WO2021213358A1 - 含硼化合物及其应用 - Google Patents

含硼化合物及其应用 Download PDF

Info

Publication number
WO2021213358A1
WO2021213358A1 PCT/CN2021/088291 CN2021088291W WO2021213358A1 WO 2021213358 A1 WO2021213358 A1 WO 2021213358A1 CN 2021088291 W CN2021088291 W CN 2021088291W WO 2021213358 A1 WO2021213358 A1 WO 2021213358A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutically acceptable
phenyl
methyl
amino
compound
Prior art date
Application number
PCT/CN2021/088291
Other languages
English (en)
French (fr)
Inventor
古鹏
刘磊
张国宝
周峰
唐任宏
任晋生
Original Assignee
江苏先声药业有限公司
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 江苏先声药业有限公司 filed Critical 江苏先声药业有限公司
Priority to CN202180027836.9A priority Critical patent/CN115667275B/zh
Publication of WO2021213358A1 publication Critical patent/WO2021213358A1/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
    • 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
    • A61K31/437Heterocyclic 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 the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/4375Heterocyclic 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 the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • 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
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • 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
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • 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
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic System
    • C07F5/02Boron compounds
    • C07F5/04Esters of boric acids

Definitions

  • the present invention relates to a novel boron-containing compound or a pharmaceutically acceptable salt thereof, a pharmaceutical composition containing them, and the use of a selective estrogen receptor degrading agent (SERD) in the prevention or treatment of related diseases.
  • SELD selective estrogen receptor degrading agent
  • Estrogen (E2) and estrogen alpha receptor (ER ⁇ ) are important driving factors for the development of breast cancer. More than two-thirds of breast cancer patients express ER transcription factors, and in most ER-positive patients, ER is still a key driving factor even in tumors that progress after early endocrine therapy. Therefore, ER is A major target for breast cancer treatment (Pharmacology&Therapeutics 186(2018)1-24).
  • the purpose of endocrine therapy is to reduce ER activity.
  • SERMs selective estrogen receptor modulators
  • tamoxifen which is an allosteric modulator of ER, which inhibits its transcriptional activity after binding to ER.
  • AIs Aromatase inhibitors
  • fulvestrant selective estrogen receptor down-regulators
  • the antagonist inhibits its activity and also has the effect of inducing ER protein degradation.
  • endocrine therapy is the first choice for patients with estrogen receptor-positive breast cancer, about 30% of patients will relapse after treatment, and almost all patients with metastatic breast cancer will develop resistance and progress.
  • One is focused on the estrogen receptor signaling pathway itself, including the activation mutation, amplification, and fusion with other genes of the gene encoding the estrogen receptor, estrogen receptor.
  • Co-regulators and downstream control of cell cycle factors, etc.; another type of mechanism includes the activation of signal pathways that cross-react with the estrogen receptor signaling pathway, such as the growth factor receptor pathway (Oncol Ther, 2017, 5:17) –29).
  • Fulvestrant is the first and only SERD drug clinically approved for the treatment of postmenopausal patients with ER-positive, metastatic breast cancer after the progression of tamoxifen or aromatase inhibitors.
  • AstraZeneca see patent application WO2018077630A1
  • Genentech see patent application WO2019245974A1 also disclosed a series of SERD compounds with novel structures and corresponding medical applications.
  • fulvestrant has not fully achieved ER degradation, but it may also be the dose of fulvestrant (the highest can only reach 500mg, mainly due to its pharmacodynamic characteristics and muscle
  • the internal route of administration limits the maximum dose that can be given to patients) and limits its efficacy. Therefore, it has higher bioavailability, higher ER antagonistic activity, and a greater degree of ER degradation, and can be used for premenopausal patients with higher estrogen levels, and is convenient for oral administration. Drugs are urgently needed clinically.
  • the present invention provides a compound represented by formula (I) or a pharmaceutically acceptable salt thereof:
  • R 1 and R 2 are independently selected from OH, or R 1 , R 2 and the boron atom to which they are connected together form a 5-6 membered heterocyclic group, and the 5-6 membered heterocyclic group is optionally substituted by C 1 -C 10 Alkyl substitution;
  • X 1 , X 2 , X 3 , and X 4 are independently selected from CR 7 or N;
  • R 7 is selected from H, F, Cl, Br, I, OH, CN, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, 3-10 membered heterocyclic group, C 1 -C 10 alkoxy Group, C 3 -C 10 cycloalkyloxy or 3-10 membered heterocyclyloxy;
  • X and Y are independently selected from O or NH;
  • R 3 is selected from H, F, Cl, Br, I, OH, CN, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, 3-10 membered heterocyclic group, C 1 -C 10 alkoxy Group, C 3 -C 10 cycloalkyloxy or 3-10 membered heterocyclyloxy;
  • R 4 is selected from H, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl or 3-10 membered heterocyclic group;
  • R 3 , R 4 and the atoms to which they are connected together form a 4-10 membered heterocyclic group
  • the 4-10 membered heterocyclic group is optionally substituted by R a1
  • the R a1 is selected from F, Cl, Br, I, OH, CN, C 1 -C 10 alkyl or C 1 -C 10 alkoxy;
  • R 5 and R 6 are independently selected from C 1 -C 6 alkyl group or phenyl group, and the C 1 -C 6 alkyl group or phenyl group is optionally substituted by R a2;
  • R 8 is selected from H or C 1 -C 6 alkyl, the C 1 -C 6 alkyl is optionally substituted by R a2;
  • R a2 is selected from F, Cl, Br, I, OH, CN or the following groups optionally substituted by R b : C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl or 3-10 membered heterocycle base;
  • R b is selected from F, Cl, Br, I, OH, CN, C 1 -C 10 alkyl or C 1 -C 10 alkoxy;
  • R 1 and R 2 are independently selected from OH, or R 1 , R 2 and the boron atom to which they are connected together form a 5-6 membered heterocyclic group, and the 5-6 membered heterocyclic group is optionally substituted by C 1 -C 10 Alkyl substitution;
  • X 1 , X 2 , X 3 , and X 4 are independently selected from CR 7 or N;
  • R 7 is selected from H, F, Cl, Br, I, OH, CN, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, 3-10 membered heterocyclic group, C 1 -C 10 alkoxy Group, C 3 -C 10 cycloalkyloxy or 3-10 membered heterocyclyloxy;
  • X and Y are independently selected from O or NH;
  • R 3 is selected from H, F, Cl, Br, I, OH, CN, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, 3-10 membered heterocyclic group, C 1 -C 10 alkoxy Group, C 3 -C 10 cycloalkyloxy or 3-10 membered heterocyclyloxy;
  • R 4 is selected from H, C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl or 3-10 membered heterocyclic group;
  • R 3 , R 4 and the atoms to which they are connected together form a 4-10 membered heterocyclic group
  • the 4-10 membered heterocyclic group is optionally substituted by R a1
  • the R a1 is selected from F, Cl, Br, I, OH, CN, C 1 -C 10 alkyl or C 1 -C 10 alkoxy;
  • R 5 and R 6 are independently selected from C 1 -C 6 alkyl group or phenyl group, and the C 1 -C 6 alkyl group or phenyl group is optionally substituted by R a2;
  • R a2 is selected from F, Cl, Br, I, OH, CN or the following groups optionally substituted by R b : C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl or 3-10 membered heterocycle base;
  • R b is selected from F, Cl, Br, I, OH, CN, C 1 -C 10 alkyl or C 1 -C 10 alkoxy;
  • R 1 and R 2 are independently selected from OH, or R 1 , R 2 and the boron atom to which they are attached together form a 5-6 membered heterocyclic group, and the 5-6 membered heterocyclic group optionally Replaced by methyl.
  • R 1 and R 2 are independently selected from OH, or R 1 , R 2 and the boron atom to which they are attached together form a 5-6 membered heterocycloalkyl group, the 5-6 membered heterocycloalkyl group Substitution by 1, 2, 3 or 4 methyl groups, in addition to boron atoms, the heteroatoms in the 5-6 membered heterocycloalkyl group also contain 2-3 oxygen atoms.
  • R 1 and R 2 are independently selected from OH, or the 5-6 membered heterocycloalkyl formed by R 1 , R 2 and the boron atom to which they are connected is selected from
  • R 1 and R 2 are independently selected from OH.
  • X 1 , X 2 , X 3 , X 4 are independently selected from CR 7 or N, and at least 2 of X 1 , X 2 , X 3 , and X 4 groups are selected from CR 7 .
  • X 1 , X 2 , X 3 , X 4 are independently selected from CR 7 or N, and at least 3 of X 1 , X 2 , X 3 , and X 4 groups are selected from CR 7 .
  • the R 7 is selected from H, F, Cl, Br, I, CN, C 1 -C 3 alkyl or C 1 -C 3 alkoxy.
  • the R 7 is selected from H, F, Cl, Br, I, CN, or C 1 -C 3 alkoxy.
  • the R 7 is selected from H, F or methoxy.
  • the R 7 is selected from H or F.
  • R 3 is selected from H, F, Cl, Br, I, OH, CN, C 1 -C 6 alkyl, or C 1 -C 6 alkoxy.
  • R 4 is selected from H or C 1 -C 6 alkyl.
  • both R 3 and R 4 are selected from H.
  • the 4-7 membered heterocyclic group is selected from a 4-6 membered monoheterocyclic group or a 6-7 membered heterocyclic group.
  • the 4-6 membered monoheterocyclic group or the 6-7 membered spiroheterocyclic group is optionally substituted by R a1.
  • R 3 , R 4 and the atoms to which they are attached together form a 4-membered heterocyclic group, which is optionally substituted by Ra1.
  • the Ra1 is selected from F, Cl, Br, I, CN, C 1 -C 6 alkyl or C 1 -C 6 alkoxy.
  • the Ra1 is selected from F, Cl, Br, I, CN, or C 1 -C 3 alkyl.
  • the Ra1 is selected from F, Cl, Br, I, CN, or methyl.
  • the Ra1 is selected from F or methyl.
  • R 3 , R 4 and the connected atoms together form
  • R 3 , R 4 and the connected atoms together form
  • R 5 and R 6 are independently selected from a C 1 -C 6 alkyl group or a phenyl group, and the C 1 -C 6 alkyl group or phenyl group is optionally substituted by Ra2.
  • R 5 and R 6 are independently selected from a C 1 -C 3 alkyl group or a phenyl group, and the C 1 -C 3 alkyl group is optionally substituted by Ra2.
  • the R a2 is selected from F, Cl, Br, I, OH, CN , or R b is optionally substituted C 3 -C 6 cycloalkyl.
  • the Ra2 is selected from F, Cl, Br, I, OH, CN, or cyclopropyl optionally substituted with R b.
  • the R a2 is selected from F, OH, CN, or cyclopropyl optionally substituted with R b.
  • the R b is selected from F, Cl, Br, I, OH, CN, or C 1 -C 6 alkyl.
  • the R b is selected from F, Cl, Br, I, or CN.
  • the R b is selected from F.
  • the Ra2 is selected from F, Cl, OH, or cyclopropyl optionally substituted with F.
  • R 5 and R 6 are independently selected from CH 2 CF 3 , CH 2 CHF 2 , CH 2 CF 2 CH 2 OH, CH 2 CF 2 CH 2 CN, Phenyl, CH 2 C(CH 3 ) 2 F, p-fluorophenyl, p-chlorophenyl,
  • R 5 and R 6 are independently selected from CH 2 CF 3 , CH 2 CHF 2 , CH 2 CF 2 CH 2 OH, CH 2 CF 2 CH 2 CN, Or phenyl.
  • R 5 and R 6 are independently selected from CH 2 CF 3 , CH 2 CF 2 CH 2 OH, Phenyl, CH 2 C(CH 3 ) 2 F, p-fluorophenyl, p-chlorophenyl,
  • R 6 is selected from CH 2 CF 3 .
  • R 8 is selected from H or C 1 -C 3 alkyl.
  • R 8 is selected from H or methyl.
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof is selected from the compound represented by formula (II) or a pharmaceutically acceptable salt thereof:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , X 1 , X 2 , X 3 , X 4 , X, and Y are as defined above.
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof is selected from the compound represented by formula (II) or a pharmaceutically acceptable salt thereof:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 8 , X 1 , X 2 , X 3 , X 4 , X, and Y are as defined above.
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof is selected from the following compounds or a pharmaceutically acceptable salt thereof:
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof is selected from the following compounds or a pharmaceutically acceptable salt thereof:
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof is selected from the following compounds or a pharmaceutically acceptable salt thereof:
  • the present invention also provides a pharmaceutical composition, which comprises a compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable carrier and/or excipient.
  • the present invention also provides a pharmaceutical composition, which comprises a compound represented by formula (I) or a pharmaceutically acceptable salt thereof and pharmaceutically acceptable auxiliary materials.
  • the present invention relates to the use of the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof in the preparation of a medicine for preventing or treating estrogen receptor-related diseases.
  • the present invention relates to the use of the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof in the prevention or treatment of estrogen receptor related diseases.
  • the present invention relates to the use of a compound represented by formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof as a selective estrogen receptor degrading agent (SERD) in the prevention or treatment of related diseases.
  • SELD selective estrogen receptor degrading agent
  • the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof for preventing or treating estrogen receptor-related diseases.
  • the present invention also relates to a method for treating estrogen receptor-related diseases, which comprises administering to a patient a therapeutically effective dose of a pharmaceutical preparation containing the compound of formula (I) of the present invention or a pharmaceutically acceptable salt thereof.
  • the estrogen receptor-related diseases include but are not limited to tumors (such as breast cancer).
  • the definitions of groups and terms described in the specification and claims of the present invention include definitions as examples, exemplary definitions, preferred definitions, definitions recorded in tables, and definitions of specific compounds in the examples. And so on, can be combined and combined with each other arbitrarily. Such combination and the group definition and compound structure after the combination should fall within the scope of the description of the present invention.
  • a specific term should not be considered uncertain or unclear without a special definition, but should be understood according to the ordinary meaning in the field. When a trade name appears in this article, it is meant to refer to its corresponding commodity or its active ingredient.
  • pharmaceutically acceptable salts refers to pharmaceutically acceptable salts of non-toxic acids or bases, including salts of inorganic acids and bases, and organic acids and bases.
  • stereoisomer refers to the isomers produced by the different arrangements of atoms in the molecule in space, including cis and trans isomers, enantiomers, diastereomers and conformational isomers.
  • the compound of the present invention may have an asymmetric atom such as a carbon atom, a sulfur atom, a nitrogen atom, a phosphorus atom, or an asymmetric double bond. Racemates, enantiomers, diastereomers, geometric isomers and individual isomers are all included in the scope of the present invention.
  • 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 Conformers, (D)-isomers, (L)-isomers, and their racemic mixtures and other mixtures, such as enantiomers or diastereomer-enriched mixtures, all of these mixtures belong to Within the scope of the present invention.
  • Additional asymmetric carbon atoms, asymmetric sulfur atoms, asymmetric nitrogen atoms, or asymmetric phosphorus atoms may be present in substituents such as alkyl groups.
  • the asymmetric atom-containing compound of the present application can be isolated in an optically pure form or a racemic form.
  • the optically active pure form can be resolved from the racemic mixture or synthesized by using chiral raw materials or chiral reagents.
  • stereoisomers include, but are not limited to:
  • tautomer refers to an isomer of a functional group resulting from the rapid movement of an atom in a molecule at two positions.
  • the compounds of the present invention may exhibit tautomerism.
  • Tautomeric compounds can exist in two or more mutually convertible species.
  • Proton shift tautomers result from the migration of covalently bonded hydrogen atoms between two atoms.
  • Tautomers generally exist in an equilibrium form, and an attempt to separate a single tautomer usually produces a mixture whose physical and chemical properties are consistent with a mixture of compounds. The position of equilibrium depends on the chemical properties of the molecule.
  • the ketone type is dominant; in phenol, the enol type is dominant.
  • the present invention encompasses all tautomeric forms of the compound.
  • pharmaceutical composition means a mixture of one or more of the compounds described in the text or their physiologically/pharmaceutically acceptable salts or prodrugs and other chemical components, such as physiologically/pharmaceutically acceptable carriers And excipients.
  • the purpose of the pharmaceutical composition is to facilitate the administration of the compound to the organism.
  • substituted means that any one or more hydrogen atoms on a specific atom are replaced by a substituent, as long as the valence of the specific atom is normal and the substituted compound is stable.
  • it means that two hydrogen atoms are replaced, and the oxo will not occur on the aromatic group.
  • the term “optional” or “optionally” means that the event or situation described later can occur or not occur, and the description includes occurrence of said event or situation and non-occurrence of said event or situation.
  • the ethyl group is "optionally" substituted by halogen, meaning that the ethyl group can be unsubstituted (CH 2 CH 3 ), monosubstituted (such as CH 2 CH 2 F), or polysubstituted (such as CHFCH 2 F, CH 2 CHF 2 etc.) or completely substituted (CF 2 CF 3 ).
  • CH 2 CH 3 unsubstituted
  • monosubstituted such as CH 2 CH 2 F
  • polysubstituted such as CHFCH 2 F, CH 2 CHF 2 etc.
  • CF 2 CF 3 completely substituted
  • halo or halogen refers to fluorine, chlorine, bromine and iodine.
  • C m -C n in this context means having an integer number of carbon atoms in the range of mn.
  • C 1 -C 10 means that the group can have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, 6 carbon atoms, 7 carbon atoms, 8 carbon atoms, 9 carbon atoms, or 10 carbon atoms.
  • alkyl refers to a hydrocarbon group of the general formula C n H 2n+1.
  • the alkyl group may be linear or branched.
  • C 1 -C 10 alkyl should be understood to mean a linear or branched saturated monovalent hydrocarbon group having 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms.
  • the alkyl group is, for example, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, 2-methylbutyl, 1-methylbutyl, 1-ethylpropyl, 1,2-dimethylpropyl, neopentyl, 1,1-dimethylpropyl, 4-methylpentyl, 3-methylpentyl Group, 2-methylpentyl, 1-methylpentyl, 2-ethylbutyl, 1-ethylbutyl, 3,3-dimethylbutyl, 2,2-dimethylbutyl, 1,1-dimethylbutyl, 2,3-dimethylbutyl, 1,3-dimethylbutyl or 1,2-dimethylbutyl, etc.; preferably, "C 1 -C 10 "Alkyl" may include "C 1 -C 6 alkyl
  • alkoxy refers to -O-alkyl, for example the term “C 1 -C 6 alkoxy” can be understood as “C 1 -C 6 alkyloxy” or “C 1 -C 6 alkyl-O ", preferably, "C 1 -C 6 alkoxy” may include "C 1 -C 3 alkoxy”.
  • cycloalkyl refers to a carbocyclic ring that is fully saturated and may exist as a monocyclic ring, a fused ring, a bridged ring, or a spiro ring. Unless otherwise indicated, the carbocyclic ring is usually a 3 to 10 membered ring.
  • C 3 -C 10 cycloalkyl should be understood to mean a saturated monovalent monocyclic, fused, spiro or bridged ring, which has 3 to 10 carbon atoms.
  • Non-limiting examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, norbornyl (bicyclo[2.2. 1]heptyl), bicyclo[2.2.2]octyl, adamantyl, spiro[4.5]decane and the like.
  • Spirocycloalkyl refers to a cycloalkyl that exists as a spiro ring.
  • C 3 -C 10 cycloalkyl may include “C 3 -C 6 cycloalkyl”, and “C 3 -C 6 cycloalkyl” should be understood to mean a saturated monovalent monocyclic or bicyclic hydrocarbon ring, which It has 3 to 6 carbon atoms, and specific examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like.
  • cycloalkyloxy can be understood as “cycloalkyl-O-”.
  • 3-10 membered heterocyclic group means a heterocyclic group having 3, 4, 5, 6, 7, 8, 9 or 10 ring atoms, and the ring atoms contain 1-5 independently selected from the above
  • "3-10 membered heterocyclic group” may include “4-10 membered heterocyclic group", “4-7 membered heterocyclic group”, 5-6 membered heterocyclic group Group” and the like, wherein non-limiting examples of 4-membered heterocyclic groups include but are not limited to azetidinyl, oxetanyl; examples of 5-membered heterocyclic groups include, but are not limited to, tetrahydrofuranyl, dihydrofuran Oxoleyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl, 4,5-dihydrooxazole or 2,5-dihydro-1H-pyrrolyl; 6-membere
  • the heterocyclic group may also be a bicyclic group, wherein examples of the 5,5-membered bicyclic group include but Not limited to hexahydrocyclopenta[c]pyrrole-2(1H)-based ring, examples of 5, 6-membered bicyclic groups include but are not limited to hexahydropyrrolo[1,2-a]pyrazine-2(1H)- Ring, 5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazinyl ring or 5,6,7,8-tetrahydroimidazo[1 ,5-a]pyrazine.
  • the heterocyclic group may be a benzo-fused ring group of the aforementioned 4-7 membered heterocyclic group, and examples include, but are not limited to, dihydroisoquinolinyl, etc.
  • the heterocyclic groups are still non-aromatic as a whole.
  • heterocyclyloxy can be understood as “heterocyclyl-O-”.
  • treatment means administering the compound or formulation described in this application to prevent, ameliorate or eliminate a disease or one or more symptoms associated with the disease, and includes:
  • terapéuticaally effective amount means (i) treatment or prevention of a particular disease, condition or disorder, (ii) reduction, amelioration or elimination of one or more symptoms of a particular disease, condition or disorder, or (iii) prevention or delay
  • the amount of the compound of the present invention that constitutes a “therapeutically effective amount” varies depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but it can be routinely determined by those skilled in the art. Determined by its own knowledge and this disclosure.
  • excipients refers to pharmaceutically acceptable inert ingredients.
  • examples of types of the term “excipient” include, without limitation, binders, disintegrants, lubricants, glidants, stabilizers, fillers, diluents, and the like. Excipients can enhance the handling characteristics of the pharmaceutical preparation, that is, make the preparation more suitable for direct compression by increasing fluidity and/or adhesion.
  • examples of typical "pharmaceutically acceptable carriers” suitable for the above formulations are: sugars, starches, cellulose and its derivatives and other auxiliary materials commonly used in pharmaceutical formulations.
  • pharmaceutically acceptable excipients refers to those excipients that have no obvious stimulating effect on the organism and will not damage the biological activity and performance of the active compound.
  • Suitable auxiliary materials are well known to those skilled in the art, such as carbohydrates, waxes, water-soluble and/or water-swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like.
  • the compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, the embodiments formed by combining them with other chemical synthesis methods, and those well known to those skilled in the art Equivalent alternatives, preferred implementations include but are not limited to the embodiments of the present invention.
  • the present application also includes compounds of the present application that are the same as those described herein, but have one or more atoms replaced by an isotope-labeled atom having an atomic weight or mass number different from the atomic weight or mass number commonly found in nature.
  • isotopes that can be bound to the compounds of the present application include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 123 I, 125 I and 36 Cl, etc.
  • isotope-labeled compounds of the application can be used in compound and/or substrate tissue distribution analysis. Tritiated (ie 3 H) and carbon-14 (ie 14 C) isotopes are especially preferred due to their ease of preparation and detectability. Positron emission isotopes such as 15 O, 13 N, 11 C, and 18 F can be used in positron emission tomography (PET) studies to determine substrate occupancy.
  • PET positron emission tomography
  • the isotopically-labeled compounds of the present application can be prepared by the following procedures similar to those disclosed in the schemes and/or examples below, by replacing non-isotopically-labeled reagents with isotope-labeled reagents.
  • substitution with heavier isotopes can provide certain therapeutic advantages resulting from higher metabolic stability (for example, increased in vivo half-life or reduced dosage requirements), and therefore in certain situations
  • deuterium substitution can be partial or complete, and partial deuterium substitution refers to the substitution of at least one hydrogen by at least one deuterium.
  • the pharmaceutical composition of the present application can be prepared by combining the compound of the present application with suitable pharmaceutically acceptable excipients, for example, can be formulated into solid, semi-solid, liquid or gaseous preparations, such as tablets, pills, capsules, and powders. , Granules, ointments, emulsions, suspensions, suppositories, injections, inhalants, gels, microspheres and aerosols.
  • Typical routes for administering the compound of the present application or a pharmaceutically acceptable salt or pharmaceutical composition thereof include, but are not limited to, oral, rectal, topical, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, Intramuscular, subcutaneous, and intravenous administration.
  • the pharmaceutical composition of the present application can be manufactured by methods well known in the art, such as conventional mixing method, dissolution method, granulation method, sugar-coated pill method, grinding method, emulsification method, freeze-drying method, etc.
  • the pharmaceutical composition is in an oral form.
  • the pharmaceutical composition can be formulated by mixing the active compound with pharmaceutically acceptable excipients well known in the art. These auxiliary materials enable the compound of the present application to be formulated into tablets, pills, lozenges, sugar-coated agents, capsules, liquids, gels, slurries, suspensions, etc., for oral administration to patients.
  • the solid oral composition can be prepared by conventional mixing, filling or tableting methods. For example, it can be obtained by the following method: mixing the active compound with solid excipients, optionally grinding the resulting mixture, adding other suitable excipients if necessary, and then processing the mixture into granules to obtain tablets Or the core of the dragee.
  • suitable excipients include but are not limited to: binders, diluents, disintegrants, lubricants, glidants, sweeteners or flavoring agents, and the like.
  • the pharmaceutical composition may also be suitable for parenteral administration, such as a sterile solution, suspension or lyophilized product in a suitable unit dosage form.
  • the daily dose is 0.01 to 100 mg/kg body weight, preferably 0.05 to 50 mg/kg body weight, more preferably 0.1 to 30 mg/kg body weight, in single or divided doses form.
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) and/or mass spectrometry (MS).
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • the unit of NMR shift is 10 -6 (ppm).
  • the solvents measured by NMR are deuterated dimethyl sulfoxide, deuterated chloroform, deuterated methanol, etc., and the internal standard is tetramethylsilane (TMS);
  • TMS tetramethylsilane
  • IC 50 refers to the half inhibitory concentration, which refers to the half of the maximum inhibitory effect concentration.
  • Step 1 Synthesis of (R)-tert-butyl(1-(3-(benzyloxy)phenyl)propan-2-yl)carbamate
  • Step 3 Synthesis of (R)-1-(3-(benzyloxy)phenyl)-N-(2,2,2-trifluoroethyl)propane-2-amine
  • Step 6 (1S,3R)-1-(5-((1-(3-fluoropropyl)azetidine-3-yl)amino)pyridin-2-yl)-3-methyl-2-( Synthesis of 2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinoline-6-phenol
  • Step 7 (1S,3R)-1-(5-((1-(3-fluoropropyl)azetidine-3-yl)amino)pyridin-2-yl)-3-methyl-2-( Synthesis of 2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinolin-6-yl trifluoromethanesulfonate
  • Step 8 ((1S,3R)-1-(5-((1-(3-fluoropropyl)azetidin-3-yl)amino)pyridin-2-yl)-3-methyl-2- Synthesis of (2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • reaction solution was stirred and reacted at 80°C for 2 hours under the protection of nitrogen.
  • the reaction solution was cooled to room temperature, poured into water (10 mL) and stirred for 10 minutes, extracted with ethyl acetate (10 mL) twice, the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated to dryness under reduced pressure.
  • Step 1 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3- Synthesis of methyl-2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinoline-6-phenol
  • Step 2 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3- Synthesis of methyl-2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinolin-6-yl trifluoromethanesulfonate
  • Step 5 ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3 -Synthesis of methyl-2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid.
  • reaction solution was stirred and reacted at 80°C for 2 hours. After the reaction is detected by LCMS, the reaction solution is cooled to room temperature, concentrated under reduced pressure, and then subjected to preparative liquid chromatography (Phenomenex Synergi C18 column, 3um silica, 30mm diameter, 75mm length); (using water (containing 0.225% formic acid) The diminishing polarity mixture of He and acetonitrile is used as the eluent), and purified to obtain ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)nitrogen heterocycle) Butane-3-yl)amino)phenyl)-3-methyl-2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinolin-6-yl ) Boric acid (11.57 mg).
  • Step 1 Synthesis of tert-butyl (1-(3-fluoropropyl)azetidin-3-yl)carbamate.
  • Step 2 Synthesis of 1-(3-fluoropropyl)azetidine-3-amine hydrochloride.
  • Step 4 Synthesis of N-(4-(diethoxymethyl)-3,5-difluorophenyl)-1-(3-fluoropropyl)azetidine-3-amine
  • Step 5 Synthesis of 2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)benzaldehyde
  • reaction solution was poured into water (200 mL), extracted three times with ethyl acetate (100 mL), the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated to dryness under reduced pressure.
  • Step 7 Synthesis of (R)-1-fluoro-N-(1-(3-hydroxyphenyl)propan-2-yl)cyclopropanamide
  • Step 8 Synthesis of (R)-3-(2-(((1-fluorocyclopropyl)methyl)amino)propyl)phenol
  • Step 9 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2- Synthesis of (((1-fluorocyclopropyl)methyl)-3-methyl1,2,3,4-tetrahydroisoquinoline-6-phenol
  • the pH was adjusted to 8-9 with saturated aqueous sodium carbonate, and then ethyl acetate (100 mL) was used. Extract three times. The combined organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated to dryness under reduced pressure.
  • Step 10 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2- Synthesis of ((1-fluorocyclopropyl)methyl)-3-1,2,3,4-tetrahydroisoquinoline-6-methyltrifluoromethanesulfonate
  • Step 11 ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2 Synthesis of -(((1-fluorocyclopropyl)methyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • Example 4 ((1S,3R)-2-(2,2-difluoro-3-hydroxypropyl)-1-(2,6-difluoro-4-((1-(3-fluoropropyl) )Azetidine-3-yl)amino)phenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • 2,2-Difluoropropane-1,3-diol (7.7g, 68.70mmol) was dissolved in tetrahydrofuran (100mL), sodium hydrogen (2.75g, 68.70mmol) was added at 0°C, and the reaction solution was heated at 25°C. The reaction was stirred for 1 hour.
  • Step 4 (R)-N-(1-(3-(Benzyloxy)phenyl)propan-2-yl)-3-((tert-butyldiphenylsilyl)oxy)-2 Synthesis of ,2-Difluoropropane-1-amine
  • Step 5 Synthesis of (R)-3-(2-((3-((tert-butyldiphenylsilyl)oxy)-2,2-difluoropropyl)amino)propyl)phenol
  • Step 6 (1S,3R)-2-(3-((tert-butyldiphenylsilyl)oxy)-2,2-difluoropropyl)-1-(2,6-difluoro -4-((1-(3-Fluoropropyl)azetidin-3-yl)amino)phenyl)-3-methyl-1,2,3,4-tetrahydroisoquinoline-6 -Phenol synthesis
  • Step 7 (1S,3R)-2-(3-((tert-butyldiphenylsilyl)oxy)-2,2-difluoropropyl)-1-(2,6-difluoro -4-((1-(3-Fluoropropyl)azetidin-3-yl)amino)phenyl)-3-methyl-1,2,3,4-tetrahydroisoquinoline-6 Synthesis of 3-Trifluoromethanesulfonate
  • Step 8 ((1S,3R)-2-(3-((tert-butyldiphenylsilyl)oxy)-2,2-difluoropropyl)-1-(2,6-di Fluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3-methyl-1,2,3,4-tetrahydroisoquinoline- Synthesis of 6-yl)boronic acid
  • Step 9 ((1S,3R)-2-(2,2-difluoro-3-hydroxypropyl)-1-(2,6-difluoro-4-((1-(3-fluoropropyl) Synthesis of azetidine-3-yl)amino)phenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • reaction solution was poured into water and stirred for 10 minutes, extracted with ethyl acetate (5 mL) twice, the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated to dryness under reduced pressure.
  • reaction solution was washed successively with water (5mL), 1mol/L aqueous hydrochloric acid (5mL) and saturated sodium bicarbonate solution (5mL).
  • the collected organic phase was dried with sodium sulfate and filtered.
  • the organic phase Concentrate to dryness under reduced pressure to obtain compound 2-fluoro-2-methylpropyl trifluoromethanesulfonate (3.60 g).
  • Step 2 Synthesis of (R)-N-(1-(3-(benzyloxy)phenyl)propan-2-yl)-2-fluoro-2-methylprop-1-amine
  • reaction solution was cooled to room temperature, a saturated sodium carbonate solution was added dropwise to the reaction solution to adjust its pH to weakly alkaline.
  • reaction solution was diluted with ethyl acetate (10 mL), it was washed three times with an aqueous solution (30 mL), and the aqueous phase and the organic phase were collected. After the aqueous phase was extracted three times with ethyl acetate (20 mL), all the organic phases were combined, dried over sodium sulfate, filtered, and the organic phase was concentrated to dryness under reduced pressure.
  • Step 5 (1S,3R)-2-(2-fluoro-2-methylpropyl)-1-(5-((1-(3-fluoropropyl)azetidin-3-yl) Synthesis of (amino)pyridin-2-yl)-3-methyl-1,2,3,4-tetrahydroisoquinoline-6-phenol
  • Step 6 (1S,3R)-2-(2-fluoro-2-methylpropyl)-1-(5-((1-(3-fluoropropyl)azetidin-3-yl) Synthesis of (amino)pyridin-2-yl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl trifluoromethanesulfonate
  • Step 7 ((1S,3R)-2-(2-fluoro-2-methylpropyl)-1-(5-((1-(3-fluoropropyl)azetidin-3-yl )Amino)pyridin-2-yl)-3-methyl 1,2,3,4-tetrahydroisoquinolin-6-yl) boronic acid
  • reaction solution was cooled to room temperature, the reaction solution was diluted with water (5 mL), and extracted with ethyl acetate (15 mL) three times. All the organic phases were combined and dried over sodium sulfate. The organic phase was depressurized Concentrate to dryness.
  • Step 3 Synthesis of 1-(5-bromobenzofuran-3-yl)-N-(2,2,2-trifluoroethyl)propane-2-amine
  • Step 4 1-(5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-yl)-N-(2 ,2,2-Trifluoroethyl)propane-2-amine synthesis
  • Step 5 Synthesis of 3-(2-(((2,2,2-trifluoroethyl)amino)propyl)benzofuran-5-ol
  • reaction solution was extracted three times with ethyl acetate (15 mL).
  • Step 6 1-(4-Bromo-2,6-difluorophenyl)-3-methyl-2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydro Synthesis of Benzofuran[2,3-c]pyridine-6-ol
  • Step 7 1-(2,6-Difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3-methyl-2-( Synthesis of 2,2,2-trifluoroethyl)-1,2,3,4-tetrahydrobenzofuran[2,3-c]pyridine-6-ol
  • Step 8 1-(2,6-Difluoro-4-(((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3-methyl-2- Synthesis of (2,2,2-trifluoroethyl)-1,2,3,4-tetrahydrobenzofuran[2,3-c]pyridin-6-yl trifluoromethanesulfonate
  • Step 9 (1-(2,6-Difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3-methyl-2- Synthesis of (2,2,2-trifluoroethyl)-1,2,3,4-tetrahydrobenzofuran[2,3-c]pyridin-6-yl)boronic acid
  • Step 1 Synthesis of tert-butyl N-[(1R)-2-(5-benzyloxy-1H-indol-3-yl)-1-methyl-ethyl]carbamate
  • Step 3 Synthesis of (2R)-1-(5-benzyloxy-1H-indol-3-yl)-N-(2,2,2-trifluoroethyl)propane-2-amine
  • Step 4 Synthesis of 3-[(2R)-2-(2,2,2-trifluoroethylamino)propyl]-1H-indol-5-phenol
  • Step 5 (1R,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3- Synthesis of methyl-2-(2,2,2-trifluoroethyl)-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indole-6-phenol
  • Step 6 (1R,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3- Methyl-2-(2,2,2-trifluoroethyl)-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indol-6-yltrifluoromethanesulfonate Synthesis of acid esters
  • Step 7 ((1R,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3 -Methyl-2-(2,2,2(trifluoroethyl)-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indol-6-yl)boronic acid synthesis
  • Step 1 tert-Butyl (3-fluoropropyl) (2-((6-((1S,3R)-6-hydroxy-3-methyl-2-(2,2,2-trifluoroethyl) -Synthesis of 1,2,3,4-tetrahydroisoquinolin-1-yl)pyridin-3-yl)oxo)ethyl)amino methyl ester
  • Step 2 (1S,3R)-1-(5-(2-((tert-butoxycarbonyl)(3-fluoropropyl)amino)ethoxy)pyridin-2-yl)-3-methyl- Synthesis of 2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinolin-6-yl trifluoromethanesulfonate
  • Step 3 ((1S,3R)-1-(5-(2-((tert-butoxycarbonyl)(3-fluoropropyl)amino)ethoxy)pyridin-2-yl)-3-methyl Synthesis of -2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • Step 4 ((1S,3R)-1-(5-(2-((3-fluoropropyl)amino)ethoxy)pyridin-2-yl)-3-methyl-2-(2,2 ,2-Trifluoroethyl)-1,2,3,4-tetrahydroisoquinolin-6-yl) boronic acid
  • Step 1 Synthesis of (R)-tert-butyl(1-(3-methoxy-2-methylphenyl)propan-2-yl)carbamate
  • Step 3 Synthesis of (R)-1-(3-methoxy-2-methylphenyl)-N-(2,2,2-trifluoroethyl)propane-2-amine
  • Step 5 (1S,3R)-1-(5-((1-(3-fluoropropyl)azetidin-3-yl)amino)pyridin-2-yl)-3,5-dimethyl Synthesis of 2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinoline-6-phenol
  • Step 6 (1S,3R)-1-(5-((1-(3-fluoropropyl)azetidin-3-yl)amino)pyridin-2-yl)-3,5-dimethyl Synthesis of 2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinolin-6-yl trifluoromethanesulfonate
  • Step 7 ((1S,3R)-1-(5-((1-(3-fluoropropyl)azetidin-3-yl)amino)pyridin-2-yl)-3,5-di Synthesis of methyl-2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • reaction solution was stirred and reacted at 80°C for 13 hours.
  • LCMS detects that the reaction is complete.
  • the reaction solution was cooled to room temperature and poured into water (12 mL) and stirred for 10 minutes, extracted with ethyl acetate (10 mL) twice, the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated to dryness under reduced pressure.
  • reaction solution was reacted at 80°C for 16 hours.
  • LCMS monitors the completion of the reaction.
  • Step 4 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3- Synthesis of methyl-2-phenyl-1,2,3,4-tetrahydroisoquinoline-6-phenol
  • Step 5 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3- Synthesis of methyl-2-phenyl-1,2,3,4-tetrahydroisoquinolin-6-yl trifluoromethanesulfonate.
  • Step 6 ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3 -Methyl-2-phenyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid and ((1R,3R)-1-(2,6-difluoro-4-(( 1-(3-Fluoropropyl)azetidin-3-yl)amino)phenyl)-3-methyl-2-phenyl-1,2,3,4-tetrahydroisoquinoline-6 -Based) boronic acid synthesis
  • reaction solution was stirred and reacted at 80°C for 2 hours. After the reaction is detected by LCMS, the reaction solution is cooled to room temperature, concentrated under reduced pressure and then subjected to preparative liquid chromatography (Phenomenex Gemini-NX C18 column, 3um silica, 30mm diameter, 75mm length); (use water (containing 0.225%) ((1R,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)nitrogen) with a mixture of formic acid) and acetonitrile with decreasing polarity as the eluent).
  • Etidine-3-yl)amino)phenyl)-3-methyl-2-phenyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid first peak, Example 10, 4.29 mg
  • ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino) Phenyl)-3-methyl-2-phenyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid second peak, Example 11, 5.19 mg).
  • Example 12 ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)oxo)phenyl) Synthesis of -3-methyl-2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • Step 1 Synthesis of tert-butyl 3-(3,5-difluoro-4-formylphenoxy)azetidine-1-carboxylate
  • Step 3 Synthesis of 2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)oxo)benzaldehyde
  • Step 4 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)oxo)phenyl)-3 Synthesis of -methyl-2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinoline-6-phenol
  • Step 5 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)oxo)phenyl)-3 Synthesis of -methyl-2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinolin-6-yl trifluoromethanesulfonate
  • Step 6 ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)oxo)phenyl)- Synthesis of 3-methyl-2-(2,2,2-trifluoroethyl)-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • reaction solution was stirred and reacted at 90°C for 2 hours.
  • LCMS detects that the reaction is complete.
  • the reaction solution was cooled to room temperature, poured into water (20 mL) and stirred for 10 minutes, extracted with ethyl acetate (20 mL) twice, the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated to dryness under reduced pressure.
  • Examples 13 and 14 ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl )-2-(4-Fluorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid and ((1R,3R)-1-(2,6 -Difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2-(4-fluorophenyl)-3-methyl-1, Synthesis of 2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • Step 4 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2- Synthesis of (4-fluorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinoline-6-phenol
  • Step 5 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2- Synthesis of (4-fluorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl trifluoromethanesulfonate
  • Step 6 ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2 -(4-Fluorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid and ((1R,3R)-1-(2,6-difluoro -4-((1-(3-Fluoropropyl)azetidin-3-yl)amino)phenyl)-2-(4-fluorophenyl)-3-methyl-1,2,3 Synthesis of ,4-tetrahydroisoquinolin-6-yl)boronic acid
  • reaction solution was stirred and reacted at 90°C for 4 hours. After the reaction is detected by LCMS, the reaction solution is cooled to room temperature, concentrated under reduced pressure and then subjected to preparative liquid chromatography (Phenomenex Gemini-NX C18 column, 3um silica, 30mm diameter, 75mm length); (use water (containing 0.225%) A mixture of formic acid) and acetonitrile with decreasing polarity was used as the eluent), and the first peak and the second peak were obtained by purification.
  • the first peak is obtained by chiral separation (DAICEL CHIRALPAK AD column, 5um silica, 30mm diameter, 250mm length); (using a mixture of ethanol (containing 0.1% ammonia) and water with decreasing polarity as the eluent) (( 1S,3R)-1-(2,6-Difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2-(4-fluoro Phenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid (Example 13, 4.41 mg).
  • DICEL CHIRALPAK AD column 5um silica, 30mm diameter, 250mm length
  • the second peak is separated by chirality (DAICEL CHIRALCEL OD-H column, 5um silica, 30mm diameter, 250mm length); (using a mixture of ethanol (containing 0.1% ammonia) and water with decreasing polarity as the eluent) And preparative liquid chromatography purification (Phenomenex Synergi C18 column, 4um silica, 30mm diameter, 150mm length); (using a mixture of water (containing 0.05% hydrochloric acid) and acetonitrile as the eluent of decreasing polarity) to obtain (( 1R,3R)-1-(2,6-Difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2-(4-fluoro Phenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid (Example 14, 1.54 mg).
  • Examples 15 and 16 ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl )-2-(2,4-Difluorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid and ((1R,3R)-1-( 2,6-Difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2-(2,4-difluorophenyl)-3 -Methyl-1,2,3,4-tetrahydroisoquinolin-6-yl) synthesis of boronic acid
  • Step 1 Synthesis of (R)-2,4-difluoro-N-(1-(3-methoxyphenyl)propan-2-yl)aniline
  • Step 3 (1S,3R)-1-(4-bromo-2,6-difluorophenyl)-2-(2,4-difluorophenyl)-3-methyl-1,2,3, Synthesis of 4-tetrahydroisoquinoline-6-phenol
  • Step 4 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2- Synthesis of (2,4-Difluorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinoline-6-phenol
  • Step 5 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2- Synthesis of (2,4-Difluorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl trifluoromethanesulfonate
  • Step 6 ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2 -(2,4-Difluorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid and ((1R,3R)-1-(2,6 -Difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2-(2,4-difluorophenyl)-3-methyl Synthesis of -1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • reaction solution was stirred and reacted at 90°C for 2 hours.
  • LCMS detects that the reaction is complete.
  • the reaction solution was cooled to room temperature, poured into water (20 mL) and stirred for 10 minutes, extracted with ethyl acetate (40 mL) twice, the organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated to dryness under reduced pressure.
  • Example 15 ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)- 2-(2,4-Difluorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid.
  • Example 16 ((1R,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)- 2-(2,4-Difluorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • Examples 17 and 18 ((1S,3R)-2-(4-cyclopropylphenyl)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)aza Cyclobutan-3-yl)amino)phenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid and ((1R,3R)-2-(4 -Cyclopropylphenyl)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3-methyl Synthesis of 1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • Step 1 Synthesis of (R)-4-cyclopropyl-N-(1-(3-methoxyphenyl)propan-2-yl)aniline
  • reaction solution was stirred and reacted at 80°C for 16 hours. TLC monitors after completion of the reaction.
  • Step 3 (1S,3R)-1-(4-bromo-2,6-difluorophenyl)-2-(4-cyclopropylphenyl)-3-methyl-1,2,3,4 -Synthesis of Tetrahydroisoquinoline-6-phenol
  • Step 4 (1S,3R)-2-(4-cyclopropylphenyl)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidine- Synthesis of 3-yl)amino)phenyl)-3-methyl-1,2,3,4-tetrahydroisoquinoline-6-phenol
  • reaction solution was stirred and reacted at 120°C for 2 hours.
  • LCMS monitored the completion of the reaction.
  • the reaction solution was concentrated to dryness under reduced pressure, it was extracted with ethyl acetate (10mL*3) and water (10mL).
  • Step 5 (1S,3R)-2-(4-cyclopropylphenyl)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidine- Synthesis of 3-yl)amino)phenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl trifluoromethanesulfonate
  • Step 6 ((1S,3R)-2-(4-cyclopropylphenyl)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidine) -3-yl)amino)phenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid and ((1R,3R)-2-(4-cyclopropyl) Phenyl)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-3-methyl-1 Synthesis of 2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • reaction solution was stirred and reacted at 90°C for 2 hours. After the reaction is detected by LCMS, the reaction solution is cooled to room temperature, concentrated under reduced pressure, and then subjected to preparative liquid chromatography (Phenomenex Gemini-NX C18 column, 3um silica, 30mm diameter, 75mm length); (use water (containing 0.225%) (1S, 3R)-2-(4-cyclopropylphenyl)-1-(2,6-difluoro-4-( (1-(3-Fluoropropyl)azetidin-3-yl)amino)phenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid (The first peak,
  • Example 17 13.93mg and ((1R,3R)-2-(4-cyclopropylphenyl)-1-(2,6-difluoro-4-((1-(3-fluoropropyl) Azetidine-3-yl)amino)phenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid (second peak, Example 18, 16.23mg).
  • Examples 19 and 20 ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl )-2-(4-chlorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid and ((1R,3R)-1-(2,6 -Difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2-(4-chlorophenyl)-3-methyl-1, Synthesis of 2,3,4-tetrahydroisoquinolin-6-yl)boronic acid
  • reaction solution was stirred and reacted at 80°C for 16 hours.
  • the reaction solution was diluted with ethyl acetate (30mL) and water (15mL). After filtration, the filtrate was extracted three times with ethyl acetate (20mL). The collected organic phase was dried with anhydrous sodium sulfate, filtered and concentrated under reduced pressure to dryness, and then passed through a column.
  • reaction solution was slowly added dropwise to ice water (20 mL) for quenching, and then saturated sodium carbonate solution was added dropwise to the solution to adjust the pH of the solution to weakly alkaline.
  • the mixed solution was extracted three times with dichloromethane (50mL), the collected organic phase was dried and filtered with anhydrous sodium sulfate, and concentrated under reduced pressure to obtain the crude product (R)-3-(2-((4-chlorophenyl)amino ) Propyl) phenol (420.00 mg).
  • Step 4 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2- Synthesis of (4-chlorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinoline-6-phenol
  • reaction solution was stirred and reacted at 120°C for 3 hours. The completion of the reaction was monitored by LCMS.
  • Step 5 (1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2- Synthesis of (4-fluorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl trifluoromethanesulfonate
  • reaction solution was stirred and reacted at 25°C for 16 hours.
  • Step 6 ((1S,3R)-1-(2,6-difluoro-4-((1-(3-fluoropropyl)azetidin-3-yl)amino)phenyl)-2 -(4-chlorophenyl)-3-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)boronic acid and ((1R,3R)-1-(2,6-difluoro -4-((1-(3-Fluoropropyl)azetidin-3-yl)amino)phenyl)-2-(4-chlorophenyl)-3-methyl-1,2,3 Synthesis of ,4-tetrahydroisoquinolin-6-yl)boronic acid
  • reaction solution was stirred and reacted at 90°C for 3 hours. After the reaction is detected by LCMS, the reaction solution is cooled to room temperature, concentrated under reduced pressure and then subjected to preparative liquid chromatography (Phenomenex Synergi C18 column, 4um silica, 30mm diameter, 150mm length); (using water (containing 0.225% formic acid) A mixture of decreasing polarity with acetonitrile was used as eluent) product (35 mg).
  • Test Example 1 Detection of the degradation effect of the compound of the present invention on the estrogen receptor in MCF7 cells
  • the purpose of this experiment is to determine the degradation activity of the compound of the present invention on the endogenously expressed estrogen receptor in MCF7 cells, and evaluate the activity of the compound based on the IC50 and the maximum degradation efficiency.
  • MCF7 cells (ATCC, HTB-22) were cultured in DMEM (Gibco, 11995-065) complete medium containing 10% fetal bovine serum. On the first day of the experiment, MCF7 cells were seeded in a 384-well plate at a density of 3000 cells/well using a complete medium, and cultured in a cell incubator at 37°C and 5% CO 2. The compounds to be tested were dissolved in DMSO at a storage concentration of 10mM, diluted with Echo 550 (Labcyte Inc.) and added to the cell culture plate.
  • the initial concentration of each compound treatment was 100nM, with a 3-fold dilution, 9 concentration points, and setting containing A blank control of 0.5% DMSO, and a double-well control for each concentration point. Incubate for 24 hours in a 37°C, 5% CO 2 cell incubator. Add paraformaldehyde to each cell culture well to fix the cells at a final concentration of about 3.7%.
  • Table 1 The inhibitory activity and maximum inhibitory rate of the compounds of the present invention on estrogen receptors in MCF7 cells
  • Example 1 Compound number ER level IC 50 (nM) Maximum inhibition rate Example 1 0.25 98% Example 2 0.13 97% Example 3 0.08 98% Example 4 0.19 100% Example 5 0.45 96% Example 6 1.85 96% Example 7 0.68 98% Example 8 1.04 85% Example 9 0.64 98% Example 10 0.51 92% Example 11 0.29 97% Example 12 0.14 97% Example 13 0.39 92% Example 14 0.24 99% Example 15 1.28 95% Example 16 0.56 99% Example 17 1.57 98% Example 18 0.5 96% Example 19 1.86 96% Example 20 1.64 94%
  • Test Example 2 Detection of the inhibitory effect of the compound of the present invention on the proliferation of MCF7 cells
  • MCF7 cells (ATCC, HTB-22) were cultured in DMEM (Gibco, 11995-065) complete medium containing 10% fetal bovine serum. On the first day of the experiment, MCF7 cells were seeded in a 384-well plate at a density of 500 cells/well using complete medium, and cultured overnight in a 37°C, 5% CO 2 cell incubator. The next day, add the test compound for drug treatment, use Echo550 (Labcyte Inc.) to dilute the compound solution with a storage concentration of 10mM and transfer it to each cell culture well.
  • Echo550 (Labcyte Inc.)
  • the initial concentration of each compound in the cell is 100nM , 3-fold gradient dilution, 9 concentration points, set a blank control containing 0.3% DMSO, and set a double-well control for each concentration point.
  • 37°C, 5% CO 2 cell incubator was cultured for 7 days, and on the eighth day, the cell culture plate was taken out.
  • join in Luminescent Cell Viability Assay Promega, G7573
  • use the multi-label microplate reader EnVision (PerkinElmer) to read the luminescence signal value
  • use XLfit to calculate the inhibitory activity IC 50 of each compound based on the compound concentration and luminescence signal value And the maximum inhibition rate Imax, the results are shown in Table 2 below.
  • Test Example 3 Determination of the metabolic stability of the compound of the present invention in liver microsomes
  • liver microsomes human liver microsomes (Corning 452117), CD-1 mouse liver microsomes (XENOTECH M1000)
  • Test Example 4 Determination of the plasma protein binding rate of the compound of the present invention
  • HTDialysis LLC Gales Ferry, CT, HTD96B
  • MWCO 12-14K #1101
  • Preparation of buffer solution with a concentration of 100 mM sodium phosphate and 150 mM NaCl prepare an alkaline solution with a concentration of 14.2 g/L Na 2 HPO 4 and 8.77 g/L NaCl with ultra-pure water, and use ultra-pure water to prepare a concentration of An acidic solution of 12.0g/L NaH 2 PO 4 and 8.77g/L NaCl. Titrate the alkaline solution with an acidic solution to a pH of 7.4 to prepare a buffer solution with a concentration of 100 mM sodium phosphate and 150 mM NaCl.
  • Preparation of the dialysis membrane soak the dialysis membrane in ultrapure water for 60 minutes to separate the membrane into two pieces, then soak it in 20% ethanol for 20 minutes, and finally soak it in the buffer for dialysis for 20 minutes.
  • Preparation of plasma Thaw the frozen plasma quickly at room temperature, then centrifuge the plasma at 4°C and 3,220g for 10 minutes to remove clots, and collect the supernatant in a new centrifuge tube. Measure and record the pH of plasma, using plasma with a pH of 7-8.
  • the peak areas of the compound on the buffer side and the plasma side were measured.
  • the formula for calculating the plasma protein binding rate of the compound is as follows:
  • % Free rate (compound peak area to internal standard peak area ratio buffer side/compound peak area to internal standard peak area ratio plasma side) * 100%
  • Table 4 The protein binding rate value of the compound of the present invention in CD-1 mouse plasma
  • Example number % Binding rate Example 1 99.8%
  • Example 2 99.9%
  • Example 3 98%
  • Example 4 97%
  • the membrane permeability and transport characteristics of the compounds of the present invention are measured using the following test methods.
  • HEPES Solarbio 804D049
  • Penicillin/Streptomycin Solarbio 20200109
  • Trypsin/EDTA Solarbio
  • PBS PBS
  • Fetal Bovine Serum (Sigma WXBD0055V), Fluorescent Yellow (Sigma MKCJ3738), NaHCO 3 (Sigma SLBZ4647)
  • Caco-2 cell culture medium high-sugar DMEM (containing L-glutamine) medium was prepared by adding FBS, penicillin, streptomycin, kanamycin and NEAA to contain 10% FBS, 0.1 mg/ mL streptomycin, 100 units of penicillin, 0.6 ⁇ g/mL kanamycin and 1 ⁇ NEAA cell culture medium.
  • the cells are cultured in a T-75 culture flask in an incubator at 37° C. and 5% CO 2 , and the culture medium is discarded when the cell growth reaches 80-90% density. Wash the cells with 5mL PBS, add 1.5mL Trypsin/EDTA, then incubate in a 37°C incubator for 5-10 minutes until the cells fall off as a quicksand, and finally neutralize the Trypsin/EDTA with a medium containing FBS.
  • TEER value TEER ( ⁇ ) measured value ⁇ film area (cm 2 )
  • the electrical resistance of the monolayer cell membrane is less than 230 ⁇ cm 2 , which indicates that the monolayer cell membrane has poor compactness and cannot be used in the test.
  • I acceptor refers to the fluorescence density on the receiving side (0.3 mL), and I donor refers to the fluorescence density on the dosing side (0.1 mL).
  • LY>1.0% means that the monolayer cell membrane has poor compactness, and the corresponding results will be excluded from the evaluation.
  • V A is the volume of the receiving end solution (A ⁇ B is 0.3 mL, B ⁇ A is 0.1 mL), Area is the membrane area of the Transwell-96-well plate (0.143 cm 2 ); incubation time is the incubation time (unit: s).
  • P app (BA) is the apparent permeability coefficient from the base end to the top
  • P app (AB) is the apparent permeability coefficient from the top end to the base end.
  • Test Example 6 The inhibitory effect of the compound of the present invention on the enzyme activities of CYP2C9, CYP2D6, and CYP3A4
  • the inhibition of the CYP2C9, CYP2D6, and CYP3A4 enzyme activities by the compounds of the present invention was determined by the following test method.
  • CD-1 mice were purchased from Beijing Weitong Lihua Laboratory Animal Technology Co., Ltd.
  • DMSO dimethyl sulfoxide
  • HP- ⁇ -CD hydroxypropyl- ⁇ -cyclodextrin
  • Tetraethylene Glycol tetraethylene glycol
  • Captisol SBE- ⁇ -CD, sulfobutyl- ⁇ -Cyclodextrin
  • Acetonitrile was purchased from Merck (USA).
  • mice 6 female CD-1 mice (20-30g, 4-6 weeks) were randomly divided into 2 groups, 3 mice in each group.
  • the first group was given the test compound by tail vein injection at a dose of 1 mg/kg with a vehicle of 5% DMSO in 10% HP- ⁇ -CD in water, and the second group was given orally with the test compound at a dose of 10 mg/kg with a vehicle of 40% Tetraethylene Glycol ( v/v), 7.5% Captisol(w/v) in water. Feed and water normally before the animal experiment.
  • Mice in each group were subjected to intravenous blood sampling at 0.083 (intravenous injection group only), 0.25, 0.5, 1, 2, 4, 6, 8 and 24 hours after administration. The collected whole blood samples were placed in a K 2 EDTA anticoagulation tube, and after centrifugation for 5 min (12000 rpm, 4° C.), plasma was taken for testing.
  • the PK test results are as follows:
  • Test Example 8 Growth inhibition experiment of compound on MCF-7 xenograft tumor
  • Human breast cancer MCF-7 cells ECACC-86012803
  • EMEM culture medium ATCC, Cat No.: 30-2003
  • Fetal Bovine Serum Hyclone; Cat No.: SV30087.03
  • Pancreatin-EDTA Gibco, Cat No.: 25200-072
  • mice Balb/c nude mice, female, 6-8 weeks old, weighing about 18-22 grams, the animals were purchased from Shanghai Lingchang Biological Technology Co., Ltd. The mice were kept in an SPF environment, each cage With separate air supply and exhaust, all animals have free access to standard certified commercial laboratory food and drinking water.
  • Cell culture Human breast cancer MCF-7 cell line is cultured in vitro, and the culture conditions are EMEM (cell culture medium) with 10% fetal bovine serum, 1% Antibiotic-Antimycotic, 37°C, 5% CO 2 incubator. Use 0.25% pancreatin-EDTA digestion solution twice a week for routine digestion and passage. When the cell saturation is 80%-90% and the number reaches the requirement, the cells are collected and counted.
  • Cell inoculation 0.2ml/(containing 1 ⁇ 10 7 ) MCF-7 cell suspension (DPBS plus Matrigel, volume ratio 1:1) was subcutaneously inoculated on the right back of each mouse, and before cell inoculation 17 ⁇ -estradiol tablets were inoculated subcutaneously in two days. On the 6th day after cell inoculation, drugs were administered in random groups according to the tumor volume, and the day of grouping was Day 0.
  • the dose of the positive drug Fulvestrant is 250 mg/kg, administered by subcutaneous injection (SC), once a week (QW) x 3 weeks; compound SCR-6139 is administered The dosage is 10 mg/kg, oral administration (PO), once a day (QD) x 3 weeks or 30 mg/kg, PO, QD x 3 weeks. Each group has 6 mice.
  • the tumor diameter was measured with vernier calipers twice a week.
  • the body weight of the mice was measured twice a week.
  • TGI tumor growth inhibition rate
  • Example 1 and Example 3 administered orally at 30 mg/kg once a day has a significant inhibitory effect on tumor growth (P ⁇ 0.01), and has a shrinking effect. The effect of tumors.
  • Example 1 and Example 3 did not significantly affect the body weight of the mice at the tried doses, nor did they cause any death of the mice, and the mice could tolerate them.

