WO2024125644A1 - 一种含羧基的杂环衍生物及其在医药上的应用 - Google Patents

一种含羧基的杂环衍生物及其在医药上的应用 Download PDF

Info

Publication number
WO2024125644A1
WO2024125644A1 PCT/CN2023/139226 CN2023139226W WO2024125644A1 WO 2024125644 A1 WO2024125644 A1 WO 2024125644A1 CN 2023139226 W CN2023139226 W CN 2023139226W WO 2024125644 A1 WO2024125644 A1 WO 2024125644A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkylene
alkyl
compound
membered
ring
Prior art date
Application number
PCT/CN2023/139226
Other languages
English (en)
French (fr)
Inventor
张晨
赵明亮
刘含波
邓华
钱玫琳
余彦
李瑶
严庞科
Original Assignee
西藏海思科制药有限公司
Priority date (The priority date 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 date listed.)
Filing date
Publication date
Application filed by 西藏海思科制药有限公司 filed Critical 西藏海思科制药有限公司
Publication of WO2024125644A1 publication Critical patent/WO2024125644A1/zh

Links

Definitions

  • the present invention relates to a compound described by general formula (I) or its stereoisomer, deuterated product, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal, and intermediates and preparation methods thereof, as well as use of the compound in preparing drugs for treating diseases related to PI3K ⁇ .
  • Phosphatidylinositol 3-kinase is a lipid kinase that, upon activation by growth factor receptors and integrins, regulates cell proliferation, survival, and migration. Up to 70% of breast cancers have some form of molecular mutation in the PI3K-AKT-mTOR pathway. Activating mutations in PIK3CA (encoding the p110 ⁇ subunit of PI3K) are highly prevalent in breast cancer and solid tumor malignancies.
  • WT PI3K ⁇ plays a central role in regulating glucose homeostasis, and PBK inhibition in patients often leads to hyperglycemia and/or hyperinsulinemia (Busaidy NL, et al, Management of metabolic effects associated with anticancer agents targeting the PBK-Akt-mTOR pathway. J Clin Oncol 2012; 30:2919-28).
  • mutant PI3K ⁇ In the case of cancers with mutant PI3K ⁇ , the development of selective inhibitors for mutant PI3K ⁇ can effectively overcome the compensatory problems of insulin and/or glucose caused by WT PI3K ⁇ inhibition, which can increase the drug administration window and thus selectively inhibit the pathological signaling of mutant PI3K ⁇ in cancer cells without affecting the host's own WT PI3Ka (Okkenhaug K, Graupera M, Vanhaesebroeck B. Targeting PBK in Cancer: Impact on Tumor Cells, Their Protective Stroma, Angiogenesis, and Immunotherapy. Cancer Discov. 2016 Oct; 6(10): 1090-1105).
  • PI3K ⁇ inhibitor development focuses on the active or orthosteric site, with equal effects on wild-type and mutant PI3K ⁇ . Its efficacy is limited by toxicity mediated by targeting WT PI3K ⁇ , including dose limitations, hyperglycemia and/or hyperinsulinemia. Because PI3K ⁇ mutations are located far from the active site, mutation-selective inhibitors have been difficult to obtain. Inhibitors targeting the second peripheral binding pocket near known mutations (e.g., H1047R) are expected to become targets for the development of new anticancer drugs.
  • the purpose of the present invention is to provide a compound capable of inhibiting a target protein or its stereoisomers, deuterated substances, solvates, prodrugs, metabolites, pharmaceutically acceptable salts or cocrystals, as well as intermediates and preparation methods, and their use in the preparation of drugs for treating diseases associated with PI3K ⁇ inhibitors.
  • the compounds of the present invention are highly selective for PI3K ⁇ H1047R, superior to WT PI3K ⁇ and other PI3K isoforms, and do not cause hyperglycemia side effects.
  • the compounds of the present invention have good bioavailability.
  • the present invention provides a compound of general formula (I) or a stereoisomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal thereof, wherein
  • X2 is selected from O, S;
  • X 1 is selected from O, S, NR x , C(R x ) 2 ;
  • X1 is selected from O, S;
  • X1 when X2 is selected from S, X1 is selected from O, S;
  • Y is selected from O, S, NR y ;
  • Y is selected from O, S, NH;
  • m is selected from 1, 2, 3, 4;
  • n is selected from 1, 2;
  • the compound of formula (I) is selected from formula (II), (III), (IV), (V) or (VI),
  • W is selected from C2-4 alkynyl, The alkynyl group is optionally substituted by 1 to 4 R k ;
  • W is selected from ethynyl, propynyl, propargyl, The ethynyl, propynyl and propargyl groups are optionally substituted with 1 to 4 R k ;
  • Z is selected from a bond or -C ⁇ C-;
  • R x and R y are each independently selected from H, deuterium, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl , -C 0-4 alkylene-C 3-10 carbocyclyl, -C 0-4 alkylene-4 to 10 membered heterocyclyl, wherein the alkyl, alkenyl, alkynyl, alkylene, carbocyclyl or heterocyclyl is optionally substituted with 1 to 4 R k ;
  • R x and R y are each independently selected from H, deuterium, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, -C 0-4 alkylene-C 3-8 carbocyclyl, -C 0-4 alkylene-4 to 8 membered heterocyclyl, wherein the alkyl, alkenyl, alkynyl, alkylene, carbocyclyl or heterocyclyl is optionally substituted with 1 to 4 R k ;
  • R x and R y are each independently selected from H, deuterium, methyl, ethyl, propyl, isopropyl, vinyl, ethynyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, and the methyl, ethyl, propyl, isopropyl, vinyl, ethynyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl is optionally substituted with 1 to 4 R k ;
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from H, deuterium, halogen, OH, CN, NO 2 , NH 2 , C 1-6 alkyl, OC 1-6 alkyl, SC 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, NHC 1-6 alkyl, N(C 1-6 alkyl) 2 , -C 0-4 alkylene-C( ⁇ O)R 1a , -C 0-4 alkylene-S( ⁇ O) 2 R 1a , -C 0-4 alkylene-P( ⁇ O)R 1a R 1b , -OC 0-4 alkylene-C 3-10 carbocyclyl, -OC 0-4 alkylene-4 to 10 membered heterocyclyl, -SC 0-4 alkylene-C 3-10 carbocyclyl, -SC 0-4 alkylene-4 to 10 membered heterocyclyl, -NH-
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from H, deuterium, halogen, OH, CN, NO 2 , NH 2 , C 1-4 alkyl, OC 1-4 alkyl, SC 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, NHC 1-4 alkyl, N(C 1-4 alkyl) 2 , -C 0-4 alkylene-C( ⁇ O)R 1a , -C 0-4 alkylene-S( ⁇ O) 2 R 1a , -C 0-4 alkylene-P( ⁇ O)R 1a R 1b , -OC 0-4 alkylene-C 3-8 carbocyclyl, -OC 0-4 alkylene-4 to 8 membered heterocyclyl, -SC 0-4 alkylene-C 3-8 carbocyclyl, -SC 0-4 alkylene-4 to 8 membered heterocyclyl, -SC 0-4 alky
  • R 5 and R 6 are each independently selected from H, deuterium, halogen, OH, CN, NO 2 , NH 2 , C 1-6 alkyl, OC 1-6 alkyl, SC 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, NHC 1-6 alkyl, N(C 1-6 alkyl) 2 , C 0-4 alkylene-5 to 10 membered heteroaryl, C 0-4 alkylene-C 3-10 carbocyclyl, C 0-4 alkylene-4 to 10 membered heterocyclyl, and the alkyl, alkylene, carbocyclyl or heterocyclyl is optionally substituted with 1 to 4 R k ;
  • R 5 and R 6 are each independently selected from H, deuterium, halogen, OH, CN, NO 2 , NH 2 , C 1-4 alkyl, OC 1-4 alkyl, SC 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl , NHC 1-4 alkyl, N(C 1-4 alkyl) 2 , C 0-4 alkylene-5 to 8 membered heteroaryl, C 0-4 alkylene-C 3-8 carbocyclyl, C 0-4 alkylene-4 to 8 membered heterocyclyl, and the alkyl, alkylene, carbocyclyl or heterocyclyl is optionally substituted with 1 to 4 R k ;
  • R 5 and R 6 are each independently selected from H, deuterium, F, Cl, Br, I, OH, CN, NO 2 , NH 2 , NHCH 3 , N(CH 3 ) 2 , methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, vinyl, ethynyl, methylthio, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, morpholinyl, wherein the methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, vinyl, ethynyl, methylthio, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, a
  • ring A is selected from C 3-12 carbocyclyl, 4 to 12 membered heterocyclyl, C 6-10 aryl or 5 to 10 membered heteroaryl, wherein the carbocyclyl, heterocyclyl, aryl or heteroaryl is optionally substituted by 1 to 4 selected from R a , and ring A is connected to Z via a carbon-carbon bond;
  • Ring A is independently selected from a 4-7 membered heteromonocyclic ring, a 5-12 membered heterocyclic ring, a 5-12 membered heterospirocyclic ring, a 7-10 membered heterobridged ring, a C 3-8 monocarbocyclyl, a C 6-12 cycloalkyl, a C 6-12 membered spirocycloalkyl, a C 5-12 membered bridged cycloalkyl, a 5 to 10 membered heteroaryl, a benzo C 3-8 carbocyclyl, a benzo 4 to 8 membered heterocyclyl, Ring A is connected to Z via a carbon-carbon bond, and the Ring A is optionally substituted by 1 to 4 selected from Ra ;
  • each ring A is independently selected from one of the following optionally substituted structures: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, 1,3-dioxolanyl, 1,4-dioxolanyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, phenyl, pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, pyrazolyl, pyrrolyl, imidazolyl, thienyl, thiazolyl, furanyl, oxazolyl, isoxazolyl, benzopyrrolyl, benzothienyl, benzofuranyl, benzothiazolyl, benzopyrazolyl
  • Each is independently selected from one of the following structures optionally substituted by 1 to 4 Ra :
  • Ring B is selected from C 3-12 carbocyclyl, 4 to 12 membered heterocyclyl, C 6-10 aryl or 5 to 10 membered heteroaryl, wherein the carbocyclyl, heterocyclyl, aryl or heteroaryl is optionally substituted by 1 to 4 selected from R b ;
  • Ring B is independently selected from a 4-7 membered heteromonocyclic ring, a 5-12 membered heterocyclic ring, a 5-12 membered heterospirocyclic ring, a 7-10 membered heterobridged ring, a C 3-8 monocarbocyclyl, a C 6-14 cycloalkyl, a C 6-12 membered spirocycloalkyl, a C 5-12 membered bridged cycloalkyl, a benzo C 3-8 carbocyclyl, a benzo 3 to 8 membered heterocyclyl, a C 6-10 aryl, a 5-10 membered heteroaryl, and the Ring B is optionally substituted by 1 to 4 R b ;
  • each ring B is independently selected from C 6-10 aryl, 5-10 membered heteroaryl, and the ring B is optionally substituted by 1 to 4 R b ;
  • each ring B is independently selected from one of the following optionally substituted structures: benzene, naphthalene, pyridine, pyrazine, pyridazine, pyrimidine, When replaced, it is replaced by 1 to 3 R b replaced;
  • B 1 , B 2 , B 3 , B 4 , and B 6 are each independently selected from N or CR b1 , and at most three of B 1 , B 2 , B 3 , B 4 , and B 6 are selected from N;
  • B 1 , B 2 , B 3 , and B 6 are each independently selected from N or CR b1 , and at most two of B 1 , B 2 , B 3 , and B 6 are selected from N;
  • three of B 1 , B 2 , B 3 , and B 6 are selected from CR b1 , and the remaining one is selected from N;
  • ring C 1 , C 2 , C 3 are each independently selected from a 4-8 membered heterocycle or a C 4-8 carbocycle, and the ring C 1 , C 2 , C 3 are optionally substituted by 1 to 4 R k ;
  • ring C 1 , C 2 , C 3 are each independently selected from one of the following groups optionally substituted with 1 to 4 R k : cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, oxetanyl, oxolyl, oxetanyl, azetidinyl, azocyclopentenyl, azocyclohexenyl, 1,3-dioxocyclopentenyl, 1,4-dioxocyclohexenyl;
  • ring C 4 is selected from a 4-8 membered heterocyclic ring, wherein said ring C 4 is optionally substituted with 1 to 4 R k ;
  • ring C 5 is selected from a 4-8 membered nitrogen-containing heterocyclic ring, wherein said ring C 5 is optionally substituted with 1 to 4 R k ;
  • ring C5 is selected from one of the following groups optionally substituted with 1 to 4 Rk : azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl;
  • Selected from Its right end is connected to ring B;
  • Selected from n is selected from 1, 2, and 3;
  • B 1 , B 2 , B 3 , and B 4 are each independently selected from N or CR b1 , and at most two of B 1 , B 2 , B 3 , and B 4 are selected from N;
  • B 5 is each independently selected from N, O, S, S(O) 2 , CR b2 , NR b2 or C(R b2 ) 2 ;
  • each R b2 is independently selected from H or R k ;
  • R b1 is independently selected from H or R b ;
  • B 1 , B 2 , and B 3 are each independently selected from CR b1 ;
  • two of B 1 , B 2 , and B 3 are selected from CR b1 , and the rest are selected from N;
  • R 1a , R 1b are each independently selected from H, OH, NH 2 , C 1-6 alkyl, C 1-6 alkoxy, NHC 1-6 alkyl, N(C 1-6 alkyl) 2 , C 3-8 carbocyclyl, 4 to 8 membered heterocyclyl, C 6-10 aryl, 5 to 10 membered heteroaryl, -OC 3-8 carbocyclyl, -O-4 to 8 membered heterocyclyl, wherein the alkyl, alkoxy, carbocyclyl, heterocyclyl, aryl or heteroaryl is optionally substituted with 1 to 4 R k ;
  • R 1a , R 1b are each independently selected from H, OH, NH 2 , C 1-4 alkyl, C 1-4 alkoxy, NHC 1-4 alkyl, N(C 1-4 alkyl) 2 , C 3-6 carbocyclyl, 4 to 8 membered heterocyclyl, C 6-10 aryl, 5 to 10 membered heteroaryl, -OC 3-8 carbocyclyl, -O-4 to 8 membered heterocyclyl, wherein the alkyl, alkoxy, carbocyclyl, heterocyclyl, aryl or heteroaryl is optionally substituted with 1 to 4 R k ;
  • R 1a , R 1b are each independently selected from H, OH, NH 2 , NHCH 3 , N(CH 3 ) 2 , methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, morpholinyl, -O-cyclopropyl, -O-cyclobutyl, imidazole, pyrazole, pyrrole or thiophene, wherein the methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidiny
  • At least one of R 2 and R 3 , R 3 and R 4 , R 4 and R 5 , R 6 and R y , and R 6 and R b and the atoms connected thereto together form a cyclobutenyl, cyclopentenyl, cyclohexenyl, azetidinyl, oxetinyl, oxetinyl, azidine, azidine, 1,3-dioxocyclopentenyl, 1,4-dioxocyclohexenyl, and the cyclobutenyl, cyclopentenyl, cyclohexenyl, azetidinyl, oxetinyl, oxetinyl, azidine, azidine, 1,3-dioxocyclopentenyl, 1,4-dioxocyclohexenyl is optionally substituted by 1 to 4 R k ;
  • X2 is selected from O, S;
  • X 1 is selected from O, S, NR x , C(R x ) 2 ;
  • Y is selected from O, S, NR y ;
  • W is selected from C2-4 alkynyl, The alkynyl group is optionally substituted by 1 to 4 R k ;
  • Z is selected from a bond or -C ⁇ C-;
  • n is selected from 1, 2, 3, 4;
  • R x and R y are each independently selected from H, deuterium, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-4 alkylene-C 3-10 carbocyclyl, -C 0-4 alkylene-4 to 10 membered heterocyclyl, wherein the alkyl, alkenyl, alkynyl, alkylene, carbocyclyl or heterocyclyl is optionally substituted by 1 to 4 R k ;
  • Ring A is selected from C 3-12 carbocyclyl, 4- to 12-membered heterocyclyl, C 6-10 aryl or 5- to 10-membered heteroaryl, wherein the carbocyclyl, heterocyclyl, aryl or heteroaryl is optionally substituted by 1 to 4 groups selected from R a ;
  • Ring B is selected from C 3-12 carbocyclyl, 4-12 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, wherein the carbocyclyl, heterocyclyl, aryl or heteroaryl is optionally substituted by 1 to 4 groups selected from R b ;
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from H, deuterium, halogen, OH, CN, NO 2 , NH 2 , C 1-6 alkyl, OC 1-6 alkyl, SC 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, NHC 1-6 alkyl, N(C 1-6 alkyl) 2 , -C 0-4 alkylene-C( ⁇ O)R 1a , -C 0-4 alkylene-S( ⁇ O) 2 R 1a , -C 0-4 alkylene-P( ⁇ O)R 1a R 1b , -OC 0-4 alkylene-C 3-10 carbocyclyl, -OC 0-4 alkylene-4 to 10 membered heterocyclyl, -SC 0-4 alkylene-C 3-10 carbocyclyl, -SC 0-4 alkylene-4 to 10 membered heterocyclyl, -NH-C -C
  • R 5 and R 6 are each independently selected from H, deuterium, halogen, OH, CN, NO 2 , NH 2 , C 1-6 alkyl, OC 1-6 alkyl, SC 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, NHC 1-6 alkyl, N(C 1-6 alkyl) 2 , C 0-4 alkylene-5 to 10 membered heteroaryl, C 0-4 alkylene-C 3-10 carbocyclyl, C 0-4 alkylene-4 to 10 membered heterocyclyl, wherein the alkyl, alkylene, carbocyclyl or heterocyclyl is optionally substituted by 1 to 4 R k ;
  • R 1a and R 1b are each independently selected from H, OH, NH 2 , C 1-6 alkyl, C 1-6 alkoxy, NHC 1-6 alkyl, N(C 1-6 alkyl) 2 , C 3-8 carbocyclyl, 4 to 8 membered heterocyclyl, C 6-10 aryl, 5 to 10 membered heteroaryl, -OC 3-8 carbocyclyl, -O-4 to 8 membered heterocyclyl, wherein the alkyl, alkoxy, carbocyclyl, heterocyclyl, aryl or heteroaryl is optionally substituted by 1 to 4 R k ;
  • R x and R y are each independently selected from H, deuterium, C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, -C 0-4 alkylene-C 3-8 carbocyclyl, -C 0-4 alkylene-4 to 8 membered heterocyclyl, wherein the alkyl, alkenyl, alkynyl, alkylene, carbocyclyl or heterocyclyl is optionally substituted by 1 to 4 R k ;
  • R 1a , R 1b are each independently selected from H, OH, NH 2 , C 1-4 alkyl, C 1-4 alkoxy, NHC 1-4 alkyl, N(C 1-4 alkyl) 2 , C 3-6 carbocyclyl, 4 to 8 membered heterocyclyl, C 6-10 aryl, 5 to 10 membered heteroaryl, -OC 3-8 carbocyclyl, -O-4 to 8 membered heterocyclyl, wherein the alkyl, alkoxy, carbocyclyl, heterocyclyl, aryl or heteroaryl is optionally substituted by 1 to 4 R k ;
  • Ring A is independently selected from a 4-7 membered heteromonocyclic ring, a 5-12 membered heterocyclic ring, a 5-12 membered heterospirocyclic ring, a 7-10 membered heterobridged ring, a C 3-8 monocarbocyclic ring, a C 6-12 cycloalkyl group, a C 6-12 membered spirocycloalkyl group, a C 5-12 membered bridged cycloalkyl group, a 5-10 membered heteroaryl group, a benzo C 3-8 carbocyclic ring group, a benzo 4-8 membered heterocyclic ring group, and the ring A is connected to Z via a carbon-carbon bond, and the ring A is optionally substituted by 1 to 4 groups selected from Ra ;
  • Ring B is independently selected from 4-7 membered heteromonocyclic ring, 5-12 membered heterocyclic ring, 5-12 membered heterospirocyclic ring, 7-10 membered heterobridged ring, C 3-8 monocarbocyclic ring, C 6-14 cycloalkyl group, C 6-12 membered spirocyclic ring alkyl group, C 5-12 membered bridged cycloalkyl group, benzo C 3-8 carbocyclic ring group, benzo 3 to 8 membered heterocyclic ring group, C 6-10 aryl group, 5-10 membered heteroaryl group, and the ring B is optionally substituted by 1 to 4 R b ;
  • Ring C 1 , C 2 , C 3 are each independently selected from a 4-8 membered heterocycle or a C 4-8 carbocycle, and the ring C 1 , C 2 , C 3 are optionally substituted by 1 to 4 R k ;
  • Ring C 4 is selected from a 4-8 membered heterocyclic ring, and the ring C 4 is optionally substituted by 1 to 4 R k ;
  • Ring C5 is selected from a 4-8 membered nitrogen-containing heterocyclic ring, and the ring C5 is optionally substituted by 1 to 4 Rk ;
  • Ring B is independently selected from C 6-10 aryl, 5-10 membered heteroaryl, and the ring B is optionally substituted by 1 to 4 R b ;
  • n is selected from 1, 2, and 3;
  • B 1 , B 2 , B 3 , and B 4 are each independently selected from N or CR b1 , and at most two of B 1 , B 2 , B 3 , and B 4 are selected from N;
  • B 5 is each independently selected from N, O, S, CR b2 , S(O) 2 , NR b2 or C(R b2 ) 2 ;
  • R b2 are each independently selected from H or R k ;
  • R b1 are each independently selected from H or R b ;
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from H, deuterium, halogen, OH, CN, NO 2 , NH 2 , C 1-4 alkyl, OC 1-4 alkyl, SC 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, NHC 1-4 alkyl, N(C 1-4 alkyl) 2 , -C 0-4 alkylene-C( ⁇ O)R 1a , -C 0-4 alkylene-S( ⁇ O) 2 R 1a , -C 0-4 alkylene-P( ⁇ O)R 1a R 1b , -OC 0-4 alkylene-C 3-8 carbocyclyl, -OC 0-4 alkylene-4 to 8 membered heterocyclyl, -SC 0-4 alkylene-C 3-8 carbocyclyl, -SC 0-4 alkylene-4 to 8 membered heterocyclyl, -NH-C 0-4
  • R 5 and R 6 are each independently selected from H, deuterium, halogen, OH, CN, NO 2 , NH 2 , C 1-4 alkyl, OC 1-4 alkyl, SC 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, NHC 1-4 alkyl, N(C 1-4 alkyl) 2 , C 0-4 alkylene-5 to 8 membered heteroaryl, C 0-4 alkylene-C 3-8 carbocyclyl, C 0-4 alkylene-4 to 8 membered heterocyclyl, wherein the alkyl, alkylene, carbocyclyl or heterocyclyl is optionally substituted by 1 to 4 R k ;
  • X 1 is selected from O, S, NR x ;
  • R x and R y are each independently selected from H, deuterium, methyl, ethyl, propyl, isopropyl, vinyl, ethynyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, piperazinyl, and morpholinyl, and the methyl, ethyl, propyl, isopropyl, vinyl, ethynyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, piperazinyl, and morpholinyl are optionally substituted with 1 to 4 R k ;
  • R 1a , R 1b are each independently selected from H, OH, NH 2 , NHCH 3 , N(CH 3 ) 2 , methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropyloxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, morpholinyl, -O-cyclopropyl, -O-cyclobutyl, imidazole, pyrazole, pyrrole or thiophene, wherein the methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropyloxy, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidiny
  • R 5 and R 6 are each independently selected from H, deuterium, F, Cl, Br, I, OH, CN, NO 2 , NH 2 , NHCH 3 , N(CH 3 ) 2 , methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, vinyl, ethynyl, methylthio, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl and morpholinyl, and the methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, isopropoxy, vinyl, ethynyl, methylthio, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidiny
  • Ring C 1 , C 2 , C 3 are each independently selected from one of the following groups optionally substituted with 1 to 4 R k : cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, oxetanyl, oxolyl, oxinyl, azetidinyl, azocyclopentenyl, azocyclohexenyl, 1,3-dioxocyclopentenyl, 1,4-dioxocyclohexenyl;
  • Ring C5 is selected from one of the following groups optionally substituted by 1 to 4 Rk : azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl;
  • W is selected from ethynyl, propynyl, propargyl, The ethynyl, propynyl and propargyl groups are optionally substituted with 1 to 4 R k ;
  • Y is selected from O, S, NH;
  • Ring B is independently selected from one of the following structures which are optionally substituted: benzene, naphthalene, pyridine, pyrazine, pyridazine, pyrimidine, When substituted, it is substituted with 1 to 3 R b ;
  • Ring A is each independently selected from one of the following optionally substituted structures: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, 1,3-dioxolanyl, 1,4-dioxolanyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, phenyl, pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, pyrazolyl, pyrrolyl, imidazolyl, thienyl, thiazolyl, furanyl, oxazolyl, isoxazolyl, benzopyrrolyl, benzothienyl, benzofuranyl, benzothiazolyl, benzopyrazolyl, benzimid
  • n1, n2, n3 are each independently selected from 1, 2, 3;
  • B 1 , B 2 , B 3 , and B 6 are each independently selected from N or CR b1 , and at most two of B 1 , B 2 , B 3 , and B 6 are selected from N;
  • the present invention relates to the following compound or its stereoisomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal, wherein the compound is selected from one of the structures in Table A below:
  • the present invention relates to a pharmaceutical composition, comprising the compound of the present invention or its stereoisomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal, and a pharmaceutically acceptable carrier.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the above-mentioned compound of the present invention or its stereoisomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal, and a pharmaceutically acceptable carrier.
  • the present invention relates to use of the compound of the present invention or its stereoisomer, deuterated product, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal in preparing a drug for treating diseases associated with PI3K ⁇ , preferably in preparing a drug for treating tumors or cancers (such as breast cancer).
  • the pharmaceutical composition of the present invention may be in the form of a unit preparation (the amount of the main drug in the unit preparation is also referred to as "preparation strength").
  • an "effective amount” or “therapeutically effective amount” as used herein refers to administering a sufficient amount of the compound disclosed herein to alleviate one or more symptoms of the disease or condition being treated (e.g., treating a disease associated with PI3K ⁇ , such as a tumor) to some extent.
  • the result is reduction and/or alleviation of signs, symptoms or causes of a disease, or any other desired alteration of a biological system.
  • an "effective amount” for therapeutic uses is the amount of a compound disclosed herein that is required to provide a clinically significant reduction in disease symptoms.
  • therapeutically effective amounts include, but are not limited to, 1-1500 mg, 1-1200 mg, 1-1000 mg, 1-900 mg, 1-800 mg, 1-700 mg, 1-600 mg, 2-600 mg, 3-600 mg, 4-600 mg, 5-600 mg, 6-600 mg, 10-600 mg, 20-600 mg, 25-600 mg, 30-600 mg, 40-600 mg, 50-600 mg, 60-600 mg, 70-600 mg, 75-600 mg, 80-600 mg, 90-600 mg, 100-600 mg, 200-600 mg, 1-500 mg, 2-500mg, 3-500mg, 4-500mg, 5-500mg, 6-500mg, 10-500mg, 20-500mg, 25-500mg, 30-500mg, 40-500mg, 50-500mg, 60-500mg, 70-500mg, 75-500mg, 80-500mg, 90-500mg, 100-500mg, 125-500mg, 150-500mg, 200-500mg, 250-500mg, 300-500m
  • the pharmaceutical composition includes but is not limited to 1-1000 mg, 20-800 mg, 40-800 mg, 40-400 mg, 25-200 mg, 1 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 110 mg, 120 mg , 125 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg, 200 mg, 210 mg, 220 mg, 230 mg, 240 mg, 250 mg, 300 mg, 320 mg, 400 mg, 480 mg, 500 mg, 600 mg, 640 mg, 840 mg of a compound of the present invention or a stereoisomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal thereof.
  • a method for treating a disease in a mammal comprising administering to a subject a therapeutically effective amount of a compound of the present invention or a stereoisomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal thereof, the therapeutically effective amount preferably being 1-1500 mg, and the disease preferably being a PI3K ⁇ inhibitor-related disease (such as a tumor).
  • a method for treating a disease in a mammal comprising administering a drug compound of the present invention or a stereoisomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal thereof to a subject at a daily dose of 1-1000 mg/day
  • the daily dose may be a single dose or divided doses, in some embodiments, the daily dose includes but is not limited to 10-1500 mg/day, 10-1000 mg/day, 10-800 mg/day, 25-800 mg/day, 50-800 mg/day, 100-800 mg/day, 200-800 mg/day , 25-400 mg/day, 50-400 mg/day, 100-400 mg/day, 200-400 mg/day, in some embodiments, daily doses include but are not limited to 10 mg/day, 20 mg/day, 25 mg/day, 50 mg/day, 80 mg/day, 100 mg/day, 125 mg/day, 150 mg/day, 160 mg/day, 200 mg/day, 300 mg/day, 320
  • the present invention relates to a kit, which may include a composition in a single-dose or multi-dose form, and the kit contains a compound of the present invention or a stereoisomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal thereof, and the amount of the compound of the present invention or its stereoisomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal is the same as that in the above-mentioned pharmaceutical composition.
  • the present invention relates to a compound of the present invention or its stereoisomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal or the above-mentioned pharmaceutical composition for use in preparing a drug for treating diseases associated with PI3K ⁇ inhibitors.
  • a compound of the present invention or its stereoisomer, deuterated substance, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal or the above-mentioned pharmaceutical composition for use in preparing a drug for treating diseases associated with PI3K ⁇ inhibitors.
  • the present invention relates to the use of the above-mentioned compound of the present invention or its stereoisomer, deuterated product, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal, or the above-mentioned pharmaceutical composition, and the disease is selected from tumors or cancers, preferably breast cancer.
  • the amount of the compound of the invention or a stereoisomer, deuterated form, solvate, prodrug, metabolite, pharmaceutically acceptable salt or cocrystal thereof is in each case calculated as the free base.
  • the carbon, hydrogen, oxygen, sulfur, nitrogen or F, Cl, Br, I involved in the groups and compounds described in the present invention all include their isotopes, and the carbon, hydrogen, oxygen, sulfur or nitrogen involved in the groups and compounds described in the present invention are optionally further replaced by one or more of their corresponding isotopes, wherein carbon isotopes include 12 C, 13 C and 14 C, hydrogen isotopes include protium (H), deuterium (D, also called heavy hydrogen), tritium (T, also called super tritium), oxygen isotopes include 16 O, 17 O and 18 O, sulfur isotopes include 32 S, 33 S, 34 S and 36 S, nitrogen isotopes include 14 N and 15 N, fluorine isotopes include 17 F and 19 F, chlorine isotopes include 35 Cl and 37 Cl, and bromine isotopes include 79 Br and 81 Br.
  • carbon isotopes include 12 C, 13 C and 14 C
  • hydrogen isotopes include pro
  • CN refers to cyano
  • Halogen refers to F, Cl, Br or I.
  • Halogen substituted refers to substitution with F, Cl, Br or I, including but not limited to substitution with 1 to 10 substituents selected from F, Cl, Br or I, substitution with 1 to 6 substituents selected from F, Cl, Br or I, and substitution with 1 to 4 substituents selected from F, Cl, Br or I.
  • Halogen substituted is abbreviated as "halo”.
  • Alkyl refers to a substituted or unsubstituted straight or branched chain saturated aliphatic hydrocarbon group, including but not limited to alkyl groups of 1 to 20 carbon atoms, alkyl groups of 1 to 8 carbon atoms, alkyl groups of 1 to 6 carbon atoms, and alkyl groups of 1 to 4 carbon atoms.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, neobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, and various branched chain isomers thereof; alkyl groups can be monovalent, divalent, trivalent, or tetravalent.
  • Heteroalkyl refers to a substituted or unsubstituted alkyl in which one or more (including but not limited to 2, 3, 4, 5 or 6) carbon atoms are replaced by heteroatoms (including but not limited to N, O or S).
  • Non-limiting examples include -X-( CH2 )vX-( CH2 )vX-( CH2 )vH (v is an integer from 1 to 5, each X is independently selected from a bond or a heteroatom, the heteroatom includes but is not limited to N, O or S, and at least one X is selected from a heteroatom, and the N or S in the heteroatom can be oxidized to various oxidation states).
  • the heteroalkyl group can be monovalent, divalent, trivalent or tetravalent.
  • Alkylene refers to a substituted or unsubstituted straight-chain or branched divalent saturated hydrocarbon group, including -(CH 2 ) v -(v is an integer from 1 to 10). Examples of alkylene include, but are not limited to, methylene, ethylene, propylene, and butylene.
  • Heteroalkylene refers to a substituted or unsubstituted alkylene in which one or more (including but not limited to 2, 3, 4, 5 or 6) carbon atoms are replaced by heteroatoms (including but not limited to N, O or S).
  • Non-limiting examples include -X-(CH 2 )vX-(CH 2 )vX-(CH 2 )v-(CH 2 )v-, v is an integer from 1 to 5, each X is independently selected from a bond, N, O or S, and at least one X is selected from N, O or S.
  • Cycloalkyl refers to a substituted or unsubstituted saturated carbocyclic hydrocarbon radical, typically having 3 to 12 carbon atoms, and the cycloalkyl can be a monocyclic, cyclic, bridged, and spirocyclic ring. Non-limiting examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclobutyl-cyclobutyl, cyclobutyl-spirocyclobutyl, adamantane, etc.
  • the cycloalkyl can be monovalent, divalent, trivalent, or tetravalent.
  • Heterocycloalkyl refers to a substituted or unsubstituted saturated cyclic hydrocarbon group containing heteroatoms, including but not limited to 3 to 12 atoms, 3 to 8 atoms, including 1 to 3 heteroatoms selected from N, O, S or Se. Can be oxidized to various oxidation states. Heterocycloalkyl can be monocyclic, cyclic, bridged and spirocyclic.
  • Heterocycloalkyl can be connected to a heteroatom or a carbon atom, and non-limiting examples include oxirane, aziridine, oxetanyl, azetidinyl, tetrahydrofuranyl, tetrahydro-2H-pyranyl, dioxolanyl, dioxane, pyrrolidinyl, piperidinyl, imidazolidinyl, oxazolidinyl, oxazolidinyl, oxazinyl, morpholinyl, hexahydropyrimidinyl, piperazinyl,
  • the heterocycloalkyl group may be monovalent, divalent, trivalent or tetravalent.
  • alkenyl refers to a substituted or unsubstituted straight or branched unsaturated hydrocarbon group having at least one, typically one, two or three carbon-carbon double bonds, with a backbone of 2 to 10, 2 to 6 or 2 to 4 carbon atoms.
  • alkenyl groups include, but are not limited to, vinyl, allyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1-methyl-1-butenyl, 2 ...
  • alkenyl group can be monovalent, divalent, trivalent or tetravalent.
  • Alkynyl refers to substituted or unsubstituted straight and branched unsaturated hydrocarbon groups having at least one, typically one, two or three carbon-carbon triple bonds, with a main chain comprising 2 to 10 carbon atoms, including but not limited to 2 to 6 carbon atoms in the main chain and 2 to 4 carbon atoms in the main chain.
  • alkynyl groups include but are not limited to ethynyl, propargyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1-pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 5-pentynyl, 6-pentynyl, 7-pentynyl, 8-pentynyl, 9-pentynyl, 10-pentynyl, 11-pentynyl, 12-pentynyl, 13-pentynyl, 14-pentynyl, 15-pentynyl, 16-pentynyl, 17-pentynyl, 18-pentynyl, 19-pentynyl, 20-pentynyl, 21-pentynyl, 22-pentynyl, 23-pent
  • Alkoxy refers to substituted or unsubstituted -O-alkyl. Non-limiting examples include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, n-hexoxy, cyclopropyloxy, and cyclobutyloxy.
  • Carbocyclyl or “carbocycle” refers to a substituted or unsubstituted aromatic or non-aromatic ring, which can be a 3-8-membered monocyclic ring, a 4-12-membered bicyclic ring, a 10-15-membered tricyclic ring, or a 12-18-membered quaternary system.
  • the carbocyclyl can be attached to an aromatic ring or a non-aromatic ring, and the ring can be optionally a monocyclic ring, a cyclic ring, a bridged ring, or a spirocyclic ring.
  • Non-limiting examples include cyclopropane, cyclobutane, cyclopentane, cyclohexane, cycloheptane, 1-cyclopentyl-1-alkenyl, 1-cyclopentyl-2-alkenyl, 1-cyclopentyl-3-alkenyl, cyclohexyl, 1-cyclohexyl-2-alkenyl, 1-cyclohexyl-3-alkenyl, cyclohexenyl, benzene ring, naphthalene ring, "Carbocyclyl” or “carbocycle” can be monovalent, divalent, trivalent or tetravalent.
  • Heterocyclyl or “heterocycle” refers to a substituted or unsubstituted aromatic or non-aromatic ring, which may be a 3-8-membered monocyclic ring, a 4-12-membered bicyclic ring, or a 10-15-membered tricyclic ring, or a 12-18-membered quaternary system, and contains one or more (including but not limited to 2, 3, 4 or 5) heteroatoms selected from N, O, S or Se, and the C, N, S or Se selectively substituted in the heterocyclyl ring may be oxidized to various oxidation states.
  • the heterocyclyl may be attached to a heteroatom or a carbon atom, and may be attached to an aromatic ring or a non-aromatic ring.
  • the heterocyclyl may be optionally a monocyclic ring, a bridged ring, a cyclic ring or a spirocyclic ring.
  • Non-limiting examples include oxirane, aziridine, oxadiazine, azetidinyl, 1,3-dioxolanyl, 1,4-dioxolanyl, 1,3-dioxolanyl, hexacyclyl, azepanyl, pyridinyl, furanyl, thienyl, pyranyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, piperidinyl, morpholinyl, thiomorpholinyl, 1,3-dithianyl, dihydrofuranyl, dihydropyranyl, dithiolanyl, tetrahydrofuranyl, tetrahydropyrrolyl, tetrahydroimidazolyl, tetrahydrothiazolyl, tetrahydropyranyl, benzimidazolyl,
  • pyrrolyl, pyrazolyl, thiazolyl, oxazolyl, pyrazinyl, indazolyl, benzothiophenyl, benzofuranyl, benzopyrrolyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, benzopyridinyl, benzopyrimidinyl, benzopyrazinyl, piperazinyl, azabicyclo[3.2.1]octyl, azabicyclo[5.2.0]nonanyl, oxatricyclo[5.3.1.1]dodecyl, azaadamantyl, oxaspiro[3.3]heptyl, "Heterocyclyl" or “heterocycle” may be monovalent, divalent, trivalent or tetravalent.
  • Spiro or “spirocyclic group” refers to a polycyclic group in which substituted or unsubstituted monocyclic rings share one atom (called a spiro atom).
  • “Spirocycle” or “spirocyclyl” may be monovalent, divalent, trivalent or tetravalent.
  • the number of ring atoms in the cyclic system includes but is not limited to 5 to 20, 5 to 14, 5 to 12, 5 to 10.
