WO2022166991A1 - 吲哚啉类化合物 - Google Patents

吲哚啉类化合物 Download PDF

Info

Publication number
WO2022166991A1
WO2022166991A1 PCT/CN2022/075562 CN2022075562W WO2022166991A1 WO 2022166991 A1 WO2022166991 A1 WO 2022166991A1 CN 2022075562 W CN2022075562 W CN 2022075562W WO 2022166991 A1 WO2022166991 A1 WO 2022166991A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
mmol
μmol
crude product
dichloromethane
Prior art date
Application number
PCT/CN2022/075562
Other languages
English (en)
French (fr)
Inventor
陆剑宇
丁照中
贺辉君
胡利红
黄元源
黎健
陈曙辉
Original Assignee
南京明德新药研发有限公司
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 南京明德新药研发有限公司 filed Critical 南京明德新药研发有限公司
Priority to CN202280011224.5A priority Critical patent/CN116745301A/zh
Publication of WO2022166991A1 publication Critical patent/WO2022166991A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention relates to a new class of indoline compounds, in particular to the application of compounds of formula (I) and their pharmaceutically acceptable salts in the preparation of medicines for treating related diseases.
  • the full name of PD-1 is programmed death receptor 1, and the English name is programmed death 1. It is an important immunosuppressive molecule and a member of the CD28 superfamily.
  • the full name of PD-L1 is programmed cell death-ligand 1, the English name programmed cell death-Ligand 1, is a 40kD transmembrane protein encoded by the CD274 gene.
  • PD-L1 can be induced to be expressed on the surface of T cells, B cells, dendritic cells, macrophages, mesenchymal stem cells, bone marrow-derived mast cells, and non-hematopoietic cells, in response to stimulation by interferon and other inflammatory factors. It may be rapidly upregulated in both tumor tissue and other tissues.
  • PD-L1 Activation of the PD-1/PD-L1 pathway suppresses the immune system in cancer, pregnancy, tissue transplantation, and autoimmune diseases.
  • PD-L1 can also bind to CD80 and competitively inhibit the T cell activation pathway of CD80 and ligand binding, which becomes another mechanism for PD-L1 to inhibit T cell activity.
  • the PD-1/PD-L1 signaling pathway can prevent excessive inflammation and autoimmune diseases induced by the immune system's excessive attack on tissues.
  • abnormal conditions such as tumor tissue and chronic HBV-infected tissue
  • PD-L1 is overexpressed.
  • the overexpression of PD-1/PD-L1 and the activation of the signaling pathway inhibit the activation and proliferation of functional T cells, inhibit the anti-tumor immune response, and make the immune system lose its inhibitory effect on the development of tumors, thereby accelerating the development and deterioration of tumors.
  • Several drugs have been approved for this pathway.
  • PD-L1 monoclonal antibodies such as Atezolizumab
  • Atezolizumab have been approved for urothelial carcinoma and non-small cell lung cancer indications, and more clinical studies are in progress in tumor-related indications.
  • macromolecular drugs compared with small molecules, macromolecular drugs have obvious disadvantages such as tissue penetration, pharmacokinetic properties, cost and usage. Therefore, the development of small molecule drugs on the PD-1/PD-L1 signaling pathway is still It is an unmet clinical need and has broad market prospects.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof
  • Ring A is selected from
  • L 1 and L 2 are each independently selected from -CH 2 - and -CH 2 -NH-CH 2 -;
  • Z and E are independently selected from CH and N, respectively;
  • Z 1 is selected from O and S;
  • Z 2 is selected from N and CR 9 ;
  • X is selected from N and CR 14 ;
  • Y is selected from N and CR 15 ;
  • R 1 is selected from H, CH 3 and CHF 2 ;
  • R 2 is selected from CH 3 and Cl
  • R 3 , R 4 , R 5 and R 6 are each independently selected from H, C 1-6 alkyl, OH, COOH and -C 1-3 alkyl-COOH;
  • R3 , R4 together with the atom to which they are attached form an azetidinyl, pyrrolidinyl, oxazolidinyl or piperidinyl group which Alkyl and piperidinyl are each independently optionally substituted with 1, 2 or 3 R 16 ;
  • R5 , R6 together with the atom to which they are attached form an azetidinyl, pyrrolidinyl, oxazolidinyl or piperidinyl group which Alkyl and piperidinyl are each independently optionally substituted with 1, 2 or 3 R 16 ;
  • R 7 is selected from H, F, Cl, CH 3 and CHF 2 ;
  • R 8 is selected from -OCH 3 , -O-CH 2 -F, and -O-CH 2 -CN;
  • R is selected from H, F and CN
  • R 14 and R 15 are each independently selected from H and C 1-6 alkyl
  • the above compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from
  • Ring B and Ring C are each independently selected from azetidinyl, pyrrolidinyl, oxazolidinyl, and piperidinyl;
  • Rings A , Z, R1 and R2 are as defined in the present invention.
  • the above compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from
  • Ring B and Ring C are each independently selected from azetidinyl, pyrrolidinyl, oxazolidinyl, and piperidinyl;
  • Z 2 is selected from CH 2 and O;
  • Z, Z 1 , Z 2 , E, R 1 , R 2 and R 8 are as defined in the present invention.
  • the above compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from
  • Z 3 is selected from -CH 2 - and -O-,
  • Z, Z 1 , Z 2 , E, R 1 , R 2 and R 8 are as defined in the present invention.
  • the above compound or a pharmaceutically acceptable salt thereof wherein R 3 , R 4 , R 5 and R 6 are independently selected from H, CH 3 , isopropyl, COOH and -C 1-3 alkyl-COOH, other variables are as defined in the present invention.
  • the above compound or a pharmaceutically acceptable salt thereof, wherein the structural unit independently selected from Other variables are as defined in the present invention.
  • the above compound or a pharmaceutically acceptable salt thereof, wherein the structural unit independently selected from Other variables are as defined in the present invention.
  • the above compound or a pharmaceutically acceptable salt thereof, wherein the structural unit independently selected from Other variables are as defined in the present invention.
  • the present invention also provides a compound of the following formula or a pharmaceutically acceptable salt thereof
  • the above compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from
  • the above-mentioned compounds or their pharmaceutically acceptable salts are used in the preparation of medicaments for the treatment of tumors.
  • the tumor is colon cancer, melanoma, non-small cell lung cancer, hepatocellular carcinoma, or renal cell carcinoma.
  • the compound of the present invention is a small molecule inhibitor with good inhibitory activity to PD-L1, which is very different from the existing treatment scheme (PD-L1 monoclonal antibody drug); Lower cost and more suitable for large-scale production; at the same time, the pharmacokinetic properties of small-molecule drugs are significantly different from those of macromolecular monoclonal antibody drugs, with faster half-life and larger tissue distribution, so small-molecule PD-L1 inhibitors Compared with macromolecular drugs in clinical application, it can have more flexible dosing schedules and more controllable side effects. After many rounds of search and evaluation, it was unexpectedly found that the compound of the present invention has good in vitro activity and pharmacokinetic properties, and showed high oral exposure in preclinical studies.
  • these compounds show significant efficacy in in vivo efficacy, and have better tumor (such as colon cancer, melanoma, non-small cell lung cancer, hepatocellular carcinoma or renal cell carcinoma, etc.) inhibitory activity.
  • tumor such as colon cancer, melanoma, non-small cell lung cancer, hepatocellular carcinoma or renal cell carcinoma, etc.
  • the term "pharmaceutically acceptable” refers to those compounds, materials, compositions and/or dosage forms that, within the scope of sound medical judgment, are suitable for use in contact with human and animal tissue , without excessive toxicity, irritation, allergic reactions or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • salts refers to salts of the compounds of the present invention, prepared from compounds with specific substituents discovered by the present invention and relatively non-toxic acids or bases.
  • base addition salts can be obtained by contacting such compounds with a sufficient amount of base in neat solution or in a suitable inert solvent.
  • Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amine or magnesium salts or similar salts.
  • acid addition salts can be obtained by contacting such compounds with a sufficient amount of acid in neat solution or in a suitable inert solvent.
  • Examples of pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, bicarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, Hydrogen sulfate, hydroiodic acid, phosphorous acid, etc.; and organic acid salts including, for example, acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, Similar acids such as fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-toluenesulfonic, citric, tartaric, and methanesulfonic acids; also include salts of amino acids such as arginine, etc. , and salts of organic acids such as glucuronic acid. Certain specific compounds of the present invention contain both basic and acidic functional groups and thus can be converted into either base
  • substituents do not specify which atom is connected to the substituted group, this substituent can be bonded through any of its atoms, for example, pyridyl can be used as a substituent through any one of the pyridine ring The carbon atom is attached to the substituted group.
  • the substituent can bond to any atom on the ring, for example, a structural unit It means that the substituent R can be substituted at any position on cyclohexyl or cyclohexadiene.
  • the direction of attachment is arbitrary, for example,
  • the linking group L in the middle is -MW-, at this time -MW- can connect ring A and ring B in the same direction as the reading order from left to right. It is also possible to connect ring A and ring B in the opposite direction to the reading order from left to right.
  • Combinations of the linking groups, substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
  • any one or more sites in the group can be linked to other groups by chemical bonds.
  • connection method of the chemical bond is not located, and there are H atoms at the connectable site, when the chemical bond is connected, the number of H atoms at the site will decrease correspondingly with the number of chemical bonds connected to the corresponding valence. the group.
  • the chemical bond connecting the site to other groups can be represented by straight solid line bonds straight dotted key or wavy lines express.
  • a straight solid bond in -OCH 3 indicates that it is connected to other groups through the oxygen atom in this group;
  • the straight dashed bond in the group indicates that it is connected to other groups through the two ends of the nitrogen atom in the group;
  • the wavy line in the phenyl group indicates that it is connected to other groups through the 1 and 2 carbon atoms in the phenyl group;
  • C 1-6 alkyl is used to denote a straight or branched chain saturated hydrocarbon group consisting of 1 to 6 carbon atoms.
  • the C 1-6 alkyl includes C 1-5 , C 1-4 , C 1-3 , C 1-2 , C 2-6 , C 2-4 , C 6 and C 5 alkyl and the like; it can be Is monovalent (eg methyl), divalent (eg methylene) or polyvalent (eg methine).
  • C 1-6 alkyl examples include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl , s-butyl and t-butyl), pentyl (including n-pentyl, isopentyl and neopentyl), hexyl, etc.
  • C 1-4 alkyl is used to denote a straight or branched chain saturated hydrocarbon group consisting of 1 to 4 carbon atoms.
  • the C 1-4 alkyl includes C 1-2 , C 1-3 and C 2-3 alkyl, etc.; it can be monovalent (such as methyl), divalent (such as methylene) or polyvalent (such as methine).
  • Examples of C 1-4 alkyl include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl , s-butyl and t-butyl) and so on.
  • C 1-3 alkyl is used to denote a straight or branched chain saturated hydrocarbon group consisting of 1 to 3 carbon atoms.
  • the C 1-3 alkyl group includes C 1-2 and C 2-3 alkyl groups, etc.; it can be monovalent (eg methyl), divalent (eg methylene) or multivalent (eg methine) .
  • Examples of C1-3 alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), and the like.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the acid or base containing parent compound by conventional chemical methods. Generally, such salts are prepared by reacting the free acid or base form of these compounds with a stoichiometric amount of the appropriate base or acid in water or an organic solvent or a mixture of the two.
  • the compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed in combination with other chemical synthesis methods, and those well known to those skilled in the art Equivalent to alternatives, preferred embodiments include, but are not limited to, the embodiments of the present invention.
  • the structure of the compound of the present invention can be confirmed by conventional methods well known to those skilled in the art. If the present invention relates to the absolute configuration of the compound, the absolute configuration can be confirmed by conventional technical means in the art. For example, single crystal X-ray diffraction method (SXRD), the cultured single crystal is collected by Bruker D8 venture diffractometer, the light source is CuK ⁇ radiation, and the scanning mode is: After scanning and collecting relevant data, the crystal structure was further analyzed by the direct method (Shelxs97), and the absolute configuration could be confirmed.
  • SXRD single crystal X-ray diffraction method
  • the cultured single crystal is collected by Bruker D8 venture diffractometer
  • the light source is CuK ⁇ radiation
  • the scanning mode is: After scanning and collecting relevant data, the crystal structure was further analyzed by the direct method (Shelxs97), and the absolute configuration could be confirmed.
  • the solvent used in the present invention is commercially available.
  • Pd(PPh 3 ) 4 stands for tetrakis(triphenylphosphine)palladium
  • Pd(dppf)Cl 2 ⁇ CH 2 Cl 2 stands for 1,1-bis(diphenylphosphine) dimethylene Iron dichloride palladium dichloromethane complex
  • NaBH(OAc) 3 represents sodium triacetyl borohydride
  • DIAD represents diisopropyl azodicarboxylate
  • Boc2O represents di-tert-butyl dicarbonate
  • t-BuXPhos-Pd -G3 represents methanesulfonic acid (2-di-tert-butylphosphino-2',4',6'-triisopropyl-1,1'-biphenyl)(2'-amino-1,1'- Biphenyl-2-yl)palladium(II); DIBAL-H for diisobutylaluminum hydride;
  • Figure 1 Inhibitory activity of compounds of the present invention on PD-1/PD-L1 binding.
  • Figure 2 shows the tumor-bearing volume of experimental animals.
  • Figure 3 shows the changes in body weight of experimental animals.
  • Figure 4 shows the proportion of CD3+ T cells in mCD5+ cells.
  • Figure 5 shows the fluorescence intensity of PD-L1 in cells.
  • the present invention will be described in detail by the following examples, but it does not mean any unfavorable limitation of the present invention.
  • the compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed in combination with other chemical synthesis methods, and those well known to those skilled in the art Equivalent to alternatives, preferred embodiments include, but are not limited to, the embodiments of the present invention. It will be apparent to those skilled in the art that various changes and modifications can be made to the specific embodiments of the present invention without departing from the spirit and scope of the invention.
  • Step A n-Butyllithium in n-hexane (2.5M, 8.88mL) was added dropwise to compound 1-1 (5g, 18.49mmol) and triisopropyl borate (4.17g, 22.19mmol) at -78°C
  • the reaction solution was stirred at -78°C for 1 hour, then slowly heated to 25°C and reacted for 1 hour, quenched by adding 80mL hydrochloric acid (1mol/L), adding 80mL After water, it was extracted with ethyl acetate (80 mL ⁇ 3), the combined organic phases were washed with saturated brine (50 mL ⁇ 2), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • the crude product was stirred in 40 mL of ethyl acetate at 25° C. for 0.5 h, and filtered to obtain product
  • Step B To a mixed solution of 30 mL of dioxane and 6 mL of water, compound 1-2 (3 g, 12.75 mmol), compound 1-3 (2.19 g, 12.75 mmol), potassium carbonate (3.52 g, 25.50 mmol) and Pd(PPh 3 ) 4 (1.47 g, 1.28 mmol), the reaction system was replaced with nitrogen three times, and the reaction was carried out at 85° C. under nitrogen protection for 12 hours.
  • Step C To 20 mL of dioxane solution was added compound 1-4 (1.29 g, 3.93 mmol), bisphosphonate boronate (2.00 g, 7.87 mmol), potassium acetate (772.34 mg, 7.87 mmol) and Pd(dppf)Cl 2 ⁇ CH 2 Cl 2 (321.33 mg, 393.48 ⁇ mol), the reaction system was replaced with nitrogen three times, and the reaction was carried out at 85° C. under nitrogen protection for 6 hours.
  • Step D compound 1-7 (2 g, 8.21 mmol), compound 1-6 (1.52 g, 9.86 mmol, 1.67 mL), sodium carbonate (2.18 g, 20.53 mmol) and Pd( PPh3 ) 4 (474.59 mg, 410.70 ⁇ mol) of a mixed solution of tert-butanol (15 mL) and water (15 mL), heated to 80° C. for 2 hours under nitrogen protection. The reactant was diluted with water, extracted with ethyl acetate (40 mL ⁇ 3), the organic phases were combined, washed with saturated brine (30 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step E To a mixed solution of compound 1-8 (1 g, 5.25 mmol) in dioxane (20 mL) and water (20 mL) was added potassium osmate dihydrate (9.66 mg, 26.23 ⁇ mol), stirred at 25° C. for 30 minutes , sodium periodate (2.63 g, 12.30 mmol) was added in portions.
  • Step F A solution of compound 1-9 (0.64 g, 3.32 mmol) and compound 1-10 (578.99 mg, 6.65 mmol) in dichloromethane (20 mL) was stirred at 25°C for 30 min, and NaBH(OAc) 3 (2.82 g, 13.29 mmol). The reactant was stirred at 25°C for 12 hours, diluted with water, and extracted with dichloromethane (50 mL ⁇ 3); the organic phase was washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step G Compounds 1-11 (0.1 g, 379.19 ⁇ mol), 1-12 (78.86 mg, 398.15 ⁇ mol) and hydrogen chloride in dioxane (4 M, 94.80 ⁇ L) were heated to 120 °C in tert-butanol (4 mL) , stir for two hours.
  • the reaction solution was cooled to room temperature, quenched with aqueous sodium bicarbonate solution, and extracted with dichloromethane (40 mL ⁇ 3); the organic phase was washed with saturated brine (20 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • the crude product was purified by preparative thin layer chromatography on silica gel plate separation (ethyl acetate) to give compound 1-13.
  • Step H Compound 1-13 (106 mg, 249.22 ⁇ mol), Compound 1-5 (93.12 mg, 249.22 ⁇ mol), sodium carbonate (66.04 mg, 623.06 ⁇ mol), and Pd(PPh 3 ) 4 (28.80 mg, 24.92 ⁇ mol)
  • a solution of dioxane (4 mL) and water (0.8 mL) was heated to 100°C under nitrogen and stirred for 1 hour.
  • the reaction solution was cooled to room temperature, 20 mL of water was added, filtered through celite, extracted with ethyl acetate (40 mL ⁇ 3), the organic phases were combined, washed with saturated brine (20 mL ⁇ 3), dried over anhydrous sodium sulfate, and filtered.
  • Step I A solution of compound 1-14 (150 mg, 253.34 ⁇ mol) and compound 1-15 (57.84 mg, 506.68 ⁇ mol) in dichloromethane (8 mL) was stirred at 25° C. for 30 min, and NaBH(OAc) 3 (2.82 g) was added thereto. , 13.29 mmol). The reactant was stirred at 25°C for 1 hour, diluted with water, and extracted with dichloromethane (20 mL ⁇ 3); the organic phase was washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step A To 10 mL of dioxane solution was added compound 1-13 (0.5 g, 1.18 mmol), bisphosphonate boronate (597.052 mg, 2.35 mmol), potassium acetate (346.12 mg, 3.53 mmol) and Pd(dppf)Cl 2 ⁇ CH 2 Cl 2 (96 mg, 117.56 ⁇ mol), the reaction system was replaced with nitrogen three times, and the reaction was carried out at 100° C. under nitrogen protection for 3 hours.
  • compound 1-13 0.5 g, 1.18 mmol
  • bisphosphonate boronate 597.052 mg, 2.35 mmol
  • potassium acetate 346.12 mg, 3.53 mmol
  • Pd(dppf)Cl 2 ⁇ CH 2 Cl 2 96 mg, 117.56 ⁇ mol
  • Step B To 40 mL of dioxane solution was added compound 2-2 (5 g, 24.10 mmol), bisphosphonate boronate (7.34 g, 28.92 mmol), potassium acetate (4.73 g, 48.20 mmol) and Pd (dppf)Cl 2 ⁇ CH 2 Cl 2 (1.97 g, 2.41 mmol), the reaction system was replaced with nitrogen three times, and the reaction was carried out at 85° C. under nitrogen protection for 12 hours. The reaction solution was cooled to room temperature and filtered through celite, then the filter cake was washed with 40 mL of dioxane solution, and the filtrates were combined to obtain crude compound 2-3.
  • Step C To a mixed solution of compound 2-3 (6.13 g, 24.08 mmol) and 16 mL of water in 80 mL of dioxane solution was added compound 2-4 (5.23 g, 24.08 mmol), potassium carbonate (8.32 g, 60.21 g mmol) and Pd(dppf)Cl 2 ⁇ CH 2 Cl 2 (1.97 g, 2.41 mmol), the reaction system was replaced with nitrogen three times, and the reaction was carried out at 85° C. under nitrogen protection for 4 hours.
  • Step D Trifluoromethanesulfonic anhydride (4.37 g, 15.47 mmol) was carefully added dropwise to compound 2-5 (3.15 g, 11.90 mmol) and N,N-diisopropylethylamine (9.23 g) at -78 °C , 71.41 mmol) in 60 mL of dichloromethane solution, the reaction solution was stirred at -78 ° C for 0.5 hours, then 50 mL of saturated citric acid solution was added to quench, 20 mL of water was added, and then extracted with dichloromethane (50 mL ⁇ 3), and the organic phases were combined.
  • Step E To a solution of compound 2-6 (1.3 g, 3.28 mmol) and compound 1-15 (748.07 mg, 6.55 mmol) in dichloromethane (12 mL) was added NaBH(OAc) 3 (3.47 g, 16.38 mmol). The reactant was stirred at 25°C for 2 hours, diluted with 100 mL of water, and extracted with ethyl acetate (100 mL ⁇ 2); the organic phase was washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • NaBH(OAc) 3 3.47 g, 16.38 mmol
  • Step F To a solution of compound 2-7 (0.5 g, 1.01 mmol) and di-tert-butyl dicarbonate (441.02 mg, 2.02 mmol) in tetrahydrofuran (14 mL) was added triethylamine (306.72 mg, 3.03 mmol), the reaction solution Stir at 25°C for 2 hours, add 100 mL of water and extract with ethyl acetate (100 mL ⁇ 2); the organic phase was washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step G Compound 2-8 (100 mg, 211.69 ⁇ mol), Compound 2-1 (125.95 mg, 211.69 ⁇ mol), sodium carbonate (56.09 mg, 529.23 ⁇ mol), and Pd(PPh 3 ) 4 (24.46 mg, 21.17 ⁇ mol)
  • the mixed solution of dioxane (2 mL) and water (0.4 mL) was replaced with nitrogen three times, heated to 100° C. under nitrogen protection, and stirred for 1 hour.
  • reaction solution was cooled to room temperature, 10 mL of water was added, filtered through celite, extracted with ethyl acetate (30 mL ⁇ 3), the organic phases were combined, washed with saturated brine (20 mL ⁇ 3), dried over anhydrous sodium sulfate, and filtered. After concentration, crude product was obtained. The crude product was purified by preparative thin layer chromatography on silica gel plate separation (ethyl acetate) to give compound 2-9. MS (ESI) m/z: 791.5 [M+H] + .
  • Step H Trifluoroacetic acid (924.00 mg, 8.10 mmol) was added to a solution of compound 2-9 (0.09 g, 113.73 ⁇ mol) in dichloromethane (1.8 mL), the reaction solution was stirred at 25° C. for 10 minutes, and concentrated to obtain the crude product .
  • the crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Synergi C18 150*25mm*10 ⁇ m; mobile phase: [pure water (0.225% formic acid)-acetonitrile]; acetonitrile%: 10%-40%, 10 minutes) and purified Compound 2 is obtained.
  • Step A Compound 3-1 (10 g, 46.08 mmol) and compound 3-2 (64.16 g, 368.63 mmol) were stirred together at 60° C. for 12 hours, and concentrated under reduced pressure to obtain compound 3-3.
  • Step D To compound 3-6 (0.35 g, 1.57 mmol), N,N-dimethylaniline (285.06 mg, 2.35 mmol) and benzyltriethylammonium chloride (714.41 mg, 3.14 mmol) in 8 mL of acetonitrile Phosphorus oxychloride (1.44 g, 9.41 mmol) was added to the solution, the reaction system was reacted at 75° C. for 2 hours, the reaction solution was cooled to room temperature and then concentrated.
  • Step E To a mixed solution of compound 3-7 (0.1 g, 248.00 ⁇ mol) in tetrahydrofuran (4 mL) and water (1 mL) was added potassium osmate dihydrate (913.77 ⁇ g, 2.48 ⁇ mol) and sodium periodate (265.23 mg) , 1.24 mmol), the reaction system was quenched with 20 mL of 50% sodium thiosulfate solution after reacting at 25 ° C for 3 hours, then extracted with dichloromethane (30 mL ⁇ 3), and the organic phases were combined and washed with saturated brine ( 30 mL ⁇ 2), dried over anhydrous sodium sulfate, filtered and concentrated to obtain crude compound 3-8.
  • Step F A solution of compound 3-8 (100 mg, 246.79 ⁇ mol) and compound 1-10 (43 mg, 493.59 ⁇ mol) in dichloromethane (6 mL) was stirred at 25° C. for 0.5 h, and NaBH(OAc) 3 (209.22 mg) was added thereto. , 987.18 ⁇ mol). The reactant was stirred at 25°C for 2 hours, then added with 10 mL of water, extracted with dichloromethane (20 mL ⁇ 3); the organic phase was washed with saturated brine (20 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude compound 3 -9.
  • Step G Compound 3-9 (90 mg, 188.95 ⁇ mol), Compound 1-5 (70.60 mg, 188.95 ⁇ mol), sodium carbonate (50.07 mg, 472.38 ⁇ mol), and Pd(PPh 3 ) 4 (65.50 mg, 56.69 ⁇ mol)
  • a solution of dioxane (5 mL) and water (1 mL) was heated to 100 °C under nitrogen and stirred for 1 hour.
  • reaction solution was added with 20 mL of dilution water, filtered through celite, extracted with ethyl acetate (30 mL ⁇ 3), the organic phases were combined and washed with saturated brine (30 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain Crude.
  • MS (ESI) m/z: 643.5 [M+H] + .
