CN113549068A - Novel imidazopyridine compound, preparation method and medical application thereof - Google Patents

Novel imidazopyridine compound, preparation method and medical application thereof Download PDF

Info

Publication number
CN113549068A
CN113549068A CN202110463162.9A CN202110463162A CN113549068A CN 113549068 A CN113549068 A CN 113549068A CN 202110463162 A CN202110463162 A CN 202110463162A CN 113549068 A CN113549068 A CN 113549068A
Authority
CN
China
Prior art keywords
methyl
alkyl
halogen
optionally substituted
formula
Prior art date
Legal status (The legal status 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 status listed.)
Granted
Application number
CN202110463162.9A
Other languages
Chinese (zh)
Other versions
CN113549068B (en
Inventor
李文明
刘宁
刘彪
刘浩淼
余健
邹昊
祝伟
李正涛
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Tuojie Biomedical Technology Co ltd
Original Assignee
Shanghai Tuojie Biomedical Technology Co ltd
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 Shanghai Tuojie Biomedical Technology Co ltd filed Critical Shanghai Tuojie Biomedical Technology Co ltd
Publication of CN113549068A publication Critical patent/CN113549068A/en
Application granted granted Critical
Publication of CN113549068B publication Critical patent/CN113549068B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/14Antitussive agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Pulmonology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The disclosure relates to a novel imidazopyridine compound, a preparation method and a medical application thereof. In particular, the compounds can be used for treating diseases related to P2X3 activity, exemplary compounds are shown in a formula (I), and the definition of groups is described in the specification.

Description

Novel imidazopyridine compound, preparation method and medical application thereof
Technical Field
The disclosure relates to the field of medicine, in particular to novel imidazopyridine compounds, and application and a preparation method of the compounds.
Background
The P2X receptor is a family of cation permeable, ligand-gated ion channels that open in response to binding of extracellular adenosine 5' -triphosphate (ATP). They belong to a larger family of receptors, called purinergic receptors. The P2X receptor is present in a variety of organisms including humans, mice, rats, rabbits, chickens, zebrafish, bullfrogs, flukes and amoebas. Seven independent genes encoding the P2X subunit have been identified and designated P2X1 through P2X 7. The different subunits exhibit different sensitivities to purinergic agonists and antagonists.
The P2X3 receptor has 4 ATP binding sites on a single subunit, consisting of 2 transmembrane domains, N-and C-termini located intracellularly, conserved sequences located in extracellular loop structures. High expression of the P2X3 receptor was found in specific medium and small diameter neurons that were associated with injury information. At the same time, the P2X3 receptor is also involved in the transmission of some non-nociceptive sensations. It has been demonstrated that the P2X3 receptor is involved in bladder sensory function, is a key receptor-mediated bladder sensory signal, and is expressed in the mucosal tissues of the bladder, which are rich in sensory nerve fibers. The expression of P2X3 is also found in sensory nerve fibers of the pharyngeal mucosa, and is involved in the conduction and formation of taste sensation.
When an organism is injured or damaged by nerves, a large amount of ATP is released to activate a presynaptic membrane P2X3 receptor, so that a large amount of Ca2+ flows in, the intracellular calcium concentration is increased to activate Protein Kinase A (PKA) and Protein Kinase C (PKC), the PKA and the PKC are phosphorylated, simultaneously, the release of glutamate is promoted, the NMDA receptor is further activated, the excitatory postsynaptic current is generated, and the central sensitization is caused. Many studies have shown that upregulation of P2X3 receptor expression can lead to the development of pain sensitivity and is involved in pain signaling.
Gefapixant is a P2X3 receptor activity antagonist with IC50 values of-30 nM and 100-250nM for human homologous recombination hP2X3 and hP2X2/3, respectively, and its use for the treatment of chronic cough patients has progressed to clinical stage III.
WO2014117274A discloses a class of P2X3 inhibitors comprising the compound methyl (S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate.
Figure BDA0003034737920000021
Disclosure of Invention
The present disclosure provides compounds of formula (I) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000022
wherein the content of the first and second substances,
R1selected from hydrogen, deuterium, alkyl optionally substituted with halogen or deuterium, halogen, alkoxy optionally substituted with halogen or deuterium, hydroxyalkyl;
R2selected from hydrogen, deuterium, halogen, cyano, cyclopropyl, alkyl optionally substituted with halogen or deuterium;
R3selected from the group consisting of alkyl optionally substituted with halogen or cyano, cycloalkyl, heterocyclyl, alkoxy optionally substituted with halogen, amino optionally substituted with alkyl;
X1is selected from N or CH;
X2oxygen, nitrogen, methylene groups, the methylene groups optionally substituted with halogen, cyclic hydrocarbon groups, alkyl groups;
z is selected from:
a) selected from hydrogen, deuterium, halogen, cyano, amino, hydroxy, optionally halogen-substituted alkyl, sulfone, sulfoxide, sulfonamide, C1-3Optionally halogen-substituted alkoxy; or
b)
Figure BDA0003034737920000023
m is selected from 0, 1 or 2, R4、R5Independently selected from the group consisting of: hydrogen, alkyl, alkoxy and cycloalkyl, or R4And R5Together with the nitrogen to which they are attached form a 4-6 membered heterocyclyl, wherein: said heterocyclyl is optionally substituted with halogen, hydroxy, cyano, haloalkyl or alkyl; and when m is 0, R4And R5A combination of (a) is other than hydrogen and methyl; or
c)
Figure BDA0003034737920000031
Heterocyclyl or heteroaryl, said heterocyclyl or heteroaryl optionally substituted with oxo, halogen, hydroxy, carbonyl, alkyl or cyano, said alkyl optionally substituted with halogen; or
d)
Figure BDA0003034737920000032
Wherein R is6Selected from the group consisting of alkyl, alkoxy, aryl, heteroaryl, cycloalkyl, heterocyclyl, cyanoalkyl, cycloalkyloxy, amino optionally substituted with alkyl;
n is an integer selected from 1 to 4.
In some embodiments, R in the compound of formula (I)1Selected from halogen, C1-3Alkyl radical, C1-3Alkoxy, said alkyl or alkoxy being optionally substituted by one or more halogen or deuterium atoms, R2、R3、X1、X2Z, n are as defined above.
In some embodiments, R in the compound of formula (I)1Selected from halogen, C1-3Alkyl radical, C1-3Alkoxy, said alkyl or alkoxy being optionally substituted with one or more halogen or deuterium atoms; z is selected from hydrogen, deuterium, halogen, cyano, amino, hydroxy, optionally halogen-substituted alkyl, sulfone, sulfoxide, sulfonamide, C1-3Optionally halogen-substituted alkoxy; r2、R3、X1、X2N is as defined above.
In some embodiments, R in the compound of formula (I)1Selected from halogen, C1-3Alkyl radical, C1-3Alkoxy, said alkyl or alkoxy being optionally substituted with one or more halogen or deuterium atoms; z is
Figure BDA0003034737920000033
m is selected from 0, 1 or 2, R4、R5Independently selected from the group consisting of: hydrogen, alkyl, alkoxy and cycloalkyl, or R4And R5Together with the nitrogen to which they are attached form a 4-6 membered heterocyclyl, wherein: said heterocyclyl is optionally substituted with halogen, hydroxy, cyano, haloalkyl or alkyl; and when m is 0, R4And R5A combination of (a) is other than hydrogen and methyl; r2、R3、X1、X2N is as defined above.
In some embodiments of the present invention, the substrate is,in the compound of formula (I) R1Selected from halogen, C1-3Alkyl radical, C1-3Alkoxy, said alkyl or alkoxy being optionally substituted with one or more halogen or deuterium atoms; z is selected from
Figure BDA0003034737920000034
Heterocyclyl or heteroaryl, said heterocyclyl or heteroaryl optionally substituted with oxo, halogen, hydroxy, carbonyl, alkyl or cyano, said alkyl optionally substituted with halogen; r2、R3、X1、X2N is as defined above.
In some embodiments, R in the compound of formula (I)1Selected from halogen, C1-3Alkyl radical, C1-3Alkoxy, said alkyl or alkoxy being optionally substituted with one or more halogen or deuterium atoms; z is selected from
Figure BDA0003034737920000035
Figure BDA0003034737920000036
Wherein R is6Selected from the group consisting of alkyl, alkoxy, aryl, heteroaryl, cycloalkyl, heterocyclyl, cyanoalkyl, cycloalkyloxy, amino optionally substituted with alkyl; r2、R3、X1、X2N is as defined above.
In some embodiments, Z in the compounds of formula (I) may be selected from:
a) selected from the group consisting of halogen, cyano, amino, sulfone, sulfonamide, alkyl substituted with one or more halogen atoms; or
b)
Figure BDA0003034737920000041
m is selected from 0, 1 or 2, R4、R5Independently selected from the group consisting of: hydrogen, alkyl, alkoxy and cycloalkyl, or R4And R5Together with the nitrogen to which they are attached form a 4-6 membered heterocyclyl, which heterocyclyl is optionally substituted by halogen, hydroxy or alkyl; and when m isAt 0, R4And R5A combination of (a) is other than hydrogen and methyl; or
c) A 4-6 membered heterocyclyl or heteroaryl group, which heterocyclyl or heteroaryl group is optionally substituted by oxo, halogen, hydroxy, carbonyl, alkyl or cyano, which alkyl group may be optionally substituted by halogen; or
d)
Figure BDA0003034737920000042
Or
e)
Figure BDA0003034737920000043
Wherein R is6Can be selected from C1-3Alkyl radical, C1-3Alkoxy, 5-6 membered aryl or heteroaryl, 3-8 membered cycloalkyl, 3-8 membered heterocyclyl, cyanoalkyl, cycloalkyloxy, amino optionally substituted with alkyl;
R1、R2、R3、X1、X2n is as defined above.
The present disclosure provides compounds of formula (I-1) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000044
wherein R is2aAnd R2bIndependently selected from hydrogen, halogen, C optionally substituted by halogen1-3Alkyl radical, R1、R3、X2Z is defined as the compound of formula (I). In some embodiments, in the compounds of formula (I-1), Z is selected from hydrogen, deuterium, halogen, cyano, amino, hydroxy, alkyl optionally substituted with halogen, sulfone, sulfoxide, sulfonamide, C1-3Optionally halogen-substituted alkoxy; r1、R3、X2As defined above. In some embodiments, in the compounds of formula (I-1), Z is
Figure BDA0003034737920000045
m is selected from 0, 1 or 2, R4、R5Independently selected from the group consisting of: hydrogen, alkyl, alkoxy and cycloalkyl, or R4And R5Together with the nitrogen to which they are attached form a 4-6 membered heterocyclyl, wherein: said heterocyclyl is optionally substituted with halogen, hydroxy, cyano, haloalkyl or alkyl; and when m is 0, R4And R5A combination of (a) is other than hydrogen and methyl; r1、R3、X2As defined above. In some embodiments, in the compounds of formula (I-1), Z is selected from
Figure BDA0003034737920000051
Heterocyclyl or heteroaryl, said heterocyclyl or heteroaryl optionally substituted with oxo, halogen, hydroxy, carbonyl, alkyl or cyano, said alkyl optionally substituted with halogen; r1、R3、X2As defined above. In some embodiments, in the compounds of formula (I-1), Z is selected from
Figure BDA0003034737920000052
Wherein R is6Selected from the group consisting of alkyl, alkoxy, aryl, heteroaryl, cycloalkyl, heterocyclyl, cyanoalkyl, cycloalkyloxy, amino optionally substituted with alkyl; r1、R3、X2As defined above.
The present disclosure provides compounds of formula (I-1) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000053
wherein R is1Selected from halogen, C optionally substituted by halogen or deuterium1-3Alkyl or alkoxy; r2aAnd R2bIndependently selected from hydrogen, halogen, C optionally substituted by halogen1-3An alkyl group; r3、X2Z is defined as the compound of formula (I). In some embodiments, in the compounds of formula (I-1), Z is selected from hydrogen, deuterium, halogen, cyano, amino, hydroxy, alkyl optionally substituted with halogen, sulfoneSulfoxide, sulfonamide, C1-3Optionally halogen-substituted alkoxy; r3、X2As defined above. In some embodiments, in the compounds of formula (I-1), Z is
Figure BDA0003034737920000054
m is selected from 0, 1 or 2, R4、R5Independently selected from the group consisting of: hydrogen, alkyl, alkoxy and cycloalkyl, or R4And R5Together with the nitrogen to which they are attached form a 4-6 membered heterocyclyl, wherein: said heterocyclyl is optionally substituted with halogen, hydroxy, cyano, haloalkyl or alkyl; and when m is 0, R4And R5A combination of (a) is other than hydrogen and methyl; r3、X2As defined above. In some embodiments, in the compounds of formula (I-1), Z is selected from
Figure BDA0003034737920000055
Heterocyclyl or heteroaryl, said heterocyclyl or heteroaryl optionally substituted with oxo, halogen, hydroxy, carbonyl, alkyl or cyano, said alkyl optionally substituted with halogen; r3、X2As defined above. In some embodiments, in the compounds of formula (I-1), Z is selected from
Figure BDA0003034737920000056
Figure BDA0003034737920000061
Wherein R is6Selected from the group consisting of alkyl, alkoxy, aryl, heteroaryl, cycloalkyl, heterocyclyl, cyanoalkyl, cycloalkyloxy, amino optionally substituted with alkyl; r3、X2As defined above.
The present disclosure provides compounds of formula (I-2) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000062
wherein R is3Selected from C optionally substituted by halogen or cyano1-6Alkyl radical, C3-8Cycloalkyl, 4-to 8-membered heterocyclic group having 1 to 3 hetero atoms, alkoxy optionally substituted with halogen or amino optionally substituted with alkyl, R1、R2、X1Z, n are as defined for compounds of formula (I). In some embodiments, in the compounds of formula (I-2), Z is selected from hydrogen, deuterium, halogen, cyano, amino, hydroxy, alkyl optionally substituted with halogen, sulfone, sulfoxide, sulfonamide, C1-3Optionally halogen-substituted alkoxy; r1、R2、X1N is as defined above. In some embodiments, in the compounds of formula (I-2), Z is
Figure BDA0003034737920000063
m is selected from 0, 1 or 2, R4、R5Independently selected from the group consisting of: hydrogen, alkyl, alkoxy and cycloalkyl, or R4And R5Together with the nitrogen to which they are attached form a 4-6 membered heterocyclyl, wherein: said heterocyclyl is optionally substituted with halogen, hydroxy, cyano, haloalkyl or alkyl; and when m is 0, R4And R5A combination of (a) is other than hydrogen and methyl; r1、R2、X1N is as defined above. In some embodiments, in the compounds of formula (I-2), Z is selected from
Figure BDA0003034737920000064
Heterocyclyl or heteroaryl, said heterocyclyl or heteroaryl optionally substituted with oxo, halogen, hydroxy, carbonyl, alkyl or cyano, said alkyl optionally substituted with halogen; r1、R2、X1N is as defined above. In some embodiments, in the compounds of formula (I-2), Z is selected from
Figure BDA0003034737920000065
Wherein R is6Selected from alkyl, alkoxy, aryl, heteroaryl, cycloalkyl, heterocyclyl, cyanoalkyl, cycloalkylAn alkoxy group, an amino group optionally substituted with an alkyl group; r1、R2、X1N is as defined above.
The present disclosure provides compounds of formula (I-2) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000071
wherein R is1Selected from halogen, C1-3Alkyl radical, C1-3Alkoxy, said alkyl or alkoxy being optionally substituted with one or more halogen or deuterium atoms; r3Selected from C optionally substituted by halogen or cyano1-6Alkyl radical, C3-8Cycloalkyl, 4-to 8-membered heterocyclic group containing 1 to 3 hetero atoms, alkoxy optionally substituted with halogen or amino optionally substituted with alkyl; r2、X1Z, n are as defined for compounds of formula (I). In some embodiments, in the compounds of formula (I-2), Z is selected from hydrogen, deuterium, halogen, cyano, amino, hydroxy, alkyl optionally substituted with halogen, sulfone, sulfoxide, sulfonamide, C1-3Optionally halogen-substituted alkoxy; r2、X1N is as defined above. In some embodiments, in the compounds of formula (I-2), Z is
Figure BDA0003034737920000072
m is selected from 0, 1 or 2, R4、R5Independently selected from the group consisting of: hydrogen, alkyl, alkoxy and cycloalkyl, or R4And R5Together with the nitrogen to which they are attached form a 4-6 membered heterocyclyl, wherein: said heterocyclyl is optionally substituted with halogen, hydroxy, cyano, haloalkyl or alkyl; and when m is 0, R4And R5A combination of (a) is other than hydrogen and methyl; r2、X1N is as defined above. In some embodiments, in the compounds of formula (I-2), Z is selected from
Figure BDA0003034737920000073
Heterocyclic ringsA heterocyclyl or heteroaryl group, said heterocyclyl or heteroaryl group optionally being substituted by oxo, halogen, hydroxy, carbonyl, alkyl or cyano, said alkyl group optionally being substituted by halogen; r2、X1N is as defined above. In some embodiments, in the compounds of formula (I-2), Z is selected from
Figure BDA0003034737920000074
Wherein R is6Selected from the group consisting of alkyl, alkoxy, aryl, heteroaryl, cycloalkyl, heterocyclyl, cyanoalkyl, cycloalkyloxy, amino optionally substituted with alkyl; r2、X1N is as defined above.
