CN118005654A - Substituted pyridopyrimidinone compounds, compositions containing the same and uses thereof - Google Patents
Substituted pyridopyrimidinone compounds, compositions containing the same and uses thereof Download PDFInfo
- Publication number
- CN118005654A CN118005654A CN202310418144.8A CN202310418144A CN118005654A CN 118005654 A CN118005654 A CN 118005654A CN 202310418144 A CN202310418144 A CN 202310418144A CN 118005654 A CN118005654 A CN 118005654A
- Authority
- CN
- China
- Prior art keywords
- compound
- compounds
- mmol
- pharmaceutically acceptable
- deuterium
- 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.)
- Pending
Links
- 239000000203 mixture Chemical class 0.000 title abstract description 37
- IAAQUOVTPAMQCR-UHFFFAOYSA-N 1h-pyrido[3,2-d]pyrimidin-2-one Chemical class C1=CC=C2NC(=O)N=CC2=N1 IAAQUOVTPAMQCR-UHFFFAOYSA-N 0.000 title description 2
- 150000001875 compounds Chemical class 0.000 claims abstract description 194
- -1 pyridopyrimidinone compound Chemical class 0.000 claims abstract description 50
- 208000002320 spinal muscular atrophy Diseases 0.000 claims abstract description 50
- 229940002612 prodrug Drugs 0.000 claims abstract description 39
- 239000000651 prodrug Substances 0.000 claims abstract description 39
- 150000003839 salts Chemical class 0.000 claims abstract description 35
- 239000013078 crystal Substances 0.000 claims abstract description 18
- 238000011282 treatment Methods 0.000 claims abstract description 11
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 45
- 239000012453 solvate Substances 0.000 claims description 34
- 229910052805 deuterium Inorganic materials 0.000 claims description 33
- 229910052739 hydrogen Inorganic materials 0.000 claims description 33
- 239000001257 hydrogen Substances 0.000 claims description 33
- YZCKVEUIGOORGS-OUBTZVSYSA-N Deuterium Chemical compound [2H] YZCKVEUIGOORGS-OUBTZVSYSA-N 0.000 claims description 28
- 201000010099 disease Diseases 0.000 claims description 26
- 239000003814 drug Substances 0.000 claims description 25
- 239000008194 pharmaceutical composition Substances 0.000 claims description 21
- 150000002431 hydrogen Chemical class 0.000 claims description 19
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 claims description 13
- 229910052736 halogen Inorganic materials 0.000 claims description 12
- 150000002367 halogens Chemical class 0.000 claims description 12
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 claims description 12
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 11
- 150000001975 deuterium Chemical group 0.000 claims description 5
- 238000004519 manufacturing process Methods 0.000 claims description 3
- 230000002265 prevention Effects 0.000 claims 1
- 239000002904 solvent Substances 0.000 abstract description 26
- 108090000623 proteins and genes Proteins 0.000 abstract description 10
- 102000047499 Survival of Motor Neuron 2 Human genes 0.000 abstract description 7
- 108700024745 Survival of Motor Neuron 2 Proteins 0.000 abstract description 7
- 238000006243 chemical reaction Methods 0.000 description 52
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical group N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 34
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 32
- 235000002639 sodium chloride Nutrition 0.000 description 32
- 239000000243 solution Substances 0.000 description 28
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 26
- 238000003786 synthesis reaction Methods 0.000 description 24
- 230000015572 biosynthetic process Effects 0.000 description 23
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 22
- 238000004809 thin layer chromatography Methods 0.000 description 22
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 20
- 239000007787 solid Substances 0.000 description 20
- 208000035475 disorder Diseases 0.000 description 19
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 19
- 125000004431 deuterium atom Chemical group 0.000 description 18
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-Dimethylformamide Chemical compound CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 17
- 229910052757 nitrogen Inorganic materials 0.000 description 17
- 239000000543 intermediate Substances 0.000 description 15
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 14
- 102100021947 Survival motor neuron protein Human genes 0.000 description 14
- 238000000034 method Methods 0.000 description 14
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 13
- 229940079593 drug Drugs 0.000 description 13
- 208000024891 symptom Diseases 0.000 description 13
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 12
- 239000004480 active ingredient Substances 0.000 description 12
- 238000010898 silica gel chromatography Methods 0.000 description 12
- 239000012074 organic phase Substances 0.000 description 11
- 241000700159 Rattus Species 0.000 description 10
- 238000002360 preparation method Methods 0.000 description 10
- 229940124597 therapeutic agent Drugs 0.000 description 10
- 210000004369 blood Anatomy 0.000 description 9
- 239000008280 blood Substances 0.000 description 9
- 150000004677 hydrates Chemical class 0.000 description 9
- 210000001853 liver microsome Anatomy 0.000 description 9
- SCVFZCLFOSHCOH-UHFFFAOYSA-M potassium acetate Chemical compound [K+].CC([O-])=O SCVFZCLFOSHCOH-UHFFFAOYSA-M 0.000 description 9
- 238000009472 formulation Methods 0.000 description 8
- 229920006395 saturated elastomer Polymers 0.000 description 8
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 7
- 229910021529 ammonia Inorganic materials 0.000 description 7
- 238000001816 cooling Methods 0.000 description 7
- 230000001965 increasing effect Effects 0.000 description 7
- 210000002161 motor neuron Anatomy 0.000 description 7
- 239000002953 phosphate buffered saline Substances 0.000 description 7
- 239000000047 product Substances 0.000 description 7
- 238000012360 testing method Methods 0.000 description 7
- 230000001225 therapeutic effect Effects 0.000 description 7
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 6
- 229920000858 Cyclodextrin Polymers 0.000 description 6
- 101150081851 SMN1 gene Proteins 0.000 description 6
- 101150015954 SMN2 gene Proteins 0.000 description 6
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 6
- HEDRZPFGACZZDS-MICDWDOJSA-N Trichloro(2H)methane Chemical compound [2H]C(Cl)(Cl)Cl HEDRZPFGACZZDS-MICDWDOJSA-N 0.000 description 6
- 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 6
- 230000008901 benefit Effects 0.000 description 6
- 239000000460 chlorine Chemical group 0.000 description 6
- 239000002552 dosage form Substances 0.000 description 6
- 230000000694 effects Effects 0.000 description 6
- 238000001727 in vivo Methods 0.000 description 6
- 230000002503 metabolic effect Effects 0.000 description 6
- 238000012544 monitoring process Methods 0.000 description 6
- NFHFRUOZVGFOOS-UHFFFAOYSA-N palladium;triphenylphosphane Chemical compound [Pd].C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 NFHFRUOZVGFOOS-UHFFFAOYSA-N 0.000 description 6
- IVDFJHOHABJVEH-UHFFFAOYSA-N pinacol Chemical compound CC(C)(O)C(C)(C)O IVDFJHOHABJVEH-UHFFFAOYSA-N 0.000 description 6
- BWHMMNNQKKPAPP-UHFFFAOYSA-L potassium carbonate Chemical compound [K+].[K+].[O-]C([O-])=O BWHMMNNQKKPAPP-UHFFFAOYSA-L 0.000 description 6
- DTQVDTLACAAQTR-UHFFFAOYSA-N trifluoroacetic acid Substances OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 6
- JIAARYAFYJHUJI-UHFFFAOYSA-L zinc dichloride Chemical compound [Cl-].[Cl-].[Zn+2] JIAARYAFYJHUJI-UHFFFAOYSA-L 0.000 description 6
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 5
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 5
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 229910052731 fluorine Inorganic materials 0.000 description 5
- 230000006870 function Effects 0.000 description 5
- 238000010438 heat treatment Methods 0.000 description 5
- JMMWKPVZQRWMSS-UHFFFAOYSA-N isopropanol acetate Natural products CC(C)OC(C)=O JMMWKPVZQRWMSS-UHFFFAOYSA-N 0.000 description 5
- 230000035772 mutation Effects 0.000 description 5
- 229930027945 nicotinamide-adenine dinucleotide Natural products 0.000 description 5
- 239000000741 silica gel Substances 0.000 description 5
- 229910002027 silica gel Inorganic materials 0.000 description 5
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical class O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 5
- KZPYGQFFRCFCPP-UHFFFAOYSA-N 1,1'-bis(diphenylphosphino)ferrocene Chemical compound [Fe+2].C1=CC=C[C-]1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=C[C-]1P(C=1C=CC=CC=1)C1=CC=CC=C1 KZPYGQFFRCFCPP-UHFFFAOYSA-N 0.000 description 4
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 4
- 239000000969 carrier Substances 0.000 description 4
- 239000003795 chemical substances by application Substances 0.000 description 4
- 238000010790 dilution Methods 0.000 description 4
- 239000012895 dilution Substances 0.000 description 4
- 238000011534 incubation Methods 0.000 description 4
- 238000002347 injection Methods 0.000 description 4
- 239000007924 injection Substances 0.000 description 4
- 238000001990 intravenous administration Methods 0.000 description 4
- 229910052740 iodine Inorganic materials 0.000 description 4
- 229940011051 isopropyl acetate Drugs 0.000 description 4
- GWYFCOCPABKNJV-UHFFFAOYSA-N isovaleric acid Chemical compound CC(C)CC(O)=O GWYFCOCPABKNJV-UHFFFAOYSA-N 0.000 description 4
- 238000001294 liquid chromatography-tandem mass spectrometry Methods 0.000 description 4
- 230000004060 metabolic process Effects 0.000 description 4
- KDLHZDBZIXYQEI-UHFFFAOYSA-N palladium Substances [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 description 4
- 235000011056 potassium acetate Nutrition 0.000 description 4
- 239000000843 powder Substances 0.000 description 4
- 102000004169 proteins and genes Human genes 0.000 description 4
- 238000000746 purification Methods 0.000 description 4
- 230000008929 regeneration Effects 0.000 description 4
- 238000011069 regeneration method Methods 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- WLPUWLXVBWGYMZ-UHFFFAOYSA-N tricyclohexylphosphine Chemical compound C1CCCCC1P(C1CCCCC1)C1CCCCC1 WLPUWLXVBWGYMZ-UHFFFAOYSA-N 0.000 description 4
- 238000005160 1H NMR spectroscopy Methods 0.000 description 3
- JWUJQDFVADABEY-UHFFFAOYSA-N 2-methyltetrahydrofuran Chemical compound CC1CCCO1 JWUJQDFVADABEY-UHFFFAOYSA-N 0.000 description 3
- YYROPELSRYBVMQ-UHFFFAOYSA-N 4-toluenesulfonyl chloride Chemical compound CC1=CC=C(S(Cl)(=O)=O)C=C1 YYROPELSRYBVMQ-UHFFFAOYSA-N 0.000 description 3
- SUBDBMMJDZJVOS-UHFFFAOYSA-N 5-methoxy-2-{[(4-methoxy-3,5-dimethylpyridin-2-yl)methyl]sulfinyl}-1H-benzimidazole Chemical compound N=1C2=CC(OC)=CC=C2NC=1S(=O)CC1=NC=C(C)C(OC)=C1C SUBDBMMJDZJVOS-UHFFFAOYSA-N 0.000 description 3
- ASKZRYGFUPSJPN-UHFFFAOYSA-N 7-(4,7-diazaspiro[2.5]octan-7-yl)-2-(2,8-dimethylimidazo[1,2-b]pyridazin-6-yl)pyrido[1,2-a]pyrimidin-4-one Chemical class CC1=CN2N=C(C=C(C)C2=N1)C1=CC(=O)N2C=C(C=CC2=N1)N1CCNC2(CC2)C1 ASKZRYGFUPSJPN-UHFFFAOYSA-N 0.000 description 3
- HBAQYPYDRFILMT-UHFFFAOYSA-N 8-[3-(1-cyclopropylpyrazol-4-yl)-1H-pyrazolo[4,3-d]pyrimidin-5-yl]-3-methyl-3,8-diazabicyclo[3.2.1]octan-2-one Chemical class C1(CC1)N1N=CC(=C1)C1=NNC2=C1N=C(N=C2)N1C2C(N(CC1CC2)C)=O HBAQYPYDRFILMT-UHFFFAOYSA-N 0.000 description 3
- NLXLAEXVIDQMFP-UHFFFAOYSA-N Ammonia chloride Chemical class [NH4+].[Cl-] NLXLAEXVIDQMFP-UHFFFAOYSA-N 0.000 description 3
- 101000617738 Homo sapiens Survival motor neuron protein Proteins 0.000 description 3
- MKXZASYAUGDDCJ-SZMVWBNQSA-N LSM-2525 Chemical compound C1CCC[C@H]2[C@@]3([H])N(C)CC[C@]21C1=CC(OC)=CC=C1C3 MKXZASYAUGDDCJ-SZMVWBNQSA-N 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- CTQNGGLPUBDAKN-UHFFFAOYSA-N O-Xylene Chemical compound CC1=CC=CC=C1C CTQNGGLPUBDAKN-UHFFFAOYSA-N 0.000 description 3
- UIIMBOGNXHQVGW-DEQYMQKBSA-M Sodium bicarbonate-14C Chemical compound [Na+].O[14C]([O-])=O UIIMBOGNXHQVGW-DEQYMQKBSA-M 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 3
- 125000004429 atom Chemical group 0.000 description 3
- 229960004853 betadex Drugs 0.000 description 3
- 238000002425 crystallisation Methods 0.000 description 3
- 230000008025 crystallization Effects 0.000 description 3
- 229960001985 dextromethorphan Drugs 0.000 description 3
- 239000012065 filter cake Substances 0.000 description 3
- 239000000706 filtrate Substances 0.000 description 3
- 238000001914 filtration Methods 0.000 description 3
- 238000004128 high performance liquid chromatography Methods 0.000 description 3
- VTVRXITWWZGKHV-UHFFFAOYSA-N imidazo[1,2-b]pyridazine Chemical compound N1=CC=CC2=NC=CN21 VTVRXITWWZGKHV-UHFFFAOYSA-N 0.000 description 3
- 238000001802 infusion Methods 0.000 description 3
- 230000002401 inhibitory effect Effects 0.000 description 3
- 210000003205 muscle Anatomy 0.000 description 3
- 229960000381 omeprazole Drugs 0.000 description 3
- 229910000027 potassium carbonate Inorganic materials 0.000 description 3
- 238000012545 processing Methods 0.000 description 3
- 230000000069 prophylactic effect Effects 0.000 description 3
- 238000011321 prophylaxis Methods 0.000 description 3
- 125000006239 protecting group Chemical group 0.000 description 3
- 238000007789 sealing Methods 0.000 description 3
- 235000017557 sodium bicarbonate Nutrition 0.000 description 3
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 3
- 238000006467 substitution reaction Methods 0.000 description 3
- 239000000758 substrate Substances 0.000 description 3
- 239000012730 sustained-release form Substances 0.000 description 3