  • Step H A solution of compound 3-10 (40 mg, 62.20 ⁇ mol) and compound 1-15 (14.20 mg, 124.40 ⁇ mol) in dichloromethane (2 mL) was stirred at 25°C for 0.5 h, and NaBH(OAc) 3 (65.91 mg) was added thereto. , 311.00 ⁇ mol).
  • the reactant was stirred at 25°C for 0.5 hours, diluted with 10 mL of water, and extracted with dichloromethane (20 mL ⁇ 5); the organic phase was washed with saturated brine (20 mL ⁇ 2), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step A To 10 mL of dioxane solution was added compound 3-9 (0.2 g, 419.89 ⁇ mol), double pinacol boronate (213.25 mg, 839.78 ⁇ mol), potassium acetate (123.62 mg, 1.26 mmol) and Pd(dppf)Cl 2 ⁇ CH 2 Cl 2 (34.29 mg, 41.99 ⁇ mol), the reaction system was replaced with nitrogen three times, and the reaction was carried out at 95° C. under nitrogen protection for 1 hour.
  • Step B Compound 4-1 (90 mg, 171.96 ⁇ mol), Compound 2-8 (112.54 mg, 189.15 ⁇ mol), sodium carbonate (36.45 mg, 343.92 ⁇ mol), and Pd(PPh 3 ) 4 (19.87 mg, 17.20 ⁇ mol)
  • the mixed solution of dioxane (8 mL) and water (2 mL) was replaced with nitrogen three times, heated to 100° C. under nitrogen protection, and stirred for 1 hour.
  • Step C Trifluoroacetic acid (3.08 g, 27.01 mmol) was added to a solution of compound 4-2 (80 mg, 94.97 ⁇ mol) in dichloromethane (6 mL), the reaction solution was stirred at 25° C. for 30 minutes, and concentrated to obtain the crude product.
  • the crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex luna C18 150*25mm*10 ⁇ m; mobile phase: [pure water (0.225% formic acid)-acetonitrile]; acetonitrile%: 13%-43%, 10 minutes) and purified
  • the formate salt of compound 4 is obtained.
  • Step A A mixed solution of compound 5-1 (500 mg, 2.31 mmol) and triethyl orthoformate (9.81 g, 66.21 mmol) was reacted at 110° C. for 5 hours. The reaction solution was concentrated to obtain the crude compound 5-2.
  • Step B To a mixed solution of 20 mL of dioxane and 4 mL of water was added compound 5-2 (550 mg, 2.43 mmol), compound 3-5 (749.53 mg, 4.87 mmol), potassium phosphate (1.29 g, 6.08 mmol) and Pd(PPh 3 ) 4 (281.18 mg, 243.33 ⁇ mol), the reaction system was replaced with nitrogen three times, and the reaction was carried out at 85° C. under nitrogen protection for 12 hours. The reaction solution was concentrated to obtain the crude compound 5-3. MS (ESI) m/z: 174.2 [M+H] + .
  • Step C To compound 5-3 (2.39 g, 13.80 mmol), N,N-dimethylaniline (2.51 g, 20.70 mmol) and benzyltriethylammonium chloride (6.29 g, 27.60 mmol) in 25 mL of acetonitrile Phosphorus oxychloride (12.70 g, 82.81 mmol) was added to the solution, and the reaction system was reacted at 75 ° C for 2 hours.
  • Step D To a solution of compound 5-4 (120 mg, 626.25 ⁇ mol) and compound 1-12 (124.03 mg, 626.25 ⁇ mol) in 5 mL of isopropanol was added methanesulfonic acid (120.38 mg, 1.25 mmol) at 80° C. The reaction was continued for 2 hours, then 20 mL of sodium bicarbonate solution was added and extracted with ethyl acetate (50 mL ⁇ 3), the organic phases were combined and washed with saturated brine (30 mL ⁇ 2), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude compound 5-5.
  • Step E To a mixed solution of compound 5-5 (227 mg, 6442.67 ⁇ mol) in tetrahydrofuran (25 mL) and water (5 mL) was added potassium osmate dihydrate (2.37 mg, 6.43 ⁇ mol) and sodium periodate (687.30 mg, 3.21 mmol), the reaction system was quenched with 20 mL of 50% sodium thiosulfate solution after 12 hours of reaction at 25 °C, and then extracted with dichloromethane (60 mL ⁇ 3), and the organic phases were combined and washed with saturated brine (30 mL). ⁇ 2), dried over anhydrous sodium sulfate, filtered and concentrated to obtain crude product.
  • Step F A solution of compound 5-6 (90 mg, 253.39 ⁇ mol) and compound 5-7 (58.34 mg, 506.77 ⁇ mol) in 6 mL of dichloromethane was stirred at 25° C. for 0.5 h, and NaBH(OAc) 3 (161.11 mg, 760.16 ⁇ mol). The reaction system was stirred at 25°C for 12 hours, 10 mL of water was added, and extracted with dichloromethane (50 mL ⁇ 5); the organic phase was washed with saturated brine (20 mL ⁇ 1), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude compound 5-8.
  • Step G Compound 5-8 (120 mg, 264.13 ⁇ mol), Compound 1-5 (128.30 mg, 343.37 ⁇ mol), sodium carbonate (69.99 mg, 660.33 ⁇ mol) and Pd(PPh 3 ) 4 (30.52 mg, 26.41 ⁇ mol)
  • a mixed solution of dioxane (10 mL) and water (2 mL) was replaced with nitrogen three times and heated to 100° C. under nitrogen protection and stirred for 1 hour.
  • the reaction solution was filtered through celite, the filter cake was washed with 100 mL of dichloromethane, the filtrates were combined and concentrated to obtain the crude product.
  • Step H A solution of compound 5-9 (45 mg, 72.45 ⁇ mol) and compound 1-15 (16.54 mg, 144.91 ⁇ mol) in 2 mL dichloromethane was stirred at 25 °C for 0.5 h, and NaBH(OAc)3 (76.78 mg) was added thereto. , 362.27 ⁇ mol).
  • the reaction system was stirred at 25°C for 12 hours, diluted with 10 mL of water, and extracted with dichloromethane (20 mL ⁇ 5). The organic phase was washed with saturated brine (20 mL ⁇ 1), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product .
  • Step A Compound 6-1 (8 g, 35.09 mmol) was added to 40 mL of dioxane, then 20 mL of water and ammonia water (18.01 g, 143.86 mmol, 28% purity) were added, and the reaction system was stirred at 25° C. for 10 minutes Sulfur powder (19.98 g, 114.74 mmol) was added in batches, and the reaction was continued at 25° C. for 5 hours. The reaction solution was filtered through celite, and the filter cake was washed with 100 mL of ethyl acetate. The filtrate was diluted with 20 mL of water and extracted with ethyl acetate (40 mL ⁇ 5).
  • Step B A mixed solution of compound 5-1 (450 mg, 2.08 mmol) and triethyl orthoacetate (7.96 g, 49.10 mmol) was reacted at 120° C. for 5 hours. The reaction solution was concentrated to obtain the crude compound 6-2. MS(ESI) m/z: 240.2 [M+H] + .
  • Step C To a mixed solution of 15 mL of dioxane and 3 mL of water was added compound 6-2 (690 mg, 2.87 mmol), compound 3-5 (885.37 mg, 5.75 mmol), potassium phosphate (1.53 g, 7.19 mmol) and Pd(PPh 3 ) 4 (332.15 mg, 287.43 ⁇ mol), the reaction system was replaced with nitrogen three times, and the reaction was carried out at 95° C. under nitrogen protection for 12 hours. The reaction solution was concentrated to obtain crude compound 6-3. MS (ESI) m/z: 188.3 [M+H] + .
  • Step D To compound 6-3 (2.2 g, 11.75 mmol), N,N-dimethylaniline (2.14 g, 17.63 mmol) and benzyltriethylammonium chloride (5.35 g, 23.50 mmol) in 40 mL of acetonitrile Phosphorus oxychloride (10.81 g, 70.51 mmol) was added to the solution, and the reaction system was reacted at 75 ° C for 2 hours.
  • Step E To a solution of compound 6-4 (230 mg, 1.12 mmol) and compound 1-12 (221.52 mg, 1.12 mmol) in 10 mL of isopropanol was added methanesulfonic acid (214.98 mg, 2.24 mmol), the system was heated at 80°C The reaction was continued for 2 hours, then 30 mL of sodium bicarbonate solution was added and extracted with ethyl acetate (50 mL ⁇ 3), the organic phases were combined, washed with saturated brine (30 mL ⁇ 2), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • methanesulfonic acid 214.98 mg, 2.24 mmol
  • Step F To a mixed solution of compound 6-5 (238 mg, 648.07 ⁇ mol) in tetrahydrofuran (25 mL) and water (5 mL) was added potassium osmate dihydrate (2.39 mg, 6.48 ⁇ mol) and sodium periodate (693.08 mg, 3.24 mmol), the reaction system was quenched with 30 mL of 50% sodium thiosulfate solution after reacting for 5 hours at 25 °C, and then extracted with dichloromethane (80 mL ⁇ 3), and the organic phases were combined and washed with saturated brine (30 mL). ⁇ 2), dried over anhydrous sodium sulfate, filtered and concentrated to obtain crude compound 6-6.
  • Step G A solution of compound 6-6 (330 mg, 893.79 ⁇ mol) and compound 5-7 (205.80 mg, 1.79 mmol) in 15 mL of dichloromethane was stirred at 25° C. for 0.5 h, and NaBH(OAc) 3 (568.29 mg, 2.68 mmol). The reaction system was stirred at 25°C for 12 hours, 10 mL of water was added, and extracted with dichloromethane (50 mL ⁇ 5); the organic phase was washed with saturated brine (20 mL ⁇ 1), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude compound 6-7.
  • Step H compound 6-7 (657 mg, 1.40 mmol), compound 1-5 (681.39 mg, 1.82 mmol), sodium carbonate (371.71 mg, 3.51 mmol), and Pd(PPh 3 ) 4 (162.10 mg, 140.28 ⁇ mol)
  • a mixed solution of dioxane (15 mL) and water (3 mL) was replaced with nitrogen three times and heated to 100° C. under nitrogen protection and stirred for 1 hour.
  • the reaction solution was filtered through celite, the filter cake was washed with 150 mL of dichloromethane, the filtrates were combined and concentrated to obtain the crude product.
  • Step I A solution of compound 6-8 (350 mg, 551.09 ⁇ mol) and compound 1-15 (125.81 mg, 1.10 mmol) in 10 mL of dichloromethane was stirred at 25° C. for 0.5 h, and NaBH(OAc) 3 (583.99 mg) was added thereto. , 2.76 mmol).
  • the reaction system was stirred at 25°C for 12 hours, diluted with 10 mL of water, and extracted with dichloromethane (20 mL ⁇ 5). The organic phase was washed with saturated brine (20 mL ⁇ 1), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product .
  • Step A Boron tribromide (21.90 g, 87.42 mmol) was slowly added dropwise to a solution of compound 1-3 (5 g, 29.14 mmol) in 60 mL of dichloromethane at 0 °C, the mixture was reacted at 0 °C for 30 minutes
  • Step B Silver carbonate (6.16 g, 22.34 mmol) was added to a solution of compound 7-1 (3.2 g, 20.31 mmol) and bromoacetonitrile (4.87 g, 40.62 mmol) in 40 mL of acetonitrile, and the reaction system was stirred at 90° C.
  • reaction solution was filtered through celite, the filter cake was washed with 30 mL of ethyl acetate, then 20 mL of water was added to the filtrate to dilute and then extracted with ethyl acetate (30 mL ⁇ 5), the organic phases were combined and washed with saturated brine (20 mL ⁇ 1), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step C compound 7-2 (0.496 g, 2.52 mmol), compound 1-2 (652.95 mg, 2.78 mmol), potassium carbonate (697.41 mg, 5.05 mmol), and Pd(PPh 3 ) 4 (291.55 mg, 252.30 ⁇ mol) ) in dioxane (10 mL) and water (2 mL) was purged three times with nitrogen and heated to 85°C under nitrogen protection and stirred for 5 hours. The reaction solution was diluted with 20 mL of water and extracted with ethyl acetate (50 mL ⁇ 3). The organic phases were combined, washed with saturated brine (30 mL ⁇ 2), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step D Compound 7-3 (630 mg, 1.79 mmol), bisphosphonate boronate (910.07 mg, 3.58 mmol), potassium acetate (351.72 mg, 3.58 mmol) and Pd(dppf)Cl 2 ⁇ CH 2 Cl
  • a solution of 2 (146.33 mg, 179.19 ⁇ mol) in 20 mL of dioxane was purged three times with nitrogen and heated to 85° C. under nitrogen and stirred for 5 hours.
  • the reaction solution was filtered through celite, the filter cake was washed with 100 mL of ethyl acetate, the filtrates were combined and concentrated to obtain the crude product.
  • Step E Compound 7-4 (100.43 mg, 251.93 ⁇ mol), Compound 3-9 (100 mg, 209.94 ⁇ mol), sodium carbonate (55.63 mg, 524.86 ⁇ mol) and Pd( PPh3 ) 4 (24.26 mg, 20.99 ⁇ mol)
  • a mixed solution of dioxane (10 mL) and water (2 mL) was replaced with nitrogen three times and heated to 100° C. under nitrogen protection and stirred for 1 hour.
  • reaction solution was filtered through celite, diluted with 20 mL of water, extracted with ethyl acetate (30 mL ⁇ 3), washed with saturated brine (30 mL ⁇ 2) after combining the organic phases, dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product .
  • MS (ESI) m/z: 668.4 [M+H] + .
  • Step F A solution of compound 7-5 (130 mg, 194.58 ⁇ mol) and compound 1-15 (44.42 mg, 389.17 ⁇ mol) in 6 mL dichloromethane was stirred at 25 °C for 0.5 h, and NaBH(OAc) 3 (206.20 mg) was added thereto. , 972.92 ⁇ mol).
  • the reaction system was stirred at 25°C for 1 hour, diluted with 10 mL of water, and then extracted with dichloromethane (20 mL ⁇ 5). The organic phase was washed with saturated brine (20 mL ⁇ 1), dried over anhydrous sodium sulfate, filtered and concentrated to obtain Crude.
  • Step A A solution of compound 3-8 (200 mg, 493.59 ⁇ mol) and compound 5-7 (113.65 mg, 987.18 ⁇ mol) in 15 mL dichloromethane was stirred at 25° C. for 0.5 h, and NaBH(OAc) 3 (313.83 mg) was added thereto. , 1.48 mmol). The reaction system was stirred at 25°C for 12 hours, diluted with 10 mL of water, and then extracted with dichloromethane (40 mL ⁇ 8). The organic phase was washed with saturated brine (20 mL ⁇ 1), dried over anhydrous sodium sulfate, filtered and concentrated to obtain Crude compound 8-1. MS (ESI) m/z: 504.3 [M+H] + .
  • Step B Compound 8-1 (150 mg, 297.43 ⁇ mol), Compound 7-4 (118.57 mg, 297.43 ⁇ mol), sodium carbonate (78.81 mg, 743.57 ⁇ mol) and Pd(PPh 3 ) 4 (34.37 mg, 29.74 ⁇ mol)
  • a mixed solution of dioxane (15 mL) and water (3 mL) was replaced with nitrogen three times and heated to 100° C. under nitrogen protection and stirred for 1 hour.
  • the reaction solution was filtered through celite, the filter cake was washed with 100 mL of ethyl acetate, the filtrates were combined and concentrated to obtain the crude product.
  • Step C A solution of compound 8-2 (80 mg, 114.93 ⁇ mol) and compound 1-15 (26.24 mg, 229.85 ⁇ mol) in 10 mL of dichloromethane was stirred at 25° C. for 0.5 h, and NaBH(OAc) 3 (121.79 mg) was added thereto. , 574.63 ⁇ mol). The reaction system was stirred at 25°C for 1 hour, diluted with 10 mL of water, and then extracted with dichloromethane (20 mL ⁇ 5). The organic phase was washed with saturated brine (20 mL ⁇ 1), dried over anhydrous sodium sulfate, filtered and concentrated to obtain Crude.
  • Step A A solution of compound 1-5 (0.5 g, 1.34 mmol) and compound 1-15 (305.50 mg, 1.34 mmol) in 10 mL of dichloromethane was stirred at 25 °C for 0.5 h, and NaBH(OAc) 3 (1.42 g, 6.69 mmol). The reaction system was stirred at 25°C for 2 hours, diluted with 20 mL of water, extracted with dichloromethane (30 mL ⁇ 5), the organic phase was washed with saturated brine (20 mL ⁇ 1), dried over anhydrous sodium sulfate, filtered and concentrated to obtain Crude compound 9-1. MS (ESI) m/z: 472.4 [M+H] + .
  • Step B To a solution of compound 9-1 (200 mg, 423.92 ⁇ mol) in 5 mL of dichloromethane was added di-tert-butyl dicarbonate (138.78 mg, 635.89 ⁇ mol) and triethylamine (85.79 mg, 847.85 ⁇ mol), the reaction system was at After stirring at 25°C for 2 hours, 10 mL of water was added to dilute, and then extracted with dichloromethane (20 mL ⁇ 3). The organic phase was washed with saturated brine (20 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product. The crude product was purified by preparative thin layer chromatography on silica gel plate separation (ethyl acetate) to give compound 9-2. MS (ESI) m/z: 572.2 [M+H] + .
  • Step C Compound 9-2 (230 mg, 402.17 ⁇ mol), Compound 3-8 (162.96 mg, 402.17 ⁇ mol), sodium carbonate (106.56 mg, 1.01 mmol) and Pd(PPh 3 ) 4 (46.47 mg, 40.22 ⁇ mol)
  • a mixed solution of dioxane (5 mL) and water (1 mL) was replaced with nitrogen three times and heated to 100° C. under nitrogen protection and stirred for 1 hour.
  • the reaction solution was diluted with 20 mL of water, filtered through celite, and then extracted with ethyl acetate (40 mL ⁇ 3).
  • Step D To a solution of compound 9-3 (30 mg, 38.95 ⁇ mol) and compound 5-7 (8.97 mg, 77.90 ⁇ mol) in dichloromethane (2 mL) was added NaBH(OAc) 3 (41.28 mg, 194.75 ⁇ mol). The reactant was stirred at 25°C for 1 hour, diluted with 50 mL of water, and extracted with dichloromethane (50 mL ⁇ 2); the organic phase was washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated to obtain crude product 9-4 . The crude product was used directly in the next step. MS (ESI) m/z: 869.3 [M+H] + .
  • Step E Trifluoroacetic acid (196.74 mg, 1.73 mmol) was added to a solution of compound 9-4 (30 mg, 34.51 ⁇ mol) in dichloromethane (2 mL), the reaction solution was stirred at 25° C. for 1 hour, and concentrated to obtain the crude product.
  • the crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex luna C18 150*25mm*10 ⁇ m; mobile phase: [pure water (0.225% formic acid)-acetonitrile]; acetonitrile%: 10%-40%, 10 minutes) and purified Compound 9 is obtained.
  • Step A Compound 4-1 (1 g, 1.91 mmol), Compound 2-6 (758.01 mg, 1.91 mmol), sodium carbonate (607.53 mg, 5.73 mmol) and Pd(dppf)Cl 2 (156.03 mg, 191.07 ⁇ mol)
  • a mixed solution of tetrahydrofuran (32 mL) and water (8 mL) was replaced with nitrogen three times and heated to 50°C under nitrogen protection, and stirred for 12 hours.
  • the reaction solution was diluted with 20 mL of water, extracted with ethyl acetate (50 mL ⁇ 3), the organic phase was washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • MS(ESI) m/z: 644.0 [M+
  • Step B Compound 10-3 (40 mg, 62.11 ⁇ mol) and Compound 5-7 (21.45 mg, 186.33 ⁇ mol) and A solution of molecular sieves (40 mg) in 5 mL of dichloromethane was stirred for 0.5 h, to which was added NaBH(OAc) 3 (65.81 mg, 310.55 ⁇ mol).
  • Step A Compound 10-3 (40 mg, 62.11 ⁇ mol) and Compound 11-1 (24.07 mg, 186.33 ⁇ mol) and A solution of molecular sieves (40 mg) in 5 mL of dichloromethane was stirred for 0.5 h, to which was added NaBH(OAc) 3 (65.81 mg, 310.55 ⁇ mol).
  • Step A Compound 10-3 (40 mg, 62.11 ⁇ mol) and Compound 1-10 (16.23 mg, 186.33 ⁇ mol) and A solution of molecular sieves (40 mg) in 5 mL of dichloromethane was stirred for 0.5 h, to which was added NaBH(OAc) 3 (65.81 mg, 310.55 ⁇ mol).
  • Step A Compound 10-3 (60 mg, 93.16 ⁇ mol) and Compound 13-1 (36.66 mg, 279.47 ⁇ mol) and A solution of molecular sieves (60 mg) in 8 mL of dichloromethane was stirred for 0.5 h, to which was added NaBH(OAc) 3 (98.72 mg, 465.79 ⁇ mol).
  • reaction system was stirred at 15 ° C for 1.5 hours and then added 2 mL of water, filtered, and the filtrate was concentrated to obtain a crude product, which was separated using preparative-grade high performance liquid chromatography (chromatographic column: YMC Triart 30*150mm*7 ⁇ m; mobile phase: [pure water (hydrochloric acid)] -acetonitrile]; % acetonitrile: 31%-51%, 7 min) purification gave compound 13 as the hydrochloride salt.
  • Step A Compound 10-3 (100 mg, 155.26 ⁇ mol) and Compound 14-1 (54.57 mg, 465.79 ⁇ mol) and A solution of molecular sieves (100 mg) in 8 mL of dichloromethane was stirred for 0.5 h, to which was added NaBH(OAc) 3 (164.53 mg, 776.31 ⁇ mol).
  • Step A A solution of compound 10-3 (50.00 mg, 77.63 ⁇ mol), compound 15-1 (20.05 mg, 155.26 ⁇ mol) and molecular sieves (0.05 g) in dichloromethane (10 mL) was stirred at 25° C. for 0.5 h, and NaBH was added to it (OAc) 3 (49.36 mg, 232.89 ⁇ mol). The reactant was stirred at 25°C for 1 hour, diluted with 20 mL of water, and extracted with dichloromethane (30 mL ⁇ 3); the organic phase was washed with saturated brine (30 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step A A solution of compound 3-10 (60 mg, 93.30 ⁇ mol) and compound 11-1 (24.10 mg, 186.60 ⁇ mol) in dichloromethane (5 mL) was stirred at 25° C. for 0.5 h, and NaBH(OAc) 3 (98.87 mg) was added thereto. , 466.50 ⁇ mol). The reactant was stirred at 25°C for 1 hour, diluted with 10 mL of water, and extracted with dichloromethane (20 mL ⁇ 3); the organic phase was washed with saturated brine (20 mL ⁇ 2), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step A A solution of compound 7-5 (0.1 g, 149.68 ⁇ mol) and compound 5-7 (34.47 mg, 299.36 ⁇ mol) in 10 mL of dichloromethane was stirred at 25 °C for 0.5 h, and NaBH(OAc) 3 (158.62 mg, 748.40 ⁇ mol). The reaction system was stirred at 25°C for 12 hours, diluted with 10 mL of water, and then extracted with dichloromethane (20 mL ⁇ 5). The organic phase was washed with saturated brine (20 mL ⁇ 1), dried over anhydrous sodium sulfate, filtered and concentrated to obtain Crude.
  • Step A To a solution of compound 3-8 (150 mg, 370.19 ⁇ mol) and compound 18-1 (74.89 mg, 740.38 ⁇ mol) in dichloromethane (20 mL) was added Molecular sieves (150 mg), stirred at 25° C. for 30 minutes, NaBH(OAc) 3 (235.38 mg, 1.11 mmol) was added thereto, the reaction solution was stirred at 25° C. for 1 hour, filtered through celite, 20 mL of water was added, and 2 Extracted with methyl chloride (40 mL ⁇ 3), combined the organic phases, washed with saturated brine (30 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain compound 18-2.
  • Step B To 10 mL of dioxane solution was added compound 18-2 (0.17 g, 346.70 ⁇ mol), bisphosphonate boronate (132.06 mg, 520.04 ⁇ mol), potassium acetate (85.06 mg, 866.74 ⁇ mol) and Pd(dppf)Cl 2 ⁇ CH 2 Cl 2 (28.31 mg, 34.67 ⁇ mol), the reaction system was replaced with nitrogen three times, and the reaction was carried out at 100° C. under nitrogen protection for 1 hour. The reaction solution was cooled to room temperature and filtered through celite. The filter cake was rinsed with dichloromethane (100 ml), and the organic phases were combined and concentrated to obtain the crude product.
  • Step C To a mixed solution of 5 mL of dioxane and 1 mL of water, compound 18-3 (65 mg, 120.95 ⁇ mol), compound 2-8 (75.56 mg, 127.00 ⁇ mol), sodium carbonate (32.05 mg, 302.38 ⁇ mol) and Pd(PPh 3 ) 4 (13.98 mg, 12.10 ⁇ mol), the reaction system was replaced with nitrogen three times, and the reaction was carried out at 100° C. under nitrogen protection for 1 hour.
  • Step D To a solution of compound 18-4 (0.1 g, 116.77 ⁇ mol) in dichloromethane (3 mL) was added trifluoroacetic acid (1.54 g, 13.51 mmol), and the reaction solution was stirred at 25° C. for 12 hours. The reaction solution was concentrated to obtain a crude product, and the crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex Synergi C18 150*25mm*10 ⁇ m; mobile phase: [pure water (0.225% formic acid)-acetonitrile]; acetonitrile%: 8% -38%, 10 min) purification gave compound 18.
  • Step A Compound 19-1 (5 g, 26.87 mmol) was dissolved in methanol (30 mL), hydrochloric acid (12 M, 6.72 mL) and water (15 mL) were added, and the temperature was lowered. At 0 °C, sodium nitrite (2.23 g, 32.25 mmol) was added. ,) was dissolved in water (15 mL) and added dropwise to the reaction system, the mixture was stirred at 0° C. for 0.5 hours, and then bicarbonate (20.47 g, 80.62 mmol) was dissolved in methanol (30 mL) and added, and the mixture was heated at 25° C. Stir for 1 hour.
  • reaction solution was quenched by adding 50 mL of water, then extracted with ethyl acetate (100 mL ⁇ 3), the organic phases were combined, washed with saturated brine (50 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step B Compound 19-2 (5.79 g, 19.50 mmol) and compound 19-3 (3.84 g, 18.57 mmol), sodium carbonate (3.94 g, 37.13 mmol) and Pd (dppf) were added to 50 mL of dioxane and 10 mL of water ) Cl 2 ⁇ CH 2 Cl 2 (1.52 g, 1.86 mmol), the reaction system was replaced with nitrogen three times, and the reaction was carried out at 50° C. under nitrogen protection for 12 hours.
  • Step C To 40 mL of methanol was added compound 19-4 (3.99 g, 11.69 mmol), sodium methoxide (4.21 g, 23.38 mmol, 4.68 mL, 30% purity). The mixture was stirred at 50°C for 12 hours. Then 8 mL of water was added and the mixture was stirred at 50°C for 2 hours. The reaction solution was cooled to room temperature, 30 mL of water was added, the pH was adjusted to 5 with 12M HCl, filtered, and the filter cake was spin-dried to obtain compound 19-5. MS (ESI) m/z: 322.7 [M+H + ].
  • Step E Compound 19-6 (1.39 g, 4.12 mmol) was added to 25 mL of tetrahydrofuran, diisobutylaluminum hydride (1 M, 10.31 mL) was added dropwise at -78 °C, and the mixture was stirred at 0 °C for 1 hour.
  • the reaction solution was warmed to room temperature, 50 mL of water was added, then extracted with ethyl acetate (50 mL ⁇ 3), the organic phases were combined, washed with saturated brine (30 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step F To 25 mL of dichloromethane was added compound 19-7 (1.33 g, 4.30 mmol), triethylamine (870.64 mg, 8.60 mmol, 1.20 mL). The temperature was lowered, and methanesulfonyl chloride (1.59 g, 13.88 mmol, 1.07 mL) was added at 0°C. The mixture was stirred at 0°C for 1 hour. The reaction solution was warmed to room temperature and added with 40 mL of water, then extracted with dichloromethane (40 mL ⁇ 3), combined with the organic phases, washed with saturated brine (30 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • dichloromethane 40 mL ⁇ 3
  • Step G To 25 mL of N,N-dimethylformamide was added compound 19-8 (0.2 g, 516.46 ⁇ mol), compound 19-9 (89.81 mg, 542.29 ⁇ mol), triethylamine (209.04 mg, 2.07 mmol) , the mixture was stirred at 25°C for 12 hours. The reaction mixture was added with 20 mL of water, then extracted with ethyl acetate (30 mL ⁇ 3), the organic phases were combined, washed with saturated brine (30 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step H compound 19-10 (52.20 mg, 124.19 ⁇ mol), compound 4-1 (65 mg, 124.19 ⁇ mol), sodium carbonate (32.91 mg, 310.48 ⁇ mol), and Pd(PPh 3 ) 4 (14.35 mg, 12.42 ⁇ mol)
  • a solution of dioxane (5 mL) and water (1 mL) was heated to 100 °C under nitrogen and stirred for 1 hour.
  • the reaction solution was diluted with 20 mL of water, filtered through celite, extracted with dichloromethane (20 mL ⁇ 3), the organic phases were combined, washed with saturated brine (30 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain Crude.
  • Step B Compound 20-2 (9.79 mg, 114.64 ⁇ mol), Compound 4-1 (60 mg, 114.64 ⁇ mol), sodium carbonate (30.38 mg, 286.60 ⁇ mol), and Pd(PPh 3 ) 4 (13.25 mg, 11.46 ⁇ mol)
  • the mixed solution of dioxane (5 mL) and water (1 mL) was replaced with nitrogen three times, heated to 100° C. under nitrogen protection, and stirred for 1 hour.
  • the reaction solution was cooled to room temperature and added with 20 mL, then extracted with dichloromethane (20 mL ⁇ 3), the organic phases were combined, washed with saturated brine (20 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • MS(ESI) m/z: 751.2[M+H] + .
