CN113372267B - C-Met kinase inhibitor - Google Patents
C-Met kinase inhibitor Download PDFInfo
- Publication number
- CN113372267B CN113372267B CN202010160921.XA CN202010160921A CN113372267B CN 113372267 B CN113372267 B CN 113372267B CN 202010160921 A CN202010160921 A CN 202010160921A CN 113372267 B CN113372267 B CN 113372267B
- Authority
- CN
- China
- Prior art keywords
- compound
- formula
- acid
- pharmaceutically acceptable
- alkyl
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Active
Links
- 229940125895 MET kinase inhibitor Drugs 0.000 title description 4
- 150000001875 compounds Chemical class 0.000 claims abstract description 156
- 238000002360 preparation method Methods 0.000 claims abstract description 35
- -1 sulfonylamidine compound Chemical class 0.000 claims abstract description 21
- 150000003839 salts Chemical class 0.000 claims abstract description 14
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 5
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 claims description 45
- 125000000217 alkyl group Chemical group 0.000 claims description 23
- 229910052736 halogen Inorganic materials 0.000 claims description 18
- 150000002367 halogens Chemical class 0.000 claims description 18
- 125000003545 alkoxy group Chemical group 0.000 claims description 16
- 125000001424 substituent group Chemical group 0.000 claims description 15
- JUJWROOIHBZHMG-UHFFFAOYSA-N Pyridine Chemical compound C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 claims description 12
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 claims description 12
- 239000000460 chlorine Substances 0.000 claims description 12
- 238000000034 method Methods 0.000 claims description 12
- 229910052757 nitrogen Inorganic materials 0.000 claims description 11
- 239000003814 drug Substances 0.000 claims description 10
- 125000004076 pyridyl group Chemical group 0.000 claims description 9
- VHYFNPMBLIVWCW-UHFFFAOYSA-N 4-Dimethylaminopyridine Chemical compound CN(C)C1=CC=NC=C1 VHYFNPMBLIVWCW-UHFFFAOYSA-N 0.000 claims description 8
- 125000000449 nitro group Chemical group [O-][N+](*)=O 0.000 claims description 8
- 150000007530 organic bases Chemical class 0.000 claims description 7
- 239000003960 organic solvent Substances 0.000 claims description 7
- 125000002943 quinolinyl group Chemical group N1=C(C=CC2=CC=CC=C12)* 0.000 claims description 7
- 238000003756 stirring Methods 0.000 claims description 7
- 125000005913 (C3-C6) cycloalkyl group Chemical group 0.000 claims description 6
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 claims description 6
- XKRFYHLGVUSROY-UHFFFAOYSA-N Argon Chemical compound [Ar] XKRFYHLGVUSROY-UHFFFAOYSA-N 0.000 claims description 6
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 claims description 6
- 206010009944 Colon cancer Diseases 0.000 claims description 6
- XEEYBQQBJWHFJM-UHFFFAOYSA-N Iron Chemical group [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 claims description 6
- 208000005718 Stomach Neoplasms Diseases 0.000 claims description 6
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 claims description 6
- 239000003054 catalyst Substances 0.000 claims description 6
- 206010017758 gastric cancer Diseases 0.000 claims description 6
- 208000002154 non-small cell lung carcinoma Diseases 0.000 claims description 6
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 claims description 6
- 230000008569 process Effects 0.000 claims description 6
- UMJSCPRVCHMLSP-UHFFFAOYSA-N pyridine Natural products COC1=CC=CN=C1 UMJSCPRVCHMLSP-UHFFFAOYSA-N 0.000 claims description 6
- 201000011549 stomach cancer Diseases 0.000 claims description 6
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 claims description 6
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 claims description 5
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-dimethylformamide Substances CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 claims description 5
- 239000002253 acid Substances 0.000 claims description 5
- 239000003638 chemical reducing agent Substances 0.000 claims description 5
- 208000029742 colonic neoplasm Diseases 0.000 claims description 5
- 229910052731 fluorine Inorganic materials 0.000 claims description 5
- 229910052739 hydrogen Inorganic materials 0.000 claims description 5
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 5
- 239000011261 inert gas Substances 0.000 claims description 5
- ITQTTZVARXURQS-UHFFFAOYSA-N 3-methylpyridine Chemical compound CC1=CC=CN=C1 ITQTTZVARXURQS-UHFFFAOYSA-N 0.000 claims description 4
- 229960000549 4-dimethylaminophenol Drugs 0.000 claims description 4
- LCTONWCANYUPML-UHFFFAOYSA-N Pyruvic acid Chemical compound CC(=O)C(O)=O LCTONWCANYUPML-UHFFFAOYSA-N 0.000 claims description 4
- 230000000259 anti-tumor effect Effects 0.000 claims description 4
- 229910052794 bromium Inorganic materials 0.000 claims description 4
- 229910052801 chlorine Inorganic materials 0.000 claims description 4
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 claims description 4
- 239000011737 fluorine Substances 0.000 claims description 4
- 239000001257 hydrogen Substances 0.000 claims description 4
- 239000004615 ingredient Substances 0.000 claims description 4
- JVTAAEKCZFNVCJ-UHFFFAOYSA-N lactic acid Chemical compound CC(O)C(O)=O JVTAAEKCZFNVCJ-UHFFFAOYSA-N 0.000 claims description 4
- 239000008194 pharmaceutical composition Substances 0.000 claims description 4
- WKBOTKDWSSQWDR-UHFFFAOYSA-N Bromine atom Chemical compound [Br] WKBOTKDWSSQWDR-UHFFFAOYSA-N 0.000 claims description 3
- ZAMOUSCENKQFHK-UHFFFAOYSA-N Chlorine atom Chemical compound [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 claims description 3
- PXGOKWXKJXAPGV-UHFFFAOYSA-N Fluorine Chemical compound FF PXGOKWXKJXAPGV-UHFFFAOYSA-N 0.000 claims description 3
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 claims description 3
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 claims description 3
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 claims description 3
- FEWJPZIEWOKRBE-UHFFFAOYSA-N Tartaric acid Natural products [H+].[H+].[O-]C(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-N 0.000 claims description 3
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical group [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 claims description 3
- 229910052786 argon Inorganic materials 0.000 claims description 3
- GDTBXPJZTBHREO-UHFFFAOYSA-N bromine Substances BrBr GDTBXPJZTBHREO-UHFFFAOYSA-N 0.000 claims description 3
- JOXIMZWYDAKGHI-UHFFFAOYSA-N toluene-4-sulfonic acid Chemical compound CC1=CC=C(S(O)(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-N 0.000 claims description 3
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 claims description 3
- BMYNFMYTOJXKLE-UHFFFAOYSA-N 3-azaniumyl-2-hydroxypropanoate Chemical compound NCC(O)C(O)=O BMYNFMYTOJXKLE-UHFFFAOYSA-N 0.000 claims description 2
- YKGBNAGNNUEZQC-UHFFFAOYSA-N 6-methyl-n,n-bis(6-methylheptyl)heptan-1-amine Chemical compound CC(C)CCCCCN(CCCCCC(C)C)CCCCCC(C)C YKGBNAGNNUEZQC-UHFFFAOYSA-N 0.000 claims description 2
- 229910021589 Copper(I) bromide Inorganic materials 0.000 claims description 2
- 229910021591 Copper(I) chloride Inorganic materials 0.000 claims description 2
- OFOBLEOULBTSOW-UHFFFAOYSA-N Propanedioic acid Natural products OC(=O)CC(O)=O OFOBLEOULBTSOW-UHFFFAOYSA-N 0.000 claims description 2
- KDYFGRWQOYBRFD-UHFFFAOYSA-N Succinic acid Natural products OC(=O)CCC(O)=O KDYFGRWQOYBRFD-UHFFFAOYSA-N 0.000 claims description 2
- GSEJCLTVZPLZKY-UHFFFAOYSA-N Triethanolamine Chemical compound OCCN(CCO)CCO GSEJCLTVZPLZKY-UHFFFAOYSA-N 0.000 claims description 2
- SRSXLGNVWSONIS-UHFFFAOYSA-N benzenesulfonic acid Chemical compound OS(=O)(=O)C1=CC=CC=C1 SRSXLGNVWSONIS-UHFFFAOYSA-N 0.000 claims description 2
- KDYFGRWQOYBRFD-NUQCWPJISA-N butanedioic acid Chemical compound O[14C](=O)CC[14C](O)=O KDYFGRWQOYBRFD-NUQCWPJISA-N 0.000 claims description 2
- OXBLHERUFWYNTN-UHFFFAOYSA-M copper(I) chloride Chemical compound [Cu]Cl OXBLHERUFWYNTN-UHFFFAOYSA-M 0.000 claims description 2
- VZCYOOQTPOCHFL-UPHRSURJSA-N maleic acid Chemical compound OC(=O)\C=C/C(O)=O VZCYOOQTPOCHFL-UPHRSURJSA-N 0.000 claims description 2
- 239000011976 maleic acid Substances 0.000 claims description 2
- 238000004519 manufacturing process Methods 0.000 claims description 2
- VZCYOOQTPOCHFL-OWOJBTEDSA-N Fumaric acid Chemical compound OC(=O)\C=C\C(O)=O VZCYOOQTPOCHFL-OWOJBTEDSA-N 0.000 claims 2
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 claims 2
- YGSDEFSMJLZEOE-UHFFFAOYSA-N salicylic acid Chemical compound OC(=O)C1=CC=CC=C1O YGSDEFSMJLZEOE-UHFFFAOYSA-N 0.000 claims 2
- QBYIENPQHBMVBV-HFEGYEGKSA-N (2R)-2-hydroxy-2-phenylacetic acid Chemical compound O[C@@H](C(O)=O)c1ccccc1.O[C@@H](C(O)=O)c1ccccc1 QBYIENPQHBMVBV-HFEGYEGKSA-N 0.000 claims 1
- WLJVXDMOQOGPHL-PPJXEINESA-N 2-phenylacetic acid Chemical compound O[14C](=O)CC1=CC=CC=C1 WLJVXDMOQOGPHL-PPJXEINESA-N 0.000 claims 1
- BSYNRYMUTXBXSQ-UHFFFAOYSA-N Aspirin Chemical compound CC(=O)OC1=CC=CC=C1C(O)=O BSYNRYMUTXBXSQ-UHFFFAOYSA-N 0.000 claims 1
- FEWJPZIEWOKRBE-JCYAYHJZSA-N Dextrotartaric acid Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-JCYAYHJZSA-N 0.000 claims 1
- IWYDHOAUDWTVEP-UHFFFAOYSA-N R-2-phenyl-2-hydroxyacetic acid Natural products OC(=O)C(O)C1=CC=CC=C1 IWYDHOAUDWTVEP-UHFFFAOYSA-N 0.000 claims 1
- 229960001138 acetylsalicylic acid Drugs 0.000 claims 1
- 229910000147 aluminium phosphate Inorganic materials 0.000 claims 1
- 229940092714 benzenesulfonic acid Drugs 0.000 claims 1
- 235000015165 citric acid Nutrition 0.000 claims 1
- 239000001530 fumaric acid Substances 0.000 claims 1
- 239000004310 lactic acid Substances 0.000 claims 1
- 235000014655 lactic acid Nutrition 0.000 claims 1
- 229960002510 mandelic acid Drugs 0.000 claims 1
- 229940098779 methanesulfonic acid Drugs 0.000 claims 1
- LNOPIUAQISRISI-UHFFFAOYSA-N n'-hydroxy-2-propan-2-ylsulfonylethanimidamide Chemical compound CC(C)S(=O)(=O)CC(N)=NO LNOPIUAQISRISI-UHFFFAOYSA-N 0.000 claims 1
- FJKROLUGYXJWQN-UHFFFAOYSA-N papa-hydroxy-benzoic acid Natural products OC(=O)C1=CC=C(O)C=C1 FJKROLUGYXJWQN-UHFFFAOYSA-N 0.000 claims 1
- 229960004838 phosphoric acid Drugs 0.000 claims 1
- 229940107700 pyruvic acid Drugs 0.000 claims 1
- 229960004889 salicylic acid Drugs 0.000 claims 1
- 229940032330 sulfuric acid Drugs 0.000 claims 1
- 239000011975 tartaric acid Substances 0.000 claims 1
- 235000002906 tartaric acid Nutrition 0.000 claims 1
- 210000004027 cell Anatomy 0.000 abstract description 35
- 210000004881 tumor cell Anatomy 0.000 abstract description 12
- 230000000694 effects Effects 0.000 abstract description 10
- 230000005764 inhibitory process Effects 0.000 abstract description 10
- 239000000651 prodrug Substances 0.000 abstract description 5
- 229940002612 prodrug Drugs 0.000 abstract description 5
- 230000035755 proliferation Effects 0.000 abstract description 5
- 239000012453 solvate Substances 0.000 abstract description 5
- 239000002246 antineoplastic agent Substances 0.000 abstract description 4
- 229940041181 antineoplastic drug Drugs 0.000 abstract description 4
- 201000011510 cancer Diseases 0.000 abstract description 4
- 239000013078 crystal Substances 0.000 abstract description 4
- 150000004677 hydrates Chemical class 0.000 abstract 1
- 230000008685 targeting Effects 0.000 abstract 1
- UHOVQNZJYSORNB-UHFFFAOYSA-N Benzene Chemical compound C1=CC=CC=C1 UHOVQNZJYSORNB-UHFFFAOYSA-N 0.000 description 53
- HEDRZPFGACZZDS-MICDWDOJSA-N Trichloro(2H)methane Chemical compound [2H]C(Cl)(Cl)Cl HEDRZPFGACZZDS-MICDWDOJSA-N 0.000 description 48
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical group CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 33
- 239000011734 sodium Substances 0.000 description 30
- 239000007787 solid Substances 0.000 description 24
- 238000006243 chemical reaction Methods 0.000 description 23
- 238000004128 high performance liquid chromatography Methods 0.000 description 23
- 239000000243 solution Substances 0.000 description 18
- 239000000203 mixture Substances 0.000 description 17
- 229920006395 saturated elastomer Polymers 0.000 description 16
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 15
- 235000002639 sodium chloride Nutrition 0.000 description 13
- 238000001644 13C nuclear magnetic resonance spectroscopy Methods 0.000 description 12
- 238000005160 1H NMR spectroscopy Methods 0.000 description 12
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 12
- 238000002844 melting Methods 0.000 description 12
- 230000008018 melting Effects 0.000 description 12
- 238000001308 synthesis method Methods 0.000 description 12
