WO2022152316A1 - 联苯衍生物及其应用 - Google Patents

联苯衍生物及其应用 Download PDF

Info

Publication number
WO2022152316A1
WO2022152316A1 PCT/CN2022/072583 CN2022072583W WO2022152316A1 WO 2022152316 A1 WO2022152316 A1 WO 2022152316A1 CN 2022072583 W CN2022072583 W CN 2022072583W WO 2022152316 A1 WO2022152316 A1 WO 2022152316A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
μmol
mmol
reaction
added
Prior art date
Application number
PCT/CN2022/072583
Other languages
English (en)
French (fr)
Inventor
吴凌云
王才林
徐雄彬
童海骏
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 CN202280010036.0A priority Critical patent/CN116710460A/zh
Publication of WO2022152316A1 publication Critical patent/WO2022152316A1/zh

Links

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
    • 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • 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
    • 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

Definitions

  • the present invention relates to a biphenyl derivative, its preparation method and its application in preparation and treatment of related diseases, in particular to a compound represented by formula (I) and a pharmaceutically acceptable salt thereof.
  • PD-1 Programmed cell death 1
  • CD279 is an important immunosuppressive molecule in the CD28/CTLA-4 receptor family. It is a membrane protein containing 268 amino acid residues and is widely expressed in T.
  • PD-L1 is a protein encoded by the CD274 gene and mainly expressed on the surface of tumor cells, dendritic cells and macrophages.
  • the PD-1/PD-L1 signaling pathway is activated, which in turn inhibits the activation of T cells, causing T cell inactivation and contributing to the immune escape of tumor cells.
  • Another ligand of PD-1, PD-L2 is mainly expressed on the surface of dendritic cells, macrophages and B cells and is associated with inflammation and autoimmune diseases.
  • PD-1 negatively regulates immune responses by binding to its ligand PD-L1 and dephosphorylating multiple key molecules in the TCR signaling pathway.
  • the activation of PD-1/PD-L1 signaling pathway can avoid the surrounding tissue damage caused by excessive immune response, thereby reducing the occurrence of autoimmune diseases.
  • the expressions of PD-1 and PD-L1 are abnormally increased, and tumor cells can successfully escape the recognition and detection of the immune system by the binding of these PD-L1 molecules to PD-1 on T cells. attack.
  • PD-(L)1 mAb can block this "tumor immune escape mechanism" and restore the patient's own immune system's anti-cancer function.
  • mAbs monoclonal antibodies
  • FDA Food and Drug Administration
  • Incyte's PD-L1 small molecule inhibitor INCB86550 (WO2018119263, WO2019191707) and Gilead's PD-L1 small molecule inhibitor GS-4224 (US20180305315, WO2019160882) ) has entered the clinical phase 2, and the small molecule PD-1/PD-L1 inhibitor of BMS benzyl phenyl ether (WO2015034820, WO2015160641) is in the preclinical research stage.
  • small-molecule drugs can cross the cell membrane and act on intracellular targets, they have the advantages of convenient storage and transportation, low production cost, no immunogenicity, and usually oral administration. Therefore, research and development of small molecule blockers of PD-1/PD-L1 has broad application prospects.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof,
  • R 1 and R 2 are each independently selected from H, F, Cl, Br, I, CN and C 1-3 alkyl optionally substituted with 1 , 2 or 3 halogens;
  • R 3 is selected from H, CN, C 1-3 alkyl, C 1-3 alkoxy and C 1-3 alkylamino, the C 1-3 alkyl, C 1-3 alkoxy and C 1- 3 alkylamino groups are each independently optionally substituted with 1, 2 or 3 halogens;
  • R 4 and R 5 are each independently selected from H, C 1-6 alkyl, C 1-6 alkylamino, C 3-6 cycloalkyl and -C 1-3 alkyl-(3-6 membered heterocycloalkane base), the C 1-6 alkyl, C 1-6 alkylamino , C 3-6 cycloalkyl and -C 1-3 alkyl-(3-6 membered heterocycloalkyl) are independently optional is substituted with 1, 2 or 3 R a ; alternatively, R 4 and R 5 are joined to form a 3-8 membered heterocycloalkyl optionally substituted by 1, 2 or 3 R b replace;
  • R 6 is selected from C 1-3 alkyl optionally substituted with 1, 2 or 3 R c ;
  • L is selected from -C 1-6 alkyl- optionally substituted with 1, 2 or 3 R d ;
  • X is selected from CH and N;
  • Y is selected from CH and N;
  • Z 1 is selected from a single bond and CH 2 ;
  • Z 2 is selected from CH and N;
  • the 3-6 membered heterocycloalkyl group and the 3-8 membered heterocycloalkyl group each independently contain 1, 2 or 3 heteroatoms or heteroatomic groups independently selected from N, O, S and NH.
  • the above Z 2 is selected from CH, and other variables are as defined in the present invention.
  • R 1 and R 2 are independently selected from H, F, Cl, Br, I, CN, CF 3 and CH 3 , and other variables are as defined in the present invention.
  • R 3 is selected from H, CN, CH 3 , -OCH 3 and The CH 3 , -OCH 3 and Each independently is optionally substituted with 1, 2 or 3 halogens, other variables are as defined herein.
  • R 3 is selected from H, CN, CH 3 , -OCH 3 , Other variables are as defined in the present invention.
  • R 4 and R 5 are each independently selected from H, C 1-4 alkyl, C 1-3 alkylamino, cyclopropyl and The C 1-4 alkyl, C 1-3 alkylamino, cyclopropyl and Each independently is optionally substituted with 1, 2 or 3 R a , other variables are as defined in the present invention.
  • R 4 and R 5 are independently selected from H, Other variables are as defined in the present invention.
  • R 4 is selected from H, Other variables are as defined in the present invention.
  • R 5 is selected from H, Other variables are as defined in the present invention.
  • R4 and R5 described above are linked to form pyrrolidinyl, 8 - azabicyclo[3.2.1]octyl, azetidinyl and 2-azaspiro[3.3]heptyl alkyl, the pyrrolidinyl, 8-azabicyclo[3.2.1]octyl, azetidinyl, and 2-azaspiro[3.3]heptyl, respectively, independently optionally replaced by 1, 2 or 3 R b substitutions, other variables are as defined in the present invention.
  • R 4 and R 5 are linked to form Other variables are as defined in the present invention.
  • R 6 is selected from CH 3 and other variables are as defined in the present invention.
  • the above L is selected from Other variables are as defined in the present invention.
  • the above-mentioned compounds are selected from
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , Z 1 are as defined in the present invention
  • the above-mentioned compounds are selected from
  • R 1 , R 2 , R 3 , R 6 , Z 1 , Z 2 , X and Y are as defined in the present invention
  • Ring A is selected from 3-8 membered heterocycloalkyl
  • the present invention also provides a compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from
  • the above-mentioned compounds are selected from
  • the present invention also provides the use of the above compounds or their pharmaceutically acceptable salts in the preparation of PD-1/PD-L1 inhibitors.
  • the above PD-1/PD-L1 inhibitor is an anti-tumor drug.
  • the compound of the present invention has a good inhibitory effect on the excessive activation of the PD-1/PD-L1 signaling pathway, thereby obtaining excellent tumor growth inhibitory activity.
  • 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
  • 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 may exist in specific geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including cis and trans isomers, (-)- and (+)-enantiomers, (R)- and (S)-enantiomers, diastereomers isomers, (D)-isomers, (L)-isomers, and racemic mixtures thereof and other mixtures, such as enantiomerically or diastereomerically enriched mixtures, all of which belong to this within the scope of the invention.
  • Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All such isomers, as well as mixtures thereof, are included within the scope of the present invention.
  • enantiomers or “optical isomers” refer to stereoisomers that are mirror images of each other.
  • cis-trans isomer or “geometric isomer” result from the inability to rotate freely due to double bonds or single bonds to ring carbon atoms.
  • diastereomer refers to a stereoisomer in which the molecule has two or more chiral centers and the molecules are in a non-mirror-image relationship.
  • tautomer or “tautomeric form” refers to isomers of different functional groups that are in dynamic equilibrium and are rapidly interconverted at room temperature.
  • a chemical equilibrium of tautomers can be achieved if tautomers are possible (eg, in solution).
  • proton tautomers also called prototropic tautomers
  • Valence tautomers include interconversions by recombination of some bonding electrons.
  • keto-enol tautomerization is the interconversion between two tautomers of pentane-2,4-dione and 4-hydroxypent-3-en-2-one.
  • the terms “enriched in one isomer”, “enriched in isomers”, “enriched in one enantiomer” or “enriched in one enantiomer” refer to one of the isomers or pairs
  • the enantiomer content is less than 100%, and the isomer or enantiomer content is greater than or equal to 60%, or greater than or equal to 70%, or greater than or equal to 80%, or greater than or equal to 90%, or greater than or equal to 95%, or Greater than or equal to 96%, or greater than or equal to 97%, or greater than or equal to 98%, or greater than or equal to 99%, or greater than or equal to 99.5%, or greater than or equal to 99.6%, or greater than or equal to 99.7%, or greater than or equal to 99.8%, or greater than or equal to 99.9%.
  • isomeric excess or “enantiomeric excess” refer to the difference between two isomers or relative percentages of two enantiomers. For example, if the content of one isomer or enantiomer is 90% and the content of the other isomer or enantiomer is 10%, the isomer or enantiomeric excess (ee value) is 80% .
  • Optically active (R)- and (S)-isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If one enantiomer of a compound of the present invention is desired, it can be prepared by asymmetric synthesis or derivatization with a chiral auxiliary, wherein the resulting mixture of diastereomers is separated and the auxiliary group is cleaved to provide pure desired enantiomer.
  • a diastereomeric salt is formed with an appropriate optically active acid or base, followed by conventional methods known in the art
  • the diastereoisomers were resolved and the pure enantiomers recovered.
  • separation of enantiomers and diastereomers is usually accomplished by the use of chromatography employing a chiral stationary phase, optionally in combination with chemical derivatization (eg, from amines to amino groups) formate).
  • the compounds of the present invention may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute the compound.
  • compounds can be labeled with radioisotopes, such as tritium ( 3 H), iodine-125 ( 125 I) or C-14 ( 14 C).
  • deuterated drugs can be formed by replacing hydrogen with deuterium, and the bonds formed by deuterium and carbon are stronger than those formed by ordinary hydrogen and carbon. Compared with non-deuterated drugs, deuterated drugs can reduce toxic side effects and increase drug stability. , enhance the efficacy, prolong the biological half-life of drugs and other advantages. All transformations of the isotopic composition of the compounds of the present invention, whether radioactive or not, are included within the scope of the present invention.
  • substituted means that any one or more hydrogen atoms on a specified atom are replaced by a substituent, which may include deuterium and hydrogen variants, as long as the valence of the specified atom is normal and the substituted compound is stable.
  • oxygen it means that two hydrogen atoms are substituted. Oxygen substitution does not occur on aromatic groups.
  • optionally substituted means that it may or may not be substituted, and unless otherwise specified, the type and number of substituents may be arbitrary on a chemically achievable basis.
  • any variable eg, R
  • its definition in each case is independent.
  • the group may optionally be substituted with up to two Rs, with independent options for R in each case.
  • combinations of substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
  • linking group When the number of a linking group is 0, such as -(CRR) 0 -, it means that the linking group is a single bond.
  • substituents When a substituent is vacant, it means that the substituent does not exist. For example, when X in A-X is vacant, it means that the structure is actually A. When the listed substituents do not indicate through which atom it is attached to the substituted group, such substituents may be bonded through any of its atoms, for example, pyridyl as a substituent may be through any one of the pyridine ring The carbon atom is attached to the substituted group.
  • 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 is an H atom at the linkable site, when the chemical bond is connected, the number of H atoms at the site will be correspondingly reduced 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;
  • the number of atoms in a ring is generally defined as the number of ring members, eg, "5-7 membered ring” refers to a “ring” of 5-7 atoms arranged around it.
  • 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.
  • C1-6alkoxy refers to those alkyl groups containing 1 to 6 carbon atoms attached to the remainder of the molecule through an oxygen atom.
  • the C 1-6 alkoxy groups include C 1-4 , C 1-3 , C 1-2 , C 2-6 , C 2-4 , C 6 , C 5 , C 4 and C 3 alkoxy groups, etc. .
  • C 1-6 alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy (including n - propoxy and isopropoxy), butoxy (including n-butoxy, isobutoxy) oxy, s-butoxy and t-butoxy), pentyloxy (including n-pentyloxy, isopentyloxy and neopentyloxy), hexyloxy and the like.
  • C1-3alkoxy refers to those alkyl groups containing 1 to 3 carbon atoms attached to the remainder of the molecule through an oxygen atom.
  • the C 1-3 alkoxy group includes C 1-2 , C 2-3 , C 3 and C 2 alkoxy and the like.
  • Examples of C 1-3 alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy (including n-propoxy and isopropoxy), and the like.
  • C 1-6 alkylamino refers to those alkyl groups containing 1 to 6 carbon atoms attached to the remainder of the molecule through an amino group.
  • the C 1-6 alkylamino includes C 1-4 , C 1-3 , C 1-2 , C 2-6 , C 2-4 , C 6 , C 5 , C 4 , C 3 and C 2 alkylamino Wait.
  • C 1-6 alkylamino examples include, but are not limited to, -NHCH 3 , -N(CH 3 ) 2 , -NHCH 2 CH 3 , -N(CH 3 )CH 2 CH 3 , -N(CH 2 CH 3 )( CH2CH3 ) , -NHCH2CH2CH3 , -NHCH2 ( CH3 ) 2 , -NHCH2CH2CH2CH3 , etc.
  • C 1-3 alkylamino refers to those alkyl groups containing 1 to 3 carbon atoms attached to the remainder of the molecule through an amino group.
  • the C 1-3 alkylamino groups include C 1-2 , C 3 and C 2 alkylamino groups and the like.
  • Examples of C 1-3 alkylamino include, but are not limited to, -NHCH 3 , -N(CH 3 ) 2 , -NHCH 2 CH 3 , -N(CH 3 )CH 2 CH 3 , -NHCH 2 CH 2 CH 3 , - NHCH 2 (CH 3 ) 2 and the like.
  • halogen or halogen by itself or as part of another substituent means a fluorine, chlorine, bromine or iodine atom.
  • C 3-6 cycloalkyl means a saturated cyclic hydrocarbon group consisting of 3 to 6 carbon atoms, which are monocyclic and bicyclic ring systems, said C 3-6 cycloalkyl including C 3-5 , C 4-5 and C 5-6 cycloalkyl and the like; it may be monovalent, divalent or polyvalent.
  • Examples of C3-6 cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
  • the term "3- to 8-membered heterocycloalkyl" by itself or in combination with other terms denotes a saturated cyclic group consisting of 3 to 8 ring atoms, respectively, of which 1, 2, 3 or 4 ring atoms are heteroatoms independently selected from O, S, and N, and the remainder are carbon atoms, where the nitrogen atom is optionally quaternized, and the nitrogen and sulfur heteroatoms are optionally oxidized (ie, NO and S(O) p , p is 1 or 2). It includes monocyclic and bicyclic ring systems, wherein bicyclic ring systems include spiro, paracyclic and bridged rings.
  • a heteroatom may occupy the position of attachment of the heterocycloalkyl to the rest of the molecule.
  • the 3-8 membered heterocycloalkyl includes 3-6 membered, 3-5 membered, 4-6 membered, 5-6 membered, 4 membered, 5 membered and 6 membered heterocycloalkyl and the like.
  • 3-8 membered heterocycloalkyl examples include, but are not limited to, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrothienyl ( Including tetrahydrothiophen-2-yl and tetrahydrothiophen-3-yl, etc.), tetrahydrofuranyl (including tetrahydrofuran-2-yl, etc.), tetrahydropyranyl, piperidinyl (including 1-piperidinyl, 2- piperidinyl and 3-piperidyl, etc.), piperazinyl (including 1-piperazinyl and 2-piperazinyl, etc.), morpholinyl (including 3-morpholinyl and 4-morpholinyl, etc.), Dioxanyl, dithianyl, isoxazolidinyl, isothiazolidinyl,
  • a heteroatom may occupy the position of attachment of the heterocycloalkyl to the remainder of the molecule.
  • the 3-6 membered heterocycloalkyl includes 4-6 membered, 5-6 membered, 4 membered, 5 membered and 6 membered heterocycloalkyl and the like.
  • Examples of 3-6 membered heterocycloalkyl include, but are not limited to, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrothienyl ( Including tetrahydrothiophen-2-yl and tetrahydrothiophen-3-yl, etc.), tetrahydrofuranyl (including tetrahydrofuran-2-yl, etc.), tetrahydropyranyl, piperidinyl (including 1-piperidinyl, 2- piperidinyl and 3-piperidyl, etc.), piperazinyl (including 1-piperazinyl and 2-piperazinyl, etc.), morpholinyl (including 3-morpholinyl and 4-morpholinyl, etc.), Dioxanyl, dithianyl, isoxazolidinyl, isothiazolidinyl
  • 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 enumerated 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.
  • the proportions of reagents used in silica gel column chromatography, silica gel column chromatography and silica gel thin-layer chromatography plates in the present invention are all volume ratios.
  • HATU O-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethylurea hexafluorophosphate
  • DMSO stands for di- Methyl sulfoxide
  • CD3OD for deuterated methanol
  • CDCl3 for deuterated chloroform
  • TBSO for tert-butyldimethylsilyloxy.
  • 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 enumerated 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.
  • MS-ESI calculated [M+H] + 760, found 760.
  • reaction solution was quenched with water (10 mL), diluted with dichloromethane (5 mL), extracted with dichloromethane (10 mL ⁇ 3), and the organic phase was washed with saturated brine (20 mL ⁇ 2), and the solution was washed with sodium sulfate. Dry, filter and concentrate.
  • MS-ESI calculated [M+H] + 401, found 403.
  • reaction solution was quenched with water (10 mL), diluted with dichloromethane (5 mL), extracted with dichloromethane (10 mL ⁇ 3), and the organic phase was washed with saturated brine (20 mL ⁇ 2), and the solution was washed with sodium sulfate. Dry, filter and concentrate.
  • reaction solution was quenched with water (10 mL), diluted with dichloromethane (5 mL), extracted with dichloromethane (10 mL ⁇ 3), and the organic phase was washed with saturated brine (20 mL ⁇ 2), and the solution was washed with sodium sulfate. Dry, filter and concentrate.
  • Small molecule compounds can competitively inhibit the binding of PD-1 and PD-L1 by binding to PD-L1; when the PD-1 molecule as the donor is very close to the PD-L1 molecule as the acceptor, the donor molecule will The energy is transferred to the receptor molecule, which in turn causes the receptor molecule to emit fluorescence; by detecting the intensity of fluorescence, the ability of small molecules to prevent the binding of PD-L1 to PD-1 can be tested.
  • a homogeneous time-resolved fluorescence (HTRF) binding assay was used to detect the ability of the compounds of the present invention to inhibit the mutual binding of PD-1/PD-L1.
  • PD-1/PD-L1TR-FRET detection kit was purchased from BPS Biosciences. Nivo Multilabel Analyzer (PerkinElmer).
  • Dilute PD1-Eu, Dye-labeled acceptor, PD-L1-biotin and test compound with the buffer in the kit.
  • the compound to be tested was diluted 5-fold to the 8th concentration with a row gun, that is, from 40 ⁇ M to 0.5 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 the compounds of the examples of the present invention on PD1/PD-L1 binding.
  • test compound IC50 (nM) Compound 2 1.49 Formate salt of compound 8 4.61 Compound 12 3.13 Compound 15 4.20
  • the compound of the present invention has a significant inhibitory effect on the binding of PD-1/PD-L1.
  • MDA-MB-231 triple-negative breast cancer cell line
  • PD-L1 molecules on the cell surface can be degraded by lysosomal and proteasome pathways, and small molecule inhibitors are added to induce PD-L1 endocytosis.
  • flow cytometry Fluorescence-activated Cell Sorting, FACS
  • FACS Fluorescence-activated Cell Sorting
  • Phosphate buffered saline 1640 medium, penicillin-streptomycin, fetal bovine serum, non-essential amino acids, ⁇ -mercaptoethanol (2-ME), human interferon ⁇ , LIVE/DEAD staining solution, staining solution (staining buffer), fixation buffer, 0.25% trypsin, EDTA, anti-human PD-L1 (Anti-human PD-L1), isotype control anti-human PD-L1 (Anti-human PD-L1 Isotype ).
  • 1640 complete medium configuration 439.5 ml of 1640 medium was added with 50 ml of fetal bovine serum, 5 ml of non-essential amino acids, 5 ml of penicillin-streptomycin and 0.5 ml of ⁇ liter mercaptoethanol, and mixed.
  • 10mM EDTA configuration add 1ml of 0.5M EDTA to 49ml of DPBS and mix.
  • MDA-MB-231 cell counting and plating remove the culture flask, remove the medium and rinse once with DPBS. After washing, add 3 ml of 0.25% trypsin to the culture flask and place it in a 37°C incubator for 1.5 min. Remove the culture flask and add 9 ml of 1640 complete medium to stop the reaction, transfer the cells to a 50 ml centrifuge tube, and centrifuge at 1000 rpm at 37°C for 5 min. Add an appropriate volume of culture medium to resuspend the cells according to the number of cells, and count with a cell counter. The cell concentration was adjusted to 5 x 10 5 cells/ml with the medium.
  • Plating A volume of 200 ⁇ L of cell suspension was added to each well of a 96-well plate, so that the number of cells in each well was 1 ⁇ 10 5 . Incubate overnight in an incubator.
  • Staining Dilute anti-human PD-L1 (2 ⁇ L per well) and LIVE/DEAD staining solution (1:1000) in staining solution, add 50 ⁇ L to each well, and stain at 4°C for 30 min. Wash twice with 200 ⁇ L of staining solution. Fixation: Add 100 ⁇ L of fixative to each well, and fix at 4°C for 15 min. Wash twice with 200 ⁇ L of staining solution. Resuspend cells in 150 ⁇ L. FACS detection. Table 2 provides the effects of the compounds of the examples of the present invention on the expression level of PD-L1 in MDA-MR-231 cells.
  • the compound of the present invention has a significant inhibitory effect on the expression level of PD-L1 in MDA-MR-231 cells.
  • the engineered T cells express PD-1 molecule and T cell receptor (TCR) on the surface, which can activate the NFAT signaling pathway of T cells after co-culture with engineered antigen presenting cells (APC).
  • TCR T cell receptor
  • APC engineered antigen presenting cells
  • the expression of PD-L1 molecules on APCs can effectively attenuate the NFAT signaling pathway in T cells; the use of PD-L1 inhibitors can effectively block the PD-1/PD-L1 regulatory mechanism, thereby reversing the weakened NFAT signaling pathway.
  • APC engineered antigen presenting cells
  • PD-1/PD-L1 NFAT 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 culture incubator overnight; the compounds to be tested were diluted 5-fold to the 8th Each concentration was diluted from 20 ⁇ M to 0.25 nM, and the DMSO concentration was 2%, and a double-well experiment was set up. Discard the TCR Activitor/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-Jurkat at a density of 4X10 5 /ml to each well. The 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).
  • Table 3 provides the inhibitory activity of the compounds of the examples of the present invention on PD-1/PD-L1 binding.
  • the compound of the present invention can inhibit the interaction of PD-1/PD-L1 at the cellular level, thereby significantly activating the NFAT signaling pathway of T cells.
  • mice Male, 8 weeks old, body weight 25g-30g
  • mice The pharmacokinetic-related parameters of the compounds of the examples of the present invention in mice are shown in the following table.
  • the compound of the present invention has good pharmacokinetic properties, including good oral bioavailability, oral exposure, half-life and clearance rate.
  • Experimental Example 5 Pharmacodynamic evaluation of the compound in the C57BL/6-hPDL1 mouse colorectal cancer MC38-hPDL1 subcutaneous transplantation model
  • Experimental purpose To evaluate the compound in the mouse colorectal cancer MC38-hPDL1 transplanted humanized mice Antitumor effects in C57BL/6-hPDL1.
  • mice mouse colon cancer cells MC38-hPDL1 were recovered, and the recovery time was Pn+6.
  • the MC38-hPDL1 cells in logarithmic growth phase were collected, and the culture medium was removed and washed twice with PBS before inoculation (before tumor-bearing, tumor-bearing
  • the survival rates of MC38-hPDL1 cells were: 97.4% and 95.0%, respectively), inoculation volume: 1 ⁇ 10 6 /100 ⁇ L/cell, inoculation location: right forelimb of mice.
  • mice On the 7th day after inoculation, when the average tumor volume reached 85.23 mm 3 , the mice were randomly divided into 5 groups according to the tumor volume, with 8 mice in each group. The day of grouping was defined as D0 day, and the administration started on D0 day. The remaining mice were used for subsequent supplementary experiments.
  • Tumor size was observed on days 0, 2, 4, 6, 8, 11, 13, 15, 18, 20, 22, and 25 after the start of administration.
  • TGItv tumor volume change
  • TGITV relative tumor inhibition rate
  • TGItv(%) [1-(meanTVtn-meanTVt0)/(meanTVvn-mean TVv0)] ⁇ 100%
  • meanTVtn the mean tumor volume of a given group when measured on day n
  • meanTVt0 the mean tumor volume of a given group when measured on day 0
  • meanTVvn mean tumor volume of the solvent control group when measured on day n
  • mean TVv0 mean tumor volume of the solvent control group measured on day 0
  • mice in the colorectal cancer MC38-hPDL1 subcutaneous transplantation model of C57BL/6-hPDL1 mice are shown in Table 8 below:
  • the compound of the present invention has excellent tumor-inhibiting effect on the colorectal cancer MC38-hPDL1 subcutaneous transplantation model of C57BL/6-hPDL1 mice, and the animal body weight does not decrease significantly during the administration process, and the tolerance is good.
  • Dosagemg/kg Dosing volume ml/kg Theoretical concentration mg/ml Dosing frequency vehicle control 0 0 0 Once a day low dose 15 5 3 Once a day medium dose 50 5 10 Once a day high dose 150 5 30 Once a day
  • the body weight of the compounds of the present invention did not decrease significantly during the administration process, and the tolerance was good; compared with the vehicle control group, gastrointestinal reactions, cough, salivation and total bilirubin (TBIL) were mainly seen.
  • Target organs were liver, lung, thymus, spleen, mesenteric lymph nodes, submandibular lymph nodes, and ileum. All the above changes can be seen to recover or recover in the recovery period, and the compounds of the present invention have good safety.