  • Step C To a solution of compound 20-3 (0.07 g, 93.23 ⁇ mol) in methanol (2 mL) and water (0.4 mL) was added lithium hydroxide monohydrate (7.82 mg, 186.46 ⁇ mol), and the reaction solution was stirred at 50°C 1 hour. After the reaction was cooled to room temperature, it was concentrated. It was then dissolved in 10 mL of water, adjusted to pH 5 with 12M HCl, and concentrated to give the crude product.
  • Step A Compound 3-8 (0.6 g, 1.48 mmol), Compound 21-1 (365.99 mg, 2.96 mmol), acetic acid (444.61 mg, 7.40 mmol) and A solution of molecular sieves (0.6 g) in 15 mL of dichloromethane was stirred at 25 °C for 0.5 h, and NaBH(OAc) 3 (941.50 mg, 4.44 mmol) was added thereto.
  • Step B Compound 21-2 (559.78 mg, 2.20 mmol), double pinacol boronate (0.7 g, 1.47 mmol), potassium acetate (360.57 mg, 3.67 mmol), and Pd(dppf)Cl 2 ⁇ CH
  • 2 Cl 2 120.01 mg, 149.96 ⁇ mol
  • the reaction solution was filtered through celite, the filter cake was washed with 100 mL of dichloromethane, and the filtrate was concentrated to obtain a crude product.
  • Step C Compound 21-3 (200 mg, 382.13 ⁇ mol), Compound 20-2 (165.97 mg, 382.13 ⁇ mol), sodium carbonate (101.26 mg, 955.33 ⁇ mol) and Pd(PPh 3 ) 4 (44.16 mg, 38.21 ⁇ mol)
  • a mixed solution of dioxane (5 mL) and water (1 mL) was replaced with nitrogen three times and heated to 100° C. under nitrogen protection and stirred for 1 hour.
  • the reaction solution was diluted with 20 mL of water and extracted with dichloromethane (40 mL ⁇ 3).
  • the organic phases were combined, washed with saturated brine (20 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step D Lithium hydroxide monohydrate (13.41 mg, 319.64 ⁇ mol) was added to a mixed solution of compound 21-4 (120 mg, 159.82 ⁇ mol) in methanol (5 mL) and water (1 mL), and the reaction system was stirred at 50° C. for 0.5 Concentrate after 1 hour, then add 10 mL of water to dilute, adjust pH to 5 with concentrated hydrochloric acid, and then concentrate to obtain crude product.
  • Step A To 10 mL of the dioxane solution was added compound 3-8 (0.5 g, 1.23 mmol), bisphosphonate boronate (470.03 mg, 1.85 mmol), potassium acetate (363.31 mg, 3.70 mmol) and Pd(dppf)Cl 2 ⁇ CH 2 Cl 2 (100.77 mg, 123.40 ⁇ mol), the reaction system was replaced with nitrogen three times, and the reaction was carried out at 100° C. under nitrogen protection for 2 hours.
  • compound 3-8 0.5 g, 1.23 mmol
  • bisphosphonate boronate 470.03 mg, 1.85 mmol
  • potassium acetate 363.31 mg, 3.70 mmol
  • Pd(dppf)Cl 2 ⁇ CH 2 Cl 2 100.77 mg, 123.40 ⁇ mol
  • Step B Compound 22-2 (2 g, 17.08 mmol) was dissolved in tetrahydrofuran (20 mL), triphenylphosphine (4.93 g, 18.79 mmol) and DIAD (3.80 g, 18.79 mmol) were added. The mixture was stirred at 25°C for 6 hours.
  • Step C To a solution of compound 22-3 (2.8 g, 10.94 mmol) in ethanol (30 mL) was added hydrazine hydrate (2.54 g, 49.72 mmol). The mixture was stirred at 50 °C for 0.5 h, and the temperature was raised to 75 °C and continued to stir for 2 Hour. A white solid was precipitated, filtered, the filtrate was concentrated and then ethanol (50 mL) was added, a white solid was precipitated, continued filtration, and the filtrate was concentrated to obtain compound 22-4.
  • Step D To a solution of compound 2-6 (256.24 mg, 645.89 ⁇ mol) in methanol (10 mL) was added NaBH(OAc) 3 (405.89 mg, 6.46 mmol) and compound 22-4 (150 mg, 1.29 mmol). The mixture was stirred at 25°C for 5 hours. Water (10 mL) was added at 0°C to quench the reaction, extracted with dichloromethane (15 mL ⁇ 3), the organic phases were combined and washed with saturated brine (15 mL ⁇ 2), dried over anhydrous sodium sulfate, filtered and concentrated to obtain compound 22- 5.
  • Step F To a mixed solution of 8 mL of tetrahydrofuran and 2 mL of water, compound 22-1 (318.19 mg, 703.56 ⁇ mol), compound 22-6 (350 mg, 586.30 ⁇ mol), sodium carbonate (155.36 mg, 1.47 mmol) and Pd (dppf) were added ) Cl 2 ⁇ CH 2 Cl 2 (47.88 mg, 58.63 ⁇ mol), the reaction system was replaced with nitrogen three times, and the reaction was carried out at 50° C. under nitrogen protection for 12 hours.
  • Step H To a solution of compound 22-9 (11 mg, 12.79 ⁇ mol) in dichloromethane (3 mL) was added trifluoroacetic acid (1.13 g, 9.90 mmol), and the reaction solution was stirred at 25° C. for 20 minutes. The reaction solution was concentrated to obtain a crude product, and the crude product was separated by preparative high performance liquid chromatography (chromatographic column: Phenomenex luna C18 150*50mm*10 ⁇ m; mobile phase: [pure water (0.225% formic acid)-acetonitrile]; acetonitrile%: 10% -40%, 10 min) purification afforded compound 22 as the formate salt.
  • Step A Dissolve compound 23-1 (3.5 g, 18.81 mmol) in methanol (40 mL), add an aqueous solution of HCl (12 M, 4.70 mL) (20 mL); stir at 0°C, add sodium nitrite (1.56 g) , 22.57 mmol) in water (10 mL). The reaction was stirred at 0°C for 30 minutes and a solution of bispinacol boronate (14.33 g, 56.44 mmol) in methanol (40 mL) was slowly added to the reaction. The reaction mixture was stirred at 20° C. for 1 hour.
  • reaction mixture was diluted with 100 mL of water, extracted with ethyl acetate (100 mL ⁇ 3), the organic phase was washed with saturated brine, dried over sodium sulfate, filtered and concentrated to obtain the crude product.
  • Step B Compound 23-2 (1.5 g, 8.74 mmol), Compound 1-3 (2.86 g, 9.62 mmol), Pd(PPh 3 ) 4 (1.01 g, 874.22 ⁇ mol) and potassium carbonate (2.42 g, 17.48 mmol)
  • the mixture was mixed with a solution of dioxane (30 mL) and water (6 mL), replaced with nitrogen, heated to 90°C, and stirred for 10 hours.
  • the reaction mixture was diluted with dichloromethane (50 mL), filtered through celite, and the filtrate was spin-dried to obtain the crude product.
  • Step C Compound 23-3 (1.4 g, 4.57 mmol), bispinacol boronate (2.32 g, 9.15 mmol) dichloromethane. bis(triphenylphosphine) palladium dichloride (373.44 mg, 457 ⁇ mol) and potassium acetate (897 mg, 9.15 mmol) in dioxane were replaced with nitrogen and then heated to 85° C. and stirred under nitrogen protection for 5 hours. The reaction mixture was diluted with dichloromethane (50 mL), filtered through celite, and the filtrate was spin-dried to obtain the crude product.
  • Step E To a solution of compound 23-5 (100 mg, 160.60 ⁇ mol) and compound 23-6 (82.97 mg, 642.40 ⁇ mol) in dichloromethane (10 mL) was added Molecular sieves (20mg), the reaction solution was stirred at 25°C for 30 minutes, NaBH(OAc) 3 (136.15mg, 642.40 ⁇ mol) was added into it, the reaction solution was stirred at 25°C for 12 hours, the reaction solution was filtered through celite, and the The filtrate was concentrated to give crude product.
  • Step A To a solution of compound 23-5 (100 mg, 160.60 ⁇ mol) and compound 24-1 (82.97 mg, 642.40 ⁇ mol) in dichloromethane (10 mL) was added Molecular sieves (20mg), the reaction solution was stirred at 25°C for 30 minutes, NaBH(OAc) 3 (136.15mg, 642.40 ⁇ mol) was added into it, the reaction solution was stirred at 25°C for 12 hours, the reaction solution was filtered through celite, and the The filtrate was concentrated to give crude product.
  • Step A Compound 10-3 (40 mg, 62.11 ⁇ mol) and Compound 25-1 (27.05 mg, 186.33 ⁇ mol) and A solution of molecular sieves (40 mg) in 5 mL of dichloromethane was stirred for 0.5 h, to which was added NaBH(OAc) 3 (65.81 mg, 310.55 ⁇ mol).
  • Step A Compound 10-3 (40 mg, 62.11 ⁇ mol) and Compound 26-1 (27.05 mg, 186.33 ⁇ mol) and A solution of molecular sieves (40 mg) in 5 mL of dichloromethane was stirred for 0.5 h, to which was added NaBH(OAc) 3 (65.81 mg, 310.55 ⁇ mol).
  • Step H 20 mL of acetic acid solution of nitric acid (9.16 g, 145.30 mmol) was added dropwise to a solution of compound 27-1 (10 g, 53.59 mmol) in 20 mL of acetic acid at 0 °C, the reaction system was stirred at 25 °C for 2 hours, and then 50 mL of water was added Diluted, filtered, and the filter cake was washed with 50 mL of water and dried to obtain crude compound 27-2.
  • Step 1 add hydrosulfite (13.53g, 77.72mmol) to the mixed solution of compound 27-2 (3g, 12.95mmol) in 30mL of methanol and 6mL of water, the reaction system was stirred at 70 ° C for 12 hours, filtered, and the filter cake was Wash with 100 mL methanol, and concentrate the filtrate to obtain crude product.
  • Step J Compound 27-3 (4.2 g, 20.83 mmol) and compound 27-4 (3.94 g, 19.79 mmol) in 100 mL of ethanol were stirred at 25 °C for 1 hour, concentrated, and then dissolved in 100 mL of dichloromethane solution, Dichlorodicyanobenzoquinone (4.49 g, 19.79 mmol) was added, the reaction system was stirred at 25°C for 12 hours, then 100 mL of saturated sodium sulfite solution was added to quench, and then extracted with dichloromethane (80 mL ⁇ 3), and the organic phases were combined.
  • Step L compound 4-1 (2.5 g, 4.78 mmol), compound 27-6 (1.68 g, 4.78 mmol), sodium carbonate (1.27 g, 11.94 mmol) and Pd(PPh 3 ) 4 (551.97 mg, 477.66 ⁇ mol)
  • a mixed solution of dioxane (15 mL) and water (3 mL) was replaced with nitrogen three times and heated to 100° C. under nitrogen protection and stirred for 12 hours.
  • the reaction solution was diluted with 30 mL of water and filtered through celite.
  • Step M Compound 27-7 (2.7 g, 4.04 mmol), potassium ferrocyanide (1.70 g, 4.04 mmol), potassium acetate (79.20 mg, 807.03 ⁇ mol) and t-BuXPhos-Pd-G3 (320.54 mg, 403.52 ⁇ mol) of a mixed solution of dioxane (30 mL) and water (30 mL) was replaced with nitrogen three times and heated to 100° C. under nitrogen protection and stirred for 3 hours. The reaction solution was diluted with 20 mL of water and filtered through celite.
  • Step N Manganese dioxide (6.33 g, 72.76 mmol) was added to a solution of compound 27-8 (2.4 g, 3.64 mmol) in 40 mL of dichloromethane, the reaction system was stirred at 50° C. for 12 hours, and the reaction solution was passed through diatomaceous earth After filtration, the filter cake was washed with 500 mL of dichloromethane, and the filtrate was concentrated to obtain the crude compound 27-9.
  • Step O A solution of compound 27-9 (2.33 g, 3.54 mmol), compound 5-7 (815.77 mg, 7.09 mmol) and triethylamine (717.00 mg, 7.09 mmol) in 30 mL of dichloromethane was stirred at 45 °C for 1 hour , cooled to room temperature, then NaBH(OAc) 3 (1.5 g, 7.09 mmol) and acetic acid (319.12 mg, 5.31 mmol) were added, and the mixture was stirred at 25° C. for 2 hours and concentrated to obtain the crude product.
  • NaBH(OAc) 3 1.5 g, 7.09 mmol
  • acetic acid 319.12 mg, 5.31 mmol
  • Step A A solution of compound 27-3 (3.0 g, 14.88 mmol) and compound 28-1 (3.10 g, 14.14 mmol) in 60 mL of ethanol was stirred at 25 °C for 1 hour, concentrated, and then dissolved in 60 mL of dichloromethane solution, Dichlorodicyanobenzoquinone (3.21 g, 14.14 mmol) was added, the reaction system was stirred at 25°C for 12 hours, then quenched by adding 30 mL of saturated sodium sulfite solution, and extracted with dichloromethane (40 mL ⁇ 3), and the organic phases were combined after Washed with saturated brine (20 mL ⁇ 3), dried over anhydrous sodium sulfate, filtered and concentrated to obtain crude product.
  • Step B DIBAL-H solution in toluene (1 mol/L, 7.68 mL) was added dropwise to a solution of compound 28-2 (1.4 g, 3.49 mmol) in 30 mL of dichloromethane at -78 °C, and the reaction system was heated at 25 °C. Stir for 2 hours, then add 50 mL of saturated potassium sodium tartrate solution to quench, extract with dichloromethane (40 mL ⁇ 3), combine the organic phases, wash with saturated brine (30 mL ⁇ 3), dry over anhydrous sodium sulfate, filter and concentrate Get crude.
  • Step C compound 28-3 (570.18 mg, 1.53 mmol), compound 4-1 (0.8 g, 1.53 mmol), sodium carbonate (405.02 mg, 3.82 mmol) and Pd(PPh 3 ) 4 (176.63 mg, 152.85 ⁇ mol)
  • a mixed solution of dioxane (15 mL) and water (3 mL) was replaced with nitrogen three times and heated to 100° C. under nitrogen protection and stirred for 12 hours.
  • the reaction solution was diluted with 20 mL of water and filtered through celite.
  • Step D Compound 28-4 (0.78 g, 1.13 mmol), potassium ferrocyanide (477.80 mg, 1.13 mmol), potassium acetate (22.20 mg, 226.24 ⁇ mol) and t-BuXPhos-Pd-G3 (89.86 mg, 113.12 ⁇ mol) of a mixed solution of dioxane (10 mL) and water (10 mL) was replaced with nitrogen three times and heated to 100° C. under nitrogen protection and stirred for 3 hours. The reaction solution was diluted with 20 mL of water and filtered through celite.
  • Step E Manganese dioxide (1.84 g, 21.17 mmol) was added to a solution of compound 28-5 (0.72 g, 1.06 mmol) in 30 mL of dichloromethane, the reaction system was stirred at 50° C. for 12 hours, and the reaction solution was passed through diatomaceous earth After filtration, the filter cake was washed with 400 mL of dichloromethane, and the filtrate was concentrated to obtain the crude compound 28-6.
  • Step F A solution of compound 28-6 (0.2 g, 294.95 ⁇ mol), compound 5-7 (67.91 mg, 589.90 mmol) and triethylamine (59.69 mg, 589.90 ⁇ mol) in 15 mL dichloromethane was stirred at 45 °C for 1 hour , cooled to room temperature, then added NaBH(OAc) 3 (125.02 mg, 589.90 ⁇ mol) and acetic acid (26.57 mg, 442.42 ⁇ mol), stirred at 25° C. for 12 hours and concentrated to obtain crude product.
  • NaBH(OAc) 3 125.02 mg, 589.90 ⁇ mol
  • acetic acid 26.57 mg, 442.42 ⁇ mol
  • Step A Compound 29-1 (5 g, 23.09 mmol), iron powder (12.89 g, 230.82 mmol) and saturated solution of ammonium chloride (5 mL) were added to 100 mL of methanol solution, and reacted at 75° C. for 2 hours. The reaction solution was cooled to room temperature and filtered through celite. After the filtrate was concentrated, 20 mL of water was added and extracted with ethyl acetate (20 mL ⁇ 2). The organic phases were combined, washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, and filtered. Concentration gave crude compound 29-2. MS (ESI) m/z: 186.9 [M+H] + .
  • Step B 100 mL of dichloromethane solution was added to compound 29-3 (16.83 g, 78.24 mmol), oxalyl chloride (24.83 g, 195.61 mmol) was added at 0 °C, and N,N-dimethylform was added at 25 °C formamide (285.96 mg, 3.91 mmol) and reacted at 25 °C for 0.5 h under nitrogen protection.
  • Step D To a solution of compound 29-5 (0.8 g, 2.30 mmol) in dichloromethane (30 mL) was slowly added dropwise a solution of DIBAL-H in toluene (1 mol/L, 4.61 mL) at -78°C under nitrogen protection , the reaction solution was stirred at minus 78°C for 1 hour. 20 mL of saturated sodium sulfate solution was slowly added to the reaction solution, and after returning to room temperature, the mixture was stirred for 0.5 hours and then filtered through celite.
  • Step F To a solution of compound 29-7 (0.6 g, 943.90 ⁇ mol) in dichloromethane (20 mL) was added manganese dioxide (1.64 g, 18.88 mmol) and stirred at 45° C. for 48 hours. After cooling, the reaction solution was filtered through celite, and the filtrate was concentrated to obtain compound 29-8. MS (ESI) m/z: 634.0 [M+H] + .
  • Step G Compound 5-7 (72.68 mg, 631.27 ⁇ mol) and triethylamine (70.27 mg, 694.39 ⁇ mol) were added to a dichloromethane solution (10 mL) of compound 29-8 (0.2 g, 315.23 ⁇ mol), the reaction solution was Stir for 1 hour at 45°C under nitrogen protection. The reaction solution was cooled to room temperature, NaBH(OAc) 3 (147.17 mg, 694.39 ⁇ mol) and acetic acid (30.33 mg, 505.01 ⁇ mol) were added, and the reaction solution was stirred at 25° C. under nitrogen protection for 12 hours.
  • Step A To a solution of compound 29-8 (60 mg, 94.69 ⁇ mol) in dichloromethane (10 mL) was added compound 23-6 (24.46 mg, 189.38 ⁇ mol) and Molecular sieves (20mg) were stirred at 25°C for 0.5 hours, then sodium borohydride acetate (60.21mg, 284.07 ⁇ mol) was added and stirred at 25°C for 1 hour. The reaction solution was filtered through celite, and the filtrate was concentrated to obtain a crude product.
  • Step B Compound 31-1 (189 mg, 520.33 ⁇ mol) and dichloromethane (10 mL) were added to a round-bottomed flask, cooled to -78°C, and a solution of DIBAL-H in toluene (1 mol/L, 1.04 mL) was added dropwise, The temperature was kept not to exceed -65°C, and after the addition was complete, the mixture was stirred at -78°C for 3 hours. Saturated sodium sulfate solution (10 mL) was carefully added with stirring, filtered, and the filtrate was concentrated to obtain compound 31-2, which was used directly in the next step.
  • Step D To a solution of compound 31-3 (110 mg, 168.78 ⁇ mol) in dichloromethane (10 mL) was added active manganese dioxide (293.48 mg, 3.38 mmol) and stirred at 45° C. for 48 hours. Filtration and concentration of the filtrate gave compound 31-4, which was used directly in the next step.
  • Step E To compound 31-4 (80 mg, 123.13 ⁇ mol), compound 1-22 (28.35 mg, 246.26 ⁇ mol), dichloromethane (10 mL) was added triethylamine (37.38 mg, 369.39 ⁇ mol), at 45°C After stirring for 1 hour, NaBH(OAc) 3 (78.29 mg, 369.39 ⁇ mol) and acetic acid (14.79 mg, 246.26 ⁇ mol) were added at 25° C. and stirring was continued for 12 hours.
  • Step A 20 mL of acetic acid solution of nitric acid (9.16 g, 145.30 mmol) was added dropwise to a solution of compound 32-1 (10 g, 53.59 mmol) in 20 mL of acetic acid at 0 °C, the reaction system was stirred at 25 °C for 2 hours, and then 50 mL of water was added Diluted, filtered, and the filter cake was washed with 50 mL of water and concentrated to obtain crude compound 32-2.
  • Step B to the mixed solution of compound 32-2 (3g, 12.95mmol) in 30mL of methanol and 6mL of water, add hydrosulfite (13.53g, 77.72mmol), the reaction system was stirred at 70 ° C for 12 hours, filtered, and the filter cake was Wash with 100 mL of methanol, and concentrate the filtrate to obtain the crude product.
  • Step C Compound 32-3 (4.2 g, 20.83 mmol) and compound 32-4 (3.94 g, 19.79 mmol) in 100 mL of ethanol were stirred at 25 °C for 1 hour, concentrated, and then dissolved in 100 mL of dichloromethane solution, Dichlorodicyanobenzoquinone (4.49 g, 19.79 mmol) was added, the reaction system was stirred at 25°C for 12 hours, then 100 mL of saturated sodium sulfite solution was added to quench, and then extracted with dichloromethane (80 mL ⁇ 3), and the organic phases were combined.
  • Step I A solution of compound 32-10 (0.15 g, 218.76 ⁇ mol) and compound 1-10 (38.12 mg, 437.52 ⁇ mol) in 5 mL of dichloromethane was stirred at 25° C. for 0.5 h, and NaBH(OAc) 3 (139.09 mg) was added. , 656.29 ⁇ mol), the reaction system was stirred at 25 °C for 12 hours. The reaction solution was concentrated to obtain the crude product.
  • Step F A solution of compound 33-5 (0.15 g, 218.76 ⁇ mol) and compound 33-6 (44.25 mg, 437.52 ⁇ mol) in 5 mL of dichloromethane was stirred at 25 °C for 0.5 h, and NaBH(OAc) 3 (139.09 mg) was added , 656.29 ⁇ mol), the reaction system was stirred at 25 °C for 12 hours.
  • preparative high performance liquid chromatography chromatographic column information: Phenomenex Synergi C18 150*25mm*10 ⁇ m; mobile phase: [pure water (formic acid)-acetonitrile]; acetonitrile %: 11%-41%, 10 minutes
  • PD1 PD-L1 TR-FRET detection kit was purchased from BPS Biosciences. Nivo Multilabel Analyzer (PerkinElmer).
  • the compounds to be tested were diluted 5 times to the 8th concentration, that is, diluted from 4 ⁇ M to 0.05 nM, and the DMSO concentration was 4%, and a double-well experiment was set up.
  • the IC 50 value can be obtained by curve fitting with four parameters (log(inhibitor) vs.response in GraphPad Prism --Variable slope mode).
  • Table 1 provides the inhibitory activity of compounds of the invention on PD1/PD-L1 binding.
  • the compound of the present invention has good PD1/PD-L1 binding inhibitory activity at the enzyme level.
  • PD1 PD-L1 TR-FRET detection kit was purchased from BPS Biosciences. Birght-Glo reagent was purchased from Promega. Nivo Multilabel Analyzer (PerkinElmer).
  • the TCR Activitor/PD-L1 CHO cells with a growth confluence of 80% were plated into the plate at 35,000 cells per well and placed in a 37°C cell incubator overnight; the compounds to be tested were diluted 5-fold with a row gun to the 8th
  • T cell receptor/PD-L1 CHO cell supernatant Discard the T cell receptor/PD-L1 CHO cell supernatant, add 50 ⁇ L of compound working solution to each well, and incubate at 37°C for 30 minutes; after the incubation, add 50 ⁇ L of PD-1/NFAT Reporter- at a density of 4 ⁇ 105/mL to each well Jurkat cell suspension was incubated at 37°C for 5 hours. After the incubation, 100 ⁇ L Bright-Glo was added to each well, and after mixing, the chemiluminescence signal was read using a Nivo multi-label analyzer.
  • the IC 50 value can be obtained by curve fitting with four parameters (log(inhibitor) vs.response in GraphPad Prism --Variable slope mode).
  • the inhibition degree of the compound on PD-1/PD-L1 was converted by the equation (Max-Min)/Min. The higher the fold, the stronger the inhibition of PD-1/PD-L1 pathway.
  • Figure 1 and Table 2 provide the inhibitory activity of the compounds of the present invention on PD-1/PD-L1 binding.
  • test compound IC50 in NFAT cells Relative DMSO activation fold 1 35 7.81 2 16 5.63 3 4.7 6.74 4 4.5 5.18 7 3.0 5.5 8 3.9 4.69 9 4.4 4.63 11 24.6 4.42 12 10.9 3.54 13 19.0 4.54 14 10.7 3.71 15 35.1 4.36 16 9.2 4.53 17 7.5 4.72 18 3.5 4.23 19 30.7 4.28 20 42.7 4.39 twenty two 22.6 3.62 twenty three 32.6 4.82 twenty four 32.6 4.79 27 6.71 5.12 28 4.67 4.43 29 6.03 4.03 30 14.54 4.13 32 8.98 4.05 33 13.62 3.84
  • the compound of the present invention can effectively block the PD-1/PD-L1 signaling pathway at the cellular level and restore the activity of T cells.
  • the LC/MS/MS method was used to determine the drug concentration in the plasma at different times after the mice were given the test compounds by gavage. To study the pharmacokinetic behavior of test compounds in mice, and to evaluate their pharmacokinetic characteristics.
  • Test animals healthy male C57BL/6 mice.
  • Drug preparation the vehicle in IV group is 5% DMSO+95% (20% hydroxypropyl- ⁇ -cyclodextrin); the vehicle in PO group is 5% DMSO+95% (20% HP- ⁇ -CD).
  • Administration The test compound was administered at a dose of 1 mg/kg IV and 10 mg/kg or 30 mg/kg PO (Note: the PO group was administered after an overnight fast).
  • the compound of the present invention has excellent pharmacokinetic properties, has a longer half-life in vivo, higher plasma exposure and bioavailability, and has good druggability.
  • Experimental example 4 PD-L1 antibody drug efficacy experiment based on MC38-hPD-L1 colon cancer animal model of B-hPD-L1 humanized mice Experimental method:
  • Mouse colon cancer MC38 cells were purchased from Shunran Shanghai Biotechnology Co., Ltd. Biositu (Beijing) Pharmaceutical Technology Co., Ltd. genetically modified MC38 to express human PD-L1, named MC38-hPD-L1.
  • the cells are adherent cells and are cultured in an incubator at 37° C. and 5% CO 2 , and the medium composition is Dulbecco's Modified Eagle's Medium medium containing 10% inactivated fetal bovine serum.
  • MC38-hPD-L1 cells resuspended in PBS were inoculated subcutaneously on the right side of B-hPD-L1 mice at a volume of 5 ⁇ 10 5 cells/0.1 mL/cell.
  • appropriate mice were selected according to the tumor volume and body weight of the mice, and they were equally distributed into each experimental group, with 8 mice in each group.
  • the administration started on the day of grouping.
  • the administration vehicle of the oral group was 5% DMSO+95% (20% HP- ⁇ -CD dissolved in water), and the administration vehicle of the intraperitoneal injection group was 0.9% sodium chloride injection.
  • the specific dosing schedule is shown in Table 5:
  • a The administration volume is calculated as 10 ⁇ L/g according to the body weight of the experimental animals; b. The dose is 20 mg/kg within 5 days after grouping, and the dose is adjusted to 50 mg/kg from the 6th day; c: p.o. refers to oral administration, i.p. Refers to intraperitoneal injection; d: TIW refers to 3 times a week, BID refers to twice a day.
  • TGI TV Tumor volume inhibition rate
  • TGI TV (%) [1-(Ti-T0)/(Vi-V0)] ⁇ 100%
  • Ti the mean tumor volume of the treatment group on the i day of administration
  • T0 the mean tumor volume of the treatment group on the 0th day of administration
  • Vi the mean tumor volume of the solvent control group on the ith day of administration
  • V0 the mean tumor volume of the solvent control group on the 0th day of administration
  • TGI TV (%) 88.4% (p ⁇ 0.0001) for atezolizumab (group G2)
  • TGI TV (%) 86.1% (p ⁇ 0.0001) for compound 4 (group G4)
  • compound 27 (group G5) ) of TGI TV (%) 86.1% (p ⁇ 0.0001)
  • the changes of tumor-bearing volume and body weight of experimental animals are shown in Figure 2 and Figure 3, respectively.
  • the tumor tissues of each group were taken for TILs (tumor infiltrating lymphocyte analysis) detection. The results of TILs detection are shown in Figure 4 and Figure 5 .
  • the compounds of the present invention can significantly inhibit the expression of tumor PD-L1 in mice, effectively activate immunity, and inhibit the growth of hPD-L1 positive MC38 tumors.
  • OBJECTIVE To evaluate the antitumor effect of the test drugs in the human melanoma A375 mixed PBMC subcutaneous xenograft model.
  • A375 cells were cultured in DMEM medium containing 10% fetal bovine serum (FBS). A375 cells in exponential growth phase were collected and resuspended in HBSS to an appropriate concentration for subcutaneous tumor inoculation in NCG mice. The experimental A375 cells were incubated with Mitomycin C and washed with PBS.
  • FBS fetal bovine serum
  • PBMCs Frozen PBMCs were purchased, recovered and counted, and the obtained PBMCs were added to A375 cells treated with Mitomycin C. PBMCs were co-cultured with A375 in RPMI 1640 medium containing IL-2 and 10% FBS.
  • mice After PBMC and A375 were co-cultured, PBMC and freshly digested A375 cells were harvested and inoculated subcutaneously on the right side of NCG mice. Then, the mice were randomly divided into groups according to their body weight.
  • the tumor volume and tumor growth inhibition rate TGI (%) were used to detect and evaluate the inhibitory effect or complete cure ability of the test drugs in human melanoma A375 mixed PBMC in vivo xenograft tumor.