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 12
- 238000010992 reflux Methods 0.000 description 11
- 125000003118 aryl group Chemical group 0.000 description 10
- 235000019441 ethanol Nutrition 0.000 description 10
- 229960004756 ethanol Drugs 0.000 description 10
- 239000012074 organic phase Substances 0.000 description 10
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 9
- 125000001188 haloalkyl group Chemical group 0.000 description 9
- HQNSWBRZIOYGAW-UHFFFAOYSA-N 2-chloro-n,n-dimethylpyridin-4-amine Chemical compound CN(C)C1=CC=NC(Cl)=C1 HQNSWBRZIOYGAW-UHFFFAOYSA-N 0.000 description 8
- QOGPNCUTXVZQSL-UHFFFAOYSA-N 6,7-dimethoxy-1h-quinolin-4-one Chemical compound C1=CC(O)=C2C=C(OC)C(OC)=CC2=N1 QOGPNCUTXVZQSL-UHFFFAOYSA-N 0.000 description 8
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 8
- 238000001514 detection method Methods 0.000 description 8
- 125000005842 heteroatom Chemical group 0.000 description 7
- 125000000592 heterocycloalkyl group Chemical group 0.000 description 7
- 229940079593 drug Drugs 0.000 description 6
- 125000005843 halogen group Chemical group 0.000 description 6
- 239000002609 medium Substances 0.000 description 6
- 239000003921 oil Substances 0.000 description 6
- 229910052760 oxygen Inorganic materials 0.000 description 6
- 229910052717 sulfur Inorganic materials 0.000 description 6
- 102000003745 Hepatocyte Growth Factor Human genes 0.000 description 5
- 108090000100 Hepatocyte Growth Factor Proteins 0.000 description 5
- 206010023774 Large cell lung cancer Diseases 0.000 description 5
- 239000012320 chlorinating reagent Substances 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 201000009546 lung large cell carcinoma Diseases 0.000 description 5
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 5
- 125000002950 monocyclic group Chemical group 0.000 description 5
- 231100000252 nontoxic Toxicity 0.000 description 5
- 238000012360 testing method Methods 0.000 description 5
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 4
- XTHFKEDIFFGKHM-UHFFFAOYSA-N Dimethoxyethane Chemical compound COCCOC XTHFKEDIFFGKHM-UHFFFAOYSA-N 0.000 description 4
- URLKBWYHVLBVBO-UHFFFAOYSA-N Para-Xylene Chemical group CC1=CC=C(C)C=C1 URLKBWYHVLBVBO-UHFFFAOYSA-N 0.000 description 4
- 239000008346 aqueous phase Substances 0.000 description 4
- 125000002618 bicyclic heterocycle group Chemical group 0.000 description 4
- MVPPADPHJFYWMZ-UHFFFAOYSA-N chlorobenzene Chemical compound ClC1=CC=CC=C1 MVPPADPHJFYWMZ-UHFFFAOYSA-N 0.000 description 4
- 238000004440 column chromatography Methods 0.000 description 4
- 238000000338 in vitro Methods 0.000 description 4
- 239000003112 inhibitor Substances 0.000 description 4
- 239000012046 mixed solvent Substances 0.000 description 4
- 230000003000 nontoxic effect Effects 0.000 description 4
- XHXFXVLFKHQFAL-UHFFFAOYSA-N phosphoryl trichloride Chemical compound ClP(Cl)(Cl)=O XHXFXVLFKHQFAL-UHFFFAOYSA-N 0.000 description 4
- 239000011780 sodium chloride Substances 0.000 description 4
- ORPHLVJBJOCHBR-UHFFFAOYSA-N 403-19-0 Chemical compound OC1=CC=C([N+]([O-])=O)C=C1F ORPHLVJBJOCHBR-UHFFFAOYSA-N 0.000 description 3
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 3
- CXQHYVUVSFXTMY-UHFFFAOYSA-N N1'-[3-fluoro-4-[[6-methoxy-7-[3-(4-morpholinyl)propoxy]-4-quinolinyl]oxy]phenyl]-N1-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide Chemical compound C1=CN=C2C=C(OCCCN3CCOCC3)C(OC)=CC2=C1OC(C(=C1)F)=CC=C1NC(=O)C1(C(=O)NC=2C=CC(F)=CC=2)CC1 CXQHYVUVSFXTMY-UHFFFAOYSA-N 0.000 description 3
- 239000004698 Polyethylene Substances 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 230000008901 benefit Effects 0.000 description 3
- 229910052799 carbon Inorganic materials 0.000 description 3
- 238000001816 cooling Methods 0.000 description 3
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 3
- 239000002552 dosage form Substances 0.000 description 3
- 239000000839 emulsion Substances 0.000 description 3
- NPTDXPDGUHAFKC-UHFFFAOYSA-N ethynylcyclopropane Chemical group C#CC1CC1 NPTDXPDGUHAFKC-UHFFFAOYSA-N 0.000 description 3
- 239000012065 filter cake Substances 0.000 description 3
- 239000000706 filtrate Substances 0.000 description 3
- 229950008692 foretinib Drugs 0.000 description 3
- 238000010438 heat treatment Methods 0.000 description 3
- 125000001072 heteroaryl group Chemical group 0.000 description 3
- 239000005457 ice water Substances 0.000 description 3
- 229940043355 kinase inhibitor Drugs 0.000 description 3
- 239000000546 pharmaceutical excipient Substances 0.000 description 3
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 3
- 125000004193 piperazinyl group Chemical group 0.000 description 3
- 125000003386 piperidinyl group Chemical group 0.000 description 3
- 239000000843 powder Substances 0.000 description 3
- 239000011541 reaction mixture Substances 0.000 description 3
- 230000019491 signal transduction Effects 0.000 description 3
- LMBFAGIMSUYTBN-MPZNNTNKSA-N teixobactin Chemical compound C([C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CO)C(=O)N[C@H](CCC(N)=O)C(=O)N[C@H]([C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CO)C(=O)N[C@H]1C(N[C@@H](C)C(=O)N[C@@H](C[C@@H]2NC(=N)NC2)C(=O)N[C@H](C(=O)O[C@H]1C)[C@@H](C)CC)=O)NC)C1=CC=CC=C1 LMBFAGIMSUYTBN-MPZNNTNKSA-N 0.000 description 3
- 125000001544 thienyl group Chemical group 0.000 description 3
- 229920000858 Cyclodextrin Polymers 0.000 description 2
- 206010059866 Drug resistance Diseases 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 239000007864 aqueous solution Substances 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- DFEXVBOMMIJOAW-UHFFFAOYSA-N carbamimidoylsulfonylmethanimidamide Chemical group NC(=N)S(=O)(=O)C(N)=N DFEXVBOMMIJOAW-UHFFFAOYSA-N 0.000 description 2
- 125000004432 carbon atom Chemical group C* 0.000 description 2
- 239000001569 carbon dioxide Substances 0.000 description 2
- 229910002092 carbon dioxide Inorganic materials 0.000 description 2
- 230000004663 cell proliferation Effects 0.000 description 2
- 239000007810 chemical reaction solvent Substances 0.000 description 2
- 125000000753 cycloalkyl group Chemical group 0.000 description 2
- 238000013461 design Methods 0.000 description 2
- GLUUGHFHXGJENI-UHFFFAOYSA-N diethylenediamine Natural products C1CNCCN1 GLUUGHFHXGJENI-UHFFFAOYSA-N 0.000 description 2
- USIUVYZYUHIAEV-UHFFFAOYSA-N diphenyl ether Chemical compound C=1C=CC=CC=1OC1=CC=CC=C1 USIUVYZYUHIAEV-UHFFFAOYSA-N 0.000 description 2
- 201000010099 disease Diseases 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 2
- 239000012091 fetal bovine serum Substances 0.000 description 2
- 239000012051 hydrophobic carrier Substances 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 229910052740 iodine Inorganic materials 0.000 description 2
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 2
- 150000002632 lipids Chemical class 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 239000008297 liquid dosage form Substances 0.000 description 2
- 125000002757 morpholinyl group Chemical group 0.000 description 2
- 239000000825 pharmaceutical preparation Substances 0.000 description 2
- RLOWWWKZYUNIDI-UHFFFAOYSA-N phosphinic chloride Chemical group ClP=O RLOWWWKZYUNIDI-UHFFFAOYSA-N 0.000 description 2
- 239000006187 pill Substances 0.000 description 2
- 125000003367 polycyclic group Chemical group 0.000 description 2
- 239000013641 positive control Substances 0.000 description 2
- 239000000047 product Substances 0.000 description 2
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 description 2
- 108090000623 proteins and genes Proteins 0.000 description 2
- 230000035484 reaction time Effects 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 239000002904 solvent Substances 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 2
- NQRYJNQNLNOLGT-UHFFFAOYSA-N tetrahydropyridine hydrochloride Natural products C1CCNCC1 NQRYJNQNLNOLGT-UHFFFAOYSA-N 0.000 description 2
- HKDZJADXHIXZLF-MXHKJMPZSA-N (1R,3S,5S,6R,8S,10S,11R,13S,15S,16R,18S,20S,21R,23S,25S,26R,28S,30S,31R,33S,35S,36S,37S,38S,39S,40S,41S,42S,43S,44S,45S,46S,47S,48S,49S)-5,15,25,35-tetrakis(hydroxymethyl)-10,20,30-tris(2-hydroxypropoxymethyl)-2,4,7,9,12,14,17,19,22,24,27,29,32,34-tetradecaoxaoctacyclo[31.2.2.23,6.28,11.213,16.218,21.223,26.228,31]nonatetracontane-36,37,38,39,40,41,42,43,44,45,46,47,48,49-tetradecol Chemical compound OC[C@@H]([C@@H]([C@H]([C@@H]1O)O)O[C@@H]2O[C@H]([C@H](O[C@@H]3O[C@@H](CO)[C@@H]([C@H]([C@@H]3O)O)O[C@@H]3O[C@@H](COCC(C)O)[C@@H]([C@H]([C@@H]3O)O)O[C@@H]3O[C@@H](CO)[C@@H]([C@H]([C@@H]3O)O)O[C@@H]3O[C@@H](COCC(C)O)[C@@H]([C@H]([C@@H]3O)O)O3)[C@@H](O)[C@@H]2O)COCC(O)C)O[C@H]1O[C@@H]1[C@@H](O)[C@H](O)[C@H]3O[C@H]1CO HKDZJADXHIXZLF-MXHKJMPZSA-N 0.000 description 1
- 125000006273 (C1-C3) alkyl group Chemical group 0.000 description 1
- 125000004169 (C1-C6) alkyl group Chemical group 0.000 description 1
- DQXKOHDUMJLXKH-PHEQNACWSA-N (e)-n-[2-[2-[[(e)-oct-2-enoyl]amino]ethyldisulfanyl]ethyl]oct-2-enamide Chemical compound CCCCC\C=C\C(=O)NCCSSCCNC(=O)\C=C\CCCCC DQXKOHDUMJLXKH-PHEQNACWSA-N 0.000 description 1
- 125000000954 2-hydroxyethyl group Chemical group [H]C([*])([H])C([H])([H])O[H] 0.000 description 1
- PXRKCOCTEMYUEG-UHFFFAOYSA-N 5-aminoisoindole-1,3-dione Chemical compound NC1=CC=C2C(=O)NC(=O)C2=C1 PXRKCOCTEMYUEG-UHFFFAOYSA-N 0.000 description 1
- BSYNRYMUTXBXSQ-FOQJRBATSA-N 59096-14-9 Chemical compound CC(=O)OC1=CC=CC=C1[14C](O)=O BSYNRYMUTXBXSQ-FOQJRBATSA-N 0.000 description 1
- 206010000830 Acute leukaemia Diseases 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- 239000005995 Aluminium silicate Substances 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 208000001333 Colorectal Neoplasms Diseases 0.000 description 1
- 229910021595 Copper(I) iodide Inorganic materials 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 229920002785 Croscarmellose sodium Polymers 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- GSNUFIFRDBKVIE-UHFFFAOYSA-N DMF Natural products CC1=CC=C(C)O1 GSNUFIFRDBKVIE-UHFFFAOYSA-N 0.000 description 1
- 235000019739 Dicalciumphosphate Nutrition 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 208000000461 Esophageal Neoplasms Diseases 0.000 description 1
- CTKXFMQHOOWWEB-UHFFFAOYSA-N Ethylene oxide/propylene oxide copolymer Chemical compound CCCOC(C)COCCO CTKXFMQHOOWWEB-UHFFFAOYSA-N 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 206010064571 Gene mutation Diseases 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 102100022623 Hepatocyte growth factor receptor Human genes 0.000 description 1
- 101000972946 Homo sapiens Hepatocyte growth factor receptor Proteins 0.000 description 1
- 238000012404 In vitro experiment Methods 0.000 description 1
- 208000008839 Kidney Neoplasms Diseases 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 231100000002 MTT assay Toxicity 0.000 description 1
- 238000000134 MTT assay Methods 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 206010027406 Mesothelioma Diseases 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 1
- 201000003793 Myelodysplastic syndrome Diseases 0.000 description 1
- 206010067482 No adverse event Diseases 0.000 description 1
- 206010030155 Oesophageal carcinoma Diseases 0.000 description 1
- 206010033128 Ovarian cancer Diseases 0.000 description 1
- 206010061535 Ovarian neoplasm Diseases 0.000 description 1
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 1
- 108010019160 Pancreatin Proteins 0.000 description 1
- YGYAWVDWMABLBF-UHFFFAOYSA-N Phosgene Chemical compound ClC(Cl)=O YGYAWVDWMABLBF-UHFFFAOYSA-N 0.000 description 1
- RVGRUAULSDPKGF-UHFFFAOYSA-N Poloxamer Chemical compound C1CO1.CC1CO1 RVGRUAULSDPKGF-UHFFFAOYSA-N 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 1
- 102000001253 Protein Kinase Human genes 0.000 description 1
- 108700020978 Proto-Oncogene Proteins 0.000 description 1
- 102000052575 Proto-Oncogene Human genes 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- 208000006265 Renal cell carcinoma Diseases 0.000 description 1
- 208000000453 Skin Neoplasms Diseases 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 208000002463 Sveinsson chorioretinal atrophy Diseases 0.000 description 1
- YTPLMLYBLZKORZ-UHFFFAOYSA-N Thiophene Chemical compound C=1C=CSC=1 YTPLMLYBLZKORZ-UHFFFAOYSA-N 0.000 description 1
- 208000024770 Thyroid neoplasm Diseases 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 239000000783 alginic acid Substances 0.000 description 1
- 235000010443 alginic acid Nutrition 0.000 description 1
- 229960001126 alginic acid Drugs 0.000 description 1
- 229920000615 alginic acid Polymers 0.000 description 1
- 150000004781 alginic acids Chemical class 0.000 description 1
