WO2024199524A1 - 作为pkmyt1抑制剂的化合物 - Google Patents
作为pkmyt1抑制剂的化合物 Download PDFInfo
- Publication number
- WO2024199524A1 WO2024199524A1 PCT/CN2024/085203 CN2024085203W WO2024199524A1 WO 2024199524 A1 WO2024199524 A1 WO 2024199524A1 CN 2024085203 W CN2024085203 W CN 2024085203W WO 2024199524 A1 WO2024199524 A1 WO 2024199524A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- alkylene
- alkyl
- cycloalkyl
- heterocyclyl
- independently selected
- Prior art date
Links
- 150000001875 compounds Chemical class 0.000 title claims abstract description 333
- 229940126642 PKMYT1 inhibitor Drugs 0.000 title abstract description 3
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 41
- 150000003839 salts Chemical class 0.000 claims abstract description 28
- 201000010099 disease Diseases 0.000 claims abstract description 26
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 16
- 238000002360 preparation method Methods 0.000 claims abstract description 7
- -1 oxopiperazinyl Chemical group 0.000 claims description 198
- 125000000217 alkyl group Chemical group 0.000 claims description 142
- 125000000623 heterocyclic group Chemical group 0.000 claims description 135
- 125000001424 substituent group Chemical group 0.000 claims description 126
- 125000001072 heteroaryl group Chemical group 0.000 claims description 118
- 229910052739 hydrogen Inorganic materials 0.000 claims description 110
- 239000001257 hydrogen Substances 0.000 claims description 108
- 125000000304 alkynyl group Chemical group 0.000 claims description 89
- 125000000008 (C1-C10) alkyl group Chemical group 0.000 claims description 87
- 125000003118 aryl group Chemical group 0.000 claims description 84
- 229910052736 halogen Inorganic materials 0.000 claims description 75
- 150000002367 halogens Chemical group 0.000 claims description 75
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 claims description 72
- 125000000753 cycloalkyl group Chemical group 0.000 claims description 63
- 125000006376 (C3-C10) cycloalkyl group Chemical group 0.000 claims description 62
- 125000002947 alkylene group Chemical group 0.000 claims description 55
- 229910052757 nitrogen Inorganic materials 0.000 claims description 53
- 150000002431 hydrogen Chemical group 0.000 claims description 52
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 claims description 49
- 125000005842 heteroatom Chemical group 0.000 claims description 47
- 229910052717 sulfur Inorganic materials 0.000 claims description 47
- 125000003342 alkenyl group Chemical group 0.000 claims description 46
- 125000006374 C2-C10 alkenyl group Chemical group 0.000 claims description 41
- 229910052760 oxygen Inorganic materials 0.000 claims description 41
- 125000004429 atom Chemical group 0.000 claims description 40
- 125000005275 alkylenearyl group Chemical group 0.000 claims description 39
- 125000005218 alkyleneheteroaryl group Chemical group 0.000 claims description 39
- 206010028980 Neoplasm Diseases 0.000 claims description 33
- 229910052799 carbon Inorganic materials 0.000 claims description 31
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 28
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 claims description 27
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 claims description 26
- 239000001301 oxygen Substances 0.000 claims description 26
- 239000011593 sulfur Substances 0.000 claims description 26
- YLQBMQCUIZJEEH-UHFFFAOYSA-N Furan Chemical compound C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 claims description 24
- LPIQUOYDBNQMRZ-UHFFFAOYSA-N cyclopentene Chemical group C1CC=CC1 LPIQUOYDBNQMRZ-UHFFFAOYSA-N 0.000 claims description 22
- 239000003814 drug Substances 0.000 claims description 22
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 claims description 22
- 125000004432 carbon atom Chemical group C* 0.000 claims description 20
- RGSFGYAAUTVSQA-UHFFFAOYSA-N pentamethylene Chemical group C1CCCC1 RGSFGYAAUTVSQA-UHFFFAOYSA-N 0.000 claims description 19
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical group C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 claims description 18
- 201000011510 cancer Diseases 0.000 claims description 18
- RAXXELZNTBOGNW-UHFFFAOYSA-N imidazole Natural products C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 claims description 18
- 239000000460 chlorine Substances 0.000 claims description 16
- HGCIXCUEYOPUTN-UHFFFAOYSA-N cis-cyclohexene Natural products C1CCC=CC1 HGCIXCUEYOPUTN-UHFFFAOYSA-N 0.000 claims description 16
- 208000035475 disorder Diseases 0.000 claims description 15
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 claims description 14
- YTPLMLYBLZKORZ-UHFFFAOYSA-N Thiophene Chemical compound C=1C=CSC=1 YTPLMLYBLZKORZ-UHFFFAOYSA-N 0.000 claims description 14
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 14
- 229910052698 phosphorus Inorganic materials 0.000 claims description 14
- 239000011574 phosphorus Substances 0.000 claims description 14
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 claims description 12
- 125000002950 monocyclic group Chemical group 0.000 claims description 12
- 206010033128 Ovarian cancer Diseases 0.000 claims description 11
- 206010061535 Ovarian neoplasm Diseases 0.000 claims description 11
- 125000006569 (C5-C6) heterocyclic group Chemical group 0.000 claims description 10
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 claims description 10
- 150000002430 hydrocarbons Chemical group 0.000 claims description 10
- 206010006187 Breast cancer Diseases 0.000 claims description 9
- 208000026310 Breast neoplasm Diseases 0.000 claims description 9
- 229910052801 chlorine Inorganic materials 0.000 claims description 9
- 125000000596 cyclohexenyl group Chemical group C1(=CCCCC1)* 0.000 claims description 9
- 229910052731 fluorine Inorganic materials 0.000 claims description 9
- UMJSCPRVCHMLSP-UHFFFAOYSA-N pyridine Chemical group COC1=CC=CN=C1 UMJSCPRVCHMLSP-UHFFFAOYSA-N 0.000 claims description 9
- LVZWSLJZHVFIQJ-UHFFFAOYSA-N Cyclopropane Chemical compound C1CC1 LVZWSLJZHVFIQJ-UHFFFAOYSA-N 0.000 claims description 8
- 208000000461 Esophageal Neoplasms Diseases 0.000 claims description 8
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 8
- 206010030155 Oesophageal carcinoma Diseases 0.000 claims description 8
- KAESVJOAVNADME-UHFFFAOYSA-N Pyrrole Chemical compound C=1C=CNC=1 KAESVJOAVNADME-UHFFFAOYSA-N 0.000 claims description 8
- 208000005718 Stomach Neoplasms Diseases 0.000 claims description 8
- FZWLAAWBMGSTSO-UHFFFAOYSA-N Thiazole Chemical compound C1=CSC=N1 FZWLAAWBMGSTSO-UHFFFAOYSA-N 0.000 claims description 8
- 201000004101 esophageal cancer Diseases 0.000 claims description 8
- 206010017758 gastric cancer Diseases 0.000 claims description 8
- DMEGYFMYUHOHGS-UHFFFAOYSA-N heptamethylene Chemical group C1CCCCCC1 DMEGYFMYUHOHGS-UHFFFAOYSA-N 0.000 claims description 8
- 201000005202 lung cancer Diseases 0.000 claims description 8
- 208000020816 lung neoplasm Diseases 0.000 claims description 8
- 125000003226 pyrazolyl group Chemical group 0.000 claims description 8
- 201000011549 stomach cancer Diseases 0.000 claims description 8
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 claims description 8
- 206010014733 Endometrial cancer Diseases 0.000 claims description 7
- 206010014759 Endometrial neoplasm Diseases 0.000 claims description 7
- HSFWRNGVRCDJHI-UHFFFAOYSA-N alpha-acetylene Natural products C#C HSFWRNGVRCDJHI-UHFFFAOYSA-N 0.000 claims description 7
- 229910052794 bromium Inorganic materials 0.000 claims description 7
- 125000002534 ethynyl group Chemical group [H]C#C* 0.000 claims description 7
- 201000007270 liver cancer Diseases 0.000 claims description 7
- 208000014018 liver neoplasm Diseases 0.000 claims description 7
- 229930192474 thiophene Natural products 0.000 claims description 7
- XDTMQSROBMDMFD-UHFFFAOYSA-N Cyclohexane Chemical group C1CCCCC1 XDTMQSROBMDMFD-UHFFFAOYSA-N 0.000 claims description 6
- NQRYJNQNLNOLGT-UHFFFAOYSA-N Piperidine Chemical group C1CCNCC1 NQRYJNQNLNOLGT-UHFFFAOYSA-N 0.000 claims description 6
- 125000002777 acetyl group Chemical group [H]C([H])([H])C(*)=O 0.000 claims description 6
- 125000004093 cyano group Chemical group *C#N 0.000 claims description 6
- 206010005003 Bladder cancer Diseases 0.000 claims description 5
- 206010008342 Cervix carcinoma Diseases 0.000 claims description 5
- 206010009944 Colon cancer Diseases 0.000 claims description 5
- 206010018338 Glioma Diseases 0.000 claims description 5
- JCXJVPUVTGWSNB-UHFFFAOYSA-N Nitrogen dioxide Chemical compound O=[N]=O JCXJVPUVTGWSNB-UHFFFAOYSA-N 0.000 claims description 5
- WTKZEGDFNFYCGP-UHFFFAOYSA-N Pyrazole Chemical compound C=1C=NNC=1 WTKZEGDFNFYCGP-UHFFFAOYSA-N 0.000 claims description 5
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 claims description 5
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 claims description 5
- 125000002393 azetidinyl group Chemical group 0.000 claims description 5
- 208000035269 cancer or benign tumor Diseases 0.000 claims description 5
- 201000010881 cervical cancer Diseases 0.000 claims description 5
- 239000003937 drug carrier Substances 0.000 claims description 5
- 201000010536 head and neck cancer Diseases 0.000 claims description 5
- 208000014829 head and neck neoplasm Diseases 0.000 claims description 5
- 201000001441 melanoma Diseases 0.000 claims description 5
- 125000002757 morpholinyl group Chemical group 0.000 claims description 5
- 125000003386 piperidinyl group Chemical group 0.000 claims description 5
- 125000004076 pyridyl group Chemical group 0.000 claims description 5
- 125000000335 thiazolyl group Chemical group 0.000 claims description 5
- 201000005112 urinary bladder cancer Diseases 0.000 claims description 5
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 claims description 4
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 claims description 4
- 101001132551 Diplobatis ommata Ras-related protein Rab-8 Proteins 0.000 claims description 4
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 claims description 4
- 125000003917 carbamoyl group Chemical group [H]N([H])C(*)=O 0.000 claims description 4
- JHIVVAPYMSGYDF-UHFFFAOYSA-N cyclohexanone Chemical group O=C1CCCCC1 JHIVVAPYMSGYDF-UHFFFAOYSA-N 0.000 claims description 4
- 238000006467 substitution reaction Methods 0.000 claims description 4
- YNGDWRXWKFWCJY-UHFFFAOYSA-N 1,4-Dihydropyridine Chemical group C1C=CNC=C1 YNGDWRXWKFWCJY-UHFFFAOYSA-N 0.000 claims description 3
- 208000001333 Colorectal Neoplasms Diseases 0.000 claims description 3
- BUDQDWGNQVEFAC-UHFFFAOYSA-N Dihydropyran Chemical group C1COC=CC1 BUDQDWGNQVEFAC-UHFFFAOYSA-N 0.000 claims description 3
- 101100406515 Drosophila melanogaster orb2 gene Proteins 0.000 claims description 3
- 208000032612 Glial tumor Diseases 0.000 claims description 3
- 101100258025 Schizosaccharomyces pombe (strain 972 / ATCC 24843) shk1 gene Proteins 0.000 claims description 3
- 208000002495 Uterine Neoplasms Diseases 0.000 claims description 3
- 208000009956 adenocarcinoma Diseases 0.000 claims description 3
- 208000024200 hematopoietic and lymphoid system neoplasm Diseases 0.000 claims description 3
- 206010073071 hepatocellular carcinoma Diseases 0.000 claims description 3
- 231100000844 hepatocellular carcinoma Toxicity 0.000 claims description 3
- 206010046766 uterine cancer Diseases 0.000 claims description 3
- 208000037965 uterine sarcoma Diseases 0.000 claims description 3
- OXBLVCZKDOZZOJ-UHFFFAOYSA-N 2,3-Dihydrothiophene Chemical group C1CC=CS1 OXBLVCZKDOZZOJ-UHFFFAOYSA-N 0.000 claims description 2
- VSWICNJIUPRZIK-UHFFFAOYSA-N 2-piperideine Chemical group C1CNC=CC1 VSWICNJIUPRZIK-UHFFFAOYSA-N 0.000 claims description 2
- ZPOODPZCZLCUAN-UHFFFAOYSA-N 3,4-dihydro-1h-pyridin-2-one Chemical group O=C1CCC=CN1 ZPOODPZCZLCUAN-UHFFFAOYSA-N 0.000 claims description 2
- 208000036762 Acute promyelocytic leukaemia Diseases 0.000 claims description 2
- 206010003571 Astrocytoma Diseases 0.000 claims description 2
- 208000003950 B-cell lymphoma Diseases 0.000 claims description 2
- 208000003174 Brain Neoplasms Diseases 0.000 claims description 2
- 101000743846 Diplobatis ommata Ras-related protein Rab-10 Proteins 0.000 claims description 2
- 201000008808 Fibrosarcoma Diseases 0.000 claims description 2
- 208000004463 Follicular Adenocarcinoma Diseases 0.000 claims description 2
- 208000022072 Gallbladder Neoplasms Diseases 0.000 claims description 2
- 206010061187 Haematopoietic neoplasm Diseases 0.000 claims description 2
- 208000002250 Hematologic Neoplasms Diseases 0.000 claims description 2
- 208000017604 Hodgkin disease Diseases 0.000 claims description 2
- 208000021519 Hodgkin lymphoma Diseases 0.000 claims description 2
- 208000010747 Hodgkins lymphoma Diseases 0.000 claims description 2
- 208000007766 Kaposi sarcoma Diseases 0.000 claims description 2
- 206010023347 Keratoacanthoma Diseases 0.000 claims description 2
- 208000008839 Kidney Neoplasms Diseases 0.000 claims description 2
- 201000003793 Myelodysplastic syndrome Diseases 0.000 claims description 2
- 208000033761 Myelogenous Chronic BCR-ABL Positive Leukemia Diseases 0.000 claims description 2
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 claims description 2
- 206010029260 Neuroblastoma Diseases 0.000 claims description 2
- 206010061902 Pancreatic neoplasm Diseases 0.000 claims description 2
- 206010060862 Prostate cancer Diseases 0.000 claims description 2
- 208000000236 Prostatic Neoplasms Diseases 0.000 claims description 2
- 206010038389 Renal cancer Diseases 0.000 claims description 2
- 201000010208 Seminoma Diseases 0.000 claims description 2
- 208000000453 Skin Neoplasms Diseases 0.000 claims description 2
- 206010041067 Small cell lung cancer Diseases 0.000 claims description 2
- 206010042971 T-cell lymphoma Diseases 0.000 claims description 2
- 208000027585 T-cell non-Hodgkin lymphoma Diseases 0.000 claims description 2
- 206010043276 Teratoma Diseases 0.000 claims description 2
- DHXVGJBLRPWPCS-UHFFFAOYSA-N Tetrahydropyran Chemical group C1CCOCC1 DHXVGJBLRPWPCS-UHFFFAOYSA-N 0.000 claims description 2
- 208000024770 Thyroid neoplasm Diseases 0.000 claims description 2
- 201000006083 Xeroderma Pigmentosum Diseases 0.000 claims description 2
- 230000001154 acute effect Effects 0.000 claims description 2
- 210000003169 central nervous system Anatomy 0.000 claims description 2
- 208000029742 colonic neoplasm Diseases 0.000 claims description 2
- FWFSEYBSWVRWGL-UHFFFAOYSA-N cyclohex-2-enone Chemical group O=C1CCCC=C1 FWFSEYBSWVRWGL-UHFFFAOYSA-N 0.000 claims description 2
- 201000010175 gallbladder cancer Diseases 0.000 claims description 2
- 201000005787 hematologic cancer Diseases 0.000 claims description 2
- 208000019691 hematopoietic and lymphoid cell neoplasm Diseases 0.000 claims description 2
- 201000010982 kidney cancer Diseases 0.000 claims description 2
- 208000032839 leukemia Diseases 0.000 claims description 2
- 201000005243 lung squamous cell carcinoma Diseases 0.000 claims description 2
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 claims description 2
- 208000007538 neurilemmoma Diseases 0.000 claims description 2
- 201000008968 osteosarcoma Diseases 0.000 claims description 2
- 201000002528 pancreatic cancer Diseases 0.000 claims description 2
- 208000008443 pancreatic carcinoma Diseases 0.000 claims description 2
- 210000001428 peripheral nervous system Anatomy 0.000 claims description 2
- XUWHAWMETYGRKB-UHFFFAOYSA-N piperidin-2-one Chemical group O=C1CCCCN1 XUWHAWMETYGRKB-UHFFFAOYSA-N 0.000 claims description 2
- 201000009410 rhabdomyosarcoma Diseases 0.000 claims description 2
- 206010039667 schwannoma Diseases 0.000 claims description 2
- 201000000849 skin cancer Diseases 0.000 claims description 2
- 208000000587 small cell lung carcinoma Diseases 0.000 claims description 2
- RAOIDOHSFRTOEL-UHFFFAOYSA-N tetrahydrothiophene Chemical group C1CCSC1 RAOIDOHSFRTOEL-UHFFFAOYSA-N 0.000 claims description 2
- 201000002510 thyroid cancer Diseases 0.000 claims description 2
- 208000030901 thyroid gland follicular carcinoma Diseases 0.000 claims description 2
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 claims 1
- 210000004369 blood Anatomy 0.000 claims 1
- 239000008280 blood Substances 0.000 claims 1
- 201000010288 cervix melanoma Diseases 0.000 claims 1
- 101000687968 Homo sapiens Membrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinase Proteins 0.000 abstract description 22
- 102100024262 Membrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinase Human genes 0.000 abstract description 22
- 238000011282 treatment Methods 0.000 abstract description 5
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 453
- 239000000543 intermediate Substances 0.000 description 388
- 238000006243 chemical reaction Methods 0.000 description 246
- 239000000243 solution Substances 0.000 description 206
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 204
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 183
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 175
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 169
- 230000015572 biosynthetic process Effects 0.000 description 162
- 238000003786 synthesis reaction Methods 0.000 description 162
- 230000002829 reductive effect Effects 0.000 description 144
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 124
- 239000000203 mixture Substances 0.000 description 120
- XKRFYHLGVUSROY-UHFFFAOYSA-N Argon Chemical compound [Ar] XKRFYHLGVUSROY-UHFFFAOYSA-N 0.000 description 116
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 105
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 96
- 239000012074 organic phase Substances 0.000 description 95
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical class O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 90
- 238000005481 NMR spectroscopy Methods 0.000 description 74
- 239000000706 filtrate Substances 0.000 description 67
- 229910052786 argon Inorganic materials 0.000 description 58
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 57
- 239000003208 petroleum Substances 0.000 description 55
- XTHFKEDIFFGKHM-UHFFFAOYSA-N Dimethoxyethane Chemical compound COCCOC XTHFKEDIFFGKHM-UHFFFAOYSA-N 0.000 description 50
- 239000012071 phase Substances 0.000 description 50
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 46
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-Dimethylformamide Chemical compound CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 42
- 238000001816 cooling Methods 0.000 description 35
- 239000003112 inhibitor Substances 0.000 description 35
- QGZKDVFQNNGYKY-UHFFFAOYSA-N Ammonia Chemical compound N QGZKDVFQNNGYKY-UHFFFAOYSA-N 0.000 description 32
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 32
- 238000010898 silica gel chromatography Methods 0.000 description 31
- 239000012065 filter cake Substances 0.000 description 28
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 27
- 238000010791 quenching Methods 0.000 description 26
- ILAHWRKJUDSMFH-UHFFFAOYSA-N boron tribromide Chemical compound BrB(Br)Br ILAHWRKJUDSMFH-UHFFFAOYSA-N 0.000 description 24
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical class [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 23
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 22
- BWHMMNNQKKPAPP-UHFFFAOYSA-L potassium carbonate Chemical compound [K+].[K+].[O-]C([O-])=O BWHMMNNQKKPAPP-UHFFFAOYSA-L 0.000 description 22
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 21
- 210000004027 cell Anatomy 0.000 description 21
- 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 21
- 239000000047 product Substances 0.000 description 21
- 238000004809 thin layer chromatography Methods 0.000 description 21
- 102100037858 G1/S-specific cyclin-E1 Human genes 0.000 description 20
- 238000010790 dilution Methods 0.000 description 20
- 239000012895 dilution Substances 0.000 description 20
- 101000738568 Homo sapiens G1/S-specific cyclin-E1 Proteins 0.000 description 19
- 238000004440 column chromatography Methods 0.000 description 19
- 238000000034 method Methods 0.000 description 19
- 238000003756 stirring Methods 0.000 description 19
- QAOWNCQODCNURD-UHFFFAOYSA-N sulfuric acid Substances OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 19
- CUONGYYJJVDODC-UHFFFAOYSA-N malononitrile Chemical compound N#CCC#N CUONGYYJJVDODC-UHFFFAOYSA-N 0.000 description 18
- 239000007787 solid Substances 0.000 description 18
- 125000001183 hydrocarbyl group Chemical group 0.000 description 17
- 238000004808 supercritical fluid chromatography Methods 0.000 description 17
- PCLIMKBDDGJMGD-UHFFFAOYSA-N N-bromosuccinimide Chemical compound BrN1C(=O)CCC1=O PCLIMKBDDGJMGD-UHFFFAOYSA-N 0.000 description 16
- KEAYESYHFKHZAL-UHFFFAOYSA-N Sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 16
- 229910021529 ammonia Inorganic materials 0.000 description 16
- 230000014509 gene expression Effects 0.000 description 16
- BDAGIHXWWSANSR-UHFFFAOYSA-N methanoic acid Natural products OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 16
- 239000012312 sodium hydride Substances 0.000 description 16
- 229910000104 sodium hydride Inorganic materials 0.000 description 16
- 238000000926 separation method Methods 0.000 description 15
- MZRVEZGGRBJDDB-UHFFFAOYSA-N N-Butyllithium Chemical compound [Li]CCCC MZRVEZGGRBJDDB-UHFFFAOYSA-N 0.000 description 14
- UZFKIHMFDWGBKC-UHFFFAOYSA-N pyridine-4-carboxamide Chemical compound NC(=O)C1=CC=NC=C1.NC(=O)C1=CC=NC=C1 UZFKIHMFDWGBKC-UHFFFAOYSA-N 0.000 description 14
- DBRPRHMYNICNTR-UHFFFAOYSA-N 3-methoxy-2,6-dimethylaniline Chemical compound COC1=CC=C(C)C(N)=C1C DBRPRHMYNICNTR-UHFFFAOYSA-N 0.000 description 13
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 13
- 238000004237 preparative chromatography Methods 0.000 description 13
- 229940124597 therapeutic agent Drugs 0.000 description 13
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 12
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 12
- 230000000694 effects Effects 0.000 description 12
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical class [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 11
- 239000007788 liquid Substances 0.000 description 11
- 229910000027 potassium carbonate Inorganic materials 0.000 description 11
- 239000007789 gas Substances 0.000 description 10
- FYSNRJHAOHDILO-UHFFFAOYSA-N thionyl chloride Chemical compound ClS(Cl)=O FYSNRJHAOHDILO-UHFFFAOYSA-N 0.000 description 10
- 239000000741 silica gel Substances 0.000 description 9
- 229910002027 silica gel Inorganic materials 0.000 description 9
- PAQZWJGSJMLPMG-UHFFFAOYSA-N 2,4,6-tripropyl-1,3,5,2$l^{5},4$l^{5},6$l^{5}-trioxatriphosphinane 2,4,6-trioxide Chemical compound CCCP1(=O)OP(=O)(CCC)OP(=O)(CCC)O1 PAQZWJGSJMLPMG-UHFFFAOYSA-N 0.000 description 8
- OSWFIVFLDKOXQC-UHFFFAOYSA-N 4-(3-methoxyphenyl)aniline Chemical compound COC1=CC=CC(C=2C=CC(N)=CC=2)=C1 OSWFIVFLDKOXQC-UHFFFAOYSA-N 0.000 description 8
- 239000007864 aqueous solution Substances 0.000 description 8
- 235000019253 formic acid Nutrition 0.000 description 8
- 125000001841 imino group Chemical group [H]N=* 0.000 description 8
- 108090000623 proteins and genes Proteins 0.000 description 8
- MFRIHAYPQRLWNB-UHFFFAOYSA-N sodium tert-butoxide Chemical compound [Na+].CC(C)(C)[O-] MFRIHAYPQRLWNB-UHFFFAOYSA-N 0.000 description 8
- 239000002904 solvent Substances 0.000 description 8
- 239000004215 Carbon black (E152) Substances 0.000 description 7
- 101710105178 F-box/WD repeat-containing protein 7 Proteins 0.000 description 7
- 102100028138 F-box/WD repeat-containing protein 7 Human genes 0.000 description 7
- 239000008346 aqueous phase Substances 0.000 description 7
- 230000008901 benefit Effects 0.000 description 7
- 229930195733 hydrocarbon Natural products 0.000 description 7
- 239000012046 mixed solvent Substances 0.000 description 7
- 125000006413 ring segment Chemical group 0.000 description 7
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 6
- 102100025064 Cellular tumor antigen p53 Human genes 0.000 description 6
- 101000621390 Homo sapiens Wee1-like protein kinase Proteins 0.000 description 6
- CDBYLPFSWZWCQE-UHFFFAOYSA-L Sodium Carbonate Chemical class [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 6
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 6
- DTQVDTLACAAQTR-UHFFFAOYSA-N Trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 6
- 102100023037 Wee1-like protein kinase Human genes 0.000 description 6
- 125000003545 alkoxy group Chemical group 0.000 description 6
- 229910002092 carbon dioxide Inorganic materials 0.000 description 6
- 239000012043 crude product Substances 0.000 description 6
- 229940079593 drug Drugs 0.000 description 6
- OKKJLVBELUTLKV-VMNATFBRSA-N methanol-d1 Chemical compound [2H]OC OKKJLVBELUTLKV-VMNATFBRSA-N 0.000 description 6
- 230000035772 mutation Effects 0.000 description 6
- 239000011541 reaction mixture Substances 0.000 description 6
- 230000001225 therapeutic effect Effects 0.000 description 6
- JLTRXTDYQLMHGR-UHFFFAOYSA-N trimethylaluminium Chemical compound C[Al](C)C JLTRXTDYQLMHGR-UHFFFAOYSA-N 0.000 description 6
- 125000002373 5 membered heterocyclic group Chemical group 0.000 description 5
- PGCAHTNLXQHIOZ-UHFFFAOYSA-N 5-methyl-1h-indazol-4-amine Chemical group CC1=CC=C2NN=CC2=C1N PGCAHTNLXQHIOZ-UHFFFAOYSA-N 0.000 description 5
- 108020004414 DNA Proteins 0.000 description 5
- ODWXUNBKCRECNW-UHFFFAOYSA-M bromocopper(1+) Chemical compound Br[Cu+] ODWXUNBKCRECNW-UHFFFAOYSA-M 0.000 description 5
- 239000001569 carbon dioxide Substances 0.000 description 5
- 239000003153 chemical reaction reagent Substances 0.000 description 5
- 238000004128 high performance liquid chromatography Methods 0.000 description 5
- 230000005764 inhibitory process Effects 0.000 description 5
- 239000002994 raw material Substances 0.000 description 5
- 229920006395 saturated elastomer Polymers 0.000 description 5
- IOGXOCVLYRDXLW-UHFFFAOYSA-N tert-butyl nitrite Chemical compound CC(C)(C)ON=O IOGXOCVLYRDXLW-UHFFFAOYSA-N 0.000 description 5
- 239000012414 tert-butyl nitrite Substances 0.000 description 5
- 108010034798 CDC2 Protein Kinase Proteins 0.000 description 4
- 102100032857 Cyclin-dependent kinase 1 Human genes 0.000 description 4
- 101000721661 Homo sapiens Cellular tumor antigen p53 Proteins 0.000 description 4
- YNAVUWVOSKDBBP-UHFFFAOYSA-N Morpholine Chemical compound C1COCCN1 YNAVUWVOSKDBBP-UHFFFAOYSA-N 0.000 description 4
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-Diisopropylethylamine (DIPEA) Chemical compound CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 0.000 description 4
- JRNVZBWKYDBUCA-UHFFFAOYSA-N N-chlorosuccinimide Chemical compound ClN1C(=O)CCC1=O JRNVZBWKYDBUCA-UHFFFAOYSA-N 0.000 description 4
- 150000001408 amides Chemical class 0.000 description 4
- 125000002619 bicyclic group Chemical group 0.000 description 4
- GDTBXPJZTBHREO-UHFFFAOYSA-N bromine Substances BrBr GDTBXPJZTBHREO-UHFFFAOYSA-N 0.000 description 4
- 238000006482 condensation reaction Methods 0.000 description 4
- 125000001559 cyclopropyl group Chemical group [H]C1([H])C([H])([H])C1([H])* 0.000 description 4
- 239000005457 ice water Substances 0.000 description 4
- 230000000415 inactivating effect Effects 0.000 description 4
- 239000004615 ingredient Substances 0.000 description 4
- 239000002207 metabolite Substances 0.000 description 4
- 239000011259 mixed solution Substances 0.000 description 4
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 4
- 238000002953 preparative HPLC Methods 0.000 description 4
- 239000000651 prodrug Substances 0.000 description 4
- 229940002612 prodrug Drugs 0.000 description 4
- 239000012453 solvate Substances 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- JOXIMZWYDAKGHI-UHFFFAOYSA-N toluene-4-sulfonic acid Chemical compound CC1=CC=C(S(O)(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-N 0.000 description 4
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 description 4
- MMWRGWQTAMNAFC-UHFFFAOYSA-N 1,2-dihydropyridine Chemical compound C1NC=CC=C1 MMWRGWQTAMNAFC-UHFFFAOYSA-N 0.000 description 3
- WFDIJRYMOXRFFG-UHFFFAOYSA-N Acetic anhydride Chemical compound CC(=O)OC(C)=O WFDIJRYMOXRFFG-UHFFFAOYSA-N 0.000 description 3
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 3
- UHNRLQRZRNKOKU-UHFFFAOYSA-N CCN(CC1=NC2=C(N1)C1=CC=C(C=C1N=C2N)C1=NNC=C1)C(C)=O Chemical compound CCN(CC1=NC2=C(N1)C1=CC=C(C=C1N=C2N)C1=NNC=C1)C(C)=O UHNRLQRZRNKOKU-UHFFFAOYSA-N 0.000 description 3
- 101150056334 Ccne1 gene Proteins 0.000 description 3
- 101100028131 Danio rerio ora4 gene Proteins 0.000 description 3
- 101150095705 FBXW7 gene Proteins 0.000 description 3
- WMFOQBRAJBCJND-UHFFFAOYSA-M Lithium hydroxide Chemical compound [Li+].[OH-] WMFOQBRAJBCJND-UHFFFAOYSA-M 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- KDLHZDBZIXYQEI-UHFFFAOYSA-N Palladium Chemical compound [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 description 3
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 3
- 101150080074 TP53 gene Proteins 0.000 description 3
- 238000005972 Thorpe-Ziegler reaction Methods 0.000 description 3
- 230000002159 abnormal effect Effects 0.000 description 3
- 239000002253 acid Substances 0.000 description 3
- 239000004480 active ingredient Substances 0.000 description 3
- 239000013543 active substance Substances 0.000 description 3
- 125000001931 aliphatic group Chemical group 0.000 description 3
- 239000000611 antibody drug conjugate Substances 0.000 description 3
- 229940049595 antibody-drug conjugate Drugs 0.000 description 3
- 125000005605 benzo group Chemical group 0.000 description 3
- 230000033228 biological regulation Effects 0.000 description 3
- 239000011203 carbon fibre reinforced carbon Substances 0.000 description 3
- 239000003054 catalyst Substances 0.000 description 3
- 230000004663 cell proliferation Effects 0.000 description 3
- 239000007810 chemical reaction solvent Substances 0.000 description 3
- 239000012141 concentrate Substances 0.000 description 3
- 235000008504 concentrate Nutrition 0.000 description 3
- 238000002474 experimental method Methods 0.000 description 3
- 238000000605 extraction Methods 0.000 description 3
- 239000011737 fluorine Substances 0.000 description 3
- 230000002401 inhibitory effect Effects 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- UKVIEHSSVKSQBA-UHFFFAOYSA-N methane;palladium Chemical compound C.[Pd] UKVIEHSSVKSQBA-UHFFFAOYSA-N 0.000 description 3
- 125000000325 methylidene group Chemical group [H]C([H])=* 0.000 description 3
- 125000004433 nitrogen atom Chemical group N* 0.000 description 3
- 230000002018 overexpression Effects 0.000 description 3
- 125000004430 oxygen atom Chemical group O* 0.000 description 3
- 108700025694 p53 Genes Proteins 0.000 description 3
- 239000011591 potassium Substances 0.000 description 3
- 229910052700 potassium Inorganic materials 0.000 description 3
- 238000011865 proteolysis targeting chimera technique Methods 0.000 description 3
- 229940124823 proteolysis targeting chimeric molecule Drugs 0.000 description 3
- 238000000746 purification Methods 0.000 description 3
- 238000007363 ring formation reaction Methods 0.000 description 3
- 108010026668 snake venom protein C activator Proteins 0.000 description 3
- 239000011343 solid material Substances 0.000 description 3
- 125000004434 sulfur atom Chemical group 0.000 description 3
- 238000010189 synthetic method Methods 0.000 description 3
- KZPYGQFFRCFCPP-UHFFFAOYSA-N 1,1'-bis(diphenylphosphino)ferrocene Chemical compound [Fe+2].C1=CC=C[C-]1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=C[C-]1P(C=1C=CC=CC=1)C1=CC=CC=C1 KZPYGQFFRCFCPP-UHFFFAOYSA-N 0.000 description 2
- OKGNMRKOGWTADH-UHFFFAOYSA-N 1,4-dihydropyrimidine Chemical compound C1C=CNC=N1 OKGNMRKOGWTADH-UHFFFAOYSA-N 0.000 description 2
- IMFQYAJJXFXVMM-UHFFFAOYSA-N 1-(isothiocyanatomethyl)-4-methoxybenzene Chemical compound COC1=CC=C(CN=C=S)C=C1 IMFQYAJJXFXVMM-UHFFFAOYSA-N 0.000 description 2
- RSEBUVRVKCANEP-UHFFFAOYSA-N 2-pyrroline Chemical compound C1CC=CN1 RSEBUVRVKCANEP-UHFFFAOYSA-N 0.000 description 2
- BFJMHTOBRRZELQ-UHFFFAOYSA-N 3-iodo-2h-pyrazolo[3,4-c]pyridine Chemical compound N1=CC=C2C(I)=NNC2=C1 BFJMHTOBRRZELQ-UHFFFAOYSA-N 0.000 description 2
- VHYFNPMBLIVWCW-UHFFFAOYSA-N 4-Dimethylaminopyridine Chemical compound CN(C)C1=CC=NC=C1 VHYFNPMBLIVWCW-UHFFFAOYSA-N 0.000 description 2
- 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 2
- NLXLAEXVIDQMFP-UHFFFAOYSA-N Ammonia chloride Chemical class [NH4+].[Cl-] NLXLAEXVIDQMFP-UHFFFAOYSA-N 0.000 description 2
- VHUUQVKOLVNVRT-UHFFFAOYSA-N Ammonium hydroxide Chemical compound [NH4+].[OH-] VHUUQVKOLVNVRT-UHFFFAOYSA-N 0.000 description 2
- 108090000461 Aurora Kinase A Proteins 0.000 description 2
- 102100032311 Aurora kinase A Human genes 0.000 description 2
- 102100032306 Aurora kinase B Human genes 0.000 description 2
- 108090000749 Aurora kinase B Proteins 0.000 description 2
- WKBOTKDWSSQWDR-UHFFFAOYSA-N Bromine atom Chemical compound [Br] WKBOTKDWSSQWDR-UHFFFAOYSA-N 0.000 description 2
- 102100021975 CREB-binding protein Human genes 0.000 description 2
- 108010019244 Checkpoint Kinase 1 Proteins 0.000 description 2
- 102000006459 Checkpoint Kinase 1 Human genes 0.000 description 2
- KZBUYRJDOAKODT-UHFFFAOYSA-N Chlorine Chemical compound ClCl KZBUYRJDOAKODT-UHFFFAOYSA-N 0.000 description 2
- ZAMOUSCENKQFHK-UHFFFAOYSA-N Chlorine atom Chemical compound [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 description 2
- 206010052360 Colorectal adenocarcinoma Diseases 0.000 description 2
- 229910021595 Copper(I) iodide Inorganic materials 0.000 description 2
- 102100033587 DNA topoisomerase 2-alpha Human genes 0.000 description 2
- 102100023933 Deoxyuridine 5'-triphosphate nucleotidohydrolase, mitochondrial Human genes 0.000 description 2
- 102100036109 Dual specificity protein kinase TTK Human genes 0.000 description 2
- YCKRFDGAMUMZLT-UHFFFAOYSA-N Fluorine atom Chemical compound [F] YCKRFDGAMUMZLT-UHFFFAOYSA-N 0.000 description 2
- 230000008051 G1/S transition checkpoint Effects 0.000 description 2
- 208000031448 Genomic Instability Diseases 0.000 description 2
- 102100021455 Histone deacetylase 3 Human genes 0.000 description 2
- 102100039489 Histone-lysine N-methyltransferase, H3 lysine-79 specific Human genes 0.000 description 2
- 101000659223 Homo sapiens Dual specificity protein kinase TTK Proteins 0.000 description 2
- 101000963360 Homo sapiens Histone-lysine N-methyltransferase, H3 lysine-79 specific Proteins 0.000 description 2
- 101001074727 Homo sapiens Ribonucleoside-diphosphate reductase large subunit Proteins 0.000 description 2
- 101000575639 Homo sapiens Ribonucleoside-diphosphate reductase subunit M2 Proteins 0.000 description 2
- 101000989953 Homo sapiens Serine/threonine-protein kinase haspin Proteins 0.000 description 2
- CPELXLSAUQHCOX-UHFFFAOYSA-N Hydrogen bromide Chemical compound Br CPELXLSAUQHCOX-UHFFFAOYSA-N 0.000 description 2
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 2
- 239000005909 Kieselgur Substances 0.000 description 2
- 108700005090 Lethal Genes Proteins 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 2
- LRHPLDYGYMQRHN-UHFFFAOYSA-N N-Butanol Chemical compound CCCCO LRHPLDYGYMQRHN-UHFFFAOYSA-N 0.000 description 2
- SECXISVLQFMRJM-UHFFFAOYSA-N N-Methylpyrrolidone Chemical compound CN1CCCC1=O SECXISVLQFMRJM-UHFFFAOYSA-N 0.000 description 2
- LQZMLBORDGWNPD-UHFFFAOYSA-N N-iodosuccinimide Chemical compound IN1C(=O)CCC1=O LQZMLBORDGWNPD-UHFFFAOYSA-N 0.000 description 2
- 102100036320 Ribonucleoside-diphosphate reductase large subunit Human genes 0.000 description 2
- 102100026006 Ribonucleoside-diphosphate reductase subunit M2 Human genes 0.000 description 2
- 230000018199 S phase Effects 0.000 description 2
- 102100035250 SUMO-activating enzyme subunit 2 Human genes 0.000 description 2
- 101710180715 SUMO-activating enzyme subunit 2 Proteins 0.000 description 2
- 102100031463 Serine/threonine-protein kinase PLK1 Human genes 0.000 description 2
- 102100030267 Serine/threonine-protein kinase PLK4 Human genes 0.000 description 2
- 101710183229 Serine/threonine-protein kinase PLK4 Proteins 0.000 description 2
- 102100029332 Serine/threonine-protein kinase haspin Human genes 0.000 description 2
- WQDUMFSSJAZKTM-UHFFFAOYSA-N Sodium methoxide Chemical compound [Na+].[O-]C WQDUMFSSJAZKTM-UHFFFAOYSA-N 0.000 description 2
- 208000000102 Squamous Cell Carcinoma of Head and Neck Diseases 0.000 description 2
- 208000034254 Squamous cell carcinoma of the cervix uteri Diseases 0.000 description 2
- 108010021188 Superoxide Dismutase-1 Proteins 0.000 description 2
- 102100038836 Superoxide dismutase [Cu-Zn] Human genes 0.000 description 2
- 102100032891 Superoxide dismutase [Mn], mitochondrial Human genes 0.000 description 2
- 108010078814 Tumor Suppressor Protein p53 Proteins 0.000 description 2
- 108010046308 Type II DNA Topoisomerases Proteins 0.000 description 2
- 102000006275 Ubiquitin-Protein Ligases Human genes 0.000 description 2
- 108010083111 Ubiquitin-Protein Ligases Proteins 0.000 description 2
- 239000012391 XPhos Pd G2 Substances 0.000 description 2
- 239000002671 adjuvant Substances 0.000 description 2
- 235000011114 ammonium hydroxide Nutrition 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 239000002246 antineoplastic agent Substances 0.000 description 2
- 239000012298 atmosphere Substances 0.000 description 2
- 206010005084 bladder transitional cell carcinoma Diseases 0.000 description 2
- 201000001528 bladder urothelial carcinoma Diseases 0.000 description 2
- XJHCXCQVJFPJIK-UHFFFAOYSA-M caesium fluoride Chemical compound [F-].[Cs+] XJHCXCQVJFPJIK-UHFFFAOYSA-M 0.000 description 2
- 230000022131 cell cycle Effects 0.000 description 2
- 230000012820 cell cycle checkpoint Effects 0.000 description 2
- 230000030833 cell death Effects 0.000 description 2
- 201000006612 cervical squamous cell carcinoma Diseases 0.000 description 2
- 238000004587 chromatography analysis Methods 0.000 description 2
- LSXDOTMGLUJQCM-UHFFFAOYSA-M copper(i) iodide Chemical compound I[Cu] LSXDOTMGLUJQCM-UHFFFAOYSA-M 0.000 description 2
- 238000005859 coupling reaction Methods 0.000 description 2
- 239000013078 crystal Substances 0.000 description 2
- 108010011219 dUTP pyrophosphatase Proteins 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- 238000001212 derivatisation Methods 0.000 description 2
- WQOXQRCZOLPYPM-UHFFFAOYSA-N dimethyl disulfide Chemical compound CSSC WQOXQRCZOLPYPM-UHFFFAOYSA-N 0.000 description 2
- 238000003821 enantio-separation Methods 0.000 description 2
- 201000003914 endometrial carcinoma Diseases 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- JYSDXODDWAQWJR-UHFFFAOYSA-N ethyl 2-amino-4,5-dimethylthiophene-3-carboxylate Chemical compound CCOC(=O)C1=C(N)SC(C)=C1C JYSDXODDWAQWJR-UHFFFAOYSA-N 0.000 description 2
- 239000012467 final product Substances 0.000 description 2
- 125000005843 halogen group Chemical group 0.000 description 2
- 201000000459 head and neck squamous cell carcinoma Diseases 0.000 description 2
- 108010074724 histone deacetylase 3 Proteins 0.000 description 2
- 230000007062 hydrolysis Effects 0.000 description 2
- 238000006460 hydrolysis reaction Methods 0.000 description 2
- 230000007794 irritation Effects 0.000 description 2
- ZLTPDFXIESTBQG-UHFFFAOYSA-N isothiazole Chemical group C=1C=NSC=1 ZLTPDFXIESTBQG-UHFFFAOYSA-N 0.000 description 2
- 230000000155 isotopic effect Effects 0.000 description 2
- 125000005647 linker group Chemical group 0.000 description 2
- ZCSHNCUQKCANBX-UHFFFAOYSA-N lithium diisopropylamide Chemical compound [Li+].CC(C)[N-]C(C)C ZCSHNCUQKCANBX-UHFFFAOYSA-N 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 230000001404 mediated effect Effects 0.000 description 2
- 125000002816 methylsulfanyl group Chemical group [H]C([H])([H])S[*] 0.000 description 2
- 238000006386 neutralization reaction Methods 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 125000004437 phosphorous atom Chemical group 0.000 description 2
- 230000000865 phosphorylative effect Effects 0.000 description 2
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 2
- 108010056274 polo-like kinase 1 Proteins 0.000 description 2
- 125000003367 polycyclic group Chemical group 0.000 description 2
- NROKBHXJSPEDAR-UHFFFAOYSA-M potassium fluoride Chemical compound [F-].[K+] NROKBHXJSPEDAR-UHFFFAOYSA-M 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 125000006239 protecting group Chemical group 0.000 description 2
- 125000003373 pyrazinyl group Chemical group 0.000 description 2
- ABMYEXAYWZJVOV-UHFFFAOYSA-N pyridin-3-ylboronic acid Chemical compound OB(O)C1=CC=CN=C1 ABMYEXAYWZJVOV-UHFFFAOYSA-N 0.000 description 2
- 125000000168 pyrrolyl group Chemical group 0.000 description 2
- 125000002294 quinazolinyl group Chemical group N1=C(N=CC2=CC=CC=C12)* 0.000 description 2
- 230000002285 radioactive effect Effects 0.000 description 2
- 230000001105 regulatory effect Effects 0.000 description 2
- 230000028617 response to DNA damage stimulus Effects 0.000 description 2
- 230000002441 reversible effect Effects 0.000 description 2
- 150000003384 small molecules Chemical class 0.000 description 2
- 239000011734 sodium Substances 0.000 description 2
- LPXPTNMVRIOKMN-UHFFFAOYSA-M sodium nitrite Chemical compound [Na+].[O-]N=O LPXPTNMVRIOKMN-UHFFFAOYSA-M 0.000 description 2
- 108010045815 superoxide dismutase 2 Proteins 0.000 description 2
- DYHSDKLCOJIUFX-UHFFFAOYSA-N tert-butoxycarbonyl anhydride Chemical compound CC(C)(C)OC(=O)OC(=O)OC(C)(C)C DYHSDKLCOJIUFX-UHFFFAOYSA-N 0.000 description 2
- HJUGFYREWKUQJT-UHFFFAOYSA-N tetrabromomethane Chemical compound BrC(Br)(Br)Br HJUGFYREWKUQJT-UHFFFAOYSA-N 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 210000001519 tissue Anatomy 0.000 description 2
- 231100000419 toxicity Toxicity 0.000 description 2
- 230000001988 toxicity Effects 0.000 description 2
- 230000007704 transition Effects 0.000 description 2
- PBIMIGNDTBRRPI-UHFFFAOYSA-N trifluoro borate Chemical compound FOB(OF)OF PBIMIGNDTBRRPI-UHFFFAOYSA-N 0.000 description 2
- LWIHDJKSTIGBAC-UHFFFAOYSA-K tripotassium phosphate Chemical compound [K+].[K+].[K+].[O-]P([O-])([O-])=O LWIHDJKSTIGBAC-UHFFFAOYSA-K 0.000 description 2
- 239000003643 water by type Substances 0.000 description 2
- KEFRAFHUHLKQCE-UHFFFAOYSA-N (1-methylpyrazol-4-yl)oxyboronic acid Chemical compound CN1C=C(OB(O)O)C=N1 KEFRAFHUHLKQCE-UHFFFAOYSA-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
- 239000001082 (3R)-3-sulfanylpentan-2-one Substances 0.000 description 1
- 125000006656 (C2-C4) alkenyl group Chemical group 0.000 description 1
- 125000006650 (C2-C4) alkynyl group Chemical group 0.000 description 1
- 125000006552 (C3-C8) cycloalkyl group Chemical group 0.000 description 1
- DELJOESCKJGFML-RQOWECAXSA-N (z)-3-aminobut-2-enenitrile Chemical compound C\C(N)=C\C#N DELJOESCKJGFML-RQOWECAXSA-N 0.000 description 1
- SCYULBFZEHDVBN-UHFFFAOYSA-N 1,1-Dichloroethane Chemical compound CC(Cl)Cl SCYULBFZEHDVBN-UHFFFAOYSA-N 0.000 description 1
- LRANPJDWHYRCER-UHFFFAOYSA-N 1,2-diazepine Chemical group N1C=CC=CC=N1 LRANPJDWHYRCER-UHFFFAOYSA-N 0.000 description 1
- WCFAPJDPAPDDAQ-UHFFFAOYSA-N 1,2-dihydropyrimidine Chemical compound C1NC=CC=N1 WCFAPJDPAPDDAQ-UHFFFAOYSA-N 0.000 description 1
- 125000005918 1,2-dimethylbutyl group Chemical group 0.000 description 1
- LMDZBCPBFSXMTL-UHFFFAOYSA-N 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide Substances CCN=C=NCCCN(C)C LMDZBCPBFSXMTL-UHFFFAOYSA-N 0.000 description 1
- ASOKPJOREAFHNY-UHFFFAOYSA-N 1-Hydroxybenzotriazole Chemical compound C1=CC=C2N(O)N=NC2=C1 ASOKPJOREAFHNY-UHFFFAOYSA-N 0.000 description 1
- 125000004972 1-butynyl group Chemical group [H]C([H])([H])C([H])([H])C#C* 0.000 description 1
- 125000006218 1-ethylbutyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C([H])([H])[H] 0.000 description 1
- 238000005160 1H NMR spectroscopy Methods 0.000 description 1
- MDELYEBAXHZXLZ-UHFFFAOYSA-N 1h-indazol-4-amine Chemical compound NC1=CC=CC2=C1C=NN2 MDELYEBAXHZXLZ-UHFFFAOYSA-N 0.000 description 1
- FJZJUSOFGBXHCV-UHFFFAOYSA-N 2,2-dimethylpropanethioamide Chemical compound CC(C)(C)C(N)=S FJZJUSOFGBXHCV-UHFFFAOYSA-N 0.000 description 1
- ZABMHLDQFJHDSC-UHFFFAOYSA-N 2,3-dihydro-1,3-oxazole Chemical group C1NC=CO1 ZABMHLDQFJHDSC-UHFFFAOYSA-N 0.000 description 1
- JKTCBAGSMQIFNL-UHFFFAOYSA-N 2,3-dihydrofuran Chemical compound C1CC=CO1 JKTCBAGSMQIFNL-UHFFFAOYSA-N 0.000 description 1
- BZYUMXXOAYSFOW-UHFFFAOYSA-N 2,3-dimethylthiophene Chemical compound CC=1C=CSC=1C BZYUMXXOAYSFOW-UHFFFAOYSA-N 0.000 description 1
- VHMOYJYPQAAQJZ-UHFFFAOYSA-N 2,4-dibromo-1,3-thiazole-5-carboxylic acid Chemical compound OC(=O)C=1SC(Br)=NC=1Br VHMOYJYPQAAQJZ-UHFFFAOYSA-N 0.000 description 1
- WAQFYSJKIRRXLP-UHFFFAOYSA-N 2,4-dibromothiophene Chemical compound BrC1=CSC(Br)=C1 WAQFYSJKIRRXLP-UHFFFAOYSA-N 0.000 description 1
- ACDLOOGOFKSUPO-UHFFFAOYSA-N 2-bromo-5-methylthiophene Chemical compound CC1=CC=C(Br)S1 ACDLOOGOFKSUPO-UHFFFAOYSA-N 0.000 description 1
- HORDCPFFRDZLRG-UHFFFAOYSA-N 2-bromo-6-fluoro-3-methylbenzaldehyde Chemical compound CC1=CC=C(F)C(C=O)=C1Br HORDCPFFRDZLRG-UHFFFAOYSA-N 0.000 description 1
- RVSXMPCELBYUSF-UHFFFAOYSA-N 2-bromothiophene-3-carboxylic acid Chemical compound OC(=O)C=1C=CSC=1Br RVSXMPCELBYUSF-UHFFFAOYSA-N 0.000 description 1
- 125000000069 2-butynyl group Chemical group [H]C([H])([H])C#CC([H])([H])* 0.000 description 1
- 125000006176 2-ethylbutyl group Chemical group [H]C([H])([H])C([H])([H])C([H])(C([H])([H])*)C([H])([H])C([H])([H])[H] 0.000 description 1
- MVIVDSWUOGNODP-UHFFFAOYSA-N 2-iodobenzoyl chloride Chemical compound ClC(=O)C1=CC=CC=C1I MVIVDSWUOGNODP-UHFFFAOYSA-N 0.000 description 1
- ZHDRDZMTEOIWSX-UHFFFAOYSA-N 2-methyl-1,3-thiazole-4-carboxylic acid Chemical compound CC1=NC(C(O)=O)=CS1 ZHDRDZMTEOIWSX-UHFFFAOYSA-N 0.000 description 1
- 125000004493 2-methylbut-1-yl group Chemical group CC(C*)CC 0.000 description 1
- 125000005916 2-methylpentyl group Chemical group 0.000 description 1
- 125000003903 2-propenyl group Chemical group [H]C([*])([H])C([H])=C([H])[H] 0.000 description 1
- MGADZUXDNSDTHW-UHFFFAOYSA-N 2H-pyran Chemical compound C1OC=CC=C1 MGADZUXDNSDTHW-UHFFFAOYSA-N 0.000 description 1
- QMDFJHAAWUGVKQ-UHFFFAOYSA-N 2h-thiopyran Chemical compound C1SC=CC=C1 QMDFJHAAWUGVKQ-UHFFFAOYSA-N 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
- SZECUQRKLXRGSJ-UHFFFAOYSA-N 3-Mercapto-2-pentanone Chemical compound CCC(S)C(C)=O SZECUQRKLXRGSJ-UHFFFAOYSA-N 0.000 description 1
- 125000000474 3-butynyl group Chemical group [H]C#CC([H])([H])C([H])([H])* 0.000 description 1
- NHQDETIJWKXCTC-UHFFFAOYSA-N 3-chloroperbenzoic acid Chemical compound OOC(=O)C1=CC=CC(Cl)=C1 NHQDETIJWKXCTC-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
- 125000005917 3-methylpentyl group Chemical group 0.000 description 1
- SVSUYEJKNSMKKW-UHFFFAOYSA-N 4,4,5,5-tetramethyl-2-prop-1-en-2-yl-1,3,2-dioxaborolane Chemical compound CC(=C)B1OC(C)(C)C(C)(C)O1 SVSUYEJKNSMKKW-UHFFFAOYSA-N 0.000 description 1
- ABMUSXPGSSMPLK-UHFFFAOYSA-N 4-bromo-2-methylthiophene Chemical compound CC1=CC(Br)=CS1 ABMUSXPGSSMPLK-UHFFFAOYSA-N 0.000 description 1
- LRFIHWGUGBXFEC-UHFFFAOYSA-N 4-methylthiophene-3-carboxylic acid Chemical compound CC1=CSC=C1C(O)=O LRFIHWGUGBXFEC-UHFFFAOYSA-N 0.000 description 1
- MRUWJENAYHTDQG-UHFFFAOYSA-N 4H-pyran Chemical compound C1C=COC=C1 MRUWJENAYHTDQG-UHFFFAOYSA-N 0.000 description 1
- STBGWNRZMPCVTG-UHFFFAOYSA-N 4h-thiopyran Chemical compound C1C=CSC=C1 STBGWNRZMPCVTG-UHFFFAOYSA-N 0.000 description 1
- ARHCLXWELPFVFQ-UHFFFAOYSA-N 5-bromo-1,3-thiazol-2-amine Chemical compound NC1=NC=C(Br)S1 ARHCLXWELPFVFQ-UHFFFAOYSA-N 0.000 description 1
- YCNXGPMGMAKDPM-UHFFFAOYSA-N 5-bromothiophene-3-carboxylic acid Chemical compound OC(=O)C1=CSC(Br)=C1 YCNXGPMGMAKDPM-UHFFFAOYSA-N 0.000 description 1
- BHOSWVZIGTVQNK-UHFFFAOYSA-N 5-chloro-1h-indazol-4-amine Chemical group NC1=C(Cl)C=CC2=C1C=NN2 BHOSWVZIGTVQNK-UHFFFAOYSA-N 0.000 description 1
- ZCYVEMRRCGMTRW-UHFFFAOYSA-N 7553-56-2 Chemical compound [I] ZCYVEMRRCGMTRW-UHFFFAOYSA-N 0.000 description 1
- WCOCYZXXISYZLN-UHFFFAOYSA-N B(F)(F)F.C=C.[K] Chemical compound B(F)(F)F.C=C.[K] WCOCYZXXISYZLN-UHFFFAOYSA-N 0.000 description 1
- ROFVEXUMMXZLPA-UHFFFAOYSA-N Bipyridyl Chemical compound N1=CC=CC=C1C1=CC=CC=N1 ROFVEXUMMXZLPA-UHFFFAOYSA-N 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 238000007125 Buchwald synthesis reaction Methods 0.000 description 1
- 125000004648 C2-C8 alkenyl group Chemical group 0.000 description 1
- 125000004649 C2-C8 alkynyl group Chemical group 0.000 description 1
- 125000001313 C5-C10 heteroaryl group Chemical group 0.000 description 1
- NJTZSWLQBQJUHK-UHFFFAOYSA-N CCCP(=O)=O Chemical compound CCCP(=O)=O NJTZSWLQBQJUHK-UHFFFAOYSA-N 0.000 description 1
- 108010040163 CREB-Binding Protein Proteins 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 102000005483 Cell Cycle Proteins Human genes 0.000 description 1
- 108010031896 Cell Cycle Proteins Proteins 0.000 description 1
- 229910021589 Copper(I) bromide Inorganic materials 0.000 description 1
- 241000938605 Crocodylia Species 0.000 description 1
- 102000003909 Cyclin E Human genes 0.000 description 1
- 108090000257 Cyclin E Proteins 0.000 description 1
- 108010024986 Cyclin-Dependent Kinase 2 Proteins 0.000 description 1
- 102100036239 Cyclin-dependent kinase 2 Human genes 0.000 description 1
- PMPVIKIVABFJJI-UHFFFAOYSA-N Cyclobutane Chemical compound C1CCC1 PMPVIKIVABFJJI-UHFFFAOYSA-N 0.000 description 1
- 108090000323 DNA Topoisomerases Proteins 0.000 description 1
- 102000003915 DNA Topoisomerases Human genes 0.000 description 1
- 239000012624 DNA alkylating agent Substances 0.000 description 1
- 230000004544 DNA amplification Effects 0.000 description 1
- 230000005778 DNA damage Effects 0.000 description 1
- 231100000277 DNA damage Toxicity 0.000 description 1
- 239000012623 DNA damaging agent Substances 0.000 description 1
- 239000012625 DNA intercalator Substances 0.000 description 1
- 230000008265 DNA repair mechanism Effects 0.000 description 1
- 230000004543 DNA replication Effects 0.000 description 1
- 102100024607 DNA topoisomerase 1 Human genes 0.000 description 1
- WLTCKEHCTUYJGI-UHFFFAOYSA-N Diethyl aminomalonate Chemical compound CCOC(=O)C(N)C(=O)OCC WLTCKEHCTUYJGI-UHFFFAOYSA-N 0.000 description 1
- 101100114828 Drosophila melanogaster Orai gene Proteins 0.000 description 1
- 101710180995 Endonuclease 1 Proteins 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- PXGOKWXKJXAPGV-UHFFFAOYSA-N Fluorine Chemical compound FF PXGOKWXKJXAPGV-UHFFFAOYSA-N 0.000 description 1
- 230000010190 G1 phase Effects 0.000 description 1
- 101710170962 G1/S-specific cyclin-E1 Proteins 0.000 description 1
- 239000007821 HATU Substances 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000896987 Homo sapiens CREB-binding protein Proteins 0.000 description 1
- 101000830681 Homo sapiens DNA topoisomerase 1 Proteins 0.000 description 1
- 101000896657 Homo sapiens Mitotic checkpoint serine/threonine-protein kinase BUB1 Proteins 0.000 description 1
- 101000794228 Homo sapiens Mitotic checkpoint serine/threonine-protein kinase BUB1 beta Proteins 0.000 description 1
- 101000967135 Homo sapiens N6-adenosine-methyltransferase catalytic subunit Proteins 0.000 description 1
- 101000693367 Homo sapiens SUMO-activating enzyme subunit 1 Proteins 0.000 description 1
- 101000785063 Homo sapiens Serine-protein kinase ATM Proteins 0.000 description 1
- 101001059454 Homo sapiens Serine/threonine-protein kinase MARK2 Proteins 0.000 description 1
- 102100030550 Menin Human genes 0.000 description 1
- 101710169972 Menin Proteins 0.000 description 1
- 108060004795 Methyltransferase Proteins 0.000 description 1
- 102000016397 Methyltransferase Human genes 0.000 description 1
- 102100021691 Mitotic checkpoint serine/threonine-protein kinase BUB1 Human genes 0.000 description 1
- 102100030144 Mitotic checkpoint serine/threonine-protein kinase BUB1 beta Human genes 0.000 description 1
- NQTADLQHYWFPDB-UHFFFAOYSA-N N-Hydroxysuccinimide Chemical compound ON1C(=O)CCC1=O NQTADLQHYWFPDB-UHFFFAOYSA-N 0.000 description 1
