WO2022262782A1 - 戊二酰亚胺取代的异噁唑稠环化合物及其应用 - Google Patents
戊二酰亚胺取代的异噁唑稠环化合物及其应用 Download PDFInfo
- Publication number
- WO2022262782A1 WO2022262782A1 PCT/CN2022/099015 CN2022099015W WO2022262782A1 WO 2022262782 A1 WO2022262782 A1 WO 2022262782A1 CN 2022099015 W CN2022099015 W CN 2022099015W WO 2022262782 A1 WO2022262782 A1 WO 2022262782A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- compound
- reaction
- ethyl acetate
- added
- reduced pressure
- Prior art date
Links
- 150000001875 compounds Chemical class 0.000 title claims abstract description 402
- -1 Glutarimide-substituted isoxazole Chemical class 0.000 title abstract description 75
- 150000003839 salts Chemical class 0.000 claims abstract description 27
- 229940079593 drug Drugs 0.000 claims description 21
- 239000003814 drug Substances 0.000 claims description 21
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 claims description 13
- 125000004432 carbon atom Chemical group C* 0.000 claims description 10
- 102000004169 proteins and genes Human genes 0.000 claims description 10
- 108090000623 proteins and genes Proteins 0.000 claims description 10
- 125000006570 (C5-C6) heteroaryl group Chemical group 0.000 claims description 9
- 125000000592 heterocycloalkyl group Chemical group 0.000 claims description 9
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 8
- 125000000753 cycloalkyl group Chemical group 0.000 claims description 7
- 229910052760 oxygen Inorganic materials 0.000 claims description 7
- 229910052739 hydrogen Inorganic materials 0.000 claims description 6
- 125000004076 pyridyl group Chemical group 0.000 claims description 5
- 125000001072 heteroaryl group Chemical group 0.000 claims description 4
- 229910052794 bromium Inorganic materials 0.000 claims description 3
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 608
- 238000006243 chemical reaction Methods 0.000 description 309
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 215
- 239000000243 solution Substances 0.000 description 202
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 198
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 176
- 238000003786 synthesis reaction Methods 0.000 description 162
- 230000015572 biosynthetic process Effects 0.000 description 161
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 154
- 239000012074 organic phase Substances 0.000 description 140
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 140
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-Dimethylformamide Chemical compound CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 132
- 229910052757 nitrogen Inorganic materials 0.000 description 124
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 110
- 239000000706 filtrate Substances 0.000 description 109
- 238000005481 NMR spectroscopy Methods 0.000 description 108
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical class O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 93
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 87
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 78
- 239000003208 petroleum Substances 0.000 description 76
- 239000012065 filter cake Substances 0.000 description 75
- 210000004027 cell Anatomy 0.000 description 70
- 239000011541 reaction mixture Substances 0.000 description 67
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 66
- 239000012043 crude product Substances 0.000 description 63
- 238000004440 column chromatography Methods 0.000 description 61
- 239000002904 solvent Substances 0.000 description 61
- 239000003480 eluent Substances 0.000 description 59
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-Diisopropylethylamine (DIPEA) Chemical compound CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 0.000 description 46
- QCQCHGYLTSGIGX-GHXANHINSA-N 4-[[(3ar,5ar,5br,7ar,9s,11ar,11br,13as)-5a,5b,8,8,11a-pentamethyl-3a-[(5-methylpyridine-3-carbonyl)amino]-2-oxo-1-propan-2-yl-4,5,6,7,7a,9,10,11,11b,12,13,13a-dodecahydro-3h-cyclopenta[a]chrysen-9-yl]oxy]-2,2-dimethyl-4-oxobutanoic acid Chemical compound N([C@@]12CC[C@@]3(C)[C@]4(C)CC[C@H]5C(C)(C)[C@@H](OC(=O)CC(C)(C)C(O)=O)CC[C@]5(C)[C@H]4CC[C@@H]3C1=C(C(C2)=O)C(C)C)C(=O)C1=CN=CC(C)=C1 QCQCHGYLTSGIGX-GHXANHINSA-N 0.000 description 39
- 239000008346 aqueous phase Substances 0.000 description 39
- 239000012141 concentrate Substances 0.000 description 39
- 238000003756 stirring Methods 0.000 description 36
- 239000000203 mixture Substances 0.000 description 34
- YLQBMQCUIZJEEH-UHFFFAOYSA-N tetrahydrofuran Natural products C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 description 33
- BWHMMNNQKKPAPP-UHFFFAOYSA-L potassium carbonate Chemical compound [K+].[K+].[O-]C([O-])=O BWHMMNNQKKPAPP-UHFFFAOYSA-L 0.000 description 32
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 30
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 30
- 239000012634 fragment Substances 0.000 description 28
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 27
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 26
- 238000012360 testing method Methods 0.000 description 26
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 24
- 230000002378 acidificating effect Effects 0.000 description 23
- BZLVMXJERCGZMT-UHFFFAOYSA-N Methyl tert-butyl ether Chemical compound COC(C)(C)C BZLVMXJERCGZMT-UHFFFAOYSA-N 0.000 description 22
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 21
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 21
- 239000012071 phase Substances 0.000 description 21
- SCVFZCLFOSHCOH-UHFFFAOYSA-M potassium acetate Chemical compound [K+].CC([O-])=O SCVFZCLFOSHCOH-UHFFFAOYSA-M 0.000 description 20
- 238000002953 preparative HPLC Methods 0.000 description 19
- 238000012054 celltiter-glo Methods 0.000 description 18
- 229910000027 potassium carbonate Inorganic materials 0.000 description 17
- 239000005457 ice water Substances 0.000 description 16
- 238000000034 method Methods 0.000 description 16
- 238000000926 separation method Methods 0.000 description 16
- LPNYRYFBWFDTMA-UHFFFAOYSA-N potassium tert-butoxide Chemical compound [K+].CC(C)(C)[O-] LPNYRYFBWFDTMA-UHFFFAOYSA-N 0.000 description 15
- WTDHULULXKLSOZ-UHFFFAOYSA-N Hydroxylamine hydrochloride Chemical compound Cl.ON WTDHULULXKLSOZ-UHFFFAOYSA-N 0.000 description 14
- 206010028980 Neoplasm Diseases 0.000 description 14
- 230000005764 inhibitory process Effects 0.000 description 14
- 206010060862 Prostate cancer Diseases 0.000 description 13
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 13
- IMNFDUFMRHMDMM-UHFFFAOYSA-N N-Heptane Chemical compound CCCCCCC IMNFDUFMRHMDMM-UHFFFAOYSA-N 0.000 description 12
- 239000007787 solid Substances 0.000 description 12
- QAOWNCQODCNURD-UHFFFAOYSA-N sulfuric acid Substances OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 12
- LWIHDJKSTIGBAC-UHFFFAOYSA-K tripotassium phosphate Chemical compound [K+].[K+].[K+].[O-]P([O-])([O-])=O LWIHDJKSTIGBAC-UHFFFAOYSA-K 0.000 description 12
- NLXLAEXVIDQMFP-UHFFFAOYSA-N Ammonia chloride Chemical class [NH4+].[Cl-] NLXLAEXVIDQMFP-UHFFFAOYSA-N 0.000 description 11
- 239000002585 base Substances 0.000 description 11
- 229920006395 saturated elastomer Polymers 0.000 description 11
- 125000001424 substituent group Chemical group 0.000 description 11
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 10
- 230000000694 effects Effects 0.000 description 10
- 239000012046 mixed solvent Substances 0.000 description 10
- 235000011056 potassium acetate Nutrition 0.000 description 10
- 235000017557 sodium bicarbonate Nutrition 0.000 description 10
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 10
- 239000000126 substance Substances 0.000 description 10
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 9
- HRPVXLWXLXDGHG-UHFFFAOYSA-N Acrylamide Chemical compound NC(=O)C=C HRPVXLWXLXDGHG-UHFFFAOYSA-N 0.000 description 9
- WMFOQBRAJBCJND-UHFFFAOYSA-M Lithium hydroxide Chemical compound [Li+].[OH-] WMFOQBRAJBCJND-UHFFFAOYSA-M 0.000 description 9
- 241000699670 Mus sp. Species 0.000 description 9
- 241000700159 Rattus Species 0.000 description 9
- 102000001307 androgen receptors Human genes 0.000 description 9
- 108010080146 androgen receptors Proteins 0.000 description 9
- 125000004429 atom Chemical group 0.000 description 9
- 238000004113 cell culture Methods 0.000 description 9
- RAXXELZNTBOGNW-UHFFFAOYSA-N imidazole Natural products C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 description 9
- NLKNQRATVPKPDG-UHFFFAOYSA-M potassium iodide Chemical compound [K+].[I-] NLKNQRATVPKPDG-UHFFFAOYSA-M 0.000 description 9
- 239000012321 sodium triacetoxyborohydride Substances 0.000 description 9
- VMHLLURERBWHNL-UHFFFAOYSA-M Sodium acetate Chemical compound [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 description 8
- 239000002253 acid Substances 0.000 description 8
- 239000006143 cell culture medium Substances 0.000 description 8
- 239000001632 sodium acetate Substances 0.000 description 8
- 235000017281 sodium acetate Nutrition 0.000 description 8
- 229910052938 sodium sulfate Inorganic materials 0.000 description 8
- GEHJYWRUCIMESM-UHFFFAOYSA-L sodium sulfite Chemical compound [Na+].[Na+].[O-]S([O-])=O GEHJYWRUCIMESM-UHFFFAOYSA-L 0.000 description 8
- 235000011152 sodium sulphate Nutrition 0.000 description 8
- 239000012224 working solution Substances 0.000 description 8
- CYPYTURSJDMMMP-WVCUSYJESA-N (1e,4e)-1,5-diphenylpenta-1,4-dien-3-one;palladium Chemical compound [Pd].[Pd].C=1C=CC=CC=1\C=C\C(=O)\C=C\C1=CC=CC=C1.C=1C=CC=CC=1\C=C\C(=O)\C=C\C1=CC=CC=C1.C=1C=CC=CC=1\C=C\C(=O)\C=C\C1=CC=CC=C1 CYPYTURSJDMMMP-WVCUSYJESA-N 0.000 description 7
- WSLDOOZREJYCGB-UHFFFAOYSA-N 1,2-Dichloroethane Chemical compound ClCCCl WSLDOOZREJYCGB-UHFFFAOYSA-N 0.000 description 7
- 210000004369 blood Anatomy 0.000 description 7
- 239000008280 blood Substances 0.000 description 7
- 230000004663 cell proliferation Effects 0.000 description 7
- 239000012295 chemical reaction liquid Substances 0.000 description 7
- IEJIGPNLZYLLBP-UHFFFAOYSA-N dimethyl carbonate Chemical compound COC(=O)OC IEJIGPNLZYLLBP-UHFFFAOYSA-N 0.000 description 7
- 229940086542 triethylamine Drugs 0.000 description 7
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 6
- VSCWAEJMTAWNJL-UHFFFAOYSA-K aluminium trichloride Chemical compound Cl[Al](Cl)Cl VSCWAEJMTAWNJL-UHFFFAOYSA-K 0.000 description 6
- 229910002091 carbon monoxide Inorganic materials 0.000 description 6
- 230000003833 cell viability Effects 0.000 description 6
- 239000003153 chemical reaction reagent Substances 0.000 description 6
- 238000001514 detection method Methods 0.000 description 6
- PQVSTLUFSYVLTO-UHFFFAOYSA-N ethyl n-ethoxycarbonylcarbamate Chemical compound CCOC(=O)NC(=O)OCC PQVSTLUFSYVLTO-UHFFFAOYSA-N 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- 230000002401 inhibitory effect Effects 0.000 description 6
- 125000005647 linker group Chemical group 0.000 description 6
- 239000007788 liquid Substances 0.000 description 6
- 229940040692 lithium hydroxide monohydrate Drugs 0.000 description 6
- GLXDVVHUTZTUQK-UHFFFAOYSA-M lithium hydroxide monohydrate Substances [Li+].O.[OH-] GLXDVVHUTZTUQK-UHFFFAOYSA-M 0.000 description 6
- 239000011259 mixed solution Substances 0.000 description 6
- 239000012299 nitrogen atmosphere Substances 0.000 description 6
- NROKBHXJSPEDAR-UHFFFAOYSA-M potassium fluoride Chemical compound [F-].[K+] NROKBHXJSPEDAR-UHFFFAOYSA-M 0.000 description 6
- 229910000160 potassium phosphate Inorganic materials 0.000 description 6
- 235000011009 potassium phosphates Nutrition 0.000 description 6
- FVAUCKIRQBBSSJ-UHFFFAOYSA-M sodium iodide Chemical compound [Na+].[I-] FVAUCKIRQBBSSJ-UHFFFAOYSA-M 0.000 description 6
- JQWHASGSAFIOCM-UHFFFAOYSA-M sodium periodate Chemical compound [Na+].[O-]I(=O)(=O)=O JQWHASGSAFIOCM-UHFFFAOYSA-M 0.000 description 6
- 239000000758 substrate Substances 0.000 description 6
- 239000000872 buffer Substances 0.000 description 5
- 229910052799 carbon Inorganic materials 0.000 description 5
- 239000012091 fetal bovine serum Substances 0.000 description 5
- 125000000524 functional group Chemical group 0.000 description 5
- 239000000463 material Substances 0.000 description 5
- XBXCNNQPRYLIDE-UHFFFAOYSA-M n-tert-butylcarbamate Chemical compound CC(C)(C)NC([O-])=O XBXCNNQPRYLIDE-UHFFFAOYSA-M 0.000 description 5
- 238000000746 purification Methods 0.000 description 5
- 239000011734 sodium Substances 0.000 description 5
- 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 4
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 4
- 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 4
- 229930040373 Paraformaldehyde Natural products 0.000 description 4
- 229960000583 acetic acid Drugs 0.000 description 4
- 239000003146 anticoagulant agent Substances 0.000 description 4
- 229940127219 anticoagulant drug Drugs 0.000 description 4
- 230000015556 catabolic process Effects 0.000 description 4
- 238000004587 chromatography analysis Methods 0.000 description 4
- 238000006731 degradation reaction Methods 0.000 description 4
- 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 4
- SACNIGZYDTUHKB-UHFFFAOYSA-N ditert-butyl-[2-[2,4,6-tri(propan-2-yl)phenyl]phenyl]phosphane Chemical group CC(C)C1=CC(C(C)C)=CC(C(C)C)=C1C1=CC=CC=C1P(C(C)(C)C)C(C)(C)C SACNIGZYDTUHKB-UHFFFAOYSA-N 0.000 description 4
- 238000011156 evaluation Methods 0.000 description 4
- 125000005842 heteroatom Chemical group 0.000 description 4
- 125000004435 hydrogen atom Chemical group [H]* 0.000 description 4
- 210000004731 jugular vein Anatomy 0.000 description 4
- 239000010410 layer Substances 0.000 description 4
- 238000001294 liquid chromatography-tandem mass spectrometry Methods 0.000 description 4
- 125000004433 nitrogen atom Chemical group N* 0.000 description 4
- 239000003960 organic solvent Substances 0.000 description 4
- 229920002866 paraformaldehyde Polymers 0.000 description 4
- 210000002381 plasma Anatomy 0.000 description 4
- 238000007747 plating Methods 0.000 description 4
- 238000002360 preparation method Methods 0.000 description 4
- 125000006413 ring segment Chemical group 0.000 description 4
- 229910052708 sodium Inorganic materials 0.000 description 4
- CWXPZXBSDSIRCS-UHFFFAOYSA-N tert-butyl piperazine-1-carboxylate Chemical compound CC(C)(C)OC(=O)N1CCNCC1 CWXPZXBSDSIRCS-UHFFFAOYSA-N 0.000 description 4
- WFDIJRYMOXRFFG-UHFFFAOYSA-N Acetic anhydride Chemical compound CC(=O)OC(C)=O WFDIJRYMOXRFFG-UHFFFAOYSA-N 0.000 description 3
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 3
- 241000699666 Mus <mouse, genus> Species 0.000 description 3
- FXHOOIRPVKKKFG-UHFFFAOYSA-N N,N-Dimethylacetamide Chemical compound CN(C)C(C)=O FXHOOIRPVKKKFG-UHFFFAOYSA-N 0.000 description 3
- 102000004142 Trypsin Human genes 0.000 description 3
- 108090000631 Trypsin Proteins 0.000 description 3
- WETWJCDKMRHUPV-UHFFFAOYSA-N acetyl chloride Chemical compound CC(Cl)=O WETWJCDKMRHUPV-UHFFFAOYSA-N 0.000 description 3
- 239000012346 acetyl chloride Substances 0.000 description 3
- 125000000217 alkyl group Chemical group 0.000 description 3
- 150000001412 amines Chemical class 0.000 description 3
- 235000019270 ammonium chloride Nutrition 0.000 description 3
- JFDZBHWFFUWGJE-UHFFFAOYSA-N benzonitrile Chemical compound N#CC1=CC=CC=C1 JFDZBHWFFUWGJE-UHFFFAOYSA-N 0.000 description 3
- 238000005119 centrifugation Methods 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 238000007796 conventional method Methods 0.000 description 3
- 239000008367 deionised water Substances 0.000 description 3
- 229910021641 deionized water Inorganic materials 0.000 description 3
- DGODWNOPHMXOTR-UHFFFAOYSA-N dipotassium;dioxido(dioxo)osmium;dihydrate Chemical compound O.O.[K+].[K+].[O-][Os]([O-])(=O)=O DGODWNOPHMXOTR-UHFFFAOYSA-N 0.000 description 3
- PQJJJMRNHATNKG-UHFFFAOYSA-N ethyl bromoacetate Chemical compound CCOC(=O)CBr PQJJJMRNHATNKG-UHFFFAOYSA-N 0.000 description 3
- 238000000605 extraction Methods 0.000 description 3
- 239000001257 hydrogen Substances 0.000 description 3
- 239000002609 medium Substances 0.000 description 3
- 125000002950 monocyclic group Chemical group 0.000 description 3
- 239000012452 mother liquor Substances 0.000 description 3
- XBXHCBLBYQEYTI-UHFFFAOYSA-N piperidin-4-ylmethanol Chemical compound OCC1CCNCC1 XBXHCBLBYQEYTI-UHFFFAOYSA-N 0.000 description 3
- 239000011698 potassium fluoride Substances 0.000 description 3
- 235000003270 potassium fluoride Nutrition 0.000 description 3
- 239000002244 precipitate Substances 0.000 description 3
- 230000017854 proteolysis Effects 0.000 description 3
- 235000010265 sodium sulphite Nutrition 0.000 description 3
- 230000008685 targeting Effects 0.000 description 3
- MHYGQXWCZAYSLJ-UHFFFAOYSA-N tert-butyl-chloro-diphenylsilane Chemical compound C=1C=CC=CC=1[Si](Cl)(C(C)(C)C)C1=CC=CC=C1 MHYGQXWCZAYSLJ-UHFFFAOYSA-N 0.000 description 3
- FAQYAMRNWDIXMY-UHFFFAOYSA-N trichloroborane Chemical compound ClB(Cl)Cl FAQYAMRNWDIXMY-UHFFFAOYSA-N 0.000 description 3
- LEIMLDGFXIOXMT-UHFFFAOYSA-N trimethylsilyl cyanide Chemical compound C[Si](C)(C)C#N LEIMLDGFXIOXMT-UHFFFAOYSA-N 0.000 description 3
- FTVLMFQEYACZNP-UHFFFAOYSA-N trimethylsilyl trifluoromethanesulfonate Chemical compound C[Si](C)(C)OS(=O)(=O)C(F)(F)F FTVLMFQEYACZNP-UHFFFAOYSA-N 0.000 description 3
- 239000012588 trypsin Substances 0.000 description 3
- 125000006273 (C1-C3) alkyl group Chemical group 0.000 description 2
- IBYHHJPAARCAIE-UHFFFAOYSA-N 1-bromo-2-chloroethane Chemical compound ClCCBr IBYHHJPAARCAIE-UHFFFAOYSA-N 0.000 description 2
- HZNVUJQVZSTENZ-UHFFFAOYSA-N 2,3-dichloro-5,6-dicyano-1,4-benzoquinone Chemical compound ClC1=C(Cl)C(=O)C(C#N)=C(C#N)C1=O HZNVUJQVZSTENZ-UHFFFAOYSA-N 0.000 description 2
- JWUJQDFVADABEY-UHFFFAOYSA-N 2-methyltetrahydrofuran Chemical compound CC1CCCO1 JWUJQDFVADABEY-UHFFFAOYSA-N 0.000 description 2
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 2
- VHYFNPMBLIVWCW-UHFFFAOYSA-N 4-Dimethylaminopyridine Chemical compound CN(C)C1=CC=NC=C1 VHYFNPMBLIVWCW-UHFFFAOYSA-N 0.000 description 2
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 2
- 238000006646 Dess-Martin oxidation reaction Methods 0.000 description 2
- YZCKVEUIGOORGS-OUBTZVSYSA-N Deuterium Chemical compound [2H] YZCKVEUIGOORGS-OUBTZVSYSA-N 0.000 description 2
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 2
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 2
- BELBBZDIHDAJOR-UHFFFAOYSA-N Phenolsulfonephthalein Chemical compound C1=CC(O)=CC=C1C1(C=2C=CC(O)=CC=2)C2=CC=CC=C2S(=O)(=O)O1 BELBBZDIHDAJOR-UHFFFAOYSA-N 0.000 description 2
- 239000012980 RPMI-1640 medium Substances 0.000 description 2
- 238000011579 SCID mouse model Methods 0.000 description 2
- UIIMBOGNXHQVGW-DEQYMQKBSA-M Sodium bicarbonate-14C Chemical compound [Na+].O[14C]([O-])=O UIIMBOGNXHQVGW-DEQYMQKBSA-M 0.000 description 2
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical group [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 2
- GLNADSQYFUSGOU-GPTZEZBUSA-J Trypan blue Chemical compound [Na+].[Na+].[Na+].[Na+].C1=C(S([O-])(=O)=O)C=C2C=C(S([O-])(=O)=O)C(/N=N/C3=CC=C(C=C3C)C=3C=C(C(=CC=3)\N=N\C=3C(=CC4=CC(=CC(N)=C4C=3O)S([O-])(=O)=O)S([O-])(=O)=O)C)=C(O)C2=C1N GLNADSQYFUSGOU-GPTZEZBUSA-J 0.000 description 2
- YRKCREAYFQTBPV-UHFFFAOYSA-N acetylacetone Chemical compound CC(=O)CC(C)=O YRKCREAYFQTBPV-UHFFFAOYSA-N 0.000 description 2
- XAGFODPZIPBFFR-UHFFFAOYSA-N aluminium Chemical compound [Al] XAGFODPZIPBFFR-UHFFFAOYSA-N 0.000 description 2
- 229910052782 aluminium Inorganic materials 0.000 description 2
- 125000003277 amino group Chemical group 0.000 description 2
- 230000003698 anagen phase Effects 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 230000001028 anti-proliverative effect Effects 0.000 description 2
- 230000000259 anti-tumor effect Effects 0.000 description 2
- 239000012298 atmosphere Substances 0.000 description 2
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical group [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 125000002619 bicyclic group Chemical group 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- ILAHWRKJUDSMFH-UHFFFAOYSA-N boron tribromide Chemical compound BrB(Br)Br ILAHWRKJUDSMFH-UHFFFAOYSA-N 0.000 description 2
- WTEOIRVLGSZEPR-UHFFFAOYSA-N boron trifluoride Chemical compound FB(F)F WTEOIRVLGSZEPR-UHFFFAOYSA-N 0.000 description 2
- 150000001721 carbon Chemical group 0.000 description 2
- 239000001569 carbon dioxide Substances 0.000 description 2
- 229910002092 carbon dioxide Inorganic materials 0.000 description 2
- 230000006037 cell lysis Effects 0.000 description 2
- 239000006285 cell suspension Substances 0.000 description 2
- 239000013078 crystal Substances 0.000 description 2
- 238000012136 culture method Methods 0.000 description 2
- 238000007405 data analysis Methods 0.000 description 2
- XUDOZULIAWNMIU-UHFFFAOYSA-N delta-hexenoic acid Chemical compound OC(=O)CCCC=C XUDOZULIAWNMIU-UHFFFAOYSA-N 0.000 description 2
- 238000001212 derivatisation Methods 0.000 description 2
- 229910052805 deuterium Inorganic materials 0.000 description 2
- 150000002148 esters Chemical class 0.000 description 2
- MVEAAGBEUOMFRX-UHFFFAOYSA-N ethyl acetate;hydrochloride Chemical compound Cl.CCOC(C)=O MVEAAGBEUOMFRX-UHFFFAOYSA-N 0.000 description 2
- 239000011888 foil Substances 0.000 description 2
- 239000012362 glacial acetic acid Substances 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 229920000669 heparin Polymers 0.000 description 2
- ZFGMDIBRIDKWMY-PASTXAENSA-N heparin Chemical compound CC(O)=N[C@@H]1[C@@H](O)[C@H](O)[C@@H](COS(O)(=O)=O)O[C@@H]1O[C@@H]1[C@@H](C(O)=O)O[C@@H](O[C@H]2[C@@H]([C@@H](OS(O)(=O)=O)[C@@H](O[C@@H]3[C@@H](OC(O)[C@H](OS(O)(=O)=O)[C@H]3O)C(O)=O)O[C@@H]2O)CS(O)(=O)=O)[C@H](O)[C@H]1O ZFGMDIBRIDKWMY-PASTXAENSA-N 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 239000012442 inert solvent Substances 0.000 description 2
- 238000006317 isomerization reaction Methods 0.000 description 2
- 238000002796 luminescence method Methods 0.000 description 2
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 2
- UHOVQNZJYSORNB-UHFFFAOYSA-N monobenzene Natural products C1=CC=CC=C1 UHOVQNZJYSORNB-UHFFFAOYSA-N 0.000 description 2
- 230000007935 neutral effect Effects 0.000 description 2
- 238000003305 oral gavage Methods 0.000 description 2
- 239000012285 osmium tetroxide Substances 0.000 description 2
- 229910000489 osmium tetroxide Inorganic materials 0.000 description 2
- 239000007800 oxidant agent Substances 0.000 description 2
- 230000001590 oxidative effect Effects 0.000 description 2
- 239000001301 oxygen Substances 0.000 description 2
- 125000004430 oxygen atom Chemical group O* 0.000 description 2
- PIBWKRNGBLPSSY-UHFFFAOYSA-L palladium(II) chloride Chemical compound Cl[Pd]Cl PIBWKRNGBLPSSY-UHFFFAOYSA-L 0.000 description 2
- 229960003531 phenolsulfonphthalein Drugs 0.000 description 2
- IVDFJHOHABJVEH-UHFFFAOYSA-N pinacol Chemical compound CC(C)(O)C(C)(C)O IVDFJHOHABJVEH-UHFFFAOYSA-N 0.000 description 2
- 230000002285 radioactive effect Effects 0.000 description 2
- 238000004467 single crystal X-ray diffraction Methods 0.000 description 2
- QDRKDTQENPPHOJ-UHFFFAOYSA-N sodium ethoxide Chemical compound [Na+].CC[O-] QDRKDTQENPPHOJ-UHFFFAOYSA-N 0.000 description 2
- 235000009518 sodium iodide Nutrition 0.000 description 2
- 125000003003 spiro group Chemical group 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 229910052717 sulfur Inorganic materials 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 238000003419 tautomerization reaction Methods 0.000 description 2
- DPKBAXPHAYBPRL-UHFFFAOYSA-M tetrabutylazanium;iodide Chemical compound [I-].CCCC[N+](CCCC)(CCCC)CCCC DPKBAXPHAYBPRL-UHFFFAOYSA-M 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- WJKHJLXJJJATHN-UHFFFAOYSA-N triflic anhydride Chemical compound FC(F)(F)S(=O)(=O)OS(=O)(=O)C(F)(F)F WJKHJLXJJJATHN-UHFFFAOYSA-N 0.000 description 2
- HVLLSGMXQDNUAL-UHFFFAOYSA-N triphenyl phosphite Chemical compound C=1C=CC=CC=1OP(OC=1C=CC=CC=1)OC1=CC=CC=C1 HVLLSGMXQDNUAL-UHFFFAOYSA-N 0.000 description 2
- RIOQSEWOXXDEQQ-UHFFFAOYSA-N triphenylphosphine Chemical compound C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 RIOQSEWOXXDEQQ-UHFFFAOYSA-N 0.000 description 2
- 210000004881 tumor cell Anatomy 0.000 description 2
- 230000001875 tumorinhibitory effect Effects 0.000 description 2
- 210000003462 vein Anatomy 0.000 description 2
- 238000005406 washing Methods 0.000 description 2
- 125000006704 (C5-C6) cycloalkyl group Chemical group 0.000 description 1
- 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 1
- SCYULBFZEHDVBN-UHFFFAOYSA-N 1,1-Dichloroethane Chemical compound CC(Cl)Cl SCYULBFZEHDVBN-UHFFFAOYSA-N 0.000 description 1
- LMDZBCPBFSXMTL-UHFFFAOYSA-N 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide Substances CCN=C=NCCCN(C)C LMDZBCPBFSXMTL-UHFFFAOYSA-N 0.000 description 1
- ASOKPJOREAFHNY-UHFFFAOYSA-N 1-Hydroxybenzotriazole Chemical compound C1=CC=C2N(O)N=NC2=C1 ASOKPJOREAFHNY-UHFFFAOYSA-N 0.000 description 1
- 125000001462 1-pyrrolyl group Chemical group [*]N1C([H])=C([H])C([H])=C1[H] 0.000 description 1
- 238000005160 1H NMR spectroscopy Methods 0.000 description 1
- ZWZFYMRKMLQMFL-UHFFFAOYSA-N 2-chloro-2-methylpropane piperazine-1-carboxylic acid Chemical compound C(C)(C)(C)Cl.N1(CCNCC1)C(=O)O ZWZFYMRKMLQMFL-UHFFFAOYSA-N 0.000 description 1
- BDDVAWDNVWLHDQ-UHFFFAOYSA-N 2-chloro-4-hydroxybenzonitrile Chemical compound OC1=CC=C(C#N)C(Cl)=C1 BDDVAWDNVWLHDQ-UHFFFAOYSA-N 0.000 description 1
- SVDDJQGVOFZBNX-UHFFFAOYSA-N 2-chloroethyl carbonochloridate Chemical compound ClCCOC(Cl)=O SVDDJQGVOFZBNX-UHFFFAOYSA-N 0.000 description 1
- 125000002941 2-furyl group Chemical group O1C([*])=C([H])C([H])=C1[H] 0.000 description 1
- LDSQQXKSEFZAPE-UHFFFAOYSA-N 2-piperidin-4-ylethanol Chemical compound OCCC1CCNCC1 LDSQQXKSEFZAPE-UHFFFAOYSA-N 0.000 description 1
- 125000004105 2-pyridyl group Chemical group N1=C([*])C([H])=C([H])C([H])=C1[H] 0.000 description 1
- 125000000389 2-pyrrolyl group Chemical group [H]N1C([*])=C([H])C([H])=C1[H] 0.000 description 1
- 125000000175 2-thienyl group Chemical group S1C([*])=C([H])C([H])=C1[H] 0.000 description 1
- FPQQSJJWHUJYPU-UHFFFAOYSA-N 3-(dimethylamino)propyliminomethylidene-ethylazanium;chloride Chemical compound Cl.CCN=C=NCCCN(C)C FPQQSJJWHUJYPU-UHFFFAOYSA-N 0.000 description 1
- 125000003682 3-furyl group Chemical group O1C([H])=C([*])C([H])=C1[H] 0.000 description 1
- 125000003349 3-pyridyl group Chemical group N1=C([H])C([*])=C([H])C([H])=C1[H] 0.000 description 1
- 125000001397 3-pyrrolyl group Chemical group [H]N1C([H])=C([*])C([H])=C1[H] 0.000 description 1
- 125000001541 3-thienyl group Chemical group S1C([H])=C([*])C([H])=C1[H] 0.000 description 1
- POILWHVDKZOXJZ-UHFFFAOYSA-N 4-hydroxypent-3-en-2-one Chemical compound CC(O)=CC(C)=O POILWHVDKZOXJZ-UHFFFAOYSA-N 0.000 description 1
- 125000000339 4-pyridyl group Chemical group N1=C([H])C([H])=C([*])C([H])=C1[H] 0.000 description 1
- KDDQRKBRJSGMQE-UHFFFAOYSA-N 4-thiazolyl Chemical group [C]1=CSC=N1 KDDQRKBRJSGMQE-UHFFFAOYSA-N 0.000 description 1
- CWDWFSXUQODZGW-UHFFFAOYSA-N 5-thiazolyl Chemical group [C]1=CN=CS1 CWDWFSXUQODZGW-UHFFFAOYSA-N 0.000 description 1
- RIMXEJYJXDBLIE-UHFFFAOYSA-N 6-bromohex-1-ene Chemical compound BrCCCCC=C RIMXEJYJXDBLIE-UHFFFAOYSA-N 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 229910015900 BF3 Inorganic materials 0.000 description 1
- BTBUEUYNUDRHOZ-UHFFFAOYSA-N Borate Chemical compound [O-]B([O-])[O-] BTBUEUYNUDRHOZ-UHFFFAOYSA-N 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- JGLMVXWAHNTPRF-CMDGGOBGSA-N CCN1N=C(C)C=C1C(=O)NC1=NC2=CC(=CC(OC)=C2N1C\C=C\CN1C(NC(=O)C2=CC(C)=NN2CC)=NC2=CC(=CC(OCCCN3CCOCC3)=C12)C(N)=O)C(N)=O Chemical compound CCN1N=C(C)C=C1C(=O)NC1=NC2=CC(=CC(OC)=C2N1C\C=C\CN1C(NC(=O)C2=CC(C)=NN2CC)=NC2=CC(=CC(OCCCN3CCOCC3)=C12)C(N)=O)C(N)=O JGLMVXWAHNTPRF-CMDGGOBGSA-N 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- UGFAIRIUMAVXCW-UHFFFAOYSA-N Carbon monoxide Chemical compound [O+]#[C-] UGFAIRIUMAVXCW-UHFFFAOYSA-N 0.000 description 1
- VGCXGMAHQTYDJK-UHFFFAOYSA-N Chloroacetyl chloride Chemical compound ClCC(Cl)=O VGCXGMAHQTYDJK-UHFFFAOYSA-N 0.000 description 1
- UFHFLCQGNIYNRP-VVKOMZTBSA-N Dideuterium Chemical compound [2H][2H] UFHFLCQGNIYNRP-VVKOMZTBSA-N 0.000 description 1
- BDAGIHXWWSANSR-UHFFFAOYSA-M Formate Chemical compound [O-]C=O BDAGIHXWWSANSR-UHFFFAOYSA-M 0.000 description 1
- 230000005526 G1 to G0 transition Effects 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 239000012359 Methanesulfonyl chloride Substances 0.000 description 1
- 239000007832 Na2SO4 Substances 0.000 description 1
- CTQNGGLPUBDAKN-UHFFFAOYSA-N O-Xylene Chemical compound CC1=CC=CC=C1C CTQNGGLPUBDAKN-UHFFFAOYSA-N 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical group C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 description 1
- KEAYESYHFKHZAL-UHFFFAOYSA-N Sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 1
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical class [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 1
- OKJPEAGHQZHRQV-UHFFFAOYSA-N Triiodomethane Natural products IC(I)I OKJPEAGHQZHRQV-UHFFFAOYSA-N 0.000 description 1
- YZCKVEUIGOORGS-NJFSPNSNSA-N Tritium Chemical compound [3H] YZCKVEUIGOORGS-NJFSPNSNSA-N 0.000 description 1
- YGYOQZPGFXHQMW-XEAPYIEGSA-N [(1s,5r)-3-azabicyclo[3.1.0]hexan-6-yl]methanol Chemical compound C1NC[C@@H]2C(CO)[C@@H]21 YGYOQZPGFXHQMW-XEAPYIEGSA-N 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 208000030961 allergic reaction Diseases 0.000 description 1
- BHELZAPQIKSEDF-UHFFFAOYSA-N allyl bromide Chemical compound BrCC=C BHELZAPQIKSEDF-UHFFFAOYSA-N 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 125000003118 aryl group Chemical group 0.000 description 1
- 238000011914 asymmetric synthesis Methods 0.000 description 1
- 239000012752 auxiliary agent Substances 0.000 description 1
- 230000001588 bifunctional effect Effects 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- GDTBXPJZTBHREO-UHFFFAOYSA-N bromine Chemical compound BrBr GDTBXPJZTBHREO-UHFFFAOYSA-N 0.000 description 1
- 239000007853 buffer solution Substances 0.000 description 1
- FJDQFPXHSGXQBY-UHFFFAOYSA-L caesium carbonate Chemical compound [Cs+].[Cs+].[O-]C([O-])=O FJDQFPXHSGXQBY-UHFFFAOYSA-L 0.000 description 1
- 229910000024 caesium carbonate Inorganic materials 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 239000012069 chiral reagent Substances 0.000 description 1
- HRYZWHHZPQKTII-UHFFFAOYSA-N chloroethane Chemical compound CCCl HRYZWHHZPQKTII-UHFFFAOYSA-N 0.000 description 1
- 238000001816 cooling Methods 0.000 description 1
- MPTQRFCYZCXJFQ-UHFFFAOYSA-L copper(II) chloride dihydrate Chemical compound O.O.[Cl-].[Cl-].[Cu+2] MPTQRFCYZCXJFQ-UHFFFAOYSA-L 0.000 description 1
- 238000012258 culturing Methods 0.000 description 1
- 125000004122 cyclic group Chemical group 0.000 description 1
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 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
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 230000000593 degrading effect Effects 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 125000000532 dioxanyl group Chemical group 0.000 description 1
- GPAYUJZHTULNBE-UHFFFAOYSA-N diphenylphosphine Chemical compound C=1C=CC=CC=1PC1=CC=CC=C1 GPAYUJZHTULNBE-UHFFFAOYSA-N 0.000 description 1
- 125000005883 dithianyl group Chemical group 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- HHRKFGMMAHZWIM-UHFFFAOYSA-N ethenoxyboronic acid Chemical compound OB(O)OC=C HHRKFGMMAHZWIM-UHFFFAOYSA-N 0.000 description 1
- CVYKQPLWSDHSSV-UHFFFAOYSA-N ethyl 2-(2-chloroethoxy)acetate Chemical compound CCOC(=O)COCCCl CVYKQPLWSDHSSV-UHFFFAOYSA-N 0.000 description 1
- ARFLASKVLJTEJD-UHFFFAOYSA-N ethyl 2-bromopropanoate Chemical compound CCOC(=O)C(C)Br ARFLASKVLJTEJD-UHFFFAOYSA-N 0.000 description 1
- MHYCRLGKOZWVEF-UHFFFAOYSA-N ethyl acetate;hydrate Chemical compound O.CCOC(C)=O MHYCRLGKOZWVEF-UHFFFAOYSA-N 0.000 description 1
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 1
- KTWOOEGAPBSYNW-UHFFFAOYSA-N ferrocene Chemical compound [Fe+2].C=1C=C[CH-]C=1.C=1C=C[CH-]C=1 KTWOOEGAPBSYNW-UHFFFAOYSA-N 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 239000012737 fresh medium Substances 0.000 description 1
- 125000002541 furyl group Chemical group 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- 150000002430 hydrocarbons Chemical group 0.000 description 1
- 125000002962 imidazol-1-yl group Chemical group [*]N1C([H])=NC([H])=C1[H] 0.000 description 1
- 125000002632 imidazolidinyl group Chemical group 0.000 description 1
- 125000002883 imidazolyl group Chemical group 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- XMBWDFGMSWQBCA-YPZZEJLDSA-N iodane Chemical compound [125IH] XMBWDFGMSWQBCA-YPZZEJLDSA-N 0.000 description 1
- 229940044173 iodine-125 Drugs 0.000 description 1
- INQOMBQAUSQDDS-UHFFFAOYSA-N iodomethane Chemical compound IC INQOMBQAUSQDDS-UHFFFAOYSA-N 0.000 description 1
- 230000007794 irritation Effects 0.000 description 1
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 230000000155 isotopic effect Effects 0.000 description 1
- 125000003965 isoxazolidinyl group Chemical group 0.000 description 1
- 125000000842 isoxazolyl group Chemical group 0.000 description 1
- 159000000003 magnesium salts Chemical class 0.000 description 1
- 108010082117 matrigel Proteins 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- QARBMVPHQWIHKH-UHFFFAOYSA-N methanesulfonyl chloride Chemical compound CS(Cl)(=O)=O QARBMVPHQWIHKH-UHFFFAOYSA-N 0.000 description 1
- 125000001434 methanylylidene group Chemical group [H]C#[*] 0.000 description 1
- 125000000325 methylidene group Chemical group [H]C([H])=* 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- XONPDZSGENTBNJ-UHFFFAOYSA-N molecular hydrogen;sodium Chemical compound [Na].[H][H] XONPDZSGENTBNJ-UHFFFAOYSA-N 0.000 description 1
- 125000004572 morpholin-3-yl group Chemical group N1C(COCC1)* 0.000 description 1
- 125000004573 morpholin-4-yl group Chemical group N1(CCOCC1)* 0.000 description 1
- 125000002757 morpholinyl group Chemical group 0.000 description 1
- 125000004123 n-propyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001624 naphthyl group Chemical group 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 125000002971 oxazolyl group Chemical group 0.000 description 1
- YJVFFLUZDVXJQI-UHFFFAOYSA-L palladium(ii) acetate Chemical compound [Pd+2].CC([O-])=O.CC([O-])=O YJVFFLUZDVXJQI-UHFFFAOYSA-L 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- KHIWWQKSHDUIBK-UHFFFAOYSA-N periodic acid Chemical compound OI(=O)(=O)=O KHIWWQKSHDUIBK-UHFFFAOYSA-N 0.000 description 1
- 230000003285 pharmacodynamic effect Effects 0.000 description 1
- 239000008363 phosphate buffer Substances 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- RFIOZSIHFNEKFF-UHFFFAOYSA-M piperazine-1-carboxylate Chemical compound [O-]C(=O)N1CCNCC1 RFIOZSIHFNEKFF-UHFFFAOYSA-M 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
- 229920001184 polypeptide Polymers 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 239000000047 product Substances 0.000 description 1
- VVWRJUBEIPHGQF-UHFFFAOYSA-N propan-2-yl n-propan-2-yloxycarbonyliminocarbamate Chemical compound CC(C)OC(=O)N=NC(=O)OC(C)C VVWRJUBEIPHGQF-UHFFFAOYSA-N 0.000 description 1
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 238000011865 proteolysis targeting chimera technique Methods 0.000 description 1
- 229940124823 proteolysis targeting chimeric molecule Drugs 0.000 description 1
- 125000003373 pyrazinyl group Chemical group 0.000 description 1
- 125000003072 pyrazolidinyl group Chemical group 0.000 description 1
- 125000003226 pyrazolyl group Chemical group 0.000 description 1
- 125000002098 pyridazinyl group Chemical group 0.000 description 1
- ZDYVRSLAEXCVBX-UHFFFAOYSA-N pyridinium p-toluenesulfonate Chemical compound C1=CC=[NH+]C=C1.CC1=CC=C(S([O-])(=O)=O)C=C1 ZDYVRSLAEXCVBX-UHFFFAOYSA-N 0.000 description 1
- 125000000246 pyrimidin-2-yl group Chemical group [H]C1=NC(*)=NC([H])=C1[H] 0.000 description 1
- 125000004527 pyrimidin-4-yl group Chemical group N1=CN=C(C=C1)* 0.000 description 1
- 125000000714 pyrimidinyl group Chemical group 0.000 description 1
- 125000000719 pyrrolidinyl group Chemical group 0.000 description 1
- 125000000168 pyrrolyl group Chemical group 0.000 description 1
- 230000006340 racemization Effects 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 239000000376 reactant Substances 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 239000013557 residual solvent Substances 0.000 description 1
- 229930195734 saturated hydrocarbon Natural products 0.000 description 1
- 238000013207 serial dilution Methods 0.000 description 1
- 239000002356 single layer Substances 0.000 description 1
- 108010026668 snake venom protein C activator Proteins 0.000 description 1
- 239000012312 sodium hydride Substances 0.000 description 1
- 229910000104 sodium hydride Inorganic materials 0.000 description 1
- RSIJVJUOQBWMIM-UHFFFAOYSA-L sodium sulfate decahydrate Chemical compound O.O.O.O.O.O.O.O.O.O.[Na+].[Na+].[O-]S([O-])(=O)=O RSIJVJUOQBWMIM-UHFFFAOYSA-L 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 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 1
- XKXIQBVKMABYQJ-UHFFFAOYSA-M tert-butyl carbonate Chemical compound CC(C)(C)OC([O-])=O XKXIQBVKMABYQJ-UHFFFAOYSA-M 0.000 description 1
- IOGXOCVLYRDXLW-UHFFFAOYSA-N tert-butyl nitrite Chemical compound CC(C)(C)ON=O IOGXOCVLYRDXLW-UHFFFAOYSA-N 0.000 description 1
- 239000012414 tert-butyl nitrite Substances 0.000 description 1
- 125000004192 tetrahydrofuran-2-yl group Chemical group [H]C1([H])OC([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000003718 tetrahydrofuranyl group Chemical group 0.000 description 1
- 125000001412 tetrahydropyranyl group Chemical group 0.000 description 1
- 125000003507 tetrahydrothiofenyl group Chemical group 0.000 description 1
- 125000003831 tetrazolyl group Chemical group 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 125000000335 thiazolyl group Chemical group 0.000 description 1
- 125000001544 thienyl group Chemical group 0.000 description 1
- 210000001519 tissue Anatomy 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 238000012549 training Methods 0.000 description 1
- 125000001425 triazolyl group Chemical group 0.000 description 1
- 229910052722 tritium Inorganic materials 0.000 description 1
- 230000004614 tumor growth Effects 0.000 description 1
- 230000034512 ubiquitination Effects 0.000 description 1
- 238000010798 ubiquitination Methods 0.000 description 1
- 238000010792 warming Methods 0.000 description 1
- 238000005303 weighing Methods 0.000 description 1
- 239000008096 xylene Substances 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/41—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
- A61K31/42—Oxazoles
- A61K31/423—Oxazoles condensed with carbocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/44—Non condensed pyridines; Hydrogenated derivatives thereof
- A61K31/445—Non condensed piperidines, e.g. piperocaine
- A61K31/4523—Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
- A61K31/4525—Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with oxygen as a ring hetero atom
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/44—Non condensed pyridines; Hydrogenated derivatives thereof
- A61K31/445—Non condensed piperidines, e.g. piperocaine
- A61K31/4523—Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
- A61K31/4545—Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/4965—Non-condensed pyrazines
- A61K31/497—Non-condensed pyrazines containing further heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/54—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
- A61K47/55—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D413/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
- C07D413/14—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
Definitions
- the present invention relates to a series of glutarimide-substituted isoxazole condensed ring compounds and applications thereof, in particular to compounds represented by formula (II) and pharmaceutically acceptable salts thereof.
