WO2024146502A1 - Wee1 degrading compounds - Google Patents
Wee1 degrading compounds Download PDFInfo
- Publication number
- WO2024146502A1 WO2024146502A1 PCT/CN2024/070070 CN2024070070W WO2024146502A1 WO 2024146502 A1 WO2024146502 A1 WO 2024146502A1 CN 2024070070 W CN2024070070 W CN 2024070070W WO 2024146502 A1 WO2024146502 A1 WO 2024146502A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- optionally substituted
- compound
- alkyl
- independently
- halogen
- Prior art date
Links
- 150000001875 compounds Chemical class 0.000 title claims abstract description 433
- 101150040313 Wee1 gene Proteins 0.000 title description 7
- 230000000593 degrading effect Effects 0.000 title description 2
- 238000000034 method Methods 0.000 claims abstract description 39
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 22
- 201000010099 disease Diseases 0.000 claims abstract description 17
- 238000011282 treatment Methods 0.000 claims abstract description 16
- 229910052736 halogen Inorganic materials 0.000 claims description 392
- 150000002367 halogens Chemical class 0.000 claims description 389
- -1 heterocyclene Chemical group 0.000 claims description 374
- 229910052739 hydrogen Inorganic materials 0.000 claims description 325
- 239000001257 hydrogen Substances 0.000 claims description 325
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 253
- 150000002431 hydrogen Chemical class 0.000 claims description 188
- 125000004093 cyano group Chemical group *C#N 0.000 claims description 185
- 125000000623 heterocyclic group Chemical group 0.000 claims description 185
- 125000006273 (C1-C3) alkyl group Chemical group 0.000 claims description 175
- 125000004169 (C1-C6) alkyl group Chemical group 0.000 claims description 167
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 claims description 117
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 claims description 116
- 125000000217 alkyl group Chemical group 0.000 claims description 108
- 125000004429 atom Chemical group 0.000 claims description 89
- 125000000753 cycloalkyl group Chemical group 0.000 claims description 87
- 125000004452 carbocyclyl group Chemical group 0.000 claims description 79
- 150000003839 salts Chemical class 0.000 claims description 76
- 125000000171 (C1-C6) haloalkyl group Chemical group 0.000 claims description 75
- 125000001309 chloro group Chemical group Cl* 0.000 claims description 73
- 125000002947 alkylene group Chemical group 0.000 claims description 69
- 125000002993 cycloalkylene group Chemical group 0.000 claims description 68
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 claims description 65
- 125000006716 (C1-C6) heteroalkyl group Chemical group 0.000 claims description 60
- 239000012453 solvate Substances 0.000 claims description 55
- 125000001072 heteroaryl group Chemical group 0.000 claims description 50
- 125000004191 (C1-C6) alkoxy group Chemical group 0.000 claims description 49
- 239000000651 prodrug Substances 0.000 claims description 40
- 229940002612 prodrug Drugs 0.000 claims description 40
- 125000006583 (C1-C3) haloalkyl group Chemical group 0.000 claims description 38
- 125000000449 nitro group Chemical group [O-][N+](*)=O 0.000 claims description 38
- 229910052757 nitrogen Inorganic materials 0.000 claims description 36
- 101000941994 Homo sapiens Protein cereblon Proteins 0.000 claims description 35
- 239000003446 ligand Substances 0.000 claims description 34
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 33
- 125000001153 fluoro group Chemical group F* 0.000 claims description 32
- 125000003118 aryl group Chemical group 0.000 claims description 30
- 125000006274 (C1-C3)alkoxy group Chemical group 0.000 claims description 29
- 125000003545 alkoxy group Chemical group 0.000 claims description 29
- 125000004433 nitrogen atom Chemical group N* 0.000 claims description 29
- 125000006577 C1-C6 hydroxyalkyl group Chemical group 0.000 claims description 26
- 125000004103 aminoalkyl group Chemical group 0.000 claims description 26
- 206010028980 Neoplasm Diseases 0.000 claims description 25
- 201000011510 cancer Diseases 0.000 claims description 23
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 claims description 20
- 125000004404 heteroalkyl group Chemical group 0.000 claims description 20
- 125000006587 (C5-C10) heteroarylene group Chemical group 0.000 claims description 19
- 125000005549 heteroarylene group Chemical group 0.000 claims description 19
- 125000001424 substituent group Chemical group 0.000 claims description 19
- 239000008194 pharmaceutical composition Substances 0.000 claims description 18
- VGMJQHONPAXABH-UHFFFAOYSA-N 4,5,6,7-tetrahydro-1h-pyrazolo[4,3-c]pyridine Chemical class C1NCCC2=C1C=NN2 VGMJQHONPAXABH-UHFFFAOYSA-N 0.000 claims description 17
- OOGKXAWZILGOBA-UHFFFAOYSA-N 4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazine Chemical class C1NCCN2N=CC=C21 OOGKXAWZILGOBA-UHFFFAOYSA-N 0.000 claims description 17
- 125000004474 heteroalkylene group Chemical group 0.000 claims description 17
- 125000006652 (C3-C12) cycloalkyl group Chemical group 0.000 claims description 16
- 125000005913 (C3-C6) cycloalkyl group Chemical group 0.000 claims description 16
- 125000006708 (C5-C14) heteroaryl group Chemical group 0.000 claims description 16
- 125000005915 C6-C14 aryl group Chemical group 0.000 claims description 16
- 125000000304 alkynyl group Chemical group 0.000 claims description 15
- 125000003342 alkenyl group Chemical group 0.000 claims description 14
- 125000001188 haloalkyl group Chemical group 0.000 claims description 13
- 125000005843 halogen group Chemical group 0.000 claims description 13
- NOIXNOMHHWGUTG-UHFFFAOYSA-N 2-[[4-[4-pyridin-4-yl-1-(2,2,2-trifluoroethyl)pyrazol-3-yl]phenoxy]methyl]quinoline Chemical compound C=1C=C(OCC=2N=C3C=CC=CC3=CC=2)C=CC=1C1=NN(CC(F)(F)F)C=C1C1=CC=NC=C1 NOIXNOMHHWGUTG-UHFFFAOYSA-N 0.000 claims description 12
- 125000001559 cyclopropyl group Chemical group [H]C1([H])C([H])([H])C1([H])* 0.000 claims description 11
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 11
- 125000000882 C2-C6 alkenyl group Chemical group 0.000 claims description 10
- 125000003601 C2-C6 alkynyl group Chemical group 0.000 claims description 10
- 125000002887 hydroxy group Chemical group [H]O* 0.000 claims description 9
- 229910052760 oxygen Inorganic materials 0.000 claims description 7
- 229910052717 sulfur Inorganic materials 0.000 claims description 7
- 241001465754 Metazoa Species 0.000 claims description 6
- JXASPPWQHFOWPL-UHFFFAOYSA-N Tamarixin Natural products C1=C(O)C(OC)=CC=C1C1=C(OC2C(C(O)C(O)C(CO)O2)O)C(=O)C2=C(O)C=C(O)C=C2O1 JXASPPWQHFOWPL-UHFFFAOYSA-N 0.000 claims description 6
- 230000001613 neoplastic effect Effects 0.000 claims description 6
- 125000004178 (C1-C4) alkyl group Chemical group 0.000 claims description 5
- 239000003814 drug Substances 0.000 claims description 5
- 238000004519 manufacturing process Methods 0.000 claims description 3
- 125000001313 C5-C10 heteroaryl group Chemical group 0.000 claims description 2
- 150000001336 alkenes Chemical class 0.000 claims description 2
- 102000015367 CRBN Human genes 0.000 claims 10
- 101000621390 Homo sapiens Wee1-like protein kinase Proteins 0.000 abstract description 43
- 102100023037 Wee1-like protein kinase Human genes 0.000 abstract description 41
- 230000004063 proteosomal degradation Effects 0.000 abstract description 4
- 238000010798 ubiquitination Methods 0.000 abstract description 3
- 230000034512 ubiquitination Effects 0.000 abstract description 3
- 239000000203 mixture Substances 0.000 description 179
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 166
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 123
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 105
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 98
- 239000000243 solution Substances 0.000 description 77
- KNCYXPMJDCCGSJ-UHFFFAOYSA-N piperidine-2,6-dione Chemical compound O=C1CCCC(=O)N1 KNCYXPMJDCCGSJ-UHFFFAOYSA-N 0.000 description 63
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 60
- 229910001868 water Inorganic materials 0.000 description 60
- 239000011541 reaction mixture Substances 0.000 description 52
- 125000004432 carbon atom Chemical group C* 0.000 description 50
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 description 44
- 230000002829 reductive effect Effects 0.000 description 44
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 43
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-Dimethylformamide Chemical compound CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 42
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 40
- 235000019439 ethyl acetate Nutrition 0.000 description 39
- 238000005160 1H NMR spectroscopy Methods 0.000 description 37
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 37
- 125000001246 bromo group Chemical group Br* 0.000 description 35
- 125000004943 pyrimidin-6-yl group Chemical group N1=CN=CC=C1* 0.000 description 35
- 150000003254 radicals Chemical class 0.000 description 35
- IAZDPXIOMUYVGZ-WFGJKAKNSA-N Dimethyl sulfoxide Chemical compound [2H]C([2H])([2H])S(=O)C([2H])([2H])[2H] IAZDPXIOMUYVGZ-WFGJKAKNSA-N 0.000 description 32
- 239000012044 organic layer Substances 0.000 description 32
- 125000004194 piperazin-1-yl group Chemical group [H]N1C([H])([H])C([H])([H])N(*)C([H])([H])C1([H])[H] 0.000 description 30
- 125000004450 alkenylene group Chemical group 0.000 description 29
- 239000007832 Na2SO4 Substances 0.000 description 28
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 28
- 229910052938 sodium sulfate Inorganic materials 0.000 description 28
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 27
- 229940093499 ethyl acetate Drugs 0.000 description 27
- 239000012267 brine Substances 0.000 description 26
- 238000006243 chemical reaction Methods 0.000 description 26
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical compound O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 26
- 102100032783 Protein cereblon Human genes 0.000 description 25
- 125000004105 2-pyridyl group Chemical group N1=C([*])C([H])=C([H])C([H])=C1[H] 0.000 description 24
- HEDRZPFGACZZDS-MICDWDOJSA-N Trichloro(2H)methane Chemical compound [2H]C(Cl)(Cl)Cl HEDRZPFGACZZDS-MICDWDOJSA-N 0.000 description 24
- FJDQFPXHSGXQBY-UHFFFAOYSA-L caesium carbonate Chemical compound [Cs+].[Cs+].[O-]C([O-])=O FJDQFPXHSGXQBY-UHFFFAOYSA-L 0.000 description 24
- 239000000126 substance Substances 0.000 description 24
- 125000004419 alkynylene group Chemical group 0.000 description 23
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 21
- 229910052799 carbon Inorganic materials 0.000 description 21
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 19
- 239000012071 phase Substances 0.000 description 19
- 238000010898 silica gel chromatography Methods 0.000 description 19
- 125000003710 aryl alkyl group Chemical group 0.000 description 16
- 239000000706 filtrate Substances 0.000 description 16
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-Diisopropylethylamine (DIPEA) Chemical compound CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 0.000 description 15
- DTQVDTLACAAQTR-UHFFFAOYSA-N Trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 15
- 125000003709 fluoroalkyl group Chemical group 0.000 description 15
- 125000004446 heteroarylalkyl group Chemical group 0.000 description 15
- 125000004415 heterocyclylalkyl group Chemical group 0.000 description 15
- WYURNTSHIVDZCO-UHFFFAOYSA-N tetrahydrofuran Substances C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 15
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 14
- KDLHZDBZIXYQEI-UHFFFAOYSA-N Palladium Chemical compound [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 description 14
- 125000004482 piperidin-4-yl group Chemical group N1CCC(CC1)* 0.000 description 14
- 239000000741 silica gel Substances 0.000 description 14
- 229910002027 silica gel Inorganic materials 0.000 description 14
- 238000003786 synthesis reaction Methods 0.000 description 14
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 13
- 238000003818 flash chromatography Methods 0.000 description 13
- 230000007115 recruitment Effects 0.000 description 13
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 12
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 12
- 229910000024 caesium carbonate Inorganic materials 0.000 description 12
- 230000015556 catabolic process Effects 0.000 description 12
- 238000006731 degradation reaction Methods 0.000 description 12
- 239000012634 fragment Substances 0.000 description 12
- IXCSERBJSXMMFS-UHFFFAOYSA-N hydrogen chloride Substances Cl.Cl IXCSERBJSXMMFS-UHFFFAOYSA-N 0.000 description 12
- 229910000041 hydrogen chloride Inorganic materials 0.000 description 12
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical class [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 11
- 125000005884 carbocyclylalkyl group Chemical group 0.000 description 11
- 239000012043 crude product Substances 0.000 description 11
- 125000004122 cyclic group Chemical group 0.000 description 11
- 239000012074 organic phase Substances 0.000 description 11
- 238000002953 preparative HPLC Methods 0.000 description 11
- KXKVLQRXCPHEJC-UHFFFAOYSA-N acetic acid trimethyl ester Natural products COC(C)=O KXKVLQRXCPHEJC-UHFFFAOYSA-N 0.000 description 10
- 239000002253 acid Substances 0.000 description 10
- 230000015572 biosynthetic process Effects 0.000 description 10
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 10
- 229910052805 deuterium Inorganic materials 0.000 description 10
- NCAIGTHBQTXTLR-UHFFFAOYSA-N phentermine hydrochloride Chemical compound [Cl-].CC(C)([NH3+])CC1=CC=CC=C1 NCAIGTHBQTXTLR-UHFFFAOYSA-N 0.000 description 10
- VZBQJKIOAOUYJL-UHFFFAOYSA-N (1-methyl-1h-imidazol-2-yl)-(3-methyl-4-{3-[(pyridin-3-ylmethyl)-amino]-propoxy}-benzofuran-2-yl)-methanone Chemical compound C=12C(C)=C(C(=O)C=3N(C=CN=3)C)OC2=CC=CC=1OCCCNCC1=CC=CN=C1 VZBQJKIOAOUYJL-UHFFFAOYSA-N 0.000 description 9
- CMRQAKJTXKOGSF-UHFFFAOYSA-N 3-(6-bromo-3-oxo-1h-isoindol-2-yl)piperidine-2,6-dione Chemical compound C1C2=CC(Br)=CC=C2C(=O)N1C1CCC(=O)NC1=O CMRQAKJTXKOGSF-UHFFFAOYSA-N 0.000 description 9
- YZCKVEUIGOORGS-OUBTZVSYSA-N Deuterium Chemical compound [2H] YZCKVEUIGOORGS-OUBTZVSYSA-N 0.000 description 9
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 9
- 210000004027 cell Anatomy 0.000 description 9
- 239000012299 nitrogen atmosphere Substances 0.000 description 9
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 9
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 8
- 150000001721 carbon Chemical group 0.000 description 8
- BWHMMNNQKKPAPP-UHFFFAOYSA-L potassium carbonate Chemical compound [K+].[K+].[O-]C([O-])=O BWHMMNNQKKPAPP-UHFFFAOYSA-L 0.000 description 8
- 238000002360 preparation method Methods 0.000 description 8
- 239000000377 silicon dioxide Substances 0.000 description 8
- DMCXZVRRTPCCRY-UHFFFAOYSA-N 1-(4-nitrophenyl)piperidine-4-carbaldehyde Chemical compound C1=CC([N+](=O)[O-])=CC=C1N1CCC(C=O)CC1 DMCXZVRRTPCCRY-UHFFFAOYSA-N 0.000 description 7
- 239000003795 chemical substances by application Substances 0.000 description 7
- 125000004431 deuterium atom Chemical group 0.000 description 7
- 238000001914 filtration Methods 0.000 description 7
- 150000002430 hydrocarbons Chemical group 0.000 description 7
- 125000001841 imino group Chemical group [H]N=* 0.000 description 7
- 125000004043 oxo group Chemical group O=* 0.000 description 7
- 239000002904 solvent Substances 0.000 description 7
- 230000008901 benefit Effects 0.000 description 6
- 239000003208 petroleum Substances 0.000 description 6
- 230000017854 proteolysis Effects 0.000 description 6
- QAEDZJGFFMLHHQ-UHFFFAOYSA-N trifluoroacetic anhydride Chemical compound FC(F)(F)C(=O)OC(=O)C(F)(F)F QAEDZJGFFMLHHQ-UHFFFAOYSA-N 0.000 description 6
- OIVLITBTBDPEFK-UHFFFAOYSA-N 5,6-dihydrouracil Chemical compound O=C1CCNC(=O)N1 OIVLITBTBDPEFK-UHFFFAOYSA-N 0.000 description 5
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 5
- VHUUQVKOLVNVRT-UHFFFAOYSA-N Ammonium hydroxide Chemical compound [NH4+].[OH-] VHUUQVKOLVNVRT-UHFFFAOYSA-N 0.000 description 5
- 239000012298 atmosphere Substances 0.000 description 5
- 239000002585 base Substances 0.000 description 5
- 238000004440 column chromatography Methods 0.000 description 5
- 125000000524 functional group Chemical group 0.000 description 5
- 125000005842 heteroatom Chemical group 0.000 description 5
- 238000004128 high performance liquid chromatography Methods 0.000 description 5
- 238000002347 injection Methods 0.000 description 5
- 239000007924 injection Substances 0.000 description 5
- 239000000543 intermediate Substances 0.000 description 5
- 125000004430 oxygen atom Chemical group O* 0.000 description 5
- 108090000623 proteins and genes Proteins 0.000 description 5
- 102000004169 proteins and genes Human genes 0.000 description 5
- 238000000746 purification Methods 0.000 description 5
- 125000003226 pyrazolyl group Chemical group 0.000 description 5
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 5
- 239000011780 sodium chloride Substances 0.000 description 5
- MBVAZGKHRAOOJA-UHFFFAOYSA-N 1-(4-aminophenyl)piperidin-4-one Chemical compound C1=CC(N)=CC=C1N1CCC(=O)CC1 MBVAZGKHRAOOJA-UHFFFAOYSA-N 0.000 description 4
- BKWJAKQVGHWELA-UHFFFAOYSA-N 1-[6-(2-hydroxypropan-2-yl)-2-pyridinyl]-6-[4-(4-methyl-1-piperazinyl)anilino]-2-prop-2-enyl-3-pyrazolo[3,4-d]pyrimidinone Chemical compound C1CN(C)CCN1C(C=C1)=CC=C1NC1=NC=C2C(=O)N(CC=C)N(C=3N=C(C=CC=3)C(C)(C)O)C2=N1 BKWJAKQVGHWELA-UHFFFAOYSA-N 0.000 description 4
- WFQDTOYDVUWQMS-UHFFFAOYSA-N 1-fluoro-4-nitrobenzene Chemical compound [O-][N+](=O)C1=CC=C(F)C=C1 WFQDTOYDVUWQMS-UHFFFAOYSA-N 0.000 description 4
- FDMZEIZGNKTOII-UHFFFAOYSA-N 3-(5-bromo-3-oxo-1H-isoindol-2-yl)piperidine-2,6-dione Chemical compound BrC1=CC=C2CN(C3CCC(=O)NC3=O)C(=O)C2=C1 FDMZEIZGNKTOII-UHFFFAOYSA-N 0.000 description 4
- WMFOQBRAJBCJND-UHFFFAOYSA-M Lithium hydroxide Chemical compound [Li+].[OH-] WMFOQBRAJBCJND-UHFFFAOYSA-M 0.000 description 4
- 229910017906 NH3H2O Inorganic materials 0.000 description 4
- 229910020889 NaBH3 Inorganic materials 0.000 description 4
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 4
- 108091000080 Phosphotransferase Proteins 0.000 description 4
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 4
- 150000007513 acids Chemical class 0.000 description 4
- 229950009557 adavosertib Drugs 0.000 description 4
- 150000001412 amines Chemical class 0.000 description 4
- 239000007864 aqueous solution Substances 0.000 description 4
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 4
- 239000011203 carbon fibre reinforced carbon Substances 0.000 description 4
- 230000022131 cell cycle Effects 0.000 description 4
- 229910052681 coesite Inorganic materials 0.000 description 4
- 229910052906 cristobalite Inorganic materials 0.000 description 4
- 125000001316 cycloalkyl alkyl group Chemical group 0.000 description 4
- MXFYYFVVIIWKFE-UHFFFAOYSA-N dicyclohexyl-[2-[2,6-di(propan-2-yloxy)phenyl]phenyl]phosphane Chemical compound CC(C)OC1=CC=CC(OC(C)C)=C1C1=CC=CC=C1P(C1CCCCC1)C1CCCCC1 MXFYYFVVIIWKFE-UHFFFAOYSA-N 0.000 description 4
- 239000003480 eluent Substances 0.000 description 4
- 238000009472 formulation Methods 0.000 description 4
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 4
- 230000000155 isotopic effect Effects 0.000 description 4
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 4
- 239000001301 oxygen Substances 0.000 description 4
- 102000020233 phosphotransferase Human genes 0.000 description 4
- 229910000027 potassium carbonate Inorganic materials 0.000 description 4
- 229920006395 saturated elastomer Polymers 0.000 description 4
- 239000007787 solid Substances 0.000 description 4
- 239000007858 starting material Substances 0.000 description 4
- 229910052682 stishovite Inorganic materials 0.000 description 4
- 125000003107 substituted aryl group Chemical group 0.000 description 4
- 239000011593 sulfur Substances 0.000 description 4
- 230000001225 therapeutic effect Effects 0.000 description 4
- 125000000464 thioxo group Chemical group S=* 0.000 description 4
- 238000012546 transfer Methods 0.000 description 4
- 229910052905 tridymite Inorganic materials 0.000 description 4
- AQRLNPVMDITEJU-UHFFFAOYSA-N triethylsilane Chemical compound CC[SiH](CC)CC AQRLNPVMDITEJU-UHFFFAOYSA-N 0.000 description 4
- 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
- JIZNOSHLAWSCKW-UHFFFAOYSA-N 1-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]piperidine-4-carbaldehyde Chemical compound O=C1NC(CCC1N1C(C2=CC=C(C=C2C1)N1CCC(CC1)C=O)=O)=O JIZNOSHLAWSCKW-UHFFFAOYSA-N 0.000 description 3
- 125000004214 1-pyrrolidinyl group Chemical group [H]C1([H])N(*)C([H])([H])C([H])([H])C1([H])[H] 0.000 description 3
- ZWCVKPZAXXWBHU-UHFFFAOYSA-N 8-azaspiro[4.5]decan-3-one Chemical compound C1C(=O)CCC21CCNCC2 ZWCVKPZAXXWBHU-UHFFFAOYSA-N 0.000 description 3
- 101150012716 CDK1 gene Proteins 0.000 description 3
- 102100025064 Cellular tumor antigen p53 Human genes 0.000 description 3
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 238000005481 NMR spectroscopy Methods 0.000 description 3
- MUBZPKHOEPUJKR-UHFFFAOYSA-N Oxalic acid Chemical compound OC(=O)C(O)=O MUBZPKHOEPUJKR-UHFFFAOYSA-N 0.000 description 3
- OFOBLEOULBTSOW-UHFFFAOYSA-N Propanedioic acid Natural products OC(=O)CC(O)=O OFOBLEOULBTSOW-UHFFFAOYSA-N 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 102000006275 Ubiquitin-Protein Ligases Human genes 0.000 description 3
- 108010083111 Ubiquitin-Protein Ligases Proteins 0.000 description 3
- 150000001732 carboxylic acid derivatives Chemical class 0.000 description 3
- 239000003153 chemical reaction reagent Substances 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
- 238000001816 cooling Methods 0.000 description 3
- XBDQKXXYIPTUBI-UHFFFAOYSA-N dimethylselenoniopropionate Natural products CCC(O)=O XBDQKXXYIPTUBI-UHFFFAOYSA-N 0.000 description 3
- 230000000694 effects Effects 0.000 description 3
- 239000012065 filter cake Substances 0.000 description 3
- 150000003840 hydrochlorides Chemical class 0.000 description 3
- PQNFLJBBNBOBRQ-UHFFFAOYSA-N indane Chemical group C1=CC=C2CCCC2=C1 PQNFLJBBNBOBRQ-UHFFFAOYSA-N 0.000 description 3
- 239000003112 inhibitor Substances 0.000 description 3
- 125000002346 iodo group Chemical group I* 0.000 description 3
- 230000037361 pathway Effects 0.000 description 3
- 230000000144 pharmacologic effect Effects 0.000 description 3
- SCVFZCLFOSHCOH-UHFFFAOYSA-M potassium acetate Chemical compound [K+].CC([O-])=O SCVFZCLFOSHCOH-UHFFFAOYSA-M 0.000 description 3
- 230000003389 potentiating effect Effects 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 230000000069 prophylactic effect Effects 0.000 description 3
- 239000000376 reactant Substances 0.000 description 3
- 238000004007 reversed phase HPLC Methods 0.000 description 3
- 239000011734 sodium Substances 0.000 description 3
- 230000003595 spectral effect Effects 0.000 description 3
- 238000006467 substitution reaction Methods 0.000 description 3
- 208000024891 symptom Diseases 0.000 description 3
- 238000010189 synthetic method Methods 0.000 description 3
- QSQWENQPOSRWLP-UHFFFAOYSA-N tert-butyl 4-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazol-1-yl]piperidine-1-carboxylate Chemical compound C1CN(C(=O)OC(C)(C)C)CCC1N1N=CC(B2OC(C)(C)C(C)(C)O2)=C1 QSQWENQPOSRWLP-UHFFFAOYSA-N 0.000 description 3
- CXWXQJXEFPUFDZ-UHFFFAOYSA-N tetralin Chemical group C1=CC=C2CCCCC2=C1 CXWXQJXEFPUFDZ-UHFFFAOYSA-N 0.000 description 3
- 239000011701 zinc Substances 0.000 description 3
- VPPLIPSVBSZVBN-UHFFFAOYSA-N 1-(2,2,2-trifluoroacetyl)pyrrolidin-3-one Chemical compound FC(F)(F)C(=O)N1CCC(=O)C1 VPPLIPSVBSZVBN-UHFFFAOYSA-N 0.000 description 2
- CRGRJUULGQHKOZ-UHFFFAOYSA-N 1-(4-nitrophenyl)piperidin-4-one Chemical compound C1=CC([N+](=O)[O-])=CC=C1N1CCC(=O)CC1 CRGRJUULGQHKOZ-UHFFFAOYSA-N 0.000 description 2
- NTSGMZSNBBYMCF-MUUNZHRXSA-N 1-[(7R)-7-ethyl-7-hydroxy-5,6-dihydrocyclopenta[b]pyridin-2-yl]-6-(4-piperazin-1-ylanilino)-2-prop-2-enylpyrazolo[3,4-d]pyrimidin-3-one Chemical compound C1=C2C(=NC(=C1)N1N(C(=O)C3=CN=C(NC4=CC=C(N5CCNCC5)C=C4)N=C13)CC=C)[C@@](CC)(CC2)O NTSGMZSNBBYMCF-MUUNZHRXSA-N 0.000 description 2
- YBYIRNPNPLQARY-UHFFFAOYSA-N 1H-indene Chemical group C1=CC=C2CC=CC2=C1 YBYIRNPNPLQARY-UHFFFAOYSA-N 0.000 description 2
- HZAXFHJVJLSVMW-UHFFFAOYSA-N 2-Aminoethan-1-ol Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 description 2
- YTKVMOBHMCOGHZ-UHFFFAOYSA-N 3-(3-oxo-6-piperazin-1-yl-1H-isoindol-2-yl)piperidine-2,6-dione Chemical compound O=C1N(CC2=C1C=CC(=C2)N1CCNCC1)C1CCC(=O)NC1=O YTKVMOBHMCOGHZ-UHFFFAOYSA-N 0.000 description 2
- GUOJSPAORSZIHQ-UHFFFAOYSA-N 3-(4-nitrophenyl)-3,9-diazaspiro[5.5]undecane Chemical compound C1=CC([N+](=O)[O-])=CC=C1N1CCC2(CCNCC2)CC1 GUOJSPAORSZIHQ-UHFFFAOYSA-N 0.000 description 2
- CMUYBPMCSWFBSY-UHFFFAOYSA-N 3-[6-(2,7-diazaspiro[3.5]nonan-2-yl)-3-oxo-1H-isoindol-2-yl]piperidine-2,6-dione Chemical compound O=C1N(CC2=C1C=CC(=C2)N1CC2(C1)CCNCC2)C1CCC(=O)NC1=O CMUYBPMCSWFBSY-UHFFFAOYSA-N 0.000 description 2
- WFNGKTSPXVLWMD-UHFFFAOYSA-N 3-[6-[4-(hydroxymethyl)piperidin-1-yl]-3-oxo-1H-isoindol-2-yl]piperidine-2,6-dione Chemical compound OCC1CCN(CC1)C1=CC=C2C(=O)N(CC2=C1)C1CCC(=O)NC1=O WFNGKTSPXVLWMD-UHFFFAOYSA-N 0.000 description 2
- NHQDETIJWKXCTC-UHFFFAOYSA-N 3-chloroperbenzoic acid Chemical compound OOC(=O)C1=CC=CC(Cl)=C1 NHQDETIJWKXCTC-UHFFFAOYSA-N 0.000 description 2
- PWRBCZZQRRPXAB-UHFFFAOYSA-N 3-chloropyridine Chemical compound ClC1=CC=CN=C1 PWRBCZZQRRPXAB-UHFFFAOYSA-N 0.000 description 2
- MSHFRERJPWKJFX-UHFFFAOYSA-N 4-Methoxybenzyl alcohol Chemical compound COC1=CC=C(CO)C=C1 MSHFRERJPWKJFX-UHFFFAOYSA-N 0.000 description 2
- YYOYIBYKZDYMGL-UHFFFAOYSA-N CC(C)(C)OC(=O)N1CC2(C1)CCC(CO)CC2 Chemical compound CC(C)(C)OC(=O)N1CC2(C1)CCC(CO)CC2 YYOYIBYKZDYMGL-UHFFFAOYSA-N 0.000 description 2
- QEZGJKGCVZCSSA-UHFFFAOYSA-N CCCC(CCC)C1=CC=CC(C(CCC)CCC)=C1N(CN1C2=C(C(CCC)CCC)C=CC=C2C(CCC)CCC)C(Cl)=C1Cl Chemical compound CCCC(CCC)C1=CC=CC(C(CCC)CCC)=C1N(CN1C2=C(C(CCC)CCC)C=CC=C2C(CCC)CCC)C(Cl)=C1Cl QEZGJKGCVZCSSA-UHFFFAOYSA-N 0.000 description 2
- PHGNMZYFCWWFPS-UHFFFAOYSA-N COC=C1CCC2(CCN(CC2)C(=O)OC(C)(C)C)CC1 Chemical compound COC=C1CCC2(CCN(CC2)C(=O)OC(C)(C)C)CC1 PHGNMZYFCWWFPS-UHFFFAOYSA-N 0.000 description 2
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 2
- 230000005778 DNA damage Effects 0.000 description 2
- 231100000277 DNA damage Toxicity 0.000 description 2
- 239000012623 DNA damaging agent Substances 0.000 description 2
- OKKJLVBELUTLKV-MZCSYVLQSA-N Deuterated methanol Chemical compound [2H]OC([2H])([2H])[2H] OKKJLVBELUTLKV-MZCSYVLQSA-N 0.000 description 2
- KRHYYFGTRYWZRS-UHFFFAOYSA-N Fluorane Chemical compound F KRHYYFGTRYWZRS-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
- 208000031448 Genomic Instability Diseases 0.000 description 2
- AEMRFAOFKBGASW-UHFFFAOYSA-N Glycolic acid Chemical compound OCC(O)=O AEMRFAOFKBGASW-UHFFFAOYSA-N 0.000 description 2
- 239000007821 HATU Substances 0.000 description 2
- 101001092185 Homo sapiens Regulator of cell cycle RGCC Proteins 0.000 description 2
- XEEYBQQBJWHFJM-UHFFFAOYSA-N Iron Chemical compound [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 2
- 125000002842 L-seryl group Chemical group O=C([*])[C@](N([H])[H])([H])C([H])([H])O[H] 0.000 description 2
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 2
- MBBZMMPHUWSWHV-BDVNFPICSA-N N-methylglucamine Chemical compound CNC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO MBBZMMPHUWSWHV-BDVNFPICSA-N 0.000 description 2
- UFWIBTONFRDIAS-UHFFFAOYSA-N Naphthalene Chemical group C1=CC=CC2=CC=CC=C21 UFWIBTONFRDIAS-UHFFFAOYSA-N 0.000 description 2
- SRAVBQRYVDKPEU-UHFFFAOYSA-N O=C(C(C(C1)=C2)=CC=C2N(CC2)CCC22OCCO2)N1C(CCC(N1)=O)C1=O Chemical compound O=C(C(C(C1)=C2)=CC=C2N(CC2)CCC22OCCO2)N1C(CCC(N1)=O)C1=O SRAVBQRYVDKPEU-UHFFFAOYSA-N 0.000 description 2
- GLUUGHFHXGJENI-UHFFFAOYSA-N Piperazine Chemical compound C1CNCCN1 GLUUGHFHXGJENI-UHFFFAOYSA-N 0.000 description 2
- NQRYJNQNLNOLGT-UHFFFAOYSA-N Piperidine Chemical compound C1CCNCC1 NQRYJNQNLNOLGT-UHFFFAOYSA-N 0.000 description 2
- 102000004245 Proteasome Endopeptidase Complex Human genes 0.000 description 2
- 108090000708 Proteasome Endopeptidase Complex Proteins 0.000 description 2
- 102000001253 Protein Kinase Human genes 0.000 description 2
- LCTONWCANYUPML-UHFFFAOYSA-N Pyruvic acid Chemical compound CC(=O)C(O)=O LCTONWCANYUPML-UHFFFAOYSA-N 0.000 description 2
- NPXOKRUENSOPAO-UHFFFAOYSA-N Raney nickel Chemical compound [Al].[Ni] NPXOKRUENSOPAO-UHFFFAOYSA-N 0.000 description 2
- 102100035542 Regulator of cell cycle RGCC Human genes 0.000 description 2
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 2
- YZCKVEUIGOORGS-NJFSPNSNSA-N Tritium Chemical compound [3H] YZCKVEUIGOORGS-NJFSPNSNSA-N 0.000 description 2
- 229940126117 ZN-c3 Drugs 0.000 description 2
- DJKPUSPBJFCEMA-UHFFFAOYSA-N [1-(4-nitrophenyl)piperidin-4-yl]methanol Chemical compound C1CC(CO)CCN1C1=CC=C([N+]([O-])=O)C=C1 DJKPUSPBJFCEMA-UHFFFAOYSA-N 0.000 description 2
- 125000000218 acetic acid group Chemical group C(C)(=O)* 0.000 description 2
- 229910052784 alkaline earth metal Inorganic materials 0.000 description 2
- IYABWNGZIDDRAK-UHFFFAOYSA-N allene Chemical group C=C=C IYABWNGZIDDRAK-UHFFFAOYSA-N 0.000 description 2
- 239000013011 aqueous formulation Substances 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- 125000000499 benzofuranyl group Chemical group O1C(=CC2=C1C=CC=C2)* 0.000 description 2
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 2
- 125000002619 bicyclic group Chemical group 0.000 description 2
- RYYVLZVUVIJVGH-UHFFFAOYSA-N caffeine Chemical compound CN1C(=O)N(C)C(=O)C2=C1N=CN2C RYYVLZVUVIJVGH-UHFFFAOYSA-N 0.000 description 2
- 230000012820 cell cycle checkpoint Effects 0.000 description 2
- 230000006369 cell cycle progression Effects 0.000 description 2
- 230000002950 deficient Effects 0.000 description 2
- NKLCNNUWBJBICK-UHFFFAOYSA-N dess–martin periodinane Chemical compound C1=CC=C2I(OC(=O)C)(OC(C)=O)(OC(C)=O)OC(=O)C2=C1 NKLCNNUWBJBICK-UHFFFAOYSA-N 0.000 description 2
- 208000035475 disorder Diseases 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 230000008029 eradication Effects 0.000 description 2
- 239000007789 gas Substances 0.000 description 2
- 102000045973 human WEE1 Human genes 0.000 description 2
- 125000004356 hydroxy functional group Chemical group O* 0.000 description 2
- 125000002632 imidazolidinyl group Chemical group 0.000 description 2
- 125000002636 imidazolinyl group Chemical group 0.000 description 2
- 238000010348 incorporation Methods 0.000 description 2
- 125000003453 indazolyl group Chemical group N1N=C(C2=C1C=CC=C2)* 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 150000007529 inorganic bases Chemical class 0.000 description 2
- 238000011068 loading method Methods 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- OKKJLVBELUTLKV-VMNATFBRSA-N methanol-d1 Chemical compound [2H]OC OKKJLVBELUTLKV-VMNATFBRSA-N 0.000 description 2
- 230000011278 mitosis Effects 0.000 description 2
- 230000006618 mitotic catastrophe Effects 0.000 description 2
- UHOVQNZJYSORNB-UHFFFAOYSA-N monobenzene Natural products C1=CC=CC=C1 UHOVQNZJYSORNB-UHFFFAOYSA-N 0.000 description 2
- 125000002950 monocyclic group Chemical group 0.000 description 2
- 238000010534 nucleophilic substitution reaction Methods 0.000 description 2
- 150000007530 organic bases Chemical class 0.000 description 2
- 229910052763 palladium Inorganic materials 0.000 description 2
- PIBWKRNGBLPSSY-UHFFFAOYSA-L palladium(II) chloride Chemical compound Cl[Pd]Cl PIBWKRNGBLPSSY-UHFFFAOYSA-L 0.000 description 2
- XBXHCBLBYQEYTI-UHFFFAOYSA-N piperidin-4-ylmethanol Chemical compound OCC1CCNCC1 XBXHCBLBYQEYTI-UHFFFAOYSA-N 0.000 description 2
- IVRIRQXJSNCSPQ-UHFFFAOYSA-N propan-2-yl carbonochloridate Chemical compound CC(C)OC(Cl)=O IVRIRQXJSNCSPQ-UHFFFAOYSA-N 0.000 description 2
- 108060006633 protein kinase Proteins 0.000 description 2
- 125000003072 pyrazolidinyl group Chemical group 0.000 description 2
- 125000000168 pyrrolyl group Chemical group 0.000 description 2
- YGSDEFSMJLZEOE-UHFFFAOYSA-N salicylic acid Chemical compound OC(=O)C1=CC=CC=C1O YGSDEFSMJLZEOE-UHFFFAOYSA-N 0.000 description 2
- 150000003384 small molecules Chemical class 0.000 description 2
- 235000017557 sodium bicarbonate Nutrition 0.000 description 2
- 238000003756 stirring Methods 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- WTAXAAQWUPUKGQ-UHFFFAOYSA-N tert-butyl 2-[2-(2,6-dioxopiperidin-3-yl)-1-oxo-3H-isoindol-5-yl]-2,7-diazaspiro[3.5]nonane-7-carboxylate Chemical compound CC(C)(C)OC(=O)N1CCC2(CN(C2)C2=CC3=C(C=C2)C(=O)N(C3)C2CCC(=O)NC2=O)CC1 WTAXAAQWUPUKGQ-UHFFFAOYSA-N 0.000 description 2
- SVMVJWMLQLOPRF-UHFFFAOYSA-N tert-butyl 4-(3-methoxycarbonylcyclobutyl)piperazine-1-carboxylate Chemical compound CC(C)(C)OC(=O)N1CCN(CC1)C2CC(C2)C(=O)OC SVMVJWMLQLOPRF-UHFFFAOYSA-N 0.000 description 2
- YRLQFRXDWBFGMK-UHFFFAOYSA-N tert-butyl 4-(4-aminophenyl)piperidine-1-carboxylate Chemical compound C1CN(C(=O)OC(C)(C)C)CCC1C1=CC=C(N)C=C1 YRLQFRXDWBFGMK-UHFFFAOYSA-N 0.000 description 2
- DSBDTGZARKKWJQ-UHFFFAOYSA-N tert-butyl 4-(4-formylpyrazol-1-yl)piperidine-1-carboxylate Chemical compound C1CN(C(=O)OC(C)(C)C)CCC1N1N=CC(C=O)=C1 DSBDTGZARKKWJQ-UHFFFAOYSA-N 0.000 description 2
- PKYRLRVLXYAKPM-UHFFFAOYSA-N tert-butyl 4-(4-nitrophenyl)-3,6-dihydro-2h-pyridine-1-carboxylate Chemical compound C1N(C(=O)OC(C)(C)C)CCC(C=2C=CC(=CC=2)[N+]([O-])=O)=C1 PKYRLRVLXYAKPM-UHFFFAOYSA-N 0.000 description 2
- WOEQSXAIPTXOPY-UHFFFAOYSA-N tert-butyl 4-methylsulfonyloxypiperidine-1-carboxylate Chemical compound CC(C)(C)OC(=O)N1CCC(OS(C)(=O)=O)CC1 WOEQSXAIPTXOPY-UHFFFAOYSA-N 0.000 description 2
- ZRDIWHVGGRNECI-UHFFFAOYSA-N tert-butyl 9-(4-nitrophenyl)-3,9-diazaspiro[5.5]undecane-3-carboxylate Chemical compound C1CN(C(=O)OC(C)(C)C)CCC21CCN(C=1C=CC(=CC=1)[N+]([O-])=O)CC2 ZRDIWHVGGRNECI-UHFFFAOYSA-N 0.000 description 2
- AXUPGUGFMSYTMC-UHFFFAOYSA-N tert-butyl 9-formyl-3-azaspiro[5.5]undecane-3-carboxylate Chemical compound C1CN(C(=O)OC(C)(C)C)CCC11CCC(C=O)CC1 AXUPGUGFMSYTMC-UHFFFAOYSA-N 0.000 description 2
- CWXPZXBSDSIRCS-UHFFFAOYSA-N tert-butyl piperazine-1-carboxylate Chemical compound CC(C)(C)OC(=O)N1CCNCC1 CWXPZXBSDSIRCS-UHFFFAOYSA-N 0.000 description 2
- YAPQBXQYLJRXSA-UHFFFAOYSA-N theobromine Chemical compound CN1C(=O)NC(=O)C2=C1N=CN2C YAPQBXQYLJRXSA-UHFFFAOYSA-N 0.000 description 2
- 125000001544 thienyl group Chemical group 0.000 description 2
- 125000004568 thiomorpholinyl group Chemical group 0.000 description 2
- JOXIMZWYDAKGHI-UHFFFAOYSA-N toluene-4-sulfonic acid Chemical compound CC1=CC=C(S(O)(=O)=O)C=C1 JOXIMZWYDAKGHI-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
- GETQZCLCWQTVFV-UHFFFAOYSA-N trimethylamine Chemical compound CN(C)C GETQZCLCWQTVFV-UHFFFAOYSA-N 0.000 description 2
- 229910052722 tritium Inorganic materials 0.000 description 2
- 230000006663 ubiquitin-proteasome pathway Effects 0.000 description 2
- 125000000391 vinyl group Chemical group [H]C([*])=C([H])[H] 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
- 125000006732 (C1-C15) alkyl group Chemical group 0.000 description 1
- 125000006527 (C1-C5) alkyl group Chemical group 0.000 description 1
- 125000006833 (C1-C5) alkylene group Chemical group 0.000 description 1
- 125000004209 (C1-C8) alkyl group Chemical group 0.000 description 1
- 125000006313 (C5-C8) alkyl group Chemical group 0.000 description 1
- WBYWAXJHAXSJNI-VOTSOKGWSA-M .beta-Phenylacrylic acid Natural products [O-]C(=O)\C=C\C1=CC=CC=C1 WBYWAXJHAXSJNI-VOTSOKGWSA-M 0.000 description 1
- 125000005988 1,1-dioxo-thiomorpholinyl group Chemical group 0.000 description 1
- 125000005871 1,3-benzodioxolyl group Chemical group 0.000 description 1
- 125000005877 1,4-benzodioxanyl group Chemical group 0.000 description 1
- CTQSHBBTVXALEN-UHFFFAOYSA-N 1,4-diazaspiro[2.2]pentane Chemical compound C1NC11NC1 CTQSHBBTVXALEN-UHFFFAOYSA-N 0.000 description 1
- KPKNTUUIEVXMOH-UHFFFAOYSA-N 1,4-dioxa-8-azaspiro[4.5]decane Chemical compound O1CCOC11CCNCC1 KPKNTUUIEVXMOH-UHFFFAOYSA-N 0.000 description 1
- HRLDFIUASRWJKK-UHFFFAOYSA-N 1,6-diazaspiro[2.4]heptane Chemical compound C1NC11CNCC1 HRLDFIUASRWJKK-UHFFFAOYSA-N 0.000 description 1
- KUYMEVLKUJVHKA-UHFFFAOYSA-N 1,6-diazaspiro[2.5]octane Chemical compound C1NC11CCNCC1 KUYMEVLKUJVHKA-UHFFFAOYSA-N 0.000 description 1
- UCSGBBSVOZFICK-UHFFFAOYSA-N 1,7-diazaspiro[2.6]nonane Chemical compound C1NC11CCNCCC1 UCSGBBSVOZFICK-UHFFFAOYSA-N 0.000 description 1
- VXNZUUAINFGPBY-UHFFFAOYSA-N 1-Butene Chemical group CCC=C VXNZUUAINFGPBY-UHFFFAOYSA-N 0.000 description 1
- AAYCHKCUROMSRP-LJQANCHMSA-N 1-[(7R)-7-ethyl-7-hydroxy-5,6-dihydrocyclopenta[b]pyridin-2-yl]-6-methylsulfanyl-2-prop-2-enylpyrazolo[3,4-d]pyrimidin-3-one Chemical compound C1=C2C(=NC(=C1)N1C3=NC(SC)=NC=C3C(=O)N1CC=C)[C@@](CC)(CC2)O AAYCHKCUROMSRP-LJQANCHMSA-N 0.000 description 1
- QQUFDFQHIUKUFP-UHFFFAOYSA-N 1-azaspiro[2.2]pentane Chemical compound C1CC11NC1 QQUFDFQHIUKUFP-UHFFFAOYSA-N 0.000 description 1
- IJBXVKITOJFLBF-UHFFFAOYSA-N 1-azaspiro[2.3]hexane Chemical compound C1NC11CCC1 IJBXVKITOJFLBF-UHFFFAOYSA-N 0.000 description 1
- IOOAVHIKEJAKPT-UHFFFAOYSA-N 1-azaspiro[2.4]heptane Chemical compound C1NC11CCCC1 IOOAVHIKEJAKPT-UHFFFAOYSA-N 0.000 description 1
- CBLWNEBYJOYFFX-UHFFFAOYSA-N 1-azaspiro[2.5]octane Chemical compound C1NC11CCCCC1 CBLWNEBYJOYFFX-UHFFFAOYSA-N 0.000 description 1
- NKRLGTMHRAQKAT-UHFFFAOYSA-N 1-azaspiro[2.6]nonane Chemical compound N1CC11CCCCCC1 NKRLGTMHRAQKAT-UHFFFAOYSA-N 0.000 description 1
- ZDFBKZUDCQQKAC-UHFFFAOYSA-N 1-bromo-4-nitrobenzene Chemical compound [O-][N+](=O)C1=CC=C(Br)C=C1 ZDFBKZUDCQQKAC-UHFFFAOYSA-N 0.000 description 1
- 125000004973 1-butenyl group Chemical group C(=CCC)* 0.000 description 1
- 125000005987 1-oxo-thiomorpholinyl group Chemical group 0.000 description 1
- LRGBDJBDJXZTTD-UHFFFAOYSA-N 1h-pyrazole-4-carbaldehyde Chemical compound O=CC=1C=NNC=1 LRGBDJBDJXZTTD-UHFFFAOYSA-N 0.000 description 1
- 125000004206 2,2,2-trifluoroethyl group Chemical group [H]C([H])(*)C(F)(F)F 0.000 description 1
- HCSBTDBGTNZOAB-UHFFFAOYSA-N 2,3-dinitrobenzoic acid Chemical class OC(=O)C1=CC=CC([N+]([O-])=O)=C1[N+]([O-])=O HCSBTDBGTNZOAB-UHFFFAOYSA-N 0.000 description 1
- UDSAJFSYJMHNFI-UHFFFAOYSA-N 2,6-diazaspiro[3.3]heptane Chemical compound C1NCC11CNC1 UDSAJFSYJMHNFI-UHFFFAOYSA-N 0.000 description 1
- OXPIFHNJGOJJQZ-UHFFFAOYSA-N 2,7-diazaspiro[3.4]octane Chemical compound C1NCC11CNCC1 OXPIFHNJGOJJQZ-UHFFFAOYSA-N 0.000 description 1
- DROZYMFJWSYDRY-UHFFFAOYSA-N 2,7-diazaspiro[3.5]nonane Chemical compound C1NCC11CCNCC1 DROZYMFJWSYDRY-UHFFFAOYSA-N 0.000 description 1
- DDVRNOMZDQTUNS-UHFFFAOYSA-N 2,7-diazaspiro[4.4]nonane Chemical compound C1NCCC11CNCC1 DDVRNOMZDQTUNS-UHFFFAOYSA-N 0.000 description 1
- OMGQHKZFOQEKMA-UHFFFAOYSA-N 2,8-diazaspiro[3.6]decane Chemical compound C1NCC11CCNCCC1 OMGQHKZFOQEKMA-UHFFFAOYSA-N 0.000 description 1
- WYZZNMWIWHRXRM-UHFFFAOYSA-N 2,8-diazaspiro[4.5]decane Chemical compound C1NCCC21CCNCC2 WYZZNMWIWHRXRM-UHFFFAOYSA-N 0.000 description 1
- UKBBHWZRMFFESL-UHFFFAOYSA-N 2,9-diazaspiro[4.6]undecane Chemical compound C1NCCC21CCNCCC2 UKBBHWZRMFFESL-UHFFFAOYSA-N 0.000 description 1
- MPQLCQKBYRSPNA-UHFFFAOYSA-N 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindole-1,3-dione Chemical compound O=C1C2=CC(F)=CC=C2C(=O)N1C1CCC(=O)NC1=O MPQLCQKBYRSPNA-UHFFFAOYSA-N 0.000 description 1
- GZNLORFTQXVXFE-UHFFFAOYSA-N 2-(2,6-dioxopiperidin-3-yl)-5-piperazin-1-ylisoindole-1,3-dione hydrochloride Chemical compound Cl.O=C1N(C2CCC(=O)NC2=O)C(=O)c2cc(ccc12)N1CCNCC1 GZNLORFTQXVXFE-UHFFFAOYSA-N 0.000 description 1
- CGBNTOUUTCHHQO-UHFFFAOYSA-N 2-[(2-methylpropan-2-yl)oxycarbonyl]-2-azaspiro[3.5]nonane-7-carboxylic acid Chemical compound C1N(C(=O)OC(C)(C)C)CC21CCC(C(O)=O)CC2 CGBNTOUUTCHHQO-UHFFFAOYSA-N 0.000 description 1
- ZYHQGITXIJDDKC-UHFFFAOYSA-N 2-[2-(2-aminophenyl)ethyl]aniline Chemical group NC1=CC=CC=C1CCC1=CC=CC=C1N ZYHQGITXIJDDKC-UHFFFAOYSA-N 0.000 description 1
- MSWZFWKMSRAUBD-IVMDWMLBSA-N 2-amino-2-deoxy-D-glucopyranose Chemical compound N[C@H]1C(O)O[C@H](CO)[C@@H](O)[C@@H]1O MSWZFWKMSRAUBD-IVMDWMLBSA-N 0.000 description 1
- QPEJAHMNOVMSOZ-UHFFFAOYSA-N 2-azaspiro[3.3]heptane Chemical compound C1CCC21CNC2 QPEJAHMNOVMSOZ-UHFFFAOYSA-N 0.000 description 1
- PSNDWZOXFDKLLH-UHFFFAOYSA-N 2-azaspiro[3.4]octane Chemical compound C1NCC11CCCC1 PSNDWZOXFDKLLH-UHFFFAOYSA-N 0.000 description 1
- IBODJKKYTBNWTD-UHFFFAOYSA-N 2-azaspiro[3.5]nonane Chemical compound C1NCC11CCCCC1 IBODJKKYTBNWTD-UHFFFAOYSA-N 0.000 description 1
- CDZMBPQIVAKQMT-UHFFFAOYSA-N 2-azaspiro[3.6]decane Chemical compound C1NCC11CCCCCC1 CDZMBPQIVAKQMT-UHFFFAOYSA-N 0.000 description 1
- NINJAJLCZUYDGV-UHFFFAOYSA-N 2-azaspiro[4.4]nonane Chemical compound C1CCCC21CNCC2 NINJAJLCZUYDGV-UHFFFAOYSA-N 0.000 description 1
- SFCOKGCNIPSUQF-UHFFFAOYSA-N 2-azaspiro[4.5]decane Chemical compound C1NCCC21CCCCC2 SFCOKGCNIPSUQF-UHFFFAOYSA-N 0.000 description 1
- UYUUEEXDJGLCLY-UHFFFAOYSA-N 2-azaspiro[4.6]undecane Chemical compound C1NCCC21CCCCCC2 UYUUEEXDJGLCLY-UHFFFAOYSA-N 0.000 description 1
- LILXDMFJXYAKMK-UHFFFAOYSA-N 2-bromo-1,1-diethoxyethane Chemical compound CCOC(CBr)OCC LILXDMFJXYAKMK-UHFFFAOYSA-N 0.000 description 1
- LDLCZOVUSADOIV-UHFFFAOYSA-N 2-bromoethanol Chemical compound OCCBr LDLCZOVUSADOIV-UHFFFAOYSA-N 0.000 description 1
- IKCLCGXPQILATA-UHFFFAOYSA-N 2-chlorobenzoic acid Chemical class OC(=O)C1=CC=CC=C1Cl IKCLCGXPQILATA-UHFFFAOYSA-N 0.000 description 1
- BFSVOASYOCHEOV-UHFFFAOYSA-N 2-diethylaminoethanol Chemical compound CCN(CC)CCO BFSVOASYOCHEOV-UHFFFAOYSA-N 0.000 description 1
- 229940013085 2-diethylaminoethanol Drugs 0.000 description 1
- WJZIPMQUKSTHLV-UHFFFAOYSA-N 2-ethyldecanoic acid Chemical compound CCCCCCCCC(CC)C(O)=O WJZIPMQUKSTHLV-UHFFFAOYSA-N 0.000 description 1
- 125000006088 2-oxoazepinyl group Chemical group 0.000 description 1
- 125000004638 2-oxopiperazinyl group Chemical group O=C1N(CCNC1)* 0.000 description 1
- 125000004637 2-oxopiperidinyl group Chemical group O=C1N(CCCC1)* 0.000 description 1
- 125000003903 2-propenyl group Chemical group [H]C([*])([H])C([H])=C([H])[H] 0.000 description 1
- 125000004485 2-pyrrolidinyl group Chemical group [H]N1C([H])([H])C([H])([H])C([H])([H])C1([H])* 0.000 description 1
- HETIDEGYTRXKCE-UHFFFAOYSA-N 3,10-diazaspiro[5.6]dodecane Chemical compound C1CNCCC21CCNCCC2 HETIDEGYTRXKCE-UHFFFAOYSA-N 0.000 description 1
- YNKVCLQNSSTHTD-UHFFFAOYSA-N 3,9-diazaspiro[5.5]undecane Chemical compound C1CNCCC21CCNCC2 YNKVCLQNSSTHTD-UHFFFAOYSA-N 0.000 description 1
- MOUYUPAFHMXXRV-UHFFFAOYSA-N 3-[3-oxo-6-(4-oxopiperidin-1-yl)-1H-isoindol-2-yl]piperidine-2,6-dione Chemical compound O=C(C(C(C1)=C2)=CC=C2N(CC2)CCC2=O)N1C(CCC(N1)=O)C1=O MOUYUPAFHMXXRV-UHFFFAOYSA-N 0.000 description 1
- BMYNFMYTOJXKLE-UHFFFAOYSA-N 3-azaniumyl-2-hydroxypropanoate Chemical compound NCC(O)C(O)=O BMYNFMYTOJXKLE-UHFFFAOYSA-N 0.000 description 1
- LIZKZVQBLDHKCY-UHFFFAOYSA-N 3-azaspiro[5.5]undecane Chemical compound C1CCCCC21CCNCC2 LIZKZVQBLDHKCY-UHFFFAOYSA-N 0.000 description 1
- MFHUQHWMLMXUAH-UHFFFAOYSA-N 3-azaspiro[5.6]dodecane Chemical compound C1CNCCC21CCCCCC2 MFHUQHWMLMXUAH-UHFFFAOYSA-N 0.000 description 1
- 125000004575 3-pyrrolidinyl group Chemical group [H]N1C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- NRVYHSVBQNFNAX-UHFFFAOYSA-N 4-piperidin-4-ylaniline Chemical compound C1=CC(N)=CC=C1C1CCNCC1 NRVYHSVBQNFNAX-UHFFFAOYSA-N 0.000 description 1
- 125000005986 4-piperidonyl group Chemical group 0.000 description 1
- RHVACWSWOHDPBX-UHFFFAOYSA-N 5-azaspiro[2.3]hexane Chemical compound C1CC11CNC1 RHVACWSWOHDPBX-UHFFFAOYSA-N 0.000 description 1
- HOXWESKAHYABGO-UHFFFAOYSA-N 5-azaspiro[2.4]heptane Chemical compound C1CC11CNCC1 HOXWESKAHYABGO-UHFFFAOYSA-N 0.000 description 1
- MWMSBQXTHIEBNT-UHFFFAOYSA-N 5-bromo-2-(2,6-dioxopiperidin-3-yl)isoindole-1,3-dione Chemical compound O=C1C2=CC(Br)=CC=C2C(=O)N1C1CCC(=O)NC1=O MWMSBQXTHIEBNT-UHFFFAOYSA-N 0.000 description 1
- GIBPTWPJEVCTGR-UHFFFAOYSA-N 6-azaspiro[2.5]octane Chemical compound C1CC11CCNCC1 GIBPTWPJEVCTGR-UHFFFAOYSA-N 0.000 description 1
- SUKDJGHHYOXCIW-UHFFFAOYSA-N 6-azaspiro[2.6]nonane Chemical compound C1CC11CCNCCC1 SUKDJGHHYOXCIW-UHFFFAOYSA-N 0.000 description 1
- NVZWMJWMTWOVNJ-UHFFFAOYSA-N 6-azaspiro[3.4]octane Chemical compound C1CCC21CNCC2 NVZWMJWMTWOVNJ-UHFFFAOYSA-N 0.000 description 1
- BSQKGAVROUDOTE-UHFFFAOYSA-N 7-azaspiro[3.5]nonane Chemical compound C1CCC21CCNCC2 BSQKGAVROUDOTE-UHFFFAOYSA-N 0.000 description 1
- VCSWIDPMTFSITK-UHFFFAOYSA-N 7-azaspiro[3.6]decane Chemical compound C1CCC21CCNCCC2 VCSWIDPMTFSITK-UHFFFAOYSA-N 0.000 description 1
- HBAQYPYDRFILMT-UHFFFAOYSA-N 8-[3-(1-cyclopropylpyrazol-4-yl)-1H-pyrazolo[4,3-d]pyrimidin-5-yl]-3-methyl-3,8-diazabicyclo[3.2.1]octan-2-one Chemical class C1(CC1)N1N=CC(=C1)C1=NNC2=C1N=C(N=C2)N1C2C(N(CC1CC2)C)=O HBAQYPYDRFILMT-UHFFFAOYSA-N 0.000 description 1
- XAJANCPFKPMWGA-UHFFFAOYSA-N 8-azaspiro[4.6]undecane Chemical compound C1CCCC21CCNCCC2 XAJANCPFKPMWGA-UHFFFAOYSA-N 0.000 description 1
- ZWAATVZJDIHKSZ-UHFFFAOYSA-N 9-azaspiro[5.6]dodecane Chemical compound C1CCCCC21CCNCCC2 ZWAATVZJDIHKSZ-UHFFFAOYSA-N 0.000 description 1
- HRPVXLWXLXDGHG-UHFFFAOYSA-N Acrylamide Chemical compound NC(=O)C=C HRPVXLWXLXDGHG-UHFFFAOYSA-N 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-N Ammonia Chemical compound N QGZKDVFQNNGYKY-UHFFFAOYSA-N 0.000 description 1
- NLXLAEXVIDQMFP-UHFFFAOYSA-N Ammonia chloride Chemical compound [NH4+].[Cl-] NLXLAEXVIDQMFP-UHFFFAOYSA-N 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 239000005711 Benzoic acid Substances 0.000 description 1
- KWIUHFFTVRNATP-UHFFFAOYSA-N Betaine Natural products C[N+](C)(C)CC([O-])=O KWIUHFFTVRNATP-UHFFFAOYSA-N 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- NMQGAXWXHWLWBP-UHFFFAOYSA-N C1NC11CNC1 Chemical compound C1NC11CNC1 NMQGAXWXHWLWBP-UHFFFAOYSA-N 0.000 description 1
- 102000009728 CDC2 Protein Kinase Human genes 0.000 description 1
- 108010034798 CDC2 Protein Kinase Proteins 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 239000004215 Carbon black (E152) Substances 0.000 description 1
- OKTJSMMVPCPJKN-NJFSPNSNSA-N Carbon-14 Chemical compound [14C] OKTJSMMVPCPJKN-NJFSPNSNSA-N 0.000 description 1
- 102100034357 Casein kinase I isoform alpha Human genes 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- WBYWAXJHAXSJNI-SREVYHEPSA-N Cinnamic acid Chemical compound OC(=O)\C=C/C1=CC=CC=C1 WBYWAXJHAXSJNI-SREVYHEPSA-N 0.000 description 1
- RYGMFSIKBFXOCR-UHFFFAOYSA-N Copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 1
- OCUCCJIRFHNWBP-IYEMJOQQSA-L Copper gluconate Chemical class [Cu+2].OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C([O-])=O.OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C([O-])=O OCUCCJIRFHNWBP-IYEMJOQQSA-L 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- 230000006820 DNA synthesis Effects 0.000 description 1
- 238000011346 DNA-damaging therapy Methods 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
- XBPCUCUWBYBCDP-UHFFFAOYSA-N Dicyclohexylamine Chemical compound C1CCCCC1NC1CCCCC1 XBPCUCUWBYBCDP-UHFFFAOYSA-N 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- PIICEJLVQHRZGT-UHFFFAOYSA-N Ethylenediamine Chemical compound NCCN PIICEJLVQHRZGT-UHFFFAOYSA-N 0.000 description 1
- 102100036816 Eukaryotic peptide chain release factor GTP-binding subunit ERF3A Human genes 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- BDAGIHXWWSANSR-UHFFFAOYSA-N Formic acid Chemical compound OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 241001272567 Hominoidea Species 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000994700 Homo sapiens Casein kinase I isoform alpha Proteins 0.000 description 1
- 101000851788 Homo sapiens Eukaryotic peptide chain release factor GTP-binding subunit ERF3A Proteins 0.000 description 1
- 101000610640 Homo sapiens U4/U6 small nuclear ribonucleoprotein Prp3 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
- LPHGQDQBBGAPDZ-UHFFFAOYSA-N Isocaffeine Natural products CN1C(=O)N(C)C(=O)C2=C1N(C)C=N2 LPHGQDQBBGAPDZ-UHFFFAOYSA-N 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-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
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 229910010084 LiAlH4 Inorganic materials 0.000 description 1
- WHXSMMKQMYFTQS-UHFFFAOYSA-N Lithium Chemical compound [Li] WHXSMMKQMYFTQS-UHFFFAOYSA-N 0.000 description 1
- 239000012448 Lithium borohydride Substances 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- KWIUHFFTVRNATP-UHFFFAOYSA-O N,N,N-trimethylglycinium Chemical compound C[N+](C)(C)CC(O)=O KWIUHFFTVRNATP-UHFFFAOYSA-O 0.000 description 1
- UEEJHVSXFDXPFK-UHFFFAOYSA-N N-dimethylaminoethanol Chemical compound CN(C)CCO UEEJHVSXFDXPFK-UHFFFAOYSA-N 0.000 description 1
- HTLZVHNRZJPSMI-UHFFFAOYSA-N N-ethylpiperidine Chemical compound CCN1CCCCC1 HTLZVHNRZJPSMI-UHFFFAOYSA-N 0.000 description 1
- GRYLNZFGIOXLOG-UHFFFAOYSA-N Nitric acid Chemical compound O[N+]([O-])=O GRYLNZFGIOXLOG-UHFFFAOYSA-N 0.000 description 1
- JCXJVPUVTGWSNB-UHFFFAOYSA-N Nitrogen dioxide Chemical compound O=[N]=O JCXJVPUVTGWSNB-UHFFFAOYSA-N 0.000 description 1
- HFZAEKGTOHBSMJ-UHFFFAOYSA-N OCC(CC1)CCN1C1=CC=C(CN(C(CCC(N2)=O)C2=O)C2=O)C2=C1 Chemical compound OCC(CC1)CCN1C1=CC=C(CN(C(CCC(N2)=O)C2=O)C2=O)C2=C1 HFZAEKGTOHBSMJ-UHFFFAOYSA-N 0.000 description 1
- 102100027069 Odontogenic ameloblast-associated protein Human genes 0.000 description 1
- 101710091533 Odontogenic ameloblast-associated protein Proteins 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 241000282579 Pan Species 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 244000025272 Persea americana Species 0.000 description 1
- 235000008673 Persea americana Nutrition 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 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
- 241000700159 Rattus Species 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 101001110823 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) 60S ribosomal protein L6-A Proteins 0.000 description 1
- 101000712176 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) 60S ribosomal protein L6-B Proteins 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- KDYFGRWQOYBRFD-UHFFFAOYSA-N Succinic acid Natural products OC(=O)CCC(O)=O KDYFGRWQOYBRFD-UHFFFAOYSA-N 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- LSNNMFCWUKXFEE-UHFFFAOYSA-N Sulfurous acid Chemical class OS(O)=O LSNNMFCWUKXFEE-UHFFFAOYSA-N 0.000 description 1
- 241000282898 Sus scrofa Species 0.000 description 1
- STSCVKRWJPWALQ-UHFFFAOYSA-N TRIFLUOROACETIC ACID ETHYL ESTER Chemical compound CCOC(=O)C(F)(F)F STSCVKRWJPWALQ-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
- 241000906446 Theraps Species 0.000 description 1
- 102100040374 U4/U6 small nuclear ribonucleoprotein Prp3 Human genes 0.000 description 1
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 description 1
- QOTUIIJRVXKSJU-RXMQYKEDSA-N [(3r)-pyrrolidin-3-yl]methanol Chemical compound OC[C@@H]1CCNC1 QOTUIIJRVXKSJU-RXMQYKEDSA-N 0.000 description 1
- IUHFWCGCSVTMPG-UHFFFAOYSA-N [C].[C] Chemical class [C].[C] IUHFWCGCSVTMPG-UHFFFAOYSA-N 0.000 description 1
- 150000001242 acetic acid derivatives Chemical class 0.000 description 1
- 125000000641 acridinyl group Chemical group C1(=CC=CC2=NC3=CC=CC=C3C=C12)* 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 125000002015 acyclic group Chemical group 0.000 description 1
- 125000005073 adamantyl group Chemical group C12(CC3CC(CC(C1)C3)C2)* 0.000 description 1
- 125000003158 alcohol group Chemical group 0.000 description 1
- 125000001931 aliphatic group Chemical group 0.000 description 1
- 239000003513 alkali Substances 0.000 description 1
- 150000001342 alkaline earth metals Chemical class 0.000 description 1
- HSFWRNGVRCDJHI-UHFFFAOYSA-N alpha-acetylene Natural products C#C HSFWRNGVRCDJHI-UHFFFAOYSA-N 0.000 description 1
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 1
- 150000001413 amino acids Chemical class 0.000 description 1
- 235000012538 ammonium bicarbonate Nutrition 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 229960003121 arginine Drugs 0.000 description 1
- 159000000032 aromatic acids Chemical class 0.000 description 1
- 150000005840 aryl radicals Chemical class 0.000 description 1
- AXMNGEUJXLXFRY-UHFFFAOYSA-N azaspirodecane Chemical compound C1CCCC21CCNCC2 AXMNGEUJXLXFRY-UHFFFAOYSA-N 0.000 description 1
- 125000002785 azepinyl group Chemical group 0.000 description 1
- 150000007514 bases Chemical class 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- SRSXLGNVWSONIS-UHFFFAOYSA-N benzenesulfonic acid Chemical class OS(=O)(=O)C1=CC=CC=C1 SRSXLGNVWSONIS-UHFFFAOYSA-N 0.000 description 1
- 125000003785 benzimidazolyl group Chemical group N1=C(NC2=C1C=CC=C2)* 0.000 description 1
- 125000005870 benzindolyl group Chemical group 0.000 description 1
- 125000005605 benzo group Chemical group 0.000 description 1
- 125000005875 benzo[b][1,4]dioxepinyl group Chemical group 0.000 description 1
- 125000005876 benzo[b][1,4]oxazinyl group Chemical group 0.000 description 1
- 125000005873 benzo[d]thiazolyl group Chemical group 0.000 description 1
- 125000000928 benzodioxinyl group Chemical group O1C(=COC2=C1C=CC=C2)* 0.000 description 1
- 125000002047 benzodioxolyl group Chemical group O1OC(C2=C1C=CC=C2)* 0.000 description 1
- 235000010233 benzoic acid Nutrition 0.000 description 1
- 150000001558 benzoic acid derivatives Chemical class 0.000 description 1
- 125000005878 benzonaphthofuranyl group Chemical group 0.000 description 1
- 125000005872 benzooxazolyl group Chemical group 0.000 description 1
- 125000004619 benzopyranyl group Chemical group O1C(C=CC2=C1C=CC=C2)* 0.000 description 1
- 125000005874 benzothiadiazolyl group Chemical group 0.000 description 1
- 125000003354 benzotriazolyl group Chemical group N1N=NC2=C1C=CC=C2* 0.000 description 1
- 125000004541 benzoxazolyl group Chemical group O1C(=NC2=C1C=CC=C2)* 0.000 description 1
- MSWZFWKMSRAUBD-UHFFFAOYSA-N beta-D-galactosamine Natural products NC1C(O)OC(CO)C(O)C1O MSWZFWKMSRAUBD-UHFFFAOYSA-N 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- 229960003237 betaine Drugs 0.000 description 1
- BVCRERJDOOBZOH-UHFFFAOYSA-N bicyclo[2.2.1]heptanyl Chemical group C1C[C+]2CC[C-]1C2 BVCRERJDOOBZOH-UHFFFAOYSA-N 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 230000036983 biotransformation Effects 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-M bisulphate group Chemical group S([O-])(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-M 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 150000001649 bromium compounds Chemical class 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
- ILAJWURWJKXJPW-UHFFFAOYSA-N butanedioic acid;octanedioic acid Chemical class OC(=O)CCC(O)=O.OC(=O)CCCCCCC(O)=O ILAJWURWJKXJPW-UHFFFAOYSA-N 0.000 description 1
- 125000000480 butynyl group Chemical group [*]C#CC([H])([H])C([H])([H])[H] 0.000 description 1
- 239000006227 byproduct Substances 0.000 description 1
- 229960001948 caffeine Drugs 0.000 description 1
- VJEONQKOZGKCAK-UHFFFAOYSA-N caffeine Natural products CN1C(=O)N(C)C(=O)C2=C1C=CN2C VJEONQKOZGKCAK-UHFFFAOYSA-N 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 125000000609 carbazolyl group Chemical group C1(=CC=CC=2C3=CC=CC=C3NC12)* 0.000 description 1
- 125000002915 carbonyl group Chemical group [*:2]C([*:1])=O 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 239000003054 catalyst Substances 0.000 description 1
- 230000025084 cell cycle arrest Effects 0.000 description 1
- 230000023359 cell cycle switching, meiotic to mitotic cell cycle Effects 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 235000010980 cellulose Nutrition 0.000 description 1
- 150000001793 charged compounds Chemical class 0.000 description 1
- PBAYDYUZOSNJGU-UHFFFAOYSA-N chelidonic acid Natural products OC(=O)C1=CC(=O)C=C(C(O)=O)O1 PBAYDYUZOSNJGU-UHFFFAOYSA-N 0.000 description 1
- 150000001805 chlorine compounds Chemical class 0.000 description 1
- VDANGULDQQJODZ-UHFFFAOYSA-N chloroprocaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1Cl VDANGULDQQJODZ-UHFFFAOYSA-N 0.000 description 1
- 229960002023 chloroprocaine Drugs 0.000 description 1
- OEYIOHPDSNJKLS-UHFFFAOYSA-N choline Chemical compound C[N+](C)(C)CCO OEYIOHPDSNJKLS-UHFFFAOYSA-N 0.000 description 1
- 229960001231 choline Drugs 0.000 description 1
- 235000013985 cinnamic acid Nutrition 0.000 description 1
- 229930016911 cinnamic acid Natural products 0.000 description 1
- 125000000259 cinnolinyl group Chemical group N1=NC(=CC2=CC=CC=C12)* 0.000 description 1
- 235000015165 citric acid Nutrition 0.000 description 1
- 150000001860 citric acid derivatives Chemical class 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 229910052802 copper Inorganic materials 0.000 description 1
- 239000010949 copper Substances 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 125000000392 cycloalkenyl group Chemical group 0.000 description 1
- 125000001162 cycloheptenyl group Chemical group C1(=CCCCCC1)* 0.000 description 1
- 125000000582 cycloheptyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000000596 cyclohexenyl group Chemical group C1(=CCCCC1)* 0.000 description 1
- 125000000113 cyclohexyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000000522 cyclooctenyl group Chemical group C1(=CCCCCCC1)* 0.000 description 1
- 125000000640 cyclooctyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- 125000002433 cyclopentenyl group Chemical group C1(=CCCC1)* 0.000 description 1
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- DEZRYPDIMOWBDS-UHFFFAOYSA-N dcm dichloromethane Chemical compound ClCCl.ClCCl DEZRYPDIMOWBDS-UHFFFAOYSA-N 0.000 description 1
- 125000005507 decahydroisoquinolyl group Chemical group 0.000 description 1
- 125000004855 decalinyl group Chemical group C1(CCCC2CCCCC12)* 0.000 description 1
- 239000001064 degrader Substances 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 150000001975 deuterium Chemical group 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 125000005509 dibenzothiophenyl group Chemical group 0.000 description 1
- 150000001991 dicarboxylic acids Chemical class 0.000 description 1
- ZBCBWPMODOFKDW-UHFFFAOYSA-N diethanolamine Chemical compound OCCNCCO ZBCBWPMODOFKDW-UHFFFAOYSA-N 0.000 description 1
- HPNMFZURTQLUMO-UHFFFAOYSA-N diethylamine Chemical compound CCNCC HPNMFZURTQLUMO-UHFFFAOYSA-N 0.000 description 1
- 125000001028 difluoromethyl group Chemical group [H]C(F)(F)* 0.000 description 1
- UXGNZZKBCMGWAZ-UHFFFAOYSA-N dimethylformamide dmf Chemical compound CN(C)C=O.CN(C)C=O UXGNZZKBCMGWAZ-UHFFFAOYSA-N 0.000 description 1
- 125000005879 dioxolanyl group Chemical group 0.000 description 1
- 235000011180 diphosphates Nutrition 0.000 description 1
- CETRZFQIITUQQL-UHFFFAOYSA-N dmso dimethylsulfoxide Chemical compound CS(C)=O.CS(C)=O CETRZFQIITUQQL-UHFFFAOYSA-N 0.000 description 1
- 238000009509 drug development Methods 0.000 description 1
- 238000007876 drug discovery Methods 0.000 description 1
- 238000000132 electrospray ionisation Methods 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- CCIVGXIOQKPBKL-UHFFFAOYSA-M ethanesulfonate Chemical compound CCS([O-])(=O)=O CCIVGXIOQKPBKL-UHFFFAOYSA-M 0.000 description 1
- PQJJJMRNHATNKG-UHFFFAOYSA-N ethyl bromoacetate Chemical compound CCOC(=O)CBr PQJJJMRNHATNKG-UHFFFAOYSA-N 0.000 description 1
- 229940012017 ethylenediamine Drugs 0.000 description 1
- 125000002534 ethynyl group Chemical group [H]C#C* 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 230000001747 exhibiting effect Effects 0.000 description 1
- RWTNPBWLLIMQHL-UHFFFAOYSA-N fexofenadine Chemical group C1=CC(C(C)(C(O)=O)C)=CC=C1C(O)CCCN1CCC(C(O)(C=2C=CC=CC=2)C=2C=CC=CC=2)CC1 RWTNPBWLLIMQHL-UHFFFAOYSA-N 0.000 description 1
- RMBPEFMHABBEKP-UHFFFAOYSA-N fluorene Chemical group C1=CC=C2C3=C[CH]C=CC3=CC2=C1 RMBPEFMHABBEKP-UHFFFAOYSA-N 0.000 description 1
- 125000004216 fluoromethyl group Chemical group [H]C([H])(F)* 0.000 description 1
- 235000019253 formic acid Nutrition 0.000 description 1
- 239000012458 free base Substances 0.000 description 1
- VZCYOOQTPOCHFL-OWOJBTEDSA-L fumarate(2-) Chemical class [O-]C(=O)\C=C\C([O-])=O VZCYOOQTPOCHFL-OWOJBTEDSA-L 0.000 description 1
- 239000001530 fumaric acid Substances 0.000 description 1
- 235000011087 fumaric acid Nutrition 0.000 description 1
- 125000003844 furanonyl group Chemical group 0.000 description 1
- 125000002541 furyl group Chemical group 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 229960002442 glucosamine Drugs 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 235000001727 glucose Nutrition 0.000 description 1
- 125000000262 haloalkenyl group Chemical group 0.000 description 1
- 125000000232 haloalkynyl group Chemical group 0.000 description 1
- 230000003862 health status Effects 0.000 description 1
- 150000002390 heteroarenes Chemical class 0.000 description 1
- 125000005114 heteroarylalkoxy group Chemical group 0.000 description 1
- 125000000592 heterocycloalkyl group Chemical group 0.000 description 1
- VKYKSIONXSXAKP-UHFFFAOYSA-N hexamethylenetetramine Chemical compound C1N(C2)CN3CN1CN2C3 VKYKSIONXSXAKP-UHFFFAOYSA-N 0.000 description 1
- 125000005980 hexynyl group Chemical group 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 229960002885 histidine Drugs 0.000 description 1
- XGIHQYAWBCFNPY-AZOCGYLKSA-N hydrabamine Chemical compound C([C@@H]12)CC3=CC(C(C)C)=CC=C3[C@@]2(C)CCC[C@@]1(C)CNCCNC[C@@]1(C)[C@@H]2CCC3=CC(C(C)C)=CC=C3[C@@]2(C)CCC1 XGIHQYAWBCFNPY-AZOCGYLKSA-N 0.000 description 1
- 150000004677 hydrates Chemical class 0.000 description 1
- 125000000717 hydrazino group Chemical group [H]N([*])N([H])[H] 0.000 description 1
- 229930195733 hydrocarbon Natural products 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
- 125000002883 imidazolyl group Chemical group 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 125000003387 indolinyl group Chemical group N1(CCC2=CC=CC=C12)* 0.000 description 1
- 125000003406 indolizinyl group Chemical group C=1(C=CN2C=CC=CC12)* 0.000 description 1
- 125000001041 indolyl group Chemical group 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- XMBWDFGMSWQBCA-YPZZEJLDSA-N iodane Chemical compound [125IH] XMBWDFGMSWQBCA-YPZZEJLDSA-N 0.000 description 1
- 150000004694 iodide salts Chemical class 0.000 description 1
- 229940044173 iodine-125 Drugs 0.000 description 1
- 239000003456 ion exchange resin Substances 0.000 description 1
- 229920003303 ion-exchange polymer Polymers 0.000 description 1
- 229910052742 iron Inorganic materials 0.000 description 1
- NNPPMTNAJDCUHE-UHFFFAOYSA-N isobutane Chemical compound CC(C)C NNPPMTNAJDCUHE-UHFFFAOYSA-N 0.000 description 1
- KQNPFQTWMSNSAP-UHFFFAOYSA-N isobutyric acid Chemical class CC(C)C(O)=O KQNPFQTWMSNSAP-UHFFFAOYSA-N 0.000 description 1
- 125000004594 isoindolinyl group Chemical group C1(NCC2=CC=CC=C12)* 0.000 description 1
- 125000000904 isoindolyl group Chemical group C=1(NC=C2C=CC=CC12)* 0.000 description 1
- JJWLVOIRVHMVIS-UHFFFAOYSA-N isopropylamine Chemical compound CC(C)N JJWLVOIRVHMVIS-UHFFFAOYSA-N 0.000 description 1
- 125000002183 isoquinolinyl group Chemical group C1(=NC=CC2=CC=CC=C12)* 0.000 description 1
- 125000005956 isoquinolyl group Chemical group 0.000 description 1
- 125000004628 isothiazolidinyl group Chemical group S1N(CCC1)* 0.000 description 1
- 125000001786 isothiazolyl group Chemical group 0.000 description 1
- 125000003965 isoxazolidinyl group Chemical group 0.000 description 1
- 125000000842 isoxazolyl group Chemical group 0.000 description 1
- 150000003893 lactate salts Chemical class 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 229910052744 lithium Inorganic materials 0.000 description 1
- 239000012280 lithium aluminium hydride Substances 0.000 description 1
- 229960003646 lysine Drugs 0.000 description 1
- 230000007347 lysosomal proteolysis Effects 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- ZLNQQNXFFQJAID-UHFFFAOYSA-L magnesium carbonate Chemical compound [Mg+2].[O-]C([O-])=O ZLNQQNXFFQJAID-UHFFFAOYSA-L 0.000 description 1
- 239000001095 magnesium carbonate Substances 0.000 description 1
- 229910000021 magnesium carbonate Inorganic materials 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 230000014759 maintenance of location Effects 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
- 150000002688 maleic acid derivatives Chemical class 0.000 description 1
- 150000004701 malic acid derivatives Chemical class 0.000 description 1
- 150000002690 malonic acid derivatives Chemical class 0.000 description 1
- 229960002510 mandelic acid Drugs 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 150000002739 metals Chemical class 0.000 description 1
- 125000005341 metaphosphate group Chemical group 0.000 description 1
- AFVFQIVMOAPDHO-UHFFFAOYSA-M methanesulfonate group Chemical class CS(=O)(=O)[O-] AFVFQIVMOAPDHO-UHFFFAOYSA-M 0.000 description 1
- 229940098779 methanesulfonic acid Drugs 0.000 description 1
- IZDROVVXIHRYMH-UHFFFAOYSA-N methanesulfonic anhydride Chemical compound CS(=O)(=O)OS(C)(=O)=O IZDROVVXIHRYMH-UHFFFAOYSA-N 0.000 description 1
- SDMCZCALYDCRBH-UHFFFAOYSA-N methoxymethyl(triphenyl)phosphanium Chemical compound C=1C=CC=CC=1[P+](C=1C=CC=CC=1)(COC)C1=CC=CC=C1 SDMCZCALYDCRBH-UHFFFAOYSA-N 0.000 description 1
- GINLBXIFGQVBHL-UHFFFAOYSA-N methyl 2-(5-bromo-2-chlorophenyl)acetate Chemical compound COC(=O)CC1=CC(Br)=CC=C1Cl GINLBXIFGQVBHL-UHFFFAOYSA-N 0.000 description 1
- IHLHSAIBOSSHQV-UHFFFAOYSA-N methyl 3-oxocyclobutane-1-carboxylate Chemical compound COC(=O)C1CC(=O)C1 IHLHSAIBOSSHQV-UHFFFAOYSA-N 0.000 description 1
- QPJVMBTYPHYUOC-UHFFFAOYSA-N methyl benzoate Chemical class COC(=O)C1=CC=CC=C1 QPJVMBTYPHYUOC-UHFFFAOYSA-N 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- WBYWAXJHAXSJNI-UHFFFAOYSA-N methyl p-hydroxycinnamate Natural products OC(=O)C=CC1=CC=CC=C1 WBYWAXJHAXSJNI-UHFFFAOYSA-N 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 125000005322 morpholin-1-yl group Chemical group 0.000 description 1
- 125000004312 morpholin-2-yl group Chemical group [H]N1C([H])([H])C([H])([H])OC([H])(*)C1([H])[H] 0.000 description 1
- 125000002757 morpholinyl group Chemical group 0.000 description 1
- ACTNHJDHMQSOGL-UHFFFAOYSA-N n',n'-dibenzylethane-1,2-diamine Chemical compound C=1C=CC=CC=1CN(CCN)CC1=CC=CC=C1 ACTNHJDHMQSOGL-UHFFFAOYSA-N 0.000 description 1
- NOUUUQMKVOUUNR-UHFFFAOYSA-N n,n'-diphenylethane-1,2-diamine Chemical compound C=1C=CC=CC=1NCCNC1=CC=CC=C1 NOUUUQMKVOUUNR-UHFFFAOYSA-N 0.000 description 1
- 125000004593 naphthyridinyl group Chemical group N1=C(C=CC2=CC=CN=C12)* 0.000 description 1
- 150000002823 nitrates Chemical class 0.000 description 1
- 229910017604 nitric acid Inorganic materials 0.000 description 1
- 150000002825 nitriles Chemical class 0.000 description 1
- 229910000069 nitrogen hydride Inorganic materials 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 125000003518 norbornenyl group Chemical group C12(C=CC(CC1)C2)* 0.000 description 1
- 125000002868 norbornyl group Chemical group C12(CCC(CC1)C2)* 0.000 description 1
- VIKNJXKGJWUCNN-XGXHKTLJSA-N norethisterone Chemical compound O=C1CC[C@@H]2[C@H]3CC[C@](C)([C@](CC4)(O)C#C)[C@@H]4[C@@H]3CCC2=C1 VIKNJXKGJWUCNN-XGXHKTLJSA-N 0.000 description 1
- NIHNNTQXNPWCJQ-UHFFFAOYSA-N o-biphenylenemethane Chemical group C1=CC=C2CC3=CC=CC=C3C2=C1 NIHNNTQXNPWCJQ-UHFFFAOYSA-N 0.000 description 1
- 125000005060 octahydroindolyl group Chemical group N1(CCC2CCCCC12)* 0.000 description 1
- 125000005061 octahydroisoindolyl group Chemical group C1(NCC2CCCCC12)* 0.000 description 1
- WWZKQHOCKIZLMA-UHFFFAOYSA-M octanoate Chemical class CCCCCCCC([O-])=O WWZKQHOCKIZLMA-UHFFFAOYSA-M 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 235000019198 oils Nutrition 0.000 description 1
- 238000011275 oncology therapy Methods 0.000 description 1
- 239000006186 oral dosage form Substances 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 238000006053 organic reaction Methods 0.000 description 1
- 125000002524 organometallic group Chemical group 0.000 description 1
- 125000001715 oxadiazolyl group Chemical group 0.000 description 1
- 150000003891 oxalate salts Chemical class 0.000 description 1
- 235000006408 oxalic acid Nutrition 0.000 description 1
- 125000000160 oxazolidinyl group Chemical group 0.000 description 1
- 125000002971 oxazolyl group Chemical group 0.000 description 1
- 125000000466 oxiranyl group Chemical group 0.000 description 1
- 125000005476 oxopyrrolidinyl group Chemical group 0.000 description 1
- FJKROLUGYXJWQN-UHFFFAOYSA-N papa-hydroxy-benzoic acid Natural products OC(=O)C1=CC=C(O)C=C1 FJKROLUGYXJWQN-UHFFFAOYSA-N 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 125000005981 pentynyl group Chemical group 0.000 description 1
- 125000001791 phenazinyl group Chemical group C1(=CC=CC2=NC3=CC=CC=C3N=C12)* 0.000 description 1
- 125000001484 phenothiazinyl group Chemical group C1(=CC=CC=2SC3=CC=CC=C3NC12)* 0.000 description 1
- 125000001644 phenoxazinyl group Chemical group C1(=CC=CC=2OC3=CC=CC=C3NC12)* 0.000 description 1
- WLJVXDMOQOGPHL-UHFFFAOYSA-N phenylacetic acid Chemical class OC(=O)CC1=CC=CC=C1 WLJVXDMOQOGPHL-UHFFFAOYSA-N 0.000 description 1
- 235000021317 phosphate Nutrition 0.000 description 1
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 230000000865 phosphorylative effect Effects 0.000 description 1
- 125000005498 phthalate group Chemical class 0.000 description 1
- 125000004592 phthalazinyl group Chemical group C1(=NN=CC2=CC=CC=C12)* 0.000 description 1
- 230000000704 physical effect Effects 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 125000004193 piperazinyl group Chemical group 0.000 description 1
- 125000000587 piperidin-1-yl group Chemical group [H]C1([H])N(*)C([H])([H])C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- 125000004483 piperidin-3-yl group Chemical group N1CC(CCC1)* 0.000 description 1
- 125000003386 piperidinyl group Chemical group 0.000 description 1
- 229920000768 polyamine Polymers 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- LPNYRYFBWFDTMA-UHFFFAOYSA-N potassium tert-butoxide Chemical compound [K+].CC(C)(C)[O-] LPNYRYFBWFDTMA-UHFFFAOYSA-N 0.000 description 1
- 150000003141 primary amines Chemical class 0.000 description 1
- MFDFERRIHVXMIY-UHFFFAOYSA-N procaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 1
- 229960004919 procaine Drugs 0.000 description 1
- 239000000047 product Substances 0.000 description 1
- 230000002062 proliferating effect Effects 0.000 description 1
- 235000019260 propionic acid Nutrition 0.000 description 1
- 125000002568 propynyl group Chemical group [*]C#CC([H])([H])[H] 0.000 description 1
- 125000006239 protecting group Chemical group 0.000 description 1
- 238000001243 protein synthesis Methods 0.000 description 1
- 125000001042 pteridinyl group Chemical group N1=C(N=CC2=NC=CN=C12)* 0.000 description 1
- 150000003212 purines Chemical class 0.000 description 1
- 125000000561 purinyl group Chemical group N1=C(N=C2N=CNC2=C1)* 0.000 description 1
- 125000003373 pyrazinyl group Chemical group 0.000 description 1
- 125000004353 pyrazol-1-yl group Chemical group [H]C1=NN(*)C([H])=C1[H] 0.000 description 1
- 125000002098 pyridazinyl group Chemical group 0.000 description 1
- 125000004076 pyridyl group Chemical group 0.000 description 1
- 125000000714 pyrimidinyl group Chemical group 0.000 description 1
- QGKLPGKXAVVPOJ-UHFFFAOYSA-N pyrrolidin-3-one Chemical compound O=C1CCNC1 QGKLPGKXAVVPOJ-UHFFFAOYSA-N 0.000 description 1
- 125000000719 pyrrolidinyl group Chemical group 0.000 description 1
- 229940107700 pyruvic acid Drugs 0.000 description 1
- 125000002294 quinazolinyl group Chemical group N1=C(N=CC2=CC=CC=C12)* 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
- 125000002943 quinolinyl group Chemical group N1=C(C=CC2=CC=CC=C12)* 0.000 description 1
- 125000001567 quinoxalinyl group Chemical group N1=C(C=NC2=CC=CC=C12)* 0.000 description 1
- 125000004621 quinuclidinyl group Chemical group N12C(CC(CC1)CC2)* 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 230000022983 regulation of cell cycle Effects 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 239000011347 resin Substances 0.000 description 1
- 229920005989 resin Polymers 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 229960004889 salicylic acid Drugs 0.000 description 1
- CXMXRPHRNRROMY-UHFFFAOYSA-N sebacic acid Chemical class OC(=O)CCCCCCCCC(O)=O CXMXRPHRNRROMY-UHFFFAOYSA-N 0.000 description 1
- 150000003335 secondary amines Chemical class 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- ILJOYZVVZZFIKA-UHFFFAOYSA-M sodium;1,1-dioxo-1,2-benzothiazol-3-olate;hydrate Chemical compound O.[Na+].C1=CC=C2C(=O)[N-]S(=O)(=O)C2=C1 ILJOYZVVZZFIKA-UHFFFAOYSA-M 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- OGNAOIGAPPSUMG-UHFFFAOYSA-N spiro[2.2]pentane Chemical compound C1CC11CC1 OGNAOIGAPPSUMG-UHFFFAOYSA-N 0.000 description 1
- FYGUBWKMMCWIKB-UHFFFAOYSA-N spiro[2.3]hexane Chemical compound C1CC11CCC1 FYGUBWKMMCWIKB-UHFFFAOYSA-N 0.000 description 1
- HEMCGZPSGYRIOL-UHFFFAOYSA-N spiro[2.4]heptane Chemical compound C1CC11CCCC1 HEMCGZPSGYRIOL-UHFFFAOYSA-N 0.000 description 1
- FOEYMRPOKBCNCR-UHFFFAOYSA-N spiro[2.5]octane Chemical compound C1CC11CCCCC1 FOEYMRPOKBCNCR-UHFFFAOYSA-N 0.000 description 1
- DVDJICIUXXAIKJ-UHFFFAOYSA-N spiro[2.6]nonane Chemical compound C1CC11CCCCCC1 DVDJICIUXXAIKJ-UHFFFAOYSA-N 0.000 description 1
- LBJQKYPPYSCCBH-UHFFFAOYSA-N spiro[3.3]heptane Chemical compound C1CCC21CCC2 LBJQKYPPYSCCBH-UHFFFAOYSA-N 0.000 description 1
- IWDANOJGJIFBEL-UHFFFAOYSA-N spiro[3.4]octane Chemical compound C1CCC21CCCC2 IWDANOJGJIFBEL-UHFFFAOYSA-N 0.000 description 1
- VMWOETMUNAQFAX-UHFFFAOYSA-N spiro[3.5]nonane Chemical compound C1CCC21CCCCC2 VMWOETMUNAQFAX-UHFFFAOYSA-N 0.000 description 1
- PLDXRPSSERMPSV-UHFFFAOYSA-N spiro[3.6]decane Chemical compound C1CCC21CCCCCC2 PLDXRPSSERMPSV-UHFFFAOYSA-N 0.000 description 1
- PHICBFWUYUCFKS-UHFFFAOYSA-N spiro[4.4]nonane Chemical compound C1CCCC21CCCC2 PHICBFWUYUCFKS-UHFFFAOYSA-N 0.000 description 1
- NWTSRIZEBKLUFM-UHFFFAOYSA-N spiro[4.6]undecane Chemical compound C1CCCC21CCCCCC2 NWTSRIZEBKLUFM-UHFFFAOYSA-N 0.000 description 1
- NECLQTPQJZSWOE-UHFFFAOYSA-N spiro[5.5]undecane Chemical compound C1CCCCC21CCCCC2 NECLQTPQJZSWOE-UHFFFAOYSA-N 0.000 description 1
- ZDNANLAHKKNCTC-UHFFFAOYSA-N spiro[5.6]dodecane Chemical compound C1CCCCC21CCCCCC2 ZDNANLAHKKNCTC-UHFFFAOYSA-N 0.000 description 1
- NGGUAMSYAYRZGK-UHFFFAOYSA-N spiro[6.6]tridecane Chemical compound C1CCCCCC21CCCCCC2 NGGUAMSYAYRZGK-UHFFFAOYSA-N 0.000 description 1
- CTDQAGUNKPRERK-UHFFFAOYSA-N spirodecane Chemical compound C1CCCC21CCCCC2 CTDQAGUNKPRERK-UHFFFAOYSA-N 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- LSNNMFCWUKXFEE-UHFFFAOYSA-L sulfite Chemical class [O-]S([O-])=O LSNNMFCWUKXFEE-UHFFFAOYSA-L 0.000 description 1
- 150000003467 sulfuric acid derivatives Chemical class 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 238000001308 synthesis method Methods 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 235000012222 talc Nutrition 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 239000011975 tartaric acid Substances 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- 150000003892 tartrate salts Chemical class 0.000 description 1
- 238000003419 tautomerization reaction Methods 0.000 description 1
- BGUYAMZPJMTFRU-UHFFFAOYSA-N tert-butyl 2,6-diazaspiro[3.4]octane-6-carboxylate Chemical compound C1N(C(=O)OC(C)(C)C)CCC11CNC1 BGUYAMZPJMTFRU-UHFFFAOYSA-N 0.000 description 1
- NRADOPGBTAJXKB-UHFFFAOYSA-N tert-butyl 2,7-diazaspiro[3.5]nonane-7-carboxylate Chemical compound C1CN(C(=O)OC(C)(C)C)CCC11CNC1 NRADOPGBTAJXKB-UHFFFAOYSA-N 0.000 description 1
- ISIJQEHRDSCQIU-UHFFFAOYSA-N tert-butyl 2,7-diazaspiro[4.5]decane-7-carboxylate Chemical compound C1N(C(=O)OC(C)(C)C)CCCC11CNCC1 ISIJQEHRDSCQIU-UHFFFAOYSA-N 0.000 description 1
- NFNCPNAVNRBDOU-UHFFFAOYSA-N tert-butyl 2,8-diazaspiro[4.5]decane-2-carboxylate Chemical compound C1N(C(=O)OC(C)(C)C)CCC21CCNCC2 NFNCPNAVNRBDOU-UHFFFAOYSA-N 0.000 description 1
- YLKHACHFJMCIRE-UHFFFAOYSA-N tert-butyl 3,9-diazaspiro[5.5]undecane-3-carboxylate Chemical compound C1CN(C(=O)OC(C)(C)C)CCC11CCNCC1 YLKHACHFJMCIRE-UHFFFAOYSA-N 0.000 description 1
- BYWAFPDHLZFJKP-UHFFFAOYSA-N tert-butyl 3-(hydroxymethyl)-8-azaspiro[4.5]decane-8-carboxylate Chemical compound CC(C)(C)OC(=O)N1CCC2(CCC(CO)C2)CC1 BYWAFPDHLZFJKP-UHFFFAOYSA-N 0.000 description 1
- ZXXJXEXKMUZKPG-UHFFFAOYSA-N tert-butyl 3-oxo-8-azaspiro[4.5]decane-8-carboxylate Chemical compound C1CN(C(=O)OC(C)(C)C)CCC11CC(=O)CC1 ZXXJXEXKMUZKPG-UHFFFAOYSA-N 0.000 description 1
- VVDCRJGWILREQH-UHFFFAOYSA-N tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,6-dihydro-2h-pyridine-1-carboxylate Chemical compound C1N(C(=O)OC(C)(C)C)CCC(B2OC(C)(C)C(C)(C)O2)=C1 VVDCRJGWILREQH-UHFFFAOYSA-N 0.000 description 1
- ZTTLVQSVYNKGJH-UHFFFAOYSA-N tert-butyl 4-[3-(hydroxymethyl)cyclobutyl]piperazine-1-carboxylate Chemical compound CC(C)(C)OC(=O)N1CCN(CC1)C2CC(C2)CO ZTTLVQSVYNKGJH-UHFFFAOYSA-N 0.000 description 1
- PWQLFIKTGRINFF-UHFFFAOYSA-N tert-butyl 4-hydroxypiperidine-1-carboxylate Chemical compound CC(C)(C)OC(=O)N1CCC(O)CC1 PWQLFIKTGRINFF-UHFFFAOYSA-N 0.000 description 1
- UXZBTEIHIQFMIT-UHFFFAOYSA-N tert-butyl 9-oxo-3-azaspiro[5.5]undecane-3-carboxylate Chemical compound C1CN(C(=O)OC(C)(C)C)CCC21CCC(=O)CC2 UXZBTEIHIQFMIT-UHFFFAOYSA-N 0.000 description 1
- 150000003512 tertiary amines Chemical class 0.000 description 1
- WHRNULOCNSKMGB-UHFFFAOYSA-N tetrahydrofuran thf Chemical compound C1CCOC1.C1CCOC1 WHRNULOCNSKMGB-UHFFFAOYSA-N 0.000 description 1
- 125000003718 tetrahydrofuranyl group Chemical group 0.000 description 1
- 125000001412 tetrahydropyranyl group Chemical group 0.000 description 1
- 125000000147 tetrahydroquinolinyl group Chemical group N1(CCCC2=CC=CC=C12)* 0.000 description 1
- CZDYPVPMEAXLPK-UHFFFAOYSA-N tetramethylsilane Chemical compound C[Si](C)(C)C CZDYPVPMEAXLPK-UHFFFAOYSA-N 0.000 description 1
- 125000003831 tetrazolyl group Chemical group 0.000 description 1
- WROMPOXWARCANT-UHFFFAOYSA-N tfa trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F.OC(=O)C(F)(F)F WROMPOXWARCANT-UHFFFAOYSA-N 0.000 description 1
- 229960004559 theobromine Drugs 0.000 description 1
- 125000001113 thiadiazolyl group Chemical group 0.000 description 1
- 125000001984 thiazolidinyl group Chemical group 0.000 description 1
- 125000000335 thiazolyl group Chemical group 0.000 description 1
- 125000005985 thienyl[1,3]dithianyl group Chemical group 0.000 description 1
- LBLYYCQCTBFVLH-UHFFFAOYSA-M toluenesulfonate group Chemical group C=1(C(=CC=CC1)S(=O)(=O)[O-])C LBLYYCQCTBFVLH-UHFFFAOYSA-M 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 238000000844 transformation Methods 0.000 description 1
- 230000001052 transient effect Effects 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- 125000004306 triazinyl group Chemical group 0.000 description 1
- 125000001425 triazolyl group Chemical group 0.000 description 1
- INQOMBQAUSQDDS-FIBGUPNXSA-N trideuterio(iodo)methane Chemical compound [2H]C([2H])([2H])I INQOMBQAUSQDDS-FIBGUPNXSA-N 0.000 description 1
- ITMCEJHCFYSIIV-UHFFFAOYSA-N triflic acid Chemical compound OS(=O)(=O)C(F)(F)F ITMCEJHCFYSIIV-UHFFFAOYSA-N 0.000 description 1
- YFTHZRPMJXBUME-UHFFFAOYSA-N tripropylamine Chemical compound CCCN(CCC)CCC YFTHZRPMJXBUME-UHFFFAOYSA-N 0.000 description 1
- 125000005455 trithianyl group Chemical group 0.000 description 1
- 210000004881 tumor cell Anatomy 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
- 229920002554 vinyl polymer Polymers 0.000 description 1
- 239000003643 water by type Substances 0.000 description 1
- 229910052725 zinc Inorganic materials 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
- C07D471/22—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed systems contains four or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
- C07D471/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
- C07D471/04—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
- C07D471/12—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
- C07D471/14—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D487/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
- C07D487/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
- C07D487/04—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D519/00—Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
Definitions
- One embodiment provides a compound, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (I) :
- R 1 , R 2 , R 3 , and R 4 are each independently selected from hydrogen, optionally substituted straight or branched C 1-6 alkyl, optionally substituted C 1-3 alkoxy, halogen, -N (R 7 ) 2 , -OR 7 , and cyano; or
- R 1 and R 2 , R 2 and R 3 , or R 3 and R 4 together with the intervening atoms to which they are attached form an optionally substituted 5-6 membered carbocyclyl or optionally substituted heterocyclyl ring;
- R 5 is or
- R 1 and R 5 are taken together with the intervening atoms to which they are attached to form an optionally substituted 10-12 membered ring;
- each R 6 is independently selected from halogen, nitro, cyano, -OR 7 , -N (R 7 ) 2 , -SR 7 , -S (O) R 7 , -S (O) 2 R 7 , -S (O) 2 N (R 7 ) 2 , -NR 7 S (O) 2 R 7 , -NR 7 S (O) 2 N (R 7 ) 2 , -C (O) R 7 , -C (O) OR 7 , -OC (O) R 7 , -OC (O) OR 7 , -OC (O) N (R 7 ) 2 , -NR 7 C (O) R 7 , -NR 7 C (O) OR 7 , -NR 7 C (O) N (R 7 ) 2 , -C (O) N (R 7 ) 2 , -P (O) (OR 7 ) 2 , -P (O) (R 7 ) 2 , optionally substituted C
- W is CH, CR 6 , or N;
- v 0, 1, 2, or 3;
- each R 7 is independently hydrogen or optionally substituted C 1-6 alkyl
- ring B is an optionally substituted C 6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;
- L is -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*;
- Ak 1 is - (CR 8 R 9 ) k -or -O-;
- Ak 3 is - (CR 8 R 9 ) n -;
- each R 8 and R 9 are independently selected from hydrogen, halogen, -NH 2 , -OH, cyano, optionally substituted C 1-3 alkyl, and optionally substituted C 3 cycloalkyl; or
- Z 1 and Z 2 are each independently selected from bond, -O-, -NR 7 -, -S-, optionally substituted 5-10 membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C 4-11 cycloalkylene;
- R 50 is hydrogen, optionally substituted straight or branched C 1-6 alkyl, or optionally substituted C 3-6 cycloalkyl;
- each R 7 is independently hydrogen or optionally substituted C 1-6 alkyl
- Ak 1 is - (CR 8 R 9 ) k -or -O-;
- Ak 2 is - (CR 8 R 9 ) m -;
- each of k, m, and n is an integer selected from 0 to 10;
- One embodiment provides a compound, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (III) :
- each R 83 is independently selected from halogen, nitro, cyano, -OR 7 , -N (R 7 ) 2 , -SR 7 , -S (O) R 7 , -S (O) 2 R 7 , -S (O) 2 N (R 7 ) 2 , -NR 7 S (O) 2 R 7 , -NR 7 S (O) 2 N (R 7 ) 2 , -C (O) R 7 , -C (O) OR 7 , -OC (O) R 7 , -OC (O) OR 7 , -OC (O) N (R 7 ) 2 , -NR 7 C (O) R 7 , -NR 7 C (O) OR 7 , -NR 7 C (O) N (R 7 ) 2 , -NR 7 C (O) OR 7 , -NR 7 C (O) N (R 7 ) 2 , -C (O) N (R 7 ) 2 , -P (
- v 0, 1, 2, or 3;
- ring B is an optionally substituted C 6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;
- L is -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*;
- Ak 1 is - (CR 8 R 9 ) k -or -O-;
- Ak 2 is - (CR 8 R 9 ) m -;
- Cycloalkylene refers to a divalent carbocyclyl or cycloalkyl linking the rest of the molecule to a radical group.
- Heterocyclene refers to a divalent heterocyclyl linking the rest of the molecule to a radical group.
- Heteroarylene refers to a divalent heteroaryl radical.
- C-heteroaryl refers to a heteroaryl radical as defined above and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a carbon atom in the heteroaryl radical.
- a C-heteroaryl radical is optionally substituted as described above for heteroaryl radicals.
- Heteroarylalkoxy refers to a radical bonded through an oxygen atom of the formula –O-R c -heteroaryl, where R c is an alkylene chain as defined above. If the heteroaryl is a nitrogen-containing heteroaryl, the heteroaryl is optionally attached to the alkyl radical at the nitrogen atom.
- the alkylene chain of the heteroarylalkoxy radical is optionally substituted as defined above for an alkylene chain.
- the heteroaryl part of the heteroarylalkoxy radical is optionally substituted as defined above for a heteroaryl group.
- CD 3 I iodomethane-d 3
- solvates refers to a composition of matter that is the solvent addition form.
- solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and are formed during the process of making with pharmaceutically acceptable solvents such as water, ethanol, and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Solvates of compounds described herein are conveniently prepared or formed during the processes described herein. The compounds provided herein optionally exist in either unsolvated as well as solvated forms.
- R 60 and R 61 are each independently hydrogen, halogen, cyano, -OR 70 , -SR 70 , -S (O) R 70 , -S (O) 2 R 70 , -N (R 70 ) 2 , or an optionally substituted C 1-3 alkyl;
- R 60 and R 61 are taken together to form -C (O) -;
- each R 62 is independently halogen, cyano, -OR 70 , -SR 70 , -S (O) R 70 , -S (O) 2 R 70 , -N (R 70 ) 2 , or an optionally substituted C 1-3 alkyl;
- R 63 is O, S, or NR 70 ;
- g 0, 1, 2 or 3;
- the recruitment motif is a CRBN ligand selected from
- Z 1 and Z 2 are each independently selected from bond, -O-, -NR 7 -, -S-, optionally substituted 5-10 membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C 4-11 cycloalkylene;
- Z 1 and Z 2 are heteroarylene, heterocyclene or cycloalkylene; and wherein the point of bonding to the CRBN ligand is indicated by the asterisk.
- the linking motif is represented by -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*and is selected from a molecular fragment illustrated below:
- X, X1, and X2 at each instance is independently CH, CR e , or N;
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- q 0, 1, 2, 3, or 4.
- k 0, 1, 2, 3, 4, 5, or 6;
- X, X1, and X2 at each instance is independently CH, CR e , or N;
- each R e in Z 1 , X, X1, and X2 is independently selected from -F, methyl, and -CF 3 ;
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- the linking motif is represented by -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*and is selected from a molecular fragment illustrated below:
- X1, X2, X3, and X4 at each instance is independently CH, CR e , or N ;
- n 0, 1, 2, 3, 4, 5 or 6;
- q 0, 1, 2, 3, or 4;
- the linking motif is represented by -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*and is selected from a molecular fragment illustrated below:
- each R L is independently hydrogen, halogen, or C 1-3 alkyl.
- X1, X2, X3, and X4 at each instance is independently CH, CR e , or N ;
- each R e in Z 1 , Z 2 , X1, X2, X3, and X4 is independently selected from halogen, -NH 2 , -OH, cyano, C 1-3 alkyl, and C 1-3 haloalkyl;
- each R L is independently hydrogen, halogen, or C 1-3 alkyl.
- q 0, 1, 2, 3, or 4;
- each R 7 is independently hydrogen or optionally substituted C 1-6 alkyl
- each R 83 is independently selected from halogen, nitro, cyano, -OR 7 , -N (R 7 ) 2 , -SR 7 , -S (O) R 7 , -S (O) 2 R 7 , -S (O) 2 N (R 7 ) 2 , -NR 7 S (O) 2 R 7 , -NR 7 S (O) 2 N (R 7 ) 2 , -C (O) R 7 , -C (O) OR 7 , -OC (O) R 7 , -OC (O) OR 7 , -OC (O) N (R 7 ) 2 , -NR 7 C (O) R 7 , -NR 7 C (O) OR 7 , -NR 7 C (O) N (R 7 ) 2 , -NR 7 C (O) OR 7 , -NR 7 C (O) N (R 7 ) 2 , -C (O) N (R 7 ) 2 , -P (
- R 85 and R 86 are taken together to form -C (O) -;
- W is CH, CR 83 , or N;
- each R 7 is independently hydrogen or optionally substituted C 1-6 alkyl
- a 1 , A 2 , A 3 , and A 4 are each independently CH or N;
- L cyc is -L1 cyc -, -L1 cyc -L2 cyc -, -L1 cyc -L2 cyc -L3 cyc -, or -L1 cyc -L2 cyc -L3 cyc -L4 cyc -;
- R c , R d , R y , R f , and R g are each independently hydrogen, halogen, -OH, -SH, nitro, cyano, -NH 2 , optionally substituted C 1-6 alkyl, optionally substituted C 1-6 haloalkyl, optionally substituted C 1-6 alkoxy, optionally substituted C 2-6 alkenyl, optionally substituted C 2-6 alkynyl, optionally substituted C 3 -12 cycloalkyl, optionally substituted 3-12 membered heterocyclyl, optionally substituted C 6-14 aryl, or optionally substituted 5-14 membered heteroaryl; or
- any two of R c , R d , R y , R f , and R g are taken together with the atoms to which they are attached to form an optionally substituted C 3-12 cycloalkyl, an optionally substituted 3-12 membered heterocyclyl, an optionally substituted C 6-14 aryl group, or an optionally substituted 5 -14 membered heteroaryl;
- n1, and n2 are each independently an integer selected from 0 to 10;
- ring B is an optionally substituted C 6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring.
- R 5 is or
- R 1 and R 5 are taken together with the intervening atoms to which they are attached to form an optionally substituted 10-12 membered ring;
- each R 6 is independently selected from halogen, nitro, cyano, -OR 7 , -N (R 7 ) 2 , -SR 7 , -S (O) R 7 , -S (O) 2 R 7 , -S (O) 2 N (R 7 ) 2 , -NR 7 S (O) 2 R 7 , -NR 7 S (O) 2 N (R 7 ) 2 , -C (O) R 7 , -C (O) OR 7 , -OC (O) R 7 , -OC (O) OR 7 , -OC (O) N (R 7 ) 2 , -NR 7 C (O) R 7 , -NR 7 C (O) OR 7 , -NR 7 C (O) N (R 7 ) 2 , -C (O) N (R 7 ) 2 , -P (O) (OR 7 ) 2 , -P (O) (R 7 ) 2 , optionally substituted C
- v 0, 1, 2, or 3;
- each R 7 is independently hydrogen or optionally substituted C 1-6 alkyl
- ring B is an optionally substituted C 6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;
- Ak 1 is - (CR 8 R 9 ) k -or -O-;
- Ak 3 is - (CR 8 R 9 ) n -;
- each R 8 and R 9 are independently selected from hydrogen, halogen, -NH 2 , -OH, cyano, optionally substituted C 1-3 alkyl, and optionally substituted C 3 cycloalkyl; or
- each - (CR 8 R 9 ) - may independently form -C (O) -;
- Z 1 and Z 2 are each independently selected from bond, -O-, -NR 7 -, -S-, optionally substituted 5-10 membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C 4-11 cycloalkylene;
- R 1 and R 2 , R 2 and R 3 , or R 3 and R 4 together with the intervening atoms to which they are attached form an optionally substituted 5-6 membered carbocyclyl or optionally substituted heterocyclyl ring, wherein the carbocyclyl and heterocyclyl ring is optionally substituted with one or more R e ;
- each R 7 is independently hydrogen or optionally substituted C 1-6 alkyl, wherein the alkyl, is optionally substituted with one or more R e ; or
- R 7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring, wherein the 3-6 membered heterocyclyl ring is optionally substituted with one or more R e ;
- ring B is an optionally substituted C 6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more R B ;
- each R 8 and R 9 are independently selected from hydrogen, halogen, -NH 2 , -OH, cyano, optionally substituted C 1-3 alkyl, and optionally substituted C 3 cycloalkyl, wherein the alkyl and cycloalkyl are each independently optionally substituted with one or more R e ; or
- Z 1 and Z 2 are heteroarylene, heterocyclene or cycloalkylene
- R 1 and R 5 are taken together to with the intervening atoms to which they are attached to form an optionally substituted 10-12 membered ring, wherein the ring is optionally substituted with one or more substituents selected from halogen, -CN, -OH, -OCH 3 , -S (O) CH 3 , -S (O) 2 CH 3 , -S (O) 2 NH 2 , -S (O) 2 NHCH 3 , -S (O) 2 N (CH 3 ) 2 , -NH 2 , -NHCH 3 , -N (CH 3 ) 2 , -C (O) CH 3 , -C (O) OH, -C (O) OCH 3 , C 1 - 6 alkyl, C 1 - 6 haloalkyl, C 1 - 6 hydroxyalkyl, C 1 - 6 aminoalkyl, and C 1 - 6 heteroalkyl.
- R 10 is selected from hydrogen, halogen, hydroxyl, and C 1 - 3 alkyl. In some embodiments of Formula (I) and Formula (I-3) , R 10 is selected from halogen, hydroxyl, and C 1 - 3 alkyl. In some embodiments of Formula (I) and Formula (I-3) , R 10 is selected from hydrogen and fluoro. In some embodiments of Formula (I) , the compound has a structure of Formula (I-3a) or Formula (I-3b) :
- each R 10 is selected from halogen, -CN, -OH, -OCH 3 , -S (O) CH 3 , -S (O) 2 CH 3 , -S (O) 2 NH 2 , -S (O) 2 NHCH 3 , -S (O) 2 N (CH 3 ) 2 , -NH 2 , -NHCH 3 , -N (CH 3 ) 2 , -C (O) CH 3 , -C (O) OH, -C (O) OCH 3 , C 1 - 6 alkyl, C 1 - 6 haloalkyl, C 1 - 6 hydroxyalkyl, C 1 - 6 aminoalkyl, and C 1 - 6 heteroalkyl.
- R 10 is selected from halogen and C 1 - 3 alkyl. In some embodiments, R 10 is fluoro.
- each R 6 is independently selected from halogen, cyano, -OR 7 , - N (R 7 ) 2 , -C (O) R 7 , -C (O) OR 7 , -OC (O) R 7 , -OC (O) OR 7 , -OC (O) N (R 7 ) 2 , -NR 7 C (O) R 7 , -NR 7 C (O) OR 7 , -NR 7 C (O) N (R 7 ) 2 , -C (O) N (R 7 ) 2 , optionally substituted C 1-6 alkyl, optionally substituted C 1-6 haloalky
- each R B is independently straight or branched C 1-3 alkyl or halogen. In some embodiments, each R B is independently methyl or fluoro. In some embodiments, each R B is independently methyl or fluoro and j is 0, 1, or 2. In some embodiments, j is 0. In some embodiments, ring B is selected from In some embodiments, ring B is selected from
- ring B is an optionally substituted C 11-14 cycloalkyl or an optionally substituted 11-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more R B .
- ring B is:
- j 0, 1, 2, 3, or 4.
- n 0, 1, 2, 3, 4, 5 or 6;
- k 0, 1, 2, 3, 4, 5, or 6;
- each R e in Z 1 , X, X1, and X2 is independently selected from -F, methyl, and -CF 3 ;
- k 0, 1, 2, 3, 4, 5, or 6;
- Formula (I) is -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*, wherein
- L is -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*, wherein
- L is -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*, wherein
- Z 1 and Z 2 are each independently selected from bond, pyrazole optionally substituted with R L , wherein each R L is independently hydrogen, halogen, or C 1-3 alkyl, 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and C 4-6 cycloalkylene.
- Formula (I) Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- X1, X2, X3, and X4 at each instance is independently CH, CR e , or N ;
- each R L is independently hydrogen, halogen, or C 1-3 alkyl.
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- q 0, 1, 2, 3, or 4;
- h 0, 1, 2, 3, or 4.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or C 1-3 alkyl.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or methyl.
- each R L is independently hydrogen, -F, -Cl, or methyl.
- R 8 and R 9 are each independently selected from hydrogen , methyl, and halogen; or each - (CR 8 R 9 ) -may independently form -C (O) -.
- Formula (I) Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- X1, X2, X3, and X4 at each instance is independently CH, CR e , or N ;
- each R e in Z 1 , Z 2 , X1, X2, X3, and X4 is independently selected from halogen, -NH 2 , -OH, cyano, C 1-3 alkyl, and C 1-3 haloalkyl;
- each R L is independently hydrogen, halogen, or C 1-3 alkyl.
- n 0, 1, 2, 3, 4, 5 or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- q 0, 1, 2, 3, or 4;
- h 0, 1, 2, 3, or 4.
- Formula (I) Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- each R e in Z 1 , Z 2 , X1, X2, X3, and X4 is independently selected from halogen, -NH 2 , -OH, cyano, C 1-3 alkyl, and C 1-3 haloalkyl;
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- h 0, 1, 2, 3, or 4.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or C 1-3 alkyl.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or methyl.
- each R L is independently hydrogen, -F, -Cl, or methyl.
- Formula (I) Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- each R e is independently selected from halogen, -OH, and methyl
- q 0, 1, or 2;
- X1 and X2 at each instance is independently CH, CR e , or N;
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6.
- k is 0, 1, or 2; and m is 0, 1, or 2.
- Formula (I) Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- R 51 and R 52 are each independently hydrogen or optionally substituted straight or branched C 1-6 alkyl; or
- Z 1 and Z 2 are heteroarylene, heterocyclene or cycloalkylene
- R 50 is methyl or ethyl. In some embodiments, R 50 is methyl. In some embodiments, R 50 is ethyl. In some embodiments, R 50 is cyclopropyl or cyclobutyl. In some embodiments, R 50 is cyclobutyl. In some embodiments, R 50 is cyclopropyl. In some embodiments, R 51 and R 52 are each independently hydrogen, optionally substituted straight or branched C 1-6 alkyl, or R 51 and R 52 are taken together to form -C (O) -.
- Y 1 and Y 2 are N or CH;
- n 0, 1, 2, 3, 4, 5 or 6;
- Formula (II) has a structure selected from:
- L is -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*, wherein
- Z 1 and Z 2 are each independently selected from bond, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C 4-6 cycloalkylene.
- Z 1 and Z 2 are each independently selected from bond, pyrazole optionally substituted with R L , wherein each R L is independently hydrogen, halogen, or C 1-3 alkyl, 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and C 4-6 cycloalkylene.
- Formula (II) has a structure selected from:
- X1, X2, X3, and X4 at each instance is independently CH, CR e , or N ;
- each R e in Z 1 , Z 2 , X1, X2, X3, and X4 is independently selected from halogen, -NH 2 , -OH, cyano, C 1-3 alkyl, and C 1-3 haloalkyl;
- each R L is independently hydrogen, halogen, or C 1-3 alkyl.
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- q 0, 1, 2, 3, or 4;
- h 0, 1, 2, 3, or 4.
- k is 0, 1, or 2; m is 0, 1, or 2; and n is 0.
- q is 0, 1, or 2; and h is 0, 1, or 2.
- q is 0 or 1; and h is 0 or 1.
- q is 0; and h is 0.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or C 1-3 alkyl.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or methyl.
- each R L is independently hydrogen, -F, -Cl, or methyl.
- R 8 and R 9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR 8 R 9 ) -may independently form -C (O) -.
- Formula (II) has a structure selected from:
- X1, X2, X3, and X4 at each instance is independently CH, CR e , or N ;
- each R L is independently hydrogen, halogen, or C 1-3 alkyl.
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- q 0, 1, 2, 3, or 4;
- h 0, 1, 2, 3, or 4.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or C 1-3 alkyl.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or methyl.
- each R L is independently hydrogen, -F, -Cl, or methyl.
- X1, X2, X3, and X4 at each instance is independently CH, CR e , or N ;
- each R e in Z 1 , Z 2 , X1, X2, X3, and X4 is independently selected from halogen, -NH 2 , -OH, cyano, C 1-3 alkyl, and C 1-3 haloalkyl;
- each R L is independently hydrogen, halogen, or C 1-3 alkyl.
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- q 0, 1, 2, 3, or 4;
- h 0, 1, 2, 3, or 4.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or C 1-3 alkyl.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or methyl.
- each R L is independently hydrogen, -F, -Cl, or methyl.
- Formula (II) has a structure selected from:
- h 0, 1, or 2.
- each R 83 is independently selected from halogen, nitro, cyano, -OR 7 , -N (R 7 ) 2 , -SR 7 , -S (O) R 7 , -S (O) 2 R 7 , -S (O) 2 N (R 7 ) 2 , -NR 7 S (O) 2 R 7 , -NR 7 S (O) 2 N (R 7 ) 2 , -C (O) R 7 , -C (O) OR 7 , -OC (O) R 7 , -OC (O) OR 7 , -OC (O) N (R 7 ) 2 , -NR 7 C (O) R 7 , -NR 7 C (O) OR 7 , -NR 7 C (O) N (R 7 ) 2 , -NR 7 C (O) OR 7 , -NR 7 C (O) N (R 7 ) 2 , -C (O) N (R 7 ) 2 , -P (
- W is CH, CR 83 , or N;
- Ak 1 is - (CR 8 R 9 ) k -or -O-;
- Ak 2 is - (CR 8 R 9 ) m -;
- each R 83 is independently selected from halogen, cyano, -OR 7 , -N (R 7 ) 2 , -C (O) R 7 , -C (O) OR 7 , -OC (O) R 7 , -OC (O) OR 7 , -OC (O) N (R 7 ) 2 , -NR 7 C (O) R 7 , -NR 7 C (O) OR 7 , -NR 7 C (O) N (R 7 ) 2 , -C (O) N (R 7 ) 2 , optionally substituted C 1-6 alkyl, optionally substituted C 1-6 haloalkyl, and optionally substituted C 1-6 alkoxy.
- W is CH or N.
- W is CR 83 or N.
- W is CH or CR 83 .
- W is CH.
- W is CR 83 .
- W is N.
- ring B is a 6-14 membered carbocyclyl ring or a 6-14 membered heterocyclyl ring selected from:
- Y 1 and Y 2 are N or CH;
- q 0, 1, 2, 3, or 4.
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6.
- Formula (III) In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- X, X1, and X2 at each instance is independently CH, CR e , or N;
- each R e in Z 1 , X, X1, and X2 is independently selected from -F, methyl, and -CF 3 ;
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- n 0, 1, 2, 3, 4, 5 or 6.
- Formula (III) In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- L is -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*, wherein
- Z 1 and Z 2 are each independently selected from bond, -O-, -NR 7 -, -S-, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C 4-6 cycloalkylene.
- Z 1 and Z 2 are each independently selected from bond, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C 4-6 cycloalkylene.
- L is -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*, wherein
- Formula (III) In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- X1, X2, X3, and X4 at each instance is independently CH, CR e , or N ;
- each R e in Z 1 , Z 2 , X1, X2, X3, and X4 is independently selected from halogen, -NH 2 , -OH, cyano, C 1-3 alkyl, and C 1-3 haloalkyl;
- each R L is independently hydrogen, halogen, or C 1-3 alkyl.
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- q 0, 1, 2, 3, or 4;
- h 0, 1, 2, 3, or 4.
- Formula (III) In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- X1, X2, X3, and X4 at each instance is independently CH, CR e , or N ;
- each R e in Z 1 , Z 2 , X1, X2, X3, and X4 is independently selected from halogen, -NH 2 , -OH, cyano, C 1-3 alkyl, and C 1-3 haloalkyl;
- each R L is independently hydrogen, halogen, or C 1-3 alkyl.
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- q 0, 1, 2, 3, or 4;
- Formula (III) In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- each R e in Z 1 , Z 2 , X1, X2, X3, and X4 is independently selected from halogen, -NH 2 , -OH, cyano, C 1-3 alkyl, and C 1-3 haloalkyl;
- each R L is independently hydrogen, halogen, or C 1-3 alkyl.
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- q 0, 1, 2, 3, or 4;
- Formula (III) In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- each R L is independently hydrogen, halogen, or C 1-3 alkyl
- each R e is independently selected from halogen, -OH, and methyl
- q 0, 1, or 2;
- h 0, 1, or 2.
- any two of R c , R d , R f , and R g are taken together with the atoms to which they are attached to form an optionally substituted C 3-12 cycloalkyl group, an optionally substituted 3-12 membered heterocyclyl, an optionally substituted C 6-14 aryl group, or an optionally substituted 5 -14 membered heteroaryl, wherein the cycloalkyl, heterocyclyl, aryl, and heteroaryl are independently optionally substituted with one or more R e ;
- each R e is independently selected from halogen, -CN, -OH, -OCH 3 , -S (O) CH 3 , -S (O) 2 CH 3 , -S (O) 2 NH 2 , -S (O) 2 NHCH 3 , -S (O) 2 N (CH 3 ) 2 , -NH 2 , -NHCH 3 , -N (CH 3 ) 2 , -C (O) CH 3 , -C (O) OH, -C (O) OCH 3 , C 1 - 6 alkyl, C 1 - 6 haloalkyl, C 1 - 6 hydroxyalkyl, C 1 - 6 aminoalkyl, and C 1 - 6 heteroalkyl; or
- Y 1 and Y 2 are N or CH;
- j 0, 1, 2, 3, or 4;
- q 0, 1, 2, 3, or 4.
- Formula (IV) In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- each R e in Z 1 , X, X1, and X2 is independently selected from halogen, -NH 2 , -OH, C 1-3 alkyl, and -CF 3 ;
- n 0, 1, 2, 3, 4, 5 or 6;
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- q 0, 1, 2, 3, or 4;
- h 0, 1, 2, 3, or 4.
- k is 0, 1, or 2; m is 0, 1, or 2; and n is 0.
- q is 0, 1, or 2; and h is 0, 1, or 2.
- q is 0 or 1; and h is 0 or 1.
- q is 0; and h is 0.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or C 1-3 alkyl.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or methyl.
- each R L is independently hydrogen, -F, -Cl, or methyl.
- R 8 and R 9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR 8 R 9 ) -may independently form -C (O) -.
- Formula (IV) In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- X1, X2, X3, and X4 at each instance is independently CH, CR e , or N ;
- each R e in Z 1 , Z 2 , X1, X2, X3, and X4 is independently selected from halogen, -NH 2 , -OH, cyano, C 1-3 alkyl, and C 1-3 haloalkyl;
- each R L is independently hydrogen, halogen, or C 1-3 alkyl.
- n 0, 1, 2, 3, 4, 5 or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- q 0, 1, 2, 3, or 4;
- h 0, 1, 2, 3, or 4.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or C 1-3 alkyl.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or methyl.
- each R L is independently hydrogen, -F, -Cl, or methyl.
- Formula (IV) In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- X1, X2, X3, and X4 at each instance is independently CH, CR e , or N ;
- each R e in Z 1 , Z 2 , X1, X2, X3, and X4 is independently selected from halogen, -NH 2 , -OH, cyano, C 1-3 alkyl, and C 1-3 haloalkyl;
- each R L is independently hydrogen, halogen, or C 1-3 alkyl.
- k 0, 1, 2, 3, 4, 5, or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- n 0, 1, 2, 3, 4, 5 or 6;
- q 0, 1, 2, 3, or 4;
- h 0, 1, 2, 3, or 4.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or C 1-3 alkyl.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or methyl.
- each R L is independently hydrogen, -F, -Cl, or methyl.
- Formula (IV) In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , -Ak 1 -Z 1 -Ak 2 -Z 2 -Ak 3 -*has a structure selected from:
- each R L is independently hydrogen, halogen, or C 1-3 alkyl
- each R e is independently selected from halogen, -OH, and methyl
- q 0, 1, or 2;
- h 0, 1, or 2.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or C 1-3 alkyl.
- each R L is independently hydrogen, -F, -Cl, -Br, -I, or methyl.
- each R L is independently hydrogen, -F, -Cl, or methyl.
- Formula (I) is a CRBN ligand having the structure
- ring A is selected from:
- R 60 and R 61 are taken together to form -C (O) -;
- R 63 is O, S, or NR 70 ;
- each R 70 is independently hydrogen or optionally substituted C 1-6 alkyl
- R 60 and R 61 are each independently hydrogen, halogen, cyano, -OR 70 , -SR 70 , -S (O) R 70 , -S (O) 2 R 70 , -N (R 70 ) 2 , or an optionally substituted C 1-3 alkyl, wherein the alkyl is optionally substituted with one or more R e ;
- ring A is In some embodiments, ring A is In some embodiments, ring A is In some embodiments, ring A is In some embodiments, ring A is In some embodiments, ring A is In some embodiments, ring A is In some embodiments, ring A is In some embodiments, ring A is In some embodiments, ring A is In some embodiments, ring A is In some embodiments, ring A is In some embodiments, ring A is In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , ring A is In some embodiments, ring A is In some embodiments, ring A is In some embodiment
- compounds as described herein has a structure provided in Table 2.
- the injection formulation is an aqueous formulation.
- the injection formulation is a non-aqueous formulation.
- the injection formulation is an oil-based formulation, such as sesame oil, or the like.
- One embodiment provides a compound of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , or a pharmaceutically acceptable salt or solvate thereof, for use in a method of treatment of the human or animal body.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Provided herein are compounds which target WEE1 kinase proteins for ubiquitination and proteasomal degradation. Also provided herein are methods for using said compounds for the treatment of diseases.
Description
WEE1 is a kinase belonging to the Ser/Thr family of protein kinases and is a key regulator of cell cycle progression. Pharmacological intervention in the cell cycle is strategy for the treatment of cancer and other proliferative disease. The present disclosure provides compounds, compositions and methods for the reduction or elimination of WEE1 activity via the targeting of WEE1 protein in a cell through ubiquitination and proteasomal degradation.
BRIEF SUMMARY OF THE INVENTION
Provided herein are compounds which target WEE1 proteins for ubiquitination and proteasomal degradation. Also provided herein are methods for using said compounds for the treatment of diseases.
One embodiment provides a compound, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (I) :
wherein,
R1, R2, R3, and R4 are each independently selected from hydrogen, optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, and cyano; or
R1 and R2, R2 and R3, or R3 and R4 together with the intervening atoms to which they are attached form an optionally substituted 5-6 membered carbocyclyl or optionally substituted heterocyclyl ring;
R5 isor
R1 and R5 are taken together with the intervening atoms to which they are attached to form an optionally substituted 10-12 membered ring;
each R6 is independently selected from halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted
C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;
W is CH, CR6, or N;
v is 0, 1, 2, or 3;
each R7 is independently hydrogen or optionally substituted C1-6 alkyl; or
two R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring;
ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;
L is -Ak1-Z1-Ak2-Z2-Ak3-*;
Ak1 is - (CR8R9) k-or -O-;
Ak2 is - (CR8R9) m-;
Ak3 is - (CR8R9) n-;
each of k, m, and n is an integer selected from 0 to 10;
each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl; or
each - (CR8R9) -may independently form -C (O) -;
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10 membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-11 cycloalkylene;
wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;
wherein the point of bonding to the CRBN ligand is indicated by the asterisk; and U is a CRBN ligand.
One embodiment provides a compound, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (II) :
wherein,
R50 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl;
R51 and R52 are each independently hydrogen or optionally substituted straight or branched C1-6 alkyl; or
R51 and R52 are taken together to form -C (O) -;
represents a single or double bond;
R53, R54, R55, R56, R57, R58, and R59 are each independently hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;
each R7 is independently hydrogen or optionally substituted C1-6 alkyl; or
two R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring;
ring B is an optionally substituted C6-14cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;
L is -Ak1-Z1-Ak2-Z2-Ak3-*;
Ak1 is - (CR8R9) k-or -O-;
Ak2 is - (CR8R9) m-;
Ak3 is - (CR8R9) n-;
each of k, m, and n is an integer selected from 0 to 10;
R8 and R9 are each independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl; or
each - (CR8R9) -may independently form -C (O) -;
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-11 cycloalkylene;
wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;
wherein the point of bonding to the CRBN ligand is indicated by the asterisk; and U is a CRBN ligand.
One embodiment provides a compound, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (III) :
wherein,
R81 and R82 are each independently selected from hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;
each R83 is independently selected from halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;
R84 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl;
R85 and R86 are each independently hydrogen or optionally substituted straight or branched C1-6 alkyl; or
R85 and R86 are taken together to form -C (O) -;
represents a single or double bond;
W is CH, CR83, or N;
v is 0, 1, 2, or 3;
each R7 is independently hydrogen or optionally substituted C1-6 alkyl; or
two R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring;
ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;
L is -Ak1-Z1-Ak2-Z2-Ak3-*;
Ak1 is - (CR8R9) k-or -O-;
Ak2 is - (CR8R9) m-;
Ak3 is - (CR8R9) n-;
each of k, m, and n is an integer selected from 0 to 10;
each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl; or
each - (CR8R9) -may independently form -C (O) -;
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-11 cycloalkylene;
wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;
wherein the point of bonding to the CRBN ligand is indicated by the asterisk; and U is a CRBN ligand;
provided that whenrepresents a double bond, then R84 and R85 are absent.
One embodiment provides a compound, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (IV) :
wherein,
A1, A2, A3, and A4 are each independently CH or N;
Lcyc is -L1cyc-, -L1cyc-L2cyc-, -L1cyc-L2cyc-L3cyc-, or -L1cyc-L2cyc-L3cyc-L4cyc-;
L1cyc, L2cyc, L3cyc, and L4cyc are each independently bond, -O-, -S-, -NRy-, -C (O) -, -S (O) m1-, - (CRcRd) n1-, - (CHRc=CHRd) n1-, -C≡C-, -NRy (CRfRg) n2-, -O (CRcRd) n1O (CRfRg) n2-, -S (CRcRd) n1S (CRfRg) n2-, - (CRcRd) n1O (CRfRg) n2-, -O (CRcRd) n1NRy (CRfRg) n2-, -O (CRcRd) n1NRy (CRfRg) n2O-, - (CRcRd) n1S (CRfRg) n2-, - (CRcRd) n1 (CRfRg) n2-, - (CHRc=CHRd) n1 (CRfRg) n2-, - (CRcRd) n1 (C≡
C) (CRfRg) n2-, - (CRcRd) n1C (O) (CRfRg) n2-, - (CRcRd) n1S (O) m1 (CRfRg) n2-, - (CRcRd) n1NRyC (O) (CRfRg) n2-, - (CRcRd) n1NRyS (O) m1 (CRfRg) n2-, - (CRcRd) n1C (O) NRy (CRfRg) n2-, - (CRcRd) n1S (O) (=NRy) (CRfRg) n2-, - (CRcRd) n1N=S (=O) Ry (CRfRg) n2-, or - (CRcRd) n1S (O) m1NRy (CRfRg) n2-;
Rc, Rd, Ry, Rf, and Rg are each
independently hydrogen, halogen, -OH, -SH, nitro, cyano, -NH2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3 -12 cycloalkyl, optionally substituted 3-12 membered heterocyclyl, optionally substituted C6-14 aryl, or optionally substituted 5-14 membered heteroaryl; or
any two of Rc, Rd, Ry, Rf, and Rg are taken together with the atoms to which they are attached to form an optionally substituted C3-12 cycloalkyl, an optionally substituted 3-12 membered heterocyclyl, an optionally substituted C6-14 aryl group, or an optionally substituted 5 -14 membered heteroaryl;
n1, and n2 are each independently an integer selected from 0 to 10;
m1 is 0, 1, or 2;
ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;
L is -Ak1-Z1-Ak2-Z2-Ak3-*;
Ak1 is - (CR8R9) k-or -O-;
Ak2 is - (CR8R9) m-;
Ak3 is - (CR8R9) n-;
each of k, m, and n is an integer selected from 0 to 10;
each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl; or
each - (CR8R9) -may independently form -C (O) -;
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-11 cycloalkylene;
wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;
wherein the point of bonding to the CRBN ligand is indicated by the asterisk; and U is a CRBN ligand.
One embodiment provides a pharmaceutical composition comprising a compound of Formula (I) , Formula (II) , Formula (III) , or Formula (IV) , or pharmaceutically acceptable salt solvate, prodrug, stereoisomer, or tautomer thereof, and at least one pharmaceutically acceptable excipient.
One embodiment provides a compound of Formula (I) , Formula (II) , Formula (III) , or Formula (IV) , or pharmaceutically acceptable salt solvate, prodrug, stereoisomer, or tautomer thereof for use in a method of treatment of a human or animal.
One embodiment provides a compound of Formula (I) , Formula (II) , Formula (III) , or Formula (IV) , or pharmaceutically acceptable salt solvate, prodrug, stereoisomer, or tautomer thereof, for use in a method of treatment of cancer or neoplastic disease.
One embodiment provides the use of a compound of Formula (I) , Formula (II) , Formula (III) , or Formula (IV) , or pharmaceutically acceptable salt solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for the treatment of cancer or neoplastic disease.
One embodiment provides a method of treating cancer in a patient in need thereof comprising administering to the patient a compound of Formula (I) , Formula (II) , Formula (III) , or Formula (IV) , or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof.
One embodiment provides a method of treating a disease or disorder in a patient in need thereof comprising administering to the patient a compound of Formula (I) , Formula (II) , Formula (III) , or Formula (IV) , or pharmaceutically acceptable salt solvate, prodrug, stereoisomer, or tautomer thereof. Another embodiment provides the method wherein the disease or disorder is cancer.
One embodiment provides a method of treating cancer in a patient in need thereof comprising administering to the patient a pharmaceutical composition comprising a compound of Formula (I) , Formula (II) , Formula (III) , or Formula (IV) , or pharmaceutically acceptable salt solvate, prodrug, stereoisomer, or tautomer thereof, and a pharmaceutically acceptable excipient.
INCORPORATION BY REFERENCE
All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference for the specific purposes identified herein.
FIG. 1 depicts the protein levels of 3 targets (Wee1, GSPT1, CK1a) in TOV-21G cell line after treatment with exemplary compounds described herein.
FIG. 2 depicts the anti-tumor efficacy of WEE1 degraders in a SK-UT-1 CDX model.
FIG. 3 depicts the In-tumor WEE1 protein level detection. (24 h after 60 mpk PO at day 14; 12 h after 30 mpk BID at day 14) .
As used herein and in the appended claims, the singular forms "a, " "and, " and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "an agent" includes a plurality of such agents, and reference to "the cell" includes reference to one or more cells (or to a plurality of cells) and equivalents thereof known to those skilled in the art, and so forth. When ranges are used herein for physical properties, such as molecular weight, or chemical properties, such as chemical formulae, all combinations and subcombinations of ranges and specific embodiments therein are intended to be included. The term "about" when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error) , and thus the number or numerical range, in some instances, will vary between 1%and 15%of the stated number or numerical range. The term "comprising" (and related terms such as "comprise" or "comprises" or "having" or "including" ) is not intended to exclude that in other certain embodiments, for example, an embodiment of any composition of matter, composition, method, or process, or the like, described herein, "consist of" or "consist essentially of" the described features.
Definitions
As used in the specification and appended claims, unless specified to the contrary, the following terms have the meaning indicated below.
"Amino" refers to the –NH2 radical.
"Cyano" refers to the -CN radical.
"Nitro" refers to the -NO2 radical.
"Oxa" refers to the -O-radical.
"Oxo" refers to the =O radical.
"Thioxo" refers to the =S radical.
"Imino" refers to the =N-H radical.
"Oximo" refers to the =N-OH radical.
"Hydrazino" refers to the =N-NH2 radical.
"Alkyl" refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to fifteen carbon atoms (e.g., C1-C15 alkyl) . In certain embodiments, an alkyl comprises one to thirteen carbon
atoms (e.g., C1-C13 alkyl) . In certain embodiments, an alkyl comprises one to eight carbon atoms (e.g., C1-C8 alkyl) . In other embodiments, an alkyl comprises one to five carbon atoms (e.g., C1-C5 alkyl) . In other embodiments, an alkyl comprises one to four carbon atoms (e.g., C1-C4 alkyl) . In other embodiments, an alkyl comprises one to three carbon atoms (e.g., C1-C3 alkyl) . In other embodiments, an alkyl comprises one to two carbon atoms (e.g., C1-C2 alkyl) . In other embodiments, an alkyl comprises one carbon atom (e.g., C1 alkyl) . In other embodiments, an alkyl comprises five to fifteen carbon atoms (e.g., C5-C15 alkyl) . In other embodiments, an alkyl comprises five to eight carbon atoms (e.g., C5-C8 alkyl) . In other embodiments, an alkyl comprises two to five carbon atoms (e.g., C2-C5 alkyl) . In other embodiments, an alkyl comprises three to five carbon atoms (e.g., C3-C5 alkyl) . In other embodiments, the alkyl group is selected from methyl, ethyl, 1-propyl (n-propyl) , 1-methylethyl (iso-propyl) , 1-butyl (n-butyl) , 1-methylpropyl (sec-butyl) , 2-methylpropyl (iso-butyl) , 1, 1-dimethylethyl (tert-butyl) , 1-pentyl (n-pentyl) . The alkyl is attached to the rest of the molecule by a single bond. Unless stated otherwise specifically in the specification, an alkyl group is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -ORa, -SRa, -OC (O) -Ra, -N (Ra) 2, -C (O) Ra, -C (O) ORa, -C (O) N (Ra) 2, -N (Ra) C (O) ORa, -OC (O) -N (Ra) 2, -N (Ra) C (O) Ra, -N (Ra) S (O) tRa (where t is 1 or 2) , -S (O) tORa (where t is 1 or 2) , -S (O) tRa (where t is 1 or 2) and -S (O) tN (Ra) 2 (where t is 1 or 2) where each Ra is independently hydrogen, alkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , fluoroalkyl, carbocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , carbocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aralkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heteroaryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , or heteroarylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) .
"Alkoxy" refers to a radical bonded through an oxygen atom of the formula –O-alkyl, where alkyl is an alkyl chain as defined above.
"Alkenyl" refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one carbon-carbon double bond, and having from two to twelve carbon atoms. In certain embodiments, an alkenyl comprises two to eight carbon atoms. In other embodiments, an alkenyl comprises two to four carbon atoms. The alkenyl is attached to the rest of the molecule by a single bond, for example, ethenyl (i.e., vinyl) ,
prop-1-enyl (i.e., allyl) , but-1-enyl, pent-1-enyl, penta-1, 4-dienyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -ORa, -SRa, -OC (O) -Ra, -N (Ra) 2, -C (O) Ra, -C (O) ORa, -C (O) N (Ra) 2, -N (Ra) C (O) ORa, -OC (O) -N (Ra) 2, -N (Ra) C (O) Ra, -N (Ra) S (O) tRa (where t is 1 or 2) , -S (O) tORa (where t is 1 or 2) , -S (O) tRa (where t is 1 or 2) and -S (O) tN (Ra) 2 (where t is 1 or 2) where each Ra is independently hydrogen, alkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , fluoroalkyl, carbocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , carbocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aralkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heteroaryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , or heteroarylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) .
"Alkynyl" refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one carbon-carbon triple bond, having from two to twelve carbon atoms. In certain embodiments, an alkynyl comprises two to eight carbon atoms. In other embodiments, an alkynyl comprises two to six carbon atoms. In other embodiments, an alkynyl comprises two to four carbon atoms. The alkynyl is attached to the rest of the molecule by a single bond, for example, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkynyl group is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -ORa, -SRa, -OC (O) -Ra, -N (Ra) 2, -C (O) Ra, -C (O) ORa, -C (O) N (Ra) 2, -N (Ra) C (O) ORa, -OC (O) -N (Ra) 2, -N (Ra) C (O) Ra, -N (Ra) S (O) tRa (where t is 1 or 2) , -S (O) tORa (where t is 1 or 2) , -S (O) tRa (where t is 1 or 2) and -S (O) tN (Ra) 2 (where t is 1 or 2) where each Ra is independently hydrogen, alkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , fluoroalkyl, carbocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , carbocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aralkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heteroaryl (optionally substituted with halogen, hydroxy, methoxy, or
trifluoromethyl) , or heteroarylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) .
"Alkylene" or "alkylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation and having from one to twelve carbon atoms, for example, methylene, ethylene, propylene, n-butylene, and the like. The alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group are through one carbon in the alkylene chain or through any two carbons within the chain. In certain embodiments, an alkylene comprises one to eight carbon atoms (e.g., C1-C8 alkylene) . In other embodiments, an alkylene comprises one to five carbon atoms (e.g., C1-C5 alkylene) . In other embodiments, an alkylene comprises one to four carbon atoms (e.g., C1-C4 alkylene) . In other embodiments, an alkylene comprises one to three carbon atoms (e.g., C1-C3 alkylene) . In other embodiments, an alkylene comprises one to two carbon atoms (e.g., C1-C2 alkylene) . In other embodiments, an alkylene comprises one carbon atom (e.g., C1 alkylene) . In other embodiments, an alkylene comprises five to eight carbon atoms (e.g., C5-C8 alkylene) . In other embodiments, an alkylene comprises two to five carbon atoms (e.g., C2-C5 alkylene) . In other embodiments, an alkylene comprises three to five carbon atoms (e.g., C3-C5 alkylene) . Unless stated otherwise specifically in the specification, an alkylene chain is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -ORa, -SRa, -OC (O) -Ra, -N (Ra) 2, -C (O) Ra, -C (O) ORa, -C (O) N (Ra) 2, -N (Ra) C (O) ORa, -OC (O) -N (Ra) 2, -N (Ra) C (O) Ra, -N (Ra) S (O) tRa (where t is 1 or 2) , -S (O) tORa (where t is 1 or 2) , -S (O) tRa (where t is 1 or 2) and -S (O) tN (Ra) 2 (where t is 1 or 2) where each Ra is independently hydrogen, alkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , fluoroalkyl, carbocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , carbocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aralkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heteroaryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , or heteroarylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) .
"Alkenylene" or "alkenylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one carbon-carbon double bond, and having from two to twelve
carbon atoms. The alkenylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. In certain embodiments, an alkenylene comprises two to eight carbon atoms (e.g., C2-C8 alkenylene) . In other embodiments, an alkenylene comprises two to five carbon atoms (e.g., C2-C5 alkenylene) . In other embodiments, an alkenylene comprises two to four carbon atoms (e.g., C2-C4 alkenylene) . In other embodiments, an alkenylene comprises two to three carbon atoms (e.g., C2-C3 alkenylene) . In other embodiments, an alkenylene comprises two carbon atoms (e.g., C2 alkenylene) . In other embodiments, an alkenylene comprises five to eight carbon atoms (e.g., C5-C8 alkenylene) . In other embodiments, an alkenylene comprises three to five carbon atoms (e.g., C3-C5 alkenylene) . Unless stated otherwise specifically in the specification, an alkenylene chain is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -ORa, -SRa, -OC (O) -Ra, -N (Ra) 2, -C (O) Ra, -C (O) ORa, -C (O) N (Ra) 2, -N (Ra) C (O) ORa, -OC (O) -N (Ra) 2, -N (Ra) C (O) Ra, -N (Ra) S (O) tRa (where t is 1 or 2) , -S (O) tORa (where t is 1 or 2) , -S (O) tRa (where t is 1 or 2) and -S (O) tN (Ra) 2 (where t is 1 or 2) where each Ra is independently hydrogen, alkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , fluoroalkyl, carbocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , carbocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aralkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heteroaryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , or heteroarylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) .
"Alkynylene" or "alkynylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one carbon-carbon triple bond, and having from two to twelve carbon atoms. The alkynylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. In certain embodiments, an alkynylene comprises two to eight carbon atoms (e.g., C2-C8 alkynylene) . In other embodiments, an alkynylene comprises two to five carbon atoms (e.g., C2-C5 alkynylene) . In other embodiments, an alkynylene comprises two to four carbon atoms (e.g., C2-C4 alkynylene) . In other embodiments, an alkynylene comprises two to three carbon atoms (e.g., C2-C3 alkynylene) . In other embodiments, an alkynylene comprises two carbon atoms (e.g., C2 alkynylene) . In other embodiments, an alkynylene comprises five to eight carbon atoms (e.g., C5-C8 alkynylene) . In
other embodiments, an alkynylene comprises three to five carbon atoms (e.g., C3-C5 alkynylene) . Unless stated otherwise specifically in the specification, an alkynylene chain is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -ORa, -SRa, -OC (O) -Ra, -N (Ra) 2, -C (O) Ra, -C (O) ORa, -C (O) N (Ra) 2, -N (Ra) C (O) ORa, -OC (O) -N (Ra) 2, -N (Ra) C (O) Ra, -N (Ra) S (O) tRa (where t is 1 or 2) , -S (O) tORa (where t is 1 or 2) , -S (O) tRa (where t is 1 or 2) and -S (O) tN (Ra) 2 (where t is 1 or 2) where each Ra is independently hydrogen, alkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , fluoroalkyl, carbocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , carbocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aralkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heteroaryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , or heteroarylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) .
"Aryl" refers to a radical derived from an aromatic monocyclic or multicyclic hydrocarbon ring system by removing a hydrogen atom from a ring carbon atom. The aromatic monocyclic or multicyclic hydrocarbon ring system contains only hydrogen and carbon from five to eighteen carbon atoms, where at least one of the rings in the ring system is fully unsaturated, i.e., it contains a cyclic, delocalized (4n+2) π–electron system in accordance with the Hückel theory. The ring system from which aryl groups are derived include, but are not limited to, groups such as benzene, fluorene, indane, indene, tetralin and naphthalene. Unless stated otherwise specifically in the specification, the term "aryl" or the prefix "ar-" (such as in "aralkyl" ) is meant to include aryl radicals optionally substituted by one or more substituents independently selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -Rb-ORa, -Rb-OC (O) -Ra, -Rb-OC (O) -ORa, -Rb-OC (O) -N (Ra) 2, -Rb-N (Ra) 2, -Rb-C (O) Ra, -Rb-C (O) ORa, -Rb-C (O) N (Ra) 2, -Rb-O-Rc-C (O) N (Ra) 2, -Rb-N (Ra) C (O) ORa, -Rb-N (Ra) C (O) Ra, -Rb-N (Ra) S (O) tRa (where t is 1 or 2) , -Rb-S (O) tRa (where t is 1 or 2) , -Rb-S (O) tORa (where t is 1 or 2) and -Rb-S (O) tN (Ra) 2 (where t is 1 or 2) , where each Ra is independently hydrogen, alkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , fluoroalkyl, cycloalkyl (optionally substituted with
halogen, hydroxy, methoxy, or trifluoromethyl) , cycloalkylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aralkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heteroaryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , or heteroarylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , each Rb is independently a direct bond or a straight or branched alkylene or alkenylene chain, and Rc is a straight or branched alkylene or alkenylene chain, and where each of the above substituents is unsubstituted unless otherwise indicated.
"Aralkyl" refers to a radical of the formula -Rc-aryl where Rc is an alkylene chain as defined above, for example, methylene, ethylene, and the like. The alkylene chain part of the aralkyl radical is optionally substituted as described above for an alkylene chain. The aryl part of the aralkyl radical is optionally substituted as described above for an aryl group.
"Aralkenyl" refers to a radical of the formula –Rd-aryl where Rd is an alkenylene chain as defined above. The aryl part of the aralkenyl radical is optionally substituted as described above for an aryl group. The alkenylene chain part of the aralkenyl radical is optionally substituted as defined above for an alkenylene group.
"Aralkynyl" refers to a radical of the formula -Re-aryl, where Re is an alkynylene chain as defined above. The aryl part of the aralkynyl radical is optionally substituted as described above for an aryl group. The alkynylene chain part of the aralkynyl radical is optionally substituted as defined above for an alkynylene chain.
"Aralkoxy" refers to a radical bonded through an oxygen atom of the formula -O-Rc-aryl where Rc is an alkylene chain as defined above, for example, methylene, ethylene, and the like. The alkylene chain part of the aralkyl radical is optionally substituted as described above for an alkylene chain. The aryl part of the aralkyl radical is optionally substituted as described above for an aryl group.
"Carbocyclyl" refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which includes fused, bridged, or spirocyclic ring systems, having from three to fifteen carbon atoms. In certain embodiments, a carbocyclyl comprises three to ten carbon atoms. In other embodiments, a carbocyclyl comprises five to seven carbon atoms. The carbocyclyl is attached to the rest of the molecule by a single bond. Carbocyclyl is saturated (i.e., containing single C-C bonds only) or unsaturated (i.e., containing one or more double bonds or triple bonds) . A fully saturated carbocyclyl radical is also referred to as "cycloalkyl. " Examples of monocyclic cycloalkyls include, e.g., cyclopropyl,
cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. An unsaturated carbocyclyl is also referred to as "cycloalkenyl. " Examples of monocyclic cycloalkenyls include, e.g., cyclopentenyl, cyclohexenyl, cycloheptenyl, and cyclooctenyl. Polycyclic carbocyclyl radicals include, for example, adamantyl, norbornyl (i.e., bicyclo [2.2.1] heptanyl) , norbornenyl, decalinyl, 7, 7-dimethyl-bicyclo [2.2.1] heptanyl, and the like. Spirocyclic carbocyclyl or cycloalkyl radicals include, for example, spiro [2.2] pentane, spiro [2.3] hexane, spiro [2.4] heptane, spiro [2.5] octane, spiro [2.6] nonane, spiro [3.3] heptane, spiro [3.4] octane, spiro [3.5] nonane, spiro [3.6] decane, spiro [4.4] nonane, spiro [4.5] decane, spiro [4.6] undecane, spiro [5.5] undecane, spiro [5.6] dodecane, spiro [6.6] tridecane, and the like. Unless otherwise stated specifically in the specification, the term "carbocyclyl" is meant to include carbocyclyl radicals that are optionally substituted by one or more substituents independently selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, oxo, thioxo, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -Rb-ORa, -Rb-OC (O) -Ra, -Rb-OC (O) -ORa, -Rb-OC (O) -N (Ra) 2, -Rb-N (Ra) 2, -Rb-C (O) Ra, -Rb-C (O) ORa, -Rb-C (O) N (Ra) 2, -Rb-O-Rc-C (O) N (Ra) 2, -Rb-N (Ra) C (O) ORa, -Rb-N (Ra) C (O) Ra, -Rb-N (Ra) S (O) tRa (where t is 1 or 2) , -Rb-S (O) tRa (where t is 1 or 2) , -Rb-S (O) tORa (where t is 1 or 2) and -Rb-S (O) tN (Ra) 2 (where t is 1 or 2) , where each Ra is independently hydrogen, alkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , fluoroalkyl, cycloalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , cycloalkylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aralkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heteroaryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , or heteroarylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , each Rb is independently a direct bond or a straight or branched alkylene or alkenylene chain, and Rc is a straight or branched alkylene or alkenylene chain, and where each of the above substituents is unsubstituted unless otherwise indicated.
“Cycloalkylene” refers to a divalent carbocyclyl or cycloalkyl linking the rest of the molecule to a radical group.
"Carbocyclylalkyl" refers to a radical of the formula –Rc-carbocyclyl where Rc is an alkylene chain as defined above. The alkylene chain and the carbocyclyl radical is optionally substituted as defined above.
"Carbocyclylalkynyl" refers to a radical of the formula –Rc-carbocyclyl where Rc is an alkynylene chain as defined above. The alkynylene chain and the carbocyclyl radical is optionally substituted as defined above.
"Carbocyclylalkoxy" refers to a radical bonded through an oxygen atom of the formula –O-Rc-carbocyclyl where Rc is an alkylene chain as defined above. The alkylene chain and the carbocyclyl radical is optionally substituted as defined above.
As used herein, “carboxylic acid bioisostere” refers to a functional group or moiety that exhibits similar physical, biological and/or chemical properties as a carboxylic acid moiety. Examples of carboxylic acid bioisosteres include, but are not limited to,
and the like.
and the like.
"Halo" or "halogen" refers to bromo, chloro, fluoro or iodo substituents.
"Fluoroalkyl" refers to an alkyl radical, as defined above, that is substituted by one or more fluoro radicals, as defined above, for example, trifluoromethyl, difluoromethyl, fluoromethyl, 2, 2, 2-trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, and the like. In some embodiments, the alkyl part of the fluoroalkyl radical is optionally substituted as defined above for an alkyl group.
“Heteroalkyl” refers to an alkyl group in which one or more skeletal atoms of the alkyl are selected from an atom other than carbon, e.g., oxygen, nitrogen (e.g., -NH-, -N (alkyl) -) , sulfur, or combinations thereof. A heteroalkyl is attached to the rest of the molecule at a carbon atom of the heteroalkyl. In one aspect, a heteroalkyl is a C1-C6 heteroalkyl wherein the heteroalkyl is comprised of 1 to 6 carbon atoms and one or more atoms other than carbon, e.g., oxygen, nitrogen (e.g. -NH-, -N (alkyl) -) , sulfur, or combinations thereof wherein the heteroalkyl is attached to the rest of the molecule at a carbon atom of the heteroalkyl. Examples of such heteroalkyl are, for example, –CH2-O-CH2-, –CH2-N (alkyl) -CH2-, –CH2-N (aryl) -CH2-, -OCH2CH2O-, –OCH2CH2OCH2CH2O-, or –OCH2CH2OCH2CH2OCH2CH2O-. Unless stated otherwise specifically in the specification, a heteroalkyl is optionally substituted for example, with oxo, halogen, amino, nitrile, nitro, hydroxyl, alkyl, alkenyl, alkynyl, haloalkyl, alkoxy, aryl,
cycloalkyl, heterocycloalkyl, heteroaryl, and the like. In some embodiments, a heteroalkyl is optionally substituted with oxo, halogen, methyl, ethyl, -CN, -CF3, -OH, -OMe, -NH2, or -NO2. In some embodiments, a heteroalkyl is optionally substituted with oxo, halogen, methyl, ethyl, -CN, -CF3, -OH, or -OMe. In some embodiments, the heteroalkyl is optionally substituted with halogen.
“Heteroalkylene” refers to a divalent heteroalkyl radical.
"Heterocyclyl" refers to a stable 3-to 18-membered non-aromatic ring radical that comprises two to twelve carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur. Unless stated otherwise specifically in the specification, the heterocyclyl radical is a monocyclic, bicyclic, tricyclic, tetracyclic, or spirocyclic ring system, which optionally includes fused or bridged ring systems. The heteroatoms in the heterocyclyl radical are optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heterocyclyl radical is partially or fully saturated. The heterocyclyl is attached to the rest of the molecule through any atom of the ring (s) . Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl [1, 3] dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1, 1-dioxo-thiomorpholinyl, 1-azaspiro [2.2] pentane, 1-azaspiro [2.3] hexane, 1-azaspiro [2.4] heptane, 1-azaspiro [2.5] octane, 1-azaspiro [2.6] nonane, 2-azaspiro [3.3] heptane, 2-azaspiro [3.4] octane, 2-azaspiro [3.5] nonane, 2-azaspiro [3.6] decane, 2-azaspiro [4.4] nonane, 2-azaspiro [4.5] decane, 2-azaspiro [4.6] undecane, 3-azaspiro [5.5] undecane, 3-azaspiro [5.6] dodecane, 3-azaspiro [6.6] tridecane, 5-azaspiro [2.3] hexane, 5-azaspiro [2.4] heptane, 6-azaspiro [2.5] octane, 6-azaspiro [2.6] nonane, 6-azaspiro [3.4] octane, 7-azaspiro [3.5] nonane, 7-azaspiro [3.6] decane, 8-azaspiro [4.5] decane, 8-azaspiro [4.6] undecane, 9-azaspiro [5.6] dodecane, 1, 4-diazaspiro [2.2] pentane, 1, 5-diazaspiro [2.3] hexane, 1, 5-diazaspiro [2.4] heptane, 1, 6-diazaspiro [2.5] octane, 1, 6-diazaspiro [2.6] nonane, 2, 6-diazaspiro [3.3] heptane, 2, 6-diazaspiro [3.4] octane, 2, 7-diazaspiro [3.5] nonane, 2, 7-diazaspiro [3.6] decane, 2, 7-diazaspiro [4.4] nonane, 2, 8-diazaspiro [4.5] decane, 2, 8-diazaspiro [4.6] undecane, 3, 9-diazaspiro [5.5] undecane, 3, 9-diazaspiro [5.6] dodecane, and 3, 10-diazaspiro [6.6] tridecane. Unless stated otherwise specifically in the specification, the term "heterocyclyl" is meant to include heterocyclyl radicals as defined above that are optionally substituted by one or more substituents selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, oxo, thioxo, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally
substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -Rb-ORa, -Rb-OC (O) -Ra, -Rb-OC (O) -ORa, -Rb-OC (O) -N (Ra) 2, -Rb-N (Ra) 2, -Rb-C (O) Ra, -Rb-C (O) ORa, -Rb-C (O) N (Ra) 2, -Rb-O-Rc-C (O) N (Ra) 2, -Rb-N (Ra) C (O) ORa, -Rb-N (Ra) C (O) Ra, -Rb-N (Ra) S (O) tRa (where t is 1 or 2) , -Rb-S (O) tRa (where t is 1 or 2) , -Rb-S (O) tORa (where t is 1 or 2) and -Rb-S (O) tN (Ra) 2 (where t is 1 or 2) , where each Ra is independently hydrogen, alkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , fluoroalkyl, cycloalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , cycloalkylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aralkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heteroaryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , or heteroarylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , each Rb is independently a direct bond or a straight or branched alkylene or alkenylene chain, and Rc is a straight or branched alkylene or alkenylene chain, and where each of the above substituents is unsubstituted unless otherwise indicated.
“Heterocyclene” refers to a divalent heterocyclyl linking the rest of the molecule to a radical group.
"N-heterocyclyl" or “N-attached heterocyclyl” refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical. An N-heterocyclyl radical is optionally substituted as described above for heterocyclyl radicals. Examples of such N-heterocyclyl radicals include, but are not limited to, 1-morpholinyl, 1-piperidinyl, 1-piperazinyl, 1-pyrrolidinyl, pyrazolidinyl, imidazolinyl, and imidazolidinyl.
"C-heterocyclyl" or “C-attached heterocyclyl” refers to a heterocyclyl radical as defined above containing at least one heteroatom and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a carbon atom in the heterocyclyl radical. A C-heterocyclyl radical is optionally substituted as described above for heterocyclyl radicals. Examples of such C-heterocyclyl radicals include, but are not limited to, 2-morpholinyl, 2-or 3-or 4-piperidinyl, 2-piperazinyl, 2-or 3-pyrrolidinyl, and the like.
"Heterocyclylalkyl" refers to a radical of the formula –Rc-heterocyclyl where Rc is an alkylene chain as defined above. If the heterocyclyl is a nitrogen-containing heterocyclyl, the
heterocyclyl is optionally attached to the alkyl radical at the nitrogen atom. The alkylene chain of the heterocyclylalkyl radical is optionally substituted as defined above for an alkylene chain. The heterocyclyl part of the heterocyclylalkyl radical is optionally substituted as defined above for a heterocyclyl group.
"Heterocyclylalkoxy" refers to a radical bonded through an oxygen atom of the formula –O-Rc-heterocyclyl where Rc is an alkylene chain as defined above. If the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl is optionally attached to the alkyl radical at the nitrogen atom. The alkylene chain of the heterocyclylalkoxy radical is optionally substituted as defined above for an alkylene chain. The heterocyclyl part of the heterocyclylalkoxy radical is optionally substituted as defined above for a heterocyclyl group.
"Heteroaryl" refers to a radical derived from a 3-to 18-membered aromatic ring radical that comprises two to seventeen carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur. As used herein, the heteroaryl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, wherein at least one of the rings in the ring system is fully unsaturated, i.e., it contains a cyclic, delocalized (4n+2) π–electron system in accordance with the Hückel theory. Heteroaryl includes fused or bridged ring systems. The heteroatom (s) in the heteroaryl radical is optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heteroaryl is attached to the rest of the molecule through any atom of the ring (s) . Examples of heteroaryls include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzindolyl, 1, 3-benzodioxolyl, benzofuranyl, benzooxazolyl, benzo [d] thiazolyl, benzothiadiazolyl, benzo [b] [1, 4] dioxepinyl, benzo [b] [1, 4] oxazinyl, 1, 4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl) , benzothieno [3, 2-d] pyrimidinyl, benzotriazolyl, benzo [4, 6] imidazo [1, 2-a] pyridinyl, carbazolyl, cinnolinyl, cyclopenta [d] pyrimidinyl, 6, 7-dihydro-5H-cyclopenta [4, 5] thieno [2, 3-d] pyrimidinyl, 5, 6-dihydrobenzo [h] quinazolinyl, 5, 6-dihydrobenzo [h] cinnolinyl, 6, 7-dihydro-5H-benzo [6, 7] cyclohepta [1, 2-c] pyridazinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, furo [3, 2-c] pyridinyl, 5, 6, 7, 8, 9, 10-hexahydrocycloocta [d] pyrimidinyl, 5, 6, 7, 8, 9, 10-hexahydrocycloocta [d] pyridazinyl, 5, 6, 7, 8, 9, 10-hexahydrocycloocta [d] pyridinyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, 5, 8-methano-5, 6, 7, 8-tetrahydroquinazolinyl, naphthyridinyl, 1, 6-naphthyridinonyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 5, 6, 6a, 7, 8, 9, 10, 10a-octahydrobenzo [h] quinazolinyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl, pyrazolo [3, 4-d] pyrimidinyl, pyridinyl, pyrido [3, 2-d] pyrimidinyl, pyrido [3, 4-d] pyrimidinyl,
pyrazinyl, pyrimidinyl, pyridazinyl, pyrrolyl, quinazolinyl, quinoxalinyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, 5, 6, 7, 8-tetrahydroquinazolinyl, 5, 6, 7, 8-tetrahydrobenzo [4, 5] thieno [2, 3-d] pyrimidinyl, 6, 7, 8, 9-tetrahydro-5H-cyclohepta [4, 5] thieno [2, 3-d] pyrimidinyl, 5, 6, 7, 8-tetrahydropyrido [4, 5-c] pyridazinyl, 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, thieno [2, 3-d] pyrimidinyl, thieno [3, 2-d] pyrimidinyl, thieno [2, 3-c] pridinyl, and thiophenyl (i.e. thienyl) . Unless stated otherwise specifically in the specification, the term "heteroaryl" is meant to include heteroaryl radicals as defined above which are optionally substituted by one or more substituents selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, haloalkenyl, haloalkynyl, oxo, thioxo, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -Rb-ORa, -Rb-OC (O) -Ra, -Rb-OC (O) -ORa, -Rb-OC (O) -N (Ra) 2, -Rb-N (Ra) 2, -Rb-C (O) Ra, -Rb-C (O) ORa, -Rb-C (O) N (Ra) 2, -Rb-O-Rc-C (O) N (Ra) 2, -Rb-N (Ra) C (O) ORa, -Rb-N (Ra) C (O) Ra, -Rb-N (Ra) S (O) tRa (where t is 1 or 2) , -Rb-S (O) tRa (where t is 1 or 2) , -Rb-S (O) tORa (where t is 1 or 2) and -Rb-S (O) tN (Ra) 2 (where t is 1 or 2) , where each Ra is independently hydrogen, alkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , fluoroalkyl, cycloalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , cycloalkylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , aralkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heterocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , heteroaryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , or heteroarylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl) , each Rb is independently a direct bond or a straight or branched alkylene or alkenylene chain, and Rc is a straight or branched alkylene or alkenylene chain, and where each of the above substituents is unsubstituted unless otherwise indicated.