- 238000005406 washing Methods 0.000 description 3
- 239000012224 working solution Substances 0.000 description 3
- 239000008096 xylene Substances 0.000 description 3
- 239000011592 zinc chloride Substances 0.000 description 3
- 235000005074 zinc chloride Nutrition 0.000 description 3
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- LOMKIDUODAAOHI-UHFFFAOYSA-N CC(C)(C)OC(=O)N1CCN(CC11CC1)C1=CN2C(=O)C=C(O)N=C2C=C1 Chemical compound CC(C)(C)OC(=O)N1CCN(CC11CC1)C1=CN2C(=O)C=C(O)N=C2C=C1 LOMKIDUODAAOHI-UHFFFAOYSA-N 0.000 description 2
- AUPXBVDHVRZMIB-UHFFFAOYSA-M C[Mg]I Chemical class C[Mg]I AUPXBVDHVRZMIB-UHFFFAOYSA-M 0.000 description 2
- 208000019505 Deglutition disease Diseases 0.000 description 2
- 208000000059 Dyspnea Diseases 0.000 description 2
- 206010013975 Dyspnoeas Diseases 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- 239000001116 FEMA 4028 Substances 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- 208000010428 Muscle Weakness Diseases 0.000 description 2
- 206010028289 Muscle atrophy Diseases 0.000 description 2
- 206010028372 Muscular weakness Diseases 0.000 description 2
- 229910019142 PO4 Inorganic materials 0.000 description 2
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical group [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- 101150113275 Smn gene Proteins 0.000 description 2
- 208000027418 Wounds and injury Diseases 0.000 description 2
- 239000002671 adjuvant Substances 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- WHGYBXFWUBPSRW-FOUAGVGXSA-N beta-cyclodextrin 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](CO)[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](CO)[C@H]([C@@H]([C@H]3O)O)O3)[C@H](O)[C@H]2O)CO)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@@H]3O[C@@H]1CO WHGYBXFWUBPSRW-FOUAGVGXSA-N 0.000 description 2
- 235000011175 beta-cyclodextrine Nutrition 0.000 description 2
- 238000009835 boiling Methods 0.000 description 2
- VQFAIAKCILWQPZ-UHFFFAOYSA-N bromoacetone Chemical compound CC(=O)CBr VQFAIAKCILWQPZ-UHFFFAOYSA-N 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- 210000003169 central nervous system Anatomy 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 238000004587 chromatography analysis Methods 0.000 description 2
- 210000000349 chromosome Anatomy 0.000 description 2
- 230000001684 chronic effect Effects 0.000 description 2
- 239000006071 cream Substances 0.000 description 2
- 239000012043 crude product Substances 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- 230000007850 degeneration Effects 0.000 description 2
- 238000012217 deletion Methods 0.000 description 2
- 230000037430 deletion Effects 0.000 description 2
- 230000002708 enhancing effect Effects 0.000 description 2
- 150000002148 esters Chemical class 0.000 description 2
- 238000000605 extraction Methods 0.000 description 2
- 239000000796 flavoring agent Substances 0.000 description 2
- 125000000524 functional group Chemical group 0.000 description 2
- 230000000415 inactivating effect Effects 0.000 description 2
- 208000014674 injury Diseases 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- KWGKDLIKAYFUFQ-UHFFFAOYSA-M lithium chloride Chemical compound [Li+].[Cl-] KWGKDLIKAYFUFQ-UHFFFAOYSA-M 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 230000004199 lung function Effects 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 238000002844 melting Methods 0.000 description 2
- 230000008018 melting Effects 0.000 description 2
- OSWPMRLSEDHDFF-UHFFFAOYSA-N methyl salicylate Chemical compound COC(=O)C1=CC=CC=C1O OSWPMRLSEDHDFF-UHFFFAOYSA-N 0.000 description 2
- 230000003228 microsomal effect Effects 0.000 description 2
- 201000000585 muscular atrophy Diseases 0.000 description 2
- 239000002674 ointment Substances 0.000 description 2
- 230000003285 pharmacodynamic effect Effects 0.000 description 2
- 230000000144 pharmacologic effect Effects 0.000 description 2
- 239000008363 phosphate buffer Substances 0.000 description 2
- 229920001223 polyethylene glycol Polymers 0.000 description 2
- 230000002035 prolonged effect Effects 0.000 description 2
- 238000010791 quenching Methods 0.000 description 2
- 230000000171 quenching effect Effects 0.000 description 2
- 239000011541 reaction mixture Substances 0.000 description 2
- 230000000241 respiratory effect Effects 0.000 description 2
- HFHDHCJBZVLPGP-UHFFFAOYSA-N schardinger α-dextrin Chemical class O1C(C(C2O)O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC(C(O)C2O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC2C(O)C(O)C1OC2CO HFHDHCJBZVLPGP-UHFFFAOYSA-N 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- GEHJYWRUCIMESM-UHFFFAOYSA-L sodium sulfite Chemical compound [Na+].[Na+].[O-]S([O-])=O GEHJYWRUCIMESM-UHFFFAOYSA-L 0.000 description 2
- 239000012089 stop solution Substances 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 238000013268 sustained release Methods 0.000 description 2
- 125000003396 thiol group Chemical group [H]S* 0.000 description 2
- 230000000699 topical effect Effects 0.000 description 2
- RIOQSEWOXXDEQQ-UHFFFAOYSA-N triphenylphosphine Chemical compound C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 RIOQSEWOXXDEQQ-UHFFFAOYSA-N 0.000 description 2
- 229910021642 ultra pure water Inorganic materials 0.000 description 2
- 239000012498 ultrapure water Substances 0.000 description 2
- 238000001291 vacuum drying Methods 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- KYVBNYUBXIEUFW-UHFFFAOYSA-N 1,1,3,3-tetramethylguanidine Chemical compound CN(C)C(=N)N(C)C KYVBNYUBXIEUFW-UHFFFAOYSA-N 0.000 description 1
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 1
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 1
- CHHHXKFHOYLYRE-UHFFFAOYSA-M 2,4-Hexadienoic acid, potassium salt (1:1), (2E,4E)- Chemical compound [K+].CC=CC=CC([O-])=O CHHHXKFHOYLYRE-UHFFFAOYSA-M 0.000 description 1
- MEAPRSDUXBHXGD-UHFFFAOYSA-N 3-chloro-n-(4-propan-2-ylphenyl)propanamide Chemical compound CC(C)C1=CC=C(NC(=O)CCCl)C=C1 MEAPRSDUXBHXGD-UHFFFAOYSA-N 0.000 description 1
- FGOWNGCSUSKHQI-UHFFFAOYSA-N 4-bromo-6-chloropyridazin-3-amine Chemical compound NC1=NN=C(Cl)C=C1Br FGOWNGCSUSKHQI-UHFFFAOYSA-N 0.000 description 1
- ATXXLNCPVSUCNK-UHFFFAOYSA-N 5-bromo-2-nitropyridine Chemical compound [O-][N+](=O)C1=CC=C(Br)C=N1 ATXXLNCPVSUCNK-UHFFFAOYSA-N 0.000 description 1
- QTBSBXVTEAMEQO-UHFFFAOYSA-M Acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 description 1
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical class CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 1
- 229920001450 Alpha-Cyclodextrin Polymers 0.000 description 1
- 206010002091 Anaesthesia Diseases 0.000 description 1
- 241000416162 Astragalus gummifer Species 0.000 description 1
- 102000004506 Blood Proteins Human genes 0.000 description 1
- 108010017384 Blood Proteins Proteins 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- WKBOTKDWSSQWDR-UHFFFAOYSA-N Bromine atom Chemical compound [Br] WKBOTKDWSSQWDR-UHFFFAOYSA-N 0.000 description 1
- 240000008213 Brosimum alicastrum Species 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical group [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- OKTJSMMVPCPJKN-NJFSPNSNSA-N Carbon-14 Chemical compound [14C] OKTJSMMVPCPJKN-NJFSPNSNSA-N 0.000 description 1
- ZAMOUSCENKQFHK-UHFFFAOYSA-N Chlorine atom Chemical group [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 206010011224 Cough Diseases 0.000 description 1
- 206010011469 Crying Diseases 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- PXGOKWXKJXAPGV-UHFFFAOYSA-N Fluorine Chemical group FF PXGOKWXKJXAPGV-UHFFFAOYSA-N 0.000 description 1
- BDAGIHXWWSANSR-UHFFFAOYSA-M Formate Chemical compound [O-]C=O BDAGIHXWWSANSR-UHFFFAOYSA-M 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical class OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 102000008100 Human Serum Albumin Human genes 0.000 description 1
- 108091006905 Human Serum Albumin Proteins 0.000 description 1
- 208000008454 Hyperhidrosis Diseases 0.000 description 1
- 206010021118 Hypotonia Diseases 0.000 description 1
- 238000004566 IR spectroscopy Methods 0.000 description 1
- 206010050740 Inability to crawl Diseases 0.000 description 1
- 206010023201 Joint contracture 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
- 239000004166 Lanolin Substances 0.000 description 1
- 241000282567 Macaca fascicularis Species 0.000 description 1
- 241000282560 Macaca mulatta Species 0.000 description 1
- 244000246386 Mentha pulegium Species 0.000 description 1
- 235000016257 Mentha pulegium Nutrition 0.000 description 1
- 235000004357 Mentha x piperita Nutrition 0.000 description 1
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 1
- 208000007379 Muscle Hypotonia Diseases 0.000 description 1
- 206010049816 Muscle tightness Diseases 0.000 description 1
- 238000005481 NMR spectroscopy Methods 0.000 description 1
- 239000006057 Non-nutritive feed additive Substances 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 102000007327 Protamines Human genes 0.000 description 1
- 108010007568 Protamines Proteins 0.000 description 1
- 208000033526 Proximal spinal muscular atrophy type 3 Diseases 0.000 description 1
- 206010057190 Respiratory tract infections Diseases 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- VMHLLURERBWHNL-UHFFFAOYSA-M Sodium acetate Chemical compound [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 description 1
- 208000003954 Spinal Muscular Atrophies of Childhood Diseases 0.000 description 1
- 229920002472 Starch Polymers 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
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical group [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 1
- 241000282898 Sus scrofa Species 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
- 229920001615 Tragacanth Polymers 0.000 description 1
- 206010044565 Tremor Diseases 0.000 description 1
- YZCKVEUIGOORGS-NJFSPNSNSA-N Tritium Chemical compound [3H] YZCKVEUIGOORGS-NJFSPNSNSA-N 0.000 description 1
- 238000002441 X-ray diffraction Methods 0.000 description 1
- LUTSRLYCMSCGCS-BWOMAWGNSA-N [(3s,8r,9s,10r,13s)-10,13-dimethyl-17-oxo-1,2,3,4,7,8,9,11,12,16-decahydrocyclopenta[a]phenanthren-3-yl] acetate Chemical compound C([C@@H]12)C[C@]3(C)C(=O)CC=C3[C@@H]1CC=C1[C@]2(C)CC[C@H](OC(=O)C)C1 LUTSRLYCMSCGCS-BWOMAWGNSA-N 0.000 description 1
- JGFACMOFRHDKKK-UHFFFAOYSA-N [N+](=O)([O-])C1=CC=C(C=N1)N1CCN(C2(CC2)C1)C(=O)OC(C)(C)C Chemical compound [N+](=O)([O-])C1=CC=C(C=N1)N1CCN(C2(CC2)C1)C(=O)OC(C)(C)C JGFACMOFRHDKKK-UHFFFAOYSA-N 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 238000009825 accumulation 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
- PBCJIPOGFJYBJE-UHFFFAOYSA-N acetonitrile;hydrate Chemical compound O.CC#N PBCJIPOGFJYBJE-UHFFFAOYSA-N 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 239000011149 active material Substances 0.000 description 1
- 238000012382 advanced drug delivery Methods 0.000 description 1
- 239000000783 alginic acid Substances 0.000 description 1
- 235000010443 alginic acid Nutrition 0.000 description 1
- 229920000615 alginic acid Polymers 0.000 description 1
- 229960001126 alginic acid Drugs 0.000 description 1
- 150000004781 alginic acids Chemical class 0.000 description 1
- PNEYBMLMFCGWSK-UHFFFAOYSA-N aluminium oxide Inorganic materials [O-2].[O-2].[O-2].[Al+3].[Al+3] PNEYBMLMFCGWSK-UHFFFAOYSA-N 0.000 description 1
- CEGOLXSVJUTHNZ-UHFFFAOYSA-K aluminium tristearate Chemical compound [Al+3].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O CEGOLXSVJUTHNZ-UHFFFAOYSA-K 0.000 description 1
- 229940063655 aluminum stearate Drugs 0.000 description 1
- 230000037005 anaesthesia Effects 0.000 description 1
- 210000002226 anterior horn cell Anatomy 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 238000011914 asymmetric synthesis Methods 0.000 description 1
- 230000037147 athletic performance Effects 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical group [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 230000037444 atrophy Effects 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 150000001558 benzoic acid derivatives Chemical class 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 230000008512 biological response Effects 0.000 description 1
- WYPCGKBOSFOHGU-UHFFFAOYSA-N bis(2,4,6-trichlorophenyl) propanedioate Chemical compound ClC1=CC(Cl)=CC(Cl)=C1OC(=O)CC(=O)OC1=C(Cl)C=C(Cl)C=C1Cl WYPCGKBOSFOHGU-UHFFFAOYSA-N 0.000 description 1
- 230000036765 blood level Effects 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 210000000133 brain stem Anatomy 0.000 description 1
- GDTBXPJZTBHREO-UHFFFAOYSA-N bromine Substances BrBr GDTBXPJZTBHREO-UHFFFAOYSA-N 0.000 description 1
- 229910052794 bromium Inorganic materials 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 150000001732 carboxylic acid derivatives Chemical class 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000006727 cell loss Effects 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 235000010980 cellulose Nutrition 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 210000002230 centromere Anatomy 0.000 description 1
- 125000003636 chemical group Chemical group 0.000 description 1
- 229910052801 chlorine Chemical group 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 229940075614 colloidal silicon dioxide Drugs 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 238000004440 column chromatography Methods 0.000 description 1
- 208000018697 congenital contractures Diseases 0.000 description 1
