WO2019154294A1 - 吡唑并[1,5-a][1,3,5]三嗪-2-胺类衍生物、其制备方法及其在医药上的应用 - Google Patents

吡唑并[1,5-a][1,3,5]三嗪-2-胺类衍生物、其制备方法及其在医药上的应用 Download PDF

Info

Publication number
WO2019154294A1
WO2019154294A1 PCT/CN2019/074324 CN2019074324W WO2019154294A1 WO 2019154294 A1 WO2019154294 A1 WO 2019154294A1 CN 2019074324 W CN2019074324 W CN 2019074324W WO 2019154294 A1 WO2019154294 A1 WO 2019154294A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound
alkyl
alkoxy
cycloalkyl
Prior art date
Application number
PCT/CN2019/074324
Other languages
English (en)
French (fr)
Inventor
陆标
桂斌
张俊珍
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 CA3089159A priority Critical patent/CA3089159A1/en
Priority to EP19752007.5A priority patent/EP3750891A4/en
Priority to JP2020564306A priority patent/JP2021512959A/ja
Priority to CN201980006595.2A priority patent/CN111511745B/zh
Priority to US16/967,370 priority patent/US20210032253A1/en
Publication of WO2019154294A1 publication Critical patent/WO2019154294A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present disclosure belongs to the field of medicine, and relates to a pyrazolo[1,5-a][1,3,5]triazin-2-amine derivative represented by the formula (I), a preparation method thereof, and the like derivatives, pharmaceutical compositions and their use as therapeutic agents, particularly as a 2a receptor antagonists of use and preparation for the treatment of a condition or disorder by inhibiting the a 2a receptor and improved in the pharmaceutical use .
  • Adenosine is a naturally occurring purine nucleoside and is an endogenous regulator of many physiological functions. It plays an important role in the regulation of cardiovascular system, central nervous system, respiratory system, kidney, fat and platelets.
  • adenosine The action of adenosine is mediated by the G-protein coupled receptor family, and at least four subtypes of adenosine receptors are currently known, classified as A 1 , A 2a , A 2b and A 3 .
  • a 1 and A 3 receptors inhibit the activity of adenylate cyclase
  • a 2a and A 2b receptors stimulate the activity of the enzyme, thereby modulating the level of cyclic AMP in the cell, through which adenosine regulates a wide range of Physiological function.
  • the A 2a receptor (A 2a R) is widely distributed in the body, and is mainly expressed in the striatum in the central nervous system, and is also expressed in tissues such as the periphery, heart, liver, lung and kidney.
  • adenosine A 2a receptor antagonists have surprising efficacy in the treatment of neurodegenerative diseases, primarily Parkinson's disease, Huntington's disease or Alzheimer's disease (Trends in Neurosci. 2006, 29(11), 647-654; Expert Opinion on Therapeutic Patents, 2007, 17, 979-991, etc.). It can also be used to treat other central nervous system (CNS) related diseases such as depression, hyperactivity syndrome, sleep disorders and anxiety (Clin. Neuropharmacol.
  • CNS central nervous system
  • adenosine A 2a receptor antagonists have therapeutic potential as neuroprotective agents (see Jenner PJ Neurol. 2000; 24 7 Supp 12: 1143-50).
  • adenosine A 2a receptors may play an important immunomodulatory role in many pathological processes such as ischemia, hypoxia, inflammation, trauma, transplantation, etc., which may be related to A 2a receptors in T cells and B cells. It is related to the high expression levels of various immune cells such as mononuclear macrophages and neutrophils.
  • the activation of A 2a receptor can promote the body to produce immune tolerance, and closely participate in the formation of "immune escape” or "immunosuppression” of tumor cells, which creates favorable conditions for the occurrence and development of tumors. Lokshin and colleagues (Cancer Res.
  • a 2a receptor antagonists can also be used in the treatment of tumors.
  • adenosine A 1 receptor in tissue, ischemia, hypoxia, in the central, circulatory, digestive system and skeletal muscle, cells in the hypoxic and hypoxic stress environment extracellular accumulation of adenosine through activation a 1 starts the corresponding membrane receptor on protection mechanisms, thus increasing cellular tolerance to hypoxic hypoxia.
  • the A 1 receptor located on immune cells promotes a cellular immune response in a hypoxic environment.
  • the A 1 receptor also reduces free fatty acids and triglycerides and is involved in the regulation of blood sugar.
  • a 3 adenosine receptor (such as Gessi S, et al., Pharmacol.Ther.117 (1), the 2008,123-140) will play a role in protecting the heart for A 3 receptors Blocking may increase the likelihood of complications caused by any pre-existing or developing ischemic heart disease, such as angina or heart failure.
  • the purpose of the present disclosure is to provide a compound of the formula (I), or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer, or In the form of a mixture thereof or a pharmaceutically acceptable salt thereof:
  • L is selected from CR 4 R 5 , O, NH or S;
  • Ring A and Ring B are the same or different and are each independently selected from the group consisting of a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group;
  • R 1 is the same or different and is each independently selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogenated alkoxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, and a hetero group.
  • Y is selected from a covalent bond or an alkylene group
  • R a is selected from a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogenated alkoxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, a heterocyclic group or a heterocyclic alkyl group.
  • R c is selected from the group consisting of a hydrogen atom, an alkyl group, a halogenated alkyl group, a hydroxyalkyl group, a cycloalkyl group, and a heterocyclic group, wherein the alkyl group, the cycloalkyl group, and the heterocyclic group are each independently optionally selected from a halogen, an alkane. Substituted by one or more substituents of a group, an alkoxy group, a halogenated alkyl group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, and a heterocyclic group;
  • R 2 is selected from the group consisting of hydrogen atom, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R 3 is selected from the group consisting of hydrogen atom, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R 4 and R 5 are the same or different and are each independently selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, a hydroxyl group, and a hydroxyalkyl group;
  • R 6 is selected from the group consisting of a hydrogen atom, a halogen, an alkoxy group, a halogenated alkoxy group, an amino group, a cycloalkyl group, a heterocyclic group, an aryl group, a heteroaryl group, and -NR 7 R 8 ;
  • R 7 and R 8 are the same or different and are each independently selected from the group consisting of a hydrogen atom, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogenated alkoxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, a heterocyclic group, an aryl group and a heteroaryl group;
  • R 7 and R 8 together with the attached nitrogen atom form a heterocyclic group, wherein the heterocyclic group optionally contains 1 to 2 identical or different selected from N, O and in addition to 1 nitrogen atom.
  • a hetero atom of S and said heterocyclic group is optionally selected from the group consisting of alkyl, alkoxy, oxo, halogen, amino, cyano, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocycle Substituted by one or more substituents in the aryl, aryl and heteroaryl;
  • R 9 is selected from the group consisting of a hydrogen atom, an alkyl group, a halogenated alkyl group, an alkoxy group, a halogenated alkoxy group, an amino group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group;
  • n 0, 1, 2, 3 or 4;
  • s 0, 1, 2 or 3;
  • n 1 or 2.
  • the compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer thereof Or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein R a is selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogenated alkoxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitrate a base, a cycloalkyl group, a heterocyclic group, a heterocyclylalkyl group, a heterocyclic oxy group, -OS(O) m R 6 , an aryl group and a heteroaryl group, wherein the alkyl group, the alkoxy group, the ring Alkyl, heterocyclyl, heterocyclylal
  • the compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer thereof Or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein R 1 is the same or different and is each independently selected from -YR a ;
  • Y is selected from a covalent bond or an alkylene group
  • R a is selected from hydrogen, halo, alkoxy, haloalkoxy, hydroxy, cyano, amino, nitro, cycloalkyl, heterocyclyl, heterocyclyloxy, - OR c , -COR 9 , -COOR 9 , -OS(O) m R 6 , aryl and heteroaryl; wherein the alkoxy, cycloalkyl, heterocyclyl, heterocyclyloxy, aryl
  • the base and heteroaryl are each independently optionally selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, heterocyclyl. Substituting one or more substituents of oxy, aryl, heteroaryl and -OS(O) m R 6 ;
  • R a is selected from a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogenated alkoxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, or a hetero group.
  • the compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer thereof Or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein R 1 is the same or different and is each independently selected from -YR a ;
  • Y is selected from a covalent bond or an alkylene group
  • R a is selected from a hydrogen atom, a halogen, -COR 9 or -COOR 9 ;
  • R a is selected from a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogenated alkoxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, or a hetero group.
  • the compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer thereof Or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein R 1 is the same or different and is each independently selected from -YR a ;
  • Y is selected from a covalent bond or an alkylene group, and the alkylene group is Wherein R e and R f are each independently hydrogen or alkyl;
  • R a is selected from a hydrogen atom, a halogen, -COR 9 or -COOR 9 ;
  • R a is selected from a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogenated alkoxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, or a hetero group.
  • the compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer thereof Or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein R 1 is the same or different and is each independently selected from -YR a ;
  • Y is selected from a covalent bond or an alkylene group, and the alkylene group is Wherein R e and R f are each independently hydrogen or alkyl;
  • R a is selected from a hydrogen atom, -COR 9 or -COOR 9 ;
  • R a is selected from a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogenated alkoxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, or a hetero group.
  • the compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer thereof Or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein R 1 is the same or different and is each independently selected from -YR a ;
  • Y is selected from a covalent bond or an alkylene group, and the alkylene group is Wherein R e and R f are each independently hydrogen or alkyl;
  • R a is selected from a hydrogen atom, -COR 9 or -COOR 9 ;
  • R a is selected from a hydrogen atom, an alkyl group, an alkoxy group, a hydroxyl group, a hydroxyalkyl group, a cycloalkyl group, a heterocyclic group, a heterocyclic alkyl group, a heterocyclic oxy group, -OR c , -COR 9 , -COOR 9 and -OS(O) m R 6 ; wherein the alkyl, alkoxy, cycloalkyl, heterocyclyl, heterocyclylalkyl and heterocyclyloxy groups are each Independently optionally substituted with one or more substituents selected from the group consisting of alkyl, alkoxy, hydroxy, hydroxyalkyl and -OS(O) m R 6 ;
  • the compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer thereof Or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein Y is a covalent bond or an alkylene group, and the alkylene group is Wherein R e and R f are each independently hydrogen or alkyl; other groups are as defined in the present disclosure.
  • the compound of the formula (IV), or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer a form, or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein for Wherein R 3a is as defined for R 3 , z is 0, 1 or 2, and other groups are as defined in the present disclosure.
  • the compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer thereof, Or a mixture thereof or a pharmaceutically acceptable salt thereof wherein the compound represented by the formula (I) is a compound represented by the formula (II):
  • R b is the same or different and is each independently selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogenated alkoxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, and a hetero group.
  • p 0, 1, 2 or 3;
  • Ring A, Ring B, L, Y, R a , R 2 , R 3 and s are as defined in the general formula (I).
  • An isomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof which is a compound of the formula (III):
  • G is selected from C, CH or N;
  • L, Y, R a , R b , R 2 , R 3 , p and s are as defined in the formula (II).
  • An isomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof which is a compound of the formula (III'):
  • G is selected from C, CH or N;
  • L, Y, R a , R 2 , R 3 and s are as defined in the formula (I).
  • An isomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof which is a compound of the formula (IV):
  • G is selected from C, CH or N;
  • R a , R b , R 2 , R 3 , p and s are as defined in the formula (I).
  • the compound of the formula (I), or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer a form, or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein said R a is selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, a halogenated alkyl group, an alkoxy group, a halogenated alkoxy group, a hydroxyl group, a hydroxyalkyl group, a heterocyclic ring.
  • the compound of the formula (I), or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer Isomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein said R a is selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, a halogenated alkyl group, an alkoxy group, a halogenated alkoxy group, a hydroxyl group, a hydroxyalkyl group, a hetero a cycloalkyl group, a heterocyclylalkyl group, a heterocyclic oxy group, -OR c , -COR 9 , -COOR 9 and -OS(O) m R 6 ; wherein said alkyl group, alkoxy group, heterocyclic group And the heterocyclylalkyl and heterocyclyloxy are each independently optionally substituted with one or more substitu
  • the compound of the formula (I), or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer Isomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein said R a is selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, a hydroxyalkyl group, a heterocyclic group, -OR c , -COR 9 , -COOR 9 and OS (O) m R 6, wherein said alkyl and heterocyclyl optionally are selected from alkyl, substituted with a hydroxy and oxo or more substituents; R c is selected from a hydrogen atom An alkyl group, a halogenated alkyl group, a hydroxyalkyl group, a cycloalkyl group and a heterocyclic group, wherein the alkyl group, the cycloalkyl group and
  • the compound of the formula (I), or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer Isomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof wherein said R b is selected from a hydrogen atom, a halogen or an alkyl group; and p is 0, 1, or 2.
  • Typical compounds of the present disclosure include, but are not limited to:
  • a tautomer a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof or a pharmaceutically acceptable salt thereof.
  • Another aspect of the present disclosure relates to a compound of the formula (IA):
  • R w is an amino protecting group, preferably a tert-butyl or tert-butoxycarbonyl group
  • R 7 is a hydrogen atom or R w ;
  • L is selected from CR 4 R 5 , O, NH or S;
  • Ring A and Ring B are the same or different and are each independently selected from the group consisting of a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group;
  • R 1 is the same or different and is each independently selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogenated alkoxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, and a hetero group.
  • Y is selected from a covalent bond or an alkylene group
  • R a is selected from a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogenated alkoxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, a heterocyclic group or a heterocyclic alkyl group.
  • R c is selected from the group consisting of a hydrogen atom, an alkyl group, a halogenated alkyl group, a hydroxyalkyl group, a cycloalkyl group, and a heterocyclic group, wherein the alkyl group, the cycloalkyl group, and the heterocyclic group are each independently optionally selected from a halogen, an alkane. Substituted by one or more substituents of a group, an alkoxy group, a halogenated alkyl group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, and a heterocyclic group;
  • R 2 is selected from the group consisting of hydrogen atom, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R 3 is selected from the group consisting of hydrogen atom, halogen, alkyl, alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl;
  • R 4 and R 5 are the same or different and are each independently selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, a hydroxyl group, and a hydroxyalkyl group;
  • R 6 is selected from the group consisting of a hydrogen atom, a halogen, an alkoxy group, a halogenated alkoxy group, an amino group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group;
  • R 9 is selected from the group consisting of a hydrogen atom, an alkyl group, a halogenated alkyl group, an alkoxy group, a halogenated alkoxy group, an amino group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group;
  • n 0, 1, 2, 3 or 4;
  • s 0, 1, 2 or 3;
  • n 1 or 2.
  • Typical compounds of the present disclosure include, but are not limited to:
  • Another aspect of the present disclosure relates to a method of preparing a compound of the formula (I), the method comprising:
  • R w is an amino protecting group, preferably a tert-butyl or tert-butoxycarbonyl group
  • R 7 is a hydrogen atom or R w ;
  • Ring A, Ring B, L, R 1 to R 3 , n and s are as defined in the general formula (I).
  • Another aspect of the present disclosure relates to a method of preparing a compound of the formula (II), the method comprising:
  • R w is an amino protecting group, preferably a tert-butyl or tert-butoxycarbonyl group
  • R 7 is a hydrogen atom or R w ;
  • Ring A, Ring B, L, Y, R a , R b , R 2 , R 3 , p and s are as defined in the formula (II).
  • Another aspect of the present disclosure relates to a method of preparing a compound of the formula (III), the method comprising:
  • R w is an amino protecting group, preferably a tert-butyl or tert-butoxycarbonyl group
  • R 7 is a hydrogen atom or R w ;
  • G is selected from C, CH or N;
  • L, Y, R a , R b , R 2 , R 3 , p and s are as defined in the formula (III).
  • Another aspect of the present disclosure relates to a method of producing a compound of the formula (III'), the method comprising:
  • the compound of the formula (IIIA') is deprotected to give an amino group protecting group to give a compound of the formula (III').
  • R w is an amino protecting group, preferably a tert-butyl or tert-butoxycarbonyl group
  • R 7 is a hydrogen atom or R w ;
  • G is selected from C, CH or N;
  • L, Y, R a , R 2 , R 3 and s are as defined in the formula (III').
  • Another aspect of the present disclosure relates to a method of preparing a compound of the formula (IV), the method comprising:
  • R w is an amino protecting group, preferably a tert-butyl or tert-butoxycarbonyl group
  • R 7 is a hydrogen atom or R w ;
  • G is selected from C, CH or N;
  • Another aspect of the present disclosure relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of the formula (I) of the present disclosure or a tautomer thereof, a mesogen, a racemic , enantiomers, diastereomers or mixtures thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the present disclosure further relates to a compound of the formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof, or Use of a pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same, in the manufacture of a medicament for inhibiting the A2a receptor.
  • the present disclosure further relates to a compound of the formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof, or pharmaceutically acceptable salt, or a pharmaceutical composition thereof for the manufacture for treating a condition or disorder by a 2a receptors to inhibition of the improved medicament.
  • the A 2a receptor inhibition by a condition ameliorated or disorder selected from cancer, depression, cognitive disorders, neurodegenerative disorders (Parkinson's disease, Huntington's disease, Alzheimer's disease Or amyotrophic lateral sclerosis, etc., attention-related disorders, extrapyramidal syndrome, abnormal dyskinesia, cirrhosis, liver fibrosis, fatty liver, cutaneous fibrosis, sleep disorders, stroke, brain injury, neuroinflammation and addiction Behavior; preferably a tumor.
  • a condition ameliorated or disorder selected from cancer, depression, cognitive disorders, neurodegenerative disorders (Parkinson's disease, Huntington's disease, Alzheimer's disease Or amyotrophic lateral sclerosis, etc., attention-related disorders, extrapyramidal syndrome, abnormal dyskinesia, cirrhosis, liver fibrosis, fatty liver, cutaneous fibrosis, sleep disorders, stroke, brain injury, neuroinflammation and addiction Behavior; preferably a tumor.
  • the present disclosure further relates to a compound of the formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof, or A pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same, for the preparation of a tumor, depression, cognitive function disorder, neurodegenerative disorder (Parkinson's disease, Huntington's disease, Alzheimer's disease or amyotrophic side) Somatorrhea, etc., attention-related disorders, extrapyramidal syndrome, abnormal dyskinesia, cirrhosis, liver fibrosis, fatty liver, cutaneous fibrosis, sleep disorders, stroke, brain injury, neuroinflammation and addictive behavior, preferably tumor Use in medicine.
  • the present disclosure further relates to a compound of the formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof, or Use of a pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same, in the manufacture of a medicament for treating a tumor.
  • the present disclosure also relates to a method of inhibiting an A2a receptor comprising administering to a subject in need thereof a therapeutically effective amount of a compound of the formula (I) or a tautomer, a mesogen thereof, a racemate thereof. , enantiomers, diastereomers or mixtures thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
  • the present disclosure also relates to a method of treating by A 2a receptor inhibition and improved condition or disorder, comprising administering to a patient in need thereof a therapeutically effective amount of a formula (I) or a compound represented tautomer, A meso form, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
  • a formula (I) or a compound represented tautomer A meso form, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
  • the present disclosure relates to the treatment of tumors, depression, cognitive function disorders, neurodegenerative disorders (Parkinson's disease, Huntington's disease, Alzheimer's disease or amyotrophic lateral sclerosis, etc.), attention-related disorders , extrapyramidal syndrome, abnormal dyskinesia, cirrhosis, liver fibrosis, fatty liver, cutaneous fibrosis, sleep disorders, stroke, brain injury, neuroinflammation and addictive behavior, preferably a method of tumor, which includes administering a desired patient An effective amount of a compound of the formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof, or A pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same.
  • a method of tumor which includes administering a desired patient
  • the present disclosure further relates to a compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer, or a mixture thereof Or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same, which is used as a medicament.
  • the present disclosure also relates to a compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer or a mixture thereof, or A pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same, for use as an A2a receptor antagonist.
  • the present disclosure also relates to a compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer or a mixture thereof, or pharmaceutically acceptable salt, or a pharmaceutical composition thereof, which by the treatment of a disorder or condition a 2a receptor inhibition ameliorated.
  • the present disclosure also relates to a compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer or a mixture thereof, or A pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same, for use in the treatment of a tumor, depression, cognitive function disorder, neurodegenerative disorder (Parkinson's disease, Huntington's disease, Alzheimer's disease or muscular atrophy) Sexual lateral sclerosis, etc., attention-related disorders, extrapyramidal syndrome, abnormal dyskinesia, cirrhosis, liver fibrosis, fatty liver, cutaneous fibrosis, sleep disorders, stroke, brain injury, neuroinflammation and addictive behavior, preferably Tumor.
  • the present disclosure further relates to a compound of the formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof, or A pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same, for use in the treatment of a tumor.
  • the tumor described in the present disclosure is selected from the group consisting of melanoma, brain tumor, esophageal cancer, gastric cancer, liver cancer, pancreatic cancer, colorectal cancer, lung cancer, kidney cancer, breast cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma.
  • sarcoma osteochondroma, osteoma, osteosarcoma, seminoma, testicular tumor, uterine cancer, head and neck cancer, multiple myeloma, malignant lymphoma, polycythemia, leukemia, thyroid neoplasms, ureteral neoplasms, bladder Cancer, gallbladder cancer, cholangiocarcinoma, chorionic epithelial cancer, and pediatric tumor; preferably lung cancer.
  • the active ingredient-containing pharmaceutical composition may be in a form suitable for oral administration, such as tablets, dragees, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or Tincture.
  • Oral compositions can be prepared according to any method known in the art for preparing pharmaceutical compositions, such compositions may contain one or more ingredients selected from the group consisting of sweeteners, flavoring agents, coloring agents, and preservatives, To provide a pleasing and tasty pharmaceutical preparation. Tablets contain the active ingredient and non-toxic pharmaceutically acceptable excipients suitable for the preparation of a tablet for admixture.
  • excipients can be inert excipients, granulating agents, disintegrating agents, binders and lubricants. These tablets may be uncoated or may be coated by masking the taste of the drug or delaying disintegration and absorption in the gastrointestinal tract, thus providing a sustained release effect over a longer period of time.
  • Oral formulations can also be provided in soft gelatine capsules in which the active ingredient is mixed with an inert solid diluent or the active ingredient in admixture with a water-soluble vehicle or an oil vehicle.
  • the aqueous suspension contains the active substance and excipients suitable for the preparation of the aqueous suspension for mixing. Such excipients are suspending, dispersing or wetting agents.
  • the 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.
  • the oil suspension can be formulated by suspending the active ingredient in vegetable oil, or mineral oil.
  • the oil suspension may contain a thickening agent.
  • the above sweeteners and flavoring agents may be added to provide a palatable preparation. These compositions can be preserved by the addition of an antioxidant.
  • the dispersible powders and granules suitable for the preparation of aqueous suspensions can be provided by the addition of water to provide the active ingredient and dispersing or wetting agents, suspending agents or one or more preservatives. Suitable dispersing or wetting agents and suspending agents can be used to illustrate the above examples. Other excipients such as sweetening, flavoring, and coloring agents may also be added. These compositions are preserved by the addition of an anti-oxidant such as ascorbic acid.
  • compositions of the present disclosure may also be in the form of an oil-in-water emulsion.
  • the oil phase can be a vegetable oil, or a mineral oil or a mixture thereof.
  • Suitable emulsifiers can be naturally occurring phospholipids, and emulsions can also contain sweeteners, flavoring agents, preservatives, and antioxidants.
  • Such formulations may also contain a demulcent, a preservative, a colorant, and an antioxidant.
  • compositions of the present disclosure may be in the form of a sterile injectable aqueous solution.
  • 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 the oily phase.
  • the injection or microemulsion is injected into the bloodstream of the patient by topical injection.
  • the solution and microemulsion are preferably administered in a manner that maintains a constant circulating concentration of the compounds of the present disclosure.
  • a continuous intravenous delivery device can be used.
  • An example of such a device is the Deltec CADD-PLUS.TM.5400 intravenous pump.
  • compositions of the present disclosure may be in the form of a sterile injectable aqueous or oily suspension for intramuscular and subcutaneous administration.
  • the suspension may be formulated according to known techniques using those suitable dispersing or wetting agents and suspending agents.
  • 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 may conveniently be employed as a solvent or suspension medium. Any blended fixed oil can be used for this purpose.
  • fatty acids can also be prepared as injections.
  • the compounds of the present disclosure can be administered in the form of a suppository 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 thus dissolves in the rectum to release the drug.
  • the dosage of the drug to be administered depends on a variety of factors including, but not limited to, the following factors: the activity of the particular compound used, the age of the patient, the weight of the patient, the health of the patient, the behavior of the patient. , the patient's diet, the time of administration, the mode of administration, the rate of excretion, the combination of drugs, etc.; in addition, the optimal treatment modality such as the mode of treatment, the daily dosage of the compound of formula (I) or the pharmaceutically acceptable salt
  • the type can be verified according to traditional treatment options.
  • alkyl refers to a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing from 1 to 20 carbon atoms, preferably an alkyl group having from 1 to 12 carbon atoms, more preferably from 1 to 6 carbons.
  • the alkyl group of the atom is a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing from 1 to 20 carbon atoms, preferably an alkyl group having from 1 to 12 carbon atoms, more preferably from 1 to 6 carbons.
  • 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-methylhexyl,
  • lower alkyl groups having from 1 to 6 carbon atoms, non-limiting examples including methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl Base, 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-dimethyl Butyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl Base, 2,3-dimethylbutyl and the like.
  • the alkyl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, preferably independently selected from a hydrogen atom, a halogen, an alkyl group, an alkane.
  • One or more substitutions of oxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl and -OS(O) m R 6 Substituted by the base.
  • alkylene refers to a saturated straight or branched aliphatic hydrocarbon radical having two residues derived from the removal of two hydrogen atoms from the same carbon atom of the parent alkane or two different carbon atoms.
