R 6a、R6b, which are identical or different from each other, are H or C 1-6 alkyl which may be substituted, or R 6a、R6b, which may form, together with the carbon to which they are bonded, a C 3-6 cycloalkyl which may be substituted or a compound containing 1 heteroatom selected from the group consisting of oxygen, sulfur and nitrogen, which may be substituted, a 4-to 6-membered saturated heterocyclic group, or a salt thereof.
(7-2)
R 6a、R6b, which are the same or different from each other, are H or C 1-3 alkyl, and the C 1-3 alkyl may be substituted with a group selected from the group consisting of F, OH, OCH 3 and N (CH 3)2), or R 6a、R6b may form a C 3-6 cycloalkyl compound or a salt thereof together with the carbon to which they are bonded.
(7-3)
R 6a、R6b, which are the same or different from each other, are H or C 1-3 alkyl, and the C 1-3 alkyl may be substituted with a group selected from the group consisting of F, OH and N (CH 3)2), or R 6a、R6b may form a cyclopropyl compound or a salt thereof together with the carbon to which they are bonded.
(7-4)
A compound wherein R 6a is H, R 6b is C 1-3 alkyl which may be substituted with OH, or a salt thereof.
(7-5)
R 6a is H, R 6b is hydroxymethyl or a salt thereof.
(8-1)
R 7 is H, halogen, C 1-3 alkyl, -SO 2CH3、C3-6 cycloalkyl, a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 to 2 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen, a 5-membered heteroaryl group which may be substituted and contains 1 to 4 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen, or a 6-membered heteroaryl group containing 1 to 3 nitrogen atoms, or a salt thereof.
(8-2)
R 7 is H, halogen, C 1-3 alkyl, -SO 2CH3、C3-6 cycloalkyl or a group selected from the group consisting of formula (IX), formula (X), formula (XI), formula (XII), formula (XIII), formula (XIV), formula (XV), formula (XVI), formula (XVII) and formula (XVIII) below,
R 7a、R7b are identical or different from each other, are H or C 1-3 -alkyl which may be substituted by OH, or salts thereof.
(8-3)
R 7 is halogen or a group selected from the group consisting of the following formula (IX), formula (X), formula (XI), formula (XII), formula (XIII) and formula (XIV),
R 7a、R7b are identical or different from each other, are H or C 1-3 -alkyl which may be substituted by OH, or salts thereof.
(8-4)
R 7 is a group of the following formula (IX), formula (X), formula (XI) or formula (XII),
R 7a is a compound of C 1-3 alkyl which may be substituted by OH or a salt thereof.
(8-5)
R 7 is the following formula (IX) or formula (XI),
A compound wherein R 7a is C 1-3 alkyl or a salt thereof.
(8-6)
R 7 is the following formula (IX),
A compound wherein R 7a is C 1-3 alkyl or a salt thereof.
(8-7)
A compound wherein R 7 is H or a salt thereof.
(9-1)
W is a phenylene group which may be substituted or a 6-membered heteroarene diradical which may be substituted and contains 1 to 3 nitrogen atoms, or a salt thereof.
(9-2)
W is the following formula (XIX),
W 1 and W 2:
(i) W 1 is CH, W 2 is C-SO 2CH3, or
(Ii) W 1、W2 are the same or different from each other, CH, CF, CCl, CCH 3 or N, wherein R 7 is a compound of H or a salt thereof in the case of the above (i).
(9-3)
W is the following formula (XIX),
W 1、W2 are identical or different from each other, are CH or N compounds or salts thereof.
(9-4)
W is the following formula (XIX),
W 1、W2 are the same or different from each other, are CH, CF, CCl, CCH 3 or N compounds or salts thereof.
(9-5)
W is the following formula (XIX),
A compound or salt thereof wherein W 1、W2 is CH.
(10-1)
Compounds in which X is a bond, -CH 2 -, -O-, -S-, or-NR 4x-,R4x is H or C 1-3 alkyl, or salts thereof.
(10-2)
A compound wherein X is-O-or-NR 4x-、R4x is H or C 1-3 alkyl, or a salt thereof.
(10-3)
And X is-O-or-NH-or a salt thereof.
(10-4)
A compound wherein X is-O-or a salt thereof.
(11-1)
And Y is phenylene or pyridyldiyl, and the phenylene may be substituted with F.
(11-2)
And Y is phenylene or pyridyldiyl or a salt thereof.
(11-3)
And Y is a phenylene group which may be substituted with F or a salt thereof.
(12-1)
L is- (L 1-L2-L3-L4) -,
L 1、L2、L3、L4, which are identical or different from one another, are selected from the group consisting of bond, -O-, -NR L1 -, pyrrolidinediyl which may be substituted, piperidinyl which may be substituted, piperazinediyl which may be substituted, C 1-3 alkylene which may be substituted and C=O,
A compound wherein R L1 is H or C 1-3 alkyl or a salt thereof.
(12-2)
L is a bond, C 1-3 alkylene, C=O or a group selected from the group consisting of formula (XX), formula (XXI), formula (XXII), formula (XXIII), formula (XXIV) and formula (XXV) below,
R L1 is H or C 1-3 alkyl,
R L2、RL3, identical or different from one another, are H, F, OH, OCH 3 or C 1-3 alkyl which may be substituted,
R L is CH or N, and the total number of the components is,
And n is an integer of 1 to 2, or a salt thereof.
(12-3)
L is a bond, C=O or a group selected from the group consisting of the following formula (XX) and formula (XXII),
R L1 is H or C 1-3 alkyl,
R L2、RL3, identical or different from one another, are H, F, OH, OCH 3 or C 1-3 alkyl which may be substituted,
And n is an integer of 1 to 2, or a salt thereof.
(12-4)
L is a bond, C=O or a group selected from the group consisting of the following formulas (XX) -1 and (XXII) -1,
(Wherein Y * represents a bond with Y.)
R L1 is a C 1-3 alkyl group,
R L2、RL3 is H, and the hydrogen atom,
A compound wherein n is 1 or a salt thereof.
(12-5)
L is a bond, c=o, or a group selected from the group consisting of formula (XX) and formula (XXII), R L1 is C 1-3 alkyl, R L2、RL3 is H, and n is 1, or a salt thereof.
(12-6)
And L is a bond, c=o, or a group selected from the group consisting of formula (XX) -1 and formula (XXII) -1, R L1 is C 1-3 alkyl, R L2、RL3 is H, and n is 1, or a salt thereof.
(12-7)
And L is c=o or a group selected from the group consisting of formula (XX) and formula (XXII), R L1 is C 1-3 alkyl, R L2、RL3 is H, and n is 1, or a salt thereof.
(12-8)
L is c=o or a group selected from the group consisting of formula (XX) -1 and formula (XXII) -1, R L1 is C 1-3 alkyl, R L2、RL3 is H, n is 1, or a salt thereof.
(12-9)
And L is a bond or a compound of c=o or a salt thereof.
(12-10)
And L is a bond or a salt thereof.
(12-11)
And L is a compound or salt of c=o.
(13-1)
Z is NH or a 5-membered heteroarene diyl compound or salt thereof containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen.
(13-2)
Z is NH or a compound selected from the group consisting of the following formulas (XXVI), formula (XXVII), formula (XXVIII) and formula (XXIX), or a salt thereof.
(13-3)
Z is NH or a compound selected from the group consisting of the following formula (XXVI) -1, formula (XXVII) -1, formula (XXVIII) -1 and formula (XXIX) -1, or a salt thereof.
(Wherein L * represents a bond with L.)
(13-4)
Z is NH or a compound selected from the group consisting of the following formula (XXVI), formula (XXVII) and formula (XXVIII), or a salt thereof.
(13-5)
Z is NH or a compound selected from the group consisting of the following formula (XXVI) -1, formula (XXVII) -1 and formula (XXVIII) -1, or a salt thereof.
(Wherein L * represents a bond with L.)
(13-6)
Z is NH or a compound selected from the group consisting of the following formula (XXVI) and formula (XXVII) or a salt thereof.
(13-7)
Z is NH or a compound selected from the group consisting of the following formulas (XXVI) -1 and (XXVII) -1, or a salt thereof.
(Wherein L * represents a bond with L.)
(13-8)
Z is a compound selected from the group consisting of the formula (XXVI) and the formula (XXVII) or a salt thereof.
(13-9)
A compound wherein Z is a group selected from the group consisting of formula (XXVI) -1 and formula (XXVII) -1, or a salt thereof.
(13-10)
A compound wherein Z is NH or a salt thereof.
(13-11)
Z is a compound of the following formula (XXVII) or a salt thereof.
(13-12)
Z is a compound of the following formula (XXVII) -1 or a salt thereof.
(Wherein L * represents a bond with L.)
(14-1)
Y-L-Z is a compound of the following formula (VIII) or a salt thereof.
(14-2)
Y-L-Z is a compound of the following formula (VIII) -1 or a salt thereof.
(Wherein O-CH 2 * represents a bond to a carbon atom of O-CH 2 bonded to Y-L-Z.)
(15)
A compound or a salt thereof which is a combination of any two or more of the above-mentioned modes (1-1) to (14-2) and is not contradictory.
The combination of (15) above is specifically exemplified by the following means.
(15-1)
A compound of formula (I) or a salt thereof.
(In the formula (I),
A is CR A or N, and the total number of the components is N,
R A is H or C 1-3 alkyl,
R 1 is a naphthyl group which may be substituted by OH or a group selected from the group consisting of the following formula (II), formula (III) and formula (IV),
R 1a、R1b are identical or different from each other and are H, methyl, F or Cl,
R 1c is F, cl, methyl or ethyl,
R 2 is H, halogen, optionally substituted C 1-3 alkyl, cyclopropyl or vinyl,
R 3 is-P-Q or V,
P is-CH 2 -, -O-or-N (R P) -,
R P is H or C 1-3 alkyl which can be substituted,
Q is the following formula (V) or formula (VI),
V is the following formula (VII),
R Q, which are identical or different from each other, are OH or C 1-3 alkyl, and R Q is bonded only to carbon atoms as constituent atoms of rings selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
M is a number from 0 to 2,
R 4 is an optionally substituted C 1-6 alkyl group, an optionally substituted 4-to 6-membered saturated heterocyclic group containing 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen, an optionally substituted 5-membered heteroaryl group containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen or an optionally substituted 6-membered heteroaryl group containing 1 to 3 nitrogen atoms,
R 5 is C 1-6 alkyl which may be substituted, C 3-6 cycloalkyl which may be substituted or a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 hetero atom selected from the group consisting of oxygen, sulfur and nitrogen,
R 6a、R6b, identical or different from one another, is H or C 1-6 -alkyl which may be substituted, or R 6a、R6b together with the carbon to which they are bonded may form C 3-6 -cycloalkyl which may be substituted or a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 heteroatom selected from the group consisting of oxygen, sulfur and nitrogen,
R 7 is H, halogen, C 1-3 alkyl, -SO 2CH3、C3-6 cycloalkyl, an optionally substituted 4-to 6-membered saturated heterocyclic group containing 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen, an optionally substituted 5-membered heteroaryl group containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen or a 6-membered heteroaryl group containing 1 to 3 nitrogen atoms,
W is optionally substituted phenylene or optionally substituted 6-membered heteroarene diyl having 1 to 3 nitrogen atoms,
X is a bond, -CH 2 -, -O-, -S-, or-NR 4x -,
R 4x is H or C 1-3 alkyl,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is- (L 1-L2-L3-L4) -,
L 1、L2、L3、L4, which are identical or different from one another, are selected from the group consisting of bond, -O-, -NR L1 -, pyrrolidinediyl which may be substituted, piperidinyl which may be substituted, piperazinediyl which may be substituted, C 1-3 alkylene which may be substituted and C=O,
R L1 is H or C 1-3 alkyl,
Z is NH or a 5-membered heteroarene diyl containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulphur and nitrogen,
Or Y-L-Z is the following formula (VIII). )
(15-2)
The compound according to the above (15-1), wherein,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is a bond, C 1-3 alkylene, C=O or a group selected from the group consisting of formula (XX), formula (XXI), formula (XXII), formula (XXIII), formula (XXIV) and formula (XXV) below,
R L1 is H or C 1-3 alkyl,
R L2、RL3, identical or different from one another, are H, F, OH, OCH 3 or C 1-3 alkyl which may be substituted,
R L is CH or N, and the total number of the components is,
N is an integer of 1 to 2,
Z is NH or a group selected from the group consisting of the following formulae (XXVI), formula (XXVII), formula (XXVIII) and formula (XXIX),
Or Y-L-Z is the following formula (VIII).
(15-3)
The compound according to the above (15-2), wherein,
R 1 is a group of formula (IIa) or formula (IIIa),
R 1a、R1b are identical or different from each other and are H, methyl, F or Cl,
R3:
(I) When A is CR A, it is-P-Q,
P is-O-,
(Ii) A is-P-Q or V when A is N,
P is-O-or-N (R P) -,
R P is H or C 1-3 alkyl,
In either case of (i) and (ii) above, Q is a compound of the following formula (V) or formula (VI),
V is the following formula (VII),
R Q is the same or different from each other and is OH or methyl, R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
M is a number from 0 to 2,
R 4 is C 1-6 alkyl which may be substituted, 4-to 6-membered saturated heterocyclic group which may be substituted containing 1 to 2 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen, pyrazolyl which may be substituted, pyridinyl which may be substituted or pyrimidinyl which may be substituted,
R 5 is methyl, ethyl, isopropyl, tert-butyl or C 3-6 cycloalkyl,
R 6a、R6b, which are identical or different from each other, are H or C 1-3 alkyl, which C 1-3 alkyl may be substituted by a group selected from the group consisting of F, OH, OCH 3 and N (CH 3)2), or R 6a、R6b may form, together with the carbon to which they are bonded, a C 3-6 cycloalkyl group,
R 7 is H, halogen, C 1-3 alkyl, -SO 2CH3、C3-6 cycloalkyl or a group selected from the group consisting of formula (IX), formula (X), formula (XI), formula (XII), formula (XIII), formula (XIV), formula (XV), formula (XVI) formula (XVII) and formula (XVIII) below,
R 7a、R7b, identical or different from one another, are H or C 1-3 alkyl which may be substituted by OH,
W is the following formula (XIX),
W 1 and W 2:
(i) W 1 is CH, W 2 is C-SO 2CH3, or
(Ii) W 1、W2 are identical or different from each other and are CH, CF, CCl, CCH 3 or N,
Wherein, when W 1 is CH and W 2 is C-SO 2CH3, R 7 is H,
X is-O-or-NR 4x -,
R 4x is H or C 1-3 alkyl,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is a bond, C=O or a group selected from the group consisting of the following formula (XX) and formula (XXII),
R L1 is H or C 1-3 alkyl,
R L2、RL3, identical or different from one another, are H, F, OH, OCH 3 or C 1-3 alkyl which may be substituted,
N is an integer of 1 to 2,
Z is NH or a group selected from the group consisting of the following formulae (XXVI), formula (XXVII), formula (XXVIII) and formula (XXIX),
Or Y-L-Z is the following formula (VIII).
(15-4)
The compound according to the above (15-3), wherein,
A is CH or N, and the A is H or N,
R 2 is halogen, C 1-3 alkyl, cyclopropyl or vinyl, the C 1-3 alkyl may be substituted with a group selected from the group consisting of OH and OCH 3,
R3:
(I) When A is CH, it is-P-Q,
P is-O-,
(Ii) A is-P-Q or V when A is N,
P is-O-or-N (R P) -,
R P is H or C 1-3 alkyl,
In either case of (i) and (ii) above, Q is a compound of the following formula (V) or formula (VI),
V is the following formula (VII),
R Q is the same or different from each other and is OH or methyl, R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
M is a number from 0 to 2,
R 4 is C 1-6 alkyl or tetrahydropyranyl which may be substituted with a group selected from the group consisting of F, OH, OCH 3、CF3、CHF2, cyclopropyl which may be substituted, pyrrolidinyl which may be substituted and tetrahydrofuranyl which may be substituted,
R 7 is halogen or a group selected from the group consisting of the following formula (IX), formula (X), formula (XI), formula (XII), formula (XIII) and formula (XIV),
R 7a、R7b, identical or different from one another, are H or C 1-3 alkyl which may be substituted by OH,
W is the following formula (XIX),
W 1、W2 are identical or different from each other and are CH or N,
X is-O-or-NH-,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is a bond and is preferably a bond,
Z is the following formula (XXVII).
(15-5)
The compound according to the above (15-3), wherein,
A is N, and the number of the A is N,
R 3 is-P-Q,
P is-O-or-N (R P) -,
R P is H or C 1-3 alkyl,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
M is a number from 0 to 2,
W is the following formula (XIX),
W 1、W2 are identical or different from each other and are CH, CF, CCl, CCH 3 or N,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is a bond, C=O or a group selected from the group consisting of the following formula (XX) and formula (XXII),
R L1 is H or C 1-3 alkyl,
R L2、RL3, identical or different from one another, are H, F, OH, OCH 3 or C 1-3 alkyl which may be substituted,
N is an integer of 1 to 2,
Z is NH or a group selected from the group consisting of the following formulas (XXVI), formula (XXVII), formula (XXVIII) and formula (XXIX).
(15-6)
The compound according to the above (15-3), wherein,
A is CH, and the A is CH,
R 3 is-P-Q,
P is-O-,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
M is a number from 0 to 2,
W is the following formula (XIX),
W 1、W2 are identical or different from each other and are CH, CF, CCl, CCH 3 or N,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is a bond, C=O or a group selected from the group consisting of the following formula (XX) and formula (XXII),
R L1 is H or C 1-3 alkyl,
R L2、RL3, identical or different from one another, are H, F, OH, OCH 3 or C 1-3 alkyl which may be substituted,
N is an integer of 1 to 2,
Z is NH or a group selected from the group consisting of the following formulas (XXVI), formula (XXVII), formula (XXVIII) and formula (XXIX).
(15-7)
The compound according to the above (15-3), wherein,
A is N, and the number of the A is N,
R 3 is V, and the R is H,
V is the following formula (VII),
R Q which are identical or different from each other are OH or methyl, R Q is bonded only to a carbon atom as a ring constituting atom of the piperazine ring represented by formula (VII),
M is a number from 0 to 2,
W is the following formula (XIX),
W 1、W2 are identical or different from each other and are CH, CF, CCl, CCH 3 or N,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is a bond, C=O or a group selected from the group consisting of the following formula (XX) and formula (XXII),
R L1 is H or C 1-3 alkyl,
R L2、RL3, identical or different from one another, are H, F, OH, OCH 3 or C 1-3 alkyl which may be substituted,
N is an integer of 1 to 2,
Z is NH or a group selected from the group consisting of the following formulas (XXVI), formula (XXVII), formula (XXVIII) and formula (XXIX).
(15-8)
The compound according to the above (15-4), wherein,
A is N, and the number of the A is N,
R 1 is the following formula (IIb),
R 3 is-P-Q,
P is-O-or-N (R P) -,
R P is H or C 1-3 alkyl,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
M is 0 or 1 and the number of the groups,
X is-O-.
(15-9)
The compound according to the above (15-4), wherein,
A is CH, and the A is CH,
R 1 is the following formula (IIb),
R 3 is-P-Q,
P is-O-,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
M is 0 or 1 and the number of the groups,
X is-O-.
(15-10)
The compound according to the above (15-4), wherein,
A is N, and the number of the A is N,
R 1 is the following formula (IIb),
R 3 is V, and the R is H,
V is the following formula (VII),
R Q which are identical or different from each other are OH or methyl, R Q is bonded only to a carbon atom as a ring constituting atom of the piperazine ring represented by formula (VII),
M is 0 or 1 and the number of the groups,
X is-O-.
(15-11)
The compound according to the above (15-8), wherein,
R 2 is cyclopropyl, and the amino group is a cyclopropyl group,
R 3 is-P-Q,
P is-N (R P) -,
R P is H or C 1-3 alkyl,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
M is 0 or 1 and the number of the groups,
R 4 is C 1-6 alkyl or tetrahydropyranyl which may be substituted by a group selected from the group consisting of OCH 3 and tetrahydrofuranyl,
R 5 is isopropyl group, and the amino group is a hydroxyl group,
R 6a is H, R 6b is C 1-3 alkyl which can be substituted by OH,
R 7 is a group of the following formula (IX), formula (X), formula (XI) or formula (XII),
R 7a is C 1-3 alkyl which may be substituted by OH,
W is the following formula (XIX),
W 1、W2 is CH, and the two are all CH,
Y is phenylene which may be substituted by F.
(15-12)
The compound according to the above (15-8), wherein,
R 2 is cyclopropyl, and the amino group is a cyclopropyl group,
R 3 is-P-Q,
P is-N (R P) -,
R P is methyl or ethyl, and the amino group is a methyl or ethyl,
Q is (VI),
M is 0 and is the number of the three-dimensional space,
R 5 is isopropyl group, and the amino group is a hydroxyl group,
R 6a is H, R 6b is hydroxymethyl,
R 7 is the following formula (IX) or formula (XI),
R 7a is a C 1-3 alkyl group,
W is the following formula (XIX),
W 1、W2 is CH, and the two are all CH,
Y is phenylene which may be substituted by F.
(15-13)
The compound according to the above (15-12), wherein,
R 7 is the following formula (IX),
R 7a is C 1-3 alkyl.
One embodiment of the compound of formula (IA) or a salt thereof in the present invention is shown below.
(16-1)
R 1 is a naphthyl group which may be substituted by OH or a group selected from the group consisting of the following formula (II), formula (III) and formula (IV),
R 1a、R1b are identical or different from each other and are H, methyl, F or Cl,
A compound wherein R 1c is F, cl, methyl or ethyl, or a salt thereof.
(16-2)
R 1 is a group of formula (IIa) or formula (IIIa),
R 1a、R1b are identical or different from each other and are H, methyl, F or Cl compounds or salts thereof.
(16-3)
R 1 is a compound of the following formula (IIb) or a salt thereof.
(17-1)
E is CH or N, and the R is H,
G is CR 2 or N, R 2 is H, halogen, C 1-3 alkyl which may be substituted, cyclopropyl or vinyl,
A is CR A or N, R A is H or C 1-3 alkyl,
R 3 is-P-Q or V,
P is-CH 2 -, -O-or-N (R P) -,
R P is H or C 1-3 alkyl which can be substituted,
Q is the following formula (V) or formula (VI),
V is the following formula (VII),
R Q, which are identical or different from each other, are OH or C 1-3 alkyl, and R Q is bonded only to carbon atoms as constituent atoms of rings selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
In addition, (i) when A is CR A, or (ii) when A is N and E or G is N,
V may be a saturated or unsaturated 7-to 8-membered bridged heterocyclic group other than the formula (VII),
A compound wherein m is 0 to 2 or a salt thereof.
(17-2)
E is CH, and the E is CH,
G is CR 2,R2 is H, halogen, C 1-3 alkyl which may be substituted, cyclopropyl or vinyl,
A is CR A,RA is H or C 1-3 alkyl,
R 3 is-P-Q or V,
P is-CH 2 -, -O-or-N (R P) -,
R P is H or C 1-3 alkyl which can be substituted,
Q is the following formula (V) or formula (VI),
V is saturated or unsaturated 7-to 8-membered bridged heterocyclic group,
A compound wherein m is 0 to 2 or a salt thereof.
(17-3)
E is CH, and the E is CH,
G is CR 2,R2 and is cyclopropyl,
A is CR A,RA is H or C 1-3 alkyl,
R 3 is-P-Q or V,
P is-CH 2 -, -O-or-N (R P) -,
R P is H or C 1-3 alkyl which can be substituted,
Q is the following formula (V) or formula (VI),
V is saturated or unsaturated 7-to 8-membered bridged heterocyclic group,
A compound wherein m is 0 to 2 or a salt thereof.
(17-4)
E is CH, and the E is CH,
G is CR 2,R2 is H, halogen, C 1-3 alkyl which may be substituted, cyclopropyl or vinyl,
A is CR A,RA and H is H,
R 3 is V, and the R is H,
V is a saturated or unsaturated 7-to 8-membered bridged heterocyclic compound or a salt thereof.
(17-5)
E is CH, and the E is CH,
G is CR 2,R2 and is cyclopropyl,
A is CR A,RA and H is H,
R 3 is V, and the R is H,
V is a saturated or unsaturated 7-to 8-membered bridged heterocyclic compound or a salt thereof.
(17-6)
E is N, and the number of the E is N,
G is CR 2,R2 is H, halogen, C 1-3 alkyl which may be substituted, cyclopropyl or vinyl,
A is N, and the number of the A is N,
R 3 is-P-Q or V,
P is-CH 2 -, -O-or-N (R P) -,
R P is H or C 1-3 alkyl which can be substituted,
Q is the following formula (V) or formula (VI),
V is a saturated or unsaturated 7-to 8-membered bridged heterocyclic group of the following formula (VII),
R Q, which are identical or different from each other, are OH or C 1-3 alkyl, and R Q is bonded only to carbon atoms as constituent atoms of rings selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
A compound wherein m is 0 to 2 or a salt thereof.
(17-7)
E is N, and the number of the E is N,
G is CR 2,R2 and is cyclopropyl,
A is N, and the number of the A is N,
R 3 is-P-Q or V,
P is-CH 2 -, -O-or-N (R P) -,
R P is H or C 1-3 alkyl which can be substituted,
Q is the following formula (V) or formula (VI),
V is a saturated or unsaturated 7-to 8-membered bridged heterocyclic group of the following formula (VII),
R Q, which are identical or different from each other, are OH or C 1-3 alkyl, and R Q is bonded only to carbon atoms as constituent atoms of rings selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
A compound wherein m is 0 to 2 or a salt thereof.
(18-1)
R 4 is C 1-6 alkyl which can be substituted, 4-to 6-membered saturated heterocyclic group which can be substituted and contains 1 to 2 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen, 5-membered heteroaryl which can be substituted and contains 1 to 4 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen, or 6-membered heteroaryl which can be substituted and contains 1 to 3 nitrogen atoms, or a salt thereof.
(18-2)
R 4 is a C 1-6 alkyl or tetrahydropyranyl compound or salt thereof that may be substituted with a group selected from the group consisting of OCH 3 and tetrahydrofuranyl.
(19-1)
R 5 is C 1-6 alkyl which can be substituted, C 3-6 cycloalkyl which can be substituted or a compound containing 1 hetero atom selected from the group consisting of oxygen, sulfur and nitrogen and 4-to 6-membered saturated heterocyclic group which can be substituted or a salt thereof.
(19-2)
A compound wherein R 5 is isopropyl or C 3-6 cycloalkyl, or a salt thereof.
(20-1)
R 6a、R6b, which are identical or different from each other, are H or C 1-6 alkyl which may be substituted, or R 6a、R6b, which may form, together with the carbon to which they are bonded, a C 3-6 cycloalkyl which may be substituted or a compound containing 1 heteroatom selected from the group consisting of oxygen, sulfur and nitrogen, which may be substituted, a 4-to 6-membered saturated heterocyclic group, or a salt thereof.
(20-2)
A compound wherein R 6a is H, R 6b is C 1-3 alkyl which may be substituted with OH, or a salt thereof.
(21-1)
R 7 is H, halogen, C 1-3 alkyl, -SO 2CH3、C3-6 cycloalkyl, a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 to 2 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen, a 5-membered heteroaryl group which may be substituted and contains 1 to 4 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen, or a 6-membered heteroaryl group containing 1 to 3 nitrogen atoms, or a salt thereof.
(21-2)
R 7 is a group of the following formula (IX), formula (X), formula (XI) or formula (XII),
R 7a is a compound of C 1-3 alkyl which may be substituted by OH or a salt thereof.
(22-1)
L Y is-O- (optionally substituted C 1-3 alkylene) -, S- (C 1-3 alkylene which may be substituted) -, -SO 2 - (optionally substituted C 1-3 alkylene) -, -NR Y - (optionally substituted C 1-3 alkylene) -, and- (C 1-3 alkylene which may be substituted) -O-, C 1-3 alkylene which may be substituted) -S-, C 1-3 alkylene which may be substituted) -SO 2 -, C 1-3 alkylene which may be substituted) -NR Y -,
A compound wherein R Y is H or C 1-3 alkyl or a salt thereof.
(22-2)
L Y is-S- (optionally substituted C 1-3 alkylene) -, -SO 2 - (optionally substituted C 1-3 alkylene) -, and-NR Y - (optionally substituted C 1-3 alkylene) -, - (C 1-3 alkylene which may be substituted) -O-, - (C 1-3 alkylene which may be substituted) -S-, - (C 1-3 alkylene which may be substituted) -SO 2 -, - (C 1-3 alkylene which may be substituted) -NR Y -,
A compound wherein R Y is H or C 1-3 alkyl or a salt thereof.
(22-3)
L Y is-S- (optionally substituted C 1-3 alkylene) -, -SO 2 - (optionally substituted C 1-3 alkylene) -, and-NR Y - (optionally substituted C 1-3 alkylene) -, - (C 1-3 alkylene which may be substituted) -S-, C 1-3 alkylene which may be substituted) -SO 2 -, C 1-3 alkylene which may be substituted) -NR Y -,
A compound wherein R Y is H or C 1-3 alkyl or a salt thereof.
(22-4)
L Y is a compound of the formula-O- (optionally substituted C 1-3 alkylene) -or a salt thereof.
(23-1)
R P1 is OH or F, and the total number of the components is H,
R P2a is H or F,
A compound wherein R P2b is H or a salt thereof.
(23-2)
R P1 is F, and the total number of the components is F,
R P2a is H or F,
A compound wherein R P2b is H or a salt thereof.
(23-3)
R P1 is OH, and the hydroxyl radical is,
R P2a is H or F,
A compound wherein R P2b is H or a salt thereof.
(23-4)
R P1 is OH, and the hydroxyl radical is,
A compound or salt thereof wherein R P2a and R P2b are both H.
(24-1)
W is a phenylene group which may be substituted or a 6-membered heteroarene diradical which may be substituted and contains 1 to 3 nitrogen atoms, or a salt thereof.