"Heteroarylene" refers to a divalent heteroaryl radical.
"N-heteroaryl" refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical. An N-heteroaryl radical is optionally substituted as described above for heteroaryl radicals.
"C-heteroaryl" refers to a heteroaryl radical as defined above and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a carbon atom in the heteroaryl radical. A C-heteroaryl radical is optionally substituted as described above for heteroaryl radicals.
"Heteroarylalkyl" refers to a radical of the formula –Rc-heteroaryl, where Rc is an alkylene chain as defined above. If the heteroaryl is a nitrogen-containing heteroaryl, the heteroaryl is optionally attached to the alkyl radical at the nitrogen atom. The alkylene chain of the heteroarylalkyl radical is optionally substituted as defined above for an alkylene chain. The heteroaryl part of the heteroarylalkyl radical is optionally substituted as defined above for a heteroaryl group.
"Heteroarylalkoxy" refers to a radical bonded through an oxygen atom of the formula –O-Rc-heteroaryl, where Rc is an alkylene chain as defined above. If the heteroaryl is a nitrogen-containing heteroaryl, the heteroaryl is optionally attached to the alkyl radical at the nitrogen atom. The alkylene chain of the heteroarylalkoxy radical is optionally substituted as defined above for an alkylene chain. The heteroaryl part of the heteroarylalkoxy radical is optionally substituted as defined above for a heteroaryl group.
The compounds disclosed herein, in some embodiments, contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that are defined, in terms of absolute stereochemistry, as (R) -or (S) -. Unless stated otherwise, it is intended that all stereoisomeric forms of the compounds disclosed herein are contemplated by this disclosure. The term “positional isomer” refers to structural isomers around a central ring, such as ortho-, meta-, and para-isomers around a benzene ring.
A "tautomer" refers to a molecule wherein a proton shift from one atom of a molecule to another atom of the same molecule is possible. The compounds presented herein, in certain embodiments, exist as tautomers. In circumstances where tautomerization is possible, a chemical equilibrium of the tautomers will exist. The exact ratio of the tautomers depends on several factors, including physical state, temperature, solvent, and pH. Some examples of tautomeric equilibrium include:
The compounds disclosed herein, in some embodiments, are used in different enriched isotopic forms, e.g., enriched in the content of 2H, 3H, 11C, 13C and/or 14C. In one particular embodiment, the compound is deuterated in at least one position. Such deuterated forms can be made by the procedure described in U.S. Patent Nos. 5,846,514 and 6,334,997. As described in U.S. Patent Nos. 5,846,514 and 6,334,997, deuteration can improve the metabolic stability and or efficacy, thus increasing the duration of action of drugs.
Unless otherwise stated, structures depicted herein are intended to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13C-or 14C-enriched carbon are within the scope of the present disclosure.
The compounds of the present disclosure optionally contain unnatural proportions of atomic isotopes at one or more atoms that constitute such compounds. For example, the compounds may be labeled with isotopes, such as for example, deuterium (2H) , tritium (3H) , iodine-125 (125I) or carbon-14 (14C) . Isotopic substitution with 2H, 11C, 13C, 14C, 15C, 12N, 13N, 15N, 16N, 16O, 17O, 14F, 15F, 16F, 17F, 18F, 33S, 34S, 35S, 36S, 35Cl, 37Cl, 79Br, 81Br, 125I are all contemplated. In some embodiments, isotopic substitution with 18F is contemplated. All isotopic variations of the compounds of the present invention, whether radioactive or not, are encompassed within the scope of the present invention.
In certain embodiments, the compounds disclosed herein have some or all of the 1H atoms replaced with 2H atoms. The methods of synthesis for deuterium-containing compounds are known in the art and include, by way of non-limiting example only, the following synthetic methods.
Deuterium substituted compounds are synthesized using various methods such as described in: Dean, Dennis C.; Editor. Recent Advances in the Synthesis and Applications of Radiolabeled Compounds for Drug Discovery and Development. [Curr., Pharm. Des., 2000; 6 (10) ] 2000, 110 pp; George W.; Varma, Rajender S. The Synthesis of Radiolabeled Compounds via Organometallic Intermediates, Tetrahedron, 1989, 45 (21) , 6601-21; and Evans, E. Anthony. Synthesis of radiolabeled compounds, J. Radioanal. Chem., 1981, 64 (1-2) , 9-32.
Deuterated starting materials are readily available and are subjected to the synthetic methods described herein to provide for the synthesis of deuterium-containing compounds. Large numbers of deuterium-containing reagents and building blocks are available commercially from chemical vendors, such as Aldrich Chemical Co.
Deuterium-transfer reagents suitable for use in nucleophilic substitution reactions, such as iodomethane-d3 (CD3I) , are readily available and may be employed to transfer a deuterium-substituted carbon atom under nucleophilic substitution reaction conditions to the reaction substrate. The use of CD3I is illustrated, by way of example only, in the reaction schemes below.
Deuterium-transfer reagents, such as lithium aluminum deuteride (LiAlD4) , are employed to transfer deuterium under reducing conditions to the reaction substrate. The use of LiAlD4 is illustrated, by way of example only, in the reaction schemes below.
Deuterium gas and palladium catalyst are employed to reduce unsaturated carbon-carbon linkages and to perform a reductive substitution of aryl carbon-halogen bonds as illustrated, by way of example only, in the reaction schemes below.
In one embodiment, the compounds disclosed herein contain one deuterium atom. In another embodiment, the compounds disclosed herein contain two deuterium atoms. In another
embodiment, the compounds disclosed herein contain three deuterium atoms. In another embodiment, the compounds disclosed herein contain four deuterium atoms. In another embodiment, the compounds disclosed herein contain five deuterium atoms. In another embodiment, the compounds disclosed herein contain six deuterium atoms. In another embodiment, the compounds disclosed herein contain more than six deuterium atoms. In another embodiment, the compound disclosed herein is fully substituted with deuterium atoms and contains no non-exchangeable 1H hydrogen atoms. In one embodiment, the level of deuterium incorporation is determined by synthetic methods in which a deuterated synthetic building block is used as a starting material.
"Pharmaceutically acceptable salt" includes both acid and base addition salts. A pharmaceutically acceptable salt of any one of the heteroaromatic WEE1 inhibitory compounds described herein is intended to encompass any and all pharmaceutically suitable salt forms. Preferred pharmaceutically acceptable salts of the compounds described herein are pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
"Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, hydroiodic acid, hydrofluoric acid, phosphorous acid, and the like. Also included are salts that are formed with organic acids such as aliphatic mono-and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and. aromatic sulfonic acids, etc. and include, for example, acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like. Exemplary salts thus include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, nitrates, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, trifluoroacetates, propionates, caprylates, isobutyrates, oxalates, malonates, succinate suberates, sebacates, fumarates, maleates, mandelates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, phthalates, benzenesulfonates, toluenesulfonates, phenylacetates, citrates, lactates, malates, tartrates, methanesulfonates, and the like. Also contemplated are salts of amino acids, such as arginates, gluconates, and galacturonates (see, for example, Berge S.M. et al., "Pharmaceutical Salts, " Journal of Pharmaceutical Science, 66: 1-19 (1997) ) . Acid addition salts of basic compounds are, in some embodiments, prepared by contacting
the free base forms with a sufficient amount of the desired acid to produce the salt according to methods and techniques with which a skilled artisan is familiar.
"Pharmaceutically acceptable base addition salt" refers to those salts that retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Pharmaceutically acceptable base addition salts are, in some embodiments, formed with metals or amines, such as alkali and alkaline earth metals or organic amines. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, for example, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, N, N-dibenzylethylenediamine, chloroprocaine, hydrabamine, choline, betaine, ethylenediamine, ethylenedianiline, N-methylglucamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like. See Berge et al., supra.
"Pharmaceutically acceptable solvate" refers to a composition of matter that is the solvent addition form. In some embodiments, solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and are formed during the process of making with pharmaceutically acceptable solvents such as water, ethanol, and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Solvates of compounds described herein are conveniently prepared or formed during the processes described herein. The compounds provided herein optionally exist in either unsolvated as well as solvated forms.
The term “subject” or “patient” encompasses mammals. Examples of mammals include, but are not limited to, any member of the Mammalian class: humans, non-human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like. In one aspect, the mammal is a human.
"Prodrug" refers to a compound that undergoes biotransformation before exhibiting its pharmacological effects. Prodrugs can thus be viewed as drugs containing specialized protective groups used in a transient manner to alter pharmacological properties in the parent molecule.
As used herein, “treatment” or “treating, ” or “palliating” or “ameliorating” are used interchangeably. These terms refer to an approach for obtaining beneficial or desired results
including but not limited to therapeutic benefit and/or a prophylactic benefit. By “therapeutic benefit” is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient is still afflicted with the underlying disorder. For prophylactic benefit, the compositions are, in some embodiments, administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease has not been made.
WEE1 Kinase
WEE1 is a kinase belonging to the Ser/Thr family of protein kinases and is a key regulator of cell cycle progression. The WEE1 kinase is encoded by the WEE1 gene. WEE1 is an important regulator of the G2/M cell cycle checkpoint. WEE1 mediates cell-cycle arrest by regulating the phosphorylation of cyclin-dependent kinase 1 (Cdk1) . WEE1 inhibits Cdk1 by phosphorylating it on two different sites, Tyr15 and Thr14. Phosphorylated Cdk1 has reduced kinase activity and therefore prevents entry into mitosis. As a cell progress through the cell cycle, WEE1 is inactivated and degraded, and the cell is allowed to enter mitosis. DNA damage caused by, for example, genomic instability, errors in DNA synthesis, or DNA damaging agents, activates p53 and reduces WEE1 degradation thereby arresting the cell cycle and preventing progression into mitosis. Cancer cells are often deficient in p53 signaling, display genomic instability, and rely on the WEE1 and G2/M cell cycle checkpoint to avoid mitotic catastrophe due to DNA damage. Inhibition or degradation of WEE1 sensitizes cancers to DNA-damaging therapies. Additional information may be found in a) LL Parker, and H Piwnica-Worms. Inactivation of the p34cdc2-cyclin B complex by the human WEE1 tyrosine kinase. Science. 1992 Sep 25; 257 (5078) : 1955-1957, b) N Watanabe, M Broome, and T Hunter. Regulation of the human WEE1Hu CDK tyrosine 15-kinase during the cell cycle. EMBO J. 1995 May 1; 14 (9) : 1878-1891, c) CH McGowan, and P Russell. Cell cycle regulation of human WEE1. EMBO J. 1995 May 15; 14 (10) : 2166-2175, and d) Philip C De Witt Hamer, Shahryar E Mir, David Noske, Cornelis J F Van Noorden, Tom Wurdinger. WEE1 kinase targeting combined with DNA-damaging cancer therapy catalyzes mitotic catastrophe. Clin Cancer Res. 1022 Jul 1; 17 (13) , 4200–4207, each of which is incorporated herein by reference.
Selective Protein Degradation
The levels of proteins within a cell are determined by both the rate of protein synthesis and the rate of protein degradation. In eukaryotic cells two pathways exist for selective protein degradation, the ubiquitin-proteasome pathway and the lysosomal proteolysis pathway. In general, selective protein degradation is mediated by the presence of a recruitment motif which
promotes binding of degradation proteins, such as proteasomal degradation proteins, or proteins associated with ubiquitin-proteasome pathway. Recruitment motifs include E3 ligase recognition agents and proteasome recognition agents. Conjugation of recruitment motifs with high affinity ligands for WEE1 kinase provides compounds capable of selectively directing pathways for protein degradation to the WEE1 kinase protein itself. Such an outcome will reduce levels of WEE1 kinase activity.
Heteroaromatic WEE1 degradation compounds
In one aspect, provided herein is a heteroaromatic WEE1 degradation compound having the general formula provided below:
wherein the WEE1 kinase affinity motif is a molecular construct having high affinity for the WEE1 kinase protein independent of the linking motif and/or the recruitment motif, the linking motif is a molecular construct providing a covalent bond to both the WEE1 kinase affinity motif and the recruitment motif, and the recruitment motif is a molecular construct having the ability to selectively target and recruit protein degradation.
Recruitment motif
Recruitment motifs include E3 ligase recognition agents and proteasome recognition agents. In some embodiments, the E3 ligase is cereblon (CRBN) . In some embodiments, the recruitment motif is a CRBN ligand. In some embodiments, the recruitment motif is a derivative of a CRBN ligand.
In some embodiments, the recruitment motif is a CRBN ligand having the structure illustrated below:
wherein,
Z3 is -CH2-, -C (O) -, -C (S) -, =CH-, =N-, or -NH-;
Z4 is
eachrepresents a single or double bond;
ring A is selected from:
R60 and R61 are each independently hydrogen, halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl;
or R60 and R61 are taken together to form -C (O) -;
each R62 is independently halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl;
R63 is O, S, or NR70;
g is 0, 1, 2 or 3;
X5, X6, and X7 are each independently -N= or =CR64-;
R64 is hydrogen, halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl; and
each R70 is independently hydrogen or optionally substituted C1-6 alkyl; or
two R70 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring;
wherein the **indicates the attachment point to the Linking Motif and the rest of the heteroaromatic compound. In some embodiments (e.g., compounds of Formula (I) , Formula (II) , Formula (III) , or Formula (IV) ) , the Linking Motif is -L-. In some embodiments,
In some embodiments, the recruitment motif is a CRBN ligand selected from
Linking Motif
The linking motif is a molecular construct providing a covalent bond to both the WEE1 kinase affinity motif and the recruitment motif. In some embodiments, the linking motif comprises a cyclic moiety. In some embodiments, the linking motif comprises an acyclic moiety. In some embodiments, the linking motif comprises a heteroarylene, a heterocyclene, or a cycloalkylene moiety. In some embodiments, the linking motif comprises between 4 and 50 non-hydrogen atoms in a linear sequence. In some embodiments, the linking motif comprises between 4 and 20 non-hydrogen atoms in a linear sequence. In some embodiments, the linking motif comprises between 4 and 25 non-hydrogen atoms in a linear sequence. In some embodiments, the linking motif comprises between 4 and 30 non-hydrogen atoms in a linear sequence. In some embodiments, the linking motif comprises between 6 and 18 atoms. In some embodiments, the linking motif comprises between 5 and 10 non-hydrogen atoms in a linear sequence. In some embodiments, the linking motif comprises 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 non-hydrogen atoms in a linear sequence.
In some embodiments, the linking motif is represented by the following formula:
-Ak1-Z1-Ak2-Z2-Ak3-*
wherein,
Ak1 is selected from - (CR8R9) k-or -O-;
Ak2 is selected from - (CR8R9) m-;
Ak3 is selected from - (CR8R9) n-;
each of k, m, and n is an integer selected from 0 to 10;
each R8 and R9 is independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl; or
each - (CR8R9) -may independently form -C (O) -
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10 membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-11 cycloalkylene;
each R7 is independently hydrogen or optionally substituted C1-6 alkyl; and
wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene; and wherein the point of bonding to the CRBN ligand is indicated by the asterisk.
In some embodiments, the linking motif is represented by -Ak1-Z1-Ak2-Z2-Ak3-*and is selected from a molecular fragment illustrated below:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6; and
n is 0, 1, 2, 3, 4, 5 or 6; and
q is 0, 1, 2, 3, or 4.
In some embodiments, the linking motif is represented by -Ak1-Z1-Ak2-Z2-Ak3-*and is selected from a molecular fragment illustrated below:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, C1-3 alkyl, and -CF3;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6; and
n is 0, 1, 2, 3, 4, 5 or 6.
In some embodiments, the linking motif is represented by -Ak1-Z1-Ak2-Z2-Ak3-*and is selected from a molecular fragment illustrated below:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from -F, methyl, and -CF3;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6; and
n is 0, 1, 2, 3, 4, 5 or 6.
In some embodiments, the linking motif is represented by -Ak1-Z1-Ak2-Z2-Ak3-*and is selected from a molecular fragment illustrated below:
In some embodiments, the linking motif is represented by -Ak1-Z1-Ak2-Z2-Ak3-*and is selected from a molecular fragment illustrated below:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3 alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments, the linking motif is represented by -Ak1-Z1-Ak2-Z2-Ak3-*and is selected from a molecular fragment illustrated below:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments, the linking motif is represented by -Ak1-Z1-Ak2-Z2-Ak3-*and is selected from a molecular fragment illustrated below:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments, the linking motif is represented by -Ak1-Z1-Ak2-Z2-Ak3-*and is selected from a molecular fragment illustrated below:
wherein,
each RL is independently hydrogen, halogen, or C1-3alkyl;
each Re is independently selected from halogen, -OH, and methyl;
q is 0, 1, or 2; and
h is 0, 1, or 2.
WEE1 Kinase Affinity Motif
Adavosertib (MK-1775, AZD1775) is a potent and selective Wee1 inhibitor with IC50 of 5.2 nM in a cell-free assay as described in Hirai et al., “Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumor cells to DNA-damaging agents” Mol. Can. Therap. 2009. DOI: 10.1158/1535-7163. MCT-09-0463 which is incorporated herein by reference.
ZN-c3 also is a potent and selective Wee1 inhibitor with IC50 of 3.8 nM in a cell-free assay as described in Huang et al., “Discovery of ZN-c3, a Highly Potent and Selective Wee1 inhibitor Undergoing Evaluation in Clinical Trials for the Treatment of Cancer” J. Med. Chem. 2021. DOI: 10.1021/acs. jmedchem. 1c01121which is incorporated herein by reference.
In some embodiments, the WEE1 Kinase Affinity Motif is represented by a molecular fragment illustrated below:
wherein,
R1, R2, R3, and R4 are each independently selected from hydrogen, optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, and cyano; or
R1 and R2, R2 and R3, or R3 and R4 together with the intervening atoms to which they are attached form an optionally substituted 5-6 membered carbocyclyl or optionally substituted heterocyclyl ring;
R5 isor
R1 and R5 are taken together with the intervening atoms to which they are attached to form an optionally substituted 10-12 membered ring;
each R6 is independently selected from halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;
W is CH, CR6, or N;
v is 0, 1, 2, or 3;
ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring; and
each R7 is independently hydrogen or optionally substituted C1-6 alkyl; or
two R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring.
In some embodiments, the WEE1 Kinase Affinity Motif is represented by a molecular fragment illustrated below:
wherein,
R50 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl;
R51 and R52 are each independently hydrogen or optionally substituted straight or branched C1-6 alkyl; or
R51 and R52 are taken together to form -C (O) -;
represents a single or double bond;
R53, R54, R55, R56, R57, R58, and R59 are each independently hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;
ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring; and
each R7 is independently hydrogen or optionally substituted C1-6 alkyl; or
two R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring.
In some embodiments, the WEE1 Kinase Affinity Motif is represented by a molecular fragment illustrated below:
wherein,
R81 and R82 are each independently selected from hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -
NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;
each R83 is independently selected from halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;
R84 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl;
R85 and R86 are each independently hydrogen or optionally substituted straight or branched C1-6 alkyl; or
R85 and R86 are taken together to form -C (O) -;
represents a single or double bond;
W is CH, CR83, or N;
v is 0, 1, 2, or 3;
ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring; and
each R7 is independently hydrogen or optionally substituted C1-6 alkyl; or
two R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring;
provided that whenrepresents a double bond, then R84 and R85 are absent.
In some embodiments, the WEE1 Kinase Affinity Motif is represented by a molecular fragment illustrated below:
wherein,
A1, A2, A3, and A4 are each independently CH or N;
Lcyc is -L1cyc-, -L1cyc-L2cyc-, -L1cyc-L2cyc-L3cyc-, or -L1cyc-L2cyc-L3cyc-L4cyc-;
L1cyc, L2cyc, L3cyc, and L4cyc are each independently bond, -O-, -S-, -NRy-, -C (O) -, -S (O) m1-, - (CRcRd) n1-, - (CHRc=CHRd) n1-, -C≡C-, -NRy (CRfRg) n2-, -O (CRcRd) n1O (CRfRg) n2-, -S (CRcRd) n1S (CRfRg) n2-, - (CRcRd) n1O (CRfRg) n2-, -O (CRcRd) n1NRy (CRfRg) n2-, -O (CRcRd) n1NRy (CRfRg) n2O-, - (CRcRd) n1S (CRfRg) n2-, - (CRcRd) n1 (CRfRg) n2-, - (CHRc=CHRd) n1 (CRfRg) n2-, - (CRcRd) n1 (C≡C) (CRfRg) n2-, - (CRcRd) n1C (O) (CRfRg) n2-, - (CRcRd) n1S (O) m1 (CRfRg) n2-, - (CRcRd) n1NRyC (O) (CRfRg) n2-, - (CRcRd) n1NRyS (O) m1 (CRfRg) n2-, - (CRcRd) n1C (O) NRy (CRfRg) n2-, - (CRcRd) n1S (O) (=NRy) (CRfRg) n2-, - (CRcRd) n1N=S (=O) Ry (CRfRg) n2-, or - (CRcRd) n1S (O) m1NRy (CRfRg) n2-;
Rc, Rd, Ry, Rf, and Rg are each independently hydrogen, halogen, -OH, -SH, nitro, cyano, -NH2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3 -12 cycloalkyl, optionally substituted 3-12 membered heterocyclyl, optionally substituted C6-14 aryl, or optionally substituted 5-14 membered heteroaryl; or
any two of Rc, Rd, Ry, Rf, and Rg are taken together with the atoms to which they are attached to form an optionally substituted C3-12 cycloalkyl, an optionally substituted 3-12 membered heterocyclyl, an optionally substituted C6-14 aryl group, or an optionally substituted 5 -14 membered heteroaryl;
n1, and n2 are each independently an integer selected from 0 to 10;
m1 is 0, 1, or 2; and
ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring.
Heteroaromatic WEE1 degradation compounds
In some embodiments, provided herein is a compound, or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (I) :
wherein,
R1, R2, R3, and R4 are each independently selected from hydrogen, optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, and cyano; or
R1 and R2, R2 and R3, or R3 and R4 together with the intervening atoms to which they are attached form an optionally substituted 5-6 membered carbocyclyl or optionally
substituted heterocyclyl ring;
R5 isor
R1 and R5 are taken together with the intervening atoms to which they are attached to form an optionally substituted 10-12 membered ring;
each R6 is independently selected from halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;
W is CH, CR6, or N;
v is 0, 1, 2, or 3;
each R7 is independently hydrogen or optionally substituted C1-6 alkyl; or
two R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring;
ring B is an optionally substituted C6-14cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;
L is -Ak1-Z1-Ak2-Z2-Ak3-*;
Ak1 is - (CR8R9) k-or -O-;
Ak2 is - (CR8R9) m-;
Ak3 is - (CR8R9) n-;
each of k, m, and n is an integer selected from 0 to 10;
each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl; or
each - (CR8R9) -may independently form -C (O) -;
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10 membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-11 cycloalkylene;
wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;
wherein the point of bonding to the CRBN ligand is indicated by the asterisk; and U is a CRBN ligand.
In some embodiments, provided herein is a compound of Formula (I) , or a pharmaceutically acceptable salt or solvate thereof.
In some embodiments of Formula (I) ,
R1, R2, R3, and R4 are each independently selected from hydrogen, optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, and cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; or
R1 and R2, R2 and R3, or R3 and R4 together with the intervening atoms to which they are attached form an optionally substituted 5-6 membered carbocyclyl or optionally substituted heterocyclyl ring, wherein the carbocyclyl and heterocyclyl ring is optionally substituted with one or more Re;
R5 isor
R1 and R5 are taken together to with the intervening atoms to which they are attached to form an optionally substituted 10-12 membered ring, wherein the 10-12 membered ring is optionally substituted with one or more Re;
each R6 is independently selected from halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl, wherein the alkyl, haloalkyl, alkoxy, heteroalkyl, carbocyclyl, and heterocyclyl are independently optionally substituted with one or more Re;
v is 0, 1, 2, or 3;
each R7 is independently hydrogen or optionally substituted C1-6 alkyl, wherein the alkyl, is optionally substituted with one or more Re; or
two R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring, wherein the 3-6 membered heterocyclyl ring is optionally substituted with one or more Re;
ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB;
L is -Ak1-Z1-Ak2-Z2-Ak3-*;
Ak1 is - (CR8R9) k-or -O-;
Ak2 is - (CR8R9) m-;
Ak3 is - (CR8R9) n-;
each of k, m, and n is an integer selected from 0 to 10;
each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl, wherein the alkyl and cycloalkyl are each independently optionally substituted with one or more Re; or
each - (CR8R9) -may independently form -C (O) -;
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10 membered heteroaryl, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene, wherein the heteroarylene, heterocyclene, and cycloalkylene are independently optionally substituted with one or more Re;
wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;
each Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl; or
two Re attached to the same atom are taken together to form -C (O) -; and
each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; or
two RB groups attached to the same atom are taken together to form -C (O) -; or
two RB groups attached to different atoms are taken together to form a bridge.
In some embodiments of Formula (I) , W is CH or N. In some embodiments, W is CR6 or N. In some embodiments, W is CH or CR6. In some embodiments, W is CH. In some embodiments, W is CR6. In some embodiments, W is N.
In some embodiments of Formula (I) , R1 is optionally substituted branched C1-6 alkyl, wherein the alkyl is optionally substituted with hydroxy; and R2, R3, and R4 are hydrogen. In
some embodiments, R1 isIn some embodiments, R5 isIn some embodiments of Formula (I) , the compound has a structure of Formula (I-1) :
In some embodiments of Formula (I) , R1 and R2 together with the intervening atoms to which they are attached form an optionally substituted 5-6 membered cycloalkyl or optionally substituted 5-6 membered heterocyclyl ring, wherein the cycloalkyl and heterocyclyl are optionally substituted with one or more substituents selected from C1-6 alkyl and hydroxyl. In some embodiments, R1 and R2 together with the intervening atoms to which they are attached formIn some embodiments, R5 isIn some embodiments of Formula (I) , the compound has a structure of Formula (I-2) :
In some embodiments of Formula (I) , R1 and R5 are taken together to with the intervening atoms to which they are attached to form an optionally substituted 10-12 membered ring, wherein the ring is optionally substituted with one or more substituents selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl. In some embodiments of Formula (I) , R1 and R5 are taken together to with the intervening atoms to which they are attached to form an optionally substituted 10-12 membered ring, wherein the ring is optionally substituted with one or more substituents selected from C1-6 alkyl, halogen, and hydroxyl. In some embodiments, the ring comprises an alkene or alkyne bond. In some embodiments of Formula (I) , the compound has a structure of Formula (I-3) :
wherein,
the wavy bondin Formula (I-3) represents an E-isomer or a Z-isomer with respect to the double bond; and
each R10 is independently selected from hydrogen, halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl.
In some embodiments of Formula (I) and Formula (I-3) , R10 is selected from hydrogen, halogen, hydroxyl, and C1-3 alkyl. In some embodiments of Formula (I) and Formula (I-3) , R10 is selected from halogen, hydroxyl, and C1-3 alkyl. In some embodiments of Formula (I) and Formula (I-3) , R10 is selected from hydrogen and fluoro. In some embodiments of Formula (I) , the compound has a structure of Formula (I-3a) or Formula (I-3b) :
wherein,
the wavy bondin Formula (I-3a) or Formula (I-3b) represents an E-isomer or a Z-isomer with respect to the double bond; and
each R10 is selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH,
-C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl.
In some embodiments, R10 is selected from halogen and C1-3 alkyl. In some embodiments, R10 is fluoro.
In some embodiments of Formula (I) , the compound has a structure of Formula (I-3c) :
wherein,
the wavy bondin Formula (I-3c) represents an E-isomer or a Z-isomer with respect to the double bond.
In some embodiments of Formula (I) and Formula (I-3) , the compound has a structure of Formula (I-3-R) :
wherein,
the wavy bondin Formula (I-3-R) represents an E-isomer or a Z-isomer with respect to the double bond; and
each R10 is independently selected from hydrogen, halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl.
In some embodiments of Formula (I-3-R) , R10 is selected from hydrogen, halogen, hydroxyl, and C1-3 alkyl. In some embodiments of Formula (I-3-R) , R10 is selected from halogen, hydroxyl, and C1-3 alkyl. In some embodiments of Formula (I-3-R) , R10 is selected from hydrogen and fluoro. In some embodiments of Formula (I-3-R) , the compound has a structure of Formula (I-3a-R) or Formula (I-3b-R) :
wherein,
the wavy bondin Formula (I-3a-R) or Formula (I-3b-R) represents an E-isomer or a Z-isomer with respect to the double bond; and
each R10 is selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl.
In some embodiments, R10 is selected from halogen and C1-3 alkyl. In some embodiments, R10 is fluoro.
In some embodiments of Formula (I-3-R) , the compound has a structure of Formula (I-3c-R) :
wherein,
the wavy bondin Formula (I-3c) represents an E-isomer or a Z-isomer with respect to the double bond.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , each R6 is independently selected from halogen, cyano, -OR7, -
N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, and optionally substituted C1-6 alkoxy. In some embodiments, each R6 is independently selected from halogen, cyano, -OR7, -N (R7) 2, and optionally substituted C1-4 alkyl. In some embodiments, each R6 is independently selected from -F, -Cl, cyano, methyl, and -CH2OH. In some embodiments, R6 is -F. In some embodiments, R6 is -Cl. In some embodiments, R6 is methyl. In some embodiments, R6 is cyano. In some embodiments, R6 is -CH2OH.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , v is 0, 1 or 2. In some embodiments, v is 0 or 1. In some embodiments, v is 0. In some embodiments, v is 1. In some embodiments, v is 2.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , ring B is a 6-14 membered carbocyclyl ring or a 6-14 membered heterocyclyl ring selected from:
wherein,
Y1 and Y2 are N, CRB, or CH;
B is a bridge selected from C1-3 alkylene and C1-2 heteroalkylene;
each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano; or
two RB groups attached to the same atom are taken together to form -C (O) -;
j is 0, 1, 2, 3, or 4; and
o and p are each independently 1 or 2.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) ,
Formula (I-3c-R) , ring B isIn some embodiments, ring B isIn some embodiments, ring B isIn some embodiments, ring B isIn some embodiments, ring B isIn some embodiments, ring B is In some embodiments, B is a bridge selected from C1-3 alkylene. In some embodiments, B is a bridge selected from C1-2 heteroalkylene. In some embodiments, B is selected from:
andIn some embodiments, ring B is a 6-14 membered carbocyclyl ring. In some embodiments, ring B is a 6-11 membered carbocyclyl ring. In some embodiments, ring B is a 6 membered carbocyclyl ring. In some embodiments, ring B is: In some embodiments, ring B is a 6-14 membered heterocyclyl ring. In some embodiments, ring B is a 6-
11 membered heterocyclyl ring. In some embodiments, ring B is selected from:
In some embodiments, each RB is independently optionally substituted straight or branched C1-3 alkyl or halogen. In some embodiments, each RB is independently straight or branched C1-3 alkyl or halogen. In some embodiments, each RB is independently methyl or fluoro. In some embodiments, each RB is independently methyl or fluoro and j is 0, 1, or 2. In some embodiments, j is 0. In some embodiments, ring B is selected from
In some embodiments, ring B is selected from
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula
(1-3b-R) , Formula (I-3c-R) , ring B is an optionally substituted C11-14 cycloalkyl or an optionally substituted 11-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB. In some embodiments, ring B is:
wherein,
Y1 and Y2 are N, CRB, or CH;
each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano; or
two RB groups attached to the same atom are taken together to form -C (O) -; and
j is 0, 1, 2, 3, or 4.
In some embodiments, ring B is selected from
In some embodiments, each RB is independently optionally substituted straight or branched C1-3 alkyl or halogen. In some embodiments, each RB is independently straight or branched C1-3 alkyl or halogen. In some embodiments, each RB is independently methyl or fluoro. In some embodiments, each RB is independently methyl or fluoro and j is 0, 1, or 2.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6; and
q is 0, 1, 2, 3, or 4.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2. In some embodiments, q is 0. In some embodiments, q is 1 or 2. In some embodiments, q is 1. In some embodiments, q is 2. In some embodiments, R8 and R9 are each independently
selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, C1-3 alkyl, and -CF3;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6; and
n is 0, 1, 2, 3, 4, 5 or 6.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from -F, methyl, and -CF3;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6; and
n is 0, 1, 2, 3, 4, 5 or 6.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , L is -Ak1-Z1-Ak2-Z2-Ak3-*, wherein
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene.
In some embodiments, L is -Ak1-Z1-Ak2-Z2-Ak3-*, wherein
Z1 and Z2 are each independently selected from bond, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene.
In some embodiments, L is -Ak1-Z1-Ak2-Z2-Ak3-*, wherein
Z1 and Z2 are each independently selected from bond, pyrazole optionally substituted with RL , wherein each RL is independently hydrogen, halogen, or C1-3 alkyl, 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and C4-6 cycloalkylene.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3 alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments , k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2; and h is 0, 1, or 2. In some embodiments, q is 0 or 1; and h is 0 or 1. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl. In some embodiments, R8 and R9 are each independently selected from hydrogen , methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2; and h is 0, 1, or 2. In some embodiments, q is 0 or 1; and h is 0 or 1. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2; and h is 0, 1, or 2. In some embodiments, q is 0 or 1; and h is 0 or 1. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
each RL is independently hydrogen, halogen, or C1-3alkyl;
each Re is independently selected from halogen, -OH, and methyl;
q is 0, 1, or 2; and
h is 0, 1, or 2.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0 or 1; and h is 0 or 1. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , at least one of Z1 and Z2 is a spirocyclic heterocyclene. In some embodiments, Z1 is a spirocyclic heterocyclene. In some embodiments, Z2 is a spirocyclic heterocyclene. In some embodiments, the spirocyclic heterocyclene is a 9-11 membered spirocyclic heterocyclene.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
X1 and X2 at each instance is independently CH, CRe, or N;
each Re in X1 and X2 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
k is 0, 1, 2, 3, 4, 5, or 6; and
m is 0, 1, 2, 3, 4, 5 or 6.
In some embodiments, k is 0, 1, or 2; and m is 0, 1, or 2.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X1 and X2 at each instance is independently CH, CRe or N;
each Re in X1 and X2 is independently selected from halogen, -NH2, -OH, C1-3 alkyl, and -CF3;
k is 0, 1, 2, 3, 4, 5, or 6; and
m is 0, 1, 2, 3, 4, 5 or 6.
In some embodiments, k is 0, 1, or 2; and m is 0, 1, or 2.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
In some embodiments, provided herein is a compound, or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (II) :
wherein,
R50 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl;
R51 and R52 are each independently hydrogen or optionally substituted straight or branched C1-6 alkyl; or
R51 and R52 are taken together to form -C (O) -;
represents a single or double bond;
R53, R54, R55, R56, R57, R58, and R59 are each independently hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;
each R7 is independently hydrogen or optionally substituted C1-6 alkyl; or
two R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring;
ring B is an optionally substituted C6-14cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;
L is -Ak1-Z1-Ak2-Z2-Ak3-*;
Ak1 is - (CR8R9) k-or -O-;
Ak2 is - (CR8R9) m-;
Ak3 is - (CR8R9) n-;
each of k, m, and n is an integer selected from 0 to 10;
each R8 and R9 is independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl; or
each - (CR8R9) -may independently form -C (O) -;
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-11 cycloalkylene;
wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;
wherein the point of bonding to the CRBN ligand is indicated by the asterisk; and
U is a CRBN ligand.
In some embodiments, provided herein is a compound of Formula (II) , or a pharmaceutically acceptable salt or solvate thereof.
In some embodiments provided herein is a compound of Formula (II) , wherein,
R50 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl, wherein the alkyl and cycloalkyl are each optionally substituted with one or more Re;
R51 and R52 are each independently hydrogen or optionally substituted straight or branched C1-6 alkyl, wherein the alkyl is optionally substituted with one or more Re;or
R51 and R52 are taken together to form -C (O) -;
represents a single or double bond;
R53, R54, R55, R56, R57, R58, and R59 are each independently hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl, wherein the alkyl, haloalkyl, alkoxy, heteroalkyl, carbocyclyl, and heterocyclyl are independently optionally substituted with one or more Re;
each R7 is independently hydrogen or optionally substituted C1-6 alkyl, wherein the alkyl, is optionally substituted with one or more Re; or
two R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring, wherein the 3-6 membered heterocyclyl ring is optionally substituted with one or more Re;
ring B is an optionally substituted C6-14cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB;
L is -Ak1-Z1-Ak2-Z2-Ak3-*;
Ak1 is - (CR8R9) k-or -O-;
Ak2 is - (CR8R9) m-;
Ak3 is - (CR8R9) n-;
each of k, m, and n is an integer selected from 0 to 10;
each R8 and R9 is independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3
cycloalkyl, wherein the alkyl and cycloalkyl are each optionally substituted with one or more Re; or
each - (CR8R9) -may independently form -C (O) -;
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene, wherein the heteroaryl, heterocyclene, and cycloalkylene are independently optionally substituted with one or more Re;
wherein at least one of Z1 and Z2 is heterocyclene or cycloalkylene;
each Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, C1-C6 aminoalkyl, and C1-C6 heteroalkyl; or
two Re attached to the same atom are taken together to form -C (O) -; and
each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; or
two RB groups attached to the same atom are taken together to form -C (O) -; or
two RB groups attached to different atoms are taken together to form a bridge.
In some embodiments of Formula (II) , represents a single bond. In some embodiments of Formula (II) , the compound has a structure of Formula (II-1) :
In some embodiments, R50 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl. In some embodiments, R50 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl, wherein the alkyl and cycloalkyl are each optionally substituted with one or more Re. In some embodiments, R50 is hydrogen, methyl, ethyl, cyclopropyl, or cyclobutyl. In some embodiments, R50 is hydrogen. In some embodiments, R50 is methyl, ethyl, cyclopropyl, or cyclobutyl. In some embodiments, R50 is methyl or ethyl. In some embodiments, R50 is methyl. In some embodiments, R50 is ethyl. In some embodiments, R50 is cyclopropyl or cyclobutyl. In some embodiments, R50 is cyclobutyl. In some embodiments, R50 is cyclopropyl. In some embodiments, R51 and R52 are each
independently hydrogen, optionally substituted straight or branched C1-6 alkyl, or R51 and R52 are taken together to form -C (O) -. In some embodiments, R51 and R52 are each independently hydrogen, optionally substituted straight or branched C1-6 alkyl, or R51 and R52 are taken together to form -C (O) -, wherein the alkyl is optionally substituted with one or more Re. In some embodiments, R51 and R52 are each independently hydrogen, optionally substituted straight or branched C1-3 alkyl, or R51 and R52 are taken together to form -C (O) -. In some embodiments, R51 and R52 are each independently hydrogen, methyl, ethyl, isopropyl, or R51 and R52 are taken together to form -C (O) -. In some embodiments, R51 and R52 are each hydrogen, or R51 and R52 are taken together to form -C (O) -. In some embodiments, R51 is methyl and R52 is hydrogen. In some embodiments, R51 and R52 are each hydrogen. In some embodiments, R51 and R52 are taken together to form -C (O) -.
In some embodiments of Formula (II) , represents a double bond. In some embodiments of Formula (II) , the compound has a structure of Formula (II-2) :
In some embodiments, R52 is hydrogen or optionally substituted straight or branched C1-6 alkyl. In some embodiments, R52 is hydrogen or optionally substituted straight or branched C1-6 alkyl, wherein the alkyl is optionally substituted with one or more Re. In some embodiments, R52 is hydrogen or optionally substituted straight or branched C1-3 alkyl. In some embodiments, R52 is hydrogen, methyl, ethyl, or isopropyl. In some embodiments, R52 is hydrogen. In some embodiments, R52 is methyl.
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , R53, R57, R58, and R59 are each independently hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl; and R54, R55, and R56 are each hydrogen. In some embodiments, R53, R54, R55, R56, R57, R58, and R59 are each independently hydrogen, halogen, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, or optionally substituted
C1-6 heteroalkyl. In some embodiments, R53 and R57 are each independently hydrogen, halogen, cyano, -OR7, -N (R7) 2, -SR7, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, or optionally substituted C1-6 heteroalkyl; R58 and R59 are each independently hydrogen, halogen, cyano, -OR7, -N (R7) 2, -SR7, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, or optionally substituted C1-6 heteroalkyl; and R54, R55, R56 are each hydrogen. In some embodiments, R53 and R57 are each independently hydrogen or halogen; and R54, R55, and R56 are each hydrogen. In some embodiments, R53 and R57 are each independently halogen; and R54, R55, and R56 are each hydrogen. In some embodiments, R53 and R57 are each independently hydrogen, fluoro or chloro; and R54, R55, and R56 are each hydrogen. In some embodiments, R53 is hydrogen, R57 is fluoro or chloro; and R54, R55, and R56 are each hydrogen. In some embodiments, R53 is fluoro or chloro; R57 is hydrogen, and R54, R55, and R56 are each hydrogen. In some embodiments, R58 and R59 are each independently hydrogen, -F, -Cl, or methyl.
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , ring B is a 6-14 membered carbocyclyl ring or a 6-14 membered heterocyclyl ring selected from:
wherein,
Y1 and Y2 are N or CH;
B is a bridge selected from C1-3 alkylene and C1-2 heteroalkylene;
each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano; or
two RB groups attached to the same atom are taken together to form -C (O) -;
j is 0, 1, 2, 3, or 4; and
o and p are each independently 1 or 2.
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , ring B is In some embodiments, ring B isIn some embodiments, ring B is
In some embodiments, ring B isIn some embodiments, ring B isIn some embodiments, ring B isIn some embodiments, B is a bridge selected from C1-3 alkylene. In some embodiments, B is a bridge selected from C1-2 heteroalkylene. In some embodiments, B is selected from:
In some embodiments, ring B is a 6-14 membered carbocyclyl ring. In some embodiments, ring B is a 6-11 membered carbocyclyl ring. In some embodiments, ring B is a 6 membered carbocyclyl ring. In some embodiments, ring B is: In some embodiments, ring B is a 6-14 membered heterocyclyl ring. In some embodiments, ring B is a 6-11 membered heterocyclyl ring. In some embodiments, ring B is selected from:
In some embodiments, each RB is independently optionally substituted straight or branched C1-3 alkyl or halogen. In some embodiments, each RB is independently straight or branched C1-3 alkyl or halogen. In some embodiments, each RB is independently methyl or fluoro; and j is 0, 1, or 2. In some embodiments, j is 0. In some embodiments, ring B is selected from
In some embodiments, ring B is selected from
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6; and
q is 0, 1, 2, 3, or 4.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2. In some embodiments, q is 0. In some embodiments, q is 1 or 2. In some embodiments, q is 1. In some embodiments, q is 2. In some embodiments, R8 and R9 are each independently selected from hydrogen and methyl; or each - (CR8R9) -may independently form -C (O) -
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, C1-3 alkyl, and -CF3;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6; and
n is 0, 1, 2, 3, 4, 5 or 6.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0.
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from -F, methyl, and -CF3;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6; and
n is 0, 1, 2, 3, 4, 5 or 6.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0.
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , L is -Ak1-Z1-Ak2-Z2-Ak3-*, wherein
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene.
In some embodiments, L is -Ak1-Z1-Ak2-Z2-Ak3-*, wherein
Z1 and Z2 are each independently selected from bond, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene.
In some embodiments, L is -Ak1-Z1-Ak2-Z2-Ak3-*, wherein
Z1 and Z2 are each independently selected from bond, pyrazole optionally substituted with RL , wherein each RL is independently hydrogen, halogen, or C1-3 alkyl, 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and C4-6 cycloalkylene.
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3 alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2; and h is 0, 1, or 2. In some embodiments, q is 0 or 1; and h is 0 or 1. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl. In some embodiments, R8 and R9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -.
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2; and h is 0, 1, or 2. In some embodiments, q is 0 or 1; and h is 0 or 1. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl.
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2; and h is 0, 1, or 2. In some embodiments, q is 0 or 1; and h is 0 or 1. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl.
In some embodiments of Formula (II) , Formula (II-1) , or Formula (II-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
each RL is independently hydrogen, halogen, or C1-3alkyl;
each Re is independently selected from halogen, -OH, and methyl;
q is 0, 1, or 2; and
h is 0, 1, or 2.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0 or 1; and h is 0 or 2. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl.
In some embodiments, provided herein is a compound, or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (III) :
wherein,
R81 and R82 are each independently selected from hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2,
-C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;
each R83 is independently selected from halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;
R84 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl;
R85 and R86 are each independently hydrogen or optionally substituted straight or branched C1-6 alkyl; or
R85 and R86 are taken together to form -C (O) -;
represents a single or double bond;
W is CH, CR83, or N;
v is 0, 1, 2, or 3;
each R7 is independently hydrogen or optionally substituted C1-6 alkyl; or
two R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring;
ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;
L is -Ak1-Z1-Ak2-Z2-Ak3-*;
Ak1 is - (CR8R9) k-or -O-;
Ak2 is - (CR8R9) m-;
Ak3 is - (CR8R9) n-;
each of k, m, and n is an integer selected from 0 to 10;
each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl; or
each - (CR8R9) -may independently form -C (O) -;
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-11 cycloalkylene;
wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;
wherein the point of bonding to the CRBN ligand is indicated by the asterisk; and
U is a CRBN ligand;
provided that whenrepresents a double bond, then R84 and R85 are absent.
In some embodiments, provided herein is a compound of Formula (III) , or a pharmaceutically acceptable salt or solvate thereof.
In some embodiments provided herein is a compound of Formula (III) , wherein,
R81 and R82 are each independently selected from hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl, wherein the alkyl, haloalkyl, alkoxy, heteroalkyl, carbocyclyl, and heterocyclyl are independently optionally substituted with one or more Re;
each R83 is independently selected from halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl, wherein the alkyl, haloalkyl, alkoxy, heteroalkyl, carbocyclyl, and heterocyclyl are independently optionally substituted with one or more Re;
R84 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl, wherein the alkyl or cycloalkyl are each optionally substituted with one or more Re;
R85 and R86 are each independently hydrogen or optionally substituted straight or branched C1-6 alkyl, wherein the alkyl is optionally substituted with one or more Re;or
R85 and R86 are taken together to form -C (O) -;
represents a single or double bond;
W is CH, CR83, or N;
v is 0, 1, 2, or 3;
each R7 is independently hydrogen or optionally substituted C1-6 alkyl, wherein the alkyl, is optionally substituted with one or more Re; or
two R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring, wherein the 3-6 membered heterocyclyl ring is optionally substituted with one or more Re;
ring B is an optionally substituted C6-14cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB;
L is -Ak1-Z1-Ak2-Z2-Ak3-*;
Ak1 is - (CR8R9) k-or -O-;
Ak2 is - (CR8R9) m-;
Ak3 is - (CR8R9) n-;
each of k, m, and n is an integer selected from 0 to 10;
each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl, wherein the alkyl and cycloalkyl are each optionally substituted with one or more Re; or
each - (CR8R9) -may independently form -C (O) -;
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene, wherein the heteroaryl, heterocyclene, and cycloalkylene are independently optionally substituted with one or more Re;
wherein at least one of Z1 and Z2 is heterocyclene or cycloalkylene;
each Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, C1-C6 aminoalkyl, and C1-C6 heteroalkyl; or
two Re attached to the same atom are taken together to form -C (O) -; and
each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; or
two RB groups attached to the same atom are taken together to form -C (O) -; or
two RB groups attached to different atoms are taken together to form a bridge.
In some embodiments of Formula (III) , represents a single bond. In some embodiments of Formula (III) , the compound has a structure of Formula (III-1) :
In some embodiments, R84 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl. In some embodiments, R84 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl, wherein the alkyl and cycloalkyl are each optionally substituted with one or more Re. In some embodiments, R84 is hydrogen, methyl, ethyl, cyclopropyl, or cyclobutyl. In some embodiments, R84 is hydrogen. In some embodiments, R84 is methyl, ethyl, cyclopropyl, or cyclobutyl. In some embodiments, R84 is methyl or ethyl. In some embodiments, R84 is methyl. In some embodiments, R84 is ethyl. In some embodiments, R84 is cyclopropyl or cyclobutyl. In some embodiments, R84 is cyclobutyl. In some embodiments, R84 is cyclopropyl. In some embodiments, R85 and R86 are each independently hydrogen, optionally substituted straight or branched C1-6 alkyl, or R85 and R86 are taken together to form -C (O) -. In some embodiments, R85 and R86 are each independently hydrogen, optionally substituted straight or branched C1-6 alkyl, or R85 and R86 are taken together to form -C (O) -, wherein the alkyl is optionally substituted with one or more Re. In some embodiments, R85 and R86 are each independently hydrogen, optionally substituted straight or branched C1-3 alkyl, or R85 and R86 are taken together to form -C (O) -. In some embodiments, R85 and R86 are each independently hydrogen, methyl, ethyl, isopropyl, or R85 and R86 are taken together to form -C (O) -. In some embodiments, R85 and R86 are each hydrogen, or R85 and R86 are taken together to form -C (O) -. In some embodiments, R85 is methyl and R86 is hydrogen. In some embodiments, R85 and R86 are each hydrogen. In some embodiments, R85 and R86 are taken together to form -C (O) -. In some embodiments, R85 and R86 are each hydrogen and R84 is methyl.
In some embodiments of Formula (III) , represents a double bond. In some embodiments of Formula (III) , the compound has a structure of Formula (III-2) :
In some embodiments, R86 is hydrogen or optionally substituted straight or branched C1-6 alkyl. In some embodiments, R86 is hydrogen or optionally substituted straight or branched C1-6 alkyl, wherein the alkyl is optionally substituted with one or more Re. In some embodiments, R86 is hydrogen or optionally substituted straight or branched C1-3 alkyl. In some embodiments, R86 is hydrogen, methyl, ethyl, or isopropyl. In some embodiments, R86 is hydrogen. In some embodiments, R86 is methyl.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , R81 and R82 are each independently hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl. In some embodiments, R81 and R82 are each independently hydrogen, halogen, cyano, hydroxy, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, or optionally substituted C1-6 alkoxy. In some embodiments, R81 and R82 are each independently hydrogen or halogen. In some embodiments, at least one of R81 and R82 is halogen. In some embodiments, R81 and R82 are each independently halogen. In some embodiments, R81 and R82 are each independently selected from fluoro, chloro, bromo, and iodo. In some embodiments, R81 and R82 are each independently selected from fluoro and chloro. In some embodiments, R81 and R82 are each chloro. In some embodiments, R81 is halogen and R82 is hydrogen. In some embodiments, R81 is selected from fluoro, chloro, bromo, and iodo; and R82 is hydrogen. In some embodiments, R81 is selected from fluoro and chloro; and R82 is hydrogen. In some embodiments, R81 is chloro and R82 is hydrogen.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , each R83 is independently selected from halogen, cyano, -OR7, -N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, and optionally substituted C1-6 alkoxy. In some embodiments, each R83 is independently selected from halogen, cyano, -OR7, -N (R7) 2, and optionally substituted C1-4 alkyl. In some embodiments, each R83 is independently selected from -F, -Cl, cyano, methyl, and -CH2OH. In some embodiments, R83 is -
Cl. In some embodiments, R83 is methyl. In some embodiments, R83 is cyano. In some embodiments, R83 is -CH2OH.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , v is 0, 1 or 2. In some embodiments, v is 0 or 1. In some embodiments, v is 0. In some embodiments, v is 1. In some embodiments, v is 2.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , W is CH or N.In some embodiments, W is CR83 or N. In some embodiments, W is CH or CR83. In some embodiments, W is CH. In some embodiments, W is CR83. In some embodiments, W is N.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , ring B is a 6-14 membered carbocyclyl ring or a 6-14 membered heterocyclyl ring selected from:
wherein,
Y1 and Y2 are N or CH;
B is a bridge selected from C1-3 alkylene and C1-2 heteroalkylene;
each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano; or
two RB groups attached to the same atom are taken together to form -C (O) -;
j is 0, 1, 2, 3, or 4; and
o and p are each independently 1 or 2.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , ring B is In some embodiments, ring B isIn some embodiments, ring B is In some embodiments, ring B isIn some embodiments, ring B isIn some embodiments, ring B isIn some embodiments, B is
a bridge selected from C1-3 alkylene. In some embodiments, B is a bridge selected from C1-2 heteroalkylene. In some embodiments, B is selected from:
In some embodiments, ring B is a 6-14 membered carbocyclyl ring. In some embodiments, ring B is a 6-11 membered carbocyclyl ring. In some embodiments, ring B is a 6 membered carbocyclyl ring. In some embodiments, ring B is: In some embodiments, ring B is a 6-14 membered heterocyclyl ring. In some embodiments, ring B is a 6-11 membered heterocyclyl ring. In some embodiments, ring B is selected from:
In some embodiments, each RB is independently
optionally substituted straight or branched C1-3 alkyl or halogen. In some embodiments, each RB is independently straight or branched C1-3 alkyl or halogen. In some embodiments, each RB is independently methyl or fluoro; and j is 0, 1, or 2. In some embodiments, j is 0. In some embodiments, ring B is selected from
In some embodiments, ring B is selected from
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6; and
q is 0, 1, 2, 3, or 4.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2. In some embodiments, q is 0. In some embodiments, q is 1 or 2. In some embodiments, q is 1. In some embodiments, q is 2. In some embodiments, R8 and R9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, C1-3 alkyl, and -CF3;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6; and
n is 0, 1, 2, 3, 4, 5 or 6.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from -F, methyl, and -CF3;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6; and
n is 0, 1, 2, 3, 4, 5 or 6.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , L is -Ak1-Z1-Ak2-Z2-Ak3-*, wherein
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene.
In some embodiments, L is -Ak1-Z1-Ak2-Z2-Ak3-*, wherein
Z1 and Z2 are each independently selected from bond, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene.
In some embodiments, L is -Ak1-Z1-Ak2-Z2-Ak3-*, wherein
Z1 and Z2 are each independently selected from bond, pyrazole optionally substituted with RL , wherein each RL is independently hydrogen, halogen, or C1-3 alkyl, 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and C4-6 cycloalkylene.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3 alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2; and h is 0, 1, or 2. In some embodiments, q is 0 or 1; and h is 0 or 1. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl. In some embodiments, R8 and R9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2; and h is 0, 1, or 2. In some embodiments, q is 0 or 1; and h is 0 or 1. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2; and h is 0, 1, or 2. In some embodiments, q is 0 or 1; and h is 0 or 1. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl.
In some embodiments of Formula (III) , Formula (III-1) , or Formula (III-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
each RL is independently hydrogen, halogen, or C1-3alkyl;
each Re is independently selected from halogen, -OH, and methyl;
q is 0, 1, or 2; and
h is 0, 1, or 2.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0 or 1; and h is 0 or 2. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl.
In some embodiments, provided herein is a compound, or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (IV) :
wherein,
A1, A2, A3, and A4 are each independently CH or N;
Lcyc is -L1cyc-, -L1cyc-L2cyc-, -L1cyc-L2cyc-L3cyc-, or -L1cyc-L2cyc-L3cyc-L4cyc-;
L1cyc, L2cyc, L3cyc, and L4cyc are each independently bond, -O-, -S-, -NRy-, -C (O) -, -S (O) m1-, - (CRcRd) n1-, - (CHRc=CHRd) n1-, -C≡C-, -NRy (CRfRg) n2-, -O (CRcRd) n1O (CRfRg) n2-, -S (CRcRd) n1S (CRfRg) n2-, - (CRcRd) n1O (CRfRg) n2-, -O (CRcRd) n1NRy (CRfRg) n2-, -O (CRcRd) n1NRy (CRfRg) n2O-, - (CRcRd) n1S (CRfRg) n2-, - (CRcRd) n1 (CRfRg) n2-, - (CHRc=CHRd) n1 (CRfRg) n2-, - (CRcRd) n1 (C≡C) (CRfRg) n2-, - (CRcRd) n1C (O) (CRfRg) n2-, - (CRcRd) n1S (O) m1 (CRfRg) n2-, - (CRcRd) n1NRyC (O) (CRfRg) n2-, - (CRcRd) n1NRyS (O) m1 (CRfRg) n2-, - (CRcRd) n1C (O) NRy (CRfRg) n2-, - (CRcRd) n1S (O) (=NRy) (CRfRg) n2-, - (CRcRd) n1N=S (=O) Ry (CRfRg) n2-, or - (CRcRd) n1S (O) m1NRy (CRfRg) n2-;
Rc, Rd, Rf, and Rg are each independently hydrogen, halogen, -OH, -SH, nitro, cyano, -NH2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3 -12 cycloalkyl, optionally substituted 3-12 membered heterocyclyl, optionally substituted C6-14 aryl, or optionally substituted 5-14 membered heteroaryl; or
any two of Rc, Rd, Rf, and Rg are taken together with the atoms to which they are attached to form an optionally substituted C3-12 cycloalkyl, an optionally substituted 3-12 membered heterocyclyl, an optionally substituted C6-14 aryl group, or an optionally substituted 5 -14 membered heteroaryl;
Ry is hydrogen, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3 -12 cycloalkyl, optionally substituted 3-12 membered heterocyclyl, optionally substituted C6-14 aryl, or optionally substituted 5-14 membered heteroaryl;
n1, and n2 are each independently an integer selected from 0 to 10;
m1 is 0, 1, or 2;
ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;
L is -Ak1-Z1-Ak2-Z2-Ak3-*;
Ak1 is - (CR8R9) k-or -O-;
Ak2 is - (CR8R9) m-;
Ak3 is - (CR8R9) n-;
each of k, m, and n is an integer selected from 0 to 10;
each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl; or
each - (CR8R9) -may independently form -C (O) -;
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-11 cycloalkylene;
wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;
wherein the point of bonding to the CRBN ligand is indicated by the asterisk; and U is a CRBN ligand.
In some embodiments, provided herein is a compound of Formula (IV) , or a pharmaceutically acceptable salt or solvate thereof.
In some embodiments of Formula (IV) ,
Rc, Rd, Rf, and Rg are each independently hydrogen, halogen, -OH, -SH, nitro, cyano, -NH2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3 -12 cycloalkyl, optionally substituted 3-12 membered heterocyclyl, optionally substituted C6-14 aryl, or optionally substituted 5-14 membered heteroaryl; wherein the alkyl, haloalkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, are independently optionally substituted with one or more Re;
or
any two of Rc, Rd, Rf, and Rg are taken together with the atoms to which they are attached to form an optionally substituted C3-12 cycloalkyl group, an optionally substituted 3-12 membered heterocyclyl, an optionally substituted C6-14 aryl group, or an optionally substituted 5 -14 membered heteroaryl, wherein the cycloalkyl, heterocyclyl, aryl, and heteroaryl are independently optionally substituted with one or more Re;
Ry is hydrogen, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3 -12 cycloalkyl, optionally substituted 3-12 membered heterocyclyl, optionally substituted C6-14 aryl, or optionally substituted 5-14 membered heteroaryl, wherein the alkyl, haloalkyl,
alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, are independently optionally substituted with one or more Re;
ring B is an optionally substituted C6-14cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB;
L is -Ak1-Z1-Ak2-Z2-Ak3-*;
Ak1 is - (CR8R9) k-or -O-;
Ak2 is - (CR8R9) m-;
Ak3 is - (CR8R9) n-;
each of k, m, and n is an integer selected from 0 to 10;
each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl, wherein the alkyl and cycloalkyl are each optionally substituted with one or more Re; or
each - (CR8R9) -may independently form -C (O) -;
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene, wherein the heteroaryl, heterocyclene, and cycloalkylene are independently optionally substituted with one or more Re;
wherein at least one of Z1 and Z2 is heterocyclene or cycloalkylene;
each Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N(CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, C1-C6 aminoalkyl, and C1-C6 heteroalkyl; or
two Re attached to the same atom are taken together to form -C (O) -; and
each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; or
two RB groups attached to the same atom are taken together to form -C (O) -; or
two RB groups attached to different atoms are taken together to form a bridge.
In some embodiments of Formula (IV) , A1 is CH. In some embodiments, A1 is N. In some embodiments, A2 is CH. In some embodiments, A2 is N. In some embodiments, A3 is CH. In some embodiments, A3 is N. In some embodiments, A4 is CH. In some embodiments, A4 is N. In some embodiments, A1 is CH, A2 is CH, A3 is CH, and A4 is CH. In some embodiments, A1 is
N, A2 is CH, A3 is CH, and A4 is CH. In some embodiments, A1 is CH, A2 is N, A3 is CH, and A4 is CH. In some embodiments, A1 is CH, A2 is CH, A3 is N, and A4 is CH. In some embodiments, A1 is CH, A2 is CH, A3 is CH, and A4 is N. In some embodiments, A1 is N, A2 is N, A3 is CH, and A4 is CH. In some embodiments, A1 is N, A2 is CH, A3 is N, and A4 is CH. In some embodiments, A1 is N, A2 is CH, A3 is CH, and A4 is N. In some embodiments, A1 is CH, A2 is N, A3 is N, and A4 is CH. In some embodiments, A1 is CH, A2 is N, A3 is CH, and A4 is N. In some embodiments, A1 is CH, A2 is CH, A3 is N, and A4 is N. In some embodiments, A1 is N, A2 is N, A3 is N, and A4 is CH. In some embodiments, A1 is N, A2 is N, A3 is CH, and A4 is N. In some embodiments, A1 is N, A2 is CH, A3 is N, and A4 is N. In some embodiments, A1 is CH, A2 is N, A3 is N, and A4 is N. In some embodiments, A1 is N, A2 is N, A3 is N, and A4 is N.
In some embodiments of Formula (IV) , A1 is CH, A3 is CH, and A4 is CH, and the compound has the structure of Formula (IV-1) :
In some embodiments of Formula (IV) or Formula (IV-1) , Lcyc is -L1cyc-. In some embodiments, Lcyc is -L1cyc-L2cyc-. In some embodiments, Lcyc is -L1cyc-L2cyc-L3cyc-. In some embodiments, Lcyc is -L1cyc-L2cyc-L3cyc-L4cyc-. In some embodiments, L1cyc, L2cyc, L3cyc, and L4cyc are each independently bond, -O-, -CH2-, -OCH2-, -CH2O-, or -CH2CH2-.
In some embodiments of Formula (IV) or Formula (IV-1) , the compound has the structure of Formula (IV-2) :
In some embodiments, A2 is CH or N. In some embodiments, A2 is N. IN some embodiments, A2 is CH.
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , L1cyc is bond, -O-, -CH2-, -OCH2-, -CH2O-, or -CH2CH2-. In some embodiments, L1cyc is bond. In some embodiments, L1cyc is -O-, -CH2-, -OCH2-, -CH2O-, or -CH2CH2-. In some embodiments, L1cyc
is -O-, -OCH2-, or -CH2O-. In some embodiments, L1cyc is -CH2-or -CH2CH2-. In some embodiments, L1cyc is bond, -O-, -CH2-, -OCH2-, -CH2O-, or -CH2CH2-.
The compound of any one of claims 139-152, wherein ring B is an optionally substituted C11-14 cycloalkyl or an optionally substituted 11-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB;
each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; or
two RB groups attached to the same atom are taken together to form -C (O) -; or
two RB groups attached to different atoms are taken together to form a bridge; and
each Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl; or
two Re attached to the same atom are taken together to form -C (O) -.
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , ring B is a 6-14 membered carbocyclyl ring or a 6-14 membered heterocyclyl ring selected from:
wherein,
Y1 and Y2 are N or CH;
B is a bridge selected from C1-3 alkylene and C1-2 heteroalkylene;
each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano; or
two RB groups attached to the same atom are taken together to form -C (O) -;
j is 0, 1, 2, 3, or 4; and
o and p are each independently 1 or 2.
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , ring B is In some embodiments, ring B isIn some embodiments, ring B is In some embodiments, ring B isIn some embodiments, ring B isIn some embodiments, ring B isIn some embodiments, B is a bridge selected from C1-3 alkylene. In some embodiments, B is a bridge selected from C1-2 heteroalkylene. In some embodiments, B is selected from:
In some embodiments, ring B is a 6-14 membered carbocyclyl ring. In some embodiments, ring B is a 6-11 membered carbocyclyl ring. In some embodiments, ring B is a 6 membered carbocyclyl ring. In some embodiments, ring B is: In some embodiments, ring B is a 6-14 membered heterocyclyl ring. In some embodiments, ring B is a 6-11 membered heterocyclyl ring. In some embodiments, ring B is selected from:
In some embodiments, each RB is independently optionally substituted straight or branched C1-3 alkyl or halogen. In some embodiments, each RB is independently straight or branched C1-3 alkyl or halogen. In some embodiments, each RB is independently methyl or fluoro; and j is 0, 1, or 2. In some embodiments, j is 0. In some embodiments, ring B is selected from
In some embodiments, ring B is selected from
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6; and
q is 0, 1, 2, 3, or 4.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2. In some embodiments, q is 0. In some embodiments, q is 1 or 2. In some embodiments, q is 1. In some embodiments, q is 2. In some embodiments, R8 and R9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, C1-3 alkyl, and -CF3;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6; and
n is 0, 1, 2, 3, 4, 5 or 6.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0.
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X, X1, and X2 at each instance is independently CH, CRe, or N;
each Re in Z1, X, X1, and X2 is independently selected from -F, methyl, and -CF3;
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6; and
n is 0, 1, 2, 3, 4, 5 or 6.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0.
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , L is -Ak1-Z1-Ak2-Z2-Ak3-*, wherein
Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene.
In some embodiments, L is -Ak1-Z1-Ak2-Z2-Ak3-*, wherein
Z1 and Z2 are each independently selected from bond, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene.
In some embodiments, L is -Ak1-Z1-Ak2-Z2-Ak3-*, wherein
Z1 and Z2 are each independently selected from bond, pyrazole optionally substituted with RL , wherein each RL is independently hydrogen, halogen, or C1-3 alkyl, 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and C4-6 cycloalkylene.
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3 alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2; and h is 0, 1, or 2. In some embodiments, q is 0 or 1; and h is 0 or 1. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl. In some embodiments, R8 and R9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -.
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2; and h is 0, 1, or 2. In some embodiments, q is 0 or 1; and h is 0 or 1. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl.
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;
each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;
each RL is independently hydrogen, halogen, or C1-3alkyl.
k is 0, 1, 2, 3, 4, 5, or 6;
m is 0, 1, 2, 3, 4, 5 or 6;
n is 0, 1, 2, 3, 4, 5 or 6;
q is 0, 1, 2, 3, or 4; and
h is 0, 1, 2, 3, or 4.
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0, 1, or 2; and h is 0, 1, or 2. In some embodiments, q is 0 or 1; and h is 0 or 1. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl.
In some embodiments of Formula (IV) , Formula (IV-1) , or Formula (IV-2) , -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,
each RL is independently hydrogen, halogen, or C1-3alkyl;
each Re is independently selected from halogen, -OH, and methyl;
q is 0, 1, or 2; and
h is 0, 1, or 2.
In some embodiments, k is 0, 1, or 2; m is 0, 1, or 2; and n is 0. In some embodiments, q is 0 or 1; and h is 0 or 2. In some embodiments, q is 0; and h is 0. In some embodiments, each RL is independently hydrogen, -F, -Cl, -Br, -I, or C1-3alkyl. each RL is independently hydrogen, -F, -Cl, -Br, -I, or methyl. In some embodiments, each RL is independently hydrogen, -F, -Cl, or methyl.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , U is a CRBN ligand having the structure
wherein,
Z3 is -CH2-, -C (O) -, -C (S) -, =CH-, =N-, or -NH-;
Z4 is
eachrepresents a single or double bond;
ring A is selected from:
R60 and R61 are each independently hydrogen, halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl;
or R60 and R61 are taken together to form -C (O) -;
each R62 is independently halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl;
R63 is O, S, or NR70;
g is 0, 1, 2 or 3;
X5, X6, and X7 are each independently -N= or =CR64-;
R64 is hydrogen, halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl; and
each R70 is independently hydrogen or optionally substituted C1-6 alkyl; or
two R70 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring;
wherein the **indicates the attachment point to -L-.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) ,
R60 and R61 are each independently hydrogen, halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl, wherein the alkyl is optionally substituted with one or more Re;
or R60 and R61 are taken together to form -C (O) -;
each R62 is independently halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl, wherein the alkyl is optionally substituted with one or more Re;
R64 is hydrogen, halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl, wherein the alkyl is optionally substituted with one or more Re; and
each Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N(CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl; or
two Re attached to the same atom are taken together to form -C (O) -.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , Z3 is -CH2-, -C (O) -, -C (S) -, =CH-, or =N-. In some embodiments, Z3 is -C (O) -, -C (S) -, or =N-. In some embodiments, Z3 is -C (O) -or =N-. In some embodiments, is
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , R60 and R61 are each hydrogen, or R60 and R61 are taken together to form -C (O) -.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , each R62 is independently halogen -OR70, or an optionally substituted C1-3 alkyl. In some embodiments, each R62 is independently fluoro, chloro, methyl, -OMe, -OEt, -CHF2, or CF3. In some embodiments, g is 0, 1, or 2. In some embodiments, g is 1 or 2. In some embodiments, g is 0. In some embodiments, g is 1. In some embodiments, g is 2.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , R63 is O.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , R64 is hydrogen, halogen, -OR70, or an optionally substituted C1-3 alkyl. In some embodiments, R64 is hydrogen or fluoro.
In some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , ring A isIn some embodiments, ring A isIn some embodiments, ring A is In some embodiments, ring A isIn some embodiments, ring A isIn some embodiments of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , the CRBN ligand has a structure selected from:
In some embodiments, is
In some embodiments, is
In some embodiments, is In some embodiments, is
In some embodiments, In some embodiments, is
In some embodiments, is
In some embodiments, is
In some embodiments, isIn some embodiments, is
In some embodiments, isIn some embodiments, is
In some embodiments,
isIn some embodiments, is
In some embodiments, g is 0, 1, or 2. In some embodiments, g is 1 or 2. In some embodiments, g is 0. In some embodiments, g is 1. In some embodiments, each R62 is independently halogen, -OR70, or C1-3 alkyl optionally substituted with halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, or C1-6 heteroalkyl. In some embodiments, each R62 is independently halogen, -OMe, -OEt, -CHF2, -CF3, or methyl. In some embodiments, R64 is independently hydrogen, halogen, -OR70, or C1-3 alkyl optionally substituted with halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, or C1-6 heteroalkyl. In some embodiments, R64 is independently hydrogen, halogen, -OMe, or methyl. In some embodiments, each R62 is independently -F, -Cl, -OMe, -OEt, -CHF2, -CF3, or methyl, and R64 is independently hydrogen, -F, -Cl, -OMe, or methyl In some embodiments, each R62 is
independently -F, -Cl, -OMe, or methyl, and R64 is independently hydrogen, -F, -Cl, -OMe, or methyl.
In some embodiments, the compounds as described herein have a structure provided in Table 1, Table 2, Table 3, Table 4, or Table 5. In some embodiments, the compounds as described herein have a structure provided in Table 1.
Table 1
In some embodiments, compounds as described herein has a structure provided in Table 2.
In some embodiments, compounds as described herein has a structure provided in Table 3.
Table 3
In some embodiments, compounds as described herein has a structure provided in Table 4.
Table 4
In some embodiments, compounds as described herein has a structure provided in Table 5.
Table 5
Preparation of Compounds
The compounds used in the reactions described herein are made according to organic synthesis techniques known to those skilled in this art, starting from commercially available chemicals and/or from compounds described in the chemical literature. "Commercially available chemicals" are obtained from standard commercial sources including Acros Organics (Pittsburgh, PA) , Aldrich Chemical (Milwaukee, WI, including Sigma Chemical and Fluka) , Apin Chemicals Ltd. (Milton Park, UK) , Avocado Research (Lancashire, U.K. ) , BDH Inc. (Toronto, Canada) , Bionet (Cornwall, U.K. ) , Chemservice Inc. (West Chester, PA) , Crescent Chemical Co. (Hauppauge, NY) , Eastman Organic Chemicals, Eastman Kodak Company (Rochester, NY) , Fisher Scientific Co. (Pittsburgh, PA) , Fisons Chemicals (Leicestershire, UK) , Frontier Scientific (Logan, UT) , ICN Biomedicals, Inc. (Costa Mesa, CA) , Key Organics (Cornwall, U.K. ) , Lancaster Synthesis (Windham, NH) , Maybridge Chemical Co. Ltd. (Cornwall, U.K. ) , Parish Chemical Co. (Orem, UT) , Pfaltz & Bauer, Inc. (Waterbury, CN) , Polyorganix (Houston, TX) , Pierce Chemical Co. (Rockford, IL) , Riedel de Haen AG (Hanover, Germany) , Spectrum Quality Product, Inc. (New Brunswick, NJ) , TCI America (Portland, OR) , Trans World Chemicals, Inc. (Rockville, MD) , and Wako Chemicals USA, Inc. (Richmond, VA) .
Suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds described herein, or provide references to articles that describe the preparation, include for example, "Synthetic Organic Chemistry" , John Wiley & Sons, Inc., New York; S.R. Sandler et al., "Organic Functional Group Preparations, " 2nd Ed., Academic Press, New York, 1983; H.O. House, "Modern Synthetic Reactions" , 2nd Ed., W.A. Benjamin, Inc. Menlo Park, Calif. 1972; T.L. Gilchrist, "Heterocyclic Chemistry" , 2nd Ed., John Wiley & Sons, New York, 1992; J. March, "Advanced Organic Chemistry: Reactions, Mechanisms and Structure" , 4th Ed., Wiley-Interscience, New York, 1992. Additional suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds described herein, or provide references to articles that describe the preparation, include for example, Fuhrhop, J. and Penzlin G. "Organic Synthesis: Concepts, Methods, Starting Materials" , Second, Revised and Enlarged Edition (1994) John Wiley & Sons ISBN: 3-527-
29074-5; Hoffman, R.V. "Organic Chemistry, An Intermediate Text" (1996) Oxford University Press, ISBN 0-19-509618-5; Larock, R.C. "Comprehensive Organic Transformations: A Guide to Functional Group Preparations" 2nd Edition (1999) Wiley-VCH, ISBN: 0-471-19031-4; March, J. "Advanced Organic Chemistry: Reactions, Mechanisms, and Structure" 4th Edition (1992) John Wiley & Sons, ISBN: 0-471-60180-2; Otera, J. (editor) "Modern Carbonyl Chemistry" (2000) Wiley-VCH, ISBN: 3-527-29871-1; Patai, S. "Patai's 1992 Guide to the Chemistry of Functional Groups" (1992) Interscience ISBN: 0-471-93022-9; Solomons, T.W. G. "Organic Chemistry" 7th Edition (2000) John Wiley & Sons, ISBN: 0-471-19095-0; Stowell, J.C., "Intermediate Organic Chemistry" 2nd Edition (1993) Wiley-Interscience, ISBN: 0-471-57456-2; "Industrial Organic Chemicals: Starting Materials and Intermediates: An Ullmann's Encyclopedia" (1999) John Wiley & Sons, ISBN: 3-527-29645-X, in 8 volumes; "Organic Reactions" (1942-2000) John Wiley & Sons, in over 55 volumes; and "Chemistry of Functional Groups" John Wiley & Sons, in 73 volumes.
Specific and analogous reactants are optionally identified through the indices of known chemicals prepared by the Chemical Abstract Service of the American Chemical Society, which are available in most public and university libraries, as well as through on-line databases (contact the American Chemical Society, Washington, D.C. for more details) . Chemicals that are known but not commercially available in catalogs are optionally prepared by custom chemical synthesis houses, where many of the standard chemical supply houses (e.g., those listed above) provide custom synthesis services. A reference useful for the preparation and selection of pharmaceutical salts of the compounds described herein is P.H. Stahl & C.G. Wermuth "Handbook of Pharmaceutical Salts" , Verlag Helvetica Chimica Acta, Zurich, 2002.
Pharmaceutical Compositions
In certain embodiments, the heteroaromatic WEE1 degradation compound described herein is administered as a pure chemical. In other embodiments, the heteroaromatic WEE1 degradation compound described herein is combined with a pharmaceutically suitable or acceptable carrier (also referred to herein as a pharmaceutically suitable (or acceptable) excipient, physiologically suitable (or acceptable) excipient, or physiologically suitable (or acceptable) carrier) selected on the basis of a chosen route of administration and standard pharmaceutical practice as described, for example, in Remington: The Science and Practice of Pharmacy (Gennaro, 21st Ed. Mack Pub. Co., Easton, PA (2005) ) .
Provided herein is a pharmaceutical composition comprising at least one heteroaromatic WEE1 degradation compound as described herein, or a stereoisomer, pharmaceutically acceptable salt, hydrate, or solvate thereof, together with one or more pharmaceutically acceptable carriers. The carrier (s) (or excipient (s) ) is acceptable or suitable if the carrier is
compatible with the other ingredients of the composition and not deleterious to the recipient (i.e., the subject or the patient) of the composition.
One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable excipient and a compound of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , or a pharmaceutically acceptable salt or solvate thereof.
One embodiment provides a method of preparing a pharmaceutical composition comprising mixing a compound of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable carrier.
In certain embodiments, the heteroaromatic WEE1 degradation compound as described by Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , or a pharmaceutically acceptable salt or solvate thereof, is substantially pure, in that it contains less than about 5%, or less than about 1%, or less than about 0.1%, of other organic small molecules, such as unreacted intermediates or synthesis by-products that are created, for example, in one or more of the steps of a synthesis method.
Suitable oral dosage forms include, for example, tablets, pills, sachets, or capsules of hard or soft gelatin, methylcellulose or of another suitable material easily dissolved in the digestive tract. In some embodiments, suitable nontoxic solid carriers are used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like. (See, e.g., Remington: The Science and Practice of Pharmacy (Gennaro, 21st Ed. Mack Pub. Co., Easton, PA (2005) ) .
In some embodiments, the heteroaromatic WEE1 degradation compound as described by Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , or pharmaceutically acceptable salt or solvate
thereof, is formulated for administration by injection. In some instances, the injection formulation is an aqueous formulation. In some instances, the injection formulation is a non-aqueous formulation. In some instances, the injection formulation is an oil-based formulation, such as sesame oil, or the like.
The dose of the composition comprising at least one heteroaromatic WEE1 degradation compound as described herein differs depending upon the subject or patient's (e.g., human) condition. In some embodiments, such factors include general health status, age, and other factors.
Pharmaceutical compositions are administered in a manner appropriate to the disease to be treated (or prevented) . An appropriate dose and a suitable duration and frequency of administration will be determined by such factors as the condition of the patient, the type and severity of the patient's disease, the particular form of the active ingredient, and the method of administration. In general, an appropriate dose and treatment regimen provides the composition (s) in an amount sufficient to provide therapeutic and/or prophylactic benefit (e.g., an improved clinical outcome, such as more frequent complete or partial remissions, or longer disease-free and/or overall survival, or a lessening of symptom severity. Optimal doses are generally determined using experimental models and/or clinical trials. The optimal dose depends upon the body mass, weight, or blood volume of the patient.
Methods of Treatment
One embodiment provides a compound of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , or a pharmaceutically acceptable salt or solvate thereof, for use in a method of treatment of the human or animal body.
One embodiment provides a compound of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , or a pharmaceutically acceptable salt or solvate thereof, for use in a method of treatment of cancer or neoplastic disease.
One embodiment provides a use of a compound of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-
2) , or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for the treatment of cancer or neoplastic disease.
In some embodiments, described herein is a method of treating cancer in a patient in need thereof comprising administering to the patient a compound of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , or a pharmaceutically acceptable salt or solvate thereof. In some embodiments, described herein is a method of treating cancer in a patient in need thereof comprising administering to the patient a pharmaceutical composition comprising a compound of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (1-3-R) , Formula (1-3a-R) , Formula (1-3b-R) , Formula (I-3c-R) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , Formula (III-2) , Formula (IV) , Formula (IV-1) , or Formula (IV-2) , or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient.
Provided herein is the method wherein the pharmaceutical composition is administered orally. Provided herein is the method wherein the pharmaceutical composition is administered by injection.
Other embodiments and uses will be apparent to one skilled in the art in light of the present disclosures. The following examples are provided merely as illustrative of various embodiments and shall not be construed to limit the invention in any way.
EXAMPLES
I. Chemical Synthesis
In some embodiments, the compounds disclosed herein are synthesized according to the following examples. As used below, and throughout the description of the invention, the following abbreviations, unless otherwise indicated, shall be understood to have the following meanings:
℃ degrees Celsius
δH chemical shift in parts per million downfield from tetramethylsilane
DCM dichloromethane (CH2Cl2)
DMF dimethylformamide
DMSO dimethylsulfoxide
EA ethyl acetate
ESI electrospray ionization
Et ethyl
g gram (s)
h hour (s)
HPLC high performance liquid chromatography
Hz hertz
J coupling constant (in NMR spectrometry)
LCMS liquid chromatography mass spectrometry
μ micro
m multiplet (spectral) ; meter (s) ; milli
M molar
M+ parent molecular ion
Me methyl
MHz megahertz
min minute (s)
mol mole (s) ; molecular (as in mol wt)
mL milliliter
MS mass spectrometry
nm nanometer (s)
NMR nuclear magnetic resonance
pH potential of hydrogen; a measure of the acidity or basicity of an aqueous
solution
PE petroleum ether
RT room temperature
s singlet (spectral)
t triplet (spectral)
T temperature
TFA trifluoroacetic acid
THF tetrahydrofuran
DIEA diisopropylethyl amine
HATU O- (7-Azabenzotriazol-1-yl) -N, N, N, N-tetramethyl uronium
hexafluorophosphate
℃ degrees Celsius
δH chemical shift in parts per million downfield from tetramethylsilane
DCM dichloromethane (CH2Cl2)
DMF dimethylformamide
DMSO dimethylsulfoxide
EA ethyl acetate
ESI electrospray ionization
Et ethyl
g gram (s)
h hour (s)
HPLC high performance liquid chromatography
Hz hertz
J coupling constant (in NMR spectrometry)
LCMS liquid chromatography mass spectrometry
μ micro
m multiplet (spectral) ; meter (s) ; milli
M molar
M+ parent molecular ion
Me methyl
MHz megahertz
min minute (s)
mol mole (s) ; molecular (as in mol wt)
mL milliliter
MS mass spectrometry
nm nanometer (s)
NMR nuclear magnetic resonance
pH potential of hydrogen; a measure of the acidity or basicity of an aqueous
solution
PE petroleum ether
RT room temperature
s singlet (spectral)
t triplet (spectral)
T temperature
TFA trifluoroacetic acid
THF tetrahydrofuran
DIEA diisopropylethyl amine
HATU O- (7-Azabenzotriazol-1-yl) -N, N, N, N-tetramethyl uronium
hexafluorophosphate
Example 1: 3- (5- {1- [1- (2- {4- [4- ( {1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperazin-1-yl} ethyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione (Cpd-001)
Cpd-001 was synthesized according to the following scheme:
Step A: tert-butyl 4- {4- [2- (2, 6-dioxopiperidin-3-yl) -1-oxo-3H-isoindol-5-yl] pyrazol-1-
yl} piperidine-1-carboxylate
A solution of tert-butyl 4- [4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazol-1-yl] piperidine-1-carboxylate (7 g, 18.553 mmol) and 3- (5-bromo-1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione (3.00 g, 9.277 mmol) , KOAc (2.70 g, 27.829 mmol) in
dimethylformamide (20 mL) was stirred at 90℃ for 16 h under N2 atmosphere. after cooling down to the room temperature, saturated sodium chloride solution (200mL) was added and the resulting mixture was extracted by EA (500 mL x 3) . The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with MeOH/DCM (0%-15%) to afford the title compound.
LCMS: MS (ESI) m/z 494.25 [M+H] +.
Step B: 3- {1-oxo-5- [1- (piperidin-4-yl) pyrazol-4-yl] -3H-isoindol-2-yl} piperidine-2, 6-dione
A solution of tert-butyl 4- {4- [2- (2, 6-dioxopiperidin-3-yl) -1-oxo-3H-isoindol-5-yl] pyrazol-1-yl} piperidine-1-carboxylate (3.1 g, 6.281 mmol) in hydrogen chloride in EA (15 mL) was stirred at 22℃ for 24 h under H2 atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was diluted with EA (200 mL) , and then concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z 394.25 [M+H] +.
Step C: 3- (5- {1- [1- (2-hydroxyethyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-
yl) piperidine-2, 6-dione
A solution of 3- {1-oxo-5- [1- (piperidin-4-yl) pyrazol-4-yl] -3H-isoindol-2-yl} piperidine-2, 6-dione (500 mg) , DIEA (492.75 mg, 3.813 mmol) and 2-bromo-1, 1-diethoxyethane (15.03 mg, 0.076 mmol) in dimethylformamide (1 mL) was stirred at 80℃ for 16 h. The residue was purified by reverse flash chromatography (MeCN/H2O (0.5%TFA) from 0-25%) to afford the title compound.
LCMS: MS (ESI) m/z 438.25 [M+H] +.
Step D: 3- (5- {1- [1- (2-chloroethyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-
yl) piperidine-2, 6-dione
A solution of 3- (5- {1- [1- (2-hydroxyethyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione (200 mg, 0.457 mmol) , thionyl chloride (81.57 mg, 0.685 mmol) in DCE (10 mL) was stirred at 80℃ for 5 h under N2 atmosphere. After filtration, the filtrate was concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z 456.20 [M+H] +.
Step E: 3- (5- {1- [1- (2- {4- [4- ( {1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -
3-oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperazin-1-
yl} ethyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione
A solution of 3- (5- {1- [1- (2-chloroethyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione (100 mg) , 1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -6- { [4- (piperazin-1-yl) phenyl] amino} -2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-3-one (75 mg,
0.146 mmol) and DIEA (85.04 mg, 0.657 mmol) in dimethylformamide (4 mL) was stirred at 80℃ for 16 h. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions [Column: Xselect CSH C18 OBD Column 30*150mm 5μm, n; Mobile Phase A: Water (0.1%FA) , Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 3%B to 32%B in 10 min, 32%B; Wave Length: 254 nm; RT1(min) : 8.28; Number Of Runs: 0] to afford the title compound.
LCMS: MS (ESI) m/z 932.25 [M+H] +.
1H NMR (300 MHz, DMSO-d6) : δ 11.00 (s, 1H) , 10.14 (s, 1H) , 8.83 (s, 1H) , 8.43 (s, 1H) , 8.02 (s, 1H) , 7.94 (d, J = 8.3 Hz, 1H) , 7.83 (s, 1H) , 7.78 –7.65 (m, 3H) , 7.59 (s, 2H) , 6.93 (d, J = 8.6 Hz, 2H) , 5.77 –5.58 (m, 1H) , 5.18 –5.02 (m, 2H) , 5.01 –4.81 (m, 3H) , 4.75 (s, 1H) , 4.64 –4.53 (m, 1H) , 4.47 (d, J = 17.3 Hz, 1H) , 4.33 (d, J = 17.1 Hz, 1H) , 4.23 –4.10 (m, 1H) , 3.19 –3.07 (m, 5H) , 3.06 –2.90 (m, 5H) , 2.89 –2.71 (m, 3H) , 2.67 –2.52 (m, 6H) , 2.49 –2.37 (m, 1H) , 2.29 –2.10 (m, 4H) , 2.09 –1.81 (m, 7H) , 1.78 –1.65 (m, 1H) .
Example 2: 3- (5- {1- [1- ( {1- [4- ( {1- [ (7R) -7-Ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperidin-4-yl} methyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione (Cpd-002)
Cpd-002 was synthesized according to the following scheme:
Step A: [1- (4-Nitrophenyl) piperidin-4-yl] methanol
A solution of 4-fluoronitrobenzene (7 g, 49.61 mmol) , piperidin-4-ylmethanol (8.57 g, 74.42 mmol) and K2CO3 (10.28 g, 74.42 mmol) in ACN (30 mL) was stirred at 80℃ for 12 h. After cooling down to room temperature, the resulting mixture was diluted with EA (50 mL) . The organic layer was washed with H2O (50 mL x 3) , dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA/PE (0-100%) to afford the title compound.
LCMS: MS (ESI) m/z 237.52 [M+H] +.
Step B: 1- (4-Nitrophenyl) piperidine-4-carbaldehyde
A solution of [1- (4-nitrophenyl) piperidin-4-yl] methanol (1 g, 4.232 mmol) and Dess-Martin periodinane (3.59 g, 8.464 mmol) in DCM (20 mL) was stirred at 25℃ for 12 h. The resulting mixture was diluted with DCM (50 mL) . The organic layer was washed with H2O (50 mLx3) , dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with MeOH/DCM (0-20%) to afford the title compound.
LCMS: MS (ESI) m/z 235.22 [M+H] +.
Step C: 3- {5- [1- (1- { [1- (4-Nitrophenyl) piperidin-4-yl] methyl} piperidin-4-yl) pyrazol-4-yl] -1-
oxo-3H-isoindol-2-yl} piperidine-2, 6-dione
To a solution of 1- (4-nitrophenyl) piperidine-4-carbaldehyde (1.66 g, 7.065 mmol) in MeOH (27.78 mL) was added 3- {1-oxo-5- [1- (piperidin-4-yl) pyrazol-4-yl] -3H-isoindol-2-yl} piperidine-2, 6-dione (2 g, 5.083 mmol) . The mixture was stirred at 60℃ for 1 h, followed by the addition of NaBH3CN (0.96 g, 15.249 mmol) in portions at 0℃. The resulting solution was stirred at 22℃ for 2 hrs. The resulting mixture was diluted with EA (50 mL) . The organic layer was washed with H2O (50 mL x 3) , dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA/PE (0-100%) to afford the title compound.
LCMS: MS (ESI) m/z 612.33 [M+H] +
Step D: 3- {5- [1- (1- { [1- (4-Aminophenyl) piperidin-4-yl] methyl} piperidin-4-yl) pyrazol-4-yl] -
1-oxo-3H-isoindol-2-yl} piperidine-2, 6-dione
A suspension of 3- {5- [1- (1- { [1- (4-nitrophenyl) piperidin-4-yl] methyl} piperidin-4-yl) pyrazol-4-yl] -1-oxo-3H-isoindol-2-yl} piperidine-2, 6-dione (200 mg, 0.327 mmol) and Pd/C (100 mg, 10%w/w? ) in MeOH (1 mL) was stirred at 25℃ for 1 h. The resulting mixture was concentrated under reduced pressure. The crude product was used in the next step directly without further purification.
LCMS: MS (ESI) m/z 582.32 [M+H] +
Step E: 3- (5- {1- [1- ( {1- [4- ( {1- [ (7R) -7-Ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-
oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperidin-4-
yl} methyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione
A solution of 3- {5- [1- (1- { [1- (4-aminophenyl) piperidin-4-yl] methyl} piperidin-4-yl) pyrazol-4-yl] -1-oxo-3H-isoindol-2-yl} piperidine-2, 6-dione (20 mg) , 1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -6-methanesulfinyl-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-3-one (13.73 mg, 0.034 mmol) and DIEA (13.33 mg, 0.102 mmol) in toluene (0.5 mL) at 80℃ was stirred for 6 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column, 19*150 mm, 5μm; Mobile Phase A: Water (0.05%TFA ) , Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 12%B to 42%B in 7 min, 42%B; Wave Length: 254 nm; RT1(min) : 6.23; Number Of Runs: 0) to afford the title compound.
LCMS: MS (ESI) m/z 917.20 [M+H] +.
1H NMR (400 MHz, DMSO-d6) : δ 11.02 (s, 1H) , 10.20 (s, 1H) , 9.27 (s, 1H) , 8.86 (s, 1H) , 8.42 (s, 1H) , 8.12 (d, J = 8.1 Hz, 1H) , 7.93 (d, J = 8.2 Hz, 1H) , 7.86 (d, J = 6.6 Hz, 1H) , 7.83 –7.75 (m, 1H) , 7.78 –7.67 (m, 2H) , 7.65 (s, 2H) , 7.07 (s, 2H) , 5.68 –2.35 (m, 1H) , 5.13-2.08 (m, 1H) , 5.01 (dd, J = 10.2, 1.5 Hz, 1H) , 4.98 -4.86 (m, 1H) , 4.75 (s, 1H) , 4.62-4.57 (m, 2H) , 4.52-4.48 (m, 1H) , 4.35-4.23 (m, 1H) , 3.19 (s, 5H) , 3.11 (s, 2H) , 3.04 –2.87 (m, 3H) , 2.85 –2.73 (m, 4H) ,
2.62-2.54 (m, 1H) , 2.43-2.38 (m, 1H) , 2.32 (d, J = 8.1 Hz, 4H) , 2.21-2.12 (m, 1H) , 2.10-2.03 (m, 3H) , 1.95 –1.86 (m, 3H) , 1.72-1.56 (m, 1H) , 1.42 (s, 2H) , 0.87 (q, J = 8.3, 7.8 Hz, 3H) .
Example 3: 3- (6- {1- [1- (2- {4- [4- ( {1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperazin-1-yl} ethyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione (Cpd-003)
Cpd-003 was synthesized according to the following scheme:
Step A: tert-butyl 4- {4- [2- (2, 6-dioxopiperidin-3-yl) -3-oxo-1H-isoindol-5-yl] pyrazol-1-
yl} piperidine-1-carboxylate
To a stirred solution of tert-butyl 4- [4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazol-1-yl] piperidine-1-carboxylate (2 g, 5.301 mmol) , 3- (6-bromo-1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione (0.86 g, 2.651 mmol) and KOAc (0.78 g, 7.952 mmol) in DMF (20 mL) was added Pd (dppf) Cl2 (0.19 g, 0.265 mmol) at 22℃ under N2 atmosphere. The resulting mixture was stirred at 90℃ for 16 hrs. The resulting mixture was diluted with EA (100 mL) . The resulting mixture was extracted with water (50 mL x 3) . The combined organic layers was washed with NaCl (50 mL) , dried over anhydrous NaSO4. After filtration, the filtrate was concentrated under
reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA/PE (0~50%) to afford the title compound.
LCMS: MS (ESI) m/z 494 [M+H] +.
Step B: 3- {1-oxo-6- [1- (piperidin-4-yl) pyrazol-4-yl] -3H-isoindol-2-yl} piperidine-2, 6-dione
A solution of tert-butyl 4- {4- [2- (2, 6-dioxopiperidin-3-yl) -3-oxo-1H-isoindol-5-yl] pyrazol-1-yl} piperidine-1-carboxylate (1 g, 2.026 mmol) and HCl (5 mL, 4N) in EA (10 mL) was stirred for 1 h at 22℃. The precipitated solid was collected by filtration and washed with EA (20 mL) to afford the title compound.
LCMS: MS (ESI) m/z 394 [M+H] +.
Step C: 3- (6- {1- [1- (2-hydroxyethyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-
yl) piperidine-2, 6-dione
To a stirred solution of 3- {1-oxo-6- [1- (piperidin-4-yl) pyrazol-4-yl] -3H-isoindol-2-yl} piperidine-2, 6-dione (700 mg) and 2-bromoethanol (266.80 mg, 2.135 mmol) in DMF (5 mL) was added DIEA (1149.75 mg, 8.895 mmol) dropwise at 22℃. The resulting mixture was stirred at 80℃for additional 16 hrs. The residue was purified by reversed-phase flash chromatography (column, C18 silica gel; mobile phase, water (0.05%TFA) in ACN, 0%to 50%gradient in 1 h; detector, UV 254 nm) to afford the title compound.
LCMS: MS (ESI) m/z 438 [M+H] +.
Step D: 3- (6- {1- [1- (2-chloroethyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-
yl) piperidine-2, 6-dione
A solution of 3- (6- {1- [1- (2-hydroxyethyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione (300 mg, 0.686 mmol) and SOCl2 (407.86 mg, 3.430 mmol) in DCE (10.00 mL, 126.382 mmol) was stirred for 16 hrs at 80℃. The resulting mixture was diluted with EA (50 mL) . The resulting mixture was washed with water (20mL x 3) . The organic layer was washed with NaCl (20 mL) , dried over anhydrous NaSO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA/PE (0~100%) to afford the title compound.
LCMS: MS (ESI) m/z 456 [M+H] +.
Step E: 3- (6- {1- [1- (2- {4- [4- ( {1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -
3-oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperazin-1-
yl} ethyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione
To a stirred solution of 3- (6- {1- [1- (2-chloroethyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione (50 mg, 0.110 mmol) and 1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -6- { [4- (piperazin-1-yl) phenyl] amino} -2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-3-one (37.48 mg, 0.073 mmol) in DMF (3 mL) was added DIEA
(37.80 mg, 0.293 mmol) at 22℃ . The resulting mixture was stirred at 80℃ for additional 16 h. The mixture was concentrated under vacuum and purified by Prep-HPLC with the following condition (Column: Xselect CSH C18 OBD Column 30*150mm 5μm, n; Mobile Phase A: Water (0.1%FA) , Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 3%B to 32%B in 10 min, 32%B; Wave Length: 254 nm; RT1 (min) : 9.38; Number Of Runs: 0) to afford the title compound.
LCMS: MS (ESI) m/z 932 [M+H] +.
1H NMR (400 MHz, DMSO-d6) : δ 11.01 (s, 1H) , 10.15 (s, 1H) , 8.83 (s, 1H) , 8.45 (s, 1H) , 8.03 (s, 1H) , 7.96-7.94 (m, 2H) , 7.87 (dd, J = 7.9, 1.7 Hz, 1H) , 7.70 (d, J = 8.1 Hz, 1H) , 7.60 (t, J = 7.4 Hz, 3H) , 6.93 (d, J = 8.7 Hz, 2H) , 5.71-5.65 (m, 1H) , 5.21-5.15 (m, 1H) , 5.07 (s, 1H) , 5.04-5.00 (m, 1H) , 4.90 –4.81 (m, 1H) , 4.76 (s, 1H) , 4.58 (s, 1H) , 4.52-4.43 (m, 1H) , 4.38-4.30 (m, 1H) , 4.21-4.15 (m, 1H) , 3.15 –3.05 (m, 4H) , 3.04 (s, 2H) , 3.00 –2.90 (m, 2H) , 2.81-2.75 (m, 1H) , 2.71-2.52 (m, 9H) , 2.43-2.39 (m, 1H) , 2.25-2.16 (m, 3H) , 2.13-2.00 (m, 6H) , 1.98-1.87 (m, 1H) , 1.73-1.69 (m, 1H) , 0.88 (t, J = 7.4 Hz, 3H) .
Example 4: 3- (6- {1- [1- (2- {4- [4- ( {1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperazin-1-yl} acetyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione (Cpd-004)
Scheme:
Step A: Methyl 2- {4- [4- ( {1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-
oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperazin-1-yl} acetate
To a stirred solution of 1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -6- { [4- (piperazin-1-yl) phenyl] amino} -2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-3-one (150 mg, 0.293 mmol) in DMF (5 mL) were added 1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -6- { [4- (piperazin-1-yl) phenyl] amino} -2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-3-one (150 mg, 0.293 mmol) and DIEA (113.46 mg, 0.879 mmol) at 22℃. The mixture was stirred at 22℃ for 15 h. The reaction mixture was diluted with EA (60 mL) and washed with brine (aq. 10 mL) , the organic phase was separated and dried over anhydrous sodium sulfate, then concentrated under reduced pressure to give crude product, which was further purified by column chromatography using EA/PE (0 -30%) to afford the title compound.
LCMS: MS (ESI) m/z 585.50 [M+H] +.
Step B: {4- [4- ( {1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-oxo-2- (prop-
2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperazin-1-yl} acetic acid
To a solution of methyl 2- {4- [4- ( {1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperazin-1-yl} acetate
(65 mg, 0.111 mmol) in THF (1 mL) and H2O (0.5 mL) was added LiOH (0.15 mL, 3M) at 22℃. The mixture was stirred at 22℃ for 1 h. The resulting mixture was concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z 394.15 [M+H] +.
Step C: 3- (6- {1- [1- (2- {4- [4- ( {1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -
3-oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperazin-1-
yl} acetyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione
To a solution of {4- [4- ( {1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperazin-1-yl} acetic acid (60 mg) in anhydrous DMF (2 mL) was added 3- {1-oxo-6- [1- (piperidin-4-yl) pyrazol-4-yl] -3H-isoindol-2-yl} piperidine-2, 6-dione (53.78 mg, 0.137 mmol) and DIEA (40.77 mg, 0.315 mmol) . The reaction mixture was stirred at 22℃ for 2 h. The reaction mixture was diluted with DCM (5 mL) . The organic phase was washed with brine NaCl (aq. 5 mL) , dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was further purified by Prep-HPLC (Column: XSelect CSH Prep C18 OBD Column, 19*250 mm, 5μm; Mobile Phase A: Water (0.1%FA) , Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 21%B to 51%B in 7 min, 51%B; Wave Length: 254 nm; RT1 (min) : 6; Number Of Runs: 0) to afford the title compound.
LCMS: MS (ESI) m/z 946.35 [M+H] +.
1H NMR (400 MHz, DMSO-d6) : δ 11.01 (s, 1H) , 10.15 (s, 1H) , 8.83 (s, 1H) , 8.46 (s, 1H) , 8.04 (s, 1H) , 7.99 –7.90 (m, 2H) , 7.89 –7.84 (m, 1H) , 7.70 (d, J = 8.2 Hz, 1H) , 7.58 (d, J = 8.0 Hz, 3H) , 6.95 (d, J = 8.6 Hz, 2H) , 5.81 –5.59 (m, 1H) , 5.21 –5.07 (m, 1H) , 5.06 (s, 1H) , 5.03 –4.91 (m, 1H) , 4.90 –4.71 (m, 2H) , 4.70 (s, 1H) , 4.50 –4.36 (m, 3H) , 4.23 (s, 2H) , 3.25 –3.00 (m, 6H) , 2.99 –2.86 (m, 2H) , 2.83 –2.70 (m, 2H) , 2.65 –2.58 (m, 4H) , 2.47 –2.31 (m, 2H) , 2.30 –2.06 (m, 4H) , 2.00 (s, 3H) , 1.96 –1.89 (m, 7.3 Hz, 1H) , 1.84 (s, 1H) , 1.76-1.68 (m, 1H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 5: 3- (6- {1- [1- ( {1- [4- ( {1- [ (7R) -7-Ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperidin-4-yl} methyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione (Cpd-005)
Cpd-005 was synthesized according to the following scheme:
Step A: Ethyl 3- {6- [1- (1- { [1- (4-nitrophenyl) piperidin-4-yl] methyl} piperidin-4-yl) pyrazol-4-
yl] -1-oxo-3H-isoindol-2-yl} piperidine-2, 6-dione
To a stirred solution of 1- (4-nitrophenyl) piperidine-4-carbaldehyde (1.01 g, 4.295 mmol) in DCE (6 mL) and DMA (6 mL) were added TEA (0.67 g, 6.608 mmol) , 3- {1-oxo-6- [1- (piperidin-4-yl) pyrazol-4-yl] -3H-isoindol-2-yl} piperidine-2, 6-dione (1.3 g, 3.304 mmol) at 22℃and stirred for 0.5 h. Then to the above mixture was added NaBH3CN (0.42 g, 6.608 mmol) at 0℃. The resulting mixture was stirred at 22℃ for 3 hrs. The reaction was quenched with 15 mL NaHCO3 (aq) . The resulting mixture was extracted with EtOAc (15 mL x 3) . The combined
organic layers was washed with NaCl (aq. 20 mL x 3) then concentrated. The residue obtained was purified by silica gel chromatography (0-15%MeOH/DCM) to afford the title compound.
LCMS: MS (ESI) m/z 612.35 [M+H] +
Step B: 3- {6- [1- (1- { [1- (4-Aminophenyl) piperidin-4-yl] methyl} piperidin-4-yl) pyrazol-4-yl] -
1-oxo-3H-isoindol-2-yl} piperidine-2, 6-dione
To a stirred solution of 3- {6- [1- (1- { [1- (4-nitrophenyl) piperidin-4-yl] methyl} piperidin-4-yl) pyrazol-4-yl] -1-oxo-3H-isoindol-2-yl} piperidine-2, 6-dione (600 mg, 0.981 mmol) in MeOH (10 mL) was acidified to pH=7 with HCl (aq) . Then to the above mixture was added Pd/C (85.72 mg, 10%w/w? ) and stirred under hydrogen atmosphere at 22℃ for 1 h. The resulting mixture was filtered, the filter cake was washed with methanol (20 mL x 3) . The filtrate was concentrated under reduced pressure. The crude was used in the next step directly without further purification.
LCMS: MS (ESI) m/z 582.30 [M+H] +.
Step C: 3- (6- {1- [1- ( {1- [4- ( {1- [ (7R) -7-Ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-
oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperidin-4-
yl} methyl) piperidin-4-yl] pyrazol-4-yl} -1-oxo-3H-isoindol-2-yl) piperidine-2, 6-dione
To a solution of 3- {6- [1- (1- { [1- (4-aminophenyl) piperidin-4-yl] methyl} piperidin-4-yl) pyrazol-4-yl] -1-oxo-3H-isoindol-2-yl} piperidine-2, 6-dione (50 mg) and 1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -6-methanesulfinyl-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-3-one (34.34 mg, 0.086 mmol) in CHCl3 (3 mL) was added DIEA (33.33 mg, 0.258 mmol) . The resulting mixture was stirred at 80℃ for 12 h. The reaction was purified by reverse phase HPLC (Column: XBridge Shield RP18 OBD Column, 19*250 mm, 10μm; Mobile Phase A: Water (0.1%FA) , Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 17%B to 47%B in 7 min, 47%B; Wave Length: 254 nm; RT1 (min) : 6; Number Of Runs: 0) to afford the title compound.
LCMS: MS (ESI) m/z 917.40 [M+H] +.
1H NMR (300 MHz, DMSO-d6) : δ 11.01 (s, 1H) , 10.16 (s, 1H) , 8.85 (s, 1H) , 8.44 (s, 1H) , 8.12 (d, J = 6.9 Hz, 1H) , 8.00 (d, J = 8.2 Hz, 1H) , 7.85-7.96 (m, 2H) , 7.70 (d, J = 8.2 Hz, 1H) , 7.56-7.67 (m, 3H) , 7.02 –7.17 (m, 2H) , 5.57-5.75 (m, 1H) , 5.09-5.20 (m, 1H) , 5.01 (d, J = 10.1 Hz, 1H) , 4.87 (d, J = 17.1 Hz, 1H) , 4.76 (d, J = 14.9 Hz, 1H) , 4.60 (d, J = 6.8 Hz, 1H) , 4.33 (d, J = 17.2 Hz, 1H) , 3.71 (s, 4H) , 3.19 (s, 2H) , 3.11 (s, 2H) , 2.09 –3.01 (m, 1H) , 2.74 –2.94 (m, 3H) , 2.62 (d, J = 17.5 Hz, 1H) , 2.38 –2.55 (m, 5H) , 2.35 (s, 4H) , 2.14 –2.27 (m, 1H) , 1.84-2.09 (m, 6H) , 1.64-1.78 (m, 1H) , 1.43 (d, J = 11.6 Hz, 2H) , 0.83-0.93 (m, 3H) .
Example 6: 5- (1- (1- ( (1- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperidin-4-yl) methyl) piperidin-4-yl) -1H-pyrazol-4-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Cpd-006)
Cpd-006 was synthesized according to the following scheme:
Step A: Tert-butyl 4- {4- [2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindol-5-yl] pyrazol-1-
yl} piperidine-1-carboxylate
To a stirred solution of tert-butyl 4- [4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazol-1-yl] piperidine-1-carboxylate (4.36 g, 11.569 mmol) and 5-bromo-2- (2, 6-dioxopiperidin-3-yl) isoindole-1, 3-dione (3 g, 8.899 mmol) was added KOAc (1.75 g, 17.798 mmol, 2 equiv) . The resulting mixture was stirred at 100℃ for 16 h. The reaction was quenched with 15 mL H2O at 22℃. The resulting mixture was extracted with EtOAc (15 mL x 3) . The combined organic
layers was washed with NaCl (aq. 15 mL x 3) , dried over anhydrous Na2SO4. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2 /MeOH (10: 1) to afford the title compound.
LCMS: MS (ESI) m/z 508.20 [M+H] +
Step B: 2- (2, 6-Dioxopiperidin-3-yl) -5- [1- (piperidin-4-yl) pyrazol-4-yl] isoindole-1, 3-dione
A solution of tert-butyl 4- {4- [2- (2, 6-dioxopiperidin-3-yl) -1, 3-dioxoisoindol-5-yl] pyrazol-1-yl} piperidine-1-carboxylate (2.2 g, 4.335 mmol) in HCl (25.00 mL, 4M gas in 1, 4-dioxane) was stirred at 22℃ for 1 h. The precipitated solids were collected by filtration and washed with EtOAc (15 mL x 3) . The crude product was used in the next step directly without further purification.
LCMS: MS (ESI) m/z 408.15 [M+H] +.
Step C: Ethyl 2- (2, 6-dioxopiperidin-3-yl) -5- [1- (1- { [1- (4-nitrophenyl) piperidin-4-
yl] methyl} piperidin-4-yl) pyrazol-4-yl] isoindole-1, 3-dione
To a stirred solution of 1- (4-nitrophenyl) piperidine-4-carbaldehyde (0.57 g, 2.454 mmol) in DCE (6 mL) and DMA (6 mL) were added TEA (496.74 mg, 4.908 mmol) , 2- (2, 6-dioxopiperidin-3-yl) -5- [1- (piperidin-4-yl) pyrazol-4-yl] isoindole-1, 3-dione (1 g) at 22℃ and stirred for 0.5 h. Then to the above mixture was added NaBH3CN (308.47 mg, 4.908 mmol) at 0℃. The resulting mixture was stirred at 22℃ for 12 h. The reaction was quenched with 15 mL NaHCO3 (aq. ) . The resulting mixture was extracted with EtOAc (15 mL x 3) . The combined organic layers was washed with NaCl (aq. 20 mL x 3) then concentrated. The residue obtained was purified by silica gel chromatography (0-15%MeOH/DCM) to afford the title compound.
LCMS: MS (ESI) m/z 626.35 [M+H] +
Step D: 5- [1- (1- { [1- (4-Aminophenyl) piperidin-4-yl] methyl} piperidin-4-yl) pyrazol-4-yl] -2-
(2, 6-dioxopiperidin-3-yl) isoindole-1, 3-dione
To a stirred solution of 2- (2, 6-dioxopiperidin-3-yl) -5- [1- (1- { [1- (4-nitrophenyl) piperidin-4-yl] methyl} piperidin-4-yl) pyrazol-4-yl] isoindole-1, 3-dione (600 mg, 0.959 mmol) in MeOH (10 mL) was acidified to pH=7 with HCl (aq. ) . Pd/C (85.72 mg, 10%w/w? ) was added and the mixture was stirred under hydrogen atmosphere at 22℃ for 1 h. The resulting mixture was filtered, the filter cake was washed with methanol (20 mL x 3) . The filtrate was concentrated under reduced pressure. The crude product was purified by reverse phase HPLC (Column: XBridge Shield RP18 OBD Column, 19*250 mm, 10μm; Mobile Phase A: Water (0.1%FA) , Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 5%B to 30%B in 7 min, 30%B; Wave Length: 254 nm; RT1 (min) : 6; Number Of Runs: 0) to afford the title compound.
LCMS: MS (ESI) m/z 596.35 [M+H] +
Step E: 5- (1- (1- ( (1- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperidin-4-yl) methyl) piperidin-4-yl) -1H-pyrazol-4-yl) -2- (2, 6-
dioxopiperidin-3-yl) isoindoline-1, 3-dione
To a solution of 5- [1- (1- { [1- (4-aminophenyl) piperidin-4-yl] methyl} piperidin-4-yl) pyrazol-4-yl] -2- (2, 6-dioxopiperidin-3-yl) isoindole-1, 3-dione (100 mg, 0.168 mmol) and 1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -6-methanesulfinyl-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-3-one (67.06 mg, 0.168 mmol) in CHCl3 (3 mL) was added DIEA (65.09 mg, 0.504 mmol) . The resulting mixture was stirred at 80℃ for 12 h. The reaction was purified by reverse phase HPLC (Column: XBridge Shield RP18 OBD Column, 19*250 mm, 10μm; Mobile Phase A: Water (0.1%FA) , Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 17%B to 47%B in 7 min, 47%B; Wave Length: 254 nm; RT1 (min) : 6; Number Of Runs: 0) to afford the title compound.
LCMS: MS (ESI) m/z 931.50 [M+H] +.
1H NMR (300 MHz, DMSO-d6) : δ 11.15 (s, 1H) , 10.12 (s, 1H) , 8.82 (s, 1H) , 8.66 (s, 1H) , 8.06 –8.22 (m, 3H) , 7.85-7.98 (m, 2H) , 7.70 (d, J = 8.1 Hz, 1H) , 7.57 (s, 2H) , 6.93 (d, J = 8.8 Hz, 2H) , 5.58-5.76 (m, 1H) , 5.11-5.22 (m, 1H) , 5.07 (s, 1H) , 5.00 (d, J = 10.1 Hz, 1H) , 4.86 (d, J = 17.3 Hz, 1H) , 4.74 (s, 1H) , 4.52 –4.63 (m, 1H) , 4.18 (s, 1H) , 3.65 (d, J = 11.8 Hz, 3H) , 2.97 (d, J = 9.6 Hz, 6H) , 2.65 (d, J = 10.5 Hz, 2H) , 2.56 (d, J = 13.4 Hz, 2H) , 2.16-2.27 (m, 3H) , 2.05–2.16 (m, 2H) , 2.06 (d, J = 5.9 Hz, 4H) , 2.01 (d, J = 4.3 Hz, 1H) , 1.86 –2.02 (m, 1H) , 1.71 –1.89 (m, 3H) , 1.64 –1.75 (m, 2H) , 0.82-0.93 (m, 3H) .
Example 7: 3- {5- [1- (7- {1- [4- ( {1- [ (7R) -7-Ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperidin-4-yl} -7-azaspiro [3.5] nonan-2-yl) pyrazol-4-yl] -1-oxo-3H-isoindol-2-yl} piperidine-2, 6-dione (Cpd-007)
Cpd-007 was synthesized according to the following scheme:
Step A: 1- (4-Aminophenyl) piperidin-4-one
A solution of 1- (4-nitrophenyl) piperidin-4-one (200 mg, 0.908 mmol) and Raney Ni (38.90 mg) in MeOH (5 mL) was stirred for 1 h at 22℃ under H2 atmosphere. The resulting mixture was filtered, the filter cake was washed with MeOH (10 mL x 3) . The filtrate was concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z 190.11 [M+H] +.
Step B: 1- [ (7R) -7-Ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -6-methanesulfinyl-2-
(prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-3-one
A solution of 1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -6- (methylsulfanyl) -2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-3-one (400 mg, 1.043 mmol) and m-CPBA (306.00 mg, 1.773 mmol) in toluene (2 mL) was stirred at 22℃ for 6 hrs. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with MeOH/DCM (0-20%) to afford the title compound.
LCMS: MS (ESI) m/z 399.13 [M+H] +
Step C: 1- [4- ( {1- [ (7R) -7-Ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-oxo-2- (prop-
2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperidin-4-one
A solution of 1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -6-methanesulfinyl-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-3-one (1 g, 2.503 mmol) and 1- (4-aminophenyl) piperidin-4-one (0.57 g) and DIEA (0.97 g, 7.509 mmol) in toluene (10 mL) was stirred at 80℃ for 2 h. The resulting mixture was diluted with EA (20 mL) . The organic layer was washed with H2O (20 mL x 3) , dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA/PE (0-100%) to afford the title compound.
LCMS: MS (ESI) m/z 526.23 [M+H] +
Step D: Tert-butyl 2- [ (4-methylbenzenesulfonyl) oxy] -7-azaspiro [3.5] nonane-7-carboxylate
A solution of tert-butyl 2-hydroxy-7-azaspiro [3.5] nonane-7-carboxylate (1.5 g, 6.216 mmol) and TsCl (1.42 g, 7.459 mmol) and Et3N (1.89 g, 18.648 mmol, ) in DCM (15 mL) was stirred for 12 h at 25℃. The resulting mixture was diluted with EA (20 mL) . The organic layer was washed with H2O (20 mL x 3) , dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA/PE (0-100%) to afford the title compound.
LCMS: MS (ESI) m/z 396.56 [M+H] +.
Step E: Tert-butyl 4- [4- ( {1- [6- (2-hydroxypropan-2-yl) pyridin-2-yl] -2-isopropyl-3-
oxopyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperazine-1-carboxylate
A solution/mixture of tert-butyl 2- [ (4-methylbenzenesulfonyl) oxy] -7-azaspiro [3.5] nonane-7-carboxylate (800 mg, 2.023 mmol) , 4-iodopyrazole (470.83 mg, 2.428 mmol) and Cs2CO3 (1647.59 mg, 5.058 mmol) in DMF (10 mL) was stirred at 25℃ for 12 h . The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA/PE (0-100%) to afford the title compound.
LCMS: MS (ESI) m/z 418.22 [M+H] +
Step F: Tert-butyl 2- [4- (4, 4, 5-trimethyl-1, 3, 2-dioxaborolan-2-yl) pyrazol-1-yl] -7-
azaspiro [3.5] nonane-7-carboxylate
A solution of tert-butyl 2- (4-iodopyrazol-1-yl) -7-azaspiro [3.5] nonane-7-carboxylate (780 mg, 1.869 mmol) in THF (10 mL, 123.428 mmol) was treated with iPrMgCl. LiCl (5.5 mL, 7.188 mmol, 1.3 M in THF) at -78 ℃ for 30 min under N2 followed by the addition of 2-isopropoxy-4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolane (3.12 g, 16.772 mmol) dropwise at -20℃. The solution was stirred at 25℃ for 1h under N2 atmosphere. The resulting mixture was diluted with EA (20 mL) . The organic layer was washed with aq. NH4Cl (20 mL x 3) , dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA/PE (0-100%) to afford the title compound. LCMS: MS (ESI) m/z 418.22 [M+H] +.
Step G: Tert-butyl 2- {4- [2- (2, 6-dioxopiperidin-3-yl) -1-oxo-3H-isoindol-5-yl] pyrazol-1-yl} -
7-azaspiro [3.5] nonane-7-carboxylate
A solution of tert-butyl 2- [4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazol-1-yl] -7-azaspiro [3.5] nonane-7-carboxylate (1056.87 mg, 2.532 mmol) , 3- [5- (4-bromophenyl) -1-oxo-3H-isoindol-2-yl] piperidine-2, 6-dione (337 mg, 0.844 mmol) , Pd (dppf) Cl2 (61.76 mg, 0.084 mmol) and KOAc (248.52 mg, 2.532 mmol) in DMF (15 mL) was stirred at 110℃ for 12 h under N2 atmosphere. The resulting mixture was diluted with EA (20 mL) . The organic layer was washed with H2O (20 mL x 3) , dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with EA/PE (0-100%) to afford the title compound.
LCMS: MS (ESI) m/z 534.22 [M+H] +
Step H: 3- [5- (1- {7-Azaspiro [3.5] nonan-2-yl} pyrazol-4-yl) -1-oxo-3H-isoindol-2-
yl] piperidine-2, 6-dione
A solution of tert-butyl 2- {4- [2- (2, 6-dioxopiperidin-3-yl) -1-oxo-3H-isoindol-5-yl] pyrazol-1-yl} -7-azaspiro [3.5] nonane-7-carboxylate (230 mg, 0.431 mmol) in HCl (gas) in 1, 4-dioxane (4M, 2 mL) at 25℃ was stirred for 1 h . The precipitated solid was collected by filtration and washed with EA (5 mL) to afford the title compound.
LCMS: MS (ESI) m/z 434.22 [M+H] +.
Step I: 3- {5- [1- (7- {1- [4- ( {1- [ (7R) -7-Ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-
oxo-2- (prop-2-en-1-yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperidin-4-yl} -7-
azaspiro [3.5] nonan-2-yl) pyrazol-4-yl] -1-oxo-3H-isoindol-2-yl} piperidine-2, 6-dione
A solution of 3- [5- (1- {7-azaspiro [3.5] nonan-2-yl} pyrazol-4-yl) -1-oxo-3H-isoindol-2-yl] piperidine-2, 6-dione (196.30 mg) in DCE (1 mL) and DMA (1 mL) was treated with 1- [4- ( {1- [ (7R) -7-ethyl-7-hydroxy-5H, 6H-cyclopenta [b] pyridin-2-yl] -3-oxo-2- (prop-2-en-1-
yl) pyrazolo [3, 4-d] pyrimidin-6-yl} amino) phenyl] piperidin-4-one (170 mg, 0.323 mmol) and AcOH (1.94 mg, 0.032 mmol) at 25℃ for 1 h followed by the addition of NaBH3CN (50.81 mg, 0.807 mmol) in portions at 25℃. A solution of at 25℃ was stirred for 2 h . The mixture was purified by Prep-HPLC with the following conditions (Column: Xselect CSH F-phenyl OBD Column, 19*250 mm, 5μm; Mobile Phase A: Water (0.1%FA) , Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 10%B to 40%B in 10 min, 40%B; Wave Length: 254 nm; RT1 (min) : 8.89; Number Of Runs: 0) to afford the title compound.
LCMS: MS (ESI) m/z 943.12 [M+H] +.
1H NMR (300 MHz, DMSO-d6) : δ 10.98 (s, 1H) , 9.78 (s, 1H) , 8.87 (s, 1H) , 8.85 (s, 1H) , 7.97 (t, J = 7.9 Hz, 1H) , 7.71 (dt, J = 6.2, 3.1 Hz, 5H) , 7.59 (dd, J = 7.7, 0.9 Hz, 1H) , 7.53 (s, 1H) , 7.46 (dd, J = 7.8, 1.4 Hz, 1H) , 7.35 (d, J = 8.4 Hz, 1H) , 7.24 (dd, J = 8.4, 2.1 Hz, 1H) , 6.90 (d, J = 9.2 Hz, 3H) , 5.28 (s, 1H) , 5.05 -5.25 (m, 2H) , 4.39 -4.52 (m, 4H) , 3.55 (s, 2H) , 3.08 (d, J = 5.9 Hz, 4H) , 2.86 -2.92 (m, 1H) , 2.58 (d, J = 4.6 Hz, 5H) , 2.29 -2.48 (m, 1H) , 2.02 (s, 1H) , 1.52 (s, 6H) , 1.45 (d, J = 6.1 Hz, 6H) .
Example 8: 5- [4- [4- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] pyrazol-1-yl] -1-piperidyl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (Cpd-008)
Cpd-008 was synthesized according to the following scheme:
Step A: tert-butyl 4-methylsulfonyloxypiperidine-1-carboxylate
To a solution of tert-butyl 4-hydroxypiperidine-1-carboxylate (2 g, 9.94 mmol) in DCM (30 mL) was added TEA (3.02 g, 29.81 mmol) and methylsulfonyl methanesulfonate (2.60 g, 14.91 mmol) at 0 ℃. The mixture was stirred at 20 ℃ for 2 hrs. The reaction mixture was quenched by addition of water (50 mL) , and then extracted with DCM (60 mL × 3) . The combined organic layers was dried over Na2SO4, filtered and concentrated under reduced pressure to afford the title compound.
1H NMR (400 MHz, CDCl3) δ: 4.80–4.89 (m, 1H) , 3.61–3.75 (m, 2H) , 3.21–3.34 (m, 2H) , 3.071 (s, 3H) , 1.88–1.98 (m, 2H) , 1.72–1.84 (m, 2H) , 1.43 (s, 9H) .
Step B: tert-butyl 4- (4-formylpyrazol-1-yl) piperidine-1-carboxylate
To a solution of tert-butyl 4-methylsulfonyloxypiperidine-1-carboxylate (1.74 g) and 1H-pyrazole-4-carbaldehyde (500 mg, 5.20 mmol) in DMF (20 mL) was added Cs2CO3 (4.24 g, 13.01 mmol) . The mixture was stirred at 90 ℃ for 16 hrs. The reaction mixture was quenched by addition water (20 mL) , and then diluted with DCM (50 mL) and extracted with DCM (50 mL × 3) . The combined organic layers was washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~50%Ethyl acetate/Petroleum ether gradient @40 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 224.1 [M+H-tBu] +.
1H NMR (400 MHz, CDCl3) δ: 9.81 (s, 1H) , 7.95 (d, J = 6.0 Hz, 2H) , 4.13–4.34 (m, 3H) , 2.87 (t, J = 12.0 Hz, 2H) , 2.13 (d, J = 12.0 Hz, 2H) , 1.82–1.95 (m, 2H) , 1.44 (s, 9H) .
Step C: 1- (4-piperidyl) pyrazole-4-carbaldehyde
To a solution of tert-butyl 4- (4-formylpyrazol-1-yl) piperidine-1-carboxylate (300 mg, 1.07 mmol) in DCM (400 uL) was added TFA (616.00 mg, 5.40 mmol) . The mixture was stirred at 20 ℃ for 5 hrs. The reaction mixture was concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 180.2 [M+H] +.
Step D: 1- [1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] -4-piperidyl] pyrazole-4-
carbaldehyde
To a solution of 1- (4-piperidyl) pyrazole-4-carbaldehyde (200 mg) and 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (308.25 mg, 1.12 mmol) in DMSO (5 mL) was added TEA (225.84 mg, 2.23 mmol) . The mixture was stirred at 120 ℃ for 2 hrs under N2. The reaction mixture was quenched by addition water (5 mL) , and then diluted with DCM (10 mL) and extracted with DCM (20 mL × 3) . The combined organic layers was washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 0~80%Ethyl acetate/Petroleum ether gradient @30 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 436.1 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 11.08 (s, 1H) , 9.78 (s, 1H) , 8.55 (s, 1H) , 8.00 (s, 1H) , 7.69 (d, J = 8.4 Hz, 1H) , 7.41 (s, 1H) , 7.32 (d, J = 8.4 Hz, 1H) , 5.08 (dd, J = 12.8, 5.2 Hz, 1H) , 4.61 (t, J = 11.6 Hz, 1H) , 4.20 (d, J = 13.2 Hz, 2H) , 3.18 (t, J = 12.4 Hz, 2H) , 2.82-2.95 (m, 1H) , 2.53-2.67 (m, 3H) , 2.13 (d, J = 11.6 Hz, 3H) , 2.03 (d, J = 6.8 Hz, 1H) .
Step E: 5- [4- [4- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] pyrazol-1-
yl] -1-piperidyl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione
To solution of 1- [1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] -4-piperidyl] pyrazole-4-carbaldehyde (25.48 mg, 58.52 umol) and 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (10 mg, 19.51 umol) in DCM (1 mL) were added NaBH (OAc) 3 (12.40 mg, 58.52 umol) , molecular sieve (10 mg) and TEA (9.87 mg, 97.54 umol) . The mixture was stirred at 20 ℃ for 16 hrs. The residue was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (NH4HCO3) -ACN] ; B%: 36%-76%, 9 min) and then lyophilized to afford the title compound.
LCMS: MS (ESI) m/z = 932.5 [M+H] +.
1H NMR (400 MHz, CD3OD) δ: 8.78 (s, 1H) , 7.85 (d, J = 8.0 Hz, 1H) , 7.68–7.74 (m, 3H) , 7.50–7.56 (m, 3H) , 7.42 (d, J = 2.4 Hz, 1H) , 7.29 (dd, J = 8.4, 2.0 Hz, 1H) , 6.94 (d, J = 9.2 Hz, 2H) , 5.64–5.81 (m, 1H) , 5.05–5.12 (m, 2H) , 4.41–4.54 (m, 1H) , 4.19 (d, J = 14.0 Hz, 2H) , 3.56 (s, 2H) , 3.19 (s, 6H) , 3.00–3.11 (m, 1H) , 2.81–2.93 (m, 2H) , 2.75–2.78 (m, 1H) , 2.73 (s, 1H) , 2.6–2.69 (m, 4H) , 2.31–2.44 (m, 1H) , 2.08–2.25 (m, 7H) , 1.99–2.05 (m, 1H) , 1.79–1.91 (m, 1H) , 1.25–1.38 (m, 4H) , 0.86–0.97 (m, 4H) .
Example 9: 3- [5- [2- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -7-azaspiro [3.5] nonan-7-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-009)
Cpd-009 was synthesized according to the following scheme:
Step A: 2- (2, 6-dioxo-3-piperidyl) -5- [2- (hydroxymethyl) -7-azaspiro [3.5] nonan-7-
yl] isoindoline-1, 3-dione
A mixture of 7-azaspiro [3.5] nonan-2-ylmethanol (200 mg, 1.04 mmol, HCl salt) , 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (430.90 mg, 1.56 mmol) and DIEA (806.5mg, 6.24 mmol) in DMSO (3 mL) was degassed and purged with N2 for 3 times, and stirred at 120 ℃ for 16 hours. The reaction was diluted with water (20 mL) and extracted with ethyl acetate (40 mL × 2) . The combined organics were washed with brine (20 mL × 2) , dried over anhydrous sodium sulfate, filtered and concentrated in vacuum to give the residue, which was purified by flash
column chromatography on silica gel (eluted with methanol in dichloromethane from 3%to 8%) to afford the title compound.
LCMS: MS (ESI) m/z = 412.1 [M+H] +.
Step B: [7- [2- (2, 6-dioxo-3-piperidyl) -3-hydroxy-1-oxo-isoindolin-5-yl] -7-
azaspiro [3.5] nonan-2-yl] methyl acetate and [7- [2- (2, 6-dioxo-3-piperidyl) -1-hydroxy-3-oxo-
isoindolin-5-yl] -7-azaspiro [3.5] nonan-2-yl] methyl acetate (a mixture)
A mixture of 2- (2, 6-dioxo-3-piperidyl) -5- [2- (hydroxymethyl) -7-azaspiro [3.5] nonan-7-yl] isoindoline-1, 3-dione (200 mg, 486.09 umol) was dissolved in CH3COOH (10 mL) , to which was added Zn (127.1 mg, 1.94 mmol) , and the mixture was stirred at 80 ℃ for 16 hours. The mixture was filtered through diatomite pad under vacuum and washed with ethyl acetate (30 mL × 3) . The filtrate was evaporated to afford a residue. The residue (150 mg) was dissolved in CH3COOH (3 mL) , to which was added Zn (216.3 mg, 3.31 mmol) , and the mixture was allowed to stir at 80 ℃ for another 2 hours. The mixture was filtered through diatomite pad under vacuum and washed with ethyl acetate (30 mL × 3) . The filtrate was evaporated to afford a mixture of [7- [2- (2, 6-dioxo-3-piperidyl) -3-hydroxy-1-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonan-2-yl] methyl acetate and [7- [2- (2, 6-dioxo-3-piperidyl) -1-hydroxy-3-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonan-2-yl] methyl acetate.
LCMS: MS (ESI) m/z = 456.2 [M+H] +.
Step C: [7- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonan-2-
yl] methyl acetate and [7- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -7-
azaspiro [3.5] nonan-2-yl] methyl acetate (a mixture)
A mixture of [7- [2- (2, 6-dioxo-3-piperidyl) -3-hydroxy-1-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonan-2-yl] methyl acetate and [7- [2- (2, 6-dioxo-3-piperidyl) -1-hydroxy-3-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonan-2-yl] methyl acetate (150 mg, mixture of two region-isomers) was dissolved in dichloromethane (3 mL) , to which were added triethylsilane (728 mg, 6.26 mmol) , and TFA (3.1 g, 27.01 mmol) . The mixture was stirred at 25℃ for 2 hours. The reaction mixture was concentrated under reduced pressure to afford [7- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonan-2-yl] methyl acetate and [7- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonan-2-yl] methyl acetate (mixture of two region-isomers, crude) .
LCMS: tR = 0.582 & 0.633 min, m/z = 440.1 [M+H] +.
Step D: 3- [5- [2- (hydroxymethyl) -7-azaspiro [3.5] nonan-7-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione and 3- [6- [2- (hydroxymethyl) -7-azaspiro [3.5] nonan-7-yl] -1-oxo-
isoindolin-2-yl] piperidine-2, 6-dione
A mixture of [7- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonan-2-yl] methyl acetate and [7- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonan-2-yl] methyl acetate (150 mg, mixture of two region-isomers) was dissolved in aq HCl (6 M, 2 mL) , and the mixture was allowed to react at 50 ℃ for one hour. The mixture was purified by reversed-phase (C18, 10%-20%CH3CN in water 0.1%HCl condition) to afford 3- [6- [2- (hydroxymethyl) -7-azaspiro [3.5] nonan-7-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione and 3- [5- [2- (hydroxymethyl) -7-azaspiro [3.5] nonan-7-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione.
Step E: 7- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonane-2-
carbaldehyde
A mixture of 3- [5- [2- (hydroxymethyl) -7-azaspiro [3.5] nonan-7-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (8 mg) was dissolved in dichloromethane (1.5 mL) and DMF (1.5 mL) , to which was added DMP (12.8 mg, 30.19 umol) , and the mixture was stirred at 25 ℃ for 2 hours. To the reaction mixture was added water (10 mL) , the pH value of the mixture was adjusted to 8 by addition of saturated sodium bicarbonate aqueous solution and extracted with ethyl acetate (20 mL × 2) . The organics were washed with water (20 mL) and brine (20 mL) , dried over anhydrous sodium sulfate, filtered and concentrated in vacuum to afford the title compound.
LCMS: MS (ESI) m/z = 396.1 [M+H] +.
Step F: 3- [5- [2- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -7-
azaspiro [3.5] nonan-7-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 7- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonane-2-carbaldehyde (6 mg) and 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (5 mg, 9.75 umol) in DMF (2 mL) was added TEA (9.87 mg, 97.54 umol) , and the mixture was stirred at 25 ℃ for 1 hour, then NaBH (OAc) 3 (6.2 mg, 29.26 umol) was added, and the mixture was stirred at 25℃ for 12 hours. The mixture was filtered through filter and the filter cake was washed with DMF (0.5 mL) . The filtrate was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 48%-78%, 8min) and lyophilized to afford the title compound.
LCMS: MS (ESI) m/z = 892.6 [M+H] +.
1H NMR (400 MHz, MeOD) δ: 8.79 (s, 1H) , 7.86 (d, J = 8.0 Hz, 1H) , 7.72 (d, J = 8.0 Hz, 1H) , 7.61 (d, J = 8.4 Hz, 1H) , 7.55 (d, J = 8.4 Hz, 2H) , 7.10-7.05 (m, 2H) , 6.96 (d, J = 9.2 Hz, 2H) , 5.79-5.64 (m, 1H) , 5.40-5.30 (m, 1H) , 5.11 (d, J = 5.2 Hz, 1H) , 5.08 (d, J = 5.2 Hz, 1H) , 5.05-
5.02 (m, 2H) , 4.39 (d, J = 6.4 Hz, 2H) , 3.38-3.35 (m, 2H) , 3.29-3.25 (m, 2H) , 3.22-3.3.18 (m, 4H) , 3.11-3.02 (m, 1H) , 2.92-2.83 (m, 2H) , 2.74-2.69 (m, 4H) , 2.64-2.61 (m, 2H) , 2.50-2.33 (m, 2H) , 2.21-2.10 (m, 5H) , 2.06-1.99 (m, 2H) , 1.83-1.79 (m, 2H) , 1.69-1.64 (m, 2H) , 1.63-1.55 (m, 3H) , 0.95 (t, J = 7.4 Hz, 3H) .
Example 10: 3- [6- [2- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -7-azaspiro [3.5] nonan-7-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-010)
Cpd-010 was synthesized according to the following scheme:
Step A: 7- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonane-2-
carbaldehyde
3- [6- [2- (hydroxymethyl) -7-azaspiro [3.5] nonan-7-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (15 mg, 37.74 umol) was dissolved in a mixture of DMF (1.5 mL) and dichloromethane (1.5 mL) , to which was added DMP (30 mg, 70.73 umol) , and the mixture was stirred at 25℃ for 2 hours. To the reaction mixture was added water (10 mL) , the pH value of mixture was adjusted to 8 by addition of saturated sodium bicarbonate aqueous solution, and extracted with ethyl acetate (20 mL × 2) . The organics were washed with water (20 mL) and brine (20 mL) , dried over anhydrous sodium sulfate, filtered and concentrated in vacuum to afford the title compound.
LCMS: MS (ESI) m/z = 396.1 [M+H] +.
Step B: 3- [6- [2- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -7-
azaspiro [3.5] nonan-7-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 7- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonane-2-carbaldehyde (15 mg) and 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (15 mg, 29.26 umol) in DMF (2 mL) was added TEA (29.6 mg, 292.62 umol) , and the mixture was stirred at 25℃ for 1 hour, then NaBH (OAc) 3 (18.6 mg, 87.79 umol) was added, and the mixture was stirred at 25 ℃ for 12 hours. The mixture was filtered through filter and washed with DMF (0.5 mL) . The mixture purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 46%-76%, 8min) : 60 mL/min) and lyophilized to afford the title compound.
LCMS: MS (ESI) m/z = 892.6 [M+H] +.
1H NMR 400 MHz, DMSO-d6) δ = 10.96 (s, 1H) , 10.20-9.98 (m, 1H) , 8.81 (s, 1H) , 7.92 (d, J =6.0 Hz, 1H) , 7.69 (d, J = 8.0 Hz, 1H) , 7.62-7.51 (m, 2H) , 7.40 (d, J = 8.4 Hz, 1H) , 7.24 (dd, J =2.0, 8.4 Hz, 1H) , 7.14 (d, J = 2.0 Hz, 1H) , 6.90 (d, J = 9.2 Hz, 2H) , 5.73-5.58 (m, 1H) , 5.13-4.96 (m, 3H) , 4.85 (d, J = 17.6 Hz, 1H) , 4.80-4.67 (m, 1H) , 4.62-4.50 (m, 1H) , 4.38-4.14 (m, 2H) , 3.30-3.29 (m, 4H) , 3.22-3.16 (m, 2H) , 3.12-3.05 (m, 6H) , 2.99-2.85 (m, 2H) , 2.82-2.70 (m, 1H) , 2.69-2.59 (m, 1H) , 2.44-2.40 (m, 2H) , 2.36-2.29 (m, 1H) , 2.24-2.13 (m, 1H) , 2.06-1.92 (m, 5H) , 1.91-1.83 (m, 1H) , 1.76-1.65 (m, 3H) , 1.61-1.55 (m, 2H) , 1.51-1.40 (m, 2H) , 0.86 (t, J = 7.2 Hz, 3H)
Example 11: 3- [5- [7- [ [1- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -4-piperidyl] methyl] -2, 7-diazaspiro [3.5] nonan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-011)
Cpd-011 was synthesized according to the following scheme:
Step A: tert-butyl 2- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -2, 7-
diazaspiro [3.5] nonane-7-carboxylate
To a mixture of 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (300 mg, 928.39 μmol) and tert-butyl 2, 7-diazaspiro [3.5] nonane-7-carboxylate (210 mg, 927.91 μmol) in dioxane (10 mL) were added Cs2CO3 (907.2 mg, 2.78 mmol) , RuPhos (86.4 mg, 185.15 μmol) and RuPhos Pd G3 (77.4 mg, 92.54 μmol) . Then the mixture was stirred at 120 ℃ for 12 h under N2. The reaction mixture was added with water (50 mL) , extracted with EtOAc (3 × 30 mL) . The combined organic layer was washed with brine (80 mL) , dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~3%Methanol/Dichloromethane gradient @40 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 360.9 [M-100+H] +.
Step B: 3- [5- (2, 7-diazaspiro [3.5] nonan-2-yl) -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
A mixture of tert-butyl 2- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -2, 7-diazaspiro [3.5] nonane-7-carboxylate (162 mg, 345.75 μmol) in TFA (1 mL) and DCM (4 mL) was stirred at 20 ℃ for 1h. The reaction mixture was concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 369.0 [M+H] +.
Step C: 3- [5- [7- [ [1- (4-nitrophenyl) -4-piperidyl] methyl] -2, 7-diazaspiro [3.5] nonan-2-yl] -1-
oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a mixture of 3- [5- (2, 7-diazaspiro [3.5] nonan-2-yl) -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (160 mg) , 1- (4-nitrophenyl) piperidine-4-carbaldehyde (77.7 mg, 331.70 μmol) in DCM (3 mL) was added TEA (167.7 mg, 1.66 mmol) . The mixture was stirred at 20 ℃ for 12 hours under N2. Then NaBH (OAc) 3 (351.4 mg, 1.66 mmol) was added into the mixture reaction and the mixture was stirred at 20 ℃ for another 4 hour under N2. The reaction mixture was concentrated under reduced pressure to give a residue, which was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~7%Methanol/Dichloromethane gradient @30 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 587.4 [M+H] +.
Step D: 3- [5- [7- [ [1- (4-aminophenyl) -4-piperidyl] methyl] -2, 7-diazaspiro [3.5] nonan-2-yl] -1-
oxo-isoindolin-2-yl] piperidine-2, 6-dione
A mixture of 3- [5- [7- [ [1- (4-nitrophenyl) -4-piperidyl] methyl] -2, 7-diazaspiro [3.5] nonan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (190 mg, 323.86 μmol) and Pd/C (100 mg, 10%w/w) in EtOAc (10 mL) was stirred at 20 ℃ for 2 h under H2 (15psi) . The reaction mixture was filtered and the solid was washed with MeOH (50 mL) . The filtrate was concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 557.4 [M+H] +.
1H NMR (ES22601-110-P1B, 400MHz, METHANOL-d4) δ: 7.30 (d, J = 8.4 Hz, 1H) , 6.88 (d, J = 8.0 Hz, 2H) , 6.77-6.66 (m, 4H) , 5.28-5.21 (m, 1H) , 5.05-4.99 (m, 1H) , 4.68-4.48 (m, 1H) , 4.28 (d, J = 8.8 Hz, 2H) , 3.66 (s, 4H) , 3.31-3.26 (m, 4H) , 2.81-2.70 (m, 4H) , 2.24-2.00 (m, 6H) , 1.57-1.40 (m, 9H) .
Step E: 3- [5- [7- [ [1- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -4-piperidyl] methyl] -2, 7-
diazaspiro [3.5] nonan-2-yl] -1-oxo-isoin-dolin-2-yl] piperidine-2, 6-dione
To a mixture of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6-methylsulfanyl-pyrazolo [3, 4-d] pyrimidin-3-one (27.55 mg, 71.85 μmol) in toluene (2 mL) was added m-CPBA (17.51 mg, 86.22 μmol, 85%purity) at 0 ℃, and the mixture was stirred at 20 ℃ for 1 h. Then 3- [5- [7- [ [1- (4-aminophenyl) -4-piperidyl] methyl] -2, 7-diazaspiro [3.5] nonan-2-yl] -1-oxo-isoindolin-2-yl] -piperidine-2, 6-dione (40 mg) was added and the mixture was stirred at 20 ℃ for 12 hrs. The reaction mixture was concentrated under reduced pressure to give a residue, which was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3·H2O+NH4HCO3) -ACN] ; B%: 50%-80%, 8min) to afford the title compound. LCMS: MS (ESI) m/z = 892.8 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ: 10.96 (s, 1H) , 10.11 (s, 1H) , 8.81 (s, 1H) , 7.92 (s, 1H) , 7.69 (d, J = 8.0 Hz, 1H) , 7.56 (s, 2H) , 7.37 (d, J = 7.6 Hz, 1H) , 7.20 (s, 1H) , 6.91 (d, J = 9.4 Hz, 2H) ,
6.68-6.71 (m, 2H) , 5.69 (d, J = 10.0 Hz, 1H) , 5.32 (s, 1H) , 5.04-5.11 (m, 3H) , 5.00 (d, J = 10.0 Hz, 1H) , 4.85 (d, J = 18.0 Hz, 1H) , 4.73 (s, 1H) , 4.56 (d, J = 14.4 Hz, 1H) , 4.27-4.35 (m, 2H) , 4.18 (d, J = 16.4 Hz, 2H) , 3.61-3.65 (m, 3H) , 2.95 (d, J = 13.2 Hz, 2H) , 2.79 (d, J = 10.0 Hz, 2H) , 2.67 (s, 2H) , 2.57 (s, 2H) , 2.35-2.39 (m, 2H) , 2.15-2.22 (m, 4H) , 2.00 (d, J = 6.8 Hz, 3H) , 1.90 (s, 1H) , 1.71-1.79 (m, 8H) , 1.45 (s, 1H) , 0.86-0.90 (m, 3H) .
Example 12: 3- [5- [8- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -2-azaspiro [4.5] decan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-012)
Cpd-012 was synthesized according to the following scheme:
Step A: 2-azaspiro [4.5] decan-8-ylmethanol
To a solution of tert-butyl 8- (hydroxymethyl) -2-azaspiro [4.5] decane-2-carboxylate (284.2 mg, 1.05 mmol) in DCM (5 mL) was added HCl/dioxane (4 M, 5 mL) at 0℃, The mixture was stirred at 25 ℃ for 2 hours. The reaction mixture was concentrated in vacuum to give a residue. The residue was suspended in toluene (10 mL) . The mixture was concentrated in vacuum to afford the title compound.
Step B: 3- [5- [8- (hydroxymethyl) -2-azaspiro [4.5] decan-2-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
A mixture of 2-azaspiro [4.5] decan-8-ylmethanol (95.5 mg, HCl salt) , 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (100 mg, 309.46 umol) , Pd-PEPPSI-IHeptCl (15.1 mg,
15.47 umol) , and Cs2CO3 (151.2 mg, 464.19 umol) in dioxane (1.5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 16 hours under N2 atmosphere. The reaction was diluted with water (20 mL) and extracted with ethyl acetate (40 mL × 2) . The combined organic layers was washed with brine (20 mL × 2 ) , dried over anhydrous sodium sulfate, filtered and concentrated in vacuum to give the residue, which was purified by prep-TLC (dichloromethane: methanol = 20: 1) to afford the title compound.
LCMS: MS (ESI) m/z = 412.1 [M+1] +.
Step C: 2- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -2-azaspiro [4.5] decane-8-
carbaldehyde
A mixture of 3- [5- [8- (hydroxymethyl) -2-azaspiro [4.5] decan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (10 mg, 24.30 umol) was dissolved in dichloromethane (1.5 mL) and DMF (1.5 mL) , to which was added DMP (20.6 mg, 48.60 umol) , and the mixture was allowed to react at 25 ℃ for 1.5 h. To the reaction mixture was added DMP (10 mg) and stirred at 25℃for another 0.5 hour. To the reaction mixture was added water (10 mL) , the pH value of the mixture was adjusted to 8 by addition of saturated sodium bicarbonate aqueous solution, and extracted with ethyl acetate (20 mL × 2) . The organic layer was separated, washed with water (20 mL) and brine (20 mL) , dried over anhydrous sodium sulfate, filtered and concentrated in vacuum to afford the title compound.
LCMS: MS (ESI) m/z = 410.1 [M+1] +.
Step D: 3- [5- [8- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -2-
azaspiro [4.5] decan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -2-azaspiro [4.5] decane-8-carbaldehyde (5 mg) and 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (6.9 mg, 13.43 umol) in DMF (2 mL) was added TEA (12.4 mg, 122.11 umol) , and the mixture was stirred at 25 ℃ for one hour, then NaBH (OAc) 3 (7.8 mg, 36.63 umol) was added, and the mixture was stirred at 25 ℃ for 12 hours. The mixture was filtered through filter and washed with DMF (0.5 mL) . The mixture was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 54%-84%, 8min) and lyophilized to afford the title compound.
LCMS: MS (ESI) m/z = 906.7 [M+H] +.
1H NMR (400 MHz, MeOD) δ: 8.79 (s, 1H) , 7.86 (d, J = 8.0 Hz, 1H) , 7.72 (d, J = 8.0 Hz, 1H) , 7.54 (d, J = 9.2 Hz, 2H) , 7.35 (d, J = 8.0 Hz, 1H) , 6.96 (d, J = 9.2 Hz, 2H) , 6.88 (d, J = 2.4 Hz, 1H) , 6.83 (dd, J = 2.4, 8.4 Hz, 1H) , 5.77-5.65 (m, 1H) , 5.34 (t, J = 4.4 Hz, 1H) , 5.19 (s, 1H) ,
5.12 (dd, J = 5.2, 13.2 Hz, 1H) , 5.06-4.97 (m, 2H) , 4.36 (d, J = 7.2 Hz, 2H) , 3.42-3.38 (m, 2H) , 3.28-3.26 (m, 2H) , 3.21-3.17 (m, 4H) , 3.13-3.11 (m, 2H) , 2.92-2.89 (m, 1H) , 2.70-2.63 (m, 4H) , 2.36-2.32 (m, 2H) , 2.21-2.17 (m, 4H) , 2.15-2.12 (m, 1H) , 2.05-2.01 (m, 4H) , 1.99-1.96 (m, 2H) , 1.83-1.78 (m, 2H) , 1.74-1.69 (m, 2H) , 1.63-1.58 (m, 3H) , 0.94 (t, J = 7.2 Hz, 3H)
Example 13: 3- (5- (4- ( (1- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperidin-4-yl) methyl) piperazin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (Cpd-013)
Cpd-013 was synthesized according to the following scheme:
Step A: tert-butyl 4- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] piperazine-1-
carboxylate
A Schlenk tube charged with 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (1 g, 3.09 mmol) , tert-butyl piperazine-1-carboxylate (1.15 g, 6.19 mmol) , Cs2CO3 (3.02 g, 9.28 mmol) , [2- (2-aminophenyl) phenyl] -methylsulfonyloxy-palladium; dicyclohexyl- [2- (2, 6-diisopropoxyphenyl) phenyl] phosphane (258.82 mg, 309.46 umol) and dicyclohexyl- [2- (2, 6-diisopropoxyphenyl) phenyl] phosphane (288.81 mg, 618.92 umol) was degassed and purged with N2 for 3 times, and then dioxane (10 mL) was added. The mixture was stirred at 120 ℃ for 4 hours. The residue was diluted with H2O (200 mL) and extracted with ethyl acetate (100 mL ×3) . The combined organic layers was washed with brine (100 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give crude product, which was purified by flash column chromatography on silica gel (ethyl acetate in dichloromethane from 56%to 60%) to afford the title compound.
LCMS: MS (ESI) m/z = 373.1 [M+H] +.
Step B: 3- (1-oxo-5-piperazin-1-yl-isoindolin-2-yl) piperidine-2, 6-dione
To a solution of tert-butyl 4- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] piperazine-1-carboxylate (260 mg, 606.80 umol) in CH2Cl2 (3 mL) was added HCl/dioxane (4 M, 3 mL) . The mixture was stirred at 25 ℃ for 3 hours. The reaction mixture concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 329.1 [M+H] +.
Step C: 3- [5- [4- [ [1- (4-nitrophenyl) -4-piperidyl] methyl] piperazin-1-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
To a solution of 3- (1-oxo-5-piperazin-1-yl-isoindolin-2-yl) piperidine-2, 6-dione (250.00 mg) in CH2Cl2 (4 mL) was added TEA (173.35 mg, 1.71 mmol) and 1- (4-nitrophenyl) piperidine-4-carbaldehyde (192.63 mg, 822.31 umol) . The mixture was stirred at 25 ℃ for 2 hours. And then NaBH (OAc) 3 (435.70 mg, 2.06 mmol) was added. The mixture was stirred at 25 ℃ for 16 hours. The residue was diluted with H2O (20 mL) and extracted with CH2Cl2 (30 mL × 3) . The combined organic layers was washed with brine (30 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash column chromatography on silica gel (MeOH in CH2Cl2 from 0%to 10 %) to afford the title compound. LCMS: MS (ESI) m/z = 547.2 [M+H] +.
Step D: 3- [5- [4- [ [1- (4-aminophenyl) -4-piperidyl] methyl] piperazin-1-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
To a solution of 3- [5- [4- [ [1- (4-nitrophenyl) -4-piperidyl] methyl] piperazin-1-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (260 mg, 475.65 umol) in THF (4 mL) was added Pd/C (0.26 g, 10%w/w) and the mixture was degassed, purged with N2 for 3 times. The mixture was stirred at 25 ℃ for 12 hours under hydrogen atmosphere (15 Psi) . The reaction mixture was
filtered through celite, and the filter cake was washed with DMF (30mL) for 3 times. The filtrate was concentrated in vacuum to afford the title compound.
LCMS: MS (ESI) m/z = 517.2 [M+H] +.
1H NMR (400 MHz, DMSO) δ: 10.91 (br s, 1H) , 7.43 (d, J = 8.4 Hz, 1H) , 7.25 (dd, J = 8.4, 2.0 Hz, 1H) , 7.16 (d, J = 2.0 Hz, 1H) , 6.68 (d, J = 8.8 Hz, 2H) , 6.48 (d, J = 8.4 Hz, 2H) , 5.09 (dd, J = 13.2, 5.2 Hz, 1H) , 4.53 (br s, 2H) , 4.33 (d, J = 16.4 Hz, 1H) , 4.21 (d, J = 16.4 Hz, 1H) , 3.22-3.15 (m, 5H) , 2.63-2.59 (m, 1H) , 2.58-2.54 (m, 1H) , 2.47-2.42 (m, 2H) , 2.41-2.34 (m, 1H) , 2.21 (d, J = 7.2 Hz, 1H) , 2.05-1.94 (m, 2H) , 1.78 (d, J = 11.6 Hz, 2H) , 1.67-1.55 (m, 1H) , 1.30-1.15 (m,5H) .
Step E: 3- (5- (4- ( (1- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperidin-4-yl) methyl) piperazin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-
dione
To a solution of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6-methylsulfanyl-pyrazolo [3, 4-d] pyrimidin-3-one (133.60 mg, 348.41 umol) in toluene (3 mL) was added m-CPBA (64.84 mg, 319.37 umol) at 0 ℃ and the mixture was stirred for 1 hour. Then 3- [5- [4- [ [1- (4-aminophenyl) -4-piperidyl] methyl] piperazin-1-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (150 mg) and DIEA (112.57 mg, 871.02 umol, 151.72 uL) were added. The mixture was stirred at 25 ℃ for 16 hours. The residue was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 47%-77%, 8min) to afford the title compound.
LCMS: MS (ESI) m/z = 852.6 [M+1] +.
1H NMR (400 MHz, DMSO) δ: 10.97 (s, 1H) , 10.09 (br s, 1H) , 8.81 (s, 1H) , 7.92 (d, J = 7.2 Hz, 1H) , 7.69 (d, J = 8.4 Hz, 2H) , 7.62-7.52 (m, 2H) , 7.43 (d, J = 8.8 Hz, 1H) , 7.36 (dd, J = 8.8, 4.2Hz, 1H) , 7.17 (d, J = 2.0 Hz, 1H) , 6.92 (d, J = 8.8 Hz, 1H) , 5.73-5.61 (m, 1H) , 5.12-5.06 (m, 1H) , 4.99 (d, J = 10.4 Hz, 1H) , 4.85 (d, J = 17.2 Hz, 1H) , 4.80-4.69 (m, 1H) , 4.62-4.51 (m, 1H) , 4.33 (d, J = 8.8Hz, 2H) , 4.33 (d, J = 16.8Hz, 1H) , 4.21 (d, J = 16.8Hz, 1H) , 3.64 (br d, J = 12.4 Hz, 2H) , 3.23-3.15 (m, 4H) , 3.03-2.85 (m, 2H) , 2.83-2.73 (m, 1H) , 2.68-2.60 (m, 3H) , 2.56-2.51 (m, 4H) , 2.42-2.34 (m, 1H) , 2.27-2.14 (m, 3H) , 2.06-1.95 (m, 2H) , 1.93-1.85 (m, 1H) , 1.85-1.76 (m, 2H) , 1.75-1.63 (m, 2H) , 1.27-1.22 (m, 2H) , 0.87 (t, J = 7.2 Hz, 3H) .
Example 14: 3- [5- [3- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-014)
Cpd-014 was synthesized according to the following scheme:
Step A: 8-azaspiro [4.5] decan-3-ylmethanol
To a solution of tert-butyl 3- (hydroxymethyl) -8-azaspiro [4.5] decane-8-carboxylate (300 mg, 1.11 mmol) in CH2Cl2 (3 mL) was added HCl/dioxane (4 M, 9.59 mL) . The mixture was stirred at 25 ℃ for 5 hours. The mixture was concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 170.1 [M+H] +.
Step B: 3- [5- [3- (hydroxymethyl) -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
Schlenk tube charged of 8-azaspiro [4.5] decan-3-ylmethanol (100 mg) , 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (196.34 mg, 607.61 umol) , Pd-PEPPSI-IHeptCl (24.11 mg, 30.38 umol) , and Cs2CO3 (296.96 mg, 911.41 umol) in dioxane (3 mL) was degassed and purged with N2 for 3 times. The reaction mixture was stirred at 100 ℃ for 12 hours. The resulting mixture was treated with water (100 mL) and extracted with ethyl acetate (100 mL ×3) . The combined organic phase was washed with brine (50 mL) and dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduce pressure to give crude product, which was purified by flash column chromatography on silica gel (MeOH in CH2Cl2 from 0%to 3 %) to afford the title compound.
LCMS: MS (ESI) m/z = 412.2 [M+H] +.
Step C: 8- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -8-azaspiro [4.5] decane-3-
carbaldehyde
To a solution of 3- [5- [3- (hydroxymethyl) -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (50 mg, 121.51 umol) in CH2Cl2 (2 mL) was added DMP (77.31 mg, 182.26 umol) at 0 ℃. Then the mixture was warmed to room temperature slowly and stirred for 12 hours. The reaction was diluted with water (20 mL) and extracted with ethyl acetate (40 mL × 2) . The organics were washed with brine (20 mL × 2) , dried over anhydrous sodium sulfate, filtered and concentrated in vacuum to afford the title compound.
LCMS: MS (ESI) m/z = 410.1 [M+H] +.
Step D: 3- [5- [3- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -8-
azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 8- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -8-azaspiro [4.5] decane-3-carbaldehyde (20 mg) in CH2Cl2 (4 mL) was added TEA (12.36 mg, 122.11 umol) , and 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (25.04 mg, 48.84 umol) . The mixture was stirred at 25 ℃ for 2 hours, and then NaBH (OAc) 3 (31.06 mg, 146.53 umol) was added. The mixture was stirred at 25 ℃ for 14 hours. The residue concentrated under reduced pressure to give a residue, which was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 53%-83%, 8min) to afford the title compound.
LCMS: MS (ESI) m/z = 906.9 [M+H] +.
1H NMR (400 MHz, CDCl3) δ: 8.81 (s, 1H) , 8.44 (br s, 1H) , 7.69 (q, J = 8.4 Hz, 2H) , 7.47 (d, J = 8.8 Hz, 2H) , 7.39 (d, J = 2.0 Hz, 1H) , 7.32 (d, J = 8.0 Hz, 1H) , 7.18 (dd, J = 8.4, 2.4Hz, 1H) , 6.9 (d, J = 9.2 Hz, 2H) , 5.75-5.63 (m, 1H) , 5.22 (dd, J = 13.2 Hz, 4.2Hz, 1H) , 5.02 (d, J = 10.0 Hz, 1H) , 4.91 (d, J = 17.2 Hz, 1H) , 4.86-4.77 (m, 1H) , 4.72-4.53 (m, 1H) , 4.40 (d, J = 15.6 Hz, 2H) , 4.27 (d, J = 15.6 Hz, 2H) , 3.31-3.17 (m, 8H) , 3.11-3.00 (m, 1H) , 2.93-2.82 (m, 3H) , 2.78-2.67 (m, 4H) , 2.53-2.46 (m, 2H) , 2.43-2.30 (m, 4H) , 2.27-2.16 (m, 3H) , 2.07-1.98 (m, 2H) , 1.86-1.83 (m, 2H) , 1.66-1.64 (m, 2H) , 1.58-1.54 (m, 2H) , 1.43-1.36 (m, 1H) , 1.20-1.13 (m, 1H) , 0.99 (t, J = 7.2 Hz, 3H) .
Example 15: 3- [5- [7- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -2-azaspiro [3.5] nonan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-015)
Cpd-015 was synthesized according to the following scheme:
Step A: O2-tert-butyl O7-isopropoxycarbonyl 2-azaspiro [3.5] nonane-2, 7-dicarboxylate
To a solution of 2-tert-butoxycarbonyl-2-azaspiro [3.5] nonane-7-carboxylic acid (200.0 mg, 742.57 umol) in THF (5 mL) was added Et3N (300.5 mg, 2.97 mmol) and isopropyl carbonochloridate (136.5 mg, 1.11 mmol) at 0 ℃. The mixture was stirred at 0 ℃ for 2 hours. To the reaction mixture was added isopropyl carbonochloridate (50.0 mg) and stirred at 0 ℃ for 2 hours. The mixture was filtered and the filter cake was washed with THF (30 mL) . The filtrate was concentrated in vacuum to afford the title compound.
LCMS: MS (ESI) m/z = 378.1 [M+Na] +.
Step B: tert-butyl 7- (hydroxymethyl) -2-azaspiro [3.5] nonane-2-carboxylate
To a solution of O2-tert-butyl O7-isopropoxycarbonyl 2-azaspiro [3.5] nonane-2, 7-dicarboxylate (200.0 mg) in THF (4 mL) and H2O (0.2 mL) was added LiBH4 (73.6 mg, 3.38 mmol) . The mixture was stirred at 0 ℃ for 2 hours. The reaction was diluted with water (20 mL) and
extracted with ethyl acetate (40 mL × 2) . The combined organic layers was washed with brine (20 mL × 2) , dried over anhydrous sodium sulfate, filtered and concentrated in vacuum to give the residue, which was purified by flash column chromatography on silica gel (eluted with methanol in dichloromethane from 0%to 10%) to afford the title compound.
LCMS: MS (ESI) m/z = 200.1 [M-56+1] +.
Step C: 2-azaspiro [3.5] nonan-7-ylmethanol
To a solution of tert-butyl 7- (hydroxymethyl) -2-azaspiro [3.5] nonane-2-carboxylate (180.0 mg, 704.91 umol) in dichloromethane (5 mL) was added HCl/dioxane (4 M, 5 mL) . The mixture was stirred at 25 ℃ for 3 hours. The reaction mixture was concentrated in vacuum to give the title compound.
Step D: 3- [5- [7- (hydroxymethyl) -2-azaspiro [3.5] nonan-2-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
Schlenk tube charged with a mixture of 2-azaspiro [3.5] nonan-7-ylmethanol (70.0 mg, HCl salt) , 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (177.0 mg, 547.74 umol) , Pd-PEPPSI-IHeptCl (28 mg, 28.78 umol) and Cs2CO3 (202.26 mg, 620.77 umol) in dioxane (5 mL) was degassed and purged with N2 for 3 times, then the reaction mixture was stirred at 100 ℃ for 16 hours. The residue was purified by flash column chromatography on silica gel (eluted with methanol in dichloromethane from 0%to 5%) to afford the title compound.
LCMS: MS (ESI) m/z = 463.2 [M+H] +.
Step E: 2- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -2-azaspiro [3.5] nonane-7-
carbaldehyde
To a mixture of 3- [5- [7- (hydroxymethyl) -2-azaspiro [3.5] nonan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (13.0 mg, 32.71 umol) in CH2Cl2 (1 mL) and DMF (1 mL) was added DMP (20.8 mg, 49.06 umol) , then the reaction mixture was stirred at 25 ℃ for 2 hours. To the reaction mixture was added water (10 mL) , the pH value of the mixture was adjusted to 8 by addition of saturated aqueous sodium bicarbonate and extracted with ethyl acetate (20 mL × 2) . The combined organic layers was washed with water (20 mL) and brine (20 mL) , dried over anhydrous sodium sulfate, filtered and concentrated in vacuum to afford the title compound.
LCMS: MS (ESI) m/z = 396.1 [M+H] +.
Step F: 3- [5- [7- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -2-
azaspiro [3.5] nonan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a mixture of 2- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -2-azaspiro [3.5] nonane-7-carbaldehyde (10.0 mg) and 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (13.0 mg, 25.29 umol) in DMF
(2 mL) was added TEA (25.59 mg, 252.88 umol) and the mixture was stirred at 25 ℃ for 1 hour. Then NaBH (OAc) 3 (16.1 mg, 75.86 umol) was added, and the mixture was stirred at 25 ℃for 12 hours. The reaction mixture was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 52%-82%, 8min) : 60 mL/min) and lyophilized to afford the title compound.
LCMS: MS (ESI) m/z = 892.6 [M+1] +.
1H NMR (400 MHz, DMSO-d6) δ: 10.97 (s, 1H) , 8.83 (s, 1H) , 7.97-7.91 (m, 1H) , 7.70 (d, J =8.4 Hz, 1H) , 7.66-7.55 (m, 2H) , 7.37 (d, J = 7.6 Hz, 1H) , 7.00-6.88 (m, 2H) , 6.72-6.62 (m, 2H) , 5.73-5.60 (m, 1H) , 5.11-5.04 (m, 2H) , 5.02-4.96 (m, 1H) , 4.91-4.81 (m, 1H) , 4.80-4.71 (m, 1H) , 4.36-4.14 (m, 2H) , 3.59-3.52 (m, 4H) , 3.31-3.30 (m, 6H) , 3.14-3.05 (m, 3H) , 3.02-2.74 (m, 2H) , 2.43-2.31 (m, 2H) , 2.25-2.11 (m, 3H) , 2.06-1.94 (m, 4H) , 1.93-1.85 (m, 3H) , 1.80-1.66 (m, 4H) , 1.56-1.44 (m, 3H) , 0.88 (t, J = 7.2 Hz, 3H) .
Example 16: 3- [6- [3- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-016)
Cpd-016 was synthesized according to the following scheme:
Step A: 3- [6- [3- (hydroxymethyl) -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
To a solution of 3- (6-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (104.72 mg, 324.06 umol) , 8-azaspiro [4.5] decan-3-ylmethanol (100 mg, 486.09 umol, HCl) and Cs2CO3 (179.49 mg, 550.90 umol) in dioxane (5 mL) was bubbled with N2 for 5 minutes. Then 1, 3-bis [2, 6-bis (1-propylbutyl) phenyl] -4, 5-dichloro-2H-imidazol-1-ium-2-ide; 3-chloropyridine; dichloropalladium (15.76 mg, 16.20 umol) was added and the mixture was stirred at 100 ℃ for 6 hrs under N2. aq. AcOH (5%w/t) (10 mL) was added at 0 ℃ and the mixture was extracted with ethyl acetate (50 mL × 3) . The combined organic phases was dried with anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 0~10%MeOH/DCM gradient @30 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 412.1 [M+H] +.
Step B: 8- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -8-azaspiro [4.5] decane-3-
carbaldehyde
To a solution of 3- [6- [3- (hydroxymethyl) -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (30 mg, 72.91 umol) in DCM (3 mL) and DMF (1 mL) was added DMP (46.38 mg, 109.36 umol, 33.86 uL) at 0℃. The mixture was stirred at 25℃ for 2 hr. The reaction mixture was quenched with sat. aq. NaHCO3 solution (10 mL) and diluted with ethyl
acetate (40 mL × 3) . The combined organic layers was dried over Na2SO4, filtered and concentrated to afford the title compound.
LCMS: MS (ESI) m/z = 410.1 [M+H] +.
Step C: 3- [6- [3- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -8-
azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To solution of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (30 mg, 58.52 umol) and 8- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -8-azaspiro [4.5] decane-3-carbaldehyde (30 mg) in DCM (2 mL) was added TEA (37.07 mg, 366.32 umol, 50.99 uL) . The mixture was stirred at 20 ℃ for 0.5 hr. NaBH (OAc) 3 (46.58 mg, 219.79 umol) was added and the mixture was stirred at 20 ℃for 3 hrs. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: C18-1 150*30mm*5um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 38%-78%, 9min) to afford the title compound.
LCMS: MS (ESI) m/z = 906.5 [M+H] +.