Abstract

本发明提供了式(I)所示的含硼化合物或其药学可接受的盐,含有它们的药物组合物以及作为选择性雌激素受体降解剂(SERD)在预防或治疗相关性疾病中的用途。

Description

含硼化合物及其应用
本申请要求2020年04月21日向中国国家知识产权局提交的,专利申请号为202010317185.4,发明名称为“含硼化合物及其应用”的在先申请的优先权。所述申请的全文通过引用的方式结合于本申请中。
技术领域
本发明涉及一种新型的含硼化合物或其药学可接受的盐,含有它们的药物组合物以及作为选择性雌激素受体降解剂(SERD)在预防或治疗相关性疾病中的用途。
背景技术
雌激素(E2)及雌激素α受体(ERα)是乳腺癌发生发展的重要驱动因子。在乳腺癌患者中有超过2/3的患者表达ER转录因子,并且在大多数ER阳性患者中,即使经过早期的内分泌治疗后进展的肿瘤中,ER仍是一个关键的驱动因子,因此ER是乳腺癌治疗的一个主要靶点(Pharmacology&Therapeutics 186(2018)1–24)。内分泌治疗目的是降低ER活性,主要有三类,包括选择性雌激素受体调节剂(SERMs),比如他莫昔芬(tamoxifen),是ER的别构调节剂,同ER结合后抑制其转录活性;芳香化酶抑制剂(aromatase inhibitors,AIs),通过抑制雄激素转化为雌激素,减低体内雌激素水平;以及选择性雌激素受体下调剂,比如氟维司群(fulvestrant),不仅作为ER的拮抗剂抑制其活性,还具有诱导ER蛋白降解的作用。虽然内分泌治疗是雌激素受体阳性乳腺癌患者的首选,但是约有30%的治疗后病人会发生复发,并且几乎所有的转移性乳腺癌患者都会产生耐药而发生进展。内分泌治疗产生耐药的机制主要有两类,一是集中在雌激素受体信号通路本身,包括编码雌激素受体的基因ESR1的激活突变,扩增,与其他基因的融合,雌激素受体共调节因子和下游控制细胞周期因子的失调等;另一类机制包括与雌激素受体信号通路有交叉反应的信号通路的激活,比如生长因子受体通路等(Oncol Ther,2017,5:17–29)。
氟维司群是首个也是唯一经临床批准用于他莫昔芬或芳香化酶抑制剂进展后治疗ER阳性、转移性乳腺癌的绝经后患者的SERD类药物。此外,阿斯利康(参见专利申请WO2018077630A1)及基因泰克公司(参见专利申请WO2019245974A1)也公开了一系列结构新颖的SERD类化合物及相应的医药用途。多项研究数据显示经氟维司群治疗的患者体内并未能完全实现ER的降解,不过也可能是氟维司群的剂量(最高只能达到500mg,主要是其药效动力学特征和肌内给药途径限制了其可给予患者的最高剂量)限制了其药效。因此,具有更高生物利用度、对ER的拮抗活性更高、并能在更大程度上的ER降解、以及可以用于雌激素水平较高的绝经前患者、方便的口服给药的SERD类药物是临床上迫切需要的。
发明内容
本发明提供一种式(I)所示化合物或其药学上可接受的盐:
Figure PCTCN2021088291-appb-000001
其中,
Figure PCTCN2021088291-appb-000002
选自
Figure PCTCN2021088291-appb-000003
R 1、R 2独立选自OH,或者R 1、R 2以及它们所连接的硼原子共同形成5-6元杂环基,所述5-6元杂环基任选被C 1-C 10烷基取代;
X 1、X 2、X 3、X 4独立地选自CR 7或N;
R 7选自H、F、Cl、Br、I、OH、CN、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基或3-10元杂环基氧基;
X、Y独立选自O或NH;
R 3选自H、F、Cl、Br、I、OH、CN、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基或3-10元杂环基氧基;
R 4选自H、C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基;
或者R 3、R 4以及它们所连接的原子共同形成4-10元杂环基,所述4-10元杂环基任选被R a1取代,所述R a1选自F、Cl、Br、I、OH、CN、C 1-C 10烷基或C 1-C 10烷氧基;
R 5、R 6独立选自C 1-C 6烷基或苯基,所述C 1-C 6烷基或苯基任选被R a2取代;
R 8选自H或C 1-C 6烷基,所述C 1-C 6烷基任选被R a2取代;
R a2选自F、Cl、Br、I、OH、CN或任选被R b取代的以下基团:C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基;
R b选自F、Cl、Br、I、OH、CN、C 1-C 10烷基或C 1-C 10烷氧基;
条件是,式(I)所示化合物不包含
Figure PCTCN2021088291-appb-000004
在一些实施方案中,所述式(I)所示化合物或其药学上可接受的盐,其中,
Figure PCTCN2021088291-appb-000005
选自
Figure PCTCN2021088291-appb-000006
R 1、R 2独立选自OH,或者R 1、R 2以及它们所连接的硼原子共同形成5-6元杂环基,所述5-6元杂环基任选被C 1-C 10烷基取代;
X 1、X 2、X 3、X 4独立地选自CR 7或N;
R 7选自H、F、Cl、Br、I、OH、CN、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基或3-10元杂环基氧基;
X、Y独立选自O或NH;
R 3选自H、F、Cl、Br、I、OH、CN、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基或3-10元杂环基氧基;
R 4选自H、C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基;
或者R 3、R 4以及它们所连接的原子共同形成4-10元杂环基,所述4-10元杂环基任选被R a1取代,所述R a1选自F、Cl、Br、I、OH、CN、C 1-C 10烷基或C 1-C 10烷氧基;
R 5、R 6独立选自C 1-C 6烷基或苯基,所述C 1-C 6烷基或苯基任选被R a2取代;
R a2选自F、Cl、Br、I、OH、CN或任选被R b取代的以下基团:C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基;
R b选自F、Cl、Br、I、OH、CN、C 1-C 10烷基或C 1-C 10烷氧基;
条件是,式(I)所示化合物不包含
Figure PCTCN2021088291-appb-000007
在一些实施方案中,
Figure PCTCN2021088291-appb-000008
选自
Figure PCTCN2021088291-appb-000009
在一些实施方案中,R 1、R 2独立选自OH,或者R 1、R 2以及它们所连接的硼原子共同形成5-6元杂环基,所述5-6元杂环基任选被甲基取代。
在一些实施方案中,R 1、R 2独立选自OH,或者R 1、R 2以及它们所连接的硼原子共同形成5-6元杂环烷基,所述5-6元杂环烷基被1个、2个、3个或4个甲基取代,除了硼原子外,所述5-6元杂环烷基中的杂原子还包含2-3个氧原子。
在一些实施方案中,R 1、R 2独立选自OH,或者R 1、R 2以及它们所连接的硼原子共同形成的5-6元杂环烷基选自
Figure PCTCN2021088291-appb-000010
在一些实施方案中,R 1、R 2独立选自OH。
在一些实施方案中,X 1、X 2、X 3、X 4独立地选自CR 7或N,且X 1、X 2、X 3、X 4基团中的至少2个选自CR 7
在一些实施方案中,X 1、X 2、X 3、X 4独立地选自CR 7或N,且X 1、X 2、X 3、X 4基团中的至少3个选自CR 7
在一些实施方案中,所述R 7选自H、F、Cl、Br、I、CN、C 1-C 3烷基或C 1-C 3烷氧基。
在一些实施方案中,所述R 7选自H、F、Cl、Br、I、CN或C 1-C 3烷氧基。
在一些实施方案中,所述R 7选自H、F或甲氧基。
在一些实施方案中,所述R 7选自H或F。
在一些实施方案中,结构单元
Figure PCTCN2021088291-appb-000011
选自
Figure PCTCN2021088291-appb-000012
在一些实施方案中,结构单元
Figure PCTCN2021088291-appb-000013
选自
Figure PCTCN2021088291-appb-000014
在一些实施方案中,R 3选自H、F、Cl、Br、I、OH、CN、C 1-C 6烷基或C 1-C 6烷氧基。
在一些实施方案中,R 4选自H或C 1-C 6烷基。
在一些实施方案中,R 3、R 4均选自H。
在一些实施方案中,R 3、R 4以及它们所连接的原子共同形成4-7元杂环基,所述4-7元杂环基选自4-6元单杂环基或6-7元螺杂环基,所述4-6元单杂环基或6-7元螺杂环基任选被R a1取代。
在一些实施方案中,R 3、R 4以及它们所连接的原子共同形成4元杂环基,所述4元杂环基任选被R a1取代。
在一些实施方案中,所述R a1选自F、Cl、Br、I、CN、C 1-C 6烷基或C 1-C 6烷氧基。
在一些实施方案中,所述R a1选自F、Cl、Br、I、CN或C 1-C 3烷基。
在一些实施方案中,所述R a1选自F、Cl、Br、I、CN或甲基。
在一些实施方案中,所述R a1选自F或甲基。
在一些实施方案中,R 3、R 4以及所连接的原子共同形成
Figure PCTCN2021088291-appb-000015
Figure PCTCN2021088291-appb-000016
在一些实施方案中,R 3、R 4以及所连接的原子共同形成
Figure PCTCN2021088291-appb-000017
在一些实施方案中,R 5、R 6独立选自C 1-C 6烷基或苯基,所述C 1-C 6烷基或苯基任选被R a2取代。
在一些实施方案中,R 5、R 6独立选自C 1-C 3烷基或苯基,所述C 1-C 3烷基任选被R a2取代。
在一些实施方案中,所述R a2选自F、Cl、Br、I、OH、CN或任选被R b取代的C 3-C 6环烷基。
在一些实施方案中,所述R a2选自F、Cl、Br、I、OH、CN或任选被R b取代的环丙基。
在一些实施方案中,所述R a2选自F、OH、CN或任选被R b取代的环丙基。
在一些实施方案中,所述R b选自F、Cl、Br、I、OH、CN或C 1-C 6烷基。
在一些实施方案中,所述R b选自F、Cl、Br、I或CN。
在一些实施方案中,所述R b选自F。
在一些实施方案中,所述R a2选自F、Cl、OH或任选被F取代的环丙基。
在一些实施方案中,R 5、R 6独立选自CH 2CF 3、CH 2CHF 2、CH 2CF 2CH 2OH、CH 2CF 2CH 2CN、
Figure PCTCN2021088291-appb-000018
苯基、CH 2C(CH 3) 2F、对氟苯基、对氯苯基、
Figure PCTCN2021088291-appb-000019
在一些实施方案中,R 5、R 6独立选自CH 2CF 3、CH 2CHF 2、CH 2CF 2CH 2OH、CH 2CF 2CH 2CN、
Figure PCTCN2021088291-appb-000020
或苯基。
在一些实施方案中,R 5、R 6独立选自CH 2CF 3、CH 2CF 2CH 2OH、
Figure PCTCN2021088291-appb-000021
苯基、CH 2C(CH 3) 2F、对氟苯基、对氯苯基、
Figure PCTCN2021088291-appb-000022
在一些实施方案中,R 6选自CH 2CF 3
在一些实施方案中,R 8选自H或C 1-C 3烷基。
在一些实施方案中,R 8选自H或甲基。
在一些实施方案中,所述式(I)所示的化合物或其药学可接受的盐,选自式(II)所示化合物或其药学可接受的盐:
Figure PCTCN2021088291-appb-000023
其中
Figure PCTCN2021088291-appb-000024
选自
Figure PCTCN2021088291-appb-000025
R 1、R 2、R 3、R 4、R 5、R 6、X 1、X 2、X 3、X 4、X、Y如上文定义。
在一些实施方案中,所述式(I)所示的化合物或其药学可接受的盐,选自式(II)所示化合物或其药学可接受的盐:
Figure PCTCN2021088291-appb-000026
其中
Figure PCTCN2021088291-appb-000027
选自
Figure PCTCN2021088291-appb-000028
R 1、R 2、R 3、R 4、R 5、R 6、R 8、X 1、X 2、X 3、X 4、X、Y如上文定义。
在一些实施方案中,结构单元
Figure PCTCN2021088291-appb-000029
选自
Figure PCTCN2021088291-appb-000030
在一些实施方案中,结构单元
Figure PCTCN2021088291-appb-000031
选自
Figure PCTCN2021088291-appb-000032
在一些实施方案中,所述式(I)所示的化合物或其药学可接受的盐,选自以下化合物或其药学可接受的盐:
Figure PCTCN2021088291-appb-000033
在一些实施方案中,所述式(I)所示的化合物或其药学可接受的盐,选自以下化合物或其药学可接受的盐:
Figure PCTCN2021088291-appb-000034
在一些实施方案中,所述式(I)所示的化合物或其药学可接受的盐,选自以下化合物或其药学可接受的盐:
Figure PCTCN2021088291-appb-000035
本发明还提供药物组合物,其包含式(I)所示化合物或其药学可接受的盐、药学可接受的载体和/或 赋形剂。
本发明还提供药物组合物,其包含式(I)所示化合物或其药学可接受的盐和药学上可接受的辅料。
进一步,本发明涉及式(I)所示的化合物或其药学上可接受的盐,或其药物组合物在制备预防或者治疗雌激素受体相关疾病的药物中的用途。
进一步,本发明涉及式(I)所示的化合物或其药学上可接受的盐,或其药物组合物在预防或者治疗雌激素受体相关疾病中的用途。
进一步,本发明涉及式(I)所示的化合物或其药学上可接受的盐,或其药物组合物作为选择性雌激素受体降解剂(SERD)在预防或治疗相关疾病中的用途。
进一步,本发明涉及预防或者治疗雌激素受体相关疾病的式(I)化合物或其药学上可接受的盐,或其药物组合物。
本发明还涉及治疗雌激素受体相关疾病的方法,该方法包括给以患者治疗上有效剂量的包含本发明所述的式(I)化合物或其药学上可接受的盐的药物制剂。
本发明的优选方案,其中所述的雌激素受体相关性疾病包括但不限于肿瘤(如乳腺癌)。
术语定义和说明
除非另有说明,本发明说明书和权利要求书中记载的基团和术语定义,包括其作为实例的定义、示例性的定义、优选的定义、表格中记载的定义、实施例中具体化合物的定义等,可以彼此之间任意组合和结合。这样的组合和结合后的基团定义及化合物结构,应当属于本发明说明书记载的范围内。一个特定的术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,而应该按照本领域普通的含义去理解。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。
本文中
Figure PCTCN2021088291-appb-000036
表示连接位点。
术语“药学上可接受的盐”是指药学上可接受的无毒酸或碱的盐,包括无机酸和碱、有机酸和碱的盐。
术语“立体异构体”是指由分子中原子在空间上排列方式不同所产生的异构体,包括顺反异构体、对映异构体、非对应异构体和构象异构体。
本发明的化合物可以具有不对称原子如碳原子、硫原子、氮原子、磷原子或不对称双键。外消旋体、对映异构体、非对映异构体、几何异构体和单个的异构体都包括在本发明的范围之内。
本文中消旋体或者对映体纯的化合物的图示法来自Maehr,J.Chem.Ed.1985,62:114-120。除非另有说明,用楔形键和虚楔键
Figure PCTCN2021088291-appb-000037
表示一个立体中心的绝对构型,用黑实键和虚键
Figure PCTCN2021088291-appb-000038
表示一个立体中心的相对构型(如脂环化合物的顺反构型)。当本文所述化合物含有烯属双键或其它几何不对称中心,除非另有规定,它们包括E、Z几何异构体。同样地,所有的互变异构形式均包括在本发明的范围之内。
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、不对称硫原子、不对称氮原子或不对称磷原子。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。本申请的含有不对称原子的化合物可以以光学活性纯的形式或外消旋形式被分离出来。光学活性纯的形式可以从外消旋混合物拆分,或通过使用手性原料或手性试剂合成。立体异构体的非限制性实例包括但不限于:
Figure PCTCN2021088291-appb-000039
术语“互变异构体”是指因分子中某一原子在两个位置迅速移动而产生的官能团异构体。本发明化合物可表现出互变异构现象。互变异构的化合物可以存在两种或多种可相互转化的种类。质子移变互变异构体来自两个原子之间共价键合的氢原子的迁移。互变异构体一般以平衡形式存在,尝试分离单一互变异构体时通常产生一种混合物,其理化性质与化合物的混合物是一致的。平衡的位置取决于分子内的化学特性。例如,在很多脂族醛和酮如乙醛中,酮型占优势;而在酚中,烯醇型占优势。本发明包含化合物的所有互变异构形式。
术语“药物组合物”表示一种或多种文本所述化合物或其生理学/药学上可接受的盐或前体药物与其它化学组分的混合物,其它组分例如生理学/药学上可接受的载体和赋形剂。药物组合物的目的是促进化合物对生物体的给药。
术语“被取代”是指特定原子上的任意一个或多个氢原子被取代基取代,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。当取代基为氧代(即=O)时,意味着两个氢原子被取代,氧代不会发生在芳香基上。
术语“任选”或“任选地”是指随后描述的事件或情况可以发生或不发生,该描述包括发生所述事件或情况和不发生所述事件或情况。例如,乙基“任选”被卤素取代,指乙基可以是未被取代的(CH 2CH 3)、单取代的(如CH 2CH 2F)、多取代的(如CHFCH 2F、CH 2CHF 2等)或完全被取代的(CF 2CF 3)。本领域技术人员可理解,对于包含一个或多个取代基的任何基团,不会引入任何在空间上不可能存在和/或不能合成的取代或取代模式。
术语“卤”或“卤素”是指氟、氯、溴和碘。
本文中的C m-C n,是指具有m-n范围中的整数个碳原子。例如“C 1-C 10”是指该基团可具有1个碳原子、2个碳原子、3个碳原子、4个碳原子、5个碳原子、6个碳原子、7个碳原子、8个碳原子、9个碳原子或10个碳原子。
术语“烷基”是指通式为C nH 2n+1的烃基。该烷基可以是直链或支链的。例如,术语“C 1-C 10烷基”应理解为表示具有1、2、3、4、5、6、7、8、9或10个碳原子的直链或支链饱和一价烃基。所述烷基是例如甲基、乙基、丙基、丁基、戊基、己基、异丙基、异丁基、仲丁基、叔丁基、异戊基、2-甲基丁基、1-甲基丁基、1-乙基丙基、1,2-二甲基丙基、新戊基、1,1-二甲基丙基、4-甲基戊基、3-甲基戊基、2-甲基戊基、1-甲基戊基、2-乙基丁基、1-乙基丁基、3,3-二甲基丁基、2,2-二甲基丁基、1,1-二甲基丁基、2,3-二甲基丁基、1,3-二甲基丁基或1,2-二甲基丁基等;优选地,“C 1-C 10烷基”可以包含“C 1-C 6烷基”或“C 1-C 3烷基”,“C 1-C 6烷基”应理解为表示具有1、2、3、4、5、6个碳原子的直链或支链饱和一价烃基,“C 1-C 3烷基”应理解为表示具有1、2、3个碳原子的直链或支链饱和一价烃基。
术语“烷氧基”指-O-烷基,例如术语“C 1-C 6烷氧基”可理解为“C 1-C 6烷基氧基”或“C 1-C 6烷基-O”,优选地,“C 1-C 6烷氧基”可以包含“C 1-C 3烷氧基”。
术语“环烷基”指完全饱和的并且可以以呈单环、并环、桥环或螺环存在的碳环。除非另有指示,该碳环通常为3至10元环。例如,术语“C 3-C 10环烷基”应理解为表示饱和的一价单环、并环、螺环或桥环,其具有3~10个碳原子。环烷基非限制性实例包括但不限于环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环壬基、环癸基,降冰片基(双环[2.2.1]庚基)、双环[2.2.2]辛基、金刚烷基、螺[4.5]癸烷等。螺环烷基指以螺环存在的环烷基。术语“C 3-C 10环烷基”可以包含“C 3-C 6环烷基”,“C 3-C 6环烷基”应理解为表示饱和的一价单环或双环烃环,其具有3~6个碳原子,具体实例包括环丙基、环丁基、环戊基、环己 基等。
术语“环烷基氧基”可理解为“环烷基-O-”。
术语“杂环基”是指完全饱和的或部分饱和的(整体上不是具有芳香性的杂芳族)一价单环、并环、螺环或桥环基团,其环原子中含有1-5个杂原子或杂原子团(即含有杂原子的原子团),所述“杂原子或杂原子团”包括但不限于氮原子(N)、氧原子(O)、硫原子(S)、磷原子(P)、硼原子(B),=O,=S,-O-N=,-C(=O)O-,-C(=O)-,-C(=S)-,-S(=O) 2-,-S(=O)-,以及任选被取代的-NH-,-S(=O)(=NH)-,-C(=O)NH-,-C(=NH)-,-S(=O) 2NH-,S(=O)NH-,-NHC(=O)NH-等。术语“3-10元杂环基”意指指环原子数目为3、4、5、6、7、8、9或10的杂环基,且其环原子中含有1-5个独立选自上文所述的杂原子或杂原子团,优选地,“3-10元杂环基”可以包括“4-10元杂环基”、“4-7元杂环基”、5-6元杂环基”等范围,其中,4元杂环基的非限制性实例包括但不限于氮杂环丁烷基、氧杂环丁烷基;5元杂环基的实例包括但不限于四氢呋喃基、二氧杂环戊烯基、吡咯烷基、咪唑烷基、吡唑烷基、吡咯啉基、4,5-二氢噁唑或2,5-二氢-1H-吡咯基;6元杂环基的实例包括但不限于四氢吡喃基、哌啶基、吗啉基、二噻烷基、硫代吗啉基、哌嗪基、三噻烷基、四氢吡啶基或4H-[1,3,4]噻二嗪基;7元杂环基的实例包括但不限于二氮杂环庚烷基。所述杂环基还可以是双环基,其中,5,5元双环基实例包括但不限于六氢环戊并[c]吡咯-2(1H)-基环,5,6元双环基实例包括但不限于六氢吡咯并[1,2-a]吡嗪-2(1H)-基环、5,6,7,8-四氢-[1,2,4]三唑并[4,3-a]吡嗪基环或5,6,7,8-四氢咪唑并[1,5-a]吡嗪。任选地,所述杂环基可以是上述4-7元杂环基的苯并稠合环基,实例包括但不限于二氢异喹啉基等。根据本发明,尽管有些双环类杂环基部分地含有一个苯环或一个杂芳环,但所述杂环基整体上仍是无芳香性的。
术语“杂环基氧基”可理解为“杂环基-O-”。
术语“治疗”意为将本申请所述化合物或制剂进行给药以预防、改善或消除疾病或与所述疾病相关的一个或多个症状,且包括:
(i)预防疾病或疾病状态在哺乳动物中出现,特别是当这类哺乳动物易患有该疾病状态,但尚未被诊断为已患有该疾病状态时;
(ii)抑制疾病或疾病状态,即遏制其发展;
(iii)缓解疾病或疾病状态,即使该疾病或疾病状态消退。
术语“治疗有效量”意指(i)治疗或预防特定疾病、病况或障碍,(ii)减轻、改善或消除特定疾病、病况或障碍的一种或多种症状,或(iii)预防或延迟本文中所述的特定疾病、病况或障碍的一种或多种症状发作的本发明化合物的用量。构成“治疗有效量”的本发明化合物的量取决于该化合物、疾病状态及其严重性、给药方式以及待被治疗的哺乳动物的年龄而改变,但可例行性地由本领域技术人员根据其自身的知识及本公开内容而确定。
术语“辅料”是指可药用惰性成分。术语“赋形剂”的种类实例非限制性地包括粘合剂、崩解剂、润滑剂、助流剂、稳定剂、填充剂和稀释剂等。赋形剂能增强药物制剂的操作特性,即通过增加流动性和/或粘着性使制剂更适于直接压缩。适用于上述制剂的典型的“药学上可接受的载体”的实例为:糖类,淀粉类,纤维素及其衍生物等在药物制剂中常用到的辅料。
术语“药学上可接受的辅料”是指对有机体无明显刺激作用,而且不会损害该活性化合物的生物活性及性能的那些辅料。合适的辅料是本领域技术人员熟知的,例如碳水化合物、蜡、水溶性和/或水可膨胀的聚合物、亲水性或疏水性材料、明胶、油、溶剂、水等。
词语“包括(comprise)”、“含有(comprise)”或“包含(comprise)”及其英文变体例如comprises或comprising应理解为开放的、非排他性的意义,即“包括但不限于”。
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。
本申请还包括与本文中记载的那些相同的,但一个或多个原子被原子量或质量数不同于自然中通常发现的原子量或质量数的原子置换的同位素标记的本申请化合物。可结合到本申请化合物的同位素的实例包括氢、碳、氮、氧、磷、硫、氟、碘和氯的同位素,诸如分别为 2H、 3H、 11C、 13C、 14C、 13N、 15N、 15O、 17O、 18O、 31P、 32P、 35S、 18F、 123I、 125I和 36Cl等。
某些同位素标记的本申请化合物(例如用 3H及 14C标记的那些)可用于化合物和/或底物组织分布分析 中。氚化(即 3H)和碳-14(即 14C)同位素对于由于它们易于制备和可检测性是尤其优选的。正电子发射同位素,诸如 15O、 13N、 11C和 18F可用于正电子发射断层扫描(PET)研究以测定底物占有率。通常可以通过与公开于下文的方案和/或实施例中的那些类似的下列程序,通过同位素标记试剂取代未经同位素标记的试剂来制备同位素标记的本申请化合物。
此外,用较重同位素(诸如氘(即 2H))取代可以提供某些由更高的代谢稳定性产生的治疗优点(例如增加的体内半衰期或降低的剂量需求),并且因此在某些情形下可能是优选的,其中氘取代可以是部分或完全的,部分氘取代是指至少一个氢被至少一个氘取代。
本申请的药物组合物可通过将本申请的化合物与适宜的药学上可接受的辅料组合而制备,例如可配制成固态、半固态、液态或气态制剂,如片剂、丸剂、胶囊剂、粉剂、颗粒剂、膏剂、乳剂、悬浮剂、栓剂、注射剂、吸入剂、凝胶剂、微球及气溶胶等。
给予本申请化合物或其药学上可接受的盐或其药物组合物的典型途径包括但不限于口服、直肠、局部、吸入、肠胃外、舌下、阴道内、鼻内、眼内、腹膜内、肌内、皮下、静脉内给药。
本申请的药物组合物可以采用本领域众所周知的方法制造,如常规的混合法、溶解法、制粒法、制糖衣药丸法、磨细法、乳化法、冷冻干燥法等。
在一些实施方案中,药物组合物是口服形式。对于口服给药,可以通过将活性化合物与本领域熟知的药学上可接受的辅料混合,来配制该药物组合物。这些辅料能使本申请的化合物被配制成片剂、丸剂、锭剂、糖衣剂、胶囊剂、液体、凝胶剂、浆剂、悬浮剂等,用于对患者的口服给药。
可以通过常规的混合、填充或压片方法来制备固体口服组合物。例如,可通过下述方法获得:将所述的活性化合物与固体辅料混合,任选地碾磨所得的混合物,如果需要则加入其它合适的辅料,然后将该混合物加工成颗粒,得到了片剂或糖衣剂的核心。适合的辅料包括但不限于:粘合剂、稀释剂、崩解剂、润滑剂、助流剂、甜味剂或矫味剂等。
药物组合物还可适用于肠胃外给药,如合适的单位剂型的无菌溶液剂、混悬剂或冻干产品。
本文所述的通式Ⅰ化合物的所有施用方法中,每天给药的剂量为0.01到100mg/kg体重,优选为0.05到50mg/kg体重,更优选0.1到30mg/kg体重,以单独或分开剂量的形式。
本发明具体实施方式的化学反应是在合适的溶剂中完成的,所述的溶剂须适合于本发明的化学变化及其所需的试剂和物料。为了获得本发明的化合物,有时需要本领域技术人员在已有实施方式的基础上对合成步骤或者反应流程进行修改或选择。
具体实施方式
下面通过实施例对发明进行详细描述,但并不意味着对本发明任何不利限制。本文已经详细地描述了本发明,其中也公开了其具体实施例方式,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改变和改进将是显而易见的。本申请所使用的所有试剂是市售的,无需进一步纯化即可使用。
化合物的结构是通过核磁共振(NMR)和/或质谱(MS)来确定的。NMR位移的单位为10 -6(ppm)。NMR测定的溶剂为氘代二甲基亚砜、氘代氯仿、氘代甲醇等,内标为四甲基硅烷(TMS);“IC 50”指半数抑制浓度,指达到最大抑制效果一半时的浓度。
实施例1:((1S,3R)-1-(5-((1-(3-氟丙基)吖丁啶-3-基)氨基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸的合成(化合物1)
Figure PCTCN2021088291-appb-000040
步骤1:(R)-叔-丁基(1-(3-(苄氧基)苯基)丙烷-2-基)氨基甲酯的合成
Figure PCTCN2021088291-appb-000041
将1-(苄氧基)-3-溴苯(6.7g,25.46mmol)溶于四氢呋喃(100mL)中,在-70℃下加入正丁基锂(2.5M,12.22mL),反应液于-70℃搅拌反应2小时。随后加入溶在四氢呋喃(30mL)中的(R)-叔丁基4-甲基-1,2,3-氧杂噻唑烷-3-甲酸基酯2,2-二氧化物(6.04g,25.46mmol),反应液于-70℃缓慢升温到25℃,搅拌反应13小时。将反应液倒入水中并搅拌10分钟,乙酸乙酯(50mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/乙酸乙酯=10/1)得到产物(R)-叔-丁基(1-(3-(苄氧基)苯基)丙烷-2-基)氨基甲酯(1.67g)。
MS m/z(ESI):364.2[M+Na] +
1H NMR(400MHz,CHLOROFORM-d)δ7.50-7.41(m,5H),7.35-7.21(m,1H),6.85-6.82(m,3H),5.07(s,2H),4.41(br s,1H),3.93(br s,1H),2.88-2.83(m,1H),2.67-2.62(m,1H),1.46(s,9H),1.10-1.09(d,J=6.5Hz,3H).
步骤2:(R)-1-(3-(苄氧基)苯基)丙烷-2-胺的合成
Figure PCTCN2021088291-appb-000042
将(R)-叔-丁基(1-(3-(苄氧基)苯基)丙烷-2-基)氨基甲酯(1.67g,4.89mmol)溶于1,4-二氧六环(10mL)中,加入HCl/1,4二氧六环(4M,18.34mL),反应液于25℃搅拌反应5小时。将反应液浓缩,用饱和碳酸氢钠中和至pH=8,乙酸乙酯(50mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干,得到产物(R)-1-(3-(苄氧基)苯基)丙烷-2-胺(1.16g)。
MS m/z(ESI):242.2[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ7.52-7.32(m,5H),7.24-7.20(t,1H),6.87-6.81(m,3H),5.10-5.05(d,J=5.6Hz,2H),3.22-3.17(m,1H),2.74-2.70(m,1H),2.54-2.49(m,1H),1.19-1.06(m,3H).
步骤3:(R)-1-(3-(苄氧基)苯基)-N-(2,2,2-三氟乙基)丙烷-2-胺的合成
Figure PCTCN2021088291-appb-000043
将(R)-1-(3-(苄氧基)苯基)丙烷-2-胺(1.16g,4.81mmol)和2,2,2-三氟乙基三氟甲磺酸酯(1.12g,4.81mmol)溶于1,4-二氧六环(20mL)中,加入N,N-二异丙基乙胺(1.86g,14.42mmol),反应液80℃搅拌反应13小时。将反应液倒入水中并搅拌10分钟,乙酸乙酯(20mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,二氯甲烷/甲醇=20/1)得到产物(R)-1-(3-(苄氧基)苯基)-N-(2,2,2-三氟乙基)丙烷-2-胺(1.36g)。
MS m/z(ESI):324.2[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ7.51-7.32(m,5H),7.24-7.22(m,1H),6.90-6.77(m,3H),5.08(s,2H),3.20-3.13(m,2H),3.09-2.98(m,1H),2.77-2.57(m,2H),1.10-1.08(d,J=6.2Hz,3H).
步骤4:(R)-3-(2-((2,2,2-三氟乙基)氨基)丙基)苯酚的合成
Figure PCTCN2021088291-appb-000044
将(R)-1-(3-(苄氧基)苯基)-N-(2,2,2-三氟乙基)丙烷-2-胺(1.36g,4.21mmol)溶于乙酸乙酯(50mL)中,加入10%钯炭(0.4g),反应液在50℃和氢气(15Psi)下搅拌反应13小时。反应液过滤,滤液减压浓缩至干得到产物(R)-3-(2-((2,2,2-三氟乙基)氨基)丙基)苯酚(1.0g)。
MS m/z(ESI):234[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ7.22-7.12(m,1H),6.86-6.59(m,3H),3.22-3.15(m,2H),3.12-2.96(m,1H),2.75-2.53(m,2H),1.11-1.10(d,J=6.3Hz,3H)
步骤5:(1S,3R)-1-(5-溴吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000045
将(R)-3-(2-((2,2,2-三氟乙基)氨基)丙基)苯酚(1.0g,4.07mmol)和5-溴-2-吡啶甲醛(984.94mg,5.30mmol)溶于二氯乙烷(90mL)中,加入三氟乙酸(1.39g,12.22mmol),反应液25℃搅拌反应16小时。将反应液倒入饱和碳酸氢钠(100mL)并搅拌10分钟,二氯甲烷(40mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/乙酸乙酯=4/1)得到产物(1S,3R)-1-(5-溴吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚(1.44g)。
MS m/z(ESI):401.1,403.1[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ8.62-8.53(m,1H),7.83-7.69(m,1H),7.38-7.33(m,1H),6.77-6.73(m,1H),6.63-6.51(m,2H),4.94(s,1H),3.53-3.37(m,1H),3.32-3.19(m,1H),3.12-3.09(dd,J=4.6,16.4Hz,1H),2.95(m,1H),2.60-2.54(dd,J=5.8,16.3Hz,1H),1.10-1.08(m,J=6.4,3H)
步骤6:(1S,3R)-1-(5-((1-(3-氟丙基)吖丁啶-3-基)氨基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000046
将(1S,3R)-1-(5-溴吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚(1.24g,3.09mmol) 和1-(3-氟丙基)吖丁啶-3-胺(828.27mg,3.71mmol,2.5当量盐酸盐)溶于1,4-二氧六环(20mL)中,加入甲磺酸(2-二环己基膦基-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)(280.16mg,309.06μmol)和叔丁醇钠(2.97g,30.91mmol),反应液80℃搅拌反应2小时。将反应液冷却至室温并倒入水(40mL)中并搅拌10分钟,乙酸乙酯(40mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,二氯甲烷/甲醇=20/1)得到产物(1S,3R)-1-(5-((1-(3-氟丙基)吖丁啶-3-基)氨基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚(0.76g)。
MS m/z(ESI):453.1[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ7.90-7.80(m,1H),7.28-7.16(m,1H),6.82-6.79(m,1H),6.73-6.63(m,1H),6.57-6.43(m,2H),4.86(s,1H),4.57(t,J=5.9Hz,1H),4.45(t,J=5.9Hz,1H),4.17-4.06(m,2H),3.79-3.71(m,2H),3.51-3.42(m,1H),3.28-3.06(m,2H),3.03-2.87(m,3H),2.68-2.59(m,2H),1.83-1.71(m,2H),1.29-1.05(m,3H).
步骤7:(1S,3R)-1-(5-((1-(3-氟丙基)吖丁啶-3-基)氨基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000047
将(1S,3R)-1-(5-((1-(3-氟丙基)吖丁啶-3-基)氨基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚(580mg,1.28mmol)和1,1,1-三氟-N-苯基-N-((三氟甲基)磺酰)甲磺酰胺(915.85mg,2.56mmol)溶于二氯甲烷(30mL)中,加入三乙胺(389.11mg,3.85mmol,535.23μL),反应液25℃搅拌反应13小时。将反应液冷却至室温并倒入水(40mL)中并搅拌10分钟,乙酸乙酯(40mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干,然后柱层析纯化(二氧化硅,二氯甲烷/甲醇=20/1)得到产物(1S,3R)-1-(5-((1-(3-氟丙基)吖丁啶-3-基)氨基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(0.65g)。
MS m/z(ESI):585.1[M+H] +
步骤8:((1S,3R)-1-(5-((1-(3-氟丙基)吖丁啶-3-基)氨基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000048
将(1S,3R)-1-(5-((1-(3-氟丙基)吖丁啶-3-基)氨基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(150mg,256.61μmol)和5,5,5',5'-四甲基-2,2'-联(1,3,2-二噁硼己环)(115.93mg,513.22μmol)溶于1,4-二氧六环(5mL)中,加入[1,1-双(二苯基膦)二茂铁]二氯化钯二氯甲烷(31.43mg,38.49μmol)和乙酸钾(75.55mg,769.82μmol)。反应液在氮气保护下80℃搅拌反应2小时。将反应液冷却至室温并倒入水(10mL)中并搅拌10分钟,乙酸乙酯(10mL)萃取2次,有机 相用无水硫酸钠干燥,过滤,减压浓缩至干。经制备液相色谱纯化(PhenomenexGemini-NX柱:3um二氧化硅,30mm直径,75mm长度;使用水(含有0.225%乙酸)和乙腈的极性递减的混合物作为洗脱液)纯化得到化合物((1S,3R)-1-(5-((1-(3-氟丙基)吖丁啶-3-基)氨基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸(8.67mg)。
MS m/z(ESI):481.3[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.86-7.74(m,1H),7.68-7.25(m,2H),7.14(d,J=8.7Hz,1H),6.98(dd,J=2.8,8.6Hz,1H),6.71(d,J=7.7Hz,1H),5.05-4.94(m,1H),4.59(t,J=5.6Hz,1H),4.48(t,J=5.6Hz,1H),4.38-4.11(m,3H),3.64-3.45(m,3H),3.41-3.35(m,1H),3.27-3.20(m,1H),3.15-3.02(m,2H),3.01-2.85(m,1H),2.68(dd,J=4.5,16.0Hz,1H),1.99-1.77(m,2H),1.08(d,J=6.5Hz,3H)。
实施例2:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸的合成(化合物2)
Figure PCTCN2021088291-appb-000049
