CN111662271A - Compound with IDH mutant inhibitory activity and preparation method and application thereof - Google Patents
Compound with IDH mutant inhibitory activity and preparation method and application thereof Download PDFInfo
- Publication number
- CN111662271A CN111662271A CN201910174468.5A CN201910174468A CN111662271A CN 111662271 A CN111662271 A CN 111662271A CN 201910174468 A CN201910174468 A CN 201910174468A CN 111662271 A CN111662271 A CN 111662271A
- Authority
- CN
- China
- Prior art keywords
- pharmaceutically acceptable
- compound
- idh2
- acceptable salt
- triazine compound
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Granted
Links
- 238000002360 preparation method Methods 0.000 title claims abstract description 35
- 150000001875 compounds Chemical class 0.000 title claims description 63
- 230000002401 inhibitory effect Effects 0.000 title description 18
- -1 s-triazine compound Chemical class 0.000 claims abstract description 68
- 101000599886 Homo sapiens Isocitrate dehydrogenase [NADP], mitochondrial Proteins 0.000 claims abstract description 37
- 102100037845 Isocitrate dehydrogenase [NADP], mitochondrial Human genes 0.000 claims abstract description 36
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 25
- 230000035772 mutation Effects 0.000 claims abstract description 20
- 150000003839 salts Chemical class 0.000 claims abstract description 20
- 239000003814 drug Substances 0.000 claims abstract description 18
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 15
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 claims description 18
- 238000006243 chemical reaction Methods 0.000 claims description 17
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 claims description 14
- 239000003112 inhibitor Substances 0.000 claims description 12
- 125000000217 alkyl group Chemical group 0.000 claims description 10
- 239000002246 antineoplastic agent Substances 0.000 claims description 9
- 201000011510 cancer Diseases 0.000 claims description 8
- 229910052736 halogen Inorganic materials 0.000 claims description 8
- 150000002367 halogens Chemical group 0.000 claims description 8
- 229910052739 hydrogen Inorganic materials 0.000 claims description 8
- 239000001257 hydrogen Substances 0.000 claims description 8
- 125000002887 hydroxy group Chemical group [H]O* 0.000 claims description 8
- 229910052757 nitrogen Inorganic materials 0.000 claims description 8
- 239000000556 agonist Substances 0.000 claims description 7
- 150000002431 hydrogen Chemical group 0.000 claims description 6
- XHXFXVLFKHQFAL-UHFFFAOYSA-N phosphoryl trichloride Chemical compound ClP(Cl)(Cl)=O XHXFXVLFKHQFAL-UHFFFAOYSA-N 0.000 claims description 6
- FYSNRJHAOHDILO-UHFFFAOYSA-N thionyl chloride Chemical compound ClS(Cl)=O FYSNRJHAOHDILO-UHFFFAOYSA-N 0.000 claims description 6
- 229910052799 carbon Inorganic materials 0.000 claims description 5
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 claims description 4
- 150000001732 carboxylic acid derivatives Chemical class 0.000 claims description 4
- 239000000463 material Substances 0.000 claims description 4
- DOWNSQADAFSSAR-UHFFFAOYSA-N 2-bromo-6-(trifluoromethyl)pyridine Chemical compound FC(F)(F)C1=CC=CC(Br)=N1 DOWNSQADAFSSAR-UHFFFAOYSA-N 0.000 claims description 3
- 229940076838 Immune checkpoint inhibitor Drugs 0.000 claims description 3
- 102000037982 Immune checkpoint proteins Human genes 0.000 claims description 3
- 108091008036 Immune checkpoint proteins Proteins 0.000 claims description 3
- 102000037984 Inhibitory immune checkpoint proteins Human genes 0.000 claims description 3
- 108091008026 Inhibitory immune checkpoint proteins Proteins 0.000 claims description 3
- OHJMTUPIZMNBFR-UHFFFAOYSA-N biuret Chemical group NC(=O)NC(N)=O OHJMTUPIZMNBFR-UHFFFAOYSA-N 0.000 claims description 3
- DOBRDRYODQBAMW-UHFFFAOYSA-N copper(i) cyanide Chemical compound [Cu+].N#[C-] DOBRDRYODQBAMW-UHFFFAOYSA-N 0.000 claims description 3
- 229940127089 cytotoxic agent Drugs 0.000 claims description 3
- 239000012274 immune-checkpoint protein inhibitor Substances 0.000 claims description 3
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 claims description 2
- XFXPMWWXUTWYJX-UHFFFAOYSA-N Cyanide Chemical compound N#[C-] XFXPMWWXUTWYJX-UHFFFAOYSA-N 0.000 claims description 2
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 claims description 2
- 150000001263 acyl chlorides Chemical class 0.000 claims description 2
- 125000003368 amide group Chemical group 0.000 claims description 2
- 150000001412 amines Chemical class 0.000 claims description 2
- 150000003927 aminopyridines Chemical class 0.000 claims description 2
- 239000003443 antiviral agent Substances 0.000 claims description 2
- 150000004982 aromatic amines Chemical class 0.000 claims description 2
- 125000001309 chloro group Chemical group Cl* 0.000 claims description 2
- 125000004029 hydroxymethyl group Chemical group [H]OC([H])([H])* 0.000 claims description 2
- 150000004702 methyl esters Chemical class 0.000 claims description 2
- 238000010534 nucleophilic substitution reaction Methods 0.000 claims description 2
- 238000010992 reflux Methods 0.000 claims description 2
- 125000001424 substituent group Chemical group 0.000 claims description 2
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 claims description 2
- 229960005486 vaccine Drugs 0.000 claims description 2
- 229960004854 viral vaccine Drugs 0.000 claims description 2
- 238000004519 manufacturing process Methods 0.000 claims 2
- 230000000118 anti-neoplastic effect Effects 0.000 claims 1
- 229910052801 chlorine Inorganic materials 0.000 claims 1
- 239000000460 chlorine Substances 0.000 claims 1
- 229940127072 targeted antineoplastic agent Drugs 0.000 claims 1
- 229940079593 drug Drugs 0.000 abstract description 15
- 201000010099 disease Diseases 0.000 abstract description 7
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 abstract description 7
- 210000004027 cell Anatomy 0.000 description 33
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 27
- 239000007787 solid Substances 0.000 description 25
- 238000005160 1H NMR spectroscopy Methods 0.000 description 20
- 230000015572 biosynthetic process Effects 0.000 description 16
- 108010075869 Isocitrate Dehydrogenase Proteins 0.000 description 15
- 102000012011 Isocitrate Dehydrogenase Human genes 0.000 description 15
- 238000002474 experimental method Methods 0.000 description 15
- 238000002330 electrospray ionisation mass spectrometry Methods 0.000 description 14
- XJLXINKUBYWONI-DQQFMEOOSA-N [[(2r,3r,4r,5r)-5-(6-aminopurin-9-yl)-3-hydroxy-4-phosphonooxyoxolan-2-yl]methoxy-hydroxyphosphoryl] [(2s,3r,4s,5s)-5-(3-carbamoylpyridin-1-ium-1-yl)-3,4-dihydroxyoxolan-2-yl]methyl phosphate Chemical compound NC(=O)C1=CC=C[N+]([C@@H]2[C@H]([C@@H](O)[C@H](COP([O-])(=O)OP(O)(=O)OC[C@@H]3[C@H]([C@@H](OP(O)(O)=O)[C@@H](O3)N3C4=NC=NC(N)=C4N=C3)O)O2)O)=C1 XJLXINKUBYWONI-DQQFMEOOSA-N 0.000 description 13
- 238000003786 synthesis reaction Methods 0.000 description 13
- 238000000034 method Methods 0.000 description 12
- 239000000243 solution Substances 0.000 description 11
- 230000000694 effects Effects 0.000 description 10
- 229930027945 nicotinamide-adenine dinucleotide Natural products 0.000 description 10
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 10
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 9
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 8
- 102100039905 Isocitrate dehydrogenase [NADP] cytoplasmic Human genes 0.000 description 8
- 230000002503 metabolic effect Effects 0.000 description 8
- 230000000144 pharmacologic effect Effects 0.000 description 8
- 101001042041 Bos taurus Isocitrate dehydrogenase [NAD] subunit beta, mitochondrial Proteins 0.000 description 7
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 description 7
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 7
- 101000960234 Homo sapiens Isocitrate dehydrogenase [NADP] cytoplasmic Proteins 0.000 description 7
- 238000001816 cooling Methods 0.000 description 7
- 238000001035 drying Methods 0.000 description 7
- 239000007924 injection Substances 0.000 description 7
- 238000002347 injection Methods 0.000 description 7
- 230000035755 proliferation Effects 0.000 description 7
- 238000003756 stirring Methods 0.000 description 7
- 238000012360 testing method Methods 0.000 description 7
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 6
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 6
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 6
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 6
- 230000008859 change Effects 0.000 description 6
- 210000001853 liver microsome Anatomy 0.000 description 6
- 239000000203 mixture Substances 0.000 description 6
- 230000008569 process Effects 0.000 description 6
- 230000002829 reductive effect Effects 0.000 description 6
- 210000004881 tumor cell Anatomy 0.000 description 6
- HWXBTNAVRSUOJR-UHFFFAOYSA-N 2-hydroxyglutaric acid Chemical compound OC(=O)C(O)CCC(O)=O HWXBTNAVRSUOJR-UHFFFAOYSA-N 0.000 description 5
- SXFBKPOTJNVNEW-UHFFFAOYSA-N 4-chloro-n-[2-(trifluoromethyl)pyridin-4-yl]-6-[6-(trifluoromethyl)pyridin-2-yl]-1,3,5-triazin-2-amine Chemical compound FC(F)(F)C1=CC=CC(C=2N=C(NC=3C=C(N=CC=3)C(F)(F)F)N=C(Cl)N=2)=N1 SXFBKPOTJNVNEW-UHFFFAOYSA-N 0.000 description 5
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 5
- 229940041181 antineoplastic drug Drugs 0.000 description 5
- 230000024245 cell differentiation Effects 0.000 description 5
- 238000000338 in vitro Methods 0.000 description 5
- 239000000725 suspension Substances 0.000 description 5
- 230000004102 tricarboxylic acid cycle Effects 0.000 description 5
- 238000005406 washing Methods 0.000 description 5
- JBXAKQJKSGOFNP-UHFFFAOYSA-N 6-[6-(trifluoromethyl)pyridin-2-yl]-1h-1,3,5-triazine-2,4-dione Chemical compound FC(F)(F)C1=CC=CC(C=2NC(=O)NC(=O)N=2)=N1 JBXAKQJKSGOFNP-UHFFFAOYSA-N 0.000 description 4
- 241000699666 Mus <mouse, genus> Species 0.000 description 4
- 238000001727 in vivo Methods 0.000 description 4
- 238000011534 incubation Methods 0.000 description 4
- 230000003834 intracellular effect Effects 0.000 description 4
- 238000001294 liquid chromatography-tandem mass spectrometry Methods 0.000 description 4
- AYABEJGGSWJVPN-UHFFFAOYSA-N methyl 6-(trifluoromethyl)pyridine-2-carboxylate Chemical compound COC(=O)C1=CC=CC(C(F)(F)F)=N1 AYABEJGGSWJVPN-UHFFFAOYSA-N 0.000 description 4
- 239000013641 positive control Substances 0.000 description 4
- 239000000758 substrate Substances 0.000 description 4
- KQIGMPWTAHJUMN-GSVOUGTGSA-N (2r)-3-aminopropane-1,2-diol Chemical compound NC[C@@H](O)CO KQIGMPWTAHJUMN-GSVOUGTGSA-N 0.000 description 3
