CN115381823A - Application of cyclin-dependent kinase 9 inhibitor - Google Patents
Application of cyclin-dependent kinase 9 inhibitor Download PDFInfo
- Publication number
- CN115381823A CN115381823A CN202210564415.6A CN202210564415A CN115381823A CN 115381823 A CN115381823 A CN 115381823A CN 202210564415 A CN202210564415 A CN 202210564415A CN 115381823 A CN115381823 A CN 115381823A
- Authority
- CN
- China
- Prior art keywords
- substituted
- unsubstituted
- independently selected
- group
- dichloromethane
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Granted
Links
- 229940124077 Cyclin-dependent kinase 9 inhibitor Drugs 0.000 title abstract description 3
- 150000001875 compounds Chemical class 0.000 claims abstract description 124
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 75
- 150000003839 salts Chemical class 0.000 claims abstract description 61
- 229940002612 prodrug Drugs 0.000 claims abstract description 56
- 239000000651 prodrug Substances 0.000 claims abstract description 56
- 201000007270 liver cancer Diseases 0.000 claims abstract description 36
- 208000014018 liver neoplasm Diseases 0.000 claims abstract description 36
- 206010006187 Breast cancer Diseases 0.000 claims abstract description 30
- 208000026310 Breast neoplasm Diseases 0.000 claims abstract description 30
- 206010060862 Prostate cancer Diseases 0.000 claims abstract description 28
- 208000000236 Prostatic Neoplasms Diseases 0.000 claims abstract description 28
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 22
- -1 Alkoxy radical Chemical class 0.000 claims description 126
- 239000003814 drug Substances 0.000 claims description 61
- 229910052757 nitrogen Inorganic materials 0.000 claims description 51
- 125000000592 heterocycloalkyl group Chemical group 0.000 claims description 48
- 125000001424 substituent group Chemical group 0.000 claims description 40
- 125000004432 carbon atom Chemical group C* 0.000 claims description 35
- 229940079593 drug Drugs 0.000 claims description 33
- 125000003118 aryl group Chemical group 0.000 claims description 28
- 125000000217 alkyl group Chemical group 0.000 claims description 27
- 125000000753 cycloalkyl group Chemical group 0.000 claims description 24
- 125000003545 alkoxy group Chemical group 0.000 claims description 23
- 239000000203 mixture Substances 0.000 claims description 23
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 21
- 125000006413 ring segment Chemical group 0.000 claims description 21
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 claims description 20
- 125000004433 nitrogen atom Chemical group N* 0.000 claims description 20
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical group C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 claims description 18
- 229910052799 carbon Inorganic materials 0.000 claims description 16
- 206010073071 hepatocellular carcinoma Diseases 0.000 claims description 16
- 229910052731 fluorine Inorganic materials 0.000 claims description 15
- 125000001072 heteroaryl group Chemical group 0.000 claims description 15
- 238000006467 substitution reaction Methods 0.000 claims description 15
- 101100383153 Caenorhabditis elegans cdk-9 gene Proteins 0.000 claims description 14
- 208000003721 Triple Negative Breast Neoplasms Diseases 0.000 claims description 12
- 231100000844 hepatocellular carcinoma Toxicity 0.000 claims description 12
- 125000005842 heteroatom Chemical group 0.000 claims description 12
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 claims description 12
- 208000022679 triple-negative breast carcinoma Diseases 0.000 claims description 12
- 210000004185 liver Anatomy 0.000 claims description 11
- 125000005913 (C3-C6) cycloalkyl group Chemical group 0.000 claims description 10
- 125000001313 C5-C10 heteroaryl group Chemical group 0.000 claims description 10
- 201000011510 cancer Diseases 0.000 claims description 10
- 125000004076 pyridyl group Chemical group 0.000 claims description 10
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 claims description 9
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 claims description 9
- 125000004438 haloalkoxy group Chemical group 0.000 claims description 9
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 claims description 9
- 125000003566 oxetanyl group Chemical group 0.000 claims description 9
- 125000003226 pyrazolyl group Chemical group 0.000 claims description 9
- HSFWRNGVRCDJHI-UHFFFAOYSA-N alpha-acetylene Natural products C#C HSFWRNGVRCDJHI-UHFFFAOYSA-N 0.000 claims description 8
- 210000000481 breast Anatomy 0.000 claims description 8
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 claims description 8
- 125000002534 ethynyl group Chemical group [H]C#C* 0.000 claims description 8
- 125000004368 propenyl group Chemical group C(=CC)* 0.000 claims description 8
- 125000000391 vinyl group Chemical group [H]C([*])=C([H])[H] 0.000 claims description 8
- 125000006583 (C1-C3) haloalkyl group Chemical group 0.000 claims description 7
- 125000000113 cyclohexyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 claims description 7
- 125000001153 fluoro group Chemical group F* 0.000 claims description 7
- 238000009472 formulation Methods 0.000 claims description 7
- 125000000842 isoxazolyl group Chemical group 0.000 claims description 7
- 229910052760 oxygen Inorganic materials 0.000 claims description 7
- 229910052717 sulfur Inorganic materials 0.000 claims description 7
- 125000000171 (C1-C6) haloalkyl group Chemical group 0.000 claims description 6
- IANQTJSKSUMEQM-UHFFFAOYSA-N 1-benzofuran Chemical group C1=CC=C2OC=CC2=C1 IANQTJSKSUMEQM-UHFFFAOYSA-N 0.000 claims description 6
- XWIYUCRMWCHYJR-UHFFFAOYSA-N 1h-pyrrolo[3,2-b]pyridine Chemical group C1=CC=C2NC=CC2=N1 XWIYUCRMWCHYJR-UHFFFAOYSA-N 0.000 claims description 6
- 150000001336 alkenes Chemical class 0.000 claims description 6
- 150000001345 alkine derivatives Chemical class 0.000 claims description 6
- 125000001041 indolyl group Chemical group 0.000 claims description 6
- 125000002393 azetidinyl group Chemical group 0.000 claims description 5
- 125000001559 cyclopropyl group Chemical group [H]C1([H])C([H])([H])C1([H])* 0.000 claims description 5
- 125000000335 thiazolyl group Chemical group 0.000 claims description 5
- 125000001544 thienyl group Chemical group 0.000 claims description 5
- 125000004737 (C1-C6) haloalkoxy group Chemical group 0.000 claims description 4
- 125000006590 (C2-C6) alkenylene group Chemical group 0.000 claims description 4
- 239000004480 active ingredient Substances 0.000 claims description 4
- 125000004419 alkynylene group Chemical group 0.000 claims description 4
- 125000001797 benzyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])* 0.000 claims description 4
- 239000003937 drug carrier Substances 0.000 claims description 4
- 125000004193 piperazinyl group Chemical group 0.000 claims description 4
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 claims description 4
- IQUPABOKLQSFBK-UHFFFAOYSA-N 2-nitrophenol Chemical group OC1=CC=CC=C1[N+]([O-])=O IQUPABOKLQSFBK-UHFFFAOYSA-N 0.000 claims description 3
- 239000002246 antineoplastic agent Substances 0.000 claims description 3
- 125000004069 aziridinyl group Chemical group 0.000 claims description 3
- 125000001188 haloalkyl group Chemical group 0.000 claims description 3
- 125000003453 indazolyl group Chemical group N1N=C(C2=C1C=CC=C2)* 0.000 claims description 3
- 239000007972 injectable composition Substances 0.000 claims description 3
- 125000002971 oxazolyl group Chemical group 0.000 claims description 3
- 230000008685 targeting Effects 0.000 claims description 3
- 125000003554 tetrahydropyrrolyl group Chemical group 0.000 claims description 3
- 125000006570 (C5-C6) heteroaryl group Chemical group 0.000 claims description 2
- 125000000094 2-phenylethyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])C([H])([H])* 0.000 claims description 2
- 125000000882 C2-C6 alkenyl group Chemical group 0.000 claims description 2
- 125000003601 C2-C6 alkynyl group Chemical group 0.000 claims description 2
- 125000000041 C6-C10 aryl group Chemical group 0.000 claims description 2
- 125000000484 butyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 claims description 2
- 229910052801 chlorine Inorganic materials 0.000 claims description 2
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 claims description 2
- 125000002757 morpholinyl group Chemical group 0.000 claims description 2
- 125000001147 pentyl group Chemical group C(CCCC)* 0.000 claims description 2
- 125000002568 propynyl group Chemical group [*]C#CC([H])([H])[H] 0.000 claims description 2
- RWRDLPDLKQPQOW-UHFFFAOYSA-N Pyrrolidine Chemical compound C1CCNC1 RWRDLPDLKQPQOW-UHFFFAOYSA-N 0.000 claims 3
- XDTMQSROBMDMFD-UHFFFAOYSA-N Cyclohexane Chemical compound C1CCCCC1 XDTMQSROBMDMFD-UHFFFAOYSA-N 0.000 claims 2
- RGSFGYAAUTVSQA-UHFFFAOYSA-N Cyclopentane Chemical compound C1CCCC1 RGSFGYAAUTVSQA-UHFFFAOYSA-N 0.000 claims 2
- 125000005330 8 membered heterocyclic group Chemical group 0.000 claims 1
- 150000001413 amino acids Chemical class 0.000 claims 1
- 229940044683 chemotherapy drug Drugs 0.000 claims 1
- DMEGYFMYUHOHGS-UHFFFAOYSA-N heptamethylene Natural products C1CCCCCC1 DMEGYFMYUHOHGS-UHFFFAOYSA-N 0.000 claims 1
- 125000000896 monocarboxylic acid group Chemical group 0.000 claims 1
- 125000005936 piperidyl group Chemical group 0.000 claims 1
- 238000000034 method Methods 0.000 abstract description 8
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 676
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 417
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 292
- 238000006243 chemical reaction Methods 0.000 description 187
- 230000015572 biosynthetic process Effects 0.000 description 181
- 238000003786 synthesis reaction Methods 0.000 description 181
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-Dimethylformamide Chemical compound CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 172
- 239000007858 starting material Substances 0.000 description 165
- 239000002904 solvent Substances 0.000 description 159
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 146
- 238000004440 column chromatography Methods 0.000 description 144
- 239000012074 organic phase Substances 0.000 description 143
- 239000000243 solution Substances 0.000 description 139
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 135
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 131
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical class [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 113
- 239000012467 final product Substances 0.000 description 100
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-Diisopropylethylamine (DIPEA) Chemical compound CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 0.000 description 98
- WFDIJRYMOXRFFG-UHFFFAOYSA-N Acetic anhydride Chemical compound CC(=O)OC(C)=O WFDIJRYMOXRFFG-UHFFFAOYSA-N 0.000 description 84
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical group N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 69
- DTQVDTLACAAQTR-UHFFFAOYSA-N Trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 68
- 210000004027 cell Anatomy 0.000 description 59
- 239000003208 petroleum Substances 0.000 description 58
- BWHMMNNQKKPAPP-UHFFFAOYSA-L potassium carbonate Chemical compound [K+].[K+].[O-]C([O-])=O BWHMMNNQKKPAPP-UHFFFAOYSA-L 0.000 description 54
- HIXDQWDOVZUNNA-UHFFFAOYSA-N 2-(3,4-dimethoxyphenyl)-5-hydroxy-7-methoxychromen-4-one Chemical compound C=1C(OC)=CC(O)=C(C(C=2)=O)C=1OC=2C1=CC=C(OC)C(OC)=C1 HIXDQWDOVZUNNA-UHFFFAOYSA-N 0.000 description 52
- RTZKZFJDLAIYFH-UHFFFAOYSA-N ether Substances CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 52
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 50
- 238000012360 testing method Methods 0.000 description 50
- 239000012043 crude product Substances 0.000 description 41
- 238000000746 purification Methods 0.000 description 40
- 230000005764 inhibitory process Effects 0.000 description 39
- CDBYLPFSWZWCQE-UHFFFAOYSA-L Sodium Carbonate Chemical compound [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 34
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 34
- 239000007795 chemical reaction product Substances 0.000 description 31
- 102100024457 Cyclin-dependent kinase 9 Human genes 0.000 description 29
- XTHFKEDIFFGKHM-UHFFFAOYSA-N Dimethoxyethane Chemical compound COCCOC XTHFKEDIFFGKHM-UHFFFAOYSA-N 0.000 description 29
- 238000012544 monitoring process Methods 0.000 description 28
- 101000980930 Homo sapiens Cyclin-dependent kinase 9 Proteins 0.000 description 27
- 229910000027 potassium carbonate Inorganic materials 0.000 description 27
- 238000000926 separation method Methods 0.000 description 25
- PSLUFJFHTBIXMW-WYEYVKMPSA-N [(3r,4ar,5s,6s,6as,10s,10ar,10bs)-3-ethenyl-10,10b-dihydroxy-3,4a,7,7,10a-pentamethyl-1-oxo-6-(2-pyridin-2-ylethylcarbamoyloxy)-5,6,6a,8,9,10-hexahydro-2h-benzo[f]chromen-5-yl] acetate Chemical compound O([C@@H]1[C@@H]([C@]2(O[C@](C)(CC(=O)[C@]2(O)[C@@]2(C)[C@@H](O)CCC(C)(C)[C@@H]21)C=C)C)OC(=O)C)C(=O)NCCC1=CC=CC=N1 PSLUFJFHTBIXMW-WYEYVKMPSA-N 0.000 description 24
- NXQGGXCHGDYOHB-UHFFFAOYSA-L cyclopenta-1,4-dien-1-yl(diphenyl)phosphane;dichloropalladium;iron(2+) Chemical compound [Fe+2].Cl[Pd]Cl.[CH-]1C=CC(P(C=2C=CC=CC=2)C=2C=CC=CC=2)=C1.[CH-]1C=CC(P(C=2C=CC=CC=2)C=2C=CC=CC=2)=C1 NXQGGXCHGDYOHB-UHFFFAOYSA-L 0.000 description 23
- 239000011541 reaction mixture Substances 0.000 description 23
- JSGHMGKJNZTKGF-DTWKUNHWSA-N (1s,3r)-3-[(2-methylpropan-2-yl)oxycarbonylamino]cyclohexane-1-carboxylic acid Chemical compound CC(C)(C)OC(=O)N[C@@H]1CCC[C@H](C(O)=O)C1 JSGHMGKJNZTKGF-DTWKUNHWSA-N 0.000 description 22
- 238000000605 extraction Methods 0.000 description 22
- VLKZOEOYAKHREP-UHFFFAOYSA-N n-Hexane Chemical compound CCCCCC VLKZOEOYAKHREP-UHFFFAOYSA-N 0.000 description 22
- IHKOBZNEWCGRPD-UHFFFAOYSA-N 5-fluoro-4-iodopyridin-2-amine Chemical compound FC=1C(=CC(=NC=1)N)I IHKOBZNEWCGRPD-UHFFFAOYSA-N 0.000 description 21
- 229920006395 saturated elastomer Polymers 0.000 description 21
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 19
- 238000001727 in vivo Methods 0.000 description 19
- 238000001035 drying Methods 0.000 description 18
- 238000005406 washing Methods 0.000 description 18
- 230000000694 effects Effects 0.000 description 16
- 230000002401 inhibitory effect Effects 0.000 description 16
- 239000005457 ice water Substances 0.000 description 15
- 229910001873 dinitrogen Inorganic materials 0.000 description 14
- 238000000338 in vitro Methods 0.000 description 14
- SCVFZCLFOSHCOH-UHFFFAOYSA-M potassium acetate Chemical compound [K+].CC([O-])=O SCVFZCLFOSHCOH-UHFFFAOYSA-M 0.000 description 14
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 14
- 235000017557 sodium bicarbonate Nutrition 0.000 description 14
- 229910000029 sodium carbonate Inorganic materials 0.000 description 14
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 13
- 241001465754 Metazoa Species 0.000 description 13
- 238000001514 detection method Methods 0.000 description 13
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 12
- IAZDPXIOMUYVGZ-WFGJKAKNSA-N Dimethyl sulfoxide Chemical compound [2H]C([2H])([2H])S(=O)C([2H])([2H])[2H] IAZDPXIOMUYVGZ-WFGJKAKNSA-N 0.000 description 12
- 239000012298 atmosphere Substances 0.000 description 12
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 12
- 238000010992 reflux Methods 0.000 description 12
- FYSNRJHAOHDILO-UHFFFAOYSA-N thionyl chloride Chemical compound ClS(Cl)=O FYSNRJHAOHDILO-UHFFFAOYSA-N 0.000 description 12
- 239000008346 aqueous phase Substances 0.000 description 11
- 239000007864 aqueous solution Substances 0.000 description 11
- 125000000623 heterocyclic group Chemical group 0.000 description 11
- 239000003112 inhibitor Substances 0.000 description 11
- 239000000463 material Substances 0.000 description 11
- 108091007914 CDKs Proteins 0.000 description 10
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 10
- WOSKHXYHFSIKNG-UHFFFAOYSA-N lenvatinib Chemical compound C=12C=C(C(N)=O)C(OC)=CC2=NC=CC=1OC(C=C1Cl)=CC=C1NC(=O)NC1CC1 WOSKHXYHFSIKNG-UHFFFAOYSA-N 0.000 description 10
- 229960003784 lenvatinib Drugs 0.000 description 10
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 9
- UIIMBOGNXHQVGW-DEQYMQKBSA-M Sodium bicarbonate-14C Chemical compound [Na+].O[14C]([O-])=O UIIMBOGNXHQVGW-DEQYMQKBSA-M 0.000 description 9
- 238000001816 cooling Methods 0.000 description 9
- 201000010099 disease Diseases 0.000 description 9
- 238000010438 heat treatment Methods 0.000 description 9
- 230000003287 optical effect Effects 0.000 description 9
- 238000002360 preparation method Methods 0.000 description 9
- FVAUCKIRQBBSSJ-UHFFFAOYSA-M sodium iodide Chemical compound [Na+].[I-] FVAUCKIRQBBSSJ-UHFFFAOYSA-M 0.000 description 9
- 102000003903 Cyclin-dependent kinases Human genes 0.000 description 8
- 108090000266 Cyclin-dependent kinases Proteins 0.000 description 8
- 241000699666 Mus <mouse, genus> Species 0.000 description 8
- KDLHZDBZIXYQEI-UHFFFAOYSA-N Palladium Chemical compound [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 description 8
- 238000007920 subcutaneous administration Methods 0.000 description 8
- 241000699670 Mus sp. Species 0.000 description 7
- 239000006285 cell suspension Substances 0.000 description 7
- 238000004587 chromatography analysis Methods 0.000 description 7
- 230000002018 overexpression Effects 0.000 description 7
- 235000011056 potassium acetate Nutrition 0.000 description 7
- 230000035897 transcription Effects 0.000 description 7
- 238000013518 transcription Methods 0.000 description 7
- 210000004881 tumor cell Anatomy 0.000 description 7
- 230000004614 tumor growth Effects 0.000 description 7
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 6
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 6
- 150000001555 benzenes Chemical group 0.000 description 6
- 239000012964 benzotriazole Substances 0.000 description 6
- 235000019441 ethanol Nutrition 0.000 description 6
- 239000012091 fetal bovine serum Substances 0.000 description 6
- 230000002062 proliferating effect Effects 0.000 description 6
- 238000003756 stirring Methods 0.000 description 6
- 238000005303 weighing Methods 0.000 description 6
- YZCUMZWULWOUMD-UHFFFAOYSA-N 5-fluoro-4-(4-fluoro-2-methoxyphenyl)-n-[4-[(methylsulfonimidoyl)methyl]pyridin-2-yl]pyridin-2-amine Chemical compound COC1=CC(F)=CC=C1C1=CC(NC=2N=CC=C(CS(C)(=N)=O)C=2)=NC=C1F YZCUMZWULWOUMD-UHFFFAOYSA-N 0.000 description 5
- 102100032857 Cyclin-dependent kinase 1 Human genes 0.000 description 5
- 101710106279 Cyclin-dependent kinase 1 Proteins 0.000 description 5
- 108091000080 Phosphotransferase Proteins 0.000 description 5
- 230000022131 cell cycle Effects 0.000 description 5
- SBZXBUIDTXKZTM-UHFFFAOYSA-N diglyme Chemical compound COCCOCCOC SBZXBUIDTXKZTM-UHFFFAOYSA-N 0.000 description 5
- 229940126364 enitociclib Drugs 0.000 description 5
- 238000011156 evaluation Methods 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 230000003902 lesion Effects 0.000 description 5
- 102000020233 phosphotransferase Human genes 0.000 description 5
- 239000000047 product Substances 0.000 description 5
- 239000002994 raw material Substances 0.000 description 5
- 239000000758 substrate Substances 0.000 description 5
- YLQBMQCUIZJEEH-UHFFFAOYSA-N tetrahydrofuran Natural products C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 description 5
- VHYFNPMBLIVWCW-UHFFFAOYSA-N 4-Dimethylaminopyridine Chemical compound CN(C)C1=CC=NC=C1 VHYFNPMBLIVWCW-UHFFFAOYSA-N 0.000 description 4
- 238000011729 BALB/c nude mouse Methods 0.000 description 4
- AEMRFAOFKBGASW-UHFFFAOYSA-N Glycolic acid Chemical compound OCC(O)=O AEMRFAOFKBGASW-UHFFFAOYSA-N 0.000 description 4
- 101000979909 Homo sapiens NMDA receptor synaptonuclear signaling and neuronal migration factor Proteins 0.000 description 4
- 102100024546 NMDA receptor synaptonuclear signaling and neuronal migration factor Human genes 0.000 description 4
- 108010012271 Positive Transcriptional Elongation Factor B Proteins 0.000 description 4
- 102000019014 Positive Transcriptional Elongation Factor B Human genes 0.000 description 4
- 125000004429 atom Chemical group 0.000 description 4
- 230000000903 blocking effect Effects 0.000 description 4
- 238000004364 calculation method Methods 0.000 description 4
- 239000004359 castor oil Substances 0.000 description 4
- 235000019438 castor oil Nutrition 0.000 description 4
- 238000004113 cell culture Methods 0.000 description 4
- 230000000857 drug effect Effects 0.000 description 4
- 238000003810 ethyl acetate extraction Methods 0.000 description 4
- ZEMPKEQAKRGZGQ-XOQCFJPHSA-N glycerol triricinoleate Natural products CCCCCC[C@@H](O)CC=CCCCCCCCC(=O)OC[C@@H](COC(=O)CCCCCCCC=CC[C@@H](O)CCCCCC)OC(=O)CCCCCCCC=CC[C@H](O)CCCCCC ZEMPKEQAKRGZGQ-XOQCFJPHSA-N 0.000 description 4
- 239000001963 growth medium Substances 0.000 description 4
- 125000004435 hydrogen atom Chemical group [H]* 0.000 description 4
- 238000002156 mixing Methods 0.000 description 4
- 239000000546 pharmaceutical excipient Substances 0.000 description 4
- 208000023958 prostate neoplasm Diseases 0.000 description 4
- UMJSCPRVCHMLSP-UHFFFAOYSA-N pyridine Natural products COC1=CC=CN=C1 UMJSCPRVCHMLSP-UHFFFAOYSA-N 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- DYHSDKLCOJIUFX-UHFFFAOYSA-N tert-butoxycarbonyl anhydride Chemical compound CC(C)(C)OC(=O)OC(=O)OC(C)(C)C DYHSDKLCOJIUFX-UHFFFAOYSA-N 0.000 description 4
- 238000010998 test method Methods 0.000 description 4
- 230000001225 therapeutic effect Effects 0.000 description 4
- 238000009210 therapy by ultrasound Methods 0.000 description 4
- 230000005029 transcription elongation Effects 0.000 description 4
- 230000002100 tumorsuppressive effect Effects 0.000 description 4
- 229910021642 ultra pure water Inorganic materials 0.000 description 4
- 239000012498 ultrapure water Substances 0.000 description 4
- UYDZUCNMZXCLJI-UHFFFAOYSA-N 4-bromo-3-methoxyphenol Chemical compound COC1=CC(O)=CC=C1Br UYDZUCNMZXCLJI-UHFFFAOYSA-N 0.000 description 3
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 3
- QGJOPFRUJISHPQ-UHFFFAOYSA-N Carbon disulfide Chemical compound S=C=S QGJOPFRUJISHPQ-UHFFFAOYSA-N 0.000 description 3
- 206010048610 Cardiotoxicity Diseases 0.000 description 3
- 108050006400 Cyclin Proteins 0.000 description 3
- 102000016736 Cyclin Human genes 0.000 description 3
- 108010025454 Cyclin-Dependent Kinase 5 Proteins 0.000 description 3
- 102100026810 Cyclin-dependent kinase 7 Human genes 0.000 description 3
- 102100026805 Cyclin-dependent-like kinase 5 Human genes 0.000 description 3
- 101000911952 Homo sapiens Cyclin-dependent kinase 7 Proteins 0.000 description 3
- 101000600434 Homo sapiens Putative uncharacterized protein encoded by MIR7-3HG Proteins 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 206010027476 Metastases Diseases 0.000 description 3
- 102100037401 Putative uncharacterized protein encoded by MIR7-3HG Human genes 0.000 description 3
- 102000009572 RNA Polymerase II Human genes 0.000 description 3
- 108010009460 RNA Polymerase II Proteins 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 239000002671 adjuvant Substances 0.000 description 3
- 150000001408 amides Chemical class 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 230000004071 biological effect Effects 0.000 description 3
- 239000011203 carbon fibre reinforced carbon Substances 0.000 description 3
- 231100000259 cardiotoxicity Toxicity 0.000 description 3
- 230000004663 cell proliferation Effects 0.000 description 3
- 238000001516 cell proliferation assay Methods 0.000 description 3
- SNRCKKQHDUIRIY-UHFFFAOYSA-L cyclopenta-1,4-dien-1-yl(diphenyl)phosphane;dichloromethane;dichloropalladium;iron(2+) Chemical compound [Fe+2].ClCCl.Cl[Pd]Cl.C1=C[CH-]C(P(C=2C=CC=CC=2)C=2C=CC=CC=2)=C1.C1=C[CH-]C(P(C=2C=CC=CC=2)C=2C=CC=CC=2)=C1 SNRCKKQHDUIRIY-UHFFFAOYSA-L 0.000 description 3
- 230000034994 death Effects 0.000 description 3
- 231100000517 death Toxicity 0.000 description 3
- 238000011161 development Methods 0.000 description 3
- 230000018109 developmental process Effects 0.000 description 3
- 238000007865 diluting Methods 0.000 description 3
- 238000006073 displacement reaction Methods 0.000 description 3
- 238000006911 enzymatic reaction Methods 0.000 description 3
- 150000002148 esters Chemical class 0.000 description 3
- 230000006870 function Effects 0.000 description 3
- 238000011081 inoculation Methods 0.000 description 3
- 238000011835 investigation Methods 0.000 description 3
- 238000004519 manufacturing process Methods 0.000 description 3
- DAZXVJBJRMWXJP-UHFFFAOYSA-N n,n-dimethylethylamine Chemical compound CCN(C)C DAZXVJBJRMWXJP-UHFFFAOYSA-N 0.000 description 3
- 230000035407 negative regulation of cell proliferation Effects 0.000 description 3
- 239000012299 nitrogen atmosphere Substances 0.000 description 3
- 125000003386 piperidinyl group Chemical group 0.000 description 3
- 239000013641 positive control Substances 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 210000002307 prostate Anatomy 0.000 description 3
- 125000002098 pyridazinyl group Chemical group 0.000 description 3
- 125000000714 pyrimidinyl group Chemical group 0.000 description 3
- 235000009518 sodium iodide Nutrition 0.000 description 3
- 239000012321 sodium triacetoxyborohydride Substances 0.000 description 3
- UCPYLLCMEDAXFR-UHFFFAOYSA-N triphosgene Chemical compound ClC(Cl)(Cl)OC(=O)OC(Cl)(Cl)Cl UCPYLLCMEDAXFR-UHFFFAOYSA-N 0.000 description 3
- KPZYAGQLBFUTMA-UHFFFAOYSA-K tripotassium;phosphate;trihydrate Chemical compound O.O.O.[K+].[K+].[K+].[O-]P([O-])([O-])=O KPZYAGQLBFUTMA-UHFFFAOYSA-K 0.000 description 3
- 239000012224 working solution Substances 0.000 description 3
- RNJQBGXOSAQQDG-JGVFFNPUSA-N (1s,3r)-3-[(2-methylpropan-2-yl)oxycarbonylamino]cyclopentane-1-carboxylic acid Chemical compound CC(C)(C)OC(=O)N[C@@H]1CC[C@H](C(O)=O)C1 RNJQBGXOSAQQDG-JGVFFNPUSA-N 0.000 description 2
- MPDDTAJMJCESGV-CTUHWIOQSA-M (3r,5r)-7-[2-(4-fluorophenyl)-5-[methyl-[(1r)-1-phenylethyl]carbamoyl]-4-propan-2-ylpyrazol-3-yl]-3,5-dihydroxyheptanoate Chemical compound C1([C@@H](C)N(C)C(=O)C2=NN(C(CC[C@@H](O)C[C@@H](O)CC([O-])=O)=C2C(C)C)C=2C=CC(F)=CC=2)=CC=CC=C1 MPDDTAJMJCESGV-CTUHWIOQSA-M 0.000 description 2
- ADJBXDCXYMCCAD-UHFFFAOYSA-N (4-fluoro-2-methoxyphenyl)boronic acid Chemical compound COC1=CC(F)=CC=C1B(O)O ADJBXDCXYMCCAD-UHFFFAOYSA-N 0.000 description 2
- 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 2
- YZUPZGFPHUVJKC-UHFFFAOYSA-N 1-bromo-2-methoxyethane Chemical compound COCCBr YZUPZGFPHUVJKC-UHFFFAOYSA-N 0.000 description 2
- KSJJMSKNZVXAND-UHFFFAOYSA-N 1-cyanocyclopropane-1-carboxylic acid Chemical compound OC(=O)C1(C#N)CC1 KSJJMSKNZVXAND-UHFFFAOYSA-N 0.000 description 2
- MCTWTZJPVLRJOU-UHFFFAOYSA-N 1-methyl-1H-imidazole Chemical compound CN1C=CN=C1 MCTWTZJPVLRJOU-UHFFFAOYSA-N 0.000 description 2
- GQHTUMJGOHRCHB-UHFFFAOYSA-N 2,3,4,6,7,8,9,10-octahydropyrimido[1,2-a]azepine Chemical compound C1CCCCN2CCCN=C21 GQHTUMJGOHRCHB-UHFFFAOYSA-N 0.000 description 2
- BWLBGMIXKSTLSX-UHFFFAOYSA-N 2-hydroxyisobutyric acid Chemical compound CC(C)(O)C(O)=O BWLBGMIXKSTLSX-UHFFFAOYSA-N 0.000 description 2
- XZXUUIMQZYMXJT-UHFFFAOYSA-N 3-cyano-1-[(2-methylpropan-2-yl)oxycarbonyl]azetidine-3-carboxylic acid Chemical compound CC(C)(C)OC(=O)N1CC(C(O)=O)(C#N)C1 XZXUUIMQZYMXJT-UHFFFAOYSA-N 0.000 description 2
- ALRHLSYJTWAHJZ-UHFFFAOYSA-N 3-hydroxypropionic acid Chemical compound OCCC(O)=O ALRHLSYJTWAHJZ-UHFFFAOYSA-N 0.000 description 2
- QGZKDVFQNNGYKY-UHFFFAOYSA-N Ammonia Chemical compound N QGZKDVFQNNGYKY-UHFFFAOYSA-N 0.000 description 2
- BTBUEUYNUDRHOZ-UHFFFAOYSA-N Borate Chemical compound [O-]B([O-])[O-] BTBUEUYNUDRHOZ-UHFFFAOYSA-N 0.000 description 2
- 229940126074 CDK kinase inhibitor Drugs 0.000 description 2
- 108010024986 Cyclin-Dependent Kinase 2 Proteins 0.000 description 2
- 108010025464 Cyclin-Dependent Kinase 4 Proteins 0.000 description 2
- 108010025468 Cyclin-Dependent Kinase 6 Proteins 0.000 description 2
- 108010025461 Cyclin-Dependent Kinase 9 Proteins 0.000 description 2
- 102100036239 Cyclin-dependent kinase 2 Human genes 0.000 description 2
- 102100036252 Cyclin-dependent kinase 4 Human genes 0.000 description 2
- 102100026804 Cyclin-dependent kinase 6 Human genes 0.000 description 2
- 102100034770 Cyclin-dependent kinase inhibitor 3 Human genes 0.000 description 2
- OKKJLVBELUTLKV-MZCSYVLQSA-N Deuterated methanol Chemical compound [2H]OC([2H])([2H])[2H] OKKJLVBELUTLKV-MZCSYVLQSA-N 0.000 description 2
- YZGQDNOIGFBYKF-UHFFFAOYSA-N Ethoxyacetic acid Chemical compound CCOCC(O)=O YZGQDNOIGFBYKF-UHFFFAOYSA-N 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 101000945639 Homo sapiens Cyclin-dependent kinase inhibitor 3 Proteins 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 2
- JVTAAEKCZFNVCJ-REOHCLBHSA-N L-lactic acid Chemical compound C[C@H](O)C(O)=O JVTAAEKCZFNVCJ-REOHCLBHSA-N 0.000 description 2
- 239000012359 Methanesulfonyl chloride Substances 0.000 description 2
- BAVYZALUXZFZLV-UHFFFAOYSA-N Methylamine Chemical compound NC BAVYZALUXZFZLV-UHFFFAOYSA-N 0.000 description 2
- FFDGPVCHZBVARC-UHFFFAOYSA-N N,N-dimethylglycine Chemical compound CN(C)CC(O)=O FFDGPVCHZBVARC-UHFFFAOYSA-N 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 150000001412 amines Chemical class 0.000 description 2
- 230000003698 anagen phase Effects 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 210000001099 axilla Anatomy 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- AGEZXYOZHKGVCM-UHFFFAOYSA-N benzyl bromide Chemical compound BrCC1=CC=CC=C1 AGEZXYOZHKGVCM-UHFFFAOYSA-N 0.000 description 2
- 125000002619 bicyclic group Chemical group 0.000 description 2
- ILAHWRKJUDSMFH-UHFFFAOYSA-N boron tribromide Chemical compound BrB(Br)Br ILAHWRKJUDSMFH-UHFFFAOYSA-N 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- FJDQFPXHSGXQBY-UHFFFAOYSA-L caesium carbonate Chemical compound [Cs+].[Cs+].[O-]C([O-])=O FJDQFPXHSGXQBY-UHFFFAOYSA-L 0.000 description 2
- 229910000024 caesium carbonate Inorganic materials 0.000 description 2
- 230000009702 cancer cell proliferation Effects 0.000 description 2
- CREMABGTGYGIQB-UHFFFAOYSA-N carbon carbon Chemical group C.C CREMABGTGYGIQB-UHFFFAOYSA-N 0.000 description 2
- 230000010261 cell growth Effects 0.000 description 2
- 239000013078 crystal Substances 0.000 description 2
- MLIREBYILWEBDM-UHFFFAOYSA-N cyanoacetic acid Chemical compound OC(=O)CC#N MLIREBYILWEBDM-UHFFFAOYSA-N 0.000 description 2
- 239000002875 cyclin dependent kinase inhibitor Substances 0.000 description 2
- 229940043378 cyclin-dependent kinase inhibitor Drugs 0.000 description 2
- 229940127089 cytotoxic agent Drugs 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 208000035475 disorder Diseases 0.000 description 2
- 239000000706 filtrate Substances 0.000 description 2
- 238000001914 filtration Methods 0.000 description 2
- 210000003194 forelimb Anatomy 0.000 description 2
- 239000011521 glass Substances 0.000 description 2
- 229910052736 halogen Inorganic materials 0.000 description 2
- 125000005843 halogen group Chemical group 0.000 description 2
- 150000002367 halogens Chemical class 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 230000036210 malignancy Effects 0.000 description 2
- 108010082117 matrigel Proteins 0.000 description 2
- 108020004999 messenger RNA Proteins 0.000 description 2
- 206010061289 metastatic neoplasm Diseases 0.000 description 2
- QARBMVPHQWIHKH-UHFFFAOYSA-N methanesulfonyl chloride Chemical compound CS(Cl)(=O)=O QARBMVPHQWIHKH-UHFFFAOYSA-N 0.000 description 2
- WSFSSNUMVMOOMR-NJFSPNSNSA-N methanone Chemical compound O=[14CH2] WSFSSNUMVMOOMR-NJFSPNSNSA-N 0.000 description 2
- 239000011259 mixed solution Substances 0.000 description 2
- 125000002950 monocyclic group Chemical group 0.000 description 2
- 125000001971 neopentyl group Chemical group [H]C([*])([H])C(C([H])([H])[H])(C([H])([H])[H])C([H])([H])[H] 0.000 description 2
- 238000011275 oncology therapy Methods 0.000 description 2
- 229910052763 palladium Inorganic materials 0.000 description 2
- PIBWKRNGBLPSSY-UHFFFAOYSA-L palladium(II) chloride Chemical compound Cl[Pd]Cl PIBWKRNGBLPSSY-UHFFFAOYSA-L 0.000 description 2
- 206010033675 panniculitis Diseases 0.000 description 2
- 230000003285 pharmacodynamic effect Effects 0.000 description 2
- 230000026731 phosphorylation Effects 0.000 description 2
- 238000006366 phosphorylation reaction Methods 0.000 description 2
- 230000000704 physical effect Effects 0.000 description 2
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 2
- 102000004169 proteins and genes Human genes 0.000 description 2
- 108090000623 proteins and genes Proteins 0.000 description 2
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 2
- 125000000719 pyrrolidinyl group Chemical group 0.000 description 2
- 230000002336 repolarization Effects 0.000 description 2
- 239000004017 serum-free culture medium Substances 0.000 description 2
- 238000010898 silica gel chromatography Methods 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- DAEPDZWVDSPTHF-UHFFFAOYSA-M sodium pyruvate Chemical compound [Na+].CC(=O)C([O-])=O DAEPDZWVDSPTHF-UHFFFAOYSA-M 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 210000004304 subcutaneous tissue Anatomy 0.000 description 2
- 239000006228 supernatant Substances 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 238000010189 synthetic method Methods 0.000 description 2
- 125000003718 tetrahydrofuranyl group Chemical group 0.000 description 2
- 125000003831 tetrazolyl group Chemical group 0.000 description 2
- 125000001113 thiadiazolyl group Chemical group 0.000 description 2
- 125000002053 thietanyl group Chemical group 0.000 description 2
- QAEDZJGFFMLHHQ-UHFFFAOYSA-N trifluoroacetic anhydride Chemical compound FC(F)(F)C(=O)OC(=O)C(F)(F)F QAEDZJGFFMLHHQ-UHFFFAOYSA-N 0.000 description 2
- XIWNHJLMNFUQIW-UHFFFAOYSA-N (1-methylpyrrolo[2,3-b]pyridin-4-yl)boronic acid Chemical compound C1=CN=C2N(C)C=CC2=C1B(O)O XIWNHJLMNFUQIW-UHFFFAOYSA-N 0.000 description 1
- AVIWDYSJSPOOAR-LSDHHAIUSA-N (1S,3R)-3-acetamido-N-[5-chloro-4-(5,5-dimethyl-4,6-dihydropyrrolo[1,2-b]pyrazol-3-yl)pyridin-2-yl]cyclohexane-1-carboxamide Chemical compound C(C)(=O)N[C@H]1C[C@H](CCC1)C(=O)NC1=NC=C(C(=C1)C1=C2N(N=C1)CC(C2)(C)C)Cl AVIWDYSJSPOOAR-LSDHHAIUSA-N 0.000 description 1
- 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 1
- JSGHMGKJNZTKGF-BDAKNGLRSA-N (1r,3s)-3-[(2-methylpropan-2-yl)oxycarbonylamino]cyclohexane-1-carboxylic acid Chemical compound CC(C)(C)OC(=O)N[C@H]1CCC[C@@H](C(O)=O)C1 JSGHMGKJNZTKGF-BDAKNGLRSA-N 0.000 description 1
- ABRTZUPJNVJMHP-UHFFFAOYSA-N (2,3-dichloropyridin-4-yl)boronic acid Chemical compound OB(O)C1=CC=NC(Cl)=C1Cl ABRTZUPJNVJMHP-UHFFFAOYSA-N 0.000 description 1
- LCCZTROJRJFXNV-UHFFFAOYSA-N (2,6-difluoropyridin-3-yl)boronic acid Chemical compound OB(O)C1=CC=C(F)N=C1F LCCZTROJRJFXNV-UHFFFAOYSA-N 0.000 description 1
- LOVFKIWGXGWXLN-UHFFFAOYSA-N (2-ethoxy-5-fluorophenyl)boronic acid Chemical compound CCOC1=CC=C(F)C=C1B(O)O LOVFKIWGXGWXLN-UHFFFAOYSA-N 0.000 description 1