  • Non-limiting examples of alkylene include, but are not limited to, 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 -) and 1,5-butylene (-CH 2 CH 2 CH 2 CH 2 CH 2 -) Wait.
  • the alkylene group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, preferably independently selected from alkyl, alkenyl, alkynyl groups. , alkoxy, alkylthio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy Substituted with one or more substituents of cycloalkylthio, heterocycloalkylthio, oxo and -OS(O) m R 6 .
  • alkoxy refers to -O-(alkyl) and -O-(unsubstituted cycloalkyl), wherein alkyl is as defined above.
  • alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy.
  • the alkoxy group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from a hydrogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group. Substituted by one or more substituents of hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl and -OS(O) m R 6 .
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 carbon atoms, preferably from 3 to 12 carbon atoms, preferably from 3 to 10 More preferably, the carbon atom contains 3 to 6 (e.g., 3, 4, 5 or 6) carbon atoms.
  • Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene
  • a polycycloalkyl group includes a spiro ring, a fused ring, and a cycloalkyl group.
  • spirocycloalkyl refers to a polycyclic group that shares a carbon atom (referred to as a spiro atom) between 5 to 20 members of a single ring, which may contain one or more double bonds, but none of the rings have a fully conjugated ⁇ electronic system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • the spirocycloalkyl group is classified into a monospirocycloalkyl group, a bispirocycloalkyl group or a polyspirocycloalkyl group, preferably a monospirocycloalkyl group and a bispirocycloalkyl group, depending on the number of common spiro atoms between the rings.
  • spirocycloalkyl groups include:
  • fused cycloalkyl refers to 5 to 20 members, and each ring in the system shares an all-carbon polycyclic group of an adjacent pair of carbon atoms with other rings in the system, wherein one or more of the rings may contain one or Multiple double bonds, but none of the rings have a fully conjugated ⁇ -electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • fused cycloalkyl groups include:
  • bridged cycloalkyl refers to an all-carbon polycyclic group of 5 to 20 members, any two rings sharing two carbon atoms which are not directly bonded, which may contain one or more double bonds, but none of the rings have complete Conjugate ⁇ -electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members. Depending on the number of constituent rings, it may be classified into a bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl group, preferably a bicyclic ring, a tricyclic ring or a tetracyclic ring, more preferably a bicyclic ring or a tricyclic ring.
  • bridged cycloalkyl groups include:
  • the cycloalkyl ring includes the above cycloalkyl (eg, monocyclic, fused, spiro, and bridged cycloalkyl) fused to an aryl, heteroaryl or heterocycloalkyl ring, wherein the parent structure is attached
  • the ring together is a cycloalkyl group, and non-limiting examples include indanyl, tetrahydronaphthyl, benzocycloheptyl, and the like; preferably phenylcyclopentyl, tetrahydronaphthyl.
  • the cycloalkyl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, which is preferably independently independently selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, Alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, heterocyclyloxy, aryl, heteroaryl and -OS(O) m R 6 Substituted by one or more substituents.
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 ring atoms wherein one or more ring atoms are selected from nitrogen, oxygen or S(O).
  • a hetero atom of m (where m is an integer of 0 to 2), but excluding the ring moiety of -OO-, -OS- or -SS-, the remaining ring atoms being carbon.
  • it contains from 3 to 12 ring atoms, wherein from 1 to 4 are heteroatoms; more preferably from 3 to 10 ring atoms, wherein 1-4 are heteroatoms; more preferably from 5 to 6 ring atoms, of which 1-3 They are heteroatoms.
  • monocyclic heterocyclic groups include pyrrolidinyl, tetrahydropyranyl, 1, 2.3.6-tetrahydropyridyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, High piperazinyl and the like.
  • Polycyclic heterocyclic groups include spiro, fused, and bridged heterocyclic groups.
  • spiroheterocyclyl refers to a polycyclic heterocyclic group in which one atom (called a spiro atom) is shared between 5 to 20 members of a single ring, wherein one or more ring atoms are selected from nitrogen, oxygen or S (O). ) m (where m is an integer 0 to 2) heteroatoms, and the remaining ring atoms are carbon. It may contain one or more double bonds, but none of the rings have a fully conjugated pi-electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members (for example, 7, 8, 9, or 10).
  • the spiroheterocyclyl group is classified into a monospiroheterocyclic group, a dispiroheterocyclic group or a polyspirocyclic group according to the number of shared spiro atoms between the ring and the ring, and is preferably a monospiroheterocyclic group and a dispiroheterocyclic group. More preferably, it is 4 yuan / 4 yuan, 4 yuan / 5 yuan, 4 yuan / 6 yuan, 5 yuan / 5 yuan or 5 yuan / 6-membered monospiroheterocyclic group.
  • Non-limiting examples of spiroheterocyclyl groups include:
  • fused heterocyclyl refers to 5 to 20 members, and each ring in the system shares an adjacent pair of atomic polycyclic heterocyclic groups with other rings in the system, and one or more rings may contain one or more Double bond, but none of the rings have a fully conjugated ⁇ -electron system in which one or more ring atoms are heteroatoms selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), and the remaining rings
  • the atom is carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members (for example, 7, 8, 9, or 10).
  • fused heterocyclic groups include:
  • bridge heterocyclyl refers to a polycyclic heterocyclic group of 5 to 14 members, any two rings sharing two atoms which are not directly bonded, which may contain one or more double bonds, but none of the rings have a total A ⁇ -electron system of a yoke in which one or more ring atoms are heteroatoms selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), the remaining ring atoms being carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members (for example, 7, 8, 9, or 10).
  • bridge heterocyclic groups include:
  • the heterocyclyl ring includes the above heterocyclic group (for example, a monocyclic ring, a fused ring, a spiro ring, and a bridged heterocyclic group) fused to an aryl group, a heteroaryl group or a cycloalkyl ring, wherein the structure is bonded to the parent structure.
  • the ring together is a heterocyclic group, non-limiting examples of which include:
  • the heterocyclic group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, which is preferably independently independently selected from a hydrogen atom, a halogen, an alkyl group, One or more of alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl and -OS(O) m R 6 Substituted by a substituent.
  • aryl refers to a 6 to 14 membered all-carbon monocyclic or fused polycyclic ring (ie, a ring that shares a pair of adjacent carbon atoms) having a conjugated ⁇ -electron system, preferably 6 to 10 members, such as benzene. Base and naphthyl.
  • the aryl ring includes the above aryl group fused to a heteroaryl group, a heterocyclic group or a cycloalkyl ring, wherein the ring bonded to the parent structure is an aryl ring, non-limiting examples of which include:
  • the aryl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, which is preferably independently independently selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, an alkane.
  • Oxyl, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, heterocyclyloxy, aryl, heteroaryl and -OS(O) m R 6 Substituted by one or more substituents.
  • heteroaryl refers to a heteroaromatic system containing from 1 to 4 heteroatoms, from 5 to 14 ring atoms, wherein the heteroatoms are selected from the group consisting of oxygen, sulfur and nitrogen.
  • the heteroaryl group is preferably 5 to 10 members, more preferably 5 or 6 members, such as furyl, thienyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, Imidazolyl, pyrazolyl, tetrazolyl, and the like.
  • the heteroaryl ring includes a heteroaryl group as defined above fused to an aryl, heterocyclic or cycloalkyl ring, wherein the ring to which the parent structure is attached is a heteroaryl ring, non-limiting examples of which include:
  • the heteroaryl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, which is preferably independently independently selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, One or more of alkoxy, haloalkyl, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl and -OS(O) m R 6 Substituted by a substituent.
  • amino protecting group is intended to keep the amino group unchanged during the reaction of other parts of the molecule, and to protect the amino group with a group which is easily removed.
  • Non-limiting examples include t-butyl, t-butoxycarbonyl, acetyl, benzyl, allyl, p-methoxybenzyl, and the like. These groups may be optionally substituted with from 1 to 3 substituents selected from halogen, alkoxy or nitro.
  • the amino protecting group is preferably a tert-butyl or a tert-butoxycarbonyl group.
  • heterocyclyloxy refers to heterocyclyl-O- wherein heterocyclyl is as defined above.
  • heterocyclylalkyl refers to an alkyl group substituted by one or more heterocyclic groups, wherein alkyl and heterocyclic groups are as defined above.
  • haloalkyl refers to an alkyl group substituted by one or more halogens, wherein alkyl is as defined above.
  • haloalkoxy means that the alkoxy group is substituted by one or more halogens, wherein alkoxy is as defined above.
  • hydroxy refers to -OH.
  • hydroxyalkyl refers to an alkyl group substituted with a hydroxy group, wherein alkyl is as defined above.
  • halogen means fluoro, chloro, bromo or iodo.
  • amino means -NH 2.
  • cyano refers to -CN.
  • nitro refers to -NO 2 .
  • heterocyclic group optionally substituted by an alkyl group means that an alkyl group may be, but not necessarily, present, and the description includes the case where the heterocyclic group is substituted with an alkyl group and the case where the heterocyclic group is not substituted with an alkyl group.
  • Substituted means that one or more hydrogen atoms in the group, preferably up to 5, more preferably 1 to 3, hydrogen atoms are independently substituted with each other by a corresponding number of substituents, wherein each substituent is independently
  • the options ie the substituents can be the same or different. It goes without saying that the substituents are only in their possible chemical positions, and those skilled in the art will be able to determine (by experiment or theory) substitutions that may or may not be possible without undue effort.
  • an amino group or a hydroxyl group having a free hydrogen may be unstable when combined with a carbon atom having an unsaturated (e.g., olefinic) bond.
  • “Pharmaceutical composition” means a mixture comprising one or more of the compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, as well as other components such as physiological/pharmaceutically acceptable carriers. And excipients.
  • the purpose of the pharmaceutical composition is to promote the administration of the organism, which facilitates the absorption of the active ingredient and thereby exerts biological activity.
  • “Pharmaceutically acceptable salt” refers to a salt of a compound of the present disclosure which is safe and effective for use in a mammal and which possesses the desired biological activity.
  • the compounds of the present disclosure may also comprise isotopic derivatives thereof.
  • isotopic derivative refers to a compound that differs in structure only in the presence of one or more isotopically enriched atoms.
  • isotopic derivative refers to a compound that differs in structure only in the presence of one or more isotopically enriched atoms.
  • having the structure of the present disclosure in place of " ⁇ " or “ ⁇ ” instead of hydrogen, or 18 F-fluorine ( 18 F isotope) instead of fluorine, or 11 C-, 13 C-, or 14 C-rich carbon set (11 C-, 13 C-, 14 C- or carbon labeled; 11 C-, 13 C-, 14 C- or isotopes) in place of carbon atoms in a compound within the scope of the present disclosure.
  • Such compounds can be used, for example, as analytical tools or probes in biological assays, or as in vivo diagnostic imaging tracers for disease, or as tracers for pharmacodynamic, pharmacokinetic or receptor studies.
  • Deuterated products generally retain activity comparable to that of non-deuterated compounds, and achieve better metabolic stability when deuterated at certain sites, resulting in certain therapeutic advantages (eg, increased in vivo half-life or reduced dosage requirements) ).
  • terapéuticaally effective amount refers to a sufficient amount of a drug or agent that is non-toxic but achieves the desired effect.
  • the determination of the effective amount will vary from person to person, depending on the age and general condition of the recipient, and also on the particular active substance, and a suitable effective amount in a case can be determined by one skilled in the art based on routine experimentation.
  • R 6 is as defined in the formula (I).
  • the present disclosure provides a novel pyrazolo[1,5-a][1,3,5]triazin-2-amine structure adenosine A 2a receptor antagonist, and found that a compound having such a structure has Strong inhibitory activity and high selectivity, and the compounds of such structures are well absorbed by the drug.
  • a method for preparing a medicinal salt comprising the steps of:
  • the compound of the formula (IA) is subjected to deprotection of an amino group under acidic conditions to give a compound of the formula (I).
  • R w is an amino protecting group, preferably a tert-butyl or tert-butoxycarbonyl group
  • R 7 is a hydrogen atom or R w ;
  • Ring A, Ring B, L, R 1 to R 3 , n and s are as defined in the general formula (I).
  • Agents providing acidic conditions include, but are not limited to, hydrogen chloride, hydrogen chloride in 1,4-dioxane solution, ammonium chloride, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid, methanesulfonic acid, nitric acid, phosphoric acid, p-benzene. Methanesulfonic acid and TMSOTf, preferably trifluoroacetic acid;
  • the above reaction is preferably carried out in a solvent including, but not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1 , 4-dioxane, ethylene glycol dimethyl ether, water or N,N-dimethylformamide and mixtures thereof.
  • a solvent including, but not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1 , 4-dioxane, ethylene glycol dimethyl ether, water or N,N-dimethylformamide and mixtures thereof.
  • a method for preparing a medicinal salt comprising the steps of:
  • R w is an amino protecting group, preferably a tert-butyl or tert-butoxycarbonyl group
  • R 7 is a hydrogen atom or R w ;
  • Ring A, Ring B, L, Y, R a , R b , R 2 , R 3 , p and s are as defined in the formula (II).
  • Agents providing acidic conditions include, but are not limited to, hydrogen chloride, hydrogen chloride in 1,4-dioxane solution, ammonium chloride, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid, methanesulfonic acid, nitric acid, phosphoric acid, p-benzene. Methanesulfonic acid and TMSOTf, preferably trifluoroacetic acid;
  • the above reaction is preferably carried out in a solvent including, but not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1 , 4-dioxane, ethylene glycol dimethyl ether, water or N,N-dimethylformamide and mixtures thereof.
  • a solvent including, but not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1 , 4-dioxane, ethylene glycol dimethyl ether, water or N,N-dimethylformamide and mixtures thereof.
  • a method for preparing a medicinal salt comprising the steps of:
  • R w is an amino protecting group, preferably a tert-butyl or tert-butoxycarbonyl group
  • R 7 is a hydrogen atom or R w ;
  • G is selected from C, CH or N;
  • L, Y, R a , R b , R 2 , R 3 , p and s are as defined in the formula (III).
  • Agents providing acidic conditions include, but are not limited to, hydrogen chloride, hydrogen chloride in 1,4-dioxane solution, ammonium chloride, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid, methanesulfonic acid, nitric acid, phosphoric acid, p-benzene. Methanesulfonic acid and TMSOTf, preferably trifluoroacetic acid;
  • the above reaction is preferably carried out in a solvent including, but not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1 , 4-dioxane, ethylene glycol dimethyl ether, water or N,N-dimethylformamide and mixtures thereof.
  • a solvent including, but not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1 , 4-dioxane, ethylene glycol dimethyl ether, water or N,N-dimethylformamide and mixtures thereof.
  • R w is an amino protecting group, preferably a tert-butyl or tert-butoxycarbonyl group
  • R 7 is a hydrogen atom or R w ;
  • G is selected from C, CH or N;
  • L, Y, R a , R 2 , R 3 and s are as defined in the formula (III').
  • Agents providing acidic conditions include, but are not limited to, hydrogen chloride, hydrogen chloride in 1,4-dioxane solution, ammonium chloride, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid, methanesulfonic acid, nitric acid, phosphoric acid, p-benzene. Methanesulfonic acid and TMSOTf, preferably trifluoroacetic acid;
  • the above reaction is preferably carried out in a solvent including, but not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1 , 4-dioxane, ethylene glycol dimethyl ether, water or N,N-dimethylformamide and mixtures thereof.
  • a solvent including, but not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1 , 4-dioxane, ethylene glycol dimethyl ether, water or N,N-dimethylformamide and mixtures thereof.
  • a method for preparing a medicinal salt comprising the steps of:
  • R w is an amino protecting group, preferably a tert-butyl or tert-butoxycarbonyl group
  • R 7 is a hydrogen atom or R w ;
  • G is selected from C, CH or N;
  • Agents providing acidic conditions include, but are not limited to, hydrogen chloride, hydrogen chloride in 1,4-dioxane solution, ammonium chloride, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid, methanesulfonic acid, nitric acid, phosphoric acid, p-benzene. Methanesulfonic acid and TMSOTf, preferably trifluoroacetic acid;
  • the above reaction is preferably carried out in a solvent including, but not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1 , 4-dioxane, ethylene glycol dimethyl ether, water or N,N-dimethylformamide and mixtures thereof.
  • a solvent including, but not limited to, acetic acid, methanol, ethanol, n-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, n-hexane, dimethyl sulfoxide, 1 , 4-dioxane, ethylene glycol dimethyl ether, water or N,N-dimethylformamide and mixtures thereof.
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) or/and mass spectrometry (MS).
  • NMR shift ( ⁇ ) is given in units of 10 -6 (ppm).
  • the NMR was measured by a Bruker AVANCE-400 nuclear magnetic apparatus, and the solvent was deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD), and the internal standard was four.
  • DMSO-d 6 dimethyl sulfoxide
  • CDCl 3 deuterated chloroform
  • CD 3 OD deuterated methanol
  • TMS Methyl silane
  • the MS was assayed using an Agilent 1200/1290 DAD-6110/6120 Quadrupole MS LC/MS (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.
  • the CombiFlash Rapid Preparer uses the Combiflash Rf200 (TELEDYNE ISCO).
  • Thin layer chromatography silica gel plate uses Yantai Yellow Sea HSGF254 or Qingdao GF254 silica gel plate.
  • the specification of silica gel plate used for thin layer chromatography (TLC) is 0.15mm ⁇ 0.2mm.
  • the specification for thin layer chromatography separation and purification is 0.4mm. ⁇ 0.5mm.
  • Silica gel column chromatography generally uses Yantai Huanghai silica gel 200-300 mesh silica gel as a carrier.
  • the known starting materials of the present disclosure may be synthesized by or according to methods known in the art, or may be purchased from ABCR GmbH & Co. KG, Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc, Dari Companies such as chemicals.
  • the reactions can all be carried out under an argon atmosphere or a nitrogen atmosphere.
  • An argon atmosphere or a nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon having a volume of about 1 L.
  • the hydrogen atmosphere means that the reaction flask is connected to a hydrogen balloon of about 1 L volume.
  • the pressurized hydrogenation reaction was carried out using a Parr Model 3916EKX hydrogenation apparatus and a clear blue QL-500 type hydrogen generator or a HC2-SS type hydrogenation apparatus.
  • the hydrogenation reaction is usually evacuated, charged with hydrogen, and operated three times.
  • the microwave reaction used a CEM Discover-S Model 908860 microwave reactor.
  • the solution means an aqueous solution.
  • reaction temperature is room temperature and is 20 ° C to 30 ° C.
  • TLC thin layer chromatography
  • the developing agent used for the reaction the column chromatography eluent system used for the purification of the compound
  • the thin layer chromatography developing solvent system including: A: Methylene chloride/methanol system, B: n-hexane/ethyl acetate system, C: petroleum ether/ethyl acetate system, D: acetone, E: dichloromethane/acetone system, F: ethyl acetate/dichloromethane system , G: ethyl acetate / dichloromethane / n-hexane, H: ethyl acetate / dichloromethane / acetone, the volume ratio of the solvent is adjusted according to the polarity of the compound, a small amount of triethylamine and acetic acid may also be added. Adjust with alkaline or acidic reagents.
  • the crude compound 1b (27.5 g, 115.91 mmol) was dissolved in 1 L of anhydrous ethanol, sodium borohydride (15.56 g, 173.87 mmol) and lithium chloride (10.50 g, 173.87 mmol) were added and heated to 50 ° C for 17 hours. . After cooling to room temperature, 400 mL of a saturated aqueous solution of ammonium chloride was added to the reaction mixture, and the reaction mixture was concentrated under reduced pressure. Water (200 mL), and ethyl acetate (300 mL ⁇ 4), and the organic phase was combined. The residue was dried over EtOAc (EtOAc)EtOAc.
  • reaction solution was cooled to room temperature, and 100 mL of a saturated sodium chloride solution was added, and the mixture was separated, and the aqueous phase was extracted with ethyl acetate (100 mL ⁇ 3). Purification with the eluent system B using a CombiFlash flash-purchase to give the title compound 16c (12.2 g, yield: 89.69%).
  • the crude compound 19c (1.0 g, 2.64 mmol) was dissolved in 30 mL of 1,4-dioxane, and 6 mL of tert-butylamine was added, and the mixture was sealed at 100 ° C for 1.5 hours. The reaction solution was cooled to room temperature, then evaporated, evaporated, evaporated
  • the compound 20 g (170 mg, 387.29 ⁇ mol) was dissolved in 20 mL of methanol, and a palladium-carbon hydrogenation catalyst (wet) (82 mg, 38.53 ⁇ mol, 5% purity) was added thereto, and the mixture was replaced with hydrogen three times, and the reaction was stirred for 1 hour. Filtration, the filtrate was concentrated under reduced pressure.
  • a palladium-carbon hydrogenation catalyst (wet) (82 mg, 38.53 ⁇ mol, 5% purity) was added thereto, and the mixture was replaced with hydrogen three times, and the reaction was stirred for 1 hour. Filtration, the filtrate was concentrated under reduced pressure.
  • the title compound 21b (3.87 g) was obtained from the title compound (3R)-tetrahydrofuran-3-ol 21a.
  • the first starting material compound 16b and the compound 21a were replaced with the compound 1-bromo-3-bromomethylbenzene ( ⁇ ( ⁇ ) Co., Ltd.) and the compound 16a, respectively, to obtain the title compound 25. (10 mg, yield: 5.2%).
  • Example 27 Using the synthetic route of Example 27, the fourth step starting material compound morpholine was replaced with the compound 2-oxa-6-azaspiro[3.3]heptane hemi-dicarboxylate (Nanjing Stone Technology Co., Ltd.). The title product was 32 (43 mg).
  • the compound 34c (160 mg, 437 ⁇ mol) was dissolved in 5 mL of trifluoroacetic acid, and the mixture was heated to 70 ° C for 16 hours. The reaction mixture was concentrated under reduced pressure. ⁇ 3) Extraction, combining the organic phases, and drying over anhydrous sodium sulfate. Filtration, the filtrate was concentrated under reduced pressure.
  • Example 2 Using the synthetic route of Example 1, the ninth starting material compound 1d was replaced with methyl 6-formyl-2-pyridinecarboxylate (Shanghai Bi De Technology Pharmaceutical Co., Ltd.) to obtain the compound 6-((2-amino-4). Methyl (5-methylfuran-2-yl)pyrazolo[1,5-a][1,3,5]triazin-8-yl)methyl)pyridinecarboxylate 38a (70 mg).
  • Test Example 1 the disclosed compounds for the adenosine A 2a receptor (adenosine A 2a receptor, A 2a R) cAMP signaling pathway, A 1 adenosine receptor (adenosine A 1 receptor, A 1 R) cAMP signaling pathway and adenosine a 3 receptor (adenosine a 3 receptor, a 3 R) cAMP signaling pathway inhibitory activity assay.
  • the following method was used to determine the inhibitory activity of the compounds of the present disclosure on the adenosine A 2a receptor cAMP signaling pathway, the adenosine A 1 receptor cAMP signaling pathway and the adenosine A 3 receptor cAMP signaling pathway.
  • the experimental method is briefly described as follows:
  • CHO-K1/A 2a R cells NM_000675.5
  • CHO-K1/A 1 R cells NM_000674.2
  • CHO-K1/A 3 R cells NM_000677.3
  • Adenosine deaminase (sigma, 10102105001)
  • CHO-K1/A 2a R cells were cultured in DMEM/F12 medium containing 10% fetal bovine serum and 800 ⁇ g/mL bleomycin. The cells were digested with cell separation buffer at the time of the experiment, and the cells were resuspended in a balanced salt buffer containing 20 mM HEPES and 0.1% bovine serum albumin, and the cell density was adjusted to 10 6 /mL. Add 5 ⁇ L of cell suspension to each well in a 384-well plate, 2.5 ⁇ L of 4 ⁇ prepared with balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 ⁇ M rolipram and 2.7 U/mL adenosine deaminase.
  • test compound Concentration of test compound was incubated for 30 minutes at room temperature. Add 2.5 ⁇ L of 4 ⁇ concentration of ethyl carbazole in a balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 ⁇ M rolipram and 2.7 U/mL adenosine deaminase per well and incubate at room temperature. 30 minutes. The final concentrations of the compounds were: 10,000, 2000, 400, 80, 16, 3.2, 0.64, 0.128, 0.0256, 0.00512, 0.001024 nM, and the final concentration of ethyl carbazole was 20 nM. Intracellular cAMP concentrations were detected using the cAMP Dynamic 2 kit.
  • cAMP-d2 and anti-cAMP-Eu-Cryptate were separately diluted 1:4 with cAMP lysis buffer. 5 ⁇ L of the diluted cAMP-d2 was added to each well, and 5 ⁇ L of the diluted anti-cAMP-Eu-cryptate was added thereto, and the mixture was incubated at room temperature for 1 hour in the dark.
  • the HTRF signal value was read using a PHERAstar multi-function microplate reader.
  • the IC 50 values of the compound inhibitory activity were calculated using Graphpad Prism software, as shown in Table 1.
  • CHO-K1/A 1 R was cultured in DMEM/F12 medium containing 10% fetal calf serum and 1 mg/mL G418.
  • the cells were digested with cell separation buffer at the time of the experiment, and then the cells were resuspended in a balanced salt buffer containing 20 mM HEPES and 0.1% bovine serum albumin, and the cell density was adjusted to 5 ⁇ 10 5 /mL.
  • the concentration of the test compound was incubated for 30 minutes at room temperature. Add 4.25 ⁇ L of 4 ⁇ concentration of forskolin and N6-ring in a balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 ⁇ M rolipram and 2.7 U/mL adenosine deaminase per well. Amyl adenosine was incubated for 30 minutes at room temperature. The final concentrations of the compounds were: 100,000, 10,000, 1000, 100, 10, 1, 0.1 and 0 nM, the final concentration of forskolin was 10 ⁇ M, and the final concentration of CPA was 10 nM. Intracellular cAMP concentrations were detected using the cAMP Dynamic 2 kit.
  • cAMP-d2 and anti-cAMP-Eu-cryptate were separately diluted with cAMP lysis buffer in a ratio of 1:4. 12.5 ⁇ L of diluted cAMP-d2 was added to each well, and 12.5 ⁇ L of the diluted anti-cAMP-Eu-cryptate was added, and the mixture was incubated at room temperature for 1 hour in the dark.
  • the HTRF signal value was read using a PHERAstar multi-function microplate reader. Calculated using Graphpad Prism software compound to inhibit the activity of IC 50 values (Table 2).
  • CHO-K1/A 3 R was cultured in DMEM/F12 medium containing 10% fetal bovine serum and 10 ⁇ g/mL puromycin.
  • the cells were digested with cell separation buffer during the experiment, and the cells were resuspended in a balanced salt buffer containing 20 mM HEPES and 0.1% bovine serum albumin, and the cell density was adjusted to 5 ⁇ 10 5 /mL.
  • the concentration of the test compound was incubated for 30 minutes at room temperature. Add 4.25 ⁇ L of 4 ⁇ concentration of forskolin and 2Cl-IB in a balanced salt buffer containing 20 mM HEPES, 0.1% bovine serum albumin, 54 ⁇ M rolipram and 2.7 U/mL adenosine deaminase per well. -MECA, incubate for 30 minutes at room temperature. The final concentrations of the compounds were: 100,000, 10,000, 1000, 100, 10, 1, 0.1 and 0 nM, the final concentration of forskolin was 10 ⁇ M, and the final concentration of 2Cl-IB-MECA was 5 nM. Intracellular cAMP concentrations were detected using the cAMP Dynamic 2 kit.
  • cAMP-d2 and anti-cAMP-Eu-cryptate were separately diluted with cAMP lysis buffer in a ratio of 1:4. 12.5 ⁇ L of diluted cAMP-d2 was added to each well, and 12.5 ⁇ L of the diluted anti-cAMP-Eu-cryptate was added, and the mixture was incubated at room temperature for 1 hour in the dark.
  • the HTRF signal value was read using a PHERAstar multi-function microplate reader.
  • the IC 50 values of the compound inhibitory activity were calculated using Graphpad Prism software, as shown in Table 3.
  • Table 1 IC 50 values of the compounds of the present disclosure for adenosine A 2a receptor cAMP signaling pathway inhibitory activity
  • the compounds of the present disclosure have significant inhibitory activity on the adenosine A 2a receptor cAMP signaling pathway.
  • the compounds of the present disclosure have a weak effect on adenosine A 1 receptor inhibitory activity, indicating that the compounds of the present disclosure have high selectivity for the A 2a receptor.
  • Test Example 2 Mouse pharmacokinetic test of the compounds of the present disclosure
  • mice Using mice as test animals, the concentration of the drug in plasma at different times after administration of the compound of Example 1, the compound of Example 18 and the compound of Example 19 by intragastric administration was determined by LC/MS/MS method. The pharmacokinetic behavior of the compounds of the present disclosure in mice was investigated and their pharmacokinetic characteristics were evaluated.
  • mice There were 27 C57 mice, female, 9 in each group, with an average of 3, purchased from Shanghai Jiesijie Experimental Animal Co., Ltd., animal production license number: SCXK (Shanghai) 2013-0006.
  • a certain amount of the drug was weighed, and 2.5% by volume of DMSO, 2.5% by volume of tween 80 and 95% physiological saline were placed in a 0.1 mg/mL colorless clear liquid.
  • mice were intragastrically administered overnight after fasting, and the dose was 2.0 mg/kg, and the administration volume was 0.2 mL/10 g.
  • Example 1 The compound of Example 1, the compound of Example 18 and the compound of Example 19 were administered by gavage, and 0.1 mL of blood was collected before administration and 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, 11.0, 24.0 hours after administration. Three animals at each time point were placed in heparinized tubes, and plasma was separated by centrifugation at 3500 rpm for 10 minutes, stored at -20 ° C, and fed 2 hours after administration.
  • the compounds of the present disclosure have better pharmacological absorption and have pharmacokinetic advantages.