Abstract

一种联苯衍生物、其制法以及其在制备治疗相关疾病药物中的应用,具体涉及式(I)所示化合物、其药学上可接受的盐。

Description

联苯衍生物及其应用
本申请主张如下优先权:
CN202110066211.5,申请日2021年01月18日。
技术领域
本发明涉及一种联苯衍生物、其制法以及其在制备治疗相关疾病中的应用,具体涉及式(I)所示化合物及其药学上可接受的盐。
背景技术
程序性细胞死亡分子1(PD-1)又被称为CD279,是CD28/CTLA-4受体家族中重要的免疫抑制分子,是一种含有268个氨基酸残基的膜蛋白,广泛表达于T细胞、巨噬细胞、B细胞等多种免疫细胞表面,其配体是PD-L1和PD-L2。PD-L1是由CD274基因编码并主要表达于肿瘤细胞、树突状细胞和巨噬细胞表面的蛋白。PD-1和PD-L1结合后,PD-1/PD-L1信号通路被激活,进而抑制T细胞的活性化,造成T细胞失能,有助于肿瘤细胞的免疫逃逸。PD-1的另一个配体PD-L2主要表达于树突状细胞、巨噬细胞和B细胞表面,与炎症及自身免疫性疾病相关。
PD-1通过与其配体PD-L1结合,并去磷酸化TCR信号通路上的多个关键分子,从而发挥对免疫应答的负性调节作用。在健康机体中,PD-1/PD-L1这一信号通路的激活可避免因过度免疫反应造成的周围组织损伤,从而减少自身免疫性疾病的发生。但在肿瘤微环境的诱导下,PD-1、PD-L1的表达均异常升高,肿瘤细胞可以通过这些PD-L1分子与T细胞上的PD-1的结合成功逃避机体免疫***的识别和攻击。PD-(L)1单抗可以阻断这种“肿瘤免疫逃逸机制”,恢复患者自身的免疫***抗癌功能。
目前,靶向PD-1/PD-L1途径的市售药物都是单克隆抗体(mAb)。2014年,美国食品和药物管理局(FDA)批准了将PD-1分子靶向市场的前两种单克隆抗体(Pembrolizumab和Nivolumab)。在接下来的几年中,另外三种靶向PD-L1分子的mAb(Atezolizumab,Durvalumab和Avelumab)出现在市场上。以上这些都是生物大分子,其自身具有显著的缺陷,如易被蛋白酶分解,在体内稳定性较差,需经注射给药;产品质量不易控制,生产技术要求高;大量制备和纯化比较困难,生产成本高及易产生免疫原性等。而小分子PD-1/PD-L1抑制剂越来越受到关注,Incyte的PD-L1小分子抑制剂INCB86550(WO2018119263,WO2019191707)和Gilead的PD-L1小分子抑制剂GS-4224(US20180305315,WO2019160882)已进入临床2期,BMS苄基苯基醚类的小分子PD-1/PD-L1抑制剂(WO2015034820,WO2015160641)在临床前研究阶段。由于小分子药物能够穿过细胞膜作用于细胞内靶点,储存运输方便,生产成本较低,无免疫原性及通常可口服给药等优势。因此,研究开发PD-1/PD-L1的小分子阻断剂具有广阔的应用前景。
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 PCTCN2022072583-appb-000001
其中,
R 1和R 2分别独立地选自H、F、Cl、Br、I、CN和C 1-3烷基,所述C 1-3烷基任选被1、2或3个卤素取代;
R 3选自H、CN、C 1-3烷基、C 1-3烷氧基和C 1-3烷氨基,所述C 1-3烷基、C 1-3烷氧基和C 1-3烷氨基分别独立地任选被1、2或3个卤素取代;
R 4和R 5分别独立地选自H、C 1-6烷基、C 1-6烷氨基、C 3-6环烷基和-C 1-3烷基-(3-6元杂环烷基),所述C 1- 6烷基、C 1-6烷氨基、C 3-6环烷基和-C 1-3烷基-(3-6元杂环烷基)分别独立地任选被1、2或3个R a取代;或者,R 4和R 5连接形成3~8元杂环烷基,所述3~8元杂环烷基任选被1、2或3个R b取代;
R 6选自任选被1、2或3个R c取代的C 1-3烷基;
L选自任选被1、2或3个R d取代的-C 1-6烷基-;
X选自CH和N;
Y选自CH和N;
Z 1选自单键和CH 2
Z 2选自CH和N;
R a选自F、Cl、Br、I、OH、=O、NH 2、C 1-3烷氨基、C 1-3烷氧基和C 1-3烷基,所述C 1-3烷氨基、C 1-3烷氧基和C 1-3烷基分别独立地任选被1、2或3个R取代;
R b选自CN、F、Cl、Br、I、OH、-C(=O)NH 2、C 1-3烷氨基、C 1-3烷氧基和C 1-3烷基;
R c选自F、Cl、Br、I、CN、OH、=O和NH 2
R d选自F、Cl、Br、I、CN、OH、=O和NH 2
R选自F、Cl、Br、I、CN、OH、=O和NH 2
所述3-6元杂环烷基和3~8元杂环烷基分别独立地包含1、2或3个分别独立地选自N、O、S和NH的杂原子或杂原子团。
在本发明的一些方案中,上述Z 2选自CH,其他变量如本发明所定义。
在本发明的一些方案中,上述R 1和R 2分别独立地选自H、F、Cl、Br、I、CN、CF 3和CH 3,其他变量如本发明所定义。
在本发明的一些方案中,上述R 3选自H、CN、CH 3、-OCH 3
Figure PCTCN2022072583-appb-000002
所述CH 3、-OCH 3
Figure PCTCN2022072583-appb-000003
分别独立地任选被1、2或3个卤素取代,其他变量如本发明所定义。
在本发明的一些方案中,上述R 3选自H、CN、CH 3、-OCH 3
Figure PCTCN2022072583-appb-000004
其他变量如本 发明所定义。
在本发明的一些方案中,上述R 4和R 5分别独立地选自H、C 1-4烷基、C 1-3烷氨基、环丙基和
Figure PCTCN2022072583-appb-000005
所述C 1-4烷基、C 1-3烷氨基、环丙基和
Figure PCTCN2022072583-appb-000006
分别独立地任选被1、2或3个R a取代,其他变量如本发明所定义。
在本发明的一些方案中,上述R a选自F、Cl、Br、I、OH、=O、NH 2、-NHCH 3、-OCH 3、-CH 2OH和CH 3,其他变量如本发明所定义。
在本发明的一些方案中,上述R 4和R 5分别独立地选自H、
Figure PCTCN2022072583-appb-000007
Figure PCTCN2022072583-appb-000008
其他变量如本发明所定义。
在本发明的一些方案中,上述R 4选自H、
Figure PCTCN2022072583-appb-000009
Figure PCTCN2022072583-appb-000010
其他变量如本发明所定义。
在本发明的一些方案中,上述R 5选自H、
Figure PCTCN2022072583-appb-000011
Figure PCTCN2022072583-appb-000012
其他变量如本发明所定义。
在本发明的一些方案中,上述R 4和R 5连接形成吡咯烷基、8-氮杂双环[3.2.1]辛烷基、氮杂环丁烷基和2-氮杂螺[3.3]庚烷基,所述吡咯烷基、8-氮杂双环[3.2.1]辛烷基、氮杂环丁烷基和2-氮杂螺[3.3]庚烷基分别独立地任选被1、2或3个R b取代,其他变量如本发明所定义。
在本发明的一些方案中,上述R b选自CN、OH、-C(=O)NH 2、-OCH 3和CH 3,其他变量如本发明所定义。
在本发明的一些方案中,上述R 4和R 5连接形成
Figure PCTCN2022072583-appb-000013
Figure PCTCN2022072583-appb-000014
其他变量如本发明所定义。
在本发明的一些方案中,上述R 6选自CH 3,其他变量如本发明所定义。
在本发明的一些方案中,上述L选自
Figure PCTCN2022072583-appb-000015
其他变量如本发明所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2022072583-appb-000016
选自
Figure PCTCN2022072583-appb-000017
Figure PCTCN2022072583-appb-000018
Figure PCTCN2022072583-appb-000019
其他变量如本发明所定义。
在本发明的一些方案中,上述结构单元
Figure PCTCN2022072583-appb-000020
选自
Figure PCTCN2022072583-appb-000021
其他变量如本发明所定义。
在本发明的一些方案中,上述化合物选自
Figure PCTCN2022072583-appb-000022
其中,R 1、R 2、R 3、R 4、R 5、R 6、Z 1如本发明所定义,R 7和R 8选自H、F、Cl、Br、I、CN、OH、=O和NH 2
在本发明的一些方案中,上述化合物选自
Figure PCTCN2022072583-appb-000023
其中,R 1、R 2、R 3、R 6、Z 1、Z 2、X和Y如本发明所定义,
环A选自3~8元杂环烷基,
R 9和R 10分别独立地选自H、CN、F、Cl、Br、I、OH、-C(=O)NH 2、C 1-3烷氨基、C 1-3烷氧基和C 1-3烷基。
本发明还提供化合物或其药学上可接受的盐,其中,化合物选自
Figure PCTCN2022072583-appb-000024
Figure PCTCN2022072583-appb-000025
Figure PCTCN2022072583-appb-000026
Figure PCTCN2022072583-appb-000027
在本发明的一些方案中,上述化合物选自
Figure PCTCN2022072583-appb-000028
本发明还提供上述化合物或其药学上可接受的盐在制备PD-1/PD-L1抑制剂中的应用。
在本发明的一些方案中,上述PD-1/PD-L1抑制剂是抗肿瘤药物。
技术效果
本发明化合物对PD-1/PD-L1信号通路过度活化有良好的抑制效果,进而获得了优良的抑制肿瘤生长的活性。
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。
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存在特定的几何或立体异构体形式。本发明设想所有的这类化合物,包括顺式和反式异构体、(-)-和(+)-对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,及其外消旋混合物和其他混合物,例如对映异构体或非对映体富集的混合物,所有这些混合物都属于本发明的范围之内。烷基等取代基中可存在另外的不对称碳原子。所有这些异构体以及它们的混合物,均包括在本发明的范围之内。
除非另有说明,术语“对映异构体”或者“旋光异构体”是指互为镜像关系的立体异构体。
除非另有说明,术语“顺反异构体”或者“几何异构体”系由因双键或者成环碳原子单键不能自由旋转而引起。
除非另有说明,术语“非对映异构体”是指分子具有两个或多个手性中心,并且分子间为非镜像的关系的立体异构体。
除非另有说明,“(+)”表示右旋,“(-)”表示左旋,“(±)”表示外消旋。
除非另有说明,用楔形实线键
Figure PCTCN2022072583-appb-000029
和楔形虚线键
Figure PCTCN2022072583-appb-000030
表示一个立体中心的绝对构型,用直形实线键
Figure PCTCN2022072583-appb-000031
和直形虚线键
Figure PCTCN2022072583-appb-000032
表示立体中心的相对构型,用波浪线
Figure PCTCN2022072583-appb-000033
表示楔形实线键
Figure PCTCN2022072583-appb-000034
或楔形虚线键
Figure PCTCN2022072583-appb-000035
或用波浪线
Figure PCTCN2022072583-appb-000036
表示直形实线键
Figure PCTCN2022072583-appb-000037
和直形虚线键
Figure PCTCN2022072583-appb-000038
除非另有说明,术语“互变异构体”或“互变异构体形式”是指在室温下,不同官能团异构体处于动态平衡,并能很快的相互转化。若互变异构体是可能的(如在溶液中),则可以达到互变异构体的化学平衡。例如,质子互变异构体(proton tautomer)(也称质子转移互变异构体(prototropic tautomer))包括通过质子迁移来进行的互相转化,如酮-烯醇异构化和亚胺-烯胺异构化。价键异构体(valence tautomer)包括一些成键电子的重组来进行的相互转化。其中酮-烯醇互变异构化的具体实例是戊烷-2,4-二酮与4-羟 基戊-3-烯-2-酮两个互变异构体之间的互变。
除非另有说明,术语“富含一种异构体”、“异构体富集”、“富含一种对映体”或者“对映体富集”指其中一种异构体或对映体的含量小于100%,并且,该异构体或对映体的含量大于等于60%,或者大于等于70%,或者大于等于80%,或者大于等于90%,或者大于等于95%,或者大于等于96%,或者大于等于97%,或者大于等于98%,或者大于等于99%,或者大于等于99.5%,或者大于等于99.6%,或者大于等于99.7%,或者大于等于99.8%,或者大于等于99.9%。
除非另有说明,术语“异构体过量”或“对映体过量”指两种异构体或两种对映体相对百分数之间的差值。例如,其中一种异构体或对映体的含量为90%,另一种异构体或对映体的含量为10%,则异构体或对映体过量(ee值)为80%。
可以通过的手性合成或手性试剂或者其他常规技术制备光学活性的(R)-和(S)-异构体以及D和L异构体。如果想得到本发明某化合物的一种对映体,可以通过不对称合成或者具有手性助剂的衍生作用来制备,其中将所得非对映体混合物分离,并且辅助基团裂开以提供纯的所需对映异构体。或者,当分子中含有碱性官能团(如氨基)或酸性官能团(如羧基)时,与适当的光学活性的酸或碱形成非对映异构体的盐,然后通过本领域所公知的常规方法进行非对映异构体拆分,然后回收得到纯的对映体。此外,对映异构体和非对映异构体的分离通常是通过使用色谱法完成的,所述色谱法采用手性固定相,并任选地与化学衍生法相结合(例如由胺生成氨基甲酸盐)。
本发明的化合物可以在一个或多个构成该化合物的原子上包含非天然比例的原子同位素。例如,可用放射性同位素标记化合物,比如氚( 3H),碘-125( 125I)或C-14( 14C)。又例如,可用重氢取代氢形成氘代药物,氘与碳构成的键比普通氢与碳构成的键更坚固,相比于未氘化药物,氘代药物有降低毒副作用、增加药物稳定性、增强疗效、延长药物生物半衰期等优势。本发明的化合物的所有同位素组成的变换,无论放射性与否,都包括在本发明的范围之内。
术语“任选”或“任选地”指的是随后描述的事件或状况可能但不是必需出现的,并且该描述包括其中所述事件或状况发生的情况以及所述事件或状况不发生的情况。
术语“被取代的”是指特定原子上的任意一个或多个氢原子被取代基取代,取代基可以包括重氢和氢的变体,只要特定原子的价态是正常的并且取代后的化合物是稳定的。当取代基为氧(即=O)时,意味着两个氢原子被取代。氧取代不会发生在芳香基上。术语“任选被取代的”是指可以被取代,也可以不被取代,除非另有规定,取代基的种类和数目在化学上可以实现的基础上可以是任意的。
当任何变量(例如R)在化合物的组成或结构中出现一次以上时,其在每一种情况下的定义都是独立的。因此,例如,如果一个基团被0-2个R所取代,则所述基团可以任选地至多被两个R所取代,并且每种情况下的R都有独立的选项。此外,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。
当一个连接基团的数量为0时,比如-(CRR) 0-,表示该连接基团为单键。
当其中一个变量选自单键时,表示其连接的两个基团直接相连,比如A-L-Z中L代表单键时表示该结构实际上是A-Z。
当一个取代基为空缺时,表示该取代基是不存在的,比如A-X中X为空缺时表示该结构实际上是 A。当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 PCTCN2022072583-appb-000039
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 PCTCN2022072583-appb-000040
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 PCTCN2022072583-appb-000041
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 PCTCN2022072583-appb-000042
直形虚线键
Figure PCTCN2022072583-appb-000043
或波浪线
Figure PCTCN2022072583-appb-000044
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 PCTCN2022072583-appb-000045
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 PCTCN2022072583-appb-000046
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 PCTCN2022072583-appb-000047
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 PCTCN2022072583-appb-000048
这4种连接方式,即使-N-上画出了H原子,但是
Figure PCTCN2022072583-appb-000049
仍包括
Figure PCTCN2022072583-appb-000050
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,环上原子的数目通常被定义为环的元数,例如,“5-7元环”是指环绕排列5-7个原子的“环”。
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。
除非另有规定,术语“C 1-6烷氧基”表示通过一个氧原子连接到分子的其余部分的那些包含1至6个碳原子的烷基基团。所述C 1-6烷氧基包括C 1-4、C 1-3、C 1-2、C 2-6、C 2-4、C 6、C 5、C 4和C 3烷氧基等。C 1- 6烷氧基的实例包括但不限于甲氧基、乙氧基、丙氧基(包括正丙氧基和异丙氧基)、丁氧基(包括n-丁氧基、异丁氧基、s-丁氧基和t-丁氧基)、戊氧基(包括n-戊氧基、异戊氧基和新戊氧基)、己氧基等。
除非另有规定,术语“C 1-3烷氧基”表示通过一个氧原子连接到分子的其余部分的那些包含1至3个碳原子的烷基基团。所述C 1-3烷氧基包括C 1-2、C 2-3、C 3和C 2烷氧基等。C 1-3烷氧基的实例包括但不限于甲氧基、乙氧基、丙氧基(包括正丙氧基和异丙氧基)等。
除非另有规定,术语“C 1-6烷氨基”表示通过氨基连接到分子的其余部分的那些包含1至6个碳原子的烷基基团。所述C 1-6烷氨基包括C 1-4、C 1-3、C 1-2、C 2-6、C 2-4、C 6、C 5、C 4、C 3和C 2烷氨基等。C 1-6烷氨基的实例包括但不限于-NHCH 3、-N(CH 3) 2、-NHCH 2CH 3、-N(CH 3)CH 2CH 3、-N(CH 2CH 3)(CH 2CH 3)、-NHCH 2CH 2CH 3、-NHCH 2(CH 3) 2、-NHCH 2CH 2CH 2CH 3等。
除非另有规定,术语“C 1-3烷氨基”表示通过氨基连接到分子的其余部分的那些包含1至3个碳原子的烷基基团。所述C 1-3烷氨基包括C 1-2、C 3和C 2烷氨基等。C 1-3烷氨基的实例包括但不限于-NHCH 3、-N(CH 3) 2、-NHCH 2CH 3、-N(CH 3)CH 2CH 3、-NHCH 2CH 2CH 3、-NHCH 2(CH 3) 2等。
除非另有规定,术语“卤代素”或“卤素”本身或作为另一取代基的一部分表示氟、氯、溴或碘原子。
除非另有规定,“C 3-6环烷基”表示由3至6个碳原子组成的饱和环状碳氢基团,其为单环和双环体系,所述C 3-6环烷基包括C 3-5、C 4-5和C 5-6环烷基等;其可以是一价、二价或者多价。C 3-6环烷基的实例包括,但不限于,环丙基、环丁基、环戊基、环己基等。
除非另有规定,术语“3-8元杂环烷基”本身或者与其他术语联合分别表示由3至8个环原子组成的饱和环状基团,其1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子,其中氮原子任选地被季铵化,氮和硫杂原子可任选被氧化(即NO和S(O) p,p是1或2)。其包括单环和双环体系,其中双环体系包括螺环、并环和桥环。此外,就该“3-8元杂环烷基”而言,杂原子可以占据杂环烷基与分子其余部分的连接位置。所述3-8元杂环烷基包括3-6元、3-5元、4-6元、5-6元、4元、5元和6元杂环烷基等。3-8元杂环烷基的实例包括但不限于氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢噻吩基(包括四氢噻吩-2-基和四氢噻吩-3-基等)、四氢呋喃基(包括四氢呋喃-2-基等)、四氢吡喃基、哌啶基(包括1-哌啶基、2-哌啶基和3-哌啶基等)、哌嗪基(包括1-哌嗪基和2-哌嗪基等)、吗啉基(包括3-吗啉基和4-吗啉基等)、二噁烷基、二噻烷基、异噁唑烷基、异噻唑烷基、1,2-噁嗪基、1,2-噻嗪基、六氢哒嗪基、高哌嗪基、高哌啶基或二氧杂环庚烷基等。
除非另有规定,术语“3-6元杂环烷基”本身或者与其他术语联合分别表示由3至6个环原子组成的 饱和环状基团,其1、2、3或4个环原子为独立选自O、S和N的杂原子,其余为碳原子,其中氮原子任选地被季铵化,碳、氮和硫杂原子可任选被氧化(即C(=O)、NO和S(O)p,p是1或2)。其包括单环和双环体系,其中双环体系包括螺环、并环和桥环。此外,就该“3-6元杂环烷基”而言,杂原子可以占据杂环烷基与分子其余部分的连接位置。所述3-6元杂环烷基包括4-6元、5-6元、4元、5元和6元杂环烷基等。3-6元杂环烷基的实例包括但不限于氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢噻吩基(包括四氢噻吩-2-基和四氢噻吩-3-基等)、四氢呋喃基(包括四氢呋喃-2-基等)、四氢吡喃基、哌啶基(包括1-哌啶基、2-哌啶基和3-哌啶基等)、哌嗪基(包括1-哌嗪基和2-哌嗪基等)、吗啉基(包括3-吗啉基和4-吗啉基等)、二噁烷基、二噻烷基、异噁唑烷基、异噻唑烷基、1,2-噁嗪基、1,2-噻嗪基、六氢哒嗪基、高哌嗪基或高哌啶基等。
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 PCTCN2022072583-appb-000051
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。
如无特殊说明,本发明中硅胶柱层析、硅胶色谱柱层析和硅胶薄层色谱板中所用试剂配比均为体积比。