- ROEQGIFOWRQYHD-UHFFFAOYSA-N (2-methoxyphenyl)boronic acid Chemical compound COC1=CC=CC=C1B(O)O ROEQGIFOWRQYHD-UHFFFAOYSA-N 0.000 description 1
- LRWJRIFKJPPAPM-JTQLQIEISA-N (2s)-2-(4-hydroxyphenyl)-2-[(2-methylpropan-2-yl)oxycarbonylamino]acetic acid Chemical compound CC(C)(C)OC(=O)N[C@H](C(O)=O)C1=CC=C(O)C=C1 LRWJRIFKJPPAPM-JTQLQIEISA-N 0.000 description 1
- TWYYFYNJOJGNFP-CUXYNZQBSA-N (2s,4r,5s,6s)-2-[(4s,5r)-4-acetyloxy-5-methyl-3-methylidene-6-phenylhexyl]-2-carbamoyl-4-[[(e,4s,6s)-4,6-dimethyloct-2-enoyl]oxymethyl]-5-hydroxy-1,3-dioxane-4,5,6-tricarboxylic acid Chemical compound O1[C@H](C(O)=O)[C@](C(O)=O)(O)[C@](COC(=O)/C=C/[C@@H](C)C[C@@H](C)CC)(C(O)=O)O[C@]1(C(N)=O)CCC(=C)[C@@H](OC(C)=O)[C@H](C)CC1=CC=CC=C1 TWYYFYNJOJGNFP-CUXYNZQBSA-N 0.000 description 1
- VSALEQTUDXMKST-UHFFFAOYSA-N (3,4-difluoro-2-methoxyphenyl)boronic acid Chemical compound COC1=C(F)C(F)=CC=C1B(O)O VSALEQTUDXMKST-UHFFFAOYSA-N 0.000 description 1
- BMQDAIUNAGXSKR-UHFFFAOYSA-N (3-hydroxy-2,3-dimethylbutan-2-yl)oxyboronic acid Chemical compound CC(C)(O)C(C)(C)OB(O)O BMQDAIUNAGXSKR-UHFFFAOYSA-N 0.000 description 1
- ISQCHQGYKNHYGP-UHFFFAOYSA-N (4,5-difluoro-2-methoxyphenyl)boronic acid Chemical compound COC1=CC(F)=C(F)C=C1B(O)O ISQCHQGYKNHYGP-UHFFFAOYSA-N 0.000 description 1
- IFPQNCKVOQKEIU-UHFFFAOYSA-N (4-fluoro-2-phenylmethoxyphenyl)boronic acid Chemical compound OB(O)C1=CC=C(F)C=C1OCC1=CC=CC=C1 IFPQNCKVOQKEIU-UHFFFAOYSA-N 0.000 description 1
- RSEFJQODCQCGRB-UHFFFAOYSA-N (4-fluoro-2-propan-2-yloxyphenyl)boronic acid Chemical compound CC(C)OC1=CC(F)=CC=C1B(O)O RSEFJQODCQCGRB-UHFFFAOYSA-N 0.000 description 1
- VUDZSIYXZUYWSC-DBRKOABJSA-N (4r)-1-[(2r,4r,5r)-3,3-difluoro-4-hydroxy-5-(hydroxymethyl)oxolan-2-yl]-4-hydroxy-1,3-diazinan-2-one Chemical compound FC1(F)[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)N[C@H](O)CC1 VUDZSIYXZUYWSC-DBRKOABJSA-N 0.000 description 1
- FMBVAOHFMSQDGT-UHFFFAOYSA-N (5-chloro-2-methoxyphenyl)boronic acid Chemical compound COC1=CC=C(Cl)C=C1B(O)O FMBVAOHFMSQDGT-UHFFFAOYSA-N 0.000 description 1
- ITGIYLMMAABTHC-ONEGZZNKSA-N (e)-4-(dimethylazaniumyl)but-2-enoate Chemical compound CN(C)C\C=C\C(O)=O ITGIYLMMAABTHC-ONEGZZNKSA-N 0.000 description 1
- SKCBKBCACWDALV-UHFFFAOYSA-N 1-(trifluoromethyl)cyclopropane-1-carboxylic acid Chemical compound OC(=O)C1(C(F)(F)F)CC1 SKCBKBCACWDALV-UHFFFAOYSA-N 0.000 description 1
- JWOHBPPVVDQMKB-UHFFFAOYSA-N 1-[(2-methylpropan-2-yl)oxycarbonyl]piperidine-4-carboxylic acid Chemical compound CC(C)(C)OC(=O)N1CCC(C(O)=O)CC1 JWOHBPPVVDQMKB-UHFFFAOYSA-N 0.000 description 1
- YCGQPIRMLGEWMW-UHFFFAOYSA-N 1-[1-butyl-4-(3-methoxyphenyl)-2-oxo-1,8-naphthyridin-3-yl]-3-[4-[(dimethylamino)methyl]-2,6-di(propan-2-yl)phenyl]urea;hydrochloride Chemical compound Cl.CC(C)C=1C=C(CN(C)C)C=C(C(C)C)C=1NC(=O)NC=1C(=O)N(CCCC)C2=NC=CC=C2C=1C1=CC=CC(OC)=C1 YCGQPIRMLGEWMW-UHFFFAOYSA-N 0.000 description 1
- FOZVXADQAHVUSV-UHFFFAOYSA-N 1-bromo-2-(2-bromoethoxy)ethane Chemical compound BrCCOCCBr FOZVXADQAHVUSV-UHFFFAOYSA-N 0.000 description 1
- BZXMUSAMMGLTMI-UHFFFAOYSA-N 1-bromo-2-(difluoromethoxy)-4-fluorobenzene Chemical compound FC(F)OC1=CC(F)=CC=C1Br BZXMUSAMMGLTMI-UHFFFAOYSA-N 0.000 description 1
- UMPNPEAXNWBDME-UHFFFAOYSA-N 1-cyanocyclobutane-1-carboxylic acid Chemical compound OC(=O)C1(C#N)CCC1 UMPNPEAXNWBDME-UHFFFAOYSA-N 0.000 description 1
- DTJLXXLXQPOQBZ-UHFFFAOYSA-N 1-ethyl-3-methylpyrazole-4-carboxylic acid Chemical compound CCN1C=C(C(O)=O)C(C)=N1 DTJLXXLXQPOQBZ-UHFFFAOYSA-N 0.000 description 1
- NDCPERCVXDYEFU-UHFFFAOYSA-N 1-fluorocyclopropane-1-carboxylic acid Chemical compound OC(=O)C1(F)CC1 NDCPERCVXDYEFU-UHFFFAOYSA-N 0.000 description 1
- GQXURJDNDYACGE-UHFFFAOYSA-N 1-hydroxycyclopropane-1-carboxylic acid Chemical compound OC(=O)C1(O)CC1 GQXURJDNDYACGE-UHFFFAOYSA-N 0.000 description 1
- DIZKLZKLNKQFGB-UHFFFAOYSA-N 1-methylcyclopropane-1-carboxylic acid Chemical compound OC(=O)C1(C)CC1 DIZKLZKLNKQFGB-UHFFFAOYSA-N 0.000 description 1
- VBEYEZPOEJIIPQ-UHFFFAOYSA-N 1-propan-2-yl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrrolo[2,3-b]pyridine Chemical compound CC(C)N1C=CC2=C1N=CC=C2B1OC(C)(C)C(C)(C)O1 VBEYEZPOEJIIPQ-UHFFFAOYSA-N 0.000 description 1
- BGZZJZIZRARGGZ-UHFFFAOYSA-N 1h-indazol-4-ylboronic acid Chemical compound OB(O)C1=CC=CC2=C1C=NN2 BGZZJZIZRARGGZ-UHFFFAOYSA-N 0.000 description 1
- USJUQEVUEBCLLR-UHFFFAOYSA-N 1h-indol-4-ylboronic acid Chemical compound OB(O)C1=CC=CC2=C1C=CN2 USJUQEVUEBCLLR-UHFFFAOYSA-N 0.000 description 1
- NWQWQKUXRJYXFH-UHFFFAOYSA-N 2,2-Dichloroacetaldehyde Chemical compound ClC(Cl)C=O NWQWQKUXRJYXFH-UHFFFAOYSA-N 0.000 description 1
- JYWKEVKEKOTYEX-UHFFFAOYSA-N 2,6-dibromo-4-chloroiminocyclohexa-2,5-dien-1-one Chemical compound ClN=C1C=C(Br)C(=O)C(Br)=C1 JYWKEVKEKOTYEX-UHFFFAOYSA-N 0.000 description 1
- BGWSMDYVVVJGBB-UHFFFAOYSA-N 2-(diethylamino)acetic acid;hydrochloride Chemical compound Cl.CCN(CC)CC(O)=O BGWSMDYVVVJGBB-UHFFFAOYSA-N 0.000 description 1
- HZAXFHJVJLSVMW-UHFFFAOYSA-N 2-Aminoethan-1-ol Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 description 1
- MSSQOQPKGAMUSY-LEAFIULHSA-N 2-[1-[2-[(4r,6s)-8-chloro-6-(2,3-dimethoxyphenyl)-4,6-dihydropyrrolo[1,2-a][4,1]benzoxazepin-4-yl]acetyl]piperidin-4-yl]acetic acid Chemical compound COC1=CC=CC([C@@H]2C3=CC(Cl)=CC=C3N3C=CC=C3[C@@H](CC(=O)N3CCC(CC(O)=O)CC3)O2)=C1OC MSSQOQPKGAMUSY-LEAFIULHSA-N 0.000 description 1
- PYRKKGOKRMZEIT-UHFFFAOYSA-N 2-[6-(2-cyclopropylethoxy)-9-(2-hydroxy-2-methylpropyl)-1h-phenanthro[9,10-d]imidazol-2-yl]-5-fluorobenzene-1,3-dicarbonitrile Chemical compound C1=C2C3=CC(CC(C)(O)C)=CC=C3C=3NC(C=4C(=CC(F)=CC=4C#N)C#N)=NC=3C2=CC=C1OCCC1CC1 PYRKKGOKRMZEIT-UHFFFAOYSA-N 0.000 description 1
- KMODISUYWZPVGV-UHFFFAOYSA-N 2-bromo-6-methoxypyridine Chemical compound COC1=CC=CC(Br)=N1 KMODISUYWZPVGV-UHFFFAOYSA-N 0.000 description 1
- LDLCZOVUSADOIV-UHFFFAOYSA-N 2-bromoethanol Chemical compound OCCBr LDLCZOVUSADOIV-UHFFFAOYSA-N 0.000 description 1
- KVVDRQDTODKIJD-UHFFFAOYSA-N 2-cyclopropylacetic acid Chemical compound OC(=O)CC1CC1 KVVDRQDTODKIJD-UHFFFAOYSA-N 0.000 description 1
- NEAQRZUHTPSBBM-UHFFFAOYSA-N 2-hydroxy-3,3-dimethyl-7-nitro-4h-isoquinolin-1-one Chemical class C1=C([N+]([O-])=O)C=C2C(=O)N(O)C(C)(C)CC2=C1 NEAQRZUHTPSBBM-UHFFFAOYSA-N 0.000 description 1
- KQDJTBPASNJQFQ-UHFFFAOYSA-N 2-iodophenol Chemical compound OC1=CC=CC=C1I KQDJTBPASNJQFQ-UHFFFAOYSA-N 0.000 description 1
- 125000004493 2-methylbut-1-yl group Chemical group CC(C*)CC 0.000 description 1
- 125000003762 3,4-dimethoxyphenyl group Chemical group [H]C1=C([H])C(OC([H])([H])[H])=C(OC([H])([H])[H])C([H])=C1* 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
- BGAJNPLDJJBRHK-UHFFFAOYSA-N 3-[2-[5-(3-chloro-4-propan-2-yloxyphenyl)-1,3,4-thiadiazol-2-yl]-3-methyl-6,7-dihydro-4h-pyrazolo[4,3-c]pyridin-5-yl]propanoic acid Chemical compound C1=C(Cl)C(OC(C)C)=CC=C1C1=NN=C(N2C(=C3CN(CCC(O)=O)CCC3=N2)C)S1 BGAJNPLDJJBRHK-UHFFFAOYSA-N 0.000 description 1
- FNXYWHTZDAVRTB-UHFFFAOYSA-N 3-methyl-1,2-oxazol-5-amine Chemical compound CC=1C=C(N)ON=1 FNXYWHTZDAVRTB-UHFFFAOYSA-N 0.000 description 1
- 125000003542 3-methylbutan-2-yl group Chemical group [H]C([H])([H])C([H])(*)C([H])(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- XOERUXUEIZCZNL-UHFFFAOYSA-N 4-(bromomethyl)pyridine;hydron;chloride Chemical compound Cl.BrCC1=CC=NC=C1 XOERUXUEIZCZNL-UHFFFAOYSA-N 0.000 description 1
- VIZMARKIEQIMIG-UHFFFAOYSA-N 4-bromo-5-chloropyridin-2-amine Chemical compound NC1=CC(Br)=C(Cl)C=N1 VIZMARKIEQIMIG-UHFFFAOYSA-N 0.000 description 1
- DQAZPZIYEOGZAF-UHFFFAOYSA-N 4-ethyl-n-[4-(3-ethynylanilino)-7-methoxyquinazolin-6-yl]piperazine-1-carboxamide Chemical compound C1CN(CC)CCN1C(=O)NC(C(=CC1=NC=N2)OC)=CC1=C2NC1=CC=CC(C#C)=C1 DQAZPZIYEOGZAF-UHFFFAOYSA-N 0.000 description 1
- YZCUMZWULWOUMD-NDEPHWFRSA-N 5-fluoro-4-(4-fluoro-2-methoxyphenyl)-N-[4-[(methylsulfonimidoyl)methyl]pyridin-2-yl]pyridin-2-amine Chemical compound COc1cc(F)ccc1-c1cc(Nc2cc(C[S@@](C)(=N)=O)ccn2)ncc1F YZCUMZWULWOUMD-NDEPHWFRSA-N 0.000 description 1
- SVVSOIGNROPKMS-UHFFFAOYSA-N 6-bromo-1-methylindazole Chemical compound C1=C(Br)C=C2N(C)N=CC2=C1 SVVSOIGNROPKMS-UHFFFAOYSA-N 0.000 description 1
- RREANTFLPGEWEN-MBLPBCRHSA-N 7-[4-[[(3z)-3-[4-amino-5-[(3,4,5-trimethoxyphenyl)methyl]pyrimidin-2-yl]imino-5-fluoro-2-oxoindol-1-yl]methyl]piperazin-1-yl]-1-cyclopropyl-6-fluoro-4-oxoquinoline-3-carboxylic acid Chemical compound COC1=C(OC)C(OC)=CC(CC=2C(=NC(\N=C/3C4=CC(F)=CC=C4N(CN4CCN(CC4)C=4C(=CC=5C(=O)C(C(O)=O)=CN(C=5C=4)C4CC4)F)C\3=O)=NC=2)N)=C1 RREANTFLPGEWEN-MBLPBCRHSA-N 0.000 description 1
- GPZPQJITKRSVQJ-UHFFFAOYSA-N 7-bromo-1-benzofuran Chemical compound BrC1=CC=CC2=C1OC=C2 GPZPQJITKRSVQJ-UHFFFAOYSA-N 0.000 description 1
- HBAQYPYDRFILMT-UHFFFAOYSA-N 8-[3-(1-cyclopropylpyrazol-4-yl)-1H-pyrazolo[4,3-d]pyrimidin-5-yl]-3-methyl-3,8-diazabicyclo[3.2.1]octan-2-one Chemical class C1(CC1)N1N=CC(=C1)C1=NNC2=C1N=C(N=C2)N1C2C(N(CC1CC2)C)=O HBAQYPYDRFILMT-UHFFFAOYSA-N 0.000 description 1
- MITGKKFYIJJQGL-UHFFFAOYSA-N 9-(4-chlorobenzoyl)-6-methylsulfonyl-2,3-dihydro-1H-carbazol-4-one Chemical compound ClC1=CC=C(C(=O)N2C3=CC=C(C=C3C=3C(CCCC2=3)=O)S(=O)(=O)C)C=C1 MITGKKFYIJJQGL-UHFFFAOYSA-N 0.000 description 1
- 229940125934 AZD4573 Drugs 0.000 description 1
- 208000019738 Abnormality of the breast Diseases 0.000 description 1
- 208000019782 Abnormality of the liver Diseases 0.000 description 1
- QTBSBXVTEAMEQO-UHFFFAOYSA-M Acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 description 1
- AYWSNOYLEWDKRX-UHFFFAOYSA-N C1(CC1)C(C(=O)O)=C=O Chemical compound C1(CC1)C(C(=O)O)=C=O AYWSNOYLEWDKRX-UHFFFAOYSA-N 0.000 description 1
- VGCXGMAHQTYDJK-UHFFFAOYSA-N Chloroacetyl chloride Chemical compound ClCC(Cl)=O VGCXGMAHQTYDJK-UHFFFAOYSA-N 0.000 description 1
- 102000002435 Cyclin T Human genes 0.000 description 1
- 108010068106 Cyclin T Proteins 0.000 description 1
- 102100036876 Cyclin-K Human genes 0.000 description 1
- 102100033245 Cyclin-dependent kinase 16 Human genes 0.000 description 1
- 229930182843 D-Lactic acid Natural products 0.000 description 1
- JVTAAEKCZFNVCJ-UWTATZPHSA-N D-lactic acid Chemical compound C[C@@H](O)C(O)=O JVTAAEKCZFNVCJ-UWTATZPHSA-N 0.000 description 1
- 101710088194 Dehydrogenase Proteins 0.000 description 1
- YIIMEMSDCNDGTB-UHFFFAOYSA-N Dimethylcarbamoyl chloride Chemical compound CN(C)C(Cl)=O YIIMEMSDCNDGTB-UHFFFAOYSA-N 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 101710199605 Endoribonuclease Proteins 0.000 description 1
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000713127 Homo sapiens Cyclin-K Proteins 0.000 description 1
- 101000702364 Homo sapiens Transcription elongation factor SPT5 Proteins 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 102000004310 Ion Channels Human genes 0.000 description 1
- WXNXCEHXYPACJF-SSDOTTSWSA-N N-acetyl-D-leucine Chemical compound CC(C)C[C@H](C(O)=O)NC(C)=O WXNXCEHXYPACJF-SSDOTTSWSA-N 0.000 description 1
- WXNXCEHXYPACJF-ZETCQYMHSA-N N-acetyl-L-leucine Chemical compound CC(C)C[C@@H](C(O)=O)NC(C)=O WXNXCEHXYPACJF-ZETCQYMHSA-N 0.000 description 1
- 102100023057 Neurofilament light polypeptide Human genes 0.000 description 1
- UXRZLDREKITWRO-UHFFFAOYSA-N P(c1ccccc1)c1ccccc1.CC1(C)c2ccccc2Oc2ccccc12 Chemical compound P(c1ccccc1)c1ccccc1.CC1(C)c2ccccc2Oc2ccccc12 UXRZLDREKITWRO-UHFFFAOYSA-N 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- SIOXPEMLGUPBBT-UHFFFAOYSA-N Picolinic acid Natural products OC(=O)C1=CC=CC=N1 SIOXPEMLGUPBBT-UHFFFAOYSA-N 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 102000001253 Protein Kinase Human genes 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 101710113029 Serine/threonine-protein kinase Proteins 0.000 description 1
- 206010042434 Sudden death Diseases 0.000 description 1
- DHXVGJBLRPWPCS-UHFFFAOYSA-N Tetrahydropyran Chemical compound C1CCOCC1 DHXVGJBLRPWPCS-UHFFFAOYSA-N 0.000 description 1
- FZWLAAWBMGSTSO-UHFFFAOYSA-N Thiazole Chemical compound C1=CSC=N1 FZWLAAWBMGSTSO-UHFFFAOYSA-N 0.000 description 1
- 102100030402 Transcription elongation factor SPT5 Human genes 0.000 description 1
- 235000005811 Viola adunca Nutrition 0.000 description 1
- 240000009038 Viola odorata Species 0.000 description 1
- 235000013487 Viola odorata Nutrition 0.000 description 1
- 235000002254 Viola papilionacea Nutrition 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 239000002250 absorbent Substances 0.000 description 1
- 230000002745 absorbent Effects 0.000 description 1
- DHKHKXVYLBGOIT-UHFFFAOYSA-N acetaldehyde Diethyl Acetal Natural products CCOC(C)OCC DHKHKXVYLBGOIT-UHFFFAOYSA-N 0.000 description 1
- 150000001241 acetals Chemical class 0.000 description 1
- WETWJCDKMRHUPV-UHFFFAOYSA-N acetyl chloride Chemical compound CC(Cl)=O WETWJCDKMRHUPV-UHFFFAOYSA-N 0.000 description 1
- 239000012346 acetyl chloride Substances 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- HFBMWMNUJJDEQZ-UHFFFAOYSA-N acryloyl chloride Chemical compound ClC(=O)C=C HFBMWMNUJJDEQZ-UHFFFAOYSA-N 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 230000001464 adherent effect Effects 0.000 description 1
- 239000000853 adhesive Substances 0.000 description 1
- 230000001070 adhesive effect Effects 0.000 description 1
- 125000001931 aliphatic group Chemical group 0.000 description 1
- 125000002947 alkylene group Chemical group 0.000 description 1
- 208000026935 allergic disease Diseases 0.000 description 1
- 150000007854 aminals Chemical class 0.000 description 1
- KRMDCWKBEZIMAB-UHFFFAOYSA-N amitriptyline Chemical compound C1CC2=CC=CC=C2C(=CCCN(C)C)C2=CC=CC=C21 KRMDCWKBEZIMAB-UHFFFAOYSA-N 0.000 description 1
- 229960000836 amitriptyline Drugs 0.000 description 1
- 229910021529 ammonia Inorganic materials 0.000 description 1
- 125000005428 anthryl group Chemical group [H]C1=C([H])C([H])=C2C([H])=C3C(*)=C([H])C([H])=C([H])C3=C([H])C2=C1[H] 0.000 description 1
- 230000000181 anti-adherent effect Effects 0.000 description 1
- 239000003911 antiadherent Substances 0.000 description 1
- 239000002518 antifoaming agent Substances 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 206010003119 arrhythmia Diseases 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 125000003785 benzimidazolyl group Chemical group N1=C(NC2=C1C=CC=C2)* 0.000 description 1
- 125000000499 benzofuranyl group Chemical group O1C(=CC2=C1C=CC=C2)* 0.000 description 1
- 125000004196 benzothienyl group Chemical group S1C(=CC2=C1C=CC=C2)* 0.000 description 1
- 230000008827 biological function Effects 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- AEILLAXRDHDKDY-UHFFFAOYSA-N bromomethylcyclopropane Chemical compound BrCC1CC1 AEILLAXRDHDKDY-UHFFFAOYSA-N 0.000 description 1
- 150000004657 carbamic acid derivatives Chemical class 0.000 description 1
- 150000001721 carbon Chemical group 0.000 description 1
- 150000004649 carbonic acid derivatives Chemical class 0.000 description 1
- 150000001732 carboxylic acid derivatives Chemical class 0.000 description 1
- 230000000747 cardiac effect Effects 0.000 description 1
- 210000004413 cardiac myocyte Anatomy 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 238000013375 chromatographic separation Methods 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 229940125876 compound 15a Drugs 0.000 description 1
- 229940126540 compound 41 Drugs 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- DMSZORWOGDLWGN-UHFFFAOYSA-N ctk1a3526 Chemical compound NP(N)(N)=O DMSZORWOGDLWGN-UHFFFAOYSA-N 0.000 description 1
- YMGUBTXCNDTFJI-UHFFFAOYSA-N cyclopropanecarboxylic acid Chemical compound OC(=O)C1CC1 YMGUBTXCNDTFJI-UHFFFAOYSA-N 0.000 description 1
- 229940022769 d- lactic acid Drugs 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 230000007423 decrease Effects 0.000 description 1
- 230000003111 delayed effect Effects 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 125000005054 dihydropyrrolyl group Chemical group [H]C1=C([H])C([H])([H])C([H])([H])N1* 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 108700003601 dimethylglycine Proteins 0.000 description 1
- 125000000532 dioxanyl group Chemical group 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- 231100000294 dose-dependent toxicity Toxicity 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 230000009982 effect on human Effects 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 150000002081 enamines Chemical class 0.000 description 1
- 239000008393 encapsulating agent Substances 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 239000000686 essence Substances 0.000 description 1
- 125000001033 ether group Chemical group 0.000 description 1
- 210000003722 extracellular fluid Anatomy 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 239000008394 flocculating agent Substances 0.000 description 1
- 239000004088 foaming agent Substances 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 239000008098 formaldehyde solution Substances 0.000 description 1
- 150000003948 formamides Chemical class 0.000 description 1
- 239000003205 fragrance Substances 0.000 description 1
- 125000002541 furyl group Chemical group 0.000 description 1
- 230000014509 gene expression Effects 0.000 description 1
- 238000007429 general method Methods 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 229930182470 glycoside Natural products 0.000 description 1
- 150000002338 glycosides Chemical class 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 150000002373 hemiacetals Chemical class 0.000 description 1
- 150000002374 hemiaminals Chemical class 0.000 description 1
- 210000003494 hepatocyte Anatomy 0.000 description 1
- 239000000833 heterodimer Substances 0.000 description 1
- 239000003906 humectant Substances 0.000 description 1
- 229910052739 hydrogen Inorganic materials 0.000 description 1
- 239000001257 hydrogen Substances 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- 230000003463 hyperproliferative effect Effects 0.000 description 1
- 125000002632 imidazolidinyl group Chemical group 0.000 description 1
- 125000002883 imidazolyl group Chemical group 0.000 description 1
- 150000002466 imines Chemical class 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 230000005917 in vivo anti-tumor Effects 0.000 description 1
- 150000002475 indoles Chemical group 0.000 description 1
- 150000007529 inorganic bases Chemical class 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 229910052740 iodine Inorganic materials 0.000 description 1
- 230000000622 irritating effect Effects 0.000 description 1
- 230000007794 irritation Effects 0.000 description 1
- 125000000959 isobutyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])* 0.000 description 1
- 125000000904 isoindolyl group Chemical group C=1(NC=C2C=CC=CC12)* 0.000 description 1
- 125000001972 isopentyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000002183 isoquinolinyl group Chemical group C1(=NC=CC2=CC=CC=C12)* 0.000 description 1
- 230000031700 light absorption Effects 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 1
- RENRQMCACQEWFC-UGKGYDQZSA-N lnp023 Chemical compound C1([C@H]2N(CC=3C=4C=CNC=4C(C)=CC=3OC)CC[C@@H](C2)OCC)=CC=C(C(O)=O)C=C1 RENRQMCACQEWFC-UGKGYDQZSA-N 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 230000003211 malignant effect Effects 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 239000002609 medium Substances 0.000 description 1
- 230000028161 membrane depolarization Effects 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 230000001394 metastastic effect Effects 0.000 description 1
- ANGDWNBGPBMQHW-UHFFFAOYSA-N methyl cyanoacetate Chemical compound COC(=O)CC#N ANGDWNBGPBMQHW-UHFFFAOYSA-N 0.000 description 1
- 125000006431 methyl cyclopropyl group Chemical group 0.000 description 1
- 239000012982 microporous membrane Substances 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 210000003470 mitochondria Anatomy 0.000 description 1
- 239000012046 mixed solvent Substances 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 125000002911 monocyclic heterocycle group Chemical group 0.000 description 1
- 229940078490 n,n-dimethylglycine Drugs 0.000 description 1
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000003136 n-heptyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001280 n-hexyl group Chemical group C(CCCCC)* 0.000 description 1
- 125000000740 n-pentyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 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
- 108010090677 neurofilament protein L Proteins 0.000 description 1
- 210000002569 neuron Anatomy 0.000 description 1
- 150000002823 nitrates Chemical class 0.000 description 1
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 150000007530 organic bases Chemical class 0.000 description 1
- 230000003204 osmotic effect Effects 0.000 description 1
- 125000001715 oxadiazolyl group Chemical group 0.000 description 1
- 125000000160 oxazolidinyl group Chemical group 0.000 description 1
- UWOTZNQZPLAURK-UHFFFAOYSA-N oxetane-3-carboxylic acid Chemical compound OC(=O)C1COC1 UWOTZNQZPLAURK-UHFFFAOYSA-N 0.000 description 1
- NFHFRUOZVGFOOS-UHFFFAOYSA-N palladium;triphenylphosphane Chemical compound [Pd].C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 NFHFRUOZVGFOOS-UHFFFAOYSA-N 0.000 description 1
- 238000012402 patch clamp technique Methods 0.000 description 1
- 125000003538 pentan-3-yl group Chemical group [H]C([H])([H])C([H])([H])C([H])(*)C([H])([H])C([H])([H])[H] 0.000 description 1
- 230000010412 perfusion Effects 0.000 description 1
- 238000009520 phase I clinical trial Methods 0.000 description 1
- 125000005561 phenanthryl group Chemical group 0.000 description 1
- AHWALFGBDFAJAI-UHFFFAOYSA-N phenyl carbonochloridate Chemical compound ClC(=O)OC1=CC=CC=C1 AHWALFGBDFAJAI-UHFFFAOYSA-N 0.000 description 1
- 235000021317 phosphate Nutrition 0.000 description 1
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 1
- 230000000865 phosphorylative effect Effects 0.000 description 1
- 125000004592 phthalazinyl group Chemical group C1(=NN=CC2=CC=CC=C12)* 0.000 description 1
- 239000004014 plasticizer Substances 0.000 description 1
- 125000003367 polycyclic group Chemical group 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 239000003380 propellant Substances 0.000 description 1
- UORVCLMRJXCDCP-UHFFFAOYSA-N propynoic acid Chemical compound OC(=O)C#C UORVCLMRJXCDCP-UHFFFAOYSA-N 0.000 description 1
- 108060006633 protein kinase Proteins 0.000 description 1
- 125000001042 pteridinyl group Chemical group N1=C(N=CC2=NC=CN=C12)* 0.000 description 1
- 125000000561 purinyl group Chemical group N1=C(N=C2N=CNC2=C1)* 0.000 description 1
- 229950010131 puromycin Drugs 0.000 description 1
- 125000004309 pyranyl group Chemical group O1C(C=CC=C1)* 0.000 description 1
- 125000003373 pyrazinyl group Chemical group 0.000 description 1
- 125000003072 pyrazolidinyl group Chemical group 0.000 description 1
- 125000001725 pyrenyl group Chemical group 0.000 description 1
- 125000000168 pyrrolyl group Chemical group 0.000 description 1
- 125000002943 quinolinyl group Chemical group N1=C(C=CC2=CC=CC=C12)* 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 230000008844 regulatory mechanism Effects 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 238000011076 safety test Methods 0.000 description 1
- 125000002914 sec-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 150000003354 serine derivatives Chemical class 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229940054269 sodium pyruvate Drugs 0.000 description 1
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical class O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 125000003003 spiro group Chemical group 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000010025 steaming Methods 0.000 description 1
- 230000001954 sterilising effect Effects 0.000 description 1
- 238000004659 sterilization and disinfection Methods 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 208000011580 syndromic disease Diseases 0.000 description 1
- AKQXKEBCONUWCL-MRVPVSSYSA-N tert-butyl (3r)-3-aminopiperidine-1-carboxylate Chemical compound CC(C)(C)OC(=O)N1CCC[C@@H](N)C1 AKQXKEBCONUWCL-MRVPVSSYSA-N 0.000 description 1
- GHDNLFGEJLLKEC-UHFFFAOYSA-N tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrrolo[2,3-b]pyridine-1-carboxylate Chemical compound C1=CN=C2N(C(=O)OC(C)(C)C)C=CC2=C1B1OC(C)(C)C(C)(C)O1 GHDNLFGEJLLKEC-UHFFFAOYSA-N 0.000 description 1
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 125000005931 tert-butyloxycarbonyl group Chemical group [H]C([H])([H])C(OC(*)=O)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 125000001973 tert-pentyl group Chemical group [H]C([H])([H])C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- VXKWYPOMXBVZSJ-UHFFFAOYSA-N tetramethyltin Chemical compound C[Sn](C)(C)C VXKWYPOMXBVZSJ-UHFFFAOYSA-N 0.000 description 1
- 125000001984 thiazolidinyl group Chemical group 0.000 description 1
- 239000002562 thickening agent Substances 0.000 description 1
- 125000004568 thiomorpholinyl group Chemical group 0.000 description 1
- VNNLHYZDXIBHKZ-UHFFFAOYSA-N thiophene-2-sulfonyl chloride Chemical compound ClS(=O)(=O)C1=CC=CS1 VNNLHYZDXIBHKZ-UHFFFAOYSA-N 0.000 description 1
- 210000001519 tissue Anatomy 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000041 toxicology testing Toxicity 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 125000004306 triazinyl group Chemical group 0.000 description 1
- 150000003852 triazoles Chemical class 0.000 description 1
- 125000001425 triazolyl group Chemical group 0.000 description 1
- 238000013414 tumor xenograft model Methods 0.000 description 1
- 230000005760 tumorsuppression Effects 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 238000012403 whole-cell patch-clamp technology Methods 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
- A61K31/435—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
- A61K31/4418—Non condensed pyridines; Hydrogenated derivatives thereof having a carbocyclic group directly attached to the heterocyclic ring, e.g. cyproheptadine
-
- 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
- A61K31/4427—Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
-
- 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
- A61K31/4427—Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
- A61K31/443—Non condensed pyridines; Hydrogenated derivatives thereof 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
- A61K31/4427—Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
- A61K31/4433—Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-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
- A61K31/4427—Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
- A61K31/4436—Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a heterocyclic ring having sulfur 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
- A61K31/4427—Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
- A61K31/4439—Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
-
- 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
- A61K31/4427—Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
- A61K31/444—Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
-
- 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
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
- C07D401/04—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
- C07D401/12—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D405/00—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
- C07D405/02—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
- C07D405/04—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D405/00—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
- C07D405/02—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
- C07D405/12—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D409/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
- C07D409/02—Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
- C07D409/12—Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- 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
- C07D413/02—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
- C07D413/12—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D417/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
- C07D417/02—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
- C07D417/12—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present application provides the use of a cyclin-dependent kinase 9 inhibitor. In particular, the present application provides compounds of formula (I) or a salt thereofThe medicinal salt, the stereoisomer or the prodrug thereof can be applied to various applications in treating solid tumors, particularly liver cancer, breast cancer or prostate cancer, and comprise related pharmaceutical applications, pharmaceutical compositions or treatment methods and the like.
Description
RELATED APPLICATIONS
This application claims priority to chinese patent application No. 202110564093.0 filed 24/5/2021, the entire contents of which are hereby incorporated by reference in their entirety for all purposes.
Technical Field
The application relates to the field of medicines, in particular to a cyclin dependent kinase 9 (CDK 9) inhibitor or a pharmaceutically acceptable salt, a stereoisomer or a prodrug thereof, a pharmaceutical composition containing the same, or a use of a medicament containing the same, and particularly relates to a use of the inhibitor or the pharmaceutically acceptable salt, the stereoisomer or the prodrug thereof, the pharmaceutical composition containing the same, or the medicament containing the same in preparation of medicaments for treating solid tumor diseases, particularly malignant solid tumors.
Background
Cyclin-dependent kinases (CDKs) are a class of serine/threonine protein kinases that play key roles in regulating the cell cycle and transcription. Up to now, there are more than 20 subtypes of human CDKs and about 30 cyclin-chaperones (Cao et al.2014) known, which can be activated by cyclins and exert different biological functions, and CDKs can be divided into two by function, one to control the cell cycle and one to regulate cell transcription. For example, CDK1, 2,3, 4 and 6 directly intervene in the cell cycle; CDK5 does not regulate the cell cycle, but plays a key role in the complex migration of post-mitotic neurons; CDK7 acts indirectly as an activator of these CDKs; CDK9 only plays a role in cellular transcription and is not involved in the regulation of the cell cycle.
CDK9 is an important member of the transcribed CDKs subfamily, a group of kinases that function as the major steps in the control of the synthesis and processing of mRNA by eukaryotic RNA polymerase II (Pol II). CDK9 is present in all mammalian cells and activation of CDK9 in vivo is dependent on its binding to the corresponding Cyclin (Cyclin T/K) to form a heterodimer, positive transcription elongation factor b (P-TEFb). When the negative transcription elongation factors (NELF and NELFs) are involved in the negative regulation of cell transcription, the transcription is inhibited, the P-TEFb is recruited into a system for inhibiting the transcription elongation by the negative transcription elongation factors, the phosphorylation of a Carbon Terminal Domain (CTD) of RNA polymerase II is catalyzed, and the phosphorylation of an SPT5 subunit of the NELFs and an RD subunit of the NELF are catalyzed, so that the negative transcription elongation factors are separated from a transcription complex, and the transcription is continued.
Tumors are often caused by either a loss of cyclin-dependent kinase inhibitor (CDKI) expression or by over-expression of cyclins that render the cells unregulated and hyperproliferative. In view of the above regulatory mechanisms, the use of CDK9 inhibitors would prevent P-TEFb from phosphorylating the carbon-terminal domain of RNA polymerase II, further blocking NEFL exit, enhancing negative inhibition, causing transcriptional arrest, allowing rapid decrease in intracellular mRNA and short-half-life protein levels, which could lead to apoptosis of tumor cells. CDK9 has become a potential protein target for the development of effective cancer therapies and CDK9 inhibitors have recently been investigated by pharmaceutical companies for cancer therapy, e.g., AZD4573 of astrazen and BAY-1251152 of bayer, both in phase I clinical trials and no CDK9 inhibitors have been approved for sale.
Although some CDK9 inhibitor small molecules (e.g., WO2009047359, WO2014076091, etc.) have been disclosed so far, there is still a need to develop new compounds with good drug efficacy and good safety, which will benefit more patients clinically.
Disclosure of Invention
In one aspect, the present application provides a compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof for use in the preparation of a medicament for the treatment of solid tumors, in particular malignant solid tumors.
In another aspect, the present application provides a compound represented by formula (I) or a pharmaceutically acceptable salt, a stereoisomer or a prodrug thereof, for use in the preparation of a medicament for treating liver cancer, breast cancer or prostate cancer, in particular CDK 9-related liver cancer, breast cancer or prostate cancer.
In another aspect, the present application provides the use of compound 45 or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof as a CDK9 inhibitor in the preparation of a medicament for the treatment of a solid tumor, in particular a malignant solid tumor.
In another aspect, the present application provides the use of compound 45 or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof as a CDK9 inhibitor in the preparation of a medicament for the treatment of liver, breast or prostate cancer, particularly CDK 9-related liver, breast or prostate cancer.
In another aspect, the present application provides a pharmaceutical composition for treating solid tumors, which comprises a compound represented by formula (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof.
In another aspect, the present application provides a pharmaceutical composition for treating liver cancer, breast cancer or prostate cancer, comprising a compound represented by formula (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof.
In another aspect, the present application provides a pharmaceutical composition for treating a solid tumor comprising compound 45 or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof.
In another aspect, the present application provides a pharmaceutical composition for treating liver cancer, breast cancer or prostate cancer, comprising compound 45 or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof.
Previous work by the inventors of the present application has developed novel compounds having cyclin dependent kinase 9 (CDK 9) inhibitor activity (see publication No. WO2021/115335A1 or application No. PCT/CN2020/134966, the entire contents of which are incorporated herein by reference in their entirety for all purposes). On the basis of this, the inventors of the present application have made further studies on the medical use of the compound, and have obtained various inventions of the present application.