- 102100040619 N6-adenosine-methyltransferase catalytic subunit Human genes 0.000 description 1
- CTQNGGLPUBDAKN-UHFFFAOYSA-N O-Xylene Chemical compound CC1=CC=CC=C1C CTQNGGLPUBDAKN-UHFFFAOYSA-N 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 108010000597 Polycomb Repressive Complex 2 Proteins 0.000 description 1
- 102000002272 Polycomb Repressive Complex 2 Human genes 0.000 description 1
- 102000001253 Protein Kinase Human genes 0.000 description 1
- RWRDLPDLKQPQOW-UHFFFAOYSA-N Pyrrolidine Chemical compound C1CCNC1 RWRDLPDLKQPQOW-UHFFFAOYSA-N 0.000 description 1
- 102100025809 SUMO-activating enzyme subunit 1 Human genes 0.000 description 1
- 101100406516 Schizosaccharomyces pombe (strain 972 / ATCC 24843) orb6 gene Proteins 0.000 description 1
- 102100020824 Serine-protein kinase ATM Human genes 0.000 description 1
- 102100028904 Serine/threonine-protein kinase MARK2 Human genes 0.000 description 1
- 108020004459 Small interfering RNA Proteins 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- OKJPEAGHQZHRQV-UHFFFAOYSA-N Triiodomethane Natural products IC(I)I OKJPEAGHQZHRQV-UHFFFAOYSA-N 0.000 description 1
- 238000006887 Ullmann reaction Methods 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- DSVGQVZAZSZEEX-UHFFFAOYSA-N [C].[Pt] Chemical compound [C].[Pt] DSVGQVZAZSZEEX-UHFFFAOYSA-N 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 125000000641 acridinyl group Chemical group C1(=CC=CC2=NC3=CC=CC=C3C=C12)* 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 125000001118 alkylidene group Chemical group 0.000 description 1
- 208000026935 allergic disease Diseases 0.000 description 1
- 230000009435 amidation Effects 0.000 description 1
- 238000007112 amidation reaction Methods 0.000 description 1
- 125000003368 amide group Chemical group 0.000 description 1
- 125000002178 anthracenyl group Chemical group C1(=CC=CC2=CC3=CC=CC=C3C=C12)* 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 125000004931 azocinyl group Chemical group N1=C(C=CC=CC=C1)* 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
- 125000004603 benzisoxazolyl group Chemical group O1N=C(C2=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
- 125000003354 benzotriazolyl group Chemical group N1N=NC2=C1C=CC=C2* 0.000 description 1
- 125000004541 benzoxazolyl group Chemical group O1C(=NC2=C1C=CC=C2)* 0.000 description 1
- ALXLNFWWLXCXSK-UHFFFAOYSA-N benzyl 3-oxopiperidine-1-carboxylate Chemical compound C1CCC(=O)CN1C(=O)OCC1=CC=CC=C1 ALXLNFWWLXCXSK-UHFFFAOYSA-N 0.000 description 1
- 125000002618 bicyclic heterocycle group Chemical group 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- HGXJOXHYPGNVNK-UHFFFAOYSA-N butane;ethenoxyethane;tin Chemical compound CCCC[Sn](CCCC)(CCCC)C(=C)OCC HGXJOXHYPGNVNK-UHFFFAOYSA-N 0.000 description 1
- 239000012830 cancer therapeutic Substances 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 125000000609 carbazolyl group Chemical group C1(=CC=CC=2C3=CC=CC=C3NC12)* 0.000 description 1
- 150000001721 carbon Chemical group 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000025084 cell cycle arrest Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 150000005829 chemical entities Chemical class 0.000 description 1
- 239000007795 chemical reaction product Substances 0.000 description 1
- 238000004296 chiral HPLC Methods 0.000 description 1
- 125000000259 cinnolinyl group Chemical group N1=NC(=CC2=CC=CC=C12)* 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 238000009833 condensation Methods 0.000 description 1
- 230000005494 condensation Effects 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- NKNDPYCGAZPOFS-UHFFFAOYSA-M copper(i) bromide Chemical compound Br[Cu] NKNDPYCGAZPOFS-UHFFFAOYSA-M 0.000 description 1
- DOBRDRYODQBAMW-UHFFFAOYSA-N copper(i) cyanide Chemical compound [Cu+].N#[C-] DOBRDRYODQBAMW-UHFFFAOYSA-N 0.000 description 1
- 239000006071 cream Substances 0.000 description 1
- DGJMPUGMZIKDRO-UHFFFAOYSA-N cyanoacetamide Chemical compound NC(=O)CC#N DGJMPUGMZIKDRO-UHFFFAOYSA-N 0.000 description 1
- 125000004122 cyclic group Chemical group 0.000 description 1
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 125000000113 cyclohexyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 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 1
- 125000002433 cyclopentenyl group Chemical group C1(=CCCC1)* 0.000 description 1
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 229940127089 cytotoxic agent Drugs 0.000 description 1
- 230000006240 deamidation Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 230000017858 demethylation Effects 0.000 description 1
- 238000010520 demethylation reaction Methods 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- NKLCNNUWBJBICK-UHFFFAOYSA-N dess–martin periodinane Chemical compound C1=CC=C2I(OC(=O)C)(OC(C)=O)(OC(C)=O)OC(=O)C2=C1 NKLCNNUWBJBICK-UHFFFAOYSA-N 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- HPNMFZURTQLUMO-UHFFFAOYSA-N diethylamine Chemical compound CCNCC HPNMFZURTQLUMO-UHFFFAOYSA-N 0.000 description 1
- 125000005045 dihydroisoquinolinyl group Chemical group C1(NC=CC2=CC=CC=C12)* 0.000 description 1
- 125000005048 dihydroisoxazolyl group Chemical group O1N(CC=C1)* 0.000 description 1
- UAOMVDZJSHZZME-UHFFFAOYSA-N diisopropylamine Chemical compound CC(C)NC(C)C UAOMVDZJSHZZME-UHFFFAOYSA-N 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- YOTZYFSGUCFUKA-UHFFFAOYSA-N dimethylphosphine Chemical compound CPC YOTZYFSGUCFUKA-UHFFFAOYSA-N 0.000 description 1
- SXZIXHOMFPUIRK-UHFFFAOYSA-N diphenylmethanimine Chemical compound C=1C=CC=CC=1C(=N)C1=CC=CC=C1 SXZIXHOMFPUIRK-UHFFFAOYSA-N 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 125000005883 dithianyl group Chemical group 0.000 description 1
- 239000003534 dna topoisomerase inhibitor Substances 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 230000037437 driver mutation Effects 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 238000006911 enzymatic reaction Methods 0.000 description 1
- QTTMOCOWZLSYSV-QWAPEVOJSA-M equilin sodium sulfate Chemical compound [Na+].[O-]S(=O)(=O)OC1=CC=C2[C@H]3CC[C@](C)(C(CC4)=O)[C@@H]4C3=CCC2=C1 QTTMOCOWZLSYSV-QWAPEVOJSA-M 0.000 description 1
- 230000032050 esterification Effects 0.000 description 1
- 238000005886 esterification reaction Methods 0.000 description 1
- AYALPHBHKQJOMW-UHFFFAOYSA-N ethene 2,2,2-trifluoroacetic acid Chemical compound FC(C(=O)O)(F)F.C=C AYALPHBHKQJOMW-UHFFFAOYSA-N 0.000 description 1
- AEOCXXJPGCBFJA-UHFFFAOYSA-N ethionamide Chemical compound CCC1=CC(C(N)=S)=CC=N1 AEOCXXJPGCBFJA-UHFFFAOYSA-N 0.000 description 1
- QRYDVMPGEGDIGJ-UHFFFAOYSA-N ethyl 2-(3-bromopyridin-2-yl)acetate Chemical compound CCOC(=O)CC1=NC=CC=C1Br QRYDVMPGEGDIGJ-UHFFFAOYSA-N 0.000 description 1
- CDYVTVLXEWMCHU-UHFFFAOYSA-N ethyl 2-amino-4,5,6,7-tetrahydro-1-benzothiophene-3-carboxylate Chemical group C1CCCC2=C1SC(N)=C2C(=O)OCC CDYVTVLXEWMCHU-UHFFFAOYSA-N 0.000 description 1
- KRHUTJDRKGFWQD-UHFFFAOYSA-N ethyl 2-bromo-4,5,6,7-tetrahydro-1-benzothiophene-3-carboxylate Chemical compound C1CCCC2=C1SC(Br)=C2C(=O)OCC KRHUTJDRKGFWQD-UHFFFAOYSA-N 0.000 description 1
- ZIUSEGSNTOUIPT-UHFFFAOYSA-N ethyl 2-cyanoacetate Chemical compound CCOC(=O)CC#N ZIUSEGSNTOUIPT-UHFFFAOYSA-N 0.000 description 1
- VICYTAYPKBLQFB-UHFFFAOYSA-N ethyl 3-bromo-2-oxopropanoate Chemical compound CCOC(=O)C(=O)CBr VICYTAYPKBLQFB-UHFFFAOYSA-N 0.000 description 1
- WAFDSQXKSGGKCD-UHFFFAOYSA-N ethyl 4-amino-2-methylsulfanyl-1,3-thiazole-5-carboxylate Chemical compound CCOC(=O)C=1SC(SC)=NC=1N WAFDSQXKSGGKCD-UHFFFAOYSA-N 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 235000003599 food sweetener Nutrition 0.000 description 1
- 238000001640 fractional crystallisation Methods 0.000 description 1
- 125000002541 furyl group Chemical group 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 238000010438 heat treatment Methods 0.000 description 1
- 125000000592 heterocycloalkyl group Chemical group 0.000 description 1
- 150000004677 hydrates Chemical class 0.000 description 1
- 229910000042 hydrogen bromide Inorganic materials 0.000 description 1
- 230000003301 hydrolyzing effect Effects 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- 125000002632 imidazolidinyl group Chemical group 0.000 description 1
- 125000002883 imidazolyl group Chemical group 0.000 description 1
- 229940044680 immune agonist Drugs 0.000 description 1
- 239000012651 immune agonist Substances 0.000 description 1
- 238000009169 immunotherapy Methods 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 125000003453 indazolyl group Chemical group N1N=C(C2=C1C=CC=C2)* 0.000 description 1
- 125000003406 indolizinyl group Chemical group C=1(C=CN2C=CC=CC12)* 0.000 description 1
- 125000001041 indolyl group Chemical group 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 229910052740 iodine Inorganic materials 0.000 description 1
- 239000011630 iodine Substances 0.000 description 1
- OCVXZQOKBHXGRU-UHFFFAOYSA-N iodine(1+) Chemical group [I+] OCVXZQOKBHXGRU-UHFFFAOYSA-N 0.000 description 1
- INQOMBQAUSQDDS-UHFFFAOYSA-N iodomethane Chemical compound IC INQOMBQAUSQDDS-UHFFFAOYSA-N 0.000 description 1
- 125000000959 isobutyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])* 0.000 description 1
- 125000004491 isohexyl group Chemical group C(CCC(C)C)* 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
- 125000005956 isoquinolyl group Chemical group 0.000 description 1
- 125000001786 isothiazolyl group Chemical group 0.000 description 1
- 125000000842 isoxazolyl group Chemical group 0.000 description 1
- 229940043355 kinase inhibitor Drugs 0.000 description 1
- CFHGBZLNZZVTAY-UHFFFAOYSA-N lawesson's reagent Chemical compound C1=CC(OC)=CC=C1P1(=S)SP(=S)(C=2C=CC(OC)=CC=2)S1 CFHGBZLNZZVTAY-UHFFFAOYSA-N 0.000 description 1
- 231100000518 lethal Toxicity 0.000 description 1
- 230000001665 lethal effect Effects 0.000 description 1
- 231100000225 lethality Toxicity 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 244000144972 livestock Species 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 239000006210 lotion Substances 0.000 description 1
- 239000007937 lozenge Substances 0.000 description 1
- 230000036210 malignancy Effects 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- VNWKTOKETHGBQD-UHFFFAOYSA-N methane Natural products C VNWKTOKETHGBQD-UHFFFAOYSA-N 0.000 description 1
- 229940098779 methanesulfonic acid Drugs 0.000 description 1
- AUTCCPQKLPMHDN-ONEGZZNKSA-N methyl (e)-3-methoxyprop-2-enoate Chemical compound CO\C=C\C(=O)OC AUTCCPQKLPMHDN-ONEGZZNKSA-N 0.000 description 1
- JEENWEAPRWGXSG-UHFFFAOYSA-N methyl 2-oxocyclohexane-1-carboxylate Chemical compound COC(=O)C1CCCCC1=O JEENWEAPRWGXSG-UHFFFAOYSA-N 0.000 description 1
- 239000003094 microcapsule Substances 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001280 n-hexyl group Chemical group C(CCCCC)* 0.000 description 1
- 125000000740 n-pentyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 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
- 125000004593 naphthyridinyl group Chemical group N1=C(C=CC2=CC=CN=C12)* 0.000 description 1
- 125000001971 neopentyl group Chemical group [H]C([*])([H])C(C([H])([H])[H])(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 125000006574 non-aromatic ring group Chemical group 0.000 description 1
- UMRZSTCPUPJPOJ-KNVOCYPGSA-N norbornane Chemical compound C1C[C@H]2CC[C@@H]1C2 UMRZSTCPUPJPOJ-KNVOCYPGSA-N 0.000 description 1
- 239000002777 nucleoside Substances 0.000 description 1
- 150000003833 nucleoside derivatives Chemical class 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 125000001715 oxadiazolyl group Chemical group 0.000 description 1
- AVPKHOTUOHDTLW-UHFFFAOYSA-N oxane-4-carboxylic acid Chemical compound OC(=O)C1CCOCC1 AVPKHOTUOHDTLW-UHFFFAOYSA-N 0.000 description 1
- 125000002971 oxazolyl group Chemical group 0.000 description 1
- 125000003566 oxetanyl group Chemical group 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- 125000005476 oxopyrrolidinyl group Chemical group 0.000 description 1
- 229910052763 palladium Inorganic materials 0.000 description 1
- PIBWKRNGBLPSSY-UHFFFAOYSA-L palladium(II) chloride Chemical compound Cl[Pd]Cl PIBWKRNGBLPSSY-UHFFFAOYSA-L 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
- 239000008177 pharmaceutical agent Substances 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 1
- 125000004592 phthalazinyl group Chemical group C1(=NN=CC2=CC=CC=C12)* 0.000 description 1
- 238000000554 physical therapy Methods 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 125000004193 piperazinyl group Chemical group 0.000 description 1
- 229910052697 platinum Inorganic materials 0.000 description 1
- 239000011698 potassium fluoride Substances 0.000 description 1
- 235000003270 potassium fluoride Nutrition 0.000 description 1
- 229910000160 potassium phosphate Inorganic materials 0.000 description 1
- 235000011009 potassium phosphates Nutrition 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 238000000634 powder X-ray diffraction Methods 0.000 description 1
- 239000002244 precipitate Substances 0.000 description 1
- 238000004262 preparative liquid chromatography Methods 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000002062 proliferating effect Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 108060006633 protein kinase Proteins 0.000 description 1
- 238000001671 psychotherapy Methods 0.000 description 1
- 125000000561 purinyl group Chemical group N1=C(N=C2N=CNC2=C1)* 0.000 description 1
- 125000003072 pyrazolidinyl group Chemical group 0.000 description 1
- 125000002098 pyridazinyl group Chemical group 0.000 description 1
- 125000000714 pyrimidinyl group Chemical group 0.000 description 1
- 125000000719 pyrrolidinyl group Chemical group 0.000 description 1
- 125000001422 pyrrolinyl group Chemical group 0.000 description 1
- 230000000171 quenching effect Effects 0.000 description 1
- 125000005493 quinolyl group Chemical group 0.000 description 1
- 125000001567 quinoxalinyl group Chemical group N1=C(C=NC2=CC=CC=C12)* 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 238000006722 reduction reaction Methods 0.000 description 1
- 230000008439 repair process Effects 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 230000007017 scission Effects 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
- 125000003548 sec-pentyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 1
- 235000017557 sodium bicarbonate Nutrition 0.000 description 1
- 229910000029 sodium carbonate Inorganic materials 0.000 description 1
- QDRKDTQENPPHOJ-UHFFFAOYSA-N sodium ethoxide Chemical compound [Na+].CC[O-] QDRKDTQENPPHOJ-UHFFFAOYSA-N 0.000 description 1
- 235000010288 sodium nitrite Nutrition 0.000 description 1
- AKHNMLFCWUSKQB-UHFFFAOYSA-L sodium thiosulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=S AKHNMLFCWUSKQB-UHFFFAOYSA-L 0.000 description 1
- 235000019345 sodium thiosulphate Nutrition 0.000 description 1
- 239000012321 sodium triacetoxyborohydride Substances 0.000 description 1
- LBJQKYPPYSCCBH-UHFFFAOYSA-N spiro[3.3]heptane Chemical compound C1CCC21CCC2 LBJQKYPPYSCCBH-UHFFFAOYSA-N 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 239000007858 starting material Substances 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 239000003765 sweetening agent Substances 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 239000006188 syrup Substances 0.000 description 1
- 235000020357 syrup Nutrition 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- WMXCDAVJEZZYLT-UHFFFAOYSA-N tert-butylthiol Chemical compound CC(C)(C)S WMXCDAVJEZZYLT-UHFFFAOYSA-N 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
- 125000003718 tetrahydrofuranyl group Chemical group 0.000 description 1
- 125000001412 tetrahydropyranyl group Chemical group 0.000 description 1
- 125000001113 thiadiazolyl group Chemical group 0.000 description 1
- 125000001544 thienyl group Chemical group 0.000 description 1
- 238000006177 thiolation reaction Methods 0.000 description 1
- 125000004568 thiomorpholinyl group Chemical group 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 125000004306 triazinyl group Chemical group 0.000 description 1
- 125000001425 triazolyl group Chemical group 0.000 description 1
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 description 1
- MWKJTNBSKNUMFN-UHFFFAOYSA-N trifluoromethyltrimethylsilane Chemical compound C[Si](C)(C)C(F)(F)F MWKJTNBSKNUMFN-UHFFFAOYSA-N 0.000 description 1
- 230000001960 triggered effect Effects 0.000 description 1
- WUUHFRRPHJEEKV-UHFFFAOYSA-N tripotassium borate Chemical compound [K+].[K+].[K+].[O-]B([O-])[O-] WUUHFRRPHJEEKV-UHFFFAOYSA-N 0.000 description 1
- HJOAXCLZLHDZDX-UHFFFAOYSA-N tris(1,2,2-trifluoroethenyl) borate Chemical compound FC(F)=C(F)OB(OC(F)=C(F)F)OC(F)=C(F)F HJOAXCLZLHDZDX-UHFFFAOYSA-N 0.000 description 1
- 125000005455 trithianyl group Chemical group 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 125000000391 vinyl group Chemical group [H]C([*])=C([H])[H] 0.000 description 1
- 229920002554 vinyl polymer Polymers 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 239000008096 xylene Substances 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—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
- A61K31/47—Quinolines; Isoquinolines
-
- 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
- A61K31/47—Quinolines; Isoquinolines
- A61K31/472—Non-condensed isoquinolines, e.g. papaverine
-
- 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
- C07D513/00—Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
- C07D513/02—Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
- C07D513/04—Ortho-condensed systems
Definitions
- the present invention relates to the field of medicinal chemistry, and in particular to a class of compounds or pharmaceutically acceptable salts used as PKMYT1 inhibitors, preparation methods thereof, pharmaceutical compositions and use thereof in treating PKMYT1-related diseases.
- PKMYT1 Protein kinase, membrane associated tyrosine/threonine 1 (PKMYT1) belongs to the WEE family of serine/threonine kinases. PKMYT1 specifically regulates the activity of cyclin-dependent kinase 1 (CDK1) by phosphorylating CDK1, thereby regulating the transition from G2 to M phase of the cell cycle. PKMYT1 is not essential for checkpoint-triggered cell cycle arrest following DNA damage in normal cells. Depletion of PKMYT1 by siRNA alone does not affect long-term cell growth, but PKMYT1 plays a more important role in cells with genomic instability or in the presence of replication stress or other impaired cell cycle checkpoint regulation.
- CDK1 cyclin-dependent kinase 1
- CCNE1 gene amplification is a common tumor driver mutation that occurs in a variety of malignancies, including ovarian cancer, breast cancer, gastric cancer, and lung cancer.
- Cyclin E1, encoded by CCNE1 binds to and activates CDK2, promoting the transition from the G1 phase to the S phase of the cell cycle.
- Overexpression of Cyclin E impairs G1/S checkpoint regulation, causing cells to enter the S phase prematurely, increasing DNA replication pressure, and leading to genomic instability.
- cells with CCNE1 amplification are more dependent on the regulation of other cell cycle checkpoints and the integrity of DNA repair mechanisms.
- PKMYT1 kinase inhibition and CCNE1 amplification are synthetically lethal.
- WEE1 which is in the same family as PKMYT1
- PKMYT1 inhibitors and WEE1 inhibitors can target the DNA damage response (DDR) in cancer, inhibit the damage and repair of tumor cell DNA, and promote their apoptosis.
- DDR DNA damage response
- WEE1 inhibitors are being developed for the treatment of tumors. Studies have shown that compared with WEE1, PKMYT1 inhibition has less effect on normal cells, so PKMYT1 inhibitors have better safety. Finding PKMYT1 inhibitors is considered a promising approach to developing new anticancer agents.
- Ring A is selected from a benzene ring, a C 5-6 hydrocarbon ring, a 5-6 membered heterocyclic ring and a 5-6 membered heteroaromatic ring;
- E is selected from O and S;
- R1 is selected from hydrogen, halogen, C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, C3-10 cycloalkyl, -C1-4 alkylene- C3- 10 Cycloalkyl, heterocyclyl, -C 1-4 alkylene-heterocyclyl, CN, NO 2 , aryl, -C 1-4 alkylene-aryl, heteroaryl, -C 1-4 alkylene-heteroaryl, -NR A1 R B1 , -OR A1 , -SR A1 , -C( ⁇ O) R A1 , -C( ⁇ NR E1 ) R A1 , -C( ⁇ N-OR B1 ) R A1 , -C( ⁇ O)OR A1 , -OC( ⁇ O) R A1 , -C( ⁇ O)NR A1 R B1 , -NR A1 C( ⁇ O) R B1 , -C( ⁇ NR
- R 3 together with the atoms to which they are attached form a C 3-10 monocyclic hydrocarbon ring or a 4-12 membered heterocyclic ring containing 1, 2 or 3 heteroatoms or a 5-10 membered heteroaromatic ring or a benzene ring containing 1, 2 or 3 heteroatoms, wherein the heteroatoms are independently selected from oxygen, sulfur, nitrogen and phosphorus, and the ring is unsubstituted or substituted by at least one substituent independently selected from RX3 ;
- R 7 and R 8 together with the atoms to which they are attached form a C 5-6 hydrocarbon ring or a 5-6 membered heterocyclic ring or a 5-6 membered heteroaromatic ring containing 1, 2 or 3 heteroatoms, wherein the heteroatoms are independently selected from oxygen, sulfur and nitrogen, and the ring is unsubstituted or substituted by at least one substituent independently selected from R X7 ;
- each of RA1, RA2 , RA3 , RA4 , RA5 , RA6 , RA7 , RA8 , RB1 , RB2 , RB3 , RB4 , RB5 , RB6 , RB7 and RB8 is independently selected from hydrogen, C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, C3-10 cycloalkyl, -C1-4 alkylene- C3-10 cycloalkyl, heterocyclyl, -C1-4 alkylene - heterocyclyl , aryl, -C1-4 alkylene-aryl, heteroaryl and -C1-4 alkylene-heteroaryl, wherein each of alkyl, alkylene, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one substituent independently selected from RX ;
- RA1 and RB1 or “RA2 and RB2 ” or “ RA3 and RB3 ” or “ RA4 and RB4 ” or “ RA5 and RB5 ” or “ RA6 and RB6 ” or “ RA7 and RB7 ” or “ RA8 and RB8 ” together with the atoms to which they are attached, form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring is unsubstituted or substituted with 1, 2 or 3 substituents selected from RX ;
- Ra1 and Rb1 is independently selected from hydrogen, C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, C3-10 cycloalkyl, -C1-4 alkylene- C3-10 cycloalkyl, heterocyclyl, -C1-4 alkylene-heterocyclyl, aryl, -C1-4 alkylene-aryl, heteroaryl and -C1-4 alkylene-heteroaryl, wherein each of alkyl, alkylene, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and hetero Aryl is unsubstituted or substituted with at least one substituent independently selected from R Y ;
- Ra1 and Rb1 together with the atoms or atoms to which they are attached together form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring is unsubstituted or substituted with 1, 2 or 3 substituents selected from RY ;
- each R c1 and R d1 is independently selected from hydrogen, halogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 3-10 cycloalkyl, -C 1-4 alkylene-C 3-10 cycloalkyl, heterocyclyl, -C 1-4 alkylene-heterocyclyl, aryl, -C 1-4 alkylene-aryl, heteroaryl and -C 1-4 alkylene-heteroaryl, wherein each alkyl, alkylene, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one substituent independently selected from RY ;
- each R c1 and R d1 together with the single or multiple carbon atoms to which they are attached form a 3-12 membered ring containing 0, 1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring is unsubstituted or substituted with 1, 2 or 3 groups independently selected from RY ;
- each R e1 is independently selected from hydrogen, C 1-10 alkyl, CN, NO 2 , —S( ⁇ O) r R a1 , —C( ⁇ O) R a1 , —C( ⁇ O) OR a1 , —C( ⁇ O) NR a1 R b1 , and —S( ⁇ O) r NR a1 R b1 , wherein alkyl is unsubstituted or substituted with at least one substituent independently selected from RY ;
- Each RY is independently selected from halogen, NO2 , -CN, C1-10 alkyl, -OH, -O( C1-10 alkyl), -O( C3-10 cycloalkyl), -O( C1-4 alkylene- C3-10 cycloalkyl), -O(heterocyclyl), -O( C1-4 alkylene-heterocyclyl), -SH, -S( C1-10 alkyl), -S( C3-10 cycloalkyl), -S(C1-4 alkylene- C3-10 cycloalkyl), -S(heterocyclyl), -S( C1-4 alkylene-heterocyclyl), -NH2 , -NH ( C1-10 alkyl), -N( C1-10 alkyl) 2 , -NH( C3-10 cycloalkyl), -NH( C1-4 alkylene- C3-10 cycloalkyl), -NH(heterocycly
- g is selected from 0, 1, 2, 3, 4, 5, 6, 7 and 8;
- Each r is independently selected from 1 and 2;
- Each t is independently selected from 0, 1, 2, 3 and 4.
- a pharmaceutical composition comprising a preventively or therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.
- synthetic lethality refers to the situation where two or more individual genes simultaneously express abnormally, leading to cell death, while only one or a portion of the genes express abnormally, leading to cell death; wherein the abnormal expression includes mutation, overexpression or gene inhibition.
- the two synthetic lethal genes are referred to as a synthetic lethal gene pair.
- cyclin E1 refers to the G1/S specific cyclin E1 (gene name: CCNE1). Cells that overexpress cyclin E1 show higher cyclin E1 activity than cells that normally express cyclin E1. CCNE1-amplified cells are cells that have a higher CCNE1 gene copy number than normal cells. Cells that overexpress cyclin E1 can be CCNE1-amplified cells, for example, in one embodiment, cells that overexpress cyclin E1 have more than 2 copies of CCNE1 compared to diploid normal cells having 2 copies of CCNE1. In addition to directly detecting the copy number of the CCNE1 gene, cyclin E1 overexpression can also be determined by identifying the expression level of the gene product in the cell (e.g., mRNA transcription level or cyclin E1 protein level).
- cancer overexpressing cyclin E1 and "CCNE1 amplified cancer” are used interchangeably.
- CCNE1 amplified cancers include, but are not limited to, uterine sarcoma, ovarian cancer, breast cancer, gastric cancer, esophageal cancer, lung cancer, liver cancer, and endometrial cancer.
- FBXW7 refers to F-box/WD repeat-containing protein 7 (gene name: FBXW7).
- An FBXW7 gene with an inactivating mutation refers to a mutant FBXW7 gene that cannot produce a normal functional FBXW7 protein or produces a reduced amount of FBXW7 protein in a cell.
- FBXW7 mutation cancers include, but are not limited to, blood tumors, gliomas, liver cancer (e.g., hepatocellular carcinoma), uterine cancer (e.g., endometrial carcinoma), colorectal cancer (e.g., colorectal adenocarcinoma), breast cancer, lung cancer (e.g., non-small cell lung cancer (NSCLC)), gastric cancer, esophageal cancer, esophageal junction adenocarcinoma, bladder cancer (e.g., urothelial bladder carcinoma), head and neck cancer (e.g., head and neck squamous cell carcinoma), cervical cancer (e.g., cervical squamous cell carcinoma), melanoma, ovarian cancer (e.g., high-grade serous ovarian cancer).
- liver cancer e.g., hepatocellular carcinoma
- uterine cancer e.g., endometrial carcinoma
- colorectal cancer e.
- p53 protein refers to tumor protein p53 (gene name: TP53).
- a TP53 gene with an inactivating mutation refers to a mutant TP53 gene that cannot produce a functional p53 protein or produces a reduced amount of p53 protein in a cell.
- C1 - C10 or " C1-10” encompasses a range of 1-10 carbon atoms and should be understood to also encompass any subranges and individual point values therein, such as C2-3 , C2-4 , C2-5 , C3-4 , C3-5 , C3-6 , C3-7 , C1-2 , C1-3, C1-4 , C1-5 , C1-6 , C1-7 , C1-8 , C1-9 , etc. , as well as C1 , C2 , C3 , C4 , C5 , C6 , C7 , C8 , C9 , C10 , etc.
- C3 - C10 or " C3-10” should also be understood in a similar manner, for example, it can include C3-4 , C3-5 , C3-6 , C3-7 , C3-8 , C4-5 , C4-6 , C4 - C7 , C5 -C6, etc., as well as C3 , C4, C5 , C6 , C7, C8, C9, C10 , etc.
- the expression “3-14 yuan” should be understood to include any sub-ranges and each point value therein, such as 3-4, 3-5 , 3-6 , 3-7 , 3-8 , 4-5 , 4-6, 4-7, 5-6, 3, 4, 5, 6 or 7 yuan, etc.
- p is an integer selected from 0 to 13
- p is any integer from 0 to 13, for example, p may be 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or 13.
- Other similar expressions such as p1 and p2 should also be understood in a similar manner.
- any variable e.g., R x
- its definition is independent at each occurrence.
- the expression "each R x is independently selected from” means that if multiple R x are contained, the options for each R x substituent are independent of each other at each occurrence.
- Other variables or expressions such as R 3 should also be understood in a similar manner.
- substituted and “substituted” refer to one or more (e.g., one, two, three or four) hydrogen atoms on the designated atom being replaced by a selection from the indicated group, provided that the normal atomic valence of the designated atom in the current situation is not exceeded and the substitution forms a stable compound. Combinations of substituents and/or variables are permitted only if such combinations form stable compounds. When a substituent is described as not present, it should be understood that the substituent can be one or more hydrogen atoms, provided that the structure can achieve a stable state of the compound.
- the point of attachment of a substituent may be from any convenient position of the substituent.
- a bond to a substituent is shown as passing through a bond connecting two atoms in a ring, then such a substituent may be bonded to any ring-forming atom in the substitutable ring.
- halo or "halogen” or “halo” is understood to mean a fluorine (F), chlorine (Cl), bromine (Br) or iodine (I) atom, preferably a fluorine, chlorine or bromine atom.
- hydrocarbyl refers to a monovalent group derived from a hydrocarbon.
- hydrocarbyl groups include, but are not limited to, alkyl, alkenyl, alkynyl, cycloalkyl, and aryl.
- alkyl refers to a saturated aliphatic hydrocarbon group consisting of carbon atoms and hydrogen atoms, which is connected to the rest of the molecule by a single bond.
- Alkyl includes straight chain alkyl and branched chain alkyl.
- Alkyl can contain 1-10 carbon atoms, known as C 1-10 alkyl, such as C 1-6 alkyl, C 1-4 alkyl, C 1-3 alkyl, C 1-2 alkyl, C 3 alkyl, C 4 alkyl, C 3-6 alkyl.
- Non-limiting examples of straight chain alkyl include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, etc.
- Non-limiting examples of branched alkyl groups include, but are not limited to, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, 2-methylbutyl, 1-methylbutyl, 1-ethylpropyl, 1,2-dimethylpropyl, neopentyl, 1,1-dimethylpropyl, 4-methylpentyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 2-ethylbutyl, 1-ethylbutyl, 3,3-dimethylbutyl, 2,2-dimethylbutyl, 1,1-dimethylbutyl, 2,3-dimethylbutyl, 1,3-dimethylbuty
- a divalent group refers to a group obtained by removing a hydrogen atom from a carbon atom with free valence electrons of a corresponding monovalent group.
- a divalent group has two attachment sites connected to the rest of the molecule, wherein the two attachment sites can be located on the same atom or two different atoms of the divalent group.
- Alkylene or “alkylidene” refers to a saturated divalent hydrocarbon group.
- Alkylene includes straight chain or branched chain alkylene. Examples of straight chain alkylene include, but are not limited to, methylene ( -CH2- ), -( CH2 ) 2- , -( CH2 ) 3- , - ( CH2) 4- , -( CH2 ) 5- , -( CH2 ) 6- , and the like.
- branched alkylene groups include, but are not limited to, -CH( CH3 )-, -CH( C2H5 )-, -CH( CH3 ) -CH2- , -CH( C3H7 ) - , -CH ( C2H5 ) -CH2- , -C( CH3 ) 2 - CH2- , -(CH (CH3 ) ) 2- , -CH( CH3 )-(CH2) 2- , -CH2 - CH (CH3 ) -CH2- , -CH(C4H9)-, -C ( CH3 ) ( C3H7 )-, -C(C2H5) 2- , -CH( C3H7 ) -CH2- , -CH ( C2H5 ) -CH( CH3 )-, -CH( C2H5 )-( CH2 ) 2- , and -CH( CH3 ) -CH2- .
- alkenyl refers to a straight or branched unsaturated aliphatic hydrocarbon group having at least one double bond consisting of carbon atoms and hydrogen atoms.
- the alkenyl group may have 2-8 carbon atoms, i.e., " C2-8 alkenyl", such as C2-4 alkenyl, C3-4 alkenyl.
- Non-limiting examples of alkenyl include, but are not limited to, vinyl, allyl, (E)-2-methylvinyl, (Z)-2-methylvinyl, (E)-but-2-enyl, (Z)-but-2-enyl, (E)-but-1-enyl, (Z)-but-1-enyl, etc.
- alkynyl refers to a straight or branched unsaturated aliphatic hydrocarbon group consisting of carbon atoms and hydrogen atoms, having at least one triple bond.
- the alkynyl group may have 2-8 carbon atoms, i.e., " C2-8 alkynyl", such as C2-4 alkynyl, C3-4 alkynyl.
- Non-limiting examples of alkynyl include, but are not limited to, ethynyl, prop-1-ynyl, prop-2-ynyl, but-1-ynyl, but-2-ynyl, but-3-ynyl, etc.
- alkoxy refers to an alkyl group as defined above that is connected to an oxygen atom by a single bond. The alkoxy group is connected to the rest of the molecule through the oxygen atom.
- the alkoxy group can be represented as -O(alkyl).
- C 1-8 alkoxy or “-O(C 1-8 alkyl)” refers to an alkoxy group containing 1 to 8 carbon atoms, wherein the alkyl portion can be a straight chain or branched structure.
- Alkoxy includes, but is not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, n-pentoxy, etc.
- hydrocarbon ring and “cycloalkyl” refer to a saturated or unsaturated non-aromatic ring system consisting of carbon atoms and hydrogen atoms.
- Hydrocarbon ring and “cycloalkyl” preferably contain 1 or 2 rings respectively.
- the “hydrocarbon ring” and “cycloalkyl” can be a monocyclic, condensed polycyclic, bridged or spirocyclic structure.
- Hydrocarbon ring” and “cycloalkyl” can have 3-10 carbon atoms respectively, i.e. "C 3-10 hydrocarbon ring” (e.g.
- C 4 hydrocarbon ring, C 5 hydrocarbon ring, C 6 hydrocarbon ring, C 7 hydrocarbon ring) and "C 3-10 cycloalkyl” e.g. C 3-8 cycloalkyl, C 5 cycloalkyl, C 6 cycloalkyl, C 7 cycloalkyl.
- hydrocarbon rings include but are not limited to cyclopentane, cyclohexane, cycloheptane, cyclopentene, cyclohexene, etc.
- Non-limiting examples of cycloalkyl include, but are not limited to, cyclopropane, cyclobutane, cyclopentane, cyclohexane, cycloheptane, bicyclo[2.2.1]heptane and spiro[3.3]heptane, cyclopentenyl, cyclohexenyl, etc.
- the C atoms in the cycloalkyl are optionally substituted with oxo.
- the C atoms in the cycloalkyl are optionally substituted with imino.
- the imino is substituted with -OR, wherein R is H or C 1-10 alkyl.
- cyclic alkyl and “cycloalkyl” have the same meaning herein and are used interchangeably. Refers to a saturated cycloalkyl group. Cycloalkyl can have 3, 4, 5, 6, 7, 8, 9 or 10 ring carbon atoms (C 3-10 ). Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl. In some embodiments, cycloalkyl is a C 3-7 monocyclic or bicyclic cycloalkyl, preferably a C 3-6 monocyclic cycloalkyl, particularly a cyclopropyl.
- heterocycle and “heterocyclyl” each refer to a monocyclic or bicyclic ring system (3-14-membered, 7-14-membered, 3-8-membered, 3-7-membered, 4-6-membered, 5-6-membered) having, for example, 3-14 ring atoms (e.g., 7-14, 3-8, 3-7, 4-6, or 5-6 ring atoms), wherein at least one ring atom (e.g., 1, 2, or 3) is a heteroatom selected from oxygen, sulfur, nitrogen, and phosphorus, and the remaining ring atoms are C.
- the ring system may be saturated (also understood as the corresponding "heterocycloalkane” or “heterocycloalkyl”) or unsaturated (i.e., having one or more double bonds and/or triple bonds in the ring).
- “heterocycle” and “heterocyclyl” each may be benzo-fused.
- “Heterocycle” and “heterocyclyl” each do not have aromaticity.
- the C, N, S, and P atoms in “heterocycle” and “heterocyclyl” are optionally substituted with oxo.
- the C, S and P atoms in "heterocycle” and “heterocyclyl” are optionally substituted by imino.
- the imino is substituted by -OR, wherein R is H or C 1-10 alkyl.
- the heterocyclic group can be, for example, a four-membered ring, such as azetidinyl, oxetanyl; or a five-membered ring, such as tetrahydrofuranyl, dioxolinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl, oxopyrrolidinyl, 2-oxoimidazolidin-1-yl; or a six-membered ring, such as tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, 1,1-dioxo-1,2-thiazin-2-yl or trithianyl; or a seven-membered ring, such as diazepine, Base ring.
- a four-membered ring such as azetidinyl, oxet
- the heterocyclic group may be bicyclic, without limitation, for example, a five-membered and a five-membered ring, such as octahydrocyclopenta[c]pyrrolyl; or a five-membered and a six-membered bicyclic ring, such as octahydropyrrolo[1,2-b]pyrazinyl.
- the heterocycle may be unsaturated, i.e., it may contain one or more double bonds, without being limited thereto, for example, an unsaturated heterocycle containing a nitrogen atom may be a 1,6-dihydropyrimidine, a 1,2-dihydropyrimidine, a 1,4-dihydropyrimidine, a 1,6-dihydropyridine, a 1,2-dihydropyridine, a 1,4-dihydropyridine, a 2,3-dihydro-1H-pyrrole, a 3,4-dihydro-1H-pyrrole, a 2,5-dihydro-1H-pyrrolyl, a 4H-[1,3,4]thiadiazinyl, a 4,5-dihydrooxazolyl or a 4H-[1,4]thiazinyl ring, an unsaturated heterocycle containing an oxygen atom may be a 2H-pyran, a 4H-pyran, a 2,
- Exemplary bicyclic heterocycles also include:
- aryl refers to an aromatic ring group of an all-carbon monocyclic or fused polycyclic (such as a bicyclic) ring having a conjugated ⁇ electron system.
- an aryl group may have 6-14 carbon atoms, preferably 6-10, more preferably 6 or 10.
- Examples of aryl groups include, but are not limited to, phenyl, naphthyl, anthracenyl, and the like.
- heteroaryl refers to a monocyclic, bicyclic or tricyclic aromatic ring system having 5, 6, 7, 8, 9 or 10 ring atoms ("5-10 membered heteroaryl”), in particular 5 or 6 or 9 or 10 ring atoms, and including at least one (suitably 1 to 4, more suitably 1, 2 or 3) heteroatoms which may be identical or different, such as oxygen, nitrogen or sulfur. Furthermore, in each case “heteroaromatic ring” and “heteroaryl” may each be benzo-fused.
- the heteroaryl group is selected from thienyl, furanyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, and the like, and benzo derivatives thereof, such as benzofuranyl, benzothienyl, benzoxazolyl, benzisoxazolyl, benzimidazolyl, benzotriazolyl, indazolyl, indolyl, isoindolyl, and the like; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, and the like, and benzo derivatives thereof, such as quinolyl, quinazolinyl, isoquinolyl, and the like; or azocinyl, indolizinyl,
- the pharmaceutically acceptable salts of the compounds of the present invention include acid addition salts and base addition salts thereof.
- the methods for preparing the pharmaceutically acceptable salts of the compounds of the present invention are known to those skilled in the art.
- the compounds of the present invention encompass pharmaceutically acceptable salts, stereoisomers, solvates, polymorphs, tautomers, isotopic compounds, metabolites or prodrugs thereof.
- the compounds of the present invention may exist in specific geometric or stereoisomeric forms. All such compounds are contemplated by the present invention.
- the compounds of the present invention may exist in E or Z configurations of carbon-carbon double bonds or carbon-nitrogen double bonds, wherein “E” represents the preferred substituents on the opposite side of the carbon-carbon double bond or carbon-nitrogen double bond according to the Cahn-Ingold-Prelog priority rules, and "Z” represents the preferred substituents on the same side of the carbon-carbon double bond or carbon-nitrogen double bond.
- the compounds of the present invention may also exist in the form of mixtures of "E" and "Z" isomers.
- Isomer forms also include cis and trans isomers, (-)- and (+)-enantiomers, (R)- and (S)-enantiomers, diastereomers, (D)-isomers, (L)-isomers, and racemic mixtures and other mixtures thereof, such as enantiomerically or diastereomerically enriched mixtures, all of which are within the scope of the present invention. Purification and separation of such materials can be achieved by standard techniques known in the art.
- the compounds of the present invention may contain a bond with hindered rotation, so that two separate torsional isomers can be separated. All possible torsional isomers are included within the scope of the present invention.
- the separation method of torsional isomers can be a known method or a method described herein.
- chiral chromatography is used to separate torsional isomers, for example, using supercritical CO2 and MeOH as mobile phases.
- Optically pure enantiomers can be obtained by resolving racemic mixtures according to conventional methods, for example, by forming diastereomeric salts using optically active acids or bases, or by forming covalent diastereomers.
- a mixture of diastereoisomers can be separated into single diastereomers based on their physical and/or chemical differences by methods known in the art (e.g., by chromatography or fractional crystallization). Then, the optically active enantiomer base or acid is released from the separated diastereomeric salts.
- Another method for separating racemic enantiomers can use chiral chromatography (e.g., a chiral HPLC column), and the separated chiral isomers can be subjected to conventional derivatization treatment or non-derivatization before separation, depending on which method can achieve more effective separation of chiral isomers.
- Enzymatic methods can also be used to separate derivatized or non-derivatized chiral isomers.
- the present invention can be obtained by chiral synthesis using optically active raw materials. Optically pure compounds invented.
- the compounds of the present invention may exist in the form of solvates (preferably hydrates), wherein the compounds of the present invention contain a solvent as a structural element of the crystal lattice of the compound, in particular water, methanol or ethanol.
- a solvent as a structural element of the crystal lattice of the compound, in particular water, methanol or ethanol.
- the amount of the solvent, in particular water may exist in a stoichiometric or non-stoichiometric ratio.
- the present invention also encompasses all possible crystalline forms or polymorphs of the compounds of the present invention, which may be a single polymorph or a mixture of more than one polymorph in any ratio.
- the compounds of the invention may exist in isotopically labeled or enriched form, containing one or more atoms having a different atomic mass and mass number from the most common atomic mass in nature.
- Isotopes may be radioactive or non-radioactive isotopes.
- Isotopes of atoms such as hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine and iodine include, but are not limited to, 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 32 P, 35 S, 18 F, 36 Cl and 125 I.
- metabolites of the compounds of the present invention i.e., substances formed in vivo when the compounds of the present invention are administered.
- Such products may be produced, for example, by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymolysis, etc. of the administered compounds.
- the present invention further includes within its scope prodrugs of the compounds of the invention, which are certain derivatives of the compounds of the invention that may themselves have little or no pharmacological activity but are converted, for example by hydrolytic cleavage, into compounds of the invention having the desired activity when administered into or onto the body.
- polymorph or “polymorph” refers to a single polymorph or a mixture of more than one polymorph in any proportion.
- crystalline form or “crystal” refers to any solid material that exhibits a three-dimensional ordering, in contrast to amorphous solid material, which produces a characteristic X-ray powder diffraction pattern with well-defined peaks.
- amorphous refers to any solid material that has no order in three dimensions.
- pharmaceutically acceptable means that it is within the scope of normal medical judgment and will not cause undue toxicity, irritation, allergic response, or the like in contact with the tissues of patients.
- pharmaceutically acceptable carrier refers to those substances that have no significant irritation to organisms and do not impair the biological activity and performance of the active compound.
- “Pharmaceutically acceptable carrier” includes, but is not limited to, glidants, sweeteners, diluents, preservatives, dyes/colorants, flavoring agents, surfactants, wetting agents, dispersants, disintegrants, stabilizers, solvents or emulsifiers.
- active ingredient refers to a chemical entity that is effective in treating or preventing a target disorder, disease, or condition.
- the term "effective amount”, “therapeutically effective amount” or “prophylactically effective amount” refers to a sufficient amount of the drug or pharmaceutical agent that can achieve the desired effect with acceptable side effects.
- the determination of the effective amount varies from person to person, depending on the age and general condition of the individual and on the specific active substance. The appropriate effective amount in each case can be determined by a person skilled in the art based on routine experiments.
- subject includes humans or non-human animals.
- exemplary human subjects include human subjects (referred to as patients) suffering from diseases (e.g., diseases described herein) or normal subjects.
- Non-human animals in the present invention include all vertebrates, such as non-mammals (e.g., birds, amphibians, reptiles) and mammals, such as non-human primates, livestock and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
- the present invention provides a compound having the structure of formula (I):
- Ring A is selected from a benzene ring, a C 5-6 hydrocarbon ring, a 5-6 membered heterocyclic ring and a 5-6 membered heteroaromatic ring;
- E is selected from O and S;
- R 3 together with the atoms to which they are attached form a C 3-10 monocyclic hydrocarbon ring or a 4-12 membered heterocyclic ring containing 1, 2 or 3 heteroatoms or a 5-10 membered heteroaromatic ring or a benzene ring containing 1, 2 or 3 heteroatoms, wherein the heteroatoms are independently selected from oxygen, sulfur, nitrogen and phosphorus, and the ring is unsubstituted or substituted by at least one substituent independently selected from RX3 ;
- R 7 and R 8 together with the atoms to which they are attached form a C 5-6 hydrocarbon ring or a 5-6 membered heterocyclic ring or a 5-6 membered heteroaromatic ring containing 1, 2 or 3 heteroatoms, wherein the heteroatoms are independently selected from oxygen, sulfur and nitrogen, and the ring is unsubstituted or substituted by at least one substituent independently selected from RX7 ;
- each of RA1, RA2 , RA3 , RA4 , RA5 , RA6 , RA7 , RA8 , RB1 , RB2 , RB3 , RB4 , RB5 , RB6 , RB7 and RB8 is independently selected from hydrogen, C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, C3-10 cycloalkyl, -C1-4 alkylene- C3-10 cycloalkyl, heterocyclyl, -C1-4 alkylene - heterocyclyl , aryl, -C1-4 alkylene-aryl, heteroaryl and -C1-4 alkylene-heteroaryl, wherein each of alkyl, alkylene, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one substituent independently selected from RX ;
- RA1 and RB1 or “RA2 and RB2 ” or “ RA3 and RB3 ” or “ RA4 and RB4 ” or “ RA5 and RB5 ” or “ RA6 and RB6 ” or “ RA7 and RB7 ” or “ RA8 and RB8 ” together with the atoms to which they are attached, form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring is unsubstituted or substituted with 1, 2 or 3 substituents selected from RX ;
- each of Ra1 and Rb1 is independently selected from hydrogen, C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, C3-10 cycloalkyl, -C1-4 alkylene- C3-10 cycloalkyl, heterocyclyl, -C1-4 alkylene-heterocyclyl, aryl, -C1-4 alkylene-aryl, heteroaryl and -C1-4 alkylene-heteroaryl, wherein each of alkyl, alkylene, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one substituent independently selected from RY ;
- Ra1 and Rb1 together with the atoms or atoms to which they are attached together form a 4-12 membered heterocyclic ring containing 0, 1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring is unsubstituted or substituted with 1, 2 or 3 substituents selected from RY ;
- each R c1 and R d1 is independently selected from hydrogen, halogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, C 3-10 cycloalkyl, -C 1-4 alkylene-C 3-10 cycloalkyl, heterocyclyl, -C 1-4 alkylene-heterocyclyl, aryl, -C 1-4 alkylene-aryl, heteroaryl and -C 1-4 alkylene-heteroaryl, wherein each alkyl, alkylene, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one substituent independently selected from RY ;
- each R c1 and R d1 together with the single or multiple carbon atoms to which they are attached form a 3-12 membered ring containing 0, 1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring is unsubstituted or substituted with 1, 2 or 3 groups independently selected from RY ;
- each R e1 is independently selected from hydrogen, C 1-10 alkyl, CN, NO 2 , —S( ⁇ O) r R a1 , —C( ⁇ O) R a1 , —C( ⁇ O) OR a1 , —C( ⁇ O) NR a1 R b1 , and —S( ⁇ O) r NR a1 R b1 , wherein alkyl is unsubstituted or substituted with at least one substituent independently selected from RY ;
- Each RY is independently selected from halogen, NO2 , -CN, C1-10 alkyl, -OH, -O( C1-10 alkyl), -O( C3-10 cycloalkyl), -O( C1-4 alkylene- C3-10 cycloalkyl), -O(heterocyclyl), -O( C1-4 alkylene-heterocyclyl), -SH, -S( C1-10 alkyl), -S( C3-10 cycloalkyl), -S(C1-4 alkylene- C3-10 cycloalkyl), -S(heterocyclyl), -S( C1-4 alkylene-heterocyclyl), -NH2 , -NH ( C1-10 alkyl), -N( C1-10 alkyl) 2 , -NH( C3-10 cycloalkyl), -NH( C1-4 alkylene- C3-10 cycloalkyl), -NH(heterocycly
- g is selected from 0, 1, 2, 3, 4, 5, 6, 7 and 8;
- Each r is independently selected from 1 and 2;
- Each t is independently selected from 0, 1, 2, 3 and 4.
- t is 0. In one embodiment, g is selected from 0, 1 and 2. In one embodiment, E is O. In one embodiment, E is S.
- each of RA1 , RA2 , RA3 , RA4 , RA5 , RA6 , RA7 , RA8, RB1 , RB2 , RB3 , RB4 , RB5 , RB6 , RB7 and RB8 is independently selected from hydrogen and C1-10 alkyl , wherein each alkyl is unsubstituted or substituted with at least one substituent independently selected from RX .
- it is selected from halogen, C 1-10 alkyl, CN, NO 2 , -(CR c1 R d1 ) t NR a1 R b1 , -(CR c1 R d1 ) t OR b1 , -(CR c1 R d1 ) t C( ⁇ O)R a1 , -(CR c1 R d1 ) t C( ⁇ O)OR b1 , -(CR c1 R d1 ) t OC( ⁇ O)R b1 , -(CR c1 R d1 ) t C( ⁇ O)NR a1 R b1 and -(CR c1 R d1 ) t NR a1 C( ⁇ O)R b1 , wherein each alkyl, alkylene, cycloalkyl and heterocyclyl is unsubstituted or substituted by at least one substituent independently selected from RY
- each RX , RX1 , RX2 , RX4 , RX5 , RX6 , RX7 and RX8 is independently selected from halogen, C 1-10 alkyl, CN and NO 2 , wherein each alkyl is unsubstituted or substituted with at least one substituent independently selected from RY .
- each R a1 and R b1 is independently selected from hydrogen and C 1-10 alkyl, wherein each alkyl is unsubstituted or substituted with at least one substituent independently selected from RY .
- each R c1 and R d1 is independently selected from hydrogen, halogen and C 1-10 alkyl, wherein each alkyl is unsubstituted or substituted with at least one substituent independently selected from RY .
- each RY is independently selected from halogen, NO2 , -CN, C1-10 alkyl, -OH, -O ( C1-10 alkyl), -NH2 , -NH( C1-10 alkyl) and -N( C1-10 alkyl) 2 , preferably selected from halogen, NO2 , -CN and C1-10 alkyl.
- R 1 is selected from C 1-10 alkyl, -NR A1 R B1 , -OR A1 and -SR A1 , preferably selected from -NR A1 R B1 and -OR A1 , in particular -NR A1 R B1 .
- each R B1 is independently selected from C 1-10 alkyl
- each R A1 is hydrogen.
- each R A1 and R B1 are hydrogen.
- each R E1 is independently selected from hydrogen and C 1-10 alkyl.
- R 1 is -NH 2.
- R 1 is selected from -NR A1 R B1 and hydrogen, preferably selected from -NH 2 and hydrogen.