Abstract

一类吲哚啉类化合物,具体公开了式(I)所示化合物以及其药学上可接受的盐在制备治疗相关疾病药物中的应用。

Description

吲哚啉类化合物
本申请主张如下优先权:
CN202110172933.9,申请日2021年02月08日;
CN202110934819.5,申请日2021年08月13日;
CN202111076891.5,申请日2021年09月14日;
CN202111204272.X,申请日2021年10月15日;
CN202111593525.7,申请日2021年12月23日。
技术领域
本发明涉及一类新的吲哚啉类化合物,具体涉及式(I)化合物及其药学上可接受的盐在制备治疗相关疾病药物中的应用。
背景技术
PD-1全称是程序性死亡受体1,英文名字为programmed death 1,是一种重要的免疫抑制分子,为CD28超家族成员。PD-L1全称是程序性死亡受体-配体1,英文名字programmed cell death-Ligand 1,是一种40kD的跨膜蛋白,由CD274基因编码。PD-L1可被诱导表达于T细胞、B细胞、树突细胞、巨噬细胞、间充质干细胞、骨髓来源的肥大细胞和非造血细胞的表面上,在干扰素及其他炎症因子刺激应答的肿瘤组织和其他组织中都可能会迅速上调。PD-1/PD-L1通路激活后,在癌症、妊娠、组织移植以及自身免疫病中抑制免疫系统。此外,PD-L1还能与CD80结合,竞争性抑制CD80与配体结合的T细胞激活通路,成为PD-L1抑制T细胞活性的另一机制。
在正常情况下,PD-1/PD-L1信号通路可以放置免疫系统对组织的过度攻击而诱导的过度炎症、自身免疫疾病。在非正常情况下,如肿瘤组织及慢性HBV感染组织内,存在PD-L1过度表达的情况。PD-1/PD-L1的过表达及信号通路激活抑制了功能性T细胞的活化及增殖,抑制抗肿瘤免疫反应,使得免疫系统对肿瘤的发展失去抑制作用,进而加速肿瘤的发展与恶化。针对该通路已经有多种药物获批。其中,PD-L1单克隆抗体,如Atezolizumab已经在尿路上皮癌及非小细胞肺癌适应证尚获批,在肿瘤相关适应证上还有更多临床研究在进行中。但是,相比小分子,大分子药物在诸如组织渗透、药代动力学性质、费用及使用方式上都存在明显的缺点,因此,在PD-1/PD-L1信号通路上开发小分子药物依旧是未被满足的临床需求,具备广阔的市场前景。
目前,百施美-施贵宝公司、Incyte制药公司及Gilead制药公司均有小分子PD-1/PD-L1抑制剂报道。Incyte制药公司的专利WO2018119224报道了该系列化合物展示了较好的细胞活性,并将化合物INCB086550推上了临床。尽管如此,上述小分子化合物系列在化合物的药理性质(体外活性和体内抑制肿瘤生长活性等),还有很大的改善空间;因此,在PD-1/PD-L1信号通路小分子抑制剂的开发上面,提示还有更大的空间及前景。
发明内容
本发明提供式(I)化合物或其药学上可接受的盐
Figure PCTCN2022075562-appb-000001
其中,
环A选自
Figure PCTCN2022075562-appb-000002
L 1和L 2分别独立地选自-CH 2-和-CH 2-NH-CH 2-;
Z和E分别独立地选自CH和N;
Z 1选自O和S;
Z 2选自N和CR 9
X选自N和CR 14
Y选自N和CR 15
R 1选自H、CH 3和CHF 2
R 2选自CH 3和Cl;
R 3、R 4、R 5和R 6分别独立地选自H、C 1-6烷基、OH、COOH和-C 1-3烷基-COOH;
或者,R 3、R 4连同它们所连接的原子一起形成氮杂环丁烷基、吡咯烷基、恶唑烷基或哌啶基,所述氮杂环丁烷基、吡咯烷基、恶唑烷基和哌啶基分别独立任选被1、2或3个R 16取代;
或者,R 5、R 6连同它们所连接的原子一起形成氮杂环丁烷基、吡咯烷基、恶唑烷基或哌啶基,所述氮杂环丁烷基、吡咯烷基、恶唑烷基和哌啶基分别独立任选被1、2或3个R 16取代;
R 7选自H、F、Cl、CH 3和CHF 2
R 8选自-OCH 3、-O-CH 2-F、和-O-CH 2-CN;
R 9选自H、F和CN;
R 14和R 15分别独立地选自H和C 1-6烷基;
R 16分别独立地选自H、C 1-6烷基、OH、=O、COOH和-C 1-3烷基-COOH。
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中,化合物选自
Figure PCTCN2022075562-appb-000003
其中,
环B和环C分别独立地选自氮杂环丁烷基、吡咯烷基、恶唑烷基和哌啶基;
R 10、R 11、R 12和R 13分别独立地选自H、C 1-4烷基、OH、=O、COOH和-C 1-3烷基-COOH;
环A、Z、R 1和R 2如本发明所定义。
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中,化合物选自
Figure PCTCN2022075562-appb-000004
其中,
环B和环C分别独立地选自氮杂环丁烷基、吡咯烷基、恶唑烷基和哌啶基;
R 10、R 11、R 12和R 13分别独立地选自H、C 1-4烷基、OH、=O、COOH和-C 1-3烷基-COOH;
Z 2选自CH 2和O;
Z、Z 1、Z 2、E、R 1、R 2和R 8如本发明所定义。
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中,化合物选自
Figure PCTCN2022075562-appb-000005
Figure PCTCN2022075562-appb-000006
其中,
Z 3选自-CH 2-和-O-,
R 10、R 11、R 12和R 13分别独立地选自H、C 1-4烷基、OH、=O、COOH和-C 1-3烷基-COOH,
Z、Z 1、Z 2、E、R 1、R 2和R 8如本发明所定义。
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中,Z 1选自O,Z 2选自C(CN),其他变量如本发明所定义。
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中,X选自N,其他变量如本发明所定义。
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中,Y选自N,其他变量如本发明所定义。
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中,R 1选自CHF 2,其他变量如本发明所定义。
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中,R 3、R 4、R 5和R 6分别独立地选自H、CH 3、异丙基、
Figure PCTCN2022075562-appb-000007
COOH和-C 1-3烷基-COOH,其他变量如本发明所定义。
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中R 7选自H,其他变量如本发明所定义。
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中,环A选自
Figure PCTCN2022075562-appb-000008
Figure PCTCN2022075562-appb-000009
其他变量如本发明所定义。
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中,结构单元
Figure PCTCN2022075562-appb-000010
分别独立地选自
Figure PCTCN2022075562-appb-000011
Figure PCTCN2022075562-appb-000012
Figure PCTCN2022075562-appb-000013
其他变量如本发明所定义。
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中,结构单元
Figure PCTCN2022075562-appb-000014
Figure PCTCN2022075562-appb-000015
分别独立地选自
Figure PCTCN2022075562-appb-000016
Figure PCTCN2022075562-appb-000017
Figure PCTCN2022075562-appb-000018
其他变量如本发明所定义。
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中,结构单元
Figure PCTCN2022075562-appb-000019
Figure PCTCN2022075562-appb-000020
分别独立地选自
Figure PCTCN2022075562-appb-000021
Figure PCTCN2022075562-appb-000022
其他变量如本发明所定义。
本发明还提供下式化合物或其药学上可接受的盐
Figure PCTCN2022075562-appb-000023
Figure PCTCN2022075562-appb-000024
Figure PCTCN2022075562-appb-000025
Figure PCTCN2022075562-appb-000026
Figure PCTCN2022075562-appb-000027
Figure PCTCN2022075562-appb-000028
在本发明的一些方案中,上述化合物或其药学上可接受的盐,其中,化合物选自
Figure PCTCN2022075562-appb-000029
Figure PCTCN2022075562-appb-000030
Figure PCTCN2022075562-appb-000031
Figure PCTCN2022075562-appb-000032
Figure PCTCN2022075562-appb-000033
Figure PCTCN2022075562-appb-000034
Figure PCTCN2022075562-appb-000035
Figure PCTCN2022075562-appb-000036
本发明还有一些方案是由上述各变量任意组合而来。
在本发明的一些方案中,上述化合物或其药学上可接受的盐在制备治疗与PD-L1有关的疾病药物中的应用。
在本发明的一些方案中,上述化合物或其药学上可接受的盐在制备治疗肿瘤药物中的应用。
在本发明的一些方案中,上述肿瘤为结肠癌、黑色素瘤、非小细胞肺癌、肝细胞癌或肾细胞癌。
技术效果
本发明化合物是对PD-L1具有较好的抑制活性的小分子抑制剂,与现有治疗方案(PD-L1单抗药物)具有很大差别;小分子抑制剂在生产上相比单抗药物成本更低,更适合大规模生产;同时,小分子药物的药代动力学性质与大分子单抗药物也显著不同,具有更快的半衰期及更大组织分布,因此小分子PD-L1抑制剂在临床应用中相比大分子药物可以有更灵活的给药方案及更可控的副作用。经过多轮的寻找及评估,意外的发现本发明所述化合物具有很好的体外活性及药代动力学性质,在临床前研究中展现了较高的口服暴露量。同时,这些化合物在在体内药效方面表现出了显著的药效,具有较好的肿瘤(如结肠癌、黑色素瘤、非小细胞肺癌、肝细胞癌或肾细胞癌等)抑制活性。这些性质支持了该发明系列化合物口服用于PD-L1阳性引起的疾病的治疗,具有良好的成药及临床应用前景。
定义和说明
除非另有说明,本文所用的下列术语和短语旨在具有下列含义。一个特定的术语或短语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文中出现商品名时,意在指代其对应的商品或其活性成分。
这里所采用的术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”是指本发明化合物的盐,由本发明发现的具有特定取代基的化合物与相对无毒的酸或碱制备。当本发明的化合物中含有相对酸性的功能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的碱与这类化合物接触的方式获得碱加成盐。药学上可接受的碱加成盐包括钠、钾、钙、铵、有机胺或镁盐或类似的盐。当本发明的化合物中含有相对碱性的官能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的酸与这类化合物接触的方式获得酸加成盐。药学上可接受的酸加成盐的实例包括无机酸盐,所述无机酸包括例如盐酸、氢溴酸、硝酸、碳酸,碳酸氢根,磷酸、磷酸一氢根、磷酸二氢根、硫酸、硫酸氢根、氢碘酸、亚磷酸等;以及有机酸盐,所述有机酸包括如乙酸、丙酸、异丁酸、马来酸、丙二酸、苯甲酸、琥珀酸、辛二酸、反丁烯二酸、乳酸、扁桃酸、邻苯二甲酸、苯磺酸、对甲苯磺酸、柠檬酸、酒石酸和甲磺酸等类似的酸;还包括氨基酸(如精氨酸等)的盐,以及如葡糖醛酸等有机酸的盐。本发明的某些特定的化合物含有碱性和酸性的官能团,从而可以被转换成任一碱或酸加成盐。
所列举的取代基中没有指明其通过哪一个原子连接到被取代的基团上时,这种取代基可以通过其任何原子相键合,例如,吡啶基作为取代基可以通过吡啶环上任意一个碳原子连接到被取代的基团上。
当一个取代基的键可以交叉连接到一个环上的两一个以上原子时,这种取代基可以与这个环上的任意原子相键合,例如,结构单元
Figure PCTCN2022075562-appb-000037
表示其取代基R可在环己基或者环己二烯上的任意一个位置发生取代。
当所列举的连接基团没有指明其连接方向,其连接方向是任意的,例如,
Figure PCTCN2022075562-appb-000038
中连接基团L为-M-W-,此时-M-W-既可以按与从左往右的读取顺序相同的方向连接环A和环B构成
Figure PCTCN2022075562-appb-000039
也可以按照与从左往右的读取顺序相反的方向连接环A和环B构成
Figure PCTCN2022075562-appb-000040
所述连接基团、取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
除非另有规定,当某一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相连。当该化学键的连接方式是不定位的,且可连接位点存在H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。所述位点与其他基团连接的化学键可以用直形实线键
Figure PCTCN2022075562-appb-000041
直形虚线键
Figure PCTCN2022075562-appb-000042
或波浪线
Figure PCTCN2022075562-appb-000043
表示。例如-OCH 3中的直形实线键表示通过该基团中的氧原子与其他基团相连;
Figure PCTCN2022075562-appb-000044
中的直形虚线键表示通过该基团中的氮原子的两端与其他基团相连;
Figure PCTCN2022075562-appb-000045
中的波浪线表示通过该苯基基团中的1和2位碳原子与其他基团相连;
Figure PCTCN2022075562-appb-000046
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
Figure PCTCN2022075562-appb-000047
这4种连接方式,即使-N-上画出了H原子,但是
Figure PCTCN2022075562-appb-000048
仍包括
Figure PCTCN2022075562-appb-000049
这种连接方式的基团,只是在连接1个化学键时,该位点的H会对应减少1个变成相应的一价哌啶基。
除非另有规定,术语“C 1-6烷基”用于表示直链或支链的由1至6个碳原子组成的饱和碳氢基团。所述C 1-6烷基包括C 1-5、C 1-4、C 1-3、C 1-2、C 2-6、C 2-4、C 6和C 5烷基等;其可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基)。C 1-6烷基的实例包括但不限于甲基(Me)、乙基(Et)、丙基(包括n-丙基和异丙基)、丁基(包括n-丁基,异丁基,s-丁基和t-丁基)、戊基(包括n-戊基,异戊基和新戊基)、己基等。
除非另有规定,术语“C 1-4烷基”用于表示直链或支链的由1至4个碳原子组成的饱和碳氢基团。所述C 1-4烷基包括C 1-2、C 1-3和C 2-3烷基等;其可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基)。C 1-4烷基的实例包括但不限于甲基(Me)、乙基(Et)、丙基(包括n-丙基和异丙基)、丁基(包括n-丁基,异丁基,s-丁基和t-丁基)等。
除非另有规定,术语“C 1-3烷基”用于表示直链或支链的由1至3个碳原子组成的饱和碳氢基团。所述C 1-3烷基包括C 1-2和C 2-3烷基等;其可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基)。C 1-3烷基的实例包括但不限于甲基(Me)、乙基(Et)、丙基(包括n-丙基和异丙基)等。
本发明的药学上可接受的盐可由含有酸根或碱基的母体化合物通过常规化学方法合成。一般情况下,这样的盐的制备方法是:在水或有机溶剂或两者的混合物中,经由游离酸或碱形式的这些化合物与化学计量的适当的碱或酸反应来制备。
本发明的化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。
本发明的化合物可以通过本领域技术人员所熟知的常规方法来确认结构,如果本发明涉及化合物的绝对构型,则该绝对构型可以通过本领域常规技术手段予以确证。例如单晶X射线衍射法(SXRD),把培养出的单晶用Bruker D8 venture衍射仪收集衍射强度数据,光源为CuKα辐射,扫描方 式:
Figure PCTCN2022075562-appb-000050
扫描,收集相关数据后,进一步采用直接法(Shelxs97)解析晶体结构,便可以确证绝对构型。
本发明所使用的溶剂可经市售获得。
本发明采用下述缩略词:Pd(PPh 3) 4代表四(三苯基膦)钯;Pd(dppf)Cl 2·CH 2Cl 2代表1,1-双(二苯基膦)二茂铁二氯化钯二氯甲烷络合物;NaBH(OAc) 3代表三乙酰基硼氢化钠;DIAD代表偶氮二甲酸二异丙酯;Boc2O代表二碳酸二叔丁酯;t-BuXPhos-Pd-G3代表甲烷磺酸(2-二叔丁基膦基-2',4',6'-三异丙基-1,1'-联苯基)(2'-氨基-1,1'-联苯-2-基)钯(II);DIBAL-H代表二异丁基氢化铝;DMSO代表二甲基亚砜。
化合物依据本领域常规命名原则或者使用
Figure PCTCN2022075562-appb-000051
软件命名,市售化合物采用供应商目录名称。
附图说明
图1本发明的化合物对PD-1/PD-L1结合的抑制活性。
图2为实验动物荷瘤体积。
图3为实验动物体重变化。
图4为CD3+T细胞在mCD5+细胞中占比。
图5为细胞PD-L1荧光强度。
具体实施方式
下面通过实施例对本发明进行详细描述,但并不意味着对本发明任何不利限制。本发明的化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。对本领域的技术人员而言,在不脱离本发明精神和范围的情况下针对本发明具体实施方式进行各种变化和改进将是显而易见的。
实施例1
Figure PCTCN2022075562-appb-000052
Figure PCTCN2022075562-appb-000053
步骤A:将正丁基锂的正己烷溶液(2.5M,8.88mL)在-78℃时滴加到化合物1-1(5g,18.49mmol)和硼酸三异丙酯(4.17g,22.19mmol)的12.5mL 2-甲基四氢呋喃和50mL甲苯的混合溶液中,反应液在-78℃搅拌1小时,然后缓慢升温至25℃下反应1小时后加入80mL盐酸(1mol/L)淬灭,加入80mL水后用乙酸乙酯萃取(80mL×3),合并有机相用饱和食盐水洗涤(50mL×2),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品在40mL乙酸乙酯中,25℃下搅拌0.5小时,过滤得到产品1-2。
步骤B:向30mL二氧六环和6mL水的混合溶液中加入化合物1-2(3g,12.75mmol),化合物1-3(2.19g,12.75mmol),碳酸钾(3.52g,25.50mmol)和Pd(PPh 3) 4(1.47g,1.28mmol),反应体系用氮气置换三次,在85℃和氮气保护下反应12小时。反应液冷却至室温后加入20mL水后通过硅藻土过滤,然后用乙酸乙酯萃取(50mL×3),合并有机相后用3饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层层析(石油醚:乙酸乙酯=0:1-1:0,v/v)分离得到化合物1-4。MS(ESI)m/z:328.2[M+H] +
步骤C:向20mL二氧六环溶液中加入化合物1-4(1.29g,3.93mmol),双联嚬哪醇硼酸酯(2.00g,7.87mmol),乙酸钾(772.34mg,7.87mmol)和Pd(dppf)Cl 2·CH 2Cl 2(321.33mg,393.48μmol),反应体系用氮气置换三次,在85℃和氮气保护下反应6小时。反应液冷却至室温后加入15mL水后通过硅藻土过滤,然后用乙酸乙酯萃取(40mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=0:1-1:0,v/v)纯化得到化合物1-5。MS(ESI)m/z:374.4[M+H] +
步骤D:化合物1-7(2g,8.21mmol),化合物1-6(1.52g,9.86mmol,1.67mL),碳酸钠(2.18g,20.53mmol)和Pd(PPh 3) 4(474.59mg,410.70μmol)的叔丁醇(15mL)和水(15mL)的混合溶液,氮气保护下加热到80℃下反应2小时。反应物用水稀释,用乙酸乙酯萃取(40mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=0:1-1:0,v/v)纯化得到化合物1-8。MS(ESI)m/z:191.1[M+H] +
步骤E:向化合物1-8(1g,5.25mmol)的二氧六环(20mL)和水(20mL)的混合溶液内加入二水合锇酸钾(9.66mg,26.23μmol),25℃搅拌30分钟,向内分批加入高碘酸钠(2.63g,12.30mmol)。反应物继续搅拌3小时,用50%硫代硫酸钠水溶液淬灭,二氯甲烷萃取(60mL×3);有机相用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品化合物1-9,直接用于下一步MS(ESI)m/z:193.3 [M+H +]。
步骤F:化合物1-9(0.64g,3.32mmol)和化合物1-10(578.99mg,6.65mmol)的二氯甲烷(20mL)溶液25℃搅拌30分钟,向内加入NaBH(OAc) 3(2.82g,13.29mmol)。反应物25℃下搅拌12小时,用水稀释,二氯甲烷萃取(50mL×3);有机相用饱和食盐水洗涤(20mL),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(甲醇:二氯甲烷=0:1-1:0,v/v)纯化得到化合物1-11。MS(ESI)m/z:264.51[M+H] +
步骤G:化合物1-11(0.1g,379.19μmol),1-12(78.86mg,398.15μmol)和氯化氢的二氧六环(4M,94.80μL)在叔丁醇(4mL)中加热到120℃,搅拌两小时。反应液冷却到室温,用碳酸氢钠水溶液淬灭,二氯甲烷萃取(40mL×3);有机相用饱和食盐水洗涤(20mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(乙酸乙酯)纯化得到化合物1-13。MS(ESI)m/z:427.3[M+H] +
步骤H:化合物1-13(106mg,249.22μmol),化合物1-5(93.12mg,249.22μmol),碳酸钠(66.04mg,623.06μmol),和Pd(PPh 3) 4(28.80mg,24.92μmol)的二氧六环(4mL)和水(0.8mL)溶液氮气保护下加热到100℃,搅拌1小时。反应液冷却至室温后加入20mL水后通过硅藻土过滤,然后用乙酸乙酯萃取(40mL×3),合并有机相后用饱和食盐水洗涤(20mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(乙酸乙酯)纯化得到化合物1-14。MS(ESI)m/z:592.1[M+H] +
步骤I:化合物1-14(150mg,253.34μmol)和化合物1-15(57.84mg,506.68μmol)的二氯甲烷(8mL)溶液25℃搅拌30分钟,向内加入NaBH(OAc) 3(2.82g,13.29mmol)。反应物25℃下搅拌1小时,用水稀释,二氯甲烷萃取(20mL×3);有机相用饱和食盐水洗涤(20mL),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 250*50mm*15μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:12%-42%,10分钟)纯化得到化合物1。 1H NMR(400MHz,DMSO-d 6)δ=8.83(d,J=2.0Hz,1H),8.21-8.17(m,2H),7.83(d,J=7.6Hz,1H),7.72(s,1H),7.65-7.59(m,2H),7.52(t,J=7.6Hz,1H),7.44(dd,J=2.0,7.6Hz,1H),7.40(d,J=5.6Hz,1H),7.28(d,J=7.2Hz,1H),7.19(t,J=8.0Hz,1H),6.81(d,J=6.8Hz,1H),4.71-4.54(m,2H),4.30-4.15(m,1H),3.92(s,3H),3.80(d,J=7.6Hz,2H),3.72(d,J=2.0Hz,2H),3.67-3.60(m,2H),3.60(bs,4H),2.98(t,J=8.0Hz,2H),2.77-2.73(m,1H),2.69-2.63(m,1H),2.56(d,J=6.0Hz,2H),2.30-2.37(m,1H),2.17-1.97(m,4H),1.78-1.50(m,2H),MS(ESI)m/z:690.5[M+H] +
实施例2
Figure PCTCN2022075562-appb-000054
Figure PCTCN2022075562-appb-000055
步骤A:向10mL二氧六环溶液中加入化合物1-13(0.5g,1.18mmol),双联嚬哪醇硼酸酯(597.052mg,2.35mmol),乙酸钾(346.12mg,3.53mmol)和Pd(dppf)Cl 2·CH 2Cl 2(96mg,117.56μmol),反应体系用氮气置换三次,在100℃和氮气保护下反应3小时。反应液冷却至室温后加入10mL水后通过硅藻土过滤,然后用乙酸乙酯萃取(30mL×3),合并有机相后用饱和食盐水洗涤(20mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=2:1~0:1,v/v)纯化得到化合物2-1。MS(ESI)m/z:473.3[M+H] +
步骤B:向40mL二氧六环溶液中加入化合物2-2(5g,24.10mmol),双联嚬哪醇硼酸酯(7.34g,28.92mmol),乙酸钾(4.73g,48.20mmol)和Pd(dppf)Cl 2·CH 2Cl 2(1.97g,2.41mmol),反应体系用氮气置换三次,在85℃和氮气保护下反应12小时。反应液冷却至室温后通过硅藻土过滤,然后用40mL二氧六环溶液洗涤滤饼,滤液合并得到粗品化合物2-3。
步骤C:向80mL二氧六环溶液中的化合物2-3(6.13g,24.08mmol)和16mL水的混合溶液中加入化合物2-4(5.23g,24.08mmol),碳酸钾(8.32g,60.21mmol)和Pd(dppf)Cl 2·CH 2Cl 2(1.97g,2.41mmol),反应体系用氮气置换三次,在85℃和氮气保护下反应4小时。反应液冷却至室温后加入20mL水后通过硅藻土过滤,然后用乙酸乙酯萃取(80mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层层析(石油醚:乙酸乙酯=1:0~0:1,v/v)分离得到化合物2-5。MS(ESI)m/z:265.1[M+H] +
步骤D:将三氟甲磺酸酐(4.37g,15.47mmol)在-78℃时小心滴加到化合物2-5(3.15g,11.90mmol) 和N,N-二异丙基乙胺(9.23g,71.41mmol)的60mL二氯甲烷溶液中,反应液在-78℃搅拌0.5小时,然后加入50mL饱和柠檬酸溶液淬灭,加入20mL水后用二氯甲烷萃取(50mL×3),合并有机相用饱和食盐水洗涤(20mL×2),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层层析(石油醚:乙酸乙酯=1:0-0:1,v/v)分离得到化合物2-6。
步骤E:向化合物2-6(1.3g,3.28mmol)和化合物1-15(748.07mg,6.55mmol)的二氯甲烷(12mL)溶液中加入NaBH(OAc) 3(3.47g,16.38mmol)。反应物在25℃下搅拌2小时,加入100mL水稀释,乙酸乙酯萃取(100mL×2);有机相用饱和食盐水洗涤(100mL),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=1:0~0:1,v/v)纯化得到化合物2-7。MS(ESI)m/z:495.1[M+H] +
步骤F:向化合物2-7(0.5g,1.01mmol)和二碳酸二叔丁酯(441.02mg,2.02mmol)的四氢呋喃(14mL)溶液中加入三乙胺(306.72mg,3.03mmol),反应液在25℃搅拌2小时,加入100mL水后用乙酸乙酯萃取(100mL×2);有机相用饱和食盐水洗涤(100mL),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=1:0~0:1,v/v)纯化得到化合物2-8。MS(ESI)m/z:595.1[M+H] +
步骤G:化合物2-8(100mg,211.69μmol),化合物2-1(125.95mg,211.69μmol),碳酸钠(56.09mg,529.23μmol),和Pd(PPh 3) 4(24.46mg,21.17μmol)的二氧六环(2mL)和水(0.4mL)的混合溶液用氮气置换三次,在氮气保护下加热到100℃,搅拌1小时。反应液冷却至室温后加入10mL水后通过硅藻土过滤,然后用乙酸乙酯萃取(30mL×3),合并有机相后用饱和食盐水洗涤(20mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(乙酸乙酯)纯化得到化合物2-9。MS(ESI)m/z:791.5[M+H] +
步骤H:向化合物2-9(0.09g,113.73μmol)的二氯甲烷(1.8mL)溶液中加入三氟乙酸(924.00mg,8.10mmol),反应液在25℃下搅拌10分钟,浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex Synergi C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:10%-40%,10分钟)纯化得到化合物2。 1H NMR(400MHz,DMSO-d 6)δ=8.89(d,J=2.0Hz,1H),8.56(s,1H),8.31(s,1H),8.22(d,J=5.6Hz,1H),8.13(s,1H),7.72-7.67(m,2H),7.65(d,J=8.0Hz,1H),7.60(t,J=7.6Hz,1H),7.57-7.53(m,1H),7.42(d,J=5.6Hz,1H),7.20(t,J=8.0Hz,1H),6.83(d,J=7.2Hz,1H),5.15-4.94(m,1H),4.76-4.53(m,2H),4.29(s,3H),4.03(s,3H),3.90-3.78(m,2H),3.04-2.95(m,6H),2.88-2.63(m,3H),2.21-2.03(m,4H),1.85-1.64(m,2H);MS(ESI)m/z:691.2[M+H] +
实施例3
Figure PCTCN2022075562-appb-000056
Figure PCTCN2022075562-appb-000057
步骤A:化合物3-1(10g,46.08mmol),化合物3-2(64.16g,368.63mmol)一起在60℃下搅拌12小时,减压浓缩得到化合物3-3。MS(ESI)m/z:277.2[M+H +]
步骤B:化合物3-3(6.38g,23.03mmol)和氨水(28.83g,230.31mmol,31.68mL,含胺28%)在闷罐中于85℃搅拌5小时。浓缩得到粗品,加入50mL二氯甲烷/甲醇=10/1的溶剂,25℃搅拌1小时,过滤,收集固体浓缩得到化合物3-4。MS(ESI)m/z:276.2[M+H +]
步骤C:向20mL二氧六环和20mL水的混合溶液中加入化合物3-4(1.7g,6.16mmol),化合物3-5(1.9g,12.32mmol),磷酸钾(3.27g,15.40mmol)和Pd(PPh 3) 4(711.66mg,615.86μmol),反应体系用氮气置换三次,在100℃和氮气保护下反应12小时。反应液浓缩得到粗品。粗品通过硅胶柱层层析(石油醚:乙酸乙酯=0:1-1:0,V/V)分离得到化合物3-6。MS(ESI)m/z:224.3[M+H] +
步骤D:向化合物3-6(0.35g,1.57mmol),N,N-二甲基苯胺(285.06mg,2.35mmol)和苄基三乙基氯化铵(714.41mg,3.14mmol)的8mL乙腈溶液中加入氧氯化磷(1.44g,9.41mmol),反应体系在75℃下反应2小时,反应液冷却至室温后浓缩。然后和化合物1-12(1.44g,9.41mmol)溶入8mL异丙醇溶液中,再加入甲磺酸(301.44mg,3.14mmol),反应体系在80℃下反应2小时后浓缩,然后溶于30mL乙酸乙酯和20mL碳酸氢钠溶液中并用乙酸乙酯萃取(30mL×2),合并有机相后用饱和食盐水洗涤(20 mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=0:1-1:0,v/v)纯化得到化合物3-7。MS(ESI)m/z:403.2[M+H] +
步骤E:向化合物3-7(0.1g,248.00μmol)的四氢呋喃(4mL)和水(1mL)的混合溶液中加入二水合锇酸钾(913.77μg,2.48μmol)和高碘酸钠(265.23mg,1.24mmol),反应体系在25℃下反应3小时后用20mL 50%的硫代硫酸钠溶液淬灭,然后用二氯甲烷萃取(30mL×3),合并有机相后用饱和食盐水洗涤(30mL×2),无水硫酸钠干燥,过滤后浓缩得到粗品化合物3-8。MS(ESI)m/z:405.2[M+H] +
步骤F:化合物3-8(100mg,246.79μmol)和化合物1-10(43mg,493.59μmol)的二氯甲烷(6mL)溶液在25℃搅拌0.5小时,向内加入NaBH(OAc) 3(209.22mg,987.18μmol)。反应物25℃下搅拌2小时后加入10mL水,用二氯甲烷萃取(20mL×3);有机相用饱和食盐水洗涤(20mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品化合物3-9。MS(ESI)m/z:478.3[M+H] +
步骤G:化合物3-9(90mg,188.95μmol),化合物1-5(70.60mg,188.95μmol),碳酸钠(50.07mg,472.38μmol),和Pd(PPh 3) 4(65.50mg,56.69μmol)的二氧六环(5mL)和水(1mL)溶液氮气保护下加热到100℃,搅拌1小时。反应液加入20mL稀释水后通过硅藻土过滤,然后用乙酸乙酯萃取(30mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(石油醚:乙酸乙酯=1:1)纯化得到化合物3-10。MS(ESI)m/z:643.5[M+H] +
步骤H:化合物3-10(40mg,62.20μmol)和化合物1-15(14.20mg,124.40μmol)的二氯甲烷(2mL)溶液25℃搅拌0.5小时,向内加入NaBH(OAc) 3(65.91mg,311.00μmol)。反应物25℃下搅拌0.5小时,用10mL水稀释,二氯甲烷萃取(20mL×5);有机相用饱和食盐水洗涤(20mL×2),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex Synergi C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:14%-44%,10分钟)纯化得到化合物3的甲酸盐。 1H NMR(400MHz,CD 3OD)δ=8.98-8.86(m,1H),8.74(d,J=8.0Hz,1H),8.50(s,1H),8.14(s,1H),7.76(d,J=7.6Hz,1H),7.61(d,J=7.6Hz,1H),7.53-7.45(m,1H),7.42-7.32(m,2H),7.26(d,J=7.2Hz,1H),7.14-6.98(m,1H),6.87-6.51(m,1H),5.08-5.02(m,2H),4.41-4.38(m,1H),4.03(s,3H),3.98-3.80(m,5H),3.20-2.98(m,2H),2.95-2.84(m,2H),2.83-2.72(m,2H),2.72-2.58(m,2H),2.44-2.11(m,4H),1.92-1.68(m,2H);MS(ESI)m/z:741.2[M+H] +