Abstract

一种通式(I)所示的吡唑并[1,5-a][1,3,5]三嗪-2-胺类衍生物、其制备方法及含有该衍生物的药物组合物以及其作为治疗剂的用途,特别是作为A 2a受体拮抗剂的用途和在制备用于治疗通过对A 2a受体的抑制而改善的病况或病症的药物中的用途。

Description

吡唑并[1,5-a][1,3,5]三嗪-2-胺类衍生物、其制备方法及其在医药上的应用
本申请要求申请日为2018年2月6日的中国专利申请CN201810118455.1的优先权。本申请引用上述中国专利申请的全文。
技术领域
本公开属于医药领域,涉及一种通式(I)所示的吡唑并[1,5-a][1,3,5]三嗪-2-胺类衍生物、其制备方法、含有该衍生物的药物组合物以及其作为治疗剂的用途,特别是作为A 2a受体拮抗剂的用途和在制备用于治疗通过对A 2a受体的抑制而改善的病况或病症的药物中的用途。
背景技术
腺苷是天然存在的嘌呤核苷,是许多生理功能的内源性调节剂。在心血管***、中枢神经、呼吸***、肾脏、脂肪和血小板的功能调节中发挥重要作用。
腺苷的作用由G蛋白偶联受体家族介导,目前已知至少有四种亚型的腺苷受体,分类为A 1、A 2a、A 2b和A 3。其中A 1和A 3受体抑制腺苷酸环化酶的活性,而A 2a和A 2b受体刺激该酶的活性,由此调节细胞中环AMP水平,通过这些受体,腺苷调节广泛的生理功能。
A 2a受体(A 2aR)在机体分布较为广泛,在中枢神经***主要表达于纹状体,在外周、心、肝、肺、肾等组织也均有表达。数个临床前研究表明,腺苷A 2a受体拮抗剂对于治疗神经变性疾病,主要是帕金森病、亨廷顿病或阿尔茨海默病具有惊人疗效(Trends in Neurosci.2006,29(11),647-654;Expert Opinion on Therapeutic Patents,2007,17,979-991等)。而且也可以用于治疗其他中枢神经***(CNS)相关的疾病例如抑郁、多动综合征、睡眠障碍和焦虑症(Clin.Neuropharmacol.2010,33,55-60;J.Neurosci.2010,30(48),16284-16292;Parkinsonisn Relat.Disord.2010,16(6),423-426;及其中的参考文献:Mov.Disorders,2010,25(2),S305)。此外,腺苷A 2a受体拮抗剂还具有作为神经保护剂的治疗潜力(参见Jenner P.J Neurol.2000;24 7Supp12:1143-50)。
近来研究表明,在缺血低氧、炎症、创伤、移植等诸多病理过程中,腺苷A 2a受体的激活可以发挥重要的免疫调节作用,这可能与A 2a受体在T细胞、B细胞、单核巨噬细胞、中性粒细胞等多种免疫细胞上表达水平较高有关。此外,A 2a受体的活化可以促使机体产生免疫耐受,密切参与了肿瘤细胞“免疫逃逸”或“免疫抑制”的形成,为肿瘤的发生发展创造了有利条件。Lokshin及其同事(Cancer Res.2006Aug1;66(15):7758-65)证实自然 杀伤细胞上的A 2a受体活化可以通过升高cAMP,激活PKA抑制自然杀伤细胞对肿瘤细胞的杀伤。还有研究表明,激活A 2a受体可以促进黑色素瘤A375细胞、成纤维瘤NIH3T3细胞及嗜铬细胞瘤PC12细胞等肿瘤细胞的增殖,其可能与T细胞上A 2a受体的活化可以抑制T细胞活化、增殖、与肿瘤细胞的黏附及对肿瘤细胞产生细胞毒性作用相关;而A 2a受体基因敲除的小鼠则可以加强CD8 +T细胞抗肿瘤的免疫作用,显著抑制肿瘤的增殖。因此,A 2a受体拮抗剂也可用于肿瘤的治疗。
尽管对多种腺苷受体亚型具有显著生物学活性的化合物可具有治疗作用,但它们可导致不想要的副作用。例如腺苷A 1受体在组织缺血/缺氧时,在中枢、循环、消化***和骨骼肌中,细胞在处于缺氧和低氧的应激环境时,胞外聚集的腺苷通过激活胞膜上的A 1受体启动相应的保护机制,从而增加细胞对缺氧低氧的耐受。位于免疫细胞上的A 1受体在低氧环境中能促进细胞免疫应答。另外,A 1受体还能降低游离脂肪酸和甘油三酯,参与调节血糖。因此,A 1受体的持续阻断可能会引起机体组织中各种不良反应的发生(Chinese Pharmacological Bulletin,2008,24(5),573-576)。如有文献报道,在动物模型上,阻断A 1受体将会产生焦虑、觉醒等不良反应(Basic&Clinical Pharmacology&Toxicology,2011,109(3),203-7)。在心肌缺血期间,腺苷受体A 3(如Gessi S等人,Pharmacol.Ther.117(1),2008,123-140所述)会对心脏的保护发挥作用,A 3受体的持续阻断可能增加由任何预先存在的或正在发展的缺血性心脏病引起的并发症的可能性,所述缺血性心脏病诸如心绞痛或心衰。
目前,虽然已有许多化合物被开发为A 2a受体的拮抗剂用于治疗很多疾病,如WO2007116106、WO2009080197、WO2009156737、WO2011159302、WO2011095625、WO2014101373、WO2015031221中所述。
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,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐:
Figure PCTCN2019074324-appb-000001
其中:
L选自CR 4R 5、O、NH或S;
环A和环B相同或不同,且各自独立地选自环烷基、杂环基、芳基和杂芳基;
R 1相同或不同,且各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基、杂芳基和-Y-R a
Y选自共价键或亚烷基;
R a选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、-OR c、-COR 9、-COOR 9、-OS(O) mR 6、芳基和杂芳基;其中所述的烷基、烷氧基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代;
R c选自氢原子、烷基、卤代烷基、羟烷基、环烷基和杂环基,其中所述的烷基、环烷基和杂环基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基和杂环基中的一个或多个取代基所取代;
R 2选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基;
R 3选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基;
R 4和R 5相同或不同,且各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基和羟烷基;
或者R 4和R 5一起形成=NH或=O;
R 6选自氢原子、卤素、烷氧基、卤代烷氧基、氨基、环烷基、杂环基、芳基、杂芳 基和-NR 7R 8
R 7和R 8相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基;
或者R 7和R 8与相连的氮原子一起形成杂环基,其中所述的杂环基除含有1个氮原子之外,还任选含有1~2个相同或不同选自N、O和S的杂原子,并且所述的杂环基任选被选自烷基、烷氧基、氧代基、卤素、氨基、氰基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
R 9选自氢原子、烷基、卤代烷基、烷氧基、卤代烷氧基、氨基、环烷基、杂环基、芳基和杂芳基;
n为0、1、2、3或4;
s为0、1、2或3;且
m为1或2。
在本公开的一些实施方案中,所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中R a选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、-OS(O) mR 6、芳基和杂芳基,其中所述的烷基、烷氧基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代;其他基团的定义如本公开中所述。
在本公开的一些实施方案中,所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中R 1相同或不同,且各自独立地选自-Y-R a
Y选自共价键或亚烷基;
当Y为共价键时,R a选自氢原子、卤素、烷氧基、卤代烷氧基、羟基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、-OR c、-COR 9、-COOR 9、-OS(O) mR 6、芳基和杂芳基;其中所述的烷氧基、环烷基、杂环基、杂环基氧基、芳基和杂芳基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代;
当Y为亚烷基时,R a选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、-OR c、 -COR 9、-COOR 9、-OS(O) mR 6、芳基和杂芳基;其中所述的烷基、烷氧基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代;
其他基团如本公开中所定义。
在本公开的一些实施方案中,所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中R 1相同或不同,且各自独立地选自-Y-R a
Y选自共价键或亚烷基;
当Y为共价键时,R a选自氢原子、卤素、-COR 9或-COOR 9
当Y为亚烷基时,R a选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、-OR c、-COR 9、-COOR 9、-OS(O) mR 6、芳基和杂芳基;其中所述的烷基、烷氧基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代;
其他基团如本公开中所定义。
在本公开的一些实施方案中,所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中R 1相同或不同,且各自独立地选自-Y-R a
Y选自共价键或亚烷基,所述的亚烷基为
Figure PCTCN2019074324-appb-000002
其中R e和R f各自独立地为氢或烷基;
当Y为共价键时,R a选自氢原子、卤素、-COR 9或-COOR 9
当Y为亚烷基时,R a选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、-OR c、-COR 9、-COOR 9、-OS(O) mR 6、芳基和杂芳基;其中所述的烷基、烷氧基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代;
其他基团如本公开中所定义。
在本公开的一些实施方案中,所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中R 1相同或不同,且各自独立地选自-Y-R a
Y选自共价键或亚烷基,所述的亚烷基为
Figure PCTCN2019074324-appb-000003
其中R e和R f各自独立地为氢或烷基;
当Y为共价键时,R a选自氢原子、-COR 9或-COOR 9
当Y为亚烷基时,R a选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、-OR c、-COR 9、-COOR 9、-OS(O) mR 6、芳基和杂芳基;其中所述的烷基、烷氧基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代;
其他基团如本公开中所定义。
在本公开的一些实施方案中,所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中R 1相同或不同,且各自独立地选自-Y-R a
Y选自共价键或亚烷基,所述的亚烷基为
Figure PCTCN2019074324-appb-000004
其中R e和R f各自独立地为氢或烷基;
当Y为共价键时,R a选自氢原子、-COR 9或-COOR 9
当Y为亚烷基时,R a选自氢原子、烷基、烷氧基、羟基、羟烷基、环烷基、杂环基、杂环基烷基、杂环基氧基、-OR c、-COR 9、-COOR 9和-OS(O) mR 6;其中所述的烷基、烷氧基、环烷基、杂环基、杂环基烷基和杂环基氧基各自独立地任选被选自烷基、烷氧基、羟基、羟烷基和-OS(O) mR 6中的一个或多个取代基所取代;
其他基团如本公开中所定义。
在本公开的一些实施方案中,所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中Y为共价键或亚烷基,所述的亚烷基为
Figure PCTCN2019074324-appb-000005
其中R e和R f各自独立地为氢或烷基;其他基团如本公开中所定义。
在本公开的一些实施方案中,所述的通式(I)所示的化合物或其互变异构体、内消旋 体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中
Figure PCTCN2019074324-appb-000006
Figure PCTCN2019074324-appb-000007
其他基团如本公开中所定义。
在本公开的一些实施方案中,所述的通式(IV)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中
Figure PCTCN2019074324-appb-000008
Figure PCTCN2019074324-appb-000009
其中R 3a的定义如R 3,z为0、1或2,其他基团如本公开中所定义。
在本公开的一些实施方案中,所述的通式(IV)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中
Figure PCTCN2019074324-appb-000010
Figure PCTCN2019074324-appb-000011
其他基团如本公开中所定义。
在本公开的一些实施方案中,所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中-Y-R a为氟、
Figure PCTCN2019074324-appb-000012
Figure PCTCN2019074324-appb-000013
在本公开一些优选的实施方案中,通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,所述的通式(I)所示的化合物为通式(II)所示的化合物:
Figure PCTCN2019074324-appb-000014
其中:
R b相同或不同,且各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;
p为0、1、2或3;
环A、环B、L、Y、R a、R 2、R 3和s如通式(I)中所定义。
在本公开一些优选的实施方案中,所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的环A和环B相同或不同,且各自独立地为芳基或杂芳基,优选选自苯基、吡啶基、呋喃基或噻吩基。
在本公开的一些实施方案中,所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的环A为苯基或吡啶基,和/或,环B为呋喃基。
在本公开一些优选的实施方案中,所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其为通式(III)所示的化合物:
Figure PCTCN2019074324-appb-000015
其中:
G选自C、CH或N;
L、Y、R a、R b、R 2、R 3、p和s如通式(II)中所定义。
在本公开一些优选的实施方案中,所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其为通式(III’)所示的化合物:
Figure PCTCN2019074324-appb-000016
其中:
G选自C、CH或N;
L、Y、R a、R 2、R 3和s如通式(I)中所定义。
在本公开一些优选的实施方案中,所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的-Y-为共价键或-CH 2-。
在本公开一些优选的实施方案中,所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其为通式(IV)所示的化合物:
Figure PCTCN2019074324-appb-000017
其中:
G选自C、CH或N;
L、R a、R b、R 2、R 3、p和s如通式(I)中所定义。
在本公开一些优选的实施方案中,所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的L选自CR 4R 5、O、NH或S;R 4和R 5为氢原子;或者R 4和R 5一起形成=NH。
在本公开一些优选的实施方案中,所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 2选自氢原子、卤素或烷基,优选为氢原子。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 3选自氢原子、卤素或烷基,优选为氢原子或C 1-6烷基。
在本公开的一些实施方案中,所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R a选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、羟烷基、杂环基、杂环基烷基、杂环基氧基和-OS(O) mR 6;其中所述的烷基、烷氧基、杂环基、杂环基烷基和杂环基氧基各自独立地任选被选自卤素、烷基、烷氧基和环烷基中的一个或多个取代基所取代;R 6为烷基或氨基。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R a选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、羟烷基、杂环基、杂环基烷基、杂环基氧基、-OR c、-COR 9、-COOR 9和-OS(O) mR 6;其中所述的烷基、烷氧基、杂环基、杂环基烷基和杂环基氧基各自独立地任选被选自卤素、烷 基、烷氧基和环烷基中的一个或多个取代基所取代;R 6为烷基或氨基;R c选自氢原子、烷基、卤代烷基、羟烷基、环烷基和杂环基,其中所述的烷基、环烷基和杂环基各自独立地任选被选自烷氧基、羟烷基、环烷基和杂环基中的一个或多个取代基所取代;R 9为烷基。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R a选自氢原子、卤素、烷基、羟烷基、杂环基、-OR c、-COR 9、-COOR 9和OS(O) mR 6,其中所述的烷基和杂环基任选被选自烷基、羟基和氧代基中的一个或多个取代基所取代;R c选自氢原子、烷基、卤代烷基、羟烷基、环烷基和杂环基,其中所述的烷基、环烷基和杂环基各自独立地任选被选自烷基、烷氧基、羟基和环烷基中的一个或多个取代基所取代;R 6为烷基或氨基;R 9为烷基。
在本公开一个优选的实施方案中,所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R b选自氢原子、卤素或烷基;p为0、1或2。
本公开的典型化合物包括但不限于:
Figure PCTCN2019074324-appb-000018
Figure PCTCN2019074324-appb-000019
Figure PCTCN2019074324-appb-000020
Figure PCTCN2019074324-appb-000021
Figure PCTCN2019074324-appb-000022
Figure PCTCN2019074324-appb-000023
Figure PCTCN2019074324-appb-000024
Figure PCTCN2019074324-appb-000025
Figure PCTCN2019074324-appb-000026
Figure PCTCN2019074324-appb-000027
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐。
本公开的另一方面涉及通式(IA)所示的化合物:
Figure PCTCN2019074324-appb-000028
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
其中:
R w为氨基保护基,优选为叔丁基或叔丁氧羰基;
R 7为氢原子或R w
L选自CR 4R 5、O、NH或S;
环A和环B相同或不同,且各自独立地选自环烷基、杂环基、芳基和杂芳基;
R 1相同或不同,且各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基、杂芳基和Y-R a
Y选自共价键或亚烷基;
R a选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、-OR c、-COR 9、-COOR 9、-OS(O) mR 6、芳基和杂芳基;其中所述的烷基、烷氧基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代;
R c选自氢原子、烷基、卤代烷基、羟烷基、环烷基和杂环基,其中所述的烷基、环烷基和杂环基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基和杂环基中的一个或多个取代基所取代;
R 2选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝 基、环烷基、杂环基、芳基和杂芳基;
R 3选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基;
R 4和R 5相同或不同,且各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基和羟烷基;
或者R 4和R 5一起形成=NH或=O;
R 6选自氢原子、卤素、烷氧基、卤代烷氧基、氨基、环烷基、杂环基、芳基和杂芳基;
R 9选自氢原子、烷基、卤代烷基、烷氧基、卤代烷氧基、氨基、环烷基、杂环基、芳基和杂芳基;
n为0、1、2、3或4;
s为0、1、2或3;且
m为1或2。
所述的通式(IA)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中,各基团可以如通式(I)中所定义。
本公开的典型化合物包括但不限于:
Figure PCTCN2019074324-appb-000029
Figure PCTCN2019074324-appb-000030
Figure PCTCN2019074324-appb-000031
Figure PCTCN2019074324-appb-000032
Figure PCTCN2019074324-appb-000033
Figure PCTCN2019074324-appb-000034
Figure PCTCN2019074324-appb-000035
本公开的另一方面涉及一种制备通式(I)所示的化合物的方法,该方法包括:
Figure PCTCN2019074324-appb-000036
通式(IA)的化合物脱去氨基保护基,得到通式(I)的化合物,
其中:
R w为氨基保护基,优选为叔丁基或叔丁氧羰基;
R 7为氢原子或R w
环A、环B、L、R 1~R 3、n和s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)所示的化合物的方法,该方法包括:
Figure PCTCN2019074324-appb-000037
通式(IIA)的化合物脱去氨基保护基,得到通式(II)的化合物,
其中:
R w为氨基保护基,优选为叔丁基或叔丁氧羰基;
R 7为氢原子或R w
环A、环B、L、Y、R a、R b、R 2、R 3、p和s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)所示的化合物的方法,该方法包括:
Figure PCTCN2019074324-appb-000038
通式(IIIA)的化合物脱去氨基保护基,得到通式(III)的化合物,
其中:
R w为氨基保护基,优选为叔丁基或叔丁氧羰基;
R 7为氢原子或R w
G选自C、CH或N;
L、Y、R a、R b、R 2、R 3、p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(III’)所示的化合物的方法,该方法包括:
Figure PCTCN2019074324-appb-000039
通式(IIIA’)的化合物脱去氨基保护基,得到通式(III’)的化合物,
其中:
R w为氨基保护基,优选为叔丁基或叔丁氧羰基;
R 7为氢原子或R w
G选自C、CH或N;
L、Y、R a、R 2、R 3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)所示的化合物的方法,该方法包括:
Figure PCTCN2019074324-appb-000040
通式(IVA)的化合物脱去氨基保护基,得到通式(IV)的化合物,
其中:
R w为氨基保护基,优选为叔丁基或叔丁氧羰基;
R 7为氢原子或R w
G选自C、CH或N;
L、R a、R b、R 2、R 3、p和s如通式(IV)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治疗有效量的本公开通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,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)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物在制备用于抑制A 2a受体的药物中的用途。
本公开进一步涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物在制备用于治疗通过对A 2a受体抑制而改善的病况或病症的药物中的用途。
在本公开中,通过对A 2a受体抑制而改善的病况或病症选自肿瘤、抑郁症、认知功能病症、神经退行性病症(帕金森氏病、亨廷顿氏病、阿尔茨海默氏病或肌萎缩性侧索硬化等)、注意力相关病症、锥体外症候群、异常运动障碍、肝硬化、肝纤维化、脂肪肝、皮肤纤维化、睡眠障碍、中风、脑损伤、神经炎症和成瘾行为;优选为肿瘤。
本公开进一步涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物在制备***、抑郁症、认知功能病症、神经退行性病症(帕金森氏病、亨廷顿氏病、阿 尔茨海默氏病或肌萎缩性侧索硬化等)、注意力相关病症、锥体外症候群、异常运动障碍、肝硬化、肝纤维化、脂肪肝、皮肤纤维化、睡眠障碍、中风、脑损伤、神经炎症和成瘾行为,优选肿瘤的药物中的用途。
本公开进一步涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物在制备***的药物中的用途。
本公开还涉及一种抑制A 2a受体的方法,其包括给予所需患者治疗有效量的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物。
本公开还涉及一种治疗通过对A 2a受体抑制而改善的病况或病症的方法,其包括给予所需患者治疗有效量的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物。
本公开涉及一种***、抑郁症、认知功能病症、神经退行性病症(帕金森氏病、亨廷顿氏病、阿尔茨海默氏病或肌萎缩性侧索硬化等)、注意力相关病症、锥体外症候群、异常运动障碍、肝硬化、肝纤维化、脂肪肝、皮肤纤维化、睡眠障碍、中风、脑损伤、神经炎症和成瘾行为,优选肿瘤的方法,其包括给予所需患者治疗有效量的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物。
本公开进一步涉及一种通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐或包含其的药物组合物,其用作药物。
本公开还涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物,其用作A 2a受体拮抗剂。
本公开还涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物,其治疗通过对A 2a受体抑制而改善的病况或病症。
本公开还涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物,其用于***、抑郁症、认知功能病症、神经退行性病症(帕金森氏病、亨廷顿氏病、阿尔茨海默氏病或肌萎缩性侧索硬化等)、注意力相关病症、锥体外症候群、异常运动障碍、 肝硬化、肝纤维化、脂肪肝、皮肤纤维化、睡眠障碍、中风、脑损伤、神经炎症和成瘾行为,优选肿瘤。
本公开进一步涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物,其用于***。
本公开中所述的肿瘤选自黑色素瘤、脑瘤、食管癌、胃癌、肝癌、胰腺癌、结肠直肠癌、肺癌、肾癌、乳腺癌、卵巢癌、***癌、皮肤癌、神经母细胞瘤、肉瘤、骨软骨瘤、骨瘤、骨肉瘤、***瘤、睾丸肿瘤、子宫癌、头颈肿瘤、多发性骨髓瘤、恶性淋巴瘤、真性红细胞增多症、白血病、甲状腺肿瘤、输尿管肿瘤、膀胱癌、胆囊癌、胆管癌、绒毛膜上皮癌和儿科肿瘤;优选为肺癌。
含活性成分的药物组合物可以是适用于口服的形式,例如片剂、糖锭剂、锭剂、水或油混悬液、可分散粉末或颗粒、乳液、硬或软胶囊,或糖浆剂或酏剂。可按照本领域任何已知制备药用组合物的方法制备口服组合物,此类组合物可含有一种或多种选自以下的成分:甜味剂、矫味剂、着色剂和防腐剂,以提供悦目和可口的药用制剂。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。
水悬浮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。
通过加入水可使适用于制备水混悬的可分散粉末和颗粒提供活性成分和用于混合的分散剂或湿润剂、悬浮剂或一种或多种防腐剂。适宜的分散剂或湿润剂和悬浮剂可说明上述的例子。也可加入其他赋形剂例如甜味剂、矫味剂和着色剂。通过加入抗氧化剂例如抗坏血酸保存这些组合物。
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。
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、***的速率、药物的组合等;另外,最佳的治疗方式如治疗的模式、通式化合物(I)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。
发明的详细说明
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个碳原子的直链或支链基团,优选含有1至12个碳原子的烷基,更优选含有1至6个碳原子的烷基。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。更优选的是含有1至6个碳原子的低级烷基,非限制性实施例包括甲基、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、2,3-二甲基丁基等。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上被取代,所述取代基优选独立地任选选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代。
术语“亚烷基”指饱和的直链或支链脂肪族烃基,其具有2个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,其为包含1至20个碳原子的直链或支链基团,优选含有1至12个碳原子,更优选含有1至6个碳原子的亚烷基。亚烷基的非限制性实例包括但不限于亚甲基(-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-)和1,5-亚丁基(-CH 2CH 2CH 2CH 2CH 2-)等。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上被取代,所述取代基优选独立地任选选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基和-OS(O) mR 6中的一个或多个取代基所取代。
术语“烷氧基”指-O-(烷基)和-O-(非取代的环烷基),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、丁氧基、环丙氧基、环丁氧基、环戊氧基、环己氧基。烷氧基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代。
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个碳原子,优选包含3至12个碳原子,优选包含3至10个碳原子,更优选包含3至6 (例如3、4、5或6)个碳原子。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、稠环和桥环的环烷基。
术语“螺环烷基”指5至20元的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,但没有一个环具有完全共轭的π电子***。优选为6至14元,更优选为7至10元。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 PCTCN2019074324-appb-000041
术语“稠环烷基”指5至20元,***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,但没有一个环具有完全共轭的π电子***。优选为6至14元,更优选为7至10元。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5元/5元或5元/6元双环烷基。稠环烷基的非限制性实例包括:
Figure PCTCN2019074324-appb-000042
术语“桥环烷基”指5至20元,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,但没有一个环具有完全共轭的π电子***。优选为6至14元,更优选为7至10元。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 PCTCN2019074324-appb-000043
所述环烷基环包括上述环烷基(例如单环、稠环、螺环和桥环环烷基)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,非限制性实例包括茚满基、四氢萘基、苯并环庚烷基等;优选苯基并环戊基、四氢萘基。
环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上被取代,所述取代基优选独立地任选选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代。
术语“杂环基”指饱和或部分不饱和单环或多环环状烃取代基,其包含3至20个环原子,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,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个环原子,其中1~4个是杂原子;更优选包含3至10个环原子,其中1-4是杂原子;更优选包含5至6个环原子,其中1-3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。多环杂环基包括螺环、稠环和桥环的杂环基。
术语“螺杂环基”指5至20元的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、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子***。优选为6至14元,更优选为7至10元(例如7、8、9或10)。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 PCTCN2019074324-appb-000044