(24-2)
W is the following formula (XIX),
A compound or salt thereof wherein W 1、W2 is CH.
(25-1)
X is a bond, -CH 2 -, -O-, -S-, or-NR 4x -,
A compound wherein R 4x is H or C 1-3 alkyl or a salt thereof.
(25-2)
A compound wherein X is-O-or a salt thereof.
(26-1)
And Y is phenylene or pyridyldiyl, and the phenylene may be substituted with F.
(26-2)
And Y is a phenylene group which may be substituted with F or a salt thereof.
(27-1)
L is- (L 1-L2-L3-L4) -,
L 1、L2、L3、L4, which are identical or different from one another, are selected from the group consisting of bond, -O-, -NR L1 -, pyrrolidinediyl which may be substituted, piperidinyl which may be substituted, piperazinediyl which may be substituted, C 1-3 alkylene which may be substituted and C=O,
A compound wherein R L1 is H or C 1-3 alkyl or a salt thereof.
(27-2)
And L is a bond or a salt thereof.
(28-1)
Z is NH or a 5-membered heteroarene diyl compound or salt thereof containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen.
(28-2)
Z is a compound of the following formula (XXVII) -1 or a salt thereof.
(Wherein L * represents a bond with L.)
(29-1)
Y-L-Z is a compound of the following formula (VIII) or a salt thereof.
(30)
A compound or a salt thereof which is a combination of any two or more of the above-mentioned modes (16-1) to (29-1) and is not contradictory.
The combination of the above (30) is specifically exemplified by the following modes.
(30-1)
A compound of formula (IA) or a salt thereof.
(In the formula (I),
A is CR A or N, and the total number of the components is N,
R A is H or C 1-3 alkyl,
E is CH or N, and the R is H,
G is CR 2 or N, and the total number of the components is N,
R 1 is a naphthyl group which may be substituted by OH or a group selected from the group consisting of the following formula (II), formula (III) and formula (IV),
R 1a、R1b are identical or different from each other and are H, methyl, F or Cl,
R 1c is F, cl, methyl or ethyl,
R 2 is H, halogen, optionally substituted C 1-3 alkyl, cyclopropyl or vinyl,
R 3 is-P-Q or V,
P is-CH 2 -, -O-or-N (R P) -,
R P is H or C 1-3 alkyl which can be substituted,
Q is the following formula (V) or formula (VI),
V is the following formula (VII),
R Q, which are identical or different from each other, are OH or C 1-3 alkyl, and R Q is bonded only to carbon atoms as constituent atoms of rings selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
In addition, (i) when A is CR A, or (ii) when A is N and E or G is N,
V may be a saturated or unsaturated 7-to 8-membered bridged heterocyclic group other than the formula (VII),
M is a number from 0 to 2,
R 4 is an optionally substituted C 1-6 alkyl group, an optionally substituted 4-to 6-membered saturated heterocyclic group containing 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen, an optionally substituted 5-membered heteroaryl group containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen or an optionally substituted 6-membered heteroaryl group containing 1 to 3 nitrogen atoms,
R 5 is C 1-6 alkyl which may be substituted, C 3-6 cycloalkyl which may be substituted or a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 hetero atom selected from the group consisting of oxygen, sulfur and nitrogen,
R 6a、R6b, identical or different from one another, is H or C 1-6 -alkyl which may be substituted, or R 6a、R6b together with the carbon to which they are bonded may form C 3-6 -cycloalkyl which may be substituted or a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 heteroatom selected from the group consisting of oxygen, sulfur and nitrogen,
R 7 is H, halogen, C 1-3 alkyl, -SO 2CH3、C3-6 cycloalkyl, an optionally substituted 4-to 6-membered saturated heterocyclic group containing 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen, an optionally substituted 5-membered heteroaryl group containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen or a 6-membered heteroaryl group containing 1 to 3 nitrogen atoms,
L Y is-O- (optionally substituted C 1-3 alkylene) -, S- (C 1-3 alkylene which may be substituted) -, -SO 2 - (optionally substituted C 1-3 alkylene) -, -NR Y - (optionally substituted C 1-3 alkylene) -, and- (C 1-3 alkylene which may be substituted) -O-, C 1-3 alkylene which may be substituted) -S-, C 1-3 alkylene which may be substituted) -SO 2 -, C 1-3 alkylene which may be substituted) -NR Y -,
R Y is H or C 1-3 alkyl,
R P1 is OH or F, and the total number of the components is H,
R P2a is H or F,
R P2b is H, and the hydrogen atom,
W is optionally substituted phenylene or optionally substituted 6-membered heteroarene diyl having 1 to 3 nitrogen atoms,
X is a bond, -CH 2 -, -O-, -S-, or-NR 4x -,
R 4x is H or C 1-3 alkyl,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is- (L 1-L2-L3-L4) -,
L 1、L2、L3、L4, which are identical or different from one another, are selected from the group consisting of bond, -O-, -NR L1 -, pyrrolidinediyl which may be substituted, piperidinyl which may be substituted, piperazinediyl which may be substituted, C 1-3 alkylene which may be substituted and C=O,
R L1 is H or C 1-3 alkyl,
Z is NH or a 5-membered heteroarene diyl containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulphur and nitrogen,
Or Y-L-Z is the following formula (VIII). )
As examples of specific compounds included in the present invention, the following compounds are given as one embodiment.
A compound selected from the group consisting of:
(4R) -1- [ (2S) -2- {4- [4- ({ [ 6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } -2- { [ (2R) -oxolan-2-yl ] methoxy } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ 6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } -2- { [ (2S) -oxolan-2-yl ] methoxy } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ 6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ 6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -N- { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } -4-hydroxy-L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ 6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) -2-fluorophenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- { (2S) -2- [4- (4- { [ (6-cyclopropyl-4- { ethyl [ (3S) -pyrrolidin-3-yl ] amino } -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] quinazolin-8-yl) oxy ] methyl } phenyl) -1H-1,2, 3-triazol-1-yl ] -3-methylbutanoyl } -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide, and
(4R) -1- [ (2S) -2- {4- [4- ({ [ 6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3R) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide.
As examples of specific compounds included in the present invention, the following compounds are given as one embodiment.
A compound selected from the group consisting of:
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } -2- { [ (2R) -oxopyrrolidin-2-yl ] methoxy } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } -2- { [ (2S) -oxopyrrolidin-2-yl ] methoxy } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -N- { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } -4-hydroxy-L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) -2-fluorophenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- (4- {4- [ ({ (7M) -6-cyclopropyl-4- { ethyl [ (3S) -pyrrolidin-3-yl ] amino } -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] quinazolin-8-yl } oxy) methyl ] phenyl } -1H-1,2, 3-triazol-1-yl) -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide, and
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3R) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide.
As examples of specific compounds included in the present invention, the following compounds are given as one embodiment.
A compound selected from the group consisting of:
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7P) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } -2- { [ (2R) -oxopyrrolidin-2-yl ] methoxy } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7P) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } -2- { [ (2S) -oxopyrrolidin-2-yl ] methoxy } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7P) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7P) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -N- { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } -4-hydroxy-L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7P) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) -2-fluorophenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- (4- {4- [ ({ (7P) -6-cyclopropyl-4- { ethyl [ (3S) -pyrrolidin-3-yl ] amino } -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] quinazolin-8-yl } oxy) methyl ] phenyl } -1H-1,2, 3-triazol-1-yl) -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide, and
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7P) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3R) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide.
In the compounds of formula (I) and formula (IA), depending on the kind of substituents, tautomers, geometrical isomers may exist. In the present specification, the compounds of formula (I) and formula (IA) are sometimes described as only one form of isomer, but the present invention also includes other isomers, isolated products of isomers, or mixtures thereof.
In addition, the compounds of formula (I) and (IA) may have asymmetric carbon atoms and asymmetric axes, and diastereoisomers based thereon may be present. The invention also includes isolated products of diastereomers of the compounds of formula (I) and formula (IA) or mixtures thereof.
Furthermore, the present invention also includes pharmaceutically acceptable prodrugs of the compounds of formula (I) and formula (IA). Pharmaceutically acceptable prodrugs refer to compounds having groups that are capable of being converted to amino, hydroxyl, carboxyl, etc. groups by solvolysis or under physiological conditions. Examples of the prodrug-forming groups include those described in prog.med.,1985,5, p.2157-2161, "development of medicines", volume 7 molecular design, book store, 1990, and p.163-198.
The salts of the compounds of the formulae (I) and (IA) are pharmaceutically acceptable salts of the compounds of the formulae (I) and (IA), and may form acid addition salts or salts with bases depending on the type of substituent. Examples of the salts include those described in P.Heinrich Stahl, handbook of Pharmaceutical Salts Properties, selection, and Use, wiley-VCH, 2008. Specifically, there may be mentioned: acid addition salts with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, nitric acid, phosphoric acid, etc., organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, mandelic acid, tartaric acid, dibenzoyl tartaric acid, xylene formyl tartaric acid, citric acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, aspartic acid, glutamic acid, etc.; salts with inorganic metals such as sodium, potassium, magnesium, calcium, and aluminum; salts with organic bases such as methylamine, ethylamine, ethanolamine, etc.; salts with various amino acids and amino acid derivatives such as acetylleucine, lysine, ornithine, etc.; ammonium salts, and the like.
Furthermore, the present invention also includes various hydrates, solvates and polymorphs of the compounds of formula (I) and formula (IA) and salts thereof.
In addition, the present invention includes all compounds of formula (I) and formula (IA) or salts thereof labeled with more than 1 pharmaceutically acceptable radioisotope, either radioactive or non-radioactive. Examples of preferred isotopes for isotopic labeling of the compounds of the present invention include isotopes of hydrogen (2 H and 3 H, etc.), carbon (11C、13 C and 14 C, etc.), nitrogen (13 N and 15 N, etc.), oxygen (15O、17 O and 18 O, etc.), fluorine (18 F, etc.), chlorine (36 Cl, etc.), iodine (123 I and 125 I, etc.), sulfur (35 S, etc.).
The isotopically labeled compounds of the present application can be used for studies such as tissue distribution studies of drugs and/or substrates. For example, from the viewpoint of ease of labeling and easiness of detection, a radioisotope such as tritium (3 H) or carbon 14 (14 C) can be used for this purpose.
Substitution to heavier isotopes, such as substitution of hydrogen to deuterium (2 H), is sometimes therapeutically advantageous (e.g., increasing in vivo half-life, reducing the necessary amount, reducing drug interactions) due to increased metabolic stability.
Substitution to positron emitting isotopes (11C、18F、15 O, 13 N, etc.) may be used in Positron Emission Tomography (PET) experiments for the purpose of assaying substrate receptor occupancy.
The isotopically labeled compounds of the present invention can be generally prepared by conventional methods known to those skilled in the art or by using an isotopically labeled suitable reagent in place of an unlabeled reagent, by the same production method as in examples or production examples, etc.
(Manufacturing method)
The compounds of formula (I) and formula (IA) and salts thereof can be produced by using various known synthetic methods based on the characteristics of the basic structure or the kind of substituents. In this case, depending on the kind of the functional group, the functional group may be replaced with an appropriate protecting group (a group that can be easily converted into the functional group) in advance at a stage from the raw material to the intermediate, which is effective in terms of manufacturing technology. Examples of such protecting groups include those described in p.g.m.wuts and t.w.greene, "Greene's Protective Groups in Organic Synthesis", 5 th edition, john Wiley & Sons inc., 2014, and the like, and may be appropriately selected and used according to the reaction conditions. In such a method, the desired compound can be obtained by introducing the protecting group and then reacting, and removing the protecting group as necessary.
In addition, prodrugs of compounds of formula (I) and formula (IA) can be produced by introducing a specific group at the stage from the starting material to the intermediate or further reacting the obtained compounds of formula (I) and formula (IA) in the same manner as the protecting group described above. The reaction may be carried out by applying a method known to those skilled in the art such as usual esterification, amidation, dehydration, etc.
Hereinafter, a representative production method of the compounds of the formulas (I) and (IA) will be described. The manufacturing methods may also be performed with reference to the references attached in this description. The manufacturing method of the present invention is not limited to the examples shown below. The compound of formula (IA) can be obtained by the same method as the typical production method of the compound of formula (I) using a starting compound corresponding to the compound of formula (IA).
In the present specification, the following abbreviations may be used.
DMF: n, N-dimethylformamide, DMAc: n, N-dimethylacetamide, THF: tetrahydrofuran, meCN: acetonitrile, meOH: methanol, etOH: ethanol, iPrOH: isopropanol, tBuOH: tertiary butanol, DOX:1, 4-dioxane, DMSO: dimethyl sulfoxide, TEA: triethylamine, DIPEA: n, N-diisopropylethylamine, tBuOK: potassium tert-butoxide, pdCl 2(dppf)·CH2Cl2: [1,1' -bis (diphenylphosphino) ferrocene ] palladium (II) dichloride dichloromethane adduct, pd/C: palladium on carbon, pyBOP: (Benzotriazol-1-yloxy) tripyrrolidinylHexafluorophosphoric acid, NMP: n-methyl-2-pyrrolidone.
(Manufacturing method 1)
( Wherein PG 1 represents a protecting group for NH contained in R 3, R 31 represents a divalent group formed by removing H from NH contained in R 3, PG 2 represents a protecting group or a hydrogen atom for NH or OH contained in R 1, and R 11 represents a divalent group formed by removing H from NH contained in R 1. The same applies to the followings )
The compound of formula (I) can be obtained by subjecting compound (1) to a deprotection reaction. Examples of the protecting group which can be deprotected under acidic conditions include t-butoxycarbonyl, triphenylmethyl, tetrahydro-2H-pyran-2-yl, methoxymethyl, dimethylmethane diyl, t-butylsulfinyl and the like.
The reaction is carried out by stirring for usually 0.1 hour to 5 days under cooling to heating reflux. Examples of the solvent used herein are not particularly limited, and include: alcohols such as MeOH and EtOH; halogenated hydrocarbons such as methylene chloride, 1, 2-dichloroethane or chloroform; ethers such as diethyl ether, THF, DOX, dimethoxyethane, etc.; DMF, DMSO, meCN or water; and mixtures of these. Examples of the deprotecting reagent include, but are not particularly limited to, acids such as hydrogen chloride (DOX solution), trifluoroacetic acid, methanesulfonic acid, phosphoric acid, and p-toluenesulfonic acid.
Deprotection can also be achieved by catalytic hydrogenation by selection of protecting groups. Examples of the protecting group include benzyl, p-methoxybenzyl, and benzyloxycarbonyl. Alternatively, deprotection may be carried out using a fluoride ion source such as tetra-n-butylammonium fluoride. Examples of the protecting group include a tert-butyl (dimethyl) silyl group, a (trimethylsilyl) ethoxymethyl group, and the like. Examples of the protecting group which can be deprotected under alkaline conditions include an acetyl group, a trifluoroacetyl group, and a benzoyl group. Alternatively, as PG 1、PG2, deprotection may be performed in stages by selecting protecting groups capable of deprotection under different deprotection conditions.
As references for this reaction, for example, the following references can be referred to.
P.g.m.wuts and t.w.greene, "Greene's Protective Groups in Organic Synthesis", 5 th edition, john Wiley & Sons inc., 2014
When the compound (1) as a starting material has an axial asymmetry, the present reaction may be carried out using a stereoisomer obtained by once separating the compound (1).
The hydrochloride of the compound of formula (I) can be obtained by applying the following operation as a salt-forming reaction to the compound of formula (I).
The compound of formula (I), which is considered to form a salt with hydrochloric acid in terms of its chemical structural characteristics, was dissolved in CH 2Cl2 and MeOH, hydrogen chloride (4M DOX solution, 10 equivalents) was added under ice-cooling, and stirred under ice-cooling for 30 minutes. The reaction mixture was concentrated under reduced pressure, diethyl ether was added to the resulting residue, and the resulting solid was collected by filtration and dried under reduced pressure to give the hydrochloride salt of the compound of formula (I).
The hydrochloride salt of the compound of formula (I) can be obtained by applying the following operation as a desalting reaction.
The hydrochloride of the compound of formula (I) was purified by ODS column chromatography (MeCN/0.1% aqueous formic acid), and the fractions containing the target were collected, made basic with saturated aqueous sodium bicarbonate, and extracted with CHCl 3/MeOH (5/1). The combined organic layers were dried over anhydrous sodium sulfate, the solution was concentrated under reduced pressure, and the resulting solid was washed with diethyl ether and dried under reduced pressure to give the compound of formula (I).
(Manufacturing method 2)
The compound of formula (IA) may be obtained by subjecting compound (87) to a deprotection reaction.
The reaction conditions were the same as in production method 1.
(Synthesis of raw materials 1)
The present production method is a first method for producing the compound (1) -1 contained in the raw material compound (1).
(First step)
This step is a method for producing compound (1) -1 by cycloaddition reaction of compound (2) and compound (3).
In the present reaction, the compound (2) and the compound (3) are used in equal amounts or in excess of one, and a mixture thereof is stirred for usually 0.1 hour to 5 days, preferably in the presence of a copper salt, further preferably in the presence of a copper salt and a reducing agent, in a solvent inert to the reaction or in the absence of a solvent, under cooling to a reflux under heating, preferably at 0℃to 100 ℃. Examples of the solvent used herein are not particularly limited, and include: halogenated hydrocarbons such as methylene chloride, 1, 2-dichloroethane, chloroform, etc.; aromatic hydrocarbons such as benzene, toluene, and xylene; ethers such as diethyl ether, THF, DOX, and 1, 2-dimethoxyethane; DMF, DMSO, ethyl acetate, meCN, tBuOH, water, and mixtures of these. Copper salts include CuI, cuSO4, and copper (I) triflate (CuOTf). Examples of the reducing agent include sodium ascorbate. The manner of carrying out the reaction in the presence of TEA, DIPEA, N-methylmorpholine (NMM), 2, 6-lutidine, tris [ (1-benzyl-1H-1, 2, 3-triazol-4-yl) methyl ] amine (TBTA) or the like is sometimes advantageous in terms of allowing the reaction to proceed smoothly.
[ Literature ]
Angew.Chem.Int.Ed.2002,41,p.2596-2599.
The present reaction may be carried out using a compound obtained by first subjecting PG 2 of compound (2) to a deprotection reaction.
(Raw material Synthesis 2)
( Wherein R represents a C 1-3 alkyl group. The same applies to the followings )
The present production method is a second method for producing the compound (1) -1 contained in the raw material compound (1).
(First step)
This step is a method for producing compound (5) by cycloaddition reaction of compound (2) and compound (4).
The reaction conditions were the same as in the first step of raw material synthesis 1.
(Second step)
This step is a method for producing the compound (6) by hydrolyzing the compound (5).
The reaction is carried out by stirring the compound (5) under cooling to a reflux under heating for usually 0.1 hour to 5 days. Examples of the solvent used herein include, but are not particularly limited to, alcohols, acetone, DMF, TMF, and the like. In addition, a mixed solvent of the solvent and water is sometimes preferable for the reaction. Examples of the hydrolysis reagent include, but are not particularly limited to, aqueous sodium hydroxide solution, aqueous potassium hydroxide solution, and trimethyltin hydroxide.
As references for this reaction, for example, the following references can be referred to.
Japanese society of chemistry "Experimental chemistry lecture (5 th edition)" 16 volume (2005) (pill)
Angew.Chem.Int.Ed.2005,44,p.1378-1382.
(Third step)
This step is a method for producing compound (1) -1 by amidation reaction of compound (6) with compound (7).
In the present reaction, the compound (6) and the compound (7) are used in equal amounts or in excess, and a mixture thereof is stirred for usually 0.1 hour to 5 days in the presence of a condensing agent in a solvent inert to the reaction, under cooling to heating, preferably-20 ℃ to 60 ℃. Examples of the solvent include, but are not particularly limited to: aromatic hydrocarbons such as toluene; ethers such as THF and DOX; halogenated hydrocarbons such as methylene chloride; alcohols; DMF, DMSO, ethyl acetate, meCN; and mixtures of these. As examples of condensing agents, there may be mentioned (benzotriazol-1-yloxy) tripyrrolidinyl hexafluorophosphate(PyBOP), O- (7-azabenzotriazol-1-yl) -N, N, N ', N' -tetramethyluronium Hexafluorophosphate (HATU), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide or its hydrochloride salt, N, N '-Dicyclohexylcarbodiimide (DCC), 1' -Carbonyldiimidazole (CDI), diphenyl azide phosphate (DPPA), and the like. The manner in which the additive (e.g., 1-hydroxybenzotriazole) is used is sometimes preferred for the reaction. The reaction may be carried out in the presence of an organic base such as TEA, DIPEA or NMM or an inorganic base such as potassium carbonate, sodium carbonate or potassium hydroxide, and it is advantageous in terms of smooth progress of the reaction.
In addition, a method of converting the compound (6) into a reactive derivative and then performing an acylation reaction may be used. Examples of the reactive derivative of carboxylic acid include an acid halide obtained by reacting with a halogenating agent such as phosphorus oxychloride or thionyl chloride, a mixed acid anhydride obtained by reacting with isobutyl chloroformate or the like, and an active ester obtained by condensing with 1-hydroxybenzotriazole or the like. The reaction of the reactive derivative with the compound (7) may be carried out in a solvent inert to the reaction, such as halogenated hydrocarbons, aromatic hydrocarbons, ethers, etc., under cooling to heating, preferably at-20℃to 120 ℃.
[ Literature ]
S.R.Sandler and W.Karo, "Organic Functional Group Preparations", 2 nd edition, volume 1, ACADEMIC PRESS Inc., 1991
Japanese society of chemistry "Experimental chemistry lecture (5 th edition)" 16 volume (2005) (pill)
(Raw material Synthesis 3)
( Wherein PG 3 represents a protecting group for OH, LG 1 represents a leaving group, and BLG represents a boric acid group or a trifluoroborate group (hereinafter, sometimes referred to as a boric acid group or the like) protected by a boric acid protecting group such as a boric acid group or a pinacol ester group. Examples of the leaving group shown here include Cl, br, methanesulfonyloxy, p-toluenesulfonyloxy, and the like. )
The present production method is a first method for producing the raw material compound (2).
(First step)
This step is a method for producing compound (10) by in situ substitution reaction of compound (8) -1 with compound (9).
In the present reaction, the compound (8) -1 and the compound (9) are used in equal amounts or in excess, and a mixture thereof is stirred for usually 0.1 hour to 5 days in a solvent inert to the reaction or in the absence of a solvent under cooling to a reflux under heating, preferably at 0℃to 80 ℃. Examples of the solvent used herein are not particularly limited, and include: halogenated hydrocarbons such as methylene chloride, 1, 2-dichloroethane, chloroform, etc.; aromatic hydrocarbons such as benzene, toluene, and xylene; ethers such as diethyl ether, THF, DOX, and 1, 2-dimethoxyethane; DMF, DMAc, DMSO, ethyl acetate, meCN; and mixtures of these. The reaction may be carried out in the presence of an organic base such as TEA, DIPEA, N-methylmorpholine (NMM) or 1, 4-diazabicyclo [2.2.2] octane (DABCO), or an inorganic base such as sodium hydride, potassium carbonate, sodium carbonate, cesium carbonate or tBuOK, which is advantageous in terms of smooth progress.
In addition, the compound (10) can also be produced by subjecting a compound obtained by a Rheumatoid-Herbac reaction of the compound (8) -1 with the compound (9) to a catalytic hydrogenation reaction.
(Second step)
This step is a method for producing compound (12) by an in-situ substitution reaction of compound (10) with compound (11).
The reaction conditions were the same as in the first step of raw material synthesis 3.
In addition, the compound (12) can also be produced by coupling a compound in which a hydrogen atom of the compound (11) is converted into halogen with the root of the compound (10).
(Third step)
This step is a method for producing compound (13) -1 by in situ substitution reaction of compound (12) with PG 3 -OH.
As examples of PG 3 -OH used herein, benzyl alcohol, p-methoxybenzyl alcohol and 1-phenylethanol may be mentioned.
The reaction conditions were the same as in the first step of raw material synthesis 3.
(Fourth step)
This step is a method for producing a compound (14) by a suzuki-miyaura coupling reaction of a compound (13) with a boric acid derivative composed of an R 2 -boric acid group or the like, wherein the compound (13) contains any one of a compound (13) -1 obtained according to the third step of the present synthetic method and a compound (13) -2 obtained according to the following (raw material synthesis 14). Examples of the boric acid group and the like used herein are not particularly limited, and boric acid groups, boric acid ester groups, boric acid pinacol ester groups, triol borate groups, and trifluoro borate groups may be mentioned.
In the present reaction, the compound (13) and the boric acid derivative composed of R 2 -boric acid group or the like are used in equal amounts or in excess, and the mixture thereof is stirred for usually 0.1 to 5 days in a solvent inert to the reaction in the presence of a base and a palladium catalyst at room temperature to a reflux under heating, preferably at 20℃to 140 ℃. Examples of the solvent used herein are not particularly limited, and include: halogenated hydrocarbons such as methylene chloride, 1, 2-dichloroethane, chloroform, etc.; aromatic hydrocarbons such as benzene, toluene, and xylene; ethers such as diethyl ether, THF, DOX, and 1, 2-dimethoxyethane; alcohols such as MeOH, etOH, isopropanol, butanol, and pentanol; DMF, DMSO, meCN, 1, 3-dimethylimidazolin-2-one, water; and mixtures of these. Examples of the base include inorganic bases such as potassium phosphate, sodium carbonate, potassium carbonate, sodium hydroxide, and barium hydroxide. Examples of the palladium catalyst include tetrakis (triphenylphosphine) palladium, bis (triphenylphosphine) palladium (II) dichloride, [1,1 '-bis (diphenylphosphino) ferrocene ] palladium (II) dichloride, methylene chloride adducts, (1 e,4 e) -1, 5-diphenylpenta-1, 4-dien-3-one/palladium (3:2), (2-dicyclohexylphosphino-2', 6 '-diisopropyloxy-1, 1' -biphenyl) [2- (2 '-amino-1, 1' -biphenyl) ] palladium (II) methanesulfonate, and palladium (II) acetate. The manner of carrying out the reaction in the presence of a ligand such as dicyclohexyl (2 ',6' -dimethoxybiphenyl-2-yl) phosphine, dicyclohexyl (2 ',6' -diisopropyloxy- [1,1 '-biphenyl ] -2-yl) phosphine, 1' -bis (diphenylphosphino) ferrocene, etc. is sometimes advantageous in terms of smooth progress of the reaction. In addition, the method of heating the mixture by microwave irradiation is sometimes advantageous in terms of smooth progress of the reaction.
[ Literature ]
J.Am.Chem.Soc.,2005,127,p.4685-4696
Org.Lett.2011,13,p.3948-3951
Org.Lett.2012,14,p.1278-1281
Compound (14) (here, R 2 is hydrogen) can be produced by dehalogenation reaction using the Pd catalyst of compound (13) and a reducing agent.
[ Literature ]
J.Org.Chem.,1977,42,p.3491-3494
Tetrahedron Letters 2013,54,5207-5210
(Fifth step)
This step is a method for producing compound (16) by a suzuki-miyaura coupling reaction of compound (14) with compound (15).
The reaction conditions were the same as in the fourth step of raw material synthesis 3.
When the compound (16) has an axiasymmetric structure, it is obtained as a mixture of stereoisomers, and each stereoisomer can be isolated by performing a conventional resolution operation, for example, resolution using ODS column chromatography or silica gel column chromatography.
(Sixth step)
This step is a method for producing the compound (17) by deprotecting the compound (16) by catalytic hydrogenation.
The reaction can be carried out by stirring the compound (16) under a hydrogen atmosphere under normal pressure to pressure in a solvent inert to the reaction such as MeOH, etOH, ethyl acetate, etc., in the presence of a metal catalyst, under cooling to heating, preferably at room temperature for 1 hour to 5 days. As the metal catalyst, a palladium catalyst such as Pd/C or palladium black, a platinum catalyst such as a platinum plate or platinum oxide, a nickel catalyst such as reduced nickel or raney nickel, or the like is used.
(Seventh step)
This step is a method for producing the compound (2) by reacting the compound (17) with the compound (18).
The reaction is carried out by using the compound (17) and the compound (18) in equal amounts or in excess and allowing a mixture thereof to react in the presence of a base in a solvent inert to the reaction under cooling to a reflux under heating, preferably at 0 to 80℃for usually 0.1 hour to 5 days. The solvent used herein is not particularly limited, and examples thereof include: aromatic hydrocarbons such as benzene, toluene, and xylene; alcohols such as MeOH and EtOH; ethers such as diethyl ether, THF, DOX, and 1, 2-dimethoxyethane; halogenated hydrocarbons such as methylene chloride, 1, 2-dichloroethane, chloroform, etc.; DMF, DMSO, ethyl acetate, meCN; and mixtures of these. Examples of the base include, but are not particularly limited to, organic bases such as TEA, DIPEA, 1, 8-diazabicyclo [5.4.0] -7-undecene, n-butyllithium, tBuOK, and inorganic bases such as sodium hydroxide, sodium carbonate, potassium carbonate, cesium carbonate, and sodium hydride. The manner in which the reaction is carried out in the presence of a tetra-n-butylammonium chloride or the like transfer catalyst is sometimes advantageous.
As references for this reaction, for example, the following references can be referred to.
Japanese chemical society, "Experimental chemistry lecture", 5 th edition, volume 14, charles, pill-like, 2005
The compound (2) may be obtained as a mixture of stereoisomers, and the compound (2) or the compound (2) having PG 2 as a protecting group may be subjected to deprotection, and the resulting compound may be subjected to a conventional resolution operation, for example, resolution using ODS column chromatography or silica gel column chromatography, to isolate each stereoisomer.
The reaction conditions for the deprotection reaction used herein are the same as those described in production method 1.