1H NMR (400 MHz, CDCl3) δ: 8.82 (s, 1H) , 8.13 (s, 1H) , 7.65–7.74 (m, 2H) , 7.47 (d, J = 8.4 Hz, 2H) , 7.40 (d, J = 2.4 Hz, 1H) , 7.32 (d, J = 8.0 Hz, 1H) , 7.19 (d, J = 10.4 Hz, 1H) , 6.91 (d, J = 8.8 Hz, 2H) , 5.63–5.77 (m, 1H) , 5.22 (dd, J = 13.2, 5.2 Hz, 1H) , 5.02 (d, J = 10.8 Hz, 1H) , 4.92 (d, J = 16.0 Hz, 1H) , 4.77–4.87 (m, 1H) , 4.64–4.72 (m, 1H) , 4.37–4.44 (m, 1H) , 4.24–4.31 (m, 1H) , 3.15–3.36 (m, 8H) , 3.00–3.10 (m, 1H) , 2.81–2.97 (m, 3H) , 2.60–2.80 (m, 3H) , 2.52 (s, 2H) , 2.33–2.44 (m, 4H) , 2.17–2.29 (m, 2H) , 1.98–2.07 (m, 1H) , 1.89–1.90 (m 1H) , 1.79–1.88 (m, 2H) , 1.39–1.48 (m, 2H) , 1.24–1.35 (m, 5H) , 1.09–1.20 (m, 1H) , 0.99 (t, J = 7.6 Hz, 3H) , 0.80–0.90 (m, 2H) .
Example 17: 3- [6- [8- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -2-azaspiro [4.5] decan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-017)
Cpd-017 was synthesized according to the following scheme:
Step A: 2-azaspiro [4.5] decan-8-ylmethanol
A mixture of tert-butyl 8- (hydroxymethyl) -2-azaspiro [4.5] decane-2-carboxylate (300 mg, 1.11 mmol) in CH2Cl2 (2 mL) and TFA (1 mL) was stirred at 25 ℃ for 16 hr. The reaction mixture was concentrated in vacuo to give the title compound.
LCMS: MS (ESI) m/z = 170.2 [M+H] +.
1H NMR (ES22580-198-P1B2, 400 MHz, DMSO-d6) δ: 4.25 (d, J = 6.4 Hz, 2H) , 4.40–4.05 (m, 1H) , 3.18–3.30 (m, 2H) , 2.88 (t, J = 5.6 Hz, 2H) , 1.98 (s, 1H) , 1.75 (t, J = 7.6 Hz, 2H) , 1.57–1.64 (m, 3H) , 1.35 (d, J = 13.2, 4.0 Hz, 2H) , 1.07–1.22 (m, 4H) .
Step B: 3- [6- [8- (hydroxymethyl) -2-azaspiro [4.5] decan-2-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
A mixture of 2-azaspiro [4.5] decan-8-ylmethanol (350.67 mg) , 3- (6-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (200 mg, 618.92 umol) , Pd-PEPPSI-IHeptCl (30.10 mg, 30.95 umol) and Cs2CO3 (604.97 mg, 1.86 mmol) in dioxane (4 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 7 hrs under N2 atmosphere. aq. AcOH (5%w/t) (10 mL) was added at 0 ℃ and the mixture was extracted with ethyl acetate (50 mL × 3) . The combined organic phases was dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (5%Methanol in Dichloromethane) to give the title compound
LCMS: MS (ESI) m/z = 412.2 [M+H] +.
Step C: 2- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -2-azaspiro [4.5] decane-8-
carbaldehyde
To a solution of 3- [6- [8- (hydroxymethyl) -2-azaspiro [4.5] decan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (55 mg, 133.66 umol) in DCM (5 mL) was added DMP (85.04 mg, 200.49 umol, 62.07 uL) and DMF (1 mL) . The mixture was stirred at 25 ℃ for 2 hr. To the reaction mixture was added water (10 mL) , the pH value of the mixture was adjusted to 8 by addition of saturated aqueous solution of NaHCO3, and extracted with DCM (10 mL × 2) . The combined organic layers was washed with water (5 mL) , dried over Na2SO4, filtered and concentrated to afford the title compound.
LCMS: MS (ESI) m/z = 410.2 [M+H] +.
Step D: 3- [6- [8- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -2-
azaspiro [4.5] decan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -2-azaspiro [4.5] decane-8-carbaldehyde (60 mg) and 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (30 mg, 58.52 umol) in DCM (2 mL) was added NaBH (OAc) 3 (39.60 mg, 186.82 umol) and Et3N (63.01 mg, 622.75 umol, 86.68 uL) . The mixture was stirred at 25 ℃ for 6 hr. The mixture was poured into water (10 mL) and extracted with DCM (50 mL) , dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 47%-77%, 8 min) to afford the title compound.
LCMS: MS (ESI) m/z = 906.5 [M+H] +.
1H NMR (400 MHz, CDCl3) δ: 8.81 (s, 1H) , 8.61 (s, 1H) , 7.69 (s, 3H) , 7.46 (d, J = 2.0 Hz, 2H) , 6.97 (s, 1H) , 6.90 (s, 2H) , 6.70–6.76 (m, 1H) , 5.62–5.75 (m, 1H) , 5.22 (d, J = 7.2 Hz, 1H) , 4.98-5.05 (m, 1H) , 4.87–4.95 (m, 1H) , 4.77–4.84 (m, 1H) , 4.63–4.72 (m, 1H) , 4.22–4.40 (m, 2H) , 3.38 (s, 2H) , 3.20 (s, 4H) , 3.00–3.15 (m, 4H) , 2.78–2.95 (m, 4H) , 2.62 (s, 4H) , 2.17–2.41 (m, 8H) , 1.98–2.10 (m, 2H) , 1.92 (s, 3H) , 1.62 (s, 2H) , 1.40–1.47 (m, 2H) , 1.10–1.19 (m, 2H) , 0.99 (s, 3H) .
Example 18: 3- [5- [7- [ [1- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -4-piperidyl] methyl] -2, 7-diazaspiro [3.4] octan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-018)
Cpd-018 was synthesized according to the following scheme:
Step A: tert-butyl 2- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -2, 7-
diazaspiro [3.4] octane-7-carboxylate
A mixture of 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (250 mg, 773.65 umol) , tert-butyl 2, 6-diazaspiro [3.4] octane-6-carboxylate (153.96 mg, 618.92 umol) , Pd-PEPPSI-IHeptCl (49.12 mg, 61.89 umol) , Cs2CO3 (378.11 mg, 1.16 mmol) in dioxane (4 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 8 hours. The resulting mixture concentrated under reduce pressure to give a residue, which was purified by flash column chromatography on silica gel (MeOH in CH2Cl2 from 4%to 4%) to afford the title compound.
LCMS: MS (ESI) m/z = 455.2 [M+H] +.
Step B: 3- [5- (2, 7-diazaspiro [3.4] octan-2-yl) -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of tert-butyl 2- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -2, 7-diazaspiro [3.4] octane-7-carboxylate (170 mg, 374.02 umol) in CH2Cl2 (2 mL) was added TFA (2 mL) . The mixture was stirred at 25 ℃ for 2 hours. The reaction mixture concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 355.1 [M+H] +.
Step C: 3- [5- [7- [ [1- (4-nitrophenyl) -4-piperidyl] methyl] -2, 7-diazaspiro [3.4] octan-2-yl] -1-
oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 3- [5- (2, 7-diazaspiro [3.4] octan-2-yl) -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (120 mg) in CH2Cl2 (3 mL) was added 1- (4-nitrophenyl) piperidine-4-carbaldehyde (79.32 mg, 338.60 umol) and TEA (85.66 mg, 846.50 umol, 117.82 uL) , the mixture was stirred at 25 ℃ for 2 hours, and then NaBH (OAc) 3 (215.29 mg, 1.02 mmol) was added. The mixture was stirred at 25 ℃ for 3 hours. The residue was diluted with H2O (20 mL) and extracted with CH2Cl2 (30 mL × 3) . The combined organic layers was washed with brine (30 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was purified by flash column chromatography on silica gel (MeOH in CH2Cl2 from 5%to 7 %) to afford the title compound.
LCMS: MS (ESI) m/z = 573.2 [M+H] +.
Step D: 3- [5- [7- [ [1- (4-aminophenyl) -4-piperidyl] methyl] -2, 7-diazaspiro [3.4] octan-2-yl] -1-
oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 3- [5- [7- [ [1- (4-nitrophenyl) -4-piperidyl] methyl] -2, 7-diazaspiro [3.4] octan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (120 mg, 209.55 umol) in THF (4 mL) was added Pd/C (0.2 g, 10%w/w) . The mixture was stirred 12 hours at room temperature (25 ℃) under hydrogen atmosphere (15 Psi) . The reaction mixture was filtered through diatomite and the solid was washed with THF for 3 times. The filtrate was concentrated in vacuum to afford the title compound.
LCMS: MS (ESI) m/z = 543.2 [M+H] +.
Step E: 3- [5- [7- [ [1- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -4-piperidyl] methyl] -2, 7-
diazaspiro [3.4] octan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6-methylsulfanyl-pyrazolo [3, 4-d] pyrimidin-3-one (70.66 mg, 184.27 umol) in toluene (1 mL) was added m-CPBA (41.15 mg, 202.70 umol, 85%purity) at 0 ℃ and the mixture was stirred for 1 hour. Then 3- [5- [7- [ [1- (4-aminophenyl) -4-piperidyl] methyl] -2, 7-diazaspiro [3.4] octan-2-yl] -1-
oxo-isoindolin-2-yl] piperidine-2, 6-dione (100 mg) and DIEA (71.45 mg, 552.82 umol) were added. The mixture was stirred at 25℃ for 16 hours. The residue was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 43%-73%, 8min) to afford the title compound.
LCMS: MS (ESI) m/z = 878.5 [M+H] +.
1H NMR (400 MHz, CDCl3) δ: 8.81 (s, 1H) , 8.57 (br s, 1H) , 7.80-7.63 (m, 3H) , 7.45 (d, J = 8.4 Hz, 2H) , 7.29 (s, 1H) , 6.95-6.89 (m, 3H) , 6.66 (dd, J = 8.0, 2.0 Hz, 1H) , 7.17 (d, J = 2.0 Hz, 1H) 5.75-5.62 (m, 1H) , 5.25-5.17 (m, 1H) , 5.01 (d, J = 10.0 Hz, 1H) , 4.91 (d, J = 16.8 Hz, 1H) , 4.86-4.78 (m, 1H) , 4.72-4.60 (m, 1H) , 4.39 (d, J = 16.8 Hz, 1H) , 4.27 (d, J = 16.8 Hz, 1H) , 3.93-3.86 (m, 2H) , 3.93 (d, J = 6.8 Hz, 2H) , 3.65 (d, J = 12.4 Hz, 2H) , 3.10-3.00 (m, 1H) , 2.95-2.80 (m, 5H) , 2.75-2.65 (m, 4H) , 2.48-2.43 (m, 2H) , 2.42-2.34 (m, 2H) , 2.27-2.14 (m, 4H) , 2.06-1.90 (m, 4H) , 1.87-1.80 (m, 2H) , 1.46-1.37 (m, 2H) , 0.99 (t, J = 7.2 Hz, 3H) .
Example 19: 3- [6- [4- [ [8- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -2, 8-diazaspiro [4.5] decan-2-yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-019)
Cpd-019 was synthesized according to the following scheme:
Step A: tert-butyl 8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decane-2-carboxylate
To a solution of 1-fluoro-4-nitro-benzene (605.8 mg, 4.29 mmol, 455.5 uL) and tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (860 mg, 3.58 mmol) in DMSO (10 mL) was added DIEA (1.39 g, 10.73 mmol) . Then the reaction mixture was stirred at 120 ℃ for 12 hours. The reaction was diluted with water (10 mL) and extracted with ethyl acetate (20 mL × 3) . The combined organic layers was washed with brine (20 mL × 3) , dried over anhydrous sodium sulfate, filtered
and concentrated in vacuum to afford the title compound. as a crude, which was used in the next step without purification.
LCMS: MS (ESI) m/z = 362.2 [M+H] +.
Step B: 8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decane
To a solution of tert-butyl 8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decane-2-carboxylate (1.2 g) in DCM (10 mL) was added TFA (3.08 g, 27.01 mmol) , then the reaction mixture was stirred at 25 ℃ for 1.5 hours. The reaction mixture was concentrated in vacuum to afford the title compound.
LCMS: MS (ESI) m/z = 262.1 [M+H] +.
Step C: 2, 2, 2-trifluoro-1- [8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decan-2-yl] ethanone
To a solution of 8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decane (870 mg) in CH2Cl2 (10 mL) was added TEA (1.01 g, 9.99 mmol) and TFAA (839.10 mg, 4.00 mmol) . The mixture was stirred at 25 ℃ for 3 hours. The reaction mixture was filtered, the filtrate was concentrated in vacuum to give crude product, which was purified by flash column chromatography on silica gel (eluted with methanol in dichloromethane from 5%to 10%) to afford the title compound.
LCMS: MS (ESI) m/z = 358.1 [M+H] +.
Step D: 1- [8- (4-aminophenyl) -2, 8-diazaspiro [4.5] decan-2-yl] -2, 2, 2-trifluoro-ethanone
To a solution of 2, 2, 2-trifluoro-1- [8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decan-2-yl] ethanone (300 mg, 839.57 umol) in THF (10 mL) was added Pd/C (100 mg, 10%w/w) under H2. The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H2 (15 psi) at 25 ℃ for 12 hours. The mixture was filtered through diatomite pad under vacuum and the solid was washed with ethyl acetate (30 mL × 3) . The filtrate was evaporated to dryness to afford the title compound as a crude.
LCMS: MS (ESI) m/z = 328.1 [M+H] +.
Step E: 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [2-
(2, 2, 2-trifluoroacetyl) -2, 8-diazaspiro [4.5] decan-8-yl] anilino] pyrazolo [3, 4-d] pyrimidin-3-
one
To a solution of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6-methylsulfanyl-pyrazolo [3, 4-d] pyrimidin-3-one (140 mg, 365.09 umol) in toluene (3 mL) was added m-CPBA (148.2 mg, 730.18 umol, 85%purity) at 0 ℃, then the reaction mixture was stirred at 25 ℃ for 1.5 hours. 1- [8- (4-aminophenyl) -2, 8-diazaspiro [4.5] decan-2-yl] -2, 2, 2-trifluoro-ethanone (119.51 mg) , DIEA (141.56 mg, 1.10 mmol) were added and the mixture was stirred at 25 ℃ for 2.5 hours. The reaction mixture was purified by Biotage flash reversed-phase C-18 column eluting with MeCN in water from 10%to 80% (addition of 0.1%NH3. H2O) and lyophilized to afford the title compound.
LCMS: MS (ESI) m/z = 663.3 [M+H] +.
Step F: 2-allyl-6- [4- (2, 8-diazaspiro [4.5] decan-8-yl) anilino] -1- [ (7R) -7-ethyl-7-hydroxy-5, 6-
dihydrocyclopenta [b] pyridin-2-yl] pyrazolo [3, 4-d] pyrimidin-3-one
To a solution of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [2- (2, 2, 2-trifluoroacetyl) -2, 8-diazaspiro [4.5] decan-8-yl] anilino] pyrazolo [3, 4-d] pyrimidin-3-one (23 mg, 34.71 umol) in MeOH (0.5 mL) and H2O (0.5 mL) was added K2CO3 (9.59 mg, 69.41 umol) . The mixture was stirred at 25 ℃ for 3 hours. The reaction mixture was concentrated in vacuum to afford the title compound, which was used into the next step without further purification.
LCMS: MS (ESI) m/z = 567.2 [M+H] +.
Step G: 2- (2, 6-dioxo-3-piperidyl) -5- [4- (hydroxymethyl) -1-piperidyl] isoindoline-1, 3-dione
A mixture of 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (1 g, 3.62 mmol) , 4-piperidylmethanol (625.4 mg, 5.43 mmol) , and DIEA (1.87 g, 14.48 mmol) in DMSO (5 mL) was stirred at 120 ℃ for 1.5 hours under N2 atmosphere. The mixture was purified by reversed-phase (C18, 5%-30%CH3CN in water 0.1%FA condition) to afford the title compound.
LCMS: MS (ESI) m/z = 372.1 [M+H] +.
Step H: 3- [3-hydroxy-5- [4- (hydroxymethyl) -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-
2, 6-dione and 3- [1-hydroxy-5- [4- (hydroxymethyl) -1-piperidyl] -3-oxo-isoindolin-2-
yl] piperidine-2, 6-dione (a mixture)
To a mixture of 2- (2, 6-dioxo-3-piperidyl) -5- [4- (hydroxymethyl) -1-piperidyl] isoindoline-1, 3-dione (1.3 g, 3.61 mmol) in CH3COOH (5 mL) was added Zn (2.4 g, 36.08 mmol) , and the mixture was stirred at 60 ℃ for one hour. The mixture was filtered through filter paper under vacuum and the solid was washed with THF (30 mL × 3) . The filtrate was concentrated in vacuum to afford the title compound.
LCMS: MS (ESI) m/z = 374.1 [M+H] +.
Step I: 3- [6- [4- (hydroxymethyl) -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 3- [3-hydroxy-5- [4- (hydroxymethyl) -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione and 3- [1-hydroxy-5- [4- (hydroxymethyl) -1-piperidyl] -3-oxo-isoindolin-2-yl] piperidine-2, 6-dione (1.3 g, mixture of two region-isomers) in dichloromethane (5 mL) were added TFA (2.60 mL) and triethylsilane (1.9 g, 16.27 mmol, 2.60 mL) . The mixture was stirred at 25 ℃ for 12 hours. The reaction mixture was concentrated in vacuum to give a residue, and then aq. HCl (6 M, 5 mL) was added at 25 ℃. The resulting mixture was stirred at 50℃ for 1 hour. The mixture was purified by reversed-phase (C18, 5%-10%CH3CN in water 0.1%FA condition) to afford the title compound.
LCMS: MS (ESI) m/z = 358.2 [M+H] +.
Step J: 1- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde
To a mixture of 3- [6- [4- (hydroxymethyl) -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (100.0 mg) in DMF (2 mL) was added DMP (157.7 mg, 371.81 umol) , and the mixture was allowed to stir at 25 ℃ for 12 hours. Water (10 mL) was added and the pH value of mixture was adjusted to 8 by addition of saturated sodium bicarbonate aqueous solution. The mixture was extracted with ethyl acetate (20 mL × 2) . The combined organic layers was washed with water (20 mL) and brine (20 mL) , dried over anhydrous sodium sulfate, filtered and concentrated in vacuum to give the residue, which was purified by flash column chromatography on silica gel (eluted with methanol in dichloromethane from 0%to 4%) to afford the title compound.
LCMS: MS (ESI) m/z = 378.0 [M+Na] +.
1H NMR (400 MHz, CDCl3) δ: 9.72 (s, 1H) , 8.21 (s, 1H) , 7.40 (d, J = 2.0 Hz, 1H) , 7.34 (d, J =8.4 Hz, 1H) , 7.20 (d, J = 8.0 Hz, 1H) , 5.22 (dd, J = 5.2 13.2 Hz, 1H) , 4.50-4.20 (m, 2H) , 3.68 (td, J = 4.0, 12.4 Hz, 2H) , 2.99-2.82 (m, 4H) , 2.49-2.38 (m, 1H) , 2.38-2.28 (m, 1H) , 2.25-2.17 (m, 1H) , 2.10-2.00 (m, 2H) , 1.87 -1.78 (m, 2H)
Step K: 3- [6- [4- [ [8- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -2, 8-diazaspiro [4.5] decan-2-
yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 1- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde (5 mg, 14.07 umol) and 2-allyl-6- [4- (2, 8-diazaspiro [4.5] decan-8-yl) anilino] -1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] pyrazolo [3, 4-d] pyrimidin-3-one (17 mg) in DMF (2 mL) was added TEA (14.24 mg, 140.69 umol) , the reaction mixture was stirred at 25 ℃ for 0.5 hour. NaBH (OAc) 3 (8.95 mg, 42.21 umol) was added and the mixture was stirred for 16 hours. The reaction mixture was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 52%-82%, 8min) : 60 mL/min) and lyophilized to afford the title compound.
LCMS: MS (ESI) m/z = 906.6 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 10.97 (s, 1H) , 10.18-10.02 (m, 1H) , 8.81 (s, 1H) , 7.98-7.89 (m, 1H) , 7.69 (d, J = 8.4 Hz, 1H) , 7.61-7.51 (m, 2H) , 7.41 (d, J = 9.2 Hz, 1H) , 7.25 (d, J = 7.2 Hz, 1H) , 7.15 (s, 1H) , 6.92 (d, J = 8.4 Hz, 2H) , 5.72-5.61 (m, 1H) , 5.12-5.05 (m, 1H) , 4.99 (d, J = 10.8 Hz, 1H) , 4.88-4.82 (m, 1H) , 4.79-4.70 (m, 1H) , 4.60-4.53 (m, 1H) , 4.35-4.29 (m, 1H) , 4.22-4.17 (m, 1H) , 3.77-3.71 (m, 2H) , 3.14-3.04 (m, 4H) , 2.99-2.86 (m, 2H) , 2.82-2.65 (m, 4H) , 2.38-2.32 (m, 4H) , 2.29-2.24 (m, 2H) , 2.21-2.15 (m, 1H) , 2.05-1.95 (m, 2H) , 1.92-1.86 (m, 1H) , 1.85-1.78 (m, 2H) , 1.70-1.54 (m, 9H) , 1.25-1.20 (m, 2H) , 0.86 (t, J = 7.2 Hz, 3H) .
Example 20: 3- [5- [4- [ [3- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -3, 9-diazaspiro [5.5] undecan-9-yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-020)
Cpd-020 was synthesized according to the following scheme:
Step A: tert-butyl 9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane-3-carboxylate
To a solution of 1-fluoro-4-nitro-benzene (1 g, 7.09 mmol) in DMF (40 mL) were added tert-butyl 3, 9-diazaspiro [5.5] undecane-3-carboxylate (1.80 g, 7.09 mmol) and DIEA (28.35 mmol, 4.94 mL) . The mixture was stirred at 90 ℃ for 12 hours. The residue was diluted with H2O (200 mL) and extracted with ethyl acetate (100 mL × 3) . The combined organic layers was washed with brine (100 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was purified by reversed-phase HPLC (56%MeOH in water 0.1%FA) to afford the title compound.
LCMS: MS (ESI) m/z = 376.1 [M+H] +.
1H NMR (400 MHz, DMSO_d6) δ: 8.08 (d, J = 9.2 Hz, 2H) , 6.95 (d, J = 9.2 Hz, 2H) , 3.53-3.42 (m, 8H) , 1.66 (t, J = 5.6 Hz, 4H) , 1.52 (t, J = 5.6 Hz, 4H) , 1.46 (s, 9H) .
Step B: 3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane
To a solution of tert-butyl 9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane-3-carboxylate (200 mg, 532.68 umol) in CH2Cl2 (3 mL) was added HCl/dioxane (4 M, 3 mL) . The mixture was stirred at 25 ℃ for 3 hours. The reaction mixture concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 275.9 [M+H] +.
1H NMR (400 MHz, DMSO_d6) δ: 8.85 (br s, 2H) , 8.04 (d, J = 9.6 Hz, 2H) , 7.01 (d, J = 9.6 Hz, 2H) , 4.62 (t, J = 5.6 Hz, 4H) , 3.07-2.98 (m, 4H) , 1.66 (t, J = 5.6 Hz, 4H) , 1.57 (t, J = 5.6 Hz, 4H) .
Step C: 2, 2, 2-trifluoro-1- [9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl] ethanone
To a solution of 3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane (140 mg) in CH2Cl2 (2 mL) were added TEA (136.30 mg, 1.35 mmol, 187.49 uL) and TFAA (94.30 mg, 449.00 umol) . The mixture was stirred at 25 ℃ for 2 hours. The reaction mixture was concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 372.1 [M+H] +.
Step D: 1- [9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl] -2, 2, 2-trifluoro-ethanone
To a solution of 2, 2, 2-trifluoro-1- [9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl] ethanone (160 mg) in THF (4 mL) was added Pd/C (0.2 g, 10%w/w) The mixture was stirred 12 hours at room temperature (25 ℃) under hydrogen atmosphere (15 Psi) . The reaction mixture was filtered through diatomite and the solid was washed with THF (40mL *3) . The filtrate was concentrated in vacuum to afford the title compound.
LCMS: MS (ESI) m/z = 342.1 [M+H] +.
1H NMR (400 MHz, DMSO) δ: 6.69 (d, J = 8.8 Hz, 2H) , 6.47 (d, J = 8.8 Hz, 2H) , 4.53 (br s, 2H) , 3.63-3.50 (m, 4H) , 2.93-2.84 (m, 4H) , 1.63-1.57 (m, 4H) , 1.55-1.47 (m, 4H) .
Step E: 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [3-
(2, 2, 2-trifluoroacetyl) -3, 9-diazaspiro [5.5] undecan-9-yl] anilino] pyrazolo [3, 4-d] pyrimidin-3-
one
To a solution of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6-methylsulfanyl-pyrazolo [3, 4-d] pyrimidin-3-one (30.33 mg, 79.09 umol) in toluene (2 mL) was added m-CPBA (17.66 mg, 87.00 umol, 85%purity) at 0 ℃, and the mixture was stirred for 1 hour. 1- [9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl] -2, 2, 2-trifluoro-ethanone (27 mg) and DIEA (30.67 mg, 237.28 umol, 41.33 uL) were added and the mixture was stirred at 25 ℃for 4 hours. The mixture was purified by reversed-phase HPLC (60%-77%acetonitrile in water 0.1%NH3H2O) to afford the title compound.
LCMS: MS (ESI) m/z = 677.3 [M+H] +.
Step F: 2-allyl-6- [4- (3, 9-diazaspiro [5.5] undecan-3-yl) anilino] -1- [ (7R) -7-ethyl-7-hydroxy-
5, 6-dihydrocyclopenta [b] pyridin-2-yl] pyrazolo [3, 4-d] pyrimidin-3-one
To a solution of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [3- (2, 2, 2-trifluoroacetyl) -3, 9-diazaspiro [5.5] undecan-9-yl] anilino] pyrazolo [3, 4-d] pyrimidin-3-one 10 mg, 14.78 umol) in MeOH (1 mL) was added K2CO3 (2.45 mg, 17.73 umol) . The mixture was stirred at 25 ℃ for 1 hour. The residue was concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 581.2 [M+H] +.
Step G: 3- [5- [4- [ [3- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -3, 9-diazaspiro [5.5] undecan-9-
yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2-allyl-6- [4- (3, 9-diazaspiro [5.5] undecan-3-yl) anilino] -1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] pyrazolo [3, 4-d] pyrimidin-3-one (8 mg) in CH2Cl2 (2 mL) was added TEA (3.48 mg, 34.45 umol) and 1- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde (5.00 mg, 14.07 umol) . The mixture was stirred at 25 ℃ for 2 hours, and then NaBH (OAc) 3 (8.76 mg, 41.34 umol) was added and the mixture was stirred at 25 ℃ for 14 hours. The mixture was concentrated under reduced pressure to give a residue, which was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 47%-77%, 8 min) to afford the title compound. LCMS: MS (ESI) m/z = 920.7 [M+H] +.
HPLC: tR = 6.002 min in 0-30AB_8 min. lcm (Ultimate C18 3 um, 3.0*50 mm) .
Example 21: 3- (6- (2- (4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (Cpd-021)
Cpd-021 was synthesized according to the following scheme:
Step A: 8-azaspiro [4.5] decan-3-one
A mixture of tert-butyl 3-oxo-8-azaspiro [4.5] decane-8-carboxylate (400 mg, 1.58 mmol) in CH2Cl2 (3 mL) and TFA (1.5 mL) was stirred at 25 ℃ for 16 hr. The reaction mixture was concentrated in vacuo to give the title compound.
LCMS: MS (ESI) m/z = 154.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 3.10–3.15 (m, 2H) , 2.94–3.04 (m, 3H) , 2.23–2.30 (m, 2H) , 2.20–2.22 (m, 2H) , 1.83 (t, J = 8.0 Hz, 2H) , 1.66 (t, J = 5.6 Hz, 4H) .
Step B: 3- [1-oxo-5- (3-oxo-8-azaspiro [4.5] decan-8-yl) isoindolin-2-yl] piperidine-2, 6-dione
Schlenk tube charged with a mixture of 3- (6-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (72.55 mg, 224.51 umol) , 8-azaspiro [4.5] decan-3-one (100 mg) , Pd-PEPPSI-IHeptCl (10.92 mg, 11.23 umol) and Cs2CO3 (219.45 mg, 673.54 umol) in dioxane (4 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 6 hr under N2 atmosphere. aq. AcOH (5%w/t) (50 mL) was added at 0 ℃, and extracted with ethyl acetate (50 mL × 3) . The combined organic phases was dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-TLC (SiO2, Dichloromethane: Methanol =10: 1) to afford the title compound.
LCMS: MS (ESI) m/z = 396.2 [M+H] +.
Step C: 3- (6- (2- (4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-
yl) piperidine-2, 6-dione
To a solution of 3- [1-oxo-5- (3-oxo-8-azaspiro [4.5] decan-8-yl) isoindolin-2-yl] piperidine-2, 6-dione (45 mg) and 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (30 mg, 58.52 umol) in CH2Cl2 (3 mL) was added NaBH (OAc) 3 (25.32 mg, 119.48 umol) and Et3N (40.30 mg, 398.28 umol, 55.44 uL) . The mixture was stirred at 25 ℃ for 16 hr. The mixture was poured into water (10 mL) and extracted with DCM (50 mL) . The organic layer was dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by prep-HPLC (column: Phenomenex C18 75*30mm*3um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 36%-76%, 9 min) to afford the title compound.
LCMS: MS (ESI) m/z = 892.7 [M+H] +.
1H NMR (400 MHz, CDCl3) δ: 8.81 (s, 1H) , 8.40–8.55 (m, 1H) , 7.61–7.78 (m, 3H) , 7.38–7.49 (m, 3H) , 7.19 (d, J = 4.8 Hz, 1H) , 6.91 (d, J = 3.2 Hz, 2H) , 5.60–5.76 (m, 1H) , 5.17–5.29 (m, 1H) , 4.99–5.07 (m, 1H) , 4.91 (d, J = 16.4 Hz, 1H) , 4.76–4.85 (m, 1H) , 4.65–4.74 (m, 1H) , 4.25–4.44 (m, 2H) , 3.24 (s, 10H) , 3.01–3.08 (m, 1H) , 2.82–2.92 (m, 3H) , 2.74 (s, 4H) , 2.18–2.45 (m, 6H) , 1.90–2.06 (m, 5H) , 1.54–1.60 (m, 2H) , 1.45–1.53 (m, 2H) , 1.25–1.34 (m, 2H) , 0.99 (s, 3H) .
Example 22: 3- (5- (4- ( (S) -3- ( (4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) pyrrolidin-1-yl) piperidin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (Cpd-022)
Cpd-022 was synthesized according to the following scheme:
Step A: 3- [5- (1, 4-dioxa-8-azaspiro [4.5] decan-8-yl) -1-oxo-isoindolin-2-yl] piperidine-2, 6-
dione
Schlenk tube charged with a mixture of 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (200 mg, 618.92 umol) , 1, 4-dioxa-8-azaspiro [4.5] decane (70.90 mg, 495.14 umol) , and Pd-PEPPSI-IHeptCl (24.56 mg, 30.95 umol) , Cs2CO3 (302.49 mg, 928.39 umol) in dioxane (3 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 8 hours. The resulting mixture was concentrated under reduce pressure to give a residue, which was purified by flash column chromatography on silica gel (MeOH in CH2Cl2 from 0%to 10%) to afford the title compound.
LCMS: MS (ESI) m/z = 386.1 [M+H] +.
Step B: 3- [1-oxo-5- (4-oxo-1-piperidyl) isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 3- [5- (1, 4-dioxa-8-azaspiro [4.5] decan-8-yl) -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (30 mg, 77.84 umol) in HCOOH (3 mL) was added TfOH (11.68 mg, 77.84 umol) at 25 ℃ for 2 hours. The reaction mixture was concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 342.1 [M+H] +.
Step C: 2, 2, 2-trifluoro-1- [ (3R) -3- (hydroxymethyl) pyrrolidin-1-yl] ethanone
A mixture of [ (3R) -pyrrolidin-3-yl] methanol (200 mg, 1.98 mmol) was dissolved in THF (2 mL) , to which ethyl 2, 2, 2-trifluoroacetate (95 mg, 2.08 mmol) dissolved in THF (2 mL) was added, and the mixture was allowed to stir at 25 ℃ for 12 hours. The reaction mixture was concentrated in vacuo to afford the title compound.
LCMS: MS (ESI) m/z = 198.1 [M+H] +.
Step D: (3R) -1- (2, 2, 2-trifluoroacetyl) pyrrolidine-3-carbaldehyde
A mixture of 2, 2, 2-trifluoro-1- [ (3R) -3- (hydroxymethyl) pyrrolidin-1-yl] ethanone (389.0 mg) was dissolved in dichloromethane (10 mL) , to which was added DMP (1.26 g, 2.96 mmol) at 0 ℃, and the mixture was allowed to react at 0 ℃ for 6 hours. The reaction was diluted with water (20 mL) and extracted with ethyl acetate (40 mL × 2) . The combined organic layers was washed with brine (20 mL × 2) , dried over anhydrous sodium sulfate, filtered and concentrated in vacuum to give the residue, which was purified by flash column chromatography on silica gel (eluted with ethyl acetate in petroleum ether from 30%to 50%) to afford the title compound.
LCMS: MS (ESI) m/z = 196.1 [M+H] +.
Step E: 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [4-
[ [ (3S) -1- (2, 2, 2-trifluoroacetyl) pyrrolidin-3-yl] methyl] piperazin-1-yl] anilino] pyrazolo [3, 4-
d] pyrimidin-3-one
To a solution of (3R) -1- (2, 2, 2-trifluoroacetyl) pyrrolidine-3-carbaldehyde (20.25 mg) in CH2Cl2 (2 mL) were added 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (25 mg, 48.77 umol) and TEA (12.34 mg, 121.93 umol, 16.97 uL) . The mixture was stirred at 25 ℃ for 2 hours and NaBH (OAc) 3 (31.01 mg, 146.31 umol) was added. The mixture was stirred at 25 ℃ for 16 hours. The residue was diluted with H2O (20 mL) and extracted with CH2Cl2 (90 mL) . The combined organic layers was washed with brine (30 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give crude product, which was purified by reversed-phase HPLC (35-50%acetonitrile in water 0.1%NH3-H2O) to afford the title compound.
LCMS: MS (ESI) m/z = 692.4 [M+H] +.
Step F: 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [4-
[ [ (3R) -pyrrolidin-3-yl] methyl] piperazin-1-yl] anilino] pyrazolo [3, 4-d] pyrimidin-3-one
To a solution of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [4- [ [ (3S) -1- (2, 2, 2-trifluoroacetyl) pyrrolidin-3-yl] methyl] piperazin-1-yl] anilino] pyrazolo [3, 4-d] pyrimidin-3-one (30 mg) in MeOH (2 mL) was added K2CO3 (5.03 mg, 36.43 umol) . The mixture was stirred at 25 ℃ for 1 hour. The reaction was concentrated under reduce pressure to afford the title compound.
LCMS: MS (ESI) m/z = 596.3 [M+H] +.
Step G: 3- (5- (4- ( (S) -3- ( (4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) pyrrolidin-1-yl) piperidin-1-yl) -1-oxoisoindolin-2-
yl) piperidine-2, 6-dione
To a solution of 3- [1-oxo-5- (4-oxo-1-piperidyl) isoindolin-2-yl] piperidine-2, 6-dione (10.3 mg) and 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [4- [ [ (3S) -pyrrolidin-3-yl] methyl] piperazin-1-yl] anilino] pyrazolo [3, 4-d] pyrimidin-3-one (18.0 mg) in CH2Cl2 (2 mL) was added TEA (7.64 mg, 75.54 umol, 10.51 uL) . The mixture was stirred at 25 ℃ for 2 hours and NaBH (OAc) 3 (19.21 mg, 90.64 umol) was added. The mixture was stirred at 25 ℃ for 16 hours. The mixture was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 42%-72%, 8min) to afford the title compound.
LCMS: MS (ESI) m/z = 921.7 [M+H] +.
1H NMR (400 MHz, CDCl3) δ: 8.83 (s, 1H) , 8.15-8.03 (m, 1H) , 7.74-7.66 (m, 2H) , 7.49-7.46 (m, 2H) , 7.43-7.38 (m, 1H) , 7.36-7.29 (m, 2H) , 7.18 (dd, J = 8.4, 2.4 Hz, 1H) , 6.90 (d, J = 8.8 Hz, 2H) , 5.77-5.64 (m, 1H) , 5.37-5.34 (m, 2H) , 5.24-5.17 (m, 1H) , 5.03 (d, J=10.4 Hz, 1H) , 4.96-4.88 (m, 1H) , 4.86-4.78 (m, 1H) , 4.73-4.64 (m, 1H) , 4.41 (d, J = 15.6 Hz, 1H) , 4.29 (d, J =15.6 Hz, 1H) , 3.88-3.81 (m, 2H) , 3.22-3.12 (m, 4H) , 3.12-3.04 (m, 2H) , 2.93-2.75 (m, 6H) , 2.74-2.67 (m, 2H) , 2.65-2.56 (m, 2H) , 2.45-2.33 (m, 4H) , 2.26-2.19 (m, 6H) , 2.03-2.00 (m, 4H) , 1.86-1.83 (m, 2H) , 0.99 (t, J = 6.4 Hz, 3H) .
Example 23: 3- (5- (4- ( (1s, 3s) -3- ( (4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) cyclobutyl) piperazin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (Cpd-023)
Cpd-023 was synthesized according to the following scheme:
Step A: tert-butyl 4- (3-methoxycarbonylcyclobutyl) piperazine-1-carboxylate
To a solution of methyl 3-oxocyclobutanecarboxylate (2 g, 15.61 mmol) , tert-butyl piperazine-1-carboxylate (2.91 g, 15.61 mmol) in DCM (20 mL) was added NaBH (OAc) 3 (3.97 g, 18.73 mmol) and TEA (2.37 g, 23.41 mmol, 3.26 mL) . The mixture was stirred at 25 ℃ for 16 hrs. The mixture was concentrated to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate = 0%to 50%) to afford the title compound.
LCMS: MS (ESI) m/z = 299.0 [M+H] +) .
1H NMR (400 MHz, CDCl3) δ: 3.67 (s, 3H) , 3.44 –3.42 (m, 4H) , 2.78 –2.72 (m, 2H) , 2.33 –2.27 (m, 6H) , 2.27 –2.15 (m, 2H) , 1.45 (s, 9H) .
Step B: 4- [3- (hydroxymethyl) cyclobutyl] piperazine-1-carboxylate
To a solution of tert-butyl 4- (3-methoxycarbonylcyclobutyl) piperazine-1-carboxylate (1 g, 3.35 mmol) in THF (15 mL) was added LiAlH4 (190.80 mg, 5.03 mmol) in THF (15 mL) at 0 ℃. The mixture was stirred at 0 ℃ for 2 hr. The reaction mixture was quenched by addition of 15 %NaOH (0.5 mL) at 0 ℃. The reaction mixture was filtered and the solid was washed with THF (30 mL × 3) . And the filtrate was concentrated in vacuum to afford the title compound.
Step C: 3-piperazin-1-ylcyclobutyl) methanol
To a solution of tert-butyl 4- [3- (hydroxymethyl) cyclobutyl] piperazine-1-carboxylate (720 mg) in DCM (5 mL) was added HCI/dioxane (4 M, 5 mL) . The mixture was stirred at 25 ℃ under N2 atmosphere for 16 hr. The mixture was concentrated to afford the title compound.
Step D: 3- [5- [4- [3- (hydroxymethyl) cyclobutyl] piperazin-1-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
To a solution of 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (100 mg, 309.46 umol) and (3-piperazin-1-ylcyclobutyl) methanol (102.35 mg , HCl salt) in dioxane (4.0 mL) was added 1, 3-bis [2, 6-bis (1-propylbutyl) phenyl] -4, 5-dichloro-2H-imidazol-1-ium-2-ide; 3-chloropyridine; dichloropalladium (30.10 mg, 30.95 umol) , Cs2CO3 (302.49 mg, 928.38 umol) . The mixture was stirred at 120 ℃ for 15 hr under nitrogen atmosphere. The reaction mixture was diluted with water (8.0 mL) , acidified with HCl (1 N) to pH 6~7 and extracted with ethyl acetate (5 mL × 2) . The aqueous layer was freeze-dried in vacuum. The obtained solid was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; B%: 0%-25%, 8min) to afford the title compound.
LCMS : MS (ESI) m/z = 413.2 [M+H] +) .
Step E: 3- [4- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] piperazin-1-
yl] cyclobutanecarbaldehyde
To a solution of 3- [5- [4- [3- (hydroxymethyl) cyclobutyl] piperazin-1-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (30 mg, 72.73 umol) in DCM (1.0 mL) and DMF (1.0 mL) was added Dess-Martin reagent (46.27 mg, 109.10 umol, 33.78 uL) . The mixture was stirred at 25 ℃ for 15 hrs. The reaction mixture was concentrated in vacuum to remove organic solvents. The residue was purified by prep-HPLC (column: YMC-Actus Triart C18 150*30mm*5um; mobile phase: [water (TFA) -ACN] ; B%: 0%-20%, 10.5min) to afford the title compound.
LCMS: MS (ESI) m/z = 411.0 [M+H] +) .
Step F: 3- (5- (4- ( (1s, 3s) -3- ( (4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) cyclobutyl) piperazin-1-yl) -1-oxoisoindolin-2-
yl) piperidine-2, 6-dione
To a solution of 3- [4- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] piperazin-1-yl] cyclobutanecarbaldehyde (17 mg, 41.42 umol) and 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (25.48 mg, 49.70 umol) in DCM (1.0 mL) and DMSO (0.5 mL) was added TEA (20.95 mg, 207.08 umol, 28.82 uL) . After stirring at 25 ℃ for 1 hr, NaBH (OAc) 3 (13.17 mg, 62.12 umol) was added. The mixture was stirred at 25 ℃ for 15 hrs. The reaction mixture was concentrated
to remove DCM, quenched with water (0.5 mL) . The residue was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 35%-55%, 16min) to afford the title compound.
LCMS: MS (ESI) m/z = 907.4 [M+H] +) .
Example 24: 3- [5- [3- [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-024)
Cpd-024 was synthesized according to the following scheme:
Step A: 3- [1-oxo-5- (3-oxo-8-azaspiro [4.5] decan-8-yl) isoindolin-2-yl] piperidine-2, 6-dione
Schlenk tube charged with a mixture of 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (102.93 mg, , 318.54 umol) , 8-azaspiro [4.5] decan-3-one (51.08 mg, 191.12 umol) , Cs2CO3 (310.97 mg, 954.43 umol) and 1, 3-bis [2, 6-bis (1-propylbutyl) phenyl] -4, 5-dichloro-2H-imidazol-1-ium-2-ide; 3-chloropyridine; dichloropalladium (15.49 mg, 15.93 umol) in dioxane (3 mL) was degassed with N2. The mixture was stirred at 95 ℃ for 16 h under N2. aq. AcOH (5%w/t) (50 mL) was added and the mixture was extracted with ethyl acetate (50 mL × 3) . The combined organic phases was dried with anhydrous Na2SO4, filtered and concentrated in under reduce pressure. The residue was purified by prep-TLC (SiO2, Dichloromethane: Methanol = 10: 1) to afford the title compound.
LCMS: MS (ESI) m/z = 396.2 [M+H] +.
Step B: 3- [5- [3- [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] -8-
azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a mixture of 3- [1-oxo-5- (3-oxo-8-azaspiro [4.5] decan-8-yl) isoindolin-2-yl] piperidine-2, 6-dione (34 mg, 85.98 umol) , 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (29.97 mg, 58.46 umol) and TEA (43.50 mg, 429.89 umol) in DCM (3 mL) was added NaBH (OAc) 3 (27.33 mg, 128.97 umol) . The mixture was stirred at 25 ℃ for 32 h under N2. H2O (50 mL) was added and the mixture was extracted with ethyl acetate (50 mL × 3) . The combined organic phases was washed dried with anhydrous Na2SO4, filtered and concentrated in under reduced pressure. The crude product was purified by prep-HPLC (column: C18-1 150*30mm*5um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 25%-65%, 9 min) to afford the title compound.
LCMS: MS (ESI) m/z = 892.4 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.83 (s, 1H) , 7.97 (s, 1H) , 7.64-7.76 (m, 3H) , 7.48 (d, J = 7.6 Hz, 2H) , 7.00 (d, J = 4.4 Hz, 1H) , 6.92 (d, J = 8.8 Hz, 2H) , 6.89 (s, 1H) , 5.65-5.77 (m, 1H) , 5.21 (dd, J = 13.6, 4.8 Hz, 1H) , 5.03 (d, J = 10.0 Hz, 1H) , 4.92 (d, J = 16.4 Hz, 1H) , 4.76-4.87 (m, 1H) , 4.63-4.72 (m, 1H) , 4.37-4.45 (m, 1H) , 4.22-4.30 (m, 1H) , 3.33 (s, 6H) , 3.05 (s, 1H) , 2.81-2.92 (m, 3H) , 2.68-2.80 (m, 2H) , 2.17-2.44 (m, 5H) , 1.97-2.06 (m, 2H) , 1.85 (dd, J = 14.2, 7.6 Hz, 2H) , 1.64-1.73 (m, 6H) , 1.26 (s, 6H) , 0.99 (t, J = 7.6 Hz, 3H) .
Example 25: 3- [5- [4- [ [3- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -3, 9-diazaspiro [5.5] undecan-9-yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-025)
Cpd-025 was synthesized according to the following scheme:
Step A: 3- [5- [4- (hydroxymethyl) -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
A mixture of 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (300 mg, 928.39 umol) , and 4-piperidylmethanol (534.62 mg, 4.64 mmol) , Pd-PEPPSI-IHeptCl (58.95 mg, 74.27 umol) , Cs2CO3 (453.73 mg, 1.39 mmol) in dioxane (3 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 5 hours. The residue was concentrated under reduced pressure, which was purified by flash column chromatography on silica gel (MeOH in CH2Cl2 from3%to 4%) to afford the title compound.
LCMS: MS (ESI) m/z = 358.1 [M+H] +.
Step B: 1- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde
To a solution of 3- [5- [4- (hydroxymethyl) -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (50 mg, 139.90 umol) in DMF (3 mL) was added DMP (89 mg, 209.85 umol) . The mixture was stirred at 0-25 ℃ for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue, which was purified by reversed-phase HPLC (60%-77%acetonitrile in water 0.1%NH3H2O) to afford the title compound.
LCMS: MS (ESI) m/z = 356.1 [M+H] +.
Step C: 3- [5- [4- [ [3- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -3, 9-diazaspiro [5.5] undecan-9-
yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2-allyl-6- [4- (3, 9-diazaspiro [5.5] undecan-3-yl) anilino] -1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] pyrazolo [3, 4-d] pyrimidin-3-one (50 mg, 86.10 umol) in CH2Cl2 (2 mL) was added TEA (21.78 mg, 215.25 umol) and 1- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde (30.60 mg, 86.10 umol) . The mixture was stirred at 25 ℃ for 2 hours, and then NaBH (OAc) 3 54.74 mg, 258.30 umol) was added. The mixture was stirred at 25 ℃ for 14 hours. The mixture was concentrated under
reduced pressure to give a residue, which was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 47%-77%, 8min) to afford the title compound.
LCMS: MS (ESI) m/z = 920.7 [M+H] +.
1H NMR (ES22702-196-P1D1, 400 MHz, DMSO-d6) δ: 10.97 (s, 1H) , 10.09 (br s, 1H) , 8.81 (s, 1H) , 7.98-7.89 (m, 1H) , 7.69 (d, J = 8.0 Hz, 1H) , 7.63-7.52 (m, 2H) , 7.40 (d, J = 8.0 Hz, 1H) , 7.24 (d, J = 8.4 Hz, 1H) , 7.18-7.15 (m, 1H) , 6.91 (d, J = 8.8 Hz, 2H) , 5.74-5.60 (m, 1H) , 5.13-5.04 (m, 2H) , 4.99 (d, J = 10.4 Hz, 1H) , 4.85 (d, J = 16.4 Hz, 1H) , 4.80-4.67 (m, 1H) , 4.60-4.50 (m, 1H) , 4.32 (d, J = 17.2 Hz, 1H) , 4.20 (d, J = 17.2 Hz, 1H) , 3.76 -3.69 (m, 2H) , 3.12-3.06 (m, 4H) , 2.97-2.93 (m, 2H) , 2.83-2.73 (m, 2H) , 2.72-2.65 (m, 2H) , 2.36-2.33 (m, 4H) , 2.23-2.15 (m, 3H) , 2.05-1.93 (m, 3H) , 1.93-1.75 (m, 4H) , 1.74-1.65 (m, 3H) , 1.55-1.42 (m, 8H) , 0.86 (t, J =7.2Hz, 3H) .
Example 26: 3- [5- [4- [ [8- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -2, 8-diazaspiro [4.5] decan-2-yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-026)
Cpd-026 was synthesized according to the following scheme:
Step A: 3- [5- [4- [ [8- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -2, 8-diazaspiro [4.5] decan-2-
yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2-allyl-6- [4- (2, 8-diazaspiro [4.5] decan-8-yl) anilino] -1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] pyrazolo [3, 4-d] pyrimidin-3-one (20 mg, 35.29 umol) and
1- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde (20 mg, 56.28 umol) in DMF (2 mL) was added TEA (56.9 mg, 562.77 umol) . After stirred at 25 ℃ for 0.5 hour, NaBH (OAc) 3 (35.78 mg, 168.83 umol) was added and the mixture was stirred at 25 ℃ for 18 hours. The reaction mixture was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 52%-82%, 8min) : 60 mL/min) and lyophilized to afford the title compound.
LCMS: MS (ESI) m/z = 906.6 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 10.95 (s, 1H) , 10.22-9.98 (m, 1H) , 8.82 (s, 1H) , 7.97-7.88 (m, 1H) , 7.69 (d, J = 8.0 Hz, 1H) , 7.63-7.52 (m, 2H) , 7.50 (d, J = 9.2 Hz, 1H) , 7.08-7.00 (m, 2H) , 6.92 (d, J = 8.8 Hz, 2H) , 5.74-5.60 (m, 1H) , 5.07 (s, 1H) , 5.05-4.98 (m, 2H) , 4.85 (d, J =17.2 Hz, 1H) , 4.80-4.70 (m, 1H) , 4.63-4.52 (m, 1H) , 4.35-4.28 (m, 1H) , 4.23-4.16 (m, 1H) , 3.87 (d, J = 12.4 Hz, 2H) , 3.13-3.04 (m, 4H) , 3.01-2.89 (m, 2H) , 2.88-2.73 (m, 4H) , 2.41-2.30 (m, 4H) , 2.28-2.23 (m, 2H) , 2.22-2.16 (m, 1H) , 2.05-1.87 (m, 3H) , 1.84-1.77 (m, 2H) , 1.73-1.55 (m, 9H) , 1.24-1.15 (m, 2H) , 0.87 (t, J = 7.2 Hz, 3H)
Example 27: 3- [6- [4- [ [3- [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] pyrrolidin-1-yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-027)
Cpd-027 was synthesized according to the following scheme:
Step A: 1- (2, 2, 2-trifluoroacetyl) pyrrolidin-3-one
TFAA (766.0 mg, 4.11 mmol) was added to pyrrolidin-3-one (500.0 mg, 4.11 mmol, HCl salt) , the reaction mixture was stirred at 25 ℃ for 2 hours. The mixture was concentrated to provide a residue. The residue was purified by flash column chromatography on silica gel (eluted with methanol in dichloromethane from 0%to 5%) to afford the title compound.
LCMS: MS (ESI) m/z = 182.1 [M+H] +.
Step B: 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [4- [1-
(2, 2, 2-trifluoroacetyl) pyrrolidin-3-yl] piperazin-1-yl] anilino] pyrazolo [3, 4-d] pyrimidin-3-
one
To a mixture of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (50.0 mg, 97.54 umol) and 1- (2, 2, 2-trifluoroacetyl) pyrrolidin-3-one (50.0 mg) in methyl alcohol (5 mL) was added AcOH (58.6 mg, 975.41 umol) , the reaction mixture was stirred at 25 ℃ for 0.5 hour. Then NaBH3CN (18.4 mg, 292.62 umol) was added, and the reaction mixture was stirred at 25 ℃ for 16 hours. The mixture was purified by flash reversed-phase C-18 column eluting with methyl alcohol in water from 10%to 80% (addition of 0.1%NH3. H2O) and to afford the title compound.
LCMS: MS (ESI) m/z = 678.3 [M+H] +.
Step C: 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- (4-
pyrrolidin-3-ylpiperazin-1-yl) anilino] pyrazolo [3, 4-d] pyrimidin-3-one
To a mixture of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [4- [1- (2, 2, 2-trifluoroacetyl) pyrrolidin-3-yl] piperazin-1-yl] anilino] pyrazolo [3, 4-d] pyrimidin-3-one (14.0 mg, 20.66 umol) in methanol (4 mL) was added K2CO3 (4.3 mg, 30.99 umol) . Then the reaction mixture was stirred at 25 ℃ for 4 hours. The reaction mixture was filtered, the filtrate was concentrated in vacuum to afford the title compound. which was used in the next step without further purification.
LCMS: MS (ESI) m/z = 582.3 [M+H] +.
Step D: 3- [6- [4- [ [3- [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-
dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-
yl] amino] phenyl] piperazin-1-yl] pyrrolidin-1-yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
To a mixture of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- (4-pyrrolidin-3-ylpiperazin-1-yl) anilino] pyrazolo [3, 4-d] pyrimidin-3-one (15.0 mg) and 1- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde (10.1 mg, 28.37 umol) in DMF (3 mL) was added TEA (26.1 mg, 257.90 umol) . After stirred at 0.5 hour, NaBH (OAc) 3 (16.4 mg, 77.37 umol) was added and the mixture was stirred at 25 ℃ for another 12 hours. The reaction mixture was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 52%-82%, 8min) : 60 mL/min) to afford the title compound.
LCMS: tm/z = 921.7 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 10.97 (s, 1H) , 10.20-10.06 (m, 1H) , 8.82 (s, 1H) , 7.92 (dd, J = 2.0, 10.4 Hz, 1H) , 7.71-7.68 (m, 1H) , 7.69 (d, J = 8.0 Hz, 1H) , 7.62-7.52 (m, 2H) , 7.41 (d, J =8.4 Hz, 1H) , 7.25 (dd, J = 2.0, 8.4 Hz, 1H) , 7.15 (d, J = 2.0 Hz, 1H) , 6.93-6.88 (m, 2H) , 5.72-5.61 (m, 1H) , 5.13-5.04 (m, 2H) , 4.99 (d, J = 10.0 Hz, 1H) , 4.85 (d, J = 17.2 Hz, 1H) , 4.80-4.69 (m, 1H) , 4.61-4.52 (m, 1H) , 4.36-4.29 (m, 1H) , 4.23-4.16 (m, 1H) , 3.78-3.70 (m, 2H) , 3.13-3.05 (m, 1H) , 3.12-3.05 (m, 3H) , 3.05-3.05 (m, 1H) , 3.00-2.78 (m, 4H) , 2.76-2.65 (m, 4H) , 2.62-2.56 (m, 2H) , 2.37-2.28 (m, 4H) , 2.26-2.14 (m, 3H) , 2.05-1.94 (m, 2H) , 1.93-1.76 (m, 5H) , 1.73-1.62 (m, 3H) , 1.27-1.18 (m, 2H) , 0.87 (t, J = 7.2 Hz, 3H) .
Example 28: 5- (4- (2- (4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) acetyl) piperazin-1-yl) -2- (2, 6-dioxopiperidin-3-yl) isoindoline-1, 3-dione (Cpd-028)
Cpd-028 was synthesized according to the following scheme:
Step A: Ethyl (R) -2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) acetate
To a solution of (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- ( (4- (piperazin-1-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (30.0 mg, 0.059 mmol) and ethyl 2-bromoacetate (15.0 mg, 0.088 mmol) in DMF (10 mL) was added K2CO3 (24.0 mg, 0.176 mmol) . The mixture was warmed to 100℃ and stirred at 100℃ for 3hrs. The reaction mixture was quenched by water (5 mL) and was extracted with ethyl acetate (10 mL x 3) . The combined organic layers was washed with brine (10 mL) , dried over Na2SO4 concentrated under reduced pressure. The crude product was purified by pre-TLC with DCM: MeOH =20: 1 to afford the title compound.
LC-MS (ESI) [M+H] + 599.3.
Step B: (R) -2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-
yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-
yl) acetic acid
To a solution of ethyl (R) -2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) acetate (31.0 mg, 0.052 mmol) in THF/MeOH/H2O (1 mL/1 mL/1 mL) was added LiOH. H2O (10.4 mg, 0.248 mmol) . The mixture was stirred at 30℃ for 2hrs. The mixture was concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 571.3.
Step C: 5- (4- (2- (4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) acetyl) piperazin-1-yl) -2- (2, 6-dioxopiperidin-3-
yl) isoindoline-1, 3-dione
To a solution of (R) -2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) acetic acid (25.0 mg) , 2- (2, 6-dioxopiperidin-3-yl) -5- (piperazin-1-yl) isoindoline-1, 3-dione hydrochloride (16.6 mg, 0.0438 mmol) and DIEA (16.9 mg, 0.131 mmol) in DMF (2 mL) was added HATU (16.6 mg, 0.0438 mmol) . The mixture was stirred at 30℃ for 1hr. The mixture was concentrated and the residue was purified by prep-HPLC (FA) (Waters 2767, Column: Pursuit XRs 10 C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 25%~35%; Retention Time: 8.7-10.5min of 18 min. ) to afford the title compound.
LC-MS (ESI) [M+H] + 895.5.
1H NMR (400 MHz, DMSO) δ 11.08 (s, 1H) , 10.12 (s, 1H) , 8.82 (s, 1H) , 8.40 (s, 1H) , 7.92 (s, 1H) , 7.77 –7.66 (m, 2H) , 7.58 (s, 2H) , 7.38 (t, J = 7.7 Hz, 2H) , 6.93 (d, J = 8.0 Hz, 2H) , 5.67 (d, J = 6.0 Hz, 1H) , 5.05 (dd, J = 44.0, 8.7 Hz, 3H) , 4.85 (d, J = 17.2 Hz, 1H) , 4.74 (s, 1H) , 4.58 (s, 1H) , 3.80 (s, 2H) , 3.67 (s, 2H) , 3.27 (s, 6H) , 3.12 (s, 4H) , 2.98 –2.72 (m, 4H) , 2.60 (s, 5H) , 2.20 (s, 1H) , 2.03 (s, 2H) , 1.88 (s, 1H) , 1.71 (s, 1H) , 0.88 (t, J = 7.3 Hz, 3H) .
Example 29: 3- (5- (1- (2- (1- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperidin-4-yl) -2-azaspiro [3.5] nonan-7-yl) -1H-pyrazol-4-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (Cpd-029)
Cpd-029 was synthesized according to the following scheme:
Step A: 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6-
(methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one
To a solution of (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylthio) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (500 mg, 1.31 mmol) in DCM (30 mL) was added a solution of m-chloroperbenzoic acid (337.81 mg, 1.96 mmol) in DCM (20 mL) under ice-cooling, and the mixture is stirred at 25 ℃ for 2 hours. The reaction mixture is washed with a saturated aqueous sodium hydrogen carbonate solution and brine, and the organic layer was dried over anhydrous sodium sulfate, and the solvent was evaporated in vacuo to afford the title compound.
LC-MS (ESI) [M+H] + 400.1
Step B: 1- (4-aminophenyl) piperidin-4-one
To a solution of 1- (4-nitrophenyl) piperidin-4-one (1000 mg, 4.54 mmol) in MeOH (10 mL) was added Raney-Ni (300 mg) . Then the reaction mixture was stirred at 25℃ for 18 hrs under H2 atmosphere. After completion of reaction, the reaction mixture was filtered, and the filtrate was concentrated by vacuum to afford the title compound., which was used in the next step without further purification.
LC-MS (ESI) [M+H] + = 191.2
Step C: (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- ( (4-
(4-oxopiperidin-1-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one
To a solution of 1- (4-aminophenyl) piperidin-4-one (50.0 mg) in DCM (1 mL) was added 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (105 mg, 0.263 mmol) . Then the reaction mixture was stirred at 25℃ for 18 h. After completion of reaction, the reaction mixture was concentrated by vacuum, and the residue was purified by flash silica gel column chromatography (DCM/MeOH=5/1) to afford the title compound.
LC-MS (ESI) [M+H] + = 526.3
Step D: tert-butyl 7- ( (methylsulfonyl) oxy) -2-azaspiro [3.5] nonane-2-carboxylate
To a solution of tert-butyl 7-hydroxy-2-azaspiro [3.5] nonane-2-carboxylate (1000 mg, 4.15 mmol) , TEA (838 mg, 8.30 mmol) in DMF (20 mL) was added MsCl (573 mg, 4.98 mmol) . Then the reaction mixture was stirred at 25℃ for 3 hrs. After completion of reaction, the reaction mixture was diluted with H2O (50 mL) and extracted with EtOAc (50 mL x 3) , the organic layers was washed with water (50 mL) and brine (50 mL) , dried with Na2SO4, filtered, the filtrate was concentrated by vacuum to afford the title compound.
Step E: tert-butyl 7- (4-bromo-1H-pyrazol-1-yl) -2-azaspiro [3.5] nonane-2-carboxylate
To a solution of tert-butyl 7- ( (methylsulfonyl) oxy) -2-azaspiro [3.5] nonane-2-carboxylate (1000 mg) , Cs2CO3 (3.06 g, 9.39 mmol) in DMF (20 mL) was added 4-bromo-1H-pyrazole (460 mg, 3.13 mmol) . Then the reaction mixture was stirred at 80℃ for 18 h. After completion of
reaction, the reaction mixture was diluted with H2O (50 mL) and extracted with EtOAc (50 mL x 3) . The combined organic layers was washed with water (50 mL × 2) and brine (50 mL) , dried with Na2SO4, filtered, and the filtrate was concentrated by vacuum, the residue was purified by flash silica gel column chromatography (PE/EA=10/1) to afford the title compound.
LC-MS (ESI) [M+H-t-Bu] + 314.2.
Step F: tert-butyl 7- (4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-pyrazol-1-yl) -2-
azaspiro [3.5] nonane-2-carboxylate
To a solution of tert-butyl 7- (4-bromo-1H-pyrazol-1-yl) -2-azaspiro [3.5] nonane-2-carboxylate (1000 mg, 2.70 mmol) , KOAc (794 mg, 8.10 mmol) in DMF (20 mL) were added B2Pin2 (1.03 g, 4.05 mmol) and Pd (dppf) Cl2 (196 mg, 0.270 mmol) . Then the reaction mixture was stirred at 80℃ for 8 hrs under argon atmosphere. The mixture was diluted with H2O (50 mL) and extracted with EtOAc (100 mL x 2) , The combined organic layers was washed with brine (50 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE/EA=5/1) to afford the title compound.
LC-MS (ESI) [1/2M+H] + = 209.0
Step G: tert-butyl 7- (4- (2- (2, 6-dioxopiperidin-3-yl) -1-oxoisoindolin-5-yl) -1H-pyrazol-1-yl) -
2-azaspiro [3.5] nonane-2-carboxylate
To a solution of tert-butyl 7- (4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-pyrazol-1-yl) -2-azaspiro [3.5] nonane-2-carboxylate (100 mg, 0.240 mmol) , Na2CO3 (76.0 mg, 0.720 mmol) in dioxane/H2O (2/0.2 mL) was added 3- (5-bromoisoindolin-2-yl) piperidine-2, 6-dione (74 mg, 0.240 mmol) and Pd (dtbpf) Cl2 (16 mg, 0.0240 mmol) . Then the reaction mixture was stirred at 100℃ for 12 h under argon atmosphere. After completion of reaction, the mixture was diluted with H2O (20 mL) and extracted with EtOAc (20 mL x 3) . The combined organic layers was washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE/EA=5/1) to afford the title compound.
Step H: 3- (5- (1- (2-azaspiro [3.5] nonan-7-yl) -1H-pyrazol-4-yl) -1-oxoisoindolin-2-
yl) piperidine-2, 6-dione
To a solution of tert-butyl 7- (4- (2- (2, 6-dioxopiperidin-3-yl) -1-oxoisoindolin-5-yl) -1H-pyrazol-1-yl) -2-azaspiro [3.5] nonane-2-carboxylate (200 mg, 0.375 mmol) in DCM (2 mL) was added TFA (0.2 mL) . Then the reaction mixture was stirred at 30℃ for 2 hrs. After completion, the reaction mixture was concentrated by vacuum to afford the title compound, which was used in the next step without further purifications.
LC-MS (ESI) [M+H] + = 434.0
Step I: 3- (5- (1- (2- (1- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperidin-4-yl) -2-azaspiro [3.5] nonan-7-yl) -1H-pyrazol-4-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a solution of 3- (5- (1- (2-azaspiro [3.5] nonan-7-yl) -1H-pyrazol-4-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (50.0 mg) in DCM (1 mL) and MeOH (1 mL) was added (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- ( (4- (4-oxopiperidin-1-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (60.5 mg) , the reaction mixture was stirred at 25℃ for 30 min, NaBH (OAc) 3 (73.1 mg, 0.345 mmol) was added. Then the reaction mixture was stirred at 25℃ for 2 h. After completion of reaction, the reaction mixture was concentrated by vacuum to give the residue, which was added Η20 (10 mL) and extracted by EtOAc (10 mL x 3) . The combined organic phase was washed with brine, dried over anhydrous Na2S04. The organic layer was concentrated and purified by pre-HPLC to afford the title compound.
LC-MS (ESI) [M+H] + = 944.1
1H NMR (400 MHz, DMSO-d6) δ 11.34 –10.73 (m, 1H) , 10.09 (s, 1H) , 8.81 (s, 1H) , 8.42 (s, 1H) , 7.98 (s, 1H) , 7.94 (s, 2H) , 7.86 (d, J = 8.2 Hz, 1H) , 7.69 (d, J = 8.0 Hz, 1H) , 7.58 (d, J = 7.7 Hz, 3H) , 6.92 (d, J = 8.8 Hz, 2H) , 5.67 (d, J = 5.7 Hz, 1H) , 5.13 (d, J = 13.4 Hz, 1H) , 4.99 (d, J =10.0 Hz, 1H) , 4.85 (d, J = 17.8 Hz, 1H) , 4.75 (s, 1H) , 4.57 (s, 1H) , 4.45 (d, J = 16.7 Hz, 1H) , 4.31 (d, J = 17.2 Hz, 1H) , 4.13 (s, 1H) , 3.52 (s, 1H) , 3.01 (s, 2H) , 2.97 –2.95 (m, 1H) , 2.91 (s, 2H) , 2.85-2.55 (m, 8H) , 2.41-2.32 (m, 1H) , 2.24-2.15 (m, 1H) , 2.04-1.92 (m, 6H) , 1.91 –1.88 (m, 1H) , 1.87 –1.83 (m, 1H) , 1.82-1.66 (m, 6H) , 1.62-1.55 (m, 2H) , 131-1.21 (m, 2H) , 0.87 (t, J = 7.6 Hz, 3H) .
Example 30: 3- (5- (1- (1- (7- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) -7-azaspiro [3.5] nonan-2-yl) piperidin-4-yl) -1H-pyrazol-4-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (Cpd-030)
Cpd-030 was synthesized according to the following scheme:
Step A: 7- (4-nitrophenyl) -7-azaspiro [3.5] nonan-2-one
To a solution of (0.952 g, 6.85 mmol) in DMSO (20 mL) was added 1-fluoro-4-nitrobenzene (0.966 g, 6.85 mmol) , DIPEA (1.42 g, 10.3 mmol) . The mixture was stirred at 80℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture was quenched with water (20 mL) , extracted with EA (3*10 mL) . The combined organic layers was washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 261.0
Step B: 7- (4-aminophenyl) -7-azaspiro [3.5] nonan-2-one
To a solution of 7- (4-nitrophenyl) -7-azaspiro [3.5] nonan-2-one (772 mg, 2.97 mmol) in MeOH (10 mL) was added Pd/C (10 mg, w/w=10%) and the reaction was stirred at 35℃ under H2 atmosphere at 2 atm for 12 hrs. The reaction solution was filtered and concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 231.2
Step C: (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- ( (4-
(2-oxo-7-azaspiro [3.5] nonan-7-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3, 4-
d] pyrimidin-3-one
To a solution of 7- (4-aminophenyl) -7-azaspiro [3.5] nonan-2-one (100 mg) in DCM (2.0 mL) was added 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6-
(methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (174 mg, 0.434 mmol) . The mixture was stirred at r. t. for 2 h. After completion of reaction, the mixture was concentrated. The residue was diluted with water (5.0 mL) , extracted with EA (3*10 mL) . The organic layers were washed with water (3*5 mL) and brine (5 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography to afford the title compound.
LC-MS (ESI) [M+H] + 566.1
Step D: 3- (5- (1- (1- (7- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) -7-azaspiro [3.5] nonan-2-yl) piperidin-4-yl) -1H-pyrazol-4-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a solution of (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- ( (4- (2-oxo-7-azaspiro [3.5] nonan-7-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (50.0 mg, 0.0884 mmol) in DCM (1 mL) and MeOH (1 mL) was added 3- (1-oxo-5- (1- (piperidin-4-yl) -1H-pyrazol-4-yl) isoindolin-2-yl) piperidine-2, 6-dione (34.8 mg, 0.0884 mmol) . The reaction mixture was stirred at 25℃ for 30 min, NaBH (OAc) 3 (18.7mg, 0.084 mmol) was added. The reaction mixture was stirred at 25℃ for 2 h. After completion of reaction, the reaction mixture was concentrated by vacuum to give the residue, which was added Η20 (10 mL) and extracted by EtOAc (10 mL x 3) . The combined organic phase was washed with brine, dried over anhydrous Na2S04. The organic layer was concentrated and the residue was purified by pre-HPLC to afford the title compound.
LC-MS (ESI) [M+H] + = 943.3
1H NMR (400 MHz, DMSO) δ 10.97 (s, 1H) , 10.11 (s, 1H) , 8.82 (s, 1H) , 8.42 (s, 1H) , 7.98 (s, 1H) , 7.94 (s, 2H) , 7.88 –7.84 (m, 1H) , 7.69 (d, J = 8.1 Hz, 1H) , 7.58 (d, J = 8.0 Hz, 3H) , 6.92 (d, J = 9.0 Hz, 2H) , 5.67 (dq, J = 10.4, 5.9 Hz, 1H) , 5.14 (dd, J = 13.3, 5.1 Hz, 1H) , 5.00 (d, J =10.0 Hz, 1H) , 4.85 (d, J = 17.3 Hz, 1H) , 4.75 (s, 1H) , 4.57 (d, J = 9.5 Hz, 1H) , 4.45 (d, J = 17.2 Hz, 1H) , 4.32 (d, J = 17.3 Hz, 1H) , 4.13 (s, 1H) , 3.54 (d, J = 11.2 Hz, 2H) , 3.02 –2.89 (m, 6H) , 2.82 –2.77 (m, 1H) , 2.75 –2.66 (m, 3H) , 2.61 (d, J = 16.3 Hz, 2H) , 2.44 –2.31 (m, 2H) , 2.19 (dd, J = 13.3, 5.3 Hz, 2H) , 2.05 –1.91 (m, 7H) , 1.80 –1.68 (m, 5H) , 1.58 (t, J = 11.2 Hz, 2H) , 1.28 (d, J = 10.3 Hz, 2H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 31: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-031)
Cpd-031 was synthesized according to the following scheme:
Step A: tert-butyl 9- (methoxymethylene) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of (methoxymethyl) triphenylphosphonium (6.92 g, 15.0 mmol) in THF (30 mL) was added LDA (2 N) (7.49 mL, THF) at -78℃ under N2 atmosphere. The mixture was stirred at -78℃ for 1 h, then tert-butyl 9-oxo-3-azaspiro [5.5] undecane-3-carboxylate (2.00 g, 7.48 mmol) in THF (10 mL) was added. The mixture was stirred at 25℃ for 18 h. The mixture was quenched with NH4Cl (aq. 50 mL) , extracted with EA (100 mL x 2) . The combined organic layers was washed with brine (50 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE/EA=10/1) to afford the title compound.
Step B: tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9- (methoxymethylene) -3-azaspiro [5.5] undecane-3-carboxylate (200 mg, 0.677 mmol) in THF (2 mL) was added aq. HCl (1 N, 2 mL) . The mixture was stirred at 20℃ for 18 h. The mixture was diluted with H2O (20 mL) and extracted with EtOAc (20 mL x 2) . The combined organic layers was washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated to afford the title compound.
Step C: tert-butyl (R) -9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate
A mixture of tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (41.0 mg) , (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- ( (4- (piperazin-1-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (74.8 mg, 0.146 mmol) in DCE (1 mL) was stirred at 25℃ for 0.5 h. Then NaBH (OAc) 3 (62.0 mg, 0.292 mmol) was added. The mixture was stirred at 25℃ for 2 h. The mixture was concentrated and purified by flash silica gel column chromatography (DCM/MeOH=10/1) to afford the title compound.
LC-MS (ESI) [M+H] + 778.6.
Step D: (R) -6- ( (4- (4- ( (3-azaspiro [5.5] undecan-9-yl) methyl) piperazin-1-yl) phenyl) amino) -2-
allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-
pyrazolo [3, 4-d] pyrimidin-3-one
A mixture of tert-butyl (R) -9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate (40.0 mg, 0.051 mmol) , aq. HCl (1 N) (2 mL) in DCM (1 mL) was stirred at 25℃ for 8 h. The mixture was quenched with NaHCO3 (aq. 10 mL) , extracted with EA (20 mL x 2) . The combined organic layers was dried with Na2SO4, filtered and concentrated to afford the title compound. and used in the next step directly.
LC-MS (ESI) [M+H] + 678.5.
Step E: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -6- ( (4- (4- ( (3-azaspiro [5.5] undecan-9-yl) methyl) piperazin-1-yl) phenyl) amino) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (40.0 mg) in DMF (1 mL) were added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (31 mg, 0.071 mmol) and DIEA (23 mg, 0.180 mmol) . The mixture was stirred at 25℃ for 1 h. The mixture
was diluted with H2O (10 mL) and extracted with EtOAc (20 mL x 2) . The combined organic layers was washed with brine (20 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by Pre-HPLC (Waters 2767 /Qda, Column: Pursuit XRs 10 C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 26%~36%; Retention Time: 8.4-9.3min of 18 min) to afford the title compound.
LC-MS (ESI) [M+H] + 928.5.
1H NMR (400 MHz, DMSO) δ 10.51 (s, 1H) , 10.16 –10.02 (m, 1H) , 8.82 (s, 1H) , 8.28 (s, 1H) , 7.91 (s, 1H) , 7.69 (d, J = 8.1 Hz, 1H) , 7.63 (d, J = 8.1 Hz, 1H) , 7.55 (s, 3H) , 7.39 (d, J = 7.5 Hz, 1H) , 6.91 (d, J = 9.0 Hz, 2H) , 5.66 (s, 1H) , 4.99 (d, J = 10.2 Hz, 2H) , 4.85 (d, J = 16.8 Hz, 1H) , 4.78 –4.70 (m, 1H) , 4.59 (s, 1H) , 3.75 (s, 2H) , 3.60 (s, 4H) , 3.08 (s, 8H) , 2.98 –2.95 (m, 1H) , 2.74 (s, 2H) , 2.17 (s, 4H) , 2.01 (s, 1H) , 1.88 (s, 1H) , 1.69 (s, 4H) , 1.59 (s, 4H) , 1.36 (s, 1H) , 1.29 (s, 1H) , 1.09 (s, 4H) , 0.87 (t, J = 7.2 Hz, 3H) .
Example 32: 3- [5- [9- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] -2-chloro-phenyl] piperidine-2, 6-dione (Cpd-032)
Cpd-032 was synthesized according to the following scheme:
Step A: 3- (5-bromo-2-chloro-phenyl) piperidine-2, 6-dione
To a mixture of methyl 2- (5-bromo-2-chloro-phenyl) acetate (1 g, 3.79 mmol) and prop-2-enamide (323.67 mg, 4.55 mmol) in DMF (8 mL) was added t-BuOK (468.41 mg, 4.17 mmol) in one portion at 0 ℃ under N2. The mixture was stirred at 0 ℃ for 1 hour. aq. AcOH (5%, w/t) (30 mL) was added and the mixture was extracted with ethyl acetate (30 mL × 3) . The combined organic phases was washed with brine (30 mL × 3) , dried with anhydrous Na2SO4, filtered and concentrated in under reduce pressure. The crude product was triturated with (Petroleum ether/Ethyl acetate = 2/1) at 25 ℃ for 30 min to afford the title compound.
1H NMR (400 MHz, DMSO-d6) δ 10.93 (s, 1H) , 7.61 (d, J = 2.0 Hz, 1H) , 7.52 (dd, J = 8.4, 2.4 Hz, 1H) , 7.40-7.46 (m, 1H) , 4.22 (dd, J = 12.8, 4.8 Hz, 1H) , 2.70-2.83 (m, 1H) , 2.55-2.61 (m, 1H) , 2.28-2.42 (m, 1H) , 1.90-2.03 (m, 1H) .
Step B: (4-methoxyphenyl) methyl 4-chloro-3- (2, 6-dioxo-3-piperidyl) benzoate
To a mixture of 3- (5-bromo-2-chloro-phenyl) piperidine-2, 6-dione (640 mg, 2.12 mmol) and (4-methoxyphenyl) methanol (1.46 g, 10.58 mmol) in DMF (10 mL) was added Et3N (1.31 g, 12.90 mmol) and Pd (dppf) Cl2 (464.34 mg, 634.60 umol) . The mixture was stirred at 80 ℃ for 24 hours under CO (50 Psi) . H2O (100 mL) was added and the mixture was extracted with ethyl acetate (150 mL × 3) . The combined organic phases was washed with brine (200 mL × 3) , dried with anhydrous Na2SO4, filtered and concentrated in under reduced pressure. The residue was
purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=100/1 to 1/1) to afford the title compound.
1H NMR (ES22593-210-P1H2, 400 MHz, DMSO-d6) δ: 10.95 (s, 1H) , 7.92 (d, J = 2.0 Hz, 1H) , 7.87 (dd, J = 8.4, 2.0 Hz, 1H) , 7.62 (d, J = 8.4 Hz, 1H) , 7.41 (d, J = 8.8 Hz, 2H) , 6.95 (d, J = 8.8 Hz, 2H) , 5.28 (s, 2H) , 4.33 (dd, J = 12.8, 4.0 Hz, 1H) , 3.76 (s, 3H) , 2.70-2.84 (m, 1H) , 2.58-2.59 (m, 1H) , 2.55-2.57 (m, 1H) , 2.34-2.41 (m, 1H) , 1.99-2.03 (m, 1H) .
Step C: 4-chloro-3- (2, 6-dioxo-3-piperidyl) benzoic acid
The mixture of (4-methoxyphenyl) methyl 4-chloro-3- (2, 6-dioxo-3-piperidyl) benzoate (340 mg, 876.71 umol) and TFA (3 mL) was stirred at 25 ℃ for 16 h. The mixture was added EtOAc (10 mL) at 0 ℃. The crude product was triturated with EtOAc (10 mL) at 25 ℃ for 1 h to afford the title compound.
1H NMR (ES22593-214-P1H1, 400 MHz, DMSO-d6) δ 12.90-13.59 (m, 1H) , 10.95 (s, 1H) , 7.90 (d, J = 2.0 Hz, 1H) , 7.85 (dd, J = 8.4, 2.0 Hz, 1H) , 7.60 (d, J = 8.4 Hz, 1H) , 4.32 (dd, J = 12.8, 4.8 Hz, 1H) , 2.72-2.85 (m, 1H) , 2.55-2.57 (m, 1H) , 2.33-2.41 (m, 1H) , 1.97-2.05 (m, 1H) .
Step D: 3- [2-chloro-5- [9- (hydroxymethyl) -3-azaspiro [5.5] undecane-3-
carbonyl] phenyl] piperidine-2, 6-dione
To a mixture of 4-chloro-3- (2, 6-dioxo-3-piperidyl) benzoic acid (120 mg) , 3-azaspiro [5.5] undecan-9-ylmethanol (82.17 mg, 448.32 umol) in DCM (2 mL) were added HATU (255.70 mg, 672.48 umol) and DIPEA (173.83 mg, 1.34 mmol) . The mixture was stirred at 25 ℃ for 3 hrs. H2O (10 mL) was added and the mixture was extracted with DCM (10 mL ×3) . The combined organic phases was dried with anhydrous Na2SO4, filtered and concentrated in under reduced pressure. The residue was purified by column chromatography (SiO2, Petroleum ether: Ethyl acetate=100/1 to 10/1) to afford the title compound.
LCMS: MS (ESI) m/z = 433.2 [M+H] +.
Step E: 3- [4-chloro-3- (2, 6-dioxo-3-piperidyl) benzoyl] -3-azaspiro [5.5] undecane-9-
carbaldehyde
To a mixture of 3- [2-chloro-5- [9- (hydroxymethyl) -3-azaspiro [5.5] undecane-3-carbonyl] phenyl] piperidine-2, 6-dione (190 mg, 438.86 umol) in DCM (2 mL) and DMF (2 mL) was added Dess-Martin reagent (279.21 mg, 658.29 umol) . The mixture was stirred at 25 ℃ for 16 h. The reaction mixture was filtered, washed with DCM (10 mL) , concentrated under reduced pressure to give crude product. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol=100/1 to 10/1) to give the title compound.
LCMS: MS (ESI) m/z = 431.1 [M+H] +.
Step F: 3- [5- [9- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -3-
azaspiro [5.5] undecane-3-carbonyl] -2-chloro-phenyl] piperidine-2, 6-dione
To a mixture of 3- [4-chloro-3- (2, 6-dioxo-3-piperidyl) benzoyl] -3-azaspiro [5.5] undecane-9-carbaldehyde (50 mg, 116.03 umol) , 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (59.48 mg, 116.03 umol) in DCM (2 mL) were added TEA (58.70 mg, 580.15 umol) and NaBH (OAc) 3 (36.89 mg, , 174.04 umol) . The mixture was stirred at 25 ℃ for 16 hrs. H2O (10 mL) was added and the mixture was extracted with DCM (10 mL × 3) . The combined organic phases were dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 44%-74%, 8 min) to afford the title compound.
LCMS: MS (ESI) m/z = 927.3 [M+H] +.
1H NMR (ES22593-226-P1H1, 400 MHz, DMSO-d6) δ: 10.93 (s, 1H) , 10.12 (s, 1H) , 8.81 (s, 1H) , 7.92 (d, J = 7.6 Hz, 1H) , 7.69 (d, J = 8.0 Hz, 1H) , 7.57 (s, 2H) , 7.52 (d, J = 8.4 Hz, 1H) , 7.36 (d, J = 1.6 Hz, 1H) , 7.29-7.34 (m, 1H) , 6.91 (d, J = 8.8 Hz, 2H) , 5.61-5.73 (m, 1H) , 5.06 (s, 1H) , 4.99 (d, J = 10.0 Hz, 1H) , 4.85 (d, J = 17.6 Hz, 1H) , 4.74 (s, 1H) , 4.56 (d, J = 10.0 Hz, 1H) , 4.26 (dd, J = 12.4, 4.8 Hz, 1H) , 3.50-3.64 (m, 2H) , 3.22-3.29 (m, 2H) , 3.08 (s, 4H) , 2.91-3.01 (m, 1H) , 2.72-2.83 (m, 2H) , 2.54-2.58 (m, 1H) , 2.46 (s, 4H) , 2.30-2.36 (m, 1H) , 2.12-2.25 (m, 3H) , 1.96-2.06 (m, 2H) , 1.84-1.93 (m, 1H) , 1.64-1.76 (m, 3H) , 1.47-1.62 (m, 4H) , 1.23-1.44 (m, 3H) , 1.00-1.17 (m, 4H) , 0.86 (t, J = 7.6 Hz, 3H) .
Example 33: 1- [5- [9- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] -2-chloro-phenyl] hexahydropyrimidine-2, 4-dione (Cpd-033)
Cpd-033 was synthesized according to the following scheme:
Step A: tert-butyl 4- (4-nitrophenyl) -3, 6-dihydro-2H-pyridine-1-carboxylate
To a solution of tert-butyl 4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydro-2H-pyridine-1-carboxylate (3 g, 9.70 mmol) , 1-bromo-4-nitro-benzene (2.35 g, 11.64 mmol) , Pd(dppf) Cl2 (354.96 mg, 485.11 umol) , K2CO3 (4.02 g, 29.11 mmol) in dioxane (30 mL) and H2O (10 mL) was purged with N2 for 3 times. The mixture was stirred at 100 ℃ for 2 hrs under N2 atmosphere. The mixture was poured into water (100 mL) and extracted with EtOAc (200 mL × 2) . The combined organic layers was washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by column chromatography on silica gel (11%EtOAc in Petroleum ether) to afford the title compound.
1H NMR (400 MHz, CDCl3) δ: 8.20 (d, J = 8.8 Hz, 2H) , 7.52 (d, J = 8.8 Hz, 2H) , 6.24 (s, 1H) , 4.14 (d, J = 2.8 Hz, 2H) , 3.67 (t, J = 5.6 Hz, 2H) , 2.56 (d, J = 1.2 Hz, 2H) , 1.50 (s, 9H) .
Step B: tert-butyl 4- (4-aminophenyl) piperidine-1-carboxylate
To a solution of tert-butyl 4- (4-nitrophenyl) -3, 6-dihydro-2H-pyridine-1-carboxylate (500 mg, 1.64 mmol) in THF (10 mL) was added Pd/C (0.1 g, 10%wt loading, wet) , Pd (OH) 2/C (0.1 g, 5%wt loading, wet) under H2. The mixture was stirred at 25 ℃ for 16 hr under H2 (15 Psi) . The
reaction mixture was filtered, washed with EtOAc (200mL) and concentrated under reduced pressure to afford the title compound.
1H NMR (400 MHz, CDCl3) δ : 7.00 (d, J = 8.4 Hz, 2H) , 6.65 (d, J = 8.4 Hz, 2H) , 4.22 (s, 2H) , 3.04-3.87 (m, 2H) , 2.78 (t, J = 11.6 Hz, 2H) , 2.47–2.62 (m, 1H) , 1.78 (d, J = 13.2 Hz, 2H) , 1.51–1.63 (m, 2H) , 1.48 (s, 9H) .
Step C: 4- (4-piperidyl) aniline
To a solution of tert-butyl 4- (4-aminophenyl) piperidine-1-carboxylate (600 mg) in DCM (1 mL) was added TFA (1.54 g, 1 mL) . The mixture was stirred at 25 ℃ for 3 hrs. The mixture was concentrated to afford the title compound.
LCMS: MS (ESI) m/z = 177.2 [M+H] +.
Step D: tert-butyl 9- (methoxymethylene) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of methoxymethyl (triphenyl) phosphonium; chloride (15 g, 43.76 mmol) in THF (150 mL) was added t-BuOK (6.30 g, 56.10 mmol) . The mixture was stirred at 0℃ for 0.5 hr. tert-butyl 9-oxo-3-azaspiro [5.5] undecane-3-carboxylate (5 g, 18.70 mmol) was added and the mixture was stirred at 25℃ for 4 hrs. The reaction was quenched with saturated aq. sat. NH4Cl (50 mL) . The resulting aqueous residue was extracted with EtOAc (100 mL ×3) . The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated in vacuo. The residue was purified by flash silica gel chromatography (80 gSilica Flash Column, Eluent of 0~10%Ethyl acetate/Petroleum ether gradient @40 mL/min) to afford the title compound.
1H NMR (400 MHz, CDCl3) δ: 5.77 (s, 1H) , 3.50–3.57 (m, 3H) , 3.32–3.43 (m, 4H) , 2.19 (t, J = 6.0 Hz, 2H) , 1.93–2.00 (m, 2H) , 1.46 (s, 9H) , 1.38–1.45 (m, 8H) .
Step E: tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9- (methoxymethylene) -3-azaspiro [5.5] undecane-3-carboxylate (600 mg, 2.03 mmol) in MeCN (15 mL) was added water (0.5 mL) and TFA (0.25 mL) . The mixture was stirred at 25 ℃ for 6 hrs. The reaction was quenched with saturated aqueous NaHCO3 solution (4 mL) under ice cooling and organic solvent was removed in vacuo. The mixture was extracted with EtOAc (50 × 3 mL) . The organic layers was washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated in vacuo to afford the title compound.
1H NMR (400 MHz, CDCl3) δ: 9.64 (s, 1H) , 4.11 (q, J = 7.2 Hz, 1H) , 3.31-3.40 (m, 4H) , 2.19-2.29 (m, 1H) , 1.73-1.82 (m, 2H) , 1.62-1.70 (m, 2H) , 1.49-1.60 (m, 2H) , 1.44 (s, 9H) , 1.39-1.43 (m, 2H) , 1.36 (d, J = 6.0 Hz, 2H) , 1.21-1.26 (m, 2H) .
Step F: 3-azaspiro [5.5] undecane-9-carbaldehyde
To a solution of tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (150 mg) in DCM (2 mL) was added TFA (2.27 g, 1.47 mL) . The mixture was stirred at 25 ℃ for 2 hrs. The mixture was concentrated to afford the title compound.
Step G: 3- [4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoyl] -3-
azaspiro [5.5] undecane-9-carbaldehyde
To a solution of 3-azaspiro [5.5] undecane-9-carbaldehyde (150 mg) , 4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoic acid (222.30 mg, 827.48 umol) , DIEA (320.83 mg, 2.48 mmol, 432.38 uL) in DMF (3 mL) was added HATU (471.95 mg, 1.24 mmol) . The mixture was stirred at 25 ℃ for 2 hr. The mixture was poured into water (50 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers was washed with brine (50 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by column chromatography on silica gel (5%MeOH in Dichloromethane) to afford the title compound. LCMS: MS (ESI) m/z = 423.2 [M+H] +
Step H: 1- [5- [9- [ [4- (4-aminophenyl) -1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-
carbonyl] -2-chloro-phenyl] hexahydropyrimidine-2, 4-dione
A mixture of 3- [4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoyl] -3-azaspiro [5.5] undecane-9-carbaldehyde (310 mg, 717.74 umol) , 4- (4-piperidyl) aniline (189.76 mg) , TEA (363.14 mg, 3.59 mmol, 499.51 uL) in DCM (5 mL) was added NaBH (OAc) 3 (456.35 mg, 2.15 mmol) . The mixture was stirred at 25 ℃ for 16 hrs. The mixture was poured into water (20 mL) and extracted with EtOAc (20 mL × 2) . The combined organic layers was washed with brine (20 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by column chromatography on silica gel (6%MeOH in Dichloromethane) to afford the title compound.
LCMS: MS (ESI) m/z = 592.3 [M+H] +
Step I: 1- [5- [9- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] -1-piperidyl] methyl] -3-
azaspiro [5.5] undecane-3-carbonyl] -2-chloro-phenyl] hexahydropyrimidine-2, 4-dione
To a solution of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6-methylsulfanyl-pyrazolo [3, 4-d] pyrimidin-3-one (30 mg, 78.23 umol) in toluene (3 mL) was added m-CPBA (23.82 mg, 117.35 umol, 85%purity) and the mixture was stirred at 25℃ for 1hr. 1- [5- [9- [ [4- (4-aminophenyl) -1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] -2-chloro-phenyl] hexahydropyrimidine-2, 4-dione (46.33 mg, 78.23 umol) was added and the resulting mixture was stirred at 25℃ for 3 hrs. The mixture was poured into water (30 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers was washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by
prep-HPLC (column: C18-1 150*30mm*5um; mobile phase: [water (FA) -ACN] ; B%: 49%-89%, 9 min) to afford the title compound.
LCMS: MS (ESI) m/z = 927.5 [M+H] +
1H NMR (ES22540-202-P1B, 400 MHz, CD3OD) δ 8.82 (s, 1H) , 7.87 (d, J = 8.0 Hz, 1H) , 7.73 (d, J = 8.0 Hz, 1H) , 7.65 (d, J = 8.4 Hz, 1H) , 7.60 (d, J = 8.4 Hz, 2H) , 7.54 (s, 1H) , 7.43 (d, J = 8.0 Hz, 1H) , 7.19 (d, J = 8.4 Hz, 2H) , 5.64–5.82 (m, 1H) , 5.04 (d, J = 10.4 Hz, 1H) , 4.95 (s, 1H) , 4.91 (d, J = 4.4 Hz, 1H) , 4.69–4.77 (m, 1H) , 3.79 (t, J = 6.4 Hz, 2H) , 3.72 (s, 2H) , 3.44 (s, 2H) , 3.02–3.18 (m, 4H) , 2.84–2.92 (m, 3H) , 2.57 (s, 1H) , 2.33–2.43 (m, 3H) , 2.12–2.26 (m, 3H) , 1.98–2.05 (m, 1H) , 1.83 –1.87 (m, 7H) , 1.60–1.73 (m, 5H) , 1.44–1.57 (m, 2H) , 1.36 (s, 1 H) , 1.29 (s, 1H) , 1.15–1.26 (m, 4H) , 0.95 (t, J = 7.6 Hz, 3H) .
Example 34: 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [4- [ [3- [4-methoxy-3- (6-oxo-4, 5-dihydro-1H-pyridazin-3-yl) benzoyl] -3-azaspiro [5.5] undecan-9-yl] methyl] piperazin-1-yl] anilino] pyrazolo [3, 4-d] pyrimidin-3-one (Cpd-034)
Cpd-034 was synthesized according to the following scheme:
Step A: methyl 3-acetyl-4-methoxy-benzoate
To a solution of 3-acetyl-4-hydroxy-benzoic acid (5.0 g, 27.7 mmol) in acetone (50.0 mL) were added MeI (19.70 g, 138.7 mmol) and K2CO3 (7.67 g, 55.5 mmol) . The mixture was stirred at 25 ℃ for 15 hrs. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was diluted with water (20.0 mL) and extracted with ethyl acetate (15 mL × 3) . The combined organic layers was washed with brine (10.0 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (40 gSilica Flash Column, Eluent of 0~20%Ethyl acetate/Petroleum ether gradient @40 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 209.2 [M+H] +.
1H NMR (400 MHz, CDCl3) δ: 8.38 (d, J = 2.4 Hz, 1H) , 8.14 (dd, J = 2.4, 8.8 Hz, 1H) , 7.01 (d, J = 8.8 Hz, 1H) , 3.97 (s, 3H) , 3.89 (s, 3H) , 2.61 (s, 3H) .
Step B: methyl 3- (2-bromoacetyl) -4-methoxy-benzoate
To a solution of methyl 3-acetyl-4-methoxy-benzoate (0.7 g, 3.36 mmol) in DCM (15.0 mL) was added NBS (600 mg, 3.37 mmol) and 4-methylbenzenesulfonic acid hydrate (64 mg, 336.240 umol) . The mixture was stirred at 0-25 ℃ for 15 hrs. The reaction mixture was quenched by addition sat. NaHCO3 (10.0 mL) , the organic layer was separated. The aqueous layer was extracted with DCM (10 mL×3) . The combined organic layers was washed with brine (10.0 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (20 gSilica Flash Column, Eluent of 0~30%Ethyl acetate/Petroleum ether gradient @18 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 286.8 [M+H] +.
1H NMR (400 MHz, CDCl3) δ: 8.49 (d, J = 2.0 Hz, 1H) , 8.23 (dd, J = 1.6, 8.8 Hz, 1H) , 7.07 (d, J = 8.8 Hz, 1H) , 4.58 (s, 2H) , 4.04 (s, 3H) , 3.93 (s, 3H) .
Step C: methyl 3- [2- (2, 2-dimethyl-4, 6-dioxo-1, 3-dioxan-5-yl) acetyl] -4-methoxy-benzoate
To a solution of NaH (107 mg, 2.67 mmol, 60%purity) in THF (3.0 mL) was added a solution of 2, 2-dimethyl-1, 3-dioxane-4, 6-dione (350 mg, 2.43 mmol) in THF (2.0 mL) dropwise at 25℃ under nitrogen atmosphere. After addition, the resulting solution was stirred at this temperature for 1 hr., and then methyl 3- (2-bromoacetyl) -4-methoxy-benzoate (837 mg, 2.91 mmol) in THF (3.0 mL) was added dropwise at 25 ℃. The resulting mixture was stirred at 25 ℃ for 15 hrs. The reaction mixture was poured into water (5.0 mL) , acidified to pH 2-3 with 1 N hydrochloric acid and extracted with ethyl acetate (5.0 mL × 3) . The combined organic layers was washed with brine (10.0 mL) , dried over anhydrous Na2SO4, concentrated in vacuum. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~60%Ethyl acetate/Petroleum ether gradient @18 mL/min) to afford the title compound.
1H NMR (400 MHz, CDCl3) δ: 8.56 (d, J = 2.4 Hz, 1H) , 8.21 (dd, J = 2.4, 8.8 Hz, 1H) , 7.05 (d, J = 8.8 Hz, 1H) , 4.03 (s, 3H) , 3.95 -3.92 (m, 2H) , 3.90 -3.88 (m, 4H) , 1.88 (d, J = 7.6 Hz, 6H) .
Step D: ethyl 4-methoxy-3- (6-oxo-4, 5-dihydro-1H-pyridazin-3-yl) benzoate
To a solution of methyl 3- [2- (2, 2-dimethyl-4, 6-dioxo-1, 3-dioxan-5-yl) acetyl] -4-methoxy-benzoate (140 mg, 399.6 umol) in DMF (1.5 mL) was added NH2NH2. H2O (84 mg, 1.68 mmol) . The mixture was stirred at 25 ℃ for 72 hrs. The reaction mixture was poured into ice-water and acidified with aq. HCl (6 N) to pH 4~5. The reaction mixture was extracted with ethyl acetate (5 mL×3) . The combined organic layers was washed with brine (10.0 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 0~50%Ethyl acetate/Petroleum ether gradient @18 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 263.2 [M+H] +) .
1H NMR (400 MHz, CDCl3) δ: 8.14 (d, J = 2.0 Hz, 1H) , 8.10 (dd, J = 2.4, 8.8 Hz, 1H) , 8.03 (s, 1H) , 6.98 (d, J = 8.8 Hz, 1H) , 3.94 (s, 3H) , 3.90 (s, 3H) , 2.96 -2.93 (m, 2H) , 2.57 (dd, J = 7.2, 8.8 Hz, 2H) .
Step E: 4-methoxy-3- (6-oxo-4, 5-dihydro-1H-pyridazin-3-yl) benzoic acid
To a solution of methyl 4-methoxy-3- (6-oxo-4, 5-dihydro-1H-pyridazin-3-yl) benzoate (50 mg, 190.6 umol) in THF (1.0 mL) , MeOH (1.0 mL) and water (0.5 mL) was added NaOH (23 mg, 575.0 umol) . The mixture was stirred at 25 ℃ for 15 hrs. The reaction mixture was concentrated under reduced pressure to remove organic solvent. The residue was diluted with water (2.0 mL) , acidified to pH = 5~6 with 1 N HCl. and extracted with ethyl acetate (3 mL× 3) . The combined organic layers was washed with brine (5.0 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to afford the title compound., which was used in the next step without further purification.
LCMS: MS (ESI) m/z = 249.0 [M+H] +) .
Step F: 3- [5- [9- (hydroxymethyl) -3-azaspiro [5.5] undecane-3-carbonyl] -2-methoxy-phenyl] -
4, 5-dihydro-1H-pyridazin-6-one
To a solution of 4-methoxy-3- (6-oxo-4, 5-dihydro-1H-pyridazin-3-yl) benzoic acid (25 mg) and DIPEA (65 mg, 503.5 umol) in DMF (1.5 mL) were added HATU (57 mg, 151.0 umol) and 3-azaspiro [5.5] undecan-9-ylmethanol (33 mg, 151.0 umol) . The mixture was stirred at 25 ℃ for 3 hrs. The reaction mixture was diluted with water (5.0 mL) and extracted with ethyl acetate (5 mL×3) . The combined organic layers was washed with brine (10.0 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-TLC (SiO2, DCM: MeOH =10: 1) to afford the title compound.
LCMS: MS (ESI) m/z = 414.2 [M+H] +) .
Step G: 3- [4-methoxy-3- (6-oxo-4, 5-dihydro-1H-pyridazin-3-yl) benzoyl] -3-
azaspiro [5.5] undecane-9-carbaldehyde
To a solution of 3- [5- [9- (hydroxymethyl) -3-azaspiro [5.5] undecane-3-carbonyl] -2-methoxy-phenyl] -4, 5-dihydro-1H-pyridazin-6-one (35 mg, 84.64 umol) in DCM (3 mL) was added Dess-Martin reagent (54 mg, 126.9 umol) . The mixture was stirred at 25 ℃ for 1 hr. The reaction mixture was filtered and the filtrate was concentrated in vacuum. The residue was purified by prep-TLC (SiO2, DCM: MeOH =10: 1) to afford the title compound.
LCMS: MS (ESI) m/z = 412.5 [M+H] +.
Step H: 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [4-
[ [3- [4-methoxy-3- (6-oxo-4, 5-dihydro-1H-pyridazin-3-yl) benzoyl] -3-azaspiro [5.5] undecan-
9-yl] methyl] piperazin-1-yl] anilino] pyrazolo [3, 4-d] pyrimidin-3-one
To a solution of 3- [4-methoxy-3- (6-oxo-4, 5-dihydro-1H-pyridazin-3-yl) benzoyl] -3-azaspiro [5.5] undecane-9-carbaldehyde (20 mg, 48.6 umol) and 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (25 mg, 48.7 umol) in DCM (2.5 mL) was added TEA (25 mg, 243.0) . The reaction mixture was stirred at 25 ℃ for 1 hr. Then NaBH (OAc) 3 (16 mg, 72.9 umol) was added to the reaction mixture. After the addition was finished, the mixture was stirred at 25 ℃ for an additional 1 hr. The reaction mixture was concentrated in vacuum, diluted with DMF (1.0 mL) and water (0.2 mL) . The reaction mixture was purified by prep-HPLC (column: Phenomenex C18 75*30mm*3um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 42%-82%, 9min) to afford the title compound.
LCMS: MS (ESI) m/z = 908.4 [M+H] +.
1H NMR (400 MHz, CD3OD) δ: 8.78 (s, 1H) , 7.86 (d, J = 8.4 Hz, 1H) , 7.72 (d, J = 8.0 Hz, 1H) , 7.53 (d, J = 8.8 Hz, 2H) , 7.51 -7.45 (m, 2H) , 7.14 (d, J = 8.4 Hz, 1H) , 6.94 (d, J =8.8 Hz, 2H) , 5.81 -5.65 (m, 1H) , 5.06 -5.01 (m, 1H) , 4.94 (s, 1H) , 4.82 (s, 1H) , 4.76 -4.69 (m, 1H) , 3.93 (s, 3H) , 3.79 -3.62 (m, 2H) , 3.56 -3.39 (m, 2H) , 3.20 -3.12 (m, 4H) , 3.10 -3.01 (m, 1H) , 2.95 (t, J = 8.4 Hz, 2H) , 2.91 -2.81 (m, 1H) , 2.64 -2.55 (m, 4H) , 2.54 -2.47 (m, 2H) , 2.42 -2.33 (m, 1H) , 2.31 -2.21 (m, 2H) , 2.15 (ddd, J = 5.6, 8.4, 13.6 Hz, 1H) , 2.07 -1.97 (m, 1H) , 1.91 -1.74 (m, 3H) , 1.72 -1.49 (m, 5H) , 1.44 (s, 1H) , 1.34 (s, 1H) , 1.26 -1.05 (m, 4H) , 0.95 (t, J = 7.4 Hz, 3H) .
Example 35: 1- [2-chloro-5- [9- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-yl] amino] phenyl] piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] phenyl] hexahydropyrimidine-2, 4-dione (Cpd-035)
Cpd-035 was synthesized according to the following scheme:
Step A: (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one
To a solution of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfanyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (100 mg, 270.6 umol) in toluene (4.0 mL) was added m-CPBA (116.78 mg, 541.3 umol, 80%purity) in portions at 25 ℃. The mixture was stirred at this temperature for 1 hr. The solution of the title compound in toluene (4.0 mL) was used in the next step directly without further purification.
LCMS: MS (ESI) m/z = 402.0 [M+H] +.
Step B: tert-butyl 4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-
yl] amino] phenyl] piperazine-1-carboxylate
To a solution of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one in toluene (4.0 mL) (as described in step A) was added DIPEA (175 mg, 1.36 mmol) and tert-butyl 4- (4-aminophenyl) piperazine-1-carboxylate (98 mg, 352.9 umol) . The mixture was stirred at 25 ℃ for 15 hrs under nitrogen atmosphere. The reaction was diluted with brine (5.0 mL) and extracted with ethyl acetate (5.0 mL × 3) . The combined organic layers was dried over
anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by prep-TLC (SiO2, Petroleum ether/Ethyl acetate=1: 2) to give the title compound.
LCMS: MS (ESI) m/z = 599.2 [M+H] +.
Step C: (12Z, 16R) -16-hydroxy-16-methyl-5- (4-piperazin-1-ylanilino) -2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one
To a solution of tert-butyl 4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-yl] amino] phenyl] piperazine-1-carboxylate (90.00 mg, 150.3 umol) in DCM (2.0 mL) was added TFA (1.0 mL) . The mixture was stirred at 25 ℃ for 3 hrs. The reaction mixture was concentrated in vacuum afford the title compound., which was used in the next step directly without further purification.
LCMS: MS (ESI) m/z = 499.0 [M+H] +.
Step D: tert-butyl 9- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-
yl]amino] phenyl] piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of (12Z, 16R) -16-hydroxy-16-methyl-5- (4-piperazin-1-ylanilino) -2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (75 mg) and tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (64 mg, 225.6 umol) in DCM (6.0 mL) was added TEA (76 mg, 752.1 umol) . After the reaction mixture was stirred at 25 ℃ for 1 hr., NaBH (OAc) 3 (48 mg, 225.6 umol) was added to the reaction mixture. The mixture was stirred at 25 ℃ for additional 1 hr. The reaction mixture was quenched by addition saturated NaHCO3 (5.0 mL, aq. ) , and extracted with DCM (5 mL × 3) . The combined organic layers was washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 0~100%Ethyl acetate/Petroleum ether gradient @18 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 764.4 [M+H] +.
Step E: (12Z, 16R) -5- [4- [4- (3-azaspiro [5.5] undecan-9-ylmethyl) piperazin-1-yl] anilino] -16-
hydroxy-16-methyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-
1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one
To a solution of tert-butyl 9- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-yl] amino] phenyl] piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate (45 mg, 58.9 umol) in DCM (1.0 mL) was added TFA (0.5 mL) . The mixture was stirred at 25 ℃ for 1 hr.
The reaction mixture was concentrated in vacuum to afford the title compound., which was used in the nest step without further purification.
LCMS: MS (ESI) m/z =664.9 [M+H] +.
Step F: 1- [2-chloro-5- [9- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-
yl] amino] phenyl] piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-
carbonyl] phenyl] hexahydropyrimidine-2, 4-dione
To a solution of (12Z, 16R) -5- [4- [4- (3-azaspiro [5.5] undecan-9-ylmethyl) piperazin-1-yl] anilino] -16-hydroxy-16-methyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (40 mg) , DIPEA (39 mg, 301.297 umol) and 4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoic acid (21 mg, 78.3 umol) in DCM (2.0 mL) was added HATU (30 mg, 78.3 umol) . The mixture was stirred at 25 ℃ for 1 hr. The reaction mixture was concentrated in vacuum, the residue was diluted with DMF (2.0 mL) and water (0.2 mL) . The mixture was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 35%-55%, 16min) to afford the title compound.
LCMS: MS (ESI) m/z = 914.3 [M+H] +) .
1H NMR (400 MHz, CDCl3) δ 8.88 (s, 1H) , 8.16 (s, 1H) , 7.89 -7.84 (m, 1H) , 7.83 -7.78 (m, 1H) , 7.55 (d, J = 8.4 Hz, 1H) , 7.48 (d, J = 8.8 Hz, 2H) , 7.43 (s, 1H) , 7.38 (d, J = 8.0 Hz, 1H) , 7.25 (s, 1H) , 6.91 (d, J = 8.8 Hz, 2H) , 5.73 (s, 2H) , 4.49 (d, J = 12.0 Hz, 1H) , 4.31 (s, 1H) , 4.24 (s, 1H) , 3.88 -3.57 (m, 5H) , 3.47 -3.32 (m, 2H) , 3.30 -3.12 (m, 5H) , 3.06 -2.91 (m, 1H) , 2.89 -2.75 (m, 1H) , 2.66 -2.57 (m, 4H) , 2.32 -2.23 (m, 2H) , 2.21 -2.09 (m, 2H) , 2.08 -1.93 (m, 2H) , 1.81 -1.74 (m, 3H) , 1.46 (s, 3H) , 1.36 -1.23 (m, 2H) , 1.21 -0.99 (m, 5H) .
Example 36: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-036)
Cpd-036 was synthesized according to the following scheme:
Step A: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-
methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -6- ( (4- (4- ( (3-azaspiro [5.5] undecan-9-yl) methyl) piperazin-1-yl) phenyl) amino) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (40.0 mg, 0.0590 mmol) in DMF (1 mL) was added perfluorophenyl 3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) -4-methoxybenzoate (30.5 mg, 0.0708 mmol) , DIEA (22.9 mg, 0.177 mmol) . The mixture was stirred at 25℃ for 1 h. The mixture was diluted with H2O (10 mL) and extracted with EtOAc (20 mL x 2) . The combined organic layers was washed with brine (20 mL) , dried over Na2SO4, filtered, concentrated and purified by Pre-HPLC (Gilson GX-281) Column: Agilent C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 36-36%; Retention Time: 6.3-7.3min of 17min) to afford the title compound.
LC-MS (ESI) [M+H] + 924.7
1H NMR (400 MHz, DMSO) δ 10.33 (s, 1H) , 10.11 (s, 1H) , 8.82 (s, 1H) , 7.92 (s, 1H) , 7.69 (d, J = 8.1 Hz, 1H) , 7.57 (s, 2H) , 7.40 –7.28 (m, 2H) , 7.15 (d, J = 8.6 Hz, 1H) , 6.91 (d, J = 8.9 Hz, 2H) , 5.67 (dq, J = 10.2, 6.1 Hz, 1H) , 5.11 –4.95 (m, 2H) , 4.85 (d, J = 17.5 Hz, 1H) , 4.74 (s, 1H) , 4.58 (s, 1H) , 3.84 (s, 3H) , 3.60 (t, J = 6.6 Hz, 2H) , 3.44 (s, 4H) , 3.30 –3.27 (m, 4H) , 3.08 (s, 4H) , 2.98 (d, J = 6.7 Hz, 1H) , 2.80 –2.75 (m, 1H) , 2.70 –2.66 (m, 2H) , 2.19 (d, J = 12.1 Hz, 3H) , 2.02 (dd, J = 11.9, 7.0 Hz, 1H) , 1.91 –1.84 (m, 1H) , 1.75 –1.67 (m, 3H) , 1.60 –1.44 (m, 5H) , 1.31 (s, 2H) , 1.10 (d, J = 10.4 Hz, 4H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 37: 1- [5- [9- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-
yl]amino] phenyl] piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] -2-methoxy-phenyl] hexahydropyrimidine-2, 4-dione (Cpd-037)
Cpd-037 was synthesized according to the following scheme:
Step A: 1- [5- [9- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-
yl] amino] phenyl] piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] -2-methoxy-
phenyl] hexahydropyrimidine-2, 4-dione
To a solution of (12Z, 16R) -5- [4- [4- (3-azaspiro [5.5] undecan-9-ylmethyl) piperazin-1-yl] anilino] -16-hydroxy-16-methyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (40 mg, 60.2 umol) , DIPEA (39 mg, 301.2 umol) and 3- (2, 4-dioxohexahydropyrimidin-1-yl) -4-methoxy-benzoic acid (21 mg, 79.4 umol) in DCM (2.0 mL) was added HATU (30 mg, 78.3 umol) . The mixture was stirred at 25 ℃ for 1 hr. The reaction mixture was concentrated in vacuum, the residue was diluted with DMF (2.0 mL) and water (0.2 mL) . The residue was purified by prep-HPLC (column: Boston Uni C18 40*150*5um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 45%-65%, 9min) , to afford the title compound.
LCMS: MS (ESI) m/z = 910.4 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.88 (s, 1H) , 8.07 (s, 1H) , 7.90 -7.78 (m, 2H) , 7.52 -7.42 (m, 3H) , 7.37 (s, 1H) , 7.25 (s, 1H) , 7.01 (d, J = 8.4 Hz, 1H) , 6.91 (d, J = 8.8 Hz, 2H) , 5.92 -5.57 (m, 2H) , 4.50 (d, J = 8.8 Hz, 1H) , 4.41 -4.20 (m, 2H) , 3.90 (s, 3H) , 3.82 -3.60 (m, 3H) , 3.60 -3.35 (m, 3H) , 3.30 -3.11 (m, 5H) , 2.92 -2.78 (m, 2H) , 2.70 -2.52 (m, 5H) , 2.32 -2.23 (m, 2H) , 2.15 (d, J = 12.0 Hz, 2H) , 2.10 -1.93 (m, 2H) , 1.83 -1.78 (m, 1H) , 1.62 -1.49 (m, 4H) , 1.46 -1.25 (m, 3H) , 1.24 -0.98 (m, 5H) .
Example 38: (R) -1- (5- (2- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) -7-azaspiro [3.5] nonane-7-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-038)
Cpd-038 was synthesized according to the following scheme:
Step A: tert-butyl (R) -2- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) -7-azaspiro [3.5] nonane-7-carboxylate
A solution of (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- ( (4- (piperazin-1-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (55.4 mg, 0.108 mmol) , tert-butyl 2-formyl-7-azaspiro [3.5] nonane-7-carboxylate (41.0 mg, 0.162 mmol) in DCM (1 mL) and MeOH (1 mL) was stirred at 25℃ for 0.5 h. Then NaBH (OAc) 3 (68.7 mg, 0.324 mmol) was added. The mixture was stirred at 25℃ for 2 h. The mixture was concentrated and purified by flash silica gel column chromatography (DCM /MeOH 10 /1) to afford the title compound.
LC-MS (ESI) [M+H] + 750.6.
Step B: (R) -6- ( (4- (4- ( (7-azaspiro [3.5] nonan-2-yl) methyl) piperazin-1-yl) phenyl) amino) -2-
allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-
pyrazolo [3, 4-d] pyrimidin-3-one
A mixture of tert-butyl (R) -2- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) -7-azaspiro [3.5] nonane-7-carboxylate (40 mg, 0.0533 mmol) , HCl (aq. 1 N) (1 mL) in THF (1 mL) was stirred at 25℃ for 8 h. The mixture was quenched with NaHCO3 (10 mL) , extracted with EA (20 mL) . The combined organic layers was washed with brine (20 mL) , dried over Na2SO4, filtered and concentrated to afford the title compound, which was used in next step directly.
LC-MS (ESI) [M+H] + 650.5.
Step C: (R) -1- (5- (2- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) -7-azaspiro [3.5] nonane-7-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -6- ( (4- (4- ( (7-azaspiro [3.5] nonan-2-yl) methyl) piperazin-1-yl) phenyl) amino) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (39.0 mg) in DMF (1 mL) were added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (30.4 mg, 0.07 mmol) , DIEA (23.3 mg, 0.18 mmol) . The mixture was stirred at 25℃ for 1 h. The mixture was purified by Pre-HPLC (Waters 2767/Qda, Column: Pursuit XRs 10 C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 25%~35%; Retention Time: 7.5-8.6min of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 900.5
1H NMR (400 MHz, DMSO) δ 10.51 (s, 1H) , 10.11 (s, 1H) , 8.82 (s, 1H) , 8.31 (s, 1H) , 7.91 (s, 1H) , 7.69 (d, J = 8.2 Hz, 1H) , 7.63 (d, J = 8.2 Hz, 1H) , 7.55 (s, 3H) , 7.38 (d, J = 10.0 Hz, 1H) , 6.90 (d, J = 9.0 Hz, 2H) , 5.75 –5.61 (m, 1H) , 4.99 (d, J = 9.3 Hz, 2H) , 4.85 (d, J = 17.1 Hz, 1H) ,
4.73 (s, 1H) , 4.58 (s, 1H) , 3.74 (s, 2H) , 3.63 (s, 4H) , 3.07 (s, 4H) , 2.96 (d, J = 6.5 Hz, 1H) , 2.79 (s, 1H) , 2.74 (d, J = 4.5 Hz, 2H) , 2.41 (s, 3H) , 2.21 (dd, J = 11.9, 6.9 Hz, 2H) , 2.02 (dd, J = 18.9, 13.8 Hz, 4H) , 1.89 (dd, J = 13.9, 7.6 Hz, 1H) , 1.74 –1.42 (m, 9H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 39: (R) -1- (5- (9- (2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) ethyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-039)
Cpd-039 was synthesized according to the following scheme:
Step A: tert-butyl 9- (2-ethoxy-2-oxoethylidene) -3-azaspiro [5.5] undecane-3-carboxylateTo a solution of tert-butyl 9-oxo-3-azaspiro [5.5] undecane-3-carboxylate (2.00 g, 7.48 mmol) in DCM (10 mL) was added ethyl 2- (diethoxyphosphoryl) acetate (2.51 g, 11.2 mmol) and NaH
(60%, 0.600 g, 15.0 mmol) at 0℃. Then the reaction was stirred at 0℃ for 3 h under N2 atmosphere. After completion of reaction, the mixture was concentrated. The residue was diluted with water (20 mL) , extracted with EA (3*10 mL) . The organic layers were washed with water (10 mL) and brine (10 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column (DCM: MeOH =10: 1) to afford the title compound.
1HNMR: 1H NMR (400 MHz, CDCl3) δ 5.63 (s, 1H) , 4.19 (d, J = 7.1 Hz, 1H) , 4.16 –4.09 (m, 2H) , 3.45 –3.36 (m, 4H) , 2.85 (d, J = 6.4 Hz, 2H) , 2.21 (d, J = 6.6 Hz, 2H) , 1.54 (dd, J = 12.6, 5.9 Hz, 4H) , 1.48 (s, 1H) , 1.46 (s, 9H) , 1.35 (t, J = 7.1 Hz, 2H) , 1.27 (dd, J = 9.8, 4.3 Hz, 5H) .
Step B: tert-butyl 9- (2-ethoxy-2-oxoethyl) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9- (2-ethoxy-2-oxoethylidene) -3-azaspiro [5.5] undecane-3-carboxylate (220 mg, 0.652 mmol) in MeOH (10 mL) was added Pd/C (10.0 mg, w/w=10%) and stirred at 35℃ for 2 h under H2 atmosphere. The reaction was filtered and concentrated to obtain the title compound.
TLC: Rf =0.5 (PE: EA =3: 1) .
Step C: tert-butyl 9- (2-hydroxyethyl) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9- (2-ethoxy-2-oxoethyl) -3-azaspiro [5.5] undecane-3-carboxylate (1.30 g) in THF (10 mL) was added LiBH4 (0.334 g, 15.3 mmol) and the reaction was stirred at 0℃ for 2 h under N2 atmosphere. The mixture was quenched with water (20 mL) , extracted with EA (3*10 mL) . The combined organic layers was washed with brine (5.0 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
TLC: Rf =0.2 (DCM: MeOH =9: 1) .
Step D: tert-butyl 9- (2-oxoethyl) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9- (2-hydroxyethyl) -3-azaspiro [5.5] undecane-3-carboxylate (308 mg, 1.04 mmol) in DCM (5.0 mL) was added DMP (882 mg, 2.08 mmol) and the reaction mixture was stirred at 35℃ under N2 atmosphere for 2 hrs. After completion of reaction, the mixture was concentrated. The residue was diluted with water (20 mL) , extracted with DCM (3*20 mL) . The combined organic layers was dried with Na2SO4 and filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (PE: EA =3: 1) to afford the title compound.
TLC: Rf =0.4 (PE: EA =1: 1) .
Step E: tert-butyl (R) -9- (2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) ethyl) -3-azaspiro [5.5] undecane-3-carboxylate
A mixture of tert-butyl 9- (2-oxoethyl) -3-azaspiro [5.5] undecane-3-carboxylate (40.5 mg, 0.137 mmol) , (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- ( (4-
(piperazin-1-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (70.0 mg, 0.137 mmol) and NaBH (OAc) 3 (87.1 mg, 0.411 mmol) in DCM (2.0 mL) and MeOH (2.0 mL) and HOAc (0.1 mL) was stirred at 0℃ for 2 hrs. After completion. The reaction mixture was quenched with water (10 mL) , extracted with EA (3*10 mL) . The organic layers were washed with brine (5.0 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 792.6
Step F: (R) -6- ( (4- (4- (2- (3-azaspiro [5.5] undecan-9-yl) ethyl) piperazin-1-yl) phenyl) amino) -2-
allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-
pyrazolo [3, 4-d] pyrimidin-3-one
To a solution of tert-butyl (R) -9- (2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) ethyl) -3-azaspiro [5.5] undecane-3-carboxylate (70.0 mg, 0.0884 mmol) in THF (4 mL) was added 1M aq. HCl in (4.5 mL) . The mixture was stirred at 35℃ for 1 h. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with EtOAc (3*10 mL) , the combined organic layers was combined and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 692.4
Step G: (R) -1- (5- (9- (2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) ethyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -6- ( (4- (4- (2- (3-azaspiro [5.5] undecan-9-yl) ethyl) piperazin-1-yl) phenyl) amino) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (35.0 mg, 0.0506 mmol) in DMF (2.0 mL) were added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (22.0 mg, 0.0506 mmol) , DIEA (19.6 mg, 0.152 mmol) . The mixture was stirred at r. t. for 2 h. After completion of reaction, the mixture was concentrated. The residue was diluted with water (5.0 mL) , extracted with EA (3*10 mL) . The organic layers were washed with water (3*5 mL) and brine (5 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (Waters 2767/Qda) Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.05%TFA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 32-42%; Retention Time: 8.4-9.2min of 16 min) to afford) to afford the title compound.
LC-MS (ESI) [M+H] + 943.6.
1H NMR (400 MHz, DMSO) δ 10.49 (s, 1H) , 10.12 (s, 1H) , 9.37 (s, 1H) , 8.84 (s, 1H) , 7.90 (d, J = 8.1 Hz, 1H) , 7.69 (d, J = 8.1 Hz, 1H) , 7.64 (d, J = 8.2 Hz, 2H) , 7.54 (s, 1H) , 7.39 (d, J = 8.8 Hz, 1H) , 6.98 (t, J = 10.0 Hz, 2H) , 5.73 –5.57 (m, 1H) , 5.11 (t, J = 67.9 Hz, 2H) , 4.86 (d, J = 16.3 Hz, 1H) , 4.73 (s, 1H) , 4.58 (s, 1H) , 3.78 (d, J = 12.4 Hz, 2H) , 3.59 (d, J = 10.7 Hz, 4H) , 3.15 (d, J = 21.5 Hz, 4H) , 2.94 (t, J = 12.6 Hz, 3H) , 2.81 –2.75 (m, 2H) , 2.25 –2.17 (m, 1H) , 2.02 (dd, J = 16.7, 10.0 Hz, 2H) , 1.90 (dd, J = 13.8, 7.3 Hz, 1H) , 1.73 (d, J = 7.5 Hz, 3H) , 1.53 (t, J = 30.8 Hz, 6H) , 1.24 (s, 7H) , 1.13 (s, 2H) , 0.87 (t, J = 7.3 Hz, 3H) .
Example 40: (R) -1- (3- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-040)
Cpd-040 was synthesized according to the following scheme:
Step A: (R) -1- (3- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-
carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -6- ( (4- (4- ( (3-azaspiro [5.5] undecan-9-yl) methyl) piperazin-1-yl) phenyl) amino) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (40.0 mg, 0.0590 mmol) in DMF (1 mL) were
added perfluorophenyl 3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (28.3 mg, 0.0708. mmol) , DIEA (22.9 mg, 0.177 mmol) . The mixture was stirred at 25℃ for 1 h. The mixture was diluted with H2O (10 mL) and extracted with EtOAc (20 mL x 2) . The combined organic layers was washed with brine (20 mL) , , dried over Na2SO4, filtered, concentrated and purified by Pre-HPLC (Gilson GX-281) Column: Agilent C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 27-32%; Retention Time: 7.5-9.5min of 18min) to afford the title compound.
LC-MS (ESI) [M+H] + 894.7
1H NMR (400 MHz, DMSO) δ 10.42 (s, 1H) , 10.12 (s, 1H) , 8.82 (s, 1H) , 8.35 (s, 1H) , 7.92 (s, 1H) , 7.69 (d, J = 8.2 Hz, 1H) , 7.57 (s, 2H) , 7.45 (t, J = 7.6 Hz, 1H) , 7.39 (d, J = 8.0 Hz, 1H) , 7.35 (s, 1H) , 7.22 (d, J = 7.3 Hz, 1H) , 6.91 (d, J = 9.0 Hz, 2H) , 5.65 (dt, J = 10.7, 8.1 Hz, 1H) , 4.99 (d, J = 9.4 Hz, 1H) , 4.85 (d, J = 16.9 Hz, 1H) , 4.74 (s, 1H) , 4.59 (s, 1H) , 3.82 (t, J = 6.7 Hz, 2H) , 3.55 (s, 4H) , 3.32 –3.27 (m, 4H) , 3.08 (s, 4H) , 3.00 –2.94 (m, 1H) , 2.79 (d, J = 5.9 Hz, 1H) , 2.73 (s, 2H) , 2.19 (d, J = 13.9 Hz, 4H) , 2.02 (d, J = 5.4 Hz, 1H) , 1.89 (dd, J = 14.0, 7.0 Hz, 1H) , 1.73 –1.66 (m, 3H) , 1.55 (d, J = 30.0 Hz, 4H) , 1.43 (s, 1H) , 1.35 (s, 1H) , 1.27 (s, 1H) , 1.09 (s, 4H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 41: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-fluorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-041)
Cpd-041 was synthesized according to the following scheme:
Step A: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-
fluorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -6- ( (4- (4- ( (3-azaspiro [5.5] undecan-9-yl) methyl) piperazin-1-yl) phenyl) amino) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (40.0 mg, 0.0590 mmol) in DMF (1 mL) were added perfluorophenyl 3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) -4-fluorobenzoate (24.7 mg, 0.0590 mmol) , DIEA (22.9 mg, 0.177 mmol) . The mixture was stirred at 25℃ for 1 h. The mixture was diluted with H2O (10 mL) and extracted with EtOAc (20 mL x 3) . The combined organic layers was washed with brine (10 mL) , dried over Na2SO4, filtered, concentrated and purified by Pre-HPLC (Waters 2767/Qda) Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 24-34%; Retention Time: 7.8-8.8min of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 912.3
1H NMR (400 MHz, DMSO) δ 10.52 (s, 1H) , 10.08 (s, 1H) , 8.81 (s, 1H) , 7.91 (s, 1H) , 7.69 (d, J = 8.2 Hz, 1H) , 7.57 (s, 2H) , 7.49 (s, 1H) , 7.41 –7.27 (m, 2H) , 6.91 (d, J = 8.7 Hz, 2H) , 5.65 (dt, J = 11.3, 8.4 Hz, 1H) , 5.04 (s, 1H) , 4.99 (d, J = 10.0 Hz, 1H) , 4.86 (d, J = 17.9 Hz, 1H) , 4.73 (s, 1H) , 4.57 (s, 1H) , 3.75 (t, J = 6.6 Hz, 2H) , 3.58 (s, 2H) , 3.29 –3.23 (m, 2H) , 3.08 (s, 4H) , 2.96 (dd, J = 15.3, 7.0 Hz, 1H) , 2.85 –2.76 (m, 1H) , 2.73 (t, J = 6.6 Hz, 2H) , 2.48 –2.41 (m, 4H) , 2.19 (d, J = 13.9 Hz, 3H) , 2.07 –1.99 (m, 1H) , 1.89 (dd, J = 13.5, 6.7 Hz, 1H) , 1.70 (d, J = 6.6 Hz, 3H) , 1.56 (d, J = 33.0 Hz, 5H) , 1.37 –1.23 (m, 2H) , 1.09 (s, 4H) , 0.87 (t, J = 7.3 Hz, 3H) .
Example 42: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-methylphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-042)
Cpd-042 was synthesized according to the following scheme:
Step A: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta
[b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl)
piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-methylphenyl)
dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -6- ( (4- (4- ( (3-azaspiro [5.5] undecan-9-yl) methyl) piperazin-1-yl) phenyl) amino) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (40.0 mg, 0.0590 mmol) in DMF (1 mL) were added perfluorophenyl 3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) -4-methylbenzoate (31.0 mg, 0.0590 mmol) , DIEA (22.9 mg, 0.177 mmol) . The mixture was stirred at 25℃ for 1 h. The mixture was diluted with H2O (10 mL) and extracted with EtOAc (20 mL x 3) . The combined organic layers was washed with brine (10 mL) , dried over Na2SO4, filtered, concentrated and purified by Pre-HPLC (Waters 2767/Qda, Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 26%~36%; Retention Time: 7.1-8.2min of 16 min) to afford the title compound.
LC-MS (ESI) 908.4 [M+H] +
1H NMR (400 MHz, DMSO) δ 10.36 (s, 1H) , 10.10 (s, 1H) , 8.81 (s, 1H) , 8.41 (s, 1H) , 7.91 (s, 1H) , 7.69 (d, J = 8.1 Hz, 1H) , 7.57 (s, 2H) , 7.34 (d, J = 7.9 Hz, 1H) , 7.31 (s, 1H) , 7.24 (d, J = 7.7 Hz, 1H) , 6.91 (d, J = 8.9 Hz, 2H) , 5.67 (dq, J = 10.6, 5.8 Hz, 1H) , 5.04 (s, 1H) , 4.99 (d, J = 10.3 Hz, 1H) , 4.86 (d, J = 17.1 Hz, 1H) , 4.73 (s, 1H) , 4.57 (s, 1H) , 3.79 (s, 1H) , 3.64 –3.45 (m, 3H) , 3.29 –3.24 (m, 3H) , 3.08 (s, 4H) , 2.96 (dd, J = 14.1, 7.9 Hz, 1H) , 2.73 (ddd, J = 21.6, 15.2, 8.3 Hz, 3H) , 2.48 –2.41 (m, 4H) , 2.21 (s, 3H) , 2.17 (s, 2H) , 2.02 (dd, J = 11.5, 6.8 Hz, 1H) , 1.89 (dd, J = 13.7, 7.4 Hz, 1H) , 1.70 (d, J = 6.8 Hz, 3H) , 1.55 (d, J = 36.3 Hz, 5H) , 1.33 (s, 2H) , 1.09 (s, 4H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 43: (R) -1- (5- (7- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) -2-azaspiro [3.5] nonane-2-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-043)
Cpd-043 was synthesized according to the following scheme:
Step A: tert-butyl 7-oxo-2-azaspiro [3.5] nonane-2-carboxylate
To a solution of tert-butyl 7-hydroxy-2-azaspiro [3.5] nonane-2-carboxylate (1.00 g, 4.15 mmol) in 50 mL of dichloromethane was added Dess-Martin reagent (3.52 g, 8.30 mmol) , the reaction mixture was stirred at room temperature overnight. After completion of reaction, 150 mL of dichloromethane and 100 mL of water were added and the mixture was filtered on a pad of celite. The organic layer was separated, wash with 100 mL of saturated brine, dried with anhydrous Na2SO4 and filtered. The filtrate was concentrated by vacuum to give a residue, which was purified by flash silica gel column chromatography (PE/EA=10/1) to afford the title compound.
Step B: tert-butyl 7- (methoxymethylene) -2-azaspiro [3.5] nonane-2-carboxylate
To a solution of (methoxymethyl) triphenylphosphonium (1.51 g, 4.40 mmol) in THF (30 mL) was added LDA (2 N in THF) (2.20 mL, 4.40 mmol) at -78℃ under N2 atmosphere. The mixture was stirred at -78℃ for 1 h, then tert-butyl 7-oxo-2-azaspiro [3.5] nonane-2-carboxylate (700 mg, 2.93 mmol) in THF (10 mL) was added. The mixture stirred at 30℃ for 18 hrs. The mixture was quenched with NH4Cl (aq. 50 mL) , extracted with EA (100 mL x 2) . The combined
organic layers was washed with brine (50 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE/EA=10/1) to afford the title compound.
Step C: tert-butyl 7-formyl-2-azaspiro [3.5] nonane-2-carboxylate
To a solution of tert-butyl 7- (methoxymethylene) -2-azaspiro [3.5] nonane-2-carboxylate (300 mg, 1.12 mmol) in THF (10 mL) was added aq. HCl (1 N, 20 mL) . The mixture was stirred at 20℃for 8 hrs. The mixture was diluted with H2O (20 mL) and extracted with EtOAc (20 mL x 2) . The combined organic layers was washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated to afford the title compound. and used in the next step directly.
Step D: tert-butyl (R) -7- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) -2-azaspiro [3.5] nonane-2-carboxylate
A solution of (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- ( (4- (piperazin-1-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (55.4 mg, 0.108 mmol) , tert-butyl 7-formyl-2-azaspiro [3.5] nonane-2-carboxylate (41.0 mg) in DCM (1 mL) was stirred at 25℃ for 0.5 h. Then NaBH (OAc) 3 (68.7 mg, 0.324 mmol) was added. The mixture was stirred at 25℃ for 2 h. The mixture was concentrated and purified by flash silica gel column chromatography (DCM /MeOH 10 /1) to afford the title compound.
LC-MS (ESI) [M+H] + 750.5.
Step E: (R) -6- ( (4- (4- ( (2-azaspiro [3.5] nonan-7-yl) methyl) piperazin-1-yl) phenyl) amino) -2-
allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-
pyrazolo [3, 4-d] pyrimidin-3-one
A mixture of tert-butyl (R) -7- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) -2-azaspiro [3.5] nonane-2-carboxylate (40.0 mg, 0.0533 mmol) , aq. HCl (1 N) (1 mL) in THF (1 mL) was stirred at 25℃ for 8 h. The mixture was quenched with NaHCO3 (10 mL) , extracted with EA (20 mL) . The combined organic layers was washed with brine (20 mL) , dried over Na2SO4, filtered and concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 650.5.
Step F: (R) -1- (5- (7- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) -2-azaspiro [3.5] nonane-2-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -6- ( (4- (4- ( (2-azaspiro [3.5] nonan-7-yl) methyl) piperazin-1-yl) phenyl) amino) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (39.0 mg) in DMF (1 mL) were added Perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (30.4 mg, 0.07 mmol) and DIEA (23.3 mg, 0.180 mmol) . The mixture was stirred at 25℃ for 1 h. The mixture was purified by Pre-HPLC (Waters 2767/Qda, Column: XBridge C18 19*250mm, 10 um; Mobile Phase A: 10mmol NH4HCO3/H2O, B: ACN; flow rate: 20ml/min; gradient: 40%~40%; Retention Time: 8.6-10.2min of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 900.6
1H NMR (400 MHz, DMSO) δ 10.52 (d, J = 3.8 Hz, 1H) , 10.12 (s, 1H) , 8.82 (s, 1H) , 7.92 (s, 1H) , 7.78 (s, 1H) , 7.69 (d, J = 8.1 Hz, 1H) , 7.65 (d, J = 3.7 Hz, 2H) , 7.58 (s, 2H) , 6.91 (d, J = 7.8 Hz, 2H) , 5.66 (ddd, J = 16.7, 10.9, 5.7 Hz, 1H) , 5.10 –4.96 (m, 2H) , 4.85 (d, J = 16.6 Hz, 1H) , 4.75 (s, 1H) , 4.58 (s, 1H) , 3.99 (d, J = 18.4 Hz, 2H) , 3.77 (s, 1H) , 3.74 (s, 1H) , 3.69 (s, 1H) , 3.62 (s, 1H) , 3.08 (s, 4H) , 3.00 –2.93 (m, 1H) , 2.76 (d, J = 5.6 Hz, 3H) , 2.47 (s, 4H) , 2.27 –2.07 (m, 4H) , 2.02 (d, J = 5.0 Hz, 1H) , 1.93 –1.84 (m, 3H) , 1.74 –1.64 (m, 3H) , 1.45 (s, 4H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 44: (R) -1- (5- (9- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-044)
Cpd-044 was synthesized according to the following scheme:
Step A: tert-butyl 9- (4- (4-nitrophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecane-3-
carboxylate
A solution of 1- (4-nitrophenyl) piperazine (100 mg, 0.483 mmol) , tert-butyl 9-oxo-3-azaspiro [5.5] undecane-3-carboxylate (258 mg 0.966 mmol) , NaBH (OAc) 3 (204 mg 0.966 mmol) , TEA (49.0 mg 0.483 mmol) and HOAc (58.0 mg 0.483 mmol) in DCM (5.0 mL) was stirred at 35℃ for 12 hrs. The mixture was quenched with water (10 mL) , extracted with EtOAc (3*10 mL) . The combined organic layers was washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
LC-MS (ESI) [M+H] +. 459.3
Step B: 9- (4- (4-nitrophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecane
To a solution of tert-butyl 9- (4- (4-nitrophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecane-3-carboxylate (121 mg, 0.264 mmol) in DCM (5.0 mL) was added HCl in dioxane (3.0 mL) and
the mixture was stirred at 25℃ for 1 h. The mixture was concentrated. The residue was concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 359.2
Step C: 1- (2-chloro-5- (9- (4- (4-nitrophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecane-3-
carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 9- (4- (4-nitrophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecane (168 mg) in DMF (5.0 mL) were added 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoic acid (120 mg, 0.469 mmol) , DIEA (173 mg, 1.41 mmol) , HATU (170 mg 0.469 mmol) . The mixture was stirred at r. t. for 2 h. After completion, the reaction mixture was quenched with water (10 mL) , extracted with EA (3*20 mL) . The combined organic layers was washed with brine (10 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 609.2
Step D: 1- (5- (9- (4- (4-aminophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (9- (4- (4-nitrophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (187 mg, 0.308 mmol) in EtOH (10 mL) and H2O (3.0 mL) was added NH4Cl (82.0 mg, 1.54 mmol) , Fe (84.0 mg 1.54 mmol) . The mixture was stirred at 90℃ for 2 h. After completion of reaction, After completion of reaction, the mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was added Η20 (50 mL) and extracted by EtOAc (30 mL x 3) . The combined organic phase was washed with brine, dried over anhydrous Na2S04. The organic layer was concentrated to obtain the title compound.
LC-MS (ESI) [M+H] + 579.2
Step E: (R) -1- (5- (9- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
A solution of (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylthio) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (100 mg, 0.261 mmol) in DCM (10 mL) was added m-CPBA (58.0 mg, 0.287 mmol) was stirred at 0℃ for 1 h. 1- (5- (9- (4- (4-aminophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (151 mg) and DIPEA (101 mg, 0.783 mmol) were added. The mixture was stirred at 25℃ for 12 hrs. After completion, the reaction mixture was quenched with water (50 mL) , the resulting solution was extracted with DCM (2*100 mL) , the organic
layers was combined and concentrated to give the crude. The crude was purified by Pre-HPLC (Gilson-GX-281, Column: Pursuit XRs 10 C18 250*21.2mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 26%~36%; Retention Time: 6.3-7.4min of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 914.3.
1H NMR (400 MHz, DMSO) δ 10.50 (s, 1H) , 10.10 (s, 1H) , 8.81 (s, 1H) , 8.18 (s, 1H) , 7.91 (s, 1H) , 7.69 (d, J = 8.2 Hz, 1H) , 7.63 (s, 1H) , 7.55 (s, 3H) , 7.39 (d, J = 8.4 Hz, 1H) , 6.90 (d, J = 8.6 Hz, 2H) , 5.67 (dd, J = 16.8, 10.5 Hz, 1H) , 4.99 (d, J = 10.2 Hz, 2H) , 4.85 (d, J = 17.4 Hz, 1H) , 4.73 (s, 1H) , 4.57 (s, 1H) , 3.74 (s, 1H) , 3.61 (d, J = 16.7 Hz, 3H) , 3.07 (s, 4H) , 2.97 (s, 2H) , 2.75 (d, J = 3.6 Hz, 4H) , 2.64 (s, 4H) , 2.21 (dd, J = 12.6, 7.1 Hz, 3H) , 2.01 (s, 1H) , 1.89 (dd, J = 13.7, 7.1 Hz, 1H) , 1.72 (dd, J = 40.0, 20.8 Hz, 6H) , 1.36 (s, 7H) , 1.13 (s, 2H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 45: (R) -1- (5- (7- ( (1- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-2-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-045)
Cpd-045 was synthesized according to the following scheme:
Step A: tert-butyl 7- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-2-
carboxylate
To a solution of 1- (4-nitrophenyl) piperidine-4-carbaldehyde (200 mg, 0.854 mmol) in DCM (5 mL) and MeOH (5 mL) were added tert-butyl 2, 7-diazaspiro [3.5] nonane-2-carboxylate (193 mg, 0.854 mmol) , NaBH (OAC) 3 (199 mg, 0.939 mmol) , the mixture was stirred at 25℃ for 12 h under N2 atmosphere. The mixture was quenched by addition of 10 mL water, the resulting solution was extracted with DCM (3 *10 mL) , the organic layers were combined and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1-1: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 445.2
Step B: 7- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane
To a solution of tert-butyl 7- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-2-carboxylate (110 mg, 0.247 mmol) in DCM (10 mL) was added HCl/dioxane (3 mL, 1 M) , the mixture was stirred at 25℃ for 2 h under N2 atmosphere. The mixture was concentrated under reduced pressure to afford the title compound, which was used direct without further purification.
LC-MS (ESI) [M+H] + 345.3
Step C: 1- (2-chloro-5- (7- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 7-
diazaspiro [3.5] nonane-2-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 7- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane (80.0 mg) in DMF (5 mL) was added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (101 mg, 0.232 mmol) and DIPEA (224 mg, 1.74 mmol) , the mixture was stirred at 25℃ for 2 hrs under N2 atmosphere. The mixture was quenched by addition of 20 mL water. The resulting solution was extracted with EtOAc (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1-1: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 595.3
Step D : 1- (5- (7- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-2-
carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (7- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-2-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (120 mg, 0.202 mmol) in EtOH (10 mL) and H2O (1 mL) were added Fe (56.4 mg, 1.01 mmol) and NH4Cl (54.0 mg, 1.01 mmol) , the mixture was stirred at 90℃ for 3 hrs under N2 atmosphere. The mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was added Η20 (50 mL) and extracted by EtOAc (30 mL x 3) . The combined organic phase was washed with brine, dried over anhydrous Na2S04 and the organic layer was concentrated. under reduced pressure to afford the title compound.
LC-MS (ESI) [M+H] + 565.3
Step E: (R) -1- (5- (7- ( (1- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-2-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (5- (7- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-2-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (100 mg) in DCM (10 mL) were added 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (70.7 mg, 0.177 mmol) and DIPEA (68.5 mg, 0.531 mmol) , the mixture was stirred at 20℃ for 18 hrs under N2 atmosphere. The mixture was quenched by addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was prep-HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%TFA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 22-32%Retention Time: 7.3-8.5of 16 min) to afford the title compound.
LC-MS: (ESI) [M+H] + 900.5
1H NMR (400 MHz, MeOD) δ 8.86 (s, 1H) , 7.88 (d, J = 8.1 Hz, 1H) , 7.79 (d, J = 16.2 Hz, 3H) , 7.71 (d, J = 8.1 Hz, 1H) , 7.68 (s, 2H) , 7.39 (s, 2H) , 5.79 –5.64 (m, 1H) , 5.04 (dd, J = 10.3, 1.2 Hz, 1H) , 4.93 (dd, J = 17.1, 1.3 Hz, 1H) , 4.81 (d, J = 5.9 Hz, 1H) , 4.73 (dd, J = 15.9, 6.1 Hz, 1H) , 4.23 (d, J = 28.4 Hz, 2H) , 4.00 (d, J = 33.5 Hz, 2H) , 3.79 (t, J = 6.8 Hz, 2H) , 3.73 (d, J = 12.0 Hz, 2H) , 3.62 (s, 2H) , 3.42 (d, J = 46.5 Hz, 1H) , 3.20 –2.98 (m, 5H) , 2.93 –2.80 (m, 3H) , 2.38 (ddd, J = 13.9, 8.6, 5.4 Hz, 1H) , 2.25 (s, 2H) , 2.20 –1.94 (m, 7H) , 1.86 (dq, J = 15.0, 7.5 Hz, 1H) , 1.73 (s, 2H) , 1.29 (s, 2H) , 0.95 (t, J = 7.5 Hz, 3H) .
Example 46: (R) -1- (5- (4- ( (8- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) -2, 8-diazaspiro [4.5] decan-2-yl) methyl) piperidine-1-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-046)
Cpd-046 was synthesized according to the following scheme:
Step A: tert-butyl 8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decane-2-carboxylate
To a solution of 1-fluoro-4-nitrobenzene (588 mg, 4.17 mmol) in DMF (15 mL) was added tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (1.00 g, 4.17 mmol) , then the reaction mixture was stirred at 80℃ for 16 hrs. After completion, the reaction mixture was quenched with H2O (10 mL) and extracted with EA (3 × 20 mL) . The combined organic layers was washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =0: 1) to afford the title compound.
LC-MS: (ESI) [M+H] + 362.2
Step B: 8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decane
To a solution of tert-butyl 8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decane-2-carboxylate (1.40 g, 3.88 mmol) in DCM (20 mL) was added HCl (10 mL, 4 M in dioxane) , then the reaction mixture was stirred at 30℃ for 2 h under N2 atmosphere. After completion, the reaction mixture was concentrated to afford the title compound., which was used in next steps without further purification.
LC-MS (ESI) [M+H] + 262.1
Step C: tert-butyl 4- ( (8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decan-2-yl) methyl) piperidine-1-
carboxylate
To a solution of 8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decane (500 mg) in DCM/MeOH (15 mL/15ml) was added tert-butyl 4-formylpiperidine-1-carboxylate (409 mg, 1.92 mmol) , then the reaction mixture was stirred at 30℃ for 30 min, NaBH3CN (181 mg, 2.88 mmol) and AcOH (1.0 mL) were added, then the mixture was stirred at 30℃ for 16 hrs under N2 atmosphere. After completion, the reaction mixture was quenched with H2O (10 mL) and extracted with EA (3 ×20 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =0: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 459.3
Step D: 8- (4-nitrophenyl) -2- (piperidin-4-ylmethyl) -2, 8-diazaspiro [4.5] decane
To a solution of tert-butyl 4- ( (8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decan-2-yl) methyl) piperidine-1-carboxylate (400 mg, 0.873 mmol) in DCM (10 mL) was added HCl (5 mL, 4 M in dioxane) , then the reaction mixture was stirred at 30℃ for 2 hrs under N2 atmosphere. After completion of reaction, the reaction mixture was concentrated to afford the title compound, which was used in next steps without further purification.
LC-MS (ESI) [M+H] + 359.2
Step E: 1- (2-chloro-5- (4- ( (8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decan-2-
yl) methyl) piperidine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 8- (4-nitrophenyl) -2- (piperidin-4-ylmethyl) -2, 8-diazaspiro [4.5] decane (200 mg) in DMF (20 mL) were added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl)benzoate (243 mg, 0.559 mmol) and DIPEA (217 mg, 1.68 mmol) , then the reaction mixture was stirred at 25℃ for 2 h under N2 atmosphere. After completion, the reaction mixture was quenched with H2O (10 mL) and extracted with EA (3 × 20 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =0: 1) to afford the title compound. LC-MS (ESI) [M+H] + 609.3
Step F: 1- (5- (4- ( (8- (4-aminophenyl) -2, 8-diazaspiro [4.5] decan-2-yl) methyl) piperidine-1-
carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (4- ( (8- (4-nitrophenyl) -2, 8-diazaspiro [4.5] decan-2-yl) methyl) piperidine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (250 mg, 0.411 mmol) in EtOH/H2O (10 mL/1 mL) were added Fe (115 mg, 2.06 mmol) and NH4Cl (110 mg, 2.06 mmol) , then the reaction mixture was stirred at 90℃ for 2 h under N2 atmosphere. The mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was added Η20 (30 mL) and extracted by EtOAc (20 mL x 3) . The combined organic phase was washed with brine, dried over anhydrous Na2S04 and the organic layer was concentrated to give a residue, which was used in next steps without further purification.
LC-MS (ESI) [M+H] + 579.3
Step G: (R) -1- (5- (4- ( (8- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) -2, 8-diazaspiro [4.5] decan-2-yl) methyl) piperidine-1-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (5- (4- ( (8- (4-aminophenyl) -2, 8-diazaspiro [4.5] decan-2-yl) methyl) piperidine-1-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (200 mg) in DCM (10 mL) was added 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (138 mg, 0.345 mmol) and DIPEA (134 mg, 1.04 mmol) , the mixture was stirred at 25℃ for 18 hrs under N2 atmosphere. The mixture was quenched with 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 17-27%Retention Time: 9-19 of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 914.5
1H NMR (400 MHz, DMSO) δ 10.51 (s, 1H) , 10.11 (s, 1H) , 8.81 (s, 1H) , 8.35 (s, 1H) , 7.92 (s, 1H) , 7.69 (d, J = 8.1 Hz, 1H) , 7.64 (d, J = 8.2 Hz, 1H) , 7.55 (d, J = 1.9 Hz, 3H) , 7.38 (dd, J = 8.2, 2.0 Hz, 1H) , 6.92 (d, J = 8.8 Hz, 2H) , 5.73 –5.59 (m, 1H) , 5.05 (s, 1H) , 4.99 (d, J = 9.4 Hz, 1H) , 4.85 (d, J = 17.8 Hz, 1H) , 4.75 (s, 1H) , 4.57 (s, 1H) , 4.45 (s, 1H) , 3.75 (s, 1H) , 3.62 (d, J = 6.1 Hz, 3H) , 3.07 (s, 5H) , 2.99 –2.94 (m, 1H) , 2.75 (dd, J = 16.6, 12.3 Hz, 4H) , 2.34 (s, 2H) , 2.22 (dd, J = 20.5, 6.3 Hz, 4H) , 2.02 (dd, J = 11.8, 6.8 Hz, 1H) , 1.92 –1.86 (m, 1H) , 1.70 (dd, J = 13.8, 7.3 Hz, 4H) , 1.59 (d, J = 7.1 Hz, 6H) , 1.08 (d, J = 14.6 Hz, 2H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 47: (R) -1- (5- (9- ( (1- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6 yl) amino) phenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3 carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-047)
Cpd-047 was synthesized according to the following scheme:
Step A: (1- (4-nitrophenyl) piperidin-4-yl) methanol
To a solution of 1-fluoro-4-nitrobenzene (5.00 g, 35.4 mmol) in DMSO (20 mL) were added piperidin-4-ylmethanol (4.08 g, 35.4 mmol) , DIEA (9.15 g, 70.8 mmol) , the mixture was stirred at 80℃ for 3 h under N2 atmosphere. The mixture was quenched with 10 mL water, the reaction mixture was filtered through celite, and the filter cake was washed with water (20 mL) . The combined filtrate was dried under reduced pressure to afford the title compound.
Step B: 1- (4-nitrophenyl) piperidine-4-carbaldehyde
To a solution of (1- (4-nitrophenyl) piperidin-4-yl) methanol (4.00 g) in EA (60 mL) was added IBX (14.3 g, 51.0 mmol) , the mixture was stirred at 80℃ for 3 h under N2 atmosphere. The reaction mixture was filtered through celite, and the filtrate was dried under reduced pressure to afford the title compound.
LC-MS (ESI) [M+H] + 235.3
Step C: tert-butyl 9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-
3-carboxylate
To a solution of 1- (4-nitrophenyl) piperidine-4-carbaldehyde (500 mg) in DCM (5 mL) and MeOH (5 mL) were added tert-butyl 3, 9-diazaspiro [5.5] undecane-3-carboxylate (543 mg, 2.13 mmol) and NaBH (OAC) 3 (1.42 g, 6.39 mmol) , the mixture was stirred at 25℃ for 12 hrs under N2 atmosphere. The mixture was quenched with 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers was combined and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1-1: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 473.3
Step D: 3- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane
To a solution of tert-butyl 9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carboxylate (880 mg, 1.86 mmol) in DCM (10 mL) was added HCl/dioxane (3 mL, 1 M) , the mixture was stirred at 25℃ for 2 h under N2 atmosphere. The mixture was dried under reduced pressure to afford the title compound, which was used in the next step directly.
LC-MS (ESI) [M+H] + 373.2
Step E: 1- (2-chloro-5- (9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-
diazaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 3- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane (220 mg) in DMF (5 mL) were added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (308 mg, 0.709 mmol) and DIEA (229 mg, 1.77 mmol) , the mixture was stirred at 25℃ for 2 hrs under N2 atmosphere. The mixture was quenched with 20 mL water. The resulting solution was extracted with EtOAc (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1-1: 1) to afford the title compound.
1H NMR (400 MHz, DMSO) δ 10.50 (s, 1H) , 8.02 (d, J = 9.4 Hz, 2H) , 7.63 (d, J = 8.2 Hz, 1H) , 7.54 (d, J = 1.8 Hz, 1H) , 7.39 (dd, J = 8.2, 1.9 Hz, 1H) , 6.99 (d, J = 9.5 Hz, 2H) , 4.01 (d, J = 7.0 Hz, 2H) , 3.74 (s, 1H) , 3.61 (d, J = 18.6 Hz, 3H) , 2.97 (t, J = 11.9 Hz, 2H) , 2.74 (dd, J = 10.3, 4.2 Hz, 2H) , 2.40 (s, 4H) , 2.21 (s, 2H) , 1.85 (s, 1H) , 1.78 (d, J = 13.4 Hz, 2H) , 1.45 (d, J = 42.9 Hz, 9H) , 1.23 (s, 1H) , 1.11 (d, J = 11.1 Hz, 2H) .
Step F: 1- (5- (9- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-
carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (210 mg, 0.337 mmol) in EtOH (10 mL) and H2O (1 mL) was added Fe (94.1 mg, 1.68 mmol) and NH4Cl (90.1 mg, 1.68 mmol) , the mixture was stirred at 90℃ for 3 hrs under N2 atmosphere. The
mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was added Η20 (30 mL) and extracted by EtOAc (20 mL x 3) . The combined organic phase was washed with brine, dried over anhydrous Na2S04, and the organic layer was concentrated under reduced pressure to afford the title compound.
LC-MS (ESI) [M+H] + 593.2
Step G: (R) -1- (5- (9- ( (1- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6
yl) amino) phenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3 carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (5- (9- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (100 mg) in DCM (10 mL) were added 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (67.3 mg, 0.169 mmol) and DIPEA (65.4 mg, 0.507 mmol) , the mixture was stirred at 25℃ for 18 h under N2 atmosphere. The mixture was quenched with 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 17-27%Retention Time: 9-19 of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 928.3
1HNMR (400 MHz, DMSO) δ 10.51 (s, 1H) , 10.10 (s, 1H) , 8.81 (s, 1H) , 8.21 (s, 1H) , 7.92 (d, J = 7.8 Hz, 1H) , 7.69 (d, J = 8.1 Hz, 1H) , 7.64 (d, J = 8.2 Hz, 1H) , 7.55 (d, J = 1.9 Hz, 2H) , 7.39 (dd, J = 8.2, 1.9 Hz, 1H) , 6.90 (d, J = 8.9 Hz, 2H) , 5.67 (dq, J = 10.4, 6.0 Hz, 1H) , 4.99 (d, J = 9.5 Hz, 1H) , 4.85 (d, J = 17.5 Hz, 1H) , 4.74 (s, 1H) , 4.56 (d, J = 9.8 Hz, 1H) , 3.80 –3.71 (m, 1H) , 3.61 (d, J = 9.2 Hz, 6H) , 3.29 (s, 2H) , 3.01 –2.92 (m, 1H) , 2.82 –2.76 (m, 1H) , 2.74 (dd, J = 8.5, 5.5 Hz, 2H) , 2.59 (t, J = 11.2 Hz, 2H) , 2.34 (s, 4H) , 2.24 –2.20 (m, 1H) , 2.18 (d, J = 6.4 Hz, 2H) , 2.05 –1.97 (m, 1H) , 1.89 (dd, J = 13.6, 7.4 Hz, 1H) , 1.76 (d, J = 12.0 Hz, 2H) , 1.70 (dd, J = 13.7, 7.4 Hz, 1H) , 1.64 (d, J = 10.9 Hz, 1H) , 1.49 (s, 6H) , 1.39 (s, 2H) , 1.24 –1.16 (m, 2H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 48: (R) -1- (5- (9- ( (1- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-048)
Cpd-048 was synthesized according to the following scheme:
Step A: 1- (2-methoxy-5- (9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-
diazaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 3- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane (220 mg, 0.591 mmol) in DMF (5 mL) were added 3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) -4-methoxybenzoic acid (125 mg, 0.472 mmol) , DIPEA (229 mg, 1.77 mmol) and HATU (449 mg, 1.18 mmol) , the mixture was stirred at 25℃ for 2 h under N2 atmosphere. The mixture was quenched by the addition of 5 mL water, the reaction mixture was filtered through celite, and the filter cake washed with water (5 mL) . The combined filtrate was concentrated under reduced pressure to afford the title compound.
Step B: 1- (5- (9- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-
carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-methoxy-5- (9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (140 mg) in EtOH (10 mL) and H2O (1 mL) were added Fe (63.2 mg, 1.13 mmol) and NH4Cl (60.5 mg, 1.13 mmol) , the mixture was stirred at 90℃ for 3 h under N2 atmosphere. The mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was added Η20 (30 mL) and extracted by EtOAc (20 mL x 3) . The combined organic phase was washed with brine, dried over anhydrous Na2S04 and the organic layer was concentrated under reduced pressure to afford the title compound.
LC-MS (ESI) [M+H] + 589.4
Step C: (R) -1- (5- (9- ( (1- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carbonyl) -2-
methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (5- (9- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (100 mg) in DCM (10 mL) was added 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (67.9 mg, 0.170 mmol) and DIPEA (65.9 mg, 0.510 mmol) , then the reaction mixture was stirred at 20℃ for 18 h under N2 atmosphere. The reaction mixture was quenched with 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 17-27%Retention Time: 9-10of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 924.3
1HNMR (400 MHz, DMSO) δ 10.34 (s, 1H) , 10.10 (s, 1H) , 8.81 (s, 1H) , 7.91 (s, 1H) , 7.69 (d, J = 8.1 Hz, 1H) , 7.55 (s, 2H) , 7.37 (dd, J = 8.5, 2.1 Hz, 1H) , 7.32 (d, J = 2.1 Hz, 1H) , 7.15 (d, J = 8.6 Hz, 1H) , 6.90 (d, J = 8.9 Hz, 2H) , 5.66 (dq, J = 10.4, 5.9 Hz, 1H) , 5.04 (s, 1H) , 4.99 (d, J = 10.3 Hz, 1H) , 4.85 (d, J = 17.0 Hz, 1H) , 4.74 (s, 1H) , 4.57 (s, 1H) , 3.84 (s, 3H) , 3.62 (s, 1H) , 3.59 (d, J = 6.6 Hz, 3H) , 3.55 –3.38 (m, 3H) , 2.96 (dd, J = 14.5, 8.0 Hz, 1H) , 2.77 (dd, J = 15.0, 7.3 Hz, 1H) , 2.68 (t, J = 6.6 Hz, 2H) , 2.60 (t, J = 11.1 Hz, 2H) , 2.32 (s, 4H) , 2.24 –2.18 (m, 1H) , 2.16 (d, J = 7.3 Hz, 2H) , 2.02 (dd, J = 11.7, 6.7 Hz, 1H) , 1.89 (dd, J = 13.6, 7.4 Hz, 1H) , 1.78 (s, 1H) , 1.75 (s, 1H) , 1.70 (dd, J = 13.6, 7.3 Hz, 1H) , 1.63 (s, 1H) , 1.49 (s, 4H) , 1.42 (s, 4H) , 1.23 (s, 1H) , 1.19 (d,J = 10.7 Hz, 2H) , 0.87 (t, J = 7.4 Hz, 3H)
Example 49: 1- (5- (2- (4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) -8-azaspiro [4.5] decane-8-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-049)
Cpd-049 was synthesized according to the following scheme:
Step A: tert-butyl 2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane-8-carboxylate
A mixture of tert-butyl 2-oxo-8-azaspiro [4.5] decane-8-carboxylate (220 mg, 0.868 mmol) , 1- (4-nitrophenyl) piperazine (180 mg, 0.868 mmol) , CH3COOH (105 mg, 1.75 mmol) and NaBH (OAc) 3 (368.35 mg, 1.738 mmol) in DCM (6 mL) was stirred at 25℃ for 16 hours. The reaction was quenched with an aqueous solution of sat. NaHCO3 (5 mL) and the layers were
separated. The aqueous layer was extracted with CH2Cl2 (3 x 5 mL) . The combined organic extracts were dried (MgSO4) and concentrated under reduced pressure. The crude material was purified via flash column chromatography eluting EtOAc: hexanes (1: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 445.3
Step B: 2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decanetert-butyl 2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane-8-carboxylate (300 mg, 0.68 mmol) was dissolved in 10 mL of solvent (1, 4-Dioxane: DCM 4: 1) and 6 mL of anhydrous 4N HCl in dioxane was added dropwise in an ice bath. The solution was stirred overnight at room temperature and the solvent was evaporated to afford the title compound.
LC-MS (ESI) [M+H] + 345.2
Step C: 1- (2-chloro-5- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane-8-
carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
A solution of 2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane (200.00 mg) , perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (378 mg, 0.870 mmol) and DIEA (225 mg, 1.74 mmol) in DMF (8 mL) was stirred at room temperature for 2hrs. Water (10 mL) was added and the mixture was extracted with ethyl acetate (20 mL x 3) . The organic layer was dried and concentrated. The residue was purified by silica gel chromatography (eluted with petroleum ether: ethyl acetate =2: 1 to 1: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 595.3
Step D: 1- (5- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane-8-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a stirred solution of 1- (2-chloro-5- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane-8-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (118 mg, 0.198 mmol) in EtOH (10 mL) and water (2 mL) was added iron powder (55.45 mg, 0.99 mmol) followed by ammonium chloride (52.96 mg, 0.99 mmol) at room temperature. The mixture was stirred at 90℃ for 30 mins. The reaction mixture was diluted with ethyl acetate (20 mL) . The organic layer was washed with water (2 x 10 mL) , brine (20 mL) , dried over Na2SO4 and concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 565.3
Step E: 1- (5- (2- (4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) -8-azaspiro [4.5] decane-8-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
A solution of 1- (5- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane-8-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (90 mg) , 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (63.86 mg, 0.16 mmol) in DCM (10 mL) was added DIPEA (62.04 mg, 0.48 mmol) . The reaction mixture was stirred at 25° C for 24 hrs. The reaction mixture was concentrated and purified by HPLC (Waters 2767/Qda, Column: XBridge C18 19*250mm, 10 um;Mobile Phase A: 10mmol NH4HCO3/H2O, B: ACN; flow rate: 20ml/min; gradient: 40%~40%; Retention Time: 8.6-10.2min of 16 min) to afford the product.
LC-MS (ESI) [M+H] + 900.4
1H NMR (400 MHz, DMSO) δ 10.50 (s, 1H) , 10.10 (s, 1H) , 8.81 (s, 1H) , 7.92 (d, J = 7.5 Hz, 1H) , 7.69 (d, J = 8.1 Hz, 1H) , 7.64 (d, J = 8.2 Hz, 1H) , 7.55 (d, J = 1.9 Hz, 3H) , 7.39 (dd, J = 8.2, 2.0 Hz, 1H) , 6.90 (d, J = 9.1 Hz, 2H) , 5.66 (dq, J = 10.5, 6.0 Hz, 1H) , 5.03 (s, 1H) , 4.99 (d, J = 10.3 Hz, 1H) , 4.85 (d, J = 17.0 Hz, 1H) , 4.73 (s, 1H) , 4.57 (d, J = 16.0 Hz, 1H) , 3.75 (s, 1H) , 3.63 (d, J = 6.3 Hz, 3H) , 3.30 –3.22 (m, 2H) , 3.08 (s, 4H) , 3.03 –2.91 (m, 1H) , 2.82 –2.70 (m, 3H) , 2.60 (s, 1H) , 2.54 (s, 3H) , 2.25 –2.16 (m, 1H) , 2.06 –1.97 (m, 1H) , 1.89 (dd, J = 13.7, 7.3 Hz, 3H) , 1.70 (dd, J = 13.5, 7.3 Hz, 1H) , 1.49 (s, 7H) , 1.31 (s, 1H) , 1.23 (s, 1H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 50: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-ethoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-050)
Cpd-050 was synthesized according to the following scheme:
Step A: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-
ethoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -6- ( (4- (4- ( (3-azaspiro [5.5] undecan-9-yl) methyl) piperazin-1-yl) phenyl) amino) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (40.0 mg, 0.0590 mmol) in DMF (1 mL) were added perfluorophenyl 3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) -4-ethoxybenzoate (31.5 mg, 0.0708 mmol) and DIEA (22.8 mg, 0.177 mmol) . The mixture was stirred at 25℃ for 1 h. The mixture was diluted with H2O (10 mL) and extracted with EtOAc (20 mL x 2) . The combined organic layers was washed with brine (20 mL) , dried over Na2SO4, filtered, and the filtrate was concentrated by vacuum, the residue was purified by Pre-HPLC (Waters 2767/Qda, Column: XBridge C18 19*250mm, 10 um; Mobile Phase A: 10mmol NH4HCO3/H2O, B: ACN; flow rate: 20ml/min; gradient: 40%~40%; Retention Time: 8.6-10.2min of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 938.5
1H NMR (400 MHz, DMSO) δ 10.33 (s, 1H) , 10.11 (s, 1H) , 8.82 (s, 1H) , 8.33 (s, 1H) , 7.92 (s, 1H) , 7.69 (d, J = 8.2 Hz, 1H) , 7.57 (s, 2H) , 7.36 –7.30 (m, 2H) , 7.13 (d, J = 8.5 Hz, 1H) , 6.91 (d, J = 8.7 Hz, 2H) , 5.67 (ddd, J = 16.6, 11.0, 6.0 Hz, 1H) , 5.01 (t, J = 13.9 Hz, 2H) , 4.85 (d, J = 16.5 Hz, 1H) , 4.74 (s, 1H) , 4.58 (s, 1H) , 4.11 (q, J = 6.8 Hz, 2H) , 3.60 (d, J = 6.7 Hz, 2H) , 3.43 (s, 4H) , 3.08 (s, 4H) , 2.98 (d, J = 7.8 Hz, 1H) , 2.80 –2.75 (m, 1H) , 2.68 (d, J = 6.4 Hz, 2H) , 2.47 (s, 4H) , 2.19 (d, J = 13.3 Hz, 3H) , 2.02 (d, J = 5.3 Hz, 1H) , 1.90 –1.85 (m, 1H) , 1.70 (d, J = 6.6 Hz, 3H) , 1.61 –1.45 (m, 6H) , 1.32 (d, J = 7.0 Hz, 4H) , 1.10 (d, J = 11.5 Hz, 4H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 51: 3- [6- [7- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -2-azaspiro [3.5] nonan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-051)
Cpd-051 was synthesized according to the following scheme:
Step A: O2-tert-butyl O7-isopropoxycarbonyl 2-azaspiro [3.5] nonane-2, 7-dicarboxylate
To a solution of 2-tert-butoxycarbonyl-2-azaspiro [3.5] nonane-7-carboxylic acid (200 mg, 742.57 umol, 1 eq) in THF (5 mL) were added Et3N (300.56 mg, 2.97 mmol, 413.42 uL) and isopropyl carbonochloridate (136.50 mg, 1.11 mmol, 154.59 uL) at 0 ℃. The mixture was stirred at 0 ℃ for 2 hours. The mixture was filtered and the filter cake was washed with THF (30 mL) . The filtrate was concentrated under vacuum to afford the title compound.
1H NMR (400 MHz, CDCl3) δ: 4.95–5.05 (m, 1H) , 3.61 (s, 2H) , 3.58 (s, 2H) , 2.37–2.41 (m, 1H) , 1.93–1.96 (m, 4H) , 1.49–1.53 (m, 4H) , 1.44 (s, 9H) , 1.35–1.37 (m, 3H) , 1.34–1.35 (m, 3H) .
Step B: tert-butyl 7- (hydroxymethyl) -2-azaspiro [3.5] nonane-2-carboxylate
To a solution of O2-tert-butyl O7-isopropoxycarbonyl 2-azaspiro [3.5] nonane-2, 7-dicarboxylate (250 mg) in THF (4 mL) and H2O (0.4 mL) was added LiBH4 (91.92 mg, 4.22 mmol) at 0 ℃ .