- 125000001931 aliphatic group Chemical group 0.000 description 1
- 239000003513 alkali Substances 0.000 description 1
- 235000012211 aluminium silicate Nutrition 0.000 description 1
- 230000003698 anagen phase Effects 0.000 description 1
- 230000006909 anti-apoptosis Effects 0.000 description 1
- 230000001028 anti-proliverative effect Effects 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 239000002585 base Substances 0.000 description 1
- 125000003785 benzimidazolyl group Chemical group N1=C(NC2=C1C=CC=C2)* 0.000 description 1
- 125000004604 benzisothiazolyl group Chemical group S1N=C(C2=C1C=CC=C2)* 0.000 description 1
- 125000004603 benzisoxazolyl group Chemical group O1N=C(C2=C1C=CC=C2)* 0.000 description 1
- 125000005605 benzo group Chemical group 0.000 description 1
- 125000000499 benzofuranyl group Chemical group O1C(=CC2=C1C=CC=C2)* 0.000 description 1
- 125000001164 benzothiazolyl group Chemical group S1C(=NC2=C1C=CC=C2)* 0.000 description 1
- 125000004196 benzothienyl group Chemical group S1C(=CC2=C1C=CC=C2)* 0.000 description 1
- 125000004541 benzoxazolyl group Chemical group O1C(=NC2=C1C=CC=C2)* 0.000 description 1
- 229920000249 biocompatible polymer Polymers 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 150000001717 carbocyclic compounds Chemical class 0.000 description 1
- 150000001721 carbon Chemical group 0.000 description 1
- MOIPGXQKZSZOQX-UHFFFAOYSA-N carbonyl bromide Chemical compound BrC(Br)=O MOIPGXQKZSZOQX-UHFFFAOYSA-N 0.000 description 1
- RVIQSSNDHKQZHH-UHFFFAOYSA-N carbonyl diiodide Chemical compound IC(I)=O RVIQSSNDHKQZHH-UHFFFAOYSA-N 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 239000004359 castor oil Substances 0.000 description 1
- 235000019438 castor oil Nutrition 0.000 description 1
- 230000012292 cell migration Effects 0.000 description 1
- 230000009087 cell motility Effects 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 125000001309 chloro group Chemical group Cl* 0.000 description 1
- 239000000084 colloidal system Substances 0.000 description 1
- NKNDPYCGAZPOFS-UHFFFAOYSA-M copper(i) bromide Chemical compound Br[Cu] NKNDPYCGAZPOFS-UHFFFAOYSA-M 0.000 description 1
- LSXDOTMGLUJQCM-UHFFFAOYSA-M copper(i) iodide Chemical compound I[Cu] LSXDOTMGLUJQCM-UHFFFAOYSA-M 0.000 description 1
- 239000008120 corn starch Substances 0.000 description 1
- 229940099112 cornstarch Drugs 0.000 description 1
- 229960001681 croscarmellose sodium Drugs 0.000 description 1
- 235000010947 crosslinked sodium carboxy methyl cellulose Nutrition 0.000 description 1
- 238000005138 cryopreservation Methods 0.000 description 1
- 238000012258 culturing Methods 0.000 description 1
- 229940045803 cuprous chloride Drugs 0.000 description 1
- WZHCOOQXZCIUNC-UHFFFAOYSA-N cyclandelate Chemical compound C1C(C)(C)CC(C)CC1OC(=O)C(O)C1=CC=CC=C1 WZHCOOQXZCIUNC-UHFFFAOYSA-N 0.000 description 1
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 229940097362 cyclodextrins Drugs 0.000 description 1
- 125000000113 cyclohexyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 125000001559 cyclopropyl group Chemical group [H]C1([H])C([H])([H])C1([H])* 0.000 description 1
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 230000003013 cytotoxicity Effects 0.000 description 1
- 238000002784 cytotoxicity assay Methods 0.000 description 1
- 231100000263 cytotoxicity test Toxicity 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- NEFBYIFKOOEVPA-UHFFFAOYSA-K dicalcium phosphate Chemical compound [Ca+2].[Ca+2].[O-]P([O-])([O-])=O NEFBYIFKOOEVPA-UHFFFAOYSA-K 0.000 description 1
- 229940038472 dicalcium phosphate Drugs 0.000 description 1
- 229910000390 dicalcium phosphate Inorganic materials 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 239000003596 drug target Substances 0.000 description 1
- 238000001035 drying Methods 0.000 description 1
- 230000009982 effect on human Effects 0.000 description 1
- 201000004101 esophageal cancer Diseases 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 125000001153 fluoro group Chemical group F* 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 125000000524 functional group Chemical group 0.000 description 1
- 125000002541 furyl group Chemical group 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 229940014259 gelatin Drugs 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 208000005017 glioblastoma Diseases 0.000 description 1
- 229960005150 glycerol Drugs 0.000 description 1
- 235000011187 glycerol Nutrition 0.000 description 1
- ZEMPKEQAKRGZGQ-XOQCFJPHSA-N glycerol triricinoleate Natural products CCCCCC[C@@H](O)CC=CCCCCCCCC(=O)OC[C@@H](COC(=O)CCCCCCCC=CC[C@@H](O)CCCCCC)OC(=O)CCCCCCCC=CC[C@H](O)CCCCCC ZEMPKEQAKRGZGQ-XOQCFJPHSA-N 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 125000004438 haloalkoxy group Chemical group 0.000 description 1
- 150000002430 hydrocarbons Chemical group 0.000 description 1
- 239000012052 hydrophilic carrier Substances 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- 125000002883 imidazolyl group Chemical group 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 125000001041 indolyl group Chemical group 0.000 description 1
- 239000012442 inert solvent Substances 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 150000007529 inorganic bases Chemical class 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000007917 intracranial administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000007914 intraventricular administration Methods 0.000 description 1
- 230000009545 invasion Effects 0.000 description 1
- 125000001786 isothiazolyl group Chemical group 0.000 description 1
- 125000000842 isoxazolyl group Chemical group 0.000 description 1
- NLYAJNPCOHFWQQ-UHFFFAOYSA-N kaolin Chemical compound O.O.O=[Al]O[Si](=O)O[Si](=O)O[Al]=O NLYAJNPCOHFWQQ-UHFFFAOYSA-N 0.000 description 1
- 229960000829 kaolin Drugs 0.000 description 1
- 201000010982 kidney cancer Diseases 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 229960001375 lactose Drugs 0.000 description 1
- 150000002611 lead compounds Chemical class 0.000 description 1
- 229940040145 liniment Drugs 0.000 description 1
- 239000000865 liniment Substances 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 201000007270 liver cancer Diseases 0.000 description 1
- 208000014018 liver neoplasm Diseases 0.000 description 1
- 239000008176 lyophilized powder Substances 0.000 description 1
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 229960001855 mannitol Drugs 0.000 description 1
- 210000002901 mesenchymal stem cell Anatomy 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 239000008108 microcrystalline cellulose Substances 0.000 description 1
- 229940016286 microcrystalline cellulose Drugs 0.000 description 1
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 1
- 239000011859 microparticle Substances 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 125000002911 monocyclic heterocycle group Chemical group 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
- 125000001624 naphthyl group Chemical group 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
- 230000007935 neutral effect Effects 0.000 description 1
- 125000006574 non-aromatic ring group Chemical group 0.000 description 1
- 125000002971 oxazolyl group Chemical group 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 208000008443 pancreatic carcinoma Diseases 0.000 description 1
- 229940055695 pancreatin Drugs 0.000 description 1
- 201000010279 papillary renal cell carcinoma Diseases 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 229940127557 pharmaceutical product Drugs 0.000 description 1
- WLJVXDMOQOGPHL-UHFFFAOYSA-N phenylacetic acid Chemical compound OC(=O)CC1=CC=CC=C1 WLJVXDMOQOGPHL-UHFFFAOYSA-N 0.000 description 1
- 150000003904 phospholipids Chemical class 0.000 description 1
- UHZYTMXLRWXGPK-UHFFFAOYSA-N phosphorus pentachloride Chemical compound ClP(Cl)(Cl)(Cl)Cl UHZYTMXLRWXGPK-UHFFFAOYSA-N 0.000 description 1
- FAIAAWCVCHQXDN-UHFFFAOYSA-N phosphorus trichloride Chemical compound ClP(Cl)Cl FAIAAWCVCHQXDN-UHFFFAOYSA-N 0.000 description 1
- 229920001983 poloxamer Polymers 0.000 description 1
- 229920001993 poloxamer 188 Polymers 0.000 description 1
- 229940044519 poloxamer 188 Drugs 0.000 description 1
- 229920001992 poloxamer 407 Polymers 0.000 description 1
- 229940044476 poloxamer 407 Drugs 0.000 description 1
- 229940068886 polyethylene glycol 300 Drugs 0.000 description 1
- 229940068918 polyethylene glycol 400 Drugs 0.000 description 1
- 239000000244 polyoxyethylene sorbitan monooleate Substances 0.000 description 1
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 1
- 229940068968 polysorbate 80 Drugs 0.000 description 1
- 229920000053 polysorbate 80 Polymers 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 229960004063 propylene glycol Drugs 0.000 description 1
- 235000013772 propylene glycol Nutrition 0.000 description 1
- 108060006633 protein kinase Proteins 0.000 description 1
- 102000004169 proteins and genes Human genes 0.000 description 1
- 239000003586 protic polar solvent Substances 0.000 description 1
- 125000003373 pyrazinyl group Chemical group 0.000 description 1
- 125000003226 pyrazolyl group Chemical group 0.000 description 1
- 125000002098 pyridazinyl group Chemical group 0.000 description 1
- 125000000714 pyrimidinyl group Chemical group 0.000 description 1
- 125000000168 pyrrolyl group Chemical group 0.000 description 1
- 125000001567 quinoxalinyl group Chemical group N1=C(C=NC2=CC=CC=C12)* 0.000 description 1
- 150000003254 radicals Chemical class 0.000 description 1
- 239000002994 raw material Substances 0.000 description 1
- 108091008598 receptor tyrosine kinases Proteins 0.000 description 1
- 102000027426 receptor tyrosine kinases Human genes 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 238000013341 scale-up Methods 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 201000000849 skin cancer Diseases 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 229960002668 sodium chloride Drugs 0.000 description 1
- 239000008109 sodium starch glycolate Substances 0.000 description 1
- 229940079832 sodium starch glycolate Drugs 0.000 description 1
- 229920003109 sodium starch glycolate Polymers 0.000 description 1
- 239000007909 solid dosage form Substances 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 238000005556 structure-activity relationship Methods 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 229960004793 sucrose Drugs 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- 125000001712 tetrahydronaphthyl group Chemical group C1(CCCC2=CC=CC=C12)* 0.000 description 1
- 125000003554 tetrahydropyrrolyl group Chemical group 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 125000000335 thiazolyl group Chemical group 0.000 description 1
- 201000002510 thyroid cancer Diseases 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000820 toxicity test Toxicity 0.000 description 1
- 230000005747 tumor angiogenesis Effects 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 239000012224 working solution Substances 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D215/00—Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
- C07D215/02—Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
- C07D215/16—Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
- C07D215/20—Oxygen atoms
- C07D215/22—Oxygen atoms attached in position 2 or 4
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D215/00—Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
- C07D215/02—Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
- C07D215/16—Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
- C07D215/20—Oxygen atoms
- C07D215/22—Oxygen atoms attached in position 2 or 4
- C07D215/233—Oxygen atoms attached in position 2 or 4 only one oxygen atom which is attached in position 4
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
- C07D401/12—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Hematology (AREA)
- Oncology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The invention provides a sulfonylamidine compound with a novel structure as well as a preparation method and application thereof. The sulfonylamidine compound is a compound shown in a formula (I), or polymorphs, prodrugs, solvates, hydrates, co-crystals, pharmaceutically acceptable salts and the like thereof. The sulfonylamidine compound provided by the invention has a good proliferation inhibition effect on various cancer cells, has a low tumor cell inhibition concentration, remarkably improves the activity of the compound, has a good selectivity on tumor cells, and is expected to be a specific antitumor drug targeting c-Met kinase.