- Protein degradation targeting chimera is a technology that uses the ubiquitin-proteasome system to target specific proteins and induce their degradation in cells.
- the ubiquitin-proteasome system is the main pathway for intracellular protein degradation. Its normal physiological function is mainly responsible for removing denatured, mutated or harmful proteins in cells. The degradation of more than 80% of proteins in cells depends on the ubiquitin-proteasome system.
- PROTACs use the cell's own protein destruction mechanism to remove specifically targeted proteins in the cell.
- the present invention provides bifunctional or proteolytically targeting chimeric (PROTAC) compounds discovered to act as modulators of targeted ubiquitination and androgen receptor (AR) degradation.
- PROTAC proteolytically targeting chimeric
- the present invention provides a compound represented by formula (II) or a pharmaceutically acceptable salt thereof,
- R 1 is selected from H and CH 3 ;
- Linker selected from Wherein, E 1 end is connected with ABM;
- R is selected from H and methyl
- E 2 is selected from single bond, -(CH 2 ) m -, and -E 5 -(CH 2 ) m -;
- E 3 is selected from -(CH 2 ) n -, -(CH 2 ) p O(CH 2 ) q -,
- E is selected from 5-6 membered heterocycloalkyl
- T and T are each independently selected from CH and N ;
- n is selected from 1, 2 and 3;
- n is selected from 1, 2, 3, 4, 5, 6 and 7;
- p and q are each independently selected from 0, 1 and 2;
- Ring Q is selected from phenyl or
- Ring A is selected from phenyl and 5-6 membered heteroaryl
- the ABM is selected from drugs or derivatives thereof that are targeted to bind to the AR protein.
- E 2 is selected from single bond, -CH 2 -, -(CH 2 ) 2 -, -(CH 2 ) 3 -, Other variables are as defined herein.
- the above-mentioned structural units -E 1 -E 2 - are selected from single bonds, -O-, -OCH 2 CH 2 -, Other variables are as defined herein.
- the above-mentioned E 3 is selected from single bond, -CH 2 -, -(CH 2 ) 2 -, -(CH 2 ) 4 -, -(CH 2 ) 5 -, Other variables are as defined herein.
- the above-mentioned structural units -E 3 -E 4 - are selected from single bonds, -CH 2 -, -(CH 2 ) 2 O-, -(CH 2 ) 5 O-, Other variables are as defined herein.
- ABM is selected from the structures of formula (ABM-1) and (ABM-2):
- R 3 and R 4 are selected from methyl
- R 3 and R 4 form a C 4-6 cycloalkyl group with the carbon atoms connected together;
- Ring B is selected from phenyl and pyridyl optionally substituted by 1, 2 or 3 R a ;
- Ring C is selected from phenyl and 6-membered heteroaryl optionally substituted by 1, 2 or 3 R b ;
- Ra is selected from F, Cl, Br, I, CN, CH 3 , CF 3 and NO 2 ;
- Rb is selected from F and Cl
- Ring D is selected from
- the above-mentioned ABM is selected from the structures of formula (ABM-1a), (ABM-2a), (ABM-2b) and (ABM-3a):
- R b1 is selected from H and F;
- t is selected from 1, 2 and 3;
- T1 is selected from CH and N ;
- R a is as defined in the present invention.
- the present invention also provides a compound represented by formula (I) or a pharmaceutically acceptable salt thereof,
- R 1 is selected from H and CH 3 ;
- Linker selected from Wherein, E 1 end is connected with ABM;
- R is selected from H and methyl
- E 2 is selected from single bond, -(CH 2 ) m -, and -E 5 -(CH 2 ) m -;
- E 3 is selected from -(CH 2 ) n -, -(CH 2 ) p O(CH 2 ) q -,
- E is selected from 5-6 membered heterocycloalkyl
- T and T are each independently selected from CH and N ;
- n is selected from 1, 2 and 3;
- n is selected from 1, 2, 3, 4, 5, 6 and 7;
- p and q are each independently selected from 0, 1 and 2;
- Ring A is selected from nonexistent, phenyl and 5-6 membered heteroaryl
- the ABM is selected from drugs or derivatives thereof that are targeted to bind to the AR protein.
- E 2 is selected from single bond, -CH 2 -, -(CH 2 ) 2 -, -(CH 2 ) 3 -, Other variables are as defined herein.
- the above-mentioned structural units -E 1 -E 2 - are selected from single bonds, -O-, -OCH 2 CH 2 -, Other variables are as defined herein.
- the above-mentioned E 3 is selected from -CH 2 -, -(CH 2 ) 2 -, -(CH 2 ) 4 -, -(CH 2 ) 5 -, Other variables are as defined herein.
- the above structural unit -E 3 -E 4 - is selected from -CH 2 -, -(CH 2 ) 2 O-, -(CH 2 ) 5 O-, Other variables are as defined herein.
- ABM is selected from the structures of formula (ABM-1) and (ABM-2):
- R 3 and R 4 are selected from methyl
- R 3 and R 4 form a C 4-6 cycloalkyl group with the carbon atoms connected together;
- Ring B is selected from phenyl and pyridyl optionally substituted by 1, 2 or 3 R a ;
- Ring C is selected from phenyl and 6-membered heteroaryl optionally substituted by 1, 2 or 3 R b ;
- Ra is selected from F, Cl, Br, I, CN, CH 3 , CF 3 and NO 2 ;
- Rb is selected from F and Cl
- Ring D is selected from
- ABM is selected from the structures of formula (ABM-1a), (ABM-2a) and (ABM-2b):
- R b1 is selected from H and F;
- t is selected from 1, 2 and 3;
- R a is as defined in the present invention.
- the present invention also provides the following compounds or pharmaceutically acceptable salts thereof:
- the compound of the present invention has excellent AR protein degradation effect, cell proliferation inhibition effect and significant antitumor effect, especially for prostate cancer, has excellent therapeutic effect, and has good druggability.
- pharmaceutically acceptable refers to those compounds, materials, compositions and/or dosage forms, which are suitable for use in contact with human and animal tissues within the scope of sound medical judgment , without undue toxicity, irritation, allergic reaction or other problems or complications, commensurate with a reasonable benefit/risk ratio.
- pharmaceutically acceptable salt refers to a salt of a compound of the present invention, which is prepared from a compound having a specific substituent found in the present invention and a relatively non-toxic acid or base.
- base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of base, either neat solution or in a suitable inert solvent.
- Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amine or magnesium salts or similar salts.
- acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the acid, either neat solution or in a suitable inert solvent.
- Certain specific compounds of the present invention contain basic and acidic functional groups and can thus be converted into either base or acid addition salts.
- the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound containing acid groups or bases by conventional chemical methods.
- such salts are prepared by reacting the free acid or base form of these compounds with a stoichiometric amount of the appropriate base or acid in water or an organic solvent or a mixture of both.
- targeting protein means a protein or polypeptide that binds a compound of the invention and is degraded.
- drug or derivative thereof includes the drug or derivative thereof that has been developed to bind to the target protein and the drug or derivative thereof that will be developed in the future to bind to the target protein.
- the compounds of the invention may exist in particular geometric or stereoisomeric forms.
- the present invention contemplates all such compounds, including cis and trans isomers, (-)- and (+)-enantiomers, (R)- and (S)-enantiomers, diastereomers isomers, (D)-isomers, (L)-isomers, and their racemic and other mixtures, such as enantiomerically or diastereomerically enriched mixtures, all of which are subject to the present within the scope of the invention.
- Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All such isomers, as well as mixtures thereof, are included within the scope of the present invention.
- enantiomer or “optical isomer” refer to stereoisomers that are mirror images of each other.
- diastereoisomer refers to stereoisomers whose molecules have two or more chiral centers and which are not mirror images of the molecules.
- keys with wedge-shaped solid lines and dotted wedge keys Indicates the absolute configuration of a stereocenter, with a straight solid-line bond and straight dashed keys Indicates the relative configuration of the stereocenter, with a wavy line Indicates wedge-shaped solid-line bond or dotted wedge key or with tilde Indicates a straight solid line key and straight dashed keys
- any one or more sites of the group can be linked to other groups through chemical bonds.
- the chemical bonds that the site connects with other groups can use straight solid line bonds Straight dotted key or tilde express.
- the straight-shaped solid-line bond in -OCH3 indicates that it is connected to other groups through the oxygen atom in the group;
- the straight dotted line bond in indicates that the two ends of the nitrogen atom in the group are connected to other groups;
- the wavy lines in indicate that the carbon atoms at positions 1 and 2 in the phenyl group are connected to other groups.
- tautomer or “tautomeric form” means that isomers with different functional groups are in dynamic equilibrium at room temperature and are rapidly interconvertible. If tautomerism is possible (eg, in solution), then chemical equilibrium of the tautomers can be achieved.
- proton tautomers also called prototropic tautomers
- proton tautomers include interconversions via migration of a proton, such as keto-enol isomerization and imine-ene Amine isomerization.
- Valence isomers (valence tautomers) involve interconversions by recombination of some bonding electrons.
- keto-enol tautomerization is the interconversion between two tautomers of pentane-2,4-dione and 4-hydroxypent-3-en-2-one.
- the terms “enriched in an isomer”, “enriched in an isomer”, “enriched in an enantiomer” or “enantiomerically enriched” refer to one of the isomers or enantiomers
- the content of the enantiomer is less than 100%, and the content of the isomer or enantiomer is greater than or equal to 60%, or greater than or equal to 70%, or greater than or equal to 80%, or greater than or equal to 90%, or greater than or equal to 95%, or Greater than or equal to 96%, or greater than or equal to 97%, or greater than or equal to 98%, or greater than or equal to 99%, or greater than or equal to 99.5%, or greater than or equal to 99.6%, or greater than or equal to 99.7%, or greater than or equal to 99.8%, or greater than or equal to 99.9%.
- the terms “isomer excess” or “enantiomeric excess” refer to the difference between the relative percentages of two isomers or two enantiomers. For example, if the content of one isomer or enantiomer is 90% and the other isomer or enantiomer is 10%, then the isomer or enantiomeric excess (ee value) is 80% .
- Optically active (R)- and (S)-isomers as well as D and L-isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If one enantiomer of a compound of the invention is desired, it can be prepared by asymmetric synthesis or derivatization with chiral auxiliary agents, wherein the resulting diastereomeric mixture is separated and the auxiliary group is cleaved to provide pure desired enantiomer.
- a diastereoisomeric salt is formed with an appropriate optically active acid or base, and then a diastereomeric salt is formed by a conventional method known in the art. Diastereomeric resolution is performed and the pure enantiomers are recovered. Furthermore, the separation of enantiomers and diastereomers is usually accomplished by the use of chromatography using chiral stationary phases, optionally in combination with chemical derivatization methods (e.g. amines to amino groups formate).
- the compounds of the present invention may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute the compounds.
- compounds may be labeled with radioactive isotopes such as tritium ( 3 H), iodine-125 ( 125 I) or C-14 ( 14 C).
- radioactive isotopes such as tritium ( 3 H), iodine-125 ( 125 I) or C-14 ( 14 C).
- heavy hydrogen can be used to replace hydrogen to form deuterated drugs.
- the bond formed by deuterium and carbon is stronger than the bond formed by ordinary hydrogen and carbon.
- deuterated drugs can reduce toxic side effects and increase drug stability. , enhance the efficacy, prolong the biological half-life of drugs and other advantages. All changes in isotopic composition of the compounds of the invention, whether radioactive or not, are included within the scope of the invention.
- substituted means that any one or more hydrogen atoms on a specified atom are replaced by a substituent, which may include deuterium and hydrogen variants, as long as the valence of the specified atom is normal and the substituted compound is stable.
- Oxygen substitution does not occur on aromatic groups.
- optionally substituted means that it may or may not be substituted, and unless otherwise specified, the type and number of substituents may be arbitrary on a chemically realizable basis.
- any variable eg, R
- its definition is independent at each occurrence.
- said group may optionally be substituted with up to two R, with independent options for each occurrence of R.
- substituents and/or variations thereof are permissible only if such combinations result in stable compounds.
- linking group When the number of a linking group is 0, such as -(CRR) 0 -, it means that the linking group is a single bond.
- substituent When a substituent is vacant, it means that the substituent does not exist. For example, when X in A-X is vacant, it means that the structure is actually A. When the enumerated substituent does not indicate which atom it is connected to the substituted group, this substituent can be bonded through any atom, for example, pyridyl as a substituent can be connected to any atom on the pyridine ring. The carbon atom is attached to the group being substituted.
- C1-3 alkyl is used to denote a straight or branched chain saturated hydrocarbon group consisting of 1 to 3 carbon atoms.
- the C 1-3 alkyl group includes C 1-2 and C 2-3 alkyl groups, etc.; it can be monovalent (such as methyl), divalent (such as methylene) or multivalent (such as methine) .
- Examples of C 1-3 alkyl include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), and the like.
- the number of atoms in a ring is generally defined as the number of ring members, eg, "5-7 membered ring” means a “ring” with 5-7 atoms arranged around it.
- C 4-6 cycloalkyl means a saturated cyclic hydrocarbon group composed of 4 to 6 carbon atoms, which includes monocyclic and bicyclic systems, wherein bicyclic systems include spiro rings, fused rings and bridge ring.
- the C 4-6 cycloalkyl group includes C 4-6 , C 4-5 or C 5-6 cycloalkyl group, etc.; it may be monovalent, divalent or multivalent.
- Examples of C 4-6 cycloalkyl include, but are not limited to, cyclobutyl, cyclopentyl, cyclohexyl and the like.
- C n-n+m or C n -C n+m includes any specific instance of n to n+m carbons, for example C 1-12 includes C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , C 10 , C 11 , and C 12 , also including any range from n to n+m, for example, C 1-12 includes C 1- 3 , C 1-6 , C 1-9 , C 3-6 , C 3-9 , C 3-12 , C 6-9 , C 6-12 , and C 9-12 etc.; similarly, n to n +m means that the number of atoms on the ring is n to n+m, for example, a 3-12-membered ring includes a 3-membered ring, a 4-membered ring, a 5-membered ring, a 6-membered ring, a 7-membered ring, an 8-membere
- the term "5-6 membered heterocycloalkyl" by itself or in combination with other terms denotes a saturated cyclic group consisting of 5 to 6 ring atoms, respectively, whose 1, 2, 3 or 4 ring atoms is a heteroatom independently selected from O, S, and N, and the rest is carbon atoms, wherein the nitrogen atom is optionally quaternized, and the nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., NO and S(O)p,p is 1 or 2). It includes monocyclic and bicyclic ring systems, wherein bicyclic ring systems include spiro, fused and bridged rings.
- a heteroatom may occupy the attachment position of the heterocycloalkyl to the rest of the molecule.
- the 5-6 membered heterocycloalkyl group includes 5-membered and 6-membered heterocycloalkyl groups.
- 5-6 membered heterocycloalkyl groups include, but are not limited to, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrothiophenyl (including tetrahydrothiophen-2-yl and tetrahydrothiophen-3-yl, etc.) , tetrahydrofuranyl (including tetrahydrofuran-2-yl, etc.), tetrahydropyranyl, piperidinyl (including 1-piperidinyl, 2-piperidinyl and 3-piperidinyl, etc.), piperazinyl (including 1 -piperazinyl and 2-piperazinyl, etc.), morpholinyl (including 3-morpholinyl and 4-morpholinyl, etc.), dioxanyl, dithianyl, isoxazolidinyl, isothiazole Alkyl, 1,2-oxazinyl, 1,2-thiazinyl, etc.
- the terms “5-6-membered heteroaryl ring” and “5-6-membered heteroaryl” in the present invention can be used interchangeably, and the term “5-6-membered heteroaryl” means that there are 5 to 6 ring atoms A monocyclic group with a conjugated ⁇ -electron system, 1, 2, 3 or 4 ring atoms are heteroatoms independently selected from O, S and N, and the rest are carbon atoms. Where the nitrogen atom is optionally quaternized, the nitrogen and sulfur heteroatoms may be optionally oxidized (ie, NO and S(O)p, where p is 1 or 2).
- the 5-6 membered heteroaryl can be attached to the rest of the molecule through a heteroatom or a carbon atom.
- the 5-6 membered heteroaryl includes 5 and 6 membered heteroaryl.
- Examples of the 5-6 membered heteroaryl groups include, but are not limited to, pyrrolyl (including N-pyrrolyl, 2-pyrrolyl and 3-pyrrolyl, etc.), pyrazolyl (including 2-pyrazolyl and 3-pyrrolyl Azolyl, etc.), imidazolyl (including N-imidazolyl, 2-imidazolyl, 4-imidazolyl and 5-imidazolyl, etc.), oxazolyl (including 2-oxazolyl, 4-oxazolyl and 5- Oxazolyl, etc.), triazolyl (1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl, 1H-1,2,4-triazolyl and 4H-1, 2,4-triazolyl, etc.
- any one or more sites of the group can be linked to other groups through chemical bonds.
- connection method of the chemical bond is not positioned, and there is an H atom at the connectable site, when the chemical bond is connected, the number of H atoms at the site will decrease correspondingly with the number of chemical bonds connected to become the corresponding valence group.
- the chemical bonds that the site connects with other groups can use straight solid line bonds Straight dotted key or tilde express.
- the straight-shaped solid-line bond in -OCH3 indicates that it is connected to other groups through the oxygen atom in the group;
- the straight dotted line bond in indicates that the two ends of the nitrogen atom in the group are connected to other groups;
- the wavy lines in indicate that the 1 and 2 carbon atoms in the phenyl group are connected to other groups;
- the compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, the embodiments formed by combining them with other chemical synthesis methods, and the methods well known to those skilled in the art Equivalent alternatives, preferred embodiments include but are not limited to the examples of the present invention.
- the structure of the compounds of the present invention can be confirmed by conventional methods known to those skilled in the art. If the present invention involves the absolute configuration of the compound, the absolute configuration can be confirmed by conventional technical means in the art. For example, in single crystal X-ray diffraction (SXRD), the cultured single crystal is collected with a Bruker D8 venture diffractometer to collect diffraction intensity data, the light source is CuK ⁇ radiation, and the scanning method is: After scanning and collecting relevant data, the absolute configuration can be confirmed by further analyzing the crystal structure by direct method (Shelxs97).
- SXRD single crystal X-ray diffraction
- the solvent used in the present invention is commercially available.
- reaction solution was lowered to room temperature, concentrated under reduced pressure to remove most of the ethanol, diluted with ice water (500mL) and dichloromethane (1L), separated into layers, the aqueous phase was extracted with dichloromethane (200mL), the organic phases were combined, and successively washed with Wash with saturated aqueous sodium bicarbonate solution (500 mL) and saturated brine (500 mL), dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure to obtain intermediate BB-1.
- reaction solution was slowly poured into saturated ammonium chloride (1200mL), extracted with ethyl acetate (200mL ⁇ 2), the organic phases were combined, washed with water (200mL ⁇ 2) and saturated brine (200mL ⁇ 2) successively, The collected organic phases were concentrated under reduced pressure.
- Step 4 Synthesis of the hydrochloride salt of intermediate BB-2
- intermediate BB-2-3 (45mg, 125.22 ⁇ mol) to ethyl acetate (2mL), slowly add hydrochloric acid/ethyl acetate solution (4M, 2mL) to the reaction solution, and replace the mixture with nitrogen for 3 times at 25°C Stir for 12 hours. After the reaction was completed, the reaction solution was concentrated under reduced pressure to obtain the hydrochloride of the intermediate BB-2.
- the intermediate BB-3-3 was dissolved in tetrahydrofuran (300 mL), dimethyl carbonate (47.05 g, 522.30 mmol, 43.97 mL) was added, the temperature was lowered to 0°C, and potassium tert-butoxide (87.91 g, 783.45 mmol) was added. Under the protection of nitrogen, the temperature was raised to 70°C and stirred for about 16 hours. After the reaction was completed, the reaction solution was slowly poured into ice water (200mL), and the pH was adjusted to 8-9 with hydrochloric acid solution (2M). 2-3, solids were precipitated, filtered, and the filter cake was washed with water (100 mL), collected and dried to obtain intermediate BB-3-4.
- 1 H NMR 400 MHz, DMSO_d 6 ) ⁇ : 12.65 (br s, 1H), 7.90 (s, 1H), 7.16 (s, 1H), 5.49 (s, 1H), 3.94 (s, 3H
- the intermediate BB-3-6 (5g, 15.92mmol) was dissolved in toluene (100mL) and water (20mL), followed by adding tert-butyl carbamate (3.73g, 31.83mmol), potassium phosphate (10.14g, 47.75mmol ), tris(dibenzylideneacetone)dipalladium (1.17g, 1.27mmol) and 2-di-tert-butylphosphino-2',4',6'-triisopropylbiphenyl (1.08g, 2.55mmol), nitrogen Replaced three times, raised the temperature to 100°C, and stirred for 36 hours. After the reaction was completed, the reaction solution was concentrated under reduced pressure to obtain a residue.
- the intermediate BB-3-8 (0.3g, 1.27mmol) was dissolved in dichloromethane (3mL), and chloroacetyl chloride (172.12mg, 1.52mmol, 121.21 ⁇ L) and triethylamine (192.76mg, 1.90 mmol, 265.15 ⁇ L), reacted for 0.5 hours, and concentrated under reduced pressure to obtain a residue.
- Xylene (3 mL) and pyridine p-toluenesulfonate (319.15 mg, 1.27 mmol) were added to the residue, the temperature was raised to 140°C, and the reaction was continued for 4 hours.
- compound BB-4-1 (10g, 78.67mmol) was dissolved in dichloromethane (120mL) and acetone (60mL), and cyanotrimethylsilane (12.45g , 125.50mmol, 15.70mL), trimethylsilyl triflate (820.00mg, 3.69mmol, 666.67 ⁇ L), and the reaction mixture was stirred at 25°C for 2 hours. After completion of the reaction, cool to 0-5°C, add water (200 mL) to dilute, and extract with ethyl acetate (200 mL ⁇ 3).
- the intermediate BB-4-2 (8 g, 45.40 mmol) was dissolved in N, N-dimethylacetamide (150 mL), 4-isothiocyanato-2-(trifluoroform Base) benzonitrile (10.36g, 45.40mmol) was added to the reaction solution in batches, the reaction mixture was stirred at 25°C for 12 hours, methanol (60mL), dilute hydrochloric acid (2M, 60mL) were added, and the reaction mixture was stirred at 70°C for 3 hours , After the reaction was completed, it was cooled to room temperature, diluted with water (500 mL), and extracted with ethyl acetate (200 mL ⁇ 3).
- the intermediate BB-4-3 (2g, 4.72mmol) was dissolved in N,N-dimethylformamide (50mL), and tert-butyl-4-(2-bromoethyl Base) piperazine-1-carboxylate (1.66g, 5.67mmol), potassium carbonate (1.31g, 9.45mmol), potassium iodide (784.17mg, 4.72mmol), and the reaction mixture was heated to 80°C and stirred for 12 hours. After the reaction was completed, it was cooled to room temperature, diluted with water (100 mL), and extracted with ethyl acetate (100 mL ⁇ 3).
- Step 4 Synthesis of the hydrochloride salt of intermediate BB-4-5
- the intermediate BB-4-4 (2.0 g, 3.15 mmol) was dissolved in ethyl acetate (50 mL), hydrochloric acid/ethyl acetate (4M, 3.93 mL) was slowly added dropwise to the reaction mixture, The reaction mixture was stirred at 25°C for 12 hours. After the reaction was completed, the reaction mixture was concentrated under reduced pressure to obtain the hydrochloride salt of intermediate BB-4-5.
- the intermediate BB-4-5 (1.5g, 2.62mmol, hydrochloride) was dissolved in acetonitrile (50mL), and ethyl bromoacetate (875.85mg, 5.24mmol, 580.04 ⁇ L) was added successively, Potassium carbonate (724.86mg, 5.24mmol), the reaction mixture was heated to 80°C and stirred for 5 hours. After the reaction was completed, it was cooled to room temperature, diluted with water (100mL), and extracted with ethyl acetate (100mL ⁇ 3).
- the intermediate BB-4-6 (1.5g, 2.42mmol) was dissolved in ethanol (50mL), lithium hydroxide (1M, 7.25mL) was slowly added dropwise to the reaction mixture, and the reaction mixture was stirred at 25°C 12 hours. After the reaction was completed, the pH of the reaction mixture was adjusted to 2-3 with hydrochloric acid solution (2M), and concentrated under reduced pressure to obtain intermediate BB-4.
- Step 2 Synthesis of the hydrochloride salt of intermediate BB-5
- Compound BB-6-1 (15.10g, 89.80mmol, 13.25mL) was added to dimethyl sulfoxide (200mL), potassium carbonate (31.03g, 224.51mmol) and 1-tert-butoxycarbonylpiperazine ( 20g, 89.80mmol), after nitrogen replacement three times, the temperature was raised to 120°C, and the reaction was carried out overnight.
- the intermediate BB-6-2 (4g, 11.96mmol) was added to tetrahydrofuran (20mL) and water (20mL), sodium hydroxide (956.84mg, 23.92mmol) was added, nitrogen was replaced three times, the temperature was raised to 70°C, and the reaction overnight. After the reaction was complete, the reaction system was concentrated under reduced pressure to obtain a residue, ethyl acetate (40 mL) and water (30 mL) were added to the residue, the layers were separated, the aqueous phase was collected, and the pH was adjusted to 5-6 with hydrochloric acid solution (1M).
- Step 3 Synthesis of the hydrochloride salt of intermediate BB-6
- the intermediate BB-6-3 (3.2g, 10.45mmol) was added to hydrochloric acid/ethyl acetate solution (4M, 30mL) and reacted at room temperature for 2 hours. After completion of the reaction, filter and rinse the filter cake with ethyl acetate (50 mL), collect the filter cake to obtain the hydrochloride salt of intermediate BB-6.
- reaction solution was lowered to room temperature, concentrated under reduced pressure to remove the solvent, added ice water (3 L) to dissolve, extracted with methyl tert-butyl ether (1 L ⁇ 2), collected the aqueous phase, and used an aqueous sodium hydroxide solution ( 2N, 500 mL ⁇ 3) to wash and collect the aqueous phase.
- the two aqueous phases were combined, and the pH was adjusted to 2-3 with hydrochloric acid solution (6N), and the solid was precipitated, filtered, and the filter cake was rinsed with water (500 mL ⁇ 3), and the filter cake was collected to obtain intermediate BB-7-2.
- the aqueous phase was collected, the organic phase was washed with saturated aqueous sodium bicarbonate (300 mL ⁇ 3), and the aqueous phase was collected.
- the two aqueous phases were combined, and the pH was adjusted to 2 with hydrochloric acid solution (2N). Solids were precipitated, dissolved and extracted with ethyl acetate (2L ⁇ 3), the organic phases were combined, washed with saturated brine (1L), and anhydrous sulfuric acid Dry over sodium, filter, and concentrate the filtrate under reduced pressure to obtain intermediate BB-7-3.