The mixture was stirred at 0 ℃ for 2 hrs. The mixture was diluted with water (5 mL) and extracted with EtOAc (30 mL × 3) . The combined organic layers was dried over Na2SO4, filtered and concentrated to afford the title compound.
1H NMR (400 MHz, CDCl3) δ: 3.59 (s, 2H) , 3.56 (s, 2H) , 3.45 (d, J = 6.4 Hz, 2H) , 1.92 (d, J =12.0 Hz, 4H) , 1.73 (d, J = 13.6 Hz, 4H) , 1.45 (s, 9H) , 1.19–1.22 (m, 1H)
Step C: 2-azaspiro [3.5] nonan-7-ylmethanol
A mixture of tert-butyl 7- (hydroxymethyl) -2-azaspiro [3.5] nonane-2-carboxylate (100 mg) in DCM (2.5 mL) and HCl/dioxane (4M, 2 mL) was stirred at 20 ℃ for 2 hrs. The reaction mixture was concentrated under reduced pressure to afford the title compound.
1H NMR (400 MHz, CDCl3) δ: 2.90–2.96 (m, 2H) , 2.37 (s, 1H) , 2.28 (s, 4H) , 1.20 (t, J = 7.2 Hz, 4H) , 0.89 (s, 4H) .
Step D: 3- [6- [7- (hydroxymethyl) -2-azaspiro [3.5] nonan-2-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
Schlenk tube charged with a mixture of 3- (6-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (99.2 mg, 306.99 umol) , 2-azaspiro [3.5] nonan-7-ylmethanol (60 mg) , 1, 3-bis [2, 6-bis (1-propylbutyl) phenyl] -4, 5-dichloro-2H-imidazol-1-ium-2-ide; 3-chloropyridine; dichloropalladium (15.22 mg, 15.65 umol) and Cs2CO3 (173.36 mg, 532.09 umol) in dioxane (5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 4 hrs under N2 atmosphere. aq. AcOH (5%w/t) (10 mL) was added at 0 ℃, and the mixture was extracted with ethyl acetate (50 mL × 3) . The combined organic phases was dried with anhydrous Na2SO4, filtered and concentrated in under reduce pressure. The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 0~10% MeOH/DCM gradient @30 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 398.2 [M+H] +.
Step E: 2- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -2-azaspiro [3.5] nonane-7-
carbaldehyde
To a solution of 3- [6- [7- (hydroxymethyl) -2-azaspiro [3.5] nonan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (30 mg, 75.48 umol) in DCM (3 mL) and DMF (1 mL) was added Dess-Martin reagent (48.02 mg, 113.22 umol) at 0℃. The mixture was stirred at 25℃ for 4 hrs. The reaction mixture was quenched with sat. aq. NaHCO3 solution (10 mL) and extracted with ethyl acetate (40 mL × 3) . The combined organic layers was dried over Na2SO4, filtered and concentrated to afford the title compound.
LCMS: MS (ESI) m/z = 396.1 [M+H] +.
Step F: 3- [6- [7- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -2-
azaspiro [3.5] nonan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To solution of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (22 mg, 42.92 umol) and 2- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -2-azaspiro [3.5] nonane-7-carbaldehyde (30 mg) in DCM (2 mL) was added TEA (38.38 mg, 379.31 umol, 52.80 uL) . The mixture was stirred at 20 ℃ for 0.5 hr. Then NaBH (OAc) 3 (48.24 mg, 227.59 umol) was added and the mixture was stirred at 20 ℃ for 3 hrs. The reaction mixture was quenched with H2O (10 mL) and extracted with ethyl acetate (50 mL × 3) . The combined organic phase was dried with anhydrous Na2SO4, filtered and concentrated under reduce pressure. The residue was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 39%-69%, 8 min) to afford the title compound.
LCMS: MS (ESI) m/z = 892.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 10.97 (s, 1H) , 10.14 (br s, 1H) , 8.83 (s, 1H) , 7.93 (s, 1H) , 7.70 (d, J = 8.0 Hz, 1H) , 7.59 (s, 2H) , 7.38 (d, J = 8.0 Hz, 1H) , 6.93 (d, J = 8.8 Hz, 2H) , 6.66-6.71 (m, 2H) , 5.61-5.74 (m, 1H) , 5.06-5.12 (m, 2H) , 5.00 (d, J = 10.2 Hz, 1H) , 4.86 (d, J = 17.6 Hz, 1H) , 4.75 (s, 1H) , 4.57 (d, J = 9.6 Hz, 1H) , 4.28-4.34 (m, 1H) , 4.16-4.22 (m, 1H) , 3.53-3.60 (m, 4H) , 3.42-3.49 (m, 2H) , 3.10 (s, 4H) , 2.87-3.03 (m, 2H) , 2.71-2.85 (m, 1H) , 2.13-2.24 (m, 3H) , 2.01 (dd, J = 13.2, 5.2 Hz, 3H) , 1.91 (d, J = 13.2 Hz, 3H) , 1.66-1.79 (m, 3H) , 1.39-1.62 (m, 4H) , 1.24 (s, 2H) , 0.98 (d, J = 12.0 Hz, 2H) , 0.88 (t, J = 7.2 Hz, 3H) .
Example 52: 3- (5- (1- ( (1s, 4s) -4- ( (4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) cyclohexyl) -1H-pyrazol-4-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (Cpd-052)
Cpd-052 was synthesized according to the following scheme:
Step A: methyl 4-methylsulfonyloxycyclohexanecarboxylate
To a solution of methyl 4-hydroxycyclohexanecarboxylate (3 g, 18.96 mmol) , DIEA (7.35 g, 56.89 mmol, 9.91 mL) in DCM (30 mL) was added methylsulfonyl methanesulfonate (3.96 g, 22.76 mmol) at 0 ℃ under N2. The mixture was stirred at 0-25 ℃ for 3 hrs under N2. The mixture was poured into water (100 mL) and extracted with DCM (200 mL × 2) . The combined organic layers was washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give the title compound.
1H NMR (400 MHz, CDCl3) δ: 4.54–4.81 (m, 1H) , 3.68 (s, 3H) , 3.02 (s, 3H) , 2.28–2.40 (m, 1H) , 2.15–2.24 (m, 2H) , 2.03–2.10 (m, 2H) , 1.59–1.65 (m, 4H) .
Step B: methyl 4- (4-bromopyrazol-1-yl) cyclohexanecarboxylate
To a solution of methyl 4-methylsulfonyloxycyclohexanecarboxylate (4 g) , 4-bromo-1H-pyrazole (2.49 g, 16.93 mmol) in DMF (50 mL) was added Cs2CO3 (11.03 g, 33.86 mmol) . The mixture was stirred at 90 ℃ for 16 hrs under N2. The mixture was poured into water (100 mL)
and extracted with EtOAc (200 mL × 2) . The combined organic layers was washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by column chromatography on silica gel (12%EtOAc in Petroleum ether) to afford the title compound.
LCMS: MS (ESI) m/z = 287.1 [M+H] +.
1H NMR (400 MHz, CDCl3) δ: 7.46 (d, J = 3.2 Hz, 1H) , 7.43 (s, 1H) , 4.10–4.24 (m, 1H) , 3.70 (s, 3H) , 2.33–2.43 (m, 1H) , 2.15–2.26 (m, 4H) , 1.63–1.84 (m, 4H) .
Step C: [4- (4-bromopyrazol-1-yl) cyclohexyl] methanol
To a solution of methyl 4- (4-bromopyrazol-1-yl) cyclohexanecarboxylate (1 g, 3.48 mmol) in THF (20 mL) was added LiBH4 (379.30 mg, 17.41 mmol) at 0 ℃. The mixture was stirred at 25 ℃ for 3 hrs under N2. The mixture was quenched with MeOH (5 mL) and stirred for 10 mins, then the mixture was concentrated to give a residue, which was purified by column chromatography on silica gel (45%EtOAc in Petroleum ether) to afford the title compound.
LCMS: MS (ESI) m/z = 261.1 [M+H] +.
1H NMR (400 MHz, CDCl3) δ: 7.45 (d, J = 6.8 Hz, 2H) , 4.02–4.12 (m, 1H) , 3.53 (d, J = 6.4 Hz, 2H) , 2.17–2.28 (m, 2H) , 1.95–2.04 (m, 2H) , 1.72–1.80 (m, 2H) , 1.65–1.71 (m, 1H) , 1.12–1.24 (m, 2H) .
Step D: [4- [4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazol-1-
yl] cyclohexyl] methanol
A microwave tube charged with a mixture of [4- (4-bromopyrazol-1-yl) cyclohexyl] methanol (200 mg, 771.78 umol) , 4, 4, 5, 5-tetramethyl-2- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1, 3, 2-dioxaborolane (293.97 mg, 1.16 mmol) , KOAc (227.23 mg, 2.32 mmol) , Pd (dppf) Cl2
. CH2Cl2 (63.03 mg, 77.18 umol) in 2-methyltetrahydrofuran (3 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 120 ℃ for 1 hr under microwave condition. The mixture was concentrated under reduced pressure to give a product, which was purified by column chromatography on silica gel (3% MeOH in Dichloromethane) to afford the title compound.
LCMS: MS (ESI) m/z = 307.2 [M+H] +
1H NMR (400 MHz, CDCl3) δ: 7.78–7.83 (m, 1H) , 7.72–7.77 (m, 1H) , 4.10–4.18 (m, 1H) , 3.73 (dd, J = 6.0, 2.4 Hz, 1H) , 3.53 (dd, J = 6.0, 2.4 Hz, 2H) , 2.24 (d, J = 10.8 Hz, 2H) , 1.94–1.99 (m, 2H) , 1.75–1.81 (m, 2H) , 1.56–1.67 (m, 3H) , 1.32 (s, 12H) .
Step E: 3- [5- [1- [4- (hydroxymethyl) cyclohexyl] pyrazol-4-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
A microwave tube charged with a mixture of [4- [4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazol-1-yl] cyclohexyl] methanol (150 mg, 489.86 umol) , 3- (5-bromo-1-oxo-isoindolin-2-
yl) piperidine-2, 6-dione (158.30 mg, 489.86 umol) , Pd (dppf) Cl2
. CH2Cl2 (40.00 mg, 48.99 umol) , KF (56.92 mg, 979.73 umol) in dioxane (5 mL) and H2O (0.5 mL) was degassed and purged with N2, and then the mixture was stirred at 120 ℃ in microwave for 1 hr under N2 atmosphere. The mixture was poured into water (20 mL) and extracted with DCM (20 mL × 2) . The combined organic layers was washed with brine (20 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by column chromatography on silica gel (8%MeOH in Dichloromethane) to afford the title compound.
LCMS: MS (ESI) m/z = 423.2 [M+H] +
Step F: 4- [4- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] pyrazol-1-
yl] cyclohexanecarbaldehyde
To a solution of 3- [5- [1- [4- (hydroxymethyl) cyclohexyl] pyrazol-4-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (100 mg, 236.70 umol) in DCM (5 mL) was added Dess-Martin reagent (150.59 mg, 355.05 umol) . The mixture was stirred at 25 ℃ for 1 hr under N2. The mixture was poured into water (20 mL) and extracted with DCM (20 mL × 2) . The combined organic layers was washed with brine (20 mL × 2) , dried over Na2SO4, filtered and concentrated to afford the title compound.
LCMS: MS (ESI) m/z = 421.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 10.96 (s, 1H) , 9.59–9.74 (m, 1H) , 8.33–8.37 (m, 1H) , 8.05 (d, J = 4.4 Hz, 1H) , 7.97–8.00 (m, 1H) , 7.73–7.76 (m, 1H) , 7.67–7.71 (m, 1H) , 5.09 (dd, J = 13.2, 5.2 Hz, 1H) , 4.29–4.52 (m, 2H) , 2.84–2.97 (m, 1H) , 2.57–2.63 (m, 1H) , 2.07–2.18 (m, 3H) , 1.98–2.04 (m, 1H) , 1.72–1.87 (m, 3H) , 1.37–1.50 (m, 1H) .
Step G: 3- (5- (1- ( (1s, 4s) -4- ( (4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) methyl) cyclohexyl) -1H-pyrazol-4-yl) -1-oxoisoindolin-2-
yl) piperidine-2, 6-dione
A mixture of 4- [4- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] pyrazol-1-yl] cyclohexanecarbaldehyde (24.61 mg) , 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (30 mg, 58.52 umol) , NaBH (OAc) 3 (62.02 mg, 292.62 umol) in DCM (3 mL) was added TEA (29.61 mg, 292.62 umol) . The mixture was stirred at 25 ℃ for 16 hrs. The mixture was poured into water (30 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers was washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by Pre-TLC (Dichloromethane: Methanol = 10: 1) and then by prep-HPLC (column: Phenomenex C18 75*30mm*3um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 35%-75%, 9 min) to afford the title compound.
LCMS: MS (ESI) m/z = 917.5 [M+H] +
1H NMR (400 MHz, CD3OD) δ 8.79 (s, 1H) , 8.21 (s, 1H) , 7.96 (s, 1H) , 7.86 (s, 1H) , 7.71–7.82 (m, 4H) , 7.49–7.59 (m, 2H) , 6.97 (d, J = 7.6 Hz, 2 H) , 4.57–4.60 (m, 6H) , 4.48–4.54 (m, 1H) , 3.15–3.25 (m, 4H) , 3.00–3.10 (m, 1H) , 2.80–2.94 (m, 2H) , 2.60–2.70 (m, 4H) , 2.45–2.57 (m, 1H) , 2.30–2.43 (m, 3H) , 2.15–2.25 (m, 4H) , 2.00–2.12 (m, 3H) , 1.80–1.95 (m, 3H) , 1.67–1.79 (m, 1H) , 1.18–1.36 (m, 8H) , 0.95 (t, J = 6.8 Hz, 3H) .
Example 53: 3- (6- (4- ( (1- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperidin-4-yl) methyl) piperazin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (Cpd-053)
Cpd-053 was synthesized according to the following scheme:
Step A: tert-butyl 4- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] piperazine-1-
carboxylate
Schlenk tube charged with a mixture of 3- (6-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (150 mg, 464.19 umol) , tert-butyl piperazine-1-carboxylate (110 mg, 590.60 umol) , Pd-PEPPSI-IHeptCl (36.84 mg, 46.42 umol) , Cs2CO3 (226.86 mg, 696.29 umol) in dioxane (3 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 120 ℃ for 4 hours. The resulting mixture concentrated under reduce pressure to give which was purified by flash column chromatography on silica gel (MeOH in CH2Cl2 from 4%to 4%) to afford the title compound.
LCMS: MS (ESI) m/z = 373.1 [M+H] +.
Step B: 3- (1-oxo-6-piperazin-1-yl-isoindolin-2-yl) piperidine-2, 6-dione
To a solution of tert-butyl 4- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] piperazine-1-carboxylate (110 mg, 256.72 umol) in CH2Cl2 (2 mL) was added TFA (2.11 g, 18.54 mmol) .
The mixture was stirred at 25 ℃ for 2 hours. The reaction was mixture concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 679.3 [M+H] +.
Step C: 3- [6- [4- [ [1- (4-nitrophenyl) -4-piperidyl] methyl] piperazin-1-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
To a solution of 3- (1-oxo-6-piperazin-1-yl-isoindolin-2-yl) piperidine-2, 6-dione (3, 95 mg) in CH2Cl2 (3 mL) was added 1- (4-nitrophenyl) piperidine-4-carbaldehyde (66.50 mg, 283.88 umol) and TEA (73.19 mg, 723.28 umol) , the mixture was stirred at 25℃ for 2 hours, NaBH (OAc) 3 (183.95 mg, 867.94 umol) was added. The mixture was stirred at 25 ℃ for 3 hours. The residue was diluted with H2O (20 mL) and extracted with CH2Cl2 (30 mL × 3) . The combined organic layers was washed with brine (30 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give crude product, which was purified by flash column chromatography on silica gel (MeOH in CH2Cl2 from 5%to 7 %) to afford the title compound.
LCMS: MS (ESI) m/z = 547.2 [M+H] +.
Step D: 3- [6- [4- [ [1- (4-aminophenyl) -4-piperidyl] methyl] piperazin-1-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
To a solution of 3- [6- [4- [ [1- (4-nitrophenyl) -4-piperidyl] methyl] piperazin-1-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (150 mg, 274.42 umol) in THF (4 mL) was added Pd/C (0.2 g, 10%w/w) . The mixture was stirred 12 hours at room under hydrogen atmosphere (15 Psi) . The reaction mixture was filtered through diatomite, and the filter cake was washed by DMAc (20 mL) , the filtrate was concentrated in vacuum to afford the title compound.
LCMS) : MS (ESI) m/z = 519.2 [M+H] +.
Step E: 3- (6- (4- ( (1- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperidin-4-yl) methyl) piperazin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-
dionee
To a solution of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6-methylsulfanyl-pyrazolo [3, 4-d] pyrimidin-3-one (59.38 mg, 154.85 umol) in toluene (1 mL) was added m-CPBA (34.58 mg, 170.33 umol, 85%purity) at 0 ℃. The mixture was stirred 1 hour. Then 3- [6- [4- [ [1- (4-aminophenyl) -4-piperidyl] methyl] piperazin-1-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (80 mg) and DIEA (60.04 mg, 464.55 umol) were added. The mixture was stirred at 25 ℃ for 16 hours. The residue was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 46%-76%, 8min) to afford the title compound.
LCMS: MS (ESI) m/z = 852.6 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 10.97 (s, 1H) , 10.1 (br s, 1H) , 8.81 (s, 1H) , 7.95-8.03 (m, 1H) , 7.69 (d, J = 8.0 Hz, 1H) , 7.60-7.53 (m, 2H) , 7.43 (d, J = 8.4 Hz, 1H) , 7.29-7.24 (m, 1H) , 7.18-7.15 (m, 1H) , 6.92 (d, J = 8.8 Hz, 2H) , 5.74-5.60 (m, 1H) , 5.13 -5.04 (m, 2H) , 5.0 (d, J = 10.0 Hz, 1H) , 4.85 (d, J = 16.0 Hz, 1H) , 4.80-4.67 (m, 1H) , 4.60-4.50 (m, 1H) , 4.33 (d, J = 17.2 Hz, 1H) , 4.21 (d, J = 17.2 Hz, 1H) , 3.68-3.58 (m, 3H) , 3.23-3.16 (m, 4H) , 3.04-3.70 (m, 4H) , 2.68-2.55 (m, 4H) , 2.42-2.33 (m, 2H) , 2.32-2.15 (m, 4H) , 2.05-1.93 (m, 3H) , 1.90-1.78 (m, 3H) , 1.74-1.65 (m, 2H) , 0.87 (t, J = 7.2Hz, 3H) .
Example 54: 3- [5- [9- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-054)
Cpd-054 was synthesized according to the following scheme:
Step A: 3-azaspiro [5.5] undecan-9-ylmethanol
A mixture of tert-butyl 9- (hydroxymethyl) -3-azaspiro [5.5] undecane-3-carboxylate (100 mg, 352.85 umol) , HCl/dioxane (4 M, 4.00 mL) in EtOAc (2 mL) was stirred at 25 ℃ for 12 hr under N2 atmosphere. The reaction was concentrated in vacuo to afford the title compound.
Step B: 3- [5- [9- (hydroxymethyl) -3-azaspiro [5.5] undecan-3-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
A Schlenk tube charged with a mixture of 3-azaspiro [5.5] undecan-9-ylmethanol (77 mg) , 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (136 mg, 420.87 umol) , 1, 3-bis [2, 6-bis (1-propylbutyl) phenyl] -4, 5-dichloro-2H-imidazol-1-ium-2-ide; 3-chloropyridine; dichloropalladium (34.0 mg, 34.95 umol) , Cs2CO3 (340.0 mg, 1.04 mmol) in dioxane (3 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 120 ℃ for 15 hrs under N2 atmosphere. H2O (30 mL) was added at 25 ℃, and then extracted with EtOAc (60 mL × 3) . The combined organic layers was washed with brine 60 mL, dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (12 g Silica Flash Column, Eluent of 0~10%MeOH/DCM gradient @50 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 426.0 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ: 10.97 (s, 1H) , 7.41 (d, J = 8.4 Hz, 1H) , 7.24 (dd, J = 2.4, 8.4 Hz, 1H) , 7.15 (d, J = 2.0 Hz, 1H) , 5.10 (dd, J = 5.2, 13.2 Hz, 1H) , 4.41 -4.30 (m, 2H) , 4.23 -4.16 (m, 1H) , 3.25 -3.15 (m, 5H) , 2.97 -2.85 (m, 1H) , 2.63 -2.56 (m, 1H) , 2.42 - 2.30 (m, 1H) , 2.03 -1.96 (m, 1H) , 1.72 -1.65 (m, 2H) , 1.62 -1.57 (m, 2H) , 1.53 (br s, 2H) , 1.47 - 1.42 (m, 2H) , 1.13 -1.04 (m, 4H) .
Step C: 3- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -3-azaspiro [5.5] undecane-9-
carbaldehyde
A mixture of 3- [5- [9- (hydroxymethyl) -3-azaspiro [5.5] undecan-3-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (25 mg, 58.75 umol) , Dess-Martin reagent (29 mg, 68.37 umol , 68.37 umol) in DCM (2 mL) was stirred at 25 ℃ for 1.5 hrs. DMP (6 mg, 14.15 umol) was added and the mixture was stirred for another 1 hr at 25 ℃. The reaction was diluted with DCM (20 mL) and sat. NaHCO3 (15 mL) , the mixture was extracted with DCM (30 mL × 3) , The combined organic layers was washed with brine (20 mL) and dried over Na2SO4, filtered and concentrated in vacuo to afford the title compound., which was used in next step without further purifications. LCMS: MS (ESI) m/z = 424.0 [M+H] +.
Step D: 3- [5- [9- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -3-
azaspiro [5.5] undecan-3-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
A mixture of 3- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] -3-azaspiro [5.5] undecane-9-carbaldehyde (23 mg, 54.31 umol) , 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (31 mg) and TEA (11 mg, 108.71 umol, 15.13 uL) in DCM (2 mL) was stirred at 25 ℃ for 5 hrs. NaBH (OAc) 3 (36 mg, 169.86 umol) was added and the mixture was stirred at 25℃ for 12 hrs. The reaction was quenched with H2O (1 mL) and DMF (2 mL) , concentrated in vacuo to remove CH2Cl2. The residue was diluted with CH3CN (2 mL) and filtered. The solid cake was purified by prep_HPLC (column: Welch Xtimate C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; B%: 16%-46%, 7min) to afford the title compound.
LCMS: MS (ESI) m/z = 920.7 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ: 11.01 -10.95 (m, 1H) , 8.82 (s, 1H) , 7.96 - 7.88 (m, 1H) , 7.77 -7.63 (m, 2H) , 7.62 -7.52 (m, 2H) , 7.41 (d, J = 8.4 Hz, 1H) , 7.27 - 7.23 (m, 1H) , 7.15 (s, 1H) , 6.94 -6.89 (m, 2H) , 5.72 -5.59 (m, 1H) , 5.12 -5.06 (m, 2H) , 5.03 - 4.98 (m, 1H) , 4.90 - 4.83 (m, 1H) , 4.38 -4.29 (m, 1H) , 4.23 -4.17 (m, 1H) , 3.22 -3.16 (m, 11H) , 3.09 (m, 4H) , 2.22 -2.17 (m, 4H) , 2.06 -1.82 (m, 6H) , 1.72 -1.68 (m, 2H) , 1.63 -1.58 (m, 4H) , 1.46 (m, 2H) , 1.24 (s, 3H) , 1.15 -1.10 (m, 4H) , 0.88 (m, 3H) .
Example 55: 3- [6- [4- [4- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] pyrazol-1-yl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-055)
Cpd-055 was synthesized according to the following scheme:
Step A: tert-butyl 4- (4-formylpyrazol-1-yl) piperidine-1-carboxylate
To a solution of 1H-pyrazole-4-carbaldehyde (200 mg, 2.08 mmol) and tert-butyl 4- (p-tolylsulfonyloxy) piperidine-1-carboxylate (887.81 mg, 2.50 mmol) in DMF (5 mL) was added Cs2CO3 (1.36 g, 4.16 mmol) . The mixture was stirred at 90 ℃ for 16 hrs. The mixture was poured into water (200 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers was washed with brine (50 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by column chromatography on silica gel (28% Ethyl acetate in Petroleum ether) to afford the title compound.
LCMS: MS (ESI) m/z = 280.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 9.78 (s, 1H) , 8.53 (s, 1H) , 8.00 (s, 1H) , 4.39–4.51 (m, 1H) , 4.03–4.09 (m, 2H) , 2.86–3.03 (m, 2H) , 2.00–2.05 (m, 2H) , 1.74–1.84 (m, 2H) , 1.41 (s, 9H) .
Step B: 1- (4-piperidyl) pyrazole-4-carbaldehyde
A mixture of tert-butyl 4- (4-formylpyrazol-1-yl) piperidine-1-carboxylate (570 mg, 2.04 mmol) in HCl/dioxane (4M, 2 mL) was stirred at 25 ℃ for 2 hrs. The reaction mixture was added H2O (20 mL) , then lyophilized directly to afford the title compound.
LCMS: MS (ESI) m/z = 180.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 9.81 (d, J = 1.2 Hz, 1H) , 8.54 (s, 1H) , 8.05 (s, 1H) , 4.56–4.66 (m, 1H) , 3.37 (d, J = 13.2 Hz, 2H) , 3.05 (d, J = 9.6 Hz, 2H) , 2.10–2.30 (m, 4H) .
Step C: 1- [1- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -4-piperidyl] pyrazole-4-
carbaldehyde
A Schlenk tube charged with a mixture of 1- (4-piperidyl) pyrazole-4-carbaldehyde (150 mg) , 3- (6-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (224.74 mg, 695.48 umol) , Pd-PEPPSI-
IHeptCl (33.83 mg, 34.77 umol) and Cs2CO3 (679.80 mg, 2.09 mmol) in dioxane (5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 6 hrs under N2 atmosphere. aq. AcOH (5%w/t) (10 mL) at 0 ℃ was added and the mixture was extracted with ethyl acetate (50 mL × 3) . The combined organic phases was dried with anhydrous Na2SO4, filtered and concentrated to give a residue. The residue was purified by column chromatography on silica gel (7%Methanol in Dichloromethane) to afford the title compound.
LCMS: MS (ESI) m/z = 422.2 [M+H] +.
Step D: 3- [6- [4- [4- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-
dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-
yl] amino] phenyl] piperazin-1-yl] methyl] pyrazol-1-yl] -1-piperidyl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
To a solution of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (14.19 mg, 27.68 umol) and 1- [1- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -4-piperidyl] pyrazole-4-carbaldehyde (20 mg, 33.22 umol, 70%purity) in DCM (1 mL) were added NaBH (OAc) 3 (8.80 mg, 41.52 umol) and Et3N (14.01 mg, 138.41 umol, 19.27 uL) . The mixture was stirred at 25 ℃ for 32 hrs. The mixture was poured into water (10 mL) and extracted with DCM (50 mL) . The organic layer was dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by prep-HPLC (column: Phenomenex C18 75*30mm*3um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 28%-68%, 9 min) to afford the title compound.
LCMS: MS (ESI) m/z = 918.4 [M+H] +.
1H NMR (400 MHz, CDCl3) δ: 8.81 (s, 1H) , 8.59 (s, 1H) , 7.64–7.71 (m, 2H) , 7.41–7.50 (m, 5H) , 7.35 (d, J = 8.4 Hz, 1H) , 7.21 (dd, J = 8.4, 1.6 Hz, 1H) , 6.89 (d, J = 8.8 Hz, 2H) , 5.63–5.73 (m, 1H) , 5.22 (dd, J = 13.2, 5.2 Hz, 1H) , 5.01 (d, J = 10.0 Hz, 1H) , 4.88–4.95 (m, 1H) , 4.82 (dd, J = 15.6, 5.2 Hz, 1H) , 4.64–4.72 (m, 1H) , 4.25–4.46 (m, 3H) , 3.88 (d, J = 12.4 Hz, 2H) , 3.53 (s, 2H) , 3.21 (s, 4H) , 2.95–3.07 (m, 3H) , 2.78–2.92 (m, 3H) , 2.66 (s, 4H) , 2.33–2.43 (m, 2H) , 2.10–2.32 (m, 7H) , 2.02 (dd, J = 14.0, 7.2 Hz, 1H) , 1.84 (dd, J = 14.0, 7.2 Hz, 1H) , 0.98 (t, J = 7.6 Hz, 3H) .
Example 56: 3- [6- [2- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-yl] amino] phenyl] piperazin-1-yl] methyl] -7-azaspiro [3.5] nonan-7-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-056)
Cpd-056 was synthesized according to the following scheme:
Step A: 3- [6- [2- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-
yl] amino] phenyl] piperazin-1-yl] methyl] -7-azaspiro [3.5] nonan-7-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
To a solution of (12Z, 16R) -16-hydroxy-16-methyl-5- (4-piperazin-1-ylanilino) -2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (70 mg, 140.4 umol) and 7- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] -7-azaspiro [3.5] nonane-2-carbaldehyde (111 mg, 280.8 umol) in DCM (3.0 mL) was added TEA (71 mg, 702.0 umol) under nitrogen atmosphere. The mixture was stirred at 25 ℃ for 1 hr. Then NaBH (OAc) 3 (45 mg, 210.6 umol) was added to the reaction mixture, the reaction mixture was stirred at 25 ℃ for additional 1 hr. The reaction mixture was concentrated in vacuum. The residue was diluted with DMSO (3.0 mL) and water (0.5 mL) . The mixture was purified by prep-HPLC (column: C18-1 150*30mm*5um; mobile phase: [water (FA) -ACN] ; B%: 10%-50%, 9min) to afford the title compound.
LCMS: MS (ESI) m/z = 878.5 [M+H] +) .
1H NMR (400 MHz, DMSO-d6) δ: 10.95 (s, 1H) , 10.05 (s, 1H) , 8.82 (s, 1H) , 8.00 (d, J=7.2 Hz, 1H) , 7.75 (d, J=8.0 Hz, 1H) , 7.57 (t, J=8.0 Hz, 3H) , 7.49 (d, J=8.8 Hz, 1H) , 7.07 - 7.00 (m, 2H) , 6.89 (d, J=8.8 Hz, 2H) , 5.38 -5.25 (m, 2H) , 5.03 (dd, J=4.8, 13.2 Hz, 1H) , 4.45 (d, J = 5.2 Hz, 2H) , 4.35 -4.27 (m, 1H) , 4.23 -4.14 (m, 1H) , 3.30 -3.25 (m, 4H) , 3.24 -3.17 (m, 3H) , 3.10 -3.03 (m, 4H) , 2.96 -2.84 (m, 1H) , 2.67 (s, 1H) , 2.63 -2.59 (m, 1H) , 2.44 - 2.38 (m, 3H) , 2.38 -2.27 (m, 2H) , 2.24 -2.12 (m, 1H) , 2.03 -1.89 (m, 5H) , 2.03 -1.89 (m, 3H) , 1.57 - 1.52 (m, 4H) , 1.49 -1.42 (m, 2H) .
Example 57: 3- [5- [4- [ [3- [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] pyrrolidin-1-yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-057)
Cpd-057 was synthesized according to the following scheme:
Step A: 3- [5- [4- [ [3- [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-
dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-
yl] amino] phenyl] piperazin-1-yl] pyrrolidin-1-yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
To a mixture of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- (4-pyrrolidin-3-ylpiperazin-1-yl) anilino] pyrazolo [3, 4-d] pyrimidin-3-one (12 mg, 20.63 umol) and 1- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde (7.33 mg, 20.63 umol) in DMF (2 mL) was added TEA (20.87 mg, 206.29 umol) . After stirred at 25 ℃ for
0.5 hour, NaBH (OAc) 3 (13.12 mg, 61.89 umol) was added and the mixture was stirred for 16 hours. The mixture was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 52%-82%, 8min) : 60 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 921.6 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 10.95 (s, 1H) , 10.22-10.02 (m, 1H) , 8.81 (s, 1H) , 7.95-7.90 (m, 1H) , 7.69 (d, J = 8.0 Hz, 1H) , 7.62-7.53 (m, 2H) , 7.50 (d, J = 9.2 Hz, 1H) , 7.06-7.01 (m, 2H) , 6.94-6.89 (m, 2H) , 5.72-5.61 (m, 1H) , 5.07-5.06 (m, 1H) , 5.05-4.97 (m, 2H) , 4.85 (d, J = 17.8 Hz, 1H) , 4.79-4.68 (m, 1H) , 4.61-4.52 (m, 1H) , 4.35-4.28 (m, 1H) , 4.22-4.16 (m, 1H) , 3.90-3.83 (m, 2H) , 3.11-3.06 (m, 4H) , 3.03-2.89 (m, 1H) , 2.88-2.74 (m, 4H) , 2.71-2.66 (m, 1H) , 2.36-2.27 (m, 6H) , 2.24-2.17 (m, 3H) , 2.06-1.74 (m, 9H) , 1.73-1.61 (m, 4H) , 1.20-1.13 (m, 2H) , 0.86 (t, J = 7.2 Hz, 3H) .
Example 58: 3- [6- [4- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-058)
Cpd-058 was synthesized according to the following scheme:
Step A: 3- [6- [4- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -1-
piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a mixture of 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (20 mg, 39.02 umol) and 1- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde (20 mg, 56.28 umol) in DMF (2 mL) was added TEA (56.95 mg, 562.77 umol) and the reaction was stirred at 25 ℃ for 0.5 hour. Then NaBH (OAc) 3 (35.7 mg, 168.83 umol) was added and stirred for 16 hours. The reaction mixture was purified by prep-HPLC (column: Phenomenex C18 80*40mm*3um; mobile phase: [water (NH3H2O+NH4HCO3) -ACN] ; B%: 52%-82%, 8min) : 60 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 852.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 10.97 (s, 1H) , 10.20-10.05 (m, 1H) , 8.81 (s, 1H) , 7.95-7.89 (m, 1H) , 7.69 (d, J = 8.4 Hz, 1H) , 7.62-7.54 (m, 2H) , 7.44-7.39 (m, 1H) , 7.28-7.22 (m, 1H) , 7.18-7.12 (m, 1H) , 6.92 (d, J = 8.4 Hz, 2H) , 5.73-5.60 (m, 1H) , 5.11-5.05 (m, 2H) , 4.99 (d, J = 10.4 Hz, 1H) , 4.85 (d, J = 17.6 Hz, 1H) , 4.78-4.70 (m, 1H) , 4.61-4.50 (m, 1H) , 4.37-4.29 (m, 1H) , 4.23-4.16 (m, 1H) , 3.79-3.72 (m, 2H) , 3.12-3.08 (m, 4H) , 3.01-2.88 (m, 2H) , 2.81-2.64 (m, 4H) , 2.40-2.30 (m, 2H) , 2.24-2.16 (m, 4H) , 2.05-1.92 (m, 3H) , 1.91-1.77 (m, 1H) , 1.74-1.67 (m, 2H) , 1.28-1.21 (m, 2H) , 0.86 (t, J = 7.2 Hz, 3H)
Example 59: 3- [6- [3- [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-yl] amino] phenyl] piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-059)
Cpd-059 was synthesized according to the following scheme:
Step A: tert-butyl 3-oxo-8-azaspiro [4.5] decane-8-carboxylate
A solution of KOH (2.34 g, 7.5 mL, 30%w/w) was added to a solution of tert-butyl 2-oxo-7-azaspiro [3.5] nonane-7-carboxylate (3 g, 1 eq) in MeOH (37 mL) and THF (37 mL) . N, 4-dimethyl-N-nitroso-benzenesulfonamide (6.98 g, 80% purity, 2.08 eq) was dissolved in H2O (114 mL) and the mixture was added drop wise to the above reaction solution at 0 ℃. The resulting mixture was warmed to 25 ℃ and stirred for 16 hours. The reaction mixture was quenched with aq. AcOH (1 M) (90 mL) at room temperature and extracted with ethyl acetate (100 mL × 2) . The combined organic phases was washed with water (100 mL) and brine (100 mL) , dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=100/1 to 15/85) to afford the title compound.
1H NMR (400 MHz, CD3OD) δ: 3.60 (t, J = 13.6, 5.2 Hz, 2H) , 3.18–3.30 (m, 2H) , 2.30 (t, J = 8.0 Hz, 2H) , 2.20 (s, 2H) , 1.90 (t, J = 8.0 Hz, 2H) , 1.54 (t, J = 5.6 Hz, 4H) , 1.46 (s, 9H) .
Step B: tert-butyl 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane-8-carboxylate
To a mixture of 1- (4-nitrophenyl) piperazine (4.79 g, 23.09 mmol) and tert-butyl 3-oxo-8-azaspiro [4.5] decane-8-carboxylate (4.5 g, 17.76 mmol) in DCM (80 mL) were added AcOH (1.07 g, , 17.76 mmol) and NaBH (OAc) 3 (11.29 g, , 53.29 mmol) . The mixture was stirred at 25 ℃ for 4 hours. The reaction mixture was quenched with H2O (300 mL) and extracted with DCM (100 mL × 3) . The combined organic phases was dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was triturated with Petroleum ether (20 mL) and Ethyl acetate (20 mL) at 25 ℃ for 16 h, filtered to afford the title compound.
LCMS: MS (ESI) m/z = 445.3 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.13 (d, J = 9.2 Hz, 2H) , 6.82 (d, J = 9.6 Hz, 2H) , 3.44 (t, J = 5.2 Hz, 4H) , 3.30-3.41 (m, 4H) , 2.63 (s, 5H) , 1.82-2.00 (m, 2H) , 1.75 (s, 1H) , 1.49-1.64 (m, 4H) , 1.46 (s, 9H) , 1.40-1.44 (m, 2H) , 1.31-1.39 (m, 1H) .
Step C: 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane
To a mixture of tert-butyl 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane-8-carboxylate (2 g) in HCl/MeOH (40 mL, 4 M) was stirred at 25 ℃ for 2 hours. The reaction mixture was concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 367.1 [M+Na] +.
1H NMR (400 MHz, CDCl3) δ 8.13 (d, J = 9.6 Hz, 2H) , 6.82 (d, J = 9.6 Hz, 2H) , 3.38-3.48 (m, 4H) , 2.74-2.84 (m, 4H) , 2.56-2.68 (m, 5H) , 1.84-1.99 (m, 2H) , 1.51-1.68 (m, 4H) , 1.39-1.48 (m, 3H) , 1.32-1.36 (m, 1H) .
Step D: 2- (2, 6-dioxo-3-piperidyl) -5- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-
azaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione
To a mixture of 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane (1 g) and 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (882.11 mg, , 3.19 mmol) in DMSO (20 mL) was added DIPEA (375.21 mg, 505.67 μL, 2.90 mmol) . The mixture was stirred at 80 ℃ for 1 hour. The mixture was allowed to cool to room temperature, diluted with H2O (60 mL) , and extracted with EtOAc (3 × 50 mL) . The combined organic phases were washed with brine (100 mL × 5) , dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was triturated with Ethyl acetate (20 mL) , Petroleum ether (5 mL) at 25 ℃ for 30 min, filtered to afford the title compound.
LCMS: MS (ESI) m/z = 601.3 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.13 (d, J = 9.6 Hz, 2H) , 7.68 (d, J = 8.4 Hz, 1H) , 7.29 (d, J = 2.0 Hz, 1H) , 7.05 (d, J = 8.8 Hz, 1H) , 6.83 (d, J = 9.2 Hz, 2H) , 4.94 (dd, J = 12.4, 5.2 Hz, 1H) , 3.44 (d, J = 4.0 Hz, 8H) , 2.62-2.67 (m, 4H) , 2.09-2.18 (m, 1H) , 1.97-2.04 (m, 1H) , 1.92 (dd, J = 12.4, 7.2 Hz, 1H) , 1.55-1.77 (m, 11H) , 1.45 (t, J = 10.4 Hz, 1H) .
Step E: 3- [6- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-
isoindolin-2-yl] piperidine-2, 6-dione and 3- [5- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-
azaspiro [4.5] decan-8-yl] -1-hydroxy-3-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a mixture of 2- (2, 6-dioxo-3-piperidyl) -5- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione (570 mg) in TFA (5.7 mL) was added Zn (1.49 g, 22.79 mmol) . The mixture was stirred at 75 ℃ for 2 hours. The reaction mixture was filtered. The residue washed with EtOAc (50 mL) , concentrated under reduced pressure to give crude product. The crude product was purified by reversed-phase column (C18, H2O/MeCN=1/0 to 0/1, 0.1%TFA condition) , freeze drying to give 3- [6- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione and 3- [5- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -1-hydroxy-3-oxo-isoindolin-2-yl] piperidine-2, 6-dione.
3- [6- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione:
LCMS: MS (ESI) m/z = 557.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 10.98 (s, 1H) , 7.43 (d, J = 8.0 Hz, 1H) , 7.29 (d, J = 8.4 Hz, 1H) , 7.18 (d, J = 11.2 Hz, 3H) , 7.07 (d, J = 8.8 Hz, 2H) , 5.09 (dd, J = 13.2, 5.2 Hz, 1H) , 4.31-4.36 (m, 1H) , 4.20 (d, J = 16.8 Hz, 1H) , 3.80 (s, 2H) , 3.61 (s, 2H) , 3.19-3.31 (m, 6H) , 3.12 (s, 3H) , 2.86 (s, 1H) , 2.61 (s, 2H) , 2.39 (s, 1H) , 1.88-2.02 (m, 2H) , 1.48-1.75 (m, 8H) .
3- [5- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -1-hydroxy-3-oxo-isoindolin-2-yl] piperidine-2, 6-dione
LCMS: MS (ESI) m/z = 557.2 [M+H] +.
Step F: 3- [6- [3- [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-
yl] amino] phenyl] piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
To a mixture of 3- [6- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (80 mg) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (45 mg) in n-BuOH (2 mL) was heated at 100 ℃ for 2 hrs. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: Phenomenex C18 75*30mm*3um; mobile phase: [water (FA) -ACN] ; B%:8%-48%, 9 min) to afford the title compound.
LCMS: MS (ESI) m/z = 878.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.97 (s, 1H) , 10.03 (br s, 1H) , 8.82 (s, 1H) , 8.15 (FA, 0.837H) , 8.01 (t, J = 7.6 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.57 (t, J = 7.6 Hz, 3H) , 7.41 (d, J =8.4 Hz, 1H) , 7.25 (d, J = 8.8 Hz, 1H) , 7.16 (d, J = 2.0 Hz, 1H) , 6.90 (d, J = 8.8 Hz, 2H) , 5.29-5.44 (m, 2H) , 5.27 (s, 1H) , 5.09 (dd, J = 13.2, 5.2 Hz, 1H) , 4.42-4.50 (m, 2H) , 4.17-4.36 (m, 2H) , 3.20 (s, 4H) , 3.09 (s, 4H) , 2.86-2.96 (m, 1H) , 2.61-2.63 (m, 1H) , 2.54-2.57 (m, 4H) , 2.43-2.46 (m, 1H) , 2.35-2.41 (m, 1H) , 2.10-2.27 (m, 1H) , 1.81-2.04 (m, 5H) , 1.69-1.75 (m, 1H) , 1.47-1.65 (m, 10H) , 1.29-1.35 (m, 1H) .
Example 60: 3- [5- [3- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-yl] amino] phenyl] piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-060)
Cpd-060 was synthesized according to the following scheme:
Step A: tert-butyl 3- (methoxymethylene) -8-azaspiro [4.5] decane-8-carboxylate
To a solution of methoxymethyl (triphenyl) phosphonium chloride (21.38 g, 62.37 mmol) in THF (200 mL) was added t-BuOK (7.00 g, 62.37 mmol) . The mixture was stirred at 0℃ for 1 hr. Then tert-butyl 3-oxo-8-azaspiro [4.5] decane-8-carboxylate (7.9 g, 31.18 mmol) was added and the mixture was stirred at 25℃ for 1 hr. The reaction was quenched with saturated aqueous NH4Cl solution (100 mL) . The resulting aqueous residue was extracted with EtOAc (200 mL ×3) . The organic layer was washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated in vacuo. The residue was purified by flash silica gel chromatography (80
gSilica Flash Column, Eluent of 0~10%Ethyl acetate/Petroleum ether gradient @80 mL/min) to afford the title compound.
1H NMR (400 MHz, CDCl3) δ: 5.89 (d, J = 2.0 Hz, 1H) , 3.56 (s, 3H) , 3.41–3.54 (m, 2H) , 3.17–3.30 (m, 2H) , 2.23–2.34 (m, 2H) , 2.17 (s, 1H) , 2.08 (s, 1H) , 1.50–1.56 (m, 2H) , 1.46 (s, 9H) , 1.41 (q, J = 5.6 Hz, 4H) .
Step B: tert-butyl 3-formyl-8-azaspiro [4.5] decane-8-carboxylate
To a solution of tert-butyl 3- (methoxymethylene) -8-azaspiro [4.5] decane-8-carboxylate (3.7 g, 13.15 mmol) in MeCN (120 mL) was added water (3.2 mL) and TFA (2 mL) . The mixture was stirred at 25℃ for 16 hr. The reaction was quenched with saturated aqueous NaHCO3 solution (20 mL) under ice cooling and organic solvent was removed in vacuo. The resulting aqueous residue was extracted with EtOAc (100 mL × 3) . The organic layer was washed with brine (100 mL) , dried over anhydrous Na2SO4, filtered and concentrated in vacuo to afford the title compound.
1H NMR (400 MHz, CD3OD) δ: 9.60 (d, J = 2.0 Hz, 1H) , 3.39 (d, J = 2.8 Hz, 4H) , 2.11–2.23 (m, 1H) , 1.91–1.97 (m, 2H) , 1.75 (d, J = 8.4 Hz, 2H) , 1.58–1.63 (m, 2H) , 1.48–1.54 (m, 4H) , 1.45 (s, 9H) .
Step C: tert-butyl 3- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decane-8-
carboxylate
To solution of tert-butyl 3-formyl-8-azaspiro [4.5] decane-8-carboxylate (2 g) and 1- (4-nitrophenyl) piperazine (1.71 g, 8.23 mmol) in DCM (20 mL) was added TEA (3.78 g, 37.40 mmol, 5.21 mL) . The mixture was stirred at 20 ℃ for 1 hr. NaBH (OAc) 3 (1.90 g, 8.98 mmol) was added and the mixture was stirred at 20 ℃ for 2 hrs. The mixture was diluted with water (20 mL) and extracted with EtOAc (100 mL × 3) . The combined organic layers was dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by flash silica gel chromatography (20 gSilica Flash Column, Eluent of 0~5% MeOH/DCM gradient @30 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 459.3 [M+H] +.
1H NMR (400 MHz, CDCl3) δ: 8.10–8.15 (m, 2H) , 6.82 (d, J = 9.6 Hz, 2H) , 3.40–3.46 (m, 4H) , 3.33–3.39 (m, 4H) , 2.53–2.61 (m, 4H) , 2.35 (d, J = 6.4 Hz, 2H) , 2.18–2.29 (m, 1H) , 1.82–1.90 (m, 1H) , 1.77 (dd, J = 12.8, 7.6 Hz, 1H) , 1.58–1.68 (m, 2H) , 1.49–1.55 (m, 2H) , 1.46 (s, 9H) , 1.39–1.42 (m, 2H) , 1.31–1.37 (m, 2H) .
Step D: 3- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decane
To a solution of tert-butyl 3- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decane-8-carboxylate (2.76 g, 6.02 mmol) in DCM (20 mL) was added HCl/dioxane (4N, 20 mL) . The mixture was stirred at 25 ℃ for 1 hr. The reaction mixture was concentrated under reduced
pressure to give a residue. Then the residue was dissolved in water (30 mL) , and washed with DCM (30 mL × 3) . The aqueous phases were adjusted to pH = 10 with sat. aq. Na2CO3. Then the mixture was extracted with DCM (50 mL × 3) . The combined organic layers was washed with brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 359.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 8.04 (d, J = 9.2 Hz, 2H) , 7.01 (d, J = 9.6 Hz, 2H) , 3.40-3.49 (m, 4H) , 3.30 (br s, 5H) , 2.61-2.66 (m, 3H) , 2.43-2.47 (m, 3H) , 2.14-2.29 (m, 3H) , 1.64-1.79 (m, 2H) , 1.38-1.46 (m, 2H) , 1.28-1.36 (m, 4H) , 1.18-1.27 (m, 2H) .
Step E: 2- (2, 6-dioxo-3-piperidyl) -5- [3- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -8-
azaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione
The mixture of 3- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decane (1.7 g) , 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (2.38 g, 8.61 mmol) in DMSO (20 mL) was added DIPEA (556.31 mg, 4.30 mmol, 749.75 μL) . The mixture was stirred at 80 ℃ for 2 h under N2. The ice-cooled water (40 mL) was added and the mixture extracted with DCM (200 mL × 3) . The combined organic layers was washed with brine (100 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (40 gSilica Flash Column, Eluent of 0~5%Dichloromethane : Methanol gradient @40 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 615.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 11.05 (s, 1H) , 7.98–8.06 (m, 2H) , 7.61 (d, J = 8.4 Hz, 1H) , 7.28 (d, J = 2.0 Hz, 1H) , 7.19 (dd, J = 8.8, 2.0 Hz, 1H) , 6.98 (d, J = 9.6 Hz, 2H) , 5.03 (dd, J = 12.8, 5.6 Hz, 1H) , 3.36–3.48 (m, 8H) , 2.77–2.92 (m, 2H) , 2.51 (s, 3H) , 2.17–2.30 (m, 3H) , 1.97–2.09 (m, 1H) , 1.67–1.83 (m, 2H) , 1.40–1.54 (m, 7H) , 1.22–1.34 (m, 1H) , 0.97–1.09 (m, 1H) .
Step F: 3- [5- [3- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -3-
hydroxy-1-oxo-isoindolin-2-yl] piperidine-2, 6-dione and 3- [5- [3- [ [4- (4-
aminophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -1-hydroxy-3-oxo-
isoindolin-2-yl] piperidine-2, 6-dione
To a mixture of 2- (2, 6-dioxo-3-piperidyl) -5- [3- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione (1.1 g, 1.79 mmol) in dioxane (15 mL) was added AcOH (1.5 mL) , H2O (1.5 mL) , Aluminum (1.45 g, 53.69 mmol, 536.46 μL) and HgCl2 (130 mg, 478.82 μmol) in one portion at 25 ℃. The mixture was stirred at 80 ℃ for 2 hours. The reaction mixture was filtered. The residue washed with acetone (100 mL ×2) , the filtrate was concentrated under reduced pressure to give 3- [5- [3- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -3-hydroxy-1-oxo-isoindolin-2-yl] piperidine-2, 6-dione and 3- [5- [3-
[ [4- (4-aminophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -1-hydroxy-3-oxo-isoindolin-2-yl] piperidine-2, 6-dione
Step G: 3- [5- [3- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -1-
oxo-isoindolin-2-yl] piperidine-2, 6-dione
The mixture of 3- [5- [3- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -1-hydroxy-3-oxo-isoindolin-2-yl] piperidine-2, 6-dione and 3- [5- [3- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -3-hydroxy-1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (1.1 g) in DCM (10 mL) was added triethylsilane (6.55 g, 56.35 mmol, 9 mL) , TFA (13.82 g, 121.16 mmol, 9 mL) . The mixture was stirred at 25 ℃ for 16 hrs. The reaction mixture was filtered and concentrated under reduced pressure to give a residue, which was purified by reversed-phase column (C18, H2O/MeCN=1/0 to 0/1, 0.1% TFA condition) to afford the title compound.
LCMS: MS (ESI) m/z = 571.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ = 10.95 (s, 1H) , 9.52 (br s, 2H) , 7.51 (d, J = 8.4 Hz, 1H) , 7.21 (d, J = 5.6 Hz, 2H) , 7.06–7.09 (m, 4H) , 5.04 (dd, J = 13.2, 4.8 Hz, 1H) , 4.28–4.35 (m, 1H) , 4.14–4.23 (m, 1H) , 3.30-3.36 (m, 5H) , 3.16-3.22 (m, 4H) , 2.95-3.11 (m, 4H) , 2.84-2.90 (m, 1H) , 2.57-2.59 (m, 1H) , 2.28-2.43 (m, 2H) , 1.86-1.99 (m, 4H) , 1.3-1.58 (m, 6H) , 1.36-1.44 (m, 1H) , 1.12-1.18 (m, 1H) .
Step H: 3- [5- [3- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
yl] amino] phenyl] piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
A solution of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (120 mg, 298.92 μmol) and 3- [5- [3- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (120 mg) in n-BuOH (2 mL) was stirred at 100 ℃ for 2 hrs. The residue was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (TFA) -ACN] ; B%: 23%-43%, 16 min) to afford the title compound.
LCMS: MS (ESI) m/z = 892.8 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 10.95 (s, 1H) , 10.08 (s, 1H) , 9.30 (s, 1H) , 8.85 (s, 1H) , 8.00 (t, J = 7.6 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.58–7.66 (m, 3H) , 7.52 (d, J = 8.4 Hz, 1H) , 7.04–7.09 (m, 2H) , 6.99 (d, J = 8.8 Hz, 2H) , 5.35 (br s, 2H) , 5.04 (dd, J = 13.2, 5.2 Hz, 1H) , 4.48 (br s, 2H) , 4.17–4.36 (m, 2H) , , 3.76 (d, J = 11.6 Hz, 2H) , 3.29–3.37 (m, 5H) , 3.15–3.22 (m, 3H) , 2.95–3.06 (m, 2H) , 2.89 (d, J = 12.0 Hz, 1H) , 2.61 (s, 1H) , 2.55–2.58 (m, 2H) , 2.30–2.36 (m,
1H) , 2.14–2.21 (m, 1H) , 1.83–2.12 (m, 6H) , 1.68–1.75 (m, 1H) , 1.51–1.64 (m, 10H) , 1.34–1.47 (m, 1H) , 1.12–1.19 (m, 1H)
Example 61: 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [4- [ [3- [4-methoxy-3- (6-oxo-1H-pyridazin-3-yl) benzoyl] -3-azaspiro [5.5] undecan-9-yl] methyl] piperazin-1-yl] anilino] pyrazolo [3, 4-d] pyrimidin-3-one (Cpd-061)
Cpd-061 was synthesized according to the following scheme:
Step A: methyl 4-methoxy-3- (6-oxo-1H-pyridazin-3-yl) benzoate
A Schlenk tube charged with a mixture of (2-methoxy-5-methoxycarbonyl-phenyl) boronic acid (1 g, 4.76 mmol) , 3-bromo-1H-pyridazin-6-one (999.95 mg, 5.71 mmol) , Pd (dppf) Cl2 (348.45 mg, 476.21 umol) , K2CO3 (1.97 g, 14.29 mmol) in dioxane (10 mL) and H2O (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90 ℃ for 2 hr
under N2 atmosphere. The mixture was poured into water (40 mL) and extracted with EtOAc (40 mL × 3) . The combined organic layers was washed with brine (30 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by flash silica gel chromatography (40 gSilica Flash Column, Eluent of 0~10%Methanol/Dichloromethane gradient @100 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 261.1 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 13.25 (s, 1H) , 8.00–8.11 (m, 2H) , 7.76 (d, J = 10.0 Hz, 1H) , 7.29 (d, J = 8.8 Hz, 1H) , 6.92 (dd, J = 10.0, 1.6 Hz, 1H) , 3.92 (s, 3H) , 3.84 (s, 3H)
Step B: 4-methoxy-3- (6-oxo-1H-pyridazin-3-yl) benzoic acid
To a solution of methyl 4-methoxy-3- (6-oxo-1H-pyridazin-3-yl) benzoate (300 mg, 1.15 mmol) in a mixture of THF (2 mL) , H2O (1 mL) and MeOH (2 mL) was added LiOH. H2O (241.87 mg, 5.76 mmol) . The mixture was stirred at 25 ℃ for 2 hrs. The pH value was adjusted to 5 by 3 M aq.HCl, the mixture was lyophilization to afford the title compound.
LCMS: MS (ESI) m/z = 247.1 [M+H] +.
Step C: 3-azaspiro [5.5] undecane-9-carbaldehyde
To a solution of tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (20 mg) in a mixture of DCM (1 mL) was added TFA (1 mL) . The mixture was stirred at 25 ℃ for 2 hr. The mixture was concentrated and lyophilized to afford the title compound.
1H NMR (400 MHz, DMSO-d6) δ: 9.57 (s, 1H) , 2.23–2.32 (m, 1H) , 1.65–1.74 (m, 4H) , 1.52-1.62 (m, 6H) , 1.36–1.51 (m, 6H) .
Step D: 3- [4-methoxy-3- (6-oxo-1H-pyridazin-3-yl) benzoyl] -3-azaspiro [5.5] undecane-9-
carbaldehyde
To a solution of 4-methoxy-3- (6-oxo-1H-pyridazin-3-yl) benzoic acid (81.50 mg) and 3-azaspiro [5.5] undecane-9-carbaldehyde (20 mg) in DCM (3 mL) were added EDCI (42.30 mg, 220.66 umol) , DIEA (71.30 mg, 551.65 umol, 96.09 uL) and HOBt (29.82 mg, 220.66 umol) . The mixture was stirred at 25 ℃ for 6 hr under N2 atmosphere. The mixture was poured into water (40 mL) and extracted with CH2Cl2 (40 mL × 3) . The combined organic layers was washed with brine (30 mL × 2) , dried over Na2SO4, filtered and concentrated to afford the title compound.
LCMS: MS (ESI) m/z = 410.2 [M+H] +.
Step F: 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- [4- [4-
[ [3- [4-methoxy-3- (6-oxo-1H-pyridazin-3-yl) benzoyl] -3-azaspiro [5.5] undecan-9-
yl] methyl] piperazin-1-yl] anilino] pyrazolo [3, 4-d] pyrimidin-3-one
To a solution of 3- [4-methoxy-3- (6-oxo-1H-pyridazin-3-yl) benzoyl] -3-azaspiro [5.5] undecane-9-carbaldehyde (19.17 mg) and 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one
(20 mg, 39.02 umol) in DCM (3 mL) were added NaBH (OAc) 3 (41.35 mg, 195.08 umol) and Et3N (19.74 mg, 195.08 umol, 27.15 uL) . The mixture was stirred at 25 ℃ for 2 hrs under N2 atmosphere. The mixture was poured into water (40 mL) and extracted with DCM (40 mL × 3) . The combined organic layers was dried over anhydrous sodium sulfate, filtered and concentrated to give a residue, which was purified by prep-HPLC (column: C18-1 150*30mm*5um; mobile phase: [water (FA) -ACN] ; B%: 3%-43%, 9min) to afford the title compound.
LCMS: MS (ESI) m/z = 906.4 [M+H] +.
1H NMR (400 MHz, CD3OD) δ: 8.74–8.82 (m, 1H) , 7.89–7.94 (m, 1H) , 7.83–7.87 (m, 1H) , 7.70–7.75 (m, 1H) , 7.63 (d, J = 2.0 Hz, 1H) , 7.54 (d, J = 8.4 Hz, 3H) , 7.21 (d, J = 8.8 Hz, 1H) , 6.93–7.00 (m, 3H) , 5.63 (s, 1H) , 5.01–5.06 (m, 2H) , 4.57–4.61 (m, 4H) , 3.85–4.03 (m, 3H) , 3.65–3.77 (m, 2H) , 3.39–3.53 (m, 2H) , 3.19 (s, 4H) , 2.98–3.12 (m, 1H) , 2.78–2.93 (m, 1H) , 2.66 (s, 4H) , 2.32–2.41 (m, 3H) , 2.09–2.21 (m, 1H) , 1.93-2.07 (m, 1H) , 1.77–1.89 (m, 4H) , 1.61–1.71 (m, 4H) , 1.12–1.26 (m, 4H) , 0.92–0.97 (m, 3H) .
Example 62: (R) -1- (5- (9- (2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) ethyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-062)
Cpd-062 was synthesized according to the following scheme:
Step A: tert-butyl 9- (2-ethoxy-2-oxoethylidene) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9-oxo-3-azaspiro [5.5] undecane-3-carboxylate (2.00 g, 7.48 mmol) in DMF (10 mL) was added ethyl 2- (diethoxyphosphoryl) acetate (2.51 g, 11.2 mmol) and NaH (60%, 0.600 g, 15.0 mmol) at 0℃. Then the reaction was stirred at 0℃ for 3 h under N2 atmosphere. After completion of reaction, the mixture was concentrated. The residue was diluted with water (20 mL) , extracted with EA (3*10 mL) . The combined organic layers was washed with water (10 mL) and brine (10 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column (DCM: MeOH =10: 1) to afford the title compound.
1H NMR (400 MHz, CDCl3) δ 5.63 (s, 1H) , 4.21-4.11 (m, 4H) , 3.42 – 3.34 (m, 4H) , 2.88 –2.80 (m, 2H) , 2.28 – 2.17 (m, 2H) , 1.57-1.52 (m, 4H) , 1.48-1.40 (m, 11H) , 1.27 (t, J = 5.5 Hz, 3H) .
Step B: tert-butyl 9- (2-ethoxy-2-oxoethyl) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9- (2-ethoxy-2-oxoethylidene) -3-azaspiro [5.5] undecane-3-carboxylate (220 mg, 0.652 mmol) in MeOH (10 mL) was added Pd/C (10.0 mg, 10% w/w) and stirred at 35℃ for 2 hrs under H2 atmosphere. The reaction was filtered and concentrated to afford the title compound.
TLC: Rf =0.5 (PE: EA =3: 1) .
Step C: tert-butyl 9- (2-hydroxyethyl) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9- (2-ethoxy-2-oxoethyl) -3-azaspiro [5.5] undecane-3-carboxylate (1.30 g) in THF (10 mL) was added LiBH4 (0.334 g, 15.3 mmol) and the reaction was stirred at 0℃for 2 hrs under N2 atmosphere. After completion, the mixture was quenched with water (20 mL) , extracted with EA (3*10 mL) . The combined organic layers was washed with brine (5.0 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
TLC: Rf =0.2 (DCM: MeOH =9: 1) .
Step D: tert-butyl 9- (2-oxoethyl) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9- (2-hydroxyethyl) -3-azaspiro [5.5] undecane-3-carboxylate (500 mg, 1.68 mmol) in DCM (5.0 mL) was added Dess-Martin reagent (1.50 g, 3.36 mmol) and the reaction mixture was stirred at 35℃ under N2 atmosphere for 3 h. After completion, the mixture was concentrated. The residue was diluted with water 20 mL) , extracted with DCM (3*20 mL) . The organic layers were dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (PE: EA =3: 1) to afford the title compound.
1H NMR (400 MHz, CDCl3) δ 9.73 (s, 1H) , 3.41-3.31 (m, 4H) , 2.38 – 2.29 (m, 1H) , 1.97 – 1.83 (m, 1H) , 1.72 – 1.63 (m, 2H) , 1.62 – 1.47 (m, 3H) , 1.45 (s, 9H) , 1.34 – 1.28 (m, 2H) , 1.26 (t, J = 7.2 Hz, 2H) , 1.20 (s, 1H) , 1.17 (d, J = 9.3 Hz, 3H) .
Step E: tert-butyl (R) -9- (2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) ethyl) -3-azaspiro [5.5] undecane-3-carboxylate
A mixture of tert-butyl 9- (2-oxoethyl) -3-azaspiro [5.5] undecane-3-carboxylate (163 mg, 0.553 mmol) , (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- ( (4- (piperazin-1-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (283 mg, 0.553 mmol) and NaBH (OAc) 3 (351 mg, 1.66 mmol) in DCM (5.0 mL) /MeOH (5.0 mL) was added HOAc (33.0 mg 0.553 mmol) and the mixture was stirred at 0℃ for 2 h. After completion, the reaction was quenched with water (10 mL) , extracted with EA (3*10 mL) . The organic layers were washed with brine (5.0 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 792.5
Step F: (R) -6- ( (4- (4- (2- (3-azaspiro [5.5] undecan-9-yl) ethyl) piperazin-1-yl) phenyl) amino) -2-
allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-
pyrazolo [3, 4-d] pyrimidin-3-one
To a solution of tert-butyl (R) -9- (2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) ethyl) -3-azaspiro [5.5] undecane-3-carboxylate (185 mg, 0.234 mmol) in THF (5 mL) was added 1M aq. HCl (12 mL) . The mixture was stirred at 35℃ for 1 h. The mixture was quenched by the addition of 10 mL water. The resulting solution was extracted with EtOAc (3*10 mL) , the organic layers was combined and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 692.4
Step G: (R) -1- (5- (9- (2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) ethyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-
methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -6- ( (4- (4- (2- (3-azaspiro [5.5] undecan-9-yl) ethyl) piperazin-1-yl) phenyl) amino) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (113 mg, 0.164 mmol) in DMF (5.0 mL) were added perfluorophenyl 3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) -4-methoxybenzoate (105 mg, 0.246 mmol) , DIEA (63.0 mg, 0.492 mmol) . The mixture was stirred at r. t. for 2 hrs. After completion of reaction, the mixture was concentrated. The residue was diluted with water (5.0 mL) , extracted with EA (3*10 mL) . The organic layers were washed with water (3*5 mL) and brine (5 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography to afford the title compound.
LC-MS (ESI) [M+H] + 938.3.
1H NMR (400 MHz, DMSO) δ 10.32 (s, 1H) , 10.12 (s, 1H) , 8.81 (s, 1H) , 8.37 (s, 1H) , 7.91 (s, 1H) , 7.69 (d, J = 8.2 Hz, 1H) , 7.58 (s, 2H) , 7.36 (d, J = 8.6 Hz, 1H) , 7.32 (s, 1H) , 7.15 (d, J = 8.5 Hz, 1H) , 6.91 (d, J = 8.8 Hz, 2H) , 5.67 (dd, J = 16.6, 10.7 Hz, 1H) , 5.04 (s, 1H) , 4.99 (d, J = 10.4 Hz, 1H) , 4.85 (d, J = 17.0 Hz, 1H) , 4.80-4.65 (m, 1H) , 4.60-4.55 (m, 1H) , 3.84 (s, 3H) , 3.59 (t, J = 6.8 Hz, 2H) , 3.52-3.36 (m, 4H) , 3.26 – 3.19 (m, 1H) , 3.08 (s, 4H) , 3.0-2.90 (m, 1H) , 2.80-2.71 (m, 1H) , 2.70-2.55 (m, 2H) , 2.47 – 2.43 (m, 1H) , 2.33 (s, 3H) , 2.25-2.18 (m, 1H) , 2.10-1.98 (m, 1H) , 1.95-1.77 (m, 1H) , 1.73-1.65 (m, 3H) , 1.64-1.41 (m, 4H) , 1.40-1.32 (m, 2H) , 1.32-1.25 (m, 4H) , 1.24-1.02 (m, 4H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 63: (R) -1- (5- (8- ( (1- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperidin-4-yl) methyl) -2, 8-diazaspiro [4.5] decane-2-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-063)
Cpd-063 was synthesized according to the following scheme:
Step A: tert-butyl 8- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 8-diazaspiro [4.5] decane-2-
carboxylate
A solution of 1- (4-nitrophenyl) piperidine-4-carbaldehyde (200 mg, 0.855 mmol) , tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (205 mg 0.855 mmol) , NaBH (OAc) 3 (544 mg 2.57 mmol) , HOAc (50.0 mg 0.855 mmol) in DCM (5.0 mL) and MeOH (5 mL) was stirred at 35℃ for 12 hrs. After completion, the reaction was quenched with water (10 mL) , extracted with EtOAc (3*10 mL) . The combined organic layers was washed with brine (10 mL) , dried with
Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
LC-MS (ESI) [M+H] +. 459.4
Step B: 8- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 8-diazaspiro [4.5] decane
To a solution of tert-butyl 8- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 8-diazaspiro [4.5] decane-2-carboxylate (307 mg, 0.670 mmol) in DCM (5.0 mL) was added HCl in dioxane (4.0 mL) and stirred at 35℃ for 1 h. The mixture was concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 359.3
Step C: 1- (2-chloro-5- (8- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 8-
diazaspiro [4.5] decane-2-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 8- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 8-diazaspiro [4.5] decane (328 mg) in DMF (5.0 mL) was added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (246 mg, 0.946 mmol) , DIEA (355 mg, 2.75 mmol) . The mixture was stirred at r. t. for 2 hrs. After completion, the reaction mixture was quenched with water (10 mL) , extracted with EA (3*20 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 609.1
Step D: 1- (5- (8- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -2, 8-diazaspiro [4.5] decane-2-
carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (8- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 8-diazaspiro [4.5] decane-2-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (115 mg, 0.189 mmol) in EtOH (10 mL) and H2O (3.0 mL) were added NH4Cl (51.0 mg, 0.945 mmol) , Fe (53.0 mg 0.945 mmol) . The mixture was stirred at 90℃ for 2 hrs. After completion of reaction, The mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was added Η20 (30 mL) and extracted by EtOAc (20 mL x 3) . The combined organic phase was washed with brine, dried over anhydrous Na2S04 and the organic layer was concentrated to give a residue, which was used in next steps without further purifications.
LC-MS (ESI) [M+H] + 579.3
Step E: (R) -1- (5- (8- ( (1- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperidin-4-yl) methyl) -2, 8-diazaspiro [4.5] decane-2-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylthio) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (100 mg, 0.261 mmol) in DCM (10 mL) was added m-CPBA (58.0 mg, 0.287 mmol, 85%purity) . The mixture was stirred at 25℃ for 2 h. 1- (5- (8- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -2, 8-diazaspiro [4.5] decane-2-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (135 mg) and DIPEA (101 mg, 0.783 mmol) were added and the mixture was stirred at 25℃ for 12 hrs. After completion, the reaction was quenched with water (50 mL) , the resulting solution was extracted with DCM (2*100 mL) , the organic layers were combined and concentrated to get the crude. The crude was purified by Pre-HPLC (Waters 2767, Column: Pursuit XRs C18, 21.2*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 22%~22%; Retention Time: 7.2-8.8min of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 914.2.
1H NMR (400 MHz, DMSO) δ 10.50 (s, 1H) , 10.17 – 9.96 (m, 1H) , 8.81 (s, 1H) , 8.31 (s, 1H) , 7.91 (s, 1H) , 7.73 –7.61 (m, 3H) , 7.55 (s, 3H) , 6.90 (s, 2H) , 5.66 (s, 1H) , 5.08 – 4.96 (m, 2H) , 4.86 (d, J = 16.2 Hz, 1H) , 4.77 – 4.69 (m, 1H) , 4.59 – 4.50 (m, 1H) , 3.76 (s, 1H) , 3.61 (s, 3H) , 3.51 (d, J = 19.6 Hz, 3H) , 2.95 (s, 2H) , 2.75 (s, 3H) , 2.68 – 2.53 (m, 3H) , 2.46 – 2.40 (m, 1H) , 2.33 (s, 2H) , 2.16 (d, J = 26.5 Hz, 4H) , 2.05 – 1.97 (m, 1H) , 1.88 (s, 1H) , 1.75 (d, J = 16.1 Hz, 5H) , 1.58 (s, 2H) , 1.46 (s, 2H) , 1.19 (s, 2H) , 0.87 (t, J = 7.6 Hz, 3H) .
Example 64: 1- (5- (9- ( ( (S) -4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) -2-methylpiperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-064)
Cpd-064 was synthesized according to the following scheme:
Step A: (S) -3-methyl-1- (4-nitrophenyl) piperazine
To a solution of 1-fluoro-4-nitrobenzene (5.00 g, 35.4 mmol) in DMSO (20 mL) was added (S) -2-methylpiperazine (3.55 g, 35.4 mmol) , N-ethyl-N-isopropylpropan-2-amine (13.7 g, 106 mmol) , the mixture was stirred at 120℃ for 12 hrs under N2 atmosphere. The mixture was quenched by addition of 10 mL water, the reaction mixture was filtered through celite, and the filter cake washed with water (20 mL) . The combined filtrate were dried under reduced pressure to afford the title compound.
1H NMR (400 MHz, DMSO) δ 8.04 (t, J = 6.4 Hz, 2H) , 7.00 (d, J = 9.5 Hz, 2H) , 3.90 – 3.80 (m, 2H) , 2.96 (dd, J = 11.8, 2.2 Hz, 1H) , 2.83 (td, J = 12.0, 3.0 Hz, 1H) , 2.70 (ddd, J = 11.7, 6.5, 2.9 Hz, 2H) , 2.36 (s, 1H) , 1.03 (d, J = 6.3 Hz, 3H) .
Step B: tert-butyl (S) -9- ( (2-methyl-4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carboxylate
To a solution of (S) -3-methyl-1- (4-nitrophenyl) piperazine (120 mg) in DCM (10 mL) were added tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (153 mg, 0.542 mmol) ,
NaBH (OAc) 3 (345 mg, 1.63 mmol) , the mixture was stirred at 25 ℃ for 12 hrs under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3*10 mL) , the organic layers were combined and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =100: 1-0: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 487.3
Step C: (S) -9- ( (2-methyl-4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane
To a solution of tert-butyl (S) -9- ( (2-methyl-4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate (150 mg, 0.308 mmol) in DCM (10 mL) was added 1M HCl/Dioxane (2 mL) at 0 ℃, the mixture was stirred at 20 ℃ for 2 hrs under N2 atmosphere. The mixture was concentrated under reduced pressure to afford the title compound.
LC-MS (ESI) [M+H] + 387.3
Step D: (S) -1- (2-chloro-5- (9- ( (2-methyl-4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (S) -9- ( (2-methyl-4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane (118 mg) in DMF (5 mL) were added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (133 mg, 0.305 mmol) , N-ethyl-N-isopropylpropan-2-amine (118 mg, 0.916 mmol) , the mixture was stirred at 25 ℃ for 2 hrs under N2 atmosphere. The mixture was quenched by the addition of 20 mL water, the resulting solution was extracted with EtOAc (3*10 mL) , the organic layers were combined and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1-1: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 637.3
Step E: (S) -1- (5- (9- ( (4- (4-aminophenyl) -2-methylpiperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (S) -1- (2-chloro-5- (9- ( (2-methyl-4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (150 mg, 0.235 mmol) in EtOH (10 mL) and H2O (1 mL) were added iron powder (65.7 mg, 1.18 mmol) and NH4Cl (63.0 mg, 1.18 mmol) , the mixture was stirred at 90 ℃ for 3 hrs under N2 atmosphere. The reaction mixture was filtered through celite, and the filtrated was concentrated under reduced pressure to afford the title compound.
LC-MS (ESI) [M+H] + 473.3
Step F: 1- (5- (9- ( ( (S) -4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) -2-methylpiperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (S) -1- (5- (9- ( (4- (4-aminophenyl) -2-methylpiperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (100 mg) in DCM (10 mL) were added 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (67.8 mg, 0.165 mmol) and DIEA (63.9 mg, 0.495 mmol) , the mixture was stirred at 25 ℃ for 18 hrs under N2 atmosphere. The mixture was quenched by addition of 10 mL water, the resulting solution was extracted with DCM (3*10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um: Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate 20ml/min; gradient: 22-32%Retention Time: 8.5-10 min) to afford the title compound.
LC-MS (ESI) [M+H] + 942.8
1H NMR (400 MHz, DMSO) δ 10.50 (s, 1H) , 10.08 (s, 1H) , 8.81 (s, 1H) , 8.31 (s, 1H) , 7.91 (s, 1H) , 7.68 (d, J = 8.0 Hz, 1H) , 7.63 (d, J = 8.1 Hz, 1H) , 7.54 (s, 3H) , 7.38 (d, J = 7.7 Hz, 1H) , 6.90 (d, J = 8.1 Hz, 2H) , 5.73 – 5.59 (m, 1H) , 4.99 (d, J = 10.0 Hz, 2H) , 4.85 (d, J = 17.2 Hz, 1H) , 4.73 (s, 1H) , 4.56 (d, J = 10.7 Hz, 1H) , 3.74 (s, 3H) , 3.60 (s, 3H) , 2.96 (dd, J = 14.8, 7.4 Hz, 1H) , 2.89 (d, J = 10.2 Hz, 1H) , 2.77 (d, J = 16.7 Hz, 5H) , 2.46 – 2.28 (m, 2H) , 2.25 – 2.16 (m, 2H) , 2.06 – 1.84 (m, 4H) , 1.69 (s, 4H) , 1.47 (s, 4H) , 1.35 (s, 1H) , 1.25 (s, 1H) , 1.23 (s, 2H) , 1.07 (s, 2H) , 1.03 (s, 4H) , 0.86 (t, J = 6.9 Hz, 3H) .
Example 65: 1- (5- (9- ( ( (R) -4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) -2-methylpiperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-065)
Cpd-065 was synthesized according to the following scheme:
Step A: (R) -3-methyl-1- (4-nitrophenyl) piperazine
To a stirred mixture of 1-fluoro-4-nitrobenzene (7.05 g, 50 mmol) in DMF (40 mL) at 25℃, (R) -2-methylpiperazine (5 g, 50 mmol) and K2CO3 (20.73 g, 150 mmol) were added and stirred at rt for 16 h. The mixture was diluted with water (100 mL) and extracted with EtOAc (3 x 150 mL) . The organic layer was washed with brine solution (50 mL) , dried over anhydrous sodium sulphate, and concentrated to provide crude residue. The crude was triturated with n-hexane and filtered to obtain the title compound.
LC-MS (ESI) [M+H] + 222.2
Step B: tert-butyl (R) -9- ( (2-methyl-4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carboxylate
A mixture of (R) -3-methyl-1- (4-nitrophenyl) piperazine (200 mg) , tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (254.34 mg, 0.90 mmol) , CH3COOH (108.09 mg, 1.80 mmol) and NaBH (OAc) 3 (381.49 mg, 1.80 mmol) in DCM (20 mL) was stirred at 25℃ for 16 hours. The reaction was quenched with an aqueous solution of sat. NaHCO3 (5 mL) and the layers were separated. The aqueous layer was extracted with CH2Cl2 (3 x 5 mL) . The combined organic extracts were dried (MgSO4) and concentrated under reduced pressure. The crude material was purified via flash column chromatography eluting EtOAc: hexanes (1: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 487.4
Step C: (R) -9- ( (2-methyl-4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane
tert-butyl (S) -9- ( (2-methyl-4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate (257 mg, 0.53 mmol) was dissolved in 10 mL of solvent (V1, 4-Dioxane: VDCM =4: 1) , 60 mL of anhydrous 4N HCl in Dioxane was added dropwise in an ice bath. The solution was stirred overnight at room temperature and the solvent was evaporated to afford the title compound.
LC-MS (ESI) [M+H] + 387.3
Step D: (R) -1- (2-chloro-5- (9- ( (2-methyl-4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
A solution of (R) -9- ( (2-methyl-4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane (195.00 mg) , perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (328.65 mg, 0.76 mmol) and DIEA (195.73 mg, 1.51 mmol) in DMF (15 mL) was stirred at room temperature for 2hrs. The solution was diluted with water (10 mL) and extracted with ethyl acetate (20 mL x 3) . The organic layer was dried and concentrated. The residue was purified by silica gel chromatography (eluted with petroleum ether: ethyl acetate =2: 1 to 1: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 637.3
Step E: (R) -1- (5- (9- ( (4- (4-aminophenyl) -2-methylpiperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a stirred solution of (R) -1- (2-chloro-5- (9- ( (2-methyl-4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (250 mg, 0.39 mmol) in EtOH (10 mL) and water (2 mL) was added iron powder (109.72 mg, 1.96 mmol) , followed by ammonium chloride (104.84 mg, 1.96 mmol) at room temperature. The mixture was stirred at 90℃ for 30 mins. The reaction mixture was diluted with ethyl acetate (20
mL) . The organic layer was washed with water (2 x 10 mL) , brine (20 mL) , dried over Na2SO4 and concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 607.3
Step F: 1- (5- (9- ( ( (R) -4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) -2-methylpiperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a mixture of (R) -1- (5- (9- ( (4- (4-aminophenyl) -2-methylpiperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (200 mg) , 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (131.72 mg, 0.33 mmol) in DCM (10 mL) was added DIPEA (127.94 mg, 0.99 mmol) . The reaction mixture was stirred at 25 ℃ for 24 hrs. The reaction mixture was concentrated and purified by RP-HPLC (Waters 2767/Qda, Column: XBridge C18 19*250mm, 10 um; Mobile Phase A: 10mmol NH4HCO3/H2O, B: ACN; flow rate: 20ml/min; gradient: 50%~50%; Retention Time: 10.5-12.1min of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 942.5
1H NMR (400 MHz, DMSO) δ 10.51 (s, 1H) , 10.10 (s, 1H) , 8.81 (s, 1H) , 7.91 (s, 1H) , 7.69 (d, J = 8.1 Hz, 1H) , 7.63 (d, J = 8.2 Hz, 1H) , 7.55 (d, J = 1.8 Hz, 3H) , 7.39 (d, J = 8.1 Hz, 1H) , 6.90 (d, J = 8.6 Hz, 2H) , 5.76 (s, 1H) , 5.72 – 5.61 (m, 1H) , 5.04 (s, 1H) , 4.99 (d, J = 11.5 Hz, 1H) , 4.85 (d, J = 17.2 Hz, 1H) , 4.74 (s, 1H) , 4.57 (s, 1H) , 3.74 (s, 1H) , 3.62 (d, J = 11.4 Hz, 3H) , 3.33 (s, 3H) , 2.95 (dd, J = 25.1, 17.9 Hz, 2H) , 2.83 – 2.63 (m, 4H) , 2.39 (d, J = 49.0 Hz, 4H) , 2.21 (dd, J = 11.9, 7.1 Hz, 2H) , 2.05 – 1.97 (m, 1H) , 1.89 (dd, J = 13.4, 7.5 Hz, 2H) , 1.75 – 1.64 (m, 4H) , 1.48 (s, 4H) , 1.36 (s, 2H) , 1.23 (s, 1H) , 1.11 (s, 2H) , 1.04 (s, 5H) , 0.87 (t, J = 7.3 Hz, 3H) .
Example 66: (R) -1- (5- (4- ( (9- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-066)
Cpd-066 was synthesized according to the following scheme:
Step A: tert-butyl 9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane-3-carboxylate
To a solution of 1-fluoro-4-nitrobenzene (588 mg, 4.17 mmol) in DMF (15 mL) was added tert-butyl 3, 9-diazaspiro [5.5] undecane-3-carboxylate (1.06 g, 4.17 mmol) , then the reaction mixture was stirred at 80℃ for 16 hrs, After completion, the reaction was quenched with H2O (10 mL) and extracted with EA (3 × 20 mL) . The combined organic layers was washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =0: 1) to afford the title compound.
LC-MS ESI [M+H] + = 376.2;
Step B: 3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane
To a solution of tert-butyl 9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane-3-carboxylat (1.407 g, 3.75 mmol) in DCM (20 mL) was added HCl. dioxane (10 mL, 4 M) , then the reaction mixture was stirred at 30℃ for 2 hrs under N2 atmosphere. After completion of reaction, the reaction mixture was concentrated to afford the title compound.
LC-MS (ESI) [M+H] + = 276.1;
Step C: tert-butyl 4- ( (9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-
1-carboxylate
To a solution of give 3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane (528 mg) in DCM/MeOH (15 mL/15ml) was added tert-butyl 4-formylpiperidine-1-carboxylate (409 mg, 1.92 mmol) , then the reaction mixture was stirred at 30℃ for 30 mins, NaBH (OAc) 3 (610 mg, 2.88 mmol) AcOH (1.0 mL) and TEA (1.0 mL) were added. The mixture was stirred at 30℃ for 4 hrs under N2 atmosphere. After completion. The mixture was quenched with H2O (10 mL) and extracted with EA (3 × 20 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =0: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 473.4
Step D: 3- (4-nitrophenyl) -9- (piperidin-4-ylmethyl) -3, 9-diazaspiro [5.5] undecane
To a solution of tert-butyl 4- ( (9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carboxylate (400 mg, 0.845 mmol) in DCM (10 mL) was added HCl. dioxane (5 mL, 4 M) , then the reaction mixture was stirred at 30℃ for 2 hrs under N2 atmosphere. After completion of reaction, the reaction mixture was concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 373.3
Step E: 1- (2-chloro-5- (4- ( (9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-
yl) methyl) piperidine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 3- (4-nitrophenyl) -9- (piperidin-4-ylmethyl) -3, 9-diazaspiro [5.5] undecane (208 mg) in DMF (20 mL) was added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (243 mg, 0.559 mmol) , DIPEA (217 mg, 1.68 mmol) , then the reaction mixture was stirred at 25℃ for 2 hrs under N2 atmosphere. After completion, the reaction was quenched with H2O (10 mL) , extracted with EA (3 × 20 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =0: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 623.3
Step F: 1- (5- (4- ( (9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-
carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (4- ( (9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (250 mg, 0.402
mmol) in EtOH/H2O (10 mL/1 mL) were added Fe (115 mg, 2.06 mmol) and NH4Cl (110 mg, 2.06 mmol) , then the reaction mixture was stirred at 90℃ for 2 hrs under N2 atmosphere. After completion, the mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was added Η20 (30 mL) and extracted by EtOAc (20 mL x 3) . The combined organic phase was washed with brine, dried over anhydrous Na2SO4. the organic layer was concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 593.3
Step G: (R) -1- (5- (4- ( (9- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (5- (4- ( (9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (90 mg) in DCM (10 mL) was added 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (50 mg, 0.125 mmol) , the mixture was stirred at 25℃ for 18 hrs under N2 atmosphere. The mixture was quenched by addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 17-27% Retention Time: 9-19 of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 928.5
1H NMR (400 MHz, DMSO) δ 10.50 (s, 1H) , 10.09 (s, 1H) , 8.81 (s, 1H) , 8.33 (s, 1H) , 7.91 (s, 1H) , 7.69 (d, J = 7.7 Hz, 1H) , 7.63 (d, J = 8.2 Hz, 1H) , 7.54 (s, 3H) , 7.38 (d, J = 8.2 Hz, 1H) , 6.91 (d, J = 8.7 Hz, 2H) , 5.67 (d, J = 5.8 Hz, 1H) , 4.99 (d, J = 10.8 Hz, 2H) , 4.86 (d, J = 16.8 Hz, 1H) , 4.74 (s, 1H) , 4.51 (d, J = 57.9 Hz, 2H) , 3.80-3.73 (m, 2H) , 3.62-3.53 (m, 3H) , 3.50-3.00 (m, 5H) , 2.96 (d, J = 9.6 Hz, 1H) , 2.80-2.68 (m, 3H) , 2.43-2.30 (m, 4H) , 2.31-2.21 (m, 1H) , 2.22-2.10 (m, 2H) , 2.08-2.01 (m, 1H) , 1.88 (d, J = 7.8 Hz, 1H) , 1.78 (d, J = 20.3 Hz, 2H) , 1.70 (dd, J = 13.3, 7.4 Hz, 2H) , 1.60-1.52 (m, 4H) , 1.52-1.43 (m, 4H) , 1.07 (d, J = 10.9 Hz, 2H) , 0.87 (t, J = 7.2 Hz, 3H) .
Example 67: (R) -1- (5- (2- ( (1- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-7-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-067)
Cpd-067 was synthesized according to the following scheme:
Step A: tert-butyl 2- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-7-
carboxylate
A solution of tert-butyl 2, 7-diazaspiro [3.5] nonane-7-carboxylate (491 mg, 2.17 mmol) , 1- (4-nitrophenyl) piperidine-4-carbaldehyde (508 mg 2.17 mmol) , NaBH (OAc) 3 (1.40 g 6.51 mmol) , HOAc (130 mg 2.17 mmol) in DCM (5.0 mL) and MeOH (5 mL) was stirred at 30℃ for 12 hrs. After completion, the reaction mixture was quenched with water (10 mL) , extracted with EtOAc
(3*10 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
LC-MS (ESI) [M+H] +. 445.3
Step B: 2- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane
To a solution of tert-butyl 2- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-7-carboxylate (755 mg, 1.70 mmol) in DCM (5.0 mL) was added HCl in dioxane (5.0 mL, 4N) . The mixture was stirred at 25℃ for 2 h. The mixture was concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 345.3
Step C: 1- (2-chloro-5- (2- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 7-
diazaspiro [3.5] nonane-7-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 2- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane (763 mg) in DMF (15.0 mL) were added 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoic acid (595 mg, 2.22 mmol) , DIEA (856 mg, 6.66 mmol) and HATU (844 mg 2.22 mmol) . The mixture was stirred at r. t. for 2 hrs. After completion, the reaction was quenched with water (10 mL) and extracted with EA (3 *20 mL) . The combined organic layers was washed with brine (10 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 595.6
Step D: 1- (5- (2- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-7-
carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (2- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-7-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (512 mg, 0.862 mmol) in EtOH (10 mL) and H2O (3.0 mL) were added NH4Cl (231 mg, 4.31 mmol) and Fe (241 mg 4.31 mmol) . The mixture was stirred at 90℃ for 2 h. After completion, the mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was added Η20 (30 mL) and extracted by EtOAc (20 mL x 3) . The combined organic phase was washed with brine, dried over anhydrous Na2S04 and the organic layer was concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 565.3
Step E: (R) -1- (5- (2- ( (1- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-7-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
A solution of (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylthio) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (100 mg, 0.261 mmol) in DCM (10 mL) was added m-CPBA (80.0 mg, 0.392 mmol) . The mixture was stirred at 25℃ for 2 h. 1- (5- (2- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -2, 7-diazaspiro [3.5] nonane-7-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (147 mg) and DIPEA (101 mg, 0.783 mmol) were added. The mixture was stirred at 25℃ for 12 hrs. After completion, the reaction mixture was quenched with water (50 mL) , the resulting solution was extracted with DCM (2*100 mL) , the organic layers was combined and concentrated to get the crude. The crude was purified by Pre-HPLC (Waters 2767/Qda, Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN; flow rate: 20ml/min; gradient: 18%~28%; Retention Time: 8.0-9.4min of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 900.2.
1H NMR (400 MHz, DMSO) δ 10.51 (s, 1H) , 10.10 (s, 1H) , 8.81 (s, 1H) , 8.18 (s, 1H) , 7.91 (s, 1H) , 7.66 (dd, J = 18.9, 8.2 Hz, 2H) , 7.55 (d, J = 2.0 Hz, 3H) , 7.38 (dd, J = 8.2, 2.0 Hz, 1H) , 6.90 (d, J = 9.0 Hz, 2H) , 5.65 (dt, J = 10.4, 8.2 Hz, 1H) , 5.10 – 4.95 (m, 2H) , 4.85 (d, J = 16.1 Hz, 1H) , 4.58 (s, 1H) , 3.74 (s, 1H) , 3.60 (d, J = 13.0 Hz, 4H) , 2.99 (s, 6H) , 2.80 – 2.71 (m, 3H) , 2.61 –2.51 (m, 4H) , 2.34 (d, J = 7.1 Hz, 3H) , 2.24 – 2.15 (m, 1H) , 2.01 (d, J = 5.0 Hz, 1H) , 1.89 (dd, J = 14.0, 7.6 Hz, 1H) , 1.70 (dd, J = 13.5, 7.4 Hz, 7H) , 1.40 (s, 1H) , 1.22 (d, J = 11.7 Hz, 2H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 68: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) -2-methylphenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-068)
Cpd-068 was synthesized according to the following scheme:
Step A: tert-butyl 4- (2-methyl-4-nitrophenyl) piperazine-1-carboxylate
To a solution of 1-fluoro-2-methyl-4-nitrobenzene (5.00 g, 32.2 mmol) in DMSO (30 mL) were added tert-butyl piperazine-1-carboxylate (6.00 g, 32.2 mmol) and DIEA (12.5 g, 96.7 mmol) , the mixture was stirred at 120 ℃ for 15 hrs under N2 atmosphere. The mixture was quenched by addition of 10 mL water, the reaction mixture was filtered through celite, and the filter cake was washed with water (30 mL) . The combined filtrate were concentrated under reduced pressure to afford the title compound.
1H NMR (400 MHz, DMSO) δ 8.03 (dd, J = 12.4, 3.6 Hz, 2H) , 7.14 (d, J = 8.8 Hz, 1H) , 3.49 (s, 4H) , 2.99 –2.94 (m, 4H) , 2.35 (s, 3H) , 1.43 (s, 9H)
Step B: 1- (2-methyl-4-nitrophenyl) piperazine
To a solution of tert-butyl 4- (2-methyl-4-nitrophenyl) piperazine-1-carboxylate (500 mg) in DCM (10 mL) was added 1M HCl/Dioxane (2 mL) 0℃ for 2 h under N2 atmosphere. The mixture was dried under reduced pressure to afford the title compound as a crude product.
LC-MS (ESI) [M+H] + 222.0
Step C: tert-butyl 9- ( (4- (2-methyl-4-nitrophenyl) piperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carboxylate
To a solution of 1- (2-methyl-4-nitrophenyl) piperazine (150 mg) in DCM (10 mL) , MeOH (1 mL) and HOAc (1 mL) was added tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (191 mg, 0.678 mmol) , NaBH (OAc) 3 (431 mg, 2.03 mmol) , the mixture was stirred at 25℃ for 12 hrs under N2 atmosphere. The mixture was quenched by addition of 10 mL water. The resulting solution was extracted with DCM (3*10 mL) , the organic layers were combined and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =100: 1-0: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 487.4
Step D: 9- ( (4- (2-methyl-4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane
To a solution of tert-butyl 9- ( (4- (2-methyl-4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate (170 mg, 0.349 mmol) in DCM (10 mL) was added 1M HCl/Dioxane (2 mL) at 0 ℃ , the mixture was stirred at 20 ℃ for 1 h under N2 atmosphere. The mixture was dried under reduced pressure to afford the title compound.
LC-MS (ESI) [M+H] + 387.3
Step E: 1- (2-chloro-5- (9- ( (4- (2-methyl-4-nitrophenyl) piperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 9- ( (4- (2-methyl-4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane (120 mg) in DMF (20 mL) were added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (135 mg, 0.310 mmol) and DIPEA (120 mg, 0.93 mmol) , Then the reaction mixture was stirred at 25 ℃ for 2 hrs under N2 atmosphere. After completion, the reaction was quenched with H2O (10 mL) , extracted with EA (3*20 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =0: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 637.1
Step F: 1- (5- (9- ( (4- (4-amino-2-methylphenyl) piperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (9- ( (4- (2-methyl-4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (150 mg, 0.235
mmol) in EtOH/H2O (10 mL/1 mL) were added Fe (66.0 mg, 1.18 mmol) and NH4Cl (63.1 mg, 1.18 mmol) then the reaction mixture was stirred at 90℃ for 2 h under N2 atmosphere. After completion, the mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was added Η20 (30 mL) and extracted by EtOAc (20 mL x 3) . The combined organic phase was washed with brine, dried over anhydrous Na2S04, and the organic layer was concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 607.3
Step G: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) -2-
methylphenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (5- (9- ( (4- (4-amino-2-methylphenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (140 mg) in DCM (10 mL) were added 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (92.2 mg, 0.231 mmol) and DIPEA (89.4 mg, 0.693 mmol) , the mixture was stirred at 25℃ for 12 hrs under N2 atmosphere. The mixture was quenched by addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 17-27%Retention Time: 9-19 of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 942.6
1H NMR (400 MHz, DMSO) δ 10.50 (s, 1H) , 10.12 (s, 1H) , 8.84 (s, 1H) , 8.33 (s, 1H) , 7.90 (d, J = 8.1 Hz, 1H) , 7.71 (d, J = 8.1 Hz, 1H) , 7.64 (d, J = 8.2 Hz, 2H) , 7.55 (s, 1H) , 7.41 (dd, J = 14.5, 9.6 Hz, 2H) , 6.99 (d, J = 8.7 Hz, 1H) , 5.73 – 5.61 (m, 1H) , 5.01 (t, J = 13.0 Hz, 2H) , 4.85 (d, J = 17.3 Hz, 1H) , 4.74 (s, 1H) , 4.56 (d, J = 9.9 Hz, 1H) , 3.74 (s, 1H) , 3.62 (d, J = 12.8 Hz, 4H) , 3.02 –2.91 (m, 1H) , 2.80 (s, 5H) , 2.77 – 2.65 (m, 3H) , 2.33 (s, 3H) , 2.23 (s, 3H) , 2.19 (d, J = 7.8 Hz, 3H) , 2.06 –1.97 (m, 1H) , 1.89 (dd, J = 13.7, 7.1 Hz, 1H) , 1.76 – 1.65 (m, 3H) , 1.60 (s, 2H) , 1.51 (s, 2H) , 1.47 –1.40 (m, 1H) , 1.36 (s, 1H) , 1.29 (s, 1H) , 1.09 (s, 5H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 69: (R) -1- (5- (9- ( (1- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperidin-4-yl) methyl) -1-oxa-4, 9-diazaspiro [5.5] undecane-4-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-069)
Cpd-069 was synthesized according to the following scheme:
Step A: tert-butyl 9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -1-oxa-4, 9-
diazaspiro [5.5] undecane-4-carboxylate
A solution of tert-butyl 1-oxa-4, 9-diazaspiro [5.5] undecane-4-carboxylate (200 mg, 0.781 mmol) , 1- (4-nitrophenyl) piperidine-4-carbaldehyde (173 mg 0.781 mmol) , NaBH (OAc) 3 (497 mg 2.34 mmol) , HOAc (47.0 mg 0.781 mmol) in DCM (5.0 mL) and MeOH (5 mL) was stirred at 30℃ for 12 hrs. After completion, the reaction was quenched with water (10 mL) , extracted with EtOAc (3*10 mL) . The combined organic layers was washed with brine (10 mL) , dried
with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
LC-MS (ESI) [M+H] +. 475.3
Step B: 9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -1-oxa-4, 9-diazaspiro [5.5] undecane
To a solution of tert-butyl 9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -1-oxa-4, 9-diazaspiro [5.5] undecane-4-carboxylate (210 mg, 0.443 mmol) in DCM (5.0 mL) was added HCl in dioxane (4.0 mL, 4N) and stirred at 25℃ for 2 h. The mixture was concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 375.3
Step C: 1- (2-chloro-5- (9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -1-oxa-4, 9-
diazaspiro [5.5] undecane-4-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -1-oxa-4, 9-diazaspiro [5.5] undecane (326 mg) in DMF (5.0 mL) were added 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoic acid (234 mg, 0.872 mmol) , DIEA (337 mg, 2.62 mmol) , HATU (331 mg 0.872 mmol) . The mixture was stirred at r. t. for 2 h. After completion, the reaction was quenched with water (10 mL) , extracted with EA (3*20 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the title compound.
LC-MS (ESI) [M+H] + 625.8
Step D: 1- (5- (9- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -1-oxa-4, 9-
diazaspiro [5.5] undecane-4-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -1-oxa-4, 9-diazaspiro [5.5] undecane-4-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (155 mg, 0.248 mmol) in EtOH (10 mL) and H2O (3.0 mL) were added NH4Cl (64.0 mg, 1.24 mmol) and Fe (67.0 mg 1.24 mmol) . The mixture was stirred at 90℃ for 2 hrs. After completion, The mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was added Η20 (30 mL) and extracted by EtOAc (20 mL x 3) . The combined organic phase was washed with brine, dried over anhydrous Na2S04 and the organic layer was concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 595.4
Step E: (R) -1- (5- (9- ( (1- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperidin-4-yl) methyl) -1-oxa-4, 9-diazaspiro [5.5] undecane-4-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylthio) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (60 mg, 0.157 mmol) in DCM (5 mL) was added m-CPBA (48.0 mg, 0.236 mmol) . The mixture was stirred at 25C for 2 hrs. 1- (5- (9- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -1-oxa-4, 9-diazaspiro [5.5] undecane-4-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (140 mg) was added. The mixture was stirred at 25℃ for 12 hrs. After completion, the reaction mixture was quenched with water (50 mL) , the resulting solution was extracted with DCM (2*100 mL) , the organic layers was combined and concentrated to get a residue, which was purified by Pre-HPLC (Waters 2767/Qda, Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 25%~35%; Retention Time: 7.6-8.9min of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 930.2.
1H NMR (400 MHz, DMSO) δ 10.51 (s, 1H) , 10.25 –9.87 (m, 1H) , 8.81 (s, 1H) , 8.17 (s, 1H) , 7.91 (s, 1H) , 7.71 –7.64 (m, 2H) , 7.56 (s, 2H) , 7.42 (s, 1H) , 6.90 (d, J = 9.0 Hz, 2H) , 5.67 (d, J =6.6 Hz, 1H) , 5.12 –4.90 (m, 2H) , 4.88 (s, 1H) , 4.79 –4.64 (m, 1H) , 4.57 (s, 1H) , 3.75 (s, 1H) , 3.62 (s, 6H) , 3.50 (s, 3H) , 2.95 (s, 2H) , 2.75 (s, 4H) , 2.59 (s, 2H) , 2.44 –2.34 (m, 2H) , 2.33 (s, 1H) , 2.20 (d, J = 5.9 Hz, 3H) , 2.03 (d, J = 7.9 Hz, 2H) , 1.93 –1.82 (m, 1H) , 1.81 –1.67 (m, 5H) , 1.65 –1.53 (m, 2H) , 1.18 (s, 2H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 70: 1- (5- (9- ( ( (1R, 4R) -5- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-070)
Cpd-070 was synthesized according to the following scheme:
Step A: tert-butyl (1R, 4R) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylate
A solution of tert-butyl (1R, 4R) -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylate (1 g, 5.04 mmol) , 1-fluoro-4-nitrobenzene (624 mg, 4.42 mmol) , DIEA (1.30 g, 10.07 mmol) in ACN (10 mL) was stirred at 80℃ for 16 hrs. After completion, Water (100 mL) was added and the solution was extracted with EtOAc (100 mL x 3) . The combined organic layers was washed with brine (100 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. the crude product was purified by silica gel chromatography eluted with PE: EA=1: 1 to afford the title compound. LC-MS (ESI) [M+H] + = 319.9
Step B: (1R, 4R) -2- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptane
To a solution of tert-butyl (1R, 4R) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylate (1.08 g, 3.39 mmol) in HCl in dioxane (10 mL, 4 M) and DCM (10 mL) was stirred at 20℃ for 1 h. After completion, the reaction mixture was concentrated under reduced pressure to dryness to afford the title compound.
LC-MS (ESI) [M+H] + = 220.1
Step C: tert-butyl 9- ( ( (1R, 4R) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-
yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate
A solution of (1R, 4R) -2- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptane (155 mg) , tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (200 mg, 0.711 mmol) , NaBH (OAc) 3 (301 mg, 1.42 mmol) , CH3COOH (85.4 mg, 1.42 mmol) in DCM (5 mL) and MeOH (5 mL) r was stirred at 25℃ for 12 hrs. The reaction mixture was quenched with water (50 mL) , and the mixture was extracted with EtOAc (50 mL x 3) . The combined organic layers was washed with brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography eluted with DCM: MeOH=10: 1 to afford the title compound.
LC-MS (ESI) [M+H] + = 485.3
Step D: 9- ( ( (1R, 4R) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-
azaspiro [5.5] undecane
To a solution of tert-butyl 9- ( ( (1R, 4R) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate (121 mg, 0.250 mmol) in HCl in dioxane (2 mL, 4 M) and DCM (2 mL) was stirred at 20℃ for 1 h. After completion, the reaction mixture was concentrated under reduced pressure to afford the title compound.
LC-MS (ESI) [M+H] + = 385.2
Step E: 1- (2-chloro-5- (9- ( ( (1R, 4R) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-
yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -
dione
A solution of 9- ( ( (1R, 4R) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-azaspiro [5.5] undecane (130 mg) , perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (220 mg, 0.508 mmol) , DIEA (87.3 mg, 0.678 mmol) in DMF (2 mL) was stirred at 25℃ for 4hrs. After completion, water (20 mL) was added and the solution was extracted with EtOAc (20 mL x 3) . The combined organic layers was washed with brine (20 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. the crude product was purified by silica gel chromatography eluted with DCM: MeOH=10: 1 to afford the title compound.
LC-MS (ESI) [M+H] + = 635.3
Step F: 1- (5- (9- ( ( (1R, 4R) -5- (4-aminophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-
azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (9- ( ( (1R, 4R) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (114 mg, 0.180 mmol) , NH4Cl (47.6 mg, 0.899 mmol) , in H2O (2 mL) and EtOH (20 mL) were
added Fe (50.3 mg, 0.899 mmol) , Then the mixture was stirred at 90℃ for 2hrs. After completion of reaction, the mixture was filtered through a Celite pad, water (200 mL) was added and the filtrate was extracted with EtOAc (200 mL x 3) . The combined organic layers was washed with brine (200 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. the crude product was purified by silica gel chromatography eluted with DCM: MeOH=10: 1 to afford the title compound.
LC-MS (ESI) [M+H] + = 605.3
Step G: 1- (5- (9- ( ( (1R, 4R) -5- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-azaspiro [5.5] undecane-3-
carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (5- (9- ( ( (1R, 4R) -5- (4-aminophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (102 mg, 0.169 mmol) in DCM (10 mL) was added 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (50.0 mg, 0.125 mmol) , the mixture was stirred at 25℃ for 16 hrs. The mixture was quenched by addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767 /Qda, Column: Pursuit XRs 10 C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 26%~36%; Retention Time: 8.4-9.3min of 18 min) to afford the title compound.
LC-MS (ESI) [M+H] +=940.5
1H NMR (400 MHz, DMSO) δ 10.49 (s, 1H) , 10.00 (s, 1H) , 8.78 (s, 1H) , 8.30 (s, 1H) , 7.87 (d, J = 8.0 Hz, 1H) , 7.69 (d, J = 8.2 Hz, 1H) , 7.62 (d, J = 8.2 Hz, 1H) , 7.51 (d, J = 16.7 Hz, 3H) , 7.37 (d, J = 8.2 Hz, 1H) , 6.55 (d, J = 8.1 Hz, 2H) , 5.66 (dd, J = 16.6, 10.3 Hz, 1H) , 4.99 (d, J = 9.9 Hz, 2H) , 4.86 (d, J = 17.2 Hz, 1H) , 4.75 (s, 1H) , 4.57 (s, 1H) , 4.25 (s, 1H) , 3.73 (s, 1H) , 3.59 (d, J = 19.9 Hz, 3H) , 3.45 (s, 1H) , 3.26 –3.18 (m, 3H) , 3.10 (d, J = 9.1 Hz, 1H) , 2.95 (d, J = 6.0 Hz, 1H) , 2.84 (d, J = 8.8 Hz, 1H) , 2.73 (d, J = 4.2 Hz, 3H) , 2.42 (d, J = 9.4 Hz, 1H) , 2.28 (s, 2H) , 2.19 (d, J = 5.4 Hz, 1H) , 2.01 (s, 1H) , 1.89 (dd, J = 13.5, 7.0 Hz, 1H) , 1.81 (s, 1H) , 1.77 –1.59 (m, 4H) , 1.48 (s, 3H) , 1.40 (s, 2H) , 1.25 (s, 2H) , 1.03 (s, 4H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 71: 3- [3- [9- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] phenyl] piperidine-2, 6-dione (Cpd-071)
Cpd-071 was synthesized according to the following scheme:
Step A: 3- (2, 6-dibenzyloxy-3-pyridyl) benzoic acid
A mixture of 3-boronobenzoic acid (1 g, 6.03 mmol) , 2, 6-dibenzyloxy-3-bromo-pyridine (2.23 g, 6.03 mmol) , Pd (dppf) Cl2
. CH2Cl2 (492.13 mg, 602.63 umol) , Na2CO3 (1.92 g, 18.08 mmol) in dioxane (20 mL) and H2O (2 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90 ℃ for 16 hrs under N2 atmosphere. The mixture was poured into water (50 mL) and extracted with DCM (40 mL × 3) . The combined organic layers was washed with brine (40 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by flash silica gel chromatography (40 gSilica Flash Column, Eluent of 0~30%Petroleum ether/Ethyl acetate@80 mL/min) to afford the title compound.
LCMS: MS (ESI) m/z = 412.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 12.92-13.09 (m, 1H) , 8.20 (t, J = 1.6 Hz, 1H) , 7.84-7.91 (m, 1H) , 7.75-7.83 (m, 2H) , 7.53 (t, J = 7.6 Hz, 1H) , 7.29-7.47 (m, 11H) , 5.43 (s, 2H) , 5.40 (s, 2H)
Step : 3- (2, 6-dioxopiperidin-3-yl) benzoic acid
To a solution of 3- (2, 6-dibenzyloxy-3-pyridyl) benzoic acid (400 mg, 972.18 umol) in MeOH (10 mL) was added Pd/C (0.2 g, 972.18 umol, 10%w/w) . The mixture was stirred at 25 ℃ for 16 h under H2 atmosphere (50 Psi) . The reaction mixture was filtered, the solid was washed with MeOH (100 mL) . The filtrate was concentrated under reduced pressure to afford the title compound.
LCMS: MS (ESI) m/z = 234.1 [M+H] +.
Step C: 3- [3- (2, 6-dioxo-3-piperidyl) benzoyl] -3-azaspiro [5.5] undecane-9-carbaldehyde
To a solution of 3- (2, 6-dioxo-3-piperidyl) benzoic acid (51.46 mg) and 3-azaspiro [5.5] undecane-9-carbaldehyde (40.00 mg, 220.66 umol) in DCM (3 mL) were added EDCI (84.60 mg, 441.32 umol) , HOBt (59.63 mg, 441.32 umol) and DIEA (142.59 mg, 1.10 mmol, 192.18 uL) . The mixture was stirred at 25 ℃ for 4 hrs under N2 atmosphere. The mixture was poured into water (40 mL) and extracted with DCM (40 mL × 3) and the organic layer was concentrated to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH = 10: 1) to afford the title compound.
LCMS: MS (ESI) m/z = 397.2 [M+H] +.
Step D: 3- [3- [9- [ [4- [4- [ [2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-
2-yl] -3-oxo-pyrazolo [3, 4-d] pyrimidin-6-yl] amino] phenyl] piperazin-1-yl] methyl] -3-
azaspiro [5.5] undecane-3-carbonyl] phenyl] piperidine-2, 6-dione
To a solution of 3- [3- (2, 6-dioxo-3-piperidyl) benzoyl] -3-azaspiro [5.5] undecane-9-carbaldehyde (29.4 mg, crude) and 2-allyl-1- [ (7R) -7-ethyl-7-hydroxy-5, 6-dihydrocyclopenta [b] pyridin-2-yl] -6- (4-piperazin-1-ylanilino) pyrazolo [3, 4-d] pyrimidin-3-one (13.30 mg, 25.95 umol) in DCM (3 mL) were added NaBH (OAc) 3 (27.50 mg, 129.77 umol) and Et3N (13.13 mg, 129.77 umol, 18.06 uL) . The mixture was stirred at 25 ℃ for 2 hrs under N2 atmosphere. The mixture was poured into water (40 mL) and extracted with DCM (40 mL × 3) . The organic layer was concentrated to give a residue. The residue was purified by prep-HPLC (column: C18-1 150*30mm*5um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 30%-70%, 9 min) to afford the title compound.
LCMS: MS (ESI) m/z = 893.4 [M+H] +.
1H NMR (400 MHz, CDCl3) δ: 8.83 (s, 1H) , 8.09 (s, 1H) , 7.66-7.72 (m, 2H) , 7.33-7.51 (m, 6H) , 6.91 (d, J = 8.8 Hz, 2H) , 5.65-5.75 (m, 1H) , 5.02 (d, J = 10.0 Hz, 1H) , 4.94 (d, J = 17.6 Hz, 1H) , 4.80-4.85 (m, 1H) , 4.65-4.70 (m, 1H) , 3.72-3.83 (m, 4H) , 3.26-3.36 (m, 7H) , 3.02-3.09 (m, 1H) , 2.78-2.92 (m, 2H) , 2.75 (t, J = 5.2 Hz, 2H) , 2.68-2.72 (m, 3H) , 2.24-2.39 (m, 9H) , 1.97-2.05 (m, 1H) , 1.84-1.88 (m, 2H) , 1.72-1.77 (m, 7H) , 1.47 (s, 4H) .
Example 72: 1- [2-chloro-5- [9- [ [4- [4- [11- (2, 6-dichlorophenyl) -10-oxo-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-4-yl] pyrazol-1-yl] -1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] phenyl] hexahydropyrimidine-2, 4-dione (Cpd-072)
Cpd-072 was synthesized according to the following scheme:
Step A: tert-butyl 4- [4- [11- (2, 6-dichlorophenyl) -10-oxo-5- (2-trimethylsilylethoxymethyl) -
5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-4-yl] pyrazol-1-
yl] piperazine-1-carboxylate and tert-butyl 4- (4- (3- (2-chlorophenyl) -4-oxo-7- ( (2-
(trimethylsilyl) ethoxy) methyl) -4, 7-dihydro-3H-pyrrolo [3', 2': 5, 6] pyrido [4, 3-d] pyrimidin-8-
yl) -1H-pyrazol-1-yl) piperidine-1-carboxylate (a mixture)
A mixture of 4-bromo-11- (2, 6-dichlorophenyl) -5- (2-trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-10-one (80 mg, 148.06 μmol) , tert-butyl 4- [4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazol-1-yl] piperazine-1-carboxylate (56.01 mg, 148.06 μmol) and KF (17.20 mg, 296.13 μmol) , ditert-butyl (cyclopentyl) phosphane; dichloropalladium; iron (9.65 mg, 14.81 μmol) in dioxane (5 mL) and H2O (0.5 mL) was stirred at 100 ℃ for 16 hr. The mixture was poured into water (100 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers was washed with brine (50 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by column chromatography on silica gel (5%MeOH in Dichloromethane) to afford the title compound. (a mixture) .
Step B: 11- (2, 6-dichlorophenyl) -4- (1-piperazin-1-ylpyrazol-4-yl) -5, 7, 11, 13-
tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-10-one and 11- (2-chlorophenyl) -
4- (1-piperazin-1-ylpyrazol-4-yl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-
pentaen-10-one
To a solution of tert-butyl 4- [4- [11- (2, 6-dichlorophenyl) -10-oxo-5- (2-trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-4-yl] pyrazol-1-yl] piperazine-1-carboxylate (70 mg, 98.35 μmol) in DCM (1 mL) was added TFA (1.53 g, 15.71 mmol, 1.0 mL) . The mixture was purified by prep-HPLC (column: Phenomenex C18 75*30mm*3um; mobile phase: [water (FA) -ACN] ; B%: 8%-48%, 9 min) and dried by lyophilization to give 11- (2, 6-dichlorophenyl) -4- (1-piperazin-1-ylpyrazol-4-yl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-10-one, and 11- (2-chlorophenyl) -4- (1-piperazin-1-ylpyrazol-4-yl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-10-one.
11- (2, 6-dichlorophenyl) -4- (1-piperazin-1-ylpyrazol-4-yl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-10-one:
LCMS: MS (ESI) m/z = 480.1 [M+H] +
11- (2-chlorophenyl) -4- (1-piperazin-1-ylpyrazol-4-yl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-10-one:
LCMS: MS (ESI) m/z = 446.2 [M+H] +
Step C: 1- [2-chloro-5- [9- [ [4- [4- [11- (2, 6-dichlorophenyl) -10-oxo-5, 7, 11, 13-
tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-4-yl] pyrazol-1-yl] -1-
piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] phenyl] hexahydropyrimidine-2, 4-
dione
To a solution of 11- (2, 6-dichlorophenyl) -4- [1- (4-piperidyl) pyrazol-4-yl] -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-10-one (10 mg, 20.82 μmol) , 3- [4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoyl] -3-azaspiro [5.5] undecane-9-carbaldehyde (13.49 mg, 31.23 μmol) , TEA (10.53 mg, 104.09 μmol) in DCM (2 mL) was added NaBH (OAc) 3 (22.06 mg, 104.09 μmol) . The mixture was stirred at 20 ℃ for 3 hrs. The mixture was concentrated to give a residue, which was purified by prep-HPLC (column: Phenomenex C18 75*30mm*3um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 41%-81%, 9 min) and to afford the title compound.
LCMS: MS (ESI) m/z = 897.4 [M+H] +
1H NMR (400 MHz, CDCl3) δ: 10.76 (s, 1H) , 9.25 (s, 1H) , 8.35–8.71 (m, 1H) , 8.05 (s, 1H) , 7.89 (s, 2H) , 7.54–7.59 (m, 3H) , 7.43–7.49 (m, 2H) , 7.39 (d, J = 8.0 Hz, 1H) , 7.09 (d, J = 2.0 Hz, 1H) , 4.22 (s, 1H) , 3.66–3.87 (m, 4H) , 3.41 (s, 2H) , 2.92–3.14 (m, 3H) , 2.79–2.90 (m, 1H) , 2.07–2.38 (m, 8H) , 1.68–1.78 (m, 4H) , 1.46 (s, 3H) , 1.33 (s, 1H) , 0.92–1.23 (m, 5H) .
Example 73: 1- [2-chloro-5- [9- [ [4- [4- [11- (2-chlorophenyl) -10-oxo-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-4-yl] pyrazol-1-yl] -1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] phenyl] hexahydropyrimidine-2, 4-dione (Cpd-073)
Cpd-073 was synthesized according to the following scheme:
Step A: 1- [2-chloro-5- [9- [ [4- [4- [11- (2-chlorophenyl) -10-oxo-5, 7, 11, 13-
tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-4-yl] pyrazol-1-yl] -1-
piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] phenyl] hexahydropyrimidine-2, 4-
dione
To a solution of 11- (2-chlorophenyl) -4- [1- (4-piperidyl) pyrazol-4-yl] -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-10-one (10 mg, 22.43 μmol) , 3- [4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoyl] -3-azaspiro [5.5] undecane-9-carbaldehyde (14.53 mg, 33.64 μmol) , TEA (11.35 mg, 112.13 μmol) in DCM (2 mL) was added NaBH (OAc) 3 (23.77 mg, 112.13 μmol) . The mixture was stirred at 20 ℃ for 3 hrs. The mixture was concentrated to give a residue, which was purified by prep-HPLC (column: Phenomenex C18 75*30mm*3um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; B%: 38%-78%, 9 min) and dried by lyophilization to afford the title compound.
LCMS: MS (ESI) m/z = 863.3 [M+H] +
1H NMR (400 MHz, CDCl3) δ: 9.23 (s, 1H) , 8.17 (s, 1H) , 7.90–8.03 (m, 1H) , 7.88 (s, 1H) , 7.63–7.68 (m, 1H) , 7.56 (d, J = 8.0 Hz, 1H) , 7.48–7.54 (m, 3H) , 7.45 (d, J = 2.0 Hz, 1H) , 7.39 (d, J =8.0 Hz, 1H) , 7.06–7.10 (m, 1H) , 7.08 (s, 1H) , 4.26 (s, 1H) , 3.58–3.86 (m, 4H) , 3.40 (s, 2H) , 3.12 (s, 1H) , 2.90 –3.04 (m, 1H) , 2.78–2.89 (m, 1H) , 2.10–2.45 (m, 6H) , 1.73 (s, 5H) , 1.46 (s, 6H) , 0.99–1.36 (m, 5H) .
Example 74: (R) -1- (5- (2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) -7-azaspiro [3.5] nonane-7-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-074)
Cpd-074 was synthesized according to the following scheme:
Step A: tert-butyl 2- (4- (4-nitrophenyl) piperazin-1-yl) -7-azaspiro [3.5] nonane-7-carboxylate
To the mixture of 1- (4-nitrophenyl) piperazine hydrochloride (5.0 g, 20.52 mmol) in dichloromethane (100 mL) were added tert-butyl 2-oxo-7-azaspiro [3.5] nonane-7-carboxylate (7.4 g, 30.78 mmol) , AcOH (2.46 g, 41.04 mmol) , TEA (6.23 g, 61.56 mmol ) and NaBH (OAc) 3 (8.7 g, 41.04 mmol) . The mixture was stirred at 25 ℃ for 16 hrs. The reaction mixture was quenched with MeOH and concentrated under reduced pressure. The residue was purified with by silica gel chromatography eluted petroleum ether/ethylacetate = 2: 1 to afford the title compound.
LC-MS (ESI) [M+H] + 431.3
Step B: 2- (4- (4-nitrophenyl) piperazin-1-yl) -7-azaspiro [3.5] nonane
To the solution of tert-butyl 2- (4- (4-nitrophenyl) piperazin-1-yl) -7-azaspiro [3.5] nonane-7-carboxylate 1 (7.8 g, 18.11 mmol) in DCM (100 mL) was added 4 M HCl/dioxane (20 mL) . The mixture was stirred at 25 C for 16 hrs. The reaction mixture was concentrated under reduced pressure. The residue was triturated with methanol (10 mL) and filtered to afford the title compound.
LC-MS (ESI) [M+H] + 331.2
Step C: 1- (2-chloro-5- (2- (4- (4-nitrophenyl) piperazin-1-yl) -7-azaspiro [3.5] nonane-7-
carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 2- (4- (4-nitrophenyl) piperazin-1-yl) -7-azaspiro [3.5] nonane (500 mg) in DMF (5.00 mL) was added 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoic acid (409 mg, 1.52 mmol) , DIEA (588 mg, 4.56 mmol) and HATU (578mg 1.52 mmol) . The reaction mixture was stirred at 25 ℃ for 2 hrs. After completion, the reaction was quenched with water (10 mL) , extracted with EA (3 × 20 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to give the title compound.
LC-MS (ESI) [M+H] + 581.7
Step D: 1- (5- (2- (4- (4-aminophenyl) piperazin-1-yl) -7-azaspiro [3.5] nonane-7-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (2- (4- (4-nitrophenyl) piperazin-1-yl) -7-azaspiro [3.5] nonane-7-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (500 mg, 0.861 mmol) in EtOH (20 mL) and H2O (4 mL) was added NH4Cl (231 mg, 4.31 mmol) and Fe (241 mg, 4.31 mmol) . The mixture was stirred at 90℃ for 2 h. After completion, The mixture was filtered through a pad of celite. The filtrate was concentrated in vacuo. The residue was added Η20 (30 mL) and extracted by EtOAc (20 mL x 3) . The combined organic phase was washed with brine, dried over anhydrous Na2S04 and the organic layer was concentrated to obtain the title compound.
LC-MS (ESI) [M+H] + 551.4
Step E: (R) -1- (5- (2- (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) piperazin-1-yl) -7-azaspiro [3.5] nonane-7-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylthio) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (100 mg, 0.261 mmol) in DCM (10 mL) was added m-CPBA (58.0 mg, 0.287 mmol) was stirred at 0℃ for 1 h. To the resulting solution was added 1- (5- (2- (4- (4-aminophenyl) piperazin-1-yl) -7-azaspiro [3.5] nonane-7-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (144 mg) and DIPEA (101 mg,
0.783 mmol) . The reaction mixture was stirred at 25℃ for 12 hrs. The reaction mixture was quenched by addition of water (50 mL) , the resulting solution was extracted with DCM (2 × 100 mL) , the organic layers was combined and concentrated to get the residue. The residue was purified by Pre-HPLC to afford the title compound.
LC-MS (ESI) [M+H] + 886.2
1H NMR (400 MHz, DMSO) δ 10.52 (s, 1H) , 10.13 (d, J = 12.2 Hz, 1H) , 8.82 (s, 1H) , 8.22 (s, 1H) , 7.92 (d, J = 7.1 Hz, 1H) , 7.75 –7.51 (m, 5H) , 7.38 (d, J = 8.0 Hz, 1H) , 6.91 (d, J = 8.5 Hz, 2H) , 5.66 (ddd, J = 16.4, 10.7, 5.7 Hz, 1H) , 5.00 (d, J = 10.1 Hz, 1H) , 4.86 (d, J = 17.2 Hz, 1H) , 4.75 (s, 1H) , 4.57 (d, J = 10.6 Hz, 1H) , 3.74 (d, J = 6.4 Hz, 5H) , 3.24 (d, J = 33.4 Hz, 2H) , 3.09 (s, 4H) , 3.02 –2.90 (m, 1H) , 2.76 (t, J = 9.7 Hz, 4H) , 2.38 (s, 4H) , 2.25 –2.16 (m, 1H) , 2.01 (d, J = 5.3 Hz, 3H) , 1.90 (dd, J = 13.6, 7.1 Hz, 1H) , 1.71 (dd, J = 13.7, 7.3 Hz, 1H) , 1.57 (d, J = 29.2 Hz, 6H) , 0.87 (t, J = 7.2 Hz, 3H)
Example 75: 1- (5- (9- ( ( (1S, 4S) -5- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-075)
Cpd-075 was synthesized according to the following scheme:
Step A: tert-butyl (1S, 4S) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylate
A solution of tert-butyl (1S, 4S) -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylate (5.0 g, 25.22 mmol) , 1-fluoro-4-nitrobenzene (3.56 g, 25.22 mmol) , DIEA (6.52 g, 50.44 mmol) in ACN (150 mL) was stirred at 80℃ for 16 hrs. After completion of reaction, water (100 ml) was added and the solution was extracted with EtOAc (100 mL x 3) . The combined organic layers was washed with brine (100 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography eluted with PE: EA=1: 1 to give the title compound.
1H NMR (400 MHz, CDCl3) δ 8.13 (d, J = 9.1 Hz, 2H) , 6.49 (d, J = 9.1 Hz, 2H) , 4.75 –4.47 (m, 2H) , 3.59 (d, J = 9.2 Hz, 1H) , 3.46 (s, 2H) , 3.32 (dd, J = 35.0, 8.8 Hz, 2H) , 2.02 (d, J = 15.5 Hz, 2H) , 1.44 (d, J = 18.8 Hz, 9H) .
Step B: (1S, 4S) -2- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptane
To a solution of tert-butyl (1S, 4S) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptane-2-carboxylate (5.2 g, 16.3 mmol) in HCl in dioxane (20 mL, 4 M) and DCM (30 mL) was stirred at 20℃ for 1 h. After completion of reaction, the reaction mixture was concentrated under reduced pressure to dryness to afford the title compound.
LC-MS (ESI) [M+H] + 220.1 Rt=0.785min
Step C: tert-butyl 9- ( ( (1S, 4S) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -
3-azaspiro [5.5] undecane-3-carboxylate
A solution of (1S, 4S) -2- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptane (150 mg) , tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (212 mg) , NaBH (OAc) 3 (174 mg, 0.821 mmol) in DCM (5 mL) and MeOH (5 mL) was stirred at 25℃ for 12 hrs. The reaction mixture was quenched with water (50 mL) , and the solution was extracted with EtOAc (50 mL x 3) . The combined organic layers was washed with brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography eluted with PE: EA=1: 4 to give the title compound.
LC-MS (ESI) [M+H] + = 485.3
Step D: 9- ( ( (1S, 4S) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-
azaspiro [5.5] undecane
To a solution of tert-butyl 9- ( ( (1S, 4S) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate (190 mg, 0.393 mmol) in HCl in dioxane (2 mL, 4 M) and DCM (2 mL) was stirred at 20℃ for 1 h. After completion of reaction, the reaction mixture was concentrated under reduced pressure to dryness to afford the title compound.
LC-MS (ESI) [M+H] + = 385.2
Step E: 1- (2-chloro-5- (9- ( ( (1S, 4S) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-
yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -
dione
A solution of 9- ( ( (1S, 4S) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-azaspiro [5.5] undecane (150 mg) , perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (170 mg, 0.390 mmol) , DIEA (151 mg, 1.17 mmol) in DMF (2 mL) was stirred at 25℃ for 4hrs. After completion of reaction, water (20 mL) was added and the solution was extracted with EtOAc (20 mL x 3) . The combined organic layers was washed with brine (20
mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. the crude product was purified by silica gel chromatography eluted with DCM: MeOH=10: 1 to give the title compound.
LC-MS (ESI) [M+H] + = 635.3
Step F: 1- (5- (9- ( ( (1S, 4S) -5- (4-aminophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-
azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (9- ( ( (1S, 4S) -5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (180 mg, 0.283 mmol) in EtOH/H2O (10 mL/1 mL) were added Fe (79.4 mg, 1.42 mmol) and NH4Cl (76.0 mg, 1.42 mmol) . The reaction mixture was stirred at 90℃ for 2 hrs under N2 atmosphere. The reaction mixture was filtered, and the filtrate was concentrated to give the title compound.
LC-MS (ESI) [M+H] + 605.6
Step G: 1- (5- (9- ( ( (1S, 4S) -5- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-
yl) amino) phenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-azaspiro [5.5] undecane-3-
carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (5- (9- ( ( (1S, 4S) -5- (4-aminophenyl) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4(1H, 3H) -dione (140 mg) in DCM (10 mL) was added 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (92.3 mg, 0.231 mmol) , DIPEA (89.6 mg, 0.693 mmol) , the mixture was stirred at 25℃ for 16 hrs under N2 atmosphere. The mixture was quenched by addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%NH3/H2O, B: ACN ; flow rate gradient: 17-27%Retention Time: 9-19 of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 940.9
1H NMR (400 MHz, DMSO) δ 10.50 (s, 1H) , 10.02 (s, 1H) , 8.78 (s, 1H) , 7.87 (d, J = 8.2 Hz, 1H) , 7.69 (d, J = 8.2 Hz, 1H) , 7.62 (d, J = 8.2 Hz, 1H) , 7.52 (t, J = 9.7 Hz, 3H) , 7.37 (dd, J = 8.2, 1.9 Hz, 1H) , 6.55 (d, J = 8.2 Hz, 2H) , 5.73 –5.60 (m, 1H) , 5.04 (s, 1H) , 4.99 (d, J = 9.2 Hz, 1H) , 4.85 (d, J = 17.2 Hz, 1H) , 4.74 (s, 1H) , 4.58 (s, 1H) , 4.26 (s, 1H) , 3.73 (s, 1H) , 3.67 –3.49 (m, 3H) , 3.45 (s, 1H) , 3.30 –3.21 (m, 2H) , 3.10 (d, J = 8.8 Hz, 1H) , 2.95 (dd, J = 15.7, 9.7 Hz, 1H) , 2.84 (d, J = 8.4 Hz, 1H) , 2.80 –2.70 (m, 3H) , 2.41 (d, J = 9.4 Hz, 1H) , 2.30 (d, J = 21.8 Hz, 2H) , 2.23 –2.14 (m, 1H) , 2.02 (dd, J = 11.9, 6.7 Hz, 1H) , 1.89 (dd, J = 13.7, 7.3 Hz, 1H) , 1.80 (s,
1H) , 1.75 (d, J = 8.0 Hz, 1H) , 1.73 –1.61 (m, 3H) , 1.49 (s, 3H) , 1.40 (s, 1H) , 1.32 (s, 1H) , 1.24 (s, 3H) , 1.02 (s, 4H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 76: 1- [2-chloro-5- [9- [ [4- [4- [ [ (12E, 16R) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-yl] amino] phenyl] piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] phenyl] hexahydropyrimidine-2, 4-dione (Cpd-076)
Cpd-076 was synthesized according to the following scheme:
Step A: (12E, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one
To a solution of (12E, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfanyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (80 mg, 206.49 μmol) in THF (0.8 mL) and water (0.8 mL) was added Oxone (317.35 mg, 516.22 μmol) in portions at 0 ℃. After addition, the mixture was stirred at 0-5 ℃ for 1 hr. The crude residue was dissolved in EtOAc (30 mL) , washed with water (10 mL) , the organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure to obtain the title compound, which was used in next step without purification
LCMS: MS (ESI) m/z = 419.9 [M+H] +.
Step B: 1- [2-chloro-5- [9- [ [4- [4- [ [ (12E, 16R) -12-fluoro-16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-
yl] amino] phenyl] piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-
carbonyl] phenyl] hexahydropyrimidine-2, 4-dione
To a solution of 1- [5- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] -2-chloro-phenyl] hexahydropyrimidine-2, 4-dione (43 mg, 72.49 μmol) and (12E, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (39.53 mg, 94.24 μmol) in n-BuOH (1.5 mL) and the mixture was stirred at 100 ℃ for 4 hrs. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC (column: Phenomenex C18 75*30mm*3um; mobile phase: [water (FA) -ACN] ; gradient: 12 %-52%B over 9 min) to afford the title compound.
LCMS: MS m/z = 932.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 10.52 (s, 1H) , 10.27 -10.00 (m, 1H) , 8.84 (s, 1H) , 8.09 -7.98 (m, 1H) , 7.81 (d, J = 8.0 Hz, 1H) , 7.68 -7.51 (m, 5H) , 7.39 (br d, J = 8.0 Hz, 1H) , 6.90 (br d, J =8.8 Hz, 2H) , 5.33 (s, 1H) , 5.15 -4.98 (m, 1H) , 4.71 -4.54 (m, 2H) , 3.83 -3.69 (m, 2H) , 3.65 -3.54 (m, 4H) , 3.07 (br s, 4H) , 2.80 -2.70 (m, 2H) , 2.46 (br d, J = 6.8 Hz, 4H) , 2.19 -2.12 (m, 2H) , 1.90 (br d, J = 9.6 Hz, 2H) , 1.75 -1.63 (m, 3H) , 1.61 -1.43 (m, 8H) , 1.39 -1.21 (m, 3H) , 1.16 -0.96 (m, 4H) .
Example 77: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) -2-fluorophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-077)
Cpd-077 was synthesized according to the following scheme:
Step A: tert-butyl 4- (2-fluoro-4-nitrophenyl) piperazine-1-carboxylate
To a solution of 1, 2-difluoro-4-nitrobenzene (3 mL, 26.8 mmol) and tert-butyl piperazine-1-carboxylate (5 g, 26.8 mmol) in DMF (30 mL) was added K2CO3 (11.1 g, 80.5 mmol) . The reaction mixture was stirred at 100 ℃ for 16 hrs. The reaction mixture was filtered and the filter cake was washed by water (300 ml) and EtOAc (300 mL) . The filtrate was diluted with water (300 mL) and the solution was extracted with EtOAc (300 mL x 3) . The combined organic layers was washed with brine (300 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to afford the title compound.
LC-MS (ESI) [M+H-Boc] + = 226.1
Step B: 1- (2-fluoro-4-nitrophenyl) piperazine
A solution of tert-butyl 4- (2-fluoro-4-nitrophenyl) piperazine-1-carboxylate (1 g, 3.08 mmol) in HCl in dioxane (10 mL, 4 M) and DCM (10 mL) was stirred at 20℃ for 1 hr. After completion of reaction, the reaction mixture was concentrated under reduced pressure to dryness to afford the title compound.
LC-MS (ESI) [M+H] + = 226.0
Step C: tert-butyl 9- ( (4- (2-fluoro-4-nitrophenyl) piperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carboxylate
A mixture of 1- (2-fluoro-4-nitrophenyl) piperazine (200 mg) , tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (250 mg, 0.888 mmol) , NaBH (OAc) 3 (377 mg, 1.78 mmol) , CH3COOH (107 mg, 1.78 mmol) in DCM (5 mL) and MeOH (5 mL) was stirred at 25℃for 12 hrs. The reaction mixture was quenched with water (50 mL) and extracted with EtOAc (50 mL x 3) . The combined organic layers was washed with brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography eluted with PE: EA=1: 1 to afford the title compound.
LC-MS (ESI) [M+H] + = 491.7
Step D: 9- ( (4- (2-fluoro-4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane
To a solution of tert-butyl 9- ( (4- (2-fluoro-4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate (191 mg, 0.390 mmol) in DCM (2 mL) was added HCl in dioxane (2 mL, 4 M) and the mixture was stirred at 20℃ for 1 hr. After completion of reaction, the reaction mixture was concentrated under reduced pressure to dryness to afford the title compound.
LC-MS (ESI) [M+H] + = 391.2
Step E: 1- (2-chloro-5- (9- ( (4- (2-fluoro-4-nitrophenyl) piperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 9- ( (4- (2-fluoro-4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane (150 mg) , 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoic acid (103 mg, 0.384 mmol) , HATU (219 mg, 0.576 mmol) in DMF (2 mL) was added DIEA (149 mg, 1.15 mmol) , Then the mixture was stirred at 20℃ for 2hrs. After completion of reaction, Water (20 mL) was added and the solution was extracted with EtOAc (20 mL x 3) . The combined organic layers was washed with brine (20 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography eluted with DCM: MeOH=10: 1 to give the title compound.
LC-MS (ESI) [M+H] + = 641.3
Step F: 1- (5- (9- ( (4- (4-amino-2-fluorophenyl) piperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
A mixture of 1- (2-chloro-5- (9- ( (4- (2-fluoro-4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (176 mg, 0.275 mmol) , NH4Cl (73.5 mg, 1.375 mmol) , in H2O (2 mL) and EtOH (20 mL) were added Fe (77.0 mg, 1.375 mmol) was stirred at 90℃ for 2 hrs. After completion of reaction, the mixture was filtered through a Celite pad, water (200 mL) was added to the filtrate and the mixture was extracted with EtOAc (200 mL x 3) . The combined organic layers was washed with brine (200 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to dryness to afford the title compound.
LC-MS (ESI) [M+H] + = 611.3
Step G: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-
cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) -2-
fluorophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-
chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (20 mg, 0.0501 mmol) in n-Butanol (5 mL) was added 1- (5- (9- ( (4- (4-amino-2-fluorophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (45.9 mg, 0.0752 mmol and DIEA (6.47 mg, 0.0501 mmol) . The mixture was stirred at 100℃ for 16 hrs under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda, Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 24%~34%; Retention Time: 10.5-11.5min of 16 min) to give (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) -2-fluorophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (8.45 mg, 17.8 %yield) as a yellow solid.
LC-MS (ESI) [M+H] + 946.5
1H NMR (400 MHz, DMSO) δ 10.51 (s, 1H) , 10.36 –10.20 (m, 1H) , 8.89 (s, 1H) , 8.21 (s, 1H) , 7.90 (d, J = 7.8 Hz, 1H) , 7.70 (d, J = 8.4 Hz, 2H) , 7.64 (d, J = 8.2 Hz, 1H) , 7.56 (s, 1H) , 7.39 (d, J = 8.9 Hz, 2H) , 7.01 (t, J = 9.5 Hz, 1H) , 5.68 (d, J = 6.0 Hz, 1H) , 5.07 (s, 1H) , 5.00 (d, J = 10.2 Hz, 1H) , 4.86 (d, J = 16.5 Hz, 1H) , 4.80 –4.71 (m, 1H) , 4.58 (s, 1H) , 3.75 (s, 1H) , 3.60 (s, 3H) , 3.25 –3.18 (m, 2H) , 2.97 (s, 5H) , 2.76 (s, 3H) , 2.47 –2.38 (m, 4H) , 2.18 (s, 3H) , 2.03 (s, 1H) ,
1.91 (s, 1H) , 1.71 (d, J = 7.0 Hz, 3H) , 1.60 (s, 5H) , 1.29 (s, 2H) , 1.10 (s, 4H) , 0.88 (t, J = 7.4 Hz, 3H) .
Example 78: 3- (7- (2- ( (4- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) piperazin-1-yl) methyl) -7-azaspiro [3.5] nonan-7-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (Cpd-078)
Cpd-078 was synthesized according to the following scheme:
Step A: 7- (2- (dimethoxymethyl) -7-azaspiro [3.5] nonan-7-yl) phthalazin-1 (2H) -one
To a solution of 7-bromophthalazin-1 (2H) -one (120 mg, 0.536 mmol) in dioxane (15 ml) was added 2- (dimethoxymethyl) -7-azaspiro [3.5] nonane (107 mg, 0.536 mmol) , Ruphos-Pd-G2 (41.6 mg, 0.0536 mmol) , t-BuONa (154 mg, 1.60 mmol) . Then the reaction mixture was stirred at 100 ℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched by H2O (10 mL) and extracted with EA (3*20 mL) . The organic layers were washed with brine (10 mL) , dried by Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA=0: 1) to give the title compound.
ESI [M+H] + =344.1
Step B: 3- (7- (2- (dimethoxymethyl) -7-azaspiro [3.5] nonan-7-yl) -1-oxophthalazin-2 (1H) -
yl) piperidine-2, 6-dione
To a solution of 6- (2- (dimethoxymethyl) -7-azaspiro [3.5] nonan-7-yl) phthalazin-1 (2H) -one (30.0 mg, 0.0874 mmol) in DMF (4 mL) was added NaH (7.00 mg, 0.175 mmol, 60%w/w) , the reaction mixture was stirred at 25 ℃ for 2 hrs under N2 atmosphere. Then 3-bromopiperidine-2, 6-dione (16.7 mg, 0.0874 mmol) was added to the mixture, the reaction mixture was stirred at 25 ℃ for 2 hrs . After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EA (3 × 10 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA=1: 2) to give the title compound.
ESI [M+H] + =455.2
Step C: 7- (3- (2, 6-dioxopiperidin-3-yl) -4-oxo-3, 4-dihydrophthalazin-6-yl) -7-
azaspiro [3.5] nonane-2-carbaldehyde
A solution of 3- (7- (2- (dimethoxymethyl) -7-azaspiro [3.5] nonan-7-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (10.0 mg, 0.0220 mmol) in DCM/FA (1 mL/1 mL ) was stirred at 25℃for 2 hrs under N2 atmosphere. After completion of reaction, the reaction mixture was concentrated to give the title compound, which was used directly for the next step without further purification.
LC-MS (ESI) [M+H] + 409.2
Step D: 3- (7- (2- ( (4- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17-
(azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-
yl) amino) phenyl) piperazin-1-yl) methyl) -7-azaspiro [3.5] nonan-7-yl) -1-oxophthalazin-
2 (1H) -yl) piperidine-2, 6-dione
To a solution of 7- (3- (2, 6-dioxopiperidin-3-yl) -4-oxo-3, 4-dihydrophthalazin-6-yl) -7-azaspiro [3.5] nonane-2-carbaldehyde (30.0 mg, 0.0735 mmol) , (R, Z) -12-hydroxy-12-methyl-2- ( (4- (piperazin-1-yl) phenyl) amino) -7, 10, 11, 12-tetrahydro-5H-13, 17-
(azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (36.6 mg, 0.0735 mmol) in DCM (1 mL) and MeOH (1 mL) was stirred at 25℃ for 0.5 h. Then NaBH (OAc) 3 (23.5 mg, 0.111 mmol) was added. The mixture was stirred at 25℃ for 2 hrs. The mixture was quenched by addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 17-27%Retention Time: 7.3-9.1 of 16 min) to give the title compound.
LC-MS (ESI) [M+H] + 891.4
1H NMR (400 MHz, DMSO) δ 11.00 (s, 1H) , 10.07 (s, 1H) , 8.82 (s, 1H) , 8.26 (s, 1H) , 8.23 (s, 1H) , 8.01 (s, 1H) , 7.75 (d, J = 8.9 Hz, 2H) , 7.62 –7.52 (m, 4H) , 7.48 (s, 1H) , 6.90 (d, J = 9.0 Hz, 2H) , 5.75 (s, 1H) , 5.27 (s, 3H) , 4.46 (s, 1H) , 3.42 (s, 5H) , 3.07 (s, 5H) , 2.92 (s, 2H) , 2.70 –2.54 (m, 2H) , 2.44 (s, 4H) , 2.10 (s, 2H) , 1.99 (s, 4H) , 1.70 (s, 3H) , 1.56 (s, 5H) , 1.49 (s, 3H) .
Example 79: 1- (2-chloro-5- (9- ( ( (R) -4- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) -2-methylpiperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-079)
Cpd-079 was synthesized according to the following scheme:
Step A: 1- (2-chloro-5- (9- ( ( (R) -4- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-
tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-
yl) amino) phenyl) -2-methylpiperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-
carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (R) -1- (5- (9- ( (4- (4-am) inophenyl) -2-methylpiperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl-2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (50 mg, 0.08 mmol) and (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (31.76 mg, 0.08 mmol) in DCM (5 mL) was added DIPEA (31.0 mg, 0.24 mmol) . The reaction mixture was stirred at 25 ℃ for 24 hrs. The reaction mixture was concentrated and purified by prep-HPLC (Waters 2767/Qda, Column: Atlantis TM T3 Prep OBD TM, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 30%~40%; Retention Time: 7.8-9.6min of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 928.2.
1H NMR (400 MHz, DMSO) δ 10.50 (s, 1H) , 10.14 –9.83 (m, 1H) , 8.82 (s, 1H) , 8.42 (s, 1H) , 8.02 (s, 1H) , 7.75 (d, J = 8.3 Hz, 1H) , 7.63 (d, J = 8.3 Hz, 1H) , 7.60 –7.49 (m, 4H) , 7.39 (d, J =8.2 Hz, 1H) , 6.89 (d, J = 9.5 Hz, 2H) , 5.26 (s, 3H) , 4.46 (s, 2H) , 3.75 (s, 1H) , 3.62 (s, 3H) , 2.90 (s, 1H) , 2.75 (s, 3H) , 2.67 (s, 1H) , 2.33 (s, 1H) , 2.25 (d, J = 27.1 Hz, 3H) , 1.94 (s, 4H) , 1.69 (s, 4H) , 1.56 (s, 3H) , 1.49 (s, 4H) , 1.40 –1.33 (m, 1H) , 1.24 (s, 5H) , 1.11 (s, 2H) , 1.04 (s, 4H) .
Example 80: (R, Z) -1- (5- (9- ( (1- (4- ( (12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-
yl) amino) phenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-080)
Cpd-080 was synthesized according to the following scheme:
Step A: 1- (2-methoxy-5- (9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-
diazaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
A solution of 3- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane (200 mg, 0.54 mmol) , perfluorophenyl 3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) -4-methoxybenzoate (277.27 mg, 0.64 mmol) and DIEA (139.58 mg, 1.08 mmol) in DMF (10 mL) was stirred at 25℃ for 2hrs. The solution was added water and extracted with ethyl acetate (20 mL x 3) . The organic layer was dried and concentrated. The residue was purified by silica gel chromatography (eluted with petroleum ether: ethyl acetate =2: 1 to 1: 1) to give the title compound.
LC-MS (ESI) [M+H] + 619.5.
Step B: 1- (5- (9- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-
carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a stirred solution of 1- (2-methoxy-5- (9- ( (1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (230 mg, 0.37 mmol) in EtOH (10 mL) and water (2 mL) was added iron powder (104 mg, 1.86 mmol) and ammonium chloride (99.49 mg, 1.86 mmol) at room temperature. The mixture was stirred at 90℃ for 30 min. The reaction mixture was diluted with DCM (20 mL) . The organic layer was washed with water (2 x 10 mL) , brine (20 mL) , dried by anhydrous Na2SO4 and concentrated to afford the title compound.
LC-MS (ESI) [M+H] + 589.3.
Step C: (R, Z) -1- (5- (9- ( (1- (4- ( (12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17-
(azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-
yl) amino) phenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carbonyl) -2-
methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (5- (9- ( (1- (4-aminophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (50 mg, 0.085 mmol) , (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (32.73 mg, 0.085 mmol) in DCM (5 mL) was added DIEA (32.96 mg, 0.26 mmol) . The reaction mixture was stirred at 25℃ for 12 hrs. The reaction mixture was concentrated and purified by prep-HPLC (Waters 2767/Qda, Column: Atlantis TM T3 Prep OBD TM, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 35%~50%; Retention Time: 7.5-9.3min of 16 min) to afford the title compound.
LC-MS (ESI) [M+H] + 910.3.
1H NMR (400 MHz, DMSO) δ 10.33 (s, 1H) , 10.04 (s, 1H) , 8.81 (s, 1H) , 8.21 (s, 1H) , 8.01 (s, 1H) , 7.75 (d, J = 8.1 Hz, 1H) , 7.56 (dd, J = 14.8, 8.2 Hz, 3H) , 7.37 (dd, J = 8.5, 2.1 Hz, 1H) , 7.32 (d, J = 2.1 Hz, 1H) , 7.15 (d, J = 8.6 Hz, 1H) , 6.89 (d, J = 9.2 Hz, 2H) , 5.26 (s, 3H) , 4.46 (s, 2H) , 3.84 (s, 3H) , 3.60 (t, J = 6.7 Hz, 4H) , 3.37 (s, 4H) , 2.68 (t, J = 6.4 Hz, 2H) , 2.59 (t, J = 11.7 Hz, 2H) , 2.33 (s, 4H) , 2.16 (d, J = 6.8 Hz, 2H) , 1.95 (d, J = 12.7 Hz, 2H) , 1.76 (d, J = 9.1 Hz, 3H) , 1.56 (s, 3H) , 1.49 (s, 4H) , 1.41 (s, 4H) , 1.27 –1.15 (m, 4H) .
Example 81: 3- [5- [4- [ [3- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-yl] amino] phenyl] -3, 9-diazaspiro [5.5] undecan-9-yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (Cpd-081)
Cpd-081 was synthesized according to the following scheme:
Step A: (12Z, 16R) -16-hydroxy-16-methyl-5- [4- [3- (2, 2, 2-trifluoroacetyl) -3, 9-
diazaspiro [5.5] undecan-9-yl] anilino] -2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one
To a solution of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (148.17 mg, 0.369 mmol) , 1- [9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl] -2, 2, 2-trifluoro-ethanone (120 mg, 351.52 μmol) in n-BuOH (5 mL) . The resulting mixture was stirred at 100 ℃for 2 hr. The mixture was concentrated to give a residue, which was purified by column chromatography on silica gel (24%MeOH in DCM) to give the title compound.
LCMS: MS (ESI) m/z = 663.3 [M+H] +
Step B: (12Z, 16R) -5- [4- (3, 9-diazaspiro [5.5] undecan-3-yl) anilino] -16-hydroxy-16-methyl-
2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-
one
To a solution of (12Z, 16R) -16-hydroxy-16-methyl-5- [4- [3- (2, 2, 2-trifluoroacetyl) -3, 9-diazaspiro [5.5] undecan-9-yl] anilino] -2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (200 mg, 301.79 μmol) in MeOH (3 mL) was added K2CO3 (83.42 mg, 603.59 μmol) . The mixture was stirred at 20 ℃ for 2 hr. The mixture was poured into water (30 mL) and extracted with EtOAc (30 mL × 2) . The combined organic layers were washed with brine (30 mL × 2) , dried over Na2SO4, filtered and concentrated to give the title compound.
LCMS: MS (ESI) m/z = 567.3 [M+H] +
Step C: 3- [5- [4- [ [3- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
yl] amino] phenyl] -3, 9-diazaspiro [5.5] undecan-9-yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-
yl] piperidine-2, 6-dione
To a solution of (12Z, 16R) -5- [4- (3, 9-diazaspiro [5.5] undecan-3-yl) anilino] -16-hydroxy-16-methyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (180 mg, 317.63 μmol) , 1- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde (135.46 mg, 381.16 μmol) , TEA (96.42 mg, 952.89 μmol) in DCM (3 mL) was added NaBH (OAc) 3 (201.96 mg, 952.89 μmol) . The mixture was stirred at 20 ℃ for 3 hr. The mixture was poured into water (20 mL) and extracted with DCM (20 mL × 2) . The combined organic layers were washed with brine (20 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by prep-HPLC (column: Phenomenex C18 75*30mm*3um; mobile phase: [water (FA) -ACN] ; B%: 6%-46%, 9 min) and dried by lyophilization to give the title compound.
LCMS: MS (ESI) m/z = 906.5 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ = 10.95 (s, 1H) , 10.05 (s, 1H) , 8.81 (s, 1H) , 8.20 (FA, 0.23H) , 7.98–8.05 (s, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.51–7.62 (m, 3H) , 7.49 (d, J = 9.2 Hz, 1H) , 7.00–7.08 (m, 2H) , 6.90 (d, J = 8.8 Hz, 2H) , 5.15–5.50 (m, 3H) , 5.04 (dd, J = 13.6, 5.2 Hz, 1H) , 4.40–4.55 (m, H) , 4.14–4.35 (m, 2H) , 3.86 (d, J = 12.4 Hz, 2H) , 3.07 (s, 4H) , 2.75–2.94 (m, 3H) , 2.55–2.68 (m, 2H) , 2.33 (s, 5H) , 2.16 (d, J = 6.0 Hz, 3H) , 2.00–2.08 (m, 1H) , 1.90–1.99 (m, 2H) , 1.72–1.79 (m, 3H) , 1.43–1.58 (m, 11H) , 1.16 (d, J = 11.6 Hz, 2H) .
Example 82: 8- (1- (1- ( (3- (4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoyl) -3-azaspiro [5.5] undecan-9-yl) methyl) piperidin-4-yl) -1H-pyrazol-4-yl) -3- (2, 6-dichlorophenyl) -1-methyl-1, 7-dihydro-2H-pyrrolo [3', 2': 5, 6] pyrido [4, 3-d] pyrimidine-2, 4 (3H) -dione (Cpd-082)
Compound 82 was prepared using the similar procedure as described in Example 72.
LCMS: MS (ESI) m/z 927.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ: 12.83 (s, 1H) , 10.52 (s, 1H) , 8.96-9.27 (TFA, 1H) , 8.78 (s, 1H) , 8.34-8.54 (m, 1H) , 8.13-8.20 (m, 1H) , 7.73 (s, 1H) , 7.71 (s, 1H) , 7.65 (d, J = 8.4 Hz, 1H) , 7.56-7.61 (m, 1H) , 7.55 (s, 1H) , 7.39 (d, J = 8.4 Hz, 1H) , 7.34 (d, J = 1.6 Hz, 1H) , 4.53 (d, J =8.6 Hz, 1H) , 4.00 (s, 3H) , 3.71-3.80 (m, 1H) , 3.64 (t, J = 6.0 Hz, 2H) , 3.52-3.60 (m, 2H) , 3.42-3.51 (m, 2H) , 3.15 (s, 4H) , 3.01 (s, 2H) , 2.75 (t, J = 6.0 Hz, 2H) , 2.26-2.33 (m, 3H) , 1.69-1.86 (m, 3H) , 1.58-1.67 (m, 2H) , 1.42-1.56 (m, 2H) , 1.25-1.40 (m, 2H) , 1.09-1.24 (m, 4H) .
Example 83: 3- (6- (2- ( (4- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) piperazin-1-yl) methyl) -7-azaspiro [3.5] nonan-7-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (Cpd-083)
Compound 83 was prepared using the similar procedure as described in Example 78.
LCMS: MS (ESI) m/z 891.6 [M+H] +.
1H NMR (400 MHz, DMSO) δ 10.99 (s, 1H) , 10.04 (s, 1H) , 8.82 (s, 1H) , 8.38 (s, 1H) , 8.22 (s, 1H) , 8.00 (d, J = 9.0 Hz, 2H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.57 (t, J = 8.1 Hz, 3H) , 7.48 (d, J = 9.4 Hz, 1H) , 7.24 (s, 1H) , 6.90 (d, J = 9.2 Hz, 2H) , 5.74 (dt, J = 12.2, 5.7 Hz, 1H) , 5.33 (ddd, J =25.2, 23.4, 9.9 Hz, 3H) , 4.51 –4.38 (m, 2H) , 3.46 –3.40 (m, 4H) , 3.21 (ddd, J = 14.2, 8.6, 3.3 Hz, 4H) , 3.07 (dd, J = 9.0, 5.3 Hz, 4H) , 2.90 (ddd, J = 11.5, 6.8, 2.8 Hz, 1H) , 2.68 –2.56 (m, 2H) , 2.44 –2.41 (m, 2H) , 2.22 –2.13 (m, 1H) , 2.12 –1.90 (m, 6H) , 1.70 (ddd, J = 10.8, 6.6, 2.6 Hz, 3H) , 1.60 –1.54 (m, 5H) , 1.48 (t, J = 9.9 Hz, 2H) .
Example 84: (R, Z) -1- (2-chloro-4-fluoro-5- (9- ( (4- (4- ( (12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-084)
Compound 84 was prepared using the similar procedure as described in Example 35.
LCMS: MS (ESI) m/z 891.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.52 (s, 1H) , 10.03 (s, 1H) , 8.82 (s, 1H) , 8.14 (s, 1H) , 8.01 (d, J = 7.4 Hz, 1H) , 7.76 (d, J = 8.0 Hz, 1H) , 7.71 (dd, J = 8.9, 3.3 Hz, 1H) , 7.63 –7.54 (m, 4H) , 6.90 (d, J = 7.6 Hz, 2H) , 5.36 –5.25 (m, 2H) , 4.47 –4.44 (m, 1H) , 3.73 (dd, J = 14.7, 8.0 Hz, 1H) , 3.65 –3.36 (m, 4H) , 3.29 –3.17 (m, 4H) , 3.08 (s, 4H) , 2.74 (dd, J = 12.4, 6.1 Hz, 2H) , 2.69 –2.53 (m, 1H) , 2.46 –2.31 (m, 2H) , 2.24 –2.12 (m, 3H) , 2.05 –1.89 (m, 2H) , 1.77 –1.67 (m, 3H) , 1.60 –1.49 (m, 7H) , 1.44 –1.25 (m, 3H) , 1.07 (dt, J = 23.7, 11.1 Hz, 4H) .
Example 85: 3- (4-fluoro-7- (2- ( (4- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) piperazin-1-yl) methyl) -7-azaspiro [3.5] nonan-7-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (Cpd-085) and 3- (4-fluoro-6- (2- ( (4- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) piperazin-1-yl) methyl) -7-azaspiro [3.5] nonan-7-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (Cpd-086) (mixture)
Compounds 85 and 86 were prepared using the similar procedure as described in Example 78, using a mixture of 7-bromo-4-fluorophthalazin-1 (2H) -one and 4, 6-difluorophthalazin-1 (2H) -one as starting material.
LCMS: MS (ESI) m/z 909.4 [M+H] +.
1H NMR (400 MHz, DMSO) δ 11.03 (s, 1H) , 10.04 (s, 1H) , 8.82 (s, 1H) , 8.23 (s, 1H) , 8.09 –7.97 (m, 2H) , 7.75 (d, J = 8.1 Hz, 1H) , 7.57 (t, J = 8.1 Hz, 4H) , 7.09 (d, J = 2.1 Hz, 1H) , 6.89 (d, J = 9.2 Hz, 2H) , 5.73 (dd, J = 12.4, 5.2 Hz, 1H) , 5.46 –5.18 (m, 3H) , 4.44 (dd, J = 14.1, 7.5 Hz, 2H) , 3.48 (dd, J = 10.1, 5.0 Hz, 3H) , 3.40 (dd, J = 8.8, 6.8 Hz, 5H) , 3.07 (s, 4H) , 2.91 (ddd, J =18.9, 13.8, 5.4 Hz, 1H) , 2.62 (ddd, J = 13.8, 10.6, 7.5 Hz, 2H) , 2.43 (t, J = 5.8 Hz, 3H) , 2.12 –2.03 (m, 2H) , 2.03 –1.95 (m, 3H) , 1.95 –1.89 (m, 1H) , 1.74 –1.65 (m, 3H) , 1.54 (d, J = 9.8 Hz, 5H) , 1.48 (t, J = 9.9 Hz, 3H) .
Example 86: 1- (5- (9- ( (4- (4- (3- (2, 6-dichlorophenyl) -4-oxo-4, 7-dihydro-3H-pyrrolo [3', 2': 5, 6] pyrido [4, 3-d] pyrimidin-8-yl) -1H-pyrazol-1-yl) piperidin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-087)
Compound 87 was prepared using the similar procedure as described in Example 72.
LCMS: MS (ESI) m/z 946.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 12.67 (s, 1H) , 10.34 (s, 1H) , 8.96 (s, 1H) , 8.62 (s, 1H) , 8.43 (s, 1H) , 8.12 (d, J = 9.3 Hz, 1H) , 7.81 (d, J = 8.2 Hz, 2H) , 7.70 –7.62 (m, 1H) , 7.37 (d, J = 8.6 Hz, 1H) , 7.33 (s, 1H) , 7.15 (d, J = 8.2 Hz, 1H) , 7.11 (s, 1H) , 4.30 –4.15 (m, 1H) , 3.84 (s, 3H) , 3.60 (t, J = 6.6 Hz, 2H) , 3.03 –2.90 (m, 2H) , 2.72 –2.65 (m, 2H) , 2.19 (dd, J = 17.6, 8.6 Hz, 2H) , 2.13 –2.03 (m, 4H) , 2.04 –1.91 (m, 3H) , 1.75 –1.68 (m, 2H) , 1.58 (dd, J = 15.9, 7.7 Hz, 3H) , 1.54 –1.42 (m, 3H) , 1.39 –1.21 (m, 3H) , 1.17 –0.94 (m, 5H) .
Example 87: (R, Z) -1- (5- (9- (2- (4- (4- ( (12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) piperazin-1-yl) ethyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-088)
Compound 88 was prepared using the similar procedure as described in Example 62.
1HNMR (400 MHz, DMSO) δ 10.32 (s, 1H) , 10.03 (s, 1H) , 8.82 (s, 1H) , 8.23 (s, 1H) , 8.00 (d, J =7.3 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.57 (t, J = 8.1 Hz, 3H) , 7.34 (dd, J = 17.8, 5.2 Hz, 2H) , 7.15 (d, J =8.6 Hz, 1H) , 6.90 (d, J = 9.1 Hz, 2H) , 5.32 (s, 2H) , 4.45 (s, 1H) , 3.84 (s, 3H) , 3.59 (t, J = 6.8 Hz, 2H) , 3.39 (s, 4H) , 3.26 –3.17 (m, 2H) , 3.07 (s, 4H) , 2.68 (t, J = 6.5 Hz, 2H) , 2.49 –2.44 (m, 4H) , 2.33 (s, 2H) , 2.18 (s, 1H) , 2.03 (s, 1H) , 1.94 (s, 1H) , 1.68 (s, 3H) , 1.56 (s, 5H) , 1.48 (s, 2H) , 1.38 (s, 2H) , 1.29 (s, 3H) , 1.10 (s, 4H) .
Example 88: (R, Z) -1- (2-chloro-5- (9- (2- (4- (4- ( (12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) piperazin-1-yl) ethyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-089)
Compound 89 was prepared using the similar procedure as described in Example 62.
1HNMR (400 MHz, DMSO) δ 10.51 (s, 1H) , 10.04 (s, 1H) , 8.81 (d, J = 8.3 Hz, 1H) , 8.19 (s, 1H) , 8.00 (d, J = 7.8 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.63 , J = 8.2 Hz, 1H) , 7.56 (dd, J = 14.2, 4.7 Hz, 4H) , 7.38 (dd, J = 8.2, 1.9 Hz, 1H) , 6.89 (d, J = 9.1 Hz, 2H) , 5.27 (s, 3H) , 4.44 (s, 2H) , 3.74 (s, 1H) , 3.69-3.48 (m, 4H) , 3.07 (s, 4 H) , 2.74 (d, J = 3.8 Hz, 2H) , 2.33 (s, 2H) , 2.17 (s, 1H) ,
2.08-1.87 (m, 2H) , 1.69 (d, J = 8.7 Hz, 3H) , 1.59-1.48 (m, 6H) , 1.38 (s, 4H) , 1.23 (s, 3H) , 1.08 (s, 4H)
Example 89: 3- (6- (2- ( (4- (4- ( (2-allyl-1- ( (R) -7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) -7-azaspiro [3.5] nonan-7-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (Cpd-090)
Compound 90 was prepared using the similar procedure as described in Example 78.
1H NMR (400 MHz, DMSO-d6) δ 10.98 (s, 1H) , 10.08 (s, 1H) , 8.81 (s, 1H) , 8.30 (s, 1H) , 8.22 (s, 1H) , 8.06 –7.88 (m, 2H) , 7.69 (d, J = 8.1 Hz, 1H) , 7.62 –7.43 (m, 3H) , 7.24 (s, 1H) , 6.91 (d, J = 8.7 Hz, 2H) , 5.70 (dd, J = 25.3, 9.4 Hz, 2H) , 5.00 (d, J = 9.8 Hz, 2H) , 4.90 –4.70 (m, 2H) , 4.59 (s, 1H) , 3.43 (s, 8H) , 3.08 (s, 4H) , 3.05 –2.85 (m, 3H) , 2.80 (s, 1H) , 2.65 (d, J = 16.8 Hz, 2H) , 2.44 (s, 2H) , 2.20 (s, 1H) , 2.00 (d, J = 9.1 Hz, 4H) , 1.88 (s, 1H) , 1.70 (s, 3H) , 1.57 (s, 2H) , 1.50 (d, J = 9.2 Hz, 2H) , 0.87 (t, J = 7.2 Hz, 3H) .
Example 90: (R) -1- (5- (9- ( (4- (4- ( (2-allyl-1- (7-ethyl-7-hydroxy-6, 7-dihydro-5H-cyclopenta [b] pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-6-yl) amino) phenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chloro-4-fluorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-091)
Compound 91 was prepared using the similar procedure as described in Example 31.
LCMS: MS (ESI) m/z 891.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.51 (s, 1H) , 10.10 (s, 1H) , 8.81 (s, 1H) , 8.31 (s, 1H) , 7.92 (dt, J = 7.5, 3.3 Hz, 1H) , 7.70 (dd, J = 11.1, 5.6 Hz, 2H) , 7.63 –7.51 (m, 3H) , 6.91 (d, J = 8.2 Hz, 2H) , 5.72 –5.61 (m, 1H) , 5.04 (s, 1H) , 4.99 (d, J = 10.5 Hz, 1H) , 4.85 (d, J = 17.6 Hz, 1H) , 4.79 –4.70 (m, 1H) , 4.57 (dd, J = 11.8, 6.5 Hz, 1H) , 3.79 –3.68 (m, 1H) , 3.65 –3.40 (m, 3H) , 3.25 –3.18 (m, 2H) , 3.08 (s, 4H) , 2.96 (dd, J = 15.8, 6.4 Hz, 1H) , 2.80 –2.66 (m, 3H) , 2.33 (s, 3H) , 2.22 –2.11 (m, 3H) , 2.06 –1.95 (m, 1H) , 1.89 (dd, J = 13.8, 7.2 Hz, 1H) , 1.75 –1.64 (m,
3H) , 1.62 –1.49 (m, 4H) , 1.45 –1.39 (m, 1H) , 1.36 (dd, J = 8.0, 4.6 Hz, 1H) , 1.28 (dt, J = 12.2, 6.0 Hz, 1H) , 1.22 –0.90 (m, 5H) , 0.87 (t, J = 7.4 Hz, 3H) .
Example 91: 1- (2-chloro-5- (9- ( (4- (4- (3- (2, 6-dichlorophenyl) -1-methyl-4-oxo-2, 3, 4, 7-tetrahydro-1H-pyrrolo [3', 2': 5, 6] pyrido [4, 3-d] pyrimidin-8-yl) -1H-pyrazol-1-yl) piperidin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (Cpd-092)
Compound 92 was prepared using the similar procedure as described in Example 72.
LCMS: MS (ESI) m/z 913.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 8.43 (s, 1H) , 8.28 (s, 1H) , 7.97 (s, 1H) , 7.64 (dd, J = 8.2, 3.2 Hz, 3H) , 7.55 (d, J = 1.8 Hz, 1H) , 7.50 –7.44 (m, 1H) , 7.39 (d, J = 10.0 Hz, 1H) , 6.96 (s, 1H) , 4.95 (s, 2H) , 4.19 –4.09 (m, 1H) , 3.78 –3.71 (m, 1H) , 3.66 –3.54 (m, 4H) , 3.45 (s, 3H) , 2.91 (d, J = 10.3 Hz, 2H) , 2.77 –2.72 (m, 2H) , 2.16 (s, 2H) , 2.04 (d, J = 11.6 Hz, 4H) , 1.92 (d, J =10.8 Hz, 2H) , 1.76 –1.68 (m, 3H) , 1.58 (d, J = 9.6 Hz, 2H) , 1.49 (ddd, J = 12.6, 7.6, 3.9 Hz, 3H) , 1.39 –1.32 (m, 1H) , 1.26 (ddd, J = 14.4, 8.5, 4.8 Hz, 2H) , 1.15 –1.00 (m, 5H) .
Example 92:
3- (5- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -7-aza-7-spiro [3.5] nonyl} -1, 3-dioxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-096)
Scheme:
Step A: tert-butyl 2- [4- (4-nitrophenyl) piperazin-1-yl] -7-azaspiro [3.5] nonane-7-carboxylate
To a solution of tert-butyl 2-oxo-7-azaspiro [3.5] nonane-7-carboxylate (2 g, 8.36 mmol) and 1-(4-nitrophenyl) piperazine (1.91 g, 9.19 mmol) in DCM (36 mL) was added AcOH (501.88 mg, 8.36 mmol) and then added NaBH (OAc) 3 (5.31 g, 25.07 mmol) . The mixture was stirred at 25 ℃for 1 hr. The crude residue was quenched with H2O (20mL) and dissolved in DCM (200 mL×3) . The organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure to obtain a residue. The crude product was triturated with EtOAc: PE=3: 1 to afford the desired product.