- 239000008120 corn starch Substances 0.000 description 1
- 229940097362 cyclodextrins Drugs 0.000 description 1
- VRLDVERQJMEPIF-UHFFFAOYSA-N dbdmh Chemical compound CC1(C)N(Br)C(=O)N(Br)C1=O VRLDVERQJMEPIF-UHFFFAOYSA-N 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 230000001934 delay Effects 0.000 description 1
- 238000010511 deprotection reaction Methods 0.000 description 1
- CSCPPACGZOOCGX-WFGJKAKNSA-N deuterated acetone Substances [2H]C([2H])([2H])C(=O)C([2H])([2H])[2H] CSCPPACGZOOCGX-WFGJKAKNSA-N 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 235000014113 dietary fatty acids Nutrition 0.000 description 1
- 150000004683 dihydrates Chemical class 0.000 description 1
- 238000007865 diluting Methods 0.000 description 1
- FSBVERYRVPGNGG-UHFFFAOYSA-N dimagnesium dioxido-bis[[oxido(oxo)silyl]oxy]silane hydrate Chemical compound O.[Mg+2].[Mg+2].[O-][Si](=O)O[Si]([O-])([O-])O[Si]([O-])=O FSBVERYRVPGNGG-UHFFFAOYSA-N 0.000 description 1
- ZPWVASYFFYYZEW-UHFFFAOYSA-L dipotassium hydrogen phosphate Chemical compound [K+].[K+].OP([O-])([O-])=O ZPWVASYFFYYZEW-UHFFFAOYSA-L 0.000 description 1
- 229910000396 dipotassium phosphate Inorganic materials 0.000 description 1
- 235000019797 dipotassium phosphate Nutrition 0.000 description 1
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 1
- 230000006806 disease prevention Effects 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- BNIILDVGGAEEIG-UHFFFAOYSA-L disodium hydrogen phosphate Chemical compound [Na+].[Na+].OP([O-])([O-])=O BNIILDVGGAEEIG-UHFFFAOYSA-L 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 238000004090 dissolution Methods 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 238000012377 drug delivery Methods 0.000 description 1
- 238000009510 drug design Methods 0.000 description 1
- 238000001035 drying Methods 0.000 description 1
- 239000003792 electrolyte Substances 0.000 description 1
- 238000002567 electromyography Methods 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- 125000004185 ester group Chemical group 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 125000004494 ethyl ester group Chemical group 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 239000000194 fatty acid Substances 0.000 description 1
- 229930195729 fatty acid Natural products 0.000 description 1
- 150000004665 fatty acids Chemical class 0.000 description 1
- KTWOOEGAPBSYNW-UHFFFAOYSA-N ferrocene Chemical compound [Fe+2].C=1C=C[CH-]C=1.C=1C=C[CH-]C=1 KTWOOEGAPBSYNW-UHFFFAOYSA-N 0.000 description 1
- 230000001605 fetal effect Effects 0.000 description 1
- 235000019634 flavors Nutrition 0.000 description 1
- 239000011737 fluorine Chemical group 0.000 description 1
- 235000013305 food Nutrition 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 235000003599 food sweetener Nutrition 0.000 description 1
- 238000007710 freezing Methods 0.000 description 1
- 230000008014 freezing Effects 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 229940121390 gene splicing modulator Drugs 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 125000002791 glucosyl group Chemical group C1([C@H](O)[C@@H](O)[C@H](O)[C@H](O1)CO)* 0.000 description 1
- 125000005456 glyceride group Chemical group 0.000 description 1
- 229960002449 glycine Drugs 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 150000004687 hexahydrates Chemical class 0.000 description 1
- 235000001050 hortel pimenta Nutrition 0.000 description 1
- 125000004435 hydrogen atom Chemical group [H]* 0.000 description 1
- 230000007062 hydrolysis Effects 0.000 description 1
- 238000006460 hydrolysis reaction Methods 0.000 description 1
- 239000008309 hydrophilic cream Substances 0.000 description 1
- 239000005457 ice water Substances 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 238000002329 infrared spectrum Methods 0.000 description 1
- 239000007972 injectable composition Substances 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 238000007917 intracranial administration Methods 0.000 description 1
- 238000007919 intrasynovial administration Methods 0.000 description 1
- 150000002500 ions Chemical class 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 201000004815 juvenile spinal muscular atrophy Diseases 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 208000030175 lameness Diseases 0.000 description 1
- 229940039717 lanolin Drugs 0.000 description 1
- 235000019388 lanolin Nutrition 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 229940067606 lecithin Drugs 0.000 description 1
- 239000006193 liquid solution Substances 0.000 description 1
- 239000006194 liquid suspension Substances 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 239000000391 magnesium silicate Substances 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 235000019793 magnesium trisilicate Nutrition 0.000 description 1
- 229940099273 magnesium trisilicate Drugs 0.000 description 1
- 229910000386 magnesium trisilicate Inorganic materials 0.000 description 1
- VXWPONVCMVLXBW-UHFFFAOYSA-M magnesium;carbanide;iodide Chemical compound [CH3-].[Mg+2].[I-] VXWPONVCMVLXBW-UHFFFAOYSA-M 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 230000007721 medicinal effect Effects 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 239000002207 metabolite Substances 0.000 description 1
- 150000004702 methyl esters Chemical class 0.000 description 1
- STZCRXQWRGQSJD-GEEYTBSJSA-M methyl orange Chemical compound [Na+].C1=CC(N(C)C)=CC=C1\N=N\C1=CC=C(S([O-])(=O)=O)C=C1 STZCRXQWRGQSJD-GEEYTBSJSA-M 0.000 description 1
- 229940012189 methyl orange Drugs 0.000 description 1
- 229960001047 methyl salicylate Drugs 0.000 description 1
- 229940016286 microcrystalline cellulose Drugs 0.000 description 1
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 1
- 239000008108 microcrystalline cellulose Substances 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 150000004682 monohydrates Chemical class 0.000 description 1
- 230000007659 motor function Effects 0.000 description 1
- 230000020763 muscle atrophy Effects 0.000 description 1
- 230000032405 negative regulation of neuron apoptotic process Effects 0.000 description 1
- 230000004770 neurodegeneration Effects 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 239000003883 ointment base Substances 0.000 description 1
- 230000005693 optoelectronics Effects 0.000 description 1
- 229940126701 oral medication Drugs 0.000 description 1
- 150000002894 organic compounds Chemical class 0.000 description 1
- 239000001301 oxygen Chemical group 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- PIBWKRNGBLPSSY-UHFFFAOYSA-L palladium(II) chloride Chemical compound Cl[Pd]Cl PIBWKRNGBLPSSY-UHFFFAOYSA-L 0.000 description 1
- YJVFFLUZDVXJQI-UHFFFAOYSA-L palladium(ii) acetate Chemical compound [Pd+2].CC([O-])=O.CC([O-])=O YJVFFLUZDVXJQI-UHFFFAOYSA-L 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 235000021317 phosphate Nutrition 0.000 description 1
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 1
- 229910052698 phosphorus Inorganic materials 0.000 description 1
- 239000011574 phosphorus Chemical group 0.000 description 1
- 230000000704 physical effect Effects 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 229920000058 polyacrylate Polymers 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 239000004302 potassium sorbate Substances 0.000 description 1
- 235000010241 potassium sorbate Nutrition 0.000 description 1
- 229940069338 potassium sorbate Drugs 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 239000002244 precipitate Substances 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 230000000750 progressive effect Effects 0.000 description 1
- 229960004604 propranolol hydrochloride Drugs 0.000 description 1
- AQHHHDLHHXJYJD-UHFFFAOYSA-N propranolol hydrochloride Natural products C1=CC=C2C(OCC(O)CNC(C)C)=CC=CC2=C1 AQHHHDLHHXJYJD-UHFFFAOYSA-N 0.000 description 1
- 229950008679 protamine sulfate Drugs 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 235000005828 ramon Nutrition 0.000 description 1
- 239000000376 reactant Substances 0.000 description 1
- 230000009257 reactivity Effects 0.000 description 1
- 238000001953 recrystallisation Methods 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 230000011514 reflex Effects 0.000 description 1
- 208000020029 respiratory tract infectious disease Diseases 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 229940121322 risdiplam Drugs 0.000 description 1
- CVHZOJJKTDOEJC-UHFFFAOYSA-N saccharin Chemical compound C1=CC=C2C(=O)NS(=O)(=O)C2=C1 CVHZOJJKTDOEJC-UHFFFAOYSA-N 0.000 description 1
- 229940081974 saccharin Drugs 0.000 description 1
- 235000019204 saccharin Nutrition 0.000 description 1
- 239000000901 saccharin and its Na,K and Ca salt Substances 0.000 description 1
- 239000012266 salt solution Substances 0.000 description 1
- 238000012284 sample analysis method Methods 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 239000001632 sodium acetate Substances 0.000 description 1
- 235000017281 sodium acetate Nutrition 0.000 description 1
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 1
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 1
- 235000010265 sodium sulphite Nutrition 0.000 description 1
- PODWXQQNRWNDGD-UHFFFAOYSA-L sodium thiosulfate pentahydrate Chemical compound O.O.O.O.O.[Na+].[Na+].[O-]S([S-])(=O)=O PODWXQQNRWNDGD-UHFFFAOYSA-L 0.000 description 1
- 239000008247 solid mixture Substances 0.000 description 1
- 238000003797 solvolysis reaction Methods 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 238000002798 spectrophotometry method Methods 0.000 description 1
- 210000000278 spinal cord Anatomy 0.000 description 1
- 238000013222 sprague-dawley male rat Methods 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 239000007858 starting material Substances 0.000 description 1
- 239000011550 stock solution Substances 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 125000001424 substituent group Chemical group 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 125000004964 sulfoalkyl group Chemical group 0.000 description 1
- 229940097346 sulfobutylether-beta-cyclodextrin Drugs 0.000 description 1
- 229910052717 sulfur Inorganic materials 0.000 description 1
- 239000011593 sulfur Chemical group 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 208000013460 sweaty Diseases 0.000 description 1
- 239000003765 sweetening agent Substances 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 210000003411 telomere Anatomy 0.000 description 1
- 102000055501 telomere Human genes 0.000 description 1
- 108091035539 telomere Proteins 0.000 description 1
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 238000003354 tissue distribution assay Methods 0.000 description 1
- 229960005371 tolbutamide Drugs 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 229910052722 tritium Inorganic materials 0.000 description 1
- 208000032527 type III spinal muscular atrophy Diseases 0.000 description 1
- 235000013311 vegetables Nutrition 0.000 description 1
- 210000003462 vein Anatomy 0.000 description 1
- 238000003260 vortexing Methods 0.000 description 1
- 239000001993 wax Substances 0.000 description 1
- 230000036266 weeks of gestation Effects 0.000 description 1
- 238000005303 weighing Methods 0.000 description 1
- 150000003751 zinc Chemical class 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D519/00—Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P21/00—Drugs for disorders of the muscular or neuromuscular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/28—Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Neurology (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Physical Education & Sports Medicine (AREA)
- Biomedical Technology (AREA)
- Neurosurgery (AREA)
- Epidemiology (AREA)
- Hospice & Palliative Care (AREA)
- Psychiatry (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The invention provides a substituted pyridopyrimidinone compound, a composition containing the compound and application thereof, wherein the compound is shown as a formula (I) or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate or solvent compound thereof. The compounds of formula (I) are useful as SMN2 (Survival of motor neuron 2) gene splice modulators for the treatment of spinal muscular atrophy (Spinal Muscular Atrophy, SMA), with high selectivity and good pharmacokinetic properties.
Description
Technical Field
The invention belongs to the technical field of biological medicines, and particularly relates to a substituted pyridopyrimidinone compound, a composition containing the compound and application thereof. More particularly, the present invention relates to certain deuterium-substituted 7- (4, 7-diazaspiro [2.5] oct-7-yl) -2- (2, 8-dimethylimidazo [1,2-b ] pyridazin-6-yl) pyrido [1,2-a ] pyrimidin-4-one compounds, and pharmaceutically acceptable salts, tautomers, stereoisomers, prodrugs, crystalline forms, hydrates, or solvates thereof. These deuterium substituted compounds and compositions thereof are useful as SMN2 (Survival of motor neuron 2) gene splice modulators for the treatment of spinal muscular atrophy (Spinal Muscular Atrophy, SMA) with high selectivity and good pharmacokinetic properties.
Background
Spinal Muscular Atrophy (SMA) is a variety of hereditary and acquired Central Nervous System (CNS) diseases characterized by progressive motor neuron injury in the spinal cord and brain stem, which causes muscle weakness and atrophy. SMA is most commonly caused by mutations in the motor neuron Survival (SMN) gene and appears to severely affect from infant to adult.