本发明采用下述缩略词:HATU代表O-(7-氮杂苯并***-1-基)-N,N,N′,N′-四甲基脲六氟磷酸盐;DMSO代表二甲亚砜;CD 3OD代表氘代甲醇;CDCl 3代表氘代氯仿;TBSO代表叔丁基二甲基硅基氧基。
化合物依据本领域常规命名原则或者使用
Figure PCTCN2022072583-appb-000052
软件命名,市售化合物采用供应商目录名称。
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。
中间体A
Figure PCTCN2022072583-appb-000053
第一步
将化合物A-1(10克,48.43毫摩尔)和化合物A-2(15.99克,62.96毫摩尔)溶于二氧六环中(100毫升),向反应液中加入1,1′-双(二苯基磷)二茂铁氯化钯(3.54克,4.84毫摩尔)和乙酸钾(11.88克,121.08毫摩尔),反应混合物置换气体后在氮气保护下80℃搅拌15小时,反应完毕,将反应混合物过滤浓缩,得粗产品中间体A。MS-ESI计算值[M+H] +254,实测值254。
中间体B
Figure PCTCN2022072583-appb-000054
第一步
将化合物B-1(20克,144.80毫摩尔)和丙酮酸乙酯(84.07克,723.99毫摩尔)混合,并在20℃条件下搅拌15分钟。然后加入三氯氧磷(222.02克,1.45摩尔)并在100℃条件下搅拌1小时。反应完毕,将反应液倒入冰水(1升),碳酸钠调节pH至7,用乙酸乙酯(150毫升×2)萃取,有机相用饱和食盐水(100毫升×2)洗涤,有机层用无水硫酸钠干燥,过滤并浓缩,残余物通过硅胶柱层析(石油醚∶乙酸乙酯=15∶1到3∶1)纯化得到化合物B-2。MS-ESI计算值[M+H] +236.8实测值236.8。
第二步
将化合物B-2(16.8克,70.99毫摩尔)和氰基硼氢化钠(13.387克,212.97毫摩尔)溶于醋酸(100毫升),反应液在氮气保护下20℃反应1小时。反应完毕后,将反应液倒入冰水,碳酸钠调节pH至7,用乙酸乙酯(200毫升×2)萃取,有机相用饱和食盐水(100毫升×2)洗涤,无水硫酸钠干燥,过滤并浓缩,得到化合物B-3直接用于下一步骤无需进一步纯化。MS-ESI计算值[M+H] +240.8,实测值240.8。
第三步
将化合物B-3(23克,95.56毫摩尔)溶于二氯甲烷(200毫升)中,加入二碳酸二叔丁酯(31.28克,143.34毫摩尔),三乙胺(29.01克,286.68毫摩尔),25℃搅拌0.5小时。反应完毕,往反应液中加水(300毫升),用二氯甲烷(200毫升×2)萃取,有机相用饱和食盐水(150毫升×2)洗涤,无水硫酸钠干燥,过滤,浓缩,残余物通过硅胶柱层析(石油醚∶乙酸乙酯=15∶1到5∶1)纯化得到化合物B-4。MS-ESI计算值[M+H] +340.8,实测值340.8。
第四步
将化合物B-4(500毫克,1.38毫摩尔)溶于甲醇(10毫升),接着向反应液中加入甲醇钠(744.99毫 克,13.79毫摩尔),在70℃下搅拌10小时。反应完毕后,将反应液倒入饱和氯化铵(10毫升)水溶液中,用乙酸乙酯30毫升(10毫升×3)萃取,合并的有机相经饱和食盐水30毫升(10毫升×3)洗涤后,用无水硫酸钠干燥,过滤减压浓缩得到化合物B-5。MS-ESI计算值[M+H] +308,实测值308。
第五步
将化合物B-5(400毫克,1.30毫摩尔)溶于N,N-二甲基甲酰胺(5毫升),接着向反应液中加入碘乙烷(1.01克,6.49毫摩尔)和碳酸钾(359.59毫克,2.59毫摩尔),在60℃下搅拌10小时。反应完成后,加水(10毫升)稀释后用乙酸乙酯40毫升(20毫升×2)萃取,合并的有机相用饱和食盐水40毫升(20毫升×2)洗涤,无水硫酸钠干燥,过滤,浓缩。粗品用硅胶柱层析纯化(石油醚 乙酸乙酯=10∶1到3∶1)得到化合物B-6。MS-ESI计算值[M+H] +337,实测值337。 1H NMR(400MHz,CDCl 3)δ=7.53(s,1H),4.80(s,2H),4.51(q,2H),3.97(s,3H),3.68(t,2H),2.79(br t,2H),1.67(br s,9H),1.43-1.48(m,3H).
第六步
将化合物B-6(300毫克,891.84微摩尔)溶于乙酸乙酯(3毫升)中,然后滴加氯化氢的乙酸乙酯溶液(4摩尔/升,1.11毫升),在20℃下搅拌0.5小时。反应完成后,减压浓缩得到化合物B-7的盐酸盐。
MS-ESI计算值[M+H] +236,实测值236。
第七步
将化合物B-7(250毫克,916.67微摩尔,盐酸盐)和化合物B-8(319.5毫克,1.83毫摩尔)溶于二氯甲烷(3毫升)中,然后加入N,N-二异丙基乙胺(118.47毫克,916.67微摩尔)和三乙氧基硼氢化钠(582.84毫克,2.75毫摩尔),在20℃下反应2小时。反应完成后,将反应液过滤减压浓缩,剩余物经硅胶薄层色谱板(石油醚∶乙酸乙酯=3∶1)分离得到中间体B。MS-ESI计算值[M+H] +395,实测值395。
中间体D
Figure PCTCN2022072583-appb-000055
第一步
将中间体A(600毫克,2.37毫摩尔)和化合物D-1(511.84毫克,1.89毫摩尔)溶于二氧六环(10毫升),然后加入1,1-双(二苯基磷)二茂铁氯化钯(173.16毫克,236.66微摩尔)和碳酸钾(981.22毫克,7.10毫摩尔),用氮气置换溶液中的空气3次,反应液在氮气保护下80℃搅拌12小时。反应完成后,用20毫升水稀释,并用乙酸乙酯60毫升(20毫升×3)萃取,有机相用饱和食盐水40毫升(20毫升×2)洗涤,并用无水硫酸钠干燥,过滤减压浓缩。剩余物经硅胶薄层色谱板(石油醚 乙酸乙酯=5∶1)分离得到化合物D-2。MS-ESI计算值[M+H]+317,实测值317。
第二步
将化合物D-2(8克,25.24毫摩尔)和化合物A-2(7.05克,27.76毫摩尔)溶于二氧六环(100毫升)中,然后加入1,1-双(二苯基磷)二茂铁氯化钯(1.85克,2.52毫摩尔)和醋酸钾(7.43克,75.72毫摩尔),用氮气置换溶液中的空气3次,反应液在氮气保护下90℃搅拌4小时。反应完成后,将反应液过滤减压浓缩,得到的剩余物经硅胶色谱柱(石油醚∶乙酸乙酯=30∶1到10∶1)分离得到中间体D。MS-ESI计算值[M+H]+364,实测值364。 1H NMR(400MHz,CDCl 3)δ=7.68(dd,J=4.3,4.9Hz,1H),7.33-7.28(m,2H),7.11(t,J=7.8Hz,1H),6.83(dd,J=1.5,8.0Hz,1H),6.64(dd,J=1.4,7.5Hz,1H),1.39(s,12H)。
中间体C
Figure PCTCN2022072583-appb-000056
第一步
将化合物C-1(10克,49.01毫摩尔)加入到乙腈(100毫升)和水(150毫升)中,加入氢碘酸水溶液(100毫升,质量分数55%),0℃下搅拌0.5小时。将亚硝酸钠(50.73克,735.21毫摩尔)溶于水(100毫升),在0℃下逐滴加入到上述反应液中。滴加完毕后先在20℃下搅拌1小时,然后50℃下继续搅拌16小时。反应完毕后,混合物冷却至20℃,滴加到氢氧化钠水溶液(600毫升,质量分数20%)中同时保持温度在0℃。混合物用乙酸乙酯萃取(500毫升×2),有机相用无水硫酸钠干燥,过滤,浓缩。残余物用硅胶柱层析(石油醚∶乙酸乙酯=100∶1)分离得到化合物C-2。 1H NMR(400MHz,CDCl 3)δ=8.08(s,1H),4.05(s,3H).
第二步
将化合物C-2(9.2克,29.22毫摩尔)溶于四氢呋喃(100毫升),零下60℃下滴加到异丙基氯化镁氯化锂复合物(1.3摩尔/升,40.45毫升)并搅拌30分钟,然后零下40℃加入N,N-二甲基甲酰胺(6.41克,87.65毫摩尔)并搅拌1小时。反应完毕后,小心向反应物中滴加饱和氯化铵水溶液(100毫升),然后用乙酸乙酯萃取(50毫升×2),有机相用无水硫酸钠干燥,过滤,浓缩。得到化合物C-3。MS-ESI计算值[M+H] +217和219,实测值217和219。
第三步
将化合物C-3(1.5克,6.91毫摩尔)溶于甲醇(15毫升)中,然后0℃下加入硼氢化钠(784.47毫 克,20.74毫摩尔),25℃搅拌2小时。反应完成后,用20毫升饱和氯化铵水溶液淬灭反应,减压浓缩大部分甲醇后,加水(20毫升)稀释,然后用乙酸乙酯(90毫升)萃取,有机相用饱和食盐水(60毫升)洗涤,并用无水硫酸钠干燥,过滤减压浓缩。剩余物经硅胶色谱柱(石油醚 乙酸乙酯=30 1到10∶1)分离得到化合物C-4。 1H NMR(400MHz,DMSO-d 6)δ=8.34(s,1H),5.24(t,J=6.0Hz,1H),4.52(d,J=6.0Hz,2H),3.94(s,3H)
第四步
将化合物C-4(1.4克,6.39毫摩尔)和中间体D(2.33克,6.39毫摩尔)溶于二氧六环(15毫升)和水(2毫升),然后加入[1,1-双(二苯基膦)二茂铁]二氯化钯(467.68毫克,639.17微摩尔)和碳酸钾(2.65克,19.17毫摩尔),用氮气置换溶液中的空气3次,反应液在氮气保护下65℃搅拌4小时。反应完成后,用水(40毫升)稀释,并用乙酸乙酯(150毫升)萃取,有机相用饱和食盐水(60毫升)洗涤,并用无水硫酸钠干燥,过滤减压浓缩。剩余物经硅胶色谱柱(石油醚 乙酸乙酯=30 1到3 1)分离得到化合物C-5。
第五步
将化合物C-5(171.63毫克,456.18微摩尔)和中间体B(200毫克,506.87微摩尔)溶于四氢呋喃(4毫升),零度氮气保护下加入双三甲硅基氨基锂的四氢呋喃溶液(1摩尔/升,1.77毫升),反应液在零度下反应1小时。反应结束后,反应液用饱和氯化铵溶液(20毫升)淬灭,乙酸乙酯(20毫升)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。粗品经硅胶薄层色谱板(石油醚 乙酸乙酯=1 2)纯化分离得到化合物C-6。MS-ESI计算值[M+H] +724,实测值724。
第六步
将化合物C-6(130毫克,186.27微摩尔)溶解于1,1-二氯乙烷(2毫升)中,加入二氧化锰(161.94毫克,1.86毫摩尔),反应液在氮气保护下加热到90℃反应2小时。反应完毕后,过滤,浓缩。得到中间体C。MS-ESI计算值[M+H] +722,实测值722。
中间体L
Figure PCTCN2022072583-appb-000057
第一步
用化合物L-1(6克,27.65毫摩尔)替换化合物K-1(5.30克,48.38毫摩尔),方法同中间体K。反应完毕,反应液减压浓缩得到化合物L-2。MS-ESI计算值[M+H] +288和290,实测值288和290。
第二步
用化合物L-2(5克,17.35毫摩尔)替换化合物K-2(9克,32.83毫摩尔),方法同中间体K。反应完毕,反应液减压浓缩得到中间体L。MS-ESI计算值[M+H] +402和404,实测值402和404。
中间体E
Figure PCTCN2022072583-appb-000058
第一步
将中间体L(100毫克,248.51微摩尔)和中间体D(99.52毫克,273.36微摩尔)溶于二氧六环(5毫升)和水(0.5毫升),然后加入[1,1-双(二苯基膦)二茂铁]二氯化钯(18.18毫克,24.85微摩尔)和碳酸钾(103.04毫克,745.53微摩尔),用氮气置换溶液中的空气3次,反应液在氮气保护下65℃搅拌4小时。反应完成后,用20毫升水稀释,并用乙酸乙酯120毫升(40毫升×3)萃取,有机相用饱和食盐水60毫升(30毫升×2)洗涤,并用无水硫酸钠干燥,过滤减压浓缩。剩余物经硅胶薄层色谱板(石油醚∶乙酸乙酯=3∶1)分离得到中间体E。MS-ESI计算值[M+H] +559,实测值559。 1H NMR(400MHz,CD 3OD)δ=8.31(s,1H),7.53(dd,J=1.7,7.7Hz,1H),7.40(t,J=7.6Hz,1H),7.26(dd,J=1.7,7.5Hz,1H),7.03(t,J=7.8Hz,1H),6.81(dd,J=1.5,8.2Hz,1H),6.52(dd,J=1.5,7.5Hz,1H),4.49(s,2H),3.97(s,3H),3.83(s,2H),3.48-3.43(m,2H),1.47(s,3H),1.11(s,1H),0.80(s,9H),0.00(s,6H)。
中间体F
Figure PCTCN2022072583-appb-000059
第一步
将化合物B-2(1克,4.23毫摩尔)溶于四氢呋喃(10毫升)和水(2毫升),接着向反应液中加入氢氧化钠(845.05毫克,21.13毫摩尔),在70℃下搅拌10小时。将反应液过滤减压浓缩得到化合物F-1的粗品。MS-ESI计算值[M+H] +191,实测值191。
第二步
将化合物F-1(0.9克,4.73毫摩尔)溶于乙醇(10毫升),接着向反应液中加入浓硫酸(2.32克,23.66毫摩尔),在70℃下搅拌2小时。反应完成后,过滤滤液减压浓缩的残渣经硅胶柱层析(石油醚 乙酸乙酯=20∶1到1∶1)纯化得到化合物F-2。MS-ESI计算值[M+H] +219,实测值219。
第三步
将化合物F-2(130毫克,583.85微摩尔)和化合物F-3(267.04毫克,1.75毫摩尔),碳酸铯(570.68毫克,1.75毫摩尔)溶于N,N二甲基甲酰胺(2毫升),在80℃下搅拌0.5小时。反应完成后,过滤减压浓缩的残渣经硅胶薄层色谱板(石油醚∶乙酸乙酯=2∶1)纯化得到化合物F-4。MS-ESI计算值[M+H] +268,实测值268。
第四步
将化合物F-4(90毫克,325.48微摩尔)溶于冰醋酸(2毫升),接着向反应液中加入氰基硼氢化钠(40.91毫克,650.97微摩尔),在20℃下搅拌0.5小时。将反应液减压浓缩得到化合物F-5的粗品。MS-ESI计算值[M+H] +272,实测值272。
第五步
将化合物F-5(80毫克,293.85微摩尔)溶于二氯甲烷(2毫升)中,然后加入化合物B-8(102.44毫克,587.70微摩尔)和三乙氧基硼氢化钠(186.84毫克,881.55微摩尔),反应液在20℃下反应1小时。反应完成后,将反应液过滤并减压浓缩。剩余物经硅胶薄层色谱板(石油醚∶乙酸乙酯=3∶1)分离得到化合物F-6。MS-ESI计算值[M+H] +431,实测值431。
第六步
将化合物F-6(1.2克,2.79毫摩尔)和化合物C-5(838.88毫克,2.23毫摩尔)溶于四氢呋喃(15毫升),接着在0℃条件下向反应液中加入二(三甲基硅)氨基锂的四氢呋喃溶液(1摩尔/升,9.75毫升),在25℃下搅拌1小时。反应完成后,在0℃下将反应液倒入饱和氯化铵水溶液(50毫升)中,然后加水(30毫升)稀释,用乙酸乙酯(50毫升×3)萃取,合并的有机相经饱和食盐水(20毫升×2)洗后,用无水硫酸钠干燥,过滤减压浓缩,剩余物经硅胶色谱柱(石油醚∶乙酸乙酯=20∶1到5∶1)分离得到中间体F。MS-ESI计算值[M+H] +760,实测值760。
中间体G
Figure PCTCN2022072583-appb-000060
第一步
将化合物B-4(1克,2.76毫摩尔)溶于N,N-二甲基乙酰胺(10毫升),接着向反应液中加入氰化锌(649.76毫克,5.52毫摩尔)和甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2,4,6-三异丙基-1,1-联苯)(2-氨基-1,1-联苯-2-基)钯(II)(500.06毫克,551.64微摩尔),氮气保护下,120℃搅拌10小时。反应完成后,将加水(10毫升)稀释后用乙酸乙酯30毫升(10毫升×3)萃取,合并的有机相用饱和食盐水30毫升(10毫升×3)洗涤,无水硫酸钠干燥,过滤,浓缩。得到残渣经高效液相色谱(色谱柱:Phenomenex luna C18 250*50毫米*10微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流 动相B:乙腈;B%:25%-55%,30分钟)分离得到化合物G-1。MS-ESI计算值[M+H] +332,实测值332。 1H NMR(400MHz,DMSO-d 6)δppm 8.23(s,1H),4.64(br s,2H),3.66-3.72(m,2H),3.02(br s,2H),1.44(s,9H)。
第二步
将化合物G-1(150毫克,452.67微摩尔)溶于二氯甲烷(5毫升)和三氟乙酸(1毫升),在20℃下搅拌1小时。反应完成后,减压浓缩得到化合物G-2的三氟乙酸盐。MS-ESI计算值[M+H] +232,实测值232。
第三步
将化合物G-2(100毫克,432.43微摩尔,三氟乙酸盐)和化合物B-8(150.76毫克,864.86微摩尔)溶于甲醇(5毫升)中,然后加入N,N-二异丙基乙胺(55.89毫克,432.43微摩尔)和氰基硼氢化钠(67.94毫克,1.08毫摩尔),反应液在20℃下反应1小时。反应完成后,将反应液倒入饱和氯化铵(30毫升)水溶液中,用乙酸乙酯60毫升(30毫升×2)萃取,合并的有机相经饱和食盐水60毫升(30毫升×2)洗后,用无水硫酸钠干燥,过滤减压减压浓缩得到中间体G。MS-ESI计算值[M+H] +389,实测值389。
中间体K
Figure PCTCN2022072583-appb-000061
第一步
将化合物C-3(7克,32.26毫摩尔)和化合物K-1(5.30克,48.38毫摩尔)溶于甲醇(100毫升),加入N,N-二异丙基乙胺(8.34克,64.51毫摩尔)并在25℃下搅拌0.5小时,然后加入氰基硼氢化钠(2.43克,38.71毫摩尔)并在25℃下搅拌0.5小时。反应完毕后,小心向反应物中加入水(200毫升),然后用乙酸乙酯萃取(100毫升×3),有机相用无水硫酸钠干燥,过滤,浓缩。得到化合物K-2。MS-ESI计算值[M+H] +274和276,实测值274和276。
第二步
将化合物K-2(9克,32.83毫摩尔)溶于二氯甲烷(100毫升),加入叔丁基二甲基氯硅烷(12.37克,82.08毫摩尔)和咪唑(7.82克,114.92毫摩尔),混合物在25℃下搅拌16小时。反应完毕后,向反应物中加入水(100毫升)和二氯甲烷(100毫升)搅拌15分钟,分离出有机相用无水硫酸钠干燥,过滤,浓缩。残余物用硅胶柱层析(石油醚∶乙酸乙酯=10∶1到3∶1)分离得到中间体K。MS-ESI计算值[M+H] +388和390,实测值388和390。
中间体I
Figure PCTCN2022072583-appb-000062
将中间体K(7.4克,19.05毫摩尔)和中间体D(6.94克,19.05毫摩尔)溶于二氧六环(150毫升)和水(15毫升),然后加入[1,1-双(二苯基膦)二茂铁]二氯化钯(1.39克,1.91毫摩尔)和碳酸钾(7.90克,57.16毫摩尔),用氮气置换3次,反应液在氮气保护和65℃下搅拌4小时。反应完成后,用100毫升水稀释,并用乙酸乙酯(100毫升×2)萃取,有机相用饱和食盐水(100毫升×2)洗涤,并用无水硫酸钠干燥,过滤减压浓缩。剩余物经硅胶层析色谱柱(石油醚∶乙酸乙酯=50∶1到5∶1)分离得到中间体I。MS-ESI计算值[M+H] +545,实测值545。 1H NMR(400MHz,CDCl 3)δ=8.43(s,1H),7.56(dd,J=1.8,7.7Hz,1H),7.38(t,J=7.6Hz,1H),7.28(dd,J=1.8,7.5Hz,1H),7.15-7.04(m,1H),6.79(dd,J=1.5,8.1Hz,1H),6.66(dd,J=1.5,7.5Hz,1H),4.47(t,J=6.2Hz,1H),4.15(br s,2H),3.99(s,3H),3.81-3.76(m,2H),3.07-2.95(m,2H),0.90-0.81(m,10H),0.01(br s,6H).
中间体J
Figure PCTCN2022072583-appb-000063
第一步
将化合物B-4(1克,2.93毫摩尔)溶于二氧六环(20毫升),加入化合物A-2(968.66毫克,3.81毫摩尔),乙酸钾(719.94毫克,7.34毫摩尔)和1,1-双(二苯磷基)二茂铁二氯化钯(II)二氯甲烷复合物(239.62毫克,293.43微摩尔),反应液在氮气保护下加热到90℃反应14小时。反应完毕后,过滤,滤液用乙酸乙酯(60毫升)稀释,饱和食盐水(60毫升)洗涤。有机相用硫酸钠干燥,过滤,浓缩。粗品经硅胶柱层析纯化(石油醚∶乙酸乙酯=15∶1)分离得到化合物J-1。 1H NMR(400MHz,CDCl 3)δ=8.07(s,1H),4.61(s,2H),4.37-4.33(m,2H),3.65-3.55(m,2H),3.10-3.06(m,2H),1.44(s,9H),1.35-1.31(m,3H),1.17(s,12H).
第二步
将化合物J-1(300毫克,693.94微摩尔)溶于四氢呋喃(3毫升)和水(1.5毫升),加入过硼酸钠四水合物(427.08毫克,2.78毫摩尔),反应液在室温下反应1小时。反应完毕后,反应液用1摩尔/升的稀盐酸调pH等于6,乙酸乙酯(20毫升)萃取,有机相用硫酸钠干燥,过滤,浓缩。粗品经硅胶薄层色谱板(乙酸乙酯)分离得到中间体J。MS-ESI计算值[M+H] +323,实测值323。
实施例1
Figure PCTCN2022072583-appb-000064
第一步
将化合物D-1(2克,7.40毫摩尔)溶于四氢呋喃(20毫升),在零下78摄氏度和氮气保护下加入正丁基锂的正己烷溶液(3.26毫升,2.5摩尔每升),反应半小时后加入硼酸三甲酯(1.54克,14.80毫摩尔),反应液在室温下反应1小时。反应完毕后,反应液用稀盐酸(1摩尔/升,40毫升)淬灭,乙酸乙酯(40毫升)萃取。有机相用硫酸钠干燥,过滤,浓缩。粗品经硅胶色谱柱纯化(石油醚 乙酸乙酯=10∶1)分离得到化合物1-1。 1H NMR(400MHz,DMSO-D 6)δ=8.42-8.46(m,2H),7.70-7.73(m,1H),7.36-7.39(m,1H),7.22-7.24(m,1H).
第二步
将化合物1-1(800毫克,3.40毫摩尔)溶于二氧六环(8毫升)和水(0.8毫升),在氮气保护下加入化合物1-2(324.13毫克,1.89毫摩尔),碳酸钾(522.16毫克,3.78毫摩尔)和四(三苯基磷)钯(152.81毫克,132.24微摩尔),反应液在氮气保护下加热到95℃反应3小时。反应完毕后,过滤,滤液用乙酸乙酯(50毫升)稀释,饱和食盐水(50毫升)洗涤。有机相用硫酸钠干燥,过滤,浓缩。粗品经硅胶色谱柱纯化(石油醚∶乙酸乙酯=30∶1)分离得到化合物1-3。MS-ESI计算值[M+H] +326,实测值326。
第三步
将化合物1-4(337.58毫克,2.24毫摩尔)溶于二氯甲烷(6毫升)和乙醇(6毫升),加入N,N-二 异丙基乙胺(482.81毫克,3.74毫摩尔)和化合物1-3(610毫克,1.87毫摩尔),搅拌半小时后加入氰基硼氢化钠(1.19克,5.60毫摩尔),反应液在室温下反应16小时。反应完毕后,反应液用二氯甲烷(50毫升)稀释,饱和食盐水洗涤(50毫升)。有机相用无水硫酸钠干燥,过滤,浓缩,得到化合物1-5。MS-ESI计算值[M+H] +424,实测值424。
第四步