术语“稠杂环基”指5至20元,***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,但没有一个环具有完全共轭的π电子***,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元(例如7、8、9或10)。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5元/5元或5元/6元双环稠杂环基。稠杂环基的非限制性实例包括:
Figure PCTCN2019074324-appb-000045
术语“桥杂环基”指5至14元,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,但没有一个环具有完全共轭的π电子***,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元(例如7、8、9或10)。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 PCTCN2019074324-appb-000046
所述杂环基环包括上述杂环基(例如单环、稠环、螺环和桥环杂环基)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 PCTCN2019074324-appb-000047
杂环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上被取代,所述取代基优选独立地任选选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代。
术语“芳基”指具有共轭的π电子体系的6至14元全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,优选为6至10元,例如苯基和萘基。所述芳基环包括上述芳基稠合于杂芳基、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 PCTCN2019074324-appb-000048
芳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上被取代,所述取代基优选独立地任选选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代。
术语“杂芳基”指包含1至4个杂原子、5至14个环原子的杂芳族体系,其中杂原子选自氧、硫和氮。杂芳基优选为5至10元,更优选为5元或6元,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、四唑基等。所述杂芳基环包括上述杂芳基稠合于芳基、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 PCTCN2019074324-appb-000049
杂芳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 上被取代,所述取代基优选独立地任选选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代。
术语“氨基保护基”是为了使分子其它部位进行反应时氨基保持不变,用易于脱去的基团对氨基进行保护。非限制性实施例包含叔丁基、叔丁氧羰基、乙酰基、苄基、烯丙基和对甲氧苄基等。这些基团可任选地被选自卤素、烷氧基或硝基中的1-3个取代基所取代。所述氨基保护基优选为叔丁基或叔丁氧羰基。
术语“氧代基”指=O。
术语“杂环基氧基”指杂环基-O-,其中杂环基如上所定义。
术语“杂环基烷基”指烷基被一个或多个杂环基取代,其中烷基和杂环基如上所定义。
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“羟基”指-OH。
术语“羟烷基”指被羟基取代的烷基,其中烷基如上所定义。
术语“卤素”指氟、氯、溴或碘。
术语“氨基”指-NH 2
术语“氰基”指-CN。
术语“硝基”指-NO 2
“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发生,该说明包括该事件或环境发生或不发生地场合。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,优选为最多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,其中每个取代基都有独立的选项(即取代基可以相同,也可以不同)。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验或理论)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。
本公开的化合物还可包含其同位素衍生物。术语“同位素衍生物”指结构不同仅在于存在一种或多种同位素富集原子的化合物。例如,具有本公开的结构,除了用“氘”或“氚”代替氢,或者用 18F-氟标记( 18F同位素)代替氟,或者用 11C-, 13C-,或者 14C-富集的碳( 11C-, 13C-,或者 14C-碳标记; 11C-, 13C-,或者 14C-同位素)代替碳原子的化合物处于本公开的范围内。这样的化合物可用作例如生物学测定中的分析工具或探针,或者可以用作疾病的体内诊断成像示踪剂,或者作为药效学、药动学或受体研究的示踪剂。氘代物通常可以保留与未氘代的化合物相当的活性,并且当氘代在某些特定位点时可以取得更好的代谢稳定性,从而获得某些治疗优势(如体内半衰期增加或剂量需求减少)。
针对药物或药理学活性剂而言,术语“治疗有效量”是指无毒的但能达到预期效果的药物或药剂的足够用量。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。
其中:R 6如通式(I)中所定义。
本公开提供一种新型吡唑并[1,5-a][1,3,5]三嗪-2-胺类结构的腺苷A 2a受体拮抗剂,并发现具有此类结构的化合物具有强抑制活性和高选择性,并且此类结构的化合物药代吸收良好。
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)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式,或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2019074324-appb-000050
通式(IA)的化合物在酸性条件下,脱去氨基保护基,得到通式(I)的化合物,
其中:
R w为氨基保护基,优选为叔丁基或叔丁氧羰基;
R 7为氢原子或R w
环A、环B、L、R 1~R 3、n和s如通式(I)中所定义。
提供酸性的条件的试剂包括但不限于氯化氢、氯化氢的1,4-二氧六环溶液、氯化铵、三氟乙酸、甲酸、乙酸、盐酸、硫酸、甲磺酸、硝酸、磷酸、对苯甲磺酸和TMSOTf,优选为三氟乙酸;
上述反应优选在溶剂中进行,所用溶剂包括但不限于:醋酸、甲醇、乙醇、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、乙二醇二甲醚、水或N,N-二甲基甲酰胺及其混合物。
方案二
本公开通式(II)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式,或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2019074324-appb-000051
通式(IIA)的化合物在酸性条件下,脱去氨基保护基,得到通式(II)的化合物,
其中:
R w为氨基保护基,优选为叔丁基或叔丁氧羰基;
R 7为氢原子或R w
环A、环B、L、Y、R a、R b、R 2、R 3、p和s如通式(II)中所定义。
提供酸性的条件的试剂包括但不限于氯化氢、氯化氢的1,4-二氧六环溶液、氯化铵、三氟乙酸、甲酸、乙酸、盐酸、硫酸、甲磺酸、硝酸、磷酸、对苯甲磺酸和TMSOTf,优选为三氟乙酸;
上述反应优选在溶剂中进行,所用溶剂包括但不限于:醋酸、甲醇、乙醇、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、乙二醇二甲醚、水或N,N-二甲基甲酰胺及其混合物。
方案三
本公开通式(III)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式,或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2019074324-appb-000052
通式(IIIA)的化合物在酸性条件下,脱去氨基保护基,得到通式(III)的化合物,其中:
R w为氨基保护基,优选为叔丁基或叔丁氧羰基;
R 7为氢原子或R w
G选自C、CH或N;
L、Y、R a、R b、R 2、R 3、p和s如通式(III)中所定义。
提供酸性的条件的试剂包括但不限于氯化氢、氯化氢的1,4-二氧六环溶液、氯化铵、三氟乙酸、甲酸、乙酸、盐酸、硫酸、甲磺酸、硝酸、磷酸、对苯甲磺酸和TMSOTf,优选为三氟乙酸;
上述反应优选在溶剂中进行,所用溶剂包括但不限于:醋酸、甲醇、乙醇、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、乙二醇二甲醚、水或N,N-二甲基甲酰胺及其混合物。
方案四
本公开通式(III’)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式,或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2019074324-appb-000053
通式(IIIA’)的化合物在酸性条件下,脱去氨基保护基,得到通式(III’)的化合物,其中:
R w为氨基保护基,优选为叔丁基或叔丁氧羰基;
R 7为氢原子或R w
G选自C、CH或N;
L、Y、R a、R 2、R 3和s如通式(III’)中所定义。
提供酸性的条件的试剂包括但不限于氯化氢、氯化氢的1,4-二氧六环溶液、氯化铵、三氟乙酸、甲酸、乙酸、盐酸、硫酸、甲磺酸、硝酸、磷酸、对苯甲磺酸和TMSOTf,优选为三氟乙酸;
上述反应优选在溶剂中进行,所用溶剂包括但不限于:醋酸、甲醇、乙醇、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、乙二醇二甲醚、水或N,N-二甲基甲酰胺及其混合物。
方案五
本公开通式(IV)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式,或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2019074324-appb-000054
通式(IVA)的化合物在酸性条件下,脱去氨基保护基,得到通式(IV)的化合物,其中:
R w为氨基保护基,优选为叔丁基或叔丁氧羰基;
R 7为氢原子或R w
G选自C、CH或N;
L、R a、R b、R 2、R 3、p和s如通式(IV)中所定义。
提供酸性的条件的试剂包括但不限于氯化氢、氯化氢的1,4-二氧六环溶液、氯化铵、三氟乙酸、甲酸、乙酸、盐酸、硫酸、甲磺酸、硝酸、磷酸、对苯甲磺酸和TMSOTf,优选为三氟乙酸;
上述反应优选在溶剂中进行,所用溶剂包括但不限于:醋酸、甲醇、乙醇、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、乙二醇二甲醚、水或N,N-二甲基甲酰胺及其混合物。
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-400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 1200/1290DAD-6110/6120Quadrupole MS液质联用仪(生产商:Agilent,MS型号:6110/6120Quadrupole MS),waters ACQuity UPLC-QD/SQD(生产商:waters,MS型号:waters ACQuity Qda Detector/waters SQ Detector),THERMO Ultimate3000-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高压液相色谱仪。
手性HPLC分析测定使用Agilent 1260DAD高效液相色谱仪。
高效液相制备使用Waters 2545-2767、Waters 2767-SQ Detecor2、Shimadzu LC-20AP和Gilson GX-281制备型色谱仪。
手性制备使用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,韶远化学科技(Accela ChemBio Inc)、达瑞化学品等公司。
实施例中无特殊说明,反应能够均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:二氯甲烷/甲醇体系,B:正己烷/乙酸乙酯体系,C:石油醚/乙酸乙酯体系,D:丙酮,E:二氯甲烷/丙酮体系,F:乙酸乙酯/二氯甲烷体系,G:乙酸乙酯/二氯甲烷/正己烷,H:乙酸乙酯/二氯甲烷/丙酮,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
(S)-4-(5-甲基呋喃-2-基)-8-((6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)甲基)吡唑并[1,5-a][1,3,5]三嗪-2-胺1
Figure PCTCN2019074324-appb-000055
第一步
(S)-6-(((四氢呋喃-3-基)氧基)甲基)-2-吡啶甲酸甲酯1b
将(S)-6-(((四氢呋喃-3-基)氧基)甲基)-2-氰基吡啶1a(37.5g,183.6mmol,采用专利申请“WO2009156737”公开的方法制备而得)溶解于700mL甲醇中,加入碳酸铯(119.65g,367.24mmol),搅拌16小时。将反应液倒入750mL 1N盐酸中,搅拌2小时。在反应液中加入饱和碳酸氢钠水溶液至pH大于7。反应液减压浓缩,水相用乙酸乙酯(500mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品标题化合物1b(40.2g),产物不经纯化直接用于下一步反应。
MS m/z(ESI):238.5[M+1]。
第二步
(S)-(6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)甲醇1c
将粗品化合物1b(27.5g,115.91mmol)溶解于1L无水乙醇中,加入硼氢化钠(15.56g,173.87mmol)和氯化锂(10.50g,173.87mmol),加热至50℃,反应17小时。冷却至室温后,在反应液中加入400mL饱和氯化铵水溶液,反应液减压浓缩,向残余物中加入200mL水,用乙酸乙酯(300mL×4)萃取,合并有机相,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系A纯化,得到标题化合物1c(22.5g,产率:92.77%)。
MS m/z(ESI):210.4[M+1]。
第三步
(S)-6-(((四氢呋喃-3-基)氧基)甲基)吡啶甲醛1d
将化合物1c(22.5g,107.53mmol)溶解于500mL甲苯,加入活性二氧化锰(46.74g,537.66mmol),加热至95℃,搅拌41小时。将反应液冷却至室温,过滤,滤饼用1L乙酸乙酯洗涤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物1d(15.2g,产率:68.21%)。
MS m/z(ESI):208.4[M+1]。
第四步
(5-甲基呋喃-2-羰基)亚氨基二硫代碳酸二甲酯1f
将5-甲基呋喃-2-甲酰胺1e(19.4g,155.04mmol,采用公知的方法“Journal of Chemical Research,2016,40(10),594-596”制备而得)、二硫化碳(47.2g,620.18mmol,37.5 mL)、碘甲烷(70.4g,496.14mmol,33.5mL)溶解于600mL四氢呋喃中,再分批加入氢化钠(12.4g,310.09mmol,60%purity),搅拌反应30分钟后再加热到回流反应4小时。冷却至室温后,将反应液倒入水中,用乙酸乙酯(300mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用柱色谱以洗脱剂体系B纯化,得到标题化合物1f(17.79g,产率:50.04%)。
MS m/z(ESI):230.1[M+1]。
第五步
4-(5-甲基呋喃-2-基)-2-(甲硫基)吡唑并[1,5-a][1,3,5]三嗪1h
在氩气气氛下,将化合物1f(20.5g,89.40mmol),3-氨基吡唑1g(7.8g,93.87mmol,韶远科技(上海)有限公司)溶解于180mL N-甲基吡咯烷酮中,加热至100℃,反应0.5小时,再升温至185℃,反应3小时。冷却到室温,将反应液倒入2L水中,过滤,滤饼用水洗,干燥,所得粗品用硅胶柱色谱法以洗脱剂体系E纯化,得到标题化合物1h(14.01g,产率:63.63%)。
MS m/z(ESI):247.0[M+1]。
第六步
4-(5-甲基呋喃-2-基)-2-(甲基磺酰基)吡唑并[1,5-a][1,3,5]三嗪1i
将化合物1h(6.85g,27.81mmol)溶解于300mL二氯甲烷中,加入间氯过氧苯甲酸(9.60g,55.63mmol),搅拌反应2小时。反应液依次用饱和碳酸氢钠水溶液(50mL×1)和饱和氯化钠溶液(50mL×1)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品标题化合物1i(8.92g),产品不经纯化直接用于下一步反应。
MS m/z(ESI):279.1[M+1]。
第七步
N-(叔丁基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺1j
在封管中将粗品化合物1i(2.5g,6.99mmol)溶解于30mL 1,4-二氧六环中,加入6mL叔丁胺,密闭后加热至100℃,反应2小时。反应液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系E纯化,得到标题化合物1j(1.9g,产率:77.51%)。
MS m/z(ESI):272.3[M+1]。
第八步
8-溴-N-(叔丁基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺1k
将化合物1j(3g,11.06mmol)溶解于80mL二氯甲烷,冷却到0℃,缓慢加入N-溴代丁二酰亚胺(2.07g,11.61mmol),加毕升温到室温反应1小时。反应液减压浓缩,残余物 用CombiFlash快速制备仪以洗脱剂体系E纯化,得到标题化合物1k(3.14g,产率:80.96%)。
MS m/z(ESI):350.1[M+1]。
第九步
(2-(叔丁胺基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)(6-((((S)-四氢呋喃-3-基)氧基)甲基)吡啶-2-基)甲醇1l
将化合物1k(1.89g,5.40mmol)溶解于50mL四氢呋喃中,冷却到-78℃,滴加入7mL的1.6M正丁基锂,反应0.5小时,再一次性加入化合物1d(1.45g,6.99mmol),反应0.5小时。在反应液中加入30mL饱和氯化铵水溶液,升温到室温,分液,水相用乙酸乙酯(100mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系C纯化,得到标题化合物1l(930mg,产率:36.01%)。
MS m/z(ESI):479.6[M+1]。
第十步
(S)-N-(叔丁基)-4-(5-甲基呋喃-2-基)-8-((6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)甲基)吡唑并[1,5-a][1,3,5]三嗪-2-胺1m
将化合物1l(350mg,731μmol)溶解于10mL二氯甲烷中,加入三氟乙酸(2.25g,19.73mmol,1.5mL),再滴加入三乙基硅烷(2.07g,17.8mmol,3mL),搅拌反应过夜。在反应液中加入饱和碳酸氢钠水溶液至pH大于7,分液,水相用二氯甲烷(25mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层层析以展开剂体系B纯化,得到标题化合物1m(280mg,产率:82.76%)。
第十一步
(S)-4-(5-甲基呋喃-2-基)-8-((6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)甲基)吡唑并[1,5-a][1,3,5]三嗪-2-胺1
将化合物1m(500mg,1.08mmol)溶解于10mL三氟乙酸中,加热至70℃反应过夜。冷却至室温,将反应液减压浓缩,残余物中加入饱和碳酸氢钠水溶液至pH大于7,加入二氯甲烷(30mL×3)萃取,合并有机相,无水硫酸钠干燥。过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系A纯化,得到标题化合物1(317mg,产率:72.15%)。
MS m/z(ESI):407.5[M+1]
1H NMR(400MHz,CD 3OD):δ8.27(d,1H),7.91(s,1H),7.72(t,1H),7.25(d,1H),7.21(d,1H),6.45-6.46(m,1H),4.61-4.66(m,2H),4.33-4.35(m,1H),4.12(s,2H),3.89-3.93(m,2H),3.80-3.84(m,2H),2.50(s,3H),2.05-2.11(m,2H)。
实施例2
(6-((2-氨基-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)甲基)吡啶-2-基)甲醇2
Figure PCTCN2019074324-appb-000056
第一步
6-((苄基氧)甲基)2-吡啶甲醛2b
将(6-((苄氧基)甲基)吡啶-2-基)甲醇2a(23.400g,102.0614mmol,采用公知的方法“Journal of organic chemistry,1998,Vol.63(12),3884-3894”制备而得)溶于600mL甲苯中,加入二氧化锰(44.364g,510.3017mmol),加热到回流过夜。冷却到室温,过滤,滤饼用乙酸乙酯洗涤三次,合并滤液,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物2b(15g,产率:64.7%)。
第二步
(6-((苄氧基)甲基)吡啶-2-基)(2-(叔丁氨基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)甲醇2c
将化合物1k(5.000g,14.2770mmol)溶于200mL四氢呋喃中,氩气置换三次,冷却到-78℃,滴加18.3mL 1.6M正丁基锂,加毕继续搅拌30分钟,再加入化合物2b(3.244g,14.2745mmol),-78℃搅拌30分钟。加饱和氯化铵水溶液,用乙酸乙酯萃取三次(100mL×3),合并有机相,无水硫酸钠干燥,减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物2c(2.1g,产率:29.5%)。
第三步
8-((6-((苄氧基)甲基)吡啶-2-基)甲基)-N-(叔丁胺基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺2d
将化合物2c(2.100g,4.2120mmol)溶于20mL二氯甲烷中,加入三氟乙酸(4.802g,42.1143mmol,3.2013mL),再滴加入三乙基硅烷(4.897g,42.1149mmol,7.0971mL),搅拌反应24小时。加入饱和碳酸氢钠水溶液后分液,水相用二氯甲烷萃取三次(20mL×3),合并有机相,无水硫酸钠干燥,减压旋干,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物2c(1.4g,产率:68.8%)。
MS m/z(ESI):483.5[M+1]。
第四步
(6-((2-氨基-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)甲基)吡啶-2-基)甲醇2
将化合物2d(1.400g,2.9011mmol)加入15mL三氟乙酸中,70℃搅拌16小时。反应液减压浓缩,加入饱和碳酸氢钠水溶液,水相用二氯甲烷萃取(30mL×2),合并有机相,无水硫酸钠干燥,减压旋干,残余物用CombiFlash快速制备仪以洗脱剂体系A纯化,得标题化合物2(709mg,产率:72.6%)。
MS m/z(ESI):337.1[M+1]。
1H NMR(400MHz,DMSO-d 6):δ8.18(d,1H),7.98(s,1H),7.66(t,1H),7.26(d,1H),7.20(s,2H),7.04(d,1H),6.52-6.53(m,1H),5.35(t,1H),4.52(d,2H),3.97(s,2H),2.45(s,3H)。
实施例3
8-((6-((2-甲氧乙氧)甲基吡啶-2-基)甲基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺3
Figure PCTCN2019074324-appb-000057
Figure PCTCN2019074324-appb-000058
第一步
8-((6-(氯甲基)吡啶-2-基)甲基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺3a
将化合物2(709mg,2.1079mmol)溶于200mL二氯甲烷中,加入氯化亚砜(8.777g,73.77mmol,5.35mL),搅拌反应1小时。加饱和碳酸氢钠溶液调pH大于7,用二氯甲烷萃取(100mL×3),合并有机相,无水硫酸钠干燥,减压旋干,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物3a(550mg,产率:73.5%)。
第二步
8-((6-((2-甲氧乙氧)甲基吡啶-2-基)甲基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺3
将乙二醇单甲醚(19mg,250μmol)溶于5mL四氢呋喃中,加入氢化钠(17mg,425μmol),反应在常温搅拌30分钟,再加入化合物3a(50mg,141μmol),搅拌反应16小时。加水,用乙酸乙酯萃取(10mL×3),合并有机相,无水硫酸钠干燥,减压浓缩,残余物用硅胶柱色谱法以展开剂体系A纯化,得到标题化合物3(10mg,产率:30.0%)。
MS m/z(ESI):395.5[M+1]。
1H NMR(400MHz,CD 3OD)δ8.26(m,1H),7.91(s,1H),7.71-7.73(t,1H),7.37-7.39(d,1H),7.20-7.22(d,1H),6.46(m,1H),4.64(s,2H),4.11(s,2H),3.72-3.73(m,2H),3.62-3.63(m,2H),3.39(s,3H),2.50(s,3H)。
实施例4
8-(2-氟苄基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺4
Figure PCTCN2019074324-appb-000059
Figure PCTCN2019074324-appb-000060
第一步
(2-(叔丁胺基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)(2-氟苯基)甲醇4b
将化合物1k(600mg,1.71mmol)溶解于30mL四氢呋喃中,冷却到-78℃,滴加入正丁基锂(1.6M,2.2mL,3.52mmol),反应0.5小时,再一次性加入2-氟苯甲醛4a(319mg,2.57mmol),反应0.5小时。在反应液中加入20mL饱和氯化铵水溶液后升温到室温,分液,水相用乙酸乙酯(50mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物4b(344mg,产率:50.78%)。
MS m/z(ESI):396.3[M+1]。
第二步
N-(叔丁胺基)-8-(2-氟苄基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺4c
将化合物4b(344mg,870μmol)溶解于10mL二氯甲烷中,加入三氟乙酸(1.05g,9.21mmol,0.7mL),再滴加入三乙基硅烷(1.04g,8.90mmol,1.5mL),搅拌反应16小时。在反应液中加入饱和碳酸氢钠水溶液至pH大于7,分液。水相用二氯甲烷(20mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层层析以展开剂体系B纯化,得到标题化合物4c(168mg,产率:50.89%)。
MS m/z(ESI):380.1[M+1]。
第三步
8-(2-氟苄基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺4
将化合物4c(168mg,447μmol)溶解于5mL三氟乙酸中,加热至70℃反应16小时。冷却至室温,将反应液减压浓缩,残余物中加入饱和碳酸氢钠水溶液至pH大于7,加入二氯甲烷(20mL×3)萃取,合并有机相,无水硫酸钠干燥。过滤,滤液减压浓缩,残余物用柱层析色谱法以洗脱剂体系B纯化,得到标题化合物4(96mg,产率:62.87%)。
MS m/z(ESI):323.9[M+1]。
1H NMR(400MHz,CDCl 3):δ8.36(d,1H),7.86(s,1H),7.24-7.27(m,1H),7.17-7.21(m,1H),7.02-7.06(m,2H),6.36-6.37(d,1H),5.42(s,2H),4.01(s,2H),2.54(s,3H)。
实施例5
4-(5-甲基呋喃-2-基)-8-((6-(***啉基甲基)吡啶-2-基)甲基)吡唑并[1,5-a][1,3,5]三嗪-2-胺5
Figure PCTCN2019074324-appb-000061
将化合物3a(50mg,140.9μmol)溶解于30mL乙腈和四氢呋喃(V:V=5:1)的混合溶剂中,加入***啉(70mg,803.4μmol),加热至60℃反应16小时。冷却至室温,将反应液减压浓缩,残余物用甲醇打浆得到标题化合物5(54mg,产率:94.50%)。
MS m/z(ESI):406.3[M+1]。
1H NMR(400MHz,DMSO-d 6):δ8.20(d,1H),7.98(s,1H),7.64(t,1H),7.25(d,1H),7.21(s,2H),7.06(d,1H),6.53-6.55(m,1H),3.98(s,2H),3.57-3.60(m,4H),3.56(s,2H),2.43(s,3H),2.40-2.42(m,4H)。
实施例6
(R)-4-(5-甲基呋喃-2-基)-8-((6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)甲基)吡唑并[1,5-a][1,3,5]三嗪-2-胺6
Figure PCTCN2019074324-appb-000062
Figure PCTCN2019074324-appb-000063
将(3R)-四氢呋喃-3-醇(37mg,420μmol)溶于5mL四氢呋喃中,加入氢化钠(16mg,696μmol),反应在常温搅拌30分钟,再加入化合物3a(50mg,141μmol),搅拌反应16小时。加水,用乙酸乙酯萃取(10mL×3),合并有机相,无水硫酸钠干燥,减压浓缩,残余物用硅胶柱色谱法以展开剂体系A纯化,得到标题化合物6(10mg,产率:17.4%)。
MS m/z(ESI):407.5[M+1]。
1H NMR(400MHz,DMSO-d 6)δ8.20(m,1H),7.99(s,1H),7.66-7.70(t,1H),7.22-7.24(brs,3H),7.10-7.12(d,1H),6.54(m,1H),4.52(s,2H),4.26(m,1H),3.99(s,2H),3.75-3.77(m,2H),3.66-3.70(m,2H),2.46(s,3H),1.95-1.99(m,2H)。
实施例7
8-((6-((2-氧杂-6-氮杂螺[3.3]庚烷-6-基)甲基)吡啶-2-基)甲基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺7
Figure PCTCN2019074324-appb-000064
Figure PCTCN2019074324-appb-000065
将碳酸氢钠(360mg,4.28mmol)和2-氧杂-6-氮杂螺[3.3]庚烷半草酸盐7a(190mg,1.32mmol,采用公知的方法“Angewandte Chemie-International Edition,2008,47(24),4512-4515”制备而得)加入到60mL乙腈中,搅拌反应30分钟。再加入化合物3a(105mg,295.9μmol)和8mL四氢呋喃,加热至60℃反应16小时。冷却至室温,反应液减压浓缩,残余物用薄层层析以展开剂体系A纯化,得到标题化合物7(75mg,产率:60.70%)。
MS m/z(ESI):418.5[M+1]。
1H NMR(400MHz,CD 3OD):δ8.27(d,1H),7.91(s,1H),7.68(t,1H),7.17-7.21(m,2H),6.45-6.46(m,1H),4.74(s,4H),4.10(s,2H),3.72(s,2H),3.52(s,4H),2.50(s,3H)。
实施例8
4-(5-甲基呋喃-2-基)-8-((6-((4-甲基哌嗪-1-基)甲基)吡啶-2-基)甲基)吡唑并[1,5-a][1,3,5]三嗪-2-胺8
Figure PCTCN2019074324-appb-000066
将化合物3a(50mg,140.9μmol),N-甲基哌嗪(70mg,698.9μmol)溶解于30mL乙腈和四氢呋喃(V:V=5:1)的混合溶剂中,加热至60℃反应16小时。冷却到室温,将反应液减压浓缩,残余物用薄层层析以展开剂体系A纯化,得到标题化合物8(47mg,产率:79.69%)。
MS m/z(ESI):419.5[M+1]。
1H NMR(400MHz,CD 3OD):δ8.27(d,1H),7.91(s,1H),7.69(t,1H),7.32(d,1H),7.20(s,1H),6.46-6.47(m,1H),4.12(s,2H),3.69(s,2H),2.50-2.57(m,11H),2.30(s,3H)。
实施例9
4-(5-甲基呋喃-2-基)-8-((6-((四氢-1H-呋喃并[3,4-c]吡咯-5(3H)-基)甲基)吡啶-2-基)甲基)吡 唑并[1,5-a][1,3,5]三嗪-2-胺9
Figure PCTCN2019074324-appb-000067
将化合物3a(105mg,295.9μmol)溶解于55mL乙腈和四氢呋喃(V:V=10:1)的混合溶剂中,加入六氢-1H-呋喃并[3,4-c]吡咯9a(90mg,795.3μmol,采用专利申请“WO2013071697”公开的方法制备而得)和N,N-二异丙基乙胺(365mg,2.82mmol,0.5mL),加热至60℃反应16小时。冷却至室温,反应液减压浓缩,残余物用薄层层析以展开剂体系A纯化,得到标题化合物9(50mg,产率:39.15%)。
MS m/z(ESI):432.5[M+1]
1H NMR(400MHz,DMSO-d 6):δ8.20(d,1H),7.98(s,1H),7.64(t,1H),7.22-7.24(m,3H),7.05(d,1H),6.53-6.54(m,1H),3.98(s,2H),3.71-3.75(m,2H),3.64(s,2H),3.39-3.41(m,2H),2.70-2.72(m,2H),2.56-2.58(m,2H),2.46(s,3H),2.37-2.39(m,2H)。
实施例10
8-(2-氟苄基)-4-(呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺10
Figure PCTCN2019074324-appb-000068
Figure PCTCN2019074324-appb-000069
第一步
4-(呋喃-2-基)-2-(甲硫基)吡唑并[1,5-a][1,3,5]三嗪10b
在氩气气氛下,将(呋喃-2-羰基)亚氨基二硫代碳酸二甲酯10a(4.7g,21.83mmol,采用公知的方法“Synthesis,1981,7,554-557”制备而得),3-氨基吡唑(1.82g,21.90mmol)溶解于50mL N-甲基吡咯烷酮中,加热至100℃反应30分钟,再升温至185℃反应3小时。冷却到室温,将反应液倒入500mL乙酸乙酯中,有机相依次用水(50mL×4)和饱和氯化钠溶液(50mL×1)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用硅胶柱色谱以洗脱剂体系E纯化,得到标题化合物10b(2.26g,产率:44.57%)。