- the intermediate BB-8-2 (4.5g, 20.34mmol) was added to N,N-dimethylformamide (50mL), potassium carbonate (8.43g, 61.03mmol, 3eq), sodium iodide (304.93 mg, 2.03mmol) and 6-bromo-1-hexene (3.98g, 24.41mmol, 3.26mL), after nitrogen replacement three times, the temperature was raised to 80°C, and the reaction was carried out for 5 hours.
- the intermediate BB-8-3 (3.2g, 10.55mmol) was added to tetrahydrofuran (30mL), acrylamide (899.74mg, 12.66mmol) and potassium tert-butoxide (1.54g, 13.71mmol) were added in sequence, and nitrogen replacement was performed three times Then, react at room temperature for 1 hour. After the reaction was completed, the reaction solution was poured into hydrochloric acid solution (1M, 20mL), water (20mL) was added, extracted with ethyl acetate (25mL ⁇ 3), the organic phases were combined, washed with saturated brine (30mL ⁇ 2), Dry over anhydrous sodium sulfate, filter, and concentrate the filtrate under reduced pressure.
- the intermediate BB-10-1 (4.7g, 16.95mmol) was added to dichloroethane (60mL), the reaction system was cooled to 0°C, and N,N-diisopropylethylamine (6.57g, 50.84mmol, 8.85mL), 4-dimethylaminopyridine (207.02mg, 1.69mmol) and methanesulfonyl chloride (3.26g, 28.46mmol, 2.20mL), after nitrogen replacement three times, keep warm for 1 hour.
- the intermediate BB-10-3 (3g, 6.73mmol) was added to tetrahydrofuran (10mL) and water (10mL), sodium hydroxide (1.35g, 33.66mmol) was added, nitrogen was replaced three times, the temperature was raised to 75°C, and the reaction 48 hours. After the reaction was complete, the reaction system was concentrated under reduced pressure to obtain a residue, ethyl acetate (40 mL) and water (30 mL) were added to the residue, the liquid was extracted and separated, the aqueous phase was collected, and the pH was adjusted to 5-5 with hydrochloric acid solution (1M). 6. Concentrate under reduced pressure to obtain intermediate BB-10-4. MS-ESI m/z: 418.3 [M+H] + .
- Step 5 Synthesis of the hydrochloride salt of intermediate BB-10
- Step 1 Synthesis of the hydrochloride salt of intermediate BB-11-2
- reaction solution was cooled to room temperature, and concentrated under reduced pressure to remove most of the organic solvent. Dilute with ethyl acetate (300 mL), wash the organic phase with water (300 mL ⁇ 2), collect the organic phase, dry over anhydrous sodium sulfate, filter, and concentrate under reduced pressure.
- intermediate BB-12-1 0.5 g, 1.06 mmol, purity: 67.60%) and acrylamide (60.00 mg, 844.10 ⁇ mol, 58.25 ⁇ L) were added to N,N-dimethylformamide ( 10 mL), potassium tert-butoxide (118.40 mg, 1.06 mmol) was slowly added to the reaction in batches, and the mixture was replaced with nitrogen three times and then stirred at 0°C for 1 hour.
- intermediate BB-12-2 (50 mg, 121.53 ⁇ mol, purity: 83.94%) into ethyl acetate (5 mL), slowly add hydrochloric acid/ethyl acetate solution (4M, 2 mL) into the reaction, and replace the mixture with nitrogen three times Then stirred at 25°C for 12 hours. The reaction solution was concentrated under reduced pressure to obtain the hydrochloride salt of intermediate BB-12.
- Step 2 Synthesis of the hydrochloride salt of intermediate BB-13-2
- Step 1 Synthesis of the hydrochloride salt of intermediate BB-14
- the intermediate BB-2-2 (50mg, 121.53 ⁇ mol) was added to ethyl acetate (5mL), hydrochloric acid/ethyl acetate (4M, 2mL) was slowly added to the reaction, and the mixture was replaced with nitrogen three times and then stirred at 25°C 12 hours.
- the reaction solution was concentrated under reduced pressure to obtain the hydrochloride salt of intermediate BB-14.
- N-benzyl-bis-o-chloroethylamino hydrochloride 24.33g, 90.57mmol
- ethanol 150mL
- compound BB-15-1 24.33g, 90.57mmol
- N , N-diisopropylethylamine 117.05g, 905.70mmol, 157.75mL
- Step 2 Synthesis of the hydrochloride salt of intermediate BB-15-3
- the intermediate BB-15-3 (3.5g, 14.91mmol, hydrochloride) was dissolved in a mixed solvent of dioxane (30mL) and water (10mL), the reaction system was cooled to 0°C, and sodium bicarbonate ( 3.76g, 44.73mmol, 1.74mL) and tert-butyl carbonate (3.25g, 14.91mmol, 3.43mL), returned to 20°C and stirred for 12 hours.
- the intermediate BB-15-4 (3.1g, 10.39mmol) was dissolved in tetrahydrofuran (35mL), and after nitrogen replacement three times, the temperature was lowered to -65°C, lithium aluminum tetrahydride (1M tetrahydrofuran solution, 31.17mL) was slowly added, 0 °C and stirred for 1 hour. After the reaction was completed, sodium sulfate decahydrate (30 g) was slowly added to the reaction liquid at 0° C., filtered, the filter cake was rinsed with dichloromethane, and the filtrate was concentrated under reduced pressure to obtain intermediate BB-15-5.
- the intermediate BB-4-3 (2.39g, 5.66mmol) was dissolved in tetrahydrofuran (50mL), and then the intermediate BB-15-5 (2.9g, 11.31mmol) and triphenylphosphine (2.97 g, 11.31 mmol), the reaction system was cooled to 0°C, and diisopropyl azodicarboxylate (2.29 g, 11.31 mmol, 2.20 mL) was added dropwise. The temperature was raised to 40°C and stirred for 12 hours.
- Step 6 Synthesis of the hydrochloride salt of intermediate BB-15-7
- the intermediate BB-15-7 (0.2g, 334.42 ⁇ mol, hydrochloride) was dissolved in tetrahydrofuran (3mL), nitrogen protection dropped to 0°C, triethylamine (203.04mg, 2.01mmol, 279.28 ⁇ L) was added dropwise, and After completion, a solution of ethyl bromoacetate (111.70 mg, 668.84 ⁇ mol, 73.97 ⁇ L) in tetrahydrofuran (1 mL) was added dropwise, and the mixture was slowly raised to 20° C. and stirred for 2 hours.
- the intermediate BB-15-8 (210 mg, 324.23 ⁇ mol) was dissolved in a mixed solvent of tetrahydrofuran (2 mL) and water (0.5 mL), lowered to 0 ° C, and lithium hydroxide monohydrate (68.02 mg, 1.62 mmol) was added, After the addition was complete, stir at 20°C for 1 hour. The pH of the reaction solution was adjusted to 3-4 with hydrochloric acid solution (1M), and concentrated under reduced pressure to obtain intermediate BB-15.
- aqueous phase and solid were collected, and the pH was adjusted to 3-4 with hydrochloric acid solution (2M). If there was a large amount of insoluble matter, ethyl acetate (500 mL) was added, and the organic phase was collected after liquid separation. The aqueous phase was extracted with ethyl acetate (300mL ⁇ 3), the organic phases were combined, washed successively with citric acid solution (10%, 300mL) and saturated brine (300mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. Intermediate BB-17-4 was obtained.
- the aqueous phase was extracted with ethyl acetate (200 mL ⁇ 4), the organic phases were combined, washed with saturated brine (300 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a residue.
- reaction solution was concentrated under reduced pressure to obtain a dark yellow residue.
- Water (100 mL) and ethyl acetate (200 mL) were added to the residue, and the organic phase was collected after separation.
- the aqueous phase was extracted with ethyl acetate (100 mL ⁇ 3), the combined organic phases were washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a residue.
- MS-ESI m/z 371.2 [M+H] + .
- the intermediate BB-17-6 (0.25g, 674.95 ⁇ mol) was dissolved in tetrahydrofuran (3mL), cooled to 0°C, and acrylamide (47.97mg, 674.95 ⁇ mol, 46.58 ⁇ L) and potassium tert-butoxide (113.61mg, 1.01 mmol).
- the reaction system was stirred at 20°C for 1 hour.
- the reaction solution was slowly poured into saturated ammonium chloride solution (10 mL), ethyl acetate (10 mL) was added, and the organic phase was separated.
- Step 7 Synthesis of the hydrochloride salt of intermediate BB-17
- the intermediate BB-17-7 (120 mg, 303.48 ⁇ mol) was dissolved in hydrochloric acid/ethyl acetate solution (5 mL, 4M), and reacted at 25° C. for 30 minutes. The reaction solution was filtered, the filter cake was rinsed with ethyl acetate (3 mL ⁇ 3), and the filter cake was collected to obtain the hydrochloride salt of intermediate BB-17.
- Step 1 Synthesis of Compound BB-19-1
- 5-Hexenoic acid (82.57 mg, 723.43 ⁇ mol, 85.92 ⁇ L) was dissolved in N,N-dimethylformamide (3 mL), followed by the addition of 2-(7-azabenzotriazole)-N, N, N', N'-tetramethyluronium hexafluorophosphate (343.84 mg, 904.29 ⁇ mol) and N, N-diisopropylethylamine (311.66 mg, 2.41 mmol, 420.03 ⁇ L), the reaction system was replaced with nitrogen three times After stirring at 25°C for 20 minutes, the hydrochloride salt of compound BB-17 (200 mg, 602.86 ⁇ mol) was added, and the reaction solution was stirred at 25°C for 12 hours.
- Step 1 Synthesis of Compound BB-20-2
- the compound BB-20-2 (10g, 29.03mmol) was dissolved in N,N-dimethylacetamide (100mL), 4-cyano-3-trifluoromethylisothiocyanate benzene Ester (6.62g, 29.03mmol) was added to the reaction solution in batches, the reaction mixture was stirred at 20°C for 3 hours, methanol (100mL) and dilute hydrochloric acid (2M, 56.32mL) were added, the reaction mixture was stirred at 70°C for 2 hours, and the reaction was completed Then, it was cooled to room temperature, diluted with water (100 mL), and extracted with ethyl acetate (100 mL ⁇ 3).
- Step 1 Synthesis of Compound BB-21-2
- reaction solution was slowly poured into saturated aqueous sodium sulfite (300 mL), stirred for 10 minutes, extracted with dichloromethane (400 mL), and the organic phase was collected, washed with saturated brine (500 mL), and dried over anhydrous sodium sulfate. Filter and concentrate under reduced pressure to obtain the crude product.
- Acetyl chloride (32.78g, 417.56mmol, 29.80mL) was dissolved in dichloromethane (1L), the reaction system was cooled to 0°C, aluminum trichloride (106.29g, 797.16mmol) was added, and under nitrogen protection at 0°C After reacting for 30 minutes, compound BB-21-3 (90 g, 379.60 mmol) was added, and the reaction was continued for 5.5 hours, then returned to room temperature and reacted for 10 hours.
- the organic phase was extracted with 2M aqueous sodium hydroxide solution (150 mL ⁇ 2), and the aqueous phase was collected. Combine the aqueous phases, adjust the pH to 2-3 with 6M hydrochloric acid solution, precipitate a solid, filter, rinse the filter cake with water (150 mL ⁇ 2), collect the filter cake to obtain compound BB-21-6.
- tert-Butyl carbamate (2.31g, 19.75mmol) was added to toluene (60mL) and water (20mL), and compound BB-21 (6g, 17.96mmol), 2-di-tert-butylphosphine-2', 4', 6'-triisopropylbiphenyl (762.45mg, 1.80mmol), tris(dibenzylideneacetone) dipalladium (822.10mg, 897.76 ⁇ mol) and potassium phosphate (15.25g, 71.82mmol), nitrogen protection The temperature was raised to 100°C to react for 12 hours.
- compound BB-22-1 (2.3g, 6.21mmol) was added into tetrahydrofuran (25mL), followed by adding acrylamide (529.63mg, 7.45mmol) and potassium tert-butoxide (1.05g, 9.31mmol), nitrogen protection The reaction was carried out for 1 hour. After the reaction was completed, the reaction solution was added to 1 mol/L hydrochloric acid solution, extracted with ethyl acetate (45mL ⁇ 3), the organic phase was collected, washed with saturated brine (30mL ⁇ 2), dried over anhydrous sodium sulfate, and filtered , and the filtrate was concentrated under reduced pressure to obtain a crude product.
- Step 4 Synthesis of the hydrochloride salt of compound BB-23
- 5-Hexenoic acid (75.69 mg, 663.14 ⁇ mol, 78.76 ⁇ L) was dissolved in N,N-dimethylformamide (5 mL), followed by the addition of 2-(7-azabenzotriazole)-N, N, N', N'-tetramethyluronium hexafluorophosphate (343.84 mg, 904.29 ⁇ mol) and N, N-diisopropylethylamine (233.75 mg, 1.81 mmol, 315.02 ⁇ L), the reaction system was replaced with nitrogen three times After stirring at 20°C for 30 minutes, the hydrochloride salt of compound BB-22 (200 mg, 602.86 ⁇ mol) was added, and the reaction solution was stirred at 20°C for 2 hours.
- compound BB-21 (300mg, 897.76 ⁇ mol), 1-tert-butyloxycarbonylpiperazine (299.91mg, 1.35mmol), tris(dibenzylideneacetone)dipalladium (41.10mg, 44.89 ⁇ mol), cesium carbonate (41.10mg, 44.89 ⁇ mol) and 2-bicyclohexylphosphine-2,6-diisopropoxy-1,1-biphenyl (41.89mg, 89.78 ⁇ mol) were dissolved in 1,4-diox Hexacyclone (5 mL), the reaction mixture was warmed to 100°C and stirred for 2 hours.
- Step 3 Synthesis of the hydrochloride salt of compound BB-25
- compound BB-26-1 25g, 144.87mmol
- 4-piperidinemethanol 16.69g, 144.87mmol
- 1,4-dioxane 500mL
- potassium carbonate 60.07 g, 434.61mmol
- tetrabutylammonium iodide 5.35g, 14.49mmol
- reaction solution was poured into dilute hydrochloric acid (1N, 800 mL) at 0-5° C., and extracted with ethyl acetate (700 mL ⁇ 3). The organic phases were combined, washed with saturated brine (700 mL ⁇ 2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product.
- compound BB-26-4 (60.5 g, 127.19 mmol) and N, N-diisopropylethylamine (82.19 g, 635.97 mmol) were dissolved in N, N-dimethylformamide (700 mL) , Add 2-(7-azabenzotriazole)-N, N, N', N'-tetramethyluronium hexafluorophosphate (58.04g, 152.63mmol), and the reaction mixture was stirred at room temperature for 5 minutes , the hydrochloride salt of compound BB-26-7 (32.88 g, 114.47 mmol) was added, and the reaction mixture was stirred overnight at room temperature.
- Step 7 Synthesis of Compound BB-26-9
- compound BB-26-8 (53g, 74.82mmol) was dissolved in N,N-dimethylformamide (530mL), potassium fluoride (30.43g, 523.74mmol) was added, and the reaction mixture was heated to 100°C , and the reaction was stirred overnight. After completion of the reaction, cool to room temperature, combine two batches, add water (3 L) to the reaction solution, and extract with ethyl acetate (1 L ⁇ 3). The organic phases were combined, washed with saturated brine (1 L ⁇ 3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product.
- N,N-dimethylformamide 530mL
- potassium fluoride 30.43g, 523.74mmol
- Step 8 Synthesis of compound BB-26
- compound BB-27-1 (0.5g, 2.90mmol) and endo-3-azabicyclo[3.1.0]hexane-6-methanol (328.16mg, 2.90mmol) were dissolved in 1,4- Potassium carbonate (1.20 g, 8.70 mmol) and tetrabutylammonium iodide (107.02 mg, 290.00 ⁇ mol) were sequentially added to dioxane (10 mL), and the reaction mixture was heated to 100° C., and stirred overnight.
- Step 1 Synthesis of Compound BB-28-2
- compound BB-28-6 (1.1g, 2.31mmol) and N,N-diisopropylethylamine (1.19g, 9.23mmol, 1.61mL) were dissolved in N,N-dimethyl Formamide (10mL), then added 2-(7-azobenzotriazole)-N,N,N,N-tetramethyluronium hexafluorophosphate (1.14g, 3.00mmol), and the reaction mixture was After the reaction was stirred for 5 minutes, the hydrochloride salt of intermediate BB-26-7 (662.77 mg, 2.31 mmol) was added, and the reaction mixture was stirred at room temperature for 55 minutes.
- compound BB-28-7 (0.5 g, 704.87 ⁇ mol) was dissolved in N,N-dimethylformamide (6 mL), potassium fluoride (409.51 mg, 7.05 mmol) was added, and the reaction mixture was heated to 100 °C, the reaction was stirred overnight. After the reaction was completed, it was cooled to room temperature. Water (30 mL) was added and extracted with ethyl acetate (20 mL ⁇ 3). The organic phases were combined, washed with saturated brine (60 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to remove the solvent.
- Step 8 Synthesis of Compound BB-28
- the compound BB-28-8 (0.257g, 545.70 ⁇ mol) was dissolved in dichloromethane (5mL), cooled to 0°C, and the Dess-Martin oxidant (347.18mg, 818.56 ⁇ mol) was added to react The mixture was stirred at room temperature for 1.5 hours. After the reaction was completed, it was filtered, and the filter cake was washed with ethyl acetate (30 mL). Saturated sodium bicarbonate (20 mL) was added to the filtrate, stirred for 5 minutes, and extracted with ethyl acetate (20 mL ⁇ 3).
- Step 1 Synthesis of Compound BB-29-2
- compound BB-26-7 (4g, 13.93mmol) and compound BB-29-3 (3.30g, 13.93mmol) were dissolved in N,N-dimethylformamide (100mL), followed by Add triethylamine (7.05g, 69.64mmol, 9.69mL), 1-hydroxybenzotriazole (2.82g, 20.89mmol) and 1-ethyl-(3-dimethylaminopropyl)carbodiimide Hydrochloride (4.01 g, 20.89 mmol), and the reaction mixture was stirred at 25°C for 12 hours.
- the reaction solution was poured into 20 mL of water and 30 mL of ethyl acetate, the organic phase was separated and washed with water (20 mL ⁇ 2), the collected organic phase was dried with anhydrous sodium sulfate, filtered, and spin-dried under reduced pressure.
- the crude product was purified by preparative HPLC (mobile phase: acetonitrile/water; acidic system: 0.05% hydrochloric acid) to obtain compound WX005.
- MS-ESI m/z 792.1[M+H] + .
- reaction solution was diluted with ethyl acetate (30mL), the organic phase was washed with water (20mL ⁇ 3), the organic phase was collected, dried over anhydrous sodium sulfate, filtered, spin-dried under reduced pressure, and the resulting residue was separated by preparative HPLC (mobile phase: acetonitrile/water ; Acidic system: 0.05% hydrochloric acid), to obtain compound WX006.
- reaction solution was poured into dilute hydrochloric acid (10mL), added 5mL of water, extracted with ethyl acetate (15mL ⁇ 3), the organic phase was washed with saturated brine (10mL ⁇ 2), dried over anhydrous sodium sulfate, and filtered , concentrated under reduced pressure. The residue was separated and purified by preparative HPLC (mobile phase: acetonitrile/water; acidic system: 0.04% hydrochloric acid) to obtain compound WX014. MS-ESI m/z: 869.3 [M+H] + .
- compound BB-23 50 mg, 127.10 ⁇ mol was added to 1,2-dichloroethane (10 mL), and the hydrochloride of compound BB-24 (72.81 mg, 152.52 ⁇ mol) and potassium acetate were added slowly (18.71 mg, 190.65 ⁇ mol), after stirring for 1 hour, sodium triacetoxyborohydride (61.96 mg, 292.33 ⁇ mol) was added, and reacted overnight under nitrogen protection.
- compound BB-4 (85.88 mg, 144.69 ⁇ mol) was added to N, N-dimethylformamide (8 mL), and 2-(7-azabenzotriazole)-N, N , N', N'-tetramethylurea hexafluorophosphate (68.77mg, 180.86 ⁇ mol) and N, N-diisopropylethylamine (77.92mg, 602.86 ⁇ mol, 105.01 ⁇ L), after stirring for 0.5 hours, add The hydrochloride salt of compound BB-22 (40 mg, 120.57 ⁇ mol) was reacted under nitrogen protection for 2.5 hours.
- the hydrochloride salt of compound BB-25 (33 mg, 82.32 ⁇ mol) was dissolved in N,N-dimethylformamide (1 mL), and compound BB-26 (38.52 mg, 82.32 ⁇ mol) was added, Potassium acetate (24.24 mg, 246.97 ⁇ mol) and glacial acetic acid (0.1 mL), the reaction mixture was stirred at room temperature for 0.5 hour, then sodium triacetoxyborohydride (52.34 mg, 246.97 ⁇ mol) was added, and the reaction mixture was stirred for 1 hour.
- compound BB-27 99 mg, 169.55 ⁇ mol was dissolved in N,N-dimethylformamide (3 mL), and compound BB-25 hydrochloride (67.97 mg, 169.55 ⁇ mol) was added, Potassium acetate (49.92 mg, 508.66 ⁇ mol) and glacial acetic acid (0.1 mL), the reaction mixture was stirred at room temperature for 0.5 hours. Then sodium triacetoxyborohydride (107.81 mg, 508.66 ⁇ mol) was added, and the stirring reaction was continued for 1 hour.
- BB-28 (0.07g, 149.27 ⁇ mol) was dissolved in N,N-dimethylformamide (3mL), and then the hydrochloride of intermediate BB-25 (59.84mg, 149.27 ⁇ mol ), potassium acetate (43.95mg, 447.82 ⁇ mol) and acetic acid (105.00mg, 1.75mmol, 0.1mL), the reaction mixture was stirred and reacted at room temperature for 0.5 hours, then added sodium triacetoxyborohydride (94.91mg, 447.82 ⁇ mol), The reaction mixture was stirred for 12 hours. After completion of the reaction, the solvent was removed by concentration under reduced pressure.
- Test Example 1 Evaluation of anti-proliferative effect in human prostate cancer LNCaP cells Experimental purpose
- the tumor cell lines were cultured in an incubator at 37°C, 5% CO 2 according to the above culture conditions. Passage regularly, and take cells in logarithmic growth phase for plating.
- Drug addition Take 10 ⁇ L of the working solution of 10 times the initial concentration of the compound and add it to the cell culture plate. Add 10 ⁇ L of DMSO-cell culture solution mixture to the vehicle control and the blank control.
- IR Inhibition rate
- the compound of the present invention exhibits excellent inhibitory effect on cell proliferation in human prostate cancer cell LNCaP.
- Test Example 2 Evaluation of antiproliferative effect in human prostate cancer VCaP cells
- the tumor cell lines were cultured in an incubator at 37°C, 5% CO 2 according to the above culture conditions. Passage regularly, and take cells in logarithmic growth phase for plating.
- Drug addition Take 10 ⁇ L of the working solution of 10 times the initial concentration of the compound and add it to the cell culture plate. Add 10 ⁇ L of DMSO-cell culture solution mixture to the vehicle control and the blank control.