实施例4:化合物4
Figure PCTCN2022075562-appb-000058
Figure PCTCN2022075562-appb-000059
步骤A:向10mL二氧六环溶液中加入化合物3-9(0.2g,419.89μmol),双联频哪醇硼酸酯(213.25mg,839.78μmol),乙酸钾(123.62mg,1.26mmol)和Pd(dppf)Cl 2·CH 2Cl 2(34.29mg,41.99μmol),反应体系用氮气置换三次,在95℃和氮气保护下反应1小时。反应液冷却至室温后加入100mL水,然后用乙酸乙酯萃取(100mL×2),合并有机相后用饱和食盐水洗涤(50mL×2),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过薄层色谱法(二氯甲烷:甲醇=10:1)纯化得到化合物4-1。MS(ESI)m/z:524.2[M+H] +
步骤B:化合物4-1(90mg,171.96μmol),化合物2-8(112.54mg,189.15μmol),碳酸钠(36.45mg,343.92μmol),和Pd(PPh 3) 4(19.87mg,17.20μmol)的二氧六环(8mL)和水(2mL)的混合溶液用氮气置换三次,在氮气保护下加热到100℃,搅拌1小时。反应液冷却至室温后加入50mL,然后用乙酸乙酯萃取(25mL×2),合并有机相后用饱和食盐水洗涤(25mL×2),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层色谱法(二氯甲烷:甲醇=10:1)纯化得到化合物4-2。MS(ESI)m/z:842.3[M+H] +
步骤C:向化合物4-2(80mg,94.97μmol)的二氯甲烷(6mL)溶液中加入三氟乙酸(3.08g,27.01mmol),反应液在25℃下搅拌30分钟,浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:13%-43%,10分钟)纯化得到化合物4的甲酸盐。 1H NMR(400MHz,DMSO-d6)δ=8.92(d,J=1.6Hz,1H),8.65(d,J=8.0Hz,1H),8.52(s,1H),8.18(d,J=1.6Hz,1H),8.14(s,1H),7.72(dd,J=,1H),7.68(s,1H),7.61(t,1H),7.54(dd,1H),7.42(t,J=8.0Hz,1H),7.13-6.75(m,2H),5.00(br t,J=8.0Hz,2H),4.28-4.20(m,1H),4.10(s,2H),4.01(s,3H),3.98-3.82(m,3H),3.79-3.69(m,1H),3.15-3.01(m,2H),2.85-2.70(m,4H),2.62-2.53(m,1H),2.46(br d,J=8.0Hz,2H),2.20-1.97(m,4H),1.80-1.68(m,1H),1.66-1.54(m,1H);MS(ESI)m/z:742.2[M+H] +
实施例5:化合物5
Figure PCTCN2022075562-appb-000060
Figure PCTCN2022075562-appb-000061
步骤A:化合物5-1(500mg,2.31mmol)和原甲酸三乙酯(9.81g,66.21mmol)的混合溶液在110℃下反应5小时。反应液浓缩得到粗品化合物5-2。
步骤B:向20mL二氧六环和4mL水的混合溶液中加入化合物5-2(550mg,2.43mmol),化合物3-5(749.53mg,4.87mmol),磷酸钾(1.29g,6.08mmol)和Pd(PPh 3) 4(281.18mg,243.33μmol),反应体系用氮气置换三次,在85℃和氮气保护下反应12小时。反应液浓缩得到粗品化合物5-3。MS(ESI)m/z:174.2[M+H] +
步骤C:向化合物5-3(2.39g,13.80mmol),N,N-二甲基苯胺(2.51g,20.70mmol)和苄基三乙基氯化铵(6.29g,27.60mmol)的25mL乙腈溶液中加入氧氯化磷(12.70g,82.81mmol),反应体系在75℃下反应2小时,反应液浓缩后加入冰块和30mL碳酸氢钠溶液,然后用乙酸乙酯萃取(50mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=1:0-0:1,v/v)纯化得到化合物5-4。MS(ESI)m/z:192.1[M+H] +
步骤D:向化合物5-4(120mg,626.25μmol)和化合物1-12(124.03mg,626.25μmol)的5mL异丙醇溶液中加入甲磺酸(120.38mg,1.25mmol),该体系在80℃下反应2小时,然后加入20mL碳酸氢钠溶液并用乙酸乙酯萃取(50mL×3),合并有机相后用饱和食盐水洗涤(30mL×2),无水硫酸钠干燥,过滤后浓缩得到粗品化合物5-5。MS(ESI)m/z:353.0[M+H] +
步骤E:向化合物5-5(227mg,6442.67μmol)的四氢呋喃(25mL)和水(5mL)的混合溶液中加入二水合锇酸钾(2.37mg,6.43μmol)和高碘酸钠(687.30mg,3.21mmol),反应体系在25℃下反应12小时后用20mL 50%的硫代硫酸钠溶液淬灭,然后用二氯甲烷萃取(60mL×3),合并有机相后用饱和食盐水洗涤(30mL×2),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(石油醚:乙酸乙酯=1:1)纯化得到化合物5-6。MS(ESI)m/z:355.3[M+H] +
步骤F:化合物5-6(90mg,253.39μmol)和化合物5-7(58.34mg,506.77μmol)的6mL二氯甲烷溶液在25℃搅拌0.5小时,向内加入NaBH(OAc) 3(161.11mg,760.16μmol)。反应体系在25℃下搅拌12小时后加入10mL水,用二氯甲烷萃取(50mL×5);有机相用饱和食盐水洗涤(20mL×1),无水硫酸 钠干燥,过滤后浓缩得到粗品化合物5-8。MS(ESI)m/z:454.2[M+H] +
步骤G:化合物5-8(120mg,264.13μmol),化合物1-5(128.30mg,343.37μmol),碳酸钠(69.99mg,660.33μmol)和Pd(PPh 3) 4(30.52mg,26.41μmol)的二氧六环(10mL)和水(2mL)的混合溶液用氮气置换三次并在氮气保护下加热到100℃,搅拌1小时。反应液通过硅藻土过滤,用100mL二氯甲烷洗涤滤饼,滤液合并后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(二氯甲烷:甲醇=5:1)纯化得到化合物5-9。MS(ESI)m/z:621.1[M+H] +
步骤H:化合物5-9(45mg,72.45μmol)和化合物1-15(16.54mg,144.91μmol)的2mL二氯甲烷溶液在25℃下搅拌0.5小时,向内加入NaBH(OAc)3(76.78mg,362.27μmol)。反应体系在25℃下搅拌12小时,加10mL水稀释后用二氯甲烷萃取(20mL×5),有机相用饱和食盐水洗涤(20mL×1),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:8%-38%,12分钟)纯化得到化合物5的甲酸盐。1H NMR(400MHz,DMSO-d6)δ=8.84(d,J=2.0Hz,1H),8.74(s,1H),8.52(d,J=8.0Hz,1H),8.23(s,1H),8.07(d,J=1.6Hz,1H),7.83(d,J=7.6Hz,1H),7.71(s,1H),7.65(dd,J=1.6,7.6Hz,1H),7.54(t,J=7.6Hz,1H),7.46(dd,J=2.0,7.6Hz,1H),7.38(t,J=7.6Hz,1H),7.29(d,J=7.6Hz,1H),7.03(d,J=6.8Hz,1H),4.99-4.88(m,2H),3.93(s,3H),3.85(d,J=11.2Hz,2H),3.73(d,J=2.0Hz,2H),3.10-2.92(m,4H),2.79-2.69(m,2H),2.62-2.55(m,4H),2.17-1.94(m,5H),1.78-1.60(m,1H);MS(ESI)m/z:719.2[M+H] +
实施例6:化合物6
Figure PCTCN2022075562-appb-000062
Figure PCTCN2022075562-appb-000063
步骤A:向40mL二氧六环中加入化合物6-1(8g,35.09mmol),然后再加入20mL水和氨水(18.01g,143.86mmol,28%纯度),反应体系在25℃下搅拌10分钟后分批加入保险粉(19.98g,114.74mmol),然后继续在25℃下反应5小时。反应液通过硅藻土过滤,用100mL乙酸乙酯洗涤滤饼,然后向滤液中加入20mL水稀释后用乙酸乙酯萃取(40mL×5),合并有机相后用饱和食盐水洗涤(20mL×1),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层层析(石油醚:乙酸乙酯=1:0-0:1,v/v)分离得到化合物5-1。
步骤B:化合物5-1(450mg,2.08mmol)和原乙酸三乙酯(7.96g,49.10mmol)的混合溶液在120℃下反应5小时。反应液浓缩得到粗品化合物6-2。MS(ESI)m/z:240.2[M+H] +
步骤C:向15mL二氧六环和3mL水的混合溶液中加入化合物6-2(690mg,2.87mmol),化合物3-5(885.37mg,5.75mmol),磷酸钾(1.53g,7.19mmol)和Pd(PPh 3) 4(332.15mg,287.43μmol),反应体系用氮气置换三次,在95℃和氮气保护下反应12小时。反应液浓缩得到粗品化合物6-3。MS(ESI)m/z:188.3[M+H] +
步骤D:向化合物6-3(2.2g,11.75mmol),N,N-二甲基苯胺(2.14g,17.63mmol)和苄基三乙基氯化铵(5.35g,23.50mmol)的40mL乙腈溶液中加入氧氯化磷(10.81g,70.51mmol),反应体系在75℃下反应2小时,反应液浓缩后加入冰块和30mL碳酸氢钠溶液,然后用乙酸乙酯萃取(50mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离 (石油醚:乙酸乙酯=1:0-0:1,v/v)纯化得到化合物6-4。MS(ESI)m/z:206.0[M+H] +
步骤E:向化合物6-4(230mg,1.12mmol)和化合物1-12(221.52mg,1.12mmol)的10mL异丙醇溶液中加入甲磺酸(214.98mg,2.24mmol),该体系在80℃下反应2小时,然后加入30mL碳酸氢钠溶液并用乙酸乙酯萃取(50mL×3),合并有机相后用饱和食盐水洗涤(30mL×2),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(石油醚:乙酸乙酯=1:1)纯化得到化合物6-5。MS(ESI)m/z:367.3[M+H] +
步骤F:向化合物6-5(238mg,648.07μmol)的四氢呋喃(25mL)和水(5mL)的混合溶液中加入二水合锇酸钾(2.39mg,6.48μmol)和高碘酸钠(693.08mg,3.24mmol),反应体系在25℃下反应5小时后用30mL 50%的硫代硫酸钠溶液淬灭,然后用二氯甲烷萃取(80mL×3),合并有机相后用饱和食盐水洗涤(30mL×2),无水硫酸钠干燥,过滤后浓缩得到粗品化合物6-6。MS(ESI)m/z:369.3[M+H] +
步骤G:化合物6-6(330mg,893.79μmol)和化合物5-7(205.80mg,1.79mmol)的15mL二氯甲烷溶液在25℃搅拌0.5小时,向内加入NaBH(OAc) 3(568.29mg,2.68mmol)。反应体系在25℃下搅拌12小时后加入10mL水,用二氯甲烷萃取(50mL×5);有机相用饱和食盐水洗涤(20mL×1),无水硫酸钠干燥,过滤后浓缩得到粗品化合物6-7。MS(ESI)m/z:468.2[M+H] +
步骤H:化合物6-7(657mg,1.40mmol),化合物1-5(681.39mg,1.82mmol),碳酸钠(371.71mg,3.51mmol),和Pd(PPh 3) 4(162.10mg,140.28μmol)的二氧六环(15mL)和水(3mL)的混合溶液用氮气置换三次并在氮气保护下加热到100℃,搅拌1小时。反应液通过硅藻土过滤,用150mL二氯甲烷洗涤滤饼,滤液合并后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=1:0-0:1,v/v)纯化得到化合物6-8。MS(ESI)m/z:635.0[M+H] +
步骤I:化合物6-8(350mg,551.09μmol)和化合物1-15(125.81mg,1.10mmol)的10mL二氯甲烷溶液在25℃下搅拌0.5小时,向内加入NaBH(OAc) 3(583.99mg,2.76mmol)。反应体系在25℃下搅拌12小时,加10mL水稀释后用二氯甲烷萃取(20mL×5),有机相用饱和食盐水洗涤(20mL×1),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 150*40mm*15μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:70%-37%,10分钟)纯化得到化合物6的甲酸盐。 1H NMR(400MHz,DMSO-d6)δ=8.73(d,J=1.6Hz,1H),8.53(d,J=8.4Hz,1H),8.20(s,1H),7.95(d,J=1.6Hz,1H),7.85(d,J=7.6Hz,1H),7.72(s,1H),7.64(dd,J=1.6,7.6Hz,1H),7.53(t,J=7.6Hz,1H),7.44(dd,J=1.6,7.6Hz,1H),7.36(t,J=7.6Hz,1H),7.30(d,J=7.6Hz,1H),7.00(d,J=7.2Hz,1H),4.89(t,J=8.0Hz,2H),3.94(s,3H),3.87-3.63(m,5H),3.09-2.90(m,3H),2.81-2.67(m,2H),2.66-2.55(m,7H),2.19-1.89(m,5H),1.78-1.62(m,1H);MS(ESI)m/z:733.2[M+H] +
实施例7:化合物7
Figure PCTCN2022075562-appb-000064
步骤A:在0℃时向化合物1-3(5g,29.14mmol)的60mL二氯甲烷溶液中缓慢滴加三溴化硼(21.90g,87.42mmol),该混合物在0℃下反应30分钟后加入80mL饱和碳酸氢钠溶液淬灭,然后加入30mL水稀释后用二氯甲烷萃取(40mL×5),合并有机相后用饱和食盐水洗涤(20mL×1),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=1:0-0:1,v/v)纯化得到化合物7-1。
步骤B:向化合物7-1(3.2g,20.31mmol)和溴乙腈(4.87g,40.62mmol)的40mL乙腈溶液中加入碳酸银(6.16g,22.34mmol),反应体系在90℃下搅拌12小时,反应液通过硅藻土过滤,用30mL乙酸乙酯洗涤滤饼,然后向滤液中加入20mL水稀释后用乙酸乙酯萃取(30mL×5),合并有机相后用饱和食盐水洗涤(20mL×1),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=10:1-5:1,v/v)纯化得到化合物7-2。
步骤C:化合物7-2(0.496g,2.52mmol),化合物1-2(652.95mg,2.78mmol),碳酸钾(697.41mg,5.05 mmol),和Pd(PPh 3) 4(291.55mg,252.30μmol)的二氧六环(10mL)和水(2mL)溶液用氮气置换三次并在氮气保护下加热到85℃,搅拌5小时。反应液加入20mL水稀释后用乙酸乙酯萃取(50mL×3),合并有机相后用饱和食盐水洗涤(30mL×2),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=1:0-0:1,v/v)纯化得到化合物7-3。MS(ESI)m/z:351.0[M+H] +
步骤D:化合物7-3(630mg,1.79mmol),双联嚬哪醇硼酸酯(910.07mg,3.58mmol),乙酸钾(351.72mg,3.58mmol)和Pd(dppf)Cl 2·CH 2Cl 2(146.33mg,179.19μmol)的20mL二氧六环溶液用氮气置换三次并在氮气保护下加热到85℃,搅拌5小时。反应液通过硅藻土过滤,用100mL乙酸乙酯洗涤滤饼,滤液合并后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=1:0-0:1,v/v)纯化得到化合物7-4。MS(ESI)m/z:399.1[M+H] +
步骤E:化合物7-4(100.43mg,251.93μmol),化合物3-9(100mg,209.94μmol),碳酸钠(55.63mg,524.86μmol)和Pd(PPh 3) 4(24.26mg,20.99μmol)的二氧六环(10mL)和水(2mL)的混合溶液用氮气置换三次并在氮气保护下加热到100℃,搅拌1小时。反应液通过硅藻土过滤后加入20mL水稀释后用乙酸乙酯萃取(30mL×3),合并有机相后用饱和食盐水洗涤(30mL×2),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(二氯甲烷:甲醇=10:1)纯化得到化合物7-5。MS(ESI)m/z:668.4[M+H] +
步骤F:化合物7-5(130mg,194.58μmol)和化合物1-15(44.42mg,389.17μmol)的6mL二氯甲烷溶液在25℃下搅拌0.5小时,向内加入NaBH(OAc) 3(206.20mg,972.92μmol)。反应体系在25℃下搅拌1小时后加入10mL水稀释,然后用二氯甲烷萃取(20mL×5),有机相用饱和食盐水洗涤(20mL×1),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex Synergi C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:9%-39%,10分钟)纯化得到化合物7的甲酸盐。 1H NMR(400MHz,DMSO-d6)δ=8.92(d,J=2.0Hz,1H),8.66(d,J=8.0Hz,1H),8.18(s,1H),8.16(d,J=1.6Hz,1H),7.97(d,J=7.6Hz,1H),7.74(dd,J=1.6,7.6Hz,1H),7.70(s,1H),7.58(t,J=7.6Hz,1H),7.53-7.47(m,2H),7.42(t,J=8.0Hz,1H),7.13-6.76(m,2H),5.28(s,2H),5.00(t,J=8.0Hz,2H),4.26-4.21(m,1H),3.94-3.63(m,7H),3.09(t,J=8.0Hz,2H),2.79-2.64(m,2H),2.59(d,J=6.0Hz,2H),2.42(dd,J=3.2,9.6Hz,1H),2.19-1.98(m,4H),1.78-1.50(m,2H);MS(ESI)m/z:766.5[M+H]+。
实施例8:化合物8
Figure PCTCN2022075562-appb-000065
Figure PCTCN2022075562-appb-000066
步骤A:化合物3-8(200mg,493.59μmol)和化合物5-7(113.65mg,987.18μmol)的15mL二氯甲烷溶液在25℃下搅拌0.5小时,向内加入NaBH(OAc) 3(313.83mg,1.48mmol)。反应体系在25℃下搅拌12小时后加入10mL水稀释,然后用二氯甲烷萃取(40mL×8),有机相用饱和食盐水洗涤(20mL×1),无水硫酸钠干燥,过滤后浓缩得到粗品化合物8-1。MS(ESI)m/z:504.3[M+H] +
步骤B:化合物8-1(150mg,297.43μmol),化合物7-4(118.57mg,297.43μmol),碳酸钠(78.81mg,743.57μmol)和Pd(PPh 3) 4(34.37mg,29.74μmol)的二氧六环(15mL)和水(3mL)的混合溶液用氮气置换三次并在氮气保护下加热到100℃,搅拌1小时。反应液通过硅藻土过滤,用100mL乙酸乙酯洗涤滤饼,滤液合并后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(二氯甲烷:甲醇=5:1)纯化得到化合物8-2。MS(ESI)m/z:696.4[M+H] +
步骤C:化合物8-2(80mg,114.93μmol)和化合物1-15(26.24mg,229.85μmol)的10mL二氯甲烷溶液在25℃下搅拌0.5小时,向内加入NaBH(OAc) 3(121.79mg,574.63μmol)。反应体系在25℃下搅拌1小时后加入10mL水稀释,然后用二氯甲烷萃取(20mL×5),有机相用饱和食盐水洗涤(20mL×1),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex Synergi C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:9%-39%,10分钟)纯化得到化合物8的甲酸盐。 1H NMR(400MHz,DMSO-d6)δ=8.92(d,J=2.0Hz,1H),8.66(d,J=8.0Hz,1H),8.20(s,1H),8.16(d,J=1.6Hz,1H),7.97(d,J=7.6Hz,1H),7.74(dd,J=1.6,7.6Hz,1H),7.70(s,1H),7.58(t,J=7.6Hz,1H),7.53-7.46(m,2H),7.42(t,J=7.6Hz,1H),7.12-6.76(m,2H),5.28(s,2H),5.01(t,J=8.0Hz,2H),3.92-3.82(m,2H),3.77(s,2H),3.69-3.62(m,1H),3.09(t,J=8.0Hz,2H),3.01-2.94(m,1H),2.80-2.70(m,2H),2.63-2.56(m,4H),2.21-1.93(m,5H),1.80-1.62(m,1H);MS(ESI)m/z:794.1[M+H] +
实施例9:化合物9
Figure PCTCN2022075562-appb-000067
步骤A:化合物1-5(0.5g,1.34mmol)和化合物1-15(305.50mg,1.34mmol)的10mL二氯甲烷溶液在25℃下搅拌0.5小时,向内加入NaBH(OAc) 3(1.42g,6.69mmol)。反应体系在25℃下搅拌2小时后加入20mL水稀释,然后用二氯甲烷萃取(30mL×5),有机相用饱和食盐水洗涤(20mL×1),无水硫酸钠干燥,过滤后浓缩得到粗品化合物9-1。MS(ESI)m/z:472.4[M+H] +
步骤B:向化合物9-1(200mg,423.92μmol)的5mL二氯甲烷溶液中加入二碳酸二叔丁酯(138.78mg,635.89μmol)和三乙胺(85.79mg,847.85μmol),反应体系在25℃下搅拌2小时后加入10mL水稀释,然后用二氯甲烷萃取(20mL×3),有机相用饱和食盐水洗涤(20mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(乙酸乙酯)纯化得到化合物9-2。MS(ESI)m/z:572.2[M+H] +
步骤C:化合物9-2(230mg,402.17μmol),化合物3-8(162.96mg,402.17μmol),碳酸钠(106.56mg,1.01mmol)和Pd(PPh 3) 4(46.47mg,40.22μmol)的二氧六环(5mL)和水(1mL)的混合溶液用氮气置换三次并在氮气保护下加热到100℃,搅拌1小时。反应液加入20mL水稀释后通过硅藻土过滤,然后用乙酸乙酯萃取(40mL×3),有机相用饱和食盐水洗涤(20mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(乙酸乙酯)纯化得到化合物9-3。MS(ESI)m/z:770.5[M+H] +
步骤D:向化合物9-3(30mg,38.95μmol)和化合物5-7(8.97mg,77.90μmol)的二氯甲烷(2mL)溶液中加入NaBH(OAc) 3(41.28mg,194.75μmol)。反应物在25℃下搅拌1小时,加入50mL水稀释,二氯甲烷萃取(50mL×2);有机相用饱和食盐水洗涤(100mL),无水硫酸钠干燥,过滤后浓缩得到粗 品9-4。粗品直接用于下一步。MS(ESI)m/z:869.3[M+H] +
步骤E:向化合物9-4(30mg,34.51μmol)的二氯甲烷(2mL)溶液中加入三氟乙酸(196.74mg,1.73mmol),反应液在25℃下搅拌1小时,浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:10%-40%,10分钟)纯化得到化合物9。 1H NMR(400MHz,DMSO-d6)δ=8.92(d,J=2.0Hz,1H),8.65(d,J=8.0Hz,1H),8.17(s,1H),7.88(br s,1H),7.69-7.63(m,2H),7.56(t,J=7.6Hz,1H),7.51-7.46(m,1H),7.42(t,J=8.0Hz,1H),7.34(br d,J=8.0Hz,1H),7.11-6.79(m,2H),5.01(br t,J=8.0Hz,2H),3.96(s,3H),3.91-3.86(m,2H),3.08(br t,J=8.0Hz,2H),3.03-2.90(m,2H),2.81-2.73(m,2H),2.64-2.56(m,2H),2.24-2.07(m,4H),2.06-1.92(m,4H),1.24(br s,4H);MS(ESI)m/z:769.2[M+H] +
实施例10:化合物10
Figure PCTCN2022075562-appb-000068
步骤A:化合物4-1(1g,1.91mmol),化合物2-6(758.01mg,1.91mmol),碳酸钠(607.53mg,5.73mmol)和Pd(dppf)Cl 2(156.03mg,191.07μmol)的四氢呋喃(32mL)和水(8mL)的混合溶液用氮气置换三次并在氮气保护下加热到50℃,搅拌12小时。反应液加入20mL水稀释,用乙酸乙酯(50mL×3)萃取,有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(甲醇/二氯甲烷=0%~8%,v/v)纯化得到化合物10-3。MS(ESI)m/z:644.0[M+H] +
步骤B:化合物10-3(40mg,62.11μmol)和化合物5-7(21.45mg,186.33μmol)以及
Figure PCTCN2022075562-appb-000069
分子筛(40mg)的5mL二氯甲烷溶液搅拌0.5小时,向内加入NaBH(OAc) 3(65.81mg,310.55μmol)。反应体系在15℃下搅拌1.5小时后加入2mL水,过滤,滤液浓缩得到粗品,粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:10%-40%,10分钟)纯化得到化合物10的甲酸盐。 1H NMR(400MHz,CD 3OD)δ=8.96(d,J=2.0Hz,1H),8.77(d,J=8.0Hz,1H),8.53(s,1H),8.40(br s,1H),8.24(d,J=1.6Hz,1H),7.70(dd,J=1.6,7.6Hz,1H),7.56(t,J=7.6Hz,1H),7.51-7.46(m,1H),7.40(t,J=8.0Hz,1H),7.08(d,J=7.6Hz,1H),6.86-6.53(m,1H),5.06(br  t,J=8.4Hz,2H),4.72-4.62(m,2H),4.52-4.43(m,1H),4.25-4.14(m,2H),4.12(s,3H),3.77(dd,J=6.0,11.2Hz,1H),3.68-3.47(m,3H),3.26-3.02(m,5H),2.99-2.84(m,2H),2.48-2.19(m,3H),1.97-1.82(m,1H);MS(ESI)m/z:743.3[M+H] +
实施例11:化合物11
Figure PCTCN2022075562-appb-000070
步骤A:化合物10-3(40mg,62.11μmol)和化合物11-1(24.07mg,186.33μmol)以及
Figure PCTCN2022075562-appb-000071
分子筛(40mg)的5mL二氯甲烷溶液搅拌0.5小时,向内加入NaBH(OAc) 3(65.81mg,310.55μmol)。反应体系在15℃下搅拌1.5小时后加入2mL水,过滤,滤液浓缩得到粗品,粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:11%-41%,10分钟)纯化得到化合物11。 1H NMR(400MHz,CD 3OD)δ=8.97(d,J=2.0Hz,1H),8.78(d,J=8.0Hz,1H),8.52(s,1H),8.24(d,J=2.0Hz,1H),7.70(dd,J=1.6,7.6Hz,1H),7.57(t,J=7.6Hz,1H),7.52-7.46(m,1H),7.41(t,J=8.0Hz,1H),7.08(d,J=7.2Hz,1H),6.89-6.51(m,1H),5.08(br t,J=8.4Hz,2H),4.73-4.62(m,2H),4.50-4.42(m,1H),4.21-4.08(m,5H),3.97(d,J=11.2Hz,1H),3.74-3.63(m,1H),3.62-3.51(m,1H),3.21(br d,J=12.8Hz,1H),3.16-3.02(m,4H),2.92-2.80(m,2H),2.56-2.44(m,1H),2.25(qd,J=7.2,14.4Hz,1H),2.02(td,J=8.8,13.1Hz,1H),1.92-1.83(m,1H),1.40(s,3H);MS(ESI)m/z:757.2[M+H] +
实施例12:化合物12
Figure PCTCN2022075562-appb-000072
步骤A:化合物10-3(40mg,62.11μmol)和化合物1-10(16.23mg,186.33μmol)以及
Figure PCTCN2022075562-appb-000073
分子筛(40mg)的5mL二氯甲烷溶液搅拌0.5小时,向内加入NaBH(OAc) 3(65.81mg,310.55μmol)。反应体系在15℃下搅拌1.5小时后加入2mL水,过滤,滤液浓缩得到粗品,粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:9%-36%,9分钟) 纯化得到化合物12。 1H NMR(400MHz,CD 3OD)δ=8.97-8.88(m,1H),8.77(br dd,J=5.0,8.1Hz,1H),8.46(d,J=2.4Hz,1H),8.19-8.10(m,1H),7.73-7.65(m,1H),7.60-7.52(m,1H),7.51-7.45(m,1H),7.41(dt,J=4.7,7.9Hz,1H),7.08(dd,J=3.1,7.6Hz,1H),6.87-6.54(m,1H),5.15-5.02(m,2H),4.60(br s,1H),4.41(br dd,J=2.9,6.6Hz,2H),4.12-4.02(m,5H),3.98-3.81(m,2H),3.20-2.99(m,4H),2.96-2.76(m,4H),2.66-2.54(m,2H),2.27-2.11(m,2H),1.88-1.69(m,2H);MS(ESI)m/z:715.1[M+H] +
实施例13:化合物13
Figure PCTCN2022075562-appb-000074
步骤A:化合物10-3(60mg,93.16μmol)和化合物13-1(36.66mg,279.47μmol)以及
Figure PCTCN2022075562-appb-000075
分子筛(60mg)的8mL二氯甲烷溶液搅拌0.5小时,向内加入NaBH(OAc) 3(98.72mg,465.79μmol)。反应体系在15℃下搅拌1.5小时后加入2mL水,过滤,滤液浓缩得到粗品,使用制备级高效液相色谱分离(色谱柱:YMC Triart 30*150mm*7μm;流动相:[纯水(盐酸)-乙腈];乙腈%:31%-51%,7分钟)纯化得到化合物13的盐酸盐。 1H NMR(400MHz,DMSO-d 6)δ=12.16-11.44(m,1H),9.54(br d,J=4.4Hz,1H),9.31-9.08(m,2H),8.75-8.56(m,3H),7.74(br d,J=7.6Hz,1H),7.68-7.54(m,2H),7.44(br t,J=7.6Hz,1H),7.14-6.82(m,2H),5.01(br t,J=8.0Hz,2H),4.71(br d,J=14.8Hz,2H),4.51-4.37(m,3H),4.04(s,3H),3.31-3.16(m,4H),3.09(br d,J=4.4Hz,3H),2.86-2.74(m,1H),2.39-2.26(m,1H),2.15-1.82(m,3H),0.93(br dd,J=7.2,9.6Hz,6H);MS(ESI)m/z:759.1[M+H] +
实施例14:化合物14
Figure PCTCN2022075562-appb-000076
步骤A:化合物10-3(100mg,155.26μmol)和化合物14-1(54.57mg,465.79μmol)以及
Figure PCTCN2022075562-appb-000077
分子筛(100mg)的8mL二氯甲烷溶液搅拌0.5小时,向内加入NaBH(OAc) 3(164.53mg,776.31μmol)。反应体 系在15℃下搅拌1.5小时后加入2mL水,过滤,滤液浓缩得到粗品,使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:15%-35%,10分钟)纯化得到化合物14。 1H NMR(400MHz,CD 3OD)δ=8.90(d,J=1.6Hz,1H),8.70(br d,J=8.4Hz,1H),8.47-8.38(m,1H),8.19(d,J=1.2Hz,1H),7.64(dd,J=1.2,7.6Hz,1H),7.50(t,J=7.6Hz,1H),7.41(dd,J=1.2,7.6Hz,1H),7.34(br t,J=8.0Hz,1H),7.02(d,J=7.6Hz,1H),6.80-6.49(m,1H),4.51-4.39(m,3H),4.24-4.12(m,2H),4.08(s,3H),3.22-3.10(m,4H),3.05-2.86(m,4H),2.31-2.17(m,1H),1.90(br dd,J=5.6,7.6Hz,1H),1.28(s,6H);MS(ESI)m/z:745.1[M+H] +
实施例15:化合物15
Figure PCTCN2022075562-appb-000078
步骤A:化合物10-3(50.00mg,77.63μmol),化合物15-1(20.05mg,155.26μmol)和分子筛(0.05g)的二氯甲烷(10mL)溶液25℃搅拌0.5小时,向内加入NaBH(OAc) 3(49.36mg,232.89μmol)。反应物25℃下搅拌1小时,用20mL水稀释,二氯甲烷萃取(30mL×3);有机相用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex Synergi C18 150*25mm*10μm;流动相:[纯水(甲酸)-乙腈];乙腈%:16%-36%,10分钟)纯化得到化合物15。 1H NMR(400MHz,CD 3OD)δ=8.96(s,1H),8.77(d,J=7.6Hz,1H),8.50(s,1H),8.23(s,1H),7.70(d,J=7.2Hz,1H),7.56(t,J=7.2Hz,1H),7.52-7.45(m,1H),7.41(t,J=7.6Hz,1H),7.08(d,J=7.6Hz,1H),6.90-6.52(m,1H),5.05(s,3H),4.59-4.42(m,2H),4.15-4.10(m,4H),3.94-3.77(m,2H),3.27-3.19(m,1H),3.17-3.03(m,4H),2.95-2.77(m,3H),2.67-2.55(m,1H),2.46-2.34(m,1H),2.31-2.08(m,3H),2.02-1.83(m,2H),1.41-1.28(m,1H);MS(ESI)m/z:757.0[M+H] +