Clinical profiles of SMA disorders have been divided into the following five groups:
(1) Type 0 SMA (intrauterine SMA) is the most severe form of disease and begins before birth. Typically, the first symptom of type 0 SMA is a decrease in fetal movement that can be observed first between 30 and 36 weeks of gestation. After birth, these newborns rarely move and dysphagia and dyspnea.
(2) Type I SMA (infant SMA or wir-Huo Ershi disease) is symptomatic for 0 to 6 months, which is also very severe. The patient is never able to sit up and, due to no respiratory support, death usually occurs in the first 2 years.
(3) The age of onset of type 2 SMA (transitional SMA) is 7-18 months. The patient can sit without support, but cannot stand or walk independently. The prognosis of this group depends largely on the degree of respiratory correlation.
(4) Type 3 SMA (adolescents or ku-wei-di disease (Kugelberg-WELANDER DISEASE)) is usually diagnosed after 18 months. Type 3 SMA individuals are able to walk independently at certain times during the course of the disease, but typically rely on wheelchairs during young or adult life.
(5) Type 4 SMA (adult onset SMA). Symptoms of weakness usually begin in the tongue, hands or feet in the late adolescence and then progress to other areas of the body. Adult SMA processes are slower and have little or no effect on life expectancy.
SMN gene maps have been obtained by linkage analysis of complex regions in chromosome 5 q. In humans, this region contains approximately 50 kilobase pair (kb) inverted copies, resulting in two nearly identical copies of the SMN gene. SMA is caused by an inactivating mutation or deletion of the telomere copy of the gene (SMN 1) on both chromosomes, resulting in loss of SMN1 gene function. However, all patients retained a centromere copy of the gene (SMN 2), and the copy number of the SMN2 gene in SMA patients was generally inversely related to disease severity; i.e. patients with less severe SMA have more copies of SMN 2. Nonetheless, SMN2 cannot fully compensate for the loss of SMN1 function due to alternative splicing of exon 7 caused by a translationally silent C-to-T mutation in exon 7. Thus, most transcripts produced by SMN2 are deleted for exon 7 (Δ7smn2) and encode truncated SMN proteins that have impaired function and are rapidly degraded.
SMN proteins are thought to play a role in RNA processing and metabolism, functioning as a well-identified mediator of the assembly of a specific class of RNA-protein complexes called snrnps. In motor neurons, SMN may have other functions, however, its role in preventing selective degeneration of motor neurons has not been well established.
In most cases, SMA is diagnosed based on clinical symptoms and by the presence of at least one SMN1 gene copy test. However, in about 5% of the conditions SMA is caused by mutations in genes other than SMN1 inactivation, some of which are known and others are not yet established. In some cases, when SMN1 gene testing is not feasible or does not show any abnormalities, other tests such as Electromyography (EMG) or muscle tissue examination may be indicated.
RISDIPLAM (chemical name 7- (4, 7-diazaspiro [2.5] oct-7-yl) -2- (2, 8-dimethylimidazo [1,2-b ] pyridazin-6-yl) pyrido [1,2-a ] pyrimidin-4-one, which has the following structural formula) is the first oral small molecule SMN2 gene splicing modulator to treat SMA worldwide, which can increase functional SMN proteins systemically.
Poor absorption, distribution, metabolism and/or excretion (ADME) properties are known to be the leading cause of failure in many candidate drug clinical trials. Many drugs currently on the market also limit their range of application due to poor ADME properties. Rapid metabolism of drugs can result in many drugs that would otherwise be effective in treating the disease being difficult to formulate due to too rapid clearance from the body's metabolism. Frequent or high dose administration, while potentially solving the problem of rapid drug clearance, can lead to problems such as poor patient compliance, side effects caused by high dose administration, and increased cost of treatment. In addition, rapidly metabolized drugs may also expose the patient to poorly toxic or reactive metabolites.
The discovery of novel potent SMN2 gene splice modulators with good oral bioavailability and patentability is a challenging task. Accordingly, there remains a need in the art to develop compounds having a higher selective inhibitory activity and/or better pharmacodynamics/pharmacokinetics for use as SMN2 gene splice modulators, and the present invention provides such compounds.
Disclosure of Invention
Aiming at the technical problems, the invention discloses a novel deuterium-substituted pyridopyrimidinone compound which is used as a novel and effective SMN2 gene splicing regulator, can increase functional SMN protein and is used for treating SMA. Furthermore, the compounds of the invention also show better metabolic stability and/or pharmacokinetic properties.
In this regard, the present invention adopts the following technical scheme:
in a first aspect of the invention, there is provided a compound of formula (I):
Wherein,
R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15 And R 16 are each independently selected from hydrogen, deuterium, halogen, or trifluoromethyl;
X 1 and X 2 are each independently selected from CH 3、CD3、CHD2 or CH 2 D;
with the proviso that said compound contains at least one deuterium atom;
Or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystalline form, hydrate, or solvate thereof.
In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystalline form, hydrate, or solvate thereof, and a pharmaceutically acceptable excipient. In a specific embodiment, the compounds of the present invention are provided in the pharmaceutical composition in an effective amount. In particular embodiments, the compounds of the present invention are provided in a therapeutically effective amount. In particular embodiments, the compounds of the present invention are provided in a prophylactically effective amount.
In another aspect, the present invention provides a method of preparing a pharmaceutical composition as described above, comprising the steps of: a pharmaceutically acceptable excipient is admixed with a compound of the invention or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystal form, hydrate, or solvate thereof, thereby forming a pharmaceutical composition.
In another aspect, the invention provides the use of a compound of the invention, or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystal form, hydrate, or solvate thereof, or a pharmaceutical composition as described above, in the manufacture of a medicament for the treatment and/or prophylaxis of Spinal Muscular Atrophy (SMA) diseases.
In another aspect, the present invention provides a method of treating and/or preventing Spinal Muscular Atrophy (SMA) disease, the method comprising administering to a subject in need thereof a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystalline form, hydrate, or solvate thereof, or a pharmaceutical composition of the invention.
Other objects and advantages of the present invention will be apparent to those skilled in the art from the detailed description, examples and claims that follow.
Definition of the definition
Herein, "deuterated" refers to a compound or group in which one or more hydrogens are replaced with deuterium, unless otherwise indicated; deuteration may be mono-, di-, poly-or full-substituted. The term "one or more deuterated" is used interchangeably with "one or more deuterated".
Herein, unless otherwise specified, "non-deuterated compound" refers to a compound having a deuterium atom content of not higher than the natural deuterium isotope content (0.015%).
As used herein, the term "subject" includes, but is not limited to: a human (i.e., male or female of any age group, e.g., pediatric subjects (e.g., infants, children, adolescents) or adult subjects (e.g., young adults, middle aged adults, or senior adults)) and/or a non-human animal, e.g., a mammal, e.g., a primate (e.g., cynomolgus monkey, rhesus monkey), cow, pig, horse, sheep, goat, rodent, cat, and/or dog. In some embodiments, the subject is a human. In other embodiments, the subject is a non-human animal.
"Disease," "disorder," and "condition" are used interchangeably herein.
As used herein, unless otherwise indicated, the term "treating" includes an effect that occurs when a subject suffers from a particular disease, disorder or condition, which reduces the severity of the disease, disorder or condition, or delays or slows the progression of the disease, disorder or condition ("therapeutic treatment"), as well as an effect that occurs before the subject begins to suffer from the particular disease, disorder or condition ("prophylactic treatment").
In general, an "effective amount" of a compound refers to an amount sufficient to elicit a biological response of interest. As will be appreciated by those of ordinary skill in the art, the effective amount of the compounds of the present invention may vary depending on the following factors: for example, biological targets, pharmacokinetics of the compound, the disease being treated, the mode of administration, and the age health and symptoms of the subject. Effective amounts include therapeutically and prophylactically therapeutically effective amounts.
As used herein, unless otherwise indicated, a "therapeutically effective amount" of a compound is an amount sufficient to provide a therapeutic benefit in the treatment of a disease, disorder, or condition, or to delay or minimize one or more symptoms associated with a disease, disorder, or condition. A therapeutically effective amount of a compound refers to the amount of therapeutic agent, alone or in combination with other therapies, that provides a therapeutic benefit in the treatment of a disease, disorder or condition. The term "therapeutically effective amount" may include an amount that improves overall treatment, reduces or avoids symptoms or causes of a disease or disorder, or enhances the therapeutic efficacy of other therapeutic agents.
As used herein, unless otherwise indicated, a "prophylactically effective amount" of a compound is an amount sufficient to prevent a disease, disorder, or condition, or to prevent one or more symptoms associated with a disease, disorder, or condition, or to prevent recurrence of a disease, disorder, or condition. A prophylactically effective amount of a compound refers to the amount of therapeutic agent used alone or in combination with other agents, which provides a prophylactic benefit in preventing a disease, disorder or condition. The term "prophylactically effective amount" may include an amount that improves overall prophylaxis, or an amount that enhances the prophylactic efficacy of other prophylactic agents.
"Combination" and related terms refer to the simultaneous or sequential administration of the therapeutic agents of the present invention. For example, the compounds of the invention may be administered simultaneously or sequentially in separate unit dosage forms with another therapeutic agent, or simultaneously in a single unit dosage form with another therapeutic agent.
Detailed Description
Compounds of formula (I)
Herein, "the compound of the present invention" refers to a compound of the following formula (I), or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystal form, hydrate, or solvent compound thereof.
In one embodiment, the present invention relates to compounds of formula (I):
Wherein,
R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15 And R 16 are each independently selected from hydrogen, deuterium, halogen, or trifluoromethyl;
X 1 and X 2 are each independently selected from CH 3、CD3、CHD2 or CH 2 D;
with the proviso that said compound contains at least one deuterium atom;
Or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystalline form, hydrate, or solvate thereof.
In a specific embodiment, the deuterium isotope content of deuterium at the deuterated position is at least greater than 0.015%, preferably greater than 30%, more preferably greater than 50%, more preferably greater than 55%, more preferably greater than 60%, more preferably greater than 65%, more preferably greater than 70%, more preferably greater than 75%, more preferably greater than 80%, more preferably greater than 85%, more preferably greater than 90%, more preferably greater than 95%, more preferably greater than 99% of the natural deuterium isotope content.
Specifically, the deuterium isotope content of each deuterated position of R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15、R16、X1 and X 2 in the present invention is at least 0.015%, more preferably greater than 1%, more preferably greater than 5%, more preferably greater than 10%, more preferably greater than 15%, more preferably greater than 20%, more preferably greater than 25%, more preferably greater than 30%, more preferably greater than 35%, more preferably greater than 40%, more preferably greater than 45%, more preferably greater than 50%, more preferably greater than 55%, more preferably greater than 60%, more preferably greater than 65%, more preferably greater than 70%, more preferably greater than 75%, more preferably greater than 80%, more preferably greater than 85%, more preferably greater than 90%, more preferably greater than 95%, more preferably greater than 99%.
In another embodiment, the compounds of the present invention contain at least one deuterium atom, more preferably two deuterium atoms, more preferably three deuterium atoms, more preferably four deuterium atoms, more preferably five deuterium atoms, more preferably six deuterium atoms, more preferably seven deuterium atoms, more preferably eight deuterium atoms, more preferably nine deuterium atoms, more preferably ten deuterium atoms, more preferably eleven deuterium atoms, more preferably twelve deuterium atoms, more preferably thirteen deuterium atoms, more preferably fourteen deuterium atoms, more preferably fifteen deuterium atoms, more preferably sixteen deuterium atoms, more preferably seventeen deuterium atoms, more preferably nineteen deuterium atoms, more preferably twenty deuterium atoms.
In another specific embodiment ,"R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 are each independently selected from hydrogen, deuterium, halogen or trifluoromethyl "including the technical scheme that R 1 is selected from hydrogen, deuterium, halogen or trifluoromethyl, R 2 is selected from hydrogen, deuterium, halogen or trifluoromethyl, R 3 is selected from hydrogen, deuterium, halogen or trifluoromethyl, and so on, until R 16 is selected from hydrogen, deuterium, halogen or trifluoromethyl. More specifically, the technical scheme includes that R 1 is hydrogen, R 1 is deuterium, R 1 is halogen (F, cl, br or I) or R 1 is trifluoromethyl, R 2 is hydrogen, R 2 is deuterium, R 2 is halogen (F, cl, br or I) or R 2 is trifluoromethyl, R 3 is hydrogen, R 3 is deuterium, R 3 is halogen (F, cl, br or I) or R 3 is trifluoromethyl, and so on until R 16 is hydrogen, R 16 is deuterium, R 16 is halogen (F, cl, br or I) or R 16 is trifluoromethyl.
In another specific embodiment, "X 1 and X 2 are each independently selected from CH 3、CD3、CHD2 or CH 2 D" includes embodiments in which X 1 is selected from CH 3、CD3、CHD2 or CH 2 D, and X 2 is selected from CH 3、CD3、CHD2 or CH 2 D. More specifically, the technical scheme includes that X 1 is CH 3、X1, CD 3、X1 is CHD 2 or X 1 is CH 2 D, and X 2 is CH 3、X2, CD 3、X2 is CHD 2 or X 2 is CH 2 D.
In another specific embodiment ,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 are each independently selected from hydrogen or deuterium.
In another specific embodiment, R 1、R2、R3、R4、R5 and R 6 are hydrogen.
In another specific embodiment, R 7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 are hydrogen.
In another specific embodiment, X 1 is CD 3 or CH 3.
In another specific embodiment, X 1 is CD 3.
In another specific embodiment, X 2 is CD 3 or CH 3.
In another specific embodiment, X 2 is CD 3.
In another specific embodiment, X 1 and X 2 are CD 3.
In some embodiments of the compounds of formula (I), preferably, the present invention relates to the above-described compounds, or pharmaceutically acceptable salts, tautomers, stereoisomers, prodrugs, crystalline forms, hydrates or solvates thereof, wherein X 1 is CD3,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15、R16 and X 2 are as defined above.