将化合物1-5(550毫克,1.29毫摩尔)溶于二氯甲烷(11毫升),加入二碳酸二叔丁酯(423.94毫克,1.94毫摩尔)和三乙胺(327.59毫克,3.24毫摩尔),反应液在室温下反应0.5小时。反应完毕后,反应液用用水(80毫升)稀释,二氯甲烷(80毫升×2)萃取,有机相用饱和食盐水洗涤(50毫升)。无水硫酸钠干燥,过滤,浓缩,得到化合物1-6。MS-ESI计算值[M+H] +526,实测值526。 1H NMR(400MHz,CD 3OD)δ=7.60(dd,J=7.88,1.50Hz,1H)7.39(br d,J=1.63Hz,2H)7.23-7.31(m,1H)7.07-7.18(m,2H)4.34-4.48(m,2H)3.90-3.94(m,3H)3.83(br s,1H)3.24-3.31(m,2H)2.04-2.35(m,4H)1.52(s,9H).
第五步
将化合物1-6(413毫克,786.92微摩尔)溶于二氧六环(8毫升)和水(0.8毫升)溶液中,加入中间体A(299.26毫克,1.18毫摩尔)和碳酸钾(326.27毫克,2.36毫摩尔)和1,1-双(二苯基磷)二茂铁氯化钯(57.58毫克,78.69微摩尔)。通过3次置换氮气后,反应液在85℃下反应4小时。反应完毕后,反应液用水(40毫升)稀释,乙酸乙酯(40毫升×2)萃取,有机相用饱和食盐水洗涤(10毫升)。无水硫酸钠干燥,过滤,浓缩,得到粗品经高效液相色谱(色谱柱:Phenomenex luna C18 150×40毫米×15微米,流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:50%-80%,10分钟)分离得到化合物1-7。MS-ESI计算值[M+H] +571,实测值571。
第六步
将化合物1-7(80毫克,139.98微摩尔)和中间体B(60.76毫克,153.98微摩尔)溶于四氢呋喃(4毫升),零度氮气保护下加入双三甲硅基氨基锂的四氢呋喃溶液(1摩尔每升,419.95微升),反应液在零度下反应1小时。反应结束后,反应液用饱和氯化铵溶液(30毫升)淬灭,乙酸乙酯(30毫升)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。粗品经硅胶薄层色谱板(二氯甲烷∶甲醇=10∶1)纯化分离得到化合物1-8。MS-ESI计算值[M+H] +919,实测值919。
第七步
将化合物1-8(101毫克,109.78微摩尔)溶于二氯甲烷(2毫升),加入氯化氢的乙酸乙酯溶液(4摩尔/升,2毫升),室温搅拌1小时。反应完毕后,浓缩。粗品经高效液相色谱(色谱柱:Unisil 3-100 C18 Ultra 150*50毫米*3微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:15%-35%,10分钟)分离得到化合物1的甲酸盐。MS-ESI计算值[M+H] +705,实测值705。 1H NMR(400MHz,CD 3OD)δ=8.61(d,J=8.4,1H),8.32-8.45(m,2H),7.83-7.88(m,1H),7.77(s,1H),7.65-7.70(m,1H),7.46-7.57(m,2H),7.38-7.43(m,1H),7.31-7.36(m,1H),7.13-7.21(m,1H),4.14-4.21(m,2H),4.04-4.12(m,8H),3.97-4.02(m,1H),3.84-3.90(m,2H),3.14-3.22(m,2H),2.94-3.09(m,6H),2.33-2.46(m,3H),1.83-1.97(m,1H).
实施例2
Figure PCTCN2022072583-appb-000065
第一步
将化合物2-1(17.90毫克,205.47微摩尔)溶于二氯甲烷(3毫升),加入N,N-二异丙基乙胺(35.41毫克,273.96微摩尔)和中间体C(99毫克,136.98微摩尔),搅拌半小时后加入氰基硼氢化钠(87.10毫克,410.94微摩尔),反应液在室温下反应12小时。反应完毕后,水(20毫升)稀释,用二氯甲烷(25毫升×2)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。残余物经硅胶薄层色谱板(二氯甲烷 甲醇=12∶1)分离得到化合物2-2。MS-ESI计算值[M+H] +793,实测值793。
第二步
将化合物2-2(62毫克,78.10微摩尔)溶于二氯甲烷(3毫升),加入氯化氢的乙酸乙酯溶液(4摩尔/升,1.5毫升),25℃搅拌0.5小时。反应完毕后,浓缩。粗品经高效液相色谱(色谱柱:Phenomenex Gemini-NX C18 75×30毫米×3微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:10%-40%,8分钟)分离得到化合物2。MS-ESI计算值[M+H] +679,实测值679。 1H NMR(400MHz,CD 3OD)δ=8.62(dd,J=8.25,1.50Hz,1H),8.50(s,1H),7.71-7.76(m,2H),7.58(t,J=7.63Hz,1H),7.44-7.52(m,2H),7.18(dd,J=7.63,1.50Hz,1H),4.47-4.54(m,1H),4.31(s,2H),4.11(s,3H),4.04(s,3H),3.79-3.88(m,4H),3.37(br s,1H),3.30(br d,J=5.38Hz,1H),3.14-3.25(m,1H),3.10(br d,J=11.01Hz,1H),2.85-2.95(m,4H),2.76-2.83(m,2H),2.20-2.31(m,1H),1.89-2.00(m,1H).
实施例3
Figure PCTCN2022072583-appb-000066
Figure PCTCN2022072583-appb-000067
第一步
将化合物3-1(24.61毫克,207.55微摩尔)溶于二氯甲烷(3毫升),加入N,N-二异丙基乙胺(35.76毫克,276.73微摩尔)和中间体C(100毫克,138.36微摩尔),搅拌半小时后加入氰基硼氢化钠(87.97毫克,415.09微摩尔),反应液在室温下反应1小时。反应完毕后,水(20毫升)稀释,用二氯甲烷(25毫升×2)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。残余物经硅胶薄层色谱板(二氯甲烷 甲醇=13∶1)分离得到化合物3-2。MS-ESI计算值[M+H] +788,实测值788。
第二步
将化合物3-2(83毫克,105.22微摩尔)溶于二氯甲烷(3毫升),加入氯化氢的乙酸乙酯溶液(4摩尔/升,1.5毫升),25℃搅拌0.5小时。反应完毕后,浓缩。粗品经高效液相色谱(色谱柱:Phenomenex Gemini-NX C18 75×30毫米×3微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:20%-30%,7分钟)分离得到化合物3。MS-ESI计算值[M+H] +692,实测值692。 1H NMR(400MHz,CD 3OD)δ=8.62(dd,J=8.32,1.19Hz,1H),8.43-8.47(m,1H),7.70-7.76(m,2H),7.57(t,J=7.63Hz,1H),7.43-7.52(m,2H),7.17(dd,J=7.63,1.25Hz,1H),4.25-4.31(m,2H),4.02-4.14(m,8H),3.88-3.97(m,4H),3.83(t,J=5.82Hz,2H),3.50-3.60(m,1H),2.94-3.00(m,2H),2.81-2.92(m,4H).
实施例4
Figure PCTCN2022072583-appb-000068
Figure PCTCN2022072583-appb-000069
第一步
将中间体C(80毫克,110.69微摩尔)和化合物4-1(28.16毫克,221.38微摩尔)溶于二氯甲烷(2毫升),然后加入三乙氧基硼氢化钠(58.65毫克,276.73微摩尔),在20℃条件下反应2小时。反应完毕后,反应液过滤浓缩,粗品经用硅胶薄层色谱板纯化(二氯甲烷∶甲醇=10∶1)得到化合物4-2。MS-ESI计算值[M+H] +833,实测值833。
第二步
将化合物4-2(30毫克,35.97微摩尔)溶于乙酸乙酯(1毫升),加入氯化氢的乙酸乙酯溶液(4摩尔/升,44.97微升),25℃搅拌0.5小时。反应完毕后,反应液过滤浓缩,粗品经高效液相色谱(色谱柱:Shim-pack C18 150*25毫米*10微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:11%-41%,10分钟)分离得到化合物4的甲酸盐。MS-ESI计算值[M+H] +719,实测值719。 1H NMR(400MHz,CD 3OD)δppm 8.61(dd,1H),8.57(s,1H),8.38-8.48(m,2H),7.70-7.77(m,2H),7.59(t,1H),7.44-7.52(m,2H),7.17(dd,1H),4.48(s,2H),4.09-4.19(m,6H),4.04(s,3H),3.94(s,2H),3.83(t,2H),2.99-3.08(m,2H),2.89(q,4H),2.64(br d,2H),2.45(br d,2H),2.28-2.40(m,2H),2.09(br d,2H).
实施例5
Figure PCTCN2022072583-appb-000070
第一步
将化合物5-1(25.65毫克,207.55微摩尔)溶于二氯甲烷(3毫升),加入N,N-二异丙基乙胺(35.76毫克,276.73微摩尔)和中间体C(100毫克,138.36微摩尔),搅拌半小时后加入氰基硼氢化钠(87.97毫克,415.09微摩尔),反应液在室温下反应1小时。反应完毕后,水(20毫升)稀释,用二氯甲烷(25毫升×2)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。残余物经硅胶薄层色谱板(二氯甲烷 甲醇=13∶1)分离得到化合物5-2。MS-ESI计算值[M+H] +793,实测值793。
第二步
将化合物5-2(62毫克,78.10微摩尔)溶于二氯甲烷(3毫升),加入氯化氢的乙酸乙酯溶液(4摩尔/升,1.5毫升),25℃搅拌0.5小时。反应完毕后,浓缩。粗品经高效液相色谱(色谱柱:Phenomenex Gemini-NX C18 75×30毫米×3微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:18%-28%,7分钟)分离得到化合物5。MS-ESI计算值[M+H] +679,实测值679。 1H NMR(400MHz,CD 3OD)δ=8.62(dd,J=8.28,1.25Hz,1H),8.44-8.47(m,1H),7.70-7.77(m,2H),7.54-7.60(m,1H),7.43-7.53(m,2H),7.18(dd,J=7.59,1.19Hz,1H),4.27-4.33(m,2H),3.97-4.14(m,8H),3.85(t,J=5.65Hz,2H),3.66(s,2H),3.04-3.11(m,2H),2.89-2.96(m,4H),0.88-0.94(m,2H),0.72-0.78(m,2H).
实施例6
Figure PCTCN2022072583-appb-000071
第一步
将中间体C(110毫克,152.20微摩尔)溶于二氯甲烷(3毫升),加入化合物6-1(17.15毫克,228.30微摩尔),三乙氧基硼氢化钠(96.77毫克,456.60微摩尔)。反应液在室温下反应2小时。反应完毕后,反应液用水(10毫升)淬灭,二氯甲烷(5毫升)稀释,二氯甲烷(10毫升×3)萃取,有机相用饱和食盐水(20毫升×2)洗涤,硫酸钠干燥,过滤,浓缩。粗品经硅胶薄层色谱板纯化(二氯甲烷∶甲醇=8∶1)分离得到化合物6-2(80毫克,粗品)。MS-ESI计算值[M+H] +781,实测值781。
第二步
将化合物6-2(80毫克,102.32微摩尔)溶于二氯甲烷(2毫升),加入氯化氢的乙酸乙酯溶液(4摩 尔/升,1毫升),室温搅拌0.25小时。反应完毕后,浓缩。粗品经高效液相色谱(色谱柱:Xtimate C18 150*25毫米*10微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:7%-37%,10分钟)分离得到化合物6的甲酸盐。MS-ESI计算值[M+H] +667,实测值667。 1H NMR(400MHz,CD 3OD)δ=8.62(dd,J=1.5,8.3Hz,1H),8.55(s,1H),8.52-8.49(m,1H),7.76-7.71(m,2H),7.59(t,J=7.6Hz,1H),7.53-7.45(m,2H),7.18(dd,J=1.5,7.6Hz,1H),4.34-4.25(m,2H),4.13(s,3H),4.05(s,3H),3.86-3.79(m,4H),3.74(t,J=5.9Hz,2H),3.16(t,J=7.2Hz,2H),2.93-2.85(m,4H),2.79(t,J=5.9Hz,2H),1.95(quin,J=6.6Hz,2H).
实施例7
Figure PCTCN2022072583-appb-000072
第一步
将中间体C(100毫克,138.36微摩尔)溶于二氯甲烷(3毫升),加入化合物7-1(25.64毫克,345.91微摩尔),三乙氧基硼氢化钠(87.98毫克,415.09微摩尔)。反应液在室温下反应1小时后再加入中间体7-1(10.26毫克,138.36微摩尔),三乙氧基硼氢化钠(29.33毫克,138.36微摩尔)。反应液在室温下反应1小时。反应完毕后,反应液用水(10毫升)淬灭,二氯甲烷(5毫升)稀释,二氯甲烷(10毫升×3)萃取,有机相用饱和食盐水(20毫升×2)洗涤,硫酸钠干燥,过滤,浓缩。粗品经硅胶薄层色谱板纯化(二氯甲烷∶甲醇=10∶1)分离得到化合物7-2。MS-ESI计算值[M+H] +780,实测值780。
第二步
将化合物7-2(35毫克,44.82微摩尔)溶于二氯甲烷(2毫升),加入氯化氢的乙酸乙酯溶液(4摩尔/升,1毫升),室温搅拌0.25小时。反应完毕后,浓缩。粗品经高效液相色谱(色谱柱:Xtimate C18 150*25毫米*10微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:15%-35%,10分钟)分离得到化合物7的甲酸盐。MS-ESI计算值[M+H] +666,实测值666。 1H NMR(400MHz,CD 3OD)δ=8.60(brd,J=8.4Hz,1H),8.55(s,1H),8.46-8.40(m,1H),7.75-7.67(m,2H),7.56(brt,J=7.5Hz,1H),7.51-7.41(m,2H),7.18-7.13(m,1H),4.08(s,3H),4.03(br d,J=3.8Hz,5H),3.80(br d,J=7.2Hz,4H),3.04(br d,J=5.5Hz,2H),2.97(br d,J=5.6Hz,2H),2.91-2.84(m,4H),2.76(br t,J=5.7Hz,2H), 2.67(s,3H).
实施例8
Figure PCTCN2022072583-appb-000073
第一步
将化合物8-1(28.21毫克,228.30微摩尔)溶于二氯甲烷(3毫升),加入N,N-二异丙基乙胺(39.34毫克,304.40微摩尔)和中间体C(110毫克,152.20微摩尔),搅拌半小时后加入氰基硼氢化钠(96.77毫克,456.60微摩尔),反应液在室温下反应12小时。反应完毕后,水(20毫升)稀释,用二氯甲烷(25毫升×2)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。残余物经硅胶薄层色谱板(二氯甲烷 甲醇=10∶1)分离得到化合物8-2。MS-ESI计算值[M+H] +793,实测值793。
第二步
将化合物8-2(68毫克,85.66微摩尔)溶于二氯甲烷(3毫升),加入氯化氢的乙酸乙酯溶液(4摩尔/升,1.5毫升),25℃搅拌0.5小时。反应完毕后,浓缩。粗品经高效液相色谱(色谱柱:Phenomenex Gemini-NX C18 75×30毫米×3微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:10%-40%,7分钟)分离得到化合物8的甲酸盐。MS-ESI计算值[M+H] +679,实测值679。 1H NMR(400MHz,CD 3OD)δ=8.58-8.65(m,1H),8.49(s,1H)8.43(br s,1H),7.70-7.78(m,2H),7.59(t,J=7.63Hz,1H),7.45-7.53(m,2H),7.18(dd,J=7.63,1.38Hz,1H),4.52(s,2H),4.30-4.41(m,3H),4.12(s,3H),4.06(s,3H),3.94-4.01(m,4H),3.81-3.87(m,2H),3.35-3.37(m,3H),3.02-3.07(m,2H),2.91(q,J=5.38Hz,4H).
实施例9
Figure PCTCN2022072583-appb-000074
Figure PCTCN2022072583-appb-000075
第一步
将化合物3-1(25.84毫克,217.92微摩尔)溶于二氯甲烷(3毫升),加入N,N-二异丙基乙胺(37.55毫克,290.56微摩尔)和中间体C(105毫克,145.28微摩尔),搅拌半小时后加入氰基硼氢化钠(92.37毫克,435.85微摩尔),反应液在室温下反应1小时。反应完毕后,水(20毫升)稀释,用二氯甲烷(25毫升×2)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。残余物经硅胶薄层色谱板(二氯甲烷 甲醇=14 1)分离得到化合物9-1。MS-ESI计算值[M+H] +788,实测值788。
第二步
将化合物9-1(51毫克,64.65微摩尔)溶于三氟乙酸(3毫升),25℃搅拌16小时。反应液经过LC-MS检测后发现有原料没有反应完,将反应液升温到30℃搅拌4小时,反应完毕后,浓缩。粗品经高效液相色谱(色谱柱:Phenomenex Gemini-NX C18 75×30毫米×3微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:10%-40%,7分钟)分离得到化合物9。MS-ESI计算值[M+H] +674,实测值674。 1H NMR(400MHz,CD 3OD)δ=8.60-8.63(m,1H),8.42(s,1H),7.83(s,1H),7.72(dd,J=7.69,1.69Hz,1H),7.55-7.60(m,1H),7.51(t,J=7.94Hz,1H),7.45(dd,J=7.57,1.69Hz,1H),7.19(dd,J=7.63,1.63Hz,1H),4.30-4.39(m,2H),4.07-4.10(m,6H),3.94(br t,J=5.38Hz,4H),3.81-3.90(m,2H),3.69(t,J=7.25Hz,2H),3.52-3.60(m,1H),3.44(br s,2H),3.25(br d,J=4.50Hz,2H),3.06(brt,J=5.88Hz,2H).
实施例10
Figure PCTCN2022072583-appb-000076
Figure PCTCN2022072583-appb-000077
第一步
将化合物L-1(1.72克,13.95毫摩尔)溶于二氯甲烷(20毫升),向反应液中加入化合物10-1(2克,9.30毫摩尔),N,N-二异丙基乙胺(3.61克,27.90毫摩尔),在25℃下搅拌0.5小时。向反应液中加入三乙酰氧基硼氢化钠(5.91克,27.90毫摩尔),在25℃下搅拌2小时。反应完毕后,向反应液中加入水(100毫升),用二氯甲烷(50毫升×3)萃取,有机相用饱和食盐水(50毫升×2)洗涤,合并有机相用无水硫酸钠干燥,过滤,减压浓缩得到粗产物。粗产物经过硅胶层析柱(二氯甲烷∶甲醇=10∶1)分离得到化合物10-2。MS-ESI计算值[M+H] +288,实测值288。
第二步
将化合物10-2(1.7克,5.94毫摩尔)溶于二氯甲烷(20毫升)向反应液中加入咪唑(1.21克,17.82毫摩尔),叔丁基二甲基氯硅烷(1.79克,17.82毫摩尔)和N,N-二甲氨基吡啶(145.15毫克,1.19毫摩尔),在氮气保护下45℃搅拌10小时。反应完毕后,反应液过滤,减压浓缩得到粗产物。粗产物经过硅胶层析柱(石油醚∶乙酸乙酯=3∶1)分离得到化合物10-3。
MS-ESI计算值[M+H] +402,实测值402。
第三步
将化合物10-3(1.6克,4.00毫摩尔),中间体D(1.45克,4.00毫摩尔),碳酸钾(1.66克,11.99毫摩尔)1,1-双(二苯基磷)二茂铁氯化钯(292.37毫克,399.58微摩尔)溶于二氧六环(20毫升)和水(2毫升)在氮气保护下75℃搅拌10小时。反应完毕后,反应液过滤,减压浓缩得到粗产物,粗产物经过硅胶层析柱(石油醚∶乙酸乙酯=8∶1到3∶1)分离得到化合物10-4。MS-ESI计算值[M+H] +557,实测值557。
第四步
将化合物10-4(200.0毫克,358.66微摩尔)溶于四氢呋喃(3毫升),向反应液中加入中间体B(183.98毫克,466.26微摩尔),二(三甲基硅)氨基锂(1摩尔/升的正己烷溶液,1.08毫升),在氮气保护下0℃搅拌1小时。反应完毕后,向反应液中加入饱和氯化铵(20毫升),用乙酸乙酯(20毫升×3)萃取,合并有机相用无水硫酸钠干燥,过滤,减压浓缩得到粗产物。粗产物经过硅胶薄层色谱板(石油醚∶ 乙酸乙酯=1∶1)分离得到化合物10-5。MS-ESI计算值[M+H] +905,实测值905。
第五步
将化合物10-5(110.0毫克,121.39微摩尔)溶于二氯甲烷(1毫升),接着向反应液中加入氯化氢的乙酸乙酯溶液(4摩尔/升,5.5毫升),在25℃下搅拌0.5小时。反应完毕后,浓缩,粗品经高效液相色谱(色谱柱:Waters Xbridge C18 150*25毫米*5微米;流动相:流动相A:体积分数10毫摩尔碳酸氢铵的水溶液;流动相B:乙腈;B%:54%-84%,10分钟)分离得到化合物10。MS-ESI计算值[M+H] +677,实测值677。 1H NMR(400MHz,CD 3OD)δ=8.58(dd,J=1.4,8.3Hz,1H),7.70(s,1H),7.49-7.41(m,3H),7.33-7.27(m,2H),7.13(dd,J=1.5,7.6Hz,1H),7.05-6.97(m,2H),4.01(s,3H),3.86(s,3H),3.82-3.73(m,6H),3.40(d,J=8.9Hz,2H),3.21(d,J=8.6Hz,2H),2.86(br dd,J=4.4,10.8Hz,4H),2.76(t,J=5.9Hz,2H),1.46(s,3H).
实施例11
Figure PCTCN2022072583-appb-000078
第一步
将化合物L-1(858.07毫克,6.94微摩尔)溶于二氯甲烷(15毫升),加入N,N-二异丙基乙胺(1.20克,9.26微摩尔)和化合物11-1(1克,4.63毫摩尔),搅拌半小时后加入氰基硼氢化钠(2.94克,13.89毫摩尔),反应液在25℃下反应14小时。反应完毕后,用二氯甲烷(120毫升×2)稀释,用水(100毫升)洗涤。有机相用无水硫酸钠干燥,过滤,浓缩。残渣经硅胶层析柱(乙酸乙酯 甲醇=30 1)分离得到化合物11-2。MS-ESI计算值[M+H] +287,实测值287。