LCMS: MS (ESI) m/z 431.1 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.12 (d, J = 9.6 Hz, 2H) , 6.82 (d, J = 9.6 Hz, 2H) , 3.49 -3.26 (m, 8H) , 2.82 -2.68 (m, 1H) , 2.55 -2.37 (m, 4H) , 2.10 -2.00 (m, 2H) , 1.73 -1.62 (m, 2H) , 1.59 -1.50 (m, 4H) , 1.47 -1.43 (s, 9H)
Step B: 2- [4- (4-nitrophenyl) piperazin-1-yl] -7-azaspiro [3.5] nonane
To a solution of tert-butyl 2- [4- (4-nitrophenyl) piperazin-1-yl] -7-azaspiro [3.5] nonane-7-carboxylate (2 g, 4.65 mmol) in DCM (10 mL) and TFA (10 mL) was stirred at 25 ℃ for 2hr. The reaction mixture was washed with water (50mL) and basified with Na2CO3. Then the
mixture was extracted with EtOAc (100 mL × 3) . The organic phase was dried over Na2SO4 and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 330.9 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.05 (d, J = 9.5 Hz, 2H) , 7.02 (d, J = 9.5 Hz, 2H) , 3.46 -3.42 (m, 1H) , 2.89 -2.81 (m, 4H) , 2.80 -2.74 (m, 1H) , 2.37 -2.31 (m, 4H) , 2.01 -1.94 (m, 2H) , 1.62 -1.53 (m, 4H) , 1.55 -1.49 (m, 2H)
Step C: 2- (2, 6-dioxo-3-piperidyl) -5- [2- [4- (4-nitrophenyl) piperazin-1-yl] -7-
azaspiro [3.5] nonan-7-yl] isoindoline-1, 3-dione
To a solution of 2- [4- (4-nitrophenyl) piperazin-1-yl] -7-azaspiro [3.5] nonane (890 mg, 2.69 mmol) and 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (744.00 mg, 2.69 mmol) in NMP (2 mL) was added DIEA (696.24 mg, 5.39 mmol) . The mixture was stirred at 100 ℃ for 2hr. The reaction mixture was quenched with ice water, and then extracted with DCM (100 mL×3) . The combined organic layers were washed with brine (100 mL×5) , dried over Na2SO4 filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (20gSilica Flash Column, Eluent of 0~20%DCM/DCM MeOH=10: 1 gradient @30 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 587.1 [M+H] +.
Step D: 5- [2- [4- (4-aminophenyl) piperazin-1-yl] -7-azaspiro [3.5] nonan-7-yl] -2- (2, 6-dioxo-3-
piperidyl) isoindoline-1, 3-dione
To solution of 2- (2, 6-dioxo-3-piperidyl) -5- [2- [4- (4-nitrophenyl) piperazin-1-yl] -7-azaspiro [3.5] nonan-7-yl] isoindoline-1, 3-dione (670 mg, 1.14 mmol) in EtOH (8 mL) and NH4Cl (2 mL) was added Fe (637.81 mg, 11.42 mmol) . The mixture was stirred at 80 ℃ for 2hr. The reaction mixture was added DCM (150 ml) and stirred at 25℃ for 30min. The mixture was filtered and the combined organic layers were concentrated to give a residue. The residue was purified by flash silica gel chromatography (20gSilica Flash Column, Eluent of 0~30%DCM/DCM: MeOH=10: 1 gradient @30 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 557.4 [M+H] +.
Step E: 3- (5- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -7-aza-7-spiro [3.5] nonyl} -1, 3-dioxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 5- [2- [4- (4-aminophenyl) piperazin-1-yl] -7-azaspiro [3.5] nonan-7-yl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (61.01 mg, 109.61 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-
1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (40 mg, 99.64 μmol) in t-BuOH (2 mL) was added TFA (50 mg, 438.51 μmol) . The mixture was stirred at 100 ℃ for 2hr. The mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC (column: Welch Xtimate C18 100*40mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 15%-45%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 878.5 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 11.09 (s, 1H) , 10.21 -10.03 (m, 1H) , 9.93 (m, 1H) , 8.84 (s, 1H) , 8.04 -7.96 (m, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.69 -7.57 (m, 4H) , 7.35 (s, 1H) , 7.26 (dd, J = 1.6, 8.8 Hz, 1H) , 7.00 (d, J = 9.2 Hz, 2H) , 5.42 -5.23 (m, 2H) , 5.07 (dd, J = 5.6, 12.8 Hz, 1H) , 4.56 -4.38 (m, 2H) , 3.93 -3.73 (m, 4H) , 3.52 -3.50 (m, 3H) , 3.07 -2.80 (m, 6H) , 2.65 -2.52 (m, 2H) , 2.32 -2.17 (m, 3H) , 2.11 -1.89 (m, 6H) , 1.82 -1.60 (m, 6H) , 1.56 (m, 3H)
Example 93:
3- (5- {4- [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] -1-piperidyl} -1, 3-dioxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-101)
Scheme:
Step A: tert-butyl 9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 3, 9-diazaspiro [5.5] undecane-3-carboxylate (3 g, 11.79 mmol) and 1-fluoro-4-nitro-benzene (1.83 g, 12.97 mmol, 1.38 mL) in DMF (15 mL) was added DIEA (3.05 g, 23.59 mmol, 4.11 mL) . The mixture was stirred at 90 ℃ for 1hr . The reaction was concentrated under reduced pressure to give a residue. The reduce was added water (100 mL) and exacted with EtOAc (100 mL×3) . The organic layers was washed with brine and dried with Na2SO4. The crude product was triturated with EtOAc: PE=3: 1 to afford the desired product. LCMS: MS (ESI) m/z 376.1 [M+H] +.
Step B: 3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane
To a solution of tert-butyl 9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane-3-carboxylate (3.4 g, 9.06 mmol) in DCM (20 mL) was added HCl/dioxane (4M, 20 mL) . The mixture was stirred at 25 ℃ for 2hr. The reaction mixture was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 275.9 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 8.93 (br s, 1H) , 8.03 (d, J = 9.6 Hz, 2H) , 7.01 (d, J = 9.6 Hz, 2H) , 4.73 (br s, 4H) , 3.03 (br s, 4H) , 1.69 -1.64 (m, 4H) , 1.59 -1.54 (m, 4H) 1
Step C: tert-butyl 4- [9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl] piperidine-1-
carboxylate
To a solution of 3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane (500 mg, 1.60 mmol, HCl) and tert-butyl 4-iodopiperidine-1-carboxylate (1.50 g, 4.81 mmol) in DMF (4 mL) was added K2CO3 (665 mg, 4.81 mmol) . The mixture was stirred at 130 ℃ for 16 hr. The reaction was concentrated under reduced pressure to give a residue. The reduce was added water (50 mL) and exacted with DCM (100mL×3) . The organic layer was washed with brine and dried with Na2SO4. The residue was purified by flash silica gel chromatography (20 gSilica Flash Column, Eluent of 0~45%DCM/DCM: MeOH=10: 1 gradient @30 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 459.1 [M+H] +.
Step D: 9- (4-nitrophenyl) -3- (4-piperidyl) -3, 9-diazaspiro [5.5] undecane
To a solution of tert-butyl 4- [9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl] piperidine-1-carboxylate (130 mg, 283.48 μmol) in DCM (2 mL) was added HCl/dioxane (4M, 2 mL) . The mixture was stirred at 25 ℃ for 1hr . The reaction mixture was added water (15 mL) and exacted with DCM (30 mL×3) . The organic layer was washed with brine and dried with Na2SO4 and was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 359.1 [M+H] +.
Step E: 2- (2, 6-dioxo-3-piperidyl) -5- [4- [9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl] -
1-piperidyl] isoindoline-1, 3-dione
To a solution of 9- (4-nitrophenyl) -3- (4-piperidyl) -3, 9-diazaspiro [5.5] undecane (150 mg, 418.44 μmol) and 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (132.92 mg, 481.20 μmol) in DMF (1 mL) was added DIEA (108.16 mg, 836.87 μmol, ) . The mixture was stirred at 90 ℃for 2hr. The reaction was added water (30 mL) and exacted with DCM (50 mL*3) . The organic layer was dried with Na2SO4 and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~80%DCM/DCM: MeOH=10: 1 gradient @30 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 615.1 [M+H] +.
Step F: 5- [4- [9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl] -1-piperidyl] -2- (2, 6-
dioxo-3-piperidyl) isoindoline-1, 3-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [4- [9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl] -1-piperidyl] isoindoline-1, 3-dione (58 mg, 94.36 μmol) in THF (10 mL) and dioxane (1 mL) was added Pd/C (1.00 g, 939.67 μmol, 10%purity) and the mixture was stirred at 25 ℃ for 16hr under H2 (15Psi) . The mixture was filtered and the organic phase was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 585.3 [M+H] +.
Step G: 3- (5- {4- [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] -1-piperidyl} -1, 3-dioxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 5- [4- [9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl] -1-piperidyl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (62 mg, 106.04 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (42.57 mg, 106.04 μmol) in t-BuOH (2 mL) was added TFA (48.36 mg, 424.14 μmol) . The mixture was stirred at 100 ℃ for 16hr. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC (column: Welch Xtimate C18 100*40mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 7%-37%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 906.6 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.10 (s, 1H) , 10.12 (m, 1H) , 9.13 (m, 1H) , 8.85 (s, 1H) , 8.00 (t, J = 7.6 Hz, 1H) , 7.77 -7.69 (m, 2H) , 7.67 -7.57 (m, 3H) , 7.42 (s, 1H) , 7.36 -7.30 (m, 1H) , 7.08 (br s, 2H) , 5.43 -5.24 (m, 2H) , 5.08 (dd, J = 5.2, 12.9 Hz, 1H) , 4.47 (br d, J = 5.2 Hz, 2H) , 4.25 (d, J = 12.8 Hz, 2H) , 3.18 (br d, J = 1.2 Hz, 4H) , 3.09 (br d, J = 11.6 Hz, 2H) , 3.00 (br t, J =12.4 Hz, 3H) , 2.94 -2.83 (m, 2H) , 2.67 -2.53 (m, 3H) , 2.21 -2.09 (m, 3H) , 2.06 -1.96 (m, 3H) , 1.95 -1.89 (m, 2H) , 1.78 (m, J = 6.0 Hz, 2H) , 1.74 -1.65 (m, 3H) , 1.560 (m, 7H) , 0.92 (d, J =6.4 Hz, 1H)
Example 94:
4- {1- [1- ( {3- [4-chloro-3- (2, 4-dioxo-1, 3, 5, 6-tetrahydro-1-pyrimidinyl) benzoyl] -3-aza-9-spiro [5.5] undecyl} methyl) -4-piperidyl] -4-pyrazolyl} -13-cyclopropyl-11- (2, 6-dichlorophenyl) -5, 7, 11, 13-tetraazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (Cpd-213)
Scheme:
Step A: 2-bromo-4- (cyclopropylamino) -N- (2, 6-dichlorophenyl) -1- (2-
trimethylsilylethoxymethyl) pyrrolo [2, 3-b] pyridine-5-carboxamide
Cyclopropanamine (249 mg, 4.37 mmol) and 2-bromo-4-chloro-N- (2, 6-dichlorophenyl) -1- (2-trimethylsilylethoxymethyl) pyrrolo [2, 3-b] pyridine-5-carboxamide (300 mg, 545.7 μmol) were taken up into a microwave tube in NMP (5.0 mL) . The sealed tube was heated at 100 ℃ for 2 hr under microwave. The reaction mixture was poured into water (5.0 mL) , white solid was precipitated. The mixture was stirred at 0 ℃ for 15 min, filtered and rinsed with water (2.0 mL×3) . The white solid was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 571.1 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 9.24 (s, 1H) , 8.67 (s, 1H) , 7.45 -7.43 (m, 1H) , 7.43 -7.41 (m, 1H) , 7.25 (d, J=1.2 Hz, 1H) , 7.22 (s, 1H) , 7.19 (t, J=3.2 Hz, 1H) , 5.77 (s, 2H) , 3.72 -3.64 (m, 2H) , 2.99 (dt, J=3.2, 6.8 Hz, 1H) , 1.02 -0.90 (m, 4H) , 0.88 -0.77 (m, 2H) , 0.00 -0.04 (m, 9H) .
Step B: 4-bromo-13-cyclopropyl-11- (2, 6-dichlorophenyl) -5- (2-trimethylsilylethoxymethyl) -
5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
2-bromo-4- (cyclopropylamino) -N- (2, 6-dichlorophenyl) -1- (2-trimethylsilylethoxymethyl) pyrrolo [2, 3-b] pyridine-5-carboxamide (310 mg, 543.5 μmol) , DIPEA (1.05 g, 8.15 mmol, 1.42 mL) and CDI (441 mg, 2.72 mmol) were taken up into a microwave tube in DMAC (1.4 mL) . The sealed tube was heated at 145 ℃ for 2hr under microwave. The reaction mixture was diluted with water (8.0 mL) and extracted with ethyl acetate (8.0 mL×3) . The combined organic layer was washed with brine (10.0 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=5/1) to afford the desired product.
LCMS: MS (ESI) m/z 597.1 [M+H] +.
Step C: tert-butyl 4- [4- [13-cyclopropyl-11- (2, 6-dichlorophenyl) -10, 12-dioxo-5- (2-
trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-
tetraen-4-yl] pyrazol-1-yl] piperidine-1-carboxylate
To a solution of 4-bromo-13-cyclopropyl-11- (2, 6-dichlorophenyl) -5- (2-trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10,12-dione (98 mg, 164.3 μmol) and tert-butyl 4- [4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazol-1-yl] piperidine-1-carboxylate (74.40 mg, 197.1 μmol) in dioxane (4.0 mL) and water (0.8 mL) was added Pd (dppf) Cl2 (12 mg, 16.4 μmol) and K2CO3 (45 mg, 328.6 μmol) . The mixture was stirred at 90 ℃ for 1hr under nitrogen atmosphere. The residue was diluted with water (5.0 mL) and extracted with ethyl acetate (5 mL ×3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure
to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1/1) to afford the desired product.
LCMS: MS (ESI) m/z 766.4 [M+H] +.
Step D: 13-cyclopropyl-11- (2, 6-dichlorophenyl) -4- [1- (4-piperidyl) pyrazol-4-yl] -5, 7, 11, 13-
tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
To a solution of tert-butyl 4- [4- [13-cyclopropyl-11- (2, 6-dichlorophenyl) -10, 12-dioxo-5- (2-trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] piperidine-1-carboxylate (100 mg, 130.4 μmol) in DCM (3.0 mL) was added TFA (3.0 mL) . The mixture was stirred at 40 ℃ for 20 hr. The reaction mixture was concentrated in vacuum. The residue was co-evaporated with toluene (1.0 mL×3) to afford the desired product.
LCMS: MS (ESI) m/z 536.3 [M+H] +.
Step E: tert-butyl 9- [ [4- [4- [13-cyclopropyl-11- (2, 6-dichlorophenyl) -10, 12-dioxo-5, 7, 11, 13-
tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] -1-
piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of 13-cyclopropyl-11- (2, 6-dichlorophenyl) -4- [1- (4-piperidyl) pyrazol-4-yl] -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (70 mg, 130.5 μmol) and tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (44 mg, 156.6 μmol) in DCM (5.0 mL) was added TEA (66 mg, 652.4 μmol) . The reaction mixture was stirred at 20 ℃for 1 hr. Then NaBH (OAc) 3 (83 mg, 391.4 μmol) was added to the reaction mixture, the mixture was stirred at 20 ℃ for 1 hr. The reaction mixture was quenched by addition sat. NaHCO3 (2.0 mL) and extracted with DCM (3.0 mL×3) . The combined organic layers were washed with bine, dried over Na2SO4, filtered and concentrated in vacuum. The residue was purified by prep-TLC (SiO2, DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 801.5 [M+H] +.
Step F: 4- [1- [1- (3-azaspiro [5.5] undecan-9-ylmethyl) -4-piperidyl] pyrazol-4-yl] -13-
cyclopropyl-11- (2, 6-dichlorophenyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-
1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
To a solution of tert-butyl 9- [ [4- [4- [13-cyclopropyl-11- (2, 6-dichlorophenyl) -10, 12-dioxo-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] -1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate (68 mg, 84.8 μmol) in DCM (1.5 mL) was added HCl/dioxane (4M) (1.5 mL) . The mixture was stirred at 20 ℃ for 1hr. The reaction mixture was concentrated in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 701.4 [M+H] +.
Step G: 4- {1- [1- ( {3- [4-chloro-3- (2, 4-dioxo-1, 3, 5, 6-tetrahydro-1-pyrimidinyl) benzoyl] -3-aza-
9-spiro [5.5] undecyl} methyl) -4-piperidyl] -4-pyrazolyl} -13-cyclopropyl-11- (2, 6-
dichlorophenyl) -5, 7, 11, 13-tetraazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-
dione
To a solution of 4- [1- [1- (3-azaspiro [5.5] undecan-9-ylmethyl) -4-piperidyl] pyrazol-4-yl] -13-cyclopropyl-11- (2, 6-dichlorophenyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (50 mg, 71.2 μmol) and 4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoic acid (20 mg, 74.4 μmol, ) in DMF (1.5 mL) were added DIPEA (46 mg, 356.2 μmol) and HATU (41 mg, 106.8 μmol) . The mixture was stirred at 20 ℃ for 1hr. The reaction was quenched with water (0.5 mL) . The reaction mixture was purified by prep-HPLC (column: Welch Xtimate C18 100*40mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 18%-48%B over 8 min) to afford the desired product.
LCMS: MS (ESI) m/z 951.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 12.74 (s, 1H) , 10.53 (s, 1H) , 9.23 (s, 1H) , 8.73 (s, 1H) , 8.38 (s, 1H) , 8.18 -8.15 (m, 1H) , 7.73 -7.68 (m, 2H) , 7.65 (d, J=8.0 Hz, 1H) , 7.61 -7.57 (m, 1H) , 7.55 (s, 1H) , 7.41 -7.36 (m, 2H) , 4.60 -4.48 (m, 1H) , 3.54 -3.43 (m, 2H) , 3.22 -2.93 (m, 3H) , 3.22 -2.93 (m, 3H) , 2.79 -2.70 (m, 2H) , 2.37 -2.22 (m, 4H) , 1.87 -1.67 (m, 4H) , 1.66 -1.42 (m, 5H) , 1.42 -1.26 (m, 5H) , 1.24 -1.07 (m, 5H) , 0.86 (m, 2H) .
Example 95:
4- {1- [1- ( {3- [4-chloro-3- (2, 4-dioxo-1, 3, 5, 6-tetrahydro-1-pyrimidinyl) benzoyl] -3-aza-9-spiro [5.5] undecyl} methyl) -4-piperidyl] -4-pyrazolyl} -11- (2, 6-dichlorophenyl) -13-ethyl-5, 7, 11, 13-tetraazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (Cpd-212)
Scheme:
Step A: 2-bromo-N- (2, 6-dichlorophenyl) -4- (ethylamino) -1- (2-
trimethylsilylethoxymethyl) pyrrolo [2, 3-b] pyridine-5-carboxamide
To a solution of 2-bromo-4-chloro-N- (2, 6-dichlorophenyl) -1- (2-trimethylsilylethoxymethyl) pyrrolo [2, 3-b] pyridine-5-carboxamide (250 mg, 454.75 μmol) and ethanamine; hydrochloride (HCl, 593.32 mg, 7.28 mmol) in NMP (1 mL) was added DIEA (1.06 g, 8.19 mmol) under microwave. The mixture was stirred at 120 ℃ for 2hr. The crude residue was dissolved in DCM (300 mL) , washed with water (50 mL) and brine (450 mL) , the organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure to obtain a residue. The residue was purified by flash silica gel chromatography (20 g
Silica Flash Column, Eluent of 0~30%PE/EtOAc gradient @30 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 558.9 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.59 (s, 1H) , 7.40 (d, J = 8.13 Hz, 2H) , 7.23 -7.15 (m, 1H) , 6.80 (s, 1H) , 5.68 (m, 2H) , 3.72 -3.64 (m, 2H) , 3.61 (m, 2H) , 1.34 (t, J = 7.2 Hz, 3H) , 0.93 -0.88 (m, 2H) , 0.00 (s, 9H)
Step B: 4-bromo-11- (2, 6-dichlorophenyl) -13-ethyl-5- (2-trimethylsilylethoxymethyl) -
5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
To a solution of 2-bromo-N- (2, 6-dichlorophenyl) -4- (ethylamino) -1- (2-trimethylsilylethoxymethyl) pyrrolo [2, 3-b] pyridine-5-carboxamide (150 mg, 268.64 μmol) and CDI (653.39 mg, 4.03 mmol) in DMAC (3 mL) was added DIPEA (2.23 g, 17.22 mmol) . The mixture was stirred at 150 ℃ for 2hr under microwave. The crude residue was dissolved in DCM (300 mL) , washed with water (50 mL) and brine (450 mL) , the organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure to obtain a residue. The residue was purified by flash silica gel chromatography (20 gSilica Flash Column, Eluent of 0~27%PE/EtOAc gradient @30 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 584.9 [M+H] +.
Step C: tert-butyl 4- [4- [11- (2, 6-dichlorophenyl) -13-ethyl-10, 12-dioxo-5- (2-
trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-
tetraen-4-yl] pyrazol-1-yl] piperidine-1-carboxylate
To a solution of 4-bromo-11- (2, 6-dichlorophenyl) -13-ethyl-5- (2-trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (59 mg, 100.96 μmol) , tert-butyl 4- [4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazol-1-yl] piperidine-1-carboxylate (60.95 mg, 161.54 μmol) in dioxane (0.5 mL) and H2O (0.1 mL) was added K2CO3 (27.91 mg, 201.93 μmol) , Pd (dppf) Cl2 (7.39 mg, 10.10 μmol) under N2. The mixture was stirred at 90 ℃ for 1hr. The crude residue was dissolved in DCM (90 mL) , washed with water (30 mL) and brine (120 mL) , the organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure to obtain a residue. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~35%PE/EtOAc gradient @30 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 417.1 [M+H] +.
Step D: 11- (2, 6-dichlorophenyl) -13-ethyl-4- [1- (4-piperidyl) pyrazol-4-yl] -5, 7, 11, 13-
tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
To a solution of tert-butyl 4- [4- [11- (2, 6-dichlorophenyl) -13-ethyl-10, 12-dioxo-5- (2-trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-
yl] pyrazol-1-yl] piperidine-1-carboxylate (90 mg, 119.2 μmol) in DCM (3.0 mL) was added TFA (3.0 mL) . The mixture was stirred at 40 ℃ for 4hr. The reaction was concentrated in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 524.3 [M+H] +.
Step E: tert-butyl 9- [ [4- [4- [11- (2, 6-dichlorophenyl) -13-ethyl-10, 12-dioxo-5, 7, 11, 13-
tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] -1-
piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of 11- (2, 6-dichlorophenyl) -13-ethyl-4- [1- (4-piperidyl) pyrazol-4-yl] -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (63 mg, 120.1 μmol) and tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (41 mg, 145.7 μmol) in DCM (5.0 mL) was added TEA (61 mg, 600.6 μmol) . The reaction mixture was stirred at 20 ℃ for 1 hr. Then NaBH (OAc) 3 (76 mg, 360.4 μmol) was added to the reaction mixture, the mixture was stirred at 20 ℃ for 1 hr. The reaction mixture was quenched by addition sat. NaHCO3 (2.0 mL) and extracted with DCM (3.0 mL×3) . The combined organic layers were washed with bine, dried over Na2SO4, filtered and concentrated in vacuum. The residue was purified by prep-TLC (SiO2, DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 789.5 [M+H] +.
Step F: 4- [1- [1- (3-azaspiro [5.5] undecan-9-ylmethyl) -4-piperidyl] pyrazol-4-yl] -11- (2, 6-
dichlorophenyl) -13-ethyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-
10, 12-dione
To a solution of tert-butyl 9- [ [4- [4- [11- (2, 6-dichlorophenyl) -13-ethyl-10, 12-dioxo-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] -1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate (90 mg, 113.95 μmol) in HCl/dioxane (4M, 3 mL) and DCM (3 mL) . The mixture was stirred at 25 ℃ for 2hr. The reaction mixture was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 689.1 [M+H] +.
Step G: 4- {1- [1- ( {3- [4-chloro-3- (2, 4-dioxo-1, 3, 5, 6-tetrahydro-1-pyrimidinyl) benzoyl] -3-aza-
9-spiro [5.5] undecyl} methyl) -4-piperidyl] -4-pyrazolyl} -11- (2, 6-dichlorophenyl) -13-ethyl-
5, 7, 11, 13-tetraazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
To a solution of 4- [1- [1- (3-azaspiro [5.5] undecan-9-ylmethyl) -4-piperidyl] pyrazol-4-yl] -11- (2, 6-dichlorophenyl) -13-ethyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10,12-dione (65 mg, 94.25 μmol) and 4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoic acid (27.85 mg, 103.67 μmol) in DMF (1 mL) were added DIPEA (60.90 mg, 471.24 μmol) and HATU (53.75 mg, 141.37 μmol) . The mixture was stirred at 25 ℃ for 2hr. The mixture was concentrated under reduced pressure to give a residue. The crude product was purified by
reversed-phase HPLC (column: Welch Xtimate C18 100*40mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 19%-49%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 941.2 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 12.86 (s, 1H) , 10.53 (s, 1H) , 9.23 (br s, 1H) , 8.81 (s, 1H) , 8.42 (s, 1H) , 8.20 (s, 1H) , 7.73 (d, J = 8.1 Hz, 2H) , 7.65 (d, J = 8.1 Hz, 1H) , 7.62 -7.58 (m, 1H) , 7.56 (br d, J = 5.5 Hz, 1H) , 7.40 (br d, J = 8.1 Hz, 1H) , 7.19 (s, 1H) , 4.54 (br d, J = 6.8 Hz, 2H) , 3.51 -3.50 (m, 1H) , 3.39 -2.98 (m, 8H) , 2.79 -2.72 (m, 2H) , 2.32 (m, 4H) , 1.85 -1.59 (m, 6H) , 1.58 -1.27 (m, 9H) , 1.17 (br s , 5H)
Example 96:
4- {1- [ (3R, 4R) -1- ( {3- [4-chloro-3- (2, 4-dioxo-1, 3, 5, 6-tetrahydro-1-pyrimidinyl) benzoyl] -3-aza-9-spiro [5.5] undecyl} methyl) -3-fluoro-4-piperidyl] -4-pyrazolyl} -11- (2, 6-dichlorophenyl) -13-methyl-5, 7, 11, 13-tetraazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (Cpd-298)
Scheme:
Step A: tert-butyl (3R, 4R) -4- [4- [11- (2, 6-dichlorophenyl) -13-methyl-10, 12-dioxo-5- (2-
trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-
tetraen-4-yl] pyrazol-1-yl] -3-fluoro-piperidine-1-carboxylate
To a solution of tert-butyl (3R, 4R) -3-fluoro-4- [4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazol-1-yl] piperidine-1-carboxylate (100.00 mg, 252.99 μmol) , 4-bromo-11- (2, 6-dichlorophenyl) -13-methyl-5- (2-trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (144.29 mg, 252.99 μmol) in dioxane (2.5 mL) and H2O (0.5 mL) was added Pd (dppf) Cl2 (18.51 mg, 25.30 μmol) , K2CO3 (69.93 mg, 505.98 μmol) under N2. The mixture was stirred at 90 ℃ for 1 h. The mixture was allowed to cool to room temperature, diluted with H2O (30 mL) , and extracted with EtOAc (3 ×30 mL) . The combined organic extracted with brine (3 × 50 mL) , dried with Na2SO4. After
filtration, the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=100/1 to 5/1) to afford the desired product.
LCMS: MS (ESI) m/z 758.5 [M+H] +.
Step B: 11- (2, 6-dichlorophenyl) -4- [1- [ (3R, 4R) -3-fluoro-4-piperidyl] pyrazol-4-yl] -5-
(hydroxymethyl) -13-methyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-
tetraene-10, 12-dione
To a solution of tert-butyl (3R, 4R) -4- [4- [11- (2, 6-dichlorophenyl) -13-methyl-10, 12-dioxo-5- (2-trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] -3-fluoro-piperidine-1-carboxylate (89 mg, 117.30 μmol) in DCM (3 mL) was added HCl/dioxane (4 M, 3 mL) and TFA (767.50 mg, 6.73 mmol, 0.5 mL) . The mixture was stirred at 20 ℃ for 1 h. The reaction mixture was concentrated under reduced pressure to give residue. Toluene (3 mL) was added to the residue. The mixture was concentrated in vacuo to give a residue. Repeat the preceding operation three times to afford the desired product.
LCMS: MS (ESI) m/z 558.2 [M+H] +.
Step C: tert-butyl 9- [ [ (3R, 4R) -4- [4- [11- (2, 6-dichlorophenyl) -13-methyl-10, 12-dioxo-
5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] -3-fluoro-
1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of 11- (2, 6-dichlorophenyl) -4- [1- [ (3R, 4R) -3-fluoro-4-piperidyl] pyrazol-4-yl] -5- (hydroxymethyl) -13-methyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (69 mg, 116.00 μmol, HCl) , tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (48.96 mg, 139.19 μmol) in DCM (3 mL) was added TEA (35.21 mg, 349.99 μmol, 48.44 μL) and NaBH (OAc) 3 (73.75 mg, 347.99 μmol) . The mixture was stirred at 20 ℃ for 1 h. The mixture was allowed to cool to room temperature, diluted with H2O (30 mL) , and extracted with EtOAc (3 × 30 mL) . The combined organic extracted with brine (3 × 50 mL) , dried with Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol=100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 793.3 [M+H] +.
Step D: 4- [1- [ (3R, 4R) -1- (3-azaspiro [5.5] undecan-9-ylmethyl) -3-fluoro-4-piperidyl] pyrazol-
4-yl] -11- (2, 6-dichlorophenyl) -13-methyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-
1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
To a solution of tert-butyl 9- [ [ (3R, 4R) -4- [4- [11- (2, 6-dichlorophenyl) -13-methyl-10, 12-dioxo-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] -3-fluoro-1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate (30 mg, 37.80 μmol) in HCl/dioxane
(4 M, 3 mL) was stirred at 20 ℃ for 16 h. The mixture was allowed to cool to room temperature, diluted with H2O (30 mL) , and extracted with EtOAc (3 × 30 mL) . The combined organic extracted with brine (3 × 50 mL) , dried with Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 693.5 [M+H] +.
Step E: 4- {1- [ (3R, 4R) -1- ( {3- [4-chloro-3- (2, 4-dioxo-1, 3, 5, 6-tetrahydro-1-
pyrimidinyl) benzoyl] -3-aza-9-spiro [5.5] undecyl} methyl) -3-fluoro-4-piperidyl] -4-
pyrazolyl} -11- (2, 6-dichlorophenyl) -13-methyl-5, 7, 11, 13-tetraazatricyclo [7.4.0.02, 6] trideca-
1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
To a solution of 4- [1- [ (3R, 4R) -1- (3-azaspiro [5.5] undecan-9-ylmethyl) -3-fluoro-4-piperidyl] pyrazol-4-yl] -11- (2, 6-dichlorophenyl) -13-methyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (15 mg, 21.63 μmol) and 4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoic acid (6.97 mg, 25.95 μmol) in DMF (1 mL) was added HATU (12.33 mg, 32.44 μmol) and DIPEA (8.38 mg, 64.88 μmol) . The mixture was stirred at 20 ℃ for 1 h. The crude product was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 6%-46%B over 9 min) to afford the desired product, then dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 944.9 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ12.81 (s, 1H) , 10.52 (s, 1H) , 8.78 (s, 1H) , 8.43 (s, 1H) , 8.16 (s, 1H) , 8.13 (FA, 0.17H) , 7.72 (s, 1H) , 7.70 (s, 1H) , 7.63 (d, J = 8.0 Hz, 1H) , 7.60 -7.57 (m, 1H) , 7.57 -7.54 (m, 1H) , 7.39 (dd, J = 1.6, 8.4 Hz, 1H) , 7.33 (d, J = 2.0 Hz, 1H) , 5.02 -4.70 (m, 1H) , 4.40 (s, 1H) , 4.00 (s, 3H) , 3.84 -3.69 (m, 1H) , 3.67 -3.49 (m, 3H) , 3.32 -3.22 (m, 4H) , 2.87 (s, 1H) , 2.79 -2.65 (m, 2H) , 2.26 (s, 2H) , 2.15 -2.00 (m, 3H) , 1.65-1.75 (m, 2H) , 1.63 -1.40 (m, 5H) , 1.39 -1.24 (m, 2H) , 1.16-1.10 (m, 4H) .
19F NMR (400 MHz, DMSO-d6) δ = -185.69
Example 97:
4- {1- [ (3S, 4S) -1- ( {3- [4-chloro-3- (2, 4-dioxo-1, 3, 5, 6-tetrahydro-1-pyrimidinyl) benzoyl] -3-aza-9-spiro [5.5] undecyl} methyl) -3-fluoro-4-piperidyl] -4-pyrazolyl} -11- (2, 6-dichlorophenyl) -13-methyl-5, 7, 11, 13-tetraazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (Cpd-300)
Scheme:
Step A: tert-butyl (3S, 4R) -3-fluoro-4- [4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-
yl) pyrazol-1-yl] piperidine-1-carboxylate
A mixture of tert-butyl (3S, 4S) -3-fluoro-4-hydroxy-piperidine-1-carboxylate (200 mg, 912.19 μmol) , 4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-pyrazole (212.40 mg, 1.09 mmol) , 2-
(tributyl-phosphanylidene) acetonitrile (484.35 mg, 2.01 mmol) in toluene (3 mL) was stirred at 100 ℃ for 16 hr under N2 atmosphere. The mixture was diluted with water (100 mL) and extracted with ethyl acetate (100 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by column chromatography on silica gel (16%EtOAc in PE) to afford the desired product.
LCMS: MS (ESI) m/z 396.4 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 7.84 (s, 1H) , 7.81 (s, 1H) , 4.15 -4.39 (m, 1H) , 4.19 -4.28 (m, 1H) , 3.75 -3.87 (m, 1H) , 2.90 -2.98 (m, 2H) , 2.28 -2.42 (m, 1H) , 1.95 -2.02 (m, 2H) , 1.34 (s, 9H) , 1.32 (m, 12H) .
Step B: tert-butyl (3S, 4R) -4- [4- [11- (2, 6-dichlorophenyl) -13-methyl-10, 12-dioxo-5- (2-
trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-
tetraen-4-yl] pyrazol-1-yl] -3-fluoro-piperidine-1-carboxylate
A mixture of 4-bromo-11- (2, 6-dichlorophenyl) -13-methyl-5- (2-trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (100 mg, 175.33 μmol) , tert-butyl (3S, 4R) -3-fluoro-4- [4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazol-1-yl] piperidine-1-carboxylate (138.61 mg, 350.67 μmol) , Pd (dppf) Cl2. CH2Cl2 (14.32 mg, 17.53 μmol) , K2CO3 (72.70 mg, 526.00 μmol) in dioxane (5 mL) and H2O (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90 ℃ for 1 hr under N2 atmosphere. The mixture was diluted with water (100 mL) and extracted with ethyl acetate (200 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by silica gel column chromatography (5%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 758.4 [M+H] +.
Step C: 11- (2, 6-dichlorophenyl) -4- [1- [ (3S, 4R) -3-fluoro-4-piperidyl] pyrazol-4-yl] -13-
methyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
To a solution of tert-butyl (3S, 4R) -4- [4- [11- (2, 6-dichlorophenyl) -13-methyl-10, 12-dioxo-5- (2-trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] -3-fluoro-piperidine-1-carboxylate (100 mg, 131.80 μmol) in DCM (2 mL) was added TFA (3.07 g, 26.92 mmol, 2 mL) . The mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 528.1 [M+H] +.
Step D: tert-butyl 9- [ [ (3S, 4R) -4- [4- [11- (2, 6-dichlorophenyl) -13-methyl-10, 12-dioxo-
5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] -3-fluoro-
1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of 11- (2, 6-dichlorophenyl) -4- [1- [ (3S, 4R) -3-fluoro-4-piperidyl] pyrazol-4-yl] -13-methyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (80 mg, 151.41 μmol) , tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (42.61 mg, 151.41 μmol) , TEA (45.96 mg, 454.23 μmol, 63.22 μL) in DCM (3 mL) was added NaBH (OAc) 3 (96.27 mg, 454.23 μmol) . The mixture was stirred at 20 ℃ for 16 hr. The mixture was diluted with water (30 mL) and extracted with ethyl acetate (50 mL × 2) . The combined organic layers were washed with brine (30 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by silica gel column chromatography (5%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 793.4 [M+H] +.
Step E: 4- [1- [ (3S, 4R) -1- (3-azaspiro [5.5] undecan-9-ylmethyl) -3-fluoro-4-piperidyl] pyrazol-
4-yl] -11- (2, 6-dichlorophenyl) -13-methyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-
1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
To a solution of tert-butyl 9- [ [ (3S, 4R) -4- [4- [11- (2, 6-dichlorophenyl) -13-methyl-10, 12-dioxo-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] -3-fluoro-1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate (100 mg, 125.98 μmol) in DCM (1 mL) was added TFA (1.54 g, 13.46 mmol, 1 mL) . The mixture was stirred at 20 ℃ for 2hr. The mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 693.3 [M+H] +.
Step F: 4- {1- [ (3S, 4S) -1- ( {3- [4-chloro-3- (2, 4-dioxo-1, 3, 5, 6-tetrahydro-1-
pyrimidinyl) benzoyl] -3-aza-9-spiro [5.5] undecyl} methyl) -3-fluoro-4-piperidyl] -4-
pyrazolyl} -11- (2, 6-dichlorophenyl) -13-methyl-5, 7, 11, 13-tetraazatricyclo [7.4.0.02, 6] trideca-
1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
A mixture of 4- [1- [ (3S, 4R) -1- (3-azaspiro [5.5] undecan-9-ylmethyl) -3-fluoro-4-piperidyl] pyrazol-4-yl] -11- (2, 6-dichlorophenyl) -13-methyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (100 mg, 144.17 μmol) , 4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoic acid (38.73 mg, 144.17 μmol) , HATU (82.22 mg, 216.25 μmol) , DIEA (93.16 mg, 720.84 μmol, 125.56 μL) in DMF (2 mL) was stirred at 20 ℃ for 16 hr. The mixture was diluted with water (30 mL) and extracted with ethyl acetate (50 mL × 2) . The combined organic layers were washed with brine (30 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by prep-HPLC (column: Welch Xtimate C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 15%-55%B over 9 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 945.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ12.80 (s, 1H) , 10.50 (s, 1H) , 8.78 (s, 1H) , 8.45 (s, 1H) , 8.17 (s, 1H) , 7.75 -7.67 (m, 2H) , 7.66 -7.51 (m, 3H) , 7.44 -7.33 (m, 2H) , 5.22 -4.33 (m, 1H) , 4.01 (s, 3H) , 3.82 -3.69 (m, 1H) , 3.68 -3.45 (m, 4H) , 3.18 -2.87 (m, 3H) , 2.83 -2.60 (m, 3H) , 2.46 -1.88 (m, 5H) , 1.71 (s, 3H) , 1.64 -1.25 (m, 8H) , 1.13 (s, 3H) .
19F NMR (400 MHz, DMSO-d6) δ –71.078
Example 98:
3- [5- (4- {2- [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] ethyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-172)
Scheme:
Step A: 2- (2, 6-dioxo-3-piperidyl) -5- [4- (2-hydroxyethyl) -1-piperidyl] isoindoline-1, 3-dione
To a solution of 2- (4-piperidyl) ethanol (1.55 g, 11.96 mmol) , 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (3 g, 10.86 mmol) in DMF (60 mL) was added DIPEA (2.81 g, 21.72 mmol) . The mixture was stirred at 80 ℃ for 16 h. The reaction mixture was added H2O (200 mL) and extracted with ethyl acetate (100 mL × 3) . The combined organic phases were washed with brine (100 mL × 3) , dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was triturated with EtOAc (30 mL) and PE (10 mL) at 20 ℃ for 16 h to afford the desired product.
LCMS: MS (ESI) m/z 386.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.08 (s, 1H) , 7.64 (d, J = 8.4 Hz, 1H) , 7.29 (d, J = 1.6 Hz, 1H) , 7.22 (dd, J = 2.0, 8.8 Hz, 1H) , 5.06 (dd, J = 5.2, 13.2 Hz, 1H) , 4.39 (t, J = 5.2 Hz, 1H) , 4.05-4.00 (m, 2H) , 3.51 -3.39 (m, 2H) , 3.10 (s, 1H) , 3.01 -2.90 (m, 2H) , 2.89 -2.82 (m, 1H) , 2.62-2.52 (m, 1H) , 2.06 -1.95 (m, 1H) , 1.73 (d, J = 12.8 Hz, 2H) , 1.72-1.63 (m, 1H) , 1.37 (q, J = 6.2 Hz, 2H) , 1.27 -1.06 (m, 2H) .
Step B: 2- [1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] -4-piperidyl] acetaldehyde
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [4- (2-hydroxyethyl) -1-piperidyl] isoindoline-1, 3-dione (1 g, 2.59 mmol) in DCM (10 mL) and DMF (1 mL) was added Dess-Martin (1.32 g, 3.11 mmol) . The mixture was stirred at 20 ℃ for 1 h. The reaction mixture was added H2O (200 mL) and extracted with ethyl acetate (100 mL × 3) . The combined organic phases were washed with brine (100 mL × 3) , dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 384.0 [M+H] +.
Step C: 2- (2, 6-dioxo-3-piperidyl) -5- [4- [2- [3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-9-
yl] ethyl] -1-piperidyl] isoindoline-1, 3-dione
To a solution of 2- [1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] -4-piperidyl] acetaldehyde (1.4 g, 2.04 mmol) , 3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane (450.44 mg, 1.64 mmol) in DCM (30 mL) was added TEA (620.76 mg, 6.13 mmol) , NaBH (OAc) 3 (1.30 g, 6.13 mmol) . The mixture was stirred at 20 ℃ for 1 h. The reaction mixture was added H2O (200 mL) and extracted with ethyl acetate (100 mL × 3) . The combined organic phases were washed with brine (100 mL × 3) , dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol = 100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 643.6 [M+H] +.
Step D: 3- [5- [4- [2- [3- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] ethyl] -1-piperidyl] -
1-oxo-isoindolin-2-yl] piperidine-2, 6-dione &3- [6- [4- [2- [3- (4-aminophenyl) -3, 9-
diazaspiro [5.5] undecan-9-yl] ethyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [4- [2- [3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] ethyl] -1-piperidyl] isoindoline-1, 3-dione (1.5 g, 2.33 mmol) in TFA (20 mL) was added Zn (3 g, 45.88 mmol) . The mixture was stirred at 75 ℃ for 1 h. The reaction mixture was filtered. The residue washed with EtOAc (100 mL) , concentrated under reduced pressure to give crude product. The crude product was purified by reversed-phase column ( [water (TFA) -ACN] ; B%: 6%-20%) to afford the desired product.
3- [5- [4- [2- [3- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] ethyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione:
LCMS: MS (ESI) m/z 599.4 [M+H] +.
2D NMR: showed the substituent site in the determined structure is confirmed by ROESY correlation between H10 and H14.
1H NMR (400 MHz, DMSO-d6) δ 10.97 (s, 1H) , 9.89 (s, 1H) , 7.46 (d, J = 8.4 Hz, 1H) , 7.34 (d, J = 8.4 Hz, 1H) , 7.25 (d, J = 13.2 Hz, 3H) , 6.95 (s, 2H) , 5.09 (dd, J = 5.2, 13.2 Hz, 1H) , 4.38 -4.18 (m, 2H) , 3.75 (d, J = 11.2 Hz, 2H) , 3.64-3.56 (m, 1H) , 3.36 (d, J = 11.2 Hz, 2H) , 3.29 (m,
4H) , 3.16 -3.09 (m, 3H) , 3.07-2.98 (m, 2H) , 2.96 -2.86 (m, 1H) , 2.85-2.75 (m, 2H) , 2.65-2.55 (m, 1H) , 2.44 -2.32 (m, 1H) , 2.02 -1.96 (m, 1H) , 1.92-1.83 (m, 2H) , 1.79 (d, J = 11.6 Hz, 2H) , 1.75-1.59 (m, 6H) , 1.53 (m, 1H) , 1.42-1.32 (m, 2H) .
3- [6- [4- [2- [3- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] ethyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione:
LCMS: MS (ESI) m/z 599.4 [M+H] +.
2D NMR: showed the substituent site in the determined structure is confirmed by ROESY correlation between H16 and H11.
1H NMR (400 MHz, DMSO-d6) δ 10.94 (s, 1H) , 10.17 (s, 1H) , 7.51 (d, J = 8.4 Hz, 1H) , 7.26 (s, 2H) , 7.11 -7.04 (m, 2H) , 6.96 (s, 2H) , 5.04 (dd, J = 5.2, 13.2 Hz, 1H) , 4.36 -4.16 (m, 2H) , 3.87 (d, J = 12.4 Hz, 2H) , 3.72 (s, 1H) , 3.42 -3.19 (m, 6H) , 3.15-3.08 (m, 2H) , 3.04-2.97 (m, 2H) , 2.95 -2.87 (m, 1H) , 2.87 -2.79 (m, 2H) , 2.61-2.55 (m, 1H) , 2.42 -2.31 (m, 1H) , 2.00 -1.92 (m, 2H) , 1.91-1.85 (m, 2H) , 1.79-1.70 (m, 3H) , 1.69-1.62 (m, 4H) , 1.61-1.50 (m, 2H) , 1.34 -1.19 (m, 2H) .
Step E: 3- [5- (4- {2- [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] ethyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -
2, 6-piperidinedione
To a solution of 3- [6- [4- [2- [3- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] ethyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (30 mg, 50.10 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (22.12 mg) in t-BuOH (1 mL) was stirred at 100 ℃ for 2 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 0%-35%B over 9 min) to afford the desired product.
LCMS: MS (ESI) m/z 920.8 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ10.96 (s, 1H) , 10.06 (brs, 1H) , 8.82 (s, 1H) , 8.10 (t, J = 2.0 Hz, 1H) , 7.75 (d, J = 8.0Hz, 1H) , 7.45-7.60 (m, 4H) , 7.00-7.10 (m, 2H) , 6.89 (d, J = 8.8Hz, 2H) , 5.25-5.42 (m, 2H) , 5.04 (dd, J = 4.0, 12.0 Hz, 1H) , 4.40-4.51 (m, 2H) , 4.10-4.35 (m, 2H) , 3.82-3.90 (m, 2H) , 2.85-2.94 (m, 1H) , 2.75-2.84 (m, 2H) , 2.55-2.63 (m, 1H) , 2.28-2.46 (m, 8H) , 2.12-2.26 (m, 1H) , 1.90-2.05 (m, 3H) , 1.65-1.77 (m, 3H) , 1.35-1.65 (m, 15H) , 1.13-1.27 (m, 3H) .
Example 99:
3- [6- (4- {2- [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] ethyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-173)
Scheme:
Step A: 3- [6- (4- {2- [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] ethyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -
2, 6-piperidinedione
To a solution of 3- [5- [4- [2- [3- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] ethyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (40 mg, 66.80 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (26.82 mg, 66.80 μmol) in n-BuOH (1 mL) was added AcOH (40.12 mg, 668.03 μmol) . The mixture was stirred at 80 ℃ for 16 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: Xtimate C18 150*40mm*10um; mobile phase: [water (FA) -ACN] ; gradient: 15%-45%B over 6 min) to afford the desired product, then dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 920.8 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.97 (s, 1H) , 10.04 (s, 1H) , 8.81 (s, 1H) , 8.22 (s, 1H) , 8.05 -7.95 (m, 1H) , 7.75 (d, J = 8.4 Hz, 1H) , 7.60 -7.51 (m, 2H) , 7.40 (d, J = 8.4 Hz, 1H) , 7.24 (br d, J = 8.8 Hz, 1H) , 7.14 (s, 1H) , 6.90 (br d, J = 9.2 Hz, 2H) , 5.40 -5.22 (m, 2H) , 5.09 (dd, J = 5.2, 12.8 Hz, 1H) , 4.44 (br s, 1H) , 4.33 (d, J = 16.8 Hz, 1H) , 4.19 (d, J = 16.8 Hz, 1H) , 3.73 (br d, J = 11.6 Hz, 2H) , 3.07 (m, 5H) , 2.96 -2.85 (m, 1H) , 2.75 -2.64 (m, 3H) , 2.64 -2.53 (m, 2H) , 2.48 -2.33 (m, 7H) , 2.08 -1.87 (m, 5H) , 1.80 -1.62 (m, 4H) , 1.60 -1.34 (m, 11H) , 1.32 -1.20 (m, 2H) .
Example 100:
3- [5- (4- {2- [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] ethyl} -1-piperidyl) -1, 3-dioxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-174)
Scheme:
Step A: 2- [1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] -4-piperidyl] acetaldehyde
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [4- (2-hydroxyethyl) -1-piperidyl] isoindoline-1, 3-dione (1 g, 2.59 mmol) in DCM (10 mL) and DMF (1 mL) was added Dess-Martin (1.32 g, 3.11 mmol) . The mixture was stirred at 20 ℃ for 1 h. The reaction mixture was added H2O (20 mL) and extracted with ethyl acetate (10 mL × 3) . The combined organic phases were washed with brine (100 mL × 3) , dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 384.3 [M+H] +.
Step B: 2- (2, 6-dioxo-3-piperidyl) -5- [4- [2- [3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-9-
yl] ethyl] -1-piperidyl] isoindoline-1, 3-dione
To a solution of 2- [1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] -4-piperidyl] acetaldehyde (500 mg, 1.30 mmol) , 3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane (359.09 mg, 1.30 mmol) in DCM (10 mL) was added TEA (395.89 mg, 3.91 mmol, 544.55 μL) and NaBH (OAc) 3 (829.20 mg, 3.91 mmol) . The mixture was stirred at 20 ℃ for 2 h. The reaction mixture was added H2O (40 mL) and extracted with DCM (30 mL × 3) . The combined organic phases were dried with anhydrous Na2SO4, filtered and concentrated under reduced
pressure. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate = 100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 643.3 [M+H] +.
Step C: 5- [4- [2- [3- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] ethyl] -1-piperidyl] -2-
(2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [4- [2- [3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] ethyl] -1-piperidyl] isoindoline-1, 3-dione (850 mg, 912.50 μmol) in EtOH (20 mL) was added Fe (509.58 mg, 9.12 mmol) and NH4Cl (2 M, 3.45 mL) . The mixture was stirred at 80 ℃ for 16 h. The reaction mixture was concentrated under reduced pressure to give crude product. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol = 100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 613.1 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.09 (s, 1H) , 7.66 (d, J = 8.8 Hz, 1H) , 7.32 (s, 1H) , 7.25 (d, J = 8.8 Hz, 1H) , 6.70 (d, J = 8.8 Hz, 2H) , 6.48 (d, J = 8.4 Hz, 2H) , 5.06 (dd, J = 5.6, 12.8 Hz, 1H) , 4.06 (d, J = 12.8 Hz, 2H) , 3.16 (d, J = 3.6 Hz, 2H) , 3.06 -3.02 (m, 5H) , 3.01 -2.91 (m, 4H) , 2.89-2.79 (m, 5H) , 2.62-2.55 (m, 2H) , 2.42 -2.35 (m, 1H) , 2.06 -1.96 (m, 1H) , 1.77-1.73 (m, 1H) , 1.63-1.48 (m, 10 H) , 1.27 -1.20 (m, 2H) .
Step D: 3- [5- (4- {2- [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] ethyl} -1-piperidyl) -1, 3-dioxo-2-
isoindolinyl] -2, 6-piperidinedione
To a solution of 5- [4- [2- [3- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] ethyl] -1-piperidyl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (50 mg, 81.60 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (32.76 mg, 81.60 μmol) in n-BuOH (2 mL) was added AcOH (49.00 mg, 815.98 μmol, 46.71 μL) . The mixture was stirred at 100 ℃ for 4 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: Xtimate C18 150*40mm*10um; mobile phase: [water (FA) -ACN] ; gradient: 30%-60%B over 6 min) to afford the desired product.
LCMS: MS (ESI) m/z 934.7 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.08 (s, 1H) , 10.04 (s, 1H) , 8.82 (s, 1H) , 8.19 (FA, 0.62H) , 8.01 (s, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.65 (d, J = 9.2 Hz, 1H) , 7.61 -7.50 (m, 3H) , 7.30 (s, 1H) , 7.23 (d, J = 8.4 Hz, 1H) , 6.90 (d, J = 8.0 Hz, 2H) , 5.45-5.20 (m, 3H) , 5.06 (dd, J = 12.8, 6.4 Hz, 1H) , 4.45 (m, 2H) , 4.04 (d, J = 12.4 Hz, 2H) , 3.07 (m, 4H) , 2.99 -2.90 (m, 2H) , 2.85-
2.80 (m, 1H) , 2.57-2.54 (m, 1H) , 2.44 (m, 4H) , 2.06 -1.90 (m, 4H) , 1.81-1.68 (m, 3H) , 1.60-1.51 (m, 9H) , 1.48 (m, 4H) , 1.43-1.35 (m, 3H) , 1.25-1.15 (m, 3H) .
Example 101:
3- [5- (4- { [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -3-aza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-179)
Scheme:
Step A: tert-butyl 9- (trifluoromethylsulfonyloxy) -3-azaspiro [5.5] undec-9-ene-3-
carboxylate
To a solution of tert-butyl 9-oxo-3-azaspiro [5.5] undecane-3-carboxylate (3 g, 11.22 mmol) in THF (20 mL) was added dropwise LDA (2.0 M, 6.73 mL) at -78℃. After addition, the mixture was stirred at -78℃ for 2 hr, and then 1, 1, 1-trifluoro-N-phenyl-N- (trifluoromethylsulfonyl) methanesulfonamide (4.01 g, 11.22 mmol) in THF (10 mL) was added dropwise at -78 ℃. The resulting mixture was stirred at -78 ℃ for 1.5hr. Then the mixture solution was warmed to 20 ℃ for 2.5 h. The reaction mixture was quenched with aq. NH4Cl (100 mL) and extracted with EtOAc (100 mL × 2) . The organic layer was washed with brine (100 mL) , dried over Na2SO4 and concentrated to give a residue, which was purified by column chromatography on silica gel (12%EtOAc in PE) to afford the desired product.
1H NMR (400 MHz, CDCl3) δ 5.69 (t, J = 4.0 Hz, 1H) , 3.52 -3.41 (m, 2H) , 3.39 -3.27 (m, 2H) , 2.34 (t, J = 1.6, 6.0 Hz, 2H) , 2.14 -2.06 (m, 2H) , 1.67 (t, J = 6.4 Hz, 2H) , 1.46 (s, 9H) , 1.44 -1.40 (m, 4H) .
19F NMR (400 MHz, DMSO-d6) δ –73.993.
Step B: tert-butyl 9- (4-nitrophenyl) -3-azaspiro [5.5] undec-9-ene-3-carboxylate
A mixture of tert-butyl 9- (trifluoromethylsulfonyloxy) -3-azaspiro [5.5] undec-9-ene-3-carboxylate (1 g, 2.50 mmol) , (4-nitrophenyl) boronic acid (501.50 mg, 3.00 mmol) , Pd (dppf) Cl2 (183.19 mg, 250.36 μmol) , Na2CO3 (796.06 mg, 7.51 mmol) in dioxane (6 mL) and H2O (3 mL) was stirred at 110 ℃ for 16 hr under N2 atmosphere. The mixture was diluted with water (100 mL) and extracted with ethyl acetate (100 mL × 2) . The combined organic layers were washed with brine (50 mL × 2) , dried over Na2SO4, filtered and concentrated to afford the desired product.
1H NMR (400 MHz, CDCl3) δ 8.17 (d, J = 8.8 Hz, 2H) , 7.52 (d, J = 8.8 Hz, 2H) , 6.28 (t, J = 4.0 Hz, 1H) , 3.61 -3.44 (m, 2H) , 3.41 -3.28 (m, 2H) , 2.46 (d, J = 2.0 Hz, 2H) , 2.18 (m, 2H) , 1.70 (m, 2H) , 1.51 -1.42 (m, 13H) .
Step C: tert-butyl 9- (4-aminophenyl) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9- (4-nitrophenyl) -3-azaspiro [5.5] undec-9-ene-3-carboxylate (300 mg, 805.46 μmol) in THF (10 mL) was added Pd/C (100 mg, 10%purity, wet) and Pd (OH) 2 /C (100 mg, 20%purity, wet) . The mixture was stirred at 20 ℃ for 16 h under H2 (50 Psi) . The reaction mixture was filtered, washed with EtOAc (200 mL) and concentrated to afford the desired product.
1H NMR (400 MHz, CDCl3) δ 7.02 (d, J = 8.4 Hz, 2H) , 6.67 -6.62 (m, 2H) , 3.40 (q, J = 6.0 Hz, 4H) , 2.32 -3.45 (m, 1H) , 1.79 (d, J = 12.8 Hz, 2H) , 1.73 -1.64 (m, 2H) , 1.61 -1.52 (m, 4H) , 1.47 (s, 9H) , 1.37 -1.31 (m, 2H) , 1.28 -1.20 (m, 2H) .
Step D: tert-butyl 9- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
yl] amino] phenyl] -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 124.55 μmol) , tert-butyl 9- (4-aminophenyl) -3-azaspiro [5.5] undecane-3-carboxylate (64.36 mg, 186.83 μmol) in n-BuOH (2 mL) . The mixture was stirred at 100 ℃ for 3 hr. The mixture was concentrated to give a residue, which was purified by silica gel column chromatography (10%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 666.5 [M+H] +.
Step E: (12Z, 16R) -5- [4- (3-azaspiro [5.5] undecan-9-yl) anilino] -16-hydroxy-16-methyl-
2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-
one
To a solution of tert-butyl 9- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-yl] amino] phenyl] -3-azaspiro [5.5] undecane-3-carboxylate (100 mg, 150.19 μmol) in DCM (2 mL) was added TFA (1.54 g, 13.46 mmol, 1 mL) . The mixture was stirred at 20 ℃ for 1hr. The mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 566.5 [M+H] +.
Step F: 3- [5- (4- { [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3-aza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -2, 6-
piperidinedione
To a solution of (12Z, 16R) -5- [4- (3-azaspiro [5.5] undecan-9-yl) anilino] -16-hydroxy-16-methyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (80 mg, 141.42 μmol) , 1- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde (75.39 mg, 212.12 μmol) , TEA (42.93 mg, 424.25 μmol) in DCM (3 mL) was added NaBH (OAc) 3 (89.92 mg, 424.25 μmol) . The mixture was stirred at 20 ℃ for 3 hr. The mixture was concentrated to give a residue, which was purified by prep-HPLC (column: Welch Xtimate C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 7%-47%B over 9 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 906.0 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ10.95 (s, 1H) , 10.14 (s, 1H) , 8.86 (s, 1H) , 8.18 (FA, 0.73H) , 7.97 -8.07 (m, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.62 (dd, J = 13.6, 8.0 Hz, 3H) , 7.50 (d, J = 8.4 Hz, 1H) , 7.18 (d, J = 8.4 Hz, 2H) , 6.98 -7.09 (m, 2H) , 5.25 -5.38 (m, 2H) , 5.04 (dd, J = 13.2,
5.2 Hz, 1H) , 4.47 (m, 2H) , 4.15 -4.35 (m, 2H) , 3.86 (d, J = 12.0 Hz, 2H) , 2.75 -2.95 (m, 5H) , 2.54 -2.70 (m, 2H) , 2.32 -2.42 (m, 6H) , 2.14 -2.23 (s, 3H) , 1.90 -2.10 (m, 3H) , 1.68 -1.80 (m, 5H) , 1.51 -1.65 (m, 8H) , 1.36 (m, 2H) , 1.11 -1.22 (m, 4H) .
Example 102:
3- [5- (4- { [9- (4- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} -2-tolyl) -3-aza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-180)
Scheme:
Step A: tert-butyl 9- (2-methyl-4-nitro-phenyl) -3-azaspiro [5.5] undec-9-ene-3-carboxylate
A mixture of tert-butyl 9- (trifluoromethylsulfonyloxy) -3-azaspiro [5.5] undec-9-ene-3-carboxylate (1 g, 2.50 mmol) , (2-methyl-4-nitro-phenyl) boronic acid (453.04 mg, 2.50 mmol) , Pd (dppf) Cl2 (183.19 mg, 250.36 μmol) , Na2CO3 (796.07 mg, 7.51 mmol) in dioxane (6 mL) and H2O (3 mL) was stirred at 110 ℃ for 16 hr under N2 atmosphere. The mixture was diluted with
water (100 mL) and extracted with ethyl acetate (100 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by silica gel column chromatography (12%EA in PE) to afford the desired product.
1H NMR (400 MHz, CDCl3) δ 8.04 (d, J = 2.0 Hz, 1H) , 7.99 (dd, J = 2.0, 8.4 Hz, 1H) , 7.20 (d, J = 8.4 Hz, 1H) , 5.56 (s, 1H) , 3.65 -3.46 (m, 2H) , 3.44 -3.28 (m, 2H) , 2.37 (s, 3H) , 2.22 (d, J =2.0 Hz, 2H) , 2.11 (d, J = 3.2 Hz, 2H) , 1.67 (t, J = 6.4 Hz, 2H) , 1.53 -1.44 (m, 13H) .
Step B: tert-butyl 9- (4-amino-2-methyl-phenyl) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9- (2-methyl-4-nitro-phenyl) -3-azaspiro [5.5] undec-9-ene-3-carboxylate (300 mg, 776.23 μmol) in THF (10 mL) was added Pd/C (100 mg, 10%purity, wet) and Pd (OH) 2 /C (100 mg, 20%purity, wet) . The mixture was stirred at 20 ℃ for 16 h under H2 (50 Psi) . The reaction mixture was filtered, washed with EtOAc (200mL) and concentrated to afford the desired product.
1H NMR (400 MHz, CDCl3) δ 7.01 (d, J = 8.0 Hz, 1H) , 6.58 -6.48 (m, 2H) , 3.40 (d, J = 3.2 Hz, 4H) , 2.67 -2.52 (m, 1H) , 2.25 (s, 3H) , 1.80 (d, J = 12.8 Hz, 2H) , 1.65 -1.52 (m, 6H) , 1.47 (s, 9H) , 1.34 (d, J = 5.2 Hz, 2H) , 1.30 -1.21 (m, 4H) .
Step C: tert-butyl 9- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-yl] amino] -2-
methyl-phenyl] -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 124.55 μmol) , tert-butyl 9- (4-amino-2-methyl-phenyl) -3-azaspiro [5.5] undecane-3-carboxylate (66.98 mg, 186.83 μmol) in n-BuOH (2 mL) . The mixture was stirred at 100 ℃ for 3 hr. The mixture was concentrated to give a residue, which was purified by silica gel column chromatography (10%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 680.4 [M+H] +.
Step D: (12Z, 16R) -5- [4- (3-azaspiro [5.5] undecan-9-yl) -3-methyl-anilino] -16-hydroxy-16-
methyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-
heptaen-9-one
To a solution of tert-butyl 9- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-yl] amino] -2-methyl-phenyl] -3-azaspiro [5.5] undecane-3-carboxylate (80 mg, 117.67 μmol) in DCM (2 mL) was added TFA (1.54 g, 13.46 mmol, 1 mL) . The mixture was stirred at 20 ℃ for 1 hr. The mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 580.4 [M+H] +.
Step E: 3- [5- (4- { [9- (4- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} -2-
tolyl) -3-aza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -2, 6-
piperidinedione
To a solution of (12Z, 16R) -5- [4- (3-azaspiro [5.5] undecan-9-yl) -3-methyl-anilino] -16-hydroxy-16-methyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (80 mg, 137.99 μmol) , 1- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde (73.56 mg, 206.99 μmol) , TEA (41.89 mg, 413.98 μmol) in DCM (3 mL) was added NaBH (OAc) 3 (87.74 mg, 413.98 μmol) . The mixture was stirred at 20 ℃ for 3hr. The mixture was concentrated to give a residue, which was purified by prep-HPLC (column: Welch Xtimate C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 10%-50%B over 9 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 920.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.95 (s, 1H) , 10.10 (s, 1H) , 8.85 (s, 1H) , 8.18 (FA, 0.66H) , 8.05 -7.98 (m, 1H) , 7.79 (d, J = 8.0 Hz, 1H) , 7.61 (d, J = 7.6 Hz, 2H) , 7.50 (d, J = 8.4 Hz, 1H) , 7.40 (d, J = 7.6 Hz, 1H) , 7.17 (d, J = 9.2 Hz, 1H) , 7.07 -7.00 (m, 2H) , 5.38 -5.25 (m, 2H) , 5.04 (dd, J = 13.2, 5.2 Hz, 1H) , 4.47 (m, 2H) , 4.35 -4.15 (m, 2H) , 3.87 (d, J = 11.6 Hz, 2H) , 2.96 -2.76 (m, 5H) , 2.58 (d, J = 14.8 Hz, 2H) , 2.38 (m, 6H) , 2.26 (m, 3H) , 2.19 (d, J = 5.6 Hz, 3H) , 2.02 –1.90 (m, 3H) , 1.83 -1.69 (m, 5H) , 1.62 (m, 2H) , 1.56 (m, 3H) , 1.54 -1.47 (m, 3H) , 1.37 (m, 2 H) , 1.25 -1.13 (m, 4H) .
Example 103:
3- {5- [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] -1-oxo-2-isoindolinyl} -2, 6-piperidinedione (Cpd-190)
Scheme:
Step A: tert-butyl 4- [4- (4-nitrophenyl) piperazin-1-yl] piperidine-1-carboxylate
To solution of 1- (4-nitrophenyl) piperazine (3 g, 14.48 mmol) and tert-butyl 4-oxopiperidine-1-carboxylate (3.00 g, 15.06 mmol) in DCE (20 mL) was added AcOH (1.05 g, 17.47 mmol, 1 mL) . The mixture was stirred at 25 ℃ for 1 hr. Then the mixture was added NaBH3CN (1.36 g, 21.72 mmol) and stirred at 25 ℃ for 1 hr. The mixture was diluted with water (20 mL) and extracted with EtOAc (100 mL × 3) . The combined organic layers were dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by flash silica gel chromatography (20 gSilica Flash Column, Eluent of 0~5%MeOH/DCM gradient @30 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 391.3 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.11 (d, J = 9.6 Hz, 2H) , 6.81 (d, J = 9.6 Hz, 2H) , 4.25 -4.04 (m, 2H) , 3.94 -3.75 (m, 1H) , 3.71 (t, J = 6.4 Hz, 1H) , 3.45 (s, 4H) , 2.75 (s, 5H) 2.58 -2.47 (m, 1H) , 2.44 (t, J = 6.0 Hz, 1H) , 1.84 (d, J = 11.6 Hz, 2H) , 1.45 (s, 9H) .
Step B: 1- (4-nitrophenyl) -4- (4-piperidyl) piperazine
To a solution of tert-butyl 4- [4- (4-nitrophenyl) piperazin-1-yl] piperidine-1-carboxylate (2.5 g, 6.40 mmol) in DCM (5 mL) was added HCl/dioxane (5 mL) . The mixture was stirred at 25 ℃for 1 hr. The reaction mixture was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 291.3 [M+H] +.
Step C: 1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] piperidine-4-carbaldehyde
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [4- (hydroxymethyl) -1-piperidyl] isoindoline-1, 3-dione (1 g, 2.69 mmol) in DCM (10 mL) was added DMP (1.37 g, 3.23 mmol, 1.00 mL) at 0 ℃. The mixture was stirred at 25℃ for 1h. The mixture was added water (20 mL) , extracted with ethyl acetate (100 mL × 3) . The combined organic layers were dried over Na2SO4, filtered and concentrated in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 370.0 [M+H] +.
Step D: 2- (2, 6-dioxo-3-piperidyl) -5- [4- [ [4- [4- (4-nitrophenyl) piperazin-1-yl] -1-
piperidyl] methyl] -1-piperidyl] isoindoline-1, 3-dione
To a solution of 1- (4-nitrophenyl) -4- (4-piperidyl) piperazine (1 g, 3.06 mmol, HCl) and 1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] piperidine-4-carbaldehyde (1.6 g, 4.33 mmol) in DCM (20 mL) was added NaBH (OAc) 3 (972.74 mg, 4.59 mmol) and TEA (928.85 mg, 9.18 mmol, 1.28 mL) . The mixture was stirred at 25℃ for 2 h. The mixture was added water (10 mL) , extracted with ethyl acetate (100 mL × 3) . The combined organic layers were dried over Na2SO4, filtered and concentrated in vacuum.
The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~10%MeOH/DCM gradient @30 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 644.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.08 (s, 1H) , 8.05 (d, J = 9.6 Hz, 2H) , 7.64 (d, J = 8.4 Hz, 1H) , 7.30 (d, J = 2.0 Hz, 1H) , 7.22 (dd, J = 2.0, 8.8 Hz, 1H) , 7.02 (d, J = 9.6 Hz, 2H) , 5.06 (dd, J = 5.2, 13.2 Hz, 1H) , 4.03 (d, J = 12.0 Hz, 2H) , 3.42 (m, 4H) , 3.31 (d, J = 8.4 Hz, 2H) , 2.99 -2.92 (m, 2H) , 2.88 -2.81 (m, 2H) , 2.66 -2.54 (m, 5H) , 2.21 (t, J = 11.2 Hz, 1H) , 2.15 -2.07 (m, 2H) , 2.04 -1.97 (m, 1H) , 1.90 -1.69 (m, 7H) , 1.49 -1.34 (m, 2H) , 1.19 -1.03 (m, 2H)
Step E: 3- [5- [4- [ [4- [4- (4-aminophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-
oxo-isoindolin-2-yl] piperidine-2, 6-dione &3- [6- [4- [ [4- [4- (4-aminophenyl) piperazin-1-yl] -1-
piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [4- [ [4- [4- (4-nitrophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] isoindoline-1, 3-dione (900 mg, 1.40 mmol) in TFA (20 mL) was added Zn (2.3 g, 35.17 mmol ) . The mixture was stirred at 75 ℃ for 2hr. The mixture was
filtered through diatomite pad under vacuum and washed with water (15mL × 3) . The filtrate was evaporated in vacuum to a residue. The crude product was purified by reversed-phase HPLC (column: Welch Xtimate C18 100*40mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 0%-17%B over 7 min) and dried by lyophilization to afford the desired product.
3- [5- [4- [ [4- [4- (4-aminophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
LCMS: MS (ESI) m/z 600.5 [M+H] +. tR = 2.220 min
1HNMR (400 MHz, DMSO-d6) : δ10.96 (s, 1H) , 7.52 (d, J = 8.4 Hz, 1H) , 7.21 (d, J = 8.8 Hz, 2H) , 7.11 -7.05 (m, 4H) , 5.05 (dd, J = 5.2, 13.3 Hz, 1H) , 4.37 -4.15 (m, 4H) , 3.91 (br d, J =12.8 Hz, 4H) , 3.72 (br d, J = 10.0 Hz, 3H) , 3.06 -2.78 (m, 9H) , 2.65 -2.54 (m, 1H) , 2.44 -2.23 (m, 4H) , 2.14 -1.91 (m, 5H) , 1.82 (br d, J = 12.0 Hz, 3H) , 1.37 -1.21 (m, 2H)
3- [6- [4- [ [4- [4- (4-aminophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
LCMS: MS (ESI) m/z 600.5 [M+H] +. tR = 2.257 min
1HNMR (400 MHz, DMSO-d6) : δ10.98 (s, 1H) , 9.72 -9.26 (m, 2H) , 7.44 (d, J = 8.4 Hz, 1H) , 7.29 (dd, J = 2.0, 8.5 Hz, 1H) , 7.26 -7.18 (m, 3H) , 7.10 (br d, J = 9.2 Hz, 2H) , 5.09 (dd, J = 5.2, 13.1 Hz, 1H) , 4.38 -4.16 (m, 6H) , 3.87 -3.67 (m, 6H) , 3.56 -3.45 (m, 2H) , 3.10 -2.84 (m, 6H) , 2.78 (br t, J = 11.6 Hz, 2H) , 2.62 (br s, 1H) , 2.46 -2.27 (m, 3H) , 2.11 -1.95 (m, 4H) , 1.85 (br d, J = 12.1 Hz, 2H) , 1.43 -1.25 (m, 2H)
Step F: 3- {5- [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] -1-oxo-2-isoindolinyl} -2, 6-
piperidinedione
To a solution of 3- [5- [4- [ [4- [4- (4-aminophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (65.74 mg, 109.61 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (40 mg, 99.64 μmol) in t-BuOH (2 mL) was added TFA (50 mg, 438.51 μmol) . The mixture was stirred at 100 ℃ for 16hr. The reaction mixture was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 921.8 [M+H] +.
HNMR (400 MHz, DMSO-d6) δ10.95 (s, 1H) , 10.19 -10.02 (m, 1H) , 9.51 -9.32 (m, 1H) , 8.85 (s, 1H) , 8.04 -7.95 (m, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.62 (dd, J = 8.4, 15.0 Hz, 3H) , 7.53 (d, J = 8.8 Hz, 1H) , 7.11 -7.04 (m, 2H) , 7.00 (br d, J = 9.2 Hz, 2H) , 5.40 -5.24 (m, 2H) , 5.04 (dd, J =4.8, 13.3 Hz, 1H) , 4.47 (br d, J = 5.2 Hz, 2H) , 4.32 (d, J = 16.8 Hz, 1H) , 4.19 (d, J = 16.8 Hz, 1H) , 4.04 -3.64 (m, 8H) , 3.09 -2.80 -2.80 (m, 10H) , 2.70 -2.53 (m, 2H) , 2.40 -2.28 (m, 3H) ,
2.27 -1.90 (m, 9H) , 1.82 (br d, J = 11.9 Hz, 2H) , 1.75 -1.66 (m, 1H) , 1.56 (m, 3H) , 1.37 -1.19 (m, 2H) .
Example 104:
3- (6- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-132)
Scheme:
Step A: tert-butyl 8- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] -2, 8-
diazaspiro [4.5] decane-2-carboxylate
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (2 g, 7.24 mmol) in DMF (40 mL) was added DIPEA (1.87 g, 14.48 mmol) and tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (1.74 g, 7.24 mmol) . The mixture was stirred at 100 ℃ for 1 hr. The mixture was cooled down to 25℃ and the mixture was poured into H2O (50 mL) . The mixture was filtered and the filter cake was dried under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 497.3 [M+H] +.
Step B: 5- (2, 8-diazaspiro [4.5] decan-8-yl) -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione
To a solution of tert-butyl 8- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] -2, 8-diazaspiro [4.5] decane-2-carboxylate (3.7 g, 7.45 mmol) in DCM (20 mL) was added HCl/dioxane (4 M, 20 mL) . The mixture was stirred at 20 ℃ for 1hr. The mixture filtered and the cake was dried in vacuo to afford the desired product.
LCMS: MS (ESI) m/z 397.1 [M+H] +.
Step C: 2- (2, 6-dioxo-3-piperidyl) -5- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-
diazaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione
To a solution of 5- (2, 8-diazaspiro [4.5] decan-8-yl) -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (2.9 g, 6.70 mmol) , 1- (4-nitrophenyl) piperidin-4-one (1.48 g, 6.70 mmol) in DCM (5 mL) was added Et3N (2.71 g, 26.80 mmol) and the mixture was stirred at 25℃ for 1 hr. The mixture was added NaBH (OAc) 3 (4.26 g, 20.10 mmol) and the mixture was stirred at 25 ℃ for 1 hr. The mixture was concentrated to give a residue. The residue was purified by flash silica gel chromatography (120 gSilica Flash Column, Eluent of 0~10 %Methanol/Dichloromethane gradient @60 mL/min) and the combined desired fractions was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 601.3 [M+H] +.
Step D: 3- [6- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-
isoindolin-2-yl] piperidine-2, 6-dione &3- [5- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-
diazaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione (1.9 g, 3.16 mmol) in TFA (30 mL) was added Zn (9.62 g, 147.12 mmol) . The mixture was stirred at 75 ℃ for 1 hr. The mixture was filtered through diatomite pad under vacuum, washed with CH3CN (50 mL) and the filtrate was evaporated in vacuum to a residue. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-21%B over 8 min) and dried by lyophilization to afford the desired product.
3- [6- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
LCMS: MS (ESI) m/z 556.8 [M+H] +. tR = 1.020 min
1H NMR (400MHz, DMSO-d6) δ10.97 (s, 1H) , 10.18 (br s, 1H) , 7.44 (d, J = 8.4 Hz, 1H) , 7.35 -7.27 (m, 1H) , 7.23 (s, 1H) , 7.18 (br d, J = 8.8 Hz, 2H) , 7.08 (br d, J = 9.2 Hz, 2H) , 5.09 (dd, J =5.2, 13.2 Hz, 1H) , 4.38 -4.31 (m, 1H) , 4.25 -4.16 (m, 1H) , 3.86 (br d, J = 12.4 Hz, 2H) , 3.66 (br s, 1H) , 3.55 (br d, J = 10.8 Hz, 1H) , 3.40 -3.30 (m, 2H) , 3.25 -3.18 (m, 1H) , 3.05 (br s, 1H) , 2.97 -2.83 (m, 1H) , 2.81 -2.66 (m, 2H) , 2.60 (br d, J = 16.8 Hz, 1H) , 2.47 -2.42 (m, 2H) , 2.42 -2.28 (m, 2H) , 2.14 (br d, J = 10.8 Hz, 2H) , 2.06 -1.91 (m, 2H) , 1.90 -1.57 (m, 8H)
3- [5- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
LCMS: MS (ESI) m/z 556.4 [M+H] +. tR = 2.534 min
Step E: 3- (6- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 3- [6- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (220 mg, 328.01 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (100 mg, 259.44 μmol) in n-BuOH (1 mL) was added AcOH (155.80 mg, 2.59 mmol) . The mixture was stirred at 100℃ for 1 hr. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: Xtimate C18 150*40mm*10um; mobile phase: [water (FA) -ACN] ; gradient: 6%-36%B over 7 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 878.8 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.97 (s, 1H) , 10.03 (br s, 1H) , 8.81 (s, 1H) , 8.21 (s, 1H) , 8.01 (br t, J = 7.6 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.62 -7.50 (m, 3H) , 7.41 (d, J = 8.4 Hz, 1H) , 7.25 (br d, J = 8.8 Hz, 1H) , 7.16 (s, 1H) , 6.91 (br d, J = 9.2 Hz, 2H) , 5.35 (br s, 2H) , 5.09 (dd, J = 5.2, 13.2 Hz, 1H) , 4.45 (br d, J = 5.6 Hz, 2H) , 4.33 (d, J = 16.8 Hz, 1H) , 4.20 (d, J = 16.8 Hz, 1H) , 3.58 (br d, J = 11.6 Hz, 2H) , 3.28 -3.17 (m, 4H) , 2.98 -2.82 (m, 1H) , 2.78 -2.59 (m, 5H) , 2.57 (br s, 2H) , 2.45 -2.31 (m, 2H) , 2.31 -2.12 (m, 2H) , 2.08 -1.87 (m, 5H) , 1.76 -1.47 (m, 12H)
Example 105:
3- (6- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -5-fluoro-1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-191)
Scheme:
Step A: tert-butyl 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane-8-carboxylate
To a mixture of tert-butyl 3-oxo-8-azaspiro [4.5] decane-8-carboxylate (1 g, 3.95 mmol) and 1- (4-
nitrophenyl) piperazine (1.23 g, 5.92 mmol) in DCM (20 mL) was added AcOH (237.04 mg, 3.95 mmol, 225.97 μL) and NaBH (OAc) 3 (1.25 g, 5.92 mmol) . The mixture was stirred at 25 ℃for 2hr. The reaction mixture was added H2O (300 mL) and extracted with DCM (150 mL × 3) . The combined organic phases were dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was triturated with EtOAc (10 mL) and Petroleum ether (30 mL) to afford the desired product.
To a mixture of tert-butyl 3-oxo-8-azaspiro [4.5] decane-8-carboxylate (1 g, 3.95 mmol) and 1- (4-
nitrophenyl) piperazine (1.23 g, 5.92 mmol) in DCM (20 mL) was added AcOH (237.04 mg, 3.95 mmol, 225.97 μL) and NaBH (OAc) 3 (1.25 g, 5.92 mmol) . The mixture was stirred at 25 ℃for 2hr. The reaction mixture was added H2O (300 mL) and extracted with DCM (150 mL × 3) . The combined organic phases were dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was triturated with EtOAc (10 mL) and Petroleum ether (30 mL) to afford the desired product.
LCMS: MS (ESI) m/z 445.1 [M+H] +.
Step B: 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane
To a solution of tert-butyl 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane-8-carboxylate (1.26 g, 2.83 mmol) in DCM (15 mL) was added HCl/dioxane (15 mL, 4 M) . The mixture was stirred at 25 ℃ for 2 hr. The reaction mixture was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 344.9 [M+H] +.
Step C: 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-6- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-
azaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione
To solution of 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane (1 g, 2.90 mmol) , 2- (2, 6-dioxo-3-piperidyl) -5, 6-difluoro-isoindoline-1, 3-dione (1 g, 3.40 mmol) in DMF (30 mL) was added DIPEA (878.58 mg, 6.80 mmol) . The mixture was stirred at 90 ℃ for 4 h. The reaction mixture was added H2O (100 mL) and extracted with ethyl acetate (200 mL × 3) . The combined organic phases were washed with brine (200 mL × 3) , dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was triturated with DCM (30 mL) and PE (10 mL) to afford the desired product.
LCMS: MS (ESI) m/z 619.2 [M+H] +.
Step D: 3- [5- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -6-fluoro-1-
hydroxy-3-oxoisoindolin-2-yl] piperidine-2, 6-dione
To solution of 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-6- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione (1.8 g, 2.91 mmol) in TFA (18 mL) was added Zn (3.82 g, 58.42 mmol) . The mixture was stirred at 75 ℃ for 1 h. The reaction mixture was filtered. The residue washed with EtOAc (100 mL) , concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 591.3 [M+H] +.
Step E: 3- [6- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -5-fluoro-1-
oxo-isoindolin-2-yl] piperidine-2, 6-dione
To solution of 3- [5- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -6-fluoro-1-hydroxy-3-oxoisoindolin-2-yl] piperidine-2, 6-dione (800 mg, 1.35 mmol) and 3- [6- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -5-fluoro-1-hydroxy-3-oxo-isoindolin-2-yl] piperidine-2, 6-dione (800.00 mg, 1.35 mmol) in DCM (20 mL) was added Et3SiH (3.01 g, 25.91 mmol) and TFA (12.70 g, 111.41 mmol) . The mixture was stirred at 20 ℃ for 16 h. The reaction mixture was filtered. The residue washed with EtOAc (100 mL) , concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column:
Xtimate C18 150*40mm*10um; mobile phase: [water (FA) -ACN] ; gradient: 8%-38%B over 7 min) to afford the desired product, then dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 575.4 [M+H] +.
2D NMR showed H11 has NOE correlation to H4, which is consistent with the determined structure.
1H NMR (400 MHz, DMSO-d6) δ 10.98 (s, 1H) , 7.44 (d, J = 11.6 Hz, 1H) , 7.33 (d, J = 7.6 Hz, 1H) , 7.13-7.08 (m, 2H) , 7.05-6.98 (s, 2H) , 5.07 (dd, J = 4.8, 13.6 Hz, 1H) , 4.42 -4.20 (m, 2H) , 3.80-3.72 (m, 2H) , 3.65-3.55 (m, 4H) , 3.14 (s, 2H) , 3.12-2.98 (m, 4H) , 2.96 -2.82 (m, 1H) , 2.70-2.58 (m, 1H) , 2.41 -2.36 (m, 1H) , 2.21-2.09 (m, 2H) , 2.02-1.97 (s, 1H) , 1.92-1.85 (m, 1H) , 1.77 -1.56 (m, 8H) .
Step F: 3- (6- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -5-fluoro-1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 3- [6- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -5-fluoro-1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (88 mg, 153.13 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 124.55 μmol) in n-BuOH (2 mL) was added AcOH (74.80 mg, 1.25 mmol) .
The mixture was stirred at 100 ℃ for 16 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: Welch Xtimate C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 7%-47%B over 9 min) to afford the desired product.
LCMS: MS (ESI) m/z 896.5 [M+H] +.
19F NMR (400 MHz, DMSO-d6) δ -116.253
1H NMR (400 MHz, DMSO-d6) δ10.98 (s, 1H) , 10.04 (s, 1H) , 8.82 (s, 1H) , 8.14 (FA, 0.32H) , 8.01 (t, J = 7.6 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.57 (t, J = 7.2 Hz, 3H) , 7.41 (d, J = 12.0 Hz, 1H) , 7.32 (d, J = 8.0 Hz, 1H) , 6.90 (d, J = 8.8 Hz, 2H) , 5.45 -5.30 (m, 2H) , 5.27 (s, 1H) , 5.08 (dd, J = 4.8, 13.2 Hz, 1H) , 4.46 (m, 2H) , 4.39 -4.21 (m, 2H) , 3.09 (m, 4H) , 2.99 (ms, 4H) , 2.96 -2.84 (m, 2H) , 2.68 -2.62 (m, 1H) , 2.58 (m, 4H) , 2.43 -2.33 (m, 1H) , 2.17 (s, 1H) , 2.02 -1.83 (m, 5H) , 1.73 -1.65 (m, 2H) , 1.62 -1.56 (m, 4H) , 1.56 (m, 3H) , 1.54 -1.47 (m, 2H) , 1.39 -1.29 (m, 1H) .
Example 106:
3- (6- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2, 4-diaza-2-indanyl) -2, 6-piperidinedione (Cpd-192)
Scheme:
Step A: methyl 5-bromo-2- (bromomethyl) pyridine-3-carboxylate
A mixture of methyl 5-bromo-2-methyl-pyridine-3-carboxylate (3.0 g, 13.04 mmol) , NBS (2.79 g, 15.65 mmol) , BPO (94.76 mg, 391.21 μmol) in CCl4 (30 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 80 ℃ for 16 hr under N2 atmosphere. The
reaction mixture was concentrated to give a residue, which was purified by silica gel column chromatography (10%EtOAc in PE) to afford the desired product.
LCMS: MS (ESI) m/z 230.0 [M+H] +.
Step B: 3- (3-bromo-5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-yl) piperidine-2, 6-dione
A mixture of methyl 5-bromo-2- (bromomethyl) pyridine-3-carboxylate (2 g, 6.47 mmol) , 3-aminopiperidine-2, 6-dione (1.07 g, 6.47 mmol, HCl) , TEA (1.97 g, 19.42 mmol, 2.70 mL) in DMF (20 mL) was stirred at 80 ℃ for 2hr. The mixture was poured into water (100 mL) and extracted with ethyl acetate (100 mL × 3) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by silica gel column chromatography (7%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 329.1 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.04 (s, 1H) , 8.94 (d, J = 2.0 Hz, 1H) , 8.42 (d, J = 2.0 Hz, 1H) , 5.19 (dd, J = 5.2, 13.2 Hz, 1H) , 4.58 -4.30 (m, 2H) , 2.99 -2.85 (m, 1H) , 2.69 -2.57 (m, 1H) , 2.46 -2.36 (m, 1H) , 2.08 -1.97 (m, 1H) .
Step C: tert-butyl 8- [6- (2, 6-dioxo-3-piperidyl) -5-oxo-7H-pyrrolo [3, 4-b] pyridin-3-yl] -2, 8-
diazaspiro [4.5] decane-2-carboxylate
To a solution of 3- (3-bromo-5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-yl) piperidine-2, 6-dione (530 mg, 1.64 mmol) and tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (432 mg, 1.80 mmol) in dioxane (15.0 mL) were added 1, 3-bis [2, 6-bis (1-propylbutyl) phenyl] -4, 5-dichloro-2H-imidazol-1-ium-2-ide; 3-chloropyridine; dichloropalladium (80 mg, 81.76 μmol) and Cs2CO3 (1.60 g, 4.91 mmol) . The mixture was stirred at 100 ℃ for 15 hr. under nitrogen atmosphere. The reaction mixture was filtered and the filtrate was concentrated in vacuum. The residue was purified by column chromatography (SiO2, DCM/MeOH = 50/1 to 20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 484.3 [M+H] +.
1H NMR (400MHz, CDCl3) δ 8.47 (s, 1H) , 8.10 (s, 1H) , 7.61 (s, 1H) , 5.24 (dd, J=5.2, 13.4 Hz, 1H) , 4.48 -4.28 (m, 2H) , 3.52 -3.23 (m, 8H) , 3.00 -2.91 (m, 1H) , 2.90 -2.78 (m, 1H) , 2.48 -2.33 (m, 1H) , 2.30 -2.20 (m, 1H) , 1.80 -1.72 (m, 6H) , 1.48 (s, 12H)
Step D: 3- [3- (2, 8-diazaspiro [4.5] decan-8-yl) -5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-
yl] piperidine-2, 6-dione
To a solution of tert-butyl 8- [6- (2, 6-dioxo-3-piperidyl) -5-oxo-7H-pyrrolo [3, 4-b] pyridin-3-yl] -2,8-diazaspiro [4.5] decane-2-carboxylate (300 mg, 620.4 μmol) in DCM (2.0 mL) was added TFA (4.0 mL) . The mixture was stirred at 20 ℃ for 2hr. The reaction mixture was concentrated in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 384.2 [M+H] +.
Step E: 3- [3- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -5-oxo-7H-
pyrrolo [3, 4-b] pyridin-6-yl] piperidine-2, 6-dione
To a solution of 3- [3- (2, 8-diazaspiro [4.5] decan-8-yl) -5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-yl] piperidine-2, 6-dione (crude 560 mg) and 1- (4-nitrophenyl) piperidin-4-one (198 mg, 899.0 μmol) in DCM (10.0 mL) was added TEA (285 mg, 2.81 mmol) . The mixture was stirred at 20 ℃ for 15 hr under nitrogen atmosphere. Then NaBH (OAc) 3 (179 mg, 844.2 μmol) was added to the reaction mixture and stirred at 20 ℃ for 1 hr. The reaction mixture was quenched by addition NaHCO3 (10.0 mL) , extracted with DCM (20 mL×4) . The combined organic layers were washed with brine (20 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (4 g Silica Flash Column, Eluent of 0~5%Ethyl acetate/Petroleum ether gradient @25 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 588.2 [M+H] +.
1H NMR (400MHz, CDCl3) δ 8.47 (d, J=2.8 Hz, 1H) , 8.16 -8.06 (m, 3H) , 7.60 (d, J=2.8 Hz, 1H) , 6.83 (d, J=9.6 Hz, 2H) , 5.24 (dd, J=5.2, 13.2 Hz, 1H) , 4.48 -4.40 (m, 1H) , 4.37 -4.29 (m, 1H) , 4.03 -3.86 (m, 2H) , 3.33 -3.19 (m, 4H) , 3.10 -3.00 (m, 2H) , 2.99 -2.92 (m, 1H) , 2.90 -2.77 (m, 2H) , 2.48 -2.33 (m, 2H) , 2.32 -2.16 (m, 2H) , 2.14 -1.98 (m, 2H) , 1.79 -1.42 (m, 9H) , 1.27 (t, J=7.1 Hz, 1H) .
Step F: 3- [3- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -5-oxo-7H-
pyrrolo [3, 4-b] pyridin-6-yl] piperidine-2, 6-dione
To a solution of 3- [3- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-yl] piperidine-2, 6-dione (260 mg, 442.4 μmol) and TEA (90 mg, 884.8 μmol) in DCM (5.0 mL) and DMAC (1.0 mL) were added PdCl2 (78 mg, 442.4 μmol) and Et3SiH (1.03 g, 8.85 mmol) . The mixture was stirred at 20 ℃ for 15hr under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was dissolved with DMF (5.0 mL) and filtered. The filtration was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-22%B over 8 min) to afford the desired product.
LCMS: MS (ESI) m/z 558.4 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.99 (s, 1H) , 8.56 (d, J=2.8 Hz, 1H) , 7.59 (d, J=2.8 Hz, 1H) , 7.22 -7.12 (m, 2H) , 7.07 (d, J=9.2 Hz, 2H) , 5.15 (dd, J=5.2, 13.6 Hz, 1H) , 4.39 (d, J=16.0 Hz, 1H) , 4.22 (d, J=16.0 Hz, 1H) , 3.85 (d, J=13.2 Hz, 2H) , 3.36 -3.31 (m, 4H) , 3.08 -3.01 (m, 1H) , 2.95 -2.85 (m, 1H) , 2.74 (br t, J=11.9 Hz, 2H) , 2.68 -2.57 (m, 2H) , 2.47 -2.41 (m, 2H) , 2.41 -2.31 (m, 1H) , 2.14 (d, J=10.8 Hz, 2H) , 2.06 -1.95 (m, 2H) , 1.89 -1.75 (m, 2H) , 1.72 -1.65 (s, 4H)
Step G: 3- (6- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2, 4-diaza-2-indanyl) -2, 6-
piperidinedione
To a solution of 3- [3- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-yl] piperidine-2, 6-dione (45 mg, 80.6 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (36.00 mg, 93.4 μmol) in n-BuOH (1.5 mL) was added HOAc (49 mg, 806.9 μmol) . The mixture was stirred at 100 ℃ for 4hr. The reaction mixture was concentrated in vacuum. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 6%-36%B over 8 min) to afford the desired product.
LCMS: MS (ESI) m/z 879.5 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 11.00 (s, 1H) , 10.07 (s, 1H) , 9.64 (s, 1H) , 8.83 (s, 1H) , 8.57 (d, J=2.4 Hz, 1H) , 8.01 (t, J=7.6 Hz, 1H) , 7.75 (d, J=8.0 Hz, 1H) , 7.59 (dd, J=2.0, 5.2 Hz, 4H) , 6.97 (d, J=8.8 Hz, 2H) , 5.41 -5.26 (m, 2H) , 5.15 (dd, J=4.8, 13.2 Hz, 1H) , 4.45 (m, 2H) , 4.42 -4.18 (m, 2H) , 3.81 (d, J=11.6 Hz, 2H) , 3.72 -3.63 (m, 2H) , 3.14 -3.01 (m, 2H) , 2.99 -2.85 (m, 2H) , 2.76 -2.56 (m, 6H) , 2.47 -2.29 (m, 2H) , 2.25 -2.06 (m, 4H) , 2.05 -1.98 (m, 3H) , 1.97 -1.90 (m, 1H) , 1.87 -1.80 (m, 1H) , 1.75 -1.65 (m, 6H) , 1.56 (m, 3H) .
Example 107:
3- {6- [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] -1-oxo-2-isoindolinyl} -2, 6-piperidinedione (Cpd-198)
Scheme:
Step A: 3- {6- [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] -1-oxo-2-isoindolinyl} -2, 6-
piperidinedione
To a solution of 3- [6- [4- [ [4- [4- (4-aminophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (65.74 mg, 109.61 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (40 mg, 99.64 μmol) in t-BuOH (2 mL) was added TFA (50 mg, 438.51 μmol) . The mixture was stirred at 100 ℃ for 16hr. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC (column: Welch Xtimate C18 100*40mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 5%-35%B over 8 min ) to afford the desired product.
LCMS: MS (ESI) m/z 921.9 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H) , 10.19 -10.04 (m, 1H) , 9.37 (m, 1H) , 8.86 (s, 1H) , 8.03 -7.97 (m, 1H) , 7.76 (d, J = 8.0 Hz, 1H) , 7.63 (br dd, J = 8.0, 15.3 Hz, 3H) , 7.45 (d, J = 8.4 Hz, 1H) , 7.30 (dd, J = 1.6, 8.4 Hz, 1H) , 7.21 (s, 1H) , 7.01 (br d, J = 8.8 Hz, 2H) , 5.40 -5.28 (m, 2H) , 5.10 (dd, J = 5.2, 13.2 Hz, 1H) , 4.48 (m, 2H) , 4.35 (d, J = 16.8 Hz, 1H) , 4.21 (d, J = 16.8 Hz, 1H) , 3.82 (m, 4H) , 3.77 -3.69 (m, 3H) , 3.09 -2.85 (m, 8H) , 2.78 (m, 2H) , 2.63 (m, 2H) , 2.46 -2.28 (m, 4H) , 2.27 -2.11 (m, 2H) , 2.08 -1.92 (m, 7H) , 1.85 (m, 2H) , 1.75 -1.68 (m, 1H) , 1.57 (m, 3H) , 1.35 (m, 2H)
Example 108:
1- {5- [ (4- { [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) carbonyl] -2-chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-156)
Scheme:
Step A: 1- (5- (4- ( (9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-
carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
A mixture of 1- (2-chloro-5- (4- ( (9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (50.0 mg, 0.0802 mmol) , iron powder (22.5 mg, 0.401 mmol) , NH4Cl (21.4 mg, 0.401 mmol) in EtOH (5 mL) and H2O (1 mL) was stirred at 90℃ for 3 hrs. After completion of reaction, the mixture reaction was filtered through a Celite pad, and the filtrate was concentrated to give the crude. The crude was quenched with H2O (10 mL) , adjusted to pH 8 with solid NaHCO3 and extracted with EtOAc (3 x 30 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 593.3 [M+H] +.
Step B: 1- {5- [ (4- { [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) carbonyl] -2-
chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a solution of (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (10.0 mg, 0.0259 mmol) in DCM (5 mL) was added 1- (5- (4- ( (9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (40.0 mg, 0.0638 mmol) . Then the reaction mixture was stirred at 35℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EA (20 mL x3) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated. The residue was purified by prep-HPLC (Waters 2767, Column: Pursuit XRs C18, 21.2*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 16%~26%Retention Time: 9.5-10.5 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 914.1 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.51 (s, 1H) , 10.01 (s, 1H) , 8.81 (s, 1H) , 8.16 (s, 1H) , 8.02 (s, 1H) , 7.76 (d, J = 7.9 Hz, 1H) , 7.64 (d, J = 8.2 Hz, 1H) , 7.60-7.55 (m, 2H) , 7.54 (d, J = 1.9 Hz, 2H) , 7.38 (dd, J = 8.3, 1.9 Hz, 1H) , 6.90 (d, J = 9.1 Hz, 2H) , 5.44 -5.18 (m, 3H) , 4.44 (m, 4H) , 3.77 (s, 1H) , 3.64-3.56 (m, 3H) , 3.08 (m, 4H) , 3.06-3.03 (m, 1H) , 2.79-2.70 (m, 4H) , 2.38-2.32 (m, 4H) , 2.21-2.14 (m, 3H) , 2.09-2.02 (m, 1H) , 1.97-1.91 (m, 1H) , 1.86-1.77 (m, 2H) , 1.75-1.66 (m, 2H) , 1.59-1.55 (m, 4H) , 1.54-1.51 (m, 3H) , 1.49-1.43 (m, 4H) , 1.12-1.01 (m, 2H) .
Example 109:
1- {5- [ (4- { [9- (4- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} -2-tolyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) carbonyl] -2-chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-157)
Scheme:
Step A: 3- (2-methyl-4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane
To a solution of tert-butyl 9- (2-methyl-4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane-3-carboxylate (560 mg, 1.44 mmol) in HCl in dioxane (10 mL, 4 M) and DCM (10 mL) was stirred at 25℃ for 3 h. After completion of reaction, the reaction mixture was concentrated under reduced pressure to dryness to afford the desired product.
LCMS: MS (ESI) m/z 290.1 [M+H] +.
Step B: tert-butyl4- ( (9- (2-methyl-4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-
yl) methyl) piperidine-1-carboxylate
To a solution of 3- (2-methyl-4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane (50 mg, 0.173 mmol) , tert-butyl 4-formylpiperidine-1-carboxylate (36.8 mg, 0.173 mmol) , NaBH (OAc) 3 (110 mg, 0.519 mmol) , CH3COOH (31.1 mg, 0.519 mmol) in DCM (5 mL) . Then the mixture was stirred at 25℃ for 16 h. The reaction mixture was quenched with water (50 mL) , and the solution was extracted with EtOAc (50 mL x 3) . The combined organic layers were washed with brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product
was purified by silica gel chromatography eluted with DCM: MeOH=10: 1 to afford the desired product.
LCMS: MS (ESI) m/z 487.3 [M+H] +.
Step C: 3- (2-methyl-4-nitrophenyl) -9- (piperidin-4-ylmethyl) -3, 9-diazaspiro [5.5] undecane
To a solution of tert-butyl4- ( (9- (2-methyl-4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carboxylate (559 mg, 1.15 mmol) in HCl in dioxane (10 mL, 4 M) and DCM (10 mL) was stirred at 25℃ for 3 h. After completion of reaction, the reaction mixture was concentrated under reduced pressure to dryness to afford the desired product.
LCMS: MS (ESI) m/z 387.2 [M+H] +.
Step D: 1- (2-chloro-5- (4- ( (9- (2-methyl-4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-
yl) methyl) piperidine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 3- (2-methyl-4-nitrophenyl) -9- (piperidin-4-ylmethyl) -3, 9-diazaspiro [5.5] undecane (390 mg, 1.01 mmol) , perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (877 mg, 2.02 mmol) , DIEA (391 mg, 3.03 mmol) in DMF (10 mL) . Then the mixture was stirred at 25℃ for 3h. Water (100 mL) was added and the solution was extracted with EtOAc (100 mL x 3) . The combined organic layers were washed with brine (100 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. the crude product was purified by silica gel chromatography eluted with DCM: MeOH=10: 1 to afford the desired product.
LCMS: MS (ESI) m/z 637.3 [M+H] +.
Step E: 1- (5- (4- ( (9- (4-amino-2-methylphenyl) -3, 9-diazaspiro [5.5] undecan-3-
yl) methyl) piperidine-1-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (4- ( (9- (2-methyl-4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (692 mg, 1.09 mmol) , NH4Cl (289 mg, 5.45 mmol) , in H2O (2 mL) and EtOH (20 mL) were added Fe (305 mg, 5.45 mmol) , Then the mixture was stirred at 90℃ for 2h. After completion of reaction, the mixture was filtered through a Celite pad, water (200 mL) was added and in filtrate the solution was extracted with EtOAc (200 mL x 3) . The combined organic layers were washed with brine (200 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to dryness to afford the desired product.
LCMS: MS (ESI) m/z 607.3 [M+H] +.
Step F: 1- {5- [ (4- { [9- (4- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} -2-
tolyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) carbonyl] -2-chlorophenyl} -1, 3, 5, 6-
tetrahydro-2, 4-pyrimidinedione
To a solution of give 1- (5- (4- ( (9- (4-amino-2-methylphenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (200 mg, 0.330 mmol) in DCM (2 mL) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20 mg, 0.0519 mmol) , the mixture was stirred at 25℃ for 16 hrs. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 3767/Qda Column: Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.05%TFA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 17-27%Retention Time: 8.0-9.4 of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 464.9 [1/2M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.51 (s, 1H) , 10.07 (s, 1H) , 8.84 (s, 1H) , 8.01 (t, J = 7.9 Hz, 1H) , 7.79 (d, J = 8.1 Hz, 1H) , 7.62 (dd, J = 17.8, 8.0 Hz, 3H) , 7.55 (d, J = 1.9 Hz, 1H) , 7.38 (d, J = 6.3 Hz, 2H) , 7.00 (d, J = 8.9 Hz, 1H) , 5.43 –5.24 (m, 3H) , 4.46 (d, J = 4.7 Hz, 3H) , 3.76 (dd, J = 8.1, 6.7 Hz, 1H) , 3.66 –3.53 (m, 2H) , 3.29 (m, 2H) , 3.18 –2.93 (m, 2H) , 2.78 –2.72 (m, 6H) , 2.67 (td, J = 4.4, 2.4 Hz, 1H) , 2.33 (m, 3H) , 2.21 (m, 3H) , 2.15 (m, 2H) , 1.96 (dd, J = 28.6, 16.3 Hz, 3H) , 1.87 –1.63 (m, 5H) , 1.56 (m, 6H) , 1.50 (m, 2H) , 1.18 –0.98 (m, 2H) .
Example 110:
1- {5- [ (4- { [3- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -3-aza-9-spiro [5.5] undecyl] methyl} -1-piperazinyl) carbonyl] -2-methoxyphenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-320)
Scheme:
Step A: tert-butyl 9- ( (4- ( (benzyloxy) carbonyl) piperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carboxylate
A mixture of benzyl piperazine-1-carboxylate (250 mg, 1.14 mmol) , S tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (479 mg, 1.71 mmol) and NaBH (OAc) 3 (723 mg, 3.42 mmol) in DCM (5.0 mL) and MeOH (5.0 mL) and HOAc (68 mg, 0.473 mmol) was stirred at 35 ℃ for 16 hrs . After completion of reaction, quenched with water (50 mL) , extracted with EtOAc (3*50 mL) . The organic layers were washed with brine (15.0 mL) , dried with Na2SO4,
filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =1: 1) to afford the desired product.
LCMS: MS (ESI) m/z 486.3 [M+H] +.
Step B: tert-butyl 9- (piperazin-1-ylmethyl) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9- ( (4- ( (benzyloxy) carbonyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate (270 mg 0.557 mmol) in MeOH (15 mL) was added Pd/C (10%, 30 mg ) and stirred at 35℃ under H2 protected reaction for 3 h. The reaction solution is filtered under diatomite, and then the filtrate is suspended dry to afford the desired product.
LCMS: MS (ESI) m/z 352.3 [M+H] +.
Step C: tert-butyl 9- ( (4- (3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) -4-
methoxybenzoyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9- (piperazin-1-ylmethyl) -3-azaspiro [5.5] undecane-3-carboxylate (190 mg, 0.541 mmol) in DMF (5 mL) was added perfluorophenyl 3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) -4-methoxybenzoate (232 mg, 0.541 mmol) , and DIEA (140 mg, 1.08 mmol) . Then the reaction was stirred at 35 ℃ for 16 hrs under N2 protection. After completion of reaction, quenched with water (50 mL) , extracted with EtOAc (3*50 mL) . The organic layers were washed with brine (50 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 598.3 [M+H] +.
Step D: 1- (5- (4- ( (3-azaspiro [5.5] undecan-9-yl) methyl) piperazine-1-carbonyl) -2-
methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of tert-butyl 9- ( (4- (3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) -4-methoxybenzoyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate (210 mg, 0.352 mmol) in DCM (5.0 mL) was added HCl in dioxane (4M, 3.0 mL) and stirred at 35℃ for 1 h. The mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 498.5 [M+H] +.
Step E: 1- (2-methoxy-5- (4- ( (3- (4-nitrophenyl) -3-azaspiro [5.5] undecan-9-
yl) methyl) piperazine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (5- (4- ( (3-azaspiro [5.5] undecan-9-yl) methyl) piperazine-1-carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (184 mg, 0.370 mmol) in DMSO (3.0 mL) was added 1-fluoro-4-nitrobenzene (52 mg, 0.370 mmol) , DIEA (143 mg, 1.11 mmol) was stirred 120 ℃ for 3 hrs under N2 atmosphere. After completion of reaction, quenched with water (100 mL) , extracted with EtOAc (3*100 mL) . The organic layers were washed with brine (50
mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 619.3 [M+H] +.
Step F: 1- (5- (4- ( (3- (4-aminophenyl) -3-azaspiro [5.5] undecan-9-yl) methyl) piperazine-1-
carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
A mixture of 1- (2-methoxy-5- (4- ( (3- (4-nitrophenyl) -3-azaspiro [5.5] undecan-9-yl) methyl) piperazine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (170 mg, 0.275 mmol) , iron powder (30.7 mg, 0.550 mmol) , NH4Cl (29.4 mg, 0.550 mmol) in EtOH (10 mL) and H2O (2 mL) was stirred at 90 ℃ for 3 hrs. After completion of reaction, the mixture reaction was filtered through a Celite pad, and the filtrate was concentrated to give the crude. The crude was quenched with H2O (10 mL) , adjusted to pH 8 with solid NaHCO3 and extracted with EtOAc (3 x 30 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 589.4 [M+H] +.
Step G: 1- {5- [ (4- { [3- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3-aza-9-spiro [5.5] undecyl] methyl} -1-piperazinyl) carbonyl] -2-
methoxyphenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a solution of (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (30.0 mg, 0.0778 mmol) in DCM (8 mL) was added 1- (5- (4- ( (3- (4-aminophenyl) -3-azaspiro [5.5] undecan-9-yl) methyl) piperazine-1-carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (137 mg, 0.238 mmol) . Then the reaction mixture was stirred at 35 ℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EA (20 mL x3) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated. The residue was purified by prep-HPLC (Waters 2767, Column: Pursuit XRs C18, 21.2*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 15%~25%Retention Time: 9.5-10.5 min of 16 min) to give the product (11.58 mg, 48.9%yield) as a light yellow solid.
LCMS: MS (ESI) m/z 910.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.34 (s, 1H) , 10.03 (s, 1H) , 8.81 (s, 1H) , 8.07 (d, J = 47.4 Hz, 1H) , 7.76 (d, J = 8.1 Hz, 1H) , 7.56 (dd, J = 13.8, 8.0 Hz, 3H) , 7.36 (dd, J = 15.7, 5.2 Hz, 2H) , 7.16 (d, J = 8.6 Hz, 1H) , 6.89 (d, J = 9.1 Hz, 2H) , 5.52 –5.29 (m, 2H) , 5.26 (s, 1H) , 4.45 (q, J = 6.3 Hz, 2H) , 3.84 (m, 3H) , 3.61 (d, J = 6.7 Hz, 2H) , 3.54 –3.42 (m, 3H) , 3.31 –3.27 (m,
2H) , 3.08 (m, 4H) , 2.77 –2.65 (m, 3H) , 2.41 –2.28 (m, 4H) , 2.25 –2.09 (m, 3H) , 2.06 –1.90 (m, 2H) , 1.75 –1.63 (m, 3H) , 1.62 –1.52 (m, 7H) , 1.47 –1.40 (m, 2H) , 1.07 (m, 4H) .
Example 111:
1- {5- [ (4- { [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) carbonyl] -2-methoxyphenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-160)
Scheme:
Step A: 1- {5- [ (4- { [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) carbonyl] -2-
methoxyphenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a solution of 1- (5- (4- ( (9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (30.0 mg,0.052 mmol) in DCM (10 mL) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (30.0 mg, 0.052 mmol) , the mixture was stirred at 30 ℃ for 16 hrs under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the
resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767, Column: Pursuit XRs C18, 21.2*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 15%~25%Retention Time: 8.5-9.5 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 910.8 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.33 (s, 1H) , 10.02 (s, 1H) , 8.82 (s, 1H) , 8.00 (s, 1H) , 7.76 (s, 1H) , 7.57 (m, 3H) , 7.34 (d, J = 16.9 Hz, 2H) , 7.18 (s, 1H) , 6.93 (m, 2H) , 5.54 –5.17 (m, 3H) , 4.57 –4.35 (m, 2H) , 3.85 (s, 3H) , 3.64 –3.55 (m, 2H) , 3.18 –3.01 (m, 7H) , 3.00 –2.74 (m, 5H) , 2.72 –2.63 (m, 3H) , 2.28 –2.10 (m, 2H) , 2.08 –1.92 (m, 3H) , 1.82 –1.69 (m, 4H) , 1.68 –1.46 (m, 9H) , 1.18 (m, 3H) .
Example 112:
1- {5- [ (4- { [9- (p- { (R, 12E) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) carbonyl] -2-chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-162)
Scheme:
Step A: 1- {5- [ (4- { [9- (p- { (R, 12E) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) carbonyl] -2-
chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a solution of (12R, E) -9-fluoro-12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (30.0 mg, 0.074 mmol) in DCM (2 mL) was added 1- (5- (4- ( (9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4(1H, 3H) -dione (43.9 mg, 0.074 mmol) , the mixture was stirred at 30 ℃ for 16 hrs under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by Pre-HPLC (Waters 2767, Column: Pursuit XRs C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 13%~20%Retention Time: 8.6-10.4 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 932.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.51 (s, 1H) , 10.11 (s, 1H) , 8.83 (s, 1H) , 8.21 (s, 1H) , 8.03 (s, 1H) , 7.82 (d, J = 8.1 Hz, 1H) , 7.64 (d, J = 8.2 Hz, 1H) , 7.59 (d, J = 7.7 Hz, 1H) , 7.54 (d, J =2.0 Hz, 3H) , 7.38 (dd, J = 8.3, 2.0 Hz, 1H) , 6.90 (d, J = 9.1 Hz, 2H) , 5.03 (d, J = 7.7 Hz, 1H) , 4.63 (d, J = 21.1 Hz, 2H) , 4.49 –4.33 (m, 1H) , 3.75 (dd, J = 13.2, 6.9 Hz, 1H) , 3.64 –3.53 (m, 3H) , 3.10 –3.05 (m, 4H) , 2.75 (m, 3H) , 2.34 (m, 4H) , 2.20 –2.08 (m, 3H) , 1.97 –1.87 (m, 2H) , 1.84 –1.72 (m, 2H) , 1.70 –1.58 (m, 3H) , 1.56 (m, 3H) , 1.53 (m, 4H) , 1.49 –1.41 (m, 4H) , 1.13 –1.00 (m, 2H) .
Example 113:
3- (6- (2- (4- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2- yl) amino) phenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2- yl) piperidine-2, 6-dione (isomer 1) (Cpd-321, isomer 1)
Scheme:
Step A: tert-butyl 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane-8-carboxylate
To a mixture of 1- (4-nitrophenyl) piperazine (9.82 g, 47.37 mmol) and tert-butyl 3-oxo-8-azaspiro [4.5] decane-8-carboxylate (8 g, 31.58 mmol) in DCM (100 mL) was added AcOH (1.90 g, 31.58 mmol) and NaBH (OAc) 3 (10.04 g, 47.37 mmol) . The mixture was stirred at 25 ℃ for 16 hours. The reaction mixture was added H2O (300 mL) and extracted with DCM (150 mL ×3) . The combined organic phases were dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The crude product was triturated with EtOAc (10 mL) and Petroleum ether (30 mL) at 20 ℃ for 2 h to afford the desired product.
LCMS: MS (ESI) m/z 445.3 [M+H] +.
Step B: tert-butyl 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane-8-
carboxylat (isomer 1 &isomer 2)
The crude product was purified by prep-SFC (column: DAICEL CHIRALPAK AD (250mm*50mm, 10um) ; mobile phase: [CO2-MeOH (0.1%NH3H2O) ] ; B%: 50%, isocratic elution mode) to afford the desired product, then dried by lyophilization to give tert-butyl 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane-8-carboxylate (isomer 1) &tert-butyl 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane-8-carboxylate (isomer 2)
tert-butyl 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane-8-carboxylate (Peak 1) (isomer 2)
LCMS: MS (ESI) m/z 445.3 [M+H] +.
SFC: tR = 1.613 min (AD_MEOH_DEA_40_4ML_5 CM) , ee = 95.74%
tert-butyl 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane-8-carboxylate (Peak 2) (isomer 1)
LCMS: MS (ESI) m/z 445.3 [M+H] +.
SFC: tR = 1.248 min (AD_MEOH_DEA_40_4ML_5 CM) , ee = 100.00%
Step C: 2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane
To a solution of tert-butyl 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane-8-carboxylate (isomer 1) (4.64 g, 10.44 mmol) in HCl/dioxane (4 M, 40.00 mL) was stirred at 20 ℃ for 16 h. The mixture was added H2O (200 mL) and adjustment of pH to 8 with NaHCO3. The aqueous phase was extracted with 5 portions of DCM/MeOH (10: 1, 100 mL) . The combined organic phases were dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 345.3 [M+H] +.
Step D: 2- (2, 6-dioxopiperidin-3-yl) -5- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-
azaspiro [4.5] decan-8-yl) isoindoline-1, 3-dione
To a mixture of 2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane (2 g, 5.81 mmol) and 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (1.76 g, 6.39 mmol) in DMF (25 mL)
was added DIPEA (1.50 g, 11.61 mmol, 2.02 mL) . The mixture was stirred at 90 ℃ for 4 hours. The reaction mixture was added H2O (100 mL) and extracted with ethyl acetate (50 mL × 3) . The combined organic phases were washed with brine (100 mL × 5) , dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, DCM/MeOH = 1/0 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 601.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.08 (s, 1H) , 8.05 (d, J = 9.6 Hz, 2H) , 7.65 (d, J = 8.4 Hz, 1H) , 7.31 (s, 1H) , 7.27 -7.20 (m, 1H) , 7.02 (d, J = 9.6 Hz, 2H) , 5.06 (dd, J = 5.2, 12.8 Hz, 1H) , 3.50-3.46 (m, 4H) , 3.44-3.40 (m, 4H) , 3.31 (m, 2H) , 3.17 (d, J = 5.2 Hz, 2H) , 2.89 -2.82 (m, 1H) , 2.68 -2.58 (m, 2H) , 2.55-2.53 (m, 1H) , 2.06 -1.94 (m, 1H) , 1.93 -1.77 (m, 2H) , 1.61 -1.44 (m, 7H) , 1.38 -1.28 (m, 1H) .
Step E: 3- (6- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a mixture of 2- (2, 6-dioxopiperidin-3-yl) -5- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) isoindoline-1, 3-dione (1.92 g, 3.20 mmol) in TFA (25 mL) was added Zn (4.4 g, 67.29 mmol) . The mixture was stirred at 75 ℃ for 1 hours. The reaction mixture was filtered. The residue washed with EtOAc (100 mL) , concentrated under reduced pressure to give crude product. The crude product was purified by reversed-phase column (column: Welch Xtimate C18 100*40mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 0%-23%B over 8 min) to afford the desired product.
LCMS: MS (ESI) m/z 557.1 [M+H] +.
2D NMR (400 MHz, DMSO-d6) showed H16 has HMBC correlation to C11, which is consistent with the determined structure.
1H NMR (400 MHz, DMSO-d6) δ 10.97 (s, 1H) , 7.44 (d, J = 8.4 Hz, 1H) , 7.30 (d, J = 6.4 Hz, 1H) , 7.24 (d, J = 8.0 Hz, 3H) , 7.10 (d, J = 8.8 Hz, 2H) , 5.09 (dd, J = 4.8, 13.2 Hz, 1H) , 4.41 -4.16 (m, 2H) , 3.86 (d, J = 11.2 Hz, 2H) , 3.74 (d, J = 8.3 Hz, 1H) , 3.61 (m, 2H) , 3.36 -3.21 (m, 2H) , 3.17-3.12 (m, 2H) , 3.05 -2.93 (m, 2H) , 2.93 -2.84 (m, 1H) , 2.62-2.58 (m, 1H) , 2.56-2.53 (m, 2H) , 2.44 -2.35 (m, 1H) , 2.19 -2.05 (m, 2H) , 2.04 -1.94 (m, 1H) , 1.92-1.85 (m, 1H) , 1.76 -1.48 (m, 7H) .
Step F: 3- (6- (2- (4- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17-
(azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-
yl) amino) phenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-
yl) piperidine-2, 6-dione (isomer 1)
To a solution of 3- (6- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (70 mg, 181.61 μmol) in n-BuOH (2 mL) was added
AcOH (109.06 mg, 1.82 mmol) . The mixture was stirred 100 ℃ for 16 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: Welch Xtimate C18 100*40mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 6%-36%B over 8 min) to afford the desired product, then dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 878.7 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.97 (s, 1H) , 10.10 (TFA, 0.809H) , 9.54 (s, 1H) , 8.84 (s, 1H) , 8.00 (t, J = 7.6 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.61 (dd, J = 8.4, 13.2 Hz, 3H) , 7.44 (d, J = 8.4 Hz, 1H) , 7.30 (d, J = 7.6 Hz, 1H) , 7.23 (s, 1H) , 7.00 (d, J = 8.8 Hz, 2H) , 5.27 -5.41 (m, 3H) , 5.09 (dd, J = 4.8, 13.6 Hz, 1H) , 4.53 -4.42 (m, 2H) , 4.18-4.38 (m, 2H) , 3.86 -3.75 (m, 2H) , 3.74 -3.72 (m, 1H) , 3.60 (m, 2H) , 3.36 -3.21 (m, 4H) , 3.21 -3.12 (m, 2H) , 2.99 -2.92 (m, 2H) , 2.91 -2.85 (m, 1H) , 2.62 -2.58 (m, 1H) , 2.45 -2.34 (m, 1H) , 2.25 -2.08 (m, 2H) , 2.05 -1.90 (m, 3H) , 1.90 -1.82 (m, 1H) , 1.77 -1.51 (m, 12H) .
Example 114:
3- (6- (2- (4- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (isomer 2) (Cpd-321, isomer 2)
Scheme:
Step A: 3- (6- (2- (4- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17-
(azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-
yl) amino) phenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-
yl) piperidine-2, 6-dione
A mixture of 3- (6- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (peak 1) (50 mg, 74.55 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (35 mg, 87.19 μmol) in n-BuOH (1 mL) was stirred at 100 ℃ for 1 hr. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: Welch Xtimate C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 0%-40%B over 9 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 878.7 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.98 (s, 1H) , 10.07 (br s, 1H) , 8.81 (s, 1H) , 8.19 (s, 1H) , 8.01 (br s, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.57 (br t, J = 6.8 Hz, 3H) , 7.40 (br d, J = 8.4 Hz, 1H) , 7.24 (br d, J = 7.6 Hz, 1H) , 7.16 (s, 1H) , 6.89 (br d, J = 8.8 Hz, 2H) , 5.34 (br s, 2H) , 5.09 (br dd, J = 5.2, 13.3 Hz, 1H) , 4.45 (br s, 2H) , 4.32 (d, J = 16.8 Hz, 1H) , 4.19 (d, J = 16.8 Hz, 1H) , 3.19 (br s, 5H) , 3.08 (br s, 4H) , 2.98 -2.83 (m, 1H) , 2.69 -2.52 (m, 6H) , 2.44 -2.31 (m, 1H) , 2.17 (br s, 1H) , 2.08 -1.77 (m, 5H) , 1.77 -1.67 (br s, 1H) , 1.66 -1.42 (m, 10H) , 1.38 -1.23 (m, 1H)
Example 115:
3- (6- (2- (4- (4- ( ( (R, E) -8-fluoro-12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (isomer 1) (Cpd-323, isomer 1)
Scheme:
Step A: (12E, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one
To a mixture of (12E, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfanyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (150 mg, 387.17 μmol) in THF (2 mL) and H2O (2 mL) was added Oxone (238.02 mg, 387.17 μmol) at 0 ℃. The mixture was stirred at 0 ℃ for 2 hours. The reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (30 mL × 7) . The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 404.2 [M+H] +.
Step B: 3- (6- (2- (4- (4- ( ( (R, E) -8-fluoro-12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-
5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-
yl) amino) phenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-
yl) piperidine-2, 6-dione (isomer 1)
To a solution of 3- (6- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (99.75 mg, 148.72 μmol, TFA salt) , (12E, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (50 mg, 123.94 μmol) in n-BuOH (2 mL) was AcOH (74.42 mg, 1.24 mmol) . The mixture was stirred at 100 ℃ for 16 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: Welch Xtimate C18 100*40mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 6%-36%B over 8 min) to afford the desired product.
LCMS: MS (ESI) m/z 896.8 [M+H] +.
19F NMR (400 MHz, DMSO-d6) δ = -74.392 (s, 1H) ,
1H NMR (400 MHz, DMSO-d6) δ 10.97 (s, 1H) , 10.16 (TFA, 0.69H) , 9.55 (s, 1H) , 8.86 (s, 1H) , 8.01 (t, J = 7.6 Hz, 1H) , 7.81 (d, J = 8.0 Hz, 1H) , 7.62 (t, J = 7.2 Hz, 3H) , 7.44 (d, J = 8.4 Hz, 1H) , 7.29 (d, J = 8.4 Hz, 1H) , 7.22 (s, 1H) , 7.00 (d, J = 9.2 Hz, 2H) , 5.09 (dd, J = 4.8, 13.6 Hz, 1H) , 5.02-4.98 (m, 1H) , 4.70 -4.60 (m, 2H) , 4.38 -4.17 (m, 2H) , 3.85 -3.69 (m, 4H) , 3.34 -3.21 (m, 4H) , 3.19-3.15 (m, 2H) , 2.99 -2.91 (m, 2H) , 2.90 -2.84 (m, 1H) , 2.61-2.57 (m, 1H) , 2.46 -2.35 (m, 1H) , 2.21 -2.05 (m, 3H) , 2.03 -1.95 (m, 1H) , 1.95 -1.79 (m, 3H) , 1.77 -1.45 (m, 12H) .
Example 116:
3- (6- (2- (4- (4- ( ( (R, E) -8-fluoro-12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (isomer 2) (Cpd-323, isomer 2)
Scheme:
Step A: 3- (6- (2- (4- (4- ( ( (R, E) -8-fluoro-12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-
5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-
yl) amino) phenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-
yl) piperidine-2, 6-dione (isomer 2)
A mixture of 3- (6- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (50 mg, 89.82 μmol) and (12E, 16R) -12-fluoro-16-
hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (35 mg, 83.45 μmol) in n-BuOH (1 mL) was stirred at 100 ℃ for 1 hr. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: Welch Xtimate C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 0%-40%B over 9 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 896.5 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.98 (s, 1H) , 10.15 (br s, 1H) , 8.84 (s, 1H) , 8.15 (s, 1H) , 8.02 (br s, 1H) , 7.81 (br d, J = 7.6 Hz, 1H) , 7.58 (br t, J = 8.0 Hz, 3H) , 7.40 (br d, J = 8.4 Hz, 1H) , 7.25 (br d, J = 8.4 Hz, 1H) , 7.16 (br s, 1H) , 6.92 (br d, J = 8.4 Hz, 2H) , 5.33 (br s, 1H) , 5.18 -4.97 (m, 2H) , 4.73 -4.52 (d, J = 21.6 Hz, 2H) , 4.32 (d, J = 16.8 Hz, 1H) , 4.19 (d, J = 16.8 Hz, 1H) , 3.28 -3.07 (m, 8H) , 2.99 -2.77 (m, 3H) , 2.71 (br s, 4H) , 2.59 (br d, J = 17.6 Hz, 1H) , 2.45 -2.29 (m, 1H) , 2.18 -2.05 (m, 1H) , 2.04 -1.80 (m, 5H) , 1.73 -1.45 (m, 11H) , 1.43 -1.33 (br d, J = 11.2 Hz, 1H)
Example 117:
3- (5- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-325)
Scheme:
Step A: tert-butyl 2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane-8-carboxylate
A solution of tert-butyl 2-oxo-8-azaspiro [4.5] decane-8-carboxylate (4.00 g, 15.8 mmol) , 1- (4-nitrophenyl) piperazine (3.27 g, 15.8 mmol) , NaBH (OAc) 3 (10.0 g, 47.4 mmol) , AcOH (0.5 mL) in DCM (20 mL) and MeOH (2 mL) was stirred at 25℃ for 16 hrs. After completion, the mixture was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 445.2 [M+H] +.
Step B: 2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane
To a solution of tert-butyl 2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane-8-carboxylate (685 mg, 1.54 mmol) in DCM (3.0 mL) was added HCl. dioxane (1.0 mL, 4 M) and stirred at 20℃ for 2 hrs. After completion of reaction, the reaction mixture was concentrated under reduced pressure to dryness to afford the desired product.
LCMS: MS (ESI) m/z 345.1 [M+H] +.
Step C: 3- (5- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
A mixture of 2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane (300 mg, 0.871 mmol) , 3- (5-bromo-1-oxoisoindolin-2-yl) piperidine-2, 6-dione (281 mg, 0.871 mmol) , Pd-PEPPSI-IPentCl (42.4 mg, 0.0436 mmol) , Cs2CO3 (851 mg, 2.61 mmol) in dioxane (5.0 mL) was stirred at 100℃ for 16 hrs under N2. After completion, the mixture was quenched with H2O (10 mL) , extracted with DCM (10 mLx3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 587.1 [M+H] +.
Step D: 3- (5- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
A mixture of 3- (5- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (200 mg, 0.341 mmol) , iron powder (95.5 mg, 1.71 mmol) , NH4Cl (91.5 mg, 1.71 mmol) in EtOH (10 mL) and H2O (2 mL) was stirred at 90℃ for 3 hrs. After completion of reaction, the mixture reaction was filtered through a Celite pad, and the filtrate was concentrated to give the crude. The crude was quenched with H2O (10 mL) , adjusted to pH 8 with solid NaHCO3 and extracted with EtOAc (3 x 30 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 557.2 [M+H] +.
Step E: 3- (5- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20.0 mg, 0.0519 mmol) in DCM (3 mL) was added 3- (5- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (75.0 mg, 0.135 mmol) and t-BuOH (12 mL) . Then the reaction mixture was stirred at 100℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EA (20 mL x3) . The organic layers were concentrated. The residue was purified by prep-HPLC (Waters 2767, Column: Pursuit XRs C18, 21.2*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 13%~23%Retention Time: 9.3-10.8 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 878.2 [M+H] +.
1HNMR (400 MHz, DMSO-d6) δ 10.93 (s, 1H) , 10.04 (s, 1H) , 8.81 (s, 1H) , 8.31 (s, 1H) , 8.01 (s, 1H) , 7.75 (d, J = 6.4 Hz, 1H) , 7.57 (m, 3H) , 7.49 (d, J = 8.8 Hz, 1H) , 7.06 (s, 2H) , 6.90 (d, J = 7.8 Hz, 2H) , 5.32 (dd, J = 30.9, 16.2 Hz, 3H) , 5.04 (t, J = 12.2 Hz, 1H) , 4.55 –4.38 (m, 2H) , 4.31 (d, J = 17.5 Hz, 1H) , 4.19 (d, J = 17.7 Hz, 1H) , 3.08 (m, 4H) , 2.94 –2.83 (m, 3H) , 2.68 –2.65 (m, 2H) , 2.60 (dd, J = 6.5, 3.7 Hz, 2H) , 2.53 (m, 4H) , 2.32 (dd, J = 6.6, 2.7 Hz, 2H) , 2.22 –2.12 (m, 2H) , 2.08 –2.00 (m, 2H) , 1.95 (dd, J = 9.6, 4.5 Hz, 2H) , 1.89 –1.83 (m, 2H) , 1.71 (dd, J = 10.8, 5.1 Hz, 1H) , 1.61 (dd, J = 10.1, 7.8 Hz, 2H) , 1.56 (m, 3H) , 1.50 (d, J = 5.6 Hz, 2H) , 1.32 (t, J = 13.8 Hz, 2H) .
Example 118:
3- (5- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-326)
Scheme:
Step A: tert-butyl 8- (2- (2, 6-dioxopiperidin-3-yl) -1-oxoisoindolin-5-yl) -2, 8-
diazaspiro [4.5] decane-2-carboxylate
To a solution of 3- (5-bromo-1-oxoisoindolin-2-yl) piperidine-2, 6-dione (1 g, 3.09 mmol) in dioxane (15.0 mL) was added tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (817 mg, 3.09 mmol) , t-BuONa (892 mg, 9.27 mmol) , Ruphos-Pd-G2 (482 mg, 0.618 mmol) , was stirred 100 ℃ for 16 h under N2 atmosphere. After completion of reaction, quenched with water (100 mL) , extracted with EtOAc (3*100 mL) . The organic layers were washed with brine (50 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 483.2 [M+H] +.
Step B: 3- (1-oxo-5- (2, 8-diazaspiro [4.5] decan-8-yl) isoindolin-2-yl) piperidine-2, 6-dione
To a solution of tert-butyl 8- (2- (2, 6-dioxopiperidin-3-yl) -1-oxoisoindolin-5-yl) -2, 8-diazaspiro [4.5] decane-2-carboxylate (76 mg, 0.158 mmol) in DCM (5.0 mL) was added HCl in dioxane (4M, 3.0 mL) and stirred at 35 ℃ for 1 h. The mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 383.2 [M+H] +.
Step C: 3- (5- (2- (1- (4-nitrophenyl) piperidin-4-yl) -2, 8-diazaspiro [4.5] decan-8-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
A mixture of 3- (1-oxo-5- (2, 8-diazaspiro [4.5] decan-8-yl) isoindolin-2-yl) piperidine-2, 6-dione (50 mg, 0.131 mmol) , 1- (4-nitrophenyl) piperidin-4-one (29 mg, 0.131 mmol) and NaBH (OAc) 3 (83 mg, 0.393 mmol) in DCM (4.0 mL) and MeOH (4.0 mL) and HOAc (0.5 mL) was stirred at 35 ℃ for 16 h . After completion of reaction, quenched with water (50 mL) , extracted with EtOAc (3*50 mL) . The organic layers were washed with brine (15.0 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 587.0 [M+H] +.
Step D: 3- [5- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-
isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 3- (5- (2- (1- (4-nitrophenyl) piperidin-4-yl) -2, 8-diazaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (50 mg, 0.0853 mmol) in EtOH (10 mL) and H2O (3 mL) was added NH4Cl (23 mg 0.427 mmol) , Fe (24 mg 0.427 mmol) . The mixture was stirred at 90 ℃ for 2 h. After completion of reaction, the reaction was filtered. The filtrate was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 557.3 [M+H] +.
Step E: 3- (5- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 3- [5- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (240 mg, 357.82 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (100 mg, 259.44 μmol) in n-BuOH (1 mL) was added AcOH (15.58 mg, 259.44 μmol) . The mixture was stirred at 100 ℃ for 1 hr. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: Xtimate C18 150*40mm*10um; mobile phase: [water (FA) -ACN] ; gradient: 6%-36%B over 7 min) and dried by lyophilization to afford the desired product.
LC-MS (ESI) [M+H] + 878.4.
1H NMR (400 MHz, DMSO-d6) δ 10.94 (s, 1H) , 10.04 (br s, 1H) , 8.81 (s, 1H) , 8.19 (s, 1H) , 8.01 (br t, J = 7.6 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.62 -7.52 (m, 3H) , 7.50 (d, J = 8.4 Hz, 1H) , 7.09 -7.01 (m, 2H) , 6.91 (br d, J = 8.8 Hz, 2H) , 5.45 -5.25 (m, 2H) , 5.04 (dd, J = 4.8, 13.2 Hz, 1H) , 4.45 (br d, J = 5.6 Hz, 2H) , 4.31 (d, J = 16.8 Hz, 1H) , 4.19 (d, J = 16.8 Hz, 1H) , 3.38 -
3.34 (m, 3H) , 3.32 -3.23 (m, 3H) , 2.95 -2.84 (m, 1H) , 2.82 -2.53 (m, 8H) , 2.43 -2.26 (m, 2H) , 2.17 (br s, 1H) , 2.08 -1.86 (m, 5H) , 1.75 -1.48 (m, 12H)
Example 119:
3- (5- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -6-fluoro-1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-327)
Scheme:
Step A: tert-butyl 8- (2- (2, 6-dioxopiperidin-3-yl) -6-fluoro-1-oxoisoindolin-5-yl) -2, 8-
diazaspiro [4.5] decane-2-carboxylate
A mixture of 3- (5-bromo-6-fluoro-1-oxoisoindolin-2-yl) piperidine-2, 6-dione (300 mg, 0.879 mmol) , tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (420 mg, 1.76 mmol) , Pd-PEPPSI-IPentCl (44.1 mg, 0.0440 mmol) , Cs2CO3 (861 mg, 2.64 mmol) in dioxane (5.0 mL) was stirred at 100℃ for 16 hrs under N2. After completion, the mixture was quenched with H2O (10 mL) , extracted with DCM (10 mLx3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 501.1 [M+H] +.
Step B: 3- (6-fluoro-1-oxo-5- (2, 8-diazaspiro [4.5] decan-8-yl) isoindolin-2-yl) piperidine-2, 6-
dione
To a solution of tert-butyl 8- (2- (2, 6-dioxopiperidin-3-yl) -6-fluoro-1-oxoisoindolin-5-yl) -2, 8-diazaspiro [4.5] decane-2-carboxylate (226 mg, 0.451 mmol) in DCM (3.0 mL) was added HCl. dioxane (1.0 mL, 4 M) and stirred at 25℃ for 2 hr. After completion of reaction, the reaction mixture was concentrated under reduced pressure to dryness to afford the desired product.
LCMS: MS (ESI) m/z 401.1 [M+H] +.
Step C: 3- (6-fluoro-5- (2- (1- (4-nitrophenyl) piperidin-4-yl) -2, 8-diazaspiro [4.5] decan-8-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
A solution of 3- (6-fluoro-1-oxo-5- (2, 8-diazaspiro [4.5] decan-8-yl) isoindolin-2-yl) piperidine-2, 6-dione (190 mg, 0.435 mmol) , 1- (4-nitrophenyl) piperidin-4-one (95.8 mg, 0.435 mmol) , AcOH (0.2 mL) in DCM (10 mL) was stirred at 25℃ for 0.5 h. Then NaBH (OAc) 3 (30.5 mg, 0.144 mmol) was added. The mixture was stirred at 25℃ for 2 hrs. After completion, the mixture was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 605.4 [M+H] +.
Step D: 3- (5- (2- (1- (4-aminophenyl) piperidin-4-yl) -2, 8-diazaspiro [4.5] decan-8-yl) -6-fluoro-
1-oxoisoindolin-2-yl) piperidine-2, 6-dione
A mixture of 3- (6-fluoro-5- (2- (1- (4-nitrophenyl) piperidin-4-yl) -2, 8-diazaspiro [4.5] decan-8-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (130 mg, 0.215 mmol) , iron powder (60.3 mg, 1.08 mmol) , NH4Cl (57.8 mg, 1.08 mmol) in EtOH (10 mL) and H2O (2 mL) was stirred at 90℃ for 3 hrs. After completion of reaction, the mixture reaction was filtered through a Celite pad, and the filtrate was concentrated to give the crude. The crude was quenched with H2O (10 mL) , adjusted to pH 8 with solid NaHCO3 and extracted with EtOAc (3 x 30 mL) . The organic layers
were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 574.5 [M+H] +.
Step E: 3- (5- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -6-fluoro-1-oxo-2-isoindolinyl) -
2, 6-piperidinedione
To a solution of (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20.0 mg, 0.0519 mmol) in DCM (8 mL) was added 3- (5- (2- (1- (4-aminophenyl) piperidin-4-yl) -2, 8-diazaspiro [4.5] decan-8-yl) -6-fluoro-1-oxoisoindolin-2-yl) piperidine-2, 6-dione (50.0 mg, 0.0870 mmol) . Then the reaction mixture was stirred at 35℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EA (20 mL x3) . The organic layers were concentrated and purified by prep-TLC to afford the crude. The crude was purified by prep-HPLC (Waters 2767, Column: Pursuit XRs C18, 21.2*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 15%~25%Retention Time: 9.5-10.5 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 896.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.96 (s, 1H) , 10.08 –9.92 (m, 1H) , 8.81 (s, 1H) , 8.33 (m, 3H) , 8.01 (s, 1H) , 7.76 (d, J = 7.8 Hz, 1H) , 7.56 (d, J = 11.7 Hz, 3H) , 7.40 (d, J = 11.4 Hz, 1H) , 7.24 (d, J = 8.3 Hz, 1H) , 6.91 (d, J = 8.9 Hz, 2H) , 5.45 –5.16 (m, 3H) , 5.09 (s, 1H) , 4.44 (dd, J = 10.3, 5.9 Hz, 2H) , 4.35 (d, J = 17.3 Hz, 1H) , 4.23 (d, J = 18.0 Hz, 1H) , 3.57 (d, J = 7.1 Hz, 2H) , 3.11 –3.06 (m, 4H) , 2.96 –2.79 (m, 2H) , 2.74 –2.70 (m, 1H) , 2.65 –2.60 (m, 2H) , 2.41 –2.35 (m, 2H) , 2.16 (ddd, J = 9.4, 8.6, 5.1 Hz, 3H) , 1.98 (dd, J = 6.7, 3.2 Hz, 2H) , 1.91 (dd, J =7.4, 1.4 Hz, 2H) , 1.73 –1.68 (m, 2H) , 1.66 –1.61 (m, 4H) , 1.56 (m, 3H) , 1.48 (dd, J = 11.9, 5.3 Hz, 2H) , 1.25 –1.22 (m, 3H) .
Example 120:
3- (6- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperidyl] -8-aza-8-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-328)
Scheme:
Step A: tert-butyl 3- [4- (4-nitrophenyl) -1-piperidyl] -8-azaspiro [4.5] decane-8-carboxylate
To a solution of tert-butyl 3-oxo-8-azaspiro [4.5] decane-8-carboxylate (1.23 g, 4.85 mmol) , 4- (4-nitrophenyl) piperidine (1 g, 4.85 mmol) in EtOH (3 mL) was added dropwise AcOH (145.59 mg, 2.42 mmol) . After addition, the mixture was stirred at 50 ℃ for 1 hr, then NaBH3CN (914.11 mg, 14.55 mmol) was added. The resulting mixture was stirred at 50 ℃ for 2 hr. The mixture was poured into water (100 mL) and extracted with EtOAc (100 mL × 3) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by silica gel column chromatography (4%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 444.3 [M+H] +.
Step B: 3- [4- (4-nitrophenyl) -1-piperidyl] -8-azaspiro [4.5] decane
To a solution of tert-butyl 3- [4- (4-nitrophenyl) -1-piperidyl] -8-azaspiro [4.5] decane-8-carboxylate (2.1 g, 4.73 mmol) in DCM (15 mL) was added TFA (7.68 g, 67.31 mmol, 5 mL) . The mixture was stirred at 20 ℃ for 2hr. The reaction mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 344.3 [M+H] +.
Step C: 2- (2, 6-dioxo-3-piperidyl) -5- [3- [4- (4-nitrophenyl) -1-piperidyl] -8-
azaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione
To a solution of 3- [4- (4-nitrophenyl) -1-piperidyl] -8-azaspiro [4.5] decane (800 mg, 2.33 mmol) , 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (643.38 mg, 2.33 mmol) in DMF (8 mL) was added DIEA (903.09 mg, 6.99 mmol) . The mixture was stirred at 100 ℃ for 2 hr. The mixture was concentrated to give a residue, which was purified by silica gel column chromatography (6%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 600.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.08 (s, 1H) , 8.17 (d, J = 8.4 Hz, 2H) , 7.65 (d, J = 8.8 Hz, 1H) , 7.55 (d, J = 8.8 Hz, 2H) , 7.32 (s, 1H) , 7.24 (dd, J = 2.0, 8.8 Hz, 1H) , 5.06 (dd, J = 5.6, 12.8 Hz, 1H) , 3.48 (s, 4H) , 3.21 -2.97 (m, 2H) , 2.97 -2.79 (m, 2H) , 2.66 -2.54 (m, 2H) , 2.19 -1.96 (m, 3H) , 1.92 -1.69 (m, 5H) , 1.68 -1.24 (m, 10H) .
Step D: 3- [6- [3- [4- (4-aminophenyl) -1-piperidyl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-
isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [3- [4- (4-nitrophenyl) -1-piperidyl] -8-azaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione (800 mg, 1.33 mmol) in TFA (8 mL) was added Zn (3.51 g, 53.68 mmol) . The mixture was stirred at 75 ℃ for 2 hr. The reaction mixture was filtered, washed with DCM (300mL) and concentrated to give crude product, which was purified by prep-HPLC (column: Welch Xtimate C18 100*40mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 0%-24%B over 6.5 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 556.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.98 (s, 1H) , 9.72 (s, 1H) , 7.45 (d, J = 8.4 Hz, 1H) , 7.36 -7.31 (m, 1H) , 7.30 (s, 1H) , 7.28 (s, 1H) , 7.27 (s, 1H) , 7.22 (s, 1H) , 7.20 (s, 1H) , 5.09 (dd, J = 5.2, 13.2 Hz, 1H) , 4.41 -4.18 (m, 2H) , 3.61 (d, J = 10.8 Hz, 2H) , 3.38 -3.16 (m, 5H) , 3.03 (m, 2H) , 2.97 -2.80 (m, 2H) , 2.60 (d, J = 16.8 Hz, 1H) , 2.45 -2.30 (m, 1H) , 2.18 -2.05 (m, 2H) , 2.05 -1.94 (m, 3H) , 1.94 -1.80 (m, 3H) , 1.77 -1.51 (m, 7H) .
2D NMR (400 MHz, DMSO-d6) showed that was right.
Step E: 3- (6- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperidyl] -8-aza-8-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
A mixture of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (50 mg, 124.55 μmol) , 3- [6- [3- [4- (4-aminophenyl) -1-piperidyl] -8-azaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (103.82 mg, 186.83 μmol) , AcOH (74.79 mg, 1.25 mmol) in n-BuOH (3 mL) was stirred at 100 ℃ for 16hr. The mixture was concentrated to give a residue, which was purified by prep-HPLC (column: Welch Xtimate C18 100*40mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 8%-38%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 877.8 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H) , 10.22 (TFA, 0.89H) , 9.47 (s, 1H) , 8.88 (s, 1H) , 7.99 (t, J = 7.6 Hz, 1H) , 7.76 (d, J = 8.0 Hz, 1H) , 7.70 (d, J = 8.0 Hz, 2H) , 7.61 (d, J = 7.6 Hz, 1H) , 7.45 (d, J = 8.4 Hz, 1H) , 7.32 (d, J = 7.6 Hz, 1H) , 7.26 (s, 1H) , 7.19 (d, J = 8.4 Hz, 2H) , 5.45 -5.20 (m, 2H) , 5.09 (dd, J = 13.2, 4.8 Hz, 1H) , 4.48 (m, 2H) , 4.18 -4.37 (m, 2H) , 3.69 -3.56 (m, 3H) , 3.34 -3.19 (m, 4H) , 3.03 (m, 2H) , 2.96 -2.77 (m, 2H) , 2.60 (d, J = 17.6 Hz, 1H) , 2.45 -2.31 (m, 1H) , 2.22 -2.07 (m, 3H) , 2.05 -1.82 (m, 8H) , 1.77 -1.48 (m, 12H) .
Example 121:
3- [6- (4- {3- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -1-pyrrolidinyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-329)
Scheme:
Step A: 3- (1-oxo-6- (1, 4-dioxa-8-azaspiro [4.5] decan-8-yl) isoindolin-2-yl) piperidine-2, 6-
dione
To a solution of 3- (6-bromo-1-oxoisoindolin-2-yl) piperidine-2, 6-dione (500 mg, 1.55 mmol) in dioxane (5.0 mL) was added 1, 4-dioxa-8-azaspiro [4.5] decane (443 mg, 3.10 mmol) , Cs2CO3 (1.51 g, 4.65 mmol) , Pd-PEPPSI-IPentCl (80 mg, 0.0775 mmol) , was stirred 100 ℃ for 16 h under N2 atmosphere. After completion of reaction, quenched with water (100 mL) , extracted with DCM (3*100 mL) . The organic layers were washed with brine (50 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 386.1 [M+H] +.
Step B: 3- (1-oxo-6- (4-oxopiperidin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione
A solution of 3- (1-oxo-6- (1, 4-dioxa-8-azaspiro [4.5] decan-8-yl) isoindolin-2-yl) piperidine-2, 6-dione (300 mg, 0.779 mmol) in 0.5M HCl (12 mL) was stirred at 25℃ for 2 hours. The resulting mixture was concentrated in vacuo. The residue was dissolved in water (100 mL) and extracted with DCM (100 mL x 3) . The combined organic layers were washed with brine (100 mL) , dried over anhydrous Na2S04, and filtered. The filtrate was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 342.2 [M+H] +.
Step C: 3- (6- (4- (3- (4- (4-nitrophenyl) piperazin-1-yl) pyrrolidin-1-yl) piperidin-1-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
A mixture of 3- (1-oxo-6- (4-oxopiperidin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione (96 mg, 0.282 mmol) , 1- (4-nitrophenyl) -4- (pyrrolidin-3-yl) piperazine (155 mg, 0.564 mmol) and NaBH (OAc) 3 (179 mg, 0.846 mmol) in DCM (5.0 mL) and MeOH (5.0 mL) and HOAc (0.5 mL) was stirred at 35℃ for 16 h . After completion of reaction, quenched with water (50 mL) , extracted with EtOAc (3*50 mL) . The organic layers were washed with brine (15.0 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 602.2 [M+H] +.
Step D: 3- (6- (4- (3- (4- (4-aminophenyl) piperazin-1-yl) pyrrolidin-1-yl) piperidin-1-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a solution of 3- (6- (4- (3- (4- (4-nitrophenyl) piperazin-1-yl) pyrrolidin-1-yl) piperidin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (130 mg, 0.216 mmol) in EtOH (10 mL) and H2O (3 ml) was added NH4Cl (58 mg 1.08 mmol) , Fe (60 mg 1.08 mmol) . The mixture was stirred at 90℃ for 3 h. After completion of reaction, the reaction was filtered. The filtrate was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 572.1 [M+H] +.
Step E: 3- [6- (4- {3- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -1-pyrrolidinyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -2, 6-
piperidinedione
To a solution of (R, Z) -12-hydroxy-12-methyl-2- (methylthio) -7, 10, 11, 12-tetrahydro-5H-13, 17-(azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20 mg, 0.0519 mmol) in DCM (10 mL) was added 3- (6- (4- (3- (4- (4-aminophenyl) piperazin-1-yl) pyrrolidin-1-yl) piperidin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (60 mg, 0.104 mmol) . Then the reaction mixture was stirred at 25℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EtOAc (20
mL x3) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated. The residue was purified by prep-HPLC (Waters 2767/Qda, Column: Pursuit XRs C18, 19.5*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 23%~33%; Retention Time: 7.8-9.0min of 16 min) to afford the desired product.
LC-MS (ESI) [M+H] + 893.3.
1H NMR (400 MHz, DMSO-d6) δ 10.96 (s, 1H) , 10.05 (s, 1H) , 8.83 (s, 1H) , 8.00 (t, J = 7.8 Hz, 1H) , 7.75 (d, J = 7.9 Hz, 1H) , 7.61 (s, 1H) , 7.60 (s, 1H) , 7.58 (s, 1H) , 7.46 (d, J = 8.5 Hz, 1H) , 7.32 (d, J = 8.3 Hz, 1H) , 7.23 (d, J = 8.8 Hz, 1H) , 6.98 (s, 1H) , 6.96 (s, 1H) , 5.40 –5.20 (m, 3H) , 5.09 (dd, J = 13.0, 4.9 Hz, 1H) , 4.46 (m, 2H) , 4.35 (d, J = 17.0 Hz, 1H) , 4.24 –4.19 (m, 1H) , 3.95 (m, 4H) , 2.92 (m, 4H) , 2.79 (m, 4H) , 2.60 (d, J = 17.6 Hz, 2H) , 2.45 –2.35 (m, 3H) , 2.16 (m, 5H) , 2.05 –1.91 (m, 5H) , 1.73 (m, 4H) , 1.55 (m, 4H) .
Example 122:
3- [5- (4- {3- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -1-pyrrolidinyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-330)
Scheme:
Step A: 3- (1-oxo-5- (1, 4-dioxa-8-azaspiro [4.5] decan-8-yl) isoindolin-2-yl) piperidine-2, 6-
dione
To a solution of 3- (5-bromo-1-oxoisoindolin-2-yl) piperidine-2, 6-dione (500 mg, 1.55 mmol) in dioxane (5.0 mL) was added 1, 4-dioxa-8-azaspiro [4.5] decane (443 mg, 3.10 mmol) , Cs2CO3 (1.51 g, 4.65 mmol) , Pd-PEPPSI-IPentCl (80 mg, 0.0775 mmol) , was stirred 100 ℃ for 2 h under N2 atmosphere. After completion of reaction, quenched with water (100 mL) , extracted with DCM (3*100 mL) . The organic layers were washed with brine (50 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 386.1 [M+H] +.
Step B: 3- (1-oxo-5- (4-oxopiperidin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione
A solution of 3- (1-oxo-5- (1, 4-dioxa-8-azaspiro [4.5] decan-8-yl) isoindolin-2-yl) piperidine-2, 6-dione (300 mg, 0.779 mmol) in 0.5M HCl (12 mL) was stirred at 25℃ for 12 hours. The resulting mixture was concentrated in vacuo. The residue was dissolved in water (100 mL) and extracted with DCM (100 mL x 3) . The combined organic layers were washed with brine (100 mL) , dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 342.2 [M+H] +.
Step C: 3- (5- (4- (3- (4- (4-nitrophenyl) piperazin-1-yl) pyrrolidin-1-yl) piperidin-1-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
A mixture of 3- (1-oxo-5- (4-oxopiperidin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione (96 mg, 0.282 mmol) , 1- (4-nitrophenyl) -4- (pyrrolidin-3-yl) piperazine (155 mg, 0.564 mmol) and NaBH (OAc) 3 (179 mg, 0.846 mmol) in DCM (5.0 mL) and MeOH (5.0 mL) and HOAc (0.5 mL) was stirred at 35℃ for 16 h . After completion of reaction, quenched with water (50 mL) , extracted with EtOAc (3*50 mL) . The organic layers were washed with brine (15.0 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 602.2 [M+H] +.
Step D: 3- (5- (4- (3- (4- (4-aminophenyl) piperazin-1-yl) pyrrolidin-1-yl) piperidin-1-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a solution of 3- (5- (4- (3- (4- (4-nitrophenyl) piperazin-1-yl) pyrrolidin-1-yl) piperidin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (130 mg, 0.216 mmol) in EtOH (10 mL) and H2O (3 mL) was added NH4Cl (58 mg 1.08 mmol) , Fe (60 mg 1.08 mmol) . The mixture was stirred at 90℃ for 2 h. After completion of reaction, the reaction was filtered. The filtrate was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 572.1 [M+H] +.
Step E: 3- [5- (4- {3- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -1-pyrrolidinyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -2, 6-
piperidinedione
To a solution of 3- (5- (4- (3- (4- (4-aminophenyl) piperazin-1-yl) pyrrolidin-1-yl) piperidin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (20 mg, 0.0519 mmol) in DCM (10 mL) was added 3- (6- (4- (3- (4- (4-aminophenyl) piperazin-1-yl) pyrrolidin-1-yl) piperidin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (60 mg, 0.104 mmol) . Then the reaction mixture was stirred at 25℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EtOAc (20 mL x3) . The organic layers were washed with
brine (10 mL) , dried over Na2SO4, filtered and concentrated. The residue was purified by prep-HPLC (Waters 2767/Qda, Column: Pursuit XRs C18, 19.5*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 23%~33%; Retention Time: 7.8-9.0min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 893.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.94 (s, 1H) , 10.04 (s, 1H) , 8.82 (s, 1H) , 8.19 (s, 1H) , 8.00 (d, J = 7.9 Hz, 1H) , 7.75 (d, J = 8.1 Hz, 1H) , 7.62 –7.45 (m, 4H) , 7.06 (d, J = 8.5 Hz, 2H) , 6.90 (d, J = 9.2 Hz, 2H) , 5.32 (m, 3H) , 5.04 (dd, J = 13.3, 4.9 Hz, 1H) , 4.45 (d, J = 7.3 Hz, 2H) , 4.32 (d, J = 17.0 Hz, 1H) , 4.20 (d, J = 16.7 Hz, 1H) , 3.79 (d, J = 12.2 Hz, 2H) , 3.08 (m, 4H) , 2.87 (m, 6H) , 2.60 (m, 6H) , 2.46 –2.31 (m, 2H) , 2.29 –2.09 (m, 2H) , 2.07 –1.85 (m, 6H) , 1.76 –1.63 (m, 2H) , 1.56 (m, 3H) , 1.54 –1.40 (m, 2H) .
Example 123:
3- [6- (4- {1- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -3-pyrrolidinyl} -1-piperazinyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-331)
Scheme:
Step A: tert-butyl 4- (2- (2, 6-dioxopiperidin-3-yl) -3-oxoisoindolin-5-yl) piperazine-1-
carboxylate
To a solution of 3- (6-bromo-1-oxoisoindolin-2-yl) piperidine-2, 6-dione (1 g, 3.10 mmol) , tert-butyl piperazine-1-carboxylate (577 mg, 3.10 mmol) in Dioxane (20 mL) was added CO3Cs2
(3.03 g, 9.30 mmol) , Ruphos (145 mg, 0.310 mmol) Ruphos-Pd-G2 (241 mg, 0.310 mmol) . Then the mixture was stirred at 100℃ for 16 h under N2 atmosphere. After completion of reaction, the reaction mixture was quenched with water (200 mL) , and the solution was extracted with EtOAc (200 mL x 3) . The combined organic layers were washed with brine (200 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography eluted with DCM: MeOH=10: 1 to afford the desired product.
LCMS: MS (ESI) m/z 429.1 [M+H] +.
Step B: 3- (1-oxo-6- (piperazin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione
To a solution of tert-butyl 4- (2- (2, 6-dioxopiperidin-3-yl) -3-oxoisoindolin-5-yl) piperazine-1-carboxylate (328 mg, 0.766 mmol) in HCl in dioxane (10 mL, 4 M) and DCM (10 mL) was stirred at 35℃ for 2 h. After completion of reaction, the reaction mixture was concentrated under reduced pressure to dryness to afford the desired product.
LCMS: MS (ESI) m/z 329.1 [M+H] +.
Step C: tert-butyl 3- (4- (2- (2, 6-dioxopiperidin-3-yl) -3-oxoisoindolin-5-yl) piperazin-1-
yl) pyrrolidine-1-carboxylate
To a solution of 3- (1-oxo-6- (piperazin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione (50 mg, 0.152 mmol) , tert-butyl 3-oxopyrrolidine-1-carboxylate (28.1 mg, 0.152 mmol) , NaBH (OAc) 3 (96.7 mg, 0.456 mmol) , CH3COOH (27.4 mg, 0.456 mmol) in DCM (2 mL) . Then the mixture was stirred at 35℃ for 16 h. The reaction mixture was quenched with water (20 mL) , and the solution was extracted with DCM (20 mL x 3) . The combined organic layers were washed with brine (20 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography eluted with DCM: MeOH=10: 1 to afford the desired product.
LCMS: MS (ESI) m/z 498.3 [M+H] +.
Step D: 3- (1-oxo-6- (4- (pyrrolidin-3-yl) piperazin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione
To a solution of tert-butyl 3- (4- (2- (2, 6-dioxopiperidin-3-yl) -3-oxoisoindolin-5-yl) piperazin-1-yl) pyrrolidine-1-carboxylate (144 mg, 0.290 mmol) in HCl in dioxane (2 mL, 4 M) and DCM (2 mL) was stirred at 35℃ for 2 h. After completion of reaction, the reaction mixture was concentrated under reduced pressure to dryness to afford the desired product.
LCMS: MS (ESI) m/z 398.2 [M+H] +.
Step E: 3- (6- (4- (1- (1- (4-nitrophenyl) piperidin-4-yl) pyrrolidin-3-yl) piperazin-1-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a solution of 3- (1-oxo-6- (4- (pyrrolidin-3-yl) piperazin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione (115 mg, 0.290 mmol) , 1- (4-nitrophenyl) piperidin-4-one (95.7 mg, 0.435 mmol) , NaBH (OAc) 3 (184 mg, 0.87 mmol) , CH3COOH (52.2 mg, 0.87mmol) in DCM (10 mL) . Then
the mixture was stirred at 35℃ for 16 h. The reaction mixture was quenched with water (100 mL) , and the solution was extracted with DCM (100 mL x 3) . The combined organic layers were washed with brine (100 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography eluted with DCM: MeOH=10: 1 to afford the desired product.
LCMS: MS (ESI) m/z 602.4 [M+H] +.
Step F: 3- (6- (4- (1- (1- (4-aminophenyl) piperidin-4-yl) pyrrolidin-3-yl) piperazin-1-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a solution of 3- (6- (4- (1- (1- (4-nitrophenyl) piperidin-4-yl) pyrrolidin-3-yl) piperazin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (105 mg, 0.175 mmol) , NH4Cl (46.4 mg, 0.875 mmol) , in H2O (2 mL) and EtOH (20 mL) were added Fe (49 mg, 0.875 mmol) , Then the mixture was stirred at 90℃ for 2h. After completion of reaction, the mixture reaction was filtered through a Celite pad, and the filtrate was concentrated to give the crude. The crude was quenched with H2O (10 mL) , adjusted to pH 8 with solid NaHCO3 and extracted with EtOAc (3 x 30 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 572.3 [M+H] +.
Step G: 3- [6- (4- {1- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -3-pyrrolidinyl} -1-piperazinyl) -1-oxo-2-isoindolinyl] -2, 6-
piperidinedione
To a solution of give 3- (6- (4- (1- (1- (4-aminophenyl) piperidin-4-yl) pyrrolidin-3-yl) piperazin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (95 mg, 0.166 mmol) in n-Butanol (10 mL) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20 mg, 0.0519 mmol) , the mixture was stirred at 100℃ for 16 hrs under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 3767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.05%TFA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 17-27%Retention Time: 8.0-9.4 of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 447.4 [1/2M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.95 (s, 1H) , 10.09 –9.83 (m, 1H) , 8.81 (s, 1H) , 8.25 (s, 1H) , 8.01 (t, J = 9.8 Hz, 1H) , 7.76 (d, J = 8.0 Hz, 1H) , 7.57 (dd, J = 12.0, 7.4 Hz, 3H) , 7.42 (d, J = 8.4 Hz, 1H) , 7.28 –7.21 (m, 1H) , 7.15 (d, J = 2.3 Hz, 1H) , 6.91 (d, J = 9.0 Hz, 2H) , 5.32 (m,
3H) , 5.09 (dd, J = 12.6, 4.5 Hz, 1H) , 4.46 (dd, J = 10.6, 4.8 Hz, 2H) , 4.33 (d, J = 16.5 Hz, 1H) , 4.21 (d, J = 16.4 Hz, 1H) , 3.62 –3.54 (m, 2H) , 3.19 (dd, J = 6.3, 4.0 Hz, 4H) , 2.92 –2.77 (m, 3H) , 2.71 –2.65 (m, 3H) , 2.57 (dd, J = 17.0, 5.7 Hz, 6H) , 2.40 (m, 2H) , 2.23 –2.11 (m, 2H) , 2.05 –1.96 (m, 3H) , 1.91 (m, 3H) , 1.77 –1.61 (m, 2H) , 1.56 (m, 3H) , 1.53 –1.43 (m, 2H) .
Example 124:
3- [5- (4- {1- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -3-pyrrolidinyl} -1-piperazinyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-332)
Scheme:
Step A: 3- (1-oxo-5- (piperazin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione
To a solution of tert-butyl 4- (2- (2, 6-dioxopiperidin-3-yl) -1-oxoisoindolin-5-yl) piperazine-1-carboxylate (231 mg, 0.54 mmol) , HCl in dioxane (1M 5 ml) in DCM (15 mL) . Then the mixture was stirred at 35℃ for 2 h. The resulting mixture was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 329.1 [M+H] +.
Step B: tert-butyl 3- (4- (2- (2, 6-dioxopiperidin-3-yl) -1-oxoisoindolin-5-yl) piperazin-1-
yl) pyrrolidine-1-carboxylate
To a solution of 3- (1-oxo-5- (piperazin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione (139 mg, 0.424 mmol) , tert-butyl 3-oxopyrrolidine-1-carboxylate (78.4 mg, 0.424 mmol) , NaBH (OAc) 3 (134.8
mg, 0.636 mmol) , DCM (15 mL) . The resulting mixture was stirred for 16 h at 35℃ in an oil bath. The residue was purified by flash silica gel column chromatography (DCM: MeOH =20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 498.1 [M+H] +.
Step C: 3- (1-oxo-5- (4- (pyrrolidin-3-yl) piperazin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione
To a solution of tert-butyl 3- (4- (2- (2, 6-dioxopiperidin-3-yl) -1-oxoisoindolin-5-yl) piperazin-1-yl) pyrrolidine-1-carboxylate (150 mg, 0.302 mmol) in DCM (10 mL) was added HCl. dioxane (5 mL, 4 M) , then the reaction mixture was stirred at 30℃for 2 h under N2 atmosphere. After completion of reaction, the reaction mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 398.1 [M+H] +.
Step D: 3- (5- (4- (1- (1- (4-nitrophenyl) piperidin-4-yl) pyrrolidin-3-yl) piperazin-1-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a solution of 3- (1-oxo-5- (4- (pyrrolidin-3-yl) piperazin-1-yl) isoindolin-2-yl) piperidine-2, 6-dione (100 mg, 0.252 mmol) , 1- (4-nitrophenyl) piperidin-4-one (55.4 mg, 0.252 mmol) , NaBH (OAc) 3 (80.1 mg, 0.378 mmol) , DCM (15 mL) . The resulting mixture was stirred for 16 h at 35℃ in an oil bath. The residue was purified by flash silica gel column chromatography (DCM: MeOH =20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 602.3 [M+H] +.
Step E: 3- (5- (4- (1- (1- (4-aminophenyl) piperidin-4-yl) pyrrolidin-3-yl) piperazin-1-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a solution of 3- (5- (4- (1- (1- (4-nitrophenyl) piperidin-4-yl) pyrrolidin-3-yl) piperazin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (80 mg, 0.133 mmol) , Fe (37.3 mg, 0.667 mmol) , NH4Cl (35.3 mg, 0.667 mmol) in EtOH/H2O (10 mL/2mL) . Then the mixture was stirred at 90℃ for 3 h. The resulting mixture was filtered, the filter cake was washed with DCM (20 mL x3) . The filtrate was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 572.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.94 (s, 1H) , 7.52 (d, J = 8.8 Hz, 1H) , 7.06 (s, 1H) , 7.05 –7.00 (m, 1H) , 6.68 (d, J = 8.7 Hz, 2H) , 6.47 (d, J = 8.7 Hz, 2H) , 5.04 (dd, J = 13.3, 5.2 Hz, 1H) , 4.26 (dd, J = 50.0, 16.9 Hz, 3H) , 4.03 (d, J = 7.1 Hz, 1H) , 3.27 (s, 4H) , 3.19 –3.04 (m, 1H) , 2.91 (d, J = 13.2 Hz, 1H) , 2.84 (d, J = 13.2 Hz, 2H) , 2.64 (d, J = 26.4 Hz, 3H) , 2.44 –2.27 (m, 3H) , 1.99 (s, 2H) , 1.66 (s, 2H) , 1.49 (s, 4H) , 1.24 (s, 3H) , 1.20 –1.15 (m, 1H) , 0.85 (d, J = 6.3 Hz, 3H) .
Step F: 3- [5- (4- {1- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -3-pyrrolidinyl} -1-piperazinyl) -1-oxo-2-isoindolinyl] -2, 6-
piperidinedione
To a solution of 3- (5- (4- (1- (1- (4-aminophenyl) piperidin-4-yl) pyrrolidin-3-yl) piperazin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (46 mg, 0.081 mmol) in DCM (10 mL) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20 mg, 0.052 mmol) , the mixture was stirred at 25℃ for 16 h under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 15-23%Retention Time: 9-19 of 18 min) to afford the desired product.
LCMS: MS (ESI) m/z 893.1 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.97 (s, 1H) , 10.04 (s, 1H) , 8.82 (s, 1H) , 8.25 (s, 1H) , 8.02 (s, 1H) , 7.76 (d, J = 8.0 Hz, 1H) , 7.61 –7.51 (m, 3H) , 7.43 (d, J = 8.4 Hz, 1H) , 7.26 (d, J = 6.3 Hz, 1H) , 7.16 (s, 1H) , 6.92 (d, J = 9.0 Hz, 2H) , 5.29 (dd, J = 38.5, 13.0 Hz, 3H) , 5.10 (dd, J = 13.1, 5.2 Hz, 1H) , 4.45 (m, 2H) , 4.34 (d, J = 16.8 Hz, 1H) , 4.21 (d, J = 16.9 Hz, 1H) , 3.60 –3.55 (m, 2H) , 3.20 –3.16 (m, 4H) , 2.91 –2.81 (m, 3H) , 2.71 –2.65 (m, 3H) , 2.60 –2.53 (m, 7H) , 2.42 –2.33 (m, 2H) , 2.14 (m, 2H) , 2.07 –1.97 (m, 2H) , 1.95 –1.88 (m, 3H) , 1.76 –1.63 (m, 2H) , 1.56 (m, 3H) , 1.53 –1.42 (m, 2H) .
Example 125:
3- (5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-333)
Scheme:
Step A: 3- (5- (9- (4- (4-aminophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecan-3-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a solution of 3- (5- (9- (4- (4-nitrophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecan-3-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (190 mg, 0.316 mmol) in EtOH (10 mL) and H2O (3 mL) was added NH4Cl (103 mg 1.91 mmol) , Fe (107 mg 1.91 mmol) . The mixture was stirred at 90℃ for 2 h. After completion of reaction, the reaction was filtered. The filtrate was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 571.4 [M+H] +.