The present disclosure provides compounds of formula (I-3) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000075
Figure BDA0003034737920000081
wherein R is2aAnd R2bIndependently selected from hydrogen, halogen, C optionally substituted by halogen1-3An alkyl group; r3Selected from C optionally substituted by halogen or cyano1-6Alkyl radical, C3-8Cycloalkyl, 4-to 8-membered heterocyclic group containing 1 to 3 hetero atoms, alkoxy optionally substituted with halogen or amino optionally substituted with alkyl; r1And Z is as defined for compounds of formula (I). In some embodiments, in the compounds of formula (I-3), Z is selected from hydrogen, deuterium, halogen, cyano, amino, hydroxy, alkyl optionally substituted with halogen, sulfone, sulfoxide, sulfonamide, C1-3Optionally halogen-substituted alkoxy; r1As defined above. In some embodiments, in the compounds of formula (I-3), Z is
Figure BDA0003034737920000082
m is selected from 0, 1 or 2, R4、R5Independently selected from the group consisting of: hydrogen, alkyl, alkoxy and cycloalkyl, or R4And R5Together with the nitrogen to which they are attached form a 4-6 membered heterocyclyl, wherein: said heterocyclyl is optionally substituted with halogen, hydroxy, cyano, haloalkyl or alkyl; and when m is 0, R4And R5A combination of (a) is other than hydrogen and methyl; r1As defined above. In some embodiments, in the compounds of formula (I-3), Z is selected from
Figure BDA0003034737920000083
Heterocyclyl or heteroaryl, said heterocyclyl or heteroaryl optionally substituted with oxo, halogen, hydroxy, carbonyl, alkyl or cyano, said alkyl optionally substituted with halogen; r1As defined above. In some embodiments, in the compounds of formula (I-3), Z is selected from
Figure BDA0003034737920000084
Figure BDA0003034737920000085
Wherein R is6Selected from the group consisting of alkyl, alkoxy, aryl, heteroaryl, cycloalkyl, heterocyclyl, cyanoalkyl, cycloalkyloxy, amino optionally substituted with alkyl; r1As defined above.
The present disclosure provides compounds of formula (I-3) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000086
wherein R is1Selected from methyl, chlorine, wherein methyl may be substituted by one or more fluorine or deuterium atoms;
R2aand R2bIndependently selected from hydrogen, fluorine atom, chlorine atom, methyl;
R3is selected from C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Fluoroalkyl, cyanomethyl, methylaminoOr a dimethylamino group;
z is selected from:
a) selected from the group consisting of halogen, cyano, amino, sulfone, sulfonamide, alkyl substituted with one or more halogen atoms; or
b)
Figure BDA0003034737920000091
m is selected from 0, 1 or 2, R4、R5Independently selected from the group consisting of: hydrogen, alkyl, alkoxy and cycloalkyl, or R4And R5Together with the nitrogen to which they are attached form a 4-6 membered heterocyclyl, wherein: said heterocyclyl is optionally substituted with halogen, hydroxy or alkyl; and when m is 0, R4And R5A combination of (a) is other than hydrogen and methyl; or
c) A 4-6 membered heterocyclyl or heteroaryl group, which heterocyclyl or heteroaryl group is optionally substituted by oxo, halogen, hydroxy, carbonyl, alkyl or cyano, which alkyl group may be optionally substituted by halogen; or
d)
Figure BDA0003034737920000092
Or
e)
Figure BDA0003034737920000093
Wherein R is6Can be selected from C1-3Alkyl radical, C1-3Alkoxy, 5-6 membered aryl or heteroaryl, 3-8 membered cycloalkyl, 3-8 membered heterocyclyl, cyanoalkyl, cycloalkyloxy, amino optionally substituted with alkyl.
The present disclosure provides compounds of formula (I-3) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000094
wherein R is1Selected from methyl, chlorine, wherein methyl may be substituted by one or more fluorine or deuterium atoms;
R2aand R2bIndependently selected from hydrogen, fluorine atom, chlorine atom, methyl;
R3is selected from C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Fluoroalkyl, cyanomethyl, methylamino, or dimethylamino;
z is selected from the group consisting of halogen, cyano, amino, sulfone, sulfonamide, alkyl substituted with one or more halogen atoms.
The present disclosure provides compounds of formula (I-3) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000101
wherein R is1Selected from methyl, chlorine, wherein methyl may be substituted by one or more fluorine or deuterium atoms;
R2aand R2bIndependently selected from hydrogen, fluorine atom, chlorine atom, methyl;
R3is selected from C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Fluoroalkyl, cyanomethyl, methylamino, or dimethylamino;
z is
Figure BDA0003034737920000102
m is selected from 0, 1 or 2, R4、R5Independently selected from the group consisting of: hydrogen, alkyl, alkoxy and cycloalkyl, or R4And R5Together with the nitrogen to which they are attached form a 4-6 membered heterocyclyl, wherein: said heterocyclyl is optionally substituted with halogen, hydroxy or alkyl; and when m is 0, R4And R5The combination of (a) is not hydrogen and methyl.
The present disclosure provides compounds of formula (I-3) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000103
wherein R is1Selected from methyl and chlorine atoms, wherein the methyl groupMay be substituted by one or more fluorine atoms or deuterium atoms;
R2aand R2bIndependently selected from hydrogen, fluorine atom, chlorine atom, methyl;
R3is selected from C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Fluoroalkyl, cyanomethyl, methylamino, or dimethylamino;
z is a 4-6 membered heterocyclyl or heteroaryl group, which heterocyclyl or heteroaryl group is optionally substituted by oxo, halogen, hydroxy, carbonyl, alkyl or cyano, which alkyl group may optionally be substituted by halogen.
The present disclosure provides compounds of formula (I-3) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000111
wherein R is1Selected from methyl, chlorine, wherein methyl may be substituted by one or more fluorine or deuterium atoms;
R2aand R2bIndependently selected from hydrogen, fluorine atom, chlorine atom, methyl;
R3is selected from C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Fluoroalkyl, cyanomethyl, methylamino, or dimethylamino;
z is
Figure BDA0003034737920000112
The present disclosure provides compounds of formula (I-3) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000113
wherein R is1Selected from methyl, chlorine, wherein methyl may be substituted by one or more fluorine or deuterium atoms;
R2aand R2bIndependently selected from hydrogen, fluorine, chlorine, and AA group;
R3is selected from C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Fluoroalkyl, cyanomethyl, methylamino, or dimethylamino;
z is
Figure BDA0003034737920000114
Wherein R is6Can be selected from C1-3Alkyl, 5-6 membered aryl or heteroaryl, 3-8 membered cycloalkyl, 3-8 membered heterocyclyl, cyanoalkyl, cycloalkyloxy, amino optionally substituted with alkyl.
In some embodiments, Z in the compound of formula (I), the compound of formula (I-1), the compound of formula (I-2), or the compound of formula (I-3) is the following optionally substituted with one or more methyl, fluorine, chlorine, halomethyl, cyano groups:
Figure BDA0003034737920000115
Figure BDA0003034737920000121
the present disclosure provides compounds of formula (I-3) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000122
wherein R is1Selected from methyl, chlorine atoms, wherein methyl is optionally substituted with one or more fluorine or deuterium atoms;
R3is selected from C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Fluoroalkyl, cyanomethyl, methylamino, or dimethylamino;
R2aand R2bIndependently selected from hydrogen, deuterium, halogen, cyano, cyclopropyl, alkyl optionally substituted with halogen or deuterium;
z is selected from the following groups optionally substituted by one or more methyl, fluorine, chlorine, halomethyl, cyano groups:
Figure BDA0003034737920000123
the present disclosure also provides a compound of formula (II), or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000131
wherein Y is selected from
Figure BDA0003034737920000132
R7 is selected from alkyl optionally substituted with halogen or cyano, cycloalkyl, heterocyclyl, alkoxy optionally substituted with halogen, amino optionally substituted with alkyl, R8 is selected from alkyl, aryl optionally substituted with alkyl;
Z1selected from heterocyclic, heteroaryl or
Figure BDA0003034737920000133
Said heterocyclyl or heteroaryl optionally substituted with oxo, halo, hydroxy, carbonyl, alkyl or cyano, said alkyl optionally substituted with halo;
p is selected from 0, 1 or 2;
and the compound of formula (II) does not include Y being
Figure BDA0003034737920000134
And Z is1Is composed of
Figure BDA0003034737920000135
And p is 1.
In some embodiments, in the compound of formula (II), or a pharmaceutically acceptable salt thereof, or an isomer thereof, Z1Selected from the following groups optionally substituted with one or more methyl, fluorine, chlorine, halomethyl, cyano groups:
Figure BDA0003034737920000136
Figure BDA0003034737920000141
the present disclosure also provides a compound of formula (III), or a pharmaceutically acceptable salt thereof, or an isomer thereof:
Figure BDA0003034737920000142
wherein R is1Selected from methyl, methoxy, methyl substituted with one or more fluorine atoms or deuterium atoms;
R2aand R2bIndependently selected from hydrogen, fluorine atom, chlorine atom, methyl;
z is selected from the following optionally substituted with one or more methyl, halomethyl, cyano groups:
Figure BDA0003034737920000143
l is a direct bond, an oxygen atom, -NH-;
R3ais selected from C1-6Alkyl radical, C3-6Cycloalkyl radical, C1-6alkyl-C3-6Cycloalkyl, 4-6 membered heterocyclyl, 5-6 membered heteroaryl, phenyl, 5-12 membered bridged cyclic group, 5-12 membered bridged spiro cyclic group, C as defined above1-6Alkyl radical, C3-6Cycloalkyl radical, C1-6alkyl-C3-6Cycloalkyl, 4-6 membered heterocyclyl, 5-6 membered heteroaryl, phenyl, 5-12 membered bridged cyclic group, 5-12 membered bridged spiro cyclic group may optionally be oxo, or optionally be substituted with one or more methyl, fluoromethyl, fluoro, cyano, hydroxy, C1-3Alkoxy or C1-3Fluoroalkoxy substitution.
The present disclosure also provides a compound as shown below, or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure BDA0003034737920000151
Figure BDA0003034737920000161
Figure BDA0003034737920000171
Figure BDA0003034737920000181
the present disclosure provides a process for preparing a compound of formula (I) or a pharmaceutically acceptable salt, or isomer thereof, comprising the steps of:
Figure BDA0003034737920000182
carrying out cyclization reaction on the compound shown in the formula (I-a) and the compounds shown in the formula (I-b) and the formula (I-c) under the condition of a catalyst to obtain the compound shown in the formula (I);
the catalyst is selected from palladium/carbon, raney nickel, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride, 1' -bis (dibenzylphosphine) dichloropentairon palladium, tris (dibenzylideneacetone) dipalladium or 2-dicyclohexylphosphine-2 ',6' -dimethoxybiphenyl, [1,1' -bis (di-tert-butylphosphino) ferrocene ] dichloropalladium (II), cuprous iodide, cuprous bromide, cuprous chloride and copper trifluoromethanesulfonate;
R1、R2、R3z, n are as defined for compounds of formula (I).
The present disclosure also provides a process for preparing a compound of formula (I) or a pharmaceutically acceptable salt thereof, or an isomer thereof, comprising the steps of:
Figure BDA0003034737920000191
carrying out cyclization reaction on the compound shown in the formula (I-d) and the compound shown in the formula (I-a) to obtain a compound shown in the formula (I-e); halogenating the compound shown in the formula (I-e) to obtain a compound shown in a formula (I-f); carrying out C-C coupling reaction on the compound of the formula (I-f) under an alkaline condition to obtain a compound shown in the formula (I);
the catalyst is selected from palladium/carbon, raney nickel, tetrakis-triphenylphosphine palladium, palladium dichloride, palladium acetate, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride, 1' -bis (dibenzylideophosphonium) dichlorodipentaerythritol palladium, tris (dibenzylideneacetone) dipalladium or 2-dicyclohexylphosphine-2 ',6' -dimethoxybiphenyl, [1,1' -bis (di-tert-butylphosphino) ferrocene ] dichloropalladium (II);
R1、R2、R3z, n are as defined for compounds of formula (I).
The present disclosure also provides a process for preparing a compound of formula (I) or a pharmaceutically acceptable salt thereof, or an isomer thereof, comprising the steps of:
Figure BDA0003034737920000192
the compound shown as the formula (I-a) and the compound shown as the formula (I-g) are subjected to cyclization reaction to obtain the compound shown as the formula (I)
R1, R2, R3, Z, n are as defined for compounds of formula (I);
z' is selected from the group consisting of halogen, sulfonyl and sulfinyl.
The present disclosure also provides a process for preparing the compound or a pharmaceutically acceptable salt thereof, or an isomer thereof. In particular, the preparation is carried out by the method of the examples.
The present disclosure also relates to a pharmaceutical composition comprising a compound described herein, or a pharmaceutically acceptable salt thereof, or an isomer thereof. Typically, the composition further comprises at least one pharmaceutically acceptable carrier, diluent or excipient.
The disclosure also provides the use of the compound or a pharmaceutically acceptable salt thereof, or an isomer thereof, or a pharmaceutical composition comprising the same, in the preparation of a medicament for treating a disease associated with P2X3 activity. In some embodiments, the disease associated with P2X3 activity refers to a disease associated with P2X3 hyperactivity. The compounds of the present disclosure are highly selective for P2X3, which can avoid taste loss. In some embodiments, the compounds of the present disclosure antagonize P2X3 cognate receptors more than 20-fold stronger than antagonizing P2X2/3 heteromeric receptors. In some embodiments, the compounds of the present disclosure antagonize P2X3 cognate receptors more than 30-fold stronger than antagonizing P2X2/3 heteromeric receptors. In some embodiments, the compounds of the present disclosure antagonize P2X3 cognate receptors more than 50-fold stronger than antagonizing P2X2/3 heteromeric receptors. In some embodiments, the compounds of the present disclosure antagonize P2X3 cognate receptors more than 100-fold stronger than antagonizing P2X2/3 heteromeric receptors.
In some embodiments, the compounds of the present disclosure or pharmaceutically acceptable salts thereof, or isomers thereof, or pharmaceutical compositions comprising the same, are useful for the treatment of pain, urinary tract disorders, cough, and the like. Pain may be, for example, chronic pain, neuropathic pain, acute pain, back pain, cancer pain, pain caused by rheumatoid arthritis, migraine, and visceral pain. Urinary tract disorders such as overactive bladder (also known as urinary incontinence), pelvic hypersensitivity, and urethritis.
In some embodiments, a compound of the present disclosure or a pharmaceutically acceptable salt thereof, or an isomer thereof, or a pharmaceutical composition comprising the same, is useful for treating gastrointestinal disorders, including, for example, constipation and functional gastrointestinal disorders (e.g., irritable bowel syndrome or functional dyspepsia); can be used for treating cancer; can be used for the treatment of cardiovascular disorders or for cardioprotection following myocardial infarction; can be used as an immunomodulator, particularly in the treatment of autoimmune diseases (e.g. arthritis), in skin transplantation, organ transplantation, or similar surgical needs, in collagen diseases, in allergies, or as an anti-tumor or anti-viral agent; can be used for treating multiple sclerosis, Parkinson's disease, and Huntington's chorea; can be used for treating depression, anxiety, stress-related disorders (e.g., post-traumatic stress disorder, panic disorder, social phobia, or obsessive compulsive disorder), premature ejaculation, psychosis, traumatic brain injury, stroke, Alzheimer's disease, spinal injury, drug addiction (e.g., treatment of alcohol, nicotine, opioid, or other drug abuse), or sympathetic nervous system disorders (e.g., hypertension); can be used for treating diarrhea; can be used for treating pulmonary disorders such as, for example, asthma, cough, or pulmonary edema.
The compounds of the present disclosure or pharmaceutically acceptable salts thereof, or isomers thereof, can be formulated in a dosage form suitable for oral, buccal, vaginal, rectal, via inhalation, via insufflation, intranasal, sublingual, topical, or parenteral (e.g., intramuscular, subcutaneous, intraperitoneal, intrathoracic, intravenous, epidural, intrathecal, intracerebroventricular, or by injection into the joints) administration.
The term "treatment" refers to the administration of a pharmaceutical composition for prophylactic and/or therapeutic purposes. By "preventing a disease" is meant prophylactically treating a subject who is not yet ill, but is susceptible to, or at risk for, a particular disease. By "treating a disease" is meant treating a patient already suffering from the disease to improve or stabilize the patient's condition.
Any isotopically-labeled (or "radiolabeled") derivative of a compound described in the present disclosure or a pharmaceutically acceptable salt thereof, or an isomer thereof, is encompassed by the present disclosure. Such derivatives are where one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of radionuclides that may be incorporated include2H (also written as "D", i.e. deuterium),3H (also written as "T", i.e. tritium),11C、13C、14C、13N、15N、15O、17O、18O、18F、36Cl、82Br、75Br、76Br、77Br、123I、124I、125I. And131I. the radionuclide used will depend on the amplificationThe specific application of the derivative of the radioactive label. For example, for in vitro receptor labeling and competition assays,3h or14C is often useful. For the application of radiological imaging,11c or18F is often useful. In some embodiments, the radionuclide is3H. In some embodiments, the radionuclide is14C. In some embodiments, the radionuclide is11C. And in some embodiments the radionuclide is18F。
Unless stated to the contrary, the following terms used in the specification and claims have the following meanings.
"halogen" refers to fluorine, chlorine, bromine and iodine.
"alkyl" means a straight or branched chain alkyl group of C1 to C6, such as methyl, ethyl, n-propyl, isopropyl, and the like.
"alkoxy" means C1-C6 alkyloxy groups such as methoxy, ethoxy and the like.
"heterocyclyl" refers to a stable 3-to 18-membered non-aromatic ring group containing 1 to 12 carbon atoms and 1 to 6 heteroatoms selected from nitrogen, oxygen, and sulfur. Unless otherwise specified in the specification, a heterocyclic group may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include a spiro or bridged ring system; and the nitrogen, carbon or sulfur atoms in the heterocyclic group may be optionally oxidized; the nitrogen atoms may optionally be quaternized; and heterocyclic groups may be partially or fully saturated.
"cycloalkyl" refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may contain a spiro or bridged ring system, having 3 to 15 carbon atoms, 3 to 10 carbon atoms, or 5 to 7 carbon atoms, and which is saturated or unsaturated and is linked to the rest of the molecule by a single bond. Monocyclic cycloalkyl groups include non-bridged cycloalkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
"Cyclohydrocarbylene" refers to a divalent cyclic hydrocarbyl group derived from a cyclic hydrocarbyl group. For example,
Figure BDA0003034737920000211
and the like.
"heteroaryl" refers to a 5-to 14-membered ring system group containing hydrogen atoms, 1 to 13 carbon atoms, 1 to 6 heteroatoms selected from nitrogen, oxygen and sulfur, and at least one aromatic ring. Heteroaryl groups can be monocyclic, bicyclic, tricyclic, or tetracyclic systems, which can include spiro ring systems; and the nitrogen, carbon or sulfur atoms in the heteroaryl group may be optionally oxidized; the nitrogen atoms may optionally be quaternized. The aromatic ring of the heteroaryl group does not necessarily contain a heteroatom as long as one ring of the heteroaryl group contains a heteroatom.