实施例16:化合物16
Figure PCTCN2022075562-appb-000079
Figure PCTCN2022075562-appb-000080
步骤A:化合物3-10(60mg,93.30μmol)和化合物11-1(24.10mg,186.60μmol)的二氯甲烷(5mL)溶液25℃搅拌0.5小时,向内加入NaBH(OAc) 3(98.87mg,466.50μmol)。反应物25℃下搅拌1小时,用10mL水稀释,二氯甲烷萃取(20mL×3);有机相用饱和食盐水洗涤(20mL×2),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Waters Xbridge 150*25mm*5μm;流动相:[纯水(碳酸氢铵)-乙腈];乙腈%:35%-65%,9分钟)纯化得到化合物16。 1H NMR(400MHz,DMSO-d 6)δ=8.92(d,J=2.0Hz,1H),8.65(d,J=8.0Hz,1H),8.17(d,J=1.6Hz,1H),7.79(d,J=7.6Hz,1H),7.67(dd,J=1.6,7.6Hz,1H),7.55(t,J=7.6Hz,1H),7.47(d,J=1.6Hz,1H),7.42(t,J=8.0Hz,1H),7.30(d,J=7.6Hz,1H),7.11-6.78(m,2H),5.00(br t,J=8.4Hz,2H),4.28-4.17(m,1H),3.99-3.79(m,6H),3.63(br d,J=4.0Hz,2H),3.12-2.97(m,4H),2.80-2.62(m,4H),2.46-2.24(m,4H),2.04(dd,J=6.8,13.2Hz,1H),1.65-1.53(m,2H),1.28(s,3H);MS(ESI)m/z:756.2[M+H] +
实施例17:化合物17
Figure PCTCN2022075562-appb-000081
步骤A:化合物7-5(0.1g,149.68μmol)和化合物5-7(34.47mg,299.36μmol)的10mL二氯甲烷溶液在25℃下搅拌0.5小时,向内加入NaBH(OAc) 3(158.62mg,748.40μmol)。反应体系在25℃下搅拌12小时后加入10mL水稀释,然后用二氯甲烷萃取(20mL×5),有机相用饱和食盐水洗涤(20mL×1),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex Synergi C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:10%-40%,10分钟)纯化得到化合物17的甲酸盐。 1H NMR(400MHz,DMSO-d6)δ=8.92(d,J=1.6Hz,1H),8.66(d,J=8.4Hz,1H),8.28-8.19(m,1H),8.16(d,J=1.6Hz,1H),7.91(d,J=7.6Hz,1H),7.75(dd,J=1.6,7.6Hz,1H),7.58(t,J=7.6Hz,1H),7.53-7.46(m,2H),7.42(t,J=8.0Hz,1H),7.12-6.78(m,2H),5.28(s,2H),5.01(t,J=8.0Hz,2H),4.26-4.21(m,1H),3.96-3.78(m,3H),3.70-3.60(m,3H),3.09(t,J=8.0Hz,2H),2.99-2.90(m,1H),2.81-2.56(m,6H),2.10-1.93(m,3H),1.68-1.51(m,1H);MS(ESI)m/z:767.1[M+H] +
实施例18:化合物18
Figure PCTCN2022075562-appb-000082
步骤A:向化合物3-8(150mg,370.19μmol)和化合物18-1(74.89mg,740.38μmol)的二氯甲烷(20mL)溶液加入
Figure PCTCN2022075562-appb-000083
分子筛(150mg),在25℃搅拌30分钟,向内加入NaBH(OAc) 3(235.38mg,1.11mmol),反应液在25℃下搅拌1小时,通过硅藻土过滤,加入20mL水然后用二氯甲烷萃取(40mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到化合物18-2。MS(ESI)m/z:491.9[M+H] +
步骤B:向10mL二氧六环溶液中加入化合物18-2(0.17g,346.70μmol),双联嚬哪醇硼酸酯(132.06mg,520.04μmol),乙酸钾(85.06mg,866.74μmol)和Pd(dppf)Cl 2·CH 2Cl 2(28.31mg,34.67μmol),反应体系用氮气置换三次,在100℃和氮气保护下反应1小时。反应液冷却至室温后通过硅藻土过滤,滤饼用二氯甲烷(100ml)冲洗,合并有机相后浓缩得到粗品。粗品通过硅胶柱层析分离(甲醇:二氯甲烷=0~10%,v/v)纯化得到化合物18-3。MS(ESI)m/z:538.2[M+H] +
步骤C:向5mL二氧六环和1mL水的混合溶液中加入化合物18-3(65mg,120.95μmol),化合物2-8(75.56mg,127.00μmol),碳酸钠(32.05mg,302.38μmol)和Pd(PPh 3) 4(13.98mg,12.10μmol),反应体系用氮气置换三次,在100℃和氮气保护下反应1小时。反应液冷却至室温后加入20mL水后用二氯甲烷萃取(30mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶板(二氯甲烷:甲醇=10:1,v/v)分离得到化合物18-4。MS(ESI)m/z:856.2[M+H] +
步骤D:向化合物18-4(0.1g,116.77μmol)的二氯甲烷(3mL)溶液中加入三氟乙酸(1.54g,13.51mmol),反应液在25℃搅拌12小时。将反应液浓缩得到粗产品,粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex Synergi C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:8%-38%,10分钟)纯化得到化合物18。 1H NMR(400MHz,DMSO-d 6)δ=8.94(d,J=2.0Hz,1H),8.66(d,J=8.0Hz,1H),8.53(s,1H),8.21(s,1H),8.14(s,1H),7.75-7.67(m,2H),7.61(t,J=7.6Hz,1H),7.57-7.52(m,1H),7.42(t,J=8.0Hz,1H),7.14-6.79(m,2H),5.00(t,J=8.0Hz,2H),4.80-4.52(m,1H),4.13(s,2H),4.01(s,3H),3.97-3.92(m,2H),3.79-3.71(m,2H),3.10-3.05(m,2H),2.85(d,J=5.6Hz,2H),2.73-2.66(m,1H),2.64-2.57(m,2H),2.19-2.07(m,3H),1.85-1.69(m,3H),1.26(s,3H);MS(ESI)m/z:756.2[M+H] +
实施例19:化合物19
Figure PCTCN2022075562-appb-000084
步骤A:化合物19-1(5g,26.87mmol)溶于甲醇(30mL)中,加入盐酸(12M,6.72mL)和水(15mL),降温,0℃时,亚硝酸钠(2.23g,32.25mmol,)溶于水(15mL)滴加进反应体系,混合物在0℃搅拌0.5小时,然后双联嚬哪醇硼酸酯(20.47g,80.62mmol)溶于甲醇(30mL)加入,混合物在25℃搅拌1小时。反应液加入50mL水淬灭,然后用乙酸乙酯萃取(100mL×3),合并有机相后用饱和食盐水洗涤(50mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=1:0~0:1)纯化得到化合物19-2。
步骤B:向50mL二氧六环和10mL水中加入化合物19-2(5.79g,19.50mmol)和化合物19-3(3.84g,18.57mmol),碳酸钠(3.94g,37.13mmol)和Pd(dppf)Cl 2·CH 2Cl 2(1.52g,1.86mmol),反应体系用氮气置换三次,在50℃和氮气保护下反应12小时。反应液冷却至室温后加入30mL水后通过硅藻土过滤,然后用乙酸乙酯萃取(80mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层层析(石油醚:乙酸乙酯=1:0~0:1,v/v)分离得到化合物19-4。MS(ESI)m/z:342.9[M+H +]。
步骤C:向40mL甲醇中加入化合物19-4(3.99g,11.69mmol),甲醇钠(4.21g,23.38mmol,4.68mL,30%纯度)。混合物在50℃搅拌12小时。然后加入8mL水,混合物在50℃搅拌2小时。反应液冷却至室温后加入30mL水,用12M HCl调节pH到5,过滤,旋干滤饼得到化合物19-5。MS(ESI)m/z:322.7[M+H +]。
步骤D:向20mL甲醇中加入化合物19-5(2g,6.19mmol),浓硫酸(60.70mg,618.92μmol),混合物 在80℃搅拌12小时。旋干反应液得到粗品,粗品通过硅胶柱层层析(石油醚:乙酸乙酯=1:0~0:1,v/v)分离得到化合物19-6。MS(ESI)m/z:338.9[M+H +]
步骤E:向25mL四氢呋喃中加入化合物19-6(1.39g,4.12mmol),-78℃滴加二异丁基氢化铝(1M,10.31mL),混合物在0℃搅拌1小时。反应液升至室温后加入50mL水,然后用乙酸乙酯萃取(50mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层层析(石油醚:乙酸乙酯=1:0~0:1,v/v)分离得到化合物19-7。
步骤F:向25mL二氯甲烷中加入化合物19-7(1.33g,4.30mmol),三乙胺(870.64mg,8.60mmol,1.20mL)。降温,0℃时加入甲烷磺酰氯(1.59g,13.88mmol,1.07mL)。混合物在0℃搅拌1小时。反应液升至室温后加入40mL水,然后用二氯甲烷萃取(40mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层层析(石油醚:乙酸乙酯=1:0~0:1,v/v)分离得到化合物19-8。MS(ESI)m/z:388.8[M+H] +
步骤G:向25mL N,N-二甲基甲酰胺中加入化合物19-8(0.2g,516.46μmol),化合物19-9(89.81mg,542.29μmol),三乙胺(209.04mg,2.07mmol),混合物在25℃搅拌12小时。反应混合物加20mL水,然后用乙酸乙酯萃取(30mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(石油醚:乙酸乙酯=1:1)纯化得到化合物19-10。MS(ESI)m/z:422.0[M+H] +
步骤H:化合物19-10(52.20mg,124.19μmol),化合物4-1(65mg,124.19μmol),碳酸钠(32.91mg,310.48μmol),和Pd(PPh 3) 4(14.35mg,12.42μmol)的二氧六环(5mL)和水(1mL)溶液氮气保护下加热到100℃,搅拌1小时。反应液加入20mL水稀释后通过硅藻土过滤,然后用二氯甲烷萃取(20mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(二氯甲烷:甲醇=10:1)纯化得到化合物19-11。MS(ESI)m/z:643.5[M+H] +。MS(ESI)m/z:737.1[M+H] +
步骤I:向化合物19-11(0.05g,67.86μmol)的甲醇(5mL)和水(1mL)的溶液中加入一水合氢氧化锂(5.70mg,135.72μmol),反应液在50℃下搅拌1小时。反应冷却到室温后,先浓缩。然后用10mL水溶解,使用浓盐酸调节pH=5,在浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex Synergi C18 150*25mm*10μm;流动相:[水(甲酸)-乙腈];乙腈%:11%-41%,10分钟)纯化得到化合物19。 1H NMR(400MHz,DMSO-d 6)δ=8.92(d,J=2.0Hz,1H),8.65(d,J=8.0Hz,1H),8.36(s,1H),8.16(d,J=2.0Hz,1H),7.53(dd,J=1.2,7.6Hz,1H),7.42(q,J=7.6Hz,2H),7.33(dd,J=1.2,7.6Hz,1H),7.09-6.78(m,2H),5.00(t,J=8.4Hz,2H),4.30-4.15(m,1H),3.96(s,3H),3.91-3.76(m,4H),3.12-3.03(m,2H),2.97-2.86(m,3H),2.77-2.57(m,6H),2.41(dd,J=3.2,9.6Hz,1H),2.16(s,3H),2.10-1.98(m,2H),1.63-1.55(m,1H);MS(ESI)m/z:723.1[M+H] +
实施例20:化合物20
Figure PCTCN2022075562-appb-000085
Figure PCTCN2022075562-appb-000086
步骤A:向2mL N,N-二甲基甲酰胺溶液中加入化合物19-8(135mg,348.61μmol),化合物20-1(65.76mg,366.04μmol),三乙胺(141.10mg,1.39mmol),混合物在25℃反应4小时。反应液冷却至室温后加入20mL水,然后用乙酸乙酯萃取(20mL×3),合并有机相后用饱和食盐水洗涤(20mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过薄层色谱法(石油醚:乙酸乙酯=1:1)纯化得到化合物20-2。MS(ESI)m/z:435.9[M+H] +
步骤B:化合物20-2(9.79mg,114.64μmol),化合物4-1(60mg,114.64μmol),碳酸钠(30.38mg,286.60μmol),和Pd(PPh 3) 4(13.25mg,11.46μmol)的二氧六环(5mL)和水(1mL)的混合溶液用氮气置换三次,在氮气保护下加热到100℃,搅拌1小时。反应液冷却至室温后加入20mL,然后用二氯甲烷萃取(20mL×3),合并有机相后用饱和食盐水洗涤(20mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层色谱法(二氯甲烷:甲醇=10:1)纯化得到化合物20-3。MS(ESI)m/z:751.2[M+H] +.
步骤C:向化合物20-3(0.07g,93.23μmol)的甲醇(2mL)和水(0.4mL)的溶液中加入一水合氢氧化锂(7.82mg,186.46μmol),反应液在50℃下搅拌1小时。反应冷却到室温后,先浓缩。然后用10mL水溶解,使用12M HCl调节pH至5,在浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:[水(甲酸)-乙腈];乙腈%:13%-43%,10分钟)纯化得到化合物20。 1H NMR(400MHz,DMSO-d 6)δ=8.91(d,J=1.6Hz,1H),8.65(d,J=8.4Hz,1H),8.37(s,1H),8.16(s,1H),7.54(d,J=7.6Hz,1H),7.41(q,J=7.6Hz,2H),7.33(d,J=7.6Hz,1H),7.10-6.76(m,2H),5.00(t,J=7.6Hz,2H),4.30-4.16(m,1H),3.96(s,3H),3.92-3.78(m,4H),3.09-2.92(m,3H),2.77-2.65(m,4H),2.41(dd,J=3.2,9.6Hz,1H),2.28-2.20(m,1H),2.16(s,3H),2.09-1.96(m,2H),1.64-1.46(m,2H),1.24(s,4H);MS(ESI)m/z:737.1[M+H] +
实施例21:化合物21
Figure PCTCN2022075562-appb-000087
Figure PCTCN2022075562-appb-000088
步骤A:化合物3-8(0.6g,1.48mmol),化合物21-1(365.99mg,2.96mmol),醋酸(444.61mg,7.40mmol)和
Figure PCTCN2022075562-appb-000089
分子筛(0.6g)的15mL二氯甲烷溶液在25℃下搅拌0.5小时,向内加入NaBH(OAc) 3(941.50mg,4.44mmol)。反应体系在25℃下搅拌12小时后通过硅藻土过滤,滤液加入20mL水稀释,然后用二氯甲烷萃取(30mL×3),有机相用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(二氯甲烷:甲醇=1:0~4:1,v/v)纯化得到化合物21-2。MS(ESI)m/z:477.9[M+H] +
步骤B:化合物21-2(559.78mg,2.20mmol),双联频哪醇硼酸酯(0.7g,1.47mmol),乙酸钾(360.57mg,3.67mmol),和Pd(dppf)Cl 2·CH 2Cl 2(120.01mg,149.96μmol)的15mL二氧六环溶液用氮气置换三次并在氮气保护下加热到100℃,搅拌1小时。反应液通过硅藻土过滤,滤饼用100mL二氯甲烷洗涤,滤液浓缩得到粗品。粗品通过硅胶柱层析分离(二氯甲烷:甲醇=1:0~4:1,v/v)纯化得到化合物21-3。MS(ESI)m/z:524.3[M+H] +
步骤C:化合物21-3(200mg,382.13μmol),化合物20-2(165.97mg,382.13μmol),碳酸钠(101.26mg,955.33μmol)和Pd(PPh 3) 4(44.16mg,38.21μmol)的二氧六环(5mL)和水(1mL)的混合溶液用氮气置换三次并在氮气保护下加热到100℃,搅拌1小时。反应液加入20mL水稀释后用二氯甲烷萃取(40mL×3),合并有机相后用饱和食盐水洗涤(20mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(二氯甲烷:甲醇=10:1)纯化得到化合物21-4。MS(ESI)m/z:751.0[M+H] +
步骤D:向化合物21-4(120mg,159.82μmol)的甲醇(5mL)和水(1mL)的混合溶液中加入一水合氢氧化锂(13.41mg,319.64μmol),反应体系在50℃下搅拌0.5小时后浓缩,然后加入10mL水稀释,用浓盐酸调节pH到5,然后浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 150*40mm*15μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:13%-43%,10分钟)纯化得到化合物21 的甲酸盐。 1H NMR(400MHz,CD 3OD)δ=8.90(s,1H),8.75(d,J=8.4Hz,1H),8.38(d,J=1.6Hz,2H),8.18(d,J=1.2Hz,1H),7.51(d,J=7.6Hz,1H),7.41(q,J=7.6Hz,2H),7.34(d,J=7.2Hz,1H),7.04(d,J=7.6Hz,1H),6.88-6.53(m,1H),5.13-5.02(m,2H),4.67(d,J=1.6Hz,2H),4.14(s,2H),4.10(s,3H),3.98(d,J=11.2Hz,1H),3.76-3.53(m,4H),3.45(d,J=8.0Hz,2H),3.23(d,J=11.2Hz,1H),3.18-3.04(m,1H),3.03-2.85(m,1H),2.57-2.42(m,1H),2.21(s,3H),2.06-1.99(m,1H),1.53(s,3H),1.41(s,3H);MS(ESI)m/z:737.0[M+H] +
实施例22:化合物22
Figure PCTCN2022075562-appb-000090
步骤A:向10mL二氧六环溶液中加入化合物3-8(0.5g,1.23mmol),双联嚬哪醇硼酸酯(470.03mg,1.85mmol),乙酸钾(363.31mg,3.70mmol)和Pd(dppf)Cl 2·CH 2Cl 2(100.77mg,123.40μmol),反应体系用氮气置换三次,在100℃和氮气保护下反应2小时。反应液冷却至室温后加入50mL水后用乙酸乙酯萃取(50mL×2),合并有机相后用饱和食盐水洗涤(50mL),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=0~20%,v/v)纯化得到化合物22-1。MS(ESI)m/z:453.1[M+H] +
步骤B:化合物22-2(2g,17.08mmol)溶于四氢呋喃(20mL),加入三苯基膦(4.93g,18.79mmol)和DIAD(3.80g,18.79mmol)。该混合物于25℃下搅拌6小时。加入石油醚(100mL),过滤,收集滤饼减 压干燥,使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18(250*70mm,10μm);流动相:[纯水(0.1%三氟乙酸)-乙腈];乙腈%:10%-40%,20分钟)分离纯化得到化合物22-3.MS(ESI)m/z:247.1[M+H] +
步骤C:向化合物22-3(2.8g,10.94mmol)的乙醇(30mL)溶液中加入水合肼(2.54g,49.72mmol).该混合物于50℃下搅拌0.5小时,升温至75℃继续搅拌2小时。白色固体析出,过滤,滤液浓缩后加入乙醇(50mL),有白色固体析出,继续过滤,滤液浓缩得到化合物22-4。
步骤D:向化合物2-6(256.24mg,645.89μmol)的甲醇(10mL)溶液中加入NaBH(OAc) 3(405.89mg,6.46mmol)和化合物22-4(150mg,1.29mmol)。该混合物在25℃搅拌5小时。0℃下加入水(10mL)淬灭反应,二氯甲烷(15mL×3)萃取,合并有机相后用饱和食盐水(15mL×2)洗涤,无水硫酸钠干燥,过滤后浓缩得到化合物22-5。
步骤E:向化合物22-5(400mg,805.08μmol)的二氯甲烷(10mL)溶液中加入Boc 2O(263.56mg,1.21mmol)和三乙胺(244.40mg,2.42mmol),该混合物于25℃下搅拌3小时。加入饱和碳酸氢钠(10mL),用二氯甲烷(20mL×2)萃取,合并有机相后用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤后浓缩得到粗品,粗品用硅胶柱层析分离(石油醚:乙酸乙酯=50:1~0:1,v/v)得到化合物22-6。
步骤F:向8mL四氢呋喃和2mL水的混合溶液中加入化合物22-1(318.19mg,703.56μmol),化合物22-6(350mg,586.30μmol),碳酸钠(155.36mg,1.47mmol)和Pd(dppf)Cl 2·CH 2Cl 2(47.88mg,58.63μmol),反应体系用氮气置换三次,在50℃和氮气保护下反应12小时。反应液冷却至室温后加入20mL水后通过硅藻土过滤,然后用乙酸乙酯萃取(20mL×2),合并有机相后用饱和食盐水洗涤(20mL),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶板(石油醚:乙酸乙酯=2:3,v/v)分离得到化合物22-7。MS(ESI)m/z:773.0[M+H] +
步骤G:向化合物22-7(70mg,90.53μmol)和化合物22-8(37.34mg,362.14μmol)的二氯甲烷(20mL)溶液加入
Figure PCTCN2022075562-appb-000091
分子筛(100mg)和醋酸(543.68μg,9.05μmol),在25℃搅拌30分钟,向内加入NaBH(OAc) 3(76.75mg,362.14μmol),反应液在25℃下搅拌2小时,通过硅藻土将反应液过滤,将滤液浓缩得到粗品。粗品通过硅胶板(甲醇:二氯甲烷=1:10,v/v)纯化得到化合物22-9。MS(ESI)m/z:860.2[M+H] +
步骤H:向化合物22-9(11mg,12.79μmol)的二氯甲烷(3mL)溶液中加入三氟乙酸(1.13g,9.90mmol),反应液在25℃搅拌20分钟。将反应液浓缩得到粗产品,粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 150*50mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:10%-40%,10分钟)纯化得到化合物22的甲酸盐。 1H NMR(400MHz,CD 3OD)δ=8.85(d,J=2.0Hz,1H),8.67(d,J=8.0Hz,1H),8.33(s,1H),8.09(d,J=2.0Hz,1H),7.57(dd,J=2.0,7.6Hz,1H),7.45(t,J=7.6Hz,1H),7.37(dd,J=2.0,7.6Hz,1H),7.31(t,J=8.0Hz,1H),6.97(dd,J=0.8,7.6Hz,1H),6.76-6.45(m,2H),5.00(s,2H),4.42(s,1H),4.12(dd,J=5.6,8.8Hz,1H),4.01(br dd,J=3.2,4.0Hz,2H),3.99-3.97(m,3H),3.96(s,2H),3.94-3.86(m,1H),3.42(s,1H),3.12-2.88(m,6H),2.80-2.75(m,2H),2.62-2.57(m,2H);MS(ESI)m/z:760.1[M+H] +
实施例23:化合物23
Figure PCTCN2022075562-appb-000092
步骤A:将化合物23-1(3.5g,18.81mmol)溶解到甲醇(40mL)中,加入HCl(12M,4.70mL)的水溶液(20mL);在0℃下搅拌,加入亚硝酸钠(1.56g,22.57mmol)的水溶液(10mL)。反应在0℃下搅拌30分钟,双联频哪醇硼酸酯(14.33g,56.44mmol)的甲醇溶液(40mL)向反应内缓慢加入。反应混合物在20℃搅拌1小时,反应混合物使用100mL水稀释,乙酸乙酯(100mL×3)萃取,有机相使用饱和食盐水洗涤,硫酸钠干燥,过滤浓缩得到粗品。粗品使用硅胶柱层析纯化(石油醚:乙酸乙酯=1:0到100:1),得到化合物23-2。
步骤B:化合物23-2(1.5g,8.74mmol)、化合物1-3(2.86g,9.62mmol)、Pd(PPh 3) 4(1.01g,874.22μmol)和碳酸钾(2.42g,17.48mmol)混合于二氧六环(30mL)和水(6mL)的溶液中,置换氮气,加热到90℃,搅拌10小时。反应混合物使用二氯甲烷(50mL)稀释,硅藻土过滤,滤液旋干后得到粗品。粗品使用硅胶柱层析纯化(石油醚:乙酸乙酯=20:1~5:1,v/v),得到化合物23-3.MS(ESI)m/z:308.0[M+H +]。
步骤C:将化合物23-3(1.4g,4.57mmol),双联频哪醇硼酸酯(2.32g,9.15mmol)二氯甲烷.二(三苯基膦)二氯化钯(373.44mg,457μmol)和醋酸钾(897mg,9.15mmol)的二氧六环溶液置换氮气后加热到85℃氮气保护下搅拌5小时。反应混合物使用二氯甲烷(50mL)稀释,硅藻土过滤,滤液旋干后得到粗品。粗品使用硅胶柱层析纯化(石油醚:乙酸乙酯=100:1~10:1,v/v),得到化合物23-4. MS(ESI)m/z:354.3[M+H +]。
步骤D:向32mL二氧六环和8mL水的混合溶液中加入化合物3-9(750mg,1.57mmol),化合物23-4(556.17mg,1.57mmol),碳酸钠(417.22mg,3.94mmol)和Pd(dppf)Cl 2·CH 2Cl 2(128.59mg,157.46μmol),反应体系用氮气置换三次,在100℃和氮气保护下反应2小时。反应液冷却至室温后在真空中浓缩,加入30mL水后用二氯甲烷萃取(30mL×2),合并有机相后浓缩得到粗品。粗品通过硅胶层析柱(甲醇:二氯甲烷=0~6%,v/v)分离得到化合物23-5。MS(ESI)m/z:623.1[M+H] +
步骤E:向化合物23-5(100mg,160.60μmol)和化合物23-6(82.97mg,642.40μmol)的二氯甲烷(10mL)溶液加入
Figure PCTCN2022075562-appb-000093
分子筛(20mg),反应液在25℃搅拌30分钟,向内加入NaBH(OAc) 3(136.15mg,642.40μmol),反应液在25℃下搅拌12小时,通过硅藻土将反应液过滤,将滤液浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Waters Xbridge 150*25mm*5μm;流动相:[纯水(碳酸氢钠)-乙腈];乙腈%:40%-70%,7分钟)纯化得到化合物23。 1H NMR(400MHz,CD 3OD)δ=9.03(d,J=2.0Hz,1H),8.78(d,J=8.0Hz,1H),8.36(d,J=2.0Hz,1H),7.93(d,J=7.6Hz,1H),7.49-7.35(m,3H),7.33-7.20(m,2H),7.06(d,J=7.6Hz,1H),6.91-6.58(m,1H),5.10(br t,J=8.4Hz,2H),4.62-4.53(m,3H),4.48(s,2H),4.10(s,3H),3.85(br d,J=10.8Hz,1H),3.58-3.46(m,3H),3.41-3.35(m,2H),3.23(br d,J=12.0Hz,1H),3.18-3.08(m,2H),3.05-2.93(m,1H),2.56-2.44(m,1H),2.40-2.27(m,1H),2.19(s,3H),2.11-1.99(m,2H),1.42(s,3H);MS(ESI)m/z:736.2[M+H] +
实施例24:化合物24
Figure PCTCN2022075562-appb-000094
步骤A:向化合物23-5(100mg,160.60μmol)和化合物24-1(82.97mg,642.40μmol)的二氯甲烷(10mL)溶液加入
Figure PCTCN2022075562-appb-000095
分子筛(20mg),反应液在25℃搅拌30分钟,向内加入NaBH(OAc) 3(136.15mg,642.40μmol),反应液在25℃下搅拌12小时,通过硅藻土将反应液过滤,将滤液浓缩得到粗品。粗品使用制备级高效液相色谱分离(色谱柱:Waters Xbridge 150*25mm*5μm;流动相:[纯水(碳酸氢钠)-乙腈];乙腈%:40%-70%,7分钟)纯化得到化合物24。 1H NMR(400MHz,CD 3OD)δ=8.95(d,J=2.0Hz,1H),8.75(d,J=8.0Hz,1H),8.17(d,J=2.0Hz,1H),7.91(d,J=7.6Hz,1H),7.51-7.44(m,1H),7.39(q,J=7.6Hz,2H),7.32-7.27(m,1H),7.22(d,J=7.6Hz,1H),7.04(d,J=7.2Hz,1H),6.90-6.52(m,1H),5.10(br t,J=8.0Hz,2H),4.70-4.50(m,3H),4.46-4.35(m,3H),4.10(s,3H),3.80-3.67(m,1H),3.59-3.46(m,1H),3.19-3.06(m,1H),3.04-2.93(m,2H),2.93-2.82(m,2H),2.68-2.58(m,2H),2.42(br dd,J=4.4,7.6Hz,1H),2.25-2.14(m,4H),2.05(s,3H),2.02-1.92(m,1H),1.85-1.71(m,1H);MS(ESI)m/z:736.3 [M+H] +
实施例25:化合物25
Figure PCTCN2022075562-appb-000096
步骤A:化合物10-3(40mg,62.11μmol)和化合物25-1(27.05mg,186.33μmol)以及
Figure PCTCN2022075562-appb-000097
分子筛(40mg)的5mL二氯甲烷溶液搅拌0.5小时,向内加入NaBH(OAc) 3(65.81mg,310.55μmol)。反应体系在15℃下搅拌1.5小时后加入2mL水,过滤,滤液浓缩得到粗品,粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:14%-44%,10分钟)纯化得到化合物25。 1H NMR(400MHz,CD 3OD)δ=9.00(d,J=2.0Hz,1H),8.84-8.74(m,1H),8.55-8.46(m,1H),8.30(d,J=2.0Hz,1H),7.74-7.66(m,1H),7.62-7.54(m,1H),7.53-7.47(m,1H),7.47-7.38(m,1H),7.13-7.04(m,1H),6.89-6.57(m,1H),5.15-5.06(m,2H),4.54-4.44(m,3H),4.38-4.25(m,2H),4.16-4.08(m,3H),3.29(br d,J=6.8Hz,3H),3.22-2.96(m,5H),2.77-2.67(m,1H),2.34-2.23(m,1H),2.00-1.89(m,1H),1.80-1.69(m,2H),1.43-1.34(m,1H),0.99(t,J=6.4Hz,6H);MS(ESI)m/z:773.1[M+H] +
实施例26:化合物26
Figure PCTCN2022075562-appb-000098
步骤A:化合物10-3(40mg,62.11μmol)和化合物26-1(27.05mg,186.33μmol)以及
Figure PCTCN2022075562-appb-000099
分子筛(40mg)的5mL二氯甲烷溶液搅拌0.5小时,向内加入NaBH(OAc) 3(65.81mg,310.55μmol)。反应体系在15℃下搅拌1.5小时后加入2mL水,过滤,滤液浓缩得到粗品,粗品使用制备级高效液相色谱分离(色谱柱:Phenomenex luna C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:14%-44%,10分 钟)纯化得到化合物26。 1H NMR(400MHz,CD 3OD)δ=8.94(d,J=2.0Hz,1H),8.77(d,J=8.0Hz,1H),8.50(s,1H),8.19(d,J=1.6Hz,1H),7.69(dd,J=1.6,7.6Hz,1H),7.56(t,J=7.6Hz,1H),7.51-7.46(m,1H),7.40(t,J=8.0Hz,1H),7.07(d,J=7.6Hz,1H),6.87-6.53(m,1H),5.06(br t,J=8.0Hz,2H),4.48-4.41(m,3H),4.12(s,3H),4.09-3.97(m,2H),3.30-3.20(m,2H),3.17-3.04(m,2H),3.04-2.92(m,2H),2.82-2.64(m,3H),2.23(qd,J=7.2,14.1Hz,1H),1.88-1.68(m,3H),1.43-1.26(m,1H),0.98(t,J=6.4Hz,6H);MS(ESI)m/z:773.1[M+H] +
实施例27:化合物27
Figure PCTCN2022075562-appb-000100
步骤H:0℃时向化合物27-1(10g,53.59mmol)的20mL醋酸溶液中滴加硝酸(9.16g,145.30mmol)的20mL醋酸溶液,反应体系在25℃下搅拌2小时后加入50mL水稀释,过滤,滤饼用50mL水洗涤后干燥得到粗品化合物27-2。
步骤I:向化合物27-2(3g,12.95mmol)的30mL甲醇和6mL水的混合溶液中加入保险粉(13.53g, 77.72mmol),反应体系在70℃下搅拌12小时,过滤,滤饼用100mL甲醇洗涤,滤液浓缩得到粗品。粗品通过硅胶柱层析分离(二氯甲烷:甲醇=1:0-4:1,v/v)纯化得到化合物27-3。MS(ESI)m/z:202.1[M+H] +
步骤J:化合物27-3(4.2g,20.83mmol)和化合物27-4(3.94g,19.79mmol)的100mL乙醇溶液在25℃下搅拌1小时后浓缩,然后溶解在100mL二氯甲烷溶液中,加入二氯二氰基苯醌(4.49g,19.79mmol),反应体系在25℃下搅拌12小时,然后加入100mL饱和亚硫酸钠溶液淬灭后用二氯甲烷萃取(80mL×3),合并有机相后用饱和食盐水洗涤(50mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:二氯甲烷1:0-0:1,v/v)纯化得到化合物27-5。MS(ESI)m/z:381.7[M+H] +