  • Non-limiting examples include: "Bicyclic" or "bicyclic group” can be monovalent, divalent, trivalent or tetravalent.
  • Carbospirocycle refers to a “spirocycle” wherein the ring system consists of only carbon atoms.
  • Carbocyclic refers to a “cyclic” ring system consisting of only carbon atoms.
  • Carbobridged ring refers to a “bridged ring” wherein the ring system consists of only carbon atoms.
  • Heteromonocycle refers to a monocyclic ring system of "heterocyclyl” or “heterocycle”,
  • Heterocyclic ring refers to a “cyclic ring” containing a heteroatom.
  • Heterospirocycle refers to a “spirocycle” containing a heteroatom.
  • Heterobridged ring refers to a “bridged ring” containing a heteroatom.
  • Aryl or “aromatic ring” refers to a substituted or unsubstituted aromatic hydrocarbon group having a single ring or a fused ring, wherein the number of ring atoms in the aromatic ring includes, but is not limited to, 6 to 18, 6 to 12, or 6 to 10 carbon atoms.
  • the aryl ring may be fused to a saturated or unsaturated carbon ring, wherein the ring connected to the parent structure is an aryl ring, non-limiting examples of which include a benzene ring, a naphthalene ring, "Aryl” or “aromatic ring” can be monovalent, divalent, trivalent or tetravalent. When divalent, trivalent or tetravalent, the point of attachment is on the aryl ring.
  • heteroaryl examples include but are not limited to pyridyl, furanyl, thienyl, selenophenyl, pyridyl, pyranyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, benzopyrazolyl, benzimidazolyl, benzopyridinyl, pyrrolopyridinyl, pyridonyl, and the like.
  • the heteroaryl ring may be fused to a saturated or unsaturated carbocyclic or heterocyclic ring, wherein the ring connected to the parent structure is an aryl ring, non-limiting examples include
  • the heteroaryl groups appearing in this article have the same definition as this definition.
  • the heteroaryl group can be monovalent, divalent, trivalent or tetravalent. When it is divalent, trivalent or tetravalent, the attachment site is located on the aromatic ring.
  • X-Y membered rings (X, Y are integers, and 3 ⁇ X ⁇ Y, X ⁇ Y ⁇ 20 are selected from any integer between 4 and 20) include X, X+1, X+2, X+3, X+4....Y membered rings.
  • Rings include heterocyclic rings, carbocyclic rings, aromatic rings, aryl groups, heteroaryl groups, cycloalkyl groups, heteromonocyclic rings, heterocyclic rings, heterospirocyclic rings or heterobridged rings.
  • 4--7 membered heteromonocyclic rings refer to 4-, 5-, 6- or 7-membered heteromonocyclic rings
  • 5--10 membered heterocyclic rings refer to 5-, 6-, 7-, 8-, 9- or 10-membered heterocyclic rings
  • Cxy carbocycle (including aryl, cycloalkyl, monocyclic carbocycle, spirocyclic carbocycle, cyclic carbocycle or bridged carbocycle) includes Cx , Cx +1 , Cx +2 , Cx +3 , Cx +4 ... Cy- membered ring (x is an integer, and 3 ⁇ x ⁇ y, y is selected from any integer between 4 and 20), for example.
  • C3-6 cycloalkyl refers to C3 , C4 , C5 or C6 cycloalkyl;
  • any one or more sites of the group can be connected to other groups through chemical bonds.
  • the chemical bond connection is non-positional and there are hydrogen atoms at the connectable sites, when the chemical bonds are connected, the number of H atoms at the site will decrease with the number of connected chemical bonds and become a group with the corresponding valence.
  • any connectable site on the piperidine group can be connected to other groups through one chemical bond, including at least These four connection methods, even if the H atom is drawn on -N-, Also included For example Indicates that the R group on the piperidinyl group can be located on C, can be located on N, and at least includes
  • connection directions include connection from left to right and from right to left in reading order, for example, A-L-B, when L is selected from -M-W-, includes A-M-W-B and A-W-M-B.
  • alkyl optionally substituted with F means that alkyl may but need not be substituted with F, and the description includes situations where alkyl is substituted with F and situations where alkyl is not substituted with F.
  • “Pharmaceutically acceptable salt” or “pharmaceutically acceptable salt thereof” refers to a salt of the compound of the present invention that retains the biological effectiveness and properties of the free acid or free base, and the free acid is obtained by reacting with a non-toxic inorganic base or organic base, and the free base is obtained by reacting with a non-toxic inorganic acid or organic acid.
  • “Pharmaceutical composition” refers to one or more compounds of the present invention, or their stereoisomers, tautomers, deuterated A mixture formed by a solvate, a prodrug, a metabolite, a pharmaceutically acceptable salt or a co-crystal and other chemical components, wherein “other chemical components” refers to a pharmaceutically acceptable carrier, an excipient and/or one or more other therapeutic agents.
  • Preparation specifications refers to the weight of the main drug contained in each vial, tablet or other unit preparation.
  • Carrier refers to a material that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
  • Prodrug refers to a compound of the present invention that can be converted into a biologically active compound through in vivo metabolism.
  • the prodrug of the present invention is prepared by modifying the amino or carboxyl group in the compound of the present invention, and the modification can be removed by conventional operations or in vivo to obtain the parent compound.
  • the prodrug of the present invention is administered to a mammalian subject, the prodrug is cleaved to form a free amino or carboxyl group.
  • Co-crystal refers to a crystal formed by the active pharmaceutical ingredient (API) and the co-crystal former (CCF) under the action of hydrogen bonds or other non-covalent bonds, in which the pure state of API and CCF are solid at room temperature and there is a fixed stoichiometric ratio between the components.
  • Co-crystal is a multi-component crystal, including binary eutectics formed between two neutral solids and multi-component eutectics formed between neutral solids and salts or solvates.
  • Animal is meant to include mammals, such as humans, companion animals, zoo animals, and livestock, preferably humans, horses, or dogs.
  • Stepoisomers refer to isomers resulting from different spatial arrangements of atoms in a molecule, including cis-trans isomers, enantiomers, diastereomers and conformational isomers.
  • Tautomers refer to functional group isomers produced by the rapid movement of an atom in a molecule between two positions, such as keto-enol isomerism and amide-imino alcohol isomerism.
  • IC50 is the concentration of a drug or inhibitor required to inhibit a specified biological process (or a component of such a process, such as an enzyme, receptor, cell, etc.) by half.
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • HPLC determination was performed using an Agilent 1260DAD high pressure liquid chromatograph (Zorbax SB-C18 100 ⁇ 4.6mm, 3.5 ⁇ M);
  • the thin layer chromatography silica gel plate uses Yantai Huanghai HSGF 254 or Qingdao GF 254 silica gel plate.
  • the silica gel plate used in thin layer chromatography (TLC) adopts a specification of 0.15mm-0.20mm, and the specification used for thin layer chromatography separation and purification products is 0.4mm-0.5mm;
  • the compounds used in the reactions described herein are prepared according to organic synthesis techniques known to those skilled in the art, starting from commercially available chemicals and/or compounds described in the chemical literature.
  • “Commercially available chemicals” are obtained from standard commercial sources, including Shanghai Aladdin Biochemical Technology Co., Ltd., Shanghai McLean Biochemical Technology Co., Ltd., Sigma-Aldrich, Alfa Aesar (China) Chemical Co., Ltd., TCI (Shanghai) Chemical Industry Development Co., Ltd., Anage Chemical, Shanghai Titan Technology Co., Ltd., Kelon Chemical, Bailingwei Technology Co., Ltd., etc.
  • THF tetrahydrofuran
  • DMF N,N-dimethylformamide
  • DIPEA N,N-diisopropylethylamine
  • HATU CAS 148893-10-1
  • Boc tert-butoxycarbonyl
  • Ts p-toluenesulfonyl
  • Cbz benzyloxycarbonyl
  • TMS trimethylsilyl.
  • [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride dichloromethane complex (44 mg, 0.05 mmol) was added to a solution of 1A (370 mg, 1.07 mmol) (obtained from patent WO2022235574), 1B (376 mg, 1.18 mmol), potassium carbonate (369 mg, 2.68 mmol) in dioxane (10 mL) and water (1 mL). The mixture was reacted at 100 °C for 16 h under a nitrogen atmosphere. The mixture was cooled to room temperature, water (20 mL) was added, and the mixture was extracted with ethyl acetate (50 mL x 3), and the organic phases were combined.
  • Trifluoroacetic acid (4 mL) was added to a solution of 1C (50 mg, 0.092 mmol) in dichloromethane (2 mL) and the mixture was reacted at room temperature for 3 h.
  • the reaction solution was concentrated under reduced pressure to obtain a residue, water (10 mL) was added to the residue, and the mixture was extracted with ethyl acetate (10 mL x 2), and the organic phases were combined.
  • the organic phases were washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain 2B (42 mg, crude product).
  • 1N aqueous hydroxide solution (1.0M, 5mmol, 5mL) was added to a solution of 3b (42mg, crude product) in methanol (5mL) and tetrahydrofuran (5mL), and the mixture was reacted at 50°C for 16h.
  • the residue was concentrated under reduced pressure, and the pH was adjusted to 6-7 with 1N aqueous hydrochloric acid solution under ice-water bath, extracted with ethyl acetate (20mL x 2), and the organic phases were combined.
  • Trifluoroacetic acid (2 mL) was added to a solution of 3C (380 mg, 0.87 mmol) in dichloromethane (5 mL), and the mixture was reacted at room temperature for 3 h under a nitrogen atmosphere. The reaction solution was concentrated under reduced pressure to obtain the trifluoroacetate salt of 3D (400 mg, crude product).
  • Methyl 6-chloro-3-fluoropyridine-2-carboxylate (68 mg, 0.36 mmol) and N,N-diisopropylethylamine (155 mg, 1.20 mmol) were added to dimethyl sulfoxide (5 mL) of the hydrochloride of 4i (109 mg, crude product) and reacted at 100°C for 16 h. After cooling to room temperature, water (20 mL) was added and extracted with ethyl acetate (30 mL x 2). The organic phases were combined, washed with saturated brine (40 mL x 3), dried over anhydrous sodium sulfate, and filtered.
  • N-Bromosuccinimide (4.68 g, 26.28 mmol) was added to a solution of 5c (5.00 g, 26.28 mmol) in N,N-dimethylformamide (50 mL) and reacted at room temperature for 16 h.
  • the reaction solution was poured into water (500 mL) and extracted with ethyl acetate (100 mL x 3). The organic phases were combined, washed with saturated brine (50 mL x 3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product.
  • Methyl 2-aminobenzoate (100 mg, 0.66 mmol) was added dropwise to N,N-dimethylformamide (5 mL) of 5i (130 mg, crude product) and reacted at 80 °C for 16 h. After cooling to room temperature, water (20 mL) was added and extracted with ethyl acetate (30 mL x 2), and the organic phases were combined.
  • Compound 5 was purified by SFC on AS column (instrument and preparation column: Waters 150 Prep-SFC F, preparation column model: Chiralcel AS column. Preparation method: Compound 5 was dissolved in acetonitrile and filtered with a 0.45 ⁇ m filter membrane to prepare a sample solution. Mobile phase system: A for CO 2 and B for EtOH (0.1% NH 3 ⁇ H 2 O).
  • Methyl 2-aminobenzoate (390 mg, 2.58 mmol) was added dropwise to 8e (475 mg, crude product) in N,N-dimethylformamide (10 mL), and the mixture was reacted at 80 °C for 16 h. The mixture was cooled to room temperature, and water (20 mL) was added, and the mixture was extracted with ethyl acetate (30 mL x 2), and the organic phases were combined.
  • N,N-diisopropylethylamine (710 mg, 5.5 mmol) was added dropwise to 8 g (250 mg, 0.55 mmol) and 5-fluoroisoindoline hydrochloride. (430 mg, 2.48 mmol) in dimethyl sulfoxide (10 mL), sealed at 100 ° C for 48 h. Cool to room temperature, add water (20 mL), extract with ethyl acetate (20 mL x 2), and combine the organic phases.
  • Preparation method Dissolve the crude product in DMF and filter with a 0.45 ⁇ m filter membrane to prepare a sample solution.
  • Mobile phase system acetonitrile/water (containing 0.05% trifluoroacetic acid).
  • Gradient elution method gradient elution from 50% methanol to 80% methanol (flow rate: 15 mL/min; elution time 15 min) to separate and purify compound 8 (15 mg, yield 19%).
  • Compound 9 was obtained with 5d and m-fluorobenzaldehyde as starting materials, referring to patent WO2022235574A1.
  • Compound 9 was purified by SFC on AD column (instrument and preparation column: Waters 150 Prep-SFC F, preparation column model: Chiralcel AD column.
  • Preparation method Compound 9 was dissolved in acetonitrile and filtered with a 0.45 ⁇ m filter membrane to prepare a sample solution.
  • Mobile phase system A for CO 2 and B for IPA (0.1% NH 3 ⁇ H 2 O).
  • Compound 10 was obtained by using 8 g of 4,4-difluoropiperidine as a substrate and referring to the synthesis method of Example 8.
  • Triphenylphosphine (1.24 g, 4.71 mmol) and phthalimide (693 mg, 4.71 mmol) were added to a solution of 11 g (1.00 g, 3.14 mmol) in tetrahydrofuran (90 mL). After nitrogen replacement, the temperature was lowered to 0 ° C, and diisopropyl azodicarboxylate (952 mg, 4.71 mmol) was added. The mixture was reacted at room temperature for 16 h. Water (100 mL) was added and extracted with ethyl acetate (50 mL x 2). The organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, and filtered.
  • N,N-diisopropylethylamine (657 mg, 5.08 mmol) was added to 12d (639 mg, 1.27 mmol) and 5-fluoroisoindoline hydrochloride.
  • Salt (441 mg, 2.54 mmol) in dimethyl sulfoxide (15 mL), react at 90 ° C for 16 h. Cool to room temperature, add water (200 mL), extract with ethyl acetate (50 mL x 3), and combine the organic phases.
  • Compound 13 was purified by SFC on AD column (instrument and preparative column: Waters 150 Prep-SFC, preparative column model: Chiral AD column. Preparation method: Compound 13 was dissolved in methanol and filtered with a 0.45 ⁇ m filter membrane to prepare a sample solution. Mobile phase system: A for CO 2 and B for i-PrOH (0.1% NH 3 ⁇ H 2 O).
  • Compound 14 was prepared with 5d and 3-(trifluoromethyl)bicyclo[1.1.1]pentane-1-carboxylic acid as starting materials, referring to Example 13.
  • Compound 14 was separated by SFC on AD column (Instrument and preparation column: Waters Prep-SFC 150AP, preparation column model: Chiralcel AD column.
  • Preparation method Compound 14 was dissolved in acetonitrile and filtered with a 0.45 ⁇ m filter membrane to prepare a sample solution.
  • Mobile phase system A for CO 2 and B for ACN (0.1% trifluoroacetic acid).
  • Compound 15 was prepared with 11c and 3-(trifluoromethyl)bicyclo[1.1.1]pentane-1-carboxylic acid as starting materials, referring to Example 13.
  • Compound 15 was separated by SFC on AD column (Instrument and preparation column: Waters Prep-SFC 150AP, preparation column model: Chiralcel AD column.
  • Preparation method Compound 15 was dissolved in acetonitrile and filtered with a 0.45 ⁇ m filter membrane to prepare a sample solution.
  • Mobile phase system A for CO 2 and B for ACN (0.1% trifluoroacetic acid).
  • Example 16a was obtained with 8e as the starting material by referring to Example 13, and compound 16 was obtained with 16a as the starting material by referring to Example 12.
  • Compound 16 was separated by SFC on IG-H column (instrument and preparation column: Waters Prep-SFC 150AP, preparation column model: Daicel IG-H column.
  • Preparation method compound 16 was dissolved in DMF and filtered with a 0.45 ⁇ m filter membrane to prepare a sample solution.
  • Mobile phase system A for CO 2 and B for ethanol.
  • Compound 17 was prepared with 11c, 3-fluorobicyclo[1.1.1]pentane-1-carboxylic acid as substrates by referring to the synthesis method of Example 5.
  • Compound 17 was purified by SFC on AD column (Instrument and preparation column: Waters 150 Prep-SFC, preparation column model: Chiralpak AD column.
  • Preparation method Compound 17 was dissolved in acetonitrile and filtered with a 0.45 ⁇ m filter membrane to prepare a sample solution.
  • Mobile phase system A for CO 2 and B for MeOH (0.1% NH 3 ⁇ H 2 O).
  • diisopropyl azodicarboxylate 425 mg, 2.10 mmol was added to a solution of triphenylphosphine (550 mg, 2.10 mmol) and 17 g (600 mg, 1.75 mmol) in tetrahydrofuran (10 mL), and the mixture was reacted at room temperature for 16 h under nitrogen atmosphere. Water (30 mL) was added, and ethyl acetate (20 mL x 2) was extracted, and the organic phases were combined.
  • Methyl 6-chloro-3-fluoropyridine-2-carboxylate (227 mg, 1.20 mmol) and N,N-diisopropylethylamine (517 mg, 4.00 mmol) were added to dimethyl sulfoxide (5 mL) of 18b (342 mg, crude product) and reacted at 100°C for 16 h. After cooling to room temperature, water (20 mL) was added and extracted with ethyl acetate (30 mL x 2). The organic phases were combined, washed with saturated brine (40 mL x 3), dried over anhydrous sodium sulfate, and filtered.
  • Aqueous hydroxide solution (2N, 8.60mmol, 4.3mL) was added to a solution of 18c (442mg, 0.86mmol) in methanol (8mL) and tetrahydrofuran (8mL) and reacted at room temperature for 1h.
  • the mixture was concentrated under reduced pressure, and the pH of the residue was adjusted to 6-7 with 1N aqueous hydrochloric acid at 0°C, and extracted with ethyl acetate (20mL x 2).
  • the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product.
  • Compound 18 was purified by SFC on AD column (instrument and preparative column: Waters 150 Prep-SFC, preparative column model: Chiralcel AD column. Preparation method: Compound 18 was dissolved in acetonitrile and filtered with a 0.45 ⁇ m filter membrane to prepare a sample solution. Mobile phase system: A for CO 2 and B for IPA (0.1% NH 3 ⁇ H 2 O).
  • N-Bromosuccinimide (5.49 g, 30.83 mmol) was added to a solution of 20a (5.00 g, 30.83 mmol) in N,N-dimethylformamide (100 mL) and reacted at room temperature for 16 h.
  • the reaction solution was poured into water (500 mL), filtered, and the filter cake was rinsed with water (100 mL x 3) and dried under reduced pressure to obtain a crude product.
  • N,N-diisopropylethylamine (646 mg, 5.00 mmol) was added to 20i (494 mg, 1.25 mmol) and 5-fluoroisoindoline hydrochloride (434 mg, 2.50 mmol) in dimethyl sulfoxide (20 mL), and the mixture was reacted at 100 °C for 16 h. Water (200 mL) was added, and the mixture was extracted with dichloromethane (50 mL x 3), and the organic phases were combined.
  • the reaction solution from the previous step was slowly added dropwise to a mixed solution of dichloromethane (20 mL) and aqueous ammonia (20 mL), and the reaction was allowed to react at room temperature for 16 h.
  • the layers were separated, the aqueous phase was extracted with dichloromethane (50 mL x 2), and the organic phases were combined.
  • Compound 21 was prepared using 9e as raw material according to Example 15.
  • Compound 21 was separated by SFC on AS column (instrument and Preparation column: Waters Prep-SFC 150AP was used, and the preparation column model was: Chiralcel AS column.
  • Preparation method Compound 15 was dissolved in methanol and filtered with a 0.45 ⁇ m filter membrane to prepare a sample solution.
  • Mobile phase system A for CO 2 and B for methanol.
  • Compound 22 was obtained by using 22i and 6-chloro-3-fluoropyridine-2-carboxylic acid methyl ester as substrates according to Example 13.
  • 24j (500 mg, 1.06 mmol, 24j was prepared from 24a in reference to Example 13) was dissolved in isopropanol (24 mL) and water (4 mL), sodium borohydride (150 mg, 3.96 mmol) was added, and the mixture was reacted at room temperature for 2 h. At 0 °C, saturated ammonium chloride was added. The aqueous solution (3mL) was concentrated under reduced pressure to obtain a residue, and water was added and extracted with dichloromethane (40mL x 2). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product.
  • Compound 24 was obtained by using 24k and 6-chloro-3-fluoropyridine-2-carboxylic acid methyl ester as substrates with reference to Example 13.
  • Compound 22 was purified by SFC on AD column (instrument and preparation column: Waters 150 Prep-SFC, preparation column model: Chiral AD column. Preparation method: Compound 22 was dissolved in acetonitrile and filtered with a 0.45 ⁇ m filter membrane to prepare a sample solution. Mobile phase system: A for CO 2 and B for isopropanol (0.1% NH 3 ⁇ H 2 O).
  • Compound 26 was separated by SFC on OD column (instrument and preparation column: Waters 150 Prep-SFC, preparation column model: Chiral OD column. Preparation method: Compound 26 was dissolved in methanol and acetonitrile, and filtered with a 0.45 ⁇ m filter membrane to prepare a sample solution.
  • Mobile phase system A for CO 2 and B for methanol (0.1% NH 3 ⁇ H 2 O).
  • Compound 27 was prepared with 23e as raw material by referring to Example 26.
  • Compound 27 was separated by SFC on AS column (Instrument and preparation column: Waters 150 Prep-SFC, preparation column model: Chiral AS column.
  • Preparation method Compound 27 was dissolved in methanol and acetonitrile, and filtered with a 0.45 ⁇ m filter membrane to prepare a sample solution.
  • Mobile phase system A for CO 2 and B for methanol (0.1% NH 3 ⁇ H 2 O).
  • Method 1 MDA-MB-453 cells (ATCC, Cat#30-2008) were cultured in L-15 medium (with 10% FBS and 1% double antibody) under CO2- free conditions. When the cell confluence reached 80%-90%, the cells were collected and plated. The medium was discarded, and trypsin (Gibco, Cat#15400-054) was added to digest after rinsing with 1 ⁇ PBS. When the cells became round and began to fall off, the medium was added to terminate the digestion. The cells were blown down and transferred to a sterile centrifuge tube, centrifuged at 1000rpm for 3 minutes, and the supernatant was removed after centrifugation. The centrifuge tube was added with medium to resuspend the cells and counted.
  • L-15 medium with 10% FBS and 1% double antibody
  • the cell suspension was adjusted to an appropriate concentration and added to a 96-well cell culture plate (Corning, Cat#3903).
  • the compound was dissolved in DMSO to 10mM and stored for later use.
  • the compound mother solution was diluted in sequence during the experiment.
  • the compound was added 24 hours after plating, and the culture was continued for 4 days at 37°C and CO2 -free conditions.
  • CELL VIABILITY reagent Promega, Cat#G7573
  • the cells were gently shaken 5 times, and then the chemiluminescence readings were detected using a BMG microplate reader (PHERAstar FSX).
  • the cell proliferation inhibition rate was calculated according to the formula [(1-(RLU compound -RLU blank )/(RLU control -RLU blank )) ⁇ 100%].
  • the IC50 value was obtained by four-parameter nonlinear fitting using GraphPad Prism software.
  • MDA-MB-453 cells ATCC, Cat#HTB-131 were cultured with L-15 medium (supplemented with 10% FBS), and cells in the logarithmic growth phase were used for plating. The culture medium was discarded, and trypsin (Gibco, Cat#15400-054) was added for digestion after rinsing with 1 ⁇ PBS. When the cells became round and began to fall off, the culture medium was added to terminate the digestion. The cells were blown down and transferred to a sterile centrifuge tube, centrifuged at 1000rpm for 3 minutes, and the supernatant was discarded after the centrifugation. The culture medium was added to the centrifuge tube to resuspend the cells and count them.
  • the cell suspension was adjusted to an appropriate concentration and added to a 96-well cell culture plate (Greiner, Cat#655090).
  • the compound was dissolved in DMSO to 10mM and stored for later use.
  • the compound stock solution was diluted in sequence.
  • the compound was added 24 hours after plating, and the culture was continued for 7 days at 37°C and 5% CO2 .
  • CellTiter was added to each well.
  • AQueous One Solution Reagent Promega, Cat#G3582
  • the cell culture plate was placed in an incubator at 37°C, 0% CO2 for 4 hours, and then OD490nm was detected using a SpectraMax 340PC microplate reader.
  • the cell proliferation inhibition rate was calculated according to the formula [1-(RLU compound-RLU blank control)/(RLU solvent control-RLU blank control)] ⁇ 100%.
  • GraphPad Prism software was used to draw the inhibition curve and calculate the IC50 value.
  • the compounds of the present invention such as the compounds in the examples, have good inhibitory activity against MDA-MB-453 cells.
  • T47D cells (ATCC, Cat#HTB-133) were cultured with RPMI Medium 1640 (with 10% FBS, 1% double antibody and 0.02mg/mL bovine insulin) at 37°C and 5% CO2. When the cell confluence reached 80%-90%, the cells were collected and plated. The culture medium was discarded, and trypsin (Gibco, Cat#15400-054) was added for digestion after rinsing with 1 ⁇ PBS. When the cells became round and began to fall off, the culture medium was added to terminate the digestion. The cells were blown down and transferred to a sterile centrifuge tube, centrifuged at 1000rpm for 3 minutes, and the supernatant was discarded.
  • RPMI Medium 1640 with 10% FBS, 1% double antibody and 0.02mg/mL bovine insulin
  • the centrifuge tube was added with culture medium to resuspend the cells and counted. According to the counting results, the cell suspension was adjusted to an appropriate concentration and added to a 96-well cell culture plate (LABSELECT, Cat#11515). The compound was dissolved to 10mM with DMSO and stored for later use. The mother solution of the compound was diluted in sequence during the experiment. Compounds were added 24 hours after plating, and culture was continued for 4 days at 37°C and 5% CO2.
  • CELL VIABILITY reagent Promega, Cat#G7573
  • PHERAstar FSX BMG microplate reader
  • the compounds of the present invention such as the compounds in the examples, have good inhibitory activity against T47D cells.
  • DMSO serially dilute the compounds in the 384 PP Plate compound dilution plate, and use Echo to transfer 0.1 ⁇ L of the compound to the 384 reaction microplate (Optiplate 384), ensuring that the final DMSO concentration is 1% (duplicate wells).
  • Add 5 ⁇ L of 2X ATP and PIP2:3PS solution to each well, centrifuge at 1000 rpm for 1 minute, and incubate at 25°C for 180 minutes.
  • IC 50 is the compound concentration corresponding to 50% inhibition, see Table 2.
  • the compounds of the present invention have good inhibitory activity against PIK3CA[H1047R] enzyme, poor inhibitory activity against WT PIK3CA enzyme, and have good selectivity.
  • the inhibitory activity IC50 of compound 6-B, compound 26-B, and compound 27-B against PIK3CA[H1047R] enzyme are 77.4 nM, 54.8 nM, and 34.6 nM, respectively.
  • MDA-MB-453 cells (ATCC, Cat#HTB-131) were cultured with L-15 medium (with 10% FBS and 1% double antibody) at 37°C without CO2 . When the cell confluence reached 80%-90%, the cells were collected and plated. The culture medium was discarded, and trypsin (Gibco, Cat#15400-054) was added for digestion after rinsing with 1 ⁇ PBS. When the cells became round and began to fall off, the culture medium was added to terminate the digestion. The cells were blown down and transferred to a sterile centrifuge tube, centrifuged at 1000rpm for 3 minutes, and the supernatant was discarded after the centrifuge was completed.
  • L-15 medium with 10% FBS and 1% double antibody
  • the centrifuge tube was added with culture medium to resuspend the cells and counted. According to the counting results, the cell suspension was adjusted to an appropriate concentration and added to a 384-well plate (Perkin Elmer, Cat: 6007680). The compound was dissolved in DMSO to 10mM and stored for standby use. The compound mother solution was diluted in sequence during the experiment. The compound was added 18 hours after plating and cultured for 2h at 37°C without CO2 . After incubation, Lysis Buffer (Perkin Elmer, Cat: ALSU-PAKT-B500) was added to each well and shaken at room temperature for 10 minutes.
  • Lysis Buffer Perkin Elmer, Cat: ALSU-PAKT-B500
  • SKBR3 cells (ATCC, Cat#HTB-30) were cultured with McCoy's 5A Medium (10% FBS and 1% double antibody) at 37°C and 5% CO2 . When the cell confluence reached 80%-90%, the cells were collected and plated. The culture medium was discarded, and trypsin (Gibco, Cat#15400-054) was added to digest after rinsing with 1 ⁇ PBS. When the cells became round and began to fall off, the culture medium was added to terminate the digestion. The cells were blown down and transferred to a sterile centrifuge tube, centrifuged at 1000rpm for 3 minutes, and the supernatant was discarded.
  • McCoy's 5A Medium 10% FBS and 1% double antibody
  • the centrifuge tube was added with culture medium to resuspend the cells and counted. According to the counting results, the cell suspension was adjusted to an appropriate concentration and added to a 384-well plate (Perkin Elmer, Cat: 6007680). The compound was dissolved to 10mM with DMSO and stored for later use. The compound mother solution was diluted in sequence during the experiment. Compounds were added 18 hours after plating, and culture was continued for 2 hours at 37°C and 5% CO2 . After incubation, Lysis Buffer (Perkin Elmer, Cat: ALSU-PAKT-B500) was added to each well and shaken at room temperature for 10 minutes.
  • Lysis Buffer Perkin Elmer, Cat: ALSU-PAKT-B500
  • the compounds of the present invention have good inhibitory activity against P-AKT in MDA-MB-453 cells, but poor inhibitory activity against P-AKT in SKBR3 cells, and have good selectivity.
  • 3T3-L1 differentiated adipocytes were cultured in 96-well plates and starved for 2 hours in DMEM medium without FBS on the day of the experiment.
  • the compounds were dissolved in DMSO to 10mM and stored for later use.
  • the compound stock solution was diluted in sequence during the experiment. After washing the 3T3-L1 adipocytes with buffer, the compounds were added to the corresponding wells and incubated at 37°C in a 5% CO2 incubator for 60 minutes.
  • Test results The compounds of the present invention, such as the compounds in the examples, have poor inhibitory activity on glucose uptake in human adipocytes and do not cause side effects of hyperglycemia.
  • mice/compound Male Balb/c mice, about 22 g, 6 to 8 weeks old, 6 mice/compound, purchased from Chengdu Dashuo Experimental Animal Co., Ltd.
  • mice were randomly divided into groups according to body weight. They were fasted but not watered for 12-14 hours one day before administration and fed 4 hours after administration.
  • Intravenous administration solvent 5% DMA + 5% Solutol + 90% Saline; intragastric administration solvent: 0.5% MC
  • the compounds of the present invention such as the compounds in the examples, have good pharmacokinetic properties and have a high Exposure, good bioavailability.
  • mice Male SD rats, about 220 g, 6 to 8 weeks old, 6 rats per compound, purchased from Chengdu Dashuo Experimental Animal Co., Ltd.
  • Intravenous administration solvent 5% DMA + 5% Solutol + 90% Saline; intragastric administration solvent: 0.5% MC
  • the compounds of the present invention have good pharmacokinetic properties. Specifically, compound 6-B and compound 26-B have higher exposure in rats and good bioavailability.
  • the purpose of this study was to evaluate the effects of the test substances on the activities of five isoenzymes (CYP1A2, CYP2C19, CYP2D6 and CYP3A4) of human liver microsomal cytochrome P450 (CYP) using an in vitro test system.
  • Specific probe substrates of CYP450 isoenzymes were incubated with human liver microsomes and different concentrations of the test substances, and reduced nicotinamide adenine dinucleotide phosphate (NADPH) was added to initiate the reaction.
  • NADPH nicotinamide adenine dinucleotide phosphate
  • the metabolites produced by the specific substrates were quantitatively detected by treating the samples and using liquid chromatography-tandem mass spectrometry (LC-MS/MS) to determine the changes in CYP enzyme activity, calculate IC50 values, and evaluate the inhibitory potential of the test substances on each CYP enzyme subtype. .
  • LC-MS/MS liquid chromatography-tandem mass spectrometry
  • the compounds of the present invention have poor inhibitory activity against CYP enzymes. Specifically, the IC50 values of compound 25-B against CYP1A2, CYP2C19, CYP2D6 and CYP3A4 are all greater than 30 ⁇ M.
  • Cell line Chinese hamster ovary (CHO) cell line stably expressing hERG potassium channel
  • CHO (Chinese Hamster Ovary) cells stably expressing hERG potassium channels were used to record hERG potassium channel currents using the whole-cell patch clamp technique at room temperature.
  • the glass microelectrode was pulled from a glass electrode blank (BF150-86-10, Sutter) by a puller.
  • the tip resistance after perfusion of the electrode liquid was about 2-5M ⁇ .
  • the glass microelectrode was inserted into the amplifier probe to connect to the patch clamp amplifier.
  • the clamping voltage and data recording were controlled and recorded by pClamp 10 software through a computer, with a sampling frequency of 10kHz and a filter frequency of 2kHz.
  • the cell was clamped at -80mV, and the step voltage to induce the hERG potassium current (I hERG ) was given a 2s depolarization voltage from -80mV to +20mV, then repolarized to -50mV, and returned to -80mV after 1s.
  • This voltage stimulation was given every 10s, and the drug administration process was started after the hERG potassium current was determined to be stable (at least 1 minute).
  • Compounds were administered for at least 1 min at each tested concentration, and at least 2 cells (n ⁇ 2) were tested at each concentration.
  • Inhibition% represents the inhibition percentage of the compound on hERG potassium current
  • I and Io represent the amplitude of hERG potassium current after and before drug addition, respectively.
  • X is the Log value of the test sample detection concentration
  • Y is the inhibition percentage at the corresponding concentration
  • Bottom and Top are the minimum and maximum inhibition percentages, respectively.
  • the compounds of the present invention have poor hERG inhibitory activity. Specifically, the IC 50 of compound 6-B and compound 25-B for inhibiting hERG potassium channel current is greater than 20 ⁇ M.
  • test substance was incubated with microsomal proteins and coenzyme NADPH. After a certain time (5, 10, 20, 30, 60 min), ice-cold acetonitrile containing internal standard was added to terminate the reaction. The concentration of the test substance in the sample was detected by LC-MS/MS. T 1/2 was calculated by the ln value of the drug residual rate in the incubation system and the incubation time, and the liver microsomal intrinsic clearance CL int(Liver) was further calculated.
  • the compounds of the present invention such as the compounds in the examples, specifically compound 13-B and compound 25-B, are expressed in human liver microsomes. Good metabolic stability.
  • Test purpose To administer the test substance to beagle dogs by single-dose intravenous and oral gavage, determine the concentration of the test substance in beagle dog plasma, and evaluate the pharmacokinetic characteristics of the test substance in beagle dogs.
  • mice Male beagle dogs, about 8-11 kg, 6 per compound, purchased from Chengdu Dashuo Experimental Animal Co., Ltd.
  • test method On the day of the test, beagle dogs were randomly divided into groups according to body weight. They were fasted but not watered for 12-14 hours one day before administration and were fed 4 hours after administration.
  • Intravenous administration solvent 5% DMA + 5% Solutol + 90% Saline; intragastric administration solvent: 0.5% MC
  • the compounds of the present invention such as the compounds in the examples, have higher exposure and good pharmacokinetic properties in beagle dogs.
  • Intravenous administration solvent 5% DMA + 5% Solutol + 90% Saline; intragastric administration solvent: 0.5% MC (containing 0.5% Tween 80);
  • the compounds of the present invention have higher exposure and good pharmacokinetic properties in monkeys.