- IR Inhibition rate
- the compound of the present invention exhibits excellent inhibitory effect on cell proliferation in human prostate cancer cell VCaP.
- Test Example 3 In vivo pharmacodynamic study of human prostate cancer LNCaP cell subcutaneous xenograft tumor CB-17 SCID model
- Human prostate cancer LNcap cells (ECACC-89110211) were cultured in a single layer in vitro.
- the culture conditions were RPMI-1640 medium plus 10% fetal bovine serum, 100 U/mL penicillin and 100 ⁇ g/mL streptomycin, and incubated at 37°C in 5% CO 2 box culture. Routine digestion with trypsin-EDTA was performed twice a week for passaging. When the cell saturation is 80%-90% and the number reaches the requirement, the cells are collected, counted, and inoculated.
- Administration volume 10 ⁇ L/g based on mouse body weight. If body weight is lost by more than 15%, the dosing regimen should be adjusted accordingly.
- Tumor diameters were measured twice a week with vernier calipers.
- TGI (%) reflects tumor growth inhibition rate.
- TGI (%) [(1-(average tumor volume at the end of administration of a certain treatment group-average tumor volume at the beginning of administration of this treatment group))/(average tumor volume at the end of treatment of the solvent control group-the beginning of treatment of the solvent control group Time-average tumor volume)] ⁇ 100%.
- the compounds of the present invention exhibit significant tumor-inhibitory effects on the human prostate cancer LNCaP xenograft tumor model.
- Test Example 4 In vitro test of regulation of AR protein level in human prostate cancer LNCaP cells
- the 384 cell plate inoculated with cells was placed in the ultra-clean bench for 10 minutes.
- the compound was serially diluted 3 times (4 ⁇ L+8 ⁇ L) in the Echo plate with a Bravo instrument.
- ZPE control wells are dimethyl sulfoxide.
- PBS Join Intercept
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Organic Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Plural Heterocyclic Compounds (AREA)
Abstract
一系列戊二酰亚胺取代的异噁唑稠环化合物及其应用,具体公开了式(II)所示化合物及其药学上可接受的盐。
Description
本申请主张如下优先权:
CN2021106741905,申请日:2021年6月17日;
CN2021112224376,申请日:2021年10月20日。
本发明涉及一系列戊二酰亚胺取代的异噁唑稠环化合物及其应用,具体涉及式(II)所示合物及其药学上可接受的盐。
蛋白降解靶向嵌合体(Proteolysis Targeting Chimera,PROTAC)是一种应用泛素-蛋白酶体系统靶向特定蛋白质并诱导其在细胞内降解的技术。泛素-蛋白酶体系统是细胞内蛋白质降解的主要途径,其正常生理功能主要负责清除细胞内变性、突变或有害蛋白质,细胞内80%以上蛋白质的降解都依赖于泛素-蛋白酶体系统。PROTAC利用细胞自身的蛋白质破坏机制,清除细胞中的特异性靶向蛋白。
本发明提供了双功能或蛋白水解靶向嵌合(PROTAC)化合物,发现其可作为靶向泛素化和雄激素受体(AR)降解的调节剂。
发明内容
本发明提供式(II)所示化合物或其药学上可接受的盐,
其中,
R
1选自H和CH
3;
R
2选自H和甲基;
E
1选自单键、O、NH和-C(=O)NH-;
E
4选自单键、O和-C(=O)NH-;
E
5选自5-6元杂环烷基;
T
1和T
2各自独立地选自CH和N;
m选自1、2和3;
n选自1、2、3、4、5、6和7;
p和q各自独立地选自0、1和2;
环A选自苯基和5-6元杂芳基;
ABM选自靶向结合于AR蛋白的药物或其衍生物。
在本发明的一些方案中,上述ABM选自式(ABM-1)和(ABM-2)的结构:
其中,
R
3和R
4选自甲基;
或者,R
3与R
4与共同相连的碳原子成C
4-6环烷基;
环B选自苯基和吡啶基,所述苯基和吡啶基任选被1、2或3个R
a取代;
环C选自苯基和6元杂芳基,所述苯基和6元杂芳基任选被1、2或3个R
b取代;
R
a选自F、Cl、Br、I、CN、CH
3、CF
3和NO
2;
R
b选自F和Cl;
在本发明的一些方案中,上述ABM选自式(ABM-1a)、(ABM-2a)、(ABM-2b)和(ABM-3a)的结构:
其中,
R
b1选自H和F;
t选自1、2和3;
T
1选自CH和N;
R
a如本发明所定义。
本发明还提供式(I)所示化合物或其药学上可接受的盐,
其中,
R
1选自H和CH
3;
R
2选自H和甲基;
E
1选自单键、O、NH和-C(=O)NH-;
E
4选自单键、O和-C(=O)NH-;
E
5选自5-6元杂环烷基;
T
1和T
2各自独立地选自CH和N;
m选自1、2和3;
n选自1、2、3、4、5、6和7;
p和q各自独立地选自0、1和2;
环A选自不存在、苯基和5-6元杂芳基;
ABM选自靶向结合于AR蛋白的药物或其衍生物。
在本发明的一些方案中,上述ABM选自式(ABM-1)和(ABM-2)的结构:
其中,
R
3和R
4选自甲基;
或者,R
3与R
4与共同相连的碳原子成C
4-6环烷基;
环B选自苯基和吡啶基,所述苯基和吡啶基任选被1、2或3个R
a取代;
环C选自苯基和6元杂芳基,所述苯基和6元杂芳基任选被1、2或3个R
b取代;
R
a选自F、Cl、Br、I、CN、CH
3、CF
3和NO
2;
R
b选自F和Cl;
在本发明的一些方案中,上述ABM选自式(ABM-1a)、(ABM-2a)和(ABM-2b)的结构:
其中,
R
b1选自H和F;
t选自1、2和3;
R
a如本发明所定义。
本发明还有一些方案是由上述各变量任意组合而来。
本发明还提供下述化合物或其药学上可接受的盐:
技术效果
本发明化合物具有优异的AR蛋白降解作用、细胞增殖抑制作用和显著的抑瘤作用,尤其是针对前列腺癌,具有优异的治疗作用;具有良好的成药性。
定义和说明
除非另有说明,本文所用的下列术语和短语旨在具有下列含义。一个特定的术语或短语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文中出现商品名时,意在指代其对应的商品或其活性成分。
这里所采用的术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”是指本发明化合物的盐,由本发明发现的具有特定取代基的化合物与相对无毒的酸或碱制备。当本发明的化合物中含有相对酸性的功能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的碱与这类化合物的中性形式接触的方式获得碱加成盐。药学上可接受的碱加成盐包括钠、钾、钙、铵、有机胺或镁盐或类似的盐。当本发明的化合物中含有相对碱性的官能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的酸与这类化合物的中性形式接触的方式获得酸加成盐。本发明的某些特定的化合物含有碱性和酸性的官能团,从而可以被转换成任一碱或酸加成盐。
本发明的药学上可接受的盐可由含有酸根或碱基的母体化合物通过常规化学方法合成。一般情况下,这样的盐的制备方法是:在水或有机溶剂或两者的混合物中,经由游离酸或碱形式的这些化合物与化学计量的适当的碱或酸反应来制备。
术语“靶向蛋白”表示结合本发明化合物并被降解的蛋白质或多肽。
术语“药物或其衍生物”包括已开发可结合于靶向蛋白的药物或其衍生物和将来开发的可结合于靶向蛋白的药物或其衍生物。
本发明的化合物可以存在特定的几何或立体异构体形式。本发明设想所有的这类化合物,包括顺式和 反式异构体、(-)-和(+)-对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,及其外消旋混合物和其他混合物,例如对映异构体或非对映体富集的混合物,所有这些混合物都属于本发明的范围之内。烷基等取代基中可存在另外的不对称碳原子。所有这些异构体以及它们的混合物,均包括在本发明的范围之内。
除非另有说明,术语“对映异构体”或者“旋光异构体”是指互为镜像关系的立体异构体。
除非另有说明,术语“非对映异构体”是指分子具有两个或多个手性中心,并且分子间为非镜像的关系的立体异构体。
除非另有说明,“(+)”表示右旋,“(-)”表示左旋,“(±)”表示外消旋。
除非另有说明,用楔形实线键
和楔形虚线键
表示一个立体中心的绝对构型,用直形实线键
和直形虚线键
表示立体中心的相对构型,用波浪线
表示楔形实线键
或楔形虚线键
或用波浪线
表示直形实线键
和直形虚线键
除非另有规定,当某一基团具有一个或多个可连接位点时,该基团的任意一个或多个位点可以通过化学键与其他基团相连。所述位点与其他基团连接的化学键可以用直形实线键
直形虚线键
或波浪线
表示。例如-OCH
3中的直形实线键表示通过该基团中的氧原子与其他基团相连;
中的直形虚线键表示通过该基团中的氮原子的两端与其他基团相连;
中的波浪线表示通过该苯基集团中的1和2位的碳原子与其他基团相连。
本发明的化合物可以存在特定的。除非另有说明,术语“互变异构体”或“互变异构体形式”是指在室温下,不同官能团异构体处于动态平衡,并能很快的相互转化。若互变异构体是可能的(如在溶液中),则可以达到互变异构体的化学平衡。例如,质子互变异构体(proton tautomer)(也称质子转移互变异构体(prototropic tautomer))包括通过质子迁移来进行的互相转化,如酮-烯醇异构化和亚胺-烯胺异构化。价键异构体(valence tautomer)包括一些成键电子的重组来进行的相互转化。其中酮-烯醇互变异构化的具体实例是戊烷-2,4-二酮与4-羟基戊-3-烯-2-酮两个互变异构体之间的互变。
除非另有说明,术语“富含一种异构体”、“异构体富集”、“富含一种对映体”或者“对映体富集”指其中一种异构体或对映体的含量小于100%,并且,该异构体或对映体的含量大于等于60%,或者大于等于70%,或者大于等于80%,或者大于等于90%,或者大于等于95%,或者大于等于96%,或者大于等于97%,或者大于等于98%,或者大于等于99%,或者大于等于99.5%,或者大于等于99.6%,或者大于等于99.7%,或者大于等于99.8%,或者大于等于99.9%。
除非另有说明,术语“异构体过量”或“对映体过量”指两种异构体或两种对映体相对百分数之间的差值。例如,其中一种异构体或对映体的含量为90%,另一种异构体或对映体的含量为10%,则异构体或对映体过量(ee值)为80%。
可以通过的手性合成或手性试剂或者其他常规技术制备光学活性的(R)-和(S)-异构体以及D和L异构体。如果想得到本发明某化合物的一种对映体,可以通过不对称合成或者具有手性助剂的衍生作用来制备,其中将所得非对映体混合物分离,并且辅助基团裂开以提供纯的所需对映异构体。或者,当分子中含有碱性官能团(如氨基)或酸性官能团(如羧基)时,与适当的光学活性的酸或碱形成非对映异构体的盐,然后通过本领域所公知的常规方法进行非对映异构体拆分,然后回收得到纯的对映体。此外,对映异构体和非对映异构体的分离通常是通过使用色谱法完成的,所述色谱法采用手性固定相,并任选地与化学衍生法相结合(例如由胺生成氨基甲酸盐)。
本发明的化合物可以在一个或多个构成该化合物的原子上包含非天然比例的原子同位素。例如,可用放射性同位素标记化合物,比如氚(
3H),碘-125(
125I)或C-14(
14C)。又例如,可用重氢取代氢形成氘代药物,氘与碳构成的键比普通氢与碳构成的键更坚固,相比于未氘化药物,氘代药物有降低毒副作用、增加药物稳定性、增强疗效、延长药物生物半衰期等优势。本发明的化合物的所有同位素组成的变换,无论放射性与否,都包括在本发明的范围之内。
术语“任选”或“任选地”指的是随后描述的事件或状况可能但不是必需出现的,并且该描述包括其中所述事件或状况发生的情况以及所述事件或状况不发生的情况。
术语“被取代的”是指特定原子上的任意一个或多个氢原子被取代基取代,取代基可以包括重氢和氢的变体,只要特定原子的价态是正常的并且取代后的化合物是稳定的。当取代基为氧(即=O)时,意味着两个氢原子被取代。氧取代不会发生在芳香基上。术语“任选被取代的”是指可以被取代,也可以不被取代,除非另有规定,取代基的种类和数目在化学上可以实现的基础上可以是任意的。
当任何变量(例如R)在化合物的组成或结构中出现一次以上时,其在每一种情况下的定义都是独立的。因此,例如,如果一个基团被0-2个R所取代,则所述基团可以任选地至多被两个R所取代,并且每种情况下的R都有独立的选项。此外,取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
当一个连接基团的数量为0时,比如-(CRR)
0-,表示该连接基团为单键。
当一个取代基为空缺时,表示该取代基是不存在的,比如A-X中X为空缺时表示该结构实际上是A。当所列举的取代基中没有指明其通过哪一个原子连接到被取代的基团上时,这种取代基可以通过其任何原子相键合,例如,吡啶基作为取代基可以通过吡啶环上任意一个碳原子连接到被取代的基团上。
除非另有规定,术语“C1-3烷基”用于表示直链或支链的由1至3个碳原子组成的饱和碳氢基团。所述 C
1-3烷基包括C
1-2和C
2-3烷基等;其可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基)。C
1-
3烷基的实例包括但不限于甲基(Me)、乙基(Et)、丙基(包括n-丙基和异丙基)等。
除非另有规定,环上原子的数目通常被定义为环的元数,例如,“5-7元环”是指环绕排列5-7个原子的“环”。
除非另有规定,“C
4-6环烷基”表示由4至6个碳原子组成的饱和环状碳氢基团,其包括单环和双环体系,其中双环体系包括螺环、并环和桥环。所述C
4-6环烷基包括C
4-6、C
4-5或C
5-6环烷基等;其可以是一价、二价或者多价。C
4-6环烷基的实例包括,但不限于,环丁基、环戊基、环己基等。
除非另有规定,C
n-n+m或C
n-C
n+m包括n至n+m个碳的任何一种具体情况,例如C
1-12包括C
1、C
2、C
3、C
4、C
5、C
6、C
7、C
8、C
9、C
10、C
11、和C
12,也包括n至n+m中的任何一个范围,例如C
1-12包括C
1-
3、C
1-6、C
1-9、C
3-6、C
3-9、C
3-12、C
6-9、C
6-12、和C
9-12等;同理,n元至n+m元表示环上原子数为n至n+m个,例如3-12元环包括3元环、4元环、5元环、6元环、7元环、8元环、9元环、10元环、11元环、和12元环,也包括n至n+m中的任何一个范围,例如3-12元环包括3-6元环、3-9元环、5-6元环、5-7元环、6-7元环、6-8元环、和6-10元环等。
除非另有规定,术语“5-6元杂环烷基”本身或者与其他术语联合分别表示由5至6个环原子组成的饱和环状基团,其1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子,其中氮原子任选地被季铵化,氮和硫杂原子可任选被氧化(即NO和S(O)p,p是1或2)。其包括单环和双环体系,其中双环体系包括螺环、并环和桥环。此外,就该“5-6元杂环烷基”而言,杂原子可以占据杂环烷基与分子其余部分的连接位置。所述5-6元杂环烷基包括5元和6元杂环烷基。5-6元杂环烷基的实例包括但不限于吡咯烷基、吡唑烷基、咪唑烷基、四氢噻吩基(包括四氢噻吩-2-基和四氢噻吩-3-基等)、四氢呋喃基(包括四氢呋喃-2-基等)、四氢吡喃基、哌啶基(包括1-哌啶基、2-哌啶基和3-哌啶基等)、哌嗪基(包括1-哌嗪基和2-哌嗪基等)、吗啉基(包括3-吗啉基和4-吗啉基等)、二噁烷基、二噻烷基、异噁唑烷基、异噻唑烷基、1,2-噁嗪基、1,2-噻嗪基、六氢哒嗪基等。
除非另有规定,本发明术语“5-6元杂芳环”和“5-6元杂芳基”可以互换使用,术语“5-6元杂芳基”表示由5至6个环原子组成的具有共轭π电子体系的单环基团,其1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子。其中氮原子任选地被季铵化,氮和硫杂原子可任选被氧化(即NO和S(O)p,p是1或2)。5-6元杂芳基可通过杂原子或碳原子连接到分子的其余部分。所述5-6元杂芳基包括5元和6元杂芳基。所述5-6元杂芳基的实例包括但不限于吡咯基(包括N-吡咯基、2-吡咯基和3-吡咯基等)、吡唑基(包括2-吡唑基和3-吡唑基等)、咪唑基(包括N-咪唑基、2-咪唑基、4-咪唑基和5-咪唑基等)、噁唑基(包括2-噁唑基、4-噁唑基和5-噁唑基等)、三唑基(1H-1,2,3-三唑基、2H-1,2,3-三唑基、1H-1,2,4-三唑基和4H-1,2,4-三唑基等)、四唑基、异噁唑基(3-异噁唑基、4-异噁唑基和5-异噁唑基等)、噻唑基(包括2-噻唑 基、4-噻唑基和5-噻唑基等)、呋喃基(包括2-呋喃基和3-呋喃基等)、噻吩基(包括2-噻吩基和3-噻吩基等)、吡啶基(包括2-吡啶基、3-吡啶基和4-吡啶基等)、吡嗪基、哒嗪基或嘧啶基(包括2-嘧啶基和4-嘧啶基等)。
除非另有规定,当某一基团具有一个或多个可连接位点时,该基团的任意一个或多个位点可以通过化学键与其他基团相连。当该化学键的连接方式是不定位的,且可连接位点存在H原子时,则连接化学键时,该位点的H原子的个数会随所连接化学键的个数而对应减少变成相应价数的基团。所述位点与其他基团连接的化学键可以用直形实线键
直形虚线键
或波浪线
表示。例如-OCH
3中的直形实线键表示通过该基团中的氧原子与其他基团相连;
中的直形虚线键表示通过该基团中的氮原子的两端与其他基团相连;
中的波浪线表示通过该苯基基团中的1和2位碳原子与其他基团相连;
表示萘环上的任意可连接位点可以通过1个化学键与其他基团相连,至少包括
这6种连接方式。
本发明的化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。
本发明的化合物可以通过本领域技术人员所熟知的常规方法来确认结构,如果本发明涉及化合物的绝对构型,则该绝对构型可以通过本领域常规技术手段予以确证。例如单晶X射线衍射法(SXRD),把培养出的单晶用Bruker D8 venture衍射仪收集衍射强度数据,光源为CuKα辐射,扫描方式:
扫描,收集相关数据后,进一步采用直接法(Shelxs97)解析晶体结构,便可以确证绝对构型。
本发明所使用的溶剂可经市售获得。
下面通过实施例对本发明进行详细描述,但并不意味着对本发明任何不利限制。本文已经详细地描述了本发明,其中也公开了其具体实施例方式,对本领域的技术人员而言,在不脱离本发明精神和范围的情况下针对本发明具体实施方式进行各种变化和改进将是显而易见的。
参考例1:片段BB-1
合成路线:
步骤1:中间体BB-1-2的合成
将化合物BB-1-1(100g,465.02mmol),碳酸二甲酯(167.55g,1.86mol,156.59mL)溶于四氢呋喃(1L)中,氮气置换三次,在氮气保护下冷却至0℃,加入叔丁醇钾(313.08g,2.79mol),升温至70℃,搅拌12小时。减压浓缩除去溶剂,加入甲基叔丁基醚(500mL),缓慢倒入冰水(1000mL)中,有机相用氢氧化钠水溶液(2N,250mL)洗涤,水相合并后用盐酸溶液(6N)调节pH至2-3,过滤,滤饼用水(500mL×3)淋洗,收集滤饼得到中间体BB-1-2。
1H NMR(400MHz,DMSO_d
6)δ:7.73(d,J=8.8Hz,1H),7.67(d,J=1.6Hz,1H),7.53(dd,J=1.8,8.2Hz,1H),5.62(s,1H)。
步骤2:中间体BB-1-3的合成
将中间体BB-1-2(120g,497.85mmol)溶于乙醇(1.2L),加入乙酸钠(142.94g,1.74mol)和盐酸羟胺(121.09g,1.74mol),氮气保护下升温至80℃搅拌15小时。将反应液降至室温,过滤,滤饼用乙醇(200mL)淋洗,滤液减压浓缩除去大部分乙醇,用碳酸氢钠水溶液调节pH=8,用乙酸乙酯萃取(500mL×2),舍弃有机相;水相用稀盐酸溶液(2N)调节pH至2-3,用乙酸乙酯(1L×2)萃取,有机相用饱和食盐水洗涤(1L),无水硫酸钠干燥,过滤,减压浓缩得到中间体BB-1-3。
1H NMR(400MHz,DMSO_d
6)δ:8.11(d,J=1.2Hz,1H),7.82(d,J=8.4Hz,1H),7.59(dd,J=1.2,8.4Hz,1H),4.11(s,2H)。
步骤3:中间体BB-1的合成
将浓硫酸(9.73g,97.25mmol,5.29mL,纯度:98%)加入中间体BB-1-3(83g,324.15mmol)的乙醇(1L)溶液中,80℃搅拌15小时,补加浓硫酸(0.5mL,纯度:98%),继续搅拌15小时。将反应液降至室温, 减压浓缩除去大部分乙醇,用冰水(500mL)和二氯甲烷(1L)稀释,分液,水相用二氯甲烷(200mL)萃取,合并有机相,依次用饱和碳酸氢钠水溶液(500mL)和饱和食盐水(500mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到中间体BB-1。
1H NMR(400MHz,CDCl
3)δ:7.78(d,J=1.2Hz,1H),7.59(d,J=8.4Hz,1H),7.46(dd,J=1.6,8.4Hz,1H),4.22(q,J=7.2Hz,2H),4.02(s,2H),1.27(t,J=7.2Hz,3H)。
参考例2:片段BB-2
合成路线:
步骤1:中间体BB-2-1的合成
将中间体BB-1(3g,10.56mmol)和氨基甲酸叔丁酯(6.18g,52.80mmol)加入到甲苯(150mL)和水(15mL)中,将磷酸钾(6.72g,31.68mmol),三(二亚苄基丙酮)二钯(676.87mg,739.16μmol)和2-二叔丁基膦基2′,4′,6′-三异丙基联苯(627.76mg,1.48mmol)依次加入上述反应液中,混合物用氮气置换3次后升温至100℃搅拌12小时。反应完毕,反应液冷却至室温,加入水(300mL)稀释,用乙酸乙酯萃取(100mL×3),合并有机相,用饱和食盐水洗涤(100mL×3),无水硫酸钠干燥,过滤,滤液减压浓缩。柱层析分离纯化(洗脱剂:石油醚/乙酸乙酯=1/0-5/1,体积比),得到中间体BB-2-1。
1H NMR(400MHz,CDCl
3)δ:7.89(s,1H),7.56(d,J=8.4Hz,1H),7.14(br d,J=8.8Hz,1H),4.12(q,J=7.1Hz,2H),3.98(s,2H),1.54(s,9H),1.28-1.23(m,3H)。
步骤2:中间体BB-2-2的合成
将中间体BB-2-1(3.5g,10.93mmol)和丙烯酰胺(621.27mg,8.74mmol,603.18μL)加入N,N-二甲基甲酰胺(5mL)中,反应液冷却至0℃,将叔丁醇钾(1.23g,10.93mmol)缓慢分批加入反应液中,混合物用氮气置换3次后在0℃搅拌1小时。反应完毕,反应液缓慢倒入饱和氯化铵(1200mL)中,用乙酸乙酯萃取(200mL×2),合并有机相,依次用水(200mL×2)和饱和食盐水洗涤(200mL×2),收集有机相减压浓缩。粗品经柱层析分离纯化(洗脱剂:石油醚/乙酸乙酯=1/0-1/1,体积比),得到中间体BB-2-2。
1H NMR(400MHz,DMSO_d
6)δ:11.06(s,1H),9.85(s,1H),7.92(s,1H),7.69(d,J=8.8Hz,1H),7.34(dd,J=1.6,8.8Hz,1H),4.51(dd,J=5.0,11.8Hz,1H),2.84-2.68(m,1H),2.65-2.55(m,1H),2.48-2.38(m,1H),2.25-2.13(m,1H),1.50(s,9H)。
步骤3:中间体BB-2-3的合成
将中间体BB-2-2(400mg,1.16mmol)加入到N,N-二甲基甲酰胺(5mL)中,将碘甲烷(246.60mg,1.74mmol,108.16μL)和碳酸钾(320.16mg,2.32mmol)缓慢加入反应液中,混合物氮气置换3次后在25℃搅拌2小时。反应完毕,反应液缓慢倒入水(20mL)中,用乙酸乙酯萃取(20mL×2),合并有机相,用饱和食盐水洗涤(20mL),无水硫酸钠干燥,过滤,减压浓缩。粗品经柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/0-3∶1,体积比),得到中间体BB-2-3。MS-ESI m/z:359.9[M+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:9.85(s,1H),7.93(s,1H),7.69(d,J=8.4Hz,1H),7.33(dd,J=1.4,8.6Hz,1H),4.61(dd,J=4.8,12.0Hz,1H),3.05(s,3H),2.95-2.82(m,1H),2.79-2.69(m,1H),2.44-2.29(m,1H),2.25-2.10(m,1H),1.50(s,9H)。
步骤4:中间体BB-2的盐酸盐的合成
将中间体BB-2-3(45mg,125.22μmol)加入乙酸乙酯(2mL)中,将盐酸/乙酸乙酯溶液(4M,2mL)缓慢加入反应液中,混合物氮气置换3次后在25℃搅拌12小时。反应完毕,反应液减压浓缩,得到中间体BB-2的盐酸盐。
参考例3:片段BB-3
合成路线:
步骤1:中间体BB-3-2的合成
将化合物BB-3-1(15g,69.11mmol)溶于二氯甲烷(400mL)中,降温至0-5℃,加入三氟化硼乙醚(98.08g,691.05mmol),随后滴加醋酸酐(56.95g,557.81mmol,52.24mL),滴加完毕后,反应液在室温下搅拌2小时。反应完毕后,向反应液中加入水(200mL),用氢氧化钠溶液(6M)调节pH至7-8,用二氯甲烷(300mL×3)萃取,合并有机相,用饱和食盐水(1000mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到残余物。残余物用石油醚(200mL)和乙酸乙酯(20mL)打浆搅拌0.5小时后过滤,收集滤饼,得到中间体BB-3-2。MS-ESI m/z:259.0[M+H]
+,261.0[M+2+H]
+。
步骤2:中间体BB-3-3的合成
将中间体BB-3-2(35g,135.09mmol)溶于二氯甲烷(300mL)中,降温至-60至-40℃,滴加三氯化硼/二氯甲烷溶液(1M,135.09mL)。滴加完毕后,氮气保护下升温至0-5℃搅拌2小时。反应完成后,将反应液缓慢倒入冰水(300mL)中,分液,水相用二氯甲烷(200mL×2)萃取,合并有机相,用饱和食盐水(500mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到残余物。残余物用石油醚(200mL)和乙酸乙酯(20mL)打浆搅拌0.5小时后过滤,收集滤饼,得到中间体BB-3-3。
步骤3:中间体BB-3-4的合成
将中间体BB-3-3溶于四氢呋喃(300mL)中,加入碳酸二甲酯(47.05g,522.30mmol,43.97mL),降温至0℃,加入叔丁醇钾(87.91g,783.45mmol)。氮气保护下,升温至70℃搅拌约16小时。反应完毕后,将反应液缓慢倒入冰水(200mL)中,用盐酸溶液(2M)调节pH至8-9,分液后收集水相,将水相继续用盐酸溶液(6M)调节pH至2-3,有固体析出,过滤,滤饼用水(100mL)洗涤,收集滤饼,干燥,得到中间体BB-3-4。
1H NMR(400MHz,DMSO_d
6)δ:12.65(br s,1H),7.90(s,1H),7.16(s,1H),5.49(s,1H),3.94(s,3H)。
步骤4:化合物BB-3-5的合成
在预先干燥的反应瓶中加入中间体BB-3-4(12g,44.27mmol)和乙醇(150mL),随后依次加入盐酸羟胺(10.77g,154.95mmol)和乙醇钠(10.54g,154.95mmol),氮气保护下升温至80℃,搅拌4小时。反应完成后,将反应液减压浓缩得到残余物,向残余物中加入水(200mL),搅拌0.5小时,过滤,滤饼用水(50mL)淋洗,收集滤饼。向滤饼中加入乙酸乙酯(50mL)和石油醚(50mL),搅拌0.5小时后过滤,收集滤饼,得到中间体BB-3-5。MS-ESI m/z:286.0[M+H]
+,288.0[M+2+H]
+。
步骤5:化合物BB-3-6的合成
将中间体BB-3-5(12g,41.95mmol)溶于乙醇(150mL)中,加入浓硫酸(3.68g,36.77mmol,2.00mL,纯度:98%),氮气保护下升温至80℃,搅拌4小时。反应完毕后,将反应液减压浓缩至1/2体积,有固体析出,加入石油醚(100mL),搅拌0.5小时,过滤,收集滤饼,得到中间体BB-3-6。
1H NMR(400MHz,CDCl
3)δ:7.89(s,1H),7.05(s,1H),4.23(q,J=7.2Hz,2H),3.99(s,3H),3.97(s,2H),1.29(t,J=7.2Hz,3H)。
步骤6:中间体BB-3-7的合成
将中间体BB-3-6(5g,15.92mmol)溶于甲苯(100mL)和水(20mL)中,依次加入氨基甲酸叔丁酯(3.73g,31.83mmol),磷酸钾(10.14g,47.75mmol),三(二亚苄基丙酮)二钯(1.17g,1.27mmol)和2-二叔丁膦基-2′,4′,6′-三异丙基联苯(1.08g,2.55mmol),氮气置换三次,升温至100℃,搅拌36小时。反应完毕后,将反应液减压浓缩得到残余物。向残余物中加入水(50mL)和二氯甲烷(50mL),过滤,将滤液分液,水相用二氯甲烷(60mL×3)萃取,合并有机相,用饱和食盐水(150mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到残余物。残余物经过柱层析(洗脱剂:石油醚/二氯甲烷/乙酸乙酯=50/50/1-25/25/1,体积比)分离,得到中间体BB-3-7。
1H NMR(400MHz,DMSO_d
6)δ:8.35(br s,1H),7.27(s,1H),7.02(s,1H),4.23(q,J=7.2Hz,2H),4.00-3.97(m,5H),1.55(s,9H),1.29(t,J=7.2Hz,3H)。
步骤7:中间体BB-3-8的合成
将中间体BB-3-7(0.9g,2.57mmol)溶于二氯甲烷(20mL)中,氮气置换三次,降温至0℃,加入无水三氯化铝(1.71g,12.84mmol,701.88μL),反应混合物升温至40℃搅拌过夜。将反应液降温至0℃,补加无水三氯化铝(1.37g,10.27mmol,561.51μL),随后升温至40℃继续反应过夜。反应完毕后,向反应液中缓慢加入水(20mL),40℃下搅拌0.5小时。分液,水相用二氯甲烷和乙醇的混合溶剂(20mL×5,10∶1,体积比)萃取,合并有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到残余物。残余物经柱层析(洗脱剂:石油醚/乙酸乙酯=10/1-1.5/1,体积比)分离,得到中间体BB-3-8。
1H NMR(400MHz,DMSO_d
6)δ:6.88(s,1H),6.74(s,1H),4.12(q,J=7.2Hz,2H),3.95(s,2H),1.19(t,J=7.2Hz,3H)。
步骤8:中间体BB-3的合成
室温下,将中间体BB-3-8(0.3g,1.27mmol)溶于二氯甲烷(3mL)中,加入氯乙酰氯(172.12mg,1.52mmol,121.21μL)和三乙胺(192.76mg,1.90mmol,265.15μL),反应0.5小时后,减压浓缩得到残余物。向残余物中加入二甲苯(3mL)和吡啶对甲苯磺酸盐(319.15mg,1.27mmol),升温至140℃,继续反应4小时。反应完毕后,将反应液降至室温,减压浓缩得到残余物。向残余物中加入水(10mL),用乙酸乙酯(10mL×3)萃取,合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品。粗品经柱层析(洗脱剂:石油醚/乙酸乙酯=10/1-5/1,体积比)分离纯化,得到中间体BB-3。
1H NMR(400MHz,DMSO_d
6)δ:8.27(s,1H),8.24(s,1H),5.12(s,2H),4.28(s,2H),4.15(q,J=7.1Hz,2H),1.20(t,J=7.0Hz,3H)。
参考例4:片段BB-4
合成路线:
步骤1:中间体BB-4-2的合成
在0-5℃和氮气保护下,将化合物BB-4-1(10g,78.67mmol)溶于二氯甲烷(120mL)和丙酮(60mL),依次缓慢滴加氰基三甲基硅烷(12.45g,125.50mmol,15.70mL),三甲基硅基三氟甲磺酸酯(820.00mg,3.69mmol,666.67μL),反应混合物在25℃搅拌2小时。反应完毕后,冷却至0-5℃,加入水(200mL)稀释, 乙酸乙酯(200mL×3)萃取。合并的有机相经饱和食盐水(100mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压除去溶剂。所得残余物经过柱层析(洗脱剂:石油醚/乙酸乙酯=10/1-5/1,体积比)分离,得到中间体BB-4-2。
1H NMR(400MHz,CDCl
3)δ:6.83(t,J=9.0Hz,1H),6.74(dd,J=2.4,12.0Hz,1H),6.66-6.59(m,1H),1.65(s,6H)。
步骤2:中间体BB-4-3的合成
在室温下和氮气保护下,将中间体BB-4-2(8g,45.40mmol)溶于N,N-二甲基乙酰胺(150mL),4-异硫氰基-2-(三氟甲基)苯甲腈(10.36g,45.40mmol)分批加入反应液中,反应混合物在25℃搅拌12小时,加入甲醇(60mL),稀盐酸(2M,60mL),反应混合物在70℃搅拌3小时,反应完毕后,冷却至室温,加入水(500mL)稀释,用乙酸乙酯(200mL×3)萃取。合并有机相,用饱和食盐水(100mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压除去溶剂。所得残余物经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=3/1-1/1,体积比),得到中间体BB-4-3。
1H NMR(400MHz,CDCl
3)δ:8.02-7.94(m,2H),7.85(dd,J=1.8,8.2Hz,1H),7.19-7.10(m,2H),6.99-6.90(m,2H),1.58(s,6H)。
步骤3:中间体BB-4-4的合成
在室温下和氮气保护下,将中间体BB-4-3(2g,4.72mmol)溶于N,N-二甲基甲酰胺(50mL),依次加入叔丁基-4-(2-溴乙基)哌嗪-1-甲酸酯(1.66g,5.67mmol),碳酸钾(1.31g,9.45mmol),碘化钾(784.17mg,4.72mmol),反应混合物加热至80℃搅拌反应12小时。反应完毕后,冷却至室温,加入水(100mL)稀释,用乙酸乙酯(100mL×3)萃取。合并有机相,用饱和食盐水(100mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压除去溶剂。所得残余物经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=3/1-1/1,体积比),得到中间体BB-4-4。MS-ESI m/z:636.3[M+H]
+。
步骤4:中间体BB-4-5的盐酸盐的合成
在室温和氮气保护下,将中间体BB-4-4(2.0g,3.15mmol)溶于乙酸乙酯(50mL),盐酸/乙酸乙酯(4M,3.93mL)慢慢滴加入反应混合物中,反应混合物在25℃搅拌12小时,反应完毕后,反应混合物减压浓缩,得到中间体BB-4-5的盐酸盐。
1H NMR(400MHz,CD
3OD)δ:8.21-8.12(m,2H),7.98(dd,J=2.0,8.4Hz,1H),7.38(t,J=9.0Hz,1H),7.30(dd,J=2.4,11.6Hz,1H),7.22(br d,J=8.8Hz,1H),4.68-4.59(m,2H),3.86-3.77(m,6H),3.73-3.63(m,4H),1.57(s,6H)。
步骤5:中间体BB-4-6的合成
在室温和氮气保护下,将中间体BB-4-5(1.5g,2.62mmol,盐酸盐)溶于乙腈(50mL),依次加入溴乙酸乙酯(875.85mg,5.24mmol,580.04μL),碳酸钾(724.86mg,5.24mmol),反应混合物加热至80℃搅拌反应5小时,反应完毕后,冷却至室温,加入水(100mL)稀释,用乙酸乙酯(100mL×3)萃取。合并有机相,用饱和食盐水(100mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压除去溶剂。所得残余物经过柱 层析分离(洗脱剂:石油醚/乙酸乙酯=3/1-1/1,体积比),得到中间体BB-4-6。MS-ESI m/z:622.2[M+H]
+。
步骤6:中间体BB-4的合成
在室温和氮气保护下,中间体BB-4-6(1.5g,2.42mmol)溶于乙醇(50mL),氢氧化锂(1M,7.25mL)缓慢滴加入反应混合物中,反应混合物在25℃搅拌12小时。反应完毕,反应混合物用盐酸溶液(2M)调节pH至2-3,减压浓缩,得到中间体BB-4。MS-ESI m/z:594.1[M+H]
+。
参考例5:片段BB-5
合成路线:
步骤1:中间体BB-5-2的合成
将化合物N-[((1,3-反式)-3-羟基-2,2,4,4-四甲基环丁基]氨基甲酸叔丁酯(0.8g,3.29mmol)加入N,N-二甲基甲酰胺(10mL)中,将反应体系降温至0℃,缓慢地加入氢化钠(262.98mg,6.58mmol,纯度:60%),氮气置换三次后,搅拌0.5小时,加入化合物BB-5-1(613.68mg,3.95mmol),升温至70℃下反应1小时。反应完毕,向反应液中加入水(10mL),室温下搅拌0.5小时,用乙酸乙酯(15mL×3)萃取,有机相用饱和食盐水(15mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品经柱层析(洗脱剂:石油醚/乙酸乙酯=8/1-4/1,体积比)分离纯化,得到中间体BB-5-2。MS-ESI m/z:379.1[M+H]
+。
1H NMR(400MHz,CDCl
3)δ:7.56(d,J=8.8Hz,1H),6.94(d,J=2.4Hz,1H),6.78(dd,J=2.4Hz,8.8Hz,1H),4.67(br d,J=7.6Hz,1H),3.68(br d,J=8.4Hz,1H),1.46(s,9H),1.19(s,6H),1.15(s,6H)。
步骤2:中间体BB-5的盐酸盐的合成
将中间体BB-5-2(0.6g,1.58mmol)加入盐酸/乙酸乙酯溶液(4M,5mL)中,室温下反应1小时。反应完毕,过滤,滤饼用乙酸乙酯(40mL)淋洗,收集滤饼,得到中间体BB-5的盐酸盐。MS-ESI m/z:279.1[M+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:8.52(br s,3H),7.89(d,J=8.8Hz,1H),7.21(d,J=2.4Hz,1H),6.99(dd,J=2.4,8.8Hz,1H),4.36(s,1H),3.06(s,1H),1.31(s,6H),1.08(s,6H)。
参考例6:片段BB-6
合成路线:
步骤1:中间体BB-6-2的合成
将化合物BB-6-1(15.10g,89.80mmol,13.25mL)加入至二甲基亚砜(200mL)中,依次加入碳酸钾(31.03g,224.51mmol)和1-叔丁氧羰基哌嗪(20g,89.80mmol),氮气置换三次后,升温至120℃,反应过夜。反应完毕,将反应液倒入水(250mL)中,用乙酸乙酯(200mL×3)萃取,有机相用饱和食盐水(300mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品,所得粗品经柱层析(洗脱剂:石油醚/乙酸乙酯=8/1-3/1,体积比)分离纯化,得到中间体BB-6-2。MS-ESI m/z:335.2[M+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:7.79(d,J=8.8Hz,2H),6.97(d,J=9.2Hz,2H),4.24(q,J=7.2Hz,2H),3.48-3.42(m,4H),3.32-3.26(m,4H),1.42(s,9H),1.28(t,J=7.0Hz,3H)。
步骤2:中间体BB-6-3的合成
将中间体BB-6-2(4g,11.96mmol)加入至四氢呋喃(20mL)和水(20mL)中,加入氢氧化钠(956.84mg,23.92mmol),氮气置换三次后,升温至70℃,反应过夜。反应完毕,将反应体系减压浓缩得到残余物,向残余物中加入乙酸乙酯(40mL)和水(30mL),分液,收集水相,用盐酸溶液(1M)调节pH至5-6,用乙酸乙酯(50mL×2)萃取,收集有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,得到中间体BB-6-3。MS-ESI m/z:307.2[M+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:7.78(d,J=8.8Hz,2H),6.95(d,J=8.8Hz,2H),3.49-3.40(m,4H),3.32-3.22(m,4H),1.42(s,9H)。
步骤3:中间体BB-6的盐酸盐的合成
将中间体BB-6-3(3.2g,10.45mmol)加入至盐酸/乙酸乙酯溶液(4M,30mL)中,室温下反应2小时。反应完毕,过滤,滤饼用乙酸乙酯(50mL)淋洗,收集滤饼,得到中间体BB-6的盐酸盐。MS-ESI m/z:207.2[M+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:9.57(br s,2H),7.80(d,J=9.2Hz,2H),7.02(d,J=9.2Hz,2H),3.60-3.50(m,4H),3.17(br s,4H)。
参考例7:片段BB-7
合成路线:
步骤1:中间体BB-7-2的合成
将化合物BB-7-1(200.00g,930.04mmol),碳酸二甲酯(335.10g,3.72mol,313.18mL)溶于四氢呋喃(2L)中,降温至0℃,分批加入叔丁醇钾(626.17g,5.58mol),缓慢升温至20℃搅拌0.5小时,随后升温至70℃搅拌12小时。反应完毕,将反应液降至室温,减压浓缩除去溶剂,加入冰水(3L)溶解,用甲基叔丁基醚(1L×2)萃取,收集水相,有机相用氢氧化钠水溶液(2N,500mL×3)洗涤,收集水相。合并两次水相,用盐酸溶液(6N)调节pH至2-3,析出固体,过滤,滤饼用水(500mL×3)淋洗,收集滤饼,得到中间体BB-7-2。
1H NMR(400MHz,DMSO_d
6)δ:12.85(br s,1H),7.89(d,J=2.4Hz,1H),7.79(dd,J=2.6Hz,8.6Hz,1H),7.35(d,J=8.8Hz,1H),5.65(s,1H)。
步骤2:中间体BB-7-3的合成
将中间体BB-7-2(255g,1.06mol)溶于乙醇(2.5L)中,20℃下加入盐酸羟胺(257.30g,3.70mol)和乙酸钠(303.74g,3.70mol),搅拌1小时。升温至80℃搅拌12小时。降至室温,过滤,滤饼用乙酸乙酯(400mL×3)淋洗,固体丢弃,母液浓缩至剩余1/3,用碳酸氢钠水溶液调节pH至8,用乙酸乙酯(2L)萃取,收集水相,有机相用饱和碳酸氢钠水溶液(300mL×3)洗涤,收集水相。合并两次水相,用盐酸溶液(2N)调节pH至2,有固体析出,用乙酸乙酯(2L×3)溶解并萃取,合并有机相,用饱和食盐水(1L)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到中间体BB-7-3。
步骤3:中间体BB-7的合成
将中间体BB-7-3(120g,468.65mmol)溶于乙醇(1200mL)中,加入硫酸(9.20g,91.92mmol,5mL,纯度:98%),升温至80℃搅拌16小时。反应完毕后,反应液降至室温,减压浓缩,加入乙酸乙酯(600mL),用饱和碳酸氢钠水溶液调节pH至8,分液,有机相用饱和碳酸氢钠水溶液(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到中间体BB-7。
1H NMR(400MHz,CDCl
3)δ:7.88(d,J=1.6Hz,1H),7.66(dd,J=1.8,9.0Hz,1H),7.48(d,J=9.2Hz,1H),4.24(q,J=7.2Hz,2H),4.02(s,2H),1.29(t,J=7.2Hz,3H)。
参考例8:片段BB-8
合成路线:
步骤1:中间体BB-8-1的合成
将中间体BB-1(6g,21.12mmol)加入至二氧六环(60mL)中,依次加入双联嚬哪醇硼酸酯(6.44g,25.34mmol),[1,1-双(二苯基膦)二茂铁]二氯化钯二氯甲烷(1.72g,2.11mmol)和乙酸钾(8.29g,84.48mmol),氮气置换三次后,升温至100℃,反应5小时。反应完毕,将反应液倒入水(55mL)中,用乙酸乙酯(65mL×3)萃取,合并有机相,用饱和食盐水(50mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品经柱层析(洗脱剂:石油醚/乙酸乙酯=10/1-6/1,体积比)分离纯化,得到中间体BB-8-1。MS-ESI m/z:332.2[M+H]