The "carbonyl" substituent means a "-CO-" group formed by an oxygen atom and a carbon atom on a cyclic group such as a heterocyclic group, a heteroaryl group or a cycloalkyl group
"Cyclohydrocarbyloxy" means C3-C8 cyclic hydrocarbyloxy derived from said cyclic hydrocarbyl group in a manner analogous to alkoxy derived from alkyl.
"optional" or "optionally" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs or does not. For example "C optionally substituted by halogen or cyano1-6Alkyl "means that halogen or cyano may, but need not, be present, and the description includes the case where alkyl is substituted with halogen or cyano and the case where alkyl is not substituted with halogen and cyano.
Detailed Description
The structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) or/and Mass Spectrometry (MS). NMR shift (. delta.) of 10-6The units in (ppm) are given. NMR was measured using a Bruker AVANCE-400 NMR spectrometer using deuterated dimethyl sulfoxide (DMSO-d)6) Deuterated chloroform (CDCl)3) And deuterated methanol (CD)3OD), internal standard Tetramethylsilane (TMS).
HPLC measurements were performed using Waters ACQUITY ultra high Performance LC, Shimadzu LC-20A systems, Shimadzu LC-2010HT series, or Agilent 1200LC HPLC (ACQUITY UPLC BEH C181.7 μm 2.1X 50mm column, Ultimate XB-C183.0X 150mm column, or XTate C182.1X 30mm column).
MS was determined using a Waters SQD2 mass spectrometer, scanning in positive/negative ion mode with a mass scan range of 100-1200.
Chiral HPLC analytical determination used Chiralpak IC-3100X 4.6mm I.D., 3 μm, Chiralpak AD-3150X 4.6mm I.D., 3 μm, Chiralpak AD-350X 4.6mm I.D., 3 μm, Chiralpak AS-3150X 4.6mm I.D., 3 μm, Chiralpak AS-3100X 4.6mm I.D., 3 μm, Chiralpak OD-3150X 4.6mm I.D., 3 μm, Chiralcel OD-3100X 4.6mm I.D., 3 μm, Chiralcel OJ-H150X 4.6mm I.D., 5 μm, Chiralcel OJ-3150X 4.6mm I.D., 3 μm chromatography columns.
The thin layer chromatography silica gel plate adopts HSGF254 of tobacco yellow sea or GF254 of Qingdao, the specification of the silica gel plate used by Thin Layer Chromatography (TLC) is 0.15 mm-0.2 mm, and the specification of the thin layer chromatography separation and purification product is 0.4 mm-0.5 mm.
The column chromatography generally uses 100-200 mesh, 200-300 mesh or 300-400 mesh silica gel of Tibet Huanghai silica gel as a carrier.
The chiral preparative column used DAICEL CHIRALPAK IC (250 mm. times.30 mm,10 μm) or Phenomenex-Amylose-1(250 mm. times.30 mm,5 μm).
Known starting materials of the present disclosure may be synthesized using or according to methods known in the art, or may be purchased from companies such as ABCR GmbH & co.kg, Acros Organics, Aldrich Chemical Company, nephelo Chemical science and technology (Accela ChemBio Inc), dare chemicals, and the like.
In the examples, the reaction can be carried out in an argon atmosphere or a nitrogen atmosphere, unless otherwise specified.
Argon atmosphere or nitrogen atmosphere means that the reaction flask is connected with an argon or nitrogen balloon having a volume of about 1L.
The hydrogen atmosphere refers to a reaction flask connected with a hydrogen balloon with the volume of about 1L.
The pressure hydrogenation reaction used a hydrogenation apparatus of Parr 3916EKX type and a hydrogen generator of Qinglan QL-500 type or a hydrogenation apparatus of HC2-SS type.
The hydrogenation reaction was usually evacuated and charged with hydrogen and repeated 3 times.
The microwave reaction was carried out using a CEM Discover-S908860 type microwave reactor.
In the examples, the solution means an aqueous solution unless otherwise specified.
In the examples, the reaction temperature is, unless otherwise specified, from 20 ℃ to 30 ℃ at room temperature.
The monitoring of the progress of the reaction in the examples employed Thin Layer Chromatography (TLC), a developing solvent used for the reaction, a system of eluents for column chromatography used for purifying compounds and a developing solvent system for thin layer chromatography including: a: dichloromethane/methanol system, B: n-hexane/ethyl acetate system, C: petroleum ether/ethyl acetate system, D: the volume ratio of petroleum ether/ethyl acetate/methanol and solvent is regulated according to the different polarity of the compound, and a small amount of basic or acidic reagents such as triethylamine, acetic acid and the like can be added for regulation.
Example 1
Methyl (R) -3- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) amino) piperidine-1-carboxylate (1)
Figure BDA0003034737920000231
The method comprises the following steps: tert-butyl (R) -3- ((2- (2, 6-difluoro-4- (methoxycarbonyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) amino) piperidine-1-carboxylate (1a)
Methyl 3, 5-difluoro-4-formylbenzoate (200mg,0.99mmol) and 4-methylpyridin-2-amine (108.0mg,0.99mmol) were dissolved in methanol (4mL), and p-toluenesulfonic acid (51.6mg,0.30mmol) and (3R) -1- [ (tert-butoxycarbonyl) -3-isocyanopiperidine (210.1mg,0.99mmol) were added at room temperature. After 12 hours of reaction at 60 ℃, the reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with (ethyl acetate/petroleum ether) ═ 0-30%) to give compound 1a (300.0mg, yield: 57%).
Step two: methyl (R) -3- ((2- (2, 6-difluoro-4- (methoxycarbonyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) amino) piperidine-1-carboxylate (1b)
Compound 1a (100mg,0.19mmol) was dissolved in methanol (2.0mL) and methanolic hydrochloric acid solution (4M,10.0mL) was added at room temperature. After the reaction mixture was stirred at room temperature for 16 hours, the mixture was concentrated under reduced pressure. After dissolving the crude product in dichloromethane (1mL), N-diisopropylethylamine (76. mu.L, 0.45mmol) was added at 0 ℃ followed by dropwise addition of methyl chloroformate (20. mu.L, 0.25mmol) and the reaction mixture was stirred at 0 ℃ for 2 hours. The reaction was quenched by the addition of water (50mL), the mixture was extracted with ethyl acetate (30 mL. times.3), the organic phases were combined, washed with saturated sodium bicarbonate solution (30mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography eluting with (ethyl acetate/petroleum ether) ═ 0-50%) to give compound 1b (70.0mg, yield: 83%).
Step three: methyl (R) -3- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) amino) piperidine-1-carboxylate (1)
Compound 1b (70mg,0.13mmol) was dissolved in THF (3mL) and water (3mL), NaOH (27.4mg,0.68mmol) was added, and the mixture was stirred at room temperature for 2 hours. The reaction mixture was concentrated under reduced pressure and the resulting residue was dissolved in DMF (0.5mL) and methylamine hydrochloride (9.1mg,0.14mmol), HOBt (24.3mg,0.18mmol), N-methylmorpholine (20. mu.L, 0.18mmol) and EDCI (32. mu.L, 0.18mmol) were added sequentially. After 2 hours of reaction at 15 ℃, the mixture was diluted with water (10mL) and extracted with ethyl acetate (10mL × 3). The organic phases were combined, washed with brine (10mL × 3), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by C18 reverse phase chromatography eluting with 0-40% methanol/water (0.1% ammonia) to give the title compound 1(15.0mg, yield: 25%).
MS(ESI)m/z=458.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.02(d,J=7.2Hz,1H),7.42(d,J=8.0Hz,2H),7.33(s,1H),6.69(d,J=7.2Hz,1H),6.50(br s,1H),3.58(br s,4H),3.09(br s,1H),3.04(d,J=4.8Hz,3H),3.01-2.91(m,1H),2.90-2.73(m,2H),2.41(s,3H),1.74(br s,2H),1.26(br s,2H).
Example 2
Methyl 1- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) piperidine-3-carboxylate (2)
Figure BDA0003034737920000241
The method comprises the following steps: 3, 5-difluoro-N-methyl-4- (7-methylimidazo [1,2-a ] pyridin-2-yl) benzamide (2a)
4- (2-Bromoacetyl) -3, 5-difluoro-N-methylbenzamide (940.0mg,3.22mmol) and 4-methylpyridin-2-amine (382.8mg,3.54mmol) were dissolved in anhydrous DMF (15mL) and stirred at 100 ℃ for 2 h. Water (15mL) was added for dilution, and extraction was performed with ethyl acetate (15 mL. times.3). The organic phases were combined, washed with brine (50mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the resulting crude product was purified by silica gel column chromatography eluting with (methanol/dichloromethane ═ 0-2%) to give compound 2a (291.7mg, yield: 30%).
MS(ESI)m/z=301.9[M+H]+.
1H NMR(400MHz,CDCl3)δ8.04(d,J=6.8Hz,1H),7.93(s,1H),7.44(br d,J=8.0Hz,2H),7.41(s,1H),6.68(br d,J=6.8Hz,1H),6.34(br s,1H),3.04(d,J=4.8Hz,3H),2.42(s,3H).
Step two: methyl 1- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) piperidine-3-carboxylate (2)
Compound 2a (50mg,0.17mmol), piperidine-3-carboxylic acid methyl ester (29.7mg,0.21mmol) and paraformaldehyde (6.2mg,0.21mmol) were dissolved in acetic acid (5mL) and stirred at 60 ℃ for 5 hours under nitrogen. The reaction mixture was concentrated under reduced pressure, and the resulting crude product was dissolved in methanol and adjusted to pH 8-9 with aqueous ammonia (0.5 mL). The crude product was prepared as a liquid phase (Column: Phenomenex Gemini-NX 80: 40 mm: 3 μm, Condition: 32-56% (A: water 0.05% NH)3·H2O+10mM NH4HCO3ACN), flow rate 30mL/min) and lyophilized to give the title compound 2(51.5mg, yield: 66%).
MS(ESI)m/z=457.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.36(d,J=7.2Hz,1H),7.42-7.36(m,3H),7.06(br s,1H),6.69(br d,J=7.2Hz,1H),3.68(s,2H),3.60(s,3H),3.03(d,J=4.8Hz,3H),2.75(br d,J=8.4Hz,1H),2.55-2.46(m,2H),2.44(s,3H),2.32(br s,1H),2.03(br d,J=11.6Hz,1H),1.78(br s,1H),1.55(br d,J=9.2Hz,1H),1.49-1.38(m,1H),1.36-1.22(m,1H).
Example 3
Methyl 4- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) piperazine-1-carboxylate (3)
Figure BDA0003034737920000251
See example 2 for the synthetic procedure for example 3. Wherein piperazine-1-carboxylic acid methyl ester is used to replace piperidine-3-carboxylic acid methyl ester.
MS(ESI)m/z=458.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.36(d,J=7.2Hz,1H),7.43-7.36(m,3H),7.19(br s,1H),6.72(br d,J=6.8Hz,1H),3.70(s,2H),3.66(s,3H),3.40(br s,4H),3.03(d,J=4.8Hz,3H),2.45(s,3H),2.30(br s,4H).
Example 4
Methyl (S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7- (trifluoromethyl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (4)
Figure BDA0003034737920000252
The method comprises the following steps: tert-butyl (S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7- (trifluoromethyl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (4a)
Tert-butyl (2S) -2-ethynylmorpholine-4-carboxylate (200.0mg,0.95mmol), 3, 5-difluoro-4-formyl-N-methylbenzamide (188.5mg,0.95mmol) and 4- (trifluoromethyl) pyridin-2-amine (153.5mg,0.95mmol) were dissolved in toluene (4mL) and cuprous chloride (28.1mg,0.28mmol) and copper triflate (102.7mg,0.28mmol) were added under nitrogen. After the mixture was stirred at 85 ℃ for 5 minutes, N-dimethylacetamide (30. mu.L, 0.28mmol) was added. The reaction mixture was stirred at 85 ℃ for 12 hours and cooled, after which water (15mL) and aqueous ammonia (8mL) were added. The reaction mixture was extracted with ethyl acetate (20mL × 3), the combined organic phases were dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to obtain a crude product, which was purified by silica gel column chromatography (petroleum ether/ethyl acetate ═ 0-50%) to obtain compound 4a (194.7mg, 24%).
MS(ESI)m/z=555.2[M+H]+.
Step two: methyl (S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7- (trifluoromethyl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (4)
Compound 4a (194.7mg,0.35mmol) was dissolved in methanol (2.0mL) and methanolic hydrochloric acid solution (4M,2.0mL) was added at room temperature. After the reaction mixture was stirred at room temperature for 4 hours, the mixture was concentrated under reduced pressure. After dissolving the crude product in dichloromethane (2mL), N-diisopropylethylamine (150. mu.L, 0.94mmol) was added at 0 ℃ followed by dropwise addition of methyl chloroformate (30. mu.L, 0.45mmol) and the reaction mixture was stirred at 0 ℃ for 2 hours. The reaction was quenched by the addition of water (30mL), the mixture was extracted with ethyl acetate (20 mL. times.3), the combined organic phases were washed with saturated sodium bicarbonate solution (30mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was subjected to preparative HPLC (column: Phenomenex Gemini-NX C1880: 30 mm: 3 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O+10mM NH4HCO3) 34% -64%; flow rate:30mL/min) and lyophilized to give the title compound 4(35.1mg, yield: 19.5%).
MS(ESI)m/z=513.1[M+H]+.
1H NMR(400MHz,CDCl3)δ8.48(d,J=7.2Hz,1H),7.96(s,1H),7.45(d,J=8.0Hz,2H),7.02(d,J=8.4Hz,1H),6.34(br d,J=4.4Hz,1H),3.96-3.75(m,3H),3.69(s,3H),3.60(br s,1H),3.38(br t,J=10.8Hz,1H),3.07(d,J=4.4Hz,3H),3.03(br d,J=6.8Hz,2H),2.91(br s,1H),2.64(br t,J=12.0Hz,1H).
Example 5
Methyl (S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7- (difluoromethyl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (5)
Figure BDA0003034737920000271
See example 4 for the synthetic procedure for example 5. Wherein 4- (trifluoromethyl) pyridin-2-amine is replaced by 4- (difluoromethyl) pyridin-2-amine.
MS(ESI)m/z=495.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.85-8.63(m,2H),7.92(br s,1H),7.70(d,J=8.3Hz,2H),7.33-6.98(m,2H),3.82(br d,J=11.8Hz,2H),3.63(br d,J=10.8Hz,2H),3.57(s,3H),3.51(br s,1H),3.28-3.05(m,3H),2.84(d,J=4.5Hz,3H),2.68(s,1H).
Example 6
(S) -4- (3- ((4-acetylmorpholin-2-yl) methyl) -7- (difluoromethyl) imidazo [1,2-a ] pyridin-2-yl) -3, 5-difluoro-N-methylbenzamide (6)
Figure BDA0003034737920000272
See example 4 for the synthetic procedure for example 6. Wherein 4- (difluoromethyl) pyridin-2-amine is substituted for 4- (trifluoromethyl) pyridin-2-amine and acetic anhydride is substituted for methyl chloroformate.
MS(ESI)m/z=479.1[M+H]+.
1H NMR(400MHz,CDCl3)δ8.78-8.62(m,1H),8.38-8.25(m,1H),7.50(br d,J=8.0Hz,2H),7.33(br d,J=7.2Hz,1H),7.01-6.63(m,2H),4.53-4.38(m,1H),3.94-3.80(m,1H),3.72-3.53(m,2H),3.39(br t,J=12.0Hz,1H),3.28-3.16(m,1H),3.10(br d,J=5.6Hz,2H),3.03(d,J=4.8Hz,3H),2.80-2.40(m,1H),2.10-2.06(m,3H).
Example 7
Methyl (S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methoxyimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (7)
Figure BDA0003034737920000281
The synthetic procedure for example 7 is as for step one in example 4. Wherein 4- (methoxy) pyridin-2-amine is substituted for 4- (trifluoromethyl) pyridin-2-amine and (2S) -2-ethynylmorpholine-4-carboxylic acid methyl ester is substituted for (2S) -2-ethynylmorpholine-4-carboxylic acid tert-butyl ester.
MS(ESI)m/z=475.3[M+H]+.
1H NMR(400MHz,CDCl3)δ8.16(br s,1H),7.42(br d,J=7.6Hz,2H),6.99(br s,1H),6.88(br s,1H),6.57(br d,J=7.2Hz,1H),3.88(s,3H),3.80(br s,3H),3.67(s,3H),3.54(br s,1H),3.38(br s,1H),3.03(br d,J=4.4Hz,3H),2.93(br s,3H),2.59(br t,J=11.2Hz,1H).
Example 8
(S) -4- (3- ((4- (2-cyanoacetyl) morpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluoro-N-methylbenzamide (8)
Figure BDA0003034737920000282
The method comprises the following steps: (S) -3, 5-difluoro-N-methyl-4- (7-methyl-3- (morpholin-2-ylmethyl) imidazo [1,2-a ] pyridin-2-yl) benzamide (8a)
Tert-butyl (S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (685.0mg,1.37mmol) was dissolved in dichloromethane (3mL), and 1, 4-dioxane hydrochloride solution (6mL) was added at room temperature to react at 40 ℃ for 1 hour. The reaction solution was concentrated under reduced pressure, and the resulting crude residue was dissolved in water (15mL), adjusted to pH 12 with a saturated NaOH solution, and then extracted with dichloromethane (20 mL. times.3) and chloroform/isopropanol (3:1,20 mL. times.1) in this order. The organic phases were combined, dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure to give compound 8a (497.0mg, 90.7%).
MS(ESI)m/z=401.2[M+H]+.