In some embodiments of the compounds of formula (I), preferably, the present invention relates to the above compound, or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystal form, hydrate, or solvent compound thereof, wherein X 1 is CD3,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 are each independently selected from hydrogen or deuterium, and X 2 is selected from CH 3、CD3、CHD2 or CH 2 D.
In some embodiments of the compounds of formula (I), preferably, the present invention relates to the above compound, or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystal form, hydrate, or solvent compound thereof, wherein X 1 is CD 3,R1、R2、R3、R4、R5 and R 6 is hydrogen, R 7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 are each independently selected from hydrogen or deuterium, and X 2 is selected from CH 3、CD3、CHD2 or CH 2 D.
In some embodiments of the compounds of formula (I), preferably, the present invention relates to the above compound, or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystal form, hydrate, or solvate thereof, wherein X 1 is CD3,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 is hydrogen, and X 2 is selected from CH 3、CD3、CHD2 or CH 2 D.
In some embodiments of the compounds of formula (I), preferably, the present invention relates to the above-described compounds, or pharmaceutically acceptable salts, tautomers, stereoisomers, prodrugs, crystalline forms, hydrates or solvates thereof, wherein X 2 is CD3,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15、R16 and X 1 are as defined above.
In some embodiments of the compounds of formula (I), preferably, the present invention relates to the above compound, or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystal form, hydrate, or solvent compound thereof, wherein X 2 is CD3,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 are each independently selected from hydrogen or deuterium, and X 1 is selected from CH 3、CD3、CHD2 or CH 2 D.
In some embodiments of the compounds of formula (I), preferably, the present invention relates to the above compound, or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystal form, hydrate, or solvent compound thereof, wherein X 2 is CD 3,R1、R2、R3、R4、R5 and R 6 is hydrogen, R 7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 are each independently selected from hydrogen or deuterium, and X 1 is selected from CH 3、CD3、CHD2 or CH 2 D.
In some embodiments of the compounds of formula (I), preferably, the present invention relates to the above compound, or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystal form, hydrate, or solvate thereof, wherein X 2 is CD3,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 is hydrogen, and X 1 is selected from CH 3、CD3、CHD2 or CH 2 D.
In some embodiments of the compounds of formula (I), preferably, the present invention relates to the above-described compounds, or pharmaceutically acceptable salts, tautomers, stereoisomers, prodrugs, crystalline forms, hydrates or solvates thereof, wherein X 1 and X 2 are CD3,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 are as defined above.
In some embodiments of the compounds of formula (I), preferably, the present invention relates to the above-described compounds, or pharmaceutically acceptable salts, tautomers, stereoisomers, prodrugs, crystalline forms, hydrates, or solvates thereof, wherein X 1 and X 2 are CD3,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 are each independently selected from hydrogen or deuterium.
In some embodiments of the compounds of formula (I), preferably, the present invention relates to the above-described compounds, or pharmaceutically acceptable salts, tautomers, stereoisomers, prodrugs, crystalline forms, hydrates, or solvates thereof, wherein X 1 and X 2 are CD 3,R1、R2、R3、R4、R5 and R 6 are hydrogen, R 7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 are each independently selected from hydrogen or deuterium.
In some embodiments of the compounds of formula (I), preferably, the present invention relates to the above-described compounds, or pharmaceutically acceptable salts, tautomers, stereoisomers, prodrugs, crystalline forms, hydrates, or solvates thereof, wherein X 1 and X 2 are CD3,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 is hydrogen.
As a preferred embodiment of the present invention, the compound, or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystal form, hydrate, or solvate thereof, is selected from any one of the following compounds:
The compounds of the invention may include one or more asymmetric centers and thus may exist in a variety of stereoisomeric forms, for example, enantiomeric and/or diastereomeric forms. For example, the compounds of the invention may be individual enantiomers, diastereomers, or geometric isomers (e.g., cis and trans isomers), or may be in the form of mixtures of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomers. The isomers may be separated from the mixtures by methods known to those skilled in the art, including: chiral High Pressure Liquid Chromatography (HPLC), formation and crystallization of chiral salts; alternatively, preferred isomers may be prepared by asymmetric synthesis.
Those skilled in the art will appreciate that the organic compound may form a complex with a solvent in or from which it reacts or from which it precipitates or crystallizes. These complexes are referred to as "solvates". When the solvent is water, the complex is referred to as a "hydrate". The present invention encompasses all solvates of the compounds of the present invention.
The term "solvate" refers to a form of a compound or salt thereof that is bound to a solvent, typically formed by a solvolysis reaction. This physical association may include hydrogen bonding. Conventional solvents include water, methanol, ethanol, acetic acid, DMSO, THF, diethyl ether, and the like. The compounds described herein may be prepared, for example, in crystalline form, and may be solvated. Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric solvates and non-stoichiometric solvates. In some cases, the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid. "solvate" includes both solvates in solution and separable solvates. Representative solvates include hydrates, ethanolates and methanolates.
The term "hydrate" refers to a compound that binds to water. Generally, the ratio of the number of water molecules contained in a hydrate of a compound to the number of molecules of the compound in the hydrate is determined. Thus, the hydrates of the compounds may be represented by, for example, the general formula R x H 2 O, where R is the compound and x is a number greater than 0. A given compound may form more than one hydrate type, including, for example, monohydrate (x is 1), lower hydrate (x is a number greater than 0 and less than 1, e.g., hemihydrate (r.0.5H 2 O)), and polyhydrate (x is a number greater than 1, e.g., dihydrate (r.2h 2 O) and hexahydrate (r.6h 2 O)).
The compounds of the present invention may be in amorphous or crystalline form (polymorphs). Furthermore, the compounds of the present invention may exist in one or more crystalline forms. Accordingly, the present invention includes within its scope all amorphous or crystalline forms of the compounds of the present invention. The term "polymorph" refers to a crystalline form (or salt, hydrate or solvate thereof) of a compound of a particular crystal stacking arrangement. All polymorphs have the same elemental composition. Different crystalline forms typically have different X-ray diffraction patterns, infrared spectra, melting points, densities, hardness, crystal shapes, optoelectronic properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature, and other factors can lead to a crystalline form predominating. Various polymorphs of a compound can be prepared by crystallization under different conditions.
The invention also includes isotopically-labeled compounds, which are identical to those of the compounds of the present invention, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, and chlorine, such as 2H、3H、13C、11C、14C、15N、18O、17O、31P、32P、35S、18F and 36 Cl, respectively. The compounds of the invention, prodrugs thereof, and pharmaceutically acceptable salts of the compounds or prodrugs thereof, which contain the isotopes described above and/or other isotopes of other atoms, are within the scope of this invention. Certain isotopically-labeled compounds of the present invention, for example those into which radioactive isotopes (e.g., 3 H and 14 C) are introduced, are useful in drug and/or substrate tissue distribution assays. Tritium, i.e., 3 H, and carbon-14, i.e., 14 C isotopes are particularly preferred because they are easy to prepare and detect. Further, substitution with heavier isotopes, such as deuterium, i.e., 2 H, may be preferred in some circumstances because greater metabolic stability may afford therapeutic benefits such as increased in vivo half-life or reduced dosage requirements. Isotopically-labeled compounds of formula (I) of the present invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes and/or examples and preparations below by substituting a readily available isotopically-labeled reagent for a non-isotopically-labeled reagent.
In addition, prodrugs are also included within the context of the present invention. The term "prodrug" as used herein refers to a compound that is converted in vivo by hydrolysis, e.g. in blood, into its active form having a medical effect. Pharmaceutically acceptable prodrugs are described in t.higuchi and v.stilla, prodrugs as Novel DELIVERY SYSTEMS, A.C.S.SYMPOSIUM Series Vol.14,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987, and d.fleisher, s.ramon and H.Barbra"Improved oral drug delivery:solubility limitations overcome by the use of prodrugs",Advanced Drug Delivery Reviews(1996)19(2)115-130, each of which are incorporated herein by reference.
Prodrugs are any covalently bonded compounds of the invention which, when administered to a patient, release the parent compound in vivo. Prodrugs are typically prepared by modifying functional groups in such a way that the modification may be performed by conventional procedures or cleavage in vivo to yield the parent compound. Prodrugs include, for example, compounds of the invention wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that, when administered to a patient, may cleave to form the hydroxy, amino, or sulfhydryl group. Representative examples of prodrugs therefore include, but are not limited to, acetate, formate and benzoate/amide derivatives of hydroxy, mercapto and amino functional groups of compounds of formula (I). In addition, in the case of carboxylic acid (-COOH), esters such as methyl ester, ethyl ester, and the like can be used. The esters themselves may be active and/or may be hydrolysed under in vivo conditions in the human body. Suitable pharmaceutically acceptable in vivo hydrolysable ester groups include those groups which readily decompose in the human body to release the parent acid or salt thereof.
Process for preparing compounds of the invention
The compounds of the present invention (including salts thereof) may be prepared using known organic synthesis techniques and may be synthesized according to any of a number of possible synthetic routes, such as those in the schemes below. The reaction for preparing the compounds of the present invention may be carried out in a suitable solvent, which may be readily selected by those skilled in the art of organic synthesis. Suitable solvents may be substantially unreactive with the starting materials (reactants), intermediates, or products at the temperature at which the reaction is carried out (e.g., at a temperature in the range of the solvent freezing temperature to the solvent boiling temperature). The given reaction may be carried out in one solvent or a mixture of more than one solvent. The skilled artisan can select the solvent for a particular reaction step depending on the particular reaction step.
The preparation of the compounds of the invention may involve protection and deprotection of different chemical groups. One skilled in the art can readily determine whether protection is desired and removal of the protection and selection of the appropriate protecting group. The chemical nature of the protecting groups can be found, for example, in Wuts and Greene, protective Groups in Organic Synthesis, 4 th edition, john Wiley & Sons: new Jersey, (2006), which is incorporated herein by reference in its entirety.
The compounds of the present invention can be prepared as individual stereoisomers thereof by reacting a racemic mixture of the compounds with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereoisomers and recovering the optically pure enantiomer. Enantiomeric resolution may be carried out using diastereomeric derivatives of the compounds of the invention, preferably dissociable complexes (e.g., crystalline diastereomeric salts). Diastereomers have significantly different physical properties (e.g., melting point, boiling point, solubility, reactivity, etc.), and can be readily separated by the advantages of these dissimilarities. Diastereomers may be separated by chromatography, preferably by separation/resolution techniques based on differences in solubility. The optically pure enantiomer is then recovered by any practical means that does not racemize, along with the resolving agent. A more detailed description of techniques suitable for resolution of stereoisomers of compounds starting from racemic mixtures can be found in Jean Jacques, andre Collet, samue1h.wilen, "enantiomers, racemates and resolution" ("Enantiomers, RACEMATES AND resolution"), john Wiley And Sons, inc.
The reaction may be monitored according to any suitable method known in the art. For example, product formation may be monitored by spectroscopic means, such as Nuclear Magnetic Resonance (NMR) spectroscopy (e.g., 1 H or 13 C), infrared (IR) spectroscopy, spectrophotometry (e.g., UV-visible), mass Spectrometry (MS)), or by chromatographic methods, such as High Performance Liquid Chromatography (HPLC) or Thin Layer Chromatography (TLC).
Pharmaceutical compositions, formulations and kits
In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention (also referred to as an "active ingredient") and a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition comprises an effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of an active ingredient. In some embodiments, the pharmaceutical composition comprises a prophylactically effective amount of the active ingredient.
Pharmaceutically acceptable excipients for use in the present invention refer to non-toxic carriers, adjuvants or vehicles that do not destroy the pharmacological activity of the co-formulated compounds. Pharmaceutically acceptable carriers, adjuvants or vehicles that can be used in the compositions of the invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (e.g., human serum albumin), buffer substances (e.g., phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (e.g., protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol, and lanolin.
The invention also includes kits (e.g., pharmaceutical packages). Kits provided can include a compound of the invention, other therapeutic agent, and first and second containers (e.g., vials, ampoules, bottles, syringes, and/or dispersible packages or other suitable containers) containing a compound of the invention, other therapeutic agent. In some embodiments, the provided kits may also optionally include a third container containing pharmaceutically acceptable excipients for diluting or suspending the compounds of the invention and/or other therapeutic agents. In some embodiments, the compounds of the invention and other therapeutic agents provided in the first and second containers are combined to form one unit dosage form.
The pharmaceutical compositions provided herein may be administered by a number of routes including, but not limited to: oral, parenteral, inhalation, topical, rectal, nasal, buccal, vaginal, by implantation or other means of administration. For example, parenteral administration as used herein includes subcutaneous, intradermal, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intramuscularly, and intracranial injection or infusion techniques.
Typically, an effective amount of a compound provided herein is administered. The amount of the compound actually administered may be determined by a physician, according to the circumstances involved, including the condition being treated, the route of administration selected, the compound actually administered, the age, weight and response of the individual patient, the severity of the patient's symptoms, and the like.
When used to prevent a disorder of the present invention, a subject at risk of developing the disorder is administered a compound provided herein, typically based on physician recommendations and administered under the supervision of a physician, at a dosage level as described above. Subjects at risk for developing a particular disorder generally include subjects having a family history of the disorder, or those subjects determined by genetic testing or screening to be particularly susceptible to developing the disorder.
The pharmaceutical compositions provided herein may also be administered chronically ("chronically"). Chronic administration refers to administration of a compound or pharmaceutical composition thereof over a prolonged period of time, e.g., 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or may continue administration indefinitely, e.g., for the remainder of the subject's life. In some embodiments, chronic administration is intended to provide a constant level of the compound in the blood over a prolonged period of time, e.g., within a therapeutic window.