+。
步骤2:中间体BB-8-2的合成
将中间体BB-8-1(8g,24.16mmol)加入至四氢呋喃(80mL)和水(40mL)中,加入碳酸氢钠(4.06g,48.31mmol),将反应体系降温至0℃,缓慢地滴加双氧水(14.32g,126.27mmol,12.13mL,纯度:30%),滴加完毕后,保温反应2小时。反应完毕后,加入15%亚硫酸钠水溶液(50mL),搅拌10分钟,用盐酸溶液(1N)调节pH至5-6,用乙酸乙酯(50mL×3)萃取,合并有机相,用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品经柱层析(洗脱剂:石油醚/乙酸乙酯=9∶1-4∶1,体积比)分离纯化,得到中间体BB-8-2。MS-ESI m/z:222.1[M+H]
+。
1H NMR(400MHz,CDCl
3)δ:7.49(d,J=8.4Hz,1H),6.93(d,J=1.6Hz,1H),6.85(dd,J=2.0,8.4Hz,1H),4.23(q,J=7.1Hz,2H),4.00(s,2H),1.28(t,J=7.0Hz,3H)。
步骤3:中间体BB-8-3的合成
将中间体BB-8-2(4.5g,20.34mmol)加入至N,N-二甲基甲酰胺(50mL)中,依次加入碳酸钾(8.43g,61.03mmol,3eq),碘化钠(304.93mg,2.03mmol)和6-溴-1-己烯(3.98g,24.41mmol,3.26mL),氮气置换三次后,升温至80℃,反应5小时。反应完毕后,将反应液倒入水(60mL)中,用乙酸乙酯(50mL×3)萃取,合并有机相,用饱和食盐水(70mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品经柱层析(洗脱剂:石油醚/乙酸乙酯=9/1-4/1,体积比)分离纯化,得到中间体BB-8-3。MS-ESI m/z:304.2[M+H]
+。
步骤4:中间体BB-8-4的合成
将中间体BB-8-3(3.2g,10.55mmol)加入至四氢呋喃(30mL)中,依次加入丙烯酰胺(899.74mg,12.66mmol)和叔丁醇钾(1.54g,13.71mmol),氮气置换三次后,室温下反应1小时。反应完毕后,将反应液倒入盐酸溶液(1M,20mL)中,加入水(20mL),用乙酸乙酯(25mL×3)萃取,合并有机相,用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品经柱层析(洗脱剂:石油醚/乙酸乙酯=5/1-1/1,体积比)分离纯化,得到中间体BB-8-4。MS-ESI m/z:329.1[M+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:11.08(s,1H),7.68(d,J=8.8Hz,1H),7.29(d,J=1.6Hz,1H),6.97(dd,J=1.6,8.8Hz,1H),5.89-5.75(m,1H),5.04(dd,J=1.4,17.0Hz,1H),4.97(dd,J=1.0,10.2Hz,1H),4.51(dd,J=5.0Hz,11.8Hz,1H),4.08(t,J=6.4Hz,2H),2.81-2.69(m,1H),2.64-2.55(m,1H),2.48-2.40(m,1H),2.23-2.14(m,1H),2.13-2.05(m,2H),1.81-1.71(m,2H),1.53-1.46(m,2H)。
步骤5:中间体BB-8的合成
将中间体BB-8-4(0.4g,1.22mmol)加入至四氢呋喃(9mL)和水(3mL)中,降温至0℃,缓慢加入二水合锇酸钾(44.88mg,121.82μmol)和高碘酸钠(1.04g,4.87mmol),室温反应1小时。反应完毕后,将反应液倒入饱和亚硫酸钠溶液(10mL)和水(30mL)中,用乙酸乙酯(10mL×3)萃取,合并有机相,用饱和食盐水(15mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到中间体BB-8。MS-ESI m/z:331.1[M+H]
+。
参考例9:片段BB-9
合成路线:
步骤1:中间体BB-9-2的合成
将化合物BB-9-1(5g,23.25mmol,1eq)和碳酸二甲酯(8.38g,93.00mmol,7.83mL,4eq)溶于四氢呋喃(200mL)中,冰浴降温至0-5℃,分批加入叔丁醇钾(15.65g,139.51mmol,6eq),加料结束后升温至70℃反应20小时,随后升温至90℃反应15小时。将反应液降温至室温,倒入盐酸溶液(1N,150mL)中,用乙酸乙酯(100mL×3)萃取,合并有机相,用饱和食盐水(150mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品中间体BB-9-2。
1H NMR(400MHz,CDCl
3)δ:12.50(s,1H),7.80-7.78(m,1H),7.67-7.65(m,1H),6.88-6.84(m,1H),4.05(s,2H),3.78(s,3H)。
步骤2:中间体BB-9-3的合成
将中间体BB-9-2(7g)和碳酸钾(4.70g,34.02mmol,1.5eq)加入N,N-二甲基甲酰胺(70mL)中,升温至90℃,敞口反应过夜。降温至室温,过滤,滤饼依次用水(50mL)和乙醇(50mL)淋洗,收集滤饼,得到中间体BB-9-3。MS-ESI m/z:241.1[M+H]
+,243.1[M+2+H]
+。
步骤3:中间体BB-9-4的合成
将中间体BB-9-3(5.1g,21.16mmol,1eq),盐酸羟胺(8.09g,116.37mmol,5.5eq)和乙酸钠(9.55g,116.37mmol,5.5eq)溶于乙醇(60mL)中,升温至90℃反应过夜。将反应液降至室温,减压浓缩除去大部分溶剂,倒入水(100mL)中,用乙酸乙酯(100mL×3)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂,得到中间体BB-9-4。MS-ESI m/z:256.0[M+H]
+,258.1[M+2+H]
+。
步骤4:中间体BB-9-5的合成
将中间体BB-9-4(2.7g,10.54mmol,1eq)和浓硫酸(1.88g,18.76mmol,1.02mL,纯度:98%,1.78eq)加入到乙醇(40mL)中,升温至90℃反应过夜。将反应液降温至40-50℃,减压浓缩除去溶剂,残余物用饱和碳酸氢钠溶液调节pH至5-6,用乙酸乙酯(30mL)萃取,有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品。粗品经柱层析(洗脱剂:乙酸乙酯/石油醚=1/5)分离纯化,得到中间体BB-9-5。
1H NMR(400MHz,CDCl
3)δ:7.73(d,J=7.6Hz,1H),7.67(d,J=8.0Hz,1H),7.23(t,J=8.0Hz,1H),4.22(q,J=7.2Hz,2H),4.04(s,2H),1.27(q,J=7.2Hz,3H)。
步骤5:中间体BB-9-6的合成
将中间体BB-9-5(1.7g,5.98mmol)加入至二氧六环(20mL)和水(4mL)中,依次加入频哪醇乙烯基硼酸酯(1.11g,7.18mmol,1.22mL),碳酸钾(1.65g,11.97mmol)和[1,1-双(二苯基膦)二茂铁]二氯化钯二氯甲烷(244.33mg,299.18μmol),氮气置换三次后,升温至90℃,反应4小时。反应完毕,将反应液倒入水(25mL)中,用乙酸乙酯(35mL×3)萃取,有机相用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品,粗品经柱层析(洗脱剂:石油醚/乙酸乙酯=10/1-5/1,体积比)分离纯化,得到中间体BB-9-6。MS-ESI m/z:232.2[M+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:7.75(d,J=7.6Hz,1H),7.69(d,J=7.2Hz,1H),7.39(t,J=7.6Hz,1H),6.97(dd,J=11.4,17.8Hz,1H),6.26(d,J=17.6Hz,1H),5.63(d,J=11.2Hz,1H),4.23(s,2H),4.14(q,J=7.2Hz,2H),1.19(t,J=7.0Hz,3H)。
步骤6:中间体BB-9-7的合成
将中间体BB-9-6(0.6g,2.59mmol)加入至四氢呋喃(13mL)中,加入丙烯酰胺(221.30mg,3.11mmol)和叔丁醇钾(320.26mg,2.85mmol),氮气置换三次后,室温下反应1小时。反应完毕后,将反应液倒入盐酸溶液中(1M,10mL),加入水(10mL),用乙酸乙酯(15mL×3)萃取,合并有机相,用饱和食盐水(15mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品,粗品经柱层析(洗脱剂:石油醚/乙酸乙酯=5/1-1/1,体积比)分离纯化,得到中间体BB-9-7。MS-ESI m/z:257.1[M+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:10.12(br s,1H),7.78(d,J=8.0Hz,1H),7.70(d,J=7.2Hz,1H),7.39(d,J=7.6Hz,1H),6.99(dd,J=11.2,17.6Hz,1H),6.27(d,J=17.6Hz,1H),5.65(d,J=11.6Hz,1H),4.64(dd,J=4.8,12.0Hz,1H),2.84-2.72(m,1H),2.68-2.59(m,1H),2.59-2.51(m,1H),2.27-2.16(m,1H)。
步骤7:中间体BB-9的合成
将化合物BB-9-7(0.16g,624.38μmol)加入至四氢呋喃(6mL)和水(2mL)中,降温至0℃,缓慢加入二水合锇酸钾(23.01mg,62.44μmol)和高碘酸钠(534.19mg,2.50mmol),氮气置换三次后,室温反应1小时。反应完毕后,将反应液倒入水(5mL)中,用乙酸乙酯(10mL×3)萃取,有机相用饱和食盐水(5mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到中间体BB-9。MS-ESI m/z:259.1[M+H]
+。
参考例10:片段BB-10
合成路线:
步骤1:中间体BB-10-1的合成
将4-哌啶乙醇(4.8g,37.15mmol)加入至N,N-二甲基甲酰胺(60mL)中,依次加入碳酸钾(15.40g,111.46mmol)和BB-6-1(6.25g,37.15mmol,5.48mL),氮气置换三次后,升温至110℃搅拌16小时。反应完毕,将反应液倒入水(70mL)中,用乙酸乙酯(65mL×3)萃取,有机相用饱和食盐水(70mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品,粗品经柱层析(洗脱剂:石油醚/乙酸乙酯=4/1-1/1,体积比)分离纯化,得到中间体BB-10-1。MS-ESI m/z:278.2[M+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:7.76(d,J=8.8Hz,2H),6.93(d,J=9.2Hz,2H),5.75(s,1H),4.23(q,J=7.2Hz,2H),3.87(d,J=12.8Hz,2H),3.50-3.44(m,2H),2.82-2.75(m,2H),1.74-1.70(m,2H),1,67-1,55(m,1H),1.40-1.36(m,2H),1.28(t,J=7.2Hz,3H),1.20-1.14(m,2H)。
步骤2:中间体BB-10-2的合成
将中间体BB-10-1(4.7g,16.95mmol)加入至二氯乙烷(60mL)中,将反应体系降温至0℃,依次加入N,N-二异丙基乙胺(6.57g,50.84mmol,8.85mL),4-二甲基氨基吡啶(207.02mg,1.69mmol)和甲磺酰氯(3.26g,28.46mmol,2.20mL),氮气置换三次后,保温反应1小时。反应完毕后,将反应液倒入水(50mL)中,用二氯甲烷(45mL×3)萃取,合并有机相,用饱和食盐水(50mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到中间体BB-10-2。MS-ESI m/z:356.2[M+H]
+。
步骤3:中间体BB-10-3的合成
将1-叔丁氧羰基哌嗪(5.26g,23.63mmol)加入至乙腈(70mL)中,依次加入碳酸钾(5.44g,39.39mmol)和中间体BB-10-2(7g,19.69mmol),氮气置换三次后,升温至60℃,反应过夜。反应完毕后,将反应液倒入水(100mL)中,用乙酸乙酯(75mL×3)萃取,合并有机相,用饱和食盐水(60mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品,粗品经柱层析(洗脱剂:石油醚/乙酸乙酯=5/1-1/1,体积比)分离纯化,得到中间体BB-10-3。MS-ESI m/z:446.4[M+H]
+。
1H NMR(400MHz,CD
3OD)δ:7.84(br d,J=8.8Hz,2H),6.91(br d,J=8.8Hz,2H),4.29(q,J=7.1Hz,2H),3.89(br d,J=12.8Hz,2H),3.43(br s,4H),2.81(br t,J=12.4Hz,2H),2.48-2.34(m,6H),1.79(br d,J=12.4Hz,2H),1.56-1.48(m,3H),1.45(s,9H),1.35(t,J=7.0 Hz,3H),1.31-1.24(m,2H)。
步骤4:中间体BB-10-4的合成
将中间体BB-10-3(3g,6.73mmol)加入至四氢呋喃(10mL)和水(10mL)中,加入氢氧化钠(1.35g,33.66mmol),氮气置换三次后,升温至75℃,反应48小时。反应完毕后,将反应体系减压浓缩得到残余物,向残余物中加入乙酸乙酯(40mL)和水(30mL),萃取分液,收集水相,用盐酸溶液(1M)调节pH至5-6,减压浓缩得到中间体BB-10-4。MS-ESI m/z:418.3[M+H]
+。
步骤5:中间体BB-10的盐酸盐的合成
将中间体BB-10-4(2.4g,5.75mmol)加入至盐酸/乙酸乙酯(4N,20mL)中,室温下搅拌2小时。反应完毕后,过滤,滤饼用乙酸乙酯(20mL)淋洗,收集滤饼,得到中间体BB-10的盐酸盐。MS-ESI m/z:318.2[M+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:12.10(br s,1H),10.18(br s,2H),7.87(br d,J=8.4Hz,2H),7.39(br s,2H),3.80-3.63(m,4H),3.53-3.25(m,6H),3.24-3.00(m,4H),1.83(br d,J=12.4Hz,2H),1.76-1.61(m,3H),1.60-1.30(m,2H)。
参考例11:片段BB-11
合成路线:
步骤1:中间体BB-11-2的盐酸盐的合成
化合物BB-11-1(5g,18.43mmol)溶于乙酸乙酯(20mL),20℃下加入盐酸/乙酸乙酯溶液(4M,23.03mL),反应液在20℃继续搅拌12小时,反应完毕,反应液减压浓缩,得到中间体BB-11-2的盐酸盐。
1H NMR(400MHz,DMSO_d
6)δ:9.45-8.62(m,2H),4.06(q,J=7.2Hz,2H),3.18(br d,J=12.8Hz,2H),2.91-2.74(m,2H),2.26(d,J=6.8Hz,2H),2.02-1.88(m,1H),1.77(br d,J=12.8Hz,2H),1.49-1.34(m,2H),1.18(t,J=7.0Hz,3H)。
步骤2:中间体BB-11-3的合成
中间体BB-11-2(2g,9.63mmol,盐酸盐)溶于N,N-二甲基甲酰胺(50mL),将1-溴-2-氯-乙烷(13.81g,96.29mmol,7.98mL),N,N-二异丙基乙胺(3.73g,28.89mmol,5.03mL)依次加入反应液中,反应混合物在25℃搅拌12小时。反应完毕,加入冰水(100mL)和乙酸乙酯(100mL),分离的有机相经饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压除去溶剂,所得残余物经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/1,体积比),得到中间体BB-11-3。
1H NMR(400MHz,CDCl
3)δ:4.13(q,J=7.1Hz,2H),3.58(t,J=7.2Hz,2H),2.90(br d,J=11.6Hz,2H),2.71(t,J=7.2Hz,2H),2.23(d,J=6.8Hz,2H),2.15-2.04(m,2H),1.76-1.66(m,3H),1.40-1.28(m,2H),1.26(t,J=7.2Hz,3H)。
步骤3:中间体BB-11-4的合成
在室温和氮气保护下,将中间体BB-11-3(1.10g,4.72mmol,1eq),BB-4-3(2g,4.72mmol),碘化钾(784.17mg,4.72mmol)和碳酸钾(1.31g,9.45mmol)依次加入到N,N-二甲基甲酰胺(50mL),反应混合物在80℃搅拌12小时,反应完毕,反应混合物降至室温,加入冰水(100mL)和乙酸乙酯(100mL),分离的有机相经饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压除去溶剂,所得残余物经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/1,体积比),得到中间体BB-11-4。MS-ESI m/z:621.2[M+H]
+。
步骤4:中间体BB-11的合成
将中间体BB-11-4(1.5g,2.42mmol)溶于乙醇(50mL),加入氢氧化锂溶液(1M,7.25mL),反应混合物在25℃搅拌12小时。反应完毕,反应混合物用稀盐酸(2M,10mL)调节pH至2-3,反应液减压浓缩,得到中间体BB-11。MS-ESI m/z:593.3[M+H]
+。
参考例12:片段BB-12
合成路线:
步骤1:中间体BB-12-1的合成
将中间体BB-7(5g,17.60mmol)和氨甲酸叔丁酯(10.31g,88.00mmol)加到甲苯(25mL)和水(2.5mL)中,将磷酸钾(11.21g,52.80mmol,3eq),三(二亚苄基丙酮)二钯(1.13g,1.23mmol)和2-二-叔丁膦基-2′,4′,6′-三异丙基联苯(1.05g,2.46mmol)依次缓慢加入反应中,混合物用氮气置换3次后升温至100℃搅拌12小时。反应液冷却至室温,减压浓缩除去大部分有机溶剂。用乙酸乙酯稀释(300mL),有机相用水(300mL×2)洗涤,收集有机相,用无水硫酸钠干燥,过滤,减压浓缩。所得粗品经柱层析(洗脱剂:石油醚/乙酸乙酯=10/1-4/1)分离,得到中间体BB-12-1(纯度:67.60%)。MS-ESI m/z:320.8[M+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:9.86(s,1H),7.92(s,1H),7.66(d,J=8.4Hz,1H),7.35(dd,J=1.8,8.6Hz,1H),4.18-4.07(m,4H),1.50(s,9H),1.18(t,J=7.2Hz,5H)。
步骤2:中间体BB-12-2的合成
0℃下,将中间体BB-12-1(0.5g,1.06mmol,纯度:67.60%)和丙稀酰胺(60.00mg,844.10μmol,58.25μL)加到N,N-二甲基甲酰胺(10mL)中,将叔丁醇钾(118.40mg,1.06mmol)缓慢分批加入反应中,混合物氮气置换3次后在0℃搅拌1小时。反应液慢慢加入饱和氯化铵中(20mL),用乙酸乙酯(20mL×2)萃取,合并有机相,用饱和食盐水洗涤(20mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩。所得粗品经柱层析(洗脱剂:石油醚/乙酸乙酯=10/1-1/1)分离,得到中间体BB-12-2(纯度:83.94%)。MS-ESI m/z:346.2[M+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:11.06(s,1H),9.85(s,1H),7.91(s,1H),7.67(d,J=8.8Hz,1H),7.32(dd,J=1.6,8.8Hz,1H),4.50(dd,J=5.0,11.8Hz,1H),2.80-2.68(m,1H),2.63-2.53(m,1H),2.46-2.37(m,1H),2.27-2.10(m,1H),1.49(s,9H)。
步骤3:中间体BB-12的合成
将中间体BB-12-2(50mg,121.53μmol,纯度:83.94%)加入乙酸乙酯(5mL)中,将盐酸/乙酸乙酯溶液(4M,2mL)缓慢加入反应中,混合物氮气置换3次后在25℃搅拌12小时。反应液减压浓缩,得到中间体BB-12的盐酸盐。
参考例13:片段BB-13
合成路线:
步骤1:中间体BB-13-1的合成
室温下,将中间体BB-4-3(0.5g,1.18mmol)和4-甲基磺酰氧基哌啶-1-羧酸叔丁酯(494.86mg,1.77mmol)加到N,N-二甲基甲酰胺(5mL)中,随后加入碳酸钾(326.43mg,2.36mmol),反应液在氮气氛围下,80℃搅拌12小时。反应完毕,加入乙酸乙酯(20mL)萃取,依次使用水(20mL×2)和饱和食盐水(20mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残余物经柱层析分离(石油醚/乙酸乙酯=1/0-5/1,体积比),得到中间体BB-13-1。MS-ESI m/z:507.1[M-100+H]
+。
步骤2:中间体BB-13-2的盐酸盐的合成
将中间体BB-13-1(0.6g,849.71μmol)加入到乙酸乙酯(10mL)中,将盐酸/乙酸乙酯(4M,4mL)缓慢的加到反应中,混合物用氮气置换3次后在25℃搅拌12小时。反应完毕,反应液减压浓缩,得到中间体BB-13-2的盐酸盐。
步骤3:中间体BB-13-3的合成
将中间体BB-13-2(0.5g,987.14μmol,盐酸盐)和溴代乙酸乙酯(329.71mg,1.97mmol,218.35μL)加到乙腈(10mL)中,将碳酸钾(272.86mg,1.97mmol)缓慢的加到反应中,混合物用氮气置换3次后在80℃搅拌2小时。反应液冷却至25℃,减压浓缩除去大部分有机溶剂。加入乙酸乙酯(20mL)和水(20mL),分离有机相,水相用乙酸乙酯萃取(20mL×2),合并有机相,用无水硫酸钠干燥,过滤,减压浓缩,所得残余物经柱层析分离(石油醚/乙酸乙酯=1/0-1/1,体积比),得到中间体BB-13-3。MS-ESI m/z:593.1[M+H]
+。
步骤4:中间体BB-13的合成
将中间体BB-13-3(500mg,843.73μmol)加到乙醇(5mL)和水(1mL)中,将一水合氢氧化锂(70.81 mg,1.69mmol)缓慢的加入反应中,混合物用氮气置换3次后在25℃搅拌2小时。反应液减压浓缩除去大部分有机溶剂,加入水(20mL),水相用稀盐酸溶液调节pH至2,水相冻干,得到中间体BB-13。MS-ESI m/z:565.5[M+H]
+。
参考例14:片段BB-14
合成路线:
步骤1:中间体BB-14的盐酸盐的合成
将中间体BB-2-2(50mg,121.53μmol)加到乙酸乙酯(5mL)中,盐酸/乙酸乙酯(4M,2mL)缓慢加到反应中,混合物氮气置换3次后在25℃搅拌12小时。反应液减压浓缩,得到中间体BB-14的盐酸盐。
参考例15:片段BB-15
合成路线:
步骤1:中间体BB-15-2的合成
在预先干燥的反应瓶中加入N-苄基-双邻氯乙基氨基盐酸盐(24.33g,90.57mmol),乙醇(150mL),化合物BB-15-1(24.33g,90.57mmol)和N,N-二异丙基乙胺(117.05g,905.70mmol,157.75mL),反应体系升温至90℃搅拌16小时。反应结束后,减压浓缩除去溶剂,向残余物中加入水(100mL),用二氯甲烷(100mL×3)萃取,合并有机相,用饱和食盐水(300mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂,所得残余物经过柱层析(洗脱剂:石油醚/乙酸乙酯=50/1-10/1,体积比)分离纯化,得到中间体BB-15-2。MS-ESI m/z:289.3[M+H]
+。
1H NMR(400MHz,CDCl
3)δ:7.29-7.18(m,5H),4.11-4.06(q,J=6.8Hz,2H),3.45(s,2H),2.92(s,4H),2.31(s,4H),1.23-1.19(m,5H),0.85-0.83(m,2H)。
步骤2:中间体BB-15-3的盐酸盐的合成
将中间体BB-15-2(8g,27.74mmol)溶于二氯甲烷(80mL)中,0℃加入氯甲酸氯乙酯(5.79g,40.50mmol),反应体系升温至20℃搅拌1小时。反应完全,将反应液减压浓缩除去溶剂。残余物重新溶解于乙醇(80mL)中,85℃继续反应16小时。反应结束后,反应液减压浓缩除去溶剂。向残余物中加入乙酸乙酯(80mL),搅拌20分钟后过滤,收集滤饼,得到中间体BB-15-3的盐酸盐。
1H NMR(400MHz,CD
3OD)δ:4.22-4.11(m,2H),3.59-3.35(m,4H),3.31-3.12(m,4H),1.57-1.27(m,4H),1.27-1.18(m,3H)。
步骤3:化合物BB-15-4的合成
将中间体BB-15-3(3.5g,14.91mmol,盐酸盐)溶于二氧六环(30mL)和水(10mL)的混合溶剂中,反应体系降温至0℃,加入碳酸氢钠(3.76g,44.73mmol,1.74mL)和碳酸叔丁酯(3.25g,14.91mmol,3.43mL),恢复至20℃搅拌12小时。反应结束后,反应液加水(20mL)和二氯甲烷(20mL),分离有机相,水相加二氯甲烷(20mL×3)萃取,合并有机相,用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。所得残余物经过柱层析(洗脱剂:石油醚/乙酸乙酯=0/1-25/2,体积比)分离纯化,得到中 间体BB-15-4。
1H NMR(400MHz,CDCl
3)δ:4.12(q,J=7.1Hz,2H),3.31(br s,4H),2.90(br s,4H),1.47(s,9H),1.32-1.27(m,2H),1.25(t,J=7.2Hz,3H),0.94(q,J=3.7Hz,2H)。
步骤4:中间体BB-15-5的合成
将中间体BB-15-4(3.1g,10.39mmol)溶于四氢呋喃(35mL)中,氮气置换三次后,降温至-65℃,缓慢加入四氢铝锂(1M四氢呋喃溶液,31.17mL),0℃搅拌1小时。反应结束后,在0℃缓慢向反应液中加入十水硫酸钠(30g),过滤,滤饼用二氯甲烷淋洗,滤液减压浓缩,得到中间体BB-15-5。
1H NMR(400MHz,CDCl
3)δ:3.58(s,2H),3.35(t,J=4.8Hz,4H),2.69(t,J=4.8Hz,4H),1.45(s,9H),0.71-0.68(m,2H),0.55-0.52(m,2H)。
步骤5:中间体BB-15-6的合成
氮气保护下,将中间体BB-4-3(2.39g,5.66mmol)溶于四氢呋喃(50mL)中,然后加入中间体BB-15-5(2.9g,11.31mmol)和三苯基膦(2.97g,11.31mmol),反应体系降温至0℃,滴加偶氮二甲酸二异丙酯(2.29g,11.31mmol,2.20mL)。升温至40℃搅拌12小时。反应结束后,向反应液中加水(20mL)和乙酸乙酯(50mL),分离有机相,水相用乙酸乙酯(50mL×3)萃取,合并有机相,用饱和食盐水(50mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂,所得残余物经过柱层析(洗脱剂:石油醚/乙酸乙酯=1/0-4/5,体积比)分离纯化,得到中间体BB-15-6。
1H NMR(400MHz,CDCl
3)δ:8.00-7.96(m,2H),7.85-7.82(m,1H),7.08-7.02(m,3H),4.05(s,2H),2.81(s,4H),2.69(s,4H),1.59(s,6H),1.46(s,9H),0.75-0.70(m,4H)。
步骤6:中间体BB-15-7的盐酸盐的合成
将中间体BB-15-6(3.2g,4.84mmol)溶于乙酸乙酯(5mL)中,加入盐酸/乙酸乙酯(4M,30mL),15℃下反应2小时。将反应液减压浓缩除去溶剂,加入甲基叔丁基醚(10mL)搅拌5分钟,过滤,滤饼用甲基叔丁基醚(2mL×2)淋洗,收集滤饼减压浓缩除去溶剂,得到中间体BB-15-7的盐酸盐。
步骤7:化合物BB-15-8的合成
将中间体BB-15-7(0.2g,334.42μmol,盐酸盐)溶于四氢呋喃(3mL),氮气保护下降至0℃,滴加三乙胺(203.04mg,2.01mmol,279.28μL),加完后滴加溴乙酸乙酯(111.70mg,668.84μmol,73.97μL)的四氢呋喃(1mL)溶液,缓慢升至20℃搅拌2小时。向反应液中加水(10mL),用乙酸乙酯萃取(10mL×2),有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,减压除去溶剂,得到中间体BB-15-8。
步骤8:中间体BB-15的合成
将中间体BB-15-8(210mg,324.23μmol)溶于四氢呋喃(2mL)和水(0.5mL)的混合溶剂中,降至0℃,加入一水合氢氧化锂(68.02mg,1.62mmol),加完后20℃搅拌1小时。用盐酸溶液(1M)调节反应液pH至3-4,减压浓缩,得到中间体BB-15。
参考例16:片段BB-16
合成路线:
步骤1:中间体BB-16-1的合成
将中间体BB-4-5(600mg,1.12mmol)和溴丙酸乙酯(405.62mg,2.24mmol,285.65μL)加到乙腈(10mL)中,随后加入碳酸钾(464.51mg,3.36mmol)和碘化钠(167.93mg,1.12mmol),在氮气氛围下,80℃搅拌2小时。反应完毕,冷却至室温,加入乙酸乙酯(50mL)和水(50mL)萃取,有机相经饱和食盐水洗涤(30mL×2),无水硫酸钠干燥,过滤,减压浓缩,所得粗品经柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/0-10/1,体积比)分离纯化,得到中间体BB-16-1。MS-ESI m/z:636.0[M+H]
+。
步骤2:中间体BB-16的合成
将中间体BB-16-1(0.7g,963.22μmol)加到乙醇(10mL)和水(2mL)中,开启搅拌,随后加入一水合氢氧化锂(80.84mg,1.93mmol),反应液在氮气氛围下,25℃搅拌12小时。反应完毕,减压浓缩除去溶剂,加入水(10mL)稀释,加入盐酸溶液(2N)调至pH至3,水相冻干,得到中间体BB-16的粗品。MS-ESI m/z:608.1[M+H]
+。
参考例17:片段BB-17
合成路线:
步骤1:中间体BB-17-2的合成
将化合物BB-17-1(25g,105.44mmol,62.50mL)溶于二氯甲烷(1000mL)中,加入乙酰氯(8.28g,105.44mmol,7.52mL),氮气置换三次,在5-15℃分批加入三氯化铝(29.53g,221.43mmol,12.10mL),室温搅拌4小时。将反应液缓慢倒入冰水(500mL)中,用二氯甲烷(200mL×3)萃取,合并有机相,用饱和食盐水(300mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到中间体BB-17-2。MS-ESI m/z:265.0[M+H]
+,267.0[M+2+H]
+。
步骤2:中间体BB-17-3的合成
将中间体BB-17-2(20g,75.44mmol)和碳酸二甲酯(27.18g,301.77mmol,25.40mL)溶于四氢呋喃(200mL)中,氮气置换三次,降温至0℃,缓慢加入叔丁醇钾(50.79g,452.66mmol),升温至70℃搅拌过夜。将反应液减压浓缩得到黄色残余物,向残余物中加入冰水(700mL),用盐酸溶液(6M)调节pH至2-3,有大量固体析出,过滤,滤饼依次用水(200mL)和甲基叔丁基醚(200mL)淋洗,滤饼减压浓缩,得到中间体BB-17-3。MS-ESI m/z:291.0[M+H]
+,293.0[M+2+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:9.91(d,J=9.2Hz,1H),8.21(s,1H),8.01(d,J=8.8Hz,1H),7.70(d,J=9.2Hz,1H),7.45(d,J=8.8Hz,1H),5.15(s,1H)。
步骤3:中间体BB-17-4的合成
将中间体BB-17-3(21g,72.14mmol)溶于乙醇(300mL)中,加入盐酸羟胺(32.59g,468.92mmol)和醋酸钠(20.71g,252.49mmol),氮气置换三次,升温至90℃搅拌96小时。将反应液冷却至室温,过滤,用乙酸乙酯(500mL)淋洗滤饼,滤液减压浓缩剩余1/4,用氢氧化钠溶液(1M)调节pH至8-9,过滤,滤饼用乙酸乙酯(200mL)淋洗,滤液分液后收集水相。收集水相和固体,用盐酸溶液(2M)调节pH至3-4,有大量不溶物,加入乙酸乙酯(500mL),分液后收集有机相。水相用乙酸乙酯(300mL×3)萃取,合并有机相,依次用柠檬酸溶液(10%,300mL)和饱和食盐水(300mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到中间体BB-17-4。
步骤4:中间体BB-17-5的合成
将中间体BB-17-4(10g,23.87mmol)溶于乙醇(100mL)中,加入浓硫酸(3.11g,31.03mmol,1.69mL,纯度:98%,1.3eq),氮气置换三次,升温至70℃搅拌过夜。将反应液减压浓缩得到深黄色残余物,向残余物中加入水(200mL)和乙酸乙酯(300mL),分液后收集有机相。水相用乙酸乙酯(200mL×4)萃取,合并有机相,用饱和食盐水(300mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到残余物。用2-甲基四氢呋喃(50mL)将残余物溶解,加入石油醚(100mL),有大量固体析出,过滤,滤饼用石油醚(10mL)淋洗,滤饼减压浓缩得到固体,滤液减压浓缩得到残余物。残余物经柱层析分离(洗脱剂:乙酸乙酯/石油醚=0/1-15/85,体积比)纯化,得到中间体BB-17-5。MS-ESI m/z:334.0[M+H]
+,336.0[M+2+H]
+。
1H NMR(400MHz,CDCl
3)δ:8.16(d,J=1.6Hz,1H),7.99(d,J=8.8Hz,1H),7.88(d,J=8.8Hz,1H),7.79-7.71(m,2H),4.32(s,2H),4.23(q,J=7.2Hz,2H),1.22(t,J=7.0Hz,3H)。
步骤5:化合物BB-17-6的合成
将中间体BB-17-5(7.9g,23.64mmol,1eq),氨基甲酸叔丁酯(3.32g,28.37mmol,1.2eq)溶于甲苯(100mL)和水(20mL),加入磷酸钾(20.07g,94.56mmol,4eq),2-二-叔丁膦基-2′,4′,6′-三异丙基联苯(1.41g,3.31mmol,0.14eq),三(二亚苄基丙酮)二钯(1.52g,1.65mmol,0.07eq),氮气置换三次,升温至100℃搅拌过夜。将反应液减压浓缩得到深黄色残余物,向残余物中加入水(100mL)和乙酸乙酯(200mL),分液后收集有机相。水相用乙酸乙酯(100mL×3)萃取,合并有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到残余物。残余物经柱层析分离(洗脱剂:乙酸乙酯/石油醚=0/1-15/85)纯化,得到中间体BB-17-6。MS-ESI m/z:371.2[M+H]
+。
1H NMR(400MHz,CDCl
3)δ:8.20(s,1H),8.03(d,J=8.8Hz,1H),7.90(d,J=9.2Hz,1H),7.68(d,J=9.2Hz,1H),7.53(dd,J=2.4Hz,8.8Hz,1H),6.71(s,1H),4.31(s,2H),4.22(q,J=7.2Hz,2H),1.57(s,9H),1.21(t,J=7.0Hz,3H)。
步骤6:中间体BB-17-7的合成
将中间体BB-17-6(0.25g,674.95μmol)溶于四氢呋喃(3mL)中,降温至0℃,加入丙烯酰胺(47.97mg,674.95μmol,46.58μL)和叔丁醇钾(113.61mg,1.01mmol)。反应体系在20℃搅拌1小时。将反应液缓慢倒入饱和氯化铵溶液(10mL)中,加入乙酸乙酯(10mL),分离有机相。水相用乙酸乙酯(10mL×3)萃取,合并有机相,用饱和食盐水(10mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经过柱层析(洗脱剂:石油醚/乙酸乙酯=0/1-1/1,体积比)分离纯化,得到中间体BB-17-7。MS-ESI m/z:396.2[M+H]
+。
1H NMR(400MHz,DMSO_d
6)δ:11.12(s,1H),9.66(s,1H),8.36(s,1H),8.12(d,J=9.2Hz,1H),8.07(d,J=9.2Hz,1H),7.85(d,J=8.8Hz,1H),7.68(dd,J=2.0,9.2Hz,1H),5.00(dd,J=4.8Hz,11.2Hz,1H),2.89-2.76(m,1H),2.68-2.53(m,2H),2.41-2.31(m,1H),1.52(s,9H)。
步骤7:中间体BB-17的盐酸盐的合成
将中间体BB-17-7(120mg,303.48μmol)溶于盐酸/乙酸乙酯溶液(5mL,4M)中,25℃下反应30分钟。将反应液过滤,用乙酸乙酯(3mL×3)淋洗滤饼,收集滤饼,得到中间体BB-17的盐酸盐。
参考例18:片段BB-18
合成路线:
步骤1:化合物BB-18-2的合成
将化合物BB-18-1(10g,53.13mmol)溶于二氯甲烷(300mL)中,依次加入乙酰氯(4.17g,53.13mmol,3.79mL)和三氯化铝(10.63g,79.68mmol,4.35mL),氮气保护下室温搅拌2小时。反应完毕后,将反应液缓慢倒入冰水(400mL)中,分液后收集有机相。水相用二氯甲烷(3×100mL)萃取,合并有机相经饱和食盐水(300mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到化合物BB-18-2。MS-ESI m/z:231.1[M+H]
+.
1H NMR(400MHz,CDCl
3)δ:7.78(d,J=9.2Hz,1H),7.70(d,J=9.2Hz,1H),7.26(d,J=9.2Hz,1H),7.17(dd,J=2.4Hz,9.2Hz,1H),7.10(d,J=2.4Hz,1H),3.96(s,3H),3.91(s,3H),2.65(s,3H).
步骤2:化合物BB-18-3的合成
将化合物BB-18-2(24.3g,105.53mmol)溶于二氯甲烷(250mL)中,置换氮气三次,降温至-60℃,并于-60℃~-40℃下滴加三氯化硼的二氯甲烷溶液(1M,105.53mL)。滴加完毕后,氮气保护下升温至0~5℃并搅拌2小时。反应完毕后,将反应液缓慢倒入冰水(300mL)中,分液后收集有机相。水相用二氯甲烷(3×100mL)萃取,合并有机相经饱和食盐水(300mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到BB-18-3。MS-ESI m/z:217.1[M+H]
+.
1H NMR(400MHz,CDCl
3)δ:13.17(s,1H),8.02(d,J=9.6Hz,1H),7.81(d,J=8.8Hz,1H),7.25(dd,J=2.8Hz,9.2Hz,1H),7.13(d,J=9.2Hz,2H),3.93(s,3H),2.85(s,3H).
步骤3:化合物BB-18-4的合成
将化合物BB-18-3(10g,46.25mmol)溶于四氢呋喃(150mL)中,加入碳酸二甲酯(16.66g,184.99 mmol,15.57mL),降温至0℃,加入叔丁醇钾(31.14g,277.48mmol)。氮气保护下反应升温至70℃搅拌12小时。反应完毕后,将反应液缓慢加入到冰水(200mL)中,用6M盐酸调节pH=2~3,有白色固体析出,过滤,滤饼用水(100mL)洗涤后减压干燥得到化合物BB-18-4。MS-ESI m/z:243.1[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:12.94(s,1H),9.18(d,J=9.6Hz,1H),8.10(d,J=9.2Hz,1H),7.49(dd,J=3.0Hz,5.8Hz,2H),7.33(dd,J=2.8Hz,9.6Hz,1H),5.80(s,1H),3.89(s,3H).
步骤4:化合物BB-18-5的合成
将化合物BB-18-4(11g,45.41mmol)溶于二氯甲烷(150mL)中,氮气置换三次,降温至-50℃,并于-50℃~-30℃下滴加三溴化硼(39.82g,158.94mmol,15.31mL),反应混合物缓慢恢复至15℃并搅拌12小时。反应完毕后,将反应液倒入冰水(300mL)中,过滤,滤饼用水(100mL)洗涤,收集滤饼。滤液用2-甲基四氢呋喃(4×200mL)萃取,合并有机相,用无水硫酸钠干燥,过滤,收集滤液。滤饼与滤液合并,减压浓缩得到化合物BB-18-5。MS-ESI m/z:229.2[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:12.76(s,1H),9.11(d,J=8.8Hz,1H),7.98(d,J=9.2Hz,1H),7.41(d,J=8.8Hz,1H),7.26-7.22(m,2H),5.72(s,1H).
步骤5:化合物BB-18-6的合成
将化合物BB-18-5(11.5g,50.39mmol)溶于乙醇(170mL)中,依次加入盐酸羟胺(12.26g,176.38mmol,)和乙醇钠(12.00g,176.38mmol),氮气保护下升温至80℃搅拌12小时。反应完毕后,将反应液减压浓缩至一半体积,向残余物中加入水(200mL),用乙酸乙酯(3×200mL)萃取,合并有机相,用饱和食盐水(2×500mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩得到化合物BB-18-6。MS-ESI m/z:244.2[M+H]
+.
步骤6:化合物BB-18-7的合成
将化合物BB-18-6(11g,39.20mmol,纯度:86.67%)溶于乙醇(110mL)中,加入硫酸(4.05g,40.45mmol,2.20mL,纯度:98%),氮气置换三次,升温至70℃搅拌12小时。反应完毕后,将反应液减压浓缩得到黄色残余物,向残余物中加入水(100mL)和乙酸乙酯(200mL),分液后收集有机相。水相用乙酸乙酯(3×100mL)萃取,合并有机相用饱和食盐水(200mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩得到黄色残余物。残余物经柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/0-4/1),得到化合物BB-18-7。MS-ESI m/z:272.1[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:9.89(s,1H),7.99(d,J=5.6Hz,1H),7.97(d,J=5.6Hz,1H),7.80(d,J=9.2Hz,1H),7.40(d,J=2.4Hz,1H),7.27(dd,J=2.4,9.2Hz,1H),4.44(s,2H),4.15(q,J=7.2Hz,2H),1.16(t,J=7.0Hz,3H).
步骤7:化合物BB-18-8的合成
将化合物BB-18-7(2g,7.37mmol)溶于N,N-二甲基甲酰胺(20mL)中,加入碳酸钾(2.04g,14.75 mmol)和烯丙基溴(891.93mg,7.37mmol),氮气置换三次,升温至50℃搅拌12小时。反应完毕后,向反应液中加入水(20mL)和乙酸乙酯(20mL)萃取,分液后收集有机相。水相用乙酸乙酯(2×20mL)萃取,合并有机相并用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到黄色残余物。残余物经柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/0-17/3),得到化合物BB-18-8。MS-ESI m/z:312.1[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:8.08(d,J=9.2Hz,1H),8.04(d,J=8.8Hz,1H),7.87(d,J=9.2Hz,1H),7.65(d,J=2.4Hz,1H),7.43(dd,J=2.6Hz,9.0Hz,1H),6.17-6.08(m,1H),5.47(dd,J=1.2Hz,17.2Hz,1H),5.31(d,J=10.4Hz,1H),4.72(d,J=5.2Hz,2H),4.47(s,2H),4.15(q,J=7.0Hz,2H),1.16(t,J=7.0Hz,3H).
步骤8:化合物BB-18-9的合成
将化合物BB-18-8(1g,3.21mmol)溶于四氢呋喃(20mL),0℃下加入叔丁醇钾(540.63mg,4.82mmol)和丙烯酰胺(228.30mg,3.21mmol),室温搅拌1小时。反应完毕后,将反应液缓慢加入到0.5M盐酸调节pH=6~7,用乙酸乙酯(3×20mL)萃取,合并有机相用饱和食盐水(30mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩得到黄色残余物。残余物经柱层析分离(洗脱剂:石油醚/乙酸乙酯=9/1-13/7),得到化合物BB-18-9。MS-ESI m/z:337.1[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.12(s,1H),8.15(d,J=9.2Hz,1H),8.09(d,J=9.2Hz,1H),7.89(d,J=9.2Hz,1H),7.66(d,J=2.4Hz,1H),7.39(dd,J=2.6Hz,9.0Hz,1H),6.18-6.08(m,1H),5.47(dd,J=1.6Hz,17.2Hz,1H),5.31(dd,J=1.6Hz,10.4Hz,1H),5.03(dd,J=4.8Hz,11.2Hz,1H),4.73(d,J=5.2Hz,2H),2.92-2.77(m,1H),2.68-2.54(m,2H),2.42-2.30(m,1H).