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

本发明涉及一种通式(I)所述的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,及其中间体和制备方法,以及在制备治疗与PI3Kα相关疾病的药物中的应用。

Description

一种含羧基的杂环衍生物及其在医药上的应用 技术领域
本发明涉及一种通式(I)所述的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,及其中间体和制备方法,以及在制备治疗与PI3Kα相关疾病的药物中的应用。
背景技术
磷脂酰肌醇3-激酶(PI3K)是一种脂质激酶,其由生长因子受体和整联蛋白激活后,调节细胞的增殖、存活和迁移。高达70%的乳腺癌具有某种形式的PI3K-AKT-mTOR途径的分子突变。PIK3CA(编码PI3K的p110α亚基)突变的激活在乳腺癌和实体瘤恶性肿瘤中非常普遍。
WT PI3Kα在调节机体葡萄糖稳态中起中心作用,患者体内的PBK抑制通常会导致高血糖和/或高胰岛素血症(Busaidy NL,et al,Management of metabolic effects associated with anticancer agents targeting the PBK-Akt-mTOR pathway.J Clin Oncol 2012;30:2919-28)。在具有突变的PI3Kα的癌症的情况下,开发对突变PI3Kα的选择性抑制剂能有效克服WT PI3Kα抑制时导致的胰岛素和/或葡萄糖的代偿性问题,这可以增加药物的给药窗口,从而选择性地抑制癌细胞中突变PI3Kα的病理信号,而不影响宿主本身的WT PI3Ka(Okkenhaug K,Graupera M,Vanhaesebroeck B.Targeting PBK in Cancer:Impact on Tumor Cells,Their Protective Stroma,Angiogenesis,and Immunotherapy.Cancer Discov.2016Oct;6(10):1090-1105)。
当前的PI3Kα抑制剂的开发集中在活性或正构位点,对野生型和突变型PI3Kα具有同等作用。其疗效受靶向WT PI3Kα介导的毒性的限制,包括剂量限制、高血糖和/或高胰岛素血症等。由于PI3Kα突变位置远离活性位点,突变选择性抑制剂一直难以获得。靶向已知突变附近的第二外周结合袋(例如H1047R)的抑制剂有望成为新型抗癌药物研发的靶点。
发明内容
本发明的目的就是提供一种能够抑制靶蛋白的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,及其中间体和制备方法,以及在制备治疗与PI3Kα抑制剂相关疾病的药物中的应用。
本发明化合物对PI3KαH1047R具有高度选择性,优于WT PI3Kα和其他PI3K亚型,并且不会引起高血糖副作用。
本发明化合物具有良好的生物利用度。
本发明提供一种通式(I)所述的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,其中
在某种实施方案中,X2选自O、S;
在某种实施方案中,X1选自O、S、NRx、C(Rx)2
在某种实施方案中,X2选自O时,X1选自O、S;
在某种实施方案中,X2选自S时,X1选自O、S;
在某种实施方案中,Y选自O、S、NRy
在某种实施方案中,Y选自O、S、NH;
在某种实施方案中,m选自1、2、3、4;
在某种实施方案中,m选自1、2;
在某种实施方案中,通式(I)化合物选自通式(II)、(III)、(IV)、(V)或(VI),
在某种实施方案中,通式(II)、(III)中,选自
在某种实施方案中,W选自C2-4炔基、所述的炔基任选被1至4个Rk所取代;
在某种实施方案中,W选自乙炔基、丙炔基、炔丙基、所述的乙炔基、丙炔基、炔丙基任选被1至4个Rk所取代;
在某种实施方案中,Z选自键或-C≡C-;
在某种实施方案中,Rx、Ry各自独立的选自H、氘、C1-6烷基、C2-6烯基、C2-6炔基、-C0-4亚烷基-C3-10碳环基、-C0-4亚烷基-4至10元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
在某种实施方案中,Rx、Ry各自独立的选自H、氘、C1-4烷基、C2-4烯基、C2-4炔基、-C0-4亚烷基-C3-8碳环基、-C0-4亚烷基-4至8元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
在某种实施方案中,Rx、Ry各自独立的选自H、氘、甲基、乙基、丙基、异丙基、乙烯基、乙炔基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、哌嗪基、吗啉基,所述的甲基、乙基、丙基、异丙基、乙烯基、乙炔基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、哌嗪基、吗啉基任选被1至4个Rk所取代;
在某种实施方案中,R1、R2、R3、R4各自独立地选自H、氘、卤素、OH、CN、NO2、NH2、C1-6烷基、OC1-6烷基、SC1-6烷基、C2-6烯基、C2-6炔基、NHC1-6烷基、N(C1-6烷基)2、-C0-4亚烷基-C(=O)R1a、-C0-4亚烷基-S(=O)2R1a、-C0-4亚烷基-P(=O)R1aR1b、-O-C0-4亚烷基-C3-10碳环基、-O-C0-4亚烷基-4至10元杂环基、-S-C0-4亚烷基-C3-10碳环基、-S-C0-4亚烷基-4至10元杂环基、-NH-C0-4亚烷基-C3-10碳环基、-NH-C0-4亚烷基-4至10元杂环基、-C0-4亚烷基-C3-10碳环基、-C0-4亚烷基-4至10元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
在某种实施方案中,R1、R2、R3、R4各自独立地选自H、氘、卤素、OH、CN、NO2、NH2、C1-4烷基、OC1-4烷基、SC1-4烷基、C2-4烯基、C2-4炔基、NHC1-4烷基、N(C1-4烷基)2、-C0-4亚烷基-C(=O)R1a、-C0-4亚烷基-S(=O)2R1a、-C0-4亚烷基-P(=O)R1aR1b、-O-C0-4亚烷基-C3-8碳环基、-O-C0-4亚烷基-4至8元杂环基、-S-C0-4亚烷基-C3-8碳环基、-S-C0-4亚烷基-4至8元杂环基、-NH-C0-4亚烷基-C3-8碳环基、-NH-C0-4亚烷基-4至8元杂环基、-C0-4亚烷基-C3-8碳环基、-C0-4亚烷基-4至8元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
在某种实施方案中,R1、R2、R3、R4各自独立地选自H、氘、F、Cl、Br、I、OH、CN、NO2、NH2、NHCH3、N(CH3)2、甲基、乙基、丙基、异丙基、乙烯基、乙炔基、丙炔基、炔丙基、甲硫基、-C(=O)R1a、-S(=O)2R1a、-P(=O)R1aR1b、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基,所述的甲基、乙基、丙基、异丙基、乙烯基、乙炔基、丙炔基、炔丙基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基任选被1至4个Rk所取代;
在某种实施方案中,R5、R6各自独立的选自H、氘、卤素、OH、CN、NO2、NH2、C1-6烷基、OC1-6烷基、SC1-6烷基、C2-6烯基、C2-6炔基、NHC1-6烷基、N(C1-6烷基)2、C0-4亚烷基-5至10元杂芳基、C0-4亚烷基-C3-10碳环基、C0-4亚烷基-4至10元杂环基,所述的烷基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
在某种实施方案中,R5、R6各自独立的选自H、氘、卤素、OH、CN、NO2、NH2、C1-4烷基、OC1-4烷基、SC1-4烷基、C2-4烯基、C2-4炔基、NHC1-4烷基、N(C1-4烷基)2、C0-4亚烷基-5至8元杂芳基、C0-4亚烷基-C3-8碳环基、C0-4亚烷基-4至8元杂环基,所述的烷基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
在某种实施方案中,R5、R6各自独立的选自H、氘、F、Cl、Br、I、OH、CN、NO2、NH2、 NHCH3、N(CH3)2、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基,所述的甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基任选被1至4个Rk所取代;
在某种实施方案中,环A选自C3-12碳环基、4至12元杂环基、C6-10芳基或5至10元杂芳基,所述的碳环基、杂环基、芳基或杂芳基任选被1至4个选自Ra所取代,环A以碳碳键与Z相连;
在某种实施方案中,环A各自独立的选自4-7元杂单环、5-12元杂并环、5-12元杂螺环、7-10元杂桥环、C3-8单碳环基、C6-12并环烷基、C6-12元螺环烷基、C5-12元桥环烷基、5至10元杂芳基、苯并C3-8碳环基、苯并4至8元杂环基,环A以碳碳键与Z相连,所述的环A任选被1至4个选自Ra所取代;
在某种实施方案中,环A各自独立的选自任选取代的如下结构之一:环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、四氢呋喃基、四氢吡喃基、1,3-二氧环戊烷基、1,4-二氧环己基、吡咯烷基、哌啶基、哌嗪基、吗啉基、苯基、吡啶基、吡嗪基、哒嗪基、嘧啶基、吡唑基、吡咯基、咪唑基、噻吩基、噻唑基、呋喃基、噁唑基、异噁唑基、苯并吡咯基、苯并噻吩基、苯并呋喃基、苯并噻唑基、苯并吡唑基、苯并咪唑基、喹啉基、异喹啉基、吡啶并吡啶基、吡啶并噻唑基、吡啶并呋喃基、吡啶并吡咯基、异吲哚啉基、吲哚啉基、 当被取代时,被1至3个Ra取代;
在某种实施方案中,各自独立的选自任选被1至4个Ra取代的如下结构之一:
在某种实施方案中,环B选自C3-12碳环基、4至12元杂环基、C6-10芳基或5至10元杂芳基,所述的碳环基、杂环基、芳基或杂芳基任选被1至4个选自Rb所取代;
在某种实施方案中,环B各自独立的选自4-7元杂单环、5-12元杂并环、5-12元杂螺环、7-10元杂桥环、C3-8单碳环基、C6-14并环烷基、C6-12元螺环烷基、C5-12元桥环烷基、苯并C3-8碳环基、苯并3至8元杂环基、C6-10芳基、5-10元杂芳基,所述的环B任选被1至4个Rb所取代;
在某种实施方案中,环B各自独立的选自C6-10芳基、5-10元杂芳基,所述的环B任选被1至4个Rb所取代;
在某种实施方案中,环B各自独立的选自任选取代的如下结构之一:苯、萘、吡啶、吡嗪、哒嗪、嘧啶、当被取代时,被1至 3个Rb所取代;
在某种实施方案中,各自独立的选自
在某种实施方案中,各自独立的选自
在某种实施方案中,各自独立的选自在某种实施方案中,B1、B2、B3、B4、B6各自独立的选自N或CRb1,B1、B2、B3、B4、B6中至多三个选自N;
在某种实施方案中,B1、B2、B3、B6各自独立的选自N或CRb1,B1、B2、B3、B6中至多两个选自N;
在某种实施方案中,B1、B2、B3、B6中三个选自CRb1,其余一个选自N;
在某种实施方案中,各自独立的选自
在某种实施方案中,环C1、C2、C3各自独立的选自4-8元杂环或C4-8碳环,所述的环C1、C2、C3任选被1至4个Rk所取代;
在某种实施方案中,环C1、C2、C3各自独立的选自任选被1至4个Rk取代的如下基团之一:环丁烯基、环戊烯基、环己烯基、环庚烯基、氧杂环丁烯基、氧杂环戊烯基、氧杂环己烯基、氮杂环丁烯基、氮杂环戊烯基、氮杂环己烯基、1,3-二氧环戊烯基、1,4-二氧环己烯基;
在某种实施方案中,环C4选自4-8元杂环,所述的环C4任选被1至4个Rk所取代;
在某种实施方案中,环C5选自4-8元含氮杂环,所述的环C5任选被1至4个Rk所取代;
在某种实施方案中,环C5选自任选被1至4个Rk取代的如下基团之一:氮杂环丁基、吡咯烷基、哌啶基、吗啉基;
在某种实施方案中,选自其右端与环B相连接;
在某种实施方案中,选自n选自1、2、3;
在某种实施方案中,B1、B2、B3、B4各自独立的选自N或CRb1,B1、B2、B3、B4中至多两个选自N;
在某种实施方案中,B5各自独立地选自N、O、S、S(O)2、CRb2、NRb2或C(Rb2)2
在某种实施方案中,Rb2各自独立的选自H或Rk
在某种实施方案中,Rb1各自独立的选自H或Rb
在某种实施方案中,选自
在某种实施方案中,选自
在某种实施方案中,选自
在某种实施方案中,选自
在某种实施方案中,选自
在某种实施方案中,B1、B2、B3各自独立的选自CRb1
在某种实施方案中,B1、B2、B3中两个选自CRb1,其余选自N;
在某种实施方案中,选自
在某种实施方案中,选自
在某种实施方案中,选自
在某种实施方案中,Ra、Rb各自独立的选自氘、卤素、=O、OH、CN、NO2、NH2、C1-6烷基、OC1-6烷基、SC1-6烷基、C2-6烯基、C2-6炔基、NHC1-6烷基、N(C1-6烷基)2、-C0-4亚烷基-C(=O)R1a、-C0-4亚烷基-S(=O)2R1a、-C0-4亚烷基-P(=O)R1aR1b、-O-C0-4亚烷基-C3-10碳环基、-O-C0-4亚烷基-4至10元杂环基、-S-C0-4亚烷基-C3-10碳环基、-S-C0-4亚烷基-4至10元杂环基、-NH-C0-4亚烷基-C3-10碳环基、-NH-C0-4亚烷基-4至10元杂环基、-C0-4亚烷基-C3-10碳环基、-C0-4亚烷基-4至10元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
在某种实施方案中,Ra、Rb各自独立的选自氘、氘、卤素、=O、OH、CN、NO2、NH2、C1-4烷基、OC1-4烷基、SC1-4烷基、C2-4烯基、C2-4炔基、NHC1-4烷基、N(C1-4烷基)2、-C0-4亚烷基-C(=O)R1a、-C0-4亚烷基-S(=O)2R1a、-C0-4亚烷基-P(=O)R1aR1b、-O-C0-4亚烷基-C3-8碳环基、-O-C0-4亚烷基-4至8元杂环基、-S-C0-4亚烷基-C3-8碳环基、-S-C0-4亚烷基-4至8元杂环基、-NH-C0-4亚烷基-C3-8碳环基、-NH-C0-4亚烷基-4至8元杂环基、-C0-4亚烷基-C3-8碳环基、-C0-4亚烷基-4至8元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
在某种实施方案中,Ra各自独立的选自氘、F、Cl、Br、I、OH、=O、NH2、CN、NO2、NHCH3、N(CH3)2、COOH、CONH2或任选被1至4个Rk取代的如下基团之一:甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、苯基、吡啶基、吡嗪基、哒嗪基、嘧啶基、吡唑基、吡咯基、咪唑基、噻吩基、噻唑基、呋喃基、噁唑基、异噁唑基;
在某种实施方案中,Ra各自独立的选自氘、F、Cl、Br、I、OH、=O、NH2、CN、NO2、NHCH3、N(CH3)2、COOH、CONH2、CF3、甲基、乙基、异丙基、环丙基;
在某种实施方案中,Rb各自独立的选自氘、F、Cl、Br、I、OH、=O、CN、NH2、NO2、NHCH3、N(CH3)2、COOH、CONH2、-CH2-C(=O)R1a、-CH2-S(=O)2R1a、-CH2-P(=O)R1aR1b、-C(=O)R1a、-S(=O)2R1a、-P(=O)R1aR1b、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基、-CH2-环丙基、-CH2-环丁基、-CH2-吡咯烷基、-O-环丙基、-O-环丁基、-O-吡咯烷基、-S-环丙基、-S-环丁基、-NH-环丙基、-NH-环丁基、-OCH2-环丙基、咪唑、吡唑、吡咯或噻吩,所述的甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基、咪唑、吡唑、吡咯或噻吩任选被1至4个Rk所取代;
在某种实施方案中,R1a、R1b各自独立的选自H、OH、NH2、C1-6烷基、C1-6烷氧基、NHC1-6烷基、N(C1-6烷基)2、C3-8碳环基、4至8元杂环基、C6-10芳基、5至10元杂芳基、-O-C3-8碳环基、-O-4至8元杂环基,所述的烷基、烷氧基、碳环基、杂环基、芳基或杂芳基任选被1至4个Rk所取代;
在某种实施方案中,R1a、R1b各自独立的选自H、OH、NH2、C1-4烷基、C1-4烷氧基、NHC1-4烷基、N(C1-4烷基)2、C3-6碳环基、4至8元杂环基、C6-10芳基、5至10元杂芳基、-O-C3-8碳环基、-O-4至8元杂环基,所述的烷基、烷氧基、碳环基、杂环基、芳基或杂芳基任选被1至4个Rk所取代;
在某种实施方案中,R1a、R1b各自独立的选自H、OH、NH2、NHCH3、N(CH3)2、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基、-O-环丙基、-O-环丁基、咪唑、吡唑、吡咯或噻吩,所述的甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基、咪唑、吡唑、吡咯或噻吩任选被1至3个Rk所取代;
在某种实施方案中,R2与R3、R3与R4、R4与R5、R6与Ry、R6与Rb中至少有一组和与其相连的原子共同形成C3-12碳环基或3至12元杂环基,所述的碳环基或杂环基任选被1至4个Rk所取代;
在某种实施方案中,R2与R3、R3与R4、R4与R5、R6与Ry、R6与Rb中至少有一组和与其相连的原子共同形成环丁烯基、环戊烯基、环己烯基、氮杂环丁烯基、氧杂环丁烯基、氧杂环戊烯基、氮杂环戊烯基、氮杂环己烯基、1,3-二氧环戊烯基、1,4-二氧环己烯基,所述的环丁烯基、环戊烯基、环己烯基、氮杂环丁烯基、氧杂环丁烯基、氧杂环戊烯基、氮杂环戊烯基、氮杂环己烯基、1,3-二氧环戊烯基、1,4-二氧环己烯基任选被1至4个Rk所取代;
在某种实施方案中,Rk各自独立的选自氘、卤素、OH、=O、CN、NH2、COOH、CONH2、C1-6烷基、OC1-6烷基、SC1-6烷基、C2-6烯基、C2-6炔基、NHC1-6烷基、N(C1-6烷基)2、-O-C3-6碳环、-O-3至7元杂环、-NH-C3-6碳环、-NH-3至7元杂环、-C0-4亚烷基-C3-6碳环、-C0-4亚烷基-3至7元杂环,所述的烷基、亚烷基、烯基、炔基、碳环或杂环任选被1至4个选自氘、卤素、=O、CN、OH、NH2、C1-6烷基、C1-6烷氧基的取代基所取代;
在某种实施方案中,Rk各自独立的选自氘、卤素、OH、=O、CN、NH2、COOH、CONH2、C1-4烷基、OC1-4烷基、SC1-4烷基、C2-4烯基、C2-4炔基、NHC1-4烷基、N(C1-4烷基)2、-O-C3-6碳环、-O-3至7元杂环、-NH-C3-6碳环、-NH-3至7元杂环、-C0-4亚烷基-C3-6碳环、-C0-4亚烷基-3至7元杂环,所述的烷基、亚烷基、烯基、炔基、碳环或杂环任选被1至4个选自氘、卤素、=O、CN、OH、NH2、C1-4烷基、C1-4烷氧基的取代基所取代;
在某种实施方案中,Rk各自独立的选自氘、F、Cl、Br、I、OH、=O、CN、NH2、COOH、CONH2、NHCH3、N(CH3)2、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、甲硫基、乙烯基、乙炔基、丙炔基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吡唑基、吡咯基、吗啉基,所述的甲基、乙基、丙基、异丙基、甲氧基、乙氧基、甲硫基、乙烯基、乙炔基、丙炔基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吡唑基、吡咯基、吗啉基任选被1至4个选自氘、F、Cl、Br、I、=O、CN、OH、NH2、C1-4烷基、C1-4烷氧基的取代 基所取代;
在某种实施方案中,Rk各自独立的选自氘、F、Cl、Br、I、OH、=O、COOH、CN、CHF2、CH2F、CF3、OCHF2、OCF3、NH2、NHCH3、N(CH3)2、CH2OH、OCH2OH、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、甲硫基、乙烯基、乙炔基、丙炔基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吡唑基、吡咯基、吗啉基。
作为本发明的第一种实施方案,前述通式(I)所示的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,
X2选自O、S;
X1选自O、S、NRx、C(Rx)2
Y选自O、S、NRy
W选自C2-4炔基、所述的炔基任选被1至4个Rk所取代;
Z选自键或-C≡C-;
m选自1、2、3、4;
Rx、Ry各自独立的选自H、氘、C1-6烷基、C2-6烯基、C2-6炔基、-C0-4亚烷基-C3-10碳环基、-C0-4亚烷基-4至10元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
环A选自C3-12碳环基、4至12元杂环基、C6-10芳基或5至10元杂芳基,所述的碳环基、杂环基、芳基或杂芳基任选被1至4个选自Ra所取代;
环B选自C3-12碳环基、4至12元杂环基、C6-10芳基或5至10元杂芳基,所述的碳环基、杂环基、芳基或杂芳基任选被1至4个选自Rb所取代;
Ra、Rb各自独立的选自氘、卤素、=O、OH、CN、NO2、NH2、C1-6烷基、OC1-6烷基、SC1-6烷基、C2-6烯基、C2-6炔基、NHC1-6烷基、N(C1-6烷基)2、-C0-4亚烷基-C(=O)R1a、-C0-4亚烷基-S(=O)2R1a、-C0-4亚烷基-P(=O)R1aR1b、-O-C0-4亚烷基-C3-10碳环基、-O-C0-4亚烷基-4至10元杂环基、-S-C0-4亚烷基-C3-10碳环基、-S-C0-4亚烷基-4至10元杂环基、-NH-C0-4亚烷基-C3-10碳环基、-NH-C0-4亚烷基-4至10元杂环基、-C0-4亚烷基-C3-10碳环基、-C0-4亚烷基-4至10元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
R1、R2、R3、R4各自独立地选自H、氘、卤素、OH、CN、NO2、NH2、C1-6烷基、OC1-6烷基、SC1-6烷基、C2-6烯基、C2-6炔基、NHC1-6烷基、N(C1-6烷基)2、-C0-4亚烷基-C(=O)R1a、-C0-4亚烷基-S(=O)2R1a、-C0-4亚烷基-P(=O)R1aR1b、-O-C0-4亚烷基-C3-10碳环基、-O-C0-4亚烷基-4至10元杂环基、-S-C0-4亚烷基-C3-10碳环基、-S-C0-4亚烷基-4至10元杂环基、-NH-C0-4亚烷基-C3-10碳环基、-NH-C0-4亚烷基-4至10元杂环基、-C0-4亚烷基-C3-10碳环基、-C0-4亚烷基-4至10元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
R5、R6各自独立的选自H、氘、卤素、OH、CN、NO2、NH2、C1-6烷基、OC1-6烷基、SC1-6烷基、C2-6烯基、C2-6炔基、NHC1-6烷基、N(C1-6烷基)2、C0-4亚烷基-5至10元杂芳基、C0-4亚烷基-C3-10碳环基、C0-4亚烷基-4至10元杂环基,所述的烷基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
R1a、R1b各自独立的选自H、OH、NH2、C1-6烷基、C1-6烷氧基、NHC1-6烷基、N(C1-6烷基)2、 C3-8碳环基、4至8元杂环基、C6-10芳基、5至10元杂芳基、-O-C3-8碳环基、-O-4至8元杂环基,所述的烷基、烷氧基、碳环基、杂环基、芳基或杂芳基任选被1至4个Rk所取代;
R2与R3、R3与R4、R4与R5、R6与Ry、R6与Rb中至少有一组和与其相连的原子共同形成C3-12碳环基或3至12元杂环基,所述的碳环基或杂环基任选被1至4个Rk所取代;
Rk各自独立的选自氘、卤素、OH、=O、CN、NH2、COOH、CONH2、C1-6烷基、OC1-6烷基、SC1-6烷基、C2-6烯基、C2-6炔基、NHC1-6烷基、N(C1-6烷基)2、-O-C3-6碳环、-O-3至7元杂环、-NH-C3-6碳环、-NH-3至7元杂环、-C0-4亚烷基-C3-6碳环、-C0-4亚烷基-3至7元杂环,所述的烷基、亚烷基、烯基、炔基、碳环或杂环任选被1至4个选自氘、卤素、=O、CN、OH、NH2、C1-6烷基、C1-6烷氧基的取代基所取代。
作为本发明的第二种实施方案,前述通式(I)、(II)、(III)、(IV)、(V)、(VI)所示的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,
Rx、Ry各自独立的选自H、氘、C1-4烷基、C2-4烯基、C2-4炔基、-C0-4亚烷基-C3-8碳环基、-C0-4亚烷基-4至8元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
R1a、R1b各自独立的选自H、OH、NH2、C1-4烷基、C1-4烷氧基、NHC1-4烷基、N(C1-4烷基)2、C3-6碳环基、4至8元杂环基、C6-10芳基、5至10元杂芳基、-O-C3-8碳环基、-O-4至8元杂环基,所述的烷基、烷氧基、碳环基、杂环基、芳基或杂芳基任选被1至4个Rk所取代;
环A各自独立的选自4-7元杂单环、5-12元杂并环、5-12元杂螺环、7-10元杂桥环、C3-8单碳环基、C6-12并环烷基、C6-12元螺环烷基、C5-12元桥环烷基、5至10元杂芳基、苯并C3-8碳环基、苯并4至8元杂环基,环A以碳碳键与Z相连,所述的环A任选被1至4个选自Ra所取代;
环B各自独立的选自4-7元杂单环、5-12元杂并环、5-12元杂螺环、7-10元杂桥环、C3-8单碳环基、C6-14并环烷基、C6-12元螺环烷基、C5-12元桥环烷基、苯并C3-8碳环基、苯并3至8元杂环基、C6-10芳基、5-10元杂芳基,所述的环B任选被1至4个Rb所取代;
环C1、C2、C3各自独立的选自4-8元杂环或C4-8碳环,所述的环C1、C2、C3任选被1至4个Rk所取代;
环C4选自4-8元杂环,所述的环C4任选被1至4个Rk所取代;
环C5选自4-8元含氮杂环,所述的环C5任选被1至4个Rk所取代;
Rk各自独立的选自氘、卤素、OH、=O、CN、NH2、COOH、CONH2、C1-4烷基、OC1-4烷基、SC1-4烷基、C2-4烯基、C2-4炔基、NHC1-4烷基、N(C1-4烷基)2、-O-C3-6碳环、-O-3至7元杂环、-NH-C3-6碳环、-NH-3至7元杂环、-C0-4亚烷基-C3-6碳环、-C0-4亚烷基-3至7元杂环,所述的烷基、亚烷基、烯基、炔基、碳环或杂环任选被1至4个选自氘、卤素、=O、CN、OH、NH2、C1-4烷基、C1-4烷氧基的取代基所取代;
其余取代基的定义与本发明方案一中所述一致。
作为本发明的第三种实施方案,前述通式(I)、(II)、(III)、(IV)、(V)、(VI)所示的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,
环B各自独立的选自C6-10芳基、5-10元杂芳基,所述的环B任选被1至4个Rb所取代;
或者选自n选自1、2、3;
B1、B2、B3、B4各自独立的选自N或CRb1,B1、B2、B3、B4中至多两个选自N;
B5各自独立的选自N、O、S、CRb2、S(O)2、NRb2或C(Rb2)2
Rb2各自独立的选自H或Rk
Rb1各自独立的选自H或Rb
R1、R2、R3、R4各自独立地选自H、氘、卤素、OH、CN、NO2、NH2、C1-4烷基、OC1-4烷基、SC1-4烷基、C2-4烯基、C2-4炔基、NHC1-4烷基、N(C1-4烷基)2、-C0-4亚烷基-C(=O)R1a、-C0-4亚烷基-S(=O)2R1a、-C0-4亚烷基-P(=O)R1aR1b、-O-C0-4亚烷基-C3-8碳环基、-O-C0-4亚烷基-4至8元杂环基、-S-C0-4亚烷基-C3-8碳环基、-S-C0-4亚烷基-4至8元杂环基、-NH-C0-4亚烷基-C3-8碳环基、-NH-C0-4亚烷基-4至8元杂环基、-C0-4亚烷基-C3-8碳环基、-C0-4亚烷基-4至8元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
R5、R6各自独立的选自H、氘、卤素、OH、CN、NO2、NH2、C1-4烷基、OC1-4烷基、SC1-4烷基、C2-4烯基、C2-4炔基、NHC1-4烷基、N(C1-4烷基)2、C0-4亚烷基-5至8元杂芳基、C0-4亚烷基-C3-8碳环基、C0-4亚烷基-4至8元杂环基,所述的烷基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
Ra、Rb各自独立的选自氘、氘、卤素、=O、OH、CN、NO2、NH2、C1-4烷基、OC1-4烷基、SC1-4烷基、C2-4烯基、C2-4炔基、NHC1-4烷基、N(C1-4烷基)2、-C0-4亚烷基-C(=O)R1a、-C0-4亚烷基-S(=O)2R1a、-C0-4亚烷基-P(=O)R1aR1b、-O-C0-4亚烷基-C3-8碳环基、-O-C0-4亚烷基-4至8元杂环基、-S-C0-4亚烷基-C3-8碳环基、-S-C0-4亚烷基-4至8元杂环基、-NH-C0-4亚烷基-C3-8碳环基、-NH-C0-4亚烷基-4至8元杂环基、-C0-4亚烷基-C3-8碳环基、-C0-4亚烷基-4至8元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
其余取代基的定义与本发明方案一、二中所述一致。
作为本发明的第四种实施方案,前述通式(I)、(II)、(III)、(IV)、(V)、(VI)所示的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,
X1选自O、S、NRx
Rx、Ry各自独立的选自H、氘、甲基、乙基、丙基、异丙基、乙烯基、乙炔基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、哌嗪基、吗啉基,所述的甲基、乙基、丙基、异丙基、乙烯基、乙炔基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、哌嗪基、吗啉基任选被1至4个Rk所取代;
R1a、R1b各自独立的选自H、OH、NH2、NHCH3、N(CH3)2、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基、-O-环丙基、-O-环丁基、咪唑、吡唑、吡咯或噻吩,所述的甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基、咪唑、吡唑、吡咯或噻吩任选被1至3个Rk所 取代;
R1、R2、R3、R4各自独立地选自H、氘、F、Cl、Br、I、OH、CN、NO2、NH2、NHCH3、N(CH3)2、甲基、乙基、丙基、异丙基、乙烯基、乙炔基、丙炔基、炔丙基、甲硫基、-C(=O)R1a、-S(=O)2R1a、-P(=O)R1aR1b、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基,所述的甲基、乙基、丙基、异丙基、乙烯基、乙炔基、丙炔基、炔丙基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基任选被1至4个Rk所取代;
R5、R6各自独立的选自H、氘、F、Cl、Br、I、OH、CN、NO2、NH2、NHCH3、N(CH3)2、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基,所述的甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基任选被1至4个Rk所取代;
Ra各自独立的选自氘、F、Cl、Br、I、OH、=O、NH2、CN、NO2、NHCH3、N(CH3)2、COOH、CONH2或任选被1至4个Rk取代的如下基团之一:甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、苯基、吡啶基、吡嗪基、哒嗪基、嘧啶基、吡唑基、吡咯基、咪唑基、噻吩基、噻唑基、呋喃基、噁唑基、异噁唑基;
Rb各自独立的选自氘、F、Cl、Br、I、OH、=O、CN、NH2、NO2、NHCH3、N(CH3)2、COOH、CONH2、-CH2-C(=O)R1a、-CH2-S(=O)2R1a、-CH2-P(=O)R1aR1b、-C(=O)R1a、-S(=O)2R1a、-P(=O)R1aR1b、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基、-CH2-环丙基、-CH2-环丁基、-CH2-吡咯烷基、-O-环丙基、-O-环丁基、-O-吡咯烷基、-S-环丙基、-S-环丁基、-NH-环丙基、-NH-环丁基、-OCH2-环丙基、咪唑、吡唑、吡咯或噻吩,所述的甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基、咪唑、吡唑、吡咯或噻吩任选被1至4个Rk所取代;
环C1、C2、C3各自独立的选自任选被1至4个Rk取代的如下基团之一:环丁烯基、环戊烯基、环己烯基、环庚烯基、氧杂环丁烯基、氧杂环戊烯基、氧杂环己烯基、氮杂环丁烯基、氮杂环戊烯基、氮杂环己烯基、1,3-二氧环戊烯基、1,4-二氧环己烯基;
环C5选自任选被1至4个Rk取代的如下基团之一:氮杂环丁基、吡咯烷基、哌啶基、吗啉基;
其余取代基的定义与本发明方案一、二、三中所述一致。
作为本发明的第五种实施方案,前述通式(I)、(II)、(III)、(IV)、(V)、(VI)所示的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,
W选自乙炔基、丙炔基、炔丙基、所述的乙炔基、丙炔基、炔丙基任选被1至4个Rk所取代;
Y选自O、S、NH;
Rk各自独立的选自氘、F、Cl、Br、I、OH、=O、CN、NH2、COOH、CONH2、NHCH3、N(CH3)2、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、甲硫基、乙烯基、乙炔基、丙炔基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吡唑基、吡咯基、吗啉基,所述的甲基、乙基、丙基、异丙基、甲氧基、乙氧基、甲硫基、乙烯基、乙炔基、丙炔基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吡唑基、吡咯基、吗啉基任选被1至4个选自氘、F、Cl、Br、I、=O、CN、OH、NH2、C1-4烷基、C1-4烷氧基的取代基所取代;
环B各自独立的选自任选取代的如下结构之一:苯、萘、吡啶、吡嗪、哒嗪、嘧啶、 当被取代时,被1至3个Rb所取代;
环A各自独立的选自任选取代的如下结构之一:环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、四氢呋喃基、四氢吡喃基、1,3-二氧环戊烷基、1,4-二氧环己基、吡咯烷基、哌啶基、哌嗪基、吗啉基、苯基、吡啶基、吡嗪基、哒嗪基、嘧啶基、吡唑基、吡咯基、咪唑基、噻吩基、噻唑基、呋喃基、噁唑基、异噁唑基、苯并吡咯基、苯并噻吩基、苯并呋喃基、苯并噻唑基、苯并吡唑基、苯并咪唑基、喹啉基、异喹啉基、吡啶并吡啶基、吡啶并噻唑基、吡啶并呋喃基、吡啶并吡咯基、异吲哚啉基、吲哚啉基、 当被取代时,被1至3个Ra取代;
或者选自
其余取代基的定义与本发明方案一、二、三、四中所述一致。
作为本发明的第六种实施方案,前述通式(I)、(II)、(III)、(IV)、(V)、(VI)所示的化合物 或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,
选自
选自
选自
n1、n2、n3各自独立的选自1、2、3;
各自独立的选自
B1、B2、B3、B6各自独立的选自N或CRb1,B1、B2、B3、B6中至多两个选自N;
或者选自
选自其右端与环B相连接;
其余取代基的定义与本发明方案一、二、三、四、五中所述一致。
作为本发明的第七种实施方案,前述通式(I)、(II)、(III)、(IV)、(V)、(VI)所示的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,
选自
选自
选自
各自独立的选自
或者选自
选自其右端与环B相连接;
其余取代基的定义与本发明方案一、二、三、四、五、六中所述一致。
本发明涉及一种下述化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,其中该化合物选自如下表A结构之一:
表A


















本发明涉及一种药物组合物,包括本发明所述的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,以及药学上可接受的载体。
本发明涉及一种药物组合物,包括治疗有效量的本发明上述的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,以及药学上可接受的载体。
本发明涉及一种本发明所述的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶在用于制备治疗与PI3Kα相关疾病的药物中的应用,优选用于制备肿瘤或癌症(例如乳腺癌)药物中的应用。
在一些实施方案中,本发明的药物组合物可以为单位制剂形式(单位制剂中主药的量也被称为“制剂规格”)。
本申请中所述“有效量”或“治疗有效量”是指给予足够量的本申请公开的化合物,其将在某种程度上缓解所治疗的疾病或病症(例如治疗与PI3Kα相关疾病如肿瘤)的一种或多种症状。在一些 实施方案中,结果是减少和/或缓和疾病的体征、症状或原因,或生物系统的任何其它希望的改变。例如,针对治疗用途的“有效量”是提供临床上显著的疾病症状降低所需的包含本申请公开的化合物的量。治疗有效量的实例包括但不限于1-1500mg、1-1200mg、1-1000mg、1-900mg、1-800mg、1-700mg、1-600mg、2-600mg、3-600mg、4-600mg、5-600mg、6-600mg、10-600mg、20-600mg、25-600mg、30-600mg、40-600mg、50-600mg、60-600mg、70-600mg、75-600mg、80-600mg、90-600mg、100-600mg、200-600mg、1-500mg、2-500mg、3-500mg、4-500mg、5-500mg、6-500mg、10-500mg、20-500mg、25-500mg、30-500mg、40-500mg、50-500mg、60-500mg、70-500mg、75-500mg、80-500mg、90-500mg、100-500mg、125-500mg、150-500mg、200-500mg、250-500mg、300-500mg、400-500mg、5-400mg、10-400mg、20-400mg、25-400mg、30-400mg、40-400mg、50-400mg、60-400mg、70-400mg、75-400mg、80-400mg、90-400mg、100-400mg、125-400mg、150-400mg、200-400mg、250-400mg、300-400mg、1-300mg、2-300mg、5-300mg、10-300mg、20-300mg、25-300mg、30-300mg、40-300mg、50-300mg、60-300mg、70-300mg、75-300mg、80-300mg、90-300mg、100-300mg、125-300mg、150-300mg、200-300mg、250-300mg、1-200mg、2-200mg、5-200mg、10-200mg、20-200mg、25-200mg、30-200mg、40-200mg、50-200mg、60-200mg、70-200mg、75-200mg、80-200mg、90-200mg、100-200mg、125-200mg、150-200mg、80-1000mg、80-800mg。
在一些实施方案中,该药物组合物包括但不限于1-1000mg、20-800mg、40-800mg、40-400mg、25-200mg、1mg、5mg、10mg、15mg、20mg、25mg、30mg、35mg、40mg、45mg、50mg、55mg、65mg、70mg、75mg、80mg、85mg、90mg、95mg、100mg、110mg、120mg、125mg、130mg、140mg、150mg、160mg、170mg、180mg、190mg、200mg、210mg、220mg、230mg、240mg、250mg、300mg、320mg、400mg、480mg、500mg、600mg、640mg、840mg的本发明化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶。
一种用于治疗哺乳动物的疾病的方法,所述方法包括给予受试者治疗有效量的本发明化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,治疗有效量优选1-1500mg,所述的疾病优选PI3Kα抑制剂相关疾病(如肿瘤)。
一种用于治疗哺乳动物的疾病的方法所述方法包括,将药物本发明化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶以1-1000mg/天的日剂量给予受试者,所述日剂量可以为单剂量或分剂量,在一些实施方案中,日剂量包括但不限于10-1500mg/天、10-1000mg/天、10-800mg/天、25-800mg/天、50-800mg/天、100-800mg/天、200-800mg/天、25-400mg/天、50-400mg/天、100-400mg/天、200-400mg/天,在一些实施方案中,日剂量包括但不限于10mg/天、20mg/天、25mg/天、50mg/天、80mg/天、100mg/天、125mg/天、150mg/天、160mg/天、200mg/天、300mg/天、320mg/天、400mg/天、480mg/天、600mg/天、640mg/天、800mg/天、1000mg/天。
本发明涉及一种试剂盒,该试剂盒可以包括单剂量或多剂量形式的组合物,该试剂盒包含本发明化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,本发明化合物的或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶的量与上述药物组合物中其量相同。
本发明涉及一种本发明上述的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶或者上述药物组合物在用于制备治疗与PI3Kα抑制剂相关疾病的药 物中的应用。
本发明涉及的本发明上述的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶、或者上述药物组合物的应用,所述的疾病选自肿瘤或癌症,优选自乳腺癌。
本发明化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶的量在每种情况下以游离碱的形式换算。
除非另有说明,在说明书和权利要求书中使用的术语具有下述含义。
本发明所述基团和化合物中所涉及的碳、氢、氧、硫、氮或F、Cl、Br、I均包括它们的同位素情况,及本发明所述基团和化合物中所涉及的碳、氢、氧、硫或氮任选进一步被一个或多个它们对应的同位素所替代,其中碳的同位素包括12C、13C和14C,氢的同位素包括氕(H)、氘(D,又叫重氢)、氚(T,又叫超重氢),氧的同位素包括16O、17O和18O,硫的同位素包括32S、33S、34S和36S,氮的同位素包括14N和15N,氟的同位素包括17F和19F,氯的同位素包括35Cl和37Cl,溴的同位素包括79Br和81Br。
“CN”是指氰基。
“卤素”是指F、Cl、Br或I。
“卤素取代的”是指F、Cl、Br或I取代,包括但不限于1至10个选自F、Cl、Br或I的取代基所取代,1至6个选自F、Cl、Br或I的取代基所取代,1至4个选自F、Cl、Br或I的取代基所取代。“卤素取代的”简称为“卤代”。
“烷基”是指取代的或者未取代的直链或支链饱和脂肪族烃基,包括但不限于1至20个碳原子的烷基、1至8个碳原子的烷基、1至6个碳原子的烷基、1至4个碳原子的烷基。非限制性实施例包括甲基、乙基、正丙基、异丙基、正丁基、仲丁基、新丁基、叔丁基、正戊基、异戊基、新戊基、正己基及其各种支链异构体;烷基可以是一价、二价、三价或四价。
“杂烷基”指取代的或者未取代的烷基中的1个或多个(包括但不限于2、3、4、5或6个)碳原子被杂原子(包括但不限于N、O或S)替换。非限制性实施例包括-X-(CH2)v-X-(CH2)v-X-(CH2)v-H(v为1至5的整数,X各自独立的选自键或杂原子,杂原子包括但不限于N、O或S,且至少有1个X选自杂原子,且杂原子中的N或S可被氧化成各种氧化态)。杂烷基可以是一价、二价、三价或四价。
“亚烷基”是指取代的或者未取代的直链和支链的二价饱和烃基,包括-(CH2)v-(v为1至10的整数),亚烷基实施例包括但不限于亚甲基、亚乙基、亚丙基和亚丁基等。
“亚杂烷基”是指取代的或者未取代的亚烷基中的1个或多个(包括但不限于2、3、4、5或6个)碳原子被杂原子(包括但不限于N、O或S)替换。非限制性实施例包括-X-(CH2)v-X-(CH2)v-X-(CH2)v-,v为1至5的整数,X各自独立的选自键、N、O或S,且至少有1个X选自N、O或S。
“环烷基”是指取代的或者未取代的饱和的碳环烃基,通常有3至12个碳原子,环烷基可以是单环、并环、桥环和螺环。非限制性实施例包括环丙基、环丁基、环戊基、环己基、环庚基、环丁基并环丁基、环丁基螺环丁基、金刚烷等。环烷基可以是一价、二价、三价或四价。
“杂环烷基”是指取代的或者未取代的饱和的含有杂原子的环烃基,包括但不限于3至12个原子、3至8个原子,包含1至3个选自N、O、S或Se的杂原子,杂环烷基的环上的C、N、S 可被氧化成各种氧化态。杂环烷基可以是单环、并环、桥环和螺环。杂环烷基可以连接在杂原子或者碳原子上,非限制性实施例包括环氧乙基、氮杂环丙基、氧杂环丁基、氮杂环丁基、四氢呋喃基、四氢-2H-吡喃基、二氧戊环基、二氧六环基、吡咯烷基、哌啶基、咪唑烷基、噁唑烷基、噁嗪烷基、吗啉基、六氢嘧啶基、哌嗪基、 杂环烷基可以是一价、二价、三价或四价。
“烯基”是指取代的或者未取代的直链和支链的不饱和烃基,其具有至少1个,通常有1、2或3个碳碳双键,主链包括但不限于2至10个、2至6个或2至4个碳原子,烯基实施例包括但不限于乙烯基、烯丙基、1-丙烯基、2-丙烯基、1-丁烯基、2-丁烯基、3-丁烯基、1-戊烯基、2-戊烯基、3-戊烯基、4-戊烯基、1-甲基-1-丁烯基、2-甲基-1-丁烯基、2-甲基-3-丁烯基、1-己烯基、2-己烯基、3-己烯基、4-己烯基、5-己烯基、1-甲基-1-戊烯基、2-甲基-1-戊烯基、1-庚烯基、2-庚烯基、3-庚烯基、4-庚烯基、1-辛烯基、3-辛烯基、1-壬烯基、3-壬烯基、1-癸烯基、4-癸烯基、1,3-丁二烯、1,3-戊二烯、1,4-戊二烯和1,4-己二烯等;烯基可以是一价、二价、三价或四价。
“炔基”是指取代的或者未取代的直链和支链的不饱和烃基,其具有至少1个,通常有1、2或3个碳碳三键,主链包括2至10个碳原子,包括但不限于在主链上有2至6个碳原子,主链上有2至4个碳原子,炔基实施例包括但不限于乙炔基、炔丙基、1-丙炔基、2-丙炔基、1-丁炔基、2-丁炔基、3-丁炔基、1-戊炔基、2-戊炔基、3-戊炔基、4-戊炔基、1-甲基-1-丁炔基、2-甲基-1-丁炔基、2-甲基-3-丁炔基、1-己炔基、2-己炔基、3-己炔基、4-己炔基、5-己炔基、1-甲基-1-戊炔基、2-甲基-1-戊炔基、1-庚炔基、2-庚炔基、3-庚炔基、4-庚炔基、1-辛炔基、3-辛炔基、1-壬炔基、3-壬炔基、1-癸炔基、4-癸炔基等;炔基可以是一价、二价、三价或四价。
“烷氧基”是指取代的或者未取代的-O-烷基。非限制性实施例包括甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、仲丁氧基、叔丁氧基、正戊氧基、正己氧基、环丙氧基和环丁氧基。
“碳环基”或“碳环”是指取代的或未取代的芳香环或者非芳香环,芳香环或者非芳香环可以是3至8元的单环、4至12元双环、10至15元三环、12至18元四元体系,碳环基可以连接在芳香环上或者非芳香环上,环任选为单环、并环、桥环或者螺环。非限制性实施例包括环丙烷、环丁烷、环戊烷、环己烷、环庚烷、1-环戊基-1-烯基、1-环戊基-2-烯基、1-环戊基-3-烯基、环己基、1-环己基-2-烯基、1-环己基-3-烯基、环己烯基、苯环、萘环、 “碳环基”或“碳环”可以是一价、二价、三价或四价。
“杂环基”或“杂环”是指取代的或未取代的芳香环或者非芳香环,芳香环或者非芳香环可以是3至8元的单环、4至12元双环或者10至15元三环、12至18元四元体系,且包含1个或多个(包括但不限于2、3、4或5个)个选自N、O、S或Se的杂原子,杂环基的环中选择性取代的C、N、S或Se可被氧化成各种氧化态。杂环基可以连接在杂原子或者碳原子上,杂环基可以连接在芳香环上或者非芳香环上,杂环基任选为单环、桥环、并环或者螺环,非限制性实施例包括环氧乙基、氮杂环丙基、氧杂环丁基、氮杂环丁基、1,3-二氧戊环基、1,4-二氧戊环基、1,3-二氧 六环基、氮杂环庚基、吡啶基、呋喃基、噻吩基、吡喃基、N-烷基吡咯基、嘧啶基、吡嗪基、哒嗪基、咪唑基、哌啶基、吗啉基、硫代吗啉基、1,3-二噻基、二氢呋喃基、二氢吡喃基、二噻戊环基、四氢呋喃基、四氢吡咯基、四氢咪唑基、四氢噻唑基、四氢吡喃基、苯并咪唑基、苯并吡啶基、吡咯并吡啶基、苯并二氢呋喃基、吡咯基、吡唑基、噻唑基、噁唑基、吡嗪基、吲唑基、苯并噻吩基、苯并呋喃基、苯并吡咯基、苯并咪唑基、苯并噻唑基、苯并噁唑基、苯并吡啶基、苯并嘧啶基、苯并吡嗪基、哌嗪基、氮杂二环[3.2.1]辛烷基、氮杂二环[5.2.0]壬烷基、氧杂三环[5.3.1.1]十二烷基、氮杂金刚烷基、氧杂螺[3.3]庚烷基、 “杂环基”或“杂环”可以是一价、二价、三价或四价。
“螺环”或“螺环基”是指取代的或未取代的单环之间共用一个原子(称螺原子)的多环基团,螺环体系中环原子的个数包括但不限于含有5至20个、6至14个、6至12个、6至10个,其中一个或多个环可以含有0个或多个(包括但不限于1、2、3或4)双键,且任选可以含有0至5个选自N、O或S(=O)n(n为0、1或2)的杂原子。