In addition, in the compound (18), a compound having a hydroxyl group in a portion corresponding to LG 1 is halogenated, whereby a compound having LG 1 as a halogen can be produced. Examples of the halogenating agent used herein are not particularly limited, and examples thereof include thionyl chloride, phosphorus oxychloride, hydrobromic acid, phosphorus tribromide, and the like.
As references for this reaction, for example, the following references can be referred to.
Japanese chemical society, "Experimental chemistry lecture", 5 th edition, volume 13, charles, pill-like, 2004 ]
Further, in the compound (18), a compound having a hydroxyl group in a portion corresponding to LG 1 is sulfonylated in the presence of a base, whereby a compound having LG 1 as a sulfonyloxy group can be produced. Examples of the sulfonylating agent used herein include, but are not particularly limited to, methanesulfonyl chloride, p-toluenesulfonyl chloride, methanesulfonyl anhydride, and the like. Examples of the base include, but are not particularly limited to, TEA, DIPEA, pyridine, and tetramethyl ethylenediamine.
As references for this reaction, for example, the following references can be referred to.
Synthesis 1999,9,p.1633-1636
(Raw material Synthesis 4)
(Wherein R LG represents a C 1-12 alkyl group and n represents 1 or 2.)
The present production method is a second method for producing the raw material compound (16).
(First step)
This step is a method for producing compound (19) by in situ substitution reaction of compound (10) with R LG -SH. Examples of R LG -SH used herein include C 1-12 alkyl mercaptans such as ethanethiol and dodecanethiol.
The reaction conditions were the same as in the first step of raw material synthesis 3.
(Second step)
This step is a process for producing compound (20) -1 by in situ substitution reaction of compound (19) with PG 3 -OH. As examples of PG 3 -OH used herein, benzyl alcohol, p-methoxybenzyl alcohol and 1-phenylethanol may be mentioned.
The reaction conditions were the same as in the first step of raw material synthesis 3.
(Third step)
This step is a method for producing compound (21) by suzuki-miyaura coupling reaction of compound (20) with a boric acid derivative composed of R 2 -boric acid group or the like, wherein compound (20) contains any one of compound (20) -1 obtained in the second step of the present synthetic method and compound (20) -2 obtained in the following (raw material synthesis 13).
The reaction conditions were the same as in the fourth step of raw material synthesis 3.
Compound (21) (here, R 2 is hydrogen) can be produced by dehalogenation reaction using the Pd catalyst of compound (20) and a reducing agent.
[ Literature ]
J.Org.Chem.,1977,42,p.3491-3494
Tetrahedron Letters 2013,54,5207-5210
(Fourth step)
This step is a method for producing compound (22) by a suzuki-miyaura coupling reaction of compound (21) with compound (15).
The reaction conditions were the same as in the fourth step of raw material synthesis 3.
(Fifth step)
This step is a method for producing the compound (23) by oxidation reaction of the compound (22).
In the present reaction, compound (22) is treated with an equal or excessive amount of an oxidizing agent in a solvent inert to the reaction under cooling to heating, preferably-20℃to 80℃for usually 0.1 hour to 3 days. In this reaction, oxidation of m-chloroperbenzoic acid, perbenzoic acid, peracetic acid, sodium hypochlorite or hydrogen peroxide is preferably used. Examples of the solvent include halogenated hydrocarbons such as aromatic hydrocarbons, ethers, and methylene chloride, DMF, DMSO, ethyl acetate, meCN, and mixtures of these. Examples of the other oxidizing agent include cumene hydroperoxide, potassium monopersulfate (Oxone), active manganese dioxide, chromic acid, potassium permanganate, sodium periodate, and the like.
[ Literature ]
Japanese chemical society, "Experimental chemistry lecture", 5 th edition, volume 17, charpy, 2004)
(Sixth step)
This step is a method for producing compound (16) by in situ substitution reaction of compound (23) with compound (24).
The reaction conditions were the same as in the first step of raw material synthesis 3.
When the compound (16) has an axiasymmetric structure, it is obtained as a mixture of stereoisomers, and each stereoisomer can be isolated by performing a conventional resolution operation, for example, resolution using ODS column chromatography or silica gel column chromatography.
(Raw material Synthesis 5)
The present production method is a second method for producing the raw material compound (2).
(First step)
This step is a method for producing the compound (25) by deprotecting the compound (23) -1 by catalytic hydrogenation.
The reaction conditions were the same as in the sixth step of raw material synthesis 3.
(Second step)
This step is a method for producing the compound (26) by reacting the compound (25) with the compound (18).
The reaction conditions were the same as in the seventh step of raw material synthesis 3.
(Third step)
This step is a method for producing compound (2) by an in-situ substitution reaction of compound (26) with compound (24).
The reaction conditions were the same as in the first step of raw material synthesis 3.
The compound (2) may be obtained as a mixture of stereoisomers, and the compound (2) or the compound (2) having PG 2 as a protecting group may be subjected to deprotection, and the resulting compound may be subjected to a conventional resolution operation, for example, resolution using ODS column chromatography or silica gel column chromatography, to isolate each stereoisomer.
The reaction conditions for the deprotection reaction used herein are the same as those described in production method 1.
(Raw material Synthesis 6)
(Wherein PG 4、PG5 represents a protecting group.)
The present production method is a method for producing the raw material compound (3).
(First step)
This step is a method for producing compound (28) by amidation reaction of compound (7) with compound (27).
The reaction conditions are the same as in the third step of raw material synthesis 2.
(Second step)
This step is a method for producing the compound (29) by subjecting the compound (28) to a deprotection reaction.
The reaction conditions were the same as those described in production method 1.
(Third step)
This step is a method for producing compound (31) by amidation reaction of compound (29) with compound (30).
The reaction conditions are the same as in the third step of raw material synthesis 2.
(Fourth step)
This step is a method for producing the compound (32) by subjecting the compound (31) to a deprotection reaction.
The reaction conditions were the same as those described in production method 1.
(Fifth step)
This step is a method for producing the compound (3) by reacting the compound (32) with a diazonium reagent.
In this reaction, compound (32) is treated with an equal or excess amount of the diazonium reagent in a solvent inert to the reaction, under cooling to heating, preferably 0℃to 50℃for usually 0.1 hour to 3 days. Examples of the diazo transfer reagent include, but are not particularly limited to, trifluoromethanesulfonyl azide, imidazole-1-sulfonyl azide or salts thereof, and 2-azide-1, 3-dimethylimidazoleHexafluorophosphate (ADMP), and the like. It is sometimes advantageous to carry out the reaction in the presence of an organic base such as TEA, 4-Dimethylaminopyridine (DMAP), 2, 6-lutidine, and a catalytic amount of a copper salt such as CuSO 4. Examples of the solvent include halogenated hydrocarbons such as THF and methylene chloride, meCN, alcohols, water, and mixtures of these.
[ Literature ]
J.Org.Chem.2012,77,p.1760-1764
Nature 2019,574,p.86-89
Org.Biomol.Chem.2014,12,p.4397-4406
(Raw material Synthesis 7)
(Wherein LG 2 represents a leaving group, PG 6 represents a protecting group for NH.)
The present production method is a method for producing the raw material compounds (1) -2 or the raw material compounds (1) -3 contained in the raw material compound (1). Here, a production method when L 2 in the starting compounds (1) to (3) is NR L1, pyrrolidinediyl, piperidediyl or piperazinediyl is shown.
(First step)
This step is a method for producing compound (2) -1 by reacting compound (17) with compound (33).
The reaction conditions were the same as in the seventh step of raw material synthesis 3.
(Second step)
This step is a method for producing the compound (2) -2 by hydrolyzing the compound (2) -1.
The reaction conditions are the same as in the second step of raw material synthesis 2.
(Third step)
This step is a method for producing compound (1) -2 by amidation reaction of compound (32) with compound (2) -2.
The reaction conditions are the same as in the third step of raw material synthesis 2.
(Fourth step)
This step is a method for producing compound (35) by amidation reaction of compound (32) with compound (34).
The reaction conditions are the same as in the third step of raw material synthesis 2.
(Fifth step, sixth step)
This step is a method for producing the compound (1) -3 by amidation reaction of the compound (2) -2 with the compound obtained by carrying out deprotection reaction of the compound (35).
The reaction conditions for the deprotection reaction are the same as those described in production method 1. Examples of the protecting group which can be deprotected under acidic conditions include t-butoxycarbonyl, triphenylmethyl, tetrahydro-2H-pyran-2-yl, methoxymethyl, dimethylmethane diyl, t-butylsulfinyl and the like.
The reaction conditions for the amidation reaction are the same as in the third step of raw material synthesis 2.
(Raw material Synthesis 8)
(Wherein when Z is NH, A 1 represents a hydrogen atom, A 2 represents a halogen atom, when Z is A5-membered heteroarene diyl group containing 1 to 4 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen, A 1 is a group selected from the group consisting of Cl, br and I, A 2 represents a boric acid group or the like, A 1 is a boric acid group or the like, A 2 represents a group selected from the group consisting of Cl, br and I.)
The present production method is a method for producing the raw material compounds (1) -4 contained in the raw material compound (1).
(First step)
This step is a method for producing the compound (38) by an in-situ reaction of the compound (36) with the compound (37) or a Buchwald-Hartmax amination reaction when Z is NH.
The reaction conditions for the in situ reaction are the same as in the first step of raw material synthesis 3.
As references for the blowald-hattery amination reaction, for example, the following references can be referred to.
J.Am.Chem.Soc.,2020,142,p.15027-15037
In addition, this step is a method for producing compound (38) by a suzuki-miyaura coupling reaction of compound (36) with compound (37) when Z is a 5-membered heteroaryl group containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen.
The reaction conditions were the same as in the fourth step of raw material synthesis 3.
Reference to the reaction when Z is the formula (XXIX) is made, for example, to the following.
J.Org.Chem.,2000,65,p.1516-1524
Chemical Communications 2014,50,p.1867-1870
Bioorg.Med.Chem.Lett.,2001,11,p.2061-2065
(Second step)
This step is a method for producing compound (39) by reacting compound (38) with compound (17).
The reaction conditions were the same as in the seventh step of raw material synthesis 3.
In addition, in this step, compound (39) can be produced by a casting reaction between a compound (17) and a compound (38) having a hydroxyl group in the portion corresponding to LG 1.
Reference to the casting reaction is made, for example, to the following.
Chem.Asian J.2007,2,p.1340-1355
(Third step)
This step is a method for producing the compound (40) by hydrolyzing the compound (39).
The reaction conditions are the same as in the second step of raw material synthesis 2.
(Fourth step)
This step is a method for producing compounds (1) -4 by amidation reaction of compound (40) with compound (29).
The reaction conditions are the same as in the third step of raw material synthesis 2.
(Raw material Synthesis 9)
(Wherein R 7 represents a group selected from the group consisting of formula (IX), formula (X), formula (XI), formula (XII), formula (XIII), formula (XIV), formula (XV), formula (XVI), formula (XVII) and formula (XVIII), PG 7 represents a protecting group, PG 8、PG9、PG10 are the same or different from each other, represent a hydrogen atom or a protecting group, A 3 represents a hydrogen atom, a carboxyl group or a boric acid group, etc., A 4 represents a hydrogen atom or a group selected from the group consisting of Cl, br and I, BLG 1 represents a boric acid group, etc.)
The present production method is a method for producing a starting compound (7) when R 7 is a group selected from the group consisting of formula (IX), formula (X), formula (XI), formula (XII), formula (XIII), formula (XIV), formula (XV), formula (XVI), formula (XVII) and formula (XVIII).
(First step)
This step is a method for producing the compound (45) by, for example, a sultam-heck reaction of the compound (42) in which R 7 is the formula (IX) and the compound (41) when a 3 is a hydrogen atom.
In the present reaction, the compound (42) and the compound (41) are used in equal amounts or in excess, and a mixture thereof is stirred for usually 0.1 hour to 5 days in a solvent inert to the reaction in the presence of a base and a palladium catalyst at room temperature to a reflux under heating, preferably at 20℃to 140 ℃. Examples of the solvent used herein include, but are not particularly limited to, ethers such as diethyl ether, THF, DOX, 1, 2-dimethoxyethane, DMF, DMAc, DMSO, meCN, 1, 3-dimethylimidazolin-2-one, ethyl acetate, water, and mixtures of these. Examples of the base include bases such as potassium phosphate, sodium carbonate, potassium carbonate, and potassium acetate. Examples of the palladium catalyst include tetrakis (triphenylphosphine) palladium, bis (triphenylphosphine) palladium (II) dichloride, [1,1 '-bis (diphenylphosphino) ferrocene ] palladium (II) dichloride, methylene chloride adducts, (1 e,4 e) -1, 5-diphenylpenta-1, 4-dien-3-one/palladium (3:2), (2-dicyclohexylphosphino-2', 6 '-diisopropyloxy-1, 1' -biphenyl) [2- (2 '-amino-1, 1' -biphenyl) ] palladium (II) methanesulfonate, and palladium (II) acetate. In addition, the method of heating the mixture by microwave irradiation is sometimes advantageous in terms of smooth progress of the reaction.
As references for the reaction, for example, the following references can be referred to.
Synthesis 2020,52,p.2521-2527
PNAS2016,113,p.7124-7129
Or for example, a method for producing the compound (45) by a ullmann reaction of the compound (42) wherein R 7 is a group selected from the group consisting of the formula (XII), the formula (XIII), the formula (XIV), the formula (XV) and the formula (XVII) with the compound (41).
As references for the reaction, for example, the following references can be referred to.
Angew.Chem.Int.Ed.,2003,42,p.5400-5449
In addition, this step is a method for producing the compound (45) by, for example, a decarbonation coupling reaction of the compound (42) in which R 7 is the formula (X) and the compound (41) when a 3 is a carboxyl group.
As references for the reaction, for example, the following references can be referred to.
Science,2006,313,p.662-664
Further, this step is a method for producing the compound (45) by, for example, a bell wood-palace coupling reaction of the compound (42) in which R 7 is a group selected from the group consisting of the formula (IX), the formula (XI) and the formula (XVI) with the compound (41) when a 3 is a boric acid group or the like.
The reaction conditions were the same as in the fourth step of raw material synthesis 3.
(Second step)
This step is a method for producing the compound (43) by a reaction in which the bromo group of the compound (41) is replaced with a boric acid group or the like.
As references for the reaction, for example, the following references can be referred to.
Eur.J.Med.Chem.,2019,162,p.407-422
J.Org.Chem.2020,85,16,p.10966-10972
J.Am.Chem.Soc.,2010,132,p.17701-17703
(Third step)
This step is a method for producing compound (45) by a suzuki-miyaura coupling reaction of compound (44) with compound (43) when a 4 is a group selected from the group consisting of Cl, br and I.
The reaction conditions were the same as in the fourth step of raw material synthesis 3.
In addition, this step is a method for producing the compound (45) by, for example, a Chan-Lam-Evans coupling reaction of the compound (44) which is a group selected from the group consisting of the formula (XII), the formula (XIII), the formula (XIV), the formula (XV) and the formula (XVII) with the compound (43) when A 4 is a hydrogen atom.
As references for the reaction, for example, the following references can be referred to.
Adv.Synth.Catal.2020,362,p.3311-3331.
(Fourth step)
This step is a method for producing compound (7) by subjecting compound (45) to a deprotection reaction. Examples of the protecting group which can be deprotected under acidic conditions include t-butoxycarbonyl, triphenylmethyl, tetrahydro-2H-pyran-2-yl, methoxymethyl, dimethylmethane diyl, t-butylsulfinyl and the like.
The reaction conditions were the same as those described in production method 1.
(Synthesis of raw materials 10)
(Wherein PG 11 represents a tert-butyl group, R 3A represents-O-or-N (R P) -)
The present production method is a method for producing the raw material compounds (2) -3.
(First step)
This step is a method for producing the compound (46) by hydrolyzing the compound (8) -1.
The reaction is carried out by stirring the compound (8) -1 under cooling to a heated reflux for usually 0.1 hour to 5 days. Examples of the solvent used herein include, but are not particularly limited to, alcohols, acetone, DMF, THF, and the like. In addition, a mixed solvent of the solvent and water is sometimes preferable for the reaction. Examples of the hydrolysis reagent include, but are not particularly limited to, aqueous sodium hydroxide solution, aqueous potassium hydroxide solution, and the like.
As references for this reaction, for example, the following references can be referred to.
Japanese society of chemistry "Experimental chemistry lecture (5 th edition)" 16 volume (2005) (pill)
Angew.Chem.Int.Ed.2005,44,p.1378-1382.
(Second step)
This step is a method for producing the compound (47) by protecting the hydroxyl group of the compound (46) with tert-butyl group (PG 11).
The reaction is carried out by stirring the compound (46) under cooling to a heated reflux for usually 0.1 hour to 5 days. Examples of the solvent used herein include, but are not particularly limited to, ethers such as THF and DOX, halogenated hydrocarbons such as methylene chloride, tBuOH, DMF, and the like. Examples of the tertiary butyl protecting agent include, but are not particularly limited to, isobutylene, 2-tertiary butyl-1, 3-diisopropylisourea, and the like.
The compound (47) can be produced by a dehydration condensation reaction between the compound (46) and tBuOH.
As references for this reaction, for example, the following references can be referred to.
P.g.m.wuts and T.W Greene, "Greene's Protective Groups in Organic Synthesis", 5 th edition, john Wiley & Sons inc., 2014
Org.Lett.,2012,14,17,p.4678-4681
(Third step)
This step is a method for producing compound (48) by in situ substitution reaction of compound (47) with R LG -SH.
The reaction conditions were the same as in the first step of raw material synthesis 4.
(Fourth step)
This step is a method for producing compound (49) by in situ substitution reaction of compound (48) with PG 3 -OH.
The reaction conditions were the same as in the third step of raw material synthesis 3.
(Fifth step)
This step is a method for producing the compound (50) by a suzuki-miyaura coupling reaction of the compound (49) with a boric acid derivative composed of an R 2 -boric acid group or the like.
The reaction conditions were the same as in the fourth step of raw material synthesis 3.
Compound (50) (here, R 2 is hydrogen) can be produced by dehalogenation reaction using the Pd catalyst of compound (49) and a reducing agent.
[ Literature ]
J.Org.Chem.,1977,42,p.3491-3494
Tetrahedron Letters 2013,54,5207-5210
(Sixth step)
This step is a method for producing compound (51) by a suzuki-miyaura coupling reaction of compound (50) with compound (15).
The reaction conditions were the same as in the fifth step of raw material synthesis 3.
(Seventh step)
This step is a method for producing the compound (52) by oxidation reaction of the compound (51).
The reaction conditions were the same as in the fifth step of raw material synthesis 4.
The compound (52) may have an axial asymmetry, and may be obtained as a mixture of stereoisomers, and each stereoisomer may be isolated by performing a conventional resolution operation, for example, resolution using ODS column chromatography or silica gel column chromatography.
In order to be able to deprotect under different conditions from the later-introduced protecting group PG 1, the compound (52) may be converted into another protecting group after the deprotection reaction.
The reaction conditions for the deprotection reaction used herein are the same as those described in production method 1.
Examples of the protecting group of the converted PG 2 include tetrahydro-2H-pyran-2-yl and the like.
As references for this reaction, for example, the following references can be referred to.
P.g.m.wuts and T.W Greene, "Greene's Protective Groups in Organic Synthesis", 5 th edition, john Wiley & Sons inc., 2014
(Eighth step)
This step is a method for producing the compound (53) by performing deprotection by catalytic hydrogenation of the compound (52).
The reaction conditions were the same as in the sixth step of raw material synthesis 3.
(Ninth step)
This step is a method for producing compound (54) by reacting compound (53) with compound (18).
The reaction conditions were the same as in the seventh step of raw material synthesis 3.
(Tenth step)
This step is a method for producing compound (55) by in situ substitution reaction of compound (54) with compound (24).
The reaction conditions were the same as in the sixth step of raw material synthesis 4.
(Eleventh step)
This step is a method for producing the compound (56) by subjecting the compound (55) to a deprotection reaction.
The reaction conditions were the same as those described in production method 1.
(Twelfth step)
This step is a method for producing compound (2) -3 by reacting compound (56) with compound (9) -1.
In the present reaction, the compound (56) and the compound (9) -1 are used in equal amounts or in excess, and a mixture thereof is stirred for usually 0.1 hour to 5 days in the presence of a condensing agent, in a solvent inert to the reaction, under cooling to heating, preferably at-20℃to 60 ℃. Examples of the solvent include, but are not particularly limited to, aromatic hydrocarbons such as toluene, ethers such as THF and DOX, halogenated hydrocarbons such as methylene chloride, alcohols, DMF, DMSO, ethyl acetate, meCN, and mixtures of these. As examples of the condensing agent, pyBOP, HATU, CDI and the like are given. The reaction may be carried out in the presence of an organic base such as TEA, DIPEA or NMM or an inorganic base such as potassium carbonate, sodium carbonate or cesium carbonate, and it is advantageous in terms of smooth progress of the reaction.
(Raw material Synthesis 11)
The present production method is a method for producing (16) -1 wherein A is N in the raw material compound (16).
(First step)
This step is a method for producing compound (57) by an in-situ substitution reaction of compound (52) -1 with compound (24).
The reaction conditions were the same as in the sixth step of raw material synthesis 4.
(Second step)
This step is a method for producing the compound (58) by subjecting the compound (57) to a deprotection reaction.
The reaction conditions were the same as those described in production method 1.
(Third step)
This step is a method for producing compound (16) -1 by reacting compound (58) with compound (9).
The reaction conditions were the same as in the twelfth step of the raw material synthesis 10.
(Raw materials Synthesis 12)
The present production method is a method for producing a raw material compound (57).
(First step)
This step is a method for producing compound (59) by an in-situ substitution reaction of compound (47) with compound (24).
The reaction conditions were the same as in the second step of raw material synthesis 3.
(Second step)
This step is a method for producing compound (60) by in situ substitution reaction of compound (59) with PG 3 -OH.
The reaction conditions were the same as in the third step of raw material synthesis 3.
(Third step)
This step is a method for producing the compound (61) by a suzuki-miyaura coupling reaction of the compound (60) with a boric acid derivative composed of an R 2 -boric acid group or the like.
The reaction conditions were the same as in the fourth step of raw material synthesis 3.
Compound (61) (here, R 2 is hydrogen) can be produced by dehalogenation reaction using the Pd catalyst of compound (60) and a reducing agent.
[ Literature ]
J.Org.Chem.,1977,42,p.3491-3494
Tetrahedron Letters 2013,54,5207-5210
(Fourth step)
This step is a method for producing compound (57) by a suzuki-miyaura coupling reaction of compound (61) with compound (15).
The reaction conditions were the same as in the fifth step of raw material synthesis 3.
(Raw material Synthesis 13)
The present production method is a method for producing the raw material compound (20) -2.
(First step)
This step is a method for producing compound (8) -2 by chlorination of compound (62).
In the present reaction, the compound (62) and the chlorinating agent are used in equal amounts or in excess, and a mixture thereof is stirred for usually 0.1 hour to 5 days in a solvent inert to the reaction or in the absence of a solvent under cooling to a heating reflux, preferably at 60℃to a heating reflux. Examples of the solvent used herein include, but are not particularly limited to, aromatic hydrocarbons such as toluene, ethers such as THF and DOX, halogenated hydrocarbons such as methylene chloride, and the like. Examples of the chlorinating agent include phosphorus oxychloride and thionyl chloride. The manner of carrying out the reaction in the presence of an organic base such as TEA, DIPEA or NMM is sometimes advantageous in terms of smooth progress of the reaction.
(Second step)
This step is a method for producing compound (63) by in situ substitution reaction of compound (8) -2 with R LG -SH.
The reaction conditions were the same as in the first step of raw material synthesis 4.
(Third step)
This step is a method for producing compound (64) by in situ substitution reaction of compound (63) with PG 3 -OH.
The reaction conditions were the same as in the third step of raw material synthesis 3.
(Fourth step)
This step is a method for producing compound (20) -2 by in situ substitution reaction of compound (64) with compound (9) -1.
The reaction conditions were the same as in the first step of raw material synthesis 3.
(Raw materials Synthesis 14)
The present production method is a method for producing the raw material compound (13) -2.
(First step)
This step is a method for producing compound (65) by in-situ substitution reaction of compound (8) -2 with compound (24).
The reaction conditions were the same as in the second step of raw material synthesis 3.
In addition, compound (65) can be produced by coupling a compound that converts a hydrogen atom of compound (24) into a halogen with the root bank of compound (8) -2.
(Second step)
This step is a method for producing compound (66) by in situ substitution reaction of compound (65) with PG 3 -OH.
The reaction conditions were the same as in the third step of raw material synthesis 3.
(Third step)
This step is a method for producing compound (13) -2 by an in-situ substitution reaction of compound (66) with compound (9) -1.
The reaction conditions are the same as in the first step of raw material synthesis 3, but the use of DMAc or the like as a solvent, the reaction at a reaction temperature of 0 ℃ to 120 ℃, and the use of potassium carbonate or the like as an inorganic base are sometimes advantageous in terms of smooth progress of the reaction.
(Raw material Synthesis 15)
(Wherein A 5 represents a group selected from the group consisting of Cl, br, mesylate and triflate, LG 3 represents a leaving group.)
The present production method is a method for producing a raw material compound (38) -1 and a raw material compound (38) -2 contained in a raw material compound (38).
(First step)
This step is a method for producing compound (70) -1 by reacting compound (67) with compound (69).
As references for the reaction, for example, the following references can be referred to.
Chem.Commun.,2014,50,15,p.1867-1870
(Second step)
This step is a method for producing compound (70) -1 by the reaction of compound (69) with glyoxylic acid using an aryl-substituted tosylmethyl isonitrile (TosMIC) reagent.
As references for the reaction, for example, the following references can be referred to.
J.Org.Chem.,2000,65,5,p.1516-1524
J.Am.Chem.Soc.,2007,129,3,p.490-491
(Third step)
This step is a process for the manufacture of compound (38) -1 by suzuki-miyaura coupling of compound (70) -1 with an alkoxymethylboronic acid derivative followed by deprotection under acidic conditions.
The reaction conditions of the suzuki-miyaura coupling reaction are the same as the fourth step of the raw material synthesis 3.
Examples of the alkoxymethylboronic acid derivative used herein include potassium (2-trimethylsilyl) -ethoxymethyltrifluoroborate.
As references for the reaction, for example, the following references can be referred to.
Org.Lett.,2008,10,11,p.2135-2128
Org.Lett.,2011,13,15,p.3948-3951
The reaction conditions for the deprotection reaction under acidic conditions to be performed subsequently are the same as those described in production method 1. Examples of the acid used herein include trifluoroacetic acid and the like.
As references for deprotection reactions, for example, the following references can be referred to.
P.g.m.wuts and T.W Greene, "Greene's Protective Groups in Organic Synthesis", 5 th edition, john Wiley & Sons inc., 2014
In addition, in this step, compound (38) -1 can be produced by suzuki-miyaura coupling reaction of compound (70) -1 with an acetoxymethylboronic acid derivative.
As references for the reaction, for example, the following references can be referred to.
Org.Lett.,2012,14,5,p.1278-1281
(Fourth step)
This step is a method for producing compound (70) -2 by reacting compound (71) with compound (69).
In the present reaction, the compound (71) and the compound (69) are used in equal amounts or in excess, and a mixture thereof is stirred for usually 0.1 hour to 5 days in a solvent inert to the reaction or in the absence of a solvent under cooling to heating reflux, preferably at room temperature to heating reflux. Examples of the solvent used herein include, but are not particularly limited to, aromatic hydrocarbons such as toluene, ethers such as THF and DOX, halogenated hydrocarbons such as methylene chloride, DMF, DMSO, ethyl acetate, meCN, and the like. The reaction may be carried out in the presence of an organic base such as TEA, DIPEA or NMM or an inorganic base such as potassium carbonate, sodium carbonate or cesium carbonate, and it is advantageous in terms of smooth progress of the reaction.
(Fifth step)
This step is a process for the manufacture of compound (38) -2 by suzuki-miyaura coupling of compound (70) -2 with an alkoxymethylboronic acid derivative followed by deprotection under acidic conditions.
The reaction conditions are the same as in the third step of the raw material synthesis 15.
In addition, in this step, compound (38) -2 can be produced by suzuki-miyaura coupling reaction of compound (70) -2 with an acetoxymethylboronic acid derivative.
(Raw materials Synthesis 16)
( Wherein R 3B represents a divalent group formed by removing 1 bond from a group selected from the group consisting of-P-Q, V and a saturated or unsaturated 7-to 8-membered bridged heterocyclic group, and X 1 represents O or NR 4x. The following is the same. )
The present production method is a method for producing a compound (87) -1 in which A and E in the raw material compound (87) of production method 2 are N, G is CR 2, X is O or NR 4x、LY is-O-CH 2 -, L is a bond, and Z is the following formula (XXVII).
(First step)
This step is a method for producing compound (74) by reacting compound (72) with compound (73).
In this reaction, after converting the compound (72) into the corresponding enolate using an orthoester such as trimethyl orthoformate under acidic conditions, the compound (73) is added in an excess amount of one or both of the equivalent amounts, and the mixture is stirred in a solvent inert to the reaction under heating reflux, preferably at 60 ℃ to heating reflux, for usually 0.1 hour to 5 days. Examples of the solvent used herein include, but are not particularly limited to, aromatic hydrocarbons such as toluene, ethers such as THF and DOX, DMF, DMAc, and the like.
(Second step)
This step is a method for producing compound (75) from compound (74).
The reaction is carried out by stirring the compound (74) in a solvent inert to the reaction under heating reflux, preferably at 150℃to heating reflux, usually for 0.1 hour to 5 days. Examples of the solvent used herein include, but are not particularly limited to, NMP and the like.
(Third step)
This step is a method for producing compound (76) from compound (75).