- GSSCQXYNRDFGAV-UHFFFAOYSA-N 2,4-dichloro-6-[6-(trifluoromethyl)pyridin-2-yl]-1,3,5-triazine Chemical compound FC(F)(F)C1=CC=CC(C=2N=C(Cl)N=C(Cl)N=2)=N1 GSSCQXYNRDFGAV-UHFFFAOYSA-N 0.000 description 3
- KJJPLEZQSCZCKE-UHFFFAOYSA-N 2-aminopropane-1,3-diol Chemical compound OCC(N)CO KJJPLEZQSCZCKE-UHFFFAOYSA-N 0.000 description 3
- KPGXRSRHYNQIFN-UHFFFAOYSA-N 2-oxoglutaric acid Chemical compound OC(=O)CCC(=O)C(O)=O KPGXRSRHYNQIFN-UHFFFAOYSA-N 0.000 description 3
- KQIGMPWTAHJUMN-UHFFFAOYSA-N 3-aminopropane-1,2-diol Chemical compound NCC(O)CO KQIGMPWTAHJUMN-UHFFFAOYSA-N 0.000 description 3
- KQIGMPWTAHJUMN-VKHMYHEASA-N 3-aminopropane-1,2-diol Chemical compound NC[C@H](O)CO KQIGMPWTAHJUMN-VKHMYHEASA-N 0.000 description 3
- LXNGKPOIZPTXDL-UHFFFAOYSA-N 6-(trifluoromethyl)pyridine-2-carbonitrile Chemical compound FC(F)(F)C1=CC=CC(C#N)=N1 LXNGKPOIZPTXDL-UHFFFAOYSA-N 0.000 description 3
- OKBHXGBLXDNJJD-UHFFFAOYSA-N 6-(trifluoromethyl)pyridine-2-carboxylic acid Chemical compound OC(=O)C1=CC=CC(C(F)(F)F)=N1 OKBHXGBLXDNJJD-UHFFFAOYSA-N 0.000 description 3
- 206010009944 Colon cancer Diseases 0.000 description 3
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 3
- 102000016680 Dioxygenases Human genes 0.000 description 3
- 108010028143 Dioxygenases Proteins 0.000 description 3
- 102000004190 Enzymes Human genes 0.000 description 3
- 108090000790 Enzymes Proteins 0.000 description 3
- 208000032612 Glial tumor Diseases 0.000 description 3
- 206010018338 Glioma Diseases 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 108010033040 Histones Proteins 0.000 description 3
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 3
- 241000699670 Mus sp. Species 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 230000004663 cell proliferation Effects 0.000 description 3
- 208000006990 cholangiocarcinoma Diseases 0.000 description 3
- 208000029742 colonic neoplasm Diseases 0.000 description 3
- 238000011161 development Methods 0.000 description 3
- 230000018109 developmental process Effects 0.000 description 3
- 238000010790 dilution Methods 0.000 description 3
- 239000012895 dilution Substances 0.000 description 3
- 238000006911 enzymatic reaction Methods 0.000 description 3
- 230000001939 inductive effect Effects 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- 239000002609 medium Substances 0.000 description 3
- 230000035790 physiological processes and functions Effects 0.000 description 3
- NLKNQRATVPKPDG-UHFFFAOYSA-M potassium iodide Chemical compound [K+].[I-] NLKNQRATVPKPDG-UHFFFAOYSA-M 0.000 description 3
- 230000001737 promoting effect Effects 0.000 description 3
- 239000011734 sodium Substances 0.000 description 3
- 229910052708 sodium Inorganic materials 0.000 description 3
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical class O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 3
- 239000002904 solvent Substances 0.000 description 3
- 238000007920 subcutaneous administration Methods 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- 239000003826 tablet Substances 0.000 description 3
- YLQBMQCUIZJEEH-UHFFFAOYSA-N tetrahydrofuran Natural products C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 description 3
- 238000002054 transplantation Methods 0.000 description 3
- WNWHHMBRJJOGFJ-UHFFFAOYSA-N 16-methylheptadecan-1-ol Chemical class CC(C)CCCCCCCCCCCCCCCO WNWHHMBRJJOGFJ-UHFFFAOYSA-N 0.000 description 2
- 208000031261 Acute myeloid leukaemia Diseases 0.000 description 2
- 208000003174 Brain Neoplasms Diseases 0.000 description 2
- 208000005623 Carcinogenesis Diseases 0.000 description 2
- 230000026641 DNA hypermethylation Effects 0.000 description 2
- 101000599885 Dictyostelium discoideum Isocitrate dehydrogenase [NADP], mitochondrial Proteins 0.000 description 2
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 2
- 102000003951 Erythropoietin Human genes 0.000 description 2
- 108090000394 Erythropoietin Proteins 0.000 description 2
- 206010064571 Gene mutation Diseases 0.000 description 2
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 description 2
- 108700020796 Oncogene Proteins 0.000 description 2
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical class [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 2
- 239000007983 Tris buffer Substances 0.000 description 2
- 238000002835 absorbance Methods 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 150000001413 amino acids Chemical class 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 230000000259 anti-tumor effect Effects 0.000 description 2
- 239000008346 aqueous phase Substances 0.000 description 2
- 230000036952 cancer formation Effects 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 231100000504 carcinogenesis Toxicity 0.000 description 2
- 230000003197 catalytic effect Effects 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 239000002552 dosage form Substances 0.000 description 2
- 230000008030 elimination Effects 0.000 description 2
- 238000003379 elimination reaction Methods 0.000 description 2
- 229940105423 erythropoietin Drugs 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- 239000000706 filtrate Substances 0.000 description 2
- 238000001914 filtration Methods 0.000 description 2
- 239000005457 ice water Substances 0.000 description 2
- 239000012535 impurity Substances 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- ODBLHEXUDAPZAU-UHFFFAOYSA-N isocitric acid Chemical compound OC(=O)C(O)C(C(O)=O)CC(O)=O ODBLHEXUDAPZAU-UHFFFAOYSA-N 0.000 description 2
- 208000032839 leukemia Diseases 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 239000002207 metabolite Substances 0.000 description 2
- BDAGIHXWWSANSR-UHFFFAOYSA-N methanoic acid Natural products OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 2
- 231100000590 oncogenic Toxicity 0.000 description 2
- 230000002246 oncogenic effect Effects 0.000 description 2
- 239000008188 pellet Substances 0.000 description 2
- 239000000546 pharmaceutical excipient Substances 0.000 description 2
- 239000012071 phase Substances 0.000 description 2
- 239000006187 pill Substances 0.000 description 2
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 2
- OXCMYAYHXIHQOA-UHFFFAOYSA-N potassium;[2-butyl-5-chloro-3-[[4-[2-(1,2,4-triaza-3-azanidacyclopenta-1,4-dien-5-yl)phenyl]phenyl]methyl]imidazol-4-yl]methanol Chemical compound [K+].CCCCC1=NC(Cl)=C(CO)N1CC1=CC=C(C=2C(=CC=CC=2)C2=N[N-]N=N2)C=C1 OXCMYAYHXIHQOA-UHFFFAOYSA-N 0.000 description 2
- 230000008672 reprogramming Effects 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 238000013112 stability test Methods 0.000 description 2
- 238000000967 suction filtration Methods 0.000 description 2
- 239000006228 supernatant Substances 0.000 description 2
- 239000000829 suppository Substances 0.000 description 2
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 2
- JNYAEWCLZODPBN-JGWLITMVSA-N (2r,3r,4s)-2-[(1r)-1,2-dihydroxyethyl]oxolane-3,4-diol Chemical compound OC[C@@H](O)[C@H]1OC[C@H](O)[C@H]1O JNYAEWCLZODPBN-JGWLITMVSA-N 0.000 description 1
- LYNBZRJTRHTSKI-UHFFFAOYSA-N 2-(trifluoromethyl)pyridin-4-amine Chemical compound NC1=CC=NC(C(F)(F)F)=C1 LYNBZRJTRHTSKI-UHFFFAOYSA-N 0.000 description 1
- VIUDTWATMPPKEL-UHFFFAOYSA-N 3-(trifluoromethyl)aniline Chemical compound NC1=CC=CC(C(F)(F)F)=C1 VIUDTWATMPPKEL-UHFFFAOYSA-N 0.000 description 1
- OSWFIVFLDKOXQC-UHFFFAOYSA-N 4-(3-methoxyphenyl)aniline Chemical compound COC1=CC=CC(C=2C=CC(N)=CC=2)=C1 OSWFIVFLDKOXQC-UHFFFAOYSA-N 0.000 description 1
- LRSASMSXMSNRBT-UHFFFAOYSA-N 5-methylcytosine Chemical compound CC1=CNC(=O)N=C1N LRSASMSXMSNRBT-UHFFFAOYSA-N 0.000 description 1
- OBYJTLDIQBWBHM-UHFFFAOYSA-N 6-chloropyridin-2-amine Chemical compound NC1=CC=CC(Cl)=N1 OBYJTLDIQBWBHM-UHFFFAOYSA-N 0.000 description 1
- 101100125460 Arabidopsis thaliana IDH3 gene Proteins 0.000 description 1
- 206010004593 Bile duct cancer Diseases 0.000 description 1
- 206010005003 Bladder cancer Diseases 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- 239000012275 CTLA-4 inhibitor Substances 0.000 description 1
- 229940045513 CTLA4 antagonist Drugs 0.000 description 1
- 206010008342 Cervix carcinoma Diseases 0.000 description 1
- 208000005243 Chondrosarcoma Diseases 0.000 description 1
- 102100031162 Collagen alpha-1(XVIII) chain Human genes 0.000 description 1
- 206010014733 Endometrial cancer Diseases 0.000 description 1
- 206010014759 Endometrial neoplasm Diseases 0.000 description 1
- 108010079505 Endostatins Proteins 0.000 description 1
- 208000000461 Esophageal Neoplasms 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
- 102100034458 Hepatitis A virus cellular receptor 2 Human genes 0.000 description 1
- 101710083479 Hepatitis A virus cellular receptor 2 homolog Proteins 0.000 description 1
- 102000008157 Histone Demethylases Human genes 0.000 description 1
- 108010074870 Histone Demethylases Proteins 0.000 description 1
- 102100032742 Histone-lysine N-methyltransferase SETD2 Human genes 0.000 description 1
- 102000006947 Histones Human genes 0.000 description 1
- 101000746373 Homo sapiens Granulocyte-macrophage colony-stimulating factor Proteins 0.000 description 1
- 101000654725 Homo sapiens Histone-lysine N-methyltransferase SETD2 Proteins 0.000 description 1
- 101000615488 Homo sapiens Methyl-CpG-binding domain protein 2 Proteins 0.000 description 1
- 101000955999 Homo sapiens V-set domain-containing T-cell activation inhibitor 1 Proteins 0.000 description 1
- 101000666896 Homo sapiens V-type immunoglobulin domain-containing suppressor of T-cell activation Proteins 0.000 description 1
- 206010021143 Hypoxia Diseases 0.000 description 1
- 101150020771 IDH gene Proteins 0.000 description 1
- 101150104906 Idh2 gene Proteins 0.000 description 1
- 229940122827 Isocitrate dehydrogenase inhibitor Drugs 0.000 description 1
- 108010044467 Isoenzymes Proteins 0.000 description 1
- 102000019065 Jumonji Domain-Containing Histone Demethylases Human genes 0.000 description 1
- 108010051918 Jumonji Domain-Containing Histone Demethylases Proteins 0.000 description 1
- 208000008839 Kidney Neoplasms Diseases 0.000 description 1
- 229940125563 LAG3 inhibitor Drugs 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 206010064912 Malignant transformation Diseases 0.000 description 1
- 206010027406 Mesothelioma Diseases 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 102100021299 Methyl-CpG-binding domain protein 2 Human genes 0.000 description 1
- 102000008109 Mixed Function Oxygenases Human genes 0.000 description 1
- 108010074633 Mixed Function Oxygenases Proteins 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-Dimethylformamide Chemical compound CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 1