步骤K:在-78℃时向化合物27-5(7.62g,20.02mmol)的120mL二氯甲烷溶液中滴加DIBAL-H的甲苯溶液(1mol/L,40.04mL),反应体系在25℃下搅拌2小时,然后加入200mL饱和酒石酸钾钠溶液淬灭后用二氯甲烷萃取(100mL×3),合并有机相后用饱和食盐水洗涤(50mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(二氯甲烷:甲醇=1:0-4:1,v/v)纯化得到化合物27-6。MS(ESI)m/z:354.0[M+H] +
步骤L:化合物4-1(2.5g,4.78mmol),化合物27-6(1.68g,4.78mmol),碳酸钠(1.27g,11.94mmol)和Pd(PPh 3) 4(551.97mg,477.66μmol)的二氧六环(15mL)和水(3mL)的混合溶液用氮气置换三次并在氮气保护下加热到100℃,搅拌12小时。反应液加入30mL水稀释后通过硅藻土过滤,滤液用二氯甲烷萃取(50mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(二氯甲烷:甲醇=1:0-3:1,v/v)纯化得到化合物27-7。MS(ESI)m/z:669.3[M+H] +
步骤M:化合物27-7(2.7g,4.04mmol),亚铁氰化钾(1.70g,4.04mmol),乙酸钾(79.20mg,807.03μmol)和t-BuXPhos-Pd-G3(320.54mg,403.52μmol)的二氧六环(30mL)和水(30mL)的混合溶液用氮气置换三次并在氮气保护下加热到100℃,搅拌3小时。反应液加入20mL水稀释后通过硅藻土过滤,滤液用二氯甲烷萃取(50mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(二氯甲烷:甲醇=1:0-3:1,v/v)纯化得到化合物27-8。MS(ESI)m/z:660.0[M+H] +
步骤N:向化合物27-8(2.4g,3.64mmol)的40mL二氯甲烷溶液中加入二氧化锰(6.33g,72.76mmol),反应体系在50℃下搅拌12小时,反应液通过硅藻土过滤,滤饼用500mL二氯甲烷洗涤,滤液浓缩得到粗品化合物27-9。MS(ESI)m/z:658.3[M+H] +
步骤O:化合物27-9(2.33g,3.54mmol),化合物5-7(815.77mg,7.09mmol)和三乙胺(717.00mg,7.09mmol)的30mL二氯甲烷溶液在45℃下搅拌1小时,冷却到室温,然后加入NaBH(OAc) 3(1.5g,7.09mmol)和醋酸(319.12mg,5.31mmol),在25℃下搅拌2小时后浓缩得到粗品。粗品先通过硅胶柱层析分离(二氯甲烷:甲醇=1:0-3:1,v/v)纯化,然后使用制备级高效液相色谱分离(色谱柱信息:Phenomenex Synergi Max-RP 250*50mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:15%-45%,20分钟)纯化得到化合物27。 1H NMR(400MHz,DMSO-d 6)δ=8.90(s,1H),8.66(d,J=8.4Hz,1H),8.19-8.07(m,3H),7.87(s,1H),7.59-7.53(m,1H),7.53-7.49(m,1H),7.42(t,J=7.6Hz,1H),7.10-6.74(m, 2H),4.97(t,J=8.4Hz,2H),4.23(s,1H),3.94-3.73(m,7H),3.09-2.82(m,4H),2.81-2.63(m,4H),2.52(s,3H),2.12-1.86(m,3H),1.61-1.58(m,1H);MS(ESI)m/z:757.3[M+H] +
实施例28:化合物28
Figure PCTCN2022075562-appb-000101
步骤A:化合物27-3(3.0g,14.88mmol)和化合物28-1(3.10g,14.14mmol)的60mL乙醇溶液在25℃下搅拌1小时后浓缩,然后溶解在60mL二氯甲烷溶液中,加入二氯二氰基苯醌(3.21g,14.14mmol),反应体系在25℃下搅拌12小时,然后加入30mL饱和亚硫酸钠溶液淬灭后用二氯甲烷萃取(40mL×3),合并有机相后用饱和食盐水洗涤(20mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:二氯甲烷1:0-0:1,v/v)纯化得到化合物28-2。MS(ESI)m/z:401.7[M+H] +
步骤B:在-78℃时向化合物28-2(1.4g,3.49mmol)的30mL二氯甲烷溶液中滴加DIBAL-H的甲 苯溶液(1mol/L,7.68mL),反应体系在25℃下搅拌2小时,然后加入50mL饱和酒石酸钾钠溶液淬灭后用二氯甲烷萃取(40mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:乙酸乙酯=1:0-0:1,v/v)纯化得到化合物28-3。MS(ESI)m/z:373.7[M+H] +
步骤C:化合物28-3(570.18mg,1.53mmol),化合物4-1(0.8g,1.53mmol),碳酸钠(405.02mg,3.82mmol)和Pd(PPh 3) 4(176.63mg,152.85μmol)的二氧六环(15mL)和水(3mL)的混合溶液用氮气置换三次并在氮气保护下加热到100℃,搅拌12小时。反应液加入20mL水稀释后通过硅藻土过滤,滤液用二氯甲烷萃取(40mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(二氯甲烷:甲醇=1:0-3:1,v/v)纯化得到化合物28-4。MS(ESI)m/z:688.9[M+H] +
步骤D:化合物28-4(0.78g,1.13mmol),亚铁氰化钾(477.80mg,1.13mmol),乙酸钾(22.20mg,226.24μmol)和t-BuXPhos-Pd-G3(89.86mg,113.12μmol)的二氧六环(10mL)和水(10mL)的混合溶液用氮气置换三次并在氮气保护下加热到100℃,搅拌3小时。反应液加入20mL水稀释后通过硅藻土过滤,滤液用二氯甲烷萃取(40mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(二氯甲烷:甲醇=1:0-3:1,v/v)纯化得到化合物28-5。MS(ESI)m/z:679.9[M+H] +
步骤E:向化合物28-5(0.72g,1.06mmol)的30mL二氯甲烷溶液中加入二氧化锰(1.84g,21.17mmol),反应体系在50℃下搅拌12小时,反应液通过硅藻土过滤,滤饼用400mL二氯甲烷洗涤,滤液浓缩得到粗品化合物28-6。MS(ESI)m/z:677.9[M+H] +
步骤F:化合物28-6(0.2g,294.95μmol),化合物5-7(67.91mg,589.90mmol)和三乙胺(59.69mg,589.90μmol)的15mL二氯甲烷溶液在45℃下搅拌1小时,冷却到室温,然后加入NaBH(OAc) 3(125.02mg,589.90μmol)和醋酸(26.57mg,442.42μmol),在25℃下搅拌12小时后浓缩得到粗品。粗品先通过制备薄层层析硅胶板分离(二氯甲烷:甲醇=10:1)纯化,然后使用制备级高效液相色谱分离(色谱柱信息:column:Phenomenex Synergi C18 150*25mm*10μm;流动相:[纯水(0.225%甲酸)-乙腈];乙腈%:16%-36%,10分钟)纯化得到化合物28。 1H NMR(400MHz,DMSO-d 6)δ=8.90(d,J=1.6Hz,1H),8.66(d,J=8.4Hz,1H),8.19(t,J=4.8Hz,1H),8.16-8.12(m,2H),7.92(s,1H),7.71(d,J=4.8Hz,2H),7.43(t,J=8.0Hz,1H),7.12-6.77(m,2H),4.98(t,J=8.8Hz,2H),4.27-4.16(m,1H),3.90-3.71(m,6H),3.05(t,J=8.0Hz,2H),2.99-2.87(m,1H),2.76-2.63(m,4H),2.41(dd,J=3.6,9.6Hz,2H),2.09-1.93(m,3H),1.64-1.51(m,1H);MS(ESI)m/z:777.3[M+H] +
实施例29:化合物29
Figure PCTCN2022075562-appb-000102
Figure PCTCN2022075562-appb-000103
步骤A:向100mL甲醇溶液中加入化合物29-1(5g,23.09mmol),铁粉(12.89g,230.82mmol)和氯化铵的饱和溶液(5mL),在75℃下反应2小时。反应液冷却至室温后通过硅藻土过滤,滤液浓缩后加入20mL水并用乙酸乙酯萃取(20mL×2),合并有机相后用饱和食盐水洗涤(20mL),无水硫酸钠干燥,过滤后浓缩得到粗品化合物29-2。MS(ESI)m/z:186.9[M+H] +
步骤B:向化合物29-3(16.83g,78.24mmol)中加入100mL二氯甲烷溶液,在0℃下加入草酰氯(24.83g,195.61mmol),然后在25℃下加入N,N-二甲基甲酰胺(285.96mg,3.91mmol)并在25℃和氮气保护下反应0.5小时。将反应液在真空中浓缩后加入100mL二氯甲烷溶液,然后在0℃下加入化合物29-2(7.3g,39.12mmol)的二氯甲烷溶液(100mL)和吡啶(15.47g,195.61mmol),在25℃和氮气保护下反应0.5小时。向反应液中加入200mL水并用二氯甲烷萃取(200mL×2),合并有机相在真空中浓缩得到粗品,然后加入200mL石油醚和50mL乙酸乙酯,室温搅拌0.5小时,过滤并将滤饼在真空中干燥后得到化合物29-4。MS(ESI)m/z:384.7[M+H] +
步骤C:向化合物29-4(2.3g,6.00mmol)的N,N-二甲基甲酰胺(40mL)溶液中加入碳酸钠(635.45mg,6.00mmol),在140℃搅拌12小时。反应液冷却后加入300mL乙酸乙酯,用水(200mL×2)和食盐水(100mL)洗涤,无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析(石油醚:乙酸乙酯=10:1,v/v)纯化得到化合物29-5。MS(ESI)m/z:348.7[M+H] +
步骤D:在零下78℃和氮气保护下,向化合物29-5(0.8g,2.30mmol)的二氯甲烷(30mL)溶液中缓慢滴加DIBAL-H的甲苯溶液(1mol/L,4.61mL),反应液在零下78℃搅拌1小时。向反应液中缓慢加入20mL饱和的硫酸钠溶液,待恢复至室温后搅拌0.5小时然后通过硅藻土过滤,滤液用二氯甲烷萃取(50mL×2),合并有机相后用饱和食盐水洗涤(50mL),无水硫酸钠干燥,过滤后浓缩得到粗品,粗品通过硅胶柱层析(二氯甲烷/甲醇=10/1,v/v)纯化得到化合物29-6。MS(ESI)m/z:319.0[M+H] +
步骤E:向化合物29-6(0.4g,1.25mmol)的二氧六环(20mL)和水(4mL)的溶液中加入化合物4-1 (655.96mg,1.25mmol),Pd(PPh 3) 4(144.83mg,125.33μmol)和碳酸钠(332.09mg,3.13mmol),在100℃和氮气保护下搅拌4小时,反应液冷却后直接浓缩得到粗品。粗品通过硅胶柱层析(二氯甲烷:甲醇=10:1,v/v)纯化得到化合物29-7。MS(ESI)m/z:636.3[M+H] +
步骤F:向化合物29-7(0.6g,943.90μmol)的二氯甲烷(20mL)溶液中加入二氧化锰(1.64g,18.88mmol),在45℃搅拌48小时。反应液冷却后通过硅藻土过滤,滤液浓缩后得到化合物29-8。MS(ESI)m/z:634.0[M+H] +
步骤G:向化合物29-8(0.2g,315.23μmol)的二氯甲烷溶液(10mL)中加入化合物5-7(72.68mg,631.27μmol)和三乙胺(70.27mg,694.39μmol),反应液在45℃和氮气保护下搅拌1小时。反应液冷却至室温后加入NaBH(OAc) 3(147.17mg,694.39μmol)和醋酸(30.33mg,505.01μmol),反应液在25℃和氮气保护下搅拌12小时。反应液在真空中浓缩得到粗品,粗品化合物通过硅胶板(二氯甲烷/甲醇=10/1,v/v)纯化,再使用制备级高效液相色谱分离(色谱柱信息:Waters Xbridge 150*25mm*5μm;流动相:[纯水(氨水)-乙腈];乙腈%:31%-61%,9分钟)纯化得到化合物29。 1H NMR(400MHz,DMSO-d 6)δ=12.07-11.56(m,1H),11.53-11.13(m,1H),9.19(br s,1H),8.70(d,J=8.4Hz,1H),8.66-8.58(m,3H),8.21(dd,J=1.6,7.7Hz,1H),7.63-7.51(m,2H),7.45(t,J=8.0Hz,1H),7.15-6.81(m,2H),5.68-5.38(m,1H),5.00(br t,J=8.8Hz,2H),4.78-4.64(m,2H),4.60(br s,2H),4.45(br s,1H),3.66-3.47(m,4H),3.17(br s,4H),3.10-3.02(m,1H),2.99-2.87(m,1H),2.55(s,3H),2.37-1.80(m,4H);MS(ESI)m/z:733.2[M+H] +
实施例30:化合物30
Figure PCTCN2022075562-appb-000104
步骤A:向化合物29-8(60mg,94.69μmol)的二氯甲烷(10mL)溶液中加入化合物23-6(24.46mg,189.38μmol)和
Figure PCTCN2022075562-appb-000105
分子筛(20mg),在25℃搅拌0.5小时,然后加入醋酸硼氢化钠(60.21mg,284.07μmol)继续在25℃搅拌1小时,反应液通过硅藻土过滤,滤液浓缩后得到粗品,粗品化合物通过硅胶板(二氯甲烷:甲醇=10:1,v/v)纯化得到粗品,粗品使用制备级高效液相色谱分离(色谱柱信息:Waters Xbridge 150*25mm*5μm;流动相:[纯水(碳酸氢铵)-乙腈];乙腈%:33%-63%,8分钟)纯化得到化合物30。 1H NMR(400MHz,DMSO-d 6)δ=8.91(d,J=2.0Hz,1H),8.67(d,J=8.4Hz,1H),8.34(d,J=2.0Hz,1H),8.24-8.13(m,3H),7.59-7.48(m,2H),7.43(t,J=8.0Hz,1H),7.09-6.77(m,2H),4.99(br t,J=8.8Hz,2H),4.27-4.17(m,1H),3.92-3.82(m,2H),3.81-3.71(m,2H),3.10-2.96(m,1H),2.96-2.90(m,2H),2.74(dd,J=6.0,9.6Hz,1H),2.69(br d,J=7.2Hz,1H),2.65-2.57(m,2H),2.54(s,3H),2.49-2.46(m, 1H),2.42(dd,J=3.6,9.6Hz,1H),2.35-2.32(m,1H),2.32-2.25(m,1H),2.07-1.97(m,1H),1.63-1.54(m,2H),1.26(s,3H);MS(ESI)m/z:747.4[M+H] +
实施例31:化合物31
Figure PCTCN2022075562-appb-000106
步骤A:在圆底烧瓶中加入化合物29-4(1g,2.61mmol),五硫化二磷(1.16g,5.21mmol),吡啶(20mL)和二甲苯(80mL)并在140℃下反应12小时。将溶剂减压浓缩后硅胶柱层析分离(乙酸乙酯/石油醚=0%~20%,v/v)得到化合物31-1.
步骤B:向圆底烧瓶中加入化合物31-1(189mg,520.33μmol)和二氯甲烷(10mL),冷却到-78℃,滴加DIBAL-H的甲苯溶液(1mol/L,1.04mL),使温度不超过-65℃,加完后在-78℃下搅拌3小时。搅拌下小心加入饱和硫酸钠溶液(10mL),过滤,滤液浓缩得到化合物31-2,直接用于下一步。
步骤C:化合物31-2(80mg,238.65μmol,1eq),化合物4-1(124.91mg,238.65μmol),碳酸钠(63.24 mg,596.63μmol),和Pd(PPh 3) 4(27.58mg,23.87μmol)的二氧六环(10mL)和水(2mL)溶液氮气保护下加热到100℃,搅拌12小时。反应液过滤后浓缩得到粗品。粗品通过制备薄层层析硅胶板分离(甲醇/二氯甲烷=0%~10%,v/v)纯化得到化合物31-3。
步骤D:向化合物31-3(110mg,168.78μmol)的二氯甲烷(10mL)溶液中加入活性二氧化锰(293.48mg,3.38mmol)并在45℃下搅拌48小时。过滤,滤液浓缩得到化合物31-4,直接用于下一步。
步骤E:向化合物31-4(80mg,123.13μmol),化合物1-22(28.35mg,246.26μmol),二氯甲烷(10mL)中加入三乙胺(37.38mg,369.39μmol),在45℃下搅拌1小时,然后在25℃下加入NaBH(OAc) 3(78.29mg,369.39μmol)和醋酸(14.79mg,246.26μmol)并继续搅拌12小时。该混合物减压浓缩后,通过制备级硅胶薄层色谱(甲醇/二氯甲烷=1/8)初步分离,并进一步使用制备级高效液相色谱(column:Phenomenex Synergi C18 150*25mm*10μm;流动相:[纯水(甲酸)-乙腈];乙腈%:13%-43%,10分钟)分离纯化得到化合物31. 1H NMR(400MHz,DMSO-d6)δ=11.89-11.52(m,1H),11.43-11.13(m,1H),9.19(br d,J=16.0Hz,1H),8.92-8.76(m,2H),8.75-8.55(m,2H),7.85(d,J=7.2Hz,1H),7.61-7.36(m,3H),7.16-6.79(m,2H),5.78-5.29(m,1H),5.00(br t,J=8.0Hz,2H),4.78-4.56(m,4H),4.52-4.39(m,1H),3.69-3.56(m,4H),3.28-3.17(m,4H),3.13-3.03(m,1H),3.01-2.90(m,1H),2.39(s,3H),2.35-2.16(m,2H),2.11-1.83(m,2H);MS(ESI)m/z:749.3[M+H] +
实施例32:化合物32
Figure PCTCN2022075562-appb-000107
Figure PCTCN2022075562-appb-000108
步骤A:0℃时向化合物32-1(10g,53.59mmol)的20mL醋酸溶液中滴加硝酸(9.16g,145.30mmol)的20mL醋酸溶液,反应体系在25℃下搅拌2小时后加入50mL水稀释,过滤,滤饼用50mL水洗涤后浓缩得到粗品化合物32-2。
步骤B:向化合物32-2(3g,12.95mmol)的30mL甲醇和6mL水的混合溶液中加入保险粉(13.53g,77.72mmol),反应体系在70℃下搅拌12小时,过滤,滤饼用100mL甲醇洗涤,滤液浓缩得到粗品。粗品通过硅胶柱层析分离(二氯甲烷:甲醇=1:0-4:1,v/v)纯化得到化合物32-3。MS(ESI)m/z:202.1[M+H] +
步骤C:化合物32-3(4.2g,20.83mmol)和化合物32-4(3.94g,19.79mmol)的100mL乙醇溶液在25℃下搅拌1小时后浓缩,然后溶解在100mL二氯甲烷溶液中,加入二氯二氰基苯醌(4.49g,19.79mmol),反应体系在25℃下搅拌12小时,然后加入100mL饱和亚硫酸钠溶液淬灭后用二氯甲烷萃取(80mL×3),合并有机相后用饱和食盐水洗涤(50mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(石油醚:二氯甲烷1:0-0:1,v/v)纯化得到化合物32-5。MS(ESI)m/z:381.7[M+H] +
步骤D:在-78℃时向化合物32-5(7.62g,20.02mmol)的120mL二氯甲烷溶液中滴加DIBAL-H的甲苯溶液(1mol/L,40.04mL),反应体系在25℃下搅拌2小时,然后加入200mL饱和酒石酸钾钠溶液淬灭后用二氯甲烷萃取(100mL×3),合并有机相后用饱和食盐水洗涤(50mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱层析分离(二氯甲烷:甲醇=1:0-4:1,v/v)纯化得到化合物32-6。MS(ESI)m/z:354.0[M+H] +
步骤E:氮气保护下,向化合物8-1(1.11g,2.20mmol)和双联频哪醇硼酸酯(1.12g,4.40mmol)的30mL二氧六环溶液中加入Pd(dppf)Cl2·CH2Cl2(179.74mg,220.10μmol)和乙酸钾(648.01mg,6.60mmol),反应体系在100℃下搅拌1小时。冷却后通过硅藻土过滤,滤饼用200mL的二氯甲烷洗涤,滤液浓缩得到粗品。粗品通过硅胶柱分离(二氯甲烷:甲醇=1:0-5:1,v/v)纯化得到化合物32-7。MS(ESI)m/z:552.2[M+H] +
步骤F:氮气保护下,向化合物32-7(0.4g,725.44μmol),化合物32-6(255.80mg,725.44μmol)的 二氧六环(10mL)和水(2mL)的混合溶液中加入碳酸钠(192.22mg,1.81mmol)和Pd(PPh 3) 4(83.83mg,72.54umol),用氮气置换三次并在氮气保护下加热到100℃,搅拌1小时。反应液浓缩得到粗品。粗品通过硅胶柱分离(二氯甲烷:甲醇=1:0-3:1,v/v)纯化得到化合物32-8。MS(ESI)m/z:697.0[M+H] +
步骤G:化合物32-8(0.515g,738.74μmol),亚铁氰化钾(468.06mg,1.11mmol),乙酸钾(14.50mg,147.75μmol)和t-BuXPhos-Pd-G3(58.68mg,73.87μmol)的二氧六环(15mL)和水(5mL)的混合溶液用氮气置换三次并在氮气保护下加热到100℃,搅拌3小时。反应液浓缩得到粗品。粗品通过硅胶柱分离(二氯甲烷:甲醇=1:0-3:1,v/v)纯化得到化合物32-9。MS(ESI)m/z:688.0[M+H] +
步骤H:向化合物32-9(0.435g,632.55μmol)的15mL二甲基亚砜溶液中加入IBX(354.25g,1.27mmol),反应体系在25℃下搅拌12小时,向反应液中缓慢加入20mL的硫代硫酸钠饱和溶液,用二氯甲烷萃取(30mL×5),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品,粗品通过硅胶柱分离(二氯甲烷:甲醇=1:0-3:1,v/v)纯化得到化合物32-10。MS(ESI)m/z:686.1[M+H] +
步骤I:化合物32-10(0.15g,218.76μmol)和化合物1-10(38.12mg,437.52μmol)的5mL二氯甲烷溶液在25℃下搅拌0.5小时后,加入NaBH(OAc) 3(139.09mg,656.29μmol),反应体系在25℃下搅拌12小时。反应液浓缩得到粗品,粗品先通过硅胶柱层析法分离(二氯甲烷:甲醇=1:0-2:1,v/v)纯化,然后使用制备级高效液相色谱分离(色谱柱信息:Phenomenex Synergi C18 150*25mm*10μm;流动相:[纯水(甲酸)-乙腈];乙腈%:11%-41%,10分钟)纯化得到化合物32。 1H NMR(400MHz,DMSO-d6)δ=8.91(d,J=2.0Hz,1H),8.68(d,J=8.0Hz,1H),8.21-8.12(m,3H),7.90(d,J=1.2Hz,1H),7.61-7.55(m,1H),7.55-7.51(m,1H),7.44(t,J=8.0Hz,1H),7.09-6.78(m,2H),5.00(br t,J=8.4Hz,2H),4.22(tt,J=3.2,6.6Hz,1H),3.90-3.81(m,3H),3.77(br d,J=12.0Hz,2H),3.03-2.92(m,3H),2.80-2.70(m,3H),2.69-2.65(m,1H),2.60(br t,J=6.8Hz,2H),2.54(br s,3H),2.40(br dd,J=3.2,9.6Hz,1H),2.07-1.95(m,3H),1.67-1.49(m,1H);MS(ESI)m/z:757.0[M+H] +
实施例33:化合物33
Figure PCTCN2022075562-appb-000109
Figure PCTCN2022075562-appb-000110
步骤A:化合物3-8(1.0g,2.47mmol)和化合物5-7(568.27mg,4.94mmol)的20mL二氯甲烷溶液在25℃下搅拌0.5小时后,加入NaBH(OAc) 3(1.57g,7.40mmol),反应体系在25℃下搅拌1小时。加入30mL水稀释,用二氯甲烷萃取(40mL×3),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品。粗品通过硅胶柱分离(二氯甲烷:甲醇=1:0-10:1,v/v)纯化得到化合物33-1。MS(ESI)m/z:504.2[M+H] +
步骤B:氮气保护下,向化合物33-1(1.11g,2.20mmol)和双联频哪醇硼酸酯(1.12g,4.40mmol)的30mL二氧六环溶液中加入Pd(dppf)Cl 2·CH 2Cl 2(179.74mg,220.10μmol)和乙酸钾(648.01mg,6.60mmol),反应体系在100℃下搅拌1小时。冷却后通过硅藻土过滤,滤饼用200mL的二氯甲烷洗涤,滤液浓缩得到粗品。粗品通过硅胶柱分离(二氯甲烷:甲醇=1:0-5:1,v/v)纯化得到化合物33-2。MS(ESI)m/z:552.2[M+H] +
步骤C:氮气保护下,向化合物33-2(0.4g,725.44μmol),化合物27-6(255.80mg,725.44μmol)的二氧六环(10mL)和水(2mL)的混合溶液中加入碳酸钠(192.22mg,1.81mmol)和Pd(PPh 3) 4(83.83mg,72.54umol),用氮气置换三次并在氮气保护下加热到100℃,搅拌1小时。反应液浓缩得到粗品。粗品通过硅胶柱分离(二氯甲烷:甲醇=1:0-3:1,v/v)纯化得到化合物33-3。MS(ESI)m/z:697.0[M+H] +
步骤D:化合物33-3(0.515g,738.74μmol),亚铁氰化钾(468.06mg,1.11mmol),乙酸钾(14.50mg,147.75μmol)和t-BuXPhos-Pd-G3(58.68mg,73.87μmol)的二氧六环(15mL)和水(5mL)的混合溶液用氮气置换三次并在氮气保护下加热到100℃,搅拌3小时。反应液浓缩得到粗品。粗品通过硅胶柱分离(二氯甲烷:甲醇=1:0-3:1,v/v)纯化得到化合物33-4。MS(ESI)m/z:688.0[M+H] +
步骤E:向化合物33-4(0.435g,632.55μmol)的15mL二甲基亚砜溶液中加入IBX(354.25g,1.27mmol),反应体系在25℃下搅拌12小时,向反应液中缓慢加入20mL的硫代硫酸钠饱和溶液,用二氯甲烷萃取(30mL×5),合并有机相后用饱和食盐水洗涤(30mL×3),无水硫酸钠干燥,过滤后浓缩得到粗品,粗品通过硅胶柱分离(二氯甲烷:甲醇=1:0-3:1,v/v)纯化得到化合物33-5。MS(ESI)m/z:686.1[M+H] +
步骤F:化合物33-5(0.15g,218.76μmol)和化合物33-6(44.25mg,437.52μmol)的5mL二氯甲烷溶液在25℃下搅拌0.5小时后,加入NaBH(OAc) 3(139.09mg,656.29μmol),反应体系在25℃下搅拌12小时。反应液浓缩得到粗品,粗品先通过硅胶柱分离(二氯甲烷:甲醇=1:0-2:1,v/v)纯化,然后使用制备级高效液相色谱分离(色谱柱信息:Phenomenex Synergi C18 150*25mm*10μm;流动相:[纯水(甲酸)- 乙腈];乙腈%:11%-41%,10分钟)纯化得到化合物33。 1H NMR(400MHz,DMSO-d 6)δ=8.91(d,J=2.0Hz,1H),8.68(d,J=8.0Hz,1H),8.23-8.12(m,3H),7.93(d,J=1.2Hz,1H),7.64-7.50(m,2H),7.44(t,J=8.0Hz,1H),7.12-6.68(m,2H),5.00(br t,J=8.8Hz,2H),3.93-3.80(m,5H),3.03-2.94(m,3H),2.81-2.77(m,1H),2.76-2.69(m,2H),2.60(br t,J=6.8Hz,3H),2.54(br s,3H),2.06-1.95(m,2H),1.88-1.65(m,2H),1.32-1.19(m,4H);MS(ESI)m/z:771.0[M+H] +
实验例1:PD-L1结合实验
实验材料:
PD1:PD-L1 TR-FRET检测试剂盒购自BPS Biosciences。Nivo多标记分析仪(PerkinElmer)。
实验方法:
使用试剂盒里的缓冲液稀释PD1-Eu、Dye-labeled acceptor(色素标记的受体)、PD-L1-biotin(PD-L1-生化素)和待测化合物。
将待测化合物进行5倍稀释至第8个浓度,即从4μM稀释至0.05nM,DMSO浓度为4%,设置双复孔实验。向微孔板中加入5μL待测化合物各浓度梯度,其中Max信号孔加入5μL含4%DMSO的缓冲液和5μL PD-L1-biotin(PD-L1-生化素)(60nM),Min信号孔只加入5μL缓冲液,25度孵育20分钟。结束孵育后每孔加入5μL稀释后PD1-Eu(10nM)和5μL稀释后的Dye-labeled acceptor(色素标记的受体)。反应体系置于25度反应90分钟。反应结束后,采用多标记分析仪读取时间分辨荧光分析信号。
数据分析:
利用方程式(样品-Min)/(Max-Min)*100%将原始数据换算成抑制率,IC 50的值即可通过四参数进行曲线拟合得出(GraphPad Prism中log(inhibitor)vs.response--Variable slope模式得出)。表1提供了本发明的化合物对PD1/PD-L1结合的抑制活性。
表1
受试化合物 PD1/PD-L1结合IC 50(nM)
1 1.95
实验结论:本发明化合物在酶水平具有较好的PD1/PD-L1结合抑制活性。
实验例2:NFAT结合实验
实验材料:
PD1:PD-L1 TR-FRET检测试剂盒购自BPS Biosciences。Birght-Glo试剂购自Promega。Nivo多标记分析仪(PerkinElmer)。
实验方法:
将生长汇合度达到80%的TCR Activitor/PD-L1 CHO细胞按照每孔35000个细胞铺到板子里面然后放入37℃细胞培养箱中过夜;将待测化合物用排枪进行5倍稀释至第8个浓度,即从100微摩尔稀释至1.28纳摩尔,DMSO浓度为100%,设置双复孔实验。向中间板中加入147μL培养基,再按照对应 位置,转移3μL每孔的梯度稀释化合物至中间板,此时化合物浓度为2微摩尔至0.0256纳摩尔,DMSO浓度为2%。弃T细胞受体/PD-L1 CHO细胞上清,每孔加入50μL化合物工作液,37℃孵育30分钟;结束孵育后每孔加入50μL密度为4×105/mL的PD-1/NFAT Reporter-Jurkat细胞悬液,37℃孵育5小时。结束孵育后每孔加入100μL Bright-Glo,混匀后使用Nivo多标分析仪读取化学发光信号。
数据分析:
利用方程式(Sample-Min)/(Max-Min)*100%将原始数据换算成抑制率,IC 50的值即可通过四参数进行曲线拟合得出(GraphPad Prism中log(inhibitor)vs.response--Variable slope模式得出)。利用方程式(Max-Min)/Min折算化合物对于PD-1/PD-L1的抑制程度,倍数越高,代表PD-1/PD-L1通路抑制越强。
实验结果:图1和表2提供了本发明的化合物对PD-1/PD-L1结合的抑制活性。
表2
受试化合物 NFAT细胞IC 50(nM) 相对DMSO激活倍数
1 35 7.81
2 16 5.63
3 4.7 6.74
4 4.5 5.18
7 3.0 5.5
8 3.9 4.69
9 4.4 4.63
11 24.6 4.42
12 10.9 3.54
13 19.0 4.54
14 10.7 3.71
15 35.1 4.36
16 9.2 4.53
17 7.5 4.72
18 3.5 4.23
19 30.7 4.28
20 42.7 4.39
22 22.6 3.62
23 32.6 4.82
24 32.6 4.79
27 6.71 5.12
28 4.67 4.43
29 6.03 4.03
30 14.54 4.13
32 8.98 4.05
33 13.62 3.84
实验结论:本发明化合物在细胞水平可以有效阻断PD-1/PD-L1信号通路,恢复T细胞活性。
实验例3:小鼠体内药代动力学研究
以雄性C57BL/6小鼠为受试动物,应用LC/MS/MS法测定小鼠灌胃给予受试化合物后不同时刻血 浆中的药物浓度。研究受试化合物在小鼠体内的药代动力学行为,评价其药动学特征。
试验动物:健康雄性C57BL/6小鼠。药物配制:IV组溶媒为5%DMSO+95%(20%羟丙基-β环糊精);PO组溶媒为5%DMSO+95%(20%HP-β-CD)。给药:受试化合物的给药剂量为IV为1mg/kg,PO为10mg/kg或30mg/kg(注:PO组禁食一夜后给药)。
实验操作:给药后,收集一定时间的全血,制备得到血浆,以LC-MS/MS方法分析药物浓度,并用Phoenix WinNonlin软件(美国Pharsight公司)计算药代参数。
实验结果:如表3和表4所示。
表3体内PK性质评价结果(一)
Figure PCTCN2022075562-appb-000111
表4体内PK性质评价结果(二)
Figure PCTCN2022075562-appb-000112
实验结论:本发明化合物药代动力学性质优良,在体内具有较长的半衰期,更高的血浆暴露量和生物利用度,具备良好的成药性。
实验例4:基于B-hPD-L1人源化小鼠的MC38-hPD-L1结肠癌动物模型的PD-L1抗体药物药效实验实验方法:
1.细胞培养
小鼠结肠癌MC38细胞购自舜冉上海生物科技有限公司。百奥赛图(北京)医药科技股份有限公司对MC38进行基因改造,使其表达人的PD-L1,命名为MC38-hPD-L1。此细胞为贴壁细胞,培养在37℃、5%CO 2的培养箱中,培养基成分为含有10%灭活胎牛血清的Dulbecco's Modified Eagle's Medium培养基。
2.肿瘤细胞的接种与分组
将PBS重悬的MC38-hPD-L1细胞以5×10 5个/0.1mL/只体积接种于B-hPD-L1小鼠的右侧皮下。当平均 肿瘤体积达到150±50mm 3时,根据小鼠肿瘤体积和体重选择合适的小鼠入组,平均分配到每个实验组中,每组8只。分组当天开始给药,口服组给药溶媒为5%DMSO+95%(20%HP-β-CD溶于水中),腹腔注射组给药溶媒为0.9%氯化钠注射液。具体给药方案见表5:
表5 MC38-hPD-L1实验给药方案
Figure PCTCN2022075562-appb-000113
注:a:给药体积依实验动物体重按10μL/g计算;b.分组后5天内剂量为20mg/kg,从第6天开始剂量调整为50mg/kg;c:p.o.指口服给药,i.p.指腹腔注射给药;d:TIW指每周给药3次,BID指每天给药两次。
3.药物评价指标
肿瘤体积抑制率(TGI TV):TGI TV(%)=[1-(Ti-T0)/(Vi-V0)]×100%(Ti:治疗组在给药第i天的肿瘤体积均值,T0:治疗组在给药第0天的肿瘤体积均值;Vi:溶剂对照组在给药第i天的肿瘤体积均值,V0:溶剂对照组在给药第0天的肿瘤体积均值)
实验结果:
阿特珠单抗(G2组)的TGI TV(%)=88.4%(p<0.0001),化合物4(G4组)的TGI TV(%)=86.1%(p<0.0001),化合物27(G5组)的TGI TV(%)=86.1%(p<0.0001),实验动物荷瘤体积和体重变化分别如图2和图3所示。末次给药4小时后,取各组的肿瘤组织做TILs(肿瘤浸润淋巴细胞分析)检测,TILs检测结果如图4和图5所示。
实验结论:
本发明化合物在小鼠体内可以显著抑制肿瘤PD-L1表达,有效激活免疫,抑制hPD-L1阳性MC38肿瘤生长。
实验例5:人源化的PBMC+A375共孵育模型药效实验
实验目的:评价受试药物在人黑色素瘤A375混合PBMC皮下移植瘤模型中的抗肿瘤作用。
实验设计:
1.细胞培养:
A375细胞培养在含10%胎牛血清(FBS)的DMEM培养液中。收集指数生长期的A375细胞,HBSS重悬至适合浓度用于NCG小鼠皮下肿瘤接种。实验用的A375细胞用Mitomycin C培养,然后PBS洗涤。
2.外周血单个核细胞的复苏及共培养:
购买冻存的PBMC,复苏计数,将得到的PBMC加入经Mitomycin C处理的A375细胞中,PBMC与A375共培养,培养液为含IL-2和10%FBS的RPMI 1640培养液。
3.肿瘤细胞接种:
PBMC与A375共培养后,收取PBMC与新鲜消化下来的A375细胞,接种于NCG小鼠右侧皮下。 之后根据小鼠体重随机进行分组给药。
4.评估指标:
通过肿瘤体积和肿瘤生长抑制率TGI(%)来检测和评价受试药物在人黑色素瘤A375混合PBMC体内移植瘤生长的抑制作用或完全治愈能力。