In particular to the application of a compound shown as a formula (I) or a pharmaceutically acceptable salt, a stereoisomer or a prodrug thereof in preparing a medicament for treating solid tumors,
wherein,
x is selected from Cl and F, wherein F is preferred;
R 1 selected from substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; r 1 Said "substituted" in (a) means substituted with 1,2, 3,4 or 5 substituents each independently selected from-F, -Cl, -Br, -NH 2 、-OH、-SH、-CN、-NO 2 、-N 3 、-C≡CH、-COOH、-R 3 、-(CH 2 ) w O(CH 2 ) n R 3 、-(CH 2 ) w NH(CH 2 ) n R 3 、-(CH 2 ) w NR 3 (CH 2 ) n R 4 、-(CH 2 ) w S(CH 2 ) n R 3 、-(CH 2 ) w C(O)(CH 2 ) n R 3 、-(CH 2 ) w C(O)O(CH 2 ) n R 3 、-(CH 2 ) w OC(O)(CH 2 ) n R 3 、-(CH 2 ) w C(O)NH(CH 2 ) n R 3 、-(CH 2 ) w NHC(O)(CH 2 ) n R 3 、-(CH 2 ) w C(O)NR 3 (CH 2 ) n R 4 、-(CH 2 ) w NR 3 C(O)(CH 2 ) n R 4 、-(CH 2 ) w OS(O) 2 (CH 2 ) n R 3 Or- (CH) 2 ) w S(O) 2 O(CH 2 ) n R 3 Substituted with a group of (a); wherein each occurrence of w, n is independently selected from 0,1, 2,3 or 4; r is 3 And R 4 Each independently selected from substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, and substituted or unsubstituted C 1-6 Alkyl, substituted or unsubstituted C 1-6 Haloalkyl, substituted or unsubstituted C 2-6 Alkenyl, substituted or unsubstituted C 2-6 Alkynyl, substituted or unsubstituted C 1-6 Alkoxy, substituted or unsubstituted C 1-6 Haloalkoxy, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, or when R is 3 、R 4 When taken together to the same nitrogen atom, R 3 、R 4 And the nitrogen atom to which they are both attached form a substituted or unsubstituted heterocycloalkyl group; r 3 And R 4 Said "substituted" in (a) means substituted with 1,2 or 3 substituents each independently selected from-F, -Cl, -Br, -NH 2 、-OH、-SH、-CN、-NO 2 、-N 3 、-C≡CH、-COOH、C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Alkoxy radical, C 1-6 Halogenoalkoxy, etc.;
ring A is selected from substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl; said "substituted" in ring A is by 1,2, 3,4 or 5 groups each independently selected from-F, -Cl, -Br, OH, NH 2 、SH、CN、NO 2 、-N 3 、-C≡CH、COOH、R 5 、OR 5 、-NHR 5 、-NR 5 R 6 、-SR 5 、-NHCOR 5 、-CONHR 5 、-NHS(O) 2 R 5 、-S(O) 2 NHR 5 、-NR 5 S(O) 2 R 6 、-S(O) 2 NR 5 R 6 Or 1,2 or more-CH in the A ring structure 2 -the group can optionally be replaced by a-C (O) -group; wherein R is 5 And R 6 Independently is C 1-6 Alkyl radical, C 1-6 A haloalkyl group.
R 2 Selected from H, R 7 、-(CH 2 ) x R 7 、-(CH 2 ) x NH(CH 2 ) y R 7 、-(CH 2 ) x O(CH 2 ) y R 7 、-(CH 2 ) x NR 7 (CH 2 ) y R 8 、-(CH 2 ) x C(O)(CH 2 ) y H、-(CH 2 ) x C(O)(CH 2 ) y R 7 、-(CH 2 ) x S(O) 2 (CH 2 ) y R 7 、-(CH 2 ) x C(O)C(O)(CH 2 ) y R 7 、-(CH 2 ) x S(O) 2 NH 2 、-(CH 2 ) x NHS(O) 2 H、-(CH 2 ) x S(O) 2 NH(CH 2 ) y R 7 、-(CH 2 ) x NHS(O) 2 (CH 2 ) y R 7 、-(CH 2 ) x S(O) 2 NR 7 (CH 2 ) y R 8 、-(CH 2 ) x NR 7 S(O) 2 (CH 2 ) y R 8 、-(CH 2 ) x C(O)O(CH 2 ) y R 7 、-(CH 2 ) x OC(O)(CH 2 ) y R 7 、-(CH 2 ) x C(O)NH 2 、-(CH 2 ) x NHC(O)H、-(CH 2 ) x C(O)NH(CH 2 ) y R 7 、-(CH 2 ) x NHC(O)(CH 2 ) y R 7 、-(CH 2 ) x C(O)NR 7 (CH 2 ) y R 8 Or- (CH) 2 ) x NR 7 C(O)(CH 2 ) y R 8 (ii) a Wherein 1,2 or more-CH 2 -the group can optionally be replaced by a-C (O) -group; x, y are independently selected for each occurrence from 0,1, 2,3 or 4;
R 7 and R 8 Independently selected from substituted or unsubstituted R 9 、OR 9 、-R 10 -O-R 9 、-R 10 -NH-R 9 、-R 10 -C(O)-R 9 、-R 10 -NHC(O)-R 9 、-R 10 -C(O)NH-R 9 、-R 10 -S-R 9 、-R 10 -S(O)-R 9 、-R 10 -S-C(O)-R 9 Cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -R 10 -aryl, -R 10 -heteroaryl, -O-R 10 -aryl, -O-R 10 -heteroaryl, -R 10 -O-aryl, -R 10 -O-heteroaryl, -cycloalkyl-aryl, -cycloalkyl-heteroaryl, -heterocycloalkyl-aryl, -heterocycloalkyl-heteroaryl, C 2-6 Olefins and C 2-6 Alkyne, or when R 7 And R 8 When they are jointly bound to the same nitrogen atom, R 7 And R 8 With the nitrogen atom to which they are both attached to form a substituted or unsubstituted heterocycloalkyl; wherein R is 9 Is C 1-6 Alkyl radical, R 10 Is C 1-6 Alkylene radical, C 2-6 Alkenylene or C 2-6 An alkynylene group; r 7 And R 8 Said "substituted" in (a) means substituted with 1,2 or 3 substituents each independently selected from-F, -Cl, -Br, -OH, -NH 2 、-SH、-CN、-NO 2 、-N 3 、-C≡CH、-COOH、C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, -NHCN, -NHCONH 2 、NHC(O)CH 3 、N(CH 3 ) 2 、N(C 2 H 5 ) 2 、-SC(O)CH 3 、-OC(O)-C 1-6 Alkyl, etc.
Definitions for "aryl", "heteroaryl", "cycloalkyl", "heterocycloalkyl" in this application are as defined in the "definitions" section below.
In one embodiment, aryl preferably contains 6 to 10 carbon atoms, cycloalkyl preferably contains 3 to 6 carbon atoms, heteroaryl preferably is 5 to 10 membered heteroaryl, and heterocycloalkyl preferably is 3 to 8 membered heterocyclyl; heteroaryl or heterocycloalkyl preferably contains 1,2 or 3 heteroatoms each independently selected from N, O or S, the remainder being carbon atoms.
With regard to "w", "n", "x", "y" in the present application, each may be independently selected from 0,1, 2,3 or 4 as previously described, when "w" and "n", "x" and "y" are simultaneously present in one group, specifically, the numerical combination of "w" and "n", "x" and "y" may be selected from (0,0), (0,1), (0,2), (0,3), (3575), (1,0), (1,1), (1,2), (1,3), (1,4), (5678 zxft 3578), (2,1), (8624), (9696 zxft 963535), (3292 zxft 32323292), (5634zxft 5292), (3535353535zzft 354258), (354235) may be applied to the combination of these values (379, 35424235, 354235, 354292, 35zxft 5292, 354235, and the combination of these values 1 、R 2 Each relevant group in the definition, for example, R 1 In definition of- (CH) 2 ) w O(CH 2 ) n R 3 Is equivalent to disclose-OR 3 、-OCH 2 R 3 、-O(CH 2 ) 2 R 3 、-O(CH 2 ) 3 R 3 、-O(CH 2 ) 4 R 3 、-CH 2 OR 3 、-CH 2 OCH 2 R 3 、-CH 2 O(CH 2 ) 2 R 3 、-CH 2 O(CH 2 ) 3 R 3 、-CH 2 O(CH 2 ) 4 R 3 、-(CH 2 ) 2 OR 3 、-(CH 2 ) 2 OCH 2 R 3 、-(CH 2 ) 2 O(CH 2 ) 2 R 3 、-(CH 2 ) 2 (CH 2 ) 3 R 3 、-(CH 2 ) 2 O(CH 2 ) 4 R 3 、-(CH 2 ) 3 OR 3 、-(CH 2 ) 3 OCH 2 R 3 、-(CH 2 ) 3 O(CH 2 ) 3 R 3 、-(CH 2 ) 3 O(CH 2 ) 3 R 3 、-(CH 2 ) 3 O(CH 2 ) 4 R 3 、-(CH 2 ) 4 OR 3 、-(CH 2 ) 4 OCH 2 R 3 、-(CH 2 ) 4 O(CH 2 ) 4 R 3 、-(CH 2 ) 4 O(CH 2 ) 3 R 3 、-(CH 2 ) 4 O(CH 2 ) 4 R 3 The same as R 1 、R 2 Other related groups in (1) also disclose such a selection, and are not described in detail.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, wherein ring a is selected from substituted or unsubstituted 4-6 membered cycloalkyl, substituted or unsubstituted 4-6 membered heterocycloalkyl, or substituted or unsubstituted 5-6 cycloalkyl, or substituted unsubstituted 5-6 membered heterocycloalkyl; further preferably, the A ring is selected from substituted or unsubstituted 5-6 cycloalkyl, or substituted unsubstituted 5-6 membered heterocycloalkyl; still more preferably, the A ring is selected from substituted or unsubstituted 5-6 cycloalkyl.
In one embodiment, ring a is selected from the group consisting of cyclohexyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, cyclopentyl, or morpholinyl, further preferably ring a is selected from the group consisting of cyclohexyl, cyclopentyl, or tetrahydropyrrolyl.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, wherein the "substitution" in ring A is by 1,2, 3,4, or 5, each independently selected from-F, -Cl, -Br, OH, NH 2 、SH、CN、R 5 、OR 5 Wherein R is 5 Is C 1-6 Alkyl or C 1-6 Alkoxy radical, R 5 Can be further selected from C 1-4 Alkyl or C 1-4 An alkoxy group.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, has the structure of formula (II):
wherein R is 1 、R 2 And X is represented by formula (I).
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, has the structure of formula (IIa):
wherein R is 1 、R 2 And X is represented by formula (I).
In one embodiment, the compound of formula (I), or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, has the structure of formula (III):
wherein R is 1 、R 2 And X is represented by formula (I).
In one embodiment, the compound of formula (I), or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, has the structure of formula (IIIa):
wherein R is 1 、R 2 And X is represented by formula (I).
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, wherein R is 1 Selected from substituted or unsubstituted 6-10 membered aryl, or substituted or unsubstituted 5-10 membered heteroaryl; the heteroaryl group contains 1 or 2 heteroatoms each independently selected from N or O; number of said substituentThe amount is selected from 1,2 or 3.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, wherein R is 1 Selected from substituted or unsubstituted benzene rings, pyridine rings, indole rings, indazole rings, benzofuran rings, pyrrolopyridine rings; further preferred are a substituted or unsubstituted benzene ring, pyridine ring, indole ring, benzofuran ring, pyrrolopyridine ring; further preferred are a substituted benzene ring, a pyridine ring and an unsubstituted indole ring, a benzofuran ring, a pyrrolopyridine ring; substituted benzene rings are even more preferred.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, wherein R is 1 The substituents on the above are respectively and independently selected from-F, -Cl, -OH and-NH 2 、-R 3 、-(CH 2 ) w O(CH 2 ) n R 3 Or- (CH) 2 ) w OC(O)(CH 2 ) n R 3 (ii) a w and n are independently selected from 0,1 or 2 at each occurrence, wherein R 3 As defined in formula (I). Preferably, said R is 1 The substituents on the substituent groups are respectively and independently selected from-F-, -Cl, -OH and-R 3 、-(CH 2 ) w O(CH 2 ) n R 3 (ii) a Further preferably, said R 1 The substituents on the substituent group are respectively and independently selected from-F-, -OH and-R 3 、-(CH 2 ) w O(CH 2 ) n R 3 . For example, when R is 1 In the case of a substituted phenyl ring, the substituent is selected from the group consisting of-F, -OH or alkoxy, preferably 1 or 2 fluorine atoms and 1 alkoxy.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, has the structure of formula (II):
wherein R is 1 Is substituted benzeneA ring, the substituents selected from-F, -OH or alkoxy; preferably 1 or 2 fluorine atoms and 1-OH or alkoxy, preferably 1 or 2 fluorine atoms and 1 alkoxy;
R 2 and X is represented by formula (I).
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, has the structure of formula (IIa):
wherein, R is 1 Is a substituted benzene ring, and the substituent is selected from-F, -OH or alkoxy; preferably 1 or 2 fluorine atoms and 1-OH or alkoxy, preferably 1 or 2 fluorine atoms and 1 alkoxy;
R 2 and X is represented by formula (I).
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, wherein R is 3 And R 4 Each independently selected from substituted or unsubstituted 6-membered aryl, substituted or unsubstituted 5-6-membered heteroaryl, substituted or unsubstituted C 1-3 Alkyl, substituted or unsubstituted C 1-3 Alkoxy, substituted or unsubstituted C 3-6 Cycloalkyl, substituted or unsubstituted C 3-6 Heterocycloalkyl, or when R is 3 、R 4 When they are jointly bound to the same nitrogen atom, R 3 、R 4 And the nitrogen atom to which they are both attached form a 3-7 membered substituted or unsubstituted heterocycloalkyl group; the heterocycloalkyl contains 1 or 2 heteroatoms independently selected from N, O or S; r 3 And R 4 The term "substituted" as used herein means substituted with 1,2 or 3 substituents each independently selected from the group consisting of-F, -Cl, -Br, -OH, -CH 3 、-C 2 H 5 、-OCH 3 、-OC 2 H 5 Substituted with the substituent(s); preferably, R 3 And R 4 Each independently selected from substituted or unsubstituted C 1-3 Alkyl, substituted or unsubstituted C 1-3 An alkoxy group.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, wherein R is 3 And R 4 Each independently selected from the group consisting of substituted or unsubstituted benzene ring, pyridine ring, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, R 3 And R 4 The term "substituted" as used herein means substituted with 1,2 or 3 substituents each independently selected from the group consisting of-F, -Cl, -Br, -OH, -CH 3 、-C 2 H 5 、-OCH 3 、-OC 2 H 5 Substituted with the substituent(s). Preferably, R 3 And R 4 Each independently selected from substituted or unsubstituted methyl, ethyl, isopropyl, methoxy, ethoxy, isopropoxy, cyclopropyl, pyridine ring; further preferably, R 3 And R 4 Each independently selected from substituted or unsubstituted methyl, ethyl, isopropyl, methoxy, ethoxy, isopropoxy; even more preferably, R 3 And R 4 Each independently selected from substituted or unsubstituted methyl, methoxy.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, wherein R is 2 Is selected from R 7 、-(CH 2 ) x R 7 、-(CH 2 ) x NH(CH 2 ) y R 7 、-(CH 2 ) x C(O)(CH 2 ) y R 7 、-(CH 2 ) x S(O) 2 (CH 2 ) y R 7 、-(CH 2 ) x C(O)C(O)(CH 2 ) y R 7 、-(CH 2 ) x C(O)O(CH 2 ) y R 7 、-(CH 2 ) x C(O)NH(CH 2 ) y R 7 、-(CH 2 ) x C(O)NR 7 (CH 2 ) y R 8 Or- (CH) 2 ) x NR 7 C(O)(CH 2 ) y R 8 Wherein, said R 7 And R 8 The same formula (I) is defined. Further preferably, R 2 Is selected from R 7 、-(CH 2 ) x R 7 、-(CH 2 ) x C(O)(CH 2 ) y R 7 、-(CH 2 ) x S(O) 2 (CH 2 ) y R 7 、-(CH 2 ) x C(O)C(O)(CH 2 ) y R 7 、-(CH 2 ) x C(O)O(CH 2 ) y R 7 、-(CH 2 ) x C(O)NH(CH 2 ) y R 7 (ii) a Even more preferably, R 2 Is selected from R 7 、-(CH 2 ) x R 7 、-(CH 2 ) x C(O)(CH 2 ) y R 7 (ii) a Even more preferably, R 2 Is selected from- (CH) 2 ) x C(O)(CH 2 ) y R 7 。
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, wherein R is 7 And R 8 Each independently selected from substituted or unsubstituted R 9 、OR 9 、-R 10 -O-R 9 、-R 10 -NH-R 9 、-R 10 -C(O)-R 9 、-R 10 -NHC(O)-R 9 、-R 10 -C(O)NH-R 9 、-R 10 -S-R 9 、-R 10 -S-C(O)-R 9 、C 3-6 Cycloalkyl, 3-6 membered heterocycloalkyl, C 6-10 Aryl, 5-10 membered heteroaryl, -R 10 -C 6-10 Aryl, -R 10 -5-10 membered heteroaryl, -O-R 10 -C 6-10 Aryl, -O-R 10 -5-10 membered heteroaryl, -R 10 -O-C 6-10 Aryl, -R 10 -O-5-10 membered heteroaryl, C 2-6 Olefins and C 2-6 Alkyne, or when R 7 And R 8 When they are jointly bound to the same nitrogen atom, R 7 And R 8 With the nitrogen atom to which they are both attached, to form a substituted or unsubstituted 3-6 membered heterocycloalkyl group; wherein R is 9 Is C 1-6 Alkyl radical, R 10 Is C 1-6 Alkylene radical, C 2-6 Alkenylene or C 2-6 An alkynylene group; r 7 And R 8 Said "substituted" in (1) means by 1,2 or3 are each independently selected from-F, -Cl, -Br, -OH and-NH 2 、-SH、-CN、C 1-3 Alkyl radical, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Haloalkoxy, -NHCN, -NHCONH 2 、NHC(O)CH 3 、N(CH 3 ) 2 、N(C 2 H 5 ) 2 、-SC(O)CH 3 、-OC(O)-C 1-6 Alkyl, etc.; preferably, R 7 Independently selected from substituted or unsubstituted R 9 、OR 9 、-R 10 -O-R 9 、-R 10 -NHC(O)-R 9 、C 3-6 Cycloalkyl, 3-6 membered heterocycloalkyl, C 6-10 Aryl, 5-10 membered heteroaryl, C 2-6 Olefins and C 2-6 Alkyne, or when R 7 And R 8 When taken together to the same nitrogen atom, R 7 And R 8 With the nitrogen atom to which they are both attached, to form a substituted or unsubstituted 3-6 membered heterocycloalkyl group; further preferably, R 7 Independently selected from substituted or unsubstituted R 9 、OR 9 、-R 10 -O-R 9 、C 3-6 Cycloalkyl, 3-6 membered heterocycloalkyl, C 6-10 Aryl, 5-10 membered heteroaryl; further preferably, R 7 Independently selected from substituted or unsubstituted R 9 、OR 9 、-R 10 -O-R 9 、C 3-6 Cycloalkyl, 3-6 membered heterocycloalkyl; further preferably, R 7 Independently selected from substituted or unsubstituted R 9 ;R 7 The term "substituted" as used herein means substituted with 1,2 or 3 substituents each independently selected from the group consisting of-F, -Cl, -Br, -OH and-NH 2 、-SH、-CN、C 1-3 Alkyl radical, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Haloalkoxy, -NHCN, -NHCONH 2 、NHC(O)CH 3 、N(CH 3 ) 2 、N(C 2 H 5 ) 2 、-SC(O)CH 3 、-OC(O)-C 1-6 Alkyl, etc.; preferably, R 7 The term "substituted" as used herein means that 1,2 or 3 of them are each independently selected from the group consisting of-F, -OH, -NH 2 、-SH、-CN、C 1-3 Alkyl radical, C 1-3 Alkoxy radical, C 1-3 HalogenatedAlkyl, NHC (O) CH 3 、N(CH 3 ) 2 、-OC(O)-C 1-6 Alkyl, etc.; further preferably, R 7 The term "substituted" as used herein means that 1,2 or 3 of them are each independently selected from the group consisting of-F, -OH, -NH 2 、-CN、C 1-3 Alkyl radical, C 1-3 Alkoxy, etc.; further preferably, R 7 The "substitution" as referred to in (1) means that 1,2 or 3 groups each independently selected from-OH, -CN, etc. are substituted; further preferably, R 7 The "substitution" as referred to in (1) means that 1 group each independently selected from-OH, -CN, etc. is substituted; further preferably, R 7 The "substitution" as described in (1) means that each of the 1 is independently substituted by a group selected from-OH groups.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, wherein R is 7 And R 8 Each independently selected from substituted or unsubstituted methyl, ethyl, propyl, isopropyl, butyl, pentyl, methoxy, ethoxy, propoxy, isopropoxy, -CH 2 OCH 3 、-CH 2 OCH 2 CH 3 、-CH 2 CH 2 OCH 3 、-CH 2 CH 2 OCH 2 CH 3 Cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, aziridinyl, azetidinyl, azacyclohexyl, oxacyclopropyl, oxetanyl, oxacyclopentyl, oxacyclohexyl, phenyl, pyridyl, pyrazolyl, oxazolyl, isoxazolyl, thienyl, thiazolyl, benzyl, phenethyl, ethenyl, propenyl, ethynyl or propynyl. Preferably, R 7 Independently selected from substituted or unsubstituted methyl, ethyl, methoxy, ethoxy, cyclopropyl, cyclobutyl, azetidinyl, azacyclohexyl, oxetanyl, oxacyclohexyl, phenyl, pyridyl, pyrazolyl, isoxazolyl, thienyl, thiazolyl, benzyl, ethenyl, propenyl or ethynyl; further preferably, R 7 Independently selected from the group consisting of substituted or unsubstituted methyl, ethyl, methoxy, cyclopropyl, cyclobutyl, azacyclohexyl, oxetanyl, oxaCyclohexyl, pyridyl, pyrazolyl, isoxazolyl, ethenyl, propenyl, or ethynyl; further preferably, R 7 Independently selected from substituted or unsubstituted methyl, ethyl, cyclopropyl, azacyclohexyl, oxetanyl, pyrazolyl, ethenyl, propenyl, or ethynyl; even more preferably, R 7 Independently selected from substituted or unsubstituted methyl, ethyl, cyclopropyl; even more preferably, R 7 Independently selected from substituted or unsubstituted methyl. R 7 And R 8 Said "substituted" in (a) means substituted with 1,2 or 3 substituents each independently selected from-F, -Cl, -Br, -OH, -NH 2 、-SH、-CN、C 1-3 Alkyl radical, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Haloalkoxy, -NHCN, -NHCONH 2 、NHC(O)CH 3 、N(CH 3 ) 2 、N(C 2 H 5 ) 2 、-SC(O)CH 3 、-OC(O)-C 1-6 Alkyl, etc.; preferably, R 7 And R 8 Said "substituted" in (a) means that the compound is substituted with 1,2 or 3 substituents each independently selected from-F, -OH, -SH, -CN, C 1-3 Alkyl radical, C 1-3 Alkoxy radical, C 1-3 Haloalkoxy, NHC (O) CH 3 、N(CH 3 ) 2 、N(C 2 H 5 ) 2 、-OC(O)-C 1-6 Alkyl, etc.; preferably, R 7 And R 8 Said "substituted" in (a) means substituted with 1,2 or 3 substituents each independently selected from-F, -OH, -NH 2 、-CN、C 1-3 Halogenoalkoxy, etc.; further preferably, R 7 And R 8 Said "substituted" in (a) means substituted with 1,2 or 3 groups each independently selected from-F, -OH, -CN, etc.; further preferably, R 7 And R 8 Said "substituted" in (a) means substituted with 1,2 or 3 groups each independently selected from-OH, -CN, etc.; further preferably, R 7 And R 8 Said "substituted" in (1) means substituted with 1 group each independently selected from-OH, -CN, etc.; further preferably, R 7 And R 8 Said "substituted" in (1) means substituted by 1 eachIndependently selected from-OH groups.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, wherein R is 9 Is C 1-4 Alkyl radical, R 10 Is C 1-4 An alkylene group.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, wherein the compound has the structure:
in one embodiment, the solid tumor is a malignant solid tumor; preferably advanced malignant solid tumors; further preferably liver cancer, breast cancer or prostate cancer.
In one embodiment, the liver, breast or prostate cancer is CDK 9-associated liver, breast or prostate cancer. Preferably, the liver cancer, breast cancer or prostate cancer is liver cancer, breast cancer or prostate cancer caused by CDK9 overexpression.
In one embodiment, the liver cancer is hepatocellular carcinoma; preferably advanced hepatocellular carcinoma; further preferred is CDK 9-associated late-stage hepatocellular carcinoma; further preferred is late hepatocellular carcinoma caused by CDK9 overexpression.
In one embodiment, the breast cancer is a triple negative breast cancer.
In one embodiment, the triple negative breast cancer is CDK 9-associated triple negative breast cancer; preferably triple negative breast cancer caused by CDK9 overexpression.
In one embodiment, said CDK9 associated is CDK9 is overexpressed.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof is the only active ingredient in the medicament.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof is used in combination with one or more other targeting or chemotherapeutic agents for the manufacture of the medicament.
In one embodiment, the medicament is formulated for clinical acceptance. In a preferred embodiment, the formulation is an oral formulation or an injectable formulation.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof is administered at a daily dosage range of from about 0.001mg/kg to about 1000 mg/kg; preferably 0.01mg/kg to about 100mg/kg, and more preferably 0.02mg/kg to about 10mg/kg.
In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof is administered at a daily dosage range of from about 0.001mg to about 1000 mg; preferably 0.01mg to about 100mg, further preferably 0.1mg to about 80mg, further preferably 1mg to about 70mg, further preferably 1.5mg to about 60mg, further preferably 2mg to about 50mg. More preferably 2mg, 3mg, 4mg, 5mg, 6mg, 8mg, 10mg, 12mg, 16mg, 18mg, 20mg, 24mg, 25mg, 30mg, 32mg, 36mg, 40mg, 42mg, 45mg, 50mg. The frequency of administration is a single administration or multiple administrations. Preferably, the frequency of administration is once daily, twice daily, three times daily, once every two days, once every three days, once every four days, once every five days, once every six days, or once every seven days.
In one embodiment, the medicament contains a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof. In a preferred embodiment, the therapeutically effective amount is 0.001-1000mg. In a preferred embodiment, the therapeutically effective amount is 0.01-100mg. In a preferred embodiment, the therapeutically effective amount is 0.1-50mg. More preferably 2mg, 3mg, 4mg, 5mg, 6mg, 8mg, 10mg, 12mg, 16mg, 18mg, 20mg, 24mg, 25mg, 30mg, 32mg, 36mg, 40mg, 42mg, 45mg, 50mg.
In one embodiment, the medicament comprises 0.01-100mg of a compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof; preferably 0.1-80mg; further preferably 1 to 60mg; further preferably 1-50mg; further preferably 2-20mg; further preferably 2 to 15mg; further preferably 2 to 10mg; further preferably 2 to 8mg; further preferably 2 to 6mg; further preferably 2 to 5mg; further preferably 2 to 4mg; more preferably 2 to 3mg.
In one embodiment, the compound represented by the formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, or a prodrug thereof is administered in a single dose or in divided doses.
In one embodiment, the medicament is administered orally or by injection. In a preferred embodiment, the medicament is administered orally.
In a second aspect of the present application, there is provided a pharmaceutical composition for treating solid tumors, comprising a compound of formula (I) as described in the first aspect, or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, and optionally a pharmaceutically acceptable carrier.
In case of no contradiction or conflict, the technical solutions or features described in the first aspect apply to the second aspect.
In a third aspect of the present application, there is provided the use of compound 45 of the present application, or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, in the manufacture of a medicament for the treatment of a solid tumor,
in one embodiment, the solid tumor is a malignant solid tumor; preferably advanced malignant solid tumors; further preferably liver cancer, breast cancer or prostate cancer.
In one embodiment, the liver, breast or prostate cancer is CDK 9-associated liver, breast or prostate cancer. Preferably, the liver cancer, breast cancer or prostate cancer is liver cancer, breast cancer or prostate cancer caused by CDK9 overexpression.
In one embodiment, the liver cancer is hepatocellular carcinoma; preferably advanced hepatocellular carcinoma; further preferred is CDK 9-associated late-stage hepatocellular carcinoma; further preferred is late hepatocellular carcinoma caused by CDK9 overexpression.
In one embodiment, the breast cancer is a triple negative breast cancer.
In one embodiment, the triple negative breast cancer is CDK 9-associated triple negative breast cancer; preferably triple negative breast cancer caused by CDK9 overexpression.
In one embodiment, said CDK9 associated is CDK9 is overexpressed.
In one embodiment, said compound 45 or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof is the only active ingredient in said medicament.
In one embodiment, said compound 45, or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, is used in combination with one or more other targeting or chemotherapeutic agents for the preparation of said medicament.
In one embodiment, the medicament is formulated for clinical acceptance. In a preferred embodiment, the formulation is an oral formulation or an injectable formulation.
In one embodiment, said compound 45, or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, is administered at a daily dosage range of from about 0.001mg/kg to about 1000 mg/kg; preferably 0.01mg/kg to about 100mg/kg, and more preferably 0.02mg/kg to about 10mg/kg.
In one embodiment, said compound 45, or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof, is administered at a daily dosage range of from about 0.001mg to about 1000 mg; preferably 0.01mg to about 100mg, further preferably 0.1mg to about 80mg, further preferably 1mg to about 70mg, further preferably 1.5mg to about 60mg, further preferably 2mg to about 50mg. More preferably 2mg, 3mg, 4mg, 5mg, 6mg, 8mg, 10mg, 12mg, 16mg, 18mg, 20mg, 24mg, 25mg, 30mg, 32mg, 36mg, 40mg, 42mg, 45mg, 50mg. The frequency of administration is a single administration or multiple administrations. Preferably, the frequency of administration is once daily, twice daily, three times daily, once every two days, once every three days, once every four days, once every five days, once every six days, or once every seven days.
In one embodiment, the medicament contains a therapeutically effective amount of compound 45 or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof. In a preferred embodiment, the therapeutically effective amount is 0.001-1000mg. In a preferred embodiment, the therapeutically effective amount is 0.01-100mg. In a preferred embodiment, the therapeutically effective amount is 0.1-50mg.
In one embodiment, the medicament comprises 0.01-100mg of compound 45, or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof; preferably 0.1-80mg; further preferably 1 to 60mg; further preferably 1-50mg; further preferably 2-20mg; further preferably 2 to 15mg; further preferably 2 to 10mg; further preferably 2 to 8mg; further preferably 2 to 6mg; further preferably 2 to 5mg; further preferably 2 to 4mg; more preferably 2 to 3mg.
In one embodiment, said compound 45 or a pharmaceutically acceptable salt, stereoisomer, or prodrug thereof is administered in a single dose or in divided doses.
In one embodiment, the medicament is administered orally or by injection. In a preferred embodiment, the medicament is administered orally.
In a fourth aspect of the present application, there is provided a pharmaceutical composition for the treatment of solid tumors comprising compound 45 of the present application, or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, and optionally a pharmaceutically acceptable carrier.
In case of no contradiction or conflict, the solution or features described in the third aspect apply to the fourth aspect.
The compound has optical activity, can be racemic, optical isomer or a mixture thereof, and the synthesis of the optical isomer in the compound can be prepared from the starting materials of the optical isomer or can be prepared by racemic separation.
Definition of
The following terms used in the present application have the following meanings, unless otherwise specified. A particular term should not be considered as indefinite or unclear unless it is specifically defined, but rather construed according to ordinary meaning in the art. When a trade name appears herein, it is intended to refer to its corresponding commodity or its active ingredient.
The terms "optionally" or "optionally" mean that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. For example, ethyl "optionally" substituted with halo means that the ethyl group may be unsubstituted (CH) 2 CH 3 ) Monosubstituted (e.g. CH) 2 CH 2 F) Polysubstituted (e.g. CHFCH) 2 F、CH 2 CHF 2 Etc.) or completely substituted (CF 2 CF 3 ). It will be appreciated by those skilled in the art that any group containing one or more substituents will not incorporate any substitution or substitution pattern which is sterically impossible and/or cannot be synthesized.
Herein C m-n It is the moiety that has an integer number of carbon atoms in the given range. E.g. "C 1-6 By "is meant that the group can have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, or 6 carbon atoms.
When any variable (e.g., R) occurs more than one time in the composition or structure of a compound, its definition in each case is independent. Thus, for example, if a group is substituted with 2R, then there are separate options for each R.
The term "alkyl" refers to a monovalent saturated aliphatic hydrocarbon group, a straight or branched chain group containing 1-20 carbon atoms, preferably 1-10 carbon atoms (i.e., C) 1-10 Alkyl group), further preferably containing 1 to 8 carbon atoms (C) 1-8 Alkyl), more preferably containing 1 to 6 carbon atoms (i.e., C) 1-6 Alkyl) such as "C 1-6 By alkyl is meant that the group is alkyl and the number of carbon atoms in the carbon chain is between 1 and 6 (specifically 1,2, 3,4, 5 or 6). Examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, neopentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, n-heptyl, n-octyl, and the like.
The term "cycloalkyl" refers to a monocyclic saturated aliphatic radical having the specified number of carbon atoms, preferably containing from 3 to 12 carbon atoms (i.e., C) 3-12 Cycloalkyl), more preferably containing 3 to 10 carbon atoms (C) 3-10 Cycloalkyl group), further preferably 3 to 6 carbon atoms (C) 3-6 Cycloalkyl), 4 to 6 carbon atoms (C) 4-6 Cycloalkyl), 5 to 6 carbon atoms (C) 5-6 Cycloalkyl groups). Examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methylcyclopropyl, 2-ethyl-cyclopentyl, dimethylcyclobutyl, and the like.
The term "alkoxy" refers to an-O-alkyl group, as defined above, i.e. containing 1 to 20 carbon atoms, preferably 1 to 10 carbon atoms, preferably 1 to 8 carbon atoms, more preferably 1 to 6 carbon atoms (in particular 1,2, 3,4, 5 or 6). Representative examples include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, butoxy, 1-methylpropoxy, 2-methylpropoxy, tert-butoxy, pentyloxy, 1-methylbutoxy, 2-methylbutoxy, 3-methylbutoxy, 1,1-dimethylpropoxy, 1,2-dimethylpropoxy, 2,2-dimethylpropoxy, 1-ethylpropoxy, and the like.
The term "halogen" or "halo" refers to F, cl, br, I. The term "haloAlkyl "means an alkyl group as defined above in which one, two or more hydrogen atoms or all hydrogen atoms are replaced by halogen. Representative examples of haloalkyl groups include CCl 3 、CF 3 、CHCl 2 、CH 2 Cl、CH 2 Br、CH 2 I、CH 2 CF 3 、CF 2 CF 3 And so on.
The term "heterocyclyl" refers to a saturated or partially unsaturated monocyclic, bicyclic, or polycyclic cyclic hydrocarbon substituent, non-aromatic in structure, containing from 3 to 20 ring atoms, wherein 1,2, 3, or more ring atoms are selected from N, O or S, and the remaining ring atoms are C. Preferably containing 3 to 12 ring atoms (C) 3-12 Heterocyclyl) and further preferably contains 3 to 10 ring atoms (C) 3-10 Heterocyclyl), or 3 to 8 ring atoms (C) 3-8 Heterocyclyl), or 3 to 6 ring atoms (C) 3-6 Heterocyclyl), or 4 to 6 ring atoms (C) 4-6 Heterocyclyl), or 5 to 6 ring atoms (C) 5-6 A heterocyclic group). The number of heteroatoms is preferably 1-4, more preferably 1-3 (i.e. 1,2 or 3). Examples of monocyclic heterocyclic groups include pyrrolidinyl, imidazolidinyl, tetrahydrofuranyl, dihydropyrrolyl, piperidinyl, piperazinyl, pyranyl, and the like. Polycyclic heterocyclic groups include spiro, fused and bridged heterocyclic groups.
The term "heterocycloalkyl" refers to a saturated "heterocyclyl" as defined above, containing 3 to 20 ring atoms, of which 1,2, 3 or more ring atoms are selected from N, O or S, with the remaining ring atoms being C. Preferably containing 3 to 12 ring atoms (C) 3-12 Heterocycloalkyl group), further preferably contains 3 to 10 ring atoms (C) 3-10 Heterocycloalkyl), or 3 to 8 ring atoms (C) 3-8 Heterocycloalkyl), or 3 to 7 ring atoms (C) 3-7 Heterocycloalkyl), or 3 to 6 ring atoms (C) 3-6 Heterocycloalkyl), or 4 to 6 ring atoms (C) 4-6 Heterocycloalkyl), or 5 to 6 ring atoms (C) 5-6 Heterocycloalkyl). The number of heteroatoms is preferably 1-4, more preferably 1-3 (i.e. 1,2 or 3). Examples include aziridinyl, oxetanyl, thietanyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, tetrahydrofuranyl, oxacyclohexane, piperidinyl, piperazinyl, morpholineMesityl, thiomorpholinyl, dioxanyl, dithiacyclohexyl, oxazolidinyl, thiazolidinyl, pyrazolidinyl, imidazolinidinyl and the like.
The term "aryl" denotes monocyclic, bicyclic and tricyclic aromatic carbocyclic ring systems containing from 6 to 16 carbon atoms, or from 6 to 14 carbon atoms, or from 6 to 12 carbon atoms, or from 6 to 10 carbon atoms, preferably from 6 to 10 carbon atoms, the term "aryl" being used interchangeably with the term "aromatic ring". Examples of the aryl group may include, but are not limited to, phenyl, naphthyl, anthryl, phenanthryl, pyrenyl, or the like.
The term "heteroaryl" denotes an aromatic monocyclic or polycyclic ring system containing a 5-12 membered structure, or preferably a 5-10 membered structure, a 5-8 membered structure, more preferably a 5-6 membered structure, wherein 1,2, 3 or more ring atoms are heteroatoms and the remaining atoms are carbon, the heteroatoms are independently selected from O, N or S, the number of heteroatoms is preferably 1,2 or 3. Examples of heteroaryl groups include, but are not limited to, furyl, thienyl, oxazolyl, thiazolyl, isoxazolyl, oxadiazolyl, thiadiazolyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, thiadiazolyl, triazinyl, phthalazinyl, quinolinyl, isoquinolinyl, pteridinyl, purinyl, indolyl, isoindolyl, indazolyl, benzofuranyl, benzothienyl, benzopyridyl, benzopyrimidinyl, benzimidazolyl, phthalizinyl, pyrrolo [ 4232 zft 4232-b ] pyridyl, imidazo [ 4234 zft 4234-a ] pyridyl, pyrazolo [ 5364 zft 5364-86564-a ] pyridyl, pyrazolo [ 8652 zxft 2-a ] pyrimidinyl, imidazo [1,2-b ] pyridazinyl, [ 3579 z3579 zft 3579 ] triazolo [ 3525 xft 3265-b ] pyridazinyl, [ 353579 zft 3735 ] triazolo [ 3527-b ] pyridazinyl, [ 3735 ] triazolo [ 385256-b ] pyrimidinyl, and the like.
The terms "pharmaceutically acceptable salt" or "pharmaceutically acceptable salt" refer to salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of mammals, especially humans, without excessive toxicity, irritation, allergic response, and the like, and are commensurate with a reasonable benefit/risk ratio.
The term "salt" encompasses salts prepared from inorganic acids. Pharmaceutically acceptable non-toxic bases include salts prepared with inorganic and organic bases if the compounds of the present application are acidic.
The term "stereoisomer" refers to isomers resulting from the different arrangement of atoms in a molecule, including configurational isomers and conformational isomers, wherein configurational isomers include geometric isomers (or cis-trans isomers) and optical isomers (including enantiomers and diastereomers).
Geometric isomers may be present in the present compounds. The compounds of the present application may contain a carbon-carbon double bond or a carbon-nitrogen double bond in either the E or Z configuration, where the term "E" represents a more sequential substituent on the opposite side of the carbon-carbon or carbon-nitrogen double bond and the term "Z" represents a more sequential substituent on the same side of the carbon-carbon or carbon-nitrogen double bond (as determined using Cahn-Ingold Prelog priority rules). The compounds of the present application may also exist as mixtures of "E" and "Z" isomers. Substituents around a cycloalkyl or heterocycloalkyl group are referred to as cis or trans configurations.
Optical isomers refer to substances having completely the same molecular structure and similar physicochemical properties, but different optical rotation.
The compounds of the present application may contain asymmetrically substituted carbon atoms in either the R or S configuration, where the terms "R" and "S" are as defined in IUPAC 1974Recommendations for Section E, functional Stereochemistry, pure appl. Chem. (1976) 45,13-10. Compounds with asymmetrically substituted carbon atoms (with equal numbers of R and S configurations) are racemic at those carbon atoms. Having an excess of atoms of one configuration (relative to the other) allows the configuration to be present in higher amounts, preferably in an excess of about 85% to 90%, more preferably in an excess of about 95% to 99%, and even more preferably in an excess of greater than about 99%. Accordingly, the present application includes racemic mixtures, relative and absolute stereoisomers and mixtures of relative and absolute stereoisomers.