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-diisopropylethylamine Substances CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 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
- 239000012270 PD-1 inhibitor Substances 0.000 description 1
- 239000012668 PD-1-inhibitor Substances 0.000 description 1
- 239000012271 PD-L1 inhibitor Substances 0.000 description 1
- 239000012272 PD-L2 inhibitor Substances 0.000 description 1
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 1
- 239000004698 Polyethylene Substances 0.000 description 1
- 102000004079 Prolyl Hydroxylases Human genes 0.000 description 1
- 108010043005 Prolyl Hydroxylases Proteins 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 1
- 229940079156 Proteasome inhibitor Drugs 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- 229940044665 STING agonist Drugs 0.000 description 1
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 1
- 208000005718 Stomach Neoplasms Diseases 0.000 description 1
- 101100215487 Sus scrofa ADRA2A gene Proteins 0.000 description 1
- 229940126547 T-cell immunoglobulin mucin-3 Drugs 0.000 description 1
- 102000043123 TET family Human genes 0.000 description 1
- 108091084976 TET family Proteins 0.000 description 1
- 229940125555 TIGIT inhibitor Drugs 0.000 description 1
- 208000024313 Testicular Neoplasms Diseases 0.000 description 1
- 206010057644 Testis cancer Diseases 0.000 description 1
- 208000024770 Thyroid neoplasm Diseases 0.000 description 1
- JLRGJRBPOGGCBT-UHFFFAOYSA-N Tolbutamide Chemical compound CCCCNC(=O)NS(=O)(=O)C1=CC=C(C)C=C1 JLRGJRBPOGGCBT-UHFFFAOYSA-N 0.000 description 1
- 229940122429 Tubulin inhibitor Drugs 0.000 description 1
- 101710165473 Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 description 1
- 102100022153 Tumor necrosis factor receptor superfamily member 4 Human genes 0.000 description 1
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 description 1
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 description 1
- 102100038929 V-set domain-containing T-cell activation inhibitor 1 Human genes 0.000 description 1
- 102100038282 V-type immunoglobulin domain-containing suppressor of T-cell activation Human genes 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 239000002168 alkylating agent Substances 0.000 description 1
- 229940100198 alkylating agent Drugs 0.000 description 1
- 229940009533 alpha-ketoglutaric acid Drugs 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 230000003698 anagen phase Effects 0.000 description 1
- 206010002449 angioimmunoblastic T-cell lymphoma Diseases 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 239000002256 antimetabolite Substances 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 239000002585 base Substances 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- 208000026900 bile duct neoplasm Diseases 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 238000006555 catalytic reaction Methods 0.000 description 1
- 230000022131 cell cycle Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 201000010881 cervical cancer Diseases 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 208000035250 cutaneous malignant susceptibility to 1 melanoma Diseases 0.000 description 1
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 230000017858 demethylation Effects 0.000 description 1
- 238000010520 demethylation reaction Methods 0.000 description 1
- 235000014113 dietary fatty acids Nutrition 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- DZHFTEDSQFPDPP-HWYNEVGZSA-L disodium;(2r)-2-hydroxypentanedioate Chemical group [Na+].[Na+].[O-]C(=O)[C@H](O)CCC([O-])=O DZHFTEDSQFPDPP-HWYNEVGZSA-L 0.000 description 1
- 239000003534 dna topoisomerase inhibitor Substances 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 238000010828 elution Methods 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 230000037149 energy metabolism Effects 0.000 description 1
- 230000006718 epigenetic regulation Effects 0.000 description 1
- 201000004101 esophageal cancer Diseases 0.000 description 1
- 235000019441 ethanol Nutrition 0.000 description 1
- 229930195729 fatty acid Natural products 0.000 description 1
- 239000000194 fatty acid Substances 0.000 description 1
- 239000012065 filter cake Substances 0.000 description 1
- IJJVMEJXYNJXOJ-UHFFFAOYSA-N fluquinconazole Chemical compound C=1C=C(Cl)C=C(Cl)C=1N1C(=O)C2=CC(F)=CC=C2N=C1N1C=NC=N1 IJJVMEJXYNJXOJ-UHFFFAOYSA-N 0.000 description 1
- 235000019253 formic acid Nutrition 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 239000012458 free base Substances 0.000 description 1
- 206010017758 gastric cancer Diseases 0.000 description 1
- 238000003304 gavage Methods 0.000 description 1
- 230000014509 gene expression Effects 0.000 description 1
- 230000030279 gene silencing Effects 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 208000005017 glioblastoma Diseases 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 150000003278 haem Chemical class 0.000 description 1
- 201000010536 head and neck cancer Diseases 0.000 description 1
- 208000014829 head and neck neoplasm Diseases 0.000 description 1
- 238000010438 heat treatment Methods 0.000 description 1
- 150000002391 heterocyclic compounds Chemical class 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 102000046157 human CSF2 Human genes 0.000 description 1
- 230000007954 hypoxia Effects 0.000 description 1
- 101150046722 idh1 gene Proteins 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 238000011081 inoculation Methods 0.000 description 1
- 208000014899 intrahepatic bile duct cancer Diseases 0.000 description 1
- 230000002601 intratumoral effect Effects 0.000 description 1
- 201000010982 kidney cancer Diseases 0.000 description 1
- 229940043355 kinase inhibitor Drugs 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 1
- 239000007791 liquid phase Substances 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 201000007270 liver cancer Diseases 0.000 description 1
- 208000014018 liver neoplasm Diseases 0.000 description 1
- 201000005202 lung cancer Diseases 0.000 description 1
- 208000020816 lung neoplasm Diseases 0.000 description 1
- 210000002751 lymph Anatomy 0.000 description 1
- 229920002521 macromolecule Polymers 0.000 description 1
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L magnesium chloride Substances [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 230000036212 malign transformation Effects 0.000 description 1
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 230000006371 metabolic abnormality Effects 0.000 description 1
- 230000037353 metabolic pathway Effects 0.000 description 1
- NDBQJIBNNUJNHA-DFWYDOINSA-N methyl (2s)-2-amino-3-hydroxypropanoate;hydrochloride Chemical compound Cl.COC(=O)[C@@H](N)CO NDBQJIBNNUJNHA-DFWYDOINSA-N 0.000 description 1
- 230000003228 microsomal effect Effects 0.000 description 1
- 210000003470 mitochondria Anatomy 0.000 description 1
- 230000007171 neuropathology Effects 0.000 description 1
- 238000010899 nucleation Methods 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 239000012074 organic phase Substances 0.000 description 1
- 230000036542 oxidative stress Effects 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 208000008443 pancreatic carcinoma Diseases 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 229940121655 pd-1 inhibitor Drugs 0.000 description 1
- 229940121656 pd-l1 inhibitor Drugs 0.000 description 1
- 229940121654 pd-l2 inhibitor Drugs 0.000 description 1
- 210000002824 peroxisome Anatomy 0.000 description 1
- 239000003208 petroleum Substances 0.000 description 1
- 239000002504 physiological saline solution Substances 0.000 description 1
- 229910052697 platinum Inorganic materials 0.000 description 1
- 229920000573 polyethylene Polymers 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 230000001376 precipitating effect Effects 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 230000002062 proliferating effect Effects 0.000 description 1
- 229960004063 propylene glycol Drugs 0.000 description 1
- 239000003207 proteasome inhibitor Substances 0.000 description 1
- 239000003909 protein kinase inhibitor Substances 0.000 description 1
- 108090000623 proteins and genes Proteins 0.000 description 1
- 102000004169 proteins and genes Human genes 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 238000002390 rotary evaporation Methods 0.000 description 1
- 229920006395 saturated elastomer Polymers 0.000 description 1
- 238000007493 shaping process Methods 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 239000000741 silica gel Substances 0.000 description 1
- 229910002027 silica gel Inorganic materials 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 201000011549 stomach cancer Diseases 0.000 description 1
- 239000012089 stop solution Substances 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 201000003120 testicular cancer Diseases 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 201000002510 thyroid cancer Diseases 0.000 description 1
- JMXKSZRRTHPKDL-UHFFFAOYSA-N titanium ethoxide Chemical compound [Ti+4].CC[O-].CC[O-].CC[O-].CC[O-] JMXKSZRRTHPKDL-UHFFFAOYSA-N 0.000 description 1
- 229960005371 tolbutamide Drugs 0.000 description 1
- 229940044693 topoisomerase inhibitor Drugs 0.000 description 1
- 230000005748 tumor development Effects 0.000 description 1
- 238000004704 ultra performance liquid chromatography Methods 0.000 description 1
- 201000005112 urinary bladder cancer Diseases 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
Classifications
-
- 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
- C07D401/14—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 three or more hetero rings
-
- 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
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
- C07D401/04—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
Landscapes
- Organic Chemistry (AREA)
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Chemical Kinetics & Catalysis (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 belongs to the field of medicines, and particularly relates to an s-triazine compound with structural characteristics of a general formula I, or a pharmaceutically acceptable salt, a pharmaceutical composition, a preparation method and application thereof in preparation of an IDH2 mutant inhibitorCan be used for preparing medicaments for preventing and/or treating various related diseases caused by IDH2 mutation, wherein the diseases comprise cancers carrying IDH2 mutation.