第二步
将化合物11-2(1.1克,3.83毫摩尔)溶于二氯甲烷(20毫升),加入N,N-二甲氨基吡啶(93.60毫克,766.14微摩尔)和咪唑(1.04克,15.32毫摩尔),反应液在0℃和氮气保护下加入叔丁基二甲基氯硅烷(1.73克,11.49微摩尔)。反应液在45℃和氮气保护下反应16小时。LC-MS监测反应原料剩 余,有产物生成。补加N,N-二甲氨基吡啶(93.60毫克,766.14微摩尔)和咪唑(1.04克,15.32毫摩尔),反应液在0℃氮气保护下加入叔丁基二甲基氯硅烷(1.73克,11.49微摩尔)。反应液在45℃氮气保护下反应14小时。反应完毕后,用水(100毫升)稀释,二氯甲烷(110毫升×2)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。粗品经高效液相色谱(色谱柱:Kromasil Eternity XT 250×80毫米×10微米,流动相:流动相A:体积分数0.05%氨溶液;流动相B:乙腈;B%:35%-95%,20分钟)分离得到粗品后,再通过硅胶薄层色谱板(二氯甲烷∶甲醇=12∶1)纯化分离得到化合物11-3。MS-ESI计算值[M+H] +401,实测值403。
第三步
将化合物11-3(90毫克,224.21微摩尔)溶于二氧六环(3毫升)和水(0.3毫升)中,加入中间体D(106.12毫克,291.47微摩尔)和碳酸钾(92.96毫克,672.62微摩尔)和1,1-双(二苯基磷)二茂铁氯化钯(24.61毫克,33.63微摩尔),反应液在65℃氮气保护下搅拌8小时。反应完毕后,反应液经过滤后,滤液用水(40毫升)稀释,用乙酸乙酯(50毫升×2)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。经硅胶薄层色谱板(二氯甲烷∶甲醇=12∶1)分离得到化合物11-4。MS-ESI计算值[M+H] +558,实测值558。
第四步
将化合物11-4(70毫克,125.31微摩尔)和中间体B(64.28毫克,162.90微摩尔)溶于四氢呋喃(2毫升),零度氮气保护下加入双三甲硅基氨基锂的四氢呋喃溶液(1摩尔每升,375.93微升),反应液在25℃下反应1小时。反应结束后,反应液用饱和氯化铵溶液(20毫升)淬灭,乙酸乙酯(25毫升×2)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。粗品经硅胶薄层色谱板(二氯甲烷 甲醇=12 1)纯化分离得到化合物11-5。MS-ESI计算值[M+H] +907,实测值906。
第五步
将化合物11-5(50毫克,55.12微摩尔)溶于二氯甲烷(5毫升),加入氯化氢的乙酸乙酯溶液(4摩尔/升,1毫升),反应液在25℃下反应0.5小时。反应完毕后,浓缩。粗品经高效液相色谱(色谱柱:Phenomenex Gemini-NX C18 75×30毫米×3微米,流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:10%-40%,7分钟)分离得到化合物11。MS-ESI计算值[M+H] +678,实测值678。 1H NMR(400MHz,CD 3OD)δ=8.61(dd,J=8.31,1.47Hz,1H),8.18-8.24(m,1H),7.72(s,1H),7.40-7.56(m,5H),7.16(dd,J=7.58,1.47Hz,1H),4.12(s,2H),4.04(s,3H),3.96(s,3H),3.78-3.84(m,4H),3.70(br d,J=9.54Hz,2H),3.53(br d,J=9.29Hz,2H),2.88(br dd,J=12.53,4.71Hz,4H),2.78(t,J=5.87Hz,2H),1.51(s,3H).
实施例12
Figure PCTCN2022072583-appb-000079
Figure PCTCN2022072583-appb-000080
第一步
将中间体C(80.0毫克,111微摩尔),12-1(36.2毫克,221微摩尔)溶于二氯甲烷(2毫升),向反应液中加入N,N-二异丙基乙胺(28.6毫克,221微摩尔),在20℃下搅拌1小时。向反应液中加入三乙酰氧基硼氢化钠(70.4毫克,332微摩尔),在20℃下搅拌2小时。反应完毕后,浓缩的残渣经硅胶薄层色谱板(二氯甲烷∶甲醇=10∶1)分离纯化得到化合物12-2。MS-ESI计算值[M+H] +833,实测值833。
第二步
将中间体12-2(30.0毫克,29.3微摩尔)溶于二氯甲烷(2毫升),接着向反应液中加入氯化氢的乙酸乙酯溶液(4摩尔/升,500微升),在20℃下搅拌0.5小时。反应完毕后,浓缩,粗品经高效液相色谱(色谱柱:Phenomenex Gemini-NX C18 75*30毫米*3微米;流动相:流动相A:体积分数0.225%甲酸的水溶液;流动相B:乙腈;B%:10%-40%,7分钟)分离得到化合物12。MS-ESI计算值[M+H] +719,实测值719。 1H NMR(400MHz,CD 3OD)δ=8.63(dd,J=1.5,8.3Hz,1H),8.47(s,1H),7.79-7.66(m,2H),7.58(t,J=7.6Hz,1H),7.52-7.43(m,2H),7.22-7.15(m,1H),4.32(s,2H),4.10(s,3H),4.05(s,3H),3.99(br d,J=13.6Hz,4H),3.88-3.76(m,4H),3.01-2.84(m,4H),2.80(t,J=5.9Hz,2H),2.45-2.20(m,4H),1.29(s,3H).
实施例13
Figure PCTCN2022072583-appb-000081
Figure PCTCN2022072583-appb-000082
第一步
将化合物B-6(300毫克,892微摩尔),中间体E(334毫克,595微摩尔)溶于四氢呋喃(6毫升),接着在0℃下向反应液中加入双(三甲硅基)氨基锂(1摩尔/升,1.49毫升),在15℃下搅拌2小时。反应完毕后,将饱和氯化铵水溶液(20毫升)加入到反应液中,乙酸乙酯萃取(15毫升×3),有机相用饱和食盐水(20毫升×3)洗涤,无水硫酸钠干燥,过滤,浓缩的残渣经硅胶薄层色谱板(石油醚∶乙酸乙酯=2∶1)分离得到化合物13-1。MS-ESI计算值[M+H] +849,实测值849。
第二步
将化合物13-1(200毫克,235.32微摩尔)溶于二氯甲烷(3毫升)中,然后加入三氟乙酸(924.00毫克,8.10毫摩尔,0.6毫升),反应液在15℃下反应0.5小时。反应完成后,将反应液减压浓缩得到化合物13-2。MS-ESI计算值[M+H] +749,实测值749。
第三步
将化合物13-2(100毫克,133.37微摩尔)和高氯酸锂(28.38毫克,266.74微摩尔)溶于乙醇(1.5毫升)中,然后加入化合物13-3(28.85毫克,400.11微摩尔),反应液置于封管中,80℃下搅拌6小时后。反应完成后,减压浓缩,剩余物经硅胶薄层色谱板(石油醚∶乙酸乙酯=1∶1)分离得到化合物13-4。MS-ESI计算值[M+H] +821,实测值821。
第四步
将化合物13-4(90毫克,109.50微摩尔)溶于二氯甲烷(1毫升)中,加入氯化氢的乙酸乙酯溶液(4摩尔/升,0.2毫升),反应液在25℃下反应1小时。反应完成后,减压浓缩得到残渣经高效液相色谱(色谱柱:Phenomenex luna C18 150*25毫米*10微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:7%-37%,10分钟)分离得到化合物13的甲酸盐。MS-ESI计算值[M+H] +707,实测值707。 1H NMR(400MHz,CD 3OD)δ=8.63(dd,J=1.4,8.3Hz,1H),8.50(s,1H),8.44(s,1H),7.78-7.68(m,2H),7.59(t,J=7.7Hz,1H),7.53-7.41(m,2H),7.17(dd,J=1.4,7.7Hz,1H),4.59(s,2H),4.19(d,J=10.9Hz,2H),4.13(s,3H),4.08-3.99(m,5H),3.94(s,2H),3.07-2.94(m,2H),2.91-2.80(m,2H),2.61(s,2H),1.58(s,3H),1.26(s,6H).
实施例14
Figure PCTCN2022072583-appb-000083
第一步
将化合物14-1(43.45毫克,345.91微摩尔)溶于二氯甲烷(3毫升),加入N,N二异丙基乙胺(53.65毫克,415.09微摩尔),中间体C(100毫克,138.36微摩尔)。反应液在室温下搅拌0.5小时后加入三乙氧基硼氢化钠(87.98毫克,415.09微摩尔)。反应液在室温下反应1小时。反应完毕后,反应液用水(10毫升)淬灭,二氯甲烷(5毫升)稀释,二氯甲烷(10毫升×3)萃取,有机相用饱和食盐水(20毫升×2)洗涤,硫酸钠干燥,过滤,浓缩。粗品经硅胶薄层色谱板纯化(二氯甲烷 甲醇=10 1)分离得到化合物14-2。MS-ESI计算值[M+H] +795,实测值795。
第二步
将化合物14-2(90毫克,113.08微摩尔)溶于二氯甲烷(2毫升),加入氯化氢的乙酸乙酯溶液(4摩尔/升,1毫升),室温搅拌0.25小时。反应完毕后,浓缩。粗品经高效液相色谱(色谱柱:Xtimate C18 150*25毫米*10微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:9%-39%,10分钟)分离得到化合物14。MS-ESI计算值[M+H] +681,实测值681。 1H NMR(400MHz,CD 3OD)δ=8.62(dd,J=1.5,8.3Hz,1H),8.55(s,1H),8.52-8.49(m,1H),7.76-7.71(m,2H),7.59(t,J=7.6Hz,1H),7.53-7.45(m,2H),7.18(dd,J=1.5,7.6Hz,1H),4.34-4.25(m,2H),4.13(s,3H),4.05(s,3H),3.86-3.79(m,4H),3.74(t,J=5.9Hz,2H),3.16(t,J=7.2Hz,2H),2.93-2.85(m,4H),2.79(t,J=5.9Hz,2H),1.95(quin,J=6.6Hz,2H).
实施例15
Figure PCTCN2022072583-appb-000084
Figure PCTCN2022072583-appb-000085
第一步
将中间体F(150毫克,197.18微摩尔)溶于1,2-二氯乙烷(2毫升),加入二氧化锰(171.42毫克,1.97毫摩尔),反应液在氮气保护下加热到90℃,反应2小时后加入二氧化锰(171.42毫克,1.97毫摩尔)。反应液在氮气保护下加热到90℃反应2小时。反应完毕后,过滤,浓缩。得到化合物15-1。MS-ESI计算值[M+H] +758,实测值758。
第二步
将化合物2-1(34.21毫克,276.78微摩尔)溶于二氯甲烷(3毫升),N,N二异丙基乙胺(35.77毫克,276.78微摩尔),化合物15-1(140毫克,184.52微摩尔),室温下搅拌0.5小时后加入三乙氧基硼氢化钠(114.37毫克,539.61微摩尔)。反应液在室温下反应1小时。反应完毕后,反应液用水(10毫升)淬灭,二氯甲烷(5毫升)稀释,二氯甲烷(10毫升×3)萃取,有机相用饱和食盐水(20毫升×2)洗涤,硫酸钠干燥,过滤,浓缩。粗品经硅胶薄层色谱板纯化(二氯甲烷∶甲醇=10∶1)分离得到中间体15-2。MS-ESI计算值[M+H] +829,实测值829。
第三步
将化合物15-2(75毫克,90.38微摩尔)二氯甲烷(2毫升),加入氯化氢的乙酸乙酯溶液(4摩尔/升,1毫升),室温搅拌0.25小时。反应完毕后,浓缩。粗品经高效液相色谱(色谱柱:Xtimate C18 150*25毫米*10微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:7%-37%,10分钟)分离得到化合物15。MS-ESI计算值[M+H] +715,实测值715。 1H NMR(400MHz,CD 3OD)δ=8.59(dd,J=1.5,8.3Hz,1H),8.46(s,1H),7.82(s,1H),7.74-7.68(m,1H),7.56(t,J=7.6Hz,1H),7.51-7.42(m,2H),7.29-7.22(m,1H),7.20-7.14(m,1H),7.34-6.95(m,1H),4.49-4.36(m,1H),4.11(s,2H),4.08(s,3H),3.91-3.85(m,2H),3.83-3.76(m,2H),3.20-3.07(m,2H),3.01-2.84(m,6H),2.81-2.74(m,2H),2.28-2.12(m,1H),1.89-1.78(m,1H).
实施例16
Figure PCTCN2022072583-appb-000086
第一步
将化合物16-1(500毫克,2.85毫摩尔)溶于N,N-二甲基甲酰胺(6毫升),向反应液中加入碳酸钾 (472毫克,3.42毫摩尔),碘乙烷(578毫克,3.71毫摩尔),在50℃下搅拌2小时。反应完毕后,加水(50毫升)稀释,乙酸乙酯萃取(15毫升×3),有机相用碳酸氢钠水溶液(20毫升×3)洗涤,有机相接着用饱和食盐水(20毫升×3)洗涤,无水硫酸钠干燥,过滤,浓缩得到化合物16-2。MS-ESI计算值[M+H] +204,实测值204。 1H NMR(400MHz,CDCl 3)δ=8.51-8.35(m,1H),7.09(dd,J=5.7,8.0Hz,1H),4.47(q,J=7.2Hz,2H),1.42(t,J=7.2Hz,3H)。
第二步
将化合物16-2(12.5克,61.40毫摩尔)溶于N,N-二甲基甲酰胺(80毫升),向反应液中加入醋酸铯(12.96克,67.54毫摩尔),氮气置换三次,然后在氮气保护下80℃搅拌2小时。TLC检测反应原料剩余,向反应液中加入醋酸铯(7.07克,36.84毫摩尔),氮气置换三次,然后在氮气保护下80℃搅拌2小时。加水(400毫升)稀释,乙酸乙酯萃取(200毫升×3),有机相接着用饱和食盐水(200毫升×2)洗涤,无水硫酸钠干燥,过滤减压浓缩得到的粗品经硅胶柱层析(石油醚∶乙酸乙酯=100∶1到10∶1)纯化分离得到化合物16-3。MS-ESI计算值[M+H] +202,实测值202。
第三步
将化合物16-3(7克,34.72毫摩尔)和化合物F-3(10.59克,69.44毫摩尔)溶于N,N二甲基甲酰胺(70毫升),然后加入碳酸钾(14.40克,104.16毫摩尔),反应液80℃下搅拌2小时。向反应液中加入90毫升水稀释,然后用乙酸乙酯(60毫升×3)萃取,合并后有机相用饱和食盐水(30毫升×3)洗涤,并用无水硫酸钠干燥。过滤减压浓缩得到的粗品经硅胶柱层析(石油醚∶乙酸乙酯=30∶1到10∶1)纯化分离得到化合物16-4。MS-ESI计算值[M+H] +252,实测值252。 1H NMR(400MHz,CDCl 3)δ=8.62-8.33(m,1H),7.19-7.03(m,1H),6.95-6.40(m,1H),4.65-4.28(m,2H),1.50-1.26(m,3H).
第四步
将化合物16-4(1.6克,6.36毫摩尔)溶于N,N二甲基乙酰胺(16毫升),加入氰化锌(1.49克,12.72毫摩尔),甲烷磺酸(2-二环己基膦)-3,6-二甲氧基-2,4,6-三异丙基-1,1-联苯)(2-氨基-1,1-联苯-2-基)钯(II)(1.15克,1.27毫摩尔)。反应液置换三次氮气后,在氮气保护下105℃搅拌12小时。过滤并加入水(50毫升),用乙酸乙酯(40毫升×3)萃取,合并后有机相用饱和食盐水(30毫升×3)洗涤,并用无水硫酸钠干燥。过滤减压浓缩得到的粗品经硅胶柱层析(石油醚∶乙酸乙酯=50∶1到10∶1)纯化分离得到化合物16-5。MS-ESI计算值[M+H] +243,实测值243。 1H NMR(400MHz,CDCl 3)δ=8.75(d,J=5.7Hz,1H),7.42(td,J=1.3,5.7Hz,1H),7.01-6.41(m,1H),4.55(q,J=7.2Hz,2H),1.48(t,J=7.2Hz,3H).
第五步
将化合物16-5(1.1克,4.54毫摩尔)溶于甲醇(40毫升),氮气保护下加入雷尼镍(389.14毫克,454.21微摩尔,10%纯度),反应液在氢气(压力50psi)环境下加热到50℃反应12小时。反应完毕后,过滤,浓缩。粗品用石油醚∶乙酸乙酯=4∶1(25毫升)打浆,过滤,滤饼干燥得到化合物16-6。MS-ESI计算值[M+H] +201,实测值201。 1H NMR(400MHz,CDCl 3-d)δ=8.72(d,J=5.7Hz,1H),7.67-7.36(m,1H),7.25(s,1H),7.18(d,J=5.7Hz,1H),6.67(br s,1H),4.57(s,2H).
第六步
将化合物16-6(780毫克,3.90毫摩尔)溶于二氯甲烷(10毫升),加入二碳酸二叔丁酯(935.62毫克,4.29毫摩尔)和4-二甲氨基吡啶(71.42毫克,584.58微摩尔)。反应液在25℃下反应0.5小时。反应液减压浓缩得到的粗品经硅胶柱层析(石油醚∶乙酸乙酯=20∶1到5∶1)纯化分离得到化合物16-7。MS-ESI计算值[M+H] +301,实测值301。
第七步
将化合物16-7(900毫克,3.00毫摩尔)溶于四氢呋喃(10毫升),零下5℃加入二异丁基氢化铝(1摩尔/升,8.99毫升),反应液在20℃下反应2小时。加入十水硫酸钠(20克)并搅拌30分钟淬灭反应,过滤,滤液浓缩得到化合物16-8。MS-ESI计算值[M+H] +303,实测值303。
第八步
将化合物16-8(800毫克,2.65毫摩尔)溶于醋酸(8毫升),加入氰基硼氢化钠(166.32毫克,2.65毫摩尔),反应液在15℃下反应1小时。将反应液缓慢滴加到饱和碳酸氢钠水溶液(50毫升)中,搅拌20分钟后,用乙酸乙酯(40毫升×3)萃取,合并后有机相用饱和食盐水(30毫升×2)洗涤,并用无水硫酸钠干燥。过滤减压浓缩得到的粗品经硅胶柱层析(石油醚∶乙酸乙酯=20∶1到5∶1)纯化分离得到化合物16-9。MS-ESI计算值[M+H] +287,实测值287。
第九步
将化合物16-9(270毫克,943.15微摩尔)溶于二氯甲烷(3毫升),加入间氯过氧苯甲酸(406.90毫克,1.89毫摩尔,80%纯度),15℃下搅拌4小时。0℃下加入15毫升的饱和亚硫酸钠水溶液淬灭反应,并用饱和碳酸氢钠水溶液调pH到7后,二氯甲烷(30毫升×3)萃取,合并后有机相用饱和食盐水(30毫升×2)洗涤,并用无水硫酸钠干燥。过滤减压浓缩得到的粗品用石油醚∶甲基叔丁基醚=1∶1(10毫升)打浆,过滤,滤饼干燥得到化合物16-10。MS-ESI计算值[M+H] +303,实测值303。 1H NMR(400MHz,CDCl 3)δ=8.14(d,J=7.2Hz,1H),7.11(br d,J=7.2Hz,1H),6.88-6.33(m,1H),4.93-4.72(m,4H),1.53(s,9H).
第十步
将化合物16-10(75毫克,248.12微摩尔)溶于N,N二甲基甲酰胺(10毫升),0℃下缓慢滴加草酰氯(725.02毫克,5.71毫摩尔),升温到30℃下搅拌12小时。0℃下将反应液逐滴加入到50毫升的饱和碳酸氢钠水溶液中淬灭反应,乙酸乙酯(30毫升×3)萃取,合并后有机相用饱和食盐水(20毫升×2)洗涤,并用无水硫酸钠干燥。过滤减压浓缩得到的粗品经硅胶薄层色谱板(石油醚∶乙酸乙酯=3∶1)纯化得到化合物16-11。MS-ESI计算值[M+H] +321,实测值321。
第十一步
将化合物16-11(30毫克,93.54微摩尔)溶于甲醇(5毫升)和N,N二甲基甲酰胺(1毫升),加入三乙胺(28.40毫克,280.62微摩尔)和1,1-双(二苯基磷)二茂铁二氯化钯(6.84毫克,9.35微摩尔),反应液在一氧化碳(压力50psi)环境下加热到80度反应12小时。过滤浓缩,用乙酸乙酯10毫升和水20毫升稀释,然后用乙酸乙酯(30毫升×2)萃取,有机相用饱和食盐水(20毫升×2)洗涤,无水硫酸钠干燥,过滤并减压浓缩,得到的粗品经硅胶薄层色谱板(石油醚∶乙酸乙酯=1∶1)纯化得到化合物16-12。MS-ESI计算值[M+H] +345,实测值345。
第十二步
将化合物16-12(30毫克,87.13微摩尔)和中间体E(43.88毫克,78.42微摩尔)溶于四氢呋喃(2毫升),0度氮气保护下加入双三甲硅基氨基锂的四氢呋喃溶液(1摩尔/升,304.96微升),反应液在15℃下反应1小时。0℃下加入饱和氯化铵溶液(20毫升)淬灭反应液,乙酸乙酯(20毫升×3)萃取。合并的有机相用饱和食盐水(15毫升×2)洗涤,无水硫酸钠干燥,过滤并减压浓缩,得到的粗品经硅胶薄层色谱板(石油醚∶乙酸乙酯=1∶1)纯化得到化合物16-13。MS-ESI计算值[M+H] +871,实测值871。
第十三步
将化合物16-13(32毫克,36.7微摩尔)溶于二氯甲烷(2毫升),加入氯化氢的乙酸乙酯溶液(4摩尔/升,432.43微升),15℃下搅拌6小时。反应液浓缩得到化合物16-14的盐酸盐。MS-ESI计算值[M+H] +657,实测值657。
第十四步
将化合物16-14(25毫克,36.03微摩尔)溶于甲醇(2毫升),加入N,N-二异丙基乙胺(4.66毫克,36.03微摩尔)和化合物B-8(15.70毫克,90.06微摩尔),15℃下搅拌0.5小时后加入氰基硼氢化钠(6.79毫克),反应液在15℃下反应1.5小时。加入10毫升水稀释反应液,并用二氯甲烷(10毫升×3)萃取。合并的有机相用饱和食盐水(10毫升×2)洗涤,无水硫酸钠干燥,过滤并减压浓缩得到化合物16-15。MS-ESI计算值[M+H] +815,实测值815。
第十五步
将化合物16-15(29毫克,35.55微摩尔)溶于乙酸乙酯(1毫升),加入氯化氢的乙酸乙酯溶液(0.5毫升,4摩尔/升),反应液在15℃下反应0.5小时。反应液减压浓缩得到的粗品经高效液相色谱(色谱柱:Phenomenex Synergi C18 150*25毫米*10微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:8%-38%,10分钟)分离得到化合物16。MS-ESI计算值[M+H] +701,实测值701。 1H NMR(400MHz,CD 3OD)δ=8.58(dd,J=1.5,8.3Hz,1H),8.46(s,1H),7.91(s,1H),7.70(dd,J=1.6,7.7Hz,1H),7.56(t,J=7.6Hz,1H),7.52-7.06(m,4H),4.45(br s,2H),4.16(br d,J=10.9Hz,4H),4.09(s,3H),4.06(br d,J=8.5Hz,2H),3.93-3.83(m,2H),3.77(t,J=5.7Hz,2H),2.97(t,J=5.7Hz,2H),1.54(s,3H).
实施例17
Figure PCTCN2022072583-appb-000087
Figure PCTCN2022072583-appb-000088