。“螺环”或“螺环基”可以是一价、二价、三价或四价。
“并环”或“并环基”是指系统中的每个环与体系中的其他环共享毗邻的一对原子的多环基团,其中一个或多个环可以含有0个或多个(包括但不限于1、2、3或4)双键,且可以是取代的或未取代,并环体系中的各个环可以含0至5个杂原子或含有杂原子的基团(包括但不限于选自N、S(=O)n或O,n为0、1或2)。并环体系中环原子的个数包括但不限于5至20个,5至14个,5至12个,5至10个。非限定性实例包括: “并环”或“并环基”可以是一价、二价、三价或四价。
“桥环”或“桥环基”是指取代的或未取代的含有任意两个不直接连接的原子的多环基团,可以 含有0个或多个双键,桥环体系中的任意环可以含0至5个选自杂原子或含有杂原子的基团(包括但不限于N、S(=O)n或O,其中n为0、1、2)。环原子个数包括但不限于5至20个、5至14个、5至12个或5至10个。非限定性实例包括立方烷、金刚烷。“桥环”或“桥环基”可以是一价、二价、三价或四价。
“碳螺环”、“螺环碳环基”、“螺碳环基”或者“碳螺环基”是指环体系仅有碳原子组成的“螺环”。
“碳并环”、“并环碳环基”、“并碳环基”或者“碳并环基”是指环体系仅有碳原子组成的“并环”。
“碳桥环”、“桥环碳环基”、“桥碳环基”或者“碳桥环基”是指环体系仅有碳原子组成的“桥环”。
“杂单环”、“单环杂环基”或“杂单环基”是指单环体系的“杂环基”或“杂环”,
“杂并环”、“杂并环基”“并环杂环基”或“并杂环基”是指含有杂原子的“并环”。
“杂螺环”、“杂螺环基”、“螺环杂环基”或“螺杂环基”是指含有杂原子的“螺环”。
“杂桥环”、“杂桥环基”、“桥环杂环基”或“桥杂环基”是指含有杂原子的“桥环”。
“芳基”或“芳环”是指取代的或者未取代的具有单环或稠合环的芳香族烃基,芳香环中环原子个数包括但不限于6至18、6至12或6至10个碳原子。芳基环可以稠合于饱和或不饱和的碳环,其中与母体结构连接在一起的环为芳基环,非限制性实施例包含苯环、萘环、“芳基”或“芳环”可以是一价、二价、三价或四价。当为二价、三价或四价时,连接位点位于芳基环上。
“杂芳基”或“杂芳环”是指取代或未取代的芳香族烃基,且含有1至5个选杂原子或含有杂原子的基团(包括但不限于N、O、S(=O)n或Se(=O)n,n为0、1、2),杂芳香环中环原子个数包括但不限于5至15、5至10或5至6个。环上的原子C、N、S任选被氧化(即C(=O)、NO、S(=O)n、Se(=O)n,n为1、2),杂芳基的非限制性实施例包括但不限于吡啶基、呋喃基、噻吩基、硒吩基、吡啶基、吡喃基、N-烷基吡咯基、嘧啶基、吡嗪基、哒嗪基、咪唑基、苯并吡唑基、苯并咪唑基、苯并吡啶基、吡咯并吡啶基、吡啶酮基等。所述杂芳基环可以稠合于饱和或不饱和的碳环或杂环上,其中与母体结构连接在一起的环为芳基环,非限制性实施例包含 本文中出现的杂芳基,其定义与本定义一致。杂芳基可以是一价、二价、三价或四价。当为二价、三价或四价时,连接位点位于具有芳香性的环上。
“取代”或“取代的”是指被1个或多个(包括但不限于2、3、4或5个)取代基所取代,取代基包括但不限于H、F、Cl、Br、I、烷基、环烷基、烷氧基、卤代烷基、硫醇、羟基、硝基、巯基、氨基、氰基、异氰基、芳基、杂芳基、杂环基、桥环基、螺环基、并环基、羟基烷基、=O、羰基、 醛、羧酸、甲酸酯、-(CH2)m-C(=O)-Ra、-O-(CH2)m-C(=O)-Ra、-(CH2)m-C(=O)-NRbRc、-(CH2)mS(=O)nRa、-(CH2)m-烯基-Ra、ORd或-(CH2)m-炔基-Ra(其中m、n为0、1或2)、芳基硫基、硫代羰基、硅烷基或-NRbRc等基团,其中Rb与Rc独立选自包括H、羟基、氨基、羰基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、磺酰基、三氟甲磺酰基,作为选择,Rb与Rc可形成五或六元环烷基或杂环基、Ra与Rd各自独立选自芳基、杂芳基、烷基、烷氧基、环烷基、杂环基、羰基、酯基、桥环基、螺环基或并环基。
“1至X个选自…..取代基所取代”是指被1、2、3….X个选自…..取代基所取代,X选自1至10之间的任意整数。如“1至4个Rk取代”是指被1、2、3或4个Rk取代。如“1至5个选自…..取代基所取代”是指被1、2、3、4或5个选自…..取代基所取代。如“杂桥环任选被1至4个选自H或F的取代基所取代”是指杂桥环任选被1、2、3或4个选自H或F的取代基所取代。
X-Y元的环(X、Y为整数,且3≤X<Y,X<Y≤20选自4至20之间的任意整数)包括了X、X+1、X+2、X+3、X+4….Y元的环。环包括了杂环、碳环、芳环、芳基、杂芳基、环烷基、杂单环、杂并环、杂螺环或杂桥环。如“4-7元杂单环”是指4元、5元、6元或7元的杂单环,“5-10元杂并环”是指5元、6元、7元、8元、9元或10元的杂并环。
Cx-y碳环(包括芳基、环烷基、单环碳环、螺环碳环、并环碳环或桥环碳环)包括了Cx、Cx+1、Cx+2、Cx+3、Cx+4….Cy元的环(x为整数,且3≤x<y,y选自4至20之间的任意整数),例如。如C3-6环烷基”是指C3、C4、C5或C6环烷基;
当某一个基团具有一个或多个可连接位点时,该基团的任意一个或多个位点可以通过化学键与其他基团连接。当该化学键的连接方式是不定位的,且可连接位点存在氢原子时,则连接化学键时,该位点的H原子的个数会随所连接化学键的个数而对应减少变成相应价数的基团。例如表示该哌啶基上的任意可连接位点可以通过1个化学键与其他基团相连,至少包括这4种连接方式,即使-N-上画出了H原子,也包括了例如表示该哌啶基上的R基团可以位于C上,可以位于N上,至少包括了
当所列举的连接基团没有指明其连接方向时,其连接方向包括了从左向右和从右向左的读取顺序的方向进行连接,例如A-L-B,L选自-M-W-时,包括了A-M-W-B和A-W-M-B。
“任选”或“任选地”是指随后所描述的事件或环境可以但不必须发生,该说明包括该事件或环境发生或不发生的场合。如:“任选被F取代的烷基”指烷基可以但不必须被F取代,说明包括烷基被F取代的情形和烷基不被F取代的情形。
“药学上可接受的盐”或者“其药学上可接受的盐”是指本发明化合物保持游离酸或者游离碱的生物有效性和特性,且所述的游离酸通过与无毒的无机碱或者有机碱,所述的游离碱通过与无毒的无机酸或者有机酸反应获得的盐。
“药物组合物”是指一种或多种本发明所述化合物、或者其立体异构体、互变异构体、氘代物、 溶剂化物、前药、代谢产物、药学上可接受的盐或共晶和其它化学组分形成的混合物,其中,“其它化学组分”是指药学上可接受的载体、赋形剂和/或一种或多种其它治疗剂。
“制剂规格”是指每一支、片或其他每一个单位制剂中含有主药的重量。
“载体”是指不会对生物体产生明显刺激且不会消除所给予化合物的生物活性和特性的材料。
“前药”是指可经体内代谢转化为具有生物活性的本发明化合物。本发明的前药通过修饰本发明化合物中的氨基或者羧基来制备,该修饰可以通过常规的操作或者在体内被除去,而得到母体化合物。当本发明的前药被施予哺乳动物个体时,前药被割裂形成游离的氨基或者羧基。
“共晶”是指活性药物成分(API)和共晶形成物(CCF)在氢键或其他非共价键的作用下结合而成的晶体,其中API和CCF的纯态在室温下均为固体,并且各组分间存在固定的化学计量比。共晶是一种多组分晶体,既包含两种中性固体之间形成的二元共晶,也包含中性固体与盐或溶剂化物形成的多元共晶。
“动物”是指包括哺乳动物,例如人、陪伴动物、动物园动物和家畜,优选人、马或者犬。
“立体异构体”是指由分子中原子在空间上排列方式不同所产生的异构体,包括顺反异构体、对映异构体、非对映异构体和构象异构体。
“互变异构体”是指分子中某一原子在两个位置迅速移动而产生的官能团异构体,如酮式-烯醇式异构和酰胺-亚胺醇式异构等。
“IC50”是对指定的生物过程(或该过程中的某个组分比如酶、受体、细胞等)抑制一半时所需的药物或者抑制剂的浓度。
具体实施方式
以下实施例详细说明本发明的技术方案,但本发明的保护范围包括但是不限于此。
化合物的结构是通过核磁共振(NMR)或(和)质谱(MS)来确定的。NMR位移(δ)以10-6(ppm)的单位给出。NMR的测定是用(Bruker Avance III 400和Bruker Avance 300)核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6),氘代氯仿(CDCl3),氘代甲醇(CD3OD),内标为四甲基硅烷(TMS);
MS的测定用(Agilent 6120B(ESI)和Agilent 6120B(APCI));
HPLC的测定使用Agilent 1260DAD高压液相色谱仪(Zorbax SB-C18 100×4.6mm,3.5μM);
薄层层析硅胶板使用烟台黄海HSGF254或青岛GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm-0.20mm,薄层层析分离纯化产品采用的规格是0.4mm-0.5mm;
柱层析一般使用烟台黄海硅胶200-300目硅胶为载体;
为了完成本发明的目的,根据本领域技术人员已知的有机合成技术,从市售的化学品和/或化学文献中描述的化合物开始,制备本文所述反应中使用的化合物“商业上可用的化学品”是从标准的商业来源获得的,包括上海阿拉丁生化科技股份有限公司,上海麦克林生化科技有限公司,Sigma-Aldrich,阿法埃莎(中国)化学有限公司,梯希爱(上海)化成工业发展有限公司,安耐吉化学,上海泰坦科技股份有限公司,科龙化工,百灵威科技有限公司等。
合成实验中,无特殊说明,温度为室温;
THF:四氢呋喃;DMF:N,N-二甲基甲酰胺;DIPEA:N,N-二异丙基乙胺;HATU:CAS  148893-10-1;Boc:叔丁氧基羰基;Ts:对甲苯磺酰基;Cbz:苄氧羰基;TMS:三甲基硅基。
实施例1:化合物1的制备
第一步:1C的制备
将[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(44mg,0.05mmol)加入到1A(370mg,1.07mmol)(参考专利WO2022235574得到)、1B(376mg,1.18mmol)、碳酸钾(369mg,2.68mmol)的二氧六环(10mL)和水(1mL)溶液中。氮气氛围下,100℃反应16h。冷却至室温,加入水(20mL),乙酸乙酯(50mL x 3)萃取,合并有机相。有机相经无水硫酸钠干燥、过滤,滤液经减压浓缩得残留物,残留物经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=5:2)得1C(500mg,产率87%)。
LCMS m/z=542.4[M+1]+
第二步:1D的制备
将钯碳(70mg,0.65mmol,10%)加入到1C(350mg,1.01mmol)的乙醇(150mL)溶液中。氢气氛围下,室温反应8h。反应完毕后,过滤,滤液经减压浓缩得残留物,残留物经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=5:1)得1D(210mg,产率60%)。
LCMS m/z=544.5[M+1]+
第三步:1F的制备
将1D(210mg,0.39mmol)溶于4N盐酸二氧六环(30mL)溶液中,室温反应2h。将反应液减压浓缩得1F的盐酸盐(185mg,粗品)。
LCMS m/z=444.5[M+1]+
第四步:化合物1的制备
将氢氧化钠(125mg,3.12mmol)加入到1F的盐酸盐(185mg,粗品)的甲醇(10mL)和水(1.5mL)溶液中。氮气氛围下,50℃反应16h。冷却至室温,用2N盐酸调节pH值至3-4,混合物减压浓缩得残留物,残留物经柱层析分离纯化(二氯甲烷:四氢呋喃(v:v)=10:1)得化合物1(40mg,两步产率24%)。
LCMS m/z=430.2[M+1]+
1H NMR(400MHz,DMSO-d6)δ12.55(s,1H),7.80(s,1H),7.66-7.56(m,4H),7.50-7.36(m,2H),7.14(d,1H),6.90(s,1H),6.54(t,1H),5.52(t,1H),3.61-3.47(m,1H),3.03-2.90(m,1H),2.41(s, 3H),2.08(s,3H).
实施例2:化合物2的的制备
第一步:2B的制备
向1C(50mg,0.092mmol)的二氯甲烷(2mL)溶液中加入三氟乙酸(4mL),室温反应3h。将反应液减压浓缩得残留物,向残留物中加入水(10mL),乙酸乙酯(10mL x 2)萃取,合并有机相。有机相用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得2B(42mg,粗品)。
LCMS m/z=442.4[M+H]+
第二步:化合物2的的制备
将1N氢氧化水溶液(1.0M,5mmol,5mL)加入到3b(42mg,粗品)的甲醇(5mL)和四氢呋喃(5mL)溶液中,50℃反应16h。减压浓缩得残留物,冰水浴下用1N盐酸水溶液调pH至6-7,乙酸乙酯(20mL x 2)萃取,合并有机相。有机相经无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经Pre-HPLC纯化(仪器及制备柱:采用Waters 2767制备液相,制备柱型号是XBridge@Prep C18,内径x长度=19mm x250mm)。制备方法:粗品用DMF溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:乙腈/水(含0.05%的三氟乙酸)。梯度洗脱方法:乙腈由30%梯度洗脱至80%(流速:12mL/min;洗脱时间15min)得化合物2(10mg,两步产率25%)。
LCMS m/z=428.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.10(s,1H),11.24(s,1H),8.02-7.97(m,1H),7.94-7.90(m,1H),7.85(d,1H),7.81-7.75(m,2H),7.74-7.68(m,1H),7.62-7.54(m,1H),7.43-7.36(m,1H),7.15(t,1H),7.00(d,1H),2.52(s,3H),2.10(s,3H).
实施例3:化合物3的制备
第一步:3B-1和3B-2的制备
将醋酸钯(21mg,0.09mmol)加入到1A(320mg,0.92mmol)、1-叔丁氧羰基-2,3-二氢吡咯(171mg,1.01mmol)、三苯基膦(48.26mg,0.18mmol)、醋酸钾(271mg,2.76mmol)的DMF(5mL)溶液中,氮气氛围下,100℃反应16h。冷却至室温,将反应液减压浓缩后得粗品,粗品经柱层析分 离纯化(石油醚:乙酸乙酯(v:v)=5:2)得3B-1和3B-2的混合物(400mg,产率100%)。
LCMS m/z=436.0[M+H]+
第二步:3C的制备
氮气保护下,将二氧化铂(52mg,0.23mmol)加入到3B-1与3B-2的混合物(400mg,0.92mmol)的乙醇(10mL)溶液中。氢气氛围下,25℃反应16h。过滤,滤液经减压浓缩得3C(380mg,产率95%)。
LCMS m/z=438.1[M+H]+
第三步:3D的制备
将三氟乙酸(2mL)加入到3C(380mg,0.87mmol)的二氯甲烷(5mL)溶液中,氮气氛围下室温反应3h。将反应液减压浓缩得3D的三氟乙酸盐(400mg,粗品)。
LCMS m/z=338.3[M+H]+
第四步:3E的制备
将2-甲基苯甲酸甲酯(136mg,0.88mmol)、DIPEA(228mg,1.76mmol)、DMAP(6mg,0.05mmol)加入到3D的三氟乙酸盐(200mg,粗品)的乙醇(10mL)溶液中,120℃微波反应7h。将反应液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=3:1)得到3E(95mg,2步产率46%)。
LCMS m/z=472.2[M+H]+
第五步:化合物3的制备
将氢氧化钠(64mg,1.60mmol)加入到3E(0.19g,0.40mmol)的甲醇(5mL)、四氢呋喃(5mL)和水(5mL)溶液中,50℃反应16h。冰水浴下,用1N盐酸水溶液调pH至4-5。减压浓缩得残留物,残留物经制备板分离纯化(100%乙酸乙酯)得化合物3。
LCMS m/z=458.5[M+H]+
实施例4:化合物4的制备
第一步:4b的制备
将4a(7.20g,33.47mmol)溶于二氯甲烷(100mL)中,加入吡啶(3.97g,50.20mmol)和丙酰氯(3.41g,36.82mmol),室温搅拌16h。加入盐酸(2N)调节pH=4-5,二氯甲烷(100mL x 2)萃取。 合并有机相,有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=95:5)得4b(7.00g,产率77%)。
LCMS m/z=269.0[M+1]+
第二步:4c的制备
将4b(3.50g,13.00mmol)溶于四氯化钛(13mL,2N in DCM)中,100℃反应16h。加入水(40mL)和乙酸乙酯(40mL),分离有机相。有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。粗品经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=94:6)得4c(1.80g,产率51%)。
LCMS m/z=269.0[M+1]+
第三步:4d的制备
将3-氟二环[1.1.1]戊烷-1-羧酸(1.74g,13.38mmol)溶于二氯甲烷(60mL)中,加入N,N'-二环己基碳二亚胺(3.04g,14.72mmol)、4-二甲氨基吡啶(163mg,1.34mmol),室温反应30min。加入4c(3.60g,13.38mmol),室温反应16h。加入水(100mL),分离有机相。有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=96:4)得4d(1.80g,产率35%)。
1H NMR(400MHz,DMSO-d6)δ7.68(s,1H),2.89(t,2H),2.82(t,2H),2.71(q,2H),2.53(d,6H),2.10-1.98(m,2H),1.03(t,3H).
第四步:4e的制备
将氢化钠(564mg,14.10mmol,60%)溶于二甲亚砜(20mL)中,缓慢加入4d(1.80g,4.70mmol),28℃反应1h。加入水(40mL)和醋酸(0.2mL),乙酸乙酯(40mL)萃取。有机相用饱和食盐水(40mL x 3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。将粗品加入到醋酸(20mL)中,缓慢滴入浓盐酸(12N,1mL),95℃反应2h。冷却至室温,减压浓缩,向残留物中加入水(40mL),乙酸乙酯(30mL x 2)萃取。合并有机相,用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=90:10)得4e(1.70g,产率99%)。
LCMS m/z=363.1[M+1]+
第五步:4f的制备
将4e(300mg,0.83mmol)、双三苯基磷二氯化钯(58mg,0.083mmol)和三丁基(1-乙氧基乙烯)锡(380mg,1.06mmol)加入到干燥的1,4-二氧六环(6mL)中,氮气保护下95℃反应16h。将反应液冷却至50℃,加入盐酸水溶液(3mL,1N),搅拌30min。冷却至室温,加入饱和氟化钾水溶液(8mL),搅拌30min。过滤,滤液中加入水(30mL),乙酸乙酯(40mL)萃取。有机相经无水硫酸钠干燥后,过滤,滤液减压浓缩得残留物,残留物经柱层析分离纯化(石油醚:四氢呋喃(v:v)=9:1)得4f(240mg,产率89%)。
LCMS m/z=327.4[M+H]+
第六步:4g的制备
将4f(200mg,0.61mmol)、(R)-(+)-叔丁基亚磺酰胺(220mg,1.83mmol),钛酸四异丙酯(690mg,2.44mmol)加入到干燥的四氢呋喃(5mL)中,氮气保护下85℃反应16h。加入水(20mL),乙酸乙酯(20mL)萃取。有机相经无水硫酸钠干燥后,过滤,滤液减压浓缩得残留物,残留物经柱层 析分离纯化(石油醚:四氢呋喃(v:v)=14:86)得4g(130mg,产率49%)。
LCMS m/z=430.2[M+H]+
第七步:4h的制备
将硼氢化钠(18mg,0.46mmol)和七水氯化铈(III)(56mg,0.15mmol)加入到4g(130mg,0.30mmol)的甲醇(5mL)溶液中,室温反应1h。加入水(1mL),减压浓缩。加入水(10mL),二氯甲烷(20mL x 2)萃取。合并有机相,有机相经无水硫酸钠干燥,过滤,滤液减压浓缩得到4h(130mg,产率100%)。
LCMS m/z=432.5[M+H]+
第八步:4i的盐酸盐制备
将4h(130mg,0.30mmol)溶于4N盐酸二氧六环(5mL)中,室温反应2h。减压浓缩得4i的盐酸盐(109mg,粗品)。
LCMS m/z=328.4[M+1]+
第九步:4g的制备
将6-氯-3-氟吡啶-2-甲酸甲酯(68mg,0.36mmol)和N,N-二异丙基乙胺(155mg,1.20mmol)加到4i的盐酸盐(109mg,粗品)的二甲亚砜(5mL)中,100℃反应16h。冷却至室温,加入水(20mL),乙酸乙酯(30mL x 2)萃取。合并有机相,有机相用饱和食盐水(40mL x 3)洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得残留物,残留物经柱层析分离纯化(石油醚:四氢呋喃(v:v)=14:86)得4g(100mg,两步产率67%)。
第十步:化合物4的的制备
将2N氢氧化水溶液(2.0M,4mmol,2mL)加入到4g(100mg,0.20mmol)的甲醇(2mL)溶液中,50℃反应2h。减压浓缩得残留物。冰水浴下用1N盐酸水溶液将残留物调pH至6-7,乙酸乙酯(10mL x 2)萃取。合并有机相,有机相经无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经制备液相(仪器:waters 2767制备液相;色谱柱:SunFire@Prep C18(19mm×150mm);流动相组成:流动相A:乙腈流动相B:水(含0.05%的乙酸铵))分离纯化得化合物4(10mg,产率10%)。
LCMS m/z=483.6[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.01(s,1H),8.44(s,1H),7.53(s,1H),7.33(d,1H),7.02(d,1H),5.15-4.99(m,1H),3.37-3.30(m,2H),2.89-2.76(m,2H),2.66(d,6H),2.08-1.95(m,5H),1.60(d,3H).
实施例5:化合物5的制备
第一步:5b的制备
将吡啶(4.36g,55.06mmol),丙酰氯(3.74g,40.38mmol)加入到5a(5.00g,37.26mmol)的二氯甲烷(100mL)溶液中,室温搅拌16h。加入水(100mL),分液,水相用二氯甲烷(50mL)萃取1次,合并有机相。有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=96:4)得5b(7.00g,产率99%)。
LCMS m/z=191.1[M+H]+
第二步:5c的制备
将三氯化铝(14.72g,110.40mmol)加入到5b(7.00g,36.80mmol)的二硫化碳(14mL)溶液中,80℃反应3.5h。冷却至室温,加入稀盐酸(200mL,1N)和乙酸乙酯(100mL),搅拌至溶清。分层,有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=98:2)得5c(6.27g,产率90%)。
1H NMR(400MHz,DMSO)δ12.21(s,1H),7.75(s,1H),6.82(s,1H),3.13-3.03(m,2H),2.87-2.78(m,4H),2.06-1.97(m,2H),1.10(t,3H).
第三步:5d的制备
将N-溴代丁二酰亚胺(4.68g,26.28mmol)加入到5c(5.00g,26.28mmol)的N,N-二甲基甲酰胺(50mL)溶液中,室温反应16h。将反应液倒入到水(500mL)中,乙酸乙酯(100mL x 3)萃取。合并有机相,有机相用饱和食盐水(50mL x 3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。粗品用混合溶剂(石油醚:乙酸乙酯(v:v)=10:1)打浆得5d(4.44g,产率63%)。
LCMS m/z=269.1[M+H]+
第四步:5e的制备
将3-氟二环[1.1.1]戊烷-1-羧酸(967mg,7.43mmol)溶于二氯甲烷(60mL)中,加入N,N'-二环己基碳二亚胺(1.69g,8.17mmol)和4-二甲氨基吡啶(91mg,0.74mmol),室温搅拌10min。加入5d(2.00g,7.43mmol),35℃搅拌16h。加入硅胶,减压浓缩得残留物,残留物经柱层析分离纯化(石油醚:二氯甲烷(v:v)=40:60)得5e(2.20g,产率78%)。
LCMS m/z=381.0[M+H]+
第五步:5f的制备
将氢化钠(625mg,15.63mmol,60%)分批加入到5e(2.00g,5.25mmol)的二甲亚砜(50mL)中, 28℃反应1h。将反应液缓慢滴加到0℃的饱和氯化铵(300mL)溶液中,乙酸乙酯(100mL)萃取。水相用1N盐酸调pH=4-5左右,乙酸乙酯(100mL)萃取。合并有机相,有机相用饱和食盐水(40mL x 3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。将粗品加入到醋酸(50mL)中,加入浓盐酸(12N,1mL),95℃反应2h。减压浓缩得粗品,粗品用乙酸乙酯(100mL)溶解,加入硅胶,减压浓缩得残留物,残留物经柱层析分离纯化(石油醚:四氢呋喃(v:v)=80:20)得5f(1.77g,产率93%)。
LCMS m/z=363.3[M+H]+
第六步:5g的制备
将5f(300mg,0.83mmol)、双三苯基磷二氯化钯(58mg,0.08mmol)和三丁基(1-乙氧基乙烯)锡(0.38g,1.06mmol)加入到干燥的1,4-二氧六环(6mL)中,氮气氛围下100℃反应16h。将反应液冷却至50℃,加入盐酸水溶液(3mL,1N),搅拌1h。冷却至室温,加入饱和氟化钾水溶液(8mL),搅拌30min。过滤,滤液中加入水(30mL),乙酸乙酯(40mL)萃取。有机相经无水硫酸钠干燥后过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=90:10)得5g(165mg,产率61%)。
第七步:5h的制备
-10℃下,将硼氢化钠(17mg,0.45mmol)分批加入到5g(115mg,0.35mmol)的二氯甲烷(5mL)和甲醇(5mL)溶液中,-10℃下反应2h。加入水(1mL),减压浓缩得残留物。加入水(30mL),乙酸乙酯(30mL x 2)萃取,合并有机相。有机相经无水硫酸钠干燥,过滤,滤液减压浓缩得到5h(110mg,产率95%)。
LCMS m/z=329.3[M+H]+
第八步:5i的制备
将三溴化硼(270mg,1.0mmol)滴加到5h(110mg,0.33mmol)的二氯甲烷(10mL)溶液中,室温反应3h。加入饱和碳酸氢钠水溶液(30mL),二氯甲烷(30mL x 2)萃取。合并有机相,有机相经无水硫酸钠干燥,过滤,滤液减压浓缩得5i(130mg,粗品)。
LCMS m/z=391.0[M+H]+
第九步:5j的制备
将2-氨基苯甲酸甲酯(100mg,0.66mmol)滴加到5i(130mg,粗品)的N,N-二甲基甲酰胺(5mL)中,80℃反应16h。冷却至室温,加入水(20mL),乙酸乙酯(30mL x 2)萃取,合并有机相。有机相用饱和食盐水(40mL x 3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=90:10)得5j(80mg,两步产率52%)。
LCMS m/z=462.7[M+H]+
第十步:化合物5的的制备
将2N氢氧化水溶液(2.0M,3mmol,1.5mL)加入到5j(80mg,0.17mmol)的甲醇(10mL)和四氢呋喃(3mL)的混合溶液中,50℃反应6h。减压浓缩得残留物,0℃下用1N盐酸水溶液调pH至6-7,乙酸乙酯(30mL x 2)萃取,合并有机相。有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经Pre-HPLC纯化(仪器及制备柱:采用Waters 2767制备液相,制备柱型号是XBridge@Prep C18,内径*长度=19mm*250mm)。制备方法:粗品用DMF溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:乙腈/水(含0.05%的乙酸铵)。梯度洗脱方法: 乙腈由50%梯度洗脱80%(流速:15mL/min;洗脱时间15min)得化合物5(15mg,产率19%)。
LCMS m/z=448.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.73(s,1H),8.54(s,1H),7.78(dd,1H),7.73(s,1H),7.22(t,1H),6.50(t,1H),6.42(d,1H),5.21-5.09(m,1H),3.26-3.15(m,1H),3.04-2.80(m,3H),2.75-2.65(m,6H),2.11-1.97(m,5H),1.64(d,3H).
实施例6:化合物6-A与化合物6-B的制备
化合物5经SFC on AS column纯化(仪器及制备柱:采用Waters 150 Prep-SFC F,制备柱型号是:Chiralcel AS column。制备方法:化合物5用乙腈溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:A for CO2and B for EtOH(0.1%NH3·H2O)。梯度洗脱方法:25%流动相B等梯度洗脱(流速:120mL/min;洗脱时间3.5min)),冻干后得化合物6-A(化合物6-A为化合物6-1与化合物6-2结构之一)和化合物6-B(化合物6-B为化合物6-1与化合物6-2结构之一)。
分析方法(仪器及制备柱:高效液相色谱仪-正相色谱,制备柱型号是:AS-H(4.6*2.50mm,5um)流动相体系:正己烷:乙醇(95:5),柱温:35℃,流速:1.0mL/min。保留时间T=15.786min为化合物6-A(化合物6-A为化合物6-1与化合物6-2结构之一)。保留时间T=25.718min为化合物6-B(化合物6-B为化合物6-1与化合物6-2结构之一)。
化合物6-A:
LCMS m/z=448.6[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.69(s,1H),8.46(d,1H),7.78(dd,1H),7.73(s,1H),7.24(t,1H),6.51(t,1H),6.44(d,1H),5.20-5.10(m,1H),3.25-3.16(m,1H),3.04-2.80(m,3H),2.74-2.66(m,6H),2.10-1.98(m,5H),1.64(d,3H).
化合物6-B:
LCMS m/z=448.6[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.67(s,1H),8.46(d,1H),7.78(dd,1H),7.73(s,1H),7.24(t,1H),6.51(t,1H),6.44(d,1H),5.20-5.10(m,1H),3.25-3.16(m,1H),3.02-2.82(m,3H),2.74-2.66(m,6H),2.09-1.97(m,5H),1.65(d,3H).
实施例7:化合物7的制备
化合物7以5d、2-茚羧酸为起始物料,参考实施例5的合成方法得到。
LCMS m/z=480.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.60(s,1H),8.19(d,1H),7.74(dd,1H),7.70(s,1H),7.30(d,1H),7.21(d,1H),7.17-7.10(m,2H),7.07(t,1H),6.47(t,1H),6.05(d,1H),4.68-4.55(m,1H),4.19-4.09(m,1H),3.53-3.38(m,2H),3.29-3.24(m,2H),3.13-3.03(m,1H),2.87-2.66(m,3H),2.09(s,3H),1.97-1.88(m,2H),1.27(d,3H).
实施例8:化合物8的制备
第一步:8a的制备
0℃下,将双(三甲基硅基)氨基钠(2.0M,14mL,28mmol,)缓慢滴加到5d(2.5g,9.29mmol)的四氢呋喃(60mL)溶液中,室温反应1h。0℃下缓慢滴加二硫化碳(2.12g,27.87mmol),30℃反应16h。0℃下加入15%稀硫酸(32mL),室温搅拌0.5h。加入水20mL,乙酸乙酯(40mL x 2)萃取,合并有机相。有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=60:40)得8a(2.25g,产率78%)。
LCMS m/z=311.1[M+H]+
第二步:8b的制备
将碳酸钾(0.91g,6.55mmol)、碘乙烷(3.43g,21.84mmol)加入到8a(1.7g,5.46mmol)的丙酮(30mL)溶液中,60℃反应3h。加入水(50mL),二氯甲烷(40mL x 2)萃取,合并有机相。有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=90:10)得8b(1.49g,产率80%)。
LCMS m/z=339.2[M+H]+
第三步:8c的制备
将8b(1.5g,4.42mmol)、双三苯基磷二氯化钯(0.31g,0.44mmol)和三丁基(1-乙氧基乙烯)锡(2.23g,6.19mmol)加入到干燥的1,4-二氧六环(30mL)中,氮气氛围下100℃反应16h。降温至50℃,加入盐酸水溶液(4.5mL,1N),搅拌1h。冷却至室温,加入饱和氟化钾水溶液(12mL),搅拌30min。过滤,滤液中加入水(30mL),乙酸乙酯(40mL)萃取。有机相经无水硫酸钠干燥后过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=93:7)得8c(800mg,产率60%)。
LCMS m/z=303.2[M+H]+
第四步:8d的制备
-10℃下,将硼氢化钠(130mg,3.44mmol)分批加入到8c(800mg,2.65mmol)的二氯甲烷(10mL)和甲醇(10mL)溶液中,室温反应2h。0℃下加入饱和氯化铵水溶液(1mL),加入水(30mL),二氯甲烷(40mL x 2)萃取,合并有机相。有机相经无水硫酸钠干燥,过滤,滤液减压浓缩得到8d(800mg,粗品)。
LCMS m/z=305.1[M+H]+
第五步:8e的制备
0℃下,将三溴化硼(2.14g,7.91mmol)滴加到8d(800mg,粗品)的二氯甲烷(25mL)溶液中,室温反应3h。在0℃下加入饱和碳酸氢钠水溶液(30mL),二氯甲烷(30mL x 2)萃取。合并有机相,有机相经无水硫酸钠干燥,过滤,滤液减压浓缩得8e(475mg,粗品)。
LCMS m/z=367.1[M+H]+
第六步:8f的制备
将2-氨基苯甲酸甲酯(390mg,2.58mmol)滴加到8e(475mg,粗品)的N,N-二甲基甲酰胺(10mL)中,80℃反应16h。冷却至室温,加入水(20mL),乙酸乙酯(30mL x 2)萃取,合并有机相。有机相用饱和食盐水(40mL x 3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=92:8)得8f(334mg,三步产率29%)。
LCMS m/z=438.2[M+H]+
第七步:8g的制备
0℃下,将间氯过氧苯甲酸(174mg,1.01mmol)加入到8f(334mg,0.76mmol)的二氯甲烷(15mL)溶液中,室温反应2h。加入硫代硫酸钠水溶液(30mL),搅拌30min,二氯甲烷(20mL x 2)萃取,合并有机相。有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=55:45)得8g(278mg,产率80%)。
LCMS m/z=454.5[M+H]+
第八步:8h的制备
将N,N-二异丙基乙胺(710mg,5.5mmol)滴加到8g(250mg,0.55mmol)和5-氟异吲哚啉盐酸盐 (430mg,2.48mmol)的二甲亚砜(10mL)中,100℃封管反应48h。冷却至室温,加入水(20mL),乙酸乙酯(20mL x 2)萃取,合并有机相。有机相用饱和食盐水(20mL x 3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=60:40)得8h(125mg,产率44%)。
LCMS m/z=513.2[M+H]+
第九步:化合物8的制备
将2N氢氧化水溶液(2.0M,9mmol,4.5mL)加入到8h(80mg,0.17mmol)的甲醇(8mL)和四氢呋喃(5mL)的混合溶液中,50℃反应6h。0℃下用1N盐酸水溶液调pH至6-7,乙酸乙酯(30mL x 2)萃取,合并有机相。有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经Pre-HPLC纯化(仪器及制备柱:采用Waters 2767制备液相,制备柱型号是XBridge@Prep C18,内径*长度=19mm*250mm)。制备方法:粗品用DMF溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:乙腈/水(含0.05%的三氟乙酸)。梯度洗脱方法:甲醇由50%梯度洗脱80%(流速:15mL/min;洗脱时间15min)分离纯化得化合物8(15mg,产率19%)。
LCMS m/z=499.6[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.63(s,1H),8.48(s,1H),7.80(dd,1H),7.69(s,1H),7.51-7.44(m,1H),7.31(dd,1H),7.24-7.12(m,2H),6.50(t,1H),6.40(d,1H),5.34-5.13(m,5H),3.23-3.13(m,1H),2.93-2.73(m,3H),2.25(s,3H),2.05-1.94(m,2H),1.68(d,3H).
实施例9:化合物9-A与化合物9-B的制备
化合物9以5d、间氟苯甲醛为起始原料,参照专利WO2022235574A1得到。化合物9经SFC on AD column纯化(仪器及制备柱:采用Waters 150 Prep-SFC F,制备柱型号是:Chiralcel AD column。制备方法:化合物9用乙腈溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:A for CO2and B for IPA(0.1%NH3·H2O)。梯度洗脱方法:25%流动相B等梯度洗脱(流速:120mL/min;洗脱时间3.2min)),冻干后得到化合物9-A(化合物9-A为化合物9-1与化合物9-2结构之一)和化合物9-B(化合物9-B为化合物9-1与化合物9-2结构之一)。
分析方法(仪器及制备柱:高效液相色谱仪-正相色谱,制备柱型号是:AD-H(4.6*2.50mm,5um)流动相体系:正己烷:乙醇(90:10),柱温:35℃,流速:1.0mL/min。保留时间T=8.549min为化合物9-A(化合物9-A为化合物9-1与化合物9-2结构之一)。保留时间T=10.887min为化合物9-B(化合物9-B为化合物9-1与化合物9-2结构之一)。
化合物9-A:
LCMS m/z=458.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.54(s,1H),8.39(d,1H),7.81(s,1H),7.77(dd,1H),7.68-7.60(m,3H),7.49-7.40(m,1H),7.23-7.16(m,1H),6.50(t,1H),6.41(d,1H),5.28-5.15(m,1H),3.26-3.18(m,1H),2.98-2.81(m,3H),2.07(s,3H),2.05-1.98(m,2H),1.62(d,3H).
化合物9-B:
LCMS m/z=458.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.60(s,1H),8.40(d,1H),7.81(s,1H),7.77(dd,1H),7.70-7.62(m,3H),7.49-7.40(m,1H),7.23-7.16(m,1H),6.50(t,1H),6.41(d,1H),5.28-5.17(m,1H),3.26-3.17(m,1H),2.98-2.79(m,3H),2.07(s,3H),2.05-1.97(m,2H),1.62(d,3H).
实施例10:化合物10的制备
化合物10以8g、4,4-二氟哌啶为底物,参考实施例8的合成方法得到。
LCMS m/z=483.2[M+H]+
实施例11:化合物11的制备
化合物11以5a、丁酰氯为底物,参考实施例8的合成方法得到。
LCMS m/z=513.3[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.62(s,1H),8.44(d,1H),7.80(dd,1H),7.69(s,1H),7.54-7.46(m,1H),7.33(dd,1H),7.25-7.13(m,2H),6.51(t,1H),6.41(d,1H),5.33-5.21(m,1H), 5.21-5.11(m,4H),3.24-3.14(m,1H),2.93-2.71(m,5H),2.07-1.93(m,2H),1.68(d,3H),1.12(t,3H).
实施例12:化合物12的制备
第一步:12a的制备
将三苯基膦(1.24g,4.71mmol),邻苯二甲酰亚胺(693mg,4.71mmol)加入到11g(1.00g,3.14mmol)的四氢呋喃(90mL)溶液中,氮气置换后,降温至0℃,加入偶氮二甲酸二异丙酯(952mg,4.71mmol),室温反应16h。加入水(100mL),乙酸乙酯(50mL x 2)萃取。合并有机相,有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=82:18)得12a(988mg,产率70%)。
LCMS m/z=448.1[M+H]+
第二步:12b的制备
将水合肼(1.5mL,80%)加入到12a(988mg,2.21mmol)的乙醇(30mL)溶液中,室温反应16h。过滤,乙醇(4mL x 3)淋洗滤饼。滤液减压浓缩后,加入乙酸乙酯(30mL)溶解,过滤,滤液减浓缩得12b(587mg,产率84%)
LCMS m/z=318.1[M+H]+
第三步:12c的制备
将6-氯-3-氟吡啶-2-甲酸甲酯(421mg,2.22mmol)和N,N-二异丙基乙胺(956mg,7.40mmol)加入到12b(587mg,1.85mmol)的二甲亚砜(15mL)中,100℃反应16h。冷却至室温,加入水(150mL),乙酸乙酯(50mL x 3)萃取。合并有机相,有机相用饱和食盐水(50mL x 3)洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=75:25)得12c(900mg,产率100%)。
LCMS m/z=487.2[M+H]+
第四步:12d的制备