Step two: (S) -4- (3- ((4- (2-cyanoacetyl) morpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluoro-N-methylbenzamide (8)
After dissolving compound 8a (10.0mg,0.025mmol) in DMF (1mL), cyanoacetic acid (4.2mg,0.05mmol), HATU (19.0mg,0.05mmol) and N, N-diisopropylethylamine (17. mu.L, 0.10mmol) were added at 0 ℃. After 2 hours of reaction at room temperature, the reaction solution was directly purified by C18 reverse phase chromatography eluting with acetonitrile/water (containing 0.05% ammonia water) (5-70%) and lyophilized to obtain the title compound 8(7.3mg, yield: 62.5%).
MS(ESI)m/z=468.1[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.77-8.64(m,1H),8.48-8.37(m,1H),7.65(d,J=8.0Hz,2H),7.35(s,1H),6.84(br d,J=6.8Hz,1H),4.16-3.98(m,2H),3.97-3.76(m,1H),3.72-3.38(m,3H),3.25-2.88(m,4H),2.82(d,J=4.4Hz,3H),2.38(s,3H).
Example 9
(S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -N, N-dimethylmorpholine-4-carboxamide (9)
Figure BDA0003034737920000291
After dissolving compound 8a (10.0mg,0.025mmol) in dichloromethane (1mL), dimethylcarbamoyl chloride (5. mu.L, 0.05mmol) and N, N-diisopropylethylamine (17. mu.L, 0.10mmol) were added at 0 ℃. After reacting at room temperature for 1 hour, the reaction solution was concentrated, and the residue was directly purified by C18 reverse phase chromatography (acetonitrile/water (containing 0.05% aqueous ammonia) ═ 5-70%) and lyophilized to obtain the title compound 9(5.3mg, yield: 45.0%).
MS(ESI)m/z=472.2[M+H]+.
Example 10
Methyl (S) -2- ((7-methyl-2- (4-methyl-6- (methylcarbamoyl) pyridin-3-yl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (10)
Figure BDA0003034737920000292
The method comprises the following steps: 5-vinyl-N, 4-dimethylpyridine-2-carboxamide (10a)
Reacting 5-bromo-N, 4-diMethylpyridine-2-carboxamide (1.00g,4.37mmol) and potassium vinyltrifluoroborate (0.88g,6.55mmol) were dissolved in THF (22.5mL) and water (7.5mL) and Pd (dppf) Cl was added under nitrogen2·CH2Cl2(0.71g,0.87mmol) and K2CO3(0.74mL,13.10 mmol). After refluxing at 80 ℃ for 16 hours under nitrogen, the mixture was suction filtered under reduced pressure and the solid was washed with ethyl acetate (10 mL. times.3). The reaction was quenched with water (20mL), extracted with ethyl acetate (30 mL. times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (ethyl acetate/petroleum ether ═ 0 to 40%) to give compound 10a (410.0mg, yield: 37%).
MS(ESI)m/z=176.9[M+H]+.
1H NMR(400MHz,CDCl3)δ8.54(s,1H),7.99(s,2H),6.89(dd,J=10.8,17.6Hz,1H),5.78(d,J=17.6Hz,1H),5.50(d,J=11.2Hz,1H),3.04(d,J=5.2Hz,3H),2.42(s,3H).
Step two: 5-formyl-N, 4-dimethylpyridine-2-carboxamide (10b)
10a (410.0mg,2.33mmol) was dissolved in dichloromethane (40mL) and ozone was bubbled through the solution at-78 deg.C until the reaction turned blue. And after the reaction is finished, introducing oxygen into the solution until the blue color of the reaction solution disappears. Dimethyl sulfide (4.0mL,54.07mmol) was added and stirred at-78 deg.C for 30 min. After the reaction solution was warmed to room temperature, the reaction solution was concentrated under reduced pressure, and the obtained crude product was purified by silica gel column chromatography (methanol/dichloromethane ═ 0 to 10%) to obtain compound 10b (113.7mg, yield: 19%).
MS(ESI)m/z=178.8[M+H]+.
1H NMR(400MHz,CDCl3)δ10.39(s,1H),8.87(s,1H),8.14(s,1H),3.07(d,J=5.2Hz,3H),2.76(s,3H).
Step three: tert-butyl (S) -2- ((7-methyl-2- (4-methyl-6- (methylcarbamoyl) pyridin-3-yl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (10c)
The synthetic procedure for example 10c is as for step one in example 4. Wherein 4-methylpyridin-2-amine is used for replacing 4- (trifluoromethyl) pyrid-2-amine, and 10b is used for replacing 3, 5-difluoro-4-formyl-N-methylbenzamide.
MS(ESI)m/z=480.1[M+H]+.
1H NMR(400MHz,CDCl3)δ8.46(s,1H),8.14(s,1H),8.10(br d,J=5.6Hz,1H),8.04(br d,J=4.8Hz,1H),7.37(s,1H),6.70(br d,J=6.8Hz,1H),3.77(br d,J=10.4Hz,3H),3.52(br s,1H),3.34(br t,J=10.0Hz,1H),3.05(br d,J=5.2Hz,3H),3.03-2.86(m,4H),2.42(s,3H),2.39(s,3H),1.43-1.41(m,9H).
Step four: methyl (S) -2- ((7-methyl-2- (4-methyl-6- (methylcarbamoyl) pyridin-3-yl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (10)
See example 4, step two for the synthetic procedure of example 10.
MS(ESI)m/z=438.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.46(s,1H),8.16(s,1H),8.10(br d,J=7.2Hz,1H),8.04(br d,J=4.4Hz,1H),7.42(s,1H),6.73(br d,J=6.4Hz,1H),3.80(br d,J=12.0Hz,3H),3.68(s,3H),3.53(br s,1H),3.41-3.30(m,1H),3.07(d,J=4.8Hz,3H),3.04-2.95(m,2H),2.94-2.83(m,1H),2.58-2.49(m,1H),2.45(s,3H),2.41(s,3H).
Example 11
Methyl (S) -2- ((7-methyl-2- (3-methyl-5- (methylcarbamoyl) pyridin-2-yl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (11)
Figure BDA0003034737920000311
The method comprises the following steps: methyl (S) -2- ((2- (5-bromo-3-methylpyridin-2-yl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (11a)
See example 10, step three for the synthetic procedure for example 11 a. Wherein 5-bromo-3-methylpyridine-2-formaldehyde is used instead of 5-formyl-N, 4-dimethylpyridine-2-carboxamide.
MS(ESI)m/z=459.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.03(s,1H),7.98(d,J=5.2Hz,1H),6.64(s,1H),6.55(s,1H),6.48(d,J=4.4Hz,1H),6.04(br d,J=12.0Hz,2H),4.75(br d,J=8.4Hz,1H),4.40-4.18(m,1H),4.06-3.86(m,2H),3.84-3.70(m,4H),3.61-3.51(m,1H),3.33-3.13(m,1H),2.43(s,3H),2.15(s,3H).
Step two: methyl (S) -2- ((7-methyl-2- (3-methyl-5- (methylcarbamoyl) pyridin-2-yl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (11)
Compound 11a (200.0mg,0.43mmol) and methylamine hydrochloride (176.4mg,2.61mmol) are dissolved in toluene (4mL) and Pd (OAc) is added sequentially2(58.6mg,0.26mmol), Xantphos (251.9mg,0.43mmol) and K3PO4(739.3mg,3.48 mmol). The reaction mixture was stirred at 80 ℃ for 16 hours under an atmosphere of carbon monoxide. The mixture was filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by preparative HPLC [ column: phenomenex Gemini-NX 80 x 40mm x3 μm, eluent: (21-51% acetonitrile, water (containing 0.05% NH)3·H2O+10mM NH4HCO3) (ii) a Flow rate:30mL/min]. Freeze-drying gave the title compound 11(24.0mg, yield: 12%).
MS(ESI)m/z=438.3[M+H]+.
1H NMR(400MHz,CDCl3)δ8.58(s,1H),7.90(br d,J=4.8Hz,1H),7.22-6.92(m,1H),6.69(d,J=20.0Hz,2H),6.53(br d,J=4.8Hz,1H),6.31(s,1H),6.19(s,1H),4.87(br d,J=7.6Hz,1H),4.38-4.16(m,1H),4.05-3.88(m,2H),3.88-3.81(m,1H),3.77(s,3H),3.56(br dd,J=9.2,12.8Hz,1H),3.32-3.18(m,1H),3.03(br d,J=4.4Hz,3H),2.44(s,2H),2.20(s,3H).
Example 12
Methyl (S) -2- ((7-methyl-2- (3-methyl-5- (methylcarbamoyl) pyridin-2-yl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (12)
Figure BDA0003034737920000321
The method comprises the following steps: methyl (S) -2- ((2- (6-chloro-2-methylpyridin-3-yl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (12a)
See example 10, step three for the synthetic procedure for example 12 a. Wherein 6-chloro-2-methylpyridine-3-formaldehyde is used instead of 5-formyl-N, 4-dimethylpyridine-2-carboxamide.
MS(ESI)m/z=415.1[M+H]+.
1H NMR(400MHz,CDCl3)δ8.06(d,J=5.6Hz,1H),7.62(d,J=7.6Hz,1H),7.37(s,1H),7.25(d,J=8.0Hz,1H),6.71(d,J=6.0Hz,1H),3.97-3.75(m,3H),3.68(s,3H),3.56-3.45(m,1H),3.43-3.25(m,1H),3.03-2.78(m,3H),2.60-2.51(m,1H),2.49(s,3H),2.44(s,3H).
Step two: methyl (S) -2- ((2- (6- (methoxycarbonylacyl) -2-methylpyridin-3-yl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (12b)
To a solution of compound 12a (150.0mg,0.36mmol) in methanol (3mL) was added dppf (400.8mg,0.72mmol), TEA (0.15mL,1.08mmol) and Pd (OAc)2(81.1mg,0.36mmol) was stirred at 80 ℃ for 16 hours under a carbon monoxide atmosphere. The reaction mixture was filtered, the filtrate was concentrated under reduced pressure, and the obtained crude product was purified by silica gel column chromatography (methanol/dichloromethane ═ 0-4%) to obtain compound 12b (120.0mg, yield: 75%).
MS(ESI)m/z=439.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.12-8.02(m,2H),7.81(d,J=8.0Hz,1H),7.42(s,1H),6.73(d,J=7.2Hz,1H),4.04(s,3H),3.97-3.72(m,3H),3.67(s,3H),3.56-3.45(m,1H),3.42-3.25(m,1H),3.03-2.82(m,3H),2.62(s,3H),2.52(br t,J=11.2Hz,1H),2.44(s,3H).
Step three: (S) -5- (3- ((4- (methoxycarbonylacyl) morpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -6-methyl-o-picolinic acid (12c)
To compound 12b (100.0mg,0.23mmol) in THF (2mL) and H2Adding LiOH & H into O (2mL) solution2O (19.1mg,0.46 mmol). After the reaction mixture was stirred at room temperature for 2.5 hours, the pH of the mixture was adjusted to 3 by adding 2N HCl. The mixture was extracted with chloroform/isopropanol (3/1,5 mL. times.6), the organic phases were combined, dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure to give a compoundSubstance 12c (90.0mg, yield: 65%).
MS(ESI)m/z=425.2[M+H]+.
Step four: methyl (S) -2- ((7-methyl-2- (3-methyl-5- (methylcarbamoyl) pyridin-2-yl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (12)
To a solution of compound 12c (90.0mg,0.21mmol) in DMF (2mL) was added DIEA (0.14mL,0.85mmol) and methylamine in ethanol (0.28mL,2.12 mmol). After the reaction mixture was stirred at 30 ℃ for 16 hours, the reaction was quenched by addition of water (5mL) and extracted with ethyl acetate (4 mL. times.5). The organic phases were combined, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to give a crude product which was purified by preparative HPLC (column: Phenomenex Gemini-NX 80 x 40mm x3 μm; eluent: 23-51% acetonitrile, water (containing 0.05% NH)3·H2O+10mM NH4HCO3) (ii) a Flow rate:30mL/min) to give the title compound 12(28.8mg, yield: 30%).
MS(ESI)m/z=438.5[M+H]+
1H NMR(400MHz,CDCl3)δ8.18-8.12(m,1H),8.12-8.04(m,2H),7.79(br d,J=7.6Hz,1H),7.39(s,1H),6.72(br d,J=6.8Hz,1H),3.96-3.71(m,3H),3.67(s,3H),3.56-3.47(m,1H),3.42-3.28(m,1H),3.08(d,J=5.2Hz,3H),3.03-2.85(m,3H),2.55(s,3H),2.54-2.47(m,1H),2.44(s,3H).
Example 13
Methyl (S) -2- ((2- (4- (azetidine-1-carbonyl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (13)
Figure BDA0003034737920000331
The method comprises the following steps: methyl (S) -2- ((2- (4-bromo-2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (13a)
See example 10, step three, for the synthetic procedure for compound 13 a. Wherein 4-bromo-2, 6-difluorobenzaldehyde is used for replacing 5-formyl-N, 4-dimethylpyridine-2-formamide.
MS(ESI)m/z=480.0[M+H]+.
1H NMR(400MHz,CDCl3)δ8.17(d,J=6.8Hz,1H),7.38(s,1H),7.25-7.18(m,2H),6.67(br d,J=6.8Hz,1H),4.01-3.74(m,3H),3.68(s,3H),3.56(br s,1H),3.44-3.34(m,1H),3.06-2.85(m,3H),2.60(dd,J=11.2,12.8Hz,1H),2.42(s,3H)
Step two: methyl (S) -2- ((2- (4- (methoxycarbonylacyl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (13b)
See example 12, step two for the synthetic procedure for compound 13 b. In which 12a is replaced by 13 a.
MS(ESI)m/z=460.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.17(d,J=6.8Hz,1H),7.74-7.60(m,2H),7.38(s,1H),6.67(br d,J=6.4Hz,1H),3.96(s,3H),3.92-3.70(m,3H),3.66(s,3H),3.55(br s,1H),3.49-3.26(m,1H),3.10-2.77(m,3H),2.57(br dd,J=11.2,12.8Hz,1H),2.40(s,3H).
Step three: (S) -3, 5-difluoro-4- (3- ((4- (methoxycarbonylacyl) morpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) benzoic acid (13c)
See example 12, step three, for the synthesis of compound 13 c. In which 12b is replaced by 13 b.
MS(ESI)m/z=446.1[M+H]+.
Step four: methyl (S) -2- ((2- (4- (azetidine-1-carbonyl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (13)
See example 12 for step four for the synthetic procedure for example 13. In which azetidine hydrochloride was substituted for the ethanolic methylamine solution and 13c for 12 c.
MS(ESI)m/z=485.2[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.42(d,J=7.2Hz,1H),7.43(d,J=7.2Hz,2H),7.35(s,1H),6.84(d,J=7.2Hz,1H),4.40(br t,J=7.6Hz,2H),4.26(t,J=7.6Hz,2H),4.08(br t,J=8.0Hz,2H),3.73(br d,J=12.0Hz,2H),3.63(br s,2H),3.55(s,3H),3.22(br t,J=10.4Hz,1H),3.04(br s,2H),2.38(s,3H),2.30(br dd,J=8.0,16.0Hz,2H).
Example 14
Methyl (S) -2- ((2- (4- (3, 3-difluoroazetidine-1-carbonyl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (14)
Figure BDA0003034737920000341
See example 12 for step four for the synthetic procedure for example 14. Wherein 3, 3-difluoroazetidine hydrochloride is substituted for the ethanolic methylamine and 13c is substituted for 12 c.
MS(ESI)m/z=521.3[M+H]+.
1H NMR(400MHz,CDCl3)δ8.19(d,J=7.2Hz,1H),7.39(s,1H),7.35-7.28(m,2H),6.69(br d,J=6.0Hz,1H),4.60(br t,J=11.6Hz,4H),3.96-3.73(m,3H),3.68(s,3H),3.57(br s,1H),3.44-3.31(m,1H),3.07-2.86(m,3H),2.66-2.55(m,1H),2.43(s,3H).
Example 15
Methyl (S) -2- ((2- (2, 6-difluoro-4- (methoxycarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (15)
Figure BDA0003034737920000351
See example 12 for step four for the synthetic procedure for example 15. Wherein methoxylamine hydrochloride is used for replacing methylamine ethanol solution, and 13c is used for replacing 12 c.
MS(ESI)m/z=475.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ12.18-11.88(m,1H),8.44(d,J=7.3Hz,1H),7.58(br d,J=7.8Hz,2H),7.36(s,1H),6.87-6.82(m,1H),3.75(s,4H),3.68-3.60(m,2H),3.56(s,3H),3.51-3.41(m,1H),3.26-3.18(m,1H),3.09-2.97(m,2H),2.69(s,2H),2.39(s,3H).
Example 16
Methyl (S) -2- ((2- (4- (cyclopropylcarbamoyl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (16)
Figure BDA0003034737920000352
See example 11, step two for the synthetic procedure of example 16. In which cyclopropylamine was used instead of methylamine hydrochloride and 13c instead of 11 a.
MS(ESI)m/z=484.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.66(d,J=4.0Hz,1H),8.42(d,J=7.2Hz,1H),7.64(d,J=8.0Hz,2H),7.35(s,1H),6.84(dd,J=1.6,7.2Hz,1H),3.73(br d,J=12.0Hz,1H),3.67-3.58(m,2H),3.55(s,3H),3.50-3.40(m,1H),3.30(br s,1H),3.22(dt,J=2.8,11.2Hz,1H),3.03(br d,J=5.2Hz,2H),2.92-2.83(m,1H),2.38(s,3H),0.77-0.70(m,2H),0.64-0.57(m,2H).
Example 17
Methyl (S) -2- ((2- (2, 6-difluoro-4- (2- (methylamino) -2-carbonylethyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (17)
Figure BDA0003034737920000361
The method comprises the following steps: methyl (S) -2- ((2- (4- (2- (tert-butoxy) -2-carbonylethyl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (17a)
To a solution of compound 13a (24.0mg,0.05mmol) in THF (1mL) at 0 deg.C under nitrogen was added 2-tert-butoxy-2-carbonylethyl zinc bromide (50. mu.L, 0.10mmol,2M in THF). After stirring the reaction mixture at 80 ℃ for 6 hours, it was filtered and concentrated under reduced pressure. The residue was purified by C18 reverse phase chromatography (acetonitrile/water (containing 0.05% aqueous ammonia) ═ 0-95%) and lyophilized to give compound 17a (20.6mg, yield: 80%).