- each RB2 is independently selected from C 1-10 alkyl
- each RA2 is hydrogen.
- each RA2 and RB2 are hydrogen.
- each RE2 is independently selected from hydrogen and C 1-10 alkyl.
- any two adjacent R3 together with the atoms to which they are attached form a C5-6 monocyclic hydrocarbon ring or a 5-6 membered heterocyclic ring containing 1, 2 or 3 heteroatoms or a 5 membered heterocyclic ring containing 1, 2 or 3 heteroatoms or a 6 membered heteroaromatic ring containing 1 or 2 heteroatoms, wherein the heteroatoms are independently selected from oxygen, sulfur, nitrogen and phosphorus, preferably selected from oxygen, sulfur and nitrogen; the ring is unsubstituted or substituted by at least one substituent independently selected from RX3 .
- g is 1 or 2.
- halogen is selected from F, Cl and Br.
- each RB3 is independently selected from C1-10 alkyl
- each RA3 is hydrogen.
- each RA3 and RB3 are hydrogen.
- each RE3 is independently selected from hydrogen and C1-10 alkyl.
- g is 2, and two R3 are adjacent, and two R3 together with the atoms to which they are attached form cyclohexene or cyclohexane, wherein the cyclohexene or cyclohexane is unsubstituted or substituted by at least one substituent independently selected from RX3 .
- each R 3 is independently selected from F, Cl, Br, CN, OH, methyl, ethyl, isopropyl, tert-butyl, cyclopropane, azetidinyl, cyclohexenyl, piperidinyl, morpholinyl, oxopiperazinyl, pyridinyl, thiazolyl, pyrazolyl, diethylamino, amino, vinyl, ethynyl, acetyl, carbamoyl, methoxy, ethoxy, methylthio, ethylthio, isopropylthio, methylsulfonyl and phenyl, wherein the methyl, ethyl, isopropyl, Tert-butyl, cyclopropane, azetidinyl, cyclohexenyl, piperidinyl, morpholinyl, oxopiperazinyl
- each RX is independently selected from F, Cl, Br, CN, OH, C1-10 alkyl, -O( C1-10 alkyl) and heterocyclyl, wherein the C1-10 alkyl is unsubstituted or substituted with at least one substituent independently selected from halogen.
- each R 3 is independently selected from F, Cl, Br, CN, OH, methyl, ethyl, isopropyl, tert-butyl, cyclopropane, diethylamino, amino, vinyl, ethynyl, acetyl, carbamoyl, methoxy, methylthio, ethylthio, isopropylthio, methylsulfonyl, phenyl,
- R is methyl.
- R is methyl or ethyl.
- R is methyl or methylthio.
- R is methylthio.
- R is methyl or cyclopropane.
- R is methyl or isopropyl. In one embodiment, R is methyl or Cl. In one embodiment, R is methyl or trifluoromethyl. In one embodiment, R is methyl or isopropyl. In one embodiment, R is isopropyl. In one embodiment, R is tert-butyl.
- any two adjacent R 3 together with the atoms to which they are attached form a ring selected from the group consisting of cyclohexene, cyclohexane, dihydropyran, tetrahydropyran, cyclohexenone, cyclohexanone, dihydropyridone, piperidone, tetrahydropyridine, piperidine, cyclopentene, cyclopentane, dihydrothiophene, tetrahydrothiophene, pyridine and dihydropyridine, which ring is unsubstituted or substituted with at least one substituent independently selected from RX 3.
- any two adjacent R 3 together with the atoms to which they are attached form a ring selected from the group consisting of:
- the ring is unsubstituted or substituted with at least one substituent independently selected from RX3 ; wherein represents the site of attachment of R 3 to Ring A.
- any two adjacent R 3 together with the atoms to which they are attached form The ring is unsubstituted or substituted with at least one substituent independently selected from RX3 ; wherein
- RX3 is C1-10 alkyl, especially methyl.
- RX3 is selected from -( CRc1Rd1 ) tNRa1Rb1 ; wherein Ra1 is selected from In one embodiment, any two adjacent R3 together with the atoms to which they are attached form a structure selected from the following:
- R is selected from C 1-10 alkyl, preferably methyl or ethyl , wherein each alkyl is unsubstituted or substituted by at least one substituent independently selected from RX4. In one embodiment, R is selected from C 1-10 alkyl . In a particular embodiment, R is methyl. In one embodiment, R is selected from halogen , especially Cl .
- R 5 is selected from hydrogen, halogen, C 1-10 alkyl, CN, NO 2 , -NR A5 R B5 and -OR A5 , in particular selected from hydrogen, halogen, C 1-10 alkyl, CN and NO 2 , wherein each alkyl group is unsubstituted or substituted with at least one substituent independently selected from RX5 .
- R 5 is selected from halogen, in particular F.
- R 5 is hydrogen.
- R 6 is selected from hydrogen, halogen, C 1-10 alkyl, CN, NO 2 , -NR A6 RB6 and -OR A6 , wherein each alkyl group is unsubstituted or substituted by at least one substituent independently selected from RX6 .
- R 6 is hydrogen.
- R 6 is F.
- R 7 is selected from halogen, C 1-10 alkyl, CN, NO 2 , -NR A7 RB7 and -OR A7 , wherein each alkyl group is unsubstituted or substituted by at least one substituent independently selected from RX7 .
- R 7 is selected from -NR A7 RB7 and -OR A7 , preferably -OR A7 .
- R 7 is -OH.
- R 7 and R 8 together with the atoms to which they are attached form a C 5-6 hydrocarbon ring or a 5-6 membered heterocyclic ring or a 5-6 membered heteroaromatic ring containing 1, 2 or 3 heteroatoms, wherein the heteroatoms are independently selected from oxygen, sulfur and nitrogen, and the ring is unsubstituted or substituted with at least one substituent independently selected from RX3 .
- R 7 and R 8 together with the atoms to which they are attached form a C 5 hydrocarbon ring or a 5-membered heterocyclic ring or a 5-membered heteroaromatic ring containing 1, 2 or 3 heteroatoms, preferably a 5-membered heterocyclic ring or a 5-membered heteroaromatic ring containing 1, 2 or 3 heteroatoms, wherein the heteroatoms are independently selected from oxygen, sulfur and nitrogen, and the ring is unsubstituted or substituted with at least one substituent independently selected from RX7 .
- R7 and R8 together with the atoms to which they are attached form a pyrazole ring, an isothiazole ring, a thiazole ring, a dihydroisoxazole ring or a dihydrooxazole ring, preferably a pyrazole ring or an isothiazole ring, in particular a pyrazole ring; the ring is unsubstituted or substituted by at least one substituent independently selected from RX7 .
- in formula (I) The structure is In one embodiment, in formula (I) The structure is selected from in particular In one embodiment, in formula (I) The structure is
- R 7 and R 8 together with the atoms to which they are attached form a ring, and formula (I) has the structure of formula (I-1):
- X 1 is selected from N(R 9a ), N, O, S, C(R 9a R 9a' ) and C(R 9a );
- X 2 is selected from N(R 9b ), N, O, S, C(R 9b R 9b' ) and C(R 9b );
- X 3 is selected from N(R 9c ), N, O, S, C(R 9c R 9c ') and C(R 9c );
- R 9a , R 9a′ , R 9b , R 9b ′, R 9c and R 9c′ are each independently selected from hydrogen and R X ;
- Ring A, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 and E are as defined in formula (I).
- X 1 is selected from S and N(R 9a ), preferably NH or N(CH 3 ), in particular NH.
- X 2 is selected from N and O.
- X 3 is selected from C(R 9c R 9c′ ) and C(R 9c ).
- X 1 is N(R 9a ), preferably NH or N(CH 3 ), especially NH; and X 2 is N.
- R 9c is selected from hydrogen and C 1-10 alkyl and R 9c′ , if applicable, is selected from hydrogen and C 1-10 alkyl; preferably, R 9c is hydrogen and R 9c′ , if applicable, is hydrogen.
- ring A is selected from a benzene ring, a C5-6 hydrocarbon ring and a 5-6 membered heteroaromatic ring. In one embodiment, ring A is selected from thiophene, thiazole, pyrazole, imidazole, pyrrole, furan, a benzene ring, pyridine, cyclohexene and cyclopentene, which are substituted by g substituents independently selected from R 3 .
- ring A is selected from a 5-membered heteroaromatic ring and cyclopentene, preferably selected from thiophene, thiazole, pyrazole, imidazole, pyrrole, furan and cyclopentene, which is substituted by g substituents independently selected from R 3.
- ring A is thiophene, which is substituted by g substituents independently selected from R 3.
- ring A is thiazole, which is substituted by g substituents independently selected from R 3.
- ring A is pyrazole, which is substituted by g substituents independently selected from R 3.
- ring A is imidazole, which is substituted by g substituents independently selected from R 3.
- ring A is pyrrole, which is substituted by g substituents independently selected from R 3.
- ring A is furan, which is substituted by g substituents independently selected from R 3 .
- ring A is selected from a 5-membered heteroaromatic ring and cyclopentene, and formula (I) has the structure of formula (II) as follows:
- a 1 is selected from N(R 3a ), N, O, S, C(R 3a R 3a' ) and C(R 3a );
- a 2 is selected from N(R 3b ), N, O, S, C(R 3b R 3b' ) and C(R 3b );
- a 3 is selected from N(R 3c ), N, O, S, C(R 3c R 3c′ ) and C(R 3c );
- R 3a , R 3a′ , R 3b , R 3b′ , R 3c and R 3c′ are each independently selected from hydrogen and R 3 ;
- R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 and E are as defined in formula (I).
- Ring A is selected from a 5-membered heteroaromatic ring.
- ring A is selected from thiophene, thiazole and imidazole, wherein A2 is C( R3b ); A1 is selected from N( R3a ), N, S and C( R3a ) and A3 is selected from N( R3c ), N, S and C( R3c ). In one embodiment, ring A is thiophene, wherein A2 is C( R3b ); A1 is C( R3a ) and A3 is S, or A1 is S and A3 is C( R3c ). In one embodiment, ring A is thiazole, A2 is C( R3b ); A1 is N and A3 is S, or A1 is S and A3 is N.
- ring A is imidazole, A2 is C( R3b ); A1 is N and A3 is N. In one embodiment, Ring A is cyclopentene, A1 is C( R3a ), A2 is C( R3b ), and A3 is C( R3c ).
- R3a and R3c are each hydrogen.
- Ring A is thiophene
- R 3b is selected from halogen, C 1-10 alkyl and C 3-10 cycloalkyl
- R 3a and R 3c are each independently selected from C 1-10 alkyl; wherein each alkyl, cycloalkyl is unsubstituted or substituted with at least one substituent independently selected from RX3 .
- Ring A is pyrazole, wherein A 2 is N; A 1 is N(R 3a ) and A 3 is C(R 3c ), or A 1 is C(R 3a ) and A 3 is N(R 3c ).
- R 3a and R 3c are each independently selected from hydrogen and C 1-10 alkyl; wherein each alkyl is unsubstituted or substituted by at least one substituent independently selected from RX3 .
- one of R3a and R3c is selected from C1-10 alkyl and the other is hydrogen.
- R3a and R3c are each independently selected from C1-10 alkyl.
- R 7 and R 8 together with the atoms to which they are attached form a ring, and formula (II) has the structure of formula (II-1):
- X 1 , X 2 , and X 3 are as defined in formula (I-1);
- a 1 , A 2 , A 3 , R 1 , R 2 , R 4 , R 5 , R 6 , and E are as defined in formula (II).
- Ring A, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , and E of formula (II-1) are as defined in formula (I).
- A1 is selected from N( R3a ), C( R3aR3a ') and C( R3a ),
- A2 is selected from N(R3b), C(R3bR3b') and C(R3b), and R3a or R3a ' and R3b or R3b ' together with the atoms to which they are attached form a ring Q, and formula (II) has the structure of the following formula (II-2):
- Y is selected from N and C;
- Y2 is selected from N and C;
- Ring Q is a C 3-10 monocyclic hydrocarbon ring or a 4-12 membered heterocyclic ring containing 1, 2 or 3 heteroatoms or a 5-10 membered heteroaromatic ring containing 1, 2 or 3 heteroatoms; wherein the heteroatoms are independently selected from oxygen, sulfur, nitrogen and phosphorus, preferably selected from oxygen, sulfur and nitrogen;
- h is selected from 0, 1, 2, 3, 4, 5 and 6;
- a 3 , R 1 , R 2 , R 4 , R 5 , R 6 , R 7 , R 8 , RX3 , and E are as defined in formula (II).
- ring Q is a C 5-6 monocyclic hydrocarbon ring. In one embodiment, ring Q is a 5-6 membered heterocyclic ring containing 1, 2 or 3 heteroatoms. In one embodiment, ring Q is a 5 membered heterocyclic ring containing 1, 2 or 3 heteroatoms. In one embodiment, ring Q is a 6 membered heteroaromatic ring containing 1 or 2 heteroatoms.
- ring Q is selected from in Indicates the site where loop Q is connected to loop A.
- ring A is selected from a benzene ring, a cyclohexene and a 6-membered heteroaromatic ring, which is substituted by g substituents independently selected from R 3.
- the 6-membered heteroaromatic ring contains 1 to 3 N atoms, and the other ring atoms are carbon atoms.
- ring A is selected from a benzene ring, pyridine and cyclohexene, especially a benzene ring.
- ring A is selected from a benzene ring, a cyclohexene ring and a 6-membered heteroaromatic ring, and formula (I) has the structure of formula (III):
- a 4 is selected from N, C(R 3d R 3d′ ) and C(R 3d );
- A6 is selected from N, C( R3fR3f ' ) and C( R3f );
- ring Q is a C 5-6 monocyclic hydrocarbon ring. In one embodiment, ring Q is a 5-6 membered heterocyclic ring containing 1, 2 or 3 heteroatoms. In one embodiment, ring Q is a 5 membered heterocyclic ring containing 1, 2 or 3 heteroatoms. In one embodiment, ring Q is a 6 membered heteroaromatic ring containing 1 or 2 heteroatoms. In one embodiment, ring Q is selected from
- formula (I) has the structure of formula (Ia) or formula (Ib).
- formula (I-1) has the structure of formula (Ia-1) or formula (Ib-1).
- formula (II) has the structure of formula (IIa) or formula (IIb).
- formula (II-1) has the structure of formula (IIa-1) or formula (IIb-1).
- formula (III) has the structure of formula (IIIa) or formula (IIIb).
- the compound of the present invention has a structure of formula (Ia), (Ia-1), (IIa), (IIa-1), (IIIa) or (IIIa-1).
- the compound of the present invention has a structure of formula (Ib), (Ib-1), (IIb), (IIb-1), (IIIb) or (IIIb-1).
- the present invention provides a compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
- the present invention provides a compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
- the present invention provides a compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
- the present invention provides an antibody-drug conjugate comprising a compound of the present invention, an antibody capable of binding to a target, and a linker connecting the compound of the present invention and the antibody.
- Another object of the present invention is to provide a pharmaceutical composition
- a pharmaceutical composition comprising a compound of the present invention or a pharmaceutically acceptable salt, stereoisomer, solvate, polymorph, tautomer, isotope compound, metabolite or prodrug thereof, and at least one pharmaceutically acceptable carrier.
- the pharmaceutical composition of the present invention can be administered in any manner as long as it achieves the effect of preventing, alleviating, preventing or treating human or animal symptoms.
- various suitable dosage forms can be prepared according to the route of administration.
- it can be administered orally or parenterally to a patient in the form of a conventional preparation.
- the conventional preparations are such as capsules, microcapsules, tablets, granules, powders, lozenges, pills, suppositories, injections, suspensions, syrups, patches, creams, lotions, ointments, gels, sprays, solutions and emulsions.
- the dosage of the compound administered to a subject can be adjusted to a considerable extent.
- the dosage can vary depending on the specific route of administration and the needs of the subject, and can be subject to the judgment of a healthcare professional.
- the present invention or its pharmaceutically acceptable salts, stereoisomers, solvates, polymorphs, tautomers, isotopic compounds, metabolites or prodrugs or pharmaceutical compositions of the present invention can be used to prevent or treat PKMYT1-mediated diseases, or diseases responsive to the inhibition of PKMYT1, including but not limited to abnormal cell proliferation diseases, such as cancer.
- the present invention provides a compound of the present invention or a pharmaceutically acceptable salt thereof or a pharmaceutical composition of the present invention, optionally in combination with a second therapeutic agent, for use in treating a cell proliferation disorder.
- the disease, disorder or condition is selected from a cancerous proliferative disease (eg, cancer).
- a cancerous proliferative disease eg, cancer
- the disease, disorder or condition is cancer, for example selected from: (a) a solid tumor or a blood-borne tumor selected from the following cancers: bladder cancer, endometrial cancer, squamous cell lung cancer, breast cancer, colon cancer, kidney cancer, liver cancer, lung cancer, small cell lung cancer, esophageal cancer, gallbladder cancer, brain cancer, head and neck cancer, ovarian cancer, pancreatic cancer, gastric cancer, cervical cancer, thyroid cancer, prostate cancer and skin cancer; (b) a hematopoietic tumor of the lymphoid lineage selected from the following: leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, Hodgkin's lymphoma, (c) a hematopoietic neoplasm of the myeloid lineage selected from the group consisting of acute and chronic myeloid leukemias, myelodysplastic syndromes and promye
- the disease, disorder or condition is selected from CCNE1 amplified cancer. In one embodiment, the disease, disorder or condition is selected from uterine sarcoma, ovarian cancer, breast cancer, gastric cancer, esophageal cancer, lung cancer, liver cancer and endometrial cancer.
- the disease, disorder or condition is selected from FBXW7 mutant cancer, such as a cancer with an inactivating mutation in the FBXW7 gene.
- the disease, disorder or condition is selected from a blood tumor, a glioma, a liver cancer (e.g., hepatocellular carcinoma), a uterine cancer (e.g., endometrial carcinoma), a colorectal cancer (e.g., colorectal adenocarcinoma), a breast cancer, a lung cancer (e.g., non-small cell lung cancer (NSCLC)), a gastric cancer, an esophageal cancer, an esophageal junction adenocarcinoma, a bladder cancer (e.g., urothelial bladder carcinoma), a head and neck cancer (e.g., head and neck squamous cell carcinoma), a cervical cancer (e.g., cervical squamous cell carcinoma), a melanoma,
- the disease, disorder or condition is selected from a TP53 mutant cancer, such as a cancer in which the TP53 gene has an inactivating mutation.
- the disease, disorder or condition is breast cancer or ovarian cancer.
- the compound of the present invention or its pharmaceutically acceptable salt or the pharmaceutical composition of the present invention can be used alone or in combination with other therapeutic agents.
- an adjuvant can enhance the therapeutic effect of the compounds of the present invention (for example, the therapeutic benefit of using an adjuvant alone is minimal, but when used in combination with another drug, the therapeutic benefit of the individual can be enhanced), or, for example, the compound of the present invention combined with another therapeutic agent that also has therapeutic effects can enhance the therapeutic benefit of the individual.
- the combined use of another drug for treating cancer may enhance the clinical benefit.
- Therapies that can be combined include but are not limited to physical therapy, psychotherapy, radiotherapy, chemotherapeutic agents, small molecule targeted therapeutic agents (such as kinase inhibitors, immune agonists), immunotherapy (anti-PD-1, anti-PDL-1, CAR-T cells), etc. Regardless of the disease, disorder or condition, the two therapies should have an additive effect or a synergistic effect to benefit the individual's treatment.
- the other therapeutic agent is selected from inhibitors of WEE family members, such as WEE1 inhibitors.
- the other therapeutic agent is selected from cancer therapeutic agents targeting DNA, such as those that directly destroy Therapeutic agents for DNA structure or DNA topoisomerase inhibitors. Examples of therapeutic agents that directly damage DNA structure include, but are not limited to, DNA double strand breakers, DNA alkylating agents, and DNA intercalators.
- the additional therapeutic agent is selected from a WEE1 inhibitor, a FEN1 (flap-specific endonuclease 1) inhibitor, a TOP1 (DNA topoisomerase I) inhibitor, a RRM1 (ribonucleotide reductase catalytic subunit M1) inhibitor, a RRM2 (ribonucleotide reductase regulatory subunit M2) inhibitor, an AURKB (aurora kinase B) inhibitor, a TOP2A (DNA topoisomerase II ⁇ ) inhibitor, an ATR (ataxia telangiectasia mutated and RAD-3-related protein kinase) inhibitor, a TTK (TTK protein kinase) inhibitor, a SOD1 (superoxide dismutase 1) inhibitor, a SOD2 (superoxide dismutase 2) inhibitor, a BUB1 (BUB1 mitotic checkpoint serine/threonine kinase B) inhibitor, a CDC7 (
- novel small molecule PKMYT1 inhibitor provided by the present invention shows outstanding PKMYT1 inhibitory activity and cell proliferation inhibitory activity.
- the compounds of the present invention can prevent or treat diseases mediated by PKMYT1, or diseases responsive to the inhibition of PKMYT1, including but not limited to abnormal cell proliferation diseases such as cancer, and have good prospects for development as drugs.
- the inventors also unexpectedly found that the torsional isomers of at least a portion of the compounds of the present invention can be obtained by chromatography. Some of the torsional isomers separated show differences in activity. By obtaining torsional isomers, the activity of the compounds of the present invention is further improved, and the above-mentioned excellent properties can be achieved.
- LCMS SHIMADZ LCMS2020, chromatographic column: Waters SunFire TM C18 5 ⁇ m 50*4.6 mm; mobile phase A: H 2 O (containing 0.1% formic acid); mobile phase B: CH 3 CN (containing 0.1% formic acid).
- the compound of formula (I) or its pharmaceutically acceptable salt can be synthesized by different methods, some exemplary methods are provided below and in the Examples. Other synthetic methods can be easily proposed by those skilled in the art based on the information disclosed in the present invention.
- the compound of formula (I) can be prepared and synthesized by various methods from an amide or thioamide compound M1 known in the literature or well known to those skilled in the art and an active methylene reagent M2.
- w1 in M1 is a leaving group, for example, selected from halogen, OTf, in particular halogen.
- M1 and M2 are subjected to a substitution reaction or a coupling reaction to obtain an intermediate M3, and the intermediate M3 is subjected to a condensation reaction to obtain a compound of formula (I).
- the coupling reaction is an Ullmann reaction or a Buchwald reaction.
- the active methylene reagent M2 is malononitrile
- the condensation reaction is, for example, an intramolecular Thorpe-Ziegler cyclization.
- the condensation reaction is carried out in the presence of a palladium reagent, for example, Pd(dppf) Cl2 can be used.
- the condensation reaction is carried out under heating conditions.
- the following process is completed in one synthesis step: M1 and M2 react to obtain M3, and M3 is converted into a compound of formula (I).
- the synthesis step is carried out in the presence of a base, such as sodium alkoxide or sodium hydride, in particular sodium hydride.
- the synthesis step is carried out in an ether solvent, such as ethylene glycol dimethyl ether.
- the amide or thioamide compound M1 as the starting material can be prepared by a known method, for example, by using the corresponding carboxyl compound and amino compound to obtain the target amide through acid-amine condensation, or by using the corresponding amide The compound is subjected to thiolation reaction to obtain the target thioamide.
- Some substituents of the compound of formula (I) can be converted to groups by known methods.
- M2 is malononitrile and the compound of formula (I) is prepared by intramolecular Thorpe-Ziegler cyclization
- the cyano group of the Thorpe-Ziegler cyclization product can be converted to an amide group by known methods, such as hydrolysis.
- the intermediate 1-2 (900 mg) was dissolved in 1,4-dioxane (10.00 mL) and water (1.00 mL), and 2,4,6-trimethylboroxane (405 mg), potassium carbonate (594 mg) and [1,1'-bis(diphenylphosphino)ferrocene] palladium dichloride (157 mg) were added in sequence, and the mixture was heated and stirred at 120°C for 2 hours under argon protection. After cooling to room temperature, the mixture was directly extracted with ethyl acetate (3x30.0 mL), and the organic phase was washed with saturated brine (2x20.0 mL) and filtered.
- the intermediate 2-2 (600 mg) was dissolved in acetic acid (6 mL), and liquid bromine (79 mg) was added, and the mixture was reacted at 25°C for 3 hours. Saturated sodium bicarbonate aqueous solution was added to the reaction solution until no bubbles were generated, and the mixture was extracted with ethyl acetate (3x10 mL). The organic phase was washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain the intermediate 2-3 (500 mg).
- N,N-diisopropylamine (373 mg) was dissolved in tetrahydrofuran (3 mL) at room temperature, and n-butyl lithium (1.6 M, 2.1 mL) was added at -68 ° C.
- n-butyl lithium 1.6 M, 2.1 mL
- a solution of 2-bromo-5-methylthiophene (500 mg) in tetrahydrofuran (1.00 mL) was slowly added at -68 ° C, and the mixture was stirred at -68 ° C for 2 hours.
- Carbon dioxide gas was introduced at -68 ° C, and then the reaction system was warmed to room temperature and reacted at room temperature for 1 hour.
- the intermediate 3-2 (250 mg) was dissolved in 1,4-dioxane (10.00 mL), and 3-methoxy-2,6-dimethylaniline (258 mg) and propylphosphonic anhydride (1.45 g) were added in sequence.
- the reaction system was heated and stirred at 70 ° C for 16 hours.
- water (10.0 mL) was added to the reaction solution to quench, and it was extracted with ethyl acetate (3x10.0 mL).
- the organic phase was washed with saturated brine (2x10.0 mL), dried over anhydrous sodium sulfate, and filtered.
- the intermediate 3-4 (60 mg) was dissolved in concentrated sulfuric acid (0.50 mL), and the mixture was heated and stirred at 40 ° C for 3 hours. After cooling to room temperature, the reaction solution was diluted with dichloromethane (5.0 mL), and the pH was adjusted to 7-8 with a saturated sodium bicarbonate solution, extracted with dichloromethane (3x5.0 mL), and the organic phase was washed with saturated brine (2x5.0 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain the intermediate 3-5 (40 mg).
- 2-Methylthiazole-4-carboxylic acid (1.00 g) was dissolved in tetrahydrofuran (10.00 mL) at room temperature, and n-butyl lithium (1.6 M, 10.9 mL) was slowly added dropwise at -68 ° C, and the mixture was stirred at -68 ° C for 1 hour.
- Liquid bromine (0.42 mL) was slowly added at -68 ° C, and then the reaction system was warmed to room temperature and reacted at room temperature for 2 hours. Water (10.0 mL) was added to the reaction solution, and the unreacted raw materials were removed by extraction with ethyl acetate (3x10.0 mL).
- the intermediate 9-2 (400 mg) was dissolved in dimethyl sulfoxide (4.00 mL), and 2-cyanoacetamide (105.9 mg), potassium carbonate (290.0 mg) and cuprous iodide (20.0 mg) were added in sequence.
- the reaction system was heated and stirred at 85 ° C in a sealed tube under argon protection for 3 hours. After cooling to room temperature, water (10.0 mL) was added to the reaction solution, and it was extracted with ethyl acetate (3x30.0 mL). The organic phase was washed with saturated brine (3x20.0 mL), dried over anhydrous sodium sulfate, and filtered.
- the intermediate 10-5 (230 mg) was dissolved in concentrated sulfuric acid (1.5 mL), reacted at room temperature for 1 hour, and then heated to 75 ° C for 1 hour.
- LCMS MS m/z (ESI): 263.0 [M+H] + .
- the intermediate 12-2 (4.70 g) was dissolved in ethanol (40.00 mL), and sodium hydroxide (1.43 g) was added under argon protection. The reaction system was heated and stirred at 70 ° C overnight. After cooling to room temperature, the reaction solvent was concentrated, and water (50.0 mL) was added to dilute it. The pH was adjusted to 3-4 with 1N hydrochloric acid. Solids precipitated and filtered. The filter cake was washed with 1% hydrochloric acid and vacuum dried to obtain intermediate 12-3 (4.0 g).
- the intermediate 12-3 (1.2 g, 5.10 mmol) was dissolved in thionyl chloride (15.00 mL), and heated and stirred in an oil bath at 80 ° C for 75 minutes. After cooling to room temperature, the reaction solution was concentrated, the residue was dissolved in dry toluene (5.0 mL), and added dropwise to a pyridine (5 mL) solution of 3-methoxy-2,6-dimethylaniline (1.16 g) under an ice bath. After the addition was complete, the reaction system was warmed to room temperature and reacted overnight under argon protection.
- 5-Bromothiazol-2-amine (10.00 g) was dissolved in dichloromethane (100.0 mL) at room temperature, and triethylamine (14.1 g), 4-dimethylaminopyridine (682.4 mg) and di-tert-butyl dicarbonate (14.6 g) were added in sequence, and the mixture was stirred and heated at 25°C for 2 hours.
- the reaction solution was diluted with water (100.0 mL) and extracted with dichloromethane (3 ⁇ 200.0 mL). The organic phases were combined and washed with saturated brine (3 ⁇ 500.0 mL), dried over anhydrous sodium sulfate, and filtered.
- the intermediate 17-3 (400 mg) was dissolved in N, N-dimethylformamide (6.00 mL), and N-iodosuccinimide (229 mg) was added, and the reaction was stirred at room temperature overnight.
- Potassium fluoride (6 mg) and cuprous iodide (21 mg) were placed in a microwave tube at room temperature, purged with nitrogen, and stirred in an oil bath at 60°C for 1 hour.
- a solution of trimethyltrifluoromethylsilane 120 mg) in N-methylpyrrolidone (0.50 mL) was added, and the reaction system was stirred in an oil bath at 50°C for 45 minutes.
- a solution of intermediate 17-4 (20 mg) in N,N-dimethylformamide (0.50 mL) was added, and the reaction system was stirred in an oil bath at 80°C overnight. Water (5.0 mL) was added to the reaction solution for dilution, and extracted with ethyl acetate (3 ⁇ 5.0 mL).
- the intermediate 23-2 (250 mg) was dissolved in N, N-dimethylformamide (3.00 mL), tributyl (1-ethoxyvinyl) tin (265 mg) and XPhos-Pd-G2 (39 mg) were added, and the reaction system was replaced with argon three times, and then heated and stirred at 100 ° C for 2 hours under argon protection.
- the reaction solution was poured into water (20.0 mL), extracted with ethyl acetate (3 ⁇ 20.0 mL), and the organic phase was washed with saturated brine (2 ⁇ 20.0 mL), dried over anhydrous sodium sulfate, and filtered.
- the intermediate 23-2 (200 mg) was dissolved in N, N-dimethylformamide (20.00 mL), and 2-(trimethyltinyl)thiazole (291 mg) and [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (29 mg) were added in sequence.
- the reaction system was heated and stirred at 80°C under argon protection for 2 hours. After cooling to room temperature, the mixture was diluted with water (10.0 mL), extracted with ethyl acetate (3 ⁇ 10.0 mL), and the organic phase was washed with saturated brine (2 ⁇ 10.0 mL), dried over anhydrous sodium sulfate, and filtered.
- step 1.1 the 3-methoxy-2,6-dimethylaniline in step 1.1 was replaced with 5-methyl-1H-indazol-4-amine to obtain the target compound 6-amino-2-methyl-5-(5-methyl-1H-indazol-4-yl)-4-oxo-4,5-dihydrothiazolo[5,4-c]pyridine-7-carboxamide (Compound 036).
- the intermediate 43-2 (10 g) was dissolved in ethanol (100 mL), and an ethanol solution of sodium ethoxide (0.5 N, 100 mL) was added under argon protection, and then the temperature was raised to 60 ° C for 16 hours. Water (100.0 mL) was added to the reaction solution to quench, and it was extracted with ethyl acetate (3 ⁇ 100.0 mL).
- the intermediate 43-3 (4.8 g) was dissolved in water (48 mL), hydrogen bromide (7.68 mL) was added, and the mixture was cooled to -5°C.
- Cuprous bromide (12.3 g) was added, the mixture was heated to room temperature and stirred for 30 minutes, and then heated to 100°C in an oil bath and stirred for 2 hours. The mixture was cooled to room temperature, and the reaction solution was extracted with ethyl acetate (3 ⁇ 100.0 mL).
- the intermediate 43-4 (900.0 mg) was dissolved in tetrahydrofuran (9.00 mL), cooled to 0°C, sodium hydride (232.6 mg) was added, stirred for 30 minutes, iodomethane (825.6 mg) was added, and the mixture was heated and stirred in a 50°C oil bath for 16 hours.
- water (20.0 mL) was added to the reaction solution, and the mixture was extracted with ethyl acetate (3 ⁇ 20.0 mL). The organic phase was washed with saturated brine (2 ⁇ 20.0 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure.
- 2,4-Dibromothiophene (10 g) was dissolved in ether (10 mL), and n-butyl lithium (25.8 mL, 1.6 M) was slowly added at -68 ° C, and stirred for 0.5 hours.
- a solution of dimethyl disulfide (4.28 g) in tetrahydrofuran (5 mL) was slowly added at -68 ° C, and continued to stir for 0.5 hours, and then the temperature was raised to 0 ° C and stirred for 1 hour.
- Saturated ammonium chloride solution 100 mL was slowly added to the reaction solution to quench, and extracted with ethyl acetate (3 ⁇ 100 mL).
- Example 28 replacing intermediate 50-3 with 44-6 to obtain the target product 6-amino-5-(3-hydroxy-2,6-dimethylphenyl)-2-(methylthio)-4-oxo-4,5-dihydrothieno[3,2-c]pyridine-7-carboxamide (Compound 044).
- lithium diisopropylamide (36.9 ml, 2M) was dissolved in tetrahydrofuran (10 ml), and a mixture of 4-methylthiophene-3-carboxylic acid (5 g) and tetrahydrofuran (40 ml) was slowly added, and stirred at -68°C for 0.5 h.
- a solution of carbon tetrabromide (12.8 g) in tetrahydrofuran (10 ml) was slowly added dropwise, and stirred at -68°C for 30 minutes. The reaction solution was heated to 25°C and stirred for 2 hours.
- the intermediate 46-2 (200 mg) was dissolved in dioxane (5 mL) and water (0.5 mL), and potassium cyclopropyl trifluoroborate (355 mg), potassium carbonate (132.6 mg), [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium (35 mg) were added, and the gas was replaced three times under nitrogen protection, and the reaction was carried out at 100°C for 16 hours. Water (20 mL) was added to the reaction solution, and it was extracted with ethyl acetate (20 mLx3), and the organic phase was washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
- the remaining demethylation method refers to the above step 25.4.
- potassium ethylene trifluoroborate in step 25.1 was replaced with 4,4,5,5-tetramethyl-2-(prop-1-en-2-yl)-1,3,2-dioxaborane to obtain compound 6-amino-5-(3-hydroxy-2,6-dimethylphenyl)-2-isopropyl-3-methyl-4-oxo-4,5-dihydrothieno[3,2-c]pyridine-7-carboxamide (Compound 048).
- Example 28 6-amino-5-(3-hydroxy-2,6-dimethylphenyl)-3-methyl-2-(methylthio)-4-oxo-4,5-dihydrothieno [3,2-c]pyridine-7-carboxamide (Compound 050), (S)-6-amino-5-(3-hydroxy-2,6-dimethylphenyl)-3-methyl-2-( methylthio )-4-oxo-4,5- dihydrothieno[3,2-c]pyridine-7-carboxamide (Compound 050-1) and (R)-6-amino-5-(3-hydroxy-2,6-dimethylphenyl)-3-methyl-2-(methylthio)-4-oxo-4,5-dihydrothieno[3,2-c]pyridine-7-carboxamide ( Compound 050-2)
- the intermediate 50-2 (145 mg) was dissolved in dioxane (2 mL) and water (0.2 mL), and 2,4,6-trimethyl-1,3,5,2,4,6-trioxatriborane (0.22 mL), 1,1'-bis(diphenylphosphino)ferrocenepalladium dichloride (54 mg) and potassium carbonate (72 mg) were added, and the reaction solution was heated to 100 ° C for 16 hours. After cooling to room temperature, water (5 mL) was added to the reaction solution for dilution, and it was extracted with ethyl acetate (3 ⁇ 10 mL).
- the intermediate 50-3 (77 mg) was dissolved in dichloromethane (1.0 mL) at room temperature, trifluoroacetic acid (0.10 mL) was added, and the reaction was stirred at room temperature for 1 hour. Saturated sodium bicarbonate solution was added to the reaction solution to adjust the pH to 7, and extracted with dichloromethane (3 ⁇ 5.0 mL). The organic phase was washed with saturated brine (2 ⁇ 5.0 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by slurrying with petroleum ether (5.0 mL) to obtain intermediate 50-4 (52 mg). LCMS: MS m/z (ESI): 372.1 [M+H] + .
- the intermediate 50-4 (52 mg) was dissolved in a mixed solution of ethanol (3.0 mL) and water (1.0 mL), and (dimethylphosphonic acid) platinum (II) hydrogen complex (3.3 mg) was added. After replacing with argon three times, the reaction system was stirred in an oil bath at 50°C under argon protection for 3 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by high performance liquid preparative chromatography to obtain 6-amino-5-(3-hydroxy-2,6-dimethylphenyl)-3-methyl-2-(methylthio)-4-oxo-4,5-dihydrothieno[3,2-c]pyridine-7-carboxamide (Compound 050) (5.66 mg).
- the intermediate 45-4 (200 mg) was dissolved in acetonitrile (4.0 mL), 1-(trifluoromethyl)-1,2-benzidoyl-3(1H)-one (559 mg) and potassium carbonate (122 mg) were added, and the reaction solution was replaced with argon three times and reacted at 80°C for 12 hours.
- the intermediate 1-2 (1.00 g) was dissolved in 1,4-dioxane (10.00 mL) and water (1.00 mL), and isopropenylboronic acid pinacol ester (400 mg), potassium carbonate (658 mg) and [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium dichloromethane complex (193 mg) were added in sequence.
- the reaction system was heated in an oil bath at 100°C for 4 hours under argon protection. After cooling to room temperature, water (10.0 mL) was added to the reaction solution, and the mixture was extracted with ethyl acetate (3 ⁇ 10.0 mL).
- the intermediate 53-3 (2 g) was dissolved in acetic acid (20 mL), and liquid bromine (0.5 mL) was added. The reaction was stirred at 50 ° C in a closed reaction system for 16 hours. After cooling to room temperature, a saturated aqueous sodium thiosulfate solution (30 mL) was added to the reaction solution to quench, and extracted with ethyl acetate (3 ⁇ 50 mL). The organic phase was washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The residue was separated and purified by silica gel column chromatography (EA/PE: 0% to 0.5%) to obtain the intermediate 53-3 (1.4 g). LCMS: MS m/z (ESI): 292.0 [M+H] + .
- LCMS MS m/z (ESI): 281.9 [M+H] + .
- the intermediate 54-2 (1.50 g) was dissolved in ethanol (10.00 mL) and water (5 mL), and sodium hydroxide (320.1 mg) was added under argon protection.
- the reaction system was heated and stirred in a 50°C oil bath overnight. After cooling to room temperature, the reaction solution After concentration under reduced pressure, solids precipitated. Water (5.0 mL) was added to dissolve the solids. The pH was adjusted to 3-4 with 1N hydrochloric acid. Solids precipitated. After stirring at room temperature for 30 minutes, the mixture was filtered and the filter cake was dried under vacuum to obtain intermediate 54-3 (1.36 g).
- the intermediate 54-3 (500 mg) was dissolved in thionyl chloride (6 mL), and the mixture was heated and stirred in an oil bath at 80°C for 75 minutes.
- the reaction solution was concentrated under reduced pressure, and the residue was dissolved in dry toluene (5.0 mL) and added dropwise to a pyridine solution of 3-((4-methoxybenzyl)oxy)-2,6-dimethylaniline (759.5 mg) under an ice bath. After the addition was complete, the reaction system was warmed to room temperature and reacted overnight under argon protection.
- the reaction solution was concentrated under reduced pressure, and the residue was diluted with water (10.0 mL) and extracted with ethyl acetate (3 ⁇ 10.0 mL).
- LCMS MS m/z (ESI): 513.3 [M+H] + .
- the intermediate 23-2 (1.0 g) was dissolved in N, N-dimethylformamide (10.0 mL), (tributyltinyl) methanol (944 mg) and XPhos-Pd-G2 (308 mg) were added in sequence, and the reaction system was heated and stirred in an oil bath at 80°C for 2 hours. After cooling to room temperature, water (20.0 mL) was added to the reaction solution for dilution, and the mixture was extracted with ethyl acetate (3 ⁇ 20.0 mL). The organic phase was washed with saturated brine (2 ⁇ 20.0 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure.
- the intermediate 59-3 (110 mg) was dissolved in dichloromethane (4.00 mL), and diethylamine (88 mg) was added. After stirring at room temperature for 10 minutes, sodium triacetoxyborohydride (153 mg) was added and stirred at room temperature for 1 hour. Water (10.00 mL) was added to the reaction solution for dilution, and it was extracted with dichloromethane (3 ⁇ 10.0 mL). The organic phase was washed with saturated brine (2 ⁇ 10.0 mL), dried over anhydrous sodium sulfate, and filtered.
- the intermediate 23-2 (120 mg) was dissolved in concentrated sulfuric acid (2.0 mL) at room temperature, and the reaction system was stirred in an oil bath at 40 ° C for 2 hours. After cooling to room temperature, a saturated sodium bicarbonate solution was added to the reaction solution for neutralization, and extracted with ethyl acetate (3 ⁇ 5.0 mL). The organic phase was washed with saturated brine (2 ⁇ 5.0 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by slurrying with petroleum ether (15.0 mL), filtered, and the filter cake was washed with petroleum ether (3 ⁇ 10.0 mL) and dried in vacuo to obtain the intermediate 60-2 (60 mg).
- the intermediate 104-6 (150 mg) was dissolved in N,N-dimethylformamide (1.5 mL), and tetrahydropyran-4-carboxylic acid (51 mg), 1-ethyl-(3-dimethylaminopropyl)carbodiimide hydrochloride (114 mg), 1-hydroxybenzotriazole (80 mg) and N,N-diisopropylethylamine (154 mg).
- the intermediate 108-2 (1.40 g) was dissolved in isopropanol (12.0 mL) and water (2.0 mL), and the reaction system was heated and stirred in an oil bath at 70°C for 0.5 hours. After cooling to room temperature, the reaction solution was poured into water (20.0 mL), extracted with dichloromethane (3 ⁇ 20.0 mL), and the organic phase was washed with saturated brine (2 ⁇ 20.0 mL), dried over anhydrous sodium sulfate, and filtered.
- the intermediate 108-3 (200 mg) was dissolved in toluene (4.0 mL) at room temperature, and benzophenone imine (190 mg), sodium tert-butoxide (118 mg) and 1,1'-binaphthyl-2,2'-bisdiphenylphosphine (409 mg) and tris(dibenzylideneacetone)dipalladium (200 mg), after replacing with argon three times, the reaction system was heated to 90°C and stirred overnight.
- intermediate 12-2 (36 mg) and intermediate 108-5 (20 mg) were dissolved in 1,4-dioxane (1.0 mL), and trimethylaluminum (0.8 mL, 1.6 M) was added dropwise under stirring.
- the gas was replaced with argon three times, and the reaction system was stirred in a sealed tube in an oil bath at 120°C for 5 hours.
- the reaction was stopped, and the reaction solution was poured into methanol (3.0 mL), extracted with dichloromethane (3 ⁇ 20.0 mL), and the organic phase was washed with saturated brine (2 ⁇ 20.0 mL), dried over anhydrous sodium sulfate, and filtered.
- the filtrate was concentrated under reduced pressure, and the residue was purified by Prep-TLC to obtain intermediate 108-6 (20 mg).
- LCMS MS m/z (ESI): 381.0 [M+H] + .
- the intermediate 108-7 (10 mg) was dissolved in ethanol (3.0 mL) and water (1.0 mL) at room temperature, and (dimethylphosphonic acid) platinum (II) hydrogen complex (1 mg) was added. After replacing with argon three times, the reaction system was stirred in an oil bath at 100°C under argon protection for 24 hours. The reaction solution was concentrated under reduced pressure and purified by high performance liquid preparative chromatography to obtain the target product 6-amino-2,3-dimethyl-5-(5-methylbenzo[d]isothiazol-4-yl)-4-oxo-4,5-dihydrothieno[3,2-c]pyridine-7-carboxamide (Compound 108) (1.13 mg).
- the intermediate 109-2 (5.77 g) was dissolved in trifluoroacetic acid (20.0 mL), and methanesulfonic acid (1.0 mL) was added, and the reaction was stirred at 25°C for 20 minutes.
- Ethyl acetate (50.0 mL) was added to the reaction solution, and the mixture was extracted with water (3 ⁇ 100 mL).
- the pH of the aqueous phase was adjusted to 8-9 with ammonia water, and extracted with ethyl acetate (3 ⁇ 150 mL).
- the organic phase was washed with saturated brine (2 ⁇ 100 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to obtain the intermediate 109-3 (2.2 g).
- the intermediate 109-4 (2.50 g) was dissolved in methanol (10.00 mL) and water (5.00 mL), and lithium hydroxide (628 mg) was added.
- the reaction system was stirred in an oil bath at 25°C for 4 hours. The reaction was stopped, and the methanol was removed by concentration under reduced pressure.
- the pH was adjusted to 3-4 with 1N hydrochloric acid solution, and extracted with ethyl acetate (3 ⁇ 50.0 mL).
- the organic phase was washed with saturated brine (2 ⁇ 50.0 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain the intermediate 109-5 (2.0 g).
- the intermediate 110-2 (10.0 g) was dissolved in 1,4-dioxane (100 mL), 5-methyl-1H-indazole-4-amine (6.27 g) and trimethylaluminum (1.6 M, 132.8 mL) were added, and the gas was replaced with argon three times.
- the reaction solution was heated to 120 ° C for 6 hours. The temperature was lowered to 0 ° C, and the reaction solution was slowly added to methanol (300 mL) to quench.
- the intermediate 110-5 (4 g) was dissolved in a mixed solvent of 1,4-dioxane (40 mL) and water (4 mL), potassium trifluoro(vinyl)borate (3.88 g), potassium carbonate (3.34 g) and [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium (707 mg) were added in sequence, replaced with argon three times, and reacted at 110°C for 16 hours.
- the intermediate 110-7 (140 mg) was dissolved in a mixed solvent of ethanol (3 mL) and water (1 mL), and (dimethylphosphonic acid) platinum (II) hydrogen complex (17 mg) was added, replaced with argon three times, and the reaction solution was heated to 100°C for 16 hours. The solvent was removed under reduced pressure, and the mixture was slurried with a mixed solvent of dichloromethane and methanol (10:1, 1.5 mL).
- the intermediate 111-4 (5.00 g) was dissolved in 1,4-dioxane (50 mL), and the intermediate 111-5 (3.03 g) was added.
- Trimethylaluminum (1.6 M, 59.3 mL) was added under stirring, and the reaction was stirred at 120 ° C for 3 hours under argon protection.
- the reaction solution was slowly added to methanol (200 mL) at 0 ° C to quench, concentrated under reduced pressure, diluted with water (100 mL), extracted with ethyl acetate (3 ⁇ 100 mL), and the organic phase was washed with saturated sodium chloride solution (200 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
- the intermediate 111-7 (2.00 g) was dissolved in a mixed solution of ethanol (21.0 mL) and water (7.0 mL), and (dimethylphosphonic acid) platinum (II) hydrogen complex (245 mg) was added, replaced with argon three times, and heated in an oil bath at 100°C for 16 hours. After cooling to room temperature, solids precipitated, which were filtered and the filter cake was purified by slurrying with methanol (20 mL) to obtain the compound 6-amino-5-(5-chloro-1H-indazol-4-yl)-2-ethyl-3-methyl-4-oxo-4,5-dihydrothieno[3,2-c]pyridine-7-carboxamide (1.48 g).
- LCMS MS m/z(ESI):402.1[M+H] + .
- the intermediate 112-2 (1.28 g) was dissolved in ethylene glycol dimethyl ether (25.0 mL), and malononitrile (433 mg), sodium tert-butoxide (630 mg) and [1,1'-bis(diphenylphosphino)ferrocene] palladium dichloride (240 mg) were added in sequence. After replacing with argon three times, the reaction system was heated and stirred in an oil bath at 100°C under argon protection for overnight reaction. The reaction was stopped, and water (50.0 mL) was added to the reaction solution for dilution.
- the intermediate 110-5 (530 mg) was dissolved in dichloromethane (5.0 mL), 3,4-dihydro-2H-pyran (215 mg) was added, and p-toluenesulfonic acid (22 mg) was added when cooled to 0°C.
- the reaction system was heated to 30°C and stirred for 1 hour.
- the reaction was stopped, water (10.0 mL) was added to the reaction solution for dilution, and it was extracted with dichloromethane (2 ⁇ 10.0 mL).
- the organic phase was washed with saturated sodium bicarbonate solution (2 ⁇ 10.0 mL) and saturated sodium chloride solution (2 ⁇ 10.0 mL) in sequence, dried over anhydrous sodium sulfate, and filtered.
- the intermediate 114-2 (487 mg) was dissolved in 1,4-dioxane (5.0 mL) and water (0.5 mL), and potassium cyclopropyl trifluoroborate (722 mg), potassium carbonate (55 mg), cesium fluoride (44 mg) and [1,1'-bis(diphenylphosphino)ferrocene] palladium dichloride (71 mg) were added in sequence.
- the reaction system was reacted at 100°C for 1 hour in a microwave under argon protection. After cooling to room temperature, water (10.0 mL) was added to the reaction solution for dilution, and extracted with ethyl acetate (3 ⁇ 10.0 mL).
- the intermediate 114-3 (240 mg) was dissolved in a methanol solution of hydrochloric acid (5.0 mL, 4 M), and the reaction system was stirred at 25°C for 1 hour. The reaction was stopped, the reaction solution was concentrated, tetrahydrofuran (10.0 mL) was added to dissolve, water (10.0 mL) was added to dilute, and extracted with ethyl acetate (3 ⁇ 10.0 mL). The organic phase was washed with saturated sodium bicarbonate solution (2 ⁇ 20.0 mL) and saturated brine (2 ⁇ 20.0 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to obtain the intermediate 114-4 (158 mg).
- Intermediate 120-2 (0.8 g) was dissolved in phosphorus oxychloride (5 mL), stirred at room temperature for 0.5 hours, and then heated to 100°C for 6.5 hours. After cooling to room temperature, the reaction solution was concentrated under reduced pressure, toluene (5 mL) was added, and the residual phosphorus oxychloride was removed by concentration under reduced pressure to obtain intermediate 120-3 (954 mg), which was directly used in the next step.
- 2,2,6,6-tetramethylpiperidine (30.0 g) was dissolved in ultra-dry tetrahydrofuran (160.0 mL) at room temperature, replaced with argon three times, and n-butyl lithium (84.0 mL, 2.5 M) was slowly added at -20°C using a constant pressure feed funnel, and the reaction system was stirred at 0°C for 0.5 hours.
- the intermediate 127-2 (32.0 g) was dissolved in dimethyl sulfoxide (320.0 mL), hydrazine hydrate (81.8 mL) was added, and the mixture was stirred at 120°C for 8 hours. The reaction was stopped, and the reaction solution was slowly added into water (3.2 L) and stirred for 1 hour. The pH was adjusted to 5 with 3M hydrochloric acid, and solids precipitated. The filter cake was filtered and dried to obtain the intermediate 127-3 (23.7 g).
- the intermediate 127-3 (23.7 g) was dissolved in dichloromethane (200.0 mL), 3,4-dihydro-2H-pyran (26.1 g) was added, and p-toluenesulfonic acid (1.77 g) was added when the temperature was lowered to 0°C, and then the reaction system was stirred at 30°C for 1 hour. The reaction was stopped, and water (300.0 mL) was added to the reaction solution for dilution, and the mixture was extracted with dichloromethane (3 ⁇ 100.0 mL).
- the intermediate 127-4 (400 mg) was dissolved in 1,4-dioxane (100.0 mL), and benzophenone imine (13.9 g), cesium carbonate (37.4 g), 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (4.40 g) and palladium acetate (868 mg) were added in sequence, and the atmosphere was replaced with argon three times.
- the reaction system was reacted at 110°C for 16 hours. The reaction was stopped, and water (150.0 mL) was added to the reaction solution for dilution, and extracted with ethyl acetate (3 ⁇ 150.0 mL).
- Compound 127 was chirally resolved by SFC to obtain compound (S)-6-amino-2-ethyl-5-(7-fluoro-5-methyl-1H-indazol-4-yl)-3-methyl-4-oxo-4,5-dihydrothieno[3,2-c]pyridine-7-carboxamide (compound 127-P1).
- This series of compounds has atropisomerism in their structures, and two stable isomers can be obtained by chiral SFC separation.
- the results of the aforementioned biological test examples show that the two isomers show large differences in activity, both in enzyme inhibition and cell proliferation inhibition.
- Two representative molecular compounds 047 and 112 were selected, and single crystal cultivation and structural analysis were carried out on the isomers with poor inhibitory activity to determine their absolute configuration.
- the single crystal cultivation method includes: gas-liquid diffusion and slow solvent evaporation crystallization.
- compound 047-2 is obtained by gas-liquid diffusion in an ethanol and n-pentane system to obtain plate-like crystals
- compound 112-P1 is obtained by slow solvent evaporation in an ethanol and n-pentane system to obtain block crystals.
- the single crystal structure analysis data is shown in the table below.
- both test compounds are in R configuration. From this result, it can be inferred that the isomer with better inhibitory activity is S configuration.
- the present invention further improves the activity of the compound by obtaining the torsional isomer.
- the inhibition of the compound on PKMYT1 kinase was detected by ADP-Glo assay.
- the reaction buffer (Assay buffer): 50mM HEPES, 10mM MgCl, 1mM EGTA, 0.01% Brij-35, 2mM DTT, and then dilute the MYT1 enzyme (Carna, 05-176, final concentration of 20nM) and the reaction substrate (ATP with a final concentration of 40 ⁇ M and inactivated CDK1 with a final concentration of 40nM, Signalchem C22-14G) with the reaction buffer. Dilute the test compound 3 times into 10 concentrations, with a starting concentration of 3 ⁇ M.
- the inhibition of the compound on WEE1 kinase was detected by ADP-Glo assay.
- the reaction buffer (Assay buffer): 50mM HEPES, 10mM MgCl, 1mM EGTA, 0.01% Brij-35, 2mM DTT, and then Dilute WEE1 enzyme (Carna, 05-177, final concentration of 10 nM) and reaction substrate (ATP with a final concentration of 20 ⁇ M and Poly (Lys-Tyr, 4:1) with a final concentration of 400 ng/ ⁇ L, BPS Bioscience, 79910) with reaction buffer. Dilute the test compound 3 times to 10 concentrations, with a starting concentration of 3 ⁇ M.
- HCC1569 cells ATCC, CRL-2330
- a 96-well flat-bottom cell culture plate Corning, #3603
- DMSO dilute it to 10 concentration points at a ratio of 1:3.
- CD-1 mice were purchased from Zhejiang Weitonglihua Experimental Animal Technology Co., Ltd., Sibeifu (Beijing) Biotechnology Co., Ltd. and Sibeifu (Suzhou) Biotechnology Co., Ltd.
- DMSO, Solutol, PEG400, acetonitrile, Methanol and VETPGS were purchased from Sigma-Aldrich.
- the LC-MS/MS system was Waters Acquity UPLC class I plus connected in series with AB Sciex Triple Quad 6500+, and the chromatographic column was Agilent Poroshell 120EC-C18 4 ⁇ m (50 ⁇ 2.1mm). All data were collected and processed by Analyst software, and pharmacokinetic parameters were calculated using Phoenix Build 8.3.
- mice Six female CD-1 mice (20-30 g, 6-8 weeks) were randomly divided into two groups according to body weight, with three mice in each group.
- the first group was given the test compound by tail vein injection at a dose of 1 mg/kg or 0.5 mg/kg, and the solvent was 5% DMSO + 5% Solutol + 90% Saline solution;
- the second group was orally given the test compound at a dose of 2 mg/kg or 5 mg/kg, and the solvent was 5% DMSO/20% PEG400/75% (15% VETPGS aqueous solution).
- the first group of animals were fed and watered normally before the experiment, while the second group of animals were fasted overnight before the experiment and fed 2 hours after the administration.
- Mouse plasma samples were added with acetonitrile solution containing internal standard compounds and vortexed for 0.5min, then centrifuged at 3900rpm for 15min, the supernatant obtained by centrifugation was transferred and diluted 3 times with aqueous solution, and 2 ⁇ L was injected into the LC-MS/MS system for quantitative analysis.
- sample concentration a female CD-1 mouse plasma standard curve (linear range: 0.5-1000ng/mL) and quality control samples (1, 2, 5, 50, 400, 800ng/mL) were used.
- HCC1569 is a human breast cancer cell.
- a model was established using NOD SCID female mice.
- 0.2 mL (3 ⁇ 10 6 ) of HCC1569 cells were subcutaneously inoculated on the right back of each mouse.
- group administration began.
- the compound was orally administered twice a day.
- the tumor diameter was measured twice a week and at the end of administration.