步骤1:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000050
将(R)-3-(2-((2,2,2-三氟乙基)氨基)丙基)苯酚(160mg,686.02μmol)溶于甲苯(4mL),加入2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯甲醛(205.46mg,754.62μmol)和乙酸(411.97mg,6.86mmol)。反应液于90℃搅拌反应16小时。LCMS检测反应完成后,将反应溶液冷却至室温后,减压浓缩干,经薄层层析纯化(二氧化硅,石油醚/四氢呋喃=1/1)得到(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚(150.00mg)。
MS m/z(ESI):488.1[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ6.55-6.44(m,3H),6.10-6.00(m,2H),5.06(s,1H),4.51(t,J=5.9Hz,1H),4.39(t,J=5.9Hz,1H),4.07-3.97(m,1H),3.80-3.74(m,2H),3.46(br dd,J=4.1,10.2Hz,1H),3.27-3.13(m,2H),3.02-2.84(m,3H),2.65-2.59(m,2H),2.54-2.45(m,1H),1.82-1.68(m,2H),1.03(d,J=6.5Hz,3H)
步骤2:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4- 四氢异喹啉-6-基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000051
将(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚(150mg,307.70μmol)溶于二氯甲烷(2mL),加入N-苯基双(三氟甲烷磺酰)亚胺(164.89mg,461.56μmol)和三乙胺(93.41mg,923.11μmol)。反应液于25℃搅拌反应16小时。LCMS检测反应完成后,将反应溶液冷却至室温后,减压浓缩干,用二氯甲烷3mL稀释后经薄层层析纯化(二氧化硅,石油醚/四氢呋喃=1/1)得到(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(120.00mg)。
MS m/z(ESI):620.1[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.14(d,J=1.5Hz,1H),7.13-7.10(m,1H),6.90(d,J=8.8Hz,1H),6.15-6.06(m,2H),5.20(s,1H),4.53(t,J=5.8Hz,1H),4.41(t,J=5.8Hz,1H),4.16-4.07(m,1H),3.98-3.91(m,2H),3.58-3.48(m,1H),3.28-3.16(m,4H),2.96-2.85(m,1H),2.81(t,J=7.5Hz,2H),2.68(dd,J=3.9,16.2Hz,1H),1.87-1.73(m,2H),1.06(d,J=6.5Hz,3H)。
步骤5:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸的合成。
Figure PCTCN2021088291-appb-000052
将(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸(60.00mg,96.85μmol)溶于二氧六环(1mL),加入5,5,5',5'-四甲基-2,2'-联(1,3,2-二噁硼己环)(43.75mg,193.69μmol),乙酸钾(28.51mg,290.54μmol),1,1-双(二苯基磷)二茂铁氯化钯(10.63mg,14.53μmol)在氮气环境下加入该体系中。反应液于80℃搅拌反应2小时。LCMS检测反应完成后,将反应溶液冷却至室温后,减压浓缩后经过制备液相色谱(Phenomenex Synergi C18柱,3um二氧化硅,30mm直径,75mm长度);(使用水(含有0.225%甲酸)和和乙腈的极性递减的混合物作为洗脱液),纯化得到((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸(11.57mg)。
MS m/z(ESI):516.1[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.48(br s,1H),7.39(br d,J=7.3Hz,1H),6.75-6.66(m,1H),6.19-6.10(m,2H),5.19(s,1H),4.60(t,J=5.5Hz,1H),4.49(t,J=5.5Hz,1H),4.32(br s,3H),3.69(br s,2H),3.58-3.49(m,1H),3.32-3.25(m,1H),3.18(br t,J=7.3Hz,3H),2.99-2.77(m,1H),2.64(dd,J=3.5,15.6Hz,1H),2.02-1.87(m,2H),1.06(d,J=6.3Hz,3H)
实施例3:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(((1-氟环丙基)甲基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000053
步骤1:(1-(3-氟丙基)氮杂环丁烷-3-基)氨基甲酸叔丁酯的合成。
Figure PCTCN2021088291-appb-000054
氮杂环丁烷-3-基氨基甲酸叔丁酯盐酸盐(50g,239.60mmol)溶于二氧六环(1L)中,加入氟碘丙烷(49.54g,263.55mmol)和碳酸钾(165.57g,1.20mol)。反应液在25℃下搅拌反应16小时。TLC(二氯甲烷:甲醇=10:1)监测反应完成后。反应液过滤,收集滤液减压浓缩,经过柱层析纯化(二氧化硅,二氯甲烷/甲醇=100/7)得到产物(1-(3-氟丙基)氮杂环丁烷-3-基)氨基甲酸叔丁酯(41.00g)。
MS m/z(ESI):233.2[M+H] +
步骤2:1-(3-氟丙基)氮杂环丁烷-3-胺盐酸盐的合成。
Figure PCTCN2021088291-appb-000055
将(1-(3-氟丙基)氮杂环丁烷-3-基)氨基甲酸叔丁酯(41.55g,178.87mmol,)溶于乙酸乙酯(200mL),慢慢滴加盐酸乙酸乙酯(447.17mL,4mol/L)。反应液在25℃下搅拌反应16小时。TLC(二氯甲烷:甲醇=10:1)监测反应完成后。反应液减压浓缩,得到粗产物1-(3-氟丙基)氮杂环丁烷-3-胺盐酸盐(40g)。
1H NMR(400MHz,DMSO-d 6)δ8.98(s,2H),4.68-4.55(m,1H),4.53-4.44(m,1H),4.30(br s,4H),4.13-3.99(m,1H),3.47-3.34(m,2H),2.02-1.86(m,2H)
步骤3:5-溴-2-(二乙氧基甲基)-1,3-二氟苯的合成
Figure PCTCN2021088291-appb-000056
将4-溴-2,6-二氟苯甲醛(40g,181.00mmol)和原甲酸三乙酯(134.12g,904.98mmol)溶于甲苯(200mL)中,加入一水合对甲苯磺酸(8.61g,45.25mmol),反应液于25℃搅拌16小时。将反应液减压浓缩至干,浓缩物经柱层析纯化(二氧化硅,石油醚/乙酸乙酯=100/1)得到产物5-溴-2-(二乙氧基甲基)-1,3-二氟苯(50g)。
1H NMR(400MHz,METHANOL-d 4)δ7.33-7.19(m,2H),3.78-3.75(m,2H),3.66-3.53(m,2H),1.26-1.19(m,6H)
步骤4:N-(4-(二乙氧基甲基)-3,5-二氟苯基)-1-(3-氟丙基)氮杂环丁烷-3-胺的合成
Figure PCTCN2021088291-appb-000057
将5-溴-2-(二乙氧基甲基)吡啶(27g,91.49mmol)和1-(3-氟丙基)氮杂环丁烷-3-胺盐酸盐(24.31g,100.64mmol)溶于四氢呋喃中(500mL),在氮气环境下加入叔丁醇钠(52.75g,548.93mmol)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)(4.15g,4.57mmol),反应液在80℃的氮气环境下搅拌4小时。将反应液浓缩至干,用水(300mL)和乙酸乙酯(300mL*3)萃取,将有机相合并,用饱和食盐水(200mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩至干。经柱层析纯化(二氧化硅,石油醚/四氢呋喃=1/1)得到产物N-(4-(二乙氧基甲基)-3,5-二氟苯基)-1-(3-氟丙基)氮杂环丁烷-3-胺(13g)。
1H NMR(400MHz,METHANOL-d 4)δ6.08(d,J=11.2Hz,2H),5.58(s,1H),4.55-4.50(t,1H),4.40(t,J=5.9Hz,1H),4.04(m,J=6.6Hz,1H),3.82-3.74(m,2H),3.71-3.63(m,2H),3.56-3.47(m,2H),3.03-2.91(m,2H),2.63(t,J=7.5Hz,2H),1.84-1.64(m,2H),1.19(m,J=7.0Hz,6H).
步骤5:2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯甲醛的合成
Figure PCTCN2021088291-appb-000058
将N-(4-(二乙氧基甲基)-3,5-二氟苯基)-1-(3-氟丙基)氮杂环丁烷-3-胺(13g,30.02mmol)溶于四氢呋喃中(100mL),加入盐酸溶液(2M,120.1mL),反应液于25℃搅拌2小时。反应完成后,反应液用饱和碳酸氢钠溶液调节PH到8~9,并用乙酸乙酯(100ml*3)萃取,将有机相合并,用无水硫酸钠干燥,过滤,减压浓缩至干得到产物N-(4-(二乙氧基甲基)-3,5-二氟苯基)-1-(3-氟丙基)氮杂环丁烷-3-胺(10g)。
MS m/z(ESI):506.2[M+H] +
步骤6:(R)-1-氟-N-(1-(3-甲氧苯基)丙烷-2-基)环丙甲酰胺的合成
Figure PCTCN2021088291-appb-000059
将(R)-1-(3-甲氧苯基)丙烷-2-胺(21g,114.38mmol)和1-氟环丙羧酸(14.29g,137.26mmol)溶于四氢呋喃(300mL)中,加入N,N-二异丙基乙胺(44.35g,343.15mmol)和O-(7-氮杂苯并三氮唑-1-基)-N,N,N,N-四甲基脲六氟膦盐(52.19g,137.26mmol),反应液于25℃搅拌16小时。反应完成后,将反应液倒入水(200mL)中,用乙酸乙酯(100mL)萃取三次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。浓缩物经柱层析纯化(二氧化硅,石油醚/乙酸乙酯=4/1)得到产物(R)-1-氟-N-(1-(3-甲氧苯基)丙烷-2-基)环丙甲酰胺(25.5g)。
1H NMR(400MHz,METHANOL-d 4)δ7.17(t,J=8.0Hz,1H),6.83-6.71(m,3H),4.21(m,J=6.8Hz,1H),3.81-3.72(m,3H),2.87-2.78(m,1H),2.75-2.66(m,1H),1.28-1.13(m,7H).
步骤7:(R)-1-氟-N-(1-(3-羟基苯基)丙烷-2-基)环丙甲酰胺的合成
Figure PCTCN2021088291-appb-000060
将(R)-1-氟-N-(1-(3-甲氧苯基)丙烷-2-基)环丙甲酰胺(25.5g,101.47mmol)溶于二氯甲烷(200mL)中,该溶液冷却到0℃后,滴加入三溴化硼(50.84g,202.95mmol),反应液于25℃下搅拌1小时。反应完成后,将反应液慢慢滴加入冰水中淬灭,用饱和碳酸钠溶液将溶液pH调节到8。然后用二氯甲烷(200mL)萃取两次,收集到的有机相用无水硫酸钠干燥过滤,减压浓缩后得到粗产物(R)-1-氟-N-(1-(3-羟基苯基)丙烷-2-基)环丙甲酰胺(23g)。
1H NMR(400MHz,METHANOL-d 4)δ8.06(br d,J=7.5Hz,1H),7.08(t,J=7.8Hz,1H),6.73-6.58(m,3H),4.20(m,J=7.1Hz,1H),2.80(dd,J=7.0,13.3Hz,1H),2.65(dd,J=7.3,13.3Hz,1H),1.30-1.13(m,7H).
步骤8:(R)-3-(2-(((1-氟环丙基)甲基)氨基)丙基)苯酚的合成
Figure PCTCN2021088291-appb-000061
将(R)-1-氟-N-(1-(3-羟基苯基)丙烷-2-基)环丙甲酰胺(23g,96.94mmol)溶于四氢呋喃(200mL)中,在0℃下缓慢滴加硼烷四氢呋喃溶液(1M,484.68mL),反应液在65℃搅拌6小时。LCMS检测反应完成。将反应液冷却到0℃,加入甲醇淬灭,将溶液减压浓缩至干并溶于甲醇(100mL)中,在65℃搅拌2小时。将反应液减压浓缩至干。浓缩物经柱层析纯化(二氧化硅,石油醚/四氢呋喃=1/1)得到产物(R)-3-(2-(((1-氟环丙基)甲基)氨基)丙基)苯酚(15.5g)。
1H NMR(400MHz,METHANOL-d 4)δ7.16-7.07(m,1H),6.73-6.62(m,3H),3.09-2.99(m,1H),2.99-2.84(m,2H),2.75-2.66(m,1H),2.55(dd,J=7.0,13.3Hz,1H),1.06(d,J=6.0Hz,3H),1.03-0.94(m,2H),0.69-0.56(m,2H).
步骤9:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(((1-氟环丙基)甲基)-3-甲基1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000062
将(R)-3-(2-(((1-氟环丙基)甲基)氨基)丙基)苯酚(7.22g,32.32mmol)和2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯甲醛(10g,29.38mmol)溶于乙醇(150mL)中,加入二苯基磷酸(9.62g,44.07mmol),反应液在70℃搅拌16小时。反应完成后,将反应液减压浓缩至干,浓缩物溶于乙酸乙酯(100mL)和水(100mL)中,用饱和碳酸钠水溶液调节PH至8~9,然后用乙酸乙酯(100mL)萃取三次。合并的有机相用无水硫酸钠干燥,过滤,减压浓缩至干。浓缩物经柱层析纯化(二氧化硅,石油醚/四氢呋喃=1/1)得到产物(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(((1-氟环丙基)甲基)-3-甲基1,2,3,4-四氢异喹啉-6-酚(9g)。
MS m/z(ESI):478.2[M+H] +
步骤10:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-((1-氟环丙基)甲基)-3-1,2,3,4-四氢异喹啉-6-甲基三氟甲磺酸甲酯的合成
Figure PCTCN2021088291-appb-000063
将(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(((1-氟环丙基)甲基)-3-甲基1,2,3,4-四氢异喹啉-6-酚(8.9g,10.06mmol)溶于二氯甲烷(100mL),加入1,1,1-三氟-N-苯基-N-((三氟甲基)磺酰)甲磺酰胺(7.19g,20.13mmol)和三乙胺(3.06g,30.19mmol)。反应液于25℃搅拌反应16个小时。反应完成后,将反应溶液减压浓缩干,经薄层层析纯化(二氧化硅,石油醚/乙酸乙酯=1/1)得到产物(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-((1-氟环丙基)甲基)-3-1,2,3,4-四氢异喹啉-6-甲基三氟甲磺酸甲酯(3.2g)。
MS m/z(ESI):610.1[M+H] +
步骤11:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(((1-氟环丙基)甲基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000064
将(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-((1-氟环丙基)甲基)-3-1,2,3,4-四氢异喹啉-6-甲基三氟甲磺酸甲酯(2.8g,4.59mmol)和5,5,5',5'-四甲基-2,2'-联(1,3,2-二噁硼己环)(1.56g,6.89mmol)溶于二氧六环(50mL),加入乙酸钾(901.57mg,9.19mmol),1,1-双(二苯 基磷)二茂铁氯化钯(336.09mg,459.32μmol)在氮气环境下加入该反应体系中。反应液于90℃搅拌反应2个小时。反应完成后,将反应溶液冷却至室温后,减压浓缩后经薄层层析纯化(二氧化硅,石油醚/四氢呋喃=1/1),并经手性制备纯化(REGIS(s,s)WHELK-O1柱:10um二氧化硅,50mm直径,250mm长度;使用45%乙醇(含有0.1%氨水)作为洗脱液)得到产物((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(((1-氟环丙基)甲基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸(341.16mg)。
MS m/z(ESI):506.1[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.45(s,1H),7.36(br d,J=7.8Hz,1H),6.69(d,J=7.8Hz,1H),6.06(d,J=11.5Hz,2H),5.07(s,1H),4.52(t,J=5.8Hz,1H),4.41(t,J=5.9Hz,1H),4.05(m,J=6.7Hz,1H),3.86-3.78(m,2H),3.70(m,J=4.3,10.3Hz,1H),3.30-3.22(m,1H),3.17-3.06(m,1H),3.05-2.96(m,1H),3.04-2.96(m,1H),2.73-2.55(m,4H),1.86-1.70(m,2H),1.01(d,J=6.4Hz,3H),0.92-0.82(m,2H),0.48-0.39(m,2H).
实施例4:((1S,3R)-2-(2,2-二氟-3-羟基丙基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000065
步骤1:2,2-二氟丙烷-1,3-二醇的合成
Figure PCTCN2021088291-appb-000066
将四氢锂铝(7.74g,203.93mmol)溶于四氢呋喃(10mL)中,在0℃下加入溶在四氢呋喃(10mL)中的二氟丙二酸二乙酯(5g,25.49mmol)溶液,反应液于25℃搅拌反应16小时。向反应液中滴加水(10mL),15%氢氧化钠水溶液(10mL)和水(30mL),无水硫酸钠干燥,过滤减压浓缩至干得到产物2,2-二氟丙烷-1,3-二醇(7.1g)。
1H NMR(400MHz,METHANOL-d 4)δ3.83-3.69(m,4H).
步骤2:3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙烷-1-醇的合成
Figure PCTCN2021088291-appb-000067
将2,2-二氟丙烷-1,3-二醇(7.7g,68.70mmol)溶于四氢呋喃(100mL)中,在0℃下加入钠氢(2.75g,68.70mmol),反应液于25℃搅拌反应1小时。在0℃下加入溶在四氢呋喃(30mL)中的叔丁基二苯基氯硅烷溶液,反应液于25℃搅拌反应15小时,将反应液倒入水中并搅拌10分钟,乙酸乙酯(50mL)萃取3次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/四氢呋喃=10/1)得到产物3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙烷-1-醇(15g)。
1H NMR(400MHz,METHANOL-d 4)δ7.70-7.67(m,4H),7.46-7.37(m,6H),3.87-3.80(m,4H),1.05(s,9H).
步骤3:3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000068
将3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙烷-1-醇(1.9g,5.42mmol)溶于二氯甲烷(20mL)中,加入2,6-二甲基吡啶(1.74g,16.26mmol)和三氟甲磺酸酐(3.06g,10.84mmol),反应液-10℃搅拌反应2小时。将反应液倒入水中并搅拌10分钟,乙酸乙酯(20mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干得到粗产物3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基三氟甲磺酸酯(3.0g)。
1H NMR(400MHz,METHANOL-d 4)δ7.70-7.67(m,4H),7.50-7.44(m,6H),4.98(t,J=12.2Hz,2H),3.93(t,J=12.2Hz,2H),1.08(s,1H).
步骤4:(R)-N-(1-(3-(苄氧基)苯基)丙烷-2-基)-3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙烷-1-胺的合成
Figure PCTCN2021088291-appb-000069
将(R)-1-(3-(苄氧基)苯基)丙烷-2-胺(1.25g,5.18mmol)溶于乙腈(10mL)中,加入碳酸钾(2.15g,15.54mmol)和3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基三氟甲磺酸酯(2.94g,5.18mmol)。反应液60℃搅拌反应16小时。将反应液过滤并减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/乙酸乙酯=20/1)得到产物(R)-N-(1-(3-(苄氧基)苯基)丙烷-2-基)-3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙烷-1-胺(1.0g)。
MS m/z(ESI):574.4[M+H] +
步骤5:(R)-3-(2-((3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基)氨基)丙基)苯酚的合成
Figure PCTCN2021088291-appb-000070
将(R)-N-(1-(3-(苄氧基)苯基)丙烷-2-基)-3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙烷-1-胺(0.2g,348.56μmol)溶于乙酸乙酯(10mL)中,加入10%氢氧化钯/钯炭(28.57mg),用氢气置换3次,反应液在氢气(15Psi)和25℃下搅拌反应16小时。将反应液过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/乙酸乙酯=3/1)得到产物(R)-3-(2-((3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基)氨基)丙基)苯酚(100mg)。
MS m/z(ESI):484.2[M+H] +
步骤6:(1S,3R)-2-(3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000071
将(R)-3-(2-((3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基)氨基)丙基)苯酚(100mg,206.76μmol)和2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯甲醛(67.55mg,248.11μmol)溶于乙酸(1mL)和甲苯(5mL)中,该反应液100℃搅拌反应32小时。将反应液减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/四氢呋喃=1/3)得到产物(1S,3R)-2-(3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(45mg)。
MS m/z(ESI):738.4[M+H] +
步骤7:(1S,3R)-2-(3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000072
将(1S,3R)-2-(3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(45mg,42.69μmol)和N-苯基双(三氟甲烷磺酰)亚胺(30.50mg,85.38μmol)溶于二氯甲烷(3mL)中,加入三乙胺(12.96mg,128.06μmol),反应液25℃搅拌反应13小时。将反应液倒入水中并搅拌10分钟,二氯甲烷(5mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/四氢呋喃=1/3)得到产物(1S,3R)-2-(3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(15mg)。
MS m/z(ESI):870.2[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ7.74-7.58(m,4H),7.47-7.36(m,6H),7.00(s,1H),6.97-6.90(m,1H),6.88-6.79(m,1H),5.85(d,J=11.0Hz,2H),5.12(s,1H),4.56(t,J=5.8Hz,1H),4.44(t,J=5.8Hz,1H),4.34(br d,J=6.0Hz,1H),4.04-3.83(m,2H),3.83-3.67(m,2H),3.65-3.41(m,2H),3.29-3.05(m,2H),2.99(br s,2H),2.86-2.71(m,1H),2.69-2.61(m,2H),2.54(dd,J=3.5,16.1Hz,1H),1.86-1.67(m,2H),1.07-1.00(m,12H).
步骤8:((1S,3R)-2-(3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000073
将(1S,3R)-2-(3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(15mg,17.24μmol)和联硼酸新戊二醇酯(7.79mg,34.48μmol)溶于二甲基亚砜(1mL)中,加入乙酸钾(5.08mg,51.72μmol)和[1,1-双(二苯基膦)二茂铁]二氯化钯二氯甲烷(2.11mg,2.59μmol),反应液80℃搅拌反应1小时。将反应液倒入水中并搅拌10分钟,乙酸乙酯(10mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干得到粗产物((1S,3R)-2-(3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸(20mg)。
MS m/z(ESI):766.4[M+H] +
步骤9:((1S,3R)-2-(2,2-二氟-3-羟基丙基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000074
将((1S,3R)-2-(3-((叔-丁基二苯基甲硅烷基)氧基)-2,2-二氟丙基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸(20mg,10.45μmol)溶于四氢呋喃(1mL)中,加入四丁基氟化铵四氢呋喃溶液(1M,20.90μmol),反应液25℃搅拌反应2小时。将反应液倒入水中并搅拌10分钟,乙酸乙酯(5mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后薄层层析纯化(二氧化硅,二氯甲烷/甲醇=10/1)得到产物((1S,3R)-2-(2,2-二氟-3-羟基丙基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸(1.52mg)。
MS m/z(ESI):528.2[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.67-6.63(m,2H),6.55-6.47(m,1H),6.16-6.05(m,2H),5.42-5.33(m,1H)5.09(br s,1H),4.56(t,J=5.8Hz,1H),4.45(t,J=5.8Hz,1H),4.15(m,1H),3.99(br s,2H),3.84-3.51(m,4H),3.27-2.99(m,4H),2.91-2.81(m,2H),2.76-2.45(m,2H),1.91-1.79(m,2H),1.04(d,J=6.5Hz,3H).
实施例5:((1S,3R)-2-(2-氟-2-甲基丙基)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3-甲基1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000075
步骤1:2-氟-2-甲基丙基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000076
将2-氟-2-甲基-1-丙醇(1g,10.86mmol)和2,6-二甲基吡啶(1.40g,13.03mmol,1.52mL)溶于二氯甲烷(25mL)中,反应瓶置于-10℃的冰盐浴中,向反应液中滴加三氟甲磺酸酐(3.37g,11.94mmol,1.97mL),反应液为橙色透明溶液。反应液于25℃搅拌反应2小时。TLC检测原料反应完全后,将反应液依次用水(5mL),1mol/L的盐酸水溶液(5mL)和饱和碳酸氢钠溶液(5mL)洗涤,收集的有机相用硫酸钠干燥后,过滤,有机相减压浓缩干得到化合物2-氟-2-甲基丙基三氟甲磺酸酯(3.60g)。
1H NMR(400MHz,CHLOROFORM-d)δ4.41(d,J=18.6Hz,2H),1.48(s,3H),1.43(s,3H).
步骤2:(R)-N-(1-(3-(苄氧基)苯基)丙-2-基)-2-氟-2-甲基丙-1-胺的合成
Figure PCTCN2021088291-appb-000077
将(R)-1-(3-(苄氧基)苯基)丙烷-2-胺(500mg,1.76mmol,85%纯度)和2-氟-2-甲基丙基三氟甲磺酸酯(723.78mg,1.94mmol,60%纯度)溶于二氧六环(15mL)中,加入N,N-二异丙基乙胺(455.22mg,3.53mmol)。反应液于80℃搅拌反应16个小时。LCMS检测反应完成后,将反应溶液冷却至室温后,减压浓缩干,经过柱层析纯化(二氧化硅,石油醚/乙酸乙酯=100/12)得到化合物(R)-N-(1-(3-(苄氧基)苯基)丙-2-基)-2-氟-2-甲基丙-1-胺(410.00mg)。
MS m/z(ESI):316.1[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ7.46-7.30(m,5H),7.23-7.18(m,1H),6.85-6.78(m,3H),5.06(s,2H),2.94-2.85(m,1H),2.79-2.62(m,3H),2.56(dd,J=6.8,13.3Hz,1H),1.37(s,3H),1.32(s,3H),1.04(d,J=6.0Hz,3H).
步骤3:(R)-3-(2-((2-氟-2-甲基丙基)氨基)丙基)苯酚的合成
Figure PCTCN2021088291-appb-000078
将(R)-N-(1-(3-(苄氧基)苯基)丙-2-基)-2-氟-2-甲基丙-1-胺(540mg,1.71mmol)溶于乙酸乙酯(20mL)中,在氮气环境下加入钯碳(100mg,10%纯度),反应液于50℃的氢气(15Psi)环境下搅拌16小时。LCMS显示反应完成。将反应液过滤浓缩,浓缩物经柱层析纯化(二氧化硅,石油醚/乙酸乙酯=0~20%)得到产物(R)-3-(2-((2-氟-2-甲基丙基)氨基)丙基)苯酚(350mg)。
1H NMR(400MHz,CHLOROFORM-d)δ7.16(t,J=8.2Hz,1H),6.79-6.65(m,3H),2.94(m,J=6.5Hz,1H),2.80-2.56(m,4H),1.37(s,3H),1.32(s,3H),1.09(d,J=6.0Hz,3H).
步骤4:(1S,3R)-1-(5-溴吡啶-2-基)-2-(2-氟-2-甲基丙基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000079
将(R)-3-(2-((2-氟-2-甲基丙基)氨基)丙基)苯酚(50mg,221.92ummol)溶于二氯乙烷(1.5mL)中,加入5-溴-吡啶-2-甲醛(45.41mg,244.12ummol)和三氟乙酸(75.91mg,665.77μmol,49.29uL)。反应液于25℃搅拌反应16个小时。LCMS检测反应未完成,反应液升温至50℃搅拌反应3小时,TLC(二氧化硅,石油醚/乙酸乙酯=3/1)监测反应完毕。反应液冷却至室温后,向反应液中滴加饱和碳酸钠溶液,将其pH调节至弱碱性。再将反应液用乙酸乙酯(10mL)稀释后,用水溶液(30mL)洗涤三次,收集水相和有机相。水相用乙酸乙酯(20mL)萃取三次后,将所有有机相合并,用硫酸钠干燥后,过滤,有机相减压浓缩干。经薄层层析纯化(二氧化硅,石油醚/乙酸乙酯=3/1)得到化合物(1S,3R)-1-(5-溴吡啶-2-基)-2-(2-氟-2-甲基丙基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(36.00mg)。
1H NMR(400MHz,CHLOROFORM-d)δ8.54(d,J=2.0Hz,1H),7.70(dd,J=2.4,8.4Hz,1H),7.34(d,J=8.6Hz,1H),6.73(d,J=8.4Hz,1H),6.58(s,1H),6.56-6.52(m,1H),4.88(s,1H),3.63-3.46(m,1H),3.04(dd,J=4.4,15.9Hz,1H),2.91-2.74(m,1H),2.54(dd,J=6.0,16.1Hz,1H),2.44-2.28(m,1H),1.31(s,3H),1.20(s,3H),1.01(d,J=6.5Hz,3H).
步骤5:(1S,3R)-2-(2-氟-2-甲基丙基)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000080
将(1S,3R)-1-(5-溴吡啶-2-基)-2-(2-氟-2-甲基丙基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(36mg,91.54μmol)和1-(3-氟丙基)氮杂环丁烷-3-胺(14.52mg,6.10μmol,3HCl)溶于二氧六环(2mL)中,在氮气环境下加入叔丁醇钠(43.98mg,457.68μmol)和BrettPhos Pd G3(80.30mg,9.15μmol)。反应液于80℃搅拌反应16个小时。LCMS检测反应完成后,将反应溶液冷却至室温后,减压浓缩干,用二氯甲烷将浓缩液溶解,之后过滤,得到滤液。经薄层层析纯化(二氧化硅,石油醚/四氢呋喃=1/2,1%NH 3.H 2O)得到化合物(1S,3R)-2-(2-氟-2-甲基丙基)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(23.00mg)。
MS m/z(ESI):443.2[M-H] +
1H NMR(400MHz,CHLOROFORM-d)δ7.78(d,J=2.8Hz,1H),7.18(d,J=8.5Hz,1H),6.76(dd,J=2.8,8.5Hz,1H),6.63(d,J=8.3Hz,1H),6.53(d,J=2.3Hz,1H),6.42(dd,J=2.5,8.3Hz,1H),4.79(s,1H),4.54(t,J=5.9Hz,1H),4.42(t,J=5.9Hz,1H),4.14-4.00(m,2H),3.77-3.71(m,2H),3.58-3.49(m,1H),3.08-2.92(m,3H),2.77(dd,J=14.8,19.6Hz,1H),2.63(t,J=7.2Hz,2H),2.48(dd,J=5.5,16.3Hz,1H),1.82-1.72(m,2H),1.24(d,J=6.8Hz,3H),1.19(d,J=12.0Hz,3H),0.98(d,J=6.5Hz,3H)
步骤6:(1S,3R)-2-(2-氟-2-甲基丙基)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000081
将(1S,3R)-2-(2-氟-2-甲基丙基)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(17mg,38.24μmol)和N-苯基双(三氟甲烷磺酰)亚胺(20.49mg,57.36μmol)溶于无水二氯甲烷(1mL)中,加入三乙胺(11.61mg,114.72μmol)。反应液于25℃搅拌反应13个小时。LCMS检测反应完成后,将反应溶液冷却至室温后,减压浓缩干。经薄层层析纯化(二氧化硅,石油醚/四氢呋喃=1/1,1%NH 3.H 2O)得到化合物(1S,3R)-2-(2-氟-2-甲基丙基)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(22.00mg)。
MS m/z(ESI):577.1[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ7.80(d,J=2.8Hz,1H),7.38-7.32(m,1H),7.23(s,1H),7.17(d,J=8.5Hz,1H),7.01(s,1H),6.79(dd,J=2.8,8.5Hz,1H),4.86(s,1H),4.53(t,J=5.9Hz,1H),4.41(t,J=5.8Hz,1H),4.17(s,2H),3.81(s,2H),3.66-3.57(m,1H),3.16-3.02(m,3H),2.85-2.54(m,4H),1.84-1.69(m,2H),1.29-1.26(m,3H),1.21(d,J=12.5Hz,3H),0.99(d,J=6.5Hz,3H).
步骤7:((1S,3R)-2-(2-氟-2-甲基丙基)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3-甲基1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000082
将(1S,3R)-2-(2-氟-2-甲基丙基)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(20mg,34.68μmol)和联硼酸新戊二醇酯(11.75mg,52.03μmol)溶于1,4-二氧六环(3mL)中,加入乙酸钾(6.81mg,69.37μmol)和1,1-双(二苯基磷)二茂铁氯化钯(2.54mg,3.47μmol),反应液用N 2置换三次,升温到80℃并搅拌反应16个小时。LCMS检测反应完成后,将反应溶液冷却至室温后,将反应液用水(5mL)稀释后,用乙酸乙酯(15mL)萃取三次,将所有有机相合并,用硫酸钠干燥后,有机相减压浓缩干。经薄层层析纯化(二氧化硅,二氯甲烷/甲醇=10/1)得到化合物((1S,3R)-2-(2-氟-2-甲基丙基)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3-甲基1,2,3,4-四氢异喹啉-6-基)硼酸(2.19mg)。
MS m/z(ESI):473.2[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ8.01(s,1H),7.69(s,1H),7.61(d,J=8.3Hz,1H),7.23(d,J=8.6Hz,1H),7.13(s,1H),7.03(d,J=7.4Hz,1H),5.63(s,1H),4.62(t,J=5.4Hz,1H),4.50(t,J=5.4Hz,1H),4.21-3.90(m,4H),3.59-3.40(m,3H),3.23(s,1H),3.10-2.90(m,4H),2.07-1.95(m,2H),1.53(d,J=5.1Hz,3H),1.48(d,J=5.3Hz,3H),1.34(d,J=6.3Hz,3H).