Description
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to a c-Met kinase inhibitor.
Background
The c-Met or HGFR is a protein product encoded by the c-Met protooncogene, is a receptor tyrosine kinase with high affinity, and is the only known high affinity receptor for Hepatocyte Growth Factor (HGF). Activation of the HGF/c-Met signaling pathway plays a very important role in cell proliferation, motility, migration and survival. However, in many common malignant tumors, abnormally high expression of c-Met and HGF results in abnormal activation of HGF/c-Met signaling pathway, which is in close and indiscriminate association with proliferation, metastasis, invasion, anti-apoptosis and tumor angiogenesis of tumor cells. Therefore, c-Met kinase and related signal pathways have become an important target for anti-tumor drug research. Of the many c-Met kinase inhibitors, the most prominent is the research directed to small molecule inhibitors. The structure-activity relationship shows that the inhibitor mainly consists of three parts, namely a framework which is easy to form hydrogen bonds with c-Met kinase, an aryl fragment which can be extended into a hydrophobic capsule and an intermediate bridging chain part, and the influence of the intermediate bridging chain part on the activity of the inhibitor is particularly remarkable. However, with the use of c-Met kinase inhibitors in large quantities, secondary gene mutations or subcloning of tumor cells in the body result in the generation and expansion of drug-resistant tumor cells containing the new mutated genes, ultimately leading to the generation of drug resistance of this class of drugs. Thus, there is an urgent need to study and develop c-Met kinase inhibitors having novel structures in order to find therapeutic methods for solving drug resistance.
The sulfonylamidine fragment is an important pharmacophore, can form hydrogen bond interaction with a drug target, and is widely applied to the design of anticancer drugs. Based on the application of the segment in the design of antitumor drugs, the study tries to take the sulfonylamidine segment as the intermediate bridge chain part of the c-Met inhibitor so as to obtain the lead compound with better antitumor activity.
The following compounds are part of the reported compounds:
Disclosure of Invention
In view of the deficiencies of the prior art, one aspect of the present application is to provide a c-Met kinase inhibitor.
Specifically, the invention provides a compound represented by the general formula I or a polymorph, prodrug, solvate, hydrate, co-crystal or pharmaceutically acceptable salt thereof:
wherein,
R 1 is C 1-6 alkyl, C 3-6 cycloalkyl, C 3-6 heterocycloalkyl, aryl, azaaryl, which are optionally substituted with: alkoxy, halogen, alkyl and haloalkyl, wherein the hetero element in the heterocycloalkyl of the C 3-6 is O, N, S;
R 2 is aryl, azaaryl, cycloalkyl of C 3-6, heterocycloalkyl of C 3-6, said aryl and azaaryl optionally being substituted with: alkoxy, halogen, alkyl, haloalkyl and nitro, wherein the hetero element in the heterocycloalkyl of the C 3-6 is O, N, S;
R 3 is aryl, heteroaryl, het selected from the group consisting of piperidinyl, pyrrolyl, pyrazolyl, piperidinyl, imidazolyl, furanyl, morpholinyl, thienyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridinyl, pyrimidinyl, piperazinyl, substituted piperazinyl, pyrazinyl, and pyridazinyl; or a bicyclic heterocycle selected from quinolinyl, quinoxalinyl, indolyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzothiazolyl, benzisothiazolyl, benzofuranyl, thiophen [3,2-B ] pyridinyl, 7H-pyrrolo [2,3-d ] pyrimidinyl, benzothienyl, 2, 3-dihydrobenzo [ B ] [1,4] dioxanyl, or benzo [ d ] [1,3] dioxolyl; each of said monocyclic or bicyclic heterocycles being optionally substituted with 1,2 or 3 substituents, each substituent being independently selected from halogen, haloalkyl, alkyl or alkoxy, or from an aliphatic carbocycle of C 3-C8, or from an aliphatic heterocycle of the following formula: tetrahydropyrrolyl, morpholinyl, alkoxymorpholinyl, piperazinyl, piperidinyl, alkylamino piperidinyl; piperidine substituted alkyl, piperazine substituted alkyl; each monocyclic or bicyclic heterocycle is optionally substituted by 1,2 or 3 substituents, and the hetero element in the heteroaryl, the monocyclic heterocycle, the aliphatic heterocycle and the bicyclic heterocycle is O, N, S;
x is hydrogen or halogen, and halogen is selected from one of fluorine, chlorine and bromine.
Preferably, R 1 is C 1-6 alkyl, C 3-6 cycloalkyl, C 3-6 heterocycloalkyl, aryl, azaaryl, which may be optionally substituted with: c 1-3 alkoxy, halogen, C 1-3 alkyl and haloalkyl, wherein the hetero element in the heterocycloalkyl of the C 3-6 is O, N, S;
R 2 is aryl, azaaryl, which may be optionally substituted with: alkoxy, halogen, alkyl, haloalkyl, nitro;
R 3 is quinolinyl, thienyl [3,2-B ] pyridinyl, 7H-pyrrolo [2,3-d ] pyrimidinyl, quinolinyl, thienyl [3,2-B ] pyridinyl, and 7H-pyrrolo [2,3-d ] pyrimidinyl optionally substituted with 1,2, or 3 substituents, each substituent independently selected from halogen, haloalkyl, alkyl, or alkoxy; piperidine substituted alkyl, piperazine substituted alkyl;
x is halogen, and halogen is selected from one of fluorine, chlorine and bromine.
Further preferably, R 1 is C 1-6 alkyl, C 3-6 cycloalkyl, phenyl, pyridinyl, which are optionally substituted with 0, 1, 2 groups: c 1-3 alkoxy, halogen, C 1-3 alkyl, haloalkyl;
R 2 is phenyl, pyridinyl, which are optionally substituted with 0,1 or 2 groups: alkoxy of C 1-6, halogen, alkyl of C 1-6, haloalkyl, nitro;
R 3 is quinolinyl, quinolinyl optionally substituted with 1,2, or 3 substituents, each substituent independently selected from halogen, haloalkyl, C 1-5 alkyl, or C 1-5 alkoxy; 2- (1-piperidine) -ethyl, 2- (1-piperazine) -ethyl;
x is fluorine.
More preferably, the compound is selected from the following compounds:
in a second aspect the present invention provides a process for the preparation of a compound of formula I, the process comprising the step of reacting a compound of formula IV, a compound of formula III and a compound of formula II to obtain a compound of formula I:
Wherein X, R 1、R2 and R 3 are as defined above.
Preferably, the synthesis method specifically comprises the following steps: the compound of the formula IV is dissolved in an organic solvent, organic alkali is added, stirring and inert gas protection are carried out, and the compound of the formula III, the compound of the formula II and a catalyst are added for reaction to obtain the compound of the formula I.
Further preferably, the organic base is selected from one of triethylamine, pyridine, triethanolamine, 3-methylpyridine, DMAP and triisooctylamine; the organic solvent is selected from one or more of dichloromethane, 1, 4-dioxane, meCN, THF, DMF and toluene; the inert gas is argon or nitrogen; the catalyst is CuX, and X is selected from one of Cl, I and Br.
The method further comprises the step of reducing the nitro group of the compound of formula V to provide the compound of formula IV:
Wherein R 3 is as defined above.
Preferably, the reducing agent used for reducing the compound of formula V is selected from iron powder or zinc powder.
The process further comprises the step of reacting a compound of formula VII with a compound of formula VI to obtain a compound of formula V:
wherein X, R 3 is as defined above.
Preferably, the molar ratio of the compound of formula VII to the compound of formula VI in the present reaction is from 1:1.0 to 1.5.
The process further comprises reacting the compound of formula VIII with a chlorinating agent to obtain a compound of formula VI:
R3-OH
VIII,
Wherein R 3 is as defined above.
The following further details the preparation of the compounds of formula I of the present invention:
At room temperature, dissolving the compound of the formula IV in an organic solvent, adding an organic base, stirring, protecting by inert gas, adding the compound of the formula III, the compound of the formula II and a catalyst to react at room temperature, adding dichloromethane and saturated NH 4 Cl aqueous solution into the reaction solution, continuously stirring, separating liquid, extracting the aqueous phase with dichloromethane, merging the organic phases, drying, filtering, concentrating under reduced pressure, and performing column chromatography to obtain the compound of the formula I.
Preferably, the catalyst is selected from one of cuprous iodide, cuprous bromide and cuprous chloride.
Preferably, the organic base is selected from one of triethylamine, DMAP and pyridine.
Preferably, the molar ratio of the compound of formula IV to the organic base is 1:2 to 3; the molar ratio of the compound of the formula IV to the compound of the formula III is 1:1.0-1.1; the molar ratio of the compound of the formula IV to the compound of the formula II is 1:1.1-1.3.
In one embodiment, the molar ratio of the compound of formula IV to the organic base is 1:2.4; the molar ratio of the compound of formula IV to the compound of formula III is 1:1; the molar ratio of the compound of formula IV to the compound of formula II is 1:1.2.
The preparation of the compound of formula IV comprises the steps of: and (3) dissolving the compound shown in the formula V in a proton solvent, heating, adding a saturated NH 4 Cl solution and a reducing agent, and heating and refluxing for reaction.