- TGI% tumor growth inhibition rate
- TGI (%) [(1-(average tumor volume at the end of administration of a treatment group-average tumor volume at the beginning of administration of the treatment group))/(average tumor volume at the end of treatment of the solvent control group-average tumor volume at the beginning of treatment of the solvent control group)] ⁇ 100%.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Life Sciences & Earth Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Epidemiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
涉及药物化学领域,特别地涉及一类用作PKMYT1抑制剂的化合物或药学可接受的盐、其制备方法、药物组合物及其在治疗PKMYT1相关疾病中的用途。
Description
本发明涉及药物化学领域,特别地涉及一类用作PKMYT1抑制剂的化合物或药学可接受的盐、其制备方法、药物组合物及其在治疗PKMYT1相关疾病中的用途。
蛋白激酶,膜相关酪氨酸/苏氨酸1(protein kinase,membraneassociated tyrosine/threonine 1,PKMYT1)属于丝氨酸/苏氨酸激酶WEE家族。PKMYT1能够通过磷酸化细胞周期蛋白依赖性激酶1(CDK1)特异性地调节CDK1的活性,进而调控细胞周期G2期向M期的转换过程。对于正常细胞DNA损伤后检查点触发的细胞周期停滞,PKMYT1不是必需的。用siRNA耗竭PKMYT1单独不影响细胞的长期生长,但在基因组不稳定或存在复制压力或其他细胞周期检查点调控受损的细胞中,PKMYT1起到更为重要的作用。
CCNE1基因扩增是常见的肿瘤驱动突变,出现在卵巢癌、乳腺癌、胃癌、肺癌等多种恶性肿瘤中。CCNE1编码的细胞周期蛋白E1(Cyclin E1)结合并激活CDK2,促进细胞周期G1期向S期的转换过程。Cyclin E的过表达损害G1/S检查点调控,使细胞提前进入S期,增加DNA复制压力,导致基因组不稳定。相比正常细胞,CCNE1扩增的细胞更加依赖其他细胞周期检查点的调控和DNA修复机制的完整性。
PKMYT1激酶抑制与CCNE1扩增是合成致死的。
与PKMYT1同家族的WEE1也能够通过磷酸化CDK1调节其活性,从而调控G1/S检查点。PKMYT1抑制剂、WEE1抑制剂可以靶向癌症中的DNA损伤反应(DDR),抑制肿瘤细胞DNA的损伤修复,并促进其凋亡。目前多个WEE1抑制剂正被开发用于治疗肿瘤。研究证明,与WEE1相比,PKMYT1抑制对正常细胞的影响更小,因此,PKMYT1抑制剂具有更好的安全性。寻找PKMYT1抑制剂被认为是有前途的开发新的抗癌剂的方法。
发明内容
在一方面,提供式(I)的化合物:
或其药学上可接受的盐,
其中,
环A选自苯环、C5-6烃环、5-6元杂环和5-6元杂芳环;
E选自O和S;
R1选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA1RB1、-ORA1、-SRA1、-C(=O)RA1、-C(=NRE1)RA1、-C(=N-ORB1)RA1、-C(=O)ORA1、-OC(=O)RA1、-C(=O)NRA1RB1、-NRA1C(=O)RB1、-C(=NRE1)NRA1RB1、-NRA1C(=NRE1)RB1、-OC(=O)NRA1RB1、-NRA1C(=O)ORB1、-NRA1C(=O)NRA1RB1、-NRA1C(=S)NRA1RB1、-NRA1C(=NRE1)NRA1RB1、-S(=O)rRA1、-S(=O)(=NRE1)RB1、-N=S(=O)RA1RB1、-S(=O)2ORA1、-OS(=O)2RA1、-NRA1S(=O)rRB1、-NRA1S(=O)(=NRE1)RB1、-S(=O)rNRA1RB1、-S(=O)(=NRE1)NRA1RB1、-NRA1S(=O)2NRA1RB1和-NRA1S(=O)(=NRE1)NRA1RB1,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX1的取代基取代;
R2选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA2RB2、-ORA2、-SRA2、-C(=O)RA2、-C(=NRE2)RA2、-C(=N-ORB2)RA2、-C(=O)ORA2、-OC(=O)RA2、-C(=O)NRA2RB2、-NRA2C(=O)RB2、-C(=NRE2)NRA2RB2、-NRA2C(=NRE2)RB2、-OC(=O)NRA2RB2、-NRA2C(=O)ORB2、-NRA2C(=O)NRA2RB2、-NRA2C(=S)NRA2RB2、-NRA2C(=NRE2)NRA2RB2、-S(=O)rRA2、-S(=O)(=NRE2)RB2、-N=S(=O)RA2RB2、-S(=O)2ORA2、-OS(=O)2RA2、-NRA2S(=O)rRB2、-NRA2S(=O)(=NRE2)RB2、-S(=O)rNRA2RB2、-S(=O)(=NRE2)NRA2RB2、-NRA2S(=O)2NRA2RB2和-NRA2S(=O)(=NRE2)NRA2RB2,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX2的取代基取代;
每个R3独立选自卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烃基、-C1-4亚烷基-C3-10环烃基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA3RB3、-ORA3、-SRA3、-C(=O)RA3、-C(=NRE3)RA3、-C(=N-ORB3)RA3、-C(=O)ORA3、-OC(=O)RA3、-C(=O)NRA3RB3、-NRA3C(=O)RB3、-C(=NRE3)NRA3RB3、-NRA3C(=NRE3)RB3、-OC(=O)NRA3RB3、-NRA3C(=O)ORB3、-NRA3C(=O)NRA3RB3、-NRA3C(=S)NRA3RB3、-NRA3C(=NRE3)NRA3RB3、-S(=O)rRA3、-S(=O)(=NRE3)RB3、-N=S(=O)RA3RB3、-S(=O)2ORA3、-OS(=O)2RA3、-NRA3S(=O)rRB3、-NRA3S(=O)(=NRE3)RB3、-S(=O)rNRA3RB3、-S(=O)(=NRE3)NRA3RB3、-NRA3S(=O)2NRA3RB3和-NRA3S(=O)(=NRE3)NRA3RB3,其中每个烷基、亚烷基、烯基、炔基、环烃基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX3的取代基取代;
或任意两个相邻的R3连同与它们相连的原子一起形成一个C3-10单环烃环或者含1、2或3个杂原子的4-12元杂环或者含1、2或3个杂原子的5-10元杂芳环或者苯环,其中杂原子独立选自氧、硫、氮和磷,该环是未被取代的或被至少一个独立选自RX3的取代基取代;
R4选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA4RB4、-ORA4、-SRA4、-C(=O)RA4、-C(=NRE4)RA4、-C(=N-ORB4)RA4、-C(=O)ORA4、-OC(=O)RA4、-C(=O)NRA4RB4、-NRA4C(=O)RB4、-C(=NRE4)NRA4RB4、-NRA4C(=NRE4)RB4、-OC(=O)NRA4RB4、-NRA4C(=O)ORB4、-NRA4C(=O)NRA4RB4、-
NRA4C(=S)NRA4RB4、-NRA4C(=NRE4)NRA4RB4、-S(=O)rRA4、-S(=O)(=NRE4)RB4、-N=S(=O)RA4RB4、-S(=O)2ORA4、-OS(=O)2RA4、-NRA4S(=O)rRB4、-NRA4S(=O)(=NRE4)RB4、-S(=O)rNRA4RB4、-S(=O)(=NRE4)NRA4RB4、-NRA4S(=O)2NRA4RB4和-NRA4S(=O)(=NRE4)NRA4RB4,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX4的取代基取代;
R5选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA5RB5、-ORA5、-SRA5、-C(=O)RA5、-C(=NRE5)RA5、-C(=N-ORB5)RA5、-C(=O)ORA5、-OC(=O)RA5、-C(=O)NRA5RB5、-NRA5C(=O)RB5、-C(=NRE5)NRA5RB5、-NRA5C(=NRE5)RB5、-OC(=O)NRA5RB5、-NRA5C(=O)ORB5、-NRA5C(=O)NRA5RB5、-NRA5C(=S)NRA5RB5、-NRA5C(=NRE5)NRA5RB5、-S(=O)rRA5、-S(=O)(=NRE5)RB5、-N=S(=O)RA5RB5、-S(=O)2ORA5、-OS(=O)2RA5、-NRA5S(=O)rRB5、-NRA5S(=O)(=NRE5)RB5、-S(=O)rNRA5RB5、-S(=O)(=NRE5)NRA5RB5、-NRA5S(=O)2NRA5RB5和-NRA5S(=O)(=NRE5)NRA5RB5,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX5的取代基取代;
R6选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA6RB6、-ORA6、-SRA6、-C(=O)RA6、-C(=NRE6)RA6、-C(=N-ORB6)RA6、-C(=O)ORA6、-OC(=O)RA6、-C(=O)NRA6RB6、-NRA6C(=O)RB6、-C(=NRE6)NRA6RB6、-NRA6C(=NRE6)RB6、-OC(=O)NRA6RB6、-NRA6C(=O)ORB6、-NRA6C(=O)NRA6RB6、-NRA6C(=S)NRA6RB6、-NRA6C(=NRE6)NRA6RB6、-S(=O)rRA6、-S(=O)(=NRE6)RB6、-N=S(=O)RA6RB6、-S(=O)2ORA6、-OS(=O)2RA6、-NRA6S(=O)rRB6、-NRA6S(=O)(=NRE6)RB6、-S(=O)rNRA6RB6、-S(=O)(=NRE6)NRA6RB6、-NRA6S(=O)2NRA6RB6和-NRA6S(=O)(=NRE6)NRA6RB6,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX6的取代基取代;
R7选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA7RB7、-ORA7、-SRA7、-C(=O)RA7、-C(=NRE7)RA7、-C(=N-ORB7)RA7、-C(=O)ORA7、-OC(=O)RA7、-C(=O)NRA7RB7、-NRA7C(=O)RB7、-C(=NRE7)NRA7RB7、-NRA7C(=NRE7)RB7、-OC(=O)NRA7RB7、-NRA7C(=O)ORB7、-NRA7C(=O)NRA7RB7、-NRA7C(=S)NRA7RB7、-NRA7C(=NRE7)NRA7RB7、-S(=O)rRA7、-S(=O)(=NRE7)RB7、-N=S(=O)RA7RB7、-S(=O)2ORA7、-OS(=O)2RA7、-NRA7S(=O)rRB7、-NRA7S(=O)(=NRE7)RB7、-S(=O)rNRA7RB7、-S(=O)(=NRE7)NRA7RB7、-NRA7S(=O)2NRA7RB7和-NRA7S(=O)(=NRE7)NRA7RB7,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX7的取代基取代;
R8选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA8RB8、-ORA8、-SRA8、-C(=O)RA8、-C(=NRE8)RA8、-C(=N-ORB8)RA8、
-C(=O)ORA8、-OC(=O)RA8、-C(=O)NRA8RB8、-NRA8C(=O)RB8、-C(=NRE8)NRA8RB8、-NRA8C(=NRE8)RB8、-OC(=O)NRA8RB8、-NRA8C(=O)ORB8、-NRA8C(=O)NRA8RB8、-NRA8C(=S)NRA8RB8、-NRA8C(=NRE8)NRA8RB8、-S(=O)rRA8、-S(=O)(=NRE8)RB8、-N=S(=O)RA8RB8、-S(=O)2ORA8、-OS(=O)2RA8、-NRA8S(=O)rRB8、-NRA8S(=O)(=NRE8)RB8、-S(=O)rNRA8RB8、-S(=O)(=NRE8)NRA8RB8、-NRA8S(=O)2NRA8RB8和-NRA8S(=O)(=NRE8)NRA8RB8,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX8的取代基取代;
或R7和R8连同与它们相连的原子一起形成一个C5-6烃环或含1、2或3个杂原子的5-6元杂环或5-6元杂芳环,其中杂原子独立选自氧、硫和氮,该环是未被取代的或被至少一个独立选自RX7的取代基取代;
每个RA1、RA2、RA3、RA4、RA5、RA6、RA7、RA8、RB1、RB2、RB3、RB4、RB5、RB6、RB7和RB8独立选自氢、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、芳基、-C1-4亚烷基-芳基、杂芳基和-C1-4亚烷基-杂芳基,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX的取代基取代;
或“RA1和RB1”或“RA2和RB2”或“RA3和RB3”或“RA4和RB4”或“RA5和RB5”或“RA6和RB6”或“RA7和RB7”或“RA8和RB8”一起连同与它们相连的单个或多个原子共同构成一个含有0、1或2个额外的独立选自氧、硫、氮和磷的杂原子的4-12元杂环,该环是未被取代的或被1、2或3个选自RX的取代基取代;
每个RE1、RE2、RE3、RE4、RE5、RE6、RE7和RE8独立选自氢、C1-10烷基、CN、NO2、-S(=O)rRa1、-C(=O)Ra1、-C(=O)ORa1、-C(=O)NRa1Rb1和-S(=O)rNRa1Rb1,其中烷基是未被取代的或被至少一个独立选自RX的取代基取代;
每个RX、RX1、RX2、RX3、RX4、RX5、RX6、RX7和RX8独立选自卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、CN、NO2、-(CRc1Rd1)tNRa1Rb1、-(CRc1Rd1)tORb1、-(CRc1Rd1)tC(=O)Ra1、-(CRc1Rd1)tC(=NRe1)Ra1、-(CRc1Rd1)tC(=O)ORb1、-(CRc1Rd1)tOC(=O)Rb1、-(CRc1Rd1)tC(=O)NRa1Rb1、-(CRc1Rd1)tNRa1C(=O)Rb1、-(CRc1Rd1)tC(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)Rb1、-(CRc1Rd1)tOC(=O)NRa1Rb1、-(CRc1Rd1)tNRa1C(=O)ORb1、-(CRc1Rd1)tNRa1C(=O)NRa1Rb1、-(CRc1Rd1)tNRa1C(=S)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)NRa1Rb1、-(CRc1Rd1)tS(=O)rRb1、-(CRc1Rd1)tS(=O)(=NRe1)Rb1、-(CRc1Rd1)tN=S(=O)Ra1Rb1、-(CRc1Rd1)tS(=O)2ORb1、-(CRc1Rd1)tOS(=O)2Rb1、-(CRc1Rd1)tNRa1S(=O)rRb1、-(CRc1Rd1)tNRa1S(=O)(=NRe1)Rb1、-(CRc1Rd1)tS(=O)rNRa1Rb1、-(CRc1Rd1)tS(=O)(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1S(=O)2NRa1Rb1和-(CRc1Rd1)tNRa1S(=O)(=NRe1)NRa1Rb1,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RY的取代基取代;
每个Ra1和Rb1独立选自氢、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、芳基、-C1-4亚烷基-芳基、杂芳基和-C1-4亚烷基-杂芳基,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂
芳基是未被取代的或被至少一个独立选自RY的取代基取代;
或Ra1和Rb1一起连同与它们相连的单个或多个原子共同构成一个含有0、1或2个额外的独立选自氧、硫、氮和磷的杂原子的4-12元杂环,该环是未被取代的或被1、2或3个选自RY的取代基取代;
每个Rc1和Rd1独立选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、芳基、-C1-4亚烷基-芳基、杂芳基和-C1-4亚烷基-杂芳基,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RY的取代基取代;
或者每个Rc1和Rd1一起连同与它们相连的单个或多个碳原子构成含有0、1或2个独立选自氧、硫和氮的杂原子的3-12元环,该环是未被取代的或被1、2或3个独立选自RY的基团取代;
每个Re1独立选自氢、C1-10烷基、CN、NO2、-S(=O)rRa1、-C(=O)Ra1、-C(=O)ORa1、-C(=O)NRa1Rb1和-S(=O)rNRa1Rb1,其中烷基是未被取代的或被至少一个独立选自RY的取代基取代;
每个RY独立选自卤素、NO2、-CN、C1-10烷基、-OH、-O(C1-10烷基)、-O(C3-10环烷基)、-O(C1-4亚烷基-C3-10环烷基)、-O(杂环基)、-O(C1-4亚烷基-杂环基)、-SH、-S(C1-10烷基)、-S(C3-10环烷基)、-S(C1-4亚烷基-C3-10环烷基)、-S(杂环基)、-S(C1-4亚烷基-杂环基)、-NH2、-NH(C1-10烷基)、-N(C1-10烷基)2、-NH(C3-10环烷基)、-NH(C1-4亚烷基-C3-10环烷基)、-NH(杂环基)和-NH(C1-4亚烷基-杂环基);
g选自0、1、2、3、4、5、6、7和8;
每个r独立选自1和2;
每个t独立选自0、1、2、3和4。
在另一方面,提供一种药物组合物,其包含预防或治疗有效量的本发明的化合物或其药学上可接受的盐,以及至少一种药学上接受的载体。
在另一方面,提供本发明的化合物或其药学上可接受的盐或者药物组合物在在制备治疗疾病、病症或病况的药物中的用途,所述疾病、病症或病况选自细胞增殖异常疾病,特别是癌症。
下面提供具体实施方案来说明本发明的技术内容。本领域技术人员通过说明书中公开的内容可以很容易地理解本发明的其他优点和效果。本发明还可以通过其他不同的具体实施方式来实施或应用。本领域技术人员可以在不脱离本发明的精神的情况下进行各种修改和变化。
定义
除非下文另有定义,本文中使用的所有技术和科学术语与本领域技术人员通常理解的含义相同。本文使用的技术是指本领域通常理解的技术,包括对本领域技术人员显而易见的变体和等效替换。尽管相信以下术语是本领域技术人员容易理解的,但阐述以下定义以更好地说明本发明。当本文出现商品名称时,是指相应的商品或其活性成分。本文引用的所有专利、公开的专利申请和出版物均通过引用并入本文。
术语“合成致死”是指当两个或更多个单独基因同时发生表达异常时导致细胞死亡,而当只有其中一个或一部分基因表达异常时不导致细胞死亡;其中所述表达异常包括突变、过表达或基因抑制(gene inhibition)。在一实施方案中,合成致死的两个基因被称为合成致死基因对。
术语“细胞周期蛋白E1”是指G1/S特异性细胞周期蛋白E1(基因名称:CCNE1)。过表达细胞周期蛋白E1的细胞比正常表达细胞周期蛋白E1的细胞表现出更高的细胞周期蛋白E1活性。CCNE1扩增的细胞是比正常细胞具有更高CCNE1基因拷贝数的细胞。过表达细胞周期蛋白E1的细胞可以是CCNE1扩增的细胞,例如,在一实施方案中,与具有2个拷贝的CCNE1的二倍体正常细胞相比,过表达细胞周期蛋白E1的细胞具有大于2个拷贝拷贝的CCNE1。除了直接检测CCNE1基因的拷贝数,细胞周期蛋白E1过表达也可以通过鉴定细胞中基因产物的表达水平(例如,mRNA转录水平或细胞周期蛋白E1蛋白水平)来测定。
在本文中,术语“过表达细胞周期蛋白E1的癌症”和“CCNE1扩增的癌症”可以互换使用。CCNE1扩增的癌症的例子包括但不限于子宫肉瘤、卵巢癌、乳腺癌、胃癌、食管癌、肺癌、肝癌和子宫内膜癌。
术语“FBXW7”是指含F-框WD重复域蛋白7(F-box/WD repeat-containing protein 7;基因名称:FBXW7)。发生失活突变的FBXW7基因是指在细胞中不能产生正常功能性FBXW7蛋白或产生减少量的FBXW7蛋白的突变FBXW7基因。FBXW7突变癌症的例子包括但不限于血液肿瘤、神经胶质瘤、肝癌(例如肝细胞癌)、子宫癌(例如子宫内膜癌)、结直肠癌(例如结直肠腺癌)、乳腺癌、肺癌(例如非小细胞肺癌(NSCLC))、胃癌、食道癌、食管胃结合部腺癌、膀胱癌(例如膀胱尿路上皮癌)、头颈癌(例如头颈部鳞状细胞癌)、宫颈癌(例如宫颈鳞状细胞癌)、黑素瘤、卵巢癌(例如高级别浆液性卵巢癌)。
术语“p53蛋白”是指肿瘤蛋白p53(基因名称:TP53)。发生失活突变的TP53基因是指在细胞中不能产生功能性p53蛋白或产生减少量的p53蛋白的突变TP53基因。
当以范围、优选范围或优选上限或优选下限的形式阐述某一量、浓度或其他数值或参数时,应理解为等同于具体揭示通过将任何上限或优选值与任何下限或优选值组合形成的任何范围,无论所述范围是否明确记载。除非另有说明,否则本文列出的数值范围旨在包括范围的端点以及范围内的所有整数和分数(小数)。例如,表述“C1-C10”或“C1-10”涵盖1-10个碳原子的范围,并应理解为还涵盖其中的任意亚范围以及每个点值,例如C2-3、C2-4、C2-5、C3-4、C3-5、C3-6、C3-7、C1-2、C1-3、C1-4、C1-5、C1-6、C1-7、C1-8、C1-9等,以及C1、C2、C3、C4、C5、C6、C7、C8、C9、C10等。表述“C3-C10”或“C3-10”也应当以类似的方式理解,例如可以涵盖C3-4、C3-5、C3-6、C3-7、C3-8、C4-5、C4-6、C4-C7、C5-C6等以及C3、C4、C5、C6、C7、C8、C9、C10等。又例如,表述“3-14元”应理解为涵盖其中的任意亚范围以及每个点值,例如3-4、3-5、3-6、3-7、3-8、4-5、4-6、4-7、5-6、3、4、5、6或7元,等。表述“3-12元”、“3-7元”、“4-8元”也应当以类似的方式理解。表述“p为选自0至13的整数”表示p是0-13的任意整数,例如p可以是0、1、2、3、4、5、6、7、8、9、10、11、12或13。其他类似的表述例如p1和p2也应以类似的方式理解。
当任何变量(例如RX)在化合物的组成或结构中出现一次以上时,其在每次出现时在每一种情况下定义都是独立的。例如,表述“每个RX独立选自”表示,如果含有多个RX,则每种情况下每个RX取代基的选项都是相互独立的。其他变量或表述例如R3也应以类似的方式理解。
除非上下文另有明确规定,否则“一种(个)”和“该种(个)”等单数形式包括复数形式。表述“一种(个)或多种(个)”或“至少一种(个)”可表示1、2、3、4、5、6、7、8、9或更多。在一实施方案中,“至少一个”表示1、2、3或4个。
术语“可选”或“任选”是指随后描述的事件可能发生但不一定发生,并且描述包括其中所述事件或情况发生或不发生的情况。
术语“取代”和“取代的”指所指定的原子上的一个或多个(例如一个、两个、三个或四个)氢被从所指出的基团的选择代替,条件是未超过所指定的原子在当前情况下的正常原子价并且所述取代形成稳定的化合物。取代基和/或变量的组合仅仅当这种组合形成稳定的化合物时才是允许的。当描述某取代基不存在时,应当理解该取代基可以为一个或多个氢原子,前提是所述结构能使化合物达到稳定的状态。
除非指明,否则如本文中所使用,取代基的连接点可来自取代基的任意适宜位置。当取代基的键显示为穿过环中连接两个原子的键时,则这样的取代基可键连至该可取代的环中的任一成环原子。
表述“包含”、“包括”、“含有”和“具有”是开放式的,并且不排除额外的未列举的元素、步骤或成分。表述“由……组成”不包括未指定的任何元素、步骤或成分。表述“基本上由...组成”是指范围限于指定的元素、步骤或成分,以及任选存在的不会实质性地影响要求保护的主题的基本和新颖特征的元素、步骤或成分。应当理解,表述“包含”包括表述“基本上由……组成”和“由……组成”。
术语“卤”或“卤素”或“卤代”应理解为表示氟(F)、氯(Cl)、溴(Br)或碘(I)原子,优选氟、氯、溴原子。
术语“烃基”是指衍生自烃的单价基团。烃基的实例包括但不限于烷基、烯基、炔基、环烃基和芳基。
术语“烷基”是指由碳原子和氢原子组成的饱和脂肪烃基团,其通过单键与分子的其余部分连接。烷基包括直链烷基和支链烷基。烷基可以含有1-10个碳原子,称为C1-10烷基,例如C1-6烷基、C1-4烷基、C1-3烷基、C1-2烷基、C3烷基、C4烷基、C3-6烷基。直链烷基的非限制性实例包括但不限于甲基、乙基、n-丙基、n-丁基、n-戊基、n-己基等。支链烷基的非限制性实例包括但不限于异丙基、异丁基、仲丁基、叔丁基、异戊基、2-甲基丁基、1-甲基丁基、1-乙基丙基、1,2-二甲基丙基、新戊基、1,1-二甲基丙基、4-甲基戊基、3-甲基戊基、2-甲基戊基、1-甲基戊基、2-乙基丁基、1-乙基丁基、3,3-二甲基丁基、2,2-二甲基丁基、1,1-二甲基丁基、2,3-二甲基丁基、1,3-二甲基丁基或1,2-二甲基丁基等。
二价基团是指由相应的一价基团的具有自由价电子的碳原子去除一个氢原子从而获得的基团。二价基团具有两个与分子其余部分相连的连接位点,其中两个连接位点可以位于所述二价基团的相同原子或两个不同原子上。
“亚烷基(alkylene)”或“亚烷基(alkylidene)”指饱和二价烃基。亚烷基包括直链或支链亚烷基。直链亚烷基的实例包括但不限于亚甲基(-CH2-)、-(CH2)2-、-(CH2)3-、-(CH2)4-、-(CH2)5-、-(CH2)6-等。支链亚烷基的实例包括但不限于-CH(CH3)-、-CH(C2H5)-、-CH(CH3)-CH2-、-CH(C3H7)-、-CH(C2H5)-CH2-、-C(CH3)2-CH2-、-(CH(CH3))2-、-CH(CH3)-(CH2)2-、-CH2-CH(CH3)-CH2-、-CH(C4H9)-、-C(CH3)(C3H7)-、-C(C2H5)2-、-CH(C3H7)-CH2-、-CH(C2H5)-CH(CH3)-、-CH(C2H5)-(CH2)2-、-CH2-CH(C2H5)-CH2-、-C(CH3)2-(CH2)2-、-CH2-C(CH3)2-CH2-、-CH(CH3)-(CH2)3-、-CH2-CH(CH3)-(CH2)2-、-CH(C5H11)-、-C(C2H5)(C3H7)-、-C(CH3)(C4H9)-、-CH(C4H9)-CH2-、-C(C2H5)2-CH2-、-C(CH3)(C3H7)-CH2-、-CH(C2H5)-CH(C2H5)-、-CH(CH3)-CH(C3H7)-、-C(CH3)2-C(CH3)2-、-CH(C3H7)-(CH2)2-、-CH2-CH(C3H7)-CH2-、-CH(C2H5)-C(CH3)2-、-C(CH3)2-CH(CH3)-CH2-、-CH(CH3)-C(CH3)2-CH2-、-CH(C2H5)-CH(CH3)-CH2-、-CH(CH3)-CH(C2H5)-CH2-、-CH(CH3)-CH2-CH(C2H5)-、-CH(CH3)-C(CH3)2-CH2-、-(CH(CH3))3-、-C(CH3)2-(CH2)3-、-CH(C2H5)-(CH2)3-、-CH2-CH(C2H5)-(CH2)2-、-CH2-CH(CH3)-CH(CH3)-CH2-、-(CH(CH3))2-(CH2)2-、-CH(CH3)-(CH2)2-CH(CH3)-、-(CH2)2-CH(CH3)-(CH2)2-、-CH2-CH(CH3)-(CH2)3-、-CH(CH3)-(CH2)4-等。
术语“烯基”是指由碳原子和氢原子组成的直链或支链的具有至少一个双键的不饱和脂肪族烃基。烯基可以具有2-8个碳原子,即“C2-8烯基”,例如C2-4烯基、C3-4烯基。烯基的非限制性实例包括但不限于乙烯基、烯丙基、(E)-2-甲基乙烯基、(Z)-2-甲基乙烯基、(E)-丁-2-烯基、(Z)-丁-2-烯基、(E)-丁-1-烯基、(Z)-丁-1-烯基等。
术语“炔基”是指由碳原子和氢原子组成的直链或支链的具有至少一个三键的不饱和脂肪族烃基。炔基可以具有2-8个碳原子,即“C2-8炔基”,例如C2-4炔基、C3-4炔基。炔基的非限制性实例包括但不限于乙炔基、丙-1-炔基、丙-2-炔基、丁-1-炔基、丁-2-炔基、丁-3-炔基等。
术语“烷氧基”,是指与氧原子以单键相连的如上定义的烷基。烷氧基与分子的其他部分通过氧原子相连。烷氧基可以表示为-O(烷基)。“C1-8烷氧基”或“-O(C1-8烷基)”是指含有1-8个碳原子的烷氧基,其中的烷基部分可为直链或支链结构。烷氧基包括但不仅限于,甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、正戊氧基等。
术语“烃环”和“环烃基”各自是指由碳原子和氢原子组成的饱和或不饱和的非芳香性的环体系。“烃环”和“环烃基”优选地各自包含1或2个环。所述“烃环”和“环烃基”可以是单环、稠合多环、桥环或螺环结构。“烃环”和“环烃基”可以各自具有3-10个碳原子,即“C3-10烃环”(例如C4烃环、C5烃环、C6烃环、C7烃环)和“C3-10环烃基”(例如C3-8环烃基、C5环烃基、C6环烃基、C7环烃基)。烃环的非限制性实例包括但不限于环戊烷、环己烷、环庚烷、环戊烯、环己烯等。环烃基的非限制性实例包括但不限于环丙烷基、环丁烷基、环戊烷基、环己烷基、环庚烷基、双环[2.2.1]庚基和螺[3.3]庚基、环戊烯基、环己烯基等。在一些实施例中,环烃基中的C原子任选被氧代取代。在一些实施例中,环烃基中的C原子任选地被亚氨基取代。在一些实施例中,亚氨基是未取代(=NH)的,或被上下文所述的基团取代。在一些实施方案中,所述亚氨基被-OR取代,其中R为H或C1-10烷基。
术语“环状烷基”和“环烷基”在本文中具有相同的含义,并且可以互换使用。环烷基
是指饱和的环烃基。环烷基可以具有3、4、5、6、7、8、9或10个成环碳原子(C3-10)。环烷基的实例包括但不限于环丙基、环丁基、环戊基、环己基。在一些实施方案中,环烷基为C3-7单环或双环环烷基,优选C3-6单环环烷基,特别是环丙基。
术语“杂环”和“杂环基”各自是指具有例如3-14个(例如具有7-14个,3-8个、3-7个、4-6个或5-6个)环原子的单环或双环的环体系(3-14元、7-14元、3-8元、3-7元、4-6元、5-6元),其中至少一个环原子(例如1、2或3个)是选自氧、硫、氮和磷的杂原子,且其余环原子是C。该环体系可以是饱和(也可以理解为相应的“杂环烷烃”或“杂环烷基”)或不饱和的(即在环内具有一个或多个双键和/或三键)。任选地,“杂环”和“杂环基”各自可以是苯并稠和的。“杂环”和“杂环基”各自不具备芳香性。在一些实施例中,“杂环”和“杂环基”中的C,N,S和P原子任选被氧代取代。在一些实施例中,“杂环”和“杂环基”中的C,S和P原子任选地被亚氨基取代。在一些实施例中,亚氨基是未取代(=NH)的,或被上下文所述的基团取代。在一些实施方案中,所述亚氨基被-OR取代,其中R为H或C1-10烷基。
杂环基可以是例如四元环,如氮杂环丁烷基、氧杂环丁烷基;或者五元环,如四氢呋喃基、二噁烷基(dioxolinyl)、吡咯烷基、咪唑烷基、吡唑烷基、吡咯啉基、氧代吡咯烷基、2-氧代咪唑烷-1-基;或者六元环,如四氢吡喃基、哌啶基、吗啉基、二噻烷基、硫代吗啉基、哌嗪基、1,1-二氧代-1,2-噻嗪烷-2-基或三噻烷基;或者七元环,如二氮杂基环。
杂环基可以是双环的,不受其限制,例如五元并五元环,如八氢环戊烷[c]吡咯基;或者五元并六元双环,如八氢吡咯并[1,2-b]吡嗪基。
如上文所提到的,杂环可以是不饱和的,即其可以包含一个或多个双键,不受其限制,例如包含氮原子的不饱和的杂环可以是1,6-二氢嘧啶、1,2-二氢嘧啶、1,4-二氢嘧啶、1,6-二氢吡啶、1,2-二氢吡啶、1,4-二氢吡啶、2,3-二氢-1H-吡咯、3,4-二氢-1H-吡咯、2,5-二氢-1H-吡咯基、4H-[1,3,4]噻二嗪基、4,5-二氢噁唑基或4H-[1,4]噻嗪基环,包含氧原子的不饱和的杂环可以是2H-吡喃、4H-吡喃、2,3-二氢呋喃,包含硫原子的不饱和的杂环可以是2H-噻喃、4H-噻喃。杂环可以是苯并稠和的,不受其限制,例如二氢异喹啉基环。
示例性的双环杂环还包括:
术语“芳基”是指具有共轭的π电子体系的全碳单环或稠合多环(如双环)的芳香环基团。例如,芳基可以具有6-14个碳原子,适合地具有6-10个,更适合地具有6个或10个。芳基的实例包括但不限于苯基、萘基和蒽基等。
术语“杂芳环”和“杂芳基”各自是指单环、双环或三环芳族环系统,其具有5、6、7、8、9或10个环原子(“5-10元杂芳基”),特别是5或6或9或10个环原子,并且环原子中包含至少一个(适合地为1-4个,更适合地为1、2或3个)可以相同或不同的杂原子,所述杂原子是例如氧、氮或硫。此外,在每种情况下“杂芳环”和“杂芳基”各自可以是苯并稠合的。特别地,杂芳基选自噻吩基、呋喃基、吡咯基、噁唑基、噻唑基、咪唑基、吡唑基、异噁唑基、异噻唑基、噁二唑基、三唑基、噻二唑基等,以及它们的苯并衍生物,例如苯并呋喃基、苯并噻吩基、苯并噁唑基、苯并异噁唑基、苯并咪唑基、苯并三唑基、吲唑基、吲哚基、异吲哚基等;或者吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基等,以及它们的苯并衍生物,例如喹啉基、喹唑啉基、异喹啉基等;或者吖辛因基(azocinyl)、吲嗪基、嘌呤基等,以及它们的苯并衍生物;或者噌啉基、酞嗪基、喹唑啉基、喹喔啉基、萘啶基、咔唑基、吖啶基等。
本发明的化合物的药学上可接受的盐包括其酸加成盐及碱加成盐。用于制备本发明的化合物的药学上可接受的盐的方法为本领域技术人员已知的。
本发明的化合物涵盖其药学上可接受的盐、立体异构体、溶剂化物、多晶型、互变异构体、同位素化合物、代谢产物或前药。
本发明的化合物可以存在特定的几何或立体异构体形式。本发明设想所有的这类化合物。例如,本发明化合物可能存在E或Z构型的碳-碳双键或碳-氮双键,其中“E”代表按Cahn-Ingold-Prelog优先规则,较优的取代基在碳-碳双键或碳-氮双键的异侧,而“Z”代表较优的取代基在碳-碳双键或碳-氮双键的同侧。本发明化合物也可能以“E”和“Z”异构体的混合物形式存在。异构体形式还包括顺式和反式异构体、(-)-和(+)-对对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,及其外消旋混合物和其他混合物,例如对映异构体或非对映体富集的混合物,所有这些混合物都属于本发明的范围之内。此类物质的纯化和分离可通过本领域已知的标准技术实现。
本发明的化合物可以含有具有旋转受阻的键,使得可以分离得到两个单独的扭转异构体。所有可能的扭转异构体均包括在本发明的范围内。扭转异构体的分离方法可以是已知方法或采用本文所述的方法。在一实施方案中,采用手性色谱拆分扭转异构体,例如采用超临界CO2和MeOH作为流动相。
根据常规方法通过拆分外消旋混合物可获得光学纯对映异构体,例如通过使用具有光学活性的酸或碱形成非对映异构体盐,或者通过形成共价非对映异构体。非对映异构体的混合物可基于它们的物理和/或化学差异,通过本领域已知的方法(例如通过色谱法或分级结晶)分离成单一的非对映异构体。然后,从分离的非对映异构体盐中释放具有光学活性的对映体碱或酸。另一种分离消旋对映异构体的方法可使用手性色谱法(例如手性HPLC柱),被分离的手性异构体可以在分离前进行常规衍生化处理或不衍生化,取决于何种方法可以实现更有效地分离手性异构体。还可以使用酶法来分离衍生化的或没被衍生化的手性异构体。同样地,可使用具有光学活性的原料,通过手性合成来获得本
发明的光学纯化合物。
本发明的化合物可以溶剂化物(优选水合物)的形式存在,其中本发明的化合物包含作为所述化合物晶格的结构要素的溶剂,特别是例如水、甲醇或乙醇。溶剂特别是水的量可以化学计量比或非化学计量比存在。
本发明还涵盖本发明的化合物的所有可能的结晶形式或多晶型物,其可为单一多晶型物或多于一种多晶型物的任意比例的混合物。
本发明化合物可以以同位素标记或富集的形式存在,包含一个或多个与自然界最普遍原子质量和质量数不同的原子。同位素可以为放射性或非放射性同位素。原子如氢、碳、氮、氧、磷、硫、氟、氯和碘的同位素包括,但不仅限于,2H、3H、13C、14C、15N、18O、32P、35S、18F、36Cl和125I。
在本发明的范围内还包括本发明的化合物的代谢产物,即在给药本发明的化合物时体内形成的物质。这样的产物可由例如由给药的化合物的氧化、还原、水解、酰胺化、脱酰胺化、酯化、酶解等产生。
本发明在其范围内进一步包括本发明的化合物的前药,其为自身可具有较小药理学活性或无药理学活性的本发明的化合物的某些衍生物当被给药至身体中或其上时可通过例如水解裂解转化成具有期望活性的本发明的化合物。
术语“多晶型”或“多晶型物”是指单一多晶型物或多于一种多晶型物的任意比例的混合物。
术语“晶型”或“晶体”是指呈现三维排序的任意固体物质,与无定形固体物质相反,其产生具有边界清楚的峰的特征性X-射线粉末衍射图谱。
术语“无定形”是指三维上无排序的任意固体物质。
术语“药学上可接受的”是指在正常的医学判断范围内与患者的组织接触而不会有不适当毒性、刺激性、过敏反应等。
术语“药学上可接受的载体”是指对有机体无明显刺激作用,而且不会损害该活性化合物的生物活性及性能的那些物质。“药学上可接受的载体”包括但不限于助流剂、增甜剂、稀释剂、防腐剂、染料/着色剂、矫味剂、表面活性剂、润湿剂、分散剂、崩解剂、稳定剂、溶剂或乳化剂。
术语“活性成分”、“治疗剂”、“活性物质”或“活性剂”是指一种化学实体,其可以有效地治疗或预防目标紊乱、疾病或病症。
针对药物、药物单元或活性成分而言,术语“有效量”、“治疗有效量”或“预防有效量”是指副作用可接受的但能达到预期效果的药物或药剂的足够用量。有效量的确定因人而异,取决于个体的年龄和一般情况,也取决于具体的活性物质,个案中合适的有效量可以由本领域技术人员根据常规试验确定。
术语“个体”包括人或非人动物。示例性人个体包括患有疾病(例如本文所述的疾病)的人个体(称为患者)或正常个体。本发明中“非人动物”包括所有脊椎动物,例如非哺乳动物(例如鸟类、两栖动物、爬行动物)和哺乳动物,例如非人灵长类、家畜和/或驯化动物(例如绵羊、犬、猫、奶牛、猪等)。
式(I)的化合物
在一方面,本发明提供具有式(I)结构的化合物:
或其药学上可接受的盐,
其中,
环A选自苯环、C5-6烃环、5-6元杂环和5-6元杂芳环;
E选自O和S;
R1选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA1RB1、-ORA1、-SRA1、-C(=O)RA1、-C(=NRE1)RA1、-C(=N-ORB1)RA1、-C(=O)ORA1、-OC(=O)RA1、-C(=O)NRA1RB1、-NRA1C(=O)RB1、-C(=NRE1)NRA1RB1、-NRA1C(=NRE1)RB1、-OC(=O)NRA1RB1、-NRA1C(=O)ORB1、-NRA1C(=O)NRA1RB1、-NRA1C(=S)NRA1RB1、-NRA1C(=NRE1)NRA1RB1、-S(=O)rRA1、-S(=O)(=NRE1)RB1、-N=S(=O)RA1RB1、-S(=O)2ORA1、-OS(=O)2RA1、-NRA1S(=O)rRB1、-NRA1S(=O)(=NRE1)RB1、-S(=O)rNRA1RB1、-S(=O)(=NRE1)NRA1RB1、-NRA1S(=O)2NRA1RB1和-NRA1S(=O)(=NRE1)NRA1RB1,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX1的取代基取代;
R2选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA2RB2、-ORA2、-SRA2、-C(=O)RA2、-C(=NRE2)RA2、-C(=N-ORB2)RA2、-C(=O)ORA2、-OC(=O)RA2、-C(=O)NRA2RB2、-NRA2C(=O)RB2、-C(=NRE2)NRA2RB2、-NRA2C(=NRE2)RB2、-OC(=O)NRA2RB2、-NRA2C(=O)ORB2、-NRA2C(=O)NRA2RB2、-NRA2C(=S)NRA2RB2、-NRA2C(=NRE2)NRA2RB2、-S(=O)rRA2、-S(=O)(=NRE2)RB2、-N=S(=O)RA2RB2、-S(=O)2ORA2、-OS(=O)2RA2、-NRA2S(=O)rRB2、-NRA2S(=O)(=NRE2)RB2、-S(=O)rNRA2RB2、-S(=O)(=NRE2)NRA2RB2、-NRA2S(=O)2NRA2RB2和-NRA2S(=O)(=NRE2)NRA2RB2,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX2的取代基取代;
每个R3独立选自卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烃基、-C1-4亚烷基-C3-10环烃基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA3RB3、-ORA3、-SRA3、-C(=O)RA3、-C(=NRE3)RA3、-C(=N-ORB3)RA3、-C(=O)ORA3、-OC(=O)RA3、-C(=O)NRA3RB3、-NRA3C(=O)RB3、-C(=NRE3)NRA3RB3、-NRA3C(=NRE3)RB3、-OC(=O)NRA3RB3、-NRA3C(=O)ORB3、-NRA3C(=O)NRA3RB3、-NRA3C(=S)NRA3RB3、-NRA3C(=NRE3)NRA3RB3、-S(=O)rRA3、-S(=O)(=NRE3)RB3、-N=S(=O)RA3RB3、-S(=O)2ORA3、-OS(=O)2RA3、-NRA3S(=O)rRB3、-NRA3S(=O)(=NRE3)RB3、-S(=O)rNRA3RB3、-S(=O)(=NRE3)NRA3RB3、-NRA3S(=O)2NRA3RB3
和-NRA3S(=O)(=NRE3)NRA3RB3,其中每个烷基、亚烷基、烯基、炔基、环烃基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX3的取代基取代;
或任意两个相邻的R3连同与它们相连的原子一起形成一个C3-10单环烃环或者含1、2或3个杂原子的4-12元杂环或者含1、2或3个杂原子的5-10元杂芳环或者苯环,其中杂原子独立选自氧、硫、氮和磷,该环是未被取代的或被至少一个独立选自RX3的取代基取代;
R4选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA4RB4、-ORA4、-SRA4、-C(=O)RA4、-C(=NRE4)RA4、-C(=N-ORB4)RA4、-C(=O)ORA4、-OC(=O)RA4、-C(=O)NRA4RB4、-NRA4C(=O)RB4、-C(=NRE4)NRA4RB4、-NRA4C(=NRE4)RB4、-OC(=O)NRA4RB4、-NRA4C(=O)ORB4、-NRA4C(=O)NRA4RB4、-NRA4C(=S)NRA4RB4、-NRA4C(=NRE4)NRA4RB4、-S(=O)rRA4、-S(=O)(=NRE4)RB4、-N=S(=O)RA4RB4、-S(=O)2ORA4、-OS(=O)2RA4、-NRA4S(=O)rRB4、-NRA4S(=O)(=NRE4)RB4、-S(=O)rNRA4RB4、-S(=O)(=NRE4)NRA4RB4、-NRA4S(=O)2NRA4RB4和-NRA4S(=O)(=NRE4)NRA4RB4,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX4的取代基取代;
R5选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA5RB5、-ORA5、-SRA5、-C(=O)RA5、-C(=NRE5)RA5、-C(=N-ORB5)RA5、-C(=O)ORA5、-OC(=O)RA5、-C(=O)NRA5RB5、-NRA5C(=O)RB5、-C(=NRE5)NRA5RB5、-NRA5C(=NRE5)RB5、-OC(=O)NRA5RB5、-NRA5C(=O)ORB5、-NRA5C(=O)NRA5RB5、-NRA5C(=S)NRA5RB5、-NRA5C(=NRE5)NRA5RB5、-S(=O)rRA5、-S(=O)(=NRE5)RB5、-N=S(=O)RA5RB5、-S(=O)2ORA5、-OS(=O)2RA5、-NRA5S(=O)rRB5、-NRA5S(=O)(=NRE5)RB5、-S(=O)rNRA5RB5、-S(=O)(=NRE5)NRA5RB5、-NRA5S(=O)2NRA5RB5和-NRA5S(=O)(=NRE5)NRA5RB5,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX5的取代基取代;
R6选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA6RB6、-ORA6、-SRA6、-C(=O)RA6、-C(=NRE6)RA6、-C(=N-ORB6)RA6、-C(=O)ORA6、-OC(=O)RA6、-C(=O)NRA6RB6、-NRA6C(=O)RB6、-C(=NRE6)NRA6RB6、-NRA6C(=NRE6)RB6、-OC(=O)NRA6RB6、-NRA6C(=O)ORB6、-NRA6C(=O)NRA6RB6、-NRA6C(=S)NRA6RB6、-NRA6C(=NRE6)NRA6RB6、-S(=O)rRA6、-S(=O)(=NRE6)RB6、-N=S(=O)RA6RB6、-S(=O)2ORA6、-OS(=O)2RA6、-NRA6S(=O)rRB6、-NRA6S(=O)(=NRE6)RB6、-S(=O)rNRA6RB6、-S(=O)(=NRE6)NRA6RB6、-NRA6S(=O)2NRA6RB6和-NRA6S(=O)(=NRE6)NRA6RB6,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX6的取代基取代;
R7选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、
-C1-4亚烷基-杂芳基、-NRA7RB7、-ORA7、-SRA7、-C(=O)RA7、-C(=NRE7)RA7、-C(=N-ORB7)RA7、-C(=O)ORA7、-OC(=O)RA7、-C(=O)NRA7RB7、-NRA7C(=O)RB7、-C(=NRE7)NRA7RB7、-NRA7C(=NRE7)RB7、-OC(=O)NRA7RB7、-NRA7C(=O)ORB7、-NRA7C(=O)NRA7RB7、-NRA7C(=S)NRA7RB7、-NRA7C(=NRE7)NRA7RB7、-S(=O)rRA7、-S(=O)(=NRE7)RB7、-N=S(=O)RA7RB7、-S(=O)2ORA7、-OS(=O)2RA7、-NRA7S(=O)rRB7、-NRA7S(=O)(=NRE7)RB7、-S(=O)rNRA7RB7、-S(=O)(=NRE7)NRA7RB7、-NRA7S(=O)2NRA7RB7和-NRA7S(=O)(=NRE7)NRA7RB7,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX7的取代基取代;
R8选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA8RB8、-ORA8、-SRA8、-C(=O)RA8、-C(=NRE8)RA8、-C(=N-ORB8)RA8、-C(=O)ORA8、-OC(=O)RA8、-C(=O)NRA8RB8、-NRA8C(=O)RB8、-C(=NRE8)NRA8RB8、-NRA8C(=NRE8)RB8、-OC(=O)NRA8RB8、-NRA8C(=O)ORB8、-NRA8C(=O)NRA8RB8、-NRA8C(=S)NRA8RB8、-NRA8C(=NRE8)NRA8RB8、-S(=O)rRA8、-S(=O)(=NRE8)RB8、-N=S(=O)RA8RB8、-S(=O)2ORA8、-OS(=O)2RA8、-NRA8S(=O)rRB8、-NRA8S(=O)(=NRE8)RB8、-S(=O)rNRA8RB8、-S(=O)(=NRE8)NRA8RB8、-NRA8S(=O)2NRA8RB8和-NRA8S(=O)(=NRE8)NRA8RB8,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX8的取代基取代;
或R7和R8连同与它们相连的原子一起形成一个C5-6烃环或含1、2或3个杂原子的5-6元杂环或5-6元杂芳环,其中杂原子独立选自氧、硫和氮,该环是未被取代的或被至少一个独立选自RX7的取代基取代;
每个RA1、RA2、RA3、RA4、RA5、RA6、RA7、RA8、RB1、RB2、RB3、RB4、RB5、RB6、RB7和RB8独立选自氢、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-
10环烷基、杂环基、-C1-4亚烷基-杂环基、芳基、-C1-4亚烷基-芳基、杂芳基和-C1-4亚烷基-杂芳基,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX的取代基取代;
或“RA1和RB1”或“RA2和RB2”或“RA3和RB3”或“RA4和RB4”或“RA5和RB5”或“RA6和RB6”或“RA7和RB7”或“RA8和RB8”一起连同与它们相连的单个或多个原子共同构成一个含有0、1或2个额外的独立选自氧、硫、氮和磷的杂原子的4-12元杂环,该环是未被取代的或被1、2或3个选自RX的取代基取代;
每个RE1、RE2、RE3、RE4、RE5、RE6、RE7和RE8独立选自氢、C1-10烷基、CN、NO2、-S(=O)rRa1、-C(=O)Ra1、-C(=O)ORa1、-C(=O)NRa1Rb1和-S(=O)rNRa1Rb1,其中烷基是未被取代的或被至少一个独立选自RX的取代基取代;
每个RX、RX1、RX2、RX3、RX4、RX5、RX6、RX7和RX8独立选自卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、CN、NO2、-(CRc1Rd1)tNRa1Rb1、-(CRc1Rd1)tORb1、-(CRc1Rd1)tC(=O)Ra1、-(CRc1Rd1)tC(=NRe1)Ra1、-(CRc1Rd1)tC(=O)ORb1、-(CRc1Rd1)tOC(=O)Rb1、-(CRc1Rd1)tC(=O)NRa1Rb1、-(CRc1Rd1)tNRa1C(=O)Rb1、-
(CRc1Rd1)tC(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)Rb1、-(CRc1Rd1)tOC(=O)NRa1Rb1、-(CRc1Rd1)tNRa1C(=O)ORb1、-(CRc1Rd1)tNRa1C(=O)NRa1Rb1、-(CRc1Rd1)tNRa1C(=S)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)NRa1Rb1、-(CRc1Rd1)tS(=O)rRb1、-(CRc1Rd1)tS(=O)(=NRe1)Rb1、-(CRc1Rd1)tN=S(=O)Ra1Rb1、-(CRc1Rd1)tS(=O)2ORb1、-(CRc1Rd1)tOS(=O)2Rb1、-(CRc1Rd1)tNRa1S(=O)rRb1、-(CRc1Rd1)tNRa1S(=O)(=NRe1)Rb1、-(CRc1Rd1)tS(=O)rNRa1Rb1、-(CRc1Rd1)tS(=O)(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1S(=O)2NRa1Rb1和-(CRc1Rd1)tNRa1S(=O)(=NRe1)NRa1Rb1,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RY的取代基取代;
每个Ra1和Rb1独立选自氢、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、芳基、-C1-4亚烷基-芳基、杂芳基和-C1-4亚烷基-杂芳基,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RY的取代基取代;
或Ra1和Rb1一起连同与它们相连的单个或多个原子共同构成一个含有0、1或2个额外的独立选自氧、硫、氮和磷的杂原子的4-12元杂环,该环是未被取代的或被1、2或3个选自RY的取代基取代;
每个Rc1和Rd1独立选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、芳基、-C1-4亚烷基-芳基、杂芳基和-C1-4亚烷基-杂芳基,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RY的取代基取代;
或者每个Rc1和Rd1一起连同与它们相连的单个或多个碳原子构成含有0、1或2个独立选自氧、硫和氮的杂原子的3-12元环,该环是未被取代的或被1、2或3个独立选自RY的基团取代;
每个Re1独立选自氢、C1-10烷基、CN、NO2、-S(=O)rRa1、-C(=O)Ra1、-C(=O)ORa1、-C(=O)NRa1Rb1和-S(=O)rNRa1Rb1,其中烷基是未被取代的或被至少一个独立选自RY的取代基取代;
每个RY独立选自卤素、NO2、-CN、C1-10烷基、-OH、-O(C1-10烷基)、-O(C3-10环烷基)、-O(C1-4亚烷基-C3-10环烷基)、-O(杂环基)、-O(C1-4亚烷基-杂环基)、-SH、-S(C1-10烷基)、-S(C3-10环烷基)、-S(C1-4亚烷基-C3-10环烷基)、-S(杂环基)、-S(C1-4亚烷基-杂环基)、-NH2、-NH(C1-10烷基)、-N(C1-10烷基)2、-NH(C3-10环烷基)、-NH(C1-4亚烷基-C3-10环烷基)、-NH(杂环基)和-NH(C1-4亚烷基-杂环基);
g选自0、1、2、3、4、5、6、7和8;
每个r独立选自1和2;
每个t独立选自0、1、2、3和4。
在一实施方案中,t为0。在一实施方案中,g选自0、1和2。在一实施方案中,E为O。在一实施方案中,E为S。
在一实施方案中,每个RA1、RA2、RA3、RA4、RA5、RA6、RA7、RA8、RB1、RB2、RB3、RB4、RB5、RB6、RB7和RB8独立选自氢和C1-10烷基,其中每个烷基是未被取代的或被至少一个独立选自RX的取代基取代。
在一实施方案中,每个RX、RX1、RX2、RX3、RX4、RX5、RX6、RX7和RX8独立选自卤素、C1-10烷基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、-(CRc1Rd1)tNRa1Rb1、-(CRc1Rd1)tORb1、-(CRc1Rd1)tC(=O)Ra1、-(CRc1Rd1)tC(=O)ORb1、-(CRc1Rd1)tOC(=O)Rb1、-(CRc1Rd1)tC(=O)NRa1Rb1、-(CRc1Rd1)tNRa1C(=O)Rb1、-(CRc1Rd1)tOC(=O)NRa1Rb1、-(CRc1Rd1)tNRa1C(=O)ORb1、-(CRc1Rd1)tNRa1C(=O)NRa1Rb1、-(CRc1Rd1)tNRa1C(=S)NRa1Rb1、-(CRc1Rd1)tS(=O)rRb1、-(CRc1Rd1)tS(=O)2ORb1、-(CRc1Rd1)tOS(=O)2Rb1、-(CRc1Rd1)tNRa1S(=O)rRb1、-(CRc1Rd1)tS(=O)rNRa1Rb1,
优选选自卤素、C1-10烷基、CN、NO2、-(CRc1Rd1)tNRa1Rb1、-(CRc1Rd1)tORb1、-(CRc1Rd1)tC(=O)Ra1、-(CRc1Rd1)tC(=O)ORb1、-(CRc1Rd1)tOC(=O)Rb1、-(CRc1Rd1)tC(=O)NRa1Rb1和-(CRc1Rd1)tNRa1C(=O)Rb1,其中每个烷基、亚烷基、环烷基和杂环基是未被取代的或被至少一个独立选自RY的取代基取代。在一实施方案中,每个RX、RX1、RX2、RX4、RX5、RX6、RX7和RX8独立选自卤素、C1-10烷基、CN和NO2,其中每个烷基是未被取代的或被至少一个独立选自RY的取代基取代。在一实施方案中,每个Ra1和Rb1独立选自氢和C1-10烷基,其中每个烷基是未被取代的或被至少一个独立选自RY的取代基取代。在一实施方案中,每个Rc1和Rd1独立选自氢、卤素和C1-10烷基,其中每个烷基是未被取代的或被至少一个独立选自RY的取代基取代。在一实施方案中,每个RY独立选自卤素、NO2、-CN、C1-10烷基、-OH、-O(C1-10烷基)、-NH2、-NH(C1-
10烷基)和-N(C1-10烷基)2,优选选自卤素、NO2、-CN和C1-10烷基。