Description
Technical Field
The invention belongs to the field of medicines, and particularly relates to an s-triazine compound with IDH mutant inhibitory activity or pharmaceutically acceptable salts thereof, a pharmaceutical composition, a preparation method thereof, and application thereof as an isocitrate dehydrogenase 2 mutant (mIDH2) inhibitor.
Background
The tricarboxylic acid cycle is a common metabolic pathway of three major nutrients of carbohydrates, lipids and amino acids in human bodies, not only is a main way for the bodies to obtain energy, but also provides important small molecule precursors for the biosynthesis of other substances. Thus, the tricarboxylic acid cycle has important physiological significance.
The tricarboxylic acid cycle consists of a series of enzymatic reactions. Among them, Isocitrate Dehydrogenase (IDH) is a key rate-limiting enzyme in the tricarboxylic acid cycle, and is responsible for catalyzing the conversion of isocitrate to α -ketoglutarate (α -KG). There are 3 kinds of IDH isozymes in human, of which IDH1 is localized to cytoplasm and peroxisome, while IDH2 and IDH3 are localized to mitochondria. Different IDH subtypes have their respective physiological functions, but in general they play important roles in processes such as energy metabolism, biosynthesis, and resistance to oxidative stress (Amino Acids,2017,49(1): 21-32).
The alpha-KG generated by IDH catalysis is not only involved in tricarboxylic acid cycle, but also is a cofactor of a plurality of dioxygenases in vivo, thus playing an important role in maintaining physiological functions of organisms. These dioxygenases include, among others, the JmjC domain-containing histone demethylases closely associated with tumor development and the TET family of 5-methylcytosine hydroxylases. They are able to regulate the demethylation process of histones and DNA, thus affecting DNA conformation, DNA stability and the way DNA interacts with proteins, ultimately altering gene expression (Cancer Lett,2015,356(2): 309-.
Rapid proliferation is the most prominent biological feature of tumor cells, while metabolic abnormalities are another fundamental property. During tumorigenesis, the cellular metabolic network needs to balance energy requirements and biosynthesis requirements through reprogramming so as to be beneficial to synthesizing biological macromolecules required by various cell structures, thereby meeting the requirement of rapid proliferation of cells. Metabolic reprogramming therefore plays an important role in the process of malignant transformation of cells as a result of genetic events (Neuropathology,2019,39(1): 3-13.). IDH1 and IDH2 gene mutations have been found to be closely related to human tumors.
In 2008, IDH gene mutation was first discovered in glioma patients by Parsons et al, university of John Hopkins (Science,2008,321(5897): 1807-1812). Subsequently, H84 and IDH 84 mutations were detected in a series of patients with colon Cancer (Oncogene,2010,29(49): 6409-. Numerous studies have shown that mutations in the IDH1 and IDH2 genes play an important role in human tumorigenesis and development, mainly in relation to the oncogenic metabolites they catalyze to produce.
Clinical studies have shown that IDH mutations in tumor patients occur predominantly at key amino acid residues in the active sites of IDH1 and IDH 2. Mutations in IDH1 occurred primarily at R132, with the highest frequency of mutations at R132H, followed by the R132C mutation. The mutation types of IDH2 include R140Q, R140L, R140W, R172K, R172M, R172S, R172G and R172W, wherein the most important mutation type is R140Q and the second is R172K mutation (Science,2008,321(5897):1807 and 1812). These IDH mutants (mIDH) lose the normal physiological functions of wild-type IDH, but gain a new catalytic function, which can convert alpha-KG to R (-) -2-hydroxyglutarate (2-HG) with the aid of NADPH, resulting in a large accumulation of intracellular 2-HG (Nature,2010,465(7300): 966; Cancer Cell,2010,17(3): 225-234). 2-HG is generally considered to be an oncogenic metabolite. This is probably due to the structural similarity of 2-HG and alpha-KG, which makes it occupy the same binding pocket of alpha-KG, thus competitively inhibiting alpha-KG dependent dioxygenase, including histone demethylase, DNA demethylase and proline hydroxylase, resulting in abnormal epigenetic regulation, causing histone and DNA hypermethylation, further inducing oncogene silencing, affecting normal differentiation of cells, promoting proliferation of cells, and finally promoting tumor generation and development (Cancer, Cell 2011,19(1): 17-30; Nature,2012,483(7390): 474-) -478). Meanwhile, excessive accumulation of 2-HG also causes the intracellular hypoxia inducible factor HIF-1 alpha to be increased, so as to reduce the content of endostatin, thereby promoting the generation and development of tumor vessels (Cancer Cell,2013,23(3): 274-276). Therefore, the IDH mutant becomes a new target for developing anti-cancer drugs.
In recent years, various academic institutions and pharmaceutical companies have successively reported IDH1 and IDH2 mutant inhibitors developed respectively, however, only a few candidate drugs are currently in clinical trials (J Med Chem,2018,61(20): 8981-. Among them, AG-221 and AG-120, developed by Agios corporation, were subsequently approved by the U.S. FDA for marketing, and were used to treat refractory and relapsed acute myeloid leukemia, respectively, carrying IDH2 and IDH1 mutations. IDH1 and IDH2 mutant inhibitors can reverse histone and DNA hypermethylation by reducing 2-HG level in tumor cells, thereby inducing tumor cell differentiation and playing the role of anti-tumor.
Disclosure of Invention
The purpose of the invention is as follows: in view of the prior art, the invention provides a heterocyclic compound or a pharmaceutically acceptable salt thereof, a preparation method, a pharmaceutical composition and an application thereof, and the compound has good mIDH2 inhibitory activity and can be used for preparing medicines for treating and/or preventing various related diseases caused by IDH2 mutation.
The technical scheme is as follows: the invention discloses an s-triazine compound shown in a general formula I or pharmaceutically acceptable salt thereof:
wherein:
T1and T2Independently selected from N or C; when T is1When is N, T2Is selected from C; when T is1When is C, T2Selected from C or N;
r is selected from hydrogen, halogen, hydroxyl, amido and C1-C4Alkyl or C1-C4Alkoxy, which alkyl may be optionally mono-or poly-substituted with halogen;
Y1、Y2and Y3Each independently selected from hydrogen and C1-C4Alkyl, hydroxy C1-C4Alkyl or carboxyl, which alkyl may optionally be mono-or polysubstituted by the following identical or different substituents: hydrogen, hydroxy, C1-C4Alkyl or hydroxy C1-C4An alkyl group.
Further, R is selected from halogen or C1-C4An alkyl group, which alkyl group may optionally be mono-or poly-substituted with halogen.
Still further, R is selected from chloro or trifluoromethyl.
Further, Y1、Y2And Y3Each independently selected from hydrogen, hydroxymethyl, 1, 2-dihydroxyethyl, 2, 3-dihydroxypropyl or carboxyl.
In particular, the compounds of the formula I are preferably selected from the following compounds I-1 to I-14:
the compound numbers referred to in the following pharmacological experiments are equivalent to the compounds corresponding to the numbers here.
The invention also discloses a preparation method of the compound shown in the general formula I, which comprises the following steps: 2-bromo-6-trifluoromethylpyridine and cuprous cyanide undergo nucleophilic substitution reaction to prepare cyanide 1,1 is hydrolyzed under the condition of concentrated hydrochloric acid to generate carboxylic acid 2, 2 and thionyl chloride are prepared into acyl chloride, and then the carboxylic acid and methanol react to prepare methyl ester 3; 3 and biuret ring to obtain an intermediate 4; 4 refluxing with phosphorus oxychloride to obtain chloride 5, and reacting with different substituted aminopyridine or aromatic amine to obtain an intermediate 6; 6 with fatty amines (Y)1Y2Y3CNH2) The compound with the general formula (I) is prepared by the following synthetic route:
wherein, R, T1、T2、Y1、Y2、Y3Have the meaning of the formula I.
The pharmaceutically acceptable salts of the compounds of formula I can be synthesized by conventional chemical methods. In general, salts can be prepared by reacting the free base or acid with a stoichiometric equivalent or excess of an acid (inorganic or organic) or base (inorganic or organic) in a suitable solvent or solvent composition.
The invention also provides a pharmaceutical composition, which consists of active components with effective treatment amount and pharmaceutically acceptable auxiliary materials; the active component comprises one or more of a compound in a general formula I and pharmaceutically acceptable salts thereof. In the pharmaceutical composition, the auxiliary materials comprise pharmaceutically acceptable carriers, diluents and/or excipients.
The pharmaceutical composition may be formulated into various types of administration unit dosage forms according to the therapeutic purpose, such as tablets, pills, powders, liquids, suspensions, emulsions, granules, capsules, suppositories, injections (solutions and suspensions), and the like, preferably tablets, capsules, liquids, suspensions, and injections (solutions and suspensions). For shaping the pharmaceutical composition in the form of tablets, pills or suppositories, any excipient known and widely used in the art can be used.
For preparing the pharmaceutical composition in the form of injection, the solution or suspension may be sterilized (preferably by adding appropriate amount of sodium chloride, glucose or glycerol) and made into injection with blood isotonic pressure. In the preparation of injection, any carrier commonly used in the art may also be used. For example: water, ethanol, propylene glycol, ethoxylated isostearyl alcohol, polyethoxylated isostearyl alcohol, and fatty acid esters of polyethylene sorbitan, and the like. In addition, usual dissolving agents, buffers and the like may be added.