第一步
将中间体G(400.0毫克,1.03毫摩尔)溶于四氢呋喃(2.5毫升),在0℃下向反应液中加入化合物C-5(347.68毫克,924.11微摩尔),二(三甲基硅)氨基锂(1摩尔/升的正己烷溶液,3.08毫升),在20℃下搅拌1小时。反应完毕后,向反应液中加入水(50毫升),用二氯甲烷(50毫升×3)萃取,有机相用饱和食盐水(30毫升×3)洗涤,合并有机相用无水硫酸钠干燥,过滤,减压浓缩得到粗产物。粗产物经过硅胶柱层析(石油醚∶乙酸乙酯=20∶1到1∶1)分离得到化合物17-1。MS-ESI计算值[M+H] +719,实测值719。
第二步
将化合物17-1(200.0毫克,277.88微摩尔)溶于二氯乙烷(3毫升)加入活性二氧化锰(241.59毫克,2.78微摩尔),在90℃下搅拌2小时。反应完毕后,过滤,减压浓缩得到化合物17-2。MS-ESI计算值[M+H] +717,实测值717。
第三步
将化合物17-2(70.0毫克,97.53微摩尔)溶于二氯甲烷(3毫升)向反应液中加入化合物12-1(23.94毫克,146.30微摩尔),N,N-二异丙基乙胺(37.82毫克,292.60微摩尔),在25℃下搅拌0.5小时。向反应液中加入三乙酰氧基硼氢化钠(62.01毫克,292.60微摩尔),在25℃下搅拌10小时。反应完毕后,向反应液中加入水(5.0毫升),用二氯甲烷24毫升(8毫升×3)萃取,有机相用饱和食盐水(10毫升×2)洗涤,合并有机相用无水硫酸钠干燥,过滤,减压浓缩得到粗产物。粗产物经过硅胶薄层色谱板(石油醚∶乙酸乙酯∶乙醇=4∶3∶1)分离得到化合物17-3。MS-ESI计算值[M+H] +828,实测值828。
第四步
将化合物17-3(50.0毫克,60.32微摩尔)溶于三氟乙酸(2毫升),在25℃下搅拌12小时。反应完毕后,浓缩,粗品经高效液相色谱(色谱柱:Unisil 3-100 C18 Ultra 150*50毫米*3微米;流动相:流动相A:体积分数0.225%甲酸的水溶液;流动相B:乙腈;B%:8%-38%,10分钟)分离得到化合物17。MS-ESI计算值[M+H] +714,实测值714。 1HNMR(400MHz,CD 3OD)δ=8.63(dd,J=1.5,8.3Hz,1H),8.47(s,1H),7.79-7.66(m,2H),7.58(t,J=7.6Hz,1H),7.52-7.43(m,2H),7.22-7.15(m,1H),4.32(s,2H),4.10(s,3H),4.05(s,3H),3.99(br d,J=13.6Hz,4H),3.88-3.76(m,4H),3.01-2.84(m,4H),2.80(t,J=5.9 Hz,2H),2.45-2.20(m,4H),1.29(s,3H).
实施例18
Figure PCTCN2022072583-appb-000089
第一步
将化合物17-2(60.0毫克,83.60微摩尔)溶于二氯甲烷(3毫升),向反应液中加入化合物2-1(15.50毫克,125.40微摩尔),N,N-二异丙基乙胺(32.41毫克,250.80微摩尔),在25℃下搅拌0.5小时。向反应液中加入三乙酰氧基硼氢化钠(53.15毫克,250.80微摩尔),在25℃下搅拌2小时。反应完毕后,向反应液中加入水(5毫升),用二氯甲烷(8毫升×3)萃取,有机相用饱和食盐水(10毫升×2)洗涤,合并有机相用无水硫酸钠干燥,过滤,减压浓缩得到粗产物。粗产物经过硅胶薄层色谱板(石油醚∶乙酸乙酯∶乙醇=4∶3∶1)分离得到化合物18-1。MS-ESI计算值[M+H] +788,实测值788。
第二步
将化合物18-1(21.0毫克,26.62微摩尔)溶于三氟乙酸(2毫升),在25℃下搅拌2小时。反应完毕后,浓缩,粗品经高效液相色谱(色谱柱:Unisil 3-100 C18 Ultra 150*50毫米*3微米;流动相:流动相A:体积分数0.225%甲酸的水溶液;流动相B:乙腈;B%:8%-38%,10分钟)分离得到化合物18。MS-ESI计算值[M+H] +674,实测值674。 1H NMR(400MHz,CD 3OD)δ=8.56(dd,J=1.1,8.3Hz,1H),8.51(s,1H),8.34(s,1H),7.73(dd,J=1.4,7.6Hz,1H),7.57(t,J=7.6Hz,1H),7.53-7.42(m,2H),7.19(dd,J=1.4,7.6Hz,1H),4.60-4.56(m,3H),4.44(br s,2H),4.10(s,3H),3.95(s,2H),3.79(t,J=5.6Hz,2H),3.19(br t,J=5.6Hz,4H),3.02-2.95(m,2H),2.80(t,J=5.7Hz,2H),2.34-2.19(m,1H),2.35-2.15(m,1H),2.09-1.92(m,1H)
实施例19
Figure PCTCN2022072583-appb-000090
Figure PCTCN2022072583-appb-000091
第一步
将化合物6-1(15.70毫克,209.00微摩尔)溶于二氯甲烷(3毫升),加入化合物17-2(100毫克,139.33微摩尔),三乙氧基硼氢化钠(88.59毫克,417.99微摩尔),反应液在室温下反应2小时。反应完毕后,加水(20毫升)稀释,用二氯甲烷(25毫升×2)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。残余物经硅胶薄层色谱板(二氯甲烷∶甲醇=6∶1)分离得到化合物19-1。MS-ESI计算值[M+H] +776,实测值776。
第二步
将化合物19-1(70毫克,90.11微摩尔)溶于二氯甲烷(2毫升),加入三氟乙酸(6毫升),20℃搅拌12小时,LC-MS监测有原料,将反应液升温至30℃搅拌3小时。反应完毕后,浓缩。粗品加入到四氢呋喃(2毫升)和饱和碳酸氢钠水溶液(2毫升)溶液中,在20℃搅拌3小时,加水(20毫升)稀释,用二氯甲烷(20毫升×2)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。粗品经高效液相色谱(色谱柱:Phenomenex Gemini-NX C18 75×30毫米×3微米;流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:10%-40%,7分钟)分离得到化合物19。MS-ESI计算值[M+H] +662,实测值662。 1H NMR(400MHz,CD 3OD)δ=8.58(d,J=8.31Hz,1H),8.51(s,1H),8.33-8.40(m,1H),7.74(dd,J=7.76,0.92Hz,1H),7.56-7.60(m,1H),7.45-7.54(m,2H),7.18-7.24(m,1H),4.33(s,2H),4.12-4.14(m,3H),3.96(s,2H),3.81(t,J=5.62Hz,2H),3.75(t,J=5.81Hz,2H),3.17-3.22(m,4H),2.98-3.04(m,2H),2.82(t,J=5.62Hz,2H),1.96(quin,J=6.48Hz,2H).
实施例20
Figure PCTCN2022072583-appb-000092
Figure PCTCN2022072583-appb-000093
第一步
将中间体J(400毫克,1.24毫摩尔)溶于丙酮(4毫升),加入2-碘丙烷(1.05毫克,6.20毫摩尔),碳酸钾(342.99毫克,2.48毫摩尔),反应液在80℃下反应4小时。反应完毕后,用乙酸乙酯(60毫升×2)稀释,并用水(60毫升)萃取。有机相用硫酸钠干燥,过滤,浓缩。残渣经硅胶薄层色谱板(石油醚∶乙酸乙酯=5∶1)分离得到化合物20-1。MS-ESI计算值[M+H] +365,实测值365 1H NMR(400MHz,CDCl 3)δppm=7.48-7.50(m,1H),4.71-4.84(m,3H),4.50(q,J=7.15Hz,2H),3.62-3.71(m,2H),2.78(brt,J=5.71Hz,2H),1.40-1.50(m,18H).
第二步
将化合物20-1(535毫克,1.47微摩尔)溶于二氯甲烷(6毫升),加入氯化氢的乙酸乙酯溶液(4摩尔/升,3.5毫升),反应液在室温25℃下反应1小时。反应完毕后,反应液浓缩后得到粗品化合物20-2。MS-ESI计算值[M+H] +265.2,实测值265.2。
第三步
将化合物20-2(440毫克,1.46微摩尔)溶于甲醇(24毫升),加入N,N-二异丙基乙胺(189.06毫克,1.46微摩尔)和化合物B-8(637.49毫克,3.66毫摩尔),搅拌半小时后加入氰基硼氢化钠(275.79毫克,4.39毫摩尔),反应液在25℃下反应16小时。反应完毕后,用乙酸乙酯(60毫升×2)稀释,用水(40毫升)洗涤。有机相用无水硫酸钠干燥,过滤,浓缩,经硅胶薄层色谱板(石油醚∶乙酸乙酯=5∶1)分离得到化合物20-3。MS-ESI计算值[M+H] +423.4,实测值423.4。 1H NMR(400MHz,CDCl 3)δ=7.38(s,1H),4.58-4.74(m,1H),4.39(q,J=7.05Hz,2H),3.64-3.93(m,4H),2.50-2.94(m,6H),1.32-1.37(m,4H),1.30(d,J=6.00Hz,6H),0.83(s,9H),0.00(s,6H).
第四步
将化合物20-3(120毫克,283.93微摩尔)和中间体I(123.93毫克,227.15微摩尔)溶于四氢呋喃(2毫升),零度氮气保护下加入双三甲硅基氨基锂的四氢呋喃溶液(1摩尔/升,851.80微升),反应液在零度下反应1小时。反应结束后,反应液用饱和氯化铵溶液(20毫升)淬灭,乙酸乙酯(20毫升)萃取。有机相用无水硫酸钠干燥,过滤,浓缩。粗品经硅胶薄层色谱板(二氯甲烷∶甲醇=10∶1)纯化 分离得到化合物20-4。MS-ESI计算值[M+H] +922,实测值921。
第五步
将化合物20-4(100毫克,85.85微摩尔)溶于二氯甲烷(5毫升),加入氯化氢的乙酸乙酯溶液(4摩尔/升,4毫升),反应液在室温25℃下反应0.5小时。反应完毕后,浓缩。粗品经高效液相色谱(色谱柱:Phenomenex Gemini-NX C18 75×30毫米×3微米,流动相:流动相A:体积分数0.225%甲酸水溶液;流动相B:乙腈;B%:12%-42%,7分钟)分离得到化合物20的甲酸盐。MS-ESI计算值[M+H] +693,实测值693。 1H NMR(400MHz,CD 3OD)δ=8.61(dd,J=8.25,1.41Hz,1H),8.44-8.52(m,2H),7.70-7.74(m,2H),7.56-7.61(m,1H),7.45-7.53(m,2H),7.17(dd,J=7.64,1.41Hz,1H),4.94(br s,1H),4.65-4.71(m,1H),4.55(s,2H),4.38-4.45(m,2H),4.12(s,3H),3.90-3.98(m,4H),3.84(t,J=5.75Hz,2H),3.00-3.07(m,2H),2.85-2.94(m,4H),1.45(d,J=5.99Hz,6H).
实验例1:PD-1/PD-L1均相时间分辨荧光(Homogenouse Time-Resolved Fluorescence,HTRF)结合实验实验原理:
小分子化合物可以通过和PD-L1结合,而竞争抑制PD-1与PD-L1的结合;当作为供体的PD-1分子与作为受体的PD-L1分子十分靠近时,供体分子将能量传递给受体分子,进而导致受体分子发出荧光;通过检测荧光强弱,可以测试小分子阻止PD-L1与PD-1结合的能力。采用均相时间分辨荧光(HTRF)结合试验来检测本发明的化合物抑制PD-1/PD-L1相互结合的能力。
实验材料:
PD-1/PD-L1TR-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,染料标记受体(Dye-labeled acceptor),PD-L1-生物素(PD-L1-biotin)和待测化合物。将待测化合物用排枪进行5倍稀释至第8个浓度,即从40μM稀释至0.5nM,DMSO浓度为4%,设置双复孔实验。向微孔板中加入5μM抑制剂各浓度梯度,其中最大(Max)信号孔和最小(Min)信号孔加入5μL含4%DMSO的缓冲液,5μL PD-L1-生物素(PD-L1-biotin)(60nM),最小(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稀释后的染料标记受体(Dye-labeled acceptor)。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TR-FRET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提供了本发明实施例的化合物对PD1/PD-L1结合的抑制活性。
表1本发明实施例化合物对PD-1/PD-L1结合的IC 50值测试结果
受试化合物 IC 50(nM)
化合物2 1.49
化合物8的甲酸盐 4.61
化合物12 3.13
化合物15 4.20
实验结论:本发明化合物对PD-1/PD-L1结合有显著的抑制效果。
实验例2:利用MDA-MR-231细胞检测化合物对PD-L1表达水平的影响
实验原理:
使用三阴性乳腺癌细胞系(MDA-MB-231)是评估PD-L1內吞的间接方法。细胞表面的PD-L1分子可以通过溶酶体和蛋白酶体途径进行降解,加入小分子抑制剂促使诱导PD-L1内吞。将小分子与MDA-MB-231细胞共同孵育24小时以后,利用流式细胞术(Fluorescence-activated Cell Sorting,FACS)检测细胞表面PD-L1的含量可以间接反映小分子诱导PD-L1內吞的效果。采用流式细胞术(FACS)来检测本发明的化合物对MDA-MR-231细胞PD-L1的表达水平的影响。
实验材料:
磷酸盐缓冲液(DPBS)、1640培养基、青-链霉素、胎牛血清、非必需氨基酸、β-巯基乙醇(2-ME)、人源干扰素γ、LIVE/DEAD染液、染色液(staining buffer)、固定液(Fixation buffer)、0.25%胰酶、EDTA、抗人源PD-L1(Anti-human PD-L1)、同型对照抗人源PD-L1(Anti-human PD-L1 Isotype)。
1640完全培养基配置:439.5毫升1640培养基中加入50毫升胎牛血清、5毫升非必需氨基酸、5毫升青-链霉素和0.5毫升β升巯基乙醇,混匀。
10mM的EDTA配置:取1毫升0.5M EDTA加入49毫升DPBS中,混匀。
实验步骤:
1)MDA-MB-231细胞计数及铺板:取出培养瓶,去除培养基并用DPBS冲洗一次。冲洗后培养瓶中加入3毫升0.25%胰酶置于37℃培养箱中处理1.5min。取出培养瓶加入9毫升的1640完全培养基终止反应,将细胞转移至50毫升离心管中,37℃1000rpm离心5min。根据细胞数量加入适当体积的培养液重悬细胞,并用细胞计数仪计数。用培养基将细胞浓度调整为5×10 5个/毫升。铺板:96孔板中每孔加入200μL体积的细胞悬液,使得每孔中细胞数目为1×10 5个。放置于培养箱中培养过夜。
2)药物孵育:配置100X化合物稀释液,并按5倍梯度稀释药品。在各孔细胞中分别加入2μL各100X化合物溶液。37℃培养箱孵育24小时。3)PD-L1细胞染色及FACS检测:取出培养板,弃去上层培养液。200μL 1XPBS洗一次。入100μL EDTA(终浓度为10mM)37℃处理10min。1500rpm离心5min后200μL染色液洗一次。染色:将anti-human PD-L1(2μL每孔)和LIVE/DEAD染液(1∶1000)于染色液中稀释,每孔加入50μL,4℃染色30min。200μL染色液洗两次。固定:每孔加入100μL的固定液,4℃固定15min。200μL染色液洗两次。150μL重悬细胞。FACS检测。表2提供了本发明实施例的化合物对MDA-MR-231细胞PD-L1表达水平的影响。
表2本发明实施例化合物对MDA-MR-231细胞PD-L1表达水平的影响的测试结果
受试化合物 IC 50(nM)
化合物1的甲酸盐 3.41
化合物2 3.52
化合物3 4.07
化合物8的甲酸盐 5.85
化合物12 3.82
化合物15 2.13
实验结论:本发明化合物对MDA-MR-231细胞PD-L1表达水平有显著的抑制效果。
实验例3:NFAT活性测试
实验原理:
工程化的T细胞表面表达PD-1分子以及T细胞受体(TCR),在和工程化的抗原递呈细胞(APC)共培养以后,可以激活T细胞的NFAT信号通路。在APC上表达PD-L1分子可以有效减弱T细胞内的NFAT信号通路;利用针对PD-L1的抑制剂可以有效阻断PD-1/PD-L1调节机制,从而逆转减弱的NFAT信号通路。将小分子与APC预处理以后,再和T细胞共培养,然后通过检测荧光素酶的表达量,间接反映T细胞内NFAT通路的激活程度。
实验材料:
PD-1/PD-L1 NFAT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20μM稀释至0.25nM,DMSO浓度为2%,设置双复孔实验。弃TCR Activitor/PD-L1 CHO细胞上清,每孔加入50微升化合物工作液,37℃孵育30分钟;结束孵育后每孔加入50μL密度为4X10 5/毫升的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(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3提供了本发明实施例的化合物对PD-1/PD-L1结合的抑制活性。
表3本发明实施例化合物对PD-1/PD-L1结合的抑制活性的测试结果
Figure PCTCN2022072583-appb-000094
实验结论:本发明化合物能在细胞水平抑制PD-1/PD-L1的相互作用,从而显著激活T细胞的NFAT信号通路。
实验例4:药代动力学测试
实验目的:研究化合物在在C57BL/6小鼠体内药代动力学
实验材料:C57BL/6小鼠(雄性,8周龄,体重25g-30g)
实验操作:以标准方案测试化合物静脉注射(IV)及口服(PO)给药后的啮齿类动物药代特征,实验中候选化合物配成1mg/mL澄清溶液,给予小鼠单次静脉注射及口服给药。静注及口服溶媒均为5%DMSO/5%15-羟基硬脂酸聚乙二醇酯(Solutol)/90%水溶液。该项目使用四只雄性C57BL/6小鼠,两只小鼠进行静脉注射给药,给药剂量为1mg/kg,收集给药后0.0833,0.25,0.5,1,2,4,6,8,24h的血浆样品,另外两只小鼠口服灌胃给药,给药剂量为10mg/kg,收集给药后0.25,0.5,1,2,4,6,8,24h的血浆样品。收集24小时内的全血样品,3000g离心15分钟,分离上清得血浆样品,加入含内标的乙腈溶液沉淀蛋白,充分混匀离心取上清液进样,以LC-MS/MS分析方法定量分析血药浓度,并计算药代参数,如达峰浓度(C max),清除率(CL),半衰期(T 1/2),组织分布(V dss),药时曲线下面积(AUC 0-last),生物利用度(F)等。
本发明实施例化合物在小鼠体内的药代动力学相关参数如下表所示。
表4药代动力学测试结果
Figure PCTCN2022072583-appb-000095
实验结论:本发明化合物具有良好的药代动力学性质,包括良好的口服生物利用度,口服暴露量,半衰期和清除率等。
实验例5:化合物在C57BL/6-hPDL1小鼠结直肠癌MC38-hPDL1皮下移植模型中的药效学评价研究实验目的:评价化合物在小鼠结直肠癌细胞MC38-hPDL1移植人源化小鼠C57BL/6-hPDL1中的抗肿瘤作用。
表5实验设计
Figure PCTCN2022072583-appb-000096
备注:G:组别;N:动物只数;p.o:口服给药;BID:一天两次。
表6实验动物
种属 小鼠
品系 C57BL/6-hPDL1
级别 SPF级
周龄 5.86~6.86
性别 雌性
实验方法:
1.肿瘤细胞接种
实验细胞:小鼠结肠癌细胞MC38-hPDL1复苏,复苏代次为Pn+6代,收集对数生长期的MC38-hPDL1细胞,去除培养液并用PBS洗两次后接种(荷瘤前、荷瘤后MC38-hPDL1细胞存活率分别为:97.4%及95.0%),接种量:1×10 6/100μL/只,接种位置:小鼠右前肢。
2.分组给药
接种后第7天,平均肿瘤体积达到85.23mm 3时,小鼠根据肿瘤体积随机分成5组,每组8只。分组当天定义为D0天,并于D0天开始给药。分剩小鼠进行后续补充实验。
3.药物配制
给药体积:根据小鼠体重调整(小鼠给药体积=10μL/g×小鼠体重(g))
4.实验观察和数据采集
开始给药后,于第0、2、4、6、8、11、13、15、18、20、22、25天观测肿瘤大小。瘤体积计算方式为:肿瘤体积(mm 3)=0.5×(肿瘤长径×肿瘤短径 2)。
5.实验终点
实验结束时,分析下列指标:1)肿瘤体积变化(TGItv);2)平均体重变化;TGITV(相对肿瘤抑制率)计算公式:
TGItv(%)=[1-(meanTVtn-meanTVt0)/(meanTVvn-mean TVv0)]×100%
meanTVtn:某给药组在第n天测量时平均瘤体积
meanTVt0:某给药组在第0天测量时平均瘤体积
meanTVvn:溶剂对照组在第n天测量时平均瘤体积
mean TVv0:溶剂对照组在第0天测量时平均瘤体积
实验结果:本发明化合物对C57BL/6-hPDL1小鼠结直肠癌MC38-hPDL1皮下移植模型的抑瘤药效评价(基于给药后第25天肿瘤体积计算得出)如下表7所示:
表7
Figure PCTCN2022072583-appb-000097
本发明化合物对C57BL/6-hPDL1小鼠结直肠癌MC38-hPDL1皮下移植模型中小鼠平均体重的影响如下表8所示:
表8
组别 给药前(第0天)平均体重(克) 给药第25天平均体重(克)
Vehicle(空白组) 19.2 20.0
化合物8的甲酸盐 19.0 19.9
化合物12 19.2 19.3
化合物15 19.5 20.0
化合物2 19.0 18.8
实验结论:本发明化合物对C57BL/6-hPDL1小鼠结直肠癌MC38-hPDL1皮下移植模型具有优异的抑瘤效果,给药过程中动物体重未见显著下降,耐受性较好。
实验例6:化合物在Beagle犬经口灌胃重复给药28天毒理学研究
实验目的:评价化合物在大鼠中,每天1次经口灌胃给予Beagle犬不同剂量的化合物,连续给药4周,停药恢复4周,观察其毒性反应及其可逆程度或可能的延迟毒性反应,探索其毒性反应情况,判断毒性靶器官或靶组织。
表9:实验设计
组别 剂量mg/kg 给药容量ml/kg 理论浓度mg/ml 给药频率
溶媒对照 0 0 0 每天一次
低剂量 15 5 3 每天一次
中剂量 50 5 10 每天一次
高剂量 150 5 30 每天一次
表10:实验动物
种属
品系 Beagle犬
级别 普通级
周龄 6~8月龄
性别 雌雄
实验方法:按照现行的指导原则和预实验结果来设计开展实验。
实验结论:本发明化合物在给药过程中动物体重未见显著下降,耐受性良好;与溶媒对照组相比,主要可见胃肠道反应、咳嗽、流涎以及总胆红素(TBIL)升高。靶器官为肝脏、肺脏、胸腺、脾脏、肠系膜***、颌下***、回肠。以上改变在恢复期均可见恢复或恢复趋势,本发明化合物安全性良好。