在一实施方案中,R1选自氢、卤素、CN和NO2。在一实施方案中,R1为氢。在一实施方案中,R1选自C1-10烷基、-NRA1RB1、-ORA1、-SRA1、-OC(=O)RA1、-NRA1C(=O)RB1、-NRA1C(=NRE1)RB1、-OC(=O)NRA1RB1、-NRA1C(=O)ORB1、-NRA1C(=O)NRA1RB1、-NRA1C(=S)NRA1RB1、-NRA1C(=NRE1)NRA1RB1、-OS(=O)2RA1、-NRA1S(=O)rRB1、-NRA1S(=O)(=NRE1)RB1、-NRA1S(=O)2NRA1RB1和-NRA1S(=O)(=NRE1)NRA1RB1,其中每个烷基是未被取代的或被至少一个独立选自RX1的取代基取代。在一实施方案中,R1选自C1-10烷基、-NRA1RB1、-ORA1、-SRA1、-OC(=O)RA1、-NRA1C(=O)RB1、-OS(=O)2RA1和-NRA1S(=O)rRB1,其中每个烷基是未被取代的或被至少一个独立选自RX1的取代基取代。在一实施方案中,R1选自C1-10烷基、-NRA1RB1、-ORA1和-SRA1,优选选自-NRA1RB1和-ORA1,特别是-NRA1RB1。在一实施方案中,每个RB1独立选自C1-10烷基,并且每个RA1为氢。在一实施方案中,每个RA1和RB1为氢。在一实施方案中,每个RE1独立选自氢和C1-10烷基。在一特别的实施方案中,R1为-NH2。在一实施方案中,R1选自-NRA1RB1和氢,优选选自-NH2和氢。
在一实施方案中,R2选自C1-10烷基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、-C(=O)RA2、-C(=NRE2)RA2、-C(=N-ORB2)RA2、-C(=O)ORA2、-C(=O)NRA2RB2、-C(=NRE2)NRA2RB2、-S(=O)rRA2、-S(=O)(=NRE2)RB2、-N=S(=O)RA2RB2、-S(=O)2ORA2、-S(=O)rNRA2RB2和-S(=O)(=NRE2)NRA2RB2,其中每个烷基、亚烷基、环烷基和杂环基是未被取代的或被至少一个独立选自RX2的取代基取代。在一实施方案中,R2选自CN、-C(=O)RA2、-C(=O)ORA2、-C(=O)NRA2RB2、-C(=NRE2)NRA2RB2、-S(=O)rRA2、-S(=O)(=NRE2)RB2、-N=S(=O)RA2RB2、-S(=O)2ORA2、-S(=O)rNRA2RB2和-
S(=O)(=NRE2)NRA2RB2,优选选自CN、-C(=O)RA2、-C(=O)ORA2、-C(=O)NRA2RB2、-S(=O)rRA2、-S(=O)2ORA2和-S(=O)rNRA2RB2。在一实施方案中,R2选自CN、-C(=O)RA2、-C(=O)NRA2RB2和-S(=O)rNRA2RB2,特别是-C(=O)NRA2RB2。在一实施方案中,每个RB2独立选自C1-10烷基,并且每个RA2为氢。在一实施方案中,每个RA2和RB2为氢。在一实施方案中,每个RE2独立选自氢和C1-10烷基。在一特别的实施方案中,R2为-C(=O)NH2。
在一实施方案中,每个R3独立选自卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烃基、-C1-4亚烷基-C3-10环烃基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA3RB3、-ORA3、-SRA3、-C(=O)RA3、-C(=O)ORA3、-OC(=O)RA3、-C(=O)NRA3RB3、-NRA3C(=O)RB3、-OC(=O)NRA3RB3、-NRA3C(=O)ORB3、-NRA3C(=O)NRA3RB3、-NRA3C(=S)NRA3RB3、-S(=O)rRA3、-N=S(=O)RA3RB3、-S(=O)2ORA3、-OS(=O)2RA3、-NRA3S(=O)rRB3、-S(=O)rNRA3RB3和-NRA3S(=O)2NRA3RB3,其中每个烷基、亚烷基、烯基、炔基、环烃基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX3的取代基取代。在一实施方案中,每个R3独立选自卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烃基、-C1-4亚烷基-C3-10环烃基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-
4亚烷基-杂芳基、-NRA3RB3、-ORA3、-SRA3、-C(=O)RA3、-C(=O)ORA3、-OC(=O)RA3、-C(=O)NRA3RB3、-NRA3C(=O)RB3、-S(=O)rRA3、-S(=O)2ORA3、-OS(=O)2RA3、-NRA3S(=O)rRB3和-S(=O)rNRA3RB3,其中每个烷基、亚烷基、烯基、炔基、环烃基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX3的取代基取代。在一实施方案中,每个R3独立选自卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烃基、杂环基、CN、杂芳基、-NRA3RB3、-ORA3、-SRA3、-C(=O)RA3、-C(=O)NRA3RB3和-S(=O)rRA3,其中每个烷基、烯基、炔基、环烃基、杂环基和杂芳基是未被取代的或被至少一个独立选自RX3的取代基取代。在一实施方案中,每个R3独立选自卤素、C1-10烷基、C3-10环烃基、CN、-NRA3RB3、-ORA3、-SRA3和-C(=O)RA3,其中每个烷基、亚烷基和环烃基是未被取代的或被至少一个独立选自RX3的取代基取代。
在一实施方案中,任意两个相邻的R3连同与它们相连的原子一起形成C5-6单环烃环或含1、2或3个杂原子的5-6元杂环或含1、2或3个杂原子的5元杂环或含1或2个杂原子的6元杂芳环,其中杂原子独立选自氧、硫、氮和磷,优选选自氧、硫和氮;该环是未被取代的或被至少一个独立选自RX3的取代基取代。
在一实施方案中,g为1或2。在一实施方案中,卤素选自F、Cl和Br。在一实施方案中,每个RB3独立选自C1-10烷基,并且每个RA3为氢。在一实施方案中,每个RA3和RB3为氢。在一实施方案中,每个RE3独立选自氢和C1-10烷基。在一实施方案中,g为2,并且两个R3相邻,并且两个R3连同与它们相连的原子一起形成环己烯或环己烷,其中所述环己烯或环己烷是未被取代的或被至少一个独立选自RX3的取代基取代。
在一实施方案中,每个R3独立选自F、Cl、Br、CN、OH、甲基、乙基、异丙基、叔丁基、环丙烷基、氮杂环丁烷基、环己烯基、哌啶基、吗啉基、氧代哌嗪基、吡啶基、噻唑基、吡唑基、二乙基胺基、氨基、乙烯基、乙炔基、乙酰基、氨基甲酰基、甲氧基、乙氧基、甲硫基、乙硫基、异丙硫基、甲磺酰基和苯基,其中所述甲基、乙基、异丙基、
叔丁基、环丙烷基、氮杂环丁烷基、环己烯基、哌啶基、吗啉基、氧代哌嗪基、吡啶基、噻唑基、吡唑基、二乙基胺基、乙烯基、乙炔基、乙酰基、甲氧基、乙氧基、甲硫基、乙硫基、异丙硫基、甲磺酰基和苯基是未被取代的或被至少一个独立选自RX3的取代基取代。在一实施方案中,每个RX3独立选自F、Cl、Br、CN、OH、C1-10烷基、-O(C1-10烷基)和杂环基,其中所述C1-10烷基是未被取代的或被至少一个独立选自卤素的取代基取代。在一实施方案中,每个R3独立选自F、Cl、Br、CN、OH、甲基、乙基、异丙基、叔丁基、环丙烷基、二乙基胺基、氨基、乙烯基、乙炔基、乙酰基、氨基甲酰基、甲氧基、甲硫基、乙硫基、异丙硫基、甲磺酰基、苯基、
在一实施方案中,R3为甲基。在一实施方案中,R3为甲基或乙基。在一实施方案中,R3为甲基或甲硫基。在一实施方案中,R3为甲硫基。在一实施方案中,R3为甲基或环丙烷基。在一实施方案中,R3为甲基或异丙基。在一实施方案中,R3为甲基或Cl。在一实施方案中,R3为甲基或三氟甲基。在一实施方案中,R3为甲基或异丙基。在一实施方案中,R3为异丙基。在一实施方案中,R3为叔丁基。
在一实施方案中,任意两个相邻的R3连同与它们相连的原子一起形成选自以下的环:环己烯、环己烷、二氢吡喃、四氢吡喃、环己烯酮、环己酮、二氢吡啶酮、哌啶酮、四氢吡啶、哌啶、环戊烯、环戊烷、二氢噻吩、四氢噻吩、吡啶和二氢吡啶,该环是未被取代的或被至少一个独立选自RX3的取代基取代。在一实施方案中,任意两个相邻的R3连同与它们相连的原子一起形成选自以下的环:
该环是未被取代的或被至少一个独立选自RX3的取代基取代;其中表示R3与环A连接的位点。在一实施方案中,任意两个相邻的R3连同与它们相连的原子一起形成该环是未被取代的或被至少一个独立选自RX3的取代基取代;其中表示R3与环A连接的位点。在一实施方案中,RX3为C1-10烷基,特别是甲基。在一实施方案中,RX3为-(CRc1Rd1)tC(=O)Ra1;其中Ra1优选选自C1-10烷基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基和-C1-4亚烷基-杂环基,特别是杂环基;其中每个烷基、亚烷基、环烷基和杂环基是未被取代的或
被至少一个独立选自RY的取代基取代。在一实施方案中,RX3为-(CRc1Rd1)tC(=O)Ra1;其中Ra1为在一实施方案中,RX3选自-(CRc1Rd1)tNRa1Rb1;其中Ra1优选选自C3-
10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、芳基、-C1-4亚烷基-芳基、杂芳基和-C1-4亚烷基-杂芳基,并且Rb1优选为H;其中每个烷基、亚烷基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RY的取代基取代。在一实施方案中,RX3选自-(CRc1Rd1)tNRa1Rb1;其中Ra1选自在一实施方案中,任意两个相邻的R3连同与它们相连的原子一起形成选自以下的结构:
在一实施方案中,R4选自卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、-NRA4RB4、-ORA4、-SRA4、-C(=O)RA4、-C(=O)ORA4、-OC(=O)RA4、-C(=O)NRA4RB4、-NRA4C(=O)RB4、-OC(=O)NRA4RB4、-NRA4C(=O)ORB4、-NRA4C(=O)NRA4RB4、-NRA4C(=S)NRA4RB4、-S(=O)rRA4、-S(=O)2ORA4、-OS(=O)2RA4、-NRA4S(=O)rRB4、-S(=O)rNRA4RB4和-NRA4S(=O)2NRA4RB4,其中每个烷基、亚烷基、烯基、炔基、环烃基和杂环基是未被取代的或被至少一个独立选自RX4的取代基取代。在一实施方案中,R4选自卤素、C1-10烷基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、-NRA4RB4、-ORA4、-SRA4、-C(=O)RA4、-C(=O)ORA4、-OC(=O)RA4、-C(=O)NRA4RB4、-NRA4C(=O)RB4、-S(=O)rRA4、-S(=O)2ORA4、-OS(=O)2RA4、-NRA4S(=O)rRB4和-S(=O)rNRA4RB4,优选选自卤素、C1-10烷基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、CN、NO2、-NRA4RB4和-ORA4,特别是选自卤素、C1-10烷基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、CN和NO2,其中每个烷基、亚烷基、炔基和环烷基是未被取代的或被至少一个独立选自RX4的取代基取代。在一实施方案中,R4选自C1-
10烷基,优选甲基或乙基,其中每个烷基是未被取代的或被至少一个独立选自RX4的取代基取代。在一实施方案中,R4选自C1-10烷基。在一特别的实施方案中,R4为甲基。在一实施方案中,R4选自卤素,特别是Cl。
在一实施方案中,R5选自氢、卤素、C1-10烷基、CN、NO2、-NRA5RB5、-ORA5、-SRA5、-C(=O)RA5、-C(=O)ORA5、-OC(=O)RA5、-C(=O)NRA5RB5、-NRA5C(=O)RB5、-OC(=O)NRA5RB5、
-NRA5C(=O)ORB5、-NRA5C(=O)NRA5RB5、-NRA5C(=S)NRA5RB5、-S(=O)rRA5、-S(=O)2ORA5、-OS(=O)2RA5、-NRA5S(=O)rRB5、-S(=O)rNRA5RB5和-NRA5S(=O)2NRA5RB5,其中每个烷基是未被取代的或被至少一个独立选自RX5的取代基取代。在一实施方案中,R5选自氢、卤素、C1-10烷基、CN、NO2、-NRA5RB5和-ORA5,特别是选自氢、卤素、C1-10烷基、CN和NO2,其中每个烷基是未被取代的或被至少一个独立选自RX5的取代基取代。在一实施方案中,R5选自卤素,特别是F。在一特别的实施方案中,R5为氢。
在一实施方案中,R6选自氢、卤素、C1-10烷基、CN、NO2、-NRA6RB6、-ORA6、-SRA6、-C(=O)RA6、-C(=O)ORA6、-OC(=O)RA6、-C(=O)NRA6RB6、-NRA6C(=O)RB6、-OC(=O)NRA6RB6、-NRA6C(=O)ORB6、-NRA6C(=O)NRA6RB6、-NRA6C(=S)NRA6RB6、-S(=O)rRA6、-S(=O)2ORA6、-OS(=O)2RA6、-NRA6S(=O)rRB6、-S(=O)rNRA6RB6和-NRA6S(=O)2NRA6RB6,其中每个烷基是未被取代的或被至少一个独立选自RX6的取代基取代。在一实施方案中,R6选自氢、卤素、C1-10烷基、CN、NO2、-NRA6RB6和-ORA6,其中每个烷基是未被取代的或被至少一个独立选自RX6的取代基取代。在一特别的实施方案中,R6为氢。在另一特别的实施方案中,R6为F。
在一实施方案中,R7选自卤素、C1-10烷基、CN、NO2、-NRA7RB7、-ORA7、-SRA7、-C(=O)RA7、-C(=O)ORA7、-OC(=O)RA7、-C(=O)NRA7RB7、-NRA7C(=O)RB7、-OC(=O)NRA7RB7、-NRA7C(=O)ORB7、-NRA7C(=O)NRA7RB7、-NRA7C(=S)NRA7RB7、-S(=O)rRA7、-S(=O)2ORA7、-OS(=O)2RA7、-NRA7S(=O)rRB7、-S(=O)rNRA7RB7和-NRA7S(=O)2NRA7RB7,其中每个烷基是未被取代的或被至少一个独立选自RX7的取代基取代。在一实施方案中,R7选自卤素、C1-10烷基、CN、NO2、-NRA7RB7和-ORA7,其中每个烷基是未被取代的或被至少一个独立选自RX7的取代基取代。在一实施方案中,R7选自-NRA7RB7和-ORA7,优选-ORA7。在一特别的实施方案中,R7为-OH。
在一实施方案中,R8选自氢、卤素、C1-10烷基、CN、NO2、-NRA8RB8、-ORA8、-SRA8、-C(=O)RA8、-C(=O)ORA8、-OC(=O)RA8、-C(=O)NRA8RB8、-NRA8C(=O)RB8、-OC(=O)NRA8RB8、-NRA8C(=O)ORB8、-NRA8C(=O)NRA8RB8、-NRA8C(=S)NRA8RB8、-S(=O)rRA8、-S(=O)2ORA8、-OS(=O)2RA8、-NRA8S(=O)rRB8、-S(=O)rNRA8RB8和-NRA8S(=O)2NRA8RB8,其中每个烷基是未被取代的或被至少一个独立选自RX8的取代基取代。在一实施方案中,R8选自氢、卤素、C1-10烷基、CN、NO2、-NRA8RB8、-ORA8、-SRA8、-C(=O)RA8、-C(=O)ORA8、-OC(=O)RA8、-C(=O)NRA8RB8、-NRA8C(=O)RB8、-S(=O)rRA8、-S(=O)2ORA8、-OS(=O)2RA8、-NRA8S(=O)rRB8和-S(=O)rNRA8RB8,优选选自卤素、C1-10烷基、CN、NO2、-NRA8RB8和-ORA8,特别是选自C1-10烷基,其中每个烷基是未被取代的或被至少一个独立选自RX8的取代基取代。在一实施方案中,R8为氢。在一实施方案中,R8选自C1-10烷基。在一特别的实施方案中,R8为甲基。在一实施方案中,R8选自卤素,特别是Cl。
在一实施方案中,式(I)中的结构选自
特别是
在一实施方案中,R7和R8连同与它们相连的原子一起形成一个C5-6烃环或含1、2或3个杂原子的5-6元杂环或5-6元杂芳环,其中杂原子独立选自氧、硫和氮,该环是未被取代的或被至少一个独立选自RX3的取代基取代。在一实施方案中,R7和R8连同与它们相连的原子一起形成一个C5烃环或含1、2或3个杂原子的5元杂环或5元杂芳环,优选地形成一个含1、2或3个杂原子的5元杂环或5元杂芳环,其中杂原子独立选自氧、硫和氮,该环是未被取代的或被至少一个独立选自RX7的取代基取代。在一实施方案中,R7和R8连同与它们相连的原子一起形成吡唑环、异噻唑环、噻唑环、二氢异噁唑环或二氢噁唑环,优选吡唑环或异噻唑环,特别是吡唑环;该环是未被取代的或被至少一个独立选自RX7的取代基取代。
在一实施方案中,式(I)中的结构是在一实施方案中,式(I)中的结构选自
特别是
在一实施方案中,式(I)中的结构是
在一实施方案中,R7和R8连同与它们相连的原子成环,式(I)具有如下式(I-1)的结构:
其中,
X1选自N(R9a)、N、O、S、C(R9aR9a')和C(R9a);
X2选自N(R9b)、N、O、S、C(R9bR9b')和C(R9b);
X3选自N(R9c)、N、O、S、C(R9cR9c')和C(R9c);
R9a、R9a'、R9b、R9b'、R9c和R9c'各自独立选自氢和RX;
为单键或双键;
环A、R1、R2、R3、R4、R5、R6、E如式(I)中所定义。
在一实施方案中,X1选自S和N(R9a),优选为NH或N(CH3),特别是NH。在一实施方案中,X2选自N和O。在一实施方案中,X3选自C(R9cR9c')和C(R9c)。
在一实施方案中,X1为N(R9a),优选为NH或N(CH3),特别是NH;X2为N。
在一实施方案中,R9c选自氢和C1-10烷基,R9c',如果适用,选自氢和C1-10烷基;优选地,R9c为氢,R9c',如果适用,为氢。
在一实施方案中,式(I-1)中的结构选自
特别是在一实施方案中,式(I-1)中的结构是
在一实施方案中,R1选自-NRA1RB1和氢,特别是-NRA1RB1;R2选自-C(=O)NRA2RB2;R7选自-ORA7。在一实施方案中,R1选自-NH2和氢,特别是-NH2;R2为-C(=O)NH2;R7为-OH。
在一实施方案中,环A选自苯环、C5-6烃环和5-6元杂芳环。在一实施方案中,环A选自噻吩、噻唑、吡唑、咪唑、吡咯、呋喃、苯环、吡啶、环己烯和环戊烯,其被g个独立选自R3的取代基取代。
在一实施方案中,环A选自5元杂芳环和环戊烯,优选选自噻吩、噻唑、吡唑、咪唑、吡咯、呋喃和环戊烯,其被g个独立选自R3的取代基取代。在一实施方案中,环A为噻吩,其被g个独立选自R3的取代基取代。在一实施方案中,环A为噻唑,其被g个独立选自R3的取代基取代。在一实施方案中,环A为吡唑,其被g个独立选自R3的取代基取代。在一实施方案中,环A为咪唑,其被g个独立选自R3的取代基取代。在一实施方案中,环A为吡咯,其被g个独立选自R3的取代基取代。在一实施方案中,环A为呋喃,其被g个独立选自R3的取代基取代。
在一实施方案中,环A选自5元杂芳环和环戊烯,并且式(I)具有如下式(II)的结构:
其中,
A1选自N(R3a)、N、O、S、C(R3aR3a')和C(R3a);
A2选自N(R3b)、N、O、S、C(R3bR3b')和C(R3b);
A3选自N(R3c)、N、O、S、C(R3cR3c')和C(R3c);
R3a、R3a'、R3b、R3b'、R3c和R3c'各自独立选自氢和R3;
为单键或双键;
R1、R2、R3、R4、R5、R6、R7、R8、E如式(I)中所定义。
在一实施方案中,环A选自5元杂芳环。
在一实施方案中,环A选自噻吩、噻唑和咪唑,其中,A2为C(R3b);A1选自N(R3a)、N、S和C(R3a)并且A3选自N(R3c)、N、S和C(R3c)。在一实施方案中,环A为噻吩,其中,A2为C(R3b);A1为C(R3a)并且A3为S,或者A1为S并且A3为C(R3c)。在一实施方案中,环A为噻唑,A2为C(R3b);A1为N并且A3为S,或者A1为S并且A3为N。在一实施方案中,环A为咪唑,A2为C(R3b);A1为N并且A3为N。在一实施方案中,环A为环戊烯,A1为C(R3a),A2为C(R3b),A3为C(R3c)。在一实施方案中,R3b选自卤素、C1-10烷基、C3-10环烃基、杂环基、CN、杂芳基、-NRA3RB3、-C(=O)RA3和-C(=O)NRA3RB3,其中每个烷基、环烃基、杂环基和杂芳基是未被取代的或被至少一个独立选自RX3的取代基取代;R3a和R3c,假如适用,各自独立地选自氢和C1-10烷基;其中每个烷基是未被取代的或被至少一个独立选自RX3的取代基取代。在一实施方案中,R3a和R3c,假如适用,各自为氢。在一实施方案中,环A为噻吩,R3b选自卤素、C1-10烷基和C3-10环烷基;R3a和R3c,假如适用,各自独立地选自C1-10烷基;其中每个烷基、环烷基是未被取代的或被至少一个独立选自RX3的取代基取代。在一实施方案中,环A为吡唑,其中,A2为N;A1为N(R3a)并且A3为C(R3c),或者A1为C(R3a)并且A3为N(R3c)。在一实施方案中,R3a和R3c各自独立地选自氢和C1-10烷基;其中每个烷基是未被取代
的或被至少一个独立选自RX3的取代基取代。在一实施方案中,R3a和R3c中的一个选自C1-10烷基,另一个为氢。在一实施方案中,R3a和R3c各自独立地选自C1-10烷基。
在一实施方案中,R7和R8连同与它们相连的原子成环,式(II)具有如下式(II-1)的结构:
其中,
为单键或双键;
X1、X2、X3如式(I-1)中所定义;
A1、A2、A3、R1、R2、R4、R5、R6、E如式(II)中所定义。
在一实施方案中,式(II-1)的环A、R1、R2、R3、R4、R5、R6、E如式(I)中所定义。
在一实施方案中,A1选自N(R3a)、C(R3aR3a')和C(R3a),A2选自N(R3b)、C(R3bR3b')和C(R3b),并且R3a或R3a'与R3b或R3b'连同与它们相连的原子一起形成环Q,式(II)具有如下式(II-2)的结构:
其中,
为单键或双键;
Y1选自N和C;
Y2选自N和C;
环Q为C3-10单环烃环或含1、2或3个杂原子的4-12元杂环或含1、2或3个杂原子的5-10元杂芳环;其中杂原子独立选自氧、硫、氮和磷,优选选自氧、硫和氮;
h选自0、1、2、3、4、5和6;
A3、R1、R2、R4、R5、R6、R7、R8、RX3、E如式(II)中所定义。
在一实施方案中,环Q为C5-6单环烃环。在一实施方案中,环Q为含1、2或3个杂原子的5-6元杂环。在一实施方案中,环Q为含1、2或3个杂原子的5元杂环。在一实施方案中,环Q为含1或2个杂原子的6元杂芳环。
在一实施方案中,环Q选自
其中表示环Q与环A连接的位点。
在一实施方案中,环A选自苯环、环己烯和6元杂芳环,其被g个独立选自R3的取代基取代。在一实施方案中,6元杂芳环含有1至3个N原子,其它环原子为碳原子。在一实施方案中,环A选自苯环、吡啶和环己烯,特别是苯环。
在一实施方案中,环A选自苯环、环己烯和6元杂芳环,并且式(I)具有如下式(III)的结构:
其中,
A4选自N、C(R3dR3d')和C(R3d);
A5选自N、C(R3eR3e')和C(R3e);
A6选自N、C(R3fR3f')和C(R3f);
A7选自N、C(R3gR3g')和C(R3g);
R3d、R3d'、R3e、R3e'、R3f、R3f'、R3g和R3g'各自独立选自氢和R3;
为单键或双键;
R1、R2、R3、R4、R5、R6、R7、R8、E如式(I)中所定义。
在一实施方案中,A7为N,A4为C(R3d),A5为C(R3e),A6为C(R3f)。在一实施方案中A4为C(R3d),A5为C(R3e),A6为C(R3f),A7为C(R3g)。在一实施方案中A4为C(R3dR3d'),A5为C(R3eR3e'),A6为C(R3fR3f'),A7为C(R3gR3g')。
在一实施方案中,R3d、R3d'、R3e'、R3f'、R3g和R3g'为氢。在一实施方案中,R3d、R3d'、R3e、R3e'、R3f、R3f'、R3g和R3g'为氢。在一实施方案中,R3e和R3f中的一个选自卤素、C1-10烷基、C3-10环烃基、杂环基、CN、杂芳基、-NRA3RB3、-C(=O)RA3和-C(=O)NRA3RB3,其中烷基、环烃基、杂环基和杂芳基是未被取代的或被至少一个独立选自RX3的取代基取代;R3e和R3f中的另一个以及R3d、R3d'、R3e'、R3f'、R3g和R3g',假如适用,各自独立地选自氢和C1-10烷基;其中每个烷基是未被取代的或被至少一个独立选自RX3的取代基取代。在一实施方案中,R3e和R3f中的另一个以及R3d和R3g各自独立地选自氢和C1-10烷基;R3d'、R3e'、R3f'和R3g',假如适用,各自为氢。在另一实施方案中,R3e和R3f中的另一个以及R3d、R3d'、R3e'、R3f'、R3g和R3g',假如适用,各自为氢。在一实施方案中,R3e和R3f各自独立地选自C1-10烷基,其中烷基是未被取代的或被至少一个独立选自RX3的取代基取代;R3d、R3d'、R3e'、R3f'、R3g和R3g'为氢。
在一实施方案中,R7和R8连同与它们相连的原子成环,式(III)具有如下式(III-1)的结构:
其中,
为单键或双键;
X1、X2、X3如式(I-1)中所定义;
A4、A5、A6、A7、R1、R2、R4、R5、R6、E如式(III)中所定义。
在一实施方案中,式(III-1)的环A、R1、R2、R3、R4、R5、R6、E如式(I)中所定义。
在一实施方案中,A4选自C(R3dR3d')和C(R3d),A5选自C(R3eR3e')和C(R3e),并且R3d或R3d'与R3e或R3e'连同与它们相连的原子一起形成环Q,式(III)具有如下式(III-2)的结构:
其中,
为单键或双键;
环Q为C3-10单环烃环或含1、2或3个杂原子的4-12元杂环或含1、2或3个杂原子的5-10元杂芳环;其中杂原子独立选自氧、硫、氮和磷,优选选自氧、硫和氮;
h选自0、1、2、3、4、5、6、7和8;
A6、A7、R1、R2、R4、R5、R6、R7、R8、RX3、E如式(III)中所定义。
在一实施方案中,环Q为C5-6单环烃环。在一实施方案中,环Q为含1、2或3个杂原子的5-6元杂环。在一实施方案中,环Q为含1、2或3个杂原子的5元杂环。在一实施方案中,环Q为含1或2个杂原子的6元杂芳环。在一实施方案中,环Q选自
其中表示环Q与环A连接的位点。
在一实施方案中,本发明的化合物存在扭转异构体,其由包含环A的稠环与R4、R5、R6、R7和R8取代的苯基之间的单键旋转受阻而形成。在一实施方案中,式(I)具有如下式(Ia)或式(Ib)的结构。在一实施方案中,式(I-1)具有如下式(Ia-1)或式(Ib-1)的结构。在一实施方案中,式(II)具有如下式(IIa)或式(IIb)的结构。在一实施方案中,式(II-1)具有如下式(IIa-1)或式(IIb-1)的结构。在一实施方案中,式(III)具有如下式(IIIa)或式(IIIb)的结
构。在一实施方案中,式(III-1)具有如下式(IIIa-1)或式(IIIb-1)的结构。在一实施方案中,本发明的化合物具有式(Ia)、(Ia-1)、(IIa)、(IIa-1)、(IIIa)或(IIIa-1)的结构。在一实施方案中,本发明的化合物具有式(Ib)、(Ib-1)、(IIb)、(IIb-1)、(IIIb)或(IIIb-1)的结构。
在一实施方案中,本发明提供化合物或其药学上可接受的盐,其中化合物选自:
在一实施方案中,本发明提供化合物或其药学上可接受的盐,其中化合物选自:
在一实施方案中,本发明提供化合物或其药学上可接受的盐,其中化合物选自:
PROTAC化合物和抗体-药物缀合物(ADC)
在又一方面,本发明提供一种PROTAC化合物,其包含本发明的化合物、E3泛素连接酶结合配体,以及将本发明的化合物和E3泛素连接酶结合配体连接起来的接头。本发明的化合物可以作为配体,结合至PKMYT1。在一实施方案中,所述PROTAC化合物可以用于促进PKMYT1经UPS降解。
在又一方面,本发明提供一种抗体-药物缀合物,其包含本发明的化合物、能够与靶标结合的抗体,以及将本发明的化合物和抗体连接起来的接头。
药物组合物和药物制剂
本发明的另一目的是提供一种药物组合物,其包含本发明的化合物或其药学上可接受的盐、立体异构体、溶剂化物、多晶型、互变异构体、同位素化合物、代谢产物或前药,以及至少一种药学上接受的载体。
本发明的药物组合物可以以任何方式给药,只要其达到预防、缓解、预防或治疗人或动物症状的效果。例如,可根据给药途径制备各种合适的剂型。例如,可以以常规制剂形式口服或肠胃外给药至患者。所述常规制剂例如胶囊、微囊、片剂、颗粒剂、散剂、锭剂、丸剂、栓剂、注射剂、混悬剂、糖浆、贴剂、乳膏剂、洗剂、软膏剂、凝胶、喷雾剂、溶液和乳剂。
给予受试者的化合物的剂量可以在相当大的程度上进行调整。剂量可以根据具体的给药途径和受试者的需要而变化,并且可以经过医疗保健专业人员的判断。
治疗方法和用途
根据本发明的某些实施方案,可使用本发明的或其药学上可接受的盐、立体异构体、溶剂化物、多晶型、互变异构体、同位素化合物、代谢产物或前药或者本发明的药物组合物预防或治疗PKMYT1介导的疾病,或对PKMYT1的抑制有响应的疾病,包括但不限于细胞增殖异常疾病,例如癌症。
因此,在又一方面,本发明还提供本发明的化合物或其药学上可接受的盐或本发明的药物组合物在制备治疗疾病、病症或病况的药物中的用途,所述疾病、病症或病况选自细胞增殖异常疾病,所述化合物或其药学上可接受的盐或药物组合物任选地与第二治疗剂联合使用。
在另一方面,本发明提供本发明的化合物或其药学上可接受的盐或本发明的药物组合物,任选地与第二治疗剂联合,用于治疗细胞增殖异常疾病。
在进一步的方面,本发明提供一种治疗细胞增殖异常疾病的方法,所述方法包括向
有此需要的个体给药有效量的本发明的化合物或其药学上可接受的盐或本发明的药物组合物,所述化合物或其药学上可接受的盐或药物组合物任选地与第二治疗剂联合使用。
在一实施方案中,所述疾病、病症或病状选自癌性增殖性疾病(例如癌症)。
在一实施方案中,所述疾病、病症或病状为癌症,例如选自:(a)选自以下癌症的实体瘤或血液源性肿瘤:膀胱癌、子宫内膜癌、肺鳞状细胞癌、乳腺癌、结肠癌、肾癌、肝癌、肺癌、小细胞肺癌、食道癌、胆囊癌、脑癌、头颈癌、卵巢癌、胰腺癌、胃癌、宫颈癌、甲状腺癌、前列腺癌和皮肤癌;(b)选自以下的淋巴系的造血肿瘤:白血病、急性淋巴细胞白血病、急性成淋巴细胞白血病、B细胞淋巴瘤、T细胞淋巴瘤、霍奇金淋巴瘤、非霍奇金淋巴瘤、毛细胞淋巴瘤和伯克特淋巴瘤;(c)选自以下的骨髓系的造血肿瘤:急性和慢性骨髓性白血病、骨髓增生异常综合征和早幼粒细胞白血病;(d)选自纤维肉瘤和横纹肌肉瘤的间质来源的肿瘤;(e)选自星形细胞瘤、神经母细胞瘤、神经胶质瘤和神经鞘瘤的中枢和周围神经系统的肿瘤;或(f)黑色素瘤、精原细胞瘤、畸胎癌、骨肉瘤、着色性干皮病、角化棘皮瘤、甲状腺滤泡癌或卡波济氏肉瘤。
在一实施方案中,所述疾病、病症或病状选自CCNE1扩增的癌症。在一实施方案中,所述疾病、病症或病状选自子宫肉瘤、卵巢癌、乳腺癌、胃癌、食管癌、肺癌、肝癌和子宫内膜癌。
在一实施方案中,所述疾病、病症或病状选自FBXW7突变癌症,例如FBXW7基因发生失活突变的癌症。在一实施方案中,所述疾病、病症或病状选自血液肿瘤、神经胶质瘤、肝癌(例如肝细胞癌)、子宫癌(例如子宫内膜癌)、结直肠癌(例如结直肠腺癌)、乳腺癌、肺癌(例如非小细胞肺癌(NSCLC))、胃癌、食道癌、食管胃结合部腺癌、膀胱癌(例如膀胱尿路上皮癌)、头颈癌(例如头颈部鳞状细胞癌)、宫颈癌(例如宫颈鳞状细胞癌)、黑素瘤、卵巢癌(例如高级别浆液性卵巢癌)。
在一实施方案中,所述疾病、病症或病状选自TP53突变癌症,例如TP53基因发生失活突变的癌症。
在一特别的实施方案中,所述疾病、病症或病状为乳腺癌、卵巢癌。
联合疗法
本发明的化合物或其药学上可接受的盐或本发明的药物组合物可单独使用,或与其他治疗剂联合使用。
例如,使用佐剂(adjuvant)可增强本发明中的化合物的治疗效果(例如,单独使用辅佐药物的治疗性获益极小,但与另一种药物合用时,可增强个体的治疗性获益),或者,例如,本发明的化合物与另一个同样具有疗效的治疗剂合用可增强个体的治疗获益。例如,治疗癌症时,使用本发明的化合物时,合并使用另一种治疗癌症的药物,有可能会增强临床获益。可以联合的疗法包括但不仅限于物理疗法、心理疗法、放射疗法、化疗剂、小分子靶向治疗剂(例如激酶抑制剂、免疫激动剂)、免疫疗法(抗例如PD-1、抗PDL-1、CAR-T细胞)等。无论何种疾病、病症或病况,两种疗法使个体的治疗受益应具有加成效应或协同效应。
在一实施方案中,所述其他治疗剂选自WEE家族成员的抑制剂,例如WEE1抑制剂。在一实施方案中,其他治疗剂选自靶向DNA的癌症治疗剂,例如可以为直接破坏
DNA结构的治疗剂或DNA拓扑异构酶抑制剂。直接破坏DNA结构的治疗剂的实例包括但不限于DNA双链破坏剂(DNA double strand breaker)、DNA烷化剂、DNA嵌入剂(DNA intercalator)。
在一实施方案中,其他治疗剂选自WEE1抑制剂、FEN1(瓣结构特异性核酸内切酶1)抑制剂、TOP1(DNA拓扑异构酶I)抑制剂、RRM1(核糖核苷酸还原酶催化亚基M1)抑制剂、RRM2(核糖核苷酸还原酶调节亚基M2)抑制剂、AURKB(aurora激酶B)抑制剂、TOP2A(DNA拓扑异构酶IIα)抑制剂、ATR(毛细管扩张共济失调突变和RAD-3相关蛋白激酶)抑制剂、TTK(TTK蛋白激酶)抑制剂、SOD1(超氧化物歧化酶1)抑制剂、SOD2(超氧化物歧化酶2)抑制剂、BUB1(BUB1有丝分裂检查点丝氨酸/苏氨酸激酶B)抑制剂、CDC7(细胞分裂周期蛋白7)抑制剂、SAE1(SUMO1激活酶亚基1)抑制剂、PLK1(polo样激酶1)抑制剂、UBA2(泛素样修饰激活酶2)抑制剂、DUT(脱氧尿苷三磷酸酶)抑制剂、HDAC3(组蛋白脱乙酰基酶3)抑制剂、CHEK1(检查点激酶1)抑制剂、AURKA(aurora激酶A)抑制剂、menin抑制剂、DOT1L(DOT1样组蛋白赖氨酸甲基转移酶)抑制剂、CREBBP(CREB结合蛋白)抑制剂、EZH2(多梳抑制性复合体2亚单位zeste基因增强子人类同源物2)抑制剂、PLK4(polo样激酶4)抑制剂、HASPIN(组蛋白H3相关蛋白激酶)抑制剂、METTL3(甲基转移酶3)抑制剂、核苷类似物、基于铂的DNA破坏剂。
本发明提供的新型小分子PKMYT1抑制剂,显示突出的PKMYT1抑制活性和细胞增殖抑制活性。
因此,本发明的化合物能够预防或治疗PKMYT1介导的疾病,或对PKMYT1的抑制有响应的疾病,包括但不限于细胞增殖异常疾病,例如癌症,具有开发为药物的良好前景。
本发明的化合物的优势在于以下中至少一个:
(1)对靶标细胞有高抑制活性。
(2)优异的物理化学性质(例如溶解度、物理和/或化学稳定性)。
(3)优异的药物代谢动力学性质(例如在血浆中良好的稳定性、合适的半衰期和作用持续时间)。
(4)优异的安全性(对非靶标的正常细胞或组织较低的毒性和/或较少的副作用,较宽的治疗窗)等。
发明人还出人意料地发现,可以通过色谱手段获取本发明的至少一部分化合物的扭转异构体。分离得到的一些扭转异构体之间体现出活性差异。通过获取扭转异构体,进一步提高了本发明化合物的活性,并能实现上述优异性质。
实施例
为了更清楚地说明本发明的目的和技术方案,下面结合具体实施例对本发明作进一步的说明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未提及的具体实验方法,均按照常规实验方法进行。
仪器、材料与试剂
除非特别说明,实施例中所使用的仪器和试剂均为可商购的。试剂可以不经进一步纯化而直接使用。
LCMS:SHIMADZ LCMS2020,色谱柱:Waters SunFireTM C18 5μm 50*4.6mm;流动相A:H2O(含0.1%甲酸);流动相B:CH3CN(含0.1%甲酸)。
NMR:Bruker Avance III 400M。
合成方案
式(I)化合物或其药学上可接受的盐可由不同方法合成,一些示例性方法提供如下和实施例。其他合成方法可由本领域技术人员根据本发明披露的信息容易地提出。
在如下所述诸反应中可能有必要对活泼基团进行保护,以免这些活性基团参与其它不期望的反应:这些基团如羟基、氨基、亚胺基、含巯基或羧基,最终产物中含有这些基团。常用的保护基团可参考T.W.Greene and P.G.M.Wuts in"Protective Groups in Organic Chemistry"John Wiley and Sons,1991。
本发明的所有化合物的合成方案由以下方案和实施例加以说明。所用原料源于市售商品或可根据已有工艺方法或者此处示例的方法制备。
如合成方案1所示,式(I)化合物可由文献已知或本领域技术人员熟知的酰胺或硫代酰胺化合物M1和活性亚甲基试剂M2经由多种方法制备合成。其中M1中的w1为离去基团,例如选自卤素、OTf,特别是卤素。例如,M1和M2经取代反应或偶联反应得到中间体M3,中间体M3经缩合反应得到式(I)化合物。在一实施方案中,所述偶联反应为Ullmann反应或Buchwald反应。在一实施方案中,所述活性亚甲基试剂M2为丙二腈,所述缩合反应例如为分子内Thorpe-Ziegler环化。在一实施方案中,所述缩合反应在钯试剂存在下进行,例如可以使用Pd(dppf)Cl2。在一实施方案中,所述缩合反应在加热条件下进行。在一实施方案中,以下过程在一个合成步骤中完成:M1和M2反应得到M3、M3转化为式(I)化合物。在一实施方案中,所述合成步骤在碱存在下进行,例如醇钠或氢化钠,特别是氢化钠。在一实施方案中,所述合成步骤在醚类溶剂中进行,例如乙二醇二甲醚。
作为起始原料的酰胺或硫代酰胺化合物M1可以使用已知方法制备,例如采用与其相应的羧基化合物和氨基化合物经酸胺缩合得到目标酰胺,又例如采用与其相应的酰胺
化合物经硫代反应得到目标硫代酰胺。
式(I)化合物的一些取代基可以采用已知方法进行基团转换。例如,当M2为丙二腈,并且通过分子内Thorpe-Ziegler环化制备式(I)化合物时,Thorpe-Ziegler环化产物的氰基可以采用已知方法转化为酰胺基,例如水解。
在某些情况下,可以改变进行上述反应方案的顺序以促进反应或避免不需要的反应产物。提供以下实施例以便可以更充分地理解本发明。这些实施例仅是说明性的,不应解释为以任何方式限制本发明。
实施例1 6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-甲基-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-
7-甲酰胺(化合物001)
1.1中间体1-2的合成:
室温下将2,4-二溴噻唑-5-羧酸(1.00g)溶于1,4-二氧六环(10.00mL)中,依次加入3-甲氧基-2,6-二甲基苯胺(632mg)和丙基磷酸酐(2.22g),于70℃加热搅拌反应16小时。冷却至室温,向反应液中加入水(10.0mL)淬灭,通过乙酸乙酯(3x30.0mL)萃取,有机相经饱和食盐水(2x20.0mL)洗涤,过滤。滤液减压浓缩,残余物通过石油醚打浆纯化,得到中间体1-2(1.2g)。LCMS:MS m/z(ESI):420.9[M+H]+。
1.2中间体1-3的合成:
室温下将中间体1-2(900mg)溶于1,4-二氧六环(10.00mL)和水(1.00mL)中,依次加入2,4,6-三甲基硼氧环烷(405mg),碳酸钾(594mg)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(157mg),氩气保护下于120℃加热搅拌反应2小时。冷却至室温,直接用乙酸乙酯(3x30.0mL)萃取,有机相经饱和食盐水(2x20.0mL)洗涤,过滤。滤液减压浓缩,残余物通过柱层析(乙酸乙酯:石油醚=1:6)纯化分离,得到中间体1-3(350mg)。LCMS:MS m/z(ESI):355.0[M+H]+。
1.3中间体1-4的合成:
室温下将丙二腈(131mg)溶于乙二醇二甲醚(5.00mL)中,加入氢化钠(79mg),室温下搅拌反应0.5小时,加入中间体1-3(350mg)的乙二醇二甲醚(5.00mL)溶液和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(72mg),在氩气保护下于110℃加热搅拌反应2小时。冷却至室温,向反应液中加入水(40mL),用乙酸乙酯(3x20.0mL)萃取,有机相经饱和食盐水(2x10.0mL)洗涤,过滤。滤液减压浓缩,残余物通过薄层色谱法(乙酸乙酯:石油醚=2:1)纯化,得到中间体1-4(120mg)。LCMS:MS m/z(ESI):341.1[M+H]+。
1.4中间体1-5的合成
室温下将中间体1-4(120mg)溶于浓硫酸(1.00mL)中,于30℃加热搅拌反应1小时。反应液用二氯甲烷(10.0mL)稀释,用饱和碳酸氢钠溶液调节pH=7~8,经二氯甲烷(3x10.0mL)萃取,有机相用饱和食盐水(2x10.0mL)洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得到中间体1-5(90mg)。LCMS:MS m/z(ESI):359.1[M+H]+。
1.5化合物001的合成:
室温下将中间体1-5(90.0mg)溶于二氯甲烷(1.00mL)中,加入三溴化硼(1.0mL),于25℃搅拌反应1小时。将反应液减压浓缩,残余物直接用高效液相制备色谱法(色谱柱:Waters Xbridge C18 10um OBD 19*250mm;流动相A:H2O(0.1%甲酸),流动相B:CH3CN,A:B=95%-5%梯度变化)纯化,得到目标产物6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-甲基-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物001)(6.33mg)。
1H NMR(400MHz,DMSO)δ9.63(s,1H),9.24(d,J=3.4Hz,1H),7.39(d,J=3.4Hz,1H),7.09(d,J=8.3Hz,1H),6.89(d,J=8.3Hz,1H),2.83(s,3H),1.85(s,3H),1.77(s,3H).LCMS:MS m/z(ESI):345.1[M+H]+。
实施例26-氨基-5-(3-羟基-2,6-二甲基苯基)-2-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-
7-甲酰胺(化合物002)
2.1中间体2-2的合成:
将4-溴-2-甲基噻吩(1g)溶于乙醚(10mL)中,降温至-68℃。向反应液中缓慢加入正丁基锂(3.9mL,1.6mol/L),在-68℃下反应1小时,随后向反应液中通入二氧化碳气体,继续反应1小时。向反应液中加水(10mL)淬灭,用乙酸乙酯(3x10mL)萃取除去未反应的原料。剩余水相用稀盐酸调节pH至2~3,用乙酸乙酯(3x10mL)萃取,有机相用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩,得到中间体2-2(600mg)。1H NMR(400MHz,DMSO)δ7.99(d,J=1.2Hz,1H),7.13–7.10(m,1H),2.45(s,3H).
2.2中间体2-3的合成:
室温下将中间体2-2(600mg)溶于醋酸6mL)中,加入液溴(79mg),25℃反应3小时。向反应液中加入饱和碳酸氢钠水溶液至无气泡产生,用乙酸乙酯(3x10mL)萃取,有机相用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩,得到中间体2-3(500mg)。
LCMS:MS m/z(ESI):221.0[M+H]+。
2.3中间体2-4的合成:
室温下将中间体2-3(0.44g)溶于二氧六环(5mL)中,加入3-甲氧基-2,6-二甲基苯胺(0.36g)和丙基磷酸酐(1.27g),70℃下反应5小时。冷却至室温,将反应液直接减压浓缩,残余物通过柱层析(石油醚:乙酸乙酯=30:1-10:1)纯化,得到中间体2-4(270mg)。LCMS:MS m/z(ESI):354.0[M+H]+。
2.4中间体2-5的合成:
将丙二腈(118.8mg)溶于乙二醇二甲醚(4mL)中,加入氢化钠(80mg,60%),室温下搅拌反应0.5小时。加入中间体2-4(255mg),1,1-双(二苯基磷)二茂铁二氯化钯(52.7mg),在氮气保护下置换气三次,于100℃反应1.5小时。冷却至室温,向反应液中加入水(10.0mL),用乙酸乙酯(3x10.0mL)萃取,有机相经饱和食盐水(2x10.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物通过薄层色谱法(石油醚:乙酸乙酯=1:1)分离纯化,得到中间体2-5(100mg)。
LCMS:MS m/z(ESI):340.3[M+H]+。
2.5中间体2-6的合成:
室温下将中间体2-5(100mg)溶于浓硫酸(1.5mL)中,在40℃下搅拌反应1小时。降温至0℃,向反应液中加冰水(5mL)稀释反应液,有固体产生。过滤,滤饼真空干燥,得到中间体2-6(100mg,粗品)。LCMS:MS m/z(ESI):358.1[M+H]+。
2.6化合物002的合成:
室温下将中间体2-6(100mg)溶于二氯甲烷(2mL)中,降温到0℃,向反应液中滴加三溴化硼(1.4mL),在25℃下搅拌反应1小时。加入甲醇(2mL)淬灭,反应液减压浓缩,残余物通过高效液相制备色谱法(色谱柱:Sunfire Prep C18 OBD 10um 19*250mm;流动相A:H2O(0.1%甲酸),流动相B:CH3CN,A:B=80%-5%梯度变化)纯化,得到目标产物6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物002)(6.54mg)。
1H NMR(400MHz,DMSO)δ9.52(s,1H),7.12(s,2H),7.06(d,J=8.3Hz,1H),6.99(s,1H),6.94(s,2H),6.87(d,J=8.3Hz,1H),2.41(s,3H),1.83(s,3H),1.75(s,3H).
LCMS:MS m/z(ESI):344.0[M+H]+。
实施例3 5-氨基-6-(3-羟基-2,6-二甲基苯基)-2-甲基-7-氧代-6,7-二氢噻吩并[2,3-c]吡啶-
4-甲酰胺(化合物003)
3.1中间体3-2的合成:
室温下将N,N-二异丙基胺(373mg)溶于四氢呋喃(3mL)中,在-68℃下加入正丁基锂(1.6M,2.1mL),并保持-68℃搅拌1小时后,在-68℃下缓慢加入2-溴-5-甲基噻吩(500mg)的四氢呋喃(1.00mL)溶液,于-68℃搅拌反应2小时。在-68℃下通入二氧化碳气体,随后将反应体系升至室温,并在室温下反应1小时。向反应液中加入水(10.0mL)淬灭,通过乙酸乙酯(3x10.0mL)萃取除去未反应的原料,水相用盐酸调节pH=2~3,有白色固体析出。将固体过滤,滤饼干燥后得到中间体3-2(300mg)。
LCMS:MS m/z(ESI):220.9[M+H]+。
3.2中间体3-3的合成:
室温下将中间体3-2(250mg)溶于1,4-二氧六环(10.00mL)中,依次加入3-甲氧基-2,6-二甲基苯胺(258mg)和丙基磷酸酐(1.45g),反应体系于70℃加热搅拌反应16小时。冷却至室温,向反应液中加入水(10.0mL)淬灭,用乙酸乙酯(3x10.0mL)萃取,有机相经饱和食盐水(2x10.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物通过柱层析(乙酸乙酯:石油醚=1:10)纯化,得到中间体3-3(250mg)。
LCMS:MS m/z(ESI):354.0[M+H]+。
3.3中间体3-4的合成:
室温下将丙二腈(94mg)溶于乙二醇二甲醚(1.00mL)中,加入氢化钠(58mg,60%),室温下搅拌反应0.5小时后,加入中间体3-3(250mg)的乙二醇二甲醚(4.00mL)溶液和
[1,1'-双(二苯基膦基)二茂铁]二氯化钯(51mg),反应体系在氩气保护下于100℃加热搅拌反应2小时。向反应液中加入水(10.0mL),用乙酸乙酯(3x10.0mL)萃取,有机相经饱和食盐水(2x10.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物通过薄层色谱法(乙酸乙酯:石油醚=1:1)纯化,得到中间体3-4(110mg)。
LCMS:MS m/z(ESI):340.1[M+H]+。
3.4中间体3-5的合成:
室温下将中间体3-4(60mg)溶于浓硫酸(0.50mL)中,于40℃加热搅拌反应3小时。冷却至室温,反应液通过二氯甲烷(5.0mL)稀释,用饱和碳酸氢钠溶液调节pH=7~8,经二氯甲烷(3x5.0mL)萃取,有机相经饱和食盐水(2x5.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,得到中间体3-5(40mg)。
LCMS:MS m/z(ESI):358.1[M+H]+。
3.5化合物003的合成:
室温下将中间体3-5(40mg)溶于二氯甲烷(0.50mL),加入三溴化硼(1.0mL),于25℃搅拌反应1小时。冷却至0℃,用甲醇(2mL)淬灭,反应液减压浓缩,残余物用高效液相制备色谱法(流动相A:H2O(0.1%甲酸),流动相B:CH3CN,A:B=80%-5%梯度变化)纯化,得到目标产物5-氨基-6-(3-羟基-2,6-二甲基苯基)-2-甲基-7-氧代-6,7-二氢噻吩并[2,3-c]吡啶-4-甲酰胺(化合物003)(7.48mg)。
1H NMR(400MHz,DMSO)δ9.51(s,1H),7.25(s,1H),7.21(s,2H),7.06(d,J=8.2Hz,1H),6.90–6.79(m,3H),2.54(s,3H),1.84(s,3H),1.76(s,3H).
LCMS:MS m/z(ESI):344.1[M+H]+。
实施例4 6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-甲基-4-氧代-4,5-二氢噻唑[4,5-c]吡啶-7-
甲酰胺(化合物004)
4.1中间体4-2的合成:
室温下将2-甲基噻唑-4-羧酸(1.00g)溶于四氢呋喃(10.00mL)中,在-68℃下缓慢滴加正丁基锂(1.6M,10.9mL),并保持-68℃搅拌1小时。在-68℃下缓慢加入液溴(0.42mL),随后将反应体系升至室温,并在室温下反应2小时。向反应液中加入水(10.0mL),通过乙酸乙酯(3x10.0mL)萃取除去未反应的原料,水相用1N盐酸调节pH=2~3,然后用乙酸乙酯(3x10.0mL)萃取,有机相经饱和食盐水(2x10.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,得到中间体4-2(1.2g)。
LCMS:MS m/z(ESI):221.9[M+H]+。
4.2中间体4-3的合成:
室温下将中间体4-2(400mg)溶于1,4-二氧六环(10.00mL)中,依次加入3-甲氧基-2,6-二甲基苯胺(328mg)和丙基磷酸酐(864mg),于70℃加热搅拌反应16小时。冷却至室温,向反应液中加入水(10.0mL),用乙酸乙酯(3x10.0mL)萃取,有机相经饱和食盐水(2x10.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物通过柱层析(乙酸乙酯:石油醚=1:10~1:3)纯化,得到中间体4-3(600mg)。
LCMS:MS m/z(ESI):355.0[M+H]+。
4.3中间体4-4的合成:
室温下将丙二腈(56mg)溶于乙二醇二甲醚(1.00mL)中,加入氢化钠(34mg,60%),室温下搅拌反应0.5小时,再加入中间体4-3(150mg)的乙二醇二甲醚(4.00mL)溶液和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(31mg),反应体系在氩气保护下于100℃加热搅拌反应2小时。冷却至室温,向反应液中加入水(10.0mL),用乙酸乙酯(3x10.0mL)萃取,有机相经饱和食盐水(2x10.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物通过薄层色谱法(乙酸乙酯:石油醚=1:0)纯化,得到中间体4-4(80mg)。
LCMS:MS m/z(ESI):341.1[M+H]+。
4.4中间体4-5的合成:
室温下将中间体4-4(80mg)溶于浓硫酸(0.50mL)中,于30℃搅拌反应2小时。反应液通过二氯甲烷(5.0mL)稀释,通过饱和碳酸氢钠溶液调节pH=7~8,用二氯甲烷(3x5.0mL)萃取,有机相经饱和食盐水(2x5.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩得到中间体4-5(80mg)。
LCMS:MS m/z(ESI):359.1[M+H]+。
4.5化合物004的合成:
室温下将中间体4-5(80mg)溶于二氯甲烷(1.00mL)中,加入三溴化硼(1.0mL),于25℃搅拌反应1小时。反应体系加入甲醇(2mL)淬灭,减压浓缩,残余物用高效液相制备色谱法(流动相A:H2O(0.1%甲酸),流动相B:CH3CN,A:B=90%-5%梯度变化)纯化,得到目标产物6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-甲基-4-氧代-4,5-二氢噻唑[4,5-c]吡啶-7-甲酰胺(化合物004)(36.48mg)。
1H NMR(400MHz,DMSO)δ9.68(s,1H),7.18(s,2H),7.09(d,J=8.4Hz,1H),7.03(s,2H),6.90(d,J=8.4Hz,1H),2.64(s,3H),1.86(s,3H),1.77(s,3H).
LCMS:MS m/z(ESI):345.1[M+H]+。
实施例5(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-甲基-4-氧代-4,5-二氢噻唑并[5,4-c]吡
啶-7-甲酰胺(化合物001-1)和(R)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-甲基-4-氧代-
4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物001-2)
将化合物001通过SFC手性制备拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2流动相B:MeOH(含0.1%7.0mol/L Ammonia in MeOH),A:B=55:45),得到两个异构体:(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-甲基-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物001-1)和(R)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-甲基-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物001-2)。
化合物001-1(Rt=2.53min):
1H NMR(400MHz,MeOD)δ7.11(d,J=8.3Hz,1H),6.90(d,J=8.3Hz,1H),2.86(s,3H),
1.95(s,3H),1.91(s,3H).
LCMS:MS m/z(ESI):345.1[M+H]+。
化合物001-2(Rt=5.93min):
1H NMR(400MHz,MeOD)δ7.11(d,J=8.3Hz,1H),6.90(d,J=8.3Hz,1H),2.86(s,3H),1.95(s,3H),1.91(s,3H).
LCMS:MS m/z(ESI):345.1[M+H]+。
实施例6 6-氨基-5-(3-羟基-2,6-二甲基苯基)-2,3-二甲基-4-氧代-4,5-二氢-3H-咪唑并
[4,5-c]吡啶-7-甲酰胺(化合物006)
参考实施例2的合成方法,将中间体2-3替换为4-溴-1,2-二甲基-1H-咪唑-5-羧酸,得到终产物6-氨基-5-(3-羟基-2,6-二甲基苯基)-2,3-二甲基-4-氧代-4,5-二氢-3H-咪唑并[4,5-c]吡啶-7-甲酰胺(化合物006)。
1H NMR(400MHz,CDCl3)δ6.97(d,J=8.3Hz,1H),6.67(d,J=8.3Hz,1H),3.92(s,3H),2.47(s,3H),1.93(s,3H),1.86(s,3H).
LCMS:MS m/z(ESI):342.1[M+H]+。
实施例7 3-氨基-2-(3-羟基-2,6-二甲基苯基)-1-氧代-1,2-二氢异喹啉-4-甲酰胺(化合物
009)
7.1中间体9-2的合成:
将2-碘苯甲酰氯(1.00g)溶于二氯甲烷(10.00mL)中,0℃下依次加入三乙胺(1.14g)和3-甲氧基-2,6-二甲基苯胺(851.2mg),反应体系于25℃下搅拌反应16小时。向反应液中加入水(10.0mL),用乙酸乙酯(3x30.0mL)萃取,有机相经饱和食盐水(3x20.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物通过柱层析(乙酸乙酯:石油醚=1:10)纯化,得到中间体9-2(813mg)。LCMS:MS m/z(ESI):382.0[M+H]+。
7.2中间体9-3的合成:
室温下将中间体9-2(400mg)溶于二甲基亚砜(4.00mL),依次加入2-氰基乙酰胺(105.9mg),碳酸钾(290.0mg)和碘化亚铜(20.0mg),反应体系在氩气保护下于封管中85℃加热搅拌反应3小时。冷却至室温,向反应液中加入水(10.0mL),用乙酸乙酯(3x30.0mL)萃取,有机相经饱和食盐水(3x20.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物通过柱层析(乙酸乙酯:石油醚=1:2)纯化,得到中间体9-3(279mg)。LCMS:MS m/z(ESI):338.2[M+H]+。
7.3化合物009的合成:
室温下将中间体9-3(50.0mg)溶于二氯甲烷(1.00mL),于-68℃加入三溴化硼(0.44mL),升温至0℃搅拌反应1小时。将反应液重新冷却至-68℃,加入甲醇(3mL)淬灭,反应液直接减压浓缩,残余物用高效液相制备色谱法(流动相A:H2O(0.1%甲酸),流动相B:CH3CN,A:B=90%-5%梯度变化)纯化,得到目标产物3-氨基-2-(3-羟基-2,6-二甲基苯基)-1-氧代-1,2-二氢异喹啉-4-甲酰胺(化合物009)(17.45mg)。
1H NMR(400MHz,DMSO)δ9.54(s,1H),8.04(dd,J=8.1,1.5Hz,1H),7.86(d,J=8.4Hz,1H),7.63–7.54(m,1H),7.48(s,2H),7.18–7.10(m,1H),7.07(d,J=8.2Hz,1H),6.88(d,J=8.3Hz,1H),6.28(s,2H),1.85(s,3H),1.78(s,3H).