The content of the composition in the pharmaceutical composition is not particularly limited, and can be selected within a wide range, and generally ranges from 5 to 95% by mass, and preferably ranges from 30 to 85% by mass.
The method of administration of the pharmaceutical composition of the present invention is not particularly limited. The formulation of various dosage forms can be selected for administration according to the age, sex and other conditions and symptoms of the patient.
The invention also discloses application of the compound in the general formula I, the pharmaceutically acceptable salt thereof or the pharmaceutical composition in preparation of an isocitrate dehydrogenase 2(IDH2) mutant inhibitor. The IDH2 mutant inhibitor is used for preventing and treating diseases carrying IDH2 mutation, and the diseases related to IDH2 mutation are cancers.
The invention also discloses an application of the compound with the general formula I, the pharmaceutically acceptable salt thereof or the pharmaceutical composition in the aspect of tumor resistance, wherein the cancer is one or more of malignant melanoma, lung cancer, breast cancer, stomach cancer, colon cancer, bladder cancer, pancreatic cancer, lymph cancer, prostate cancer, testicular cancer, kidney cancer, brain cancer, head and neck cancer, ovarian cancer, cervical cancer, endometrial cancer, mesothelioma, thyroid cancer, liver cancer, esophageal cancer, leukemia, cholangiocarcinoma, chondrosarcoma or angioimmunoblastic T-cell lymphoma; the leukemia is acute myelogenous leukemia; the bile duct cancer is intrahepatic bile duct cancer; the brain tumor is primary glioma or secondary glioma.
The invention also discloses application of the compound with the general formula I, the pharmaceutically acceptable salt thereof or the pharmaceutical composition in combination with one or more chemotherapeutic agents, targeted antitumor drugs, immune checkpoint inhibitors, immune checkpoint agonists, antitumor vaccines, antiviral agents and antiviral vaccines in preparation of drugs for treating IDH2 mutation-carrying cancers. The chemotherapeutic agent is an alkylating agent, a tubulin inhibitor, a topoisomerase inhibitor, a platinum drug, an antimetabolite drug or a hormone antineoplastic drug; the targeted antitumor drug is a protein kinase inhibitor, a proteasome inhibitor, an isocitrate dehydrogenase inhibitor, an antitumor drug based on epigenetics or a cell cycle signal pathway inhibitor; the immune checkpoint inhibitor is a CTLA-4 inhibitor, a PD-1 inhibitor, a PD-L1 inhibitor, a PD-L2 inhibitor, a TIM-3 inhibitor, a VISTA inhibitor, a LAG3 inhibitor, a TIGIT inhibitor, an A2AR inhibitor or a VTCN1 inhibitor; the immune checkpoint agonist is a STING agonist, a 4-1BB agonist, an OX40 agonist, a ROR γ agonist, or an ICOS agonist.
Has the advantages that: the invention provides an s-triazine compound with a brand-new structure, a pharmaceutically acceptable salt thereof, a pharmaceutical composition, a preparation method thereof and application of the compounds in preparation of IDH2 mutant inhibitors. Pharmacological experiment results show that the compound has obvious inhibition effect on the activity of IDH2 mutant (mIDH2), can effectively inhibit the process that mIDH2 catalyzes alpha-ketoglutaric acid to generate 2-hydroxyglutaric acid, and can be used for preparing medicines for preventing and/or treating various related diseases caused by IDH2 mutation, wherein the diseases comprise cancers carrying IDH2 mutation.
Detailed Description
To further illustrate the present invention, a series of examples are given below, which are purely illustrative and are intended to be a detailed description of the invention only and should not be understood as limiting the invention.
Wherein, the positive control drug AG-221 used in the experiment is purchased from MCE company; all cell lines used were purchased from ATCC.
Example 1
Preparation of 6- (trifluoromethyl) pyridine-2-carbonitrile (1)
In a dry 500mL round bottom flask were added 2-bromo-6- (trifluoromethyl) pyridine (25.0g, 110.6mmol), N-dimethylformamide (80mL), cuprous cyanide (14.9g,165.9mmol) and potassium iodide (27.5g, 165.9mmol), respectively, and the mixture was heated to 130 ℃ for 12 h. After cooling, 600mL of ice water was poured in, 300mL of ethyl acetate was added, stirring was carried out for 15 minutes, the solid was filtered off, the filtrate was extracted with ethyl acetate (50 mL. times.12), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a pale yellow liquid which was directly put into the next reaction.
Preparation of 6- (trifluoromethyl) -pyridine-2-carboxylic acid (2)
Adding the intermediate 1 and concentrated hydrochloric acid (80mL) into a dry 250mL round-bottom flask respectively, heating to 80 ℃ to react for 6h, cooling, adding 500mL of water for dilution, adjusting the pH value of a saturated sodium bicarbonate solution to 8, extracting impurities by ethyl acetate (50mL × 6), adjusting the pH value of an aqueous phase to 5 by concentrated hydrochloric acid, precipitating a large amount of white solid, carrying out suction filtration, and drying to obtain 16.5g of white solid.1H NMR(DMSO-d6,300MHz)(ppm):13.10(s,1H),7.99-8.17(m,3H).
Preparation of methyl 6- (trifluoromethyl) -pyridine-2-carboxylate (3)
Intermediate 2(16.5g, 86.3mmol) and methanol (80mL) were added to a dry 250mL round bottom flask, and thionyl chloride (15.4g, 129.5mmol) was slowly added dropwise with stirring, and the mixture was allowed to rise to 80 ℃ for 12 h. Cooling, concentrating to dryness, adjusting pH to 8 with saturated sodium bicarbonate solution, filtering, and drying to obtain white solid 16.0g with yield of 90.4%.1H NMR(DMSO-d6,300MHz)(ppm):8.03-8.18(m,3H),3.80(s,3H).
Preparation of 6- (6- (trifluoromethyl) pyridin-2-yl) -1,3, 5-triazine-2, 4- (1H,3H) -dione (4)
Adding the intermediate 3(16.0g, 78.0mmol), biuret (9.6g, 93.2mmol) and absolute ethyl alcohol (80mL) into a dry 500mL round-bottom flask respectively, stirring at room temperature for 20min, adding ethyl titanate (53.4g, 234.2mmol), stirring for 1h under the protection of nitrogen, chopping sodium blocks (7.2g, 313.0mmol), adding into 80mL absolute ethyl alcohol, placing at room temperature under the protection of nitrogen to dissolve the sodium blocks, adding into the reaction solution, reacting at 80 ℃ under the protection of nitrogen for 72h, cooling, carrying out rotary evaporation concentration, adding 400mL water, stirring fully, adjusting the pH to 8 with concentrated hydrochloric acid, carrying out suction filtration, washing with water (200mL × 4), discarding filter cakes, combining filtrates, adjusting the pH to 5 with concentrated hydrochloric acid, extracting with ethyl acetate (50mL × 20), washing with saturated water, drying with anhydrous sodium sulfate to obtain a light yellow solid, adding 40mL methanol, stirring for 20min, drying to obtain a white solid, and obtaining a yield of 44.7%.1H NMR(DMSO-d6,300MHz)(ppm):12.32(s,1H),11.50(s,1H),8.48(d,J=8.7Hz,1H),8.35(t,J=7.5Hz,1H),8.19(d,J=9.0Hz,1H).
Preparation of 2, 4-dichloro-6- (6- (trifluoromethyl) pyridin-2-yl) -1,3, 5-triazine (5)
A100 mL dry round bottom flask was charged with intermediate 4(2g, 7.8mmol) and phosphorus oxychloride (20mL) separately and reacted at 110 ℃ for 48h under nitrogen. After cooling, 500mL of ice water was slowly added dropwise with stirring, and the mixture was extracted with dichloromethane (50 mL. times.12), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a red oil which was directly used in the next reaction.
Preparation of 4-chloro-6- (6- (trifluoromethyl) pyridin-2-yl) -N- (2- (trifluoromethyl) pyridin-4-yl) -1,3, 5-triazin-2-amine (6-1)
To 100mL dryThe intermediate 5 obtained in the previous step, 2-trifluoromethyl-4-aminopyridine (660mg, 4.1mmol), N-diisopropylethylamine (1.1g, 8.1mmol) and anhydrous tetrahydrofuran (40mL) were added to a dry round-bottom flask, reacted at 70 ℃ for 12 hours, cooled, diluted with 100mL of water, extracted with ethyl acetate (50mL × 3), washed with saturated brine, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and purified with silica gel column (petroleum ether: ethyl acetate: 25: 1) to obtain 0.9g of a pale yellow solid.1H NMR(DMSO-d6,300MHz)(ppm):11.66(s,1H),8.63-8.79(m,3H),8.37(t,J=7.8Hz,1H),8.19(d,J=6.3Hz,1H),7.93(s,1H).
Preparation of 2- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((2- (trifluoromethyl) pyridin-4-yl) amino) -1,3, 5-triazin-2-yl) amino-1, 3-propanediol (I-1)
Adding intermediate 6-1(100mg, 0.2mmol), serinol (50mg, 0.5mmol) and tetrahydrofuran (15mL) into a 50mL dry round-bottom flask respectively, reacting at 70 ℃ for 12H, cooling, adding 100mL water for dilution, extracting with ethyl acetate (50mL × 3), washing with saturated saline, drying with anhydrous sodium sulfate, and concentrating under reduced pressure to obtain a pale yellow solid 110mg, yield 97.4%, mp199-201 ℃ and ES-MS:476.2[ M + H ], (M + H)]+;1H NMR(DMSO-d6,300MHz)(ppm):10.67(s,1H),8.54-8.68(m,3H),8.33(t,J=7.5Hz,1H),8.11(d,J=6.2Hz,1H),7.78-8.03(m,2H),4.76(dt,J=21.2,5.9Hz,2H),4.05-4.31(m,1H),3.55-3.69(m,4H).
Example 2
Preparation of 3- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((2- (trifluoromethyl) pyridin-4-yl) amino) -1,3, 5-triazin-2-yl) amino-1, 2-propanediol (I-2)
Referring to the synthesis of I-1, the intermediate 6-1 was reacted with 3-amino-1, 2-propanediol to obtain a pale yellow solid with a yield of 88.5%, mp 131-. ES-MS 476.2[ M + H ]]+;1H NMR(DMSO-d6,300MHz)(ppm):10.59(s,1H),8.36-8.60(m,3H),8.23(t,J=8.6Hz,1H),8.12(d,J=20.7Hz,1H),7.98-8.03(m,2H),4.79(dd,J=23.9,6.2Hz,1H),4.57(t,J=4.5Hz,1H),3.64-3.69(m,1H),3.49(dd,J=11.7,6.5Hz,2H),3.33(dd,J=5.6,2.8Hz,2H).