0℃下,将间氯过氧苯甲酸(351mg,2.04mmol)加入到12c(900mg,1.85mmol)的二氯甲烷(15mL)溶液中,室温反应1h。加入二氯甲烷(50mL),混合物依次用饱和硫代硫酸钠水溶液(20mL),饱和碳酸氢钠水溶液(20mL),饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=60:40)得12d(639mg,产率69%)。
LCMS m/z=503.2[M+H]+
第五步:12e的制备
将N,N-二异丙基乙胺(657mg,5.08mmol)加入到12d(639mg,1.27mmol)和5-氟异吲哚啉盐酸 盐(441mg,2.54mmol)的二甲亚砜(15mL)中,90℃反应16h。冷却至室温,加入水(200mL),乙酸乙酯(50mL x 3)萃取,合并有机相。有机相用饱和食盐水(50mL x 3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=70:30)得12e(430mg,产率60%)。
LCMS m/z=562.1[M+H]+
第六步:化合物12的制备
将氢氧化锂一水合物(164mg,3.90mmol)的水(6mL)溶液加入到12e(220mg,0.39mmol)的四氢呋喃(20mL)的溶液中,室温反应16h。0℃下用1N盐酸水溶液调pH至4-5,加入饱和食盐水(30mL),乙酸乙酯(30mL x3)萃取,合并有机相。有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品用乙腈(10mL)打浆,过滤。滤饼经打浆(甲醇:二氯甲烷(v:v)=1:10,10mL)纯化,冻干得化合物12(78mg,产率36%)。
LCMS m/z=548.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.02(s,1H),8.47(s,1H),7.70(s,1H),7.49(dd,,1H),7.32(d,2H),7.22-7.12(m,1H),6.94(d,1H),5.40-4.92(m,5H),3.22-3.10(m,1H),2.94-2.69(m,5H),2.05-1.94(m,2H),1.69(d,3H),1.11(t,3H).
实施例13:化合物13-A与化合物13-B的制备
第一步:13a的制备
将5i(408mg,1.04mmol)溶于N,N-二甲基甲酰胺(15mL)中,加入邻苯二甲酰亚胺钾盐(210mg,1.13mmol),室温搅拌16h。加入水(30mL),乙酸乙酯(30mL x 2)萃取。合并有机相,有机相用饱和食盐水(30mL x 3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。粗品经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=85:15)得13a(215mg,产率45%)。
第二步:13b的制备
将水合肼(515mg,8.23mmol,80%)滴入到13a(215mg,0.47mmol)的无水乙醇溶液(10mL)中,室温反应16h。减压浓缩,加入乙酸乙酯(10mL),过滤,滤饼用乙酸乙酯冲洗。滤液减压浓缩得13b(150mg,产率97%)。
LCMS m/z=328.2[M+1]+
第三步:13c的制备
将13b(485mg,1.48mmol)和6-氯-3-氟吡啶-2-甲酸甲酯(337mg,1.78mmol)溶于N,N-二甲基甲酰胺(15mL)中。加入N,N-二异丙基乙胺(765mg,5.92mmol),100℃反应16h。冷却至室温,加入水(30mL),乙酸乙酯(30mL x 2)萃取。合并有机相,有机相用饱和食盐水(30mL x 3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=87:13)得13c(476mg,产率65%)。
LCMS m/z=497.3[M+1]+
第四步:化合物13的制备
将13c(376mg,0.76mmol)溶于四氢呋喃(30mL)和水(11mL)中,加入一水合氢氧化锂(160mg,3.82mmol),室温反应4h。0℃下加入盐酸(1N)调节pH至4-5,乙酸乙酯(50mL x 2)萃取。合并有机相,有机相用饱和食盐水(60mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(二氯甲烷:四氢呋喃(v:v)=80:20)得化合物13(340mg,产率93%)。
LCMS m/z=481.3[M-1]-
第五步:化合物13-A与化合物13-B的制备
化合物13经SFC on AD column纯化(仪器及制备柱:采用Waters 150 Prep-SFC,制备柱型号:Chiral AD column。制备方法:化合物13用甲醇溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:A for CO2and B for i-PrOH(0.1%NH3·H2O)。梯度洗脱方法:20%流动相B等梯度洗脱(流速:10mL/min;洗脱时间6min))得化合物13-A(化合物13-A为化合物13-1与化合物13-2结构之一)和化合物13-B(化合物13-B为化合物13-1与化合物13-2结构之一)。
分析方法(仪器及制备柱:高效液相色谱仪-正相色谱,制备柱型号是:AD-H(4.6*2.50mm,5um)流动相体系:正己烷(0.1%二乙胺):乙醇(90:10),柱温:35℃,流速:1.0mL/min。保留时间T=10.255min为化合物13-A(化合物13-A为化合物13-1与化合物13-2结构之一)。保留时间T=12.455min为化合物13-B(化合物13-B为化合物13-1与化合物13-2结构之一)。
化合物13-A:
LCMS m/z=483.1[M+H]+
1H NMR(400MHz,CDCl3)δ10.64(s,1H),8.34(d,1H),7.93(s,1H),7.18(d,1H),6.81(d,1H),5.16-5.07(m,1H),3.22-3.09(m,1H),3.00-2.82(m,3H),2.66(d,6H),2.16-2.05(m,5H),1.73(d,3H).
化合物13-B:
LCMS m/z=483.1[M+H]+
1H NMR(400MHz,CDCl3)δ8.34(d,1H),7.93(s,1H),7.18(d,1H),6.81(d,1H),5.18-5.06(m,1H),3.21-3.09(m,1H),3.00-2.82(m,3H),2.66(d,6H),2.15-2.05(m,5H),1.73(d,3H).
实施例14:化合物14-A与化合物14-B的制备
化合物14以5d、3-(三氟甲基)双环[1.1.1]戊烷-1-羧酸为起始原料,参照实施例13得到。化合物14经SFC on AD column拆分(仪器及制备柱:采用Waters Prep-SFC 150AP,制备柱型号是:Chiralcel AD column。制备方法:化合物14用乙腈溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:A for CO2and B for ACN(0.1%三氟乙酸)。梯度洗脱方法:15%流动相B等梯度洗脱(流速:40mL/min;洗脱时间15min))得化合物14-A(化合物14-A为化合物14-1与化合物14-2结构之一)和化合物14-B(化合物14-B为化合物14-1与化合物14-2结构之一)。
分析方法(仪器及制备柱:高效液相色谱仪-正相色谱,制备柱型号是:AD-H(4.6*2.50mm,5um)流动相体系:正己烷:乙醇(0.1%三氟乙酸)(90:10),柱温:35℃,流速:1.0mL/min。保留时间T=8.051min为化合物14-A(化合物14-A为化合物14-1与化合物14-2结构之一)。保留时间T=11.667min为化合物14-B(化合物14-B为化合物14-1与化合物14-2结构之一)。
化合物14-A:
LCMS m/z=533.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.97(s,1H),8.44(d,1H),7.75(s,1H),7.38(d,1H),7.04(d,1H),5.27-5.18(m,1H),3.22-3.13(m,1H),2.98-2.81(m,3H),2.61(s,6H),2.08-1.98(m,5H),1.66(d,3H).
化合物14-B:
LCMS m/z=533.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.97(s,1H),8.44(d,1H),7.75(s,1H),7.38(d,1H),7.05(d,1H),5.27-5.16(m,1H),3.22-3.13(m,1H),2.96-2.82(m,3H),2.61(s,6H),2.08-2.00(m,5H),1.66(d,3H).
实施例15:化合物15-A与化合物15-B的制备
化合物15以11c、3-(三氟甲基)双环[1.1.1]戊烷-1-羧酸为起始原料,参照实施例13得到。化合物15经SFC on AD column拆分(仪器及制备柱:采用Waters Prep-SFC 150AP,制备柱型号是:Chiralcel AD column。制备方法:化合物15用乙腈溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:A for CO2and B for ACN(0.1%三氟乙酸)。梯度洗脱方法:15%流动相B等梯度洗脱(流速:40mL/min;洗脱时间15min))得化合物15-A(化合物15-A为化合物15-1与化合物15-2结构之一)和化合物15-B(化合物15-B为化合物15-1与化合物15-2结构之一)。
分析方法(仪器及制备柱:高效液相色谱仪-正相色谱,制备柱型号是:AD-H(4.6*2.50mm,5um)流动相体系:正己烷:乙醇(0.1%三氟乙酸)(90:10),柱温:35℃,流速:1.0mL/min。保留时间T=5.908min为化合物15-A(化合物15-A为化合物15-1与化合物15-2结构之一)。保留时间T=8.258min为化合物15-B(化合物15-B为化合物15-1与化合物15-2结构之一)。
化合物15-A:
LCMS m/z=547.2[M+H]+
1H NMR(400MHz,CDCl3)δ8.34(d,1H),7.93(s,1H),7.19(d,1H),6.81(d,1H),5.19-5.08(m,1H),3.21-3.10(m,1H),2.99-2.82(m,3H),2.67-2.59(m,2H),2.56(s,6H),2.16-2.03(m,2H),1.74(d,3H),1.15(t,3H).
化合物15-B:
LCMS m/z=547.2[M+H]+
1H NMR(400MHz,CDCl3)δ8.34(d,1H),7.93(s,1H),7.19(d,1H),6.81(d,1H),5.19-5.09(m,1H),3.21-3.10(m,1H),2.96-2.83(m,3H),2.67-2.59(m,2H),2.57(s,6H),2.15-2.05(m,2H),1.74(d,3H),1.16(t,3H).
实施例16:化合物16-A与化合物16-B的制备
16a以8e为起始原料,参照实施例13得到,化合物16以16a为起始原料,参照实施例12得到。化合物16经SFC on IG-H column拆分(仪器及制备柱:采用Waters Prep-SFC 150AP,制备柱型号:大赛璐IG-H column。制备方法:化合物16用DMF溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:A for CO2and B for乙醇。梯度洗脱方法:60%流动相B等梯度洗脱(流速:35mL/min;洗脱时间28min))得化合物16-A(保留时间T=19min为化合物16-A,化合物16-A为化合物16-1与化合物16-2结构之一)和化合物16-B(保留时间T=24min为化合物16-B,化合物16-B为化合物16-1与化合物16-2结构之一)。
化合物16-A:
LCMS m/z=534.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.01(s,1H),8.44(d,1H),7.70(s,1H),7.46(dd,,1H),7.36-7.26(m,2H),7.21-7.13(m,1H),6.96(d,1H),5.35-5.08(m,5H),3.22-3.10(m,1H),2.93-2.69(m,3H),2.25(s,3H),2.05-1.94(m,2H),1.69(d,3H).
化合物16-B:
LCMS m/z=534.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.02(s,1H),8.44(d,1H),7.70(s,1H),7.46(dd,,1H),7.36-7.26(m,2H),7.21-7.13(m,1H),6.96(d,1H),5.32-5.11(m,5H),3.22-3.11(m,1H),2.92-2.69(m,3H),2.25(s,3H),2.06-1.93(m,2H),1.69(d,3H)..
实施例17:化合物17-A与化合物17-B的制备
化合物17以11c、3-氟二环[1.1.1]戊烷-1-羧酸为底物,参考实施例5的合成方法得到。化合物17经SFC on AD column纯化(仪器及制备柱:采用Waters 150 Prep-SFC,制备柱型号是:Chiralpak AD column。制备方法:化合物17用乙腈溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:A for CO2and B for MeOH(0.1%NH3·H2O)。梯度洗脱方法:25%流动相B等梯度洗脱(流速:70mL/min;洗脱时间6min))得化合物17-A(化合物17-A为化合物17-1与化合物17-2结构之一)和化合物17-B(化合物17-B为化合物17-1与化合物17-2结构之一)。
分析方法(仪器及制备柱:高效液相色谱仪-正相色谱,制备柱型号是:Chiralpak AD column,流动相体系:A for CO2;B for 0.05%DEA in MEOH,柱温:35℃,流速:3.0mL/min。保留时间T=1.190min为化合物17-A(化合物17-A为化合物17-1与化合物17-2结构之一)。保留时间T=1.309min为化合物17-B(化合物17-B为化合物17-1与化合物17-2结构之一)。
化合物17-A:
LCMS m/z=460.2[M-H]-
1H NMR(400MHz,CDCl3)δ10.24(s,1H),8.19(d,1H),7.98(dd,1H),7.92(s,1H),7.24-7.21(m,1H),6.59(t,1H),6.41(d,1H),5.26-5.13(m,1H),3.28-3.12(m,1H),3.04-2.81(m,3H),2.70-2.56(m,8H),2.16-2.02(m,2H),1.70(d,3H),1.15(t,3H).
化合物17-B:
LCMS m/z=460.2[M-H]-
1H NMR(400MHz,CDCl3)δ10.22(s,1H),8.19(d,1H),7.98(dd,1H),7.91(s,1H),7.27-7.22(m,1H),6.59(t,1H),6.41(d,1H),5.27-5.12(m,1H),3.29-3.12(m,1H),3.03-2.80(m,3H),2.71-2.54(m,8H),2.16-2.02(m,2H),1.70(d,3H),1.14(t,3H).
实施例18:化合物18-A与化合物18-B的制备
第一步:化合物18a的制备
0℃下,将偶氮二甲酸二异丙酯(425mg,2.10mmol)加入到三苯基膦(550mg,2.10mmol),17g(600mg,1.75mmol)的四氢呋喃(10mL)溶液中,氮气氛围下室温反应16h。加入水(30mL),乙酸乙酯(20mL x 2)萃取,合并有机相。有机相经饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩后得粗品,粗品经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=83:17)得18a(660mg,产率80%)。
LCMS m/z=472.2[M+H]+
第二步:化合物18b的制备
将水合肼(1mL,80%)加入到18a(660mg,1.40mmol)的乙醇(20mL)溶液中,室温反应16h。过滤,滤饼用乙酸乙酯洗涤(10mL x 3),滤液经减压浓缩得粗品。粗品用乙酸乙酯(10mL)打浆,过滤,滤饼用乙酸乙酯洗涤(10mL x 3),滤液经减压浓缩得化合物18b(342mg,粗品)。
LCMS m/z=342.4[M+H]+
第三步:化合物18c的制备
将6-氯-3-氟吡啶-2-甲酸甲酯(227mg,1.20mmol)和N,N-二异丙基乙胺(517mg,4.00mmol)加到18b(342mg,粗品)的二甲亚砜(5mL)中,100℃反应16h。冷却至室温,加入水(20mL),乙酸乙酯(30mL x 2)萃取。合并有机相,有机相用饱和食盐水(40mL x 3)洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=82:18)得18c(442mg,两步产率62%)。
LCMS m/z=511.1[M+H]+
第四步:化合物18的的制备
将氢氧化水溶液(2N,8.60mmol,4.3mL)加入到18c(442mg,0.86mmol)的甲醇(8mL)和四氢呋喃(8mL)溶液中,室温反应1h。减压浓缩,0℃下用1N盐酸水溶液将残留物调pH至6-7,乙酸乙酯(20mL x 2)萃取。合并有机相,有机相经无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经打浆(石油醚:乙酸乙酯(v:v)=20:1,30mL)得化合物18(318mg,产率74%)。
LCMS m/z=497.2[M+H]+
1H NMR(400MHz,Chloroform-d)δ10.64(s,1H),8.33(d,1H),7.93(s,1H),7.19(d,1H),6.81(d,1H),5.19-5.06(m,1H),3.22-3.09(m,1H),3.02-2.79(m,3H),2.65(d,6H),2.63-2.56(m,2H),2.15-2.05(m,2H),1.73(d,3H),1.13(t,3H).
第五步:化合物18-A与化合物18-B的制备
化合物18经SFC on AD column纯化(仪器及制备柱:采用Waters 150 Prep-SFC,制备柱型号是:Chiralcel AD column。制备方法:化合物18用乙腈溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:A for CO2and B for IPA(0.1%NH3·H2O)。梯度洗脱方法:30%流动相B等梯度洗脱(流速:120mL/min;洗脱时间5min))得化合物18-A(化合物18-A为化合物18-1与化合物18-2结构之一)和化合物18-B(化合物18-B为化合物18-1与化合物18-2结构之一)。
分析方法(仪器及制备柱:高效液相色谱仪-正相色谱,分析柱型号是:Chiralpak AS column流动相体系:A for CO2;B for 0.05%DEA in IPA,柱温:35℃,流速:3.0mL/min。保留时间T=1.289min为化合物18-A(化合物18-A为化合物18-1与化合物18-2结构之一)。保留时间T=1.519min为化合物18-B(化合物18-B为化合物18-1与化合物18-2结构之一)。
化合物18-A:
LCMS m/z=497.1[M+H]+
1H NMR(400MHz,CDCl3)δ10.26(s,1H),8.34(d,1H),7.93(s,1H),7.18(d,1H),6.81(d,1H),5.19-5.05(m,1H),3.24-3.05(m,1H),3.01-2.79(m,3H),2.71-2.52(m,8H),2.18-2.03(m,2H),1.73(d,3H),1.13(t,3H).
化合物18-B:
LCMS m/z=497.1[M+H]+
1H NMR(400MHz,CDCl3)δ8.35(d,1H),7.93(s,1H),7.18(d,1H),6.81(d,1H),5.19-5.05(m,1H),3.22-3.09(m,1H),3.02-2.80(m,3H),2.72-2.52(m,8H),2.18-2.01(m,2H),1.73(d,3H),1.13(t,3H).
实施例19:化合物19的制备
化合物19以5d、1-三氟甲基环丙烷-1-甲酸为起始物料,参考实施例18的合成方法得到。
LCMS m/z=505.4[M-H]-
1H NMR(400MHz,DMSO-d6)δ13.00(s,1H),8.40(d,1H),7.77(s,1H),7.30(d,1H),6.88(d,1H),5.25-5.08(m,1H),3.30-3.15(m,1H),3.00-2.80(m,3H),2.12-1.99(m,5H),1.66(d,3H),1.65-1.40(m,4H).
实施例20:化合物20的制备
第一步:20b的制备
将N-溴代丁二酰亚胺(5.49g,30.83mmol)加入到20a(5.00g,30.83mmol)的N,N-二甲基甲酰胺(100mL)溶液中,室温反应16h。将反应液倒入到水(500mL)中,过滤,滤饼用水(100mL x 3)淋洗后,减压干燥得粗品,粗品经打浆(石油醚:乙酸乙酯(v:v)=10:1,100mL)得20b(6.30g,产率85%)。
LCMS m/z=241.0[M+H]+
第二步:20c的制备
将硼氢化钠(1.57g,41.50mmol)分批加入到20b(2.00g,8.30mmol)的甲醇(100mL)和四氢呋喃(20mL)溶液中,室温反应16h。滴加1N盐酸水溶液调pH至5左右,加入硅胶,减压浓缩得残留物,残留物经柱层析分离纯化(石油醚:四氢呋喃(v:v)=80:20)得20c(1.64g,产率81%)。
LCMS m/z=225.1[M-18]+
第三步:20d的制备
将钠氢(444mg,11.10mmol,60%)分批加入到20c(0.90g,3.70mmol)的N,N-二甲基甲酰胺(40mL)溶液中,室温反应0.5h。缓慢滴加苄溴(1.90g,11.10mmol),室温反应16h。加入饱和氯化铵溶液(200mL),乙酸乙酯(50mL x 3)萃取,合并有机相。有机相经饱和食盐水洗涤(50mL x 5),无水硫酸钠干燥,抽滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=84:6)得20d(1.57g,产率100%)。
第四步:20e的制备
-78℃下,将丁基锂(14.5mL,2.5M,36.25mmol)滴加到20d(6.10g,14.41mmol)的四氢呋喃(60mL)溶液中,-78℃反应1h。加入N-甲氧基-N-甲基-丙酰胺(4.22g,36.02mmol),-78℃反应20h。-78℃下,加入饱和氯化铵水溶液(200mL),乙酸乙酯(50mL x 3)萃取,合并有机相。有机相经饱和食盐水洗涤(50mL),无水硫酸钠干燥,抽滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=84:6)得20e(2.30g,产率40%)。
LCMS m/z=401.1[M+H]+
第五步:20f的制备
将钯碳(212mg,10%)加入到20e(2.10g,5.24mmol)的甲醇(100mL)溶液中,氢气氛围下室温反应16h。垫硅藻土过滤,甲醇(30x 3)淋洗,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=86:4)得20f(1.60g,产率98%)。
LCMS m/z=309.1[M-H]-
第六步:20g的制备
0℃下,将双(三甲基硅基)氨基钠(3.15mL,2.0M,6.30mmol)滴加到20f(653mg,2.10mmol)的四氢呋喃(17mL)溶液中,室温反应1h。0℃下滴加二硫化碳(480mg,6.30mmol),30℃反应24h。0℃下加入稀硫酸(15%,8.5mL),室温搅拌0.5h。加入水20mL,乙酸乙酯(40mL x 2)萃取,合并有机相。有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经打浆(石油醚:乙酸乙酯(v:v)=10:1,15mL)得20g(714mg,产率96%)。
LCMS m/z=353.1[M+H]+
第七步:20h的制备
将碳酸钾(0.39g,2.81mmol)、碘乙烷(1.47g,9.36mmol)加入到20g(823mg,2.34mmol)的丙酮(20mL)溶液中,60℃反应3h。冷却至室温,加入硅胶,减压浓缩得残留物,残留物经柱层析分离纯化(石油醚:乙酸乙酯(v:v)=85:15)得20h(848mg,产率95%)。
LCMS m/z=381.3[M+H]+
第八步:20i的制备
0℃下,将间氯过氧苯甲酸(408mg,2.36mmol)加入到20h(748mg,1.97mmol)的二氯甲烷(35mL)溶液中,室温反应1h。加入二氯甲烷(50mL),混合物依次用饱和硫代硫酸钠水溶液(20mL),饱和碳酸氢钠水溶液(20mL),饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=70:30)得20i(544mg,产率70%)。
第九步:20j的制备
将N,N-二异丙基乙胺(646mg,5.00mmol)加到20i(494mg,1.25mmol)和5-氟异吲哚啉盐酸盐(434mg,2.50mmol)的二甲亚砜(20mL)中,100℃反应16h。加入水(200mL),二氯甲烷(50mL x 3)萃取,合并有机相。有机相用饱和食盐水(50mL x 2)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=60:40)得20j(418mg,产率74%)。
LCMS m/z=456.2[M+H]+
第十步:20k的制备
将氢氧化钯(212mg,10%)加入到20j(212mg,0.47mmol)的甲醇(212mL)溶液中,室温反应96h。垫硅藻土过滤,甲醇(50x 3)淋洗,滤液减压浓缩得20k(136mg,产率80%)。。
LCMS m/z=366.3[M+H]+
第十一步:20l的制备
0℃下,将三溴化磷200mg,0.74mmol)滴加到20k(136mg,0.37mmol)的二氯甲烷(20mL)溶液中,室温反应2h。反应液直接用于下一步。
第十二步:20m的制备
0℃下,将上一步反应液缓慢滴加到二氯甲烷(20mL)和氨水(20mL)的混合溶液中,室温反应16h。分层,水相用二氯甲烷(50mL x 2)萃取,合并有机相。无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(二氯甲烷:甲醇(v:v)=94:6)得20m(32mg,两步产率24%)。
LCMS m/z=365.1[M+H]+
第十三步:20n的制备
将6-氯-3-氟吡啶-2-甲酸甲酯(31mg,0.18mmol)和N,N-二异丙基乙胺(46mg,0.35mmol)加到20m(32mg,0.09mmol)的二甲亚砜(2mL)中,100℃反应48h。冷却至室温,加入水(20mL),乙酸乙酯(20mL x 3)萃取。合并有机相,有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤。滤液减压浓缩得粗品,粗品经柱层析分离纯化(石油醚:四氢呋喃(v:v)=50:50)得20n(33mg,产率70%)。
LCMS m/z=534.2[M+H]+
第十四步:化合物20的制备
将氢氧化锂一水合物(55mg,1.30mmol)的水(2mL)溶液加入到20n(70mg,0.13mmol)的四氢呋喃(7mL)的溶液中,室温反应16h。0℃下用1N盐酸水溶液调pH至4-5,加入食盐水(30mL),乙酸乙酯(30mL x3)萃取,合并有机相。有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经制备板纯化(二氯甲烷:甲醇(v:v)=10:1),冻干得化合物20(2mg,产率3%)。
LCMS m/z=520.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ9.50-9.04(m,1H),7.77(s,1H),7.48-7.29(m,2H),7.21-7.04(m,2H),6.92(d,1H),5.60-5.40(m,1H),5.00-4.60(m,4H),3.16-3.04(m,1H),2.95-2.83(m,1H),2.73-2.61(m,1H),2.33(s,3H),2.18(s,3H),2.06-1.94(m,1H).
实施例21:化合物21-A与化合物21-B的制备
化合物21以9e为原料,参照实施例15得到。化合物21经SFC on AS column拆分(仪器及 制备柱:采用Waters Prep-SFC 150AP,制备柱型号是:Chiralcel AS column。制备方法:化合物15用甲醇溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:A for CO2and B for甲醇。梯度洗脱方法:45%流动相B等梯度洗脱(流速:31mL/min;洗脱时间11.5min))得化合物21-A(化合物21-A为化合物21-1与化合物21-2结构之一)和化合物21-B(化合物21-B为化合物21-1与化合物21-2结构之一)。
分析方法(仪器及制备柱:高效液相色谱仪-正相色谱,制备柱型号是:OJ-H(4.6*2.50mm,5um)流动相体系:正己烷:乙醇(80:20),柱温:35℃,流速:1.0mL/min。保留时间T=11.643min为化合物21-A(化合物21-A为化合物21-1与化合物21-2结构之一)。保留时间T=18.430min为化合物21-B(化合物21-B为化合物21-1与化合物21-2结构之一)。
化合物21-A:
LCMS m/z=493.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.97(s,1H),8.36(d,1H),7.83(s,1H),7.70-7.60(m,3H),7.48-7.41(m,1H),7.32(d,1H),6.97(d,1H),5.26-5.15(m,1H),3.27-3.14(m,1H),2.99-2.77(m,3H),2.11-1.96(m,5H),1.64(d,3H).
化合物21-B:
LCMS m/z=493.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ8.81-8.54(m,1H),7.82(s,1H),7.70-7.61(m,3H),7.50-7.40(m,1H),7.24(d,1H),6.90(d,1H),5.24-5.13(m,1H),3.25-3.14(m,1H),2.97-2.78(m,3H),2.10-1.94(m,5H),1.62(d,3H).
实施例22:化合物22的制备
22i的制备
将22h(800mg,1.91mmol,22h以5a为原料,参考实施例5得到)溶于1,4-二氧六环(20mL),加入氨水(10mL,28%),50℃下,封管反应2h。冷却至室温,减压浓缩,加入水,二氯甲烷(30mL x 2)萃取。合并有机相,有机相经无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。粗品经柱层析分离纯化(二氯甲烷:甲醇(v:v)=95:5)得22i(100mg,产率17%)。
化合物22以22i、6-氯-3-氟吡啶-2-甲酸甲酯为底物,参考实施例13得到。
LCMS m/z=469.4[M+H]+
实施例23:化合物23的制备
23a的制备
0℃下,将氢氧化钾水溶液(3mL,40%wt)缓慢加入到22c(2.0g,7.84mmol)、间氟苯甲醛(1.21g,9.72mmol)的无水乙醇(40mL)溶液中,室温反应16h。0℃下,用1N盐酸调PH值至5-6,有固体析出。过滤,滤饼经干燥得到23a(2.5g,产率51%)。
LCMS m/z=361.1[M+H]+
化合物23以23a为原料,参考实施例22得到。
LCMS m/z=479.1[M+H]+
实施例24:化合物24的制备
24k的制备
将24j(500mg,1.06mmol,24j以24a为原料,参考实施例13得到)溶于异丙醇(24mL)和水(4mL)溶液中,加入硼氢化钠(150mg,3.96mmol),室温反应2h。0℃下,加入饱和氯化铵 水溶液(3mL),减压浓缩得残余物,加入水,二氯甲烷(40mL x 2)萃取。合并有机相,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。将粗品溶于异丙醇(27mL),加入乙酸(4.5mL),80℃下封管反应3h。冷却至室温,减压浓缩,加入水,0℃下,用氨水(28%)调PH值至8左右,二氯甲烷萃取(30mL x 5)。合并有机相,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩得粗品,粗品经柱层析分离纯化(二氯甲烷:甲醇(v:v)=95:5)得24k(182mg,产率50%)。
化合物24以24k、6-氯-3-氟吡啶-2-甲酸甲酯为底物,参考实施例13得到。
LCMS m/z=497.2[M+H]+
实施例25:化合物25-A与化合物25-B的制备
化合物22经SFC on AD column纯化(仪器及制备柱:采用Waters 150 Prep-SFC,制备柱型号是:Chiral AD column。制备方法:化合物22用乙腈溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:A for CO2and B for isopropanol(0.1%NH3·H2O)。梯度洗脱方法:20%流动相B等梯度洗脱(流速:120mL/min;洗脱时间3.5min)),冻干后得到化合物25-A(化合物25-A为化合物25-1与化合物25-2结构之一)和化合物25-B(化合物25-B为化合物25-1与化合物25-2结构之一)。
分析方法(仪器及制备柱:高效液相色谱仪-正相色谱,制备柱型号是:OJ-H(4.6*2.50mm,5um)流动相体系:正己烷:乙醇(80:20),柱温:35℃,流速:1.0mL/min。
化合物25-A:保留时间T=15.244min(化合物25-A为化合物25-1与化合物25-2结构之一)
LCMS m/z=469.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.05(s,1H),8.46(d,1H),7.75(s,1H),7.39(d,1H),7.01(d,1H),6.27(s,1H),5.20-5.10(m,1H),3.26-3.17(m,1H),3.04-2.84(m,3H),2.62-2.53(m,6H),2.16-2.00(m,2H),1.66(d,3H).
化合物25-B:保留时间T=21.290min(化合物25-B为化合物25-1与化合物25-2结构之一)
LCMS m/z=469.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ13.06(s,1H),8.45(d,1H),7.75(s,1H),7.39(d,1H),7.01(d,1H),6.27(s,1H),5.21-5.10(m,1H),3.28-3.18(m,1H),3.05-2.84(m,3H),2.61-2.52(m,6H),2.16-1.99(m,2H),1.66(d,3H).
实施例26:化合物26-A与化合物26-B的制备
化合物26的制备
将26a(400mg,0.82mmol,26a以22h为原料,参考实施例5得到)溶于二氯甲烷(10mL),加入三氟乙酸(10mL),30℃下,封管反应16h,50℃下反应2h。冷却至室温,加入水,二氯甲烷(20mL x 2)萃取。合并有机相,有机相经无水硫酸钠干燥,过滤,滤液减压浓缩得粗品。粗品经制备板分离纯化(石油醚:乙酸乙酯(v:v)=5:2)得化合物26(220mg,产率62%)。
化合物26经SFC on OD column拆分(仪器及制备柱:采用Waters 150 Prep-SFC,制备柱型号是:Chiral OD column。制备方法:化合物26用甲醇和乙腈溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:A for CO2and B for methanol(0.1%NH3·H2O)。梯度洗脱方法:25%流动相B等梯度洗脱(流速:100mL/min;洗脱时间3.2min))得化合物26-A(化合物26-A为化合物26-1与化合物26-2结构之一)和化合物26-B(化合物26-B为化合物26-1与化合物26-2结构之一)。
分析方法(仪器及制备柱:SHIMADZU LC-30AD sf,制备柱型号是:Chiral OD column。流动相体系:A for CO2and B for methanol(0.05%DEA),梯度洗脱方法:5%-40%流动相B等梯度洗脱,柱温:35℃,流速:3.0mL/min。保留时间T=1.583min为化合物26-A(化合物26-A为化合物26-1与化合物26-2结构之一)。保留时间T=1.765min为化合物26-B(化合物26-B为化合物26-1与化合物26-2结构之一)。
化合物26-A:
LCMS m/z=434.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.70(s,1H),8.51(s,1H),7.78(dd,1H),7.73(s,1H),7.23(t,1H),6.50(t,1H),6.43(d,1H),6.26(s,1H),5.18-5.07(m,1H),3.26-3.18(m,1H),3.08-2.98(m,1H),2.97-2.83(m,2H),2.62-2.53(m,6H),2.17-1.98(m,2H),1.64(d,3H).
化合物26-B:
LCMS m/z=434.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.73(s,1H),8.52(s,1H),7.78(dd,1H),7.73(s,1H),7.23(t,1H),6.50(t,1H),6.43(d,1H),6.26(s,1H),5.18-5.07(m,1H),3.26-3.18(m,1H),3.08-2.98(m,1H),2.97-2.84(m,2H),2.62-2.53(m,6H),2.16-1.98(m,2H),1.64(d,3H).
实施例27:化合物27-A与化合物27-B的制备
化合物27以23e为原料,参照实施例26得到。化合物27经SFC on AS column拆分(仪器及制备柱:采用Waters 150 Prep-SFC,制备柱型号是:Chiral AS column。制备方法:化合物27用甲醇和乙腈溶解,并用0.45μm滤膜过滤,制备成样品液。流动相体系:A for CO2and B for methanol(0.1%NH3·H2O)。梯度洗脱方法:15%流动相B等梯度洗脱(流速:80mL/min;洗脱时间4.0min))得化合物27-A(化合物27-A为化合物27-1与化合物27-2结构之一)和化合物27-B(化合物27-B为化合物27-1与化合物27-2结构之一)。
分析方法(仪器及制备柱:SHIMADZU LC-30AD sf,制备柱型号是:Chiral AS column。流动相体系:A for CO2and B for methanol(0.05%DEA),梯度洗脱方法:5%-40%流动相B等梯度洗脱,柱温:35℃,流速:3.0mL/min。保留时间T=1.888min为化合物27-A(化合物27-A为化合物27-1与化合物27-2结构之一)。保留时间T=2.404min为化合物27-B(化合物27-B为化合物27-1与化合物27-2结构之一)。
化合物27-A:
LCMS m/z=444.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.61(s,1H),8.54(d,1H),8.04-7.93(m,2H),7.82-7.75(m,2H),7.70-7.61(m,1H),7.51-7.42(m,1H),7.24-7.16(m,1H),7.14(s,1H),6.55-6.46(m,2H),5.47-5.36(m,1H),3.26-3.19(m,1H),2.98-2.80(m,3H),2.09-1.98(m,2H),1.72(d,3H).
化合物27-B:
LCMS m/z=444.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ12.62(s,1H),8.54(d,1H),8.03-7.93(m,2H),7.82-7.75(m,2H),7.70-7.60(m,1H),7.50-7.43(m,1H),7.24-7.16(m,1H),7.14(s,1H),6.56-6.45(m,2H),5.47-5.35(m,1H),3.26-3.19(m,1H),2.99-2.80(m,3H),2.07-1.99(m,2H),1.72(d,3H).
化合物A与化合物B参考专利WO2022235574A1的合成方法得到。
生物测试例
1、MDA-MB-453细胞增殖抑制实验
方法1:MDA-MB-453细胞(ATCC,Cat#30-2008)用L-15培养基(添加10%FBS和1%双抗)在无CO2条件下培养,待细胞汇合度达到80%-90%时收集细胞铺板。弃培养基,用1×PBS润洗后加入胰酶(Gibco,Cat#15400-054)消化,待细胞变圆并开始脱落时加入培养基终止消化,将细胞吹打下来并转移至无菌离心管中,1000rpm离心3分钟,离心完毕取出,弃上清液。离心管加入培养基重悬细胞,计数。根据计数结果将细胞悬液调整到适当浓度后加入96孔细胞培养板(Corning,Cat#3903)中。用DMSO将化合物溶解至10mM保存备用,实验时将化合物母液依次进行梯度稀释。铺板24小时后加入化合物,在37℃、无CO2条件下继续培养4天。孵育结束后每孔加入CELL VIABILITY试剂(Promega,Cat#G7573),室温孵育10分钟,孵育结束后轻轻振摇5次,然后用BMG酶标仪(PHERAstar FSX)检测化学发光读数。按公式[(1-(RLUcompound-RLUblank)/(RLUcontrol-RLU blank))×100%]计算细胞增殖抑制率。使用GraphPad Prism软件通过四参数非线性拟合获得IC50值。
方法2(MTS检测):MDA-MB-453细胞(ATCC,Cat#HTB-131)用L-15培养基(添加10%FBS)培养,取处于对数生长期的细胞用于铺板。弃培养基,用1×PBS润洗后加入胰酶(Gibco,Cat#15400-054)消化,待细胞变圆并开始脱落时加入培养基终止消化,将细胞吹打下来并转移至无菌离心管中,1000rpm离心3分钟,离心完毕取出,弃上清液。离心管加入培养基重悬细胞,计数。根据计数结果将细胞悬液调整到适当浓度后加入96孔细胞培养板(Greiner,Cat#655090)中。用DMSO将化合物溶解至10mM保存备用,实验时将化合物母液依次进行梯度稀释。铺板24小时后加入化合物,在37℃、5%CO2条件下继续培养7天。孵育结束后每孔加入CellTiterAQueous One Solution Reagent(Promega,Cat#G3582),将细胞培养板放置于在37℃,0%CO2的培养箱孵育4小时,然后用SpectraMax 340PC酶标仪检测OD490nm。按公式[1-(RLU化合物-RLU空白对照)/(RLU溶媒对照-RLU空白对照)]×100%计算细胞增殖抑制率。使用GraphPad Prism软件作抑制曲线图和计算IC50值。
表1化合物对MDA-MB-453细胞的细胞增殖抑制活性