LCMS:MS m/z(ESI):324.2[M+H]+。
实施例8 3-氨基-2-(3-羟基-2,6-二甲基苯基)-1-氧代-1,2,5,6,7,8-六氢异喹啉-4-甲酰胺
(化合物010)
8.1中间体10-2的合成:
将2-氧代环己烷-1-甲酸甲酯(5g)溶于水(50mL)中,于0℃加入氢氧化钠(1.4g),0℃下反应3小时,升至室温继续反应16小时。反应液用浓盐酸调节pH=2,用乙醚(3x50mL)萃取,有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,得到中间体10-2(3.7g)。
1H NMR(400MHz,CDCl3)δ11.94(s,1H),2.34–2.24(m,4H),1.75–1.62(m,4H).
8.2中间体10-3的合成:
将中间体10-2(3.1g)溶于丙酮(3.1mL)中,在0℃下依次加入醋酸酐(4.5mL),浓硫酸(534mg),于0℃反应4h。反应液用10%碳酸氢钠水溶液(80mL)调节pH=8,用乙酸乙酯(3x50mL)萃取。有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,得到中间体10-3(3.1g)。
LCMS:MS m/z(ESI):183.1[M+H]+。
8.3中间体10-4的合成:
将中间体10-3(2g)溶于二甲苯(15mL)中,加入3-甲氧基-2,6-二甲基苯胺(2.4g),150℃下微波反应1.5小时。冷却至室温,加入水(40mL)稀释,用乙酸乙酯(3x40mL)萃取,饱和食盐水(40mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物经硅胶柱(石油醚:乙酸乙酯=80:1~10:1)纯化分离,得到中间体10-4(1.08g)。
LCMS:MS m/z(ESI):276.1[M+H]+。
8.4中间体10-5的合成:
将中间体10-4(200mg)溶于乙醇(2mL)中,依次加入丙二腈(95mg),哌啶(33.7mg),在氮气保护下100℃反应2小时。冷却至室温,反应液直接减压浓缩,残余物经薄层制备色谱(石油醚:乙酸乙酯=1.5:1)纯化分离,得到中间体10-5(230mg)。
LCMS:MS m/z(ESI):324.1[M+H]+。
8.5中间体10-6的合成:
将中间体10-5(230mg)溶于浓硫酸(1.5mL)中,于室温下反应1小时,升温至75℃下反应1h。反应液用饱和碳酸氢钠水溶液调节pH=8,用乙酸乙酯(3x10mL)萃取,有机相用饱和食盐水(10mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物经薄层制备色谱(石油醚:乙酸乙酯=0:1)纯化分离,得到中间体10-6(70mg)。
LCMS:MS m/z(ESI):342.1[M+H]+。
8.6化合物010的合成:
将中间体10-6(40mg)溶于二氯甲烷(2mL)中,于0℃下滴加三溴化硼(0.58mL),反应体系在25℃下搅拌60分钟。用饱和碳酸氢钠溶液淬灭,并调节pH=7。反应液减压浓缩,残余物溶于甲醇(5mL)中,过滤除去不溶物,滤饼用甲醇(3x5mL)洗涤,将滤液浓缩,残余物通过高效液相制备色谱法(流动相A:H2O(0.1%甲酸),流动相B:CH3CN,A:B=90%-5%梯度变化)纯化得到目标产物3-氨基-2-(3-羟基-2,6-二甲基苯基)-1-氧代-1,2,5,6,7,8-六氢异喹啉-4-甲酰胺(化合物010)(8.83mg)。
1H NMR(400MHz,DMSO)δ9.44(s,1H),7.30(s,2H),7.01(d,J=8.3Hz,1H),6.82(d,J=8.3Hz,1H),5.74(s,2H),2.69–2.64(m,2H),2.33–2.27(m,2H),1.82(s,3H),1.74(s,3H),1.66–1.61(m,4H).
LCMS:MS m/z(ESI):328.1[M+H]+。
实施例9 6-氨基-5-(3-羟基-2,6-二甲基苯基)-2,3-二甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡
啶-7-甲酰胺(化合物012)
9.1中间体12-2的合成:
冰盐浴下将亚硝酸叔丁酯(3.11g)溶于干燥的乙腈(50.00mL)中,在氩气保护下分三
批次加入溴化铜(7.85g),搅拌15分钟后,再分三批次加入2-氨基-4,5-二甲基噻吩-3-羧酸乙酯(5.00g),并保持冰盐浴搅拌2小时,升至室温,并在室温下反应过夜。反应液通过200~300目硅胶垫底抽滤,滤饼用二氯甲烷和乙酸乙酯洗涤,滤液减压浓缩,残余物通过柱层析(乙酸乙酯:石油醚=0:1~1:100)纯化分离,得到中间体12-2(4.7g)。LCMS:MS m/z(ESI):263.0[M+H]+。
9.2中间体12-3的合成:
室温下将中间体12-2(4.70g)溶于乙醇(40.00mL)中,在氩气保护下加入氢氧化钠(1.43g),反应体系于70℃加热搅拌反应过夜。冷却至室温,将反应溶剂浓缩后,加入水(50.0mL)稀释,用1N盐酸调节pH=3~4,有固体析出,过滤,滤饼用1%盐酸洗涤,真空干燥,得到中间体12-3(4.0g)。LCMS:MS m/z(ESI):233.1[M-H]。
9.3中间体12-4的合成:
在氩气保护下将中间体12-3(1.2g,5.10mmol)溶于氯化亚砜(15.00mL)中,于80℃油浴中加热搅拌反应75分钟。冷却至室温,将反应液浓缩,残余物溶于干燥的甲苯(5.0mL)中,并在冰浴下滴加到3-甲氧基-2,6-二甲基苯胺(1.16g)的吡啶(5mL)溶液中,滴加完毕后,将反应体系升至室温,并在氩气保护下反应过夜。减压浓缩,加入水(30.0mL)稀释,用二氯甲烷(3x20.0mL)萃取,有机相经饱和食盐水(2x20.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物通过柱层析(乙酸乙酯:石油醚=1:30~1:10)纯化分离,得到中间体12-4(900mg)。LCMS:MS m/z(ESI):368.0[M+H]+。
9.4化合物012的合成:
其余步骤参考实施例2,将中间体2-4替换为中间体12-4,得到化合物012。
1H NMR(400MHz,DMSO)δ9.50(s,1H),7.12(s,2H),7.06(d,J=8.4Hz,1H),6.88–6.86(m,3H),2.30(s,3H),2.28(s,3H),1.85(s,3H),1.77(s,3H)。
LCMS:MS m/z(ESI):358.1[M+H]+。
实施例10(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2,3-二甲基-4-氧代-4,5-二氢噻吩并
[3,2-c]吡啶-7-甲酰胺(化合物012-1)和(R)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2,3-二甲
基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物012-2)
将化合物012通过SFC手性制备拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2流动相B:MeOH(含0.1%7.0mol/L Ammonia in MeOH),A:B=60:40),得到两个异构体:(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2,3-二甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物012-1,Rt=2.56min)和(R)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2,3-二甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物012-2,Rt=4.20min)。
化合物012-1:
1H NMR(400MHz,DMSO)δ:9.52(s,1H),7.13(s,2H),7.05(d,J=8.3Hz,1H),6.88-6.85(m,3H),2.29(s,3H),2.28(s,3H),1.84(s,3H),1.76(s,3H)。LCMS:MS m/z(ESI):358.2[M+H]+。
化合物012-2:
1H NMR(400MHz,DMSO)δ:9.52(s,1H),7.13(s,2H),7.05(d,J=8.3Hz,1H),6.88-6.85(m,3H),2.29(s,3H),2.28(s,3H),1.84(s,3H),1.76(s,3H)。LCMS:MS m/z(ESI):358.2[M+H]+。
实施例11 6-氨基-2-溴-5-(3-羟基-2,6-二甲基苯基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡
啶-7-甲酰胺(化合物007)
11.1中间体7-2的合成:
室温下将5-溴噻唑-2-胺(10.00g)溶于二氯甲烷(100.0mL)中,依次加入三乙胺(14.1g)、4-二甲氨基吡啶(682.4mg)和二碳酸二叔丁酯(14.6g),于25℃加热搅拌反应2小时。反应液加入水(100.0mL)稀释,用二氯甲烷(3×200.0mL)萃取,有机相合并后经饱和食盐水(3×500.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物通过柱层析(乙酸乙酯:石油醚=20:1)纯化,得到中间体7-2(13.8g)。LCMS:MS m/z(ESI):224.9[M+H-55]+。
11.2中间体7-3的合成:
室温下将正丁基锂(2M,36.0mL)溶于四氢呋喃(50.00mL),于-68℃下加入中间体7-2(5g)的四氢呋喃溶液(50mL),于-68℃下反应2小时。-68℃下通入二氧化碳,反应液缓慢恢复至25℃并在25℃下反应2小时。向反应液中加水(100.0mL)淬灭,用乙酸乙酯(3×100.0mL)反萃取,水相用1N盐酸调节pH至6~7,有大量白色固体析出。过滤,滤饼真空干燥,得到中间体7-3(2.50g)。
11.3中间体7-4的合成:
室温下将中间体7-3(450mg)溶于1,4-二氧六环(4.00mL)中,依次加入3-甲氧基-2,6-二甲基苯胺(254mg)和丙基磷酸酐(1.34g),于70℃油浴中加热搅拌反应16小时。冷却至室温,向反应液中加入水(5.0mL)淬灭,用乙酸乙酯(3×10.0mL)萃取,有机相经饱和食盐水(3×30.0mL)洗涤,过滤,减压浓缩,残余物通过柱层析(乙酸乙酯:石油醚=1:10~1:3)纯化,得到中间体7-4(332mg)。LCMS:MS m/z(ESI):456.1[M+H]+。
11.4中间体7-5的合成:
室温下将丙二腈(96.1mg)溶于乙二醇二甲醚(1.00mL)中,加入氢化钠(58.2mg),室温下搅拌反应0.5小时后,加入中间体7-4(332mg)的乙二醇二甲醚(3.00mL)溶液和[1,1'-双(二苯基膦)二茂铁]二氯化钯(60.0mg),在氩气保护下于100℃加热搅拌反应2小时。冷却至室温,加入水(10.0mL)淬灭,用乙酸乙酯(3×10.0mL)萃取,有机相经饱和食盐水(2×10.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物通过柱层析(乙酸乙酯:石油醚=2:1~1:1)纯化,得到中间体7-5(227mg)。LCMS:MS m/z(ESI):442.2[M+H]+。
11.5中间体7-6的合成:
室温下将中间体7-5(100mg)溶于二氯甲烷(1.00mL)中,加入三氟乙酸(0.3mL),室
温下搅拌反应1小时,加入饱和碳酸钠溶液淬灭,调节pH=8~9,用乙酸乙酯(3×5.0mL)萃取,有机相经饱和食盐水(3×10.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,得到中间体7-6(69.7mg)。LCMS:MS m/z(ESI):342.1[M+H]+。
11.6中间体7-7的合成:
冰盐浴下将亚硝酸叔丁酯(60.4mg)溶于干燥乙腈(5.00mL)中,在氩气保护下分批加入溴化铜(130.9mg),搅拌15分钟后,再分批加入中间体7-6(200mg),并保持冰盐浴搅拌2小时,随后升至室温,并在室温下反应过夜。反应液通过200~300目硅胶垫底抽滤,滤饼用二氯甲烷(10.0mL)和乙酸乙酯(10.0mL)洗涤,滤液减压浓缩,残余物通过硅胶柱层析(乙酸乙酯:石油醚=0:1~1:100)分离纯化,得到中间体7-7(107mg)。LCMS:MS m/z(ESI):405.0[M+H]+。
11.7中间体7-8的合成:
将中间体7-7(75mg)溶于硫酸(1.5mL)中,在25℃下搅拌2小时。向反应液置于冰浴中,缓慢滴加饱和碳酸钠溶液淬灭,并调节pH=8。用二氯甲烷(10mLx3)萃取,饱和食盐水(10mL)洗涤,无水硫酸钠干燥后,过滤,滤液减压浓缩,得中间体7-8粗品(39mg)。LCMS:MS m/z(ESI):423.0[M+H]+。
11.8化合物007的合成:
室温下中间体7-8(37mg)溶于二氯甲烷(1.00mL)中,加入三溴化硼(0.43mL),于25℃搅拌反应1小时。冷却至-78℃,反应液加甲醇(3mL)淬灭,反应液减压浓缩,残余物用高效液相制备色谱法纯化,得到目标产物6-氨基-2-溴-5-(3-羟基-2,6-二甲基苯基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物007)(5.72mg)。
1H NMR(400MHz,DMSO)δ9.71(s,1H),8.70(s,1H),7.56(s,1H),7.18(d,J=8.4Hz,1H),6.99(d,J=8.4Hz,1H),1.94(s,3H),1.86(s,3H).
LCMS:MS m/z(ESI):409.0[M+H]+。
实施例12 3-氨基-2-(3-羟基-2,6-二甲基苯基)-1-氧代-1,2,6,7,8,9-六氢苯并[4,5]噻吩并
[3,2-c]吡啶-4-甲酰胺(化合物013)
参考实施例9,将步骤9.1中的2-氨基-4,5-二甲基噻吩-3-羧酸乙酯替换为2-氨基-4,5,6,7-四氢苯并[b]噻吩-3-羧酸乙酯,最终得到3-氨基-2-(3-羟基-2,6-二甲基苯基)-1-氧代-1,2,6,7,8,9-六氢苯并[4,5]噻吩并[3,2-c]吡啶-4-甲酰胺(化合物013)。
1H NMR(400MHz,DMSO)δ:9.59(s,1H),7.18(s,2H),7.13(d,J=8.2Hz,1H),6.98(s,2H),6.93(d,J=8.2Hz,1H),2.87(brs,2H),2.73(brs,2H),1.92(s,3H),1.89 -1.81(m,7H).LCMS:MS m/z(ESI):384.2[M+H]+。
实施例13(S)-3-氨基-2-(3-羟基-2,6-二甲基苯基)-1-氧代-1,2,6,7,8,9-六氢苯并[4,5]噻吩
并[3,2-c]吡啶-4-甲酰胺(化合物013-1)和(R)-3-氨基-2-(3-羟基-2,6-二甲基苯基)-1-氧代
-1,2,6,7,8,9-六氢苯并[4,5]噻吩并[3,2-c]吡啶-4-甲酰胺(化合物013-2)
将化合物013通过SFC手性制备拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2流动相B:MeOH(含0.1%7.0mol/L Ammonia in MeOH),A:B=65:35),得到两个异构体(S)-3-氨基-2-(3-羟基-2,6-二甲基苯基)-1-氧代-1,2,6,7,8,9-六氢苯并[4,5]噻吩并[3,2-c]吡啶-4-甲酰胺(化合物013-1,Rt=4.2min)和(R)-3-氨基-2-(3-羟基-2,6-二甲基苯基)-1-氧代-1,2,6,7,8,9-六氢苯并[4,5]噻吩并[3,2-c]吡啶-4-甲酰胺(化合物013-2,Rt=6.5min)。
化合物013-1:
1H NMR(400MHz,DMSO)δ9.54(s,1H),7.12(s,2H),7.06(d,J=8.2Hz,1H),6.92(s,2H),6.87(d,J=8.2Hz,1H),2.83–2.78(m,2H),2.69–2.64(m,2H),1.85(s,3H),1.80–1.67(m,7H).LCMS:MS m/z(ESI):384.2[M+H]+
化合物013-2:
1H NMR(400MHz,DMSO)δ:9.53(s,1H),7.11(s,2H),7.05(d,J=8.2Hz,1H),6.91(s,2H),6.88(d,J=8.2Hz,1H),2.83-2.80(m,2H),2.67 -2.65(m,2H),1.84(s,3H),1.80 -1.69(m,7H).LCMS:MS m/z(ESI):384.2[M+H]+.
实施例14 5-氨基-6-(3-羟基-2,6-二甲基苯基)-2,3-二甲基-7-氧代-6,7-二氢噻吩并[2,3-c]
吡啶-4-甲酰胺(化合物014)
14.1中间体14-2的合成:
将2,3-二甲基噻吩(5g)溶于N,N-二甲基甲酰胺(50mL)中,加入N-溴代琥珀酰亚胺(27.78g),于70℃反应16小时。冷却至室温,反应液中加入水(100mL),用乙酸乙酯(100mLx3)萃取,经饱和食盐水(100mLx3)洗涤,无水硫酸钠干燥后过滤,减压浓缩,残余物用硅胶柱分离纯化,得到中间体14-2(1.3g)。1H NMR(400MHz,CDCl3)δ:2.33(s,3H),2.15(s,3H).
14.2中间体14-3的合成:
将乙醚(13mL)置于三口瓶中,在氮气保护下置换三次,降温至-68℃,加入正丁基锂溶液(6mL,1.6M),然后缓慢加入含有中间体14-2(1.3g)的乙醚溶液(6mL),于-68℃反应30分钟。在二氧化碳保护下置换气三次,于-68℃反应30分钟,然后升温至室温。反应液加水(0.5mL)淬灭反应,用10%氢氧化钠水溶液(30mLx3)洗涤,合并水相,水相用6N的盐酸调节PH至2,有白色固体析出,过滤,滤饼真空干燥,得中间体14-3(390mg)。1H NMR(400MHz,CDCl3)δ:2.46(s,3H),2.19(s,3H).
其余步骤参考实施例3,得到化合物5-氨基-6-(3-羟基-2,6-二甲基苯基)-2,3-二甲基-7-氧代-6,7-二氢噻吩并[2,3-c]吡啶-4-甲酰胺(化合物014)。
1H NMR(400MHz,DMSO)δ9.51(s,1H),7.62(s,1H),7.45(s,1H),7.05(d,J=8.2Hz,1H),6.86(d,J=8.2Hz,1H),5.48(s,2H),2.41(s,3H),2.17(s,3H),1.84(s,3H),1.76(s,3H).LCMS:MS m/z(ESI):358.1[M+H]+。
实施例15 6-氨基-5-(3-羟基-2,6-二甲基苯基)-4-氧代-2-(吡啶-3-基)-4,5-二氢噻吩并[3,2-
c]吡啶-7-甲酰胺(化合物016)
15.1中间体16-2的合成:
将5-溴噻吩-3-羧酸(1g)溶于二氯亚砜(5mL)中,于75℃反应2小时。将反应液减压浓缩至干,加入甲苯(5mL),减压浓缩,得到粗品放置待用。将3-甲氧基-2,6-二甲基苯胺(942.8mg)溶于吡啶(5mL)中,在氮气保护下,冷却至0℃后加入上述中间体,然后升至室温反应16小时。反应液减压浓缩,加入水(30mL)稀释,用乙酸乙酯(30mLx3)萃取,有机相经饱和食盐水(30mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物经硅胶柱(石油醚:乙酸乙酯=30:1~10:1)分离纯化,得到中间体16-2(0.97g)。LCMS:MS m/z(ESI):340.0[M+H]+。
15.2中间体16-3的合成:
将中间体16-2(0.6g)溶于二氧六环(6mL)和水(0.6mL)中,依次加入吡啶-3-基硼酸(435.6mg),碳酸钾(489.2mg),1,1-双(二苯基磷)二茂铁二氯化钯(129.5mg),在氮气保护下置换三次,于100℃反应16小时。冷却至室温,减压浓缩,残余物经硅胶柱层析(石油醚:乙酸乙酯=30:1~1:1)分离纯化,得到中间体16-3(550mg)。LCMS:MS m/z(ESI):339.1[M+H]+。
15.3中间体16-4的合成:
将中间体16-3(540mg)溶于N,N-二甲基甲酰胺(8mL)中,加入N-溴代琥珀酰亚胺
(312.6mg),于25℃反应16小时。加入水(20mL)稀释,用乙酸乙酯(3×200.0mL)萃取,合并有机相经饱和食盐水(20mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物经硅胶柱(石油醚:乙酸乙酯=10:1~1:1)分离纯化,得到中间体16-4(80mg)。LCMS:MS m/z(ESI):417.1[M+H]+。
其余步骤参考实施例2,将中间体2-4替换为16-4,得到目标产物6-氨基-5-(3-羟基-2,6-二甲基苯基)-4-氧代-2-(吡啶-3-基)-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物016)。
1H NMR(400MHz,DMSO)δ9.87(s,1H),8.95(s,1H),8.55(d,J=4.8Hz,1H),8.47(s,1H),8.22(s,1H),8.13(dd,J=8.2,1.9Hz,1H),7.62(s,2H),7.51–7.47(m,1H),7.45(s,1H),6.80(s,2H),2.24(s,3H),2.22(s,3H).
LCMS:MS m/z(ESI):407.1[M+H]+。
实施例16 6-氨基-5-(3-羟基-2,6-二甲基苯基)-4-氧代-2-(三氟甲基)-4,5-二氢噻吩并[3,2-
c]吡啶-7-甲酰胺(化合物017)
16.1中间体17-2的合成:
在氩气保护下将2-溴噻吩-3-羧酸(1.0g)溶于氯化亚砜(10.0mL)中,反应体系于80℃油浴中加热搅拌反应2小时。将反应溶剂浓缩后,溶于甲苯(10.0mL)中,并在冰水浴下滴加到3-((4-甲氧基苄基氧基)-2,6-二甲基苯胺(1.37g)的吡啶(10.0mL)溶液中,滴加完毕后,将反应体系恢复室温,并在氩气保护下反应过夜。将反应溶剂浓缩后,加入水(20.0mL)稀释,用二氯甲烷(3×20.0mL)萃取,有机相经饱和食盐水(2×20.0mL)洗涤,无水硫酸钠干燥,减压浓缩。残余物经硅胶柱层(乙酸乙酯:二氯甲烷=0:1~1:100)纯化,得到中间体17-2(1.5g)。LCMS:MS m/z(ESI):446[M+H]+。
16.2中间体17-3的合成:
室温下将丙二腈(264mg)溶于乙二醇二甲醚(10.00mL)中,加入氢化钠(160mg),室温下搅拌反应10分钟后,再加入中间体17-2(900mg)的乙二醇二甲醚(5.00mL)溶液和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(146mg),反应体系在氩气保护下于100℃油浴中加热搅拌反应1.5小时。向反应液中加入水(10.0mL),用乙酸乙酯(3×10.0mL)萃取,有机相经饱和食盐水(2×10.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩。残余物经硅胶柱层(乙酸乙酯:二氯甲烷=0:1~1:50)纯化,得到中间体17-3(800mg)。LCMS:MS m/z(ESI):432.1[M+H]+。
16.3中间体17-4的合成:
室温下将中间体17-3(400mg)溶于N,N-二甲基甲酰胺(6.00mL)中,加入N-碘代丁二酰亚胺(229mg),室温下搅拌反应过夜。向反应液中加入水(10.0mL)稀释,用乙酸乙酯(3×10.0mL)萃取,有机相经饱和食盐水(4×10.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物用乙酸乙酯(5.00mL)打浆,过滤,滤饼用乙酸乙酯(3×5.0mL)洗涤,真空干燥后得到中间体17-4(340mg)。LCMS:MS m/z(ESI):558.1[M+H]+。
16.4中间体17-5的合成:
室温下将氟化钾(6mg)和碘化亚铜(21mg)置于微波管中,用氮气吹扫后,在60℃油浴中搅拌反应1小时。再加入三甲基三氟甲基硅烷(120mg)的N-甲基吡咯烷酮(0.50mL)溶液,反应体系在50℃油浴中搅拌反应45分钟。再加入中间体17-4(20mg)的N,N-二甲基甲酰胺(0.50mL)溶液,反应体系在80℃油浴中搅拌反应过夜。向反应液中加入水(5.0mL)稀释,用乙酸乙酯(3×5.0mL)萃取,有机相经饱和食盐水(3×5.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩后,残余物用Prep-TLC(乙酸乙酯:石油醚=1:2)纯化,得到中间体17-5(5mg)。
16.5化合物017的合成:
室温下将中间体17-5(5mg)溶于浓硫酸(0.50mL)中,反应体系在40℃搅拌反应3小时。停止反应,向反应液中加入水(5.00mL)稀释,用乙酸乙酯(3×5.0mL)萃取,有机相经饱和食盐水(2×5.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物用高效液相制备色谱法纯化,得到目标产物6-氨基-5-(3-羟基-2,6-二甲基苯基)-4-氧代-2-(三氟甲基)-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物017)。
1H NMR(400MHz,DMSO)δ:9.61(s,1H),7.77(s,1H),7.38(s,2H),7.08(d,J=8.4Hz,1H),6.89(d,J=8.4Hz,1H),1.86(s,3H),1.78(s,3H)。
LCMS:MS m/z(ESI):398.1[M+H]+。
实施例17 6-氨基-2-环丙基-5-(3-羟基-2,6-二甲基苯基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡
啶-7-甲酰胺(化合物021)
参考实施例15,将步骤15.2中的吡啶-3-基硼酸替换为环丙基三氟硼酸钾,得到化合物021。
1H NMR(400MHz,DMSO)δ9.53(s,1H),7.13(s,2H),7.06(d,J=8.3Hz,1H),6.95(s,1H),6.93(s,2H),6.87(d,J=8.3Hz,1H),2.11–2.02(m,1H),1.83(s,3H),1.75(s,3H),1.02–0.93(m,2H),0.77–0.69(m,2H).
LCMS:MS m/z(ESI):370.1[M+H]+。
实施例18 2-乙酰基-6-氨基-5-(3-羟基-2,6-二甲基苯基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡
啶-7-甲酰胺(化合物023)
18.1中间体23-2的合成:
室温下将中间体17-3(600mg)溶于乙腈(12.00mL)中,加入N-溴代琥珀酰亚胺(272mg),室温下搅拌反应2小时后,减压浓缩,残余物用二氯甲烷(10.00mL)打浆纯化,过滤,滤饼用二氯甲烷(3×10.0mL)洗涤,真空干燥,得到中间体23-2(600mg)。LCMS:MS m/z(ESI):510.1[M+H]+。
18.2中间体23-3的合成:
在氩气保护下将中间体23-2(250mg)溶于N,N-二甲基甲酰胺(3.00mL)中,加入三丁基(1-乙氧基乙烯基)锡(265mg)和XPhos-Pd-G2(39mg),反应体系用氩气置换3次后,在氩气保护下于100℃加热搅拌反应2小时。将反应液倒入水(20.0mL)中,用乙酸乙酯(3×20.0mL)萃取,有机相经饱和食盐水(2×20.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物通过薄层色谱(石油醚:乙酸乙酯=1:1)纯化,得到中间体23-3(95mg)。LCMS:MS m/z(ESI):502.2[M+H]+。
18.3化合物023的合成:
室温下将中间体23-3(95mg)溶于浓硫酸(2.0mL)中,于50℃加热搅拌反应1小时。冷却至室温后,向反应液中加入乙酸乙酯(10.00mL)稀释,在冰水浴下通过饱和碳酸氢钠溶液中和,用乙酸乙酯(3×10.0mL)萃取,有机相经饱和食盐水(2×10.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,过滤,残余物用高效液相制备色谱法纯化,得到目标产物2-乙酰基-6-氨基-5-(3-羟基-2,6-二甲基苯基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物023)(9.06mg)。
1H NMR(400MHz,DMSO)δ:9.61(s,1H),8.12(s,1H),7.36(s,4H),7.09(d,J=8.0Hz,1H),6.90(d,J=8.0Hz,1H),2.53(s,3H),1.86(s,3H),1.78(s,3H)。
LCMS:MS m/z(ESI):372.1[M+H]+。
实施例19 6-氨基-5-(3-羟基-2,6-二甲基苯基)-4-氧代-2-(噻唑-2-基)-4,5-二氢噻吩并[3,2-
c]吡啶-7-甲酰胺(化合物024)
19.1中间体24-2的合成:
室温下将中间体23-2(200mg)溶于N,N-二甲基甲酰胺(20.00mL)中,依次加入2-(三甲基锡基)噻唑(291mg)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(29mg),反应体系在氩气保护下于80℃加热搅拌反应2小时。冷却至室温,加水(10.0mL)稀释,用乙酸乙酯(3×10.0mL)萃取,有机相经饱和食盐水(2×10.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物通过薄层色谱法(乙酸乙酯:石油醚=1:1)纯化分离,得到中间体24-2(20mg)。LCMS:MS m/z(ESI):515.2[M+H]+。
其余步骤参考实施例18,将中间体23-3替换为24-2,得到目标产物6-氨基-5-(3-羟基-2,6-二甲基苯基)-4-氧代-2-(噻唑-2-基)-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物024)。
1H NMR(400MHz,DMSO)δ:9.60(s,1H),7.81-7.71(m,3H),7.34(s,2H),7.23-7.08(m,3H),6.97-6.89(m,1H),1.87(s,3H),1.79(s,3H)。
LCMS:MS m/z(ESI):413.1[M+H]+。
实施例20 6-氨基-2-甲基-5-(5-甲基-1H-吲唑-4-基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-
甲酰胺(化合物036)
参考实施例1,将步骤1.1中的3-甲氧基-2,6-二甲基苯胺,替换为5-甲基-1H-吲唑-4-胺,得到目标化合物6-氨基-2-甲基-5-(5-甲基-1H-吲唑-4-基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物036)。
1H NMR(400MHz,DMSO)δ13.24(s,1H),9.25(s,1H),8.11(s,1H),7.82(s,1H),7.57(s,1H),7.37(s,1H),2.84(s,3H),2.03(s,3H).
LCMS:MS m/z(ESI):355.1[M+H]+。
实施例21 5-氨基-6-(3-羟基-2,6-二甲基苯基)-1,2-二甲基-7-氧代-6,7-二氢-1H-吡咯并
[2,3-c]吡啶-4-甲酰胺(化合物043)
21.1中间体43-2的合成:
室温下将2-氨基丙二酸二乙酯(10.00g)溶于甲醇(200.00mL)中,加入3-氨基丁-2-烯腈(27.08g),于25℃油浴中加热搅拌反应5小时。反应液减压浓缩,用石油醚(200.0mL)打浆,过滤,滤饼用石油醚(3×100.0mL)洗涤,干燥,得到中间体43-2(25g)。LCMS:MS m/z(ESI):241.2[M+H]+。
21.2中间体43-3的合成:
室温下将中间体43-2(10g)溶于乙醇(100mL),在氩气保护下加入乙醇钠的乙醇溶液(0.5N,100mL),随后升温至60℃反应16小时。向反应液加水(100.0mL)淬灭,用乙酸乙酯(3×100.0mL)萃取,有机相经饱和食盐水(2×100.0mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,用石油醚(50.0mL)和乙酸乙酯(50.0mL)打浆,过滤,滤饼真空干燥,得到中间体43-3(3.2g)。LCMS:MS m/z(ESI):169.1[M+H]+。
21.3中间体43-4的合成:
室温下将中间体43-3(4.8g)溶于水(48mL)中,加入溴化氢(7.68mL),冷却至-5℃下,将亚硝酸钠(5.9g)的水(4.8mL)溶液缓慢滴加入混合溶液中,并在-5℃下反应30分钟,加入溴化亚铜(12.3g),升温至室温下搅拌30分钟,随后于油浴中升温至100℃搅拌反应2小时。冷却至室温,反应液用乙酸乙酯(3×100.0mL)萃取,有机相经饱和食盐水(2×100.0mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,残余物通过硅胶柱层析(石油醚:
乙酸乙酯=20:1~5:1)分离纯化,得到中间体43-4(925mg)。LCMS:MS m/z(ESI):232.0[M+H]+。
21.4中间体43-5的合成:
室温下将中间体43-4(900.0mg)溶于四氢呋喃(9.00mL)中,降温至0℃,加入氢化钠(232.6mg),搅拌30分钟后加入碘甲烷(825.6mg),于50℃油浴中加热搅拌反应16小时。冷却至室温,反应液中加入水(20.0mL),用乙酸乙酯(3×20.0mL)萃取,有机相经饱和食盐水(2×20.0mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,残余物通过硅胶柱层析(石油醚:乙酸乙酯=50:1~10:1)分离纯化,得到中间体43-5(825mg)。LCMS:MS m/z(ESI):246.1[M+H]+。
21.5中间体43-6的合成:
室温下将中间体43-5(350mg)和3-甲氧基-2,6-二甲基苯胺(322mg)溶于二氧六环(3.50mL)中,加入三甲基铝(8.88mL),在氩气保护下,于封管中120℃加热搅拌反应3小时。冷却至室温,将反应液加入到甲醇(10.0mL)中淬灭,加入水(10.0mL)稀释,用二氯甲烷(3×10.0mL)萃取,有机相经饱和食盐水(2×10.0mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,残余物通过硅胶柱层析分离纯化,得到中间体43-6(349mg)。LCMS:MS m/z(ESI):351.0[M+H]+。
21.6化合物043的合成:
其余步骤参考实施例1,将中间体1-3替换为中间体43-6,最终得到目标化合物5-氨基-6-(3-羟基-2,6-二甲基苯基)-1,2-二甲基-7-氧代-6,7-二氢-1H-吡咯并[2,3-c]吡啶-4-甲酰胺(化合物043)。
1H NMR(400MHz,DMSO)δ9.45(s,1H),7.04(d,J=8.2Hz,1H),6.88–6.82(m,3H),6.77(s,2H),6.27(s,1H),3.87(s,3H),2.30(s,3H),1.84(s,3H),1.76(s,3H).
LCMS:MS m/z(ESI):341.1[M+H]+。
实施例22 6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-(甲硫基)-4-氧代-4,5-二氢噻吩并[3,2-c]
吡啶-7-甲酰胺(化合物044)
22.1中间体44-2的合成:
将2,4-二溴噻吩(10g)溶于乙醚(10mL)中,在-68℃缓慢加入正丁基锂(25.8mL,1.6M),搅拌0.5小时,在-68℃下缓慢加入二甲基二硫醚(4.28g)的四氢呋喃(5mL)溶液,继续搅拌0.5小时后,升温至0℃搅拌反应1小时。向反应液中缓慢加入饱和氯化铵溶液(100mL)淬灭,用乙酸乙酯(3×100mL)萃取,有机相用饱和食盐水(3×100mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩。残余物用硅胶柱层析(EA/PE:0%~5%)分离纯化,得到中间体44-2(8.1g)。1H NMR(400MHz,CDCl3)δ:7.17(d,J=1.2Hz,1H),6.95(d,J=1.2Hz,1H),2.48(s,3H).
22.2中间体44-3的合成:
将中间体44-2(4.30g)溶于乙醚(10mL),冷却至-68℃缓慢加入正丁基锂(12.0mL,1.6M),在-68℃搅拌0.5小时,用二氧化碳置换三次。升温至0℃下搅拌反应1小时。缓慢加入饱和氯化铵(40mL)溶液淬灭,用盐酸(1M)调节pH=1,用乙酸乙酯(3×40mL)萃取。有机相经饱和食盐水(3×50mL)洗涤,无水硫酸钠干燥,减压浓缩。残余物用硅胶柱层析(EA/PE:0%~20%)分离纯化,得到中间体44-3(2.1g)。1H NMR(400MHz,CDCl3)δ:8.16(d,J=1.2Hz,1H),7.49(d,J=1.2Hz,1H),2.52(s,3H).
22.3中间体44-4的合成:
将中间体44-3(1.0g)溶于N,N-二甲基甲酰胺(10mL)中,依次加入3-((4-甲氧基苄基)氧基)-2,6-二甲基苯胺(1.77g),N,N-二异丙基乙胺(1.85g)和HATU(2.42g),升温到40℃反应2小时。冷却至室温,向反应液中加水(10mL),用乙酸乙酯(3×10mL)萃取,有机相用饱和食盐水(3×10mL)洗涤,无水硫酸钠干燥,减压浓缩。残余物用硅胶柱层析(EA/PE:0%~33%)分离纯化,得到中间体44-4(2.1g)。LCMS:MS m/z(ESI):414.2[M+H]+。
22.4中间体44-5的合成:
将中间体44-4(500mg)溶于乙腈(5.0mL)中,加入N-溴代丁二酰亚胺(193.7mg),25℃下反应1小时。向反应液加水(10mL),用乙酸乙酯(3×10mL)萃取,有机相用饱和食盐水(3×10mL)洗涤,无水硫酸钠干燥,减压浓缩。残余物用硅胶柱层析(EA/PE:0%~33%)分离纯化,得到中间体44-5(416mg)。LCMS:MS m/z(ESI):492.1[M+H]+。
22.5中间体44-6的合成:
室温下将丙二腈(98mg)溶于乙二醇二甲醚(1.00mL)中,加入氢化钠(60mg),室温下搅拌反应0.5小时后,再加入中间体44-5(366mg)的乙二醇二甲醚溶液(3mL)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(54mg),反应体系在氩气保护下于80℃油浴中加热搅拌反应1h。加入水(5.0mL)稀释,用乙酸乙酯(3×5.0mL)萃取,有机相经饱和食盐水(2×10.0mL)洗涤、无水硫酸钠干燥、减压浓缩后,残余物用硅胶柱层析分离纯化,得到中间体44-6(108mg)。LCMS:MS m/z(ESI):478.2[M+H]+。
22.6化合物044的合成:
其余步骤参考实施例28,将中间体50-3替换为44-6,得到目标产物6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-(甲硫基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物044)。
1H NMR(400MHz,DMSO)δ9.59(s,1H),7.27(s,1H),7.21(s,2H),7.08(d,J=8.2Hz,1H),6.99(s,2H),6.89(d,J=8.2Hz,1H),2.51(s,3H),1.85(s,3H),1.77(s,3H).
LCMS:MS m/z(ESI):376.1[M+H]+。
实施例23 6-氨基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶
-7-甲酰胺(化合物045)
23.1中间体45-2的合成:
-68℃下将二异丙基氨基锂(36.9ml,2M)溶于四氢呋喃(10ml)中,缓慢加入4-甲基噻吩-3-羧酸(5g)和四氢呋喃(40ml)的混合液,于-68℃搅拌0.5h,缓慢滴加四溴化碳(12.8g)的四氢呋喃(10ml)溶液,在-68℃搅拌30分钟后,将反应液加热到25℃搅拌反应2小时。用盐酸(1M)调节pH至1,用乙酸乙酯(3×100ml)萃取,有机相用饱和盐水(100ml)洗涤,无水硫酸钠干燥后浓缩。残余物采用柱层析(EA/PE=0%~5%)纯化,得到中间体45-2(3g)。1H NMR:(400MHz,DMSO)δ7.24(s,2H),2.29(s,3H).
23.2中间体45-3的合成:
将中间体45-2(200mg)溶于1,4-二氧六环(2ml),再依次加入3-甲氧基-2,6-二甲基苯胺(178mg)和1-丙基磷酸环酐(869mg),将反应液加热到100℃反应16小时。反应液冷却到室温,向反应液加水(5ml)淬灭,用乙酸乙酯(3×10ml)萃取,有机相用饱和盐水(10ml)洗涤,无水硫酸钠干燥。采用柱层析(EA/PE=0%~33%)纯化,得到化中间体45-3(236mg)。LCMS:MS m/z(ESI):354.0[M+H]+。
23.3中间体45-4的合成:
将丙二腈(88mg)溶于乙二醇二甲醚(1ml),加入氢化钠(32mg),在室温搅拌30分钟,再依次加入中间体45-3(236mg)的乙二醇二甲醚溶液(3ml),以及1,1'-双二苯基膦二茂铁二氯化钯(49mg),用氩气置换三次,加热至100℃反应2小时。向反应液加水(5ml),用乙酸乙酯(3×10ml)萃取,有机相用饱和盐水(10ml)洗涤,无水硫酸钠干燥,减压浓缩。残余物经硅胶柱层(EA/PE=0%~50%)分离纯化,得到中间体45-4(80mg)。LCMS:MS m/z(ESI):340.1[M+H]+。
23.4中间体45-5的合成:
将中间体45-4(76mg)溶于浓硫酸(2ml)中,在25℃下反应2小时。在0℃下加入乙酸乙酯(2ml)稀释,用饱和的碳酸钠水溶液调节pH=7,用乙酸乙酯(3×10ml)萃取,有机
相用饱和盐水(10ml)洗涤,无水硫酸钠干燥,减压浓缩,残余物经硅胶柱(EA/PE=0%~50%)分离纯化,得到中间体45-5(50mg)。LCMS:MS m/z(ESI):358.1[M+H]+。
23.5化合物045的合成:
将中间体45-5(50mg)溶于二氯甲烷(2ml)中,在0℃下加入三溴化硼(2ml),25℃下反应2小时。反应液冷却到0℃下,加入甲醇(0.5ml)淬灭,反应液减压浓缩,残余物用用高效液相制备色谱法纯化,得到化合物6-氨基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物045)(7.69mg)。
1H NMR(400MHz,DMSO)δ9.52(s,1H),7.15(s,2H),7.06(d,J=8.2Hz,1H),6.96(s,2H),6.87(d,J=8.2Hz,1H),6.80(d,J=1.3Hz,1H),2.38(d,J=1.3Hz,3H),1.85(s,3H),1.77(s,3H).
LCMS:MS m/z(ESI):344.1[M+H]+。
实施例24 6-氨基-2-环丙基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并
[3,2-c]吡啶-7-甲酰胺(化合物046)、(S)-6-氨基-2-环丙基-5-(3-羟基-2,6-二甲基苯基)-3-
甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物046-1)和(R)-6-氨基-2-环丙基
-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物
046-2)
24.1中间体46-2的合成:
将中间体45-4(500mg)溶于乙腈(5mL),加入N-溴代丁二酰亚胺(235mg)。在25℃下反应2小时。反应液中有大量固体析出,过滤,滤饼用乙腈洗涤三遍,滤饼真空干燥,得到中间体46-2(450mg)。LCMS:MS m/z(ESI):418.0[M+H]+。
24.2中间体46-3的合成:
将中间体46-2(200mg)溶于二氧六环(5mL)和水(0.5mL)中,加入环丙基三氟硼酸钾(355mg),碳酸钾(132.6mg),[1,1'-二(二苯基膦)二茂铁]二氯化钯(35mg),在氮气保护下置换气三次,于100℃反应16小时。反应液加水(20mL),用乙酸乙酯(20mLx3)萃取,有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,减压浓缩。残余物经Prep-TLC(石油醚/乙酸乙酯=2/1)分离纯化,得到中间体46-3(150mg)。LCMS:MS m/z(ESI):380.2[M+H]+。
24.3中间体46-4的合成:
将中间体46-3(200mg)溶于二氯甲烷(4mL)中,加入三溴化硼(2.1mL),于25℃反应1小时。反应液于0℃加入1mL甲醇淬灭,减压浓缩,得到粗品,加入甲醇(3mL)打浆过滤,滤饼真空干燥,得到中间体46-4(80mg)。LCMS:MS m/z(ESI):366.2[M+H]+。
24.4化合物046的合成:
将中间体46-4(80mg)溶于乙醇(6mL)和水(2mL)中,加入(二甲基膦酸)铂(II)氢化络合物(18mg),在氮气保护下置换气三次,于60℃反应4小时。反应液减压浓缩,残余物通过高效液相制备色谱法纯化,得到化合物6-氨基-2-环丙基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物046)(10.88mg)。
1H NMR(400MHz,DMSO)δ9.54(s,1H),7.12(s,2H),7.05(d,J=8.2Hz,1H),6.89(s,2H),6.86(d,J=8.2Hz,1H),2.40(s,3H),2.05–1.94(m,1H),1.84(s,3H),1.76(s,3H),1.03–0.94(m,2H),0.68–0.60(m,2H).
LCMS:MS m/z(ESI):384.1[M+H]+。
24.5化合物046-1和046-2:
将化合物046通过SFC手性制备拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2,流动相B:IPA(含0.1%7.0mol/L Ammonia in MeOH),A:B=70:30),得到两个异构体(S)-6-氨基-2-环丙基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物046-1,Rt=12.17min)和(R)-6-氨基-2-环丙基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物046-2,Rt=15.02min)。
化合物046-1:
1H NMR(400MHz,DMSO)δ9.49(s,1H),7.11(s,2H),7.05(d,J=8.2Hz,1H),6.88(s,2H),6.86(d,J=8.2Hz,1H),2.41(s,3H),2.05–1.94(m,1H),1.84(s,3H),1.76(s,3H),1.03–0.94(m,2H),0.69–0.60(m,2H).LCMS:MS m/z(ESI):384.1[M+H]+。
化合物046-2:
1H NMR(400MHz,DMSO)δ9.49(s,1H),7.11(s,2H),7.05(d,J=8.0Hz,1H),6.93–6.82(m,3H),2.40(s,3H),2.02–1.97(m,1H),1.84(s,3H),1.76(s,3H),1.01–0.95(m,2H),0.67–0.61(m,2H).LCMS:MS m/z(ESI):384.1[M+H]+。
实施例25(S)-6-氨基-2-乙基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并
[3,2-c]吡啶-7-甲酰胺(化合物047-1)、(R)-6-氨基-2-乙基-5-(3-羟基-2,6-二甲基苯基)-3-
甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物047-2)和6-氨基-2-乙基-5-
(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物
047)
25.1中间体47-2的合成:
将中间体46-2(300mg)溶于1,4-二氧六环(5mL)和水(0.5mL)中,依次加入乙烯三氟
硼酸钾(116mg)、碳酸钾(199mg)和1,1'-双二苯基膦二茂铁二氯化钯(53mg)用氩气置换三次,将反应液加热到120℃搅拌反应7小时。冷却至室温,向反应液中加入水(10mL)稀释,用乙酸乙酯(3×10mL)萃取,有机相用饱和盐水(10mL)洗涤,无水硫酸钠干燥,减压浓缩。残余物用硅胶柱层析(EA/PE=0%~50%)纯化分离,得到中间体47-2(80mg)。LCMS:MS m/z(ESI):366.1[M+H]+。
25.2中间体47-3的合成:
将中间体47-2(80mg)溶于甲醇(10mL)中,再加入湿钯碳催化剂(233mg),用氢气置换三次,在25℃下反应1.5小时。反应液用硅藻土过滤,滤液减压浓缩,得到中间体47-3(60mg)。LCMS:MS m/z(ESI):368.1[M+H]+。
25.3中间体47-4的合成:
将中间体47-3(614mg)溶于乙醇:水=3:1的混合溶剂中(14mL),加入(二甲基膦酸)铂(II)氢化络合物(72mg),在氮气保护下置换气三次,于100℃反应16小时。将反应液减压浓缩,残余物通过柱层析(乙酸乙酯:石油醚=1:30~1:4)分离纯化,得到中间体47-4(479.97mg)。LCMS:MS m/z(ESI):386.2[M+H]+。
25.4化合物047的合成
将中间体47-4(60mg)溶于二氯甲烷(1mL)中,加入三溴化硼(1mL)。在25℃下反应1小时。加入甲醇(2mL)淬灭,反应液减压浓缩,残余物用高效液相制备色谱法纯化,得到6-氨基-2-乙基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物047)(7.36mg)。
1H NMR(400MHz,DMSO)δ9.53(s,1H),7.13(s,2H),7.06(d,J=8.3Hz,1H),6.91–6.84(m,3H),2.71(q,J=7.5Hz,2H),2.32(s,3H),1.85(s,3H),1.77(s,3H),1.20(t,J=7.5Hz,3H).LCMS:MS m/z(ESI):372.1[M+H]+。
25.5中间体47-4的手性分离:
将中间体47-4通过SFC手性拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in MeOH),A:B=70:30),得到两个异构体47-4-1(Rt=4.9min)和47-4-2(Rt=7.2min)。
25.6化合物047-1和化合物047-2的合成:
剩余脱甲基方法参考上述步骤25.4,将中间体47-4-1和47-4-2分别脱甲基后,得到(S)-6-氨基-2-乙基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物047-1)和(R)-6-氨基-2-乙基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物047-2)。
化合物047-1:
1H NMR(400MHz,DMSO-d6)δ9.49(s,1H),7.13(s,2H),7.05(d,J=8.2Hz,1H),6.88(s,2H),6.86(d,J=8.2Hz,1H),2.71(q,J=7.5Hz,2H),2.31(s,3H),1.85(s,3H),1.77(s,3H),1.20(t,J=7.5Hz,3H).LCMS:MS m/z(ESI):372.1[M+H]+。
化合物047-2:
1H NMR(400MHz,DMSO-d6)δ9.50(s,1H),7.13(s,2H),7.05(d,J=8.2Hz,1H),6.90–6.85(m,3H),2.70(q,J=7.5Hz,2H),2.31(s,3H),1.85(s,3H),1.77(s,3H),1.20(t,J=7.5Hz,3H).LCMS:MS m/z(ESI):372.1[M+H]+。
实施例26 6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-异丙基-3-甲基-4-氧代-4,5-二氢噻吩并
[3,2-c]吡啶-7-甲酰胺和(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-异丙基-3-甲基-4-氧代-
4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物048-1)
参考实施例25,将步骤25.1中的乙烯三氟硼酸钾替换为4,4,5,5-四甲基-2-(丙-1-烯-2-基)-1,3,2-二氧杂硼烷,得到化合物6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-异丙基-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物048)。通过SFC手性拆分,得到化合物(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-异丙基-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]
吡啶-7-甲酰胺(化合物048-1)。
1H NMR(400MHz,DMSO-d6)δ9.50(s,1H),7.14(s,2H),7.06(d,J=8.2Hz,1H),6.95–6.83(m,3H),2.35(s,3H),2.11–2.05(m,1H),1.85(s,3H),1.77(s,3H),1.27(d,J=6.1Hz,6H).LCMS:MS m/z(ESI):386.2[M+H]+。
实施例27 6-氨基-2-氯-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]
吡啶-7-甲酰胺(化合物049)和(S)-6-氨基-2-氯-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧
代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物049-1)
27.1中间体49-2的合成:
将中间体45-4(400mg)溶于四氢呋喃(4mL)中,加入N-氯代琥珀酰亚胺(158mg),加热到80℃搅拌反应16小时。冷却至室温,加水(10mL)淬灭,用乙酸乙酯(3×20mL)萃取,有机相用饱和盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用二氯甲烷(10mL)打浆,过滤。所得滤饼真空干燥,得中间体49-2(168mg)。LCMS:MS m/z(ESI):374.1[M+H]+。
27.2化合物049-1的合成:
剩余步骤参考实施例23,得到6-氨基-2-氯-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物049)。
将化合物049通过SFC手性拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in MeOH),A:B=65:35),得到(S)-6-氨基-2-氯-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物049-1,Rt=3.5min)。
1H NMR(400MHz,DMSO-d6)δ9.56(s,1H),7.24(s,2H),7.07(d,J=8.3Hz,1H),6.88(d,J=8.3Hz,1H),6.73(s,2H),2.34(s,3H),1.86(s,3H),1.78(s,3H).LCMS:MS m/z(ESI):378.0[M+H]+。
实施例28 6-氨基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-2-(甲硫基)-4-氧代-4,5-二氢噻吩并
[3,2-c]吡啶-7-甲酰胺(化合物050)、(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-2-(甲
硫基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物050-1)和(R)-6-氨基-5-(3-羟
基-2,6-二甲基苯基)-3-甲基-2-(甲硫基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化
合物050-2)
28.1中间体50-2的合成:
将中间体44-6(200mg)溶于乙腈(10.0mL),加入N-溴代丁二酰亚胺(82mg),室温反应1小时。向反应液加水(10mL),用乙酸乙酯(3×10mL)萃取,有机相经饱和食盐水(3×10mL)洗涤,无水硫酸钠干燥,减压浓缩。残余物用反相柱层析(ACN in H2O:0%~100%)分离纯化,得到中间体50-2(145mg)。LCMS:MS m/z(ESI):556.1[M+H]+。
28.2中间体50-3的合成:
将中间体50-2(145mg)溶于二氧六环(2mL)和水(0.2mL)中,加入2,4,6-三甲基-1,3,5,2,4,6-三氧杂三硼杂环己烷(0.22mL),1,1'-双二苯基膦二茂铁二氯化钯(54mg)和碳酸钾(72mg),用氩气置换三次,将反应液加热到100℃反应16小时。冷却至室温,向反应液加水(5mL)稀释,用乙酸乙酯(3×10mL)萃取,有机相用饱和盐水(10mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩。残余物用硅胶柱层析分离纯化,得到化中间体50-3(87mg)。LCMS:MS m/z(ESI):492.2[M+H]+。
28.3中间体50-4的合成:
室温下将中间体50-3(77mg)溶于二氯甲烷(1.0mL)中,加入三氟乙酸(0.10mL),室温下搅拌反应1小时。向反应液中加入饱和碳酸氢钠溶液调节pH至7,用二氯甲烷(3×5.0mL)萃取,有机相经饱和食盐水(2×5.0mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,残余物通过石油醚(5.0mL)打浆纯化,得到中间体50-4(52mg)。LCMS:MS m/z(ESI):372.1[M+H]+。
28.4化合物050的合成:
室温下将中间体50-4(52mg)溶于乙醇(3.0mL)和水(1.0mL)的混合溶液中,加入(二甲基膦酸)铂(II)氢化络合物(3.3mg),用氩气置换3次后,反应体系在氩气保护下于50℃油浴中搅拌反应3小时。将反应液减压浓缩,残余物通过高效液相制备色谱法纯化,得到6-氨基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-2-(甲硫基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物050)(5.66mg)。1H NMR(400MHz,DMSO)δ9.53(s,1H),7.21(s,2H),7.07(d,J=8.2Hz,1H),6.97(s,2H),6.87(d,J=8.2Hz,1H),2.45(s,3H),2.35(s,3H),1.85(s,3H),1.77(s,3H).LCMS:MS m/z(ESI):390.1[M+H]+。
28.5化合物050-1和050-2:
将化合物050通过手性SFC拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in MeOH),A:B=75:25),得到两个异构体:(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-2-(甲硫基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物050-1,Rt=5.88min)和(R)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-2-(甲硫基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物050-2,Rt=8.72min)。
化合物050-1:
1H NMR(400MHz,DMSO)δ9.56(s,1H),7.21(s,2H),7.07(d,J=8.2Hz,1H),6.97(s,2H),6.87(d,J=8.2Hz,1H),2.45(s,3H),2.36(s,3H),1.86(s,3H),1.77(s,3H).LCMS:MS m/z(ESI):390.1[M+H]+。
化合物050-2:
1H NMR(400MHz,DMSO)δ9.56(s,1H),7.21(s,2H),7.07(d,J=8.2Hz,1H),6.97(s,2H),6.88(d,J=8.2Hz,1H),2.45(s,3H),2.36(s,3H),1.86(s,3H),1.77(s,3H).LCMS:MS m/z(ESI):390.1[M+H]+。
实施例29 6-氨基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-2-(三氟甲基)-4,5-二氢噻吩
并[3,2-c]吡啶-7-甲酰胺(化合物051)和(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-
4-氧代-2-(三氟甲基)-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物051-1)
29.1中间体51-2的合成:
室温下将中间体45-4(200mg)溶于乙腈(4.0mL)中,加入1-(三氟甲基)-1,2-苯碘酰-3(1H)-酮(559mg)和碳酸钾(122mg),反应液用氩气置换三次,80℃下反应12小时。向反应液中加水(10.0mL),用乙酸乙酯(3×10.0mL)萃取,有机相经饱和食盐水(2×10.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物用Prep-TLC(乙酸乙酯:石油醚=2:1)分离纯化,得到中间体51-2(60mg)。LCMS:MS m/z(ESI):408.1[M+H]+。
29.2目标化合物51-3的合成:
室温下将中间体51-2(60mg)溶于二氯甲烷(1.0mL)中,在0℃下加入三溴化硼(0.2mL),反应液在25℃下反应1小时。在冰浴下加入饱和碳酸氢钠溶液(5.0mL)淬灭,用乙酸乙酯(3×5.0mL)萃取。有机相经饱和氯化钠溶液(2×5.0mL)洗涤,无水硫酸钠干燥,减压浓缩,得到中间体51-3(40mg)粗品。LCMS:MS m/z(ESI):394.1[M+H]+。
29.3化合物051的合成:
室温下将中间体51-3(40mg)溶于乙醇(1.5mL)和水(0.5mL)中,加入(二甲基膦酸)铂(II)氢化络合物(4mg),反应液在100℃下反应2小时。反应液减压浓缩,残余物通过高
效液相制备色谱法纯化,得到6-氨基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-2-(三氟甲基)-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物051)(19.87mg)。
1H NMR(400MHz,DMSO)δ9.56(s,1H),7.33(s,2H),7.08(d,J=8.2Hz,1H),6.97(s,2H),6.89(d,J=8.2Hz,1H),2.56–2.50(m,3H),1.87(s,3H),1.79(s,3H).LCMS:MS m/z(ESI):412.1[M+H]+。
29.4化合物051-1:
将化合物051通过SFC手性制备拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2,流动相B:MeOH(含0.1%7.0mol/L Ammonia in MeOH),A:B=85:15),得到化合物(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-3-甲基-4-氧代-2-(三氟甲基)-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物051-1,Rt=3.90min)。
1H NMR(400MHz,DMSO)δ9.56(s,1H),7.34(s,2H),7.08(d,J=8.2Hz,1H),6.96(s,2H),6.89(d,J=8.2Hz,1H),2.56–2.50(m,3H),1.87(s,3H),1.79(s,3H).LCMS:MS m/z(ESI):412.1[M+H]+。
实施例30(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-异丙基-4-氧代-4,5-二氢噻唑并[5,4-
c]吡啶-7-甲酰胺(化合物052-1)和(R)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-异丙基-4-
氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物052-2)
30.1中间体52-2的合成:
室温下将中间体1-2(1.00g)溶于1,4-二氧六环(10.00mL)和水(1.00mL)中,依次加入异丙烯基硼酸频哪醇酯(400mg),碳酸钾(658mg)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯二氯甲烷络合物(193mg),反应体系在氩气保护下于100℃油浴中加热反应4小时。冷却至室温,向反应液中加入水(10.0mL),用乙酸乙酯(3×10.0mL)萃取,有机相经饱和食盐水(2×20.0mL)洗涤,减压浓缩,残余物通过硅胶柱层析(乙酸乙酯:石油醚=1:10)分离纯化,得到中间体50-2(416mg)。LCMS:MS m/z(ESI):381.0[M+H]+。
30.2中间体52-3的合成:
室温下将中间体52-2(416mg)溶于甲醇(5.00mL)中,加入铂碳(50mg),氢气球体系下室温搅拌反应2小时。停止反应,反应液通过硅藻土过滤,滤液减压浓缩,得到中间体52-3(250mg)。LCMS:MS m/z(ESI):383.0[M+H]+。
其余步骤参考实施例1,得到6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-异丙基-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物052)。
1H NMR(400MHz,DMSO)δ9.62(s,1H),9.24(s,1H),7.37(s,1H),7.09(d,J=8.2Hz,1H),6.90(d,J=8.2Hz,1H),3.44(p,J=6.8Hz,1H),1.85(s,3H),1.78(s,3H),1.41(d,J=6.8Hz,6H).LCMS:MS m/z(ESI):373.1[M+H]+。
30.3化合物052-1和052-2:
将化合物052通过SFC手性拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2,流动相B:IPA(含0.1%7.0mol/L Ammonia in MeOH),A:B=80:20),得到两个异构体:(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-异丙基-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物052-1,Rt=3.7min)和(R)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-异丙基-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物052-2,Rt=6.9min)
化合物052-1:
1H NMR(400MHz,DMSO)δ9.58(s,1H),9.24(d,J=3.4Hz,1H),7.36(d,J=3.4Hz,1H),7.08(d,J=8.2Hz,1H),6.89(d,J=8.2Hz,1H),3.51–3.39(m,1H),1.85(s,3H),1.77(s,3H),1.41(d,J=6.8Hz,6H).LCMS:MS m/z(ESI):373.1[M+H]+。
化合物052-2:
1H NMR(400MHz,DMSO)δ9.59(s,1H),9.24(d,J=3.5Hz,1H),7.36(d,J=3.5Hz,1H),7.08(d,J=8.2Hz,1H),6.89(d,J=8.3Hz,1H),3.47–3.44(m,1H),1.85(s,3H),1.77(s,3H),1.41(d,J=6.9Hz,6H).LCMS:MS m/z(ESI):373.1[M+H]+。
实施例31(S)-6-氨基-2-(叔丁基)-5-(3-羟基-2,6-二甲基苯基)-4-氧代-4,5-二氢噻唑并
[5,4-c]吡啶-7-甲酰胺(化合物053-1)和(R)-6-氨基-2-(叔丁基)-5-(3-羟基-2,6-二甲基苯
基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物053-2)
31.1中间体53-2的合成:
将2,2,2-三甲基硫代乙酰胺(5g)溶于乙醇(50mL)中,加入3-溴-2-氧代丙酸乙酯(8.3g),25℃下搅拌反应16小时。反应液减压浓缩,残余物加入饱和碳酸氢钠(40mL),用二氯甲烷(3×50mL)萃取。有机相用饱和盐水(50mL)洗涤,无水硫酸钠干燥,减压浓缩。残余物用硅胶柱层析(EA/PE:0%~2%)分离纯化,得到中间体53-2(6.2g)。LCMS:MS m/z(ESI):214.1[M+H]+。
31.2中间体53-3的合成:
将中间体53-3(2g)溶于醋酸(20mL)中,加入液溴(0.5mL),密闭反应体系下50℃搅拌反应16小时。冷却至室温,向反应液加饱和硫代硫酸钠水溶液(30mL)淬灭,用乙酸乙酯(3×50mL)萃取。有机相用饱和盐水(50mL)洗涤,无水硫酸钠干燥,减压浓缩。残余物用硅胶柱层析(EA/PE:0%~0.5%)分离纯化,得到中间体53-3(1.4g)。LCMS:MS m/z(ESI):292.0[M+H]+。
其余步骤参考实施例9,将步骤9.2中的中间体12-2替换为中间体53-3,得到6-氨基-2-(叔丁基)-5-(3-羟基-2,6-二甲基苯基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物053)。
1H NMR:(400MHz,DMSO)δ9.60(s,1H),7.20(s,2H),7.09(d,J=8.3Hz,1H),7.03(s,2H),6.89(d,J=8.3Hz,1H),1.86(s,3H),1.77(s,3H),1.42(s,9H).LCMS:MS m/z(ESI):387.1[M+H]+。
31.3化合物053-1和053-2:
将化合物053通过SFC手性拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in MeOH),A:B=70:30),得到两个异构体:(S)-6-氨基-2-(叔丁基)-5-(3-羟基-2,6-二甲基苯基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物053-1,Rt=4.1min)和(R)-6-氨基-2-(叔丁基)-5-(3-羟基-2,6-二甲基苯基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物053-2,Rt=6.4min)。
化合物053-1:
1H NMR(400MHz,DMSO)δ:9.58(s,1H),7.19(s,2H),7.08(d,J=8.4Hz,1H),7.03(s,2H),6.89(d,J=8.4Hz,1H),1.85(s,3H),1.77(s,3H),1.42(s,9H)。LCMS:MS m/z(ESI):387.2[M+H]+。
化合物053-2:
1H NMR(400MHz,DMSO)δ:9.58(s,1H),7.19(s,2H),7.09(d,J=8.4Hz,1H),7.03(s,2H),6.89(d,J=8.4Hz,1H),1.85(s,3H),1.77(s,3H),1.42(s,9H)。LCMS:MS m/z(ESI):387.2[M+H]+。
实施例32(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-(甲硫基)-4-氧代-4,5-二氢噻唑并
[5,4-c]吡啶-7-甲酰胺(化合物054-1)和(R)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-(甲硫
基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物054-2)
32.1中间体54-2的合成:
冰盐浴下将亚硝酸叔丁酯(1.89g)溶于干燥的乙腈(30.00mL)中,在氩气保护下加入溴化铜(4.58g),搅拌15分钟后,加入4-氨基-2-(甲硫基)噻唑-5-羧酸乙酯(3.20g),并保持冰盐浴搅拌2小时,升至室温搅拌过夜。反应液通过200~300目硅胶抽滤,滤饼用二氯甲烷(40.00mL)和乙酸乙酯(40.00mL)洗涤,滤液减压浓缩,残余物通过硅胶柱层析(乙酸乙酯:石油醚=15:1)分离纯化,得到中间体54-2(3.33g)。LCMS:MS m/z(ESI):281.9[M+H]+。
32.2中间体54-3的合成:
室温下将中间体54-2(1.50g)溶于乙醇(10.00mL)和水(5mL)中,在氩气保护下加入氢氧化钠(320.1mg),反应体系于50℃油浴中加热搅拌反应过夜。冷却至室温,反应液
减压浓缩有固体析出,加入水(5.0mL)溶解固体,用1N盐酸调节pH=3~4,有固体析出。室温下搅拌30分钟后,抽滤,滤饼真空干燥,得到中间体54-3(1.36g)。LCMS:MS m/z(ESI):253.9[M+H]+。
32.3中间体54-4的合成:
在氩气保护下将中间体54-3(500mg)溶于氯化亚砜(6mL)中,80℃油浴中加热搅拌反应75分钟。反应液减压浓缩,残余物溶于干燥的甲苯(5.0mL)中,并在冰浴下滴加到3-((4-甲氧基苄基)氧基)-2,6-二甲基苯胺(759.5mg)的吡啶溶液中,滴加完毕后,将反应体系升至室温,并在氩气保护下反应过夜。反应液减压浓缩,残余物加入水(10.0mL)稀释,用乙酸乙酯(3×10.0mL)萃取,有机相经饱和食盐水(2×20.0mL)洗涤,无水硫酸钠干燥后减压浓缩,残余物用硅胶柱层析(乙酸乙酯:石油醚=1:30~1:10)分离纯化,得到中间体54-4(657mg)。LCMS:MS m/z(ESI):493.1[M-H]+。
32.4中间体54-5的合成:
室温下将丙二腈(56.6mg)溶于乙二醇二甲醚(1.00mL)中,加入氢化钠(32.4mg),室温下搅拌反应0.5小时,再加入中间体54-4(200mg)的乙二醇二甲醚(2.00mL)溶液和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(29.4mg),反应体系在氩气保护下于80℃油浴中加热搅拌反应1h。停止反应,加入水(5.0mL)稀释,用乙酸乙酯(3×5.0mL)萃取,有机相经饱和食盐水(2×10.0mL)洗涤,无水硫酸钠干燥后减压浓缩,残余物用Prep-TLC(乙酸乙酯:石油醚=2:1)分离纯化,得到中间体54-5(139mg)。LCMS:MS m/z(ESI):479.2[M-H]+。
32.5化合物054的合成:
室温下将中间体54-5(60mg)溶于浓硫酸(1.00mL)中,于40℃油浴中加热搅拌反应3小时。反应液加入二氯甲烷(5.0mL)稀释,用饱和碳酸钠溶液调节pH=8~9,用二氯甲烷(3×5.0mL)萃取。有机相用饱和食盐水(2×5.0mL)洗涤,无水硫酸钠干燥后减压浓缩,
残余物经Prep-TLC纯化(二氯甲烷:甲醇=10:1)纯化,得到6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-(甲硫基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物054)(4.54mg)。
1H NMR(400MHz,DMSO)δ9.66(s,1H),8.97(d,J=3.3Hz,1H),7.37(d,J=3.3Hz,1H),7.08(d,J=8.3Hz,1H),6.90(d,J=8.3Hz,1H),2.82(s,3H),1.86(s,3H),1.78(s,3H).LCMS:MS m/z(ESI):377.1[M+H]+。
32.6化合物054-1和054-2:
将化合物054通过SFC手性拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in MeOH),A:B=55:45),得到两个异构体:(S)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-(甲硫基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物054-1,Rt=2.17min)和(R)-6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-(甲硫基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物054-2,Rt=4.75min)。
化合物054-1:
1H NMR(400MHz,DMSO)δ9.61(s,1H),8.97(d,J=3.3Hz,1H),7.36(d,J=3.3Hz,1H),7.08(d,J=8.3Hz,1H),6.89(d,J=8.3Hz,1H),2.81(s,3H),1.85(s,3H),1.77(s,3H).LCMS:MS m/z(ESI):377.1[M+H]+。
化合物054-2:
1H NMR(400MHz,DMSO)δ:9.61(s,1H),8.96(s,1H),7.36(d,J=3.4Hz,1H),7.08(d,J=8.4Hz,1H),6.89(d,J=8.4Hz,1H),2.81(s,3H),1.85(s,3H),1.77(s,3H).LCMS:MS m/z(ESI):377.1[M+H]+。
实施例33 6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-(甲磺酰基)-4-氧代-4,5-二氢噻唑并[5,4-
c]吡啶-7-甲酰胺(化合物055)
33.1中间体55-2的合成:
室温下将中间体54-5(100mg)溶于二氯乙烷(1.00mL)中,加入间氯过氧苯甲酸(144mg),室温下搅拌反应16小时。反应液中加入饱和碳酸氢钠水溶液淬灭,调节pH=7~8左右。反应液通过乙酸乙酯(3×5.0mL)萃取,有机相经饱和食盐水(2×10.0mL)洗涤,无水硫酸钠干燥后减压浓缩,残余物用Prep-TLC(乙酸乙酯:石油醚=1:1)分离纯化,得到中
间体55-2(15mg)。LCMS:MS m/z(ESI):509.1[M-H]。
33.2化合物055的合成:
室温下将中间体55-2(15mg)溶于浓硫酸(1.0mL)中,于25℃油浴中搅拌反应1小时。停止反应,向反应液中加入乙酸乙酯(5.0mL)稀释,在冰水浴下通过饱和碳酸氢钠溶液调节pH=7~8左右,用乙酸乙酯(3×5.0mL)萃取。有机相经饱和食盐水(2×5.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物通过高效液相制备色谱法纯化,得到目标产物6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-(甲磺酰基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物055)(1.01mg)。
1H NMR(400MHz,MeOD)δ:7.14(d,J=8.4Hz,1H),6.93(d,J=8.4Hz,1H),3.51(s,3H),1.97(s,3H),1.92(s,3H).