第二步
8-溴-4-(呋喃-2-基)-2-(甲硫基)吡唑并[1,5-a][1,3,5]三嗪10c
将化合物10b(1.78g,7.67mmol)溶解于100mL二氯甲烷中,加入N-溴代丁二酰亚胺(1.52g,8.54mmol),搅拌反应2小时。将反应液减压浓缩,残余物用硅胶柱色谱以洗脱剂体系E纯化,得到标题化合物10c(2.68g,产率:112.2%)。
第三步
8-溴-4-(呋喃-2-基)-2-(甲砜基)吡唑并[1,5-a][1,3,5]三嗪10d
将化合物10c(2.86g,9.19mmol)溶解于150mL二氯甲烷中,加入间氯过氧苯甲酸(3.15g,18.25mmol),搅拌反应2小时。加入50mL饱和碳酸氢钠水溶液后分液,有机相用水(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品标题化合物10d(4.04g),产品不经纯化直接用于下一步反应。
第四步
8-溴-N-(叔丁基)-4-(呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺10e
在封管中,将粗品化合物10d(4.04g,11.77mmol)加入到70mL二氧六环中,再加入10mL叔丁胺,密封后100℃反应3小时。冷却到室温,将反应液减压浓缩,残余物用 CombiFlash快速制备仪以洗脱剂体系E纯化,得到标题化合物10e(2.34g,产率:59.12%)。
第五步
(2-(叔丁胺基)-4-(呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)(2-氟苯基)甲醇10f
将化合物10e(800mg,2.38mmol)溶解于40mL四氢呋喃中,冷却到-78℃,滴加入正丁基锂(1.6M,2mL),反应30分钟。再加入化合物4a(443mg,3.57mmol),反应30分钟。加入30mL饱和氯化铵水溶液,升温到室温,分液,水相用乙酸乙酯(50mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物10f(168mg,产率:18.51%)。
第六步
N-(叔丁基)-8-(2-氟苄基)-4-(呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺10g
将化合物10f(168mg,440μmol)溶解于10mL二氯甲烷中,加入三氟乙酸(1.5g,13.15mmol,1mL),再滴加入三乙基硅烷(1.38g,11.86mmol,2mL),搅拌反应16小时,加入20mL饱和碳酸氢钠水溶液,分液。水相用二氯甲烷(20mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物10g(168mg,产率:18.51%)。
第七步
8-(2-氟苄基)-4-(呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺10
将化合物10g(90mg,246μmol)溶解于5mL三氟乙酸中,加热至70℃反应16小时,减压浓缩反应液,残余物中加入饱和碳酸氢钠水溶液至pH大于7,加入二氯甲烷(30mL×3)萃取,合并有机相,无水硫酸钠干燥。过滤,滤液减压浓缩,残余物用薄层色谱法以洗脱剂体系B纯化,得到标题化合物10(54mg,产率:70.88%)。
MS m/z(ESI):310.3[M+1]。
1H NMR(400MHz,CD 3OD):δ8.34(d,1H),7.97(s,1H),7.84(s,1H),7.21-7.26(m,2H),7.05-7.07(m,2H),6.79-6.81(m,1H),3.96(s,2H)。
实施例11
(6-((2-氨基-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)甲基)吡啶-2-基)甲基氨基磺酸酯11
Figure PCTCN2019074324-appb-000070
将化合物2(60mg,178μmol)溶于3mL N,N-二甲基乙酰胺中,加入1mL吡啶,再加入1mL氨基磺酰氯(41mg,354μmol)的二氯甲烷溶液,常温搅拌2小时。加入饱和氯化铵溶液,乙酸乙酯萃取(10mL×3),合并有机相,无水硫酸钠干燥,减压浓缩,残余物用硅胶柱色谱法以展开剂体系A纯化,得到标题化合物11(10mg,产率:13.5%)。
MS m/z(ESI):415.9[M+1]。
1H NMR(400MHz,DMSO-d 6)δ8.19-8.20(m,1H),8.01(s,1H),7.73-7.75(m,1H),7.70(brs,2H),7.30-7.32(m,1H),7.18-7.24(m,3H),6.55(m,1H),5.10(s,2H),4.02(s,2H),2.46(s,3H)。
实施例12
(S)-4-(5-甲基呋喃-2-基)-8-(3-(((四氢呋喃-3-基)氧基)甲基)苯氧基)吡唑并[1,5-a][1,3,5]三嗪-2-胺12
Figure PCTCN2019074324-appb-000071
Figure PCTCN2019074324-appb-000072
第一步
3-((四氢-2H-吡喃-2-基)氧基)苯甲醛12b
将3-羟基苯甲醛12a(10.5g,85.98mmol,采用公知的方法“Synthetic Communications,2008,38(15),2638-2645”制备而得)、4-甲基苯磺酸吡啶鎓(648mg,2.58mmol)溶解于100mL二氯甲烷中,加入3,4-二氢-2H-吡喃(21.70g,257.94mmol),搅拌反应48小时。停止反应,加入200mL二氯甲烷,用水洗(100mL×2),有机相减压浓缩,残余物用硅胶色谱法以洗脱剂体系B纯化,得标题化合物12b(17.7g,产率:99.8%)。
第二步
(3-((四氢-2H-吡喃-2-基)氧基)苯基)甲醇12c
将化合物12b(8.0g,38.79mmol)溶解于100mL甲醇中,0℃条件下,加入硼氢化钠(1.69g,44.61mmol),搅拌20分钟。停止反应,加入100mL水,减压浓缩,用乙酸乙酯萃取(100mL×2),合并有机相,减压浓缩,残余物用硅胶色谱法以洗脱剂体系B纯化,得标题化合物12c(6.51g,产率:80.5%)。
MS m/z(ESI):209.4[M+1]
第三步
2-(3-(溴甲基)苯氧基)四氢-2H-吡喃12d
依次加入化合物12c(1.0g,4.80mmol)、四溴化碳(1.75g,5.28mmol)、三苯基膦(1.40g,5.33mmol)和N,N-二异丙基乙胺(670mg,5.19mmol)溶解于40mL二氯甲烷中,搅拌1小时。停止反应,加入30mL水,用二氯甲烷萃取(30mL×3),合并有机相,减压浓缩,残余物用硅胶色谱法以洗脱剂体系B纯化,得标题化合物12d(957mg,产率:73.5%)。
第四步
2-(3-((((S)-四氢呋喃-3-基)氧基)甲基)苯氧基)四氢-2H-吡喃12e
将S-3-羟基四氢呋喃(373mg,4.24mmol,采用已公开的专利方法“WO2005/121111”制备而得)溶解于20mL四氢呋喃中,加入氢化钠(110mg,4.59mmol),搅拌40分钟,加入化合物12d(957mg,3.53mmol)的四氢呋喃溶液1mL,0℃条件下,搅拌17小时。 停止反应,加入50mL水,用乙酸乙酯萃取(50mL×3),合并有机相,减压浓缩,残余物用硅胶色谱法以洗脱剂体系B纯化,得标题化合物12e(788mg,产率:80.2%)。
MS m/z(ESI):279.5[M+1]。
第五步
(S)-3-(((四氢呋喃-3-基)氧基)甲基)苯酚12f
将化合物12e(788mg,2.83mmol)溶解于20mL甲醇中,加入4-甲基苯磺酸吡啶鎓(22mg,0.084mmol),搅拌65小时。停止反应,减压浓缩,残余物用硅胶色谱法以洗脱剂体系B纯化,得标题化合物12f(473mg,产率:86.0%)。
MS m/z(ESI):195.4[M+1]。
第六步
(S)-N-(叔丁基)-4-(5-甲基呋喃-2-基)-8-(3-(((四氢呋喃-3-基)氧基)甲基)苯氧基)吡唑并[1,5-a][1,3,5]三嗪-2-胺12g
依次加入化合物12f(266mg,1.37mmol)、化合物1k(240mg,0.685mmol)和碳酸铯(447mg,1.37mmol)溶解于5mL N,N-二甲基甲酰胺中,95℃条件下,搅拌65小时。停止反应,加入30mL水,用乙酸乙酯萃取(30mL×3),合并有机相,减压浓缩,残余物用硅胶色谱法以洗脱剂体系B纯化,得标题化合物12g(51mg,产率:16.1%)。
MS m/z(ESI):464.8[M+1]。
第七步
(S)-4-(5-甲基呋喃-2-基)-8-(3-(((四氢呋喃-3-基)氧基)甲基)苯氧基)吡唑并[1,5-a][1,3,5]三嗪-2-胺12
将化合物12g(51mg,0.110mmol)溶解于2mL三氟乙酸中,回流条件下,搅拌2小时。停止反应,减压浓缩,加入50mL乙酸乙酯,用饱和碳酸氢钠溶液(30mL×3)洗,合并有机相,减压浓缩,残余物用薄层色谱法以展开剂体系B纯化,得标题化合物12(21.2mg,产率:47.3%)。
MS m/z(ESI):408.5[M+1]。
1H NMR(400MHz,DMSO-d 6)δ7.91-7.90(m,1H),7.44-7.40(m,1H),7.27-7.19(m,5H),6.49-6.48(m,1H),5.47(s,1H),4.51-4.50(m,2H),4.20(s,1H),3.76-3.63(m,4H),2.43(s,3H),1.96-1.91(m,2H)。
实施例13
8-((6-(甲氧基甲基)吡啶-2-基)甲基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺13
Figure PCTCN2019074324-appb-000073
将化合物3a(30mg,84μmol)溶于5mL甲醇中,加入甲醇钠(9mg,166μmol),反应在70℃搅拌16小时。加水,用乙酸乙酯萃取(20mL×3),合并有机相,无水硫酸钠干燥,减压浓缩,残余物用硅胶柱色谱法以展开剂体系A纯化,得到标题化合物13(5mg,产率:16.9%)。
MS m/z(ESI):351.4[M+1]。
1H NMR(400MHz,CD 3OD)δ8.28(m,1H),7.91(s,1H),7.71-7.73(t,1H),7.32-7.34(d,1H),7.20-7.22(d,1H),6.46-6.47(m,1H),4.56(s,2H),4.12(s,2H),3.47(s,3H),2.50(s,3H)。
实施例14
8-((6-((2-氟乙氧基)甲基)吡啶-2-基)甲基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺14
Figure PCTCN2019074324-appb-000074
Figure PCTCN2019074324-appb-000075
将2-氟乙醇(27mg,421μmol)溶于5mL四氢呋喃中,加入氢化钠(33mg,825μmol),反应在常温搅拌30分钟,再加入化合物3a(30mg,84μmol),搅拌反应16小时。加水,用乙酸乙酯萃取(10mL×3),合并有机相,无水硫酸钠干燥,减压浓缩,残余物用硅胶柱色谱法以展开剂体系A纯化,得到标题化合物14(19mg,产率:49.7%)。
MS m/z(ESI):383.4[M+1]。
1H NMR(400MHz,CD 3OD)δ8.28(m,1H),7.91(s,1H),7.71-7.73(t,1H),7.38-7.39(d,1H),7.21-7.23(d,1H),6.5-6.47(m,1H),4.68(s,2H),4.65-4.67(m,1H),4.53-4.55(m,1H),4.12(s,2H),3.85-3.87(m,1H),3.785-3.80(m,1H),2.50(s,3H)。
实施例15
(S)-4-(5-甲基呋喃-2-基)-8-((6-(((1-甲基吡咯烷-3-基)氧杂)甲基)吡啶-2-基)甲基)吡唑并[1,5-a][1,3,5]三嗪-2-胺15
Figure PCTCN2019074324-appb-000076
将(3S)-3-羟基-1-甲基吡咯烷(42mg,415μmol)溶于5mL四氢呋喃中,加入氢化钠(16mg,695μmol),搅拌反应30分钟,再加入化合物3a(50mg,140μmol),搅拌反应16小时。加水,用乙酸乙酯萃取(10mL×3),合并有机相,无水硫酸钠干燥,减压浓缩,残余物用硅胶柱色谱法以展开剂体系A纯化,得到标题化合物15(5mg,产率:8.4%)。
MS m/z(ESI):420.5[M+1]。
1H NMR(400MHz,CD 3OD)δ8.26(m,1H),7.90(s,1H),7.70-7.73(t,1H),7.35-7.37(d,1H),7.20-7.22(d,1H),6.45(m,1H),4.58(s,2H),4.25(brs,1H),4.10(s,2H),2.75-2.85(m,3H),2.50-2.56(m,1H),2.49(s,3H),2.41(s,3H),2.17-2.23(m,1H),1.96-1.99(m,1H)。
实施例16
(S)-8-(亚氨基(6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)甲基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺16
Figure PCTCN2019074324-appb-000077
第一步
(S)-2-溴-6-(((四氢呋喃-3-基)氧基)甲基)吡啶16c
将(3S)-四氢呋喃-3-醇16a(5.06g,57.43mmol,4.6mL)溶解于200mL四氢呋喃中,加入氢化钠(2.63g,65.88mmol,60%purity),搅拌反应0.5小时。再加入2-溴-6-(氯甲基)吡啶16b(10.88g,52.70mmol,采用公知的方法“Journal of Medicinal Chemistry,2010,53(23),8421-8439”制备而得),加热到70℃反应16小时。将反应液冷却至室温,加入100mL饱和氯化钠溶液,分液,水相用乙酸乙酯(100mL×3)萃取,合并有机相,无水硫酸钠干燥, 过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物16c(12.2g,产率:89.69%)。
MS m/z(ESI):257.9[M+1]。
第二步
4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺16d
在封管中,将化合物1i(3.77g,13.54mmol)加入到50mL二氧六环中,再加入10mL氨水,密封后加热至100℃反应2小时。冷却至室温,反应液减压浓缩,得到粗品标题化合物16d(3.28g),产物不经纯化,直接用于下一步反应。
MS m/z(ESI):216.2[M+1]。
第三步
8-溴-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺16e
将粗品化合物16d(1.8g,8.36mmol)溶解于250mL二氯甲烷中,加入N-溴代丁二酰亚胺(1.65g,9.27mmol),搅拌反应1小时。反应液减压浓缩,残余物用硅胶柱色谱法以洗脱剂体系D纯化,得标题化合物16e(2.25g,产率:91.46%)。
MS m/z(ESI):294.0[M+1]。
第四步
2-氨基-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-甲腈16f
在氩气气氛下,将化合物16e(1g,3.4mmol)、氰化亚铜(1.52g,16.97mmol)、碘化亚铜(120mg,630μmol)加入到30mL二甲亚砜中,加热至165℃反应5小时。将反应液冷却至室温,加入250mL乙酸乙酯,过滤,滤饼用50mL乙酸乙酯洗涤,合并滤液,依次用水(30mL×4)和饱和氯化钠溶液(30mL×1)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得粗品标题化合物16f(600mg),产物不经纯化,直接用于下一步反应。
MS m/z(ESI):241.1[M+1]。
第五步
N-叔丁氧羰基-N-[8-氰基-4-(5-甲基-2-呋喃基)吡唑并[1,5-a][1,3,5]三嗪-2-基]氨基甲酸叔丁酯16g
将粗品化合物16f(600mg,2.50mmol)溶解于50mL四氢呋喃中,加入二碳酸二叔丁酯(2.72g,12.46mmol)和4-二甲氨基吡啶(3mg,24.3μmol),搅拌反应过夜。将反应液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物16g(220mg,产率:20.0%)。
第六步
N-叔丁氧羰基-N-[4-(5-甲基-2-呋喃基)-8-[6-[[(3S)-四氢呋喃-3-基]氧基甲基]吡啶-2-甲酰亚氨]吡唑并[1,5-a][1,3,5]三嗪-2-基]氨基甲酸叔丁酯16i
将化合物16c(176mg,681.9μmol)溶解于30mL四氢呋喃中,冷却到-78℃,滴加入正丁基锂(1.6M,0.42mL),反应0.5小时,再加入化合物16g(200mg,454.1μmol)的10mL四氢呋喃溶液,反应1小时。加入10mL饱和氯化铵水溶液,升温到室温,分液,水相用乙酸乙酯(30mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物16i(53mg,产率:18.83%)。
第七步
(S)-8-(亚氨基(6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)甲基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺16
将化合物16i(53mg,85.5μmol)溶解于2mL二氯甲烷中,加入1mL三氟乙酸,搅拌反应1小时。反应液减压浓缩,残余物中加入饱和碳酸氢钠水溶液至pH大于7,加入二氯甲烷(20mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用薄层层析以展开剂体系A纯化,得到标题化合物16(20mg,产率:55.75%)。
MS m/z(ESI):420.2[M+1]。
1H NMR(400MHz,DMSO-d 6):δ8.55(d,1H),7.87(t,1H),7.76(s,1H),7.49(d,1H),6.99(d,1H),6.71(brs,2H),6.08-6.09(m,1H),6.01-6.02(m,1H),4.57-4.61(m,2H),4.27-4.29(m,1H),3.74-3.80(m,2H),3.67-3.70(m,2H),2.39(s,3H),1.94-1.99(m,2H)。
实施例17
8-((6-((环丙甲氧基)甲基)吡啶-2-基)甲基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺17
Figure PCTCN2019074324-appb-000078
Figure PCTCN2019074324-appb-000079
将环丙基甲醇(30mg,422μmol)溶于5mL四氢呋喃中,加入氢化钠(33mg,1.41mmol),反应在常温搅拌30分钟,再加入化合物3a(50mg,140μmol),反应在常温搅拌16小时。加水,用乙酸乙酯萃取(10mL×3),合并有机相,无水硫酸钠干燥,减压浓缩,残余物用硅胶柱色谱法以展开剂体系A纯化,得到标题化合物17(8mg,产率:14.5%)。
MS m/z(ESI):391.1[M+1]。
1H NMR(400MHz,CD 3OD)δ8.23-8.24(m,1H),7.88(s,1H),7.68-7.72(t,1H),7.33-7.35(d,1H),7.18-7.20(d,1H),6.44(m,1H),4.61(s,2H),4.09(s,2H),3.40-3.42(d,2H),2.48(s,3H),1.10-1.12(m,1H),0.52-0.55(m,2H),0.22-0.24(m,2H)。
实施例18
(S)-4-(5-甲基呋喃-2-yl)-N 8-(6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺18
Figure PCTCN2019074324-appb-000080
第一步
N-(叔丁基)-4-(5-甲基呋喃-2-基)-8-亚硝基吡唑并[1,5-a][1,3,5]三嗪-2-胺18a
将化合物1j(11.03g,40.65mmol)溶解于250mL乙醇中,冷却到0℃,加入氯化氢二氧六环溶液(4M,30mL),再滴加入亚硝酸异戊酯(4.78g,40.80mmol,5.5mL),加毕,升 温至室温搅拌2小时。将反应液倒入2L饱和碳酸氢钠水溶液中,搅拌5分钟后过滤,滤饼水洗,再将滤饼用500mL二氯甲烷溶解,无水硫酸钠干燥,过滤,滤液减压浓缩,粗品用100mL二氯甲烷和正己烷混合溶剂(V:V=1:10)打浆,过滤,滤饼干燥得到标题化合物18a(10.7g,产率:87.64%)。
MS m/z(ESI):300.9[M+1]。
第二步
(S)-N 2-(叔丁基)-4-(5-甲基呋喃-2-基)-N 8-(6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺18b
在氩气气氛下,将化合物16c(13g,50.36mmol)溶解于150mL四氢呋喃中,冷却到-78℃,滴加入正丁基锂(1.6M,30mL),搅拌反应30分钟,将上述反应液一次性加入到已经预冷至-78℃的化合物18a(6.75g,22.47mmol)的100mL四氢呋喃溶液中,搅拌反应1小时。加入50mL饱和氯化铵水溶液,升温到室温,分液,水相用乙酸乙酯(150mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物18b(1.6g,产率:15.35%)。
MS m/z(ESI):463.9[M+1]。
第三步
(S)-4-(5-甲基呋喃-2-yl)-N 8-(6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺18
将化合物18b(2g,4.31mmol)溶解于30mL三氟乙酸中,加热至70℃反应16小时。冷却至室温,将反应液减压浓缩,残余物中加入饱和碳酸氢钠水溶液至pH大于7,加入二氯甲烷(100mL×3)萃取,合并有机相,无水硫酸钠干燥。过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系A纯化,得到标题化合物18(1.2g,产率:68.26%)。
MS m/z(ESI):408.4[M+1]。
1H NMR(400MHz,DMSO-d 6):δ8.29(s,1H),8.23(d,1H),8.05(s,1H),7.41(t,1H),7.18(s,2H),6.63(d,1H),6.56(d,1H),6.44(d,1H),4.36-4.39(m,2H),4.23-4.26(m,1H),3.72-3.79(m,2H),3.66-3.69(m,2H),2.47(s,3H),1.76-1.99(m,2H)。
实施例19
(S)-4-(5-甲基呋喃-2-基)-8-((6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)氧基)吡唑并[1,5-a][1,3,5]三嗪-2-胺19
Figure PCTCN2019074324-appb-000081
第一步
4-(5-甲基呋喃-2-基)-2-(甲硫基)吡唑并[1,5-a][1,3,5]三嗪-8-甲醛19a
将化合物1h(4g,16.24mmol)溶解于50mL N,N-二甲基甲酰胺中,加入三氯氧磷(3.79g,24.75mmol),加热到80℃反应1小时。将反应液冷却到室温后倒入500mL饱和碳酸氢钠水溶液中,搅拌15分钟,过滤,滤饼依次用水洗,少量甲醇洗,***洗涤,真空干燥得到标题化合物19a(4.3g,产率:96.52%)。
第二步
4-(5-甲基呋喃-2-基)-2-(甲基磺酰基)吡唑并[1,5-a][1,3,5]三嗪-8-酚19b
将化合物19a(4.3g,15.67mmol)溶解于100mL氯仿中,加入间氯过氧苯甲酸(10.8g,62.58mmol),加热到65℃反应1小时。将反应液冷却到室温,依次用饱和碳酸氢钠水溶液(100mL×2)、水(50mL×1)、饱和氯化钠溶液(50mL×1)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系C纯化,得到标题化合物19b(650mg,产率:14.08%)。
第三步
4-(5-甲基呋喃-2-基)-2-(甲基磺酰基)-8-((四氢-2H-吡喃-2-基)氧基)吡唑并[1,5-a][1,3,5]三嗪19c
将化合物19b(650mg,2.21mmol)溶解于50mL二氯甲烷中,加入3,4-二氢-2H-吡喃(6.45g,76.72mmol)和吡啶对甲苯磺酸盐(55mg,218.8μmol),搅拌反应1小时。再加入2mL叔丁胺后旋干反应液,得到粗品标题化合物19c(1g),产品不经纯化直接用于下一步。
第四步
N-(叔丁基)-4-(5-甲基呋喃-2-基)-8-((四氢-2H-吡喃-2-基)氧基)吡唑并[1,5-a][1,3,5]三嗪-2-胺19d
将粗品化合物19c(1.0g,2.64mmol)溶解于30mL 1,4-二氧六环中,加入6mL叔丁胺,封管100℃反应1.5小时。将反应液冷却到室温,减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物19d(420mg,产率:42.78%)。
第五步
2-(叔丁氨基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-酚19e
将化合物19d(420mg,1.13mmol)溶解于30mL甲醇中,加入对甲苯磺酸吡啶盐(28mg,111.4μmol),搅拌反应16小时。将反应液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物19e(140mg,43.09%)。
第六步
(S)-N-(叔丁基)-4-(5-甲基呋喃-2-基)-8-((6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)氧基)吡唑并[1,5-a][1,3,5]三嗪-2-胺19f
在氩气氛下,将化合物19e(50mg,0.174mmol)、化合物16c(54mg,0.209mmol)、三二亚苄基丙酮二钯(8mg,0.009mmol)、2-(二环己基膦)-3,6-二甲氧基-2'-4'-6'-三-I-丙基-11'-联苯(10mg,0.019mmol)和碳酸铯(114mg,0.350mmol)溶解于4mL甲苯中,升温至95℃,搅拌反应3小时。反应液减压浓缩,残留物用硅胶柱色谱法以洗脱剂体系B纯化,得到标题化合物19f(54mg,产率:66.80%)。
MS m/z(ESI):465.2[M+1]。
第七步
(S)-4-(5-甲基呋喃-2-基)-8-((6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)氧基)吡唑并[1,5-a][1,3,5]三嗪-2-胺19
将化合物19f(77mg,0.166mmol)溶解于4mL三氟乙酸中,加热至回流,搅拌2小时。冷却至室温,将反应液减压浓缩,残余物中加入饱和碳酸氢钠水溶液至pH大于7,加入乙酸乙酯(30mL×3)萃取,合并有机相,无水硫酸钠干燥。过滤,滤液减压浓缩,残余物用硅胶柱色谱法以洗脱剂体系B纯化,得到标题化合物19(53.4mg,产率:78.88%)。
MS m/z(ESI):409.2[M+1]。
1H NMR(400MHz,DMSO-d 6):δ8.23-8.22(m,1H),8.20(s,1H),7.82-7.78(m,1H),7.31(brs,2H),7.12-7.10(m,1H),6.91-6.89(m,1H),6.58-6.57(m,1H),4.37(s,2H),4.21-4.20(m,1H),3.74-3.60(m,4H),2.47(s,3H),1.92-1.88(m,2H)。
实施例20
4-(5-甲基呋喃-2-基)-8-((6-丙基吡啶-2-基)甲基)吡唑并[1,5-a][1,3,5]三嗪-2-胺20
Figure PCTCN2019074324-appb-000082
第一步
2-溴-6-(1-((叔丁基二甲基硅烷基)氧基)丙基)吡啶20b
将1-(6-溴吡啶-2-基)丙基-1-醇20a(760mg,3.52mmol,采用公知的方法“Tetrahedron Letters,2014,55(41),5591-5594”制备而得)溶于20mL二氯甲烷中,加入叔丁基二甲基氯硅烷(795mg,5.27mmol)和咪唑(359mg,5.27mmol),搅拌反应16小时。反应液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物20b(830mg,产率:71.44%)。
第二步
6-(1-((叔丁基二甲基硅烷基)氧基)丙基)吡啶-2-甲醛20c
将化合物20b(830mg,2.51mmol)溶于10mL四氢呋喃中,冷却到-78℃,滴加正丁基锂1.6M(1.6M,1.73mL),搅拌反应1小时,再加入N,N-二甲基甲酰胺(367mg,5.02 mmol,387.13uL),反应继续搅拌1小时。加饱和氯化铵水溶液,用乙酸乙酯萃取三次(20mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物20c(480mg,产率:68.36%)。
MS m/z(ESI):280.2[M+1]。
第三步
(2-(叔丁氨基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)(6-(1-((叔丁基二甲基硅烷基)氧基)丙基)吡啶-2-基)甲醇20d
将化合物1k(600mg,1.71mmol)溶于10mL四氢呋喃中,冷却到-78℃,滴加正丁基锂(1.6M,2.20mL),加毕继续搅拌30分钟,再加入化合物20c(480mg,1.72mmol),反应在-78℃搅拌30分钟。加饱和氯化铵水溶液,用乙酸乙酯萃取三次(20mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物20d(300mg,产率:31.79%)。
MS m/z(ESI):551.3[M+1]。
第四步
N-(叔丁基)-8-((6-(1-((叔丁基二甲基硅烷基)氧基)丙基)吡啶-2-基)甲基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺20e
将化合物20d(300mg,544.70μmol)溶于二氯甲烷中,加入三氟乙酸(621mg,5.45mmol,414.00uL),再滴加入三乙基硅烷(633mg,5.44mmol,917.39uL),搅拌反应24小时。加入饱和碳酸氢钠水溶液后分液,水相用二氯甲烷萃取三次(20mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物20e(110mg,产率:37.76%)。
MS m/z(ESI):535.3[M+1]。
第五步
1-(6-((2-(叔丁氨基)-4-(5-甲基呋喃2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)甲基)吡啶-2-基)丙基-1-醇20f
将化合物20e(110mg,205.70μmol)加入10mL四氢呋喃中,再加入正四丁基氟化铵(69mg,306.43μmol),搅拌反应2小时。加水,用乙酸乙酯萃取三次(20mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品标题化合物20f(72mg),产物不经纯化,直接用于下一步反应。
MS m/z(ESI):421.3[M+1]。
第六步
N-(叔丁基)-8-((6-(1-氯丙基)吡啶-2-基)甲基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺20g
将粗品化合物20f(170mg,404.27μmol)溶解于20mL二氯甲烷中,再加入氯化亚砜(96mg,806.93μmol),搅拌反应2小时。将反应液倒入冰水中,用二氯甲烷萃取三次(20mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品标题化合物20g(170mg),产物不经纯化,直接用于下一步反应。
MS m/z(ESI):439.2[M+1]。
第七步
N-(叔丁基)-4-(5-甲基呋喃-2-基)-8-((6-丙基吡啶-2-基)甲基)吡唑并[1,5-a][1,3,5]三嗪-2-胺20h
将化合物20g(170mg,387.29μmol)溶解于20mL甲醇中,再加入钯碳加氢催化剂(湿)(82mg,38.53μmol,5%purity),用氢气置换三次,搅拌反应1小时。过滤,滤液减压浓缩,残余物用硅胶柱色谱法以展开剂体系A纯化得到标题化合物20h(72mg,产率:45.96%)。
MS m/z(ESI):405.2[M+1]。
第八步
4-(5-甲基呋喃-2-基)-8-((6-丙基吡啶-2-基)甲基)吡唑并[1,5-a][1,3,5]三嗪-2-胺20
将化合物20h(72mg,177.99μmol)溶于5mL三氟乙酸中,70℃搅拌16小时。冷却,减压浓缩,残余物加水,用饱和碳酸氢钠调PH大于7,乙酸乙酯萃取三次(20mL×3),合并有机相,无水硫酸钠干燥,过滤,减压浓缩,残余物用硅胶柱色谱法以展开剂体系A纯化得到标题化合物20(41mg,产率:66.11%)。
MS m/z(ESI):349.2[M+1]。
1H NMR(400MHz,DMSO-d 6):δ8.19-8.20(m,1H),7.98(s,1H),7.54-7.56(m,1H),7.22(br,2H),7.04-7.05(d,1H),6.98-7.00(d,1H),6.53-6.54(m,1H),3.97(s,2H),2.65-2.69(t,2H),2.46(s,3H),1.65-1.71(m,2H),0.89-0.93(t,3H)。
实施例21
(R)-4-(5-甲基呋喃-2-基)-N 8-(6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺21
Figure PCTCN2019074324-appb-000083
第一步
(S)-2-溴-6-(((四氢呋喃-3-基)氧基)甲基)吡啶21b
采用实施例16中化合物16c的合成路线,将第一步原料化合物16a替换为原料化合物(3R)-四氢呋喃-3-醇21a,制得标题化合物21b(3.87g)。
采用实施例18的合成路线,将第二步原料化合物16c替换为原料化合物21b,制得标题化合物21(125mg)。
MS m/z(ESI):408.3[M+1]。
1H NMR(400MHz,DMSO-d 6)δ8.29(s,1H),8.23(d,1H),8.05(s,1H),7.41(t,1H),7.18(s,2H),6.63(d,1H),6.57(d,1H),6.44(d,1H),4.36(s,2H),4.24-4.25(m,1H),3.73-3.79(m,2H),3.66-3.71(m,2H),2.47(s,3H),1.93-1.99(m,2H)。
实施例22
8-((6-((环戊基氧基)甲基)吡啶-2-基)甲基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺22
Figure PCTCN2019074324-appb-000084
采用实施例17的合成路线,将原料环丙基甲醇替换成环戊醇,制得目标产物22(5 mg)。
MS m/z(ESI):405.0[M+1]。
1H NMR(400MHz,CD 3OD):δ8.25-8.26(m,1H),7.90(s,1H),7.71-7.75(m,1H),7.35(d,1H),7.21(d,1H),6.44-6.45(m,1H),4.57(s,2H),4.09-4.11(m,3H),2.49(s,3H),1.71-1.78(m,6H),1.56-1.58(m,2H)。
实施例23
4-(5-甲基呋喃-2-基)-8-(吡啶-2-基氧基)吡唑并[1,5-a][1,3,5]三嗪-2-胺23
Figure PCTCN2019074324-appb-000085
采用实施例19的合成路线,将第六步原料16c替换成2-溴吡啶,制得目标产物23(5mg)。
MS m/z(ESI):309.1[M+1]。
1H NMR(400MHz,CD 3OD):δ8.32-8.33(m,1H),8.05-8.06(m,1H),8.05(s,1H),7.79-7.83(m,1H),7.03-7.10(m,2H),6.47-6.48(m,1H),2.50(s,3H)。
实施例24
(S)-4-(5-甲基呋喃-2-基)-8-(3-(((四氢呋喃-3-基)氧基)甲基)苄基)吡唑并[1,5-a][1,3,5]三嗪-2-胺24
Figure PCTCN2019074324-appb-000086