备注:*为方法1测试结果。
结论:本发明的化合物,例如实施例化合物,对MDA-MB-453细胞有良好的抑制活性。
2、T47D细胞增殖抑制实验
T47D细胞(ATCC,Cat#HTB-133)用RPMI Medium 1640培养基(添加10%FBS,1%双抗及0.02mg/mL牛胰岛素)在37℃,5%CO2条件下培养,待细胞汇合度达到80%-90%时收集细胞铺板。弃培养基,用1×PBS润洗后加入胰酶(Gibco,Cat#15400-054)消化,待细胞变圆并开始脱落时加入培养基终止消化,将细胞吹打下来并转移至无菌离心管中,1000rpm离心3分钟,离心完毕取出,弃上清液。离心管加入培养基重悬细胞,计数。根据计数结果将细胞悬液调整到适当浓度后加入96孔细胞培养板(LABSELECT,Cat#11515)中。用DMSO将化合物溶解至10mM保存备用,实验时将化合物母液依次进行梯度稀释。铺板24小时后加入化合物,在37℃、5%CO2下继续培养4天。孵育结束后每孔加入CELL VIABILITY试剂(Promega,Cat#G7573),轻轻振摇3分钟后,室温孵育10分钟,然后用BMG酶标仪(PHERAstar FSX)检测化学发光读数。按公式[(1-(RLUcompound-RLUblank)/(RLUcontrol-RLUblank))×100%]计算细胞增殖抑制率。使用GraphPad Prism软件通过四参数非线性拟合获得IC50值。
表2化合物对T47D细胞的细胞增殖抑制活性
结论:本发明的化合物,例如实施例化合物,对T47D细胞有良好的抑制活性。
3、PI3K酶活测试方法
在384 PP Plate化合物稀释板中使用DMSO对化合物进行梯度稀释,使用Echo转移0.1μL化合物到384反应微孔板(Optiplate 384)中,确保DMSO终浓度为1%(双复孔)。向384反应微孔板每孔中加入5μL 2X酶溶液(PIK3CA、PIK3CA[H1047R]),1000rpm离心1分钟,在25℃孵育10分钟。向每孔中加入5μL 2X ATP与PIP2:3PS溶液,1000rpm离心1分钟,在25℃孵育180分钟。向每孔中加入5μL ADP-Glo Reagent,1000rpm离心1分钟,在25℃孵育40分钟。向每孔中加入10μL ADP-Glo Detection buffer,1000rpm离心1分钟,在25℃孵育40分钟。在BMG(PHERAstar FSX)酶标仪上读取RLU(Relative luminescence unit)信号。含有1%DMSO和酶的孔作为High Control,含有1%DMSO和缓冲液的孔作为Low Control。化合物处理孔的抑制百分比在High Control和Low Control之间标准化(%inhibition=(RLU High Control-RLU化合物读值)/(RLU High Control-RLU Low Control)*100)。然后通过XLfit 5.5.0拟合四参数IC50曲线并分析,IC50是抑制率为50%所对应的化合物浓度,见表2。
表3化合物对PIK3CA[H1047R]、WT PIK3CA酶的抑制活性