Preferably, the protic solvent is selected from ethanol, water or a mixed solvent of both; the reducing agent is selected from iron powder or zinc powder.
In one embodiment, the volume ratio of ethanol to water is 9:1; the reducing agent is iron powder.
The preparation of the compound of formula V comprises the steps of: and dissolving the compound of the formula VI and the compound of the formula VII in an organic solvent, heating and refluxing, and reacting for 15-32 hours.
Preferably, the organic solvent is selected from one of chlorobenzene, ethylene glycol dimethyl ether, paraxylene, diphenyl ether, 1, 4-dioxane, pyridine/1, 4-dioxane mixed solvent, NMP/1, 4-dioxane mixed solvent, DMF/pyridine mixed solvent.
Preferably, the molar ratio of the compound of formula VII to the compound of formula VI is from 1:1.0 to 1.5; the reaction time is 28 to 32 hours.
In one embodiment, the molar ratio of the compound of formula VII to the compound of formula VI is 1:1.2; the reaction time was 30 hours.
The preparation of the compound of formula VI comprises the steps of: the compound of formula VIII and the chloro reagent are dissolved in the reaction solvent and heated to reflux for reaction.
Preferably, the reaction solvent is POCl 3 or acetonitrile.
Preferably, the molar ratio of the compound of formula VIII to the chlorinating agent is from 1:15 to 28.
Further preferably, the chlorinating agent is selected from one of phosphorus oxychloride, phosphorus trichloride and phosphorus pentachloride.
In one embodiment, the molar ratio of the compound of formula VIII to the chlorinating agent is 1:21.5; the chlorinating agent is phosphorus oxychloride.
In a preferred embodiment, the process for the preparation of the compounds of formula I comprises the steps of: the hydroxyl on the compound of the formula VIII is chlorinated and then reacts with the compound of the formula VII to obtain the compound of the formula V; reducing the nitro group on the compound of formula V to provide a compound of formula IV; finally, the compound of formula IV, the compound of formula III and the compound of formula II react to obtain the compound of formula I.
The synthetic route is as follows:
the compound of the invention can be prepared by the preparation method or the preparation method similar to the preparation method, and corresponding raw materials are selected according to different substituents and different substituent positions.
The synthetic route of the compound I-1 in the invention is as follows:
In a third aspect the present invention provides a pharmaceutical composition comprising a compound of formula I or a polymorph, prodrug, solvate, hydrate, co-crystal, pharmaceutically acceptable salt and other pharmaceutically acceptable ingredients thereof.
The compositions of the present invention refer to pharmaceutical products comprising a therapeutically effective amount of the specified ingredients, as well as any products that result directly or indirectly from the combination of the specified ingredients in the specified amounts.
The compositions of the present invention are particularly pharmaceutical compositions which are generally safe, non-toxic and biologically desirable, and therefore, the pharmaceutically acceptable carriers or excipients of the present invention are non-toxic and safe, and their combination with the compounds of the present invention are also non-toxic and safe. Pharmaceutically acceptable carriers and excipients as described herein are generally well known to those skilled in the art or can be determined by those skilled in the art depending on the actual situation. Examples of suitable carriers and excipients include dextrose, water, glycerol, ethanol, propylene glycol, corn starch, gelatin, lactose, sucrose, alginic acid, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride, croscarmellose sodium, sodium starch glycolate, polysorbate 80, polyethylene glycol 300, polyethylene glycol 400, cyclodextrins or derivatives thereof, such as ((2-hydroxypropyl) -cyclodextrin) and (2-hydroxyethyl) -cyclodextrin, also known as HPCD, pegylated castor oil, poloxamers (such as poloxamer 407 or 188); hydrophilic carriers, hydrophobic carriers, combinations thereof, or the like. Hydrophobic carriers include, for example, fat emulsions, lipids, pegylated phospholipids, biocompatible polymers, lipid globules, liposomes, vesicles, polymeric matrices, particles, and the like.
The carrier may be present in the pharmaceutical composition in an amount of 1% to 98% by weight, typically about 80% by weight.
The composition of the present invention may be administered in any manner selected from the group consisting of: oral, spray inhalation, rectal, nasal, vaginal, topical, parenteral, such as subcutaneous, intravenous, intramuscular, intraperitoneal, intrathecal, intraventricular, intrasternal or intracranial injection or infusion, or by means of an explanted reservoir, with oral, intramuscular, intraperitoneal or intravenous modes of administration being preferred.
The compounds of the present invention or compositions or pharmaceutical preparations containing the same may be administered in unit dosage form. The administration dosage form may be liquid dosage form or solid dosage form. The liquid dosage form can be true solution, colloid, microparticle, emulsion, and mixed rotation. Other dosage forms such as tablet, capsule, dripping pill, aerosol, pill, powder, solution, suspension, emulsion, granule, suppository, lyophilized powder for injection, clathrate, landfill, patch, liniment, etc.
In a fourth aspect, the invention provides the use of a compound of formula I or a polymorph, prodrug, solvate, hydrate, co-crystal or pharmaceutically acceptable salt thereof in the preparation of an anti-tumour medicament.
Preferably, the tumor is selected from: non-small cell lung cancer, liver cancer, papillary renal cell carcinoma, gastric cancer, esophageal cancer, glioblastoma, head and neck squamous cell, renal cancer, acute leukemia, prostate cancer, thyroid cancer, skin cancer, colorectal cancer, pancreatic cancer, ovarian cancer, breast cancer, myelodysplastic syndrome, mesothelioma, or the like.
Interpretation of the terms
The following sets forth definitions of various terms used to describe the application. These definitions apply to the terms used throughout the specification and claims unless otherwise limited in specific instances, either alone or as part of a larger group.
The term "alkyl" in the present invention refers to saturated straight or branched chain hydrocarbon groups, in certain embodiments containing 1 to 6 or 1 to 3 carbon atoms, respectively, examples of C 1-6 alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, neopentyl, n-hexyl, or the like, and examples of C 1-3 alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, or the like.
The term "halo" as used herein refers to a group formed by substitution of a halogen atom for a hydrogen atom on a carbon atom, wherein the halogen atom includes, but is not limited to F, cl, br, I.
The term "alkoxy" in the present invention refers to-O-alkyl, wherein the alkyl includes, but is not limited to, C 1-3 alkyl, C 1-6 alkyl, and C 3-6 cycloalkyl, and specific examples include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy, and halogenated versions thereof.
The term "aryl" in the present invention refers to a mono-or polycyclic carbocyclic ring system having one or more aromatic rings, either fused or unfused, including but not limited to phenyl, naphthyl, tetrahydronaphthyl, and the like, and which ring carbon hydrogen atoms may also be substituted with one or more substituents including but not limited to alkyl, alkoxy, halo, haloalkoxy.
The term "heterocycloalkyl" according to the present invention refers to a mono-or polycyclic non-aromatic ring system containing 2 to 6 ring carbon atoms and 1 to 3 ring heteroatoms, wherein the heteroatoms are selected from N, O, S.
The term "cycloalkyl" as used herein refers to a monovalent radical of a monocyclic or polycyclic saturated or partially unsaturated carbocyclic compound, C 3-6 cycloalkyl including, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like, and the hydrogen atoms on its ring carbon may also be substituted with one or more substituents including, but not limited to, alkyl, alkoxy, halo.
The term "heteroaryl" according to the invention refers to an aromatic ring system containing 1 to 6 carbons and at least one heteroatom selected from N, S, O; and the hydrogen atoms on the ring carbons thereof may also be substituted with one or more substituents including, but not limited to, alkyl, alkoxy, halo, nitro.
The term "pharmaceutically acceptable salt" as used herein refers to salts of the compounds of the present invention prepared from the compounds of the present invention having the specified substituents found herein and relatively non-toxic acids or bases. The relatively basic functional groups of the present invention may be present in pure solution or in a suitable inert solvent to obtain acid addition salts, including inorganic and organic acid salts, by contacting the neutral form of such compounds with a sufficient amount of the acid. The inorganic acid salts include, but are not limited to, hydrochloric, hydrobromic, sulfuric, phosphoric, methanesulfonic, benzenesulfonic, p-toluenesulfonic, citric, tartaric, lactic, pyruvic, acetic, maleic or succinic acid, fumaric, salicylic, phenylacetic, mandelic, sulfamic, acetylsalicylic acid, and also include the acid salts of inorganic bases and the like.
The "solvate" as used herein refers to a form containing a stoichiometric or non-stoichiometric addition of a solvent selected from methanol, ethanol, acetone, DMF, DMSO, and the like.
As used herein, "hydrate" refers to a form containing water addition in either a stoichiometric or non-stoichiometric amount.
"Prodrug" as used herein refers to a compound that is metabolically convertible in vivo to provide any compound described by the formulas of the present application, and various forms of drugs are known in the art.
An "effective amount" as used herein refers to a dosage that achieves the desired therapeutic effect in the desired subject without undue adverse effects, as may be generally determined by one of skill in the art as desired.
The term "treatment" as used herein refers to a method of alleviating or moderating a disease or its complications; by prevention is meant reducing or eliminating the onset of symptoms or complications of a disease, condition or disorder.
It should be understood that other terms not explained hereinabove but appearing in the present invention should be defined as commonly understood by one of ordinary skill in the art.
It is to be understood that within the scope of the present invention, the above-described technical features of the present invention and technical features specifically described below (including embodiments) may be combined with each other to constitute new or preferred technical solutions.
Compared with the prior art, the invention has the main advantages that:
(1) Compared with the existing compounds, the sulfonyl amidine compound has lower tumor cell inhibition concentration and obviously enhanced anti-tumor activity.
(2) Compared with the known similar compounds, the sulfonyl amidine compound of the formula I provided by the invention has the advantages of shorter synthetic route, simpler operation and suitability for industrial scale-up production.
Detailed Description
The advantages of the invention will now be further described by the following examples, which are given for illustrative purposes only and do not limit the scope of the invention, while variations and modifications apparent to those skilled in the art in light of the present disclosure are included within the scope of the invention.
The synthetic route of the compound of the formula I-1 is as follows:
example 1: preparation of Compounds of formula VI
4-Hydroxy-6, 7-dimethoxyquinoline (VIII) (4.10 g,20.0 mmol) and POCl 3 (40 mL) were placed in a reaction flask and the flask was placed in an oil bath under reflux for 6h. After completion of the TLC detection reaction, the reaction mixture was cooled to room temperature and concentrated under reduced pressure. To the resulting residue was poured ice water (100 mL) and vigorously stirred, and the pH was adjusted to 8 with concentrated NH 4 OH, controlling the temperature to within 20 ℃. The mixture was extracted twice with dichloromethane (320 mL. Times.2). The combined organic phases were washed successively with saturated aqueous NaCl and water, dried over anhydrous Na 2SO4, filtered and concentrated to give a white solid, the compound of formula VI, yield 68.3% and HPLC purity 98.11%. m.p.134-136 ℃; ESI-HRMS (m/z): 246.0413[ M+Na ] +.
Example 2: preparation of Compounds of formula VI
4-Hydroxy-6, 7-dimethoxyquinoline (VIII) (4.10 g,20.0 mmol) and PCl 3 (40 ml,0.458 mol) were dissolved in dry acetonitrile (40 ml) and the reaction flask was placed in an oil bath and heated under reflux for 5h. After completion of the TLC detection reaction, the reaction mixture was cooled to room temperature and concentrated under reduced pressure. To the resulting residue was poured ice water (100 ml) and vigorously stirred, and the pH was adjusted to 8 with concentrated NH 4 OH at a temperature of within 20 ℃. The mixture was extracted twice with dichloromethane (320 ml. Times.2). The combined organic phases were washed successively with saturated aqueous NaCl and water, dried over anhydrous Na 2SO4, filtered and concentrated to a white solid, yield: 66.2%, HPLC purity: 98.06%. m.p.134-136 ℃; ESI-HRMS (m/z): 246.0415[ M+Na ] +.
Example 3: preparation of Compounds of formula VI
4-Hydroxy-6, 7-dimethoxyquinoline (VIII) (4.10 g,20.0 mmol) and PCl 5 (40 ml,0.307 mol) were dissolved in dry acetonitrile (40 ml) and the reaction flask was placed in an oil bath and heated under reflux for 7h. After completion of the TLC detection reaction, the reaction mixture was cooled to room temperature and concentrated under reduced pressure. To the resulting residue was poured ice water (100 ml) and vigorously stirred, and the pH was adjusted to 8 with concentrated NH 4 OH at a temperature of within 20 ℃. The mixture was extracted twice with dichloromethane (320 ml. Times.2). The combined organic phases were washed successively with saturated aqueous NaCl and water, dried over anhydrous Na 2SO4, filtered and concentrated to a white solid, yield: 65.1%, HPLC purity: 98.97%. m.p.134-136 ℃; ESI-HRMS (m/z): 246.0412[ M+Na ] +.