Example 3
Preparation of (S) -3- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((2- (trifluoromethyl) pyridin-4-yl) amino) -1,3, 5-triazin-2-yl) amino-1, 2-propanediol (I-3)
Referring to the synthesis of I-1, a white solid was obtained from the reaction of intermediate 6-1 with (S) -3-amino-1, 2-propanediol in 88.5% yield, mp 213-. ES-MS 476.2[ M + H ]]+;1H NMR(DMSO-d6,300MHz)(ppm):10.59(s,1H),8.37-8.60(m,3H),8.23(t,J=9.2Hz,1H),8.12(d,J=21.1Hz,1H),7.98-8.04(m,2H),4.74-4.84(dd,J=23.7,6.0Hz,1H),4.56(t,J=6.1Hz,1H),3.64-3.69(m,1H),3.49(dd,J=14.9,5.8Hz,2H),3.33(dd,J=5.7,3.3Hz,2H).
Example 4
Preparation of (R) -3- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((2- (trifluoromethyl) pyridin-4-yl) amino) -1,3, 5-triazin-2-yl) amino-1, 2-propanediol (I-4)
Referring to the synthesis of I-1, a white solid was obtained from the reaction of intermediate 6-1 with (R) -3-amino-1, 2-propanediol in 88.5% yield, mp 211-. ES-MS 476.2[ M + H ]]+;1H NMR(DMSO-d6,300MHz)(ppm):10.59(s,1H),8.37-8.60(m,3H),8.23(t,J=9.2Hz,1H),8.12(d,J=21.3Hz,1H),7.98-8.03(m,2H),4.79(dd,J=23.8,6.1Hz,1H),4.57(t,J=6.3Hz,1H),3.62-3.72(m,1H),3.49(dd,J=14.7,6.2Hz,2H),3.33(dd,J=5.9,3.1Hz,2H).
Example 5
Preparation of (L) -N- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((2- (trifluoromethyl) pyridin-4-yl) amino) -1,3, 5-triazin-2-yl) serine (I-5)
Adding the intermediate 6-1(420mg, 1mmol), (L) -serine methyl ester hydrochloride (330mg, 2.1mmol), triethylamine (320mg, 3.1mmol) and tetrahydrofuran (20mL) into a 50mL dry round-bottom flask respectively, reacting for 12H at 70 ℃, adding 1mol/L sodium hydroxide solution (2.5mL), continuing to react for 12H at 70 ℃, cooling, adding 100mL water for dilution, adjusting the pH to 9 with 1mol/L sodium hydroxide solution, extracting impurities from ethyl acetate (50mL × 2), adjusting the pH of the aqueous phase to 5 with concentrated hydrochloric acid, extracting with ethyl acetate (50mL × 3), washing with saturated brine, drying with anhydrous sodium sulfate, concentrating under reduced pressure to obtain 180mg of white solid with the yield of 36.8%, and obtaining an mp 207 ℃. (ES-MS): 490.2[ M + H ], (209 ℃)]+;1H NMR(DMSO-d6,300MHz)(ppm):12.79(s,1H),10.75(s,1H),8.53-8.62(m,2H),8.29-8.43(m,3H),8.12(dd,J=6.1,3.1Hz,1H),8.02(d,J=3.4Hz,1H),4.62(dd,J=12.6,3.1Hz,1H),4.54(t,J=3.2Hz,1H),3.88(d,J=2.9Hz,2H).
Example 6
Preparation of sodium (L) -N- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((2- (trifluoromethyl) pyridin-4-yl) amino) -1,3, 5-triazin-2-yl) serine salt (I-6)
To a 50mL dry round-bottom flask were added compound I-5(150mg, 0.3mmol), solid sodium hydroxide (10mg, 0.25mmol) and methanol (10mL), respectively, and reacted at room temperature for 12 h. Concentrating under reduced pressure to precipitate a small amount of white solid, filtering, and drying to obtain 20mg of white solid with the yield of 12.8 percent and the temperature of mp 221-. ES-MS 512.2[ M + H ]]+;1H NMR(DMSO-d6,300MHz)(ppm):10.75(s,1H),8.53-8.62(m,2H),8.29-8.43(m,3H),8.12(dd,J=5.8,2.9Hz,1H),8.02(d,J=3.1Hz,1H),4.62(dd,J=12.2,3.2Hz,1H),4.54(t,J=3.3Hz,1H),3.88(d,J=3.0Hz,2H).
Example 7
Preparation of 4-chloro-6- (6- (trifluoromethyl) pyridin-2-yl) -N- (3- (trifluoromethyl) phenyl) -1,3, 5-triazin-2-amine (6-2)
Referring to the synthesis of intermediate 6-1, a white solid was prepared from intermediate 5 by reaction with 3-trifluoromethylaniline.1HNMR(DMSO-d6,300MHz)(ppm):11.33(s,1H),8.62-8.69(m,2H),8.36(t,J=8.8Hz,1H),8.19(d,J=9.1Hz,1H),7.96(dd,J=27.2,8.9Hz,1H),7.48-7.64(m,1H).
Preparation of 2- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((3- (trifluoromethyl) phenyl) amino) -1,3, 5-triazin-2-yl) amino-1, 3-propanediol (I-7)
Referring to the synthesis of I-1, a white solid was prepared from intermediate 6-2 by reaction with serinol in 97.4% yield, mp161-163 ℃. ES-MS 475.2[ M + H ]]+;1H NMR(DMSO-d6,300MHz)(ppm):10.21(s,1H),8.48-8.65(m,2H),8.30(t,J=7.5Hz,1H),8.05(dd,J=20.8,8.9Hz,2H),7.45-7.73(m,2H),7.34(d,J=6.2Hz,1H),4.72(dt,J=27.1,6.3Hz,2H),4.05-4.26(m,1H),3.61(t,J=4.5Hz,4H).
Example 8
Preparation of 3- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((3- (trifluoromethyl) phenyl) amino) -1,3, 5-triazin-2-yl) amino-1, 2-propanediol (I-8)
Referring to the synthesis of I-1, a white solid was obtained in 88.5% yield from intermediate 6-2 by reaction with 3-amino-1, 2-propanediol, mp 193-. ES-MS 475.2[ M + H ]]+;1H NMR(DMSO-d6,300MHz)(ppm):10.24(s,1H),8.49-8.66(m,2H),8.30(t,J=7.5Hz,1H),8.05-8.21(m,2H),7.51-7.93(m,2H),7.34(d,J=9.4Hz,1H),4.85(dd,J=29.6,6.1Hz,1H),4.64(dt,J=21.2,6.3Hz,1H),3.70-3.78(m,1H),3.39-3.63(m,4H).
Example 9
Preparation of (S) -3- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((3- (trifluoromethyl) phenyl) amino) -1,3, 5-triazin-2-yl) amino-1, 2-propanediol (I-9)
Referring to the synthesis of I-1, a white solid was obtained from the reaction of intermediate 6-2 with (S) -3-amino-1, 2-propanediol in 88.5% yield, mp 211-. ES-MS 475.2[ M + H ]]+;1H NMR(DMSO-d6,300MHz)(ppm):10.24(s,1H),8.49-8.66(m,2H),8.30(t,J=7.5Hz,1H),8.05-8.21(m,2H),7.51-7.94(m,2H),7.34(d,J=6.4Hz,1H),4.86(dd,J=30.1,6.2Hz,1H),4.64(dt,J=21.2,6.1Hz,1H),3.71-3.78(m,1H),3.39-3.64(m,4H).
Example 10
Preparation of (R) -3- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((3- (trifluoromethyl) phenyl) amino) -1,3, 5-triazin-2-yl) amino-1, 2-propanediol (I-10)
Referring to the synthesis of I-1, a white solid was obtained in 88.5% yield from intermediate 6-2 reacted with (R) -3-amino-1, 2-propanediol at 209 ℃ under mp 207-. ES-MS 475.2[ M + H ]]+;1H NMR(DMSO-d6,300MHz)(ppm):10.24(s,1H),8.48-8.66(m,2H),8.30(t,J=8.9Hz,1H),8.05-8.21(m,2H),7.51-7.94(m,2H),7.34(d,J=6.2Hz,1H),4.85(dd,J=30.1,6.2Hz,1H),4.64(dt,J=20.9,5.9Hz,1H),3.70-3.77(m,1H),3.39-3.64(m,4H).
Example 11
Preparation of 4-chloro-6- (6- (trifluoromethyl) pyridin-2-yl) -N- (6-chloropyridin-2-yl) -1,3, 5-triazin-2-amine (6-3)
Referring to the synthesis of intermediate 6-1, a pale cyan solid was prepared from intermediate 5 by reaction with 2-amino-6-chloropyridine.1HNMR(DMSO-d6,300MHz)(ppm):11.05(s,1H),8.54(d,J=9.2Hz,1H),8.41(t,J=7.5Hz,1H),8.22(t,J=6.1Hz,2H),7.94(t,J=7.4Hz,1H),7.28(d,J=9.2Hz,1H).
Preparation of 2- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((6-chloropyridin-2-yl) amino) -1,3, 5-triazin-2-yl) amino-1, 3-propanediol (I-11)
Referring to the synthesis of I-1, a white solid was prepared from intermediate 6-3 by reaction with serinol in 97.4% yield, mp221-223 ℃. ES-MS 440.1[ M-H ]]+;1H NMR(DMSO-d6,300MHz)(ppm):10.36(s,1H),8.57-8.62(m,1H),8.28-8.39(m,2H),8.10(d,J=8.9Hz,1H),7.63-7.92(m,2H),7.12-7.16(m,1H),4.74(dt,J=24.2,6.1Hz,2H),4.03-4.23(m,1H),3.51-3.64(m,4H).
Example 12
Preparation of 3- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((6-chloropyridin-2-yl) amino) -1,3, 5-triazin-2-yl) amino-1, 2-propanediol (I-12)
Referring to the synthesis of I-1, the intermediate 6-3 was reacted with 3-amino-1, 2-propanediol to obtain a pale yellow solid with a yield of 97.4% and mp 111-. ES-MS 440.1[ M-H ]]+;1H NMR(DMSO-d6,300MHz)(ppm):10.38(s,1H),8.41-8.61(m,2H),8.06-8.29(m,3H),7.77-7.91(m,1H),7.12(d,J=9.3Hz,1H),4.63-4.93(m,2H),3.26-3.72(m,5H).