Claims (20)

  1. 式(I)化合物或其药学上可接受的盐,
    Figure PCTCN2022072583-appb-100001
    其中,
    R 1和R 2分别独立地选自H、F、Cl、Br、I、CN和C 1-3烷基,所述C 1-3烷基任选被1、2或3个卤素取代;
    R 3选自H、CN、C 1-3烷基、C 1-3烷氧基和C 1-3烷氨基,所述C 1-3烷基、C 1-3烷氧基和C 1-3烷氨基分别独立地任选被1、2或3个卤素取代;
    R 4和R 5分别独立地选自H、C 1-6烷基、C 1-6烷氨基、C 3-6环烷基和-C 1-3烷基-(3-6元杂环烷基),所述C 1- 6烷基、C 1-6烷氨基、C 3-6环烷基和-C 1-3烷基-(3-6元杂环烷基)分别独立地任选被1、2或3个R a取代;
    或者,R 4和R 5连接形成3~8元杂环烷基,所述3~8元杂环烷基任选被1、2或3个R b取代;
    R 6选自任选被1、2或3个R c取代的C 1-3烷基;
    L选自任选被1、2或3个R d取代的-C 1-6烷基-;
    X选自CH和N;
    Y选自CH和N;
    Z 1选自单键和CH 2
    Z 2选自CH和N;
    R a选自F、Cl、Br、I、OH、=O、NH 2、C 1-3烷氨基、C 1-3烷氧基和C 1-3烷基,所述C 1-3烷氨基、C 1-3烷氧基和C 1-3烷基分别独立地任选被1、2或3个R取代;
    R b选自CN、F、Cl、Br、I、OH、-C(=O)NH 2、C 1-3烷氨基、C 1-3烷氧基和C 1-3烷基;
    R c选自F、Cl、Br、I、CN、OH、=O和NH 2
    R d选自F、Cl、Br、I、CN、OH、=O和NH 2
    R选自F、Cl、Br、I、CN、OH、=O和NH 2
    所述3-6元杂环烷基和3~8元杂环烷基分别独立地包含1、2或3个分别独立地选自N、O、S和NH的杂原子或杂原子团。
  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,R 1和R 2分别独立地选自H、F、Cl、Br、I、CN、CF 3和CH 3
  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,R 3选自H、CN、CH 3、-OCH 3
    Figure PCTCN2022072583-appb-100002
    所述CH 3、-OCH 3
    Figure PCTCN2022072583-appb-100003
    分别独立地任选被1、2或3个卤素取代。
  4. 根据权利要求3所述的化合物或其药学上可接受的盐,其中,R 3选自H、CN、CH 3、-OCH 3
    Figure PCTCN2022072583-appb-100004
    Figure PCTCN2022072583-appb-100005
  5. 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 4和R 5分别独立地选自H、C 1-4烷基、C 1-3烷氨基、环丙基和
    Figure PCTCN2022072583-appb-100006
    所述C 1-4烷基、C 1-3烷氨基、环丙基和
    Figure PCTCN2022072583-appb-100007
    分别独立地任选被1、2或3个R a取代。
  6. 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 a选自F、Cl、Br、I、OH、=O、NH 2、-NHCH 3、-OCH 3、-CH 2OH和CH 3
  7. 根据权利要求5或6所述的化合物或其药学上可接受的盐,其中,R 4和R 5分别独立地选自H、
    Figure PCTCN2022072583-appb-100008
  8. 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 4和R 5连接形成吡咯烷基、8-氮杂双环[3.2.1]辛烷基、氮杂环丁烷基和2-氮杂螺[3.3]庚烷基,所述吡咯烷基、8-氮杂双环[3.2.1]辛烷基、氮杂环丁烷基和2-氮杂螺[3.3]庚烷基分别独立地任选被1、2或3个R b取代。
  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 b选自CN、OH、-C(=O)NH 2、-OCH 3和CH 3
  10. 根据权利要求8或9所述的化合物或其药学上可接受的盐,其中,R 4和R 5连接形成
    Figure PCTCN2022072583-appb-100009
    Figure PCTCN2022072583-appb-100010
  11. 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 6选自CH 3
  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,L选自
    Figure PCTCN2022072583-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 PCTCN2022072583-appb-100012
    选自
    Figure PCTCN2022072583-appb-100013
    Figure PCTCN2022072583-appb-100014
  14. 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 PCTCN2022072583-appb-100015
    选自
    Figure PCTCN2022072583-appb-100016
  15. 根据权利要求1~12任意一项所述的化合物或其药学上可接受的盐,其中,化合物选自
    Figure PCTCN2022072583-appb-100017
    其中,R 1、R 2、R 3、R 4、R 5、R 6、Z 1如权利要求1~12所定义,
    R 7和R 8选自H、F、Cl、Br、I、CN、OH、=O和NH 2
  16. 根据权利要求1~4所述的化合物或其药学上可接受的盐,其中,化合物选自
    Figure PCTCN2022072583-appb-100018
    其中,R 1、R 2、R 3、R 6、Z 1、Z 2、X和Y如权利要求1~4所定义,
    环A选自3~8元杂环烷基,
    R 9和R 10分别独立地选自H、CN、F、Cl、Br、I、OH、-C(=O)NH 2、C 1-3烷氨基、C 1-3烷氧基和C 1-3烷基。
  17. 化合物或其药学上可接受的盐,其中,化合物选自
    Figure PCTCN2022072583-appb-100019
    Figure PCTCN2022072583-appb-100020
    Figure PCTCN2022072583-appb-100021
  18. 根据权利要求17所述的化合物或其药学上可接受的盐,其中,化合物选自
    Figure PCTCN2022072583-appb-100022
  19. 根据权利要求1-18任意一项所述的化合物或其药学上可接受的盐在制备PD-1/PD-L1抑制剂中的应用。
  20. 根据权利要求19所述的应用,其中,所述PD-1/PD-L1抑制剂是抗肿瘤药物。
PCT/CN2022/072583 2021-01-18 2022-01-18 联苯衍生物及其应用 WO2022152316A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280010036.0A CN116710460A (zh) 2021-01-18 2022-01-18 联苯衍生物及其应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110066211 2021-01-18
CN202110066211.5 2021-01-18