MS(ESI)m/z=516.2[M+H]+.
Step two: (S) -2- (3, 5-difluoro-4- (3- ((4- (methoxycarbonyl) morpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) phenyl) acetic acid (17b)
To a solution of compound 17a (20.6mg,0.04mmol) in dichloromethane (1mL) was added trifluoroacetic acid (0.2 mL). After the reaction mixture was stirred at room temperature for 1 hour, it was concentrated under reduced pressure. The residue was purified by C18 reverse phase chromatography (acetonitrile/water (containing 0.05% trifluoroacetic acid) ═ 0-95%) and lyophilized to give compound 17b (16.5mg, yield: 90%).
MS(ESI)m/z=460.2[M+H]+.
Step three: methyl (S) -2- ((2- (2, 6-difluoro-4- (2- (methylamino) -2-carbonylethyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (17)
See example 12 for step four for the synthetic procedure for example 17. In which methylamine hydrochloride is used to replace methylamine ethanol solution, and 17b is used to replace 12 c.
MS(ESI)m/z=473.1[M+H]+.
Example 18
Methyl (S) -2- ((2- (2, 6-difluoro-4- (3-hydroxyoxetan-3-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (18)
Figure BDA0003034737920000371
A solution of compound 13a (48.0mg,0.10mmol) in THF (5mL) was cooled to-78 deg.C, n-butyllithium (50. mu.L, 2.5M in hexane) was added dropwise quickly, the reaction stirred for 5 seconds, and oxetanone (11.0mg,0.15mmol) was added. The reaction was slowly warmed to room temperature and stirred for another 1 hour, the reaction was quenched by the addition of saturated aqueous ammonium chloride (5mL) and ethyl acetate (5mL), the aqueous phase after separation was extracted with ethyl acetate (5 mL. times.3), the combined organic phases were dried over anhydrous sodium sulfate, filtered, the solvent was dried and separated by reverse phase to give the title compound 18(19.0mg, yield: 40.0%).
MS(ESI)m/z=474.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.41(d,J=7.2Hz,1H),7.44-7.32(m,3H),6.83(dd,J=1.2,6.8Hz,1H),6.69(s,1H),4.80(d,J=6.8Hz,2H),4.74(d,J=6.8Hz,2H),3.75-3.58(m,3H),3.55(s,3H),3.52-3.44(m,1H),3.28-3.18(m,2H),3.03(br d,J=6.0Hz,2H),2.81(br s,1H),2.38(s,3H).
Example 19
Methyl (S) -2- ((2- (2, 6-difluoro-4- (methylsulfonyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (19)
Figure BDA0003034737920000372
L-proline (9.6mg,0.083mmol) and NaOH (3.3mg, 0.083mmol) were dissolved in DMSO (1mL) and stirred at room temperature for 0.5 h, then compound 13a (50.0mg,0.10mmol), sodium methanesulfonate (85.0mg, 0.83mmol) and CuI (15.9mg,0.083mmol) were added and the reaction mixture was stirred at 120 ℃ for 14 h. LCMS showed complete detection reaction. The crude product was purified by preparative HPLC (column: Phenomenex Gemini-NX C1880: 40 mm: 3 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O+10mM NH4HCO3) 27% -53%; flow rate:30mL/min) and lyophilized to give the title compound 19(16.6mg, yield: 33.2%).
MS(ESI)m/z=480.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.20(d,J=6.8Hz,1H),7.63(d,J=6.0Hz,2H),7.40(s,1H),6.75-6.68(m,1H),3.93-3.76(m,3H),3.69(s,3H),3.56(br s,1H),3.35(br s,1H),3.15(s,3H),3.02-2.91(m,3H),2.67-2.58(m,1H),2.43(s,3H)
Example 20
Methyl (S) -2- ((2- (2, 6-difluoro-4- (1H-imidazol-2-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (20)
Figure BDA0003034737920000381
To a solution of compound 13a (50.0mg,0.10mmol) in NMP (4mL) were added imidazole (57.0mg,0.83mmol), CuI (40.0mg,0.21mmol), and Pd (OAc) in that order2(5.0mg,0.02 mmol). The reaction solution was bubbled with nitrogen for 2 minutes, and then stirred at microwave 180 ℃ for 165 minutes.The reaction mixture was filtered, and the filtrate was directly purified by C18 reverse phase chromatography (methanol/water (containing 0.05% ammonia) ═ 0-40%) and lyophilized to give the title compound 20(21.0mg, yield: 45%).
MS(ESI)m/z=468.0[M+H]+.
1H NMR(400MHz,DMSO-d6)δ12.38(br s,1H),8.85(d,J=6.8Hz,1H),8.16(d,J=8.8Hz,2H),7.85-7.66(m,2H),7.65-7.53(m,1H),7.24(dd,J=1.6,7.2Hz,1H),4.34-4.17(m,3H),4.11-4.04(m,1H),4.02(s,3H),3.79(dt,J=2.8,11.6Hz,1H),3.63-3.52(m,2H),3.22-3.18(m,1H),3.12-2.99(m,1H),2.87(s,3H).
Example 21
Methyl (S) -2- ((2- (2, 6-difluoro-4- (oxazol-2-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (21)
Figure BDA0003034737920000382
See example 20 for the synthetic procedure for example 21. Wherein oxazole is substituted for imidazole.
MS(ESI)m/z=469.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.21(d,J=7.2Hz,1H),7.78(s,1H),7.72(d,J=8.0Hz,2H),7.48(br s,1H),7.31(s,1H),6.72(d,J=6.8Hz,1H),3.97-3.74(m,3H),3.67(s,3H),3.62-3.55(m,1H),3.46-3.31(m,1H),3.08-2.95(m,2H),2.94-2.85(m,1H),2.65-2.57(m,1H),2.44(s,3H).
Example 22
Methyl (S) -2- ((2- (2, 6-difluoro-4- (5-methyl-1H-imidazol-2-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (22)
Figure BDA0003034737920000391
See example 20 for the synthetic procedure for example 22. Wherein imidazole is replaced by 5-methyl-1H-imidazole.
MS(ESI)m/z=482.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ12.49(br s,1H),8.42(br d,J=6.8Hz,1H),7.66(br d,J=7.6Hz,2H),7.35(br s,1H),7.07(br s,1H),6.83(d,J=7.2Hz,1H),3.76-3.59(m,3H),3.54(s,3H),3.48(br d,J=7.6Hz,1H),3.30(br s,1H),3.28-3.17(m,1H),3.05(br d,J=5.2Hz,2H),2.81(br s,1H),2.38(s,3H),2.31-2.16(m,3H).
Example 23
Methyl (S) -2- ((2- (2, 6-difluoro-4- (5-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (23)
Figure BDA0003034737920000392
See example 20 for the synthetic procedure for example 23. Wherein 3-methyl-4H-1, 2, 4-triazole is used for replacing imidazole.
MS(ESI)m/z=483.3[M+H]+.
1H NMR(400MHz,CDCl3)δ8.22(br d,J=7.2Hz,1H),7.65(br d,J=7.2Hz,2H),7.46(br s,1H),6.71(br d,J=6.8Hz,1H),3.80(br s,3H),3.67(s,3H),3.60(br s,1H),3.40(br s,1H),3.09-2.96(m,2H),2.89(br s,1H),2.62(dd,J=10.8,12.8Hz,1H),2.50(s,3H),2.44(s,3H).
Example 24
Methyl (S) -2- ((2- (2, 6-difluoro-4- (1H-pyrazol-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (24)
Figure BDA0003034737920000401
13a (120.0mg,0.25mmol) was dissolved in NMP (3mL) in a microwave tube, and 1H-pyrazole (51.0mg,0.75mmol), cuprous chloride (24.7mg,0.25mmol), acetylacetone (51. mu.L, 0.50mmol) and K were added in that order2CO3(103.6mg,0.75 mmol). After sealing, the reaction mixture was heated to 140 ℃ under microwave for 10 hours. LCMS showed reaction completion. The reaction mixture was poured into water (10mL) and NH3·H2O (5mL) and extracted with ethyl acetate (20mL x 3), the organic phases were combined, washed with brine, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified by preparative HPLC (column: Phenomenex Gemini-NX C1875 x 30mm x3 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 19% -59%; flow rate:30mL/min) and lyophilized to give the title compound 24(13.2mg, yield: 11.0%).
MS(ESI)m/z=468.2[M+H]+.
1H NMR(400MHz,CD3OD)δ8.40(br d,J=2.8Hz,2H),7.79(s,1H),7.65(d,J=8.8Hz,2H),7.34(s,1H),6.86(d,J=7.2Hz,1H),6.59(s,1H),3.89-3.74(m,3H),3.64(s,3H),3.60(br s,1H),3.40-3.34(m,1H),3.16-3.07(m,2H),2.89(br s,1H),2.64(br s,1H),2.45(s,3H);
Example 25
Methyl (S) -2- ((2- (2, 6-difluoro-4- (2-methyl-1H-imidazol-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (25)
Figure BDA0003034737920000402
See example 24 for the synthetic procedure for example 25. Wherein 2-methyl-1H-imidazole is used to replace 1H-pyrazole.
MS(ESI)m/z=482.3[M+H]+.
1H NMR(400MHz,CD3OD)δ8.44(br d,J=6.8Hz,1H),7.44-7.30(m,4H),7.07(br s,1H),6.90(d,J=7.2Hz,1H),3.91-3.76(m,3H),3.67(s,3H),3.64-3.58(m,1H),3.43-3.35(m,1H),3.14(br d,J=6.0Hz,2H),2.91(br s,1H),2.66(br s,1H),2.55-2.41(m,6H).
Example 26
Methyl (S) -2- ((2- (2, 6-difluoro-4- (1H-pyrazol-5-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (26)
Figure BDA0003034737920000411
To a solution of 13a (48.0mg,0.10mmol) in 1, 4-dioxane/water (5/1,3.6mL) in a microwave tube was added (1H-pyrazol-3-yl) -boronic acid (44.7mg,0.40mmol), Cs2CO3(65.2mg,0.20mmol) and PdCl2Dppf (14.8mg,0.02 mmol). The reaction solution was bubbled with nitrogen for 2 minutes, and then stirred at microwave 90 ℃ for 1 hour. The reaction mixture was suction-filtered, and the filtrate was concentrated to give a residue which was purified by C18 reverse phase chromatography (acetonitrile/water (containing 0.05% aqueous ammonia) ═ 0 to 95%, flow rate: 60mL/min) and lyophilized to give the title compound 26(33.0mg, yield: 70.6%).
MS(ESI)m/z=468.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.42(d,J=7.3Hz,1H),7.84(d,J=1.8Hz,1H),7.65(d,J=8.8Hz,2H),7.36(s,1H),6.93(d,J=2.0Hz,1H),6.83(dd,J=1.4,7.2Hz,1H),3.75-3.61(m,3H),3.55(s,3H),3.50(br d,J=8.3Hz,2H),3.25(br s,1H),3.06(br d,J=6.0Hz,2H),2.81(br s,1H),2.39(s,3H).
Example 27
Methyl (S) -2- ((2- (2, 6-difluoro-4- (1H-pyrazol-4-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (27)
Figure BDA0003034737920000412
See example 26 for the synthetic procedure for example 27. Wherein 4-pyrazole boronic acid pinacol ester is used instead of (1H-pyrazole-3-yl) -boronic acid.
MS(ESI)m/z=468.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.40(d,J=7.0Hz,1H),8.26(s,2H),7.52(d,J=8.8Hz,2H),7.34(s,1H),6.85-6.78(m,1H),3.78-3.60(m,3H),3.54(s,3H),3.48(br s,1H),3.27-3.15(m,1H),3.03(br d,J=6.0Hz,2H),2.80(br s,1H),2.44(br s,1H),2.38(s,3H).
Example 28
Methyl (S) -2- ((2- (2, 6-difluoro-4- (1-methyl-1H-pyrazol-3-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (28)
Figure BDA0003034737920000421
See example 26 for the synthetic procedure for example 28. Wherein 4-pyrazole boronic acid pinacol ester is used instead of 1-methylpyrazole-3-boronic acid pinacol ester.
MS(ESI)m/z=482.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.42(d,J=7.5Hz,1H),7.82(s,1H),7.60(br d,J=8.3Hz,2H),7.35(s,1H),6.92(s,1H),6.83(br d,J=6.8Hz,1H),3.92(s,3H),3.79-3.57(m,4H),3.55(s,3H),3.50(br s,1H),3.24(br t,J=11.0Hz,1H),3.05(br d,J=5.5Hz,2H),2.80(br s,1H),2.39(s,3H).
Example 29
Methyl (S) -2- ((2- (4- (4, 5-dihydro-1H-imidazol-2-yl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (29)
Figure BDA0003034737920000422
To a solution of 13a (46.0mg,0.096mmol) in toluene (2mL) were added tert-butylisocyano (13.0mg,0.156mmol), ethylenediamine (50.0mg,0.832mmol), cesium carbonate (58.0mg,0.178mmol) and PdCl in that order2Dppf (13.0mg,0.018 mmol). Reflux under nitrogen for 16 h and LCMS indicated complete reaction of starting material. The reaction mixture was suction filtered, and the filtrate was concentrated to give a residue which was purified by C18 reverse phase chromatography (acetonitrile/water (containing 0.05% aqueous ammonia) ═ 0-40%) and lyophilized to give the title compound 29(16.0mg, yield: 35%).
MS(ESI)m/z=470.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ10.97-10.81(m,2H),8.68(br dd,J=7.5,12.8Hz,1H),7.92(br t,J=6.7Hz,2H),7.59(br d,J=13.1Hz,1H),7.24-7.06(m,1H),4.14-4.05(m,7H),3.88(br s,12H),3.66-3.59(m,2H),3.57(s,3H),3.54-3.36(m,2H),3.20(br dd,J=9.0,11.5Hz,2H),3.12-3.01(m,1H),2.77(br s,1H),2.47(br d,J=3.5Hz,3H).
Example 30
Methyl (S) -2- ((2- (4-acetylamino-2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (30)
Figure BDA0003034737920000431
To a solution of 13a (50.0mg,0.10mmol) in 1, 4-dioxane (1.5mL) was added acetamide (8.6mg,0.15mmol), Xantphos (12.0mg,0.02mmol), Cs in that order2CO3(67.8mg,0.21mmol) and Pd (OAc)2(3.5mg,0.015 mmol). The reaction solution was bubbled with nitrogen for 2 minutes, and then stirred at 140 ℃ for 35 minutes under microwave. The mixture was diluted with methanol (20mL), filtered, the filtrate concentrated under reduced pressure to give the crude product which was purified by preparative HPLC (column: Welch Xtimate C18150 mm 25mm 5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 25% -50%; flow rate:30mL/min) and lyophilized to give the title compound 30(21.5mg, yield: 32.0%).
MS(ESI)m/z=459.2[M+H]+.
1H NMR(400MHz,CDCl3)δ9.28(br s,1H),8.19(d,J=7.2Hz,1H),7.37(s,1H),7.19(br d,J=9.2Hz,2H),6.69(br d,J=6.4Hz,1H),3.97-3.73(m,3H),3.68(s,3H),3.57(br s,1H),3.46-3.32(m,1H),3.03-2.84(m,3H),2.61(dd,J=10.8,12.8Hz,1H),2.44(s,3H),2.18(s,3H).
Example 31
Methyl (S) -2- ((2- (4- ((dimethyl (carbonyl) -lambda)6-sulfanylidene) amino) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a]Pyridin-3-yl) methyl) morpholine-4-carboxylate (31)
Figure BDA0003034737920000432
See example 30 for the synthetic procedure for example 31. Wherein dimethyl sulfinyl imine is used to replace acetamide, and Pd is used2(dba)3Alternative Pd (OAc)2
MS(ESI)m/z=493.2[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.37(d,J=7.3Hz,1H),7.31(s,1H),6.79(dd,J=1.5,7.0Hz,1H),6.66(d,J=9.8Hz,2H),3.74-3.60(m,3H),3.55(s,3H),3.48(br s,1H),3.29-3.21(m,1H),3.06-2.94(m,2H),2.83(br s,1H),2.63-2.52(m,1H),2.50(br s,6H),2.37(s,3H).
Example 32
Methyl (S) -2- ((2- (4- (cyclopropylamino) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (32)
Figure BDA0003034737920000441
The method comprises the following steps: methyl (S) -2- ((2- (4- (tert-butoxycarbonylamino) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (32a)
To a solution of compound 13a (1.00g,2.08mmol) in toluene (30mL) were added tert-butyl carbamate (0.49g,4.16mmol), XPhos (0.20g,0.42mmol), and Cs in that order2CO3(2.03g,6.25mmol) and Pd2(dba)3(0.19g,0.21 mmol). Stirring was carried out at 110 ℃ for 12 hours under nitrogen. Water (40mL) was added to the mixture, the mixture was extracted with ethyl acetate (60 mL. times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered with suction and concentrated under reduced pressure. The obtained residue was purified by silica gel column chromatography (mobile phase: petroleum ether/ethyl acetate 0-100%; flow rate: 35mL/min) to obtain compound 32a (760.0mg, yield: 57.2%).
MS(ESI)m/z=517.1[M+H]+.
1H NMR(400MHz,CDCl3)δ8.18(d,J=7.0Hz,1H),7.41(br s,1H),7.14(br d,J=9.3Hz,2H),6.87(br s,1H),6.68(br d,J=5.8Hz,1H),3.82(br s,3H),3.69(s,3H),3.57(br s,1H),3.41(br s,1H),3.02-2.86(m,3H),2.61(dd,J=10.7,12.9Hz,1H),2.43(s,3H),1.55(s,9H).
Step two: methyl (S) -2- ((2- (4-amino-2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (32b)
To a solution of compound 32a (760.0mg,1.47mmol) in dichloromethane (10mL) was added a solution of hydrochloric acid/1, 4-dioxane (4N,10 mL). The reaction mixture was stirred at room temperature for 12 hours and LCMS showed the reaction was complete. The mixture was concentrated under reduced pressure and the residue was taken up with saturated NaHCO3The solution was adjusted to pH 8, extracted with ethyl acetate (50 mL. times.3), the organic phases combined, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give compound 32b (560.0mg, 82.3%).