The term "prodrug" or "prodrug" is a derivative of an active drug that is designed to improve some defined, undesirable physical or biological property. Physical properties are often associated solubility (too high or insufficient lipid or water solubility) or stability, while problematic biological properties include too rapid metabolism or poor bioavailability, which may itself be associated with physicochemical properties.
Prodrugs are generally prepared as follows: a) forming esters, half-esters, carbonates, nitrates, amides, hydroxamic acids, carbamates, imines, mannich bases, phosphates and enamines of the active drug, b) functionalizing the drug with azo, glycoside, peptide and ether functionalities, c) using the aminal, hemiaminal, polymer, salt, complex, phosphoramide, acetal, hemiacetal and ketal forms of the drug. See, for example, andrejus Korolkovass's, "essences of Medicinal Chemistry", john Wiley-Interscience Pulications, john Wiley and Sons, new York (1988), pp.97-118, the contents of which are incorporated herein by reference in their entirety. Esters can be prepared from substrates containing hydroxyl or carboxyl groups using general methods known to those skilled in the art. Typical reactions for these compounds are substitutions in which one heteroatom is replaced by another atom. Amides can be prepared in a similar manner from substrates containing amino or carboxyl groups. Esters may also react with amines or ammonia to form amides. Another way to prepare the amide is to heat the carboxylic acid and the amine together.
The term "liver cancer" refers to any proliferative lesion or proliferative abnormality of the liver, for example, a malignancy derived from hepatocytes that account for a large portion of the liver, including all types of malignancies that begin within the liver and metastatic liver cancer that spreads elsewhere to the liver.
The term "breast cancer" or "breast tumor" refers to any proliferative lesion or proliferative abnormality of the breast, including, for example, benign lesions, premalignant and malignant lesions, solid tumors, and metastatic disease (local metastases, e.g., stage III, and more extensive metastases, e.g., stage IV), including but not limited to triple negative breast cancer.
The term "prostate cancer" or "prostate tumor" refers to any proliferative lesion or proliferative abnormality of the prostate gland that develops in the prostate gland, is histologically or cytologically confirmed as prostate cancer or prostate tumor, including but not limited to primary prostate tumors and metastases of said primary prostate tumors (which may be located anywhere in the body).
The term "treating" means administering a compound or formulation described herein to ameliorate or eliminate a disease or one or more symptoms associated with the disease, and includes: (ii) (i) inhibiting the disease or disease state, i.e., arresting its development; (ii) Alleviating the disease or condition, i.e., causing regression of the disease or condition.
The term "therapeutically effective amount" means an amount of a compound of the present application that (i) treats a particular disease, condition, or disorder, or (ii) reduces, ameliorates, or eliminates one or more symptoms of a particular disease, condition, or disorder. The amount of a compound of the present application that constitutes a "therapeutically effective amount" varies depending on the compound, the disease state and its severity, the mode of administration, and the individual characteristics to be treated (e.g., sensitivity, weight, age, etc.), but can be routinely determined by those skilled in the art with their own knowledge and this disclosure.
The term "pharmaceutical composition" refers to a mixture of one or more pharmaceutically acceptable salts of the compounds of the present application, stereoisomers or prodrugs thereof, and pharmaceutically acceptable excipients. The purpose of the pharmaceutical composition is to facilitate the administration of a pharmaceutically acceptable salt of a compound of the present application, a stereoisomer, or a prodrug thereof, to an organism. The pharmaceutical composition of the present invention can be prepared by a conventional method in the art.
In the context of the present application, the terms "pharmaceutically acceptable carrier" or "excipient" or "pharmaceutically acceptable adjuvant" refer to those adjuvants which do not have a significant irritating effect on the organism and do not impair the biological activity and performance of the active compound. The term "pharmaceutically acceptable excipients" includes: solvents, propellants, solubilizers, emulsifiers, colorants, adhesives, disintegrants, fillers, lubricants, wetting agents, osmotic pressure regulators, stabilizers, glidants, flavoring agents, preservatives, suspending agents, coating materials, fragrances, anti-adherents, antioxidants, chelating agents, permeation promoters, pH regulators, buffers, plasticizers, surfactants, foaming agents, antifoaming agents, thickeners, encapsulating agents, humectants, absorbents, diluents, flocculants and deflocculants, filter aids, release retardants, and the like. The skilled person can select specific pharmaceutically acceptable excipients according to the actual need. Knowledge about adjuvants is well known to the person skilled in the art, for example from pharmacy (eds Cui Fude, 5 th edition, national institutes of health, 2003).
The words "comprise" or "comprise" and variations thereof such as "comprises" or "comprising," are to be understood in an open, non-exclusive sense, i.e., "including but not limited to.
Within the scope of the present application, the various options for any feature may be combined with the various options for other features to form many different embodiments. The present application is intended to include all possible embodiments consisting of various options for all technical features.
In addition to the specific embodiments listed below, the compounds of the present application may be prepared by other synthetic methods, including the specific embodiments listed below, embodiments formed by combinations thereof with other chemical synthetic methods, and equivalents thereof known to those skilled in the art, with preferred embodiments including, but not limited to, the examples of the present application.
The term "subject" or "subject" as used herein refers to a cell or mammal, such as a human, but may also be other mammals, such as domestic or laboratory animals and the like.
The above embodiments represent exemplary embodiments of the present application, but the present application is not limited to the above embodiments. In addition, the various features of the above embodiments of the present application may be combined with one another to form one or more new solutions, which also fall within the scope of the present application, as long as such new solutions are technically feasible.
The application has one or more of the following beneficial effects:
in order to prove that the compound shown in the formula (I) is a CDK9 inhibitor effective on solid tumors (such as liver cancer, prostate cancer and breast cancer), the application evaluates the in vitro kinase inhibition activity of the compound shown in the formula (I) on different subtypes of CDKs and the proliferation inhibition activity of various liver cancer, breast cancer and prostate cancer cell strains, and further evaluates the tumor growth inhibition effect of the compound shown in the formula (I) (particularly the compound 45) on a liver cancer tumor xenograft model.
In vitro kinase activity test and cell test results show that the compound has good in vitro kinase inhibition activity on CDK9 and good selectivity on other CDK subtypes; has strong inhibition effect on various liver cancer cell lines, prostate cancer cell lines and breast cancer cell lines.
In vivo test results show that compared with a contrast compound, the compound has a better in vivo anti-tumor effect, is good in tolerance and higher in possibility of patent medicine, and provides a better choice for CDK9 target inhibition medicines.
In vitro and in vivo safety test results show that compared with a control compound, the compound shown in the formula (I) has no obvious inhibitory activity on an in vitro hERG channel in a detection concentration range, which indicates that the compound has lower cardiotoxicity risk, and the compound is taken by a mouse for 7 days continuously, and no animal death occurs along with increasing of dose, so that the compound has good animal tolerance.
The development of the compounds of the application expands the selection of drugs for treating cancers.
Examples
The present application is further illustrated with reference to specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present application. The experimental procedures, in which specific conditions are not noted in the following examples, are generally carried out under conventional conditions or under conditions recommended by the manufacturers. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In addition, any methods and materials similar or equivalent to those described herein can be used in the methods of the present application. The preferred embodiments and materials are shown herein for illustrative purposes only.
Example 1:
synthesis of intermediate 1 a:
4-bromo-5-chloropyridin-2-amine (3.00g, 14.50mmol) was dissolved in ethylene glycol dimethyl ether (50 mL) and water (10 mL), followed by addition of 4-fluoro-2-methoxyphenylboronic acid (2.50g, 14.70mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (1.06g, 1.45mmol) and potassium carbonate (6.00g, 44.10mmol), and substitution with nitrogen gas three times to place the entire system under an atmosphere of nitrogen gas. The system was stirred at 100 ℃ under reflux, reacted for 4 hours, and TLC monitored that no starting material remained. Concentrated under reduced pressure and purified by column chromatography (dichloromethane: methanol =50, 1-10) to give 1a (3.11 g, 85% yield).
Synthesis of intermediate 1 b:
1a (0.89g, 3.50mmol) was dissolved in N, N-dimethylformamide (30 mL), followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclopentanecarboxylic acid (0.85g, 3.50mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (1.60g, 4.20mmol) and N, N-diisopropylethylamine (0.91g, 7.00mmol), the whole was stirred at room temperature overnight, and TLC monitored for no residue of starting material. Water (100 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50: 1-20.
Synthesis of intermediate 1 c:
dissolve 1b (0.96g, 2.07mmol) in dichloromethane (30 mL), then add trifluoroacetic acid (2 mL) in an ice bath, stir the whole at room temperature overnight, TLC monitor until no starting material remains. Water (100 mL) was added to the reaction solution, followed by adjustment of PH =9-10 with a saturated aqueous solution of sodium bicarbonate, extraction with dichloromethane (50 mL × 3), combination of organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, removal of the solvent under reduced pressure, and purification by column chromatography (dichloromethane: methanol =50, 1-8:1) to obtain 1c (0.66 g, 88% yield).
Synthesis of final product 1:
1c (0.66g, 1.80mmol) was dissolved in dichloromethane (35 mL) followed by the addition of acetic anhydride (0.92g, 9)00 mmol) and triethylamine (0.91g, 9.00mmol). The system was stirred at room temperature and monitored by TLC until no starting material remained. Water (50 mL) was added to the reaction solution, dichloromethane was extracted (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and column chromatography separation and purification (petroleum ether: ethyl acetate =10: 1-2:1) gave final product 1 (0.48 g, 64% yield). MS m/z (ESI) 406.1[ 2 ] M + H] + 。
1 H NMR(600MHz,CDCl 3 )δ9.16(s,1H),8.28-8.25(m,2H),7.55(s,1H),7.18(d,J=7.2Hz,1H),6.79-6.71(m,2H),4.44(s,1H),3.80(s,3H),2.98(t,J=4.8Hz,1H),2.20-2.17(m,3H),1.97(s,3H),1.88-1.82(m,3H).
Example 2:
synthesis of intermediate 2 a:
5-fluoro-4-iodopyridin-2-amine (1.00g, 4.20mmol) was dissolved in ethylene glycol dimethyl ether (20 mL) and water (4 mL), followed by addition of 4-fluoro-2-methoxyphenylboronic acid (0.71g, 4.20mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (0.31g, 0.42mmol) and potassium carbonate (1.70g, 12.60mmol), and substitution with nitrogen gas was carried out three times to place the whole system under an atmosphere of nitrogen gas. The system was stirred at 100 ℃ under reflux, reacted for 4 hours, and no starting material was left as monitored by TLC. Concentrated under reduced pressure and purified by column chromatography (dichloromethane: methanol =50, 1-10) to give 2a (0.80 g, 81% yield).
Synthesis of intermediate 2 b:
2a (0.80g, 3.40mmol) was dissolved in N, N-dimethylformamide (30 mL), followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclopentanecarboxylic acid (0.83g, 3.40mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (1.56g, 4.10mmol) and N, N-diisopropylethylamine (0.88g, 6.80mmol), the whole was stirred at room temperature overnight, and TLC detected that no starting material remained. To the reaction solution was added water (100 mL), extracted with ethyl acetate (50 mL × 3), and the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50 1-20) to obtain 2b (0.82 g, yield 54%).
Synthesis of intermediate 2 c:
dissolve 2b (0.82g, 1.83mmol) in dichloromethane (30 mL), then add trifluoroacetic acid (2 mL) in an ice-water bath, stir the whole system at room temperature overnight, TLC check until no starting material remains. To the reaction solution was added water (100 mL), followed by adjustment of PH =9-10 with saturated aqueous sodium bicarbonate solution, extraction with dichloromethane (50 mL × 3), combination of organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, removal of the solvent under reduced pressure, and purification by column chromatography (dichloromethane: methanol =50, 1-8:1) to obtain 2c (0.59 g, 93% yield).
Synthesis of final product 2:
dissolve 2c (0.59g, 1.70mmol) in dichloromethane (35 mL), then add acetic anhydride (0.87g, 8.50mmol) and triethylamine (0.86g, 8.50mmol). The system is stirred at room temperature and monitored by TLC tracking until no more starting material remains. To the reaction solution was added water (50 mL), extracted with dichloromethane (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (petroleum ether: ethyl acetate =10: 1-2:1) to obtain the final product 2 (0.42 g, yield 63%). MS m/z (ESI) 390.2[ deg. ] M + H] + 。
1 H NMR(600MHz,CDCl 3 )δ9.20(s,1H),8.32(s,1H),8.12(s,1H),7.30(d,J=6.6Hz,1H),6.82-6.75(m,3H),4.44(s,1H),3.84(s,3H),2.99(q,J=3.6Hz,1H),2.22-2.15(m,3H),1.99(s,3H),1.89-1.85(m,3H).
Example 3:
synthesis of intermediate 3 a:
2a (0.80g, 3.40mmol) was dissolved in N, N-dimethylformamide (30 mL), followed by the addition of cis-3- [ (tert-butoxycarbonyl) amino ] cyclohexylalkanecarboxylic acid (0.83g, 3.40mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (1.56g, 4.10 mmol) and N, N-diisopropylethylamine (0.88g, 6.80mmol), the whole system was stirred at room temperature overnight, and TLC monitored that no starting material remained. Water (100 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50: 1-20.
Synthesis of intermediate 3 b:
dissolving 3a (0.90g, 1.95mmol) in dichloromethane (30 mL), followed by addition of trifluoroacetic acid (2 mL) under ice bath, the whole system stirred at room temperature overnight, TLC monitored no remaining starting material, added water (100 mL) to the reaction solution, adjusted PH =9-10 with saturated aqueous sodium bicarbonate, extracted with dichloromethane (50 mL × 3), combined organic phases, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, removed solvent under reduced pressure, purified by column chromatography (dichloromethane: methanol =50, 1-8:1), yielded 3b (0.61 g, yield 87%).
Synthesis of final product 3:
3b (0.61g, 1.69mmol) was dissolved in dichloromethane (35 mL) followed by the addition of acetic anhydride (0.86g, 8.40mmol) and triethylamine (0.85g, 8.40mmol). The system was stirred at room temperature and no starting material was left as monitored by TLC. Water (50 mL) was added to the reaction solution, dichloromethane was extracted (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and separation and purification by column chromatography (petroleum ether: ethyl acetate =10: 1-2:1) gave the final product 3 (0.38 g, 56% yield). MS m/z (ESI) 404.2[ 2 ] M + H] + 。
1 H NMR(600MHz,CDCl 3 ) 1 H NMR(600MHz,CDCl 3 )δ8.88(s,1H),8.29(s,1H),8.10(s,1H),7.28(s,1H),6.77-6.72(m,3H),3.82(s,3H),2.52-2.49(m,1H),2.24-2.22(m,1H),2.00-1.95(m,4H),1.98(s,3H),1.48-1.38(m,3H),1.18-1.13(m,1H).
Example 4:
synthesis of intermediate 4 a:
1a (0.40g, 1.58mmol) was dissolved in N, N-dimethylformamide (30 mL) followed by addition of (1R, 3S) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.38g, 1.58mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (0.72g, 1.90mmol) and N, N-diisopropylethylamine (0.41g, 3.16mmol), the system was stirred at room temperature overnight and TLC monitored that no starting material remained. To the reaction solution was added water (100 mL), extracted with ethyl acetate (50 mL × 3), and the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50 1-20) to obtain 4a (0.45 g, yield 60%).
Synthesis of intermediate 4 b:
4a (0.45g, 0.94mmol) was dissolved in dichloromethane (30 mL) followed by the addition of 2mL of trifluoroacetic acid in an ice water bath and the whole was stirred at room temperature overnight with TLC detection until no starting material remained. To the reaction solution was added water (100 mL), followed by adjustment of PH =9-10 with saturated aqueous sodium bicarbonate solution, extraction with dichloromethane (50 mL × 3), combination of organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, removal of the solvent under reduced pressure, and purification by column chromatography (dichloromethane: methanol =50, 1-8:1) to obtain 4b (0.30 g, yield 85%).
Synthesis of end product 4:
4b (0.30g, 0.80mmol) was dissolved in dichloromethane (35 mL), followed by the addition of acetic anhydride (0.24g, 2.39mmol) and triethylamine (0.24g, 2.39mmol), the system was stirred at room temperature and TLC monitored that no starting material remained. To the reaction solution was added water (50 mL), extracted with dichloromethane (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50 1-10) to obtain a final product 4 (0.17 g, yield 51%). MS m/z (ESI) 420.14[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,CDCl 3 ) 1 H NMR(600MHz,DMSO-d6)δ10.69(s,1H),8.41(s,1H),8.05(s,1H),7.78-7.73(m,1H),7.27-7.22(m,1H),7.10-7.08(m,1H),6.91-6.88(m,1H),3.76(s,3H),3.57-3.54(m,1H),2.62-2.59(m,1H),1.86(d,J=12.6Hz,1H),1.76(s,6H),1.31-1.23(m,3H),1.07-1.05(m,1H).
Example 5:
synthesis of intermediate 5 a:
2a (0.80g, 3.40mmol) was dissolved in N, N-dimethylformamide (30 mL), followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.83g, 3.40mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (1.56g, 4.10mmol) and N, N-diisopropylethylamine (0.88g, 6.80mmol), the whole was stirred at room temperature overnight, and TLC monitored that no starting material remained. Water (100 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50: 1-20.
Synthesis of intermediate 5 b:
5a (0.90g, 1.95mmol) was dissolved in dichloromethane (30 mL) and trifluoroacetic acid (2 mL) was added under an ice water bath, and the whole was stirred at room temperature overnight with TLC detection until no starting material remained. To the reaction solution was added water (100 mL), followed by adjustment of PH =9-10 with saturated aqueous sodium bicarbonate solution, extraction with dichloromethane (50 mL × 3), combination of organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, removal of the solvent under reduced pressure, and purification by column chromatography (dichloromethane: methanol =50, 1-8:1) to obtain 5b (0.61 g, yield 87%).
Synthesis of final product 5:
5b (0.61g, 1.69mmol) was dissolved in dichloromethane (35 mL), followed by the addition of acetic anhydride (0.86g, 8.40mmol) and triethylamine (0.85g, 8.40mmol), the system was stirred at room temperature and monitored by TLC follow-up until no starting material remained. Water (50 mL) was added to the reaction solution, dichloromethane was extracted (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and separation and purification by column chromatography (petroleum ether: ethyl acetate =10: 1-2:1) gave final product 5 (0.38 g, 56% yield). MS m/z (ESI) 404.2[ 2 ] M + H] + 。
1 H NMR(600MHz,CD 3 OD)δ8.18(s,1H),8.09(s,1H),7.33-7.29(m,1H),6.94(d,J=10.8Hz,1H),6.84-6.81(m,1H),3.83(s,3H),3.76-3.72(m,1H),2.60-2.57(m,1H),2.06(d,J=12.0Hz,1H),
1.96-1.90(m,3H),1.93(s,3H),1.51-1.39(m,3H),1.24-1.21(m,1H).
Example 6:
synthesis of intermediate 6 a:
3,4-difluoro-2-methoxyphenylboronic acid (0.57g, 3.03mmol) was dissolved in dioxane (50 mL), followed by the addition of 5-fluoro-4-iodopyridin-2-amine (0.60g, 2.52mmol), tetrakistriphenylphosphine palladium (150mg, 0.13mmol) and potassium phosphate trihydrate (1.00g, 3.78mmol), the system warmed to 100 ℃ under nitrogen, reacted for 4 hours, and TLC follow-up monitored for no residue of starting material. The reaction was cooled to room temperature and the crude product was purified by column chromatography (petroleum ether: ethyl acetate =10: 1-1:1) to afford 6a (0.40 g, 52% yield).
Synthesis of intermediate 6 b:
(1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (348mg, 1.43mmol) was dissolved in dichloromethane (50 mL), followed by addition of pyridine (572mg, 7.24mmol) and thionyl chloride (300mg, 2.52mmol) and reaction at room temperature for 4 hours, and then 6a (400mg, 1.57mmol) was directly added to the above reaction solution. The reaction was continued overnight at room temperature and the starting material was monitored by TLC tracking as not remaining. To the reaction solution was added water (30 mL) and extracted with ethyl acetate (20 mL × 3), the organic phases were combined, washed with saturated sodium chloride (20 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and separated and purified by column chromatography (petroleum ether: ethyl acetate =10: 1-2:1) to obtain 6b (250 mg, yield 33%).
Synthesis of intermediate 6 c:
6b (250mg, 0.52mmol) was dissolved in dichloromethane (10 mL), followed by the addition of trifluoroacetic acid (1 mL) and reaction at room temperature for 1.5 h, with TLC follow-up monitoring no starting material remaining. To the reaction solution was added saturated sodium bicarbonate (30 mL), extracted with dichloromethane (20 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (20 mL. Times.2), dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure to give 6c (130 mg, 65% yield).
Synthesis of final product 6:
6c (130mg, 0.34mmol) was dissolved in dichloromethane (10 mL), followed by the addition of acetic anhydride (45mg, 0.44mmol), and triethylamine (44mg, 0.44mmol), reacted at room temperature for 1.5 h with TLC follow-up monitoring no starting material remaining. To the reaction solution was added saturated sodium bicarbonate (30 mL), extracted with dichloromethane (20 mL × 3), the organic phases were combined, washed with saturated sodium chloride (20 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50: 1-30. MS m/z (ESI) 422.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d6)δ10.68(s,1H),8.43(s,1H),8.20(s,1H),7.78(d,J=7.8Hz,1H),7.33(t,J=7.8Hz,1H),7.21(t,J=7.8Hz,1H),3.95(s,3H),3.59-3.54(m,1H),2.63-2.59(m,1H),1.88-1.84(m,1H),1.80-1.72(m,3H),1.78(s,3H),1.31-1.25(m,3H),1.10-1.04(m,1H).
Example 7:
synthesis of intermediate 7 a:
2-amino-5-fluoro-4-iodopyridine (0.50g, 2.10mmol) and 5-fluoro-2-ethoxyphenylboronic acid (0.46g, 2.50mmol) were dissolved in ethylene glycol dimethyl ether (10 mL) and water (2 mL), and [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (71mg, 0.10mmol), potassium carbonate (0.87g, 6.30mmol) were added. The entire system was placed under a nitrogen atmosphere by replacing with nitrogen three times. The system was reacted at 100 ℃ for 2 hours. TLC monitoring indicated no starting material remained. After cooling, the solvent was removed by concentration. The crude product was isolated and purified by column chromatography (petroleum ether: ethyl acetate =10, 1-2:1) to afford 7a (0.50 g, 95% yield).
Synthesis of intermediate 7 b:
the compound (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.29g, 1.20mmol) was dissolved in dichloromethane (10 mL), pyridine (395mg, 5.00mmol) and thionyl chloride (202mg, 1.70mmol) were added in an ice bath, and the system was reacted at room temperature for 2 hours, then concentrated, and the solvent and excess thionyl chloride were removed. Then dichloromethane (10 mL) and compound 7a (250mg, 1.00mmol) were added. The system was left to react overnight at room temperature. TLC monitoring indicated no starting material remained. The system was concentrated and the crude product was isolated and purified by column chromatography (petroleum ether: ethyl acetate =10: 1-1:1) to give 7b (100 mg, 21% yield).
Synthesis of intermediate 7 c:
7b (47mg, 0.10 mmol) was dissolved in dichloromethane (5 mL), and trifluoroacetic acid (2 mL) was added. The system was left to react at room temperature for 1 hour. TLC monitoring showed no starting material remaining. The system was concentrated to give 7c (50 mg, crude). The product was used in the next reaction without further purification.
Synthesis of final product 7:
7c (37mg, 0.10 mmol) was dissolved in methylene chloride (2 mL), and triethylamine (20mg, 0.20mmol) and acetic anhydride (20mg, 0.20mmol) were added. The system was left to react at room temperature for 1 hour. TLC monitoring indicated no starting material remained. The system was concentrated, and the resulting crude product was isolated and purified by thin plate chromatography (dichloromethane: methanol =10: 1) to give the final product 7 (30 mg, yield 72%). MS (m/z, ESI) 417.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,DMSO-d6)δ10.57(s,1H),8.33(s,1H),8.11(d,J=5.4Hz,1H),7.79(d,J=7.8Hz,1H),7.35(d,J=7.8Hz,1H),7.09(d,J=11.4Hz,1H),6.90(d,J=7.8Hz,1H),4.10-4.07(m,2H),3.57-3.55(m,1H),2.59-2.57(m,1H),1.87-1.84(m,1H),1.77-1.75(m,5H),1.70-1.50(m,1H),1.31-1.26(m,3H),1.24-1.21(m,3H),1.07-1.05(m,1H).
Example 8:
synthesis of intermediate 8 a:
5-fluoro-4-iodopyridin-2-amine (1.00g, 4.20mmol) was dissolved in ethylene glycol dimethyl ether (20 mL) and water (4 mL), followed by addition of 4-fluoro-2-isopropoxyphenylboronic acid (0.83g, 4.20mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (0.31g, 0.42mmol) and potassium carbonate (1.74g, 12.60mmol), substitution three times with nitrogen, placing the whole system under an atmosphere of nitrogen, stirring the system at reflux at 100 ℃ for 4 hours, and TLC monitored that no starting material remained. Concentrated under reduced pressure and purified by column chromatography (dichloromethane: methanol =50, 1-10) to give 8a (0.85 g, 77% yield).
Synthesis of intermediate 8 b:
after 8a (0.85g, 3.20mmol) was dissolved in N, N-dimethylformamide (30 mL), and (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.78g, 3.20mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (1.44g, 3.80mmol) and N, N-diisopropylethylamine (0.83g, 6.40mmol) were added, the whole was stirred at room temperature overnight, and no residue was left by TLC. Water (100 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50: 1-20.
Synthesis of intermediate 8 c:
8b (0.90g, 1.84mmol) was dissolved in dichloromethane (30 mL) and trifluoroacetic acid (2 mL) was added under an ice water bath and the whole was stirred at room temperature overnight with TLC check until no starting material remained. To the reaction solution was added water (100 mL), followed by adjustment of PH =9-10 with saturated aqueous sodium bicarbonate solution, extraction with dichloromethane (50 mL × 3), combination of organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, and then removal of the solvent under reduced pressure, followed by column chromatography separation and purification (dichloromethane: methanol =50, 1-8:1) to obtain 8c (0.64 g, yield 89%).
Synthesis of final product 8:
8c (0.64g, 1.64mmol) was dissolved in dichloromethane (35 mL), followed by the addition of acetic anhydride (0.84g, 8.20mmol) and triethylamine (0.83g, 8.20mmol). The system was stirred at room temperature and monitored by TLC tracking until no starting material remained. To the reaction solution was added water (50 mL), extracted with dichloromethane (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and subjected to column chromatography separation and purification (petroleum ether: ethyl acetate =10: 1-2:1) to obtain the final product 8 (0.42 g, yield 59%). MS (m/z, ESI) 431.2[ deg. ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.57(s,1H),8.32(s,1H),8.12(s,1H),7.78(d,J=7.8Hz,1H),7.34(d,J=7.2Hz,1H),7.10(s,1H),6.88(d,J=8.4Hz,1H),4.72-4.69(m,1H),3.56(s,1H),2.62-2.58(m,1H),1.78(s,6H),1.31-1.23(m,4H),1.20(s,6H),1.09-1.04(m,1H).
Example 9:
synthesis of intermediate 9 a:
1-bromo-2-difluoromethoxy-4-fluorobenzene (1.00g, 4.15mmol), bis (pinacolato) borate (1.26g, 4.98mmol), potassium acetate (1.22g, 12.45mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (0.24g, 0.33mmol), ethylene glycol dimethyl ether (30 mL) were added to a reaction flask, heated to 100 ℃ under nitrogen for 8 hours, and TLC monitored for no material remaining. The heating was stopped and the temperature was reduced to room temperature. Concentrated under reduced pressure, and separated by column chromatography (n-hexane: ethyl acetate = 10) to obtain 9a (0.50 g, yield 42%).
Synthesis of intermediate 9 b:
9a (0.50g, 1.74mmol), 5-fluoro-4-iodo-pyridin-2-amine (0.33g, 1.39mmol), palladium tetrakistriphenylphosphine (0.12g, 0.10mmol), tripotassium phosphate trihydrate (0.60g, 2.26mmol), dioxane (30 mL) was added to the reaction flask, heated to 100 ℃ under nitrogen protection for 8 hours, and TLC monitored that no starting material remained. Stopping heating, and cooling to room temperature. Concentrated under reduced pressure and separated by column chromatography (n-hexane: ethyl acetate = 1:1) to give 9b (0.39 g, 83% yield).
Synthesis of intermediate 9 c:
9b (0.39g, 1.43mmol), (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.35g, 1.43mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (0.65g, 1.72mmol), N, N-diisopropylethylamine (0.37g, 2.86mmol) and N, N-dimethylformamide (20 mL) were added to a reaction flask, reacted at room temperature for 15 hours, and TLC monitored that no starting material remained. Water (30 mL) was added to the reaction solution, extracted with ethyl acetate (30 mL × 3), the organic phases were combined, washed with saturated sodium chloride (30 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and separated and purified by column chromatography (n-hexane: ethyl acetate = 1:1) to obtain 9c (0.16 g, yield 23%).
Synthesis of intermediate 9 d:
9c (0.16g, 0.33mmol) was dissolved in dichloromethane (20 mL), trifluoroacetic acid (4 mL) was added and the reaction was carried out at room temperature for 4 hours, and TLC monitored for no residue of starting material. The reaction solution was washed with water (20 mL. Times.3), the aqueous phases were combined, the pH of the aqueous phase was adjusted to 8-9 with sodium carbonate, dichloromethane was extracted (30 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure to give 9d (0.10 g, 76% yield).
Synthesis of final product 9:
9d (0.10g, 0.26mmol) was dissolved in methylene chloride (20 mL), acetic anhydride (0.05g, 0.52mmol) and triethylamine (0.05g, 0.52mmol) were added to the solution, and the mixture was reacted at room temperature for 2 hours, and TLC was performed to monitor that no residue was left. The reaction was directly subjected to column chromatography (dichloromethane: methanol =25 = 1) to obtain the final product 9 (0.05 g, yield 44%). MS m/z (ESI) 440.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.67(s,1H),8.41(s,1H),8.12(d,J=5.4Hz,1H),7.78(d,J=7.2Hz,1H),7.58-7.56(m,1H),7.42(s,1H),7.34-7.28(m,1H),3.57-3.56(m,1H),2.61-2.59(m,1H),1.89-1.78(m,4H),1.77(s,3H),1.31-1.24(m,3H),1.08-1.06(m,1H).
Example 10:
synthesis of intermediate 10 a:
5-fluoro-4-iodopyridin-2-amine (500mg, 2.10mmol) was dissolved in a mixed solvent of DME (20 mL) and water (4 mL), followed by addition of 2-benzyloxy-4-fluorobenzeneboronic acid (620mg, 2.52mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (90mg, 0.11mmol) and potassium carbonate (870mg, 6.30mmol), and substitution with nitrogen gas was carried out three times to subject the whole system to an atmosphere of nitrogen gas. The system was stirred at 100 ℃ under reflux, reacted for 4 hours, and TLC monitored complete reaction of starting materials. The reaction was directly purified by column chromatography (petroleum ether: ethyl acetate = 5:1-2:1) to afford 10a (0.50 g, 76% yield).
Synthesis of intermediate 10 b:
10a (0.30g, 0.96mmol) was dissolved in N, N-dimethylformamide (30 mL) followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.25g, 1.01mmol), 2- (7-oxybenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (0.43g, 1.15mmol) and N, N-diisopropylethylamine (0.25g, 1.92mmol), the system was stirred at room temperature overnight with TLC monitoring no residue of starting material. Water (100 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (petroleum ether: ethyl acetate =10: 1-2:1) to give 10b (0.30 g, 58% yield).
Synthesis of intermediate 10 c:
10b (0.30g, 0.56mmol) was dissolved in dichloromethane (30 mL) and trifluoroacetic acid (2 mL) was added under an ice-water bath, the system was stirred at room temperature for 2 hours and TLC monitored that no starting material remained. To the reaction solution was added water (100 mL), followed by adjustment of PH =9-10 with a saturated aqueous solution of sodium bicarbonate, extraction with dichloromethane (50 mL × 3), combination of organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, and then removal of the solvent under reduced pressure, and the crude product was subjected to column chromatography separation purification (dichloromethane: methanol =50, 1-8:1) to give 10c (0.20 g, 82% yield).
Synthesis of final product 10:
10c (0.20g, 0.46mmol) was dissolved in methylene chloride (10 mL), and acetic anhydride (94mg, 0.92mmol) and triethylamine (93mg, 0.92mmol) were added to react at room temperature for 1.5 hours, and TLC (ethyl acetate) was used to monitor the completion of the reaction of the raw materials. To the reaction solution was added saturated sodium bicarbonate (30 mL), extracted with dichloromethane (20 mL × 3), the organic phases were combined, washed with saturated sodium chloride (20 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (dichloromethane: methanol =50: 1-20. MS m/z (ESI) 480.2[ deg. ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.56(s,1H),8.32(s,1H),8.14(d,J=5.4Hz,1H),7.76(d,J=7.8Hz,1H),7.37(dd,J=7.8Hz,J=7.2Hz,1H),7.32-7.27(m,5H),7.16(d,J=11.4Hz,1H),6.92(dd,J=8.4Hz,J=8.4Hz,1H),5.15(s,2H),3.55-3.54(m,1H),2.58-2.56(m,1H),1.86-1.84(m,1H),1.75-1.70(m,6H),1.33-1.15(m,4H).
Example 11:
synthesis of intermediate 11 a:
5 (0.10g, 0.25mmol) was dissolved in methylene chloride (20 mL), boron tribromide (0.12g, 0.50mmol) was added, the reaction was carried out at room temperature for 4 hours, and the starting material was monitored by TLC to be not remained. The reaction solution was adjusted to pH 6 with an aqueous sodium bicarbonate solution, the organic phase was separated and dried over anhydrous sodium sulfate. The solvent was distilled off under reduced pressure to give 11a (0.08 g, yield 82%).
Synthesis of final product 11:
11a (0.08g, 0.21mmol) was dissolved in N, N-dimethylformamide (10 mL), and 2-bromoethyl methyl ether (0.03g, 0.25mmol), potassium carbonate (0.06g, 0.42mmol) were added and reacted at room temperature for 8 hours with TLC monitoring that no starting material remained. Water (20 mL) was added to the reaction mixture, extracted with ethyl acetate (20 mL. Times.3), and the organic phases were combined and dried over anhydrous sodium sulfate. The organic phase was subjected to column chromatography (dichloromethane: methanol =25 = 1) to obtain the final product 11 (0.05 g, yield 53%). MS m/z (ESI) 448.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.56(s,1H),8.31(s,1H),8.11-8.10(m,1H),7.78-7.58(m,1H),7.36-7.33(m,1H),7.12-7.01(m,1H),6.92-6.89(m,1H),4.15-4.13(m,2H),3.55-3.54(m,2H),3.53-3.52(m,1H),3.15(s,3H),2.59-2.58(m,1H),1.85(s,3H),1.83-1.47(m,4H),1.29-1.04(m,4H).
Example 12:
synthesis of intermediate 12 a:
2-amino-5-fluoro-4-iodopyridine (0.50g, 2.10mmol) and 5-chloro-2-methoxyphenylboronic acid (0.47g, 2.50mmol) were dissolved in ethylene glycol dimethyl ether (10 mL), and [1,1-bis (diphenylphosphino) ferrocene ] palladium dichloride (73mg, 0.10mmol), potassium carbonate (0.87g, 6.3mmol) and water (2 mL) were added. The entire system was placed under a nitrogen atmosphere by replacing with nitrogen three times. The system was reacted at 100 ℃ for 2 hours. TLC monitoring indicated no starting material remained. After cooling, the solvent was removed by concentration. The crude product was isolated and purified by column chromatography (petroleum ether: ethyl acetate =10, 1-2:1) to afford 12a (0.50 g, 95% yield).
Synthesis of intermediate 12 b:
(1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.29g, 1.20mmol) was dissolved in dichloromethane (10 mL), pyridine (0.40g, 5.00mmol) and thionyl chloride (0.20g, 1.70mmol) were added in an ice bath, and the system was reacted at room temperature for 2 hours, then concentrated, and the solvent and excess thionyl chloride were removed. Then dichloromethane (10 mL) and compound 12a (0.25g, 1.00mmol) were added. The system was left to react overnight at room temperature. TLC monitoring indicated no starting material remained. The system was concentrated and the crude product was isolated and purified by column chromatography (petroleum ether: ethyl acetate =10: 1-1:1) to afford 12b (0.10 g, 21% yield).
Synthesis of intermediate 12 c:
12b (48mg, 0.10 mmol) was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (2 mL) was added. The system was left to react at room temperature for 1 hour. TLC monitoring showed no starting material remaining. The system was concentrated to give 12c (50 mg, crude). The product was used in the next reaction without further purification.
Synthesis of end product 12:
12c (50mg, 0.10 mmol) was dissolved in dichloromethane (2 mL), and triethylamine (20mg, 0.20mmol) and acetic anhydride (20mg, 0.20mmol) were added. The system was left to react at room temperature for 1 hour. TLC monitoring indicated no starting material remained. The system was concentrated, and the resulting crude product was isolated and purified by thin plate chromatography (dichloromethane: methanol =10: 1) to give the final product 12 (30 mg, yield 72%). MS (m/z, ESI) 420.1[ 2 ], [ M ] +H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.60(s,1H),8.34(s,1H),8.08(d,J=5.4Hz,1H),7.78(d,J=7.8Hz,1H),7.33(d,J=8.4Hz,1H),7.27(s,1H),7.20-7.15(m,1H),3.80(s,3H),3.65-3.55(m,1H),2.68-2.62(m,1H),1.90-1.85(m,1H),1.76-1.55(m,6H),1.28-1.23(m,3H),1.15-1.10(m,1H).
Example 13:
synthesis of intermediate 13 a:
3-methoxy-4-bromophenol (3.00g, 14.80mmol) was dissolved in acetone (50 mL), followed by addition of bromomethylcyclopropane (2.20g, 16.30mmol), sodium iodide (1.11g, 7.40mmol), cesium carbonate (9.64g, 29.60mmol), stirring under reflux conditions, reaction for 8 hours, and TLC monitoring of the starting material without residue. Stopping heating, and cooling to room temperature. Acetone was evaporated under reduced pressure, water (50 mL) was added to the residue, extraction was performed with ethyl acetate (30 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, and the solvent was removed by concentration under reduced pressure to give 13a (3.70 g, yield 97%).
Synthesis of intermediate 13 b:
13a (3.20g, 12.40mmol) was dissolved in ethylene glycol dimethyl ether (50 mL), followed by addition of bisphenopinacol borate (3.79g, 14.90mmol), potassium acetate (3.65g, 37.2mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (0.88g, 1.2mmol), heating to 100 ℃ under nitrogen for 8 hours, and TLC monitoring for no material remaining. Stopping heating, and cooling to room temperature. The solvent was removed under reduced pressure and purified by column chromatography (n-hexane: ethyl acetate = 5:1) to give 13b (2.50 g, 66% yield).
Synthesis of intermediate 13 c:
13b (2.19g, 7.20mmol) was dissolved in dioxane (50 mL), followed by addition of 5-fluoro-4-iodo-pyridin-2-amine (1.38g, 5.80mmol), tetratriphenylphosphine palladium (0.46g, 0.40mmol) and potassium phosphate trihydrate (2.50g, 9.40mmol), heating to 100 ℃ under nitrogen protection for 8 hours, and TLC monitored that no starting material remained. Stopping heating, and cooling to room temperature. The solvent was removed by concentration under reduced pressure, and the residue was purified by column chromatography (n-hexane: ethyl acetate = 1:1) to give 13c (1.90 g, yield 92%).
Synthesis of intermediate 13 d:
13c (0.60g, 2.10mmol) was dissolved in N, N-dimethylformamide (20 mL), followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.51g, 2.10mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (0.95g, 2.50mmol) and N, N-diisopropylethylamine (0.54g, 4.20mmol), reaction at room temperature for 15 hours, and TLC monitoring of the absence of the remaining raw material. Water (50 mL) was added to the reaction solution, extracted with ethyl acetate (30 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and subjected to column chromatography for purification (n-hexane: ethyl acetate = 1:1) to obtain 13d (0.50 g, yield 46%).