Various methods of administration may be used to further deliver the pharmaceutical compositions of the present invention. For example, in some embodiments, the pharmaceutical composition may be administered as a bolus, e.g., in order to rapidly increase the concentration of the compound in the blood to an effective level. Bolus doses depend on the targeted systemic level of the active ingredient, e.g., intramuscular or subcutaneous bolus doses cause slow release of the active ingredient, whereas bolus injections delivered directly to veins (e.g., by IV intravenous drip) can be delivered more rapidly, causing the concentration of the active ingredient in the blood to rise rapidly to effective levels. In other embodiments, the pharmaceutical composition may be administered in the form of a continuous infusion, for example, by IV intravenous drip, thereby providing a steady state concentration of the active ingredient in the subject's body. Furthermore, in other embodiments, a bolus dose of the pharmaceutical composition may be administered first, followed by continuous infusion.
Oral compositions may take the form of bulk liquid solutions or suspensions or bulk powders. More typically, however, the compositions are provided in unit dosage form in order to facilitate accurate dosing. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active material suitable for producing the desired therapeutic effect in association with a suitable pharmaceutical excipient. Typical unit dosage forms include pre-filled, pre-measured ampoules or syringes of liquid compositions, or in the case of solid compositions, pills, tablets, capsules and the like. In such compositions, the compound is typically a minor component (about 0.1 to about 50 wt.%, or preferably about 1 to about 40 wt.%) with the remainder being various carriers or excipients and processing aids useful for forming the desired administration form.
For oral doses, a typical regimen is one to five oral doses per day, especially two to four oral doses, typically three oral doses. Using these modes of dosing, each dose provides from about 0.01 to about 20mg/kg of a compound of the invention, with preferred doses each providing from about 0.1 to about 10mg/kg, especially from about 1 to about 5mg/kg.
In order to provide similar blood levels to, or lower than, the use of an injected dose, a transdermal dose is typically selected in an amount of about 0.01 to about 20% by weight, preferably about 0.1 to about 10% by weight, and more preferably about 0.5 to about 15% by weight.
From about 1 to about 120 hours, especially 24 to 96 hours, the injection dosage level is in the range of about 0.1 mg/kg/hour to at least 10 mg/kg/hour. To achieve adequate steady state levels, a preloaded bolus of about 0.1mg/kg to about 10mg/kg or more may also be administered. For human patients of 40 to 80kg, the maximum total dose cannot exceed about 2 g/day.
Liquid forms suitable for oral administration may include suitable aqueous or nonaqueous carriers, buffers, suspending and dispersing agents, colorants, flavors, and the like. Solid forms may include, for example, any of the following components, or compounds having similar properties: binders, for example microcrystalline cellulose, gum tragacanth or gelatin; excipients, for example starch or lactose, disintegrants, for example alginic acid, primogel or corn starch; lubricants, for example, magnesium stearate; glidants, for example, colloidal silicon dioxide; sweeteners, for example, sucrose or saccharin; or a flavoring agent, for example, peppermint, methyl salicylate, or orange flavoring.
Injectable compositions are typically based on sterile saline or phosphate buffered saline for injectable use, or other injectable excipients known in the art. As previously mentioned, in such compositions, the active compound is typically a minor component, often about 0.05 to 10% by weight, the remainder being an injectable excipient or the like.
Transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient. When formulated as ointments, the active ingredients are typically combined with a paraffinic or a water-miscible ointment base. Alternatively, the active ingredient may be formulated as a cream with, for example, an oil-in-water cream base. Such transdermal formulations are well known in the art and typically include other components for enhancing stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and compositions are included within the scope provided by the present invention.
The compounds of the invention may also be administered via a transdermal device. Transdermal administration may thus be achieved using a reservoir (reservoir) or porous membrane type, or a variety of solid matrix patches.
The above components of the compositions for oral administration, injection or topical administration are merely representative. Other materials and processing techniques, etc. are set forth in Remington's Pharmaceutical Sciences, part 8 of 17th edition,1985,Mack Publishing Company,Easton,Pennsylvania, incorporated herein by reference.
The compounds of the present invention may also be administered in sustained release form, or from a sustained release delivery system. A description of representative sustained release materials can be found in Remington's Pharmaceutical Sciences.
The invention also relates to pharmaceutically acceptable formulations of the compounds of the invention. In one embodiment, the formulation comprises water. In another embodiment, the formulation comprises a cyclodextrin derivative. The most common cyclodextrins are α -, β -and γ -cyclodextrins consisting of 6, 7 and 8 α -1, 4-linked glucose units, respectively, optionally including one or more substituents on the linked sugar moiety, including but not limited to: methylated, hydroxyalkylated, acylated and sulfoalkyl ether substitutions. In some embodiments, the cyclodextrin is a sulfoalkyl ether β -cyclodextrin, e.g., sulfobutyl ether β -cyclodextrin, also known as Captisol. See, for example, U.S.5,376,645. In some embodiments, the formulation comprises hexapropyl- β -cyclodextrin (e.g., 10-50% in water).
Indication of disease
In another aspect, there is provided a compound of formula (I) as disclosed herein (including all individual embodiments and subsets of the classes disclosed herein) or a tautomer, stereoisomer, prodrug, crystal form, pharmaceutically acceptable salt, hydrate or solvate thereof, having valuable pharmacological properties and which is found to enhance the inclusion of exon 7 of SMN1 and/or SMN2 into mRNA transcribed from the SMN1 and/or SMN2 gene, thereby increasing the expression of SMN protein in a human subject in need thereof.
The compounds of the invention may be used alone or in combination with other drugs for the treatment or prevention of diseases caused by inactivating mutations or deletions of the SMN1 gene and/or associated with loss of function or deficiency of the SMN1 gene. These diseases include, but are not limited to, spinal Muscular Atrophy (SMA).
The compounds of the invention may be used alone or in combination with other drugs to treat or prevent Spinal Muscular Atrophy (SMA). Treating SMA includes one or more of the following effects: (1) reducing or improving the severity of SMA; (2) delaying the onset of SMA; (3) inhibiting SMA progression; (4) reducing admission of the subject; (5) reducing the length of time for admission to the subject; (6) increasing survival of the subject; (7) improving the quality of life of the subject; (8) reducing the number of SMA-related symptoms; (9) Reducing or ameliorating the severity of one or more symptoms associated with SMA; (9) inhibiting the development or onset of SMA symptoms; and/or (10) inhibit progression of symptoms associated with SMA.
Symptoms of SMA include muscle weakness, low muscle tension, crying weakness, cough weakness, lameness or tendency to fall, sucking or dysphagia, dyspnea, accumulation of secretions in the lungs or throat, fist held with a sweaty hand, tongue tremor/vibration, frequent tendency to one side of the head (even when lying down), tendency to be weaker than the legs of the arms, frequent legs in the "frog leg" position, difficulty feeding, increased sensitivity to respiratory tract infections, bowel/bladder weakness, less than normal body weight, inability to sit unsupported, inability to walk, inability to crawl, hypotonia, loss of reflex and associated multiple congenital contractures (joint contractures) of the anterior horn cell loss.
The compounds of the invention may be used alone or in combination with other drugs to treat SMA, with one or more of the following beneficial effects: (1) reducing muscle strength injury; (2) increasing muscle strength; (3) reducing muscle atrophy; (4) reducing loss of motor function; (5) increasing motor neurons; (6) reducing motor neuron loss; (7) protecting SMA-deficient motor neurons from degeneration; (8) enhancing athletic performance; (9) increase lung function; and/or (10) reduce loss of lung function.
Examples
The invention will be further illustrated with reference to specific examples. It is to be understood that these examples are illustrative of the present invention and are not intended to limit the scope of the present invention. The experimental methods, in which specific conditions are not noted in the following examples, are generally conducted under conventional conditions or under conditions recommended by the manufacturer. Parts and percentages are parts by weight and percentages by weight unless otherwise indicated.
Abbreviations:
Pd (dppf) Cl 2: [1,1' -bis (triphenylphosphine) ferrocene ] palladium dichloride
Pd (OAc) 2 palladium acetate
PCy 3: tricyclohexylphosphorus
TsCl: para-toluenesulfonyl chloride
KOAc: potassium acetate
Isopropyl acetate: acetic acid isopropyl ester
TFA: trifluoroacetic acid
DMSO: dimethyl sulfoxide
DMF: n, N-dimethylformamide
EA: acetic acid ethyl ester
Xylene: xylene (P)
MeOH: methanol
Dioxide:1, 4-Dioxahexacyclic ring
DCM: dichloromethane (dichloromethane)
Preparation of intermediate compound A-17- (4-oxo-2- (p-toluenesulfonyloxy) -4H-pyrido [1,2-a ] pyrimidin-7-yl) -4, 7-diazaspiro [2.5] octa-4-carboxylic acid tert-butyl ester
The following synthetic route was adopted
Step 1 Synthesis of the Compound 7- (6-nitropyridin-3-yl) -4, 7-diazaspiro [2.5] oct-4-carboxylic acid tert-butyl ester
5-Bromo-2-nitropyridine (2.0 g,10 mmol) and tert-butyl 4, 7-diazaspiro [2.5] octa-4-carboxylate (2.23 g,10.5 mmol) were dissolved in 10ml dimethyl sulfoxide, lithium chloride (1.47 g,35 mmol) and tetramethylguanidine (4.4 ml,35 mmol) were added sequentially, the reaction solution was heated to 80℃under nitrogen protection and stirred overnight, TLC monitoring was completed, cooled to room temperature, the reaction solution was slowly added dropwise to ice water, a pale yellow solid was precipitated, filtered, and the filter cake was dried in vacuo to give a yellow solid 2.76g, yield 82.6%. LC-MS (APCI): M/z=335.1 (m+1) +.
Step 2 Synthesis of the Compound 7- (6-aminopyridin-3-yl) -4, 7-diazaspiro [2.5] octa-4-carboxylic acid tert-butyl ester
7- (6-Nitropyridin-3-yl) -4, 7-diazaspiro [2.5] octane-4-carboxylic acid tert-butyl ester (2.76 g,8.26 mmol) obtained in the previous step was placed in a 100ml flask, 10ml ethyl acetate and 10ml methanol were added for dissolution, 10% wt Pd/C (300 mg) was added under nitrogen protection, three times replaced with hydrogen, stirred at room temperature for 2-4 hours, TLC monitored the reaction was complete, celite was used for filtration, and the filtrate was concentrated to dryness. 2.55g of crude product was obtained and was directly fed to the next reaction without purification. LC-MS (APCI): M/z=305.4 (m+1) +.
Step 3 Synthesis of the Compound 7- (2-hydroxy-4-oxo-4H-pyrido [1,2-a ] pyrimidin-7-yl) -4, 7-diazaspiro [2.5] octane-4-carboxylic acid tert-butyl ester
7- (6-Aminopyridin-3-yl) -4, 7-diazaspiro [2.5] octyl-4-carboxylate (2.0 g,6.58 mmol) and bis (2, 4, 6-trichlorophenyl) malonate (3.66 g,7.89 mmol) were added to the reaction flask, the reaction mixture was heated to 150℃with the addition of 25ml of xylene and stirred for 1-2 hours under nitrogen protection, TLC monitoring was completed, cooling to room temperature to precipitate a pale yellow solid, filtration, washing the filter cake with a small amount of ethyl acetate, and vacuum drying gave 1.92g of solid, yield 78.4%. LC-MS (APCI) M/z=373.4 (M+1) +.
Step 4 Synthesis of intermediate Compound A-1
7- (2-Hydroxy-4-oxo-4H-pyrido [1,2-a ] pyrimidin-7-yl) -4, 7-diazaspiro [2.5] octane-4-carboxylic acid tert-butyl ester (1.12 g,3.0 mmol) was dissolved in 40ml of anhydrous dichloromethane, triethylamine (0.54 ml,3.9 mmol) was added thereto under nitrogen protection, tsCl (0.63 g,3.3 mmol) was added thereto in portions, and the reaction was stirred at room temperature for 2 to 4 hours after the addition, and the completion of the reaction was monitored by TLC. 30ml of water is added for quenching reaction, dichloromethane is used for extraction for 3-4 times, the organic phases are combined, saturated saline water is used for washing, anhydrous sodium sulfate is used for drying, filtration is carried out, filtrate is concentrated, and silica gel column chromatography is used for purifying to obtain 1.4g of product, the yield is: 88.7%. LC-MS (APCI): M/z= 527.8 (m+1) +.
Preparation of intermediate B-1 2-methyl-8- (methyl-d 3) -6- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) imidazo [1,2-B ] pyridazine
The following synthetic route was adopted
Step 1 Synthesis of the Compound 4-bromo-6-chloropyridazine-3-amine
6-Chloropyridazine-3-ammonia (4.0 g,31 mmol), sodium acetate (2.03 g,24.8 mmol) and acetic acid (0.5 g,8.27 mmol) were placed in a 100ml flask, 40ml methanol was added, 1, 3-dibromo-5, 5-dimethylhydantoin (5.3 g,18.5 mmol) was added in portions, stirred overnight at room temperature, TLC monitored, cooled to 0 ℃, quenched by slowly adding 10ml of saturated aqueous sodium sulfite solution, concentrated to remove methanol, diluted with a small amount of water, adjusted to pH 7-8 with 1N sodium hydroxide, a pale yellow solid was precipitated, filtered, the filter cake was washed with water, and 4.5g of the target product was obtained after vacuum drying, yield: 70.1% of the reaction mixture was directly fed to the next reaction without purification. LC-MS (APCI): M/z=208.1 (m+1) +.