步骤9:化合物BB-18的合成
将化合物BB-18-9(120mg,356.78μmol)溶于四氢呋喃(3mL)和水(1mL)的混合溶剂中,0℃下加入二水合锇酸钾(13.15mg,35.68μmol)和高碘酸钠(305.25mg,1.43mmol,79.08μL),反应体系置换氮气三次,室温反应3小时。反应完毕后,向反应液中加入水(5mL)中,用乙酸乙酯(10mL×3)萃取,收集有机相,用饱和食盐水(10mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到化合物BB-18。MS-ESI m/z:339.1[M+H]
+.
参考例19:片段BB-19
合成路线:
步骤1:化合物BB-19-1的合成
将5-己烯酸(82.57mg,723.43μmol,85.92μL)溶于N,N-二甲基甲酰胺(3mL)中,随后加入2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(343.84mg,904.29μmol)和N,N-二异丙基乙胺(311.66mg,2.41mmol,420.03μL),反应体系置换氮气三次,25℃搅拌20分钟后加化合物BB-17的盐酸盐(200mg,602.86μmol),反应液在25℃搅拌12小时。反应完毕,反应液加水(10mL)和乙酸乙酯(10mL),分离有机相,水相加乙酸乙酯(20mL×3)萃取,合并的有机相经饱和食盐水(20mL×2)洗涤,用无水硫酸钠干燥,过滤,减压浓缩得到粗产物经板层析分离(展开剂:石油醚∶乙酸乙酯=0∶1),得到化合物BB-19-1。MS-ESI m/z:392.2[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.13(s,1H),10.20(s,1H),8.52(d,J=2.0Hz,1H),8.18(J=9.2Hz,1H),8.10(d,J=8.8Hz,1H),7.88(d,J=9.2Hz,1H),7.79(dd,J=2.2Hz,9.0Hz,1H),5.90-5.79(m,1H),5.09-4.98(m,3H),2.88-2.78(m,1H),2.68-2.61(m,1H),2.60-2.55(m,1H),2.54-2.51(m,1H),2.40(t,J=7.6Hz,2H),2.10(q,J=7.0Hz,2H),1.78-1.68(m,2H).
步骤2:化合物BB-19的合成
将化合物BB-19-1(120mg,306.58μmol)溶于四氢呋喃(3mL)和水(1mL)的混合溶剂中,0℃下将加入高碘酸钠(262.30mg,1.23mmol,67.95μL)和二水合四氧化锇(11.30mg,30.66μmol),反应体系置换氮气三次,25℃搅拌3小时,反应完毕,向反应液加水(5mL),然后加乙酸乙酯(10mL)稀释,分离有机相,水相加乙酸乙酯(20mL×3)萃取,合并有机相的经饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到化合物BB-19。MS-ESI m/z:394.2[M+H]
+.
参考例20:片段BB-20
合成路线:
步骤1:化合物BB-20-2的合成
0℃和氮气保护下,将化合物BB-20-1(5g,18.03mmol)溶于二氯甲烷(50mL)和丙酮(25mL)中,依次慢慢滴加氰基三甲基硅烷(2.68g,27.04mmol,3.38mL),三甲基硅基三氟甲磺酸酯(400.67mg,1.80mmol,325.74μL),反应混合物在20℃搅拌2小时。反应完毕后,反应液减压除去溶剂。所得残余物经过柱层析(洗脱剂:石油醚/乙酸乙酯=1/0-4/1,体积比)分离,得到化合物BB-20-2。
步骤2:化合物BB-20的合成
室温和氮气保护下,将化合物BB-20-2(10g,29.03mmol)溶于N,N-二甲基乙酰胺(100mL),4-氰基-3-三氟甲基异硫氰酸苯酯(6.62g,29.03mmol)分批加入反应液中,反应混合物在20℃搅拌3小时,加入甲醇(100mL),稀盐酸(2M,56.32mL),反应混合物在70℃搅拌2小时,反应完毕后,冷却至室温,加入水(100mL)稀释,用乙酸乙酯(100mL×3)萃取。合并有机相,用饱和食盐水(100mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压除去溶剂。所得残余物经过柱层析分离(洗脱剂:二氯甲烷/甲醇=20/1-10/1,体积比),得到化合物BB-20。
1H NMR(400MHz,CDCl
3)δ:7.99(d,J=8.8Hz,1H),7.96(d,J=1.6Hz,1H),7.84(dd,J=2.0,8.4Hz,1H),7.22(d,J=9.2Hz,2H),7.04(d,J=8.8Hz,2H),3.64-3.57(m,4H),3.45-3.38(m,4H),1.58(s,6H).
参考例21:片段BB-21
合成路线:
步骤1:化合物BB-21-2的合成
将亚磷酸三苯酯(63.92g,206.00mmol,54.17mL)溶于二氯甲烷(400mL)中,氮气保护下将反应体系降温至-70℃,依次加入液溴(35.91g,224.73mmol,11.59mL),三乙胺(24.64g,243.46mmol,33.89mL)和化合物BB-21-1(33g,187.28mmol)的二氯甲烷(100mL)溶液,滴加完毕后,将反应体系缓慢升温至室温并搅拌反应15小时。反应完毕后,将反应液缓慢倒入饱和亚硫酸钠水溶液(300mL)中,搅拌10分钟,用二氯甲烷(400mL)萃取,收集有机相,用饱和食盐水洗涤(500mL),无水硫酸钠干燥,过滤,减压浓缩得到粗品。粗品通过柱层析(洗脱剂:石油醚∶乙酸乙酯=20/1-7/1)分离纯化得到化合物BB-21-2。
1H NMR(400MHz,DMSO_d
6)δ:7.33(d,J=8.4Hz,1H),6.83-6.76(m,2H),6.33(t,J=4.8Hz,1H),3.76(s,3H),2.77(t,J=8.2Hz,2H),2.33-2.25(m,2H).
步骤2:化合物BB-21-3的合成
将化合物BB-21-2(40g,167.29mmol)加入至甲苯(500mL)中,在0℃下缓慢地加入二氯二氰基苯醌(41.77g,184.02mmol),0℃下搅拌1小时后恢复至室温搅拌14小时。反应完毕后,向反应液中滴加饱和亚硫酸钠水溶液(200mL),搅拌10分钟,加入1mol/L的氢氧化钠水溶液(150mL),用乙酸乙酯(240mL×3)萃取,收集有机相,用饱和食盐水(320mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品。粗品通过柱层析(洗脱剂:石油醚∶乙酸乙酯=5/1-1/1)分离纯化得到化合物BB-21-3。
1H NMR(400MHz,DMSO_d
6)δ:8.02(d,J=9.2Hz,1H),7.87(d,J=8.4Hz,1H),7.68(d,J=7.2Hz,1H),7.42(s,1H),7.37(t,J=7.8Hz,1H),7.31(d,J=9.2Hz,1H),3.90(s,3H).
步骤3:化合物BB-21-4的合成
将乙酰氯(32.78g,417.56mmol,29.80mL)溶于二氯甲烷(1L)中,将反应体系降温至0℃,加入三氯化铝(106.29g,797.16mmol),氮气保护下0℃下反应30分钟,加入化合物BB-21-3(90g,379.60mmol),继续反应5.5小时后,恢复至室温反应10小时。反应完毕后,将反应液倒入水(1000mL)中,用二氯甲 烷(500mL×3)萃取,收集有机相,用饱和食盐水(800mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到化合物BB-21-4。MS-ESI m/z:279.0[M+H]
+.281.0[M+H+2]
+.
步骤4:化合物BB-21-5的合成
将化合物BB-21-4(42.5g,152.26mmol)加入至二氯甲烷(600mL)中,将反应体系降温至-60℃,缓慢地加入三氯化硼(26.76g,228.39mmol,29.70mL),氮气保护下,恢复至室温反应5小时。反应完毕后,将反应液倒入1mol/L盐酸(100mL)中,室温搅拌0.5小时,用二氯甲烷(500mL×3)萃取,收集有机相,用饱和食盐水(600mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到化合物BB-21-5。MS-ESI m/z:265.0[M+H]
+.267.0[M+H+2]
+.
步骤5:化合物BB-21-6的合成
将碳酸二甲酯(54.37g,603.54mmol,50.81mL)加入四氢呋喃(1L)中,慢慢地加入化合物BB-21-5(40g,150.89mmol)和叔丁醇钾(101.58g,905.31mmol),氮气保护下升温至70℃反应12小时。反应完毕后,将反应液降至室温,减压浓缩除去溶剂,加入冰水(250mL),用甲基叔丁基醚(250mL×2)萃取,收集水相。有机相用2M氢氧化钠水溶液(150mL×2)萃取,收集水相。合并水相,用6M盐酸溶液调节pH=2-3,析出固体,过滤,用水(150mL×2)淋洗滤饼,收集滤饼得到化合物BB-21-6。MS-ESI m/z:291.0[M+H]
+,292.9[M+H+2]
+.
1H NMR(400MHz,DMSO_d
6)δ:9.38(d,J=8.8Hz,1H),8.42(d,J=9.2Hz,1H),7.96(d,J=6.8Hz,1H),7.67(d,J=9.2Hz,1H),7.58(t,J=7.6Hz,1H),5.89(s,1H).
步骤6:化合物BB-21-7的合成
将化合物BB-21-6(80g,274.82mmol)加入至乙醇(1L)中,慢慢地加入盐酸羟胺(66.84g,961.88mmol)和乙酸钠(78.90g,961.88mmol),氮气保护下升温至80℃反应12小时。反应完毕后,将反应液降温至室温,过滤,滤饼用乙酸乙酯(600mL)淋洗,滤饼丢弃,滤液浓缩至剩余1/3,用碳酸氢钠水溶液调节pH=8,用乙酸乙酯(600mL)萃取,有机相用饱和碳酸氢钠水溶液(200mL×2)洗涤,合并两次水相,用4mol/L盐酸调节溶液pH=2,过滤,收集滤饼得到化合物BB-21-7。MS-ESI m/z:306.0[M+H]
+,308.0[M+H+2]
+.
1H NMR(400MHz,CD
3OD)δ:8.40(d,J=9.2Hz,1H),8.32(d,J=8.4Hz,1H),7.83(d,J=6.8Hz,1H),7.79(d,J=9.2Hz,1H),7.52(t,J=8.0Hz,1H),4.15(s,2H).
步骤7:化合物BB-21的合成
将化合物BB-21-7(30g,98.00mmol)加入至乙醇(300mL)中,慢慢地加入浓硫酸(9.81g,98.00mmol,5.33mL,纯度:98%),氮气保护下升温至70℃下反应2小时。反应完毕后,将反应液降温至室温,减压浓缩除去溶剂,加入乙酸乙酯(200mL),用饱和碳酸氢钠调节溶液pH=8,萃取,有机相用饱和碳酸氢钠水溶液(150mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品。粗品通过柱层析(洗脱剂:石油醚∶乙酸乙酯=8/1-3/1)分离纯化得到化合物BB-21。MS-ESI m/z:334.1[M+H]
+,336.1[M+H+2]
+.
1H NMR (400MHz,DMSO_d
6)δ:8.41(d,J=9.6Hz,1H),8.17(d,J=8.4Hz,1H),8.06(d,J=9.2Hz,1H),7.99(d,J=7.6Hz,1H),7.66(t,J=8.0Hz,1H),4.53(s,2H),4.14(q,J=7.2Hz,2H),1.15(t,J=7.2Hz,3H).
参考例22:片段BB-22
合成路线:
步骤1:化合物BB-22-1的合成
将氨基甲酸叔丁酯(2.31g,19.75mmol)加入至甲苯(60mL)和水(20mL)中,依次加入化合物BB-21(6g,17.96mmol),2-二叔丁基膦-2′,4′,6′-三异丙基联苯(762.45mg,1.80mmol),三(二亚苄基丙酮)二钯(822.10mg,897.76μmol)和磷酸钾(15.25g,71.82mmol),氮气保护下升温至100℃反应12小时。反应完毕后,将反应液倒入水(100mL)中,用乙酸乙酯(90mL×3)萃取,收集有机相,用饱和食盐水(70mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品。粗品通过柱层析(洗脱剂:石油醚∶乙酸乙酯=6/1-2/1)分离纯化得到化合物BB-22-1。
1H NMR(400MHz,DMSO_d
6)δ:9.47(s,1H),8.29(d,J=9.6Hz,1H),7.92(d,J=7.6Hz,1H),7.91(d,J=9.2Hz,1H),7.70(t,J=8.0Hz,1H),7.65(d,J=6.8Hz,1H),4.50(s,2H),4.15(q,J=7.0Hz,2H),1.50(s,9H),1.16(t,J=7.2Hz,3H).
步骤2:化合物BB-22-2的合成
室温下,将化合物BB-22-1(2.3g,6.21mmol)加入四氢呋喃(25mL)中,依次加入丙烯酰胺(529.63mg,7.45mmol)和叔丁醇钾(1.05g,9.31mmol),氮气保护下反应1小时。反应完毕后,将反应液加入至1 mol/L盐酸溶液中,用乙酸乙酯(45mL×3)萃取,收集有机相,用饱和食盐水(30mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品。粗品通过柱层析(洗脱剂:石油醚∶乙酸乙酯=5/1-1/1)分离纯化得到化合物BB-22-2。MS-ESI m/z:396.2[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.14(s,1H),9.47(s,1H),8.28(d,J=9.6Hz,1H),8.04(d,J=7.2Hz,1H),7.93(d,J=9.6Hz,1H),7.68(t,J=7.8Hz,1H),7.63(d,J=7.6Hz,1H),5.07(dd,J=4.6Hz,11.4Hz,1H),2.89-2.81(m,1H),2.65-2.55(m,2H),2.41-2.33(m,1H),1.50(s,9H).
步骤3:化合物BB-22盐酸盐的合成
室温下,将化合物BB-22-2(0.7g,1.77mmol)加入至盐酸乙酸乙酯(4M,10mL)中,搅拌2小时。反应完毕后,将反应液过滤,滤饼用乙酸乙酯(30mL)淋洗,收集滤饼得到化合物BB-22的盐酸盐。MS-ESI m/z:296.1[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.14(s,1H),8.34(d,J=9.6Hz,1H),7.98(d,J=9.2Hz,1H),7.90(s,1H),7.63(t,J=8.0Hz,1H),7.39(d,J=7.6Hz,1H),5.05(dd,J=4.4Hz,11.4Hz,1H),2.89-2.80(m,1H),2.68-2.61(m,1H),2.60-2.53(m,1H),2.41-2.35(m,1H).
参考例23:片段BB-23
合成路线:
步骤1:化合物BB-23-2的合成
将BB-23-1(10g,57.95mmol)溶于N,N-二甲基甲酰胺(100mL)中,加入哌嗪-1-甲酸叔丁酯(10.79g,57.95mmol)和N,N-二异丙基乙胺(22.47g,173.84mmol),反应混合物升温至100℃反应12小时。反应完毕后,冷却至室温,过滤,滤饼用甲基叔丁基醚(150mL)淋洗,收集滤饼,得到化合物BB-23-2。MS-ESI m/z:323.2[M+H]
+.
步骤2:化合物BB-23-3的合成
将化合物BB-23-2(2.6g,8.07mmol)溶于甲醇(5mL)、四氢呋喃(30mL)和水(7mL)的混合溶剂中,加入一水合氢氧化锂(676.92mg,16.13mmol),氮气保护下反应在20℃搅拌14小时。反应完毕,反应液减压浓缩,残余物用1mol/L盐酸调节pH=2-3,有固体析出。过滤,收集滤饼,真空干燥得到化合物BB-23-3。
1H NMR(400MHz,DMSO_d
6)δ:7.85(d,J=9.6Hz,1H),7.29(d,J=9.6Hz,1H),3.74(t,J=5.4Hz,4H),3.47(t,J=5.0Hz,4H),1.43(s,9H).
步骤3:化合物BB-23-5的合成
将化合物BB-23-4的盐酸盐(0.78g,2.72mmol)和BB-23-3(837.44mg,2.72mmol)溶于N,N-二甲基甲酰胺(10mL)中,之后依次加入2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(1.55g,4.07mmol)和N,N-二异丙基乙胺(1.05g,8.15mmol,1.42mL),氮气保护下反应在20℃搅拌12小时。反应完毕,向反应液中加入水(30mL),搅拌0.5小时后过滤,收集滤饼,真空干燥得到化合物BB-23-5。
1H NMR(400MHz,CDCl
3)δ:8.04(d,J=9.6Hz,1H),7.86(d,J=8.0Hz,1H),7.56(d,J=8.8Hz,1H),7.03-6.96(m,2H),6.86(dd,J=2.4Hz,8.8Hz,1H),4.37-4.26(m,1H),4.11-4.00(m,1H),3.82-3.71(m,4H),3.66-3.55(m,4H),2.26-2.13(m,4H),1.75-1.64(m,2H),1.50(s,9H),1.49-1.38(m,2H).
步骤4:化合物BB-23的盐酸盐的合成
将化合物BB-23-5(1.25g,2.31mmol)溶于二氯甲烷(3mL)中,加入盐酸乙酸乙酯(4M,8mL),在20℃下反应12小时。反应液过滤,滤饼用乙酸乙酯(10mL)洗涤,收集滤饼干燥得到化合物BB-23的盐酸盐。MS-ESI m/z:441.1[M+H]
+.
1H NMR(400MHz,CD
3OD)δ:8.36(d,J=9.6Hz,1H),8.00(d,J=10.0Hz,1H),7.69(d,J=8.8Hz,1H),7.20(d,J=2.4Hz,1H),7.05(dd,J=2.4,8.8Hz,1H),4.57-4.45(m,1H),4.16(t,J=5.4Hz,4H),4.04-3.94(m,1H),3.50(t,J=5.4Hz,4H),2.26-2.18(m,2H),2.15-2.05(m,2H),1.74-1.61(m,4H).
参考例24:片段BB-24
合成路线:
步骤1:化合物BB-24-1的合成
将5-己烯酸(75.69mg,663.14μmol,78.76μL)溶于N,N-二甲基甲酰胺(5mL)中,随后加入2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(343.84mg,904.29μmol)和N,N-二异丙基乙胺(233.75mg,1.81mmol,315.02μL),反应体系置换氮气三次,20℃搅拌30分钟后加化合物BB-22的盐酸盐(200mg,602.86μmol),反应液在20℃搅拌2小时。反应完毕,反应液加水(15mL),用乙酸乙酯(20mL×3)萃取,合并的有机相经饱和食盐水(15mL×2)洗涤,用无水硫酸钠干燥,过滤,减压浓缩得到粗产物经柱层析分离(洗脱剂:石油醚/乙酸乙酯=2/1-1/2,体积比),得到化合物BB-24-1。MS-ESI m/z:392.2[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.15(s,1H),10.11(s,1H),8.28(d,J=9.6Hz,1H),8.08(s,1H),7.97(d,J=9.6Hz,1H),7.75-7.65(m,2H),5.95-5.82(m,1H),5.16-4.95(m,3H),2.88-2.78(m,1H),2.69-2.61(m,2H),2.59-2.55(m,1H),2.43-2.32(m,2H),2.20-2.10(m,2H),1.82-1.72(m,2H).
步骤2:化合物BB-24的合成
将化合物BB-24-1(100mg,255.48μmol)溶于四氢呋喃(12mL)和水(4mL)的混合溶剂中,0℃下将加入高碘酸钠(218.58mg,1.02mmol,56.63μL)和二水合四氧化锇(9.41mg,25.55μmol),反应体系置换氮气三次,20℃搅拌1小时,反应完成,反应液倒入水中(20mL),水相加乙酸乙酯(15mL×3)萃取,合并有机相的经饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到化合物BB-24。MS-ESI m/z:394.2[M+H]
+.
参考例25
合成路线:
步骤1:化合物BB-25-1的合成
室温和氮气保护下,将化合物BB-21(300mg,897.76μmol),1-叔丁基氧羰基哌嗪(299.91mg,1.35mmol),三(二亚苄基丙酮)二钯(41.10mg,44.89μmol),碳酸铯(41.10mg,44.89μmol)和2-双环己基膦-2,6-二异丙氧基-1,1-联苯(41.89mg,89.78μmol)溶于1,4-二氧六环(5mL)中,反应混合物升温至100℃,搅拌反应2小时。反应完毕后,冷却至室温,加入水(20mL),用乙酸乙酯(30mL×3)萃取。合并有机相,用饱和食盐水(30mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去残余溶剂。所得残余物经过柱层析(洗脱剂:石油醚/乙酸乙酯=5/1-1/1,体积比)分离,得到化合物BB-25-1。MS-ESI m/z:440.3[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:8.47(d,J=9.2Hz,1H),7.89(d,J=9.6Hz,1H),7.84(d,J=8.4Hz,1H),7.67(t,J=8.0Hz,1H),7.30(d,J=7.6Hz,1H),4.48(s,2H),4.15(q,J=7.2Hz,2H),3.95-3.40(m,4H),3.15-2.80(m,4H),1.45(s,9H),1.17(t,J=7.0Hz,3H).
步骤2:化合物BB-25-2的合成
室温和氮气保护下,将化合物BB-25-1(100mg,227.53μmol)和丙烯酰胺(19.41mg,273.04umol,18.84μL)溶于四氢呋喃(2mL)中,冷却至0℃,加入叔丁醇钾(38.30mg,341.29μmol),反应混合物室温下搅拌反应1小时。反应结束后,将反应液倒入稀盐酸(0.1N,10mL)中,用乙酸乙酯(20mL×2)萃取。合并有机相,用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂,所得残余物经过板分离(展开剂:石油醚/乙酸乙酯=1/1,体积比)纯化,得到化合物BB-25-2。MS-ESI m/z:465.3[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.12(s,1H),8.49(d,J=9.6Hz,1H),7.95(d,J=7.2Hz,1H),7.91(d,J=9.2Hz,1H),7.66(t,J=7.8Hz,1H),7.31(d,J=8.0Hz,1H),5.04(dd,J=4.6,11.4Hz,1H),4.04-3.50(m,4H),3.15-2.90(m,4H),2.90-2.79(m,1H),2.70-2.59(m,1H),2.40-2.31(m,2H),1.45(s,9H).
步骤3:化合物BB-25的盐酸盐合成
室温下,将化合物BB-25-2(40mg,86.11μmol)溶于乙酸乙酯(2mL)中,加入盐酸的乙酸乙酯溶液(3mL,4M),反应混合物室温下搅拌反应1小时。反应完成后,减压浓缩除去溶剂,得到化合物BB-25的盐酸盐。MS-ESI m/z:365.0[M+H]
+.
参考例26:片段BB-26
合成路线:
步骤1:化合物BB-26-2的合成
室温下,将化合物BB-26-1(25g,144.87mmol)和4-哌啶甲醇(16.69g,144.87mmol)溶于1,4-二氧六环(500mL)中,依次加入碳酸钾(60.07g,434.61mmol)和四丁基碘化铵(5.35g,14.49mmol),反应混合物升温至100℃,搅拌反应48小时。反应完毕后,冷却至室温,过滤,滤饼用二氯甲烷(700mL)淋洗,母液中加入水(300mL),用二氯甲烷/甲醇混合溶液(10/1,体积比,300mL×5)萃取,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品。向粗品中加入石油醚/二氯甲烷混合溶液(1/2,体积比,250mL),室温下搅拌1小时,过滤,滤饼用石油醚/二氯甲烷混合溶液(1/2,体积比,70mL×2)淋洗,收集滤饼,真空干燥,得到化合物BB-26-2。
1H NMR(400MHz,CDCl
3)δ:7.85(d,J=9.6Hz,1H),6.87(d,J=9.6Hz,1H),4.61(d,J=13.2Hz,2H),3.99(s,3H),3.55(d,J=5.6Hz,2H),3.10-3.00(m,2H),1.97-1.84(m,3H),1.39-1.23(m,2H).
步骤2:化合物BB-26-3的合成
室温下,将化合物BB-26-2(71.5g,284.54mmol)和咪唑(23.25g,341.45mmol)溶于二氯甲烷(700mL)中,冷却至0-10℃,滴加叔丁基二苯基氯硅烷(78.21g,284.54mmol,73.09mL),反应混合物室温下搅拌反应1小时。反应完毕后,加入饱和氯化铵水溶液(500mL),用二氯甲烷(300mL×3)萃取。合并有机相,用饱和氯化铵水溶液(500mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品。向粗品 中加入石油醚(300mL),室温下搅拌0.5小时,过滤,滤饼用石油醚(50mL×3)淋洗,收集滤饼,真空干燥,得到化合物BB-26-3。
步骤3:化合物BB-26-4的合成
室温下,将化合物BB-26-3(42g,85.77mmol)溶于四氢呋喃(1.25L)中,加入氢氧化钠(2M,85.77mL),反应混合物室温下搅拌反应1小时。反应完毕后,三批合并处理,用2N盐酸调节pH=6~7,减压浓缩除去溶剂,得到化合物BB-26-4。
步骤4:化合物BB-26-6的合成
室温和氮气保护下,将钠氢(27.87g,696.74mmol,纯度:60%)溶于N,N-二甲基甲酰胺(500mL)中,冷却至0℃,滴加化合物BB-26-5(100g,464.49mmol)的N,N-二甲基甲酰胺(750mL)溶液,反应混合物在0℃搅拌反应0.5小时。滴加2-氯-4-羟基苯甲腈(86.71g,557.39mmol)的N,N-二甲基甲酰胺(300mL)溶液,反应混合物在0℃搅拌反应0.5小时。然后慢慢升温至室温搅拌反应2小时。反应完毕后,将反应液倒入0-5℃的稀盐酸(1N,800mL)中,用乙酸乙酯(700mL×3)萃取。合并有机相,用饱和食盐水(700mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品。向粗品中加入正庚烷(500mL),室温下搅拌30分钟,过滤,滤饼用正庚烷(100mL×3)淋洗,收集滤饼,真空干燥,得到化合物BB-26-6。
1H NMR(400MHz,CDCl
3)δ:7.55(d,J=8.8Hz,1H),6.97(d,J=2.0Hz,1H),6.83(dd,J=2.4,8.8Hz,1H),4.54-4.33(m,1H),4.30-4.20(m,1H),3.53(br s,1H),2.19-2.02(m,4H),1.69-1.54(m,2H),1.46(s,9H),1.36-1.21(m,2H).
步骤5:化合物BB-26-7的盐酸盐合成
室温下,将化合物BB-26-6(92g,262.23mmol)溶于乙酸乙酯(60mL)中,加入盐酸的乙酸乙酯溶液(4M,750mL),反应混合物室温下搅拌反应过夜。反应完毕后,两批合并,过滤,滤饼用乙酸乙酯(100mL×2)淋洗,收集滤饼,真空干燥,得到化合物BB-26-7的盐酸盐。
1H NMR(400MHz,D
2O)δ:7.64(d,J=8.8Hz,1H),7.14(d,J=2.4Hz,1H),6.96(dd,J=2.4,8.8Hz,1H),4.50-4.39(m,1H),3.34-3.17(m,1H),2.28-2.07(m,4H),1.68-1.42(m,4H).
步骤6:化合物BB-26-8的合成
室温下,将化合物BB-26-4(60.5g,127.19mmol)和N,N-二异丙基乙胺(82.19g,635.97mmol)溶于N,N-二甲基甲酰胺(700mL)中,加入2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(58.04g,152.63mmol),反应混合物室温下搅拌反应5分钟,加入化合物BB-26-7的盐酸盐(32.88g,114.47mmol),反应混合物室温下搅拌反应过夜。反应完毕后,向反应液中加入水(3000mL),用乙酸乙酯(1000mL×3)萃取。合并有机相,用饱和食盐水(1000mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品。 向粗品中加入甲基叔丁基醚(300mL),室温下搅拌0.5小时,过滤,滤饼用甲基叔丁基醚(100mL×2)淋洗,收集滤饼,真空干燥,得到化合物BB-26-8。
1H NMR(400MHz,DMSO_d
6)δ:8.58(d,J=8.4Hz,1H),7.85(d,J=8.8Hz,1H),7.80(d,J=9.2Hz,1H),7.63-7.56(m,4H),7.50-7.40(m,6H),7.38(d,J=2.0Hz,1H),7.34(d,J=9.6Hz,1H),7.14(dd,J=2.4,8.8Hz,1H),4.63-4.44(m,3H),3.94-3.77(m,1H),3.54(d,J=6.0Hz,2H),3.07-2.96(m,2H),2.17-2.06(m,2H),1.96-1.86(m,3H),1.84-1.74(m,2H),1.71-1.59(m,2H),1.58-1.45(m,2H),1.32-1.21(m,2H),0.99(s,9H).
步骤7:化合物BB-26-9的合成
室温下,将化合物BB-26-8(53g,74.82mmol)溶于N,N-二甲基甲酰胺(530mL)中,加入氟化钾(30.43g,523.74mmol),反应混合物升温至100℃,搅拌反应过夜。反应完毕后,冷却至室温,合并两个批次,向反应液中加入水(3L),用乙酸乙酯(1L×3)萃取。合并有机相,用饱和食盐水(1L×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品。粗品经柱层析(洗脱剂:乙酸乙酯/石油醚=1/2-1/0,体积比)纯化,得到化合物BB-26-9。MS-ESI m/z:470.2[M+H]
+.
1H NMR(400MHz,CDCl
3)δ:7.99(d,J=9.6Hz,1H),7.87(br d,J=8.0Hz,1H),7.57(d,J=8.8Hz,1H),7.04-6.97(m,2H),6.86(dd,J=2.4,8.4Hz,1H),4.56(br d,J=12.8Hz,2H),4.38-4.28(m,1H),4.13-3.99(m,1H),3.57(br d,J=5.6Hz,2H),3.16-3.02(m,2H),2.26-2.12(m,4H),1.97-1.84(m,3H),1.76-1.62(m,4H),1.54-1.44(m,2H).
步骤8:化合物BB-26的合成
20℃下,将BB-26-9(2g,4.26mmol)加入至二氯甲烷(30mL),慢慢加入戴斯-马丁氧化剂(2.17g,5.11mmol,1.58mL),氮气氛围下反应2小时,反应完毕,将反应液过滤,向滤液中加入饱和碳酸氢钠溶液(20mL)搅拌15分钟,用二氯甲烷(25mL×2)萃取,收集有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗品经柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/1-1/5,体积比),得到BB-26。MS-ESI m/z:468.2[M+H]
+.
参考例27:片段BB-27
合成路线:
步骤1:化合物BB-27-2的合成
室温下,将化合物BB-27-1(0.5g,2.90mmol)和内-3-氮杂二环[3.1.0]己烷-6-甲醇(328.16mg,2.90mmol)溶于1,4-二氧六环(10mL)中,依次加入碳酸钾(1.20g,8.70mmol)和四丁基碘化铵(107.02mg,290.00μmol),反应混合物升温至100℃,搅拌反应过夜。反应完毕后,冷却至室温,过滤,滤饼用二氯甲烷(70mL)淋洗,母液加入水(30mL),用二氯甲烷/甲醇混合溶液(10/1,体积比,30mL×5)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂。所得粗品中加入石油醚/二氯甲烷混合溶液(2/1,体积比,10mL),室温下搅拌1小时,过滤,滤饼用石油醚/二氯甲烷混合溶液(2/1,体积比,5mL×2)淋洗,收集滤饼,真空干燥,得到化合物BB-27-2。
1H NMR(400MHz,CDCl
3)δ:7.87(d,J=9.6Hz,1H),6.58(d,J=9.6Hz,1H),4.20-3.75(m,5H),3.73-3.52(m,4H),1.74(br s,2H),1.12-0.97(m,1H).
步骤2:化合物BB-27-3的合成
室温和氮气保护下,将化合物BB-27-2(0.541g,2.17mmol)和咪唑(192.08mg,2.82mmol)溶于二氯甲烷(10mL)中,冷却至0℃,滴加叔丁基二苯基氯硅烷(596.55mg,2.17mmol),反应混合物缓慢升温至室温,搅拌反应1小时。反应完毕后,加入饱和氯化铵水溶液(10mL),用二氯甲烷(5mL×3)萃取。合并有机相,用饱和食盐水(10mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂。粗品中加入正庚烷(5mL),室温下搅拌10分钟,过滤,滤饼用正庚烷(2mL×3)淋洗,收集滤饼,真空干燥,得到化合物BB-27-3。
1H NMR(400MHz,CDCl
3)δ:7.86(d,J=9.6Hz,1H),7.71-7.65(m,4H),7.47-7.36(m,6H),6.55(d,J=9.6Hz,1H),4.20-3.72(m,5H),3.67(d,J=6.0Hz,2H),3.63-3.51(m,2H),1.62(br s,2H),1.06(s,9H),0.97-0.90(m,1H).
步骤3:化合物BB-27-4的合成
室温下,将化合物BB-27-3(901mg,1.85mmol)溶于四氢呋喃(10mL)中,加入氢氧化钠水溶液(2M,1.85mL),反应混合物室温下搅拌反应2小时。反应完毕后,向反应液中加入盐酸(1N)调节pH=6-7,减压浓缩除去溶剂,得到化合物BB-27-4。
步骤4:化合物BB-27-5的合成
室温下,将化合物BB-27-4(1.02g,2.15mmol)和N,N-二异丙基乙胺(1.39g,10.77mmol)溶于N,N-二甲基甲酰胺(10mL)中,加入2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(781.60mg,2.06mmol),反应混合物室温下搅拌反应0.5小时。再加入化合物BB-26-7的盐酸盐(491.92mg,1.71mmol),反应混合物继续搅拌反应过夜。反应完毕后,加入水(50mL),用乙酸乙酯(20mL×3)萃取。合并有机相,用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂。所得粗品经柱层析(洗脱剂:乙酸乙酯/石油醚=1/3-1/2,体积比)纯化,得到化合物BB-27-5。
步骤5:化合物BB-27-6的合成
室温下,将化合物BB-27-5(1g,1.42mmol)溶于N,N-二甲基甲酰胺(20mL)中,加入氟化钾(575.75mg,9.91mmol),反应混合物升温至110℃,搅拌反应过夜。反应完毕后,冷却至室温,加入水(50mL),用乙酸乙酯(20mL×3)萃取。合并有机相,用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂。所得粗品中加入甲基叔丁基醚(3mL),室温下搅拌20分钟,过滤,滤饼用甲基叔丁基醚(2mL×2)淋洗,收集滤饼,真空干燥,得到化合物BB-27-6。
1H NMR(400MHz,CDCl
3)δ:7.98(d,J=9.6Hz,1H),7.89(br d,J=8.0Hz,1H),7.57(d,J=8.8Hz,1H),7.01(d,J=2.4Hz,1H),6.86(dd,J=2.4,8.8Hz,1H),6.68(d,J=9.2Hz,1H),4.38-4.28(m,1H),4.11-4.00(m,1H),3.49(br s,2H),3.70-3.57(m,4H),2.25-2.13(m,4H),1.77(br s,2H),1.72-1.63(m,2H),1.52-1.40(m,2H),1.11-1.03(m,1H).
步骤6:化合物BB-27的合成
室温和氮气保护下,将化合物BB-27-6(100mg,213.70μmol)溶于二氯甲烷(2mL)中,加入戴斯-马丁试剂(135.96mg,320.55μmol),反应混合物室温下搅拌反应3小时。反应结束后,过滤,向滤液中加入饱和碳酸氢钠溶液(10mL),室温下搅拌5分钟,用二氯甲烷(20mL×2)萃取,收集有机相,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂,得到化合物BB-27。MS-ESI m/z:466.1[M+H]
+.
参考例28:片段BB-28
合成路线:
步骤1:化合物BB-28-2的合成
室温下,将化合物BB-28-1(9.5g,54.29mmol)和三乙胺(10.99g,108.58mmol,15.11mL)溶于乙醇(100mL)中,加入醋酸钯(609.43mg,2.71mmol)和4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(2.51g,4.34mmol),反应混合物在室温和一氧化碳(15psi)氛围下升温至85℃,搅拌反应14小时。反应完毕后,冷却至室温,加入乙酸乙酯(100mL),过滤,滤饼用乙酸乙酯(100mL×4)淋洗,滤液减压浓缩除去溶剂。所得残余物经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=3/1-1/1,体积比),得到化合物BB-28-2。MS-ESI m/z:169.2[M+H]
+.
1H NMR(400MHz,CDCl
3)δ:8.80(s,1H),6.54-5.89(m,2H),4.51(q,J=7.2Hz,2H),1.45(t,J=7.2Hz,3H).
步骤2:化合物BB-28-3的合成
室温和氮气保护下,将化合物BB-28-2(1.8g,10.70mmol)和二水合氯化铜(2.19g,12.85mmol)溶于乙腈(30mL)中,加入亚硝酸叔丁酯(1.66g,16.06mmol,1.91mL),反应混合物升温至60℃,搅拌反应1小时。反应完毕后,冷却至室温,加入盐酸(1N,30mL),用乙酸乙酯(30mL×3)萃取。合并有机相,用饱和食盐水(80mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂。所得残余物经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=15/1-5/1,体积比),得到化合物BB-28-3。MS-ESI m/z:188.0[M+H]
+.