In the present reaction, the compound (75) and the brominating agent are used in equal amounts or in excess, and a mixture thereof is stirred in a solvent inert to the reaction or in the absence of a solvent, under cooling to a reflux under heating, preferably at room temperature, for usually 0.1 hour to 5 days. Examples of the solvent used herein include, but are not particularly limited to, aromatic hydrocarbons such as toluene, ethers such as THF and DOX, halogenated hydrocarbons such as methylene chloride, DMF, and the like. Examples of the brominating agent include N-bromosuccinimide, N-bromophthalimide, 1, 3-dibromo-5, 5-dimethylhydantoin, dibromoisocyanuric acid, and the like.
(Fourth step)
This step is a method for producing compound (77) from compound (76).
The reaction conditions were the same as in the first step of the raw material synthesis 13.
(Fifth step)
This step is a method for producing compound (79) by in situ substitution reaction of compound (77) with compound (78).
The reaction conditions were the same as in the first step of raw material synthesis 3.
(Sixth step)
This step is a method for producing compound (81) by in situ substitution reaction of compound (79) with compound (80).
The reaction conditions were the same as in the first step of raw material synthesis 3.
(Seventh step)
This step is a method for producing compound (82) by in situ substitution reaction of compound (81) with PG 3 -OH.
As examples of PG 3 -OH used herein, benzyl alcohol, p-methoxybenzyl alcohol and 1-phenylethanol may be mentioned.
The reaction conditions were the same as in the first step of raw material synthesis 3.
(Eighth step)
This step is a method for producing compound (83) by a suzuki-miyaura coupling reaction of compound (82) with compound (15) which is a boric acid derivative.
The reaction conditions were the same as in the fourth step of raw material synthesis 3.
When the compound (83) has an axiasymmetric structure, it is obtained as a mixture of stereoisomers, and each stereoisomer can be isolated by performing a conventional resolution operation, for example, resolution using ODS column chromatography or silica gel column chromatography.
In order to be able to deprotect under different conditions from the later-introduced protecting group PG 1, the compound (83) may be converted into another protecting group after the deprotection reaction.
The reaction conditions for the deprotection reaction used herein are the same as those described in production method 1.
Examples of the protecting group of the converted PG 2 include tetrahydro-2H-pyran-2-yl and the like.
As references for this reaction, for example, the following references can be referred to.
P.g.m.wuts and T.W Greene, "Greene's Protective Groups in Organic Synthesis", 5 th edition, john Wiley & Sons inc., 2014
(Ninth step)
This step is a method for producing the compound (84) by deprotecting the compound (83) by catalytic hydrogenation.
The reaction conditions were the same as in the sixth step of raw material synthesis 3.
(Tenth step)
This step is a method for producing compound (85) by reacting compound (84) with compound (18).
The reaction conditions were the same as in the seventh step of raw material synthesis 3.
(Eleventh step)
This step is a method for producing compound (87) -1 by reacting compound (85) with compound (86).
The reaction conditions were the same as in the first step of raw material synthesis 1.
The compounds of formula (I) and formula (IA) are isolated and purified as free compounds, salts, hydrates, solvates or polymorphs thereof or as amorphous solid forms of matter. Salts of the compounds of formula (I) and formula (IA) may also be produced by salt-forming reactions which are provided in conventional processes.
Isolation and purification can be carried out by ordinary chemical procedures such as extraction, fractional crystallization, and various fractional chromatographs.
The individual isomers may be produced by selecting an appropriate starting compound, or may be separated by utilizing differences in physicochemical properties between the isomers. For example, the optical isomer may be obtained by a general optical resolution method of a racemate (for example, fractional crystallization to a stereoisomer salt with an optically active base or acid, chromatography using a chiral column or the like), or may be produced from an appropriate optically active starting compound.
In addition, the compounds of formula (I) and formula (IA) or intermediates thereof sometimes have axial asymmetry, are obtained as a mixture of stereoisomers, and each stereoisomer may be isolated by performing a conventional resolution operation, for example, resolution using Octadecylsilyl (ODS) column chromatography, silica gel column chromatography.
The pharmacological activity of the compounds of formula (I) and formula (IA) was confirmed by the following test.
Experimental example 1 evaluation of KRAS decomposition of human G12D mutant KRAS-positive pancreatic cancer Strain AsPC-1 (CRL-1682; ATCC)
The KRAS degradation by the test compound was evaluated by measuring the KRAS G12D expression level by the cell ELISA method.
AsPC-1 cells were seeded at 2.0X10 4 cells per well in 384-well plates (Greiner bio-one Co.) in 20. Mu.L each. For cell culture conditions, RPMI1640 (Sigma-Aldrich) medium containing 10% fetal bovine serum (Cytiva Co.) was used in the presence of 5% CO 2 at 37 ℃.
The next day, the test compound (final concentration in the range of 10. Mu.M to 0.3nM, 10 spots) and DMSO (Fuji photo-pure Co., ltd.) as a solvent for the test compound as a negative control were diluted 500-fold with fresh medium, and 20. Mu.L of each of the wells was added thereto, followed by culturing overnight.
The next day, the culture supernatant was removed, 20. Mu.L of 4% paraformaldehyde phosphate buffer (Fuji photo-Co., ltd.) was added to each well, and the mixture was allowed to stand at room temperature for 30 minutes to fix the cells. The supernatant was removed, and 20. Mu.L of phosphate-buffered saline (PBS; fuji film and Wako pure chemical industries, ltd.) containing 0.1% Triton X-100 (Amersham Biosciences Co.) was added to each well. After standing at room temperature for 10 minutes, the supernatant was removed, 25. Mu.L of PBS was added to each well, and the supernatant was removed, thereby washing each well. The washing was performed 2 times in total. Then, the supernatant was removed, and 20. Mu.L of PBS containing 0.5% sodium dodecyl sulfate (SDS; invitrogen) was added to each well. After standing at room temperature for 10 minutes, the supernatant was removed by centrifugation (using a centrifugal dehydrator, the supernatant was removed by the same method as described below), 25. Mu.L of PBS was added to each well, and the supernatant was removed, thereby washing each well. The washing was performed 2 times in total. The supernatant was removed, and 20. Mu.L of a blocking solution (INTERCEPT BLOCKING BUFFER; li-COR Biosciences Co.) was added to each well. After leaving at room temperature for 30 minutes, the supernatant was removed, 20. Mu.L of a 1000-fold solution of an Anti- β -actin antibody (Anti-beta Actin antibody [ mAbcam 8226] -Loading Control; abcam) diluted with a blocking solution was added to the wells used as a positive control, and 20. Mu.L of a 1000-fold solution of an Anti-Ras (G12D Mutant Specific) antibody (Ras (G12 DMutant Specific) (D8H 7) Rabbit mAb #14429;Cell Signaling Technology) and an Anti- β -actin antibody (abcam) were added to the other wells and allowed to stand at 4℃overnight.
The next day, the supernatant was removed, 25. Mu.L of PBS was added to each well, and the supernatant was removed, thereby washing each well. The washing was performed 2 times in total. The supernatant was removed, and 20. Mu.L of a 1000-fold dilution of donkey anti-mouse IgG H & L (IRDye 680 RD) (Li-COR Biosciences) and goat anti-rabbit IgG H & L (IRDye 800 CW) (Li-COR Biosciences) as secondary antibodies was added to each well. After standing at room temperature for 1 hour, the supernatant was removed, 25. Mu.L of PBS was added to each well, and the supernatant was removed, thereby washing each well. The washing was performed 2 times in total. After removing the supernatant, the plate was air-dried at room temperature for 2 hours or more, and fluorescence signals at 700nm and 800nm were measured with Aerius (Li-COR Biosciences).
After correcting the signal value of RAS measured at a fluorescence wavelength of 800nm with the signal value of beta-actin measured at a fluorescence wavelength of 700nm, the signal value when DMSO was added was set to 0%, the signal value when staining with only anti-beta-actin antibody was set to 100%, and the 50% decomposition value (DC 50) of KRAS was calculated by nonlinear regression analysis of the Sigmoid-Emax model. The results of several test compounds of formula (I) and formula (IA) are shown in the following table.
[ Table 1-1]
Ex | DC50(nM) | Ex | DC50(nM) | Ex | DC50(nM) |
1 | 13 | 19 | 91 | 37 | 58 |
2 | 20 | 20 | 41 | 38 | 25 |
3 | 47 | 21 | 33 | 39 | 37 |
4 | 35 | 22 | 59 | 40 | 47 |
5 | 9 | 23 | 31 | 41 | 29 |
6 | 24 | 24 | 48 | 42 | 91 |
7 | 40 | 25 | 75 | 43 | 41 |
8 | 18 | 26 | 22 | 44 | 43 |
9 | 15 | 27 | 68 | 45 | 27 |
10 | 18 | 28 | 47 | 46 | 25 |
11 | 10 | 29 | 20 | 47 | 26 |
12 | 61 | 30 | 121 | 48 | 38 |
13 | 10 | 31 | 59 | 49 | 25 |
14 | 20 | 32 | 51 | 50 | 18 |
15 | 99 | 33 | 33 | 51 | 14 |
16 | 26 | 34 | 53 | 52 | 11 |
17 | 74 | 35 | 63 | 53 | 7 |
18 | 508 | 36 | 82 | 54 | 34 |
[ Tables 1-2]
Ex | DC50(nM) |
55 | 10 |
Test example 2 evaluation of ERK phosphorylation inhibition by human G12D mutant KRAS positive pancreatic cancer strain AsPC-1
Phosphorylation of threonine 202 (Thr 202) and tyrosine 204 (Tyr 204) of ERK downstream of KRAS signal was measured by cell ELISA, whereby inhibition of ERK phosphorylation by test compounds was assessed.
AsPC-1 cells were seeded at 36 μl/well in 384-well plates at 2.0x10 4 cells per well. For cell culture conditions, RPMI1640 medium containing 10% fetal bovine serum was used, performed in the presence of 5% co 2 at 37 ℃.
The next day, the test compound (final concentration ranging from 10. Mu.M to 0.3nM, 10 spots), trimetinib (MEK inhibitor) as a positive control, and solvent DMSO as a negative control, were diluted 100-fold with fresh medium, and after adding 4. Mu.L each to each well, cultured overnight. After the incubation, 30. Mu.L of 30% glyoxal (40% glyoxal (Nacalai Tesque) was diluted with PBS) was added to each well, and the mixture was allowed to stand at room temperature for 60 minutes to fix the cells. After that, the plate was centrifuged to remove the supernatant (using a centrifugal dehydrator, the supernatant was removed by the same method as described below), and 20. Mu.L of PBS containing 0.1% Triton X-100 was added to each well. After leaving at room temperature for 10 minutes, the supernatant was removed, and the same operation was repeated. Then, 20. Mu.L of PBS containing 0.5% SDS was added to each well, and the mixture was allowed to stand at room temperature for 30 minutes, followed by removal of the supernatant. Next, 20. Mu.L of the blocking solution (INTERCEPT BLOCKING BUFFER) was added to each well, and the mixture was allowed to stand at room temperature for 1 hour. The supernatant was removed, and 15. Mu.L of a phosphorylated antibody (Phospho-p 44/42MAPK (Erk 1/2) (Thr 202/Tyr 204) (D13.14.4E) XP Rabbit mAb; cell SignalingTechnology) of ERK (Thr 202/Tyr 204) diluted 2500-fold with a blocking solution as a primary antibody was added to each well and allowed to stand at 4℃overnight.
The next day, the supernatant was removed, and 50. Mu.L of PBS containing 0.05% Tween-20 (Thermo FISHER SCIENTIFIC Co.) was added to each well, and the supernatant was removed, whereby each well was washed. The washing was performed 3 times in total. After washing, 15. Mu.L of goat anti-rabbit IgG H & L (IRDye 800 CW) diluted 1000-fold with blocking solution as secondary antibody was added to each well and allowed to stand at room temperature for 1 hour. The supernatant was removed, and each well was washed 3 times similarly after the reaction with PBS containing 0.05% Tween-20 and primary antibody. After removing the supernatant, the plate was air-dried at room temperature for 3 hours or more, and the fluorescent signal at 800nm was measured by Aerius.
The 50% inhibition value (IC 50) was calculated by nonlinear regression analysis of the Sigmoid-Emax model, with the signal value at the time of DMSO addition set to 100% and the signal value at the time of 1. Mu.M addition set to 0%. The results of several test compounds of formula (I) and formula (IA) are shown in the following table.
[ Table 2-1]
Ex | IC50(nM) | Ex | IC50(nM) | Ex | IC50(nM) |
1 | 7 | 19 | 95 | 37 | 32 |
2 | 9 | 20 | 38 | 38 | 36 |
3 | 16 | 21 | 47 | 39 | 57 |
4 | 8 | 22 | 5 | 40 | 7 |
5 | 12 | 23 | 13 | 41 | 9 |
6 | 5 | 24 | 15 | 42 | 6 |
7 | 9 | 25 | 6 | 43 | 7 |
8 | 20 | 26 | 2 | 44 | 31 |
9 | 11 | 27 | 7 | 45 | 16 |
10 | 8 | 28 | 5 | 46 | 6 |
11 | 24 | 29 | 6 | 47 | 22 |
12 | 42 | 30 | 10 | 48 | 15 |
13 | 8 | 31 | 20 | 49 | 32 |
14 | 4 | 32 | 37 | 50 | 8 |
15 | 7 | 33 | 45 | 51 | 59 |
16 | 16 | 34 | 21 | 52 | 4 |
17 | 7 | 35 | 12 | 53 | 9 |
18 | 100 | 36 | 21 | 54 | 5 |
[ Table 2-2]
Ex | IC50(nM) |
55 | 4 |
56 | 17 |
Test example 3 evaluation of the inhibition of adherent independent cell proliferation of human G12D mutant KRAS-positive pancreatic cancer Strain AsPC-1
The inhibition of cell proliferation by the test compound was evaluated by spherical three-dimensional culture.
AsPC-1 cells were seeded at 36 μl/well in a cell low adsorption U-bottom 384-well plate (PrimeSurface: sumitomo electric company) at 5×10 2 cells per well. For cell culture conditions, RPMI1640 medium containing 10% fetal bovine serum was used, performed in the presence of 5% co 2 at 37 ℃.
The next day, the test compound (final concentration ranging from 10. Mu.M to 0.3nM, 10 spots or 6 spots) and the solvent DMSO of the test compound as a negative control were diluted 100-fold with fresh medium, and 4. Mu.L of each was added to each well. After 6 days of incubation at 37℃in the presence of 5% CO 2, 20. Mu.L of CellTiter-Glo2.0 (Promega Co.) was added to each well. After stirring at normal temperature for 1 hour using a flat plate mixer (FINEPCR Co.), the luminescence signal was measured by ARVO X3 (Perkinelmer Co.).
The signal value at DMSO treatment was set to 100%, the signal value at medium alone in the absence of cells was set to 0%, and the 50% inhibition value (IC 50) was calculated by Sigmoid-Emax model nonlinear regression analysis. The results of several test compounds of formula (I) and formula (IA) are shown in the following table.
[ Table 3-1]
Ex | IC50(nM) | Ex | IC50(nM) | Ex | IC50(nM) |
1 | 9 | 19 | 50 | 37 | 32 |
2 | 92 | 20 | 21 | 38 | 36 |
3 | 7 | 21 | 53 | 39 | 39 |
4 | 7 | 22 | 5 | 40 | 6 |
5 | 10 | 23 | 12 | 41 | 8 |
6 | 8 | 24 | 26 | 42 | 4 |
7 | 23 | 25 | 6 | 43 | 6 |
8 | 16 | 26 | 3 | 44 | 17 |
9 | 9 | 27 | 7 | 45 | 15 |
10 | 6 | 28 | 5 | 46 | 5 |
11 | 19 | 29 | 7 | 47 | 15 |
12 | 41 | 30 | 12 | 48 | 16 |
13 | 10 | 31 | 18 | 49 | 10 |
14 | 4 | 32 | 24 | 50 | 8 |
15 | 7 | 33 | 24 | 51 | 44 |
16 | 14 | 34 | 13 | 52 | 4 |
17 | 8 | 35 | 9 | 53 | 10 |
18 | 73 | 36 | 19 | 54 | 7 |
[ Table 3-2]
Ex | IC50(nM) |
55 | 15 |
56 | 73 |
[ Tables 3-3]
Ex | IC50(nM) |
57 | 95 |
Test example 4 evaluation of antitumor Effect of human G12D mutant KRAS-positive pancreatic cancer Strain PK-59 tumor-bearing mice
PK-59 cells (RIKEN BRC, RCB 1901) were cultured in RPMI1640 medium containing 10% fetal bovine serum in the presence of 5% CO 2 at 37 ℃. PK-59 cells were recovered, suspended in PBS, prepared to 1.0 to 2.0X10/7/mL by adding an equal amount of matrigel (Becton, dickinson and Company), and the resulting cell suspension was subcutaneously implanted in 4 to 5-week-old male nude mice (CanN.Cg-Foxnl nu/CrlCrlj (nu/nu), CHARLES RIVER Laboratories Japan) at a capacity of 100. Mu.L. After about 2 weeks of implantation, the groups were grouped in such a way that the tumor volume and body weight were approximately the same between the groups, and administration of the test compound was started the next day. The test was performed in 5 groups of solvent and test compound administration. The compound was dissolved in ethanol (Fuji photo-pure chemical), 5% dextrose solution (tsukamurelca pharmaceutical), 1M hydrochloric acid (kandong chemical), 50% (2-hydroxypropyl) -beta-cyclodextrin (HP-beta CD) aqueous solution (ROQUETTE), HCO-40 (solar chemical), 1M sodium hydroxide aqueous solution (kandong chemical) at a liquid ratio of 4:84.4:1.1:1:9:0.5 in a solvent. The test compound or solvent dissolved with each solvent is administered into the tail vein. The administration was twice at intervals of once a week. Tumor diameter and body weight measurements were performed 2 times every 1 week. The tumor volume was calculated using the following formula.
[ Tumor volume (mm 3) ]= [ tumor major diameter (mm) ]× [ tumor minor diameter (mm) ] 2 ×0.5
The tumor growth inhibition (%) by the test compound was calculated by setting the tumor volume of the test compound administration group at the day before the start of administration to 100% inhibition and the tumor volume of the solvent group at 2 weeks after the first administration to 0% inhibition. When the tumor volume of the test compound administration group was lower than the tumor volume of the day before the administration start, the tumor volume of the day before the administration start was set to 0% regression, and the tumor volume 0 was set to 100% regression, and the tumor regression rate (%) of the test compound was calculated. The results of several test compounds of formula (I) and formula (IA) are shown in the following table.
TABLE 4
Ex | Dosage (mg/kg) | The first administration has anti-tumor effect after 2 weeks |
9 | 10 | 81% Inhibition |
11 | 10 | 6% Withdrawal |
13 | 10 | 26% Recession |
50 | 10 | 20% Withdrawal |
53 | 10 | 53% Withdrawal |
Test example 5 evaluation of KRAS G12D/SOS/c-Raf Complex formation inhibitory Effect
The inhibition of complex formation of these proteins by test compounds was investigated by time resolved fluorescence resonance energy transfer (TR-FRET) using human recombinant KRAS G12D, SOS and c-Raf proteins.
To 384 well plates (Corning Corp.) 40000nM to 40nM of biotinylated AviTag-KRAS G12D (amino acid region 1-185, GDP) (2.5. Mu.L; 400 nM) and test compound dissolved in assay buffer (50 mM HEPES [ Jena Corp. ],150mM NaCl[Nacalai Tesque Co., 5mM MgCl 2 [ Thermo FISHER SCIENTIFIC Co., 0.05% Tween 20[ Sigma-Aldrich Corp. ], pH 7.0) were added in an amount of 2.5. Mu.L. To this was added c-Raf (amino acid region 51-131) GST (2.5. Mu.L; 130 nM) containing Son of Sevenless (SOS) (amino acid region 564-1049,2.5. Mu.L; 1.3. Mu.M), GTP (Sigma-Aldrich Co.; 2. Mu.M) and allowed to stand at room temperature for 1 hour. Next, a mixture (10. Mu.L) of LANCE Ulight-anti-GST (Perkinelmer; 120 nM) and LANCE Eu-W1024 labeled Streptoavidin (Perkinelmer; 100 ng/mL) was added, and the fluorescence intensities at 620nM and 665nM were measured using EnVision 2104 (Perkinelmer) at an excitation wavelength of 337 nM. After normalizing the value with the fluorescence intensity of the reference wavelength of 620nm, the signal value at the solvent treatment was set to 0% inhibition, the signal value at the time of no addition of GTP was set to 100% inhibition, and the 50% inhibition concentration (IC 50) was calculated by Sigmoid-Emax model nonlinear regression analysis.
As a result of the above-described test, G12D mutant KRAS decomposition was confirmed for several compounds of the formula (I) and the formula (IA) (test example 1). In addition, inhibition of G12D mutant KRAS was confirmed (test example 5). Furthermore, the compounds of the formula (IA) and the dry type (I) showed an inhibition of the phosphorylation of ERK located downstream of the KRAS signal (test example 2). In addition, several compounds of the formula (I) and the formula (IA) were confirmed to have an inhibitory effect on cell proliferation of a human G12D mutant KRAS-positive pancreatic cancer strain (test example 3) and an antitumor effect in tumor-bearing mice of a human G12D mutant KRAS-positive pancreatic cancer strain (test example 4). Thus, the compounds of formula (I) and formula (IA) may be used in the treatment of pancreatic cancer, in particular G12D mutant KRAS positive pancreatic cancer, etc.
The pharmaceutical composition containing 1 or 2 or more of the compounds of formula (I) and formula (IA) or salts thereof as an active ingredient can be prepared by a method generally used using an excipient commonly used in the art, that is, a pharmaceutical excipient or a pharmaceutical carrier, and the like.
The administration may be by oral administration such as tablet, pill, capsule, granule, powder, liquid, etc., or parenteral administration such as intra-articular, intravenous, intramuscular, etc., by injection, transmucosal, inhalation, etc.
As the solid composition for oral administration, tablets, powders, granules and the like can be used. In such solid compositions, 1 or more than 2 active ingredients are mixed with at least one inert excipient. The compositions may contain inert additives, such as lubricants, disintegrants, stabilizers, dissolution aids, according to conventional methods. The tablets or pills may be coated with sugar coatings or films of gastric or enteric substances, as desired.
Liquid compositions for oral administration contain pharmaceutically acceptable emulsions, solutions, suspensions, syrups or elixirs and the like, containing inert diluents commonly used, such as purified water or EtOH. The liquid composition may contain, in addition to the inert diluent, an auxiliary agent such as a solubilizer, a wetting agent, a suspending agent, a sweetener, a flavoring agent, a fragrance, and a preservative.
Injections for parenteral administration contain sterile aqueous or nonaqueous solutions, suspensions or emulsions. The aqueous solvent includes, for example, distilled water for injection or physiological saline. Examples of the nonaqueous solvent include alcohols such as EtOH. Such compositions may also contain isotonic agents, preservatives, wetting agents, emulsifiers, dispersants, stabilizers or dissolution aids. They are sterilized, for example, by filtration through a bacteria-retaining filter, compounding of bactericides or irradiation. In addition, they can also be prepared as sterile solid compositions and used dissolved or suspended in sterile water or sterile injectable solvents prior to use.
The transmucosal agent such as inhalant and nasal agent may be solid, liquid or semi-solid, and may be produced by a conventionally known method. For example, a known excipient, a pH adjuster, a preservative, a surfactant, a lubricant, a stabilizer, a thickener, and the like can be appropriately added. Administration may use means for appropriate inhalation or blowing. For example, the compounds may be administered as solutions or suspensions, alone or in the form of powders forming a formulated mixture, or in combination with pharmaceutically acceptable carriers, using well-known devices such as metered dose inhalation devices or nebulizers. The dry powder inhaler and the like may be one or more dose inhalers, and dry powder or powder-containing capsules may be used. Or may be a pressurized aerosol spray using a suitable propellant, for example, a suitable gas such as chlorofluoroalkane or carbon dioxide.
In general, in the case of oral administration, the amount of administration for 1 day is suitably about 0.001 to 100mg/kg, preferably 0.1 to 30mg/kg, more preferably 0.1 to 10mg/kg per unit weight, and it is administered 1 time or divided into 2 to 4 times. In the case of intravenous administration, the amount administered per day is suitably from about 0.0001 to 10mg/kg per unit body weight, and is divided into a plurality of administrations from once a day. As the transmucosal agent, administration is carried out once a day to several times per unit weight of about 0.001 to 100 mg/kg. The amount to be administered may be appropriately determined in consideration of symptoms, age, sex, etc., according to the respective cases.
The pharmaceutical composition of the present invention contains 0.01 to 100% by weight, and in one embodiment contains 0.01 to 50% by weight of 1 or more compounds of formula (I) and formula (IA) or salts thereof as an active ingredient, although it varies depending on the route of administration, dosage form, site of administration, excipient or kind of additive.
The compounds of formula (I) and formula (IA) may be used in combination with various therapeutic or prophylactic agents for diseases in which the compounds of formula (I) and formula (IA) described above are considered to be effective. The combination may be administered simultaneously, or separately, either sequentially or at desired intervals. The simultaneous administration preparation may be a compounding agent or may be formulated separately.
Examples
The following describes in further detail the methods for producing the compounds of formula (I) and formula (IA) based on examples. The present invention is not limited to the compounds described in the following examples. The production methods of the raw material compounds are shown in the production examples. The production methods of the compounds of the formula (I) and the formula (IA) are not limited to the production methods of the specific examples shown below, and the compounds of the formula (I) and the formula (IA) may be produced by a combination of these production methods or a method obvious to those skilled in the art.
In the present specification, naming software such as ACD/Name (registered trademark, ADVANCED CHEMISTRY Development, inc.) may be used for naming the compound.
In addition, for convenience, the concentration mol/L is denoted as M. For example, 1M aqueous sodium hydroxide solution refers to 1mol/L aqueous sodium hydroxide solution.
The term "amorphous solid form" as used herein includes: the powder X-ray diffraction (XRD) pattern showed no peak morphology and a morphology with low crystallinity.
XRD was performed using Empyrean at the tube ball: cu, tube current: 40mA, tube voltage: 45kV, step size: 0.013 °, wavelength: Determination of diffraction angle range (2θ): measured at 2.5-40 deg.
Production example 1
7-Bromo-2, 4-dichloro-8-fluoro-6-iodoquinazoline (40 g) was suspended in THF (400 mL), and aqueous sodium hydroxide solution (1M, 190 mL) was added dropwise under ice cooling at an internal temperature of 10℃or less, followed by stirring for 2 hours. Into a conical flask containing hydrochloric acid (1M, 190 mL) and ice water (about 900 g), the reaction solution was poured in one portion and stirred at room temperature for about 30 minutes (until ice melted). Insoluble material was collected by filtration while washing with water, and dried under reduced pressure, whereby 7-bromo-2-chloro-8-fluoro-6-iodoquinazolin-4-ol (33.56 g) was obtained as a solid.
Production example 2
To a mixture of 7-bromo-2-chloro-8-fluoro-6-iodoquinazolin-4-ol (24.6 g) and THF (260 mL) heated to 60 ℃ was added dropwise 2-tert-butyl-1, 3-diisopropylisourea (73.4 g) under a nitrogen stream over 15 minutes. Stirred at this temperature for 2.5 hours. The mixture was cooled to room temperature and the colorless solid was filtered off while washing with THF (about 500 mL). To the solid obtained by concentrating the filtrate was added MeOH (210 mL), and the mixture was stirred at room temperature for 1 hour to be washed in suspension. The solid was filtered using MeOH (100 mL) to give 7-bromo-4-tert-butoxy-2-chloro-8-fluoro-6-iodoquinazoline (23.2 g) as a solid.
Production example 3
To a suspension of 7-bromo-4-tert-butoxy-2-chloro-8-fluoro-6-iodoquinazoline (21 g) in CH 2Cl2 (200 mL) was added ethanethiol (3.6 mL) and DABCO (7.7 g) at room temperature, and stirred overnight at room temperature under an argon atmosphere. The reaction was stopped by adding water under ice-cooling. CHCl 3 was added, the organic and aqueous layers were separated, and the aqueous layer was extracted 3 times with CHCl 3.
The collected organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 7-bromo-4-tert-butoxy-2- (ethylsulfanyl) -8-fluoro-6-iodoquinazoline (23.4 g) as a solid.
Production example 5
To a suspension of 7-bromo-4-tert-butoxy-2-chloro-8-fluoro-6-iodoquinazoline (15 g) and tetrahydro-2H-pyran-4-ol (4.67 mL) in DMF (75 mL)/THF (75 mL) under nitrogen atmosphere was added cesium carbonate (21.3 g) and DABCO (360 mg) under ice-cooling, stirred at the temperature of this state for 10 minutes, and stirred at room temperature overnight. The reaction mixture was filtered through celite (registered trademark), washed with ethyl acetate, water was added to the filtrate, and extracted 2 times with ethyl acetate. The combined organic layers were washed with a half saturated aqueous sodium chloride solution and a saturated aqueous sodium chloride solution in this order, dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure. To the resulting solid was added MeOH (50 ml) and stirred at room temperature for 30 min. The resulting solid was collected by filtration, washed with MeOH (40 ml), and dried under reduced pressure to give 7-bromo-4-tert-butoxy-8-fluoro-6-iodo-2- [ ("A") as a solidAlk-4-yl) oxy ] quinazoline (13.83 g).
Production example 6
To a solution of 7-bromo-4-tert-butoxy-2- (ethylsulfanyl) -8-fluoro-6-iodoquinazoline (32 g) and (1S) -1-phenylethan-1-ol (11 mL) in THF (400 mL) was added tBuOK (10 g) under ice cooling, and stirred under ice cooling under argon atmosphere for 1 hour. A saturated aqueous ammonium chloride solution was added under ice cooling to stop the reaction. Water and ethyl acetate were added, and the organic layer and the aqueous layer were separated, and the organic layer was washed with a saturated aqueous sodium chloride solution and dried over anhydrous sodium sulfate. Filtration and concentration under reduced pressure, and the residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give 7-bromo-4-tert-butoxy-2- (ethylsulfanyl) -6-iodo-8- [ (1S) -1-phenylethoxy ] quinazoline (36.6 g) as an oil.