Example 13
Preparation of (S) -3- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((6-chloropyridin-2-yl) amino) -1,3, 5-triazin-2-yl) amino-1, 2-propanediol (I-13)
Referring to the synthesis of I-1, the intermediate 6-3 was reacted with (S) -3-amino-1, 2-propanediol to obtain a pale yellow solid with a yield of 97.4%, mp 181-. ES-MS 440.1[ M-H ]]+;1H NMR(DMSO-d6,300MHz)(ppm):10.38(s,1H),8.42-8.62(m,2H),8.06-8.31(m,3H),7.77-7.91(m,1H),7.13(d,J=9.1Hz,1H),4.63-4.92(m,2H),3.23-3.73(m,5H).
Example 14
Preparation of (R) -3- ((4- (6- (trifluoromethyl) pyridin-2-yl) -6- ((6-chloropyridin-2-yl) amino) -1,3, 5-triazin-2-yl) amino-1, 2-propanediol (I-14)
Referring to the synthesis of I-1, the intermediate 6-3 was reacted with (R) -3-amino-1, 2-propanediol to obtain a pale yellow solid with a yield of 97.4% and mp 179. sup. 181 ℃. ES-MS 440.1[ M-H ]]+;1H NMR(DMSO-d6,300MHz)(ppm):10.39(s,1H),8.42-8.62(m,2H),8.06-8.31(m,3H),7.77-7.93(m,1H),7.12(dd,J=9.1,3.1Hz,1H),4.89(dd,J=33.1,6.2Hz,1H),4.69(dt,J=18.1,6.0Hz,1H),3.22-3.78(m,5H).
Example 15
Pharmacological experiment 1: compound pair IDH2R140QMeasurement of inhibitory Activity
IDH2R140QThe mutant can catalyze the conversion of α -KG into 2-HG and simultaneously oxidize NADPH into NADP+Therefore, IDH2 can be evaluated by measuring the consumption value of NADPH by the compound of the present inventionR140QInhibitory activity of the mutant. The positive drug is AG-221.
The experimental method comprises the following steps: a96-well plate was added with a solution containing 25mM Tris (pH7.4), 150mM NaCl, 10mM MgCl20.03% BSA, followed by addition of a concentration gradient of the compound, and the substrate for the enzymatic reaction, 1mM α -KG and 100. mu. MNADPH, and finally addition of IDH2 at a concentration of 0.5. mu.g/mLR140QThe total volume was 200. mu.L. Continuous assay at 340nM wavelength at room temperature using Thermo enzyme readerThe absorbance change of NADPH was measured. Calculation of Compound Pair IDH2 at different time points of incubation based on NADPH consumptionR140QAnd IC was calculated by GraphPad Prism 5 software50The value is obtained. The test data are shown in Table 1.
Pharmacological experiment 2: compound pair IDH2WTMeasurement of inhibitory Activity
Wild type IDH2 in NADP+Under the assistance of the catalytic action, α -KG is generated by isocitrate, and NADPH is generated, so that the inhibitory activity of the compound on wild-type IDH2 can be detected by detecting the increase of NADPH.
The experimental method comprises the following steps: add 50mM Tris (pH7.4), 5mM MgCl to 96-well plates20.03% BSA, then 200. mu.M ICT and 200. mu.M NADP as substrate for the enzymatic reaction, and finally 0.3. mu.g/mL IDH2 in a total volume of 200. mu.L. The change in absorbance of NADPH was continuously measured at 340nM wavelength at room temperature using a Thermo microplate reader. Inhibition of IDH2 activity by compounds at various time points of incubation was calculated from the increase in NADPH and IC was calculated by GraphPad Prism 5 software50The value is obtained. The results are shown in Table 1.
Compounds IC in Table 150Range of values: a represents 1-300 nM, B represents 300-1000 nM, and C represents 1000nM or more.
TABLE 1 inhibitory Activity and Selectivity of the Compounds of the invention against IDH2
The experimental result shows that the compound of the invention can effectively inhibit IDH2 at very low concentrationR140QThe activity of the process. IC of which I-7, I-8, I-9 and I-1050Is obviously superior to a positive control drug AG-221, in particular to I-10 to IDH2R140QThe inhibiting activity of the compound is 58 times higher than that of AG-221. More rarely, the compound of the invention has weak inhibitory effect on the activity of wild type IDH2, shows extremely high selectivity and is remarkably superior to the positive drug AG-221.
Pharmacological experiment 3: compound (I)For IDH2R140QMutant TF-1 cell 2-HG inhibitory Activity assay
Use carrying IDH2R140QMutated TF-1 cells were evaluated for the inhibitory activity of compounds against 2-HG in tumor cells. TF-1_ IDH2R140QCells were purchased from ATCC. The intracellular 2-HG content was determined using LC-MS/MS. The positive control drug was AG-221.
The experimental method comprises the following steps: TF-1_ IDH2R140QThe cells are 5 × 104The cells were collected by seeding in 12-well plates at a density of/mL and incubating for 72 hours with a gradient of test compound concentration. The cell pellet was suspended in 100. mu.L of 80% aqueous methanol, centrifuged at 13000rpm for 10min to remove the pellet at 4 ℃ and stored in a freezer at-80 ℃. Before injection, the sample is centrifuged again at 13000rmp for 10min, and the supernatant (about 100. mu.L) is transferred to the liquid phase inner insert tube for injection. The standard substance is D-2-hydroxy glutaric acid disodium salt (Sigma-Aldrich)#H8378) The concentrations were set at 13.6ng/mL,68ng/mL,340ng/mL, 1.70. mu.g/mL, 8.48. mu.g/mL, and 42.4. mu.g/mL. The test instrument is Q activeTMPlus-OrbitrapTMMS, Thermo Scientific bench quadrupole-orbitrap high resolution mass spectrometer. The chromatographic column is WaterTMACQUITYTMUPLC HSS-T3-1.8 μm,21mm × 100mm, Part No186003539, mobile Phase aqueous formic acid (chromatographically pure), Phase B acetonitrile (chromatographically pure).
TABLE 2 LC-MS elution conditions
Time | Flow[mL/min] | %B |
0.000 | Run | |
0.000 | 0.200 | 1% |
1.500 | 0.200 | 1% |
2.000 | 0.200 | 70% |
3.500 | 0.200 | 70% |
3.600 | 0.200 | 1% |
5.000 | 0.200 | 1% |
5.000 | Stop Run |
IC calculation by GraphPad Prism 5 software50The value is obtained. Partial compound pair TF-1_ IDH2 of the present inventionR140QThe inhibitory activity at intracellular 2-HG levels is shown in Table 3.
TABLE 3 Compound of the invention vs TF-1_ IDH2R140QCell 2-HGInhibiting activity of
Compound numbering | IC50(nM) |
AG-221 | 25.0 |
I-7 | 70.6 |
I-9 | 198.6 |
I-10 | 13.2 |
Experimental results show that the compound can effectively inhibit TF-1_ IDH2 at nM concentrationR140QThe activity of the compound I-10 is obviously better than that of a positive drug AG-221 at the level of 2-HG of cells.
Pharmacological experiment 4: compound in vitro liver microsome metabolic stability test
The in vitro metabolic stability of some of the compounds of the invention was evaluated using a liver microsome assay. Metabolic stability the in vitro elimination half-life (T) was calculated by analyzing the change in the residual substrate concentration level of the compound over time by the substrate elimination method1/2)。
Experimental materials:
name (R) | Suppliers of goods | Goods number | Batch number |
Human liver microsomes | IVT | X008070 | IQF |
Mouse liver microsome | XENOTECH | M1000 | 1610148 |
The test compound was incubated with human and murine liver microsomes in a total system volume of 100. mu.L. mu.L of the compound (final concentration 1. mu.M), 50. mu.L of liver microsomes (final concentration 0.5mg/mL), 40. mu.L of NADPH reaction system were added. Incubation is carried out in a constant temperature oscillating water bath (37 ℃) at medium temperature, 300 mu L of stop solution (acetonitrile solution with the final concentration of 100ng/mL tolbutamide) is added when the incubation time is 0, 5, 10, 20, 30 and 60min respectively, and the reaction is stopped. The tube was kept open throughout the experiment to ensure oxygen participation. After the reaction was terminated, the sample was centrifuged at 13000 Xg for 10min (4 ℃ C.), 5. mu.L of the supernatant was collected, and LC-MS/MS analysis was performed to analyze the remaining amount of the compound, and 3 times of experiments were performed in parallel to investigate the metabolic stability of the compound. Specific test data are shown in table 4.
TABLE 4 in vitro liver microsomal enzyme metabolic stability test data for compounds of the invention
The experimental result shows that the compound I-10 of the invention shows excellent in vitro metabolic stability which is obviously superior to 3 positive control drugs.
Pharmacological experiment 5: compound pair IDH2R140QEffect of mutated TF-1 cell proliferation and differentiation
Dependence of different TF-1 cell lines on GM-CSF: TF-1 cell lines depend on GM-CSF for proliferation. TF-1_ IDH2 is first putWT、TF-1_IDH2R140QThe cells were cultured in whole medium, GM-CSF was removed from the medium before measuring proliferation potency, cultured for 3 days, and TF-1_ IDH2 was examined by MTT methodWTAnd TF-1_ IDH2R140QThe proliferation potency of the transfected cell line was determined by the GM-CSF-dependent change in the transfected cell line.
Effect of compounds on cell proliferation: TF-1_ IDH2WTAnd TF-1_ IDH2R140QCells were incubated with a concentration of compound for 7 days, and the MTT method was used to determine the change in proliferative capacity of both cells in the presence and absence of the compound.
Effect of compounds on cell differentiation: TF-1_ IDH2WTAnd TF-1_ IDH2R140QThe cells are respectively incubated with a compound with a certain concentration for 7 days, GM-CSF in a culture medium is removed, Erythropoietin (EPO) is added to induce the cells to differentiate for 7 days, the cells are collected by centrifugation, PBS is used for washing, and the color change generated by cell heme is observed and photographed as an index of cell differentiation.
The test results show that the tested compound I-10 can obviously inhibit GM-CSF independent TF-1_ IDH2 at the concentration of 1 mu MR140QProliferation of cells and significant induction of TF-1_ IDH2R140QRe-differentiation of the cells.