Synthesis of intermediate 13 e:
13d (0.50g, 0.97mmol) was dissolved in dichloromethane (20 mL), trifluoroacetic acid (4 mL) was added and the reaction was allowed to proceed at room temperature for 4 hours, and TLC monitored that no starting material remained. The reaction solution was washed with water (30 mL. Times.3), the aqueous phases were combined, the pH of the aqueous phase was adjusted to 8-9 with sodium carbonate, dichloromethane was extracted (30 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), and dried over anhydrous sodium sulfate. The solvent was removed by concentration under reduced pressure to give 13e (0.35 g, yield 88%).
Synthesis of final product 13:
13e (0.06g, 0.14mmol) was dissolved in dichloromethane (20 mL), followed by addition of acetic anhydride (0.03g, 0.28mmol), triethylamine (0.03g, 0.28mmol), reaction at room temperature for 2h, and absence of residual starting material monitored by TLC. The reaction was directly subjected to column chromatography (DCM: meOH =25: 1) to give the final product 13 (0.02 g, 31% yield). MS m/z (ESI) 456.2[ deg. ] M + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.52(s,1H),8.28(s,1H),8.06(d,J=4.8Hz,1H),7.78(d,J=7.8Hz,1H),7.18(d,J=8.4Hz,1H),6.71(s,1H),6.63(d,J=8.4Hz,1H),3.89(d,J=7.2Hz,2H),3.76(s,3H),3.57-3.55(m,1H),2.61-2.60(m,1H),1.88-1.86(m,1H),1.77(s,3H),1.76-1.75(m,3H),1.31-1.23(m,4H),1.09-1.04(m,1H),0.60-0.59(m,2H),0.35-0.34(m,2H).
Example 14:
synthesis of intermediate 14 a:
4-bromo-3-methoxyphenol (0.40g, 2.00mmol) was dissolved in dioxane (50 mL), followed by the addition of bisphenopinacol diborane (0.60g, 2.40mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (0.12g, 0.16mmol) and potassium acetate (0.59g, 6.00mmol), the system was protected with nitrogen, stirred at 100 deg.C, reacted for 5 hours, and TLC monitored that the starting material was reacted. To the reaction solution was added water (100 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and separated and purified by column chromatography (petroleum ether-petroleum ether: ethyl acetate = 4:1) to give 14a (0.36 g, yield 72%).
Synthesis of intermediate 14 b:
14a (0.36g, 1.44mmol) was dissolved in diethylene glycol dimethyl ether (50 mL) and water (10 mL), followed by the addition of the compounds 5-fluoro-4-iodo-pyridin-2-amine (0.29g, 1.20mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (0.44g, 0.60mmol) and potassium carbonate (0.50g, 3.60mmol). The whole system was stirred at 80 ℃ for 5 hours and no starting material remained as detected by TLC. Water (100 mL) was added to the reaction mixture, ethyl acetate extraction (100 mL. Times.3) was performed, the organic phases were combined, washed with saturated sodium chloride (100 mL. Times.2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the mixture was purified by column chromatography (petroleum ether: ethyl acetate = 4:1-2:3) to obtain 14b (0.25 g, 74% yield)
Synthesis of intermediate 14 c:
14b (0.15g, 0.64mmol) was dissolved in N, N-dimethylformamide (50 mL), followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.18g, 0.72mmol), 2- (7-oxybenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (0.58g, 0.72mmol) and diisopropylethylamine (0.20mL, 1.20mmol) the entire system was stirred at 25 ℃ for 15 hours, and TLC monitored for no residue of starting material. Water (100 mL) was added to the reaction mixture, extraction was performed with ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (100 mL. Times.2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the mixture was separated and purified by column chromatography (petroleum ether: ethyl acetate =)
4:1-2:1) to yield 14c (0.15 g, 51% yield).
Synthesis of final product 14:
14c (0.15g, 0.32mmol) was dissolved in dichloromethane (50 mL), and trifluoroacetic acid (2 mL) was added thereto and stirred at room temperature for 4 hours, and no starting material remained by TLC. Saturated aqueous sodium carbonate solution was added to the system, the pH of the reaction system was adjusted to 9 to 10, methylene chloride was extracted (50 mL. Times.3), the combined organic phases were concentrated to a volume of about 50mL, triethylamine (2 mL) and acetic anhydride (2 mL) were added and the reaction was carried out at room temperature for 30 minutes, and then the organic phase was washed by adding aqueous sodium carbonate solution. The liquid was separated, extracted with dichloromethane (50 mL × 3), concentrated under reduced pressure, and separated by column chromatography (ethyl acetate) to obtain the final product 14 (90 mg, yield 63%). MS m/z (ESI) 444.2[ 2 ] M + H] + 。
1 H NMR(600MHz,CD 3 OD)δ8.16(s,1H),8.10(s,1H),7.29(d,J=7.8Hz,1H),6.87(s,1H),6.78(t,J=7.8Hz,1H),3.79(s,3H),3.78-3.73(m,1H),2.72(t,J=12.0Hz,1H),2.31-2.29(m,1H),2.20-2.10(m,1H),2.00-1.80(m,6H),1.51-1.38(m,3H),1.27-1.21(m,2H).
Example 15:
synthesis of intermediate 15 a:
3-methoxy-4-bromophenol (540mg, 2.66mmol) was dissolved in N, N-dimethylformamide (50 mL), followed by addition of potassium carbonate (735mg, 5.32mmol) and benzyl bromide (910mg, 5.32mmol) to react and stirred at room temperature for 15 hours, TLC detected that no raw material remained, water (50 mL) was added to the system, the organic phase was extracted with ethyl acetate (50 mL. Times.3), washed with a saturated aqueous sodium chloride solution (50 mL. Times.2), dried over anhydrous sodium sulfate, concentrated under reduced pressure, and purified by column chromatography (petroleum ether-petroleum ether: ethyl acetate = 4:1) to give 15a (662 mg, 85% yield).
Synthesis of intermediate 15 b:
compound 15a (662mg, 2.27mmol), bis-pinacoldiborane (1.15g, 4.54mmol), potassium acetate (667mg, 6.81mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (146mg, 0.20mmol) were dissolved in anhydrous 1,4-dioxane (100 mL). The reaction was protected with nitrogen and reacted at 100 ℃ for 4 hours, and no starting material remained as determined by TLC. To the reaction system was added water (100 mL), extracted with ethyl acetate (100 mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, concentrated under reduced pressure to remove the solvent, and purified by column chromatography (petroleum ether-petroleum ether: ethyl acetate = 4:1) to give 15b (510 mg, yield 65%).
Synthesis of intermediate 15 c:
15b (510mg, 1.50mmol) was dissolved in diethylene glycol dimethyl ether (150 mL), 5-fluoro-4-iodopyridin-2-amine (536mg, 2.25mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (110mg, 0.15mmol) and potassium carbonate (621mg, 4.50mmol) were added at room temperature, the reaction was stirred at 80 ℃ for 4 hours, and no starting material remained as detected by TLC. Water (100 mL) was added to the reaction system, extracted with ethyl acetate (100 mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (petroleum ether-petroleum ether: ethyl acetate = 1:1) to give 15c (263 mg, yield 54%).
Synthesis of intermediate 15 d:
15c (263mg, 0.81mmol) and (1S, 3R) -3- [ (t-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (290mg, 1.23mmol) were dissolved in N, N-dimethylformamide (20 mL), and 2- (7-oxybenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (470mg, 1.23mmol) and diisopropylethylamine (480mg, 3.69mmol) were added in this order, and the reaction system was reacted at room temperature for 4 hours with no remaining starting material by TLC. To the reaction system was added water (100 mL), extracted with ethyl acetate (100 mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (petroleum ether: ethyl acetate = 4:1-2:1) to give 15d (250 mg, yield 56%).
Synthesis of intermediate 15 e:
15d (250mg, 0.45mmol) was dissolved in dichloromethane (50 mL), followed by the addition of trifluoroacetic acid (5 mL) and stirring at room temperature for 4 hours with TLC detection of no starting material remaining. Adding saturated sodium carbonate aqueous solution into the system, and adjusting the pH value of the reaction system to 9-10. Separating, extracting with dichloromethane (100 mL. Times.3), combining the organic phases, concentrating to a volume of about 50mL, adding triethylamine (2 mL) and acetic anhydride (2 mL) to react at room temperature for 30 minutes, adding aqueous sodium carbonate to wash the organic phases, extracting with dichloromethane (20 mL. Times.3), combining the organic phases, drying over anhydrous sodium sulfate, removing the solvent under reduced pressure, and separating by column chromatography (ethyl acetate) to give 15e (176 mg, 79% yield)
Synthesis of final product 15:
15e (175mg, 0.36mmol) was dissolved in methanol (20 mL), palladium on carbon (10 mg) was added, the reaction system was protected with hydrogen, reacted at room temperature for 10 hours, LC-MS showed no remaining starting material, and after palladium on carbon was filtered off, the reaction solution was concentrated under reduced pressure to obtain the final product 15 (117 mg, yield 81%). MS m/z (ESI) 402.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.49(s,1H),10.09(s,1H),8.25(s,1H),8.05(s,J=5.4Hz,1H),7.78(d,J=7.8Hz,1H),7.07(d,J=8.4Hz,1H),6.55(d,J=2.4Hz,1H),6.48(s,1H),3.71(s,3H),3.55-3.53(m,1H),2.61-2.57(m,1H),1.87(d,J=11.4Hz,1H),1.77(s,3H),1.77-1.75(m,3H),1.29-1.27(m,4H).
Example 16:
synthesis of intermediate 16 a:
2- (3,4-dimethoxyphenyl) -4,4,5,5-tetramethyl-1,3,2-dioxaborane (150mg, 0.57mmol) and 5-fluoro-4-iodopyridin-2-amine (202mg, 0.85mmol) were dissolved in diethylene glycol dimethyl ether (50 mL), and [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (37mg, 0.05mmol), potassium carbonate (117mg, 0.85mmol) and water (10 mL) were added in this order, and the reaction was reacted at 80 ℃ for 4 hours with nitrogen protection, and no residue was left by TLC. To the reaction mixture was added water (50 mL), extracted with ethyl acetate (100 mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (petroleum ether: ethyl acetate = 4:1-1:1) to give 16a (124 mg, yield 88%).
Synthesis of intermediate 16 b:
compound 16a (36mg, 0.15mmol) was dissolved in N, N-dimethylformamide (10 mL), followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (71mg, 0.29mmol), 2- (7-benzotriazol oxide) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (110mg, 0.29mmol) and diisopropylethylamine (57mg, 0.44mmol), the whole system was stirred at 25 ℃ for 15 hours, and TLC monitored that no starting material remained. To the reaction solution was added water (15 mL), extracted with ethyl acetate (20 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (petroleum ether: ethyl acetate = 4:1-2:1) to give 16b (35 mg, yield 49%).
Synthesis of final product 16:
16b (35mg, 0.074mmol) was dissolved in dichloromethane (20 mL) followed by trifluoroacetic acid (2 mL) and stirred at room temperature for 4 hours with no starting material remaining as detected by TLC. To the system was added saturated aqueous sodium carbonate solution, the pH of the reaction system was adjusted to 9-10, extracted with dichloromethane (50 mL. Times.3), the organic phases were combined, concentrated to a volume of about 20mL, and triethylamine (2 mL) and acetic anhydride (2 mL) were added. The reaction was carried out at room temperature for 30 minutes, and the organic phase was washed by adding an aqueous solution of sodium carbonate. The aqueous phase was extracted with dichloromethane (20 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and separated by silica gel column chromatography (ethyl acetate) to give the final product 16 (15 mg, 49% yield). MS m/z (ESI) 416.2[ 2 ], [ M ] +H] + 。
1 H NMR(600MHz,CD 3 OD)δ8.12(s,1H),8.08(d,J=5.4Hz,1H),7.19(d,J=8.4Hz,1H),6.64(s,1H),6.60(s,1H),3.88(s,3H),3.79(s,3H),3.71(t,J=4.8Hz,2H),2.71-2.57(m,1H),2.24-2.22(m,1H),1.91(s,3H),1.89(s,2H),1.49-1.19(m,6H).
Example 17:
synthesis of intermediate 17 a:
2-Methoxyphenylboronic acid (0.42g, 2.77mmol) was dissolved in diethylene glycol dimethyl ether (30 mL) and water (6 mL), followed by the addition of the compounds 5-fluoro-4-iodopyridin-2-amine (0.60g, 2.52mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (95mg, 0.13mmol) and potassium carbonate (1.04g, 7.56mmol). The system was stirred at 100 ℃ for 4 hours under nitrogen protection, and no residue was left after TLC monitoring. To the reaction system was added water (50 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (petroleum ether: ethyl acetate = 4:1-2:3) to give 17a (0.60 g, yield 99%).
Synthesis of intermediate 17 b:
17a (0.60g, 2.77mmol) was dissolved in N, N-dimethylformamide (50 mL), followed by addition of the compounds (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.79g, 3.20mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (1.22g, 3.20mmol) and diisopropylethylamine (1.2mL, 7.00mmol), and the whole was stirred at room temperature for 15 hours with TLC detection of no remaining starting material. To the reaction system was added water (100 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (petroleum ether: ethyl acetate = 4:1-2:3) to give 17b (0.60 g, yield 49%).
Synthesis of intermediate 17 c:
17b (0.60g, 1.35mmol) was dissolved in dichloromethane (50 mL) followed by trifluoroacetic acid (3 mL) and the entire system was stirred at 25 ℃ and followed by TLC until no starting material remained. To the reaction solution was added aqueous sodium carbonate (50 mL), extracted with dichloromethane (50 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure, and the crude product 17c was directly subjected to the next reaction.
Synthesis of end product 17
Dissolve 17c in dichloromethane (50 mL), then add triethylamine (0.4 mL, 2.70mmol) and acetic anhydride (0.3 mL, 2.70mmol) and stir at room temperature for 30 minutes, TLC detects the completion of the starting material reaction. Adding saturated sodium carbonate into the systemAqueous solution, dichloromethane extraction (50 mL × 3), combined organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, removal of solvent under reduced pressure, and purification by column chromatography (petroleum ether: ethyl acetate = 4:1-2:1) to give final product 17 (0.35 g, 68% yield). MS m/z (ESI) 386.2[ deg. ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.55(s,1H),8.30(s,1H),8.07(d,J=5.4Hz,1H),7.76(d,J=7.8Hz,1H),7.46(t,J=7.8Hz,1H),7.27(d,J=7.2Hz,1H),7.15(d,J=8.4Hz,1H),7.06(t,J=7.8Hz,1H),3.75(s,3H),3.55-3.54(m,1H),2.56-2.48(m,1H),1.86-1.84(m,1H),1.77-1.74(m,6H),1.31-1.02(m,4H).
Example 18:
synthesis of intermediate 18 a:
4- (bromomethyl) pyridine hydrochloride (1.72g, 6.81mmol) was dissolved in 20mL of N, N-dimethylformamide, followed by addition of 2-iodophenol (1.50g, 6.81mmol), potassium carbonate (2.83g, 20.45mmol), and sodium iodide (1.02g, 6.81mmol), the system was stirred at room temperature for 4 hours, and TLC monitored that no starting material remained. The reaction was directly purified by column chromatography (petroleum ether: ethyl acetate = 2:1-petroleum ether: ethyl acetate = 1:1) to give 18a (1.90 g, 90% yield).
Synthesis of intermediate 18 b:
18a (1.00g, 3.21mmol) was dissolved in 15mL of ethylene glycol dimethyl ether, followed by addition of bis pinacol borate (980mg, 3.85mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (234mg, 0.32mmol) and potassium acetate (0.95g, 9.64mmol), and displacement with nitrogen was carried out three times to leave the whole system under an atmosphere of nitrogen. The system was stirred at 100 ℃ under reflux, reacted for 4 hours, and TLC monitored that no starting material remained. The reaction was directly purified by column chromatography (petroleum ether: ethyl acetate = 2:1-petroleum ether: ethyl acetate = 1:1) to give compound 18b (0.80 g, 80% yield).
Synthesis of intermediate 18 c:
5-fluoro-4-iodopyridin-2-amine (802mg, 3.37mmol) was dissolved in 20mL of ethylene glycol dimethyl ether, followed by addition of 18b (700mg, 2.25mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (168mg, 0.23mmol) and potassium carbonate (932mg, 6.75mmol), and displacement with nitrogen was carried out three times to leave the whole system under an atmosphere of nitrogen. The system was stirred at 100 ℃ under reflux, reacted for 4 hours, and TLC monitored that no starting material remained. The reaction was directly purified by column chromatography (dichloromethane: methanol = 50-1-dichloromethane: methanol = 10) to give 18c (220 mg, 33% yield).
Synthesis of intermediate 18 d:
18c (220mg, 0.75mmol) was dissolved in 10mL of N, N-dimethylformamide, followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (217mg, 0.89mmol), 2- (7-oxybenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (340mg, 0.89mmol) and N, N-dimethylethylamine (192mg, 1.49mmol), and the whole was stirred at room temperature overnight with no remaining starting material by TLC. To the reaction solution, 100mL of water was added, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol = 50-dichloromethane: methanol = 20).
Synthesis of intermediate 18 e:
18d (0.30g, 0.58mmol) was dissolved in 30mL of dichloromethane, followed by addition of 2mL of trifluoroacetic acid in an ice-water bath, and the whole was stirred at room temperature overnight with TLC until no starting material remained. To the reaction solution, 100mL of water was added, followed by adjustment of PH =9-10 with a saturated aqueous solution of sodium bicarbonate, extraction with dichloromethane (50 mL × 3), combination of organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, and then removal of the solvent under reduced pressure gave 18e (0.16 g, 66% yield) after purification by column chromatography (dichloromethane: methanol = 50-dichloromethane: methanol = 8:1).
Synthesis of end product 18:
18e (160mg, 0.38mmol) was dissolved in dichloromethane (5 mL), followed by the addition of acetic anhydride (116mg, 1.14mmol) and triethylamine (115mg, 1.14mmol). The system is stirred at room temperature,follow-up by TLC until no starting material remained. To the reaction solution, 50mL of water was added, dichloromethane was extracted (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and column chromatography separation and purification (petroleum ether: ethyl acetate = 10-petroleum ether: ethyl acetate = 2:1) gave the final product 18 (60 mg, yield 34%). MS m/z (ESI) 463.2[ m + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.61(s,1H),8.50(d,J=5.4Hz,2H),8.36(s,1H),8.20(d,J=5.4Hz,1H),7.80(d,J=7.8Hz,1H),7.47-7.44(m,1H),7.35(d,J=7.2Hz,1H),7.30(d,J=5.4Hz,2H),7.18(d,J=9.0Hz,1H),7.15(t,J=7.8Hz,1H),5.22(s,2H),3.58-3.57(m,1H),2.62-2.53(m,1H),1.89-1.75(m,7H),1.30-1.04(m,4H).
Example 19:
synthesis of intermediate 19a
7-bromobenzofuran (600mg, 3.00mmol) was dissolved in ethylene glycol dimethyl ether (50 mL), followed by the addition of pinacol diboron ester (928mg, 3.70mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (163mg, 0.20mmol) and potassium acetate (892mg, 9.10mmol), the system was warmed to 100 ℃ under nitrogen protection for 4 hours, and TLC monitored for complete reaction of the starting material. The reaction was cooled to room temperature, and the crude product was purified by column chromatography (petroleum ether: ethyl acetate = 100.
Synthesis of intermediate 19b
19a (450mg, 1.84mmol) was dissolved in ethylene glycol dimethyl ether (50 mL) and water (10 mL), followed by the addition of 5-fluoro-4-iodopyridin-2-amine (350mg, 1.47mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (110mg, 0.15mmol) and potassium carbonate (405mg, 2.94mmol), the system was warmed to 100 ℃ under nitrogen protection for 4 hours, TLC monitored for complete reaction of the starting materials. The reaction was cooled to room temperature and the crude product was purified by column chromatography (petroleum ether: ethyl acetate =10: 1-2:1) to afford 19b (300 mg, 71% yield).
Synthesis of intermediate 19c
(1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (292mg, 1.20mmol) was dissolved in methylene chloride (50 mL), followed by addition of pyridine (474mg, 6.00mmol) and thionyl chloride (242mg, 2.04mmol), reaction at room temperature for 4 hours, followed by addition of 19b (300mg, 1.32mmol) to the above reaction mixture and reaction at room temperature overnight. TLC monitored no starting material remaining. The reaction solution was diluted with water (30 mL), the organic phase was collected by extraction, concentrated under reduced pressure, and the crude product was purified by column chromatography (petroleum ether: ethyl acetate =10: 1-2:1) to give 19c (200 mg, 39% yield).
Synthesis of intermediate 19d
19c (200mg, 0.47mmol) was dissolved in dichloromethane (10 mL), followed by the addition of trifluoroacetic acid (1 mL) and reaction at room temperature for 1.5 h, and TLC monitored for complete reaction of the starting materials. To the reaction mixture was added saturated sodium bicarbonate (30 mL), extracted with dichloromethane (20 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (20 mL. Times.2), dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure to give 19d (128 mg, 83% yield).
Synthesis of end product 19
19d (128mg, 0.39mmol) was dissolved in dichloromethane (10 mL), followed by addition of acetic anhydride (48mg, 0.47mmol) and triethylamine (47mg, 0.47mmol), reaction at room temperature for 1.5 hours, and TLC monitored for complete reaction of the starting material. To the reaction solution was added saturated sodium bicarbonate (30 mL), extracted with dichloromethane (20 mL × 3), the organic phases were combined, washed with saturated sodium chloride (20 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (dichloromethane: methanol =50: 1-20. MS m/z (ESI) 396.2[ m ] +H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.68(s,1H),8.48(s,1H),8.41(d,J=5.4Hz,1H),8.09(s,1H),7.83-7.78(m,2H),7.46(d,J=7.2Hz,1H),7.41(dd,J=7.2Hz,J=7.2Hz,1H),7.09(s,1H),3.58-3.56(m,1H),2.64-2.60(m,1H),1.90-1.84(m,1H),1.78-1.76(m,6H),1.31-1.25(m,3H),1.10-1.06(m,1H).
Example 20:
synthesis of intermediate 20 a:
2,6-difluoropyridine-3-boronic acid (550mg, 3.14mmol) and the compound 5-fluoro-4-iodopyridin-2-amine (898mg, 3.77mmol) were dissolved in 1,4-dioxane (15 mL), and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (219mg, 0.30mmol) and potassium carbonate (1.30g, 9.42mmol) were added in sequence, followed by water (6 mL). The system was stirred at 100 ℃ for 5 hours under nitrogen blanket and the starting material was not left by TLC. To the reaction solution was added water (100 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (petroleum ether: ethyl acetate = 4:1-2:3) to give 20a (700 mg, 99% yield).
Synthesis of intermediate 20 b:
20a (700mg, 3.14mmol) was dissolved in acetonitrile (50 mL), and then the whole system of compound (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (778mg, 3.20mmol), tetramethylchlorourea hexafluorophosphate (898mg, 3.20mmol) and N-methylimidazole (0.87mL, 11.00mmol) was added and stirred at room temperature for 15 hours, and no starting material was left by TLC detection. After concentrating the solvent, it was purified by column chromatography (petroleum ether: ethyl acetate = 4:1-2:3) to obtain 20b (1.40 g, yield 99%)
Synthesis of final product 20:
20b (1.40g, 3.14mmol) was dissolved in dichloromethane (50 mL), 5mL of trifluoroacetic acid was added thereto at room temperature, and the reaction was carried out for 4 hours with no residue on the starting material by TLC. The reaction system was washed to weak alkalinity with saturated aqueous sodium carbonate solution, extracted with dichloromethane (30 mL. Times.3), and the organic phases were combined and dried over anhydrous sodium sulfate. The solvent was concentrated to 50mL, 2mL of triethylamine and 2mL of acetic anhydride were added, stirred at room temperature for 30 minutes and TLC monitored that no starting material remained. Washing with anhydrous sodium carbonate solution, extraction with dichloromethane (30 mL. Times.3), combining the organic phases, concentrating under reduced pressure to remove the solvent, and column chromatography of the crude product to give the final product 20 (750 mg, 61% yield). MS m/z (ESI) 393.2[ m ] +H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.74(s,1H),8.50(s,1H),8.40(dd,J=16.8Hz,J=7.8Hz,1H),8.26(d,J=6.0Hz,1H),7.79(d,J=7.8Hz,1H),7.41(dd,J=7.8Hz,J=2.4Hz,1H),3.60-3.55(m,1H),2.64-2.60(m,1H),1.91-1.88(m,1H),1.78-1.76(m,6H),1.29-1.27(m,3H),1.09-1.06(m,1H).
Example 21:
synthesis of intermediate 21 a:
5-fluoro-4-iodopyridin-2-amine (1.00g, 4.20mmol) was dissolved in ethylene glycol dimethyl ether (20 mL) and water (4 mL), followed by the addition of indazole-4-boronic acid (0.68g, 4.20mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (0.34g, 0.42mmol) and potassium carbonate (1.74g, 12.60mmol), which were replaced three times with nitrogen gas, so that the entire system was under an atmosphere of nitrogen gas, the system was stirred at 100 ℃ under reflux, reacted for 4 hours, and no excess of starting material was monitored by TLC. Concentrated under reduced pressure and purified by column chromatography (dichloromethane: methanol =50, 1-10) to give 21a (0.77 g, yield 80%).
Synthesis of intermediate 21 b:
21a (0.75g, 3.30mmol) was dissolved in N, N-dimethylformamide (30 mL), followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.80g, 3.30mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (1.56g, 4.10mmol) and N, N-diisopropylethylamine (0.85g, 6.60mmol), the whole was stirred at room temperature overnight, and TLC monitored that no starting material remained. Water (100 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50: 1-20.
Synthesis of intermediate 21 c:
21b (0.91g, 2.00mmol) was dissolved in dichloromethane (30 mL) followed by the addition of 2mL of trifluoroacetic acid in an ice-water bath, and the whole was stirred at room temperature overnight with no starting material remaining by TLC. To the reaction solution was added water (100 mL), followed by adjustment of PH =9-10 with a saturated aqueous solution of sodium bicarbonate, extraction with dichloromethane (50 mL × 3), combination of organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, removal of the solvent under reduced pressure, and purification by column chromatography (dichloromethane: methanol =50, 1-8:1) to obtain 21c (0.61 g, yield 86%).
Synthesis of final product 21:
21c (0.61g, 1.72mmol) was dissolved in dichloromethane (35 mL), followed by the addition of acetic anhydride (0.53g, 5.20mmol) and triethylamine (0.52g, 5.20mmol), the system was stirred at room temperature, and TLC monitored that no starting material remained. Water (50 mL) was added to the reaction solution, dichloromethane was extracted (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and separation and purification by column chromatography (petroleum ether: ethyl acetate =10: 1-2:1) gave final product 21 (0.32 g, 47% yield). MS m/z (ESI) 396.1[ 2 ] M + H] + 。
1 H NMR(600MHz,CDCl 3 )δ8.60(s,1H),8.27(s,1H),8.14(s,1H),7.63(d,J=7.8Hz,1H),7.51(d,J=7.8Hz,1H),7.39(d,J=6.6Hz,1H),7.15(d,J=7.2Hz,1H),3.89-3.87(m,1H),2.54-2.50(m,1H),2.29(d,J=12.0Hz,1H),1.79-1.77(m,6H),1.44-1.28(m,3H),1.12-1.09(m,1H).
Example 22:
synthesis of intermediate 22 a:
5-fluoro-4-iodopyridin-2-amine (1.00g, 4.20mmol) was dissolved in ethylene glycol dimethyl ether (20 mL) and water (4 mL), followed by addition of indole-4-boronic acid (0.68g, 4.20mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (0.34g, 0.42mmol) and potassium carbonate (1.74g, 12.60mmol), and substitution with nitrogen gas was carried out three times to place the whole system under an atmosphere of nitrogen gas. The system was stirred at 100 ℃ under reflux, reacted for 4 hours, and no starting material was left as monitored by TLC. Concentrated under reduced pressure and purified by column chromatography (dichloromethane: methanol =50, 1-10) to give 22a (0.78 g, 82% yield).
Synthesis of intermediate 22 b:
22a (0.78g, 3.40mmol) was dissolved in N, N-dimethylformamide (30 mL), followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.83g, 3.40mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (1.56g, 4.10mmol) and N, N-diisopropylethylamine (0.88g, 6.80mmol), the whole was stirred at room temperature overnight, TLC monitored for no remaining starting material to the reaction solution, water (100 mL) was added, ethyl acetate extracted (50 mL. Times.3), the organic phase was combined, washed with saturated sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50 1-20), to give 22b (0.92 g, yield 59%).
Synthesis of intermediate 22 c:
22b (0.92g, 2.03mmol) was dissolved in dichloromethane (30 mL), followed by addition of 2mL trifluoroacetic acid under ice-water bath, the whole was stirred at room temperature overnight, TLC detected no material remaining, water (100 mL) was added to the reaction solution, pH =9-10 was adjusted with saturated aqueous sodium bicarbonate solution, dichloromethane was extracted (50 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and after purification by column chromatography (dichloromethane: methanol =50, 1-8:1), 22c (0.61 g, 85% yield) was obtained.
Synthesis of end product 22:
22c (0.61g, 1.73mmol) was dissolved in dichloromethane (35 mL), followed by addition of acetic anhydride (0.53g, 5.20mmol) and triethylamine (0.52g, 5.20mmol), the system was stirred at room temperature, and TLC monitored that no starting material remained. Water (50 mL) was added to the reaction solution, dichloromethane was extracted (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and separation and purification by column chromatography (petroleum ether: ethyl acetate =10: 1-2:1) gave the final product 22 (0.35 g, 51% yield). MS m/z (ESI) 395.1[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ11.38(s,1H),10.59(s,1H),8.42(s,1H),8.36(d,J=6.0Hz,1H),7.77(d,J=7.8Hz,1H),7.54(d,J=8.4Hz,1H),7.46(d,J=3.0Hz,1H),7.23(d,J=8.4Hz,1H),7.15(d,J=7.2Hz,1H),6.41(s,1H),3.60-3.54(m,1H),2.64-2.60(m,1H),1.89(d,J=12.0Hz,1H),1.77-1.76(m,6H),1.34-1.23(m,3H),1.10-1.08(m,1H).
Example 23:
synthesis of intermediate 23 a:
5-fluoro-4-iodopyridin-2-amine (1.00g, 4.20mmol) was dissolved in N, N-dimethylformamide (20 mL), followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (1.32g, 5.40mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (2.39g, 6.29mmol) and N, N-dimethylethylamine (2.08mL, 12.6 mmol), and the whole was stirred at room temperature overnight with no remaining starting material for TLC. Water (100 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50: 1-20.
Synthesis of intermediate 23 b:
23a (806mg, 1.74mmol) was dissolved in dichloromethane (10 mL) and then 5mL trifluoroacetic acid was added under an ice-water bath, and the whole was stirred at room temperature for 2 hours with no material remaining by TLC. Water (100 mL) was added to the reaction solution, followed by adjustment of PH =9-10 with saturated aqueous sodium bicarbonate, extraction with dichloromethane (50 mL × 3), combination of organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, and removal of the solvent under reduced pressure to give 23b (770 mg, crude product).
Synthesis of intermediate 23 c:
23b (770 mg, crude) was dissolved in dichloromethane (10 mL) followed by triethylamine (883. Mu.L, 6.35 mmol), acetic anhydride (297. Mu.L, 3.18 mmol). The system was stirred at room temperature and no starting material was left as monitored by TLC. To the reaction solution was added water (50 mL), extracted with dichloromethane (50 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (ethyl acetate) to give 23c (490 mg, 69% yield).
Synthesis of intermediate 23 d:
23c (100mg, 0.25mmol) was dissolved in 1,4 dioxane (10 mL) and water (5 mL), followed by addition of 4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) -1H-pyrrolo [2,3-b ] pyridine-1-carboxylic acid tert-butyl ester (167mg, 0.49mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (15mg, 0.02mmol) and potassium carbonate (68mg, 0.49mmol), and the whole was replaced three times with nitrogen gas to leave the system under an atmosphere of nitrogen gas. The system was stirred at 100 ℃ under reflux, reacted for 4 hours, and TLC monitored that no starting material remained. The reaction was directly purified by column chromatography (dichloromethane: methanol = 50.
Synthesis of end product 23:
23d (122mg, 0.24mmol) was dissolved in dichloromethane (10 mL), followed by the addition of 5mL trifluoroacetic acid in an ice-water bath, and the whole was stirred at room temperature for 2 hours with no residue on TLC. To the reaction solution was added water (100 mL), followed by adjustment of PH =9-10 with a saturated aqueous solution of sodium bicarbonate, extraction with dichloromethane (50 mL × 3), combination of organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, and then removal of the solvent under reduced pressure, followed by separation and purification by column chromatography (dichloromethane: methanol =50, 1-10) to obtain a final product 23 (57 mg, yield 59%). MS m/z (ESI) 396.2[ m ] +H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ11.97(s,1H),10.70(s,1H),8.42(s,1H),8.41(s,1H),8.36(d,J=4.8Hz,1H),7.79(d,J=7.8Hz,1H),7.63-7.62(m,1H),7.23(d,J=4.2Hz,1H),6.47(s,1H),3.58-3.56(m,1H),2.62-2.61(m,1H),1.99(d,J=4.8Hz,1H),1.91-1.89(m,6H),1.39-1.21(m,3H),1.20-1.09(m,1H).
Example 24:
synthesis of intermediate 24 a:
(1-methyl-1H-pyrrolo [2,3-b ] pyridin-4-yl) boronic acid (176mg, 1.00mmol) and 23a (461mg, 1.00mmol) were dissolved in 20mL of 1, 4-dioxane, and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (73mg, 0.10mmol) and potassium carbonate (414mg, 3.00mmol) were added in this order. The system was stirred at 100 ℃ for 4.5 hours under nitrogen blanket and TLC monitored for completion of the starting material reaction. The reaction was stopped and water (50 mL) was added to the reaction system, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure and purified by column chromatography (petroleum ether: ethyl acetate = 4:1-petroleum ether: ethyl acetate = 1:1) to obtain 24a (400 mg, yield 86%).
Synthesis of final product 24:
24a (400mg, 0.86mmol) was dissolved in dichloromethane (25 mL), trifluoroacetic acid (3 mL) was added at room temperature, the reaction was carried out at room temperature for 4 hours, TLC monitored that no starting material remained, the mixture was washed with saturated sodium carbonate solution to weak alkalinity, the aqueous phase was extracted with dichloromethane (30 mL. Times.3), and the organic phases were combined and dried over anhydrous sodium sulfate. The solvent was concentrated to a volume of 15mL, triethylamine (2 mL) and acetic anhydride (2 mL) were added to react at room temperature for 30 minutes, TLC monitored that no starting material remained, saturated sodium carbonate solution washed the reaction system, and the solvent was concentrated and subjected to silica gel column chromatography (dichloromethane: methanol =50 1-10. MS m/z (ESI) 410.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.69(s,1H),8.50(s,1H),8.41(s,1H),8.40(s,1H),7.77(d,J=7.8Hz,1H),7.68(d,J=3.6Hz,1H),7.26(d,J=4.8Hz,1H),6.47(d,J=2.4Hz,1H),3.89(s,3H),3.58-3.56(m,1H),2.64-2.60(m,1H),1.91-1.89(m,1H),1.79-1.77(m,6H),1.30-1.26(m,3H),1.09-1.07(m,1H).
Example 25:
synthesis of final product 25:
5b (0.61g, 1.69mmol) was dissolved in dichloromethane (35 mL), followed by the addition of methanesulfonyl chloride (0.29g, 2.54mmol) and triethylamine (0.34g, 3.38mmol), the system was stirred at room temperature and monitored by TLC follow-up until no starting material remained. Water (50 mL) was added to the reaction mixture, methylene chloride was extracted (50 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, and the residue was extracted with dichloromethaneThe solvent was removed under pressure and purified by column chromatography (petroleum ether: ethyl acetate =10: 1-2:1) to give final product 25 (0.41 g, 55% yield). MS m/z (ESI) 440.1[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.60(s,1H),8.33(s,1H),8.08(d,J=4.8Hz,1H),7.34(d,J=7.2Hz,1H),7.10(d,J=7.8Hz,2H),6.92(d,J=8.4Hz,1H),3.80(s,3H),3.13-3.11(m,1H),2.92(s,3H),2.62-2.57(m,1H),2.03(d,J=12Hz,1H),2.25(d,J=12Hz,1H),1.77-1.73(m,2H),1.36-1.11(m,4H).
Example 26:
synthesis of intermediate 26 a:
5-fluoro-4-iodopyridin-2-amine (0.50g, 2.10mmol) was dissolved in ethylene glycol dimethyl ether (20 mL) and water (4 mL), followed by addition of 4,5-difluoro-2-methoxyphenylboronic acid (0.39g, 2.10mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (0.15g, 0.21mmol) and potassium carbonate (0.87g, 6.3mmol), and replacement with nitrogen gas was carried out three times to place the entire system under an atmosphere of nitrogen gas. The system was stirred at 100 ℃ under reflux, reacted for 4 hours, and TLC monitored that no starting material remained. The reaction was directly purified by column chromatography (dichloromethane: methanol = 50.
Synthesis of intermediate 26 b:
26a (0.30g, 1.18mmol) was dissolved in N, N-dimethylformamide (30 mL), followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.35g, 1.42mmol), 2- (7-oxybenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (0.54g, 1.42mmol) and N, N-dimethylethylamine (0.30g, 2.36mmol), and the whole was stirred at room temperature overnight with TLC detection of no remaining starting material. Water (100 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50: 1-20.
Synthesis of intermediate 26 c:
26b (0.20g, 0.42mmol) was dissolved in dichloromethane (30 mL), followed by trifluoroacetic acid (2 mL) added under an ice-water bath, and the whole was stirred at room temperature overnight with TLC detection until no starting material remained. To the reaction solution was added water (100 mL), followed by adjustment of PH =9-10 with saturated aqueous sodium bicarbonate solution, extraction with dichloromethane (50 mL × 3), combination of organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, and then removal of the solvent under reduced pressure, followed by column chromatography separation purification (dichloromethane: methanol =50, 1-8:1) to obtain 26c (0.11 g, yield 70%).
Synthesis of final product 26:
26c (0.11g, 0.29mmol) was dissolved in dichloromethane (35 mL), followed by the addition of methanesulfonyl chloride (0.40g, 0.35mmol) and triethylamine (44mg, 0.46mmol). The system was stirred at room temperature and monitored by TLC tracking until no starting material remained. To the reaction solution was added water (50 mL), extracted with dichloromethane (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and subjected to column chromatography for purification (petroleum ether: ethyl acetate =10: 1-2:1) to obtain the final product 26 (40 mg, yield 30%). MS m/z (ESI) 458.1[ 2 ] M + H] + 。
1 H NMR(600MHz,CDCl 3 )δ8.59(s,1H),8.31(s,1H),8.14(d,J=10.8Hz,1H),7.17(d,J=9.0Hz,1H),6.85-6.82(m,1H),5.46(s,1H),3.88(s,1H),3.83(s,3H),2.82(s,1H),2.48(s,1H),2.27-2.25(m,1H),2.05-1.94(m,5H),1.45-1.16(m,4H).
Example 27:
synthesis of final product 27:
5b (0.61g, 1.69mmol) was dissolved in dichloromethane (35 mL) followed by the addition of thiophenesulfonyl chloride (0.46g, 2.53mmol) and triethylamine (0.34g, 3.38mmol). The system was stirred at room temperature overnight and monitored by TLC tracking until no starting material remained. Water (50 mL) was added to the reaction mixture, methylene chloride was used for extraction (50 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2),dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and column chromatography separation and purification (petroleum ether: ethyl acetate =10: 1-2:1) gave final product 27 (0.52 g, 61% yield). MS m/z (ESI) 508.1[ 2 ], [ M ] +H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.55(s,1H),8.31(s,1H),8.03(s,1H),7.96(d,J=7.2Hz,1H),7.89(s,1H),7.57(s,1H),7.32(d,J=7.2Hz,1H),7.15(s,1H),7.08(d,J=11.4Hz,1H),6.89(d,J=8.4Hz,1H),4.02(s,3H),3.04-3.03(m,1H),1.78-1.62(m,5H),1.17-1.08(m,4H).