Figure PCTCN2019074324-appb-000087
第一步
(S)-3-((3-(溴甲基)苄基)氧基)四氢呋喃24b
将化合物16a(8.41g,95.47mmol,7.65mL)溶于300mL四氢呋喃中,加入钠氢(4.00g,100.02mmol,60%purity),室温搅拌1小时。再加入1,3-二(溴甲基)苯24a(24g,90.92mmol,阿达玛斯试剂有限公司),反应回流16小时。加入饱和氯化钠溶液100mL和200mL乙酸乙酯,分液,水相用乙酸乙酯萃取三次(100mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物24b(13.75g,产率:55.77%)。
第二步
(S)-3-(((四氢呋喃-3-基)氧基)甲基)苯甲醛24c
将化合物24b(4.42g,16.30mmol),分子筛(2g,16.30mmol)加入到200mL乙腈中,加入N-甲基-N-氧化***啉(3.82g,32.60mmol),反应1小时。过滤,滤饼用乙酸乙酯洗涤一次,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物24c(2.14g,产率:63.57%)。
采用实施例1的合成路线,将第九步原料化合物1d替换为化合物24c,制得标题产物24(15mg)。
MS m/z(ESI):406.2[M+1]。
1H NMR(400MHz,CDCl 3):δ8.37-8.39(m,1H),7.85(s,1H),7.22-7.24(m,2H),6.39-6.40(m,1H),5.35-5.36(m,2H),4.51-4.53(m,2H),4.23-4.25(m,1H),3.84-4.02(m,6H),2.57(s,3H),2.05-2.07(m,2H)。
实施例25
(S)-4-(5-甲基呋喃-2-基)-N 8-(3-(((四氢呋喃-3-基)氧基)甲基)苯基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺25
Figure PCTCN2019074324-appb-000088
采用实施例21的合成路线,将第一步原料化合物16b和化合物21a分别替换为化合物1-溴-3-溴甲基苯(韶远科技(上海)有限公司)和化合物16a,得到标题化合物25(10mg,产率:5.2%)。
MS m/z(ESI):407.2[M+1]。
1H NMR(400MHz,DMSO-d 6):δ8.23(d,1H),8.05(s,1H),7.24(brs,2H),7.16(s,1H),7.01(t,1H),6.56(brs,2H),6.47-6.53(m,2H),4.30(s,2H),4.13(s,1H),3.60-3.71(m,4H),2.47(s,3H),1.89(brs,2H)。
实施例26
(S)-N 8-(2-氟-3-(((四氢呋喃-3-基)氧基)甲基)苯基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺26
Figure PCTCN2019074324-appb-000089
第一步
N 2-(叔丁基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺26c
将化合物18a(950mg,3.16mmol)溶于20mL乙醇和10mL水中,加热至70℃,分别加入氯化铵(1.38g,25.32mmol)和铁粉(884mg,15.83mmol)。保温搅拌1小时,硅藻土 过滤,乙酸乙酯洗涤(50mL×3),滤液减压浓缩,再加入饱和碳酸氢钠溶液中和,乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠溶液洗涤(20mL),硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物26c(780mg,,产率:86.11%)。
第二步
(S)-3-((3-溴-2-氟苄基)氧基)四氢呋喃26b
将化合物16a(658mg,7.47mmol)溶于20mL N,N二甲基甲酰胺中,加入钠氢(299mg,7.48mmol,60%purity),反应1小时。再加入1-溴-3-(溴甲基)-2-氟苯26a(1.00g,3.73mmol,采用专利申请“US2012172448A1”公开的方法制备而得),反应继续17小时。加水,用乙酸乙酯萃取(20mL×3),合并有机相,用水洗(20mL×2),饱和氯化钠溶液洗涤(20mL),无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物26b(950mg,产率:92.51%)。
第三步
(S)-N 2-(叔丁基)-N 8-(2-氟-3-(((四氢呋喃-3-基)氧基)甲基)苯基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺26d
在氩气氛下,依次将化合物26b(500mg,1.82mmol),化合物26c(496mg,1.73mmol),三(二亚苄基丙酮)二钯(159mg,173.63μmol),4,5-双二苯基膦-9,9-二甲基氧杂蒽(200mg,345.65μmol)和叔丁醇钠(167mg,1.74mmol)加入到50mL甲苯中,100℃搅拌17小时。冷却,硅藻土过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物26d(200mg,产率:24.04%)。
MS m/z(ESI):481.3[M+1]。
第四步
(S)-N 8-(2-氟-3-(((四氢呋喃-3-基)氧基)甲基)苯基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺26
将化合物26d(200mg,416.20μmol)溶解于5mL三氟乙酸中,加热至70℃反应7小时。冷却至室温,将反应液减压浓缩,残余物中加入饱和碳酸氢钠水溶液至pH大于7,加入二氯甲烷(100mL×3)萃取,合并有机相,无水硫酸钠干燥。过滤,滤液减压浓缩,残余物用硅胶柱色谱法以展开剂体系A纯化,得到标题化合物26(45mg,产率:25.47%)。
MS m/z(ESI):425.1[M+1]。
1H NMR(400MHz,CDCl 3):δ8.33-8.34(m,1H),8.03(s,1H),6.71-6.91(m,1H),6.69-6.72(m,1H),6.56-6.60(m,1H),6.49-6.50(m,1H),4.59-4.61(m,2H),4.31-4.32(m,1H), 3.81-3.90(m,4H),2.52(s,3H),2.00-2.10(m,2H)。
实施例27
4-(5-甲基呋喃-2-基)-N 8-(6-(***啉基甲基)吡啶-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺27
Figure PCTCN2019074324-appb-000090
第一步
N 2-(叔丁基)-N 8-(6-(((叔丁基二甲基硅烷基)氧基)甲基)吡啶-2-基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺27b
将2-溴-6-(((叔丁基二甲基硅烷基)氧基)甲基)吡啶27a(2.013g,6.66mmol)溶于10mL四氢呋喃中,冷却到-78℃,滴加入正丁基锂(1.6M,4.17mL),加毕,-78℃反应30分钟,将上述反应液加入到已经预冷至-78℃的化合物18a(1.00g,3.33mmol)30mL四氢呋喃溶液中,-78℃反应1小时。加入饱和氯化铵水溶液,分液,水相用乙酸乙酯萃取三次(50mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物27b(300mg,产率:8.87%)。
第二步
(6-((2-氨基-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)氨基)吡啶-2-基)甲醇27c
将化合物27b(300mg,590.90μmol)溶于10mL三氟乙酸中,70℃搅拌16小时。溶剂减压浓缩,残余物中加水,用饱和碳酸氢钠调节PH大于7,用二氯甲烷萃取三次(20mL×3),合并有机相,无水硫酸钠干燥,滤液减压浓缩,得粗品标题化合物27c(199mg),产物不经纯化,直接用于下一步反应。
第三步
N 8-(6-(氯甲基)吡啶-2-基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺27d
将化合物27c(199mg,589.92μmol)溶于10mL二氯甲烷中,加入氯化亚砜(140mg, 1.18mmol),搅拌1小时。加入碳酸氢钠水溶液并调节pH大于7,分液,水相用二氯甲烷萃取三次(20mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物27d(120mg,57.17%)。
MS m/z(ESI):356.1[M+1]。
第四步
4-(5-甲基呋喃-2-基)-N 8-(6-(***啉基甲基)吡啶-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺27
将化合物27d(30mg,84.3215μmol)和***啉(73mg,837.92μmol)加入10mL四氢呋喃中,70℃搅拌16小时。冷却,减压浓缩,残余物用硅胶柱色谱法以展开剂体系A纯化,得到粗品,粗品用洗脱剂体系B洗涤二次,得到标题化合物27(21mg,产率:61.27%)。
MS m/z(ESI):407.2[M+1]。
1H NMR(400MHz,DMSO-d 6):δ8.30-8.31(m,1H),8.23-8.24(m,1H),8.01(s,1H),7.36-7.40(m,1H),7.17(brs,2H),6.64(d,1H),6.56-6.57(m,1H),6.39(d,1H),3.57-3.60(m,4H),3.40(s,2H),2.47(s,3H),2.42-2.43(m,4H)。
实施例28
(S)-4-(5-甲基呋喃-2-基)-8-((6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)硫代)吡唑并[1,5-a][1,3,5]三嗪-2-胺28
Figure PCTCN2019074324-appb-000091
第一步
(S)-2-(苄氧基)-6-(((四氢呋喃-3-基)氧基)甲基)吡啶28a
将化合物16c(7.00g,27.12mmol),苯甲醇(3.52g,32.55mmol)溶解于200mL N,N-二甲基甲酰胺中,加入氢化钠(1.62g,40.67mmol,60%purity),加热到90℃搅拌17小时。将反应液冷却至室温,加入500mL水,用乙酸乙酯(250mL×3)萃取,合并有机相,依次用水(250mL)、饱和氯化钠溶液(250mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物28a(7.32g,产率:94.59%)。
第二步
(S)-6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-酚28b
将化合物28a(7.32g,25.65mmol)溶解于500mL甲醇,加入10%钯碳(1.36g,12.82mmol,含水50%),在氢气气氛下搅拌4小时。将反应液过滤,滤液浓缩得到标题化合物28b(4.8g,产率:95.84%)。
第三步
(S)-6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-硫酚28c
在氩气气氛下,将化合物28b(1.37g,7.01mmol),1,3,2,4-二硫,2,4-双(4-甲氧基苯基)-2,4-二硫化物(1.72g,4.25mmol)加入到50mL甲苯中,加热到115℃反应16小时。将反应液冷却到室温,旋干,用CombiFlash快速制备仪以洗脱剂体系A纯化后,再加入20mL 3M氢氧化钠水溶液,用甲基叔丁基醚(50mL×3)萃取,水相用柠檬酸调节至pH小于7,再用二氯甲烷(100mL×4)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩得到标题化合物28c(580mg,产率:39.11%)。
第四步
1,2-双(6-((((S)-四氢呋喃-3-基)氧基)甲基)吡啶-2-基)二硫醚28d
将氢氧化钠(126mg,3.15mmol)溶解于5mL水中,把所得溶液加入到化合物28c(580mg,2.74mmol)中,搅拌0.5小时。再加入铁***(1.04g,3.15mmol)的10mL水溶液,搅拌16小时。将反应液过滤,滤饼用水彻底洗净,真空干燥得标题化合物28d(385mg,产率;33.35%)。
第五步
(S)-N-(叔丁基)-4-(5-甲基呋喃-2-基)-8-((6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)硫代)吡唑并[1,5-a][1,3,5]三嗪-2-胺28e
将化合物1k(217mg,619.2μmol)溶解于20mL四氢呋喃中,冷却到-78℃,滴加入 0.78mL的1.6M正丁基锂,反应0.5小时,再加入化合物28d的10mL四氢呋喃溶液,反应1小时。在反应液中加入30mL饱和氯化铵水溶液后升温到室温,分液,水相用乙酸乙酯(50mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物28e(120mg,产率:40.29%)。
第六步
(S)-4-(5-甲基呋喃-2-基)-8-((6-(((四氢呋喃-3-基)氧基)甲基)吡啶-2-基)硫代)吡唑并[1,5-a][1,3,5]三嗪-2-胺28
将化合物28e(120mg,249.6μmol)溶解于5mL三氟乙酸中,加热到70℃反应过夜。冷却至室温,将反应液减压浓缩,残余物中加入饱和碳酸氢钠水溶液至pH大于7,加入二氯甲烷(30mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系A纯化,得到标题化合物28(43mg,产率:40.57%)。
MS m/z(ESI):425.2[M+1]。
1H NMR(400MHz,DMSO-d 6)δ8.24(s,1H),8.23(d,1H),7.61(s,2H),7.56(t,1H),7.10(d,1H),6.72(d,1H),6.58-6.59(m,1H),4.47(s,2H),4.24-4.25(m,1H),3.73-3.79(m,2H),3.66-3.69(m,2H),2.48(s,3H),1.93-1.99(m,2H)。
实施例29
4-(5-甲基呋喃-2-基)-N 8-(6-((4-甲基哌嗪-1-基)甲基)吡啶-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺29
Figure PCTCN2019074324-appb-000092
采用实施例27的合成路线,将第四步原料化合物***啉替换为化合物N-甲基哌嗪,制得标题产物29(5mg,产率:14.13%)。
MS m/z(ESI):420.2[M+1]。
1H NMR(400MHz,CD 3OD)δ8.27-8.31(m,2H),7.43-7.45(m,1H),6.72-6.77(m,1H),6.45-6.47(m,2H),3.52(s,2H),2.50(s,3H),2.47-2.70(m,8H),2.29(s,3H)。
实施例30
4-(5-甲基呋喃-2-基)-N 8-(6-((四氢-1H-呋喃并[3,4-c]吡咯-5(3H)-基)甲基)吡啶-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺30
Figure PCTCN2019074324-appb-000093
采用实施例27的合成路线,将第四步原料化合物***啉替换为化合物六氢-1H-呋喃并[3,4-c]吡咯,制得标题产物30(18mg,产率:29.61%)。
MS m/z(ESI):433.2[M+1]。
1H NMR(400MHz,DMSO-d 6)δ8.30(s,1H),8.23-8.24(m,1H),8.00-8.01(m,1H),7.36-7.41(m,1H),7.18(brs,2H),6.63(d,1H),6.56-6.57(m,1H),6.41-6.43(m,1H),3.70-3.72(m,2H),3.44-3.53(m,3H),3.24-3.25(m,1H),2.70-2.71(m,2H),2.47(s,3H),2.39-2.41(m,2H),1.23-1.24(m,1H),0.81-0.82(m,1H)。
实施例31
N 8-(2-氟苯基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺31
Figure PCTCN2019074324-appb-000094
第一步
N 2-(叔丁基)-N 8-(2-氟苯基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺31b
在氩气气氛下,将2-氟溴苯(160mg,914.2μmol),化合物26a(250mg,873.1μmol),叔丁醇钠(84mg,874.0μmol),4,5-双(二苯基膦)-9,9-二甲基氧杂蒽(101mg,174.5μmol)和三(二亚苄基丙酮)二钯(80mg,87.36μmol)加入到15mL甲苯中,加热到95℃反应16小 时。冷却到室温后减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物31b(226mg,产率:68.04%)。
第二步
N-(2-氨基-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)-2,2,2-三氟-N-(2-氟苯基)乙酰胺31c
将化合物31b(220mg,578.31μmol)溶解于10mL三氟乙酸中,加热到70℃反应16小时。将反应液冷却到室温,减压浓缩,残余物用饱和碳酸氢钠水溶液调节至pH大于7,加入二氯甲烷(30mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物31c(75mg,产率:30.85%)。
第三步
N 8-(2-氟苯基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺31
将化合物31c(75mg,178.4μmol)加入到10mL甲醇中,加入碳酸钾(200mg,1.45mmol),加热到50℃反应2小时。将反应液冷却到室温,减压浓缩,加入20mL水和20mL二氯甲烷,分液,水相用二氯甲烷(30mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化得到标题化合物31(30mg,产率:51.84%)。
MS m/z(ESI):325.1[M+1]。
1H NMR(400MHz,DMSO-d 6)δ8.24(d,1H),8.05(s,1H),7.28(s,2H),7.03-7.06(m,1H),7.00(s,1H),6.85(d,1H),6.56-6.57(m,2H),6.45-6.47(m,1H),2.48(s,3H)。
实施例32
N 8-(6-((2-氧杂-6-氮杂螺[3.3]庚烷-6-基)甲基)吡啶-2-基)-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2,8-二胺32
Figure PCTCN2019074324-appb-000095
采用实施例27的合成路线,将第四步原料化合物***啉替换为化合物2-氧杂-6-氮杂螺[3.3]庚烷半乙二酸盐(南京药石科技股份有限公司),制得标题产物32(43mg)。
MS m/z(ESI):419.2[M+1]。
1H NMR(400MHz,CD 3OD)δ8.30-8.31(m,1H),8.27(s,1H),7.41-7.45(t,1H),6.59(d,1H),6.46-6.50(m,2H),4.72(s,4H),3.61(s,2H),3.58(s,4H),2.49(s,3H)。
实施例33
8-苄基-4-(呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺33
Figure PCTCN2019074324-appb-000096
采用实施例10的合成路线,将第五步原料化合物4a替换为化合物苯甲醛,制得标题产物33(82mg,产率:69.85%)。
MS m/z(ESI):292.2[M+1]。
1H NMR(400MHz,DMSO-d 6):δ8.24-8.25(m,1H),8.17-8.18(m,1H),7.95-7.96(m,1H),7.23-7.24(m,4H),7.15-7.17(m,1H),6.87-6.89(m,1H),3.87(s,2H)。
实施例34
8-(3-氟苄基)-4-(呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺34
Figure PCTCN2019074324-appb-000097
第一步
(2-(叔丁氨基)-4-(呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)(3-氟苯基)甲醇34b
将化合物10e(600mg,1.78mmol)溶解于50mL四氢呋喃中,冷却到-78℃,滴加入正丁基锂(1.6M,2.2mL),反应30分钟。再加入化合物3-氟苯甲醛34a(332mg,2.67mmol),反应30分钟。加入30mL饱和氯化铵水溶液,升温到室温,分液,水相用乙酸乙酯(50mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速 制备仪以洗脱剂体系B纯化,得到标题化合物34b(210mg,产率:30.85%)。
第二步
N-(叔丁基)-8-(3-氟苄基)-4-(呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺34c
将化合物34b(210mg,550μmol)溶解于10mL二氯甲烷中,加入三氟乙酸(1.25g,10.96mmol),再滴加入三乙基硅烷(1.28g,11.01mmol),搅拌反应16小时,加入20mL饱和碳酸氢钠水溶液,分液。水相用二氯甲烷(20mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物34c(160mg,产率:79.53%)。
第三步
8-(3-氟苄基)-4-(呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-2-胺34
将化合物34c(160mg,437μmol)溶解于5mL三氟乙酸中,加热至70℃反应16小时,减压浓缩反应液,残余物中加入饱和碳酸氢钠水溶液至pH大于7,加入二氯甲烷(30mL×3)萃取,合并有机相,无水硫酸钠干燥。过滤,滤液减压浓缩,残余物用薄层色谱法以洗脱剂体系B纯化,得到标题化合物34(10mg,产率:7.38%)。
MS m/z(ESI):309.9[M+1]。
1H NMR(400MHz,CD 3OD)δ8.35(d,1H),7.99(s,1H),7.89(s,1H),7.30-7.25(m,1H),7.11-7.09(m,1H),7.03-7.00(m,1H),6.92-6.83(m,2H),3.97(s,2H)。
实施例35
2-(6-((2-氨基-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)氧基)吡啶-2-基)丙基-2-醇35
1-(6-((2-氨基-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)氧基)吡啶-2-基)乙基-1-酮36
Figure PCTCN2019074324-appb-000098
Figure PCTCN2019074324-appb-000099
采用实施例19的合成方法,将第六步原料16c替换成6-溴吡啶甲酸甲酯,得到化合物6-((2-氨基-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)氧基)吡啶甲酸甲酯35a(60mg,产率:93.49%)。
将化合物35a(60mg,163.7μmol)溶解于10mL四氢呋喃中,加入甲基溴化镁(3M,0.28mL),搅拌反应30分钟,再补加入甲基溴化镁(3M,0.28mL),搅拌反应30分钟。加入10mL饱和氯化铵水溶液,分液,水相用乙酸乙酯(30mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用制备板以体系A纯化,得到标题化合物35(16mg)和化合物36(20mg)。
化合物35
MS m/z(ESI):367.2[M+1]。
1H NMR(400MHz,DMSO-d 6)δ8.20-8.21(m,2H),7.72(t,1H),7.29(d,1H),7.25(s,2H),6.74(d,1H),6.53-6.54(m,1H),5.09(s,1H),2.44(s,3H),1.28(s,6H)。
化合物36
MS m/z(ESI):351.1[M+1]。
1H NMR(400MHz,DMSO-d 6)δ8.29(s,1H),8.20(d,1H),7.96(t,1H),7.63(d,1H),7.26-7.28(m,3H),6.52(d,1H),2.43(s,3H),2.38(s,3H)。
实施例37
1-(6-((2-氨基-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)氧基)吡啶-2-基)乙基-1-醇37
Figure PCTCN2019074324-appb-000100
将化合物36(6mg,17.12μmol)溶解于6mL甲醇中,加入硼氢化锂(2M,0.1mL),搅拌反应1小时。加入15mL氯化铵水溶液,搅拌5分钟,用二氯甲烷(30mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用制备板以体系A纯化,得到标题化合物37(2mg,产率:33.14%)。
MS m/z(ESI):353.1[M+1]。
1H NMR(400MHz,CD 3OD)δ8.34(d,1H),8.12(d,1H),7.78(t,1H),7.22(d,1H),6.85(d,1H),6.48(d,1H),4.70(q,1H),2.51(d,3H),1.40(d,3H)。
实施例38
2-(6-((2-氨基-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)甲基)吡啶-2-基丙基-2-醇38
Figure PCTCN2019074324-appb-000101
Figure PCTCN2019074324-appb-000102
采用实施例1的合成路线,将第九步原料化合物1d替换成6-甲酰基-2-吡啶甲酸甲酯(上海毕得科技医药有限公司),制得化合物6-((2-氨基-4-(5-甲基呋喃-2-基)吡唑并[1,5-a][1,3,5]三嗪-8-基)甲基)吡啶甲酸甲酯38a(70mg)。
将化合物38a(70mg,192μmol)溶解于20mL四氢呋喃中,冷却到0℃,加入甲基溴化镁(91mg,768μmol),缓慢升至室温,继续反应2小时,加入30mL饱和氯化铵水溶液,升温到室温,分液,水相用乙酸乙酯(50mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,残余物用CombiFlash快速制备仪以洗脱剂体系B纯化,得到标题化合物38(20mg,产率:28.56%)。
MS m/z(ESI):365.3[M+1]。
1H NMR(400MHz,CDCl 3):δ8.35(d,1H),7.96(s,1H),7.64(s,1H),7.20-7.14(m,2H),5.41(s,2H),4.20(s,2H),2.54(s,3H),1.55(s,6H)。
测试例:
生物学评价
测试例1、本公开化合物对腺苷A 2a受体(adenosine A 2a receptor,A 2aR)cAMP信号通路,腺苷A 1受体(adenosine A 1 receptor,A 1R)cAMP信号通路和腺苷A 3受体(adenosine A 3 receptor,A 3R)cAMP信号通路抑制活性的测定。
以下方法用来测定本公开化合物对腺苷A 2a受体cAMP信号通路,腺苷A 1受体cAMP信号通路和腺苷A 3受体cAMP信号通路的抑制活性。实验方法简述如下:
一、实验材料及仪器
1.CHO-K1/A 2aR细胞(NM_000675.5)或CHO-K1/A 1R细胞(NM_000674.2)或CHO-K1/A 3R细胞(NM_000677.3)
2.胎牛血清(Gibco,10099-141)
3.博来霉素(Thermo,R25001)或G418(ENZO,ALX-380-013-G005)或嘌呤霉素(Thermo,10687-010)
4.DMEM/F12培养基(GE,SH30023.01)
5.细胞分离缓冲液(Thermo Fisher,13151014)
6.HEPES(Gibco,42360-099)
7.牛血清白蛋白(MP Biomedicals,219989725)
8.咯利普兰(sigma,R6520-10MG)
9.腺苷脱氨酶(sigma,10102105001)
10.毛喉素(sigma,F6886)
11.2Cl-IB-MECA(Tocrics,1104/10)
12.N6-环戊基腺苷(Tocris,1702/50)
13.平衡盐缓冲液(Thermo,14025-092)
14.cAMP动态2试剂盒(cAMP dynamic 2 kit)(Cisbio,62AM4PEB)
15.384孔板(Corning,4514)或(Nunc,267462#)
16.乙基咔唑(Torcis,1691/10)
17.PHERAstar多功能酶标仪(Cisbio,62AM4PEB)
二、实验步骤
2.1腺苷A 2a受体
CHO-K1/A 2aR细胞用含有10%胎牛血清和800μg/mL博来霉素的DMEM/F12培养基进行培养。实验时使用细胞分离缓冲液消化细胞,用含有20mM HEPES和0.1%牛血清白蛋白的平衡盐缓冲液重悬细胞并计数,将细胞密度调整为10 6个/mL。在384孔板中每孔加入5μL细胞悬液,2.5μL用含有20mM HEPES,0.1%牛血清白蛋白,54μM咯利普兰和2.7U/mL腺苷脱氨酶的平衡盐缓冲液配制的4×浓度的受试化合物,室温孵育30分钟。每孔再加入2.5μL用含有20mM HEPES,0.1%牛血清白蛋白,54μM咯利普兰和2.7U/mL腺苷脱氨酶的平衡盐缓冲液配制的4×浓度的乙基咔唑,室温孵育30分钟。化合物终浓度是:10000,2000,400,80,16,3.2,0.64,0.128,0.0256,0.00512,0.001024nM,乙基咔唑终浓度是20nM。细胞内cAMP浓度使用cAMP动态2试剂盒检测。用cAMP裂解缓冲液按1:4的比例分别稀释cAMP-d2和抗cAMP-Eu-穴状化合物(Anti-cAMP-Eu-Cryptate)。每孔加入5μL稀释后的cAMP-d2,再加入5μL稀释后的抗cAMP-Eu-穴状化合物,室温避光孵育1小时。采用PHERAstar多功能酶标仪读取HTRF信号值。用Graphpad Prism软件计算化合物抑制活性的IC 50值,见表1。
2.2腺苷A 1受体
CHO-K1/A 1R用含有10%胎牛血清和1mg/mL G418的DMEM/F12培养基进行培养。 实验时使用细胞分离缓冲液消化细胞,然后用含有20mM HEPES和0.1%牛血清白蛋白的平衡盐缓冲液重悬细胞并计数,将细胞密度调整为5×10 5个/mL。在384孔板中每孔加入12.5μL细胞悬液,6.25μL用含有20mM HEPES,0.1%牛血清白蛋白,54μM咯利普兰和2.7U/mL腺苷脱氨酶的平衡盐缓冲液配制的4×浓度的受试化合物,室温孵育30分钟。每孔再加入6.25μL用含有20mM HEPES,0.1%牛血清白蛋白,54μM咯利普兰和2.7U/mL腺苷脱氨酶的平衡盐缓冲液配制的4×浓度的毛喉素和N6-环戊基腺苷,室温孵育30分钟。化合物终浓度是:100000,10000,1000,100,10,1,0.1和0nM,毛喉素的终浓度是10μM,CPA的终浓度是10nM。细胞内cAMP浓度使用cAMP动态2试剂盒检测。用cAMP裂解缓冲液按照1:4的比例分别稀释cAMP-d2和抗cAMP-Eu-穴状化合物。每孔加入12.5μL稀释后的cAMP-d2,再加入12.5μL稀释后的抗cAMP-Eu-穴状化合物,室温避光孵育1小时。采用PHERAstar多功能酶标仪读取HTRF信号值。用Graphpad Prism软件计算化合物抑制活性的IC 50值,见表2。
2.3腺苷A 3受体
CHO-K1/A 3R用含有10%胎牛血清和10μg/mL嘌呤霉素的DMEM/F12培养基进行培养。实验时使用细胞分离缓冲液消化细胞,用含有20mM HEPES和0.1%牛血清白蛋白的平衡盐缓冲液重悬细胞并计数,将细胞密度调整为5×10 5/mL。在384孔板中每孔加入12.5μL细胞悬液,6.25μL用含有20mM HEPES,0.1%牛血清白蛋白,54μM咯利普兰和2.7U/mL腺苷脱氨酶的平衡盐缓冲液配制的4×浓度的受试化合物,室温孵育30分钟。每孔再加入6.25μL用含有20mM HEPES,0.1%牛血清白蛋白,54μM咯利普兰和2.7U/mL腺苷脱氨酶的平衡盐缓冲液配制的4×浓度的毛喉素和2Cl-IB-MECA,室温孵育30分钟。化合物终浓度是:100000,10000,1000,100,10,1,0.1和0nM,毛喉素的终浓度是10μM,2Cl-IB-MECA的终浓度是5nM。细胞内cAMP浓度使用cAMP动态2试剂盒检测。用cAMP裂解缓冲液按照1:4的比例分别稀释cAMP-d2和抗cAMP-Eu-穴状化合物。每孔加入12.5μL稀释后的cAMP-d2,再加入12.5μL稀释后的抗cAMP-Eu-穴状化合物,室温避光孵育1小时。采用PHERAstar多功能酶标仪读取HTRF信号值。用Graphpad Prism软件计算化合物抑制活性的IC 50值,见表3。
表1本公开化合物对腺苷A 2a受体cAMP信号通路抑制活性的IC 50
实施例编号 IC 50/nM(A 2aR)
1 0.33
2 0.87
3 0.45
4 0.14
5 0.55
6 0.4
7 1.18
8 1.32
9 1.64
10 1.71
11 1.86
13 1.7
14 0.68
15 1.83
17 0.72
18 0.87
19 0.36
20 0.09
21 0.4
22 0.57
23 0.58
24 1.2
25 1.64
26 1.66
27 1.9
28 2.13
29 2.8
30 2.87
31 3.53
32 3.74
33 4.14
34 4.64
结论:本公开化合物对腺苷A 2a受体cAMP信号通路具有明显的抑制活性。
表2本公开化合物对腺苷A 1受体cAMP信号通路抑制活性的IC 50
Figure PCTCN2019074324-appb-000103
结论:本公开化合物对腺苷A 1受体抑制活性作用较弱,说明本公开化合物对A 2a受 体具有高选择性。
表3本公开化合物对腺苷A 3受体cAMP信号通路抑制活性的IC 50
Figure PCTCN2019074324-appb-000104
Figure PCTCN2019074324-appb-000105
结论:本公开化合物对A 3受体抑制活性作用较弱,说明本公开化合物对A 2a受体具有高选择性。
药代动力学评价
测试例2、本公开化合物的小鼠药代动力学测试
1、摘要
以小鼠为受试动物,应用LC/MS/MS法测定了小鼠灌胃给予实施例1化合物、实施例18化合物和实施例19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
实施例1化合物、实施例18化合物和实施例19化合物。
2.2试验动物
C57小鼠27只,雌性,每组9只,平均分成3只,购自上海杰思捷实验动物有限公司,动物生产许可证号:SCXK(沪)2013-0006。
2.3药物配制
称取一定量药物,加2.5%体积的DMSO、2.5%体积的tween80和95%生理盐水配置成0.1mg/mL无色澄清透明液体。
2.4给药
C57小鼠禁食过夜后灌胃给药,给药剂量均为2.0mg/kg,给药体积均为0.2mL/10g。
3、操作
小鼠灌胃给药实施例1化合物、实施例18化合物和实施例19化合物,于给药前及给药后0.5,1.0,2.0,4.0,6.0,8.0,11.0,24.0小时采血0.1mL(每个时间点3只动物),置于肝素化试管中,3500转/分钟离心10分钟分离血浆,于-20℃保存,给药后2小时进食。
测定不同浓度的药物灌胃给药后小鼠血浆中的待测化合物含量:取给药后各时刻的小鼠血浆25μL,加入内标溶液喜树碱30μL(100ng/mL),乙腈200μL,涡旋混合5分钟,离心10分钟(4000转/分钟),取上清液3μL进行LC/MS/MS分析。
4、药代动力学参数结果
本公开化合物的药代动力学参数如下表4:
表4本公开化合物的药代动力学参数
Figure PCTCN2019074324-appb-000106
结论:本公开化合物的药代吸收较好,具有药代动力学优势。