MS(ESI)m/z=417.0[M+H]+.
1H NMR(400MHz,CD3OD)δ8.35(d,J=7.0Hz,1H),7.29(s,1H),6.82(d,J=5.8Hz,1H),6.35(d,J=10.0Hz,2H),3.81(br d,J=11.8Hz,3H),3.68-3.65(m,3H),3.63-3.50(m,2H),3.43-3.35(m,1H),3.11-2.97(m,2H),2.64(br t,J=11.8Hz,1H),2.44(s,3H).
Step three: methyl (S) -2- ((2- (4- (cyclopropylamino) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (32)
To compound 32b (40.0mg,0.096mmol) in DMF (2mL) was added successively cyclopropylcarboxylic acid (15.2mg,0.18mmol), HATU (50.4mg,0.13mmol) and N, N-diisopropylethylamine (58. mu.L, 0.35 mmol). After stirring at room temperature for 72 hours, the reaction mixture was directly purified by C18 reverse phase chromatography (acetonitrile/water (containing 0.05% ammonia water) ═ 5-95%) and lyophilized to give the title compound 32(11.1mg, yield: 24%).
MS(ESI)m/z=485.1[M+H]+.
1H NMR(400MHz,DMSO-d6)δ10.67(s,1H),8.38(d,J=7.2Hz,1H),7.42(d,J=9.6Hz,2H),7.32(s,1H),6.81(dd,J=1.6,7.2Hz,1H),4.21(s,2H),3.75-3.60(m,3H),3.55(s,3H),3.51-3.34(m,3H),3.28-3.20(m,1H),3.00(br d,J=6.0Hz,2H),2.81(br s,1H),2.37(s,3H),1.83-1.74(m,1H),1.71-1.58(m,1H),0.94-0.88(m,2H).
Example 33
Methyl (S) -2- ((2- (4- (2-cyanoacetamido) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (33)
Figure BDA0003034737920000451
See example 32, step three for the synthetic procedure of example 33. Wherein cyanoacetic acid is substituted for cyclopropylcarboxylic acid.
MS(ESI)m/z=484.0[M+H]+.
1H NMR(400MHz,DMSO-d6)δ10.78(s,1H),8.39(d,J=7.2Hz,1H),7.38(d,J=9.2Hz,2H),7.33(s,1H),6.81(dd,J=1.6,7.2Hz,1H),3.99(s,2H),3.75-3.59(m,3H),3.55(s,3H),3.51-3.38(m,2H),3.30-3.17(m,1H),3.01(br d,J=5.6Hz,2H),2.81(br s,1H),2.37(s,3H).
Example 34
Methyl (S) -2- ((2- (4-benzoylamino-2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (34)
Figure BDA0003034737920000452
To a solution of compound 32b (10.0mg,0.024mmol) in dichloromethane (1mL) was added benzoyl chloride (5. mu.L, 0.044mmol) and triethylamine (13. mu.L, 0.096mmol) in that order at 0 ℃. After stirring at room temperature for 3 hours, the reaction was quenched by addition of saturated sodium bicarbonate solution, concentrated under reduced pressure, and the residue was directly purified by C18 reverse phase chromatography (acetonitrile/water (containing 0.05% aqueous ammonia) ═ 5-95%) and lyophilized to give the title compound 34(2.1mg, yield: 17%).
MS(ESI)m/z=521.1[M+H]+.
Example 35
Methyl (S) -2- ((2- (4- (3, 3-dimethylureido) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (35)
Figure BDA0003034737920000461
See example 30 for the synthetic procedure for example 35. Wherein 3, 3-dimethyl urea is used to replace acetamide, and Pd is used2(dba)3Substitute Pd(OAc)2
MS(ESI)m/z=488.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.98(s,1H),8.81(br d,J=7.0Hz,1H),7.70(s,1H),7.52(d,J=11.0Hz,2H),7.35(br d,J=5.6Hz,1H),3.88(br d,J=12.3Hz,1H),3.64(br d,J=9.3Hz,3H),3.58(s,3H),3.24-3.16(m,3H),3.15-3.05(m,2H),2.97(s,6H),2.53(s,3H).
Example 36
Methyl (S) -2- ((2- (4- (cyclopentylamino) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (36)
Figure BDA0003034737920000462
To a solution of compound 13a (50.0mg,0.10mmol) in toluene (1.5mL) were added cyclopentylamine (20. mu.L, 0.21mmol), BINAP (13.0mg,0.02mmol), tBuONa (20.0mg,0.21mmol), and Pd in that order2(dba)3(14.3mg,0.02 mmol). Stirring was carried out at 100 ℃ for 16 hours under a nitrogen atmosphere. The reaction mixture was filtered, the filtrate was concentrated under reduced pressure and the crude product obtained was purified by preparative HPLC (column: Phenomenex Gemini-NX C1875: 30 mm. times.3 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O+10mM NH4HCO3) 45% -63%; flow rate:30mL/min) and lyophilized to give the title compound 36(14.0mg, yield: 28.0%).
MS(ESI)m/z=485.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.15(d,J=6.0Hz,1H),7.37(s,1H),6.62(d,J=6.4Hz,1H),6.26-6.15(m,2H),4.04-3.95(m,1H),3.90-3.74(m,3H),3.68(s,3H),3.63-3.55(m,1H),3.47-3.35(m,1H),3.03-2.83(m,3H),2.62(dd,J=10.8,13.2Hz,1H),2.40(s,3H),2.12-2.02(m,2H),1.82-1.72(m,2H),1.72-1.66(m,2H),1.57-1.45(m,2H).
Example 37
Methyl (S) -2- ((2- (2, 6-difluoro-4- (2-carbonylpiperidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (37)
Figure BDA0003034737920000471
To a solution of compound 13a (48.0mg,0.10mmol) in 1, 4-dioxane (2mL) was added 2-piperidone (37. mu.L, 0.40mmol), N' -dimethylethylenediamine (6. mu.L, 0.05mmol), cesium carbonate (195.5mg,0.60mmol), and cuprous iodide (9.5mg,0.05mmol) in that order in a microwave tube. The reaction mixture was heated to 150 ℃ under microwave for 1 hour. LCMS showed reaction completion. The reaction mixture was concentrated under reduced pressure, the residue was dissolved in acetonitrile and filtered, and the filtrate was directly subjected to C18 reverse phase chromatography (acetonitrile/water (containing 0.05% NH)3·H2O) 5-95%, flow rate: 60mL/min) and lyophilized to give the title compound 37(33.0mg, yield: 53.9%).
MS(ESI)m/z=499.4[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.83(br d,J=6.8Hz,1H),7.72(br s,1H),7.48-7.32(m,3H),3.91(br d,J=12.4Hz,3H),3.73(br t,J=5.2Hz,3H),3.62(br s,2H),3.59-3.56(m,3H),3.30-3.03(m,4H),2.82(br s,1H),2.70-2.59(m,1H),2.54(s,3H),1.96-1.79(m,3H).
Example 38
Methyl (S) -2- ((2- (2, 6-difluoro-4- (3-carbonylmorpholin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (38)
Figure BDA0003034737920000472
See example 37 for synthetic procedure for example 38. Wherein 3-morpholone is used for replacing 2-piperidone.
MS(ESI)m/z=501.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.42(d,J=7.3Hz,1H),7.44(d,J=9.5Hz,2H),7.35(s,1H),6.83(dd,J=1.5,7.0Hz,1H),4.27(s,2H),4.04-3.99(m,2H),3.88-3.84(m,2H),3.76(br d,J=12.8Hz,1H),3.70-3.61(m,2H),3.56(s,3H),3.49(br s,1H),3.29-3.23(m,1H),3.04(br d,J=6.0Hz,2H),2.90-2.66(m,1H),2.38(s,3H),1.24(s,1H)
Example 39
Methyl (S) -2- ((2- (2, 6-difluoro-4- (4-methyl-2-carbonylpiperazin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (39)
Figure BDA0003034737920000481
See example 37 for synthetic procedure for example 39. Wherein 4-methylpiperazin-2-one is used instead of 2-piperidone.
MS(ESI)m/z=513.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.41(d,J=7.2Hz,1H),7.38(s,1H),7.35(d,J=2.6Hz,2H),6.83(dd,J=1.5,7.0Hz,1H),3.81-3.75(m,2H),3.74-3.59(m,1H),3.72-3.59(m,3H),3.55(s,3H),3.48(br s,2H),3.27-3.20(m,2H),3.17(s,2H),3.03(br d,J=6.0Hz,2H),2.78-2.72(m,2H),2.37(s,3H),2.29(s,3H).
Example 40
Methyl (S) -2- ((2- (2, 6-difluoro-4- (2-carbonylpyrrolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (40)
Figure BDA0003034737920000482
See example 37 for synthetic procedure for example 40. Wherein 2-pyrrolidone is substituted for 2-piperidone.
MS(ESI)m/z=485.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.40(d,J=7.3Hz,1H),7.60(d,J=10.0Hz,2H),7.34(s,1H),6.82(dd,J=1.4,7.2Hz,1H),3.88(t,J=7.2Hz,2H),3.75-3.60(m,3H),3.55(s,3H),3.46(br d,J=7.3Hz,1H),3.28-3.19(m,1H),3.01(br d,J=6.0Hz,2H),2.80(br s,1H),2.57(t,J=8.0Hz,3H),2.37(s,3H),2.08(quin,J=7.6Hz,2H).
EXAMPLE 41
Methyl (S) -2- ((2- (2, 6-difluoro-4- (2-methyl-5-carbonylpyrrolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (41)
Figure BDA0003034737920000483
See example 30 for the synthetic procedure for example 41. Wherein 5-methyl-2-pyrrolidone is used instead of acetamide.
MS(ESI)m/z=499.3[M+H]+.
1H NMR(400MHz,CDCl3)δ8.24-8.11(m,1H),7.37(s,1H),7.32-7.27(m,2H),6.66(d,J=7.2Hz,1H),4.43-4.28(m,1H),4.00-3.73(m,3H),3.68(d,J=2.0Hz,3H),3.59(br s,1H),3.46-3.34(m,1H),3.08-2.80(m,3H),2.78-2.68(m,1H),2.67-2.51(m,2H),2.46-2.35(m,4H),1.90-1.76(m,1H),1.34(d,J=6.4Hz,3H).
Example 42
Methyl (S) -2- ((2- (2, 6-difluoro-4- (2-carbonyloxazolidin-3-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (42)
Figure BDA0003034737920000491
See example 37 for synthetic procedure for example 42. Wherein 2-oxazolidinone is used for replacing 2-piperidone.
MS(ESI)m/z=487.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.41(d,J=7.3Hz,1H),7.51-7.45(m,2H),7.35(s,1H),6.83(dd,J=1.4,7.2Hz,1H),4.53-4.47(m,2H),4.15-4.09(m,2H),3.78-3.57(m,4H),3.56(s,3H),3.51-3.45(m,1H),3.28-3.21(m,1H),3.05-2.98(m,2H),2.89-2.73(m,1H),2.38(s,3H).
Example 43
Methyl (S) -2- ((2- (2, 6-difluoro-4- (2-carbonylimidazolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (43)
Figure BDA0003034737920000492
See example 37 for synthetic procedure for example 43. Wherein 2-imidazolidinone is substituted for 2-piperidone.
MS(ESI)m/z=486.4[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.38(d,J=7.0Hz,1H),7.43(d,J=10.6Hz,2H),7.32(s,2H),6.81(dd,J=1.5,7.1Hz,1H),3.94-3.86(m,2H),3.73-3.60(m,3H),3.55(s,3H),3.48-3.40(m,3H),3.30-3.19(m,2H),3.03-2.98(m,2H),2.80(br s,1H),2.90-2.74(m,1H),2.37(s,3H).
Example 44
Methyl (S) -2- ((2- (2, 6-difluoro-4- (3-methyl-2-carbonylimidazolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (44)
Figure BDA0003034737920000501
See example 37 for synthetic procedure for example 44. Wherein 1-methylimidazolidin-2-one is substituted for 2-piperidone.
MS(ESI)m/z=499.4[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.39(d,J=7.0Hz,1H),7.49-7.42(m,2H),7.36-7.31(m,1H),6.82(d,J=7.0Hz,1H),3.88-3.81(m,2H),3.80-3.58(m,4H),3.55(s,3H),3.52-3.46(m,3H),3.31-3.18(m,2H),3.06-2.96(m,2H),2.80(s,3H),2.38(s,3H).
Example 45
Methyl (S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7- (methyl-d)3) Imidazo [1,2-a ]]Pyridin-3-yl) methyl) morpholine-4-carboxylate (45)
Figure BDA0003034737920000502
The method comprises the following steps: tert-butyl (4- (methyl-d)3) Pyridin-2-yl) carbamate (45a)
To a suspension of magnesium turnings (0.40g,16.56mmol) in anhydrous ether (5mL) was added iodine (0.35g,1.38mmol) and stirred under nitrogen for 10 min. The reaction mixture was cooled to 0 ℃ and then slowly deuterated iodomethane (0.86mL,13.80mmol) was added, allowed to return to room temperature and stirred for 2.5 hours. The mixture prepared was used directly in the next step without further purification. To a solution of tert-butyl 4-bromopyridine-2-carbamate (400.0mg,1.46mmol) and bis triphenylphosphine palladium dichloride (25.7mg,0.037mmol) in THF (10mL) at room temperature under a nitrogen atmosphere was added dropwise the Grignard reagent prepared above. After the reaction mixture was stirred at 80 ℃ for 2 hours, LCMS showed complete disappearance of starting material. Adding saturated NH4The reaction was quenched with Cl solution (20mL), extracted with ethyl acetate (20mL × 3), the organic phases combined, washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (ethyl acetate/petroleum ether ═ 0 to 15%, flow rate:30mL/min) to give compound 45a (290.0mg, 84.4%).
MS(ESI)m/z=212.0[M+H]+.
1H NMR(400MHz,CDCl3)δ8.12(d,J=5.2Hz,1H),7.81(s,1H),7.60(br s,1H),6.80(dd,J=1.6,5.2Hz,1H),1.55(s,9H).
Step two: tert-butyl (4- (methyl-d)3) Pyridin-2-yl) carbamate (45b)
To a solution of compound 45a (150.0mg,0.71mmol) in dichloromethane (10mL) was added dropwise a solution of hydrochloric acid in methanol (4M,5mL) at 0 deg.C, followed by stirring at room temperature for 12 hours. LCMS showed complete disappearance of starting material. The mixture was concentrated under reduced pressure, diluted with water (10mL), adjusted to pH 12 with saturated NaOH solution and extracted with dichloromethane (20mL × 2), the organic phases combined, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give compound 45b (60.0mg, 76.3%).
MS(ESI)m/z=112.2[M+H]+.
1H NMR(400MHz,CDCl3)δ7.59(br t,J=5.8Hz,1H),6.93(s,1H),6.60(d,J=6.4Hz,1H).
Step three: methyl (S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7- (methyl-d)3) Imidazo [1,2-a ]]Pyridin-3-yl) methyl) Morpholine-4-carboxylate (45)
See example 10 for step three, where 4- (methyl-d) is used3) Pyridin-2-amine replaces 4- (trifluoromethyl) pyridin-2-amine.
MS(ESI)m/z=462.2[M+H]+.
1H NMR(400MHz,CD3OD)δ8.42(d,J=7.2Hz,1H),7.61(d,J=8.4Hz,2H),7.35(s,1H),6.88(dd,J=1.6,7.2Hz,1H),3.89-3.69(m,4H),3.68-3.65(m,3H),3.62-3.53(m,1H),3.15-3.06(m,2H),2.97(s,3H),2.93-2.81(m,1H),2.74-2.51(m,1H).
Example 46
Methyl 3- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -4, 4-difluoropiperidine-1-carboxylate (46)
Figure BDA0003034737920000511
The method comprises the following steps: dimethyl 4-carbonylpiperidine-1, 3-dicarboxylate (46a)
To a solution of methyl 4-oxapiperidine-3-carboxylate (11.00g,56.81mmol) in dichloromethane (200mL) at 0 deg.C under nitrogen was added methyl chloroformate (5.3mL,68.15mmol) and diisopropylethylamine (28.2mL,170.43mmol), and the reaction mixture was stirred at room temperature for 12 hours. TLC (petroleum ether/ethyl acetate 2/1) showed the reaction was complete. The reaction solution was washed with water (80 mL. times.2) and brine (80mL), and the organic phase was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (ethyl acetate/petroleum ether ═ 0 to 60%, flow rate: 60mL/min) to give compound 46a (9.70g, 79.3%).
1H NMR(400MHz,CDCl3)δ11.91(s,1H),4.04(br s,2H),3.72(s,3H),3.66(s,3H),3.55(br s,2H),2.33(br s,2H).
Step two: dimethyl 1, 4-dioxa-8-azaspiro [4.5] decane-6, 8-dicarboxylate (46b)
To a solution of compound 46a (2.00g,9.29mmol) in toluene (40mL) was added 1, 2-ethanediol (5.77g,92.94mmol) and p-toluenesulfonic acid (80.0mg,0.46 mmol). Stirring was carried out at 130 ℃ for 12 hours under nitrogen and water was removed from the mixture at the same time using a Dean-Stark trap. TLC (petroleum ether/ethyl acetate 2/1) showed the reaction was complete. The mixture was concentrated under reduced pressure, and water (30mL) was added to the residue, followed by extraction with ethyl acetate (50 mL. times.2). The organic phases were combined, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (ethyl acetate/petroleum ether 0 to 60%, flow rate: 40mL/min) to give compound 46b (2.00g, 83.0%).
Step three: methyl 6- (hydroxymethyl) -1, 4-dioxa-8-azaspiro [4.5] decane-8-carboxylate (46c)
At 0 deg.C under nitrogen protection to NaBH4(3.52g,104.14mmol) in ethanol (135mL) was added CaCl2(5.78g,52.07 mmol). After the mixture was stirred at 0 ℃ for 30 minutes, a solution of 46b (9.00g,34.71mmol) in THF (135mL) was added, followed by stirring at room temperature for 12 hours. TLC (petroleum ether/ethyl acetate 1/1) showed substantial completion of the reaction. To the mixture was added water (80mL) and filtered, the filtrate was concentrated under reduced pressure, the residue was extracted with ethyl acetate (60mL × 5), the organic phases were combined, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (ethyl acetate/petroleum ether 0 to 80%, flow rate: 60mL/min) to give compound 46c (5.40g, 63.9%).