Step B: 3- (5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 3- (5- (4- (3- (4- (4-aminophenyl) piperazin-1-yl) pyrrolidin-1-yl) piperidin-1-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (30 mg, 0.077 mmol) in DCM (3 mL) was added 3- (5- (9- (4- (4-aminophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecan-3-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (66 mg, 0.116 mmol) . Then the reaction mixture was stirred at 35℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EtOAc (20 mL x3) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated. The residue was purified by prep-HPLC (Waters 2767/Qda, Column: Pursuit XRs C18, 19.5*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 23%~33%; Retention Time: 7.8-9.0min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 892.4 [M+H] +.
1H NMR (400 MHz, DMSO) δ 10.94 (s, 1H) , 10.03 (s, 1H) , 8.82 (s, 1H) , 8.36 (m, 2H) , 8.02 (s, 1H) , 7.76 (d, J = 7.9 Hz, 1H) , 7.58 (m, 3H) , 7.50 (d, J = 8.5 Hz, 1H) , 7.04 (s, 1H) , 6.90 (d, J = 8.8 Hz, 2H) , 5.34 (m, 3H) , 5.05 (s, 1H) , 4.45 (m, 2H) , 4.32 (s, 1H) , 4.21 (s, 1H) , 3.07 (m, 4H) , 3.00 (d, J = 6.3 Hz, 2H) , 2.94 –2.82 (m, 4H) , 2.37 (d, J = 10.0 Hz, 1H) , 2.32 (d, J = 13.7 Hz, 1H) , 2.25 –2.18 (m, 3H) , 2.01 (s, 1 H) , 1.96 (m, 3H) , 1.75 (m, 3H) , 1.65 (m, 3H) , 1.56 (m, 5H) , 1.41 (m, 4H) , 1.16 (m, 2H) .
Example 126:
3- (6- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-334)
Scheme:
Step A: 3- (6- (9- (4- (4-nitrophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecan-3-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a solution of 9- (4- (4-nitrophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecane (200 mg, 0.559 mmol) , 3- (6-bromo-1-oxoisoindolin-2-yl) piperidine-2, 6-dione (180.4 mg, 0.559 mmol) , Pd-PEPPSI-IPentCl (27.1 mg, 0.0279 mmol) , Cs2CO3 (546 mg, 1.68 mmol) in dioxane (20 mL) . Then the mixture was stirred at 100℃ for 16 h under N2. The resulting mixture was filtered, the filter cake was washed with DCM (20 mL x3) . The filtrate was concentrated under reduced pressure. The crude product was purified by silica gel chromatography eluted with
DCM: MeOH=10: 1 to afford the desired product.
LCMS: MS (ESI) m/z 601.1 [M+H] +.
Step B: 3- (6- (9- (4- (4-aminophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecan-3-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a solution of 3- (6- (9- (4- (4-nitrophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecan-3-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (120 mg, 0.20 mmol) , Fe (67 mg, 1.20 mmol) , NH4Cl (63 mg, 1.20 mmol) in EtOH/H2O (10 mL/2mL) . Then the mixture was stirred at 90℃ for 3 h. The resulting mixture was filtered, the filter cake was washed with DCM (20 ml x3) . The filtrate was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 571.2 [M+H] +.
Step C: 3- (6- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 3- (6- (9- (4- (4-aminophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecan-3-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (77 mg, 0.135 mmol) in DCM (10 mL) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20 mg, 0.052 mmol) , the mixture was stirred at 25℃ for 16 h under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 12-19%Retention Time: 9-23 of 14 min) to afford the desired product.
LCMS: MS (ESI) m/z 892.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.01 –10.75 (m, 1H) , 10.02 (s, 1H) , 8.82 (s, 1H) , 8.23 –8.12 (m, 1H) , 8.01 (s, 1H) , 7.82 –7.68 (m, 1H) , 7.63 –7.43 (m, 4H) , 7.11 –6.98 (m, 2H) , 6.93 –6.80 (m, 2H) , 5.50 –5.14 (m, 3H) , 5.04 (dd, J = 14.9, 6.4 Hz, 1H) , 4.49 –4.39 (m, 2H) , 4.35 –4.27 (m, 1H) , 4.25 –4.14 (m, 1H) , 3.82 –3.74 (m, 2H) , 3.07 (d, J = 2.4 Hz, 4H) , 2.93 –2.80 (m, 5H) , 2.69 –2.53 (m, 5H) , 2.44 –2.30 (m, 5H) , 2.21 (dd, J = 17.2, 10.8 Hz, 2H) , 2.06 –1.95 (m, 2H) , 1.94 –1.84 (m, 4H) , 1.74 –1.62 (m, 2H) , 1.56 (m, 3H) , 1.45 (m, 2H) .
Example 127:
3- (6- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -7-aza-7-spiro [3.5] nonyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-335)
Scheme:
Step A: 3- (6- (2, 2-dimethoxy-7-azaspiro [3.5] nonan-7-yl) -1-oxoisoindolin-2-yl) piperidine-
2, 6-dione
To a solution of 3- (6-bromo-1-oxoisoindolin-2-yl) piperidine-2, 6-dione (1000 mg, 3 mmol) in dioxane (10 ml) was added 2, 2-dimethoxy-7-azaspiro [3.5] nonane (572 mg, 3 mmol) , RuPhosPdG2 (489 mg, 0.263mmol) and t-BuONa (865 mg, 9 mmol) . The mixture was stirred at 100℃ for 16 hrs under N2 atmosphere. After completion of reaction, quenched with water (30 mL) , extracted with EA (3*50 mL) . The organic layers were washed with brine (20 mL) , dried over Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (DCM: MeOH =20: 1) to afford the desired product.
LC-MS (ESI) [M+H] + 428.2
Step B: 3- (1-oxo-6- (2-oxo-7-azaspiro [3.5] nonan-7-yl) isoindolin-2-yl) piperidine-2, 6-dione
To a solution of 3- (6- (2, 2-dimethoxy-7-azaspiro [3.5] nonan-7-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (200 mg, 0.468 mmol) in DCM/TFA (1 mL/1 mL ) was stirred at 25℃
for 2 h under N2 atmosphere. After completion of reaction, the reaction mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 382.0 [M+H] +.
Step C: 3- (7- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -4-fluoro-1-
oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione
To a solution of 3- (1-oxo-6- (2-oxo-7-azaspiro [3.5] nonan-7-yl) isoindolin-2-yl) piperidine-2, 6-dione (115 mg, 0.302 mmol) , 1- (4-nitrophenyl) piperazine (62.5 mg, 0.302 mmol) in DCM (5 mL) and MeOH (5 mL) was stirred at 25 ℃ for 0.5 h. Then NaBH (OAc) 3 (319 mg, 1.51 mmol) was added. The mixture was stirred at 25 ℃ for 48 h. Water (40 mL) was added and the solution was extracted with EtOAc (10 mL x 3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography eluted with PE: EA=1: 1 to afford the desired product.
LCMS: MS (ESI) m/z 573.2 [M+H] +.
Step D: 3- (6- (2- (4- (4-aminophenyl) piperazin-1-yl) -7-azaspiro [3.5] nonan-7-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a solution of 3- (7- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -4-fluoro-1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (150 mg, 0.262 mmol) in EtOH/H2O (10 mL/1 mL) was added Fe (73.2 mg, 1.31 mmol) , NH4Cl (70.1 mg, 1.31 mmol) then the reaction mixture was stirred at 90 ℃ for 2 h under N2 atmosphere. After completion of reaction, filtered, the filtrate was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 543.2 [M+H] +.
Step E: 3- (6- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -7-aza-7-spiro [3.5] nonyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 3- (6- (2- (4- (4-aminophenyl) piperazin-1-yl) -7-azaspiro [3.5] nonan-7-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (70.0 mg, 0.129 mmol) in DCM (10 ml) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20.0 mg, 0.052 mmol) then the reaction mixture was stirred at 25 ℃ for 16 h under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 ×10 mL) . the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase
A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 17-27%Retention Time: 9-19 of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 864.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.97 (s, 1H) , 10.10 (brs, 1H) , 9.74 (brs, 1H) , 8.85 (s, 1H) , 8.00 (t, J = 8.0 Hz, 1H) , 7.75 (d, J = 7.8 Hz, 1H) , 7.67 -7.57 (m, 3H) , 7.43 (d, J = 8.5 Hz, 1H) , 7.32 -7.25 (m, 1H) , 7.21 -7.17 (m, 1H) , 7.00 (d, J = 9.1 Hz, 2H) , 5.48 -5.20 (m, 3H) , 5.09 (dd, J = 13.1, 4.9 Hz, 1H) , 4.53 -4.43 (m, 2H) , 4.38 -4.29 (m, 1H) , 4.25 -4.15 (m, 1H) , 3.86 -3.76 (m, 2H) , 3.52 -3.44 (m, 2H) , 3.29 -3.21 (m, 2H) , 3.20 -3.13 (m, 2H) , 3.02 –2.87 (m, 5H) , 2.66 -2.54 (m, 1H) , 2.44 -2.34 (m, 2H) , 2.29 -2.18 (m, 3H) , 2.10 -2.02 (m, 3H) , 2.01 -1.91 (m, 2H) , 1.75 –1.66 (m, 5H) , 1.57 (m, 3H) .
Example 128:
3- (6- {8- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -2-aza-2-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-336)
Scheme:
Step A: tert-butyl 8- (4- (4-nitrophenyl) piperazin-1-yl) -2-azaspiro [4.5] decane-2-carboxylate
A solution of tert-butyl 8-oxo-2-azaspiro [4.5] decane-2-carboxylate (500 mg, 0.987 mmol) , 1- (4-nitrophenyl) piperazine (204 mg, 1.97 mmol) , HOAc (0.5 mL) in DCM (10 mL) and MeOH (1 mL) was stirred at 25℃ for 0.5 h. Then NaBH (OAc) 3 (628 mg, 2.96 mmol) was added. The mixture was stirred at 25℃ for 16 hrs. After completion, the mixture was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 445.3 [M+H] +.
Step B: 8- (4- (4-nitrophenyl) piperazin-1-yl) -2-azaspiro [4.5] decane
To a solution of tert-butyl 8- (4- (4-nitrophenyl) piperazin-1-yl) -2-azaspiro [4.5] decane-2-carboxylate (890 mg, 2.00 mmol) in DCM (10 mL) was added HCl in dioxane (3 mL, 4 M) and stirred at 20℃ for 2 hr. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with DCM (20 mL x3) . The organic layers were washed with brine
(10 mL) , dried over Na2SO4, filtered and concentrated. under reduced pressure to dryness to afford the desired product.
LCMS: MS (ESI) m/z 345.1 [M+H] +.
Step C: 3- (6- (8- (4- (4-nitrophenyl) piperazin-1-yl) -2-azaspiro [4.5] decan-2-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
A mixture of 8- (4- (4-nitrophenyl) piperazin-1-yl) -2-azaspiro [4.5] decane (290 mg, 0.843 mmol) , 3- (6-bromo-1-oxoisoindolin-2-yl) piperidine-2, 6-dione (272 mg, 0.843 mmol) , Pd-PEPPSI-IPentCl (41.0 mg, 0.0422 mmol) , Cs2CO3 (825 mg, 2.53 mmol) in dioxane (5.0 mL) was stirred at 100℃ for 16 hrs under N2. After completion, the mixture was quenched with H2O (10 mL) , extracted with DCM (10 mLx3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 587.3 [M+H] +.
Step D: 3- (6- (8- (4- (4-aminophenyl) piperazin-1-yl) -2-azaspiro [4.5] decan-2-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
A mixture of 3- (6- (8- (4- (4-nitrophenyl) piperazin-1-yl) -2-azaspiro [4.5] decan-2-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (100 mg, 0.171 mmol) , iron powder (47.8 mg, 0.855 mmol) , NH4Cl (37.2 mg, 0.855 mmol) in EtOH (10 mL) and H2O (2 mL) was stirred at 90℃ for 2 hrs. After completion of reaction, the mixture reaction was filtered through a Celite pad, and the filtrate was concentrated to give the crude. The crude was quenched with H2O (10 mL) , adjusted to pH 8 with solid NaHCO3 and extracted with EtOAc (3 x 30 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 557.2 [M+H] +.
Step E: 3- (6- {8- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -2-aza-2-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20.0 mg, 0.0556 mmol) in DCM (3 mL) was added 3- (6- (8- (4- (4-aminophenyl) piperazin-1-yl) -2-azaspiro [4.5] decan-2-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (80.0 mg, 0.144 mmol) in DCM (8 mL) . Then the reaction mixture was stirred at 35℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and
extracted with EA (20 mL x3) . The organic layers were concentrated. The residue was purified by prep-HPLC (Waters 2767, Column: Pursuit XRs C18, 21.2*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 16%~24%Retention Time: 9.7-10.5 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 878.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.95 (s, 1H) , 10.08 (s, 1H) , 9.36 (s, 1H) , 8.84 (s, 1H) , 8.00 (t, J = 7.8 Hz, 1H) , 7.75 (d, J = 8.3 Hz, 1H) , 7.65 –7.60 (m, 3H) , 7.39 (d, J = 7.7 Hz, 1H) , 7.00 (d, J = 9.1 Hz, 2H) , 6.83 (d, J = 7.7 Hz, 2H) , 5.44 –5.20 (m, 3H) , 5.09 (d, J = 18.2 Hz, 1H) , 4.46 (m, 2H) , 4.31 (d, J = 16.2 Hz, 1H) , 4.19 (d, J = 16.0 Hz, 1H) , 3.81 (m, 2H) , 3.52 (m, 2H) , 3.35 (m, 2H) , 3.24 (m, 3H) , 2.95 (d, J = 11.3 Hz, 2H) , 2.59 (ddd, J = 14.0, 7.3, 3.8 Hz, 2H) , 2.40 (d, J = 4.9 Hz, 2H) , 2.21 (dd, J = 23.3, 11.6 Hz, 2H) , 2.07 –2.04 (m, 2H) , 1.98 (d, J = 8.6 Hz, 2H) , 1.85 (d, J = 9.0 Hz, 2H) , 1.78 (m, 2H) , 1.72 –1.63 (m, 3H) , 1.57 (m, 3H) , 1.45 (m, 3H) .
Example 129:
3- (5- {7- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -2-aza-2-spiro [3.5] nonyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-337)
Scheme:
Step A: 3- (5- (7- (4- (4-nitrophenyl) piperazin-1-yl) -2-azaspiro [3.5] nonan-2-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
A mixture of 7- (4- (4-nitrophenyl) piperazin-1-yl) -2-azaspiro [3.5] nonane (200 mg, 0.606 mmol) , 3- (5-bromo-1-oxoisoindolin-2-yl) piperidine-2, 6-dione (196 mg, 0.606 mmol) , Pd-PEPPSI-IPentCl (29.5 mg, 0.0303 mmol) , Cs2CO3 (593 mg, 1.82 mmol) in dioxane (5.0 mL) was stirred at 100℃ for 16 hrs under N2. After completion, the mixture was quenched with H2O (10 mL) , extracted with DCM (10 mLx3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 573.2 [M+H] +.
Step B: 3- (5- (7- (4- (4-aminophenyl) piperazin-1-yl) -2-azaspiro [3.5] nonan-2-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
A mixture of 3- (5- (7- (4- (4-nitrophenyl) piperazin-1-yl) -2-azaspiro [3.5] nonan-2-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (120 mg, 0.210 mmol) , iron powder (58.6 mg, 1.05 mmol) , NH4Cl (56.2 mg, 1.05 mmol) in EtOH (10 mL) and H2O (2 mL) was stirred at 90℃ for 2 hrs. After completion of reaction, the mixture reaction was filtered through a Celite pad, and the filtrate was concentrated to give the crude. The crude was quenched with H2O (10 mL) ,
adjusted to pH 8 with solid NaHCO3 and extracted with EtOAc (3 x 30 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 543.4 [M+H] +.
Step C: 3- (5- {7- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -2-aza-2-spiro [3.5] nonyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 3- (5- (7- (4- (4-aminophenyl) piperazin-1-yl) -2-azaspiro [3.5] nonan-2-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (30.0 mg, 0.0778 mmol) in DCM (3 mL) was added 3- (5- (8- (4- (4-aminophenyl) piperazin-1-yl) -2-azaspiro [4.5] decan-2-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (100 mg, 0.184 mmol) in DCM (8 mL) . Then the reaction mixture was stirred at 35℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EA (20 mL x3) . The organic layers were concentrated. The residue was purified by prep-HPLC (Waters 2767, Column: Pursuit XRs C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 13%~20%Retention Time: 8.9-10.9 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 864.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.92 (s, 1H) , 10.02 (s, 1H) , 8.82 (s, 1H) , 8.23 (s, 1H) , 8.02 (s, 1H) , 7.76 (d, J = 8.1 Hz, 1H) , 7.58 (t, J = 8.3 Hz, 3H) , 7.48 (d, J = 8.3 Hz, 1H) , 6.90 (d, J = 9.1 Hz, 2H) , 6.58 –6.39 (m, 2H) , 5.41 –5.24 (m, 2H) , 5.03 (dd, J = 13.1, 5.3 Hz, 1H) , 4.53 –4.33 (m, 2H) , 4.24 (dd, J = 50.8, 16.8 Hz, 2H) , 3.65 (m, 2H) , 3.59 (m, 2H) , 3.07 (m, 5H) , 2.88 (d, J = 13.0 Hz, 2H) , 2.72 –2.55 (m, 6H) , 2.34 –2.27 (m, 2H) , 1.96 (m, 4H) , 1.84 –1.67 (m, 4H) , 1.56 (m, 3H) , 1.51 (m, 2H) , 1.38 –1.26 (m, 2H) .
Example 130:
3- (6- {7- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -2-aza-2-spiro [3.5] nonyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-338)
Scheme:
Step A: 3- (6- (7- (4- (4-aminophenyl) piperazin-1-yl) -2-azaspiro [3.5] nonan-2-yl) -1-
oxoisoindolin-2-yl) piperidine-2, 6-dione
To a solution of 3- (6- (7- (4- (4-nitrophenyl) piperazin-1-yl) -2-azaspiro [3.5] nonan-2-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (130 mg, 0.23 mmol) , Fe (76 mg, 1.36 mmol) , NH4Cl (72 mg, 1.36 mmol) in EtOH (10 mL) . Then the mixture was stirred at 90℃ for 2 h. The resulting mixture was filtered, the filter cake was washed with DCM (20 mL x3) . The filtrate was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 543.3 [M+H] +.
Step B: 3- (6- {7- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -2-aza-2-spiro [3.5] nonyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 3- (6- (7- (4- (4-aminophenyl) piperazin-1-yl) -2-azaspiro [3.5] nonan-2-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (90 mg, 0.166 mmol) in DCM (10 mL) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (30 mg, 0.078 mmol) ,
the mixture was stirred at 25℃ for 16 h under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 17-27%Retention Time: 9-19 of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 864.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.92 (s, 1H) , 9.97 (s, 1H) , 8.82 (s, 1H) , 8.22 (s, 1H) , 8.01 (dt, J = 6.4, 3.9 Hz, 1H) , 7.75 (dd, J = 10.3, 7.2 Hz, 1H) , 7.58 (t, J = 8.2 Hz, 3H) , 7.47 (t, J = 6.9 Hz, 1H) , 6.89 (t, J = 8.4 Hz, 2H) , 6.52 –6.42 (m, 2H) , 5.46 –5.15 (m, 3H) , 5.07 –4.95 (m, 1H) , 4.45 (dd, J = 6.3, 3.7 Hz, 2H) , 4.30 (d, J = 16.5 Hz, 1H) , 4.17 (d, J = 16.9 Hz, 1H) , 3.66 (d, J = 8.0 Hz, 2H) , 3.61 –3.56 (m, 2H) , 3.10-3.04 (m, 4H) , 2.94 –2.84 (m, 2H) , 2.64 (d, J = 2.2 Hz, 3H) , 2.60 –2.57 (m, 2H) , 2.35 –2.27 (m, 2H) , 2.22 –2.12 (m, 1H) , 2.07 –2.01 (m, 1H) , 1.98 –1.92 (m, 3H) , 1.79 –1.68 (m, 3H) , 1.56 (s, 3H) , 1.51 (m, 2H) , 1.39 –1.27 (m, 2H) .
Example 131:
3- (6- (4- ( (9- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidin-1-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (Cpd-147)
Scheme:
Step A: 7-6- (4- (dimethoxymethyl) piperidin-1-yl) phthalazin-1 (2H) -one
To a solution of 6-bromophthalazin-1 (2H) -one (567 mg, 2.52 mmol) in Dioxane (10 mL) was added 4- (dimethoxymethyl) piperidine (400 mg, 2.52 mmol) , Ruphos Pd-G2 (196 mg, 0.252 mmol) , t-BuONa (727 mg, 7.56 mmol) then the reaction mixture was stirred at 100 ℃ for 16 hrs under N2 atmosphere. After completion of reaction, quenched with H2O (10 mL) , extracted with EA (3*20 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =0: 1) to afford the desired product.
LCMS: MS (ESI) m/z 304.1 [M+H] +.
Step B: 3- (6- (4- (dimethoxymethyl) piperidin-1-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-
dione
To a solution of 7-6- (4- (dimethoxymethyl) piperidin-1-yl) phthalazin-1 (2H) -one (100 mg, 0.330 mmol) in DMF (10 ml) was added NaH (26.4 mg, 0.660 mmol) at 0℃, 3-bromopiperidine-2, 6-dione (63.4 mg, 0.330 mmol) , then the reaction mixture was stirred at 30℃ for 16 hrs under N2 atmosphere. the reaction mixture quenched with H2O (10 mL) , extracted with EA (3 × 10 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and
concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =1: 2) to afford the desired product.
LCMS: MS (ESI) m/z 415.2 [M+H] +.
Step C: 1- (2- (2, 6-dioxopiperidin-3-yl) -1-oxo-1, 2-dihydrophthalazin-6-yl) piperidine-4-
carbaldehyde
To a solution of 3- (6- (4- (dimethoxymethyl) piperidin-1-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (100 mg, 0.241 mmol) in FA (1 mL ) was stirred at 50℃ for 3 hrs under N2 atmosphere. After completion of reaction, the reaction mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 369.1 [M+H] +.
Step D: 3- (6- (4- ( (9- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17-
(azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) -3, 9-
diazaspiro [5.5] undecan-3-yl) methyl) piperidin-1-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-
2, 6-dione
To a solution of 1- (2- (2, 6-dioxopiperidin-3-yl) -1-oxo-1, 2-dihydrophthalazin-6-yl) piperidine-4-carbaldehyde (19.5 mg, 0.053 mmol) , (R, Z) -2- ( (4- (3, 9-diazaspiro [5.5] undecan-3-yl) phenyl) amino) -12-hydroxy-12-methyl-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (30.0 mg, 0.053 mmol) in DCM (5 mL) was stirred at 25 ℃ for 0.5 h. Then NaBH (OAc) 3 (16.7 mg, 0.080 mmol) was added. The mixture was stirred at 25 ℃ for 16 h. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 17-27%Retention Time: 9.1-12.3 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 919.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H) , 8.82 (s, 1H) , 8.30 (s, 1H) , 8.23 (s, 1H) , 8.01 (d, J = 8.7 Hz, 2H) , 7.76 (d, J = 7.8 Hz, 1H) , 7.58 (d, J = 7.4 Hz, 3H) , 7.48 (s, 1H) , 7.23 (s, 1H) , 6.90 (d, J = 9.4 Hz, 2H) , 5.76 (m, 1H) , 5.26 (m, 3H) , 4.45 (m, 2H) , 4.03 (m, 2H) , 3.08 (m, 4H) , 2.93 (m, 3H) , 2.67 (m, 1H) , 2.62 (d, J = 5.6 Hz, 1H) , 2.33 (m, 4H) , 2.15 (m, 2H) , 2.07 (d, J = 10.7 Hz, 2H) , 1.93 (d, J = 2.9 Hz, 1H) , 1.81 (m, 3H) , 1.74 –1.67 (m, 1H) , 1.56 (m, 7H) , 1.48 (m, 4H) , 1.27 –1.08 (m, 3H) .
Example 132:
3- (7- (4- ( (9- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) -3, 9-
diazaspiro [5.5] undecan-3-yl) methyl) piperidin-1-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (Cpd-148)
Scheme:
Step A: 7- (4- (dimethoxymethyl) piperidin-1-yl) phthalazin-1 (2H) -one
To a solution of 7-bromophthalazin-1 (2H) -one (567 mg, 2.52 mmol) in Dioxane (10 ml) was added 4- (dimethoxymethyl) piperidine (400 mg, 2.52 mmol) , Ruphos Pd-G2 (196 mg, 0.252 mmol) , t-BuONa (727 mg, 7.56 mmol) then the reaction mixture was stirred at 100 ℃ for 16 h under N2 atmosphere. After completion of reaction, quenched with H2O (10 mL) , extracted with EA (3*20 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =0: 1) to afford the desired product.
LCMS: MS (ESI) m/z 304.1 [M+H] +.
Step B: 3- (7- (4- (dimethoxymethyl) piperidin-1-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-
dione
To a solution of 7- (4- (dimethoxymethyl) piperidin-1-yl) phthalazin-1 (2H) -one (100 mg, 0.330 mmol) in DMF (10 ml) was added NaH (26.4 mg, 0.660 mmol) , 3-bromopiperidine-2, 6-dione (63.4 mg, 0.330 mmol) , then the reaction mixture was stirred at 25℃ for 16 h under N2 atmosphere. quenched with H2O (10 mL) , extracted with EA (3 × 10 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =1: 2) to afford the desired product.
LCMS: MS (ESI) m/z 415.2 [M+H] +.
Step C: 1- (3- (2, 6-dioxopiperidin-3-yl) -4-oxo-3, 4-dihydrophthalazin-6-yl) piperidine-4-
carbaldehyde
To a solution of 3- (7- (4- (dimethoxymethyl) piperidin-1-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (100 mg, 0.241 mmol) in FA (2 mL ) was stirred at 50 ℃ for 3 h under N2 atmosphere After completion of reaction, the reaction mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 369.2 [M+H] +.
Step D: 3- (7- (4- ( (9- (4- ( ( (R, Z) -12-hydroxy-12-methyl-5-oxo-7, 10, 11, 12-tetrahydro-5H-13, 17-
(azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-2-yl) amino) phenyl) -3, 9-
diazaspiro [5.5] undecan-3-yl) methyl) piperidin-1-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-
2, 6-dione
To a solution of 1- (3- (2, 6-dioxopiperidin-3-yl) -4-oxo-3, 4-dihydrophthalazin-6-yl) piperidine-4-carbaldehyde (19.5 mg, 0.053 mmol) , (R, Z) -2- ( (4- (3, 9-diazaspiro [5.5] undecan-3-yl) phenyl) amino) -12-hydroxy-12-methyl-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (30.0 mg, 0.053 mmol) in DCM (5 mL) and MeOH (5 mL) was stirred at 25 ℃ for 0.5 h. Then NaBH (OAc) 3 (16.7 mg, 0.080 mmol) was added. The mixture was stirred at 25 ℃ for 16 h. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 17-27%Retention Time: 9-19 of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 919.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.00 (s, 1H) , 10.02 (s, 1H) , 8.81 (s, 1H) , 8.26 (s, 1H) , 8.23 (s, 1H) , 8.02 (s, 1H) , 7.76 (d, J = 9.0 Hz, 2H) , 7.62 –7.51 (m, 4H) , 7.48 (s, 1H) , 6.90 (d, J = 9.1 Hz, 2H) , 5.76 (d, J = 5.9 Hz, 1H) , 5.55 –5.04 (m, 3H) , 4.44 (dd, J = 7.9, 2.5 Hz, 2H) , 4.12 –
3.86 (m, 2H) , 3.12 –3.04 (m, 4H) , 2.91 (dd, J = 21.5, 8.6 Hz, 3H) , 2.71 –2.58 (m, 4H) , 2.40 –2.28 (m, 5H) , 2.19 –2.05 (m, 4H) , 1.95 (dd, J = 13.4, 1.8 Hz, 1H) , 1.85 –1.69 (m, 4H) , 1.59 –1.53 (m, 6H) , 1.51 –1.43 (m, 4H) , 1.27 –1.08 (m, 2H) .
Example 133:
3- [7- (4- { [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) -4-fluoro-1-oxo-1, 2-dihydro-2-phthalazinyl] -2, 6-piperidinedione (Cpd-149)
Scheme:
Step A: 7- (4- (dimethoxymethyl) piperidin-1-yl) -4-fluorophthalazin-1 (2H) -one
To a solution of 7-bromophthalazin-1 (2H) -one (610 mg, 2.52 mmol) in Dioxane (10 ml) was added 4- (dimethoxymethyl) piperidine (400 mg, 2.52 mmol) , Ruphos Pd-G2 (196 mg, 0.252 mmol) , t-BuONa (727 mg, 7.56 mmol) then the reaction mixture was stirred at 100 ℃ for 16h under N2 atmosphere. After completion of reaction, quenched with H2O (10 mL) , extracted with EA (3*20 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =0: 1) to afford the desired product.
LCMS: MS (ESI) m/z 322.1 [M+H] +.
Step B: 3- (7- (4- (dimethoxymethyl) piperidin-1-yl) -4-fluoro-1-oxophthalazin-2 (1H) -
yl) piperidine-2, 6-dione
To a solution of 7- (4- (dimethoxymethyl) piperidin-1-yl) -4-fluorophthalazin-1 (2H) -one (106 mg, 0.330 mmol) in DMF (10 ml) was added NaH (26.4 mg, 0.660 mmol) , 3-bromopiperidine-2, 6-dione (63.4 mg, 0.330 mmol) , then the reaction mixture was stirred at 0 -25℃for 16h under N2 atmosphere. quenched with H2O (10 mL) , extracted with EA (3 × 10 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =1: 2) to afford the desired product.
LCMS: MS (ESI) m/z 433.0 [M+H] +.
Step C: 1- (3- (2, 6-dioxopiperidin-3-yl) -1-fluoro-4-oxo-3, 4-dihydrophthalazin-6-
yl) piperidine-4-carbaldehyde
To a solution of 3- (7- (4- (dimethoxymethyl) piperidin-1-yl) -4-fluoro-1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (100 mg, 0.231 mmol) in FA (2mL ) stirred at 50℃ for 3 hrs under N2 atmosphere. After completion of reaction, the reaction mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 387.2 [M+H] +.
Step D: 3- [7- (4- { [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) -4-fluoro-1-oxo-1, 2-
dihydro-2-phthalazinyl] -2, 6-piperidinedione
To a solution of 1- (3- (2, 6-dioxopiperidin-3-yl) -1-fluoro-4-oxo-3, 4-dihydrophthalazin-6-yl) piperidine-4-carbaldehyde (20.4 mg, 0.053 mmol) , (R, Z) -2- ( (4- (3, 9-diazaspiro [5.5] undecan-3-yl) phenyl) amino) -12-hydroxy-12-methyl-7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (30.0 mg, 0.053 mmol) in DCM (5 mL) and MeOH (5 mL) was stirred at 25 ℃ for 0.5 h. Then NaBH (OAc) 3 (16.7 mg, 0.080 mmol) was added. The mixture was stirred at 25 ℃ for 16h. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3×
10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 17-27%Retention Time: 9-19 of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 937.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.04 (s, 1H) , 10.03 (s, 1H) , 8.81 (s, 1H) , 8.26 (s, 1H) , 8.02 (s, 1H) , 7.79 –7.72 (m, 2H) , 7.62 (d, J = 9.6 Hz, 1H) , 7.57 (dd, J = 12.4, 8.4 Hz, 3H) , 7.51 (s, 1H) , 6.90 (d, J = 9.0 Hz, 2H) , 5.75 (dd, J = 11.6, 4.0 Hz, 1H) , 5.45 –5.12 (m, 3H) , 4.45 (m, 2H) , 4.05 (d, J = 12.6 Hz, 2H) , 3.08 (m, 4H) , 3.03 –2.96 (m, 3H) , 2.96 –2.86 (m, 2H) , 2.69 –2.58 (m, 4H) , 2.34 (m, 4H) , 2.16 (d, J = 6.4 Hz, 3H) , 2.12 –1.87 (m, 5H) , 1.82 (m, 3H) , 1.75 –1.68 (m, 1H) , 1.47 (s, 4H) , 1.20 –1.09 (m, 2H) .
Example 134:
3- (6- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -4-fluoro-1-oxo-1, 2-dihydro-2-phthalazinyl) -2, 6-piperidinedione (Cpd-146)
Scheme:
Step A: 4-fluoro-6- (2-oxo-8-azaspiro [4.5] decan-8-yl) phthalazin-1 (2H) -one
A mixture of 6-bromo-4-fluorophthalazin-1 (2H) -one (120 mg, 0.494 mmol) , 8-azaspiro [4.5] decan-2-one (113 mg, 0.741 mmol) , Pd-PEPPSI-IPentCl (24.1 mg, 0.0247 mmol) , Cs2CO3 (482 mg, 1.48 mmol) in dioxane (8.0 mL) was stirred at 100℃ for 2hrs under N2. After completion, the mixture was quenched with H2O (10 mL) , extracted with DCM (10 mLx3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 316.0 [M+H] +.
Step B: 4-fluoro-6- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-
yl) phthalazin-1 (2H) -one
To a mixture of 4-fluoro-6- (2-oxo-8-azaspiro [4.5] decan-8-yl) phthalazin-1 (2H) -one (75.0 mg, 0.238 mmol) , 1- (4-nitrophenyl) piperazine (74.0 mg, 0.357 mmol) in DMSO (2 mL) was added ZnCl2 (1 M in THF, 0.4 mL) . The mixture was stirred at 25℃ for 1 hr. Then NaBH3CN (59.8
mg, 0.952 mmol) in MeOH (0.6 mL) was added and stirred at 25℃ for 16 hrs. After completion of reaction, the cooled reaction mixture was filtered and the precipitated was dried in vacuo to afford the desired product.
LCMS: MS (ESI) m/z 507.1 [M+H] +.
Step C: 3- (4-fluoro-6- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-
oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione
A mixture of 4-fluoro-6- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) phthalazin-1 (2H) -one (110 mg, 0.217 mmol) in DMF (8.0 mL) was treated with NaH (60wt. %, 60.8 mg, 1.52 mmol) at 0℃ for 1 h. Then 3-bromopiperidine-2, 6-dione (417 mg, 2.17 mmol) was added. The reaction was stirred at 25℃ under N2 atmosphere for 4hrs. After completion, the mixture was diluted with H2O (5 mL) , extracted with EtOAc (10 mLx3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 618.1 [M+H] +.
Step D: 3- (6- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -4-fluoro-1-
oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione
A mixture of 3- (4-fluoro-6- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (100 mg, 0.162 mmol) , iron powder (45.2 mg, 0.810 mmol) , NH4Cl (43.3 mg, 0.810 mmol) in EtOH (10 mL) and H2O (2 mL) was stirred at 90℃ for 2hrs. After completion of reaction, the mixture reaction was filtered through a Celite pad, and the filtrate was concentrated to give the crude. The crude was quenched with H2O (10 mL) , adjusted to pH 8 with solid NaHCO3 and extracted with EtOAc (3 x 30 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 588.2 [M+H] +.
Step E: 3- (6- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -4-fluoro-1-oxo-1, 2-dihydro-2-
phthalazinyl) -2, 6-piperidinedione
To a solution of (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20.0 mg, 0.0519 mmol) in DCM (3 mL) was added 3- (6- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -4-fluoro-1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (70.0 mg, 0.119 mmol) and n-BuOH (12 mL) . Then the reaction mixture was stirred at 100℃ for 16 hrs
under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EA (20 mL x3) . The organic layers were concentrated. The residue was purified by prep-HPLC (Waters 2767, Column: Pursuit XRs C18, 21.2*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 15%~25%Retention Time: 9.0-10.0 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 909.1 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.03 (s, 1H) , 10.05 (s, 1H) , 8.82 (s, 1H) , 8.21 –8.17 (m, 1H) , 8.05 (d, J = 9.2 Hz, 2H) , 7.75 (d, J = 6.4 Hz, 1H) , 7.57 (m, 4H) , 7.10 (s, 1H) , 6.90 (d, J = 6.9 Hz, 2H) , 5.72 (d, J = 12.1 Hz, 1H) , 5.44 –5.20 (m, 3H) , 4.44 (s, 2H) , 3.52 –3.46 (m, 4H) , 3.10 –3.03 (m, 4H) , 2.96 –2.85 (m, 2H) , 2.64 –2.57 (m, 4H) , 2.55 (m, 3H) , 2.20 –2.05 (m, 3H) , 1.97 –1.82 (m, 4H) , 1.73 –1.60 (m, 3H) , 1.55 (m, 3H) , 1.52 (d, J = 8.8 Hz, 2H) , 1.40 –1.19 (m, 2H) .
Example 135:
3- (6- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -1-oxo-2-phthalazinyl) -2, 6-piperidinedione (Cpd-144)
Scheme:
Step A: 6- (2-oxo-8-azaspiro [4.5] decan-8-yl) phthalazin-1 (2H) -one
A mixture of 6-bromophthalazin-1 (2H) -one (165 mg, 0.741 mmol) , Pd-PEPPSI-IPentCl (24.1 mg, 0.0247 mmol) , Cs2CO3 (482 mg, 1.48 mmol) in dioxane (8.0 mL) was stirred at 100℃ for 2 hrs under N2. After completion, the mixture was quenched with H2O (10 mL) , extracted with DCM (10 mLx3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 298.1 [M+H] +.
Step B: 6- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) phthalazin-1 (2H) -
one
To a mixture of 6- (2-oxo-8-azaspiro [4.5] decan-8-yl) phthalazin-1 (2H) -one (71 mg, 0.238 mmol) , 1- (4-nitrophenyl) piperazine (74.0 mg, 0.357 mmol) in DMSO (2 mL) was added ZnCl2 (1 M in THF, 0.4 mL) . The mixture was stirred at 25℃ for 1 hr. Then NaBH3CN (59.8 mg, 0.952 mmol) in MeOH (0.6 mL) was added and stirred at 25℃ for 16 hrs. After completion of reaction, the cooled reaction mixture was filtered and the precipitated was dried in vacuo to afford the desired product.
LC-MS (ESI) [M+H] + = 489.1
Step C: 3- (6- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-
oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione
A mixture of 6- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) phthalazin-1 (2H) -one (105 mg, 0.217 mmol) in DMF (8.0 mL) was treated with NaH (60wt. %, 60.8 mg, 1.52 mmol) at 0℃ for 1 h. Then 3-bromopiperidine-2, 6-dione (417 mg, 2.17 mmol) was added. The reaction was stirred at 25℃ under N2 atmosphere for 16 hrs. After completion, the mixture was diluted with H2O (5 mL) , extracted with EtOAc (10 mLx3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LC-MS (ESI) [M+H] + = 600.1
Step D: 3- (6- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-
oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione
A mixture of 3- (6- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (97 mg, 0.162 mmol) , iron powder (45.2 mg, 0.810 mmol) , NH4Cl (43.3 mg, 0.810 mmol) in EtOH (10 mL) and H2O (2 mL) was stirred at 90℃ for 3 hrs. After completion of reaction, the mixture reaction was filtered through a Celite pad, and the filtrate was concentrated to give the crude. The crude was quenched with H2O (10 mL) , adjusted to pH 8 with solid NaHCO3 and extracted with EtOAc (3 x 30 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LC-MS (ESI) [M+H] + = 570.1
Step E: 3- (6- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -1-oxo-2-phthalazinyl) -2, 6-
piperidinedione
To a solution of (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20.0 mg, 0.0519 mmol) in DCM (3 mL) was added 3- (6- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (70.0 mg, 0.119 mmol) . Then the reaction mixture was stirred at 25℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EA (20 mL x3) . The organic layers were concentrated. The residue was purified by prep-HPLC (Waters 2767, Column: Pursuit XRs C18, 21.2*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 15%~25%Retention Time: 9.0-10.0 min of 16 min) to afford the desired product.
LC-MS (ESI) [M+H] + =891.2
1H NMR (400 MHz, CDCl3) δ 8.85 (s, 1H) , 8.23 (d, J = 8.9 Hz, 1H) , 8.11 (s, 1H) , 8.02 (s, 1H) , 7.86 (d, J = 7.6 Hz, 1H) , 7.79 (s, 1H) , 7.49 (d, J = 8.6 Hz, 2H) , 7.31 (d, J = 9.1 Hz, 1H) , 6.97 –6.87 (m, 3H) , 5.75 (dt, J = 47.1, 17.2 Hz, 3H) , 4.50 (t, J = 14.5 Hz, 1H) , 4.42 –3.97 (m, 2H) , 3.42 (m, 4H) , 3.32 (m, 4H) , 2.95 –2.83 (m, 5H) , 2.81 –2.72 (m, 2H) , 2.30 (dt, J = 17.4, 11.2 Hz, 2H) , 2.19 –2.10 (m, 2H) , 2.08 –1.93 (m, 5H) , 1.71 –1.57 (m, 10H) .
Example 136:
3- (6- {9- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -3, 9-diaza-3-spiro [5.5] undecyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-339)
Scheme:
Step A: tert-butyl 9- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] -3, 9-
diazaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 3, 9-diazaspiro [5.5] undecane-3-carboxylate (500 mg, 1.97 mmol) in DMF (5 mL) was added DIPEA (508.10 mg, 3.93 mmol) and 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (542.95 mg, 1.97 mmol) . The mixture was stirred at 100 ℃ for 4 hr.The mixture was added H2O (50 mL) , extracted with EtOAc (50 mL × 3) . The combined organic layers were washed with brine (15 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 511.3 [M+H] +.
Step B: 5- (3, 9-diazaspiro [5.5] undecan-3-yl) -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-
dione
To a solution of tert-butyl 9- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] -3, 9-diazaspiro [5.5] undecane-3-carboxylate (1.2 g, 2.35 mmol) in DCM (10 mL) was added HCl/dioxane (4 M) . The mixture was stirred at 20 ℃ for 0.5 hr. The mixture was filtered and the cake was dried in vacuo to afford the desired product.
LCMS: MS (ESI) m/z 411.3 [M+H] +.
Step C: 2- (2, 6-dioxo-3-piperidyl) -5- [3- [1- (4-nitrophenyl) -4-piperidyl] -3, 9-
diazaspiro [5.5] undecan-9-yl] isoindoline-1, 3-dione
To a solution of 5- (3, 9-diazaspiro [5.5] undecan-3-yl) -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (1 g, 2.44 mmol) , 1- (4-nitrophenyl) piperidin-4-one (536.52 mg, 2.44 mmol) in DCM (10 mL) was added Et3N (986.09 mg, 9.75 mmol) . The mixture was stirred at 20 ℃ for 4 hr and then NaBH (OAc) 3 (1.03 g, 4.87 mmol) was added. The mixture was stirred at 20℃ for 12 hr. The mixture was added H2O (30 mL) , extracted with DCM (30 mL × 3) . The combined organic layers were washed with brine (15 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (40 g Silica Flash Column, Eluent of 0~10 %Methanol/Dichloromethane gradient @30 mL/min) and the combined desired fractions was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 615.3 [M+H] +.
Step D: 3- [6- [3- [1- (4-aminophenyl) -4-piperidyl] -3, 9-diazaspiro [5.5] undecan-9-yl] -1-oxo-
isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [3- [1- (4-nitrophenyl) -4-piperidyl] -3, 9-diazaspiro [5.5] undecan-9-yl] isoindoline-1, 3-dione (450 mg, 732.08 μmol) in TFA (4.5 mL) was added Zn (3.85 g, 58.88 mmol) slowly. The mixture was stirred at 75℃ for 1hr. The mixture was filtered through diatomite pad under vacuum and washed with CH3CN (30 mL) and the filtrate was evaporated in vacuum to a residue. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-21%B over 7.5 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 571.4 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.97 (s, 1H) , 9.41 (br s, 1H) , 7.48 (d, J = 8.4 Hz, 1H) , 7.39 -7.28 (m, 2H) , 7.19 (br d, J = 8.8 Hz, 2H) , 7.08 (br d, J = 8.8 Hz, 2H) , 5.09 (dd, J = 5.2, 13.2 Hz, 1H) , 4.36 (d, J = 16.8 Hz, 1H) , 4.23 (d, J = 16.8 Hz, 1H) , 3.88 (br d, J = 12.4 Hz, 2H) , 3.50 -3.34 (m, 3H) , 3.33 -3.21 (m, 4H) , 3.10 -3.00 (m, 2H) , 2.98 -2.84 (m, 1H) , 2.76 (br t, J = 12.0
Hz, 2H) , 2.60 (br d, J = 16.8 Hz, 1H) , 2.45 -2.35 (m, 1H) , 2.16 (br d, J = 11.8 Hz, 2H) , 2.05 -1.88 (m, 3H) , 1.86 -1.67 (m, 4H) , 1.66 -1.48 (m, 5H)
Step E: 3- (6- {9- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -3, 9-diaza-3-spiro [5.5] undecyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 3- [6- [3- [1- (4-aminophenyl) -4-piperidyl] -3, 9-diazaspiro [5.5] undecan-9-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (50 mg, 73.02 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (30 mg, 77.83 μmol) in n-BuOH (5 mL) was added AcOH (43.85 mg, 730.20 μmol) . The mixture was stirred at 100 ℃ for 2 hr. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: Agela DuraShell 150mm_25mm_5um; mobile phase: [water (0.05%HCl) -ACN] ; B%: 0%-31%, 10min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 892.8 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.96 (s, 1H) , 10.02 (br s, 1H) , 8.82 (s, 1H) , 8.19 (s, 1H) , 8.10 -7.95 (m, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.62 -7.50 (m, 3H) , 7.41 (d, J = 8.8 Hz, 1H) , 7.24 (br d, J = 8.4 Hz, 1H) , 7.15 (s, 1H) , 6.91 (br d, J = 9.2 Hz, 2H) , 5.40 -5.25 (m, 3H) , 5.09 (dd, J = 5.2, 13.2 Hz, 1H) , 4.44 (br s, 2H) , 4.32 (br d, J = 16.8 Hz, 1H) , 4.20 (br d, J = 16.8 Hz, 1H) , 3.68 (br d, J = 10.8 Hz, 2H) , 3.23 -3.08 (m, 4H) , 2.97 -2.84 (m, 1H) , 2.65 -2.53 (m, 7H) , 2.43 -2.31 (m, 2H) , 2.25 -2.13 (m, 1H) , 2.08 -1.90 (m, 3H) , 1.86 (br d, J = 11.6 Hz, 2H) , 1.76 -1.66 (m, 1H) , 1.65 -1.53 (m, 9H) , 1.52-1.45 (m, 4H)
Example 137:
3- (5- {9- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -3, 9-diaza-3-spiro [5.5] undecyl} -1, 3-dioxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-340)
Scheme:
Step A: tert-butyl 9- [1- (4-nitrophenyl) -4-piperidyl] -3, 9-diazaspiro [5.5] undecane-3-
carboxylate
A mixture of 1- (4-nitrophenyl) piperidin-4-one (190.47 mg, 864.89 μmol) , tert-butyl 3, 9-diazaspiro [5.5] undecane-3-carboxylate (200 mg, 786.26 μmol) and Et3N (181.75 mg, 1.80 mmol) in DCM (5 mL) was stirred at 25 ℃ for 1 hr, then NaBH (OAc) 3 (520 mg, 2.45 mmol) was added and the mixture was stirred for another 1 hr at 25℃. The mixture was concentrated to give a residue. The residue was purified by flash silica gel chromatography (12 g Silica Flash Column, Eluent of 0~10 %Methanol/Dichloromethane gradient @30 mL/min) and the combined desired fractions was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 459.4 [M+H] +.
Step B: 3- [1- (4-nitrophenyl) -4-piperidyl] -3, 9-diazaspiro [5.5] undecane
To a solution of tert-butyl 9- [1- (4-nitrophenyl) -4-piperidyl] -3, 9-diazaspiro [5.5] undecane-3-carboxylate (390 mg, 850.43 μmol) in DCM (4 mL) was added HCl/dioxane (4 M, 212.61 μL) . The mixture was stirred at 25℃ for 1 hr. The mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 359.3 [M+H] +.
Step C: 2- (2, 6-dioxo-3-piperidyl) -5- [3- [1- (4-nitrophenyl) -4-piperidyl] -3, 9-
diazaspiro [5.5] undecan-9-yl] isoindoline-1, 3-dione
To a solution of 3- [1- (4-nitrophenyl) -4-piperidyl] -3, 9-diazaspiro [5.5] undecane (100 mg, 253.20 μmol) , 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (90 mg, 325.83 μmol) in DMF (2 mL) was added DIPEA (98.17 mg, 759.61 μmol) . The mixture was stirred at 100 ℃for 1 hr. The mixture was cooled down to 25℃ and the mixture was poured into H2O (200 mL) . The mixture was filtered and the filter cake was dried under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 615.4 [M+H] +.
Step D: 5- [3- [1- (4-aminophenyl) -4-piperidyl] -3, 9-diazaspiro [5.5] undecan-9-yl] -2- (2, 6-
dioxo-3-piperidyl) isoindoline-1, 3-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [3- [1- (4-nitrophenyl) -4-piperidyl] -3, 9-diazaspiro [5.5] undecan-9-yl] isoindoline-1, 3-dione (130 mg, 211.49 μmol) in DCM (5 mL) and DMAC (1 mL) was added PdCl2 (37.50 mg, 211.49 μmol) and Et3SiH (245.91 mg, 2.11 mmol) , Et3N (42.80 mg, 422.98 μmol) . The mixture was stirred at 25 ℃ for 4hr. The mixture filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Welch Xtimate C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 4%-34%B over 7 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 585.5 [M+H] +.
Step E: 3- (5- {9- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -3, 9-diaza-3-spiro [5.5] undecyl} -1, 3-dioxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 5- [3- [1- (4-aminophenyl) -4-piperidyl] -3, 9-diazaspiro [5.5] undecan-9-yl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (20 mg, 28.62 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (11.03 mg, 28.62 μmol) in n-BuOH (1 mL) was added AcOH (17.19 mg, 286.23 μmol) . The mixture was stirred at 100 ℃ for 2 hr. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: C18 150×30mm; mobile phase: [water (FA) -ACN] ; gradient: 21%-51%B over 6 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 906.8 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 11.07 (s, 1H) , 10.04 (br s, 1H) , 8.82 (s, 1H) , 8.19 (s, 1H) , 8.02 (t, J = 7.2 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.65 (d, J = 8.4 Hz, 1H) , 7.57 (dd, J = 8.4, 12.0 Hz, 3H) , 7.30 (s, 1H) , 7.22 (br d, J = 8.4 Hz, 1H) , 6.91 (br d, J =9.2 Hz, 2H) , 5.32 (m, 3H) , 5.06 (dd, J = 5.2, 12.8 Hz, 1H) , 4.46 (br s, 2H) , 3.68 (br d, J =11.2 Hz, 2H) , 3.50 -3.40 (m, 6H) , 2.92 -2.83 (m, 1H) , 2.65 -2.59 (m, 2H) , 2.57 -2.52 (m, 4H) , 2.45 -2.35 (m, 2H) , 2.26 -2.12 (m, 1H) , 2.07 -1.79 (m, 5H) , 1.76 -1.67 (m, 1H) , 1.53 (m, 12H)
Example 138:
3- (5- {2- [1- (p- { (R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1, 3-dioxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-341)
Scheme:
Step A: 5- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -2- (2, 6-dioxo-
3-piperidyl) isoindoline-1, 3-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione (230 mg, 382.91 μmol) in DCM (5 mL) and DMAC (1 mL) was added PdCl2 (67.90 mg, 382.91 μmol) , Et3N (77.49 mg, 765.82 μmol) and Et3SiH (445.24 mg, 3.83 mmol, 611.59 μL) . The mixture was stirred at 20 ℃ for 4 hr. The mixture was concentrated to give residue. The residue was purified by prep-HPLC (column: Welch Xtimate C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 3%-33%B over 7 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 571.4 [M+H] +.
Step B: 3- (5- {2- [1- (p- { (R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1, 3-dioxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 5- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (60 mg, 87.63 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (33.78 mg, 87.63 μmol) in n-BuOH (3 mL) was added AcOH (52.62 mg, 876.29 μmol) . The mixture was stirred at 100 ℃ for 2 hr. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: C18 150×30mm; mobile phase: [water (FA) -ACN] ; gradient: 22%-52%B over 6 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 892.7 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ11.07 (s, 1H) , 10.04 (br s, 1H) , 8.81 (s, 1H) , 8.18 (s, 1H) , 8.01 (br t, J = 7.6 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.65 (d, J = 8.4 Hz, 1H) , 7.61 -7.51 (m, 3H) , 7.32 (s, 1H) , 7.24 (br d, J = 8.8 Hz, 1H) , 6.91 (br d, J = 9.2 Hz, 2H) , 5.37 (br s, 2H) , 5.06 (dd, J = 5.2, 12.8 Hz, 1H) , 4.45 (br d, J = 6.0 Hz, 2H) , 3.48 -3.38 (m, 6H) , 2.95 -2.81 (m, 2H) , 2.77 (br t, J = 6.4 Hz, 2H) , 2.68 (br t, J = 11.2 Hz, 2H) , 2.61 (br s, 2H) , 2.59 -2.52 (m, 2H) , 2.36 -2.25 (m, 1H) , 2.17 (br s, 1H) , 2.08 -1.98 (m, 2H) , 1.98 -1.87 (m, 3H) , 1.79 -1.45 (m, 12H)
Example 139:
3- (6- {8- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-2-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-342)
Scheme:
Step A: tert-butyl 8- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decane-2-
carboxylate
To a solution of 1- (4-nitrophenyl) piperidin-4-one (1.83 g, 8.32 mmol) and tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (2 g, 8.32 mmol) in DCM (40 mL) was added AcOH (499.72 mg, 8.32 mmol) . The mixture was stirred at 25 ℃ for 14hr . Then the mixture was added NaBH (OAc) 3 (5.29 g, 24.96 mmol) and stirred at 25 ℃ for 14hr. The reaction mixture was quenched with water, and then extracted with DCM (150mL ×3) . The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (80g Silica Flash Column, Eluent of 0~10%DCM/MeOH gradient @60 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 445.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 8.03 (d, J = 9.6 Hz, 2H) , 7.02 (d, J = 9.6 Hz, 2H) , 4.07 (br d, J = 13.2 Hz, 2H) , 3.26 (br s, 2H) , 3.04 (br s, 2H) , 2.97 (br t, J = 12.0 Hz, 2H) , 2.72 -2.53 (m, 3H) , 1.91 (m, 2H) , 1.90 -1.81 (m, 2H) , 1.65 (br d, J = 6.4 Hz, 2H) , 1.49 (m, 6H) , 1.38 (s, 9H)
Step B: 8- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decane
To a solution of tert-butyl 8- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decane-2-carboxylate (0.85 g, 1.91 mmol) in DCM (8 mL) was added HCl/dioxane (4 M, 8.29 mL) . The mixture was stirred at 25 ℃ for 1hr. The crude product was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 345.3 [M+H] +.
Step C: 2- (2, 6-dioxo-3-piperidyl) -5- [8- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-
diazaspiro [4.5] decan-2-yl] isoindoline-1, 3-dione
To a solution of 8- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decane (650 mg, 1.71 mmol) in DMF (6 mL) was added DIPEA (661.63 mg, 5.12 mmol) and 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (530 mg) . The mixture was stirred at 100 ℃ for 2hr. The mixture was added H2O (50 mL) , extracted with EtOAc (50 mL *3) . The combined organic layers were washed with brine (30 mL *3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (40 gSilica Flash Column, Eluent of 0~10 %Methanol/Dichloromethane gradient @30 mL/min) and the combined desired fractions was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 601.5 [M+H] +.
Step D: 3- [5- [8- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-2-yl] -1-oxo-
isoindolin-2-yl] piperidine-2, 6-dione & 3- [6- [8- [1- (4-aminophenyl) -4-piperidyl] -2, 8-
diazaspiro [4.5] decan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [8- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-2-yl] isoindoline-1, 3-dione (880 mg, 1.47 mmol) in TFA (10 mL) was added Zn (3.82 g, 58.42 mmol) . The mixture was stirred at 75 ℃ for 2hr. The reaction mixture was filtered, washed with MeCN (15mL × 3) and concentrated to give crude product. The crude product was purified by reversed-phase HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-28%B over 8 min) and dried by lyophilization to give the product. 3- [5- [8- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
LCMS: MS (ESI) m/z 556.9 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.93 (s, 1H) , 9.61 -9.48 (m, 1H) , 7.50 (br t, J = 8.8 Hz, 1H) , 7.19 (br d, J = 8.8 Hz, 2H) , 7.08 (br d, J = 8.8 Hz, 2H) , 6.65 (br d, J = 5.2 Hz, 2H) , 5.03 (br dd,
J = 3.6, 12.8 Hz, 1H) , 4.35 -4.25 (m, 1H) , 4.24 -4.14 (m, 1H) , 3.89 (br d, J = 12.0 Hz, 2H) , 3.55 -3.39 (m, 5H) , 3.20 (br s, 1H) , 3.11 (br s, 2H) , 3.00 -2.84 (m, 1H) , 2.84 -2.71 (m, 2H) , 2.58 (br d, J = 17.2 Hz, 2H) , 2.45 -2.28 (m, 2H) , 2.16 (br d, J = 10.4Hz, 2H) , 2.09 -2.01 (m, 2H) , 1.99 -1.81 (m, 6H) , 1.75 (br d, J = 12.0 Hz, 2H)
3- [6- [8- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
LCMS: MS (ESI) m/z 557.2 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.96 (s, 1H) , 9.60 -9.41 (m, 1H) , 7.42 -7.35 (m, 1H) , 7.19 (br d, J = 8.4 Hz, 2H) , 7.09 (br d, J = 8.8 Hz, 2H) , 6.88 -6.76 (m, 2H) , 5.09 (m, 1H) , 4.30 (d, J = 16.8Hz, 1H) , 4.21 (d, J = 16.8Hz, 1H) , 3.89 (d, J = 12.0 Hz, 2H) , 3.49 (br d, J = 11.2 Hz, 2H) , 3.43 -3.35 (m, 3H) , 3.16 (br d, J = 18.0 Hz, 3H) , 2.96 -2.85 (m, 1H) , 2.78 (br t, J = 12.0 Hz, 2H) , 2.59 (br d, J = 17.6 Hz, 2H) , 2.47 -2.30 (m, 2H) , 2.16 (br d, J = 10.8 Hz, 2H) , 2.09 -1.94 (m, 3H) , 1.92 -1.81 (m, 5H) , 1.80 -1.69 (m, 2H)
Step E: 3- (6- {8- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-2-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 3- [6- [8- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (57.77 mg, 103.78 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (40 mg, 103.78 μmol) in n-BuOH (2 mL) was added AcOH (62.32 mg, 1.04 mmol) and the mixture was stirred at 100 ℃ for12hr. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 10%-40%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 878.8 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.96 (s, 1H) , 10.18 -9.95 (m, 1H) , 9.38 -9.23 (m, 1H) , 8.83 (s, 1H) , 8.01 (br t, J = 7.6 Hz, 1H) , 7.75 (d, J = 8.1 Hz, 1H) , 7.59 (br d, J = 7.6 Hz, 3H) , 7.39 (dd, J = 4.4, 8.2 Hz, 1H) , 6.99 (br d, J = 9.2 Hz, 2H) , 6.89 -6.73 (m, 2H) , 5.44 -5.23 (m, 2H) , 5.09 (td, J = 4.7, 13.2 Hz, 1H) , 4.53 -4.41 (m, 2H) , 4.31 (d, J = 16.8 Hz, 1H) , 4.19 (d, J = 16.8 Hz, 1H) , 3.83 -3.81 (m, 2H) , 3.50 (br d, J = 11.6 Hz, 2H) , 3.44 -3.28 (m, 5H) , 3.22 -3.06 (m, 3H) , 2.97 -2.84 (m, 1H) , 2.78 -2.66 (m, 2H) , 2.59 (br d, J = 16.4 Hz, 1H) , 2.44 -2.30 (m, 1H) , 2.25 -2.10 (m, 3H) , 2.07 -1.68 (m, 12H) , 1.56 (s, 3H)
Example 140:
3- (5- {8- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-2-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-343)
Scheme:
Step A: 3- (5- {8- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-2-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 3- [5- [8- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-2-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (57.77 mg, 103.78 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (40 mg, 103.78 μmol) in n-BuOH (2 mL) was added AcOH (62.32 mg, 1.04 mmol) and the mixture was stirred at 100 ℃ for 12hr. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -
ACN] ; gradient: 9%-39%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 878.8 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.94 (s, 1H) , 10.16 -9.96 (m, 1H) , 9.31 (m, 1H) , 8.83 (s, 1H) , 8.01 (br t, J = 8.0 Hz, 1H) , 7.75 (d, J = 8.2 Hz, 1H) , 7.59 (br d, J = 7.6 Hz, 3H) , 7.51 (t, J = 8.8 Hz, 1H) , 6.97 (br d, J = 9.2 Hz, 2H) , 6.69 -6.58 (m, 2H) , 5.43 -5.24 (m, 2H) , 5.03 (br dd, J =12.4 Hz, 1H) , 4.46 (br d, J = 5.6 Hz, 2H) , 4.30 (dd, J = 16.8 Hz, 1H) , 4.19 (dd, J = 16.8 Hz, 1H) , 3.82 (br d, J = 12.4 Hz, 2H) , 3.21 (m, 2H) , 3.17 -3.03 (m, 3H) , 2.97 -2.82 (m, 2H) , 2.70 (br t, J = 12.0 Hz, 3H) , 2.62 -2.54 (m, 2H) , 2.43 -2.28 (m, 2H) , 2.23 -2.10 (m, 3H) , 2.09 -1.64 (m, 13H) , 1.56 (s, 3H)
Example 141:
3- (6- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -5-fluoro-1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-344)
Scheme:
Step A: tert-butyl 8- [2- (2, 6-dioxo-3-piperidyl) -6-fluoro-1, 3-dioxo-isoindolin-5-yl] -2, 8-
diazaspiro [4.5] decane-2-carboxylate
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5, 6-difluoro-isoindoline-1, 3-dione (1.22 g, 4.16 mmol) , tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (1 g, 4.16 mmol) in DMF (12 mL) was added DIEA (1.61 g, 12.48 mmol) . The mixture was stirred at 100 ℃ for 2hr. The mixture was concentrated to give a residue, which was purified by silica gel column chromatography (3%MeOH in DCM) to afford the desired product.
1H NMR (400 MHz, DMSO-d6) δ 8.06 (s, 1H) , 7.48 (d, J = 11.2 Hz, 1H) , 7.45 (d, J = 7.2 Hz, 1H) , 4.94 (dd, J = 5.2, 12.3 Hz, 1H) , 3.44 (t, J = 7.2 Hz, 2H) , 3.37 -3.29 (m, 2H) , 3.27 (m, 2H) , 3.24 -3.16 (m, 2H) , 2.88 -2.69 (m, 3H) , 2.20 -2.09 (m, 1H) , 1.83 -1.74 (m, 6H) , 1.48 (s, 9H) .
Step B: 5- (2, 8-diazaspiro [4.5] decan-8-yl) -2- (2, 6-dioxo-3-piperidyl) -6-fluoro-isoindoline-1, 3-
dione
To a solution of tert-butyl 8- [2- (2, 6-dioxo-3-piperidyl) -6-fluoro-1, 3-dioxo-isoindolin-5-yl] -2, 8-diazaspiro [4.5] decane-2-carboxylate (1.5 g, 2.92 mmol) in DCM (10 mL) was added TFA (3.45 g, 30.29 mmol) . The mixture was stirred at 20 ℃ for 2 hr. The reaction mixture was poured into water (50 mL) and adjusted to pH=8 with aq. Na2CO3 (50 mL) . Then extracted with petroleum ether (100 mL × 2) . The combined organic layer was washed with brine (100 mL) , dried over anhydrous Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 415.2 [M+H] +.
Step C: 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-6- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-
diazaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione
To a solution of 5- (2, 8-diazaspiro [4.5] decan-8-yl) -2- (2, 6-dioxo-3-piperidyl) -6-fluoro-isoindoline-1, 3-dione (1.5 g, 3.62 mmol) , 1- (4-nitrophenyl) piperidin-4-one (797.09 mg, 3.62 mmol) in EtOH (20 mL) was added dropwise AcOH (108.68 mg, 1.81 mmol) . After addition, the mixture was stirred at 50 ℃ for 1 hr, and then NaBH3CN (682.36 mg, 10.86 mmol) was added. The resulting mixture was stirred at 50 ℃ for 1.5 hr. The mixture was poured into water (100 mL) and extracted with EtOAc (200 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by silica gel column chromatography (5%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 619.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.10 (s, 1H) , 8.07 (d, J = 9.6 Hz, 2H) , 7.73 (d, J = 11.2 Hz, 1H) , 7.46 (d, J = 7.6 Hz, 1H) , 7.09 (d, J = 9.2 Hz, 2H) , 5.10 (dd, J = 5.2, 12.8 Hz, 1H) , 4.17 (s, 1H) , 3.31 (m, 4H) , 3.26 (m, 4H) , 3.17 (d, J = 5.2 Hz, 3H) , 3.03 (t, J = 12.4 Hz, 2H) , 2.94 -2.83 (m, 1H) , 2.65 -2.54 (m, 3H) , 2.18 -1.97 (m, 3H) , 1.85 -1.65 (m, 5H) , 1.61 -1.49 (m, 1H) .
Step D: 3- [6- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -5-fluoro-1-
oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-6- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione (1.7 g, 2.75 mmol) in TFA (17 mL) was added Zn (7.42 g, 113.47 mmol) . The mixture was stirred at 75 ℃ for 2 hr. The reaction mixture was filtered, washed with EtOAc (300mL) and concentrated to give crude product, which was purified by prep-HPLC (column: C18 100 × 40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-25%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 575.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.97 (s, 1H) , 10.23 (s, 1H) , 7.47 -7.39 (m, 1H) , 7.35 (d, J = 8.0 Hz, 1H) , 7.19 (d, J = 8.8 Hz, 2H) , 7.08 (d, J = 9.2 Hz, 2H) , 5.08 (dd, J = 4.8, 13.2 Hz, 1H) , 4.44 -4.19 (m, 2H) , 3.86 (d, J = 12.4 Hz, 2H) , 3.67 (s, 1H) , 3.57 (d, J = 10.0 Hz, 1H) , 3.32 (d, J = 7.2 Hz, 2H) , 3.15 -2.99 (m, 5H) , 2.97 -2.83 (m, 1H) , 2.76 (t, J = 12.0 Hz, 2H) , 2.60 (d, J = 17.2 Hz, 1H) , 2.45 -2.31 (m, 1H) , 2.15 (d, J = 8.4 Hz, 2H) , 2.06 -1.93 (m, 2H) , 1.90 -1.62 (m, 7H) .
Step E: 3- (6- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -5-fluoro-1-oxo-2-isoindolinyl) -
2, 6-piperidinedione
A mixture of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) , 3- [6- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -5-fluoro-1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (74.55 mg, 129.72 μmol) , AcOH (77.90 mg, 1.30 mmol, 74.26 μL) in n-BuOH (3 mL) was stirred at 100 ℃ for 16hr. The mixture was concentrated to give a residue, which was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 9%-39%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 896.7 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H) , 10.06 (TFA, 0.77H) , 9.80 (s, 1H) , 8.84 (s, 1H) , 8.02 (t, J = 8.0 Hz, 1H) , 7.76 (d, J = 8.0 Hz, 1H) , 7.60 (d, J = 8.0 Hz, 3H) , 7.49 -7.42 (m, 1H) , 7.37 (d, J = 8.0 Hz, 1H) , 6.99 (d, J = 8.8 Hz, 2H) , 5.23 -5.46 (m, 2H) , 5.09 (dd, J = 5.2, 13.2 Hz, 1H) , 4.47 (d, J = 6.0 Hz, 3H) , 4.42 -4.22 (m, 2H) , 3.82 (d, J = 12.4 Hz, 2H) , 3.67 (s, 1H) , 3.61 -3.51 (m, 1H) , 3.25 -3.40 (m, 2H) , 3.22 -2.99 (m, 5H) , 2.98 -2.83 (m, 1H) , 2.71 (t, J = 12.4 Hz, 2H) , 2.61 (d, J = 17.6 Hz, 1H) , 2.47 -2.32 (m, 1H) , 2.10 -2.25 (m, 3H) , 2.08 -1.91 (m, 4H) , 1.90 -1.65 (m, 8H) , 1.57 (s, 3H) .
19F NMR (400 MHz, DMSO-d6) δ = –116.466.
Example 142:
3- (7- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2-phthalazinyl) -2, 6-piperidinedione (Cpd-345)
Scheme:
Step A: tert-butyl 8- (4-oxo-3H-phthalazin-6-yl) -2, 8-diazaspiro [4.5] decane-2-carboxylate
To a solution of 7-bromo-2H-phthalazin-1-one (1 g, 4.44 mmol) , tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (1.07 g, 4.44 mmol) in dioxane (20 mL) was added 1, 3-bis[2, 6-bis (1-ethylpropyl) phenyl] -2Himidazole; 3-chloropyridine; dichloropalladium (176.34 mg, 222.18 μmol) and t-BuONa (1.28 g, 13.33 mmol) under N2. The mixture was stirred at 100 ℃ for 16 h. The reaction mixture was added H2O (100 mL) at 0 ℃, extracted with ethyl acetate (50 mL × 3) . The combined organic phases were dried with anhydrous Na2SO4, filtered
and concentrated in under reduce pressure. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate = 100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 385.3 [M+H] +.
Step B: tert-butyl 8- [3- [1- [ (4-methoxyphenyl) methyl] -2, 6-dioxo-3-piperidyl] -4-oxo-
phthalazin-6-yl] -2, 8-diazaspiro [4.5] decane-2-carboxylate
To a solution of tert-butyl 8- (4-oxo-3H-phthalazin-6-yl) -2, 8-diazaspiro [4.5] decane-2-carboxylate (400 mg, 1.04 mmol) in THF (8 mL) was added t-BuOK (140.09 mg, 1.25 mmol) at 0 ℃, the reaction solution was stirred for 30 min at this temperature, then to this was added [1- [ (4-methoxyphenyl) methyl] -2, 6-dioxo-3-piperidyl] trifluoromethanesulfonate (436.40 mg, 1.14 mmol) in THF (4 mL) dropwise. The resulting solution was stirred at 0 ℃ for 2 h. The mixture quenched with saturated aq. sat. NH4Cl (50 mL) . The aqueous layer was extracted with EtOAc (3 × 60 mL) and the combined organic layers were dried with anhydrous Na2SO4, filtered then concentrated to afford the desired product.
LCMS: MS (ESI) m/z 616.4 [M+H] +.
Step C: 3- [7- (2, 8-diazaspiro [4.5] decan-8-yl) -1-oxo-phthalazin-2-yl] -1- [ (4-
methoxyphenyl) methyl] piperidine-2, 6-dione
To a solution of tert-butyl 8- [3- [1- [ (4-methoxyphenyl) methyl] -2, 6-dioxo-3-piperidyl] -4-oxo-phthalazin-6-yl] -2, 8-diazaspiro [4.5] decane-2-carboxylate (620 mg, 845.84 μmol) in HCl/dioxane (5 mL) was stirred at 20 ℃ for 16h. The reaction mixture was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 516.4 [M+H] +.
Step D: 1- [ (4-methoxyphenyl) methyl] -3- [7- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-
diazaspiro [4.5] decan-8-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 3- [7- (2, 8-diazaspiro [4.5] decan-8-yl) -1-oxo-phthalazin-2-yl] -1- [ (4-methoxyphenyl) methyl] piperidine-2, 6-dione (650 mg, 718.22 μmol, HCl) , 1- (4-nitrophenyl) piperidin-4-one (158.17 mg, 718.22 μmol) in DCM (10 mL) was added TEA (145.35 mg, 1.44 mmol) and NaBH (OAc) 3 (456.66 mg, 2.15 mmol) . The mixture was stirred at 25 ℃ for 2 h. The reaction mixture was concentrated under reduced pressure to give crude product. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol = 100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 720.5 [M+H] +.
Step E: 3- [7- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-
phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 1- [ (4-methoxyphenyl) methyl] -3- [7- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (511 mg, 709.89 μmol)
in DCE (25 mL) was added AlCl3 (946.58 mg, 7.10 mmol) . The mixture was stirred at 85 ℃ for 1 h. The mixture was added H2O (60 mL) . The aqueous layer was extracted with DCM: MeOH (10: 1) (3 × 100 mL) and the combined organic layers were dried with anhydrous Na2SO4, filtered then concentrated. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol = 100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 600.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.98 (s, 1H) , 8.23 (s, 1H) , 8.05 (d, J = 8.8 Hz, 2H) , 7.76 (d, J = 8.8 Hz, 1H) , 7.61 (d, J = 8.4 Hz, 1H) , 7.50 (s, 1H) , 7.05 (d, J = 8.4 Hz, 2H) , 5.78 -5.74 (m, 1H) , 4.08 (d, J = 4.8 Hz, 2H) , 3.47 (m, 4H) , 3.17 (m, 2H) , 3.16 (m, 2H) , 3.03 (m, 2H) , 2.99 -2.85 (m, 2H) , 2.65 -2.52 (m, 3H) , 2.16 -2.03 (m, 2H) , 1.85-1.45 (m, 8H) .
Step F: 3- [7- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-
phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 3- [7- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (326 mg, 472.95 μmol) in DCM (5 mL) and DMAC (1 mL) was added PdCl2 (83.87 mg, 472.95 μmol) , Et3SiH (1.10 g, 9.46 mmol) and Et3N (95.72 mg, 945.91 μmol) . The mixture was stirred at 20 ℃ for 16 h. The reaction mixture was added aq. sat. Na2CO3 (30 mL) and extracted with ethyl acetate (30 mL × 3) . The combined organic phases were dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol = 100/1 to 5/1) to afford the desired product.
LCMS: MS (ESI) m/z 570.4 [M+H] +.
Step G: 3- (7- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2-phthalazinyl) -2, 6-
piperidinedione
To a solution of 3- [7- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (100 mg, 175.53 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (67.66 mg, 175.53 μmol) in n-BuOH (2 mL) was added AcOH (105.41 mg, 1.76 mmol) . The mixture was stirred at 100 ℃ for 2 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: Welch Xtimate C18 150*25mm*5um; mobile phase: [water (TFA) -ACN] ; gradient: 25%-55%B over 7 min) to afford the desired product.
LCMS: MS (ESI) m/z 891.7 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.01 (s, 1H) , 10.00 (m, 1H) , 8.83 (s, 1H) , 8.25 (s, 1H) , 8.01 (t, J = 8.0 Hz, 1H) , 7.78 (d, J = 8.8 Hz, 1H) , 7.75 (d, J = 8.4 Hz, 1H) , 7.67 -7.62 (m, 1H) , 7.62 -7.60 (m, 1H) , 7.59 (d, J = 7.6 Hz, 2H) , 7.52 (d, J = 2.0 Hz, 1H) , 6.99 (d, J = 8.8 Hz, 2H) , 5.75 (dd, J = 4.8, 11.2 Hz, 1H) , 5.25-5.45 (m, 2H) , 4.46 (d, J = 5.6 Hz, 2H) , 3.80 (d, J = 11.6 Hz, 2H) , 3.67 (s, 1H) , 3.60 -3.54 (m, 1H) , 3.50 (m, 4H) , 3.38-3.27 (m, 2H) , 3.11 -3.03 (m, 1H) , 2.98 -2.86 (m, 1H) , 2.75-2.65 (m, 2H) , 2.64 -2.53 (m, 2H) , 2.26 -1.92 (m, 7H) , 1.90 -1.76 (m, 2H) , 1.66-1.74 (m, 6H) , 1.56 (m, 3H) .
Example 143:
3- (6- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2-phthalazinyl) -2, 6-piperidinedione (Cpd-346)
Scheme:
Step A: tert-butyl 8- (1-oxo-2H-phthalazin-6-yl) -2, 8-diazaspiro [4.5] decane-2-carboxylate
To a solution of 6-bromo-2H-phthalazin-1-one (1 g, 4.44 mmol) , tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (1.07 g, 4.44 mmol) in dioxane (20 mL) was added 1, 3-bis [2, 6-bis (1-ethylpropyl) phenyl] -2Himidazole; 3-chloropyridine; dichloropalladium (176.34 mg,
222.18 μmol) and t-BuONa (1.28 g, 13.33 mmol) under N2. The mixture was stirred at 100 ℃for 16 h. The reaction mixture was added H2O (100 mL) at 0 ℃, and extracted with ethyl acetate (50 mL × 3) . The combined organic phases were dried with anhydrous Na2SO4, filtered and concentrated in under reduce pressure. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate = 100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 385.3 [M+H] +.
Step B: tert-butyl 8- [2- [1- [ (4-methoxyphenyl) methyl] -2, 6-dioxo-3-piperidyl] -1-oxo-
phthalazin-6-yl] -2, 8-diazaspiro [4.5] decane-2-carboxylate
To a solution of tert-butyl 8- (1-oxo-2H-phthalazin-6-yl) -2, 8-diazaspiro [4.5] decane-2-carboxylate (600.00 mg, 1.56 mmol) in THF (15 mL) was added t-BuOK (210.14 mg, 1.87 mmol) at 0 ℃, the reaction solution was stirred for 30 min at this temperature, then to this was added [1- [ (4-methoxyphenyl) methyl] -2, 6-dioxo-3-piperidyl] trifluoromethanesulfonate (654.60 mg, 1.72 mmol) in THF (7.5 mL) dropwise. The resulting solution was stirred at 0 ℃ for 2 h. The mixture quenched with saturated aq. sat. NH4Cl. The aqueous layer was extracted with EtOAc (3 × 30 mL) and the combined organic layers were washed with brine (3 × 30 mL) , dried with anhydrous Na2SO4, filtered then concentrated. The crude product was triturated with EtOAc (10 mL) to afford the desired product.
LCMS: MS (ESI) m/z 616.4 [M+H] +.
2D NMR: showed C13 appears at 58.23ppm, which is consistent with the determined structure.
1H NMR (400 MHz, DMSO-d6) δ 8.23 (s, 1H) , 8.02 (d, J = 9.2 Hz, 1H) , 7.49 (d, J = 9.2 Hz, 1H) , 7.25 (s, 1H) , 7.19 (d, J = 8.4 Hz, 2H) , 6.86 (d, J = 8.4 Hz, 2H) , 5.90 (dd, J = 4.8, 12.4 Hz, 1H) , 4.86 -4.69 (m, 2H) , 3.72 (s, 3H) , 3.60 -3.38 (m, 4H) , 3.31 (s, 2H) , 3.15 (s, 2H) , 3.12 -3.01 (m, 1H) , 2.83-2.76 (m, 1H) , 2.64 -2.53 (m, 1H) , 2.17 -2.06 (m, 1H) , 1.80-1.70 (m, 2H) , 1.59 (m, 4H) , 1.40 (s, 9H) .
Step C: 3- [6- (2, 8-diazaspiro [4.5] decan-8-yl) -1-oxo-phthalazin-2-yl] -1- [ (4-
methoxyphenyl) methyl] piperidine-2, 6-dione
To a solution of tert-butyl 8- [2- [1- [ (4-methoxyphenyl) methyl] -2, 6-dioxo-3-piperidyl] -1-oxo-phthalazin-6-yl] -2, 8-diazaspiro [4.5] decane-2-carboxylate (700 mg, 1.14 mmol) in DCM (5 mL) was added TFA (1.54 g, 13.46 mmol, 1 mL) , and then stirred at 20 ℃ for 2 h. The reaction mixture was concentrated under reduced pressure to give residue. To the residue was added toluene (4 mL) and MeOH (2 mL) . The mixture was concentrated in vacuo to give a residue. Repeat the preceding operation three times to afford the desired product.
LCMS: MS (ESI) m/z 516.5 [M+H] +.
Step D: 1- [ (4-methoxyphenyl) methyl] -3- [6- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-
diazaspiro [4.5] decan-8-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 3- [6- (2, 8-diazaspiro [4.5] decan-8-yl) -1-oxo-phthalazin-2-yl] -1- [ (4-methoxyphenyl) methyl] piperidine-2, 6-dione (890 mg, 1.41 mmol, TFA) , 1- (4-nitrophenyl) piperidin-4-one (373.55 mg, 1.70 mmol) in DCM (10 mL) was added TEA (286.07 mg, 2.83 mmol) and NaBH (OAc) 3 (898.76 mg, 4.24 mmol) . The mixture was stirred at 20 ℃for 16 h. The reaction mixture was concentrated under reduced pressure to give crude product. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol = 100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 720.5 [M+H] +.
Step E: 3- [6- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-
phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 1- [ (4-methoxyphenyl) methyl] -3- [6- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (690 mg, 958.56 μmol) in DCE (34 mL) was added AlCl3 (1.28 g, 9.59 mmol) . The mixture was stirred at 85 ℃ for 1 h. The mixture was added H2O (60 mL) . The aqueous layer was extracted with DCM: MeOH (10: 1) (3 × 100 mL) and the combined organic layers were dried with anhydrous Na2SO4, filtered then concentrated. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol = 100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 600.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.98 (s, 1H) , 8.23 (s, 1H) , 8.11 -8.00 (m, 3H) , 7.50 (d, J = 8.8 Hz, 1H) , 7.27 (s, 1H) , 7.07 (m, 2H) , 5.73 (d, J = 5.2 Hz, 1H) , 4.08 (q, J = 5.2 Hz, 2H) , 3.49 (m, 4H) , 3.18 (m, 2H) , 3.17 (m, 2H) , 3.03 (m, 2H) , 2.98 -2.86 (m, 2H) , 2.65-2.53 (m, 3H) , 2.13 -2.03 (m, 2H) , 1.87 -1.51 (m, 8H) .
Step F: 3- [6- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-
phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 3- [6- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (463 mg, 671.71 μmol) in DCM (5 mL) and DMAC (1 mL) was added PdCl2 (119.11 mg, 671.71 μmol) , Et3SiH (1.56 g, 13.43 mmol) , Et3N (135.94 mg, 1.34 mmol) . The mixture was stirred at 20 ℃ for 16 h. The reaction mixture was added aq. sat. Na2CO3 (30 mL) and extracted with ethyl acetate (30 mL × 3) . The combined organic phases were dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol = 100/1 to 5/1) to afford the desired product.
LCMS: MS (ESI) m/z 569.4 [M+H] +.
Step G: 3- (6- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2-phthalazinyl) -2, 6-
piperidinedione
To a solution of 3- [6- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (55 mg, 96.54 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl 2, 4, 6, 10, 21pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (37.21 mg, 96.54 μmol) in n-BuOH (2 mL) was added AcOH (57.98 mg, 965.43 μmol) . The mixture was stirred at 100 ℃ for 2 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: Welch Xtimate C18 150*25mm*5um; mobile phase: [water (TFA) -ACN] ; gradient: 25%-55%B over 7 min) to afford the desired product.
LCMS: MS (ESI) m/z 891.8 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ11.00 (s, 1H) , 10.05 (TFA, 0.82H) , 9.77 (s, 1H) , 8.83 (s, 1H) , 8.23 (s, 1H) , 8.02 -7.98 (m, 1H) , 8.02 -7.97 (m, 1H) , 7.75 (d, J = 8.4 Hz, 1H) , 7.59 (d, J = 8.0 Hz, 3H) , 7.52 (d, J = 9.2 Hz, 1H) , 7.28 (s, 1H) , 6.97 (d, J = 9.2 Hz, 2H) , 5.74 (dd, J = 5.2, 12.4 Hz, 1H) , 5.41 -5.20 (m, 2H) , 4.46 (d, J = 5.6 Hz, 2H) , 3.81 (d, J = 12.8 Hz, 2H) , 3.75-3.52 (m, 6H) , 3.34 (m, 2H) , 3.10 -3.04 (m, 1H) , 2.95 -2.87 (m, 1H) , 2.74 -2.64 (m, 2H) , 2.63 -2.53 (m, 2H) , 2.15 -1.93 (m, 7H) , 1.88 -1.76 (m, 2H) , 1.72-1.62 (m, 6H) , 1.56 (s, 3H) .
Example 144:
3- (6- {2- [1- (p- { (R) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-347)
Scheme:
Step A: 2- (6-bromo-2-pyridyl) propan-2-ol
To a solution of 2, 6-dibromopyridine (100.0 g, 422.1 mmol) in DCM (500.0 mL) was added n-BuLi (2.5 M, 141.8 mL) dropwise slowly at -70 ℃ under nitrogen atmosphere. The resulting mixture was stirred at -70 ℃ for 1 hr. Then acetone (46.6 mL, 633.8 mmol) was added dropwise
to the reaction mixture at same temperature. After the addition was finished, the reaction mixture was stirred at -70 ℃ for 0.5 hr. The reaction mixture was quenched by addition saturated NH4Cl (50.0 mL) , and then diluted with water (100.0 mL) . The organic layer was separated and washed with water (100 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 215.7, 217.7 [M+H] +.
1H NMR (400MHz, CDCl3) δ 7.58 -7.54 (m, 1H) , 7.37 (t, J=8.0 Hz, 2H) , 4.11 (s, 1H) , 1.59 -1.51 (m, 6H) .
Step B: 2-bromo-6-isopropenyl-pyridine
2- (6-bromo-2-pyridyl) propan-2-ol (106.6 g, crude) was added dropwise to H2SO4 (91.0 mL) . After the addition was finished, the mixture was stirred at 60 ℃ for 16 hr. The reaction mixture was poured into ice water (270 mL) and adjusted to pH=9~10 with 6 N NaOH (~600 mL) . Then extracted with petroleum ether (100 mL×3) . The combined organic layer was washed with brine (50.0 mL) , dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by column chromatography (SiO2, Petroleum ether) to afford the desired product.
LCMS: MS (ESI) m/z 197.8, 199.8 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 7.53 -7.48 (m, 1H) , 7.40 (dd, J=0.8, 7.6 Hz, 1H) , 7.34 (dd, J=0.8, 7.6 Hz, 1H) , 5.94 (d, J=0.8 Hz, 1H) , 5.35 -5.32 (m, 1H) , 2.18 (dd, J=0.8, 1.6 Hz, 3H) .
Step C: (2S) -2- (6-bromo-2-pyridyl) propane-1, 2-diol
To a suspension of potassium carbonate (104.7 g, 757.3 mmol) , tripotassium; hexacyanoiron (3-) (249.4 g, 757.345 mmol) and methanesulfonamide (26.4 g, 277.7 mmol) in t-BuOH (500.0 mL) and water (500.0 mL) were added 4- [ (R) - [ (2R, 4R, 5S) -5-ethylquinuclidin-2-yl] - (6-methoxy-4-quinolyl) methoxy] -1- [ (R) - [ (2R, 4S, 5R) -5-ethylquinuclidin-2-yl] - (6-methoxy-4-quinolyl) methoxy] phthalazine (1.97 g, 2.52 mmol, ) and dipotassium; dioxido (dioxo) osmium; dihydrate (930 mg, 2.52 mmol) . After the addition was finished, the reaction mixture was stirred at 20 ℃ for 30 min. Then the reaction mixture was cooled to 0 ℃, 2-bromo-6-isopropenyl-pyridine (50.0 g, 252.4 mmol) was added dropwise. The mixture was stirred at 20 ℃ for 15 hr. under nitrogen atmosphere. The reaction mixture was quenched by addition saturated Na2SO3 (100.0 mL) , extracted with ethyl acetate (75 mL× 2) . The combined organic layers were washed with brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was triturated with petroleum ether (100.0 mL) at 20 ℃ for 1 hr., filtered and rinsed with petroleum ether (25.0 mL× 3) to afford the desired product.
LCMS: MS (ESI) m/z 232.2, 234.2 [M+H] +.
1H NMR (400 MHz, CDCl3) : δ 7.61 -7.55 (m, 1H) , 7.46 -7.43 (m, 1H) , 7.39 (dd, J=0.8, 8.0 Hz, 1H) , 4.37 (s, 1H) , 3.86 (d, J=11.2 Hz, 1H) , 3.68 (d, J=11.2 Hz, 1H) , 1.47 (s, 3H) .
Step D: 2-bromo-6- [ (2S) -2-methyloxiran-2-yl] pyridine
To a solution of (2S) -2- (6-bromo-2-pyridyl) propane-1, 2-diol (32.8 g, 141.3 mmol) and DBU (63.9 mL, 424.0 mmol) in toluene (320.0 mL) was added 1, 1, 2, 2, 3, 3, 4, 4, 4-nonafluorobutane-1-sulfonyl fluoride (38.6 mL, 219.0 mmol) dropwise at 0 ℃. After the addition was finished, the mixture was stirred at 20 ℃ for 15 hr. under nitrogen atmosphere. The reaction mixture was poured into water (30.0 mL) , the organic layer was separated and the aqueous layer was extracted with toluene (15 mL×2) . The combined organic layers were washed with brine (20 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=50/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 214.1, 216.1 [M+H] +
1H NMR (400 MHz, CDCl3) : δ 7.56 -7.49 (m, 1H) , 7.39 (d, J=7.6 Hz, 1H) , 7.31 (d, J=7.6 Hz, 1H) , 3.01 (d, J=5.4 Hz, 1H) , 2.87 (d, J=5.4 Hz, 1H) , 1.78 (s, 3H) .
Step E: (2R) -2- (6-bromo-2-pyridyl) hex-5-en-2-ol
To a solution of allyl (bromo) magnesium (in ether) (1 M, 43.7 mL) was added dropwise a solution of 2-bromo-6- [ (2S) -2-methyloxiran-2-yl] pyridine (10.4 g, 48.58 mmol) in DCM (50.0 mL) at -60 ℃ under nitrogen atmosphere. After addition, the mixture was stirred at this temperature for 1 hr. The reaction mixture was poured into saturated NH4Cl (100.0 mL) , adjusted pH=10 by with NH3·H20 (25%) and extracted with ethyl acetate (80.0 mL×3) . The combined organic layers were washed with brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=20/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 238.1, 241.0 [M+H-H2O] +
1H NMR (400 MHz, CDCl3) δ 7.60 -7.54 (m, 1H) , 7.39 (d, J=7.6 Hz, 1H) , 7.32 (d, J=7.6 Hz, 1H) , 5.76 (tdd, J=6.4, 10.2, 16.8 Hz, 1H) , 4.95 (d, J=17.2 Hz, 1H) , 4.90 (d, J=10.4 Hz, 1H) , 2.20 -2.08 (m, 1H) , 1.96 -1.87 (m, 2H) , 1.86 -1.75 (m, 1H) , 1.54 (s, 3H) .
Step F: [ (1R) -1- (6-bromo-2-pyridyl) -1-methyl-pent-4-enoxy] -triethyl-silane
To a solution of (2R) -2- (6-bromo-2-pyridyl) hex-5-en-2-ol (5.0 g, 19.52 mmol) and 2, 6-dimethylpyridine (2.73 mL, 23.42 mmol) in DCM (50.0 mL) was added triethylsilyl trifluoromethanesulfonate (5.30 mL, 23.42 mmol) at 0 ℃. The mixture was stirred at 0 ℃ for 2hr under nitrogen atmosphere. The mixture was diluted with H2O (50 mL) , the organic layer was separated, the aqueous layer was extracted with DCM (20 mL×2) . The combined organic
were washed with brine (30 mL) , dried over Na2SO4, filtrated and the filtrate was concentrated to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=100/1 to 50/1) to afford the desired product.
1H NMR (400 MHz, CDCl3) δ 7.60 (dd, J=0.8, 7.6 Hz, 1H) , 7.49 (t, J=7.6 Hz, 1H) , 7.29 (dd, J=0.8, 7.6 Hz, 1H) , 5.72 (m, 1H) , 4.96 -4.82 (m, 2H) , 2.18 -2.01 (m, 2H) , 1.80 -1.70 (m, 1H) , 1.64 (s, 3H) , 1.63 -1.57 (m, 1H) , 1.02 -0.96 (m, 9H) , 0.67 (m, 6H) .
Step G: (4R) -4- (6-bromo-2-pyridyl) -4-triethylsilyloxy-pentanal
To a solution of [ (1R) -1- (6-bromo-2-pyridyl) -1-methyl-pent-4-enoxy] -triethyl-silane (2.0 g, 5.40 mmol) in dioxane (60 mL) and water (20 mL) were added 2, 6-dimethylpyridine (1.45 mL, 12.42 mmol) , dipotassium; dioxido (dioxo) osmium; dihydrate (40 mg, 107.9 μmol) and sodium periodate (4.62 g, 21.60 mmol) . The mixture was stirred at 18 ℃ for 2hr. The reaction mixture was filtrated and the filtrate was diluted with water (100.0 mL) , extracted with EtOAc (100 mL×2) . The combined organic layer washed with brine (50 mL) , dried over Na2SO4, filtrated and the filtrate was concentrated to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=50/1 to 20/1) to afford the desired product.
1H NMR (400 MHz, CDCl3) δ 9.68 -9.62 (m, 1H) , 7.59 -7.55 (m, 1H) , 7.55 -7.50 (m, 1H) , 7.32 (dd, J=1.2, 7.6 Hz, 1H) , 2.43 -2.28 (m, 2H) , 2.18 -1.99 (m, 2H) , 1.68 (s, 3H) , 1.01 -0.96 (m, 9H) , 0.70 -0.63 (m, 6H) .
Step H: ethyl (Z, 6R) -6- (6-bromo-2-pyridyl) -2-fluoro-6-triethylsilyloxy-hept-2-enoate
To a solution of ethyl 2-diethoxyphosphoryl-2-fluoro-acetate (3.82 g, 15.79 mmol) in DMSO (40.0 mL) was added DBU (4.01 g, 26.32 mmol) . After stirring at 20 ℃ for 15 min. (4R) -4- (6-bromo-2-pyridyl) -4-triethylsilyloxy-pentanal (4.9 g, 13.16 mmol) was added to the reaction mixture, the mixture was stirred at 20 ℃ for 1hr. The reaction mixture was diluted with water (400.0 mL) and extracted with Petroleum ether/Ethyl acetate (5/1, 100 mL×3) . The combined organic layers were washed with bine (50.0 mL) , dried over Na2SO4, filtered and concentrated in vacuum. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=100/1 to 20/1) to afford the desired product.
LCMS: MS (ESI) m/z 460.2, 462.2 [M+H] +
1H NMR (400 MHz, CDCl3) δ 7.61 (d, J=7.2 Hz, 1H) , 7.51 (t, J=7.6 Hz, 1H) , 7.31 -7.28 (m, 1H) , 6.07 -5.83 (m, 1H) , 4.24 (q, J=7.2 Hz, 2H) , 2.34 -2.15 (m, 2H) , 1.92 -1.74 (m, 2H) , 1.64 (s, 3H) , 1.32 (t, J=7.1 Hz, 3H) , 1.03 -0.97 (m, 9H) , 0.71 -0.64 (m, 6H) .
Step I: (Z, 6R) -6- (6-bromo-2-pyridyl) -2-fluoro-6-triethylsilyloxy-hept-2-en-1-ol
To a solution of ethyl (Z, 6R) -6- (6-bromo-2-pyridyl) -2-fluoro-6-triethylsilyloxy-hept-2-enoate (3.4 g, 7.38 mmol) in DCM (34.0 mL) was added DIBAL-H (in toluene, 1 M, 18.46 mL) under
nitrogen atmosphere. The mixture was stirred at 0 ℃ for 2hr. The reaction mixture was quenched by addition NH4Cl (20.0 mL) at 0 ℃, filtered through a pad of Celite and rinsed with DCM (10.0 mL×3) . The combined organic layers were washed with brine (20 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=50/1 to 10/1) to afford the desired product.
1H NMR (400 MHz, CDCl3) δ 7.80 -7.74 (m, 1H) , 7.59 (d, J=7.6 Hz, 1H) , 7.50 (d, J=8.0 Hz, 1H) , 5.08 (t, J=5.6 Hz, 1H) , 4.82 -4.61 (m, 1H) , 3.81 (dd, J=5.6, 14.8 Hz, 2H) , 2.10 -1.88 (m, 2H) , 1.69 (dt, J=3.6, 12.0 Hz, 1H) , 1.55 (s, 4H) , 0.93 (t, J=8.0 Hz, 9H) , 0.61 (q, J=8.0 Hz, 6H) .
Step J: 2-allyl-1- [6- [ (Z, 1R) -5-fluoro-6-hydroxy-1-methyl-1-triethylsilyloxy-hex-4-enyl] -2-
pyridyl] -6-methylsulfanyl-pyrazolo [3, 4-d] pyrimidin-3-one
To a solution of (Z, 6R) -6- (6-bromo-2-pyridyl) -2-fluoro-6-triethylsilyloxy-hept-2-en-1-ol (1.4 g, 3.35 mmol) , K2CO3 (1.16 g, 8.36 mmol) and 2-allyl-6-methylsulfanyl-1H-pyrazolo [3, 4-d] pyrimidin-3-one (819 mg, 3.68 mmol) in dioxane (15.0 mL) were added (1R, 2R) -N1, N2-dimethylcyclohexane-1, 2-diamine (142.78 mg, 1.00 mmol) and CuI (96 mg, 501.8 μmol) . The mixture was stirred at 105 ℃ for 15hr . The mixture was diluted with ethyl acetate (10.0 mL) , filtered through diatomite pad under vacuum and washed with ethyl acetate (5 mL × 3) . The filtrate was evaporated in vacuum. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1/1) to afford the desired product.
LCMS: MS (ESI) m/z 560.2 [M+H] +
1H NMR (400 MHz, CDCl3) δ 8.94 (s, 1H) , 7.90 -7.83 (m, 1H) , 7.71 (d, J=7.6 Hz, 1H) , 7.64 (d, J=7.6 Hz, 1H) , 5.65 (tdd, J=6.4, 10.4, 16.8 Hz, 1H) , 5.03 (d, J=10.0 Hz, 1H) , 4.92 -4.76 (m, 3H) , 4.73 -4.57 (m, 1H) , 3.99 (dd, J=6.4, 16.0 Hz, 2H) , 2.59 (s, 3H) , 2.21 -2.06 (m, 2H) , 1.82 -1.67 (m, 2H) , 1.63 (s, 3H) , 1.03 -0.98 (m, 9H) , 0.72 -0.65 (m, 6H) .
Step K: 1- [6- [ (Z, 1R) -5-fluoro-6-hydroxy-1-methyl-1-triethylsilyloxy-hex-4-enyl] -2-
pyridyl] -6-methylsulfanyl-2H-pyrazolo [3, 4-d] pyrimidin-3-one
To a solution of 2-allyl-1- [6- [ (Z, 1R) -5-fluoro-6-hydroxy-1-methyl-1-triethylsilyloxy-hex-4-enyl] -2-pyridyl] -6-methylsulfanyl-pyrazolo [3, 4-d] pyrimidin-3-one (1.47 g, 2.63 mmol) in Dioxane (6.0 mL) was added ammonia; formic acid (199 mg, 3.15 mmol) and Pd (dppf) Cl2. CH2Cl2 (107 mg, 131.3 μmol) . The mixture was stirred at 100 ℃ for 2hr under nitrogen atmosphere. The reaction mixture was diluted with water (30.0 mL) and extracted with ethyl acetate (15 mL×3) . The combined organic layers were washed with brine (15 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, DCM/MeOH=50/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 520.2 [M+H] +
1H NMR (400 MHz, CDCl3) δ 8.97 -8.92 (m, 1H) , 8.34 (d, J=8.4 Hz, 1H) , 7.88 -7.82 (m, 1H) , 7.56 (d, J=7.6 Hz, 1H) , 4.78 -4.59 (m, 1H) , 4.05 -3.94 (m, 2H) , 2.70 (s, 3H) , 2.40 -2.18 (m, 2H) , 1.84 (dt, J=4.0, 12.0 Hz, 1H) , 1.77 -1.71 (m, 3H) , 1.66 (d, J=8.4 Hz, 1H) , 1.05 -1.00 (m, 9H) , 0.74 -0.68 (m, 6H) .
Step L: (12Z, 16R) -12-fluoro-16-methyl-5-methylsulfanyl-16-triethylsilyloxy-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one
To a solution of 1- [6- [ (Z, 1R) -5-fluoro-6-hydroxy-1-methyl-1-triethylsilyloxy-hex-4-enyl] -2-pyridyl] -6-methylsulfanyl-2H-pyrazolo [3, 4-d] pyrimidin-3-one (1.3 g, 2.50 mmol) in DCM (30.0 mL) were added DEAD (585 mg, 3.36 mmol) and PPh3 (1.0 g, 3.81 mmol) . The mixture was stirred at 20 ℃ for 15hr under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was purified by flash silica gel chromatography (20 g Silica Flash Column, Eluent of 30~50%Ethyl acetate/Petroleum ether gradient @25 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 502.1 [M+H] +
1H NMR (400 MHz, CDCl3) δ 7.91 -7.84 (m, 1H) , 7.75 (d, J=7.6 Hz, 1H) , 7.52 (d, J=7.6 Hz, 1H) , 5.19 (d, J=15.6 Hz, 1H) , 3.86 -3.68 (m, 1H) , 2.60 -2.52 (m, 3H) , 2.23 (d, J=10.4 Hz, 1H) , 1.90 -1.69 (m, 3H) , 1.69 -1.66 (m, 3H) , 0.98 (t, J=8.0 Hz, 9H) , 0.65 (q, J=8.0 Hz, 6H) .
Step M: (12Z, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfanyl-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one
To a solution of (12Z, 16R) -12-fluoro-16-methyl-5-methylsulfanyl-16-triethylsilyloxy-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (510 mg, 1.02 mmol) in THF (10.0 mL) was added TBAF (1 M, 1.52 mL) . The mixture was stirred at 20 ℃ for 2hr under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was purified by flash silica gel chromatography (12 g Silica Flash Column, Eluent of 50~80%Ethyl acetate/Petroleum ether gradient @25 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 388.1 [M+H] +
1H NMR (400 MHz, CDCl3) δ 8.98 -8.95 (m, 1H) , 7.97 -7.90 (m, 1H) , 7.62 (d, J=8.0 Hz, 1H) , 7.32 (d, J=7.6 Hz, 1H) , 5.25 (d, J=15.6 Hz, 1H) , 4.59 (s, 1H) , 3.99 -3.78 (m, 1H) , 3.17 -3.00 (m, 1H) , 2.55 (s, 3H) , 2.07 (td, J=4.0, 14.4 Hz, 1H) , 1.76 (td, J=3.6, 14.0 Hz, 1H) , 1.68 (s, 3H) , 1.61 -1.49 (m, 2H) .
Step N: (12Z, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one
To a solution of (12Z, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfanyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (140 mg,
361.3 μmol) in THF (2.0 mL) and Water (2.0 mL) was added Oxone (222 mg, 361.3 μmol) at 0℃. After addition, the mixture was stirred at 0~5 ℃ for 1.5 hr. The reaction mixture was diluted with water (5.0 mL) and extracted with ethyl acetate (5 mL×3) . The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 404.1 [M+H] +
Step O: 3- (6- {2- [1- (p- { (R) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of (12Z, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (45 mg, 111.5 μmol) and 3- [6- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (60 mg, 107.7 μmol) in n-BuOH (1.5 mL) was added HOAc (67 mg, 1.12 mmol) . The mixture was stirred at 100 ℃ for 3hr under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was purified by prep-HPLC (column: Welch Xtimate C18 150*25mm*5um; mobile phase: [water (TFA) -ACN] ; gradient: 20%-50%B over 7 min) to afford the desired product.
LCMS: MS (ESI) m/z 896.7 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ 10.97 (s, 1H) , 9.69 (s, 1H) , 8.83 (s, 1H) , 8.06 (t, J=7.6 Hz, 1H) , 7.71 (d, J=8.0 Hz, 1H) , 7.60 (m, 3H) , 7.44 (d, J=8.4 Hz, 1H) , 7.29 (d, J=8.0 Hz, 1H) , 7.22 (s, 1H) , 6.94 (d, J=8.8 Hz, 2H) , 5.09 (dd, J=4.8, 13.2 Hz, 1H) , 4.85 (d, J=16.0 Hz, 1H) , 4.37 -4.18 (m, 2H) , 3.79 (d, J=13.6 Hz, 2H) , 3.38 -3.23 (m, 5H) , 3.03 -3.09 (m, 1H) , 2.96 -2.84 (m, 1H) , 2.74 -2.61 (m, 4H) , 2.44 -2.30 (m, 3H) , 2.17 -2.06 (m, 3H) , 2.05 -1.94 (m, 3H) , 1.85 -1.76 (m, 3H) , 1.73 -1.65 (m, 5H) , 1.63 -1.53 (m, 2H) , 1.49 (s, 3H) .
Example 145:
3- (5- {2- [1- (p- { (R) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-348)
Scheme:
Step A: 3- (5- {2- [1- (p- { (R) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of (12Z, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (45 mg, 111.5 μmol) and 3- [5- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (60 mg, 107.7 μmol) in n-BuOH (2.0 mL) was added HOAc (67 mg, 1.12 mmol) . The mixture was stirred at 100 ℃ for 2 hr under nitrogen atmosphere. The reaction was concentrated in vacuum. The residue was purified by prep-HPLC (column: Welch Xtimate C18 150*25mm*5um; mobile phase: [water (TFA) -ACN] ; gradient: 20%-50%B over 7 min) to afford the desired product.
LCMS: MS (ESI) m/z 896.8 [M+H] +
1H NMR (400MHz, DMSO-d6) δ 10.94 (s, 1H) , 10.12 (s, 1H) , 9.73 (s, 1H) , 8.88 -8.80 (m, 1H) , 8.06 (t, J = 7.6 Hz, 1H) , 7.71 (d, J = 7.6 Hz, 1H) , 7.60 (d, J = 7.6 Hz, 3H) , 7.52 (d, J = 8.4 Hz, 1H) , 7.12 -7.05 (m, 2H) , 6.94 (d, J = 8.8 Hz, 2H) , 5.04 (dd, J = 4.8, 13.2 Hz, 1H) , 4.85 (d, J = 15.6 Hz, 1H) , 4.36 -4.16 (m, 2H) , 3.92 -3.74 (m, 2H) , 3.44 -3.23 (m, 4H) , 3.10 -3.01 (m, 1H) , 2.96 -2.85 (m, 1H) , 2.74 -2.52 (m, 6H) , 2.45 -2.27 (m, 2H) , 2.21 -2.06 (m, 3H) , 2.05 -1.91 (m, 3H) , 1.88 -1.79 (m, 2H) , 1.78 -1.63 (m, 7H) , 1.63 -1.53 (m, 2H) , 1.49 (m, 3H) .
Example 146:
3- {5- [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] -1, 3-dioxo-2-isoindolinyl} -2, 6-piperidinedione (Cpd-349)
Scheme:
Step A: 2- (2, 6-dioxo-3-piperidyl) -5- [4- (hydroxymethyl) -1-piperidyl] isoindoline-1, 3-dione
A mixture of 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (2 g, 7.24 mmol) , 4-piperidylmethanol (1.67 g, 14.48 mmol) , DIEA (2.81 g, 21.72 mmol, 3.78 mL) in DMF (20 mL) was stirred at 100 ℃ for 4 hr. The mixture was concentrated to give a residue, which was purified by silica gel column chromatography (3%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 372.2 [M+H] +
Step B: 1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] piperidine-4-carbaldehyde
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [4- (hydroxymethyl) -1-piperidyl] isoindoline-1, 3-dione (1 g, 2.69 mmol) in DMSO (24 mL) was added IBX (1.06 g, 3.77 mmol) . The mixture was stirred at 20 ℃ for 1hr. The mixture was poured into water (100 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers were dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 370.2 [M+H] +
Step C: tert-butyl 4- [4- (4-nitrophenyl) piperazin-1-yl] piperidine-1-carboxylate
To a solution of 1- (4-nitrophenyl) piperazine (5 g, 24.13 mmol) , tert-butyl 4-oxopiperidine-1-carboxylate (4.81 g, 24.13 mmol) in EtOH (50 mL) was added dropwise AcOH (724.44 mg, 12.06 mmol, 690.60 μL) . After addition, the mixture was stirred at 50 ℃ for 0.5 hr, and then NaBH3CN (4.55 g, 72.38 mmol) was added. The resulting mixture was stirred at 50 ℃ for 2 hr. The mixture was poured into water (100 mL) and extracted with EtOAc (200 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by triturated with EtOAc (10 mL) at 20 ℃ for 1 hr. The mixture was filtered and the solid was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 391.2 [M+H] +
1H NMR (400 MHz, CDCl3) δ 8.13 (d, J = 9.2 Hz, 2H) , 6.83 (d, J = 9.6 Hz, 2H) , 4.25 -4.07 (m, 2H) , 3.47 -3.41 (m, 4H) , 2.81 -2.65 (m, 6H) , 2.48 (t, J = 11.2 Hz, 1H) , 1.84 (d, J = 12.4 Hz, 2H) , 1.47 (s, 9H) , 1.45 -1.39 (m, 2H) .
Step D: 1- (4-nitrophenyl) -4- (4-piperidyl) piperazine
To a solution of tert-butyl 4- [4- (4-nitrophenyl) piperazin-1-yl] piperidine-1-carboxylate (1 g, 2.56 mmol) in DCM (9 mL) was added TFA (4.61 g, 40.39 mmol, 3 mL) . The mixture was stirred at 20 ℃ for 1hr. The reaction mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 291.3 [M+H] +
Step E: 2- (2, 6-dioxo-3-piperidyl) -5- [4- [ [4- [4- (4-nitrophenyl) piperazin-1-yl] -1-
piperidyl] methyl] -1-piperidyl] isoindoline-1, 3-dione
To a solution of 1- (4-nitrophenyl) -4- (4-piperidyl) piperazine (1 g, 3.44 mmol) , 1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] piperidine-4-carbaldehyde (1.27 g, 3.44 mmol) in DCM (10 mL) was added dropwise TEA (1.05 g, 10.33 mmol, 1.44 mL) at 20 ℃. After addition, the mixture was stirred at this temperature for 0.5 hr, then NaBH (OAc) 3 (2.19 g, 10.33 mmol) was added. The mixture was stirred at 20℃ for 3hr. The mixture was poured into water (100 mL) and extracted with DCM (100 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue, which was purified by silica gel column chromatography (18%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 644.4 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ 8.14 (d, J = 9.2 Hz, 2H) , 7.68 (d, J = 8.4 Hz, 1H) , 7.29 (d, J = 2.0 Hz, 1H) , 7.06 (d, J = 7.2 Hz, 1H) , 6.84 (d, J = 9.2 Hz, 2H) , 4.94 (dd, J = 5.2, 12.4 Hz, 1H) , 3.44 (m, 8H) , 3.02 -2.45 (m, 9H) , 2.19 -2.09 (m, 1H) , 2.05 -1.85 (m, 2H) , 1.82 -1.38 (m, 12H) .
Step F: 5- [4- [ [4- [4- (4-aminophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -2- (2, 6-
dioxo-3-piperidyl) isoindoline-1, 3-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [4- [ [4- [4- (4-nitrophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] isoindoline-1, 3-dione (300.46 mg, 466.75 μmol) in DCM (5 mL) and DMAC (1 mL) was added PdCl2 (82.77 mg, 466.75 μmol) , Et3N (94.46 mg, 933.50 μmol) and Et3SiH (1.09 g, 9.33 mmol, 1.49 mL) . The mixture was stirred at 25 ℃ for 4 hr. The reaction mixture was poured into water (50 mL) and adjusted to pH = 8 with aq. Na2CO3 (50 mL) . Then extracted with petroleum ether (100 mL × 2) . The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated to give a residue, which was purified by silica gel column chromatography (18%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 614.4 [M+H] +
Step G: 3- {5- [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] -1, 3-dioxo-2-isoindolinyl} -
2, 6-piperidinedione
A mixture of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) , 5- [4- [ [4- [4- (4-aminophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (79.62 mg, 129.72 μmol) , AcOH (77.90 mg, 1.30 mmol) in n-BuOH (3 mL) was stirred at 100 ℃ for 2hr. The mixture was concentrated to give a residue, which was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 7%-37%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 935.8 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ 11.09 (s, 1H) , 10.51 -9.97 (TFA, 1.38H) , 9.43 (s, 1H) , 8.85 (s, 1H) , 8.00 (t, J = 8.0 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.72 -7.53 (m, 4H) , 7.37 (s, 1H) , 7.28 (d, J = 8.8 Hz, 1H) , 7.00 (d, J = 8.8 Hz, 2H) , 5.45 -5.20 (m, 2H) , 5.07 (dd, J = 5.2, 12.8 Hz, 1H) , 4.47 (m, 2H) , 4.11 (d, J = 12.4 Hz, 2H) , 3.60 -3.10 (m, 8H) , 3.10 -2.75 (m, 8H) , 2.72 -2.53 (m, 1H) , 2.45 -2.25 (m, 6H) , 2.16 (m, 2H) , 2.07 -1.90 (m, 5H) , 1.83 (d, J = 12.0 Hz, 2H) , 1.71 (s, 1H) , 1.56 (m, 3H) , 1.35 -1.15 (m, 2H) .
Example 147:
3- [5- (4- {3- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -1-pyrrolidinyl} -1-piperidyl) -1, 3-dioxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-350)
Scheme:
Step A: 2- (2, 6-dioxo-3-piperidyl) -5- (4-oxocyclohexyl) isoindoline-1, 3-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (2 g, 7.24 mmol) and piperidin-4-one (861.32 mg, 8.69 mmol) in DMF (20 mL) was added DIEA (2.81 g, 21.72 mmol) and the mixture was stirred at 100 ℃ for 16hr. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (40gSilica Flash Column, Eluent of 0~7%DCM/MeOH gradient @40mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 356.1 [M+H] +
1H NMR (400MHz, DMSO-d6) δ 11.14 (s, 1H) , 8.01 (dd, J = 4.5, 8.2 Hz, 1H) , 7.85 (dd, J = 2.4, 7.6 Hz, 1H) , 7.74 -7.70 (m, 1H) , 3.86 (t, J = 6.0 Hz, 1H) , 2.89 (s, 4H) , 2.73 (m, 5H) , 2.65 -2.53 (m, 2H) , 2.10 -2.03 (m, 1H)
Step B: tert-butyl 3- [4- (4-nitrophenyl) piperazin-1-yl] pyrrolidine-1-carboxylate
To a solution of 1- (4-nitrophenyl) piperazine (5 g, 24.13 mmol) and tert-butyl 3-oxopyrrolidine-1-carboxylate (4.47 g, 24.13 mmol) in EtOH (50 mL) was added ACOH (724.44 mg, 12.06 mmol, ) and the mixture was stirred at 50 ℃ for 0.5hr. Then the mixture was added NaBH3CN (4.55 g, 72.38 mmol) and stirred at 50 ℃ for 2 hr. The reaction mixture was quenched with water, and then extracted with DCM (300mL×3) . The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was triturated with EtOAc to afford the desired product.
LCMS: MS (ESI) m/z 321.0 [M+H] +
1H NMR (400 MHz, CDCl3) δ 8.12 (br d, J = 9.2 Hz, 2H) , 6.82 (br d, J = 9.2 Hz, 2H) , 3.75 -3.51 (m, 2H) , 3.48 -3.37 (m, 4H) , 3.34 -3.26 (m, 1H) , 3.21 -3.08 (m, 1H) , 2.84 (m, 1H) , 2.75 -2.51 (m, 4H) , 2.17 -2.06 (m, 1H) , 1.81 m, 1H) , 1.46 (s, 9H)
Step C: 1- (4-nitrophenyl) -4-pyrrolidin-3-yl-piperazine
To a solution of tert-butyl 3- [4- (4-nitrophenyl) piperazin-1-yl] pyrrolidine-1-carboxylate (1 g, 2.66 mmol) in DCM (9 mL) was added TFA (3 mL) and the mixture was stirred at 25 ℃ for 1hr. The reaction mixture was washed with H2O (20mL) and basified with Na2CO3 to pH=9, extracted with DCM (100×3) . The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 277.0 [M+H] +
Step D: 2- (2, 6-dioxo-3-piperidyl) -5- [4- [3- [4- (4-nitrophenyl) piperazin-1-yl] pyrrolidin-1-yl] -
1-piperidyl] isoindoline-1, 3-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- (4-oxocyclohexyl) isoindoline-1, 3-dione (1.12 g, 3.16 mmol) and 1- (4-nitrophenyl) -4-pyrrolidin-3-yl-piperazine (900 mg, 3.26 mmol) in EtOH (10 mL) was added AcOH (94.90 mg, 1.58 mmol) and the mixture was stirred at 50 ℃ for 14hr . Then the mixture was added NaBH3CN (595.85 mg, 9.48 mmol) and stirred at 50 ℃ for 2hr.
The reaction mixture was quenched with water, and then extracted with DCM (100mL×3) . The combined organic layers were washed with brine (100mL*3) , dried over Na2SO4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (40gSilica Flash Column, Eluent of 0~14 %DCM/MeOH gradient @30 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 616.4 [M+H] +
Step E: 5- [4- [3- [4- (4-aminophenyl) piperazin-1-yl] pyrrolidin-1-yl] -1-piperidyl] -2- (2, 6-
dioxo-3-piperidyl) isoindoline-1, 3-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [4- [3- [4- (4-nitrophenyl) piperazin-1-yl] pyrrolidin-1-yl] -1-piperidyl] isoindoline-1, 3-dione (200.00 mg, 324.85 μmol) in DCM (5 mL) and DMAC (1 mL) was added PdCl2 (57.60 mg, 324.85 μmol) , Et3N (65.74 mg, 649.69 μmol) and Et3SiH (755.43 mg, 6.50 mmol) . The mixture was stirred at 25 ℃ for 12hr . The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~9%DCM/MeOH gradient @15 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 586.2 [M+H] +
Step F: 3- [5- (4- {3- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -1-pyrrolidinyl} -1-piperidyl) -1, 3-dioxo-2-isoindolinyl] -2, 6-
piperidinedione
To a solution of 5- [4- [3- [4- (4-aminophenyl) piperazin-1-yl] pyrrolidin-1-yl] -1-piperidyl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (75.98 mg, 129.72 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) in n-BuOH (2 mL) was added AcOH (77.90 mg, 1.30 mmol, ) and the mixture was stirred at 100 ℃ for 12hr. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 8%-38%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 907.8 [M+H] +
1H NMR (400MHz, DMSO-d6) δ 11.08 (s, 1H) , 10.17 -9.97 (m, 1H) , 8.83 (s, 1H) , 7.99 (t, J = 8.0 Hz, 1H) , 7.77 -7.68 (m, 2H) , 7.60 (br t, J = 8.4 Hz, 3H) , 7.42 (s, 1H) , 7.36 -7.30 (m, 1H) , 6.98 (br d, J = 9.2 Hz, 2H) , 5.42 -5.23 (m, 2H) , 5.08 (dd, J = 5.2, 12.8 Hz, 1H) , 4.46 (br d, J = 6.4 Hz, 2H) , 4.22 (br d, J = 12.4Hz, 3H) , 3.34 -3.10 (m, 8H) , 3.04 -2.81 (m, 5H) , 2.64 -2.53 (m, 2H) , 2.42 -2.09 (m, 6H) , 2.08 -1.88 (m, 4H) , 1.76 -1.52 (m, 7H)
Example 148:
1- {5- [ (9- { [4- (4- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} -2-tolyl) -1-piperidyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-351)
Scheme:
Step A: tert-butyl 4- (2-methyl-4-nitrophenyl) -3, 6-dihydropyridine-1 (2H) -carboxylate
To a solution of 1-bromo-2-methyl-4-nitrobenzene (1.40 g, 6.47 mmol) , tert-butyl 4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydropyridine-1 (2H) -carboxylate (2.00 g, 6.47
mmol) , Na2CO3 (1.37 g, 12.9 mmol) , Pd (dppf) Cl2 (473 mg, 0.647 mmol) in Dioxane (30 mL) and H2O (3 mL) . Then the mixture was stirred at 100℃ for 16 h under N2 atmosphere. The mixture was filtered through a Celite pad, water (300 mL) was added and in filtrate the solution was extracted with EtOAc (300 mL x 3) . The combined organic layers were washed with brine (300 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. the crude product was purified by silica gel chromatography eluted with PE: EA=5: 1 to afford the desired product.
LCMS: MS (ESI) m/z 219.1 [M+H-tBu] +
Step B: tert-butyl 4- (4-amino-2-methylphenyl) piperidine-1-carboxylate
To a solution of tert-butyl 4- (2-methyl-4-nitrophenyl) -3, 6-dihydropyridine-1 (2H) -carboxylate (1.81 g, 5.70 mmol) in MeOH (20 mL) was added Pd/C (180 mg, w/w=10%) and stirred at 25℃under H2 atmosphere at 2 atm for 3 hrs. The mixture was filtered through a Celite pad, Then the reaction was filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 119.1 [M+H] +
Step C: tert-butyl 4- (4- ( ( ( (9H-fluoren-9-yl) methoxy) carbonyl) amino) -2-
methylphenyl) piperidine-1-carboxylate
To a solution of tert-butyl 4- (4-amino-2-methylphenyl) piperidine-1-carboxylate (600 mg, 2.07 mmol) , DIEA (534 mg, 4.14 mmol) in DCM (10 mL) at 0℃ was added Fmoc-Cl (590 mg, 2.28 mmol) . Then the mixture was stirred at 25℃ for 3h, After completion of reaction, Water (100 ml) was added and the solution was extracted with EtOAc (100 mL x 3) . The combined organic layers were washed with brine (100 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. the crude product was purified by silica gel chromatography eluted with PE:EA=5: 1 to afford the desired product.
LCMS: MS (ESI) m/z 535.2 [M+ Na] +
Step D: 9H-fluoren-9-yl) methyl (3-methyl-4- (piperidin-4-yl) phenyl) carbamate
hydrochloride
To a solution of tert-butyl 4- (4- ( ( ( (9H-fluoren-9-yl) methoxy) carbonyl) amino) -2-methylphenyl) piperidine-1-carboxylate (567 mg, 1.11 mmol) in HCl in dioxane (10 mL, 4 M) and DCM (10 mL) was stirred at 25℃ for 3 h. After completion of reaction, the reaction mixture was concentrated under reduced pressure to dryness to afford the desired product.
LCMS: MS (ESI) m/z 413.5 [M+H] +
Step E: tert-butyl 9- ( (4- (4- ( ( ( (9H-fluoren-9-yl) methoxy) carbonyl) amino) -2-
methylphenyl) piperidin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of 9H-fluoren-9-yl) methyl (3-methyl-4- (piperidin-4-yl) phenyl) carbamate hydrochloride (293 mg, 0.710 mmol) , tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-
carboxylate (400 mg, 1.42 mmol) , NaBH (OAc) 3 (301 mg, 1.42 mmol) , CH3COOH (85.2 mg, 1.42 mmol) in DCM (10 mL) was added TEA (215 mg, 2.13 mmol) . Then the mixture was stirred at 25℃ for 16 h. The reaction mixture was quenched with water (100 mL) , and the solution was extracted with DCM (100 mL x 3) . The combined organic layers were washed with brine (100 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography eluted with DCM: MeOH=10: 1 to afford the desired product.
LCMS: MS (ESI) m/z 678.9 [M+H] +
Step F: (9H-fluoren-9-yl) methyl (4- (1- ( (3-azaspiro [5.5] undecan-9-yl) methyl) piperidin-4-
yl) -3-methylphenyl) carbamate hydrochloride
To a solution of tert-butyl 9- ( (4- (4- ( ( ( (9H-fluoren-9-yl) methoxy) carbonyl) amino) -2-methylphenyl) piperidin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate (400 mg, 0.590 mmol) in HCl in dioxane (10 mL, 4 M) and DCM (10 mL) was stirred at 25℃ for 3 h. After completion of reaction, the reaction mixture was concentrated under reduced pressure to dryness to afford the desired product.
LCMS: MS (ESI) m/z 578.9 [M+H] +
Step G: 1- (5- (9- ( (4- (4-amino-2-methylphenyl) piperidin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of (9H-fluoren-9-yl) methyl (4- (1- ( (3-azaspiro [5.5] undecan-9-yl) methyl) piperidin-4-yl) -3-methylphenyl) carbamate hydrochloride (330 mg, 0.570 mmol) , perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (248 mg, 0.570 mmol) in DMF (5 mL) was added TEA (173 mg, 1.71 mmol) . Then the mixture was stirred at 25℃ for 16h. Water (50 mL) was added and the solution was extracted with EtOAc (50 mL x 3) . The combined organic layers were washed with brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. the crude product was purified by silica gel chromatography eluted with DCM: MeOH=5: 1 to afford the desired product.
LCMS: MS (ESI) m/z 606.3 [M+H] +
Step H: 1- {5- [ (9- { [4- (4- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} -2-
tolyl) -1-piperidyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-chlorophenyl} -1, 3, 5, 6-
tetrahydro-2, 4-pyrimidinedione
To a solution of give 1- (5- (9- ( (4- (4-amino-2-methylphenyl) piperidin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (50 mg, 0.0779 mmol) in DCE (10 mL) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-
a] [1, 2] diazacyclotridecin-5-one (20.0 mg, 0.0519 mmol) , the mixture was stirred at 80℃ for 16 hrs under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda, Column: Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 29%~32%; Retention Time: 6.23-7.55min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 949.7 [M+Na] +
1H NMR (400 MHz, DMSO-d6) δ 10.51 (s, 1H) , 10.12 (s, 1H) , 8.86 (s, 1H) , 8.14 (s, 1H) , 8.00 (t, J = 7.8 Hz, 1H) , 7.80 (d, J = 7.8 Hz, 1H) , 7.62 (m, 3H) , 7.55 (s, 1H) , 7.44 (d, J = 8.4 Hz, 1H) , 7.39 (d, J = 8.3 Hz, 1H) , 7.14 (d, J = 8.1 Hz, 1H) , 5.42 –5.23 (m, 3H) , 4.48 (d, J = 5.6 Hz, 2H) , 3.87 –3.69 (m, 2H) , 3.66 –3.53 (m, 4H) , 3.15 (m, 3H) , 2.84 –2.69 (m, 4H) , 2.28 (m, 3H) , 2.22 –2.12 (m, 1H) , 2.08 –1.90 (m, 3H) , 1.86 –1.77 (m, 2H) , 1.72 (m, 5H) , 1.61 (m, 3H) , 1.57 –1.50 (m, 4H) , 1.48 –1.42 (m, 1H) , 1.39 –1.33 (m, 1H) , 1.31 –1.26 (m, 1H) , 1.25-1.21 (m, 1H) , 1.20 –1.06 (m, 4H) .
Example 149:
1- [2-chloro-5- ( {9- [ (4- {4- [6- (2, 6-dichlorophenyl) -8-methyl-5-oxo-7, 8-dihydro-1, 3, 6, 8-tetraaza-2-naphthylamino] -2-tolyl} -1-piperidyl) methyl] -3-aza-3-spiro [5.5] undecyl} carbonyl) phenyl] -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-352)
Scheme:
Step A: 3- (2, 6-dichlorophenyl) -1-methyl-7- (methylsulfinyl) -2, 3-dihydropyrimido [4, 5-
d] pyrimidin-4 (1H) -one
To a solution of 3- (2, 6-dichlorophenyl) -1-methyl-7- (methylthio) -2, 3-dihydropyrimido [4, 5-d] pyrimidin-4 (1H) -one (160.0 mg, 0.451 mmol) in DCM (3 mL) was added m-CPBA (85%purity) (138 mg, 0.677 mmol) , the mixture was stirred at 0℃ for 1 hr under N2 atmosphere. The mixture was used directly.
LCMS: MS (ESI) m/z 371.0 [M+H] +
Step B: 1- [2-chloro-5- ( {9- [ (4- {4- [6- (2, 6-dichlorophenyl) -8-methyl-5-oxo-7, 8-dihydro-
1, 3, 6, 8-tetraaza-2-naphthylamino] -2-tolyl} -1-piperidyl) methyl] -3-aza-3-
spiro [5.5] undecyl} carbonyl) phenyl] -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a solution of 3- (2, 6-dichlorophenyl) -1-methyl-7- (methylsulfinyl) -2, 3-dihydropyrimido [4, 5-d] pyrimidin-4 (1H) -one (20.0 mg, 0.054 mmol) in DCE (3 mL) was added 1- (5- (9- ( (4- (4-amino-2-methylphenyl) piperidin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (39 mg, 0.065 mmol) , the mixture was stirred at 50℃ for 16 hrs under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 8%-38%B over 8 min) to afford the desired product.
LCMS: MS (ESI) m/z 914.5 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ 10.51 (s, 1H) , 9.89 –9.78 (m, 1H) , 9.12 –8.98 (m, 1H) , 8.48 (s, 1H) , 7.65 (m, 4H) , 7.59 (d, J = 9.3 Hz, 1H) , 7.55 (s, 1H) , 7.51 –7.46 (m, 1H) , 7.39 (d, J = 7.4 Hz, 1H) , 7.10 (d, J = 8.3 Hz, 1H) , 4.98 (m, 2H) , 3.62 –3.55 (m, 5H) , 3.31 (d, J = 5.1 Hz, 2H) , 3.12 (m, 3H) , 3.09 –3.05 (m, 2H) , 2.96 (m, 3H) , 2.76 –2.73 (m, 2H) , 2.66 (d, J = 3.7 Hz, 1H) , 2.31 (m, 3H) , 1.96 (d, J = 5.5 Hz, 2H) , 1.89 –1.83 (m, 2H) , 1.78 –1.70 (m, 3H) , 1.63 (dd, J = 13.9, 5.5 Hz, 2H) , 1.55 –1.43 (m, 2H) , 1.39 –1.27 (m, 2H) , 1.22 –1.11 (m, 4H) .
Example 150:
3- {5- [ (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-methoxyphenyl} -2, 6-piperidinedione (Cpd-353)
Scheme:
Step A: 3- {5- [ (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-
methoxyphenyl} -2, 6-piperidinedione
A mixture of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) , 3- [5- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] -2-methoxy-phenyl] piperidine-2, 6-dione (76.24 mg, 129.72 μmol) , AcOH (77.90 mg, 1.30 mmol, 74.26 μL) in n-BuOH (3 mL) was stirred at 100 ℃ for 2 hr. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: C18
100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 15%-45%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 909.6 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.77 (s, 1H) , 10.09 (TFA, 1H) , 9.44 (s, 1H) , 8.84 (s, 1H) , 8.00 (t, J = 8.0 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.61 (dd, J = 8.4, 12.0 Hz, 3H) , 7.32 (dd, J = 2.0, 8.4 Hz, 1H) , 7.20 (d, J = 2.0 Hz, 1H) , 7.05 (d, J = 8.8 Hz, 1H) , 6.98 (d, J = 9.2 Hz, 2H) , 5.33 (d, J = 19.2 Hz, 2H) , 4.47 (d, J = 6.0 Hz, 2H) , 3.99 (dd, J = 5.2, 12.0 Hz, 1H) , 3.79 (m, 3H) , 3.74 (d, J = 12.4 Hz, 2H) , 3.57 (d, J = 10.8 Hz, 3H) , 3.52 -3.31 (m, 3H) , 3.20 -2.95 (m, 6H) , 2.75 -2.62 (m, 1H) , 2.49 -2.43 (m, 1H) , 2.35 -2.11 (m, 2H) , 2.10 -1.87 (m, 3H) , 1.86 -1.67 (m, 4H) , 1.66 -1.59 (m, 2H) , 1.56 (m, 3H) , 1.49 (s, 2H) , 1.32 (s, 2H) , 1.25 -1.07 (m, 4H) .
Example 151:
3- (7- {9- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -3, 9-diaza-3-spiro [5.5] undecyl} -1-oxo-2-phthalazinyl) -2, 6-piperidinedione (Cpd-354)
Scheme:
Step A: 7- (9- (1- (4-nitrophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecan-3-yl) phthalazin-
1 (2H) -one
A mixture of 3- (1- (4-nitrophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecane (200 mg, 0.558mmol) , 7-bromophthalazin-1 (2H) -one (126 mg, 0.558 mmol) , Pd-PEPPSI-IPentCl (27.1 mg, 0.0279 mmol) , Cs2CO3 (544 mg, 1.67 mmol) in dioxane (10 mL) was stirred at 100℃ for 16 hrs under N2. After completion, the mixture was quenched with H2O (10 mL) , extracted with DCM (10 mLx3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product. LCMS: MS (ESI) m/z 503.3 [M+H] +
Step B: 3- (7- (9- (1- (4-nitrophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecan-3-yl) -1-
oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione
A mixture of 7- (9- (1- (4-nitrophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecan-3-yl) phthalazin-1 (2H) -one (120 mg, 0.239 mmol) in DMF (5 mL) was treated sequentially with NaH (60wt. %, 66.8 mg, 1.67 mmol) at 0℃ for 1 h. Then 3-bromopiperidine-2, 6-dione (459 mg, 2.39 mmol)
was added. The reaction was stirred at 20℃ under N2 atmosphere for 16 hrs. The mixture was diluted with H2O (5 mL) , extracted with EtOAc (10 mLx3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 614.2 [M+H] +
Step C: 3- (7- (9- (1- (4-aminophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecan-3-yl) -1-
oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione
A mixture of 3- (7- (9- (1- (4-nitrophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecan-3-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (50 mg) as a yellow solid (50.0 mg, 0.0815 mmol) , iron powder (22.8mg, 0.408 mmol) , NH4Cl (21.8 mg, 0.408 mmol) in EtOH (10 mL) and H2O (2 mL) was stirred at 90℃ for 2 hrs. After completion of reaction, the mixture reaction was filtered through a Celite pad, and the filtrate was concentrated to give the crude. The crude was quenched with H2O (10 mL) , adjusted to pH 8 with solid NaHCO3 and extracted with DCM/MeOH (10: 1; 3 x 10 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered, concentrated and purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 583.3 [M+H] +
Step D: 3- (7- {9- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -3, 9-diaza-3-spiro [5.5] undecyl} -1-oxo-2-phthalazinyl) -2, 6-
piperidinedione
To a solution of (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20.0 mg, 0.0519 mmol) in DCM (3 mL) was added 3- (7- (9- (1- (4-aminophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecan-3-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (45.0 mg, 0.248 mmol) in DCM (7 mL) . Then the reaction mixture was stirred at 40℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EA (20 mL x3) . The organic layers were concentrated. The residue was purified by prep-HPLC (Waters 2767, Column: Pursuit XRs C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 16%~21%Retention Time: 8.9-10.8 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 905.7 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ 11.00 (s, 1H) , 10.03 (brs, 1H) , 8.83 (s, 1H) , 8.25 (s, 1H) , 8.15 (s, 0.55 H, FA) , 8.02 (brs, 1H) , 7.82 -7.73 (m, 2H) , 7.65 -7.54 (m, 4H) , 7.50 (s, 1H) , 6.95
(d, J = 8.8 Hz, 2H) , 5.80 –5.69 (m, 1H) , 5.45 –5.21 (m, 3H) , 4.54 –4.40 (m, 2H) , 3.82 -3.70 (m, 2H) , 3.50 -3.43 (m, 4H) , 2.97 -2.87 (m, 2H) , 2.68 -2.60 (m, 5H) , 2.36 –2.29 (m, 2H) , 2.11 –1.91 (m, 6H) , 1.73 –1.55 (m, 14H) .
Example 152:
3- (6- {9- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -3, 9-diaza-3-spiro [5.5] undecyl} -1-oxo-2-phthalazinyl) -2, 6-piperidinedione (Cpd-355)
Scheme:
Step A: 6- (9- (1- (4-nitrophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecan-3-yl) phthalazin-
1 (2H) -one
A mixture of 3- (1- (4-nitrophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecane (350 mg, 0.978 mmol) , 6-bromophthalazin-1 (2H) -one (220 mg, 0.978 mmol) , Pd-PEPPSI-IPentCl (47.6 mg, 0.0489 mmol) , Cs2CO3 (956 mg, 2.93 mmol) in dioxane (15 mL) was stirred at 100℃ for 16 hrs under N2. After completion, the mixture was quenched with H2O (10 mL) , extracted with DCM (10 mLx3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 503.3 [M+H] +
Step B: 3- (6- (9- (1- (4-nitrophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecan-3-yl) -1-
oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione
A mixture of 6- (9- (1- (4-nitrophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecan-3-yl) phthalazin-1 (2H) -one (180 mg, 0.358 mmol) in DMF (8.0 mL) was treated sequentially with NaH (60wt. %, 100 mg, 2.51 mmol) at 0℃ for 1 h. Then 3-bromopiperidine-2, 6-dione (687 mg, 3.58 mmol) was added. The reaction was stirred at 20℃ under N2 atmosphere for 16 hrs. After completion, the mixture was diluted with H2O (5 mL) , extracted with EtOAc (10 mLx3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 613.8 [M+H] +
Step C: 3- (6- (9- (1- (4-aminophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecan-3-yl) -1-
oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione
A mixture of 3- (6- (9- (1- (4-nitrophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecan-3-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (170 mg, 0.277 mmol) , iron powder (77.6 mg, 1.39 mmol) , NH4Cl (74.4 mg, 1.39 mmol) in EtOH (10 mL) and H2O (2 mL) was stirred at 90℃ for 2 hrs. After completion of reaction, the mixture reaction was filtered through a Celite pad, and the filtrate was concentrated to give the crude. The crude was quenched with H2O (10 mL) , adjusted to pH 8 with solid NaHCO3 and extracted with EtOAc (3 x 30 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered, concentrated and purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 583.2 [M+H] +
Step D: 3- (6- {9- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -3, 9-diaza-3-spiro [5.5] undecyl} -1-oxo-2-phthalazinyl) -2, 6-
piperidinedione
To a solution of (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (50.0 mg, 0.0519 mmol) in DCM (3 mL) was added 3- (6- (9- (1- (4-aminophenyl) piperidin-4-yl) -3, 9-diazaspiro [5.5] undecan-3-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (75.0 mg, 0.135 mmol) in DCM (7 mL) . Then the reaction mixture was stirred at 40℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EA (20 mL x3) . The organic layers were concentrated. The residue was purified by prep-HPLC (Waters 2767, Column: Pursuit XRs C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 16%~21%Retention Time: 9.0-10.9 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 905.7 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H) , 10.03 (brs, 1H) , 8.82 (s, 1H) , 8.27 -8.16 (m, 2H) , 8.08 -7.96 (m, 2H) , 7.76 (d, J = 8.1 Hz, 1H) , 7.62 -7.52 (m, 3H) , 7.50 -7.44 (m, 1H) , 7.25 -7.19 (m, 1H) , 6.92 (d, J = 9.0 Hz, 2H) , 5.74 (dd, J = 11.9, 5.1 Hz, 1H) , 5.57 –5.08 (m, 3H) , 4.53 -4.39 (m, 2H) , 3.69 (d, J = 11.6 Hz, 2H) , 3.45 –3.43 (m, 4H) , 2.98 –2.85 (m, 2H) , 2.69 -2.59 (m, 5H) , 2.30 –2.02 (m, 4H) , 1.97 –1.81 (m, 3H) , 1.79 –1.70 (m, 1H) , 1.64 -1.45 (m, 14H) .
Example 153:
3- {6- [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] -1-oxo-2-phthalazinyl} -2, 6-piperidinedione (Cpd-356)
Scheme:
Step A: 3- (6-bromo-1-oxo-phthalazin-2-yl) -1- [ (4-methoxyphenyl) methyl] piperidine-2, 6-
dione
To a solution of 6-bromo-2H-phthalazin-1-one (1.0 g, 4.44 mmol) in THF (20 mL) was added t-BuOK (598 mg, 5.33 mmol) at 0 ℃ under nitrogen atmosphere. The mixture was stirred at 0 ℃for 0.5 hr. Then a solution of [1- [ (4-methoxyphenyl) methyl] -2, 6-dioxo-3-piperidyl] trifluoromethanesulfonate (1.86 g, 4.89 mmol) in THF (10 mL) was added to the reaction mixture. The reaction mixture was stirred at 0 ℃ for 2 hr. The reaction mixture was quenched by addition NH4Cl (50 mL) , extracted with ethyl acetate (50 mL×3) . The combined organic layers were washed with brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was triturated with ethyl acetate (10.0 mL) at 20 ℃ for 30 min, filtered and the filtered cake was dried in vacuum to afford the desired product. LCMS: MS (ESI) m/z 456.1, 458.1 [M+H] +
1H NMR (400 MHz, CDCl3) δ 8.30 (d, J=8.4 Hz, 1H) , 8.11 (s, 1H) , 7.92 -7.86 (m, 2H) , 7.35 (d, J=8.8 Hz, 2H) , 6.85 -6.79 (m, 2H) , 5.86 (dd, J=5.2, 12.0 Hz, 1H) , 5.06 -4.80 (m, 2H) , 3.79 (m, 3H) , 3.01 (td, J=3.6, 16.8 Hz, 1H) , 2.89 -2.78 (m, 1H) , 2.78 -2.66 (m, 1H) , 2.29 -2.18 (m, 1H) .
Step B: 3- [6- [4- (hydroxymethyl) -1-piperidyl] -1-oxo-phthalazin-2-yl] -1- [ (4-
methoxyphenyl) methyl] piperidine-2, 6-dione
To a solution of 3- (6-bromo-1-oxo-phthalazin-2-yl) -1- [ (4-methoxyphenyl) methyl] piperidine-2, 6-dione (300 mg, 657.4 μmol) , K3PO4 (419 mg, 1.97 mmol) and 4-piperidylmethanol (114 mg, 986.2 μmol) in dioxane (5.0 mL) were added Pd2 (dba) 3 (60 mg, 65.7 μmol) and RuPhos (15 mg, 32.8 μmol) . The mixture was stirred at 100 ℃ for 15 hr. under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was purified by flash silica gel chromatography (4 g Silica Flash Column, Eluent of 0~2.5%MeOH/DCM gradient @25 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 491.3 [M+H] +
1H NMR (400 MHz, CDCl3) δ 8.26 -8.17 (m, 1H) , 8.01 (s, 1H) , 7.36 (d, J=8.8 Hz, 2H) , 7.33 -7.29 (m, 1H) , 6.90 (d, J=2.4 Hz, 1H) , 6.85 -6.79 (m, 2H) , 5.94 -5.76 (m, 1H) , 5.03 -4.84 (m, 2H) , 3.71 (m, 3H) , 3.57 (d, J=6.4 Hz, 1H) , 3.04 -2.90 (m, 2H) , 2.89 -2.70 (m, 2H) , 2.28 -2.16 (m, 1H) , 1.91 (d, J=12.8 Hz, 2H) , 1.81 (s, 1H) , 1.73 -1.52 (m, 3H) , 1.47 -1.34 (m, 2H) .
Step C: 1- [2- [1- [ (4-methoxyphenyl) methyl] -2, 6-dioxo-3-piperidyl] -1-oxo-phthalazin-6-
yl] piperidine-4-carbaldehyde
To a solution of 3- [6- [4- (hydroxymethyl) -1-piperidyl] -1-oxo-phthalazin-2-yl] -1- [ (4-methoxyphenyl) methyl] piperidine-2, 6-dione (300 mg, 611.5 μmol) in DMSO (3.0 mL) was added IBX (240 mg, 856.1 μmol) . The mixture was stirred at 20 ℃ for 1hr. The reaction mixture was diluted with water (20.0 mL) and extracted with ethyl acetate (10.0 mL×3) . The combined organic layer was washed with brine (15 mL) , dried over anhydrous Na2SO4, filtered and concentrated in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 489.3 [M+H] +.
Step D: 1- [ (4-methoxyphenyl) methyl] -3- [6- [4- [ [4- [4- (4-nitrophenyl) piperazin-1-yl] -1-
piperidyl] methyl] -1-piperidyl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 1- [2- [1- [ (4-methoxyphenyl) methyl] -2, 6-dioxo-3-piperidyl] -1-oxo-phthalazin-6-yl] piperidine-4-carbaldehyde (300 mg, 614.0 μmol) and 1- (4-nitrophenyl) -4- (4-piperidyl) piperazine (196 mg, 675.4 μmol) in DCM (5.0 mL) was added TEA (311 mg, 3.07 mmol) at 20 ℃. After stirring at 20 ℃ for 2 hr. NaBH (OAc) 3 (260 mg, 1.23 mmol) was added to the reaction mixture. The mixture was stirred at 20 ℃ for 0.5 hr. The reaction mixture was quenched by addition NaHCO3 (10.0 mL) , extracted with DCM (10 mL ×3) . The combined
organic layers were washed with brine (15 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 2~5%MeOH/DCM gradient @20 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 763.4 [M+H] +.
Step E: 3- [6- [4- [ [4- [4- (4-nitrophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-
oxo-phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 1- [ (4-methoxyphenyl) methyl] -3- [6- [4- [ [4- [4- (4-nitrophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (120 mg, 157.3 μmol) in DCE (6.0 mL) was added AlCl3 (210 mg, 1.57 mmol) . The mixture was stirred at 85 ℃ for 1hr. The reaction mixture was quenched by addition water (10.0 mL) at 20 ℃, extracted with a solution of DCM/MeOH (10/1, 10 mL × 5) . The combined organic layers were washed with brine (15 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (4 g Silica Flash Column, Eluent of 10~20%Ethyl acetate/Petroleum ether gradient @18 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 643.4 [M+H] +.
Step F: 3- [6- [4- [ [4- [4- (4-aminophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-
oxo-phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 3- [6- [4- [ [4- [4- (4-nitrophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (140 mg, 217.8 μmol) and TEA (44 mg, 435.6 μmol) in DCM (5.0 mL) and DMAC (1.0 mL) were added PdCl2 (39 mg, 217.8 μmol) and Et3SiH (506 mg, 4.36 mmol) . The mixture was stirred at 20 ℃ for 15hr under nitrogen atmosphere. The reaction mixture was concentrated in vacuum by oil pump. The residue was purified by prep-TLC (SiO2, DCM: MeOH =10: 1, 1%TEA) to afford the desired product.
LCMS: MS (ESI) m/z 613.5 [M+H] +.
Step G: 3- {6- [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] -1-oxo-2-phthalazinyl} -
2, 6-piperidinedione
To a suspension of 3- [6- [4- [ [4- [4- (4-aminophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (70 mg, 114.2 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (30 mg, 77.8 μmol) in n-BuOH (1.5 mL) was added AcOH (47 mg, 778.3 μmol) . The mixture was stirred at 100 ℃ for 2 hr under nitrogen
atmosphere. The reaction mixture was concentrated in vacuum. The residue was purified by prep-HPLC (column: Welch Xtimate C18 150*25mm*5um; mobile phase: [water (TFA) -ACN] ; gradient: 20%-50%B over 7 min) to afford the desired product after freeze-drying.
LCMS: MS (ESI) m/z 934.8 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H) , 10.08 (s, 1H) , 8.85 (s, 1H) , 8.24 (s, 1H) , 8.05 (d, J=8.8 Hz, 1H) , 7.99 (t, J=7.6 Hz, 1H) , 7.75 (d, J=8.0 Hz, 1H) , 7.62 (dd, J=8.4, 13.6 Hz, 3H) , 7.51 (d, J=8.4 Hz, 1H) , 7.27 (s, 1H) , 7.00 (d, J=8.8 Hz, 2H) , 5.79 -5.70 (m, 1H) , 5.37 -5.23 (m, 2H) , 4.48 (m, 2H) , 4.07 (d, J=11.6 Hz, 2H) , 3.77 -3.57 (m, 4H) , 3.06 -2.87 (m, 10H) , 2.74 -2.57 (m, 3H) , 2.40 -2.25 (m, 3H) , 2.24 -1.91 (m, 9H) , 1.86 (d, J=10.4 Hz, 2H) , 1.79 -1.66 (m, 2H) , 1.57 (m, 3H) , 1.39 -1.21 (m, 3H) .
Example 154:
3- {7- [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] -1-oxo-2-phthalazinyl} -2, 6-piperidinedione (Cpd-357)
Scheme:
Step A: 3- (7-bromo-1-oxo-phthalazin-2-yl) -1- [ (4-methoxyphenyl) methyl] piperidine-2, 6-
dione
To a solution of 7-bromo-2H-phthalazin-1-one (400 mg, 1.78 mmol) in THF (10 mL) was added t-BuOK (239 mg, 2.13 mmol) at 0 ℃. After stirring at 0 ℃ for 0.5 hr. under nitrogen atmosphere, [1- [ (4-methoxyphenyl) methyl] -2, 6-dioxo-3-piperidyl] trifluoromethanesulfonate (746 mg, 1.96 mmol) was added to the reaction mixture at 0 ℃. After the addition was finished, the reaction mixture was stirred at 0 ℃ for 2 hr. The reaction mixture was quenched by addition NH4Cl (30 mL) , extracted with ethyl acetate (20 mL×3) . The combined organic layers were washed with brine (20.0 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was triturated with ethyl acetate (10.0 mL) at 20 ℃ for 30 min, filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 456.1, 458.1 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.60 (s, 1H) , 8.16 (s, 1H) , 7.95 (dd, J=2.0, 8.4 Hz, 1H) , 7.60 (d, J=8.4 Hz, 1H) , 7.35 (d, J=8.8 Hz, 2H) , 6.83 (d, J=8.8 Hz, 2H) , 5.86 (dd, J=5.2, 11.6 Hz, 1H) , 5.03 -4.85 (m, 2H) , 3.79 (s, 3H) , 3.09 -2.95 (m, 1H) , 2.89 -2.67 (m, 2H) , 2.31 -2.17 (m, 1H) .
Step B: 3- [7- [4- (hydroxymethyl) -1-piperidyl] -1-oxo-phthalazin-2-yl] -1- [ (4-
methoxyphenyl) methyl] piperidine-2, 6-dione
To a solution of 3- (7-bromo-1-oxo-phthalazin-2-yl) -1- [ (4-methoxyphenyl) methyl] piperidine-2, 6-dione (460 mg, 1.01 mmol) , K3PO4 (642 mg, 3.02 mmol) and 4-piperidylmethanol (174 mg, 1.51 mmol) in dioxane (6.0 mL) were added Pd2 (dba) 3 (92 mg, 100.8 μmol) and RuPhos (24 mg, 50.4 μmol) . The mixture was stirred at 100 ℃ for 15 hr. under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~2.5%MeOH/DCM gradient @20 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 491.3 [M+H] +.
Step C: 1- [3- [1- [ (4-methoxyphenyl) methyl] -2, 6-dioxo-3-piperidyl] -4-oxo-phthalazin-6-
yl] piperidine-4-carbaldehyde
To a solution of 3- [7- [4- (hydroxymethyl) -1-piperidyl] -1-oxo-phthalazin-2-yl] -1- [ (4-methoxyphenyl) methyl] piperidine-2, 6-dione (470 mg, 958.1 μmol) in DMSO (4.0 mL) was added IBX (376 mg, 1.34 mmol) . The mixture was stirred at 20 ℃ for 15 hr. . The reaction mixture was poured into water (40.0 mL) , yellow solid was precipitated. The suspension was filtered and rinsed with water (5.0 mL×3) . The filtered cake was dissolved with ethyl acetate (20 mL) , washed with brine (10.0 mL) , dried over anhydrous Na2SO4, filtered and concentrated in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 489.1 [M+H] +.
Step D: 1- [ (4-methoxyphenyl) methyl] -3- [7- [4- [ [4- [4- (4-nitrophenyl) piperazin-1-yl] -1-
piperidyl] methyl] -1-piperidyl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 1- [3- [1- [ (4-methoxyphenyl) methyl] -2, 6-dioxo-3-piperidyl] -4-oxo-phthalazin-6-yl] piperidine-4-carbaldehyde (468 mg, 957.9 μmol) and 1- (4-nitrophenyl) -4- (4-piperidyl) piperazine (306 mg, 1.05 mmol) in DCM (6.0 mL) was added TEA (485 mg, 4.79 mmol) at 20 ℃ under nitrogen atmosphere . After stirring at 20 ℃ for 15 hr., NaBH (OAc) 3 (406 mg, 1.92 mmol) was added to the reaction mixture. The reaction mixture was stirred at 20 ℃ for 0.5 hr. The reaction mixture was quenched by addition NaHCO3 (15.0mL) , extracted with DCM (10 mL×3) . The combined organic layers were washed with brine (15 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel
chromatography (12 gSilica Flash Column, Eluent of 1~2%MeOH/DCM gradient @20 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 763.4 [M+H] +.
Step E: 3- [7- [4- [ [4- [4- (4-nitrophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-
oxo-phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 1- [ (4-methoxyphenyl) methyl] -3- [7- [4- [ [4- [4- (4-nitrophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (260 mg, 340.8 μmol) in DCE (15 mL) was added AlCl3 (454 mg, 3.41 mmol) . The mixture was stirred at 85 ℃for 3hr under nitrogen atmosphere. The reaction mixture was quenched by addition water (10.0 mL) , extracted with DCM/MeOH (10/1, 10 mL ×4) . The combined organic layers were washed with brine (15 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 10~20%MeOH/DCM gradient @20 mL/min) to afford the desired product. LCMS: MS (ESI) m/z 643.3 [M+H] +.
1H NMR (400 MHz, CDCl3) δ8.14 -8.05 (m, 2H) , 8.01 (br d, J=4.0 Hz, 1H) , 7.63 (br s, 1H) , 7.58 -7.50 (m, 1H) , 7.34 (br d, J=8.8 Hz, 1H) , 6.85 -6.77 (m, 2H) , 5.77 (d, J=6.4 Hz, 1H) , 3.98 (d, J=11.1 Hz, 2H) , 3.48 -3.39 (m, 6H) , 3.38 -3.32 (m, 1H) , 2.94 -2.90 (m, 8H) , 2.78 -2.66 (m, 6H) , 1.99 -1.85 (m, 6H) , 1.35 (br d, J=10.0 Hz, 2H) , 1.22 (br s, 1H) .
Step F: 3- [7- [4- [ [4- [4- (4-aminophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-
oxo-phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 3- [7- [4- [ [4- [4- (4-nitrophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (180 mg, 280.0 μmol) and TEA (57 mg, 560.1 μmol) in DCM (6 mL) and DMAc (1.5 mL) were added PdCl2 (50 mg, 280.0 μmol) and Et3SiH (651 mg, 5.60 mmol) . The mixture was stirred at 20 ℃ for 15hr under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 10~20%MeOH/DCM gradient @20 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 613.4 [M+H] +.
Step G: 3- {7- [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] -1-oxo-2-phthalazinyl} -
2, 6-piperidinedione
To a suspension of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (40 mg, 103.7 μmol) and 3- [7- [4- [ [4- [4- (4-aminophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-
oxo-phthalazin-2-yl] piperidine-2, 6-dione (128 mg, 208.8 μmol) in n-BuOH (5.0 mL) was added AcOH (62 mg, 1.04 mmol) . The mixture was stirred at 100 ℃ for 15 hr. under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 6%-36%B over 8 min) , after freeze-dying to afford the desired product.
LCMS: MS (ESI) m/z 934.8 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.00 (s, 1H) , 10.08 (s, 1H) , 9.44 (s, 1H) , 8.84 (s, 1H) , 8.25 (s, 1H) , 7.99 (t, J=8.0 Hz, 1H) , 7.77 (dd, J=8.4, 13.2 Hz, 2H) , 7.67 -7.58 (m, 4H) , 7.52 (s, 1H) , 7.00 (d, J=8.8 Hz, 2H) , 5.74 (dd, J=5.6, 12.0 Hz, 1H) , 5.42 -5.24 (m, 2H) , 4.47 (d, J=5.6 Hz, 2H) , 4.05 (d, J=12.0 Hz, 2H) , 3.95 -3.74 (m, 6H) , 3.37 -3.19 (m, 1H) , 3.14 -2.80 (m, 10H) , 2.69 -2.51 (m, 4H) , 2.34 (d, J=11.6 Hz, 2H) , 2.27 -1.91 (m, 8H) , 1.87 (d, J=12.4 Hz, 2H) , 1.78 -1.67 (m, 1H) , 1.57 (s, 3H) , 1.38 -1.23 (m, 2H) .
Example 155:
N- (2, 6-dioxo-3-piperidyl) -5- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -2-pyrimidinecarboxamide (Cpd-358)
Scheme:
Step A: tert-butyl 8- (2-methoxycarbonylpyrimidin-5-yl) -2, 8-diazaspiro [4.5] decane-2-
carboxylate
A mixture of methyl 5-bromopyrimidine-2-carboxylate (1 g, 4.61 mmol) , tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (1.22 g, 5.07 mmol) , Pd2 (dba) 3 (210.98 mg, 230.39 μmol) , RuPhos (107.51 mg, 230.39 μmol) , K3PO4 (2.93 g, 13.82 mmol) in dioxane (5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 16hr
under N2 atmosphere. The mixture was concentrated to give a residue. The residue was purified by silica gel column chromatography (80%EtOAc in PE) to afford the desired product.
LCMS: MS (ESI) m/z 377.0 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 8.55 (s, 2H) , 3.82 (s, 3H) , 3.45 (d, J = 7.6 Hz, 4H) , 3.34 -3.29 (m, 2H) , 3.14 (s, 2H) , 1.76 (t, J = 6.0 Hz, 2H) , 1.64 -1.54 (m, 4H) , 1.40 (s, 9H) .
Step B: methyl 5- (2, 8-diazaspiro [4.5] decan-8-yl) pyrimidine-2-carboxylate
To a solution of tert-butyl 8- (2-methoxycarbonylpyrimidin-5-yl) -2, 8-diazaspiro [4.5] decane-2-carboxylate (1.5 g, 3.98 mmol) in DCM (10 mL) was added TFA (4.61 g, 40.39 mmol, 3 mL) . The mixture was stirred at 20 ℃ for 2 hr. The reaction mixture was concentrated and dried by lyophilization to give to afford the desired product.
LCMS: MS (ESI) m/z 276.8 [M+H] +.
Step C: methyl 5- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-
yl] pyrimidine-2-carboxylate
To a solution of methyl 5- (2, 8-diazaspiro [4.5] decan-8-yl) pyrimidine-2-carboxylate (1 g, 3.62 mmol) , 1- (4-nitrophenyl) piperidin-4-one (956.34 mg, 4.34 mmol) in DCM (20 mL) was added dropwise TEA (1.83 g, 18.09 mmol, 2.52 mL) . After addition, the mixture was stirred at 20 ℃for 2 hr, and then NaBH (OAc) 3 (2.30 g, 10.86 mmol) was added. The resulting mixture was stirred at 20 ℃ for 14 hr. The mixture was poured into water (100 mL) and extracted with DCM (100 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by silica gel column chromatography (5%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 481.4 [M+H] +.
Step D: 5- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] pyrimidine-2-
carboxylic acid
To a solution of methyl 5- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] pyrimidine-2-carboxylate (1.2 g, 2.50 mmol) in THF (10 mL) , MeOH (10 mL) and H2O (5 mL) was added LiOH. H2O (523.94 mg, 12.49 mmol) . The mixture was stirred at 50 ℃ for 2 hr. The mixture was poured into aq. HCl (10 mL, 2 M) and then the mixture was filtered and the solid was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 467.3 [M+H] +.
Step E: N- (2, 6-dioxo-3-piperidyl) -5- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-
diazaspiro [4.5] decan-8-yl] pyrimidine-2-carboxamide
To a solution of 5- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] pyrimidine-2-carboxylic acid (1 g, 2.14 mmol) , 3-aminopiperidine-2, 6-dione (529.19 mg, 3.22 mmol, HCl) , TEA (1.08 g, 10.72 mmol, 1.49 mL) in DMF (10 mL) was added HATU (1.22 g, 3.22 mmol) .
The mixture was stirred at 20 ℃ for 2hr. The reaction mixture was poured into water (50 mL) . Then extracted with DCM (100 mL × 2) . The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated to give a residue. The residue was purified by silica gel column chromatography (23%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 577.4 [M+H] +.
Step F: 5- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -N- (2, 6-dioxo-3-
piperidyl) pyrimidine-2-carboxamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -5- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] pyrimidine-2-carboxamide (500 mg, 867.08 μmol) in DCM (5 mL) and DMAC (1 mL) was added PdCl2 (153.76 mg, 867.08 μmol) , TEA (175.48 mg, 1.73 mmol) and Et3SiH (2.02 g, 17.34 mmol) . The mixture was stirred at 25 ℃ for 16 hr. The reaction mixture was concentrated to give a residue. The residue was purified by silica gel column chromatography (28%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 547.2 [M+H] +.
Step G: N- (2, 6-dioxo-3-piperidyl) -5- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -2-pyrimidinecarboxamide
A mixture of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) , 5- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -N- (2, 6-dioxo-3-piperidyl) pyrimidine-2-carboxamide (70.91 mg, 129.72 μmol) , AcOH (77.90 mg, 1.30 mmol, 74.26 μL) in n-BuOH (3 mL) was stirred at 100 ℃ for 2 hr. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 5%-35%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 868.7 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.88 (s, 1H) , 10.08 (TFA, 1H) , 9.75 (s, 1H) , 8.85 (d, J = 8.4 Hz, 1H) , 8.84 (s, 1H) , 8.60 (s, 2H) , 8.01 (t, J = 8.0 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.59 (d, J = 8.0 Hz, 3H) , 6.98 (d, J = 9.2 Hz, 2H) , 5.45 -5.20 (m, 2H) , 4.82 -4.69 (m, 1H) , 4.55 -4.37 (m, 2H) , 3.81 (d, J = 11.6 Hz, 2H) , 3.49 -3.45 (m, 5H) , 3.40 -3.20 (m, 2H) , 3.11 -3.03 (m, 1H) , 2.86 -2.74 (m, 1H) , 2.69 (t, J = 11.2 Hz, 2H) , 2.54 (s, 1H) , 2.28 -1.90 (m, 9H) , 1.89 -1.61 (m, 9H) , 1.56 (s, 3H) .
Example 156:
N- (2, 6-dioxo-3-piperidyl) 3- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -2-fluorobenzamide (Cpd-359)
Scheme:
Step A: tert-butyl 8- (2-fluoro-3- (methoxycarbonyl) phenyl) -2, 8-diazaspiro [4.5] decane-2-
carboxylate
To a solution of methyl 3-bromo-2-fluorobenzoate (500 mg, 2.15 mmol) in dioxane (10.0 mL) was added tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (515 mg, 2.15 mmol) , and Cs2CO3 (1.40 g, 4.30 mmol) and Pd2 (dba) 3 (200 mg 0.215 mmol) and BINAP (134 mg, 0.215 mmol) . Then the reaction was stirred at 120 ℃ for 16 hrs under N2 protection. The mixture was quenched by the addition of 100 mL water, the resulting solution was extracted with EtOAc (3*100 mL) , the organic layers was combined and concentrated. The residue was purified by flash silica gel column chromatography (PE: EtOAc =3: 1) to afford the desired product.
LCMS: MS (ESI) m/z 393.1 [M+H] +.
Step B: methyl 2-fluoro-3- (2, 8-diazaspiro [4.5] decan-8-yl) benzoate hydrochloride
To a solution of tert-butyl 8- (2-fluoro-3- (methoxycarbonyl) phenyl) -2, 8-diazaspiro [4.5] decane-2-carboxylate (650 mg, 1.66 mmol) in DCM (5.0 mL) was added HCl in dioxane (5.0 mL) and stirred at 35 ℃ for 1 hrs. The mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 293.0 [M+H] +.
Step C: methyl 2-fluoro-3- (2- (1- (4-nitrophenyl) piperidin-4-yl) -2, 8-diazaspiro [4.5] decan-8-
yl) benzoate
A solution of methyl 2-fluoro-3- (2, 8-diazaspiro [4.5] decan-8-yl) benzoate hydrochloride (480 mg 1.64 mmol) , 1- (4-nitrophenyl) piperidin-4-one (724 mg 3.28 mmol) and NaBH3CN (206 mg 3.28 mmol) in MeOH (20.0 mL) and TEA (496 mg , 4.92 mmol) and HOAc (197 mg 3.28 mmol) was stirred at 60 ℃ for 16 hrs. The mixture was quenched by the addition of 100 mL water. The resulting solution was extracted with EtOAc (3*100 mL) , the organic layers was combined and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 497.3 [M+H] +.
Step D: 2-fluoro-3- (2- (1- (4-nitrophenyl) piperidin-4-yl) -2, 8-diazaspiro [4.5] decan-8-
yl) benzoic acid
To a solution of methyl 2-fluoro-3- (2- (1- (4-nitrophenyl) piperidin-4-yl) -2, 8-diazaspiro [4.5] decan-8-yl) benzoate (600 mg, 1.21 mmol) in THF (3 mL) and MeOH (3 mL) and H2O (3 mL) was added LiOH. H2O (87 mg, 3.63 mmol) . Then the reaction was stirred at 60 ℃for 1 hrs under N2 protection. After completion of reaction, the pH was adjusted with 12N HCl to become acidic, quenched with water (100 mL) , extracted with DCM (3*100 mL) . The organic layers were washed with brine (50 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 483.3 [M+H] +.
Step E: N- (2, 6-dioxopiperidin-3-yl) -2-fluoro-3- (2- (1- (4-nitrophenyl) piperidin-4-yl) -2, 8-
diazaspiro [4.5] decan-8-yl) benzamide
To a solution of 2-fluoro-3- (2- (1- (4-nitrophenyl) piperidin-4-yl) -2, 8-diazaspiro [4.5] decan-8-yl) benzoic acid (450 mg, 0.934 mmol) in DMF (5.00 mL) was added 3-aminopiperidine-2, 6-dione hydrochloride (139 mg, 0.841 mmol) , DIEA (361 mg, 2.80 mmol) , HATU (532 mg 1.40 mmol) . The mixture was stirred at 35 ℃ for 2 hrs. After completion of reaction, quenched with water (50 mL) , extracted with EtOAc (3*150 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 593.3 [M+H] +.
Step F: 3- (2- (1- (4-aminophenyl) piperidin-4-yl) -2, 8-diazaspiro [4.5] decan-8-yl) -N- (2, 6-
dioxopiperidin-3-yl) -2-fluorobenzamide
To a solution of N- (2, 6-dioxopiperidin-3-yl) -2-fluoro-3- (2- (1- (4-nitrophenyl) piperidin-4-yl) -2, 8-diazaspiro [4.5] decan-8-yl) benzamide (490 mg, 0.216 mmol) in EtOH (20.0 mL) and H2O (6.0 mL) was added NH4Cl (231 mg, 4.14 mmol) , Fe (242 mg, 4.14 mmol) . The mixture was stirred at 90 ℃ for 2 hrs. After completion of reaction, the reaction was filtered. The filtrate was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 563.3 [M+H] +.
Step G: N- (2, 6-dioxo-3-piperidyl) 3- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -2-fluorobenzamide
To a solution of (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20 mg, 0.0519 mmol) in DCM (10 mL) was added 3- (2- (1- (4-aminophenyl) piperidin-4-yl) -2, 8-diazaspiro [4.5] decan-8-yl) -N- (2, 6-dioxopiperidin-3-yl) -2-fluorobenzamide (70 mg, 0.104 mmol) . Then the reaction mixture was stirred at 35℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with DCM (20 mL x3) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated. The residue was purified by prep-HPLC (Waters 2767/Qda, Column: XBridge C18, 19*250mm, 10 um; Mobile Phase A: 10mmol/L%NH4HCO3/H2O, B: ACN ; flow rate: 20ml/min; gradient: 37%~37%; Retention Time: 7.50-9.23min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 884.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.03 (brs, 1H) , 8.82 (s, 1H) , 8.57 (d, J = 8.5 Hz, 1H) , 8.07 -7.93 (m, 1H) , 7.76 (d, J = 8.1 Hz, 1H) , 7.65 –7.47 (m, 3H) , 7.24 -7.07 (m, 3H) , 6.91 (d, J = 9.0 Hz, 2H) , 5.45 -5.20 (m, 3H) , 4.81 –4.70 (m, 1H) , 4.53 -4.35 (m, 2H) , 3.65 -3.50 (m, 2H) , 3.33 -3.28 (m, 2H) , 3.06 -2.91 (m, 4H) , 2.84 –2.72 (m, 2H) , 2.69 -2.65 (m, 1H) , 264 -2.58 (m, 2H) , 2.25 -2.16 (m, 1H) , 2.15 –1.96 (m, 5H) , 1.95 -1.86 (m, 2H) , 1.76 -1.59 (m, 8H) , 1.56 (s, 3H) , 1.53 -1.46 (m, 2H) . 、
Example 157:
1- (5- { [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] carbonyl} -2-chlorophenyl) -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-360)
Scheme:
Step A: tert-butyl 4- ( (4- (4- (4-nitrophenyl) piperazin-1-yl) piperidin-1-yl) methyl) piperidine-
1-carboxylate
To a solution of 1- (4-nitrophenyl) -4- (piperidin-4-yl) piperazine (220 mg, 0.759 mmol) , tert-butyl 4-formylpiperidine-1-carboxylate (162 mg, 0.759 mmol) in DCM (15 mL) was added NaBH (OAc) 3 (880 mg, 4.15 mmol) . The resulting mixture was stirred for 16 h at 25℃ in an oil bath. The residue was purified by flash silica gel column chromatography (DCM: MeOH =20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 488.5 [M+H] +.