Example 28:
synthesis of end product 28:
5b (79mg, 0.22mmol) was dissolved in acetonitrile (5 mL), and sodium carbonate (47mg, 0.44mmol), and benzyl bromide (37mg, 0.22mmol) were added. The system was left to react overnight at room temperature. TLC monitoring indicated no starting material remained. The solvent was removed by concentration, and the crude product was isolated and purified by thin plate chromatography (dichloromethane: methanol =10 = 1) to give the final product 28 (30 mg, yield 30%). MS (m/z, ESI) 451.2[ deg. ] M + H] + 。
1 H NMR(600MHz,CD 3 OD)δ8.16(s,1H),8.09(d,J=4.8Hz,1H),7.37-7.32(m,4H),7.30-7.26(m,2H),6.93(d,J=11.4Hz,1H),6.80(dd,J=8.4Hz,1H),3.86(s,2H),3.81(s,3H),2.71-2.67(m,1H),2.51-2.47(m,1H),2.20-2.18(m,1H),2.06-2.04(m,1H),1.89-1.85(m,2H),1.49-1.39(m,3H),1.21-1.16(m,1H).
Example 29:
synthesis of final product 29:
5b (0.61g, 1.69mmol) was dissolved in N, N-dimethylformamide (35 mL), followed by the addition of bromoethyl methyl ether (0.26g, 1.86mmol) and potassium carbonate (0.47g, 3.38mmol). The system was stirred at room temperature and no starting material was left as monitored by TLC. Water (50 mL) and B were added to the reaction mixtureExtraction with ethyl acid (50 mL × 3), combination of organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, removal of the solvent under reduced pressure, and column chromatography separation purification (dichloromethane: methanol =50, 1-20. MS m/z (ESI) 420.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,CDCl 3 )δ8.25(d,J=5.4Hz,1H),8.12(s,1H),7.25(d,J=6.6Hz,1H),6.75-6.70(m,2H),3.80(s,3H),3.57(t,J=4.8Hz,2H),3.36(s,3H),2.90(t,J=4.8Hz,2H),2.70(s,1H),2.37(t,J=5.4Hz,1H),2.26(d,J=12.0Hz,1H),2.05-1.90(m,3H),1.52-1.22(m,4H).
Example 30:
synthesis of final product 30:
5b (155mg, 0.43mmol) was dissolved in acetonitrile (5 mL), and triethylamine (86mg, 0.86mmol), bromoethanol (54mg, 0.43mmol) and sodium iodide (10mg, 0.06mmol) were added. The system was left to react at 60 ℃ for 12 hours. TLC monitoring indicated no starting material remained. After cooling, the solvent was removed by concentration. The crude product was isolated and purified by thin plate chromatography (dichloromethane: methanol: triethylamine = 40. MS m/z (ESI) 406.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,CD 3 OD)δ8.18(s,1H),8.08(d,J=4.8Hz,1H),7.28(d,J=7.2Hz,1H),6.92(d,J=11.4Hz,1H),6.79(d,J=8.4,1H),3.82-3.80(m,5H),3.20-3.25(m,1H),3.20-3.18(m,2H),2.65-2.62(m,1H),2.31-2.29(m,1H),2.18-2.16(m,1H),1.99-1.91(m,2H),1.70-1.64(m,1H),1.53-1.34(m,3H).
Example 31:
synthesis of final product 31:
5b (79mg, 0.22mmol) was dissolved in dioxane (5 mL), and the compound 2-bromo-6-methoxypyridine (40 mg) was added0.22 mmol), tris (dibenzylideneacetone) dipalladium (20mg, 0.022mmol), 4,5-bis diphenylphosphine-9,9-dimethylxanthene (12.7mg, 0.022mmol) and cesium carbonate (215mg, 0.66mmol). The entire system was placed under a nitrogen atmosphere by replacing with nitrogen three times. The system was left to react at 120 ℃ for 2 hours. TLC monitoring showed no starting material remaining. After cooling, concentration was performed under reduced pressure, and the crude product was isolated and purified by thin plate chromatography (dichloromethane: methanol =10 = 1) to obtain a final product 31 (30 mg, yield 29%). MS (m/z, ESI) 469.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.56(s,1H),8.31(s,1H),8.07(d,J=5.4Hz,1H),7.32(d,J=7.2Hz,1H),7.24(d,J=7.8Hz,1H),7.08(d,J=11.4Hz,1H),6.89(d,J=8.4Hz,1H),6.33(d,J=7.2Hz,1H),5.97(d,J=7.8Hz,1H),5.80(d,J=7.8Hz,1H),3.76(s,3H),3.72(s,3H),3.63-3.61(m,1H),2.64-2.62(m,1H),2.06-2.04(m,1H),1.97-1.95(m,1H),1.79-1.77(m,2H),1.36-1.30(m,3H),1.11-1.09(m,1H).
Example 32:
synthesis of end product 32:
5b (61mg, 0.17mmol) was dissolved in N, N-dimethylformamide (5 mL), followed by addition of cyanoacetic acid (1695g, 0.18mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (69mg, 0.18mmol) and N, N-diisopropylethylamine (83uL, 0.50mmol), the whole was stirred at room temperature, reacted for 10 hours, followed by TLC monitoring until no starting material remained. To the reaction solution was added water (50 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and subjected to column chromatography for purification (dichloromethane: methanol =40: 1-20. MS m/z (ESI) 429.2[ 2 ], [ M ] +H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.58(s,1H),8.31(s,1H),8.20(d,J=7.8Hz,1H),8.06(d,J=5.4Hz,1H),7.32(t,J=7.2Hz,1H),7.08(d,J=11.4Hz,1H),6.90(t,J=8.4Hz,1H),3.77(s,3H),3.56(m,3H),2.61-2.57(m,1H),1.89-1.76(m,4H),1.30-1.07(m,4H).
Example 33:
synthesis of end product 33:
5b (0.16g, 0.43mmol) was dissolved in N, N-dimethylformamide (30 mL), followed by the addition of 1-cyano-1-cyclopropanecarboxylic acid (72mg, 0.65mmol), 2- (7-oxybenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (178mg, 0.47mmol) and N, N-diisopropylethylamine (111mg, 0.86mmol), the system was stirred at room temperature overnight, and TLC monitored for no residue of starting material. Water (100 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (dichloromethane: methanol = 50. MS m/z (ESI) 455.2[ deg. ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.56(s,1H),8.31(s,1H),8.05(d,J=4.8Hz,1H),8.09(d,J=7.8Hz,1H),7.32(dd,J=7.8Hz,J=7.2Hz,1H),7.09(d,J=11.4Hz,1H),6.90(dd,J=8.4Hz,J=7.8Hz,1H),3.77(s,3H),3.68-3.56(m,1H),3.59-3.55(m,1H),1.81-1.67(m,4H),1.53-1.46(m,5H)),1.28-1.23(m,3H).
Example 34:
synthesis of final product 34:
5b (0.16g, 0.43mmol) was dissolved in N, N-dimethylformamide (30 mL), followed by the addition of 1-cyanocyclobutanecarboxylic acid (81mg, 0.65mmol), 2- (7-benzotriazol oxide) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (178mg, 0.47mmol) and N, N-diisopropylethylamine (111mg, 0.86mmol), the system was stirred at room temperature overnight and TLC monitored for no residual starting material. Water (100 mL) was added to the reaction mixtureEthyl acetate extraction (50 mL × 3), organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (dichloromethane: methanol =50: 1-20. MS m/z (ESI) 469.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,DMSO-d6)δ10.56(s,1H),8.31(s,1H),8.05(d,J=4.8Hz,1H),8.09(d,J=7.8Hz,1H),7.34-7.31(m,1H),7.09(s,1H),6.91-6.88(m,1H),3.77(s,3H),3.68-3.56(m,1H),2.59-2.55(m,1H),1.81-1.67(m,4H),1.53-1.46(m,5H),1.30-1.23(m,5H).
Example 35:
synthesis of intermediate 35 a:
2,2' -dibromodiethyl ether (12.85g, 55.60mmol) was dissolved in N, N-dimethylformamide (10 mL), followed by the addition of methyl cyanoacetate (5.00g, 50.45mmol) and 1,8-diazabicyclo [5.4.0] undec-7-ene (11.50g, 75.68mmol), and displacement with nitrogen three times, leaving the entire system under an atmosphere of nitrogen. The system was stirred at 85 ℃ and reacted for 4 hours with no starting material remaining as monitored by TLC. To the reaction solution was added water (100 mL), extracted with ethyl acetate (50 mL × 3), combined organic phases, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and purified by column chromatography (petroleum ether: ethyl acetate = 1:1) to give 35a (5.00 g, 59% yield).
Synthesis of intermediate 35 b:
35a (2.37g, 14.00mmol) was dissolved in ethanol (36 mL) and water (5 mL), followed by sodium hydroxide (2.24g, 56.00mmol), and the whole was stirred at room temperature for 4 hours with no residual starting material on TLC. Water (100 mL) was added to the reaction mixture, the pH was adjusted to 8 with aqueous sodium bicarbonate, the mixture was extracted with ethyl acetate (50 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure to give 35b (1.50 g, 69% yield).
Synthesis of final product 35:
35b (123mg, 0.79mmol) was dissolved in N, N-dimethylformamide (5 mL), followed by addition of (1S, 3R) -3-amino-N- (5-fluoro-4- (4-fluoro-2-methoxyphenyl) pyridin-2-yl) cyclohexanamide (260mg, 0.72mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (548mg, 1.44mmol) and N, N-diisopropylethylamine (357. Mu.L, 2.16 mmol), the whole system was stirred at room temperature for 10 hours and TLC monitored that no starting material remained. To the reaction solution was added water (50 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and subjected to column chromatography for purification (dichloromethane: methanol =40: 1-20. MS m/z (ESI) 499.2[ deg. ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.58(s,1H),8.33(s,1H),8.17(d,J=7.8Hz,1H),8.08(d,J=6.0Hz,1H),7.35-7.33(m,1H),7.11-7.09(m,1H),6.93-6.89(m,1H),3.88-3.86(m,2H),3.78(s,3H),3.66-3.63(m,1H),3.52-3.51(m,2H),2.63(m,1H),1.97-1.96(m,4H),1.85-1.75(m,4H),1.38-1.21(m,4H).
Example 36:
synthesis of the end product 36:
triphosgene (33mg, 0.11mmol) was dissolved in dichloromethane (2 mL), followed by dropwise addition of a solution of 5b (61mg, 0.17mmol) and triethylamine (22mg, 0.22mmol) in dichloromethane (2 mL), and the whole was stirred at room temperature for 2 hours, followed by addition of a methanol solution (2 mL), reaction was continued for 3 hours, monitored by TLC until no starting material remained. Water (50 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and column chromatography separation and purification (dichloromethane: methanol =40: 1-20. MS m/z (ESI) 420.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.56(s,1H),8.31(s,1H),8.06(d,J=5.4Hz,1H),7.33(d,J=7.8Hz,1H),7.10-7.07(m,2H),6.90(d,J=8.4Hz,1H),3.77(s,3H),3.48(s,3H),3.32(m,1H),2.56(m,1H),1.87-1.71(m,4H),1.31-1.06(m,4H).
Example 37:
synthesis of final product 37:
triphosgene (33mg, 0.11mmol) was dissolved in dichloromethane (2 mL), followed by dropwise addition of a solution of 5b (61mg, 0.17mmol) and triethylamine (22mg, 0.22mmol) in dichloromethane, the whole was stirred at room temperature, reacted for 2 hours, followed by addition of 3mL of 33% methylamine alcohol solution, the reaction was continued for 3 hours, monitored by TLC until no starting material remained. To the reaction solution was added water (50 mL), extracted with ethyl acetate (50 mL × 3), and the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and subjected to column chromatography for purification (dichloromethane: methanol =40, 1-20) to obtain a final product 37 (15 mg, yield 21%). MS m/z (ESI) 419.2[ deg. ] M + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.56(s,1H),8.31(s,1H),8.06(d,J=5.4Hz,1H),7.33(t,J=8.4Hz,1H),7.10-7.07(m,1H),6.91-6.88(m,1H),5.78(d,J=8.4Hz,1H),5.56(d,J=4.8Hz,1H),3.76(s,3H),3.36-3.34(m,1H),2.65-2.55(m,1H),2.51(s,3H),1.88-1.73(m,4H),1.28-0.82(m,4H).
Example 38:
synthesis of end product 38:
5b (0.61g, 1.69mmol) was dissolved in dichloromethane (35 mL) followed by the addition of dimethylcarbamoyl chloride (0.27g, 2.54mmol) and triethylamine (0.34g, 3.38mmol). The system was stirred at room temperature overnight and no starting material was left as monitored by TLC. Water (50 mL) was added to the reaction mixture, methylene chloride was extracted (50 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), and dried over anhydrous sodium sulfateThe solvent was removed under reduced pressure and purified by column chromatography (dichloromethane: methanol =100: 1-40). MS m/z (ESI) 433.1[ 2 ], [ M ] +H] + 。
1 H NMR(600MHz,CDCl 3 )δ8.28-8.25(m,2H),8.11(s,1H),7.27-7.24(m,2H),6.76-6.70(m,2H),3.81(s,3H),3.76-3.73(m,1H),2.88(s,6H),2.47-2.43(m,1H),2.30(d,J=12.0Hz,1H),2.02-1.90(m,3H),1.47-1.12(m,4H).
Example 39:
synthesis of intermediate 39 a:
5b (0.12g, 0.33mmol) was dissolved in N, N-dimethylformamide (10 mL), and (S) -2- ((tert-butoxycarbonyl) amino) -2- (4-hydroxyphenyl) acetic acid (0.10g, 0.36mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (0.15g, 0.40mmol), N, N-diisopropylethylamine (0.09g, 0.66mmol) were added, reacted at room temperature for 8 hours, and TLC monitored that no residue was left on the starting material. Water (20 mL) was added to the reaction mixture, extracted with ethyl acetate (15 mL. Times.3), and the organic phases were combined and dried over anhydrous sodium sulfate. The organic phase was directly subjected to column chromatography (dichloromethane: methanol =10 = 1) to yield 39a (0.12 g, 60% yield).
Synthesis of end product 39:
39a (0.12g, 0.20mmol) was dissolved in methylene chloride (20 mL), and trifluoroacetic acid (4 mL) was added to react at room temperature for 4 hours, and the starting material was monitored by TLC to be not remained. The reaction solution was washed with water (20 mL. Times.3), the aqueous phases were combined, the pH of the aqueous phase was adjusted to 8-9 with sodium carbonate, dichloromethane was extracted (30 mL. Times.3), the organic phases were combined, and dried over anhydrous sodium sulfate. The organic phase was distilled off under reduced pressure to give the final product 39 (0.04 g, 39% yield). MS m/z (ESI) 511.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.56(s,1H),9.23(s,1H),8.32(s,1H),8.08(d,J=5.4Hz,1H),7.90-7.89(m,1H),7.36-7.34(m,1H),7.17-7.15(m,2H),7.12-7.10(m,1H),6.93-6.91(m,1H),6.68-6.66(m,2H),4.19(s,1H),4.04(d,J=7.2Hz,2H),3.80(s,3H),3.58-3.57(m,1H),2.59-2.57(m,1H),1.80-1.76(m,4H),1.33-1.25(m,4H).
Example 40:
synthesis of final product 40:
5b (0.20g, 0.55mmol) was dissolved in N, N-dimethylformamide (30 mL), and N, N-dimethylglycine (67mg, 0.65mmol), 2- (7-benzotriazol oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate (178mg, 0.47mmol) and N, N-diisopropylethylamine (111mg, 0.86mmol) were added, and the system was stirred at room temperature overnight, and the starting material was monitored by TLC to be free of residue. To the reaction solution was added water (100 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (dichloromethane: methanol = 50. MS m/z (ESI) 447.2[ m ] +H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.56(s,1H),8.31(s,1H),8.06(d,J=4.2Hz,1H),7.58(d,J=8.4Hz,1H),7.32(dd,J=7.8Hz,J=7.2Hz,1H),7.08(d,J=10.8Hz,1H),6.90(dd,J=7.8Hz,J=7.8Hz,1H),3.76(s,3H),3.65-3.63(m,1H),2.84-2.77(m,2H),2.66-2.60(m,1H),2.16(s,6H),1.85-1.84(m,1H),1.75-1.69(m,3H),1.41-1.35(m,1H),1.32-1.23(m,3H).
Example 41:
synthesis of compound 41:
triphosgene (0.027g, 0.09mmol) was dissolved in anhydrous tetrahydrofuran (20 mL), and a solution of 3-methylisoxazole-5-amine (0.027g, 0.28mmol) in tetrahydrofuran and triethylamine (0.028g, 0.28mmol) were added and reacted at room temperature for 6 hours. A solution of 5b (0.10g, 0.28mmol) in tetrahydrofuran and triethylamine (0.028g, 0.28mmol) were added to the reaction mixture, and the mixture was heated to 70 ℃ and reacted for 6 hours. TLC monitorNo starting material remained, the reaction was stopped, cooled to room temperature, and the reaction solution was separated by column chromatography (petroleum ether: ethyl acetate = 1:1) to obtain final product 41 (45 mg, yield 33%). MS m/z (ESI) 486.2[ deg. ] M + H] + 。
1 H NMR(600MHz,CDCl 3 )δ8.65(s,1H),8.29(s,1H),8.12(s,1H),6.76-6.71(m,2H),6.23(s,2H),5.43(s,1H),4.03-4.01(m,1H),3.81(s,3H),2.54-2.53(m,1H),2.35(s,3H),2.36-2.24(m,2H),2.05-1.94(m,3H),1.56-1.50(m,3H).
Example 42:
synthesis of final product 42:
5b (100mg, 0.28mmol) was dissolved in dichloromethane (10 mL), and trifluoroacetic anhydride (56mg, 0.55mmol), and triethylamine (56mg, 0.55mmol) were added and reacted at room temperature for 1.5 hours, and TLC monitored that no starting material remained. To the reaction solution was added saturated sodium bicarbonate (30 mL), extracted with dichloromethane (20 mL × 3), the organic phases were combined, washed with saturated sodium chloride (20 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (dichloromethane: methanol =50: 1-20. MS m/z (ESI) 458.1[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.59(s,1H),9.33(d,J=8.4Hz,1H),8.31(s,1H),8.05(d,J=6Hz,1H),7.32(dd,J=7.8Hz,J=7.2Hz,1H),7.08(m,1H),6.89(m,1H),3.76(s,3H),3.70-3.68(m,1H),2.63-2.59(m,1H),1.86-1.83(m,1H),1.80-1.76(m,2H),1.51-1.44(m,1H),1.33-1.24(m,4H).
Example 43:
synthesis of compound 43:
5b (0.11g, 0.30mmol), 2-pyridinecarboxylic acid (0.04g, 0.33mmol) were dissolved in N, N-dimethylformamide (15 mL), and 2- (7-azobenzene was addedAnd triazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (0.14g, 0.36mmol) and N, N-diisopropylethylamine (0.08g, 0.6 mmol) were reacted at room temperature for 8 hours, and TLC monitored that no starting material remained. Water (30 mL) was added to the reaction mixture to precipitate a solid, which was then filtered to obtain the final product 43 (0.07g, 50%). MS m/z (ESI) 467.2[ deg. ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.63(s,1H),8.63-8.62(m,2H),8.31(s,1H),8.07(d,J=5.4Hz,1H),8.01(d,J=7.8Hz,1H),7.98-7.95(m,1H),7.58-7.56(m,1H),7.34-7.31(m,1H),7.09-7.07(m,1H),6.91-6.88(m,1H),3.89-3.87(m,1H),3.77(s,3H),2.71-2.63(m,1H),1.92-1.91(m,1H),1.82-1.77(m,3H),1.61-1.55(m,1H),1.45-1.28(m,3H).
Example 44:
synthesis of final product compound 44:
5b (188mg, 0.52mmol) and 1-ethyl-3-methyl-1H-pyrazole-4-carboxylic acid (161mg, 1.04mmol) were dissolved in N, N-dimethylformamide (5 mL), 2- (7-oxybenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (395mg, 1.04mmol) and diisopropylethylamine (0.26mL, 1.56mmol) were added in this order, and the reaction system was reacted at room temperature for 15 hours with no remaining starting material by TLC. To the reaction solution was added water (50 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (petroleum ether: ethyl acetate = 4:1-2:1) to obtain the final product 44 (230 mg, yield 88%). MS m/z (ESI) 498.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.63(s,1H),8.34(s,1H),8.22(d,J=8.4Hz,1H),8.09(d,J=6.0Hz,1H),7.35(m,1H),7.11(m,1H),6.92(m,1H),6.59(s,1H),4.38(dd,J=7.2Hz,J=3.0Hz,2H),3.79(s,4H),2.15(s,3H),2.00(d,J=11.4Hz,1H),1.84-1.80(m,4H),1.49-1.46(m,2H),δ1.32-1.30(m,5H).
Example 45:
synthesis of final product 45:
5b (50mg, 0.138mmol) was dissolved in N, N-dimethylformamide (5 mL), then glycolic acid (1695g, 0.21mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (80mg, 0.21mmol), and N, N-diisopropylethylamine (69. Mu.L, 0.42 mmol) were added, the whole system was stirred at room temperature, reacted for 10 hours, and monitored by TLC until no starting material remained. Water (50 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and column chromatography separation and purification (dichloromethane: methanol =40: 1-20. MS m/z (ESI) 420.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.56(s,1H),8.31(s,1H),8.06(d,J=5.4Hz,1H),7.56(d,J=9.0Hz,1H),7.34-7.31(m,1H),7.09-7.07(m,1H),6.91-6.88(m,1H),5.38-5.36(m,1H),3.77-3.75(m,5H),3.66-3.63(m,1H),2.61-2.57(m,1H),1.83-1.68(m,4H),1.34-1.26(m,4H).
Example 46:
synthesis of end product 46:
5b (0.20g, 0.55mmol) was dissolved in N, N-dimethylformamide (30 mL), and then ethoxyacetic acid (68mg, 0.65mmol), 2- (7-benzotriazol oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate (178mg, 0.47mmol) and N, N-diisopropylethylamine (111mg, 0.86mmol) were added, and the system was stirred at room temperature overnight, and the starting material was monitored by TLC to be free of residue. To the reaction solution was added water (100 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (dichloromethane: methanol =50 1-20Yield 39%). MS m/z (ESI) 448.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.58(s,1H),8.33(s,1H),8.07(d,J=5.4Hz,1H),7.59(d,J=8.4Hz,1H),7.35-7.33(m,1H),7.11-7.09(m,1H),6.93-6.90(m,1H),3.78-3.76(m,5H),3.69-3.67(m,1H),3.47-3.44(m,2H),2.63-2.61(m,1H),1.84-1.69(m,4H),1.47-1.43(m,1H),1.30-1.28(m,6H).
Example 47:
synthesis of end product 47:
45 (0.20g, 0.48mmol) was dissolved in methylene chloride (30 mL), followed by addition of acetyl chloride (45mg, 0.57mmol) and triethylamine (97mg, 0.96mmol), and the system was stirred at room temperature for 1.5 hours, and the starting material was monitored by TLC to be free from residue. The solvent was removed under reduced pressure, and the crude product was purified by column chromatography (dichloromethane: methanol = 50. MS m/z (ESI) 462.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.58(s,1H),8.33(s,1H),8.07(d,J=5.4Hz,1H),7.59(d,J=8.4Hz,1H),7.35-7.34(m,1H),7.33-7.11(m,1H),7.09-6.90(m,1H),4.39(s,2H),3.78(s,3H),3.65-3.63(m,1H),2.66-2.60(m,1H),2.58(s,3H),1.85-1.84(m,1H),1.75-1.69(m,3H),1.39-1.24(m,3H),1.16-1.14(m,1H).
Example 48:
synthesis of final product 48:
5b (0.20g, 0.55mmol) was dissolved in N, N-dimethylformamide (30 mL), and then L-lactic acid (58.5mg, 0.65mmol), 2- (7-benzotriazol oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate (178mg, 0.47mmol) and N, N-diisopropylethylamine (111mg, 0.86mmol) were added, and the system was stirred at room temperature overnight, and the starting material was monitored by TLC to be free of residue. Adding water (to the reaction solution)100 mL), extraction with ethyl acetate (50 mL × 3), combination of the organic phases, washing with saturated sodium chloride (50 mL × 2), drying over anhydrous sodium sulfate, then removal of the solvent under reduced pressure, and purification of the crude product by column chromatography (dichloromethane: methanol =50, 1-20) to give the final product 48 (96 mg, 40% yield). MS m/z (ESI) 434.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.57(s,1H),8.31(s,1H),8.05(d,J=6Hz,1H),7.51(d,J=8.4Hz,1H),7.34-7.31(m,1H),7.10-7.07(m,1H),6.91-6.88(m,1H),5.38(d,J=5.4Hz,1H),3.92-3.88(m,1H),3.77(s,3H),3.62-3.59(m,1H),2.60-2.56(m,1H),1.83-1.81(m,1H),1.77-1.72(m,2H),1.41-1.35(m,1H),1.31-1.26(m,4H),1.23(d,J=13.2Hz,3H).
Example 49:
synthesis of end product 49:
5b (0.20g, 0.55mmol) was dissolved in N, N-dimethylformamide (30 mL), and then D-lactic acid (58.5mg, 0.65mmol), 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate (178mg, 0.47mmol) and N, N-diisopropylethylamine (111mg, 0.86mmol) were added, and the system was stirred at room temperature overnight, and the starting material was monitored by TLC to be free from residue. To the reaction solution was added water (100 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (dichloromethane: methanol = 50. MS m/z (ESI) 434.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.57(s,1H),8.30(s,1H),8.05(d,J=4.8Hz,1H),7.51(d,J=7.8Hz,1H),7.33(t,J=7.8Hz,1H),7.08(d,J=11.4Hz,1H),6.89(t,J=7.8Hz,1H),5.38(d,J=5.4Hz,1H),3.92-3.88(m,1H),3.77(s,3H),3.62-3.59(m,1H),2.60-2.56(m,1H),1.81(d,J=12Hz,1H),1.77-1.72(m,2H),1.67(d,J=11.4Hz,1H),1.41-1.35(m,1H),1.31-1.25(m,4H),1.23(s,3H).
Example 50:
synthesis of final product 50:
5b (50mg, 0.138mmol) was dissolved in N, N-dimethylformamide (5 mL), followed by the addition of 3-hydroxypropionic acid (19mg, 0.21mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (79mg, 0.21mmol), and N, N-diisopropylethylamine (69. Mu.L, 0.42 mmol), the entire system was stirred at room temperature for 10 hours, monitored by TLC until no starting material remained. To the reaction solution was added water (50 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and subjected to column chromatography for purification (dichloromethane: methanol =40: 1-20. MS m/z (ESI) 434.2[ 2 ], [ M ] +H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.56(s,1H),8.31(s,1H),8.06-8.05(d,J=5.4Hz,1H),7.57-7.55(d,J=9.0Hz,1H),7.34-7.31(m,1H),7.09-7.07(m,1H),6.91-6.88(m,1H),5.38-5.36(t,J=5.4Hz,1H),3.77-3.75(m,5H),3.66-3.63(m,1H),2.61-2.56(m,3H),1.83-1.68(m,4H),1.34-1.26(m,4H).
Example 51:
synthesis of final product 51:
5b (50mg, 0.138mmol) was dissolved in N, N-dimethylformamide (5 mL), followed by addition of 1-hydroxycyclopropanecarboxylic acid (21mg, 0.21mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (79mg, 0.21mmol) and N, N-diisopropylethylamine (69. Mu.L, 0.42 mmol), the whole was stirred at room temperature for 10 hours, and TLC was monitored until no starting material remained. Water (50 mL) was added to the reaction mixture, ethyl acetate extraction (50 mL. Times.3) was performed, the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the column was filled with the mixturePurification by chromatographic separation (dichloromethane: methanol =40: 1-20. MS m/z (ESI) 446.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.61(s,1H),8.34(s,1H),8.09-8.08(m,1H),7.73-7.71(m,1H),7.37-7.34(m,1H),7.13-7.10(m,1H),6.95-6.91(m,1H),3.80(s,3H),2.61-2.55(m,2H),1.85-1.72(m,4H),1.50-1.47(m,1H),1.32-1.30(m,3H),1.00-0.98(m,2H),0.81-0.80(m,2H).
Example 52:
synthesis of final product 52:
5b (50mg, 0.138mmol) was dissolved in N, N-dimethylformamide (5 mL), followed by the addition of 3-oxetanecarboxylic acid (21mg, 0.21mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (79mg, 0.21mmol) and N, N-diisopropylethylamine (69. Mu.L, 0.42 mmol), the whole was stirred at room temperature for 10 hours, monitored by TLC until no starting material remained. To the reaction solution was added water (50 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and subjected to column chromatography separation and purification (dichloromethane: methanol =40: 1-20. MS m/z (ESI) 446.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.59(s,1H),8.33(s,1H),8.07-8.06(m,1H),7.98-7.97(m,1H),7.35-7.32(m,1H),7.10-7.08(m,1H),6.93-6.91(m,1H),4.61-4.56(m,2H),3.78(s,3H),3.69-3.67(m,2H),3.42-3.40(m,1H),2.69-2.55(m,2H),1.89-1.87(m,1H),1.77-1.75(m,3H),1.29-1.25(m,3H),1.18-1.09(m,1H).
Example 53:
synthesis of final product 53:
5b (50mg, 0.138mmol) was dissolved in N, N-dimethylformamide (5 mL), followed by addition of 1-methylcyclopropane-1-carboxylic acid (21mg, 0.21mmol), 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate (79mg, 0.21mmol) and N, N-diisopropylethylamine (69. Mu.L, 0.42 mmol), the whole was stirred at room temperature for 10 hours, and TLC was monitored until no starting material remained. Water (50 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and column chromatography separation and purification (dichloromethane: methanol =40: 1-20. MS m/z (ESI) 444.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.57(s,1H),8.32(s,1H),8.08-8.06(m,1H),7.34-7.32(m,1H),7.24-7.22(m,1H),7.11-7.09(m,1H),6.93-6.91(m,1H),3.78(s,3H),3.66-3.58(m,1H),2.59-2.50(m,1H),1.80-1.69(m,4H),1.47-1.44(m,1H),1.30-1.22(m,6H),0.92-0.90(m,2H),0.46-0.44(m,2H).
Example 54:
synthesis of end product 54:
5b (50mg, 0.138mmol) was dissolved in N, N-dimethylformamide (5 mL), followed by the addition of 1-fluorocyclopropanecarboxylic acid (22mg, 0.21mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (79mg, 0.21mmol) and N, N-diisopropylethylamine (69. Mu.L, 0.42 mmol), the whole was stirred at room temperature for 10 hours and monitored by TLC until no starting material remained. To the reaction solution was added water (50 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and subjected to column chromatography for purification (dichloromethane: methanol =40: 1-20. MS m/z (ESI) 448.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,CDCl 3 )δ8.23-8.20(m,1H),8.13(s,1H),7.86(s,1H),7.27-7.25(m,1H),6.80-6.70(m,2H),6.40-6.25(m,1H),4.00-3.85(m,1H),3.82(s,3H),2.50-2.40(m,1H),2.35-2.25(m,1H),2.10-1.90(m,3H),1.55-1.45(m,3H),1.40-1.30(m,2H),1.25-1.15(m,3H).
Example 55:
synthesis of final product 55:
5b (50mg, 0.138mmol) was dissolved in N, N-dimethylformamide (5 mL), followed by addition of 1-trifluoromethylcyclopropane-1-carboxylic acid (32mg, 0.21mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (79mg, 0.21mmol) and N, N-diisopropylethylamine (69. Mu.L, 0.42 mmol), the whole was stirred at room temperature for 10 hours, monitored by TLC until no starting material remained. To the reaction solution was added water (50 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and subjected to column chromatography separation purification (dichloromethane: methanol =40: 1-20. MS m/z (ESI) 498.2[ 2 ] M + H] + 。
1 H NMR(600MHz,CDCl 3 )δ8.23-8.20(m,1H),8.13(s,1H),7.86(s,1H),7.27-7.25(m,1H),6.79-6.69(m,2H),6.05-5.95(m,1H),4.00-3.86(m,1H),3.82(s,3H),2.50-2.35(m,1H),2.30-2.20(m,1H),2.05-1.90(m,3H),1.56-1.35(m,5H),1.21-1.15(m,3H).
Example 56:
synthesis of intermediate 56 a:
glycine (500mg, 6.70mmol) was dissolved in dioxane (10 mL) and water (10 mL), and triethylamine (1.40g, 14.40mmol) and di-tert-butyl dicarbonate (1.74g, 8.00mmol) were added. The system was left to react overnight at room temperature. TLC monitoring indicated complete reaction of starting material. The system was concentrated, an aqueous sodium carbonate solution (100 mL) was added, the mixture was washed with ethyl acetate (100 mL. Times.3), the aqueous phase was adjusted to pH 3 with dilute hydrochloric acid, extracted with ethyl acetate (100 mL. Times.3), and the organic phases were combined and dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated to give 56a (1.00 g, 85% yield).
Synthesis of intermediate 56 b:
56a (46mg, 0.26mmol) was dissolved in N, N-dimethylformamide (3 mL), 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (125mg, 0.33mmol) and diisopropylethylamine (57mg, 0.44mmol) were added, and after the mixture was stirred for 10 minutes, 5b (78mg, 0.22mmol) was added. The system was left to react overnight at room temperature. TLC monitoring indicated complete reaction of starting material. Water (100 mL) was added to the reaction system, followed by extraction with ethyl acetate (100 mL. Times.3), and the organic phases were combined and washed with saturated brine (50 mL. Times.3). Drying with anhydrous sodium sulfate, filtering, and concentrating the filtrate. The crude product was isolated and purified by chromatography on a thin plate (dichloromethane: methanol =10 = 1) to give 56b (80 mg, yield 70%).
Synthesis of end product 56:
56b (80mg, 0.22mmol) was dissolved in dichloromethane (4 mL) and trifluoroacetic acid (1 mL) was added. The system was left at room temperature for 2 hours and TLC monitoring indicated complete reaction. The system was concentrated and the crude product was isolated and purified by thin plate chromatography (dichloromethane: methanol =10 1) to give the final product 56 (40 mg, 43% yield). MS m/z (ESI) 419.2[ deg. ] M + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.59(s,1H),8.31(s,1H),8.05(d,J=6.0Hz,1H),7.94(br,2H),7.32-7.30(m,1H),7.09-7.07(m,1H),6.91-6.88(m,1H),3.76(s,3H),3.64-3.59(m,1H),3.15(d,J=4.8Hz,2H),2.62-2.59(m,1H),1.89-1.87(m,1H),1.79-1.77(m,3H),1.34-1.28(m,3H),1.18-1.10(m,1H).
Example 57:
synthesis of end product 57:
compound 56 (205mg, 0.49mmol) was dissolved in dichloromethane (35 mL), followed by the addition of acetic anhydride (149mg, 1.47mmol) and triethylamine(148mg, 1.47mmol), the system was stirred at room temperature for 4 hours and no starting material was left by TLC. To the reaction solution was added water (50 mL), extracted with dichloromethane (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (petroleum ether: ethyl acetate =10: 1-2:1) to obtain a final product 57 (138 mg, yield 61%). MS m/z (ESI) 461.2[ m ] +H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.60(s,1H),8.33(s,1H),8.07(d,J=6.0Hz,1H),7.95-7.92(m,2H),7.35-7.33(m,1H),7.11(d,J=1.8Hz,1H),6.93-6.90(m,1H),4.09(s,2H),3.79(s,3H),3.58-3.56(m,1H),2.59-2.50(m,1H),1.86-1.73(m,4H),1.83(s,3H),1.39-1.14(m,4H).
Example 58:
synthesis of final product 58:
5b (0.20g, 0.55mmol) was dissolved in N, N-dimethylformamide (30 mL), and N-acetyl-L-leucine (112mg, 0.65mmol), 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate (178mg, 0.47mmol) and N, N-diisopropylethylamine (111mg, 0.86mmol) were added, and the system was stirred at room temperature overnight, and the starting material was monitored by TLC for no residue. To the reaction solution was added water (100 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (dichloromethane: methanol = 50. MS m/z (ESI) 517.2[ m ] +H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.60(s,1H),8.33(s,1H),8.07(d,J=6.0Hz,1H),7.94(dd,J=10.2,Hz,J=8.4Hz,2H),7.35(dd,J=7.8Hz,J=7.2Hz,1H),7.11-7.10(m,1H),6.93-6.90(m,1H),4.24-4.23(m,1H),3.79(s,3H),3.58-3.56(m,1H),2.59-2.50(m,1H),1.83-1.71(m,7H),1.53-1.51(m,1H),1.38-1.36(m,2H),1.31-1.28(m,6H),0.91(m,6H).
Example 59:
synthesis of final product 59:
5b (200mg, 0.55mmol) was dissolved in N, N-dimethylformamide (30 mL), followed by the addition of N-acetyl-D-leucine (112mg, 0.65mmol), 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate (178mg, 0.47mmol) and N, N-diisopropylethylamine (111mg, 0.86mmol), the system was stirred at room temperature overnight, and TLC monitored that no starting material remained. To the reaction solution was added water (100 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (dichloromethane: methanol = 50. MS m/z (ESI) 517.2[ m ] +H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.59(s,1H),8.33(s,1H),8.07(d,J=6.0Hz,1H),7.93(dd,J=10.2,Hz,J=8.4Hz,2H),7.34(dd,J=7.8Hz,J=7.2Hz,1H),7.11-7.10(m,1H),6.92-6.89(m,1H),4.24-4.23(m,1H),3.80(s,3H),3.57-3.55(m,1H),2.59-2.50(m,1H),1.83-1.71(m,7H),1.53-1.51(m,1H),1.38-1.36(m,2H),1.30-1.27(m,6H),0.91(m,6H).
Example 60:
synthesis of intermediate 60 a:
5b (500mg, 1.385mmol) was dissolved in N, N-dimethylformamide (15 mL), followed by addition of 1-Boc-4-piperidinecarboxylic acid (380mg, 1.66mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (787 mg, 2.07mmol) and N, N-diisopropylethylamine (687 uL, 4.15mmol), and the whole was stirred at room temperature for 10 hours, and TLC was used to monitor that no starting material remained. Water (50 mL) was added to the reaction mixture, which was stirred for 30 minutes, filtered, and dried over anhydrous sodium sulfate to give 60a (623 mg, yield 79%).
Synthesis of intermediate 60 b:
60a (623mg, 1.09mmol) was dissolved in methylene chloride (20 mL), followed by trifluoroacetic acid (5 mL), and the whole was stirred at room temperature for 2 hours with no residue on TLC. Water (100 mL) was added to the reaction mixture, sodium bicarbonate was added to adjust the pH to 8, ethyl acetate was extracted (50 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure to give 60b (500 mg, 97% yield).
Synthesis of final product 60:
60b (300mg, 0.63mmol) was dissolved in methanol (10 mL), followed by the addition of 1mL of aqueous formaldehyde, sodium triacetoxyborohydride (269mg, 1.27mmol), and 1 drop of acetic acid. The whole was stirred at room temperature and reacted for 10 hours, with no starting material remaining as monitored by TLC. Water (50 mL) was added to the reaction mixture, the pH was adjusted to 8 with sodium bicarbonate, extracted with ethyl acetate (50 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure to give the final product 60 (214 mg, 70% yield). MS m/z (ESI) 487.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.59(s,1H),8.34(s,1H),8.08(d,J=4.8Hz,1H),7.68(d,J=7.8Hz,1H),7.35(d,J=7.8Hz,1H),7.11(d,J=11.4Hz,1H),6.93(d,J=8.4Hz,1H),3.79(s,3H),3.58-3.56(m,1H),2.76-2.74(m,2H),2.62-2.58(m,1H),2.51(s,3H),1.99-1.96(m,1H),1.86-1.84(m,1H),1.81-1.72(m,5H),1.57-1.53(m,4H),1.31-1.24(m,3H),1.18-1.08(m,1H).
Example 61:
synthesis of end product 61:
5b (0.07g, 0.19mmol) was dissolved in N, N-dimethylformamide (10 mL), cyclopropylacetic acid (0.02g, 0.21mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (0.09g, 0.23mmol) and N, N-diisopropylethylamine (0.05g, 0.38mmol) were added and reacted at room temperature for 8 hours, and TLC monitored that no starting material remained. In the reverse directionTo the reaction solution was added water (20 mL), extracted with ethyl acetate (15 mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and subjected to column chromatography separation and purification (dichloromethane: methanol =20 1) to obtain a final product 61 (0.04 g, yield 47%). MS m/z (ESI) 444.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.58(s,1H),δ8.33(s,1H),8.07(d,J=5.4Hz,1H),7.63(d,J=8.4Hz,1H),7.35-7.33(m,1H),7.11-7.01(m,1H),6.93-6.90(m,1H),3.79(s,3H),3.60-3.57(m,1H),2.60-2.61(m,1H),1.94-1.93(m,2H),1.88-1.75(m,4H),1.32-1.23(m,4H),1.10-1.08(m,1H),0.54-0.45(m,2H),0.19-0.15(m,2H).