Step 2 Synthesis of 6-chloro-4- (methyl-d 3) pyridazine-3-amine as Compound
4-Bromo-6-chloropyridazine-3-ammonia (2.07 g,10 mmol) obtained in the above step was dissolved in 100ml of anhydrous THF, deuterated methyl magnesium iodide (27 ml,27 mmol) was slowly added dropwise under nitrogen protection, a solution of zinc chloride in 2-methyltetrahydrofuran (2.5 ml,5 mmol) and tetrakis (triphenylphosphine) palladium (116 mg,0.1 mmol) were sequentially added after the addition, and the reaction was stirred for 4-6 hours at 50℃with heating to complete the TLC. The reaction solution was cooled to room temperature, quenched by adding 50ml of saturated ammonium chloride, extracted 3-4 times with ethyl acetate, combined with organic phases, concentrated and purified by silica gel column chromatography to obtain 1.34g of the product, yield: 44.3%. LC-MS (APCI) M/z=147.5 (M+1) +.
Step 3 Synthesis of the Compound 6-chloro-2-methyl-8- (methyl-d 3) imidazo [1,2-b ] pyridazine
6-Chloro-4- (methyl-d 3) pyridazine-3-ammonia (194 mg,1.33 mmol) was dissolved in 10ml anhydrous DMF, heated to 100deg.C, and bromoacetone (200 mg,1.46 mmol) was slowly added dropwise to 3ml DMF solution, and the reaction was stirred for 2-3 hours at 110deg.C under nitrogen protection, and TLC monitoring the reaction was complete. After cooling to room temperature, the reaction was quenched by adding 10ml of saturated aqueous sodium bicarbonate, extracted 3-4 times with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to give 140mg of solid in 57.1% yield. LC-MS (APCI): M/z=185.5 (m+1) +.
Step 4 Synthesis of intermediate Compound B-1
6-Chloro-2-methyl-8- (methyl-d 3) imidazo [1,2-b ] pyridazine (90 mg,0.5 mmol), pinacol biborate (380 mg,1.5 mmol), pd (OAc) 2(6mg,0.025mmol)、PCy3 (14 mg,0.05 mmol) and potassium acetate (100 mg,1.0 mmol) were added to a 20ml microwave tube, 8ml isopropyl acetate was added under nitrogen protection, the reaction was carried out after sealing by microwave heating to 80℃for 1 hour, TLC was monitored to complete the reaction, the solvent was removed by concentration after cooling to room temperature, and silica gel column chromatography was purified to give 108mg solid, yield: 79.1%. LC-MS (APCI): M/z=277.2 (m+1) +.
Preparation of intermediate B-28-methyl-2- (methyl-d 3) -6- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) imidazo [1,2-B ] pyridazine
The following synthetic route was adopted
Step 1 Synthesis of Compound 1-bromopropane-2-one-1, 3-d 5
Deuterated acetone (5 g,78.1 mmol) was added to 100ml methanol, cooled to 0 ℃, and a dioxane complex of bromine (19.4 g,78.1 mmol) was added in portions, stirred under ice bath for 3-4 hours, after the reaction was monitored by GC, 80ml water was added for quenching reaction, 80ml diethyl ether was used for extraction 3-4 times, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated at low temperature to remove the solvent to obtain crude product, which was directly put into the next reaction without purification.
Step 2: synthesis of 6-chloro-4-methylpyridazine-3-ammonia as Compound
1-Bromopropane-2-one-1, 3-d 5 (2.07 g,10 mmol) was dissolved in 100ml anhydrous THF, methyl magnesium iodide (27 ml,27 mmol) was slowly added dropwise under nitrogen protection, a solution of zinc chloride in 2-methyltetrahydrofuran (2.5 ml,5 mmol) and tetrakis (triphenylphosphine) palladium (116 mg,0.1 mmol) were sequentially added after the addition, the temperature was raised to 50℃and the reaction was stirred for 4-6 hours, and TLC was monitored to complete the reaction. The reaction solution was cooled to room temperature, quenched by adding 50ml of saturated ammonium chloride, extracted 3-4 times with ethyl acetate, combined with organic phases, concentrated and purified by silica gel column chromatography to obtain 1.18g of the product, yield: 39.0%. LC-MS (APCI): M/z=144.2 (m+1) +.
Step 3: synthesis of Compound 6-chloro-8-methyl-2- (methyl-d 3) imidazo [1,2-b ] pyridazine
6-Chloro-4-methylpyridazine-3-ammonia (194 mg,1.34 mmol) was dissolved in 10ml of anhydrous DMF, heated to 100deg.C, 3ml of DMF solution of 1-bromopropane-2-one-1, 3-d 5 (200 mg,1.40 mmol) was slowly added dropwise, and the reaction was stirred for 2-3 hours while heating to 110deg.C under nitrogen protection, and TLC monitored the reaction. After cooling to room temperature, the reaction was quenched by adding 10ml of saturated aqueous sodium bicarbonate, extracted 3-4 times with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to give 152mg of solid in 61.9% yield. LC-MS (APCI): M/z=185.5 (m+1) +.
Step 4: synthesis of intermediate compound B-2
6-Chloro-8-methyl-2- (methyl-d 3) imidazo [1,2-b ] pyridazine (90 mg,0.5 mmol), pinacol biborate (380 mg,1.5 mmol), pd (OAc) 2(6mg,0.025mmol)、PCy3 (14 mg,0.05 mmol) and potassium acetate (100 mg,1.0 mmol) were added to a 20ml microwave tube, 8ml isopropyl acetate was added under nitrogen protection, the reaction was carried out after sealing by microwave heating to 80℃for 1 hour, TLC was monitored to complete the reaction, the solvent was removed by concentration after cooling to room temperature, and silica gel column chromatography was purified to give 111mg solid, yield: 81.3 percent. LC-MS (APCI): M/z=277.2 (m+1) +.
Preparation of intermediate compound B-2, 8-bis (methyl-d 3) -6- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) imidazo [1,2-B ] pyridazine
The following synthetic route was adopted
Step 1 Synthesis of 6-chloro-4- (methyl-d 3) pyridazine-3-amine as Compound
4-Bromo-6-chloropyridazine-3-ammonia (2.07 g,10 mmol) was dissolved in 100ml of anhydrous THF, deuterated methyl magnesium iodide (27 ml,27 mmol) was slowly added dropwise under nitrogen protection, and after the addition, zinc chloride in 2-methyltetrahydrofuran (2.5 ml,5 mmol) and tetrakis (triphenylphosphine) palladium (116 mg,0.1 mmol) were sequentially added, and the temperature was raised to 50℃and stirred for 4-6 hours, after which the reaction was monitored by TLC. The reaction solution was cooled to room temperature, quenched by adding 50ml of saturated ammonium chloride, extracted 3-4 times with ethyl acetate, combined with organic phases, concentrated and purified by silica gel column chromatography to obtain 1.32g of the product, yield: 43.6%. LC-MS (APCI): M/z=144.2 (m+1) +.
Step 2 Synthesis of the Compound 6-chloro-2, 8-bis (methyl-d 3) imidazo [1,2-b ] pyridazine
6-Chloro-4- (methyl-d 3) pyridazine-3-ammonia (194 mg,1.32 mmol) was dissolved in 10ml anhydrous DMF, heated to 100deg.C, 3ml DMF solution of 1-bromopropane-2-one-1, 3-d 5 (200 mg,1.40 mmol) was slowly added dropwise, the temperature was raised to 110deg.C under nitrogen protection, stirred for 2-3 hours, and TLC monitored the reaction was completed. After cooling to room temperature, the reaction was quenched by adding 10ml of saturated aqueous sodium bicarbonate, extracted 3-4 times with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and separated by a silica gel column to give 163mg of solid with a yield of 66.4%. LC-MS (APCI) M/z=188.5 (M+1) +.
Step 3 Synthesis of intermediate Compound B-3
6-Chloro-2, 8-bis (methyl-d 3) imidazo [1,2-b ] pyridazine (90 mg,0.5 mmol), pinacol biborate (380 mg,1.5 mmol), pd (OAc) 2(6mg,0.025mmol)、PCy3 (14 mg,0.05 mmol) and potassium acetate (100 mg,1.0 mmol) were added to a 20ml microwave tube, 8ml isopropyl acetate was added under nitrogen protection, the reaction was carried out after sealing by microwave heating to 80℃for 1 hour, TLC was monitored to complete the reaction, the solvent was removed by concentration after cooling to room temperature, and silica gel column chromatography was purified to give 95mg solid, yield: 69.6%. LC-MS (APCI): M/z=280.2 (m+1) +.
Example 1 7- (4, 7-diazaspiro [2.5] oct-7-yl) -2- (2-methyl-8- (methyl-d 3) imidazo [1,2-b ] pyridazin-6-yl) pyrido [1,2-a ] pyrimidin-4-one (Compound T-1) preparation
The following synthetic route was adopted
Step 1 Synthesis of the Compound 7- (2- (2-methyl-8- (methyl-d 3) imidazo [1,2-b ] pyridazin-6-yl) -4-oxopyrido [1,2-a ] pyrimidin-7-yl) -4, 7-diazaspiro [2.5] octa-4-carboxylic acid tert-butyl ester
Intermediate compound a-1 (90 mg,0.17 mmol) and intermediate compound B-1 (52 mg,0.19 mmol) were added to 10ml acetonitrile and 2ml water, pd (dppf) Cl 2 (12 mg,0.017 mmol) and potassium carbonate (47 mg,0.34 mmol) were added under nitrogen, heated to 80 ℃ and stirred overnight, after TLC monitoring the reaction, cooled to room temperature and concentrated to remove solvent, and purified by silica gel column chromatography to give 69mg of solid, yield: 81.1%. LC-MS (APCI): M/z=505.2 (m+1) +.
Step 2 Synthesis of Compound T-1
7- (2- (2-Methyl-8- (methyl-d 3) imidazo [1,2-b ] pyridazin-6-yl) -4-oxopyrido [1,2-a ] pyrimidin-7-yl) -4, 7-diazaspiro [2.5] oct-4-carboxylic acid tert-butyl ester (69 mg,0.14 mmol) obtained in the above step was added to 5ml of dichloromethane, TFA (0.2 ml,2.8 mmol) was added, the reaction was stirred at room temperature for 0.5-1 hour, TLC was monitored to complete the reaction, 10ml of dichloromethane was added to dilute, and the mixture was washed 2-3 times with 20ml of saturated aqueous sodium bicarbonate solution and saturated brine in sequence, and the organic phase was concentrated and purified by column chromatography on silica gel to give 48mg of off-white solid in yield :87.2%.LC-MS(APCI):m/z=405.5(M+1)+.1H NMR(400MHz,CDCl3)δ8.45(d,J=2.4Hz,1H),7.92(d,J=1.0Hz,1H),7.73(d,J=9.6Hz,1H),7.80(s,1H),7.70(dd,J=9.7,2.5Hz,1H),7.38(s,1H),3.31-3.22(m,2H),3.20-3.16(m,2H),3.08(s,2H),2.55(s,3H),1.68(br s,1H),0.77-0.75(m,2H),0.67-0.64(m,2H).
Example 2 7- (4, 7-diazaspiro [2.5] oct-7-yl) -2- (8-methyl-2- (methyl-d 3) imidazo [1,2-b ] pyridazin-6-yl) pyrido [1,2-a ] pyrimidin-4-one (Compound T-2) preparation
The following synthetic route was adopted
Step 1 Synthesis of the Compound 7- (2- (2- (methyl-d 3) -8-methylimidazo [1,2-b ] pyridazin-6-yl) -4-oxopyrido [1,2-a ] pyrimidin-7-yl) -4, 7-diazaspiro [2.5] octa-4-carboxylic acid tert-butyl ester
Intermediate compound a-1 (90 mg,0.17 mmol) and intermediate compound B-2 (52 mg,0.19 mmol) were added to 10ml acetonitrile and 2ml water, pd (dppf) Cl 2 (12 mg,0.017 mmol) and potassium carbonate (47 mg,0.34 mmol) were added under nitrogen, heated to 80 ℃ and stirred overnight, after TLC monitoring the reaction, cooled to room temperature and concentrated to remove solvent, and purified by silica gel column chromatography to give 74mg of solid, yield: 87.0%. LC-MS (APCI): M/z=505.2 (m+1) +.
Step 2 Synthesis of Compound T-2
7- (2- (2- (Methyl-d 3) -8-methylimidazo [1,2-b ] pyridazin-6-yl) -4-oxopyrido [1,2-a ] pyrimidin-7-yl) -4, 7-diazaspiro [2.5] octa-4-carboxylic acid tert-butyl ester (74 mg,0.15 mmol) obtained in the above step was added to 5ml of methylene chloride, TFA (0.2 ml,2.8 mmol) was added, stirred at room temperature for 0.5 to 1 hour, TLC was monitored to complete the reaction, 10ml of methylene chloride was added to dilute, and the mixture was washed 2 to 3 times with 20ml of saturated aqueous sodium bicarbonate solution and saturated brine in sequence, and the organic phase was concentrated and purified by silica gel column chromatography to give 51mg of an off-white solid in yield :92.6%.LC-MS(APCI):m/z=405.5(M+1)+.1H NMR(400MHz,CDCl3)δ8.46(d,J=2.4Hz,1H),7.94(d,J=1.0Hz,1H),7.73(d,J=9.6Hz,1H),7.81(s,1H),7.73(dd,J=9.7,2.5Hz,1H),7.38(s,1H),3.31-3.22(m,2H),3.20-3.16(m,2H),3.09(s,2H),2.74(d,J=0.9Hz,3H),1.69(br s,1H),0.77-0.75(m,2H),0.68-0.64(m,2H).
Example 3 7- (4, 7-diazaspiro [2.5] oct-7-yl) -2- (2, 8-bis (methyl-d 3) imidazo [1,2-b ] pyridazin-6-yl) pyrido [1,2-a ] pyrimidin-4-one (Compound T-3) preparation
The following synthetic route was adopted
Step 1 Synthesis of the Compound 7- (2, 8-bis (methyl-d 3) imidazo [1,2-b ] pyridazin-6-yl) -4-oxopyrido [1,2-a ] pyrimidin-7-yl) -4, 7-diazaspiro [2.5] octa-4-carboxylic acid tert-butyl ester
Intermediate compound a-1 (90 mg,0.17 mmol) and intermediate compound B-2 (52 mg,0.19 mmol) were added to 10ml acetonitrile and 2ml water, pd (dppf) Cl 2 (12 mg,0.017 mmol) and potassium carbonate (47 mg,0.34 mmol) were added under nitrogen protection, heated to 80 ℃ and stirred overnight for reaction, after TLC monitoring the reaction, cooled to room temperature and concentrated to remove solvent, silica gel column chromatography purification gave 58mg of solid, yield: 68.2%. LC-MS (APCI): M/z=508.2 (m+1) +.