Step B: 1- (4-nitrophenyl) -4- (1- (piperidin-4-ylmethyl) piperidin-4-yl) piperazine
hydrochloride
To a solution of tert-butyl 4- ( (4- (4- (4-nitrophenyl) piperazin-1-yl) piperidin-1-yl) methyl) piperidine-1-carboxylate (280 mg, 0.575 mmol) , HCl in dioxane (1M 5 ml) \in DCM (15 mL) . Then the mixture was stirred at 35℃ for 2 h. The resulting mixture was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 388.2 [M+H] +.
Step C: 1- (2-chloro-5- (4- ( (4- (4- (4-nitrophenyl) piperazin-1-yl) piperidin-1-
yl) methyl) piperidine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (4-nitrophenyl) -4- (1- (piperidin-4-ylmethyl) piperidin-4-yl) piperazine hydrochloride (230 mg, 0.594 mmol) in DMF (10 mL) was added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (260 mg, 0.598 mmol) and DIPEA (230 mg, 1.78 mmol) . Then the reaction mixture was stirred at 25℃ for 1 h under N2 atmosphere. After completion of reaction, diluted with water (20 mL) , and the solution was extracted with EtOAc (20 mL × 3) . The organic layers were washed with water (20 mL × 2) and brine (20 mL) , dried with Na2SO4, filtered, the filtrate was concentrated by vacuum, The residue was purified by flash silica gel column chromatography (0 %-100 %EA in PE) to afford the desired product. LCMS: MS (ESI) m/z 638.3 [M+H] +.
Step D: 1- (5- (4- ( (4- (4- (4-aminophenyl) piperazin-1-yl) piperidin-1-yl) methyl) piperidine-1-
carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (4- ( (4- (4- (4-nitrophenyl) piperazin-1-yl) piperidin-1-yl) methyl) piperidine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (190 mg, 0.298 mmol) , Fe (100 mg, 1.79 mmol) , NH4Cl (95 mg, 1.78 mmol) in EtOH/H2O (10 mL/2mL) . Then the mixture was stirred at 90℃ for 3 h. The resulting mixture was filtered, the filter cake was washed with DCM (20 ml x3) . The filtrate was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 608.3 [M+H] +.
Step E: 1- (5- { [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] carbonyl} -2-
chlorophenyl) -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a solution of 1- (5- (4- ( (4- (4- (4-aminophenyl) piperazin-1-yl) piperidin-1-yl) methyl) piperidine-1-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (60 mg, 0.099 mmol) in DCM(10 mL) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20 mg, 0.052 mmol) , the mixture was stirred at 25℃ for 16 h under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3
× 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 18-25%Retention Time: 9-19 of 12 min) to afford the desired product.
LCMS: MS (ESI) m/z 929.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.51 (s, 1H) , 10.04 (s, 1H) , 8.82 (s, 1H) , 8.08 -7.96 (m, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.64 (d, J = 8.2 Hz, 1H) , 7.61 -7.52 (m, 4H) , 7.42 –7.35 (m, 1H) , 6.90 (d, J = 8.8 Hz, 2H) , 5.42 -5.29 (m, 2H) , 5.27 (s, 1H) , 4.51 -4.40 (m, 2H) , 3.81 –3.71 (m, 1H) , 3.67 -3.50 (m, 2H) , 3.30 –3.28 (m, 4H) , 3.18 -3.00 (m, 4H) , 2.80 -2.71 (m, 3H) , 2.64 –2.58 (m, 3H) , 2.21 –2.13 (m, 2H) , 2.04 -1.90 (m, 3H) , 1.87 -1.70 (m, 6H) , 1.56 (m, 3H) , 1.24 (m, 3H) , 1.15 –1.05 (m, 3H) , 0.92 –0.73 (m, 2H) .
Example 158:
1- {5- [ (9- { [4- (4- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} -2-tolyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-361)
Scheme:
Step A: 1- {5- [ (9- { [4- (4- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} -2-
tolyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-chlorophenyl} -1, 3, 5, 6-
tetrahydro-2, 4-pyrimidinedione
To a solution of 1- (5- (9- ( (4- (4-amino-2-methylphenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (100.0 mg,0.165 mmol) in DCM (10 mL) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (30.0 mg, 0.078 mmol) , the mixture was stirred at 25℃ for 16 hrs under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by prep-HPLC (Waters 2767, Column: Pursuit XRs C18, 21.2*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 15%~25%Retention Time: 9.0-10.0 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 928.7 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.51 (s, 1H) , 10.09 (brs, 1H) , 8.84 (s, 1H) , 8.25 (s, 1H) , 8.01 (t, J = 7.9 Hz, 1H) , 7.79 (d, J = 7.9 Hz, 1H) , 7.64 (d, J = 8.1 Hz, 1H) , 7.60 (d, J = 7.6 Hz, 1H) , 7.56 -7.53 (m, 1H) , 7.43 -7.36 (m, 2H) , 6.98 (d, J = 8.5 Hz, 1H) , 5.42 –5.21 (m, 3H) , 4.50 –4.41 (m, 2H) , 3.80 –3.72 (m, 3H) , 3.65 -3.56 (m, 5H) , 2.84 -2.78 (m, 4H) , 2.76 -2.73 (m, 2H) , 2.22 (s, 3H) , 2.19 –2.15 (m, 3H) , 2.06 -2.00 (m, 1H) , 1.98 –1.90 (m, 2H) , 1.76 –1.67 (m,
4H) , 1.62 –1.58 (m, 2H) , 1.56 (s, 3H) , 1.53 -1.46 (m, 3H) , 1.37 –1.28 (m, 2H) , 1.14 –1.03 (m, 4H) .
Example 159:
3- {5- [ (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-chlorophenyl} -2, 6-piperidinedione (Cpd-362)
Scheme:
Step A: 3- [2-chloro-5- [9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-
azaspiro [5.5] undecane-3-carbonyl] phenyl] piperidine-2, 6-dione
To a solution of 4-chloro-3- (2, 6-dioxo-3-piperidyl) benzoic acid (300 mg, 1.12 mmol) , 9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane (400 mg, 1.07 mmol) in DCM (4 mL) and DMF (1 mL) was added DIPEA (434.57 mg, 3.36 mmol) and HATU (639.25 mg, 1.68 mmol) . The mixture was stirred at 20 ℃ for 1 h. The reaction mixture was added H2O (30 mL) and extracted with ethyl acetate (30 mL × 3) . The combined organic phases were dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol = 100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 622.4 [M+H] +.
Step B: 3- [5- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-
carbonyl] -2-chlorophenyl] piperidine-2, 6-dione
To a solution of 3- [2-chloro-5- [9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] phenyl] piperidine-2, 6-dione (530 mg, 851.88 μmol) in DCM (5 mL) and DMAC (1 mL) was added PdCl2 (151.06 mg, 851.88 μmol) , TEA (172.40 mg, 1.70
mmol) and Et3SiH (1.98 g, 17.04 mmol) . The mixture was stirred at 20 ℃ for 16 h. The reaction mixture was added aq. sat. NaHCO3 (30 mL) and extracted with ethyl acetate (50 mL ×3) . The combined organic phases were dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol = 100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 592.4 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.92 (s, 1H) , 7.52 (d, J = 8.0 Hz, 1H) , 7.39 -7.29 (m, 2H) , 6.67 (d, J = 8.8 Hz, 2H) , 6.49 (d, J = 8.8 Hz, 2H) , 4.26 (dd, J = 4.8, 12.4 Hz, 1H) , 3.58 (m, 2H) , 3.29 (m, 2H) , 2.91 (m, 4H) , 2.80-2.72 (m, 1H) , 2.69 -2.63 (m, 1H) , 2.57-2.55 (m, 1H) , 2.35-2.30 (m, 1H) , 1.75-1.65 (m, 2H) , 1.61-1.47 (m, 5H) , 1.45 -1.30 (m, 3H) , 1.30-1.21 (m, 5H) , 1.12-0.95 (m, 5H) .
Step C: 3- {5- [ (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-
chlorophenyl} -2, 6-piperidinedione
To a solution of 3- [5- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] -2-chlorophenyl] piperidine-2, 6-dione (80 mg, 135.10 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (52.07 mg, 135.10 μmol) in n-BuOH (3 mL) was added AcOH (81.13 mg, 1.35 mmol) . The mixture was stirred at 100 ℃ for 1 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: Welch Xtimate C18 150*25mm*5um; mobile phase: [water (TFA) -ACN] ; gradient: 30%-60%B over 7 min) to afford the desired product.
LCMS: MS (ESI) m/z 913.6 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.92 (s, 1H) , 10.07 (TFA, 0.86H) , 9.37 (s, 1H) , 8.84 (s, 1H) , 8.06 -7.94 (m, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.64-7.56 (m, 3H) , 7.53 (d, J = 8.0 Hz, 1H) , 7.36 (s, 1H) , 7.33 (d, J = 8.0 Hz, 1H) , 6.98 (d, J = 8.8 Hz, 2H) , 5.40-5.25 (m, 2H) , 4.47 (d, J = 6.0 Hz, 2H) , 4.27 (dd, J = 4.8, 12.4 Hz, 1H) , 3.76-3.63 (m, 4H) , 3.28 (m, 2H) , 3.20-3.11 (m, 2H) , 3.09-2.96 (m, 4H) , 2.84 -2.72 (m, 1H) , 2.62-2.55 (m, 1H) , 2.53 -2.51 (m, 1H) , 2.40 -2.27 (m, 1H) , 2.19 (s, 1H) , 2.08 -1.90 (m, 3H) , 1.85-1.78 (m, 1H) , 1.76-1.68 (m, 3H) , 1.65-1.58 (m, 2H) , 1.56 (s, 3H) , 1.55 -1.24 (m, 5H) , 1.21-1.09 (m, 4H) .
Example 160:
3- {5- [ (9- { [4- (p- { (R, 12E) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-chlorophenyl} -2, 6-piperidinedione (Cpd-363)
Scheme:
Step A: 3- {5- [ (9- { [4- (p- { (R, 12E) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-
chlorophenyl} -2, 6-piperidinedione
To a solution of 3- [5- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] -2-chlorophenyl] piperidine-2, 6-dione (80 mg, 135.10 μmol) , (12E, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (54.50 mg, 135.10 μmol) in n-BuOH (3 mL) was added AcOH (81.13 mg, 1.35 mmol) . The mixture was stirred at 100 ℃ for 16 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: Welch Xtimate C18 150*25mm*5um; mobile phase: [water (TFA) -ACN] ; gradient: 30%-60%B over 7 min) to afford the desired product.
LCMS: MS (ESI) m/z 931.6 [M+H] +.
19F NMR (400MHz, DMSO-d6) δ = -74.187
1H NMR (400MHz, DMSO-d6) δ 10.93 (s, 1H) , 10.16 (TFA, 0.78H) , 9.27 (s, 1H) , 8.86 (s, 1H) , 8.05 -7.98 (m, 1H) , 7.81 (d, J = 8.0 Hz, 1H) , 7.64-7.56 (m, 3H) , 7.53 (d, J = 8.0 Hz, 1H) , 7.36 (s, 1H) , 7.33 (d, J = 8.0 Hz, 1H) , 6.99 (d, J = 9.2 Hz, 2H) , 5.17 -4.95 (m, 1H) , 4.73 -4.57 (m, 2H) , 4.27 (dd, J = 4.8, 12.4 Hz, 1H) , 3.76-3.63 (m, 4H) , 3.28 (s, 2H) , 3.20-3.11 (m, 2H) , 3.10 -2.97 (m, 4H) , 2.86 -2.72 (m, 1H) , 2.62-2.55 (m, 1H) , 2.53 -2.51 (m, 1H) , 2.40-2.27 (m, 1H) , 2.20-2.09 (m, 1H) , 2.04 -1.96 (m, 1H) , 1.95-1.86 (m, 2H) , 1.85 -1.77 (m, 1H) , 1.75-1.70 (m, 2H) , 1.68-1.60 (m, 3H) , 1.57 (m, 3H) , 1.55 -1.25 (m, 5H) , 1.17-1.09 (m, 4H) .
Example 161:
3- {5- [ (9- { [4- (p- { (R, 12E) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-methoxyphenyl} -2, 6-piperidinedione (Cpd-154)
Scheme:
Step A: methyl 2- (5-bromo-2-methoxy-phenyl) acetate
To a solution of 2- (5-bromo-2-methoxy-phenyl) acetic acid (3 g, 12.24 mmol) in MeOH (30 mL) was added HCl/dioxane (4 M, 10 mL) . The mixture was stirred at 80 ℃ for 3 hr. The mixture was concentrated to afford the desired product.
1H NMR (400 MHz, DMSO-d6) δ 7.48 -7.38 (m, 2H) , 6.96 (d, J = 8.4 Hz, 1H) , 3.75 (s, 3H) , 3.61 (s, 2H) , 3.59 (s, 3H) .
Step B: 3- (5-bromo-2-methoxy-phenyl) piperidine-2, 6-dione
To a solution of methyl 2- (5-bromo-2-methoxy-phenyl) acetate (3 g, 11.58 mmol) in DMF (30 mL) was added t-BuOK (1.56 g, 13.89 mmol) and prop-2-enamide (987.59 mg, 13.89 mmol) at
0 ℃. The mixture was stirred at 20 ℃ for 2 hr. The mixture was poured into aq. NH4Cl (100 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers were washed with brine (50 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue. The crude product was triturated with EtOAc (10 mL) at 25 ℃ for 1 hr, the mixture was filtered and the solid was concentrated to afford the desired product.
1H NMR (400 MHz, DMSO-d6) δ 10.77 (s, 1H) , 7.43 (dd, J = 2.8, 8.8 Hz, 1H) , 7.35 (d, J = 2.4 Hz, 1H) , 6.99 (d, J = 8.8 Hz, 1H) , 3.95 (dd, J = 5.2, 12.4 Hz, 1H) , 3.74 (s, 3H) , 2.76 -2.61 (m, 1H) , 2.49 -4.43 (m, 1H) , 2.30 -2.15 (m, 1H) , 1.95 -1.82 (m, 1H) .
Step C: (4-methoxyphenyl) methyl 3- (2, 6-dioxo-3-piperidyl) -4-methoxy-benzoate
To a mixture of 3- (5-bromo-2-methoxy-phenyl) piperidine-2, 6-dione (3 g, 10.06 mmol) and (4-methoxyphenyl) methanol (6.95 g, 50.31 mmol) in DMF (40 mL) was added Et3N (6.21 g, 61.38 mmol, 8.54 mL) and Pd (dppf) Cl2 (2.21 g, 3.02 mmol) . The mixture was stirred at 80 ℃ for 72 hours under CO (50 Psi) . The reaction mixture was added H2O (100 mL) and extracted with ethyl acetate (100 mL × 2) . The combined organic phases were washed with brine (100 mL ×2) , dried with anhydrous Na2SO4, filtered and concentrated to give a residue. The residue was purified by silica gel column chromatography (53%EtOAc in PE) to afford the desired product.
1H NMR (400 MHz, DMSO-d6) δ 10.77 (s, 1H) , 7.91 (dd, J = 2.0, 8.8 Hz, 1H) , 7.77 (d, J = 2.0 Hz, 1H) , 7.39 (d, J = 8.8 Hz, 2H) , 7.13 (d, J = 8.8 Hz, 1H) , 6.95 (d, J = 8.8 Hz, 2H) , 5.24 (s, 2H) , 4.03 -3.99 (m, 1H) , 3.82 (m, 3H) , 3.76 (m, 3H) , 2.69 -2.63 (m, 1H) , 2.47 -2.43 (m, 1H) , 2.30 -2.18 (m, 1H) , 1.94 -1.86 (m, 1H) .
Step D: 3- (2, 6-dioxo-3-piperidyl) -4-methoxy-benzoic acid
To a solution of (4-methoxyphenyl) methyl 3- (2, 6-dioxo-3-piperidyl) -4-methoxy-benzoate (3.8 g, 9.91 mmol) in TFA (20 mL) . The mixture was stirred at 20 ℃ for 16 hr. The reaction mixture was concentrated to give crude product. The crude product was triturated with EtOAc (10 mL) at 25 ℃ for 1 hr, the mixture was filtered and the solid was concentrated to afford the desired product.
1H NMR (400 MHz, DMSO-d6) δ 10.78 (s, 1H) , 7.88 (dd, J = 2.0, 8.8 Hz, 1H) , 7.74 (d, J = 2.0 Hz, 1H) , 7.11 (d, J = 8.8 Hz, 1H) , 4.02 (dd, J = 5.2, 12.0 Hz, 1H) , 3.82 (s, 3H) , 2.72 -2.63 (m, 1H) , 2.49 -2.43 (m, 1H) , 2.30 -2.17 (m, 1H) , 1.97 -1.87 (m, 1H) .
Step E: 3- [2-methoxy-5- [9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-
azaspiro [5.5] undecane-3-carbonyl] phenyl] piperidine-2, 6-dione
To a solution of 9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane (400 mg, 1.07 mmol) , 3- (2, 6-dioxo-3-piperidyl) -4-methoxy-benzoic acid (339.21 mg, 1.29 mmol) , DIEA (416.34 mg, 3.22 mmol) in DCM (10 mL) was added HATU (612.45 mg, 1.61 mmol) . The mixture was stirred at 20 ℃ for 2hr. The mixture was poured into water (50 mL) and extracted
with DCM (100 mL × 2) . The combined organic layers were washed with brine (50 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by silica gel column chromatography (5%MeOH in Dichloromethane) to afford the desired product.
LCMS: MS (ESI) m/z 618.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.76 (s, 1H) , 8.05 (d, J = 9.2 Hz, 2H) , 7.32 (dd, J = 2.0, 8.4 Hz, 1H) , 7.19 (d, J = 1.6 Hz, 1H) , 7.03 (dd, J = 9.2, 12.8 Hz, 3H) , 4.01 -3.96 (m, 1H) , 3.79 (m, 3H) , 3.43 (m, 6H) , 2.76 -2.62 (m, 1H) , 2.45 (m, 4H) , 2.30 -2.11 (m, 3H) , 1.96 -1.87 (m, 1H) , 1.77 -1.66 (m, 2H) , 1.58 (d, J = 10.8 Hz, 2H) , 1.52 -1.41 (m, 3H) , 1.35 -1.21 (m, 5H) , 1.13-0.95 (m, 4H) .
Step F: 3- [5- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-
carbonyl] -2-methoxy-phenyl] piperidine-2, 6-dione
To a solution of 3- [2-methoxy-5- [9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] phenyl] piperidine-2, 6-dione (580 mg, 938.92 μmol) in DCM (5 mL) and DMAC (1 mL) was added PdCl2 (166.50 mg, 938.92 μmol) , Et3N (190.02 mg, 1.88 mmol, 261.37 μL) and Et3SiH (2.18 g, 18.78 mmol) . The mixture was stirred at 25 ℃ for 4 hr. The reaction mixture was poured into water (50 mL) and adjusted to pH = 8 with aq. Na2CO3 (50 mL) . Then extracted with petroleum ether (100 mL × 2) . The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated to give a residue. The residue was purified by silica gel column chromatography (13%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 588.5 [M+H] +.
Step G: 3- {5- [ (9- { [4- (p- { (R, 12E) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-
methoxyphenyl} -2, 6-piperidinedione
A mixture of (12E, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (50 mg, 123.94 μmol) , 3- [5- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] -2-methoxy-phenyl] piperidine-2, 6-dione (72.84 mg, 123.94 μmol) , AcOH (74.42 mg, 1.24 mmol, 70.95 μL) in n-BuOH (3 mL) was stirred at 100 ℃ for 2 hr. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 15%-45%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 927.8 [M+H] +.
19F NMR (400 MHz, DMSO-d6) δ = -74.436
1H NMR (400 MHz, DMSO-d6) δ 10.79 (s, 1H) , 10.19 (TFA, 1H) , 9.26 (s, 1H) , 8.86 (s, 1H) , 8.08 -7.97 (m, 1H) , 7.81 (d, J = 8.0 Hz, 1H) , 7.61 (dd, J = 4.4, 8.0 Hz, 3H) , 7.32 (dd, J = 2.0, 8.4 Hz, 1H) , 7.20 (d, J = 2.0 Hz, 1H) , 7.05 (d, J = 8.8 Hz, 1H) , 6.99 (d, J = 9.2 Hz, 2H) , 5.17 -4.96 (m, 1H) , 4.74 -4.53 (m, 2H) , 3.99 (dd, J = 4.8, 12.0 Hz, 1H) , 3.79 (m, 3H) , 3.76 -3.71 (m, 6H) , 3.60 -3.64 (m, 3H) , 3.20 -2.98 (m, 6H) , 2.77 -2.61 (m, 1H) , 2.49 -2.43 (m, 1H) , 2.31 -2.06 (m, 2H) , 1.98 -1.86 (m, 3H) , 1.83 -1.70 (m, 3H) , 1.69 -1.59 (m, 3H) , 1.57 (m, 3H) , 1.47 (m, 2H) , 1.32 (m, 2H) , 1.25 -1.08 (m, 4H) .
Example 162:
1- (5- { [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] carbonyl} -2-chlorophenyl) -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-364)
Scheme:
Step A: tert-butyl 4- ( (4- (4- (4-nitrophenyl) piperazin-1-yl) piperidin-1-yl) methyl) piperidine-
1-carboxylate
To a solution of 1- (4-nitrophenyl) -4- (piperidin-4-yl) piperazine (220 mg, 0.759 mmol) , tert-butyl 4-formylpiperidine-1-carboxylate (162 mg, 0.759 mmol) in DCM (15 mL) , was added NaBH (OAc) 3 (880 mg, 4.15 mmol) . The resulting mixture was stirred for 16 h at 25℃ in an oil bath. The residue was purified by flash silica gel column chromatography (DCM: MeOH =20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 488.5 [M+H] +.
Step B: 1- (4-nitrophenyl) -4- (1- (piperidin-4-ylmethyl) piperidin-4-yl) piperazine
hydrochloride
To a solution of tert-butyl 4- ( (4- (4- (4-nitrophenyl) piperazin-1-yl) piperidin-1-yl) methyl) piperidine-1-carboxylate (280 mg, 0.575 mmol) , HCl in dioxane (1M 5 ml) \in DCM (15 mL) . Then the mixture was stirred at 35℃ for 2 h. The resulting mixture was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 388.2 [M+H] +.
Step C: 1- (2-chloro-5- (4- ( (4- (4- (4-nitrophenyl) piperazin-1-yl) piperidin-1-
yl) methyl) piperidine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (4-nitrophenyl) -4- (1- (piperidin-4-ylmethyl) piperidin-4-yl) piperazine hydrochloride (230 mg, 0.594 mmol) in DMF (10 mL) was added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (260 mg, 0.598 mmol) and DIPEA (230 mg, 1.78 mmol) . Then the reaction mixture was stirred at 25℃ for 1 h under N2 atmosphere. After completion of reaction, diluted with water (20 mL) , and the solution was extracted with EtOAc (20 mL × 3) . The organic layers were washed with water (20 mL × 2) and brine (20 mL) , dried with Na2SO4, filtered, the filtrate was concentrated by vacuum, The residue was purified by flash silica gel column chromatography (0 %-100 %EA in PE) to afford the desired product.
LCMS: MS (ESI) m/z 638.3 [M+H] +.
Step D: 1- (5- (4- ( (4- (4- (4-aminophenyl) piperazin-1-yl) piperidin-1-yl) methyl) piperidine-1-
carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (4- ( (4- (4- (4-nitrophenyl) piperazin-1-yl) piperidin-1-yl) methyl) piperidine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (190 mg, 0.298 mmol) , Fe (100 mg, 1.79 mmol) , NH4Cl (95 mg, 1.78 mmol) in EtOH/H2O (10 mL/2mL) . Then the mixture was stirred at 90℃ for 3 h. The resulting mixture was filtered, the filter cake was washed with DCM (20 mL x3) . The filtrate was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 608.3 [M+H] +.
Step E: 1- (5- { [4- ( {4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -1-piperidyl} methyl) -1-piperidyl] carbonyl} -2-
chlorophenyl) -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a solution of 1- (5- (4- ( (4- (4- (4-aminophenyl) piperazin-1-yl) piperidin-1-yl) methyl) piperidine-1-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (60 mg, 0.099 mmol) in DCM(10 mL) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (20 mg, 0.052 mmol) , the mixture was stirred at 25℃ for 16 h under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3
× 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 18-25%Retention Time: 9-19 of 12 min) to afford the desired product.
LCMS: MS (ESI) m/z 929.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.51 (s, 1H) , 10.04 (s, 1H) , 8.82 (s, 1H) , 8.08 -7.96 (m, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.64 (d, J = 8.2 Hz, 1H) , 7.61 -7.52 (m, 4H) , 7.42 –7.35 (m, 1H) , 6.90 (d, J = 8.8 Hz, 2H) , 5.42 -5.29 (m, 2H) , 5.27 (s, 1H) , 4.51 -4.40 (m, 2H) , 3.81 –3.71 (m, 1H) , 3.67 -3.50 (m, 2H) , 3.30 –3.28 (m, 4H) , 3.18 -3.00 (m, 4H) , 2.80 -2.71 (m, 3H) , 2.64 –2.58 (m, 3H) , 2.21 –2.13 (m, 2H) , 2.04 -1.90 (m, 3H) , 1.87 -1.70 (m, 6H) , 1.56 (m, 3H) , 1.24 (m, 3H) , 1.15 –1.05 (m, 3H) , 0.92 –0.73 (m, 2H) .
Example 163:
3- (6- {9- [4- (p- { (R, 12E) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-365)
Scheme:
Step A: 3- (6- {9- [4- (p- { (R, 12E) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
To a solution of 3- (6- (9- (4- (4-aminophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecan-3-yl) -1-oxoisoindolin-2-yl) piperidine-2, 6-dione (339.52 mg, 594.90 μmol) and (12E, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (200 mg, 495.75 μmol) in n-BuOH (5 mL) was added AcOH (297.71 mg, 4.96 mmol) and stirred at 100 ℃ for 2hr. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 6%-36%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 910.8 [M+H] +.
19F NMR (400 MHz, DMSO-d6) δ =-74.463
1H NMR (400 MHz, DMSO-d6) δ11.00 (s, 1H) , 10.29 -10.12 (m, 1H) , 9.48 (br s, 1H) , 8.87 (s, 1H) , 8.07 -7.96 (m, 1H) , 7.81 (d, J = 8.0 Hz, 1H) , 7.62 (br t, J = 7.6 Hz, 3H) , 7.47 (d, J = 8.4 Hz, 1H) , 7.34 (br d, J = 8.0 Hz, 1H) , 7.30 (br s, 1H) , 7.01 (br d, J = 8.8 Hz, 2H) , 5.13 -4.99 (m, 2H) , 4.65 (d, J = 8.0 Hz, 2H) , 4.35 (d, J = 16.8 Hz, 1H) , 4.22 (d, J = 16.8 Hz, 1H) , 3.82 (br d, J = 12.0 Hz, 3H) , 3.31 -3.17 (m, 8H) , 2.99 -2.86 (m, 3H) , 2.60 (br d, J = 16.4 Hz, 1H) , 2.38 (dq,
J = 4.4, 13.6 Hz, 1H) , 2.17 -2.07 (m, 1H) , 2.04 -1.82 (m, 8H) , 1.71 -1.60 (m, 5H) , 1.57 (s, 3H) , 1.50 (br s, 2H) , 1.19 (br t, J = 12.0 Hz, 2H)
Example 164:
1- {5- [ (4- { [9- (p- { (R, 12E) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) carbonyl] -2-methoxyphenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-161)
Scheme:
Step A: 1- (2-methoxy-5- (4- ( (9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-
yl) methyl) piperidine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
A solution of 3- (4-nitrophenyl) -9- (piperidin-4-ylmethyl) -3, 9-diazaspiro [5.5] undecane (225 mg, 0.60 mmol) , perfluorophenyl 3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) -4-methoxybenzoate (335.6 mg, 0.78 mmol) and DIEA (232 mg, 1.8 mmol) in DMF (10 mL) was stirred at room temperature for 2hrs. The solution was added water and extracted with ethyl acetate (20 mL x 3) . The organic layer was dried and concentrated. The residue was purified by silica gel chromatography (eluted with petroleum ether: ethyl acetate =2: 1 to 1: 1) to afford the desired product.
LCMS: MS (ESI) m/z 619.3 [M+H] +.
Step B: 1- (5- (4- ( (9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-
carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a stirred solution of 1- (2-methoxy-5- (4- ( (9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (298 mg, 0.48 mmol) in EtOH (10 mL) and water (2 mL) was added iron powder (134.4 mg, 2.4 mmol) followed ammonium chloride (128.4 mg, 2.4 mmol) at room temperature and heated to 90℃ for 30 min. The reaction mixture was diluted with DCM (20 mL) . The organic layer was washed with water (2 x 10 mL) , brine (20 mL) , dried by anhydrous Na2SO4 and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 589.4 [M+H] +.
Step C: 1- {5- [ (4- { [9- (p- { (R, 12E) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) carbonyl] -2-
methoxyphenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a solution of (12R, E) -9-fluoro-12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (30.0 mg, 0.074 mmol) in DCM (2 mL) was added 1- (5- (4- ( (9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidine-1-carbonyl) -2-methoxyphenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (100 mg, 0.17 mmol) , the mixture was stirred at 25℃ for 16 hrs under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC to afford the desired product.
LCMS: MS (ESI) m/z 928.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.34 (s, 1H) , 10.13 (s, 1H) , 8.84 (s, 2H) , 8.01 (s, 1H) , 7.81 (d, J = 8.1 Hz, 1H) , 7.58 (dd, J = 15.9, 8.3 Hz, 3H) , 7.42 –7.29 (m, 2H) , 7.17 (d, J = 8.6 Hz, 1H) , 6.93 (d, J = 9.0 Hz, 2H) , 5.32 (s, 1H) , 5.05 (d, J = 16.5 Hz, 1H) , 4.64 (d, J = 20.1 Hz, 2H) , 3.85 (s, 3H) , 3.60 (dd, J = 7.8, 5.0 Hz, 2H) , 3.17 –2.98 (m, 9H) , 2.69 (dd, J = 11.7, 5.1 Hz, 3H) , 2.12 (dt, J = 20.8, 10.3 Hz, 2H) , 2.01 –1.68 (m, 9H) , 1.68 –1.42 (m, 9H) , 1.20 (dd, J = 20.5, 9.1 Hz, 2H) .
Example 165:
1- {5- [ (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] methyl} -9-methyl-3-aza-3-spiro [5.5] undecyl) carbonyl] -2-chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-110)
Scheme:
Step A: tert-butyl 9-formyl-9-methyl-3-azaspiro [5.5] undecane-3-carboxylate
A solution of tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylatee (1000 mg, 4.27 mmol) , t-BuOK (575.4 mg 5.13 mmol) and CH3I (1818.2 mg 12.81 mmol) in DCM (15 mL)
was stirred at 35℃ for 3 h . After completion of reaction, quenched with water (50 mL) , extracted with EtOAc (3*50 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =2: 1) to afford the desired product.
1H NMR (400 MHz, CDCl3) δ 9.43 (s, 1H) , 3.55 (dd, J = 11.5, 5.5 Hz, 4H) , 1.87-1.76 (m, 2H) , 1.50 (dd, J = 13.4, 5.9 Hz, 2H) , 1.45 (m, 10H) , 1.39-1.20 (m, 7H) , 1.01 (s, 3H) .
Step B: tert-butyl 9-methyl-9- ( (4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carboxylate
A solution of 4-methyl-1- (4-nitrophenyl) piperidine-4-carbaldehyde (300 mg, 1.2 mmol) , tert-butyl 3, 9-diazaspiro [5.5] undecane-3-carboxylate (369.2 mg1.45 mmol) and NaBH (OAc) 3 (769.1 mg 3.63 mmol) in DCM (5 mL) and MeOH (5 ml) and HOAc (60 mg 0.709 mmol) was stirred at 35℃ for 2 h. After completion of reaction, quenched with water (50 mL) , extracted with EtOAc (3*50 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =1: 1) to afford the desired product.
LCMS: MS (ESI) m/z 487.2 [M+H] +.
Step C: 9-methyl-9- ( (4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane
To a solution of tert-butyl 9-methyl-9- ( (4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate (90 mg, 0.1800 mmol) in DCM (5.0 mL) was added HCl in dioxane (4.0 mL) and stirred at 35℃ for 1 hrs. The mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 387.2 [M+H] +.
Step D: 1- (2-chloro-5- (9-methyl-9- ( (4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-
azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 9-methyl-9- ( (4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane (80 mg, 0.2 mmol) in DMF (5.0 mL) was added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (135.2 mg, 0.31 mmol) , and DIPEA (77.5 mg, 0.6 mmol) . Then the reaction was stirred at 35℃ for 12 h under N2 protection. After completion of reaction, quenched with water (30 mL) , extracted with EA (3*50 mL) . The organic layers were washed with brine (20 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 637.3 [M+H] +.
Step E: 1- (5- (9- ( (4- (4-aminophenyl) piperazin-1-yl) methyl) -9-methyl-3-
azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 1- (2-chloro-5- (9-methyl-9- ( (4- (4-nitrophenyl) piperazin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (150 mg 0.23 mmol) in EtOH (10 mL) and H2O (3 mL) was added NH4Cl (62.01 mg, 1.17 mmol ) and Fe (65.9 mg, 1.17 mmol) . The mixture was stirred at 90℃ under N2 for 2 hrs. The reaction solution was filtered under diatomite, and then the filtrate is suspended dry to afford the desired product.
LCMS: MS (ESI) m/z 607.3 [M+H] +.
Step F: 1- {5- [ (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] methyl} -9-methyl-3-aza-3-spiro [5.5] undecyl) carbonyl] -2-
chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a solution of (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (40.0 mg, 0.104 mmol) in DCM (5 mL) was added 1- (5- (9- ( (4- (4-aminophenyl) piperazin-1-yl) methyl) -9-methyl-3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (120 mg, 0.197 mmol) . Then the reaction mixture was stirred at 35℃ for 12hrs. The mixture was quenched by the addition of 50 mL water, the resulting solution was extracted with DCM (2*50 mL) , the organic layer was combined and concentrated to get the crude. The residue was purified by Pre-HPLC (Column: Agilent C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 25-25%; Retention Time: 8.1-9.3min of 16min) to afford the desired product.
LCMS: MS (ESI) m/z 928.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.51 (s, 1H) , 10.02 (s, 1H) , 8.82 (s, 1H) , 8.33 (s, 1H) , 8.02 (s, 1H) , 7.76 (d, J = 8.4 Hz, 1H) , 7.64 (d, J = 8.1 Hz, 1H) , 7.61 –7.51 (m, 4H) , 7.39 (d, J = 7.7 Hz, 1H) , 6.90 (d, J = 8.5 Hz, 2H) , 5.42 –5.13 (m, 3H) , 4.52 –4.40 (m, 2H) , 3.80 –3.72 (m, 2H) , 3.59 (dd, J = 11.0, 8.1 Hz, 4H) , 2.93 –2.88 (m, 2H) , 2.77 –2.72 (m, 2H) , 2.45 (dd, J = 10.0, 6.9 Hz, 4H) , 2.18 (dd, J = 18.4, 13.9 Hz, 3H) , 2.03 –1.93 (m, 2H) , 1.74 –1.69 (m, 1H) , 1.65 –1.56 (m, 5H) , 1.51 –1.43 (m, 6H) , 1.38 –1.30 (m, 3H) , 1.27 –1.21 (m, 3H) , 0.91 (s, 3H) .
Example 166:
1- {5- [ (9- { [4- (p- { (R, 12E) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-163)
Scheme:
Step A: (12E, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one
To a solution of (12E, 16R) -16-hydroxy-16-methyl-5-methylsulfanyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (140 mg, 378.95 μmol) in THF (3 mL) and H2O (3 mL) was added Oxone (873.63 mg, 1.42 mmol) at 0-20 ℃. The mixture was stirred at 0~20 ℃ for 2 h. The reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (30 mL × 3) . The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 386.1 [M+H] +.
Step B: 1- {5- [ (9- { [4- (p- { (R, 12E) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-
chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a solution of (12E, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (38.41 mg, 99.64 μmol) , 1- [5- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] -2-chloro-phenyl] hexahydropyrimidine-2, 4-dione (59.10 mg, 99.64 μmol) in t-BuOH (3 mL) was added TFA (80.00 mg, 701.61 μmol) . The mixture was stirred at 100 ℃ for 2 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: Welch Xtimate C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 15%-55%B over 9 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 914.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.52 (s, 1H) , 10.07 (TFA, 1H) , 8.79 (s, 1H) , 8.20 (s, 1H) , 8.07 (s, 1H) , 7.69 -7.54 (m, 6H) , 7.39 (d, J = 6.8 Hz, 1H) , 6.90 (d, J = 8.8 Hz, 2H) , 5.58 -5.30 (m, 3H) , 4.12 (s, 1H) , 3.75 (d, J = 7.2 Hz, 1H) , 3.61 (m, 5H) , 3.07 (m, 5H) , 2.92 (s, 2H) , 2.74 (d, J = 4.4 Hz, 2H) , 2.46 (m, 4H) , 2.16 (m, 2H) , 1.71 (d, J = 11.2 Hz, 2H) , 1.57 (d, J = 12.0 Hz, 2H) , 1.54 -1.42 (m, 4H) , 1.38 (m, 4H) , 1.28 (s, 1H) , 1.10 (m, 5H) .
Example 167:
1- {5- [ (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperidyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-151)
Scheme:
Step A: 1- {5- [ (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperidyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] -2-
chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a solution of 1- (5- (9- ( (4- (4-aminophenyl) piperidin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (153 mg, 0.25 mmol) in DCM (3 mL) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (100 mg, 0.25mmol) . The mixture was stirred at 35℃ for 1 8 hrs. lcms showed TM. the mixture was diluted with H2O (10 mL) and extracted with EtOAc (20 mL x 2) . Washed with brine (20 mL) , The combined organic layers were concentrated and The residue was purified by HPLC (Waters 2767/Qda Column: SunFire Sunfire C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate gradient: 17-27%Retention Time: 9-10.4 of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 913.9 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.51 (s, 1H) , 10.15 (s, 1H) , 8.87 (s, 1H) , 8.03 (t, J = 7.7 Hz, 1H) , 7.77 (d, J = 8.2 Hz, 1H) , 7.69 –7.50 (m, 5H) , 7.42 –7.35 (m, 1H) , 7.18 (d, J = 8.5 Hz, 2H) , 5.43 –5.21 (m, 3H) , 4.49 (m, 2H) , 3.80 –3.71 (m, 1H) , 3.68 –3.52 (m, 3H) , 3.32 (m, 4H) , 2.96 –2.85 (m, 2H) , 2.74 (dd, J = 9.3, 5.2 Hz, 2H) , 2.69 –2.65 (m, 1H) , 2.41 (dd, J = 13.9, 6.7 Hz, 1H) , 2.15 (dd, J = 25.6, 9.0 Hz, 2H) , 1.95 (dt, J = 14.7, 9.8 Hz, 3H) , 1.68 (m, 8H) , 1.56 (m, 3H) , 1.48 (dd, J = 15.1, 7.8 Hz, 3H) , 1.31 (ddd, J = 25.6, 17.5, 7.3 Hz, 2H) , 1.06 (dt, J = 27.3, 16.0 Hz, 4H) .
Example 168:
3- [5- (4- { [9- (p- { (R, 12E) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-171)
Scheme:
Step A: 3- [5- [4- [ [3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] methyl] -1-piperidyl] -
1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 1- [2- (2, 6-dioxo-3-piperidyl) -1-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde (200 mg, 562.77 μmol) , 3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane (154.96 mg, 562.77 μmol) in DCM (2 mL) was added NaBH (OAc) 3 (238.55 mg, 1.13 mmol) and Et3N (170.84 mg, 1.69 mmol) . The mixture was stirred at 15 ℃ for 2hr. The mixture was added H2O (10 mL) , extracted with EtOAc (30 mL × 3) . The combined organic layers were washed with brine (5 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue to afford the desired product.
LCMS: MS (ESI) m/z 615.4 [M+H] +.
Step B: 3- [5- [4- [ [3- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] methyl] -1-piperidyl] -
1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 3- [5- [4- [ [3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (350 mg, 569.35 μmol) in EtOH (5 mL) , THF (5 mL) and H2O (2.5 mL) was added Fe (317.96 mg, 5.69 mmol) and NH4Cl (456.83 mg, 8.54 mmol) . The mixture was stirred at 60 ℃ for 1hr. The mixture was concentrated to give a residue. The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 0~15 %Methanol/Dichloromethane gradient @30 mL/min) and the combined desired fractions was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 585.3 [M+H] +.
Step C: 3- [5- (4- { [9- (p- { (R, 12E) -12-fluoro-16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -
2, 6-piperidinedione
A mixture of 3- [5- [4- [ [3- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (70 mg, 119.71 μmol) and (12E, 16R) -12-fluoro-16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 119.21 μmol) in n-BuOH (3 mL) was stirred at 100 ℃ for 2hr. The mixture was concentrated to give a residue. The crude product was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (ammonia hydroxide v/v) -ACN] ; gradient: 0%-37%B over 9 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 924.7 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.94 (s, 1H) , 10.10 (br s, 1H) , 8.83 (s, 1H) , 8.22 (s, 1H) , 8.03 (br s, 1H) , 7.82 (d, J = 8.0 Hz, 1H) , 7.54 (dt, J = 8.4, 19.6 Hz, 4H) , 7.12 -6.98 (m, 2H) , 6.90 (br d, J = 9.2 Hz, 2H) , 5.31 (br s, 1H) , 5.04 (br dd, J = 5.2, 13.2 Hz, 2H) , 4.76 -4.52 (m, 2H) , 4.36 -4.13 (m, 2H) , 3.86 (br d, J = 12.0 Hz, 2H) , 3.08 (br s, 4H) , 2.95 -2.76 (m, 3H) , 2.63 -2.51 (m, 2H) , 2.34 (m, 4H) , 2.20 -2.07 (m, 3H) , 1.98 -1.84 (m, 3H) , 1.80 -1.70 (m, 3H) , 1.65 (br s, 1H) , 1.60 -1.43 (m, 11H) , 1.20 -1.10 (m, 2H) .
Example 169:
1- {2-chloro-5- [ (9- { [4- (4- {11- (2, 6-dichlorophenyl) -10-oxo-5, 7, 11, 13-tetraazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-4-yl} -3-methyl-1-pyrazolyl) -1-
piperidyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] phenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-273)
Scheme:
Step A: tert-butyl 4- (4-bromo-3-methyl-1H-pyrazol-1-yl) piperidine-1-carboxylate
A solution of 4-bromo-3-methyl-1H-pyrazole (2.00 g, 12.4 mmol) in DMA (40 mL) was treated sequentially with NaH (60 wt. %mineral oil dispersion; 992 mg, 24.8 mmol) and tert-butyl 4-bromopiperidine-1-carboxylate (4.91 g, 18.6 mmol) . The reaction was stirred at 70℃ for 16 hrs under N2 atmosphere. After cooling to ambient temperature, the reaction mixture was quenched with water (20 mL) , extracted with EtOAc (50 mL x 3) . The combined organic layers were
washed with brine (10 mL) , dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (eluted with PE: EtOAc=5: 1) to afford the desired product.
LCMS: MS (ESI) m/z 346.1 [M+H] +.
Step B: tert-butyl 4- (3-methyl-4- (4, 4, 5-trimethyl-1, 3, 2-dioxaborolan-2-yl) -1H-pyrazol-1-
yl) piperidine-1-carboxylate
A mixture of tert-butyl 4- (4-bromo-3-methyl-1H-pyrazol-1-yl) piperidine-1-carboxylate (823 mg, 2.39 mmol) , 4, 4, 4', 4', 5, 5, 5', 5'-octamethyl-2, 2'-bi (1, 3, 2-dioxaborolane) (909 mg, 3.58 mmol) , KOAc (704 mg, 7.17 mmol) and Pd (dppf) Cl2 (145 mg, 0.239 mmol) in 1, 4-dioxane (5 mL) was stirred at 100℃ under N2 atmosphere for 16 hrs. Water (40 mL) was added and the solution was extracted with EtOAc (100 mL x 3) . The combined organic layers were washed with brine (40 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography eluted with PE: EtOAc=12: 1 to afford the desired product.
LCMS: MS (ESI) m/z 391.8 [M+H] +.
Step C: tert-butyl 4- (4- (3- (2, 6-dichlorophenyl) -4-oxo-7- ( (2- (trimethylsilyl) ethoxy) methyl) -
4, 7-dihydro-3H-pyrrolo [3', 2': 5, 6] pyrido [4, 3-d] pyrimidin-8-yl) -3-methyl-1H-pyrazol-1-
yl) piperidine-1-carboxylate
A mixture of tert-butyl 4- (3-methyl-4- (4, 4, 5-trimethyl-1, 3, 2-dioxaborolan-2-yl) -1H-pyrazol-1-yl) piperidine-1-carboxylate (127 mg, 0.325 mmol) , 8-bromo-3- (2, 6-dichlorophenyl) -7- ( (2- (trimethylsilyl) ethoxy) methyl) -3H-pyrrolo [3', 2': 5, 6] pyrido [4, 3-d] pyrimidin-4 (7H) -one (70.2 mg, 0.130 mmol) , K2CO3 (53.9 mg, 0.390 mmol) and Pd (dppf) Cl2 (14.3 mg, 0.0195 mmol) in 1, 4-dioxane (5 mL) and H2O (1 mL) was stirred at 100℃ under N2 atmosphere for 16 hrs. Water (40 mL) was added and the solution was extracted with EtOAc (100 mL x 3) . The combined organic layers were washed with brine (40 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography eluted with PE: EtOAc=0: 100 to afford the desired product.
LCMS: MS (ESI) m/z 724.2 [M+H] +.
Step D: 3- (2, 6-dichlorophenyl) -8- (3-methyl-1- (piperidin-4-yl) -1H-pyrazol-4-yl) -7- ( (2-
(trimethylsilyl) ethoxy) methyl) -3H-pyrrolo [3', 2': 5, 6] pyrido [4, 3-d] pyrimidin-4 (7H) -one
To a solution of tert-butyl 4- (4- (3- (2, 6-dichlorophenyl) -4-oxo-7- ( (2- (trimethylsilyl) ethoxy) methyl) -4, 7-dihydro-3H-pyrrolo [3', 2': 5, 6] pyrido [4, 3-d] pyrimidin-8-yl) -3-methyl-1H-pyrazol-1-yl) piperidine-1-carboxylate (42.0 mg, 0.0580 mmol) in DCM (2 mL) was added HCl in dioxane (2 mL, 4 M) and stirred at 20℃ for 1 hr. After completion of reaction, the
reaction mixture was concentrated under reduced pressure to dryness to afford the desired product.
LCMS: MS (ESI) m/z 624.2 [M+H] +.
Step E: tert-butyl 9- ( (4- (4- (3- (2, 6-dichlorophenyl) -4-oxo-4, 7-dihydro-3H-
pyrrolo [3', 2': 5, 6] pyrido [4, 3-d] pyrimidin-8-yl) -3-methyl-1H-pyrazol-1-yl) piperidin-1-
yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate
A solution of 3- (2, 6-dichlorophenyl) -8- (3-methyl-1- (piperidin-4-yl) -1H-pyrazol-4-yl) -7- ( (2- (trimethylsilyl) ethoxy) methyl) -3H-pyrrolo [3', 2': 5, 6] pyrido [4, 3-d] pyrimidin-4 (7H) -one (30.0 mg, 0.0480 mmol) , tert-butyl 9-formyl-3, 9-diazaspiro [5.5] undecane-3-carboxylate (16.3 mg, 0.0576 mmol) , AcOH (0.2 mL) in DCM (5 mL) and MeOH (5 mL) was stirred at 25℃ for 0.5 h. Then NaBH (OAc) 3 (30.5 mg, 0.144 mmol) was added. was added. The mixture was stirred at 25℃ for 16 hrs. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with DCM (3 x 30 mL) . The organic layers were washed with brine (30 mL) , dried over Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (DCM: MeOH =20: 1) to afford the desired product.
LCMS: MS (ESI) m/z 889.3 [M+H] +.
Step F: 8- (1- (1- (3-azaspiro [5.5] undecan-9-ylmethyl) piperidin-4-yl) -3-methyl-1H-pyrazol-4-
yl) -3- (2, 6-dichlorophenyl) -3H-pyrrolo [3', 2': 5, 6] pyrido [4, 3-d] pyrimidin-4 (7H) -one
To a solution of tert-butyl 9- ( (4- (4- (3- (2, 6-dichlorophenyl) -4-oxo-4, 7-dihydro-3H-pyrrolo [3', 2': 5, 6] pyrido [4, 3-d] pyrimidin-8-yl) -3-methyl-1H-pyrazol-1-yl) piperidin-1-yl) methyl) -3-azaspiro [5.5] undecane-3-carboxylate (40.0 mg, 0.0526) in DCM (3 mL) was added TFA (3 mL) , then the reaction mixture was stirred at 25℃ for 2 hrs under N2 atmosphere, the reaction mixture was concentrated. The residue was dissolved in ACN (10 mL) , NH3. H2O (2mL) was added. The mixture was stirred at 70℃ for 2 hrs. After completion of reaction, the reaction mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 659.3 [M+H] +.
Step G: 1- {2-chloro-5- [ (9- { [4- (4- {11- (2, 6-dichlorophenyl) -10-oxo-5, 7, 11, 13-
tetraazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7, 12-pentaen-4-yl} -3-methyl-1-pyrazolyl) -1-
piperidyl] methyl} -3-aza-3-spiro [5.5] undecyl) carbonyl] phenyl} -1, 3, 5, 6-tetrahydro-2, 4-
pyrimidinedione
To a solution of 8- (1- (1- (3-azaspiro [5.5] undecan-9-ylmethyl) piperidin-4-yl) -3-methyl-1H-pyrazol-4-yl) -3- (2, 6-dichlorophenyl) -3H-pyrrolo [3', 2': 5, 6] pyrido [4, 3-d] pyrimidin-4 (7H) -one (25.0 mg, 0.0379 mmol) in DMF (4 mL) was added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (33.0 mg, 0.0758 mmol) , DIEA (14.7 mg, 0.114 mmol) . Then the reaction mixture was stirred at 25℃ for 2 hrs under N2 atmosphere. After
completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EtOAc (3 x 20 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated. The residue was purified by prep-HPLC (Waters 2767/Qda, Column: Pursuit XRs C18, 21.2*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 28~38%Retention Time: 8.0-8.8 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 911.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 12.55 (d, J = 40.2 Hz, 1H) , 10.51 (s, 1H) , 8.98 (d, J = 1.8 Hz, 1H) , 8.62 (d, J = 1.7 Hz, 1H) , 8.29 (s, 1H) , 7.98 (s, 1H) , 7.81 (d, J = 8.1 Hz, 2H) , 7.66 (dd, J =17.0, 8.1 Hz, 2H) , 7.56 (s, 1H) , 7.39 (d, J = 7.3 Hz, 1H) , 6.91 (s, 1H) , 4.17 (dt, J = 26.7, 14.9 Hz, 1H) , 3.76 (d, J = 8.0 Hz, 1H) , 3.61 (dd, J = 11.9, 4.7 Hz, 3H) , 2.95 –2.88 (m, 2H) , 2.78 –2.71 (m, 2H) , 2.45 (m, 3H) , 2.20 –2.14 (m, 2H) , 2.05 (t, J = 11.7 Hz, 4H) , 1.95 –1.82 (m, 2H) , 1.81 –1.61 (m, 3H) , 1.61 –1.54 (m, 2H) , 1.53 –1.41 (m, 3H) , 1.39 –1.25 (m, 2H) , 1.22 –0.89 (m, 5H) .
Example 170:
1- {5- [ (9- { [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-methyl-4-piperidyl] methyl} -3, 9-diaza-3-spiro [5.5] undecyl) carbonyl] -2-chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-366)
Scheme:
Step A: 4-methyl-1- (4-nitrophenyl) piperidine-4-carbaldehyde
A solution of 1- (4-nitrophenyl) piperidine-4-carbaldehyde (1000 mg, 4.27 mmol) , t-BuOK (575.4 mg 5.13 mmol) and CH3I (1818.2 mg 12.81 mmol) in DCM (15 mL) was stirred at 35 ℃for 3 h . After completion of reaction, quenched with water (50 mL) , extracted with EtOAc (3*50 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and
concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =2: 1) to afford the desired product.
LCMS: MS (ESI) m/z 249.1 [M+H] +.
Step B: tert-butyl 9- ( (4-methyl-1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-
diazaspiro [5.5] undecane-3-carboxylate
A solution of 4-methyl-1- (4-nitrophenyl) piperidine-4-carbaldehyde (300 mg, 1.2 mmol) , tert-butyl 3, 9-diazaspiro [5.5] undecane-3-carboxylate (369.2 mg1.45 mmol) and NaBH (OAc) 3 (769.1 mg 3.63 mmol) in DCM (5 mL) and MeOH (5 ml) and HOAc (60 mg 0.709 mmol) was stirred at 35 ℃ for 2 h . After completion of reaction, quenched with water (50 mL) , extracted with EtOAc (3*50 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =1: 1) to afford the desired product.
LCMS: MS (ESI) m/z 487.2 [M+H] +.
Step C: 3- ( (4-methyl-1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane
To a solution of tert-butyl 9- ( (4-methyl-1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carboxylate (90 mg, 0.1800 mmol) in DCM (5.0 mL) was added HCl in dioxane (4.0 mL) and stirred at 35℃ for 1 hrs. The mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 387.3 [M+H] +.
Step D: 1- (2-chloro-5- (9- ( (4-methyl-1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-
diazaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 3- ( (4-methyl-1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane (80 mg, 0.2 mmol) in DMF (5.0 mL) was added perfluorophenyl 4-chloro-3- (2, 4-dioxotetrahydropyrimidin-1 (2H) -yl) benzoate (135.2 mg, 0.31 mmol) , and DIPEA (77.5 mg, 0.6 mmol) . Then the reaction was stirred at 35℃ for 12 h under N2 protection. After completion of reaction, quenched with water (30 mL) , extracted with EA (3*50 mL) . The organic layers were washed with brine (20 mL) , dried with Na2SO4, filtered. The filtrate was concentrated and purified by flash silica gel column chromatography (DCM: MeOH =10: 1) to afford the desired product.
LCMS: MS (ESI) m/z 637.3 [M+H] +.
Step E: 1- (5- (9- ( (1- (4-aminophenyl) -4-methylpiperidin-4-yl) methyl) -3, 9-
diazaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione
To a solution of 2 1- (2-chloro-5- (9- ( (4-methyl-1- (4-nitrophenyl) piperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carbonyl) phenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (150 mg 0.23 mmol) in EtOH (10 mL) and H2O (3 mL) was added NH4Cl (62.01 mg, 1.17 mmol ) and Fe
(65.9 mg, 1.17 mmol) was stirred at 90℃ under N2 for 2 hrs. The reaction solution is filtered under diatomite, and then the filtrate is suspended dry to afford the desired product.
LCMS: MS (ESI) m/z 387.3 [M+H] +.
Step F: 1- {5- [ (9- { [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-methyl-4-piperidyl] methyl} -3, 9-diaza-3-spiro [5.5] undecyl) carbonyl] -2-
chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a solution of (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (40.0 mg, 0.104 mmol) in DCM (5 mL) was added 1- (5- (9- ( (1- (4-aminophenyl) -4-methylpiperidin-4-yl) methyl) -3, 9-diazaspiro [5.5] undecane-3-carbonyl) -2-chlorophenyl) dihydropyrimidine-2, 4 (1H, 3H) -dione (120 mg, 0.197 mmol) . Then the reaction mixture was stirred at 35℃ for 12hrs . The mixture was quenched by the addition of 50 mL water, the resulting solution was extracted with DCM (2*100 mL) , the organic layer was combined and concentrated to get the crude. The residue was purified by Pre-HPLC (Column: Agilent C18, 19*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20ml/min; gradient: 25-25%; Retention Time: 8.1-9.3min of 16min) to afford the desired product.
LCMS: MS (ESI) m/z 928.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.51 (s, 1H) , 10.02 (s, 1H) , 8.82 (s, 1H) , 8.33 (s, 1H) , 8.02 (s, 1H) , 7.76 (d, J = 8.4 Hz, 1H) , 7.64 (d, J = 8.1 Hz, 1H) , 7.61 –7.51 (m, 4H) , 7.39 (d, J = 7.7 Hz, 1H) , 6.90 (d, J = 8.5 Hz, 2H) , 5.42 –5.13 (m, 3H) , 4.52 –4.40 (m, 2H) , 3.80 –3.72 (m, 2H) , 3.59 (dd, J = 11.0, 8.1 Hz, 4H) , 2.93 –2.88 (m, 2H) , 2.77 –2.72 (m, 2H) , 2.45 (dd, J = 10.0, 6.9 Hz, 4H) , 2.18 (dd, J = 18.4, 13.9 Hz, 3H) , 2.03 –1.93 (m, 2H) , 1.74 –1.69 (m, 1H) , 1.65 –1.56 (m, 5H) , 1.51 –1.43 (m, 6H) , 1.38 –1.30 (m, 3H) , 1.27 –1.21 (m, 3H) , 0.91 (m, 3H) .
Example 171:
3- [5- (2- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] methyl} -8-aza-8-spiro [4.5] decyl) -1, 3-dioxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-093)
Scheme:
Step A: 3- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decane
To a solution of tert-butyl 3- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decane-8-carboxylate (500 mg, 1.09 mmol) in HCl/dioxane (4 M, 10.91 mL) . The mixture was stirred at 20 ℃ for 2 hr under N2. The reaction mixture was poured into water (50 mL) and adjusted to pH = 8~9 with aq. Na2CO3 (50 mL) , extracted with petroleum ether (100 mL × 2) . The combined organic layer was washed with brine (100 mL) , dried over anhydrous Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 359.2 [M+H] +.
Step B: 2- (2, 6-dioxo-3-piperidyl) -5- [3- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -8-
azaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione
To a solution of 3- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decane (400 mg, 1.12 mmol) , 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (308.21 mg, 1.12 mmol) , DIEA (288.43 mg, 2.23 mmol, 388.72 μL) in NMP (5 mL) . The mixture was stirred at 100 ℃for 2 hr under N2. The mixture was poured into water (50 mL) and extracted with ethyl acetate (100 mL × 2) . The combined organic layers were washed with brine (100 mL × 3) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 615.3 [M+H] +.
Step C: 5- [3- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -2- (2, 6-
dioxo-3-piperidyl) isoindoline-1, 3-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [3- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione (800 mg, 1.30 mmol) , Fe (726.81 mg, 13.01 mmol) and NH4Cl (2 M, 6 mL) in EtOH (9 mL) . The mixture was stirred at 80 ℃ for 16 hr. The mixture was filtered and concentrated to give a residue, which was purified by silica gel column chromatography (8%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 585.3 [M+H] +.
Step D: 3- [5- (2- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] methyl} -8-aza-8-spiro [4.5] decyl) -1, 3-dioxo-2-isoindolinyl] -
2, 6-piperidinedione
A mixture of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (30 mg, 74.73 μmol) , TFA (40.00 mg, 350.81 μmol) and 5- [3- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -8-azaspiro [4.5] decan-8-yl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (43.70 mg, 74.73 μmol) in n-BuOH (3 mL) was stirred at 100 ℃ for 2 hr. The mixture was concentrated to give a residue, which was purified by prep-HPLC (column: Welch Xtimate C18 100*40mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 17%-47%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 906.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ11.08 (s, 1H) , 10.10 (TFA, 0.91H) , 9.32 (s, 1H) , 8.84 (s, 1H) , 8.05 -7.93 (m, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.69 -7.57 (m, 4H) , 7.33 (s, 1H) , 7.25 (d, J = 8.4 Hz, 1H) , 6.99 (d, J = 9.2 Hz, 2 H) , 5.42 -5.22 (m, 2H) , 5.06 (dd, J = 12.8, 5.6 Hz, 1H) , 4.47 (d, J = 5.2 Hz, 2H) , 3.76 (d, J = 12.4 Hz, 2H) , 3.60 (d, J = 11.2 Hz, 2H) , 3.26 -3.09 (m, 6H) , 3.05 -2.96 (m, 2H) , 2.94 -2.83 (m, 1H) , 2.64 –2.52 (m, 2H) , 2.48 -2.41 (m, 2H) , 2.26 -2.13 (m, 1H) , 2.11 -1.80 (m, 6H) , 1.77 -1.67 (m, 1H) , 1.66 -1.46 (m, 10H) , 1.46 -1.35 (m, 1H) , 1.22 -1.12 (m, 1H) .
Example 172:
3- (5- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -1, 3-dioxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-094)
Scheme:
Step A: 5- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -2- (2, 6-dioxo-3-
piperidyl) isoindoline-1, 3-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] isoindoline-1, 3-dione (1 g, 1.17 mmol) in EtOH (20 mL) was added Fe (650.81 mg, 11.65 mmol) and NH4Cl (2 M, 5 mL) . The mixture was stirred at 80 ℃ for 16 h. The reaction mixture was filtered, washed with EtOAc (5 mL) , concentrated under reduced pressure to give crude product. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol=100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 571.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ11.08 (s, 1H) , 7.66 (d, J = 8.4 Hz, 1H) , 7.34 (s, 1H) , 7.25 (dd, J = 2.0, 8.4 Hz, 1H) , 6.74 (d, J = 8.4 Hz, 2H) , 6.54 (d, J = 8.4 Hz, 2H) , 5.06 (dd, J = 5.6, 12.8 Hz, 1H) , 3.61 -3.36 (m, 8H) , 3.17 (d, J = 4.8 Hz, 4H) , 2.95 -2.82 (m, 1H) , 2.61 (s, 1H) , 2.57-2.53 (m, 1H) , 2.17 -1.95 (m, 4H) , 1.92 (s, 1H) , 1.74 -1.62 (m, 3H) , 1.59 -1.45 (m, 4H) .
Step B: 3- (5- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -1, 3-dioxo-2-isoindolinyl) -2, 6-
piperidinedione
To a mixture of 5- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (51.17 mg, 89.66 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (30 mg, 74.73 μmol) in n-BuOH (2 mL) was heated at 100 ℃ for 3 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 6%-46%B over 9 min) to afford the desired product.
LCMS: MS (ESI) m/z 892.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.08 (s, 1H) , 10.06 (s, 1H) , 8.82 (s, 1H) , 8.14 (s, 0.2 FA) , 8.05 -7.93 (m, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.65 (d, J = 8.8 Hz, 1H) , 7.57 (t, J = 7.6 Hz, 3H) , 7.32 (s, 1H) , 7.20 -7.26 (m, 1H) , 6.90 (d, J = 9.2 Hz, 2H) , 5.42-5.23 (m, 3H) , 5.06 (dd, J = 12.8, 5.2 Hz, 1H) , 4.45 (d, J = 6.0 Hz, 2H) , 3.47 (s, 4H) , 3.09 (s, 4H) , 2.93-2.83 (m, 1H) , 2.61 -2.59 (m, 1H) , 2.57 (s, 4H) , 2.54 -2.52 (m, 1H) , 2.25 -2.15 (m, 1H) , 2.08 -1.94 (m, 3H) , 1.82 -1.94 (m, 2H) , 1.75 -1.67 (m, 1H) , 1.65 -1.45 (m, 11H) , 1.36 -1.30 (m, 1H) .
Example 173:
3- [5- (2- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] methyl} -7-aza-7-spiro [3.5] nonyl) -1, 3-dioxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-095)
Scheme:
Step A: 5- [2- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -7-azaspiro [3.5] nonan-7-yl] -2- (2, 6-
dioxo-3-piperidyl) isoindoline-1, 3-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [2- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -7-azaspiro [3.5] nonan-7-yl] isoindoline-1, 3-dione (300 mg, 499.45 μmol) , NH4Cl (2 M, 2 mL) in EtOH (3 mL) was added Fe (278.92 mg, 4.99 mmol) . The mixture was stirred at 80 ℃ for 16 hr under N2. The mixture was filtered and concentrated to give a residue, which was purified by silica gel column chromatography (8%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 571.3 [M+H] +.
Step B: 3- [5- (2- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] methyl} -7-aza-7-spiro [3.5] nonyl) -1, 3-dioxo-2-isoindolinyl] -
2, 6-piperidinedione
A mixture of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (30 mg, 74.73 μmol) , 5- [2- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -7-azaspiro [3.5] nonan-7-yl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (51.18 mg, 89.68 μmol) in n-BuOH (3 mL) was stirred at 100 ℃ for 2 hr. LCMS (ES22540-317-P1A) showed 51%desired mass was detected. The mixture was concentrated to give a residue, which was purified by prep-HPLC (column: Welch Xtimate C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 10%-50%B over 9 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 892.7 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.87 (s, 1H) , 8.30 (s, 1H) , 8.17 (FA, 0.1H) , 7.88 -7.94 (m, 1H) , 7.45 -7.80 (m, 1H) , 7.68 (d, J = 8.4 Hz, 1H) , 7.53 (d, J = 8.4 Hz, 2H) , 7.29 -7.32 (m, 1H) , 7.05 (d, J = 8.4 Hz, 1H) , 6.92 (d, J = 8.4 Hz, 2H) , 5.60 -5.90 (m, 1H) , 4.95 (dd, J = 12.0, 4.8 Hz, 1H) , 4.45 -4.60 (m, 1 H) , 4.20 -4.35 (m, 1 H) , 3.57 (m, 4H) , 3.31 -3.45 (m, 5H) , 3.15 -3.25 (m, 2H) , 3.08 (m, 3H) , 2.68 -2.97 (m, 4H) , 2.24 (s, 3H) , 2.15 (d, J = 9.6 Hz, 3H) , 1.94 -2.06 (m, 2H) , 1.92 -2.07 (m, 1H) , 1.77 -1.83 (m, 2H) , 1.63 -1.66 (m, 2H) , 1.26 (m, 4H) , 0.76 -0.93 (m, 2H) .
Example 174:
3- (7- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -1-oxo-2-phthalazinyl) -2, 6-piperidinedione (Cpd-143)
Scheme:
Step A: 7- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) phthalazin-1 (2H) -
one
A mixture of 7-bromophthalazin-1 (2H) -one (131 mg, 0.581 mmol) , 2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decane (200 mg, 0.581 mmol) , Pd-PEPPSI-IPentCl (56.6 mg, 0.0581 mmol) , Cs2CO3 (566 mg, 1.74 mmol) in dioxane (10 mL) was stirred at 100℃for 2 hrs under N2. After completion, the mixture was quenched with H2O (10 mL) , extracted with DCM (10 mL x 3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LC-MS (ESI) [M+H] + = 489.4
Step B: 3- (7- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-
oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione
A mixture of 7- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) phthalazin-1 (2H) -one (200 mg, 0.409 mmol) in DMF (8.0 mL) was treated with NaH (60wt. %, 60.8 mg, 1.52 mmol) at 0℃ for 1 h. Then 3-bromopiperidine-2, 6-dione (781 mg, 4.09 mmol) was added. The reaction was stirred at 25℃ under N2 atmosphere for 3 hrs. After completion, the mixture was diluted with H2O (5 mL) , extracted with EtOAc (10 mLx3) . The combined organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (eluted with DCM: MeOH=20: 1) to afford the desired product.
LC-MS (ESI) [M+H] + = 600.2
Step C: 3- (7- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-
oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione
A mixture of 3- (7- (2- (4- (4-nitrophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan-8-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (180 mg, 0.300 mmol) , iron powder (84 mg, 1.5 mmol) , NH4Cl (79.5 mg, 1.5 mmol) in EtOH (10 mL) and H2O (2 mL) was stirred at 90℃ for 3 hrs. After completion of reaction, the mixture reaction was filtered through a Celite pad, and the filtrate was concentrated to give the crude. The crude was quenched with H2O (10 mL) , adjusted to pH 8 with solid NaHCO3 and extracted with EtOAc (3 x 30 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LC-MS (ESI) [M+H] + = 570.1
Step D: 3- (7- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -1-oxo-2-phthalazinyl) -2, 6-
piperidinedione
To a solution of 3- (7- (2- (4- (4-aminophenyl) piperazin-1-yl) -8-azaspiro [4.5] decan -8-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (100.0 mg, 0.175 mmol) in DCM (10 mL) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (33.8 mg, 0.088 mmol) . Then the reaction mixture was stirred at 25℃ for 16 hrs under N2 atmosphere. After completion of reaction, the mixture reaction was quenched with H2O (10 mL) and extracted with EA (20 mL x3) . The organic layers were concentrated. The residue was purified by prep-HPLC (Waters 2767, Column: Pursuit XRs C18, 21.2*250mm, 10 um; Mobile Phase A: 0.1%FA/H2O, B: ACN ; flow rate: 20mL/min; gradient: 15%~25%Retention Time: 8.0-111.0 min of 16 min) to afford the desired product.
LCMS: MS (ESI) m/z 891.8 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H) , 8.82 (s, 1H) , 8.23 (s, 1H) , 8.05 –7.92 (m, 2H) , 7.75 (d, J = 8.2 Hz, 1H) , 7.57 (dt, J = 11.9, 6.1 Hz, 3H) , 7.50 –7.44 (m, 1H) , 7.23 (d, J = 2.3 Hz, 1H) , 6.90 (d, J = 6.3 Hz, 2H) , 5.84 –5.66 (m, 1H) , 5.36 –5.22 (m, 3H) , 4.56 –4.32 (m, 2H) , 3.44 (m, 4H) , 3.08 (dd, J = 9.8, 6.7 Hz, 4H) , 2.92 –2.87 (m, 1H) , 2.68 –2.64 (m, 3H) , 2.31 (s, 1H) , 2.08 (dd, J = 8.3, 5.7 Hz, 2H) , 1.99 (d, J = 1.4 Hz, 2H) , 1.90 (d, J = 2.2 Hz, 2H) , 1.77 –1.69 (m, 2H) , 1.64 –1.60 (m, 2H) , 1.56 (d, J = 1.7 Hz, 3H) , 1.51 (d, J = 5.4 Hz, 2H) , 1.26 (m, 2H) , 1.24 (d, J = 1.5 Hz, 3H) , 0.88 –0.80 (m, 2H) .
Example 175:
3- [6- (4- { [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) -4-fluoro-1-oxo-1, 2-dihydro-2-phthalazinyl] -2, 6-piperidinedione (Cpd-150)
Scheme:
Step A: 6- (4- (dimethoxymethyl) piperidin-1-yl) -4-fluorophthalazin-1 (2H) -one
To a solution of 6-bromo-4-fluorophthalazin-1 (2H) -one (200 mg, 0.829 mmol) in Dioxane (10 ml) was added 4- (dimethoxymethyl) piperidine (131 mg, 0.829 mmol) , Ruphos Pd-G2 (64 mg, 0.0829 mmol) , t-BuONa (238 mg, 2.487 mmol) then the reaction mixture was stirred at 100 ℃for 16 h under N2 atmosphere. After completion of reaction, quenched with H2O (10 mL) , extracted with EA (3*20 mL) . The organic layers were washed with brine (10 mL) , dried with
Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =0: 1) to afford the desired product.
LCMS: MS (ESI) m/z 322.2 [M+H] +.
Step B: 3- (6- (4- (dimethoxymethyl) piperidin-1-yl) -4-fluoro-1-oxophthalazin-2 (1H) -
yl) piperidine-2, 6-dione
To a solution of 6- (4- (dimethoxymethyl) piperidin-1-yl) -4-fluorophthalazin-1 (2H) -one (110 mg, 0.342 mmol) in DMF (10 ml) was added NaH (26.4 mg, 0.660 mmol) , 3-bromopiperidine-2, 6-dione (649 mg, 3.42 mmol) , then the reaction mixture was stirred at 0 -25℃ for 16 h under N2 atmosphere . the mixture was quenched with H2O (10 mL) , extracted with EA (3 × 10 mL) . The organic layers were washed with brine (10 mL) , dried with Na2SO4, filtered and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =1: 2) to afford the desired product.
LCMS: MS (ESI) m/z 433.2 [M+H] +.
Step C: 1- (2- (2, 6-dioxopiperidin-3-yl) -4-fluoro-1-oxo-1, 2-dihydrophthalazin-6-
yl) piperidine-4-carbaldehyde
To a solution of 3- (6- (4- (dimethoxymethyl) piperidin-1-yl) -4-fluoro-1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (100 mg, 0.231 mmol) in DCM/FA (1 ml/1 ml ) was stirred at 50℃ for 3 hrs under N2 atmosphere. After completion of reaction the reaction mixture was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 387.3 [M+H] +.
Step D: 3- (4-fluoro-6- (4- ( (9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-
yl) methyl) piperidin-1-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione
To a solution of 1- (2- (2, 6-dioxopiperidin-3-yl) -4-fluoro-1-oxo-1, 2-dihydrophthalazin-6-yl) piperidine-4-carbaldehyde (110 mg, 0.284 mmol) , 3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane (78.1 mg, 0.284 mmol) in DCM (2 mL) and MeOH (2 mL) was stirred at 25 ℃ for 0.5 h. Then NaBH (OAc) 3 (59.9mg, 0.284 mmol) was added. The mixture was stirred at 25 ℃ for 16 hrs. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by flash silica gel column chromatography (PE: EA =1: 2) to afford the desired product.
LCMS: MS (ESI) m/z 646.3 [M+H] +.
Step E: 3- (6- (4- ( (9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidin-1-
yl) -4-fluoro-1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione
A mixture of 3- (4-fluoro-6- (4- ( (9- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidin-1-yl) -1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (100 mg, 0.155 mmol) ,
iron powder (43.4mg, 0.775 mmol) , NH4Cl (41.1 mg, 0.775 mmol) in EtOH (10 mL) and H2O (2 mL) was stirred at 90℃ for 2 hrs. After completion of reaction, the mixture reaction was filtered through a Celite pad, and the filtrate was concentrated to give the crude. The crude was quenched with H2O (10 mL) , adjusted to pH 8 with solid NaHCO3 and extracted with EtOAc (3 x 30 mL) . The organic layers were washed with brine (10 mL) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 616.3 [M+H] +.
Step F: 3- [6- (4- { [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) -4-fluoro-1-oxo-1, 2-
dihydro-2-phthalazinyl] -2, 6-piperidinedione
To a solution of 3- (6- (4- ( (9- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-3-yl) methyl) piperidin-1-yl) -4-fluoro-1-oxophthalazin-2 (1H) -yl) piperidine-2, 6-dione (50.0 mg, 0.081 mmol) in DCM (8 mL) was added (12R, Z) -12-hydroxy-12-methyl-2- (methylsulfinyl) -7, 10, 11, 12-tetrahydro-5H-13, 17- (azeno) pyrimido [4', 5': 3, 4] pyrazolo [1, 2-a] [1, 2] diazacyclotridecin-5-one (16.0 mg, 0.041 mmol) , the mixture was stirred at 25℃ for 16 hrs under N2 atmosphere. The mixture was quenched by the addition of 10 mL water, the resulting solution was extracted with DCM (3 × 10 mL) , the organic layers were combined and concentrated. The residue was purified by HPLC to afford the desired product.
LC-MS (ESI) [M+H] + 937.9
1H NMR (400 MHz, DMSO-d6) δ 11.04 (s, 1H) , 10.11 –9.95 (m, 1H) , 8.81 (s, 1H) , 8.26 (s, 1H) , 8.02 (s, 1H) , 7.78 –7.74 (m, 2H) , 7.65 –7.51 (m, 5H) , 6.90 (d, J = 9.0 Hz, 2H) , 5.74 (dt, J = 10.5, 4.8 Hz, 1H) , 5.34 (ddd, J = 24.8, 23.7, 18.1 Hz, 3H) , 4.45 (ddd, J = 8.1, 5.9, 3.1 Hz, 2H) , 4.05 (dt, J = 10.5, 4.5 Hz, 2H) , 3.08 (s, 4H) , 2.99 (ddd, J = 14.3, 8.1, 5.6 Hz, 3H) , 2.87 (s, 1H) , 2.70 –2.65 (m, 2H) , 2.64 –2.60 (m, 2H) , 2.35 –2.32 (m, 4H) , 2.16 (d, J = 5.8 Hz, 3H) , 2.09 –1.92 (m, 3H) , 1.86 –1.79 (m, 3H) , 1.75 –1.69 (m, 1H) , 1.56 (s, 3H) , 1.54 (d, J = 2.3 Hz, 2H) , 1.47 (dd, J = 5.5, 2.2 Hz, 3H) , 1.19 –1.10 (m, 2H) .
Example 176:
4- {1- [ (3S, 4S) -1- ( {3- [4-chloro-3- (2, 4-dioxo-1, 3, 5, 6-tetrahydro-1-pyrimidinyl) benzoyl] -3-aza-9-spiro [5.5] undecyl} methyl) -3-fluoro-4-piperidyl] -4-pyrazolyl} -11- (2, 6-dichlorophenyl) -13-methyl-5, 7, 11, 13-tetraazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (Cpd-299)
Scheme:
Step A: methyl 4-chloro-1- (2-trimethylsilylethoxymethyl) pyrrolo [2, 3-b] pyridine-5-
carboxylate
To a mixture of methyl 4-chloro-1H-pyrrolo [2, 3-b] pyridine-5-carboxylate (11 g, 52.23 mmol) in THF (110 mL) was added NaH (3.13 g, 78.34 mmol, 60%purity) at 0 ℃ under N2. The mixture was stirred at 0 ℃ for 30 min. The mixture was added SEM-Cl (11.32 g, 67.90 mmol, 12.02 mL) at 0 ℃ under N2, then heated to 15 ℃ and stirred for 1.5 hours. The reaction mixture was quenched by addition aq. NH4Cl (50 mL) at 0 ℃ and then diluted with H2O (50 mL) and extracted with EtOAc (100 mL × 3) . The combined organic layers were washed with brine (50 mL × 3) , dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=100/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 341.1 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.87 (s, 1H) , 7.43 (d, J = 3.6 Hz, 1H) , 6.76 (d, J = 3.6 Hz, 1H) , 5.69 (s, 2H) , 3.99 (s, 3H) , 3.59 -3.50 (m, 2H) , 0.95 -0.87 (m, 2H) , 0.09 -0.03 (m, 9H) .
Step B: methyl 2-bromo-4-chloro-1- (2-trimethylsilylethoxymethyl) pyrrolo [2, 3-b] pyridine-
5-carboxylate
To a solution of 2, 2, 6, 6-tetramethylpiperidine (4.97 g, 35.20 mmol, 5.98 mL) in THF (12.59 mL) was added n-BuLi (2.5 M, 14.12 mL) dropwise at -70 ℃. The solution was stirred at 0 ℃for 15 min. To a solution of methyl 4-chloro-1- (2-trimethylsilylethoxymethyl) pyrrolo [2, 3-
b]pyridine-5-carboxylate (6 g, 17.60 mmol) in THF (50 mL) was added LTMP (1 M, 17.60 mL, 1.2 eq) at -70 ℃ dropwise. The mixture was stirred at -70 ℃ for 15 min. The mixture was added 1,2-dibromo-1, 1, 2, 2-tetrachloro-ethane (6.31 g, 19.36 mmol, 2.32 mL) and THF (20 mL) at -70 ℃ dropwise. The mixture was stirred at -70 ℃ for 1 h. The reaction mixture was quenched with saturated NH4Cl solution (150 mL) at 0 ℃, and stirred 10 min. The mixture was allowed to warm to 25 ℃ naturally, and stirred over 10 min at 20~25 ℃. The mixture was extracted with EtOAc (100 mL × 3) twice. The organic layer was concentrated to give crude. The crude was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=100/1 to 5/1) to afford the desired product.
LCMS: MS (ESI) m/z 420.9 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 8.75 (s, 1H) , 7.04 (s, 1H) , 5.68 (s, 2H) , 3.90 (s, 3H) , 3.55 (t, J = 8.0 Hz, 2H) , 0.82 (t, J = 8.0 Hz, 2H) , -0.12 (s, 9H) .
2D NMR showed C11 has HMBC correlation to H23, which is consistent with the determined structure.
Step C: 2-bromo-4-chloro-N- (2, 6-dichlorophenyl) -1- (2-
trimethylsilylethoxymethyl) pyrrolo [2, 3-b] pyridine-5-carboxamide
To a mixture of methyl 2-bromo-4-chloro-1- (2-trimethylsilylethoxymethyl) pyrrolo [2, 3-b] pyridine-5-carboxylate (1.3 g, 3.10 mmol) and 2, 6-dichloroaniline (752.63 mg, 4.65 mmol) in toluene (15 mL) was added AlMe3 (2 M, 3.10 mL) at 20 ℃. The mixture was stirred at 20 ℃under N2 for 30 min. The mixture was stirred at 100 ℃ under N2 for 15 h. The reaction mixture was added H2O (60 mL) and extracted with ethyl acetate (60 mL × 3) . The combined organic phases were washed with brine (30 mL × 3) , dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=100/1 to 5/1) to afford the desired product.
LCMS: MS (ESI) m/z 550.0 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.58 (s, 1H) , 8.52 (s, 1H) , 7.63 (s, 1H) , 7.62 -7.60 (m, 1H) , 7.45 -7.40 (m, 1H) , 7.05 -7.02 (m, 1H) , 5.72 (s, 2H) , 3.58 (t, J = 8.0 Hz, 2H) , 0.85 (t, J = 8.0 Hz, 2H) , -0.08 (s, 9H) .
Step D: 2-bromo-N- (2, 6-dichlorophenyl) -4- (methylamino) -1- (2-
trimethylsilylethoxymethyl) pyrrolo [2, 3-b] pyridine-5-carboxamide
A solution of 2-bromo-4-chloro-N- (2, 6-dichlorophenyl) -1- (2-trimethylsilylethoxymethyl) pyrrolo [2, 3-b] pyridine-5-carboxamide (850 mg, 1.55 mmol) , MeNH2 (970 mg, 9.37 mmol, 1 mL, 30%purity) were taken up into a microwave tube in NMP (7 mL) . The sealed tube was heated at 100 ℃ for 1 hr under microwave. The mixture was allowed to cool to room temperature, diluted with brine (60 mL) , and extracted with EtOAc (3 ×
50 mL) . The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=100/1 to 5/1) to afford the desired product.
LCMS: MS (ESI) m/z 545.1 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.60 (s, 1H) , 7.44 (s, 1H) , 7.42 (s, 1H) , 7.25 -7.19 (m, 1H) , 6.93 (s, 1H) , 5.31 (s, 2H) , 3.68 -3.61 (m, 2H) , 3.32 (d, J = 5.6 Hz, 3H) , 0.98 -0.90 (m, 2H) , -0.02 (s, 9H) .
Step E: 4-bromo-11- (2, 6-dichlorophenyl) -13-methyl-5- (2-trimethylsilylethoxymethyl) -
5, 7, 11, 13-tetrazatricyclo [7.4.0.02
, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
A solution of 2-bromo-N- (2, 6-dichlorophenyl) -4- (methylamino) -1- (2-trimethylsilylethoxymethyl) pyrrolo [2, 3-b] pyridine-5-carboxamide (700 mg, 1.29 mmol) , di (imidazol-1-yl) methanone (1.04 g, 6.44 mmol) and DIPEA (2.60 g, 20.09 mmol, 3.50 mL) were taken up into a microwave tube in DMAC (3.5 mL) . The sealed tube was heated at 145 ℃for 2 hr under microwave. The mixture was allowed to cool to room temperature, diluted with H2O (60 mL) , and extracted with EtOAc (3 × 50 mL) . The combined organic extracted with brine (3 × 50 mL) , dried with Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=100/1 to 5/1) to afford the desired product.
LCMS: MS (ESI) m/z 571.1 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 8.89 (s, 1H) , 7.73 (s, 1H) , 7.71 (s, 1H) , 7.61 -7.56 (m, 1H) , 7.48 (s, 1H) , 5.75 (s, 2H) , 3.94 (s, 3H) , 3.58 (t, J = 8.0 Hz, 2H) , 0.85 (t, J = 8.0 Hz, 2H) , -0.14 -0.03 (m, 9H) .
Step F: tert-butyl (3S, 4S) -4- [4- [11- (2, 6-dichlorophenyl) -13-methyl-10, 12-dioxo-5- (2-
trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02
, 6] trideca-1 (9) , 2 (6) , 3, 7-
tetraen-4-yl] pyrazol-1-yl] -3-fluoro-piperidine-1-carboxylate
To a solution of tert-butyl (3S, 4S) -3-fluoro-4- [4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrazol-1-yl] piperidine-1-carboxylate (100.00 mg, 252.99 μmol) , 4-bromo-11- (2, 6-dichlorophenyl) -13-methyl-5- (2-trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (144.29 mg, 252.99 μmol) in dioxane (2.5 mL) and H2O (0.5 mL) was added Pd (dppf) Cl2 (18.51 mg, 25.30 μmol) , K2CO3 (69.93 mg, 505.98 μmol) under N2. The mixture was stirred at 90 ℃ for 1 h. The mixture was allowed to cool to room temperature, diluted with H2O (30 mL) , and extracted with EtOAc (3 ×30 mL) . The combined organic extracted with brine (3 × 50 mL) , dried with Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by
column chromatography (SiO2, Petroleum ether/Ethyl acetate=100/1 to 5/1) to afford the desired product.
LCMS: MS (ESI) m/z760.1 [M+H] +.
Step G: 11- (2, 6-dichlorophenyl) -4- [1- [ (3S, 4S) -3-fluoro-4-piperidyl] pyrazol-4-yl] -5-
(hydroxymethyl) -13-methyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-
tetraene-10, 12-dione
To a solution of tert-butyl (3S, 4S) -4- [4- [11- (2, 6-dichlorophenyl) -13-methyl-10, 12-dioxo-5- (2-trimethylsilylethoxymethyl) -5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] -3-fluoro-piperidine-1-carboxylate (105 mg, 138.39 μmol) in DCM (3 mL) was added HCl/dioxane (4 M, 3 mL) and TFA (767.50 mg, 6.73 mmol, 0.5 mL) . The mixture was stirred at 20 ℃ for 1 h. The reaction mixture was concentrated under reduced pressure to give residue. To the residue was added toluene (3 mL) . The mixture was concentrated in vacuo to give a residue. Repeat the preceding operation three times to afford the desired product.
LCMS: MS (ESI) m/z 558.2 [M+H] +.
Step H: tert-butyl 9- [ [ (3S, 4S) -4- [4- [11- (2, 6-dichlorophenyl) -13-methyl-10, 12-dioxo-
5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] -3-fluoro-
1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate
To a solution of 11- (2, 6-dichlorophenyl) -4- [1- [ (3S, 4S) -3-fluoro-4-piperidyl] pyrazol-4-yl] -5- (hydroxymethyl) -13-methyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (82 mg, 137.85 μmol, HCl) , tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (58.18 mg, 165.42 μmol) in DCM (3 mL) was added TEA (41.85 mg, 413.55 μmol, 57.56 μL) and NaBH (OAc) 3 (87.65 mg, 413.55 μmol) . The mixture was stirred at 20 ℃ for 1 h. The mixture was allowed to cool to room temperature, diluted with H2O (30 mL) , and extracted with EtOAc (3 × 30 mL) . The combined organic extracted with brine (3 × 50 mL) , dried with Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol=100 /1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 793.7 [M+H] +.
Step I: 4- [1- [ (3S, 4S) -1- (3-azaspiro [5.5] undecan-9-ylmethyl) -3-fluoro-4-piperidyl] pyrazol-4-
yl] -11- (2, 6-dichlorophenyl) -13-methyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-
1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
To a solution of tert-butyl 9- [ [ (3S, 4S) -4- [4- [11- (2, 6-dichlorophenyl) -13-methyl-10, 12-dioxo-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraen-4-yl] pyrazol-1-yl] -3-fluoro-1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate (57 mg, 71.81 μmol) in HCl/dioxane (4 M, 3 mL) was stirred at 20 ℃ for 16 h. The mixture was allowed to cool to room temperature,
diluted with H2O (30 mL) , and extracted with EtOAc (3 × 30 mL) . The combined organic extracted with brine (3 × 50 mL) , dried with Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 693.4 [M+H] +.
Step J: 4- {1- [ (3S, 4S) -1- ( {3- [4-chloro-3- (2, 4-dioxo-1, 3, 5, 6-tetrahydro-1-
pyrimidinyl) benzoyl] -3-aza-9-spiro [5.5] undecyl} methyl) -3-fluoro-4-piperidyl] -4-
pyrazolyl} -11- (2, 6-dichlorophenyl) -13-methyl-5, 7, 11, 13-tetraazatricyclo [7.4.0.02, 6] trideca-
1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione
To a solution of 4- [1- [ (3S, 4S) -1- (3-azaspiro [5.5] undecan-9-ylmethyl) -3-fluoro-4-piperidyl] pyrazol-4-yl] -11- (2, 6-dichlorophenyl) -13-methyl-5, 7, 11, 13-tetrazatricyclo [7.4.0.02, 6] trideca-1 (9) , 2 (6) , 3, 7-tetraene-10, 12-dione (37 mg, 53.34 μmol) and 4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoic acid (17.20 mg, 64.01 μmol) in DMF (1 mL) was added HATU (30.42 mg, 80.01 μmol) and DIPEA (20.68 mg, 160.03 μmol, 27.87 μL) . The mixture was stirred at 20 ℃ for 1 h. The crude product was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 6%-46%B over 9 min) to afford the desired product.
LCMS: MS (ESI) m/z 945.1 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 12.81 (s, 1H) , 10.52 (s, 1H) , 8.78 (s, 1H) , 8.43 (s, 1H) , 8.16 (s, 1H) , 7.72 (s, 1H) , 7.70 (s, 1H) , 7.63 (d, J = 8.0 Hz, 1H) , 7.60 -7.57 (m, 1H) , 7.57 -7.54 (m, 1H) , 7.39 (dd, J = 1.6, 8.4 Hz, 1H) , 7.33 (d, J = 2.0 Hz, 1H) , 5.02 -4.70 (m, 1H) , 4.40 (s, 1H) , 4.00 (s, 3H) , 3.84 -3.69 (m, 1H) , 3.67 -3.49 (m, 3H) , 3.32 -3.22 (m, 4H) , 2.87 (s, 1H) , 2.79 -2.65 (m, 2H) , 2.26 (s, 2H) , 2.15 -2.00 (m, 3H) , 1.75-1.65 (m, 2H) , 1.63 -1.40 (m, 5H) , 1.39 -1.24 (m, 2H) , 1.16-1.10 (m, 4H) .
19F NMR (400 MHz, DMSO-d6) δ = -185.69
Example 177:
3- (6- {4- [ (4- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -1, 1-difluoroethyl} -1-piperidyl) methyl] -1-piperidyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-367)
Scheme:
Step A: tert-butyl 4- [2- [4- (4-nitrophenyl) piperazin-1-yl] acetyl] piperidine-1-carboxylate
To a solution of 1- (4-nitrophenyl) piperazine (338.39 mg, 1.63 mmol) and tert-butyl 4- (2-bromoacetyl) piperidine-1-carboxylate (500 mg, 1.63 mmol) in MeCN (5 mL) was added K2CO3 (225.68 mg, 1.63 mmol) and stirred at 80 ℃ for 12hr. The reaction mixture was concentrated under reduced pressure to give a residue and purified by flash silica gel chromatography (20gSilica Flash Column, Eluent of 0~3%DCM/MeOH gradient @25 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 433.3 [M+H] +.
Step B: tert-butyl 4- [1, 1-difluoro-2- [4- (4-nitrophenyl) piperazin-1-yl] ethyl] piperidine-1-
carboxylate
To a solution of tert-butyl 4- [2- [4- (4-nitrophenyl) piperazin-1-yl] acetyl] piperidine-1-carboxylate (600 mg, 1.39 mmol) in DCE (6 mL) was added DAST (6.71 g, 41.62 mmol) at 0-25 ℃ and stirred at 25 ℃ for 12hr. The reaction mixture was quenched with ice water, and then extracted with DCM (50mL *3) . The combined organic layers were dried over Na2SO4 filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (12gSilica Flash Column, Eluent of 5%DCM/MeOH gradient @15 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 455.2 [M+H] +.
Step C: 1- [2, 2-difluoro-2- (4-piperidyl) ethyl] -4- (4-nitrophenyl) piperazine
To a solution of tert-butyl 4- [1, 1-difluoro-2- [4- (4-nitrophenyl) piperazin-1-yl] ethyl] piperidine-1-carboxylate (500 mg, 1.10 mmol) in DCM (3 mL) was added TFA (1 mL) and stirred at 25 ℃for 2hr . The reaction mixture was concentrated under reduced pressure, then washed with water (10ml) and basified with Na2CO3. Then the mixture was extracted with DCM (50mL*3) . The organic phase was dried over Na2SO4 and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 355.5 [M+H] +.
Step D: 5- [4- [ [4- [1, 1-difluoro-2- [4- (4-nitrophenyl) piperazin-1-yl] ethyl] -1-
piperidyl] methyl] -1-piperidyl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione
To a solution of 1- [2, 2-difluoro-2- (4-piperidyl) ethyl] -4- (4-nitrophenyl) piperazine (500 mg, 1.41 mmol) and 1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] piperidine-4-carbaldehyde (521.13 mg, 1.41 mmol) in DCM (10 mL) was added TEA (285.53 mg, 2.82 mmol) , NaBH (OAc) 3 (897.06 mg, 4.23 mmol, ) . The mixture was stirred at 25 ℃ for 12hr. The reaction mixture was quenched with water and then extracted with EA/MeOH=10: 1 (50 mL *3) . The combined organic layers were dried overNa2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (
20 gFlash Column, Eluent of0~10%PE/EA/MeOH=10: 10: 1gradient @30 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 708.5 [M+H] +.
Step E: 3- [6- [4- [ [4- [2- [4- (4-aminophenyl) piperazin-1-yl] -1, 1-difluoro-ethyl] -1-
piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 5- [4- [ [4- [1, 1-difluoro-2- [4- (4-nitrophenyl) piperazin-1-yl] ethyl] -1-piperidyl] methyl] -1-piperidyl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (390 mg, 551.03 μmol) in TFA (4 mL) was added Zn (1.550 g, 23.70 mmol) and the mixture was stirred at 75 ℃for 12hr. The reaction mixture was filtered, washed with MeCN (5mL × 3) and concentrated to give crude product. The crude product was purified by reversed-phase HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-22%B over 6 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 664.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ10.99 (s, 1H) , 7.44 (d, J = 8.4 Hz, 1H) , 7.30 (dd, J = 2.0 Hz, 1H) , 7.24 -7.17 (m, 3H) , 7.05 (d, J = 8.8 Hz, 2H) , 5.09 (dd, J = 5.2, 13.3 Hz, 1H) , 4.34 (d, J =16.8 Hz, 1H) , 4.21 (d, J = 16.8 Hz, 1H) , 3.79 (br d, J = 12.0Hz, 2H) , 3.62 (br d, J = 11.2 Hz, 2H) , 3.30 (m, 6H) , 3.10 -2.85 (m, 9H) , 2.79 (br t, J = 11.6 Hz, 2H) , 2.70 -2.55 (m, 1H) , 2.46 -2.29 (m, 3H) , 2.08 -1.92 (m, 4H) , 1.90 -1.69 (m, 5H) , 1.41 -1.28 (m, 2H) .
Step F: 3- (6- {4- [ (4- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -1, 1-difluoroethyl} -1-piperidyl) methyl] -1-piperidyl} -1-oxo-
2-isoindolinyl) -2, 6-piperidinedione
To the solution of 3- [6- [4- [ [4- [2- [4- (4-aminophenyl) piperazin-1-yl] -1, 1-difluoro-ethyl] -1-piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (70 mg, 105.45 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (40.65 mg, 105.45 μmol) in n-BuOH (2 mL) was added AcOH (63.33 mg, 1.05 mmol) , the mixture was stirred at 100 ℃ for 2hr. The reaction mixture was concentrated under reduced pressure to give a residue and purified by reversed-phase HPLC (column: Xtimate C18 100*30mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 16%-46%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 985.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.97 (s, 1H) , 10.14 -9.96 (m, 1H) , 8.82 (s, 1H) , 8.19 (s, 1H) , 8.07 -7.96 (m, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.58 m, 3H) , 7.40 (d, J = 8.4 Hz, 1H) , 7.24 (dd, J = 8.4 Hz, 1H) , 7.14 (d, J = 2.0 Hz, 1H) , 6.91 (d, J = 9.2 Hz, 2H) , 5.44 -5.21 (m, 3H) , 5.09 (dd,
J =13.2 Hz, 1H) , 4.51 -4.39 (m, 2H) , 4.32 (dd, J = 16.8 Hz, 1H) , 4.19 (dd, J = 16.8 Hz, 1H, 3.74 (br d, J = 11.6 Hz, 2H) , 3.08 (m, 4H) , 2.92 (m, 3H) , 2.80 (br t, J = 14.8 Hz, 2H) , 2.73 -2.64 (m, 6H) , 2.59 (br d, J = 17.2 Hz, 1H) , 2.37 (m, J = 4.4, 13.3 Hz, 2H) , 2.14 (br d, J = 6.8 Hz, 2H) , 2.04 -1.90 (m, 4H) , 1.88 -1.66 (m, 8H) , 1.56 (m, 3H) , 1.49 -1.37 (m, 2H) , 1.26 -1.14 (m, 2H) .
Example 178:
3- (7- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2-phthalazinyl) -2, 6-piperidinedione (Cpd-368)
Scheme:
Step A: 3- (7-bromo-1-oxo-phthalazin-2-yl) -1- [ (4-methoxyphenyl) methyl] piperidine-2, 6-
dione
To a solution of 7-bromo-2H-phthalazin-1-one (1.5 g, 6.67 mmol) in THF (30 mL) was added t-BuOK (898 mg, 8.00 mmol) at 0 ℃. After stirring at 0 ℃ for 0.5 hr under nitrogen atmosphere, [1- [ (4-methoxyphenyl) methyl] -2, 6-dioxo-3-piperidyl] trifluoromethanesulfonate (2.80 g, 7.33 mmol) was added to the reaction mixture at 0 ℃. After the addition was finished, the reaction mixture was stirred at 0 ℃ for 2 hr. The reaction mixture was quenched by addition NH4Cl (30 mL) , extracted with ethyl acetate (30 mL×3) . The combined organic layers were washed with brine (30.0 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a
residue, which was triturated with ethyl acetate (15.0 mL) at 20 ℃ for 30 min, filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 455.9, 457.9 [M+H] +.
1H NMR (400MHz, CDCl3) δ 8.59 (d, J = 1.6 Hz, 1H) , 8.16 (s, 1H) , 7.95 (dd, J = 2.0, 8.4 Hz, 1H) , 7.59 (d, J = 8.4 Hz, 1H) , 7.38 -7.31 (m, 2H) , 6.86 -6.79 (m, 2H) , 5.86 (dd, J = 5.2, 12.0 Hz, 1H) , 5.02 -4.86 (m, 2H) , 3.81 -3.75 (m, 3H) , 3.06 -2.97 (m, 1H) , 2.90 -2.66 (m, 2H) , 2.29 -2.19 (m, 1H) .
Step B: 1- [ (4-methoxyphenyl) methyl] -3- [7- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 3- (7-bromo-1-oxo-phthalazin-2-yl) -1- [ (4-methoxyphenyl) methyl] piperidine-2, 6-dione (500 mg, 1.10 mmol) , K3PO4 (698 mg, 3.29 mmol) and 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (400 mg, 1.12 mmol) in dioxane (15 mL) was added Pd2 (dba) 3 (100 mg, 110 μmol) and RuPhos (26 mg, 54.7 μmol) . The mixture was stirred at 100 ℃ for 15 hr under nitrogen atmosphere. The reaction mixture was diluted with ethyl acetate (15 mL) and filtered through a pad of Celite. The filter was concentrated in vacuum. The residue was triturated with ethyl acetate (15 mL) at 20 ℃ for 30 min, filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 734.3 [M+H] +.
1H NMR (400MHz, CDCl3) δ 8.13 (d, J = 9.2 Hz, 2H) , 8.02 (s, 1H) , 7.69 (s, 1H) , 7.55 (d, J =8.8 Hz, 1H) , 7.40 -7.34 (m, 3H) , 6.83 (dd, J = 6.0, 8.8 Hz, 4H) , 5.89 -5.81 (m, 1H) , 5.01 -4.89 (m, 2H) , 3.79 (s, 3H) , 3.48 -2.41 (m, 8H) , 3.03 -2.95 (m, 1H) , 2.88 -2.77 (m, 2H) , 2.76 -2.72 (m, 4H) , 2.33 -2.16 (m, 2H) , 1.81 (t, J = 13.2 Hz, 4H) , 1.71 (br s, 2H) , 1.56 -1.39 (m, 3H) , 1.29 -1.18 (m, 3H) .
Step C: 3- [7- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-
phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 1- [ (4-methoxyphenyl) methyl] -3- [7- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (600 mg, 817.6 μmol) in DCE (30.0 mL) was added AlCl3 (1.09 g, 8.18 mmol) . The mixture was stirred at 85℃ for 2 hr. The reaction mixture was quenched by addition water (20.0 mL) , adjusted pH=7 by saturated Na2CO3 (aq. ) , extracted with DCM/MeOH (10/1, 15 mL ×5) . The combined organic layers were washed with brine (20 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was triturated with ethyl acetate (15.0 mL) at 20 ℃ for 30 min, filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 614.4 [M+H] +.
Step D: 3- [7- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-
phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 3- [7- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (200 mg, 325.8 μmol) in DMF (2.0 mL) was added hypoboric acid (87.65 mg, 977.6 μmol) and 4- (4-pyridyl) pyridine (0.5 mg, 3.26 μmol) . The mixture was stirred at 20 ℃ for 5 min. The reaction mixture was diluted with ethyl acetate (8.0 mL) , yellow solid was precipitated, filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 584.4 [M+H] +.
Step E: 3- (7- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2-phthalazinyl) -2, 6-
piperidinedione
To a suspension of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (100 mg, 259.4 μmol) and 3- [7- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (227 mg, 389.1 μmol) in n-BuOH (6.0 mL) was added HOAc (155 mg, 2.59 mmol) . The mixture was stirred at 100 ℃ for 2 hr. under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was diluted with DMF (5.0 mL) . The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 30%-60%B over 8 min) , after freeze-drying to give a yellow solid (32 mg) , . which was repurified by prep-HPLC (column: Welch Xtimate C18 150*25mm*5um; mobile phase: [water (NH4HCO3) -ACN] ; gradient: 55%-85%B over 7 min) , after freeze-drying to afford the desired product.
LCMS: MS (ESI) m/z 905.9 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ11.01 (br s, 1H) , 10.07 (br s, 1H) , 8.82 (s, 1H) , 8.23 (s, 1H) , 8.02 (s, 1H) , 7.75 (d, J = 8.4 Hz, 2H) , 7.62 -7.52 (m, 4H) , 7.47 (s, 1H) , 6.90 (d, J = 9.2 Hz, 2H) , 5.76 (d, J = 6.8 Hz, 1H) , 5.45 -5.29 (m, 2H) , 5.28 (s, 1H) , 4.52 -4.37 (m, 2H) , 3.12 -3.00 (m, 5H) , 3.02 -3.00 (m, 1H) , 2.98 -2.86 (m, 1H) , 2.67 -2.60 (m, 5H) , 2.59 -2.54 (m, 2H) , 2.27 -2.15 (m, 2H) , 2.13 -2.02 (m, 2H) , 2.01 -1.88 (m, 2H) , 1.78 -1.58 (m, 8H) , 1.56 (m, 3H) , 1.48 -1.34 (m, 4H) , 1.19 -1.10 (m, 2H) .
Example 179:
3- [5- (4- { [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) -1, 3-dioxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-100)
Scheme:
Step A: 2- (2, 6-dioxo-3-piperidyl) -5- [4- (hydroxymethyl) -1-piperidyl] isoindoline-1, 3-dione
The mixture of 4-piperidylmethanol (7 g, 60.78 mmol) , 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (13.43 g, 48.62 mmol) in NMP (50 mL) was added DIEA (15.71 g, 121.56 mmol, 21.17 mL) . The mixture was stirred at 100 ℃ for 2 h under N2. The reaction mixture was quenched by addition water 20 mL, and then extracted with EtOAc (100 mL × 3) . The combined organic layers were washed with brine (100 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by triturated from Ethyl acetate (15 mL) and Petroleum ether (5 mL) at 25 ℃ for 16 hr. The reaction mixture was filtered to afford the desired product.
LCMS MS (ESI) m/z = 372.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.07 (s, 1H) , 7.64 (d, J = 8.4 Hz, 1H) , 7.30 (d, J = 2.0 Hz, 1H) , 7.23 (dd, J = 2.4, 8.8 Hz, 1H) , 5.06 (dd, J = 5.2, 12.8 Hz, 1H) , 4.49 (t, J = 5.2 Hz, 1H) , 4.14 -3.98 (m, 2H) , 3.29 -3.24 (m, 2H) , 3.02 -2.90 (m, 2H) , 2.90 -2.77 (m, 1H) , 2.64 -2.52 (m, 2H) , 2.06 -1.95 (m, 1H) , 1.73 (d, J = 13.2 Hz, 2H) , 1.69 -1.60 (m, 1H) , 1.25 -1.11 (m, 2H)
Step B: 1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] piperidine-4-carbaldehyde
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [4- (hydroxymethyl) -1-piperidyl] isoindoline-1, 3-dione (1 g, 2.69 mmoL) in DCM (10 mL) was added DMP (1.37 g, 3.23 mmol, 1.00 mL) at 0℃. The mixture was stirred at 25℃ for 1 hr. The mixture was added water 10 mL, extracted with ethyl acetate (100 mL × 3) . The combined organic layers were dried over Na2SO4, filtered and concentrated in vacuum to afford the desired product.
LCMS MS (ESI) m/z = 370.2 [M+H] +.
Step C: 2- (2, 6-dioxo-3-piperidyl) -5- [4- [ [3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-9-
yl] methyl] -1-piperidyl] isoindoline-1, 3-dione
To solution of 3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecane (350 mg, 1.27 mmol) and 1- [2- (2, 6-dioxo-3-piperidyl) -1, 3-dioxo-isoindolin-5-yl] piperidine-4-carbaldehyde (516.47 mg, 1.40 mmol) in DCM (10 mL) was added TEA (385.88 mg, 3.81 mmol, 530.78 μL) . The mixture was stirred at 20 ℃ for 1 hr. Then the mixture was added NaBH (OAc) 3 (404.11 mg, 1.91 mmol) and stirred at 20 ℃ for 1 hr. The mixture was diluted with water (20 mL) and extracted with EtOAc (100 mL × 3) . The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~5%MeOH/DCM gradient @30 mL/min) to afford the desired product.
LCMS MS (ESI) m/z = 629.5 [M+H] +.
Step D: 5- [4- [ [3- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] methyl] -1-piperidyl] -2-
(2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [4- [ [3- (4-nitrophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] methyl] -1-piperidyl] isoindoline-1, 3-dione (150 mg, 238.58 μmol) in THF (10 mL) and DCM (1 mL) was added Pd/C (20 mg, 18.79 μmol, 10%purity) under N2 atmosphere. The suspension was degassed and purged with H2 for 3 times. The mixture was stirred under H2 (15 Psi) at 25 ℃ for 6 hr. The reaction mixture was filtered via celite pad and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 0~10%MeOH/DCM gradient @30 mL/min) to afford the desired product.
LCMS MS (ESI) m/z = 599.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ11.09 (s, 1H) , 7.66 (d, J = 8.4 Hz, 1H) , 7.33 (s, 1H) , 7.25 (s, 1H) , 6.69 (d, J = 8.8 Hz, 2H) , 6.48 (d, J = 8.4 Hz, 2H) , 5.07 (dd, J = 5.6, 12.8 Hz, 1H) , 4.10 (d, J = 5.2 Hz, 1H) , 3.17 (d, J = 5.2 Hz, 2H) , 2.97 (t, J = 12.4 Hz, 2H) , 2.89 (d, J = 6.8 Hz, 4H) , 2.62 -2.55 (m, 6H) , 2.42 -2.39 (m, 2H) , 1.80 (d, J = 11.6 Hz, 3H) , 1.55 (m, 8H) .
Step E: 3- [5- (4- { [9- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -3, 9-diaza-3-spiro [5.5] undecyl] methyl} -1-piperidyl) -1, 3-dioxo-2-
isoindolinyl] -2, 6-piperidinedione
To a solution of 5- [4- [ [3- (4-aminophenyl) -3, 9-diazaspiro [5.5] undecan-9-yl] methyl] -1-piperidyl] -2- (2, 6-dioxo-3-piperidyl) isoindoline-1, 3-dione (35 mg, 58.46 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfonyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (23.47 mg, 58.46 μmol) in t-BuOH (3 mL) was added TFA (60 mg, 526.21 μmol, 39.09 μL) . The mixture was stirred at 100 ℃ for 2 hr. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Boston Prime C18 150*30mm*5um; mobile phase: [water (TFA) -ACN] ; gradient: 16%-56%B over 9 min ) to afford the desired product.
LCMS MS (ESI) m/z = 920.3 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ11.08 (s, 1H) , 10.09 (s, 1H) , 8.84 (s, 2H) , 8.04 -7.96 (m, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.68 (d, J = 8.4 Hz, 1H) , 7.54 -7.65 (m, 3H) , 7.37 (s, 1H) , 7.28 (d, J =8.0 Hz, 1H) , 7.03 (s, 1H) , 5.30 (s, 2H) , 5.07 (dd, J = 5.6, 12.8 Hz, 1H) , 4.47 (m, 2H) , 4.10 (d, J = 12.4 Hz, 2H) , 3.39 (d, J = 12.6 Hz, 1H) , 3.17 (m, 4H) , 3.10 -2.97 (m, 6H) , 2.97 -2.81 (m, 2H) , 2.61 (s, 1H) , 2.58-2.53 (m, 2H) , 2.15 (m, 3H) , 2.05 -1.93 (m, 3H) , 1.90 -1.75 (m, 6H) , 1.74 -1.59 (m, 4H) , 1.56 (m, 3H) , 1.32 -1.21 (m, 2H) .
Example 180:
3- (6- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2-phthalazinyl) -2, 6-piperidinedione (Cpd-369)
Scheme:
Step A: 1- [ (4-methoxyphenyl) methyl] -3- [6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione
A mixture of 3- (6-bromo-1-oxo-phthalazin-2-yl) -1- [ (4-methoxyphenyl) methyl] piperidine-2, 6-dione (500 mg, 1.10 mmol) , 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (392.82 mg, 1.10 mmol) , RuPhos (25.57 mg, 54.79 μmol) , Pd2 (dba) 3 (100.34 mg, 109.58 μmol) , K3PO4 (697.80 mg, 3.29 mmol) in dioxane (10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 16 h under N2 atmosphere. The mixture was filtered and concentrated to give a residue. The crude product was triturated with EtOAc (3
mL) at 25 ℃ for 1 hr, then the mixture was filtered and the solid was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 734.4 [M+H] +
Step B: 3- [6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-
phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 1- [ (4-methoxyphenyl) methyl] -3- [6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (700 mg, 953.87 μmol) in DCE (20 mL) was added AlCl3 (1.27 g, 9.54 mmol) . The mixture was stirred at 100 ℃ for 2 h. The mixture was poured into water (100 mL) and extracted with DCM (100 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 614.3 [M+H] +
Step C: 3- [6- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-
phthalazin-2-yl] piperidine-2, 6-dione
To a solution of 3- [6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (700 mg, 1.14 mmol) in DMF (10 mL) were added hypoboric acid (329.27 mg, 3.67 mmol) and 4- (4-pyridyl) pyridine (1.78 mg, 11.41 μmol) . The mixture was stirred at 20 ℃ for 0.5 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 2%-32%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 584.2 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ 11.00 (s, 1H) , 9.73 (TFA, 3H) , 8.23 (s, 1H) , 8.02 (d, J = 9.2 Hz, 1H) , 7.48 (dd, J = 2.0, 9.2 Hz, 1H) , 7.23 (d, J = 2.0 Hz, 1H) , 7.20 -7.13 (m, 2H) , 7.07 (d, J = 8.8 Hz, 2H) , 5.74 (dd, J = 5.2, 11.6 Hz, 1H) , 3.83 (s, 1H) , 3.45 (m, 7H) , 3.24 (d, J = 11.6 Hz, 4H) , 3.08 -2.85 (m, 2H) , 2.65 -2.56 (m, 1H) , 2.54 (d, J = 2.4 Hz, 1H) , 2.16 -2.02 (m, 1H) , 2.01 -1.81 (m, 4H) , 1.74 -1.54 (m, 4H) , 1.45 (m, 2H) , 1.20 (t, J = 12.4 Hz, 2H) .
Step D: 3- (6- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2-phthalazinyl) -2, 6-
piperidinedione
To a solution of 3- [6- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-phthalazin-2-yl] piperidine-2, 6-dione (151.44 mg, 259.44 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (100 mg, 259.44 μmol) in n-BuOH (5 mL) was added AcOH (77.90 mg, 1.30 mmol, 74.26 μL) and then the mixture was stirred at 100 ℃ for 3 h. The
reaction mixture concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 15%-45%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 905.6 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ11.00 (s, 1H) , 10.11 (TFA, 1H) , 9.41 (s, 1H) , 8.85 (s, 1H) , 8.23 (s, 1H) , 7.94-8.08 (m, 2H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.55-7.69 (m, 3H) , 7.45-7.53 (m, 1H) , 7.24 (s, 1H) , 7.00 (d, J = 8.8 Hz, 2H) , 5.69-5.79 (m, 1H) , 5.23-5.43 (m, 2H) , 4.37-4.58 (m, 2H) , 3.82 (d, J = 12.4 Hz, 2H) , 3.60-3.66 (m, 2H) , 3.42-3.45 (m, 3H) , 3.12-3.32 (m, 4H) , 2.85-3.00 (m, 3H) , 2.51-2.73 (m, 2H) , 2.13-2.34 (m, 1H) , 2.01-2.12 (m, 2H) , 1.78-2.01 (m, 6H) , 1.59-1.75 (m, 5H) , 1.56 (m, 3H) , 1.46 (m, 2H) , 1.15-1.27 (m, 2H) .
Example 181:
3- [6- (4- { [ (3R, 4S) -4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-fluoro-1-piperidyl] methyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-370)
Scheme:
Step A: tert-butyl 3-fluoro-4- [4- (4-nitrophenyl) piperazin-1-yl] piperidine-1-carboxylate
To a solution of 1- (4-nitrophenyl) piperazine (5 g, 24.13 mmol) , tert-butyl 3-fluoro-4-oxo-piperidine-1-carboxylate (6.29 g, 28.95 mmol) in DCM (100 mL) was added Et3N (7.32 g, 72.38 mmol) and NaBH (OAc) 3 (10.23 g, 48.26 mmol) . The mixture was stirred at 25℃ for 16hr. The mixture was added H2O (200 mL) , extracted with DCM (150 mL × 3) . The combined organic layers were washed with brine (60 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was triturated with EtOAc (30 mL) at 25℃ for 20 min. The mixture was filtered and the cake was dried in vacuo to afford the desired product.
LCMS MS (ESI) m/z 409.3 [M+H] +.
Step B: tert-butyl (3R, 4S) -3-fluoro-4- [4- (4-nitrophenyl) piperazin-1-yl] piperidine-1-
carboxylate&tert-butyl (3S, 4R) -3-fluoro-4- [4- (4-nitrophenyl) piperazin-1-yl] piperidine-1-
carboxylate
The crude product was purified by SFC (column: DAICEL CHIRALPAK IG (250mm*50mm, 10um) ; mobile phase: [CO2-EtOH (0.1%NH3H2O) ] ; B%: 50%, isocratic elution mode) and concentrated to give the product 1 and product 2.
The product 2 was purified by SFC (column: DAICEL CHIRALPAK IG (250mm*30mm, 10um) ; mobile phase: [CO2-EtOH (0.1%NH3H2O) ] ; B%: 50%%, isocratic elution mode) and concentrated to give the product 2. tert-butyl (3R, 4S) -3-fluoro-4- [4- (4-nitrophenyl) piperazin-1-yl] piperidine-1-carboxylate
LCMS MS (ESI) m/z 409.4 [M+H] +.
SFC: Rt = 5.920 min tert-butyl (3S, 4R) -3-fluoro-4- [4- (4-nitrophenyl) piperazin-1-yl] piperidine-1-carboxylate LCMS MS (ESI) m/z 409.3 [M+H] +.
SFC: Rt = 7.165 min
Step C: 1- [ (3R, 4S) -3-fluoro-4-piperidyl] -4- (4-nitrophenyl) piperazine
To a solution of tert-butyl (3R, 4S) -3-fluoro-4- [4- (4-nitrophenyl) piperazin-1-yl] piperidine-1-carboxylate (360 mg, 881.35 μmol) in DCM (5 mL) was added TFA (11.05 g, 96.93 mmol, 7.20 mL) . The mixture was stirred at 25 ℃ for 1hr. The mixture was concentrated to give a residue to afford the desired product.
LCMS MS (ESI) m/z 309.1 [M+H] +
Step D: 3- [6- [4- [ [ (3R, 4S) -3-fluoro-4- [4- (4-nitrophenyl) piperazin-1-yl] -1-
piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 1- [ (3R, 4S) -3-fluoro-4-piperidyl] -4- (4-nitrophenyl) piperazine (100 mg, 324.31 μmol) , 1- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde (138.31 mg, 389.17 μmol) in DCM (5 mL) was added Et3N (98.45 mg, 972.92 μmol) and NaBH (OAc) 3 (206.20 mg, 972.92 μmol) . The mixture was stirred at 25℃ for 1hr. The mixture was added H2O (10 mL) , and extracted with DCM (20 mL × 3) . The combined organic layers were washed with brine (5 mL × 3) dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. TLC (Dichloromethane : Methanol = 10: 1, Rf = 0.4) . The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 0~10 %Methanol/Dichloromethane gradient @20 mL/min) and the combined desired fractions was concentrated under reduced pressure to afford the desired product.
LCMS MS (ESI) m/z 648.4 [M+H] +.
Step E: 3- [6- [4- [ [ (3R, 4S) -3-fluoro-4- [4- (4-nitrophenyl) piperazin-1-yl] -1-
piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 3- [6- [4- [ [ (3R, 4S) -3-fluoro-4- [4- (4-nitrophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (100.00 mg, 154.38
μmol) in DMF (1 mL) was added 4- (4-pyridyl) pyridine (24.11 mg, 154.38 μmol) and hypoboric acid (13.84 mg, 154.38 μmol) . The mixture was stirred at 25℃ for 5min. The mixture was added H2O (2 mL) . The crude product was purified by reversed-phase column ( [water (0.05%TFA) -ACN] ; B%: 0%-50%, 15 min) and dried by lyophilization to afford the desired product.
LCMS MS (ESI) m/z 618.1 [M+H] +.
Step F: 3- [6- (4- { [ (3R, 4S) -4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-fluoro-1-piperidyl] methyl} -1-piperidyl) -1-oxo-2-
isoindolinyl] -2, 6-piperidinedione
To a solution of 3- [6- [4- [ [ (3R, 4S) -4- [4- (4-aminophenyl) piperazin-1-yl] -3-fluoro-1-piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (80 mg, 129.50 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (49.91 mg, 129.50 μmol) in n-BuOH (2 mL) was added AcOH (77.77 mg, 1.30 mmol) . The mixture was stirred at 100 ℃ for 1 hr. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 3%-33%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS MS (ESI) m/z 939.5 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.99 (s, 1H) , 10.10 (br s, 2H) , 8.85 (s, 1H) , 8.00 (t, J = 8.0 Hz, 1H) , 7.76 (d, J = 8.0 Hz, 1H) , 7.62 (dd, J = 8.4, 14.0 Hz, 3H) , 7.45 (d, J = 8.4 Hz, 1H) , 7.30 (br d, J = 8.4 Hz, 1H) , 7.21 (s, 1H) , 7.01 (br d, J = 8.8 Hz, 1H) , 7.05 -6.97 (m, 1H) , 5.72 -5.44 (m, 1H) , 5.42 -5.22 (m, 2H) , 5.09 (dd, J = 5.2, 13.2 Hz, 1H) , 4.48 (br s, 2H) , 4.38 -4.31 (m, 1H) , 4.26 -4.17 (m, 1H) , 3.94 (br s, 2H) , 3.80 (br s, 2H) , 3.45 -3.28 (m, 10H) , 3.04 (br s, 3H) , 2.97 -2.85 (m, 2H) , 2.79 (br t, J = 12.4 Hz, 2H) , 2.60 (br d, J = 16.8 Hz, 1H) , 2.43 -2.31 (m, 2H) , 2.19 (br s, 2H) , 2.08 -1.91 (m, 4H) , 1.82 (br s, 2H) , 1.71 (br s, 1H) , 1.57 (s, 2H) , 1.61 -1.52 (m, 1H) , 1.36 (br d, J = 11.6 Hz, 2H)
Example 182:
3- [6- (4- { [ (3S, 4R) -4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-fluoro-1-piperidyl] methyl} -1-piperidyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-371)
Scheme:
Step A: 1- [ (3S, 4R) -3-fluoro-4-piperidyl] -4- (4-nitrophenyl) piperazine
To a solution of tert-butyl (3S, 4R) -3-fluoro-4- [4- (4-nitrophenyl) piperazin-1-yl] piperidine-1-carboxylate (400.00 mg, 979.27 μmol) in DCM (4 mL) and TFA (1 mL) was and stirred at 25 ℃for 2hr. The reaction mixture was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 309.1 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 9.67 -9.38 (m, 1H) , 9.21 -8.91 (m, 1H) , 8.12 (d, J = 9.2 Hz, 2H) , 7.13 (d, J = 9.6 Hz, 2H) , 5.66 -5.48 (m, 1H) , 3.97 -3.96 (m, 1H) , 4.01 -3.57 (m, 5H) , 3.52 -3.17 (m, 6H) , 3.03 (br d, J = 8.4 Hz, 1H) , 2.36 -2.23 (m, 1H) , 2.16 -2.02 (m, 1H)
Step B: 3- [6- [4- [ [ (3S, 4R) -3-fluoro-4- [4- (4-nitrophenyl) piperazin-1-yl] -1-
piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 1- [ (3S, 4R) -3-fluoro-4-piperidyl] -4- (4-nitrophenyl) piperazine (100.00 mg, 324.31 μmol) , 1- [2- (2, 6-dioxo-3-piperidyl) -3-oxo-isoindolin-5-yl] piperidine-4-carbaldehyde (400 mg, 1.13 mmol) in DCM (3 mL) was added Et3N (98.45 mg, 972.92 μmol ) and NaBH (OAc) 3 (206.20 mg, 972.92 μmol) . The mixture was added H2O (30 mL) , extracted with DCM (20 mL × 3) . The combined organic layers were washed with brine (15 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 0~10 %Methanol/Dichloromethane gradient @30 mL/min) and the combined desired fractions was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 648.4 [M+H] +.
Step C: 3- [6- [4- [ [ (3S, 4R) -4- [4- (4-aminophenyl) piperazin-1-yl] -3-fluoro-1-
piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 3- [6- [4- [ [ (3S, 4R) -3-fluoro-4- [4- (4-nitrophenyl) piperazin-1-yl] -1-piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (200 mg, 308.77 μmol) in DMF (1 mL) was added 4- (4-pyridyl) pyridine (0.5 mg, 3.09 μmol) and hypoboric acid (84 mg, 926.30 μmol) . The mixture was stirred at 25 ℃ for 5min. Upon completion, the mixture was concentrated in vacuo. The crude product was purified by reversed-phase column (0.05 %TFA condition) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 618.1 [M+H] +.
Step D: 3- [6- (4- { [ (3S, 4R) -4- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-fluoro-1-piperidyl] methyl} -1-piperidyl) -1-oxo-2-
isoindolinyl] -2, 6-piperidinedione
To a solution of 3- [6- [4- [ [ (3S, 4R) -4- [4- (4-aminophenyl) piperazin-1-yl] -3-fluoro-1-piperidyl] methyl] -1-piperidyl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (50 mg, 80.94 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (32 mg, 80.94 μmol) in n-BuOH (1 mL) was added AcOH (47 μL, 809.38 μmol) . The mixture was stirred at 100 ℃ for 1.5 hr. Upon completion, the mixture was concentrated in vacuo. The crude product
was purified by reversed-phase HPLC (column: C18 100 × 40 mm; mobile phase: [water (TFA) -ACN] ; gradient: 3 %-33 %B over 8 min) to afford the desired product.
LCMS: MS (ESI) m/z 939.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H) , 10.26 -10.00 (m, 1H) , 8.85 (s, 1H) , 8.01 (s, 1H) , 7.76 (d, J = 8.0 Hz, 1H) , 7.67 -7.58 (m, 3H) , 7.44 (d, J = 8.0 Hz, 1H) , 7.29 (dd, J = 1.6, 7.6 Hz, 1H) , 7.21 (s, 1H) , 7.00 (d, J = 8.8 Hz, 2H) , 5.48 -5.19 (m, 3H) , 5.16 -5.04 (m, 1H) , 4.48 (d, J = 1.2 Hz, 2H) , 4.38 -4.31 (m, 1H) , 4.25 -4.18 (m, 1H) , 3.89 -3.76 (m, 6H) , 3.15 -3.04 (m, 3H) , 2.92 (m, 3H) , 2.78 (d, J = 3.2 Hz, 3H) , 2.69 -2.56 (m, 3H) , 2.46 -2.31 (m, 3H) , 2.30 -2.13 (m, 3H) , 2.00 (dd, J = 4.2, 11.2 Hz, 5H) , 1.81 (d, J = 4.2 Hz, 3H) , 1.75 -1.67 (m, 1H) , 1.57 (s, 3H) , 1.43 -1.28 (m, 2H) .
Example 183:
3- (6- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2, 4-diaza-2-indanyl) -2, 6-piperidinedione (Cpd-372)
Scheme:
Step A: methyl 5-bromo-2- (bromomethyl) pyridine-3-carboxylate
To a solution of methyl 5-bromo-2-methyl-pyridine-3-carboxylate (3.0 g, 13.04 mmol) in CCl4 (30 mL) were added NBS (2.79 g, 15.65 mmol) and BPO (95 mg, 391.2 μmol) . The mixture was stirred at 80 ℃ for 15 hr. The reaction mixture was filtered and the filter was concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=20/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 307.9, 309.9 [M+H] +.
Step B: 3- (3-bromo-5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-yl) piperidine-2, 6-dione
To a solution of methyl 5-bromo-2- (bromomethyl) pyridine-3-carboxylate (3.8 g, 12.30 mmol) and 3-aminopiperidine-2, 6-dione; hydrochloride (2.02 g, 12.30 mmol) in DMF (40.0 mL) was added TEA (3.73 g, 36.90 mmol) . The mixture was stirred at 80 ℃ for 2hr. The reaction mixture was concentrated in vacuum. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 0/100) to afford the desired product.
LCMS: MS (ESI) m/z 323.9, 325.9 [M+H] +.
Step C: 3- [3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -5-oxo-7H-
pyrrolo [3, 4-b] pyridin-6-yl] piperidine-2, 6-dione
To a solution of 3- (3-bromo-5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-yl) piperidine-2, 6-dione (300 mg, 925.5 μmol) and 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (365 mg, 1.02 mmol) in dioxane (5 mL) was added 1, 3-bis [2, 6-bis (1-propylbutyl) phenyl] -4, 5-dichloro-2H-imidazol-1-ium-2-ide; 3-chloropyridine; dichloropalladium (45.02 mg, 46.2 μmol) and Cs2CO3 (905 mg, 2.78 mmol) . The mixture was stirred at 100 ℃ for 15 hr under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 0~5 MeOH/DCM gradient @18 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 602.4 [M+H] +.
Step D: 3- [3- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -5-oxo-7H-
pyrrolo [3, 4-b] pyridin-6-yl] piperidine-2, 6-dione
To a solution of 3- [3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-yl] piperidine-2, 6-dione (260 mg, 432.1 μmol) in DMF (3.0 mL) were added hypoboric acid (116.22 mg, 1.30 mmol) and 4- (4-pyridyl) pyridine (3.37 mg, 21.6 μmol) . The mixture was stirred at 20 ℃ for 5min. The reaction mixture was concentrated in vacuum. The residue was triturated with ethyl acetate (10.0 mL) , gray solid was precipitated, filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 572.4 [M+H] +.
Step E: 3- (6- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2, 4-diaza-2-indanyl) -2, 6-
piperidinedione
To a solution of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (45 mg, 116.75 μmol, 1 eq) and 3- [3- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-yl] piperidine-2, 6-dione (135 mg, 236.1 μmol) in n-BuOH (5.0 mL) was added HOAc (70.11 mg, 1.17 mmol) . The mixture was stirred at 100 ℃ for 2hr. The reaction mixture was concentrated in vacuum. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 12%-42%B over 8 min) , after freeze-drying to give a yellow solid, which was repurified by prep-HPLC (column: Welch Xtimate C18 150*25mm*5um; mobile phase: [water (NH4HCO3) -ACN] ; gradient: 35%-65%B over 7 min) , after freeze-drying to afford the desired product.
LCMS: MS (ESI) m/z 893.6 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.00 (s, 1H) , 10.06 (s, 1H) , 8.82 (s, 1H) , 8.53 (d, J = 2.0 Hz, 1H) , 8.02 (br s, 1H) , 7.75 (d, J = 8.4Hz, 1H) , 7.57 (br t, J = 8.0 Hz, 3H) , 7.52 (d, J = 2.4 Hz, 1H) , 6.90 (d, J = 9.2 Hz, 2H) , 5.42 -5.30 (m, 2H) , 5.29 (s, 1H) , 5.15 (dd, J = 5.6, 13.2 Hz, 1H) , 4.45 (br s, 2H) , 4.38 (br d, J = 17.2 Hz, 1H) , 4.24 -4.18 (m, 1H) , 3.16 -3.02 (m, 6H) , 2.97 -2.85 (m, 2H) , 2.72 -2.61 (m, 4H) , 2.57 (br s, 1H) , 2.40 -2.35 (m, 2H) , 2.33 -2.19 (m, 2H) , 2.05 -1.91 (m, 4H) , 1.80 -1.68 (m, 2H) , 1.67 -1.60 (m, 4H) , 1.56 (s, 3H) , 1.48 -1.39 (m, 4H) , 1.14 (t, J = 11.2 Hz, 2H) .
Example 184:
3- (5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10. 03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2, 4-diaza-2-indanyl) -2, 6-piperidinedione (Cpd-373)
Scheme:
Step A: methyl 2- (bromomethyl) -6-chloro-pyridine-3-carboxylate
To a solution of methyl 6-chloro-2-methyl-pyridine-3-carboxylate (3.0 g, 16.16 mmol) in CCl4 (150 mL) was added AIBN (531 mg, 3.23 mmol) and NBS (2.53 g, 14.22 mmol) . The mixture was stirred at 80 ℃ for 15 hr. under nitrogen atmosphere. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=20/1 to 10/1) to afford the desired product.
LCMS: MS (ESI) m/z 264.0, 266.0 [M+H] +.
Step B: methyl 6-chloro-2- [ [ (2, 6-dioxo-3-piperidyl) amino] methyl] pyridine-3-carboxylate
To a solution of methyl 2- (bromomethyl) -6-chloro-pyridine-3-carboxylate (3.5 g, 13.23 mmol) and 3-aminopiperidine-2, 6-dione; hydrochloride (1.40 g, 8.51 mmol) in MeCN (35.0 mL) was added DIPEA (10.26 g, 79.39 mmol) . The mixture was stirred at 85 ℃ for 3hr. The reaction
mixture was concentrated in vacuum. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 0/1) to afford the desired product.
LCMS: MS (ESI) m/z 321.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ11.04 (s, 1H) , 8.21 (d, J = 8.0 Hz, 1H) , 7.68 (d, J = 8.0 Hz, 1H) , 5.18 (dd, J = 5.2, 13.2 Hz, 1H) , 4.61 -4.50 (m, 1H) , 4.43 -4.32 (m, 1H) , 2.99 -2.85 (m, 1H) , 2.60 (d, J = 17.6 Hz, 1H) , 2.42 (dd, J = 4.4, 13.2 Hz, 1H) , 2.04 -1.97 (m, 1H) .
Step C: 3- (2-chloro-5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-yl) piperidine-2, 6-dione
To a solution of methyl 6-chloro-2- [ [ (2, 6-dioxo-3-piperidyl) amino] methyl] pyridine-3-carboxylate (1.1 g, 3.53 mmol) in MeCN (20.0 mL) was added DIPEA (1.0 mL, 5.74 mmol) . The mixture was stirred at 85 ℃ for 15 hr. The reaction mixture was concentrated in vacuum. The residue was triturated with Petroleum ether: Ethyl acetate (1: 1, 10.0 mL) at 20 ℃ for 30 min, filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 280.0 [M+H] +.
Step D: 3- [2- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -5-oxo-7H-
pyrrolo [3, 4-b] pyridin-6-yl] piperidine-2, 6-dione
To a solution of 3- (2-chloro-5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-yl) piperidine-2, 6-dione (300 mg, 1.07 mmol) and 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (423 mg, 1.18 mmol) in DMAc (5.0 mL) was added DIPEA (693 mg, 5.36 mmol) . The mixture was stirred at 100 ℃ for 15 hr. The reaction mixture was concentrated in vacuum. The crude product was triturated with ethyl acetate (5.0 mL) at 20 ℃ for 30 min, filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 602.3 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.17 -8.10 (m, 2H) , 7.83 (d, J = 8.8 Hz, 1H) , 6.83 (d, J = 9.6 Hz, 3H) , 6.65 (d, J = 8.8 Hz, 1H) , 5.21 (dd, J = 5.2, 13.2 Hz, 1H) , 4.34 -4.15 (m, 2H) , 3.69 -3.62 (m, 4H) , 3.46 -3.40 (m, 4H) , 2.91 -2.85 (m, 1H) , 2.77 -2.71 (m, 4H) , 2.46 -2.15 (m, 4H) , 1.82 (br t, J = 14.8 Hz, 4H) , 1.72 -1.60 (m, 4H) , 1.53 -1.39 (m, 4H) .
Step E: 3- [2- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -5-oxo-7H-
pyrrolo [3, 4-b] pyridin-6-yl] piperidine-2, 6-dione
To a solution of 3- [2- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-yl] piperidine-2, 6-dione (250 mg, 415.4 μmol) and TEA (84 mg, 830.9 μmol) in DCM (8.0 mL) and DMAc (2 mL) were added PdCl2 (74 mg, 415.4 μmol) and Et3SiH (1.33 mL, 8.31 mmol) . The mixture was stirred at 20 ℃ for 15hr under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 0~10 MeOH/DCM gradient @18 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 572.4 [M+H] +.
Step F: 3- (5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2, 4-diaza-2-indanyl) -2, 6-
piperidinedione
To a suspension of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (45 mg, 116.7 μmol) and 3- [2- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -5-oxo-7H-pyrrolo [3, 4-b] pyridin-6-yl] piperidine-2, 6-dione (134 mg, 233.5 μmol) in n-BuOH (5.0 mL) was added HOAc (70 mg, 1.17 mmol) . The mixture was stirred at 100 ℃ for 2hr under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 12%-42%B over 8 min) , after freeze-drying to afford the desired product.
LCMS: MS (ESI) m/z 893.6 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.97 (s, 1H) , 10.14 (s, 1H) , 9.53 (s, 1H) , 8.85 (s, 1H) , 8.01 (t, J = 7.6 Hz, 1H) , 7.75 (d, J = 8.8 Hz, 2H) , 7.61 (dd, J = 8.0, 13.6 Hz, 3H) , 7.00 (d, J = 9.2 Hz, 2H) , 6.91 (d, J = 8.8 Hz, 1H) , 5.46 -5.20 (m, 2H) , 5.07 (dd, J = 5.2, 13.2 Hz, 1H) , 4.47 (br s, 2H) , 4.32 -4.03 (m, 2H) , 3.65 -3.54 (m, 6H) , 3.35 -3.10 (m, 4H) , 3.01 -2.85 (m, 3H) , 2.69 -2.56 (m, 1H) , 2.42 -2.30 (m, 1H) , 2.27 -2.11 (m, 1H) , 2.10 -1.78 (m, 8H) , 1.77 -1.57 (m, 5H) , 1.56 (m, 3H) , 1.44 -1.34 (m, 2H) , 1.19 (t, J = 12.0 Hz, 2H) .
Example 185:
N- (2, 6-dioxo-3-piperidyl) -6- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-pyridinecarboxamide (Cpd-374)
Scheme:
Step A: methyl 6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] pyridine-2-carboxylate
To a solution of 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (500 mg, 1.39 mmol) , methyl 6-bromopyridine-2-carboxylate (361.58 mg, 1.67 mmol) , Pd2 (dba) 3 (127.72 mg, 139.48 μmol) , RuPhos (32.54 mg, 69.74 μmol) and K3PO4 (888.20 mg, 4.18 mmol) in dioxane (5 mL) was degassed and purged with N2 for 3 times, then the mixture was stirred at 100 ℃ for 3 h under N2 atmosphere. The mixture was poured into water (50 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers were washed with brine (50 mL *2) , dried over Na2SO4, filtered and concentrated to give a residue. The reaction mixture was triturated with EtOAc (10 mL) for 1 h at 20 ℃ to afford the desired product.
LCMS: MS (ESI) m/z 494.3 [M+H] +.
Step B: 6- (9- (4- (4-nitrophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecan-3-yl) picolinic acid
To a solution of methyl 6- (9- (4- (4-nitrophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecan-3-yl) picolinate (516 mg, 1.05 mmol) in THF (6 mL) , MeOH (2 mL) and H2O (2 mL) was added LiOH (75.11 mg, 3.14 mmol) . The mixture was stirred at 20 ℃ for 2.5 h. The solution was diluted with water (30 mL) and filtered. The reaction mixture was concentrated under reduced pressure to give crude product. The mixture was poured into water (30 mL) and extracted with EtOAc (30 mL) . Then the aqueous layers were adjusted the pH = 3 by aq HCl (3 M) . Then the mixture was filtered to get the residue to afford the desired product.
LCMS: MS (ESI) m/z 480.3 [M+H] +.
Step C: N- (2, 6-dioxo-3-piperidyl) -6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] pyridine-2-carboxamide
To a solution of 6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridine-2-carboxylic acid (254.8 mg, 531.31 μmol) and 3-aminopiperidine-2, 6-dione (81.69 mg, 637.57 μmol) in DMF (5 mL) was added HATU (303.03 mg, 796.96 μmol) and DIEA (206.00 mg, 1.59 mmol, 277.63 μL) . The mixture was stirred at 25 ℃ for 3 h. The reaction mixture was concentrated under reduced pressure. And added H2O (50 mL) and extracted with EtOAc (100 mL × 2) . The combined organic phases were dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 590.3 [M+H] +.
Step D: 6- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-
3-piperidyl) pyridine-2-carboxamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridine-2-carboxamide (273.9 mg, 464.49 μmol) in DMF (3 mL) was added hypoboric acid (124.93 mg, 1.39 mmol) and 4- (4-pyridyl) pyridine (725.45 μg, 4.64 μmol) . The mixture stirred at 20 ℃ for 5 min. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by reversed-phase column (C18, MeOH in H2O = 40%, 0.1%TFA) , then dried by lyophilization to give the desired product to afford the desired product.
LCMS: MS (ESI) m/z 560.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.88 (s, 1H) , 8.75 (d, J = 8.0 Hz, 1H) , 7.67 (t, J = 8.0 Hz, 1H) , 7.27 (d, J = 8.4 Hz, 3H) , 7.10 (d, J = 8.8 Hz, 2H) , 7.04 (d, J = 8.8 Hz, 1H) , 4.76 -4.68 (m, 4H) , 3.86-3.84 (m, 1H) , 3.59 (s, 6H) , 3.22-3.12 (m, 5H) , 2.84 -2.75 (m, 1H) , 2.68 (s, 1H) , 2.54 (s, 1H) , 2.27-2.16 (m, 1H) , 2.01 -1.98 (m, 2H) , 1.88 (d, J = 12.8 Hz, 2H) , 1.77 -1.68 (m, 2H) , 1.60 (m, 2H) , 1.40 (m, 2H) , 1.23 -1.15 (m, 2H) .
Step E: N- (2, 6-dioxo-3-piperidyl) -6- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-pyridinecarboxamide
To a solution of 6- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-3-piperidyl) pyridine-2-carboxamide (104.88 mg, 155.67 μmol, TFA) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) in n-BuOH (2 mL) was added AcOH (77.90 mg, 1.30 mmol, 74.26 μL) . The mixture was stirred at 100 ℃ for 1 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: C18 150×30mm; mobile phase: [water (TFA) -ACN] ; gradient: 15%-55%B over 9 min) to afford the desired product.
LCMS: MS (ESI) m/z 881.6 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.90 (s, 1H) , 10.22 -9.92 (TFA, 0.87H) , 9.42 -9.38 (m, 1H) , 8.85 (s, 1H) , 8.75 (d, J = 8.0 Hz, 1H) , 8.00 (t, J = 7.6 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.70 -7.59 (m, 4H) , 7.28 (d, J = 7.2 Hz, 1H) , 7.05 (d, J = 8.8 Hz, 1H) , 7.00 (d, J = 9.2 Hz, 2H) , 5.39 -5.24 (m, 2H) , 4.77-4.70 (m, 1H) , 4.48-4.46 (m, 1H) , 3.83-3.79 (m, 1H) , 3.60 (m, 6H) , 3.29 -3.18 (m, 4H) , 2.98 -2.89 (m, 2H) , 2.85-2.76 (m, 1H) , 2.55-2.54 (m, 1H) , 2.26-2.15 (m, 2H) , 2.04 -2.01 (m, 2H) , 2.00 -1.88 (m, 6H) , 1.73 -1.56 (m, 9H) , 1.41 (m, 2H) , 1.23-1.18 (m, 2H) .
Example 186:
N- (2, 6-dioxo-3-piperidyl) -5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-pyrimidinecarboxamide (Cpd-375)
Scheme:
Step A: methyl 5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] pyrimidine-2-carboxylate
To a solution of 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (170 mg, 474.2 μmol) , K3PO4 (302 mg, 1.42 mmol) and methyl 5-bromopyrimidine-2-carboxylate (103 mg, 474.2 μmol) in dioxane (5 mL) was added Pd2 (dba) 3 (44 mg, 47.4 μmol) and RuPhos (11 mg, 23.7 μmol) . The mixture was stirred at 100 ℃ for 15 hr. under nitrogen atmosphere. The reaction mixture was poured into water (25.0 mL) , yellow solid was precipitated, filtered. The filtered cake was triturated with MeOH (5.0 mL) at 20 ℃ for 30 min, filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 495.3 [M+H] +.
Step B: 5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyrimidine-2-
carboxylic acid
To a solution of methyl 5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyrimidine-2-carboxylate (190 mg, 384.2 μmol) in MeOH (2.0 mL) , THF (2.0 mL) and water (1.0 mL) was added LiOH·H2O (81 mg, 1.92 mmol) . The mixture was stirred at 60 ℃ for 2 hr.
The reaction was acidified with 1 M HCl (aq) to pH=5, the reaction mixture was concentrated to remove organic solvent, yellow solid was precipitated. The suspension was diluted with water (5.0 mL) , filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 481.4 [M+H] +.
Step C: N- (2, 6-dioxo-3-piperidyl) -5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] pyrimidine-2-carboxamide
To a solution of 5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyrimidine-2-carboxylic acid (140 mg, 291.3 μmol) and 3-aminopiperidine-2, 6-dione; hydrochloride (58 mg, 349.5 μmol) in DMF (2.5 mL) were added DIPEA (188 mg, 1.46 mmol) and HATU (166 mg, 436.9 μmol) . The mixture was stirred at 20 ℃ for 1hr. The reaction mixture was poured into water (15.0 mL) , and extracted with DCM/MeOH (10/1, 20 mL×3) , the combined organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 591.2 [M+H] +.
Step D: 5- (9- (4- (4-aminophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecan-3-yl) -N- (2, 6-
dioxopiperidin-3-yl) pyrimidine-2-carboxamide
To a solution of N- (2, 6-dioxopiperidin-3-yl) -5- (9- (4- (4-nitrophenyl) piperazin-1-yl) -3-azaspiro [5.5] undecan-3-yl) pyrimidine-2-carboxamide (145 mg, 245.4 μmol) in DMF (2.0 mL) was added hypoboric acid (66 mg, 736.4 μmol) and 4- (4-pyridyl) pyridine (0.4 mg, 2.45 μmol) . The mixture was stirred at 20 ℃ for 5 min . The reaction mixture was filtered and the filter was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-30%B over 6 min) , after freeze-drying to afford the desired product.
LCMS: MS (ESI) m/z 561.5 [M+H] +.
Step E: N- (2, 6-dioxo-3-piperidyl) -5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-pyrimidinecarboxamide
To a suspension of 5- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-3-piperidyl) pyrimidine-2-carboxamide (30 mg, 53.5 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (21 mg, 54.4 μmol) in n-BuOH (1.5 mL) was added HOAc (32 mg, 535.0 μmol) . The mixture was stirred at 100 ℃ for 2 hr under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 12%-42%B over 8 min) , after freeze-drying to afford the desired product.
LCMS: MS (ESI) m/z 882.9 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.86 (s, 1H) , 10.10 (br s, 1H) , 9.60 (br s, 1H) , 8.87 -8.79 (m, 2H) , 8.56 (m, 2H) , 8.00 (t, J=7.6 Hz, 1H) , 7.75 (d, J=8.0 Hz, 1H) , 7.62 (dd, J=8.4, 13.6 Hz, 3H) , 7.00 (d, J=9.2 Hz, 2H) , 5.42 -5.22 (m, 2H) , 4.80 -4.70 (m, 1H) , 4.47 (d, J=6.8 Hz, 2H) , 3.81 (d, J=11.6 Hz, 2H) , 3.62 (d, J=11.2 Hz, 2H) , 3.49 -3.37 (m, 4H) , 3.32 -3.14 (m, 3H) , 3.03 -2.92 (m, 2H) , 2.86 -2.73 (m, 1H) , 2.55 (br s, 1H) , 2.27 -2.10 (m, 2H) , 2.08 -1.85 (m, 7H) , 1.64 (m, 5H) , 1.76 -1.60 (m, 3H) , 1.50 -1.42 (m, 2H) , 1.20 (t, J=12.0 Hz, 2H) .
Example 187:
N- (2, 6-dioxo-3-piperidyl) -6- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -3-pyridazinecarboxamide (Cpd-376)
Scheme:
Step A: methyl 6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] pyridazine-3-carboxylate
To a solution of 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (200 mg, 557.9 μmol) and methyl 6-chloropyridazine-3-carboxylate (101 mg, 585.8 μmol) in dioxane (5.0 mL) was added DIPEA (216 mg, 1.67 mmol) . The mixture was stirred at 100 ℃ for 15 hr. The reaction mixture was concentrated in vacuum. The residue was triturated with ethyl acetate (20.0 mL) at 20 ℃ for 30 min. yellow solid was precipitated, filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 495.3 [M+H] +.
Step B: 6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridazine-3-
carboxylic acid
To a solution of methyl 6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridazine-3-carboxylate (200 mg, 404.3 μmol) in THF (2.0 mL) , MeOH (2.0 mL) and water (1.0 mL) was added LiOH·H2O (85 mg, 2.02 mmol) . The mixture was stirred at 60 ℃ for 2 hr. The reaction was acidified with 2 M HCl (aq) to pH=4, the reaction mixture was concentrated to
remove organic solvent, yellow solid was precipitated. The suspension was diluted with water (5.0 mL) , filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 481.4 [M+H] +.
Step C: N- (2, 6-dioxo-3-piperidyl) -6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] pyridazine-3-carboxamide
To a solution of 6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridazine-3-carboxylic acid (120 mg, 249.7 μmol) and 3-aminopiperidine-2, 6-dione; hydrochloride (49 mg, 299.6 μmol) in DMF (2.0 mL) were added DIPEA (161 mg, 1.25 mmol) and HATU (142 mg, 374.5 μmol) . The mixture was stirred at 20 ℃ for 1 hr. The reaction mixture was poured into water (15.0 mL) , extracted with DCM/MeOH (10/1, 20 mL×3) , the combined organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 591.3 [M+H] +.
Step D: 6- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-
3-piperidyl) pyridazine-3-carboxamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -6- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridazine-3-carboxamide (145 mg, crude) in DMF (2.0 mL) was added hypoboric acid (66 mg, 736.4 μmol) and 4- (4-pyridyl) pyridine (0.4 mg, 2.45 μmol) . The mixture was stirred at 20 ℃ for 5 min. The reaction mixture was filtered and the filter was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-22%B over 6 min) , after freeze-drying to afford the desired product.
LCMS: MS (ESI) m/z 561.3 [M+H] +.
Step E: N- (2, 6-dioxo-3-piperidyl) -6- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -3-pyridazinecarboxamide
To a suspension of 6- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-3-piperidyl) pyridazine-3-carboxamide (85 mg, 151.6 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (58 mg, 151.6 μmol) in n-BuOH (2 mL) was added HOAc (91 mg, 1.52 mmol) . The mixture was stirred at 100 ℃ for 2 hr under nitrogen atmosphere. The reaction mixture was concentrated in vacuum. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 8%-38%B over 8 min) , after freeze-drying to afford the desired product.
LCMS: MS (ESI) m/z 882.8 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ10.86 (s, 1H) , 10.12 (s, 1H) , 9.46 (s, 1H) , 9.07 (d, J=8.4 Hz, 1H) , 8.85 (s, 1H) , 8.00 (t, J=7.6 Hz, 1H) , 7.85 (d, J=9.6 Hz, 1H) , 7.75 (d, J=8.0 Hz, 1H) , 7.62 (dd, J=8.4, 13.6 Hz, 3H) , 7.38 (d, J=9.6 Hz, 1H) , 7.00 (d, J=9.2 Hz, 2H) , 5.42 -5.22 (m, 2H) , 4.85 -4.74 (m, 1H) , 4.48 -4.44 (m, 2H) , 3.82 (d, J=12.4 Hz, 2H) , 3.77 -3.69 (m, 4H) , 3.62 (d, J=10.8 Hz, 2H) , 3.33 -3.15 (m, 3H) , 2.95 (t, J=11.6 Hz, 2H) , 2.86 -2.74 (m, 1H) , 2.55 (s, 1H) , 2.28 -2.14 (m, 2H) , 2.06 -1.88 (m, 7H) , 1.75 -1.59 (m, 5H) , 1.56 (m, 3H) , 1.46 -1.39 (m, 2H) , 1.27 -1.15 (m, 2H) .
Example 188:
3- [5- (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-377)
Scheme:
Step A: 3- (5-bromo-1-oxo-isoindolin-2-yl) -1- [ (4-methoxyphenyl) methyl] piperidine-2, 6-
dione
To a solution of 3- (5-bromo-1-oxo-isoindolin-2-yl) piperidine-2, 6-dione (2 g, 6.19 mmol) , TBAI (457.22 mg, 1.24 mmol) , K2CO3 (1.71 g, 12.38 mmol) in DMF (20 mL) was added PMB-Cl (1.45 g, 9.28 mmol, 1.26 mL) at 0 ℃. The mixture was stirred at 25 ℃ for 16 h. The mixture was poured into water (100 mL) and extracted with EtOAC (200 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue. The crude product was triturated with EtOAc (10 mL) at 25 ℃ for 1 hr, then the mixture was filtered and the solid was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 443.2 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ 7.90 (s, 1H) , 7.76 -7.63 (m, 2H) , 7.18 (d, J = 8.8 Hz, 2H) , 6.85 (d, J = 8.8 Hz, 2H) , 5.24 (dd, J = 5.2, 13.6 Hz, 1H) , 4.75 (q, J = 14.4 Hz, 2H) , 4.55 -4.24 (m, 2H) , 3.71 (s, 3H) , 3.14 -3.02 (m, 1H) , 2.84 -2.75 (m, 1H) , 2.49 -2.35 (m, 1H) , 2.10 -2.01 (m, 1H) .
Step B: 1- [ (4-methoxyphenyl) methyl] -3- [5- [9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-
azaspiro [5.5] undecan-3-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione
A mixture of 9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane (500 mg, 1.34 mmol) , 3- (5-bromo-1-oxo-isoindolin-2-yl) -1- [ (4-methoxyphenyl) methyl] piperidine-2, 6-dione (595.01 mg, 1.34 mmol) , Pd2 (dba) 3 (122.91 mg, 134.23 μmol) , RuPhos (31.32 mg, 67.11 μmol) , K3PO4 (854.76 mg, 4.03 mmol) in dioxane (5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 16 h under N2 atmosphere. The mixture was poured into water (100 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue. The crude product was triturated with EtOAc (5 mL) at 25℃ for 1 hr, then the mixture was filtered and the solid was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 735.6 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ8.05 (d, J = 9.6 Hz, 2H) , 7.49 (d, J = 9.2 Hz, 1H) , 7.18 (d, J = 8.8 Hz, 2H) , 7.06 -6.99 (m, 4H) , 6.85 (d, J = 8.8 Hz, 2H) , 5.16 (dd, J = 5.2, 13.6 Hz, 1H) , 4.86 -4.65 (m, 2H) , 4.39 -4.10 (m, 2H) , 3.71 (s, 3H) , 3.44 (s, 4H) , 3.29 (s, 3H) , 3.14 -3.00 (m, 1H) , 2.78 (d, J = 16.4 Hz, 1H) , 2.44 (s, 4H) , 2.41 -2.31 (m, 1H) , 2.17 (d, J = 6.8 Hz, 2H) , 2.01 (d, J = 5.6 Hz, 1H) , 1.75 -1.47 (m, 8H) , 1.42 (s, 2H) , 1.14 -1.01 (m, 4H) .
Step C: 3- [5- [9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecan-3-yl] -1-
oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 1- [ (4-methoxyphenyl) methyl] -3- [5- [9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (620 mg, 843.67 μmol) in DCE (10 mL) was added AlCl3 (1.12 g, 8.44 mmol) . The mixture was stirred at 100 ℃ for 2 h. The mixture was poured into water (100 mL) and extracted with DCM (100 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 615.3 [M+H] +
Step D: 3- [5- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecan-3-yl] -1-
oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 3- [5- [9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (600 mg, 976.03 μmol) in DMF (10 mL) was added hypoboric acid (281.76 mg, 3.14 mmol) and 4- (4-pyridyl) pyridine (1.52 mg, 9.76 μmol) .
The mixture was stirred at 25 ℃ for 0.5 h. The mixture was concentrated to give a residue. The residue was purified by silica gel column chromatography (20%EtOAc in MeOH) to afford the desired product.
LCMS: MS (ESI) m/z 585.4 [M+H] +
Step E: 3- [5- (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) -1-oxo-2-isoindolinyl] -
2, 6-piperidinedione
A mixture of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) , 3- [5- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (113.78 mg, 194.58 μmol) , AcOH (77.90 mg, 1.30 mmol, 74.26 μL) in n-BuOH (3 mL) was stirred at 100 ℃ for 2 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: Xtimate C18 100*30mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 17%-47%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 906.7 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ10.96 (s, 1H) , 10.11 (TFA, 1H) , 9.28 (s, 1H) , 8.85 (s, 1H) , 8.07 -7.94 (m, 1H) , 7.76 (d, J = 8.0 Hz, 1H) , 7.62 (dd, J = 8.4, 11.2 Hz, 3H) , 7.52 (d, J = 8.8 Hz, 1H) , 7.10 -7.03 (m, 2H) , 6.99 (d, J = 9.2 Hz, 2H) , 5.50 -5.20 (m, 2H) , 5.05 (dd, J = 5.2, 13.2 Hz, 1H) , 4.56 -4.55 (m, 2H) , 4.37 -4.15 (m, 2H) , 3.75 (d, J = 12.4 Hz, 2H) , 3.38 -3.26 (m, 5H) , 3.23 -2.99 (m, 7H) , 2.96 -2.84 (m, 1H) , 2.59 (d, J = 16.8 Hz, 1H) , 2.46 -2.29 (m, 2H) , 2.19 (s, 1H) , 2.10 -1.90 (m, 3H) , 1.83 (s, 1H) , 1.73 (d, J = 8.8 Hz, 3H) , 1.62 (m, 4H) , 1.57 (m, 3H) , 1.46 (m, 2H) , 1.27 -1.14 (m, 4H) .
Example 189:
3- [6- (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-378)
Scheme:
Step A: tert-butyl 9- (methoxymethylene) -3-azaspiro [5.5] undecane-3-carboxylate
To a mixture of methoxymethyl (triphenyl) phosphonium; chloride (9.62 g, 28.05 mmol) in THF (30 mL) was added t-BuOK (3.78 g, 33.66 mmol) at 0 ℃ under N2. The mixture was stirred at 0℃ for 0.5 h and to the resulting mixture was added tert-butyl 9-oxo-3-azaspiro [5.5] undecane-3-carboxylate (3 g, 11.22 mmol) at 0 ℃ and the mixture was stirred for 3 hr at 25℃ under N2. The mixture was poured into aq. sat. NH4Cl (100 mL) , water (100 mL) and extracted with EtOAC (200 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by silica gel column chromatography (4%EtOAc in Petroleum ether) to afford the desired product.
1H NMR (400 MHz, CDCl3) δ5.77 (s, 1H) , 3.53 (s, 3H) , 3.40 -3.35 (m, 4H) , 2.18 (t, J = 6.4 Hz, 2H) , 2.00 -1.91 (m, 2H) , 1.46 (s, 9H) , 1.44 -1.37 (m, 8H) .
Step B: tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate
To a solution of tert-butyl 9- (methoxymethylene) -3-azaspiro [5.5] undecane-3-carboxylate (3 g, 10.16 mmol) in MeCN (60 mL) and H2O (2 mL) was added TFA (1.54 g, 13.46 mmol, 1 mL) . The mixture was stirred at 25 ℃ for 16 h. The mixture was poured into H2O (200 mL) and extracted with EtOAc (200 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to afford the desired product.
1H NMR (400 MHz, CDCl3) δ 9.66 (s, 1H) , 3.38 (d, J = 6.0, 12.0 Hz, 4H) , 2.32 -2.21 (m, 1H) , 1.85 -1.74 (m, 2H) , 1.73 -1.63 (m, 2H) , 1.61 -1.50 (m, 2H) , 1.47 (s, 9H) , 1.44 (d, J = 6.0 Hz, 2H) , 1.38 (t, J = 6.0 Hz, 2H) , 1.29 -1.23 (m, 2H) .
Step C: tert-butyl 9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-
carboxylate
To a solution of tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (3 g, 10.66 mmol) , 1- (4-nitrophenyl) piperazine (2.21 g, 10.66 mmol) in DCM (30 mL) was added TEA (3.24 g, 31.98 mmol, 4.45 mL) . After addition, the mixture was stirred at 25 ℃ for 1 h, and then NaBH (OAc) 3 (6.78 g, 31.98 mmol) was added. The mixture was stirred at 25 ℃ for 15 h. The mixture was poured into water (100 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by silica gel column chromatography (45%EtOAc in PE) to afford the desired product.
LCMS: MS (ESI) m/z 473.3 [M+H] +
Step D: 9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane
To a solution of tert-butyl 9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate (4.3 g, 9.10 mmol) in DCM (30 mL) was added TFA (9.21 g, 80.78 mmol, 6 mL) . The mixture was stirred at 25 ℃ for 2 h. The mixture was concentrated and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 373.3 [M+H] +
Step E: 2- (2, 6-dioxo-3-piperidyl) -5- [9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-
azaspiro [5.5] undecan-3-yl] isoindoline-1, 3-dione
To a solution of 9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane (1 g, 2.68 mmol) , 2- (2, 6-dioxo-3-piperidyl) -5-fluoro-isoindoline-1, 3-dione (741.52 mg, 2.68 mmol) in DMF (10 mL) was added DIEA (1.04 g, 8.05 mmol, 1.40 mL) . The mixture was stirred at 100 ℃ for 2 h. The mixture was poured into water (100 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue. The crude product was triturated with EtOAc (10 mL) at 25 ℃ for 1 hr, then the mixture was filtered and the solid was concentrated to afford the desired product.
LCMS: MS (ESI) m/z 629.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ11.06 (s, 1H) , 8.05 (d, J = 9.6 Hz, 2H) , 8.01 (dd, J = 4.4, 8.4 Hz, 1H) , 7.28 (s, 1H) , 7.24 -7.17 (m, 1H) , 7.02 (d, J = 9.6 Hz, 2H) , 5.06 (dd, J = 5.6, 12.8 Hz, 1H) , 3.44 (m, 8H) , 2.88 -2.82 (m, 1H) , 2.64 -2.54 (m, 3H) , 2.46 -2.42 (m, 4H) , 2.18 (d, J = 6.4 Hz, 2H) , 2.07 -2.00 (m, 1H) , 1.68 (d, J = 10.0 Hz, 2H) , 1.63 -1.54 (m, 4H) , 1.42 (m, 2H) , 1.15 -1.04 (m, 4H) .
Step F: 3- [6- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecan-3-yl] -1-
oxo-isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 2- (2, 6-dioxo-3-piperidyl) -5- [9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecan-3-yl] isoindoline-1, 3-dione (1 g, 1.59 mmol) in TFA (10 mL) was added Zn (4.11 g, 62.85 mmol) . The mixture was stirred at 75 ℃ for 2 h. The reaction mixture was filtered, washed with EtOAc (300mL) and concentrated to give crude product. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-26%B over 7 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 585.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H) , 9.84 (s, 2H) , 7.49 (d, J = 8.4 Hz, 1H) , 7.40 (d, J = 12.4 Hz, 2H) , 7.26 (d, J = 8.8 Hz, 2H) , 7.10 (d, J = 8.8 Hz, 2H) , 5.10 (dd, J = 5.2, 13.2 Hz, 1H) , 4.41 -4.20 (m, 2H) , 3.82 (s, 1H) , 3.59 (m, 2H) , 3.28 (m, 4H) , 3.22 -3.02 (m, 6H) , 2.98 -2.82 (m, 1H) , 2.60 (d, J = 16.8 Hz, 1H) , 2.45 -2.30 (m, 1H) , 2.05 -1.92 (m, 1H) , 1.89 -1.32 (m, 10H) , 1.24 -1.10 (m, 4H) .
2D NMR (400 MHz, DMSO-d6) was right.
Step G: 3- [6- (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] methyl} -3-aza-3-spiro [5.5] undecyl) -1-oxo-2-isoindolinyl] -
2, 6-piperidinedione
A mixture of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) , 3- [6- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (75.85 mg, 129.72 μmol) , AcOH (77.90 mg, 1.30 mmol, 74.26 μL) in n-BuOH (3 mL) was stirred at 100 ℃ for 2 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: Xtimate C18 100*30mm*3um; mobile phase: [water (TFA) -ACN] ; gradient: 17%-47%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 906.7 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.98 (s, 1H) , 10.12 (TFA, 1H) , 9.24 (s, 1H) , 8.85 (s, 1H) , 8.07 -7.93 (m, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.61 (dd, J = 8.4, 11.2 Hz, 3H) , 7.44 (d, J = 8.4 Hz, 1H) , 7.31 (d, J = 8.8 Hz, 1H) , 7.24 (s, 1H) , 6.99 (d, J = 9.2 Hz, 2H) , 5.45 -5.20 (m, 2H) , 5.09 (dd, J = 5.2, 13.2 Hz, 1H) , 4.55 -4.40 (m, 2H) , 4.39 -4.17 (m, 2H) , 3.75 (d, J = 12.0 Hz, 3H) , 3.29 -3.21 (m, 4H) , 3.19 -2.85 (m, 8H) , 2.65 -2.56 (m, 1H) , 2.44 -2.30 (m, 2H) , 2.18 (s, 1H) , 2.08 -1.90 (m, 3H) , 1.82 (s, 1H) , 1.72 (s, 3H) , 1.64 (m, 4H) , 1.56 (m, 3H) , 1.49 (m, 2H) , 1.26 -1.13 (m, 4H) .
Example 190:
N- (2, 6-dioxo-3-piperidyl) -4- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-pyridinecarboxamide (Cpd-379)
Scheme:
Step A: methyl 4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] pyridine-2-carboxylate
A mixture of 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (350 mg, 976.35 μmol) , methyl 4-bromopyridine-2-carboxylate (253.11 mg, 1.17 mmol) , Pd2 (dba) 3 (89.41 mg, 97.64 μmol) , RuPhos (22.78 mg, 48.82 μmol) and K3PO4 (621.74 mg, 2.93 mmol) in dioxane (10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 3 hr under N2 atmosphere. The mixture was poured into water (50 mL) and extracted with EtOAC (100 mL x 2) . The combined organic layers were dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by flash silica gel chromatography
(12 gSilica Flash Column, Eluent of 0~10%MeOH/DCM gradient @30 mL/min) to afford the desired product.
LCMS MS (ESI) m/z = 494.2 [M+H] +.
Step B: 4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridine-2-
carboxylic acid
To a solution of methyl 4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridine-2-carboxylate (200 mg, 405.19 μmol) in MeOH (2 mL) , THF (2 mL) and H2O (1 mL) was added LiOH. H2O (85.01 mg, 2.03 mmol) . The mixture was stirred at 25 ℃ for 3 hr. The reaction mixture was concentrated under reduced pressure and lyophilized directly to afford the desired product.
LCMS MS (ESI) m/z = 480.1 [M+H] +.
Step C: N- (2, 6-dioxo-3-piperidyl) -4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] pyridine-2-carboxamide
To a solution of 4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridine-2-carboxylic acid (150 mg, 312.78 μmol) and 3-aminopiperidine-2, 6-dione (61.78 mg, 375.34 μmol, HCl) in DMF (2 mL) was added HATU (178.39 mg, 469.17 μmol) and DIPEA (121.27 mg, 938.34 μmol, 163.44 μL) . The mixture was stirred at 20 ℃ for 1 hr. The combined organic layers were washed with brine (100 mL × 2) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS MS (ESI) m/z = 590.2 [M+H] +.
Step D: 4- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-
3-piperidyl) pyridine-2-carboxamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridine-2-carboxamide (180 mg, 305.25 μmol) in DMF (3 mL) were added hypoboric acid (82.10 mg, 915.75 μmol) and 4- (4-pyridyl) pyridine (476.74 μg, 3.05 μmol) . The mixture was stirred at 20 ℃ for 5 min. The reaction mixture was filtered to give a residue. The crude product was purified by reversed-phase column (C18, MeOH in H2O = 30%, 0.1%TFA) , then dried by lyophilization to afford the desired product.
LCMS MS (ESI) m/z = 560.0 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.98 (s, 1H) , 10.54 (s, 1H) , 8.16 (d, J = 7.2 Hz, 1H) , 7.81 (s, 1H) , 7.22 (d, J = 8.8 Hz, 3H) , 7.08 (br d, J = 9.2 Hz, 2H) , 4.99 -4.68 (m, 1H) , 3.83 (s, 1H) , 3.68 (m, 9H) , 3.15 (d, J = 12.0 Hz, 4H) , 2.93 -2.75 (m, 1H) , 2.44 (d, J = 2.0 Hz, 2H) , 2.22 (dt, J = 8.4, 12.8 Hz, 1H) , 2.07 -1.96 (m, 3H) , 1.90 (br d, J = 12.8 Hz, 2H) , 1.77 -1.62 (m, 4H) , 1.47 (m, 2H) , 1.31 -1.19 (m, 2H)
Step E: N- (2, 6-dioxo-3-piperidyl) -4- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-pyridinecarboxamide
To a solution of 4- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-3-piperidyl) pyridine-2-carboxamide (104.88 mg, 155.67 μmol, TFA) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) in n-BuOH (2 mL) was added AcOH (77.90 mg, 1.30 mmol, 74.26 μL) . The mixture was stirred at 100 ℃ for 16 hr. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: C18 150×30mm; mobile phase: [water (TFA) -ACN] ; gradient: 8%-48%B over 9 min) to afford the desired product.