1H NMR(400MHz,CDCl3)δ3.94(s,4H),3.77-3.65(m,2H),3.64(s,3H),3.59-3.23(m,4H),2.80-2.21(m,1H),2.84-2.20(m,1H),1.91(br s,1H),1.70(br s,1H),1.49(br s,1H).
Step four: methyl 6-formyl-1, 4-dioxa-8-azaspiro [4.5] decane-8-carboxylate (46d)
Dess-Martin reagent (11.00g,25.95mmol) was added portionwise to a solution of compound 46c (5.00g,21.62mmol) in dichloromethane (100mL) at 0 deg.C under nitrogen. After the reaction mixture was stirred at 0 ℃ for 2 hours, TLC (petroleum ether/ethyl acetate 1/1) showed the reaction was complete. Dichloromethane (100mL) was added to the mixture, followed by saturated NaHCO3Solution (50 mL. times.2), saturated Na2SO3The solution (50 mL. times.2) and brine (50mL) were washed, the organic phases combined, dried over anhydrous sodium sulfate, filtered andconcentration under reduced pressure gave compound 46d (4.90g, 98.9%).
1H NMR(400MHz,CDCl3)δ9.82(d,J=0.9Hz,1H),4.14-3.99(m,5H),3.88(br d,J=12.6Hz,1H),3.72(s,3H),3.48(dd,J=9.3,13.8Hz,1H),3.28(ddd,J=3.7,10.1,13.5Hz,1H),2.70(br s,1H),1.85-1.77(m,1H),1.75-1.66(m,1H).
Step five: methyl 6-ethynyl-1, 4-dioxa-8-azaspiro [4.5] decane-8-carboxylic acid ester (46e)
To a solution of compound 46d (4.90g,18.06mmol) in methanol (100mL) at 0 deg.C under nitrogen was added K2CO3(11.82g,85.50mmol) and dimethyl (1-diazo-2-oxopropyl) phosphonate (4.93g,25.65 mmol). After the reaction mixture was stirred at room temperature for 12 hours, TLC (petroleum ether/ethyl acetate 3/1) showed the reaction was complete. To the mixture was added water (50mL), and concentrated under reduced pressure to remove methanol, the residue was extracted with ethyl acetate (80mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (ethyl acetate/petroleum ether: 0 to 55%, flow rate: 40mL/min) to give compound 46e (3.00g, 55.9%).
1H NMR(400MHz,CDCl3)δ4.25-4.11(m,2H),4.09-4.00(m,2H),3.99-3.78(m,1H),3.73(s,3H),3.72-3.66(m,1H),3.53(br d,J=11.9Hz,2H),2.69(br s,1H),2.13(d,J=2.4Hz,1H),1.98-1.82(m,1H),1.66-1.59(m,1H).
Step six: methyl 6- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -1, 4-dioxa-8-azaspiro [4.5] decane-8-carboxylate (46f)
See example 4 for a synthetic procedure for compound 46f, wherein 4-methyl-pyridin-2-amine is substituted for 4- (trifluoromethyl) pyridin-2-amine and compound 46e is substituted for tert-butyl (2S) -2-ethynylmorpholine-4-carboxylate.
MS(ESI)m/z=515.1[M+H]+.
Step seven: methyl 3- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -4-carbonylpiperidine-1-carboxylate (46g)
To a solution of compound 46f (300.0mg,0.58mmol) in THF (10mL) was added 6N HCl (3mL, 18.00 mmol). The reaction mixture was stirred at 70 ℃ for 2 hours and LCMS showed the reaction was complete. With saturated NaHCO3The solution was adjusted to pH 8, extracted with ethyl acetate (40 mL. times.3), the organic phases combined, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was subjected to silica gel column chromatography (dichloromethane/methanol (2mL of NH per 500mL of solvent)3·H2O) ═ 0-20%, flow rate 35mL/min) to give 46g (180.0mg, 59.0%) of the compound.
MS(ESI)m/z=471.0[M+H]+.
1H NMR(400MHz,CDCl3)δ7.98(d,J=7.3Hz,1H),7.44(d,J=7.5Hz,4H),6.77(d,J=5.8Hz,1H),4.24(s,1H),4.16-3.90(m,1H),3.65(br s,3H),3.49(br d,J=15.3Hz,1H),3.16(br t,J=9.8Hz,1H),3.06(d,J=5.0Hz,3H),2.81-2.62(m,3H),2.45(s,3H),2.34(s,2H).
Step eight: methyl 3- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -4, 4-difluoropiperidine-1-carboxylate (46)
To a solution of compound 46g (50.0mg,0.11mmol) in dichloromethane (4mL) was added (diethylamino) sulfur trifluoride (39. mu.L, 0.32mmol) at-70 ℃ under nitrogen. The reaction mixture was allowed to return to room temperature and stirred for one hour, saturated NaHCO was added3The solution was quenched, extracted with dichloromethane (20mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was chromatographed on C18 reverse phase (acetonitrile/water (0.05% NH)3·H2O) 5-95%, flow rate: 40mL/min) and lyophilized to give the title compound 46(5.0mg, yield: 9%).
MS(ESI)m/z=493.2[M+H]+.
1H NMR(400MHz,CD3OD)δ8.22(d,J=6.8Hz,1H),7.62(d,J=8.0Hz,2H),7.40(s,1H),6.96(d,J=6.8Hz,1H),3.96(br d,J=12.8Hz,1H),3.78-3.64(m,1H),3.63-3.49(m,3H),3.39-3.37(m,1H),3.17-3.00(m,1H),2.97(s,3H),2.92-2.90(m,1H),2.69-2.67(m,1H),2.49(s,3H),2.33-2.16(m,1H),2.08-2.06(m,1H),1.99-1.78(m,1H).
Example 47
Methyl (S) -2- ((2- (2, 6-difluoro-4- (5- (trifluoromethyl) -1H-imidazol-2-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl)
Methyl) morpholine-4-carboxylate (47)
Figure BDA0003034737920000541
The method comprises the following steps: methyl (S) -2- ((2- (2, 6-difluoro-4-vinylphenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (47a)
To a solution of compound 13a (300.0mg,0.62mmol) in 1, 4-dioxane (8mL) and water (2mL) was added 2-vinyl-4, 4,5, 5-tetramethyl-1, 3, 2-dioxaborane (115.4mg,0.75mmol), Cs2CO3(407.0mg,1.25mmol) and Pd (dppf) Cl2(46.4mg,0.062 mmol). The reaction mixture was degassed and replaced 3 times with nitrogen, then the mixture was stirred at 90 ℃ for 12 hours under nitrogen, LCMS showed complete consumption of starting material. Water (5mL) was added to the mixture, and extracted with ethyl acetate (10 mL. times.3), the organic phases were combined, washed with brine (8mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (ethyl acetate/petroleum ether: 0 to 70%, flow rate: 35mL/min) to give compound 47a (200.0mg, 67.4%).
MS(ESI)m/z=428.2[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.40(d,J=7.2Hz,1H),7.43-7.31(m,3H),6.85-6.72(m,2H),6.07(d,J=17.6Hz,1H),5.46(d,J=11.2Hz,1H),3.78-3.58(m,3H),3.55(s,3H),3.47(br d,J=8.0Hz,1H),3.31-3.18(m,2H),3.02(d,J=6.0Hz,2H),2.80(br s,1H),2.38(s,3H).
Step two: methyl (S) -2- ((2- (2, 6-difluoro-4-formylphenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (47b)
To a solution of compound 47a (200.0mg,0.47mmol) in 1, 4-dioxane (10mL) and water (2mL) was added an aqueous osmium tetroxide solution (1.8mL,0.070mmol) and stirred at room temperature for 10 minutes, followed by sodium periodate (550.0mg,1.87 mmol). After the reaction mixture was stirred at room temperature for 1 hour, LCMS showed complete consumption of starting material. The reaction mixture was poured into water (10mL) and extracted with ethyl acetate (20mL × 3), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give compound 47b (150.0mg, 52.2%).
MS(ESI)m/z=430.2[M+H]+.
1H NMR(400MHz,CD3OD)δ10.01(s,1H),8.38(d,J=7.1Hz,1H),7.74-7.63(m,1H),7.35-7.31(m,1H),7.23(d,J=8.3Hz,1H),6.86-6.81(m,1H),3.88-3.80(m,1H),3.65(s,3H),3.61-3.50(m,2H),3.10-3.03(m,2H),2.86(br s,2H),2.60(br s,2H),2.44(s,3H).
Step three: methyl (S) -2- ((2- (2, 6-difluoro-4- (5- (trifluoromethyl) -1H-imidazol-2-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (47)
To a solution of NaOAc (54.6mg,0.66mmol) in water (3mL) was added 3, 3-dibromo-1, 1, 1-trifluoropropan-2-one (50. mu.L, 0.36mmol) and stirred at 100 ℃ for 0.5 h, then cooled to room temperature. To the solution were added a solution of 47b (130.0mg,0.30mmol) in methanol (6mL) and aqueous ammonia (2mL), followed by stirring at room temperature for 12 hours. LCMS monitors the reaction, concentrates under reduced pressure to remove methanol, extracts the remaining aqueous phase with dichloromethane (10 mL. times.2), combines the organic phases, dries over anhydrous sodium sulfate, filters and concentrates the resulting residue by preparative HPLC (column: Boston Prime C18150: 25 mm. times.5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 40% -65%; flow rate:30mL/min) and lyophilized to give the title compound 47(36.0mg, yield: 21.5%).
MS(ESI)m/z=536.2[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.43(d,J=7.2Hz,1H),8.05(s,1H),7.77(d,J=8.0Hz,2H),7.36(s,1H),6.84(dd,J=1.6,7.2Hz,1H),3.78-3.58(m,3H),3.54(s,3H),3.48(br d,J=7.2Hz,1H),3.23(dt,J=2.8,12.0Hz,1H),3.05(br d,J=6.1Hz,2H),2.79(br s,1H),2.52(br s,1H),2.38(s,3H).
Example 48
Methyl (S) -2- ((2- (4- (5-cyano-1H-imidazol-2-yl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (48)
Figure BDA0003034737920000551
Compound 47(30.0mg,0.056mmol) was dissolved in aqueous ammonia (5mL), and the reaction mixture was stirred at 60 ℃ for 0.5 h. LCMS showed complete consumption of starting material. After the reaction solution was returned to room temperature, the reaction solution was concentrated under reduced pressure, and the crude product was directly subjected to preparative HPLC (column: Phenomenex Gemini-NX C1875: 30 mm. times.3 μm) and mobile phase: acetonitrile/water (0.05% NH)3·H2O+10mM NH4HCO3) 45% -63%; flow rate:30mL/min) and lyophilized to give the title compound 48(15.0mg, yield: 54.4%).
MS(ESI)m/z=493.2[M+H]+.
1H NMR(400MHz,DMSO-d6)δ13.72(br s,1H),8.43(d,J=7.2Hz,1H),8.38(s,1H),7.77(d,J=8.4Hz,2H),7.36(s,1H),6.84(dd,J=1.6,7.2Hz,1H),3.74(br d,J=11.6Hz,1H),3.69-3.59(m,2H),3.54(s,3H),3.48(br s,1H),3.23(br dd,J=8.8,11.6Hz,1H),3.06(br s,2H),2.78(br s,1H),2.52(br s,1H),2.38(s,3H).
Example 49
Methyl (S) -2- ((2- (4- ((cyclopentyloxycarbonyl) amino) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (49)
Figure BDA0003034737920000561
To a solution of compound 32b (50mg,0.12mmol) in dichloromethane (3mL) was added a solution of diisopropylethylamine (99. mu.L, 0.60mmol) and triphosgene (42.8mg,0.14mmol) in dichloromethane (0.5mL) at 0 deg.C under nitrogen. After the reaction mixture was stirred at 0 ℃ for 1 hour, cyclopentanol (33. mu.L, 0.36mmol) was added and stirred at room temperature for 2 hours. LCMS monitors the reaction, the reaction mixture is concentrated under reduced pressure and the residue is subjected to preparative HPLC (column: Phenomenex Gemini-NX C1875 x 30mm x 3. mu.l)m; mobile phase: acetonitrile/water (0.05% NH)3·H2O+10mM NH4HCO3) 45% -63%; flow rate:30mL/min) and lyophilized to give the title compound 49(15.0mg, yield: 23.7%).
MS(ESI)m/z=529.3[M+H]+.
Example 50
Methyl (S) -2- ((7-chloro-2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -6-fluoroimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (50)
Figure BDA0003034737920000571
The method comprises the following steps: tert-butyl N- (4-chloro-5-fluoropyridin-2-yl) carbamate (50a)
To a solution of 2-bromo-4-chloro-5-fluoropyridine (2.0g,9.50mmol) and tert-butyl carbamate (1.1g,9.50mmol) in 1, 4-dioxane (120mL) under nitrogen was added Cs2CO3(6.2g,19.01mmol)、Pd(dba)2(546.5mg,0.95mmol) and Xantphos (1.1g,1.90 mmol). The reaction mixture was then stirred at 100 ℃ for 4 hours. LCMS showed reaction complete. The mixture was concentrated under reduced pressure and H was added to the residue2O (50mL), extracted with ethyl acetate (80 mL. times.3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product is purified by column chromatography on silica gel (
Figure BDA0003034737920000574
80g
Figure BDA0003034737920000575
Silica Flash Column, 0-5% ethyl acetate/petroleum ether as eluent, 60mL/min) to give the title compound (1.8g, yield: 69.1%).
1H NMR(400MHz,CDCl3)δ8.17-8.11(m,2H),7.53(br s,1H),1.55(s,9H).
Step two: 4-chloro-5-fluoropyridin-2-amine (50b)
To tert-butyl N- (4-chloro-5-fluoropyridin-2-yl) amino at 0 deg.CTo a solution of the formate ester (1.8g,7.30mmol) in DCM (20mL) was added TFA (5mL,67.31 mmol). The mixture was stirred at room temperature for 12 hours. TLC (petroleum ether/ethyl acetate 3/1) showed the reaction was complete. The mixture was concentrated under reduced pressure and the residue was taken up with saturated NaHCO3The solution was basified to pH 8 and extracted with DCM (60mL × 3). The combined organic phases are dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure, and the residue is purified by flash chromatography on silica gel
Figure BDA0003034737920000572
40g
Figure BDA0003034737920000573
Silica Flash Column, 0-50% ethyl acetate/petroleum ether, 40mL/min eluent) gave the title compound 50b (1.0g, 88.8%).
1H NMR(400MHz,CDCl3)δ7.88(s,1H),6.48(d,J=4.8Hz,1H),4.56-3.97(m,2H).
Step three: methyl (S) -2- ((7-chloro-2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -6-fluoroimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (50)
See example 4 for synthetic procedure one. Wherein 4- (trifluoromethyl) pyridin-2-amine is replaced by 4-chloro-5-fluoropyridin-2-amine and tert-butyl (2S) -2-ethynylmorpholine-4-carboxylate is replaced by methyl (2S) -2-ethynylmorpholine-4-carboxylate.
MS(ESI)m/z=497.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.46(d,J=4.4Hz,1H),7.70(d,J=6.8Hz,1H),7.44(d,J=8.0Hz,2H),6.44(br d,J=4.4Hz,1H),4.00-3.78(m,3H),3.70(s,3H),3.58(br s,1H),3.40(t,J=10.4Hz,1H),3.05(d,J=4.8Hz,3H),2.94(d,J=5.6Hz,3H),2.72-2.55(m,1H).
Example 51
Methyl (3S) -3- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -4-fluoropiperidine-1-carboxylate (51)
Figure BDA0003034737920000581
The method comprises the following steps: methyl 3- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -4-hydroxypiperidine-1-carboxylate (51a)
Add NaBH to compound 46g (100.0mg,0.21mmol) in methanol (4mL) at 0 deg.C under nitrogen4(21.6mg,0.64 mmol). After stirring the mixture at room temperature for 12 h, LCMS showed the reaction was complete. Adding H to the mixture2O (15mL), extracted with DCM (20 mL. times.3), and the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the title compound 51a (100.0mg, yield: 89.6%).
MS(ESI)m/z=472.9[M+H]+.
1H NMR(400MHz,CDCl3)δ8.10(d,J=7.0Hz,1H),7.85(d,J=7.0Hz,1H),7.42-7.39(m,3H),6.74-6.71(m,1H),3.97(br d,J=19.6Hz,1H),3.70(br d,J=15.3Hz,1H),3.57(br s,3H),3.41-3.19(m,3H),3.03(br s,3H),2.81-2.62(m,2H),2.44(s,3H),2.26-2.19(m,1H),1.86(br d,J=13.3Hz,2H).
Step two: methyl (3S) -3- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -4-fluoropiperidine-1-carboxylate (51)
To compound 51a (95.0mg,0.20mmol) in DCM (5mL) was added diethylaminosulfur trifluoride (123. mu.L, 1.00mmol) at 0 ℃ under nitrogen. After stirring the mixture at room temperature for 24 h, LCMS showed the reaction was complete. Adding H to the mixture2O (15mL), extracted with DCM (20mL × 3), the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by preparative TLC (DCM/MeOH-10/1) to give the crude product (20.0 mg). It was then purified by preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 27% -50%; flow rate: 25mL/min) and lyophilized to give the title compound 51(3.4mg, yield: 3.5%).
MS(ESI)m/z=475.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.69(br d,J=4.8Hz,1H),8.41(d,J=7.3Hz,1H),7.64(d,J=8.1Hz,2H),7.38(s,1H),6.87(br d,J=6.8Hz,1H),4.68-4.46(m,1H),3.69(br s,2H),3.50(s,3H),3.32-3.30(m,3H),2.98-2.88(m,2H),2.82(d,J=4.4Hz,2H),2.39(s,3H),2.02-1.78(m,2H),1.66-1.48(m,1H).