测试结果:本发明的化合物,例如实施例化合物对PIK3CA[H1047R]酶具有良好的抑制活性,对WT PIK3CA酶抑制活性差,具有良好的选择性,具体的如化合物6-B、化合物26-B、化合物27-B对PIK3CA[H1047R]酶的抑制活性IC50分别为77.4nM、54.8nM、34.6nM。
4、化合物对细胞中P-AKT的抑制作用
4.1化合物对MDA-MB-453细胞中P-AKT的抑制作用
MDA-MB-453细胞(ATCC,Cat#HTB-131)用L-15培养基(添加10%FBS和1%双抗)在37℃无CO2条件下培养,待细胞汇合度达到80%-90%时收集细胞铺板。弃培养基,用1×PBS润洗后加入胰酶(Gibco,Cat#15400-054)消化,待细胞变圆并开始脱落时加入培养基终止消化,将细胞吹打下来并转移至无菌离心管中,1000rpm离心3分钟,离心完毕取出,弃上清液。离心管加入培养基重悬细胞,计数。根据计数结果将细胞悬液调整到适当浓度后加入384孔板(Perkin Elmer,Cat:6007680)中。用DMSO将化合物溶解至10mM保存备用,实验时将化合物母液依次进行梯度稀释。铺板18小时后加入化合物,在37℃、无CO2条件下继续培养2h。孵育结束后每孔加入Lysis Buffer(Perkin Elmer,Cat:ALSU-PAKT-B500),室温震荡10分钟。每孔加入Acceptor Mix,室温震荡1-2min混合均匀后,室温静置孵育1h。每孔加入Donor Mix,避光室温震荡1-2min混合均匀后,室温静置孵育1h。然后用BMG酶标仪(PHERAstar FSX)检测,使用GraphPad Prism软件通过四参数非线性拟合获得IC50值。
4.2化合物对SKBR3细胞中P-AKT的抑制作用
SKBR3细胞(ATCC,Cat#HTB-30)用McCoy's 5A Medium培养基(添加10%FBS和1%双抗)在37℃、5%CO2条件下培养,待细胞汇合度达到80%-90%时收集细胞铺板。弃培养基,用1×PBS润洗后加入胰酶(Gibco,Cat#15400-054)消化,待细胞变圆并开始脱落时加入培养基终止消化,将细胞吹打下来并转移至无菌离心管中,1000rpm离心3分钟,离心完毕取出,弃上清液。离心管加入培养基重悬细胞,计数。根据计数结果将细胞悬液调整到适当浓度后加入384孔板(Perkin Elmer,Cat:6007680)中。用DMSO将化合物溶解至10mM保存备用,实验时将化合物母液依次进行梯度稀释。铺板18小时后加入化合物,在37℃、5%CO2条件下继续培养2h。孵育结束后每孔加入Lysis Buffer(Perkin Elmer,Cat:ALSU-PAKT-B500),室温震荡10分钟。每孔加入Acceptor Mix,室温震荡1-2min混合均匀后,室温静置孵育1h。每孔加入Donor Mix,避光室温震荡1-2min混合均匀后,室温静置孵育1h。然后用BMG酶标仪(PHERAstar FSX)检测,使用GraphPad Prism软件通过四参数非线性拟合获得IC50值。
表3化合物对细胞中P-AKT的抑制活性