Claims (21)

  1. 一种通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐:
    Figure PCTCN2019074324-appb-100001
    其中:
    L选自CR 4R 5、O、NH或S;
    环A和环B相同或不同,且各自独立地选自环烷基、杂环基、芳基和杂芳基;
    R 1相同或不同,且各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基、杂芳基和-Y-R a
    Y选自共价键或亚烷基;
    R a选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、-OR c、-COR 9、-COOR 9、-OS(O) mR 6、芳基和杂芳基;其中所述的烷基、烷氧基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代;
    R c选自氢原子、烷基、卤代烷基、羟烷基、环烷基和杂环基,其中所述的烷基、环烷基和杂环基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基和杂环基中的一个或多个取代基所取代;
    R 2选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基;
    R 3选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基;
    R 4和R 5相同或不同,且各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、 羟基和羟烷基;
    或者R 4和R 5一起形成=NH或=O;
    R 6选自氢原子、卤素、烷氧基、卤代烷氧基、氨基、环烷基、杂环基、芳基、杂芳基和-NR 7R 8
    R 7和R 8相同或不同,且各自独立地选自氢原子、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基;
    或者R 7和R 8与相连的氮原子一起形成杂环基,其中所述的杂环基除含有1个氮原子之外,还任选含有1~2个相同或不同选自N、O和S的杂原子,并且所述的杂环基任选被选自烷基、烷氧基、氧代基、卤素、氨基、氰基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    R 9选自氢原子、烷基、卤代烷基、烷氧基、卤代烷氧基、氨基、环烷基、杂环基、芳基和杂芳基;
    n为0、1、2、3或4;
    s为0、1、2或3;且
    m为1或2。
  2. 根据权利要求1所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R a选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、-OS(O) mR 6、芳基和杂芳基,其中所述的烷基、烷氧基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代;R 6如权利要求1中所定义。
  3. 根据权利要求1或2中所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其为通式(II)所示的化合物:
    Figure PCTCN2019074324-appb-100002
    其中:
    R b相同或不同,且各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基;
    p为0、1、2或3;
    环A、环B、L、Y、R a、R 2、R 3和s如权利要求1中所定义。
  4. 根据权利要求1~3中任一项所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的环A和环B相同或不同,且各自独立地为芳基或杂芳基,优选选自苯基、吡啶基、呋喃基或噻吩基。
  5. 根据权利要求1~4中任一项所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其为通式(III)所示的化合物:
    Figure PCTCN2019074324-appb-100003
    其中:
    G选自C、CH或N;
    L、Y、R a、R b、R 2、R 3、p和s如权利要求1中所定义。
  6. 根据权利要求1~5中任一项所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的-Y-为共价键或-CH 2-。
  7. 根据权利要求1~6中任一项所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其为通式(IV)所示的化合物:
    Figure PCTCN2019074324-appb-100004
    其中:
    G选自C、CH或N;
    L、R a、R b、R 2、R 3、p和s如权利要求1中所定义。
  8. 根据权利要求1~7中任一项所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的L选自CR 4R 5、O、NH或S;R 4和R 5为氢原子;或者R 4和R 5一起形成=NH。
  9. 根据权利要求1~8中任一项所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 2选自氢原子、卤素或烷基。
  10. 根据权利要求1~9中任一项所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R 3选自氢原子、卤素或烷基。
  11. 根据权利要求1和3-10中任一项所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R a选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟基、羟烷基、杂环基、杂环基烷基、杂环基氧基、-OR c、-COR 9、-COOR 9和-OS(O) mR 6;其中所述的烷基、烷氧基、杂环基、杂环基烷基和杂环基氧基各自独立地任选被选自卤素、烷 基、烷氧基和环烷基中的一个或多个取代基所取代;R 6为烷基或氨基;R c选自氢原子、烷基、卤代烷基、羟烷基、环烷基和杂环基,其中所述的烷基、环烷基和杂环基各自独立地任选被选自烷基、烷氧基、羟基、羟烷基、环烷基和杂环基中的一个或多个取代基所取代;R 9为烷基。
  12. 根据权利要求3~11中任一项所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其中所述的R b选自氢原子、卤素或烷基;p为0、1或2。
  13. 根据权利要求1~6中任一项所述的通式(I)所示的化合物,或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,其选自以下任一化合物:
    Figure PCTCN2019074324-appb-100005
    Figure PCTCN2019074324-appb-100006
    Figure PCTCN2019074324-appb-100007
  14. 一种通式(IA)所示的化合物:
    Figure PCTCN2019074324-appb-100008
    或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,
    其中:
    R w为氨基保护基,优选为叔丁基或叔丁氧羰基;
    R 7为氢原子或R w
    L选自CR 4R 5、O、NH或S;
    环A和环B相同或不同,且各自独立地选自环烷基、杂环基、芳基和杂芳基;
    R 1相同或不同,且各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基、杂芳基和Y-R a
    Y选自共价键或亚烷基;
    R a选自氢原子、卤素、烷基、烷氧基、卤代烷基、卤代烷氧基、羟基、羟烷基、氰 基、氨基、硝基、环烷基、杂环基、杂环基烷基、杂环基氧基、-OR c、-COR 9、-COOR 9、-OS(O) mR 6、芳基和杂芳基;其中所述的烷基、烷氧基、环烷基、杂环基、杂环基烷基、杂环基氧基、芳基和杂芳基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、杂环基氧基、芳基、杂芳基和-OS(O) mR 6中的一个或多个取代基所取代;
    R c选自氢原子、烷基、卤代烷基、羟烷基、环烷基和杂环基,其中所述的烷基、环烷基和杂环基各自独立地任选被选自卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基和杂环基中的一个或多个取代基所取代;
    R 2选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基;
    R 3选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基、羟烷基、氰基、氨基、硝基、环烷基、杂环基、芳基和杂芳基;
    R 4和R 5相同或不同,且各自独立地选自氢原子、卤素、烷基、烷氧基、卤代烷基、羟基和羟烷基;
    或者R 4和R 5一起形成=NH或=O;
    R 6选自氢原子、卤素、烷氧基、卤代烷氧基、氨基、环烷基、杂环基、芳基和杂芳基;
    R 9选自氢原子、烷基、卤代烷基、烷氧基、卤代烷氧基、氨基、环烷基、杂环基、芳基和杂芳基;
    n为0、1、2、3或4;
    s为0、1、2或3;且
    m为1或2。
  15. 根据权利要求14所述的通式(IA)所示的化合物,其选自以下任一化合物:
    Figure PCTCN2019074324-appb-100009
    Figure PCTCN2019074324-appb-100010
    Figure PCTCN2019074324-appb-100011
  16. 一种制备根据权利要求1所述的通式(I)所示的化合物的方法,该方法包括:
    Figure PCTCN2019074324-appb-100012
    通式(IA)的化合物脱去氨基保护基,得到通式(I)的化合物,
    其中:
    R w为氨基保护基,优选为叔丁基或叔丁氧羰基;
    R 7为氢原子或R w
    环A、环B、L、R 1~R 3、n和s如权利要求1中所定义。
  17. 一种药物组合物,所述药物组合物含有治疗有效量的根据权利要求1~13中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式或其可药用的盐,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
  18. 根据权利要求1~13中任一项所述的通式(I)所示的化合物或其互变异构体、内消 旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用的盐或根据权利要求17所述的药物组合物在制备用于抑制A 2a受体的药物中的用途。
  19. 根据权利要求1~13中任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用的盐或根据权利要求17所述的药物组合物在制备用于治疗通过对A 2a受体的抑制而改善的病况或病症的药物中的用途。
  20. 根据权利要求19所述的用途,其中所述的通过对A 2a受体的抑制而改善的病况或病症选自肿瘤、抑郁症、认知功能病症、神经退行性病症、注意力相关病症、锥体外症候群、异常运动障碍、肝硬化、肝纤维化、脂肪肝、皮肤纤维化、睡眠障碍、中风、脑损伤、神经炎症和成瘾行为;优选为肿瘤。
  21. 根据权利要求20所述的用途,其中所述的肿瘤选自黑色素瘤、脑瘤、食管癌、胃癌、肝癌、胰腺癌、结肠直肠癌、肺癌、肾癌、乳腺癌、卵巢癌、***癌、皮肤癌、神经母细胞瘤、肉瘤、骨软骨瘤、骨瘤、骨肉瘤、***瘤、睾丸肿瘤、子宫癌、头颈肿瘤、多发性骨髓瘤、恶性淋巴瘤、真性红细胞增多症、白血病、甲状腺肿瘤、输尿管肿瘤、膀胱癌、胆囊癌、胆管癌、绒毛膜上皮癌和儿科肿瘤。
PCT/CN2019/074324 2018-02-06 2019-02-01 吡唑并[1,5-a][1,3,5]三嗪-2-胺类衍生物、其制备方法及其在医药上的应用 WO2019154294A1 (zh)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CA3089159A CA3089159A1 (en) 2018-02-06 2019-02-01 Pyrazolo[1,5-a][1,3,5]triazine-2-amine derivative, preparation method therefor and medical use thereof
EP19752007.5A EP3750891A4 (en) 2018-02-06 2019-02-01 PYRAZOLO [1,5-A] [1,3,5] TRIAZINE-2-AMINE DERIVATIVE, METHOD OF PREPARATION AND MEDICAL USE
JP2020564306A JP2021512959A (ja) 2018-02-06 2019-02-01 ピラゾロ[1,5−a][1,3,5]トリアジン−2−アミン誘導体、その製造法、およびその医薬用途
CN201980006595.2A CN111511745B (zh) 2018-02-06 2019-02-01 吡唑并[1,5-a][1,3,5]三嗪-2-胺类衍生物、其制备方法及其应用
US16/967,370 US20210032253A1 (en) 2018-02-06 2019-02-01 Pyrazolo[1,5-a][1,3,5]triazine-2-amine derivative, preparation method therefor and medical use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810118455.1 2018-02-06
CN201810118455 2018-02-06