Production example 9
7-Bromo-4-tert-butoxy-2- (ethylsulfanyl) -6-iodo-8- [ (1S) -1-phenylethoxy ] quinazoline (36.6 g), cyclopropylboronic acid (7.5 g), pdCl 2(dppf)·CH2Cl2 (7.6 g), potassium phosphate (53 g), meCN (440 mL) and water (80 mL) were mixed at room temperature and stirred under argon atmosphere at 90℃for 4 hours. After allowing the reaction solution to return to room temperature, it was diluted with ethyl acetate and water. The organic layer and the aqueous layer were separated, and the organic layer was washed with a saturated aqueous sodium chloride solution and dried over anhydrous sodium sulfate. Filtration and concentration under reduced pressure, and the residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give 7-bromo-4-tert-butoxy-6-cyclopropyl-2- (ethylsulfanyl) -8- [ (1S) -1-phenylethoxy ] quinazoline (22.9 g) as an oil.
PREPARATION EXAMPLE 13
To 7-bromo-4-tert-butoxy-6-cyclopropyl-2- (ethylsulfanyl) -8- [ (1S) -1-phenylethoxy ] quinazoline (14.21 g) was added 6-fluoro-5-methyl-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2- (triphenylmethyl) -2H-indazole (19.3 g), palladium (II) acetate (0.67 g), dicyclohexyl (2 ',6' -diisopropyloxy- [1,1' -biphenyl ] -2-yl) phosphine (2.67 g), anhydrous barium hydroxide (14.6 g) and DOX (500 mL)/water (100 mL), and after multiple degassing and argon substitution operations were performed, the mixture was heated and stirred overnight at 50℃under argon atmosphere. The reaction suspension which was left to cool naturally was filtered through celite (registered trademark) while being washed with ethyl acetate to remove grey insoluble matters. After concentrating the filtrate under reduced pressure to about 1/4, water was added and extraction was performed 2 times with ethyl acetate. The collected organic layer was washed with saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. Concentrated under reduced pressure and the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give 4-tert-butoxy-6-cyclopropyl-2- (ethylsulfanyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinazoline (from an about 3.3:1 diastereomer mixture of axi-asymmetry, 16.44 g) as a solid.
PREPARATION EXAMPLE 14
Allowing 7-bromo-4-tert-butoxy-6-cyclopropyl-2- [ (] at room temperatureAlk-4-yl) oxy ] -8- [ (1S) -1-phenylethoxy ] quinazoline (500 mg) and 6-fluoro-5-methyl-4- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2- (triphenylmethyl) -2H-indazole (670 mg) were dissolved in DOX (10 mL), palladium (II) acetate (22 mg), dicyclohexyl (2 ',6' -diisopropyloxy- [1,1' -biphenyl ] -2-yl) phosphine (87 mg), anhydrous barium hydroxide (870 mg) and water (1 mL) were added at room temperature and stirred for 45 minutes at 50 ℃. DOX (5 mL) and 6-fluoro-5-methyl-4- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2- (triphenylmethyl) -2H-indazole (90 mg) were added at the temperature in this state, and stirred at the temperature in this state for 30 minutes. The reaction mixture was cooled naturally, ethyl acetate and celite (registered trademark) were added, and after stirring at room temperature for 30 minutes, the reaction solution was filtered through celite (registered trademark), and the filtrate was concentrated. The resulting residue was purified by silica gel chromatography (basic silica gel, hexane/ethyl acetate). To the resulting solid was added iPrOH (40 mL), stirred at 50℃for 1 hour, at room temperature for 2 hours, and the resulting solid was collected by filtration to give 4-tert-butoxy-6-cyclopropyl-7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -2- [ (-2- [ ] as a solidAlk-4-yl) oxy ] -8- [ (1S) -1-phenylethoxy ] quinazoline (from an axi-asymmetric about 1:1, a mixture of diastereomers of formula 1 296 mg). Concentrating the filtrate to obtain the target compound (7M) -4-tert-butoxy-6-cyclopropyl-7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -2- [ (-) -2- [ ] in solid formAlk-4-yl) oxy ] -8- [ (1S) -1-phenylethoxy ] quinazoline (single diastereomer, 409 mg).
Production example 15
(3S) -3- ({ 7-bromo-6-cyclopropyl-2- (ethylsulfanyl) -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } oxy) pyrrolidine-1-carboxylic acid tert-butyl ester (2.71 g), 6-fluoro-5-methyl-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -2- (triphenylmethyl) -2H-indazole (3.44 g), dicyclohexyl (2 ',6' -diisopropyloxy- [1,1' -biphenyl ] -2-yl) phosphine (531 mg), palladium (II) acetate (106 mg), anhydrous barium hydroxide (2.29 g) were suspended in DOX (110 mL) and water (22 mL) under an argon atmosphere, and stirred at 50℃for 15 minutes. 6-fluoro-5-methyl-4- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2- (triphenylmethyl) -2H-indazole (1.6 g) was added and stirred at 50℃for a further 15 minutes. Cooled to room temperature, ethyl acetate and water were added, insoluble matters were removed by filtration through celite (registered trademark), and two layers of the filtrate were separated. The aqueous layer was extracted with ethyl acetate, and the combined organic layers were washed with saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. After filtering off the drying agent, concentration under reduced pressure, the residue was purified by silica gel column chromatography (basic silica gel, hexane/ethyl acetate) to give (3S) -3- ({ 6-cyclopropyl-2- (ethylsulfanyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } oxy) pyrrolidine-1-carboxylic acid tert-butyl ester (from an about 5:1 diastereomeric mixture of axial asymmetry, 3.88 g) as an oil.
To tert-butyl (3S) -3- ({ 6-cyclopropyl-2- (ethylsulfanyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } oxy) pyrrolidine-1-carboxylate (from a mixture of diastereomers of about 5:1, which were axi-asymmetric, 100 mg) was added MeOH (2 mL) and heated to 70℃to dissolve it. Stir at room temperature overnight for a further 3 days. The precipitation of solids (pale yellow suspension) was confirmed. The solid was filtered off and washed with a small amount of MeOH to give (3S) -3- ({ 6-cyclopropyl-2- (ethylsulfanyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } oxy) pyrrolidine-1-carboxylic acid tert-butyl ester (from an axiasymmetric about 1:1 mixture of diastereomers, 21 mg) as a powder. The filtrate was concentrated to give the target tert-butyl (3S) -3- ({ 6-cyclopropyl-2- (ethylsulfanyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } oxy) pyrrolidine-1-carboxylate (single diastereomer of unknown configuration, 70 mg) as a solid.
Likewise, to tert-butyl (3S) -3- ({ 6-cyclopropyl-2- (ethylsulfanyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } oxy) pyrrolidine-1-carboxylate (from a mixture of diastereomers of about 5:1, from the axis asymmetry, 2.95 g) was added EtOH (6 mL) and heated to 70deg.C to dissolve it. A trace of the powder of the axiasymmetric mixture (obtained as described above) was added at room temperature and stirred at room temperature overnight. The precipitated solid was collected by filtration and washed with a small amount of EtOH to give (3S) -3- ({ 6-cyclopropyl-2- (ethylsulfanyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } oxy) pyrrolidine-1-carboxylic acid tert-butyl ester (from an axi-asymmetric about 1:1 mixture of diastereomers, 699 mg) as a colorless powder. The filtrate was concentrated to give the target tert-butyl (3S) -3- ({ 6-cyclopropyl-2- (ethylsulfanyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } oxy) pyrrolidine-1-carboxylate (single diastereomer of unknown configuration, 2.33 g) as a foamy solid.
Production example 17
4-Tert-butoxy-6-cyclopropyl-2- (ethylsulfanyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinazoline (diastereomer mixture from about 3.3:1 of axial asymmetry, 33.71 g) was dissolved in CH 2Cl2 (500 mL), and m-chloroperbenzoic acid (containing about 30% and 22.4 g) (inner temperature: 5-10 ℃ C.) was added under ice cooling, and stirred at room temperature for 2 hours. To the reaction solution, an aqueous solution (300 mL) of sodium thiosulfate pentahydrate (11 g) and a saturated aqueous solution (300 mL) of sodium hydrogencarbonate were poured under ice-cooling, and after stirring at room temperature for 30 minutes, the mixture was extracted 2 times with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium bicarbonate and saturated aqueous sodium chloride, dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure to give 4-tert-butoxy-6-cyclopropyl-2- (ethanesulfonyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinazoline (from an axiasymmetric diastereomer mixture). To the resulting residue was added iPrOH (1000 mL), and stirred at room temperature overnight. The resulting solid was collected by filtration, washed with iPrOH, and dried under reduced pressure to give 4-tert-butoxy-6-cyclopropyl-2- (ethanesulfonyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinazoline (about 1:1 diastereomer mixture from axial asymmetry, 11.52 g) as a solid. The filtrate was concentrated under reduced pressure to give the targeted (7M) -4-tert-butoxy-6-cyclopropyl-2- (ethanesulfonyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinazoline (single diastereomer, 23.29 g) as a solid.
Production example 19
To a solution of (7M) -4-tert-butoxy-6-cyclopropyl-2- (ethanesulfonyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinazoline (2.00 g) in THF (50 mL) under a nitrogen atmosphere was added 4-methylbenzene-1-sulfonic acid monohydrate (230 mg) at room temperature and stirred at 50 ℃ for 1 hour. After the reaction solution was naturally cooled to room temperature, TEA (350. Mu.L) was added, followed by concentration under reduced pressure. The resulting residue was purified by silica gel chromatography (basic silica gel, hexane/ethyl acetate) to give (7M) -4-tert-butoxy-6-cyclopropyl-2- (ethanesulfonyl) -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -8- [ (1S) -1-phenylethoxy ] quinazoline (740 mg) as a foamy solid.
Production example 20
To (7M) -4-tert-butoxy-6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -2- [ (-d eTo an alk-4-yloxy ] quinazoline (2.11 g) was added THF (20 mL), and methanesulfonic acid (480. Mu.L) was added under ice-cooling, followed by stirring at room temperature for 4 hours. The reaction was stopped by pouring into a saturated aqueous sodium bicarbonate solution, and extraction was performed 3 times with ethyl acetate. The combined organic layers were dried over anhydrous magnesium sulfate, filtered, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (CHCl 3/MeOH). Purifying again by silica gel column chromatography (basic silica gel, CHCl 3/MeOH) to obtain (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [. Sup.th-of-the-day in solid formAlk-4-yl) oxy ] quinazolin-4-ol (1.02 g).
Production example 21
To a solution of (7M) -4-tert-butoxy-6-cyclopropyl-2- (ethanesulfonyl) -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -8- [ (1S) -1-phenylethoxy ] quinazoline (735 mg) in THF (10 mL) under a nitrogen atmosphere was added 3, 4-dinitro-2H-pyran (900 μl) and 4-methylbenzene-1-sulfonic acid monohydrate (35 mg) at room temperature. Stirred at room temperature overnight and the reaction was concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography (basic silica gel, hexane/ethyl acetate) to give (7M) -4-tert-butoxy-6-cyclopropyl-2- (ethanesulfonyl) -7- [ 6-fluoro-5-methyl-1- ] as a foamy solidAlk-2-yl) -1H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinazoline (735 mg).
Production example 23
Under argon atmosphere, to (7M) -4-tert-butoxy-6-cyclopropyl-2- (ethylsulfonyl) -7- [ 6-fluoro-5-methyl-1- ]To a solution of alk-2-yl) -1H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinazoline (730 mg) in MeOH (15 mL)/THF (15 mL) was added sodium bicarbonate (450 mg) and 10% Pd/C (aqueous about 50%, 250 mg) at room temperature. Stirring was carried out overnight at room temperature under normal pressure under a hydrogen atmosphere. After the substitution with argon gas, the reaction solution was filtered through celite (registered trademark) and washed with ethyl acetate. Concentrating the filtrate under reduced pressure to obtain (7M) -4-tert-butoxy-6-cyclopropyl-2- (ethanesulfonyl) -7- [ 6-fluoro-5-methyl-1- ] as a foamy solidAlk-2-yl) -1H-indazol-4-yl ] quinazolin-8-ol (630 mg).
PREPARATION EXAMPLE 25
To (3S) -3- [ { (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ]To tert-butyl (1.90 g) of alk-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinazolin-4-yl } (meth) amino ] pyrrolidine-1-carboxylate was added MeOH (30 mL), 10% Pd/C (aqueous about 50%, 1.2 g) was added, and the mixture was stirred overnight at room temperature under a hydrogen atmosphere. The reaction mixture was filtered through celite (registered trademark), and the filtrate was concentrated under reduced pressure and azeotroped with EtOH. Purifying the obtained residue by silica gel column chromatography (hexane/ethyl acetate), and obtaining (3S) -3- [ { (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ] as a foamy solidAlkan-2-yl) -1H-indazol-4-yl ] -8-hydroxy-2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (1.5 g).
Production example 29
Under nitrogen flow, (7M) -4-tert-butoxy-6-cyclopropyl-2- (ethylsulfonyl) -7- [ 6-fluoro-5-methyl-1- ] is addedThe alk-2-yl) -1H-indazol-4-yl ] quinazolin-8-ol (1.4 g) and cesium carbonate (2.4 g) were suspended in DMF (17 mL), 1- (chloromethyl) -4-ethynylbenzene (500 mg) was added at room temperature, and stirred at 60℃for 1.5 hours. To the reaction mixture was added water (100 ml), and extracted 2 times with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate, filtered, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (7M) -4-tert-butoxy-6-cyclopropyl-2- (ethanesulfonyl) -8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as a foamy solidAlk-2-yl) -1H-indazol-4-yl ] quinazoline (1.47 g).
Production example 30
Under nitrogen atmosphere, to (3S) -3- [ { (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ]To a mixture of tert-butyl (150 mg) of alk-2-yl) -1H-indazol-4-yl ] -8-hydroxy-2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (meth) amino ] pyrrolidine-1-carboxylate, 4- (chloromethyl) -1-ethynyl-2-fluorobenzene (52 mg) and DMF (2 mL) was added cesium carbonate (130 mg) at room temperature and stirred at 60℃for 1 hour. After the reaction mixture was naturally cooled to room temperature, ethyl acetate was added, the organic layer was washed with water and a saturated aqueous sodium chloride solution, dried over anhydrous magnesium sulfate, insoluble matter was filtered off, and the filtrate was concentrated under reduced pressure. Purifying the obtained residue by silica gel column chromatography (basic silica gel, hexane/ethyl acetate), and obtaining (3S) -3- [ { (7M) -6-cyclopropyl-8- [ (4-ethynyl-3-fluorophenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] in solid formAlkan-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (meth) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (117 mg).
Production example 36
To a solution of (7M) -4-tert-butoxy-6-cyclopropyl-2- (ethanesulfonyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinazoline (5.01 g) and (2S) -2-methoxypropane-1-ol (0.94 mL) in THF (60 mL) under nitrogen atmosphere was added tBuOK (1.00 g) under ice cooling, and stirred at this temperature for 30 minutes. Ice water and an aqueous ammonium chloride solution were added to the reaction solution to stop the reaction. The organic layer was washed with saturated aqueous sodium chloride solution, extracted 2 times with ethyl acetate, and dried over anhydrous magnesium sulfate. Purification by silica gel column chromatography (hexane/ethyl acetate) gave (7M) -4-tert-butoxy-6-cyclopropyl-7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinazoline (4.07 g) as a foamy solid.
Production example 54
Under nitrogen atmosphere, to (7M) -4-tert-butoxy-6-cyclopropyl-2- (ethylsulfonyl) -8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ]To a solution of alk-2-yl) -1H-indazol-4-yl ] quinazoline (1.47 g) and (2S) -2-methoxypropan-1-ol (0.25 mL) in THF (11 mL) was added tBuOK (275 mg) with cooling from an ice/MeOH bath (-20 ℃ C. To-15 ℃ C.) and stirring at this temperature for 30 min. To the reaction mixture, a saturated aqueous ammonium chloride solution was added at this temperature, and extraction was performed 2 times with ethyl acetate. The combined organic layers were washed with water and saturated brine, dried over anhydrous magnesium sulfate, and filtered. Concentrating the filtrate under reduced pressure to obtain (7M) -4-tert-butoxy-6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as a foamy solidAlk-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazoline (1.43 g).
Production example 57
Under argon atmosphere, to (7M) -4-tert-butoxy-6-cyclopropyl-2- (ethylsulfonyl) -8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1-To a solution of alk-2-yl) -1H-indazol-4-yl ] quinazoline (400 mg) and [ (2R) -oxolan-2-yl ] methanol (82 mg) in THF (8 mL) was added tBuOK (85 mg) under ice-cooling, and stirred at this temperature for 30 minutes. To the reaction mixture was added a saturated aqueous ammonium chloride solution, and extracted 2 times with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride, dried over anhydrous magnesium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (7M) -4-tert-butoxy-6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as a foamy solidAlk-2-yl) -1H-indazol-4-yl ] -2- { [ (2R) -oxolan-2-yl ] methoxy } quinazoline (261 mg).
Production example 58
Under argon atmosphere, to (7M) -4-tert-butoxy-6-cyclopropyl-2- (ethylsulfonyl) -8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1-To a solution of alk-2-yl) -1H-indazol-4-yl ] quinazoline (400 mg) and [ (2S) -oxolan-2-yl ] methanol (82 mg) in THF (8 mL) was added tBuOK (85 mg) under ice-cooling, and stirred at this temperature for 30 minutes. To the reaction mixture was added a saturated aqueous ammonium chloride solution, and extracted 2 times with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride, dried over anhydrous magnesium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (7M) -4-tert-butoxy-6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as a foamy solidAlk-2-yl) -1H-indazol-4-yl ] -2- { [ (2S) -oxolan-2-yl ] methoxy } quinazoline (330 mg).
Production example 59
To (7M) -4-tert-butoxy-6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] under nitrogen atmosphereTo a solution of alk-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazoline (1.43 g) in THF (15 mL) was added 4-methylbenzene-1-sulfonic acid monohydrate (120 mg) at room temperature and stirred at 50℃for 3 hours. The reaction solution was concentrated under reduced pressure, and the obtained residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as a foam solidAlk-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-ol (1.05 g).
Production example 60
To (7M) -4-tert-butoxy-6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1-To a solution of alk-2-yl) -1H-indazol-4-yl ] -2- { [ (2R) -oxolan-2-yl ] methoxy } quinazoline (261 mg) in THF (5 mL) was added 4-methylbenzene-1-sulfonic acid monohydrate (10 mg) and 3, 4-dinitro-2H-pyran (1 mL) at room temperature, and stirred at that temperature overnight. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (basic silica gel, CHCl 3/MeOH) to give (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as a foamy solidAlk-2-yl) -1H-indazol-4-yl ] -2- { [ (2R) -oxolan-2-yl ] methoxy } quinazolin-4-ol (215 mg).
Production example 61
To (7M) -4-tert-butoxy-6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1-To a solution of alk-2-yl) -1H-indazol-4-yl ] -2- { [ (2S) -oxolan-2-yl ] methoxy } quinazoline (330 mg) in THF (5 mL) was added 4-methylbenzene-1-sulfonic acid monohydrate (11 mg) and 3, 4-dinitro-2H-pyran (1 mL) at room temperature, and stirred at that temperature overnight. The reaction mixture was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (basic silica gel, CHCl 3/MeOH) to give (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as a foamy solidAlk-2-yl) -1H-indazol-4-yl ] -2- { [ (2S) -oxolan-2-yl ] methoxy } quinazolin-4-ol (282 mg).
Production example 62
Under the atmosphere of nitrogen, to (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ]To a solution of alk-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinazolin-4-ol (1.63 g) in THF (20 mL) was added cesium carbonate (4.25 g) and PyBOP (2 g) in this order at room temperature, and the mixture was stirred at room temperature for 1 hour. Tert-butyl (3S) -3- (methylamino) pyrrolidine-1-carboxylate (1.58 g) was added at room temperature and stirred at 60℃for 2 hours. After the reaction mixture was cooled to room temperature, ethyl acetate was added thereto, and then it was filtered through celite (registered trademark). Concentrating the filtrate under reduced pressure, purifying the obtained residue by silica gel column chromatography (basic silica gel, hexane/ethyl acetate), and obtaining (3S) -3- [ { (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ] as an oilAlkan-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (1.9 g).
Production example 63
Under the atmosphere of nitrogen, to (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ]To a solution of alk-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-ol (200 mg) in THF (4 mL) was added PyBOP (335 mg) and cesium carbonate (205 mg) at room temperature, and the mixture was stirred at room temperature for 1 hour. A solution of (3R) -3- (methylamino) pyrrolidine-1-carboxylic acid tert-butyl ester (300 mg) in THF (1 mL) and cesium carbonate (500 mg) were added at room temperature and stirred at 60℃for 2 hours. After the reaction mixture was cooled to room temperature, ethyl acetate was added thereto, and then it was filtered through celite (registered trademark). The filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (basic silica gel, hexane/ethyl acetate) to give (3R) -3- [ { (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as an oil containing impuritiesAlkan-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (meth) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (313 mg).
PREPARATION EXAMPLE 73
Under the atmosphere of nitrogen, to (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ]To a solution of alk-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-ol (1.05 g) in THF (15 mL) was added cesium carbonate (2.68 g) and PyBOP (1.20 g) in this order at room temperature, and the mixture was stirred at room temperature for 1 hour. Tert-butyl (3S) -3- (methylamino) pyrrolidine-1-carboxylate (0.94 mL) was added at room temperature and stirred overnight at 60 ℃. Ethyl acetate was added to the reaction solution, and the insoluble material was filtered through celite (registered trademark). The filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (basic silica gel, hexane/ethyl acetate) to give (3S) -3- [ { (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as an oilAlkan-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (meth) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (1.62 g).
Production example 74
Under the atmosphere of nitrogen, to (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ]To a solution of alk-2-yl) -1H-indazol-4-yl ] -2- { [ (2R) -oxolan-2-yl ] methoxy } quinazolin-4-ol (106 mg) in THF (4 mL) was added cesium carbonate (265 mg) and PyBOP (120 mg) in this order at room temperature, and the mixture was stirred at room temperature for 1 hour. Tert-butyl (3S) -3- (methylamino) pyrrolidine-1-carboxylate (62. Mu.L) was added at room temperature and stirred overnight at room temperature. Tert-butyl (3S) -3- (methylamino) pyrrolidine-1-carboxylate (100. Mu.L) was added and stirred at 80℃for 2 hours. Ethyl acetate was added to the reaction solution, and insoluble matters were filtered off through celite (registered trademark). The filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (basic silica gel, hexane/ethyl acetate) to give (3S) -3- { [ (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as an oilAlkan-2-yl) -1H-indazol-4-yl ] -2- { [ (2R) -oxolan-2-yl ] methoxy } quinazolin-4-yl ] (methyl) amino } pyrrolidine-1-carboxylic acid tert-butyl ester (211 mg).
Production example 75
Under the atmosphere of nitrogen, to (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ]To a solution of alk-2-yl) -1H-indazol-4-yl ] -2- { [ (2S) -oxolan-2-yl ] methoxy } quinazolin-4-ol (164 mg) in THF (4 mL) was added cesium carbonate (410 mg) and PyBOP (185 mg) in this order at room temperature, and stirred at room temperature for 1 hour. (3S) -3- (methylamino) pyrrolidine-1-carboxylic acid tert-butyl ester (96. Mu.L) was added and stirred overnight at room temperature. Tert-butyl (3S) -3- (methylamino) pyrrolidine-1-carboxylate (100. Mu.L) was added and stirred at 80℃for 2 hours. Ethyl acetate was added to the reaction solution, insoluble matters were filtered off through celite (registered trademark), and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (basic silica gel, hexane/ethyl acetate) to give (3S) -3- { [ (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as an oilAlkan-2-yl) -1H-indazol-4-yl ] -2- { [ (2S) -oxolan-2-yl ] methoxy } quinazolin-4-yl ] (methyl) amino } pyrrolidine-1-carboxylic acid tert-butyl ester (199 mg).
Production example 79
Under the atmosphere of nitrogen, to (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ]To a solution of alk-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-ol (100 mg) in THF (2 mL) was added PyBOP (165 mg), cesium carbonate (255 mg) in this order at room temperature, and the mixture was stirred at room temperature for 1 hour. Cesium carbonate (105 mg) and tert-butyl (3S) -3-aminopyrrolidine-1-carboxylate (130. Mu.L) were added at room temperature, and stirred at room temperature overnight. Ethyl acetate was added to the reaction solution, and insoluble materials were filtered through celite (registered trademark). The filtrate was concentrated, and the resulting residue was purified by silica gel column chromatography (basic silica gel, hexane/ethyl acetate) to give (3S) -3- ({ (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as an oilAlkan-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } amino) pyrrolidine-1-carboxylic acid tert-butyl ester (143 mg).
Production example 83
Under nitrogen, to (3R) -3- [ { (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] under an atmosphere of nitrogenTo a solution of tert-butyl (311 mg) of alk-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (meth) amino ] pyrrolidine-1-carboxylate and (4R) -1- [ (2S) -2-azido-3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (170 mg) in tBuOH (3 mL), THF (3 mL) and water (3 mL) were added sodium ascorbate (110 mg) and anhydrous copper (II) sulfate (30 mg) at room temperature and stirred at room temperature for 2 hours. An aqueous solution (13 mL) of disodium ethylenediamine tetraacetate (650 mg) was added, and after stirring at room temperature for 30 minutes, the mixture was extracted with ethyl acetate. The organic layer was washed with water and saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (basic silica gel, CHCl 3/MeOH) to give (3R) -3- [ { (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ] as a solidAlkylin-2-yl) -1H-indazol-4-yl ] -8- { [4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (312 mg).
Production example 84
To (3S) -3- [ { (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] aTo a mixture of tert-butyl (133 mg), (4R) -1- [ (2S) -2-azido-3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (80 mg), tBuOH (1.2 mL), THF (1.2 mL) and water (1.2 mL) was added sodium ascorbate (66 mg) and anhydrous copper (II) sulfate (27 mg) at room temperature, and stirred at room temperature for 3 hours. 2% aqueous ethylenediamine tetraacetic acid disodium salt solution and saturated aqueous sodium chloride solution were added, extraction was performed 2 times with CH 2Cl2, and the combined organic layers were dried over anhydrous magnesium sulfate. Insoluble matter was filtered off, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (basic silica gel, CHCl 3/MeOH) to give (3S) -3- [ { (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ] as a solidAlkylin-2-yl) -1H-indazol-4-yl ] -8- { [4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (ethyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (178 mg).
Production example 92
To (3S) -3- [ { (7M) -6-cyclopropyl-8- [ (4-ethynyl-3-fluorophenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ]To a solution of tert-butyl (2S) -2-methoxypropoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylate (115 mg) and (4R) -1- [ (2S) -2-azido-3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (70 mg) in tBuOH (1 mL), THF (1 mL) and water (1 mL) was added sodium ascorbate (41 mg) and anhydrous copper (II) sulfate (11 mg) at room temperature and the mixture was stirred at room temperature for 1 hour. An aqueous solution (5 mL) of disodium ethylenediamine tetraacetate (230 mg) was added thereto, and the mixture was stirred at room temperature for 30 minutes, followed by extraction with ethyl acetate. The organic layer was washed with water and saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (basic silica gel, CHCl 3/MeOH) to give (3S) -3- [ { (7M) -6-cyclopropyl-8- { [ 3-fluoro-4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -7- [ 6-fluoro-5-methyl-1- ] in solid formAlkan-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (meth) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (171 mg).
Production example 114
To (4R) -1- [ (2S) -2-azido-3-methylbutanoyl ] -N- { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } -4-hydroxy-L-prolinamide (80 mg), (3S) -3- [ { (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1-Alkyl-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (125 mg) tBuOH/THF/water (1: 1:1, 2.0 mL) was added with anhydrous copper (II) sulfate (10 mg) and sodium ascorbate (30 mg) at room temperature, and stirred at room temperature for 1 hour. To the reaction mixture was added ethylenediamine tetraacetic acid disodium salt (175 mg) at room temperature, and stirred at that temperature for 30 minutes. Water is added to the reaction mixture, stirred, and the resulting insoluble matter is collected by filtration to give (3S) -3- [ { (7M) -6-cyclopropyl-8- { [4- (1- { (2S) -1- [ (2S, 4R) -2- ({ (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } carbamoyl) -4-hydroxypyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -7- [ 6-fluoro-5-methyl-1- ] as a solidAlkan-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (meth) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (225 mg).
Production example 115
To (4R) -1- [ (2S) -2-azido-3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (120 mg) and (3S) -3- { [ (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] under an argon atmosphereTo a solution of tert-butyl (2S) -oxolan-4-yl ] -2- { [ (2S) -oxolan-2-yl ] methoxy } quinazolin-4-yl ] (methyl) amino } pyrrolidine-1-carboxylate (199 mg) in tBuOH (1 mL), THF (1 mL) and water (1 mL), anhydrous copper (II) sulfate (18 mg) and sodium ascorbate (80 mg) were added at room temperature, and the mixture was stirred at room temperature for 1 hour. To the reaction mixture was added ethylenediamine tetraacetic acid disodium salt (300 mg) at room temperature, and the mixture was stirred at that temperature for 1 hour. Water and ethyl acetate were added, and extraction was performed with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (basic silica gel, CHCl 3/MeOH) to give (3S) -3- { [ (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ] as a foamy solidAlkan-2-yl) -1H-indazol-4-yl ] -8- { [4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2 (2S) -oxopyrrolidin-2-yl ] methoxy } quinazolin-4-yl ] (methyl) amino } pyrrolidine-1-carboxylic acid tert-butyl ester (236 mg).