Example 62:
synthesis of end product 62:
5b (0.07g, 0.19mmol) was dissolved in dichloromethane (20 mL), cyclopropylcarboxylic acid (0.02g, 0.21mmol), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (0.07g, 0.38mmol) and 4-dimethylaminopyridine (2.3mg, 0.019mmol) were added and reacted at room temperature for 8 hours with TLC monitoring no residue of starting material. The reaction solution was washed with water and an aqueous sodium bicarbonate solution in this order, and dried over anhydrous sodium sulfate. The organic phase was directly separated by column chromatography (dichloromethane: methanol =50 = 1) to yield the final product 62 (0.04 g, 48% yield). MS m/z (ESI) 430.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.58(s,1H),8.32(s,1H),8.08(d,J=5.4Hz,1H),7.99(d,J=8.4Hz,1H),7.36-7.33(m,1H),7.11-7.09(m,1H),6.93-6.90(m,1H),3.79(s,3H),3.60-3.58(m,1H),2.59-2.50(m,1H),1.89-1.77(m,4H),1.50-1.47(m,1H),1.34-1.26(m,3H),1.10-1.08(m,1H),0.64-0.60(m,4H).
Example 63:
synthesis of the end product 63:
5b (0.10g, 0.28mmol) was dissolved in N, N-dimethylformamide (25 mL), followed by addition of 2-cyclopropyl-2-carbonylacetic acid (35mg, 0.31mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (118mg, 0.31mmol) and N, N-diisopropylethylamine (0.07g, 0.55mmol). The system was stirred at room temperature and monitored by TLC until no starting material remained. To the reaction solution was added water (50 mL), extracted with dichloromethane (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and column chromatography was performed to separate and purify (dichloromethane: methanol =100 1-40) to obtain a final product 63 (54 mg, yield 43%). MS m/z (ESI) 458.1[ m ] +H] + 。
1 H NMR(600MHz,CDCl 3 )δ8.55(s,1H),8.32(d,J=6.0Hz,1H),8.11(s,1H),7.28-7.25(m,1H),6.94(d,J=8.4Hz,1H),6.77-6.71(m,2H),3.86-3.82(m,1H),3.81(s,3H),3.10-3.08(m,1H),2.48-2.46(m,1H),2.25(d,J=12.0Hz,1H),2.04-1.95(m,3H),1.53-1.47(m,3H),1.26-1.24(m,1H),1.18-1.15(m,4H).
Example 64:
synthesis of final product 64:
dissolve 5b (0.16g, 0.44mmol) in dichloromethane (20 mL), add triethylamine (0.05g, 0.53mmol), add acryloyl chloride (0.05g, 0.53mmol) in ice bath, react at room temperature for 4 hours, TLC monitor no residual starting material. Water (30 mL) was added to the reaction solution, dichloromethane was extracted (20 mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the crude product was separated by column chromatography (dichloromethane: methanol =50 1) to obtain a final product 64 (0.10 g, yield 55%). MS m/z (ESI) 416.2[ 2 ], [ M ] +H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.59(s,1H),8.31(s,1H),8.06(d,J=6.0Hz,1H),8.02(d,J=7.8Hz,1H),7.34-7.31(m,1H),7.10-7.07(m,1H),6.91-6.89(m,1H),6.18-6.14(m,1H),6.06-6.03(m,1H),5.55-5.53(m,1H),3.77(s,3H),3.65-3.63(m,1H),2.61-2.60(m,1H),1.90-1.77(m,4H),1.34-1.08(m,4H).
Example 65:
synthesis of end product 65:
5b (180mg, 0.50mmol) and propiolic acid (70mg, 1.00mmol) are dissolved in N, N-dimethylformamide (20 mL), 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethylurea hexafluorophosphate (380mg, 1.00mmol) and N, N-diisopropylethylamine (0.25mL, 1.50mmol) are sequentially added, and the reaction system is reacted at room temperature for 15 hours, and no residual raw material is detected by TLC. To the reaction solution was added water (100 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (petroleum ether: ethyl acetate = 4:1-2:1) to obtain the final product 65 (200 mg, yield 97%). MS m/z (ESI) 414.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.60(s,1H),8.73(d,J=7.8Hz,1H),8.33(s,1H),8.08(d,J=5.4Hz,1H),7.35(dd,J=8.4Hz,J=7.8Hz,1H),7.11(dd,J=11.4Hz,J=2.4Hz,1H),6.92-6.90(m,1H),4.12(s,1H),3.79(s,3H),3.64-3.62(m,1H),2.61-2.57(m,1H),1.85-1.74(m,4H),1.35-1.23(m,4H).
Example 66:
synthesis of final product 66:
5b (0.25g, 0.69mmol) was dissolved in N, N-dimethylformamide (20 mL), and trans-4-dimethylaminocrotonate (0.13g, 0.76mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (0.32g, 0.83mmol), N, N-diisopropylethylamine (0.27g, 2.07mmol) were added and reacted at room temperature for 8 hours, with TLC monitoring no residue of starting material. Water (30 mL) was added to the reaction mixture, extracted with ethyl acetate (20 mL. Times.5), and the organic phases were combined and dried over anhydrous sodium sulfate. Direct column chromatography of the organic phase (dichloromethane: methanol =)10, 1) to give the final product 66 (0.18 g, 55% yield). MS m/z (ESI) 472.2[ deg. ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.61(s,1H),8.33(s,1H),8.08(d,J=6.0Hz,1H),8.01(d,J=6.0Hz,1H),7.35-7.33(m,1H),7.11-7.09(m,1H),6.93-6.90(m,1H),6.56-6.51(m,1H),6.04(d,J=15.0Hz,1H),3.78(s,3H),3.66(m,1H),3.14(s,2H),2.64-2.60(m,1H),2.25(s,6H),1.91-1.90(m,1H),1.84-1.78(m,3H),1.35-1.13(m,4H).
Example 67:
synthesis of final product 67:
26c (99mg, 0.26mmol) was dissolved in N, N-dimethylformamide (5 mL), followed by the addition of 1-cyano-1-cyclopropanecarboxylic acid (44mg, 0.40mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (152mg, 0.40mmol) and N, N-diisopropylethylamine (131. Mu.L, 0.79 mmol), and the entire system was stirred at room temperature overnight with no residual starting material detected by TLC. Water (100 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50: 1-20. MS m/z (ESI) 473.2[ m ] +H] + 。
1 H NMR(600MHz,CDCl 3 )δ8.32-8.31(m,1H),8.13(s,1H),7.18-7.15(m,1H),6.84-6.81(m,1H),6.38-6.37(m,1H),3.90-3.85(m,1H),3.79(s,3H),2.49-2.47(m,1H),2.27-2.25(m,1H),2.01-1.94(m,3H),1.70-1.62(m,2H),1.60-48(m,5H),1.32-1.23(m,1H).
Example 68:
synthesis of final product 68:
26c (99mg, 0.26mmol) was dissolved in NN-dimethylformamide (5 mL), followed by glycolic acid (20mg, 0.26mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (152mg, 0.40mmol) and N, N-diisopropylethylamine (131. Mu.L, 0.79 mmol), the whole was stirred at room temperature overnight with no starting material remaining by TLC. Water (100 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50: 1-20. MS m/z (ESI) 438.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.60(s,1H),8.35(s,1H),8.08(d,J=6.0Hz,1H),7.58-7.56(m,1H),7.53-7.51(m,1H),7.37-7.34(m,1H),5.38(t,J=6.0Hz,1H),3.82(s,5H),3.74-3.63(m,1H),2.69-2.61(m,1H),1.85-1.70(m,4H),1.33-1.22(m,4H).
Example 69:
synthesis of intermediate 69 a:
5b (0.07g, 0.19mmol) was dissolved in absolute ethanol (20 mL), triethylamine (0.02g, 0.19mmol) and carbon disulfide (0.02g, 0.26mmol) were added to the solution, and the reaction was carried out at room temperature for 30 minutes, di-tert-butyl dicarbonate (0.04g, 0.19mmol) and 4-dimethylaminopyridine (2.3mg, 0.019mmol) were added to the reaction solution and reacted at room temperature for 4 hours, and TLC was used to monitor that no starting material remained. Ethanol was evaporated under reduced pressure and the residue was directly subjected to column chromatography (dichloromethane: methanol =50: 1) to give 69a (0.03 g, yield 39%).
Synthesis of end product 69:
69a (0.03g, 0.07mmol) was dissolved in absolute ethanol (10 mL), 2.0M aminoethanol solution (0.004g, 0.28mmol) was added, heated to 90 deg.C, and the tube was sealed for 4 hours, and TLC monitored that no starting material remained. The solvent was distilled off under reduced pressure, the residue was dissolved in anhydrous ethanol (10 mL), a 40% aqueous solution of dichloroacetaldehyde (0.005g, 0.07mmol) was added, the reaction was heated to 90 ℃ for 8 hours, and the starting material was monitored by TLC to be free from residue. Reduced pressure steaming and removingAnhydrous ethanol, and the residue was directly subjected to column chromatography (dichloromethane: methanol =25 = 1) to obtain the final product 69 (0.02 g, yield 64%). MS m/z (ESI) 445.1[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.67(s,1H),8.33(s,1H),8.33-8.08(m,1H),7.50(d,J=7.8Hz,1H),7.35-7.33(m,1H),7.11(d,J=2.4Hz,1H),7.09(d,J=2.4Hz,1H),6.99-6.90(m,1H),6.58(d,J=4.2Hz,1H),3.79(s,3H),3.52-3.50(m,1H),2.63(m,1H),2.14-2.12(m,1H),2.02-2.00(m,1H),1.82-1.78(m,2H),1.36-1.30(m,3H),1.14-1.12(m,1H).
Example 70:
synthesis of intermediate 70 a:
2a (160mg, 0.68mmol) was dissolved in tetrahydrofuran (10 mL), followed by addition of phenyl chloroformate (191mg, 1.22mmol), potassium carbonate (187mg, 1.35mmol), the entire system was stirred overnight, and TLC monitored that no starting material remained. The reaction was directly purified by column chromatography (petroleum ether: ethyl acetate =10, 1-2:1) to afford 70a (200 mg, 83% yield).
Synthesis of intermediate 70 b:
(R) -1-tert-Butoxycarbonyl-3-aminopiperidine (1.00g, 5.00mmol) was dissolved in methylene chloride (30 mL), followed by the addition of triethylamine (1.52g, 15.0 mmol), acetic anhydride (701. Mu.L, 7.50 mmol). The system was stirred at room temperature overnight and no starting material was left as monitored by TLC. Water (50 mL) was added to the reaction mixture, dichloromethane was extracted (50 mL. Times.3), the organic phases were combined, washed with saturated aqueous sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure to give 70b (1.17 g, 97% yield).
Synthesis of intermediate 70 c:
70b (1.17g, 4.83mmol) was dissolved in dichloromethane (30 mL), followed by the addition of 10mL of trifluoroacetic acid in an ice-water bath, and the whole was stirred at room temperature for 3 hours. The solvent was removed under reduced pressure to give 70c (1.50 g) which was used directly in the next step.
Synthesis of final product 70:
70a (235mg, 0.66mmol) was dissolved in tetrahydrofuran (10 mL) followed by 70c (339mg, 1.32mmol) and triethylamine (458. Mu.L, 3.30 mmol). The system was stirred at room temperature for 10 hours and TLC monitored that no starting material remained. To the reaction solution was added water (50 mL), extracted with dichloromethane (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and column chromatography was performed to separate and purify (dichloromethane: methanol =40, 1-20. MS m/z (ESI) 405.2[ 2 ], [ M + H ]] + 。
1 H NMR(600MHz,DMSO-d 6 )δ9.22(s,1H),8.25(s,1H),7.84(d,J=7.2Hz,1H),7.75(d,J=5.4Hz,1H),7.33(t,J=7.8Hz,1H),7.10(d,J=11.4Hz,1H),6.92(t,J=7.8Hz,1H),3.93(d,J=12.6Hz,1H),3.83(d,J=12.6Hz,1H),3.80(s,3H),3.64-3.59(m,1H),2.98-2.93(m,1H),2.79-2.76(m,1H),1.82-1.79(m,4H),1.71-1.69(m,1H),1.45-1.36(m,2H).
Example 71:
synthesis of intermediate 71 a:
1a (0.40g, 1.58mmol) was dissolved in N, N-dimethylformamide (30 mL), followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.38g, 1.58mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (0.72g, 1.90mmol) and N, N-diisopropylethylamine (0.41g, 3.16mmol), the whole was stirred at room temperature overnight and TLC monitored that no starting material remained. Water (100 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (dichloromethane: methanol =50: 1-20.
Synthesis of intermediate 71 b:
71a (0.45g, 0.94mmol) was dissolved in dichloromethane (30 mL), followed by addition of 2mL of trifluoroacetic acid under ice-water bath, the whole system was stirred at room temperature overnight, TLC detected no remaining starting material, water (100 mL) was added to the reaction solution, PH =9-10 was adjusted with saturated aqueous sodium bicarbonate solution, dichloromethane was extracted (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and after purification by column chromatography (dichloromethane: methanol = 50.
Synthesis of final product 71:
71b (0.31g, 0.82mmol) was dissolved in dichloromethane (35 mL), followed by the addition of acetic anhydride (0.25g, 2.47mmol) and triethylamine (0.25g, 2.47mmol), the system was stirred at room temperature and TLC monitored that no starting material remained. Water (50 mL) was added to the reaction solution, dichloromethane was extracted (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and the product was purified by column chromatography (dichloromethane: methanol =50: 1-10. MS m/z (ESI) 420.1[ 2 ], [ M + H ]] + 。 1 H NMR(600MHz,CDCl 3 )δ 1 H NMR(600MHz,DMSO-d 6 )δ10.69(s,1H),8.41(s,1H),8.05(s,1H),7.78(d,J=7.8Hz,1H),7.25(d,J=7.2Hz,1H),7.09(d,J=2.4Hz,1H),6.91-6.88(m,1H),3.76(s,3H),3.57-3.54(m,1H),2.62-2.59(m,1H),1.86-1.84(m,1H),1.76-1.74(m,6H),1.31-1.23(m,3H),1.07-1.05(m,1H).
Example 72:
synthesis of intermediate 72 a:
6-bromo-1-methyl-1H-indazole (211mg, 1.00mmol), bis-pinacoldiborane (508mg, 2.00mmol), potassium acetate (294mg, 3.00mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (73mg, 0.10mmol) were dissolved in 1,4-dioxane (20 mL). The reaction system is protected by nitrogen and reacted for 4 hours at 100 ℃, no raw material is left by TLC detection, and heating is stopped. To the reaction solution was added water (100 mL), extracted with ethyl acetate (100 mL × 3), the organic phases were combined, washed with saturated sodium chloride (100 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and separated and purified by column chromatography (petroleum ether-petroleum ether: ethyl acetate = 4:1) to give 72a (240 mg, yield 93%).
Synthesis of intermediate 72 b:
72a (258mg, 1.00mmol) was dissolved in diethylene glycol dimethyl ether (40 mL), 5-fluoro-4-iodopyridin-2-amine (286mg, 1.20mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (73mg, 0.10mmol), potassium carbonate (414mg, 3.00mmol) and water (10 mL) were added at room temperature, the reaction system was stirred at 80 ℃ for 4 hours, and TLC detection showed no remaining starting material, and the reaction was stopped. To the reaction solution was added water (100 mL), extracted with ethyl acetate (100 mL × 3), the organic phases were combined, washed with saturated sodium chloride (100 mL × 2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (petroleum ether-petroleum ether: ethyl acetate = 1:1) to give 72b (175 mg, yield 72%).
Synthesis of intermediate 72 c:
72b (175mg, 0.72mmol) and (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (350mg, 1.44mmol) were dissolved in N, N-dimethylformamide (20 mL), 2- (7-oxybenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (548mg, 1.44mmol) and diisopropylethylamine (0.36mL, 2.16mmol) were added in this order, the reaction system was reacted at room temperature for 4 hours with no remaining starting material detected by TLC, water (50 mL) was added to the reaction solution, ethyl acetate was extracted (100 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (100 mL. Times.2), dried over anhydrous sodium sulfate, the solvent was removed under reduced pressure, and purified by column chromatography (petroleum ether: ethyl acetate = 4:1-2:1), yielding 72c (200 mg, 56%).
Synthesis of final product 72:
72c (200mg, 0.40mmol) was dissolved in dichloromethane (50 mL) and trifluoroacetic acid (5.00 mL) was added and stirred at room temperature for 4 hours, and no starting material remained as detected by TLC. Adding saturated sodium carbonate aqueous solution into the system, and adjusting the pH value of the reaction system to be alkalescent. After separation, extraction with dichloromethane (100 mL. Times.3), concentration of the combined organic phases to a volume of about 50mL, addition of triethylamine (2.00 mL) and acetic anhydride (2.00 mL) for 30 minutes at room temperature, and washing of the organic phase with aqueous sodium carbonate. Separating, extracting with dichloromethane (20 mL. Times.3), combining the organic phases, washing with saturated sodium chloride (50 mL. Times.2), drying over anhydrous sodium sulfate, removing the solvent under reduced pressure, and separating by column chromatography (ethyl acetate) to give the final product 72 (R)135mg, 83% yield). MS m/z (ESI) 410.2[ 2 ] M + H] + 。
1 H NMR(600MHz,DMSO-d 6 )δ10.64(s,1H),8.44(s,1H),8.36(s,1H),8.14(s,1H),7.96(s,1H),7.90(s,1H),7.79(d,J=7.8Hz,1H),7.33(d,J=7.8Hz,1H),4.12(s,3H),3.59-3.58(m,1H),2.99-2.95(m,1H),1.91(s,3H),1.82-1.78(m,4H),1.14-1.12(m,4H).
Example 73:
synthesis of final product 73:
23c (101mg, 0.25mmol) was dissolved in 1,4 dioxane (10 mL) and water (5 mL) followed by the addition of 1-isopropyl-4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) -1H-pyrrolo [2,3-b]Pyridine (106mg, 0.37mmol), [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride (15mg, 0.02mmol) and potassium carbonate (68mg, 0.49mmol) were replaced with nitrogen three times to place the whole system under an atmosphere of nitrogen. The system was stirred at 100 ℃ under reflux, reacted for 4 hours, and TLC monitored that no starting material remained. The reaction was directly purified by column chromatography (dichloromethane: methanol =50, 1-10) to give the final product 73 (60 mg, 55% yield). MS m/z (ESI) 438.2[ 2 ], [ M + H ]] +
1 H NMR(600MHz,DMSO-d6)δ10.70(s,1H),8.51(s,1H),8.41-8.38(m,2H),7.83(d,J=3.6Hz,1H),7.79(d,J=7.8Hz,1H),7.25-7.24(m,1H),6.50-6.49(m,1H),5.17-5.15(m,1H),3.58-3.56(m,1H),2.64-2.60(m,1H),1.90-1.88(m,1H),1.77-1.76(m,6H),1.51(s,3H),1.49(s,3H),1.40-1.25(m,3H),1.15-1.05(m,1H).
Example 74:
synthesis of intermediate 74 a:
2,3-dichloropyridine-4-boronic acid (1.05g, 5.50mmol) was dissolved in dioxane (30 mL) and water (5 mL), followed by the addition of 5-fluoro-4-iodo-pyridin-2-amine (1.00g, 4.20mmol), 1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex (0.34g, 0.42mmol), potassium carbonate (1.74g, 12.6 mmol), heated to 100 ℃ under nitrogen for 8 hours with TLC monitoring no residue of starting material. Stopping heating, and cooling to room temperature. The reaction was directly subjected to column chromatography (n-hexane: ethyl acetate = 1:1) to give intermediate 74a (0.40 g, 37% yield).
Synthesis of intermediate 74 b:
74a (0.13g, 0.50mmol) was dissolved in acetonitrile (10 mL), followed by addition of (1S, 3R) -3- [ (tert-butoxycarbonyl) amino ] cyclohexanecarboxylic acid (0.116g, 0.65mmol), N, N, N ', N' -tetramethylchloroformamidine hexafluorophosphate (0.17g, 0.60mmol) and N-methylimidazole (0.14g, 1.75mmol), reaction at room temperature for 12 hours, TLC monitoring no residual starting material. The reaction solution was directly subjected to column chromatography for separation and purification (n-hexane: ethyl acetate = 2:1) to obtain 74b (0.12 g,50% yield).
Synthesis of intermediate 74 c:
74b (0.12g, 0.25mmol) was dissolved in dichloromethane (20 mL), and 4mL of trifluoroacetic acid was added to react at room temperature for 4 hours, and TLC monitored that no starting material remained. The reaction solution was washed with water (30 mL. Times.3), the aqueous phases were combined, the pH of the aqueous phase was adjusted to 8-9 with sodium carbonate, dichloromethane was extracted (30 mL. Times.3), the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), and dried over anhydrous sodium sulfate. The solvent was removed by concentration under reduced pressure to give 74c (0.08 g, yield 84%).
Synthesis of end product 74:
74c (0.08g, 0.21mmol) was dissolved in dichloromethane (20 mL) followed by the addition of acetic anhydride (0.04g, 0.42mmol), triethylamine (0.04g, 0.42mmol) and reaction at room temperature for 2 hours with TLC monitoring no residue of starting material. The reaction solution was directly subjected to column chromatography (dichloromethane: methanol =50 = 1) to obtain a final product 74 (0.02 g, yield 22%). MS m/z (ESI) 426.2[ 2 ] M + H] + .
1 H NMR(600MHz,DMSO-d6)δ10.81(s,1H),8.55-8.54(m,2H),8.18(d,J=5.4Hz,1H),7.80(d,J=7.8Hz,1H),7.6(d,J=5.4Hz,1H),3.60-3.54(m,1H),2.64-2.60(m,1H),1.90-1.88(m,1H),1.77-1.76(m,6H),1.40-1.20(m,3H),1.19-1.05(m,1H).
Example 76:
synthesis of intermediate 76 a:
5b (500mg, 1.385mmol) was dissolved in N, N-dimethylformamide (15 mL), followed by addition of 1- [ (tert-butoxy) carbonyl ] -3-cyanoazetidine-3-carboxylic acid (375mg, 1.66mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (787 mg, 2.07mmol) and N, N-diisopropylethylamine (687 uL, 4.15mmol), and the whole was stirred at room temperature for 10 hours and TLC monitored that no starting material remained. Water (50 mL) was added to the reaction mixture, which was stirred for 30 minutes, filtered, and dried over anhydrous sodium sulfate to give 76a (497 mg, 63% yield).
Synthesis of end product 76:
76a (497mg, 0.87mmol) was dissolved in dichloromethane (20 mL), followed by trifluoroacetic acid (5 mL) and the whole was stirred at room temperature for 2 hours with no residue on TLC. To the reaction solution was added water (100 mL), sodium bicarbonate was added to adjust PH to 8, ethyl acetate was extracted (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure to perform column chromatography separation (dichloromethane: methanol =40: 1) to obtain a final product 76 (396 mg, yield 97%). MS m/z (ESI) 470.2[ 2 ] M + H] + 。 1 H NMR(600MHz,CD 3 OD)δ8.38(s,1H),7.75(d,J=5.4Hz,1H),7.50-7.42(m,1H),7.04(d,J=10.8Hz,1H),6.96-6.86(m,1H),4.60-4.32(m,4H),3.93-3.76(m,4H),2.70(m,1H),2.23(m,1H),2.00(m,3H),1.71-1.46(m,3H),1.43-1.27(m,1H).
Example 77:
synthesis of end product 77:
76 (296mg, 0.63mmol) was dissolved in methanol (10 mL), followed by the addition of 1mL of aqueous formaldehyde, sodium triacetoxyborohydride (249mg, 1.27mmol), and 1 drop of acetic acid.The whole was stirred at room temperature and reacted for 10 hours, with no starting material remaining as monitored by TLC. Water (50 mL) was added to the reaction mixture, sodium bicarbonate was adjusted to pH 8, ethyl acetate extraction (50 mL. Times.3) was performed, the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure to give the final product 77 (186 mg, 61% yield). MS m/z (ESI) 484.2[ 2 ] M + H] +
1 H NMR(600MHz,DMSO-d 6 )δ10.56(s,1H),8.32(s,1H),8.05(d,J=4.8Hz,1H),8.10(d,J=7.8Hz,1H),7.34-7.31(m,1H),7.09(s,1H),6.91-6.88(m,1H),3.79(s,3H),3.68-3.56(m,1H),2.98-2.73(m,4H),2.59-2.55(m,1H),2.42(s,3H),1.81-1.67(m,4H),1.30-1.09(m,4H).
Example 78:
synthesis of intermediate 78 a:
5b (500mg, 1.385mmol) was dissolved in N, N-dimethylformamide (15 mL), followed by addition of 1- [ (tert-butoxy) carbonyl ] -3-cyanoazetidine-3-carboxylic acid (375mg, 1.66mmol), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (787 mg, 2.07mmol) and N, N-diisopropylethylamine (687 uL, 4.15mmol), and the whole was stirred at room temperature for 10 hours and TLC monitored that no starting material remained. Water (50 mL) was added to the reaction mixture, which was stirred for 30 minutes, filtered, and dried over anhydrous sodium sulfate to give 78a (504 mg, yield 61%).
Synthesis of the end product 78:
78a (504mg, 0.84mmol) was dissolved in dichloromethane (20 mL), trifluoroacetic acid (5 mL) was added, the whole was stirred at room temperature for 2 hours, and no starting material remained by TLC. To the reaction solution was added water (100 mL), sodium bicarbonate was added to adjust PH to 8, ethyl acetate was extracted (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure to perform column chromatography separation (dichloromethane: methanol =40 1) to obtain the final product 78 (384 mg, yield 92%). MS m/z (ESI) 498.7[ 2 ] M + H] + 。 1 H NMR(600MHz,CD3OD)δ8.32(s,1H),8.07(d,J=8.4Hz,1H),7.35-7.33(m,1H),7.11-7.10(m,1H),7.08-6.91(m,1H),3.78(s,3H),3.78-3.66(m,1H),3.30-3.28(m,2H),2.99-2.97(m,2H),2.62-2.52(m,1H),2.30-2.22(m,4H),1.79-1.77(m,4H),1.46-1.43(m,1H),1.29-1.26(m,3H).
Example 79:
synthesis of final product 79:
5b (0.16g, 0.43mmol) was dissolved in N, N-dimethylformamide (30 mL), followed by the addition of N, N-diethylglycinate hydrochloride (109mg, 0.65mmol), 2- (7-oxybenzotriazole) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (178mg, 0.47mmol) and N, N-diisopropylethylamine (111mg, 0.86mmol), the system was stirred at room temperature overnight, and TLC monitored for no residual starting material. Water (100 mL) was added to the reaction solution, extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (dichloromethane: methanol = 50. MS m/z (ESI) 475.3[ 2 ] M + H] +
1 H NMR(600MHz,CDCl 3 )δ8.23(d,J=7.8Hz,1H).8.13(s,1H),8.07(s,1H),7.40(m,1H),7.277.22(m,1H),6.87-6.60(m,2H),3.94-3.84(m,1H),3.81(s,3H),3.01(s,2H),2.54(m,4H),2.45(m,1H),2.22(m,1H),1.96(m,3H),1.56-1.36(m,3H),1.26-1.12(m,1H),1.01(m,6H).
Example 80:
synthesis of intermediate 80 a:
5b (0.30g, 0.83mmol) was dissolved in dichloromethane (10 mL) followed by addition of triethylamine (420mg, 4.15mmol) and chloroacetyl chloride (186mg, 1.66mmol) at 0 deg.C, the system was stirred at room temperature overnight and TLC monitored that no starting material remained. To the reaction solution was added water (20 mL), extracted with ethyl acetate (10 mL × 3), the organic phases were combined, washed with saturated sodium chloride (10 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (n-hexane: ethyl acetate = 1:0-0:1) to give 80a (200 mg, yield 55%).
Synthesis of final product 80:
80a (200mg, 0.46mmol) was dissolved in N, N-dimethylformamide (10 mL), potassium thioacetate (78.25mg, 0.69mmol) was added, and the reaction was carried out at room temperature for 2 hours, and no residue was left by TLC. The reaction solution was added to 1M aqueous HCl (10 mL), followed by water (10 mL) and dichloromethane (15 mL). Extraction with dichloromethane (10 mL × 3), combination of organic phases, washing with saturated sodium chloride (10 mL × 2), drying over anhydrous sodium sulfate, and then removal of the solvent under reduced pressure, and separation and purification of the crude product by column chromatography (n-hexane: ethyl acetate = 1:0-0:1) gave the final product 80 (200 mg, 91% yield). MS m/z (ESI) 478.2[ 2 ] M + H] +
1 H NMR(600MHz,DMSO-d 6 )δ10.58(s,1H),8.33(s,1H),8.07(d,J=5.4Hz,1H),7.59(d,J=8.4Hz,1H),7.35-7.34(m,1H),7.33-7.11(m,1H),7.09-6.90(m,1H),4.39(s,2H),3.78(s,3H),3.65-3.63(m,1H),2.66-2.60(m,1H),2.58(s,3H),1.85-1.69(m,4H),1.24-1.14(m,4H).
Example 81:
synthesis of end product 81:
80 (220mg, 0.46mmol) was dissolved in methanol (5 mL), potassium carbonate (317mg, 2.30mmol) was added thereto and reacted at room temperature for 20 minutes, and then heated to 55 ℃ and reacted for 40 minutes. TLC monitored no starting material remaining. To the reaction solution was added water (20 mL), extracted with ethyl acetate (10 mL × 3), the organic phases were combined, washed with saturated sodium chloride (10 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and the crude product was purified by column chromatography (n-hexane: ethyl acetate = 1:0-0:1) to obtain a final product 81 (41 mg, yield 20%). MS m/z (ESI) 436.2[ deg. ] M + H] +
1 H NMR(600MHz,DMSO-d 6 )δ10.55(s,1H),8.31(s,1H),8.07(d,J=5.4Hz,1H),7.56(d,J=9.0Hz,1H),7.35-7.32(m,1H),7.09-7.07(m,1H),6.91-6.88(m,1H),3.89-3.84(m,4H),3.66-3.63(m,1H),3.24-3.22(m,2H),2.61-2.57(m,1H),1.83-1.68(m,4H),1.34-1.26(m,4H).
Example 82:
synthesis of end product 82:
78 (300mg, 0.60mmol) was dissolved in methanol (10 mL), followed by addition of 1mL of aqueous formaldehyde solution, sodium triacetoxyborohydride (235mg, 1.20mmol), and 1 drop of acetic acid. The whole was stirred at room temperature and reacted for 10 hours, with no starting material remaining as monitored by TLC. Water (50 mL) was added to the reaction mixture, sodium bicarbonate was adjusted to pH 8, ethyl acetate extraction (50 mL. Times.3) was performed, the organic phases were combined, washed with saturated sodium chloride (50 mL. Times.2), dried over anhydrous sodium sulfate, and the solvent was removed under reduced pressure to give the final product 82 (205 mg, 67% yield). MS m/z (ESI) 512.2[ 2 ] M + H] +
1 H NMR(600MHz,DMSO-d 6 )δ10.28(s,1H),8.28(s,1H),8.22-8.09(m,1H),8.05(d,J=8.4Hz,1H),7.32-7.30(m,1H),7.09-7.02(m,1H),6.91-6.87(m,1H),3.80(s,3H),3.75-3.66(m,1H),3.57-3.52(m,1H),3.16-2.96(m,2H),2.81-2.59(m,4H),2.50-2.48(m,2H),2.43-2.31(m,3H),2.00-1.74(m,4H),1.52-1.48(m,1H),1.41-1.22(m,3H).
Example 83:
synthesis of end product 49:
5b (0.20g, 0.55mmol) was dissolved in N, N-dimethylformamide (30 mL), followed by addition of 2-hydroxyisobutyric acid (68mg, 0.65mmol), 2- (7-oxybenzotriazole) -N, N, N ', N' -tetramethylurea hexafluorophosphate (178mg, 0.47mmol) and N, N-diisopropylethylamine (111mg, 0.86mmol), the system was stirred at room temperature overnight, and the starting material was monitored by TLC to be free of residue. Water (10) was added to the reaction solution0 mL), extracted with ethyl acetate (50 mL × 3), the organic phases were combined, washed with saturated sodium chloride (50 mL × 2), dried over anhydrous sodium sulfate, and then the solvent was removed under reduced pressure, and the crude product was isolated and purified by column chromatography (dichloromethane: methanol =50, 1-20) to give final product 83 (81 mg, 33% yield). MS m/z (ESI) 448.2[ 2 ], [ M + H ]] +
1 H NMR(600MHz,CDCl 3 )δ8.31(s,1H),8.24(d,J=8.4Hz,1H),8.12(s,1H),7.26-7.24(m,1H),6.76-6.73(m,2H),6.63(d,J=12.6Hz,1H),3.90-3.82(m,1H),3.81(s,3H),2.65(s,1H),2.56-2.42(m,1H),2.26-2.23(m,1H),2.00-1.89(m,3H),1.61-1.49(m,2H),1.45(d,J=4.8Hz,6H),1.30-1.18(m,1H).
Examples 75, 84 to 90:
compounds 75, 84-90 were synthesized according to the methods of examples 1-72, selecting the corresponding starting materials, and the structures were as follows:
test example 1: assay of inhibitory Effect of Compounds of the present application on CDK9, CDK1, CDK2, CDK4, CDK5, CDK6 and CDK7
1. Purpose of experiment
Testing the inhibitory effect of compounds on CDK1/2/4/5/6/7/9 kinase and formulating an effective IC 50 The value is obtained.
CDKs family of assays
CDK1/CDK2/CDK4/CDK5/CDK6/CDK7/CDK9
Table 1: information relating to kinases, substrates and ATP in vitro assays
3. Detection process
3.1 dilution of the Compound
Compounds were diluted 11 concentrations in DMSO, 3-fold, with the highest concentration of test compound being 10 μ M.
3.2 enzymatic reactions
Compounds (50 nL) dissolved in DMSO were transferred to an enzyme reaction plate using the sonic technique (Echo). mu.L CDK enzyme dilution was added to the enzyme reaction plate, centrifuged and incubated for 10min at room temperature. mu.L of the substrate premix was added to the plate, and the final substrate and ATP concentrations in each well are shown in Table 1. After centrifugation, the reaction was carried out at 30 ℃ for 120 minutes.
3.3 termination reactions and Signal detection
mu.L of stop buffer was added to each well, centrifuged, incubated at room temperature for 120 minutes, and then incubated at 4 ℃ overnight. Reading signal values using HTRF program on Envision instrument and performing data analysis, IC 50 The (inhibitory concentration at 50% of maximal effect) values are expressed in nM. The results are shown in Table 2.
Table 2 inhibitory effects (IC) of the compounds of the present application on CDK1, 2,4, 5, 6, 7 and 9 50 ,nM)
The above tests demonstrate that the compounds of the present application have a selective inhibitory effect on CDK 9.
Test example 2: in vitro inhibition effect of CDK9 inhibitor on human liver cancer cell proliferation
1. Purpose of the experiment
The in vitro inhibition effect of the synthetic compound on human hepatoma cell proliferation is investigated.
2. Principle of experiment
MTT is known under the trade name thiazole blue, a tetrazolium salt of a dye that accepts hydrogen atoms. Amber dehydrogenase in mitochondria of living cells can reduce exogenous MTT into difficultly soluble bluish purple crystals and deposit in cells, while dead cells do not have the function. Dimethyl sulfoxide can be dissolvedThe blue-violet complex in the cell can indirectly reflect the cell number by using an enzyme linked immunosorbent assay detector to measure the light absorption value at 490-550nm wavelength. The amount of MTT crystals formed is proportional to the number of cells over a range of cell numbers. Sequentially diluting the drug to be tested to different concentrations, adding into 96-well plate, acting for a certain time, determining OD value which can reflect the number of living cells, and calculating IC with SPSS19.0 50 The value is obtained.
3. Testing instrument
371 type CO 2 An incubator: thermo
Model IX70-142 inverted fluorescence microscope: olympus
HFsafe-1500 type biological safety cabinet: shanghai Li Shen scientific instruments Co Ltd
Varloskan flash microplate reader: thermo Corp Ltd
A precision electronic balance: type Mettler AL204
TD6 centrifuge: changsha Xiangrui centrifuge, inc
4. Test materials:
4.1 cells and culture Medium
Note: the cell culture conditions were 37 ℃ and 5% CO 2 。
4.2 test materials
Name (R) | Specification of | Production ofManufacturer of the product |
Fetal bovine serum | 500 mL/bottle | Gibco |
PBS | 500 mL/bottle | Gibco |
DMSO | 500 mL/bottle | Kemi Euro Chemicals Ltd |
MTT | 5 g/bottle | Amresco |
0.25%Trypsin-EDTA | 500 mL/bottle | Gibco |
MEM NEAA | 100 mL/bottle | Gibco |
Sodium Pyruvate | 100 mL/bottle | Gibco |
Puromycin | 1mL | Gibco |
4.3 reagent preparation
5mg/mL MTT working solution: 0.5g of MTT was weighed and dissolved in 100mL of PBS, filtered through a 0.22 μm microporous membrane for sterilization, and stored in a refrigerator at 4 ℃ for a long period of time (used within two weeks) or at-20 ℃.
5. Test method
Cells in logarithmic growth phase are inoculated in a 96-well plate (100 mu L/well) in a certain quantity, 100 mu L of culture solution containing different concentration gradient test compounds is added into each well 24h after adherence, each drug concentration is provided with 3 multiple wells, and corresponding blank wells (only culture medium) and normal wells (drug concentration is 0) are arranged. After 72 hours of drug action, MTT working solution (5 mg/mL) was added at 20. Mu.L per well; acting at 37 ℃ for 4 hours, removing supernatant by plate throwing, and adding 150 mu L of DMSO (analytically pure); the plate was wiped clean by shaking and mixing with a micropore oscillator, and the Optical Density (OD) value was measured at 550nm using an microplate reader.
The inhibition rate of cell growth was calculated using the following formula: inhibition (%) = (OD value normal well-OD value administration well)/(OD value normal well-OD value blank well) × 100%
From each concentration inhibition ratio, the half inhibitory concentration IC of the drug was calculated using SPSS19.0 50 . The results of cell proliferation inhibition by the compounds are shown in tables 3-1 and 3-2:
TABLE 3-1 inhibition of cell proliferation assay data (IC) for compounds of the present application 50 ,nM)
Cell species | Compound 45 | Compound 68 | BAY1251152 |
Hep3B-Luc | 71 | 55 | 310 |
SMMC7721-Luc | 119 | 145 | 182 |
PLC/PRF/5 | 292 | 203 | 808 |
TABLE 3-2 inhibition of cell proliferation assay data (IC) for compounds of the present application 50 ,nM)
Cell species | Compound 45 | Lenvatinib |
HepG2-Luc | 29.2 | 1881 |
Hep3B-Luc | 450 | 9826 |
HuH7-Luc | 122 | 1858 |
SMMC7721-Luc | 28.2 | 6349 |
BEL7402-Luc | 85.7 | 8595 |
SK-Hep-1 | 73.4 | >100μM |
Hep3B | 856 | 14733 |
PLC/PRF/5 | 1124 | 1172 |
And (4) experimental conclusion: according to the data in tables 3-1 and 3-2, the compound has strong inhibition effect on various liver cancer cells and in vitro cell inhibition activity IC 50 Basically all are below 1000nM, and the best can reach dozens of nM.