Using the same procedure, the present invention also tested compound I-10 versus other IDH 2-carrying compoundsR140QThe proliferation inhibitory activity of the mutant tumor cell lines includes colon cancer HCT116 cells and glioblastoma U87 cells. The results showed that Compound I-10 was conjugated to HCT116_ IDH2R140QAnd U87_ IDH2R140QThe cell proliferation has inhibitory effect.
Pharmacological experiment 6: TF-1_ IDH2R140QEvaluation of drug efficacy of mutant cell subcutaneous transplantation tumor in vivo
Evaluation of in vivo 2-HG inhibitory Activity of Compound I-10 TF-1_ IDH 2-containingR140QNCG mice with mutant cell subcutaneous transplantation tumorThe process is carried out. NCG mice were purchased at the university of south kyo animal model research institute.
Amplification of TF-1_ IDH2R140QCells, logarithmic growth phase of tumor cells for in vivo tumor inoculation according to 5 × 106The cell amount/mouse is respectively inoculated to the lower part of the lower back of the right side of the body of the NCG mouse irradiated by sublethal dose, and the GM-CSF is intraperitoneally administered twice a day to a human until the tumor volume reaches 200mm3Groups were made at time and human GM-CSF was administered continuously. The control group without mutation was inoculated with TF-1 cell line, and the control group with the compound and a solvent (2% absolute ethanol: 10% Solutol: 88% physiological saline (v/v/v)) was inoculated with TF-1_ IDH2R140QAnd (4) inoculating. The compound solutions of the corresponding concentrations were administered to each group by gavage in a volume of 100. mu.L/10 g body weight, and the control group was administered the same volume of the blank vehicle. Mice were sacrificed 10 days after administration, tumors were detached, homogenized, and the 2-HG content in the tumors was examined. See example 17 Experimental methods section for LC-MS/MS analysis conditions. The 2-HG concentration of each animal tumor homogenate in each group was calculated as a percentage (2-HG%) by LC-MS/MS.
The relative percentage of 2-HG in the mouse tumor (mean) after administration of the test compound is shown in Table 5. The results showed that Compound I-10 was present against TF-1_ IDH2R140QThe inhibitory activity of 2-HG in the cell subcutaneous transplantation tumor is stronger than that of AG-221.
TABLE 5 intratumoral 2-HG% after 10 days of administration
Group of | Dosage (mg/kg) | 2-HG% |
TF-1 control group | - | 0 |
TF-1_IDH2R140QControl group | - | 100 |
AG-221 | 7.5 | 50.3 |
AG-221 | 15 | 9.7 |
AG-221 | 30 | -4.6 |
I-10 | 7.5 | 60.5 |
I-10 | 15 | 5.2 |
I-10 | 30 | -8.6 |
Claims (10)
1. An s-triazine compound shown in a general formula I or a pharmaceutically acceptable salt thereof:
wherein:
T1and T2Independently selected from N or C; when T is1When is N, T2Is selected from C; when T is1When is C, T2Selected from C or N;
r is selected from hydrogen, halogen, hydroxyl, amido and C1-C4Alkyl or C1-C4Alkoxy, which alkyl may be optionally mono-or poly-substituted with halogen;
Y1、Y2and Y3Each independently selected from hydrogen and C1-C4Alkyl, hydroxy C1-C4Alkyl or carboxyl, which alkyl may optionally be mono-or polysubstituted by the following identical or different substituents: hydrogen, hydroxy, C1-C4Alkyl or hydroxy C1-C4An alkyl group.
2. The s-triazine compound or the pharmaceutically acceptable salt thereof according to claim 1, wherein R is selected from halogen or C1-C4An alkyl group, which alkyl group may optionally be mono-or poly-substituted with halogen.
3. The s-triazine compound or the pharmaceutically acceptable salt thereof according to claim 1, wherein R is selected from chlorine or trifluoromethyl.
4. The s-triazine compound or the pharmaceutically acceptable salt thereof according to claim 1, wherein Y is1、Y2And Y3Each independently selected from hydrogen, hydroxymethyl, 1, 2-dihydroxyethyl, 2, 3-dihydroxypropyl or carboxyl.
6. the process for preparing the s-triazine compound according to any one of claims 1 to 5, which comprises the steps of: 2-bromo-6-trifluoromethylpyridine and cuprous cyanide undergo nucleophilic substitution reaction to prepare cyanide 1,1 is hydrolyzed under the condition of concentrated hydrochloric acid to generate carboxylic acid 2, 2 and thionyl chloride are prepared into acyl chloride, and then the carboxylic acid and methanol react to prepare methyl ester 3; 3 and biuret ring to obtain an intermediate 4; 4 refluxing with phosphorus oxychloride to obtain chloride 5, and reacting with different substituted aminopyridine or aromatic amine to obtain an intermediate 6; 6 with fatty amines (Y)1Y2Y3CNH2) The compound with the general formula I is prepared by the following reaction route:
7. a pharmaceutical composition, which comprises the s-triazine compound and/or pharmaceutically acceptable salt thereof according to any one of claims 1 to 5, and pharmaceutically acceptable auxiliary materials.
8. Use of the s-triazine compound of any one of claims 1 to 5 and/or a pharmaceutically acceptable salt thereof for preparing an IDH2 mutant inhibitor.
9. Use of the s-triazine compound of any one of claims 1 to 5 and/or a pharmaceutically acceptable salt thereof for the preparation of a medicament for treating a cancer carrying an IDH2 mutation.
10. Use of the s-triazine compound of any one of claims 1 to 5 and/or a pharmaceutically acceptable salt thereof in combination with one or more chemotherapeutic agents, targeted antineoplastic agents, immune checkpoint inhibitors, immune checkpoint agonists, antineoplastic vaccines, antiviral agents, antiviral vaccines for the manufacture of a medicament for the treatment of a cancer carrying the IDH2 mutation.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201910174468.5A CN111662271B (en) | 2019-03-08 | 2019-03-08 | Compound with IDH mutant inhibitory activity and preparation method and application thereof |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201910174468.5A CN111662271B (en) | 2019-03-08 | 2019-03-08 | Compound with IDH mutant inhibitory activity and preparation method and application thereof |
Publications (2)
Publication Number | Publication Date |
---|---|
CN111662271A true CN111662271A (en) | 2020-09-15 |
CN111662271B CN111662271B (en) | 2023-11-14 |
Family
ID=72381304
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN201910174468.5A Active CN111662271B (en) | 2019-03-08 | 2019-03-08 | Compound with IDH mutant inhibitory activity and preparation method and application thereof |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN111662271B (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN114292618A (en) * | 2022-01-13 | 2022-04-08 | 泰州亚德胶粘制品有限公司 | UV (ultraviolet) blocking optical adhesive and preparation method thereof |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN104114543A (en) * | 2012-01-06 | 2014-10-22 | 安吉奥斯医药品有限公司 | Therapeutically active compounds and their methods of use |
CN105473560A (en) * | 2013-07-11 | 2016-04-06 | 安吉奥斯医药品有限公司 | Therapeutically active compounds and use methods thereof |
CN107207469A (en) * | 2015-10-21 | 2017-09-26 | 纽弗姆制药有限公司 | For treat the deuterated compound of hematologic malignancies with and combinations thereof and method |
-
2019
- 2019-03-08 CN CN201910174468.5A patent/CN111662271B/en active Active
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN104114543A (en) * | 2012-01-06 | 2014-10-22 | 安吉奥斯医药品有限公司 | Therapeutically active compounds and their methods of use |
CN105473560A (en) * | 2013-07-11 | 2016-04-06 | 安吉奥斯医药品有限公司 | Therapeutically active compounds and use methods thereof |
CN107207469A (en) * | 2015-10-21 | 2017-09-26 | 纽弗姆制药有限公司 | For treat the deuterated compound of hematologic malignancies with and combinations thereof and method |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN114292618A (en) * | 2022-01-13 | 2022-04-08 | 泰州亚德胶粘制品有限公司 | UV (ultraviolet) blocking optical adhesive and preparation method thereof |
Also Published As
Publication number | Publication date |
---|---|
CN111662271B (en) | 2023-11-14 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN109952303B (en) | TYK2 inhibitor and application thereof | |
TW202144345A (en) | Kras mutant protein inhibitors | |
TWI527800B (en) | 1-(arylmethyl)quinazoline-2,4(1h,3h)-diones as parp inhibitors and the use thereof | |
EP3398950B1 (en) | Novel kinase inhibitor against wild-type egfr and mutated egfr | |
EP2896620A1 (en) | Alkynyl heteroaromatic ring compound and application thereof | |
CN102887895B (en) | Pyridopyrimidine class mTOR inhibitors | |
CN114057771B (en) | Macrocyclic compounds, their preparation and use | |
CN103421005A (en) | Acetylene derivative capable of resisting activity of tumor | |
CN110563703B (en) | Compound for inducing PARP-1 degradation based on CRBN ligand, preparation method and application | |
CN112979679B (en) | Fluoroheterocyclic derivative having macrocyclic structure and use thereof | |
CN116283953B (en) | Indoline compound containing thiazole structure, and preparation method and application thereof | |
CN112194659A (en) | Alkyne derivative and preparation method and application thereof | |
Hao et al. | Design, synthesis and pharmacological evaluation of a novel mTOR-targeted anti-EV71 agent | |
CN111662271B (en) | Compound with IDH mutant inhibitory activity and preparation method and application thereof | |
CN117105936A (en) | Imidazo [1,2-a ] pyridine compound serving as FLT3 inhibitor, and preparation method and application thereof | |
Liu et al. | Design, synthesis, and bioactivity study on Lissodendrins B derivatives as PARP1 inhibitor | |
Min et al. | Discovery of potent and selective c-Met inhibitors for MET-amplified hepatocellular carcinoma treatment | |
TWI546304B (en) | Protein tyrosine kinase inhibitors and their use | |
CN116249531A (en) | Compositions and methods for activating pyruvate kinase | |
CN114315837A (en) | Crystalline form of ERK inhibitor and preparation method thereof | |
TW201738227A (en) | Fused ring compound and preparation method, application and intermediate compound thereof | |
US11021479B2 (en) | Pyridoquinazoline derivatives useful as protein kinase inhibitors | |
AU2021226411A1 (en) | Pyridopyrimidine derivatives useful in modulation of AhR signalling | |
CN106674200B (en) | A kind of compound and its preparation method and application containing L- prolineamide segment | |
CN115073361B (en) | Rosin diterpenoid compound and preparation method and application thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination | ||
GR01 | Patent grant | ||
GR01 | Patent grant |