Production example 116
To (4R) -1- [ (2S) -2-azido-3-methylbutyryl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (75 mg) and (3S) -3- [ { (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] under an atmosphere of argonAlkyl-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (230 mg) tBuOH/THF/water (1: 1:1, 2.0 mL) was added with anhydrous copper (II) sulfate (10 mg) and sodium ascorbate (30 mg) at room temperature, and stirred at room temperature for 1 hour. To the reaction mixture was added ethylenediamine tetraacetic acid disodium salt (175 mg) at room temperature, and the mixture was stirred at that temperature for 1 hour. To the reaction mixture was added water and CHCl 3/MeOH (9/1). The aqueous and organic layers were separated and the aqueous layer was extracted with CHCl3/MeOH (9/1). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (basic silica gel, CHCl 3/MeOH) to give (3S) -3- [ { (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ] as a foamy solidAlkylin-2-yl) -1H-indazol-4-yl ] -8- { [4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (197 mg).
Production example 117
To (4R) -1- [ (2S) -2-azido-3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (75 mg) and (3S) -3- { [ (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] under an argon atmosphereTo a solution of tert-butyl (2R) -oxolan-2-yl ] -2- { [ (2R) -oxolan-2-yl ] methoxy } quinazolin-4-yl ] (methyl) amino } pyrrolidine-1-carboxylate (123 mg) in tBuOH (1 mL), THF (1 mL) and water (1 mL), anhydrous copper (II) sulfate (11 mg) and sodium ascorbate (50 mg) were added at room temperature, and the mixture was stirred at room temperature for 1 hour. To the reaction mixture was added ethylenediamine tetraacetic acid disodium salt (200 mg) at room temperature, and the mixture was stirred at that temperature for 1 hour. Water and ethyl acetate were added, and extraction was performed with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (basic silica gel, CHCl 3/MeOH) to give (3S) -3- { [ (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ] as a foamy solidAlkan-2-yl) -1H-indazol-4-yl ] -8- { [4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2 (2R) -oxopyrrolidin-2-yl ] methoxy } quinazolin-4-yl ] (methyl) amino } pyrrolidine-1-carboxylic acid tert-butyl ester (156 mg).
Production example 124
To (3S) -3- ({ (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (-d-a-M)To tert-butyl (75 mg) of alkyl-4-yl) oxy ] quinazolin-4-yl } amino) pyrrolidine-1-carboxylate were added tBuOH (1 mL), THF (1 mL) and water (1 mL), and (4R) -1- [ (2S) -2-azido-3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (50 mg), copper (I) (10 mg) and sodium ascorbate (30 mg) were added while stirring at room temperature, and the mixture was stirred at 50℃for 3 hours. The reaction solution was cooled naturally, and 2% disodium edetate aqueous solution and ethyl acetate were added thereto, followed by vigorous stirring for 30 minutes. The organic layer was washed with saturated aqueous sodium chloride solution, dried over anhydrous magnesium sulfate, and concentrated under reduced pressure. The residue was dissolved in THF (1 mL) and water (0.10 mL), tris (2-carboxyethyl) phosphine hydrochloride (10 mg) was added and stirred overnight at room temperature, the solvent was distilled off, and the resulting residue was purified by silica gel column chromatography (basic silica gel, CHCl 3/MeOH) to give (3S) -3- ({ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -8- { [4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2- [ -phenyl ] methoxy } -2- ] as a solidAlkyl-4-yl) oxy ] quinazolin-4-yl } amino) pyrrolidine-1-carboxylic acid tert-butyl ester (77 mg).
Production example 125
To a solution of tert-butyl [ (1R) -1- (4-bromophenyl) -2-hydroxyethyl ] carbamate (5.01 g) in DMAc (80 mL) was added 4-methyl-1, 3-thiazole (2.88 mL) and potassium acetate (3.11 g) at room temperature, each of which was degassed and replaced with argon 3 times, and then palladium acetate (356 mg) was added at room temperature and stirred at 100℃for 16 hours. After cooling at room temperature, ethyl acetate and water were added to the reaction mixture, and insoluble matter was removed by filtration through celite (registered trademark). To the filtrate was added water, and the aqueous layer was extracted 3 times with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride solution and dried over anhydrous sodium sulfate. Filtration, concentration, and purification of the residue by silica gel column chromatography (basic silica gel, hexane/ethyl acetate) gave tert-butyl { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamate (4.66 g) as a solid.
Production example 126
To a solution of tert-butyl { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamate (4.41 g) in CH 2Cl2 (50 mL) and MeOH (40 mL) was added a small amount of hydrogen chloride (4M DOX solution, 20 mL) at a time under ice-cooling, and the mixture was stirred at room temperature for 6 hours. Diethyl ether was added to the reaction mixture, and the solid was collected by filtration, washed with diethyl ether and dried under reduced pressure to give (2R) -2-amino-2- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethan-1-ol n hydrochloride (2.12 g) as a solid. The filtrate was concentrated under reduced pressure, and dried under reduced pressure with heating to give (2R) -2-amino-2- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethan-1-ol n hydrochloride (2.01 g) as a solid.
Production example 131
To a solution of tert-butyl { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } carbamate (3.34 g) in CH 2Cl2 (25 mL) and MeOH (25 mL) was added hydrogen chloride (4M DOX solution, 25.6 mL) at-20℃to-10℃under cooling, and the mixture was stirred at room temperature for 5 hours. The reaction mixture was concentrated under reduced pressure to give (2R) -2-amino-2- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] ethan-1-ol n-hydrochloride (3.06 g) as a solid.
Production example 133
A mixture of (2R) -2-amino-2- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethan-1-ol n hydrochloride (2.12 g), (4R) -1- (tert-butoxycarbonyl) -4-hydroxy-L-proline (1.76 g) and DMF (22 mL) was cooled with ice under an argon gas stream, DIPEA (4.7 mL) was added, and then HATU (3.02 g) was added in small amounts each time so that the internal temperature was maintained at 5℃or lower. The reaction mixture was stirred under ice-cooling for 1 hour, and at room temperature for 1 hour. The reaction mixture was cooled with ice, and after adding water (120 mL) and a saturated aqueous sodium chloride solution (50 mL), it was extracted 3 times with ethyl acetate. The aqueous layer was further extracted 3 times with ethyl acetate/iPrOH (9/1), and the combined organic layers were washed with saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. Concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (CHCl 3/MeOH) to give (2 s, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidine-1-carboxylic acid tert-butyl ester (3.09 g) as an oil.
Production example 134
To a mixture of (2R) -2-amino-2- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] ethan-1-ol n hydrochloride (3.43 g), (4R) -1- (tert-butoxycarbonyl) -4-hydroxy-L-proline (2.81 g) and DMF (40 mL) was added DIPEA (7.8 mL) under ice cooling, HATU (4.5 g) was added small amounts each time under ice cooling, stirred under ice cooling for 1 hour, and stirred at room temperature for 1 hour. Water, a saturated aqueous sodium chloride solution and ethyl acetate were added under ice cooling, and the aqueous layer was separated. After the aqueous layer was extracted with ethyl acetate, it was extracted with ethyl acetate/iPrOH (9/1), and the organic layer was washed with a saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. Insoluble matter was filtered off, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (CHCl 3/MeOH) to give (2 s, 4R) -2- ({ (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } carbamoyl) -4-hydroxypyrrolidine-1-carboxylic acid tert-butyl ester (5.01 g) as an oil.
Production example 135
To a solution of tert-butyl (2 s, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidine-1-carboxylate (3.09 g) in CH 2Cl2 (18 mL), meOH (18 mL) was added hydrogen chloride (4M DOX solution, 17 mL) under ice cooling, stirred under ice cooling for 1 hour, and stirred at room temperature for 4 hours. Concentrated under reduced pressure and dried to give (4R) -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide N-hydrochloride (2.92 g) as a solid.
Production example 136
To a solution of (2 s, 4R) -2- ({ (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } carbamoyl) -4-hydroxypyrrolidine-1-carboxylic acid tert-butyl ester (5.01 g) in CH 2Cl2 (35 mL), meOH (30 mL) was added hydrogen chloride (4M DOX solution, 28 mL) under cooling at-20 to-10 ℃ and stirred at room temperature for 5 hours. The reaction mixture was concentrated under reduced pressure to give (4R) -N- { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } -4-hydroxy-L-prolinamide N hydrochloride (4.71 g) as a solid.
Production example 137
To a mixture of (4R) -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide N hydrochloride (3.81 g), N- (tert-butoxycarbonyl) -L-valine (2.16 g) and DMF (45 mL) was added DIPEA (6.2 mL), and HATU (3.61 g) was added in small amounts each time under ice cooling, stirred under ice cooling for 1 hour, and stirred at room temperature for 1 hour. Water, a saturated aqueous sodium chloride solution and ethyl acetate were added under ice cooling, and the aqueous layer was separated. After extracting the aqueous layer with ethyl acetate, it was extracted with ethyl acetate/iPrOH (9/1), and the combined organic layers were washed with saturated aqueous sodium chloride solution and dried over anhydrous sodium sulfate. Insoluble matter was filtered off, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (CHCl 3/MeOH) to give N- (tert-butoxycarbonyl) -L-valyl- (4R) -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (4.43 g) as a solid.
Production example 138
To a solution of (4R) -N- { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } -4-hydroxy-L-prolinamide N hydrochloride (4.7 g), N- (tert-butoxycarbonyl) -L-valine (2.7 g) in DMF (55 mL) was added DIPEA (7.7 mL), and HATU (4.5 g) was added in small portions each time under ice cooling. After stirring for 1 hour under ice-cooling and stirring for 1 hour at room temperature, water/saturated brine (1/1) and ethyl acetate were added thereto under ice-cooling, and the mixture was separated. The aqueous layer was extracted 2 times with ethyl acetate and 2 times with ethyl acetate/iPrOH (9/1). The combined organic layers were washed with saturated aqueous sodium chloride solution and dried over anhydrous sodium sulfate. Insoluble matter was filtered off, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (CHCl 3/MeOH) to give N- (tert-butoxycarbonyl) -L-valyl- (4R) -N- { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } -4-hydroxy-L-prolinamide (5.01 g) as a solid.
Production example 139
To a solution of N- (tert-butoxycarbonyl) -L-valyl- (4R) -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (4.43 g) in CH 2Cl2 (35 mL), meOH (35 mL) was added hydrogen chloride (4 MDOX solution, 20 mL) at-20-15℃under cooling, and the mixture was stirred at room temperature for 6 hours. The reaction mixture was concentrated under reduced pressure to give L-valyl- (4R) -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide N-hydrochloride (4.21 g) as a solid.
Production example 140
To a solution of N- (tert-butoxycarbonyl) -L-valyl- (4R) -N- { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } -4-hydroxy-L-prolinamide (4.92 g) in CH 2Cl2 (30 mL) and MeOH (30 mL) was slowly added hydrogen chloride (4M DOX solution, 22 mL) at-20 to-15℃under cooling, and the mixture was stirred at room temperature for 5 hours. Concentrated under reduced pressure to give L-valyl- (4R) -N- { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } -4-hydroxy-L-prolinamide N hydrochloride (5.18 g) as a solid.
Production example 141
To a mixture of L-valyl- (4R) -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide N hydrochloride (1.71 g), TEA (3.2 mL), THF (20 mL), meCN (20 mL), 2-azido-1, 3-dimethylimidazole was added in small portions each time for 10 minutes or more under ice coolingA solution of hexafluorophosphate (1.06 g) in MeCN (5 mL) was stirred under ice-cooling for 5 hours. Water, saturated aqueous sodium chloride solution and ethyl acetate were added, and the aqueous layer and the organic layer were separated. The aqueous layer was extracted with ethyl acetate, and the combined organic layers were dried over anhydrous sodium sulfate. Insoluble matter was filtered off, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (CHCl 3/MeOH) to give (4R) -1- [ (2S) -2-azido-3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (1.07 g) as a solid.
Production example 142
2-Azido-1, 3-dimethylimidazole was cooled with an ice/sodium chloride aqueous bath under an argon atmosphereA solution of hexafluorophosphate (2.7 g) in MeCN (10 mL) was added in small amounts over 20 minutes to a mixture of L-valyl- (4R) -N- { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } -4-hydroxy-L-prolinamide N-hydrochloride (4.27 g), DIPEA (8.54 mL), THF (55 mL) and MeCN (55 mL) at a time (internal temperature below 3 ℃). The reaction mixture was stirred for 2 hours with cooling with an ice/sodium chloride aqueous solution bath. Water, saturated aqueous sodium chloride solution and ethyl acetate were added, and the aqueous layer and the organic layer were separated. The aqueous layer was extracted with ethyl acetate, and the combined organic layers were dried over sodium sulfate. After filtering off the drying agent, it was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (CHCl 3/MeOH) to give (4R) -1- [ (2S) -2-azido-3-methylbutanoyl ] -N- { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } -4-hydroxy-L-prolinamide (2.81 g) as a solid.
Production example 143
Under argon atmosphere, to (3S) -3- ({ (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] aTo a mixture of tert-butyl (220 mg) of alk-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } amino) pyrrolidine-1-carboxylate and DMF (2 mL) was added sodium hydride (55% mineral oil dispersion, 26 mg) with cooling with ice/MeOH bath, and stirred in this state for 30 minutes. Subsequently, ethyl iodide (0.08 mL) was added thereto, followed by stirring at room temperature for 1.5 hours. Ice water and saturated aqueous ammonium chloride solution were added, and extraction was performed 2 times with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. The drying agent was filtered off, and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (3S) -3- [ { (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as a solidAlkan-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (ethyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (137 mg).
Production example 144
To a mixture of tert-butyl N- [ (1R) -1- (4-bromophenyl) -2-hydroxyethyl ] carbamate (4.43 g), 1-ethyl-5- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -1H-pyrazole (4.67 g), potassium carbonate (3.87 g), DOX (80 mL) and water (8 mL) was added PdCl 2(dppf)·CH2Cl2 (1.14 g) under an argon atmosphere, and the mixture was stirred at 100℃for 16 hours. After naturally cooling to room temperature, ethyl acetate was added thereto, followed by filtration through celite (registered trademark), and concentration under reduced pressure was performed. The residue was purified by silica gel column chromatography (basic silica gel, hexane/ethyl acetate) to give tert-butyl { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } carbamate (3.74 g) as a solid.
Production example 146
To a solution of trimethyltin (IV) hydroxide (575 mg) in 1, 2-dichloroethane (10 mL) under a nitrogen atmosphere was added (4R) -1- [ (2S) -2- (4- {4- [ ({ (7M) -4- { [ (3S) -1- (tert-butoxycarbonyl) pyrrolidin-3-yl ] (methyl) amino } -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ] at room temperatureAlkan-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-8-yl } oxy) methyl ] phenyl } -1H-1,2, 3-triazol-1-yl) -3-methylbutanoyl ] -4-hydroxy-L-prolinemethyl ester (695 mg) was stirred at 80℃for 65 hours. After cooling to room temperature, ice-cold 1M hydrochloric acid was added and extracted 2 times with CHCl 3/MeOH (9/1). The combined organic layers were washed with 1M hydrochloric acid, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (CHCl 3/MeOH) to give (4R) -1- [ (2S) -2- (4- {4- [ ({ (7M) -4- { [ (3S) -1- (tert-butoxycarbonyl) pyrrolidin-3-yl ] (methyl) amino } -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ] as a solidAlk-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-8-yl } oxy) methyl ] phenyl } -1H-1,2, 3-triazol-1-yl) -3-methylbutanoyl ] -4-hydroxy-L-proline (663 mg).
Production example 149
To (2R) -2-amino-2- (4-fluorophenyl) ethan-1-ol (18 mg), (4R) -1- [ (2S) -2- (4- {4- [ ({ (7M) -4- { [ (3S) -1- (tert-butoxycarbonyl) pyrrolidin-3-yl ] (methyl) amino } -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ]To a mixture of alk-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] quinazolin-8-yl } oxy) methyl ] phenyl } -1H-1,2, 3-triazol-1-yl) -3-methylbutanoyl ] -4-hydroxy-L-proline (90 mg) and DMF (0.25 mL) was added DIPEA (80. Mu.L) and HATU (40 mg) under ice-cooling. The mixture was warmed to room temperature and stirred overnight. Water (5 mL) and saturated aqueous sodium bicarbonate (1 mL) were added and stirred for 1 hour. The resulting solid was collected by filtration to give (3S) -3- [ { (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ] as a colorless solidAlkan-2-yl) -1H-indazol-4-yl ] -8- { [4- (1- { (2S) -1- [ (2S, 4R) -2- { [ (1R) -1- (4-fluorophenyl) -2-hydroxyethyl ] carbamoyl } -4-hydroxypyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (87.5 mg).
Production example 163
To (7M) -4-tert-butoxy-6-cyclopropyl-7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinazoline (2.89 g) was added THF (40 mL), 3, 4-dinitro-2H-pyran (3.1 mL) and 4-methylbenzene-1-sulfonic acid monohydrate (330 mg), and after stirring at 50℃for 6 hours. After the reaction mixture was naturally cooled to room temperature, it was concentrated under reduced pressure, a saturated aqueous sodium hydrogencarbonate solution was added thereto, and extraction was performed 2 times with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate, filtered, concentrated, and the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ] as a foam solidAlk-2-yl) -1H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinazolin-4-ol (1.63 g).
Production example 171
To a mixture of tert-butyl (3S) -3- (methylamino) pyrrolidine-1-carboxylate (1.52 g), THF (15 mL), DOX (30 mL) and DIPEA (5 mL) under an argon atmosphere was added 7-bromo-2, 4-dichloro-8-fluoro-6-iodoquinazoline (3 g) with stirring under cooling with an ice/MeOH bath, and stirring at this temperature for 1.5 hours. A saturated aqueous ammonium chloride solution and ice water were poured into the reaction vessel, stirred in this state for 1.5 hours, and then insoluble matter was collected by filtration while washing with water, and dried under reduced pressure at 40℃for 5 hours to obtain tert-butyl (3S) -3- [ (7-bromo-2-chloro-8-fluoro-6-iodoquinazolin-4-yl) (methyl) amino ] pyrrolidine-1-carboxylate (3.82 g) as a solid.
Production example 172
To a mixture of tert-butyl (3S) -3- [ { 6-cyclopropyl-2- (ethylsulfanyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylate (a mixture of diastereomers from the axis asymmetry of about 2.5:1, 25.5 g) and CH 2Cl2 (350 mL) was added m-chloroperbenzoic acid (containing about 30%, 14.5 g) with stirring under ice cooling, and stirred under argon atmosphere at room temperature for 2.5 hours. The reaction mixture was poured with ice water/saturated aqueous sodium thiosulfate solution/saturated aqueous sodium bicarbonate solution, stirred at room temperature for about 20 minutes, and then extracted with CHCl 3. The organic layer was dried over anhydrous magnesium sulfate, and the solvent was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (3S) -3- [ { 6-cyclopropyl-2- (ethanesulfonyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinazolin-4-yl } (meth) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (from a mixture of diastereomers of about 4.3:1, which was axiasymmetric, 5.37 g) as a solid, and (3S) -3- [ { 6-cyclopropyl-2- (ethanesulfonyl) -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -8- [ (1S) -1-phenylethoxy ] quinazolin-4-yl } (meth) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (from a mixture of about 2:1, which was axiasymmetric, 8.25 g) as a solid.
The resulting mixture of tert-butyl (3S) -3- [ { 6-cyclopropyl-2- (ethanesulfonyl) -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -8- [ (1S) -1-phenylethoxy ] quinazolin-4-yl } (meth) amino ] pyrrolidine-1-carboxylate (from a mixture of diastereomers of about 2:1 that is axi-asymmetric, 7.9 g) and EtOH (80 mL) was stirred at room temperature for 3 days. Insoluble material was filtered off while rinsing with a small amount of EtOH, and dried under reduced pressure at 40 ℃ to give (3S) -3- [ { (7M) -6-cyclopropyl-2- (ethanesulfonyl) -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -8- [ (1S) -1-phenylethoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (single diastereomer, 4.42 g) as a solid.
For the (3S) -3- [ { 6-cyclopropyl-2- (ethanesulfonyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (from the diastereomeric mixture of about 4.3:1, which was axiasymmetric), obtained in the previous reaction was similarly converted to (3S) -3- [ { (7M) -6-cyclopropyl-2- (ethanesulfonyl) -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -8- [ (1S) -1-phenylethoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (single diastereomer) by the following procedure.
To a mixture of tert-butyl (3S) -3- [ { 6-cyclopropyl-2- (ethanesulfonyl) -7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -8- [ (1S) -1-phenylethoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylate (from a mixture of diastereomers of about 4.3:1, which was axiasymmetric, 6.33 g) and THF (70 mL) was added 4-methylbenzene-1-sulfonic acid monohydrate (0.13 g), and stirred overnight under an argon atmosphere. 4-methylbenzene-1-sulfonic acid monohydrate (0.15 g) was added thereto, and the mixture was stirred at room temperature for 1 day. The reaction solvent was concentrated under reduced pressure and the residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (3S) -3- [ { 6-cyclopropyl-2- (ethanesulfonyl) -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -8- [ (1S) -1-phenylethoxy ] quinazolin-4-yl } (meth) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (from about 5:1 diastereomer mixture of axial asymmetry, 3.21 g) as a solid. The resulting unprotected body (about 5:1 mixture of diastereomers from axial asymmetry, 3.2 g) was dissolved in EtOH (30 mL) and the mixture was stirred at room temperature for 1 day. The resulting insoluble material was filtered off while rinsing with a small amount of EtOH and dried overnight at 40 ℃ under reduced pressure to give (3S) -3- [ { (7M) -6-cyclopropyl-2- (ethanesulfonyl) -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -8- [ (1S) -1-phenylethoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (single diastereomer, 2.23 g) as a solid.
Production example 173
To a mixture of 4-bromo-6-fluoro-1H-indazole (235 g), TEA (183 mL), CH 2Cl2 (1880 mL) was added 1,1' - (chloromethane triyl) triphenyl (335 g) at room temperature, and the mixture was stirred at 25℃for 16 hours. The reaction mixture was poured into ice water (1.5L), the organic layer and the aqueous layer were separated, and the aqueous layer was extracted 3 times with CH 2Cl2 (400 mL). The combined organic layers were dried over anhydrous sodium sulfate, insoluble matter was filtered off, and the filtrate was concentrated under reduced pressure. Petroleum ether (550 mL) was added to the resulting residue and powdered (0 ℃ C., 2 hours), then filtered off, and dried under reduced pressure to give 4-bromo-6-fluoro-2- (triphenylmethyl) -2H-indazole (508.98 g) as a solid.
Production example 174
To a mixture of 4-bromo-6-fluoro-2- (triphenylmethyl) -2H-indazole (100 g) in 2-methyltetrahydrofuran (1000 mL) was added lithium diisopropylamide (2M THF solution, 214.28 mL) at-78deg.C under nitrogen atmosphere, and the mixture was stirred at-78deg.C for 2.5 hours. Methyl iodide (26.68 mL) was added at-78deg.C and stirred at 25deg.C for 2.5 hours. The reaction was stopped by adding water (2000 mL) and extracted 2 times with ethyl acetate (800 mL). The combined organic layers were dried over anhydrous sodium sulfate, insoluble matter was filtered off, and the filtrate was concentrated under reduced pressure. To the resulting residue was added ethyl acetate (50 mL)/petroleum ether (50 mL) and powdered, and then filtered off, and dried under reduced pressure to give 4-bromo-6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazole (81 g) as a solid.
Production example 175
To 4-bromo-6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazole (100 g), 4', 5', to a mixture of 5 '-octamethyl-2, 2' -bis-1, 3, 2-dioxaborane (61.42 g), triphenylphosphine (10.57 g), potassium acetate (59.34 g) and DOX (1000 mL) was added palladium acetate (4.52 g) at room temperature under a nitrogen atmosphere. After degassing and nitrogen filling of the reaction mixture were each performed 3 times, stirring was performed at 100℃for 12 hours under a nitrogen atmosphere. After cooling, water (1500 mL) was added and extracted 3 times with ethyl acetate (900 mL). The combined organic layers were dried over anhydrous sodium sulfate, and insoluble matter was filtered off. To the resulting solution was added activated carbon (50 g), and the mixture was stirred at 20℃for 1 hour and filtered while being washed 3 times with ethyl acetate (50 ml). The filtrate was concentrated, meOH (200 mL) was added to the resulting residue and powdered, filtered off, and dried under reduced pressure to give 6-fluoro-5-methyl-4- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2- (triphenylmethyl) -2H-indazole (110 g) as a solid.
Production example 176
DIPEA (200. Mu.L) was added to a solution of (4-ethynyl-3-fluorophenyl) methanol (110 mg) in CH 2Cl2 (2 mL) under ice cooling under nitrogen atmosphere, followed by addition of methanesulfonyl chloride (80. Mu.L). After stirring the reaction mixture at room temperature overnight, water was added and extracted with CHCl 3. The organic layer was washed with water and saturated aqueous sodium chloride solution, and dried over anhydrous magnesium sulfate. Insoluble materials were filtered off, and concentrated under reduced pressure to give 4- (chloromethyl) -1-ethynyl-2-fluorobenzene (122 mg) as an oil.
Production example 177
To a mixture of 2-amino-4-bromo-3-fluoro-5-iodobenzoic acid (10 g) and N-methyl-2-pyrrolidone (20 mL) was added trimethyl orthoacetate (10.5 mL), and the mixture was stirred at 110℃for 12 hours. After cooling to room temperature, meOH (20 ml) was added, and the resulting solid was collected by filtration while being washed with cooled methanol (about 60 ml), and dried under reduced pressure to give methyl 2-acetamide-4-bromo-3-fluoro-5-iodobenzoate (6.67 g) as a solid.
Production example 178
To a mixture of lithium bis (trimethylsilyl) amide (1.3M in THF, 100 mL) and THF (150 mL) under an argon atmosphere was added a small amount of methyl 2-acetamide-4-bromo-3-fluoro-5-iodobenzoate (18 g) at a time with cooling in a water bath. After stirring at 40℃for 1 hour, it was cooled to room temperature, water was added, and the mixture was washed with ethyl acetate 2 times. Hydrochloric acid (1M, 140 ml) and ice water were added to the aqueous layer under ice cooling to make it acidic, and stirred for a while. The precipitated solid was collected by filtration and dried under reduced pressure. The resulting solid was collected by filtration while being washed with methanol, and dried under reduced pressure to give 7-bromo-8-fluoro-4-hydroxy-6-iodoquinolin-2 (1H) -one (13.6 g) as a solid.
Production example 179
To 7-bromo-8-fluoro-4-hydroxy-6-iodoquinolin-2 (1H) -one (5 g) was added phosphorus oxychloride (24 mL) under an argon atmosphere, DIPEA (7 mL) was slowly added under ice cooling, and the mixture was stirred at 110℃for 1 hour. The reaction solution was cooled to room temperature, and concentrated under reduced pressure. Ice water was added to the residue and stirred for 30 minutes. The precipitated solid was collected by filtration, washed with water, and dried under reduced pressure to give 7-bromo-2, 4-dichloro-8-fluoro-6-iodoquinoline (5.03 g).
Production example 180
DABCO (1.8 g) was added to a mixture of 7-bromo-2, 4-dichloro-8-fluoro-6-iodoquinoline (6.4 g) and DMAc (70 mL) under argon atmosphere, and stirred at 40℃for 2 hours. Ethanethiol (1.4 mL) was added and stirred at 60℃for 1 hour. Cooled to room temperature, water was added, stirred for 5 minutes, the resulting solid was filtered off and dried under reduced pressure. The obtained solid was purified by silica gel column chromatography (hexane/ethyl acetate) to obtain 7-bromo-4-chloro-2- (ethylsulfanyl) -8-fluoro-6-iodoquinoline (5.76 g) as a solid.
Production example 181
To a solution of (1S) -1-phenylethan-1-ol (1.56 mL) in dehydrated THF (40 mL) under argon atmosphere was added tBuOK (1.39 g), and the mixture was stirred at room temperature for 30 minutes, and the prepared mixture was added dropwise to a mixture of 7-bromo-4-chloro-2- (ethylsulfanyl) -8-fluoro-6-iodoquinoline (5.5 g) and dehydrated THF (40 mL) under ice cooling for 20 minutes, and stirred at that temperature for 10 minutes. To a solution of (1S) -1-phenylethan-1-ol (0.45 mL) in dehydrated THF (10 mL) was additionally added tBuOK (415 mg) under an argon atmosphere, and the mixture was stirred at room temperature for 10 minutes, and the prepared mixture was slowly added dropwise to the previous reaction mixture under ice-cooling, and stirred at that temperature for 10 minutes. Saturated aqueous ammonium chloride, ice and ethyl acetate were added and the solution was separated. The aqueous layer was extracted with ethyl acetate 2 times, and the combined organic layers were washed with saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. Insoluble matter was filtered off, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give 7-bromo-4-chloro-2- (ethylsulfanyl) -6-iodo-8- [ (1S) -1-phenylethoxy ] quinoline (4.9 g) as an oil. The fractions mixed with a small amount of impurities were concentrated to give 7-bromo-4-chloro-2- (ethylsulfanyl) -6-iodo-8- [ (1S) -1-phenylethoxy ] quinoline (1.56 g) as an oil.
Production example 182
To a solution of tert-butyl (3S) -3-hydroxypyrrolidine-1-carboxylate (2.7 g) in DMAc (20 mL) under argon atmosphere was added tBuOK (1.59 g), and the mixture was stirred at room temperature for 10 minutes, and the prepared mixture was added dropwise to a mixture of 7-bromo-4-chloro-2- (ethylsulfanyl) -6-iodo-8- [ (1S) -1-phenylethoxy ] quinoline (6.46 g) and DMAc (40 mL) under ice-cooling, and stirred at that temperature for 30 minutes. Saturated aqueous ammonium chloride, ice and ethyl acetate were added, stirred, and the mixture was separated. The aqueous layer was extracted with ethyl acetate 2 times, and the combined organic layers were washed with saturated aqueous sodium chloride solution, dried over anhydrous magnesium sulfate, and concentrated under reduced pressure. Purification by silica gel column chromatography (hexane/ethyl acetate) gave (3S) -3- ({ 7-bromo-2- (ethylsulfanyl) -6-iodo-8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } oxy) pyrrolidine-1-carboxylic acid tert-butyl ester (4.6 g) as a solid.