Publications (1)

Publication Number Publication Date
WO2019154294A1 true WO2019154294A1 (zh) 2019-08-15

Family

ID=67548154

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/074324 WO2019154294A1 (zh) 2018-02-06 2019-02-01 吡唑并[1,5-a][1,3,5]三嗪-2-胺类衍生物、其制备方法及其在医药上的应用

Country Status (7)

Country Link
US (1) US20210032253A1 (zh)
EP (1) EP3750891A4 (zh)
JP (1) JP2021512959A (zh)
CN (1) CN111511745B (zh)
CA (1) CA3089159A1 (zh)
TW (1) TWI701251B (zh)
WO (1) WO2019154294A1 (zh)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10793561B2 (en) 2017-07-18 2020-10-06 Nuvation Bio Inc. 1,8-naphthyridinone compounds and uses thereof
CN112028891A (zh) * 2019-07-30 2020-12-04 杭州阿诺生物医药科技有限公司 腺苷受体拮抗剂
US11028058B2 (en) 2017-07-18 2021-06-08 Nuvation Bio Inc. Heterocyclic compounds as adenosine antagonists
US11254670B2 (en) 2019-01-18 2022-02-22 Nuvation Bio Inc. 1,8-naphthyridinone compounds and uses thereof
US11306071B2 (en) 2019-01-18 2022-04-19 Nuvation Bio Inc. Heterocyclic compounds as adenosine antagonists

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000059907A2 (en) * 1999-04-06 2000-10-12 Du Pont Pharmaceuticals Company Pyrazolotriazines as crf antagonists
CN1278819A (zh) * 1997-09-02 2001-01-03 杜邦药品公司 作为促肾上腺皮质素释放激素拮抗剂、可用于治疗cns障碍和紧张相关性疾病的杂环基取代的环稠合吡啶和嘧啶
WO2004011464A2 (fr) * 2002-07-26 2004-02-05 Greenpharma Nouvelles pyrazolo[1,5-a]-1,3,5-triazines substituees et leurs analogiques, compositions pharmaceutiques les contenant, utilisation a titre de medicament et procedes pour leur preparation
WO2005121111A1 (ja) 2004-06-14 2005-12-22 Mitsui Chemicals, Inc. 3-アミノメチルテトラヒドロフラン誘導体の製造方法
WO2007116106A1 (es) 2006-04-12 2007-10-18 Palobiofarma, S.L. Nuevos compuestos como antagonistas de los receptores a1 de adenosina
WO2009080197A1 (de) 2007-12-20 2009-07-02 Bayer Schering Pharma Aktiengesellschaft Substituierte pyrrolo[2, 3-b]- und pyrazolo[3, 4-b] pyridine als adenosin rezeptor liganden
WO2009156737A1 (en) 2008-06-25 2009-12-30 Vernalis (R&D) Limited Triazolo [4, 5-d] pyramidine derivatives and their use as purine receptor antagonists
WO2011095625A1 (en) 2010-02-05 2011-08-11 Heptares Therapeutics Limited 1,2,4-triazine-4-amine derivatives
WO2011159302A1 (en) 2010-06-17 2011-12-22 Janssen Pharmaceutica Nv Arylindenopyrimidines for treating neurodegenerative and movement disorders while minimizing cardiac toxicity
US20120172448A1 (en) 2010-12-07 2012-07-05 Bayer Pharma Aktiengesellschaft Substituted 1-benzylcycloalkylcarboxylic acids and the use thereof
WO2013071697A1 (en) 2011-11-14 2013-05-23 Sunshine Lake Pharma Co., Ltd. Aminoquinazoline derivatives and their salts and methods of use
WO2014101373A1 (en) 2012-12-28 2014-07-03 Merck Sharp & Dohme Corp. Heterobicyclo-substituted-[1,2,4]triazolo[1,5-c]quinazolin-5-amine compounds for treatment of central nervous system disorder
WO2015031221A1 (en) 2013-08-29 2015-03-05 Merck Sharp & Dohme Corp. 2,2-difluorodioxolo a2a receptor antagonists
WO2017055305A1 (en) * 2015-09-30 2017-04-06 Ucb Biopharma Sprl Fused pyrazole derivatives as kinase inhibitors

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0459702A1 (en) * 1990-05-29 1991-12-04 Zeneca Limited Azole Derivatives
US5356894A (en) * 1990-05-29 1994-10-18 Rodney Peter W Morpholinyl substituted [1,2,4]-triazolo[1,5-a]triazine as antagonist
PT915880E (pt) * 1996-07-24 2007-12-31 Bristol Myers Squibb Pharma Co Azol triazinas e pirimidinas
US7329658B2 (en) * 2003-02-06 2008-02-12 Pfizer Inc Cannabinoid receptor ligands and uses thereof
WO2004110454A1 (ja) * 2003-06-13 2004-12-23 Ishihara Sangyo Kaisha, Ltd. アデノシンA2a受容体アゴニストの投与が必要な疾患を治療又は予防するための組成物
EP3594216B1 (en) * 2017-03-07 2022-04-06 Guangzhou Maxinovel Pharmaceuticals Co., Ltd. Aminopyrimidine five-membered heterocyclic compound, and intermediate, preparation method, pharmaceutical composition and application thereof

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1278819A (zh) * 1997-09-02 2001-01-03 杜邦药品公司 作为促肾上腺皮质素释放激素拮抗剂、可用于治疗cns障碍和紧张相关性疾病的杂环基取代的环稠合吡啶和嘧啶
WO2000059907A2 (en) * 1999-04-06 2000-10-12 Du Pont Pharmaceuticals Company Pyrazolotriazines as crf antagonists
WO2004011464A2 (fr) * 2002-07-26 2004-02-05 Greenpharma Nouvelles pyrazolo[1,5-a]-1,3,5-triazines substituees et leurs analogiques, compositions pharmaceutiques les contenant, utilisation a titre de medicament et procedes pour leur preparation
WO2005121111A1 (ja) 2004-06-14 2005-12-22 Mitsui Chemicals, Inc. 3-アミノメチルテトラヒドロフラン誘導体の製造方法
WO2007116106A1 (es) 2006-04-12 2007-10-18 Palobiofarma, S.L. Nuevos compuestos como antagonistas de los receptores a1 de adenosina
WO2009080197A1 (de) 2007-12-20 2009-07-02 Bayer Schering Pharma Aktiengesellschaft Substituierte pyrrolo[2, 3-b]- und pyrazolo[3, 4-b] pyridine als adenosin rezeptor liganden
WO2009156737A1 (en) 2008-06-25 2009-12-30 Vernalis (R&D) Limited Triazolo [4, 5-d] pyramidine derivatives and their use as purine receptor antagonists
WO2011095625A1 (en) 2010-02-05 2011-08-11 Heptares Therapeutics Limited 1,2,4-triazine-4-amine derivatives
WO2011159302A1 (en) 2010-06-17 2011-12-22 Janssen Pharmaceutica Nv Arylindenopyrimidines for treating neurodegenerative and movement disorders while minimizing cardiac toxicity
US20120172448A1 (en) 2010-12-07 2012-07-05 Bayer Pharma Aktiengesellschaft Substituted 1-benzylcycloalkylcarboxylic acids and the use thereof
WO2013071697A1 (en) 2011-11-14 2013-05-23 Sunshine Lake Pharma Co., Ltd. Aminoquinazoline derivatives and their salts and methods of use
WO2014101373A1 (en) 2012-12-28 2014-07-03 Merck Sharp & Dohme Corp. Heterobicyclo-substituted-[1,2,4]triazolo[1,5-c]quinazolin-5-amine compounds for treatment of central nervous system disorder
WO2015031221A1 (en) 2013-08-29 2015-03-05 Merck Sharp & Dohme Corp. 2,2-difluorodioxolo a2a receptor antagonists
WO2017055305A1 (en) * 2015-09-30 2017-04-06 Ucb Biopharma Sprl Fused pyrazole derivatives as kinase inhibitors

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
ANGEWANDTE CHEMIE-INTERNATIONAL EDITION, vol. 47, no. 24, 2008, pages 4512 - 4515
BASIC & CLINICAL PHARMACOLOGY & TOXICOLOGY, vol. 109, no. 3, 2011, pages 203 - 7
CHINESE PHARMACOLOGICAL BULLETIN, vol. 24, no. 5, 2008, pages 573 - 576
CLIN. NEUROPHARMACOL., vol. 33, 2010, pages 55 - 60
EXPERT OPINION ON THERAPEUTIC PATENTS, vol. 17, 2007, pages 979 - 991
GESSI S ET AL., PHARMACOL. THER., vol. 117, no. 1, 2008, pages 123 - 140
J. NEUROSCI., vol. 30, no. 48, 2010, pages 16284 - 16292
JENNER P., J NEUROL., vol. 24, no. 12, 2000, pages 1143 - 50
JOURNAL OF CHEMICAL RESEARCH, vol. 40, no. 10, 2016, pages 594 - 596
JOURNAL OF MEDICINAL CHEMISTRY, vol. 53, no. 23, 2010, pages 8421 - 8439
JOURNAL OF ORGANIC CHEMISTRY, vol. 63, no. 12, 1998, pages 3884 - 3894
LOKSHIN, CANCER RES., vol. 66, no. 15, 1 August 2006 (2006-08-01), pages 7758 - 65
MOV. DISORDERS, vol. 25, no. 2, 2010, pages S305
PARKINSONISN RELAT. DISORD., vol. 16, no. 6, 2010, pages 423 - 426
See also references of EP3750891A4
SYNTHESIS, vol. 7, 1981, pages 554 - 557
SYNTHETIC COMMUNICATIONS, vol. 38, no. 15, 2008, pages 2638 - 2645
TETRAHEDRON LETTERS, vol. 55, no. 41, 2014, pages 5591 - 5594
TRENDS IN NEUROSCI., vol. 29, no. 11, 2006, pages 647 - 654

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10793561B2 (en) 2017-07-18 2020-10-06 Nuvation Bio Inc. 1,8-naphthyridinone compounds and uses thereof
US11028058B2 (en) 2017-07-18 2021-06-08 Nuvation Bio Inc. Heterocyclic compounds as adenosine antagonists
US11254670B2 (en) 2019-01-18 2022-02-22 Nuvation Bio Inc. 1,8-naphthyridinone compounds and uses thereof
US11306071B2 (en) 2019-01-18 2022-04-19 Nuvation Bio Inc. Heterocyclic compounds as adenosine antagonists
CN112028891A (zh) * 2019-07-30 2020-12-04 杭州阿诺生物医药科技有限公司 腺苷受体拮抗剂
WO2021018173A1 (zh) * 2019-07-30 2021-02-04 杭州阿诺生物医药科技有限公司 腺苷受体拮抗剂
CN112608316A (zh) * 2019-07-30 2021-04-06 杭州阿诺生物医药科技有限公司 一种吡唑并三嗪类腺苷受体拮抗剂
CN112028891B (zh) * 2019-07-30 2022-07-05 厦门宝太生物科技股份有限公司 腺苷受体拮抗剂

Also Published As

Publication number Publication date
EP3750891A4 (en) 2021-08-11
CN111511745A (zh) 2020-08-07
TWI701251B (zh) 2020-08-11
CN111511745B (zh) 2022-05-27
JP2021512959A (ja) 2021-05-20
CA3089159A1 (en) 2019-08-15
US20210032253A1 (en) 2021-02-04
TW201934554A (zh) 2019-09-01
EP3750891A1 (en) 2020-12-16

Similar Documents

Publication Publication Date Title
CN109963854B (zh) 杂芳基并[4,3-c]嘧啶-5-胺类衍生物、其制备方法及其在医药上的应用
TWI701251B (zh) 吡唑并[1,5-a][1,3,5]三-2-胺類衍生物、其製備方法及其在醫藥上的應用
TWI458723B (zh) 1,2-雙取代雜環化合物
TW202115076A (zh) 嘧啶并五員氮雜環類衍生物、其製備方法及其在醫藥上的應用
WO2021203768A1 (zh) 嘧啶并二环类衍生物、其制备方法及其在医药上的应用
CN110016021B (zh) 一种免疫调节剂
WO2016169421A1 (zh) 咪唑并异吲哚类衍生物、其制备方法及其在医药上的应用
WO2020011246A1 (zh) 含苯环的化合物、其制备方法及应用
WO2018130184A1 (zh) 1,2,4-三嗪-3-胺类衍生物、其制备方法及其在医药上的应用
WO2021121210A1 (zh) 并环类衍生物、其制备方法及其在医药上的应用
TW202102505A (zh) 吡咯并雜環類衍生物、其製備方法及其在醫藥上的應用
WO2020168927A1 (zh) 含氮并环化合物、其制备方法及用途
CN111247152A (zh) 作为激酶抑制剂的环状亚氨基嘧啶衍生物
WO2022017365A1 (zh) 含硫异吲哚啉类衍生物、其制备方法及其在医药上的应用
WO2020011220A1 (zh) 杂芳基类衍生物、其制备方法及其在医药上的应用
WO2022166860A1 (zh) Pim激酶抑制剂
CN108467386B (zh) 稠杂芳基取代的1,2,4-三嗪-3-胺类衍生物、其制备方法及其在医药上的应用
WO2020259703A1 (zh) 一种吡唑酮并嘧啶类化合物、其制备方法及应用
WO2023125935A1 (zh) 苯并哌啶衍生物调节剂、其制备方法和应用
CN112574212B (zh) 一种嘧啶并五元氮杂环类衍生物、其制备方法及其在医药上的应用
WO2022222911A1 (zh) 嘧啶酮化合物及其用途
CN112996783B (zh) 2-氨基嘧啶类衍生物、其制备方法及其在医药上的应用
CN114072399A (zh) 芳甲酰胺类化合物及其制备方法和医药用途
WO2021114691A1 (zh) 含氮并环化合物、其制备方法及用途
KR20210141921A (ko) 펜타미딘 유사체 및 그의 용도

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: 19752007

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3089159

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2020564306

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019752007

Country of ref document: EP

Effective date: 20200907