Test example 3: in vitro inhibition of prostate and breast cancer cell proliferation by compounds
1. Testing instrument
371 type CO 2 An incubator: thermo
Model IX70-142 inverted fluorescence microscope: olympus
HFsafe-1500 type biological safety cabinet: shanghai Li Shen scientific instruments Co Ltd
Varloskan flash microplate reader: thermo Corp Ltd
A precision electronic balance: mettler MS105 type
TD6 centrifuge: changsha Xiangrui centrifuge, inc
2. Test materials:
2.1 cells and culture Medium
Note: the cell culture conditions were 37 ℃ and 5% CO 2 。
2.2 test materials
Name (R) | Specification of | Manufacturer of the product |
Fetal bovine serum | 500 mL/bottle | Cia Minhai, lanzhou Ltd |
PBS | 500mL | Gibco |
DMSO | 500 mL/bottle | Kemi Euro Chemicals Ltd |
MTT | 5 g/bottle | Amresco |
3. The test method comprises the following steps:
cells in logarithmic growth phase are inoculated in a 96-well plate (100 mu L/well) in a certain quantity, 100 mu L of culture solution containing different concentrations of test compounds is added into each well after adherent cells adhere for 24 hours, 100 mu L of culture solution containing different concentrations of test compounds is added on the day of suspension cell inoculation, 3 multiple wells are arranged for each drug concentration, and corresponding blank wells (only culture medium) and normal wells (drug concentration is 0) are arranged. After 72 hours of drug action, MTT working solution (5 mg/mL) was added at 20. Mu.L per well; 4 hours at 37 ℃, removing supernatant, adding DMSO (analytically pure) 150 mu L; the plate was wiped clean by shaking and mixing with a micropore oscillator, and the Optical Density (OD) value was measured at 550nm using an microplate reader.
The inhibition rate of cell growth was calculated using the following formula: inhibition (%) = (OD value) Normal hole OD value Medicine feeding hole ) /(OD value) Normal hole OD value Blank hole )×100%
From the inhibition rates of the respective concentrations, the half inhibitory concentration IC of the drug was calculated using SPSS19.0 50 。
The results of cell proliferation inhibition by the compounds are shown in table 4:
TABLE 4 inhibition of cell proliferation assay data (IC) for compounds of the present application 50 ,nM)
Cell name | Compound 45 |
DU145 | 85.5 |
MDA-MB-231 | 64.4 |
DU145: human prostate cancer cells; MDA-MB-231: human triple negative breast cancer cells.
And (4) test conclusion: according to the data in the table 4, the compound of the application has strong inhibition effect on human prostate cancer cell lines and breast cancer cell lines, and the in vitro cell inhibition activity is basically below 100 nM.
Test example 4: investigation of in vivo drug tumor inhibiting Activity-in vivo pharmacodynamics of the Compounds of the present application in human hepatoma Huh7 cell subcutaneous xenograft tumor model
Cell culture: high-glucose DMEM medium containing 10% Fetal Bovine Serum (FBS), 37 ℃ C., 5% CO 2 。
BALB/c Nude mice, female, weighing approximately 18-22 grams, each mouse inoculated with 0.1mL (5X 10) of subcutaneous tissue in the axilla of the forelimb 6 One) Huh7 cells. When the mean tumor volume reached 150 cubic millimeters, dosing was initiated, with the doses and pattern shown in the table below. Tumor volumes were measured 2 times per week, volumes measured in cubic millimeters, and dosing was terminated when the mean tumor volume in the solvent group grew to above 800 cubic millimeters to compare the difference in mean tumor volume between the test compound group and the solvent group. Tumor suppressive therapeutic effect of the compounds TGI (%) evaluation. TGI (%), reflecting the rate of tumor growth inhibition.
Solvent composition of solvent group and administration group: DMSO, DMSO: HP- β -CD (0.5 g/mL): the proportion of water is 2%:20%:78% (v/v/v)
Calculation of TGI (%): TGI (%) = [1- (average tumor volume at the end of administration of a certain treatment group-average tumor volume at the start of the treatment group)/(average tumor volume at the end of administration of a solvent control group-average tumor volume at the start of the solvent control group) ] × 100%.
The results are shown in tables 5 to 6.
TABLE 5 in vivo tumor inhibition test data
TABLE 6 in vivo tumor inhibition test data
And (4) test conclusion:
the compound shows good in-vivo drug effect and tolerance in a human liver cancer Huh7 cell subcutaneous xenograft tumor model, and has obvious tumor inhibition effect and better tolerance compared with BAY1251152 (2 animals die and have poor tolerance under the administration dosage of 5 mg/kg).
Test example 5: investigation of in vivo drug tumor inhibiting Activity-in vivo pharmacodynamics of the Compounds of the present application in the model of human hepatoma SMMC-7721 cells subcutaneous xenograft tumor
Cell culture: RPMI-1640 medium containing 10% Fetal Bovine Serum (FBS), 37 ℃ C., 5% CO 2 。
BALB/c Nude mice, female, 6-8 weeks, weighing about 18-22 g, inoculated with 0.1mL (1X 10) of subcutaneous tissue from axilla of forelimb per mouse 8 One) SMMC-7721 cells. When the mean tumor volume reached 150 mm, dosing was initiated, with the doses and modes of administration shown in the table below. Tumor volumes were measured 2 times per week, volumes were measured in cubic millimeters, and dosing was terminated when the mean tumor volume in the solvent group grew to over 800 cubic millimeters to compare the difference in mean tumor volume between the test compound group and the solvent group. Tumor suppressive therapeutic effect of the compounds TGI (%) evaluation. TGI (%), reflecting the rate of tumor growth inhibition.
Solvent composition of solvent group and administration group: DMSO, DMSO: HP- β -CD (0.5 g/mL): the water proportion is 2%:20%:78% (v/v/v)
Calculation of TGI (%): TGI (%) = [1- (average tumor volume at the end of administration of a certain treatment group-average tumor volume at the start of the treatment group)/(average tumor volume at the end of administration of a solvent control group-average tumor volume at the start of the solvent control group) ] × 100%.
The results are shown in Table 7.
TABLE 7 in vivo tumor inhibition test data
And (4) test conclusion:
the compound shows good in-vivo drug effect and tolerance in a human liver cancer SMMC-7721 cell subcutaneous xenograft tumor model, the tolerance of a mouse is obviously superior to that of a control drug, 4 animals in a BAY1251152 group die under the same dosage, the weight of the animals is obviously reduced, and the tolerance is poor.
Test example 6: study on tumor inhibiting activity of the drug in vivo-the in vivo efficacy of the compound in a human liver cancer HepG2-Luc cell subcutaneous xenograft tumor model.
BALB/c Nude mice, weighing about 17-20 grams.
Resuscitating in vitro passage cells to desired cell number, diluting cells with serum-free culture medium, counting under microscope, adjusting tumor cell number, adding matrigel, and mixing to obtain final cell number of about 5 × 10 7 Putting the cell suspension in an ice water bath; on the day of inoculation, the mouse is disinfected by wiping the axillary skin with alcohol, 0.1mL of the tumor cell suspension is extracted by a sterile syringe and is only inoculated under the axillary skin of the mouse, and the tumor cell suspension is about 5 multiplied by 10 6 And (4) respectively. Tumor volume up to 231mm 3 On the left and right, mice with good tumor growth are selected, animals are evenly divided into groups according to the tumor volume, and administration is started, wherein the administration dosage and the administration mode are shown in the table below. Tumor volumes were measured 2-3 times per week, volumes measured in cubic millimeters, and dosing was terminated when the mean tumor volume in the solvent group grew to over 800 cubic millimeters to compare the difference in mean tumor volume between the test compound group and the solvent group. Tumor suppressive therapeutic effect of the compounds TGI (%) evaluation. TGI (%), reflecting the rate of tumor growth inhibition.
Calculation of TGI (%): TGI (%) = [1- (average tumor volume at the end of administration of a certain treatment group-average tumor volume at the start of the treatment group)/(average tumor volume at the end of administration of a solvent control group-average tumor volume at the start of the solvent control group) ] × 100%.
The results are shown in Table 8.
TABLE 8 in vivo tumor inhibition test data
Group of | Number of animals | Mode of administration | Dosage to be administered | Days of administration | TGI(%) |
Solvent set | 8 | qd,p.o. | -- | 12 | -- |
Compound 45 | 8 | qd,p.o. | 5mg/kg | 12 | 45.6 |
Compound 45 | 8 | qd,p.o. | 7.5mg/kg | 12 | 76.7 |
Lenvatinib | 8 | qd,p.o. | 10mg/kg | 12 | 44.1 |
Solvent: an aqueous solution containing 2% DMSO and 20% HP- β -CD at a concentration of 0.5g/mL (DMSO: HP- β -CD (0.5 g/mL): water ratio 2%:20%:78%, v/v/v).
Lenvatinib preparation: weighing a proper amount of Lenvatinib (approved by FDA and used for first-line treatment of unresectable hepatocellular carcinoma patients), adding into a mixed solution of castor oil and absolute ethyl alcohol (the volume ratio is 1:1), performing vortex ultrasonic treatment to obtain a uniform solution, adding ultrapure water, and performing vortex ultrasonic treatment to obtain a uniform solution. (the volume ratio of the castor oil to the absolute ethyl alcohol to the ultrapure water is 1.
Administration volume: 10mL/kg
And (4) test conclusion:
the compound shows good in-vivo drug effect and tolerance in a human liver cancer HepG2-Luc cell subcutaneous xenograft tumor model; at an administration dose of 5mg/kg, the degree of tumor inhibition of Compound 45 was comparable to that of Lenvatinib (administration dose: 10 mg/kg); under the administration dosage of 7.5mg/kg, the tumor inhibition degree of the compound 45 is obviously superior to that of Lenvatinib; in the test process, the tolerance of the mice is good.
Test example 7: investigation of in vivo tumor inhibiting activity of drug-the in vivo efficacy of the compound in a human hepatoma Hep3B cell subcutaneous xenograft tumor model.
BALB/c Nude mice, weighing about 17-20 grams.
Resuscitating in vitro passage cells to desired cell number, diluting cells with serum-free culture medium, counting under microscope, adjusting tumor cell number, adding matrigel, and mixing to obtain final cell number of about 5 × 10 7 Putting the cell suspension in an ice water bath; on the day of inoculation, the mouse is disinfected by wiping the axillary skin with alcohol, 0.1mL of the tumor cell suspension is extracted by a sterile syringe and is only inoculated under the axillary skin of the mouse, and the tumor cell suspension is about 5 multiplied by 10 6 And (4) respectively. Tumor volume up to 230mm 3 On the left and right sides, selecting mice with good tumor growth, dividing the animals into groups according to the tumor volume, and starting to administer the drugs, wherein the dose and the administration mode are shown belowShown in the table. Tumor volumes were measured 2-3 times per week, volumes measured in cubic millimeters, and dosing was terminated when the mean tumor volume in the solvent group grew to over 800 cubic millimeters to compare the difference in mean tumor volume between the test compound group and the solvent group. Tumor suppressive therapeutic effect of the compounds TGI (%) evaluation. TGI (%), reflecting the rate of tumor growth inhibition.
Calculation of TGI (%): TGI (%) = [1- (average tumor volume at the end of administration of a certain treatment group-average tumor volume at the start of the treatment group)/(average tumor volume at the end of administration of a solvent control group-average tumor volume at the start of the solvent control group) ] × 100%.
The results are shown in Table 9.
TABLE 9 in vivo tumor inhibition test data
Group of | Number of animals | Mode of administration | Administration dosage | Days of administration | TGI(%) |
Solvent set | 8 | qd,p.o. | -- | 12 | -- |
Compound 45 | 8 | qd,p.o. | 5mg/kg | 12 | 43.9 |
Compound 45 | 8 | qd,p.o. | 7.5mg/kg | 12 | 64.6 |
Lenvatinib | 8 | qd,p.o. | 10mg/kg | 12 | 66.6 |
Solvent: an aqueous solution containing 2% DMSO and 20% HP- β -CD at a concentration of 0.5g/mL (DMSO: HP- β -CD (0.5 g/mL): water ratio 2%:20%:78%, v/v/v).
Lenvatinib preparation: weighing a proper amount of Lenvatinib, adding into a mixed solution of castor oil and absolute ethyl alcohol (the volume ratio is 1:1), performing vortex ultrasonic treatment to obtain a uniform solution, adding ultrapure water, and performing vortex ultrasonic treatment to obtain a uniform solution. (the volume ratio of the castor oil to the absolute ethyl alcohol to the ultrapure water is 1.
The administration volume: 10mL/kg
And (4) test conclusion:
the compound shows good in-vivo drug effect and tolerance in a human liver cancer Hep3B cell subcutaneous xenograft tumor model; the degree of tumor suppression of Compound 45 was comparable to that of Lenvatinib (administration dose: 10 mg/kg) at an administration dose of 7.5 mg/kg; in the test process, the tolerance of the mice is good.
Test example 8: in vitro hERG inhibitory Activity Studies
1. The purpose of the test is as follows:
rapidly activated human delayed rectifier outward potassium current (IKr) is mediated primarily by the hERG ion channel and is involved in human cardiomyocyte repolarization. Blocking this current by drugs is the leading cause of the clinical QT interval prolongation syndrome, even acute cardiac arrhythmias and even sudden death. Detecting the blocking effect of a compound on an hERG channel on a CHO-K1 cell line stably expressing the hERG channel by using a whole-cell patch clamp technology and determining the half inhibition concentration IC of the compound 50 . As part of a comprehensive cardiac safety assessment, they were initially evaluated in a safe in vitro screen for cardiotoxicity.
2. The test method comprises the following steps:
this test includes the following aspects:
recording the hERG current on a CHO-K1 cell strain stably expressing the hERG channel by using a manual patch clamp technology;
calculate inhibition for each concentration from hERG tail current;
test 5 concentrations per compound, calculate IC 50 A value;
3 cells tested per concentration;
one positive control drug.
Whole-cell patch clamp technique was used to record hERG current. The cell suspension was taken, placed in a cell well, and placed on an upright microscope stage. After the cells adhere to the wall, the cells are perfused by extracellular fluid with the flow rate of 1-2mL/min. The glass microelectrode is drawn by a microelectrode drawing instrument in two steps, and the water inlet resistance value of the glass microelectrode is 2-5M omega. After whole cell recording was established, the clamp potential was maintained at-80 mV. Depolarization to +60mV when given voltage stimulation, and then repolarization to-50 mV elicits hERG tail current. All recordings were made after the current had stabilized. The extracellular perfusion administration is started from low concentration, each concentration is 5-10min until the current is stable, and then the next concentration is given. Half maximal Inhibitory Concentration (IC) of test compound 50 ) Best fit from Logistic equation.
Amitripyrine is one of the most widely used tools for blocking hERG current and has been used as a positive control in this study.
3. The results are shown in Table 10:
TABLE 10 IC of hERG Current for compounds recorded on CHO-K1 stable cell lines 50 Numerical value
In the above assay, IC of hERG current inhibition by the positive control drug Amitriptyline 50 The result is consistent with the historical result of the testing party and is consistent with the result reported in the literature, which shows that the result of the test is credible. The results of the above experiments indicate that the compounds tested do not achieve half inhibition of hERG current at the highest concentration tested and therefore IC cannot be determined 50 The results show that the compound has no obvious inhibition effect on the hERG channel in the detection concentration range of the test, can reflect that the compound has low or no cardiotoxicity to a certain extent, and has positive significance on drug safety evaluation.
Test example 9: toxicity study of drug on mice
Animals for the test: ICR mice (5 weeks), male and female halves
Solvent: DMSO, DMSO: HP- β -CD (0.5 g/mL): the proportion of water is 2%:20%:78% (v/v/v)
The test method comprises the following steps: ICR mice were divided into 7 groups of 5 males and females per group according to weight balance. The administration mode is intragastric administration, once a day, and 7 days of continuous administration, and the administration dosage and the results are shown in the following table.
And (3) test results:
and (4) test conclusion: as the dose was escalated, the control drug BAY1251152 exhibited dose-dependent toxicity, increasing the number of animal deaths; no experimental animal death event occurred in compound 45 of the present application at the same dose as the control drug, and it can be seen that compound 45 of the present application is significantly better tolerated in female and male animals than the control drug BAY1251152.
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this application that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.
Claims (14)
1. An application of a compound shown as a formula (I) or a pharmaceutically acceptable salt, a stereoisomer or a prodrug thereof in preparing a medicament for treating solid tumors,
wherein,
x is selected from Cl and F; preferably F;
R 1 selected from substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; r 1 Said "substituted" in (a) means substituted with 1,2, 3,4 or 5 substituents each independently selected from-F, -Cl, -Br, -NH 2 、-OH、-SH、-CN、-NO 2 、-N 3 、-C≡CH、-COOH、-R 3 、-(CH 2 ) w O(CH 2 ) n R 3 、-(CH 2 ) w NH(CH 2 ) n R 3 、-(CH 2 ) w NR 3 (CH 2 ) n R 4 、-(CH 2 ) w S(CH 2 ) n R 3 、-(CH 2 ) w C(O)(CH 2 ) n R 3 、-(CH 2 ) w C(O)O(CH 2 ) n R 3 、-(CH 2 ) w OC(O)(CH 2 ) n R 3 、-(CH 2 ) w C(O)NH(CH 2 ) n R 3 、-(CH 2 ) w NHC(O)(CH 2 ) n R 3 、-(CH 2 ) w C(O)NR 3 (CH 2 ) n R 4 、-(CH 2 ) w NR 3 C(O)(CH 2 ) n R 4 、-(CH 2 ) w OS(O) 2 (CH 2 ) n R 3 Or- (CH) 2 ) w S(O) 2 O(CH 2 ) n R 3 Substituted with a group of (a); wherein each occurrence of w, n is independently selected from 0,1, 2,3 or 4;
R 3 and R 4 Each independently selected from substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, and substituted or unsubstituted C 1-6 Alkyl, substituted or unsubstituted C 1-6 Haloalkyl, substituted or unsubstituted C 2-6 Alkenyl, substituted or unsubstituted C 2-6 Alkynyl, substituted or unsubstituted C 1-6 Alkoxy, substituted or unsubstituted C 1-6 Haloalkoxy, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, or when R is 3 、R 4 When they are jointly bound to the same nitrogen atom, R 3 、R 4 And the nitrogen atom to which they are both attached form a substituted or unsubstituted heterocycloalkyl group; r 3 And R 4 Said "substituted" in (a) means substituted with 1,2 or 3 substituents each independently selected from-F, -Cl, -Br, -NH 2 、-OH、-SH、-CN、-NO 2 、-N 3 、-C≡CH、-COOH、C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Alkoxy radical, C 1-6 Halogenated alkoxy, etc.;
ring A is selected from substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl; said "substituted" in ring A is by 1,2, 3,4 or 5 groups each independently selected from-F, -Cl, -Br, OH, NH 2 、SH、CN、NO 2 、-N 3 、-C≡CH、COOH、R 5 、OR 5 、-NHR 5 、-NR 5 R 6 、-SR 5 、-NHCOR 5 、-CONHR 5 、-NHS(O) 2 R 5 、-S(O) 2 NHR 5 、-NR 5 S(O) 2 R 6 、-S(O) 2 NR 5 R 6 Or 1,2 or 1 in the A ring structuremultiple-CH 2 -the group can optionally be replaced by a-C (O) -group; wherein R is 5 And R 6 Independently is C 1-6 Alkyl radical, C 1-6 A haloalkyl group;
R 2 selected from H, R 7 、-(CH 2 ) x R 7 、-(CH 2 ) x NH(CH 2 ) y R 7 、-(CH 2 ) x O(CH 2 ) y R 7 、-(CH 2 ) x NR 7 (CH 2 ) y R 8 、-(CH 2 ) x C(O)(CH 2 ) y H、-(CH 2 ) x C(O)(CH 2 ) y R 7 、-(CH 2 ) x S(O) 2 (CH 2 ) y R 7 、-(CH 2 ) x C(O)C(O)(CH 2 ) y R 7 、-(CH 2 ) x S(O) 2 NH 2 、-(CH 2 ) x NHS(O) 2 H、-(CH 2 ) x S(O) 2 NH(CH 2 ) y R 7 、-(CH 2 ) x NHS(O) 2 (CH 2 ) y R 7 、-(CH 2 ) x S(O) 2 NR 7 (CH 2 ) y R 8 、-(CH 2 ) x NR 7 S(O) 2 (CH 2 ) y R 8 、-(CH 2 ) x C(O)O(CH 2 ) y R 7 、-(CH 2 ) x OC(O)(CH 2 ) y R 7 、-(CH 2 ) x C(O)NH 2 、-(CH 2 ) x NHC(O)H、-(CH 2 ) x C(O)NH(CH 2 ) y R 7 、-(CH 2 ) x NHC(O)(CH 2 ) y R 7 、-(CH 2 ) x C(O)NR 7 (CH 2 ) y R 8 Or- (CH) 2 ) x NR 7 C(O)(CH 2 ) y R 8 (ii) a Wherein, 1,2 or more-CH 2 -the group can optionally be replaced by a-C (O) -group; x and y are independently selected at each occurrence0.1, 2,3 or 4;
R 7 and R 8 Independently selected from substituted or unsubstituted R 9 、OR 9 、-R 10 -O-R 9 、-R 10 -NH-R 9 、-R 10 -C(O)-R 9 、-R 10 -NHC(O)-R 9 、-R 10 -C(O)NH-R 9 、-R 10 -S-R 9 、-R 10 -S(O)-R 9 、-R 10 -S-C(O)-R 9 Cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -R 10 -aryl, -R 10 -heteroaryl, -O-R 10 -aryl, -O-R 10 -heteroaryl, -R 10 -O-aryl, -R 10 -O-heteroaryl, -cycloalkyl-aryl, -cycloalkyl-heteroaryl, -heterocycloalkyl-aryl, -heterocycloalkyl-heteroaryl, C 2-6 Olefins and C 2-6 Alkyne, or when R 7 And R 8 When they are jointly bound to the same nitrogen atom, R 7 And R 8 With the nitrogen atom to which they are both attached to form a substituted or unsubstituted heterocycloalkyl; wherein R is 9 Is C 1-6 Alkyl radical, R 10 Is C 1-6 Alkylene radical, C 2-6 Alkenylene or C 2-6 An alkynylene group; r is 7 And R 8 Said "substituted" in (a) means substituted with 1,2 or 3 substituents each independently selected from-F, -Cl, -Br, -OH, -NH 2 、-SH、-CN、-NO 2 、-N 3 、-C≡CH、-COOH、C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, -NHCN, -NHCONH 2 、NHC(O)CH 3 、N(CH 3 ) 2 、N(C 2 H 5 ) 2 、-SC(O)CH 3 、-OC(O)-C 1-6 Alkyl, etc.;
said aryl group having no ring atom number defined above preferably contains 6 to 10 carbon atoms, cycloalkyl group preferably contains 3 to 6 carbon atoms, heteroaryl group preferably is 5 to 10-membered heteroaryl group, and heterocycloalkyl group preferably is 3 to 8-membered heterocyclic group; heteroaryl or heterocycloalkyl preferably contains 1,2 or 3 heteroatoms each independently selected from N, O or S, the remainder being carbon atoms.
2. A pharmaceutical composition for treating a solid tumor, comprising a compound represented by formula (I) as defined in claim 1, or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, and optionally a pharmaceutically acceptable carrier.
3. The use according to claim 1 or the pharmaceutical composition according to claim 2, wherein the solid tumor is a malignant solid tumor; preferably advanced malignant solid tumors; further preferably liver cancer, breast cancer or prostate cancer; further preferably, the liver, breast or prostate cancer is CDK 9-associated liver, breast or prostate cancer.
4. The use according to claim 3, wherein the liver cancer is hepatocellular carcinoma; preferably advanced hepatocellular carcinoma; further preferred is CDK 9-associated late-stage hepatocellular carcinoma; the breast cancer is triple negative breast cancer; preferably CDK 9-associated triple negative breast cancer.
5. The use or pharmaceutical composition according to any one of claims 1 to 4, wherein ring A is selected from substituted or unsubstituted 4-6 membered cycloalkyl, substituted or unsubstituted 4-6 membered heterocycloalkyl, substituted or unsubstituted 5-6 membered cycloalkyl, or substituted unsubstituted 5-6 membered heterocycloalkyl; preferably a substituted or unsubstituted 5-6 membered cycloalkyl group, or a substituted or unsubstituted 5-6 membered heterocycloalkyl group; further preferred is a substituted or unsubstituted 5-6 cycloalkyl group; or, the A ring is selected from cyclohexane, tetrahydropyrrole, piperidyl, piperazinyl, cyclopentyl or morpholinyl; preferably a cyclohexane, cyclopentane or tetrahydropyrrole group; the "substitution" means that the amino acid is substituted by 1,2, 3,4 or 5 groups each independently selected from-F, -Cl, -Br, OH, NH 2 、SH、CN、R 5 、OR 5 Wherein R is 5 Is C 1-6 Alkyl or C 1-6 An alkoxy group.
7. the use or pharmaceutical composition according to any one of claims 1 to 6, wherein R is 1 Selected from substituted or unsubstituted 6-10 membered aryl, or substituted or unsubstituted 5-10 membered heteroaryl; the heteroaryl group contains 1 or 2 heteroatoms each independently selected from N or O; the number of said substituents is selected from 1,2 or 3; further, R 1 Selected from substituted or unsubstituted benzene rings, pyridine rings, indole rings, indazole rings, benzofuran rings, pyrrolopyridine rings; preferably a substituted or unsubstituted benzene ring, pyridine ring, indole ring, benzofuran ring, pyrrolopyridine ring; further preferred are a substituted benzene ring, a pyridine ring and an unsubstituted indole ring, a benzofuran ring, a pyrrolopyridine ring; further preferred are substituted benzene rings; said R is 1 The substituents on the above are respectively and independently selected from-F, -Cl, -OH and-NH 2 、-R 3 、-(CH 2 ) w O(CH 2 ) n R 3 Or- (CH) 2 ) w OC(O)(CH 2 ) n R 3 (ii) a w and n are each independently selected from 0,1 or 2; preferably, said R is 1 The substituents on the substituent groups are respectively and independently selected from F-, -Cl, -OH and-R 3 、-(CH 2 ) w O(CH 2 ) n R 3 (ii) a Further preferably, said R 1 The substituents on the substituent group are respectively and independently selected from-F-, -OH and-R 3 、-(CH 2 ) w O(CH 2 ) n R 3 (ii) a When R is 1 In the case of a substituted phenyl ring, the substituents are selected from the group consisting of-F, -OH or alkoxy, preferably 1 or 2 fluorine atoms substituted and 1-OH or alkoxy substituted, preferably 1 or 2 fluorine atoms substituted and 1 alkoxy substituted.
8. The use or pharmaceutical composition according to any one of claims 1 to 7, wherein R is 3 And R 4 Each independently selected from substituted or unsubstituted 6-membered aryl, substituted or unsubstituted 5-6-membered heteroaryl, substituted or unsubstituted C 1-3 Alkyl, substituted or unsubstituted C 1-3 Alkoxy, substituted or unsubstituted C 3-6 Cycloalkyl, substituted or unsubstituted C 3-6 Heterocycloalkyl, or when R is 3 、R 4 When taken together to the same nitrogen atom, R 3 、R 4 And the nitrogen atom to which they are both attached form a 3-7 membered substituted or unsubstituted heterocycloalkyl group; the heterocycloalkyl contains 1 or 2 heteroatoms independently selected from N, O or S; r 3 And R 4 The term "substituted" as used herein means substituted with 1,2 or 3 substituents each independently selected from the group consisting of-F, -Cl, -Br, -OH, -CH 3 、-C 2 H 5 、-OCH 3 、-OC 2 H 5 Substituted with the substituent(s); preferably, R 3 And R 4 Each independently selected from substituted or unsubstituted C 1-3 Alkyl, substituted or unsubstituted C 1-3 An alkoxy group; further, R 3 And R 4 Each independently selected from the group consisting of substituted or unsubstituted benzene ring, pyridine ring, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, R 3 And R 4 The term "substituted" as used herein means substituted with 1,2 or 3 substituents each independently selected from the group consisting of-F, -Cl, -Br, -OH, -CH 3 、-C 2 H 5 、-OCH 3 、-OC 2 H 5 Substituted with the substituent(s); preferably, R 3 And R 4 Each independently selected from substituted or unsubstituted methyl, ethyl, isopropyl, methoxy, ethoxy, isopropoxy, cyclopropyl, pyridine ring; further preferably, R 3 And R 4 Are respectively independentIs selected from substituted or unsubstituted methyl, ethyl, isopropyl, methoxy, ethoxy, isopropoxy; even more preferably, R 3 And R 4 Each independently selected from substituted or unsubstituted methyl, methoxy.
9. The use or pharmaceutical composition according to any one of claims 1 to 8, wherein R is 2 Is selected from R 7 、-(CH 2 ) x R 7 、-(CH 2 ) x NH(CH 2 ) y R 7 、-(CH 2 ) x C(O)(CH 2 ) y R 7 、-(CH 2 ) x S(O) 2 (CH 2 ) y R 7 、-(CH 2 ) x C(O)C(O)(CH 2 ) y R 7 、-(CH 2 ) x C(O)O(CH 2 ) y R 7 、-(CH 2 ) x C(O)NH(CH 2 ) y R 7 、-(CH 2 ) x C(O)NR 7 (CH 2 ) y R 8 Or- (CH) 2 ) x NR 7 C(O)(CH 2 ) y R 8 (ii) a Further preferably, R 2 Is selected from R 7 、-(CH 2 ) x R 7 、-(CH 2 ) x C(O)(CH 2 ) y R 7 、-(CH 2 ) x S(O) 2 (CH 2 ) y R 7 、-(CH 2 ) x C(O)C(O)(CH 2 ) y R 7 、-(CH 2 ) x C(O)O(CH 2 ) y R 7 、-(CH 2 ) x C(O)NH(CH 2 ) y R 7 (ii) a Even more preferably, R 2 Is selected from R 7 、-(CH 2 ) x R 7 、-(CH 2 ) x C(O)(CH 2 ) y R 7 (ii) a Even more preferably, R 2 Is selected from- (CH) 2 ) x C(O)(CH 2 ) y R 7 。
10. According to claims 1 to E9, wherein R is 7 And R 8 Each independently selected from substituted or unsubstituted R 9 、OR 9 、-R 10 -O-R 9 、-R 10 -NH-R 9 、-R 10 -C(O)-R 9 、-R 10 -NHC(O)-R 9 、-R 10 -C(O)NH-R 9 、-R 10 -S-R 9 、-R 10 -S-C(O)-R 9 、C 3-6 Cycloalkyl, 3-6 membered heterocycloalkyl, C 6-10 Aryl, 5-10 membered heteroaryl, -R 10 -C 6-10 Aryl, -R 10 -5-10 membered heteroaryl, -O-R 10 -C 6-10 Aryl, -O-R 10 -5-10 membered heteroaryl, -R 10 -O-C 6-10 Aryl, -R 10 -O-5-10 membered heteroaryl, C 2-6 Olefins and C 2-6 Alkyne, or when R 7 And R 8 When they are jointly bound to the same nitrogen atom, R 7 And R 8 With the nitrogen atom to which they are both attached, to form a substituted or unsubstituted 3-6 membered heterocycloalkyl group; wherein R is 9 Is C 1-6 Alkyl radical, R 10 Is C 1-6 Alkylene radical, C 2-6 Alkenylene or C 2-6 An alkynylene group; r 7 And R 8 Said "substituted" in (a) means substituted with 1,2 or 3 substituents each independently selected from-F, -Cl, -Br, -OH, -NH 2 、-SH、-CN、C 1-3 Alkyl radical, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Haloalkoxy, -NHCN, -NHCONH 2 、NHC(O)CH 3 、N(CH 3 ) 2 、N(C 2 H 5 ) 2 、-SC(O)CH 3 、-OC(O)-C 1-6 Alkyl, etc.; preferably, R 7 Independently selected from substituted or unsubstituted R 9 、OR 9 、-R 10 -O-R 9 、-R 10 -NHC(O)-R 9 、C 3-6 Cycloalkyl, 3-6 membered heterocycloalkyl, C 6-10 Aryl, 5-10 membered heteroaryl, C 2-6 Olefins and C 2-6 Alkyne, or when R 7 And R 8 When they are jointly bound to the same nitrogen atom, R 7 And R 8 With groups of nitrogen atoms co-boundSubstituted or unsubstituted 3-6 membered heterocycloalkyl; further preferably, R 7 Independently selected from substituted or unsubstituted R 9 、OR 9 、-R 10 -O-R 9 、C 3-6 Cycloalkyl, 3-6 membered heterocycloalkyl, C 6-10 Aryl, 5-10 membered heteroaryl; further preferably, R 7 Independently selected from substituted or unsubstituted R 9 、OR 9 、-R 10 -O-R 9 、C 3-6 Cycloalkyl, 3-6 membered heterocycloalkyl; further preferably, R 7 Independently selected from substituted or unsubstituted R 9 ;R 7 The term "substituted" as used herein means substituted with 1,2 or 3 substituents each independently selected from the group consisting of-F, -Cl, -Br, -OH and-NH 2 、-SH、-CN、C 1-3 Alkyl radical, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Haloalkoxy, -NHCN, -NHCONH 2 、NHC(O)CH 3 、N(CH 3 ) 2 、N(C 2 H 5 ) 2 、-SC(O)CH 3 、-OC(O)-C 1-6 Alkyl, etc.; preferably, R 7 The term "substituted" as used herein means that 1,2 or 3 of them are each independently selected from the group consisting of-F, -OH, -NH 2 、-SH、-CN、C 1-3 Alkyl radical, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, NHC (O) CH 3 、N(CH 3 ) 2 、-OC(O)-C 1-6 Alkyl, etc.; further preferably, R 7 The term "substituted" as used herein means that 1,2 or 3 of them are each independently selected from the group consisting of-F, -OH, -NH 2 、-CN、C 1-3 Alkyl radical, C 1-3 Alkoxy, etc.; further preferably, R 7 The "substitution" as referred to in (1) means that 1,2 or 3 groups each independently selected from-OH, -CN, etc. are substituted; further preferably, R 7 The "substitution" as referred to in (1) means that 1 group each independently selected from-OH, -CN, etc. is substituted; further preferably, R 7 The term "substituted" as used herein means that 1 is substituted by an independently selected-OH group.
11. Use or medicament according to any one of claims 1 to 10Composition of matter, characterized in that R 7 And R 8 Each independently selected from substituted or unsubstituted methyl, ethyl, propyl, isopropyl, butyl, pentyl, methoxy, ethoxy, propoxy, isopropoxy, -CH 2 OCH 3 、-CH 2 OCH 2 CH 3 、-CH 2 CH 2 OCH 3 、-CH 2 CH 2 OCH 2 CH 3 Cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, aziridinyl, azetidinyl, azacyclohexyl, oxacyclopropyl, oxacyclobutyl, oxacyclopentyl, oxacyclohexyl, phenyl, pyridyl, pyrazolyl, oxazolyl, isoxazolyl, thienyl, thiazolyl, benzyl, phenethyl, ethenyl, propenyl, ethynyl or propynyl; preferably, R 7 Independently selected from substituted or unsubstituted methyl, ethyl, methoxy, ethoxy, cyclopropyl, cyclobutyl, azetidinyl, azacyclohexyl, oxetanyl, oxacyclohexyl, phenyl, pyridyl, pyrazolyl, isoxazolyl, thienyl, thiazolyl, benzyl, ethenyl, propenyl or ethynyl; further preferably, R 7 Independently selected from substituted or unsubstituted methyl, ethyl, methoxy, cyclopropyl, cyclobutyl, azacyclohexyl, oxetanyl, oxacyclohexyl, pyridyl, pyrazolyl, isoxazolyl, ethenyl, propenyl or ethynyl; even more preferably, R 7 Independently selected from substituted or unsubstituted methyl, ethyl, cyclopropyl, azacyclohexyl, oxetanyl, pyrazolyl, ethenyl, propenyl or ethynyl; even more preferably, R 7 Independently selected from substituted or unsubstituted methyl, ethyl, cyclopropyl; even more preferably, R 7 Independently selected from substituted or unsubstituted methyl; r 7 And R 8 Said "substituted" in (1) means that the substituted is substituted with 1,2 or 3 substituents each independently selected from-F, -Cl, -Br, -OH, -NH 2 、-SH、-CN、C 1-3 Alkyl radical, C 1-3 Alkoxy radical, C 1-3 Haloalkyl, C 1-3 Haloalkoxy, -NHCN, -NHCONH 2 、NHC(O)CH 3 、N(CH 3 ) 2 、N(C 2 H 5 ) 2 、-SC(O)CH 3 、-OC(O)-C 1-6 Alkyl, etc.
13. the use or pharmaceutical composition according to any one of claims 1 to 12, wherein the compound of formula (I) or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof is the only active ingredient in the medicament; and/or the compound shown in the formula (I) or the pharmaceutically acceptable salt, the stereoisomer or the prodrug thereof is combined with one or more other targeting drugs or chemotherapeutic drugs for preparing the drug.
14. The use or pharmaceutical composition according to any one of claims 1 to 13, wherein the medicament is formulated for clinical acceptance; preferably, the formulation is an oral formulation or an injectable formulation.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN2021105640930 | 2021-05-24 | ||
CN202110564093 | 2021-05-24 |
Publications (2)
Publication Number | Publication Date |
---|---|
CN115381823A true CN115381823A (en) | 2022-11-25 |
CN115381823B CN115381823B (en) | 2024-11-05 |
Family
ID=
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN102471310A (en) * | 2009-09-04 | 2012-05-23 | 诺瓦提斯公司 | Bipyridines useful for the treatment of proliferative diseases |
CN102482265A (en) * | 2009-09-04 | 2012-05-30 | 诺瓦提斯公司 | Pyrazinylpyridines useful for the treatment of proliferative diseases |
WO2014063068A1 (en) * | 2012-10-18 | 2014-04-24 | Dana-Farber Cancer Institute, Inc. | Inhibitors of cyclin-dependent kinase 7 (cdk7) |
CN114787142A (en) * | 2019-12-09 | 2022-07-22 | 石药集团中奇制药技术(石家庄)有限公司 | Compounds as cyclin-dependent kinase 9 inhibitors and uses thereof |
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN102471310A (en) * | 2009-09-04 | 2012-05-23 | 诺瓦提斯公司 | Bipyridines useful for the treatment of proliferative diseases |
CN102482265A (en) * | 2009-09-04 | 2012-05-30 | 诺瓦提斯公司 | Pyrazinylpyridines useful for the treatment of proliferative diseases |
WO2014063068A1 (en) * | 2012-10-18 | 2014-04-24 | Dana-Farber Cancer Institute, Inc. | Inhibitors of cyclin-dependent kinase 7 (cdk7) |
CN114787142A (en) * | 2019-12-09 | 2022-07-22 | 石药集团中奇制药技术(石家庄)有限公司 | Compounds as cyclin-dependent kinase 9 inhibitors and uses thereof |
Also Published As
Publication number | Publication date |
---|---|
WO2022247785A1 (en) | 2022-12-01 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN111484477B (en) | Benzopyridone heterocyclic compound and application thereof | |
CN114787142A (en) | Compounds as cyclin-dependent kinase 9 inhibitors and uses thereof | |
CA2988816A1 (en) | Ezh2 inhibitors for treating lymphoma | |
CN112745335A (en) | Tri-heterocyclic compound and application thereof | |
EP3471830A1 (en) | Ezh2 inhibitors for treating cancer | |
WO2022253101A1 (en) | Pyridazinone compound as parp7 inhibitor | |
CN112851663A (en) | Fused heterocyclic compound and application thereof | |
CN112159401A (en) | Biased agonist and medical application thereof | |
CN113045569B (en) | Compounds useful as RET kinase inhibitors and uses thereof | |
CN111718332B (en) | 2-substituted pyrazol amino-4-substituted amino-5-pyrimidine formamide compound, composition and application thereof | |
CN115381823A (en) | Application of cyclin-dependent kinase 9 inhibitor | |
TWI782523B (en) | Compounds as RET kinase inhibitors and their applications | |
CN115381823B (en) | Application of cyclin-dependent kinase 9 inhibitor | |
WO2021228213A1 (en) | Combination comprising tricycle compound and use thereof in preparation of medicament for treating hbv | |
CN115381824A (en) | Use of cyclin-dependent kinase 9 inhibitors | |
CN115433207A (en) | Macrocyclic heterocyclic compound as EGFR inhibitor and application thereof | |
CN115381824B (en) | Use of cyclin-dependent kinase 9 inhibitors | |
CN115385912A (en) | Pyrazinopyrazinoquinolinone derivatives, preparation method and medical application thereof | |
RU2819762C1 (en) | Compound as cyclin-dependent kinase 9 inhibitor and use thereof | |
CN114276328B (en) | Compound as small molecule immunosuppressant, preparation method and application thereof | |
WO2024222630A1 (en) | Quinolone derivative and use thereof against tumors | |
CN104211687B (en) | The indole of heterocyclic substituted naphthalene ketone derivant, its preparation method, medical composition and its use | |
WO2024120360A1 (en) | Parp7 inhibitor, preparation method therefor and use thereof | |
CN113968861A (en) | Compound with PI3K delta/BTK double-target-point activity and preparation method and application thereof | |
CN116157401A (en) | Heterocyclic derivative and preparation method and application thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination | ||
GR01 | Patent grant |