测试结果:本发明的化合物,例如实施例化合物对MDA-MB-453细胞中P-AKT具有良好抑制活性,对SKBR3细胞中P-AKT抑制活性差,具有良好的选择性。
5.化合物对人脂肪细胞中葡萄糖摄取的抑制作用
将3T3-L1分化的脂肪细胞在96孔板中培养,在实验当天将其改为不含FBS的DMEM培养基饥饿培养2小时。用DMSO将化合物溶解至10mM保存备用,实验时将化合物母液依次进行梯度稀释。用缓冲液清洗3T3-L1脂肪细胞后,将化合物加入对应孔中,在5%CO2培养箱中37℃孵育60分钟。在人胰岛素对照孔、加药孔、阴性孔(含100μM cytochalasin B)中加入25μL人胰岛素,在空白对照孔中加入25μL实验缓冲液,在5%CO2培养箱中37℃孵育10分钟。用实验缓冲液制备[3H]-脱氧-d-葡萄糖溶液,将其加入检测孔中,在5%CO2培养箱中37℃孵育20分钟。除去检测孔中的溶液,用冷DPBS清洗以停止反应。脂肪细胞用10%NaOH溶解,将溶解液转移到Pico Prias管中,并加入Ultima Gold。然后使用Tri-Carb(PerkinElmer,4910TR)读数,使用GraphPad Prism软件通过四参数非线性拟合获得IC50值。
表4化合物对人脂肪细胞中葡萄糖摄取的抑制活性
测试结果:本发明的化合物,例如实施例化合物对人脂肪细胞中葡萄糖摄取的抑制活性差,不会引起高血糖副作用。
5.小鼠药代动力学测试
5.1试验动物:雄性Balb/c小鼠,22g左右,6~8周龄,6只/化合物。购于成都达硕实验动物有限公司。
5.2试验设计:试验当天,Balb/c小鼠按体重随机分组。给药前1天禁食不禁水12~14h,给药后4h给食。
表5.给药信息
静脉给药溶媒:5%DMA+5%Solutol+90%Saline;灌胃给药溶媒:0.5%MC
于给药前及给药后异氟烷麻醉经眼眶取血0.03ml,置于EDTAK2离心管中,5000rpm,4℃离心10min,收集血浆。静脉组和灌胃组采血时间点:0,2,5,15,30min,1,2,4,7h;分析检测前,所有样品存于-80℃。
结论:本发明化合物,例如实施例化合物具有良好的药代动力学性质,在小鼠上具有较高的 暴露量,良好的生物利用度。
6、大鼠药代动力学测试
6.1试验动物:雄性SD大鼠,220g左右,6~8周龄,6只/化合物。购于成都达硕实验动物有限公司。
6.2试验设计:试验当天,SD大鼠按体重随机分组。给药前1天禁食不禁水12~14h,给药后4h给食。
表7.给药信息
注:静脉给药溶媒:5%DMA+5%Solutol+90%Saline;灌胃给药溶媒:0.5%MC
(DMA:二甲基乙酰胺;Solutol:聚乙二醇-15-羟基硬脂酸酯;Saline:生理盐水;MC:甲基纤维素)
于给药前及给药后异氟烷麻醉经眼眶取血0.15ml,置于EDTAK2离心管中,5000rpm,4℃离心10min,收集血浆。静脉组和灌胃组采血时间点均为:0,5,15,30min,1,2,4,6,8,24h。分析检测前,所有样品存于-80℃,用LC-MS/MS对样品进行定量分析。
表8.测试化合物在大鼠血浆中的药代动力学参数
-:不适用。
结论:本发明化合物,例如实施例化合物具有良好的药代动力学性质,具体的如化合物6-B、化合物26-B在大鼠上具有较高的暴露量,良好的生物利用度。
7.CYP450酶抑制测试
本项研究的目的是应用体外测试体系评价受试物对人肝微粒体细胞色素P450(CYP)的5种同工酶(CYP1A2、CYP2C19、CYP2D6和CYP3A4)活性的影响。CYP450同工酶的特异性探针底物分别与人肝微粒体以及不同浓度的受试物共同孵育,加入还原型烟酰胺腺嘌呤二核苷酸磷酸(NADPH)启动反应,在反应结束后,通过处理样品并采用液相色谱-串联质谱联用(LC-MS/MS)法定量检测特异性底物产生的代谢产物,测定CYP酶活性的变化,计算IC50值,评价受试物对各CYP酶亚型的抑制潜能。。
结论:本发明化合物,例如实施例化合物对CYP酶抑制活性差,具体的如化合物25-B对CYP1A2、CYP2C19、CYP2D6和CYP3A4的IC50值均大于30μM。
8.hERG钾离子通道作用测试
实验平台:电生理手动膜片钳系统
细胞系:稳定表达hERG钾离子通道的中国仓鼠卵巢(CHO)细胞系
实验方法:稳定表达hERG钾通道的CHO(Chinese Hamster Ovary)细胞,在室温下用全细胞膜片钳技术记录hERG钾通道电流。玻璃微电极由玻璃电极毛胚(BF150-86-10,Sutter)经拉制仪拉制而成,灌注电极内液后的尖端电阻为2-5MΩ左右,将玻璃微电极插入放大器探头即可连接至膜片钳放大器。钳制电压和数据记录由pClamp 10软件通过电脑控制和记录,采样频率为10kHz,滤波频率为2kHz。在得到全细胞记录后,细胞钳制在-80mV,诱发hERG钾电流(IhERG)的步阶电压从-80mV给予一个2s的去极化电压到+20mV,再复极化到-50mV,持续1s后回到-80mV。每10s给予此电压刺激,确定hERG钾电流稳定后(至少1分钟)开始给药过程。化合物每个测试浓度至少给予1分钟,每个浓度至少测试2个细胞(n≥2)。
数据处理:数据分析处理采用pClamp 10,GraphPad Prism 5和Excel软件。不同化合物浓度对hERG钾电流(-50mV时诱发的hERG尾电流峰值)的抑制程度用以下公式计算:
Inhibition%=[1-(I/Io)]×100%
其中,Inhibition%代表化合物对hERG钾电流的抑制百分率,I和Io分别表示在加药后和加药前hERG钾电流的幅度。
化合物IC50使用GraphPad Prism 5软件通过以下方程拟合计算得出:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))
其中,X为供试品检测浓度的Log值,Y为对应浓度下抑制百分率,Bottom和Top分别为最小和最大抑制百分率。
表9:化合物对hERG钾通道电流抑制作用的IC50
结论:本发明化合物,例如实施例化合物对hERG抑制活性差,具体的如化合物6-B和化合物25-B对hERG钾通道电流抑制作用的IC50大于20μM。
9.肝微粒体稳定性测试
本实验采用人肝微粒体作为体外模型来评价受试物的代谢稳定性。
在37℃条件下,1μM的受试物与微粒体蛋白、辅酶NADPH共同孵育,反应至一定时间(5,10,20,30,60min)加入冰冷含内标的乙腈终止反应,采用LC-MS/MS方法检测样品中受试物浓度,以孵育体系中药物剩余率的ln值和孵育时间求得T1/2,并进一步计算肝微粒体肝固有清除率CLint(Liver)
表10.人肝微粒体稳定性结果
结论:本发明化合物,例如实施例化合物,具体的如化合物13-B、化合物25-B在人肝微粒 体代谢稳定性良好。
10.比格犬药代动力学测试
实验目的:通过单剂量静脉和灌胃给予受试物于比格犬,测定比格犬血浆中受试物的浓度,评价受试物在比格犬体内药代特征。
试验动物:雄性比格犬,8~11kg左右,6只/化合物,购于成都达硕实验动物有限公司。
试验方法:试验当天,比格犬按体重随机分组。给药前1天禁食不禁水12~14h,给药后4h给食。
表11.给药信息
静脉给药溶媒:5%DMA+5%Solutol+90%Saline;灌胃给药溶媒:0.5%MC
取样:于给药前及给药后通过四肢静脉取血1ml,置于EDTAK2离心管中。5000rpm,4℃离心10min,收集血浆。
采血时间点:0,5,15,30min,1,2,4,6,8,10,12,24h。分析检测前,所有样品存于-80℃。用LC-MS/MS对样品进行定量分析。
结论:本发明化合物,例如实施例化合物在比格犬具有较高的暴露量,良好的药代动力学性质。
11.猴药代动力学测试
11.1试验动物:雄性食蟹猴,3~5kg,3~6年龄,6只/化合物。购于苏州西山生物技术有限公司。
11.2试验方法:试验当天,食蟹猴按体重随机分组。给药前1天禁食不禁水14~18h,给药后4h给食。
表12.给药信息
注:静脉给药溶媒:5%DMA+5%Solutol+90%Saline;灌胃给药溶媒:0.5%MC(含0.5%Tween 80);
(DMA:二甲基乙酰胺;Solutol:聚乙二醇-15-羟基硬脂酸酯;Saline:生理盐水;MC:甲基纤维素溶液;)
*剂量以游离碱计。
于给药前及给药后通过四肢静脉取血1.0mL,置于EDTAK2离心管中。5000rpm,4℃离心 10min,收集血浆。静脉组和灌胃组采血时间点均为:0,5min,15min,30min,1,2,4,6,8,10,12,24,48h。分析检测前,所有样品存于-80℃,用LC-MS/MS对样品进行定量分析。
结论:本发明的化合物,例如实施例化合物,在猴上具有较高的暴露量,良好的药代动力学性质。

Claims (11)

  1. 一种化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,其中化合物选自通式(I)所述的化合物,
    X2选自O、S;
    X1选自O、S、NRx、C(Rx)2
    Y选自O、S、NRy
    W选自C2-4炔基、所述的炔基任选被1至4个Rk所取代;
    Z选自键或-C≡C-;
    m选自1、2、3、4;
    Rx、Ry各自独立的选自H、氘、C1-6烷基、C2-6烯基、C2-6炔基、-C0-4亚烷基-C3-10碳环基、-C0-4亚烷基-4至10元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
    环A选自C3-12碳环基、4至12元杂环基、C6-10芳基或5至10元杂芳基,所述的碳环基、杂环基、芳基或杂芳基任选被1至4个选自Ra所取代;
    环B选自C3-12碳环基、4至12元杂环基、C6-10芳基或5至10元杂芳基,所述的碳环基、杂环基、芳基或杂芳基任选被1至4个选自Rb所取代;
    Ra、Rb各自独立的选自氘、卤素、=O、OH、CN、NO2、NH2、C1-6烷基、OC1-6烷基、SC1-6烷基、C2-6烯基、C2-6炔基、NHC1-6烷基、N(C1-6烷基)2、-C0-4亚烷基-C(=O)R1a、-C0-4亚烷基-S(=O)2R1a、-C0-4亚烷基-P(=O)R1aR1b、-O-C0-4亚烷基-C3-10碳环基、-O-C0-4亚烷基-4至10元杂环基、-S-C0-4亚烷基-C3-10碳环基、-S-C0-4亚烷基-4至10元杂环基、-NH-C0-4亚烷基-C3-10碳环基、-NH-C0-4亚烷基-4至10元杂环基、-C0-4亚烷基-C3-10碳环基、-C0-4亚烷基-4至10元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
    R1、R2、R3、R4各自独立地选自H、氘、卤素、OH、CN、NO2、NH2、C1-6烷基、OC1-6烷基、SC1-6烷基、C2-6烯基、C2-6炔基、NHC1-6烷基、N(C1-6烷基)2、-C0-4亚烷基-C(=O)R1a、-C0-4亚烷基-S(=O)2R1a、-C0-4亚烷基-P(=O)R1aR1b、-O-C0-4亚烷基-C3-10碳环基、-O-C0-4亚烷基-4至10元杂环基、-S-C0-4亚烷基-C3-10碳环基、-S-C0-4亚烷基-4至10元杂环基、-NH-C0-4亚烷基-C3-10碳环基、-NH-C0-4亚烷基-4至10元杂环基、-C0-4亚烷基-C3-10碳环基、-C0-4亚烷基-4至10元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
    R5、R6各自独立的选自H、氘、卤素、OH、CN、NO2、NH2、C1-6烷基、OC1-6烷基、SC1-6 烷基、C2-6烯基、C2-6炔基、NHC1-6烷基、N(C1-6烷基)2、C0-4亚烷基-5至10元杂芳基、C0-4亚烷基-C3-10碳环基、C0-4亚烷基-4至10元杂环基,所述的烷基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
    R1a、R1b各自独立的选自H、OH、NH2、C1-6烷基、C1-6烷氧基、NHC1-6烷基、N(C1-6烷基)2、C3-8碳环基、4至8元杂环基、C6-10芳基、5至10元杂芳基、-O-C3-8碳环基、-O-4至8元杂环基,所述的烷基、烷氧基、碳环基、杂环基、芳基或杂芳基任选被1至4个Rk所取代;
    R2与R3、R3与R4、R4与R5、R6与Ry、R6与Rb中至少有一组和与其相连的原子共同形成C3-12碳环基或3至12元杂环基,所述的碳环基或杂环基任选被1至4个Rk所取代;
    Rk各自独立的选自氘、卤素、OH、=O、CN、NH2、COOH、CONH2、C1-6烷基、OC1-6烷基、
    SC1-6烷基、C2-6烯基、C2-6炔基、NHC1-6烷基、N(C1-6烷基)2、-O-C3-6碳环、-O-3至7元杂环、-NH-C3-6碳环、-NH-3至7元杂环、-C0-4亚烷基-C3-6碳环、-C0-4亚烷基-3至7元杂环,所述的烷基、亚烷基、烯基、炔基、碳环或杂环任选被1至4个选自氘、卤素、=O、CN、OH、NH2、C1-6烷基、C1-6烷氧基的取代基所取代。
  2. 根据权利要求1所述化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,其中化合物选自通式(II)、(III)、(IV)、(V)、(VI),
    Rx、Ry各自独立的选自H、氘、C1-4烷基、C2-4烯基、C2-4炔基、-C0-4亚烷基-C3-8碳环基、-C0-4亚烷基-4至8元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
    R1a、R1b各自独立的选自H、OH、NH2、C1-4烷基、C1-4烷氧基、NHC1-4烷基、N(C1-4烷基)2、C3-6碳环基、4至8元杂环基、C6-10芳基、5至10元杂芳基、-O-C3-8碳环基、-O-4至8元杂环基,所述的烷基、烷氧基、碳环基、杂环基、芳基或杂芳基任选被1至4个Rk所取代;
    环A各自独立的选自4-7元杂单环、5-12元杂并环、5-12元杂螺环、7-10元杂桥环、C3-8单碳环基、C6-12并环烷基、C6-12元螺环烷基、C5-12元桥环烷基、5至10元杂芳基、苯并C3-8碳环基、苯并4至8元杂环基,环A以碳碳键与Z相连,所述的环A任选被1至4个选自Ra所取代;
    环B各自独立的选自4-7元杂单环、5-12元杂并环、5-12元杂螺环、7-10元杂桥环、C3-8单 碳环基、C6-14并环烷基、C6-12元螺环烷基、C5-12元桥环烷基、苯并C3-8碳环基、苯并3至8元杂环基、C6-10芳基、5-10元杂芳基,所述的环B任选被1至4个Rb所取代;
    环C1、C2、C3各自独立的选自4-8元杂环或C4-8碳环,所述的环C1、C2、C3任选被1至4个Rk所取代;
    环C4选自4-8元杂环,所述的环C4任选被1至4个Rk所取代;
    环C5选自4-8元含氮杂环,所述的环C5任选被1至4个Rk所取代;
    Rk各自独立的选自氘、卤素、OH、=O、CN、NH2、COOH、CONH2、C1-4烷基、OC1-4烷基、SC1-4烷基、C2-4烯基、C2-4炔基、NHC1-4烷基、N(C1-4烷基)2、-O-C3-6碳环、-O-3至7元杂环、-NH-C3-6碳环、-NH-3至7元杂环、-C0-4亚烷基-C3-6碳环、-C0-4亚烷基-3至7元杂环,所述的烷基、亚烷基、烯基、炔基、碳环或杂环任选被1至4个选自氘、卤素、=O、CN、OH、NH2、C1-4烷基、C1-4烷氧基的取代基所取代。
  3. 根据权利要求2所述化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,其中,
    环B各自独立的选自C6-10芳基、5-10元杂芳基,所述的环B任选被1至4个Rb所取代;
    或者选自n选自1、2、3;
    B1、B2、B3、B4各自独立的选自N或CRb1,B1、B2、B3、B4中至多两个选自N;
    B5各自独立的选自N、O、S、S(O)2、CRb2、NRb2或C(Rb2)2
    Rb2各自独立的选自H或Rk
    Rb1各自独立的选自H或Rb
    R1、R2、R3、R4各自独立地选自H、氘、卤素、OH、CN、NO2、NH2、C1-4烷基、OC1-4烷基、SC1-4烷基、C2-4烯基、C2-4炔基、NHC1-4烷基、N(C1-4烷基)2、-C0-4亚烷基-C(=O)R1a、-C0-4亚烷基-S(=O)2R1a、-C0-4亚烷基-P(=O)R1aR1b、-O-C0-4亚烷基-C3-8碳环基、-O-C0-4亚烷基-4至8元杂环基、-S-C0-4亚烷基-C3-8碳环基、-S-C0-4亚烷基-4至8元杂环基、-NH-C0-4亚烷基-C3-8碳环基、-NH-C0-4亚烷基-4至8元杂环基、-C0-4亚烷基-C3-8碳环基、-C0-4亚烷基-4至8元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
    R5、R6各自独立的选自H、氘、卤素、OH、CN、NO2、NH2、C1-4烷基、OC1-4烷基、SC1-4烷基、C2-4烯基、C2-4炔基、NHC1-4烷基、N(C1-4烷基)2、C0-4亚烷基-5至8元杂芳基、C0-4亚烷基-C3-8碳环基、C0-4亚烷基-4至8元杂环基,所述的烷基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代;
    Ra、Rb各自独立的选自氘、氘、卤素、=O、OH、CN、NO2、NH2、C1-4烷基、OC1-4烷基、SC1-4烷基、C2-4烯基、C2-4炔基、NHC1-4烷基、N(C1-4烷基)2、-C0-4亚烷基-C(=O)R1a、-C0-4亚烷基-S(=O)2R1a、-C0-4亚烷基-P(=O)R1aR1b、-O-C0-4亚烷基-C3-8碳环基、-O-C0-4亚烷基-4至8元杂环基、-S-C0-4亚烷基-C3-8碳环基、-S-C0-4亚烷基-4至8元杂环基、-NH-C0-4亚烷基-C3-8碳环基、-NH-C0-4亚烷基-4至8元杂环基、-C0-4亚烷基-C3-8碳环基、-C0-4亚烷基-4至8元杂环基,所述的烷基、烯基、炔基、亚烷基、碳环基或杂环基任选被1至4个Rk所取代。
  4. 根据权利要求3所述化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,
    X1选自O、S、NRx
    Rx、Ry各自独立的选自H、氘、甲基、乙基、丙基、异丙基、乙烯基、乙炔基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、哌嗪基、吗啉基,所述的甲基、乙基、丙基、异丙基、乙烯基、乙炔基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、哌嗪基、吗啉基任选被1至4个Rk所取代;
    R1a、R1b各自独立的选自H、OH、NH2、NHCH3、N(CH3)2、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基、-O-环丙基、-O-环丁基、咪唑、吡唑、吡咯或噻吩,所述的甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基、咪唑、吡唑、吡咯或噻吩任选被1至3个Rk所取代;
    R1、R2、R3、R4各自独立地选自H、氘、F、Cl、Br、I、OH、CN、NO2、NH2、NHCH3、N(CH3)2、甲基、乙基、丙基、异丙基、乙烯基、乙炔基、丙炔基、炔丙基、甲硫基、-C(=O)R1a、-S(=O)2R1a、-P(=O)R1aR1b、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基,所述的甲基、乙基、丙基、异丙基、乙烯基、乙炔基、丙炔基、炔丙基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基任选被1至4个Rk所取代;
    R5、R6各自独立的选自H、氘、F、Cl、Br、I、OH、CN、NO2、NH2、NHCH3、N(CH3)2、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基,所述的甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基任选被1至4个Rk所取代;
    Ra各自独立的选自氘、F、Cl、Br、I、OH、=O、NH2、CN、NO2、NHCH3、N(CH3)2、COOH、CONH2或任选被1至4个Rk取代的如下基团之一:甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、苯基、吡啶基、吡嗪基、哒嗪基、嘧啶基、吡唑基、吡咯基、咪唑基、噻吩基、噻唑基、呋喃基、噁唑基、异噁唑基;
    Rb各自独立的选自氘、F、Cl、Br、I、OH、=O、CN、NH2、NO2、NHCH3、N(CH3)2、COOH、CONH2、-CH2-C(=O)R1a、-CH2-S(=O)2R1a、-CH2-P(=O)R1aR1b、-C(=O)R1a、-S(=O)2R1a、-P(=O)R1aR1b、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基、-CH2-环丙基、-CH2-环丁基、-CH2-吡咯烷基、-O-环丙基、-O-环丁基、-O-吡咯烷基、-S-环丙基、-S-环丁基、-NH-环丙基、-NH-环丁基、-OCH2-环丙基、咪唑、吡唑、吡咯或噻吩,所述的甲基、乙基、丙基、异丙基、甲氧基、乙氧基、异丙氧基、乙烯基、乙炔基、甲硫基、环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吗啉基、咪唑、吡唑、吡咯或噻吩 任选被1至4个Rk所取代;
    环C1、C2、C3各自独立的选自任选被1至4个Rk取代的如下基团之一:环丁烯基、环戊烯基、环己烯基、环庚烯基、氧杂环丁烯基、氧杂环戊烯基、氧杂环己烯基、氮杂环丁烯基、氮杂环戊烯基、氮杂环己烯基、1,3-二氧环戊烯基、1,4-二氧环己烯基;
    环C5选自任选被1至4个Rk取代的如下基团之一:氮杂环丁基、吡咯烷基、哌啶基、吗啉基。
  5. 根据权利要求4所述化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,
    W选自乙炔基、丙炔基、炔丙基、所述的乙炔基、丙炔基、炔丙基、任选被1至4个Rk所取代;
    Y选自O、S、NH;
    Rk各自独立的选自氘、F、Cl、Br、I、OH、=O、CN、NH2、COOH、CONH2、NHCH3、N(CH3)2、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、甲硫基、乙烯基、乙炔基、丙炔基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吡唑基、吡咯基、吗啉基,所述的甲基、乙基、丙基、异丙基、甲氧基、乙氧基、甲硫基、乙烯基、乙炔基、丙炔基、环丙基、环丁基、氮杂环丁基、氧杂环丁基、吡咯烷基、哌啶基、吡唑基、吡咯基、吗啉基任选被1至4个选自氘、F、Cl、Br、I、=O、CN、OH、NH2、C1-4烷基、C1-4烷氧基的取代基所取代;
    环B各自独立的选自任选取代的如下结构之一:苯、萘、吡啶、吡嗪、哒嗪、嘧啶、 当被取代时,被1至3个Rb所取代;
    环A各自独立的选自任选取代的如下结构之一:环丙基、环丁基、环戊基、环己基、氮杂环丁基、氧杂环丁基、四氢呋喃基、四氢吡喃基、1,3-二氧环戊烷基、1,4-二氧环己基、吡咯烷基、哌啶基、哌嗪基、吗啉基、苯基、吡啶基、吡嗪基、哒嗪基、嘧啶基、吡唑基、吡咯基、咪唑基、噻吩基、噻唑基、呋喃基、噁唑基、异噁唑基、苯并吡咯基、苯并噻吩基、苯并呋喃基、苯并噻唑基、苯并吡唑基、苯并咪唑基、喹啉基、异喹啉基、吡啶并吡啶基、吡啶并噻唑基、吡啶并呋喃基、吡啶并吡咯基、异吲哚啉基、吲哚啉基、 当被取代时,被1至3个Ra取代;
    或者选自
  6. 根据权利要求5所述化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,
    选自
    选自
    选自
    各自独立的选自
    或者选自
    选自其右端与环B相连接。
  7. 根据权利要求1所述化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,其中该化合物选自如表A结构之一。
  8. 一种药物组合物,包括权利要求1-7任意一项所述的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,以及药学上可接受的载体。
  9. 根据权利要求1-7任意一项所述的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶、或权利要求8所述的药物组合物在用于制备治疗与PI3Kα相关疾病的药物中的应用。
  10. 根据权利要求9所述的应用,其中,所述的疾病选自肿瘤或癌症,优选自乳腺癌。
  11. 一种用于治疗哺乳动物的疾病的方法,所述方法包括给予受试者治疗有效量的权利要求1-8任意一项所述的化合物或者其立体异构体、氘代物、溶剂化物、前药、代谢产物、药学上可接受的盐或共晶,治疗有效量优选1-1500mg,所述的疾病优选与PI3Kα相关疾病。
PCT/CN2023/139226 2022-12-16 2023-12-15 一种含羧基的杂环衍生物及其在医药上的应用 WO2024125644A1 (zh)

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
CN202211584724 2022-12-16
CN202211584724.6 2022-12-16
CN202310039796 2023-01-13
CN202310039796.0 2023-01-13
CN202310352369 2023-04-04
CN202310352369.8 2023-04-04
CN202310648744 2023-06-02
CN202310648744.3 2023-06-02
CN202310924194.3 2023-07-26
CN202310924194 2023-07-26
CN202311209072.2 2023-09-19
CN202311209072 2023-09-19

Publications (1)

Publication Number Publication Date
WO2024125644A1 true WO2024125644A1 (zh) 2024-06-20

Family

ID=91484405

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/139226 WO2024125644A1 (zh) 2022-12-16 2023-12-15 一种含羧基的杂环衍生物及其在医药上的应用

Country Status (1)

Country Link
WO (1) WO2024125644A1 (zh)

Similar Documents

Publication Publication Date Title
CN115192577A (zh) Kras突变蛋白抑制剂
EP3766882B1 (en) Phthalazine isoxazole alkoxy derivatives, preparation method thereof, pharmaceutical composition and use thereof
WO2019232724A1 (en) Compounds as nuclear transport modulators and uses thereof
AU2007279166A1 (en) N-(aminoheteroaryl)-1H-indole-2-carboxamide derivatives, preparation thereof and therapeutic use thereof
CN118043314A (zh) 一种苯并氮杂芳环衍生物及其在医药上的应用
WO2019076336A1 (zh) 含吡唑基的三并环类衍生物、其制备方法和应用
WO2023143424A1 (zh) 一种氮杂并环衍生物及其在医药上的应用
WO2020062883A1 (zh) 基于知母菝契皂苷元结构的衍生物、药物组合物及其应用
WO2023030453A1 (zh) 一种降解Bcl-2家族蛋白的化合物及其在医药上的应用
WO2023125182A1 (zh) 一种丙酸衍生物及其在医药上的应用
WO2024125644A1 (zh) 一种含羧基的杂环衍生物及其在医药上的应用
WO2023025324A1 (zh) 作为p53调节剂的化合物
WO2021228216A1 (zh) 可用作RORγ调节剂的联芳基类化合物
WO2024109918A1 (zh) 一种gspt1降解剂及其在医药上的应用
WO2023246833A1 (zh) 一种五并五元环衍生物的药物组合物及其在医药上的应用
WO2023143384A1 (zh) 一种抑制或降解hpk1激酶的化合物及其在医药中的用途
WO2024032600A1 (zh) 一种杂环衍生物及其组合物和药学上的应用
WO2023151697A1 (zh) 一种mettl3抑制剂及组合物及其在医药上的应用
WO2023284837A1 (zh) 芳氨基衍生物雌激素受体调节剂及其用途
WO2024012570A1 (zh) 一种含氮杂环衍生物及其组合物和药学上的应用
WO2023202687A1 (zh) 一种吡嗪酮衍生物及其在医药上的应用
WO2024109799A1 (zh) 一种嘧啶衍生物及其在医药上的应用
WO2023131167A1 (zh) 一种抑制并降解irak4的化合物及其药物组合物和药学上的应用
WO2023208165A1 (zh) 一种含氮杂环衍生物及其组合物和药学上的应用
WO2023109902A1 (zh) 一种并环杂环衍生物及其在医药上的应用