Example 4: preparation of Compounds of formula V
The compound of formula VI (4.46 g,20.0 mmol) and 2-fluoro-4-nitrophenol (VII) (4.78 g,24.1 mmol) were dissolved in chlorobenzene (40 mL) and the mixture was heated to reflux for 20h. The reaction is stopped when the residual amount of the compound of formula VI is less than 5%. And (5) cooling to room temperature, and distilling under reduced pressure. The residue was dissolved in dichloromethane (80 mL), washed twice with saturated K 2CO3 (20 mL. Times.2) aqueous solution and once with water (20 mL). The organic phase is dried by anhydrous Na 2SO4, filtered, and the solid obtained by concentration is recrystallized by absolute ethyl alcohol to obtain light yellow solid, and the yield is: 80.4% of HPLC purity 98.90%. m.p.161-163 ℃; ESI-HRMS (m/z): 367.0809[ M+Na ] +.
Example 5: preparation of Compounds of formula V
The compound of formula VI (4.46 g,20.0 mmol) and 2-fluoro-4-nitrophenol (compound of formula VII) (3.14 g,20.0 mmol) were dissolved in paraxylene (40 ml) and the mixture was heated to reflux for 32h. The reaction is stopped when the residual amount of the compound of formula VI is less than 5%. And (5) cooling to room temperature, and distilling under reduced pressure. The residue was dissolved in dichloromethane (80 ml), washed twice with saturated K 2CO3 solution (20 ml. Times.2) and once with water (20 ml). The organic phase was dried over anhydrous Na 2SO4, filtered and the solid obtained by concentration was recrystallized from anhydrous ethanol to give a pale yellow solid with a yield of 78.9% and an HPLC purity of 98.09%. m.p.161-163 ℃; ESI-HRMS (m/z): 367.0811[ M+Na ] +.
Example 6: preparation of Compounds of formula V
The compound of formula VI (4.46 g,20.0 mmol) and 2-fluoro-4-nitrophenol (compound of formula VI) (4.71 g,30.0 mmol) were dissolved in ethylene glycol dimethyl ether (40 ml) and the mixture was heated to reflux for 28h. The reaction is stopped when the residual amount of the compound of formula VI is less than 5%. And (5) cooling to room temperature, and distilling under reduced pressure. The residue was dissolved in dichloromethane (80 ml), washed twice with saturated K 2CO3 solution (20 ml. Times.2) and once with water (20 ml). The organic phase was dried over anhydrous Na 2SO4, filtered and the solid obtained by concentration was recrystallized from anhydrous ethanol to give a pale yellow solid with a yield of 77.2% and an HPLC purity of 98.79%. m.p.161-163 ℃; ESI-HRMS (m/z): 367.0808[ M+Na ] +.
Example 7: preparation of Compounds of formula IV
The compound of formula V (6.88 g,20.0 mmol) was dissolved in ethanol/water (90 mL,9:1 v/v) and saturated NH 4 Cl solution (90 mL) and iron powder (11.17 g,0.2 mol) were added to the mixture at reflux for 5h when the oil bath temperature was raised to 78deg.C. The reaction was detected by TLC and immediately filtered, the filter cake was rinsed with ethanol, and the filtrate was concentrated under reduced pressure to give a white solid in 90.6% yield and 99.06% purity by HPLC. m.p.193-195 ℃; ESI-HRMS (m/z): 337.1105[ M+Na ] +.
Example 8: preparation of Compounds of formula IV
The compound of formula V (6.88 g,20.0 mmol) was dissolved in ethanol (90 ml), and when the oil bath temperature was raised to 78 ℃, saturated NH 4 Cl solution (90 ml) and Fe powder (11.17 g,0.2 mol) were added, and the mixture was refluxed for 4h. The reaction is immediately filtered after TLC detection is complete, filter cakes are leached by ethanol, filtrate is decompressed and concentrated to obtain white solid, and the yield is: 88.2%, HPLC purity: 99.05%. m.p.193-195 ℃; ESI-HRMS (m/z): 337.1108[ M+Na ] +.
Example 9: preparation of Compounds of formula IV
The compound of formula V (6.88 g,20.0 mmol) was dissolved in ethanol/water (90 ml,9:1 v/v) and saturated NH 4 Cl solution (90 ml) and Zn powder (13.08 g,0.2 mol) were added to the mixture at reflux for 6h when the oil bath temperature was raised to 78 ℃. The reaction is immediately filtered after TLC detection is complete, filter cakes are leached by ethanol, filtrate is decompressed and concentrated to obtain white solid, and the yield is: 89.6%, HPLC purity: 99.16%. m.p.193-195 ℃; ESI-HRMS (m/z): 337.1109[ M+Na ] +.
Example 10: preparation of Compound I-1
Triethylamine (2.42 g,24.0 mmol) was slowly added dropwise to a dichloromethane solution (400 mL) of the compound of formula IV (3.14 g,10.0 mmol) at room temperature, stirred for 10min, after which cyclopropylacetylene (III) (0.66 g,10.0 mmol), benzenesulfonyl azide (II) (2.20 g,12.0 mmol) and iodoketone (190.45 mg,1.0 mmol) were added to the reaction flask under N 2 protection. The mixture was reacted at room temperature for 2 to 6 hours, after completion of the TLC detection reaction, it was diluted with DCM (45 mL) and saturated aqueous NH 4 Cl (66 mL), stirring was continued for 30min to separate layers, and the aqueous phase was extracted three times with DCM (40 mL. Times.3). The combined organic phases were dried over anhydrous Na 2SO4, filtered, concentrated under reduced pressure, and purified by column chromatography (PE: ea=10:1) to give the desired compound I-1. White solid, yield 82.1%, HPLC purity: 98.87%, melting point 189-191℃,1H NMR(400Hz,CDCl3)δ:9.89(s,1H,-NH-),9.03(d,J=8.1Hz,1H,-N=CH-CH=),7.80-7.59(m,6H,Ph-H,-N=CH-CH=),7.30(s,1H,quinoline-H),7.07(s,1H,quinoline-H),6.55-6.53(m,3H,Ph-H),3.92(s,3H,-OCH3),3.83(s,3H,-OCH3),2.20(d,J=6.3Hz,2H,-CH2-cyclopropare),1.02(m,1H,cyclopropare-H),0.40(m,4H,cyclopropare-H);13C NMR(100MHz,CDCl3)δ:157.7,156.7,151.3,150.9,150.6,146.2,145.2,144.1,136.0,133.7,129.7(2C),125.5(2C),120.0(2C),117.3(2C),114.5,107.9,106.2,100.7,56.1(2C),42.6,6.1(2C),1.2.ESI-HRMS(m/z):558.1577[M+Na]+
Example 11: preparation of Compound I-1
DMAP (2.93 g,24.0 mmol) was slowly added dropwise to a solution of the compound of formula IV (3.14 g,10.0 mmol) in acetonitrile (400 mL) at room temperature, stirred for 10min, after which cyclopropylacetylene (III) (0.66 g,10.0 mmol), benzenesulfonyl azide (II) (2.20 g,12.0 mmol) and bromoketone (143.45 mg,1.0 mmol) were added to the reaction flask under argon. The mixture was reacted at room temperature for 2 to 6 hours, after completion of the TLC detection reaction, it was diluted with DCM (45 mL) and saturated aqueous NH 4 Cl (66 mL), stirring was continued for 30min to separate layers, and the aqueous phase was extracted three times with DCM (40 mL. Times.3). The combined organic phases were dried over anhydrous Na 2SO4, filtered, concentrated under reduced pressure, and purified by column chromatography (PE: ea=10:1) to give the desired compound I-1, yield: 83.25%, HPLC purity: 99.13%.
Example 12: preparation of Compound I-1
Pyridine (1.90 g,24.0 mmol) was slowly added dropwise to a THF solution (400 mL) of the compound of formula IV (3.14 g,10.0 mmol) at room temperature, stirred for 10min, after which cyclopropylacetylene (III) (0.66g, 10.0 mmol), benzenesulfonyl azide (II) (2.20 g,12.0 mmol) and chloroketone (99.0 mg,1.0 mmol) were added to the reaction flask under N 2 protection. The mixture was reacted at room temperature for 2 to 6 hours, after completion of the TLC detection reaction, it was diluted with DCM (45 mL) and saturated aqueous NH 4 Cl (66 mL), stirring was continued for 30min to separate layers, and the aqueous phase was extracted three times with DCM (40 mL. Times.3). The combined organic phases were dried over anhydrous Na 2SO4, filtered, concentrated under reduced pressure, and purified by column chromatography (PE: ea=10:1) to give the desired compound I-1, yield: 82.03%, HPLC purity: 99.00%.
Example 13: preparation of Compound I-2
The synthesis method is the same as that of I-1, white solid, yield 81.6% and HPLC purity: 99.07%, melting point 177-179℃.1H NMR(400Hz,CDCl3)δ:9.87(s,1H,-NH-),9.22(d,J=8.3Hz,1H,-N=CH-CH=),8.11-7.97(m,3H,quinoline-H),7.80(d,J=5.1Hz,1H,quinoline-H),7.48(dd,J=0.6Hz,8.1Hz,2H,Ph-H),7.24(dd,J=1.3Hz,12.0Hz,2H,Ph-H),6.73(t,J=4.5Hz,1H,Ph-H),6.64(t,J=6.0Hz,1H,Ph-H),6.20(d,J=4.3Hz,1H,Ph-H),2.43(s,3H,-Ph-CH3),2.20(s,2H,-CH2-cyclopropare),1.02(m,1H,cyclopropare-H),0.40-0.15(m,4H,cyclopropare-H);13C NMR(100MHz,CDCl3)δ:159.8,158.1,156.7,152.6,148.6,141.9,141.1,139.4,132.6,132.0,131.3,130.0,128.2(2C),125.5(2C),123.7,119.2,118.5,112.9,110.7,105.8,42.6,6.1(2C),1.2.ESI-HRMS(m/z):546.1133[M+Na]+.
Example 14: preparation of Compound I-3
The synthesis method is the same as that of I-1, white solid, yield 80.1% and HPLC purity: 98.76%, melting point 193-195℃.1H NMR(400Hz,CDCl3)δ:10.01(s,1H,-NH-),9.03(d,J=7.0Hz,1H,-N=CH-CH=),7.80-7.59(m,6H,Ph-H,-N=CH-CH=),7.30(s,1H,quinoline-H),7.08(s,1H,quinoline-H),6.55-6.53(m,3H,Ph-H),3.92(s,3H,-OCH3),3.83(s,3H,-OCH3),3.81(s,3H,-OCH3),2.20(d,J=8.1Hz,2H,-CH2-cyclopropare),1.02(m,1H,cyclopropare-H),0.40(m,4H,cyclopropare-H);13C NMR(100MHz,CDCl3)δ:157.7,156.1,151.3,150.7,150.4,146.2,145.2,144.1,136.0,133.7,129.7(2C),125.5(2C),120.0(2C),117.3(2C),114.5,107.9,106.2,100.7,56.1(2C),55.8,42.6,6.1(2C),1.2.ESI-HRMS(m/z):588.1683[M+Na]+.
Example 15: preparation of Compound I-4
The synthesis method is the same as that of I-1. White solid, yield 81.2%, HPLC purity: 98.75%, melting point 179-181℃.1H NMR(400Hz,CDCl3)δ:9.88(s,1H,-NH-),9.03(d,J=8.4Hz,1H,-N=CH-CH=),7.78(m,2H,Ph-H),7.59-7.50(m,2H,Ph-H,-N=CH-CH=),7.31(s,1H,quinoline-H),7.05(s,1H,quinoline-H),6.73(t,J=6.0Hz,1H,Ph-H),6.64(t,J=8.1Hz,1H,Ph-H),6.20(t,J=4.5Hz,1H,Ph-H),3.92(s,3H,-OCH3),3.83(s,3H,-OCH3),2.20(d,J=5.3Hz,2H,-CH2-cyclopropare),1.02(m,1H,cyclopropare-H),0.40-0.15(m,4H,cyclopropare-H);13C NMR(100MHz,CDCl3)δ:167.9,159.9,157.7,156.5,151.3,150.7,150.6,146.2,141.9,139.7,131.3,129.9(2C),123.7,116.5(2C),114.5,112.0,107.9,106.2,105.8,100.7,56.1(2C),42.6,6.1(2C),1.2.ESI-HRMS(m/z):576.1483[M+Na]+.