LCMS:MS m/z(ESI):409.1[M+H]+。
实施例34 6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-(1-甲基-1H-吡唑-4-基)-4-氧代-4,5-二氢
噻唑并[5,4-c]吡啶-7-甲酰胺(化合物056)
34.1中间体56-2的合成:
将中间体55-2(150mg)溶于二氧六环(3mL)和水(0.3mL)的混合溶剂中,依次加入(1-甲基-1H-吡唑-4-基)硼酸(73mg),磷酸钾(123.1mg),1,1-双(二苯基磷)二茂铁二氯化钯(21.2mg),在氮气保护下置换气三次,于100℃反应16小时。冷却至室温,反应液减压浓缩,残余物经Prep-TLC(石油醚:乙酸乙酯=1:4)分离纯化,得到中间体56-2(130mg)。LCMS:MS m/z(ESI):513.3[M+H]+。
34.2化合物056的合成:
将中间体56-2(130mg)溶于硫酸(2mL)中,在55℃下搅拌反应1小时。冷却至0℃,加入饱和碳酸氢钠水溶液调节pH至8,用乙酸乙酯(3x30mL)萃取,有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥后减压浓缩。残余物通过高效液相制备色谱法纯化,得到目标产物6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-(1-甲基-1H-吡唑-4-基)-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物056)(3.41mg)。
1H NMR(400MHz,DMSO)δ:9.57(s,1H),9.23(s,1H),8.64(s,1H),8.16(s,1H),7.36(s,1H),7.09(d,J=8.4Hz,1H),6.89(d,J=8.4Hz,1H),3.93(s,3H),1.87(s,3H),1.79(s,3H).LCMS:MS m/z(ESI):411.1[M+H]+。
实施例35 6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-吗啉-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶
-7-甲酰胺(化合物057)
35.1中间体57-2的合成:
将中间体55-2(140mg)溶于N,N-二甲基甲酰胺(2mL)中,加入吗啉(477.1mg),在封管中于25℃反应16小时。反应液加入水(10mL),有白色固体析出。过滤,滤饼真空干燥,得到中间体57-2(110mg)。LCMS:MS m/z(ESI):518.2[M+H]+。
35.2化合物057的合成:
将中间体57-2(100mg)溶于硫酸(2mL)中,在55℃下搅拌反应1小时。反应液冷却至0℃,加入饱和碳酸氢钠水溶液调节pH至8,用乙酸乙酯(3x30mL)萃取。有机相经
饱和食盐水(30mL)洗涤,无水硫酸钠干燥后减压浓缩,残余物通过高效液相制备色谱法纯化,得到目标产物6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-吗啉-4-氧代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物057)(12.4mg)。
1H NMR(400MHz,DMSO)δ9.61(s,1H),9.12(s,1H),7.19(s,1H),7.05(d,J=8.4Hz,1H),6.90–6.83(m,1H),3.77–3.71(m,4H),3.61–3.56(m,4H),1.84(s,3H),1.76(s,3H).LCMS:MS m/z(ESI):416.2[M+H]+。
实施例36 6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-甲基-4-硫代-4,5-二氢噻唑并[5,4-c]吡啶
-7-甲酰胺(化合物058)
36.1中间体58-2的合成:
将中间体1-4(150mg)溶于甲苯(1mL),加入Lawesson试剂(535mg),加热到120℃搅拌反应3小时。冷却至室温,向反应液加水(5mL),用乙酸乙酯(3×10mL)萃取。有机相经饱和食盐水(10mL)洗涤,无水硫酸钠干燥后过滤,减压浓缩。残余物用硅胶柱层析(EA/PE:0%~10%)分离纯化,得到中间体58-2(40mg)。LCMS:MS m/z(ESI):357.1[M+H]+。36.2化合物058的合成:
剩余步骤参考实施例1,得到目标化合物6-氨基-5-(3-羟基-2,6-二甲基苯基)-2-甲基-4-硫代-4,5-二氢噻唑并[5,4-c]吡啶-7-甲酰胺(化合物058)。
1H NMR:(400MHz,CDCl3)δ7.46(s,1H),7.09(d,J=8.0Hz,1H),6.83(d,J=8.0Hz,1H),6.93(s,2H),2.73(s,3H),2.15(s,3H),1.93(s,3H).
LCMS:MS m/z(ESI):361.2[M+H]+。
实施例37 6-氨基-2-((二乙基胺基)甲基)-5-(3-羟基-2,6-二甲基苯基)-4-氧代-4,5-二氢噻
吩并[3,2-c]吡啶-7-甲酰胺(化合物059)
37.1中间体59-2的合成:
在氩气保护下将中间体23-2(1.0g)溶于N,N-二甲基甲酰胺(10.0mL)中,依次加入(三丁基锡基)甲醇(944mg)和XPhos-Pd-G2(308mg),反应体系于80℃油浴中加热搅拌反应2小时。冷却至室温,向反应液中加入水(20.0mL)稀释,用乙酸乙酯(3×20.0mL)萃取,有机相经饱和食盐水(2×20.0mL)洗涤,无水硫酸钠干燥后过滤,减压浓缩,残余物经硅胶柱层析(乙酸乙酯:石油醚=1:20~1:1)纯化,得到中间体59-2(210mg)。LCMS:MS m/z(ESI):462.2[M+H]+。
37.2中间体59-3的合成:
室温下将中间体59-2(210mg)溶于二氯甲烷(6.00mL)中,加入Dess-Martin试剂(286mg),室温下搅拌反应1小时。向反应液中加入饱和碳酸氢钠溶液(10ml)淬灭,用二氯甲烷(3×10.0mL)萃取,有机相经饱和食盐水(2×10.0mL)洗涤,无水硫酸钠干燥后过滤,减压浓缩,残余物经Prep-TLC(乙酸乙酯:石油醚=1:1)纯化,得到中间体59-3(150mg)。LCMS:MS m/z(ESI):460.2[M+H]+。
37.3中间体59-4的合成:
室温下将中间体59-3(110mg)溶于二氯甲烷(4.00mL)中,加入二乙胺(88mg),室温下搅拌10分钟后,再加入三乙酰氧基硼氢化钠(153mg),室温下搅拌反应1小时。向反应液中加入水(10.00mL)稀释,用二氯甲烷(3×10.0mL)萃取,有机相经饱和食盐水(2×10.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物经Prep-TLC(乙酸乙酯:石油醚=2:1)纯化,得到中间体59-4(100mg)。LCMS:MS m/z(ESI):517.3[M+H]+。
37.4化合物059的合成:
其余步骤参考实施例16,得到目标产物6-氨基-2-((二乙基胺基)甲基)-5-(3-羟基-2,6-二甲基苯基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物059)。
1H NMR(400MHz,DMSO)δ9.71(s,1H),7.28-7.18(m,3H),7.06(d,J=8.3Hz,1H),7.01–6.91(m,3H),3.87(s,2H),2.67–2.62(m,4H),1.84(s,3H),1.76(s,3H),1.10(s,6H).LCMS:MS m/z(ESI):413.2[M-H]。
实施例38 6-氨基-2-氰基-5-(3-羟基-2,6-二甲基苯基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶
-7-甲酰胺(化合物060)
38.1中间体60-2的合成:
室温下将中间体23-2(120mg)溶于浓硫酸(2.0mL)中,反应体系在40℃油浴中搅拌反应2小时。冷却至室温,向反应液中加入饱和碳酸氢钠溶液中和,用乙酸乙酯(3×5.0mL)萃取。有机相经饱和食盐水(2×5.0mL)洗涤,无水硫酸钠干燥后过滤,减压浓缩,残余物通过石油醚(15.0mL)打浆纯化,过滤,滤饼用石油醚(3×10.0mL)洗涤,真空干燥,得到中间体60-2(60mg)。LCMS:MS m/z(ESI):408.0[M+H]+。
38.2化合物060的合成:
室温下将中间体60-2(60mg)溶于N-甲基吡咯烷酮(1.0mL)中,加入氰化亚铜(39mg),于微波180℃条件下反应1小时。向反应液中加入水(5.0mL)稀释,用乙酸乙酯(3×5.0mL)萃取,有机相经饱和食盐水(2×5.0mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩。残余物通过薄层色谱法(甲醇:二氯甲烷=1:15)粗纯化,得到粗产物40mg,再通过高效液相制备色谱法纯化,得到目标产物6-氨基-2-氰基-5-(3-羟基-2,6-二甲基苯基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物060)(1.03mg)。
1H NMR(400MHz,DMSO)δ:9.63(s,1H),8.10(s,1H),7.40(s,2H),7.11-7.08(m,3H),6.90(d,J=8.4Hz,1H),1.86(s,3H),1.78(s,3H)。
LCMS:MS m/z(ESI):355.1[M+H]+。
实施例39 3-氨基-2-(3-羟基-2,6-二甲基苯基)-1-氧代-8-(四氢-2H-吡喃-4-羰基)-
1,2,6,7,8,9-六氢噻吩并[3,2-c:4,5-c']联吡啶-4-甲酰胺(化合物104)
39.1中间体104-2的合成:
将3-氧代哌啶-1-甲酸苄酯(10g)溶于乙醇(150mL),依次加入2-氰基乙酸乙酯(6.3g)、升华硫(6.9g)和吗啉(3.7g),在50℃下搅拌反应16小时。减压除去溶剂,残余物用硅胶柱层析纯化(PE:EA=1:0~14:1),得到中间体104-2(16.7g)。
LCMS:MS m/z(ESI):361.2[M+H]+。
39.2中间体104-3的合成:
将亚硝酸叔丁酯(5.7g)溶于乙腈(57mL),再加入溴化铜(14.5g),在0℃下搅拌15分钟,在此温度下缓慢加入中间体104-2(16.7g)的乙腈(167mL)溶液,在0℃下搅拌15分钟后将反应液加热到25℃反应16小时。向反应液加水(200mL),用乙酸乙酯(3×200mL)萃取、饱和盐水(200mL)洗涤、无水硫酸钠干燥后减压浓缩。残余物采用柱层析(PE:EA=1:0~5:1)纯化得到粗品。粗品用甲醇打浆,所得固体干燥,得到中间体104-3(8g)。LCMS:MS m/z(ESI):446.0[M+Na]+。
39.3中间体104-4的合成:
将中间体104-3(3.2g)溶于1,4-二氧六环(32mL),再加入3-甲氧基-2,6-二甲基苯胺
(1.7g)和三甲基铝(1.6M,47.3mL),将反应液加热到80℃反应1小时。降温至0℃,将反应液缓慢加到甲醇(30mL)中淬灭,将反应液浓缩,加水(30mL)稀释,用乙酸乙酯(3×10mL)萃取,饱和盐水(20mL)洗涤,无水硫酸钠干燥后减压浓缩。残余物用硅胶柱层析纯化,得到中间体104-4(3.4g)。LCMS:MS m/z(ESI):529.2[M+H]+。
39.4中间体104-5的合成:
将丙二腈(352mg)溶于乙二醇二甲醚(14mL),加入叔丁醇钠(509mg),在室温搅拌30分钟,再加入中间体104-4(1.4g)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(194mg)。用氩气置换三次,将反应液加热到100℃反应16小时。向反应液中加水(20mL),用乙酸乙酯(3×30mL)萃取,有机相经饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥后减压浓缩,残余物用硅胶柱层析(PE:EA=1:0~1:3)纯化,得到中间体104-5(750mg)。LCMS:MS m/z(ESI):515.2[M+H]+。
39.5中间体104-6的合成:
将中间体104-5(300mg)溶于四氢呋喃(10mL),加入钯碳(30mg),用氢气置换三次,反应于氢气氛围下30℃搅拌1小时。用硅藻土过滤,滤液减压浓缩,得到中间体104-6(150mg)。LCMS:MS m/z(ESI):381.1[M+H]+。
39.6中间体104-7的合成:
将中间体104-6(150mg)溶于N,N-二甲基甲酰胺(1.5mL),依次加入四氢吡喃-4-甲酸(51mg)、1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐(114mg)、1-羟基苯并三唑(80
mg)和N,N-二异丙基乙胺(154mg)。在25℃反应1小时。向反应液加水(10mL),用乙酸乙酯(3×10mL)萃取,有机相经饱和盐水(20mL)洗涤,无水硫酸钠干燥后浓缩。残余物用硅胶柱层析(PE:EA=1:4),得到中间体104-7(77mg)。LCMS:MS m/z(ESI):493.3[M+H]+。
39.7中间体104-8的合成:
将中间体104-7(77mg)溶于二氯甲烷(2mL),缓慢加入三溴化硼(0.4mL),在25℃下反应1小时。在0℃下,用饱和碳酸氢钠溶液淬灭并调节pH=7,反应液用乙酸乙酯和四氢呋喃的混合溶液(1:1,10mLx3)萃取,饱和盐水(20mL)洗涤,无水硫酸钠干燥后减压浓缩,得到中间体104-8(60mg)。LCMS:MS m/z(ESI):479.2[M+H]+。
39.8化合物104的合成:
将中间体104-8(60mg)溶于乙醇(3mL)和水(1mL),加入帕金斯催化剂(2mg)。将反应加热到100℃,回流16小时。减压除去溶剂,加入N,N-二甲基甲酰胺(2mL)稀释,用高效液相制备色谱法纯化得到化合物3-氨基-2-(3-羟基-2,6-二甲基苯基)-1-氧代-8-(四氢-2H-吡喃-4-羰基)-1,2,6,7,8,9-六氢噻吩并[3,2-c:4,5-c']联吡啶-4-甲酰胺(化合物104)(1.02mg)。
1H NMR(400MHz,DMSO)δ8.35(s,1H),7.15(s,2H),7.06(d,J=8.2Hz,1H),6.94(s,2H),6.88(d,J=8.2Hz,1H),4.32–4.23(m,1H),4.19–4.08(m,1H),3.80–3.61(m,4H),2.70–2.66(m,2H),2.56(t,J=6.9Hz,2H),2.35–2.30(m,1H),1.97–1.86(m,2H),1.85(s,3H),1.77(s,3H),1.63–1.53(m,2H).
LCMS:MS m/z(ESI):497.2[M+H]+。
实施例40 6-氨基-2,3-二甲基-5-(5-甲基苯并[d]异噻唑-4-基)-4-氧代-4,5-二氢噻吩并
[3,2-c]吡啶-7-甲酰胺(化合物108)
40.1中间体108-2的合成:
室温下将2-溴-6-氟-3-甲基苯甲醛(4.00g)溶于N,N-二甲基甲酰胺(40.0mL)中,加入碳酸钾(3.84g)和2-甲基丙烷-2-硫醇(4.84g),反应体系于50℃油浴中加热搅拌反应过夜。向反应液中加入水(50.0mL)稀释,通过乙酸乙酯(3×50.0mL)萃取,有机相经饱和食盐水(2×50.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物用硅胶柱层析(石油醚:乙酸乙酯=1:0~10:1)纯化,得到中间体108-2(1.40g)。1H NMR(400MHz,DMSO)δ:10.41(s,1H),7.66-7.59(m,2H),2.49(s,3H),1.31(s,9H)。
40.2中间体108-3的合成:
室温下将中间体108-2(1.40g)溶于异丙醇(12.0mL)和水(2.0mL)中,反应体系于70℃油浴中加热搅拌反应0.5小时。冷却至室温,将反应液倒入水(20.0mL)中,通过二氯甲烷(3×20.0mL)萃取,有机相经饱和食盐水(2×20.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩后,残余物溶于正丁醇(10.0mL)中,加入对甲苯磺酸(1.68g),反应体系于120℃油浴中加热搅拌反应过夜。向反应液中加入水(30.0mL)稀释,通过乙酸乙酯(3×30.0mL)萃取,有机相依次经饱和碳酸氢钠溶液(2×30.0mL)、饱和食盐水(2×30.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,得到中间体108-3(1.03g)。LCMS:MS m/z(ESI):228.0[M+H]+。
40.3中间体108-4的合成:
室温下将中间体108-3(200mg)溶于甲苯(4.0mL)中,依次加入二苯甲酮亚胺(190
mg),叔丁醇钠(118mg)和1,1'-联萘-2,2'-双二苯基膦(409mg)和三(二亚苄基丙酮)二钯(200mg),用氩气置换3次后,将反应体系升温至90℃搅拌过夜。停止反应,反应液减压浓缩后,加入水(20.0mL)稀释,通过乙酸乙酯(3×20.0mL)萃取,有机相经饱和食盐水(2×20.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,用Prep-TLC(石油醚:乙酸乙酯=5:1)纯化,得到中间体108-4(150mg)。LCMS:MS m/z(ESI):329.2[M+H]+。
40.4中间体108-5的合成:
室温下将中间体108-4(150mg)溶于盐酸/1,4-二氧六环溶液中(3.00mL,4M),反应体系在25℃下搅拌1小时。停止反应,反应液通过饱和碳酸氢钠溶液中和,用乙酸乙酯(3×20.0mL)萃取,有机相经饱和食盐水(2×20.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,用Prep-TLC纯化,得到中间体108-5(20mg)。LCMS:MS m/z(ESI):165.2[M+H]+。
40.5中间体108-6的合成:
室温下将中间体12-2(36mg)和中间体108-5(20mg)溶于1,4-二氧六环(1.0mL)中,在搅拌下滴加三甲基铝(0.8mL,1.6M),用氩气置换三次,反应体系于封管中在120℃油浴中搅拌反应5小时。停止反应,将反应液倒入甲醇(3.0mL)中,用二氯甲烷(3×20.0mL)萃取,有机相经饱和食盐水(2×20.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物通过Prep-TLC纯化,得到中间体108-6(20mg)。LCMS:MS m/z(ESI):381.0[M+H]+。
40.6中间体108-7的合成:
室温下将丙二腈(7mg)溶于乙二醇二甲醚(1.0mL)中,加入氢化钠(4mg),室温下搅拌反应10分钟后,再加入中间体108-6(20mg)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(4mg),用氩气置换3次后,反应体系在氩气保护下于100℃加热搅拌反应5小时。停止反应,向反应液中加入水(10.0mL)稀释,通过乙酸乙酯(3×10.0mL)萃取,有机相经饱和食盐水(2×10.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,用Prep-TLC(石油
醚:乙酸乙酯=4:1)纯化,得到中间体108-7(10mg)。LCMS:MS m/z(ESI):367.0[M+H]+。
40.7化合物108的合成:
室温下将中间体108-7(10mg)溶于乙醇(3.0mL)和水(1.0mL)中,加入(二甲基膦酸)铂(II)氢化络合物(1mg),用氩气置换3次后,反应体系在氩气保护下于100℃油浴中搅拌反应24小时。将反应液减压浓缩后,通过高效液相制备色谱法纯化,得到目标产物6-氨基-2,3-二甲基-5-(5-甲基苯并[d]异噻唑-4-基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物108)(1.13mg)。
1H NMR(400MHz,MeOD)δ8.57(s,1H),8.17(d,J=8.4Hz,1H),7.70(d,J=8.4Hz,1H),2.36(s,6H),2.27(s,3H).LCMS:MS m/z(ESI):385.2[M+H]+。
实施例41(S)-7-氨基-8-(5-甲基-1H-吲唑-4-基)-9-氧代-8,9-二氢噻吩并[3,2-b:4,5-c']二吡
啶-6-甲酰胺(化合物109-P1)和(R)-7-氨基-8-(5-甲基-1H-吲唑-4-基)-9-氧代-8,9-二氢噻
吩并[3,2-b:4,5-c']二吡啶-6-甲酰胺(化合物109-P2)
41.1中间体109-2的合成:
室温下将2-(3-溴吡啶-2-基)乙酸乙酯(5.00g)溶于二甲亚砜(30.0mL)中,加入氢化钠(590mg),室温下搅拌反应1小时,再加入4-甲氧基苄基异硫氰酸酯(4.41g),室温下搅拌反应0.5小时后,用氩气置换3次。反应体系在氩气保护下于120℃油浴中加热搅拌反应1小时。停止反应,向反应液中加入水(100mL)稀释,通过乙酸乙酯(3×200mL)萃取,有机相经饱和食盐水(2×150.0mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物通过硅胶柱层析(二氯甲烷:甲醇=30:1)纯化,得到中间体109-2(5.77g)。LCMS:MS m/z(ESI):343.1[M+H]+。
41.2中间体109-3的合成:
室温下将中间体109-2(5.77g)溶于三氟乙酸(20.0mL)中,加入甲基磺酸(1.0mL),于25℃搅拌反应20分钟。向反应液中加入乙酸乙酯(50.0mL),用水(3×100mL)萃取,水相用氨水调节pH至8~9,用乙酸乙酯(3×150mL)萃取。有机相经饱和食盐水(2×100mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,得到中间体109-3(2.2g)。LCMS:MS m/z(ESI):223.0[M+H]+。
41.3中间体109-4的合成:
室温下将中间体109-3(2.20g)和溴化铜(3.10g)溶于乙腈(20.0mL)中,在0℃下加入亚硝酸叔丁酯(1.23g),并在0℃下搅拌2小时后,恢复室温搅拌过夜。停止反应,向反应液中加入水(50.0mL),用乙酸乙酯(3×100.0mL)萃取,有机相经氨水(2×50.0mL),饱和食盐水(2×50.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,得到中间体109-4(2.5g)。LCMS:MS m/z(ESI):285.9[M+H]+
41.4中间体109-5的合成:
室温下将中间体109-4(2.50g)溶于甲醇(10.00mL)和水(5.00mL)中,加入氢氧化锂(628mg),反应体系于25℃油浴中搅拌反应4小时。停止反应,减压浓缩除掉甲醇,用1N盐酸溶液调节pH至3~4,用乙酸乙酯(3×50.0mL)萃取,有机相经饱和食盐水(2×50.0mL)洗涤,无水硫酸钠干燥,过滤,减压浓缩,得到中间体109-5(2.0g)。LCMS:MS m/z(ESI):257.9[M+H]+。
41.5中间体109-6的合成:
室温下将中间体109-5(420mg)溶于乙腈(10.0mL)中,依次加入5-甲基-1H-吲唑-4-
胺(240mg),四甲基氯代脲六氟磷酸酯(TCFH,686mg)和N-羟基丁二酰亚胺(401mg),反应体系在30℃油浴中搅拌反应2小时。停止反应,有固体析出,过滤,滤饼用乙酸乙酯(30.0mL)洗涤,干燥后得到中间体109-6(470mg)。LCMS:MS m/z(ESI):387.0[M+H]+。
41.6中间体109-7的合成:
室温下将丙二腈(160mg)溶于乙二醇二甲醚(10.0mL)中,加入氢化钠(97mg),室温下搅拌反应15分钟后,加入中间体109-6(470mg)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(178mg),用氩气置换3次后,反应体系在氩气保护下于100℃油浴中加热搅拌反应2小时。冷却至室温,向反应液中加入水(30.0mL)稀释,通过乙酸乙酯(3×30.0mL)萃取,有机相经饱和食盐水(2×30.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物通过薄层色谱(二氯甲烷:甲醇=15:1)纯化,得到中间体109-7(120mg)。LCMS:MS m/z(ESI):373.2[M+H]+。
41.7化合物109的合成:
室温下将中间体109-7(120mg)溶于乙醇(3.0mL)和水(1.0mL)中,加入(二甲基膦酸)铂(II)氢化络合物(14mg),用氩气置换3次后,反应体系在氩气保护下于100℃油浴中搅拌反应2小时。停止反应,将反应液减压浓缩,残余物通过高效液相制备色谱法纯化,得到7-氨基-8-(5-甲基-1H-吲唑-4-基)-9-氧代-8,9-二氢噻吩[3,2-b:4,5-c']二吡啶-6-甲酰胺(化合物109)(24.03mg)。LCMS:MS m/z(ESI):391.1[M+H]+。
41.8化合物109-P1和109-P2的分离:
将化合物109通过SFC手性拆分(手性柱:250*30mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in MeOH),A:B=70:30),得到两个异构体:(S)-7-氨基-8-(5-甲基-1H-吲唑-4-基)-9-氧代-8,9-二氢噻吩并[3,2-b:4,5-c']二吡啶-6-甲酰胺(化合物109-P1,Rt=7.9min)和(R)-7-氨基-8-(5-甲基-1H-吲唑-4-基)-9-氧代-8,9-二氢噻吩并[3,2-b:4,5-c']二吡啶-6-甲酰胺(化合物
109-P2,Rt=14.2min)。
化合物109-P1:
1H NMR(400MHz,MeOD)δ8.56(dd,J=4.8,1.5Hz,1H),8.26(dd,J=8.0,1.5Hz,1H),7.79(s,1H),7.68(d,J=8.6Hz,1H),7.51(d,J=8.6Hz,1H),7.29(dd,J=8.0,4.8Hz,1H),2.26(s,3H).LCMS:MS m/z(ESI):391.2[M+H]+。
化合物109-P2:
1H NMR(400MHz,MeOD)δ8.59–8.54(m,1H),8.26(dd,J=8.0,1.5Hz,1H),7.79(s,1H),7.68(d,J=8.6Hz,1H),7.50(d,J=8.6Hz,1H),7.29(dd,J=8.1,4.8Hz,1H),2.26(s,3H).LCMS:MS m/z(ESI):391.2[M+H]+。
实施例42(S)-6-氨基-2-乙基-3-甲基-5-(5-甲基-1H-吲唑-4-基)-4-氧代-4,5-二氢噻吩并
[3,2-c]吡啶-7-甲酰胺(化合物110-P1)和(R)-6-氨基-2-乙基-3-甲基-5-(5-甲基-1H-吲唑-
4-基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物110-P2)
42.1中间体110-2的合成:
将中间体45-2(13.6g)溶于甲醇(136mL),在0℃下,缓慢加入二氯亚砜(22.4mL)。升温至80℃,反应16小时。减压除去溶剂,加入乙酸乙酯(80mL)稀释,反应液先后用饱和碳酸氢钠溶液(3×100mL)和饱和氯化钠溶液(100mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,得到中间体110-2(13.7g)。LCMS:MS m/z(ESI):234.9[M+H]+。
42.2中间体110-3的合成:
将中间体110-2(10.0g)溶于1,4-二氧六环(100mL),加入5-甲基-1H-吲唑-4-胺(6.27g)和三甲基铝(1.6M,132.8mL),用氩气置换三次,将反应液加热到120℃反应6小时。降温至0℃,将反应液缓慢加入到甲醇(300mL)中淬灭。反应液浓缩,加水(100mL)稀释,用乙酸乙酯(3×200mL)萃取,有机相经饱和氯化钠溶液(400mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物用石油醚和乙酸乙酯的混合溶剂(PE:EA=10:1,50
mL)打浆,所得固体过滤,滤饼干燥后得到中间体110-3(10.1g)。LCMS:MS m/z(ESI):350.0[M+H]+。
42.3中间体110-4的合成:
将丙二腈(3.80g)溶于乙二醇二甲醚(120mL),加入叔丁醇钠(5.51g),在室温下搅拌30分钟,再依次加入中间体110-3(10.1g)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(2.11g)。用氩气置换三次,将反应液加热到100℃反应16小时。停止反应,向反应液中加水(240mL),用乙酸乙酯(3×400mL)萃取,有机相经饱和氯化钠溶液(600mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物用甲醇(50mL)打浆,所得固体过滤,滤饼干燥后得到中间体110-4(6.8g)。LCMS:MS m/z(ESI):336.1[M+H]+。
42.4中间体110-5的合成:
将中间体110-4(6.5g)溶于乙腈(325mL),0℃下分批缓慢加入N-溴代丁二酰亚胺(3.3g),加入完毕后于25℃反应2小时。将反应液过滤,滤饼用乙腈洗涤,滤饼干燥后得到中间体110-5(4g)。LCMS:MS m/z(ESI):414.0[M+H]+。
42.5中间体110-6的合成:
将中间体110-5(4g)溶于1,4-二氧六环(40mL)和水(4mL)的混合溶剂中,依次加入三氟(乙烯基)硼酸钾(3.88g),碳酸钾(3.34g)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(707mg),用氩气置换三次,在110℃下反应16小时。停止反应,向反应液中加水(40mL),用乙酸乙酯(3×40mL)萃取,有机相经饱和氯化钠溶液(40mL)洗涤,无水硫酸钠干燥后,减压浓缩,残余物用柱层析(DCM:MeOH=1:0~80:1)纯化,得到中间体110-6(950mg)。
LCMS:MS m/z(ESI):362.2[M+H]+。
42.6中间体110-7的合成:
将中间体110-6(950mg)溶于四氢呋喃(20mL),加入钯碳催化剂(10%,95mg),用氢气置换三次。将反应置于氢气氛下,加热到30℃反应2小时。反应液通过硅藻土过滤,浓缩滤液后再加入N,N-二甲基甲酰胺溶解,用反相柱(MeCN in H2O:0~39%)纯化,得到中间体110-7(140mg)。LCMS:MS m/z(ESI):364.1[M+H]+。
42.7化合物110的合成:
将中间体110-7(140mg)溶于乙醇(3mL)和水(1mL)的混合溶剂中,加入(二甲基膦酸)铂(II)氢化络合物(17mg),用氩气置换三次,将反应液加热到100℃反应16小时。减压除去溶剂,用二氯甲烷和甲醇的混合溶剂(10:1,1.5mL)打浆,所得固体过滤,滤饼干燥后得到化合物6-氨基-2-乙基-3-甲基-5-(5-甲基-1H-吲唑-4-基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物110)(90mg)。LCMS:MS m/z(ESI):382.1[M+H]+。
42.8化合物110-P1和110-P2的分离:
将化合物110通过SFC手性拆分(手性柱:250*30mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in MeOH),A:B=75:25),得到两个异构体:(S)-6-氨基-2-乙基-3-甲基-5-(5-甲基-1H-吲唑-4-基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物110-P1,Rt=6.2min)和(R)-6-氨基-2-乙基-3-甲基-5-(5-甲基-1H-吲唑-4-基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物110-P2,Rt=10.3min)。
化合物110-P1:
1H NMR(400MHz,DMSO)δ13.24(s,1H),7.68(s,1H),7.61(d,J=8.5Hz,1H),7.41(d,J=8.5Hz,1H),7.14(s,2H),6.98(s,2H),2.72(q,J=7.4Hz,2H),2.30(s,3H),2.08(s,3H),1.21(t,J=7.4Hz,3H).LCMS:MS m/z(ESI):382.1[M+H]+。
化合物110-P2:
1H NMR(400MHz,DMSO)δ13.24(s,1H),7.68(s,1H),7.61(d,J=8.5Hz,1H),7.41(d,J
=8.5Hz,1H),7.14(s,2H),6.98(s,2H),2.72(q,J=7.4Hz,2H),2.30(s,3H),2.08(s,3H),1.21(t,J=7.4Hz,3H).LCMS:MS m/z(ESI):382.1[M+H]+。
实施例43(R)-6-氨基-5-(5-氯-1H-吲唑-4-基)-2-乙基-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-
c]吡啶-7-甲酰胺(化合物111-P1)和(S)-6-氨基-5-(5-氯-1H-吲唑-4-基)-2-乙基-3-甲基-4-
氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物111-P2)
43.1中间体111-2的合成:
将3-巯基-2-戊酮(25.00g)溶于甲苯(300.0mL)中,降温至0℃,加入甲醇钠(1.14g)后缓慢加入3-甲氧基丙烯酸甲酯(24.60g),0℃下搅拌反应16小时。LCMS检测到中间体111-2的质谱信号,MS m/z(ESI):235.1[M+H]+。反应液直接用于下一步。
43.2中间体111-3的合成:
0℃下,将浓盐酸(12M,35.25mL)滴加至上一步得到的反应液中,缓慢升至室温,搅拌过夜。加入水(1.0L)稀释,用乙酸乙酯(3×1.0L)萃取,有机相经饱和氯化钠溶液(3×500.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物通过硅胶柱层析(石油醚:乙酸乙酯=100:1~40:1)纯化,得到中间体111-3(29.9g)。LCMS:MS m/z(ESI):185.2[M+H]+。
43.3中间体111-4的合成:
将中间体111-3(10.0g)溶于N,N-二甲基甲酰胺(200mL),加入N-溴代丁二酰亚胺(10.6g),于50℃油浴中加热搅拌反应16小时。加入水(2.0L)稀释,用乙酸乙酯(4×500.0
mL)萃取,有机相经饱和氯化钠溶液(4×500.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物通过硅胶柱层析(石油醚:乙酸乙酯=100:1~30:1)纯化,得到中间体111-4(6.0g)。LCMS:MS m/z(ESI):262.9[M+H]+。
43.4中间体111-5的合成:
室温下将1H-吲唑-4-胺(10.0g)溶于四氢呋喃(150.0mL)中,冷却至-68℃,滴加浓硫酸(1.3mL),并在-68℃下搅拌5分钟。于-68℃下加入N-氯代丁二酰亚胺(10.0g)的四氢呋喃(50.0mL)溶液,并在-68℃下搅拌2小时。停止反应,在-68℃下向反应液中加入碳酸钠固体中和,恢复室温,加水(200.0mL)稀释,用乙酸乙酯(3×200.0mL)萃取,有机相经饱和氯化钠溶液(2×200.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物通过硅胶柱层析(石油醚:乙酸乙酯=4:1~2:1)分离纯化,得到中间体111-5(10.2g)。LCMS:MS m/z(ESI):168.2[M+H]+。
43.5中间体111-6的合成:
将中间体111-4(5.00g)溶于1,4-二氧六环(50mL)中,加入中间体111-5(3.03g),在搅拌下加入三甲基铝(1.6M,59.3mL),反应于氩气保护下120℃搅拌反应3小时。在0℃下将反应液缓慢加入到甲醇(200mL)中淬灭,减压浓缩,加水(100mL)稀释,用乙酸乙酯(3×100mL)萃取,有机相经饱和氯化钠溶液(200mL)洗涤,无水硫酸钠干燥,降压浓缩。残余物用乙酸乙酯(100mL)打浆、过滤、滤饼干燥后得到中间体111-6(4.75g)。LCMS:MS m/z(ESI):398.0[M+H]+。
43.6中间体111-7的合成:
室温下将丙二腈(2.07g)溶于乙二醇二甲醚(20.0mL)中,加入氢化钠(1.41g),室温下搅拌反应0.5小时后,加入中间体111-6(5.00g)的乙二醇二甲醚(50.0mL)溶液和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(917mg),在氩气的保护下,于120℃加热搅拌反应16小时。冷却至室温,向反应液中加水(200mL),用乙酸乙酯(3×200mL)萃取,有机相经饱
和氯化钠溶液(2×200mL)洗涤,无水硫酸钠干燥,减压浓缩。残余物用二氯甲烷和甲醇的混合溶剂(DCM:MeOH=10:1)打浆、过滤,干燥滤饼得到中间体111-7(1.08g)。LCMS:MS m/z(ESI):384.1[M+H]+。
43.7化合物111的合成:
将中间体111-7(2.00g)溶于乙醇(21.0mL)和水(7.0mL)的混合溶液中,加入(二甲基膦酸)铂(II)氢化络合物(245mg),用氩气置换三次,于100℃油浴中加热搅拌反应16小时。冷却至室温,有固体析出,过滤,滤饼用甲醇(20mL)打浆纯化,得到化合物6-氨基-5-(5-氯-1H-吲唑-4-基)-2-乙基-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(1.48g)。LCMS:MS m/z(ESI):402.1[M+H]+。
43.8化合物111-P1和111-P2的分离:
将化合物111通过SFC手性拆分(手性柱:250*30mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in MeOH),A:B=65:35),得到两个异构体:(R)-6-氨基-5-(5-氯-1H-吲唑-4-基)-2-乙基-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物111-P1,Rt=2.8min)和(S)-6-氨基-5-(5-氯-1H-吲唑-4-基)-2-乙基-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物111-P2,Rt=5.1min)。
化合物111-P1:
1H NMR(400MHz,DMSO)δ13.52(s,1H),7.87(s,1H),7.76(d,J=8.8Hz,1H),7.60(d,J=8.8Hz,1H),7.21(s,2H),7.16(s,2H),2.73(q,J=7.5Hz,2H),2.30(s,3H),1.21(t,J=7.5Hz,3H).LCMS:MS m/z(ESI):402.2[M+H]+。
化合物111-P2:
1H NMR(400MHz,DMSO)δ13.52(s,1H),7.87(s,1H),7.75(d,J=8.8Hz,1H),7.59(d,J=8.8Hz,1H),7.21(s,2H),7.16(s,2H),2.72(q,J=7.5Hz,2H),2.29(s,3H),1.21(t,J=7.5Hz,3H).LCMS:MS m/z(ESI):402.2[M+H]+。
实施例44(R)-3-氨基-2-(5-甲基-1H-吲唑-4-基)-1-氧代-1,2,6,7,8,9-六氢苯并[4,5]噻吩并
[3,2-c]吡啶-4-甲酰胺(化合物112-P1)和(S)-3-氨基-2-(5-甲基-1H-吲唑-4-基)-1-氧代-
1,2,6,7,8,9-六氢苯并[4,5]噻吩并[3,2-c]吡啶-4-甲酰胺(化合物112-P2)
44.1中间体112-2的合成:
室温下将2-溴-4,5,6,7-四氢苯并[b]噻吩-3-羧酸乙酯(2.50g)和5-甲基-1H-吲唑-4-胺(1.15g)溶于1,4-二氧六环(20.0mL)中,在搅拌下滴加三甲基铝(16.2mL,1.6M),用氩气置换三次,反应体系于封管中在120℃油浴中搅拌反应3小时。停止反应,将反应液倒入甲醇(25.0mL)中,用二氯甲烷(3×50.0mL)萃取,有机相经饱和食盐水(2×50.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物通过柱层析(石油醚:乙酸乙酯=15:1~1:1)纯化,得到中间体112-2(1.43g)。LCMS:MS m/z(ESI):390.1[M+H]+。
44.2中间体112-3的合成:
室温下将中间体112-2(1.28g)溶于乙二醇二甲醚(25.0mL)中,依次加入丙二腈(433mg),叔丁醇钠(630mg)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(240mg),用氩气置换3次后,反应体系在氩气保护下于100℃油浴中加热搅拌反应过夜。停止反应,向反应液中加入水(50.0mL)稀释,通过乙酸乙酯(3×50.0mL)萃取,有机相经饱和食盐水(2×50.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物通过甲醇(20.0mL)打浆纯化,得到中间体112-3(960mg)。LCMS:MS m/z(ESI):376.2[M+H]+。
44.3化合物112的合成:
室温下将中间体112-3(960mg)溶于乙醇(15.0mL)和水(5.0mL)中,加入(二甲基膦
酸)铂(II)氢化络合物(110mg),用氩气置换3次后,反应体系在氩气保护下于100℃油浴中搅拌反应过夜。停止反应,将反应液减压浓缩后,残余物通过柱层析(二氯甲烷:乙醇=4:1)纯化,得到化合物112(660mg)。LCMS:MS m/z(ESI):394.2[M+H]+。
44.4化合物112-P1和112-P2的分离:
将化合物112通过SFC手性拆分(手性柱:250*30mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in MeOH),A:B=65:35),得到两个异构体:(R)-3-氨基-2-(5-甲基-1H-吲唑-4-基)-1-氧代-1,2,6,7,8,9-六氢苯并[4,5]噻吩并[3,2-c]吡啶-4-甲酰胺(化合物112-P1,Rt=5.5min)和(S)-3-氨基-2-(5-甲基-1H-吲唑-4-基)-1-氧代-1,2,6,7,8,9-六氢苯并[4,5]噻吩并[3,2-c]吡啶-4-甲酰胺(化合物112-P2,Rt=10.2min)。
化合物112-P1:
1H NMR(400MHz,DMSO)δ13.25(s,1H),7.68(s,1H),7.62(d,J=8.6Hz,1H),7.42(d,J=8.6Hz,1H),7.14(s,2H),7.02(s,2H),2.80(t,J=6.8Hz,2H),2.67(t,J=6.3Hz,2H),2.09(s,3H),1.82–1.75(m,2H),1.73–1.68(m,2H).LCMS:MS m/z(ESI):394.2[M+H]+。
化合物112-P2:
1H NMR(400MHz,DMSO)δ13.26(s,1H),7.69(s,1H),7.63(d,J=8.4Hz,1H),7.42(d,J=8.4Hz,1H),7.15(s,2H),7.03(s,2H),2.81(t,J=6.5Hz,2H),2.68(t,J=5.9Hz,2H),2.10(s,3H),1.82–1.75(m,2H),1.75–1.67(m,2H).LCMS:MS m/z(ESI):394.1[M+H]+。
实施例45(S)-3-氨基-2-(5-氯-1H-吲唑-4-基)-1-氧代-1,2,6,7,8,9-六氢苯并[4,5]噻吩并
[3,2-c]吡啶-4-甲酰胺(化合物113-P1)和(R)-3-氨基-2-(5-氯-1H-吲唑-4-基)-1-氧代-
1,2,6,7,8,9-六氢苯并[4,5]噻吩并[3,2-c]吡啶-4-甲酰胺(化合物113-P2)
参考实施例44的合成方法,将5-甲基-1H-吲唑-4-胺替换为5-氯-1H-吲唑-4-胺,得到化合物3-氨基-2-(5-氯-1H-吲唑-4-基)-1-氧代-1,2,6,7,8,9-六氢苯并[4,5]噻吩并[3,2-c]4-吡啶-4-甲酰胺(化合物113)。
将化合物113通过SFC手性拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in
MeOH),A:B=65:35),得到两个异构体:(S)-3-氨基-2-(5-氯-1H-吲唑-4-基)-1-氧代-1,2,6,7,8,9-六氢苯并[4,5]噻吩并[3,2-c]吡啶-4-甲酰胺(化合物113-P1,Rt=5.1min)和(R)-3-氨基-2-(5-氯-1H-吲唑-4-基)-1-氧代-1,2,6,7,8,9-六氢苯并[4,5]噻吩并[3,2-c]吡啶-4-甲酰胺(化合物113-P2,Rt=7.7min)。
化合物113-P1:
1H NMR(400MHz,DMSO)δ13.52(s,1H),7.86(s,1H),7.75(d,J=8.8Hz,1H),7.59(d,J=8.8Hz,1H),7.24(s,2H),7.15(s,2H),2.78(t,J=6.7Hz,2H),2.66(t,J=6.7Hz,2H),1.80–1.73(m,2H),1.73–1.67(m,2H).LCMS:MS m/z(ESI):414.2[M+H]+。
化合物113-P2:
1H NMR(400MHz,DMSO)δ13.51(s,1H),7.85(s,1H),7.75(d,J=8.9Hz,1H),7.59(d,J=8.9Hz,1H),7.24(s,2H),7.15(s,2H),2.78(t,J=6.8Hz,2H),2.69–2.65(m,2H),1.80–1.75(m,2H),1.72–1.68(m,2H).LCMS:MS m/z(ESI):414.1[M+H]+。
实施例46(R)-6-氨基-2-环丙基-3-甲基-5-(5-甲基-1H-吲唑-4-基)-4-氧代-4,5-二氢噻吩并
[3,2-c]吡啶-7-甲酰胺(化合物114-P1)和(S)-6-氨基-2-环丙基-3-甲基-5-(5-甲基-1H-吲
唑-4-基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物114-P2)
46.1中间体114-2的合成:
室温下将中间体110-5(530mg)溶于二氯甲烷(5.0mL)中,加入3,4-二氢-2H-吡喃(215mg),冷却至0℃下加入对甲苯磺酸(22mg),反应体系升温至30℃搅拌反应1小时。停止反应,向反应液中加入水(10.0mL)稀释,用二氯甲烷(2×10.0mL)萃取,有机相依次经饱和碳酸氢钠溶液(2×10.0mL)、饱和氯化钠溶液(2×10.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物通过硅胶柱层析(石油醚:乙酸乙酯=5:1~3:1)纯化,得到中间体114-2(487mg)。LCMS:MS m/z(ESI):498.0[M+H]+。
46.2中间体114-3的合成:
室温下将中间体114-2(487mg)溶于1,4-二氧六环(5.0mL)和水(0.5mL)中,依次加入环丙基三氟硼酸钾(722mg)、碳酸钾(55mg),氟化铯(44mg)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(71mg),用氩气置换3次后,反应体系在氩气保护下微波100℃反应1小时。冷却至室温,向反应液中加入水(10.0mL)稀释,用乙酸乙酯(3×10.0mL)萃取,有机相经饱和食盐水(2×20.0mL)洗涤,无水硫酸钠干燥,减压浓缩,残余物通过Prep-TLC(石油醚:乙酸乙酯=1:1)纯化,得到中间体114-3(240mg)。LCMS:MS m/z(ESI):460.2[M+H]+。
46.3中间体114-4的合成:
室温下将中间体114-3(240mg)溶于盐酸的甲醇溶液中(5.0mL,4M)中,反应体系在25℃搅拌反应1小时。停止反应,将反应液浓缩后,加入四氢呋喃(10.0mL)溶解,加入水(10.0mL)稀释,用乙酸乙酯(3×10.0mL)萃取,有机相依次经饱和碳酸氢钠溶液(2×20.0mL)、饱和食盐水(2×20.0mL)洗涤,无水硫酸钠干燥,减压浓缩,得到中间体114-4(158mg)。LCMS:MS m/z(ESI):376.1[M+H]+。
46.4化合物114的合成:
室温下将中间体114-4(158mg)溶于乙醇(12.0mL)和水(4.0mL)中,加入(二甲基膦酸)铂(II)氢化络合物(18mg),用氩气置换3次后,反应体系在氩气保护下于110℃搅拌反应3小时。停止反应,将反应液减压浓缩后,通过高效液相制备色谱法纯化,得到化合物114(22.13mg)。LCMS:MS m/z(ESI):394.2[M+H]+。
46.5化合物114-P1和114-P2的分离:
将化合物114通过SFC手性拆分(手性柱:250*30mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in MeOH),A:B=70:30),得到两个异构体:(R)-6-氨基-2-环丙基-3-甲基-5-(5-甲基-1H-吲唑-4-基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物114-P1,Rt=4.2min)和(S)-6-氨基-2-环丙基-3-甲基-5-(5-甲基-1H-吲唑-4-基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物114-P2,Rt=6.2min)。
化合物114-P1:
1H NMR(400MHz,DMSO)δ13.25(s,1H),7.67(s,1H),7.61(d,J=8.5Hz,1H),7.41(d,J=8.5Hz,1H),7.13(s,2H),6.98(s,2H),2.39(s,3H),2.09(s,3H),2.07–1.96(m,1H),1.03–0.95(m,2H),0.69–0.63(m,2H).LCMS:MS m/z(ESI):394.2[M+H]+。
化合物114-P2:
1H NMR(400MHz,DMSO)δ13.24(s,1H),7.67(s,1H),7.61(d,J=8.5Hz,1H),7.41(d,J=8.5Hz,1H),7.13(s,2H),6.98(s,2H),2.39(s,3H),2.08(s,3H),2.00(td,J=8.4,4.3Hz,1H),1.04–0.95(m,2H),0.70–0.62(m,2H).LCMS:MS m/z(ESI):394.2[M+H]+。
实施例47(S)-6-氨基-5-(5-氯-1H-吲唑-4-基)-2-环丙基-3-甲基-4-氧代-4,5-二氢噻吩并
[3,2-c]吡啶-7-甲酰胺(化合物115-P1)和(R)-6-氨基-5-(5-氯-1H-吲唑-4-基)-2-环丙基-3-
甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物115-P2)
参考实施例42和实施例46的合成方法,将5-甲基-1H-吲唑-4-胺替换为5-氯-1H-吲唑-4-胺,最终得到6-氨基-5-(5-氯-1H-吲唑-4-基)-2-环丙基-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物115)。将化合物115通过SFC手性拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in MeOH),A:B=80:20),得到两个异构体:(S)-6-氨基-5-(5-氯-1H-吲唑-4-基)-2-环丙基-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物115-P1,Rt=3.8min)和(R)-6-氨基-5-(5-氯-1H-吲唑-4-基)-2-环丙基-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物115-P2,Rt=6.1min)。
化合物115-P1:
1H NMR(400MHz,DMSO)δ13.53(s,1H),7.87(s,1H),7.76(d,J=8.9Hz,1H),7.60(d,J=8.9Hz,1H),7.22(s,2H),7.15(s,2H),2.39(s,3H),2.01(td,J=8.2,4.6Hz,1H),1.05–0.96(m,2H),0.70–0.61(m,2H).LCMS:MS m/z(ESI):414.1[M+H]+。
化合物115-P2:
1H NMR(400MHz,DMSO)δ13.52(s,1H),7.87(s,1H),7.75(d,J=8.9Hz,1H),7.59(d,J=8.9Hz,1H),7.21(s,2H),7.15(s,2H),2.38(s,3H),2.06–1.95(m,1H),1.04–0.94(m,2H),
0.71–0.60(m,2H).LCMS:MS m/z(ESI):414.1[M+H]+。
实施例48(S)-6-氨基-2-氯-3-甲基-5-(5-甲基-1H-吲唑-4-基)-4-氧代-4,5-二氢噻吩并[3,2-
c]吡啶-7-甲酰胺(化合物116-P1)和(R)-6-氨基-2-氯-3-甲基-5-(5-甲基-1H-吲唑-4-基)-
4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物116-P2)
48.1中间体116-2的合成:
将中间体110-4(730mg)溶于乙腈(10mL)中,加入N-氯代丁二酰亚胺(350mg),将反应液加热到80℃反应16小时。冷却至室温,有固体析出,过滤,滤饼用乙腈洗涤,真空干燥后得中间体116-2(630mg)。LCMS:MS m/z(ESI):370.1[M+H]+。
48.2化合物116的合成:
将中间体116-2(690mg)溶于乙醇(10mL)和水(2mL),加入帕金斯催化剂(80mg),用氩气置换三次,反应液在100℃下反应16小时。冷却至室温,反应液减压浓缩,残余物用硅胶柱层析(DCM:MeOH=1:0~30:1)纯化,得到化合物6-氨基-2-氯-3-甲基-5-(5-甲基-1H-吲唑-4-基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物116)(350mg)。LCMS:MS m/z(ESI):388.0[M+H]+。
48.3化合物116-P1和116-P2的分离:
将化合物116通过SFC手性拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in
MeOH),A:B=70:30),得到两个异构体:(S)-6-氨基-2-氯-3-甲基-5-(5-甲基-1H-吲唑-4-基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物116-P1,Rt=5.7min)和(R)-6-氨基-2-氯-3-甲基-5-(5-甲基-1H-吲唑-4-基)-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物116-P2,Rt=9.6min)。
化合物116-P1:
1H NMR(400MHz,DMSO)δ13.26(s,1H),7.72(s,1H),7.63(d,J=8.5Hz,1H),7.42(d,J=8.5Hz,1H),7.25(s,2H),6.83(s,2H),2.33(s,3H),2.10(s,3H).LCMS:MS m/z(ESI):388.0[M+H]+。
化合物116-P2:
1H NMR(400MHz,DMSO)δ13.26(s,1H),7.73(s,1H),7.63(d,J=8.4Hz,1H),7.42(d,J=8.4Hz,1H),7.25(s,2H),6.84(s,2H),2.33(s,3H),2.10(s,3H).LCMS:MS m/z(ESI):388.0[M+H]+。
实施例49(R)-6-氨基-2-氯-5-(5-氯-1H-吲唑-4-基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]
吡啶-7-甲酰胺(化合物117-P1)和(S)-6-氨基-2-氯-5-(5-氯-1H-吲唑-4-基)-3-甲基-4-氧
代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物117-P2)