LCMS MS (ESI) m/z = 881.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.99 (s, 1H) , 10.09 (TFA, 1H) , 9.61 (s, 1H) , 9.50 (s, 1H) , 8.84 (s, 1H) , 8.17 (d, J = 7.2 Hz, 1H) , 8.00 (t, J = 8.0 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.68 (s, 1H) , 7.64 -7.57 (m, 3H) , 7.21 (d, J = 5.6 Hz, 1H) , 7.00 (d, J = 9.2 Hz, 2H) , 5.33 (d, J = 10.4 Hz, 2H) , 4.91 -4.80 (m, 1H) , 4.46 (m, 2H) , 3.81 (d, J = 12.0 Hz, 2H) , 3.70 -3.59 (m, 5H) , 3.27 -3.16 (m, 2H) , 2.96 (t, J = 12.0 Hz, 3H) , 2.89 -2.77 (m, 1H) , 2.62 -2.54 (m, 1H) , 2.26 -2.11 (m, 2H) , 2.09 -1.84 (m, 8H) , 1.78 -1.61 (m, 5H) , 1.56 (m, 3H) , 1.48 (br s, 2H) , 1.29 -1.12 (m, 3H) .
Example 191:
N- (2, 6-dioxo-3-piperidyl) -5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-pyridinecarboxamide (Cpd-380)
Scheme:
Step A: methyl 5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] pyridine-2-carboxylate
To a solution of 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (250 mg, 697.40 μmol) , methyl 5-bromopyridine-2-carboxylate (180.79 mg, 836.87 μmol) , Pd2 (dba) 3 (63.86 mg, 69.74 μmol) , RuPhos (16.27 mg, 34.87 μmol) and K3PO4 (444.10 mg, 2.09 mmol) in dioxane (3 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 3 h under N2 atmosphere. The mixture was poured into water (50 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers were washed with brine (50 mL × 2) , dried
over Na2SO4, filtered and concentrated to give a residue. The reaction mixture was triturated with EtOAc (10 mL) for 1 h at 20 ℃ to afford the desired product.
LCMS: MS (ESI) m/z 494.3 [M+H] +.
Step B: 5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridine-2-
carboxylic acid
To a solution of methyl 5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridine-2-carboxylate (275.2 mg, 557.54 μmol) in DMAC (3 mL) and H2O (0.1 mL) was added LiOH (66.77 mg, 2.79 mmol) . The mixture was stirred at 60 ℃ for 2.5 h. The reaction mixture were adjusted the pH < 7 by aq HCl (3 M) . Then the mixture was concentrated under reduced pressure to give crude product to afford the desired product.
LCMS: MS (ESI) m/z 480.3 [M+H] +.
Step C: N- (2, 6-dioxo-3-piperidyl) -5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] pyridine-2-carboxamide
To a solution of 5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridine-2-carboxylic acid (275 mg, 573.43 μmol) and 3-aminopiperidine-2, 6-dione (88.17 mg, 688.12 μmol) in DMAC (3 mL) was added HATU (1.09 g, 2.87 mmol) and DIEA (741.12 mg, 5.73 mmol, 998.81 μL) . The mixture was stirred at 25 ℃ for 3 h. The mixture was poured into water (50 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers were washed with brine (50 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue to afford the desired product.
LCMS: MS (ESI) m/z 590.6 [M+H] +.
Step D: 5- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-
3-piperidyl) pyridine-2-carboxamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridine-2-carboxamide (412.3 mg, 699.19 μmol) in DMF (5 mL) was added hypoboric acid (188.05 mg, 2.10 mmol) and 4- (4-pyridyl) pyridine (1.09 mg, 6.99 μmol) . The mixture stirred at 20 ℃ for 5 min. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by reversed-phase column (C18, MeOH in H2O = 40%, 0.1%TFA) , then dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 560.1 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.85 (s, 1H) , 8.69 (d, J = 8.4 Hz, 1H) , 8.31 (d, J = 2.8 Hz, 1H) , 7.85 (d, J = 9.2 Hz, 1H) , 7.41 (dd, J = 2.8, 8.8 Hz, 1H) , 7.23 (d, J = 8.8 Hz, 2H) , 7.10 (d, J = 8.8 Hz, 2H) , 4.77-4.71 (m, 2H) , 3.87 (d, J = 12.4 Hz, 2H) , 3.63 (d, J = 11.6 Hz, 2H) , 3.37 (s, 4H) , 3.29 -3.17 (m, 4H) , 3.02 -2.97 (m, 2H) , 2.84 -2.75 (m, 1H) , 2.58-2.51 (m, 1H) , 2.22 -
2.10 (m, 1H) , 2.03 -1.98 (m, 1H) , 1.96 -1.86 (m, 4H) , 1.68 -1.60 (m, 4H) , 1.45 (m, 2H) , 1.22 -1.16 (m, 2H) .
Step E: N- (2, 6-dioxo-3-piperidyl) -5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-pyridinecarboxamide
To a solution of 5- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-3-piperidyl) pyridine-2-carboxamide (104.88 mg, 155.67 μmol, TFA) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) in n-BuOH (2 mL) was added AcOH (77.90 mg, 1.30 mmol, 74.26 μL) . The mixture was stirred at 100 ℃ for 1 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: C18 150×30mm; mobile phase: [Water (FA) -MeCN] ; gradient: 4%-44%B over 9 min) to afford the desired product.
LCMS: MS (ESI) m/z 881.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ10.84 (s, 1H) , 10.03 (TFA, 0.83H) , 8.81 (s, 1H) , 8.69 (d, J = 8.0 Hz, 1H) , 8.28 (d, J = 2.4 Hz, 1H) , 8.19 (s, 1H) , 8.01 (t, J = 7.6 Hz, 1H) , 7.84 (d, J = 8.8 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.57 (t, J = 6.4 Hz, 3H) , 7.37 (dd, J = 2.4, 9.2 Hz, 1H) , 6.88 (d, J = 9.2 Hz, 2H) , 5.37 -5.32 (m, 2H) , 4.77 -4.70 (m, 1H) , 4.44 (d, J = 5.2 Hz, 2H) , 3.32 (d, J = 3.6 Hz, 5H) , 3.06 (m, 4H) , 2.83 -2.74 (m, 1H) , 2.64 (s, 3H) , 2.54 (d, J = 3.2 Hz, 1H) , 2.27 -2.12 (m, 3H) , 2.03 -1.91 (m, 3H) , 1.74 -1.55 (m, 11H) , 1.41 -1.34 (m, 4H) , 1.11 (t, J = 12.4 Hz, 2H) .
Example 192:
N- (2, 6-dioxo-3-piperidyl) 3- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-fluorobenzamide (Cpd-381)
Scheme:
Step A: methyl 2-fluoro-3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] benzoate
To a solution of 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (400 mg, 1.12 mmol) , methyl 3-bromo-2-fluoro-benzoate (312.03 mg, 1.34 mmol) in dioxane (10 mL) was added Pd2 (dba) 3 (102.18 mg, 111.58 μmol) , RuPhos (26.03 mg, 55.79 μmol) and K3PO4 (710.56 mg, 3.35 mmol) under N2. The mixture was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 3 h under N2. The mixture was added H2O (10 mL) , and extracted withe EtOAc (30 mL × 3) . The combined organic phase were washed with brine (20 mL × 3) , dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure. The
residue was purified by silica gel column chromatography (6%MeOH in DCM) to afford the desired product.
LCMS: MS (ESI) m/z 511.4 [M+H] +.
Step B: 2-fluoro-3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] benzoic acid
To a solution of methyl 2-fluoro-3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzoate (240 mg, 470.04 μmol) in DMAC (4 mL) and H2O (0.04 mL) was added LiOH H2O (98.62 mg, 2.35 mmol) . The mixture was stirred at 60 ℃ for 16 h to afford the desired product.
LCMS: MS (ESI) m/z 497.2 [M+H] +.
Step C: N- (2, 6-dioxo-3-piperidyl) -2-fluoro-3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] benzamide
To a solution of 2-fluoro-3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzoic acid (240 mg, 391.48 μmol) , 3-aminopiperidine-2, 6-dione; hydrochloride (77.32 mg, 469.78 μmol) in DMAC (4 mL) was added HATU (223.28 mg, 587.23 μmol, ) and DIPEA (151.79 mg, 1.17 mmol, 204.57 μL) . The mixture was stirred at 25 ℃ for 2 h. The mixture was added H2O (30 mL) , filtered to give residue was desired product. The residue was wash DCM (30 mL) to give the desired product DCM solution, and dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 607.1 [M+H] +.
Step D: 3- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-
3-piperidyl) -2-fluoro-benzamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -2-fluoro-3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzamide (251 mg, 413.72 μmol) , in DMF (3 mL) was added hypoboric acid (111.27 mg, 1.24 mmol) and 4- (4-pyridyl) pyridine (1.94 mg, 12.41 μmol) . The mixture was stirred at 25 ℃ for 1 h. The mixture was filtered to give the crude product. The crude product was purified by reversed phase column (C18, MeOH in H2O = 27%, 0.1%TFA) , then dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 576.9 [M+H] +.
Step E: N- (2, 6-dioxo-3-piperidyl) 3- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-fluorobenzamide
To a solution of 3- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-3-piperidyl) -2-fluoro-benzamide (110 mg, 159.25 μmol, TFA salt) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-
1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) in n-BuOH (2 mL) was added AcOH (77.90 mg, 1.30 mmol, 74.26 μL) . The mixture was stirred at 100 ℃ for 1 h. The mixture was concentrated under reduce pressure to give a residue. The residue was purified by prep-HPLC (column: C18 150×30mm; mobile phase: [water (TFA) -ACN] ; gradient: 10%-50%B over 9 min) , then dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 898.6 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.88 (s, 1H) , 10.12 (TFA, 0.77H) , 9.29 (s, 1H) , 8.85 (s, 1H) , 8.58 (d, J=8.8 Hz, 1H) , 8.00 (t, J=7.6 Hz, 1H) , 7.75 (d, J=8.0 Hz, 1H) , 7.68 -7.56 (m, 3H) , 7.22 -7.12 (m, 3H) , 7.00 (br d, J=8.8 Hz, 2H) , 5.30 (m, 2H) , 4.84 -4.68 (m, 1H) , 4.47 (m, 2H) , 3.82 (d, J=13.2 Hz, 2H) , 3.69 -3.64 (m, 2H) , 3.29 -3.18 (m, 4H) , 3.00 (m, 4H) , 2.97 -2.88 (m, 2H) , 2.85 -2.73 (m, 1H) , 2.58 -2.53 (m, 1H) , 2.11 -1.85 (m, 9H) , 1.73 -1.60 (m, 5H) , 1.56 (m, 3H) , 1.52 -1.45 (m, 2H) , 1.25 -1.12 (m, 2H) .
19F NMR (400MHz, DMSO-d6) δ = -124.19
Example 193:
N- (2, 6-dioxo-3-piperidyl) 5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-fluorobenzamide (Cpd-382)
Scheme:
Step A: methyl 2-fluoro-5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] benzoate
A mixture of 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (300 mg, 836.87 μmol) , methyl 5-bromo-2-fluoro-benzoate (292.53 mg, 1.26 mmol) , Pd2 (dba) 3 (76.63 mg, 83.69 μmol) , K3PO4 (532.92 mg, 2.51 mmol) and RuPhos (19.53 mg, 41.84 μmol) in dioxane (10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100℃ for 16hr under N2 atmosphere. The mixture was concentrated to give a residue. The residue was purified by flash silica gel chromatography (8 gSilica Flash Column, Eluent of 0~100 %Ethyl acetate/Petroleum ether gradient @20 mL/min) and the combined desired fractions was concentrated under reduced pressure to afford the desired product.
LCMS MS (ESI) m/z 511.3 [M+H] +.
Step B: 2-fluoro-5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] benzoic acid
To a solution of methyl 2-fluoro-5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzoate (240 mg, 470.04 μmol) in THF (4 mL) and H2O (2 mL) was added LiOH·H2O (98.62 mg, 2.35 mmol) . The mixture was stirred at 60 ℃ for 2 hr . The reaction mixture was removed THF and the 1M HCl was added to adjust the pH = 3~4, the mixture was filtered and the cake was dried under reduced pressure to afford the desired product.
LCMS MS (ESI) m/z 497.3 [M+H] +
Step C: N- (2, 6-dioxo-3-piperidyl) -2-fluoro-5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] benzamide
To a solution of 2-fluoro-5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzoic acid (240 mg, 483.31 μmol) , 3-aminopiperidine-2, 6-dione (238.65 mg, 1.45 mmol) in DCM (10 mL) was added HATU (275.66 mg, 724.97 μmol) and DIPEA (312.32 mg, 2.42 mmol) . The mixture was stirred at 25 ℃ for 1 hr. The mixture was added H2O (30 mL) , and extracted with DCM (20 mL × 3) . The combined organic layers were washed with brine (15 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS MS (ESI) m/z 607.3 [M+H] +.
Step D: 5- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-
dioxo-3-piperidyl) -2-fluoro-benzamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -2-fluoro-5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzamide (250 mg, 412.07 μmol) in DMF (2 mL) was added 4- (4-pyridyl) pyridine (643.59 μg, 4.12 μmol) and hypoboric acid (110.83 mg, 1.24 mmol) . The mixture was stirred at 25℃ for 10min. The mixture was added DMF (1 mL) . The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-17%B over 7 min) and dried by lyophilization to afford the desired product.
LCMS MS (ESI) m/z 577.4 [M+H] +.
Step E: N- (2, 6-dioxo-3-piperidyl) 5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-fluorobenzamide
To a solution of 5- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-3-piperidyl) -2-fluoro-benzamide (80 mg, 138.72 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (53.47 mg, 138.72 μmolq) in n-BuOH (2 mL) was added AcOH (83.30 mg, 1.39 mmol, 79.41 μL) . The mixture was stirred at 100℃ for 2hr. The mixture was
concentrated to give a residue. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 7%-37%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS MS (ESI) m/z 898.4 [M+H] +.
1H NMR (400MHz, DMSO-d6) δ 10.87 (s, 1H) , 10.10 (br s, 1H) , 9.56 (br s, 1H) , 8.85 (s, 1H) , 8.48 (dd, J = 3.2, 8.0 Hz, 1H) , 8.00 (t, J = 8.0 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.61 (dd, J = 8.4, 13.2 Hz, 3H) , 7.17 (br d, J = 7.6 Hz, 3H) , 7.00 (d, J = 9.2 Hz, 2H) , 5.32 (br d, J = 18.4 Hz, 2H) , 4.79 -4.71 (m, 1H) , 4.46 (br s, 2H) , 3.81 (br d, J = 12.4 Hz, 2H) , 3.61 (br d, J = 11.2 Hz, 2H) , 3.30 -3.18 (m, 3H) , 3.15 (m, 4H) , 2.96 (br t, J = 11.6 Hz, 2H) , 2.86 -2.72 (m, 1H) , 2.55 (br s, 1H) , 2.27 -1.81 (m, 10H) , 1.74 -1.60 (m, 5H) , 1.56 (m, 3H) , 1.48 (br s, 2H) , 1.18 (br t, J = 12.4 Hz, 2H) .
Example 194:
N- (2, 6-dioxo-3-piperidyl) -5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-pyrazinecarboxamide (Cpd-383)
Scheme:
Step A: methyl 5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] pyrazine-2-carboxylate
To a solution of 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (550 mg, 1.53 mmol) and methyl 5-chloropyrazine-2-carboxylate (317.72 mg, 1.84 mmol) in dioxane (8 mL) was added DIPEA (991.47 mg, 7.67 mmol and the mixture was stirred at 100 ℃ for 12 hr. The reaction mixture was added EtOAc (10mL) , filtered and the cake was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 495.3 [M+H] +.
Step B: 5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyrazine-2-
carboxylic acid
To a solution of methyl 5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyrazine-2-carboxylate (504 mg, 1.02 mmol) in MeOH (8 mL) and H2O (2 mL) was added NaOH (407.59 mg, 10.19 mmol) and the mixture was stirred at 60 ℃ for 12hr. The reaction was acidified with 2 M HCl (aq) to pH=4, the reaction mixture was concentrated to remove organic solvent, yellow solid was precipitated. The suspension was diluted with water (5.0 mL) , filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 481.1 [M+H] +.
Step C: N- (2, 6-dioxo-3-piperidyl) -5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] pyrazine-2-carboxamide
To a solution of 5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyrazine-2-carboxylic acid (520 mg, 1.08 mmol) and HATU (617.16 mg, 1.62 mmol) in DMAC (1 mL) was added DIPEA (419.55 mg, 3.25 mmol) , then added 3-aminopiperidine-2, 6-dione (166.37 mg, 1.30 mmol) and the mixture was stirred at 25 ℃ for 1hr. The reaction mixture was poured into ice water, and then extracted with EtOAc (50mL *3) . The combined organic layers were dried over Na2SO4 filtered and concentrated under reduced pressure to give a residue to afford the desired product.
LCMS: MS (ESI) m/z 591.4 [M+H] +.
Step D: 5- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-
3-piperidyl) pyrazine-2-carboxamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyrazine-2-carboxamide (457 mg, 773.70 μmol) in DMF (5 mL) was added 4- (4-pyridyl) pyridine (1.21 mg, 7.74 μmol) , then added hypoboric acid (223.35 mg, 2.49 mmol) and the mixture was stirred at 25 ℃ for 5 min. The reaction mixture was concentrated under reduced pressure to give a residue and purified by reversed-phase HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-25%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 560.4 [M+H] +.
Step E: N- (2, 6-dioxo-3-piperidyl) -5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-pyrazinecarboxamide
To a solution of 5- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-3-piperidyl) pyrazine-2-carboxamide (100 mg, 178.35 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (68.74 mg, 178.35 μmol) in n-BuOH (2 mL) was added AcOH (107.10 mg, 1.78 mmol) and the mixture was stirred at 100 ℃ for 4 hr. The reaction
mixture was concentrated under reduced pressure to give a residue and purified by reversed-phase HPLC (column: Welch Xtimate C18 150*30mm*5um; mobile phase: [water (FA) -ACN] ; gradient: 25%-45%B over 7 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 882.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.85 (s, 1H) , 10.16 -9.95 (m, 1H) , 8.82 (s, 1H) , 8.64 -8.54 (m, 2H) , 8.28 (s, 1H) , 8.08 -7.97 (m, 1H) , 7.76 (d, J = 8.0 Hz, 1H) , 7.58 (br t, J = 7.6 Hz, 3H) , 6.90 (br d, J = 9.2 Hz, 2H) , 5.45 -5.23 (m, 3H) , 4.82 -4.70 (m, 1H) , 4.46 (br d, J = 4.8 Hz, 2H) , 3.70 (m, 4H) , 3.08 (m, 4H) , 2.80 (m, 1H) , 2.66 (m, 4H) , 2.54 (s, 1H) , 2.34 -2.13 (m, 3H) , 2.07 -1.91 (m, 3H) , 1.79 (br d, J = 12.0 Hz, 2H) , 1.75 -1.64 (m, 3H) , 1.56 (m, 5H) , 1.47 -1.36 (m, 4H) , 1.21 -1.10 (m, 2H) .
Example 195:
N- (2, 6-dioxo-3-piperidyl) m- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} benzamide (Cpd-384)
Scheme:
Step A: methyl 3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] benzoate
A mixture of 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (400 mg, 1.12 mmol) , methyl 3-bromobenzoate (360 mg, 1.67 mmol) , Pd2 (dba) 3 (103 mg, 111.58 μmol) , RuPhos (27 mg, 55.79 μmol) and Cs2CO3 (1.09 g, 3.35 mmol) in dioxane (5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 13 hr under N2 atmosphere. Upon completion, the mixture was concentrated in vacuo. The residue was purified by column chromatography (SiO2, Ethyl acetate /Petroleum = 63 /37) to afford the desired product.
LCMS: MS (ESI) m/z 493.2 [M+H] +.
Step B: 3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzoic acid
A mixture of methyl 3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzoate (327 mg, 663.81 μmol) , LiOH·H2O (279 mg, 6.64 mmol) in THF (4 mL) , H2O (1
mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 60 ℃ for 16 hr. The mixture was added 1M HCl to pH =3~4, and diluted with DCM (20 mL) and washed with H2O (3 × 20 mL) , brine (10 mL) , dried over Na2SO4, filtered and concentrated in vacuo to afford the desired product.
LCMS: MS (ESI) m/z 479.2 [M+H] +.
Step C: N- (2, 6-dioxo-3-piperidyl) -3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] benzamide
To a solution of 3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzoic acid (356 mg, 743.86 μmol) , 3-aminopiperidine-2, 6-dione (147 mg, 892.64 μmol, HCl) in DMAc (5 mL) was added HATU (425 mg, 1.12 mmol) and DIPEA (389 μL, 2.23 mmol) . The mixture was stirred at 25 ℃ for 3hr. The mixture was poured into H2O (30 mL) and filtered, the filtered cake was dissolved with DCM (30 mL) to dried with anhydrous Na2SO4, filtered and the filter was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 589.3 [M+H] +.
Step D: 3- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-
3-piperidyl) benzamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -3- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzamide (437 mg, 742.32 μmol) in DMF (2 mL) were added 4- (4-pyridyl) pyridine (1.2 mg, 7.42 μmol) and hypoboric acid (200 mg, 2.23 mmol) . The mixture was stirred at 25 ℃ for 5min. Upon completion, the mixture was concentrated in vacuo. The residue was purified by reversed-phase HPLC (column: C18 100 × 40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0 %-20 %B over 5.5 min) , after freeze-drying to afford the desired product.
LCMS: MS (ESI) m/z 559.3 [M+H] +.
Step E: N- (2, 6-dioxo-3-piperidyl) m- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} benzamide
To a solution of 3- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-3-piperidyl) benzamide (100 mg, 178.98 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (68.99 mg, 178.98 μmol) in n-BuOH (1 mL) was added HOAc (102.46 μL, 1.79 mmol) . The mixture was stirred at 100 ℃ for 16 hr. Upon completion, the mixture was concentrated in vacuo. The crude product was purified by reversed-phase HPLC (column: C18 100 × 40mm; mobile phase: [water (TFA) -ACN] ; gradient: 6%-36%B over 8 min) , after freeze-drying to afford the desired product.
LCMS: MS (ESI) m/z 880.9 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.88 (s, 1H) , 10.20 -10.00 (m, 1H) , 9.57 -9.38 (m, 1H) , 8.85 (s, 1H) , 8.71 (d, J = 8.4 Hz, 1H) , 8.01 (s, 1H) , 7.76 (d, J = 8.0 Hz, 1H) , 7.62 (dd, J = 8.4, 13.2 Hz, 3H) , 7.46 (s, 1H) , 7.38 -7.29 (m, 2H) , 7.19 (d, J = 7.6 Hz, 1H) , 7.01 (d, J = 9.2 Hz, 2H) , 5.41 -5.25 (m, 2H) , 4.83 -4.72 (m, 1H) , 4.48 (d, J = 4.8 Hz, 2H) , 3.82 (d, J = 12.4 Hz, 4H) , 3.24 (m, 8H) , 2.96 (m, 2H) , 2.80 (d, J = 11.6 Hz, 1H) , 2.57 (d, J = 3.6 Hz, 1H) , 2.26 -2.08 (m, 2H) , 2.01 -1.84 (m, 7H) , 1.76 -1.61 (m, 5H) , 1.57 (m, 3H) , 1.49 (m, 2H) , 1.21 (m, 2H) .
Example 196:
N- (2, 6-dioxo-3-piperidyl) -4- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -2-pyridinecarboxamide (Cpd-385)
Scheme:
Step A: methyl 4- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] pyridine-
2-carboxylate
To a solution of 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane (350 mg, 1.02 mmol) and methyl 4-bromopyridine-2-carboxylate (263.42 mg, 1.22 mmol) in dioxane (10 mL) was added K3PO4 (647.06 mg, 3.05 mmol) Pd2 (dba) 3 (93.05 mg, 101.61 μmol) and RuPhos (23.71 mg, 50.81 μmol) . The mixture was stirred at 100 ℃ for 6hr. The mixture was poured into water (50 mL) and extracted with EtOAC (100 mL × 3) . The combined organic layers were dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by flash silica gel chromatography (8 gSilica Flash Column, Eluent of 0~3%MeOH/DCM gradient @30 mL/min) to afford the desired product.
LCMS MS (ESI) m/z = 480.2 [M+H] +.
Step B: 4- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] pyridine-2-
carboxylic acid
To a solution of methyl 4- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] pyridine-2-carboxylate (200 mg, 333.63 μmol) in H2O (0.1 mL) and DMAC (3 mL) was added LiOH. H2O (70.00 mg, 1.67 mmol) . The mixture was stirred at 60 ℃ for 16 hr. The reaction mixture was adjusted to pH = 6 with 12 M HCl, the reaction mixture was filtered, collected solids. The solid was dissolved in DCM, and dried over Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS MS (ESI) m/z = 466.2 [M+H] +.
Step C: N- (2, 6-dioxo-3-piperidyl) -4- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-
azaspiro [4.5] decan-8-yl] pyridine-2-carboxamide
To a solution of 4- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] pyridine-2-carboxylic acid (200 mg, 429.61 μmol) and 3-aminopiperidine-2, 6-dione (84.85 mg, 515.53 μmol, HCl) in DMAC (2 mL) was added HATU (245.02 mg, 644.41 μmol) and DIPEA (555.22 mg, 4.30 mmol, 748.28 μL) . The mixture was stirred at 20 ℃ for 2 hr. The mixture was poured into water (30 mL) . The reaction mixture was filtered and collected solids. The solid was dissolved in DCM, and dried over Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS MS (ESI) m/z = 576.4 [M+H] +.
Step D: 4- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -N- (2, 6-dioxo-3-
piperidyl) pyridine-2-carboxamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -4- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] pyridine-2-carboxamide (300 mg, 521.14 μmol) in DMF (3 mL) were added hypoboric acid (140.16 mg, 1.56 mmol) and 4- (4-pyridyl) pyridine (813.92 μg, 5.21 μmol) . The mixture was stirred at 20 ℃ for 5 min. The crude product was purified by reversed-phase column (C18, MeOH in H2O = 23%, 0.1%TFA) , then dried by lyophilization to afford the desired product.
LCMS MS (ESI) m/z = 546.4
1H NMR (400 MHz, DMSO-d6) δ 11.00 (s, 1H) , 10.44 (s, 1H) , 9.64 (br d, J = 8.4 Hz, 1H) , 8.17 (d, J = 7.2 Hz, 1H) , 7.75 (s, 1H) , 7.31 -7.19 (m, 3H) , 7.10 (d, J = 9.2 Hz, 1H) , 7.15 -7.04 (m, 1H) , 4.91 –4.80 (m, 1H) , 4.66 -4.14 (m, 2H) , 3.86 (br s, 1H) , 3.80 -3.50 (m, 7H) , 3.22 -2.94 (m, 3H) , 2.92 -2.72 (m, 1H) , 2.47 -2.34 (m, 1H) , 2.24 -1.98 (m, 4H) , 1.96 -1.83 (m, 1H) , 1.80 -1.65 (m, 3H) , 1.63 -1.47 (m, 4H)
Step E: N- (2, 6-dioxo-3-piperidyl) -4- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -2-pyridinecarboxamide
To a solution of 4- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -N- (2, 6-dioxo-3-piperidyl) pyridine-2-carboxamide (140 mg, 212.22 μmol, TFA) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50.00 mg, 129.72 μmol) in n-BuOH (3 mL) was added AcOH (77.90 mg, 1.30 mmol, 74.26 μL) . The mixture was stirred at 100 ℃ for 16 hr. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: C18 150×30mm; mobile phase: [water (TFA) -ACN] ; gradient: 3%-43%B over 9 min) to afford the desired product.
LCMS MS (ESI) m/z = 867.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 11.00 (s, 1H) , 10.19 -9.90 (m, 2H) , 9.56 (s, 1H) , 8.84 (s, 1H) , 8.17 (d, J = 7.2 Hz, 1H) , 7.99 (t, J = 8.0 Hz, 1H) , 7.74 (d, J = 8.0 Hz, 2H) , 7.65 -7.56 (m, 3H) , 7.26 (br d, J = 5.2 Hz, 1H) , 6.99 (d, J = 9.6 Hz, 2H) , 5.45 -5.18 (m, 2H) , 4.94 -4.79 (m, 1H) , 4.47 (m, 2H) , 3.86 -3.57 (m, 4H) , 3.39 -3.27 (m, 6H) , 3.16 (s, 2H) , 3.01 -2.91 (m, 2H) , 2.90 -2.77 (m, 1H) , 2.64 -2.54 (m, 1H) , 2.25 -2.11 (m, 4H) , 2.08 -2.01 (m, 2H) , 1.99 -1.84 (m, 2H) , 1.76 -1.65 (m, 4H) , 1.63 -1.50 (m, 7H)
Example 197:
N- (2, 6-dioxo-3-piperidyl) 3- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -4-fluorobenzamide (Cpd-386)
Scheme:
Step A: methyl 4-fluoro-3- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-
yl] benzoate
To a solution of 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane (330 mg, 958.05 μmol) and methyl 3-bromo-4-fluoro-benzoate (334.89 mg, 1.44 mmol) in dioxane (10 mL) was added K3PO4 (610.10 mg, 2.87 mmol) , Pd2 (dba) 3 (87.73 mg, 95.80 μmol) and RuPhos (22.35 mg, 47.90 μmol) . The mixture was stirred at 100 ℃ for 6 hr under N2. The mixture was poured into water (20 mL) and extracted with DCM (100 mL × 3) . The combined organic layers were dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by flash silica gel chromatography (8 gSilica Flash Column, Eluent of 0~2%MeOH/DCM gradient @30 mL/min) to afford the desired product.
LCMS MS (ESI) m/z = 497.2 [M+H] +.
Step B: 4-fluoro-3- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] benzoic
acid
To a solution of methyl 4-fluoro-3- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] benzoate (170 mg, 273.88 μmol) in H2O (0.1 mL) and DMAC (3 mL) was added LiOH. H2O
(57.46 mg, 1.37 mmol) . The mixture was stirred at 60 ℃ for 16 hr. The reaction mixture was concentrated under reduced pressure and lyophilized directly to afford the desired product.
LCMS MS (ESI) m/z = 483.3 [M+H] +.
Step C: N- (2, 6-dioxo-3-piperidyl) -4- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-
azaspiro [4.5] decan-8-yl] pyridine-2-carboxamide
To a solution of 4-fluoro-3- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] benzoic acid (200 mg, 414.47 μmol) and 3-aminopiperidine-2, 6-dione (81.86 mg, 497.36 μmol, HCl) in DMAc (5 mL) was added HATU (236.39 mg, 621.70 μmol) and DIPEA (535.66 mg, 4.14 mmol, 721.91 μL) . The mixture was stirred at 20 ℃ for 2 hr. The mixture was poured into water (30 mL) . The reaction mixture was filtered and collected solids. The solids was dissolved in DCM, and dried over Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS MS (ESI) m/z = 593.1 [M+H] +.
Step D: 3- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -N- (2, 6-dioxo-3-
piperidyl) -4-fluoro-benzamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -4-fluoro-3- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] benzamide (300 mg, 506.19 μmol) in DMF (3 mL) were added hypoboric acid (136.14 mg, 1.52 mmol) and 4- (4-pyridyl) pyridine (790.57 μg, 5.06 μmol) . The mixture was stirred at 20 ℃ for 5 min. The crude product was purified by reversed-phase column (C18, MeOH in H2O = 23%, 0.1%TFA) , then dried by lyophilization to afford the desired product.
LCMS MS (ESI) m/z = 563.4
1H NMR (400 MHz, DMSO-d6) δ 10.88 (s, 1H) , 8.78 (d, J = 8.4 Hz, 1H) , 7.55 (d, J = 8.4 Hz, 1H) , 7.52 -7.48 (m, 1H) , 7.28 -7.18 (m, 3H) , 7.11 (d, J = 9.2 Hz, 2H) , 4.86 -4.65 (m, 1H) , 3.87 (d, J = 12.0 Hz, 2H) , 3.78 -3.68 (m, 1H) , 3.62 (d, J = 9.6 Hz, 2H) , 3.16 (d, J = 7.2 Hz, 2H) , 3.07 -2.94 (m, 6H) , 2.89 -2.72 (m, 1H) , 2.59 -2.52 (m, 1H) , 2.20 -2.05 (m, 3H) , 2.02 -1.83 (m, 2H) , 1.79 -1.63 (m, 4H) , 1.62 -1.51 (m, 3H)
Step E: N- (2, 6-dioxo-3-piperidyl) 3- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -4-fluorobenzamide
To a solution of 3- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -N- (2, 6-dioxo-3-piperidyl) -4-fluoro-benzamide (100 mg, 147.78 μmol, TFA) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) in n-BuOH (3 mL) were added AcOH (77.90 mg, 1.30 mmol, 74.26 μL) . The mixture was stirred at 100 ℃ for 3 h. The reaction
mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: C18 150×30 mm; mobile phase: [water (TFA) -ACN] ; gradient: 12%-52%B over 9 min) to afford the desired product.
LCMS MS (ESI) m/z = 884.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.88 (s, 1H) , 10.11 (TFA, 1H) , 9.69 (s, 1H) , 8.84 (s, 1H) , 8.76 (d, J = 8.4 Hz, 1H) , 8.07 -7.95 (m, 1H) , 7.75 (d, J =8.0 Hz, 1H) , 7.65 -7.58 (m, 3H) , 7.55 (d, J = 8.4 Hz, 1H) , 7.50 (d, J = 4.0 Hz, 1H) , 7.29 -7.21 (m, 1H) , 7.00 (d, J = 9.2 Hz, 2H) , 5.30 (m, 2H) , 4.86 -4.65 (m, 1H) , 4.48 (m, 2H) , 3.88 -3.72 (m, 2H) , 3.69 -3.60 (m, 4H) , 3.16 (m, 2H) , 3.09 -2.90 (m, 6H) , 2.86 -2.74 (m, 1H) , 2.57 (br s, 1H) , 2.49 -2.43 (m, 1H) , 2.25 -2.21 (m, 4H) , 2.04 -1.82 (m, 3H) , 1.79 -1.58 (m, 8H) , 1.56 (m, 3H)
19F NMR (400 MHz, DMSO-d6) δ = -74.342, -118.319
Example 198:
N- (2, 6-dioxo-3-piperidyl) -6- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -3-pyridazinecarboxamide (Cpd-387)
Scheme:
Step A: tert-butyl 8- (6-methoxycarbonylpyridazin-3-yl) -2, 8-diazaspiro [4.5] decane-2-
carboxylate
To a solution of methyl 6-chloropyridazine-3-carboxylate (200.00 mg, 1.16 mmol) , tert-butyl 2, 8-diazaspiro [4.5] decane-2-carboxylate (278.54 mg, 1.16 mmol) in DCM (5 mL) was added DIPEA (179.74 mg, 1.39 mmol) . The mixture was stirred at 25 ℃ for 16hr. The mixture was added H2O (10 mL) , extracted with DCM (10 mL × 3) . The combined organic layers were washed with brine (10 mL × 3) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~10 %Methanol/Dichloromethane gradient @25 mL/min) and the combined desired fractions was concentrated under reduced pressure to afford the desired product.
LCMS MS (ESI) m/z 377.3 [M+H] +.
1H NMR (400MHz, CDCl3) δ 7.88 (d, J = 9.6 Hz, 1H) , 6.89 (br d, J = 9.6 Hz, 1H) , 4.00 (s, 4H) , 3.85 (br s, 1H) , 3.77 (br s, 1H) , 3.64 (br s, 1H) , 3.51 -3.38 (m, 2H) , 3.33 (br s, 1H) , 3.24 (br s, 1H) , , 1.80 (t, J = 7.2 Hz, 2H) , 1.75 -1.67 (m, 4H) , 1.48 (s, 9H)
Step B: methyl 6- (2, 8-diazaspiro [4.5] decan-8-yl) pyridazine-3-carboxylate
To a solution of tert-butyl 8- (6-methoxycarbonylpyridazin-3-yl) -2, 8-diazaspiro [4.5] decane-2-carboxylate (300 mg, 796.9 μmol) in DCM (2.0 mL) was added HCl/dioxane (4 M, 4.0 mL) . The mixture was stirred at 20 ℃ for 15hr. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was diluted with saturated Na2CO3 (5.0 mL, aq. ) and extracted with DCM/MeOH (10 mL ×5) . The combined organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 277.3 [M+H] +.
Step C: methyl 6- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-
yl] pyridazine-3-carboxylate
To a solution of methyl 6- (2, 8-diazaspiro [4.5] decan-8-yl) pyridazine-3-carboxylate (200 mg, 723.7 μmol) and 1- (4-nitrophenyl) piperidin-4-one (175 mg, 796.1 μmol) in DCM (20.0 mL) was added TEA (366.18 mg, 3.62 mmol) at 20 ℃ under nitrogen atmosphere . The mixture was stirred at 20 ℃ for 15 hr. Then NaBH (OAc) 3 (307 mg, 1.45 mmol) was added to the reaction mixture and stirred for 0.5hr. The reaction mixture was quenched by addition NaHCO3 (15.0mL) , extracted with DCM (10 mL ×3) . The combined organic layers were washed with brine (15 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 0~5%MeOH/DCM gradient @20 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 481.3 [M+H] +.
Step D: 1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] pyridazine-3-
carboxylic acid
To a solution of methyl 6- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] pyridazine-3-carboxylate (210 mg, 436.99 μmol) in MeOH (2.0 mL) , THF (2.0 mL) and water (1 mL) was added LiOH. H2O (92 mg, 2.18 mmol) . The mixture was stirred at 20 ℃ for 15hr. The reaction was acidified with 2 M HCl (aq) to pH=4, the reaction mixture was concentrated to remove organic solvent, yellow solid was precipitated. The suspension was diluted with water (5.0 mL) , filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 467.2 [M+H] +.
Step E: N- (2, 6-dioxo-3-piperidyl) -6- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-
diazaspiro [4.5] decan-8-yl] pyridazine-3-carboxamide
To a solution of 6- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] pyridazine-3-carboxylic acid (90 mg, 192.9 μmol) and 3-aminopiperidine-2, 6-dione; hydrochloride (38 mg, 231.5 μmol) in DMF (1.0 mL) were added DIPEA (125 mg, 964.5 μmol) and HATU (110 mg, 289.3 μmol) . The mixture was stirred at 20 ℃ for 1hr. The reaction mixture was poured into water (5.0 mL) , yellow solid was precipitated, filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 577.1 [M+H] +.
Step F: 6- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -N- (2, 6-dioxo-3-
piperidyl) pyridazine-3-carboxamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -6- [2- [1- (4-nitrophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] pyridazine-3-carboxamide (100 mg, 173.4 μmol) in DMF (1.0 mL) were added hypoboric acid (50 mg, 557.7 μmol) and 4- (4-pyridyl) pyridine (0.3 mg, 1.73 μmol) . The mixture was stirred at 20 ℃ for 5 min. The reaction mixture was diluted with ethyl acetate (5.0 mL) , yellow solid was precipitated, filtered and the filtered cake was dried in vacuum to afford the desired product.
LCMS: MS (ESI) m/z 547.3 [M+H] +.
Step G: N- (2, 6-dioxo-3-piperidyl) -6- {2- [1- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -4-piperidyl] -2, 8-diaza-8-spiro [4.5] decyl} -3-pyridazinecarboxamide
To a solution of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (45 mg, 116.7 μmol) and 6- [2- [1- (4-aminophenyl) -4-piperidyl] -2, 8-diazaspiro [4.5] decan-8-yl] -N- (2, 6-dioxo-3-piperidyl) pyridazine-3-carboxamide (99 mg, 181.1 μmol) in n-BuOH (1.5 mL) was added HOAc (70 mg, 1.17 mmol) . The mixture was stirred at 100 ℃ for 2hr. The reaction mixture was concentrated in vacuum. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 5%-35%B over 8 min) , after freeze-drying to afford the desired product.
LCMS: MS (ESI) m/z 868.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ10.87 (s, 1H) , 10.09 (br s, 1H) , 9.80 (br s, 1H) , 9.09 (d, J = 8.4 Hz, 1H) , 8.84 (s, 1H) , 8.01 (t, J = 7.6 Hz, 1H) , 7.87 (d, J = 9.6 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.59 (d, J = 8.0 Hz, 3H) , 7.43 (d, J = 9.6 Hz, 1H) , 6.98 (d, J = 9.2 Hz, 2H) , 5.42 -5.25 (m, 2H) , 4.85 -4.74 (m, 1H) , 4.54 -4.41 (m, 2H) , 3.86 -3.77 (m, 6H) , 3.43 -3.23 (m, 2H) , 3.12 -3.04 (m, 1H) , 2.85 -2.76 (m, 1H) , 2.69 (br t, J = 11.6 Hz, 2H) , 2.55 (br s, 1H) , 2.25 -2.09 (m, 4H) , 2.08 -1.95 (m, 4H) , 1.94 -1.80 (m, 2H) , 1.79 -1.60 (m, 8H) , 1.56 (m, 3H) .
Example 199:
N- (2, 6-dioxo-3-piperidyl) p- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} benzamide (Cpd-388)
Scheme:
Step A: methyl 4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] benzoate
To a solution of 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (400 mg, 1.12 mmol) , methyl 4-bromobenzoate (263.95 mg, 1.23 mmol) , RuPhos (26.03 mg, 55.79 μmol) , Cs2CO3 (1.09 g, 3.35 mmol) in dioxane (5 mL) was added Pd2 (dba) 3 (102.18 mg, 111.58 μmol) and the mixture was stirred at 100 ℃ for 16 hr under N2. The resulting mixture was extracted with ethyl acetate (60 mL × 3) . The combined organic phase was washed with water (50 mL) , brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was triturated with ethyl acetate (15mL) at 20 ℃ for 30 min, filtered to afford the desired product.
LCMS: MS (ESI) m/z 493.3 [M+H] +
Step B: 4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzoic acid
To a solution of methyl 4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzoate (500 mg, 1.02 mmol) in H2O (1.5 mL) and DMAC (5 mL) was added NaOH (202.99 mg, 5.08 mmol) and the mixture was stirred at 60 ℃ for 16 hr. The resulting mixture was concentrated under reduced pressure to give a residue. The residue was added H2O (15 mL) , adjusted to pH = 3 with aq. HCl (12 M) , filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 479.3 [M+H] +
Step C: N- (2, 6-dioxo-3-piperidyl) -4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] benzamide
To a solution of 4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzoic acid (150 mg, 313.43 μmol) , 3-aminopiperidine-2, 6-dione; hydrochloride (61.90 mg, 376.11 μmol) , DIPEA (121.52 mg, 940.28 μmol) in DMAC (5 mL) was added HATU (178.76 mg, 470.14 μmol) and the mixture was stirred at 20 ℃ for 16 hr, then filtered to afford the desired product.
LCMS: MS (ESI) m/z 589.3 [M+H] +
Step D: 4- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-
3-piperidyl) benzamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzamide (180 mg, 305.76 μmol) , 4- (4-pyridyl) pyridine (9.55 mg, 61.15 μmol) in DMAC (5 mL) was added hypoboric acid (82.23 mg, 917.28 μmol) and the mixture was stirred at 20℃ for 1 hr. The resulting mixture was extracted with ethyl acetate (60 mL × 3) . The combined organic phase was washed with water (50 mL) , brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue
was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-22%B over 7 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 559.3 [M+H] +
Step E: N- (2, 6-dioxo-3-piperidyl) p- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} benzamide
To a solution of 4- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-3-piperidyl) benzamide (100 mg, 178.98 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (68.99 mg, 178.98 μmol) in n-BuOH (2 mL) was added AcOH (53.74 mg, 894.91 μmol) and the mixture was stirred at 100 ℃ for 1 hr. The resulting mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 15%-45%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 880.5 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ 10.83 (s, 1H) , 10.10 (TFA, 1H) , 9.50 (s, 1H) , 8.85 (s, 1H) , 8.45 (d, J = 8.4 Hz, 1H) , 7.93-8.06 (m, 1H) , 7.75 (d, J = 9.2 Hz, 3H) , 7.52-7.67 (m, 3H) , 6.99 (t, J = 9.2 Hz, 4H) , 5.24-5.40 (m, 2H) , 4.67-4.80 (m, 2H) , 4.44-4.48 (m, 2H) , 3.81 (d, J = 12.4 Hz, 2H) , 3.62 (d, J = 11.2 Hz, 2H) , 3.14-3.34 (m, 7H) , 2.96 (t, J = 12.0 Hz, 2H) , 2.70-2.87 (m, 1H) , 2.53-2.59 (m, 1H) , 1.83-2.24 (m, 9H) , 1.52-1.76 (m, 8H) , 1.37-1.47 (m, 2H) , 1.13-1.27 (m, 2H) .
Example 200:
N- (2, 6-dioxo-3-piperidyl) 3- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -5-fluorobenzamide (Cpd-389)
Scheme:
Step A: methyl 3-fluoro-5- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-
yl] benzoate
To a solution of 3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decane (350 mg, 1.02 mmol) , methyl 3-bromo-5-fluoro-benzoate (284.15 mg, 1.22 mmol) , Pd2 (dba) 3 (93.05 mg, 101.61 μmol) , RuPhos (23.71 mg, 50.81 μmol) and K3PO4 (647.06 mg, 3.05 mmol) in dioxane (5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 3 h under N2 atmosphere. The mixture was poured into water (50 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers were washed with brine (50 mL × 2) , dried over Na2SO4, filtered and concentrated to give a residue. The reaction mixture was triturated with EtOAc (10 mL) for 1 h at 20 ℃, filtered to afford the desired product.
LCMS: MS (ESI) m/z 497.2 [M+H] +.
Step B: 3-fluoro-5- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] benzoic
acid
To a solution of methyl 3-fluoro-5- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] benzoate (245.7 mg, 494.79 μmol) in DMAC (3 mL) and H2O (0.1 mL) was added LiOH
(59.25 mg, 2.47 mmol) . The mixture was stirred at 60 ℃ for 2.5 h. The reaction mixture was concentrated under reduced pressure to give crude product. The mixture was poured into water (30 mL) and extracted with EtOAc (30 mL) . The reaction mixture were adjusted the pH = 3 by aq. HCl (3 M) . Then the mixture was filtered to afford the desired product.
LCMS: MS (ESI) m/z 483.3 [M+H] +.
Step C: N- (2, 6-dioxo-3-piperidyl) -3-fluoro-5- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-
azaspiro [4.5] decan-8-yl] benzamide
To a solution of 3-fluoro-5- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] benzoic acid (240 mg, 497.36 μmol) and 3-aminopiperidine-2, 6-dione (76.47 mg, 596.83 μmol) in DMAC (3 mL) was added HATU (945.56 mg, 2.49 mmol) and DIEA (642.81 mg, 4.97 mmol, 866.31 μL) . The mixture was stirred at 25 ℃ for 3 h. The mixture was poured into water (50 mL) and extracted with EtOAc (100 mL × 2) . The combined organic layers were washed with brine (50 mL × 2) , dried over Na2SO4, filtered and concentrated to afford the desired product.
LCMS: MS (ESI) m/z 593.3 [M+H] +.
Step D: 3- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -N- (2, 6-dioxo-3-
piperidyl) -5-fluoro-benzamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -3-fluoro-5- [3- [4- (4-nitrophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] benzamide (328.6 mg, 554.45 μmol) in DMF (2 mL) was added hypoboric acid (149.12 mg, 1.66 mmol) and 4- (4-pyridyl) pyridine (865.96 μg, 5.54 μmol) . The mixture stirred at 20 ℃ for 5 min. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by reversed-phase column (C18, MeOH in H2O = 40%, 0.1%TFA) , then dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 563.3 [M+H] +.
Step E: N- (2, 6-dioxo-3-piperidyl) 3- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -8-aza-8-spiro [4.5] decyl} -5-fluorobenzamide
To a solution of 3- [3- [4- (4-aminophenyl) piperazin-1-yl] -8-azaspiro [4.5] decan-8-yl] -N- (2, 6-dioxo-3-piperidyl) -5-fluoro-benzamide (105.34 mg, 155.67 μmol, TFA) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) in n-BuOH (2 mL) was added AcOH (77.90 mg, 1.30 mmol, 74.26 μL) . The mixture was stirred at 100 ℃ for 1 h. The reaction mixture was concentrated under reduced pressure to give crude product. The crude product was purified by prep-HPLC (column: C18 150×30mm; mobile phase: [Water (NH4HCO3) -MeCN] ; gradient: 43%-63%B over 9 min) to afford the desired product.
LCMS: MS (ESI) m/z 884.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.06 -9.99 (m, 1H) , 8.82 (s, 1H) , 8.76 (d, J = 8.4 Hz, 1H) , 8.01 (t, J = 8.0 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.59-7.55 (m, 3H) , 7.23 (s, 1H) , 6.97 -6.89 (m, 4H) , 5.36 -5.28 (m, 3H) , 4.79 -4.73 (m, 1H) , 4.45 (d, J = 4.8 Hz, 2H) , 3.08 (m, 6H) , 2.85 -2.76 (m, 1H) , 2.62 -2.52 (m, 6H) , 2.22 -2.06 (m, 3H) , 2.04 -1.81 (m, 6H) , 1.75 -1.68 (m, 1H) , 1.65 -1.47 (m, 12H) .
19F NMR (400 MHz, DMSO-d6) δ = -112.11
Example 201:
(S) -3- [6- (2- { [4- (p- { (R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] methyl} -7-aza-7-spiro [3.5] nonyl) -1-oxo-2-isoindolinyl] -2, 6-piperidinedione (Cpd-399)
Scheme:
Step A: tert-butyl 2- ( (4- (4-aminophenyl) piperazin-1-yl) methyl) -7-azaspiro [3.5] nonane-7-
carboxylate
To a solution of tert-butyl 2- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -7-azaspiro [3.5] nonane-7-carboxylate (2 g, 4.50 mmol) in MeOH (40 mL) was added Pd/C (wet) (500 mg, 469.84 μmol, 10%purity) under N2. The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H2 (15 psi) at 25 ℃ for 15 hours. The reaction mixture was diluted with MeOH 100 mL, washed with MeOH (100 mL ×3) . Then the mixture was filtered and the filter was concentrated. Upon completion, the mixture was concentrated in vacuo. The crude product was used for the next step without purification, after freeze-drying to afford the desired product.
LCMS: MS (ESI) m/z 415.7 [M+H] +.
1H NMR (400 MHz, CD3OD) δ 6.84 (d, J = 8.8 Hz, 2H) , 6.71 (d, J = 8.8 Hz, 2H) , 3.37 (d, J =5.0 Hz, 2H) , 3.30 -3.25 (m, 2H) , 3.04 -3.00 (m, 4H) , 2.63 -2.59 (m, 4H) , 2.57 -2.50 (m, 3H) , 2.08 -2.01 (m, 2H) , 1.63 -1.58 (m, 2H) , 1.56 -1.49 (m, 3H) , 1.47 (m, 3H) , 1.44 (s, 9H)
Step B: tert-butyl 2- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
yl] amino] phenyl] piperazin-1-yl] methyl] -7-azaspiro [3.5] nonane-7-carboxylate
To a solution of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (2.21 g, 4.58 mmol) and tert-butyl 2- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -7-azaspiro [3.5] nonane-7-carboxylate (1.9 g, 4.58 mmol) in n-BuOH (40 mL) was stirred at 100 ℃ for 1 h. The reaction mixture was diluted with MeOH 100 mL, washed with MeOH (100 mL ×3) . The reaction mixture was concentrated under reduced pressure to give crude product. The residue was purified by column chromatography (SiO2, Dichloromethane/Methanol=100/1 to 96/4) to afford the desired product.
LCMS: MS (ESI) m/z 736.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.16 -9.90 (m, 1H) , 8.88 -8.72 (m, 1H) , 8.00 (t, J = 7.2 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.57 (t, J = 7.2 Hz, 3H) , 6.88 (d, J = 8.8 Hz, 2H) , 5.51 -5.13 (m, 3H) , 4.44 (d, J = 3.6 Hz, 2H) , 3.25 (m, 2H) , 3.16 (m, 2H) , 3.04 (m, 3H) , 2.45 (m, 6H) , 2.37 (d, J = 6.2 Hz, 2H) , 2.25 -2.11 (m, 1H) , 2.09 -1.85 (m, 4H) , 1.75 -1.65 (m, 1H) , 1.55 (m, 3H) , 1.48 (m, 2H) , 1.38 (s, 9H)
Step C: (12Z, 16R) -5- [4- [4- (7-azaspiro [3.5] nonan-2-ylmethyl) piperazin-1-yl] anilino] -16-
hydroxy-16-methyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-
1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one
To a solution of tert-butyl 2- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-yl] amino] phenyl] piperazin-1-yl] methyl] -7-azaspiro [3.5] nonane-7-carboxylate (8.4 g, 11.41 mmol) in DCM (20 mL) was added TFA (15.35 g, 134.62 mmol) . The mixture was stirred at 25 ℃ for 16 hr. The mixture was adjusted to pH = 11 with aq. sat. Na2CO3, and extracted with DCM : MeOH (10: 1) (200 mL × 3) , the combined organic phase was dried with anhydrous Na2SO4, and then concentrated under reduce pressure to afford the desired product.
LCMS: MS (ESI) m/z 636.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.20 -9.89 (m, 1H) , 8.82 (s, 1H) , 8.10 -7.95 (m, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.57 (t, J = 8.0 Hz, 3H) , 6.89 (d, J = 9.6 Hz, 2H) , 5.47 -5.21 (m, 3H) , 4.64 -4.33 (m, 3H) , 3.05 (m, 4H) , 2.87 -2.79 (m, 2H) , 2.73 (t, J = 4.8 Hz, 2H) , 2.46 (m, 6H) , 2.37 (d, J = 6.8 Hz, 3H) , 2.21 -2.10 (m, 1H) , 2.07 -2.00 (m, 1H) , 1.96 -1.89 (m, 3H) , 1.75 -1.66 (m, 2H) , 1.61 (d, J = 4.4 Hz, 2H) , 1.56 (m, 3H) , 1.51 -1.47 (m, 3H) , 1.45 -1.33 (m, 4H)
Step D: methyl 2-formyl-5- [2- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-
yl] amino] phenyl] piperazin-1-yl] methyl] -7-azaspiro [3.5] nonan-7-yl] benzoate
To a solution of (12Z, 16R) -5- [4- [4- (7-azaspiro [3.5] nonan-2-ylmethyl) piperazin-1-yl] anilino] -16-hydroxy-16-methyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (5 g, 7.86 mmol) , methyl 5-fluoro-2-formyl-benzoate (1.86 g, 10.22 mmol) in DMAC (37 mL) was added DIPEA (1.02 g, 7.86 mmol, 1.37 mL) and H2O (100.03 mg, 5.55 mmol) . The mixture was stirred at 120 ℃ for 16 h. The reaction mixture was added H2O (100 mL) and extracted with DCM (300 mL × 3) . The combined organic phases were washed with brine (300 mL × 3) , dried with anhydrous Na2SO4, filtered and concentrated under reduced pressure.
The crude reaction mixture on notebook page ES22579-684 was combined to ES22579-685 for purification.
The residue was purified by flash silica gel chromatography (120 g Silica Flash Column, Eluent of 0~6%MeOH/DCM gradient @100 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 798.8 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.12 -10.02 (m, 1H) , 9.99 (s, 1H) , 8.90 -8.77 (m, 1H) , 8.08 -7.96 (m, 1H) , 7.75 (dd, J = 4.4, 8.4 Hz, 2H) , 7.58 (t, J = 7.6 Hz, 3H) , 7.20 -7.11 (m, 2H) , 6.90 (d, J = 9.2 Hz, 2H) , 5.43 -5.25 (m, 3H) , 4.54 -4.36 (m, 2H) , 3.86 (m, 3H) , 3.45 -3.39 (m, 2H) , 3.34 -3.34 (m, 1H) , 3.36 -3.33 (m, 1H) , 3.31 (s, 2H) , 3.07 (m, 4H) , 2.49 -2.46 (m, 4H) , 2.42 (d, J = 6.4 Hz, 2H) , 2.24 -2.10 (m, 1H) , 2.01 -2.00 (m, 1H) , 1.99 -1.90 (m, 4H) , 1.78 -1.69 (m, 1H) , 1.69 -1.62 (m, 2H) , 1.56 (m, 3H) , 1.55 -1.50 (m, 2H) , 1.50 -1.44 (m, 2H)
Step E: (S) -3- [6- (2- { [4- (p- { (R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] methyl} -7-aza-7-spiro [3.5] nonyl) -1-oxo-2-isoindolinyl] -2, 6-
piperidinedione
To a solution of methyl 2-formyl-5- [2- [ [4- [4- [ [ (12Z, 16R) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-yl] amino] phenyl] piperazin-1-yl] methyl] -7-azaspiro [3.5] nonan-7-yl] benzoate (500 mg, 626.61 μmol) , (3S) -3-aminopiperidine-2, 6-dione (134.07 mg, 814.60 μmol, HCl) in MeOH (5 mL) and THF (5 mL) was added HOAc (376.29 mg, 6.27 mmol) and NaBH3CN (118.13 mg, 1.88 mmol) at 0 ℃. The mixture was stirred at 20 ℃ for 4 h. The mixture was adjusted to pH=7 with aq. sat. NaHCO3, and extracted with DCM: MeOH (10: 1) (200 mL × 3) , the combined organic phase was dried with anhydrous Na2SO4, and then concentrated under reduce pressure to give crude
product. The residue was purified by prep-HPLC (column: C18 100×40 mm; mobile phase: [water (TFA) -ACN] ; gradient: 6%-36%B over 8 min) to afford the desired product.
LCMS: MS (ESI) m/z 878.6 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.98 (s, 1H) , 10.23 -9.93 (TFA, 1H) , 9.58 (s, 1H) , 8.84 (s, 1H) , 8.00 (t, J = 8.0 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.61 (dd, J = 8.4, 10.6 Hz, 3H) , 7.44 (d, J = 8.4 Hz, 1H) , 7.30 (d, J = 9.2 Hz, 1H) , 7.22 (s, 1H) , 6.98 (d, J = 9.2 Hz, 2H) , 5.30 (m, 2H) , 5.09 (dd, J = 4.8, 13.6 Hz, 1H) , 4.46 (m, 2H) , 4.34 (d, J = 16.8 Hz, 1H) , 4.20 (d, J = 16.8 Hz, 1H) , 3.77 (d, J = 12.8 Hz, 2H) , 3.49 (d, J = 12.0 Hz, 2H) , 3.35 -3.06 (m, 9H) , 3.01 -2.85 (m, 3H) , 2.81 -2.69 (m, 1H) , 2.59 (d, J = 16.0 Hz, 1H) , 2.38 (dd, J = 4.8, 12.8 Hz, 1H) , 2.19 (s, 1H) , 2.13 -1.90 (m, 5H) , 1.80 -1.69 (m, 3H) , 1.68 -1.60 (m, 4H) , 1.56 (m, 3H)
Example 202:
N- (2, 6-dioxo-3-piperidyl) 4- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-fluorobenzamide (Cpd-437)
Scheme:
Step A: methyl 2-fluoro-4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] benzoate
To a solution of 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (400 mg, 1.12 mmol) , methyl 4-bromo-2-fluoro-benzoate (286.03 mg, 1.23 mmol) , RuPhos (26.03 mg, 55.79 μmol) , Cs2CO3 (1.09 g, 3.35 mmol) in dioxane (5 mL) was added Pd2 (dba) 3 (102.18 mg, 111.58 μmol) and the mixture was stirred at 100 ℃ for 3hr under N2. The resulting mixture was added water (30 mL) , extracted with ethyl acetate (60 mL × 3) . The combined organic phase was washed with brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was triturated with ethyl acetate (15mL) at 20 ℃ for 30 min, filtered to afford the desired product.
LCMS: MS (ESI) m/z 511.4 [M+H] +
Step B: 2-fluoro-4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzoic
acid
To a solution of methyl 2-fluoro-4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzoate (540 mg, 1.06 mmol) in MeOH (2 mL) , THF (2 mL) and H2O (1 mL) was added
NaOH (211.50 mg, 5.29 mmol) and the mixture was stirred at 60 ℃ for 2 hr. The resulting mixture was concentrated under reduced pressure to give a residue. The residue was added water (20 mL) , adjusted to pH = 3 with aq. HCl (3 M) , filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 497.4 [M+H] +
Step C: N- (2, 6-dioxo-3-piperidyl) -2-fluoro-4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] benzamide
To a solution of 2-fluoro-4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzoic acid (450 mg, 906.21 μmol) , 3-aminopiperidine-2, 6-dione; hydrochloride (178.98 mg, 1.09 mmol) , DIPEA (585.61 mg, 4.53 mmol) in DMAc (3 mL) was added HATU (516.85 mg, 1.36 mmol) and the mixture was stirred at 25 ℃ for 2 hr, and filtered to afford the desired product.
LCMS: MS (ESI) m/z 607.6 [M+H] +
Step D: 4- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-
3-piperidyl) -2-fluoro-benzamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -2-fluoro-4- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] benzamide (500 mg, 824.15 μmol) , 4- (4-pyridyl) pyridine (25.74 mg, 164.83 μmol) in DMAc (3 mL) was added 4, 4’-hypoboric acid (221.66 mg, 2.47 mmol) and the mixture was stirred at 25 ℃ for 20 min. The resulting mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~12%Methanol : Dichloromethane ether gradient @35 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 577.1 [M+H] +
Step E: N- (2, 6-dioxo-3-piperidyl) 4- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -2-fluorobenzamide
A mixture of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) , 4- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-3-piperidyl) -2-fluoro-benzamide (74.81 mg, 129.72 μmol) , AcOH (7.79 mg, 129.72 μmol, 7.43 μL) in n-BuOH (3 mL) was stirred at 100 ℃ for 2 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 16%-46%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 898.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.84 (s, 1H) , 10.09 (TFA, 1H) , 9.30 (s, 1H) , 8.85 (s, 1H) , 8.07 -7.94 (m, 2H) , 7.75 (d, J = 7.6 Hz, 1H) , 7.68 -7.55 (m, 4H) , 7.00 (d, J = 8.8 Hz, 2H) , 6.84 -6.71 (m, 2H) , 5.45 -5.20 (m, 2H) , 4.79 -4.66 (m, 1H) , 4.53 -4.41 (m, 2H) , 3.81 (d, J = 12.0 Hz, 2H) , 3.62 (d, J = 11.6 Hz, 1H) , 3.33 (m, 5H) , 3.27 -3.15 (m, 4H) , 2.94 (t, J = 12.0 Hz, 2H) , 2.84 -2.71 (m, 1H) , 2.21 -2.06 (m, 2H) , 2.05 -1.91 (m, 5H) , 1.86 (d, J = 12.8 Hz, 2H) , 1.77 -1.49 (m, 9H) , 1.41 (m, 2H) , 1.19 (t, J = 12.4 Hz, 2H) .
19F NMR (400 MHz, DMSO-d6) δ = -110.646
Example 203:
3- (6- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperidyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2-isoindolinyl) -2, 6-piperidinedione (Cpd-418)
Scheme:
Step A: methyl 2-formyl-5- [9- [4- (4-nitrophenyl) -1-piperidyl] -3-azaspiro [5.5] undecan-3-
yl] benzoate
To a solution of 9- [4- (4-nitrophenyl) -1-piperidyl] -3-azaspiro [5.5] undecane (400 mg, 1.12 mmol) , methyl 5-fluoro-2-formyl-benzoate (407.62 mg, 2.24 mmol) in DMAC (4 mL) was added DIEA (433.84 mg, 3.36 mmol) and then the mixture was stirred at 100 ℃ for 3 hr. The reaction mixture was added water (60 mL) , extracted with ethyl acetate (60 mL × 3) . The combined organic phase was washed with water (50 mL) , brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~6%Methanol: Dichloromethane ether gradient @30 mL/min) to afford the desired product. LCMS: MS (ESI) m/z 520.2 [M+H] +
Step B: 3- [6- [9- [4- (4-nitrophenyl) -1-piperidyl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-
isoindolin-2-yl] piperidine-2, 6-dione
To a solution of methyl 2-formyl-5- [9- [4- (4-nitrophenyl) -1-piperidyl] -3-azaspiro [5.5] undecan-3-yl] benzoate (300 mg, 577.33 μmol) , 3-aminopiperidine-2, 6-dione (221.92 mg, 1.73 mmol) ,
AcOH (69.34 mg, 1.15 mmol) in MeOH (2 mL) and THF (2 mL) was added NaBH3CN (108.84 mg, 1.73 mmol) and then the mixture was stirred at 25 ℃ for 16hr. The resulting mixture was extracted with DCM (60 mL × 3) . The combined organic phase was washed with water (50 mL) , brine (50 mL) , dried over Na2SO4, filtered and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 600.4 [M+H] +
Step C: 3- [6- [9- [4- (4-aminophenyl) -1-piperidyl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-
isoindolin-2-yl] piperidine-2, 6-dione
To a solution of 3- [6- [9- [4- (4-nitrophenyl) -1-piperidyl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (200 mg, 333.49 μmol) in DMAC (3 mL) was added hypoboric acid (89.69 mg, 1.00 mmol) and 4- (4-pyridyl) pyridine (10.42 mg, 66.70 μmol) , then the mixture was stirred at 20 ℃ for 10 min. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (12 gSilica Flash Column, Eluent of 0~12%Methanol: Dichloromethane ether gradient @30 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 570.3 [M+H] +
Step D: 3- (6- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperidyl] -3-aza-3-spiro [5.5] undecyl} -1-oxo-2-isoindolinyl) -2, 6-
piperidinedione
A mixture of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (20.30 mg, 52.66 μmol) , 3- [6- [9- [4- (4-aminophenyl) -1-piperidyl] -3-azaspiro [5.5] undecan-3-yl] -1-oxo-isoindolin-2-yl] piperidine-2, 6-dione (30 mg, 52.66 μmol) , AcOH (31.62 mg, 526.56 μmol) in n-BuOH (3 mL) was stirred at 100 ℃ for 3 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 8%-38%B over 8 min) and dried by lyophilization to afford the desired product. LCMS: MS (ESI) m/z 891.7 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ 10.97 (s, 1H) , 10.21 (s, 1H) , 9.03 (s, 1H) , 8.89 (s, 1H) , 8.02 -7.95 (m, 1H) , 7.76 (d, J = 8.0 Hz, 1H) , 7.71 (d, J = 8.8 Hz, 2H) , 7.61 (d, J = 7.6 Hz, 1H) , 7.44 (d, J = 8.4 Hz, 1H) , 7.29 (d, J = 8.8 Hz, 1H) , 7.19 (d, J = 8.8 Hz, 3H) , 5.45 -5.20 (m, 2H) , 5.09 (dd, J = 5.2, 13.2 Hz, 1H) , 4.58 -4.42 (m, 2H) , 4.39 -4.14 (m, 2H) , 3.57 (d, J = 11.2 Hz, 1H) , 3.24 (m, 6H) , 3.18 -3.05 (m, 3H) , 2.99 -2.76 (m, 2H) , 2.60 (d, J = 16.8 Hz, 1H) , 2.47 -2.32 (m, 2H) , 2.20 (m, 1H) , 2.06 -1.96 (m, 4H) , 1.93 -1.80 (m, 6H) , 1.75 -1.61 (m, 5H) , 1.57 (m, 3H) , 1.48 (m, 2H) , 1.29 -1.14 (m, 2H) .
Example 204:
N- (2, 6-dioxo-3-piperidyl) -5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -3-fluoro-2-pyridinecarboxamide (Cpd-436)
Scheme:
Step A: Methyl 3-fluoro-5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] pyridine-2-carboxylate
A mixture of 9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecane (250 mg, 697.40 μmol, 1 eq) , methyl 5-bromo-3-fluoro-pyridine-2-carboxylate (490 mg, 2.09 mmol) , Pd2 (dba) 3 (63.8 mg, 69.74 μmol) , RuPhos (16.3 mg, 34.87 μmol μmol) and Cs2CO3 (681.7 mg, 2.09 mmol) in dioxane (6 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 ℃ for 13 hr under N2 atmosphere. Upon completion, the mixture was
concentrated in vacuo. The residue was purified by column chromatography (SiO2, Ethyl acetate /Petroleum = 1 /0) to afford the desired product.
LCMS: MS (ESI) m/z 512.3 [M+H] +.
Step B: 3-fluoro-5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-
yl] pyridine-2-carboxylic acid
A mixture of methyl 3-fluoro-5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridine-2-carboxylate (193 mg, 377.26 μmol) in DMAc (4 mL) , H2O (0.04 mL) was added LiOH·H2O (158.4 mg, 3.77 mmol) . The mixture was stirred at 60 ℃ for 3hr. The mixture was added 1M HCl to pH =3~4, filtered and concentrated in vacuo to afford the desired product.
LCMS: MS (ESI) m/z 498.3 [M+H] +.
Step C: N- (2, 6-dioxo-3-piperidyl) -3-fluoro-5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-
azaspiro [5.5] undecan-3-yl] pyridine-2-carboxamide
To a solution of3-fluoro-5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridine-2-carboxylic acid (187.8 mg, 377.26 μmol) , 3-aminopiperidine-2, 6-dione; hydrochloride (74.6 mg, 452.71 μmol) , in DMAc (4 mL) was added HATU (215.2 mg, 565.89 mmol) and DIPEA (146.3 mg, 1.13 m mmol) . The mixture was stirred at 25 ℃ for 1hr. The mixture was poured into H2O (6 mL) and filtered, the filtered cake was dissolved with EtOAc (3 × 20 mL) to dried with anhydrous Na2SO4, filtered and the filter was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 608.4 [M+H] +.
Step D: 5- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-
3-piperidyl) -3-fluoro-pyridine-2-carboxamide
To a solution of N- (2, 6-dioxo-3-piperidyl) -3-fluoro-5- [9- [4- (4-nitrophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] pyridine-2-carboxamide (229 mg, 376.85 μmol) in DMF (2 mL) were added 4- (4-pyridyl) pyridine (589 μg, 3.77 μmol) and hypoboric acid (101.4 mg, 1.13 mmol) . The mixture was stirred at 25 ℃ for 5 min. Upon completion, the mixture was concentrated in vacuo. The residue was purified by reversed-phase HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-30%B over 8 min) , after freeze-drying to afford the desired product.
LCMS: MS (ESI) m/z 578.3 [M+H] +.
Step E: N- (2, 6-dioxo-3-piperidyl) -5- {9- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-
2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -3-aza-3-spiro [5.5] undecyl} -3-fluoro-2-
pyridinecarboxamide
To a solution of 5- [9- [4- (4-aminophenyl) piperazin-1-yl] -3-azaspiro [5.5] undecan-3-yl] -N- (2, 6-dioxo-3-piperidyl) -3-fluoro-pyridine-2-carboxamide (35 mg, 60.59 μmol) , (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-9-one (26 mg, 60.59 μmol) in n-BuOH (1 mL) was added HOAc (34.68 μL, 605.86 μmol) . The mixture was stirred at 100 ℃ for 16 hr. Upon completion, the mixture was concentrated in vacuo. The crude product was purified by reversed-phase HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 14%-44%B over 8 min) , after freeze-drying to afford the desired product.
LCMS: MS (ESI) m/z 899.5 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.86 (s, 1H) , 10.26 -9.99 (m, 1H) , 9.41 -9.24 (m, 1H) , 8.85 (s, 1H) , 8.59 (d, J = 8.4 Hz, 1H) , 8.19 (s, 1H) , 8.07 -7.96 (m, 1H) , 7.76 (d, J = 8.4 Hz, 1H) , 7.70 -7.53 (m, 3H) , 7.24 (d, J = 15.2 Hz, 1H) , 7.01 (d, J = 9.2 Hz, 2H) , 5.43 -5.22 (m, 2H) , 4.78 -4.65 (m, 1H) , 4.48 (d, J = 4.4 Hz, 2H) , 3.92 -3.75 (m, 2H) , 3.62 (d, J = 11.2 Hz, 3H) , 3.33 -3.12 (m, 6H) , 3.01 -2.89 (m, 2H) , 2.87 -2.73 (m, 1H) , 2.55 (m, 2H) , 2.26 -2.10 (m, 2H) , 1.97 (d, J = 11.2 Hz, 7H) , 1.62 (m, 5H) , 1.57 (m, 3H) , 1.44 (m, 2H) , 1.20 (m, 2H)
Example 205:
1- [5- ( {9- [ (4- {4- [8-bromo-6- (2, 6-dichlorophenyl) -5-oxo-5, 6-dihydro-1, 3, 6-triaza-2-naphthylamino] -1-pyrazolyl} -1-piperidyl) methyl] -3-aza-3-spiro [5.5] undecyl} carbonyl) -2-chlorophenyl] -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-394)
Scheme:
Step A: N- (2, 6-dichlorophenyl) -4-methyl-2-methylsulfanyl-pyrimidine-5-carboxamide
To a solution of ethyl 4-methyl-2-methylsulfanyl-pyrimidine-5-carboxylate (3.00 g, 14.1 mmol) and 2, 6-dichloroaniline (3.43 g, 21.2 mmol) in toluene (30 mL) was added trimethylaluminum (2 M in toluene, 14.1 mL, 28.2 mmol) at 20 ℃ under N2. The mixture was stirred at 20 ℃ for 0.5 h and then allowed to warm to 110 ℃, and stirred at 110 ℃ for 16 h. The mixture was cooled down to 10 ℃, and quenched with water (50 mL) dropwise at 10~25 ℃. Lots of precipitate were formed. The mixture was diluted with EtOAc (50 mL) , and filtered. The filtrate was separated. The water layer was extracted with EtOAc (30 mL *2) . The organic layer was
concentrated to give crude (2.20 g) as a yellow solid. The crude was suspended in PE (20 mL) , and stirred at 20 ℃ for 1 h. The mixture was filtered, and the filter cake was dried under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 327.9 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.53 (s, 1H) , 8.73 (s, 1H) , 7.57-7.65 (m, 2H) , 7.43 (t, J = 6.4 Hz, 1H) , 2.61 (s, 3H) , 2.57 (s, 3H) .
Step B: 6- (2, 6-dichlorophenyl) -2-methylsulfanyl-pyrido [4, 3-d] pyrimidin-5-one
To a solution of N- (2, 6-dichlorophenyl) -4-methyl-2-methylsulfanyl-pyrimidine-5-carboxamide (1.00 g, 3.05 mmol) in DMF (10 mL) was added DMF-DMA (1.09 g, 9.14 mmol) under N2. The mixture was stirred at 100 ℃ for 5 h. The yellow solution was concentrated to give crude. The crude was purified by Combi-flash (silica gel, 10~80%EtOAc in PE) to afford the desired product.
LCMS: MS (ESI) m/z 337.8 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 9.44 (s, 1H) , 7.53-7.60 (m, 2H) , 7.46 (d, J = 8.8 Hz, 1H) , 7.29 (t, J = 7.6 Hz, 1H) , 6.70 (d, J = 7.6 Hz, 1H) , 2.71 (s, 3H) .
Step C: 8-bromo-6- (2, 6-dichlorophenyl) -2-methylsulfanyl-pyrido [4, 3-d] pyrimidin-5-one
To a solution of 6- (2, 6-dichlorophenyl) -2-methylsulfanyl-pyrido [4, 3-d] pyrimidin-5-one (200 mg, 591 μmol) in MeCN (6 mL) was added NBS (116 mg, 652 μmol) under N2. The mixture was stirred at 80 ℃ for 16 h. The solution was concentrated to give a residue. The residue was suspended in EtOH (1.5 mL) , and stirred at 20 ℃ for 1 h. The mixture was filtered, and the filter cake was washed with EtOH (1 mL) , dried under reduced pressure to afford the desired product. LCMS: MS (ESI) m/z 417.9 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 9.37 (s, 1H) , 7.58 (s, 1H) , 7.48-7.56 (m, 3H) , 7.43 (t, J = 7.2 Hz, 1H) , 2.74 (s, 3H) .
Step D: 8-bromo-6- (2, 6-dichlorophenyl) -2-methylsulfinyl-pyrido [4, 3-d] pyrimidin-5-one
To a solution of 8-bromo-6- (2, 6-dichlorophenyl) -2-methylsulfanyl-pyrido [4, 3-d] pyrimidin-5-one (590 mg, 1.41 mmol) in DCM (10 mL) was added m-CPBA (430 mg, 2.12 mmol, 85%purity) at 20 ℃ in batches. The mixture was stirred at 20 ℃ for 1 h. The reaction mixture was treated with saturated Na2SO3 aq. (10 mL) , and stirred at 20 ℃ for 0.5 h. The mixture was extracted with DCM (10 mL *3) . The organic layer was concentrated to give crude. The crude was purified by Combi-flash (silica gel, 40~100%EtOAc in PE) to afford the desired product. LCMS: MS (ESI) m/z 433.8 [M+H] +.
Step E: tert-butyl 9- [ [4- [4- [ [8-bromo-6- (2, 6-dichlorophenyl) -5-oxo-pyrido [4, 3-d] pyrimidin-
2-yl] amino] pyrazol-1-yl] -1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate
To the mixture of 8-bromo-6- (2, 6-dichlorophenyl) -2-methylsulfinyl-pyrido [4, 3-d] pyrimidin-5-one (90 mg, 208 μmol) and tert-butyl 9- [ [4- (4-aminopyrazol-1-yl) -1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate (179 mg, 416 μmol) in DCE (4 mL) was added AcOH (12 mg, 208 μmol) . The resulting mixture was stirred at 50 ℃ for 16 h under N2. The mixture was concentrated under reduced pressure to give crude product as a pink solid. The crude was purified by Combi-flash (silica gel, 4 g, 25 mL/min, 0~6%MeOH in DCM) . The fraction containing the major component was combined and concentrated to afford the desired product. LCMS: MS (ESI) m/z 801.1 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 9.24 (s, 1H) , 8.51 (s, 1H) , 8.30 (s, 1H) , 7.74 (s, 1H) , 7.53 (d, J =8.8 Hz, 3H) , 7.38-7.45 (m, 1H) , 4.21 (s, 1H) , 3.50 (s, 2H) , 3.32-3.41 (m, 5H) , 3.07 (s, 2H) , 2.13-2.35 (m, 8H) , 1.61-1.73 (m, 4H) , 1.46 (s, 9H) , 1.27-1.34 (m, 2H) , 1.12 (d, J = 9.4 Hz, 4H) .
Step F: 2- [ [1- [1- (3-azaspiro [5.5] undecan-9-ylmethyl) -4-piperidyl] pyrazol-4-yl] amino] -8-
bromo-6- (2, 6-dichlorophenyl) pyrido [4, 3-d] pyrimidin-5-one
To the mixture of tert-butyl 9- [ [4- [4- [ [8-bromo-6- (2, 6-dichlorophenyl) -5-oxo-pyrido [4, 3-d] pyrimidin-2-yl] amino] pyrazol-1-yl] -1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate (130 mg, 162 μmol) in dioxane (1 mL) was added HCl/dioxane (4 M, 3 mL) and stirred at 20 ℃ for 16 h under N2. The mixture was concentrated under reduced pressure to afford the desired product.
1H NMR (400 MHz, DMSO) δ 10.76 (s, 1H) , 9.10 (s, 1H) , 8.80 (s, 1H) , 8.52 (s, 1H) , 8.40 (s, 1H) , 7.72-7.77 (m, 2H) , 7.57-7.65 (m, 1H) , 5.76 (s, 1H) , 4.79 (s, 7H) , 3.05-3.16 (m, 2H) , 2.87-3.05 (m, 8H) , 2.17-2.28 (m, 2H) , 1.59-1.77 (m, 8H) , 1.42-1.52 (m, 2H) , 1.09-1.20 (m, 5H) .
Step G: tert-butyl 4- (4-nitropyrazol-1-yl) piperidine-1-carboxylate
To the mixture of tert-butyl 4-hydroxypiperidine-1-carboxylate (2 g, 9.94 mmol) and 4-nitro-1H-pyrazole (1.12 g, 9.94 mmol) in THF (60 mL) was added PPh3 (3.13 g, 11.9 mmol) . The mixture was cooled down to 0~5 ℃ with an ice water. DIAD (2.41 g, 11.9 mmol) was added dropwise to the above mixture at 0~5 ℃. The resulting mixture was stirred at 20 ℃ for 16 h under N2. The mixture was diluted with water (30 mL) , and extracted with ethyl acetate (20 mL ×3) , combined organic phase was dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give crude as a yellow oil. The crude was purified by combi-flash (silica gel, 40 g, 45 mL/min, 15~30%ethyl acetate in petroleum ether) , the fraction containing the major component was combined and concentrated to afford the desired product. LCMS: MS (ESI) m/z 951.0 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 8.96 (s, 1H) , 8.88 (s, 1H) , 8.28 (s, 1H) , 4.74-4.81 (m, 1H) , 4.40-4.51 (m, 1H) , 2.89 (s, 1H) , 2.00-2.08 (m, 2H) , 1.81 (d, J = 12.2, 4.4 Hz, 2H) , 1.41 (s, 9H) , 1.18 (d, J = 6.0 Hz, 7H) .
Step H: 4- (4-nitropyrazol-1-yl) piperidine
To the mixture of tert-butyl 4- (4-nitropyrazol-1-yl) piperidine-1-carboxylate (2.90 g, 9.79 mmol) in DCM (15 mL) was added HCl/dioxane (4 M, 10 mL) . Then the mixture was stirred at 20 ℃for 16 h under N2. The mixture was filtered and the filter cake was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 197.1 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 8.95 (s, 1H) , 8.33 (s, 1H) , 4.54-4.66 (m, 1H) , 3.28-3.42 (m, 2H) , 3.04 (d, J = 8.2 Hz, 2H) , 2.09-2.27 (m, 4H) .
Step I: tert-butyl 9- [ [4- (4-nitropyrazol-1-yl) -1-piperidyl] methyl] -3-azaspiro [5.5] undecane-
3-carboxylate
To the mixture of 4- (4-nitropyrazol-1-yl) piperidine (500 mg, 2.55 mmol) and tert-butyl 9-formyl-3-azaspiro [5.5] undecane-3-carboxylate (717 mg, 2.55 mmol) in DCM (7 mL) was added acetic acid (76 mg, 1.27 mmol) . The mixture was stirred at 20 ℃ for 48 h under N2.
NaBH (OAc) 3 (1.62 g, 7.65 mmol) was added to the above mixture and stirred at 20 ℃ for 2 h under N2. The mixture was diluted with water (~10 mL) and extracted with DCM (8 mL ×3) , combined organic phase was dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give crude (1.90 g) as a pink oil. The crude was purified by Combi-flash (silica gel, 12 g, 45 mL/min, 40~70%ethyl acetate in petroleum ether) to afford the desired product.
1H NMR (400 MHz, DMSO-d6) δ 8.92 (s, 1H) , 8.27 (s, 1H) , 4.16-4.29 (m, 1H) , 3.22-3.34 (m, 5H) , 2.83-2.94 (m, 2H) , 2.13 (d, J = 7.0 Hz, 2H) , 1.91-2.03 (m, 6H) , 1.59-1.68 (m, 2H) , 1.50-1.58 (m, 2H) , 1.42-1.47 (m, 1H) , 1.38 (s, 9H) , 1.19-1.24 (m, 3H) , 0.98-1.08 (m, 4H) .
Step J: tert-butyl 9- [ [4- (4-aminopyrazol-1-yl) -1-piperidyl] methyl] -3-
azaspiro [5.5] undecane-3-carboxylate
Wet Pd/C (50 mg, 46.98 μmol, 10%purity) was weigh to the hydrogen cylinder under Ar, then MeOH (10 mL) was slowly added to the hydrogen cylinder under Ar. tert-butyl 9- [ [4- (4-nitropyrazol-1-yl) -1-piperidyl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate (220 mg, 477 μmol) was dissolved in MeOH (5 mL) and was added to the above mixture. The suspension was degassed under vacuum and purged with Ar three times, then the suspension was degassed under vacuum and purged with H2 three times. The resulting black mixture was stirred at 20 ℃ for 16 h under H2 (15 Psi) . The mixture was filtered through a pad of celite with MeOH (~60 mL) , then the filtrate was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 432.3 [M+H] +.
Step K: 1- [5- ( {9- [ (4- {4- [8-bromo-6- (2, 6-dichlorophenyl) -5-oxo-5, 6-dihydro-1, 3, 6-triaza-2-
naphthylamino] -1-pyrazolyl} -1-piperidyl) methyl] -3-aza-3-spiro [5.5] undecyl} carbonyl) -2-
chlorophenyl] -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a solution of 2- [ [1- [1- (3-azaspiro [5.5] undecan-9-ylmethyl) -4-piperidyl] pyrazol-4-yl] amino] -8-bromo-6- (2, 6-dichlorophenyl) pyrido [4, 3-d] pyrimidin-5-one (140 mg, 189.97 μmol, HCl) and 4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoic acid (51 mg, 190 μmol) in DCM (3 mL) was added HATU (108 mg, 285 μmol) and DIEA (86 mg, 6659 μmol) . Then the reaction mixture was stirred at 20 ℃ for 1 h under N2. The mixture was concentrated under reduced pressure to give crude. The crude was dissolved in MeOH (~1.5 mL) and purified by prep-HPLC (Column: Boston Prime C18 150 *30 mm *5 um; mobile phase: [Water (ammonia hydroxide v/v) -ACN] ; B%: 53%-73%, 10 min) . The obtained fraction was dried through lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 951.1 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.71 (s, 1H) , 10.53 (s, 1H) , 9.09 (s, 1H) , 8.44 (s, 1H) , 8.36 (s, 1H) , 7.82 (s, 1H) , 7.74 (d, J = 8.1 Hz, 2H) , 7.53-7.66 (m, 3H) , 7.38 (d, J = 8.1 Hz, 1H) , 4.07-4.18 (m, 1H) , 3.71-3.81 (m, 1H) , 3.53-3.67 (m, 3H) , 3.25-3.29 (m, 1H) , 2.83-2.93 (m, 2H) , 2.70-2.78 (m, 2H) , 2.14 (m, 2H) , 1.97-2.08 (m, 4H) , 1.84-1.97 (m, 2H) , 1.64-1.75 (m, 2H) , 1.40-1.59 (m, 5H) , 1.19-1.39 (m, 3H) , 0.96-1.15 (m, 4H) .
Example 206:
1- {5- [ (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] methyl} -9-hydroxy-3-aza-3-spiro [5.5] undecyl) carbonyl] -2-chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-397)
Scheme:
Step A: tert-butyl 2-oxa-9-azadispiro [2.2.56.23] tridecane-9-carboxylate
To a compound of TMSOI (905 mg, 4.11 mmol) in DMSO (7.4 mL) was added tBuOK (503 mg, 4.49 mmol) under N2 at 0 ℃, and the mixture was stirred at 20 ℃ for 1 h. The mixture was cooled down to 0 ℃, tert-butyl 9-oxo-3-azaspiro [5.5] undecane-3-carboxylate (1 g, 3.74 mmol, 1 eq) in DMSO (3.6 mL) was added to this solution and the solution was stirred at 0 ℃ for 3 h. The mixture was diluted with water (20 mL) and extracted with MTBE (20 mL x3) . The combined organic phase was washed with water (20 mL x2) , dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 226.1 [M+H-tBu] +.
1H NMR (400 MHz, DMSO-d6) δ 3.35-3.45 (m, 4H) , 2.63 (s, 2H) , 1.61-1.71 (m, 6H) , 1.41-1.54 (m, 15H) .
Step B: tert-butyl 9-hydroxy-9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-
azaspiro [5.5] undecane-3-carboxylate
To a mixture of tert-butyl 2-oxa-9-azadispiro [2.2.56.23] tridecane-9-carboxylate (900 mg, 3.20 mmol) and 1- (4-nitrophenyl) piperazine (500 mg, 2.41 mmol) in EtOH (10 mL) was added DIEA
(1.56 g, 12.1 mmol) under N2 and the solution was stirred at 80 ℃ for 16 h. The suspension was filtered and the filter cake was dried under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 489.3 [M+H] +.
1H NMR (400 MHz, CDCl3) δ 8.13 (d, J = 9.2 Hz, 2H) , 6.82 (d, J = 9.2 Hz, 2H) , 3.32-3.46 (m, 8H) , 2.85 (s, 1H) , 2.75-2.85 (m, 4H) , 2.40 (s, 2H) , 1.51-1.62 (m, 6H) , 1.39-1.51 (m, 15H) .
Step C: 9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecan-9-ol
To a mixture of tert-butyl 9-hydroxy-9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carboxylate (765 mg, 1.57 mmol) in HCl/dioxane (4 M, 8 mL) under N2 and the solution was stirred at 25 ℃ for 16 h. LCMS: ES23048-300-P1A showed the starting material was consumed and desired mass ion in one main peak. The suspension was diluted with EtOAc (20 mL x3) and concentrated under reduced pressure to remove HCl/dioxane to afford the desired product.
LCMS: MS (ESI) m/z 389.2 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 8.88 (s, 1H) , 8.12 (d, J = 7.6 Hz, 2H) , 7.12 (d, J = 7.6 Hz, 2H) , 4.00-4.10 (m, 2H) , 3.62-3.71 (m, 4H) , 3.20-3.31 (m, 4H) , 2.96-3.05 (m, 4H) , 1.60-1.68 (m, 4H) , 1.42-1.57 (m, 8H) .
Step D: 1- [2-chloro-5- [9-hydroxy-9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-
azaspiro [5.5] undecane-3-carbonyl] phenyl] hexahydropyrimidine-2, 4-dione
To a mixture of 9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecan-9-ol (370 mg, 784 μmol, HCl) and 4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoic acid (212 mg, 789 μmol) in DCM (3.5 mL) were added HATU (447 mg, 1.18 mmol) , DIEA (355 mg, 2.75 mmol) under N2 and the solution was stirred at 25 ℃ for 3 h. The crude was concentrated under reduced pressure to remove DCM and give crude as light brown oil. The crude was purified by Combi-flash (silica gel 4 g, 20 mL/min, 0-12%dichloromethane in methanol, 254, 280 nm, UV) to afford the desired product.
LCMS: MS (ESI) m/z 639.2 [M+H] +.
Step E: 1- [5- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -9-hydroxy-3-
azaspiro [5.5] undecane-3-carbonyl] -2-chloro-phenyl] hexahydropyrimidine-2, 4-dione
Wet Pd/C (130 mg, 122 μmol, 10%purity) was weigh to the hydrogen cylinder under Ar, MeOH (2 mL) was slowly added to the hydrogen cylinder under Ar. 1- [2-chloro-5- [9-hydroxy-9- [ [4- (4-nitrophenyl) piperazin-1-yl] methyl] -3-azaspiro [5.5] undecane-3-carbonyl] phenyl] hexahydropyrimidine-2, 4-dione (620 mg, 970) was dissolved in MeOH (10 mL) , then the solution was added to the above mixture. The suspension was degassed under vacuum and purged with Ar for three times, then the suspension was degassed under vacuum and purged with H2 for three times. The mixture was stirred at 20℃ for 16 hr under H2 (15 Psi) .
The mixture was filtered through a pad of celite with MeOH (30 mL) , the filtrate was concentrated under reduced pressure to give crude as light purple oil. The crude was purified by Combi-flash (silica gel 4 g, 20 mL/min, 0-15%dichloromethane in methanol, 254, 280 nm, UV) to afford the desired product.
LCMS: MS (ESI) m/z 609.2 [M+H] +.
Step F: 1- {5- [ (9- { [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] methyl} -9-hydroxy-3-aza-3-spiro [5.5] undecyl) carbonyl] -2-
chlorophenyl} -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
To a mixture of 1- [5- [9- [ [4- (4-aminophenyl) piperazin-1-yl] methyl] -9-hydroxy-3-azaspiro [5.5] undecane-3-carbonyl] -2-chloro-phenyl] hexahydropyrimidine-2, 4-dione (170 mg, 279 μmol) and (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (55 mg, 143 μmol) in DCE (5.5 mL) was added AcOH (8.6 mg, 143 μmol) under N2 and the solution was stirred at 50 ℃ for 16 h. The mixture was concentrated under reduced pressure to remove DCE and give crude as a brown solid (240 mg) . The crude solid was diluted with DMF (2.5 mL) and the solution was purified by prep-HPLC (column: Boston Prime C18 150 *30 mm *5 um; mobile phase: [water (FA) -ACN] ; gradient: 20%-40%B over 10 min) , and the desired fraction was concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 930.4 [M+H] +.
1H NMR (400 MHz, DMSO-d6) δ 10.50 (s, 1H) , 10.05 (brs, 1H) , 8.82 (s, 1H) , 7.96-8.07 (m, 1H) , 7.75 (d, J = 7.6 Hz, 1H) , 7.63 (d, J = 6.8 Hz, 1H) , 7.53-7.60 (m, 4H) , 7.38 (dd, J = 1.6, 6.4 Hz, 1H) , 6.85-6.94 (m, 2H) , 5.12-5.45 (m, 3H) , 4.35-4.54 (m, 2H) , 3.72-3.81 (m, 1H) , 3.53-3.67 (m, 5H) , 2.99-3.13 (m, 4H) , 2.71-2.78 (m, 2H) , 2.63-2.69 (m, 4H) , 2.25-2.32 (m, 2H) , 2.12-2.23 (m, 2H) , 1.92-1.98 (m, 1H) , 1.67-1.76 (m, 1H) , 1.56 (s, 3H) , 1.44-1.54 (m, 5H) , 1.23-1.44 (m, 8H) .
Example 207:
1- [5- ( {4- [ (4- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-ylamino} phenyl) -1-piperazinyl] -1, 1-difluoroethyl} -1-piperidyl) methyl] -1-piperidyl} carbonyl) -2-chlorophenyl] -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione (Cpd-402)
Scheme:
Step A: tert-butyl 4- [ [4- [1, 1-difluoro-2- [4- (4-nitrophenyl) piperazin-1-yl] ethyl] -1-
piperidyl] methyl] piperidine-1-carboxylate
To A solution of 1- [2, 2-difluoro-2- (4-piperidyl) ethyl] -4- (4-nitrophenyl) piperazine (500 mg, 1.41 mmol) and tert-butyl 4-formylpiperidine-1-carboxylate (300.90 mg, 1.41 mmol) in DCM (10 mL) was added Et3N (285.53 mg, 2.82 mmol) and NaBH (OAc) 3 (897.06 mg, 4.23 mmol) . The mixture was stirred at 25 ℃ for 12hr. The reaction mixture was quenched with water, and then extracted with EA (50mL *3) . The combined organic layers were dried over Na2SO4 filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (20gSilica Flash Column, Eluent of 0~2%EA/MeOH gradient @30 mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 552.4 [M+H] +.
Step B: 1- [2, 2-difluoro-2- [1- (4-piperidylmethyl) -4-piperidyl] ethyl] -4- (4-
nitrophenyl) piperazine
To a solution of tert-butyl 4- [ [4- [1, 1-difluoro-2- [4- (4-nitrophenyl) piperazin-1-yl] ethyl] -1-piperidyl] methyl] piperidine-1-carboxylate (421 mg, 763.14 μmol) in DCM (6 mL) and TFA (2 mL) was stirred at 25 ℃ for 2hr. The reaction mixture was concentrated under reduced pressure, then washed with water (10ml) and basified with Na2CO3 to pH=9. Then the mixture was extracted with EtOAc (50ml×3) . The organic phase was dried over Na2SO4 and concentrated under reduced pressure to afford the desired product.
LCMS: MS (ESI) m/z 452.6 [M+H] +.
Step C: 1- [2-chloro-5- [4- [ [4- [1, 1-difluoro-2- [4- (4-nitrophenyl) piperazin-1-yl] ethyl] -1-
piperidyl] methyl] piperidine-1-carbonyl] phenyl] hexahydropyrimidine-2, 4-dione
To a solution of 1- [2, 2-difluoro-2- [1- (4-piperidylmethyl) -4-piperidyl] ethyl] -4- (4-nitrophenyl) piperazine (800 mg, 1.77 mmol) , 4-chloro-3- (2, 4-dioxohexahydropyrimidin-1-yl) benzoic acid (475.96 mg, 1.77 mmol) in DCM (8 mL) was added HATU (1.01 g, 2.66 mmol) and Et3N (740 μL, 5.32 mmol) . The mixture was stirred at 25 ℃ for 3hr. Upon completion, the mixture was concentrated in vacuo. The residue was purified by flash silica gel chromatography (4 gSilica Flash Column, Eluent of 0~18%Ethyl acetate/Petroleum ether gradient @35mL/min) to afford the desired product.
LCMS: MS (ESI) m/z 702.4 [M+H] +.
Step D: 1- [5- [4- [ [4- [2- [4- (4-aminophenyl) piperazin-1-yl] -1, 1-difluoro-ethyl] -1-
piperidyl] methyl] piperidine-1-carbonyl] -2-chloro-phenyl] hexahydropyrimidine-2, 4-dione
To a solution of 1- [2-chloro-5- [4- [ [4- [1, 1-difluoro-2- [4- (4-nitrophenyl) piperazin-1-yl] ethyl] -1-piperidyl] methyl] piperidine-1-carbonyl] phenyl] hexahydropyrimidine-2, 4-dione (388 mg, 552.56 μmol) in DMF (5 mL) were added hypoboric acid (148.61 mg, 1.66 mmol) and 4- (4-pyridyl) pyridine (8.63 mg, 55.26 μmol) . The mixture was stirred at 25 ℃ for 0.5 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: C18
100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 0%-24%B over 6.5 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 672.4 [M+H] +
Step E: 1- [5- ( {4- [ (4- {2- [4- (p- { (R, 12Z) -16-hydroxy-16-methyl-9-oxo-2, 4, 6, 10, 21-
pentaazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3 (8) , 4, 6, 12, 17, 19-heptaen-5-
ylamino} phenyl) -1-piperazinyl] -1, 1-difluoroethyl} -1-piperidyl) methyl] -1-
piperidyl} carbonyl) -2-chlorophenyl] -1, 3, 5, 6-tetrahydro-2, 4-pyrimidinedione
A mixture of (12Z, 16R) -16-hydroxy-16-methyl-5-methylsulfinyl-2, 4, 6, 10, 21-pentazatetracyclo [15.3.1.02, 10.03, 8] henicosa-1 (21) , 3, 5, 7, 12, 17, 19-heptaen-9-one (50 mg, 129.72 μmol) , 1- [5- [4- [ [4- [2- [4- (4-aminophenyl) piperazin-1-yl] -1, 1-difluoro-ethyl] -1-piperidyl] methyl] piperidine-1-carbonyl] -2-chloro-phenyl] hexahydropyrimidine-2, 4-dione (87.20 mg, 129.72 μmol) , AcOH (77.90 mg, 1.30 mmol) in n-BuOH (3 mL) was stirred at 100 ℃ for 2 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: C18 100×40mm; mobile phase: [water (TFA) -ACN] ; gradient: 6%-36%B over 8 min) and dried by lyophilization to afford the desired product.
LCMS: MS (ESI) m/z 993.5 [M+H] +
1H NMR (400 MHz, DMSO-d6) δ 10.52 (s, 1H) , 10.09 (s, 1H) , 9.08 (s, 1H) , 8.84 (s, 1H) , 8.00 (t, J = 7.6 Hz, 1H) , 7.75 (d, J = 8.0 Hz, 1H) , 7.66 (d, J = 8.0 Hz, 1H) , 7.59 (d, J = 6.0 Hz, 3H) , 7.55 (s, 1H) , 7.39 (d, J = 8.0 Hz, 1H) , 6.95 (d, J = 6.8 Hz, 2H) , 5.33 (d, J = 18.0 Hz, 2H) , 4.46 (m, 2H) , 3.67 -3.54 (m, 7H) , 3.37 -3.05 (m, 8H) , 3.04 -2.82 (m, 8H) , 2.75 (t, J = 6.4 Hz, 2H) , 2.35 (d, J = 12.4 Hz, 1H) , 2.14 (m, 2H) , 2.08 -1.91 (m, 4H) , 1.89 -1.66 (m, 5H) , 1.56 (m, 3H) , 1.30 -1.05 (m, 2H) .
19F NMR (400 MHz, DMSO-d6) δ = -105.259
II. Biological Evaluation
Example B1: Evaluation of Anti-proliferation Effects of Compounds in TOV21G Cell Line by Cell Viability Assay
Cell Culture
TOV-21G cell line was maintained in culture conditions at 37℃ in an atmosphere with 5%CO2 in air. The tumor cells were routinely subcultured. The cells growing in an exponential growth phase were harvested and counted for plating.
Cell Plating
(1) . Count cells by haemocytometer with Trypan blue staining.
(2) . Adjust the cell concentration to proper cell density (4000/per 96-well) .
(3) . Plate 90 μL of cell suspension into the assay plates according to the plate map. Add 90 μL of
assay medium into the Blank wells.
(4) . Incubate the plates at 37℃, 5%CO2, 95%air and 100%relative humidity overnight.
Compound Stock Plate Preparation
Preparation of compound stock plate (1000X stock plates) : Serially dilute the stock solution from highest concentration (10 μM) down to lowest (1.52 μM) in DMSO.
Compound Plate (10X) Preparation and Compound Treatment
(1) . 10X concentrate compound plate preparation: Add 198 μL of assay medium into each well of the V-bottom plate; then transfer 2 μL of the stock compound solution of each concentration from the stock plate. Add 2 μL of DMSO into the Blank and Control wells. Pipette up and down to mix well. This V-plate is designated as the 10X concentrate compound plate.
(2) . For day 0 plate, add 10 μL of the DMSO-medium into the Control wells. The final DMSO concentration was 0.1%. Then directly proceed to cell viability assay.
(3) . Compound Treatment: add 10 μL compound-medium of each well from the 10X concentrate compound plate into the cells in 96-well assay plate according to the plate map. For Vehicle and Blank group, Add 10 μL of the DMSO-medium into the wells. The final DMSO concentration was 0.1%.
(4) . Return the assay plate into incubator and incubate for 3 days.
CellTiter-Glo Luminescent Cell Viability Assay
The procedures were performed according to the Promega CellTiter-Glo Luminescent Cell Viability Assay Kit manual (Promega-G7573) .
(1) . Thaw the CellTiter-Glo buffer and equilibrate to room temperature prior to use.
(2) . Equilibrate the lyophilized CellTiter-Glo Substrate to room temperature prior to use.
(3) . Transfer the entire liquid volume of CellTiter-Glo Buffer into the amber bottle containing CellTiter-Glo Substrate to reconstitute the lyophilized enzyme/substrate mixture. This forms the CellTiter-Glo Reagent.
(4) . Mix by gently vortexing to obtain a homogeneous solution.
(5) . Equilibrate the plate and its contents to room temperature for approximately 30 minutes.
(6) . Add 50 μL (equal to the half volume of culture medium present in each well) CellTiter-Glo Reagent in each well. Cover plates with aluminum foil to protect from light.
(7) . Mix contents for 2 minutes on an orbital shaker to induce cell lysis.
(8) . Allow the plate to incubate at room temperature for 10 minutes to stabilize luminescent signal.
(9) . Record luminescence on the 2104 EnVision plate reader.
Data Analysis
Inhibition rate (IR) of the tested compounds was determined by the following formula: IR (%) =
(1– (RLU compound –RLU blank) / (RLU control –RLU blank) ) *100%. The inhibitions of different dose of compound were calculated in Excel file, and then were used to plot inhibition curves and evaluate related parameters, such as Bottom (%) , Top (%) and Relative IC50. The data were interpreted by GraphPad Prism.
Representative cell viability from exemplary compounds is presented in Table 6.
Table 6:
Example B1-1
For some series of the compounds of the present disclosure, it was found that the stereoisomerism of WEE1 kinase affinity motifs was important to their degrading activity. In some specific embodiments of compounds having the macrocyclic WEE1 kinase affinity motif, degraders with R-configuration showed significant improved potency than those with S-configuration. For example, Ref-1, Ref-2 and Ref-3 are S-isomers comparing to Cpd-037, 035 and 076, respectively. However, all of them displayed > 10-fold potency loss than the corresponding degraders with R configuration macrocyclic WEE1 kinase affinity motif. (Table 6.1) .
Table 6.1
Example B2: Evaluation of the Effect of Compounds on Protein Levels of 3
Targets in TOV-21G Cell by Western
The protein levels of 3 targets (Wee1, GSPT1, CK1a) in TOV-21G cell line after 4h treatment with compounds were evaluated by Western blot.
Cell culture
The cancer cell line was maintained in culture conditions at 37℃ in an atmosphere of 5%CO2 in air. The tumor cells were routinely subcultured. The cells growing in an exponential growth phase were harvested and counted for plating.
Cell plating
(1) . Count live cells by haemocytometer with Trypan blue staining.
(2) . Adjust the cell concentration to proper cell density (1000000 per 6-well) .
(3) . Add 1.9 mL of cell suspension into one well of 6-well plate .
(4) . Incubate the plates at 37℃, 5%CO2, 95%air and 100%relative humidity overnight.
Test article stock plate preparation
Preparation of 1000X test article stock plate (1000 μM, 100 μM, 10 μM, 1 μM) .
Test article plate (20X) preparation and test article treatment
(1) . 20X concentrate test article plate preparation: add 490 μL of assay medium into each well of deep well plate; then transfer 10 μL of the Test Article stock solution of each concentration from the stock plate (1000X stock) . Add 10 μL of DMSO into Control wells. Pipette up and down to mix well. This plate is designated as the 20X concentrate compound plate.
(2) . Compound treatment: Add 100 μL of the compound-medium of each well from the 20X concentrate compound plate into the cells in 6-well assay plate. Add 100 μL of the DMSO-medium into Control wells. The final DMSO concentration was 0.1%.
(3) . Return the assay plate into incubator and incubate for 4 hours.
Protein extraction &protein quantification
1) After overnight incubation, collect cells by centrifugation and rinse cells with 1 mL of ice-
cold 1X PBS once.
2) Remove PBS and add ice-cold 1X RIPA Buffer (contains 1%Protease Inhibitor Cocktail and 1%Phosphatase Inhibitor Cocktail 2) .
3) Collect the cells into 1.5 mL Eppendorf tubes and incubate the tubes on ice for 30 mins.
4) Spin the tubes at 14,000 rpm for 10 mins and then transfer the supernatant to a new 1.5 mL tube.
5) Measure protein concentrations by PierceTM BCA Protein Assay Kit.
6) According to the result of BCA protein quantification, dilute all samples to the same final concentration (1μg/μL) using RIPA buffer plus 4X LDS Sample Buffer and 10X Sample Reducing Agent. Heat the samples at 100 ℃ for 10min.
7) Proceed to Western blotting or keep the denatured samples in the -80℃ refrigerator for long term storage.
Western blotting
1) Loaded protein samples toNovex 4-12%Bis-Tris gel, 10 μL per slot.
2) Ran electrophoresis with MES running buffer, 80V for 30 min and then 120V for 90 min.
3) Transferred protein to NC (nitrocellulose) membranes withGel Transfer Device, P3, 7 min.
4) Cut the membrane and kept the interest parts, washed the membranes with 5-10 mL 1X TBS, once for 5 min.
5) Blocked the non-specific protein by 5-10 mL InterceptTM (TBS) Blocking Buffer at room temperature, 1 h.
6) Washed the membrane with 5-10 mL 1X TBST two times for 5 min each.
7) Incubated membrane with 5-10 mL diluted primary antibody in InterceptTM (TBS) Blocking Buffer at 4℃, with gentle agitation overnight.
8) Washed the membrane with 5-10 mL 1X TBST three times for 10 min each.
9) Incubated membrane with 5-10 mL800CW Goat Anti-Rabbit IgG secondary antibody (generally 1: 10000) with gentle agitation in the dark for 1h at room temperature.
10) Washed the membrane with 5-10 mL 1X TBST three times for 10 min each.
11) Detect the near-infrared fluorescence signal by Odyssey Clx system.
Quantification of band intensity
Intensity of individual bands was quantified using Image Studio software.
Data analysis
Intensity of individual bands were got for analysis. Protein expression can be quantitated in relation to a housekeeping protein, and then the relative protein expression of each sample/group
were normalized to vehicle group. Normalized relative folds of each sample are plotted in bar chart underneath the gel pictures.
The protein levels of 3 targets (Wee1, GSPT1, CK1a) in TOV-21G cell line after treated with exemplary compounds are shown in FIG. 1.
Example B3: In Vivo Pharmacokinetic Determinations
The study was conducted in male CD-1 mice with an age of 6-8 weeks. Animals received intravenous injection at the dose of 1.0 mpk or 2 mpk, and oral administration at the dose of 5.0 mg/kg (cassette) or 40-60 mpk. Blood samples were taken from three different mice at certain time points (e.g.: 15, 30 min, 1, 2, 4, 6, 8, 24 hours post dose, etc. ) . Blood samples were taken from mice and transferred to EDTA tubes. Blood was centrifuged in a 4℃ centrifuge to obtain plasma. All plasma samples were stored in a freezer at -75±15℃ prior to analysis. WinNonlin (PhoenixTM) software was used for pharmacokinetic calculations and the following pharmacokinetic parameters were calculated from the plasma concentration versus time data: T1/2, AUC, Cl, and VDss.
Representative pharmacokinetic parameters for exemplary compounds are presented in Table 7 and Table 8.
Table 7 Mouse PK:
Table 8 Rat PK
Example B4: Evaluation of Anti-proliferation Effects of Compounds in SK-UT-1 Cell Line by Cell Viability Assay
Thawing SK-UT-1 cell
1) Thaw the vial by gentle agitation in a 37℃ water bath. To reduce the possibility of contamination, keep the O-ring and cap out of the water. Thawing should be rapid (approximately 2 minutes) .
2) Remove the vial from the water bath as soon as the contents are thawed, and decontaminate by dipping in or spraying with 70%ethanol. All of the operations from this point on should be carried out under strict aseptic conditions.
3) Transfer the vial contents to a centrifuge tube containing 9.0 mL complete growth medium and spin at approximately 250 xg for 5 minutes.
4) Resuspend cell pellet with the recommended complete growth medium and dispense into a 25 cm2 or a 75 cm2 culture flask.
5) Incubate the cells at a 37℃, 5% (v/v) CO2 incubator.
Cell Cryopreservation
1) Preparation of frozen medium: 10%DMSO + 90%FBS.
2) The cells in good growth state or in logarithmic phase were selected for cryopreservation. Transfer the cells into a clean 50 ml tube and centrifuge at 250 xg for 5 min. Resuspend the cell pellet with appropriate volume of frozen medium and aliquot.
3) Next day, transfer to liquid nitrogen dewar for long-term storage.
Prepare Lysate samples
1) Compounds are dissolved at 10 mM stock solution. And transfer 20 uL of diluted solution to a 96 PCR-plate (Axygen, PCR-96M2-S-C) . Perform 3 fold, 9-point dilution via transferring 10 uL compound into 20 μL DMSO.
2) Prepare 10X compounds via transfer 2ul compounds from compound source plate into 198 ul medium in 96-well cell plate (Corning 3894) .
3) On the day of experiment cells, transfer the cells into a clean 50ml tube and centrifuge at 250 xg for 5 min.
4) Resuspend the cell pellet with appropriate volume of pre-warmed complete growth medium and aliquot a sample for counting using theAutomated Cell Counter.
5) According to the cell density, dilute cells to designed concentration (5000 cells/90ul/well) and seed cells into 96 well cell plates (PE Culture-96) . Keep some wells for no cell control (only add medium) which will also be tested the CTG signal.
6) The next day, transfer 10ul 10X compounds into cell suspension.
7) To mix, agitate on a plate shaker at 400 rpm for 1-2 minutes . Incubate for 72 hr at 37℃ , 5%CO2 incubator.
CTG assay and plate Read
1) Remove cell plate from the incubator and equilibrate to room temperature.
2) Add a volume of2.0 Assay equal to the culture medium in each well, Agitate on a plate shaker at 600 rpm for 10 minutes at room temperature, and stay out of the light.
3) Read on Ensight.
Representative cell viability from exemplary compounds is presented in Table 9.
Table 9: SKUT1 CTG
Example B5: Evaluation of WEE1 Degradation and Anti-tumor Effects of Compounds in CDX model.
Cell Culture: Tumor cells (TOV21G, SK-UT-1 and OVCAR3) were cultured at 37 ℃ in an atmosphere of 5%CO2 in air. The tumor cells were routinely subcultured twice weekly. The cells growing in an exponential growth phase were harvested and counted for tumor inoculation.
Animals: Balb/c nude mice, female, 6-8 weeks, weighing approximately 18-22g, mice were purchased from Beijing VR Laboratory Animal Co., LTD. or other certified vendors.
Tumor Cell Inoculation: Each mouse was inoculated subcutaneously at the right flank with tumor cells for tumor development. The animals were randomized and treatment will be started when the average tumor volume reached approximately 100-150 mm3. The test articles were dosed orally.
Anti-tumor efficacy study:
Tumor sizes were measured twice per week in two dimensions using a caliper, and the volume was expressed in mm3 using the formula: V = 0.5 a x b2 where a and b are the long and short diameters of the tumor, respectively. The tumor sizes were then used for the calculations of T/C and TGI values. The T/C value (in percent) is an indication of antitumor effectiveness; T and C are the mean volume of the treated and control groups, respectively, on a given day. TGI is calculated for each group using the formula: TGI (%) = [1- (Ti-T0) / (Vi-V0) ] ×100; Ti is the average tumor volume of a treatment group on a given day, T0 is the average tumor volume of the treatment group on the first day of treatment, Vi is the average tumor volume of the vehicle
control group on the same day with Ti, and V0 is the average tumor volume of the vehicle group on the first day of treatment.
PD analysis:
Western blotting protocol
Protein Extraction &Protein Quantification
1. Take the snap-frozen tissues out from -80℃ freezer or liquid nitrogen tank.
2. Cut up to about 30-100mg of tissue. Place the tissues in a 2mL microcentrifuge tube, and add 400μL of RIPA Buffer (contains 1%Protease Inhibitor Cocktail and 1%Phosphatase Inhibitor Cocktail 2) .
3. Grind tumors with Tissuelyser at 50Hz for 5min.
4. Keep the tissue lysates on ice for 30min.
5. Centrifuge at 12,000 rpm at 4℃ for 10min; gently transfer the supernatants to a new microcentrifuge tube kept on ice.
6. Measure protein concentrations by PierceTM BCA Protein Assay Kit.
7. According to the result of BCA protein quantification, dilute all samples to the same final concentration (at 4μg/μL) using RIPA buffer plus 4X LDS Sample Buffer and 10X Sample Reducing Agent . Heat the samples at 100℃ for 10min.
8. Proceed to western blotting or keep the denatured samples in the -80℃ refrigerator for long term storage.
Western Blotting
1. Add protein samples to4-12%Bis-Tris Gel (26-well) , 10 μL per slot.
2. Electrophoresis with MES running buffer, 80 V for 30 min and then 120 V for 90 min.
3. Transfer protein to nitrocellulose membranes withGel Transfer Device, 20 V, 7 min.
4. Cut the membrane and keep the interest parts, wash the membranes with 5-10 mL 1x TBS, three times for 5 min each.
5. Block the non-specific protein by 5-10 mL Odyssey blocking buffer at room temperature, 1 h.
6. Wash the membrane with 1x TBST five times for 5 min each.
7. Incubate membrane with 5-10 mL freshly prepared primary antibody in Odyssey blocking buffer with 0.1%Tween 20 at 4 ℃, with gentle agitation overnight.
8. Wash the membrane with 5-10 mL 1x TBST five times for 5 min each.
9. These next steps should be operated in the dark. Incubate membrane with5-10 mL secondary antibodies mixture in Odyssey blocking buffer with 0.1%Tween20 with gentle agitation for 1 h at room temperature.
10. Wash the membrane with 5-10 mL 1x TBST five times for 5 min each.
11. Wash the membrane with 5-10 mL 1x TBS twice for 5 min each.
12. Detect the near-infrared fluorescence signal by Odyssey Clx system.
Quantification of band intensity
Intensity of individual bands were quantified using Image Studio Lite Ver 5.2 software.
Data Analysis
Band intensity of individual bands were got for analysis. Protein expression can be quantitated in relation to a reference protein, and then the normalized folds of protein expression in each sample were ratios to vehicle group. The normalized data were interpreted by GraphPad Prism.
Representative data from exemplary compounds is presented in Table 10, Table 11, Fig. 2 and Fig. 3.
Table 10: Evaluation of in-vivo WEE1 degradation potency of compounds in TOV21G CDX model after single dose:
Table 11: Anti-tumor efficacy of WEE1 degraders in SK-UT-1 CDX model
III. Preparation of Pharmaceutical Dosage Forms
Example C1: Oral capsule
The active ingredient is a compound of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , or Formula (III-2) , a compound of Table 1, Table 2, Table 3, Table 4, or Table 5, or a pharmaceutically acceptable salt or solvate thereof. A capsule for oral administration is prepared by mixing 1-1000 mg of active ingredient with starch or other suitable powder blend. The mixture is incorporated into an oral dosage unit such as a hard gelatin capsule, which is suitable for oral administration.
Example C2: Solution for injection
The active ingredient is a compound of Formula (I) , Formula (I-1) , Formula (I-2) , Formula (1-3) , Formula (1-3a) , Formula (1-3b) , Formula (I-3c) , Formula (II) , Formula (II-1) , Formula (II-2) , Formula (III) , Formula (III-1) , or Formula (III-2) , a compound of Table 1, Table 2, Table 3, Table 4, or Table 5, or a pharmaceutically acceptable salt thereof, and is formulated as a solution in sesame oil at a concentration of 50 mg-eq/mL.
The examples and embodiments described herein are for illustrative purposes only and various modifications or changes suggested to persons skilled in the art are to be included within the spirit and purview of this application and scope of the appended claims.
Claims (226)
- A compound, or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (I) :
wherein,R1, R2, R3, and R4 are each independently selected from hydrogen, optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, and cyano; orR1 and R2, R2 and R3, or R3 and R4 together with the intervening atoms to which they are attached form an optionally substituted 5-6 membered carbocyclyl or optionally substituted heterocyclyl ring;R5 isorR1 and R5 are taken together with the intervening atoms to which they are attached to form an optionally substituted 10-12 membered ring;each R6 is independently selected from halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;W is CH, CR6, or N;v is 0, 1, 2, or 3;each R7 is independently hydrogen or optionally substituted C1-6 alkyl; ortwo R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring;ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;L is -Ak1-Z1-Ak2-Z2-Ak3-*;Ak1 is - (CR8R9) k-or -O-;Ak2 is - (CR8R9) m-;Ak3 is - (CR8R9) n-;each of k, m, and n is an integer selected from 0 to 10;R8 and R9 are each independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl; oreach - (CR8R9) -may independently form -C (O) -;Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10 membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-11 cycloalkylene;wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;wherein the point of bonding to the CRBN ligand is indicated by the asterisk; andU is a CRBN ligand. - The compound of claim 1, whereinR1, R2, R3, and R4 are each independently selected from hydrogen, optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, and cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; orR1 and R2, R2 and R3, or R3 and R4 together with the intervening atoms to which they are attached form an optionally substituted 5-6 membered carbocyclyl or optionally substituted heterocyclyl ring, wherein the carbocyclyl and heterocyclyl ring is optionally substituted with one or more Re;R5 isorR1 and R5 are taken together to with the intervening atoms to which they are attached to form an optionally substituted 10-12 membered ring, wherein the 10-12 membered ring is optionally substituted with one or more Re;each R6 is independently selected from halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl, wherein the alkyl, haloalkyl, alkoxy, heteroalkyl, carbocyclyl, and heterocyclyl are independently optionally substituted with one or more Re;v is 0, 1, 2, or 3;each R7 is independently hydrogen or optionally substituted C1-6 alkyl, wherein the alkyl, is optionally substituted with one or more Re; ortwo R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring, wherein the 3-6 membered heterocyclyl ring is optionally substituted with one or more Re;ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB;L is -Ak1-Z1-Ak2-Z2-Ak3-*;Ak1 is - (CR8R9) k-or -O-;Ak2 is - (CR8R9) m-;Ak3 is - (CR8R9) n-;each of k, m, and n is an integer selected from 0 to 10;R8 and R9 are each independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl, wherein the alkyl and cycloalkyl are each independently optionally substituted with one or more Re; oreach - (CR8R9) -may independently form -C (O) -;Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10 membered heteroaryl, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene, wherein the heteroarylene, heterocyclene, and cycloalkylene are independently optionally substituted with one or more Re;wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;each Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl; ortwo Re attached to the same atom are taken together to form -C (O) -; andeach RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; ortwo RB groups attached to the same atom are taken together to form -C (O) -; ortwo RB groups attached to different atoms are taken together to form a bridge.
- The compound of claim 1 or claim 2, wherein R1 is optionally substituted branched C1-6 alkyl, wherein the alkyl is optionally substituted with hydroxy; andR2, R3, and R4 are hydrogen.
- The compound of any one of claims 1-3, wherein R1 is
- The compound of claim 1 or claim 2, wherein R1 and R2 together with the intervening atoms to which they are attached form an optionally substituted 5-6 membered cycloalkyl or optionally substituted 5-6 membered heterocyclyl ring, wherein the cycloalkyl and heterocyclyl are optionally substituted with one or more substituents selected from C1-6 alkyl and hydroxyl.
- The compound of claim 1 or claim 5, wherein R1 and R2 together with the intervening atoms to which they are attached form
- The compound of any one of claims 1-6, wherein R5 is
- The compound of claim 1 or claim 2, wherein R1 and R5 are taken together to with the intervening atoms to which they are attached to form an optionally substituted 10-12 membered ring, wherein the ring is optionally substituted with one or more substituents selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl.
- The compound of claim 8, wherein the ring comprises an alkene or alkyne bond.
- The compound of any one of claims 1-9, wherein W is CH or CR6.
- The compound of any one of claims 1, 2, or 8-10, wherein R1 and R5 are taken together to with the intervening atoms to which they are attached to form a compound of Formula (I-3) :
wherein,the wavy bondin Formula (I-3) represents an E-isomer or a Z-isomer with respect to the double bond; andeach R10 is independently selected from hydrogen, halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl. - The compound of claim 11, wherein the compound of Formula (I-3) has the structure of Formula (I-3-R) :
- The compound of claim 11 or 12, wherein each R10 is independently selected from hydrogen, halogen, hydroxyl, and C1-3 alkyl.
- The compound of any one of claims 11-13, wherein each R10 is independently selected from hydrogen or fluoro.
- The compound of any one of claims 1-14, wherein each R6 is independently selected from halogen, cyano, -OR7, -N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, and optionally substituted C1-6 alkoxy.
- The compound of any one of claims 1-15, wherein each R6 is independently selected from halogen, cyano, -OR7, -N (R7) 2, and optionally substituted C1-4 alkyl.
- The compound of any one of claims 1-16, wherein each R6 is independently selected from -F, -Cl, cyano, methyl, and -CH2OH.
- The compound of any one of claims 1-17, wherein v is 0, 1, or 2.
- The compound of any one of claims 1-18, wherein v is 0 or 1.
- The compound of any one of claims 1-19, wherein v is 0.
- The compound of any one of claims 1-20, wherein ring B is an optionally substituted C11-14 cycloalkyl or an optionally substituted 11-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB;each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; ortwo RB groups attached to the same atom are taken together to form -C (O) -; ortwo RB groups attached to different atoms are taken together to form a bridge; andeach Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl; ortwo Re attached to the same atom are taken together to form -C (O) -.
- The compound of any one of claims 1-21, wherein ring B is a 6-14 membered carbocyclyl ring or a 6-14 membered heterocyclyl ring selected from:
wherein,Y1 and Y2 are N, CRB, or CH;B is a bridge selected from C1-3 alkylene and C1-2 heteroalkylene;each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano; ortwo RB groups attached to the same atom are taken together to form -C (O) -;j is 0, 1, 2, 3, or 4; ando and p are each independently 1 or 2. - The compound of any one of claims 1-22, wherein ring B has the structure:
wherein,Y1 and Y2 are N, CRB, or CH;each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano; ortwo RB groups attached to the same atom are taken together to form -C (O) -; andj is 0, 1, 2, 3, or 4. - The compound of claim 22, wherein ring B is
- The compound of claim 22, wherein ring B is
- The compound of claim 22, wherein ring B is
- The compound of any one of claims 2-26, wherein each RB is independently optionally substituted straight or branched C1-3 alkyl or halogen.
- The compound of any one of claims 2-27, wherein each RB is independently methyl or fluoro.
- The compound of any one of claims 22, or 26-28, wherein B is C1-3 alkylene.
- The compound of any one of claims 22, or 26-28, wherein B is C1-2 heteroalkylene.
- The compound of claim 22, wherein ring B is selected from:
- The compound of any one of claims 22-31, wherein each RB is independently methyl or fluoro; and j is 0, 1, or 2.
- The compound of any one of claims 1-32, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X, X1, and X2 at each instance is independently CH, CRe, or N;each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6;n is 0, 1, 2, 3, 4, 5 or 6; andq is 0, 1, 2, 3, or 4. - The compound of any one of claims 1-33, wherein R8 and R9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -.
- The compound of any one of claims 1-34, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X, X1, and X2 at each instance is independently CH, CRe or N;each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, C1-3 alkyl, and -CF3;k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6; andn is 0, 1, 2, 3, 4, 5 or 6. - The compound of any one of claims 1-35, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X, X1, and X2 at each instance is independently CH, CRe, or N;each Re in Z1, X, X1, and X2 is independently selected from -F, methyl, and -CF3;k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6; andn is 0, 1, 2, 3, 4, 5 or 6. - The compound of any one of claims 1-36, whereink is 0, 1, or 2;m is 0, 1, or 2; andn is 0.
- The compound of any one of claims 1-37, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
- The compound of any one of claims 1-32, wherein Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene.
- The compound of any one of claims 1-32, wherein-Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;each RL is independently hydrogen, halogen, or C1-3 alkyl.k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6;n is 0, 1, 2, 3, 4, 5 or 6;q is 0, 1, 2, 3, or 4; andh is 0, 1, 2, 3, or 4. - The compound of claim 40, wherein R8 and R9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -.
- The compound of any one of claims 1-32, 40, or 41, wherein-Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;each RL is independently hydrogen, halogen, or C1-3alkyl.k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6;n is 0, 1, 2, 3, 4, 5 or 6;q is 0, 1, 2, 3, or 4; andh is 0, 1, 2, 3, or 4. - The compound of any one of claims 1-32, or 40-42, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;each RL is independently hydrogen, halogen, or C1-3alkyl.k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6;n is 0, 1, 2, 3, 4, 5 or 6;q is 0, 1, 2, 3, or 4; andh is 0, 1, 2, 3, or 4. - The compound of any one of claims 40-43, whereink is 0, 1, or 2;m is 0, 1, or 2; andn is 0.
- The compound of any one of claims 1-32 or 40-44, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,each RL is independently hydrogen, halogen, or C1-3alkyl;each Re is independently selected from halogen, -OH, and methyl;q is 0, 1, or 2; andh is 0, 1, or 2. - The compound of any one of claims 1-32, wherein at least one of Z1 and Z2 is a spirocyclic heterocyclene.
- The compound of claim 46, wherein the spirocyclic heterocyclene is a 9-11 membered spirocyclic heterocyclene.
- The compound of claim 46 or 47, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
X1 and X2 at each instance is independently CH, CRe, or N;each Re in X1 and X2 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;k is 0, 1, 2, 3, 4, 5, or 6; andm is 0, 1, 2, 3, 4, 5 or 6. - The compound of any one of claims 46-48, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X1 and X2 at each instance is independently CH, CRe or N;each Re in X1 and X2 is independently selected from halogen, -NH2, -OH, C1-3 alkyl, and -CF3;k is 0, 1, 2, 3, 4, 5, or 6; andm is 0, 1, 2, 3, 4, 5 or 6. - The compound of claim 48 or 49, whereink is 0, 1, or 2; andm is 0, 1, or 2.
- The compound of any one of claims 1-39 or 46-50, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
- A compound, or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (II) :
wherein,R50 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl;R51 and R52 are each independently hydrogen or optionally substituted straight or branched C1-6 alkyl; orR51 and R52 are taken together to form -C (O) -;represents a single or double bond;R53, R54, R55, R56, R57, R58, and R59 are each independently hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;each R7 is independently hydrogen or optionally substituted C1-6 alkyl; ortwo R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring;ring B is an optionally substituted C6-14cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;L is -Ak1-Z1-Ak2-Z2-Ak3-*;Ak1 is - (CR8R9) k-or -O-;Ak2 is - (CR8R9) m-;Ak3 is - (CR8R9) n-;each of k, m, and n is an integer selected from 0 to 10;each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl; oreach - (CR8R9) -may independently form -C (O) -;Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-11 cycloalkylene;wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;wherein the point of bonding to the CRBN ligand is indicated by the asterisk; andU is a CRBN ligand;provided that whenrepresents a double bond, then R50 and R51 are absent. - The compound of claim 52, wherein,R50 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl, wherein the alkyl or cycloalkyl are each optionally substituted with one or more Re;R51 and R52 are each independently hydrogen or optionally substituted straight or branched C1-6 alkyl, wherein the alkyl is optionally substituted with one or more Re; orR51 and R52 are taken together to form -C (O) -;represents a single or double bond;R53, R54, R55, R56, R57, R58, and R59 are each independently hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12- membered heterocyclyl, wherein the alkyl, haloalkyl, alkoxy, heteroalkyl, carbocyclyl, and heterocyclyl are independently optionally substituted with one or more Re;each R7 is independently hydrogen or optionally substituted C1-6 alkyl, wherein the alkyl, is optionally substituted with one or more Re; ortwo R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring, wherein the 3-6 membered heterocyclyl ring is optionally substituted with one or more Re;ring B is an optionally substituted C6-14cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB;L is -Ak1-Z1-Ak2-Z2-Ak3-*;Ak1 is - (CR8R9) k-or -O-;Ak2 is - (CR8R9) m-;Ak3 is - (CR8R9) n-;each of k, m, and n is an integer selected from 0 to 10;each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl, wherein the alkyl and cycloalkyl are each optionally substituted with one or more Re; oreach - (CR8R9) -may independently form -C (O) -;Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene, wherein the heteroaryl, heterocyclene, and cycloalkylene are independently optionally substituted with one or more Re;wherein at least one of Z1 and Z2 is heterocyclene or cycloalkylene;each Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, C1-C6 aminoalkyl, and C1-C6 heteroalkyl; ortwo Re attached to the same atom are taken together to form -C (O) -; andeach RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; ortwo RB groups attached to the same atom are taken together to form -C (O) -; ortwo RB groups attached to different atoms are taken together to form a bridge.
- The compound of claim 52 or claim 53, wherein R50 is hydrogen, methyl, ethyl, or cyclopropyl.
- The compound of any one of claims 52-54, wherein R50 is methyl.
- The compound of any one of claims 52-54, wherein R50 is hydrogen.
- The compound of any one of claims 52-56, wherein R51 and R52 are each hydrogen, or R51 and R52 are taken together to form -C (O) -.
- The compound of any one of claims 52-57, wherein R51 and R52 are each hydrogen.
- The compound of any one of claims 52-57, wherein R51 and R52 are taken together to form -C (O) -.
- The compound of any one of claims 52-59, whereinrepresents a single bond.
- The compound of any one of claims 52-59, whereinrepresents a double bond.
- The compound of any one of claims 52-61, wherein R53, R57, R58, and R59 are each independently hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl; andR54, R55, and R56 are each hydrogen.
- The compound of any one of claims 52-62, wherein R53, R54, R55, R56, R57, R58, and R59 are each independently hydrogen, halogen, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, or optionally substituted C1-6 heteroalkyl.
- The compound of any one of claims 52-63, wherein R53 and R57 are each independently halogen, cyano, -OR7, -N (R7) 2, -SR7, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, or optionally substituted C1-6 heteroalkyl;R58 and R59 are each independently hydrogen, halogen, cyano, -OR7, -N (R7) 2, -SR7, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, or optionally substituted C1-6 heteroalkyl; andR54, R55, and R56 are each hydrogen.
- The compound of any one of claims 52-64, wherein R53 and R57 are each independently halogen; andR54, R55, and R56 are each hydrogen.
- The compound of any one of claims 52-65, wherein R53 and R57 are each independently fluoro or chloro; and R54, R55, and R56 are each hydrogen.
- The compound of any one of claims 52-63, wherein R53 is hydrogen.
- The compound of any one of claims 52-63 or 67, wherein R57 is hydrogen.
- The compound of any one of claims 52-68, wherein R58 and R59 are each independently hydrogen, -F, -Cl, or methyl.
- The compound of any one of claims 52-69, wherein ring B is an optionally substituted C11-14 cycloalkyl or an optionally substituted 11-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB;each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; ortwo RB groups attached to the same atom are taken together to form -C (O) -; ortwo RB groups attached to different atoms are taken together to form a bridge; andeach Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl; ortwo Re attached to the same atom are taken together to form -C (O) -.
- The compound of any one of claims 52-70, wherein ring B is a C6-14 cycloalkyl ring or a 6-14 membered heterocyclyl ring selected from:
wherein,Y1 and Y2 are N or CH;B is a bridge selected from C1-3 alkylene and C1-2 heteroalkylene;each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano; ortwo RB groups attached to the same atom are taken together to form -C (O) -;j is 0, 1, 2, 3, or 4; ando and p are each independently 1 or 2. - The compound of claim 71, wherein ring B is
- The compound of claim 71, wherein ring B is
- The compound of claim 71, wherein ring B is
- The compound of any one of claims 53-74, wherein each RB is independently optionally substituted straight or branched C1-3 alkyl or halogen.
- The compound of any one of claims 53-75, wherein each RB is independently methyl or fluoro.
- The compound of any one of claims 71, or 74-76, wherein B is C1-3 alkylene.
- The compound of any one of claims 71, or 74-76, wherein B is C1-2 heteroalkylene.
- The compound of claim 71, wherein ring B is selected from:
- The compound of any one of claims 71-79, wherein each RB is independently methyl or fluoro; and j is 0, 1, or 2.
- The compound of any one of claims 52-80, wherein wherein-Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X, X1, and X2 at each instance is independently CH, CRe, or N;each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6; andn is 0, 1, 2, 3, 4, 5 or 6; andq is 0, 1, 2, 3, or 4. - The compound of any one of claims 52-81, wherein R8 and R9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -.
- The compound of any one of claims 52-82, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X, X1, and X2 at each instance is independently CH, CRe, or N;each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, C1-3 alkyl, and -CF3;k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6; andn is 0, 1, 2, 3, 4, 5 or 6. - The compound of any one of claims 52-83, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X, X1, and X2 at each instance is independently CH, CRe, or N;each Re in Z1, X, X1, and X2 is independently selected from -F, methyl, and -CF3;k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6; andn is 0, 1, 2, 3, 4, 5 or 6. - The compound of any one of claims 52-84, whereink is 0, 1, or 2;m is 0, 1, or 2; andn is 0.
- The compound of any one of claims 52-85, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
- The compound of any one of claims 52-80, wherein Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene.
- The compound of any one of claims 52-80, wherein-Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;each RL is independently hydrogen, halogen, or C1-3 alkyl.k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6;n is 0, 1, 2, 3, 4, 5 or 6;q is 0, 1, 2, 3, or 4; andh is 0, 1, 2, 3, or 4. - The compound of any one of claims 52-80, or 88, wherein R8 and R9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -.
- The compound of any one of claims 52-80, 88, or 89, wherein-Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;each RL is independently hydrogen, halogen, or C1-3alkyl.k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6;n is 0, 1, 2, 3, 4, 5 or 6;q is 0, 1, 2, 3, or 4; andh is 0, 1, 2, 3, or 4. - The compound of any one of claims 52-80, or 88-90, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;each RL is independently hydrogen, halogen, or C1-3alkyl.k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6;n is 0, 1, 2, 3, 4, 5 or 6;q is 0, 1, 2, 3, or 4; andh is 0, 1, 2, 3, or 4. - The compound of any one of claims 52-80, or 88-91, whereink is 0, 1, or 2;m is 0, 1, or 2; andn is 0.
- The compound of any one of claims 52-80 or 88-92, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,each RL is independently hydrogen, halogen, or C1-3alkyl;each Re is independently selected from halogen, -OH, and methyl;q is 0, 1, or 2; andh is 0, 1, or 2. - A compound, or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (III) :
wherein,R81 and R82 are each independently selected from hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;each R83 is independently selected from halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, - C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl;R84 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl;R85 and R86 are each independently hydrogen or optionally substituted straight or branched C1-6 alkyl; orR85 and R86 are taken together to form -C (O) -;represents a single or double bond;W is CH, CR83, or N;v is 0, 1, 2, or 3;each R7 is independently hydrogen or optionally substituted C1-6 alkyl; ortwo R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring;ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;L is -Ak1-Z1-Ak2-Z2-Ak3-*;Ak1 is - (CR8R9) k-or -O-;Ak2 is - (CR8R9) m-;Ak3 is - (CR8R9) n-;each of k, m, and n is an integer selected from 0 to 10;each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl; oreach - (CR8R9) -may independently form -C (O) -;Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-11 cycloalkylene;wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;wherein the point of bonding to the CRBN ligand is indicated by the asterisk; andU is a CRBN ligand;provided that whenrepresents a double bond, then R84 and R85 are absent. - The compound of claim 94, wherein,R81 and R82 are each independently selected from hydrogen, halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl, wherein the alkyl, haloalkyl, alkoxy, heteroalkyl, carbocyclyl, and heterocyclyl are independently optionally substituted with one or more Re;each R83 is independently selected from halogen, nitro, cyano, -OR7, -N (R7) 2, -SR7, -S (O) R7, -S (O) 2R7, -S (O) 2N (R7) 2, -NR7S (O) 2R7, -NR7S (O) 2N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, -P (O) (OR7) 2, -P (O) (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C1-6 heteroalkyl, optionally substituted C3-12 carbocyclyl, and optionally substituted 3-to 12-membered heterocyclyl, wherein the alkyl, haloalkyl, alkoxy, heteroalkyl, carbocyclyl, and heterocyclyl are independently optionally substituted with one or more Re;R84 is hydrogen, optionally substituted straight or branched C1-6 alkyl, or optionally substituted C3-6 cycloalkyl, wherein the alkyl or cycloalkyl are each optionally substituted with one or more Re;R85 and R86 are each independently hydrogen or optionally substituted straight or branched C1-6 alkyl, wherein the alkyl is optionally substituted with one or more Re;orR85 and R86 are taken together to form -C (O) -;represents a single or double bond;W is CH, CR83, or N;v is 0, 1, 2, or 3;each R7 is independently hydrogen or optionally substituted C1-6 alkyl, wherein the alkyl, is optionally substituted with one or more Re; ortwo R7 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring, wherein the 3-6 membered heterocyclyl ring is optionally substituted with one or more Re;ring B is an optionally substituted C6-14cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB;L is -Ak1-Z1-Ak2-Z2-Ak3-*;Ak1 is - (CR8R9) k-or -O-;Ak2 is - (CR8R9) m-;Ak3 is - (CR8R9) n-;each of k, m, and n is an integer selected from 0 to 10;each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl, wherein the alkyl and cycloalkyl are each optionally substituted with one or more Re; oreach - (CR8R9) -may independently form -C (O) -;Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene, wherein the heteroaryl, heterocyclene, and cycloalkylene are independently optionally substituted with one or more Re;wherein at least one of Z1 and Z2 is heterocyclene or cycloalkylene;each Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, C1-C6 aminoalkyl, and C1-C6 heteroalkyl; ortwo Re attached to the same atom are taken together to form -C (O) -; andeach RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; ortwo RB groups attached to the same atom are taken together to form -C (O) -; ortwo RB groups attached to different atoms are taken together to form a bridge.
- The compound of claim 94 or claim 95, wherein R84 is hydrogen, methyl, ethyl, or cyclopropyl.
- The compound of any one of claims 94-96, wherein R84 is methyl.
- The compound of any one of claims 94-96, wherein R84 is hydrogen.
- The compound of any one of claims 94-97, wherein R85 and R86 are each hydrogen, or R85 and R86 are taken together to form -C (O) -.
- The compound of any one of claims 94-99, wherein R85 and R86 are each hydrogen.
- The compound of any one of claims 94-99, wherein R85 and R86 are taken together to form -C (O) -.
- The compound of any one of claims 94-101, whereinrepresents a single bond.
- The compound of any one of claims 94-101, whereinrepresents a double bond.
- The compound of any one of claims 94-103, wherein R81 and R82 are each independently selected from hydrogen, halogen, cyano, hydroxy, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, and optionally substituted C1-6 alkoxy.
- The compound of any one of claims 94-104, wherein R81 and R82 are each independently selected from hydrogen and halogen.
- The compound of any one of claims 94-105, wherein at least one of R81 and R82 is halogen.
- The compound of any one of claims 94-106, wherein R81 and R82 are each independently fluoro or chloro.
- The compound of any one of claims 94-107, wherein each R83 is independently selected from halogen, cyano, -OR7, -N (R7) 2, -C (O) R7, -C (O) OR7, -OC (O) R7, -OC (O) OR7, -OC (O) N (R7) 2, -NR7C (O) R7, -NR7C (O) OR7, -NR7C (O) N (R7) 2, -C (O) N (R7) 2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, and optionally substituted C1-6 alkoxy.
- The compound of any one of claims 94-108, wherein each R83 is independently selected from halogen, cyano, -OR7, -N (R7) 2, and optionally substituted C1-4 alkyl.
- The compound of any one of claims 94-109, wherein each R83 is independently selected from -F, -Cl, cyano, methyl, and -CH2OH.
- The compound of any one of claims 94-110, wherein W is CH or CR83.
- The compound of any one of claims 94-111, wherein v is 0, 1, or 2.
- The compound of any one of claims 94-112, wherein v is 0 or 1.
- The compound of any one of claims 94-113, wherein v is 0.
- The compound of any one of claims 94-114, wherein ring B is an optionally substituted C11-14 cycloalkyl or an optionally substituted 11-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB;each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; ortwo RB groups attached to the same atom are taken together to form -C (O) -; ortwo RB groups attached to different atoms are taken together to form a bridge; andeach Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl; ortwo Re attached to the same atom are taken together to form -C (O) -.
- The compound of any one of claims 94-115, wherein ring B is a C6-14cycloalkyl ring or a 6-14 membered heterocyclyl ring selected from:
wherein,Y1 and Y2 are N or CH;B is a bridge selected from C1-3 alkylene and C1-2 heteroalkylene;each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano; ortwo RB groups attached to the same atom are taken together to form -C (O) -;j is 0, 1, 2, 3, or 4; ando and p are each independently 1 or 2. - The compound of claim 116, wherein ring B is
- The compound of claim 116, wherein ring B is
- The compound of claim 116, wherein ring B is
- The compound of any one of claims 95-119, wherein each RB is independently optionally substituted straight or branched C1-3 alkyl or halogen.
- The compound of any one of claims 95-120, wherein each RB is independently methyl or fluoro.
- The compound of any one of claims 116, or 119-121, wherein B is C1-3 alkylene.
- The compound of any one of claims 116, or 119-121, wherein B is C1-2 heteroalkylene.
- The compound of claim 116, wherein ring B is selected from:
- The compound of any one of claims 116-124, wherein each RB is independently methyl or fluoro; and j is 0, 1, or 2.
- The compound of any one of claims 94-125, wherein wherein-Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X, X1, and X2 at each instance is independently CH, CRe, or N;each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6; andn is 0, 1, 2, 3, 4, 5 or 6; andq is 0, 1, 2, 3, or 4. - The compound of any one of claims 94-126, wherein R8 and R9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -.
- The compound of any one of claims 94-127, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X, X1, and X2 at each instance is independently CH, CRe, or N;each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, C1-3 alkyl, and -CF3;k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6; andn is 0, 1, 2, 3, 4, 5 or 6. - The compound of any one of claims 94-128, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X, X1, and X2 at each instance is independently CH, CRe, or N;each Re in Z1, X, X1, and X2 is independently selected from -F, methyl, and -CF3;k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6; andn is 0, 1, 2, 3, 4, 5 or 6. - The compound of any one of claims 94-129, whereink is 0, 1, or 2;m is 0, 1, or 2; andn is 0.
- The compound of any one of claims 94-130, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
- The compound of any one of claims 94-125, wherein Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene.
- The compound of any one of claims 94-125, wherein-Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;each RL is independently hydrogen, halogen, or C1-3 alkyl.k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6;n is 0, 1, 2, 3, 4, 5 or 6;q is 0, 1, 2, 3, or 4; andh is 0, 1, 2, 3, or 4. - The compound of any one of claims 94-125, or 133, wherein R8 and R9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -.
- The compound of any one of claims 94-125, 133, or 134, wherein-Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;each RL is independently hydrogen, halogen, or C1-3alkyl.k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6;n is 0, 1, 2, 3, 4, 5 or 6;q is 0, 1, 2, 3, or 4; andh is 0, 1, 2, 3, or 4. - The compound of any one of claims 94-125, or 133-135, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;each RL is independently hydrogen, halogen, or C1-3alkyl.k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6;n is 0, 1, 2, 3, 4, 5 or 6;q is 0, 1, 2, 3, or 4; andh is 0, 1, 2, 3, or 4. - The compound of any one of claims 94-125, or 133-136, whereink is 0, 1, or 2;m is 0, 1, or 2; andn is 0.
- The compound of any one of claims 94-125, or 133-137, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,each RL is independently hydrogen, halogen, or C1-3alkyl;each Re is independently selected from halogen, -OH, and methyl;q is 0, 1, or 2; andh is 0, 1, or 2. - A compound, or a pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, having the structure of Formula (IV) :
wherein,A1, A2, A3, and A4 are each independently CH or N;Lcyc is -L1cyc-, -L1cyc-L2cyc-, -L1cyc-L2cyc-L3cyc-, or -L1cyc-L2cyc-L3cyc-L4cyc-;L1cyc, L2cyc, L3cyc, and L4cyc are each independently bond, -O-, -S-, -NRy-, -C (O) -, -S (O) m1-, - (CRcRd) n1-, - (CHRc=CHRd) n1-, -C≡C-, -NRy (CRfRg) n2-, -O (CRcRd) n1O (CRfRg) n2-, -S (CRcRd) n1S (CRfRg) n2-, - (CRcRd) n1O (CRfRg) n2-, -O (CRcRd) n1NRy (CRfRg) n2-, -O (CRcRd) n1NRy (CRfRg) n2O-, - (CRcRd) n1S (CRfRg) n2-, - (CRcRd) n1 (CRfRg) n2-, - (CHRc=CHRd) n1 (CRfRg) n2-, - (CRcRd) n1 (C≡C) (CRfRg) n2-, - (CRcRd) n1C (O) (CRfRg) n2-, - (CRcRd) n1S (O) m1 (CRfRg) n2-, - (CRcRd) n1NRyC (O) (CRfRg) n2-, - (CRcRd) n1NRyS (O) m1 (CRfRg) n2-, - (CRcRd) n1C (O) NRy (CRfRg) n2-, - (CRcRd) n1S (O) (=NRy) (CRfRg) n2-, - (CRcRd) n1N=S (=O) Ry (CRfRg) n2-, or - (CRcRd) n1S (O) m1NRy (CRfRg) n2-;Rc, Rd, Rf, and Rg are each independently hydrogen, halogen, -OH, -SH, nitro, cyano, -NH2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-12 cycloalkyl, optionally substituted 3-12 membered heterocyclyl, optionally substituted C6-14 aryl, or optionally substituted 5-14 membered heteroaryl; orany two of Rc, Rd, Rf, and Rg are taken together with the atoms to which they are attached to form an optionally substituted C3-12 cycloalkyl, an optionally substituted 3-12 membered heterocyclyl, an optionally substituted C6-14 aryl group, or an optionally substituted 5 -14 membered heteroaryl;Ry is hydrogen, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-12 cycloalkyl, optionally substituted 3-12 membered heterocyclyl, optionally substituted C6-14 aryl, or optionally substituted 5-14 membered heteroaryl;n1, and n2 are each independently an integer selected from 0 to 10;m1 is 0, 1, or 2;ring B is an optionally substituted C6-14 cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring;L is -Ak1-Z1-Ak2-Z2-Ak3-*;Ak1 is - (CR8R9) k-or -O-;Ak2 is - (CR8R9) m-;Ak3 is - (CR8R9) n-;each of k, m, and n is an integer selected from 0 to 10;each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl; oreach - (CR8R9) -may independently form -C (O) -;Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-11 cycloalkylene;wherein at least one of Z1 and Z2 is heteroarylene, heterocyclene or cycloalkylene;wherein the point of bonding to the CRBN ligand is indicated by the asterisk; andU is a CRBN ligand. - The compound of claim 139, wherein,Rc, Rd, Rf, and Rg are each independently hydrogen, halogen, -OH, -SH, nitro, cyano, -NH2, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-12 cycloalkyl, optionally substituted 3-12 membered heterocyclyl, optionally substituted C6-14 aryl, or optionally substituted 5-14 membered heteroaryl; wherein the alkyl, haloalkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, are independently optionally substituted with one or more Re;orany two of Rc, Rd, Rf, and Rg are taken together with the atoms to which they are attached to form an optionally substituted C3-12 cycloalkyl group, an optionally substituted 3-12 membered heterocyclyl, an optionally substituted C6-14 aryl group, or an optionally substituted 5 -14 membered heteroaryl, wherein the cycloalkyl, heterocyclyl, aryl, and heteroaryl are independently optionally substituted with one or more Re;Ry is hydrogen, optionally substituted C1-6 alkyl, optionally substituted C1-6 haloalkyl, optionally substituted C1-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-12 cycloalkyl, optionally substituted 3-12 membered heterocyclyl, optionally substituted C6-14 aryl, or optionally substituted 5-14 membered heteroaryl, wherein the alkyl, haloalkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, are independently optionally substituted with one or more Re;ring B is an optionally substituted C6-14cycloalkyl or optionally substituted 6-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB;L is -Ak1-Z1-Ak2-Z2-Ak3-*;Ak1 is - (CR8R9) k-or -O-;Ak2 is - (CR8R9) m-;Ak3 is - (CR8R9) n-;each of k, m, and n is an integer selected from 0 to 10;each R8 and R9 are independently selected from hydrogen, halogen, -NH2, -OH, cyano, optionally substituted C1-3 alkyl, and optionally substituted C3 cycloalkyl, wherein the alkyl and cycloalkyl are each optionally substituted with one or more Re; oreach - (CR8R9) -may independently form -C (O) -;Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted 5-10-membered heteroarylene, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene, wherein the heteroaryl, heterocyclene, and cycloalkylene are independently optionally substituted with one or more Re;wherein at least one of Z1 and Z2 is heterocyclene or cycloalkylene;each Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, C1-C6 aminoalkyl, and C1-C6 heteroalkyl; ortwo Re attached to the same atom are taken together to form -C (O) -; andeach RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; ortwo RB groups attached to the same atom are taken together to form -C (O) -; ortwo RB groups attached to different atoms are taken together to form a bridge.
- The compound of claim 139 or claim 140, whereinA1 is CH.
- The compound of claim 139 or claim 140, whereinA1 is N.
- The compound of any one of claims 139-142, whereinA3 is CH.
- The compound of any one of claims 139-142, whereinA3 is N.
- The compound of any one of claims 139-144, whereinA4 is CH.
- The compound of any one of claims 139-144, whereinA4 is N.
- The compound of claim 139 or claim 140, wherein the compound has the structure of Formula (IV-1) :
- The compound of any one of claims 139-147, wherein L1cyc, L2cyc, L3cyc, and L4cyc are each independently bond, -O-, -CH2-, -OCH2-, -CH2O-, or -CH2CH2-.
- The compound of claim 139 or claim 140, wherein the compound has the structure of Formula (IV-2) :
- The compound of any one of claims 139-149, whereinA2 is CH.
- The compound of any one of claims 139-149, whereinA2 is N.
- The compound of any one of claims 139-151, wherein L1cyc is bond, -O-, -CH2-, -OCH2-, -CH2O-, or -CH2CH2-.
- The compound of any one of claims 139-152, wherein ring B is an optionally substituted C11-14 cycloalkyl or an optionally substituted 11-14 membered heterocyclyl ring, wherein the cycloalkyl or heterocyclyl ring is optionally substituted with one or more RB;each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano, wherein the alkyl and alkoxy are independently optionally substituted with one or more Re; ortwo RB groups attached to the same atom are taken together to form -C (O) -; ortwo RB groups attached to different atoms are taken together to form a bridge; andeach Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl; ortwo Re attached to the same atom are taken together to form -C (O) -.
- The compound of any one of claims 139-153, wherein ring B is a C6-14cycloalkyl ring or a 6-14 membered heterocyclyl ring selected from:
wherein,Y1 and Y2 are N or CH;B is a bridge selected from C1-3 alkylene and C1-2 heteroalkylene;each RB is independently optionally substituted straight or branched C1-6 alkyl, optionally substituted C1-3 alkoxy, halogen, -N (R7) 2, -OR7, or cyano; ortwo RB groups attached to the same atom are taken together to form -C (O) -;j is 0, 1, 2, 3, or 4; ando and p are each independently 1 or 2. - The compound of claim 154, wherein ring B is
- The compound of claim 154, wherein ring B is
- The compound of claim 154, wherein ring B is
- The compound of any one of claims 140-157, wherein each RB is independently optionally substituted straight or branched C1-3 alkyl or halogen.
- The compound of any one of claims 140-158, wherein each RB is independently methyl or fluoro.
- The compound of any one of claims 154, or 157-159, wherein B is C1-3 alkylene.
- The compound of any one of claims 154, or 157-159, wherein B is C1-2 heteroalkylene.
- The compound of claim 154, wherein ring B is selected from:
- The compound of any one of claims 154-162, wherein each RB is independently methyl or fluoro; and j is 0, 1, or 2.
- The compound of any one of claims 139-163, wherein wherein-Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X, X1, and X2 at each instance is independently CH, CRe, or N;each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6; andn is 0, 1, 2, 3, 4, 5 or 6; andq is 0, 1, 2, 3, or 4. - The compound of any one of claims 139-164, wherein R8 and R9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -.
- The compound of any one of claims 139-165, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X, X1, and X2 at each instance is independently CH, CRe, or N;each Re in Z1, X, X1, and X2 is independently selected from halogen, -NH2, -OH, C1-3 alkyl, and -CF3;k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6; andn is 0, 1, 2, 3, 4, 5 or 6. - The compound of any one of claims 139-166, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X, X1, and X2 at each instance is independently CH, CRe, or N;each Re in Z1, X, X1, and X2 is independently selected from -F, methyl, and -CF3;k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6; andn is 0, 1, 2, 3, 4, 5 or 6. - The compound of any one of claims 139-167, whereink is 0, 1, or 2;m is 0, 1, or 2; andn is 0.
- The compound of any one of claims 139-168, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
- The compound of any one of claims 139-163, wherein Z1 and Z2 are each independently selected from bond, -O-, -NR7-, -S-, optionally substituted pyrazole, optionally substituted 4, 5, 6, 7-tetrahydropyrazolo [1, 5-a] pyrazine, optionally substituted 4, 5, 6, 7-tetrahydro-1H-pyrazolo [4, 3-c] pyridine, optionally substituted 4-11 membered heterocyclene, and optionally substituted C4-6 cycloalkylene.
- The compound of any one of claims 139-163, wherein-Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;each RL is independently hydrogen, halogen, or C1-3 alkyl.k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6;n is 0, 1, 2, 3, 4, 5 or 6;q is 0, 1, 2, 3, or 4; andh is 0, 1, 2, 3, or 4. - The compound of any one of claims 139-163, or 171, wherein R8 and R9 are each independently selected from hydrogen, methyl, and halogen; or each - (CR8R9) -may independently form -C (O) -.
- The compound of any one of claims 139-163, 171, or 172, wherein-Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;each RL is independently hydrogen, halogen, or C1-3alkyl.k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6;n is 0, 1, 2, 3, 4, 5 or 6;q is 0, 1, 2, 3, or 4; andh is 0, 1, 2, 3, or 4. - The compound of any one of claims 139-163, or 171-173, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,X1, X2, X3, and X4 at each instance is independently CH, CRe, or N ;each Re in Z1, Z2, X1, X2, X3, and X4 is independently selected from halogen, -NH2, -OH, cyano, C1-3 alkyl, and C1-3 haloalkyl;each RL is independently hydrogen, halogen, or C1-3alkyl.k is 0, 1, 2, 3, 4, 5, or 6;m is 0, 1, 2, 3, 4, 5 or 6;n is 0, 1, 2, 3, 4, 5 or 6;q is 0, 1, 2, 3, or 4; andh is 0, 1, 2, 3, or 4. - The compound of any one of claims 139-163, or 171-174, whereink is 0, 1, or 2;m is 0, 1, or 2; andn is 0.
- The compound of any one of claims 139-163, or 171-175, wherein -Ak1-Z1-Ak2-Z2-Ak3-*has a structure selected from:
wherein,each RL is independently hydrogen, halogen, or C1-3alkyl;each Re is independently selected from halogen, -OH, and methyl;q is 0, 1, or 2; andh is 0, 1, or 2. - The compound of any one of claims 1-176, whereinU is a CRBN ligand having the structure
wherein,Z3 is -CH2-, -C (O) -, -C (S) -, =CH-, =N-, or -NH-;Z4 iseachrepresents a single or double bond;ring A is selected from:
R60 and R61 are each independently hydrogen, halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl;or R60 and R61 are taken together to form -C (O) -;each R62 is independently halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl;R63 is O, S, or NR70;g is 0, 1, 2 or 3;X5, X6, and X7 are each independently -N= or =CR64-;R64 is hydrogen, halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl; andeach R70 is independently hydrogen or optionally substituted C1-6 alkyl; ortwo R70 together with the nitrogen atom to which they are attached may form an optionally substituted 3-6 membered heterocyclyl ring;wherein the **indicates the attachment point to -L-. - The compound of claim 177, whereinR60 and R61 are each independently hydrogen, halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl, wherein the alkyl is optionally substituted with one or more Re;or R60 and R61 are taken together to form -C (O) -;each R62 is independently halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl, wherein the alkyl is optionally substituted with one or more Re;R64 is hydrogen, halogen, cyano, -OR70, -SR70, -S (O) R70, -S (O) 2R70, -N (R70) 2, or an optionally substituted C1-3 alkyl, wherein the alkyl is optionally substituted with one or more Re; andeach Re is independently selected from halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, - N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, and C1-6 heteroalkyl; ortwo Re attached to the same atom are taken together to form -C (O) -.
- The compound of claim 177 or 178, wherein Z3 is -CH2-, -C (O) -, -C (S) -, =CH-, or =N-.
- The compound of any one of claims 177-179, wherein Z3 is -C (O) -, -C (S) -, or =N-.
- The compound of any one of claims 177-180, wherein Z3 is -C (O) -or =N-.
- The compound of any one of claims 177-181, whereinis
- The compound of any one of claims 177-182, wherein R60 and R61 are each hydrogen, or R60 and R61 are taken together to form -C (O) -.
- The compound of any one of claims 177-183, wherein each R62 is independently halogen, -OR70, or an optionally substituted C1-3 alkyl.
- The compound of any one of claims 177-184, wherein R63 is O.
- The compound of any one of claims 177-185, wherein R64 is hydrogen, halogen, -OR70, or an optionally substituted C1-3 alkyl.
- The compound of any one of claims 177-186, wherein ring A is
- The compound of any one of claims 177-186, wherein ring A is
- The compound of any one of claims 177-186, wherein ring A is
- The compound of any one of claims 177-186, wherein ring A is
- The compound of any one of claims 177-186, wherein ring A is
- The compound of any one of claims 177-186, wherein the CRBN ligand has a structure selected from:
- The compound of claim 192, whereinis
- The compound of claim 192, whereinis
- The compound of claim 192, whereinis
- The compound of claim 192, whereinis
- The compound of claim 192, whereinis
- The compound of claim 192, whereinis
- The compound of any one of claims 177-198, wherein g is 0, 1, or 2.
- The compound of any one of claims 177-199, wherein g is 0 or 1.
- The compound of any one of claims 177-200, wherein g is 0.
- The compound of nay one of claims 177-200, wherein g is 1.
- The compound of any one of claims 177-202, wherein each R62 is independently halogen, -OR70, or C1-3 alkyl optionally substituted with halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, or C1-6 heteroalkyl.
- The compound of any one of claims 177-203, wherein each R62 is independently halogen, -OMe, -OEt, -CH2F2, -CF3, or methyl.
- The compound of any one of claims 177-204, wherein R64 is hydrogen, halogen, -OR70, or C1-3 alkyl optionally substituted with halogen, -CN, -OH, -OCH3, -S (O) CH3, -S (O) 2CH3, -S (O) 2NH2, -S (O) 2NHCH3, -S (O) 2N (CH3) 2, -NH2, -NHCH3, -N (CH3) 2, -C (O) CH3, -C (O) OH, -C (O) OCH3, C1-6 alkyl, C1-6 haloalkyl, C1-6 hydroxyalkyl, C1-6 aminoalkyl, or C1-6 heteroalkyl.
- The compound of any one of claims 177-205, wherein R64 is hydrogen, halogen, -OMe, or methyl.
- The compound of any one of claims 177-206, wherein each R62 is independently -F, -Cl, -OMe, or methyl; and each R64 is independently hydrogen, -F, -Cl, -OMe, or methyl.
- The compound of claim 1, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, selected from Table 1.
- The compound of claim 52, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, selected from Table 2.
- The compound of claim 94, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, selected from Table 3.
- The compound of claim 139, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, selected from Table 4.
- A pharmaceutical composition comprising a compound of Formula (I) , or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, as described in any one of claims 1-51 or 177-208.
- A pharmaceutical composition comprising a compound of Formula (II) , or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, as described in any one of claims 52-93, 177-207, or 209.
- A pharmaceutical composition comprising a compound of Formula (III) , or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, as described in any one of claims 94-138, 177-207, or 210.
- A pharmaceutical composition comprising a compound of Formula (IV) , or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, as described in any one of claims 139-207, or 211.
- A compound of any one of claims 1-211 or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, for use in a method of treatment of a human or animal body.
- A compound of any one of claims 1-211, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, for use in a method of treatment of cancer or neoplastic disease.
- Use of a compound of any one of claims 1-211, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for the treatment of cancer or neoplastic disease.
- A method of treating cancer in a patient in need thereof comprising administering to the patient a compound of Formula (I) as described in any one of claims 1-51 or 177-208, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof.
- A method of treating cancer in a patient in need thereof comprising administering to the patient a compound of Formula (II) as described in any one of claims 52-93, 177-207, or 209, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof.
- A method of treating cancer in a patient in need thereof comprising administering to the patient a compound of Formula (III) as described in any one of claims 94-138, 177-207, or 210, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof.
- A method of treating cancer in a patient in need thereof comprising administering to the patient a compound of Formula (IV) as described in any one of claims 139-207, or 211, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof.
- A method of treating cancer in a patient in need thereof comprising administering to the patient a pharmaceutical composition comprising a compound of Formula (I) as described in any one of claims 1-51 or 177-208, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, and a pharmaceutically acceptable excipient.
- A method of treating cancer in a patient in need thereof comprising administering to the patient a pharmaceutical composition comprising a compound of Formula (II) as described in any one of claims 52-93, 177-207, or 209, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, and a pharmaceutically acceptable excipient.
- A method of treating cancer in a patient in need thereof comprising administering to the patient a pharmaceutical composition comprising a compound of Formula (III) as described in any one of claims 94-138, 177-207, or 210, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, and a pharmaceutically acceptable excipient.
- A method of treating cancer in a patient in need thereof comprising administering to the patient a pharmaceutical composition comprising a compound of Formula (IV) as described in any one of claims 139-207, or 211, or pharmaceutically acceptable salt, solvate, prodrug, stereoisomer, or tautomer thereof, and a pharmaceutically acceptable excipient.
Applications Claiming Priority (6)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN2023070066 | 2023-01-03 | ||
CNPCT/CN2023/070066 | 2023-01-03 | ||
CNPCT/CN2023/083831 | 2023-03-24 | ||
CN2023083831 | 2023-03-24 | ||
CNPCT/CN2023/113732 | 2023-08-18 | ||
CN2023113732 | 2023-08-18 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024146502A1 true WO2024146502A1 (en) | 2024-07-11 |
Family
ID=91803583
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2024/070070 WO2024146502A1 (en) | 2023-01-03 | 2024-01-02 | Wee1 degrading compounds |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024146502A1 (en) |
Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN113402520A (en) * | 2020-03-16 | 2021-09-17 | 中国科学院上海药物研究所 | WEE1 protein degrader |
WO2021185291A1 (en) * | 2020-03-17 | 2021-09-23 | 南京明德新药研发有限公司 | Proteolysis regulator and method for using same |
US20210403451A1 (en) * | 2018-10-26 | 2021-12-30 | Wenyuan Qian | Pyrimidopyrazolone derivative as wee1 inhibitor and use thereof |
WO2022251224A1 (en) * | 2021-05-28 | 2022-12-01 | Recurium Ip Holdings, Llc | Wee1 inhibitors and methods for treating cancer |
WO2023083194A1 (en) * | 2021-11-09 | 2023-05-19 | 杭州格博生物医药有限公司 | Wee1 protein kinase degradation agent and use thereof |
WO2023125944A1 (en) * | 2021-12-31 | 2023-07-06 | 正大天晴药业集团股份有限公司 | Compound containing heterocyclic ring |
CN116829570A (en) * | 2022-04-08 | 2023-09-29 | 北京星奇原生物科技有限公司 | WEE1 degrading compounds |
-
2024
- 2024-01-02 WO PCT/CN2024/070070 patent/WO2024146502A1/en unknown
Patent Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20210403451A1 (en) * | 2018-10-26 | 2021-12-30 | Wenyuan Qian | Pyrimidopyrazolone derivative as wee1 inhibitor and use thereof |
CN113402520A (en) * | 2020-03-16 | 2021-09-17 | 中国科学院上海药物研究所 | WEE1 protein degrader |
WO2021185291A1 (en) * | 2020-03-17 | 2021-09-23 | 南京明德新药研发有限公司 | Proteolysis regulator and method for using same |
WO2022251224A1 (en) * | 2021-05-28 | 2022-12-01 | Recurium Ip Holdings, Llc | Wee1 inhibitors and methods for treating cancer |
WO2023083194A1 (en) * | 2021-11-09 | 2023-05-19 | 杭州格博生物医药有限公司 | Wee1 protein kinase degradation agent and use thereof |
WO2023125944A1 (en) * | 2021-12-31 | 2023-07-06 | 正大天晴药业集团股份有限公司 | Compound containing heterocyclic ring |
CN116829570A (en) * | 2022-04-08 | 2023-09-29 | 北京星奇原生物科技有限公司 | WEE1 degrading compounds |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US10125134B2 (en) | KRas G12C inhibitors | |
AU2017346516B2 (en) | Piperidine derivatives as inhibitors of ubiquitin specific protease 7 | |
CN115448923B (en) | Pyrimidine-fused ring compound, preparation method and application thereof | |
EP4182313A1 (en) | Kras g12d inhibitors | |
EP3697784B9 (en) | Imidazo[4,5-b]pyridine compounds and pharmaceutical compositions thereof for the treatment of inflammatory disorders | |
KR20200120713A (en) | Pyrimidine-fused ring compound and its preparation method and use | |
WO2022221528A2 (en) | Kras g12c inhibitors | |
EA028756B1 (en) | Fused heterocyclic compounds as protein kinase inhibitors | |
EP2629777A2 (en) | Bicyclic diamines as janus kinase inhibitors | |
AU2021264916A1 (en) | Degradation of Bruton's tyrosine kinase (BTK) by conjugation of BTK inhibitors with E3 ligase ligand and methods of use | |
AU2020304472A1 (en) | Pyrazolone and pyrimidine compound, and preparation method and use therefor | |
WO2024103069A1 (en) | COMPOUNDS AND COMPOSITIONS AS eIF4E INHIBITORS AND USES THEREOF | |
WO2024146502A1 (en) | Wee1 degrading compounds | |
WO2024054512A1 (en) | Akt1 modulators | |
RU2785126C2 (en) | New compounds and their pharmaceutical compositions for treatment of inflammatory diseases | |
WO2024107565A1 (en) | Akt1 modulators | |
WO2025106482A1 (en) | Compounds and compositions as smarca2/4 degraders and uses thereof | |
WO2024102621A1 (en) | Akt1 modulators | |
WO2024026061A1 (en) | Compounds for treating huntington's disease | |
WO2024020419A1 (en) | Aza-quinazoline compounds and methods of use | |
WO2024020380A1 (en) | Quinazoline compounds and methods of use | |
WO2024238221A1 (en) | Substituted quinoxalines | |
WO2024229121A1 (en) | Compounds for treating cancer | |
WO2024216229A1 (en) | Novel inhibitors of pikfyve and methods using same |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24738435 Country of ref document: EP Kind code of ref document: A1 |