Production example 183
To a mixture of tert-butyl N- [ (1R) -1- (4-bromophenyl) -2-hydroxyethyl ] carbamate (1 g), 2-dimethoxypropane (3.3 mL) and acetone (15 mL) was added boron trifluoride diethyl ether complex (26. Mu.L), and the mixture was stirred at room temperature for 1 hour. TEA (66. Mu.L) was added and stirred at room temperature for 10 minutes. Concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (4R) -4- (4-bromophenyl) -2, 2-dimethyl-1, 3-propanediol in solid formOxazolidine-3-carboxylic acid tert-butyl ester (1.09 g).
Production example 184
To (4R) -4- (4-bromophenyl) -2, 2-dimethyl-1, 3-)Oxazolidine-3-carboxylic acid tert-butyl ester (300 mg), 1,3-Copper (I) iodide (32 mg), rac- (1R, 2R) -cyclohexane-1, 2-diamine (20. Mu.L), and potassium carbonate (290 mg) were added to a solution of oxazolidin-2-one (183 mg) in DOX (1.69 mL) at room temperature. Stirring was carried out at 140℃for 2 hours under microwave irradiation and at 150℃for 1 hour. Ethyl acetate and water were added thereto, and filtration was performed through celite (registered trademark). Concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (4R) -2, 2-dimethyl-4- [4- (2-oxo-1, 3- ] in solid formOxazolidin-3-yl) phenyl ] -1,3-Oxazolidine-3-carboxylic acid tert-butyl ester (120 mg).
Production example 185
(4R) -4- (4-bromophenyl) -2, 2-dimethyl-1, 3-)Oxazolidine-3-carboxylic acid tert-butyl ester (858 mg), 2-methyl-1H-imidazole (500 mg), copper (I) iodide (95 mg), quinolin-8-ol (138 mg), potassium carbonate (670 mg) were suspended in DMSO (10 mL) and reacted under microwave irradiation under argon atmosphere at 150℃for 3 hours. After cooling to room temperature, ethyl acetate and water were added, extraction was performed 2 times with ethyl acetate, and the combined organic layers were washed with a saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. Insoluble matter was filtered off, and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (basic silica gel, hexane/ethyl acetate) to give (4R) -2, 2-dimethyl-4- [4- (2-methyl-1H-imidazol-1-yl) phenyl ] -1, 3-propanediol as an oilOxazolidine-3-carboxylic acid tert-butyl ester (500 mg).
Production example 187
Bis (tri-t-butylphosphine) palladium (0) (18 mg) was added to 4-methyl-1, 3-Oxazole-5-carboxylic acid (178 mg), tetra-n-butylammonium chloride (195 mg), (4R) -4- (4-bromophenyl) -2, 2-dimethyl-1, 3-In a mixture of tert-butyl oxazolidine-3-carboxylate (250 mg), cesium carbonate (344 mg) and DMF (2.5 mL) was stirred at 170℃for 30 min under microwave irradiation. After cooling to room temperature, the mixture was diluted with ethyl acetate, insoluble matters were removed by filtration through celite (registered trademark), and the filtrate was washed with water and a saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. Insoluble matter was filtered off, and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (CHCl 3/MeOH) to give (4R) -2, 2-dimethyl-4- [4- (4-methyl-1, 3- ] in solid formOxazol-5-yl) phenyl ] -1, 3-)Oxazolidine-3-carboxylic acid tert-butyl ester (215 mg).
Production example 188
Addition of (3S) -3- { [ (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] anAlkyl-2-yl) -1H-indazol-4-yl ] -2- { [ (2S, 3S) -3-hydroxybutan-2-yl ] oxy } quinazolin-4-yl ] (methyl) amino } pyrrolidine-1-carboxylic acid tert-butyl ester (60 mg), dehydrated THF (2 mL) and methyl iodide (35 μl) were added sodium hydride (55% dispersion in mineral oil, 10 mg) under stirring under an argon atmosphere with cooling in an ice/MeOH bath, and stirred at room temperature overnight. An ice/saturated aqueous ammonium chloride solution was added to the reaction vessel, and extraction was performed 2 times with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. The solvent was distilled off under reduced pressure, and the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (3S) -3- { [ (7M) -6-cyclopropyl-8- [ (4-ethynylphenyl) methoxy ] -7- [ 6-fluoro-5-methyl-1- ] as a solidAlkan-2-yl) -1H-indazol-4-yl ] -2- { [ (2S, 3S) -3-methoxybutan-2-yl ] oxy } quinazolin-4-yl ] (methyl) amino } pyrrolidine-1-carboxylic acid tert-butyl ester (50 mg).
Production example 192
Cesium carbonate (2.3 g) was added to a mixture of tetrahydro-2H-pyran-4-ol (272. Mu.L), 7-bromo-2, 4-dichloro-8-fluoro-6-iodoquinoline (1 g), DABCO (280 mg) and DMAc (10 mL) under an argon atmosphere, stirred at 60℃for 6 hours and 80℃for 3 hours. After naturally cooling to room temperature, ethyl acetate and water were added, and the mixture was stirred at room temperature for 30 minutes. The precipitated solid was collected by filtration and dried under reduced pressure. Purifying the obtained solid by silica gel column chromatography (hexane/ethyl acetate) to obtain 7-bromo-4-chloro-8-fluoro-6-iodo-2- [ (] in solid formAlk-4-yl) oxy ] quinoline (692 mg).
Production example 203
To a mixture of 7-bromo-2, 4-dichloro-8-fluoro-6-iodoquinoline (10 g), DABCO (3.3 g), cesium carbonate (23.3 g) and DMAc (100 mL) was added (2S) -2-methoxypropane-1-ol (3 mL) under an argon atmosphere, and after stirring at room temperature for 1.5 hours, stirring at 60℃for 12 hours. Naturally cooling to room temperature, adding water and ethyl acetate, stirring for a period of time, separating the two layers, and extracting the water layer with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. Insoluble matter was filtered off, and concentrated. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give 7-bromo-4-chloro-8-fluoro-6-iodo-2- [ (2S) -2-methoxypropoxy ] quinoline (5.97 g) as a solid.
Production example 204
To a solution of 7-bromo-4-chloro-8-fluoro-6-iodo-2- [ (2S) -2-methoxypropoxy ] quinoline (6.47 g) and (1S) -1-phenylethan-1-ol (3.34 mL) in THF (50 mL) under an argon atmosphere was added tBuOK (3.06 g) in a cooling bath (about-10 ℃ C.) and stirred at this temperature for 30 minutes. Saturated aqueous ammonium chloride was added thereto under ice-cooling, and extraction was performed 2 times with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. Insoluble matter was filtered off, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give 7-bromo-4-chloro-6-iodo-2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinoline (4.67 g) as an oil.
Production example 205
To a solution of 7-bromo-4-chloro-6-iodo-2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinoline (1.6 g) in DMAc (5 mL) was added (1S, 4S) -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylic acid tert-butyl ester (832 mg) and potassium carbonate (768 mg) at room temperature, and the mixture was stirred overnight at 120 ℃. (1S, 4S) -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylic acid tert-butyl ester (278 mg) and potassium carbonate (384 mg) were added at 120℃and stirred at this temperature overnight. Cooled to room temperature, saturated aqueous ammonium chloride was added, and extraction was performed 2 times with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. Concentrated under reduced pressure and the residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (1S, 4S) -5- { 7-bromo-6-iodo-2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylic acid tert-butyl ester (1.02 g) as a foamy solid.
Production example 206
A mixture of 7-bromo-4-chloro-6-iodo-2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinoline (377 mg), (3S) -3-aminopyrrolidine-1-carboxylic acid tert-butyl ester (250 mg), potassium carbonate (130 mg), DMAc (5 mL) was stirred under argon atmosphere at 130℃for 7 days. Cooled to room temperature, water was added, and extraction was performed 2 times with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride solution, dried over magnesium sulfate, and concentrated under reduced pressure. Purification by silica gel column chromatography (hexane/ethyl acetate) gave (3S) -3- ({ 7-bromo-6-iodo-2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } amino) pyrrolidine-1-carboxylic acid tert-butyl ester (259 mg) as a foamy solid.
Production example 207
(3S) -3- ({ 7-bromo-6-iodo-2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } amino) pyrrolidine-1-carboxylic acid tert-butyl ester (285 mg) was dissolved in DMF (3 mL), sodium hydride (60% in oil dispersion, 19 mg) was added under ice cooling, and stirred at room temperature for 1 hour. Methyl iodide (28. Mu.L) was added at room temperature, and stirred at that temperature for 1 hour. Ice and saturated aqueous ammonium chloride solution were added, and extraction was performed 2 times with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride solution, dried over magnesium sulfate, and concentrated under reduced pressure. Purification by silica gel column chromatography (hexane/ethyl acetate) gave (3S) -3- [ { 7-bromo-6-iodo-2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylic acid tert-butyl ester (238 mg) as a foamy solid.
Production example 210
A mixture of (1S, 4S) -5- { 7-bromo-6-cyclopropyl-2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylic acid tert-butyl ester (0.75 g), 6-fluoro-5-methyl-4- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2- (triphenylmethyl) -2H-indazole (900 mg), dicyclohexyl (2 ',6' -diisopropyloxy- [1,1' -biphenyl ] -2-yl) phosphine (138 mg), palladium (II) acetate (31 mg), anhydrous barium hydroxide (0.61 g), DOX (30 mL) and water (6 mL) was stirred at 50℃for 30 minutes under an argon atmosphere. 6-fluoro-5-methyl-4- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) -2- (triphenylmethyl) -2H-indazole (600 mg) was added at this temperature and stirred at 50℃for a further 1 hour. Cooled at room temperature, ethyl acetate and water were added, insoluble matters were removed by filtration through celite (registered trademark), and two layers of the filtrate were separated. The aqueous layer was extracted with ethyl acetate, and the combined organic layers were washed with saturated aqueous sodium chloride solution and dried over anhydrous magnesium sulfate. After filtration of the insoluble material, concentration under reduced pressure, the residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (1S, 4S) -5- { 6-cyclopropyl-7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylic acid tert-butyl ester (from about 4:1 diastereomeric mixture of axi, 866 mg) as a solid. To the resulting solid (866 mg) was added MeOH (5 mL), and the mixture was heated to 60 ℃ to dissolve the MeOH, and stirred at room temperature under an argon atmosphere for 3 days. The precipitated solid was collected by filtration while being washed with a small amount of MeOH to give (1S, 4S) -5- { 6-cyclopropyl-7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -2- [ (2S) -2-methoxypropoxy ] -8- [ (1S) -1-phenylethoxy ] quinolin-4-yl } -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylic acid tert-butyl ester (almost single diastereomer of unknown configuration, 200 mg) as a solid.
Production example 218
To a mixture of methyl 3-cyclopropyl-3-oxopropionate (17 mL) and trimethyl orthoformate (20 mL) was added concentrated sulfuric acid (500. Mu.L) under ice-cooling, and the mixture was stirred at room temperature under argon atmosphere overnight. The reaction solution was concentrated under reduced pressure at room temperature and dried for 4 hours. To the residue, 5-aminopyrimidine-2, 4 (1H, 3H) -dione (11.4 g) and DMAc (300 mL) were added at room temperature and stirred overnight at 110℃under an argon atmosphere. The reaction solution was concentrated under reduced pressure, water was added to the residue, and the resulting solid was collected by filtration, and dried overnight at 50℃under reduced pressure to give methyl 3-cyclopropyl-3- [ (2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) amino ] prop-2-enoate (22.3 g) as a solid.
Production example 219
Methyl 3-cyclopropyl-3- [ (2, 4-dioxo-1, 2,3, 4-tetrahydropyrimidin-5-yl) amino ] prop-2-enoate (870 mg) was dissolved in NMP (17 mL) at room temperature and stirred under argon atmosphere at 220℃for 5 hours. The reaction solution was concentrated under reduced pressure, water was added to the residue, and the resultant solid was collected by filtration, and dried at 50℃under reduced pressure for 1 hour to give 6-cyclopropylpyrido [3,2-d ] pyrimidine-2, 4, 8-triol (230 mg) as a solid.
Production example 220
To a solution of 6-cyclopropylpyrido [3,2-d ] pyrimidine-2, 4, 8-triol (5.71 g) in DMF (90 mL) was added N-bromosuccinimide (9.3 g) under ice-cooling, and the mixture was stirred at room temperature overnight. Water was added under ice-cooling, and the resulting solid was collected by filtration and dried at 50℃under reduced pressure for 3 hours to give 7-bromo-6-cyclopropylpyrido [3,2-d ] pyrimidine-2, 4, 8-triol (2.12 g) as a solid.
Production example 221
To a mixture of 7-bromo-6-cyclopropylpyrido [3,2-d ] pyrimidine-2, 4, 8-triol (2.11 g) and N, N-diethylaniline (3.4 mL) was added phosphorus oxychloride (38.2 g) at room temperature and stirred overnight at 100℃under an argon atmosphere. The reaction solution was concentrated under reduced pressure, meCN (35 mL) and DIPEA (6 mL) were added to the residue under ice-cooling, and after stirring for 1 minute, tert-butyl (1 s,4 s) -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylate (1.96 g) was added under ice-cooling, and stirring was continued at room temperature for 1 hour. The reaction solution was concentrated under reduced pressure, and the obtained residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (1S, 4S) -5- (7-bromo-2, 8-dichloro-6-cyclopropylpyrido [3,2-d ] pyrimidin-4-yl) -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylic acid tert-butyl ester (1.29 g) as a solid.
Manufacturing example 222
(1S, 4S) -5- (7-bromo-2, 8-dichloro-6-cyclopropylpyrido [3,2-d ] pyrimidin-4-yl) -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylic acid tert-butyl ester (2.4 g), DMF (20 mL), THF (20 mL) and tetrahydro-2H-pyran-4-ol (2.2 mL) were mixed at room temperature, and cesium carbonate (7.6 g) and DABCO (100 mg) were added under ice cooling. After stirring overnight at 50℃under an argon atmosphere, a saturated aqueous ammonium chloride solution was added under ice-cooling to stop the reaction. Water and ethyl acetate were added, the organic layer and the aqueous layer were separated, and the aqueous layer was extracted 3 times with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give (1S, 4S) -5- { 7-bromo-8-chloro-6-cyclopropyl-2- [ (-) -Alkan-4-yl) oxy ] pyrido [3,2-d ] pyrimidin-4-yl } -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylic acid tert-butyl ester (1.23 g).
Production example 225
To (1S, 4S) -5- { 6-cyclopropyl-7- [ 6-fluoro-5-methyl-2- (triphenylmethyl) -2H-indazol-4-yl ] -2- {To a solution of tert-butyl alk-4-yl) oxy ] -8- [ (1S) -1-phenylethoxy ] pyrido [3,2-d ] pyrimidin-4-yl } -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylate (470 mg) in THF (10 mL) was added 4-methylbenzene-1-sulfonic acid monohydrate (60 mg) and 3, 4-dinitro-2H-pyran (1.1 mL) under ice-cooling, and the mixture was stirred overnight at room temperature under an argon atmosphere. After TEA (0.4 mL) was added under ice-cooling, the reaction solution was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (basic silica gel, hexane/ethyl acetate) to give (1S, 4S) -5- { 6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ] as a solidAlkan-2-yl) -1H-indazol-4-yl ] -2- [ ((x-x) -2- [ (x-y) and x-y)Alkan-4-yl) oxy ] -8- [ (1S) -1-phenylethoxy ] pyrido [3,2-d ] pyrimidin-4-yl } -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylic acid tert-butyl ester (291 mg).
The compounds shown in the following tables were produced in the same manner as in the production method of the production example shown above. The production methods, structures, and physicochemical data of the compounds of each production example are shown in the following tables.
Example 2
To 3- ({ (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ]Alkan-2-yl) -1H-indazol-4-yl ] -8- { [4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2- [. Sup.To a mixture of tert-butyl (355 mg) of alk-4-yloxy ] quinazolin-4-yl } oxy) azetidine-1-carboxylate and CH 2Cl2 (10 mL) was added phosphoric acid (85%, 500. Mu.L), and the mixture was stirred at room temperature overnight. After adding saturated aqueous sodium bicarbonate to the mixture, CHCl 3/MeOH (5/1) and water were added and stirred at room temperature for 30 minutes. Extraction with CHCl 3/MeOH (5/1), drying the combined organic layers over anhydrous sodium sulfate, filtration, concentration under reduced pressure, and recovery of (4R) -1- [ (2S) -2- (4- {4- [ ({ (7M) -4- [ (azetidin-3-yl) oxy ] -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (-a-M-ethyl) was achieved as a solidAlkan-4-yl) oxy ] quinazolin-8-yl } oxy) methyl ] phenyl } -1H-1,2, 3-triazol-1-yl) -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (355 mg).
Example 9
Under nitrogen atmosphere, to (3S) -3- { [ (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ]Alkan-2-yl) -1H-indazol-4-yl ] -8- { [4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2 (2R) -oxopyrrolidin-2-yl ] methoxy } quinazolin-4-yl ] (methyl) amino } pyrrolidine-1-carboxylic acid tert-butyl ester (156 mg) in CH 2Cl2 (2 mL) was added with trifluoroacetic acid (0.5 mL) under ice-cooling and stirred at room temperature for 2 hours. The reaction mixture was concentrated under reduced pressure, meCN, saturated aqueous sodium bicarbonate and water were added to the residue, and stirred at room temperature for 20 minutes. The reaction solution was purified by ODS column chromatography (MeCN/0.1% aqueous formic acid), and the fraction containing the objective was concentrated. The residue was dissolved in CHCl 3/iPrOH (9/1), saturated aqueous sodium bicarbonate was added and extracted 2 times with CHCl 3/MeOH (5/1). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to give (4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } -2- { [ (2R) -oxolan-2-yl ] methoxy } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (47 mg) as a foamy solid.
Example 10
Under nitrogen atmosphere, to (3S) -3- { [ (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ]Alkan-2-yl) -1H-indazol-4-yl ] -8- { [4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2 (2S) -oxopyrrolidin-2-yl ] methoxy } quinazolin-4-yl ] (methyl) amino } pyrrolidine-1-carboxylic acid tert-butyl ester (236 mg) in CH 2Cl2 (2 mL) was added with trifluoroacetic acid (0.5 mL) under ice-cooling and stirred at room temperature for 2 hours. The reaction mixture was concentrated under reduced pressure, meCN, saturated aqueous sodium bicarbonate and water were added to the residue, and stirred at room temperature for 20 minutes. The reaction solution was purified by ODS column chromatography (MeCN/0.1% aqueous formic acid), and the fraction containing the objective was concentrated. The residue was dissolved in CHCl 3/iPrOH (9/1), saturated aqueous sodium bicarbonate was added and extracted 2 times with CHCl 3/MeOH (5/1). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to give (4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } -2- { [ (2S) -oxolan-2-yl ] methoxy } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide as a foamy solid (103 mg).
Example 11
Under nitrogen atmosphere, to (3S) -3- [ { (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ]To a solution of tert-butyl 8- { [4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylate (197 mg) in CH 2Cl2 (2 mL) was added trifluoroacetic acid (0.5 mL) under ice cooling and stirred at room temperature for 2 hours. The reaction mixture was concentrated under reduced pressure, meCN, saturated aqueous sodium bicarbonate and water were added to the residue, and stirred at room temperature for 20 minutes. The reaction solution was purified by ODS column chromatography (MeCN/0.1% aqueous formic acid), and the fraction containing the objective was concentrated. The residue was dissolved in CHCl 3/iPrOH (9/1), saturated aqueous sodium bicarbonate was added and extracted 2 times with CHCl 3/MeOH (5/1). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to give (4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (65 mg) as a foamy solid.
Example 13
To (3S) -3- [ { (7M) -6-cyclopropyl-8- { [4- (1- { (2S) -1- [ (2S, 4R) -2- ({ (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } carbamoyl) -4-hydroxypyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -7- [ 6-fluoro-5-methyl-1- (-d-a-To a solution of tert-butyl (2S) -2-methoxypropoxy ] quinazolin-4-yl (methyl) amino ] pyrrolidine-1-carboxylate (224 mg) in CH 2Cl2 (1 mL) was added TFA (1 mL) at room temperature. Stir at room temperature for 2 hours. The reaction solvent was concentrated under reduced pressure, meCN, saturated aqueous sodium bicarbonate and water were added to the residue, and stirred at room temperature for 20 minutes.
The resulting suspension was purified by ODS column chromatography (MeCN/0.1% aqueous formic acid), and the target fraction was concentrated under reduced pressure. The residue was dissolved in CHCl 3/iPrOH (9/1), saturated aqueous sodium bicarbonate was added and extracted 2 times with CHCl 3/MeOH (5/1). The combined organic layers were dried over anhydrous sodium sulfate, concentrated under reduced pressure, and concentrated as a foamy solid to give (4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -N- { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } -4-hydroxy-L-prolinamide (109 mg).
Example 44
To (3S) -3- [ { (7M) -6-cyclopropyl-8- { [ 3-fluoro-4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -7- [ 6-fluoro-5-methyl-1- ] under a nitrogen atmosphereTo a solution of tert-butyl (2S) -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (meth) amino ] pyrrolidine-1-carboxylate (169 mg) in CH 2Cl2 (2 mL), trifluoroacetic acid (1 mL) was added under ice cooling, and the mixture was stirred at room temperature for 2 hours. To the reaction mixture was added saturated aqueous sodium bicarbonate solution and extracted 2 times with CHCl 3/MeOH (9/1). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure. The resulting residue was purified by ODS column chromatography (MeCN/0.1% aqueous formic acid). To the fractions containing the target were added saturated aqueous sodium bicarbonate and extracted 2 times with CHCl 3/MeOH (9/1). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to give (4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) -2-fluorophenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (92 mg) as a solid.
Example 50
To (3S) -3- [ { (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ]To a mixture of tert-butyl 8- { [4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (ethyl) amino ] pyrrolidine-1-carboxylate (175 mg) and CH 2Cl2 (3 mL) was added trifluoroacetic acid (1 mL) under ice/MeOH bath cooling and stirred at room temperature for 2 hours under argon atmosphere. The resulting reaction mixture was concentrated under reduced pressure, ice water, saturated aqueous sodium bicarbonate and CHCl 3/MeOH (10/1) were added to the residue, and the mixture was stirred for 20 minutes and then separated. The aqueous layer was again extracted with CHCl 3/MeOH (10/1) and the combined organic layers were dried over anhydrous magnesium sulfate. The solution was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (basic silica gel, CHCl 3/MeOH) to give (4R) -1- [ (2S) -2- (4- {4- [ ({ (7M) -6-cyclopropyl-4- { ethyl [ (3S) -pyrrolidin-3-yl ] amino } -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] quinazolin-8-yl } oxy) methyl ] phenyl } -1H-1,2, 3-triazol-1-yl) -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (101 mg) as a solid.
Example 53
Under nitrogen atmosphere, to (3R) -3- [ { (7M) -6-cyclopropyl-7- [ 6-fluoro-5-methyl-1- ]To a solution of tert-butyl 8- { [4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2- [ (2S) -2-methoxypropoxy ] quinazolin-4-yl } (methyl) amino ] pyrrolidine-1-carboxylate (310 mg) in CH 2Cl2 (3 mL) was added trifluoroacetic acid (1.5 mL) under ice cooling, followed by stirring at room temperature for 2 hours. To the mixture was added saturated aqueous sodium bicarbonate and extracted 2 times with CHCl 3/MeOH (9/1). The combined organic layers were dried over anhydrous sodium sulfate, and the solution was concentrated under reduced pressure. The resulting residue was purified by ODS column chromatography (MeCN/0.1% aqueous formic acid). Fractions containing the target were combined, saturated aqueous sodium bicarbonate was added and extracted 2 times with CHCl 3/MeOH (9/1). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure. After dissolving the residue in EtOH, it was concentrated under reduced pressure to give (4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3R) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (117 mg) as a solid.
Example 57
To (1S, 4S) -5- { 6-cyclopropyl-7- [ 6-fluoro-5-methyl-1-Alkan-2-yl) -1H-indazol-4-yl ] -8- { [4- (1- { (2S) -1- [ (2S, 4R) -4-hydroxy-2- ({ (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } carbamoyl) pyrrolidin-1-yl ] -3-methyl-1-oxobutan-2-yl } -1H-1,2, 3-triazol-4-yl) phenyl ] methoxy } -2- [. Sup.To a solution of tert-butyl (axi-asymmetric mixture, 120 mg) of Alkyloxy ] pyrido [3,2-d ] pyrimidin-4-yl } -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylate (5 mL) was added methanesulfonic acid (0.3 mL) under ice-cooling, and the mixture was stirred at 50℃for 3 hours. After TEA (2 mL) was added under ice-cooling, the reaction solution was concentrated under reduced pressure. The residue was purified by ODS column chromatography (0.1% aqueous formic acid/0.1% MeCN) and fractions on the low polarity side of the two peaks having the molecular weight of the target were collected, and CHCl3/iPrOH (3/1) and saturated aqueous sodium hydrogencarbonate solution were added at room temperature. The organic and aqueous layers were separated and the aqueous layer was extracted 3 times with CHCl3/iPrOH (3/1). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was dissolved in a small amount of ethyl acetate/iPrOH (10/1), and hexane was added. The precipitated solid was collected by filtration, washed with water and dried overnight at 50℃under reduced pressure to give (4R) -1- [ (2S) -2- (4- {4- [ ({ 6-cyclopropyl-4- [ (1S, 4S) -2, 5-diazabicyclo [2.2.1] heptan-2-yl ] -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ ] as a solidAlkan-4-yl) oxy ] pyrido [3,2-d ] pyrimidin-8-yl } oxy) methyl ] phenyl } -1H-1,2, 3-triazol-1-yl) -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide (single axis asymmetry, 40.4 mg).
The example compounds shown in the following table were produced in the same manner as the production method of the example shown above. The production methods and physicochemical data of the compounds of the examples are shown in the following tables.
In the tables described below, the following abbreviations may be used.
PEx: manufacturing example No. Ex: example No. PSyn: manufacturing example number, syn manufactured by the same method: example numbers produced by the same method (e.g., syn:9 indicates that produced by the same method as in example 9), str: the chemical formula (a compound with a "×" in the chemical formula indicates that the axial or central asymmetry of the compound is unique). n HCl: n hydrochloride (the compound with the manufacturing example number represents monohydrochloride to dihydrochloride), DAT: physicochemical data, esi+: m/z values in mass spectrometry (ionization method ESI, when not stated, [ M+H ] +), ESI-: the M/z values in the mass spectrometry (ionization ESI, when not stated, are the delta value (ppm) of the peak in 1 H-NMR (500 MHz) at 27℃in [ M-H ] -)、NMR:DMSO-d6 ], the delta value (ppm) of the peak in 1 H-NMR (500 MHz) at 100℃in DMSO-d 6, s: singlet (spectrum), d: doublet (spectrum), dd: doublet (spectrum), ddd: doublet (spectrum), t: triplet (spectrum), q: quartet (spectrum), M: multiplet (spectrum), br: broad (spectrum) (e.g., br s).