Claims (19)

  1. 式(I)化合物或其药学上可接受的盐
    Figure PCTCN2022075562-appb-100001
    其中,
    环A选自
    Figure PCTCN2022075562-appb-100002
    L 1和L 2分别独立地选自-CH 2-和-CH 2-NH-CH 2-;
    Z和E分别独立地选自CH和N;
    Z 1选自O和S;
    Z 2选自N和CR 9
    X选自N和CR 14
    Y选自N和CR 15
    R 1选自H、CH 3和CHF 2
    R 2选自CH 3和Cl;
    R 3、R 4、R 5和R 6分别独立地选自H、C 1-6烷基、OH、COOH和-C 1-3烷基-COOH;
    或者,R 3、R 4连同它们所连接的原子一起形成氮杂环丁烷基、吡咯烷基、恶唑烷基或哌啶基,所述氮杂环丁烷基、吡咯烷基、恶唑烷基和哌啶基分别独立任选被1、2或3个R 16取代;
    或者,R 5、R 6连同它们所连接的原子一起形成氮杂环丁烷基、吡咯烷基、恶唑烷基或哌啶基,所述氮杂环丁烷基、吡咯烷基、恶唑烷基和哌啶基分别独立任选被1、2或3个R 16取代;
    R 7选自H、F、Cl、CH 3和CHF 2
    R 8选自-OCH 3、-O-CH 2-F、和-O-CH 2-CN;
    R 9选自H、F和CN;
    R 14和R 15分别独立地选自H和C 1-6烷基;
    R 16分别独立地选自H、C 1-6烷基、OH、=O、COOH和-C 1-3烷基-COOH。
  2. 根据权利要求1所述的化合物或其药学上可接受的盐,其中,化合物选自
    Figure PCTCN2022075562-appb-100003
    其中,
    环B和环C分别独立地选自氮杂环丁烷基、吡咯烷基、恶唑烷基和哌啶基;
    R 10、R 11、R 12和R 13分别独立地选自H、C 1-4烷基、OH、=O、COOH和-C 1-3烷基-COOH;环A、Z、R 1和R 2如权利要求1所定义。
  3. 根据权利要求1所述的化合物或其药学上可接受的盐,其中,化合物选自
    Figure PCTCN2022075562-appb-100004
    其中,
    环B和环C分别独立地选自氮杂环丁烷基、吡咯烷基、恶唑烷基和哌啶基;
    R 10、R 11、R 12和R 13分别独立地选自H、C 1-4烷基、OH、=O、COOH和-C 1-3烷基-COOH;Z 3选自CH 2和O;
    Z、Z 1、Z 2、E、R 1、R 2和R 8如权利要求1所定义。
  4. 据权利要求1或3所述的化合物或其药学上可接受的盐,其中,化合物选自
    Figure PCTCN2022075562-appb-100005
    其中,
    Z 3选自CH 2和O,
    R 10、R 11、R 12和R 13分别独立地选自H、C 1-4烷基、OH、=O、COOH和-C 1-3烷基-COOH,
    Z、Z 1、Z 2、E、R 1、R 2和R 8如权利要求1或3所定义。
  5. 根据权利要求1-4任意一项所述的化合物或其药学上可接受的盐,其中,Z 1选自O,Z 2选自C(CN)。
  6. 根据权利要求1-4任意一项所述的化合物或其药学上可接受的盐,其中,X选自N。
  7. 根据权利要求1或6所述的化合物或其药学上可接受的盐,其中,Y选自N。
  8. 根据权利要求1-4任意一项所述的化合物或其药学上可接受的盐,其中,R 1选自CHF 2
  9. 根据权利要求1所述的化合物或其药学上可接受的盐,其中,R 3、R 4、R 5和R 6分别独立地选自H、CH 3、异丙基、
    Figure PCTCN2022075562-appb-100006
    COOH和-C 1-3烷基-COOH。
  10. 根据权利要求1所述的化合物或其药学上可接受的盐,其中R 7选自H。
  11. 根据权利要求1或2所述的化合物或其药学上可接受的盐,其中,环A选自
    Figure PCTCN2022075562-appb-100007
    Figure PCTCN2022075562-appb-100008
  12. 根据权利要求1所述的化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2022075562-appb-100009
    分别独立地选自
    Figure PCTCN2022075562-appb-100010
    Figure PCTCN2022075562-appb-100011
  13. 根据权利要求1所述的化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2022075562-appb-100012
    分别独立地选自
    Figure PCTCN2022075562-appb-100013
    Figure PCTCN2022075562-appb-100014
  14. 根据权利要求2-4任意一项所述的化合物或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2022075562-appb-100015
    Figure PCTCN2022075562-appb-100016
    分别独立地选自
    Figure PCTCN2022075562-appb-100017
    Figure PCTCN2022075562-appb-100018
  15. 下式化合物或其药学上可接受的盐
    Figure PCTCN2022075562-appb-100019
    Figure PCTCN2022075562-appb-100020
    Figure PCTCN2022075562-appb-100021
    Figure PCTCN2022075562-appb-100022
    Figure PCTCN2022075562-appb-100023
    Figure PCTCN2022075562-appb-100024
  16. 根据权利要求15所述的化合物或其药学上可接受的盐,其中,化合物选自
    Figure PCTCN2022075562-appb-100025
    Figure PCTCN2022075562-appb-100026
    Figure PCTCN2022075562-appb-100027
    Figure PCTCN2022075562-appb-100028
    Figure PCTCN2022075562-appb-100029
    Figure PCTCN2022075562-appb-100030
    Figure PCTCN2022075562-appb-100031
    Figure PCTCN2022075562-appb-100032
  17. 根据权利要求1~16任意一项所述的化合物或其药学上可接受的盐在制备治疗与PD-L1有关的疾病的药物中的应用。
  18. 根据权利要求17所述的应用,其中,所述与PD-L1有关的疾病为肿瘤。
  19. 根据权利要求18所述的应用,其中,所述肿瘤为结肠癌、黑色素瘤、非小细胞肺癌、肝细胞癌或肾细胞癌。
PCT/CN2022/075562 2021-02-08 2022-02-08 吲哚啉类化合物 WO2022166991A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280011224.5A CN116745301A (zh) 2021-02-08 2022-02-08 吲哚啉类化合物

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
CN202110172933 2021-02-08
CN202110172933.9 2021-02-08
CN202110934819.5 2021-08-13
CN202110934819 2021-08-13
CN202111076891.5 2021-09-14
CN202111076891 2021-09-14
CN202111204272 2021-10-15
CN202111204272.X 2021-10-15
CN202111593525.7 2021-12-23
CN202111593525 2021-12-23

Publications (1)

Publication Number Publication Date
WO2022166991A1 true WO2022166991A1 (zh) 2022-08-11

Family

ID=82742015

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/075562 WO2022166991A1 (zh) 2021-02-08 2022-02-08 吲哚啉类化合物

Country Status (2)

Country Link
CN (1) CN116745301A (zh)
WO (1) WO2022166991A1 (zh)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106674209A (zh) * 2016-12-23 2017-05-17 深圳先进技术研究院 程序性死亡受体1基因抑制剂及其制备方法与应用
WO2017118762A1 (en) * 2016-01-08 2017-07-13 Rijksuniversiteit Groningen Inhibitors of the pd-1/pd-l1 protein/protein interaction
CN108774219A (zh) * 2018-05-17 2018-11-09 中国药科大学 抑制pd-1/pd-l1的小分子化合物及其用途
CN108863963A (zh) * 2017-05-08 2018-11-23 南京圣和药业股份有限公司 作为pd-l1抑制剂的杂环类化合物
CN109096219A (zh) * 2017-06-20 2018-12-28 广州丹康医药生物有限公司 一种新型抗pd-l1化合物、其应用及含其的组合物
CN109776377A (zh) * 2019-02-01 2019-05-21 沈阳药科大学 吲哚啉类化合物及其制备方法和应用
CN110128415A (zh) * 2019-05-31 2019-08-16 沈阳药科大学 用作免疫调节剂的吲哚啉类化合物及其制备方法

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017118762A1 (en) * 2016-01-08 2017-07-13 Rijksuniversiteit Groningen Inhibitors of the pd-1/pd-l1 protein/protein interaction
CN106674209A (zh) * 2016-12-23 2017-05-17 深圳先进技术研究院 程序性死亡受体1基因抑制剂及其制备方法与应用
CN108863963A (zh) * 2017-05-08 2018-11-23 南京圣和药业股份有限公司 作为pd-l1抑制剂的杂环类化合物
CN109096219A (zh) * 2017-06-20 2018-12-28 广州丹康医药生物有限公司 一种新型抗pd-l1化合物、其应用及含其的组合物
CN108774219A (zh) * 2018-05-17 2018-11-09 中国药科大学 抑制pd-1/pd-l1的小分子化合物及其用途
CN109776377A (zh) * 2019-02-01 2019-05-21 沈阳药科大学 吲哚啉类化合物及其制备方法和应用
CN110128415A (zh) * 2019-05-31 2019-08-16 沈阳药科大学 用作免疫调节剂的吲哚啉类化合物及其制备方法

Also Published As

Publication number Publication date
CN116745301A (zh) 2023-09-12

Similar Documents

Publication Publication Date Title
JP6877407B2 (ja) Ntrk関連障害の治療に有用な化合物および組成物
WO2022214102A1 (zh) 作为kras g12d抑制剂的杂环化合物
WO1995003308A1 (en) Morphinan derivative and medicinal use
WO2021180235A1 (zh) Zeste增强子同源物2抑制剂及其用途
EP4083038A1 (en) Pyridazinyl thiazolecarboxamide compound
JP2020505457A (ja) 2−アリールスルホンアミド−N−アリールアセトアミド誘導体化Stat3
TW202039474A (zh) 吡唑基-氨基-嘧啶基衍生物的苯甲醯胺及其組合物和方法
WO2018084321A1 (ja) Egfr阻害及び腫瘍治療に有用な新規化合物
CN112745335A (zh) 一种三并杂环化合物及其用途
CN114728967A (zh) 作为jak抑制剂的三并杂环类化合物及其应用
KR20230093300A (ko) 치환된 벤조 또는 피리도피리미딘 아민계 억제제 및 이의 제조 방법과 응용
WO2022002077A1 (zh) 一种芳基并芳杂环衍生物及其制备方法和用途
JP5616628B2 (ja) ピログルタミン酸誘導体の合成および使用
WO2022199670A1 (zh) 6-氨基甲酸酯取代的杂芳环衍生物
CN108779090B (zh) 2环性杂环化合物
CN112955453B (zh) 作为选择性Trk抑制剂的吡唑并嘧啶衍生物
WO2022166991A1 (zh) 吲哚啉类化合物
WO2020169058A1 (zh) Pd-l1拮抗剂化合物
WO2023143147A1 (zh) 一种哒嗪并吡啶酮类化合物、其药物组合物及应用
WO2023280317A1 (zh) 苄氨基三并环类化合物及其应用
WO2022199635A1 (zh) 苄氨基喹唑啉类衍生物
WO2022222911A1 (zh) 嘧啶酮化合物及其用途
TWI652265B (zh) 氮雜吲哚衍生物
JP2023526346A (ja) 三環式化合物を含む組み合わせ及びhbv治療薬物の製造におけるその使用
WO2021233133A1 (zh) 用作ret激酶抑制剂的化合物及其应用

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22749270

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202280011224.5

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22749270

Country of ref document: EP

Kind code of ref document: A1