Example 16: preparation of Compound I-5
The synthesis method is the same as that of I-1. White solid, yield 82.4%, HPLC purity: 98.89%, melting point 148-150℃.1H NMR(400Hz,CDCl3)δ:9.86(s,1H,-NH-),9.04(d,J=8.1Hz,1H,-N=CH-CH=),7.80(d,J=7.0Hz,2H,Ph-H),7.59(d,J=8.6Hz,1H,quinoline-H),7.59(d,J=4.7Hz,1H,quinoline-H),7.32(s,1H,quinoline-H),7.12(t,J=12.1Hz,2H,Ph-H),7.06(s,1H,quinoline-H),6.73(d,J=3.8Hz,1H,Ph-H),6.64(d,J=4.3Hz,1H,Ph-H),6.21(d,J=6.5Hz,1H,Ph-H),3.92(s,3H,-OCH3),3.84(s,3H,-OCH3),2.14(s,2H,-CH2-C(CH3)3),0.94(s,9H,-C(CH3)3);13C NMR(100MHz,CDCl3)δ:159.9,157.7,156.9,152.9,151.3,150.7,150.5,146.2,141.9,136.4,131.3,128.6,129.3(2C),123.7,115.3(2C),114.5,113.2,107.9,106.3,105.8,100.7,56.2(2C),45.1,32.9,29.6(3C).ESI-HRMS(m/z):658.1713[M+Na]+.
Example 17: preparation of Compound I-6
The synthesis method is the same as that of I-1. White solid, yield 82.7%, HPLC purity: 99.20%, melting point 149-151℃.1H NMR(400Hz,CDCl3)δ:9.85(s,1H,-NH-),9.14(d,J=8.2Hz,1H,-N=CH-CH=),8.48(d,J=7.1Hz,2H,Ph-H),8.10(s,1H,quinoline-H),8.06(d,J=6.4Hz,2H,Ph-H),7.70(d,J=8.5Hz,1H,quinoline-H),7.37(s,1H,quinoline-H),6.73(d,J=4.2Hz,1H,Ph-H),6.64(d,J=5.0Hz,1H,Ph-H),6.29(d,J=5.8Hz,1H,Ph-H),2.63(t,J=8.0Hz,4H,CH3-CH2-CH2-),2.14(s,2H,-CH2-(CH3)3),2.14(s,2H,-CH2-C(CH3)3),1.64(m,4H,CH3-CH2-CH2-),0.94(t,15H,-CH2-CH2-CH3,-CH2-(CH3)3);13C NMR(100MHz,CDCl3)δ:159.8,158.1,156.7,152.9,151.1,150.8,145.6,141.9,141.0,137.9,131.3,129.2(2C),127.6,124.9(2C),123.7,119.2,115.7,112.9,108.3,105.8,45.1,36.1,35.7,29.6(3C),26.7,24.4(2C),13.7(2C).ESI-HRMS(m/z):643.2469[M+Na]+.
Example 18: preparation of Compound I-7
The synthesis method is the same as that of I-1. White solid, 86.3% yield, HPLC purity: 98.88%, melting point 198-200℃.1H NMR(400Hz,CDCl3)δ:9.91(s,1H,-NH-),9.01(d,J=8.5Hz,1H,-N=CH-CH=),8.14(d,J=7.0Hz,1H,Pyridine-H),8.35(dd,J=1.3Hz,8.3Hz,1H,Pyridine-H),7.80(d,J=4.7Hz,2H,Pyridine-H,Ph-H),7.75(t,J=6.0Hz,1H,Ph-H),7.68(t,J=4.1Hz,2H,Ph-H),7.67(t,J=5.0Hz,1H,Ph-H),7.68(t,J=3.3Hz,2H,Ph-H),7.59(d,J=6.4Hz,1H,Pyridine-H),7.30(s,1H,quinoline-H),7.24(t,J=5.7Hz,1H,Pyridine-H),7.08(s,1H,quinoline-H),6.73(d,J=12.0Hz,1H,Ph-H),6.64(t,J=4.1Hz,1H,Ph-H),6.30(d,J=8.0Hz,1H,Ph-H),3.92(s,3H,-OCH3),3.92(s,3H,-OCH3),3.83(s,2H,Ph-CH2-),3.58(s,2H,Pyridine-CH2-);13C NMR(100MHz,CDCl3)δ:157.9,157.0,151.5,150.7,150.3,146.0,145.2,143.9,136.3,135.6,134.0,129.7(2C),129.0(2C),125.7,125.5(2C),120.0(2C),117.3(2C),114.5,107.9,106.2,100.7,56.1(2C),39.5.ESI-HRMS(m/z):595.1530[M+Na]+.
Example 19: preparation of Compound I-8
The synthesis method is the same as that of I-1. White solid, yield 84.1%, HPLC purity: 98.96%, melting point 190-192℃.1H NMR(400Hz,CDCl3)δ:9.87(s,1H,-NH-),9.17(d,J=8.3Hz,1H,-N=CH-CH=),8.55(d,J=7.0Hz,2H,Pyridine-H),7.89(d,J=4.4Hz,1H,quinoline-H),7.73(d,J=4.7Hz,2H,Ph-H),7.69(d,J=6.3Hz,1H,quinoline-H),7.63(s,H,quinoline-H),7.56(t,J=5.0Hz,2H,Ph-H),7.45(d,J=8.6Hz,1H,quinoline-H),7.22(dd,J=1.9Hz,8.5Hz,2H,Pyridine-H),6.73(d,J=12.0Hz,1H,Ph-H),6.64(t,J=7.1Hz,1H,Ph-H),6.30(d,J=8.4Hz,1H,Ph-H),4.46(s,2H,-OCH2-CF3),4.36(s,2H,Pyridine-CH2-);13C NMR(100MHz,CDCl3)δ:160.0,158.0,157.5,156.7,150.2,149.8(2C),146.5,143.8,142.2,141.9,139.3,131.3,130.0,129.8(2C),127.7(2C),124.2(2C),123.7,122.8,122.4,118.8,112.9,110.1,105.7,99.2,82.5,39.5.ESI-HRMS(m/z):667.0908[M+Na]+.
Example 20: preparation of Compound I-9
The synthesis method is the same as that of I-1. White solid, yield 78.1%, HPLC purity: 98.37%, melting point 203-205℃.1H NMR(400Hz,CDCl3)δ:9.84(s,1H,-NH-),9.18(d,J=8.5Hz,1H,-N=CH-CH=),8.33(d,J=7.0Hz,1H,quinoline-H),7.77(d,J=6.1Hz,1H,quinoline-H),7.52(t,J=3.9Hz,1H,quinoline-H),7.49(d,J=9.0Hz,1H,quinoline-H),7.46(m,2H,Ph-H),7.24(m,2H,Ph-H),7.14(m,2H,Ph-H),6.73-6.64(m,4H,Ph-H),6.30(t,J=5.6Hz,1H,Ph-H),4.64(s,2H,Cl-CH2-),3.81(s,3H,Ph-OCH3),3.58(s,2H,Ph-CH2-),2.43(s,1H,Ph-CH3);13C NMR(100MHz,CDCl3)δ:159.7,158.6,157.6,156.9,152.2,147.6,141.9,141.1,139.4,139.0,131.3,130.0(2C),128.8,128.4,128.2(2C),127.7,125.5(2C),123.7,120.9,115.8,114.2(2C),112.9,108.5,105.8,55.8,46.2,39.5,21.3.ESI-HRMS(m/z):626.1395[M+Na]+.
Example 21: preparation of Compound I-10
The synthesis method is the same as that of I-1. White solid, yield 78.4%, HPLC purity: 98.73%, melting point 201-203℃.1H NMR(400Hz,CDCl3)δ:9.82(s,1H,-NH-),9.05(d,J=8.1Hz,1H,-N=CH-CH=),7.59(d,J=7.0Hz,1H,quinoline-H),7.46(dd,J=1.7Hz,6.4Hz,2H,Ph-H),7.34(s,1H,quinoline-H),7.24(dd,J=2.9Hz,8.6Hz,2H,Cl-Ph-H),7.14(m,2H,Ph-H),7.09(s,1H,quinoline-H),6.72(dd,J=2.2Hz,7.4Hz,2H,Ph-H),6.55-6.53(m,4H,Ph-H),3.92(s,3H,-OCH3),3.83(s,3H,-OCH3),3.81(s,3H,Ph-OCH3),3.58(s,2H,Ph-CH2-),2.43(s,1H,Ph-CH3);13C NMR(100MHz,CDCl3)δ:157.8,157.1,156.6,150.9,150.7,150.4,147.4,145.7,145.2,136.4(2C),129.8(2C),128.5(2C),127.8(2C),127.6,124.1,120.6(2C),116.9(2C),114.5,114.2(2C),108.3,106.6,100.5,55.9(2C),55.7,39.8.ESI-HRMS(m/z):692.1557[M+Na]+.
Example 22: preparation of Compound I-11
The synthesis method is the same as that of I-1. White solid, yield 78.5%, HPLC purity: 98.54%, melting point 212-213℃.1H NMR(400Hz,CDCl3)δ:9.84(s,1H,-NH-),9.19(d,J=8.4Hz,1H,-N=CH-CH=),8.37(d,J=7.0Hz,1H,quinoline-H),7.78(d,J=5.3Hz,1H,quinoline-H),7.60(t,J=4.2Hz,2H,Ph-H),7.55(dd,J=2.7Hz,6.4Hz,2H,Ph-H),7.52(t,J=5.7Hz,1H,quinoline-H),7.51(d,J=3.9Hz,1H,quinoline-H),6.73(d,J=6.1Hz,1H,Ph-H),6.64(d,J=6.0Hz,1H,Ph-H),2.72(m,2H,-CH2-CH3),2.69(t,J=8.2Hz,2H,Piperidine-CH2-CH2-),2.67(t,J=7.6Hz,2H,Piperidine-CH2-CH2-),2.42(m,4H,Piperidine-H),2.20(d,J=4.8Hz,2H,Cyclohexane-H),1.53-1.37(m,17H,Piperidine-H,Cyclohexane-H),1.18(t,J=4.6Hz,3H,-CH2-CH3);13C NMR(100MHz,CDCl3)δ:159.5,158.8,156.9,152.2,149.3,147.6,141.9,141.3,139.0,131.3,128.8,128.7(2C),128.4,128.2(2C),123.7,120.9,115.8,112.9,108.5,105.8,60.7,57.1(2C),37.9,33.4(2C),28.2,26.4,26.0,25.9(2C),25.5(2C),24.5,14.5.ESI-HRMS(m/z):679.3196[M+Na]+.
Example 23: preparation of Compound I-12
The synthesis method is the same as that of I-1. White solid, yield 82.1%, HPLC purity: 98.85%, melting point 186-188℃.1H NMR(400Hz,CDCl3)δ:9.93(s,1H,-NH-),9.13(d,J=8.3Hz,1H,-N=CH-CH=),8.91(d,J=4.9Hz,1H,Pyridine-H),8.89(t,J=6.0Hz,1H,Pyridine-H),8.43(t,J=7.5Hz,1H,Pyridine-H),8.37(t,J=5.5Hz,1H,quinoline-H),7.78(d,J=7.1Hz,1H,quinoline-H),7.69(m,1H,Pyridine-H),7.52(t,J=8.3Hz,1H,quinoline-H),7.51(s,1H,quinoline-H),6.73(d,J=4.6Hz,1H,Ph-H),6.64(d,J=6.0Hz,1H,Ph-H),6.20(t,J=4.4Hz,1H,Ph-H),2.69-2.65(m,8H,Piperazine-H,-CH2-CH2-),2.34(m,4H,Piperazine-H),2.20(d,J=8.7Hz,2H,-CH2-cyclopropare),1.07(s,1H,-NH-),1.02(m,1H,cyclopropare-H),0.40-0.15(m,4H,cyclopropare-H);13C NMR(100MHz,CDCl3)δ:159.7,158.7,156.7,154.6,152.2,147.8,147.6,141.9,139.0,133.7,133.6,131.3,128.8,128.4,124.8,123.7,120.9,115.8,114.2,108.5,105.8,60.4,57.6(2C),46.2(2C),42.6,33.4,6.3(2C),1.4.ESI-HRMS(m/z):611.2319[M+Na]+.
Biological Activity Studies
1. In vitro tumor cell proliferation inhibition experiment
In order to study the capability of the synthesized target compound to inhibit the proliferation of tumor cells in the experiment, the in vitro cytotoxicity of the compound to four tumor cells, namely human colon cancer cells (HT-29), human non-small cell lung cancer cells (A549), human large cell lung cancer cells (H460) and human gastric cancer cells (MKN-45), is measured, and Foretinib is used as a positive control. The assay used was the standard MTT assay.