1H NMR(400MHz,CDCl
3)δ:9.12(s,1H),4.59(q,J=7.2Hz,2H),1.50(t,J=7.2Hz,3H).
步骤3:化合物BB-28-4的合成
室温和氮气保护下,将中间体BB-28-3(0.65g,3.47mmol)和4-哌啶甲醇(598.63mg,5.20mmol)溶于1,4-二氧六环(10mL)中,加入N,N-二异丙基乙胺(1.34g,10.40mmol,1.81mL),反应混合物升温至100℃,搅拌反应14小时。反应完毕后,冷却至室温,减压浓缩除去溶剂。残余物中加入水(20mL),用乙酸乙酯(20mL×4)萃取。合并有机相,用饱和食盐水(80mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂。所得残余物中加入石油醚(20mL)和乙酸乙酯(4mL),室温下搅拌0.5小时,过滤,滤饼经石油醚(20mL)洗涤,收集滤饼,真空干燥,得到化合物BB-28-4。MS-ESI m/z:267.1[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:8.68(s,1H),5.22-4.56(m,2H),4.53(t,J=5.2Hz,1H),4.35(q,J=7.2Hz,2H),3.28(t,J=5.6Hz,2H),3.17-3.00(m,2H),1.87-1.68(m,3H),1.32(t,J=7.2Hz,3H),1.22-1.06(m,2H).
步骤4:化合物BB-28-5的合成
室温和氮气保护下,将化合物BB-28-4(0.54g,2.03mmol)和咪唑(186.37mg,2.74mmol)溶于二氯甲烷(10mL)中,加入叔丁基二苯基氯硅烷(613.11mg,2.23mmol,573.00μL)。反应混合物室温下搅拌反应1小时。反应完毕后,加入饱和氯化铵溶液(20mL),用二氯甲烷(20mL×2)萃取。合并有机相,分别用饱和氯化铵溶液(40mL×3)和饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂,得到化合物BB-28-5。MS-ESI m/z:505.3[M+H]
+。
步骤5:化合物BB-28-6的合成
室温和氮气保护下,化合物BB-28-5(1g,1.98mmol)溶于乙醇(10mL)、四氢呋喃(10mL)和水(3mL)中,加入一水合氢氧化锂(249.44mg,5.94mmol)。反应混合物室温下搅拌反应过夜。反应完毕后,减压浓缩除去溶剂。残余物中加入水(10mL),用盐酸(1N)调节pH=2-3,用乙酸乙酯(20mL×4)萃取。合并有机相,用饱和食盐水(80mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂,得到化合物BB-28-6。MS-ESI m/z:477.2[M+H]
+.
步骤6:化合物BB-28-7的合成
室温和氮气保护下,将化合物BB-28-6(1.1g,2.31mmol)和N,N-二异丙基乙胺(1.19g,9.23mmol,1.61mL)溶于N,N-二甲基甲酰胺(10mL)中,再加入2-(7-偶氮苯并三氮唑)-N,N,N,N-四甲基脲六氟磷酸酯(1.14g,3.00mmol),反应混合物室温下搅拌反应5分钟,加入中间体BB-26-7的盐酸盐(662.77mg,2.31mmol),反应混合物室温下继续搅拌55分钟。反应完毕后,加入水(20mL),用乙酸乙酯(20mL×3)萃取。合并有机相,用饱和食盐水(50mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂。所得残余 物经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=3/1-1/1,体积比),得到化合物BB-28-7。MS-ESI m/z:709.3[M+H]
+.
1H NMR(400MHz,CDCl
3)δ:8.82(s,1H),7.68-7.64(m,4H),7.57(d,J=8.8Hz,2H),7.46-7.38(m,6H),7.01(d,J=2.4Hz,1H),6.86(dd,J=2.4,8.8Hz,1H),5.17-4.70(m,2H),4.35-4.28(m,1H),4.09-4.03(m,1H),3.55(d,J=5.6Hz,2H),3.12-3.00(m,2H),2.24-2.13(m,4H),1.99-1.86(m,3H),1.76-1.64(m,2H),1.52-1.43(m,2H),1.36-1.29(m,2H),1.06(s,9H).
步骤7:化合物BB-28-8的合成
室温下,将化合物BB-28-7(0.5g,704.87μmol)溶于N,N-二甲基甲酰胺(6mL)中,加入氟化钾(409.51mg,7.05mmol),反应混合物升温至100℃,搅拌反应过夜。反应完毕后,冷却至室温。加入水(30mL),用乙酸乙酯(20mL×3)萃取。合并有机相,用饱和食盐水(60mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂。所得残余物经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=3/1-1/2,体积比),得到化合物BB-28-8。MS-ESI m/z:471.2[M+H]
+.
1H NMR(400MHz,CDCl
3)δ:8.82(s,1H),7.56(d,J=8.4Hz,2H),7.00(d,J=2.0Hz,1H),6.86(dd,J=2.4,8.8Hz,1H),4.98(br s,2H),4.37-4.25(m,1H),4.09-4.02(m,1H),3.57(d,J=5.6Hz,2H),3.07(brt,J=12.4Hz,2H),2.22-2.14(m,4H),1.99-1.82(m,3H),1.75-1.67(m,2H),1.52-1.41(m,2H),1.37-1.28(m,2H).
步骤8:化合物BB-28的合成
室温和氮气保护下,将化合物BB-28-8(0.257g,545.70μmol)溶于二氯甲烷(5mL)中,冷却至0℃,加入戴斯-马丁氧化剂(347.18mg,818.56μmol),反应混合物室温下搅拌反应1.5小时。反应完毕后,过滤,滤饼用乙酸乙酯(30mL)洗涤。向滤液中加入饱和碳酸氢钠(20mL),搅拌5分钟,用乙酸乙酯(20mL×3)萃取。合并有机相,用饱和食盐水(60mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂,得到化合物BB-28。MS-ESI m/z:469.2[M+H]
+。
参考例29:片段BB-29
合成路线:
步骤1:化合物BB-29-2的合成
化合物BB-29-1(5g,28.97mmol)溶于二甲亚砜(80mL),依次加入4-哌啶甲醇(3.34g,28.97mmol),N,N-二异丙基乙胺(7.49g,57.95mmol,10.09mL),在氮气保护下反应混合物在120℃搅拌12小时,反应完毕,反应混合物冷却至室温,加入乙酸乙酯(200mL)和去离子水(200mL),分离有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压除去溶剂,所得粗品经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/1,体积比),得到化合物BB-29-2。MS-ESI m/z:273.9[M+Na]
+.
1H NMR(400MHz,DMSO_d
6)δ:8.62(d,J=1.2Hz,1H),8.35(d,J=1.2Hz,1H),4.55-4.46(m,3H),3.80(s,3H),3.27(t,J=5.8Hz,2H),3.03-2.92(m,2H),1.80-1.64(m,3H),1.20-1.05(m,2H).
步骤2:化合物BB-29-3的合成
化合物BB-29-2(5g,19.90mmol)溶于甲醇(100mL)和去离子水(25mL)混合溶剂中,加入氢氧化锂(2.50g,59.69mmol),在氮气保护下反应混合物在25℃搅拌12小时,反应完毕后,反应混合物减压浓缩除去甲醇,在0-5℃冷却下,用稀盐酸(1M)调节溶液pH=3-4,反应混合物有白色沉淀析出,减压过滤,滤饼用去离子水洗涤,收集滤饼,40-45℃下真空干燥,得到化合物BB-29-3。MS-ESI m/z:237.9[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:12.63(br s,1H),8.61(s,1H),8.33(s,1H),4.63-4.39(m,3H),3.27(br d,J=6.0Hz,2H),3.02-2.89(m,2H),1.80-1.63(m,3H),1.19-1.04(m,2H).
步骤3:化合物BB-29-4的合成
室温和氮气保护下,将化合物BB-26-7(4g,13.93mmol)和化合物BB-29-3(3.30g,13.93mmol)溶于N,N-二甲基甲酰胺(100mL)中,依次加入三乙胺(7.05g,69.64mmol,9.69mL),1-羟基苯并三唑(2.82g,20.89mmol)和1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐(4.01g,20.89mmol),反应混合物在25℃搅拌12小时。反应完毕,加入冰水(300mL)和乙酸乙酯(200mL),分离有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压除去溶剂,所得粗品经过柱层析分离(洗脱剂:石油醚/乙酸乙酯 =1/1,体积比),得到化合物BB-29-4。MS-ESI m/z:470.1[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:8.58(d,J=0.8Hz,1H),8.23(d,J=1.2Hz,1H),8.07(d,J=8.4Hz,1H),7.85(d,J=8.8Hz,1H),7.35(d,J=2.0Hz,1H),7.11(dd,J=2.4,8.8Hz,1H),4.56-4.41(m,4H),3.90-3.73(m,1H),3.27(t,J=5.4Hz,2H),3.00-2.88(m,2H),2.14-2.02(m,2H),1.92-1.82(m,2H),1.80-1.66(m,3H),1.65-1.43(m,4H),1.19-1.05(m,2H).
步骤4:化合物BB-29的合成
在室温和氮气保护下,化合物BB-29-4(2g,4.26mmol)溶于二氯甲烷(100mL),加入戴斯-马丁试剂(2.71g,6.38mmol,1.98mL),反应混合物在室温搅拌2小时,反应完毕,加入亚硫酸钠饱和溶液(50mL)淬灭,分离有机相,用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压除去溶剂,所得粗品经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/1,体积比),得到化合物BB-29。MS-ESI m/z:468.2[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:9.62(s,1H),8.59(d,J=0.8Hz,1H),8.27(d,J=0.8Hz,1H),8.10(d,J=8.4Hz,1H),7.85(d,J=8.8Hz,1H),7.36(d,J=2.0Hz,1H),7.12(dd,J=2.2,9.0Hz,1H),4.57-4.45(m,1H),4.35-4.20(m,2H),3.90-3.75(m,1H),3.29-3.18(m,2H),2.75-2.60(m,1H),2.15-2.02(m,2H),2.00-1.83(m,4H),1.69-1.44(m,6H).
实施例1
合成路线:
步骤1:化合物WX001-1的合成
将化合物BB-3(0.14g,475.08μmol)和化合物BB-4-5的盐酸盐(285.34mg,498.83μmol)溶于乙腈(8mL)中,之后加入N,N-二异丙基乙胺(184.20mg,1.43mmol,248.25μL),氮气保护下升温至50℃搅拌 12小时。反应完毕后,将反应液减压浓缩得到残余物。残余物通过柱层析(洗脱剂:二氯甲烷/甲醇=100/1-30/1,体积比)纯化得到化合物WX001-1。
1H NMR(400MHz,DMSO_d
6)δ:8.39(d,J=8.4Hz,1H),8.27(d,J=1.6Hz,1H),8.17(dd,J=0.8Hz,4.8Hz,2H),8.06(dd,J=1.6Hz,8.4Hz,1H),7.35(t,J=9.2Hz,1H),7.30(dd,J=2.4Hz,12.0Hz,1H),7.15(d,J=8.8Hz,1H),4.27(s,2H),4.22(t,J=5.4Hz,2H),4.15(q,J=7.2Hz,2H),3.90(s,2H),2.81-2.72(m,2H),2.65-2.52(m,8H),1.50(s,6H),1.20(t,J=7.2Hz,3H).
步骤2:化合物WX001的合成
将化合物WX001-1(0.012g,15.12μmol)溶于无水四氢呋喃(0.8mL)中,降温至-60℃,加入丙烯酰胺(2.15mg,30.23μmol)和叔丁醇钾(5.09mg,45.35μmol),氮气置换三次,-60℃下搅拌1小时,缓慢恢复至15℃搅拌12小时。反应完毕后,将反应液减压浓缩得到残余物。残余物经制备HPLC(流动相:乙腈/水;酸性体系:0.04%盐酸)分离纯化得到化合物WX001。MS-ESI m/z:819.3[M+H]
+.
1H NMR(400MHz,CD
3OD)δ:8.15(d,J=8.0Hz,2H),8.11(s,1H),7.97(dd,J=1.6Hz,8.4Hz,1H),7.91(s,1H),7.35(t,J=9.2Hz,1H),7.31(dd,J=1.6Hz,11.6Hz,1H),7.24-7.20(m,1H),4.62(dd,J=4.8Hz,10.8Hz,1H),4.56(t,J=4.4Hz,2H),4.13(s,2H),3.72(t,J=4.4Hz,2H),3.47-3.35(m,4H),3.28-3.19(m,4H),2.86-2.80(m,2H),2.70-2.58(m,1H),2.45-2.37(m,1H),1.56(s,6H).
实施例2
合成路线:
步骤1:化合物WX002-1的合成
将化合物BB-8(0.4g,1.21mmol)加入至1,2-二氯乙烷(10mL)中,依次加入化合物BB-6的盐酸盐(293.89mg,1.21mmol)和乙酸钾(118.84mg,1.21mmol),室温下搅拌0.5小时,慢慢加入三乙酰氧基硼氢化钠(384.96mg,1.82mmol),室温反应11.5小时。反应完毕后,向反应液中加入1mol/L盐酸(10mL),20℃下搅拌0.5小时,过滤,收集滤饼得到化合物WX002-1。MS-ESI m/z:519.3[M-H]
+.
步骤2:化合物WX002的合成
将化合物WX002-1(0.12g,230.51μmol)加入至N,N-二甲基甲酰胺(3mL)中,依次加入2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(113.94mg,299.67μmol),N,N-二异丙基乙胺(89.38mg,691.54μmol,120.45μL),氮气置换三次后,升温至50℃反应1小时。降温至50℃,加入化合物BB-5的盐酸盐(72.67mg,230.51μmol),20℃下反应2小时。反应完毕后,将反应液倒入水(5mL)中,用乙酸乙酯(5mL×3)萃取,有机相用饱和食盐水(10mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品经制备HPLC(流动相:乙腈/水;酸性体系:0.04%盐酸)分离纯化得到化合物WX002。MS-ESI m/z:781.4[M+H]
+.
1H NMR(400MHz,CD
3OD)δ:7.80(d,J=8.8Hz,2H),7.72(d,J=8.8Hz,1H),7.64(d,J=8.8Hz,1H),7.16(d,J=1.6Hz,1H),7.13(d,J=2.4Hz,1H),7.09(d,J=8.8Hz,2H),6.99(t,J=2.8Hz,1H),6.97(t,J=2.8Hz,1H),4.44(d,J=5.0Hz,10.6Hz,1H),4.29(s,1H),4.15(t,J=6.0Hz,3H),4.05(d,J=13.6Hz,2H),3.71(d,J=12.0Hz,2H),3.29-3.21(m,4H),3.15(t,J=12.2Hz,2H),2.82-2.76(m,2H),2.58-2.47(m,1H),2.39-2.31(m,1H),1.98-1.85(m,4H),1.69-1.61(m,2H),1.28(s,6H),1.23(s,6H).
实施例3
合成路线:
步骤1:化合物WX003-1的合成
将化合物BB-9(0.12g,464.70μmol)加入至1,2-二氯乙烷(8mL)中,依次加入化合物BB-10的盐酸盐(164.45mg,464.70μmol)和乙酸钾(45.61mg,464.70μmol),氮气置换三次后,20℃下反应0.5小时,加入三乙酰氧基硼氢化钠(147.73mg,697.06μmol),20℃下继续搅拌11.5小时。反应完毕后,向反应液中加入1mol/L盐酸(5mL),20℃下搅拌0.5小时,过滤,收集滤饼得到化合物WX003-1。MS-ESI m/z:560.2[M+H]
+.
步骤2:化合物WX003的合成
将化合物WX003-1(0.13g,232.29μmol)加入至N,N-二甲基甲酰胺(3mL)中,依次加入2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(114.82mg,301.97μmol)和N,N-二异丙基乙胺(90.06mg,696.86μmol,121.38μL),氮气置换三次后,升温至50℃反应1小时。降温至20℃,加入化合物BB-5的盐酸盐(73.23mg,232.29μmol),20℃下继续反应2小时。反应完毕后,将反应液倒入水(5mL)中,用乙酸乙酯(5mL×3)萃取,有机相用饱和食盐水(10mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品经制备HPLC(流动相:乙腈/水;酸性体系:0.04%盐酸)分离纯化得到化合物WX003。MS-ESI m/z:820.4[M+H]
+.
1H NMR(400MHz,CD
3OD)δ:8.06-7.98(m,3H),7.95-7.88(m,1H),7.84(d,J=8.4Hz,2H),7.73(d,J=8.8Hz,1H),7.57-7.50(m,1H),7.13(d,J=2.0Hz,1H),6.99(dd,J=2.4Hz,8.8Hz,1H),4.82(s,2H),4.61(dd,J=5.0Hz,11.0Hz,1H),4.30(s,1H),4.18(s,1H),3.95-3.80(m,3H),3.79-3.65(m,9H),3.41(t,J=7.0Hz,2H),2.86-2.81(m,1H),2.64-2.58(m,1H),2.51-2.43(m,1H),2.42-2.33(m,1H),2.17(d,J=14.4Hz,2H),2.04- 1.98(m,1H),1.94-1.84(m,4H),1.30(s,6H),1.23(s,6H).
实施例4
合成路线:
步骤1:化合物WX004盐酸盐的合成
将化合物BB-11(0.2g,337.49μmol)和BB-12(82.76mg,337.49μmol)依次加入N,N-二甲基甲酰胺(10mL),随后加入N,N-二异丙基乙胺(218.09mg,1.69mmol,293.93μL)和2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(192.49mg,506.24μmol),反应混合物在25℃搅拌3小时。反应完毕,加入冰水(50mL),乙酸乙酯(50mL)萃取,有机相经饱和食盐水(50mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压除去溶剂,所得残余物经过制备HPLC分离(流动相:乙腈/水;酸性体系:0.05%盐酸),得到化合物WX004的盐酸盐。MS-ESI m/z:820.3[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.15(s,1H),10.72(s,1H),10.39(s,1H),8.40(d,J=8.4Hz,1H),8.28(d,J=0.4Hz,1H),8.21-8.15(m,1H),8.07(d,J=8.0Hz,1H),7.76-7.69(m,2H),7.45-7.36(m,2H),7.24(t,J=8.0Hz,1H),4.65-4.54(m,3H),3.64-3.50(m,4H),3.21-3.06(m,2H),2.87-2.74(m,1H),2.71-2.60(m,1H),2.46-2.37(m,1H),2.34(d,J=6.8Hz,2H),2.27-2.18(m,1H),2.13-2.01(m,1H),1.90(d,J=13.6Hz,2H),1.74-1.60(m,2H),1.52(s,6H).
实施例5
合成路线:
步骤1:化合物WX005的合成
将化合物BB-13(120mg,212.56μmol)和化合物BB-12的盐酸盐(87.33mg,277.57μmol)加到N,N-二甲基甲酰胺(3mL)中,将2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(121.23mg,318.84μmol)和三乙胺(64.53mg,637.67μmol)慢慢的加到反应中,混合物用氮气置换3次后在25℃搅拌2小时。反应液倒入20mL水和30mL乙酸乙酯中,分出有机相经水洗涤(20mL×2),收集有机相用无水硫酸钠干燥,过滤,减压旋干。粗品经制备HPLC纯化(流动相:乙腈/水;酸性体系:0.05%盐酸),得到化合物WX005。MS-ESI m/z:792.1[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.15(s,1H),10.38(br s,1H),8.39(d,J=8.4Hz,1H),8.27(d,J=1.6Hz,1H),8.18(d,J=6.0Hz,1H),8.06(dd,J=1.6,8.0Hz,1H),7.82-7.72(m,2H),7.51-7.41(m,1H),7.37(d,J=12.8Hz,1H),7.25-7.15(m,1H),4.92-4.68(m,1H),4.63(dd,J=5.0,11.8Hz,1H),4.28(d,J=13.2Hz,2H),3.71-3.50(m,1H),3.48-3.35(m,2H),2.90-2.75(m,1H),2.72-2.59(m,1H),2.45-2.36(m,1H),2.35-2.20(m,4H),2.19-1.98(m,2H),1.52(s,6H).
实施例6
合成路线:
步骤1:化合物WX006的合成
将化合物BB-4(100mg,168.47μmol)和化合物BB-2的盐酸盐(54.80mg,185.32μmol)加到N,N-二甲基甲酰胺(20mL)中,将2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(128.11mg,336.94μmol)和N,N-二异丙基乙胺(54.43mg,421.18μmol,73.36μL)慢慢加到反应中,混合物氮气置换3次后在25℃搅拌12小时。反应液用乙酸乙酯(30mL)稀释,有机相用水洗涤(20mL×3),收集有机相用无水硫酸钠干燥,过滤,减压旋干所得残渣经制备HPLC分离(流动相:乙腈/水;酸性体系:0.05%盐酸),得到化合物WX006。MS-ESI m/z:835.2[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.20(s,1H),8.39(d,J=8.0Hz,1H),8.28(s,1H),8.22(s,1H),8.06(d,J=8.4Hz,1H),7.82(d,J=8.8Hz,1H),7.52(d,J=8.0Hz,1H),7.43(t,J=9.0Hz,1H),7.38(dd,J=1.8,11.8Hz,1H),7.22(d,J=8.4Hz,1H),4.66(dd,J=5.0,11.8Hz,1H),4.64-4.58(m,2H),4.20-3.91(m,2H),3.90-3.62(m,10H),3.05(s,3H),2.97-2.83(m,1H),2.81-2.70(m,1H),2.47-2.27(m,1H),2.25-2.13(m,1H),1.52(s,6H).
实施例7
合成路线:
步骤1:化合物WX007的盐酸盐的合成
将化合物BB-20的盐酸盐(77.78mg,152.52μmol)溶于1,2-二氯乙烷(2mL)中,随后加入化合物BB-19(60mg,152.52μmol)和醋酸钾(25.15mg,256.23μmol),反应体系置换氮气三次,25℃搅拌20分钟后,加醋酸硼氢化钠(48.49mg,228.78μmol),25℃搅拌12小时。反应完毕,向反应液加水(5mL)和二氯甲烷(10mL),分离有机相,水相加二氯甲烷(20mL×3)萃取合并有机相,用饱和食盐水(20mL×2)洗涤,然后用无水硫酸钠干燥,过滤,减压浓缩得到粗产物经制备HPLC分离(流动相:乙腈/水;酸性体系:0.04%盐酸),得到化合物WX007的盐酸盐。MS-ESI m/z:851.4[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.14(s,1H),10.61(s,1H),10.43(s,1H),8.55(s,1H),8.38(d,J=8.0Hz,1H),8.29(s,1H),8.19(d,J=8.0Hz, 1H),8.09(t,J=9.2Hz,2H),7.87(t,J=10.8Hz,2H),7.24(d,J=8.0Hz,2H),7.14(d,J=8.0Hz,2H),5.02(dd,J=3.8,11.0Hz,1H),3.93(d,J=11.6Hz,2H),3.61(d,J=9.6Hz,2H),3.23-3.12(m,6H),2.90-2.75(m,1H),2.70-2.55(m,3H),2.45-2.30(m,2H),1.88-1.77(m,2H),1.76-1.64(m,2H),1.48(s,6H).
实施例8
合成路线:
步骤1:化合物WX008的合成
将化合物BB-4(60mg,101.08μmol)和化合物BB-14的盐酸盐(34.17mg,121.30μmol)加到N,N-二甲基甲酰胺(10mL)中,将2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(76.87mg,202.16μmol)和N,N-二异丙基乙胺(39.19mg,303.24μmol,52.82μL)慢慢加到反应中,混合物氮气置换3次后在25℃搅拌30分钟。反应液用乙酸乙酯(30mL)稀释,有机相用水洗涤(20mL×3),收集有机相用无水硫酸钠干燥,过滤,减压旋干。所得粗品经制备HPLC分离(流动相:乙腈/水;酸性体系:0.05%盐酸),得到化合物WX008。MS-ESI m/z:821.4[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.09(s,1H),10.97(s,1H),8.40(d,J=8.4Hz,1H),8.28(s,1H),8.22(s,1H),8.06(d,J=8.0Hz,1H),7.81(d,J=8.4Hz,1H),7.50(d,J=8.4Hz,1H),7.43(d,J=8.8Hz,1H),7.38(dd,J=2.4,11.6Hz,1H),7.22(d,J=8.8Hz,1H),4.63-4.52(m,3H),4.18-3.60(m,12H),2.85-2.72(m,1H),2.69-2.57(m,1H),2.48-2.32(m,1H),2.26-2.16(m,1H),1.52(s,6H).
实施例9
合成路线:
步骤1:化合物WX009的合成
将化合物BB-15(0.08g,129.11μmol)置于N,N-二甲基甲酰胺(2mL)中,加入N,N-二异丙基乙胺(50.06mg,387.33μmol,67.46μL)和2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸盐(98.18mg,258.22μmol),随后加入化合物BB-14的盐酸盐(44.33mg,180.75μmol),反应液在20℃搅拌12小时,随后升温至45℃搅拌6小时。反应完毕,反应液直接浓缩得到粗品,通过制备HPLC分离(流动相:乙腈/水;酸性体系:0.04%盐酸)纯化,得到化合物WX009。MS-ESI m/z:847.4[M+H]
+.
1H NMR(400MHz,CD
3OD)δ:8.22(d,J=1.2Hz,1H),8.16(d,J=2.8Hz,1H),8.14(d,J=3.6Hz,1H),7.98(dd,J=1.8,8.2Hz,1H),7.75(d,J=8.4Hz,1H),7.34(dd,J=1.6,8.8Hz,1H),7.29(d,J=2.4Hz,1H),7.28-7.23(m,1H),7.20-7.17(m,1H),4.49(dd,J=5.0,11.0Hz,1H),4.27(s,2H),4.14(s,2H),3.48-3.35(m,8H),2.82-2.77(m,2H),2.60-2.50(m,1H),2.40-2.30(m,1H),1.57(s,6H),1.02-0.88(m,4H).
实施例10
合成路线:
步骤1:化合物WX010的合成
将化合物BB-16(100mg,164.58μmol)和化合物BB-14的盐酸盐(86.95mg,197.50μmol,纯度:63.98%)加到N,N-二甲基甲酰胺(10mL)中,随后加入2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸盐(125.15mg,329.16μmol)和N,N-二异丙基乙胺(63.81mg,493.74μmol,86.00μL),反应液在氮气氛围下,25℃搅拌2小时。反应完毕,加入乙酸乙酯(30mL)稀释,有机相经水洗涤(20mL×3),合并的有机相经无水硫酸钠干燥,过滤,减压旋干所得粗品经制备HPLC分离(流动相:乙腈/水;酸性体系:0.05%盐酸),得到化合物WX010。MS-ESI m/z:835.2[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.08(s,1H),10.81(s,1H),8.40(d,J=8.4Hz,1H),8.27(d,J=1.2Hz,1H),8.22(s,1H),8.06(dd,J=1.6,8.0Hz,1H),7.78(d,J=8.8Hz,1H),7.45-7.33(m,3H),7.24-7.16(m,1H),4.54(d,J=4.8,11.6Hz,1H),4.52-4.35(m,2H),3.77-3.45(m,10H),3.13-2.85(m,4H),2.82-2.73(m,1H),2.69-2.64(m,1H),2.40-2.30(m,1H),2.25-2.15(m,1H),1.52(s,6H).
实施例11
合成路线:
步骤1:化合物WX011的合成
将化合物BB-4(107.36mg,180.86μmol)溶于N,N-二甲基甲酰胺(2mL)中,然后加入2-(7-氮杂苯并 三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(137.53mg,361.71μmol)和N,N-二异丙基乙胺(116.87mg,904.29μmol,157.51μL),在25℃下搅拌30分钟,加入化合物BB-17的盐酸盐(60mg,180.86μmol),25℃搅拌3小时。向反应液中加入水(5mL)和乙酸乙酯(10mL),分离有机相,水相用乙酸乙酯(20mL×3)萃取,合并有机相用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物经制备HPLC分离(流动相:乙腈/水;酸性体系:0.04%盐酸)纯化得到化合物WX011。MS-ESI m/z:871.3[M+H]
+.
1H NMR(400MHz,CD
3OD)δ:8.42(d,J=2.0Hz,1H),8.22-8.13(m,3H),8.03(d,J=9.2Hz,1H),7.98(dd,J=1.8,8.2Hz,1H),7.89(dd,J=2.0,8.8Hz,1H),7.78(d,J=9.2Hz,1H),7.36(t,J=9.2Hz,1H),7.31(dd,J=2.0,11.2Hz,1H),7.22(d,J=8.8Hz,1H),4.95(dd,J=5.0,10.2Hz,1H),4.54(t,J=4.6Hz,2H),3.71-3.62(m,4H),3.60-3.50(m,3H),3.28-3.05(m,3H),2.92-2.75(m,2H),2.74-2.62(m,1H),2.55-2.47(m,1H),2.03(s,2H),1.57(s,6H).
实施例12
合成路线:
步骤1:化合物WX012的合成
将化合物BB-4-5的盐酸盐(185.99mg,325.14μmol)溶于1,2-二氯乙烷(5mL)中,加入化合物BB-18(110mg,325.14μmol)和乙酸钠(53.35mg,650.28μmol),搅拌30分钟,加入三乙酰氧基硼氢化钠(137.82mg,650.28μmol),25℃下反应12小时。反应完毕,加入水(5mL)和二氯甲烷(10mL),分离有机相,水相用二氯甲烷(20mL×3)萃取,合并有机相,用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩。残余物经制备HPLC分离(流动相:乙腈/水;酸性体系:0.04%盐酸)纯化得到化合物WX012。MS-ESI m/z:858.3[M+H]
+.
1H NMR(400MHz,CD
3OD)δ:8.20-8.14(m,3H),8.05(d,J=9.6Hz,1H),7.98(dd,J=1.8Hz,8.2Hz,1H),7.78(d,J=9.2Hz,1H),7.65(d,J=2.8Hz,1H),7.49(dd,J=2.6,9.0Hz,1H),7.37(t,J=8.8Hz,1H),7.31(dd,J=2.4,11.6Hz,1H),7.25-7.21(m,1H),4.93(dd,J=5.0,10.2Hz,2H),4.62-4.55 (m,4H),3.82-3.65(m,11H),2.90-2.75(m,2H),2.73-2.60(m,1H),2.55-2.45(m,1H),1.56(s,6H).
实施例13
合成路线:
步骤1:化合物WX013-1的合成
将化合物BB-20(0.2g,422.37μmol)和2-氯乙氧基乙酸乙酯(140.74mg,844.75μmol)溶于N,N-二甲基甲酰胺(3mL)中,加入碳酸钾(233.50mg,1.69mmol)和碘化钾(14.02mg,84.47μmol),氮气保护下反应在90℃搅拌15小时。反应液中加入水(10mL),用乙酸乙酯(3×10mL)萃取,合并有机相经饱和食盐水(3×30mL)洗涤,用无水硫酸钠干燥,过滤,滤液减压浓缩。所得残留物经柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/1-0/1,体积比),得到化合物WX013-1。MS-ESI m/z:604.3[M+H]
+.
1H NMR(400MHz,CDCl
3)δ:8.01-7.95(m,2H),7.85(dd,J=1.6,8.0Hz,1H),7.17(d,J=8.4Hz,2H),7.01(d,J=8.8Hz,2H),4.25(q,J=7.2Hz,2H),4.14(s,2H),3.95-3.60(m,4H),3.50-3.20(m,4H),2.85-2.60(m,4H),1.58(s,6H),1.31(t,J=7.2Hz,3H).
步骤2:化合物WX013-2的合成
将化合物WX013-1(0.135g,223.64μmol)溶于乙醇(0.5mL)、四氢呋喃(2mL)和水(0.5mL)的混合溶剂中,随后加入一水合氢氧化锂(28.15mg,670.91μmol),反应液在氮气保护下在20℃搅拌14小时。反应完毕,减压浓缩除去溶剂,所得残余物加入1M盐酸水溶液(10mL),用二氯甲烷/乙醇(10/1,体积比)的混合溶剂(5×10mL)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩得到化合物WX013-2。
1H NMR(400MHz,CDCl
3)δ:12.81(s,1H),8.01-7.95(m,2H),7.84(dd,J=1.8Hz,8.2Hz,1H),7.30-7.28(m,2H),7.21(d,J=8.4Hz,2H),4.20(s,2H),4.14-4.08(m,2H),4.00-3.86(m,4H),3.83-3.77(m,2H),3.43-3.38(m, 2H),3.37-3.28(m,2H),1.59(s,6H).
步骤3:化合物WX013的合成
将化合物BB-17的盐酸盐(28.82mg,86.87μmol)和化合物WX013-2(0.05g,86.87μmol)溶于N,N-二甲基甲酰胺(2mL)中,之后加入2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(49.54mg,130.30μmol)和N,N-二异丙基乙胺(33.68mg,260.60μmol,45.39μL),氮气保护下反应在20℃搅拌14小时。反应物完毕,过滤,滤液经制备制备HPLC分离(流动相:乙腈/水;酸性体系:0.04%盐酸)纯化得到化合物WX013。MS-ESIm/z:853.3[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.13(s,1H),10.30(s,1H),8.54(d,J=1.6Hz,1H),8.39(d,J=8.4Hz,1H),8.29(d,J=1.6Hz,1H),8.23(d,J=8.8Hz,1H),8.13(d,J=8.8Hz,1H),8.08(dd,J=1.6,8.4Hz,1H),7.91(d,J=9.2Hz,2H),7.25(d,J=8.8Hz,2H),7.17(d,J=9.2Hz,2H),5.03(dd,J=4.8Hz,11.6Hz,1H),4.31(s,2H),4.03-3.95(m,4H),3.83-3.73(m,2H),3.57-3.47(m,2H),3.31-3.18(m,4H),2.88-2.77(m,1H),2.69-2.59(m,3H),1.49(s,6H).
实施例14
合成路线:
步骤1:化合物WX014-2的合成
室温下,将化合物WX014-1(10g,78.67mmol)溶于二氯甲烷(100ml)和丙酮(50mL)中,0℃下向其中加入三甲基氰硅烷(11.71g,118.00mmol,14.76mL)和三甲硅基三氟甲磺酸酯(1.75g,7.87mmol,1.42mL),反应混合物在20℃和氮气保护下搅拌反应2小时。反应完毕后,滤液减压浓缩除去溶剂。所得残余物经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/0-4/1,体积比),得到化合物WX014-2。
步骤2:化合物WX014-3的合成
室温下,将化合物WX014-2(9.83g,50.62mmol)溶于N,N-二甲基乙酰胺(90mL)中,向其中加入4-氰基-3-三氟甲基异硫氰酸苯酯(11.55g,50.62mmol),反应混合物在室温和氮气保护下搅拌反应3小时,然后分别加入盐酸(2M,98.20mL)和甲醇(90mL),反应混合物加热至70℃并搅拌反应2小时。反应完毕后,冷却至室温,向混合物加入水(150mL),用乙酸乙酯(150mL×3)萃取。合并有机相,依次用饱和食盐水(150mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂。所得残余物经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/0-4/1,体积比),得到化合物WX014-3。
1H NMR(400MHz,CDCl
3)δ:7.98(d,J=8.4Hz,1H),7.96(d,J=1.6Hz,1H),7.84(dd,J=2.0,8.4Hz,1H),7.16(t,J=8.8Hz,1H),7.06(dd,J=1.2,10.8Hz,1H),7.01-6.95(m,1H),1.59(s,6H).
步骤3:化合物WX014-4的合成
室温下,将化合物WX014-3(1g,2.22mmol,纯度:93.85%)溶于二氯甲烷(10mL)中,向其中加入三氟甲磺酸酐(938.11mg,3.32mmol,548.60μL)和三乙胺(672.91mg,6.65mmol,925.59μL),反应混合物在室温氮气保护下搅拌反应2小时。反应完毕后,向混合物加入水(15mL),用乙酸乙酯(15mL×3)萃取。 合并有机相,依次用饱和食盐水(15mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂。所得残余物经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/0-9/1,体积比),得到化合物WX014-4。
步骤4:化合物WX014-5的合成
室温和氮气保护下,将化合物WX014-4(756mg,1.36mmol)和哌嗪-1-羧酸叔丁酯盐酸盐(1.52g,6.81mmol)中溶于甲苯(10mL)和水(1mL)中,分别加入磷酸钾(866.74mg,4.08mmol),三(二亚苄基丙酮)二钯(87.25mg,95.27μmol)和2-二叔丁基膦-2′,4′,6′-三异丙基联苯(80.92mg,190.55μmol),反应混合物加热至100℃并搅拌反应12小时。反应完毕后,冷却至室温,向混合物加入水(10mL),用乙酸乙酯(10mL×3)萃取。合并有机相,依次用饱和食盐水(10mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩除去溶剂。所得残余物经过柱层析分离(洗脱剂:石油醚/乙酸乙酯=1/0-5/1,体积比),得到化合物WX014-5。
步骤5:化合物WX014-6的合成
室温下,将化合物WX014-5(333mg,562.86μmol)溶于盐酸乙酯(4M,2.83mL)中,反应混合物在20℃氮气保护下搅拌反应12小时。反应完毕后,滤液减压浓缩除去溶剂。得到化合物WX014-6。
1H NMR(400MHz,CDCl
3)δ:10.0(br s,1H),7.99(d,J=8.0Hz,1H),7.95(s,1H),7.83(d,J=8.0Hz,1H),7.18-6.97(m,3H),3.75-3.25(m,8H),1.60(s,6H).