实施例6:(1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢苯并呋喃[2,3-c]吡啶-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000083
步骤1:1-(5-溴苯并呋喃-3-基)丙烷-2-酮的合成
Figure PCTCN2021088291-appb-000084
将5-溴苯并呋喃-3(2H)-酮(1g,4.69mmol)和1-(三苯基正膦亚基)丙烷-2-酮(2.24g,7.04mmol)溶于二甲苯(20mL)中,反应液于140℃搅拌22小时。将反应液减压浓缩至干,浓缩物经柱层析纯化(二氧化硅,石油醚/乙酸乙酯=10/1)得到产物1-(5-溴苯并呋喃-3-基)丙烷-2-酮(800mg)。
1H NMR(400MHz,CHLOROFORM-d)δ7.63(s,1H),7.59(d,J=1.8Hz,1H),7.42-7.37(m,1H),7.37-7.33(m,1H),3.75(s,2H),3.70(s,1H),2.25(s,3H).
步骤2:1-(5-溴苯并呋喃-3-基)丙烷-2-胺的合成
Figure PCTCN2021088291-appb-000085
将1-(5-溴苯并呋喃-3-基)丙烷-2-酮(800mg,3.16mmol)溶于甲醇(30mL)中,然后加入乙酸铵(2.44g,31.61mmol),乙酸钠(259.30mg,3.16mmol),氰基硼氢化钠(297.96mg,4.74mmol)和醋酸(94.91mg,1.58mmol),反应液于25℃搅拌16小时。将反应液浓缩至干,经柱层析纯化(二氧化硅,二氯甲烷/甲醇=10/1,1%氨水)得到产物1-(5-溴苯并呋喃-3-基)丙烷-2-胺(740mg)。
1H NMR(400MHz,CHLOROFORM-d)δ7.69(d,J=1.8Hz,1H),7.59(s,1H),7.42-7.31(m,2H),3.64-3.53(m,1H),3.06(dd,J=6.4,14.4Hz,1H),2.89(dd,J=7.5,14.6Hz,1H),1.36(d,J=6.4Hz,3H).
步骤3:1-(5-溴苯并呋喃-3-基)-N-(2,2,2-三氟乙基)丙烷-2-胺的合成
Figure PCTCN2021088291-appb-000086
将1-(5-溴苯并呋喃-3-基)丙烷-2-胺(400mg,1.57mmol)和三氟甲磺酸三氟乙酯(400.84mg,1.73mmol)溶于二氧六环(5mL)中,加入N,N-二异丙基乙胺(608.73mg,4.71mmol),反应液于80℃搅拌16小时。将反应液减压浓缩至干,经柱层析纯化(二氧化硅,石油醚/乙酸乙酯=10/1)得到产物1-(5-溴苯并呋喃-3-基)-N-(2,2,2-三氟乙基)丙烷-2-胺(150mg)。
1H NMR(400MHz,CHLOROFORM-d)δ7.67(d,J=1.8Hz,1H),7.50(s,1H),7.43-7.31(m,2H),3.29-3.09(m,3H),2.71(m,J=6.5,14.5Hz,2H),1.13(d,J=6.4Hz,3H).
步骤4:1-(5-(4,4,5,5-四甲基-1,3,2-二噁硼戊环-2-基)苯并呋喃-3-基)-N-(2,2,2-三氟乙基)丙烷-2-胺的合成
Figure PCTCN2021088291-appb-000087
将1-(5-溴苯并呋喃-3-基)-N-(2,2,2-三氟乙基)丙烷-2-胺(320mg,951.96μmol)和双联嚬哪醇硼酸酯(362.61mg,1.43mmol)溶于二氧六环(10mL)中,加入醋酸钾(186.86mg,1.9mmol),在氮气保护下加入1,1-双(二苯基磷)二茂铁氯化钯(69.66mg,95.20μmol),反应液于80℃搅拌16小时。将反应液浓缩至干,得到粗品产物1-(5-(4,4,5,5-四甲基-1,3,2-二噁硼戊环-2-基)苯并呋喃-3-基)-N-(2,2,2-三氟乙基)丙烷-2-胺(364.80mg)。
步骤5:3-(2-(((2,2,2-三氟乙基)氨基)丙基)苯并呋喃-5-醇的合成
Figure PCTCN2021088291-appb-000088
将1-(5-(4,4,5,5-四甲基-1,3,2-二噁硼戊环-2-基)苯并呋喃-3-基)-N-(2,2,2-三氟乙基)丙烷-2-胺(364mg,949.87μmol)溶于四氢呋喃(6mL)中,加入双氧水(73.85mg,759.89μmol,35%含量)和氢氧化钠溶液(4M,118.73μL),反应液于40℃搅拌16小时。将反应液冷却到室温,加入饱和硫代硫酸钠溶液(5mL)搅拌0.5小时,反应液用乙酸乙酯(15mL)萃取三次,将有机相减压浓缩至干,经薄层色谱纯化(二氧化硅,石油醚/乙酸乙酯=3/1)得到产物3-(2-(((2,2,2-三氟乙基)氨基)丙基)苯并呋喃-5-醇(150mg)。
1H NMR(400MHz,CHLOROFORM-d)δ7.46(s,1H),7.32(d,J=8.8Hz,1H),6.95(d,J=2.5Hz,1H),6.81(dd,J=2.5,8.8Hz,1H),3.27-3.10(m,3H),2.78-2.62(m,2H),1.13(d,J=6.0Hz,3H).
步骤6:1-(4-溴-2,6-二氟苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢苯并呋喃[2,3-c]吡啶-6-醇的合成
Figure PCTCN2021088291-appb-000089
将3-(2-(((2,2,2-三氟乙基)氨基)丙基)苯并呋喃-5-醇(150mg,548.95μmol)和4-溴-2,6-二氟苯甲醛(133.45mg,603.84μmol)溶于二氧六环(5mL)中,加入三氟醋酸(187.78mg,1.65mmol),反应液于90℃搅拌16小时。将反应液减压浓缩至干,经薄层色谱纯化(二氧化硅,石油醚/四氢呋喃=3/1)得到产物1-(4-溴-2,6-二氟苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢苯并呋喃[2,3-c]吡啶-6-醇(180mg)。
1H NMR(400MHz,CHLOROFORM-d)δ7.20(d,J=8.8Hz,1H),7.07(d,J=8.0Hz,2H),6.88(d,J=2.5Hz,1H),6.74(dd,J=2.6,8.7Hz,1H),5.23(s,1H),4.70(br s,1H),3.56-3.46(m,1H),3.30-3.16(m,1H),3.00-2.86(m,2H),2.51(dd,J=1.5,5.5,15.8Hz,1H),1.19(d,J=6.8Hz,3H).
步骤7:1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢苯并呋喃[2,3-c]吡啶-6-醇的合成
Figure PCTCN2021088291-appb-000090
将1-(4-溴-2,6-二氟苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢苯并呋喃[2,3-c]吡啶-6-醇(160mg,335.97μmol)和1-(3-氟丙基)氮杂环丁烷-3-胺盐酸盐(89.27mg,369.57μmol)溶于二氧六环(4mL)中,加入叔丁醇钠(193.73mg,2.02mmol),在氮气保护下加入甲烷磺酸(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)(30.46mg,33.60μmol),反应液于80℃搅拌16小时。将反应液冷却到室温,减压浓缩至干,经薄层色谱纯化(二氧化硅,石油醚/四氢呋喃=1/2)得到产物1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢苯并呋喃[2,3-c]吡啶-6-醇(68mg)。
MS m/z(ESI):528.1[M+H] +
步骤8:1-(2,6-二氟-4-(((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢苯并呋喃[2,3-c]吡啶-6-基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000091
将1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢苯并呋喃[2,3-c]吡啶-6-醇(65mg,123.22μmol)和)N-苯基双(三氟甲烷磺酰)亚胺(88.04mg,246.45μmol)溶于二氯甲烷(2mL)中,加入三乙胺(37.41mg,369.67μmol),反应液于25℃搅拌13小时。将反应液减压浓缩至干,经薄层色谱纯化(二氧化硅,石油醚/四氢呋喃=1/2)得到化合物1-(2,6-二氟-4-(((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢苯并呋喃[2,3-c]吡啶-6-基三氟甲磺酸酯(50mg)。
1H NMR(400MHz,METHANOL-d 4)δ7.53-7.46(m,2H),7.23-7.19(m,1H),6.12(d,J=11.5Hz,2H),5.19(s,1H),4.54(t,J=5.8Hz,1H),4.42(t,J=5.9Hz,1H),4.12(t,J=6.8Hz,1H),3.91(m,2H),3.62-3.53(m,1H),3.44-3.33(m,2H),3.15(m,2H),3.02-2.91(m,1H),2.77(t,J=7.4Hz,2H),2.67-2.58(m,1H),1.88-1.74(m,2H),1.20(d,J=6.8Hz,3H).
步骤9:(1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢苯并呋喃[2,3-c]吡啶-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000092
将1-(2,6-二氟-4-(((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢苯并呋喃[2,3-c]吡啶-6-基三氟甲磺酸酯(45mg,68.23μmol)和联硼酸新戊二醇酯(30.82mg,136.45μmol)溶于二氧六环(1mL)中,加入醋酸钾(13.39mg,136.45μmol),在氮气保护下加入1,1-双(二苯基磷)二茂铁氯化钯(69.66mg,95.20μmol),反应液于90℃搅拌2小时。LCMS显示反应完成。将反应液冷却到室温,减压浓缩至干,浓缩物经制备液相色谱纯化(Phenomenex Gemini-NX C18柱:3um二氧化硅,30mm直径,75mm长度;使用水(含有0.225%甲酸)和乙腈的极性递减的混合物作为洗脱液)得到产物(1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢苯并呋喃[2,3-c]吡啶-6-基)硼酸(15mg)。
MS m/z(ESI):556.1[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.99-7.73(m,1H),7.69-7.45(m,1H),7.32(br s,1H),6.17-6.07(m,2H),5.16(s,1H),4.56(t,J=5.7Hz,1H),4.44(t,J=5.7Hz,1H),4.27-4.17(m,1H),4.13-4.01(m,2H),3.62-3.51(m,1H),3.45-3.33(m,3H),3.07-2.88(m,4H),2.61(dd,J=4.0,15.7Hz,1H),1.94-1.76(m,2H),1.20(d,J=6.8Hz,3H).
实施例7:((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2(三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000093
步骤1:叔丁基N-[(1R)-2-(5-苄氧基-1H-吲哚-3-基)-1-甲基-乙基]氨基甲酯的合成
Figure PCTCN2021088291-appb-000094
将5-苄氧基吲哚(2.12g,9.48mmol)和氯化亚铜(813.61mg,8.22mmol)溶解在四氢呋喃(20mL)中,降温到0℃,然后甲基溴化镁(3M,2.74mL)加入到上述体系中,在0℃搅拌30分钟,然后再将(R)-4-甲基-2,2-二氧代-[1,2,3]氧杂噻唑烷-3-甲酸叔丁酯(1.5g,6.32mmol)的四氢呋喃(10mL)溶液滴加到反应液中,滴加完后反应液升到室温搅拌过夜。LCMS监测反应完毕。向反应液中加水(2mL),然后用乙酸乙酯(20mL)萃取两次,有机相合并后用无水硫酸钠干燥,过滤,浓缩到干。然后经柱层析纯化(二氧化硅,石油醚/ 四氢呋喃=1/3)得化合物叔丁基N-[(1R)-2-(5-苄氧基-1H-吲哚-3-基)-1-甲基-乙基]氨基甲酯(1.1g)。
MS m/z(ESI):325.1[M+H-56] +
1H NMR(400MHz,CHLOROFORM-d)δ7.99(brs,1H),7.51(d,J=7.2Hz,2H),7.44-7.38(m,2H),7.37-7.32(m,1H),7.28(s,1H),7.20(brs,1H),7.03(s,1H),6.96(dd,J=8.8,2.4Hz,1H),5.15(s,2H),4.56-4.39(m,1H),4.03(brs,1H),2.97-2.89(m,1H),2.87-2.77(m,1H),1.45(s,9H),1.14(d,J=6.8Hz,3H).
步骤2:(2R)-1-(5-苄氧基-1H-吲哚-3-基)丙烷-2-胺的合成
Figure PCTCN2021088291-appb-000095
将叔丁基N-[(1R)-2-(5-苄氧基-1H-吲哚-3-基)-1-甲基-乙基]氨基甲酯(1.1g,2.89mmol)溶解在二氧六环(5mL)中,然后加入4M盐酸二氧六环(7.3mL),反应液室温搅拌1小时。LCMS监测反应完毕。反应液浓缩后加入水(10mL),然后用碳酸钾调节pH到8-9。用二氯甲烷(50mL)萃取三次,合并有机相,用无水硫酸钠干燥,过滤,浓缩得化合物(2R)-1-(5-苄氧基-1H-吲哚-3-基)丙烷-2-胺(600mg)。
MS m/z(ESI):281.1[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.48(d,J=7.2Hz,2H),7.40-7.35(m,2H),7.34-7.29(m,1H),7.26(d,J=8.8Hz,1H),7.13(d,J=2.4Hz,1H),7.06(s,1H),6.86(dd,J=8.8,2.4Hz,1H),5.11(s,2H),3.22-3.14(m,1H),2.84-2.76(m,1H),2.73-2.65(m,1H),1.14(d,J=6.4Hz,3H)
步骤3:(2R)-1-(5-苄氧基-1H-吲哚-3-基)-N-(2,2,2-三氟乙基)丙烷-2-胺的合成
Figure PCTCN2021088291-appb-000096
将(2R)-1-(5-苄氧基-1H-吲哚-3-基)丙烷-2-胺(600mg,2.14mmol)和二异丙基乙胺(331.91mg,2.57mmol)溶解在二氧六环(3mL)中,然后将三氟甲磺酸三氟乙酯(596.06mg,2.57mmol)加入到反应液中,将反应升温到80℃搅拌过夜。LCMS监测反应完毕。反应液浓缩到干,然后经柱层析纯化(二氧化硅,石油醚/四氢呋喃=1/3)得化合物(2R)-1-(5-苄氧基-1H-吲哚-3-基)-N-(2,2,2-三氟乙基)丙烷-2-胺(410mg)。
MS m/z(ESI):363.2[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.47(d,J=7.2Hz,2H),7.41-7.35(m,2H),7.33-7.28(m,1H),7.26(d,J=8.8Hz,1H),7.12(d,J=2.4Hz,1H),7.06(s,1H),6.86(dd,J=8.8,2.4Hz,1H),5.11(s,2H),3.27-3.15(m,2H),3.09-2.98(m,1H),2.89-2.81(m,1H),2.75-2.68(m,1H),1.08(d,J=6.4Hz,3H)
步骤4:3-[(2R)-2-(2,2,2-三氟乙胺基)丙基]-1H-吲哚-5-酚的合成
Figure PCTCN2021088291-appb-000097
将(2R)-1-(5-苄氧基-1H-吲哚-3-基)-N-(2,2,2-三氟乙基)丙烷-2-胺(410mg,1.13mmol)溶解在甲醇(15mL)中,然后加入Pd/C(100mg,10%),反应液在15psi氢气压力下室温搅拌5小时。LCMS监测反应完毕。反 应液过滤,滤液浓缩到干得化合物3-[(2R)-2-(2,2,2-三氟乙胺基)丙基]-1H-吲哚-5-酚(295mg)。
MS m/z(ESI):273.0[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.18(d,J=8.8Hz,1H),7.03(s,1H),6.93(d,J=2.4Hz,1H),6.69(dd,J=8.6,2.4Hz,1H),3.25(q,J=9.8Hz,2H),3.12-3.01(m,1H),2.87-2.79(m,1H),2.74-2.65(m,1H),1.11(d,J=6.4Hz,3H).
步骤5:(1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-6-酚的合成
Figure PCTCN2021088291-appb-000098
将3-[(2R)-2-(2,2,2-三氟乙胺基)丙基]-1H-吲哚-5-酚(60mg,220.37μmol)溶解在甲苯(10mL)和乙酸(2mL)中,然后加入2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯甲醛(66.00mg,242.41μmol),反应液升温到80℃搅拌过夜。LCMS监测反应完毕。反应液浓缩到干,然后经柱层析纯化(二氧化硅,石油醚/四氢呋喃=1/5)得化合物(1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-6-酚(70mg)。
MS m/z(ESI):527.3[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.04(d,J=8.4Hz,1H),6.81(d,J=2.4Hz,1H),6.66-6.52(m,1H),6.13(d,J=11.6Hz,2H),5.20(s,1H),4.59-4.54(m,1H),4.48-4.42(m,1H),4.22-4.14(m,1H),4.01(brt,J=8.0Hz,2H),3.58-3.51(m,1H),3.29(brd,J=5.6Hz,2H),3.26(brs,1H),3.18-3.08(m,1H),3.03-2.83(m,5H),2.55(dd,J=14.6,4.8Hz,1H),1.92-1.85(m,1H),1.84-1.79(m,1H),1.18(d,J=6.4Hz,3H)
步骤6:(1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-6-基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000099
在冰浴冷却下,将(1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-6-酚(70mg,132.95μmol)和1,1,1-三氟-N-苯基-N-(三氟甲磺酰)甲磺酰胺(94.99mg,265.90μmol)溶解在二氯甲烷(5mL)中,然后将三乙胺(40.36mg,398.85μmol)加到反应液中,反应液升到室温后搅拌过夜。LCMS监测反应完毕。反应液浓缩到干,然后经柱层析纯化(二氧化硅,石油醚/四氢呋喃=1/3)得化合物(1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-6-基三氟甲磺酸酯(55mg)。
MS m/z(ESI):659.1[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.37(d,J=2.4Hz,1H),7.11-7.06(m,1H),6.99(dd,J=2.4Hz 8.8Hz,1H),6.15(d,J=11.6Hz,2H),5.25(s,1H),4.56(t,J=5.6Hz,1H),4.45(t,J=5.6Hz,1H),4.20-4.12(m,1H),4.00(br t,J=7.6Hz,2H),3.61-3.55(m,1H),3.26(br t,J=7.6Hz,2H),3.06-2.94(m,2H),2.90-2.83(m,3H),2.64(dd,J=15.2,4.8Hz,1H),1.90-1.86(m,1H),1.85-1.78(m,1H),1.19(d,J=6.4Hz,3H).
步骤7:((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2(三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000100
将(1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2-三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-6-基三氟甲磺酸酯(52mg,78.96μmol),2-(5,5-二甲基-1,3,2-二噁硼己环-2-基)-5,5-二甲基-1,3,2-二噁硼己环(53.51mg,236.87μmol)和乙酸钾(23.25mg,236.87μmol)溶解在二氧六环(3mL)中,然后将[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(6.45mg,7.90μmol)加到上述溶液中,反应液氮气置换三次,加热到80℃搅拌过夜。反应液浓缩到干然后经制备液相色谱纯化(Phenomenex Synergi C18柱:4um二氧化硅,30mm直径,150mm长度;使用水(含有0.225%甲酸)和乙腈的极性递减的混合物作为洗脱液)得到化合物((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-(2,2,2(三氟乙基)-2,3,4,9-四氢-1H-吡啶并[3,4-b]吲哚-6-基)硼酸(5.2mg)。
MS m/z(ESI):555.2[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.97-7.74(m,1H),7.53-7.31(m,1H),7.26-7.14(m,1H),6.16(brd,J=11.6Hz,2H),5.24(s,1H),4.60(brs,2H),4.48(brs,1H),4.29(brs,2H),3.57(br d,J=6.2Hz,3H),3.38(br s,1H),3.15(br s,2H),3.07-2.94(m,2H),2.66(br d,J=14.2Hz,1H),2.01-1.86(m,2H),1.19(d,J=6.4Hz,3H).
实施例8:((1S,3R)-1-(5-(2-((3-氟丙基)氨基)乙氧基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000101
步骤1:叔丁基(3-氟丙基)(2-((6-((1S,3R)-6-羟基-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-1-基)吡啶-3-基)氧代)乙基)氨基甲酯的合成
Figure PCTCN2021088291-appb-000102
将(1S,3R)-1-(5-溴吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚(200mg,498.48μmol)和叔丁基(3-氟丙基)(2-羟基乙基)氨基甲酯(132.36mg,598.18μmol)溶于甲苯(4mL)中,在25℃下加入碳酸铯(487.24mg,1.50mmol)和甲磺酸-2-(二叔丁基膦基)-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)(42.59mg,49.85μmol),反应液于90℃搅拌反应13小时。反应液减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/乙酸乙酯=2/1)得到产物叔丁基(3-氟丙基)(2-((6-((1S,3R)-6-羟基-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-1-基)吡啶-3-基)氧代)乙基)氨基甲酯(160mg)。
MS m/z(ESI):542.2[M+H] +
步骤2:(1S,3R)-1-(5-(2-((叔-丁氧羰基)(3-氟丙基)氨基)乙氧基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000103
将叔丁基(3-氟丙基)(2-((6-((1S,3R)-6-羟基-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-1-基)吡啶-3-基)氧代)乙基)氨基甲酯(160mg,165.44μmol)和N-苯基双(三氟甲烷磺酰)亚胺(118.21mg,330.89μmol)溶于二氯甲烷(3mL)中,加入三乙胺(50.22mg,496.33μmol),反应液于25℃搅拌反应13小时。将反应液倒入水中并搅拌10分钟,二氯甲烷(10mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/乙酸乙酯=2/1)得到产物(1S,3R)-1-(5-(2-((叔-丁氧羰基)(3-氟丙基)氨基)乙氧基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(50mg)。
MS m/z(ESI):674.1[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ8.19(d,J=2.8Hz,1H),7.31(br s,1H),7.21(br s,1H),7.07(s,1H),6.98(s,2H),5.02(s,1H),4.56(br s,1H),4.45(br s,1H),4.14(br d,J=19.6Hz,2H),3.63(br s,2H),3.55(br d,J=6.3Hz,1H),3.46(br t,J=6.8Hz,2H),3.24(br dd,J=6.4,15.7Hz,2H),3.04-2.92(m,1H),2.66(dd,J=5.5,16.6Hz,1H),2.01(br s,2H),1.48(s,9H),1.11(d,J=6.5Hz,3H).
步骤3:((1S,3R)-1-(5-(2-((叔-丁氧羰基)(3-氟丙基)氨基)乙氧基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000104
将(1S,3R)-1-(5-(2-((叔-丁氧羰基)(3-氟丙基)氨基)乙氧基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(50mg,74.22μmol)和联硼酸新戊二醇酯(33.53mg,148.45μmol)溶于二甲基亚砜(2mL)中,加入乙酸钾(21.85mg,222.67μmol)和[1,1-双(二苯基膦)二茂铁]二氯化钯二氯甲烷(9.09mg,11.13μmol),反应液80℃搅拌反应2小时。将反应液倒入水中并搅拌10分钟,乙酸乙酯(10mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干得到粗产物((1S,3R)-1-(5-(2-((叔-丁氧羰基)(3-氟丙基)氨基)乙氧基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸(60mg)。
MS m/z(ESI):570.1[M+H] +
步骤4:((1S,3R)-1-(5-(2-((3-氟丙基)氨基)乙氧基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000105
将((1S,3R)-1-(5-(2-((叔-丁氧羰基)(3-氟丙基)氨基)乙氧基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸(60mg,52.69μmol)溶于二氯甲烷(0.5mL)中,加入盐酸1,4-二氧六环溶液(4M,131.72μL)。反应液25℃搅拌反应2小时。将反应液减压浓缩至干。经制备液相色谱纯化(Phenomenex Gemini-NX柱:3um二氧化硅,30mm直径,150mm长度;使用水(含有10mM碳酸氢铵)和乙腈的极性递减的混合物作为洗脱液)纯化得到化合物((1S,3R)-1-(5-(2-((3-氟丙基)氨基)乙氧基)吡啶-2-基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸(7.92mg)。
MS m/z(ESI):470.3[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ8.18(d,J=2.8Hz,1H),7.60-7.48(m,1H),7.44-7.34(m,2H),7.33-7.27(m,1H),6.70(d,J=7.8Hz,1H),5.00-4.95(m,1H),4.60(t,J=5.8Hz,1H),4.48(t,J=5.8Hz,1H),4.20(t,J=5.1Hz,2H),3.61-3.48(m,1H),3.45-3.34(m,1H),3.28-3.16(m,1H),3.09(t,J=5.1Hz,2H),3.01-2.79(m,3H),2.69(dd,J=4.6,15.9Hz,1H),2.08-1.82(m,2H),1.09(d,J=6.5Hz,3H).
实施例9:((1S,3R)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3,5-二甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000106
步骤1:(R)-叔-丁基(1-(3-甲氧基-2-甲基苯基)丙烷-2-基)氨基甲酯的合成
Figure PCTCN2021088291-appb-000107
将1-溴-3-甲氧基-2-甲苯(4g,19.89mmol)溶于四氢呋喃(40mL)中,在-70℃下加入正丁基锂(2.5M,9.55mL),反应液于-70℃搅拌反应1小时。随后加入溶在四氢呋喃(20mL)中的(R)-4-甲基-2,2-二氧-[1,2,3]氧杂噻唑烷-3-甲酸叔丁酯(5.19g,21.88mmol),反应液于-70℃下搅拌2小时,然后缓慢升温到25℃,搅拌反应13小时。TLC(石油醚:乙酸乙酯=10:1)检测反应完成。向该反应液中滴加氯化铵溶液(10mL)和水(20mL)并搅拌10分钟,乙酸乙酯(50mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/乙酸乙酯=10/1)得到产物(R)-叔-丁基(1-(3-甲氧基-2-甲基苯基)丙烷-2-基)氨基甲酯(1.2g)。
MS m/z(ESI):224.0[M+H-56] +
1H NMR(400MHz,CHLOROFORM-d)δ7.11(t,J=8.0Hz,1H),6.77(t,J=7.3Hz,2H),4.44(s,1H),3.84(s,3H),2.95(s,1H),2.67-2.60(m,1H),2.25(s,3H),1.44(s,9H),1.11(d,J=6.8Hz,3H)
步骤2:(R)-1-(3-甲氧基-2-甲基苯基)丙烷-2-胺的合成
Figure PCTCN2021088291-appb-000108
将(R)-叔-丁基(1-(3-甲氧基-2-甲基苯基)丙烷-2-基)氨基甲酯(1.2g,3.01mmol)溶于1,4-二氧六环(10mL)中,加入盐酸/1,4-二氧六环(4M,11.28mL),反应液于25℃搅拌反应13小时。LCMS检测反应完成。将反应液浓缩,用饱和碳酸氢钠中和至pH=8,乙酸乙酯(20mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干,得到产物(R)-1-(3-甲氧基-2-甲基苯基)丙烷-2-胺(0.5g)。
MS m/z(ESI):180.0[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.11(t,J=8.0Hz,1H),6.84-6.74(m,2H),3.83(s,3H),3.15-3.07(m,J=6.4Hz,1H),2.71(d,J=6.8Hz,2H),2.18(s,3H),1.11(d,J=6.3Hz,3H)
步骤3:(R)-1-(3-甲氧基-2-甲基苯基)-N-(2,2,2-三氟乙基)丙烷-2-胺的合成
Figure PCTCN2021088291-appb-000109
将(R)-1-(3-甲氧基-2-甲基苯基)丙烷-2-胺(0.5g,2.79mmol)和三氟甲磺酸三氟乙酯(712.13mg,3.07mmol)溶于1,4-二氧六环(5mL)中,加入N,N-二异丙基乙胺(1.08g,8.37mmol),反应液80℃搅拌反应13小时。LCMS检测反应完成。将反应液减压浓缩,得到的粗品倒入水中并搅拌10分钟,乙酸乙酯(20mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干,得到产物(R)-1-(3-甲氧基-2-甲基苯基)-N-(2,2,2-三氟乙基)丙烷-2-胺(0.5g)。
MS m/z(ESI):262.1[M+H] +
步骤4:(R)-2-甲基-3-(2-((2,2,2-三氟乙基)氨基)丙基)苯酚的合成
Figure PCTCN2021088291-appb-000110
将(R)-1-(3-甲氧基-2-甲基苯基)-N-(2,2,2-三氟乙基)丙烷-2-胺(450mg,1.72mmol)溶于二氯甲烷(8mL)中,在0℃下加入三溴化硼(1.29g,5.17mmol),反应液在25℃下搅拌反应4小时。LCMS检测反应完成。将反应液倒入冰水(10mL)中并用饱和碳酸氢钠中和至pH=8,二氯甲烷(20mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干,得到产物(R)-2-甲基-3-(2-((2,2,2-三氟乙基)氨基)丙基)苯酚(0.39g)。
MS m/z(ESI):248.0[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ7.03(t,J=7.8Hz,1H),6.76(d,J=7.5Hz,1H),6.69(d,J=7.9Hz,1H),4.72(br d,J=8.8Hz,1H),3.18(q,J=9.4Hz,2H),3.04-3.02(m,1H),2.86-2.73(m,1H),2.72-2.59(m,1H),2.23(s,3H),1.11(d,J=6.2Hz,3H)
步骤5:(1S,3R)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3,5-二甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000111
将(R)-2-甲基-3-(2-((2,2,2-三氟乙基)氨基)丙基)苯酚(410mg,1.66mmol)和5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)甲基吡啶醛(460mg,1.94mmol)溶于1,2-二氯乙烷(40mL)中,加入三氟乙酸(567.20mg,4.97mmol),反应液25℃搅拌反应13小时。LCMS检测反应完成。将反应液倒入饱和碳酸氢钠(50mL)并搅拌10分钟,二氯甲烷(40mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,二氯甲烷/甲醇=10/1)得到产物(1S,3R)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3,5-二甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚(410mg)。
MS m/z(ESI):467.2[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.75(d,J=2.8Hz,1H),7.08(d,J=8.8Hz,1H),6.96-6.91(m,1H),6.53(d,J=8.3Hz,1H),6.42(d,J=8.5Hz,1H),4.92(s,1H),4.60-4.49(m,1H),4.47-4.39(m,1H),4.19-4.06(m,1H),3.84(br t,J=7.2Hz,2H),3.56-3.42(m,1H),3.32-3.22(m,1H),3.10-3.00(m,2H),3.00-2.79(m,2H), 2.74-2.66(m,2H),2.58(dd,J=5.8,16.6Hz,1H),2.20-2.05(m,3H),1.87-1.71(m,2H),1.09(d,J=6.8Hz,3H)
步骤6:(1S,3R)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3,5-二甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000112
将(1S,3R)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3,5-二甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚(410.00mg,790.97μmol)和1,1,1-三氟-N-苯基-N-((三氟甲基)磺酰)甲磺酰胺(565.15mg,1.58mmol)溶于二氯甲烷(10mL)中,加入三乙胺(240.12mg,2.37mmol),反应液25℃搅拌反应13小时。LCMS检测反应完成。将反应液倒入水(10mL)中并搅拌10分钟,二氯甲烷(20mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干,然后柱层析纯化(二氧化硅,二氯甲烷/甲醇=10/1)得到产物(1S,3R)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3,5-二甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(0.9g)。
MS m/z(ESI):599.1[M+H] +
步骤7:((1S,3R)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3,5-二甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000113
将(1S,3R)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3,5-二甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(0.2g,167.06μmol)和5,5,5',5'-四甲基-2,2'-联(1,3,2-二噁硼己环)(113.21mg,501.19μmol)溶于二甲基亚砜(4mL)中,加入[1,1-双(二苯基膦)二茂铁]二氯化钯二氯甲烷(20.46mg,25.06μmol)和乙酸钾(49.19mg,501.19μmol)。反应液80℃搅拌反应13小时。LCMS检测反应完成。将反应液冷却至室温并倒入水(12mL)中并搅拌10分钟,乙酸乙酯(10mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。经制备液相色谱纯化(Phenomenex Gemini-NX C18柱:3um二氧化硅,30mm直径,75mm长度;使用水(含有0.225%乙酸)和乙腈的极性递减的混合物作为洗脱液)纯化得到化合物((1S,3R)-1-(5-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)吡啶-2-基)-3,5-二甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸(30mg)。
MS m/z(ESI):495.2[M+H] +
1H NMR(400MHz,METHANOL-d4)δ8.52(s,1H),7.87-7.73(m,1H),7.24(d,J=8.6Hz,1H),7.13(d,J=8.6Hz,1H),7.05-6.88(m,2H),6.63(br d,J=7.5Hz,1H),5.07(s,1H),4.99-4.94(m,1H),4.59(t,J=5.7Hz,1H),4.47(t,J=5.6Hz,1H),4.37-4.26(m,1H),4.22(br t,J=8.2Hz,2H),3.62-3.48(m,3H),3.46-3.34(m,1H),3.08(t,J=7.4Hz,2H),3.03-2.82(m,2H),2.62(dd,J=5.7,16.6Hz,1H),2.40-2.19(m,4H),1.99-1.82(m,3H),1.35(d,J=6.2Hz,1H),1.10(d,J=6.5Hz,3H))
实施例10和11:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-基)硼酸和((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000114
步骤1:(R)-N-(1-(3-(苄氧基)苯基)丙烷-2-基)苯胺的合成
Figure PCTCN2021088291-appb-000115
将(R)-1-(3-(苄氧基)苯基)丙烷-2-胺(500mg,2.07mmol,1eq)溶于甲苯(20mL),加入溴苯(390.36mg,2.49mmol,1.2eq)和叔丁醇钠(398.23mg,4.14mmol,2eq),在氮气氛围下将三(二亚苄基丙酮)二钯(189.73mg,207.19μmol,0.1eq)和2-二叔丁基膦-2′,4′,6′-三异丙基联苯(87.98mg,207.19μmol,0.1eq)加入该体系。反应液在80℃下反应16小时。LCMS监测反应完成。反应液冷却至室温减压浓缩后经过柱层析纯化(二氧化硅,石油醚/乙酸乙酯=100/25)得到产物(R)-N-(1-(3-(苄氧基)苯基)丙烷-2-基)苯胺(380.00mg)。
MS m/z(ESI):318.1[M+H] +
步骤2:(R)-3-(2-(苯基氨基)丙基)苯酚的合成
Figure PCTCN2021088291-appb-000116
将(R)-N-(1-(3-(苄氧基)苯基)丙烷-2-基)苯胺(350mg,1.10mol,1eq)溶于乙酸乙酯(30mL)中,在氢气环境下加入氢氧化钯碳(100mg,10%纯度),反应液于25℃的氢气(15Psi)环境下搅拌16小时。LCMS显示反应完成。将反应液过滤浓缩后经过薄层层析纯化(二氧化硅,石油醚/乙酸乙酯=3/1)得到产物(R)-3-(2-(苯基氨基)丙基)苯酚(190.00mg)。
MS m/z(ESI):228.0[M+H] +
1H NMR(400MHz,METHANOL-d4)δ7.18-7.05(m,3H),6.72-6.66(m,4H),6.66-6.60(m,2H),3.73-3.63(m,1H),2.92(dd,J=4.7,13.2Hz,1H),2.51(dd,J=8.1,13.1Hz,1H),1.12(d,J=6.5Hz,3H).
步骤3:(1S,3R)-1-(4-溴-2,6-二氟苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000117
将(R)-3-(2-(苯基氨基)丙基)苯酚(190mg,835.90μmol)溶于乙腈(32mL),加入4-溴-2,6-二氟苯(甲)醛(203.21mg,919.49μmol)和乙酸(8mL)。反应液于100℃搅拌反应16小时。LCMS和TLC(石油醚:乙酸乙酯=3:1)监测反应完成后,将反应溶液冷却至室温后,减压浓缩干,用二氯甲烷(3mL稀释)后经过薄层层析纯化(二氧化硅,石油醚/乙酸乙酯=5/1)得到(1S,3R)-1-(4-溴-2,6-二氟苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-酚(227.00mg)。
MS m/z(ESI):431.8[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ7.23-7.19(m,2H),7.00(d,J=8.0Hz,2H),6.95-6.90(m,2H),6.82(t,J=7.3Hz,1H),6.66-6.61(m,2H),6.55(d,J=1.3Hz,1H),6.04(s,1H),4.76(br s,1H),3.60-3.51(m,1H),2.65(dd,J=2.3,15.6Hz,1H),0.97(d,J=6.5Hz,3H)。
步骤4:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000118
将(1S,3R)-1-(4-溴-2,6-二氟苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-酚(227.00mg,527.56μmol)溶于叔戊醇(10mL),加入1-(3-氟丙基)氮杂环丁烷-3-胺(152.93mg,633.07μmol),在氮气环境下将碳酸铯(1.03g,3.17mmol)和Brettphos Pd G3(47.82mg,52.76μmol)加入该反应体系。反应液在120℃下搅拌反应13小时。LCMS监测反应完成,反应液减压浓缩干后,用乙酸乙酯稀释(5mL),然后用食盐水(10mL)洗涤三次,收集到的水相用乙酸乙酯(15mL)萃取三次,有机相用硫酸钠干燥后减压浓缩干,然后经薄层层析纯化(二氧化硅,石油醚/四氢呋喃=1/1,1%氨水)得(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-酚(100.00mg)。
MS m/z(ESI):482.2[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.15-7.10(m,2H),6.98(d,J=8.0Hz,2H),6.72(t,J=7.3Hz,1H),6.55(d,J=2.5Hz,2H),6.49-6.43(m,1H),5.92(d,J=11.8Hz,2H),5.48(s,1H),4.52-4.48(m,1H),4.40-4.36(m,1H),3.93(td,J=3.2,6.7Hz,1H),3.74-3.70(m,4H),3.55-3.49(m,1H),2.88-2.84(m,2H),2.59-2.56(m,2H),1.78-1.69(m,2H),0.92(d,J=6.5Hz,3H)
步骤5:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯的合成。
Figure PCTCN2021088291-appb-000119
将(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-酚(100mg,207.66μmol)溶于二氯甲烷(2mL),加入N-苯基双(三氟甲烷磺酰)亚胺(111.28mg,311.49μmol)和三乙胺(63.04mg,622.99μmol)。反应液于25℃搅拌反应16个小时。LCMS检测反应完成后,将反应溶液冷却至室温后,减压浓缩干,用二氯甲烷(2mL)稀释后经薄层层析纯化(二氧化硅,石油醚/四氢呋喃=1/1)得到(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(95.00mg)。
MS m/z(ESI):614.1[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.19-7.14(m,4H),7.07-7.03(m,3H),6.86-6.80(m,1H),5.96(d,J=7.8Hz,2H),5.91(s,1H),4.52-4.47(m,1H),4.40-4.35(m,1H),4.05-3.97(m,1H),3.90-3.81(m,3H),3.63-3.56(m,1H),3.14-3.05(m,3H),2.79-2.75(m,2H),1.85-1.74(m,2H),0.94(d,J=6.5Hz,3H)
步骤6:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-基)硼酸和((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000120
将(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(95.00mg,154.82μmol)溶于二氧六环(1.5mL),加入5,5,5',5'-四甲基-2,2'-联(1,3,2-二噁硼己环)(69.94mg,309.64μmol),乙酸钾(45.58mg,464.46μmol),1,1-双(二苯基磷)二茂铁氯化钯(16.99mg,23.22μmol)在氮气环境下加入该反应体系中。反应液于80℃搅拌反应2小时。LCMS检测反应完成后,将反应溶液冷却至室温后,减压浓缩后经制备液相色谱(Phenomenex Gemini-NX C18柱,3um二氧化硅,30mm直径,75mm长度);(使用水(含有0.225%甲酸)和和乙腈的极性递减的混合物作为洗脱液),纯化得到((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-基)硼酸(第一个峰,实施例10,4.29mg)和((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-2-苯基-1,2,3,4-四氢异喹啉-6-基)硼酸(第二个峰,实施例11,5.19mg)。
MS m/z(ESI):510.0[M+H] +
实施例10:
1H NMR(400MHz,METHANOL-d 4)δ7.49(br s,1H),7.36(br s,1H),7.18-7.11(m,2H),7.10-6.97(m,3H),6.65(d,J=7.8Hz,1H),5.96-5.85(m,2H),5.57(s,1H),4.54(t,J=5.8Hz,1H),4.42(t,J=5.6Hz,1H),4.12-4.02(m,1H),4.00-3.91(m,2H),3.52(br s,1H),3.23(br s,2H),3.08-2.91(m,2H),2.85(br t,J=7.4Hz,2H),1.89-1.73(m,2H),0.91(d,J=6.0Hz,3H)
实施例11(峰2):
1H NMR(400MHz,METHANOL-d 4)δ7.55-7.39(m,2H),7.19-7.09(m,2H),7.04(d,J=7.8Hz,2H),6.97(br d,J=7.8Hz,1H),6.80-6.73(m,1H),6.01-5.90(m,3H),4.52(t,J=5.6Hz,1H),4.40(t,J=5.8Hz,1H),4.34(br dd,J=3.3,8.3Hz,1H),4.12-4.03(m,1H),3.97(br t,J=7.8Hz,2H),3.60(br dd,J=4.8,15.3Hz,1H),3.27-3.18(m,2H),2.86(br t,J=7.4Hz,2H),2.77-2.68(m,1H),1.90-1.72(m,2H),0.92(d,J=6.5Hz,3H)
实施例12:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氧代)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000121
步骤1:叔-丁基3-(3,5-二氟-4-甲酰基苯氧基)氮杂环丁烷-1-甲酸基酯的合成
Figure PCTCN2021088291-appb-000122
将2,6-二氟-4-羟基苯甲醛(0.8g,5.06mmol)和叔丁基-3-碘氮杂环丁烷-1-甲酸基酯(3.58g,12.65mmol)和碳酸铯(4.12g,12.65mmol)溶于N,N-二甲基甲酰胺(27mL)中,在150℃下微波反应1.5小时。TLC(石油 醚:乙酸乙酯=3:1)检测反应完成。将反应液倒入水中并搅拌10分钟,乙酸乙酯(30mL)萃取3次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/乙酸乙酯=3/1)得到产物叔-丁基3-(3,5-二氟-4-甲酰基苯氧基)氮杂环丁烷-1-甲酸基酯(0.3g)。
1H NMR(400MHz,CHLOROFORM-d)δ10.13(s,1H),6.29(s,1H),6.27(s,1H),4.83(tt,J=4.0,6.4Hz,1H),4.28(dd,J=0.9,6.4Hz,1H),4.26(dd,J=0.9,6.4Hz,1H),3.97-3.95(m,1H),3.94-3.90(m,1H),1.38(s,9H).
步骤2:4-(氮杂环丁烷-3-氧基)-2,6-二氟苯甲醛盐酸盐的合成
Figure PCTCN2021088291-appb-000123
将叔-丁基3-(3,5-二氟-4-甲酰基苯氧基)氮杂环丁烷-1-甲酸基酯(300mg,957.56μmol)溶于二氯甲烷(6mL)中,在0℃下加入盐酸/1,4-二氧六环溶液(4M,3mL),反应液于25℃搅拌反应2小时。TLC(石油醚:乙酸乙酯=5:1)检测反应完成。将反应液减压浓缩至干,得到产物4-(氮杂环丁烷-3-氧基)-2,6-二氟苯甲醛盐酸盐(0.23g)。
1H NMR(400MHz,DMSO-d6)δ10.08(s,1H),7.00-6.83(m,2H),5.33-5.13(m,1H),4.59-4.36(m,2H),4.10-3.91(m,2H)
步骤3:2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氧代)苯甲醛的合成
Figure PCTCN2021088291-appb-000124
将4-(氮杂环丁烷-3-氧基)-2,6-二氟苯甲醛盐酸盐(200mg,801.15μmol)和1-氟-3-碘-丙烷(301.20mg,1.60mmol)溶于乙腈(10mL)中,加入碳酸钾(332.18mg,2.40mmol),反应液50℃搅拌反应13小时。TLC(二氯甲烷:甲醇=10:1)检测反应完成。将反应液过滤减压浓缩至干。然后柱层析纯化(二氧化硅,二氯甲烷:甲醇=10:1)得到产物2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氧代)苯甲醛(60mg)。
1H NMR(400MHz,METHANOL-d 4)δ10.24-10.12(m,1H),6.73-6.44(m,2H),5.04-4.93(m,1H),4.54(t,J=5.9Hz,1H),4.43(t,J=5.9Hz,1H),3.89-3.77(m,2H),3.31-3.19(m,2H),2.70(t,J=7.5Hz,2H),1.88-1.68(m,2H)
步骤4:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氧代)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000125
将2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氧代)苯甲醛(60mg,219.58μmol)和(R)-3-(2-((2,2,2-三氟乙基)氨基)丙基)苯酚(51.21mg,219.58μmol)溶于甲苯(3mL)中,加入乙酸(630.00mg,10.49mmol),反应液于90℃下搅拌反应13小时。LCMS检测反应完成。将反应液减压浓缩至干。然后柱层析纯化(二氯甲烷: 甲醇=10:1)得到产物(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氧代)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚(50mg)。
MS m/z(ESI):489.2[M+H] +
步骤5:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氧代)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000126
将(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氧代)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-酚(50mg,102.36μmol)和1,1,1-三氟-N-苯基-N-((三氟甲基)磺酰)甲磺酰胺(109.71mg,307.08μmol)溶于二氯甲烷(3mL)中,加入三乙胺(31.07mg,307.08μmol),反应液25℃搅拌反应13小时。LCMS检测反应完成。将反应液倒入水中并搅拌10分钟,二氯甲烷(20mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/乙酸乙酯=3/1)得到产物(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氧代)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(50mg)。
MS m/z(ESI):621.0[M+H] +
步骤6:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氧代)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000127
将(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氧代)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(50mg,80.58μmol)和5,5,5',5'-四甲基-2,2'-联(1,3,2-二噁硼己环)(54.60mg,241.73μmol)溶于二甲基亚砜(2mL)中,加入[1,1-双(二苯基膦)二茂铁]二氯化钯二氯甲烷(9.87mg,12.09μmol)和乙酸钾(23.72mg,241.73μmol)。反应液90℃搅拌反应2小时。LCMS检测反应完成。将反应液冷却至室温并倒入水(20mL)中并搅拌10分钟,乙酸乙酯(20mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。经制备液相色谱纯化(Phenomenex Gemini-NX C18柱:3um二氧化硅,30mm直径,75mm长度;使用水(含有0.225%乙酸)和乙腈的极性递减的混合物作为洗脱液)纯化得到化合物((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氧代)苯基)-3-甲基-2-(2,2,2-三氟乙基)-1,2,3,4-四氢异喹啉-6-基)硼酸(14.46mg)。
MS m/z(ESI):517.1[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.58-7.29(m,2H),6.71(d,J=7.5Hz,1H),6.52-6.39(m,2H),5.26(s,1H),4.87-4.81(m,1H),4.54(t,J=5.8Hz,1H),4.42(t,J=5.8Hz,1H),3.85(br s,2H),3.55(br d,J=6.3Hz,1H),3.42-3.34(m,1H),3.30(br s,3H),2.99-2.83(m,1H),2.78-2.61(m,3H),1.89-1.68(m,2H),1.07(d,J=6.5Hz,3H)
实施例13和14:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸和((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氟 苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000128
步骤1:(R)-4-氟-N-(1-(3-甲氧苯基)丙烷-2-基)苯胺的合成
Figure PCTCN2021088291-appb-000129
将(R)-1-(3-甲氧苯基)丙烷-2-胺(500mg,2.42mmol)和1-溴-4-氟苯(508.37mg,2.91mmol)溶于甲苯(15mL),在氮气环境下将叔丁醇钠(465.30mg,4.84mmol)和2-二叔丁基膦-2′,4′,6′-三异丙基联苯(102.80mg,242.08μmol),三(二亚苄基丙酮)二钯(221.68mg,242.08μmol)加入该体系。反应液在80℃下搅拌反应16小时。TLC监测反应完成后。反应液用乙酸乙酯(10mL)和水(20mL)稀释,有机层用食盐水(20mL)洗涤三次,收集到的水相用乙酸乙酯(20mL)萃取两次,收集到的有机相用硫酸钠干燥后减压浓缩干,然后经过柱层析纯化(二氧化硅,石油醚/乙酸乙酯=100/10)得到(R)-4-氟-N-(1-(3-甲氧苯基)丙烷-2-基)苯胺(350.00mg)。
1H NMR(400MHz,METHANOL-d 4)δ7.19-7.13(m,1H),6.89-6.81(m,2H),6.78-6.70(m,3H),6.66-6.58(m,2H),3.76-3.70(m,3H),3.63(m,J=6.4,12.8Hz,1H),2.88(dd,J=5.0,13.3Hz,1H),2.56(dd,J=7.8,13.3Hz,1H),1.08(d,J=6.3Hz,3H)
步骤2:(R)-3-(2-((4-氟苯基)氨基)丙基)苯酚的合成
Figure PCTCN2021088291-appb-000130
将(R)-4-氟-N-(1-(3-甲氧苯基)丙烷-2-基)苯胺(420mg,1.62mmol)溶于二氯甲烷(10mL)中,该溶液冷却到0℃后,滴加入三溴化硼(811.51mg,3.24mmol,2eq),反应液于25℃下搅拌0.5小时。TLC(石油醚:乙酸乙酯=5:1)显示反应完成。反应液慢慢滴加入冰水中淬灭后,向溶液中加入碳酸钠溶液,将溶液pH调节到弱碱性。有机相用食盐水(10mL)洗涤三次,收集到的水相用二氯甲烷(20mL)萃取两次,收集到的有机相用无水硫酸镁干燥过滤,减压浓缩后将反应液过滤浓缩得到粗产物(R)-3-(2-((4-氟苯基)氨基)丙基)苯酚(200.00mg)。
1H NMR(400MHz,METHANOL-d 4)δ7.09(t,J=7.7Hz,1H),6.92(t,J=8.8Hz,2H),6.74(br dd,J=4.3,8.8Hz,2H),6.70-6.60(m,3H),3.67-3.60(m,1H),2.90(dd,J=4.8,13.3Hz,1H),2.51(dd,J=8.3,13.3Hz,1H),1.11(d,J=6.5Hz,3H)
步骤3:(1S,3R)-1-(4-溴-2,6-二氟苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000131
将(R)-3-(2-((4-氟苯基)氨基)丙基)苯酚(150mg,611.52μmol)溶于乙腈(15mL),加入4-溴-2,6-二氟苯(甲)醛(148.66mg,672.67μmol)和乙酸(367.23mg,6.12mmol)。反应液于100℃搅拌反应16个小时。LCMS监测反应完成后,将反应溶液冷却至室温,减压浓缩干后经过薄层层析纯化(二氧化硅,石油醚/乙酸乙酯=10/1)得到(1S,3R)-1-(4-溴-2,6-二氟苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(120.00mg)。
MS m/z(ESI):449.8[M+H] +
步骤4:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000132
将(1S,3R)-1-(4-溴-2,6-二氟苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(150.00mg,334.62μmol)溶于叔戊醇(5mL),加入1-(3-氟丙基)氮杂环丁烷-3-胺(97.00mg,401.54μmol),碳酸铯(654.15mg,2.01mmol),Brettphos Pd G3(30.33mg,33.46μmol)在氮气环境下加入该反应体系。反应液在120℃下搅拌反应16小时。LCMS监测反应完成,反应液减压浓缩干后,用乙酸乙酯稀释(5mL),然后用食盐水(15mL)洗涤三次,收集到的水相用乙酸乙酯(20mL)萃取两次,有机相用硫酸钠干燥后减压浓缩干,然后经薄层层析纯化(二氧化硅,石油醚/四氢呋喃=1/1,1%氨水)得(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(78.00mg)。
MS m/z(ESI):500.2[M+H] +
步骤5:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000133
(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(78mg,156.41μmol)溶于二氯甲烷(2mL),加入1,1,1-三氟-N-苯基-N-((三氟甲基)磺酰)甲磺酰胺(83.67mg,234.21μmol)和三乙胺(47.40mg,468.43μmol)。反应液于25℃搅拌反应16个小时。LCMS和TLC(石油醚:四氢呋喃=1:1)监测反应完成后,将反应溶液冷却至室温后,减压浓缩干,用二氯甲烷(1.5mL)稀释后经薄层层析纯化(二氧化硅,石油醚/四氢呋喃=1/1,0.5%氨水)得到(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(68.00mg)。
MS m/z(ESI):632.1[M+H] +
步骤6:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸和((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000134
将(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(68.00mg,107.66μmol)溶于二氧六环(3mL),加入5,5,5',5'-四甲基-2,2'-联(1,3,2-二噁硼己环)(48.64mg,215.32μmol),乙酸钾(31.70mg,322.99μmol),1,1-双(二苯基磷)二茂铁氯化钯(7.88mg,10.77μmol)在氮气环境下加入该反应体系中。反应液于90℃搅拌反应4个小时。LCMS检测反应完成后,将反应溶液冷却至室温后,减压浓缩后经制备液相色谱(Phenomenex Gemini-NX C18柱,3um二氧化硅,30mm直径,75mm长度);(使用水(含有0.225%甲酸)和乙腈的极性递减的混合物作为洗脱液),纯化得到第一个峰和第二个峰。
第一个峰经手性分离(DAICEL CHIRALPAK AD柱,5um二氧化硅,30mm直径,250mm长度);(使用乙醇(含有0.1%氨水)和水的极性递减的混合物作为洗脱液)得到((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸(实施例13,4.41mg)。
MS m/z(ESI):528.1[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.41(s,1H),7.30(d,J=6.6Hz,1H),7.14-7.07(m,2H),6.90(t,J=8.7Hz,2H),6.74-6.62(m,1H),5.94(d,J=11.1Hz,2H),5.52(s,1H),4.55(t,J=5.7Hz,1H),4.44(t,J=5.7Hz,1H),4.07(t,J=6.2Hz,1H),3.94(s,2H),3.51-3.41(m,1H),3.26-3.12(m,2H),3.03-2.89(m,2H),2.84(s,2H),1.89-1.76(m,2H),0.91(d,J=6.1Hz,3H)
第二个峰经过手性分离(DAICEL CHIRALCEL OD-H柱,5um二氧化硅,30mm直径,250mm长度);(使用乙醇(含有0.1%氨水)和水的极性递减的混合物作为洗脱液)和制备液相色谱纯化(Phenomenex Synergi C18柱,4um二氧化硅,30mm直径,150mm长度);(使用水(含有0.05%盐酸)和乙腈的极性递减的混合物作为洗脱液)纯化得到((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸(实施例14,1.54mg)。
MS m/z(ESI):528.3[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.70-7.49(m,2H),7.27(s,2H),7.16-7.02(m,2H),6.93(d,J=7.5Hz,1H),6.19-6.05(m,3H),4.62(d,J=5.5Hz,1H),4.51(t,J=5.4Hz,1H),4.47-4.33(m,3H),4.18(s,1H),3.98-3.89(m,1H),3.61(dd,J=5.0,16.8Hz,1H),3.49-3.39(m,3H),3.07(s,1H),2.08-1.95(m,2H),1.15(d,J=6.3Hz,3H)
实施例15和16:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸和((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000135
步骤1:(R)-2,4-二氟-N-(1-(3-甲氧苯基)丙烷-2-基)苯胺的合成
Figure PCTCN2021088291-appb-000136
将(R)-1-(3-甲氧苯基)丙烷-2-胺(0.5g,2.42mmol)和1-溴-2,4-二氟苯(560.63mg,2.91mmol)溶于甲苯(10mL)中,在25℃下加入三(二亚苄基丙酮)二钯(221.68mg,242.08μmol),叔丁醇钠(465.30mg,4.84mmol)和2-二叔丁基膦-2′,4′,6′-三异丙基联苯(102.80mg,242.08μmol),反应液在氮气保护下于80℃搅拌反应13小时。LCMS检测反应完成。将反应液倒入水中并搅拌10分钟,乙酸乙酯(20mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/乙酸乙酯=10/1)得到产物(R)-2,4-二氟-N-(1-(3-甲氧苯基)丙烷-2-基)苯胺(530mg)。
MS m/z(ESI):278.0[M+H] +
1H NMR(400MHz,CHLOROFORM-d)δ7.24(t,J=7.8Hz,1H),6.85-6.74(m,4H),6.73(d,J=1.8Hz,1H),6.72-6.63(m,1H),3.82(s,3H),3.73(td,J=6.7,13.6Hz,1H),3.67-3.56(m,1H),2.92(dd,J=5.0,13.4Hz,1H),2.71(dd,J=7.1,13.4Hz,1H),1.22-1.18(m,3H)
步骤2:(R)-3-(2-((2,4-二氟苯基)氨基)丙基)苯酚的合成
Figure PCTCN2021088291-appb-000137
将(R)-2,4-二氟-N-(1-(3-甲氧苯基)丙烷-2-基)苯胺(530mg,1.91mmol)溶于二氯甲烷(10mL)中,在0℃下加入三溴化硼(1.44g,5.73mmol),反应液于25℃搅拌反应1小时。TLC(石油醚:乙酸乙酯=10:1)检测反应完成。将反应液倒入水中并搅拌10分钟,用饱和碳酸氢钠中和至pH=8,二氯甲烷(40mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干,得到产物(R)-3-(2-((2,4-二氟苯基)氨基)丙基)苯酚(0.6g)。
MS m/z(ESI):263.9[M+H] +
步骤3:(1S,3R)-1-(4-溴-2,6-二氟苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000138
将(R)-3-(2-((2,4-二氟苯基)氨基)丙基)苯酚(350mg,1.06mmol)和4-溴-2,6-二氟苯甲醛(258.54mg,1.17mmol)溶于乙腈(6mL)中,加入乙酸(1.28g,21.27mmol),反应液100℃搅拌反应13小时。TLC(石油醚:乙酸乙酯=3:1)检测反应完成。将反应液减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/乙酸乙酯=3/1)得到产物(1S,3R)-1-(4-溴-2,6-二氟苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(330mg)。
MS m/z(ESI):466.0,468.0[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.19-7.07(m,1H),7.06-6.96(m,2H),6.89-6.72(m,2H),6.67-6.42(m, 3H),5.77(s,1H),3.58(br s,1H),3.02-2.87(m,1H),2.86-2.72(m,1H),0.93(d,J=6.1Hz,3H)
步骤4:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000139
将(1S,3R)-1-(4-溴-2,6-二氟苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(330mg,707.75μmol)和1-(3-氟丙基)氮杂环丁烷-3-胺盐酸盐溶于叔戊醇(6mL)中,加入碳酸铯(1.38g,4.25mmol)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)(64.16mg,70.78μmol),反应液在氮气保护下于120℃下搅拌反应2小时。LCMS检测反应完成。将反应液减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/四氢呋喃(1%氨水)=1/1)得到产物(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(275mg)。
MS m/z(ESI):518.2[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.06(br d,J=7.0Hz,1H),6.85-6.66(m,2H),6.64(d,J=8.5Hz,1H),6.60-6.40(m,2H),5.96-5.86(m,2H),5.83(s,1H),5.61(s,1H),4.57-4.48(m,1H),4.43-4.33(m,1H),4.03-3.89(m,1H),3.80-3.67(m,2H),3.63-3.50(m,1H),3.44-3.37(m,1H),2.98-2.86(m,2H),2.68-2.56(m,2H),1.83-1.67(m,2H),0.92(d,J=6.3Hz,3H)
步骤5:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000140
将(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(150mg,289.84μmol)和1,1,1-三氟-N-苯基-N-((三氟甲基)磺酰)甲磺酰胺(207.09mg,579.67μmol)溶于二氯甲烷(10mL)中,加入三乙胺(87.99mg,869.51μmol),反应液25℃搅拌反应13小时。LCMS检测反应完成。将反应液倒入水中并搅拌10分钟,二氯甲烷(40mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。然后柱层析纯化(二氧化硅,石油醚/四氢呋喃(1%氨水)=1/1)得到产物(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(280mg)。
MS m/z(ESI):650.1[M+H] +
步骤6:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸和((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000141
将(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(175.26mg,775.87μmol)和5,5,5',5'-四甲基-2,2'-联(1,3,2-二噁硼己环)(175.26mg,775.87μmol)溶于二甲基亚砜(3mL)中,加入[1,1-双(二苯基膦)二茂铁]二氯化钯二氯甲烷(31.68mg,38.79μmol)和乙酸钾(76.14mg,775.87μmol)。反应液90℃搅拌反应2小时。LCMS检测反应完成。将反应液冷却至室温并倒入水(20mL)中并搅拌10分钟,乙酸乙酯(40mL)萃取2次,有机相用无水硫酸钠干燥,过滤,减压浓缩至干。经制备液相色谱纯化(Phenomenex Gemini-NX C18柱:3um二氧化硅,30mm直径,75mm长度;使用水(含有0.225%乙酸)和乙腈的极性递减的混合物作为洗脱液)和制备手性分离(DAICEL CHIRALPAK IG柱:10um二氧化硅,30mm直径,250mm长度;使用乙醇(含有0.1%氨水)的极性一致作为洗脱液)纯化得到化合物((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸(第一个峰,实施例15,30mg)和((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸(第二个峰,实施例16,15mg)。
实施例15:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸。
MS m/z(ESI):546.1[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.57-7.19(m,2H),7.08(m,1H),6.82(m,1H),6.75-6.63(m,2H),5.94(d,J=11.3Hz,2H),5.71(s,1H),4.56(t,J=5.7Hz,1H),4.44(t,J=5.7Hz,1H),4.15-4.05(m,1H),4.05-3.93(m,2H),3.60(s,1H),3.30-3.20(m,2H),3.03-2.81(m,4H),1.92-1.77(m,2H),0.95(d,J=6.1Hz,3H)
实施例16:((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(2,4-二氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸
MS m/z(ESI):546.1[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.47-7.15(m,2H),7.14-7.03(m,1H),6.76-6.60(m,3H),5.87-5.76(m,3H),4.42(t,J=5.7Hz,1H),4.31(t,J=5.8Hz,1H),3.96-3.92(m,1H),3.86-3.71(m,3H),3.43-3.33(m,1H),3.13-3.00(m,2H),2.72(br t,J=7.3Hz,2H),2.60(dd,J=3.8,15.4Hz,1H),1.75-1.65(m,2H),0.90(d,J=6.4Hz,3H)
实施例17和18:((1S,3R)-2-(4-环丙基苯基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸和((1R,3R)-2-(4-环丙基苯基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000142
步骤1:(R)-4-环丙基-N-(1-(3-甲氧苯基)丙烷-2-基)苯胺的合成
Figure PCTCN2021088291-appb-000143
将(R)-1-(3-甲氧苯基)丙烷-2-胺(500mg,2.42mmol,80%纯度)和1-溴-4-环丙基苯(572.50mg,2.91mmol)溶于甲苯(15mL),加入叔丁醇钠(465.30mg,4.84mmol)和2-二叔丁基膦-2,4,6-三异丙基联苯(102.80mg,242.08μmol),三(二亚苄基丙酮)二钯(221.68mg,242.08μmol)在氮气环境下加入该体系。反应液在80℃下搅拌反应16小时。TLC监测反应完成后。反应液用乙酸乙酯(30mL,三次)和水(30mL)萃取,收集到的有机相用硫酸钠干燥后减压浓缩干,然后经过柱层析纯化(二氧化硅,石油醚/乙酸乙酯=16/1)得到(R)-4-环丙基-N-(1-(3-甲氧苯基)丙烷-2-基)苯胺(350.00mg)。
1H NMR(400MHz,METHANOL-d 4)δ7.22-7.13(m,1H),6.92-6.86(m,2H),6.83-6.69(m,3H),6.60(d,J=8.5Hz,2H),3.79-3.72(m,3H),3.69-3.60(m,1H),2.90(dd,J=4.8,13.3Hz,1H),2.55(dd,J=7.9,13.2Hz,1H),1.83-1.73(m,1H),1.08(d,J=6.4Hz,3H),0.86-0.78(m,2H),0.58-0.50(m,2H).
步骤2:(R)-3-(2-((4-环丙基苯基)氨基)丙基)苯酚的合成
Figure PCTCN2021088291-appb-000144
将(R)-4-环丙基-N-(1-(3-甲氧苯基)丙烷-2-基)苯胺(340mg,1.21mmol)溶于二氯甲烷(10mL)中,该溶液冷却到0℃后,滴加入三溴化硼(605.41mg,2.42mmol),反应液于25℃下搅拌1小时。LCMS显示反应完成。反应液慢慢滴加入冰水中淬灭后,向溶液中加入碳酸钠溶液,将溶液pH调节到8。有机相用食盐水(10mL)洗涤三次,收集到的水相用二氯甲烷(20mL)萃取两次,收集到的有机相用无水硫酸钠干燥过滤,减压浓缩后得到粗产物(R)-3-(2-((4-环丙基苯基)氨基)丙基)苯酚(320.00mg,70%纯度)。
MS m/z(ESI):268.1[M+H] +
步骤3:(1S,3R)-1-(4-溴-2,6-二氟苯基)-2-(4-环丙基苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000145
将(R)-3-(2-((4-环丙基苯基)氨基)丙基)苯酚(320mg,789.93μmol)和4-溴-2,6-二氟苯甲醛(192.03mg,868.93μmol)溶于乙腈(5mL),加入乙酸(474.37mg,7.90mmol)。反应液于100℃搅拌反应16个小时。LCMS监测反应完成后,将反应溶液冷却至室温,减压浓缩干后经过薄层层析纯化(二氧化硅,石油醚/乙酸乙酯=10/1)得到(1S,3R)-1-(4-溴-2,6-二氟苯基)-2-(4-环丙基苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(204mg)。
MS m/z(ESI):470.2/472.2[M+H] +
步骤4:(1S,3R)-2-(4-环丙基苯基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000146
将(1S,3R)-1-(4-溴-2,6-二氟苯基)-2-(4-环丙基苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(200mg,425.22μmol)和1-(3-氟丙基)氮杂环丁胺-3-胺(123.26mg,510.26μmol)溶于叔戊醇(8mL),加入碳酸铯(692.72mg,2.13mmol),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)(38.55mg,42.52μmol)在氮气环境下加入该反应体系。反应液在120℃下搅拌反应2小时。LCMS监测反应完成,反应液减压浓缩干后,用乙酸乙酯(10mL*3)和水(10mL)萃取,有机相用硫酸钠干燥后减压浓缩干,然后经薄层层析纯化(二氧化硅,石油醚/四氢呋喃=1/1,1%氨水)得(1S,3R)-2-(4-环丙基苯基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(200mg)。
MS m/z(ESI):522.3[M+H] +
步骤5:(1S,3R)-2-(4-环丙基苯基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000147
将(1S,3R)-2-(4-环丙基苯基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(150mg,287.57μmol)溶于二氯甲烷(2mL),加入1,1,1-三氟-N-苯基-N-((三氟甲基)磺酰)甲磺酰胺(205.47mg,575.14μmol)和三乙胺(87.30mg,862.71μmol)。反应液于25℃搅拌反应16个小时。LCMS监测反应完成后,将反应溶液用水(4ml*2)洗涤后,减压浓缩干,经薄层层析纯化(二氧化硅,石油醚/四氢呋喃=1/1,0.5%氨水)得到(1S,3R)-2-(4-环丙基苯基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(200mg)。
MS m/z(ESI):654.1[M+H] +
步骤6:((1S,3R)-2-(4-环丙基苯基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸和((1R,3R)-2-(4-环丙基苯基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000148
将(1S,3R)-2-(4-环丙基苯基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(180mg,275.37μmol)和5,5,5',5'-四甲基-2,2'-联(1,3,2-二噁硼己环)(124.40mg,550.73μmol)溶于二氧六环(4mL),加入乙酸钾(81.07mg,826.10μmol),1,1-双(二苯基磷)二茂铁氯化钯(20.15mg,27.54μmol)在氮气环境下加入该反应体系中。反应液于90℃搅拌反应2个小时。LCMS检测反应完成后,将反应溶液冷却至室温后,减压浓缩后经过制备液相色谱(Phenomenex Gemini-NX C18柱,3um二氧化硅,30mm直径,75mm长度);(使用水(含有0.225%甲酸)和乙腈的极性递减的混合物作为洗脱液),纯化得到((1S,3R)-2-(4-环丙基苯基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸(第一个峰,
实施例17,13.93mg)和((1R,3R)-2-(4-环丙基苯基)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸(第二个峰,实施例18,16.23mg)。
实施例17:
MS m/z(ESI):550.3[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.54-7.24(m,2H),6.99-6.92(m,2H),6.89-6.81(m,2H),6.64(br d,J=7.5Hz,1H),5.92(d,J=11.0Hz,2H),5.53(s,1H),4.55(t,J=5.6Hz,1H),4.43(t,J=5.6Hz,1H),4.18-4.00(m,3H),3.53-3.35(m,3H),3.00-2.85(m,4H),1.94-1.75(m,3H),0.93-0.85(m,5H),0.64-0.52(m,2H).
实施例18:
MS m/z(ESI):550.3[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.52-7.30(m,2H),7.00-6.82(m,5H),5.96(d,J=11.5Hz,2H),5.89(s,1H),4.53(t,J=5.6Hz,1H),4.41(t,J=5.6Hz,1H),4.22(br s,1H),4.13-3.97(m,3H),3.56(br dd,J=4.6,15.2Hz,1H),3.39-3.32(m,2H),2.92(t,J=7.5Hz,2H),2.71(dd,J=2.8,15.3Hz,1H),1.93-1.71(m,3H),0.91(d,J=6.4Hz,3H),0.88-0.80(m,2H),0.60-0.50(m,2H).
实施例19和20:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸和((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000149
步骤1:(R)-4-氯-N-(1-(3-甲氧苯基)丙烷-2-基)苯胺的合成
Figure PCTCN2021088291-appb-000150
将(R)-1-(3-甲氧苯基)丙烷-2-胺(500mg,2.42mmol,80%纯度,1eq),1-溴-4-氯苯(556.17mg,2.91mmol,1.2eq),叔丁醇钠(465.29mg,4.84mmol),2-二叔丁基膦-2’,4’,6’-三异丙基联苯(102.80mg,242.08μmol,0.1eq)和三(二亚苄基丙酮)二钯(221.68mg,242.08μmol,0.1eq)溶于甲苯(15mL),用氮气排三次空气。反应液在80℃下搅拌反应16小时。TLC(石油醚:乙酸乙酯=3:1)监测反应完成后。反应液用乙酸乙酯(30mL)和水(15mL)稀释,过滤后滤液用乙酸乙酯(20mL)萃取三次,收集到的有机相用无水硫酸钠干燥后过滤减压浓缩干,然后经过柱层析纯化(二氧化硅,石油醚/乙酸乙酯=100/35)得到(R)-4-氯-N-(1-(3-甲氧苯基)丙烷-2-基)苯胺(400.00mg)。
步骤2:(R)-3-(2-((4-氯苯基)氨基)丙基)苯酚的合成
Figure PCTCN2021088291-appb-000151
将(R)-4-氯-N-(1-(3-甲氧苯基)丙烷-2-基)苯胺(480mg,1.74mmol,1eq)溶于二氯甲烷(10mL)中,该溶液冷却到0℃后,滴加入三溴化硼(1.31g,5.22mmol,3eq),反应液于25℃下搅拌2小时。TLC(石油醚:乙酸乙酯=3:1)显示反应完成。反应液慢慢滴加入冰水(20mL)中淬灭后,向溶液中滴加饱和碳酸钠溶液,将溶液pH调节到弱碱性。混合溶液用二氯甲烷(50mL)萃取三次,收集到的有机相用无水硫酸钠干燥过滤,减压浓缩后得到粗产物(R)-3-(2-((4-氯苯基)氨基)丙基)苯酚(420.00mg)。
1H NMR(400MHz,METHANOL-d 4)δ7.42(br d,J=8.5Hz,2H),7.18(br d,J=8.3Hz,2H),7.12(t,J=7.8Hz,1H),6.72-6.62(m,3H),3.85-3.76(m,1H),3.01(dd,J=4.8,13.3Hz,1H),2.64(dd,J=9.3,13.3Hz,1H),1.18(d,J=6.5Hz,3H).
步骤3:(1S,3R)-1-(4-溴-2,6-二氟苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000152
将(R)-3-(2-((4-氯苯基)氨基)丙基)苯酚(370mg,1.41mmol,1eq)溶于乙腈(12mL),加入4-溴-2,6-二氟苯甲醛(328.02mg,1.48mol,1.05eq)和乙酸(3mL)。反应液于100℃搅拌反应16小时。LCMS监测反应完成后,将反应溶液冷却至室温,减压浓缩干后经过柱层析纯化(二氧化硅,石油醚/乙酸乙酯=100/25)得到(1S,3R)-1-(4-溴-2,6-二氟苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(275.00mg)。
MS m/z(ESI):466.0[M+H] +
步骤4:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚的合成
Figure PCTCN2021088291-appb-000153
将(1S,3R)-1-(4-溴-2,6-二氟苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(275mg,591.74μmol,1eq)溶于叔戊醇(15mL),加入1-(3-氟丙基)氮杂环丁烷-3-胺(171.53mg,710.09μmol,1.2eq),碳酸铯(1.16g,3.55mmol,6eq),Brettphos Pd G3(53.64mg,59.17μmol,0.1eq)在氮气环境下加入该反应体系。反应液在120℃下搅拌反应3小时。LCMS监测反应完成,反应液用水(10mL)稀释后,用乙酸乙酯(20mL)萃取三次,收集到的有机相用无水硫酸钠干燥后过滤减压浓缩干,然后经薄层层析纯化(二氧化硅,石油醚/四氢呋喃=1/1,1%氨甲醇(7mol/L))得(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(135.00mg)。
MS m/z(ESI):516.1[M+H] +
步骤5:(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氟苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯的合成
Figure PCTCN2021088291-appb-000154
将(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-酚(135mg,261.63μmol,1eq)溶于二氯甲烷(10mL),加入1,1,1-三氟-N-苯基-N-((三氟甲基)磺酰)甲磺酰胺(186.93mg,523.26μmol,2eq)和三乙胺(79.42mg,784.89μmol,3eq)。反应液于25℃搅拌反应16个小时。LCMS和TLC(石油醚:四氢呋喃=1:1)监测反应完成后,将反应溶液冷却至室温后,减压浓缩干经薄层层析纯化(二氧化硅,石油醚/四氢呋喃=1/1)得到(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(135.00mg)。
MS m/z(ESI):648.1[M+H] +
步骤6:((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸和((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸的合成
Figure PCTCN2021088291-appb-000155
将(1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基三氟甲磺酸酯(105.00mg,162.02μmol,1eq)溶于二氧六环(4mL),加入5,5,5',5'-四甲基-2,2'-联(1,3,2-二噁硼己环)(73.20mg,324.04μmol,2eq),乙酸钾(47.70mg,486.07μmol,3eq),1,1-双(二苯基磷)二茂铁氯化钯(11.86mg,16.20μmol,0.1eq)在氮气环境下加入该反应体系中。反应液于90℃搅拌反应3个小时。LCMS检测反应完成后,将反应溶液冷却至室温后,减压浓缩后经制备液相色谱(Phenomenex Synergi C18柱,4um二氧化硅,30mm直径,150mm长度);(使用水(含有0.225%甲酸)和乙腈的极性递减的混合物作为洗脱液)产物(35mg)。然后经手性分离(DAICEL CHIRALPAK AD柱,10um二氧化硅,30mm直径,250mm长度);(使用乙醇(含有0.1%氨水)和甲醇的极性递减的混合物作为洗脱液)得到((1S,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸(第一个峰,实施例19,6.66mg)和((1R,3R)-1-(2,6-二氟-4-((1-(3-氟丙基)氮杂环丁烷-3-基)氨基)苯基)-2-(4-氯苯基)-3-甲基-1,2,3,4-四氢异喹啉-6-基)硼酸(第二个峰,实施例20,9.99mg)。
实施例19:
MS m/z(ESI):544.2[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.58-7.25(m,2H),7.21-7.11(m,2H),7.09-6.96(m,2H),6.69(br d,J=7.0Hz,1H),5.96(d,J=11.3Hz,2H),5.57(s,1H),4.55(t,J=5.6Hz,1H),4.43(t,J=5.8Hz,1H),4.12-4.01(m,1H),3.93(br s,2H),3.55(br d,J=6.8Hz,1H),3.19(br s,2H),3.04-2.91(m,2H),2.83(br s,2H),1.92-1.75(m,2H),0.98-0.92(m,3H)
实施例20:
MS m/z(ESI):544.3[M+H] +
1H NMR(400MHz,METHANOL-d 4)δ7.67-7.28(m,3H),7.15-7.09(m,2H),7.01(br d,J=9.0Hz,3H),6.04(d,J=11.8Hz,2H),5.93(s,1H),4.58(t,J=5.5Hz,1H),4.46(t,J=5.5Hz,1H),4.37(br s,1H),4.24(br s,3H),3.68-3.57(m,3H),3.16(br s,2H),2.76(br d,J=17.6Hz,1H),2.01-1.86(m,2H),0.96(d,J=6.5Hz,3H)
生物学活性及相关性质测试例
测试例1:本发明化合物对MCF7细胞内雌激素受体降解效果检测
1.实验目的:
本实验的目的是测定本发明化合物对MCF7细胞内内源表达的雌激素受体的降解活性,根据IC50及最大降解效率评价化合物的活性。
2.实验方法:
MCF7细胞(ATCC,HTB-22)用含10%胎牛血清的DMEM(Gibco,11995-065)完全培养基进行培养。实验第一天,使用完全培养基将MCF7细胞以3000个/孔的密度种于384孔板,37℃,5%CO 2细胞培养箱培养。待测化合物溶解于DMSO,储存浓度为10mM,用Echo 550(Labcyte Inc.)稀释并加入细胞培养板内,各化合物处理的起始浓度为100nM,3倍梯度稀释,9个浓度点,设置含0.5%DMSO的空白对照,各浓度点设双复孔对照。37℃,5%CO 2细胞培养箱培养24小时。各细胞培养孔内加入多聚甲醛至细胞培养液内,终浓度约3.7%固定细胞,作用30分钟后,弃上清,加入50μL PBS每孔洗涤一次;加入PBS含0.5%v/v Tween-20处理细胞30分钟,PBS洗涤一次;加入封闭液(PBS内含5%BSA,0.05%Tween-20)室温孵育1小时;去封闭液加入一抗混合液(抗-ER单抗,Estrogen Receptorα(D8H8)Rabbit mAb,GST,#8644S,1:1000稀释;抗-GAPDH单抗,GAPDH(D4C6R)Mouse mAb,GST,#97166S,1:2000稀释)室温孵育3小时;用PBST(PBS内含0.05%Tween-20)洗涤3次;加入检测二抗(800CW-羊抗兔IgG,LI-COR,P/N:926-32211,1:1000稀释;680RD-羊抗鼠IgG,LI-COR,#925-68070,1:1000稀释),室温,避光孵 育45分钟;PBST洗涤3次,使用Odyssey CLx读取各孔荧光信号。数据处理用XLfit,根据化合物的浓度和荧光信号值计算各化合物的抑制活性IC 50及最大抑制率Imax,结果见下表1。
表1本发明化合物对MCF7细胞内雌激素受体的抑制活性及最大抑制率
化合物编号 ER level IC 50(nM) 最大抑制率
实施例1 0.25 98%
实施例2 0.13 97%
实施例3 0.08 98%
实施例4 0.19 100%
实施例5 0.45 96%
实施例6 1.85 96%
实施例7 0.68 98%
实施例8 1.04 85%
实施例9 0.64 98%
实施例10 0.51 92%
实施例11 0.29 97%
实施例12 0.14 97%
实施例13 0.39 92%
实施例14 0.24 99%
实施例15 1.28 95%
实施例16 0.56 99%
实施例17 1.57 98%
实施例18 0.5 96%
实施例19 1.86 96%
实施例20 1.64 94%
测试例2:本发明化合物对MCF7细胞增殖的抑制效果检测
1.实验目的:
本实验的目的是测定本发明化合物对MCF7细胞体外增殖的抑制影响,根据IC 50及最大抑制效率评价化合物的活性。
2.实验方法:
MCF7细胞(ATCC,HTB-22)用含10%胎牛血清的DMEM(Gibco,11995-065)完全培养基进行培养。实验第一天,使用完全培养基将MCF7细胞以500个/孔的密度种于384孔板,37℃,5%CO 2细胞培养箱过夜培养。第二天,加入待测化合物进行药物处理,采用Echo550(Labcyte Inc.)将储存浓度为10mM的化合物溶液进行稀释及转移至各细胞培养孔内,各化合物在细胞内的处理起始浓度为100nM,3倍梯度稀释,9个浓度点,设置含0.3%DMSO的空白对照,各浓度点设双复孔对照。37℃,5%CO 2细胞培养箱培养7天,第八天,取出细胞培养板。加入
Figure PCTCN2021088291-appb-000156
Luminescent Cell Viability Assay(Promega,G7573),室温放置10分钟后,使用多标记酶标仪EnVision(PerkinElmer)读取发光信号值,用XLfit根据化合物的浓度和发光信号值计算各化合物的抑制活性IC 50及最大抑制率Imax,结果见下表2。
表2本发明化合物对MCF7细胞增殖的抑制活性及最大抑制率
化合物编号 MCF-7细胞增殖抑制IC 50(nM) 最大抑制率
实施例1 0.38 98%
实施例2 0.61 98%
实施例3 0.29 103%
实施例4 0.51 99%
实施例5 2.02 95%
实施例7 2.14 95%
实施例9 1.5 98%
实施例11 0.53 96%
测试实施例3、本发明化合物在肝微粒体中的代谢稳定性测定
一、试验材料及仪器
1.肝微粒体来源:人肝微粒体(Corning 452117),CD-1小鼠肝微粒体(XENOTECH M1000)
2.NADPH(Solarbio 1216C022)
3.阳性对照化合物维拉帕米(Sigma MKBV4993V)
4.AB Sciex Triple Quad 4000液质联用仪
二、试验步骤
1. 100mM磷酸缓冲液(PBS)的配制:称取7.098g Na 2HPO 4,加入500mL纯水超声溶解,作为溶液A。称取3.400g KH 2PO 4,加入250mL纯水超声溶解,作为溶液B。将A溶液放置在搅拌器上缓慢加入B溶液直到pH值达到7.4配制成100mM的PBS缓冲液。
2.反应体系的配制
按下表配制反应体系
Figure PCTCN2021088291-appb-000157
3.将反应体系置于37℃水浴中预孵育10分钟。向反应体系中加入40μL 10mM NADPH溶液(NADPH由100mM的磷酸缓冲液溶解),NADPH的最终浓度为1mM。用40μL磷酸缓冲液代替NADPH溶液作为阴性对照。阴性对照的作用是排除化合物自身化学稳定性的影响。
4.在反应体系中加入4μL 100μM的本发明化合物和阳性对照化合物维拉帕米启动反应,化合物的最终浓度为1μM。
5.在0.5、15、30、45和60分钟,涡旋振荡器充分混匀后,分别取出50μL孵育样品,用4倍的含有内标的冰乙腈终止反应。样品在3,220g转速下离心45分钟。离心结束后转移90μL上清液到进样板,加入90μL超纯水混匀,用于LC-MS/MS分析。
所有的数据均通过Microsoft Excel软件进行计算。通过提取离子图谱检测峰面积。通过对母药消除百分比的自然对数与时间进行线性拟合,检测母药的体外半衰期(t 1/2)。
体外半衰期(t 1/2)通过斜率计算:
in vitro t 1/2=0.693/k
经上述公式计算得到的t 1/2值见表3。
表3本发明化合物肝微粒体中的半衰期值
Figure PCTCN2021088291-appb-000158
Figure PCTCN2021088291-appb-000159
测试实施例4、本发明化合物的血浆蛋白结合率测定
一、试验材料及仪器
1.CD-1小鼠血浆(BioIVT)
2. 96孔平衡透析板(HTDialysis LLC,Gales Ferry,CT,HTD96B),平衡透析膜(MWCO 12-14K,#1101)
3.阳性对照化合物华法林
4.ABI QTrap 5500液质联用仪
二、试验步骤
1.浓度为100mM磷酸钠盐和150mM NaCl的缓冲液的配制:用超纯水配制浓度为14.2g/L Na 2HPO 4和8.77g/L NaCl的碱性溶液,用超纯水配制浓度为12.0g/L NaH 2PO 4和8.77g/L NaCl的酸性溶液。用酸性溶液滴定碱性溶液至pH值为7.4配制成浓度为100mM磷酸钠盐和150mM NaCl的缓冲液。
2.透析膜的准备:将透析膜浸泡在超纯水中60分钟以便将膜分离成两片,然后用20%乙醇浸泡20分钟,最后用透析所用缓冲液浸泡20分钟。
3.血浆的准备:将冷冻的血浆迅速在室温下解冻,然后将血浆在4℃、3,220g下离心10分钟去除凝块,并将上清收集到新的离心管中。测定和记录血浆的pH值,使用pH值为7-8的血浆。
3.含化合物的血浆样品的配制:用DMSO稀释10mM的本发明化合物或阳性对照化合物的储备液得到200μM的工作液。597μl小鼠血浆中加入3μl 200μM的化合物工作液得到终浓度为1μM的血浆样品。
4.平衡透析步骤:按照操作说明将透析装置组装起来。在透析膜的一侧加入120μL含1μM化合物的血浆样品,另一侧加入等体积的透析液(磷酸盐缓冲液)。试验设双样本。封上透析板,放入孵育装置,在37℃、5%CO 2及约100rpm转速下孵育6小时。孵育结束后,去除封膜,从每个孔的缓冲液和血浆侧吸取50μl到新板的不同孔中。在磷酸盐缓冲液样品中加入50μl空白血浆,在血浆样品中加入等体积的空白磷酸盐缓冲液,然后加入300μl含内标的乙腈沉淀蛋白。涡旋5分钟,在4℃、3,220g下离心30分钟。取100μl上清液至进样板,加入100μL超纯水混匀,用于LC-MS/MS分析。
测定化合物在缓冲液侧和血浆侧的峰面积。计算化合物的血浆蛋白结合率公式如下:
%游离率=(化合物峰面积与内标峰面积比值缓冲液侧/化合物峰面积与内标峰面积比值血浆侧)*100%
%结合率=1-%游离率
所有的数据均通过Microsoft Excel软件进行计算。计算得到的本发明化合物的血浆蛋白结合率值见表4。
表4本发明化合物在CD-1小鼠血浆中的蛋白结合率值
实施例编号 %结合率
实施例1 99.8%
实施例2 99.9%
实施例3 98%
实施例4 97%
测试例5、本发明化合物的膜渗透性及转运特性测定
本发明化合物的膜渗透性及转运特性采用如下试验方法测定。
一、试验材料及仪器
1.Caco-2细胞(ATCC)
2.HEPES(Solarbio 804D049)、青霉素/链霉素(Solarbio 20200109)和Trypsin/EDTA(Solarbio)、PBS(Solarbio 20200620)
3.胎牛血清(FBS)(Sigma WXBD0055V)、荧光黄(Sigma MKCJ3738)、NaHCO 3(Sigma SLBZ4647)
4.Hank’s平衡盐溶液(Gibco 2085528)和非必需氨基酸(NEAA)(Gibco 2211548)、Trypsin/EDTA(Gibco 2120732)
5.高糖DMEM(Corning 20319014)
6.HTS Transwell-96 Well Permeable(Corning,3391)
7.电阻检测仪(Millipore,
Figure PCTCN2021088291-appb-000160
ERS-2)
8.
Figure PCTCN2021088291-appb-000161
Vision(Nexcelom Bioscience)
9.Infinite 200 PRO酶标仪(Tecan,Infinite M200PRO)
10.阳性对照化合物Metoprolol(Sinopharm 100084-201403)、Erythromycin(MCE 84550)和Cimetidine(Sinopharm 100158-201406)
11.ABI QTrap 5500液质联用仪
二、试验步骤
1.Caco-2细胞培养
1)转运缓冲液(含25mM HEPES的HBSS,pH 7.4)的配制:精确称量5.958g HEPES和0.35g NaHCO 3,加900mL纯水让其溶解,然后加100mL 10×HBSS搅拌均匀,调pH至7.4,过滤。
2)Caco-2细胞培养基的配制:高糖DMEM(含有L-谷氨酰胺)培养基中加入FBS、青霉素、链霉素、卡那霉素和NEAA配制成含10%FBS、0.1mg/mL链霉素、100单位青霉素、0.6μg/mL卡那霉素和1×NEAA的细胞培养基。
3)在37℃、5%CO 2的培养箱中用T-75培养瓶培养细胞,细胞生长达到80-90%密度时弃去培养基。用5mL PBS冲洗细胞,加入1.5mL Trypsin/EDTA,然后在37℃培养箱中孵育5-10分钟直至细胞呈流沙状脱落,最后用含FBS的培养基中和Trypsin/EDTA。
4)细胞混悬液在120x g下离心10分钟,弃去上清液。
5)加细胞培养基重悬细胞,调至密度为6.86×10 5cells/mL的细胞悬浮液。
2.Caco-2细胞接种
1)Transwell小室每孔加入50μL培养基,下层加入25mL培养基,置于37℃,5%CO 2培养箱中预热1小时。
2)预热的Transwell小室每孔加入50μL细胞悬浮液,最终接种密度为2.4×10 5cells/cm 2
3)培养14-18天,隔一天换一次培养基,在最初种板以后的48小时之内更换培养基。实验前一天培养基必须更换。
3.评估单层细胞膜完整性
1)细胞培养14天后融合并且分化,准备进行转运实验。
2)用电阻仪测量单层膜电阻,记录每孔电阻。
3)测量完毕后,将Transwell培养板重新孵育
4)计算TEER值:
TEER值=TEER(Ω)测量值×膜面积(cm 2)
单层细胞膜的电阻<230Ω·cm 2,指示单层细胞膜致密性差,不能用于试验。
4.转运实验
1)用DMSO稀释10mM的本发明化合物或阳性对照化合物的储备液得到2mM的储备液,然后用转运缓冲液稀释2mM的储备液得到10μM的本发明化合物或阳性对照化合物的工作液。
2)从培养箱中取出Caco-2细胞板,然后用预热的转运缓冲液清洗Transwell培养板两次,再置于37℃培养箱孵育30分钟。
3)为测定化合物从顶端到基底端(A→B)的转运速率,加108μL化合物的工作液到Transwell小室(顶端),同时立即从顶端取出8μL样品至72μL转运缓冲液中,加入240μL含内标的终止液终止转运以作为初始顶端样品。同时,接收端(基底端)加入300μL转运缓冲液。试验设双样本。
4)为测定化合物从基底端到顶端(B→A)的转运速率,加308μL化合物的工作液到接收端(基底端),同时立即从基底端取出8μL样品至72μL转运缓冲液中,加入240μL含内标的终止液终止转运以作为初始基底端样品。同时,Transwell小室(顶端)加入300μL转运缓冲液。试验设双样本。
5)将细胞培养板置于37℃ CO 2培养箱中孵育2小时。
6)转运实验结束后,从给药端(即A→B方向的顶端和B→A方向的基底端)取8μL样品至72μL转运缓冲液中,然后加入240μL含内标的终止液终止转运。从接收端(即A→B方向的基底端和B→A方向的顶端)取80μL样品至240μL含内标的终止液中,1000rpm下涡旋10分钟,3,220g下离心30分钟。取100μl上清液至进样板,加入100μL超纯水混匀,用于LC-MS/MS分析。
7)转运实验结束后测量荧光值,用水配制10mM荧光黄储备液,然后用转运缓冲溶液稀释至100μM。往Transwell小室(顶端)中加入100μL荧光黄溶液,基底端加入300μL转运缓冲溶液,置于37℃的CO 2培 养箱中孵育30分钟。从基底端取80μL溶液至96孔板中,在激发波长为485nm及发射波长为530nm下用酶标仪测量细胞荧光值(检测膜完整性)。
用以下公式计算Caco-2细胞单层膜的荧光值:
LY Leakage={I acceptor×0.3/(I acceptor×0.3+I donor×0.1)}×100%
I acceptor指接收侧(0.3mL)的荧光密度,I donor指给药侧(0.1mL)的荧光密度。LY>1.0%表示单层细胞膜致密性差,相应的结果将从评估中排除。
测定化合物在给药侧和接收侧的峰面积。计算化合物的表观渗透系数(P app,单位:cm/s)和外排比(Efflux ratio,ER):
P app={V A×[drug] acceptor/(Area×incubation time×[drug] initial dono}
V A为接收端溶液的体积(A→B是0.3mL,B→A是0.1mL),Area为Transwell-96孔板膜面积(0.143cm 2);incubation time为孵育时间(单位:s)。
Figure PCTCN2021088291-appb-000162
P app(B-A)为由基底端到顶端的表观渗透系数;P app(A-B)为由顶端到基底端的表观渗透系数。
计算得到的本发明化合物的表观渗透系数和外排比值见表5。
表5本发明化合物的表观渗透系数和外排比值
Figure PCTCN2021088291-appb-000163
测试例6、本发明化合物对CYP2C9、CYP2D6、CYP3A4酶活性的抑制作用
本发明化合物对CYP2C9、CYP2D6、CYP3A4酶活性的抑制采用如下试验方法测定。
一、试验材料及仪器
1.人肝微粒体(Corning 452117)
2.Na 2HPO 4(Sigma SLBZ6180)
3.KH 2PO 4(Sigma SLBT6559)
4.NADPH(Solarbio 705Y021)
5.NADPH(Solarbio 705Y021)
6.阳性底物双氯芬酸(Sigma SLBV3438)、右美沙芬(TRC 3-EDO-175-1)和咪达唑仑(Cerilliant FE01161704)
7.阳性抑制剂磺胺苯吡唑(D.Ehrenstorfer GmbH 109012)、奎尼丁(TCI WEODL-RE)和酮康唑(Sigma 100M1091V)
7.AB Sciex Triple Quad 5500液质联用仪
二、试验步骤
1. 100mM磷酸缓冲液(PBS)的配制:称取7.098g Na 2HPO 4,加入500mL纯水超声溶解,作为溶液A。称取3.400g KH 2PO 4,加入250mL纯水超声溶解,作为溶液B。将A溶液放置在搅拌器上缓慢加入B溶液直到pH值达到7.4配制成100mM的PBS缓冲液。
2.用100mM的PBS缓冲液配制10mM的NADPH溶液。用DMSO稀释10mM的本发明化合物储备液得到200×浓度的化合物工作液(6000、2000、600、200、60、20、0μM)。用DMSO稀释阳性抑制剂储备液得到200×浓度的阳性抑制剂工作液(磺胺苯吡唑,1000、300、100、30、10、3、0μM;奎尼丁/酮康唑,100、30、10、3、1、0.3、0μM)。用水、乙腈或乙腈/甲醇配制200×浓度的底物工作液(120μM双氯芬酸、400μM右美沙芬和200μM咪达唑仑)。
3.取2μl 20mg/ml的肝微粒体溶液、1μl底物工作液、1μl化合物工作液和176μl PBS缓冲液,混 合均匀,置于37℃水浴中预孵育15分钟。阳性对照组加入1μl双氯芬酸、右美沙芬或咪达唑仑工作液代替化合物工作液。同时将10mM的NADPH溶液一起在37℃水浴中预孵育15分钟。15分钟后,取20μl NADPH加入到各个孔中,启动反应,37℃下孵育5分钟(CYP2C9)、20分钟(CYP2D6)或5分钟(CYP3A4)。所有孵育样品设双样本。孵育相应时间后向所有样本中加入400ul含内标的冰甲醇终止反应。涡旋混匀,3220g、4℃离心40分钟。离心结束后转移100μL上清液到进样板,加入100μL超纯水混匀,用于LC-MS/MS分析。
数值经Excel XLfit 5.3.1.3计算得到本发明化合物对CYP2C9、CYP2D6和CYP3A4的IC 50值见表6。
表6本发明化合物对CYP2C9、CYP2D6和CYP3A4的IC 50
实施例编号 CYP2C9(μM) CYP2D6(μM) CYP3A4(μM)
实施例1 >30 10 3.8
实施例2 10.4 9.5 7.6
实施例3 8.9 25 24
实施例4 2.6 2.3 6.1
测试实施例7:药代动力学评价
Mouse药代动力学实验
实验材料:
CD-1小鼠购自北京维通利华实验动物技术有限公司。
DMSO(二甲基亚砜)、HP-β-CD(羟丙基-β-环糊精)、Tetraethylene Glycol(三缩四乙二醇),Captisol(SBE-β-CD,磺丁基-β-环糊精)购自Sigma。
乙腈购自Merck(USA)。
实验方法:
雌性CD-1小鼠6只(20-30g,4-6周),随机分成2组,每组3只。第1组尾静脉注射给予测试化合物,剂量为1mg/kg,溶媒5%DMSO in 10%HP-β-CD in water,第2组口服给予测试化合物,剂量10mg/kg,溶媒40%Tetraethylene Glycol(v/v),7.5%Captisol(w/v)in water。动物实验前正常喂食喂水。每组小鼠于给药前及给药后0.083(仅静脉注射组)、0.25、0.5、1、2、4、6、8和24h进行静脉采血。收集的全血样品置于K 2EDTA抗凝管中,离心5min后(12000rpm,4℃)取血浆待测。
取小鼠血浆样品10μL,加入150μL乙腈溶剂(其中含内标化合物)沉淀蛋白,涡旋5min后,离心(14000rpm)5min,上清液用含0.1%(v/v)FA的水稀释2倍,于LC-MS/MS系统(AB Sciex Triple Quad 6500+)进行定量检测。在测定样品浓度时随行CD-1小鼠血浆标准曲线和质控样品。对10x稀释样品,取2μL样品加入18μL的空白血浆,涡旋0.5min后,加入300μL乙腈溶剂(其中含内标化合物)沉淀蛋白,其余处理步骤同不稀释样品。
PK测试结果如下所示:
表7:本发明化合物小鼠PK
Figure PCTCN2021088291-appb-000164
测试实施例8:化合物对MCF-7异种移植瘤的生长抑制实验
实验试剂:
人乳腺癌MCF-7细胞:ECACC-86012803
17β-雌二醇片:Innovative Research of America,Cat No.:SE-121,60-day release,0.18mg/pellet
EMEM培养液:ATCC,Cat No.:30-2003
胎牛血清:Hyclone;Cat No.:SV30087.03
Antibiotic-Antimycotic:Gibco,Cat No.:15240-062
0.25%胰酶-EDTA:Gibco,Cat No.:25200-072
DPBS:Corning,Cat No.:21-031-CVR
基质胶:Corning,Cat No.:354234
实验方法:
动物信息:Balb/c裸小鼠,雌性,6-8周,体重约18-22克,动物购自上海灵畅生物科技有限公司,将小鼠饲养在SPF级的环境中,每个笼位单独送排风,所有动物都可以自由获取标准认证的商业实验室饮食和自由饮水。
细胞培养:人乳腺癌MCF-7细胞株体外培养,培养条件为EMEM(细胞培养液)中加入10%胎牛血清,1%Antibiotic-Antimycotic,37℃、5%CO 2孵箱。一周两次用0.25%胰酶-EDTA消化液进行常规消化处理传代。当细胞饱和度为80%-90%,数量达到要求时,收取细胞,计数。
细胞接种:将0.2ml/(含1×10 7)MCF-7细胞悬液(DPBS加基质胶,体积比为1:1)皮下接种于每只小鼠的右后背,并于细胞接种前两天皮下接种17β-雌二醇片。在接种细胞后第6天,依据肿瘤体积随机分组给药,分组当天为Day 0。
给药:阳性药Fulvestrant(氟维司群,阿斯利康)的给药剂量为250mg/kg,皮下注射给药(SC),每周一次给药(QW)x3周;化合物SCR-6139的给药剂量为10mg/kg,口服给药(PO),每天一次给药(QD)x3周或30mg/kg,PO,QDx3周。每组6只小鼠。
肿瘤测量和实验指标:
每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:V=0.5a x b 2,a和b分别表示肿瘤的长径和短径。每周两次测量小鼠体重。
化合物的抑瘤疗效用肿瘤生长抑制率TGI(%)来评价。TGI(%)=[(1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积)/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)]x100%。
实验结果:
见表8,在小鼠皮下移植瘤MCF-7模型中,本发明化合物实施例1和实施例3在30mg/kg一天一次口服给药对肿瘤生长具有显著抑制作用(P<0.01),具有缩小肿瘤的效果。实施例1和实施例3在所尝试剂量下未显著影响小鼠体重,也未引起任何小鼠死亡,小鼠可以耐受。
表8 MCF-7皮下瘤模型肿瘤体积
Figure PCTCN2021088291-appb-000165

Claims (19)

  1. 式(I)所示化合物或其药学上可接受的盐:
    Figure PCTCN2021088291-appb-100001
    其中,
    Figure PCTCN2021088291-appb-100002
    选自
    Figure PCTCN2021088291-appb-100003
    R 1、R 2独立选自OH,或者R 1、R 2以及它们所连接的硼原子共同形成5-6元杂环基,所述5-6元杂环基任选被C 1-C 10烷基取代;
    X 1、X 2、X 3、X 4独立地选自CR 7或N;
    R 7选自H、F、Cl、Br、I、OH、CN、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基或3-10元杂环基氧基;
    X、Y独立选自O或NH;
    R 3选自H、F、Cl、Br、I、OH、CN、C 1-C 10烷基、C 3-C 10环烷基、3-10元杂环基、C 1-C 10烷氧基、C 3-C 10环烷基氧基或3-10元杂环基氧基;
    R 4选自H、C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基;
    或者R 3、R 4以及它们所连接的原子共同形成4-10元杂环基,所述4-10元杂环基任选被R a1取代,所述R a1选自F、Cl、Br、I、OH、CN、C 1-C 10烷基或C 1-C 10烷氧基;
    R 5、R 6独立选自C 1-C 6烷基或苯基,所述C 1-C 6烷基或苯基任选被R a2取代;
    R 8选自H或C 1-C 6烷基,所述C 1-C 6烷基任选被R a2取代;
    R a2选自F、Cl、Br、I、OH、CN或任选被R b取代的以下基团:C 1-C 10烷基、C 3-C 10环烷基或3-10元杂环基;
    R b选自F、Cl、Br、I、OH、CN、C 1-C 10烷基或C 1-C 10烷氧基;
    条件是,式(I)所示化合物不包含
    Figure PCTCN2021088291-appb-100004
  2. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R 1、R 2独立选自OH,或者R 1、R 2以及它们所连接的硼原子共同形成5-6元杂环基,所述5-6元杂环基任选被甲基取代。
  3. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,X 1、X 2、X 3、X 4独立地选自CR 7或N,且X 1、X 2、X 3、X 4基团中的至少2个选自CR 7
  4. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R 7选自H、F、Cl、Br、I、CN、C 1-C 3烷基或C 1-C 3烷氧基。
  5. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R 3、R 4均选自H,或者R 3、R 4以及它们所连接的原子共同形成4-7元杂环基,所述4-7元杂环基选自4-6元单杂环基或6-7元螺杂环基,所述4-6元单杂环基或6-7元螺杂环基任选被R a1取代。
  6. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,所述R a1选自F、Cl、Br、I、CN、C 1-C 6烷基或C 1-C 6烷氧基。
  7. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R 5、R 6独立选自C 1-C 6烷基或苯基,所述C 1-C 6烷基或苯基任选被R a2取代。
  8. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,所述R a2选自F、Cl、Br、I、OH、CN或任选被R b取代的C 3-C 6环烷基。
  9. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,所述R b选自F、Cl、Br、I、OH、CN或C 1-C 6烷基。
  10. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R 3、R 4以及所连接的原子共同形成
    Figure PCTCN2021088291-appb-100005
  11. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R 5、R 6独立选自CH 2CF 3、CH 2CF 2CH 2OH、
    Figure PCTCN2021088291-appb-100006
    苯基、CH 2C(CH 3) 2F、对氟苯基、对氯苯基、
    Figure PCTCN2021088291-appb-100007
  12. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,所述式(I)所示化合物或其药学上可接受的盐选自式(II)所示化合物或其药学可接受的盐:
    Figure PCTCN2021088291-appb-100008
    其中
    Figure PCTCN2021088291-appb-100009
    选自
    Figure PCTCN2021088291-appb-100010
    R 1、R 2、R 3、R 4、R 5、R 6、R 8、X 1、X 2、X 3、X 4、X、Y如权利要求1所定义。
  13. 根据权利要求1至12任一项所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R 8选自H或C 1-C 3烷基。
  14. 根据权利要求1至12任一项所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,R 8选自H。
  15. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,所述式(I)所示化合物或其药学上可接受的盐选自以下化合物或其药学可接受的盐:
    Figure PCTCN2021088291-appb-100011
    Figure PCTCN2021088291-appb-100012
  16. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,所述式(I)所示化合物或其药学上可接受的盐选自以下化合物或其药学可接受的盐:
    Figure PCTCN2021088291-appb-100013
    Figure PCTCN2021088291-appb-100014
  17. 根据权利要求1所述的式(I)所示化合物或其药学上可接受的盐,其特征在于,所述式(I)所示化合物或其药学上可接受的盐选自以下化合物或其药学可接受的盐:
    Figure PCTCN2021088291-appb-100015
    Figure PCTCN2021088291-appb-100016
  18. 一种药物组合物,所述组合物包含权利要求1至17任一项的化合物或其药学上可接受的盐,以及药学上可接受的辅料。
  19. 权利要求1至17任一项的化合物或其药学上可接受的盐在制备预防或者治疗雌激素受体相关疾病的药物中的用途。
PCT/CN2021/088291 2020-04-21 2021-04-20 含硼化合物及其应用 WO2021213358A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202180027836.9A CN115667275B (zh) 2020-04-21 2021-04-20 含硼化合物及其应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010317185 2020-04-21
CN202010317185.4 2020-04-21