The experimental method specifically comprises the following steps:
The cell cryopreservation tube was removed from liquid nitrogen, rapidly thawed at 39 ℃ and transferred to a 15mL centrifuge tube, 10mL of 10% fbs-containing medium was added, centrifuged for 5min (1000 rpm), the medium was removed, 10% fbs-containing medium and diabody-containing medium was added again, and transferred to a flask for culturing. Taking cells in logarithmic growth phase, removing culture solution in a culture flask, washing the cells once by PBS, carrying out pancreatin digestion and centrifugation collection, re-suspending the cells by using a culture medium containing 10% fetal bovine serum, counting and adjusting the cells to a proper concentration (the cell density is 5 multiplied by 104/mL, the cell activity is more than 90%), and adding the cell suspension into a 96-micro-well plate with 100 mu L per well. The target compounds were diluted with DMSO to 20. Mu.L solution, and the test target compounds were diluted with DMSO in a 3-fold gradient. 5. Mu.L of the diluted compound solution was added to 495. Mu.L of a medium containing 10% FBS, respectively, to prepare test compounds. 100. Mu.L of the solution containing the compound to be tested was added to the corresponding wells of a 96-well plate and cultured in a carbon dioxide cell incubator for 72 hours. The medium was removed, and 150. Mu.L of 0.3 mg/mL -1 MTT working solution (0.00265 mg/mL -1 PMS) was added to each well, followed by placing the mixture in a carbon dioxide incubator for 2 hours. The 96-well plate was shaken in an oscillator for 5min, and the absorbance A450 (450 nm) was read with an ELISA reader. All experiments were performed in 3 parallel groups or in triplicate. Finally, the median inhibitory concentration (IC 50) of the test compound is calculated. The test results are the mean ± Standard Deviation (SD) of three experiments.
The results of in vitro toxicity tests of the compounds I-1 to 12 on proliferation inhibition of four cancer cells, namely human colon cancer cells (HT 29), human non-small cell lung cancer cells (A549), human large cell lung cancer cells (H460) and human gastric cancer cells (MKN-45), are shown in Table 1.
TABLE 1
Note that: (1) screening method: MTT method; (2) action time: 72 hours. (3) P < 0.05.
In vitro experiments show that the compound I-1-12 has stronger inhibition activity on four cancer cells, namely human colon cancer cells (HT 29), human non-small cell lung cancer cells (A549), human large cell lung cancer cells (H460), and human gastric cancer cells (MKN-45), and most of the compound activities are equivalent to clinical medicines Foretinib, wherein the inhibition activity of the compound I-8 on the human non-small cell lung cancer cells (A549) and the human large cell lung cancer cells (H460) is obviously better than that of a control medicine, and the inhibition activity of the compound I-11 on the human colon cancer cells (HT 29), the human large cell lung cancer cells (H460) and the human gastric cancer cells (MKN-45) is obviously better than that of the control medicine, so that the compound I-8 has a good application prospect.
2. In vitro cytotoxicity assay on Normal cells
The invention selects the compounds I-8 and I-11 with better antiproliferative activity on tumor cells, tests the inhibition effect on human umbilical mesenchymal stem cells (normal cells), discovers that the cell survival rate of the compounds is 76.2% and 84.5% respectively at the concentration of 100uM, and shows that the compounds have no obvious toxicity on the normal cells and have certain cell selectivity on the tumor cells.
In general, the series of compounds of the invention have stronger proliferation inhibition capability on tumor cells HT29, A549, H460 and MKN-45, and the activity of part of the compounds is better than that of a positive control drug Foretinib, and the compounds have no toxic effect on normal cells, thus the compounds have very good development prospect.
Claims (8)
1. A compound or pharmaceutically acceptable salt represented by formula I:
,
wherein,
R 1 is C 1-6 alkyl, C 3-6 cycloalkyl, phenyl, pyridyl, said phenyl and pyridyl optionally substituted with 0, 1,2 groups: c 1-3 alkoxy, halogen, C 1-3 alkyl;
R 2 is phenyl, pyridinyl, which are optionally substituted with 0,1 or 2 groups: alkoxy of C 1-6, halogen, alkyl of C 1-6, nitro;
R 3 is quinolinyl, quinolinyl optionally substituted with 1,2, or 3 substituents, each substituent independently selected from halogen, C 1-5 alkyl, C 1-5 alkoxy, 2- (1-piperidine) -ethyl, 2- (1-piperazine) -ethyl;
x is hydrogen or halogen, and halogen is selected from one of fluorine, chlorine and bromine.
2. The compound of claim 1, wherein the compound is the following:
。
3. The compound or pharmaceutically acceptable salt of claim 1, wherein the pharmaceutically acceptable salt comprises an acid addition salt of a compound of formula I with: hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, lactic acid, pyruvic acid, acetic acid, maleic acid, succinic acid, fumaric acid, salicylic acid, phenylacetic acid, mandelic acid, sulfamic acid or acetylsalicylic acid.
4. A process for the preparation of a compound as claimed in claim 1, comprising the steps of: dissolving a compound of a formula IV in an organic solvent, adding an organic base, stirring, protecting by inert gas, and adding a compound of a formula III, a compound of a formula II and a catalyst to react to obtain a compound of a formula I;
,
Wherein X, R 1、R2 and R 3 are as defined in claim 1;
the organic base is selected from one of triethylamine, pyridine, triethanolamine, 3-picoline, DMAP and triisooctyl amine; the organic solvent is selected from one or more of dichloromethane, 1, 4-dioxane, meCN, THF, DMF and toluene; the inert gas is argon or nitrogen; the catalyst is one selected from CuCl, cuI and CuBr.
5. The method of claim 4, comprising the step of reducing the nitro group of the compound of formula V to provide the compound of formula IV:
,
Wherein X, R 3 is as defined in claim 1;
the reducing agent used for reducing the compound of the formula V is selected from iron powder or zinc powder.
6. The method of claim 5, comprising the step of reacting a compound of formula VII with a compound of formula VI to provide a compound of formula V:
,
Wherein X, R 3 is as defined in claim 1;
The mol ratio of the compound of the formula VII to the compound of the formula VI is 1:1.0-1.5.
7. A pharmaceutical composition comprising a compound or pharmaceutically acceptable salt of claim 1 and other pharmaceutically acceptable ingredients.
8. Use of a compound or pharmaceutically acceptable salt according to claim 1 for the manufacture of an anti-tumor medicament, said tumor being selected from the group consisting of non-small cell lung cancer, stomach cancer, colon cancer.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202010160921.XA CN113372267B (en) | 2020-03-10 | 2020-03-10 | C-Met kinase inhibitor |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202010160921.XA CN113372267B (en) | 2020-03-10 | 2020-03-10 | C-Met kinase inhibitor |
Publications (2)
Publication Number | Publication Date |
---|---|
CN113372267A CN113372267A (en) | 2021-09-10 |
CN113372267B true CN113372267B (en) | 2024-05-24 |
Family
ID=77568582
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202010160921.XA Active CN113372267B (en) | 2020-03-10 | 2020-03-10 | C-Met kinase inhibitor |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN113372267B (en) |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN116354823A (en) * | 2021-12-27 | 2023-06-30 | 武汉珈海智药科技有限公司 | Preparation method and application of a class of compounds |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN102212062A (en) * | 2010-04-02 | 2011-10-12 | 东莞市长安东阳光新药研发有限公司 | Derivative of amino ester, salt thereof and using method |
CN106349158A (en) * | 2016-08-03 | 2017-01-25 | 杭州市西溪医院 | c-Met small-molecule inhibitor, pharmaceutical composition containing same and pharmaceutical application of pharmaceutical composition containing same |
CN106543145A (en) * | 2016-10-28 | 2017-03-29 | 山西医科大学 | C Met kinases suppression 5 benzoic acid amides derivant of agent 3 (4 fluorophenyl) pyrimidone, preparation method and application |
CN106831707A (en) * | 2016-12-28 | 2017-06-13 | 杭州市西溪医院 | As the benzheterocycle analog derivative and its medical application of c Met kinase inhibitors |
Family Cites Families (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN106467541B (en) * | 2015-08-18 | 2019-04-05 | 暨南大学 | Substituted quinolone analog derivative or its pharmaceutically acceptable salt or stereoisomer and its Pharmaceutical composition and application |
-
2020
- 2020-03-10 CN CN202010160921.XA patent/CN113372267B/en active Active
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN102212062A (en) * | 2010-04-02 | 2011-10-12 | 东莞市长安东阳光新药研发有限公司 | Derivative of amino ester, salt thereof and using method |
CN106349158A (en) * | 2016-08-03 | 2017-01-25 | 杭州市西溪医院 | c-Met small-molecule inhibitor, pharmaceutical composition containing same and pharmaceutical application of pharmaceutical composition containing same |
CN106543145A (en) * | 2016-10-28 | 2017-03-29 | 山西医科大学 | C Met kinases suppression 5 benzoic acid amides derivant of agent 3 (4 fluorophenyl) pyrimidone, preparation method and application |
CN106831707A (en) * | 2016-12-28 | 2017-06-13 | 杭州市西溪医院 | As the benzheterocycle analog derivative and its medical application of c Met kinase inhibitors |
Non-Patent Citations (4)
Title |
---|
Copper-catalyzed three-component synthesis of aminonaphthoquinone–sulfonylamidine conjugates and in vitro evaluation of their antiproliferative activity;Thachapully D. Suja 等;<Bioorganic & Medicinal Chemistry Letters>;20161231;第26卷;2072-2076 * |
Qiaoling Xu 等.Design, synthesis, and biological evaluation of 4-((6,7-dimethoxyquinoline- 4-yl)oxy)aniline derivatives.《Bioorganic & Medicinal Chemistry Letters》.2019,第29卷1-7. * |
synthesis and in vitro activity of N-sulfonylamidine-derived pyrimidine analogues;luka krstulovic 等;《croat.chem.acta》;20171231;第90卷(第4期);625-636 * |
TS-Chemscore, a Target-Specific Scoring Function, Significantly Improves the Performance of Scoring in Virtual Screening;Wen-Jing Wang 等;《Chem Biol Drug Des》;20151231;第86卷;1-8 * |
Also Published As
Publication number | Publication date |
---|---|
CN113372267A (en) | 2021-09-10 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2021169963A1 (en) | Aromatic compound and use thereof in preparing antineoplastic drugs | |
CN102796124B (en) | Double β carbolines alkaloid compounds, its preparation method and its pharmaceutical composition and purposes | |
WO2023280182A1 (en) | Compound serving as kat6 inhibitor | |
CN104812761B (en) | Double B-carboline alkaloid compounds, its preparation method and its pharmaceutical composition and purposes | |
CN109134586B (en) | Triptolide derivatives and their applications | |
CN115322158A (en) | Substituted quinazolines as KRASG12C protein inhibitors | |
WO2021249558A1 (en) | Pteridone derivative as rsk inhibitor and application thereof | |
WO2013107428A1 (en) | 7-substituted hanfangichin b derivative, and preparation method and use thereof | |
CN113372267B (en) | C-Met kinase inhibitor | |
CN103922992B (en) | A kind of antitumour activity indolone derivatives, preparation method and use | |
CN111732597B (en) | Preparation and application of 2-aminopyrimidine heterocyclic compound containing 4-amidophenoxy | |
US20130225641A1 (en) | Anticancer compounds and preparation methods thereof | |
CN110467601B (en) | A kind of pyrazole bipyridone compound, intermediate and preparation method and application thereof | |
CN110407839B (en) | Preparation and application of triazole heterocyclic compound containing heteroaryl amide structure | |
CN103910643B (en) | Anti-cancer activity ketone derivative as well as synthetic method and application thereof | |
CN104586842B (en) | Anti-cancer activity indole derivative, synthesis method and uses thereof | |
US20170029404A1 (en) | Polysubstituted pyridine compound, preparation method, use and pharmaceutical composition | |
WO2022166990A1 (en) | Anti-tumor pharmaceutical combination | |
CN102688234A (en) | Synthesis of indolone derivative as RSK2 inhibitor and application | |
CN102584679B (en) | Benzocarbazole acylamide compound and preparation method and application thereof | |
CN107935995A (en) | A kind of new 2 anilino-pyrimidine derivative and its application in antitumor drug is prepared | |
CN114751858A (en) | Tranexamic acid derivatives containing quinoline group and their preparation and application | |
CN112876457A (en) | Novel 4-phenoxypyridine derivative and application thereof | |
CN115490689A (en) | Irreversible KRAS G12C Preparation of inhibitors and uses thereof | |
CN102558144A (en) | Aryl urea derivatives |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination | ||
GR01 | Patent grant | ||
GR01 | Patent grant |