步骤6:化合物WX014的合成
将化合物WX014-6的盐酸盐(80.52mg,152.52μmol)溶于1,2-二氯乙烷(5mL)中,加入化合物BB-24(50mg,127.10μmol)和乙酸钾(18.71mg,190.65μmol),在20℃搅拌1小时,加入三乙酰氧基硼氢化钠(61.96mg,292.33μmol),氮气氛围下20℃下反应11小时。反应完毕,反应液倒入稀盐酸中(10mL),加入5mL的水,用乙酸乙酯萃取(15mL×3),有机相用饱和食盐水洗涤(10mL×2),无水硫酸钠干燥,过滤,减压浓缩。残余物经制备HPLC分离(流动相:乙腈/水;酸性体系:0.04%盐酸)纯化得到化合物WX014。MS-ESI m/z:869.3[M+H]
+.
1H NMR(400MHz,CD
3OD)δ:8.26(d,J=9.6Hz,1H),8.19-8.14(m,3H),7.98(dd,J=1.6,8.4Hz,1H),7.84(d,J=9.2Hz,1H),7.74(t,J=7.8Hz,1H),7.68(d,J=7.2Hz,1H),7.27-7.16(m,3H),5.00(dd,J=5.0Hz,10.2Hz,1H),3.72(d,J=11.6Hz,4H),3.42-3.33(m,4H),3.22(t,J=11.6Hz,2H),2.93-2.78(m,2H),2.75-2.65(m,3H),2.56-2.47(m,1H),2.03-1.86(m,4H),1.56(s,6H).
实施例15
合成路线:
步骤1:化合物WX015的合成
将化合物BB-23的盐酸盐(75.24mg,157.60μmol)溶于1,2-二氯乙烷(2mL)中,加入化合物BB-19(62mg,157.60μmol)、三乙酰氧基硼氢化钠(66.81mg,315.21μmol)和醋酸(0.1mL),氮气氛围下20℃搅拌20分钟,加入乙酸钾(30.94mg,315.21μmol),在20℃下继续搅拌12小时。反应完毕,加入水(5mL)和二氯甲烷(10mL),分离有机相,水相加二氯甲烷(20mL×3)萃取,合并有机相经饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤,减压浓缩得到残余物经制备HPLC分离(流动相:乙腈/水;酸性体系:0.04%盐酸)纯化得到化合物WX015。MS-ESI m/z:818.4[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.13(s,1H),10.80(s,1H),10.40(s,1H),8.68(d,J=8.4Hz,1H),8.53(d,J=1.2Hz,1H),8.19(d,J=8.4Hz,1H),8.09(d,J=8.8Hz,1H),7.97-7.80(m,4H),7.48(d,J=9.6Hz,1H),7.38(d,J=2.4Hz,1H),7.13(dd,J=2.4Hz,8.8Hz,1H),5.03(dd,J=4.8Hz,11.2Hz,1H),4.59(d,J=14.4Hz,2H),4.54-4.47(m,1H),3.92-3.83(m,1H),3.61(d,J=10.8Hz,2H),3.56-3.48(m,2H),3.23-3.05(m,4H),2.88-2.77(m,1H),2.70-2.58(m,2H),2.45-2.32(m,2H),2.15-2.05(m,2H),1.95-1.77(m,4H),1.75-1.58(m,4H),1.57-1.45(m,2H).
实施例16
合成路线:
步骤1:化合物WX016的合成
室温下,将化合物BB-23(50mg,127.10μmol)加入至1,2-二氯乙烷(10mL)中,慢慢加入化合物BB-24的盐酸盐(72.81mg,152.52μmol)和乙酸钾(18.71mg,190.65μmol),搅拌1小时后,加入三乙酰氧基硼氢化钠(61.96mg,292.33μmol),氮气保护下反应过夜。将反应液倒入稀盐酸中(10mL),加入水(5mL),用乙酸乙酯(15mL×3)萃取,收集有机相,用饱和食盐水(10mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品。粗品经制备HPLC(流动相:乙腈/水;酸性体系:0.04%盐酸)分离纯化得到化合物WX016。MS-ESI m/z:818.4[M+H]
+.
1H NMR(400MHz,CD
3OD)δ:8.25(d,J=9.6Hz,1H),8.18-8.11(m,2H),7.83(d,J=9.6Hz,1H),7.73(t,J=8.0Hz,1H),7.71-7.65(m,2H),7.62(d,J=9.6Hz,1H),7.20(d,J=2.4Hz,1H),7.04(dd,J=2.4,8.8Hz,1H),5.05-4.95(m,5H),4.75-4.65(m,2H),4.55-4.46(m,1H),4.04-3.95(m,1H),3.84-3.73(m,2H),3.61-3.52(m,2H),2.90-2.79(m,2H),2.76-2.66(m,3H),2.57-2.47(m,1H),2.25-2.17(m,2H),2.13-2.06(m,2H),2.02-1.95(m,2H),1.94-1.84(m,2H),1.71-1.58(m,4H).
实施例17
合成路线:
步骤1:化合物WX017的合成
室温下,将化合物BB-4(85.88mg,144.69μmol)加入至N,N-二甲基甲酰胺(8mL)中,依次加入2-(7-氮杂苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(68.77mg,180.86μmol)和N,N-二异丙基乙胺(77.92mg,602.86μmol,105.01μL),搅拌0.5小时后,加入化合物BB-22的盐酸盐(40mg,120.57μmol),氮气保 护下反应2.5小时。将反应液倒入水中(15mL),用乙酸乙酯(10mL×3)萃取,收集有机相,用饱和食盐水(15mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到粗品。粗品经制备HPLC(流动相:乙腈/水;酸性体系:0.04%HCl)分离纯化得到化合物WX017。MS-ESI m/z:871.3[M+H]
+.
1H NMR(400MHz,CD
3OD)δ:8.27(d,J=9.6Hz,1H),8.22-8.13(m,3H),7.98(d,J=8.4Hz,1H),7.84(d,J=9.2Hz,1H),7.77-7.73(m,2H),7.36(t,J=9.6Hz,1H),7.31(dd,J=1.6,11.2Hz,1H),7.23(d,J=8.4Hz,1H),4.99(dd,J=5.2,10.4Hz,1H),4.58(t,J=4.0Hz,2H),4.18-4.12(m,2H),3.80-3.73(m,4H),3.63-3.55(m,3H),2.92-2.78(m,2H),2.77-2.60(m,2H),2.56-2.47(m,1H),1.57(s,6H),1.45-1.35(m,1H),1.34-1.23(m,1H).
实施例18
合成路线:
室温和氮气保护下,将化合物BB-25的盐酸盐(33mg,82.32μmol)溶于N,N-二甲基甲酰胺(1mL)中,加入化合物BB-26(38.52mg,82.32μmol),乙酸钾(24.24mg,246.97μmol)和冰乙酸(0.1mL),反应混合物室温下搅拌反应0.5小时,再加入三乙酰氧基硼氢化钠(52.34mg,246.97μmol),反应混合物继续搅拌1小时。反应完成后,减压浓缩除去溶剂,向残余物中加入N,N-二甲基甲酰胺(1mL)和盐酸(1N,1mL),过滤。滤液经制备HPLC分离(流动相:乙腈/水;酸性体系:0.04%HCl),得到化合物WX018的盐酸盐。MS-ESI m/z:816.3[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.15(s,1H),10.06(br s,1H),8.60(br d,J=8.4Hz,1H),8.47(br d,J=9.6Hz,1H),8.07-7.92(m,2H),7.85(br d,J=8.0Hz,2H),7.71(br t,J=6.8Hz,1H),7.47-7.32(m,3H),7.13(br d,J=8.0Hz,1H),5.11-5.01(m,1H),4.66-4.41(m,4H),3.94-3.82(m,4H),3.74-3.71(m,2H),3.14-3.03(m,4H),2.92-2.78(m,3H),2.75-2.60(m,4H),2.16-2.06(m,2H),2.02-1.83(m,4H),1.73-1.43(m,4H),1.37-1.24(m,2H).
实施例19
合成路线:
室温和氮气保护下,将化合物BB-27(99mg,169.55μmol)溶于N,N-二甲基甲酰胺(3mL)中,加入化合物BB-25的盐酸盐(67.97mg,169.55μmol),乙酸钾(49.92mg,508.66μmol)和冰乙酸(0.1mL),反应混合物室温下搅拌反应0.5小时。再加入三乙酰氧基硼氢化钠(107.81mg,508.66μmol),继续搅拌反应1小时。反应完成后,减压浓缩除去溶剂,向残余物中加入N,N-二甲基甲酰胺(1mL)和盐酸(1N,1mL),过滤。滤液经制备HPLC分离(流动相:乙腈/水;酸性体系:0.04%盐酸)得到化合物WX019的盐酸盐。MS-ESI m/z:814.4[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.14(s,1H),10.95(s,1H),8.57(d,J=8.4Hz,1H),8.47(d,J=9.6Hz,1H),8.01(d,J=7.6Hz,1H),7.95(d,J=9.2Hz,1H),7.90-7.83(m,2H),7.70(t,J=8.0Hz,1H),7.38(d,J=2.4Hz,1H),7.35(d,J=7.6Hz,1H),7.13(dd,J=2.4,8.8Hz,1H),7.06(d,J=9.6Hz,1H),5.06(dd,J=4.6,11.4Hz,1H),4.59-4.48(m,1H),4.00-3.90(m,2H),3.90-3.79(m,1H),3.76-3.66(m,2H),3.64-3.57(m,2H),3.36-3.20(m,6H),2.93-2.79(m,1H),2.70-2.56(m,3H),2.41-2.30(m,1H),2.16-2.01(m,5H),1.95-1.85(m,2H),1.71-1.57(m,2H),1.57-1.44(m,2H),1.14(s,1H).
实施例20
合成路线:
室温和氮气保护下,将BB-28(0.07g,149.27μmol)溶于N,N-二甲基甲酰胺(3mL)中,随后加入中间体BB-25的盐酸盐(59.84mg,149.27μmol),乙酸钾(43.95mg,447.82μmol)和醋酸(105.00mg,1.75mmol,0.1mL),反应混合物室温下搅拌反应0.5小时,再加入三乙酰氧基硼氢化钠(94.91mg,447.82μmol),反应混合物继续搅拌反应12小时。反应完毕后,减压浓缩除去溶剂。向残余物中加入N,N-二甲基甲酰胺(1mL)和盐酸(1N,1mL),过滤,滤液经制备HPLC(流动相:乙腈/水;酸性体系:0.04%HCl)分离纯化,得到化合物WX020的盐酸盐。MS-ESI m/z:817.3[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.14(s,1H),10.31(br s,1H),8.73(s,1H),8.69(d,J=8.0Hz,1H),8.48(d,J=9.2Hz,1H),8.05-7.99(m,1H),7.96(d,J=9.2Hz,1H),7.86(d,J=8.8Hz,1H),7.71(t,J=8.0Hz,1H),7.39(d,J=2.4Hz,1H),7.36(d,J=7.6Hz,1H),7.14(dd,J=2.4,8.8Hz,1H),5.06(br dd,J=4.6,11.4Hz,1H),4.79(br s,2H),4.60-4.46(m,1H),3.95-3.81(m,1H),3.77-3.65(m,2H),3.55-3.44(m,4H),3.23-3.13(m,4H),2.93-2.78(m,1H),2.70-2.52(m,4H),2.44-2.34(m,2H),2.17-2.07(m,2H),2.06-1.97(m,2H),1.94-1.83(m,2H),1.75-1.59(m,2H),1.58-1.47(m,2H),1.39-1.25(m,2H).
实施例21
合成路线:
将化合物BB-29(119.75mg,255.90μmol)和BB-25的盐酸盐(102.58mg,255.90μmol)加到二氯甲烷(5mL)中,开启搅拌,将三乙酰基硼氢化钠(81.35mg,383.85μmol)和醋酸钠(41.98mg,511.80μmol)慢慢加入到反应中,反应液用氮气置换3次后在25℃搅拌2小时。反应液慢慢加入水中(50mL),用二氯甲烷萃取(50mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩。粗品经板层析纯化(展开剂: 乙酸乙酯)得到化合物WX021。MS-ESI m/z:816.6[M+H]
+.
1H NMR(400MHz,DMSO_d
6)δ:11.14(s,1H),8.59(d,J=0.8Hz,1H),8.44(d,J=9.6Hz,1H),8.25(s,1H),8.07(d,J=8.4Hz,1H),7.96-7.88(m,2H),7.85(d,J=8.8Hz,1H),7.65(t,J=7.8Hz,1H),7.37(d,J=2.4Hz,1H),7.30(d,J=7.6Hz,1H),7.12(dd,J=2.4,8.8Hz,1H),5.03(br dd,J=5.0,11.4Hz,1H),4.65-4.35(m,3H),3.91-3.73(m,1H),3.16-2.94(m,6H),2.90-2.76(m,2H),2.75-2.52(m,5H),2.42-2.21(m,3H),2.15-2.02(m,2H),1.98-1.79(m,5H),1.66-1.43(m,4H),1.20-1.10(m,2H).
生物测试
测试例1:在人前列腺癌LNCaP细胞中的抗增殖作用评估实验目的
本实验通过检测受试化合物在人前列腺癌LNCaP细胞中对细胞增殖的抑制作用。
实验材料:
1.细胞系及培养方法(见表1):
表1
细胞系 | 肿瘤类型 | 生长特点 | 培养方法 |
LNCaP | 人前列腺癌 | 贴壁 | 无酚红1640+10%FBS |
2.培养基及试剂(见表2):
表2
培养基及试剂 | 生产商 | 货号 |
无酚红RPMI 1640 | GIBCO | 11835030 |
Dulbecco′s PBS | CORNING | 21-031-CVC |
FBS | ExCell Bio | FSP500 |
Penicillin-Streptomycin Solution | HyClone | SV30010 |
0.25%Trypsin | GIBCO | 25200072 |
3.多孔板
4.细胞活性实验所用试剂及仪器
(1)Promega CellTiter-Glo发光法细胞活性检测试剂盒(Promega-G7573)。
实验方案
1.细胞培养
将肿瘤细胞系按上述培养条件在37℃,5%CO
2的培养箱中进行培养。定期传代,取处于对数生长期的细胞用于铺板。
2.细胞铺板
(1).用台盼兰进行细胞染色并计数活细胞;
(2).将细胞浓度调整至合适浓度(见表3);
表3
Cell line | Density(per 96-well) |
LNCaP | 4500 |
(3).按上表所示在培养板中每孔加入90μL细胞悬液,在空白对照空中加入不含细胞的培养液;
(4).将培养板在37℃,5%CO
2,及100%相对湿度的培养箱中培养过夜。
3.化合物存储板制备
制备化合物起始浓度400倍浓度的母液存储板:将化合物用DMSO从最高浓度梯度稀释至最低浓度。每次现用现配。
化合物起始浓度10倍浓度的工作液的配制及化合物处理细胞
(1).在V形底的96孔板中加入78μL细胞培养液,从化合物起始浓度400倍浓度的母液存储板中吸取2μL化合物加入96孔板的细胞培养液中。在溶媒对照和空白对照中加入2μL DMSO。加入化合物或DMSO后用排枪吹打混匀。
(2).加药:取10μL的化合物起始浓度10倍浓度的工作液加入到细胞培养板中。在溶媒对照和空白对照中加入10μL DMSO-细胞培养液混合液。
(3).将96孔细胞板放回培养箱中培养6天。
4.CellTiter-Glo发光法细胞活性检测
以下步骤按照Promega CellTiter-Glo发光法细胞活性检测试剂盒(Promega-G7573)的说明书来进行。
(1).将CellTiter-Glo缓冲液融化并放置至室温;
(2).将CellTiter-Glo底物放置至室温;
(3).在一瓶CellTiter-Glo底物中加入100mL CellTiter-Glo缓冲液以溶解底物,从而配制CellTiter-Glo工作液;
(4).缓慢涡旋震荡使充分溶解;
(5).取出细胞培养板放置30分钟使其平衡至室温;
(6).在每孔中加入50μL(等于每孔中细胞培养液一半体积)的CellTiter-Glo工作液。用铝箔纸包裹细 胞板以避光;
(7).将培养板在轨道摇床上振摇2分钟以诱导细胞裂解;
(8).培养板在室温放置10分钟以稳定发光信号;
(9).在2104 EnVision读板器上检测发光信号。
5.数据分析
用下列公式来计算检测化合物的抑制率(Inhibition rate,IR):IR(%)=(RLU溶媒对照-RLU化合物)/(RLU溶媒对照-RLU空白对照)*100%.在Excel中计算不同浓度化合物的抑制率,然后用GraphPad Prism软件作抑制曲线图和计算相关参数,包括最小抑制率,最大抑制率及IC
50。
实验结果:
测试结果见表4。
表4本发明化合物在LNCaP细胞系中的细胞增殖抑制作用
化合物 | LNCaP IC 50(nM) |
WX008 | 117 |
WX009 | 125 |
WX020盐酸盐 | 89 |
WX021 | 130 |
结论:本发明化合物在人前列腺癌细胞LNCaP中展现出优异细胞增殖的抑制作用。
测试例2:在人前列腺癌VCaP细胞中的抗增殖作用评估
实验目的
本实验通过检测受试化合物在人前列腺癌VCaP细胞中对细胞增殖的抑制作用。
实验材料:
1.细胞系及培养方法见表5.
表5
细胞系 | 肿瘤类型 | 生长特点 | 培养条件 |
VCaP | 人前列腺癌 | 贴壁 | DMEM+10%FBS |
2.培养基及试剂见表6.
表6
培养基及试剂 | 生产商 | 货号 |
DMEM | GIBCO | 11995-065 |
Dulbecco′s PBS | Thermo | SH30028.02B |
FBS | ExCell Bio | FSP500 |
Penicillin-Streptomycin Solution | HyClone | SV30010 |
0.25%Trypsin | GIBCO | 25200072 |
3.多孔板
4.细胞活性实验所用试剂及仪器
(1)Promega CellTiter-Glo发光法细胞活性检测试剂盒(Promega-G7573)。
实验方案
1.细胞培养
将肿瘤细胞系按上述培养条件在37℃,5%CO
2的培养箱中进行培养。定期传代,取处于对数生长期的细胞用于铺板。
2.细胞铺板
(1).用台盼兰进行细胞染色并计数活细胞;
(2).将细胞浓度调整至合适浓度;
(3).在培养板中每孔加入90μL细胞悬液,在空白对照空中加入不含细胞的培养液;
(4).将培养板在37℃,5%CO
2,及100%相对湿度的培养箱中培养过夜。
3.化合物存储板制备
制备化合物起始浓度400倍浓度的母液存储板:将化合物用DMSO从最高浓度梯度稀释至最低浓度。每次现用现配。
化合物起始浓度10倍浓度的工作液的配制及化合物处理细胞
(1).在V形底的96孔板中加入78μL细胞培养液,从化合物起始浓度400倍浓度的母液存储板中吸取2μL化合物加入96孔板的细胞培养液中。在溶媒对照和空白对照中加入2μL DMSO。加入化合物或DMSO后用排枪吹打混匀。
(2).加药:取10μL的化合物起始浓度10倍浓度的工作液加入到细胞培养板中。在溶媒对照和空白对照中加入10μL DMSO-细胞培养液混合液。
(3).将96孔细胞板放回培养箱中培养6天。
4.CellTiter-Glo发光法细胞活性检测
以下步骤按照Promega CellTiter-Glo发光法细胞活性检测试剂盒(Promega-G7573)的说明书来进行。
(1).将CellTiter-Glo缓冲液融化并放置至室温;
(2).将CellTiter-Glo底物放置至室温;
(3).在一瓶CellTiter-Glo底物中加入100mL CellTiter-Glo缓冲液以溶解底物,从而配制CellTiter-Glo工作液;
(4).缓慢涡旋震荡使充分溶解;
(5).取出细胞培养板放置30分钟使其平衡至室温;
(6).在每孔中加入50μL(等于每孔中细胞培养液一半体积)的CellTiter-Glo工作液。用铝箔纸包裹细胞板以避光;
(7).将培养板在轨道摇床上振摇2分钟以诱导细胞裂解;
(8).培养板在室温放置10分钟以稳定发光信号;
(9).在2104 EnVision读板器上检测发光信号。
5.数据分析
用下列公式来计算检测化合物的抑制率(Inhibition rate,IR):IR(%)=(RLU溶媒对照-RLU化合物)/(RLU溶媒对照-RLU空白对照)*100%.在Excel中计算不同浓度化合物的抑制率,然后用GraphPad Prism软件作抑制曲线图和计算相关参数,包括最小抑制率,最大抑制率及IC
50。
实验结果:
表7本发明化合物在VCaP细胞系中的细胞增殖抑制作用
化合物 | VCaP IC 50(nM) |
WX008 | 81 |
结论:本发明化合物在人前列腺癌细胞VCaP中展现出优异细胞增殖的抑制作用。
测试例3:人前列腺癌LNCaP细胞皮下异种移植肿瘤CB-17 SCID模型的体内药效学研究
细胞培养
人前列腺癌LNcap细胞(ECACC-89110211)体外单层培养,培养条件为RPMI-1640培养基中加10%胎牛血清,100U/mL青霉素和100μg/mL链霉素,37℃5%CO
2孵箱培养。一周两次用胰酶-EDTA进行常规消化处理传代。当细胞饱和度为80%-90%,数量到达要求时,收取细胞,计数,接种。
实验动物
CB-17 SCID小鼠,6-8周龄,雄性,体重18-22克。
实验方案
将0.2mL(1×10
7个)LNcap细胞(加基质胶,体积比为1∶1)皮下接种于每只小鼠的右后背,肿瘤平均体积达到约80mm
3时对小鼠进行去势手术,166mm
3时开始分组给药。
表8实验动物分组及给药方案
注:
1.N:每组小鼠数目
2.给药容积:根据小鼠体重10μL/g。如果体重下降超过15%,给药方案应做出相应调整。
3.BID时间间隔为8hr
肿瘤测量和实验指标
每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:V=0.5a×b
2,a和b分别表示肿瘤的长径和短径。
化合物的抑瘤疗效用TGI(%)评价。TGI(%),反映肿瘤生长抑制率。TGI(%)=[(1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积))/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)]×100%。
实验结果
测试结果见表9。
表9化合物对LNCaP异种移植瘤模型的抑瘤效果
结论:本发明化合物在人前列腺癌LNCaP异种移植瘤模型上展示了的显著的抑瘤作用。
测试例4:人前列腺癌LNCaP细胞的AR蛋白水平的调控的体外测试
实验目的:用ICW方法测试化合物在LNCap细胞中对雄激素受体AR的降解作用
实验方法:
第一天
1.将细胞培养基、0.025%胰蛋白酶和磷酸盐缓冲液于37℃水浴锅中预热。
2.胰蛋白酶消化的细胞用1000转离心5分钟,弃上清后用新鲜培养基重悬细胞。
3.将细胞按4k/36μL/孔的密度接种于COL-Coated CellCarrier-384板中。
4.接种细胞的384细胞板放在超净台中静置10分钟。
5.将细胞板放置二氧化碳细胞培养箱中培养过夜。
第二天
1.将10mM浓度的化合物在进行倍比稀释前稀释到3mM(3μL+7μL二甲基亚砜)。
2.用Bravo仪器将化合物在Echo板中进行3倍系列稀释(4μL+8μL)。
3.设置ZPE和HPE对照孔,ZPE对照孔为二甲基亚砜。
4.将Echo板用1000转离心1分钟后,用Echo将稀释好的化合物从Echo板转移至中间板中,200nL/孔。
5.用Multidrop combi将细胞培养基加入中间板中,20μL/孔。
6.将中间板放置于水平震荡器上震荡1分钟,使化合物与培养基充分混合。
7.用Bravo将中间稀释板中的化合物转移至细胞板中,4μL/孔。
8.将细胞板放置于水平震荡器上震荡1分钟,800转离心30秒。
9.将细胞板放回二氧化碳培养箱中继续培养24小时。
第三天
1.冰箱中取出4%的多聚甲醛放至室温。
2.将细胞板从培养箱中取出。
3.细胞板中直接加入4%多聚甲醛,40μL/孔。
4.室温孵育20分钟
5.手动甩去细胞板中的培养基,倒置于离心机中,800转离心7秒,充分去除残留的培养基。
6.细胞板加入4%的多聚甲醛,25μL/孔。
7.室温孵育30分钟。
8.手动甩去细胞板中的多聚甲醛溶液,倒置于离心机中,800转离心7秒。
9.加入0.1%Trition,25μL/孔。
10.室温孵育10分钟。
11.手动甩去细胞板中0.1%的Trition溶液,倒置于离心机中,800转离心7秒。
13.室温孵育1小时。
15.加入一抗溶液,20μL/孔。细胞板置于离心机中,800转离心7秒。
16.将细胞板封膜后放置4℃孵育过夜。
第四天
1.手动甩去细胞板中的一抗溶液,倒置于离心机中,800转离心7秒。
2.110μL PBS洗3次。
3.细胞板倒置于离心机中,800转离心7秒。
4.加入二抗和DAPI溶液,20μL/孔。细胞板置于离心机中,800转离心7秒。
5.室温孵育1小时。
6.手动甩去细胞板中的二抗和DAPI溶液,倒置于离心机中,800转离心7秒。
7.用110μL PBS洗3次后,细胞板中残留30μL PBS
8.细胞板置于离心机中,800转离心7秒。
9.Operetta仪器扫描细胞板
实验结果
测试结果见表10。
表10本发明化合物在LNCaP细胞系中对AR蛋白降解作用
# | 化合物编号 | DC 50(nM) | D max |
1 | WX018的盐酸盐 | 42.0 | 106.15 |
2 | WX019的盐酸盐 | 9.5 | 80.77 |
3 | WX020的盐酸盐 | 7.94 | 110.84 |
4 | WX021 | 13.4 | 107.74 |
结论:本发明化合物对AR蛋白展现出良好的降解活性。
测试例5:化合物的小鼠药代动力学评价
实验目的:
本研究受试化合物选用CB-17 SCID雄性小鼠,应用LCMS/MS法定量测定小鼠静脉注射或口服给予测试化合物和参比化合物不同时间点的血浆中的药物浓度,以评价受试药物在小鼠的药代动力学特征。
实验材料:
CB-17 SCID小鼠(雄性,20-30g,7-10周龄,北京维通利华或上海斯莱克)。
实验操作:
将测试化合物的澄清溶液或混悬溶液经尾静脉注射到小鼠体内(不禁食),或灌胃给予到小鼠(不禁食)。静脉注射给药于0h(给药前)和给药后0.083,0.25,0.5,1,2,4,8,24h从颈静脉穿刺采血,置于添加了EDTA-K2的抗凝管中,在4℃,将混合物充分涡旋混合并在13000rpm下离心10分钟;口服灌胃给药于0h(给药前)和给药后0.25,0.5,1,2,4,8,24h从颈静脉穿刺采血,置于添加了EDTA-K2的抗凝管中,将混合物充分涡旋混合并在13000rpm下离心10分钟。采用LC-MS/MS法测定血药浓度,使用WinNonlinTMVersion 6.3(Pharsight,MountainView,CA)药代学软件,以非房室型线性对数梯形法计算相关药代动力学参数。
实验结果:测试结果见表11。
表11.本发明化合物在小鼠中的药代动力学参数
结论:本发明化合物在小鼠体内展示良好的成药性。
测试例6:化合物的大鼠药代动力学评价
实验目的:
本研究受试化合物选用SD雄性大鼠,应用LCMS/MS法定量测定大鼠静脉注射或口服给予测试化合物和参比化合物不同时间点的血浆中的药物浓度,以评价受试药物在大鼠的药代动力学特征。
实验材料:
SD大鼠(雄性,北京维通利华)。
实验操作:
将测试化合物的澄清溶液或混悬溶液经尾静脉注射到大鼠体内(不禁食),或灌胃给予到大鼠(不禁食)。静脉注射给药于0h(给药前)和给药后0.083h,0.25h,0.5h,1h,2h,4h,6h,8h,24h从颈静脉穿刺采血,置于添加了肝素钠的抗凝管中,血液样本采集后置于冰上,并于1小时之内离心分离血浆(离心条件:6000g,3分钟,2-8℃);口服灌胃给药于0h(给药前)和给药后0.25h,0.5h,1h,2h,4h,6h,8h,24h从颈静脉穿刺采血,置于添加了肝素钠的抗凝管中,血液样本采集后置于冰上,并于1小时之内离心分离血浆(离心条件:6000g,3分钟,2-8℃)。采用LC-MS/MS法测定血药浓度,使用 WinNonlinTMVersion 8.2.0(Pharsight,Mountain View,CA)药代学软件,以非房室型线性对数梯形法计算相关药代动力学参数。
实验结果:测试结果见表12。
表12.本发明化合物在大鼠中的药代动力学参数
结论:本发明化合物在大鼠体内展示良好的成药性。
Claims (11)
- 式(II)所示化合物或其药学上可接受的盐,其中,R 1选自H和CH 3;R 2选自H和甲基;E 1选自单键、O、NH和-C(=O)NH-;E 4选自单键、O和-C(=O)NH-;E 5选自5-6元杂环烷基;T 1和T 2各自独立地选自CH和N;m选自1、2和3;n选自0、1、2、3、4、5、6和7;p和q各自独立地选自0、1和2;环A选自苯基和5-6元杂芳基;ABM选自靶向结合于AR蛋白的药物或其衍生物。
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202280043489.3A CN117580575A (zh) | 2021-06-17 | 2022-06-15 | 戊二酰亚胺取代的异噁唑稠环化合物及其应用 |
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202110674190.5 | 2021-06-17 | ||
CN202110674190 | 2021-06-17 | ||
CN202111222437.6 | 2021-10-20 | ||
CN202111222437 | 2021-10-20 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2022262782A1 true WO2022262782A1 (zh) | 2022-12-22 |
Family
ID=84526867
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2022/099015 WO2022262782A1 (zh) | 2021-06-17 | 2022-06-15 | 戊二酰亚胺取代的异噁唑稠环化合物及其应用 |
Country Status (2)
Country | Link |
---|---|
CN (1) | CN117580575A (zh) |
WO (1) | WO2022262782A1 (zh) |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2024037616A1 (zh) * | 2022-08-19 | 2024-02-22 | 正大天晴药业集团股份有限公司 | 包含环己基的化合物 |
WO2024146617A1 (zh) * | 2023-01-04 | 2024-07-11 | 甘李药业股份有限公司 | 用于雄激素受体靶向降解的化合物和方法 |
WO2024199146A1 (zh) * | 2023-03-24 | 2024-10-03 | 漳州片仔癀药业股份有限公司 | 萘并异噁唑类化合物的制备方法 |
Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN107428734A (zh) * | 2015-01-20 | 2017-12-01 | 阿尔维纳斯股份有限公司 | 用于雄激素受体的靶向降解的化合物和方法 |
CN109641874A (zh) * | 2016-05-10 | 2019-04-16 | C4医药公司 | 用于靶蛋白降解的c3-碳连接的戊二酰亚胺降解决定子体 |
CN110317192A (zh) * | 2018-03-28 | 2019-10-11 | 上海美志医药科技有限公司 | 一类具有降解雄激素受体活性的化合物 |
CN110746399A (zh) * | 2018-07-23 | 2020-02-04 | 上海美志医药科技有限公司 | 具有降解雄激素受体活性的化合物 |
WO2020048548A1 (zh) * | 2018-09-07 | 2020-03-12 | 正大天晴药业集团股份有限公司 | 一种作用于crbn蛋白的三并环类化合物 |
WO2021185291A1 (zh) * | 2020-03-17 | 2021-09-23 | 南京明德新药研发有限公司 | 蛋白降解调节剂与其使用方法 |
WO2021262812A1 (en) * | 2020-06-24 | 2021-12-30 | Celgene Corporation | Cereblon binding compounds, compositions thereof, and methods of treatment therewith |
-
2022
- 2022-06-15 CN CN202280043489.3A patent/CN117580575A/zh active Pending
- 2022-06-15 WO PCT/CN2022/099015 patent/WO2022262782A1/zh active Application Filing
Patent Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN107428734A (zh) * | 2015-01-20 | 2017-12-01 | 阿尔维纳斯股份有限公司 | 用于雄激素受体的靶向降解的化合物和方法 |
CN109641874A (zh) * | 2016-05-10 | 2019-04-16 | C4医药公司 | 用于靶蛋白降解的c3-碳连接的戊二酰亚胺降解决定子体 |
CN110317192A (zh) * | 2018-03-28 | 2019-10-11 | 上海美志医药科技有限公司 | 一类具有降解雄激素受体活性的化合物 |
CN110746399A (zh) * | 2018-07-23 | 2020-02-04 | 上海美志医药科技有限公司 | 具有降解雄激素受体活性的化合物 |
WO2020048548A1 (zh) * | 2018-09-07 | 2020-03-12 | 正大天晴药业集团股份有限公司 | 一种作用于crbn蛋白的三并环类化合物 |
WO2021185291A1 (zh) * | 2020-03-17 | 2021-09-23 | 南京明德新药研发有限公司 | 蛋白降解调节剂与其使用方法 |
WO2021262812A1 (en) * | 2020-06-24 | 2021-12-30 | Celgene Corporation | Cereblon binding compounds, compositions thereof, and methods of treatment therewith |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2024037616A1 (zh) * | 2022-08-19 | 2024-02-22 | 正大天晴药业集团股份有限公司 | 包含环己基的化合物 |
WO2024146617A1 (zh) * | 2023-01-04 | 2024-07-11 | 甘李药业股份有限公司 | 用于雄激素受体靶向降解的化合物和方法 |
WO2024199146A1 (zh) * | 2023-03-24 | 2024-10-03 | 漳州片仔癀药业股份有限公司 | 萘并异噁唑类化合物的制备方法 |
Also Published As
Publication number | Publication date |
---|---|
CN117580575A (zh) | 2024-02-20 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2021147967A1 (zh) | 作为kras抑制剂的大环类化合物 | |
WO2021180181A1 (zh) | 嘧啶并杂环类化合物及其应用 | |
WO2022262782A1 (zh) | 戊二酰亚胺取代的异噁唑稠环化合物及其应用 | |
JP2023517393A (ja) | タンパク質分解調整剤およびその使用方法 | |
WO2022171147A1 (zh) | 嘧啶并芳香环类化合物 | |
WO2020048548A1 (zh) | 一种作用于crbn蛋白的三并环类化合物 | |
WO2020239076A1 (zh) | 作为甲状腺素受体-β激动剂的哒嗪酮类衍生物及其应用 | |
WO2021259331A1 (zh) | 八元含n杂环类化合物 | |
WO2021129653A1 (zh) | 一种蛋白降解剂化合物的制备方法和应用 | |
WO2020048547A1 (zh) | 三环并呋喃取代哌啶二酮类化合物 | |
WO2022161443A1 (zh) | 嘧啶并吡喃类化合物 | |
WO2022194221A1 (zh) | 呋喃稠环取代的戊二酰亚胺类化合物 | |
WO2021047674A1 (zh) | 作为crbn蛋白调节剂的双并环类化合物 | |
WO2023138662A9 (zh) | 苯并嘧啶类化合物及其应用 | |
WO2023217045A1 (zh) | 选择性抑制parp1的氟代喹喔啉酮衍生物 | |
WO2023001028A1 (zh) | 杂芳-3-哌啶二酮类化合物及其应用 | |
WO2022063308A1 (zh) | 一类1,7-萘啶类化合物及其应用 | |
WO2023001069A1 (zh) | 大环酰胺类化合物及其应用 | |
WO2023036175A1 (zh) | 戊二酰亚胺类化合物与其应用 | |
WO2023241618A1 (zh) | 氨基嘧啶类化合物及其应用 | |
WO2023208127A1 (zh) | 杂芳基取代的双环化合物及其应用 | |
WO2023246914A1 (zh) | 杂环取代的嘧啶并吡喃类化合物及其应用 | |
WO2024012456A1 (zh) | 哌嗪桥取代的杂环并嘧啶类化合物 | |
JP2022521673A (ja) | Pd-l1免疫調整剤であるフルオロビニルベンズアミド化合物 | |
TWI814092B (zh) | 稠合的三并環衍生物及其在藥學上的應用 |
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: 22824257 Country of ref document: EP Kind code of ref document: A1 |
|
WWE | Wipo information: entry into national phase |
Ref document number: 202280043489.3 Country of ref document: CN |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 22824257 Country of ref document: EP Kind code of ref document: A1 |