Publications (1)

Publication Number Publication Date
WO2022152316A1 true WO2022152316A1 (zh) 2022-07-21

Family

ID=82446957

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/072583 WO2022152316A1 (zh) 2021-01-18 2022-01-18 联苯衍生物及其应用

Country Status (2)

Country Link
CN (1) CN116710460A (zh)
WO (1) WO2022152316A1 (zh)

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018119236A1 (en) * 2016-12-22 2018-06-28 Incyte Corporation Triazolo[1,5-a]pyridine derivatives as immunomodulators
WO2018119221A1 (en) * 2016-12-22 2018-06-28 Incyte Corporation Pyridine derivatives as immunomodulators
CN110267953A (zh) * 2016-12-22 2019-09-20 因赛特公司 四氢咪唑并[4,5-c]吡啶衍生物作为pd-l1内在化诱导剂
WO2019217821A1 (en) * 2018-05-11 2019-11-14 Incyte Corporation Tetrahydro-imidazo[4,5-c]pyridine derivatives as pd-l1 immunomodulators
CN111039942A (zh) * 2018-10-12 2020-04-21 上海长森药业有限公司 含氮杂环类化合物,及其制备方法、药物组合物和应用
CN111936475A (zh) * 2018-04-03 2020-11-13 贝达药业股份有限公司 免疫调节剂及其组合物和制备方法
WO2021063404A1 (zh) * 2019-09-30 2021-04-08 南京明德新药研发有限公司 作为pd-1/pd-l1小分子抑制剂的化合物及其应用
WO2021254005A1 (zh) * 2020-06-17 2021-12-23 上海和誉生物医药科技有限公司 一种免疫抑制剂、其制备方法和应用

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018119236A1 (en) * 2016-12-22 2018-06-28 Incyte Corporation Triazolo[1,5-a]pyridine derivatives as immunomodulators
WO2018119221A1 (en) * 2016-12-22 2018-06-28 Incyte Corporation Pyridine derivatives as immunomodulators
CN110267953A (zh) * 2016-12-22 2019-09-20 因赛特公司 四氢咪唑并[4,5-c]吡啶衍生物作为pd-l1内在化诱导剂
CN111936475A (zh) * 2018-04-03 2020-11-13 贝达药业股份有限公司 免疫调节剂及其组合物和制备方法
WO2019217821A1 (en) * 2018-05-11 2019-11-14 Incyte Corporation Tetrahydro-imidazo[4,5-c]pyridine derivatives as pd-l1 immunomodulators
CN111039942A (zh) * 2018-10-12 2020-04-21 上海长森药业有限公司 含氮杂环类化合物,及其制备方法、药物组合物和应用
WO2021063404A1 (zh) * 2019-09-30 2021-04-08 南京明德新药研发有限公司 作为pd-1/pd-l1小分子抑制剂的化合物及其应用
WO2021254005A1 (zh) * 2020-06-17 2021-12-23 上海和誉生物医药科技有限公司 一种免疫抑制剂、其制备方法和应用

Also Published As

Publication number Publication date
CN116710460A (zh) 2023-09-05

Similar Documents

Publication Publication Date Title
TWI719209B (zh) 嘧啶酪氨酸激酶抑制劑
JP7088906B2 (ja) Fgfr4阻害剤並びにその製造方法及び使用
JP6986032B2 (ja) Jak阻害剤としてのピロロピリミジン化合物の結晶
TWI758999B (zh) 作為erk抑制劑的噻唑并內醯胺類化合物及其應用
JP7260718B2 (ja) ジアザインドール誘導体及びそのChk1阻害剤としての使用
CA3023444A1 (en) Antimicrobials and methods of making and using same
WO2022037649A1 (en) Heterocyclic compounds as btk inhibitors
CN113825755B (zh) 作为irak4抑制剂的咪唑并吡啶类化合物
KR20210125024A (ko) Parp 억제제로서의 인돌로 헵타밀 옥심 유사체
CN113316575B (zh) 作为TGF-βR1激酶抑制剂的5-(4-吡啶氧基)吡唑类化合物
CN112839946B (zh) 作为TGF-βR1激酶抑制剂的双环吡唑类化合物
WO2022152316A1 (zh) 联苯衍生物及其应用
JP7296017B2 (ja) ベンゾスルタムを含む化合物
CN114072401B (zh) 作为irak4和btk多靶点抑制剂的噁唑类化合物
TW202330515A (zh) 聯苯衍生物及其應用
WO2021139814A1 (zh) 喹啉并咪唑类化合物及其应用
JP7237169B2 (ja) Pd-l1免疫調整剤であるフルオロビニルベンズアミド化合物
TW202233626A (zh) 三氮唑類三并環衍生物及其製備方法和應用
RU2789450C1 (ru) Фторвинилбензамидное соединение в качестве иммуномодулятора PD-L1
CN116745301A (zh) 吲哚啉类化合物
CN116120291B (zh) 吲唑类化合物及其制备方法和应用
WO2022199578A1 (zh) 氟代乙烯基联苯衍生物及其应用
TW202330534A (zh) 吡唑並環化合物及其應用
WO2022127915A1 (zh) 吡啶并嘧啶酮类化合物
TW202241893A (zh) 四氫萘啶化合物晶型、鹽型及其製備方法

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: 22739191

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202280010036.0

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: 22739191

Country of ref document: EP

Kind code of ref document: A1