参照实施例42和实施例48的合成方法,将5-甲基-1H-吲唑-4-胺替换为5-氯-1H-吲唑-4-胺,最终得到6-氨基-2-氯-5-(5-氯-1H-吲唑-4-基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物117)。将化合物117通过SFC手性拆分(手性柱:250*30mm 10μm;流动相A:Supercritical CO2,流动相B:甲醇(含0.1%7.0mol/L Ammonia in MeOH),A:B=70:30),得到两个异构体:(R)-6-氨基-2-氯-5-(5-氯-1H-吲唑-4-基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物117-P1,Rt=3.5min)和(S)-6-氨基-2-氯-5-(5-氯-1H-吲唑-4-基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物117-P2,Rt=7.2min)。
化合物117-P1:
1H NMR(400MHz,DMSO)δ13.55(s,1H),7.92(s,1H),7.77(d,J=8.8Hz,1H),7.61(d,J=8.8Hz,1H),7.28(s,2H),7.10(s,2H),2.33(s,3H).LCMS:MS m/z(ESI):408.0[M+H]+。
化合物117-P2:
1H NMR(400MHz,DMSO)δ13.55(s,1H),7.92(s,1H),7.77(d,J=8.9Hz,1H),7.61(d,J=8.9Hz,1H),7.28(s,2H),7.10(s,2H),2.33(s,3H).LCMS:MS m/z(ESI):408.1[M+H]+。
实施例50(R)-6-氨基-5-(5-氯-1H-吲唑-4-基)-2,3-二甲基-4-氧代-4,5-二氢噻吩并[3,2-c]
吡啶-7-甲酰胺(化合物118-P1)和(S)-6-氨基-5-(5-氯-1H-吲唑-4-基)-2,3-二甲基-4-氧代
-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物118-P2)
参照实施例9、实施例43的合成方法,得到6-氨基-5-(5-氯-1H-吲唑-4-基)-2,3-二甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物118)。将化合物118通过SFC手性拆分(手性柱:250*25mm 10μm;流动相A:Supercritical CO2,流动相B:异丙醇(含0.1%7.0mol/L Ammonia in MeOH),A:B=80:20),得到两个异构体:(R)-6-氨基-5-(5-氯-1H-吲唑-4-基)-2,3-二甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物118-P1,Rt=2.7min)和(S)-6-氨基-5-(5-氯-1H-吲唑-4-基)-2,3-二甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物118-P2,Rt=6.9min)。
化合物118-P1:
1H NMR(400MHz,DMSO)δ13.53(s,1H),7.86(s,1H),7.75(d,J=8.8Hz,1H),7.60(d,J=8.8Hz,1H),7.20(s,2H),7.16(s,2H),2.29(s,3H),2.28(s,3H).LCMS:MS m/z(ESI):388.2[M+H]+。
化合物118-P2:
1H NMR(400MHz,DMSO)δ13.52(s,1H),7.85(s,1H),7.75(d,J=8.8Hz,1H),7.59(d,J=8.8Hz,1H),7.20(s,2H),7.15(s,2H),2.29(s,3H),2.27(s,3H).LCMS:MS m/z(ESI):388.1[M+H]+。
实施例51 7-氨基-6-(3-羟基-2,6-二甲基苯基)-2,3-二甲基-5-氧代-5,6-二氢-1,6-萘啶-8-甲
酰胺(化合物120)
51.1中间体120-2的合成:
将2-羟基-5,6-二甲基烟酸腈(3g)溶于硫酸(50%,15mL)中,150℃搅拌反应2小时。反应液室温下加水(100mL)稀释,有大量固体析出,过滤,将滤饼真空干燥,再加入石油醚和乙酸乙酯(20mL,1:1)打浆,所得固体过滤,滤饼干燥后得到中间体120-2(2.8g)。LCMS:MS m/z(ESI):168.1[M+H]+。
51.2中间体120-3的合成:
将中间体120-2(0.8g)溶于三氯氧磷(5mL)中,先室温搅拌0.5小时,再升温至100℃反应6.5小时。冷却至室温,反应液减压浓缩,加入甲苯(5mL),减压浓缩带出残留三氯氧磷,得到中间体120-3(954mg),直接用于下一步反应。
51.3中间体120-4的合成:
将3-((4-甲氧基苄基)氧基)-2,6-二甲基苯胺(1.08g)溶于吡啶(5mL)中,于0℃下加入中间体120-3(954mg)的甲苯(5mL)溶液,于25℃反应16小时。减压浓缩,残余物用硅胶柱层析分离(石油醚:乙酸乙酯:二氯甲烷=30:1:0到0:6:1)纯化,得到中间体120-4(960mg)。LCMS:MS m/z(ESI):425.2[M+H]+。
51.4中间体120-5的合成:
将中间体120-4(210mg)溶于N,N-二甲基甲酰胺(4mL)中,加入丙二腈(66mg)和碳酸钾(138mg),在氮气保护下于120℃反应16小时。停止反应,向反应液加水(30mL),用乙酸乙酯(30mLx3)萃取,有机相经饱和食盐水(30mLx3)洗涤,无水硫酸钠干燥,减压浓缩,残余物经Prep-TLC(石油醚:乙酸乙酯=2:1)纯化,得到中间体120-5(50mg)。LCMS:MS m/z(ESI):455.3[M+H]+。
51.5中间体120-6的合成:
将中间体120-5(50mg)溶于硫酸(1mL)中,反应体系在25℃下搅拌60分钟。停止反应,于0℃下用饱和碳酸氢钠溶液将反应液pH调至7,反应液加水(10mL)稀释,用二氯甲烷(10mLx3)萃取,有机相经饱和食盐水(10mL)洗涤,无水硫酸钠干燥,减压浓
缩,残余物经Prep-TLC(石油醚:乙酸乙酯=1.5:1)纯化,得到中间体120-6(20mg)。LCMS:MS m/z(ESI):335.2[M+H]+。
51.6化合物120的合成:
将中间体120-6(20mg)溶于乙醇(2mL)和水(0.5mL)中,加入(二甲基膦酸)铂(II)氢化络合物(5mg),在氮气保护下置换气三次,反应体系在120℃下搅拌16小时。反应液过滤,滤液减压浓缩,用甲醇(1mL)稀释,通过高效液相制备色谱法纯化,得到纯目标产物7-氨基-6-(3-羟基-2,6-二甲基苯基)-2,3-二甲基-5-氧代-5,6-二氢-1,6-萘啶-8-甲酰胺(化合物120)(1.01mg)。
1H NMR(400MHz,DMSO)δ10.89(d,J=5.0Hz,1H),9.60(s,1H),8.13(s,1H),7.26(d,J=5.0Hz,1H),7.09(d,J=8.2Hz,1H),6.90(d,J=8.2Hz,1H),2.56(s,3H),2.32(s,3H),1.85(s,3H),1.78(s,3H).LCMS:MS m/z(ESI):353.1[M+H]+。
实施例52 6-氨基-2-乙基-5-(7-氟-5-甲基-1H-吲唑-4-基)-3-甲基-4-氧代-4,5-二氢噻吩并
[3,2-c]吡啶-7-甲酰胺(化合物127)和(S)-6-氨基-2-乙基-5-(7-氟-5-甲基-1H-吲唑-4-基)-
3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物127-P1)
52.1中间体127-2的合成:
室温下将2,2,6,6-四甲基哌啶(30.0g)溶于超干的四氢呋喃(160.0mL)中,用氩气置换三次后,在-20℃下用恒压进料漏斗缓慢加入正丁基锂(84.0mL,2.5M),反应体系在0℃下搅拌反应0.5小时。冷却至-60℃下用恒压进料漏斗缓慢加入2-溴-4,5-二氟甲苯(40.0g)的超干四氢呋喃(240.0mL)溶液,并在-60℃下搅拌1小时,再缓慢加入超干N,N-二甲基甲酰胺(16.0mL),升温至-20℃搅拌反应1小时。停止反应,在0℃下向反应液中缓慢加入稀盐酸(400mL,1M)淬灭,用乙酸乙酯(3×400.0mL)萃取,有机相依次经饱和碳酸氢钠溶液(2×400.0mL)、饱和食盐水(2×300.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压
浓缩,残余物用石油醚(50.0mL)打浆,所得固体过滤,滤饼真空干燥后,得到中间体127-2(32.0g)。1H NMR(400MHz,CDCl3)δ10.35(dd,J=0.8Hz,1.7Hz,1H),7.31(dd,J=8.0Hz,10.3Hz,1H),2.44(s,3H)。19F NMR(400MHz,CDCl3)δ-137.39(d,J=19.6Hz),-143.27(d,J=20.4Hz)。
52.2中间体127-3的合成:
室温下将中间体127-2(32.0g)溶于二甲基亚砜(320.0mL)中,加入水合肼(81.8mL),120℃下搅拌反应8小时。停止反应,将反应液缓慢加入到水(3.2L)中搅拌1小时,用3M盐酸调节pH=5,有固体析出。过滤,干燥滤饼得到中间体127-3(23.7g)。LCMS:MS m/z(ESI):229.0[M+H]+。
52.3中间体127-4的合成:
室温下将中间体127-3(23.7g)溶于二氯甲烷(200.0mL)中,加入3,4-二氢-2H-吡喃(26.1g),降温至0℃下加入对甲苯磺酸(1.77g),然后反应体系在30℃下搅拌反应1小时。停止反应,向反应液中加入水(300.0mL)稀释,用二氯甲烷(3×100.0mL)萃取,有机相依次经饱和碳酸氢钠溶液(2×100.0mL)、饱和食盐水(2×100.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩后,残余物用硅胶柱层析(石油醚:乙酸乙酯=200:1~30:1)纯化,得到中间体127-4(28.2g)。1H NMR(400MHz,DMSO)δ8.10(d,J=1.6Hz,1H),7.34(d,J=12.8Hz,1H),5.81(d,J=1.6Hz,1H),3.73-3.62(m,2H),2.43(s,3H),2.07-2.04(m,2H),1.76-1.72(m,2H),1.57-1.44(m,2H)。19F NMR(400MHz,DMSO)δ-133.65(s)。
52.4中间体127-5的合成:
室温下将中间体127-4(400mg)溶于1,4-二氧六环(100.0mL)中,依次加入二苯甲酮亚胺(13.9g),碳酸铯(37.4g),4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(4.40g)和醋酸钯(868mg),用氩气置换3次,反应体系在110℃反应16小时。停止反应,向反应液中加入水(150.0mL)稀释,通过乙酸乙酯(3×150.0mL)萃取。有机相经饱和食盐水(2×150.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物用硅胶柱层析(石油醚:乙酸乙酯=50:1~20:1)纯化,得到中间体127-5(10.5g)。LCMS:MS m/z(ESI):414.3[M+H]+。
52.5中间体127-6的合成:
室温下将中间体127-5(10.5g)溶于盐酸甲醇溶液(100.0mL,4M)中,反应体系在25℃搅拌反应1小时。停止反应,将反应液浓缩后,用石油醚(60.0mL)打浆,所得固体过滤,滤饼干燥后得到中间体127-6(4.0g)。LCMS:MS m/z(ESI):166.3[M+H]+。
52.6中间体127-7的合成:
室温下将中间体111-4(1.50g)和中间体127-6(1.04g)溶于甲苯(12.0mL)中,在搅拌下滴加三甲基铝(10.7mL,1.6M),用氩气置换三次,反应体系在100℃搅拌反应1小时。停止反应,将反应液倒入甲醇(40.0mL)中,将溶剂浓缩后,加入四氢呋喃(30.0mL)溶解,加入乙酸乙酯(30.0mL)稀释,用饱和食盐水(2×30.0mL)洗涤,无水硫酸钠干燥后过滤。滤液减压浓缩,残余物用石油醚和乙酸乙酯(3:1)打浆,所得固体过滤,滤饼干燥后得到中间体127-7(1.64g)。LCMS:MS m/z(ESI):396.1[M+H]+。
52.7中间体127-8的合成:
室温下将丙二腈(547mg)溶于乙二醇二甲醚(20.0mL)中,加入氢化钠(331mg),室温下搅拌反应20分钟,再加入中间体127-7(1.64g)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(303mg),用氩气置换3次后,反应体系在氩气保护下于110℃加热搅拌反应16小时。停止反应,向反应液中加入水(40.0mL)稀释,通过乙酸乙酯(3×40.0mL)萃取,有机相经饱和食盐水(2×40.0mL)洗涤,无水硫酸钠干燥后过滤,滤液减压浓缩,残余物用硅胶柱层析(石油醚:乙酸乙酯=1:0~3:1)纯化,得到中间体127-8(90mg)。LCMS:MS m/z(ESI):382.2[M+H]+。
52.8化合物127的合成:
室温下将中间体127-8(90mg)溶于乙醇(3.0mL)和水(1.0mL)中,加入(二甲基膦酸)铂(II)氢化络合物(10mg),用氩气置换3次后,反应体系在氩气保护下于100℃搅拌反应16小时。停止反应,将反应液减压浓缩,残余物用甲醇(3.0mL)打浆纯化,得到6-氨基-2-乙基-5-(7-氟-5-甲基-1H-吲唑-4-基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物127)(32.69mg)。1H NMR(400MHz,DMSO)δ13.86(s,1H),7.85(s,1H),7.38(d,J=11.5Hz,1H),7.20(s,2H),7.17(s,2H),2.77(q,J=7.5Hz,2H),2.36(s,3H),2.12(s,3H),1.26(t,J=7.5Hz,3H).19F NMR(400MHz,DMSO)δ-130.61(s)。LCMS:MS m/z(ESI):400.2[M+H]+。
将化合物127通过SFC手性拆分,得到化合物(S)-6-氨基-2-乙基-5-(7-氟-5-甲基-1H-吲唑-4-基)-3-甲基-4-氧代-4,5-二氢噻吩并[3,2-c]吡啶-7-甲酰胺(化合物127-P1)。
1H NMR(400MHz,DMSO)δ13.86(s,1H),7.85(s,1H),7.38(d,J=11.2Hz,1H),7.20(s,2H),7.17(s,2H),2.77(q,J=7.6Hz,2H),2.36(s,3H),2.12(s,3H),1.26(t,J=7.5Hz,3H).LCMS:MS m/z(ESI):400.1[M+H]+。
实施例53化合物绝对构型的确证
该系列化合物结构中具有阻转异构特性,能够通过手性SFC拆分获得两个稳定的异构体。通过前述的生物测试例结果可知,两个异构体无论在酶学抑制活性还是细胞增殖抑制活性中,均表现出较大的活性差异。选取两个代表性分子化合物047和112,对其抑制活性较差的异构体开展单晶培养和结构解析,以确定其绝对构型。
单晶培养方法包括:气液扩散和溶剂缓慢挥发结晶。其中,化合物047-2,在乙醇和正戊烷体系通过气液扩散,获得板状晶体;化合物112-P1,在乙醇和正戊烷体系通过溶剂缓慢挥发,获得块状晶体。
测试仪器:偏振光显微镜(PLM),型号:Primotech。单晶X射线衍射仪(SCXRD),型号:Bruker D8 Venture。
单晶结构解析数据见下表。根据轴手性化合物R/S构型定义方法,两个测试化合物均为R构型。由此结果可以推测,抑制活性较好的异构体为S构型。本发明通过获取扭转异构体,进一步提高了化合物的活性。
单晶结构解析数据表
参考上述实施例的合成方法,合成了化合物035、061-090,100、105、119和121-125,下表1给出了这些化合物的结构、名称以及表征数据。
表1
表2中列出的化合物005、008、011、015、018-020、022、025-034、037-042、091-104、106-107、126、128-170基本上按照与实施例1-52相同的方法制备,使用的起始物料是商购或者根据文献方法制得。表2给出了这些化合物的名称及结构。
表2
效果例1PKMYT1酶学活性抑制实验
化合物对PKMYT1激酶的抑制由ADP-Glo实验检测得出。首先配制反应缓冲液(Assay buffer):50mM HEPES,10mM MgCl,1mM EGTA,0.01%Brij-35,2mM DTT,然后用反应缓冲液分别稀释MYT1酶(Carna,05-176,终浓度为20nM)和反应底物(终浓度为40μM的ATP和终浓度为40nM的未激活的CDK1,Signalchem C22-14G)。将待测化合物3倍梯度稀释成10个浓度,起始浓度为3μM。将稀释好的化合物转移50nL到384孔板中,转移5μL MYT1酶溶液到384孔板中,25℃孵育10分钟。转移5μL反应底物溶液到384孔板中,25℃孵育180分钟。然后转移5μL ADP-Glo Reagent(Promega,V9101)试剂到384孔板中,25分钟孵育40分钟,最后转移10μL ADP-Glo Detection试剂到384孔板中,25℃孵育40分钟。使用酶标仪(BMG)来测量样品的发光强度。采用XLfit 5.5.0软件建立非线性回归方程,进行浓度-响应曲线拟合和IC50测定。实验结果见表3。
表3化合物PKMYT1酶学活性测试结果
效果例2 WEE1酶学活性抑制实验
化合物对WEE1激酶的抑制由ADP-Glo实验检测得出。首先配制反应缓冲液(Assay buffer):50mM HEPES,10mM MgCl,1mM EGTA,0.01%Brij-35,2mM DTT,然后
用反应缓冲液分别稀释WEE1酶(Carna,05-177,终浓度为10nM)和反应底物(终浓度为20μM的ATP和终浓度为400ng/μL的Poly(Lys-Tyr,4:1),BPS Bioscience,79910)。将待测化合物3倍梯度稀释成10个浓度,起始浓度为3μM。将稀释好的化合物转移25nL到384孔板中,转移2.5μL WEE1酶溶液到384孔板中,25℃孵育10分钟。转移2.5μL反应底物溶液到384孔板中,25℃孵育180分钟。然后转移4μL ADP-Glo Reagent(Promega,V9101)试剂到384孔板中,25分钟孵育40分钟,最后转移8μL ADP-Glo Detection试剂到384孔板中,25℃孵育40分钟。使用酶标仪(BMG)来测量样品的发光强度。采用XLfit 5.5.0软件建立非线性回归方程,进行浓度-响应曲线拟合和IC50测定。实验结果见表4。
表4化合物WEE1酶学活性测试结果
效果例3肿瘤细胞增殖抑制试验
制备10000个细胞/mL的HCC1569细胞(ATCC,CRL-2330)混悬液,并转移195μL至96孔透底的平底细胞培养板(Corning,#3603)中,37℃、5% CO2条件下过夜培养。将待测化合物储液先使用DMSO稀释至中间浓度,并按1:3倍比稀释成10个浓度点。再转移5μL稀释好的化合物溶液至过夜孵育好的细胞孔中,起始最高浓度为10μM。于37℃、5%CO2条件下孵育7天。在第八天时,从培养箱中取出细胞培养板,让其恢复至室温,加入100μL的Celltiter-Glo试剂(Promega,G7573),振荡2分钟至完全混匀,然后再室温下避光孵育30min。用BMG酶标仪(BMG,PHERAstar FSX)进行定量。采用XLfit 5.5.0软件建立非线性回归方程,进行浓度-响应曲线拟合和IC50测定。测试结果见表5。
表5化合物对于HCC1569细胞增殖的抑制活性测试结果
效果例4小鼠体内药代动力学实验
实验材料:CD-1小鼠购自浙江维通利华实验动物技术有限公司、斯贝福(北京)生物技术有限公司和斯贝福(苏州)生物技术有限公司。DMSO、Solutol、PEG400、乙腈、
甲醇、VETPGS购自Sigma-Aldrich公司等。
实验设备:LC-MS/MS系统为Waters Acquity UPLC class I plus串联AB Sciex Triple Quad 6500+,色谱柱为Agilent Poroshell 120EC-C18 4μm(50×2.1mm)。所有数据由Analyst软件采集并处理,使用Phoenix Build 8.3进行药代动力学参数计算。
实验方法:雌性CD-1小鼠6只(20~30g,6-8周),根据体重随机分成2组,每组3只。第1组尾静脉注射给予待测化合物,剂量为1mg/kg或者0.5mg/kg,溶媒为5%DMSO+5% Solutol+90% Saline溶液;第2组口服给予待测化合物,剂量为2mg/kg或者5mg/kg,溶媒为5%DMSO/20% PEG400/75%(15%VETPGS的水溶液)。第1组动物实验前正常喂食喂水,第2组动物实验前禁食过夜,给药2hr后加食。小鼠于给药前及给药后0.083(仅静脉注射组)、0.25、0.5、1、2、4、8和24hr于足背静脉采血。采集的全血样品置于K2·EDTA抗凝管中,离心(4000g,5min,4℃)制备血浆,血浆样品保存于-75±15℃超低温冰箱直至检测。取小鼠血浆样品加入含内标化合物的乙腈溶液并涡旋0.5min,随后于3900rpm离心15min,转移离心所得上清液并用水溶液稀释3倍,进样2μL至LC-MS/MS系统进行定量分析。在测定样品浓度时随行雌性CD-1小鼠血浆标准曲线(线性范围:0.5~1000ng/mL)和质控样品(1,2,5,50,400,800ng/mL)。部分药代动力学测试结果见表7。
表7小鼠药代动力学测试结果
效果例5体内药效实验:小鼠HCC1569皮下移植瘤模型
选取本发明部分优选化合物进行小鼠体内药效研究。HCC1569为人乳腺癌细胞,采用NOD SCID雌性小鼠建立模型,将0.2mL(3×106个)HCC1569细胞皮下接种于每只小鼠的右后背,在肿瘤平均体积达到100-150mm3时开始分组给药,每天经口给药两次化合物。每周两次以及给药终点测量肿瘤直径。肿瘤体积的计算公式为:V=0.5a×b2,a和b分别表示肿瘤的长径和短径。化合物的抑瘤疗效用肿瘤生长抑制率(TGI%)评价。其中TGI(%)=[(1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积))/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)]×100%。
Claims (23)
- 式(I)的化合物:
或其药学上可接受的盐,其中,环A选自苯环、C5-6烃环、5-6元杂环和5-6元杂芳环;E选自O和S;R1选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3- 10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA1RB1、-ORA1、-SRA1、-C(=O)RA1、-C(=NRE1)RA1、-C(=N-ORB1)RA1、-C(=O)ORA1、-OC(=O)RA1、-C(=O)NRA1RB1、-NRA1C(=O)RB1、-C(=NRE1)NRA1RB1、-NRA1C(=NRE1)RB1、-OC(=O)NRA1RB1、-NRA1C(=O)ORB1、-NRA1C(=O)NRA1RB1、-NRA1C(=S)NRA1RB1、-NRA1C(=NRE1)NRA1RB1、-S(=O)rRA1、-S(=O)(=NRE1)RB1、-N=S(=O)RA1RB1、-S(=O)2ORA1、-OS(=O)2RA1、-NRA1S(=O)rRB1、-NRA1S(=O)(=NRE1)RB1、-S(=O)rNRA1RB1、-S(=O)(=NRE1)NRA1RB1、-NRA1S(=O)2NRA1RB1和-NRA1S(=O)(=NRE1)NRA1RB1,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX1的取代基取代;R2选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3- 10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA2RB2、-ORA2、-SRA2、-C(=O)RA2、-C(=NRE2)RA2、-C(=N-ORB2)RA2、-C(=O)ORA2、-OC(=O)RA2、-C(=O)NRA2RB2、-NRA2C(=O)RB2、-C(=NRE2)NRA2RB2、-NRA2C(=NRE2)RB2、-OC(=O)NRA2RB2、-NRA2C(=O)ORB2、-NRA2C(=O)NRA2RB2、-NRA2C(=S)NRA2RB2、-NRA2C(=NRE2)NRA2RB2、-S(=O)rRA2、-S(=O)(=NRE2)RB2、-N=S(=O)RA2RB2、-S(=O)2ORA2、-OS(=O)2RA2、-NRA2S(=O)rRB2、-NRA2S(=O)(=NRE2)RB2、-S(=O)rNRA2RB2、-S(=O)(=NRE2)NRA2RB2、-NRA2S(=O)2NRA2RB2和-NRA2S(=O)(=NRE2)NRA2RB2,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX2的取代基取代;每个R3独立选自卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烃基、-C1-4亚烷基-C3-10环烃基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA3RB3、-ORA3、-SRA3、-C(=O)RA3、-C(=NRE3)RA3、-C(=N- ORB3)RA3、-C(=O)ORA3、-OC(=O)RA3、-C(=O)NRA3RB3、-NRA3C(=O)RB3、-C(=NRE3)NRA3RB3、-NRA3C(=NRE3)RB3、-OC(=O)NRA3RB3、-NRA3C(=O)ORB3、-NRA3C(=O)NRA3RB3、-NRA3C(=S)NRA3RB3、-NRA3C(=NRE3)NRA3RB3、-S(=O)rRA3、-S(=O)(=NRE3)RB3、-N=S(=O)RA3RB3、-S(=O)2ORA3、-OS(=O)2RA3、-NRA3S(=O)rRB3、-NRA3S(=O)(=NRE3)RB3、-S(=O)rNRA3RB3、-S(=O)(=NRE3)NRA3RB3、-NRA3S(=O)2NRA3RB3和-NRA3S(=O)(=NRE3)NRA3RB3,其中每个烷基、亚烷基、烯基、炔基、环烃基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX3的取代基取代;或任意两个相邻的R3连同与它们相连的原子一起形成一个C3-10单环烃环或者含1、2或3个杂原子的4-12元杂环或者含1、2或3个杂原子的5-10元杂芳环或者苯环,其中杂原子独立选自氧、硫、氮和磷,该环是未被取代的或被至少一个独立选自RX3的取代基取代;R4选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3- 10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA4RB4、-ORA4、-SRA4、-C(=O)RA4、-C(=NRE4)RA4、-C(=N-ORB4)RA4、-C(=O)ORA4、-OC(=O)RA4、-C(=O)NRA4RB4、-NRA4C(=O)RB4、-C(=NRE4)NRA4RB4、-NRA4C(=NRE4)RB4、-OC(=O)NRA4RB4、-NRA4C(=O)ORB4、-NRA4C(=O)NRA4RB4、-NRA4C(=S)NRA4RB4、-NRA4C(=NRE4)NRA4RB4、-S(=O)rRA4、-S(=O)(=NRE4)RB4、-N=S(=O)RA4RB4、-S(=O)2ORA4、-OS(=O)2RA4、-NRA4S(=O)rRB4、-NRA4S(=O)(=NRE4)RB4、-S(=O)rNRA4RB4、-S(=O)(=NRE4)NRA4RB4、-NRA4S(=O)2NRA4RB4和-NRA4S(=O)(=NRE4)NRA4RB4,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX4的取代基取代;R5选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3- 10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA5RB5、-ORA5、-SRA5、-C(=O)RA5、-C(=NRE5)RA5、-C(=N-ORB5)RA5、-C(=O)ORA5、-OC(=O)RA5、-C(=O)NRA5RB5、-NRA5C(=O)RB5、-C(=NRE5)NRA5RB5、-NRA5C(=NRE5)RB5、-OC(=O)NRA5RB5、-NRA5C(=O)ORB5、-NRA5C(=O)NRA5RB5、-NRA5C(=S)NRA5RB5、-NRA5C(=NRE5)NRA5RB5、-S(=O)rRA5、-S(=O)(=NRE5)RB5、-N=S(=O)RA5RB5、-S(=O)2ORA5、-OS(=O)2RA5、-NRA5S(=O)rRB5、-NRA5S(=O)(=NRE5)RB5、-S(=O)rNRA5RB5、-S(=O)(=NRE5)NRA5RB5、-NRA5S(=O)2NRA5RB5和-NRA5S(=O)(=NRE5)NRA5RB5,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX5的取代基取代;R6选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3- 10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA6RB6、-ORA6、-SRA6、-C(=O)RA6、-C(=NRE6)RA6、-C(=N-ORB6)RA6、-C(=O)ORA6、-OC(=O)RA6、-C(=O)NRA6RB6、-NRA6C(=O)RB6、-C(=NRE6)NRA6RB6、- NRA6C(=NRE6)RB6、-OC(=O)NRA6RB6、-NRA6C(=O)ORB6、-NRA6C(=O)NRA6RB6、-NRA6C(=S)NRA6RB6、-NRA6C(=NRE6)NRA6RB6、-S(=O)rRA6、-S(=O)(=NRE6)RB6、-N=S(=O)RA6RB6、-S(=O)2ORA6、-OS(=O)2RA6、-NRA6S(=O)rRB6、-NRA6S(=O)(=NRE6)RB6、-S(=O)rNRA6RB6、-S(=O)(=NRE6)NRA6RB6、-NRA6S(=O)2NRA6RB6和-NRA6S(=O)(=NRE6)NRA6RB6,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX6的取代基取代;R7选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3- 10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA7RB7、-ORA7、-SRA7、-C(=O)RA7、-C(=NRE7)RA7、-C(=N-ORB7)RA7、-C(=O)ORA7、-OC(=O)RA7、-C(=O)NRA7RB7、-NRA7C(=O)RB7、-C(=NRE7)NRA7RB7、-NRA7C(=NRE7)RB7、-OC(=O)NRA7RB7、-NRA7C(=O)ORB7、-NRA7C(=O)NRA7RB7、-NRA7C(=S)NRA7RB7、-NRA7C(=NRE7)NRA7RB7、-S(=O)rRA7、-S(=O)(=NRE7)RB7、-N=S(=O)RA7RB7、-S(=O)2ORA7、-OS(=O)2RA7、-NRA7S(=O)rRB7、-NRA7S(=O)(=NRE7)RB7、-S(=O)rNRA7RB7、-S(=O)(=NRE7)NRA7RB7、-NRA7S(=O)2NRA7RB7和-NRA7S(=O)(=NRE7)NRA7RB7,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX7的取代基取代;R8选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3- 10环烷基、杂环基、-C1-4亚烷基-杂环基、CN、NO2、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、-NRA8RB8、-ORA8、-SRA8、-C(=O)RA8、-C(=NRE8)RA8、-C(=N-ORB8)RA8、-C(=O)ORA8、-OC(=O)RA8、-C(=O)NRA8RB8、-NRA8C(=O)RB8、-C(=NRE8)NRA8RB8、-NRA8C(=NRE8)RB8、-OC(=O)NRA8RB8、-NRA8C(=O)ORB8、-NRA8C(=O)NRA8RB8、-NRA8C(=S)NRA8RB8、-NRA8C(=NRE8)NRA8RB8、-S(=O)rRA8、-S(=O)(=NRE8)RB8、-N=S(=O)RA8RB8、-S(=O)2ORA8、-OS(=O)2RA8、-NRA8S(=O)rRB8、-NRA8S(=O)(=NRE8)RB8、-S(=O)rNRA8RB8、-S(=O)(=NRE8)NRA8RB8、-NRA8S(=O)2NRA8RB8和-NRA8S(=O)(=NRE8)NRA8RB8,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX8的取代基取代;或R7和R8连同与它们相连的原子一起形成一个C5-6烃环或含1、2或3个杂原子的5-6元杂环或5-6元杂芳环,其中杂原子独立选自氧、硫和氮,该环是未被取代的或被至少一个独立选自RX7的取代基取代;每个RA1、RA2、RA3、RA4、RA5、RA6、RA7、RA8、RB1、RB2、RB3、RB4、RB5、RB6、RB7和RB8独立选自氢、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3- 10环烷基、杂环基、-C1-4亚烷基-杂环基、芳基、-C1-4亚烷基-芳基、杂芳基和-C1-4亚烷基-杂芳基,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RX的取代基取代;或“RA1和RB1”或“RA2和RB2”或“RA3和RB3”或“RA4和RB4”或“RA5和RB5”或“RA6和 RB6”或“RA7和RB7”或“RA8和RB8”一起连同与它们相连的单个或多个原子共同构成一个含有0、1或2个额外的独立选自氧、硫、氮和磷的杂原子的4-12元杂环,该环是未被取代的或被1、2或3个选自RX的取代基取代;每个RE1、RE2、RE3、RE4、RE5、RE6、RE7和RE8独立选自氢、C1-10烷基、CN、NO2、-S(=O)rRa1、-C(=O)Ra1、-C(=O)ORa1、-C(=O)NRa1Rb1和-S(=O)rNRa1Rb1,其中烷基是未被取代的或被至少一个独立选自RX的取代基取代;每个RX、RX1、RX2、RX3、RX4、RX5、RX6、RX7和RX8独立选自卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、芳基、-C1-4亚烷基-芳基、杂芳基、-C1-4亚烷基-杂芳基、CN、NO2、-(CRc1Rd1)tNRa1Rb1、-(CRc1Rd1)tORb1、-(CRc1Rd1)tC(=O)Ra1、-(CRc1Rd1)tC(=NRe1)Ra1、-(CRc1Rd1)tC(=O)ORb1、-(CRc1Rd1)tOC(=O)Rb1、-(CRc1Rd1)tC(=O)NRa1Rb1、-(CRc1Rd1)tNRa1C(=O)Rb1、-(CRc1Rd1)tC(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)Rb1、-(CRc1Rd1)tOC(=O)NRa1Rb1、-(CRc1Rd1)tNRa1C(=O)ORb1、-(CRc1Rd1)tNRa1C(=O)NRa1Rb1、-(CRc1Rd1)tNRa1C(=S)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)NRa1Rb1、-(CRc1Rd1)tS(=O)rRb1、-(CRc1Rd1)tS(=O)(=NRe1)Rb1、-(CRc1Rd1)tN=S(=O)Ra1Rb1、-(CRc1Rd1)tS(=O)2ORb1、-(CRc1Rd1)tOS(=O)2Rb1、-(CRc1Rd1)tNRa1S(=O)rRb1、-(CRc1Rd1)tNRa1S(=O)(=NRe1)Rb1、-(CRc1Rd1)tS(=O)rNRa1Rb1、-(CRc1Rd1)tS(=O)(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1S(=O)2NRa1Rb1和-(CRc1Rd1)tNRa1S(=O)(=NRe1)NRa1Rb1,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RY的取代基取代;每个Ra1和Rb1独立选自氢、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、芳基、-C1-4亚烷基-芳基、杂芳基和-C1-4亚烷基-杂芳基,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RY的取代基取代;或Ra1和Rb1一起连同与它们相连的单个或多个原子共同构成一个含有0、1或2个额外的独立选自氧、硫、氮和磷的杂原子的4-12元杂环,该环是未被取代的或被1、2或3个选自RY的取代基取代;每个Rc1和Rd1独立选自氢、卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烷基、-C1-4亚烷基-C3-10环烷基、杂环基、-C1-4亚烷基-杂环基、芳基、-C1-4亚烷基-芳基、杂芳基和-C1-4亚烷基-杂芳基,其中每个烷基、亚烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基是未被取代的或被至少一个独立选自RY的取代基取代;或者每个Rc1和Rd1一起连同与它们相连的单个或多个碳原子构成含有0、1或2个独立选自氧、硫和氮的杂原子的3-12元环,该环是未被取代的或被1、2或3个独立选自RY的基团取代;每个Re1独立选自氢、C1-10烷基、CN、NO2、-S(=O)rRa1、-C(=O)Ra1、-C(=O)ORa1、-C(=O)NRa1Rb1和-S(=O)rNRa1Rb1,其中烷基是未被取代的或被至少一个独立选自RY的 取代基取代;每个RY独立选自卤素、NO2、-CN、C1-10烷基、-OH、-O(C1-10烷基)、-O(C3-10环烷基)、-O(C1-4亚烷基-C3-10环烷基)、-O(杂环基)、-O(C1-4亚烷基-杂环基)、-SH、-S(C1-10烷基)、-S(C3-10环烷基)、-S(C1-4亚烷基-C3-10环烷基)、-S(杂环基)、-S(C1-4亚烷基-杂环基)、-NH2、-NH(C1-10烷基)、-N(C1-10烷基)2、-NH(C3-10环烷基)、-NH(C1-4亚烷基-C3-10环烷基)、-NH(杂环基)和-NH(C1-4亚烷基-杂环基);g选自0、1、2、3、4、5、6、7和8;每个r独立选自1和2;每个t独立选自0、1、2、3和4。 - 权利要求1的化合物,其中R1选自C1-10烷基、-NRA1RB1、-ORA1、-SRA1、-OC(=O)RA1、-NRA1C(=O)RB1、-OS(=O)2RA1和-NRA1S(=O)rRB1,其中每个烷基是未被取代的或被至少一个独立选自RX1的取代基取代;优选地,R1选自-NRA1RB1和氢,特别是-NH2。
- 权利要求1或2的化合物,其中R2选自CN、-C(=O)RA2、-C(=O)NRA2RB2和-S(=O)rNRA2RB2,优选-C(=O)NH2。
- 权利要求1-3中任一项的化合物,其中每个R3独立选自卤素、C1-10烷基、C2-10烯基、C2-10炔基、C3-10环烃基、杂环基、CN、杂芳基、-NRA3RB3、-ORA3、-SRA3、-C(=O)RA3、-C(=O)NRA3RB3和-S(=O)rRA3,其中每个烷基、烯基、炔基、环烃基、杂环基和杂芳基是未被取代的或被至少一个独立选自RX3的取代基取代。
- 权利要求1-4中任一项的化合物,其中每个R3独立选自F、Cl、Br、CN、OH、甲基、乙基、异丙基、叔丁基、环丙烷基、氮杂环丁烷基、环己烯基、哌啶基、吗啉基、氧代哌嗪基、吡啶基、噻唑基、吡唑基、二乙基胺基、氨基、乙烯基、乙炔基、乙酰基、氨基甲酰基、甲氧基、乙氧基、甲硫基、乙硫基、异丙硫基、甲磺酰基和苯基,其中所述甲基、乙基、异丙基、叔丁基、环丙烷基、氮杂环丁烷基、环己烯基、哌啶基、吗啉基、氧代哌嗪基、吡啶基、噻唑基、吡唑基、二乙基胺基、乙烯基、乙炔基、乙酰基、甲氧基、乙氧基、甲硫基、乙硫基、异丙硫基、甲磺酰基和苯基是未被取代的或被至少一个独立选自RX3的取代基取代。
- 权利要求5的化合物,其中每个R3独立选自F、Cl、Br、CN、OH、甲基、乙基、异丙基、叔丁基、环丙烷基、二乙基胺基、氨基、乙烯基、乙炔基、乙酰基、氨基甲酰基、甲氧基、甲硫基、乙硫基、 异丙硫基、甲磺酰基、苯基、
- 权利要求1-3中任一项的化合物,其中任意两个相邻的R3连同与它们相连的原子一起形成选自以下的环:环己烯、环己烷、二氢吡喃、四氢吡喃、环己烯酮、环己酮、二氢吡啶酮、哌啶酮、四氢吡啶、哌啶、环戊烯、环戊烷、二氢噻吩、四氢噻吩、吡啶和二氢吡啶,该环是未被取代的或被至少一个独立选自RX3的取代基取代。
- 权利要求7的化合物,其中任意两个相邻的R3连同与它们相连的原子一起形成选自以下的结构:
- 权利要求1-7中任一项的化合物,其中R4选自C1-10烷基和卤素,其中每个烷基是未被取代的或被至少一个独立选自RX4的取代基取代;优选地,R4为甲基或Cl。
- 权利要求1-9中任一项的化合物,其中R5选自氢、卤素、C1-10烷基、CN、NO2、-NRA5RB5和-ORA5,其中每个烷基是未被取代的或被至少一个独立选自RX5的取代基取代;优选地,R5为氢或F,特别是氢。
- 权利要求1-10中任一项的化合物,其中R6选自氢、卤素、C1-10烷基、CN、NO2、-NRA6RB6和-ORA6,其中每个烷基是未被取代的或被至少一个独立选自RX6的取代基取代;优选地,R6为氢或F,特别是氢。
- 权利要求1-11中任一项的化合物,其中R7选自卤素、C1-10烷基、CN、NO2、-NRA7RB7和-ORA7,优选-OH。
- 权利要求1-12中任一项的化合物,其中R8选自氢、C1-10烷基和卤素,优选选自C1-10烷基和卤素,其中每个烷基是未被取代的或被至少一个独立选自RX8的取代基取代;优选地,R8为甲基或Cl。
- 权利要求1-13中任一项的化合物,其中(i)环A选自5元杂芳环和环戊烯,并且式(I)具有如下式(II)的结构:
其中,A1选自N(R3a)、N、O、S、C(R3aR3a')和C(R3a);A2选自N(R3b)、N、O、S、C(R3bR3b')和C(R3b);A3选自N(R3c)、N、O、S、C(R3cR3c')和C(R3c);R3a、R3a'、R3b、R3b'、R3c和R3c'各自独立选自氢和R3;为单键或双键;或者(ii)环A选自苯环、环己烯和6元杂芳环,并且式(I)具有如下式(III)的结构:
其中,A4选自N、C(R3dR3d')和C(R3d);A5选自N、C(R3eR3e')和C(R3e);A6选自N、C(R3fR3f')和C(R3f);A7选自N、C(R3gR3g')和C(R3g);R3d、R3d'、R3e、R3e'、R3f、R3f'、R3g和R3g'各自独立选自氢和R3;为单键或双键。 - 权利要求14的化合物,其具有如下式(II-1)或(III-1)的结构:
其中,X1选自N(R9a)、N、O、S、C(R9aR9a')和C(R9a);X2选自N(R9b)、N、O、S、C(R9bR9b')和C(R9b);X3选自N(R9c)、N、O、S、C(R9cR9c')和C(R9c);R9a、R9a'、R9b、R9b'、R9c和R9c'各自独立选自氢和RX;为单键或双键。 - 权利要求1-13中任一项的化合物,其中环A选自5元杂芳环和环戊烯,优选选自噻吩、噻唑、吡唑、咪唑、吡咯、呋喃和环戊烯,其被g个独立选自R3的取代基取代。
- 权利要求1-13中任一项的化合物,其中环A选自苯环、吡啶和环己烯,其被g个独立选自R3的取代基取代。
- 权利要求1-17中任一项的的化合物,其具有如下式(Ia)、(Ia-1)、(IIa)、(IIa-1)、(IIIa)或(IIIa-1)的结构:
- 化合物,选自:
- 一种药物组合物,其包含预防或治疗有效量的权利要求1-19中任一项的化合物或其药学上可接受的盐,以及至少一种药学上接受的载体。
- 权利要求1-19中任一项的化合物或其药学上可接受的盐或者权利要求20的药物组合物在在制备治疗疾病、病症或病况的药物中的用途,所述疾病、病症或病况选自细胞增殖异常疾病。
- 权利要求1-19中任一项的化合物或其药学上可接受的盐或者权利要求20的药物组合物在在制备治疗疾病、病症或病况的药物中的用途,其中所述疾病、病症或病况为癌症;优选地,所述癌症选自:(a)选自以下癌症的实体瘤或血液源性肿瘤:膀胱癌、子宫内膜癌、肺鳞细胞癌、乳腺癌、结肠癌、肾癌、肝癌、肺癌、小细胞肺癌、食道癌、胆囊癌、脑癌、头颈癌、卵巢癌、胰腺癌、胃癌、宫颈癌、甲状腺癌、前列腺癌和皮肤癌;(b)选自以下的淋巴系的造血肿瘤:白血病、急性淋巴细胞白血病、急性成淋巴细胞白血病、B细胞淋巴瘤、T细胞淋巴瘤、霍奇金淋巴瘤、非霍奇金淋巴瘤、毛细胞淋巴瘤和伯克特淋巴瘤;(c)选自以下的骨髓系的造血肿瘤:急性和慢性骨髓性白血病、骨髓增生异常综合征和早幼粒细胞白血病;(d)选自纤维肉瘤和横纹肌肉瘤的间质来源的肿瘤;(e)选自星形细胞瘤、神经母细胞瘤、神经胶质瘤和神经鞘瘤的中枢和周围神经系统的肿瘤;或(f)黑色素瘤、精原细胞瘤、畸胎癌、骨肉瘤、着色性干皮病、角化棘皮瘤、甲状腺滤泡癌或卡波济氏肉瘤。
- 权利要求22的用途,其中所述癌症选自子宫肉瘤、卵巢癌、乳腺癌、胃癌、食管癌、肺癌、肝癌、子宫内膜癌、血液肿瘤、神经胶质瘤、肝癌、子宫癌、结直肠癌、食道癌、食管胃结合部腺癌、膀胱癌、头颈癌、宫颈癌和黑素瘤。
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202310335595.5 | 2023-03-31 | ||
CN202310335595 | 2023-03-31 | ||
CN202311719463 | 2023-12-14 | ||
CN202311719463.9 | 2023-12-14 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024199524A1 true WO2024199524A1 (zh) | 2024-10-03 |
Family
ID=92903411
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2024/085203 WO2024199524A1 (zh) | 2023-03-31 | 2024-04-01 | 作为pkmyt1抑制剂的化合物 |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024199524A1 (zh) |
Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
DD149666A1 (de) * | 1979-06-27 | 1981-07-22 | Karl Gewald | Verfahren zur herstellung von 6-amino-und 6-hydroxy-1-aryl-5-cyan-2(1h)-pyridinonen und-thionen |
CN103432133A (zh) * | 2007-01-22 | 2013-12-11 | Gtx公司 | 核受体结合剂的用途 |
CN104163799A (zh) * | 2014-07-01 | 2014-11-26 | 苏州大学 | 3-氨基-4-氰基异喹啉-1(2h)-酮衍生物的制备方法 |
CN106083718A (zh) * | 2016-06-14 | 2016-11-09 | 苏州大学 | 一种异喹啉‑1,3,4‑三酮衍生物及其制备方法 |
CN106366085A (zh) * | 2015-07-25 | 2017-02-01 | 复旦大学 | 异喹啉酮类化合物或其盐及其制备方法和用途 |
WO2023022659A1 (en) * | 2021-08-19 | 2023-02-23 | Engine Biosciences Pte. Ltd. | Compositions and methods for generating synthetic lethality in tumors |
-
2024
- 2024-04-01 WO PCT/CN2024/085203 patent/WO2024199524A1/zh unknown
Patent Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
DD149666A1 (de) * | 1979-06-27 | 1981-07-22 | Karl Gewald | Verfahren zur herstellung von 6-amino-und 6-hydroxy-1-aryl-5-cyan-2(1h)-pyridinonen und-thionen |
CN103432133A (zh) * | 2007-01-22 | 2013-12-11 | Gtx公司 | 核受体结合剂的用途 |
CN104163799A (zh) * | 2014-07-01 | 2014-11-26 | 苏州大学 | 3-氨基-4-氰基异喹啉-1(2h)-酮衍生物的制备方法 |
CN106366085A (zh) * | 2015-07-25 | 2017-02-01 | 复旦大学 | 异喹啉酮类化合物或其盐及其制备方法和用途 |
CN106083718A (zh) * | 2016-06-14 | 2016-11-09 | 苏州大学 | 一种异喹啉‑1,3,4‑三酮衍生物及其制备方法 |
WO2023022659A1 (en) * | 2021-08-19 | 2023-02-23 | Engine Biosciences Pte. Ltd. | Compositions and methods for generating synthetic lethality in tumors |
Non-Patent Citations (4)
Title |
---|
AL-SAID MANSOUR SULAIMAN, EL-GAZZAR MARWA GALAL, GHORAB MOSTAFA MOHAMMED: "In-vitro cytotoxic and radiosensitizing evaluation of novel 2-pyridone, isoquinoline, chromene and chromenopyridone derivatives", EUROPEAN JOURNAL OF CHEMISTRY, vol. 3, no. 2, 30 June 2012 (2012-06-30), pages 228 - 234, XP093219435, ISSN: 2153-2249, DOI: 10.5155/eurjchem.3.2.228-234.596 * |
DATABASE REGISTRY 21 September 2006 (2006-09-21), ANONYMOUS: "4H-Pyrazolo[4,3-c]pyridin-4-on e, 7-acetyl-3-amino-5-(4-chl orophenyl)-1,5- dihydro- (9CI) (CA INDEX NAME)", XP093219508, Database accession no. 908069-29-4 * |
FENG XIAN, WANG JIAN-JUN, XUN ZHAN, HUANG ZHI-BIN, SHI DA-QING: "Multicomponent Strategy to Indeno[2,1- c ]pyridine and Hydroisoquinoline Derivatives through Cleavage of Carbon–Carbon Bond", THE JOURNAL OF ORGANIC CHEMISTRY, AMERICAN CHEMICAL SOCIETY, UNITED STATES, vol. 80, no. 2, 16 January 2015 (2015-01-16), United States, pages 1025 - 1033, XP093219437, ISSN: 0022-3263, DOI: 10.1021/jo5025199 * |
ZHAO, HONGTAO ET AL.: "Discovery of a Novel Chemotype of Tyrosine Kinase Inhibitors by Fragment-Based Docking and Molecular Dynamics", ACS MEDICINAL CHEMISTRY LETTERS, vol. 3, 23 August 2012 (2012-08-23), pages 834 - 838, XP055130698, DOI: 10.1021/ml3001984 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN109475531B (zh) | Ptpn11的杂环抑制剂 | |
JP7299837B2 (ja) | 化合物、組成物、および使用方法 | |
CN109970743B (zh) | 为jak抑制剂的5-氯-2-二氟甲氧基苯基吡唑并嘧啶化合物 | |
WO2021169990A1 (zh) | 用于癌症治疗的kras抑制剂 | |
TW202115076A (zh) | 嘧啶并五員氮雜環類衍生物、其製備方法及其在醫藥上的應用 | |
WO2019174533A1 (zh) | Pd-1/pd-l1类小分子抑制剂及其在药物中的应用 | |
CN112312904A (zh) | 螺环化合物 | |
CN105566321B (zh) | 杂芳化合物及其在药物中的应用 | |
JP2019524872A (ja) | 癌の治療に有用なイミダゾピリミジン化合物 | |
WO2022121914A1 (zh) | 氧代氮环类衍生物调节剂、其制备方法和应用 | |
CN107921044B (zh) | 用于治疗疾病的gls1抑制剂 | |
CN111560012B (zh) | 一种作为irak抑制剂的化合物 | |
JP2021519308A (ja) | 転写活性化タンパク質のイミダゾピペラジン阻害剤 | |
WO2016197078A1 (en) | Compounds for the modulation of myc activity | |
WO2020156479A1 (zh) | 环丙烯并苯并呋喃取代的氮杂芳基化合物、其中间体、制备方法及应用 | |
WO2022272106A1 (en) | Cdk2 inhibitors and methods of using the same | |
WO2022089389A1 (zh) | 杂环化合物及其制备方法、药物组合物和应用 | |
US11858938B2 (en) | Imidazo-fused heterocycles and uses thereof | |
CN103313599A (zh) | 取代的4-(芳基氨基)硒吩并嘧啶化合物及其使用方法 | |
KR20230035036A (ko) | 이중 키나제-브로모도메인 억제제 | |
WO2021032004A9 (zh) | 氮杂芳基化合物及其应用 | |
CN108948019B (zh) | 黏着斑激酶抑制剂及其用途 | |
CA3180058A1 (en) | Pyrazolo[1,5-a]pyrimidine derivatives having multimodal activity against pain | |
CN109790160A (zh) | 吡啶并五元芳香环类化合物、其制备方法及用途 | |
WO2024199524A1 (zh) | 作为pkmyt1抑制剂的化合物 |