Step 2 Synthesis of Compound T-3
7- (2, 8-Bis (methyl-d 3) imidazo [1,2-b ] pyridazin-6-yl) -4-oxopyrido [1,2-a ] pyrimidin-7-yl) -4, 7-diazaspiro [2.5] oct-4-carboxylic acid tert-butyl ester (58 mg,0.12 mmol) obtained in the above step was added to 5ml of dichloromethane, TFA (0.2 ml,2.8 mmol) was added, the reaction was stirred at room temperature for 0.5-1 hour, TLC was monitored to complete the reaction, 10ml of dichloromethane was added to dilute, and washing with 20ml of saturated aqueous sodium bicarbonate solution and saturated saline was sequentially carried out for 2-3 times, the organic phase was concentrated and purified by silica gel column chromatography to give 35mg of off-white solid in yield :63.5%.LC-MS(APCI):m/z=408.5(M+1)+.1H NMR(400MHz,CDCl3):δ8.45(d,J=2.4Hz,1H),7.93(d,J=1.0Hz,1H),7.74(d,J=9.6Hz,1H),7.81(s,1H),7.70(dd,J=9.7,2.5Hz,1H),7.39(s,1H),3.30-3.22(m,2H),3.20-3.16(m,2H),3.08(s,2H),1.69(br s,1H),0.78-0.75(m,2H),0.68-0.62(m,2H).
Biological activity testing.
(1) Evaluation of Metabolic stability
Metabolic stability is generally used to describe the rate and extent to which a compound is metabolized and is one of the primary factors affecting pharmacokinetic properties. Many compounds are substrates for CYP450 enzymes and other drug metabolizing enzymes, and liver microsomes are CYP 450-rich systems, and the aim of this experiment was to conduct in vitro stability studies by incubating the compounds of the invention with human and SD rat liver microsomes, respectively, and using LC-MS/MS to detect the remaining proportion of the compounds.
① Preparation of the solution
Phosphate Buffer (PBS): 150mL of a pre-prepared KH 2PO4 (0.5M) solution and 700mL of a K 2HPO4 (0.5M) solution were mixed, the pH of the mixture was adjusted to 7.4 with a K 2HPO4 (0.5M) solution, and the mixture was stored at 4℃as 5-fold concentration PBS for use. Before use, the solution was diluted 5-fold with ultrapure water, and 3.3mM magnesium chloride was added to obtain phosphate buffer PBS (100 mM).
NADPH regeneration system solution: NADPH solution containing 6.5mM NADP,16.5mM G-6-P,3U/mL G-6-P D was formulated with 5mL PBS.
Internal standard stop solution: propranolol hydrochloride (50 ng/mL) and tolbutamide (200 ng/mL) were prepared with acetonitrile as internal standard working solutions.
Human liver microsomal solution: 0.31mL of human liver microsome (25 mg/mL) was added to 0.961mL of PBS (pH 7.4) and mixed to obtain a human liver microsome dilution with a protein concentration of 0.625 mg/mL.
SD rat liver microsomal solution: 0.31mLSD rat liver microsomes (25 mg/mL) were added to 0.961mL PBS (pH 7.4) and mixed to obtain SD rat liver microsome dilution with a protein concentration of 0.625 mg/mL.
Sample working solution: the compound of the present invention and non-deuterated compound powder, positive control dextromethorphan powder and omeprazole powder were formulated with DMSO to 10mM as sample stock solutions. Then, the mixture was diluted with 70% acetonitrile-water to obtain a 0.25mM sample working solution.
② Sample incubation
398 Μl of human liver microsome dilution was added to 96 Kong Fuyo plates (n=2), and 2 μl of 0.25mM test compound and dextromethorphan were added, respectively, and mixed well.
398 Μl of SD rat liver microsome dilution was added to 96 Kong Fuyo plates (N=2), and 2 μl of 0.25mM test compound and omeprazole were added, respectively, and mixed well.
Each well was filled with 300. Mu.L of pre-chilled stop solution into a 96-well deep well plate and placed on ice as a stop plate.
The 96 Kong Fuyo plates and NADPH regeneration system were placed in a 37℃water bath, shaken at 100 revolutions per minute, and pre-incubated for 5min. 80. Mu.L of the incubation solution was removed from each well of the incubation plate, added to the termination plate, mixed well, and supplemented with 20. Mu.L of NADPH regeneration system solution as a 0min sample. Then 80. Mu.L of NADPH regeneration system solution was added to each well of the incubation plate, the reaction was started and timing was started. The reaction concentration of the compound to be tested is 1 mu M, and the protein concentration is 0.5mg/mL.
100. Mu.L of each reaction solution was added to the termination plate at 10, 30 and 90min, and the reaction was terminated by vortexing for 3 min.
The final plates were centrifuged at 5000rpm at 4℃for 15min. 200. Mu.L of the supernatant was mixed with a 96-well plate to which 200. Mu.L of ultrapure water had been added in advance, and the mixture was analyzed by LC-MS/MS to obtain a sample of 10. Mu.L.
③ Sample analysis method
The LC-MS/MS system is used in the experiment to detect the peak areas of the compound to be detected, dextromethorphan, omeprazole and an internal standard, and the ratio of the peak areas of the compound to the internal standard is calculated.
④ Data processing
The peak areas of the sample and the internal standard are obtained by a mass spectrometer and analysis software, and a single-exponential degradation model of GRAPHPAD PRISM 7.0.0 software is used for plotting the residual amount (R%) of the compound with time to obtain a substrate elimination rate constant K
Ct/C0=exp(-K*t)
And half-life T 1/2 and intrinsic clearance CL int were calculated according to the following formulas, where V/M is equal to 1/C (protein).
t1/2(min);CLint(μL/min/mg)。
Experimental results: the compounds of the present invention and their non-deuterated compounds were simultaneously tested and compared to evaluate their metabolic stability in human and SD rat liver microsomes. The compounds of the invention have a longer half-life T 1/2 and lower clearance CL int than non-deuterated compounds, which can significantly improve metabolic stability. The results of the compounds of the examples are summarized in Table 1 below.
TABLE 1
(2) Rat pharmacokinetic experiments
6 Male Sprague-Dawley rats, 7-8 weeks old, weighing about 210g, were divided into 2 groups of 3 animals each, and their pharmacokinetic differences were compared by intravenous or oral administration of a single dose of the compound (oral administration 10 mg/kg).
Rats were fed with standard feed and given water. Fasted food was started 16 hours prior to the trial. The drug was dissolved with 10mM ascorbic acid/0.01 mg/mL sodium thiosulfate pentahydrate. The eyebox was sampled at 0.083 hours, 0.25 hours, 0.5 hours, 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours and 24 hours post-administration.
Rats were briefly anesthetized after inhalation of diethyl ether and 300 μl of blood was collected from the orbit in a tube. There was 30. Mu.L of 1% heparin salt solution in the tube. Before use, the tube was baked overnight at 60 ℃. After blood collection was completed at the last time point, rats were sacrificed after ether anesthesia.
Immediately after blood sample collection, the tube was gently inverted at least 5 times, ensuring that the mix was well placed on ice. The blood sample was centrifuged at 5000rpm at 4℃for 5 minutes to separate the plasma from the erythrocytes. 100. Mu.L of plasma was aspirated with a pipette into a clean plastic centrifuge tube, indicating the name and time point of the compound. Plasma was stored at-80 ℃ prior to analysis. The concentration of the compounds of the invention in plasma was determined by LC-MS/MS. Pharmacokinetic parameters were calculated based on the blood concentration of each animal at different time points.
Experiments show that the compound has better pharmacokinetic properties in animals, thus having better pharmacodynamics and treatment effect. The results of the compounds of the examples are summarized in Table 2 below.
TABLE 2
The foregoing is a further detailed description of the invention in connection with the preferred embodiments, and it is not intended that the invention be limited to the specific embodiments described. It will be apparent to those skilled in the art that several simple deductions or substitutions may be made without departing from the spirit of the invention, and these should be considered to be within the scope of the invention.
Claims (11)
1. A compound of formula (I), or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystal form, hydrate, or solvate thereof:
Wherein,
R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15 And R 16 are each independently selected from hydrogen, deuterium, halogen, or trifluoromethyl;
X 1 and X 2 are each independently selected from CH 3、CD3、CHD2 or CH 2 D;
with the proviso that the above-mentioned compounds contain at least one deuterium atom.
2. The compound of claim 1, wherein ,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 are each independently selected from hydrogen or deuterium.
3. The compound of claim 1 or 2, wherein R 1、R2、R3、R4、R5 and R 6 are hydrogen.
4. A compound according to any one of claims 1-3, wherein R 7、R8、R9、R10、R11、R12、R13、R14、R15 and R 16 are hydrogen.
5. The compound of any one of claims 1-4, wherein X 1 is CD 3 or CH 3.
6. The compound of any one of claims 1-5, wherein X 1 is CD 3.
7. The compound of any one of claims 1-6, wherein X 2 is CD 3 or CH 3.
8. The compound of any one of claims 1-7, wherein X 2 is CD 3.
9. The compound of claim 1, or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystal form, hydrate, or solvate thereof, wherein the compound is selected from the group consisting of:
10. A pharmaceutical composition comprising a pharmaceutically acceptable excipient and a compound of any one of claims 1-9, or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystal, hydrate, or solvate thereof.
11. Use of a compound according to any one of claims 1-9, or a pharmaceutically acceptable salt, tautomer, stereoisomer, prodrug, crystalline form, hydrate, or solvate thereof, or a pharmaceutical composition according to claim 10, for the manufacture of a medicament for the treatment and/or prevention of Spinal Muscular Atrophy (SMA) disease.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202210403015 | 2022-04-18 | ||
CN2022104030157 | 2022-04-18 |
Publications (1)
Publication Number | Publication Date |
---|---|
CN118005654A true CN118005654A (en) | 2024-05-10 |
Family
ID=88419196
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202310418144.8A Pending CN118005654A (en) | 2022-04-18 | 2023-04-17 | Substituted pyridopyrimidinone compounds, compositions containing the same and uses thereof |
Country Status (2)
Country | Link |
---|---|
CN (1) | CN118005654A (en) |
WO (1) | WO2023202501A1 (en) |
Family Cites Families (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US10882868B2 (en) * | 2014-05-15 | 2021-01-05 | Hoffmann-La Roche Inc. | Compounds for treating spinal muscular atrophy |
WO2015173181A1 (en) * | 2014-05-15 | 2015-11-19 | F. Hoffmann-La Roche Ag | Compounds for treating spinal muscular atrophy |
MX2018005890A (en) * | 2015-11-12 | 2018-08-15 | Hoffmann La Roche | Compositions for treating spinal muscular atrophy. |
-
2023
- 2023-04-17 CN CN202310418144.8A patent/CN118005654A/en active Pending
- 2023-04-17 WO PCT/CN2023/088563 patent/WO2023202501A1/en unknown
Also Published As
Publication number | Publication date |
---|---|
WO2023202501A1 (en) | 2023-10-26 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN109574995B (en) | Substituted pyridazinone compounds | |
CN107428791B (en) | Crystalline forms of nicotinamide riboside | |
CN111285886B (en) | Substituted pyrazolo [1,5-a ] pyridine compounds, compositions comprising the same and uses thereof | |
JP7092405B2 (en) | Di (hetero) aryl macrocycle to inhibit kinase activity | |
EP3686199B9 (en) | Fused ring derivative as a2a receptor inhibitor | |
US10662189B2 (en) | PDE4 inhibitor | |
KR102283677B1 (en) | SGLT inhibitors and their applications | |
US20240199571A1 (en) | Thiophene compound and application thereof | |
JP5616628B2 (en) | Synthesis and use of pyroglutamic acid derivatives | |
EP3845538B1 (en) | Dihydroimidazopyrazinone compound, composition including same, and use thereof | |
WO2020156189A1 (en) | Camptothecin derivative and water-soluble prodrug thereof, pharmaceutical composition containing same, preparation method, and use | |
CA3203698A1 (en) | 2-pyridone derivative, and preparation method therefor and pharmaceutical application thereof | |
EP3492465B1 (en) | S1p1 agonist and application thereof | |
CN118005654A (en) | Substituted pyridopyrimidinone compounds, compositions containing the same and uses thereof | |
EP3816162A1 (en) | Diarylpyrazole compound, composition comprising same, and use thereof | |
CN115368378B (en) | Substituted macrocyclic compounds, compositions comprising the same and uses thereof | |
CN115785094B (en) | Benzyl substituted alpha-carboline compound or medicinal salt thereof, pharmaceutical composition thereof, preparation method and application thereof | |
WO2000068231A1 (en) | Purine derivative dihydrate, drugs containing the same as the active ingredient and intermediate in the production thereof | |
US20230331675A1 (en) | Aromatic ethylene compound and preparation method therefor, and intermediate, pharmaceutical composition, and application thereof | |
WO2023241414A1 (en) | Pyridazine compound, pharmaceutical composition thereof and application thereof | |
TW202421624A (en) | Carbonyl fused heterocyclic derivative used as ubiquitin-specific protease inhibitor | |
CN118359632A (en) | Camptothecin derivatives, preparation method and application thereof | |
CN118414335A (en) | Compounds as Src inhibitors | |
CN115215883A (en) | USP7 inhibitors | |
CN116332947A (en) | Pyrimidine-2 (1H) -keto bicyclic compounds with MAT2A inhibitory activity 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 |