[ Table 5-1]
[ Table 5-2]
[ Tables 5-3]
[ Tables 5-4]
[ Tables 5-5]
[ Tables 5-6]
[ Tables 5-7]
[ Tables 5-8]
[ Tables 5 to 9]
[ Tables 5 to 10]
[ Tables 5-11]
[ Tables 5-12]
[ Tables 5-13]
[ Tables 5 to 14]
[ Tables 5 to 15]
[ Tables 5 to 16]
[ Tables 5-17]
[ Tables 5-18]
[ Tables 5 to 19]
[ Tables 5-20]
[ Tables 5-21]
[ Tables 5-22]
[ Tables 5-23]
[ Tables 5 to 24]
[ Tables 5-25]
[ Tables 5-26]
[ Tables 5-27]
[ Tables 5-28]
[ Tables 5-29]
[ Tables 5-30]
[ Tables 5-31]
[ Tables 5-32]
[ Tables 5-33]
[ Tables 5-34]
[ Tables 5-35]
[ Tables 5-36]
[ Tables 5-37]
[ Tables 5-38]
[ Tables 5-39]
[ Tables 5-40]
[ Tables 5-41]
[ Tables 5-42]
[ Tables 5-43]
[ Tables 5-44]
[ Tables 5-45]
[ Tables 5-46]
[ Tables 5-47]
[ Tables 5-48]
[ Tables 5-49]
[ Tables 5-50]
[ Tables 5-51]
[ Tables 5-52]
[ Tables 5-53]
[ Tables 5-54]
[ Tables 5-55]
[ Tables 5-56]
[ Tables 5-57]
[ Tables 5-58]
[ Tables 5-59]
[ Tables 5-60]
[ Tables 5-61]
[ Tables 5-62]
[ Tables 5-63]
[ Tables 5-64]
[ Tables 5-65]
[ Tables 5-66]
[ Tables 5-67]
[ Table 6-1]
[ Table 6-2]
[ Table 6-3]
PEx | PSyn | DAT |
34 | 29 | ESI+:820.7 |
35 | 29 | ESI+:877.6[M+Na]+ |
36 | 36 | ESI+:841.6 |
37 | 36 | ESI+:823.7 |
38 | 36 | ESI+:823.7 |
39 | 36 | ESI+∶807.7 |
40 | 36 | ESI+∶845.7 |
41 | 36 | ESI+∶841.7 |
42 | 36 | ESI+∶819.8 |
43 | 36 | ESI+:803.6 |
44 | 36 | ESI+:821.7 |
45 | 36 | ESI+:807.8 |
46 | 36 | ESI+:843.7 |
47 | 36 | ESI+:825.7 |
48 | 36 | ESI+:807.8 |
49 | 36 | ESI+:837.7 |
50 | 36 | ESI+:863.6 |
51 | 36 | ESI+:963.7 |
52 | 36 | ESI+:851.8 |
53 | 36 | ESI+:876.8 |
54 | 54 | ESI+:693.6 |
55 | 36 | ESI+:863.4 |
56 | 36 | ESI+:851.5 |
57 | 57 | ESI+:705.5 |
58 | 58 | ESI+:705.5 |
59 | 59 | ESI+:637.4 |
60 | 60 | ESI+∶649.5 |
61 | 61 | ESI+∶649.5 |
62 | 62 | ESI+:809.8 |
63 | 63 | ESI+∶819.8 |
64 | 62 | ESI+∶805.8 |
65 | 62 | ESI+∶806.6 |
66 | 62 | ESI+∶805.6 |
67 | 62 | ESI+∶831.7 |
68 | 62 | ESI+∶819.7 |
69 | 62 | ESI+∶844.8 |
[ Tables 6 to 4]
PEx | PSyn | DAT |
70 | 62 | ESI+∶818.7 |
71 | 62 | ESI+∶831.7 |
72 | 62 | ESI+:819.7 |
73 | 73 | ESI+:819.7 |
74 | 74 | ESI+:831.8 |
75 | 75 | ESI+:831.8 |
76 | 62 | ESI+:805.5 |
77 | 62 | ESI+:833.4 |
78 | 62 | ESI+:831.7 |
79 | 79 | ESI+:805.5 |
80 | 62 | ESI+:806.3 |
81 | 62 | ESI+:804.4 |
82 | 62 | ESI+:733.4 |
83 | 83 | ESI+:1313.8[M+Ma]+ |
84 | 84 | ESI+:1305.6 |
85 | 83 | ESI+:1277.8 |
86 | 83 | ESI+:1279.7 |
87 | 83 | ESI+:1317.7 |
88 | 83 | ESI-:1307.7 |
89 | 83 | ESI+:1279.8 |
90 | 83 | ESI+:1310.2 |
91 | 83 | ESI+:1314.7[M+Ma]+ |
92 | 92 | ESI+:1332.5[M+Ma]+ |
93 | 83 | ESI+:1327.8 |
94 | 83 | ESI+:1314.7 |
95 | 83 | ESI+:1279.7 |
96 | 83 | ESI+:1315.8 |
97 | 83 | ESI+:1298.4 |
98 | 83 | ESI+:1280.8 |
99 | 83 | ESI+:1276.5 |
100 | 83 | ESI+:1293.7 |
101 | 83 | ESI+:1313.7[M+Na]+ |
102 | 83 | ESI+:1279.3 |
103 | 83 | ESI+:1304.6 |
104 | 83 | ESI+:1101.8 |
105 | 83 | ESI+:1101.8 |
[ Tables 6 to 5]
PEx | PSyn | DAT |
106 | 83 | ESI+:1457.6[M+Na]+ |
107 | 83 | ESI+:1328.1[M+Na]+ |
1 08 | 83 | ESI+:1292.9 |
109 | 83 | ESI+:1318.0 |
1 10 | 83 | ESI+:1313.0[M+Na]+ |
111 | 83 | ESI+:1089.8 |
112 | 83 | ESI+:1304.3 |
113 | 83 | ESI+:1314.8[M+Na]+ |
114 | 114 | ESI+:1312.0[M+Na]+ |
115 | 115 | ESI+:1303.3 |
116 | 116 | ESI+:1291.9 |
117 | 117 | ESI+:1 304.2 |
118 | 83 | ESI+:1277.8 |
119 | 83 | ESI+:1277.2 |
120 | 83 | ESI+:1306.5 |
121 | 83 | ESI+:1326.0[M+Na]+ |
122 | 83 | ESI+:1277.4 |
123 | 83 | ESI+:1278.3 |
124 | 124 | ESI+:1205.4 |
125 | 125 | ESI+:335.2 |
126 | 126 | ESI+:235.2 |
127 | 126 | ESI+:205.3 |
128 | 126 | ESI+:233.3 |
129 | 126 | ESI+:218.2 |
130 | 126 | ESI+:219.1 |
131 | 131 | ESI+:232.2 |
132 | 126 | ESI+:245.1[M+Na]+ |
133 | 133 | ESI+:448.3 |
134 | 134 | ESI+:445.3 |
135 | 135 | ESI+:348.2 |
136 | 136 | ESI+:345.2 |
1 37 | 137 | ESI+:547.4 |
138 | 1 38 | ESI+:544.3 |
139 | 139 | ESI+:447.3 |
140 | 140 | ESI+:444.3 |
141 | 141 | ESI+:473.3 |
[ Tables 6 to 6]
PEx | PSyn | DAT |
142 | 142 | ESI+:470.3 |
143 | 143 | ESI+:833.6 |
144 | 144 | ESI+:332.2 |
145 | 144 | ESI+:333.4 |
146 | 146 | ESI+:1076.1 |
147 | 146 | ESI+:1087.8 |
148 | 146 | ESI+:1087.8 |
149 | 149 | ESI+:1213.5 |
150 | 149 | ESI+:1262.7 |
151 | 149 | ESI+:1286.8 |
152 | 149 | ESI+:1287.7 |
153 | 149 | ESI+:1301.6 |
154 | 149 | ESI+:1323.7[M+Na]+ |
155 | 149 | ESI+:1286.9 |
156 | 149 | ESI+:1288.6 |
157 | 149 | ESI+:1300.9 |
158 | 149 | ESI+:1290.2 |
159 | 149 | ESI+:1297.0[M+Na]+ |
160 | 149 | ESI+:1280.3 |
161 | 149 | ESI+:1276.2 |
162 | 149 | ESI+:1273.7 |
163 | 163 | ESI+:627.5 |
164 | 163 | ESI+:623.5 |
165 | 163 | ESI+:649.6 |
166 | 163 | ESI+:637.3 |
167 | 163 | ESI+:662.5 |
168 | 163 | ESI+:649.3 |
169 | 163 | ESI+:637.5 |
170 | 163 | ESI+:649.4 |
171 | 171 | ESI+:585.2,587.1 |
[ Tables 6 to 7]
[ Tables 6 to 8]
PEx | PSyn | DAT |
191 | 188 | ESI+:837.7 |
192 | 192 | ESI+:485.9,487.9,489.8 |
193 | 181 | ESI+:588.0 |
194 | 182 | ESI+:741.1 |
195 | 9 | ESI+:655.2 |
196 | 13 | ESI+:965.3 |
197 | 23 | ESI+:861.4 |
198 | 29 | ESI+:975.4 |
199 | 19 | ESI+:733.4 |
200 | 124 | ESI+:1205.8 |
201 | 139 | ESI+:245.2 |
202 | 141 | ESI+:271.2 |
[ Tables 6 to 9]
PEx | PSyn | DAT |
203 | 203 | ESI+:474.0 |
204 | 204 | ESI+:598.1,600.2[M+Na]+ |
205 | 205 | ESI+:740.3 |
206 | 206 | ESI+:728.2 |
207 | 207 | ESI+:740.2 |
208 | 9 | ESI+:654.4 |
209 | 9 | ESI+:656.6 |
210 | 210 | ESI+:964.7 |
211 | 210 | ESI+:966.8 |
212 | 23 | ESI+:860.6 |
213 | 23 | ESI+:862.7 |
214 | 29 | ESI+:974.7 |
215 | 29 | ESI+:976.8 |
216 | 117 | ESI+:1446.7 |
217 | 117 | ESI+:1448.7 |
[ Tables 6 to 10]
PEx | PSyn | DAT |
218 | 218 | ESI+:252.1 |
219 | 219 | ESI+:220.1 |
220 | 220 | ESI+:298.0 |
221 | 221 | ESI+:514.2,516.2 |
222 | 222 | ESI+:580.3,582.2 |
223 | 6 | ESI+:668.5 |
224 | 13 | ESI+:978.6 |
225 | 225 | ESI+:820.6 |
226 | 23 | ESI+:716.5 |
227 | 29 | ESI+:830.6 |
228 | 117 | ESI+:1324.8[M+Na]+ |
[ Table 7-1]
[ Table 7-2]
[ Table 7-3]
[ Tables 7 to 4]
[ Tables 7 to 5]
[ Tables 7 to 6]
[ Tables 7 to 7]
[ Tables 7 to 8]
[ Tables 7 to 9]
[ Tables 7 to 10]
[ Tables 7 to 11]
[ Tables 7 to 12]
[ Tables 7 to 13]
[ Tables 7 to 14]
[ Tables 7 to 15]
[ Tables 7 to 16]
[ Tables 7 to 17]
Tables 7 to 18
[ Tables 7 to 19]
[ Tables 7 to 20]
[ Table 8-1]
[ Table 8-2]
[ Tables 8-3]
[ Tables 8 to 4]
[ Tables 8 to 5]
Ex | Syn | DAT |
55 | 9 | ESI+:1104.4 |
56 | 9 | ESI+:1107.9 |
[ Tables 8 to 6]
Ex | Syn | DAT |
57 | 57 | ES1+:1118.5 |
As examples of specific compounds of the formula (I) and the formula (IA) included in the present invention, compounds having any of the following structures are shown. These compounds may also be produced by the representative production methods, production examples and production methods of the examples shown above or a combination of these production methods or a method obvious to those skilled in the art.
Further, these compounds have excellent action of inducing the degradation of G12D mutant KRAS protein, and are expected to be useful as inhibitors of G12D mutant KRAS, and can be used as active ingredients in pharmaceutical compositions, for example, pharmaceutical compositions for the treatment of pancreatic cancer.
[ Table 9-1]
[ Table 9-2]
[ Tables 9-3]
[ Tables 9 to 4]
[ Tables 9 to 5]
[ Tables 9 to 6]
[ Tables 9 to 7]
[ Tables 9 to 8]
[ Tables 9 to 9]
[ Tables 9 to 10]
[ Tables 9 to 11]
Industrial applicability
The compound of the present invention or a salt thereof has an excellent effect of inducing the decomposition of G12D mutant KRAS protein, is useful as a G12D mutant KRAS inhibitor, and can be used as an active ingredient of a pharmaceutical composition, for example, a pharmaceutical composition for the treatment of pancreatic cancer.
Claims (17)
1. A compound of formula (IA) or a salt thereof,
In the formula (IA),
A is CR A or N, and the total number of the components is N,
R A is H or C 1-3 alkyl,
E is CH or N, and the R is H,
G is CR 2 or N, and the total number of the components is N,
R 1 is a naphthyl group which may be substituted by OH or a group selected from the group consisting of the following formula (II), formula (III) and formula (IV),
R 1a、R1b are identical or different from each other and are H, methyl, F or Cl,
R 1c is F, cl, methyl or ethyl,
R 2 is H, halogen, optionally substituted C 1-3 alkyl, cyclopropyl or vinyl,
R 3 is-P-Q or V,
P is-CH 2 -, -O-or-N (R P) -,
R P is H or C 1-3 alkyl which can be substituted,
Q is the following formula (V) or formula (VI),
V is the following formula (VII),
R Q, which are identical or different from each other, are OH or C 1-3 alkyl, and R Q is bonded only to carbon atoms as constituent atoms of rings selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
In addition, (i) when A is CR A, or (ii) when A is N and E or G is N,
V may be a saturated or unsaturated 7-to 8-membered bridged heterocyclic group other than the formula (VII),
M is a number from 0 to 2,
R 4 is an optionally substituted C 1-6 alkyl group, an optionally substituted 4-to 6-membered saturated heterocyclic group containing 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen, an optionally substituted 5-membered heteroaryl group containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen or an optionally substituted 6-membered heteroaryl group containing 1 to 3 nitrogen atoms,
R 5 is C 1-6 alkyl which may be substituted, C 3-6 cycloalkyl which may be substituted or a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 hetero atom selected from the group consisting of oxygen, sulfur and nitrogen,
R 6a、R6b, identical or different from one another, is H or C 1-6 -alkyl which may be substituted, or R 6a、R6b together with the carbon to which they are bonded may form C 3-6 -cycloalkyl which may be substituted or a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 heteroatom selected from the group consisting of oxygen, sulfur and nitrogen,
R 7 is H, halogen, C 1-3 alkyl, -SO 2CH3、C3-6 cycloalkyl, an optionally substituted 4-to 6-membered saturated heterocyclic group containing 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen, an optionally substituted 5-membered heteroaryl group containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen or a 6-membered heteroaryl group containing 1 to 3 nitrogen atoms,
L Y is-O- (optionally substituted C 1-3 alkylene) -, S- (C 1-3 alkylene which may be substituted) -, -SO 2 - (optionally substituted C 1-3 alkylene) -, -NR Y - (optionally substituted C 1-3 alkylene) -, and- (C 1-3 alkylene which may be substituted) -O-, C 1-3 alkylene which may be substituted) -S-, C 1-3 alkylene which may be substituted) -SO 2 -, C 1-3 alkylene which may be substituted) -NR Y -,
R Y is H or C 1-3 alkyl,
R P1 is OH or F, and the total number of the components is H,
R P2a is H or F,
R P2b is H, and the hydrogen atom,
W is optionally substituted phenylene or optionally substituted 6-membered heteroarene diyl having 1 to 3 nitrogen atoms,
X is a bond, -CH 2 -, -O-, -S-, or-NR 4x -,
R 4x is H or C 1-3 alkyl,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is- (L 1-L2-L3-L4) -,
L 1、L2、L3、L4, which are identical or different from one another, are selected from the group consisting of bond, -O-, -NR L1 -, pyrrolidinediyl which may be substituted, piperidinyl which may be substituted, piperazinediyl which may be substituted, C 1-3 alkylene which may be substituted and C=O,
R L1 is H or C 1-3 alkyl,
Z is NH or a 5-membered heteroarene diyl containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulphur and nitrogen,
Or Y-L-Z is the following formula (VIII),
2. A compound of formula (I) or a salt thereof,
In the formula (I) of the present invention,
A is CR A or N, and the total number of the components is N,
R A is H or C 1-3 alkyl,
R 1 is a naphthyl group which may be substituted by OH or a group selected from the group consisting of the following formula (II), formula (III) and formula (IV),
R 1a、R1b are identical or different from each other and are H, methyl, F or Cl,
R 1c is F, cl, methyl or ethyl,
R 2 is H, halogen, optionally substituted C 1-3 alkyl, cyclopropyl or vinyl,
R 3 is-P-Q or V,
P is-CH 2 -, -O-or-N (R P) -,
R P is H or C 1-3 alkyl which can be substituted,
Q is the following formula (V) or formula (VI),
V is the following formula (VII),
R Q, which are identical or different from each other, are OH or C 1-3 alkyl, and R Q is bonded only to carbon atoms as constituent atoms of rings selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
M is a number from 0 to 2,
R 4 is an optionally substituted C 1-6 alkyl group, an optionally substituted 4-to 6-membered saturated heterocyclic group containing 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen, an optionally substituted 5-membered heteroaryl group containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen or an optionally substituted 6-membered heteroaryl group containing 1 to 3 nitrogen atoms,
R 5 is C 1-6 alkyl which may be substituted, C 3-6 cycloalkyl which may be substituted or a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 hetero atom selected from the group consisting of oxygen, sulfur and nitrogen,
R 6a、R6b, identical or different from one another, is H or C 1-6 -alkyl which may be substituted, or R 6a、R6b together with the carbon to which they are bonded may form C 3-6 -cycloalkyl which may be substituted or a 4-to 6-membered saturated heterocyclic group which may be substituted and contains 1 heteroatom selected from the group consisting of oxygen, sulfur and nitrogen,
R 7 is H, halogen, C 1-3 alkyl, -SO 2CH3、C3-6 cycloalkyl, an optionally substituted 4-to 6-membered saturated heterocyclic group containing 1 to 2 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen, an optionally substituted 5-membered heteroaryl group containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulfur and nitrogen or a 6-membered heteroaryl group containing 1 to 3 nitrogen atoms,
W is optionally substituted phenylene or optionally substituted 6-membered heteroarene diyl having 1 to 3 nitrogen atoms,
X is a bond, -CH 2 -, -O-, -S-, or-NR 4x -,
R 4x is H or C 1-3 alkyl,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is- (L 1-L2-L3-L4) -,
L 1、L2、L3、L4, which are identical or different from one another, are selected from the group consisting of bond, -O-, -NR L1 -, pyrrolidinediyl which may be substituted, piperidinyl which may be substituted, piperazinediyl which may be substituted, C 1-3 alkylene which may be substituted and C=O,
R L1 is H or C 1-3 alkyl,
Z is NH or a 5-membered heteroarene diyl containing 1 to 4 heteroatoms selected from the group consisting of oxygen, sulphur and nitrogen,
Or Y-L-Z is the following formula (VIII),
3. The compound or salt according to claim 2, wherein,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is a bond, C 1-3 alkylene, C=O or a group selected from the group consisting of formula (XX), formula (XXI), formula (XXII), formula (XXIII), formula (XXIV) and formula (XXV) below,
R L1 is H or C 1-3 alkyl,
R L2、RL3, identical or different from one another, are H, F, OH, OCH 3 or C 1-3 alkyl which may be substituted,
R L is CH or N, and the total number of the components is,
N is an integer of 1 to 2,
Z is NH or a group selected from the group consisting of the following formulas (XXVI), formula (XXVII), formula (XXVIII) and formula (XXIX),
Or Y-L-Z is the following formula (VIII),
4. The compound according to claim 3, wherein R 1 is the following formula (IIa) or formula (IIIa),
R 1a、R1b are identical or different from each other and are H, methyl, F or Cl, R 3:
(i) When A is CR A, it is-P-Q,
P is-O-,
(Ii) When A is N, it is-P-Q or V, P is-O-or-N (R P) -,
R P is H or C 1-3 alkyl,
In either case of (i) and (ii), Q is a compound of formula (V) or formula (VI) below,
V is the following formula (VII),
R Q is the same or different from each other and is OH or methyl, R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
M is a number from 0 to 2,
R 4 is C 1-6 alkyl which may be substituted, 4-to 6-membered saturated heterocyclic group which may be substituted containing 1 to 2 hetero atoms selected from the group consisting of oxygen, sulfur and nitrogen, pyrazolyl which may be substituted, pyridinyl which may be substituted or pyrimidinyl which may be substituted,
R 5 is methyl, ethyl, isopropyl, tert-butyl or C 3-6 cycloalkyl,
R 6a、R6b, which are identical or different from each other, are H or C 1-3 alkyl, which C 1-3 alkyl may be substituted by a group selected from the group consisting of F, OH, OCH 3 and N (CH 3)2), or R 6a、R6b may form, together with the carbon to which they are bonded, a C 3-6 cycloalkyl group,
R 7 is H, halogen, C 1-3 alkyl, -SO 2CH3、C3-6 cycloalkyl or a group selected from the group consisting of formula (IX), formula (X), formula (XI), formula (XII), formula (XIII), formula (XIV), formula (XV), formula (XVI), formula (XVII) and formula (XVIII) below,
R 7a、R7b, identical or different from one another, are H or C 1-3 alkyl which may be substituted by OH,
W is the following formula (XIX),
W 1 and W 2:
(i) W 1 is CH, W 2 is C-SO 2CH3, or
(Ii) W 1、W2 are identical or different from each other and are CH, CF, CCl, CCH 3 or N, where R 7 is H when W 1 is CH and W 2 is C-SO 2CH3,
X is-O-or-NR 4x -,
R 4x is H or C 1-3 alkyl,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is a bond, C=O or a group selected from the group consisting of the following formula (XX) and formula (XXII),
R L1 is H or C 1-3 alkyl,
R L2、RL3, identical or different from one another, are H, F, OH, OCH 3 or C 1-3 alkyl which may be substituted,
N is an integer of 1 to 2,
Z is NH or a group selected from the group consisting of the following formulae (XXVI), formula (XXVII), formula (XXVIII) and formula (XXIX),
Or Y-L-Z is the following formula (VIII),
5. The compound or salt according to claim 4, wherein,
A is CH or N, and the A is H or N,
R 2 is halogen, C 1-3 alkyl, cyclopropyl or vinyl, the C 1-3 alkyl may be substituted with a group selected from the group consisting of OH and OCH 3,
R3:
(I) When A is CH, it is-P-Q,
P is-O-,
(Ii) A is-P-Q or V when A is N,
P is-O-or-N (R P) -,
R P is H or C 1-3 alkyl,
In either case of (i) and (ii), Q is a compound of formula (V) or formula (VI) below,
V is the following formula (VII),
R Q is the same or different from each other and is OH or methyl, R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V), a pyrrolidine ring represented by formula (VI) and a piperazine ring represented by formula (VII),
M is a number from 0 to 2,
R 4 is C 1-6 alkyl or tetrahydropyranyl which may be substituted with a group selected from the group consisting of F, OH, OCH 3、CF3、CHF2, cyclopropyl which may be substituted, pyrrolidinyl which may be substituted and tetrahydrofuranyl which may be substituted,
R 7 is halogen or a group selected from the group consisting of the following formula (IX), formula (X), formula (XI), formula (XII), formula (XIII) and formula (XIV),
R 7a、R7b, identical or different from one another, are H or C 1-3 alkyl which may be substituted by OH,
W is the following formula (XIX),
W 1、W2 are identical or different from each other and are CH or N,
X is-O-or-NH-,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is a bond and is preferably a bond,
Z is the following formula (XXVII),
6. The compound or salt according to claim 4, wherein,
A is N, and the number of the A is N,
R 3 is-P-Q,
P is-O-or-N (R P) -,
R P is H or C 1-3 alkyl,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
M is a number from 0 to 2,
W is the following formula (XIX),
W 1、W2 are identical or different from each other and are CH, CF, CCl, CCH 3 or N,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is a bond, C=O or a group selected from the group consisting of the following formula (XX) and formula (XXII),
R L1 is H or C 1-3 alkyl,
R L2、RL3, identical or different from one another, are H, F, OH, OCH 3 or C 1-3 alkyl which may be substituted,
N is an integer of 1 to 2,
Z is NH or a group selected from the group consisting of the following formulae (XXVI), formula (XXVII), formula (XXVIII) and formula (XXIX),
7. The compound or salt according to claim 4, wherein,
A is CH, and the A is CH,
R 3 is-P-Q,
P is-O-,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
M is a number from 0 to 2,
W is the following formula (XIX),
W 1、W2 are identical or different from each other and are CH, CF, CCl, CCH 3 or N,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is a bond, C=O or a group selected from the group consisting of the following formula (XX) and formula (XXII),
R L1 is H or C 1-3 alkyl,
R L2、RL3, identical or different from one another, are H, F, OH, OCH 3 or C 1-3 alkyl which may be substituted,
N is an integer of 1 to 2,
Z is NH or a group selected from the group consisting of the following formulae (XXVI), formula (XXVII), formula (XXVIII) and formula (XXIX),
8. The compound or salt according to claim 4, wherein,
A is N, and the number of the A is N,
R 3 is V, and the R is H,
V is the following formula (VII),
R Q which are identical or different from each other are OH or methyl, R Q is bonded only to a carbon atom as a ring constituting atom of the piperazine ring represented by formula (VII),
M is a number from 0 to 2,
W is the following formula (XIX),
W 1、W2 are identical or different from each other and are CH, CF, CCl, CCH 3 or N,
Y is phenylene or pyridyldiyl, which phenylene may be substituted by F,
L is a bond, C=O or a group selected from the group consisting of the following formula (XX) and formula (XXII),
R L1 is H or C 1-3 alkyl,
R L2、RL3, identical or different from one another, are H, F, OH, OCH 3 or C 1-3 alkyl which may be substituted,
N is an integer of 1 to 2,
Z is NH or a group selected from the group consisting of the following formulae (XXVI), formula (XXVII), formula (XXVIII) and formula (XXIX),
9. The compound or salt according to claim 5, wherein,
A is N, and the number of the A is N,
R 1 is the following formula (IIb),
R 3 is-P-Q,
P is-O-or-N (R P) -,
R P is H or C 1-3 alkyl,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
M is 0 or 1 and the number of the groups,
X is-O-.
10. The compound or salt according to claim 9, wherein,
R 2 is cyclopropyl, and the amino group is a cyclopropyl group,
R 3 is-P-Q,
P is-N (R P) -,
R P is H or C 1-3 alkyl,
Q is the following formula (V) or formula (VI),
R Q are the same or different from each other and are OH or methyl, and R Q is bonded only to a carbon atom as a constituent atom of a ring selected from the group consisting of an azetidine ring represented by formula (V) and a pyrrolidine ring represented by formula (VI),
M is 0 or 1 and the number of the groups,
R 4 is C 1-6 alkyl or tetrahydropyranyl which may be substituted by a group selected from the group consisting of OCH 3 and tetrahydrofuranyl,
R 5 is isopropyl group, and the amino group is a hydroxyl group,
R 6a is H, R 6b is C 1-3 alkyl which can be substituted by OH,
R 7 is a group of the following formula (IX), formula (X), formula (XI) or formula (XII),
R 7a is C 1-3 alkyl which may be substituted by OH,
W is the following formula (XIX),
W 1、W2 is CH, and the two are all CH,
Y is phenylene which may be substituted by F.
11. A compound or salt thereof according to claim 1 or claim 2, wherein the compounds of formula (I) and formula (IA) are selected from the group consisting of:
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } -2- { [ (2R) -oxopyrrolidin-2-yl ] methoxy } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } -2- { [ (2S) -oxopyrrolidin-2-yl ] methoxy } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -N- { (1R) -1- [4- (1-ethyl-1H-pyrazol-5-yl) phenyl ] -2-hydroxyethyl } -4-hydroxy-L-prolinamide,
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3S) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) -2-fluorophenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide,
(4R) -1- [ (2S) -2- (4- {4- [ ({ (7M) -6-cyclopropyl-4- { ethyl [ (3S) -pyrrolidin-3-yl ] amino } -7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] quinazolin-8-yl } oxy) methyl ] phenyl } -1H-1,2, 3-triazol-1-yl) -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide, and
(4R) -1- [ (2S) -2- {4- [4- ({ [ (7M) -6-cyclopropyl-7- (6-fluoro-5-methyl-1H-indazol-4-yl) -2- [ (2S) -2-methoxypropoxy ] -4- { methyl [ (3R) -pyrrolidin-3-yl ] amino } quinazolin-8-yl ] oxy } methyl) phenyl ] -1H-1,2, 3-triazol-1-yl } -3-methylbutanoyl ] -4-hydroxy-N- { (1R) -2-hydroxy-1- [4- (4-methyl-1, 3-thiazol-5-yl) phenyl ] ethyl } -L-prolinamide.
12. A pharmaceutical composition comprising a compound of claim 1 or claim 2, or a salt thereof, and one or more pharmaceutically acceptable excipients.
13. The pharmaceutical composition according to claim 12, which is a pharmaceutical composition for the treatment of pancreatic cancer.
14. Use of a compound of claim 1 or claim 2, or a salt thereof, in the manufacture of a pharmaceutical composition for the treatment of pancreatic cancer.
15. A compound according to claim 1 or claim 2, or a salt thereof, for use in the treatment of pancreatic cancer.
16. Use of a compound of claim 1 or claim 2, or a salt thereof, in the treatment of pancreatic cancer.
17. A method of treating pancreatic cancer comprising the step of administering to a subject an effective amount of a compound of claim 1 or claim 2, or a salt thereof.
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2022-038252 | 2022-03-11 | ||
JP2022126816 | 2022-08-09 | ||
JP2022-126816 | 2022-08-09 | ||
PCT/JP2023/009205 WO2023171781A1 (en) | 2022-03-11 | 2023-03-10 | Heterocyclic compound for inducing degradation of g12d mutant kras protein |
Publications (1)
Publication Number | Publication Date |
---|---|
CN118829640A true CN118829640A (en) | 2024-10-22 |
Family
ID=93065240
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202380025718.3A Pending CN118829640A (en) | 2022-03-11 | 2023-03-10 | Heterocyclic compounds for inducing G12D mutant KRAS protein degradation |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN118829640A (en) |
-
2023
- 2023-03-10 CN CN202380025718.3A patent/CN118829640A/en active Pending
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7169729B1 (en) | Quinazoline compounds for inducing degradation of G12D mutant KRAS protein | |
CN112266384B (en) | ErbB receptor inhibitors | |
JPWO2018143315A1 (en) | Quinazoline compounds | |
WO2023171781A1 (en) | Heterocyclic compound for inducing degradation of g12d mutant kras protein | |
CN118317774A (en) | Pharmaceutical compositions comprising quinazoline compounds | |
JP7440710B1 (en) | Heterocyclic compound that acts on G12D mutant KRAS protein | |
CN116867788A (en) | 4-aminoquinazoline compounds | |
WO2016058501A1 (en) | 5-methyl-2-(pyridine-2-ylamino)-8h-pyridino[2,3-d]pyrimidine-7-one compound | |
WO2024034657A1 (en) | Pharmaceutical composition comprising heterocyclic compound | |
TW202412789A (en) | Heterocyclic compound for inducing degradation of mutant kras protein | |
CN117624170A (en) | Compounds with anti-KRAS mutant tumor activity | |
CN116801883A (en) | Heteroaromatic ring compounds, preparation method and application thereof | |
WO2024034593A1 (en) | Heterocyclic compound for inducing degradation of g12v mutant kras protein | |
WO2024034591A1 (en) | Heterocyclic compound for inhibiting and/or inducing degradation of kras protein | |
CN118829640A (en) | Heterocyclic compounds for inducing G12D mutant KRAS protein degradation | |
KR20240162042A (en) | Heterocyclic compounds for inducing degradation of G12D mutant KRAS protein | |
CN117800975A (en) | Compounds with anti-KRAS mutant tumor activity | |
WO2023072913A1 (en) | Benzo[h]quinazoline-4-amine derivatives for the treatment of cancer | |
WO2023196629A1 (en) | Benzo[h]quinazoline-4-amine derivatives for the treatment of cancer | |
JP2023544847A (en) | Benzo[h]quinazolin-4-amine and thieno[3,2-h]quinazolin-4-amine derivatives for the treatment of cancer | |
EP4423070A1 (en) | Benzo[h]quinazoline-4-amine derivatives for the treatment of cancer |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication |