WO2023016484A1 - 磺酰胺衍生物、其制备方法及其在医药上的应用 - Google Patents

磺酰胺衍生物、其制备方法及其在医药上的应用 Download PDF

Info

Publication number
WO2023016484A1
WO2023016484A1 PCT/CN2022/111395 CN2022111395W WO2023016484A1 WO 2023016484 A1 WO2023016484 A1 WO 2023016484A1 CN 2022111395 W CN2022111395 W CN 2022111395W WO 2023016484 A1 WO2023016484 A1 WO 2023016484A1
Authority
WO
WIPO (PCT)
Prior art keywords
general formula
cancer
group
alkyl
pharmaceutically acceptable
Prior art date
Application number
PCT/CN2022/111395
Other languages
English (en)
French (fr)
Inventor
张晓敏
胡伟民
贺峰
陶维康
Original Assignee
江苏恒瑞医药股份有限公司
上海恒瑞医药有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 江苏恒瑞医药股份有限公司, 上海恒瑞医药有限公司 filed Critical 江苏恒瑞医药股份有限公司
Priority to EP22855464.8A priority Critical patent/EP4385989A1/en
Priority to JP2024507166A priority patent/JP2024529068A/ja
Priority to CN202280046780.6A priority patent/CN117597341A/zh
Priority to CA3228411A priority patent/CA3228411A1/en
Priority to KR1020247007144A priority patent/KR20240046530A/ko
Priority to AU2022325367A priority patent/AU2022325367A1/en
Priority to MX2024001777A priority patent/MX2024001777A/es
Publication of WO2023016484A1 publication Critical patent/WO2023016484A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/424Oxazoles condensed with heterocyclic ring systems, e.g. clavulanic acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the disclosure belongs to the field of medicine, and relates to a sulfonamide derivative, its preparation method and its application in medicine.
  • the present disclosure relates to a sulfonamide derivative represented by general formula (I), its preparation method and a pharmaceutical composition containing the derivative, and its use as a KAT inhibitor in the preparation of a drug for treating and/or preventing cancer use.
  • Lysine acetyltransferases are a class of enzymes that can catalyze the transfer of acetyl groups from acetyl-CoA to lysine ⁇ -amino groups in protein substrates.
  • the acetylation of lysine can affect the function of proteins, thus playing an important regulatory role in chromosome structure, gene transcription regulation, DNA binding ability, enzyme activity and stability, protein interaction and intracellular localization.
  • KATs are divided into several subfamilies, among which MYST (MOZ, YBF2/SAS3, SAS2, TIP60) is the largest one, including KAT5 (TIP60), KAT6A (MOZ; MYST3), KAT6B (MORF; MYST4), KAT7 (HBO; MYST2 ) and KAT8 (MOF; MYST1).
  • KAT6A/B as the main member of the MYST family, plays a vital role in development, maintenance of stem cells in the hematopoietic and immune systems, and tumor development and drug resistance.
  • KAT6A and KAT6B were amplified in a variety of tumors.
  • KAT6A is located in the amplicon region of chromosome 8p11-p12, which is amplified in 10-15% of breast cancers, and its copy number is positively correlated with mRNA expression and associated with poor prognosis.
  • Both KAT6A and KAT6B were significantly highly expressed in breast cancer.
  • Further subtype analysis showed that there was a certain correlation between the high expression of KAT6A/B and the expression level of ER ⁇ , revealing that KAT6A/B may be a potential target of ER + /HER2 - breast cancer.
  • KAT6A/B selective inhibitor CTx-648 showed antitumor activity in ER+ breast cancer both in vitro and in vivo, and the expression level of KAT6A was correlated with the sensitivity of CTx-648. In ER + breast cancer cells with high KAT6A expression, CTx-648 can downregulate ER ⁇ expression, and H3K23Ac can be used as a pharmacodynamic biomarker of KAT6 inhibitors.
  • KAT6A/B inhibitors have clinical development value as single agents or in combination with existing ER + /HER2 - breast cancer therapies such as fulvestrant, CDK4/6 inhibitors, and even SERD and SERCA. of.
  • KAT6A/B inhibitors have potential application prospects in glioma, B cell lymphoma, liver cancer, ovarian cancer and other tumor types, and can be used as indication expansion.
  • Published KAT6 inhibitor patent applications include WO2016198507A1, WO2019243491A1, WO2019043139A1, WO2019108824A1, WO2020216701A1, WO2020002587A1, WO2020254946A1 and WO2020254989A1, etc.
  • Ring A is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • Ring B is cycloalkyl or heterocyclyl
  • L is a chemical bond, an alkylene group or a heteroalkylene group; wherein each of the alkylene or heteroalkylene groups is independently selected from the group consisting of hydroxyl, halogen, alkyl, haloalkyl, hydroxyalkyl, alkoxy and one or more substituents in haloalkoxy;
  • Each R 1 , each R 2 , and R 3 are the same or different, and each independently selected from a hydrogen atom, halogen, cyano, nitro, oxo, alkenyl, alkynyl, alkyl, alkoxy, cycloalkane radical, heterocyclyl, aryl, heteroaryl, -OR 5 , -C(O)R 6 , -C(O)OR 6 , -OC(O)R 6 , -NHC(O)OR 6 , - NR 7 R 8 , -C(O)NR 7 R 8 , -S(O) r R 6 and -S(O) r NR 7 R 8 ; the alkenyl, alkynyl, alkyl, alkoxy radical, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently selected from the group consisting of hydroxyl, halogen, cyano, amino, nitro, oxo, alken
  • R4 is a hydrogen atom
  • Ring C is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R is selected from hydrogen atom, hydroxyl, halogen, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy and cycloalkyl;
  • Each R 4a is the same or different, and each independently selected from hydrogen atom, hydroxyl, halogen, cyano, nitro, oxo, alkenyl, alkynyl, alkyl, haloalkyl, hydroxyalkyl, alkoxy, Haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkylalkyl, heterocyclylalkyl, cycloalkyloxy, heterocyclyloxy and -NR 9 R 10 ;
  • R 5 and R 6 are the same or different, and are each independently selected from a hydrogen atom, alkenyl, alkynyl, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl; wherein the alkenyl, alkynyl radical, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently selected from the group consisting of hydroxyl, halogen, cyano, amino, nitro, oxo, alkenyl, alkynyl, alkane radical, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkylalkyl, heterocyclylalkyl, cycloalkyloxy and One or more substituents in heterocyclyloxy are substituted;
  • R 7 , R 8 , R 9 , and R 10 are the same or different, and are each independently selected from a hydrogen atom, an alkyl group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group; wherein the alkyl group, ring Alkyl, heterocyclyl, aryl and heteroaryl are each independently selected from the group consisting of hydroxy, halo, cyano, amino, nitro, oxo, alkenyl, alkynyl, alkyl, haloalkyl, hydroxy Alkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkylalkyl, heterocyclylalkyl, cycloalkyloxy and heterocyclyloxy substituted by one or more substituents;
  • R 7 and R 8 form a heterocyclic group together with the connected N atom
  • R 9 and R 10 form a heterocyclic group together with the connected N atom
  • the heterocyclic group is optionally selected from from hydroxy, halogen, cyano, amino, nitro, oxo, alkenyl, alkynyl, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl
  • p 0, 1, 2, 3 or 4;
  • q 0, 1, 2, 3 or 4;
  • n 0, 1, 2, 3 or 4;
  • n 0, 1, 2, 3 or 4;
  • r 0, 1 or 2.
  • Ring A is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • Ring B is selected from cycloalkyl or heterocyclyl
  • L is a chemical bond, an alkylene group or a heteroalkylene group; wherein each of the alkylene or heteroalkylene groups is independently selected from the group consisting of hydroxyl, halogen, alkyl, haloalkyl, hydroxyalkyl, alkoxy and one or more substituents in haloalkoxy;
  • Each R 1 , each R 2 , and R 3 are the same or different, and each independently selected from a hydrogen atom, halogen, cyano, nitro, oxo, alkenyl, alkynyl, alkyl, alkoxy, cycloalkane radical, heterocyclyl, aryl, heteroaryl, -OR 5 , -C(O)R 6 , -C(O)OR 6 , -OC(O)R 6 , -NHC(O)OR 6 , - NR 7 R 8 , -C(O)NR 7 R 8 , -S(O) r R 6 and -S(O) r NR 7 R 8 ; the alkenyl, alkynyl, alkyl, alkoxy radical, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently selected from the group consisting of hydroxyl, halogen, cyano, amino, nitro, oxo, alken
  • R4 is a hydrogen atom
  • Ring C is aryl or heteroaryl
  • R is selected from hydrogen atom, hydroxyl, halogen, alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy and cycloalkyl;
  • Each R 4a is the same or different, and each independently selected from hydrogen atom, hydroxyl, halogen, cyano, nitro, oxo, alkenyl, alkynyl, alkyl, haloalkyl, hydroxyalkyl, alkoxy, Haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkylalkyl, heterocyclylalkyl, cycloalkyloxy, heterocyclyloxy and -NR 9 R 10 ;
  • R 5 and R 6 are the same or different, and are each independently selected from a hydrogen atom, alkenyl, alkynyl, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl; wherein the alkenyl, alkynyl radical, alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently selected from the group consisting of hydroxyl, halogen, cyano, amino, nitro, oxo, alkenyl, alkynyl, alkane radical, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkylalkyl, heterocyclylalkyl, cycloalkyloxy and One or more substituents in heterocyclyloxy are substituted;
  • R 7 , R 8 , R 9 , and R 10 are the same or different, and are each independently selected from a hydrogen atom, an alkyl group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group; wherein the alkyl group, ring Alkyl, heterocyclyl, aryl and heteroaryl are each independently selected from the group consisting of hydroxy, halo, cyano, amino, nitro, oxo, alkenyl, alkynyl, alkyl, haloalkyl, hydroxy Alkyl, alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkylalkyl, heterocyclylalkyl, cycloalkyloxy and heterocyclyloxy substituted by one or more substituents;
  • R 7 and R 8 form a heterocyclic group together with the connected N atom
  • R 9 and R 10 form a heterocyclic group together with the connected N atom
  • the heterocyclic group is optionally selected from Hydroxy, Halo, Cyano, Amino, Nitro, Oxo, Alkenyl, Alkynyl, Alkyl, Haloalkyl, Hydroxyalkyl, Alkoxy, Haloalkoxy, Cycloalkyl, Heterocyclyl, Aryl , heteroaryl, cycloalkylalkyl, heterocyclylalkyl, cycloalkyloxy and heterocyclyloxy are substituted by one or more substituents;
  • p 0, 1, 2, 3 or 4;
  • q 0, 1, 2, 3 or 4;
  • n 0, 1, 2, 3 or 4;
  • n 0, 1, 2, 3 or 4;
  • r 0, 1 or 2.
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein R 3 is selected from hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1- 6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy and 3 to 8 membered cycloalkyl; preferably, R 3 is a hydrogen atom.
  • the compound represented by general formula (I) or a pharmaceutically acceptable salt thereof wherein ring C is selected from 3 to 8 membered cycloalkyl, 3 to 8 membered heterocyclyl, 6 to 10-membered aryl and 5- to 10-membered heteroaryl; preferably, ring C is 5- to 10-membered heteroaryl or 6- to 10-membered aryl; more preferably, ring C is 5- or 6-membered heteroaryl ; Most preferably, ring C is pyrazolyl.
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein ring C is a 5-membered or 6-membered heterocyclic group, a 5-membered or 6-membered heteroaryl group;
  • ring C is selected from pyrazolyl, pyridyl, furyl and tetrahydrofuryl; more preferably, ring C is pyrazolyl.
  • the compound represented by the general formula (I) or a pharmaceutically acceptable salt thereof wherein R 0 is selected from hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1- 6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy and 3 to 8 membered cycloalkyl; preferably, R is selected from hydrogen atom, hydroxyl, halogen and C 1-6 alkyl; preferably, R 0 is selected from a hydrogen atom, hydroxyl and Cl; more preferably, R 0 is a hydrogen atom.
  • m is 0 or 1, preferably 1.
  • R4 is R 4a and n are as defined in general formula (I);
  • R4 is
  • the compound represented by general formula (I) or a pharmaceutically acceptable salt thereof is a compound represented by general formula (II) or a pharmaceutically acceptable salt thereof:
  • Ring C is a 5- to 10-membered heteroaryl group, preferably a 5-membered or 6-membered heteroaryl group;
  • Ring A, ring B, L, R 1 , R 2 , R 4a , p, q and n are as defined in the general formula (I).
  • R4 is R 4a and n are as defined in general formula (I);
  • R 4 is R 4a and n are as defined in general formula (I);
  • R4 is
  • R 4 is
  • the compound represented by general formula (I) or a pharmaceutically acceptable salt thereof is a compound represented by general formula (Ii) or a pharmaceutically acceptable salt thereof:
  • Ring C is a 3- to 8-membered heterocyclic group or a 5- to 10-membered heteroaryl group, preferably a 5- or 6-membered heterocyclic group, a 5- or 6-membered heteroaryl group;
  • Ring A, ring B, L, R 1 , R 2 , R 4a , p, q and n are as defined in the general formula (I).
  • the compound represented by general formula (I), general formula (II) or general formula (Ii) or a pharmaceutically acceptable salt thereof wherein ring A is a 6- to 10-membered aryl group Or 5 to 10 membered heteroaryl, the 6 to 10 membered aryl is preferably phenyl or naphthyl; the 5 to 10 membered heteroaryl is preferably pyridyl, quinolinyl and benzoxazolyl.
  • the compound represented by general formula (I), general formula (II) or general formula (Ii) or a pharmaceutically acceptable salt thereof wherein ring A is a 6- to 10-membered aryl group Or 5 to 10 membered heteroaryl, the 6 to 10 membered aryl is preferably selected from phenyl, naphthyl, The 5- to 10-membered heteroaryl is preferably pyridyl, quinolinyl and benzoxazolyl.
  • Ring B is a 4 to 7 membered heterocyclyl or a 4 to 7 membered cycloalkyl
  • ring B is a 4- to 7-membered heterocyclic group
  • Ring B is a 4- to 7-membered heterocyclic group, wherein the 4- to 7-membered heterocyclic group contains 1-3 oxygen atoms;
  • ring B is a 5-membered or 6-membered heterocyclic group, wherein the 5-membered or 6-membered heterocyclic group contains 1 or 2 oxygen atoms.
  • the compound represented by general formula (I), general formula (II) or general formula (Ii) or a pharmaceutically acceptable salt thereof, wherein ring B is selected from preferably R 2 can be substituted at any substitutable position on ring B.
  • ring B is selected from More preferably, Ring B is R 2 can be substituted at any substitutable position on ring B.
  • the compound represented by general formula (I) or (II) or a pharmaceutically acceptable salt thereof is a compound represented by general formula (III) or a pharmaceutically acceptable salt thereof the salt:
  • Each R a , R b , R c and R d are the same or different, and each independently selected from a hydrogen atom, a hydroxyl group, a halogen, a cyano group, an amino group, an alkenyl group, an alkynyl group, an alkyl group, a haloalkyl group, a hydroxyalkyl group, Alkoxy, haloalkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkyloxy and heterocyclyloxy;
  • s 0, 1, 2 or 3;
  • L, R 1 , R 4a , p and n are as defined in the general formula (I).
  • each Rc and Rd are the same or different, and are each independently selected from hydrogen atom, halogen, C1-6 alkyl and C1-6 alkoxy;
  • each R c and R d are the same or different, and each independently is a hydrogen atom or a halogen;
  • each Rc and Rd are the same or different, and are each independently a hydrogen atom or a fluorine atom;
  • both R c and R d are hydrogen atoms.
  • the compound represented by general formula (I), general formula (II) or general formula (III) or a pharmaceutically acceptable salt thereof is represented by general formula (IV) Compound or its pharmaceutically acceptable salt:
  • L, X, R 1 , R 4a , p, n and s are as defined in general formula (III).
  • each R a and R b are the same or different, and are each independently selected from a hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy , C 1-6 haloalkoxy, 3 to 8 membered cycloalkyl, 3 to 8 membered heterocyclyl, 3 to 8 membered cycloalkyloxy and 3 to 8 membered heterocyclyloxy;
  • each R a and R b are the same or different, and are each independently selected from a hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl and C 1-6 alkoxy ;
  • each R a and R b are the same or different, and are each independently selected from a hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6
  • the compound represented by general formula (III) or general formula (IV) or a pharmaceutically acceptable salt thereof wherein X is O or CH 2 ; preferably, X is CH 2 .
  • the compound represented by general formula (III) or a pharmaceutically acceptable salt thereof wherein R c and R d are the same or different, and each independently is a hydrogen atom or a halogen; and/ or X is O or CH2 ; and/or s is 1 or 2.
  • each R 1 is the same or different, and each independently selected from a hydrogen atom, a hydroxyl group, a halogen, a cyano group, an amino group, a C 1-6 alkyl group, a C 1-6 haloalkyl group, a C 1-6 hydroxyalkyl group, C 1-6 alkoxy, C 1-6 haloalkoxy, oxo, 3 to 8 membered cycloalkyl, C 1-6 alkoxy C 1-6 alkyl, -OR 5 , -C(O )R 6 , -C(O)OR 6 , -C(O)NR 7 R 8 and -S(O) r R 6 , r, R 5 , R 6 , R 7 and R 8 such as general formula (I) defined in; preferably
  • each R 1 is the same or different, and each independently selected from hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkane Oxygen, C 1-6 haloalkoxy, C 1-6 alkoxy C 1-6 alkyl, -C(O)OCH 3 and -NR 7 R 8 ;
  • R 7 and R 8 are the same or different, and each are independently a hydrogen atom or a C 1-6 alkyl group;
  • each R 1 is the same or different, and each independently selected from hydrogen atom, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy and -NR 7 R 8 ;
  • R 7 and R 8 are the same or different, and each independently is a hydrogen atom or a C 1-6 alkyl group;
  • each R 1 is the same or different, and is independently C 1-6 alkoxy.
  • each R 1 is the same or different, and each independently is methoxy.
  • each R 1 is the same or different, and each independently selected from a hydrogen atom, a fluorine atom, a chlorine atom, a methyl group, an ethyl group, an isopropyl group, a methoxy group, an ethoxy group, a trifluoromethoxy group, a methylamino, dimethylamino and Preferably, R 1 is methoxy.
  • each R 2 is the same or different, and each independently selected from hydrogen atom, hydroxyl, halogen, cyano, amino, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy, C 1-6 Haloalkoxy, 3 to 8 membered cycloalkyl, 3 to 8 membered heterocyclyl, oxo, 3 to 8 membered cycloalkyloxy and 3 to 8 membered heterocyclyloxy; preferably, each R The same or different, and each independently selected from a hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy and oxo ; Further preferably, each R 2 is the same or different, and is independently selected from hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C
  • each R 4a is the same or different, and each independently selected from a hydrogen atom, hydroxyl, halogen, cyano, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1 -6 alkoxy, C 1-6 haloalkoxy, -NR 9 R 10 , 3 to 8 membered cycloalkyl, 3 to 8 membered heterocyclyl, 3 to 8 membered cycloalkyloxy and 3 to 8 membered Heterocyclyloxy, R 9 and R 10 are as defined in the general formula (I); preferably, each R 4a is the same or different, and each independently selected from hydrogen atom, hydroxyl, halogen, C 1-6 alkyl , C 1-6 hydroxyalkyl and C 1-6 alkoxy
  • the compound represented by general formula (I), general formula (II), general formula (Ii), general formula (III) or general formula (IV) or its pharmaceutically acceptable Salt wherein L is a chemical bond or a C 1-6 alkylene group; preferably selected from chemical bond, -CH 2 - or -CH 2 CH 2 -, more preferably chemical bond and -CH 2 -, most preferably chemical bond.
  • the compound represented by general formula (I), general formula (II), general formula (Ii), general formula (III) or general formula (IV) or its pharmaceutically acceptable Salt where n is 1 or 2.
  • the compound represented by general formula (III) or general formula (IV) or a pharmaceutically acceptable salt thereof wherein each R 4a is the same or different, and each independently selected from halogen, Hydroxy, C 1-6 alkyl, C 1-6 hydroxyalkyl and C 1-6 alkoxy, and n is 1 or 2, or R 4a is a hydrogen atom, and n is 3.
  • the compound represented by general formula (I), general formula (II), general formula (Ii), general formula (III) or general formula (IV) or its pharmaceutically acceptable Salt wherein R 5 and R 6 are the same or different, and are each independently selected from a hydrogen atom, C 1-6 alkyl, 3 to 8 membered cycloalkyl, 3 to 12 membered heterocyclic group, 6 to 10 membered aryl group and 5 to 10 membered heteroaryl; wherein said C 1-6 alkyl, 3 to 8 membered cycloalkyl, 3 to 12 membered heterocyclic group, 6 to 10 membered aryl and 5 to 10 membered heteroaryl Each independently optionally selected from hydroxyl, halogen, cyano, amino, nitro, oxo, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 One or more substituents in alkoxy, C 1-6 haloalkoxy
  • R 5 and R 6 are the same or different, and are independently selected from C 1-6 alkyl, C 1-6 haloalkyl and 3-8 membered cycloalkyl.
  • the compound represented by general formula (I), general formula (II), general formula (Ii), general formula (III) or general formula (IV) or its pharmaceutically acceptable Salt wherein R 7 and R 8 are the same or different, and each independently selected from hydrogen atom, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, 3 to 8 membered cycloalkyl and a 3- to 12-membered heterocyclic group; preferably, R 7 and R 8 are the same or different, and each independently is a hydrogen atom or a C 1-6 alkyl group; more preferably, R 7 and R 8 are hydrogen atoms.
  • the compound represented by general formula (I), general formula (II), general formula (Ii), general formula (III) or general formula (IV) or its pharmaceutically acceptable Salt wherein R 9 and R 10 are the same or different, and are each independently selected from a hydrogen atom, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, 3 to 8 membered cycloalkyl and 3- to 12-membered heterocyclyl; preferably, R9 and R10 are hydrogen atoms.
  • the compound represented by general formula (I), general formula (II) and general formula (Ii) or a pharmaceutically acceptable salt thereof, wherein for R 1a and R 1b are the same or different, and each independently selected from a hydrogen atom, hydroxyl, halogen, cyano, amino, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, oxo, 3 to 8 membered cycloalkyl, C 1-6 alkoxy C 1-6 alkyl, -OR 5 , -C(O) R 6 , -C(O)OR 6 , -C(O)NR 7 R 8 and -S(O) r R 6 , r, R 5 , R 6 , R 7 and R 8 are as in general formula (I) defined; preferably, R 1a and R 1b are the same or different, and each independently selected from hydrogen atom,
  • the compound represented by general formula (I), general formula (II) and general formula (Ii) or a pharmaceutically acceptable salt thereof, wherein for R 1a and R 1b are the same or different, and each independently selected from a hydrogen atom, hydroxyl, halogen, cyano, amino, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, oxo, 3 to 8 membered cycloalkyl, C 1-6 alkoxy C 1-6 alkyl, -OR 5 , -C(O) R 6 , -C(O)OR 6 , -C(O)NR 7 R 8 and -S(O) r R 6 , r, R 5 , R 6 , R 7 and R 8 are as in general formula (I) defined; preferably, R 1a and R 1b are the same or different, and each independently selected from hydrogen atom,
  • the compound represented by general formula (III) or a pharmaceutically acceptable salt thereof wherein for Preferably, selected from More preferably, for
  • the compound represented by general formula (IV) or a pharmaceutically acceptable salt thereof wherein selected from Preferably, selected from More preferably, for
  • each R 1 is the same or different, and each independently selected from hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 1-6 alkoxy C 1-6 alkyl and -C(O)OCH 3 ;
  • p is 1, 2 or 3;
  • each R 2 is the same or different, and each independently selected from a hydrogen atom, halogen, C 1-6 alkyl and C 1-6 alkoxy;
  • q is 1;
  • R 3 is a hydrogen atom;
  • R 4 is Each R 4a is the same or different, and each independently selected from a hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 hydroxyalkyl and C 1-6 alkoxy;
  • n is 1 or 2;
  • ring C is a 5- to 10-member
  • each R 1 is the same or different, and each independently selected from hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 1-6 alkoxy C 1-6 alkyl , -C(O)OCH 3 and -NR 7 R 8 ;
  • R 7 and R 8 are the same or different, and each independently is a hydrogen atom or a C 1-6 alkyl group;
  • p is 0, 1, 2, 3 or 4 ;
  • Each R 2 is the same or different, and each independently selected from a hydrogen atom, halogen, C 1-6 alkyl and C 1-6 alkoxy; q is 0, 1, 2, 3 or 4;
  • R 3 is hydrogen atom;
  • R 4 is Each R 4a is the same or different, and each independently selected from hydrogen atom, hydroxyl,
  • each R 1 is the same or different, and each independently selected from hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 1-6 alkoxy C 1-6 alkyl and -C(O)OCH 3 ;
  • p is 1, 2 or 3;
  • each R 2 is the same or different, and each independently selected from a hydrogen atom, halogen, C 1-6 alkyl and C 1-6 alkoxy;
  • q is 1;
  • each R 4a is the same or different, and each independently selected from a hydrogen atom, halogen, hydroxyl, C 1-6 alkyl, C 1-6 hydroxyalkyl and C 1-6 alkoxy;
  • n is 1 or 2;
  • Ring A is a 6- to 10-membered aryl group;
  • Ring B is a 4-
  • each R 1 is the same or different, and each independently selected from hydrogen atom, halogen, C 1- 6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy and -NR 7 R 8 ;
  • R 7 and R 8 are the same or different, and each independently represents a hydrogen atom or C 1-6 alkyl;
  • p is 0, 1, 2 or 3;
  • each R 2 is the same or different, and each independently is a hydrogen atom or a halogen;
  • q is 0, 1 or 2;
  • each R 4a is the same or different, and Each independently selected from hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 hydroxyalkyl and C 1-6 alkoxy;
  • n is 0, 1, 2 or 3;
  • ring A is 6 to 10-membered aryl;
  • Ring B is a 4- to 7-membered heterocyclic group;
  • each R 1 is the same or different, and each independently selected from hydrogen atom, halogen, C 1- 6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy and -NR 7 R 8 ;
  • R 7 and R 8 are the same or different, and each independently represents a hydrogen atom or C 1-6 alkyl;
  • p is 0, 1, 2 or 3;
  • each R 2 is the same or different, and each independently is a hydrogen atom or a halogen;
  • q is 0, 1 or 2;
  • each R 4a is the same or different, and Each independently selected from hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 hydroxyalkyl and C 1-6 alkoxy;
  • n is 0, 1, 2 or 3;
  • ring A is 6 to 10-membered aryl;
  • ring B is a 4- to 7-membered heterocyclic group;
  • each R 1 is the same or different, and each independently selected from hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 1-6 alkoxy C 1-6 alkyl and -C(O)OCH 3 ;
  • p is 1, 2 or 3;
  • X is O or CH 2 ;
  • R c and R d are both hydrogen atoms;
  • each R 4a is the same or different, and each independently selected from halogen, hydroxyl , C 1-6 alkyl, C 1-6 hydroxyalkyl and C 1-6 alkoxy, and n is 1 or 2, or R 4a are all hydrogen atoms, and n is 3;
  • s is 1 or 2;
  • L is a chemical bond or -CH 2 -.
  • each R 1 is the same or different, and each independently selected from hydrogen atom, halogen, C 1- 6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy and -NR 7 R 8 ;
  • R 7 and R 8 are the same or different, and each independently represents a hydrogen atom or C 1-6 alkyl;
  • p is 0, 1, 2 or 3;
  • X is O or CH 2 ;
  • each R c and R d are the same or different, and each independently is a hydrogen atom or a halogen;
  • R 4a is a hydrogen atom ;
  • n is 3;
  • s is 1 or 2; and
  • L is a chemical bond.
  • the compound represented by general formula (III) or a pharmaceutically acceptable salt thereof wherein each R 1 is the same or different, and each independently selected as C 1-6 alkoxy; p is 2; X is O or CH 2 ; each R c and R d is the same or different, and each independently is a hydrogen atom or a fluorine atom; R 4a is a hydrogen atom; n is 3; s is 1 or 2; L is a chemical bond.
  • the compound represented by general formula (III) or a pharmaceutically acceptable salt thereof wherein each R 1 is the same or different, and each independently selected as C 1-6 alkoxy; p is 1 or 2; X is CH 2 ; each of R c and R d is the same or different, and each independently is a hydrogen atom or a fluorine atom; n is 0; s is 1 or 2; and L is a chemical bond.
  • each R 1 is the same or different, and each independently selected from hydrogen atom, hydroxyl, halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 1-6 alkoxy C 1-6 alkyl and -C(O)OCH 3 ;
  • p is 1, 2 or 3;
  • X is O or CH 2 ;
  • each R 4a is the same or different, and each independently selected from halogen, hydroxyl, C 1-6 alkyl, C 1 -6 hydroxyalkyl and C 1-6 alkoxy, and n is 1 or 2, or R 4a are both hydrogen atoms, and n is 3;
  • s is 1 or 2; and L is a chemical bond or -CH 2 -.
  • each R 1 is the same or different, and each independently selected from hydrogen atom, halogen, C 1- 6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy and -NR 7 R 8 ;
  • R 7 and R 8 are the same or different, and each independently represents a hydrogen atom or C 1-6 alkyl; p is 0, 1, 2 or 3; X is O or CH 2 ; R 4a are all hydrogen atoms; n is 3; s is 1 or 2;
  • the compound represented by general formula (IV) or a pharmaceutically acceptable salt thereof wherein each R 1 is the same or different, and each independently is C 1-6 alkoxy; p is 1 or 2; X is CH 2 ; R 4a are all hydrogen atoms; n is 3; s is 1 or 2; and L is a chemical bond.
  • the compound represented by general formula (IV) or a pharmaceutically acceptable salt thereof wherein each R 1 is the same or different, and each independently is C 1-6 alkoxy; p is 2; X is O or CH 2 ; R 4a are all hydrogen atoms; n is 3; s is 1 or 2; and L is a chemical bond.
  • the compound represented by general formula (IV) or a pharmaceutically acceptable salt thereof wherein each R 1 is the same or different, and each independently is C 1-6 alkoxy; p is 1 or 2 ; X is CH2; n is 0; s is 1 or 2; and L is a chemical bond.
  • Typical compounds of the present disclosure include, but are not limited to:
  • Ring B, R 2 , R 3 , R 4 and q are as defined in the general formula (I).
  • Ring B, ring C, R 2 , R 4a , q and n are as defined in the general formula (II).
  • Ring B, ring C, R 2 , R 4a , q and n are as defined in the general formula (Ii).
  • X is halogen , preferably bromine
  • Ring A, ring B, R 1 , R 2 , R 3 , L, p and q are as defined in the general formula (I).
  • X is halogen , preferably bromine
  • Ring A, ring B, R 1 , R 2 , L, p and q are as defined in the general formula (II).
  • Another aspect of the present disclosure relates to a compound or a salt thereof represented by general formula (IIIA'):
  • X is halogen , preferably bromine
  • R 1 , R c , R d , s, p, X and L are as defined in general formula (III).
  • X is halogen , preferably bromine
  • R 1 , s, p, X and L are as defined in general formula (IV).
  • Typical intermediate compounds of the present disclosure include, but are not limited to:
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by general formula (I) or a pharmaceutically acceptable salt thereof, the method comprising the following steps:
  • Ring A, ring B, L, R 1 to R 4 , p and q are as defined in the general formula (I).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by general formula (II) or a pharmaceutically acceptable salt thereof, the method comprising the following steps:
  • Ring A, ring B, ring C, L, R 1 , R 2 , R 4a , p, q and n are as defined in the general formula (II).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by general formula (Ii) or a pharmaceutically acceptable salt thereof, the method comprising the following steps:
  • Ring A, ring B, ring C, L, R 1 , R 2 , R 4a , p, q and n are as defined in the general formula (Ii).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by general formula (III) or a pharmaceutically acceptable salt thereof, the method comprising the following steps:
  • L, X, R 1 , R 4a , R c , R d , p, n and s are as defined in general formula (III).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by general formula (IV) or a pharmaceutically acceptable salt thereof, the method comprising the following steps:
  • a compound represented by general formula (IVA) or a salt thereof reacts with a compound represented by general formula (IIIB) or a salt thereof to obtain a compound represented by general formula (IV) or a pharmaceutically acceptable salt thereof,
  • X, L, R 1 , R 4a , p, n and s are as defined in general formula (IV).
  • Another aspect of the present disclosure relates to a method for preparing a compound represented by general formula (Ii) or a pharmaceutically acceptable salt thereof, the method comprising the following steps:
  • Ring D is a 3- to 8-membered heterocyclic group containing at least one double bond in the ring, preferably a 5-membered or 6-membered heterocyclic group containing at least one double bond in the ring, more preferably
  • Ring C is a 3- to 8-membered heterocyclic group, preferably a 5-membered or 6-membered heterocyclic group, more preferably
  • X is halogen , preferably bromine
  • Ring A, ring B, L, R 1 , R 2 , R 4a , p, q and n are as defined in the general formula (Ii).
  • compositions which contains the general formula (I), general formula (II), general formula (Ii), general formula (III), general formula (IV) of the present disclosure Or a compound shown in Table A or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the present disclosure further relates to compounds shown in general formula (I), general formula (II), general formula (Ii), general formula (III), general formula (IV) or Table A or their pharmaceutically acceptable salts or including their Use of the pharmaceutical composition of the invention in the preparation of a medicament for inhibiting KAT; wherein the KAT is preferably KAT6, more preferably KAT6A and/or KAT6B.
  • the present disclosure further relates to compounds shown in general formula (I), general formula (II), general formula (Ii), general formula (III), general formula (IV) or Table A or their pharmaceutically acceptable salts or including their
  • the pharmaceutical composition of the invention is used in the preparation of medicines for treating and/or preventing KAT-mediated diseases; wherein the KAT is preferably KAT6, more preferably KAT6A and/or KAT6B.
  • the present disclosure further relates to compounds shown in general formula (I), general formula (II), general formula (Ii), general formula (III), general formula (IV) or Table A or their pharmaceutically acceptable salts or including their The purposes of the pharmaceutical composition in the preparation of the medicament for treating and/or preventing cancer;
  • said cancer is preferably selected from lung cancer (such as NCSLC, SCLC), mesothelioma, bone cancer, pancreatic cancer, skin cancer, head and neck cancer Cancer, brain cancer, melanoma, anal cancer, liver cancer, breast cancer, fallopian tube cancer, endometrial cancer, cervical cancer, ovarian cancer, vaginal cancer, vulvar cancer, Hodgkin's lymphoma, esophagus cancer, colorectal cancer, small bowel cancer Cancer, stomach cancer, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, penile cancer, testicular cancer, prostate cancer, leukemia, B cell lymphoma, bladder cancer, urethral cancer, ure
  • the present disclosure further relates to a method for inhibiting KAT, which comprises administering an effective amount of general formula (I), general formula (II), general formula (Ii), general formula (III), general formula (IV) to the required patient Or a compound shown in Table A or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising it, wherein the KAT is preferably KAT6, more preferably KAT6A and/or KAT6B.
  • the present disclosure also relates to a method for treating and/or preventing KAT-mediated diseases, which comprises administering a therapeutically effective dose and/or a preventively effective dose of general formula (I), general formula (II), general formula ( Ii), the compound shown in general formula (III), general formula (IV) or table A or its pharmaceutically acceptable salt, or the pharmaceutical composition comprising it, wherein said KAT is preferably KAT6, more preferably KAT6A and / or KAT6B, .
  • the present disclosure further relates to a method for treating and/or preventing cancer, which comprises administering a therapeutically effective amount and/or a preventively effective amount of general formula (I), general formula (II), general formula (Ii), general formula
  • said cancer is preferably selected from lung cancer (such as NCSLC, SCLC), intermediate Skin tumor, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, brain cancer, melanoma, anal cancer, liver cancer, breast cancer, fallopian tube cancer, endometrial cancer, cervical cancer, ovarian cancer, vaginal cancer, vulvar cancer, hormonal cancer Oddkin's lymphoma, esophageal cancer, colorectal cancer, small bowel cancer, gastric cancer, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, penile cancer, testicular cancer, prostate cancer, leukemia, B-cell lymph
  • the present disclosure further relates to a compound represented by general formula (I), general formula (II), general formula (Ii), general formula (III), general formula (IV) or Table A or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising it, for use as a medicament.
  • the present disclosure further relates to a compound represented by general formula (I), general formula (II), general formula (Ii), general formula (III), general formula (IV) or Table A or a pharmaceutically acceptable salt thereof, Or a pharmaceutical composition comprising it, which is used as a drug for inhibiting KAT, wherein the KAT is preferably KAT6, more preferably KAT6A and/or KAT6B.
  • the present disclosure further relates to a compound represented by general formula (I), general formula (II), general formula (Ii), general formula (III), general formula (IV) or Table A or a pharmaceutically acceptable salt thereof, Or a pharmaceutical composition comprising the same, which is used for inhibiting KAT, wherein the KAT is preferably KAT6, more preferably KAT6A and/or KAT6B.
  • the present disclosure further relates to a compound represented by general formula (I), general formula (II), general formula (Ii), general formula (III), general formula (IV) or Table A or a pharmaceutically acceptable salt thereof, Or a pharmaceutical composition comprising it, which is used as a KAT inhibitor, wherein the KAT is preferably KAT6, more preferably KAT6A and/or KAT6B.
  • the present disclosure also relates to the compound shown in general formula (I), general formula (II), general formula (Ii), general formula (III), general formula (IV) or Table A or its pharmaceutically acceptable salt, or A pharmaceutical composition comprising it, which is used for treating and/or preventing KAT-mediated diseases, wherein the KAT is preferably KAT6, more preferably KAT6A and/or KAT6B.
  • the present disclosure further relates to a compound represented by general formula (I), general formula (II), general formula (Ii), general formula (III), general formula (IV) or Table A or a pharmaceutically acceptable salt thereof, Or a pharmaceutical composition comprising it, for the treatment and/or prevention of cancer; wherein said cancer is preferably selected from lung cancer (such as NCSLC, SCLC), mesothelioma, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, brain cancer Cancer, melanoma, anal cancer, liver cancer, breast cancer, fallopian tube cancer, endometrial cancer, cervical cancer, ovarian cancer, vaginal cancer, vulvar cancer, Hodgkin's lymphoma, esophagus cancer, colorectal cancer, small bowel cancer, stomach cancer , thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, penile cancer, testicular cancer, prostate cancer, leukemia, B cell lymphoma, bladder cancer, urethral cancer, ureter
  • the KAT6 is KAT6A and/or KAT6B.
  • the cancer is breast cancer.
  • the breast cancer is ER + breast cancer.
  • the breast cancer is ER + /HER2 - breast cancer.
  • the breast cancer is locally advanced or metastatic ER + /HER2 - breast cancer.
  • the lung cancer eg, NCSLC, SCLC
  • the lung cancer is non-small cell lung cancer.
  • the lung cancer (such as NCSLC, SCLC) is locally advanced or metastatic non-small cell lung cancer.
  • the prostate cancer is castration-resistant prostate cancer.
  • the prostate cancer is locally advanced or metastatic castration-resistant prostate cancer.
  • ER + estrogen receptor positive
  • HER2 ⁇ human epidermal growth factor receptor 2 negative
  • NSCLC non-small cell lung cancer
  • CRPC castration-resistant Prostate cancer
  • the active compounds are prepared in a form suitable for administration by any suitable route, and the compositions of the present disclosure are formulated by conventional methods using one or more pharmaceutically acceptable carriers. Accordingly, the active compounds of the present disclosure may be formulated in various dosage forms for oral administration, injection (eg, intravenous, intramuscular or subcutaneous), administration by inhalation or insufflation.
  • the disclosed compounds can also be formulated into dosage forms such as tablets, hard or soft capsules, aqueous or oily suspensions, emulsions, injections, dispersible powders or granules, suppositories, lozenges or syrups.
  • the active compound is preferably presented in unit dose form, or in such a form that the patient can self-administer it as a single dose.
  • a unit dosage form of a compound or composition of the present disclosure may be presented as a tablet, capsule, cachet, bottle, powder, granule, lozenge, suppository, reconstituted powder or liquid.
  • a suitable unit dosage may be from 0.1 to 1000 mg.
  • the pharmaceutical composition of the present disclosure may contain one or more auxiliary materials selected from the following components: fillers (diluents), binders, wetting agents, disintegrants or excipients wait.
  • auxiliary materials selected from the following components: fillers (diluents), binders, wetting agents, disintegrants or excipients wait.
  • the compositions may contain from 0.1 to 99% by weight of active compound.
  • Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients suitable for the manufacture of tablets.
  • excipients may be inert excipients, granulating agents, disintegrants, binders and lubricants.
  • These tablets may be uncoated or may be coated by known techniques to mask the taste of the drug or to delay disintegration and absorption in the gastrointestinal tract, thus providing sustained release over an extended period of time.
  • Oral formulations can also be provided in soft gelatin capsules, wherein the active ingredient is mixed with an inert solid diluent, or where the active ingredient is mixed with a water-soluble carrier or an oil vehicle.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending, dispersing or wetting agents. Aqueous suspensions may also contain one or more preservatives, one or more coloring agents, one or more flavoring agents and one or more sweetening agents.
  • Oily suspensions can be formulated by suspending the active ingredient in a vegetable or mineral oil.
  • the oily suspensions may contain a thickening agent.
  • Sweetening and flavoring agents as mentioned above may be added to provide a palatable preparation. These compositions can be preserved by adding antioxidants.
  • compositions of the present disclosure may also be in the form of oil-in-water emulsions.
  • the oily phase may be vegetable oil, or mineral oil or mixtures thereof.
  • Suitable emulsifiers may be naturally occurring phospholipids, and the emulsions may also contain sweetening agents, flavoring agents, preservatives and antioxidants.
  • Such formulations may also contain a demulcent, a preservative, coloring agents and antioxidants.
  • compositions of the present disclosure may be in the form of sterile injectable aqueous solutions.
  • acceptable vehicles or solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • the sterile injectable preparation may be a sterile injectable oil-in-water microemulsion in which the active ingredient is dissolved in an oily phase.
  • the injection or microemulsion may be injected into the patient's bloodstream by local bulk injection.
  • solutions and microemulsions are preferably administered in a manner that maintains a constant circulating concentration of the disclosed compounds.
  • a continuous intravenous delivery device can be used.
  • An example of such a device is the Deltec CADD-PLUS.TM. Model 5400 IV pump.
  • compositions of the present disclosure may be in the form of sterile injectable aqueous or oily suspensions for intramuscular and subcutaneous administration.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension prepared in a parenterally acceptable non-toxic diluent or solvent.
  • sterile fixed oils are conveniently employed as a solvent or suspending medium. For this purpose, any blended and fixed oil may be used.
  • fatty acids are also used in the preparation of injectables.
  • the disclosed compounds may be administered in the form of suppositories for rectal administration.
  • These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid in the rectum and will therefore melt in the rectum to release the drug.
  • Aqueous suspensions of dispersible powders and granules can be prepared by the addition of water to administer the disclosed compounds.
  • These pharmaceutical compositions can be prepared by mixing the active ingredient with a dispersing or wetting agent, suspending agent or one or more preservatives.
  • the dosage of the drug to be administered depends on many factors, including but not limited to the following factors: the activity of the specific compound used, the age of the patient, the weight of the patient, the state of health of the patient, the behavior of the patient , patient's diet, administration time, administration method, rate of excretion, combination of drugs, severity of disease, etc.; in addition, the optimal treatment mode such as the mode of treatment, the daily dosage of the compound or the content of the pharmaceutically acceptable saltkinds can be validated against traditional treatment regimens.
  • alkyl refers to a saturated aliphatic hydrocarbon group comprising 1 to 20 (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 , 16, 17, 18, 19 or 20) carbon atoms straight or branched (ie C 1-20 alkyl), preferably containing 1 to 12 (eg 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 and 12) carbon atoms (i.e. C 1-20 alkyl), more preferably an alkyl group containing 1 to 6 carbon atoms (i.e. C 1- 6 alkyl).
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1 ,2-Dimethylpropyl, 2,2-Dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2- Methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3 - Dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2 -Methylhexyl, 3-methylhexyl, 4-methylhex
  • Alkyl groups may be substituted or unsubstituted, and when substituted, they may be substituted at any available point of attachment, the substituents being preferably selected from D atoms, halogen, alkoxy, haloalkyl, haloalkoxy, One or more of cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • alkylene refers to a saturated straight or branched chain aliphatic hydrocarbon group, which is a residue derived from the same carbon atom or two different carbon atoms of a parent alkane by removing two hydrogen atoms, which has 1 to 20 (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) carbon atoms (i.e. C 1-20 alkylene), preferably 1 to 12 carbon atoms (ie C 1-12 alkylene), more preferably 1 to 6 carbon atoms (ie C 1-6 alkylene).
  • C 1-20 alkylene preferably 1 to 12 carbon atoms (ie C 1-12 alkylene), more preferably 1 to 6 carbon atoms (ie C 1-6 alkylene).
  • Non-limiting examples include: methylene (-CH 2 -), 1,1-ethylene (-CH(CH 3 )-), 1,2-ethylene (-CH 2 CH 2 )-, 1,1-propylene (-CH(CH 2 CH 3 )-), 1,2-propylene (-CH 2 CH(CH 3 )-), 1,3-propylene (-CH 2 CH 2 CH 2 -), 1,4-butylene (-CH 2 CH 2 CH 2 CH 2 -), etc.
  • the alkylene group may be substituted or unsubstituted, and when substituted, it may be substituted at any available point of attachment, the substituents being preferably selected from D atoms, halogen, alkoxy, haloalkyl, haloalkoxy, One or more of cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • heteroalkylene means that one or more -CH 2 - in the alkylene group is replaced by one or more selected from N, O, S, S(O) and S(O) 2 ; wherein
  • the alkyl group is as defined above; the heteroalkylene group can be substituted or unsubstituted, and when substituted, the substituent can be substituted at any available attachment point, and the substituent is preferably selected from D atom, halogen, Alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, and heteroaryl One or more of the bases.
  • alkenyl refers to an alkyl compound containing at least one carbon-carbon double bond in the molecule, wherein the definition of the alkyl group is as described above, and it has 2 to 12 (such as 2, 3, 4, 5, 6, 7, 8 , 9, 10, 11 or 12) carbon atoms (ie C 2-12 alkenyl).
  • the alkenyl group is preferably an alkenyl group having 2 to 6 carbon atoms (ie, a C 2-6 alkenyl group).
  • Alkenyl may be substituted or unsubstituted, and when substituted, the substituent is preferably selected from the group consisting of alkoxy, halo, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl
  • the substituent is preferably selected from the group consisting of alkoxy, halo, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl
  • radical, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl are examples of radical, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • alkynyl refers to an alkyl compound containing at least one carbon-carbon triple bond in the molecule, wherein the definition of the alkyl group is as described above, and it has 2 to 12 (such as 2, 3, 4, 5, 6, 7, 8 , 9, 10, 11 or 12) carbon atoms (ie C 2-12 alkynyl).
  • the alkynyl group is preferably an alkynyl group having 2 to 6 carbon atoms (ie, a C 2-6 alkynyl group).
  • Alkynyl may be substituted or unsubstituted, and when substituted, the substituent is preferably selected from the group consisting of alkoxy, halo, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl
  • substituent is preferably selected from the group consisting of alkoxy, halo, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl
  • radical, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl are examples of radical, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, and the cycloalkyl ring contains 3 to 20 (such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20) carbon atoms (ie 3 to 20 membered cycloalkyl), preferably containing 3 to 12 (eg 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12) carbon atoms (ie 3 to 12 membered cycloalkyl), preferably containing 3 to 8 (eg 3, 4, 5, 6, 7 and 8) carbon atoms ( i.e.
  • 3 to 8 membered cycloalkyl further preferably comprising 4 to 7 (eg 4, 5, 6 and 7) carbon atoms (ie 4 to 7 membered cycloalkyl), more preferably comprising 3 to 6 (eg 3, 4, 5 and 6) carbon atoms (ie 3 to 6 membered cycloalkyl).
  • Non-limiting examples of monocyclic cycloalkyls include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene Base, cyclooctyl, etc.; polycyclic cycloalkyl includes spirocycloalkyl, fused cycloalkyl and bridged cycloalkyl.
  • spirocycloalkyl refers to 5 to 20 membered (e.g. 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 ring atoms, i.e. 5- to 20-membered spirocycloalkyl), a polycyclic group that shares one carbon atom (called a spiro atom) between monocyclic rings, which may contain one or more double bonds. It is preferably 6 to 14 membered (ie 6 to 14 membered spirocycloalkyl), more preferably 7 to 10 membered (eg 7, 8, 9 or 10 membered, ie 7 to 10 membered spirocycloalkyl).
  • the spirocycloalkyl group can be divided into single spirocycloalkyl, double spirocycloalkyl or polyspirocycloalkyl, preferably single spirocycloalkyl and double spirocycloalkyl. More preferably, it is a 3-membered/5-membered, 3-membered/6-membered, 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered monospirocycloalkyl group.
  • Non-limiting examples of spirocycloalkyl groups include:
  • fused cycloalkyl refers to 5 to 20 membered (e.g. 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 ring atoms, i.e. 5 to 20-membered fused cycloalkyl), an all-carbon polycyclic group in which each ring in the system shares an adjacent pair of carbon atoms with other rings in the system, one or more of which may contain one or more bis key. It is preferably 6 to 14 membered (ie, 6 to 14 membered fused cycloalkyl), more preferably 7 to 10 membered (eg 7, 8, 9 or 10 membered, ie 7 to 10 membered fused cycloalkyl).
  • bicyclic, tricyclic, tetracyclic or polycyclic fused cycloalkyl preferably bicyclic or tricyclic, more preferably 3-membered/4-membered, 3-membered/5-membered, 3-membered/6-membered , 4 yuan/4 yuan, 4 yuan/5 yuan, 4 yuan/6 yuan, 5 yuan/4 yuan, 5 yuan/5 yuan, 5 yuan/6 yuan, 6 yuan/3 yuan, 6 yuan/4 yuan, 6 yuan Member/5 member and 6 member/6 member bicyclic fused cycloalkyl.
  • fused cycloalkyl groups include:
  • bridged cycloalkyl refers to 5 to 20 membered (e.g. 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 carbon atoms, i.e. 5- to 20-membered bridged cycloalkyl), any two rings share two carbon atoms not directly connected to the full-carbon polycyclic group, which may contain one or more double bonds.
  • it is 6 to 14 membered (ie, 6 to 14 membered bridged cycloalkyl group), more preferably 7 to 10 membered (eg 7, 8, 9 or 10 membered, ie 7 to 10 membered bridged cycloalkyl group).
  • bridged cycloalkyl groups preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic.
  • bridged cycloalkyl groups include:
  • the cycloalkyl ring includes a cycloalkyl group as described above (including monocyclic cycloalkyl, spirocycloalkyl, fused cycloalkyl and bridged cycloalkyl) fused to an aryl, heteroaryl or heterocycloalkane
  • a radical ring where the ring attached to the parent structure is a cycloalkyl, non-limiting examples include etc.; preferably
  • Cycloalkyl may be substituted or unsubstituted and when substituted it may be substituted at any available point of attachment, the substituents are preferably selected from halogen, alkyl, alkoxy, haloalkyl, haloalkoxy , cycloalkyloxy, heterocyclyloxy, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, and heteroaryl.
  • alkoxy refers to -O-(alkyl), wherein alkyl is as defined above.
  • alkoxy include: methoxy, ethoxy, propoxy and butoxy.
  • Alkoxy may be optionally substituted or unsubstituted, and when substituted it is preferably one or more of the following groups independently selected from D atom, halogen, alkoxy, haloalkyl, haloalkoxy , cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic ring substituent comprising 3 to 20 ring atoms (e.g. 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 ring atoms, i.e. 3 to 20 membered heterocyclyl), wherein one or more ring atoms are heteroatoms selected from nitrogen, oxygen and sulfur,
  • the sulfur can be optionally oxoated (ie to form a sulfoxide or sulfone), excluding the -O-O-, -O-S- or -S-S- ring moieties, the remaining ring atoms being carbon.
  • ring atoms ie 3 to 12 membered heterocyclyl
  • 1 to 4 eg 1 , 2, 3 and 4
  • 3 to 8 ring atoms such as 3, 4, 5, 6, 7 and 8, ie 3 to 8 membered heterocyclyl
  • 1-3 for example 1, 2 and 3
  • 4 to 7 ring atoms for example 4, 5, 6 and 7, i.e. 4 to 7 membered heterocyclyl
  • 1-3 e.g. 1, 2 and 3
  • heterocyclyl 3, 4, 5 and 6, i.e. 3 to 6 membered heterocyclyl), wherein 1-3 (e.g. 1 , 2 and 3) are heteroatoms; most preferably contain 5 or 6 ring atoms (ie 5 or 6 membered heterocyclyl), of which 1-2 (eg 1, 2) are heteroatoms.
  • monocyclic heterocyclyl groups include pyrrolidinyl, tetrahydropyranyl, 1,2,3,6-tetrahydropyridyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholine base, homopiperazinyl, etc.
  • Polycyclic heterocyclyls include spiroheterocyclyls, fused heterocyclyls and bridged heterocyclyls.
  • spiroheterocyclyl refers to 5 to 20 membered (e.g. 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 ring atoms, i.e. 5- to 20-membered spiro heterocyclic group), a polycyclic heterocyclic group that shares one atom (called spiro atom) between monocyclic rings, wherein one or more ring atoms are heteroatoms selected from nitrogen, oxygen and sulfur, so
  • the sulfur described above can be optionally oxoated (ie to form a sulfoxide or sulfone), with the remaining ring atoms being carbon. It may contain one or more double bonds.
  • the spiroheterocyclyl is 6-14 membered (ie 6-14 membered spiroheterocyclyl), more preferably 7-10 membered (eg 7, 8, 9 or 10 membered, ie 7-10 membered spiroheterocyclyl).
  • the spiroheterocyclyl can be divided into single spiroheterocyclyl, double spiroheterocyclyl or polyspiroheterocyclyl, preferably single spiroheterocyclyl and double spiroheterocyclyl.
  • spiroheterocyclyls include:
  • fused heterocyclyl refers to 5 to 20 membered (e.g. 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 ring atoms, i.e. 5 to 20-membered fused heterocyclic group), each ring in the system shares an adjacent pair of atoms with other rings in the system, and one or more rings may contain one or more double bonds,
  • One or more of the ring atoms are heteroatoms selected from nitrogen, oxygen and sulfur, said sulfur being optionally oxoated (ie to form sulfoxides or sulfones), and the remaining ring atoms are carbon.
  • it is 6 to 14 membered (ie 6 to 14 membered fused heterocyclic group), more preferably 7 to 10 membered (eg 7, 8, 9 or 10 membered, ie 7 to 10 membered fused heterocyclic group).
  • bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic groups preferably bicyclic or tricyclic, more preferably 3-membered/4-membered, 3-membered/5-membered, 3-membered/6-membered , 4 yuan/4 yuan, 4 yuan/5 yuan, 4 yuan/6 yuan, 5 yuan/4 yuan, 5 yuan/5 yuan, 5 yuan/6 yuan, 6 yuan/3 yuan, 6 yuan/4 yuan, 6 yuan 5-membered and 6-membered/6-membered bicyclic condensed heterocyclic groups.
  • fused heterocyclic groups include:
  • bridged heterocyclyl refers to 5 to 14 membered (eg 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 ring atoms, i.e. 5 to 14 membered bridged heterocyclyl), any two A polycyclic heterocyclic group in which two rings share two atoms not directly connected, which may contain one or more double bonds, wherein one or more ring atoms are heteroatoms selected from nitrogen, oxygen and sulfur, said Sulfur can be optionally oxoated (ie to form a sulfoxide or sulfone), with the remaining ring atoms being carbon.
  • it can be divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged heterocyclic groups, preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic.
  • bridged heterocyclyl groups include:
  • the heterocyclyl ring includes a heterocyclyl as described above (including monocyclic heterocyclyl, spiro heterocyclyl, fused heterocyclyl and bridged heterocyclyl) fused to an aryl, heteroaryl or cycloalkyl On the ring, where the ring attached to the parent structure is a heterocyclyl, non-limiting examples of which include:
  • the heterocyclyl group may be substituted or unsubstituted and when substituted it may be substituted at any available point of attachment, the substituents are preferably selected from halogen, alkyl, alkoxy, haloalkyl, haloalkoxy , cycloalkyloxy, heterocyclyloxy, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, and heteroaryl.
  • aryl refers to a 6 to 14 membered (e.g. 6, 7, 8, 9, 10, 11, 12, 13 or 14 ring atoms, i.e. 6 to 14 membered aryl) all group having a conjugated ⁇ electron system.
  • Carbon monocyclic or fused polycyclic (fused polycyclic rings are rings sharing adjacent pairs of carbon atoms) groups, preferably 6 to 10 membered (ie 6 to 10 membered aryl), eg phenyl and naphthyl.
  • the aryl ring includes an aryl ring as described above fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring bonded to the parent structure is an aryl ring, non-limiting examples of which include :
  • Aryl may be substituted or unsubstituted, and when substituted, it may be substituted at any available point of attachment, the substituents being preferably selected from halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, One or more of cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
  • heteroaryl refers to a group containing 1 to 4 (eg 1, 2, 3 and 4) heteroatoms, 5 to 14 ring atoms (eg 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 ring atoms, ie 5 to 14 membered heteroaryl), wherein the heteroatoms are selected from oxygen, sulfur and nitrogen.
  • Heteroaryl is preferably 5 to 10 membered (e.g. 5, 6, 7, 8, 9 or 10 membered, i.e. 5 to 10 membered heteroaryl), more preferably 5 or 6 membered (i.e. 5 or 6 membered heteroaryl).
  • Aryl such as furyl, thienyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, etc.
  • the heteroaryl ring includes a heteroaryl as described above fused to an aryl, heterocyclyl or cycloalkyl ring, wherein the ring attached to the parent structure is a heteroaryl ring, non-limiting examples of which include :
  • Heteroaryl may be substituted or unsubstituted and when substituted it may be substituted at any available point of attachment, the substituents are preferably selected from halogen, alkyl, alkoxy, haloalkyl, haloalkoxy , cycloalkyloxy, heterocyclyloxy, hydroxyl, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, and heteroaryl.
  • cycloalkyl, heterocyclyl, aryl and heteroaryl groups mentioned above include residues derived by removing one hydrogen atom from a parent ring atom, or residues derived by removing two hydrogen atoms from the same or two different ring atoms of the parent Derivative residues, i.e. "divalent cycloalkyl", “divalent heterocyclyl” (e.g. etc.), "arylene” and "heteroarylene”.
  • cycloalkylalkyl refers to an alkyl group substituted by one or more cycloalkyl groups, wherein cycloalkyl and alkyl are as defined above.
  • heterocyclylalkyl refers to an alkyl group substituted by one or more heterocyclyl groups, wherein heterocyclyl and alkyl are as defined above.
  • heteroarylalkyl refers to an alkyl group substituted by one or more heteroaryl groups, wherein heteroaryl and alkyl are as defined above.
  • cycloalkyloxy refers to cycloalkyl-O-, wherein cycloalkyl is as defined above.
  • heterocyclyloxy refers to heterocyclyl-O-, wherein heterocyclyl is as defined above.
  • alkylthio refers to alkyl-S-, wherein alkyl is as defined above.
  • haloalkyl refers to an alkyl group substituted with one or more halogens, wherein alkyl is as defined above.
  • haloalkoxy refers to an alkoxy group substituted with one or more halogens, wherein alkoxy group is as defined above.
  • alkoxyalkyl refers to an alkyl group substituted by one or more alkoxy groups, wherein alkyl and alkoxy group are as defined above.
  • hydroxyalkyl refers to an alkyl group substituted with one or more hydroxy groups, wherein alkyl is as defined above.
  • halogen refers to fluorine, chlorine, bromine or iodine.
  • hydroxyl refers to -OH.
  • mercapto refers to -SH.
  • amino refers to -NH2 .
  • cyano refers to -CN.
  • nitro refers to -NO2 .
  • aldehyde refers to -C(O)H
  • carboxylate refers to -C(O)O(alkyl), -C(O)O(cycloalkyl)(alkyl)C(O)O- or (cycloalkyl)C(O) O-, wherein alkyl and cycloalkyl are as defined above.
  • the disclosed compounds may exist in particular geometric or stereoisomeric forms.
  • This disclosure contemplates all such compounds, including cis and trans isomers, (-)- and (+)-enantiomers, (R)- and (S)-enantiomers, diastereomers isomers, (D)-isomers, (L)-isomers, and their racemic and other mixtures, such as enantiomerically or diastereomerically enriched mixtures, all of which are within the scope of this disclosure.
  • Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All such isomers, as well as mixtures thereof, are included within the scope of this disclosure.
  • Optically active (R)- and (S)-isomers as well as D and L-isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If one enantiomer of a compound of the present disclosure is desired, it can be prepared by asymmetric synthesis or derivatization with chiral auxiliary agents, wherein the resulting diastereomeric mixture is separated and the auxiliary group is cleaved to provide pure desired enantiomer.
  • a diastereoisomeric salt is formed with an appropriate optically active acid or base, and then a diastereomeric salt is formed by a conventional method known in the art. Diastereomeric resolution is performed and the pure enantiomers are recovered. Furthermore, the separation of enantiomers and diastereomers is usually accomplished by the use of chromatography using chiral stationary phases, optionally in combination with chemical derivatization methods (e.g. amines to amino groups formate).
  • the bond Indicates unassigned configuration, i.e. if chiral isomers exist in the chemical structure, the bond can be or both Two configurations.
  • the bond If the configuration is not specified, it can be the Z configuration or the E configuration, or both configurations.
  • tautomer or tautomeric form refers to structural isomers of different energies that can interconvert via a low energy barrier.
  • proton tautomers also known as prototropic tautomers
  • lactam-lactim equilibrium is between A and B as shown below.
  • the present disclosure also includes certain isotopically labeled compounds of the disclosure that are identical to those described herein, but wherein one or more atoms are replaced by an atom of an atomic mass or mass number different from that normally found in nature.
  • isotopes that can be incorporated into compounds of the present disclosure 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.
  • the compounds of the present disclosure may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute the compounds.
  • compounds can be labeled with radioactive isotopes, such as tritium ( 3 H), and heavy hydrogen can be used to replace hydrogen to form deuterated drugs.
  • radioactive isotopes such as tritium ( 3 H)
  • heavy hydrogen can be used to replace hydrogen to form deuterated drugs.
  • the bond formed by deuterium and carbon is stronger than the bond formed by ordinary hydrogen and carbon.
  • Deuterated drugs have the advantages of reducing toxic and side effects, increasing drug stability, enhancing curative effect, and prolonging the biological half-life of drugs. All permutations of isotopic composition of the disclosed compounds, whether radioactive or not, are included within the scope of the present disclosure.
  • substitution with heavier isotopes such as deuterium may confer certain therapeutic advantages resulting from greater metabolic stability (e.g. increased in vivo half-life or reduced dosage requirements), and thus in some cases
  • deuterium substitution may be partial or complete, partial deuterium substitution meaning at least one hydrogen is replaced by at least one deuterium.
  • deuterium when a position is specifically designated as deuterium (D), the position is understood to have an abundance of deuterium (i.e., at least 10 % deuterium incorporation).
  • exemplary compounds having a natural abundance greater than deuterium can be at least 1000 times more abundant deuterium, at least 2000 times more abundant deuterium, at least 3000 times more abundant deuterium, at least 4000 times more abundant deuterium, at least 5000 times more abundant deuterium, at least 6000 times more abundant deuterium, or more abundant deuterium.
  • the present disclosure also includes various deuterated forms of compounds of formula (I). Each available hydrogen atom attached to a carbon atom can be independently replaced by a deuterium atom.
  • deuterated starting materials can be used in the preparation of deuterated forms of compounds of formula (I), or they can be synthesized using conventional techniques using deuterated reagents, including but not limited to deuterated borane, trideuterated Borane tetrahydrofuran solution, deuterated lithium aluminum hydride, deuterated ethyl iodide and deuterated methyl iodide, etc.
  • Optional or “optionally” means that the subsequently described event or circumstance can but need not occur, and that the description includes instances where the event or circumstance occurs or does not occur.
  • a heterocyclic group optionally substituted with an alkyl group means that an alkyl group may but need not be present, and the description includes cases where the heterocycle group is substituted with an alkyl group and cases where the heterocycle group is not substituted with an alkyl group .
  • Substituted means that one or more hydrogen atoms in a group, preferably 1 to 5, more preferably 1 to 3 hydrogen atoms are independently substituted by a corresponding number of substituents. Possible or impossible substitutions can be determined (by experiment or theory) by those skilled in the art without undue effort. For example, an amino or hydroxyl group with free hydrogen may be unstable when bonded to a carbon atom with an unsaturated (eg, ethylenic) bond.
  • “Pharmaceutical composition” means a mixture containing one or more compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, and other components such as a physiologically/pharmaceutically acceptable carrier and excipients.
  • the purpose of the pharmaceutical composition is to promote the administration to the organism, facilitate the absorption of the active ingredient and thus exert biological activity.
  • “Pharmaceutically acceptable salt” refers to the salt of the disclosed compound, which is safe and effective when used in mammals, and has proper biological activity. Salts can be prepared separately during the final isolation and purification of the compounds, or by reacting the appropriate group with a suitable base or acid.
  • Bases commonly used to form pharmaceutically acceptable salts include inorganic bases, such as sodium hydroxide and potassium hydroxide, and organic bases, such as ammonia. Acids commonly used to form pharmaceutically acceptable salts include inorganic acids as well as organic acids.
  • the terms “therapeutically effective amount”, “inhibitory effective amount” or “prophylactically effective amount” refer to the amount of drug or agent that is sufficient to achieve or partially achieve the desired effect.
  • the determination of the effective amount varies from person to person, depending on the age and general condition of the recipient, and also depends on the specific active substance. The appropriate effective amount in each case can be determined by those skilled in the art according to routine experiments.
  • the term "pharmaceutically acceptable” means those compounds, materials, compositions and/or dosage forms, which are suitable for use in contact with patient tissues without undue toxicity, irritation, allergic reaction or Other problems or complications that have a reasonable benefit/risk ratio and are valid for the intended use.
  • the compound represented by general formula (I) of the present disclosure, or the preparation method of pharmaceutically acceptable salt thereof, comprises the following steps:
  • L, ring A, ring B, R 1 to R 4 , p and q are as defined in the general formula (I).
  • the preparation method of the compound represented by the general formula (II) of the present disclosure, or a pharmaceutically acceptable salt thereof, comprises the following steps:
  • Ring A, ring B, ring C, L, R 1 , R 2 , R 4a , p, q and n are as defined in the general formula (II).
  • the preparation method of the compound represented by the general formula (Ii) of the present disclosure, or a pharmaceutically acceptable salt thereof, comprises the following steps:
  • Ring A, ring B, ring C, L, R 1 , R 2 , R 4a , p, q and n are as defined in the general formula (Ii).
  • the preparation method of the compound represented by the general formula (III) of the present disclosure or a pharmaceutically acceptable salt thereof comprises the following steps:
  • L, X, R 1 , R 4a , R c , R d , p, n and s are as defined in general formula (III).
  • the preparation method of the compound represented by the general formula (IV) of the present disclosure or a pharmaceutically acceptable salt thereof comprises the following steps:
  • X, L, R 1 , R 4a , p, n and s are as defined in general formula (IV).
  • the preparation method of the compound represented by the general formula (II) of the present disclosure or a pharmaceutically acceptable salt thereof comprises the following steps:
  • the first step the compound represented by the general formula (IIA') or its salt and the compound represented by the general formula (IIB') or its salt, under the action of a base (such as n-butyllithium), nucleophilic addition occurs Reaction, obtains the compound or its pharmaceutically acceptable salt shown in general formula (IIa);
  • a base such as n-butyllithium
  • Second step the compound represented by general formula (IIa) or its salt undergoes chlorination reaction (for example under the effect of PCl or SOCl 2 ), to obtain the compound represented by general formula (IIb) or its pharmaceutically acceptable salt ;
  • the third step the compound represented by the general formula (IIb) or its pharmaceutically acceptable salt undergoes a reduction reaction under the action of a metal (such as zinc powder or iron powder) to obtain the compound represented by the general formula (II) or its pharmaceutically acceptable salt the salt used;
  • a metal such as zinc powder or iron powder
  • X is halogen , preferably bromine
  • Ring A, ring B, ring C, L, R 1 , R 2 , R 4a , p, q and n are as defined in the general formula (II).
  • the preparation method of the compound represented by the general formula (Ii) of the present disclosure or a pharmaceutically acceptable salt thereof comprises the following steps:
  • Ring D is a 3- to 8-membered heterocyclic group containing at least one double bond in the ring, preferably a 5-membered or 6-membered heterocyclic group containing at least one double bond in the ring, more preferably
  • Ring C is a 3- to 8-membered heterocyclic group, preferably a 5-membered or 6-membered heterocyclic group, more preferably
  • X is halogen , preferably bromine
  • Ring A, ring B, L, R 1 , R 2 , R 4a , p, q and n are as defined in the general formula (Ii).
  • the bases include organic bases and inorganic bases
  • the organic bases include but not limited to triethylamine, pyridine, 3,5-lutidine, N,N- Diisopropylethylamine, n-butyllithium, lithium diisopropylamide, lithium bis(trimethylsilyl)amide, sodium acetate, potassium acetate, sodium tert-butoxide, potassium tert-butoxide or 1,8 -diazabicycloundec-7-ene
  • the inorganic bases include but not limited to sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide and potassium hydroxide ; the base described in scheme one to four, preferably selected from pyridine, bis(trimethylsilyl)amide lithium and 3,5-lutidine; the base described in scheme five, preferably n-butyl Lithium; the base described in scheme six, preferably potassium carbonate.
  • the metal catalysts include but are not limited to palladium acetate, tetrakis(triphenylphosphine)palladium, tris(dibenzylideneacetone)dipalladium, 1,1'-bis(diphenylphosphino)bis Ferrocenepalladium(II)chloride, bis(acetonitrile)palladium(II)chloride and palladium on carbon, preferably palladium acetate.
  • the ligands include but not limited to triphenylphosphine, tri(o-tolyl)phosphine and 1,1'-binaphthyl-2,2'-bisdiphenylphosphine (BINAP), preferably Triphenylphosphine.
  • the reaction temperature is 100-150°C, preferably 120°C.
  • the reaction time is 0.5-6 hours, preferably 2-3 hours, more preferably 3 hours.
  • the reactions of the above schemes 1 to 6 are preferably carried out in a solvent, and the solvents used include but are not limited to: ethylene glycol dimethyl ether, acetic acid, methanol, ethanol, acetonitrile, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether , ethyl acetate, n-hexane, dimethyl sulfoxide, 1,4-dioxane, water, N,N-dimethylformamide, N,N-dimethylacetamide, 1,2-di Bromoethane, pyridine and mixtures thereof.
  • the solvents used include but are not limited to: ethylene glycol dimethyl ether, acetic acid, methanol, ethanol, acetonitrile, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether , ethyl acetate,
  • NMR nuclear magnetic resonance
  • MS mass spectroscopy
  • MS was determined with Agilent 1200/1290 DAD-6110/6120 Quadrupole MS liquid mass spectrometer (manufacturer: Agilent, MS model: 6110/6120 Quadrupole MS), waters ACQuity UPLC-QD/SQD (manufacturer: waters, MS Model: waters ACQuity Qda Detector/waters SQ Detector), THERMO Ultimate 3000-Q Exactive (manufacturer: THERMO, MS model: THERMO Q Exactive).
  • HPLC High performance liquid chromatography
  • Chiral HPLC analysis was performed using an Agilent 1260 DAD high performance liquid chromatograph.
  • Chiral preparative chromatography uses a Shimadzu LC-20AP preparative chromatograph.
  • the CombiFlash rapid preparation instrument uses Combiflash Rf200 (TELEDYNE ISCO).
  • the thin-layer chromatography silica gel plate uses Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate.
  • the specification of the silica gel plate used in thin-layer chromatography (TLC) is 0.15mm-0.2mm, and the specification of thin-layer chromatography separation and purification products is 0.4mm. ⁇ 0.5mm.
  • Silica gel column chromatography generally uses Yantai Huanghai silica gel 200-300 mesh silica gel as the carrier.
  • the known starting materials of the present disclosure can be adopted or synthesized according to methods known in the art, or can be purchased from ABCR GmbH&Co.KG, Acros Organics, Aldrich Chemical Company, Shaoyuan Technology (Shanghai) Co., Ltd., Darui Chemical Products, Shanghai Titan Technology, Aladdin, Anaiji Chemicals, China National Pharmaceutical Group Co., Ltd., Adamas Reagent Co., Ltd., Sigma-Aldrich (Shanghai) Trading Co., Ltd., Shanghai Pide Pharmaceutical Technology Co., Ltd., Shanghai Haohong Bio Pharmaceutical Technology Co., Ltd., Thermo Fisher Scientific (China) Technology Co., Ltd. and other companies.
  • the reactions can all be carried out under an argon atmosphere or a nitrogen atmosphere.
  • the argon atmosphere or nitrogen atmosphere means that the reaction bottle is connected to an argon or nitrogen balloon with a volume of about 1 L.
  • the hydrogen atmosphere means that the reaction bottle is connected to a hydrogen balloon with a capacity of about 1L.
  • the pressurized hydrogenation reaction uses Parr 3916EKX hydrogenation instrument and Qinglan QL-500 hydrogen generator or HC2-SS hydrogenation instrument.
  • the hydrogenation reaction is usually vacuumized and filled with hydrogen, and the operation is repeated 3 times.
  • the solution refers to an aqueous solution.
  • reaction temperature is room temperature, which is 20°C to 30°C.
  • the monitoring of the reaction process in the embodiment adopts thin-layer chromatography (TLC), the developer used for reaction, the eluent system of the column chromatography that purifies compound adopts and the developer system of thin-layer chromatography comprise: A: n-hexane/ethyl acetate system, B: dichloromethane/methanol system, the volume ratio of the solvent is adjusted according to the polarity of the compound, and can also be adjusted by adding a small amount of basic or acidic reagents such as triethylamine and acetic acid.
  • TLC thin-layer chromatography
  • reaction solution was cooled to room temperature, filtered under reduced pressure, the filtrate was diluted with ethyl acetate (500 mL), washed with saturated sodium chloride solution (30 mL ⁇ 5), the obtained organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure , to obtain the title product 1b (36.9 g, yield: 100%).
  • the product was directly used in the next reaction without further purification.
  • reaction solution was quenched with water (10 mL), extracted with ethyl acetate (50 mL ⁇ 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the obtained product was purified by silica gel column chromatography with eluent system A As a residue, the title product If (1.11 g, yield: 94.7%) was obtained.
  • reaction solution was quenched with water (5 mL), concentrated under reduced pressure, diluted with ethyl acetate (150 mL), washed with saturated sodium chloride solution (10 mL ⁇ 2).
  • the obtained organic phase was dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the obtained residue was purified by silica gel column chromatography with eluent system A to obtain the title product 1 g (800 mg, yield: 71.8%).
  • reaction solution was cooled to room temperature, concentrated under reduced pressure, diluted with ethyl acetate (150 mL), washed with saturated sodium chloride solution (20 mL ⁇ 3), and the obtained organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure and used The resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 1j (289 mg, yield: 51.4%).
  • reaction solution was cooled to room temperature, quenched with water (1 mL), diluted with ethyl acetate (100 mL), washed with saturated sodium chloride solution (20 mL ⁇ 2), and the obtained organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was decompressed Concentrated, and the resulting residue was purified by silica gel column chromatography with eluent system A to obtain the title product 1k (350 mg, yield: 91.9%).
  • reaction solution was cooled to room temperature, quenched with water (1 mL), diluted with ethyl acetate (100 mL), washed with saturated sodium chloride solution (50 mL ⁇ 2), and the obtained organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was decompressed After concentration, the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 2d (1.84 g, yield: 99.5%).
  • reaction solution was cooled to room temperature, the reaction solution was filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to obtain the title product 4d (503 mg, yield: 87.2%).
  • reaction solution was cooled to room temperature, quenched with water (1 mL), diluted with ethyl acetate (30 mL), washed with saturated sodium chloride solution (30 mL ⁇ 2), combined organic phases, dried over anhydrous sodium sulfate, filtered, and the filtrate was Concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to obtain the title product 4e (323 mg, yield: 73.2%).
  • reaction solution was quenched with saturated sodium bisulfite solution (20mL), washed with saturated sodium chloride solution (60mL ⁇ 2), combined organic phases, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and purified by silica gel column chromatography The resulting residue was purified with eluent system A to give the title product 5b (4.8 g, yield: 51.7%).
  • reaction solution was cooled to room temperature, quenched with water (5 mL), diluted with ethyl acetate (60 mL), washed with saturated sodium chloride solution (30 mL ⁇ 2), combined organic phases, dried over anhydrous sodium sulfate, filtered, and the filtrate was decompressed After concentration, the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 5c (3.2 g, yield: 73.2%).
  • 4-bromo-3-methoxyaniline 11a (2.0 g, 9.90 mmol), 37% aqueous formaldehyde (9.0 g, 97.8 mmol), acetic acid (9.0 g, 150 mmol), acetonitrile (30 mL), room temperature Stir for 30 minutes, add sodium cyanoborohydride (800 mg, 12.7 mmol) under ice-cooling, and stir at room temperature for 16 hours.
  • reaction solution was diluted with ethyl acetate (100 mL), then washed with water (100 mL) and saturated aqueous sodium chloride solution (100 mL), respectively, concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to obtain the title Product 14c (2.4 g, 1 NMR showed ⁇ 10% Br in the isopropyl para isomer, overall yield: 77.7%, sent directly to the next step).
  • reaction solution was washed with saturated sodium bisulfite solution (20 mL) and saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to obtain the title product 17b (390.0 mg, yield: 78.0%).
  • 2-Methoxy-4-methylbenzenesulfonyl chloride 22a (78mg, 35mmol) and compound 1n (30mg, 0.12mmol) were dissolved in pyridine (5.0mL), and 4-dimethylaminopyridine (3mg, 0.02mmol ), replacing nitrogen 3 times.
  • the reaction solution was microwaved at 120° C. for 3 hours.
  • test examples are not meant to limit the scope of the present disclosure.
  • Test example 1 KAT6 enzyme activity detection (AlphaScreen method)
  • EDTA 0.5M
  • pH 8.0 pH 8.0
  • RNase-free Thermofisher, AM9260G
  • Acetyl-CoA (Ac-CoA, CAYMAN, Cat.No.16160)
  • AlphaScreen Streptavidin Donor beads (AlphaScreen Streptavidin Donor beads) 5mg (PerkinElmer, 6760002)
  • AlphaScreen Protein A Acceptor beads (AlphaScreen Protein A Acceptor beads), 5mg (PerkinElmer, 6760137M)
  • Acetylated-Lysine Antibody (Acetylated-Lysine Antibody#9441) (CST 9441S)
  • 1 ⁇ detection buffer 100mM Tris-HCL, pH7.8; 15mM NaCl; 1mM EDTA; 0.01% Tween-20; 1mM DTT; 0.01% m/v ovalbumin.
  • KAT enzyme solution 1 ⁇ detection buffer to prepare a final concentration of 1.25nM.
  • Ac-CoA and H3 mixed substrate Prepare a mixed substrate of Ac-CoA with a final concentration of 1000 nM and H3 with a final concentration of 55 nM in 1 ⁇ detection buffer.
  • Detection reagent AlphaScreen protein A acceptor beads with a final concentration of 8 ng/ ⁇ L prepared in 1 ⁇ detection buffer, AlphaScreen streptavidin donor beads at 8 ng/ ⁇ L, acetylated lysine antibody diluted 1:1500, 100 ⁇ M Anacardic acid.
  • the disclosed compound has a good inhibitory effect on KAT6A.
  • Test example 2 KAT6B enzyme activity detection (AlphaScreen method)
  • Bovine serum albumin (Sanko, A500023-0100)
  • Acetyl-CoA (Ac-CoA, CAYMAN, Cat.No.16160)
  • AlphaScreen protein A receptor beads 5 mg (PerkinElmer, 6760137M)
  • a. 1 ⁇ buffer 2 50mM Tris-HCl, pH7.8; 0.1mM EDTA; 0.01% v/v Tween-20; 1mM DTT; 0.01% m/v bovine serum albumin.
  • c.Ac-CoA and H3 mixed substrate buffer 2 to prepare a final concentration of 30nM Ac-CoA and 30nM H3 mixed substrate.
  • Detection reagent Prepare AlphaScreen protein A acceptor beads with a final concentration of 8ng/ ⁇ L in buffer 2, AlphaScreen streptavidin donor beads with a final concentration of 8ng/ ⁇ L, acetylated lysine antibody diluted 1:1000, and 100 ⁇ M anacardic acid .
  • the disclosed compound has a good inhibitory effect on KAT6B.
  • Test example 3 U2OS cell H3K23 acetylation IF detection (Immunofluorescence)
  • Bovine serum albumin (BSA) (Sangon Biotech, A500023-0100)
  • Triton X-100 (Solarbio, T8200)
  • Blocking buffer PBS (Shanghai Yuanpei) + BSA (final concentration 1%) + Triton X-100 (final concentration 0.5%).
  • Washing buffer PBS (20 ⁇ PBS diluted to 1 ⁇ PBS)+Tween-20 (final concentration 0.1%).
  • Secondary antibody solution goat anti-rabbit IgG (H+L), Superclonal TM recombinant secondary antibody, Alexa Fluor 488 at a dilution ratio of 1:1000, Hoechst 33342 at a dilution ratio of 1:5000, diluted with blocking buffer.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

提供了一种通式(I)所示的磺酰胺衍生物、其制备方法及含有该衍生物的药物组合物及其作为治疗剂的用途,特别是作为赖氨酸乙酰基转移酶(KAT)抑制剂的用途和在制备用于治疗和/或预防癌症的药物中的用途。

Description

磺酰胺衍生物、其制备方法及其在医药上的应用 技术领域
本公开属于医药领域,涉及一种磺酰胺衍生物、其制备方法及其在医药上的应用。特别地,本公开涉及通式(I)所示的磺酰胺衍生物、其制备方法及含有该衍生物的药物组合物,以及其作为KAT抑制剂在制备治疗和/或预防癌症的药物中的用途。
背景技术
赖氨酸乙酰基转移酶(Lysine acetyltransferases;KATs)是一类能够催化乙酰基从乙酰辅酶A上转移到蛋白质底物中赖氨酸ε-氨基上的酶。赖氨酸的乙酰化能够对蛋白质的功能产生影响,从而在染色体结构、基因转录调控、DNA结合能力、酶活性及稳定性、蛋白质相互作用及胞内定位中发挥着重要调节作用。KATs分为数个亚家族,其中MYST(MOZ,YBF2/SAS3,SAS2,TIP60)是最大的一个,包括KAT5(TIP60),KAT6A(MOZ;MYST3),KAT6B(MORF;MYST4),KAT7(HBO;MYST2)和KAT8(MOF;MYST1)。KAT6A/B作为MYST家族中的主要成员,对发育,造血和免疫系统中的干细胞维持,和肿瘤发生发展及耐药,起着至关重要的作用。
TCGA数据库分析表明,KAT6A和KAT6B在多种肿瘤中均有扩增。其中KAT6A位于染色体8p11-p12扩增子区域,在10-15%的乳腺癌中扩增,其拷贝数与mRNA表达正相关,且与愈后不良相关。而KAT6A和KAT6B都在乳腺癌中显著高表达。进一步亚型分析显示,KAT6A/B高表达和ERα表达水平之间有一定的相关性,揭示KAT6A/B可能为ER +/HER2 -乳腺癌的潜在靶点。
文献报道,在KAT6A扩增的luminal型乳腺癌细胞SUM-52中敲低KAT6A,克隆形成相比非成瘤细胞MCF10A有明显抑制。而RNAseq分析显示,KAT6A敲低后一些基因发生下调,其中包括ESR1和荷尔蒙应激通路相关基因。进一步的研究表明,在KAT6A高表达的ER +乳腺癌细胞系T47D、CAMA1中,KAT6A的敲低可以抑制克隆形成,但是在低表达的细胞系MCF7、SKBR3中则无此现象。T47D、CAMA1中敲低KAT6A可以下调ERα的表达,反之,MCF7、LY2中过表达野生型KAT6A可以上调ERα,而KAT活性丢失的突变型则无此作用,揭示了KAT功能的重要性。在T47D中过表达ERα,能够逆转KAT6A敲低对克隆形成的抑制作用,说明KAT6A的功能可能是通过对ERα表达的调控介导的。与此一致,在ESR1基因的启动子区域发现了KAT6A的富集。T47D模型体内药效实验也看到了KAT6A敲低的抑瘤效果和对ERα的下调。在T47D中,KAT6A和KAT6B的敲低都可以使ERα表达下调并抑制克隆形成,而且KAT6A作用大于KAT6B,如果两 者同时敲低,则作用更明显,展现了叠加的效果。KAT6A/B选择性抑制剂CTx-648在体外和体内均显示了在ER+乳腺癌中的抗肿瘤活性,且KAT6A的表达水平与CTx-648的敏感性有一定相关性。在KAT6A高表达的ER +乳腺癌细胞中,CTx-648可以下调ERα表达,H3K23Ac则可作为KAT6抑制剂的药效动力学生物标志物。综上所述,KAT6A/B抑制剂作为单药或与ER +/HER2 -乳腺癌现有疗法比如氟维司群、CDK4/6抑制剂,乃至SERD、SERCA等联用,是有临床开发价值的。
除了ER +/HER2 -乳腺癌之外,KAT6A/B抑制剂在脑胶质瘤、B细胞淋巴瘤、肝癌、卵巢癌等瘤种中均具有潜在的应用前景,可作为适应症扩展。
公开的KAT6的抑制剂专利申请包括WO2016198507A1、WO2019243491A1、WO2019043139A1、WO2019108824A1、WO2020216701A1、WO2020002587A1、WO2020254946A1和WO2020254989A1等。
发明内容
本公开的目的在于提供一种通式(I)所示的化合物或其可药用的盐:
Figure PCTCN2022111395-appb-000001
其中:
环A选自环烷基、杂环基、芳基和杂芳基;
环B为环烷基或杂环基;
L为化学键、亚烷基或杂亚烷基;其中所述的亚烷基或杂亚烷基各自独立地任选被选自羟基、卤素、烷基、卤代烷基、羟烷基、烷氧基和卤代烷氧基中的一个或多个取代基所取代;
各个R 1、各个R 2、R 3相同或不同,且各自独立地选自氢原子、卤素、氰基、硝基、氧代基、烯基、炔基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-OR 5、-C(O)R 6、-C(O)OR 6、-OC(O)R 6、-NHC(O)OR 6、-NR 7R 8、-C(O)NR 7R 8、-S(O) rR 6和-S(O) rNR 7R 8;其中所述的烯基、炔基、烷基、烷氧基、环烷基、杂环基、芳基和杂芳基各自独立地任选被选自羟基、卤素、氰基、氨基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基和杂环基氧基中的一个或多个取代基所取代;
R 4为氢原子或
Figure PCTCN2022111395-appb-000002
环C选自环烷基、杂环基、芳基和杂芳基;
R 0选自氢原子、羟基、卤素、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基和环烷基;
各个R 4a相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基和-NR 9R 10
R 5、R 6相同或不同,且各自独立地选自氢原子、烯基、炔基、烷基、环烷基、杂环基、芳基和杂芳基;其中所述的烯基、炔基、烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被选自羟基、卤素、氰基、氨基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基和杂环基氧基中的一个或多个取代基所取代;
R 7、R 8、R 9、R 10相同或不同,且各自独立地选自氢原子、烷基、环烷基、杂环基、芳基和杂芳基;其中所述的烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被选自羟基、卤素、氰基、氨基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基和杂环基氧基中的一个或多个取代基所取代;
或者,R 7和R 8与相连接的N原子一起形成一个杂环基,或者,R 9和R 10与相连接的N原子一起形成一个杂环基,所述的杂环基任选被选自羟基、卤素、氰基、氨基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基和杂环基氧基中的一个或多个取代基所取代;
p为0、1、2、3或4;
q为0、1、2、3或4;
m为0、1、2、3或4;
n为0、1、2、3或4;且
r为0、1或2。
在本公开一些的实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中:
环A选自环烷基、杂环基、芳基和杂芳基;
环B选自环烷基或杂环基;
L为化学键、亚烷基或杂亚烷基;其中所述的亚烷基或杂亚烷基各自独立地任选被选自羟基、卤素、烷基、卤代烷基、羟烷基、烷氧基和卤代烷氧基中的一个或多个取代基所取代;
各个R 1、各个R 2、R 3相同或不同,且各自独立地选自氢原子、卤素、氰基、 硝基、氧代基、烯基、炔基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-OR 5、-C(O)R 6、-C(O)OR 6、-OC(O)R 6、-NHC(O)OR 6、-NR 7R 8、-C(O)NR 7R 8、-S(O) rR 6和-S(O) rNR 7R 8;其中所述的烯基、炔基、烷基、烷氧基、环烷基、杂环基、芳基和杂芳基各自独立地任选被选自羟基、卤素、氰基、氨基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基和杂环基氧基中的一个或多个取代基所取代;
R 4为氢原子或
Figure PCTCN2022111395-appb-000003
环C为芳基或杂芳基;
R 0选自氢原子、羟基、卤素、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基和环烷基;
各个R 4a相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基和-NR 9R 10
R 5、R 6相同或不同,且各自独立地选自氢原子、烯基、炔基、烷基、环烷基、杂环基、芳基和杂芳基;其中所述的烯基、炔基、烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被选自羟基、卤素、氰基、氨基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基和杂环基氧基中的一个或多个取代基所取代;
R 7、R 8、R 9、R 10相同或不同,且各自独立地选自氢原子、烷基、环烷基、杂环基、芳基和杂芳基;其中所述的烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被选自羟基、卤素、氰基、氨基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基和杂环基氧基中的一个或多个取代基所取代;
或者,R 7和R 8与相连接的N原子一起形成一个杂环基,或者R 9和R 10与相连接的N原子一起形成一个杂环基,所述的杂环基任选被选自羟基、卤素、氰基、氨基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基和杂环基氧基中的一个或多个取代基所取代;
p为0、1、2、3或4;
q为0、1、2、3或4;
m为0、1、2、3或4;
n为0、1、2、3或4;且
r为0、1或2。
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中R 3选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基和3至8元环烷基;优选地,R 3为氢原子。
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中环C选自3至8元环烷基、3至8元杂环基、6至10元芳基和5至10元杂芳基;优选地,环C为5至10元杂芳基或6至10元芳基;更优选地,环C为5元或6元杂芳基;最优选地,环C为吡唑基。
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中环C为5元或6元杂环基、5元或6元杂芳基;优选地,环C选自吡唑基、吡啶基、呋喃基和四氢呋喃基;更优选地,环C为吡唑基。
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中环C选自
Figure PCTCN2022111395-appb-000004
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中R 4
Figure PCTCN2022111395-appb-000005
环C选自3至8元环烷基、3至8元杂环基、6至10元芳基和5至10元杂芳基;R 0、R 4a、m和n如通式(I)中所定义。
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中R 4
Figure PCTCN2022111395-appb-000006
环C为5至10元杂芳基或6至10元芳基;R 0、R 4a、m和n如通式(I)中所定义。
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中R 0选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基和3至8元环烷基;优选地,R 0选自氢原子、羟基、卤素和C 1-6烷基;优选地,R 0选自氢原子、羟基和Cl;更优选地,R 0为氢原子。
本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中m为0或1,优选为1。
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中R 4
Figure PCTCN2022111395-appb-000007
环C为5至10元杂芳基,优选为5元或6元杂芳基;R 4a和n如通式(I)中所定义;
优选地,R 4
Figure PCTCN2022111395-appb-000008
R 4a和n如通式(I)中所定义;
更优选地,R 4
Figure PCTCN2022111395-appb-000009
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其为通式(II)所示的化合物或其可药用的盐:
Figure PCTCN2022111395-appb-000010
其中:
环C为5至10元杂芳基,优选为5元或6元杂芳基;
环A、环B、L、R 1、R 2、R 4a、p、q和n如通式(I)中所定义。
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中R 4
Figure PCTCN2022111395-appb-000011
环C为3至8元杂环基或5至10元杂芳基,优选为5元或6元杂环基、5元或6元杂芳基;R 4a和n如通式(I)中所定义;
优选地,R 4
Figure PCTCN2022111395-appb-000012
R 4a和n如通式(I)中所定义;
进一步优选地,R 4
Figure PCTCN2022111395-appb-000013
R 4a和n如通式(I)中所定义;
更优选地,R 4
Figure PCTCN2022111395-appb-000014
更进一步优选地,R 4
Figure PCTCN2022111395-appb-000015
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其为通式(Ii)所示的化合物或其可药用的盐:
Figure PCTCN2022111395-appb-000016
其中:
环C为3至8元杂环基或5至10元杂芳基,优选为5元或6元杂环基、5元或6元杂芳基;
环A、环B、L、R 1、R 2、R 4a、p、q和n如通式(I)中所定义。
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中R 4
Figure PCTCN2022111395-appb-000017
优选地,R 4
Figure PCTCN2022111395-appb-000018
在本公开一些实施方案中,所述的通式(I)、通式(II)或通式(Ii)所示的化合物或其可药用的盐,其中环A为6至10元芳基或5至10元杂芳基,所述6至10元芳基优选为苯基或萘基;所述5至10元杂芳基优选为吡啶基、喹啉基和苯并噁唑基。
在本公开一些实施方案中,所述的通式(I)、通式(II)或通式(Ii)所示的化合物或其可药用的盐,其中环A为6至10元芳基或5至10元杂芳基,所述6至10元芳基优选选自苯基、萘基、
Figure PCTCN2022111395-appb-000019
所述5至10元杂芳基优选为吡啶基、喹啉基和苯并噁唑基。
在本公开一些实施方案中,所述的通式(I)、通式(II)或通式(Ii)所示的化合物或其可药用的盐,其中环A为6至10元芳基,优选选自苯基、
Figure PCTCN2022111395-appb-000020
为苯基;更优选为苯基。
在本公开一些实施方案中,所述的通式(I)、通式(II)或通式(Ii)所示的化合物或其可药用的盐,其中
Figure PCTCN2022111395-appb-000021
选自
Figure PCTCN2022111395-appb-000022
p1为0、1、2或3;R 1和p如通式(I)中所定义。
在本公开一些实施方案中,所述的通式(I)、通式(II)或通式(Ii)所示的化合物或其可药用的盐,其中
Figure PCTCN2022111395-appb-000023
Figure PCTCN2022111395-appb-000024
R 1和p如通式(I)中所定义。
在本公开一些实施方案中,所述的通式(I)、通式(II)或通式(Ii)所示的化合物或其可药用的盐,其中环B为3至8元杂环基或3至8元环烷基;
优选地,环B为4至7元杂环基或4至7元环烷基;
进一步优选地,环B为4至7元杂环基;
更进一步优选地,环B为4至7元杂环基,其中所述4至7元杂环基内含有1-3个氧原子;
最优选地,环B为5元或6元杂环基,其中所述5元或6元杂环基内含有1或2个氧原子。
在本公开一些实施方案中,所述的通式(I)、通式(II)或通式(Ii)所示的化合物或 其可药用的盐,其中环B选自
Figure PCTCN2022111395-appb-000025
优选为
Figure PCTCN2022111395-appb-000026
R 2可取代在环B任意可取代位置。
在本公开一些实施方案中,所述的通式(I)、通式(II)或通式(Ii)所示的化合物或其可药用的盐,其中环B选自
Figure PCTCN2022111395-appb-000027
优选地,环B选自
Figure PCTCN2022111395-appb-000028
更优选地,环B为
Figure PCTCN2022111395-appb-000029
R 2可取代在环B任意可取代位置。
在本公开一些实施方案中,所述的通式(I)或通式(II)所示的化合物或其可药用的盐,其为通式(III)所示的化合物或其可药用的盐:
Figure PCTCN2022111395-appb-000030
其中:
X选自O、CR aR b和C=O;
各个R a、R b、R c和R d相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、氨基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基氧基和杂环基氧基;
或者,R c、R d与所连碳原子一起形成一个C=O;
s为0、1、2或3;
L、R 1、R 4a、p和n如通式(I)中所定义。
在本公开一些实施方案中,所述的通式(III)所示的化合物或其可药用的盐,其中各个R c和R d相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、氨基、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、3至8元环烷基、3至8元杂环基、3至8元环烷基氧基和3至8元杂环基氧基;或者,R c、R d与所连碳原子一起形成一个C=O;
优选地,各个R c和R d相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基和C 1-6烷氧基;或者,R c、R d与所连碳原子一起形成一个C=O;
进一步优选地,各个R c和R d相同或不同,且各自独立地选自氢原子、卤素、 C 1-6烷基和C 1-6烷氧基;
再进一步优选地,各个R c和R d相同或不同,且各自独立地为氢原子或卤素;
甚至进一步优选地,各个R c和R d相同或不同,且各自独立地为氢原子或氟原子;
最优选地,R c和R d均为氢原子。
在本公开一些实施方案中,所述的通式(I)、通式(II)或通式(III)所示的化合物或其可药用的盐,其为通式(IV)所示的化合物或其可药用的盐:
Figure PCTCN2022111395-appb-000031
其中:
L、X、R 1、R 4a、p、n和s如通式(III)中所定义。
在本公开一些实施方案中,所述的通式(III)或通式(IV)所示的化合物或其可药用的盐,其中X为O或CR aR b;各个R a和R b相同或不同,各自独立地选自氢原子、羟基、卤素、氰基、氨基、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、3至8元环烷基、3至8元杂环基、3至8元环烷基氧基和3至8元杂环基氧基;优选地,各个R a和R b相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基和C 1-6烷氧基;进一步优选地,各个R a和R b相同或不同,且各自独立地选自氢原子、卤素、C 1-6烷基和C 1-6烷氧基;最优选地,R a和R b均为氢原子。
在本公开一些实施方案中,所述的通式(III)或通式(IV)所示的化合物或其可药用的盐,其中X为O或CH 2;优选地,X为CH 2
在本公开一些实施方案中,所述的通式(III)或通式(IV)所示的化合物或其可药用的盐,其中s为1或2,优选为1。
在本公开一些实施方案中,所述的通式(III)所示的化合物或其可药用的盐,其中R c和R d相同或不同,且各自独立地为氢原子或卤素;和/或X为O或CH 2;和/或s为1或2。
在本公开一些实施方案中,所述的通式(IV)所示的化合物或其可药用的盐,其中X为O或CH 2;和/或s为1或2。
在本公开一些实施方案中,所述的通式(I)、通式(II)或通式(Ii)所示的化合物或其可药用的盐,其中q为0、1或2;优选地,q为0。
在本公开一些实施方案中,所述的通式(I)、通式(II)或通式(Ii)所示的化合物或其可药用的盐,其中q为1。
在本公开一些实施方案中,所述的通式(I)、通式(II)、通式(Ii)、通式(III)或通式(IV)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、氨基、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、氧代基、3至8元环烷基、C 1-6烷氧基C 1-6烷基、-OR 5、-C(O)R 6、-C(O)OR 6、-C(O)NR 7R 8和-S(O) rR 6,r、R 5、R 6、R 7和R 8如通式(I)中所定义;优选地,各个R 1相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6烷氧基C 1-6烷基和-C(O)OCH 3;更优选地,各个R 1相同或不同,且各自独立地为C 1-6烷氧基;最优选地,R 1为甲氧基。
在本公开一些实施方案中,所述的通式(I)、通式(II)、通式(Ii)、通式(III)或通式(IV)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、氧代基、3至8元环烷基、C 1-6烷氧基C 1-6烷基、-NR 7R 8、-OR 5、-C(O)R 6、-C(O)OR 6、-C(O)NR 7R 8和-S(O) rR 6,r、R 5、R 6、R 7和R 8如通式(I)中所定义;
优选地,各个R 1相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6烷氧基C 1-6烷基、-C(O)OCH 3和-NR 7R 8;R 7和R 8相同或不同,且各自独立地为氢原子或C 1-6烷基;
进一步优选地,各个R 1相同或不同,且各自独立地选自氢原子、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基和-NR 7R 8;R 7和R 8相同或不同,且各自独立地为氢原子或C 1-6烷基;
更优选地,各个R 1相同或不同,且各自独立地为C 1-6烷氧基。
最优选地,各个R 1相同或不同,且各自独立地为甲氧基。
在本公开一些实施方案中,所述的通式(I)、通式(II)、通式(Ii)、通式(III)或通式(IV)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选自氢原子、氟原子、氯原子、甲基、乙基、异丙基、甲氧基、乙氧基、三氟甲氧基、一甲基氨基、二甲基氨基和
Figure PCTCN2022111395-appb-000032
优选地,R 1为甲氧基。
在本公开一些实施方案中,所述的通式(I)、通式(II)或通式(Ii)所示的化合物或其可药用的盐,其中各个R 2相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、氨基、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、3至8元环烷基、3至8元杂环基、氧代基、3至8元环烷基氧基和3至8元杂环基氧基;优选地,各个R 2相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基和氧代基;进一步优选地,各个R 2相同或不同,且各自独立地选自氢原子、卤素、C 1-6烷基和C 1-6烷氧基;更优选 地,各个R 2相同或不同,且各自独立地为氢原子或卤素;甚至更优选地,各个R 2相同或不同,且各自独立地为氢原子或氟原子;最优选地,R 2为氢原子。
在本公开一些实施方案中,所述的通式(I)、通式(II)、通式(Ii)、通式(III)或通式(IV)所示的化合物或其可药用的盐,其中各个R 4a相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、-NR 9R 10、3至8元环烷基、3至8元杂环基、3至8元环烷基氧基和3至8元杂环基氧基,R 9和R 10如通式(I)中所定义;优选地,各个R 4a相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6羟烷基和C 1-6烷氧基;更优选地,R 4a为氢原子。
在本公开一些实施方案中,所述的通式(I)、通式(II)、通式(Ii)、通式(III)或通式(IV)所示的化合物或其可药用的盐,其中p为0、1、2或3;优选地,p为1、2或3;更优选地,p为2。
在本公开一些实施方案中,所述的通式(I)、通式(II)、通式(Ii)、通式(III)或通式(IV)所示的化合物或其可药用的盐,其中p为0、1或2;优选地,p为1或2。
在本公开一些实施方案中,所述的通式(I)、通式(II)、通式(Ii)、通式(III)或通式(IV)所示的化合物或其可药用的盐,其中L为化学键或C 1-6亚烷基;优选选自化学键、-CH 2-或-CH 2CH 2-,更优选为化学键和-CH 2-,最优选为化学键。
在本公开一些实施方案中,所述的通式(I)、通式(II)、通式(Ii)、通式(III)或通式(IV)所示的化合物或其可药用的盐,其中n为1或2。
在本公开一些实施方案中,所述的通式(I)、通式(II)、通式(Ii)、通式(III)或通式(IV)所示的化合物或其可药用的盐,其中n为0、1、2或3;优选地,n为0。
在本公开一些实施方案中,所述的通式(III)或通式(IV)所示的化合物或其可药用的盐,其中各个R 4a相同或不同,且各自独立地选自卤素、羟基、C 1-6烷基、C 1-6羟烷基和C 1-6烷氧基,且n为1或2,或者R 4a均为氢原子,且n为3。
在本公开一些实施方案中,所述的通式(III)或通式(IV)所示的化合物或其可药用的盐,其中R 4a均为氢原子,且n为3。
在本公开一些实施方案中,所述的通式(I)、通式(II)、通式(Ii)、通式(III)或通式(IV)所示的化合物或其可药用的盐,其中R 5、R 6相同或不同,且各自独立地选自氢原子、C 1-6烷基、3至8元环烷基、3至12元杂环基、6至10元芳基和5至10元杂芳基;其中所述的C 1-6烷基、3至8元环烷基、3至12元杂环基、6至10元芳基和5至10元杂芳基各自独立地任选被选自羟基、卤素、氰基、氨基、硝基、氧代基、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基和3至8元环烷基中的一个或多个取代基所取代;
优选地,R 5、R 6相同或不同,且各自独立地选自C 1-6烷基、C 1-6卤代烷基和3至8元环烷基。
在本公开一些实施方案中,所述的通式(I)、通式(II)、通式(Ii)、通式(III)或通 式(IV)所示的化合物或其可药用的盐,其中R 7和R 8相同或不同,且各自独立地选自氢原子、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、3至8元环烷基和3至12元杂环基;优选地,R 7和R 8相同或不同,且各自独立地为氢原子或C 1-6烷基;更优选地,R 7和R 8为氢原子。
在本公开一些实施方案中,所述的通式(I)、通式(II)、通式(Ii)、通式(III)或通式(IV)所示的化合物或其可药用的盐,其中R 9和R 10相同或不同,且各自独立地选自氢原子、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、3至8元环烷基和3至12元杂环基;优选地,R 9和R 10为氢原子。
在本公开一些实施方案中,所述的通式(I)、通式(II)和通式(Ii)所示的化合物或其可药用的盐,其中
Figure PCTCN2022111395-appb-000033
Figure PCTCN2022111395-appb-000034
R 1a和R 1b相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、氨基、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、氧代基、3至8元环烷基、C 1-6烷氧基C 1-6烷基、-OR 5、-C(O)R 6、-C(O)OR 6、-C(O)NR 7R 8和-S(O) rR 6,r、R 5、R 6、R 7和R 8如通式(I)中所定义;优选地,R 1a和R 1b相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6烷氧基C 1-6烷基和-C(O)OCH 3;更优选地,R 1a为C 1-6烷氧基,和/或R 1b为C 1-6烷氧基;最优选地,R 1a和R 1b均为甲氧基。
在本公开一些实施方案中,所述的通式(III)或通式(IV)所示的化合物或其可药用的盐,其中
Figure PCTCN2022111395-appb-000035
Figure PCTCN2022111395-appb-000036
R 1a和R 1b相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、氨基、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、氧代基、3至8元环烷基、C 1-6烷氧基C 1-6烷基、-OR 5、-C(O)R 6、-C(O)OR 6、-C(O)NR 7R 8和-S(O) rR 6,r、R 5、R 6、R 7和R 8如通式(I)中所定义;优选地,R 1a和R 1b相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6烷氧基C 1-6烷基和-C(O)OCH 3;更优选地,R 1a为C 1-6烷氧基,和/或R 1b为C 1-6烷氧基;最优选地,R 1a和R 1b均为甲氧基。
在本公开一些实施方案中,所述的通式(I)、通式(II)和通式(Ii)所示的化合物或其可药用的盐,其中
Figure PCTCN2022111395-appb-000037
Figure PCTCN2022111395-appb-000038
R 1a和R 1b相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、氨基、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、氧代基、3至8元环烷基、C 1-6烷氧基C 1-6烷基、-OR 5、-C(O)R 6、-C(O)OR 6、-C(O)NR 7R 8和-S(O) rR 6,r、R 5、R 6、R 7和R 8 如通式(I)中所定义;优选地,R 1a和R 1b相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6烷氧基C 1-6烷基和-C(O)OCH 3;更优选地,R 1a为氢原子或C 1-6烷氧基,和/或R 1b为氢原子或C 1-6烷氧基;最优选地,R 1a和R 1b均为甲氧基;或R 1a为氢原子,R 1b为甲氧基。
在本公开一些实施方案中,所述的通式(III)或通式(IV)所示的化合物或其可药用的盐,其中
Figure PCTCN2022111395-appb-000039
Figure PCTCN2022111395-appb-000040
R 1a和R 1b相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、氨基、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、氧代基、3至8元环烷基、C 1-6烷氧基C 1-6烷基、-OR 5、-C(O)R 6、-C(O)OR 6、-C(O)NR 7R 8和-S(O) rR 6,r、R 5、R 6、R 7和R 8如通式(I)中所定义;优选地,R 1a和R 1b相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6烷氧基C 1-6烷基和-C(O)OCH 3;更优选地,R 1a为氢原子或C 1-6烷氧基,和/或R 1b为氢原子或C 1-6烷氧基;最优选地,R 1a和R 1b均为甲氧基;或R 1a为氢原子,R 1b为甲氧基。
在本公开一些实施方案中,所述的通式(III)所示的化合物或其可药用的盐,其中
Figure PCTCN2022111395-appb-000041
Figure PCTCN2022111395-appb-000042
优选地,
Figure PCTCN2022111395-appb-000043
选自
Figure PCTCN2022111395-appb-000044
更优选地,
Figure PCTCN2022111395-appb-000045
Figure PCTCN2022111395-appb-000046
Figure PCTCN2022111395-appb-000047
在本公开一些实施方案中,所述的通式(IV)所示的化合物或其可药用的盐,其中
Figure PCTCN2022111395-appb-000048
选自
Figure PCTCN2022111395-appb-000049
优选地,
Figure PCTCN2022111395-appb-000050
选自
Figure PCTCN2022111395-appb-000051
Figure PCTCN2022111395-appb-000052
更优选地,
Figure PCTCN2022111395-appb-000053
Figure PCTCN2022111395-appb-000054
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中
Figure PCTCN2022111395-appb-000055
选自
Figure PCTCN2022111395-appb-000056
L为化学键;环B为
Figure PCTCN2022111395-appb-000057
各个R 2相同或不同,且各自独立地为氢原子或氟原子;q为0、1或2;R 3为氢原子;R 4
Figure PCTCN2022111395-appb-000058
环C为5元或6元杂环基、5元或6元杂芳基;m为0或1;n为0;R 0选自氢原子、羟基和Cl;各个R 1相同或不同,且各自独立地选自氢原子、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基和-NR 7R 8;R 7和R 8相同或不同,且各自独立地为氢原子或C 1-6烷基;p为0、1或2;p1为0、1、2或3。
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6烷氧基C 1-6烷基和-C(O)OCH 3;p为1、2或3;各个R 2相同或不同,且各自独立地选自氢原子、卤素、C 1-6烷基和C 1-6烷氧基;q为1;R 3为氢原子;R 4
Figure PCTCN2022111395-appb-000059
各个R 4a相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6羟烷基和C 1-6烷氧基;n为1或2;环C为5至10元杂芳基;环A为6至10元芳基;环B为4至7元杂环基;且L为化学键或-CH 2-。
在本公开一些实施方案中,所述的通式(I)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6烷氧基C 1-6烷基、-C(O)OCH 3和-NR 7R 8;R 7和R 8相同或不同,且各自独立地为氢原子或C 1-6烷基;p为0、1、2、3或4;各个R 2相同或不同,且各自独立地选自氢原子、卤素、C 1-6烷基和C 1-6烷氧基;q为0、1、2、3或4;R 3为氢原子;R 4
Figure PCTCN2022111395-appb-000060
各个R 4a相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6羟烷基和C 1-6烷氧基;n为0、1、2、3或4;m为0或1;环C选自3至8元环烷基、3至8元杂环基、6至10元芳基和5至10元杂芳基;R 0选自氢原子、羟基、卤素和C 1-6烷基;环A为6至10元芳基;环B为4至7元杂环基;且L为化学键或-CH 2-。
在本公开一些实施方案中,所述的通式(II)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6烷氧基C 1-6烷基和 -C(O)OCH 3;p为1、2或3;各个R 2相同或不同,且各自独立地选自氢原子、卤素、C 1-6烷基和C 1-6烷氧基;q为1;各个R 4a相同或不同,且各自独立地选自氢原子、卤素、羟基、C 1-6烷基、C 1-6羟烷基和C 1-6烷氧基;n为1或2;环A为6至10元芳基;环B为4至7元杂环基;环C为5至10元杂芳基;且L为化学键或-CH 2-。
在本公开一些实施方案中,所述的通式(II)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选自氢原子、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基和-NR 7R 8;R 7和R 8相同或不同,且各自独立地为氢原子或C 1-6烷基;p为0、1、2或3;各个R 2相同或不同,且各自独立地为氢原子或卤素;q为0、1或2;各个R 4a相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6羟烷基和C 1-6烷氧基;n为0、1、2或3;环A为6至10元芳基;环B为4至7元杂环基;环C为5元或6元杂芳基;且L为化学键。
在本公开一些实施方案中,所述的通式(Ii)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选自氢原子、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基和-NR 7R 8;R 7和R 8相同或不同,且各自独立地为氢原子或C 1-6烷基;p为0、1、2或3;各个R 2相同或不同,且各自独立地为氢原子或卤素;q为0、1或2;各个R 4a相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6羟烷基和C 1-6烷氧基;n为0、1、2或3;环A为6至10元芳基;环B为4至7元杂环基;环C为5元或6元杂环基、5元或6元杂芳基;且L为化学键。
在本公开一些实施方案中,所述的通式(III)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6烷氧基C 1-6烷基和-C(O)OCH 3;p为1、2或3;X为O或CH 2;R c和R d均为氢原子;各个R 4a相同或不同,且各自独立地选自卤素、羟基、C 1-6烷基、C 1-6羟烷基和C 1-6烷氧基,且n为1或2,或者R 4a均为氢原子,且n为3;s为1或2;且L为化学键或-CH 2-。
在本公开一些实施方案中,所述的通式(III)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选自氢原子、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基和-NR 7R 8;R 7和R 8相同或不同,且各自独立地为氢原子或C 1-6烷基;p为0、1、2或3;X为O或CH 2;各个R c和R d相同或不同,且各自独立地为氢原子或卤素;R 4a均为氢原子;n为3;s为1或2;且L为化学键。
在本公开一些实施方案中,所述的通式(III)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选为C 1-6烷氧基;p为2;X为O或CH 2;各个R c和R d相同或不同,且各自独立地为氢原子或氟原子;R 4a均为氢原子;n为3;s为1或2;且L为化学键。
在本公开一些实施方案中,所述的通式(III)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选为C 1-6烷氧基;p为1或2;X为CH 2;各个R c和R d相同或不同,且各自独立地为氢原子或氟原子;n为0;s为1或2;且L为化学键。
在本公开一些实施方案中,所述的通式(IV)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6烷氧基C 1-6烷基和-C(O)OCH 3;p为1、2或3;X为O或CH 2;各个R 4a相同或不同,且各自独立地选自卤素、羟基、C 1-6烷基、C 1-6羟烷基和C 1-6烷氧基,且n为1或2,或者R 4a均为氢原子,且n为3;s为1或2;且L为化学键或-CH 2-。
在本公开一些实施方案中,所述的通式(IV)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选自氢原子、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基和-NR 7R 8;R 7和R 8相同或不同,且各自独立地为氢原子或C 1-6烷基;p为0、1、2或3;X为O或CH 2;R 4a均为氢原子;n为3;s为1或2;且L为化学键。
在本公开一些实施方案中,所述的通式(IV)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地为C 1-6烷氧基;p为1或2;X为CH 2;R 4a均为氢原子;n为3;s为1或2;且L为化学键。
在本公开一些实施方案中,所述的通式(IV)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地为C 1-6烷氧基;p为2;X为O或CH 2;R 4a均为氢原子;n为3;s为1或2;且L为化学键。
在本公开一些实施方案中,所述的通式(IV)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地为C 1-6烷氧基;p为1或2;X为CH 2;n为0;s为1或2;且L为化学键。
表A本公开的典型化合物包括但不限于:
Figure PCTCN2022111395-appb-000061
Figure PCTCN2022111395-appb-000062
Figure PCTCN2022111395-appb-000063
Figure PCTCN2022111395-appb-000064
Figure PCTCN2022111395-appb-000065
Figure PCTCN2022111395-appb-000066
Figure PCTCN2022111395-appb-000067
Figure PCTCN2022111395-appb-000068
本公开的另一方面涉及通式(IA)所示的化合物或其盐:
Figure PCTCN2022111395-appb-000069
其中:
环B、R 2、R 3、R 4和q如通式(I)中所定义。
本公开的另一方面涉及通式(IIA)所示的化合物或其盐:
Figure PCTCN2022111395-appb-000070
其中:
环B、环C、R 2、R 4a、q和n如通式(II)中所定义。
本公开的另一方面涉及通式(IiA)所示的化合物或其盐:
Figure PCTCN2022111395-appb-000071
其中:
环B、环C、R 2、R 4a、q和n如通式(Ii)中所定义。
本公开的另一方面涉及通式(IIIA)所示的化合物或其盐:
Figure PCTCN2022111395-appb-000072
其中:
X、R 4a、R c、R d、n和s如通式(III)中所定义。
本公开的另一方面涉及通式(IVA)所示的化合物或其盐:
Figure PCTCN2022111395-appb-000073
其中:
X、R 4a、n和s如通式(IV)中所定义。
本公开的另一方面涉及通式(IA’)所示的化合物或其盐:
Figure PCTCN2022111395-appb-000074
其中:
X 1为卤素,优选为溴;
环A、环B、R 1、R 2、R 3、L、p和q如通式(I)中所定义。
本公开的另一方面涉及通式(IIA’)所示的化合物或其盐:
Figure PCTCN2022111395-appb-000075
其中:
X 1为卤素,优选为溴;
环A、环B、R 1、R 2、L、p和q如通式(II)中所定义。
本公开的另一方面涉及通式(IIIA’)所示的化合物或其盐:
Figure PCTCN2022111395-appb-000076
其中:
X 1为卤素,优选为溴;
R 1、R c、R d、s、p、X和L如通式(III)中所定义。
本公开的另一方面涉及通式(IVA’)所示的化合物或其盐:
Figure PCTCN2022111395-appb-000077
其中:
X 1为卤素,优选为溴;
R 1、s、p、X和L如通式(IV)中所定义。
表B本公开的典型中间体化合物包括但不限于:
Figure PCTCN2022111395-appb-000078
Figure PCTCN2022111395-appb-000079
本公开的另一方面涉及一种制备通式(I)所示的化合物或其可药用的盐的方法,该方法包括以下步骤:
Figure PCTCN2022111395-appb-000080
通式(IA)所示的化合物或其盐与通式(IB)所示的化合物或其盐反应,得到通式(I)所示的化合物或其可药用的盐,
其中:
环A、环B、L、R 1至R 4、p和q如通式(I)中所定义。
本公开的另一方面涉及一种制备通式(II)所示的化合物或其可药用的盐的方法,该方法包括以下步骤:
Figure PCTCN2022111395-appb-000081
通式(IIA)所示的化合物或其盐与通式(IB)所示的化合物或其盐反应,得到通式(II)所示的化合物或其可药用的盐,
其中:
环A、环B、环C、L、R 1、R 2、R 4a、p、q和n如通式(II)中所定义。
本公开的另一方面涉及一种制备通式(Ii)所示的化合物或其可药用的盐的方法,该方法包括以下步骤:
Figure PCTCN2022111395-appb-000082
通式(IiA)所示的化合物或其盐与通式(IB)所示的化合物或其盐反应,得到通式(Ii)所示的化合物或其可药用的盐,
其中:
环A、环B、环C、L、R 1、R 2、R 4a、p、q和n如通式(Ii)中所定义。
本公开的另一方面涉及一种制备通式(III)所示的化合物或其可药用的盐的方法,该方法包括以下步骤:
Figure PCTCN2022111395-appb-000083
通式(IIIA)所示的化合物或其盐与通式(IIIB)所示的化合物或其盐反应,得到通式(III)所示的化合物或其可药用的盐,
其中:
L、X、R 1、R 4a、R c、R d、p、n和s如通式(III)中所定义。
本公开的另一方面涉及一种制备通式(IV)所示的化合物或其可药用的盐的方法,该方法包括以下步骤:
Figure PCTCN2022111395-appb-000084
通式(IVA)所示的化合物或其盐与通式(IIIB)所示的化合物或其盐反应,得到通式(IV)所示的化合物或其可药用的盐,
其中:
X、L、R 1、R 4a、p、n和s如通式(IV)中所定义。
本公开的另一方面涉及一种制备通式(Ii)所示的化合物或其可药用的盐的方法,该方法包括以下步骤:
Figure PCTCN2022111395-appb-000085
通式(IIA’)所示的化合物或其盐与通式(IiB’)所示的化合物或其盐发生偶联反应,得到通式(Ii)所示的化合物或其可药用的盐,
其中:
环D为至少含有一个环内双键的3至8元杂环基,优选为至少含有一个环内双键的5元或6元杂环基,更优选为
Figure PCTCN2022111395-appb-000086
环C为3至8元杂环基,优选为5元或6元杂环基,更优选为
Figure PCTCN2022111395-appb-000087
X 1为卤素,优选为溴;
环A、环B、L、R 1、R 2、R 4a、p、q和n如通式(Ii)中所定义。
本公开的另一方面涉及一种药物组合物,所述药物组合物含有本公开通式(I)、通式(II)、通式(Ii)、通式(III)、通式(IV)或表A所示的化合物或其可药用的盐,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
本公开进一步涉及通式(I)、通式(II)、通式(Ii)、通式(III)、通式(IV)或表A所示 的化合物或其可药用的盐或包括其的药物组合物在制备用于抑制KAT的药物中的用途;其中所述KAT优选为KAT6,更优选为KAT6A和/或KAT6B。
本公开进一步涉及通式(I)、通式(II)、通式(Ii)、通式(III)、通式(IV)或表A所示的化合物或其可药用的盐或包括其的药物组合物在制备用于治疗和/或预防KAT介导的疾病的药物中的用途;其中所述KAT优选为KAT6,更优选为KAT6A和/或KAT6B。
本公开进一步涉及通式(I)、通式(II)、通式(Ii)、通式(III)、通式(IV)或表A所示的化合物或其可药用的盐或包括其的药物组合物在制备用于治疗和/或预防癌症的药物中的用途;其中所述的癌症优选选自肺癌(如NCSLC,SCLC)、间皮瘤、骨癌、胰腺癌、皮肤癌、头颈癌、脑癌、黑色素瘤、肛门癌、肝癌、乳腺癌、输卵管癌、子宫内膜癌、宫颈癌、卵巢癌、阴道癌、外阴癌、霍奇金淋巴瘤、食道癌、结直肠癌、小肠癌、胃癌、甲状腺癌、甲状旁腺癌、肾上腺癌、软组织肉瘤、阴茎癌、睾丸癌、前列腺癌、白血病、B细胞淋巴瘤、膀胱癌、尿道癌、输尿管癌、肾细胞癌、肾盂癌、中枢神经系统肿瘤(CNS)、原发性CNS淋巴瘤、脊髓肿瘤、胶质细胞瘤、脑胶质瘤、垂体腺瘤和鳞状细胞癌;优选为乳腺癌、前列腺癌、肺癌(如NCSLC,SCLC)、胰腺癌、卵巢癌、宫颈癌、子宫内膜癌、膀胱癌、脑胶质瘤、B细胞淋巴瘤、肝癌和白血病;其中所述乳腺癌优选为ER +乳腺癌或ER +/HER2 -乳腺癌;其中所述肺癌(如NCSLC,SCLC)优选为非小细胞肺癌;其中所述前列腺癌优选为去势抵抗性前列腺癌。
本公开进一步涉及一种抑制KAT的方法,其包括给予所需患者抑制有效量的通式(I)、通式(II)、通式(Ii)、通式(III)、通式(IV)或表A所示的化合物或其可药用的盐、或包括其的药物组合物,其中所述KAT优选为KAT6,更优选为KAT6A和/或KAT6B,。
本公开还涉及一种治疗和/或预防KAT介导的疾病的方法,其包括给予所需患者治疗有效量和/或预防有效量的通式(I)、通式(II)、通式(Ii)、通式(III)、通式(IV)或表A所示的化合物或其可药用的盐、或包含其的药物组合物,其中所述KAT优选为KAT6,更优选为KAT6A和/或KAT6B,。
本公开进一步涉及一种治疗和/或预防癌症的方法,其包括给予所需患者治疗有效量和/或预防有效量的通式(I)、通式(II)、通式(Ii)、通式(III)、通式(IV)或表A所示的化合物或其可药用的盐、或包括其的药物组合物;其中所述的癌症优选选自肺癌(如NCSLC,SCLC)、间皮瘤、骨癌、胰腺癌、皮肤癌、头颈癌、脑癌、黑色素瘤、肛门癌、肝癌、乳腺癌、输卵管癌、子宫内膜癌、宫颈癌、卵巢癌、阴道癌、外阴癌、霍奇金淋巴瘤、食道癌、结直肠癌、小肠癌、胃癌、甲状腺癌、甲状旁腺癌、肾上腺癌、软组织肉瘤、阴茎癌、睾丸癌、前列腺癌、白血病、B细胞淋巴瘤、膀胱癌、尿道癌、输尿管癌、肾细胞癌、肾盂癌、中枢神经系统肿瘤(CNS)、原发性CNS淋巴瘤、脊髓肿瘤、胶质细胞瘤、脑胶质瘤、垂体腺瘤和 鳞状细胞癌;优选为乳腺癌、前列腺癌、肺癌(如NCSLC,SCLC)、胰腺癌、卵巢癌、宫颈癌、子宫内膜癌、膀胱癌、脑胶质瘤、B细胞淋巴瘤、肝癌和白血病;其中所述乳腺癌优选为ER +乳腺癌或ER +/HER2 -乳腺癌;其中所述肺癌(如NCSLC,SCLC)优选为非小细胞肺癌;其中所述前列腺癌优选为去势抵抗性前列腺癌。
本公开进一步涉及一种通式(I)、通式(II)、通式(Ii)、通式(III)、通式(IV)或表A所示的化合物或其可药用的盐、或包括其的药物组合物,其用作药物。
本公开进一步涉及一种通式(I)、通式(II)、通式(Ii)、通式(III)、通式(IV)或表A所示的化合物或其可药用的盐、或包括其的药物组合物,其用作抑制KAT的药物,其中所述KAT优选为KAT6,更优选为KAT6A和/或KAT6B。
本公开进一步涉及一种通式(I)、通式(II)、通式(Ii)、通式(III)、通式(IV)或表A所示的化合物或其可药用的盐、或包括其的药物组合物,其用于抑制KAT,其中所述KAT优选为KAT6,更优选为KAT6A和/或KAT6B。
本公开进一步涉及一种通式(I)、通式(II)、通式(Ii)、通式(III)、通式(IV)或表A所示的化合物或其可药用的盐、或包括其的药物组合物,其用作KAT抑制剂,其中所述KAT优选为KAT6,更优选为KAT6A和/或KAT6B。
本公开还涉及通式(I)、通式(II)、通式(Ii)、通式(III)、通式(IV)或表A所示的化合物或或其可药用的盐,或包含其的药物组合物,其用于治疗和/或预防KAT介导的疾病,其中所述KAT优选为KAT6,更优选为KAT6A和/或KAT6B。
本公开进一步涉及一种通式(I)、通式(II)、通式(Ii)、通式(III)、通式(IV)或表A所示的化合物或其可药用的盐、或包括其的药物组合物,用于治疗和/或预防癌症;其中所述的癌症优选选自肺癌(如NCSLC,SCLC)、间皮瘤、骨癌、胰腺癌、皮肤癌、头颈癌、脑癌、黑色素瘤、肛门癌、肝癌、乳腺癌、输卵管癌、子宫内膜癌、宫颈癌、卵巢癌、阴道癌、外阴癌、霍奇金淋巴瘤、食道癌、结直肠癌、小肠癌、胃癌、甲状腺癌、甲状旁腺癌、肾上腺癌、软组织肉瘤、阴茎癌、睾丸癌、前列腺癌、白血病、B细胞淋巴瘤、膀胱癌、尿道癌、输尿管癌、肾细胞癌、肾盂癌、中枢神经系统肿瘤(CNS)、原发性CNS淋巴瘤、脊髓肿瘤、胶质细胞瘤、脑胶质瘤、垂体腺瘤和鳞状细胞癌;优选为乳腺癌、前列腺癌、肺癌(如NCSLC,SCLC)、胰腺癌、卵巢癌、宫颈癌、子宫内膜癌、膀胱癌、脑胶质瘤、B细胞淋巴瘤、肝癌和白血病;其中所述乳腺癌优选为ER +乳腺癌或ER +/HER2 -乳腺癌;其中所述肺癌(如NCSLC,SCLC)优选为非小细胞肺癌;其中所述前列腺癌优选为去势抵抗性前列腺癌。
在本公开一些实施方案中,所述KAT6为KAT6A和/或KAT6B。
在本公开一些实施方案中,所述癌症为乳腺癌。
在本公开一些实施方案中,所述乳腺癌为ER +乳腺癌。
在本公开一些实施方案中,所述乳腺癌为ER +/HER2 -乳腺癌。
在本公开一些实施方案中,所述乳腺癌为局部晚期或转移性ER +/HER2 -乳腺癌。
在本公开一些实施方案中,所述肺癌(如NCSLC,SCLC)为非小细胞肺癌。
在本公开一些实施方案中,所述肺癌(如NCSLC,SCLC)为局部晚期或转移性非小细胞肺癌。
在本公开一些实施方案中,所述前列腺癌为去势抵抗性前列腺癌。
在本公开一些实施方案中,所述前列腺癌为局部晚期或转移去势抵抗性前列腺癌。
为了方便起见,本文可以使用某些众所周知的缩写,包括:雌激素受体阳性(ER +),人表皮生长因子受体2阴性(HER2 -),非小细胞肺癌(NSCLC)和去势抵抗性前列腺癌(CRPC)。
可将活性化合物制成适合于通过任何适当途径给药的形式,通过常规方法使用一种或多种药学上可接受的载体来配制本公开的组合物。因此,本公开的活性化合物可以配制成用于口服给药、注射(例如静脉内、肌肉内或皮下)给药,吸入或吹入给药的各种剂型。本公开的化合物也可以配制成例如片剂、硬或软胶囊、水性或油性混悬液、乳剂、注射液、可分散性粉末或颗粒、栓剂、锭剂或糖浆等剂型。
作为一般性指导,活性化合物优选是以单位剂量的方式,或者是以患者可以以单剂自我给药的方式。本公开化合物或组合物的单位剂量的表达方式可以是片剂、胶囊、扁囊剂、瓶装药水、药粉、颗粒剂、锭剂、栓剂、再生药粉或液体制剂。合适的单位剂量可以是0.1~1000mg。
本公开的药物组合物除活性化合物外,可含有一种或多种辅料,所述辅料选自以下成分:填充剂(稀释剂)、粘合剂、润湿剂、崩解剂或赋形剂等。根据给药方法的不同,组合物可含有0.1至99重量%的活性化合物。
片剂含有活性成分和用于混合的适宜制备片剂的无毒的可药用的赋形剂。这些赋形剂可以是惰性赋形剂、造粒剂、崩解剂、粘合剂和润滑剂。这些片剂可以不包衣或可通过掩盖药物的味道或在胃肠道中延迟崩解和吸收,因而在较长时间内提供缓释作用的已知技术将其包衣。
也可用其中活性成分与惰性固体稀释剂或其中活性成分与水溶性载体或油溶媒混合的软明胶胶囊提供口服制剂。
水混悬液含有活性物质和用于混合的适宜制备水悬浮液的赋形剂。此类赋形剂是悬浮剂、分散剂或湿润剂。水混悬液也可以含有一种或多种防腐剂、一种或多种着色剂、一种或多种矫味剂和一种或多种甜味剂。
油混悬液可通过使活性成分悬浮于植物油,或矿物油配制而成。油悬浮液可含有增稠剂。可加入上述的甜味剂和矫味剂,以提供可口的制剂。可通过加入抗氧化剂保存这些组合物。
本公开的药物组合物也可以是水包油乳剂的形式。油相可以是植物油,或矿物油或其混合物。适宜的乳化剂可以是天然产生的磷脂,乳剂也可以含有甜味剂、矫味剂、防腐剂和抗氧剂。此类制剂也可含有缓和剂、防腐剂、着色剂和抗氧剂。
本公开的药物组合物可以是无菌注射水溶液形式。可以使用的可接受的溶媒或溶剂有水、林格氏液和等渗氯化钠溶液。无菌注射制剂可以是其中活性成分溶于油相的无菌注射水包油微乳可通过局部大量注射,将注射液或微乳注入患者的血流中。或者,最好按可保持本公开化合物恒定循环浓度的方式给予溶液和微乳。为保持这种恒定浓度,可使用连续静脉内递药装置。这种装置的实例是Deltec CADD-PLUS.TM.5400型静脉注射泵。
本公开的药物组合物可以是用于肌内和皮下给药的无菌注射水或油混悬液的形式。可按已知技术,用上述那些适宜的分散剂或湿润剂和悬浮剂配制该混悬液。无菌注射制剂也可以是在肠胃外可接受的无毒稀释剂或溶剂中制备的无菌注射溶液或混悬液。此外,可方便地用无菌固定油作为溶剂或悬浮介质。为此目的,可使用任何调和固定油。此外,脂肪酸也可以制备注射剂。
可按用于直肠给药的栓剂形式给予本公开化合物。可通过将药物与在普通温度下为固体但在直肠中为液体,因而在直肠中会溶化而释放药物的适宜的无刺激性赋形剂混合来制备这些药物组合物。
可通过加入水来制备水混悬的可分散粉末和颗粒给予本公开化合物。可通过将活性成分与分散剂或湿润剂、悬浮剂或一种或多种防腐剂混合来制备这些药物组合物。
如本领域技术人员所熟知的,药物的给药剂量依赖于多种因素,包括但并非限定于以下因素:所用具体化合物的活性、患者的年龄、患者的体重、患者的健康状况、患者的行为、患者的饮食、给药时间、给药方式、排泄的速率、药物的组合、疾病的严重性等;另外,最佳的治疗方式如治疗的模式、化合物的日用量或可药用的盐的种类可以根据传统的治疗方案来验证。
术语说明
除非有相反陈述,在说明书和权利要求书中使用的术语具有下述含义。
术语“烷基”指饱和脂肪族烃基团,其为包含1至20个(例如1、2、3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)碳原子的直链或带有支链基团(即C 1-20烷基),优选含有1至12个(例如1、2、3、4、5、6、7、8、9、10、11和12个)碳原子的烷基(即C 1-20烷基),更优选为含有1至6个碳原子的烷基(即C 1-6烷基)。非限制性实例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3- 二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基、正壬基、2-甲基-2-乙基己基、2-甲基-3-乙基己基、2,2-二乙基戊基、正癸基、3,3-二乙基己基、2,2-二乙基己基,及其各种支链异构体等。烷基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点上被取代,取代基优选选自D原子、卤素、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“亚烷基”指饱和的直链或支链的脂肪族烃基,其为从母体烷的相同碳原子或两个不同的碳原子上除去两个氢原子所衍生的残基,其具有1至20个(例如1、2、3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)碳原子(即C 1-20亚烷基),优选1至12个碳原子(即C 1-12亚烷基),更优选1至6个碳原子(即C 1-6亚烷基)。非限制性的实例包括:亚甲基(-CH 2-)、1,1-亚乙基(-CH(CH 3)-)、1,2-亚乙基(-CH 2CH 2)-、1,1-亚丙基(-CH(CH 2CH 3)-)、1,2-亚丙基(-CH 2CH(CH 3)-)、1,3-亚丙基(-CH 2CH 2CH 2-)、1,4-亚丁基(-CH 2CH 2CH 2CH 2-)等。亚烷基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点被取代,取代基优选选自D原子、卤素、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“杂亚烷基”指亚烷基中的一个或多个-CH 2-被选自N、O、S、S(O)和S(O) 2中的一个或多个所替代;其中所述的烷基如上所定义;杂亚烷基可以是取代的或非取代的,当被取代时,取代基可以在任何可使用的连接点被取代,取代基优选选自D原子、卤素、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“烯基”指分子中含有至少一个碳碳双键的烷基化合物,其中烷基的定义如上所述,其具有2至12个(例如2、3、4、5、6、7、8、9、10、11或12个)碳原子(即C 2-12烯基)。所述烯基优选具有2至6个碳原子的烯基(即C 2-6烯基)。烯基可以是取代的或非取代的,当被取代时,取代基优选选自烷氧基、卤素、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“炔基”指分子中含有至少一个碳碳三键的烷基化合物,其中烷基的定义如上所述,其具有2至12个(例如2、3、4、5、6、7、8、9、10、11或12个)碳 原子(即C 2-12炔基)。所述炔基优选具有2至6个碳原子的炔基(即C 2-6炔基)。炔基可以是取代的或非取代的,当被取代时,取代基优选选自烷氧基、卤素、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“环烷基”指饱和或部分不饱和单环或多环环状烃取代基,环烷基环包含3至20个(例如3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个)碳原子(即3至20元环烷基),优选包含3至12个(例如3、4、5、6、7、8、9、10、11或12个)碳原子(即3至12元环烷基),优选包含3至8个(例如3、4、5、6、7和8个)碳原子(即3至8元环烷基),进一步优选包含4至7个(例如4、5、6和7个)碳原子(即4至7元环烷基),更优选包含3至6个(例如3、4、5和6个)碳原子(即3至6元环烷基)。单环环烷基的非限制性实例包括环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等;多环环烷基包括螺环烷基、稠环烷基和桥环烷基。
术语“螺环烷基”指5至20元(例如5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个环原子,即5至20元螺环烷基),单环之间共用一个碳原子(称螺原子)的多环基团,其可以含有一个或多个双键。优选为6至14元(即6至14元螺环烷基),更优选为7至10元(例如7、8、9或10元,即7至10元螺环烷基)。根据环与环之间共用螺原子的数目将螺环烷基分为单螺环烷基、双螺环烷基或多螺环烷基,优选为单螺环烷基和双螺环烷基。更优选为3元/5元、3元/6元、4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺环烷基。螺环烷基的非限制性实例包括:
Figure PCTCN2022111395-appb-000088
术语“稠环烷基”指5至20元(例如5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个环原子,即5至20元稠环烷基),系统中的每个环与体系中的其他环共享毗邻的一对碳原子的全碳多环基团,其中一个或多个环可以含有一个或多个双键。优选为6至14元(即6至14元稠环烷基),更优选为7至10元(例如7、8、9或10元,即7至10元稠环烷基)。根据组成环的数目可以分为双环、三环、四环或多环稠环烷基,优选为双环或三环,更优选为3元/4元、3元/5元、3元/6元、4元/4元、4元/5元、4元/6元、5元/4元、5元/5元、5元/6元、6元/3元、6元/4元、6元/5元和6元/6元的双环稠环烷基。稠环烷基的非限制性实例包括:
Figure PCTCN2022111395-appb-000089
术语“桥环烷基”指5至20元(例如5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个碳原子,即5至20元桥环烷基),任意两个环共用两个不直接连接的碳原子的全碳多环基团,其可以含有一个或多个双键。优选为6至14元(即6至14元桥环烷基),更优选为7至10元(例如7、8、9或10元,即7至10元桥环烷基)。根据组成环的数目可以分为双环、三环、四环或多环桥环烷基,优选为双环、三环或四环,更优选为双环或三环。桥环烷基的非限制性实例包括:
Figure PCTCN2022111395-appb-000090
所述环烷基环包括如上所述的环烷基(包括单环环烷基、螺环烷基、稠环烷基和桥环烷基)稠合于芳基、杂芳基或杂环烷基环上,其中与母体结构连接在一起的环为环烷基,非限制性实例包括
Figure PCTCN2022111395-appb-000091
等;优选为
Figure PCTCN2022111395-appb-000092
环烷基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点上被取代,取代基优选选自卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“烷氧基”指-O-(烷基),其中烷基的定义如上所述。烷氧基的非限制性实例包括:甲氧基、乙氧基、丙氧基和丁氧基。烷氧基可以是任选取代的或非取代的,当被取代时,其优选为一个或多个以下基团,其独立地选自D原子、卤素、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基。
术语“杂环基”指饱和或部分不饱和单环或多环环状取代基,其包含3至20个环原子(例如3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个环原子,即3至20元杂环基),其中一个或多个环原子为选自氮、氧和硫的杂原子,所述的硫可任选被氧代(即形成亚砜或砜),但不包括-O-O-、-O-S-或 -S-S-的环部分,其余环原子为碳。优选包含3至12个(例如3、4、5、6、7、8、9、10、11和12个)环原子(即3至12元杂环基),其中1~4个(例如1、2、3和4个)是杂原子;更优选包含3至8个环原子(例如3、4、5、6、7和8个,即3至8元杂环基),其中1-3个(例如1、2和3个)是杂原子;进一步优选包含4至7个环原子(例如4、5、6和7个,即4至7元杂环基),其中1-3个(例如1、2和3个)是杂原子;更优选包含3至6个环原子(例如3、4、5和6个,即3至6元杂环基),其中1-3个(例如1、2和3个)是杂原子;最优选包含5元或6个环原子(即5元或6元杂环基),其中1-2个(例如1、2个)是杂原子。单环杂环基的非限制性实例包括吡咯烷基、四氢吡喃基、1,2,3,6-四氢吡啶基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基等。多环杂环基包括螺杂环基、稠杂环基和桥杂环基。
术语“螺杂环基”指5至20元(例如5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个环原子,即5至20元螺杂环基),单环之间共用一个原子(称螺原子)的多环杂环基团,其中一个或多个环原子为选自氮、氧和硫的杂原子,所述的硫可任选被氧代(即形成亚砜或砜),其余环原子为碳。其可以含有一个或多个双键。优选为6至14元(即6至14元螺杂环基),更优选为7至10元(例如7、8、9或10元,即7至10元螺杂环基)。根据环与环之间共用螺原子的数目将螺杂环基分为单螺杂环基、双螺杂环基或多螺杂环基,优选为单螺杂环基和双螺杂环基。更优选为3元/5元、3元/6元、4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺杂环基。螺杂环基的非限制性实例包括:
Figure PCTCN2022111395-appb-000093
术语“稠杂环基”指5至20元(例如5、6、7、8、9、10、11、12、13、14、15、16、17、18、19或20个环原子,即5至20元稠杂环基),系统中的每个环与体系中的其他环共享毗邻的一对原子的多环杂环基团,一个或多个环可以含有一个或多个双键,其中一个或多个环原子为选自氮、氧和硫的杂原子,所述的硫可任选被氧代(即形成亚砜或砜),其余环原子为碳。优选为6至14元(即6至14元稠杂环基),更优选为7至10元(例如7、8、9或10元,即7至10元稠杂环基)。根据组成环的数目可以分为双环、三环、四环或多环稠杂环基,优选为双环或三环,更优选为3元/4元、3元/5元、3元/6元、4元/4元、4元/5元、4元/6元、5元/4元、5元/5元、5元/6元、6元/3元、6元/4元、6元/5元和6元/6元双环稠杂环基。稠杂环基的非限制性实例包括:
Figure PCTCN2022111395-appb-000094
术语“桥杂环基”指5至14元(例如5、6、7、8、9、10、11、12、13或14个环原子,即5至14元桥杂环基),任意两个环共用两个不直接连接的原子的多环杂环基团,其可以含有一个或多个双键,其中一个或多个环原子为选自氮、氧和硫的杂原子,所述的硫可任选被氧代(即形成亚砜或砜),其余环原子为碳。优选为6至14元(即6至14元桥杂环基),更优选为7至10元(例如7、8、9或10元,即7至10元桥杂环基)。根据组成环的数目可以分为双环、三环、四环或多环桥杂环基,优选为双环、三环或四环,更优选为双环或三环。桥杂环基的非限制性实例包括:
Figure PCTCN2022111395-appb-000095
所述杂环基环包括如上所述的杂环基(包括单环杂环基、螺杂环基、稠杂环基和桥杂环基)稠合于芳基、杂芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基,其非限制性实例包括:
Figure PCTCN2022111395-appb-000096
等。
杂环基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点上被取代,取代基优选选自卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“芳基”指具有共轭的π电子体系的6至14元(例如6、7、8、9、10、11、12、13或14个环原子,即6至14元芳基)全碳单环或稠合多环(稠合多环是共享毗邻碳原子对的环)基团,优选为6至10元(即6至10元芳基),例如苯基和萘基。所述芳基环包括如上所述的芳基环稠合于杂芳基、杂环基或环烷基环上, 其中与母体结构连接在一起的环为芳基环,其非限制性实例包括:
Figure PCTCN2022111395-appb-000097
芳基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接点上被取代,取代基优选选自卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
术语“杂芳基”指包含1至4个(例如1、2、3和4个)杂原子、5至14个环原子(例如5、6、7、8、9、10、11、12、13或14个环原子,即5至14元杂芳基)的杂芳族体系,其中杂原子选自氧、硫和氮。杂芳基优选为5至10元(例如5、6、7、8、9或10元,即5至10元杂芳基),更优选为5元或6元(即5元或6元杂芳基),例如呋喃基、噻吩基、吡啶基、吡咯基、N-烷基吡咯基、嘧啶基、吡嗪基、哒嗪基、咪唑基、吡唑基、三唑基、四唑基等。所述杂芳基环包括如上述的杂芳基稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,其非限制性实例包括:
Figure PCTCN2022111395-appb-000098
杂芳基可以是取代的或非取代的,当被取代时,其可以在任何可使用的连接 点上被取代,取代基优选选自卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、环烷基氧基、杂环基氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基中的一个或多个。
上述环烷基、杂环基、芳基和杂芳基包括从母体环原子上除去一个氢原子所衍生的残基,或从母体的相同或两个不同的环原子上除去两个氢原子所衍生的残基,即“二价环烷基”、“二价杂环基”(例如
Figure PCTCN2022111395-appb-000099
Figure PCTCN2022111395-appb-000100
等)、“亚芳基”和“亚杂芳基”。
术语“环烷基烷基”指烷基被一个或多个环烷基取代,其中环烷基和烷基如上所定义。
术语“杂环基烷基”指烷基被一个或多个杂环基取代,其中杂环基和烷基如上所定义。
术语“杂芳基烷基”指烷基被一个或多个杂芳基取代,其中杂芳基和烷基如上所定义。
术语“环烷基氧基”指环烷基-O-,其中环烷基如上所定义。
术语“杂环基氧基”指杂环基-O-,其中杂环基如上所定义。
术语“烷硫基”指烷基-S-,其中烷基如上所定义。
术语“卤代烷基”指烷基被一个或多个卤素取代,其中烷基如上所定义。
术语“卤代烷氧基”指烷氧基被一个或多个卤素取代,其中烷氧基如上所定义。
术语“烷氧基烷基”指烷基被一个或多个烷氧基取代,其中烷基和烷氧基如上所定义。
术语“羟烷基”指烷基被一个或多个羟基取代,其中烷基如上所定义。
术语“卤素”指氟、氯、溴或碘。
术语“羟基”指-OH。
术语“巯基”指-SH。
术语“氨基”指-NH 2
术语“氰基”指-CN。
术语“硝基”指-NO 2
术语“氧代基”或“氧代”指“=O”。
术语“羰基”指C=O。
术语“醛基”指-C(O)H
术语“羧基”指-C(O)OH。
术语“羧酸酯基”指-C(O)O(烷基)、-C(O)O(环烷基)(烷基)C(O)O-或(环烷基)C(O)O-,其中烷基和环烷基如上所定义。
另一方面,本公开化合物可以存在特定的几何或立体异构体形式。本公开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公开的范围之内。烷基等取代基中可存在另外的不对称碳原子。所有这些异构体以及它们的混合物,均包括在本公开的范围之内。可以通过的手性合成或手性试剂或者其他常规技术制备光学活性的(R)-和(S)-异构体以及D和L异构体。如果想得到本公开某化合物的一种对映体,可以通过不对称合成或者具有手性助剂的衍生作用来制备,其中将所得非对映体混合物分离,并且辅助基团裂开以提供纯的所需对映异构体。或者,当分子中含有碱性官能团(如氨基)或酸性官能团(如羧基)时,与适当的光学活性的酸或碱形成非对映异构体的盐,然后通过本领域所公知的常规方法进行非对映异构体拆分,然后回收得到纯的对映体。此外,对映异构体和非对映异构体的分离通常是通过使用色谱法完成的,所述色谱法采用手性固定相,并任选地与化学衍生法相结合(例如由胺生成氨基甲酸盐)。
本公开所述化合物的化学结构中,键
Figure PCTCN2022111395-appb-000101
表示未指定构型,即如果化学结构中存在手性异构体,键
Figure PCTCN2022111395-appb-000102
可以为
Figure PCTCN2022111395-appb-000103
或者同时包含
Figure PCTCN2022111395-appb-000104
Figure PCTCN2022111395-appb-000105
两种构型。本公开所述化合物的化学结构中,键
Figure PCTCN2022111395-appb-000106
并未指定构型,即可以为Z构型或E构型,或者同时包含两种构型。
另外,本公开的化合物和中间体还可以以不同的互变异构体形式存在,并且所有这样的形式包含于本公开的范围内。术语“互变异构体”或“互变异构体形式”是指可经由低能垒互变的不同能量的结构异构体。例如,质子互变异构体(也称为质子转移互变异构体)包括经由质子迁移的互变,如酮-烯醇及亚胺-烯胺异构化。内酰胺-内酰亚胺平衡实例是在如下所示的A和B之间。
Figure PCTCN2022111395-appb-000107
本公开中的所有化合物可以被画成A型或B型。所有的互变异构形式在本公开的范围内。化合物的命名不排除任何互变异构体。
本公开还包括一些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公开化合物。可结合到本公开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。
本公开的化合物可以在一个或多个构成该化合物的原子上包含非天然比例的原子同位素。例如,可用放射性同位素标记化合物,比如氚( 3H),可用重氢取代氢 形成氘代药物,氘与碳构成的键比普通氢与碳构成的键更坚固,相比于未氘化药物,氘代药物有降低毒副作用、增加药物稳定性、增强疗效、延长药物生物半衰期等优势。本公开的化合物的所有同位素组成的变换,无论放射性与否,都包括在本公开的范围之内。
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。
除另有说明,当一个位置被特别地指定为氘(D)时,该位置应理解为具有大于氘的天然丰度(其为0.015%)至少1000倍的丰度的氘(即,至少10%的氘掺入)。示例中化合物的具有大于氘的天然丰度可以是至少1000倍的丰度的氘、至少2000倍的丰度的氘、至少3000倍的丰度的氘、至少4000倍的丰度的氘、至少5000倍的丰度的氘、至少6000倍的丰度的氘或更高丰度的氘。本公开还包括各种氘化形式的式(I)化合物。与碳原子连接的各个可用的氢原子可独立地被氘原子替换。本领域技术人员能够参考相关文献合成氘化形式的式(I)化合物。在制备氘代形式的式(I)化合物时可使用市售的氘代起始物质,或它们可使用常规技术采用氘代试剂合成,氘代试剂包括但不限于氘代硼烷、三氘代硼烷四氢呋喃溶液、氘代氢化锂铝、氘代碘乙烷和氘代碘甲烷等。
“任选”或“任选地”意味着随后所描述的事件或环境可以但不必然发生,该说明包括该事件或环境发生或不发生的情形。例如,“任选被烷基取代的杂环基团”意味着烷基可以但不必须存在,该说明包括杂环基团被烷基取代的情形和杂环基团不被烷基取代的情形。
“取代的”指基团中的一个或多个氢原子,优选为1~5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代。本领域技术人员能够在不付出过多努力的情况下(通过实验或理论)确定可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。
“药物组合物”表示含有一种或多种本文所述化合物或其生理学上/可药用的盐或前体药物与其他化学组分的混合物,以及其他组分例如生理学/可药用的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
“可药用的盐”是指本公开化合物的盐,这类盐用于哺乳动物体内时具有安全性和有效性,且具有应有的生物活性。可以在化合物的最终分离和纯化过程中,或通过使合适的基团与合适的碱或酸反应来单独制备盐。通常用于形成药学上可接受的盐的碱包括无机碱,例如氢氧化钠和氢氧化钾,以及有机碱,例如氨。通常用于形成药学上可接受的盐的酸包括无机酸以及有机酸。
针对药物或药理学活性剂而言,术语“治疗有效量”、“抑制有效量”或“预防 有效量”是指足以达到或部分达到预期效果的药物或药剂的用量。有效量的确定因人而异,取决于受体的年龄和一般情况,也取决于具体的活性物质,个案中合适的有效量可以由本领域技术人员根据常规试验确定。
本文所用的术语“药学上可接受的”是指这些化合物、材料、组合物和/或剂型,在合理的医学判断范围内,适用于与患者组织接触而没有过度毒性、刺激性、过敏反应或其他问题或并发症,具有合理的获益/风险比,并且对预期的用途是有效。
本文所使用的,单数形式的“一个”、“一种”和“该”包括复数引用,反之亦然,除非上下文另外明确指出。
当将术语“约”应用于诸如pH、浓度、温度等的参数时,表明该参数可以变化±10%,并且有时更优选地在±5%之内。如本领域技术人员将理解的,当参数不是关键的时,通常仅出于说明目的给出数字,而不是限制。
本公开化合物的合成方法
为了完成本公开的目的,本公开采用如下技术方案:
方案一
本公开通式(I)所示的化合物,或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2022111395-appb-000108
通式(IA)所示的化合物或其盐与通式(IB)所示的化合物或其盐,在碱的作用下,任选在微波中发生亲核取代反应,得到通式(I)所示的化合物或其可药用的盐,
其中:
L、环A、环B、R 1至R 4、p和q如通式(I)中所定义。
方案二
本公开通式(II)所示的化合物,或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2022111395-appb-000109
通式(IIA)所示的化合物或其盐与通式(IB)所示的化合物或其盐,在碱的作用下,任选在微波中发生亲核取代反应,得到通式(II)所示的化合物或其可药用的盐,
其中:
环A、环B、环C、L、R 1、R 2、R 4a、p、q和n如通式(II)中所定义。
方案二-1
本公开通式(Ii)所示的化合物,或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2022111395-appb-000110
通式(IiA)所示的化合物或其盐与通式(IB)所示的化合物或其盐,在碱的作用下,任选在微波中发生亲核取代反应,得到通式(Ii)所示的化合物或其可药用的盐,
其中:
环A、环B、环C、L、R 1、R 2、R 4a、p、q和n如通式(Ii)中所定义。
方案三
本公开通式(III)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2022111395-appb-000111
通式(IIIA)所示的化合物或其盐与通式(IIIB)所示的化合物或其盐,在碱的作用下,任选在微波中发生亲核取代反应,得到通式(III)所示的化合物或其可药用的盐,
其中:
L、X、R 1、R 4a、R c、R d、p、n和s如通式(III)中所定义。
方案四
本公开通式(IV)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2022111395-appb-000112
通式(IVA)所示的化合物或其盐与通式(IIIB)所示的化合物或其盐,在碱的作用下,任选在微波中发生亲核取代反应,得到通式(IV)所示的化合物或其可药用的盐,
其中:
X、L、R 1、R 4a、p、n和s如通式(IV)中所定义。
方案五
本公开通式(II)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2022111395-appb-000113
第一步:通式(IIA’)所示的化合物或其盐与通式(IIB’)所示的化合物或其盐,在碱的作用下(例如正丁基锂),发生亲核加成反应,得到通式(IIa)所示的化合物或其可药用的盐;
第二步:通式(IIa)所示的化合物或其盐发生氯代反应(例如在PCl 3或者SOCl 2的作用下),得到通式(IIb)所示的化合物或其可药用的盐;
第三步:通式(IIb)所示的化合物或其可药用的盐在金属作用下(例如锌粉或铁粉)发生还原反应,得到通式(II)所示的化合物或其可药用的盐;
其中:
X 1为卤素,优选为溴;
环A、环B、环C、L、R 1、R 2、R 4a、p、q和n如通式(II)中所定义。
方案六
本公开通式(Ii)所示的化合物或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2022111395-appb-000114
通式(IIA’)所示的化合物或其盐与通式(IiB’)所示的化合物或其盐,在碱和金属催化剂的作用下,任选加入配体,发生偶联反应,得到通式(Ii)所示的化合物或其可药用的盐,
其中:
环D为至少含有一个环内双键的3至8元杂环基,优选为至少含有一个环内双键的5元或6元杂环基,更优选为
Figure PCTCN2022111395-appb-000115
环C为3至8元杂环基,优选为5元或6元杂环基,更优选为
Figure PCTCN2022111395-appb-000116
X 1为卤素,优选为溴;
环A、环B、L、R 1、R 2、R 4a、p、q和n如通式(Ii)中所定义。
上述方案一至方案六的反应中,所述的碱包括有机碱和无机碱类,所述的有机 碱类包括但不限于三乙胺、吡啶、3,5-二甲基吡啶、N,N-二异丙基乙胺、正丁基锂、二异丙基氨基锂、双(三甲基硅基)胺基锂、醋酸钠、醋酸钾、叔丁醇钠、叔丁醇钾或1,8-二氮杂二环十一碳-7-烯;所述的无机碱类包括但不限于氢化钠、磷酸钾、碳酸钠、碳酸钾、碳酸铯、氢氧化钠、氢氧化锂和氢氧化钾;方案一至方案四中所述的碱,优选选自吡啶、双(三甲基硅基)胺基锂和3,5-二甲基吡啶;方案五中所述的碱,优选为正丁基锂;方案六中所述的碱,优选为碳酸钾。
上述方案六中,所述的金属催化剂包括但不限于醋酸钯、四(三苯基膦)钯、三(二亚苄基丙酮)二钯、1,1'-二(二苯膦基)二茂铁二氯化钯(II)、双(乙腈)氯化钯(II)和钯/碳,优选为醋酸钯。
上述方案六中,所述的配体包括但不限于三苯基膦、三(邻甲苯基)膦和1,1'-联萘-2,2'-双二苯膦(BINAP),优选为三苯基膦。
上述方案一至方案四中的反应,任选在催化剂的作用下进行,所述催化剂包括但不限于4-二甲氨基吡啶和二甲亚砜(DMSO)。
当在微波中进行上述方案一至方案六的反应时,反应温度为100~150℃,优选为120℃。
当在微波中进行上述方案一至方案六的反应时,反应时间为0.5~6小时,优选2~3小时,更优选3小时。
上述方案一至方案六的反应优选在溶剂中进行,所用的溶剂包括但不限于:乙二醇二甲醚、醋酸、甲醇、乙醇、乙腈、正丁醇、甲苯、四氢呋喃、二氯甲烷、石油醚、乙酸乙酯、正己烷、二甲基亚砜、1,4-二氧六环、水、N,N-二甲基甲酰胺、N,N-二甲基乙酰胺、1,2-二溴乙烷、吡啶及其混合物。
具体实施方式
以下结合实施例用于进一步描述本公开,但这些实施例并非限制着本公开的范围。
实施例
化合物的结构是通过核磁共振(NMR)或/和质谱(MS)来确定的。NMR位移(δ)以10 -6(ppm)的单位给出。NMR的测定是用Bruker AVANCE NEO 500M核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d 6)、氘代氯仿(CDCl 3)、氘代甲醇(CD 3OD),内标为四甲基硅烷(TMS)。
MS的测定用Agilent 1200/1290 DAD-6110/6120 Quadrupole MS液质联用仪(生产商:Agilent,MS型号:6110/6120 Quadrupole MS)、waters ACQuity UPLC-QD/SQD(生产商:waters,MS型号:waters ACQuity Qda Detector/waters SQ Detector)、THERMO Ultimate 3000-Q Exactive(生产商:THERMO,MS型号:THERMO Q Exactive)。
高效液相色谱法(HPLC)分析使用Agilent HPLC 1200DAD、Agilent HPLC  1200VWD和Waters HPLC e2695-2489高效液相色谱仪。
手性HPLC分析测定使用Agilent 1260 DAD高效液相色谱仪。
高效液相制备色谱法使用Waters 2767、Waters 2767-SQ Detecor2、Shimadzu LC-20AP和Gilson-281制备型色谱仪。
手性制备色谱法使用Shimadzu LC-20AP制备型色谱仪。
CombiFlash快速制备仪使用Combiflash Rf200(TELEDYNE ISCO)。
薄层层析硅胶板使用烟台黄海HSGF254或青岛GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm~0.2mm,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm。
硅胶柱色谱法一般使用烟台黄海硅胶200~300目硅胶为载体。
激酶平均抑制率及IC 50值的测定用NovoStar酶标仪(德国BMG公司)。
本公开的已知的起始原料可以采用或按照本领域已知的方法来合成,或可购买自ABCR GmbH&Co.KG,Acros Organics,Aldrich Chemical Company,韶远科技(上海)有限公司、达瑞化学品、上海泰坦科技、阿拉丁、安耐吉化学、中国医药集团有限公司、阿达玛斯试剂有限公司、西格玛奥德里奇(上海)贸易有限公司、上海毕得医药科技有限公司、上海皓鸿生物医药科技有限公司、赛默飞世尔科技(中国)科技有限公司等公司。
实施例中无特殊说明,反应均能够在氩气氛或氮气氛下进行。
氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
氢气氛是指反应瓶连接一个约1L容积的氢气气球。
加压氢化反应使用Parr 3916EKX型氢化仪和清蓝QL-500型氢气发生器或HC2-SS型氢化仪。
氢化反应通常抽真空,充入氢气,反复操作3次。
微波反应使用CEM Discover-S 908860型微波反应器。
实施例中无特殊说明,溶液是指水溶液。
实施例中无特殊说明,反应的温度为室温,为20℃~30℃。
实施例中的反应进程的监测采用薄层色谱法(TLC),反应所使用的展开剂,纯化化合物采用的柱层析的洗脱剂的体系和薄层色谱法的展开剂体系包括:A:正己烷/乙酸乙酯体系,B:二氯甲烷/甲醇体系,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的三乙胺和醋酸等碱性或酸性试剂进行调节。
实施例1
N-(4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2,6-二甲氧基苯磺酰胺1
Figure PCTCN2022111395-appb-000117
第一步
4-溴-2-((2,4-二甲氧基苄基)氧基)-6-氟苯甲腈1b
将4-溴-2,6-二氟苯甲腈1a(22g,101mmol)和2,4-二甲氧基苯甲醇(18.5g,110mmol)溶于N,N-二甲基甲酰胺(200mL)中,加入碳酸铯(49g,150mmol)。反应液在60℃下搅拌16小时。反应液冷却至室温,减压抽滤,滤液加入乙酸乙酯(500mL)稀释,用饱和氯化钠溶液洗涤(30mL×5),所得有机相经无水硫酸钠干燥,过滤,滤液减压浓缩,得到标题产物1b(36.9g,产率:100%)。该产品不经纯化,直接用于下一步反应。
第二步
4-溴-2-氟-6-羟基苯甲腈1c
将化合物1b(36.9g,100.7mmol)溶于二氯甲烷(250mL)中,冷却至0℃,滴加三氟乙酸(39g,342mmol),反应液升至室温搅拌1小时。反应液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物1c(9.7g,产率: 44.5%)。
第三步
2-(烯丙氧基)-4-溴-6-氟苯甲腈1d
将化合物1c(10.7g,49.5mmol)溶于N,N-二甲基甲酰胺(120mL)中,反应液冷却至0℃,加入碳酸铯(24g,73.7mmol)和烯丙基溴(11.2g,92.6mmol),反应液升至室温搅拌4小时。反应液减压抽滤,滤液加入乙酸乙酯(400mL)稀释,用饱和氯化钠溶液洗涤(30mL×3),所得有机相经无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物1d(11.7g,产率:92%)。
1H NMR(500MHz,CDCl 3)δ7.02(dt,1H),6.95(t,1H),6.04(m,1H),5.57-5.47(m,1H),5.41(dt,1H),4.75-4.64(m,2H)。
第四步
3-烯丙基-4-溴-6-氟-2-羟基苯甲腈1e
将化合物1d(3.35g,13.1mmol)溶于1,2-二氯苯(80mL)中,置换氮气三次,反应液在180℃下搅拌13小时。反应液冷却至室温,用硅胶柱色谱法(湿法上样)以洗脱剂体系A纯化所得残余物,得到标题产物1e(2.77g,产率:82.7%)。
1H NMR(500MHz,CDCl 3)δ7.07(dd,1H),6.45(s,1H),5.90(dddd,1H),5.24-5.10(m,2H),3.68-3.55(m,2H)。
第五步
3-烯丙基-4-溴-6-氟-2-(甲氧基甲氧基)苯甲腈1f
将化合物1e(1g,3.91mmol)溶于乙腈(15mL)中,加入碳酸钾(1.07g,7.74mmol)和溴(溴甲氧基)甲烷(MOMBr,634mg,5.08mmol),搅拌反应2小时。反应液加水(10mL)淬灭,用乙酸乙酯萃取(50mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物1f(1.11g,产率:94.7%)。
1H NMR(500MHz,CDCl 3)δ7.28(d,1H),5.89(ddt,1H),5.26(d,2H),5.08-4.98(m,2H),3.66(s,3H),3.58(s,2H)。
第六步
4-溴-6-氟-3-(2-羟乙基)-2-(甲氧基甲氧基)苯甲腈1g
将化合物1f(1.1g,3.66mmol)溶于50mL甲醇和四氢呋喃(V:V=1:1)混合溶剂中,将反应液冷却至-78℃,通入干燥的臭氧,通气1小时。反应液加入三苯基膦(1.05g,4.0mmol)淬灭,缓慢升至室温,搅拌反应0.5小时。再将反应液冷却至0℃,分批加入硼氢化钠(560mg,14.8mmol),搅拌反应1小时。反应液加水(5mL)淬灭,减压浓缩,加入乙酸乙酯(150mL)稀释,用饱和氯化钠溶液洗涤(10mL×2)。所得有机相经无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物1g(800mg,产率:71.8%)。
1H NMR(500MHz,CDCl 3)δ7.28(d,1H),5.32(s,2H),3.82(t,2H),3.67(s,3H),3.10 (t,2H),2.39(s,1H)。
第七步
4-溴-6-氟-2-羟基-3-(2-羟乙基)苯甲腈1h
将化合物1g(800mg,2.63mmol)溶于甲醇(25mL)中,将反应液冷却至0℃,加入盐酸二氧六环溶液(4M,24mmol,6mL),反应液升至室温搅拌2小时。反应液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物1h(680mg,产率:99%)。
1H NMR(500MHz,CDCl 3)δ7.07-6.95(m,1H),4.01(t,2H),3.14(t,2H)。
第八步
4-溴-6-氟-2,3-二氢苯并呋喃-7-甲腈1i
将化合物1h(680mg,2.61mmol)溶于四氢呋喃(100mL)中,将反应液冷却至0℃,加入三苯基膦(2.05g,7.81mmol)和偶氮二甲酸二异丙酯(1.58g,7.81mmol),反应液升至室温搅拌2小时。反应液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物1i(570mg,产率:90%)。
1H NMR(500MHz,CDCl 3)δ6.84(dd,1H),4.84(td,2H),3.24(tt,2H)。
第九步
7-氰基-6-氟-2,3-二氢苯并呋喃-4-羧酸甲酯1j
将化合物1i(615mg,2.54mmol)溶于20mL甲醇和N,N-二甲基甲酰胺的混合溶剂(V:V=1:3)中,依次加入1,1'-双二苯基膦二茂铁二氯化钯(185mg,252mmmol)和三乙胺(771mg,7.62mmol)。一氧化碳置换3次,一氧化碳保护下,反应液在80℃下搅拌12小时。反应液冷却至室温,减压浓缩,加入乙酸乙酯(150mL)稀释,用饱和氯化钠溶液洗涤(20mL×3),所得有机相经无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物1j(289mg,产率:51.4%)。
1H NMR(500MHz,CDCl 3)δ7.24(d,1H),4.84(td,2H),3.94(s,3H),3.57(td,2H)。
第十步
6-氟-4-(羟甲基)-2,3-二氢苯并呋喃-7-甲腈1k
将化合物1j(436mg,1.97mmol)溶于干燥四氢呋喃(10mL)中,置换氮气3次,将反应液冷却至0℃,加入硼氢化锂(2M,5mmol,2.5mL)。反应液升温至70℃搅拌2小时。反应液冷却至室温,加水(1mL)淬灭,加入乙酸乙酯(100mL)稀释,用饱和氯化钠溶液洗涤(20mL×2),所得有机相经无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物1k(350mg,产率:91.9%)。
1H NMR(500MHz,CDCl 3)δ6.76(dd,1H),4.81(td,2H),4.66(s,2H),3.20(t,2H)。
第十一步
(7-氰基-6-氟-2,3-二氢苯并呋喃-4-基)甲磺酸甲酯1l
将化合物1k(350mg,1.81mmol)溶于二氯甲烷(20mL)中,冷却至0℃,加入三乙胺(2.2g,21.74mmol)和甲烷磺酰氯(1.24g,10.82mmol)。反应液升至室温搅拌2小时。反应液加饱和碳酸氢钠溶液(10mL)淬灭,加入乙酸乙酯(100mL)稀释,用饱和氯化钠溶液洗涤(20mL×2),所得有机相经无水硫酸钠干燥,过滤,滤液减压浓缩,得到标题产物1l(491mg,产率:99%)。该产品不经纯化,直接用于下一步反应。
第十二步
4-((1H-吡唑-1-基)甲基)-6-氟-2,3-二氢苯并呋喃-7-甲腈1m
将化合物1l(491mg,1.81mmol)溶于N,N-二甲基甲酰胺(15mL)中,加入碳酸钾(1.25g,9.04mmol)和吡唑(369mg,5.42mmol)。反应液在60℃下搅拌12小时。反应液过滤,所得滤液加入乙酸乙酯(100mL)稀释,用饱和氯化钠溶液洗涤(20mL×3),所得有机相经无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物1m(361mg,产率:82%)。
MS m/z(ESI):244.0[M+1]。
第十三步
4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-胺1n
将化合物1m(361mg,1.48mmol)和乙酰氧肟酸(334mg,4.45mmol)溶于N,N-二甲基甲酰胺(15mL)中,加入碳酸钾(1.0g,7.23mmol)。反应液在60℃下搅拌12小时。反应液过滤,加入乙酸乙酯(100mL)稀释,用饱和氯化钠溶液洗涤(20mL×3),所得有机相经无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物1n(189mg,产率:49.7%)。
MS m/z(ESI):257.0[M+1]。
第十四步
N-(4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2,6-二甲氧基苯磺酰胺1
将化合物1n(189mg,737μmol)和2,6-二甲氧基苯磺酰氯1o(265mg,1.12mmol,采用专利申请“WO2020254946A1中说明书第70页的方案15”公开的方法制备而得)溶于吡啶(8mL)中,置换氮气3次。反应液在120℃下微波反应2小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Welch Xtimate C18 5μm 30×150mm;流动相:A-水相(0.1%甲酸):B-乙腈=30%-45%(15min),流速:30mL/min),得到标题产物1(44mg,产率:13%)。
MS m/z(ESI):457.0[M+1]。
1H NMR(500MHz,CD 3OD)δ7.71(s,1H),7.55(s,1H),7.47(td,1H),6.74(dd,2H),6.58(s,1H),6.36(q,1H),5.42(s,2H),4.80(td,2H),3.83(s,6H),3.10(t,2H)。
实施例2
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-2,6-二甲氧基苯磺酰胺2
Figure PCTCN2022111395-appb-000118
第一步
4-溴-6-氟-2-羟基-3-(3-羟丙基)苯甲腈2a
将化合物1e(4.8g,18.7mmol)溶于无水四氢呋喃(100mL)中,0℃下,滴加硼烷四氢呋喃溶液(1.0M,22mL,22mmol)。反应液在冰浴下搅拌2小时。冰浴下,先后加入3M氢氧化钠水溶液(13mL,39mmol)和30%的双氧水(3.0mL),加完搅拌10min。用2M盐酸调节反应液pH=2,用乙酸乙酯(100mL×2)萃取,收集有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物2a(3.5g,产率:68.1%)。
MS m/z(ESI):275.8[M+1]。
1H NMR(500MHz,CDCl 3)δ7.04(d,1H),3.71(t,2H),3.00-2.98(m,2H),2.01-1.96(m,2H)。
第二步
5-溴-7-氟色烷-8-甲腈2b
将化合物2a(3.8g,13.9mmol)溶于无水四氢呋喃(80mL)中,将反应液冷却至0℃,加入三苯基膦(4.4g,16.8mmol)和偶氮二甲酸二异丙酯(3.4g,16.8mmol),反应液升至室温搅拌2小时。反应液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物2b(3.0g,产率:84.5%)。
MS m/z(ESI):257.8[M+1]。
1H NMR(500MHz,CDCl 3)δ7.03(d,1H),4.33(t,2H),2.77-2.74(m,2H),2.12-2.08(m,2H)。
第三步
8-氰基-7-氟色烷-5-羧酸甲酯2c
将化合物2b(2.6g,10.2mmol)溶于40mL甲醇和N,N-二甲基甲酰胺(V:V=1:3)的混合溶剂中,依次加入1,1'-双二苯基膦二茂铁二氯化钯(800mg,1.09mmol)和三乙胺(3.0g,2.93mmol)。一氧化碳置换3次,在10bar、90℃条件下搅拌16小时。反应液冷却至室温,减压浓缩,加入乙酸乙酯(150mL)稀释,用饱和氯化钠溶液洗涤(50mL×3),所得有机相经无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物2c(2.1g,产率:87.9%)。MS m/z(ESI):235.9[M+1]。
1H NMR(500MHz,CDCl 3)δ7.26(d,1H),4.38-4.36(m,2H),3.93(s,3H),3.10-3.07(m,2H),2.08-2.03(m,2H)。
第四步
7-氟-5-(羟甲基)色烷-8-甲腈2d
将化合物2c(2.1g,8.93mmol)溶于干燥四氢呋喃(40mL)中,置换氮气3次,将反应液冷却至0℃,加入硼氢化锂(2M,18mmol,9.0mL)。反应液升温至70℃搅拌2小时。反应液冷却至室温,加水(1mL)淬灭,加入乙酸乙酯(100mL)稀释,用饱和氯化钠溶液洗涤(50mL×2),所得有机相经无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物2d(1.84g,产率:99.5%)。
MS m/z(ESI):207.9[M+1]。
第五步
5-((1H-吡唑-1-基)甲基)-7-氟色烷-8-甲腈2f
将化合物2d(1.8g,8.69mmol)和1-(甲基磺酰基)-1H-吡唑2e(1.5g,10.3mmol,采用专利申请“WO2020254946A1中说明书第63页方案8中间体13”公开的方法制备而得)溶于乙腈(30mL)中,加入碳酸铯(4.2g,12.9mmol),70℃反应1小时。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物2f(1.9g,产率:85.0%)。
MS m/z(ESI):258.0[M+1]。
第六步
5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-胺2g
将化合物2f(1.9g,7.39mmol)和乙酰氧肟酸(1.7g,22.2mmol,Adamas)溶于N,N-二甲基甲酰胺(30mL)和水(4.0mL)中,加入碳酸钾(6.2g,44.9mmol)。反应液在70℃下搅拌24小时。反应液冷却到室温,加入水(100mL),过滤,收集滤饼,干燥,得到标题产物2g(1.65g,产率:82.7%)。
MS m/z(ESI):271.0[M+1]。
1H NMR(500MHz,DMSO-d 6)δ7.78(d,1H),7.52(d,1H),6.34(s,1H),6.32(t,1H),5.85(s,2H),5.39(s,2H),4.25-4.23(m,2H),2.68(t,2H),2.03-1.98(m,2H)。
第七步
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-2,6-二甲氧基苯磺酰胺2
将化合物2g(200mg,0.740mmol)和化合物1o(300mg,1.27mmol)溶于吡啶(5.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Xtimate phenyl-hexyl Prep C18 5μm,30×150mm;流动相:A-水相(0.1%氨水):B-乙腈=5%-45%(20min),流速:30mL/min),得到标题产物2(40mg,产率:11.5%)。
MS m/z(ESI):470.8[M+1]。
1H NMR(500MHz,DMSO-d 6)δ9.40(s,1H),7.79(d,1H),7.52(d,1H),7.48(t,1H),6.77(d,2H),6.43(s,1H),6.32(t,1H),5.42(s,2H),4.25(t,2H),3.78(s,6H),2.70(t,2H),2.04-1.99(m,2H)。
实施例3
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-2-甲氧基苯磺酰胺3
Figure PCTCN2022111395-appb-000119
将化合物2g(300mg,1.11mmol)和2-甲氧基苯磺酰氯3a(460mg,2.22mmol)溶于吡啶(6.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 5μm 30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=20%-45%(15min),流速:30mL/min),得到标题产物3(100mg,产率:20.5%)。
MS m/z(ESI):440.8[M+1]。
1H NMR(500MHz,DMSO-d 6)δ9.96(s,1H),7.81-7.78(m,2H),7.62(d,1H),7.51(d,1H),7.20(d,1H),7.09(d,1H),6.45(s,1H),6.32(t,1H),5.41(s,2H),4.19(t,2H),3.81(s,3H),2.69(t,2H),2.01-1.96(m,2H)。
实施例4
N-(5-((1H-吡唑-1-基)甲基)-2,3-二氢-[1,4]二噁英并[2',3':5,6]苯并[1,2-d]异噁唑-9-基)-2-甲氧基苯磺酰胺4
Figure PCTCN2022111395-appb-000120
第一步
4-溴-5-氟-2,3-二羟基苯甲酸甲酯4b
将化合物5-氟-2,3-二氢苯甲酸甲酯4a(2.26g,12.1mmol,采用文献“J.Med.Chem.2010,53,7035–7047”公开的方法制备而得)溶于二氯甲烷(60mL)中,将反应液冷却至0℃,分批加入N-溴代丁二酰亚胺(2.6g,14.6mmol),反应液在室温下搅拌3天。反应液用饱和亚硫酸氢钠溶液洗涤(30mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物4b(1.8g,产率:55.9%)。
1H NMR(500MHz,CDCl 3)δ10.84(s,1H),7.14(d,1H),3.97(s,3H)。
第二步
8-溴-7-氟-2,3-二氢苯并[b][1,4]二噁英-5-甲酸甲酯4c
将化合物4b(1.0g,3.77mmol)与1,2-二溴乙烷(1.1g,5.85mmol)溶于N,N-二甲基甲酰胺(10mL)中,加入碳酸铯(2.46g,7.55mmol),反应液于80℃搅拌2小时。反应液过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物4c(873mg,产率:79.5%)。
MS m/z(ESI):292.9[M+1]。
1H NMR(500MHz,CDCl 3)δ7.26(d,1H),4.45-4.43(m,2H),4.39-4.36(m,2H),3.91 (s,3H)。
第三步
8-氰基-7-氟-2,3-二氢苯并[b][1,4]二噁英-5-甲酸甲酯4d
将化合物4c(708mg,2.43mmol)、氰化锌(714mg,6.08mmol)与甲磺酸(2-二环己基膦基-2',6'-二异丙氧基-1,1'-联苯基)(2-氨基-1,1'-联苯-2-基)钯(306mg,0.365mmol,毕得)溶于N,N-二甲基甲酰胺(15mL)中,置换氮气3次。反应液在110℃下反应16小时。反应液冷却至室温,反应液过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物4d(503mg,产率:87.2%)。
MS m/z(ESI):238.0[M+1]。
1H NMR(500MHz,CDCl 3)δ7.20(d,1H),4.51-4.48(m,2H),4.42-4.40(m,2H),3.94(s,3H)。
第四步
6-氟-8-(羟甲基)-2,3-二氢苯并[b][1,4]二噁英-5-甲腈4e
将化合物4d(500mg,2.1mmol)溶于干燥四氢呋喃(15mL)中,置换氮气3次,将反应液冷却至0℃,加入硼氢化锂(2M,4mmol,2.0mL)。反应液升温至70℃搅拌2小时。反应液冷却至室温,加水(1mL)淬灭,加入乙酸乙酯(30mL)稀释,用饱和氯化钠溶液洗涤(30mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物4e(323mg,产率:73.2%)。
1H NMR(500MHz,CDCl 3)δ6.87(dd,1H),4.72(s,2H),4.46-4.42(m,2H),4.36-4.32(m,2H)。
第五步
(8-氰基-7-氟-2,3-二氢苯并[b][1,4]二噁英-5-基)甲磺酸甲酯4f
将化合物4e(323mg,1.54mmol)溶于二氯甲烷(10mL)中,冷却至0℃,加入三乙胺(780mg,7.71mmol)和甲烷磺酰氯(355mg,3.1mmol)。反应液升至室温搅拌2小时。反应液加饱和碳酸氢钠溶液(5mL)淬灭,加入乙酸乙酯(30mL)稀释,用饱和氯化钠溶液洗涤(20mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,得到标题产物4f(200mg,产率:45.1%)。该产品不经纯化,直接用于下一步反应。
第六步
8-((1H-吡唑-1-基)甲基)-6-氟-2,3-二氢苯并[b][1,4]二噁英-5-甲腈4g
将化合物4f(200mg,0.69mmol)溶于N,N-二甲基甲酰胺(7mL)中,加入碳酸钾(240mg,1.73mmol)和吡唑(120mg,1.76mmol)。反应液在60℃下搅拌12小时。反应液过滤,所得滤液加入乙酸乙酯(30mL)稀释,用饱和氯化钠溶液洗涤(20mL×3),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物4g(155mg,产率:85.8%)。
MS m/z(ESI):260.0[M+1]。
1H NMR(500MHz,CDCl 3)δ7.63(d,1H),7.48(dd,1H),6.34(t,1H),6.22(d,1H),5.34(s,2H),4.47-4.42(m,2H),4.38-4.35(m,2H)。
第七步
5-((1H-吡唑-1-基)甲基)-2,3-二氢-[1,4]二噁英并[2',3':5,6]苯并[1,2-d]异噁唑-9-胺4h
将化合物4g(155mg,0.6mmol)和乙酰氧肟酸(135mg,1.8mmol)溶于4mL N,N-二甲基甲酰胺与水的混合溶剂(V:V=7:1)中,加入碳酸钾(495mg,3.58mmol)。反应液在70℃下搅拌24小时。反应液冷却到室温,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物4h(110mg,产率:67.5%)。
MS m/z(ESI):273.0[M+1]。
1H NMR(500MHz,CDCl 3)δ7.61-7.55(m,1H),7.49(d,1H),6.51(s,1H),6.32(t,1H),5.39(s,2H),4.60(s,2H),4.42(dd,2H),4.37(dd,2H)。
第八步
N-(5-((1H-吡唑-1-基)甲基)-2,3-二氢-[1,4]二噁英并[2',3':5,6]苯并[1,2-d]异噁唑-9-基)-2-甲氧基苯磺酰胺4
将化合物4h(40mg,0.147mmol)和化合物3a(152mg,0.735mmol)溶于吡啶(3.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 5μm 30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=15%-35%(20min),流速:30mL/min),得到标题产物4(25mg,产率:38.4%)。
MS m/z(ESI):443.0[M+1]。
1H NMR(500MHz,DMSO-d 6)δ10.33(s,1H),7.80(d,1H),7.77(dd,1H),7.58(s,1H),7.50(d,1H),7.18(s,1H),7.05(s,1H),6.41(s,1H),6.30(t,1H),5.37(s,2H),4.33(q,4H),3.80(s,3H)。
实施例5
N-(4-((1H-吡唑-1-基)甲基)-2,2-二氟-[1,3]二氧杂戊环并[4',5':5,6]苯并[1,2-d]异噁唑-8-基)-2,6-二甲氧基苯磺酰胺5
Figure PCTCN2022111395-appb-000121
Figure PCTCN2022111395-appb-000122
第一步
7-溴-6-氟-2-硫亚基苯并[d][1,3]二氧杂环戊烯-4-甲酸甲酯5a
将化合物4b(16.7g,63mmol)溶于四氢呋喃(200mL)中,将反应液冷却至0℃,分批加入N,N'-硫羰基二咪唑(18g,101mmol),反应液在室温下搅拌3小时。反应液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物5a(9.1g,产率:47.0%)。
1H NMR(500MHz,CDCl 3)δ7.71(d,1H),4.05(s,3H)。
第二步
7-溴-2,2,6-三氟苯并[d][1,3]二氧杂环戊烯-4-甲酸甲酯5b
-40℃下将化合物5a(9.1g,29.6mmol)溶于二氯甲烷(160mL)中,于氮气保护下加入氟化氢吡啶溶液(42.3g,427mmol),反应液在-40℃下反应5分钟。随后分批加入N-碘代丁二酰亚胺(20g,88.9mmol),反应液继续在-40℃下反应30分钟。反应液加饱和亚硫酸氢钠溶液(20mL)淬灭,用饱和氯化钠溶液洗涤(60mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物5b(4.8g,产率:51.7%)。
1H NMR(500MHz,CDCl 3)δ7.49(d,1H),4.00(s,3H)。
第三步
(7-溴-2,2,6-三氟苯并[d][1,3]二氧杂环戊烯-4-基)甲醇5c
将化合物5b(4.8g,15.3mmol)溶于干燥四氢呋喃(150mL)中,置换氮气3次,将反应液冷却至0℃,加入硼氢化锂(2M,27.6mmol,13.8mL)。反应液升温至70℃搅拌2小时。反应液冷却至室温,加水(5mL)淬灭,加入乙酸乙酯(60mL)稀释,用饱和氯化钠溶液洗涤(30mL×2),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物5c(3.2g,产率:73.2%)。
1H NMR(500MHz,CDCl 3)δ7.04(d,1H),4.76(d,2H)。
第四步
1-((7-溴-2,2,6-三氟苯并[d][1,3]二氧杂环戊烯-4-基)甲基)-1H-吡唑5d
将化合物5c(3.2g,11.2mmol)和化合物2e(1.8g,12.3mmol)溶于乙腈(55mL) 中,加入碳酸铯(5.5g,16.9mmol),70℃反应1小时。反应液过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物5d(2.61g,产率:69.3%)。
MS m/z(ESI):336.9[M+1]。
1H NMR(500MHz,CDCl 3)δ7.59(d,1H),7.51(d,1H),6.67(d,1H),6.35(t,1H),5.33(s,2H)。
第五步
7-((1H-吡唑-1-基)甲基)-2,2,5-三氟苯并[d][1,3]二氧杂环戊烯-4-甲腈5e
将化合物5d(1.6g,4.77mmol)和氰化亚铜(3.2g,11.2mmol)溶于N-甲基吡咯烷酮(50mL)中,反应液在200℃反应1小时。反应液冷却至室温,加入二氯甲烷(60mL)稀释,用饱和氯化钠溶液洗涤(60mL×2),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物5e(465mg,产率:34.6%)。
MS m/z(ESI):282.0[M+1]。
第六步
4-((1H-吡唑-1-基)甲基)-2,2-二氟-[1,3]二氧杂戊环并[4',5':5,6]苯并[1,2-d]异噁唑-8-胺5f
将化合物5e(1g,3.55mmol)和乙酰氧肟酸(800mg,10.66mmol)溶于20mL N,N-二甲基甲酰胺与水的混合溶剂(V:V=7:1)中,加入碳酸钾(2.95g,21.33mmol)。反应液在70℃下搅拌30分钟。反应液冷却到室温,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物5f(60mg,产率:5.7%)。MS m/z(ESI):295.0[M+1]。
第七步
N-(4-((1H-吡唑-1-基)甲基)-2,2-二氟-[1,3]二氧杂戊环并[4',5':5,6]苯并[1,2-d]异噁唑-8-基)-2,6-二甲氧基苯磺酰胺5
将化合物5f(60mg,0.20mmol)与化合物1o(144mg,0.61mmol)溶于乙腈(5mL)中,加入二甲亚砜(1mg,0.01mmol)与3,5-二甲基吡啶(87mg,0.81mmol),反应液在35℃下反应2天。反应液过滤,滤液减压浓缩,所得残余物经高效液相色谱法纯化(welch Prep C18 5μm 30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=25%-45%(20min),流速:30mL/min),得到标题产物5(55mg,产率:54.5%)。
MS m/z(ESI):495.0[M+1]。
1H NMR(500MHz,CDCl 3)δ7.65-7.41(m,2H),7.29(m,2H),6.90(s,1H),6.65(t,2H),6.35(s,1H),5.48(s,2H),3.99-3.89(s,6H)。
实施例6
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-6-甲氧基-2,3-二 氢-1H-茚-5-磺酰胺6
Figure PCTCN2022111395-appb-000123
将化合物2g(50mg,0.185mmol)和6-甲氧基-2,3-二氢-1H-茚-5-磺酰氯6a(200mg,0.811mmol,采用专利申请“WO2019243491A1中说明书第114页的中间体I108”公开的方法制备而得)溶于吡啶(2.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 5μm 30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=20%-45%(15min),流速:30mL/min),得到标题产物6(30mg,产率:33.7%)。
MS m/z(ESI):480.8[M+1]。
1H NMR(500MHz,DMSO-d 6)δ9.59(s,1H),7.78(d,1H),7.63(s,1H),7.51(d,1H),7.08(s,1H),6.44(s,1H),6.31(d,1H),5.41(s,2H),4.24(t,2H),3.79(s,3H),2.89(t,2H),2.83(t,2H),2.69(t,2H),2.04-1.99(m,4H).
实施例7
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-2-乙氧基-4-甲苯磺酰胺7
Figure PCTCN2022111395-appb-000124
Figure PCTCN2022111395-appb-000125
第一步
5-溴-2-乙氧基-4-甲苯磺酸7b
将化合物1-溴-4-乙氧基-2-甲苯7a(2.0g,9.30mmol)溶于浓硫酸(3.6mL)中,室温搅拌过夜。将反应液倒入冰水(20mL)中,减压浓缩除去大部分水,用环己烷(20mL)洗涤,过滤,滤饼用乙酸乙酯(20mL)洗涤,再用乙醚(20mL)洗涤,收集滤饼,真空干燥,得到标题产物7b(2.5g,产率:91.1%)。
MS m/z(ESI):295.1[M-1]
第二步
2-乙氧基-4-甲苯磺酸7c
将化合物7b(2.5g,8.47mmol)溶于甲醇(20mL)中,加钯碳(226mg,50%water),通入氢气,升温至70℃反应16小时。过滤,滤液减压浓缩,得到标题产物7c(1.8g,产率:98.3%)。
MS m/z(ESI):215.1[M-1]
第三步
2-乙氧基-4-甲苯磺酰氯7d
将化合物7c(200mg,0.92mmol)加入到烧瓶中,缓慢滴加二氯亚砜(771mg,6.48mmol),85℃反应3小时。反应液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物7d(200mg,产率:92.1%)。
第四步
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-2-乙氧基-4-甲苯磺酰胺7
将化合物2g(50mg,0.185mmol)和化合物7d(100mg,0.426mmol)溶于吡啶(2.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 5μm 30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=20%-45%(15min),流速:30mL/min),得到标题产物7(30mg,产率:34.6%)。
MS m/z(ESI):468.8[M+1]。
1H NMR(500MHz,DMSO-d 6)δ9.32(s,1H),7.78(d,1H),7.71(d,1H),7.51(d,1H),7.01(s,1H),6.89(d,1H),6.44(s,1H),6.32(t,1H),5.41(s,2H),4.25-4.19(m,2H),4.10(q,2H),2.70(t,2H),2.34(s,3H),2.02-1.97(m,2H),1.26(t,3H).
实施例8
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-4-甲基-2-(2,2,2-三氟乙氧基)苯磺酰胺8
Figure PCTCN2022111395-appb-000126
第一步
5-溴-4-甲基-2-(2,2,2-三氟乙氧基)苯磺酸8b
将1-溴-2-甲基-4-(2,2,2-三氟乙氧基)苯8a(1.2g,4.46mmol,采用专利申请“WO2020069322A1中说明书第71页的中间体A30”公开的方法制备而得)溶于浓硫酸(2mL)中,室温搅拌过夜。将反应液倒入冰水(20mL)中,减压浓缩除去大部分水,用环己烷(20mL)洗涤,过滤,滤饼用乙酸乙酯(20mL)洗涤,再用乙醚(20mL)洗涤,收集滤饼,真空干燥,得到标题产物8b(1.5g,产率:96.3%)。
MS m/z(ESI):349.1[M-1]
第二步
4-甲基-2-(2,2,2-三氟乙氧基)苯磺酸8c
将化合物8b(1.7g,4.87mmol)溶于甲醇(10mL)中,加钯碳(130mg,50%water),通入氢气,升温至70℃反应16小时。过滤,滤液减压浓缩,得到标题产物8c(1.3g,产率:98.8%)。
MS m/z(ESI):269.2[M-1]
第三步
4-甲基-2-(2,2,2-三氟乙氧基)苯磺酰氯8d
将化合物8c(500mg,1.85mmol)加入到烧瓶中,缓慢滴加二氯亚砜(1.55g,13.02mmol),85℃反应3小时。反应液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物8d(400mg,产率:74.9%)。
MS m/z(ESI):287.2[M-1]
第四步
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-4-甲基-2-(2,2,2-三氟乙氧基)苯磺酰胺8
将化合物2g(50mg,0.185mmol)和化合物8d(130mg,0.450mmol)溶于吡啶(2.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 5μm 30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=20%-45%(15min),流速:30mL/min),得到标题产物8(10mg,产率:10.3%)。
MS m/z(ESI):522.8[M+1]。
1H NMR(500MHz,DMSO-d 6)δ9.39(s,1H),7.79-7.77(m,2H),7.51(d,1H),7.20(s,1H),7.04(d,1H),6.44(s,1H),6.32(t,1H),5.41(s,2H),4.89(d,2H),4.21(t,2H),2.69(t,2H),2.37(s,3H),2.01-1.98(m,2H)。
实施例9
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-5-氟-2-甲氧基苯磺酰胺9
Figure PCTCN2022111395-appb-000127
将化合物2g(50mg,0.185mmol)和5-氟-2-甲氧基苯磺酰氯9a(200mg,0.890mmol)溶于吡啶(2.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 5μm 30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=30%-45%(15min),流速:30mL/min),得到标题产物9(15mg,产率:17.6%)。
MS m/z(ESI):458.9[M+1]。
1H NMR(500MHz,DMSO-d 6)δ10.45(s,1H),7.79(d,1H),7.57(dd,1H),7.58-7.56 (m,2H),7.27(m,1H),6.47(s,1H),6.32(t,1H),5.42(s,2H),4.17(t,2H),3.79(s,3H),2.69(t,2H),2.10-1.94(m,2H)。
实施例10
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-2-甲氧基-6-(三氟甲氧基)苯磺酰胺10
Figure PCTCN2022111395-appb-000128
第一步
2-甲氧基-6-(三氟甲氧基)苯磺酰氯10b
向100mL三口烧瓶中加入1-甲氧基-3-(三氟甲氧基)苯10a(500mg,2.55mmol)、无水四氢呋喃(10mL)和四甲基乙二胺(616mg,5.30mmol),氮气氛围下,冷却到-78℃滴加正丁基锂(1.3mL,2.5M,3.25mmol),加毕,反应在-78℃继续搅拌1小时,加入硫酰氯(0.3mL,3.71mmol),加毕,反应升到室温搅拌1小时。加水(20mL),乙酸乙酯(20mL)萃取,浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物10b(200mg,产率:26.9%)。
1H NMR(500MHz,CDCl 3)δ7.70(t,1H),7.12(d,1H),7.05(dt,1H),4.10(s,3H)。
第二步
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-2-甲氧基-6-(三氟甲氧基)苯磺酰胺10
将化合物2g(50mg,0.185mmol)和化合物10b(200mg,0.688mmol)溶于吡啶(2.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 5μm30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=20%-45%(15min),流速:30mL/min),得到标题产物10(3.0mg,产率:3.09%)。
MS m/z(ESI):524.8[M+1]。
1H NMR(500MHz,CD 3OD)δ7.66(d,1H),7.60-7.56(m,2H),7.15(d,1H),7.02(d,1H),6.40-6.38(m,2H),5.43(s,2H),4.28-4.26(m,2H),3.85(s,3H),2.69(t,2H),2.12-2.07(m,2H)。
实施例11
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-4-(二甲氨基)-2-甲氧基苯磺酰胺11
Figure PCTCN2022111395-appb-000129
第一步
4-溴-3-甲氧基-N,N-二甲基苯胺11b
向100mL烧瓶中加入4-溴-3-甲氧基苯胺11a(2.0g,9.90mmol),37%甲醛水溶液(9.0g,97.8mmol),乙酸(9.0g,150mmol),乙腈(30mL),室温搅拌30分钟,冰浴下,加入氰基硼氢化钠(800mg,12.7mmol),室温搅拌16小时。加水(100mL),乙酸乙酯(50mL)萃取,有机相浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物11b(930mg,产率:40.8%)。
MS m/z(ESI):230.0[M+1].
1H NMR(500MHz,CDCl 3)δ7.33(d,1H),6.28(d,1H),6.24(dd,1H),3.91(s,3H),2.97(s,6H).
第二步
4-(二甲氨基)-2-甲氧基苯磺酰氯11c
向100mL三口烧瓶中加入11b(900mg,3.91mmol),无水四氢呋喃(15mL),氮气氛围下,冷却到-70℃,滴加正丁基锂(2.0mL,2.5M,5.0mmol),加毕,-70℃继续搅拌1小时,通入自制二氧化硫10分钟,然后搅拌30分钟,再加入N-氯代丁二酰亚胺(700mg,5.24mmol),加毕,升到室温反应30分钟。加水(20mL),乙酸乙酯(20mL)萃取,浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物11c(50mg,产率:5.11%)。
MS m/z(ESI):232.0[M-Cl+OH+1]。
第三步
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-4-(二甲氨基)-2-甲氧基苯磺酰胺11
将化合物2g(40mg,0.148mmol)和化合物11c(50mg,0.200mmol)溶于吡啶(2.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 5μm30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=10%-45%(15min),流速:30mL/min),得到标题产物11(2.0mg,产率:2.79%)。
MS m/z(ESI):483.9[M+1]。
1H NMR(500MHz,CD 3OD)δ7.72(d,1H),7.66(d,1H),7.57(d,1H),6.43-6.38(m,2H),6.33(dd,1H),6.19(d,1H),5.42(s,2H),4.38(t,2H),3.87(s,3H),3.03(s,6H),2.70(t,2H),2.18-2.12(m,2H)。
实施例12
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-2,3-二氢苯并呋喃-7-磺酰胺12
Figure PCTCN2022111395-appb-000130
第一步
2,3-二氢苯并呋喃-7-磺酰氯12b
向100mL烧瓶中加入2,3-二氢苯并呋喃12a(2.0g,16.6mmol),三氧化硫N,N-二甲基甲酰胺络合物(3.0g,19.6mmol),1,2-二氯乙烷(10mL),反应在80℃搅拌1小时。冷却到室温,滴加氯化亚砜(2.2g,18.5mmol),加毕,反应升温至70℃反应2小时。然后冷却到室温,倒入冰水(50mL),乙酸乙酯(50mL)萃取,浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物12b(3.2g,产率:87.9%)。
1H NMR(500MHz,CDCl 3)δ7.89-7.86(m,2H),6.93(d,1H),4.78(t,2H),3.35(t,2H)。
第二步
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-2,3-二氢苯并呋 喃-7-磺酰胺12
将化合物2g(50mg,0.185mmol)和化合物12b(200mg,0.915mmol)溶于吡啶(2.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 5μm 30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=30%-45%(15min),流速:30mL/min),得到标题产物12(20.0mg,产率:23.9%)。
MS m/z(ESI):452.9[M+1]。
1H NMR(500MHz,DMSO-d 6)δ10.56(s,1H),7.83(d,1H),7.78(d,1H),7.76(d,1H),7.52(d,1H),6.93(d,1H),6.44(s,1H),6.32(t,1H),5.42(s,2H),4.64(t,2H),4.30-4.20(m,2H),3.25(t,2H),2.69(t,2H),2.17-1.95(m,2H)。
实施例13
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-4-乙基-2-甲氧基苯磺酰胺13
Figure PCTCN2022111395-appb-000131
第一步
4-溴-3-乙基苯酚13b
将化合物3-乙基苯酚13a(4.0g,32.7mmol,adamas)溶于二氯甲烷(30mL),加入四丁基三溴化铵(16.0g,33.2mmol),室温搅拌2小时。反应液分别用1M稀盐酸(50mL)、水、饱和氯化钠水溶液洗涤,减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物13b(6.3g,产率:95.7%)。
1H NMR(500MHz,CDCl 3)δ7.38(d,1H),6.76(d,1H),6.58(dd,1H),4.83(s,1H),2.71(q,2H),1.23(t,3H)。
第二步
1-溴-2-乙基-4-甲氧基苯13c
将化合物13b(3.0g,14.9mmol)溶于N,N-二甲基甲酰胺(40mL)中,加入碳酸钾(4.1g,29.7mmol)和碘甲烷(2.6g,18.3mmol),室温反应16小时。反应液用乙酸乙酯(100mL)稀释,然后分别用水(100mL)、饱和氯化钠水溶液(100mL)洗涤,减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物13c(2.5g,产率:77.9%)。
1H NMR(500MHz,CDCl 3)δ7.42(d,1H),6.81(d,1H),6.64(dd,1H),3.81(s,3H),2.74(q,2H),1.24(t,3H)。
第三步
5-溴-4-乙基-2-甲氧基苯磺酸13d
冰浴下,将化合物13c(2.5g,11.6mmol)慢慢加入浓硫酸(6.0mL)中,室温搅拌2小时。将反应液倒入冰水(90mL)中,用20%氢氧化钠水溶液调节反应液pH=3,过滤,滤饼用水(100mL)洗涤,干燥,得到标题产物13d(1.5g,产率:43.7%)。MS m/z(ESI):293.1[M-1]。
第四步
4-乙基-2-甲氧基苯磺酸13e
将化合物13d(1.5g,5.08mmol)溶于甲醇(20mL)中,加钯碳(600mg,10%,50%water),通入氢气,反应升温至60℃搅拌16小时。过滤,滤液减压浓缩,得到标题产物13e(1.1g,产率:100%)。
MS m/z(ESI):215.2[M-1]。
第五步
4-乙基-2-甲氧基苯磺酰氯13f
将化合物13e(400mg,1.85mmol)加入到烧瓶中,缓慢滴加二氯亚砜(4.0mL),85℃反应3小时。反应液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物13f(200mg,产率:46.1%)。
MS m/z(ESI):215.2[M-Cl+OH-1]。
第六步
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-4-乙基-2-甲氧基苯磺酰胺13
将化合物13f(200mg,0.852mmol)和化合物2g(60mg,0.222mmol)溶于吡啶(2.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 5μm 30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=15%-45%(15min),流速:30mL/min),得到标题产物13(15.0mg,产率:14.4%)。
MS m/z(ESI):468.9[M+1]。
1H NMR(500MHz,DMSO-d 6)δ9.77(s,1H),7.78(d,1H),7.70(d,1H),7.51(d,1H), 7.03(s,1H),6.92(d,1H),6.43(s,1H),6.31(t,1H),5.40(s,2H),4.24-4.18(m,2H),3.81(s,3H),2.69(t,2H),2.64(t,2H),2.01-1.97(m,2H),1.19(t,3H).
实施例14
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-4-异丙基-2-甲氧基苯磺酰胺14
Figure PCTCN2022111395-appb-000132
第一步
4-溴-3-异丙基苯酚14b
将化合物3-异丙基苯酚14a(2.0g,14.7mmol)溶于二氯甲烷(30mL)和甲醇(10mL),加入四丁基三溴化铵(7.1g,14.7mmol),室温搅拌2小时。反应液分别用1M稀盐酸(50mL)、水、饱和氯化钠水溶液洗涤,减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物14b(2.9g, 1NMR显示含~20%Br在异丙基对位的异构体,总产率:95.7%,直接投下一步)。
1H NMR(500MHz,CDCl 3)δ7.38(d,1H),6.79(d,1H),6.57(dd,1H),4.99(s,1H),3.31(m,1H),1.24(d,6H)。
第二步
1-溴-2-异丙基-4-甲氧基苯14c
将化合物14b(2.9g,13.5mmol)溶于N,N-二甲基甲酰胺(30mL)中,加入碳酸钾(3.7g,26.8mmol)和碘甲烷(2.28g,16.1mmol),室温反应16小时。反应液用乙酸乙酯(100mL)稀释,然后分别用水(100mL)、饱和氯化钠水溶液(100mL)洗涤,减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物14c(2.4g, 1NMR显示含~10%Br在异丙基对位的异构体,总产率:77.7%,直接投下一步)。
1H NMR(500MHz,CDCl 3)δ7.44(d,1H),6.86(d,1H),6.64(dd,1H),3.81(s,3H), 3.33(m,1H),1.25(d,6H)。
第三步
5-溴-4-异丙基-2-甲氧基苯磺酸14d
冰浴下,将化合物14c(2.4g,10.5mmol)慢慢加入浓硫酸(6.0mL)中,室温搅拌2小时。将反应液倒入冰水(90mL)中,用20%氢氧化钠水溶液调节反应液pH=3,过滤,滤饼用水(100mL)洗涤,干燥,得到标题产物14d(2.4g,产率:74.1%)。
MS m/z(ESI):307.1[M-1]。
第四步
4-异丙基-2-甲氧基苯磺酸14e
将化合物14d(2.4g,7.76mmol)溶于甲醇(30mL)中,加钯碳(1.0g,10%,50%water),通入氢气,升温至60℃反应16小时。过滤,滤液减压浓缩,得到标题产物14e(1.3g,产率:72.7%)。
MS m/z(ESI):229.2[M-1]。
第五步
4-异丙基-2-甲氧基苯磺酰氯14f
将化合物14e(360mg,1.56mmol)加入到烧瓶中,缓慢滴加二氯亚砜(4.0mL),85℃反应3小时。反应液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物14f(250mg,产率:64.3%)。
MS m/z(ESI):229.2[M-Cl+OH-1]。
第六步
N-(5-((1H-吡唑-1-基)甲基)-3,4-二氢-2H-色烯并[8,7-d]异噁唑-9-基)-4-异丙基-2-甲氧基苯磺酰胺14
将化合物14f(250mg,1.01mmol)和化合物2g(60mg,0.222mmol)溶于吡啶(2.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 5μm 30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=15%-45%(15min),流速:30mL/min),得到标题产物14(10.0mg,产率:9.34%)。
MS m/z(ESI):482.9[M+1]。
1H NMR(500MHz,DMSO-d 6)δ9.94(s,1H),7.75(d,1H),7.68(d,1H),7.51(d,1H),6.94(s,1H),6.87(d,1H),6.31(d,2H),5.37(s,2H),4.24-4.16(m,2H),3.77(s,3H),2.91(m,1H),2.67(t,2H),2.06-1.94(m,2H),1.21(d,6H)。
实施例15
N-(4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-6-甲氧基-2,3-二氢-1H-茚-5-磺酰胺15
Figure PCTCN2022111395-appb-000133
将化合物6a(289mg,1.17mmol)和化合物1n(150mg,0.59mmol)溶于吡啶(5.0mL)中,加入4-二甲氨基吡啶(15mg,0.12mmol),置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Xtimate phenyl-hexyl Prep C18 5μm,30×150mm;流动相:A-水相(0.1%氨水):B-乙腈=5%-45%(20min),流速:30mL/min),得到标题产15(30mg,产率:11.0%)。
MS m/z(ESI):467.5[M+1]。
1H NMR(400MHz,CDCl 3):δ7.90(s,1H),7.57-7.56(d,1H),7.42-7.41(d,1H),6.84(s,1H),6.64(s,1H),6.33-6.32(m,1H),5.33(s,2H),4.83-4.80(m,2H),3.94(s,3H),3.09-3.05(m,2H),2.94-2.88(m,4H),2.13-2.07(m,2H)。
实施例16
N-(4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2-乙氧基-4-甲基苯磺酰胺16
Figure PCTCN2022111395-appb-000134
将化合物7d(92mg,0.39mmol)和化合物1n(50mg,0.20mmol)溶于吡啶(5.0mL)中,加入4-二甲氨基吡啶(3mg,0.02mmol),置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Xtimate phenyl-hexyl Prep C18 5μm,30×150mm;流动相:A-水相(0.1%氨水):B-乙腈=5%-45%(20min),流速:30mL/min),得到标题产16(10mg,产率:11.3%)。
MS m/z(ESI):455.5[M+1]。
1H NMR(400MHz,CDCl 3):δ7.97-7.96(d,1H),7.56-7.55(d,1H),7.42-7.41(d,1H),6.89-6.87(d,1H),6.75(s,1H),6.66(s,1H),6.33-6.32(m,1H),5.33(s,2H),4.83-4.79(m,2H),4.19-4.15(m,2H),3.09-3.06(m,2H),2.37(s,3H),1.53-1.50(m,3H)。
实施例17
N-(4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2-甲氧基-6-甲基苯磺酰胺17
Figure PCTCN2022111395-appb-000135
第一步
2-甲氧基-6-甲基苯磺酰氯17b
-70℃下,向2-溴-1-甲氧基-3-甲基苯17a(500.0mg,2.5mmol)的无水乙醚(10mL)溶液中,滴加正丁基锂(1.0mL,2.5mmol,2.5M正己烷溶液)。氮气保护下,-70℃搅拌反应1小时。-70℃下,向反应液中通二氧化硫气体30分钟。加入N-氯代丁二酰亚胺(496.0g,3.8mmol),缓慢升至室温后,室温反应2小时。反应液用饱和亚硫酸氢钠溶液(20mL)和饱和氯化钠溶液(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物17b(390.0mg,产率:78.0%)。
第二步
N-(4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2-甲氧基-6-甲基苯磺酰胺17
-70℃下,向化合物1n(60.0mg,0.24mmol)无水四氢呋喃(3mL)溶液中,滴加双(三甲基硅基)胺基锂(0.4mL,0.4mmol,1M四氢呋喃溶液)。氮气保护下,-70℃搅拌1小时。-70℃下,滴加化合物17b(78.0mg,0.35mmol)的四氢呋喃(0.5mL)溶液。反应体系缓慢升至室温后,室温搅拌反应16小时。向反应液中加入饱和氯化铵溶液(5mL),用乙酸乙酯萃取(20mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相色谱法纯化(Welch Xtimate C18,5μm,30mm*150mm,洗脱体系:水(10mM碳酸氢铵)、乙腈,乙腈在14分钟内由20%(v/v) 升至34%(v/v),检测波长214&254nm),得到标题产物17(25.0mg,产率:23.7%)。
MS m/z(ESI):440.9[M+1]。
1H NMR(500MHz,CD 3OD):δ7.70(d,1H),7.54(d,1H),7.36(dd,1H),6.95(d,1H),6.87(d,1H),6.53(s,1H),6.36(t,1H),5.40(s,2H),4.77(t,2H),3.82(s,3H),3.07(t,2H),2.65(s,3H)。
实施例18
N-(4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2-甲氧基-5-甲基苯磺酰胺18
Figure PCTCN2022111395-appb-000136
将化合物2-甲氧基-5-甲基苯磺酰氯18a(130mg,0.59mmol)和化合物1n(50mg,0.20mmol)溶于吡啶(5.0mL)中,加入4-二甲氨基吡啶(5mg,0.04mmol),置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Xtimate phenyl-hexyl Prep C18 5μm,30×150mm;流动相:A-水相(0.1%氨水):B-乙腈=5%-45%(20min),流速:30mL/min),得到标题产18(3mg,产率:3.5%)。
MS m/z(ESI):441.3[M+1]。
1H NMR(400MHz,CDCl 3):δ7.80(s,1H),7.57-7.56(m,2H),7.49-7.48(m,2H),6.30-6.29(m,2H),5.37(s,2H),4.70-4.67(m,2H),3.71(s,3H),3.09-3.06(m,2H),2.25(s,3H)。
实施例19
N-(4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-5-甲氧基-2,3-二氢苯并呋喃-6-磺酰胺19
Figure PCTCN2022111395-appb-000137
Figure PCTCN2022111395-appb-000138
第一步
6-溴-5-甲氧基-2,3-二氢苯并呋喃19b
将5-甲氧基-2,3-二氢苯并呋喃19a(250mg,1.66mmol,采用专利申请“WO2017218960A1中说明书第65页的中间体1D”公开的方法制备而得)加入二氯甲烷(5mL)中,0℃下,缓慢加入1,3-二溴-5,5-二甲基咪唑啉-2,4-二酮(291mg,0.67mmol),0℃反应2小时。反应液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物19b(330mg,产率:86.5%)。
MS m/z(ESI):229.1[M+1]。
第二步
5-甲氧基-2,3-二氢苯并呋喃-6-磺酰氯19c
将化合物19b(390mg,1.70mmol)溶于乙醚(10mL)中,-78℃滴加正丁基锂(0.69mL,1.72mmol,2.5M四氢呋喃溶液),-78℃搅拌1小时。向反应液中通二氧化硫气体30分钟,加入N-氯代丁二酰亚胺(342mg,2.56mmol),室温搅拌2小时,加入饱和氯化铵溶液(5mL),用乙酸乙酯萃取(20mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物19c(200mg,产率:47.2%)。
第三步
N-(4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-5-甲氧基-2,3-二氢苯并呋喃-6-磺酰胺19
将化合物19c(194mg,0.78mmol)和化合物1n(100mg,0.39mmol),3,5-二甲基吡啶(168mg,1.57mmol),二甲亚砜(2mg,0.03mmol)溶于乙腈(10mL)中,反应液在室温搅拌过夜。减压浓缩,所得残余物经高效液相色谱法纯化(Xtimate phenyl-hexyl Prep C18 5μm,30×150mm;流动相:A-水相(0.1%氨水):B-乙腈=5%-45%(20min),流速:30mL/min),得到标题产19(50mg,产率:27.3%)。
MS m/z(ESI):469.1[M+1]。
1H NMR(400MHz,DMSO-d 6):δ7.80(s,1H),7.49-7.48(m,1H),7.09-7.06(m,3H),6.29-6.28(m,1H),5.36(s,2H),4.70-4.67(m,2H),4.52-4.48(m,2H),3.68(s,3H),3.08-3.07(m,2H),3.06-3.05(m,2H)。
实施例20
N-(4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2-甲氧基苯磺酰胺20
Figure PCTCN2022111395-appb-000139
将化合物1n(100mg,0.39mmol)和化合物3a(120mg,0.58mmol)溶于吡啶(5.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 5μm 30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=20%-45%(15min),流速:30mL/min),得到标题产物20(19mg,产率:11.4%)。
MS m/z(ESI):427.1[M+1]。
1H NMR(500MHz,CD 3OD)δ7.91(dd,1H),7.72(d,1H),7.62-7.57(m,1H),7.56(d,1H),7.14(d,1H),7.07(t,1H),6.59(s,1H),6.38(t,1H),5.42(s,2H),4.78(t,2H),3.87(s,3H),3.09(t,2H)。
实施例21
N-(4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-4-甲基-2-(2,2,2-三氟乙氧基)苯磺酰胺21
Figure PCTCN2022111395-appb-000140
将化合物8d(113mg,0.39mmol)和化合物1n(50mg,0.20mmol)溶于吡啶(5.0mL)中,加入4-二甲氨基吡啶(5mg,0.04mmol),置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Xtimate phenyl-hexyl Prep C18 5μm,30×150mm;流动相:A-水相(0.1%氨水):B-乙腈=5%-45%(20min),流速:30mL/min),得到标题产物21(35mg,产率:35.3%)。
MS m/z(ESI):509.3[M+1]。
1H NMR(400MHz,CDCl 3):δ8.07-8.05(d,1H),7.56-7.55(d,1H),7.41-7.40(d,1H),7.06-7.04(d,1H),6.78(s,1H),6.64(s,1H),6.33-6.32(m,1H),5.33(s,2H),4.82-4.78(m,2H),4.52-4.48(m,2H),3.08-3.04(m,2H),2.42(s,3H)。
实施例22
N-(4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2-甲氧基-4-甲基苯磺酰胺22
Figure PCTCN2022111395-appb-000141
将2-甲氧基-4-甲基苯磺酰氯22a(78mg,35mmol)和化合物1n(30mg,0.12mmol)溶于吡啶(5.0mL)中,加入4-二甲氨基吡啶(3mg,0.02mmol),置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Xtimate phenyl-hexyl Prep C18 5μm,30×150mm;流动相:A-水相(0.1%氨水):B-乙腈=5%-45%(20min),流速:30mL/min),得到标题产22(5mg,产率:9.7%)。
MS m/z(ESI):441.5[M+1]。
1H NMR(400MHz,DMSO-d 6):δ7.82(s,1H),7.62-7.60(m,1H),7.50-7.49(m,1H),6.99-6.97(m,1H),6.84-6.82(m,1H),6.65-6.63(m,1H),6.30(s,1H),5.40(s,2H),4.73-4.69(m,2H),3.76(s,3H),3.11-3.08(m,2H),2.35(s,3H)。
实施例23
N-(4-((1H-吡唑-1-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2-甲氧基-6-(三氟甲氧基)苯磺酰胺23
Figure PCTCN2022111395-appb-000142
Figure PCTCN2022111395-appb-000143
将化合物1n(80mg,0.31mmol)与化合物10b(185mg,0.63mmol)溶于乙腈(3mL)中,加入二甲亚砜(3mg,0.04mmol)与3,5-二甲基吡啶(135mg,1.26mmol),反应液在室温下反应16小时。反应液过滤,滤液减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 5μm 30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=20%-40%(10min),流速:30mL/min),得到标题产物23(16mg,产率:10.0%)。
MS m/z(ESI):511.0[M+1]。
1H NMR(500MHz,DMSO-d 6)δ7.79(s,1H),7.48(s,1H),7.38(s,1H),7.10(s,2H),6.86(s,1H),6.37(s,1H),6.29(m,1H),5.34(s,2H),4.65(t,2H),3.68(s,3H),3.05(t,2H)。
实施例24
2,6-二甲氧基-N-(4-(吡啶-2-基甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)苯磺酰胺24
Figure PCTCN2022111395-appb-000144
第一步
4-溴-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-胺24a
将化合物1i(2.3g,9.59mmol)和乙酰氧肟酸(2.2g,28.85mmol)溶于N,N-二甲基甲酰胺(25mL)和水(2.5mL)的混合液中,加入碳酸钾(7.9g,57.54mmol)。反应液在65℃下搅拌16小时。反应液过滤,得到标题产物24a(1.7g,产率:67.5%)。
MS m/z(ESI):255.0[M+1]。
第二步
N-(4-溴-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2,6-二甲氧基苯磺酰胺24b
将化合物24a(300mg,1.18mmol)和化合物1o(835mg,3.53mmol)溶于吡啶(8mL)中,加入4-二甲氨基吡啶(29mg,0.24mmol),置换氮气3次。反应液在120℃下微波反应2小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(Welch Xtimate C18 5μm 30×150mm;流动相:A-水相(0.1%甲酸):B-乙腈=30%-45%(15min),流速:30mL/min),得到标题产物24b(130mg,产率:24.3%)。MS m/z(ESI):455.1[M+1]。
第三步
(±)-N-(4-(羟基(吡啶-2-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2,6-二甲氧基苯磺酰胺24c
将化合物24b(30mg,0.065mmol)溶于四氢呋喃(5mL)中,-78℃滴加正丁基锂(0.06mL,0.16mmol,2.5M四氢呋喃溶液),-78℃搅拌1小时。向反应液中加入吡啶-2-甲醛(9mg,0.084mmol),室温搅拌过夜,向反应液中加饱和氯化铵溶液(5mL),用乙酸乙酯萃取(20mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物24c(5mg,产率:15.7%)。
MS m/z(ESI):484.3[M+1]。
第四步
(±)-N-(4-(氯(吡啶-2-基)甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2,6-二甲氧基苯磺酰胺24d
将化合物24c(5mg,0.01mmol)加入到二氯甲烷(5mL)中,缓慢滴加二氯亚砜(13mg,0.1mmol),室温反应3小时。反应液减压浓缩,溶于乙酸乙酯(20mL)中,用饱和碳酸氢钠溶液(20mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到标题产物24d(5mg,产率:96.3%)。
MS m/z(ESI):502.3[M+1]。
第五步
2,6-二甲氧基-N-(4-(吡啶-2-基甲基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)苯磺酰胺24
将化合物24d(5mg,0.01mmol)加入到乙酸(1mL)中,加锌粉(1mg,0.015mmol),60℃反应3小时。反应液减压浓缩,所得残余物经高效液相色谱法纯化(Xtimate phenyl-hexyl Prep C18 5μm,30×150mm;流动相:A-水相(0.1%氨水):B-乙腈=5%-45%(20min),流速:30mL/min),得到标题产物24(2.5mg,产率:53.7%)。MS m/z(ESI):468.4[M+1]。
1H NMR(400MHz,DMSO-d 6):δ8.49-8.48(m,1H),7.80-7.77(m,2H),7.42(s,1H), 7.32-7.28(m,2H),6.72-6.71(m,2H),4.77-4.76(m,2H),4.60(s,3H),4.20(s,2H),3.78(s,3H),3.13-3.10(m,2H)。
实施例25
N-(4-(呋喃-2-基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2-甲氧基苯磺酰胺25
Figure PCTCN2022111395-appb-000145
第一步
4-(4,5-二氢呋喃-2-基)-6-氟-2,3-二氢苯并呋喃-7-甲腈25b
将化合物1i(300mg,1.24mmol)和2,3-二氢呋喃25a(435mg,6.20mmol)溶于N,N-二甲基甲酰胺(10mL)中,置换氮气3次,加入醋酸钯(28mg,0.12mmol)、三苯基膦(65mg,0.24mmol)与碳酸钾(345mg,2.49mmol),反应液在110℃反应16小时。反应液冷却至室温,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物25b(100mg,产率:34.9%)。
MS m/z(ESI):232.0[M+1]。
第二步
4-(呋喃-2-基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-胺25c
将化合物25b(100mg,0.43mmol)和乙酰氧肟酸(100mg,1.33mmol)溶于4mL N,N-二甲基甲酰胺与水的混合溶剂(V:V=7:1)中,加入碳酸钾(360mg,2.60mmol)。反应液在70℃下搅拌30分钟。反应液冷却到室温,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题产物25c(22mg,产率:21.0%)。
MS m/z(ESI):243.0[M+1]。
1H NMR(500MHz,CDCl 3)δ7.56(dd,1H),7.27(s,1H),6.69(dd,1H),6.56(dd,1H),4.86(t,2H),4.52(s,2H),3.49(t,2H)。
第三步
N-(4-(呋喃-2-基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)-2-甲氧基苯磺酰胺25
将化合物25c(22mg,0.09mmol)和化合物3a(95mg,0.46mmol)溶于吡啶(1.0mL)中,置换氮气3次。反应液在120℃下微波反应3小时。反应液冷却至室温,减压浓缩,所得残余物经高效液相色谱法纯化(welch Prep C18 5μm 30×150mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=25%-45%(20min),流速:30mL/min),得到标题产物25(5mg,产率:13.3%)。
MS m/z(ESI):413.0[M+1]。
1H NMR(500MHz,DMSO-d 6)δ10.85(s,1H),7.88(s,1H),7.78(dd,1H),7.61(s,1H),7.37(s,1H),7.19(s,1H),7.06(s,1H),7.01(s,1H),6.70(dd,1H),4.79(t,2H),3.79(s,3H),3.44(t,2H)。
实施例26
2,6-二甲氧基-N-(4-(四氢呋喃-2-基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)苯磺酰胺26
Figure PCTCN2022111395-appb-000146
将化合物24b(500mg,1.09mmol)和化合物25a(430mg,6.13mmol)溶于N,N-二甲基甲酰胺(10mL)中,置换氮气3次,加入醋酸钯(30mg,0.13mmol)、三苯基膦(60mg,0.22mmol)与碳酸钾(320mg,2.31mmol),反应液在110℃反应16小时。反应液冷却至室温,置换氢气3次,于室温下继续反应6小时。反应液过滤,滤液减压浓缩,所得残余物经高效液相色谱法纯化(Sharpsil-T Prep C18 8μm 50×250mm;流动相:A-水相(10mM碳酸氢铵):B-乙腈=18%-38%(20min),流速:80mL/min),得到标题产物26(203mg,产率:41.4%)。
MS m/z(ESI):447.0[M+1]。
1H NMR(500MHz,CDCl 3)δ7.40(t,1H),6.99(s,1H),6.61(d,2H),4.94(t,1H),4.90-4.78(m,2H),4.12(dt,1H),3.96(dt,1H),3.93(s,6H),3.25(t,2H),2.36(dq,1H),2.09-1.99(m,2H),1.76(dq,1H)。
实施例26-1和实施例26-2
(R)-2,6-二甲氧基-N-(4-(四氢呋喃-2-基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)苯 磺酰胺26-1
(S)-2,6-二甲氧基-N-(4-(四氢呋喃-2-基)-2,3-二氢苯并呋喃并[7,6-d]异噁唑-8-基)苯磺酰胺26-2
Figure PCTCN2022111395-appb-000147
将化合物26(203mg,0.45mmol)进行手性制备(分离条件:CHIRALPAK IE手性制备柱,20mm×250mm;流动相:正己烷/乙醇/三氟乙酸=60/40/0.1(V/V/V),流速:20mL/min),收集其相应组分,减压浓缩,分别得到标题产物26-1(60mg)和26-2(56mg)。
26-1(较长保留时间,60mg):
MS m/z(ESI):447.0[M+1]。
手性HPLC分析:保留时间36.6分钟,手性纯度:100%(色谱柱:CHIRALPAK IE,20mm×250mm,5μm;流动相:正己烷/乙醇/三氟乙酸=60/40/0.1(V/V/V)。
1H NMR(500MHz,CDCl 3)δ7.40(t,1H),6.99(s,1H),6.61(d,2H),4.94(t,1H),4.90-4.78(m,2H),4.12(dt,1H),3.96(dt,1H),3.93(s,6H),3.25(t,2H),2.36(dq,1H),2.09-1.99(m,2H),1.76(dq,1H)。
26-2(较短保留时间,56mg):
MS m/z(ESI):447.0[M+1]。
手性HPLC分析:保留时间26.3分钟,手性纯度:100%(色谱柱:CHIRALPAK IE,20mm×250mm,5μm;流动相:正己烷/乙醇/三氟乙酸=60/40/0.1(V/V/V)。
1H NMR(500MHz,CDCl 3)δ7.40(t,1H),6.99(s,1H),6.61(d,2H),4.94(t,1H),4.90-4.78(m,2H),4.12(dt,1H),3.96(dt,1H),3.93(s,6H),3.25(t,2H),2.36(dq,1H),2.09-1.99(m,2H),1.76(dq,1H)。
生物学评价
以下结合测试例进一步描述解释本公开,但这些测试例并非意味着限制本公开的范围。
测试例1 KAT6酶活性检测(AlphaScreen法)
一、试剂与仪器
1.KAT6A(Chempartner订制)
2.卵清蛋白(Sigma-Aldrich,A5378-5G)
3. 2M Tris-HCl溶液,pH 7.8,无菌(生工B548140-0500)
4. 5M NaCl溶液(生工,B548121-0100)
5.EDTA(0.5M),pH 8.0,RNase-free(Thermofisher,AM9260G)
6.吐温-20(生工,A100777-0500)
7.DTT,1M(Invitrogen,P2325)
8.乙酰辅酶A(Ac-CoA,CAYMAN,Cat.No.16160)
9.生物素标记重组组蛋白H3.1(人)(Recombinant Histone H3.1 biotinylated(Human))(Active Motif 31696)
10. 384孔板,浅灰色(Perkin Elmer,Cat.No.6005350)
11.漆树酸(MCE,Cat.No.HY-N2020)
12.AlphaScreen链霉亲和素供体珠(AlphaScreen Streptavidin Donor beads)5mg(PerkinElmer,6760002)
13.AlphaScreen蛋白A受体珠(AlphaScreen Protein A Acceptor beads),5mg(PerkinElmer,6760137M)
14.乙酰化赖氨酸抗体(Acetylated-Lysine Antibody#9441)(CST 9441S)
15.PHERA star酶标仪(BMG labtech)
二、实验方法
1.试剂准备
a.1×检测缓冲液:100mM Tris-HCL,PH7.8;15mM NaCl;1mM EDTA;0.01%吐温-20;1mM DTT;0.01%m/v卵清蛋白。
b.KAT酶溶液:1×检测缓冲液配制终浓度1.25nM。
c.Ac-CoA和H3混合底物:1×检测缓冲液配制终浓度1000nM的Ac-CoA和终浓度55nM的H3的混合底物。
d.化合物:起始浓度100μM,3倍稀释,10个浓度梯度。所有浓度化合物用1×检测缓冲液稀释83倍,备用。
e.检测试剂:1×检测缓冲液配制终浓度8ng/μL的AlphaScreen蛋白A受体珠,8ng/μL的AlphaScreen链霉亲和素供体珠,乙酰化赖氨酸抗体1:1500稀释,100μM漆树酸。
2.实验步骤
a.384孔板,每孔加入3μL配制好的酶溶液,第23和24列孔(Min)加3μL1×检测缓冲液。
b.每孔加3μL化合物溶液,Min每孔加入3μL buffer;第1和2列(Max)每孔加入3μL DMSO溶液作对照。离心,混匀震荡2分钟;室温孵育15分钟。
c.每孔加入6μL Ac-CoA和H3混合底物,离心,混匀震荡2分钟,室温孵育20分钟。
d.每孔加6μL检测试剂,离心,混匀震荡2分钟,室温孵育120分钟,避光。
e.酶标仪读板,记录AlphaScreen计数的数值。
f.Graphpad软件作图,计算化合物IC 50值。
表1本公开化合物对人KAT6A酶抑制的IC 50
化合物编号 KAT6A/IC 50(nM)
1 0.6
2 0.3
3 0.5
4 4.2
5 1.0
6 0.4
7 0.5
8 0.7
9 0.7
10 0.9
11 1.6
12 12.5
13 0.3
15 1.2
16 1.9
17 10.5
18 12.5
19 2.7
20 2.9
21 4.6
22 4.7
23 4.8
26 12.2
26-1 5.2
结论:本公开化合物对KAT6A具有很好的抑制作用。
测试例2 KAT6B酶活性检测(AlphaScreen法)
一、试剂与仪器
1.KAT6B(718-1008)(ActiveMotif,81224)
2.牛血清白蛋白(生工,A500023-0100)
3. 2M Tris-HCl溶液,pH 7.8,无菌(生工B548140-0500)
4.EDTA(0.5M),pH 8.0,RNase-free(Thermofisher,AM9260G)
5.吐温-20(生工,A100777-0500)
6.DTT,1M(Invitrogen,P2325)
7.乙酰辅酶A(Ac-CoA,CAYMAN,Cat.No.16160)
8.生物素标记重组组蛋白H3.1(人)(Active Motif 31696)
9. 384孔板,浅灰色(Perkin Elmer,Cat.No.6007290)
10.漆树酸(MCE,Cat.No.HY-N2020)
11.AlphaScreen链霉亲和素供体珠5mg(PerkinElmer,6760002)
12.AlphaScreen蛋白A受体珠,5mg(PerkinElmer,6760137M)
13.乙酰化赖氨酸抗体#9441(CST 9441S)
14.DMSO(Tansoole,G7592B)
15.PHERA star酶标仪(BMG labtech)
二、实验方法
1.试剂准备
a.1×缓冲液2:50mM Tris-HCl,pH7.8;0.1mM EDTA;0.01%v/v吐温-20;1mM DTT;0.01%m/v牛血清白蛋白。
b.KAT6B酶溶液:缓冲液2配制终浓度3nM
c.Ac-CoA和H3混合底物:缓冲液2配制终浓度30nM Ac-CoA和30nM H3混合底物。
d.化合物:起始浓度10mM,4倍稀释,10个浓度梯度。所有浓度化合物用缓冲液2稀释2500倍,备用。
e.检测试剂:缓冲液2配制终浓度8ng/μL的AlphaScreen蛋白A受体珠,8ng/μL的AlphaScreen链霉亲和素供体珠,乙酰化赖氨酸抗体1:1000稀释,100μM漆树酸。
2.实验步骤
a.384孔板,每孔加2μL配制好的化合物溶液,Min和Max每孔加入2μL缓冲液2(含0.04%DMSO)作对照,离心。
b.每孔加入2μL配制好的酶溶液,Min加2μL缓冲液2。离心,混匀震荡2分钟;室温孵育10分钟。
c.每孔加入4μL Ac-CoA和H3混合底物,离心,混匀震荡2分钟,室温孵育120分钟。
d.每孔加4μL检测试剂,离心,混匀震荡2分钟,避光室温孵育120分钟。
e.酶标仪读板,记录AlphaScreen计数的数值。
f.使用graphpad prism软件的log(抑制剂)vs.应答,横坐标为化合物浓度对数值,纵坐标为计算的酶活性抑制率,绘制量效曲线并计算IC 50
表2本公开化合物对人KAT6B酶抑制的IC 50
化合物编号 KAT6B/IC 50(nM)
1 1.1
2 0.7
3 1.4
5 1.5
结论:本公开化合物对KAT6B具有很好的抑制作用。
测试例3 U2OS细胞H3K23乙酰化IF检测(Immunofluorescence)
一、试剂与仪器
1.U-2 OS(ATCC HTB-96)
2.重组抗组蛋白H3(乙酰基K23)抗体(Recombinant Anti-Histone H3(acetyl K23)antibody)(Abcam,ab177275)
3.山羊抗兔IgG(H+L)(Goat anti-Rabbit IgG(H+L)),Superclonal TM重组二抗(Superclonal TMRecombinant Secondary Antibody),Alexa Fluor 488(Thermofisher,A27034)
4.Hoechst 33342(Sigma-Aldrich,B2261-25MG)
5. 96孔底部透明黑色检测板(Assay plate,96well,Black with clear bottom)(Corning,3603)
6.牛血清白蛋白(BSA)(Sangon Biotech,A500023-0100)
7.甲醇(GENMERAL-REAGENT,G75851D)
8.吐温-20(生工,A100777-0500)
9.Triton X-100(Solarbio,T8200)
10.PBS(上海源培生物科技股份有限公司,B320KJ)
11. 20X PBS缓冲液(Sangon Biotech,B548117-0500)
12.McCoy's 5A培养基(Gibco,16600082)
13. 0.25%胰蛋白酶-EDTA(1x)(Gibco,25200-072)
14.Pen strep(Gibco,15140-122)
15.DPBS(1x)(Gibco,14190-144)
16.FBS(Gibco,10091148)
17.自动细胞计数器(Countstar,IC1000)
18.恒温培养箱(Thermo,I160)
19.
Figure PCTCN2022111395-appb-000148
Micro Confocal(Molecular Device)
二、实验方法
1.试剂准备
a.封闭缓冲液:PBS(上海源培)+BSA(终浓度1%)+Triton X-100(终浓度0.5%)。
b.洗涤缓冲液:PBS(20×PBS稀释为1×PBS)+吐温-20(终浓度0.1%)。
c.一抗溶液:重组抗组蛋白H3(乙酰基K23)抗体按1:1000稀释比,用封闭缓冲液稀释。
d.二抗溶液:山羊抗兔IgG(H+L),Superclonal TM重组二抗,Alexa Fluor 488按1:1000稀释比,Hoechst 33342按1:5000稀释比,用封闭缓冲液稀释。
e.化合物:起始浓度100μM,3倍稀释,9个浓度梯度。所有浓度化合物用McCoy's 5A培养基稀释500倍,备用。
2.实验步骤
2.1处理细胞(第一天)
a.显微镜下观察U-2OS细胞状态,确保细胞的融合度在90%左右。
b.弃去细胞的上清,DPBS润洗一次,倒去DPBS。加入适量胰蛋白酶消化细胞,室温或37℃静置5分钟。
c.用等体积含10%FBS的培养基终止消化,收集细胞悬液。300g,离心3分钟。用适量新鲜培养基悬浮细胞。
d.取重悬的细胞悬液进行计数。
e.稀释细胞悬液,按9000个细胞/50μL/孔进行铺板。
f.100μL PBS封周围孔。
g.将细胞培养板放在37℃,5%二氧化碳的培养箱中培养过夜。
2.2加药(第二天)
a.在每孔50μL的细胞上清中加入50μL的稀释好的化合物到每个细胞板中。
b.将加完药的细胞板放置在37℃,5%二氧化碳的培养箱培养24小时。
2.3免疫荧光染色及检测(第三天至第四天)
a.取出培养箱培养24小时的细胞板,弃去培养基,加入预冷甲醇室温固定10分钟。
b.弃去固定液,用洗涤缓冲液快洗3次后再慢洗3次(5分钟/次)。
c.弃去洗涤缓冲液,加入封闭缓冲液室温孵育60分钟。
d.弃去封闭缓冲液,加入配制的一抗溶液4℃过夜。
e.弃去一抗溶液,加入配制的二抗溶液室温孵育60分钟。
f.弃去二抗溶液,用洗涤缓冲液快洗3次后再慢洗5次(5分钟/次)。
g.使用
Figure PCTCN2022111395-appb-000149
Micro Confocal检测。
h.根据每个细胞的平均荧光强度的数据进行Graphpad软件作图,计算化合物IC 50值和Imax%。
表3本公开化合物对H3K23乙酰化抑制的IC 50值及最大抑制率
化合物编号 H3K23 ac/IC 50(nM) 最大抑制率/Imax%
1 1.1 101.8
2 0.4 98.5
3 0.9 106.3
4 1.5 105.5
5 0.6 104.2
7 1.4 97.4
8 2.8 99.4
9 0.75 99.5
10 1.2 106.3
11 2.1 103.1
20 4.6 99.4
22 2.9 104.8
结论:本公开化合物对H3K23乙酰化具有很好的抑制作用。
测试例4 ZR-75-1增殖实验
一.试剂与仪器
1.ZR-75-1(ATCC CRL1500)
2. 1640培养基(Gibco,22400-089)
3. 0.25%胰蛋白酶-EDTA(1x)(Gibco,25200-072)
4.青霉素-链霉素(Gibco,15140-122)
5.DPBS(1×)(Gibco,14190-144)
6.FBS(Gibco,10091148)
7. 96孔底部透明黑色检测板(Corning,3603)
8. 96孔圆底配药板(96well non-treated round bottom)(JET BIOFIL,TCP-002-096)
9.CellTiter-Glo缓冲液(Promega,G756B)
10.CellTiter-Glo底物(Promega,G755B)
11.自动化细胞计数仪(Countstar,IC1000)
12.恒温培养箱(Thermo,I160)
13.PHERAstar FS(BMG labtech,PHERAstar FS)
二.实验方法
1.细胞铺板(第0天)
a.显微镜下观察细胞状态,确保细胞的融合度在90%左右。
b.弃去细胞的上清,DPBS润洗一次,倒去DPBS。加入适量胰蛋白酶消化细胞,37℃静置5分钟。
c.用等体积含10%FBS的1640培养基终止消化,收集细胞悬液。300g,离心3分钟。用适量新鲜培养基悬浮细胞。
d.取重悬的细胞悬液进行计数。
e.用含10%FBS的1640培养基稀释细胞悬液到5×10 4/mL,50μL/孔。 ZR-75-1为2500个细胞/孔。
f.将细胞培养板放在37℃,5%二氧化碳的培养箱中培养过夜。
2.加药(第1天)
a.用DMSO将每个化合物梯度稀释成9个浓度点(起始浓度100μM,3倍稀释;不同化合物根据IC 50的不同最高浓度可作相应调整)。例如,在96孔圆底配药板中,将3μL化合物依次梯度稀释至6μL DMSO中。
b.将每个化合物各浓度点按500倍稀释到相应体积1640培液中。
c.向每个细胞板中50μL/孔的细胞上清中依次加入50μL上述稀释好的化合物溶液。
d.将加完药的细胞板放置在37℃,5%二氧化碳的培养箱培养。
3.重新消化铺板并加药(第7天)
a.加药6天后,弃去含药培养液,然后按150μL/孔加入DPBS润洗一次,立即吸去DPBS。
b.加入50μL胰蛋白酶消化细胞,37℃静置3分钟,然后按150μL/孔加入含10%FBS的1640培养基终止消化。
c.用排枪吹打混匀细胞,按照1:8的比例进行重新铺板,即吸取25μL细胞悬液到新的96孔板中(新板中提前加入25μL含10%FBS的1640培养基)。
d.按照2中a.到c.步骤进行化合物配制和加药,每孔50μL。
e.将加完药的细胞板放置在37℃,5%二氧化碳的培养箱中培养。
4.CTG检测(第14天)
a.使用前将CellTiter-Glo缓冲液和冻干的CellTiter-Glo底物放置平衡到室温,两者混合充分混匀配制成100mL的CellTiter-Glo试剂(或将已混好的CellTiter-Glo试剂从-20℃拿出平衡到室温)。
b.将待检测的板子从培养箱中取出,平衡至室温,每孔加入50μL CellTiter-Glo试剂。
c.振荡混匀2分钟使细胞充分裂解。
d.室温放置28分钟待信号稳定后在PHERAstar FS上进行检测。
表4本公开化合物对ZR-75-1增殖抑制的IC 50值及最大抑制率
化合物编号 ZR-75-1/IC 50(nM) 最大抑制率/Imax%
1 1.4 96
2 0.4 96
3 1.9 95
5 0.9 98
6 0.6 95.4
7 1.6 95.6
8 2.6 96.3
9 1.6 96.2
10 2.95 94.1
20 8.6 94.9
23 10.9 90.8
结论:本公开化合物对ZR-75-1增殖具有很好的抑制作用。
测试例5药代动力学评价
一、SD大鼠试验
1、摘要
以SD大鼠为受试动物,应用LC/MS/MS法测定了SD大鼠灌胃(i.g.)给予本公开化合物后不同时刻血浆中的药物浓度。研究本公开化合物在SD大鼠体内的药代动力学行为,评价其药动学特征。
2、试验方案
2.1试验药品
化合物2、化合物7、化合物21和化合物23。
2.2试验动物
SD大鼠16只,雌雄各半,平均分成4组,由维通利华实验动物技术有限公司提供。禁食一夜后分别灌胃给药。
2.3药物配制
分别称取一定量的受试化合物,加5%DMSO+5%吐温80+90%生理盐水,配制成0.2mg/mL无色澄明溶液。
2.4给药
给药剂量为2.0mg/kg,给药体积为10.0mL/kg。
3.操作
于给药前及给药后0.25、0.5、1.0、2.0、4.0、6.0、8.0、11.0、24.0小时,由眼眶采血0.1mL,置EDTA-K2抗凝试管中,10000rpm离心1分钟(4℃),1小时内分离血浆,-20℃保存待测。采血至离心过程在冰浴条件下操作。给药后2小时进食。
测定不同浓度的药物给药后SD大鼠血浆中的待测化合物含量:取给药后各时刻的SD大鼠血浆样品25μL,每个样品加入25μL喜树碱(化合物2的内标,100ng/mL)或50μL甲苯磺丁脲(化合物7的内标,100ng/mL)或50μL维拉帕米(化合物21,23的内标,100ng/mL),并用200μL乙腈沉淀蛋白质,涡旋混合5分钟,并在3700rpm下离心10分钟。取120μL上清液,加入30μL水,涡旋5分钟,进样5μL进行LC/MS/MS分析。
4、药代动力学参数结果
表5本公开化合物的药代动力学参数
Figure PCTCN2022111395-appb-000150
结论:本公开化合物在SD大鼠体内血药浓度高,暴露量大,清除率低,具有药代动力学优势。
二、C57小鼠试验
1、摘要
以C57小鼠为受试动物,应用LC/MS/MS法测定了C57小鼠灌胃(i.g.)/静脉注射(i.v.)给予本公开化合物后不同时刻血浆中的药物浓度。研究本公开化合物在C57小鼠体内的药代动力学行为,评价其药动学特征。
2、试验方案
2.1试验药品
化合物2和化合物3。
2.2试验动物
C57小鼠36只,雌雄各半,平均分成4组,9只一组,每组每个时间点3只小鼠,由维通利华实验动物技术有限公司提供。分别灌胃及静脉注射给药。
2.3药物配制
分别称取一定量的受试化合物,加5%DMSO+5%吐温80+90%生理盐水,配制成0.1mg/mL无色澄明溶液(灌胃给药组)和0.1mg/mL无色澄明溶液(静脉注射给药组)。
2.4给药
灌胃给药组:给药剂量为2.0mg/kg,给药体积为0.2mL/10g。
静脉注射给药组:给药剂量为1.0mg/kg,给药体积为0.1mL/10g。
3.操作
灌胃给药组:于给药前及给药后0.25、0.5、1.0、2.0、4.0、6.0、8.0、11.0、24.0小时,由采血0.1mL,置EDTA-K2抗凝试管中,10000rpm离心1分钟(4℃),1小时内分离血浆,-80℃保存待测。采血至离心过程在冰浴条件下操作。
静脉注射给药组:于给药前及给药后5分钟,0.25、0.5、1.0、2.0、4.0、8.0、11.0、24小时采血,处理同灌胃给药组。
测定不同浓度的药物给药后C57小鼠血浆中的待测化合物含量:取给药后各 时刻的C57小鼠血浆样品25μL,每个样品加入50μL双氯芬酸(化合物2的内标,100ng/mL)或25μL甲苯磺丁脲(化合物3的内标,10μg/mL,购自英国LGC公司),并用200μL乙腈沉淀蛋白质,涡旋混合5分钟,并在3700rpm下离心10分钟。取120μL上清液,加入30μL水,涡旋5分钟,进样5μL进行LC/MS/MS分析。
4、药代动力学参数结果
表6本公开化合物的药代动力学参数
Figure PCTCN2022111395-appb-000151
结论:本公开化合物在C57小鼠体内血药浓度高,暴露量高,清除率低,半衰期较长,具有药代动力学优势。

Claims (20)

  1. 一种通式(I)所示的化合物或其可药用的盐:
    Figure PCTCN2022111395-appb-100001
    其中:环A选自环烷基、杂环基、芳基和杂芳基;
    环B为环烷基或杂环基;
    L为化学键、亚烷基或杂亚烷基;其中所述的亚烷基或杂亚烷基各自独立地任选被选自羟基、卤素、烷基、卤代烷基、羟烷基、烷氧基和卤代烷氧基中的一个或多个取代基所取代;
    各个R 1、各个R 2、R 3相同或不同,且各自独立地选自氢原子、卤素、氰基、硝基、氧代基、烯基、炔基、烷基、烷氧基、环烷基、杂环基、芳基、杂芳基、-OR 5、-C(O)R 6、-C(O)OR 6、-OC(O)R 6、-NHC(O)OR 6、-NR 7R 8、-C(O)NR 7R 8、-S(O) rR 6和-S(O) rNR 7R 8;其中所述的烯基、炔基、烷基、烷氧基、环烷基、杂环基、芳基和杂芳基各自独立地任选被选自羟基、卤素、氰基、氨基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基和杂环基氧基中的一个或多个取代基所取代;
    R 4为氢原子或
    Figure PCTCN2022111395-appb-100002
    环C选自环烷基、杂环基、芳基和杂芳基;
    R 0选自氢原子、羟基、卤素、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基和环烷基;
    各个R 4a相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基、杂环基氧基和-NR 9R 10
    R 5、R 6相同或不同,且各自独立地选自氢原子、烯基、炔基、烷基、环烷基、杂环基、芳基和杂芳基;其中所述的烯基、炔基、烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被选自羟基、卤素、氰基、氨基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基和杂环基氧基中的一个或多个取 代基所取代;
    R 7、R 8、R 9、R 10相同或不同,且各自独立地选自氢原子、烷基、环烷基、杂环基、芳基和杂芳基;其中所述的烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被选自羟基、卤素、氰基、氨基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基和杂环基氧基中的一个或多个取代基所取代;
    或者,R 7和R 8与相连接的N原子一起形成一个杂环基,或者R 9和R 10与相连接的N原子一起形成一个杂环基,所述的杂环基任选被选自羟基、卤素、氰基、氨基、硝基、氧代基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基烷基、杂环基烷基、环烷基氧基和杂环基氧基中的一个或多个取代基所取代;
    p为0、1、2、3或4;
    q为0、1、2、3或4;
    m为0、1、2、3或4;
    n为0、1、2、3或4;且
    r为0、1或2。
  2. 根据权利要求1所述的通式(I)所示的化合物或其可药用的盐,其中环C为芳基或杂芳基。
  3. 根据权利要求1或2所述的通式(I)所示的化合物或其可药用的盐,其中R 3为氢原子。
  4. 根据权利利要求1至3中任一项所述的通式(I)所示的化合物或其可药用的盐,其中R 4
    Figure PCTCN2022111395-appb-100003
    环C为5至10元杂芳基,优选为5元或6元杂芳基;R 4a和n如权利要求1中所定义。
  5. 根据权利要求1至4中任一项所述的通式(I)所示的化合物或其可药用的盐,其为通式(II)所示的化合物或其可药用的盐:
    Figure PCTCN2022111395-appb-100004
    其中:
    环C为5至10元杂芳基,优选为5元或6元杂芳基;
    环A、环B、L、R 1、R 2、R 4a、p、q和n如权利要求1中所定义。
  6. 根据权利要求1至5中任一项所述的通式(I)所示的化合物或其可药用的盐,其中环A为6至10元芳基,优选选自苯基、
    Figure PCTCN2022111395-appb-100005
  7. 根据权利要求1至6中任一项所述的通式(I)所示的化合物或其可药用的盐,其中环B为4至7元杂环基。
  8. 根据权利要求1至7中任一项所述的通式(I)所示的化合物或其可药用的盐,其为通式(III)所示的化合物或其可药用的盐:
    Figure PCTCN2022111395-appb-100006
    其中:
    X选自O、CR aR b和C=O;
    各个R a、R b、R c和R d相同或不同,且各自独立地选自氢原子、羟基、卤素、氰基、氨基、烯基、炔基、烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、环烷基、杂环基、芳基、杂芳基、环烷基氧基和杂环基氧基;
    或者,R c、R d与所连碳原子一起形成一个C=O;
    s为0、1、2或3;
    L、R 1、R 4a、p和n如权利要求1中所定义。
  9. 根据权利要求8所述的通式(I)所示的化合物或其可药用的盐,其中R c和R d相同或不同,且各自独立地为氢原子或卤素;和/或X为O或CH 2;和/或s为1或2。
  10. 根据权利要求1至9中任一项所述的通式(I)所示的化合物或其可药用的盐,其中各个R 1相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6烷氧基C 1-6烷基和-C(O)OCH 3和-NR 7R 8;R 7和R 8相同或不同,且各自独立地为氢原子或C 1-6烷基;优选地,各个R 1相同或不同,且各自独立地选自氢原子、卤素、C 1-6烷基、C 1-6 卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基和-NR 7R 8;R 7和R 8相同或不同,且各自独立地为氢原子或C 1-6烷基;更优选地,各个R 1相同或不同,且各自独立地为C 1-6烷氧基。
  11. 根据权利要求1至7和10中任一项所述的通式(I)所示的化合物或其可药用的盐,其中各个R 2相同或不同,且各自独立地选自氢原子、卤素、C 1-6烷基和C 1-6烷氧基,优选为氢原子或卤素。
  12. 根据权利要求1至11中任一项所述的通式(I)所示的化合物或其可药用的盐,其中各个R 4a相同或不同,且各自独立地选自氢原子、羟基、卤素、C 1-6烷基、C 1-6羟烷基和C 1-6烷氧基,优选为氢原子。
  13. 根据权利要求1至12中任一项所述的通式(I)所示的化合物或其可药用的盐,其中L为化学键或-CH 2-,优选为化学键。
  14. 根据权利要求1至13中任一项所述的通式(I)所示的化合物或其可药用的盐,其选自如下化合物:
    Figure PCTCN2022111395-appb-100007
    Figure PCTCN2022111395-appb-100008
  15. 一种通式(IA)所示的化合物或其盐:
    Figure PCTCN2022111395-appb-100009
    其中:
    环B、R 2、R 3、R 4和q如权利要求1中所定义。
  16. 化合物或其盐,选自以下化合物或其盐:
    Figure PCTCN2022111395-appb-100010
  17. 一种制备根据权利要求1所述的通式(I)所示的化合物或其可药用的盐的方法,所述方法包括:
    Figure PCTCN2022111395-appb-100011
    通式(IA)所示的化合物或其盐与通式(IB)所示的化合物或其盐反应,得到通式(I)所示的化合物或其可药用的盐,
    其中:
    环A、环B、L、R 1至R 4、p和q如权利要求1中所定义。
  18. 一种药物组合物,所述药物组合物含有根据权利要求1至14中任一项所述的通式(I)所示的化合物或其可药用的盐,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
  19. 根据权利要求1至14中任一项所述的通式(I)所示的化合物或其可药用的盐或根据权利要求18所述的药物组合物在制备用于抑制KAT的药物中的用途;其中所述KAT优选为KAT6,更优选为KAT6A和/或KAT6B。
  20. 根据权利要求1至14中任一项所述的通式(I)所示的化合物或其可药用的盐或根据权利要求18所述的药物组合物在制备用于治疗和/或预防癌症的药物中的用途,其中所述的癌症优选选自肺癌、间皮瘤、骨癌、胰腺癌、皮肤癌、头颈癌、脑癌、黑色素瘤、肛门癌、肝癌、乳腺癌、输卵管癌、子宫内膜癌、宫颈癌、卵巢癌、阴道癌、外阴癌、霍奇金淋巴瘤、食道癌、结直肠癌、小肠癌、胃癌、甲状腺癌、甲状旁腺癌、肾上腺癌、软组织肉瘤、阴茎癌、睾丸癌、前列腺癌、白血病、B细胞淋巴瘤、膀胱癌、尿道癌、输尿管癌、肾细胞癌、肾盂癌、中枢神经系统肿瘤(CNS)、原发性CNS淋巴瘤、脊髓肿瘤、胶质细胞瘤、脑胶质瘤、垂体腺瘤和鳞状细胞癌;更优选选自乳腺癌、前列腺癌、肺癌、胰腺癌、卵巢癌、宫颈癌、子宫内膜癌、膀胱癌、脑胶质瘤、B细胞淋巴瘤、肝癌和白血病;其中所述乳腺癌优选为ER +乳腺癌或ER +/HER2 -乳腺癌;其中所述肺癌优选为非小细胞肺癌;其中所述前列腺癌优选为去势抵抗性前列腺癌。
PCT/CN2022/111395 2021-08-10 2022-08-10 磺酰胺衍生物、其制备方法及其在医药上的应用 WO2023016484A1 (zh)

Priority Applications (7)

Application Number Priority Date Filing Date Title
EP22855464.8A EP4385989A1 (en) 2021-08-10 2022-08-10 Sulfonamide derivative, preparation method therefor and medical use thereof
JP2024507166A JP2024529068A (ja) 2021-08-10 2022-08-10 スルファミド誘導体、その調製方法及びその医薬的使用
CN202280046780.6A CN117597341A (zh) 2021-08-10 2022-08-10 磺酰胺衍生物、其制备方法及其在医药上的应用
CA3228411A CA3228411A1 (en) 2021-08-10 2022-08-10 Sulfonamide derivative, preparation method therefor and medical use thereof
KR1020247007144A KR20240046530A (ko) 2021-08-10 2022-08-10 설폰아미드 유도체, 이의 제조 방법 및 이의 의학적 용도
AU2022325367A AU2022325367A1 (en) 2021-08-10 2022-08-10 Sulfonamide derivative, preparation method therefor and medical use thereof
MX2024001777A MX2024001777A (es) 2021-08-10 2022-08-10 Derivado de sulfonamida, metodo de preparacion y uso medico del mismo.

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN202110913249.1 2021-08-10
CN202110913249 2021-08-10
CN202111142090 2021-09-28
CN202111142090.4 2021-09-28
CN202111500231.5 2021-12-09
CN202111500231 2021-12-09

Publications (1)

Publication Number Publication Date
WO2023016484A1 true WO2023016484A1 (zh) 2023-02-16

Family

ID=85200573

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/111395 WO2023016484A1 (zh) 2021-08-10 2022-08-10 磺酰胺衍生物、其制备方法及其在医药上的应用

Country Status (9)

Country Link
EP (1) EP4385989A1 (zh)
JP (1) JP2024529068A (zh)
KR (1) KR20240046530A (zh)
CN (1) CN117597341A (zh)
AU (1) AU2022325367A1 (zh)
CA (1) CA3228411A1 (zh)
MX (1) MX2024001777A (zh)
TW (1) TW202321263A (zh)
WO (1) WO2023016484A1 (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023114710A1 (en) * 2021-12-13 2023-06-22 Aurigene Oncology Limited Fused benzoisoxazolyl compounds as kat6a inhibitors
US11976075B2 (en) 2022-03-28 2024-05-07 Isosterix, Inc. Inhibitors of the MYST family of lysine acetyl transferases
WO2024165035A1 (zh) * 2023-02-10 2024-08-15 江苏恒瑞医药股份有限公司 一种磺酰胺衍生物结晶形式及其制备方法
US12091406B2 (en) 2021-11-16 2024-09-17 Insilico Medicine Ip Limited Lysine acetyltransferase 6A (KAT6A) inhibitors and uses thereof

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1065267A (zh) * 1991-03-28 1992-10-14 美国辉瑞有限公司 杂环环胺衍生物
US20020028834A1 (en) * 1991-03-28 2002-03-07 Anabella Villalobos Heterocyclic-cyclic amine derivatives
CN1639146A (zh) * 2002-02-22 2005-07-13 拜尔药品公司 用于治疗过度增生疾病的稠合三环杂环
CN101287493A (zh) * 2005-08-18 2008-10-15 根马布股份公司 采用cd4结合肽和辐射的疗法
WO2016198507A1 (en) 2015-06-09 2016-12-15 Monash University Aryl sulfonohydrazides
WO2017218960A1 (en) 2016-06-17 2017-12-21 Fronthera U.S. Pharmaceuticals Llc Hemoglobin modifier compounds and uses thereof
WO2019043139A1 (en) 2017-08-31 2019-03-07 Ctxt Pty Limited FUSED [1,2,4] THIADIAZINE DERIVATIVES AS KAT INHIBITORS OF THE MYST FAMILY
WO2019108824A1 (en) 2017-11-29 2019-06-06 Epizyme, Inc. Myst family histone acetyltransferase inhibitors
WO2019243491A1 (en) 2018-06-20 2019-12-26 Ctxt Pty Limited Compounds
WO2020002587A1 (en) 2018-06-28 2020-01-02 Ctxt Pty Limited Compounds
WO2020048548A1 (zh) * 2018-09-07 2020-03-12 正大天晴药业集团股份有限公司 一种作用于crbn蛋白的三并环类化合物
WO2020069322A1 (en) 2018-09-28 2020-04-02 Praxis Precision Medicines, Inc. Ion channel modulators
WO2020216701A1 (en) 2019-04-25 2020-10-29 Bayer Aktiengesellschaft Acyl sulfonamides for treating cancer
WO2020254946A1 (en) 2019-06-18 2020-12-24 Pfizer Inc. Benzisoxazole sulfonamide derivatives
WO2020254989A1 (en) 2019-06-19 2020-12-24 Pfizer Inc. Cycloalkyl and heterocycloalkyl benzisoxazole sulfonamide derivatives

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1065267A (zh) * 1991-03-28 1992-10-14 美国辉瑞有限公司 杂环环胺衍生物
US20020028834A1 (en) * 1991-03-28 2002-03-07 Anabella Villalobos Heterocyclic-cyclic amine derivatives
CN1639146A (zh) * 2002-02-22 2005-07-13 拜尔药品公司 用于治疗过度增生疾病的稠合三环杂环
CN101287493A (zh) * 2005-08-18 2008-10-15 根马布股份公司 采用cd4结合肽和辐射的疗法
WO2016198507A1 (en) 2015-06-09 2016-12-15 Monash University Aryl sulfonohydrazides
WO2017218960A1 (en) 2016-06-17 2017-12-21 Fronthera U.S. Pharmaceuticals Llc Hemoglobin modifier compounds and uses thereof
WO2019043139A1 (en) 2017-08-31 2019-03-07 Ctxt Pty Limited FUSED [1,2,4] THIADIAZINE DERIVATIVES AS KAT INHIBITORS OF THE MYST FAMILY
WO2019108824A1 (en) 2017-11-29 2019-06-06 Epizyme, Inc. Myst family histone acetyltransferase inhibitors
WO2019243491A1 (en) 2018-06-20 2019-12-26 Ctxt Pty Limited Compounds
WO2020002587A1 (en) 2018-06-28 2020-01-02 Ctxt Pty Limited Compounds
WO2020048548A1 (zh) * 2018-09-07 2020-03-12 正大天晴药业集团股份有限公司 一种作用于crbn蛋白的三并环类化合物
WO2020069322A1 (en) 2018-09-28 2020-04-02 Praxis Precision Medicines, Inc. Ion channel modulators
WO2020216701A1 (en) 2019-04-25 2020-10-29 Bayer Aktiengesellschaft Acyl sulfonamides for treating cancer
WO2020254946A1 (en) 2019-06-18 2020-12-24 Pfizer Inc. Benzisoxazole sulfonamide derivatives
WO2020254989A1 (en) 2019-06-19 2020-12-24 Pfizer Inc. Cycloalkyl and heterocycloalkyl benzisoxazole sulfonamide derivatives

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
J. MED. CHEM., vol. 53, 2010, pages 7035 - 7047

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US12091406B2 (en) 2021-11-16 2024-09-17 Insilico Medicine Ip Limited Lysine acetyltransferase 6A (KAT6A) inhibitors and uses thereof
WO2023114710A1 (en) * 2021-12-13 2023-06-22 Aurigene Oncology Limited Fused benzoisoxazolyl compounds as kat6a inhibitors
US11976075B2 (en) 2022-03-28 2024-05-07 Isosterix, Inc. Inhibitors of the MYST family of lysine acetyl transferases
WO2024165035A1 (zh) * 2023-02-10 2024-08-15 江苏恒瑞医药股份有限公司 一种磺酰胺衍生物结晶形式及其制备方法

Also Published As

Publication number Publication date
CN117597341A (zh) 2024-02-23
EP4385989A1 (en) 2024-06-19
CA3228411A1 (en) 2023-02-16
JP2024529068A (ja) 2024-08-01
MX2024001777A (es) 2024-02-29
TW202321263A (zh) 2023-06-01
AU2022325367A1 (en) 2024-02-08
KR20240046530A (ko) 2024-04-09

Similar Documents

Publication Publication Date Title
WO2023016484A1 (zh) 磺酰胺衍生物、其制备方法及其在医药上的应用
CN111902417B (zh) 一种二芳基巨环化合物、药物组合物以及其用途
KR20180096823A (ko) 신규한 소염제
WO2020200069A1 (zh) 吡咯并杂环类衍生物、其制备方法及其在医药上的应用
WO2022105852A1 (zh) 三嗪二酮类衍生物、其制备方法及其在医药上的应用
TW201934554A (zh) 吡唑并[1,5-a][1,3,5]三<img align="absmiddle" height="18px" width="27px" file="d10999.TIF" alt="其他非圖式 d10999.TIF" img-content="tif" orientation="portrait" inline="yes"></img>-2-胺類衍生物、其製備方法及其在醫藥上的應用
WO2022247920A1 (zh) 喹啉胺类化合物、其制备方法及其在医药上的应用
CN115594695A (zh) 大环类化合物、其制备方法及其在医药上的应用
CN111094254B (zh) 杂芳基类衍生物、其制备方法及其在医药上的应用
WO2020221209A1 (zh) 一种cd73抑制剂,其制备方法和应用
WO2023066371A1 (zh) 含氮的四环化合物、其制备方法及其在医药上的应用
WO2023072297A1 (zh) 含氮的四环化合物、其制备方法及其在医药上的应用
CN115611877A (zh) 磺酰胺类化合物、其制备方法及其在医药上的应用
WO2023030335A1 (zh) 作为tyk2/jak1假激酶结构域抑制剂的化合物及合成和使用方法
TW201823231A (zh) 氮雜雙環基取代的三唑類衍生物、其製備方法及其在醫藥上的應用
CN112996783A (zh) 2-氨基嘧啶类衍生物、其制备方法及其在医药上的应用
CN112341434B (zh) PI3K/mTOR蛋白降解靶向嵌合体类化合物及其制备方法和医药用途
WO2024140754A1 (zh) 一种萘酰胺类化合物、其制备方法及其应用
WO2023011581A1 (zh) 含氮杂环化合物、其制备方法及其在医药上的应用
WO2022161447A1 (zh) 二甲酰胺类化合物、其制备方法及其在医药上的应用
WO2023186058A1 (zh) 吲唑类化合物、其制备方法及其在医药上的应用
WO2024104398A1 (zh) 吡啶氮氧化物类衍生物及其药物组合物、制备方法和用途
WO2022135360A1 (zh) 嘌呤酮衍生物、其制备方法及其在医药上的应用
TW202333698A (zh) 喹啉胺類化合物、其製備方法及其在醫藥上的應用
CN118344364A (zh) 氨基取代的稠合杂芳基类化合物、其制备方法及其在医药上的应用

Legal Events

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

Ref document number: 22855464

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202280046780.6

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2022325367

Country of ref document: AU

Ref document number: AU2022325367

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2401000743

Country of ref document: TH

WWE Wipo information: entry into national phase

Ref document number: 2024507166

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 3228411

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2022325367

Country of ref document: AU

Date of ref document: 20220810

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112024002510

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 202490333

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 20247007144

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 11202400353T

Country of ref document: SG

WWE Wipo information: entry into national phase

Ref document number: 202427016473

Country of ref document: IN

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022855464

Country of ref document: EP

Effective date: 20240311

ENP Entry into the national phase

Ref document number: 112024002510

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20240207