Publications (1)

Publication Number Publication Date
WO2021213358A1 true WO2021213358A1 (zh) 2021-10-28

Family

ID=78270780

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/088291 WO2021213358A1 (zh) 2020-04-21 2021-04-20 含硼化合物及其应用

Country Status (2)

Country Link
CN (1) CN115667275B (zh)
WO (1) WO2021213358A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024046837A1 (en) * 2022-08-31 2024-03-07 Cybin Irl Limited Tryptamine compounds, compositions, and methods of use

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107108611A (zh) * 2014-12-18 2017-08-29 豪夫迈·罗氏有限公司 四氢‑吡啶并[3,4‑b]吲哚雌激素受体调节剂及其用途
WO2018102261A2 (en) * 2016-11-29 2018-06-07 Xavier University Of Louisiana Boronic derivatives hydroxamates as anticancer agents
CN109219604A (zh) * 2016-04-08 2019-01-15 豪夫迈·罗氏有限公司 四氢异喹啉雌激素受体调节剂及其用途
CN109415388A (zh) * 2016-05-06 2019-03-01 路易斯安那泽维尔大学 选择性雌激素受体下调剂(serds)
WO2019228443A1 (en) * 2018-05-30 2019-12-05 Dizal (Jiangsu) Pharmaceutical Co., Ltd. Selective estrogen receptor downregulators and uses thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107108611A (zh) * 2014-12-18 2017-08-29 豪夫迈·罗氏有限公司 四氢‑吡啶并[3,4‑b]吲哚雌激素受体调节剂及其用途
CN109219604A (zh) * 2016-04-08 2019-01-15 豪夫迈·罗氏有限公司 四氢异喹啉雌激素受体调节剂及其用途
CN109415388A (zh) * 2016-05-06 2019-03-01 路易斯安那泽维尔大学 选择性雌激素受体下调剂(serds)
WO2018102261A2 (en) * 2016-11-29 2018-06-07 Xavier University Of Louisiana Boronic derivatives hydroxamates as anticancer agents
WO2019228443A1 (en) * 2018-05-30 2019-12-05 Dizal (Jiangsu) Pharmaceutical Co., Ltd. Selective estrogen receptor downregulators and uses thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LIU JIAWANG, ZHENG SHILONG, GUO SHANCHUN, ZHANG CHANGDE, ZHONG QIU, ZHANG QIANG, MA PENG, SKRIPNIKOVA ELENA V., BRATTON MELYSSA R.: "Rational Design of a Boron-Modified Triphenylethylene (GLL398) as an Oral Selective Estrogen Receptor Downregulator", ACS MEDICINAL CHEMISTRY LETTERS, vol. 8, no. 1, 12 January 2017 (2017-01-12), US , pages 102 - 106, XP055860947, ISSN: 1948-5875, DOI: 10.1021/acsmedchemlett.6b00410 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024046837A1 (en) * 2022-08-31 2024-03-07 Cybin Irl Limited Tryptamine compounds, compositions, and methods of use

Also Published As

Publication number Publication date
CN115667275A (zh) 2023-01-31
CN115667275B (zh) 2024-02-23

Similar Documents

Publication Publication Date Title
CN109219604B (zh) 四氢异喹啉雌激素受体调节剂及其用途
TWI699363B (zh) 四氫-吡啶并[3,4-b]吲哚雌激素受體調節劑及其用途
CN107735399B (zh) 作为蛋白质激酶的调节剂的手性二芳基大环
US10577366B2 (en) Crystalline forms of a compound that inhibits bromodomain
CN109311876B (zh) 杂芳基雌激素受体调节剂及其用途
US10398689B2 (en) Benzopiperidine derivative, preparation method thereof and medical use thereof
WO2022017339A1 (zh) 稠合哒嗪类衍生物、其制备方法及其在医药上的应用
WO2020064002A1 (zh) 异吲哚啉类化合物、其制备方法、药物组合物及用途
CN104024255A (zh) 作为btk活性的抑制剂的烷基化哌嗪化合物
WO2021228210A1 (zh) 吡咯烷类化合物及其应用
WO2021190417A1 (zh) 新型氨基嘧啶类egfr抑制剂
WO2021249533A1 (zh) 雌激素受体调节剂化合物及其用途
TWI826819B (zh) 一種作為btk抑制劑的化合物及其製備方法與用途
US9884874B2 (en) Bromodomain-inhibiting compounds and pharmaceutical composition comprising same for preventing or treating a cancer
US9724331B2 (en) Use of maleimide derivatives for preventing and treating leukemia
WO2022247816A1 (zh) 含氮杂环类化合物、其制备方法及其在医药上的应用
WO2021139756A1 (zh) 三环四氢异喹啉类衍生物、其制备方法及其在医药上的应用
TW202110848A (zh) 取代的稠合雙環類衍生物、其製備方法及其在醫藥上的應用
WO2021213358A1 (zh) 含硼化合物及其应用
WO2022166860A1 (zh) Pim激酶抑制剂
WO2020020377A1 (zh) 用作fgfr4抑制剂的稠环衍生物
WO2023125928A1 (zh) Menin抑制剂及其用途
CN113286786A (zh) Nlrp3调节剂
TW201835079A (zh) 6-吡唑-[1,2,4]三唑並[4,3-α]吡啶-3-醯胺類衍生物、其製備方法及其在醫藥上的應用
TW202214631A (zh) 作為Akt激酶抑制劑的化合物

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: 21791701

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21791701

Country of ref document: EP

Kind code of ref document: A1