Example 52
Methyl 2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -1, 4-oxazepin-4-carboxylate (52)
Figure BDA0003034737920000591
The method comprises the following steps: tert-butyl-2-formyl-1, 4-oxazepine-4-carboxylic acid ester (52a)
See example 46 for step four for the synthesis of example 52 a. Wherein tert-butyl 2- (hydroxymethyl) -1, 4-oxazepine-4-carboxylate is substituted for compound 46 c.
1H NMR(400MHz,CDCl3)δ9.58(br d,J=13.5Hz,1H),4.25-3.98(m,2H),3.87-3.46(m,3H),3.42-3.13(m,2H),1.84(br s,2H),1.40(br s,9H).
Step two: tert-butyl 2-ethynyl-1, 4-oxazepine-4-carboxylate (52b)
See example 46 for synthesis procedure five. Wherein compound 52a is substituted for compound 46 d.
1H NMR(400MHz,CDCl3)δ4.43(br d,J=2.0Hz,1H),4.18-3.86(m,3H),3.69-3.59(m,1H),3.32(br d,J=9.0Hz,1H),3.15-3.02(m,1H),2.51(br d,J=2.8Hz,1H),2.07-1.84(m,2H),1.49(s,9H).
Step three: methyl 2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -1, 4-oxazepin-4-carboxylate (52c)
2-ethynyl-1, 4-oxaheptane-4-carboxylic acid tert-butyl ester (35.0mg,0.16mmol), 3, 5-difluoro-4-formyl-N-methylbenzamide (30.9mg,0.16mmol), 4-methylpyridin-2-amine (16.8mg,0.16mmol), copper (II) trifluoroethanesulfonate (16.9mg,0.05mmol), copper (I) chloride (I) (II)4.61mg,0.05mmol) was dissolved in a mixed solution of N, N-dimethylacetamide (4.3. mu.L, 0.05mmol) and toluene (1mL), and bubbled with nitrogen for 30 seconds; the mixed reaction solution was stirred at 140 ℃ for 5 hours by microwave reaction. The reaction solution was concentrated under reduced pressure, and the residue was purified by preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 40% -60%; flow rate: 25mL/min) and lyophilized to give the title compound 52c (8.0mg, yield: 10.0%).
Step four: methyl 2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -1, 4-oxazepin-4-carboxylate (52)
Under nitrogen protection, N-diisopropylethylamine (9.6. mu.L, 0.06mmol) was added dropwise to a solution of 52c (8.0mg,0.02mmol) in dichloromethane (2mL) in an ice-water bath, ethyl chloroformate (1.8. mu.L, 0.03mmol) was then added dropwise slowly, the ice bath was removed after the addition was complete, and the reaction was continued at room temperature for 1 hour. After completion of the reaction, the reaction liquid was concentrated under reduced pressure, and the residue was purified by preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% FA), 0% -40%; flow rate: 25mL/min) and lyophilized to give the title compound 52(2.2mg, yield: 23.9%).
MS(ESI)m/z=473.2[M+H]+.
1H NMR(400MHz,MeOD)δ8.43(dd,J=3.6,6.7Hz,1H),7.65-7.57(m,2H),7.36(s,1H),6.89(br d,J=6.8Hz,1H),3.88-3.82(m,1H),3.80-3.60(m,6H),3.30-3.19(m,2H),3.14-3.01(m,3H),2.97(s,3H),2.48(s,3H),1.81(br s,2H).
Example 53
Methyl (S) -2- ((2- (4-cyano-2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (53)
Figure BDA0003034737920000601
Under nitrogen, cuprous cyanide (11.0mg,0.12mmol) was added to a solution of 13a (48.0mg,0.10mmol) in N-methylpyrrolidone (3mL) and the reaction was stirred at 150 ℃ for 1 hour. After completion of the reaction, filtration, the filtrate was purified by preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% FA), 0% -40%; flow rate: 25mL/min) and lyophilized to give the title compound 53(4.0mg, yield: 9.4%).
1H NMR(400MHz,CDCl3)δ8.28-8.11(m,1H),7.42(s,1H),7.34(br d,J=5.6Hz,2H),6.73(br d,J=6.8Hz,1H),3.99-3.74(m,3H),3.69(s,3H),3.60-3.51(m,1H),3.44-3.29(m,1H),3.10-2.83(m,3H),2.67-2.53(m,1H),2.43(s,3H).
Example 54
Methyl (S) -2- ((2- (4-acetylamino-2, 3, 6-trifluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (54)
Figure BDA0003034737920000611
The method comprises the following steps: methyl 4-acetylamino-2, 3, 6-trifluorobenzoate (54a)
Methyl 4-bromo-2, 3, 6-trifluorobenzoate (100.0mg,0.37mmol), acetamide (30.8mg,0.52mmol), palladium acetate (12.6mg,0.06mmol), 4, 5-bis (diphenylphosphino) -9, 9-dimethylxanthene (43.0mg,0.08mmol), and cesium carbonate (242.2mg,0.74mmol) were dissolved in 1, 4-dioxane (1.5mL), and the mixture was bubbled with nitrogen for 30 seconds, and then stirred at 150 ℃ for 35 minutes by microwave reaction. The reaction solution was filtered using celite, and the filtrate was concentrated to give a crude product, which was purified by silica gel column chromatography (ethyl acetate/petroleum ether ═ 0 to 11%, flow rate: 35mL/min) to give the title compound 54a (57.0mg, 62.0%).
Step two: n- (2,3, 5-trifluoro-4- (hydroxymethyl) phenyl) acetamide (54b)
To a dry tetrahydrofuran solution (6mL) in which lithium aluminum hydride (202.7mg,5.34mmol) was suspended was added methyl 4-acetamido-2, 3, 6-trifluorobenzoate (660.0mg,2.67mmol) in portions under ice-water bath and nitrogen protection, and the reaction was stirred for 1.5 hours under ice-water bath. Methanol was added to the reaction solution for quenching, the reaction solution was concentrated, and the crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether: 0 to 25%, flow rate: 35mL/min) to give the title compound 54b (578.0mg, 98.7%).
Step three: n- (2,3, 5-trifluoro-4-formylphenyl) acetamide (54c)
Freshly prepared manganese dioxide (357.3mg,4.21mmol) was added to a solution of 54b (90.0mg,0.40mmol) in tetrahydrofuran (5mL) under nitrogen and stirred at 60 ℃ for 4 h. The reaction solution was filtered, concentrated, and the crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether: 0-55%, flow rate: 35mL/min) to give the title compound 54c (33.0mg, 37.0%).
1H NMR(400MHz,CDCl3)δ10.22(s,1H),8.25-8.16(m,1H),7.87(br s,1H),2.31(s,3H).
Step four: methyl (S) -2- ((2- (4-acetylamino-2, 3, 6-trifluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (54)
The synthetic procedure for example 54 is as for step one in example 4. Wherein compound 54c is substituted for 3, 5-difluoro-4-formyl-N-methylbenzamide, 4-methylpyridin-2-amine is substituted for 4- (trifluoromethyl) pyridin-2-amine, and methyl (2S) -2-ethynylmorpholine-4-carboxylate is substituted for tert-butyl (2S) -2-ethynylmorpholine-4-carboxylate.
MS(ESI)m/z=477.4[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.38(d,J=7.0Hz,1H),7.32(s,1H),6.96(d,J=9.0Hz,2H),6.81(dd,J=1.3,7.0Hz,1H),3.73-3.61(m,4H),3.55(s,3H),3.47(br dd,J=5.6,10.7Hz,2H),3.29-3.22(m,1H),3.18(s,3H),3.01(br d,J=6.3Hz,2H),2.37(s,3H).
Example 55
Methyl (S) -2- ((2- (4- (2, 5-dicarbonylpyrrolidin-1-yl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (55)
Figure BDA0003034737920000621
Under nitrogen, 32b (100.0mg,0.24mmol) and succinic anhydride (72.1mg,0.72mmol) were dissolved in glacial acetic acid (6mL), and the reaction mixture was stirred at 110 ℃ for 12 hours. Reaction solution is reducedConcentrated under pressure, neutralized with sodium bicarbonate solution, extracted with ethyl acetate, concentrated under pressure with organic phase, and the crude product purified by preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3.H2O), 30% -60%; flow rate: 25mL/min) and lyophilized to give the title compound 55(40.4mg, yield: 32.9%).
MS(ESI)m/z=499.2[M+H]+.
1H NMR(400MHz,CD3OD)δ8.41(d,J=7.2Hz,1H),7.35(s,1H),7.26(d,J=8.4Hz,2H),6.88(dd,J=1.6,7.2Hz,1H),3.96-3.69(m,4H),3.67(s,3H),3.60(br s,1H),3.42-3.34(m,1H),3.18-3.06(m,2H),2.90(s,4H),2.65(br t,J=11.6Hz,1H),2.46(s,3H).
Example 56
Methyl (S) -2- ((2- (4- (1, 1-dioxoisothiazolidin-2-yl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (56)
Figure BDA0003034737920000631
See example 30 for the synthetic procedure for example 56. Wherein 1, 1-dioxoisothiazole is used instead of acetamide.
MS(ESI)m/z=521.2[M+H]+.
1H NMR(400MHz,CD3OD)δ8.37(d,J=7.1Hz,1H),7.31(s,1H),7.04-6.97(m,2H),6.84(dd,J=1.6,7.1Hz,1H),3.85(t,J=6.5Hz,2H),3.78(br d,J=12.8Hz,3H),3.65(s,3H),3.61-3.55(m,1H),3.52(t,J=7.4Hz,2H),3.39-3.34(m,1H),3.12-2.98(m,2H),2.89(br s,1H),2.67-2.60(m,1H),2.55(quin,J=6.9Hz,2H),2.46-2.42(m,1H),2.44(s,2H).
Example 57
(S) -1- (4- (3- ((4-acetylmorpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluorophenyl) pyrrolidin-2-one (57)
Figure BDA0003034737920000632
The method comprises the following steps: tert-butyl (S) -2- ((2- (4-bromo-2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (57a)
See example 4 for a step one for the synthesis of compound 57a wherein 4-methyl-2-aminopyridine is substituted for 4- (trifluoromethyl) pyridin-2-amine and 4-bromo-2, 6-difluorobenzaldehyde is substituted for 5-formyl-N, 4-dimethylpyridine-2-carboxamide.
MS(ESI)m/z=522.2[M+H]+.
1H NMR(400MHz,CD3OD)δ8.38(d,J=7.2Hz,1H),7.42(d,J=6.8Hz,2H),7.32(s,1H),6.86(d,J=7.2Hz,1H),3.83-3.66(m,3H),3.58-3.48(m,1H),3.38-3.32(m,1H),3.08-3.02(m,2H),2.81(br.s,1H),2.54(t,J=11.6Hz,1H),2.44(s,3H),1.41(s,9H).
Step two: tert-butyl (S) -2- ((2- (2, 6-difluoro-4- (2-carbonylpyrrolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (57b)
See example 37 for a synthetic procedure for compound 57 b. Wherein 2-pyrrolidone is used for replacing 2-piperidone, and the compound 57a is used for replacing the compound 13 a.
MS(ESI)m/z=527.2[M+H]+.
1H NMR(400MHz,CDCl3)δ8.19(d,J=7.2Hz,1H),7.44(d,J=10.0Hz,3H),6.68(d,J=5.6Hz,1H),3.88(t,J=6.8Hz,2H),3.78(br s,3H),3.56(br s,1H),3.47-3.31(m,1H),3.05-2.92(m,2H),2.83(br s,1H),2.68(t,J=7.6Hz,2H),2.53(dd,J=10.8,13.2Hz,1H),2.42(s,3H),2.26-2.17(m,2H),1.43(s,9H).
Step three: 1- [3, 5-difluoro-4- (7-methyl-3- { [ (2S) -morpholin-2-yl ] methyl } imidazo [1,2-a ] pyridin-2-yl) phenyl ] pyrrolidin-2-one hydrochloride (57c)
Compound 57b (430.0mg,0.82mmol) was dissolved in dichloromethane (3mL), 4M 1,4 dioxane hydrochloride solution (1.0mL) was added dropwise, the reaction was stirred at room temperature for 2 hours, and concentrated under reduced pressure to give compound 57c (382.2mg, yield: 91%).
MS(ESI)m/z=427.2[M+H]+.
1H NMR(400MHz,DMSO-d6)δ9.49(br s,2H),8.84(d,J=7.2Hz,1H),7.84-7.67(m,3H),7.41(d,J=7.2Hz,1H),3.91(br t,J=7.2Hz,3H),3.76(br dd,J=3.6,12.8Hz,1H),3.58(br s,1H),3.53(br s,1H),3.31-3.16(m,2H),3.05(d,J=12.4Hz,1H),2.93-2.70(m,2H),2.60(t,J=8.0Hz,2H),2.56(s,3H),2.17-2.05(m,2H).
Step four: (S) -1- (4- (3- ((4-acetylmorpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluorophenyl) pyrrolidin-2-one (57)
To compound 57c (30.0mg,0.065mmol) in DMF (1mL) was added acetic acid (7.8mg,0.13mmol), HATU (49.3mg,0.13mmol) and N, N-diisopropylethylamine (43. mu.L, 0.26mmol) in that order. After stirring at room temperature for 1 hour, the reaction mixture was directly purified by C18 reverse phase chromatography (acetonitrile/water (containing 0.05% ammonia water) ═ 10-75%) and lyophilized to give the title compound 57(14.7mg, yield: 48%).
MS(ESI)m/z=469.4[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.40(dd,J=7.1,11.1Hz,1H),7.60(br d,J=10.0Hz,2H),7.33(s,1H),6.86-6.79(m,1H),4.16-3.98(m,1H),3.88(t,J=7.0Hz,2H),3.74-3.62(m,2H),3.56(br d,J=13.2Hz,1H),3.32-3.23(m,1H),3.04-2.99(m,2H),2.77(br dd,J=10.6,12.8Hz,1H),2.57(br s,2H),2.37(s,3H),2.31-2.21(m,1H),2.08(quin,J=7.5Hz,2H),1.91(d,J=9.8Hz,3H).
Example 58
(S) -1- (3, 5-difluoro-4- (7-methyl-3- ((4-propionylmorpholin-2-yl) methyl) imidazo [1,2-a ] pyridin-2-yl) phenyl) pyrrolidin-2-one (58)
Figure BDA0003034737920000651
See example 57, step four for the synthetic procedure for compound 58. In which propionic acid is substituted for acetic acid.
MS(ESI)m/z=483.5[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.40(br dd,J=7.3,10.5Hz,1H),7.60(d,J=10.3Hz,2H),7.33(s,1H),6.82(br t,J=5.2Hz,1H),4.16-4.01(m,1H),3.88(br t,J=7.0Hz,2H),3.74-3.55(m,2H),3.46(br s,1H),3.11-2.93(m,3H),2.62-2.51(m,3H),2.37(s,3H),2.34-2.02(m,5H),0.91(t,J=7.4Hz,3H).
Example 59
(S) -1- (4- (3- ((4- (cyclopropylcarbonyl) morpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluorophenyl) pyrrolidin-2-one (59)
Figure BDA0003034737920000652
The synthetic procedure for example 59 is as in step four of example 57, with cyclopropanecarboxylic acid replacing acetic acid.
MS(ESI)m/z=495.4[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.41(br s,1H),7.60(d,J=10.0Hz,2H),7.34(s,1H),6.83(br d,J=6.0Hz,1H),4.11(br d,J=12.5Hz,1H),4.02(br d,J=11.0Hz,1H),3.89(t,J=7.0Hz,2H),3.74(br d,J=10.8Hz,1H),3.57-3.42(m,1H),3.05(br s,2H),2.57(t,J=8.0Hz,2H),2.52-2.50(m,3H),2.38(s,3H),2.09(quin,J=7.5Hz,2H),1.93-1.75(m,1H),0.67(br d,J=6.8Hz,4H).
Example 60
(S) -1- (4- (3- ((4- (2-cyclopropylacetyl) morpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluorophenyl) pyrrolidin-2-one (60)
Figure BDA0003034737920000661
The synthetic procedure for example 60 is as in step four of example 57, with cyclopropylacetic acid replacing acetic acid.
1H NMR(400MHz,DMSO-d6)δ9.49(br s,2H),8.84(d,J=7.2Hz,1H),7.84-7.67(m,3H),7.41(br d,J=7.2Hz,1H),3.91(br t,J=7.2Hz,3H),3.76(br dd,J=3.6,12.8Hz,1H),3.58(br s,1H),3.53(br s,1H),3.31-3.16(m,2H),3.05(br d,J=12.4Hz,1H),2.93-2.70(m,2H),2.60(t,J=8.0Hz,2H),2.56(s,3H),2.17-2.05(m,2H).
Example 61
(S) -1- (4- (3- ((4- (2, 2-difluoropropionyl) morpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluorophenyl) pyrrolidin-2-one (61)
Figure BDA0003034737920000662
The synthetic procedure for example 61 is as in step four of example 57, with 2, 2-difluoropropionic acid substituted for acetic acid.
MS(ESI)m/z=519.4[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.40(br t,J=6.1Hz,1H),7.60(br d,J=10.1Hz,2H),7.34(s,1H),6.82(br d,J=6.7Hz,1H),4.13-3.97(m,1H),3.90-3.73(m,4H),3.54(br d,J=7.3Hz,1H),3.22-3.13(m,1H),3.09-3.01(m,2H),2.95-2.79(m,1H),2.61-2.51(m,3H),2.38(s,3H),2.09(q,J=7.4Hz,2H),1.82-1.63(m,3H).
Example 62
(S) -3- (2- ((2- (2, 6-difluoro-4- (2-carbonylpyrrolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholino) -3-carbonylpropionitrile (62)
Figure BDA0003034737920000671
The synthetic procedure for example 62 is as in step four of example 57, with 2-cyanoacetic acid replacing acetic acid.
MS(ESI)m/z=494.5[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.40(br dd,J=7.3,10.5Hz,1H),7.60(d,J=10.3Hz,2H),7.33(s,1H),6.82(br t,J=5.2Hz,1H),4.16-4.01(m,1H),3.88(br t,J=7.0Hz,2H),3.74-3.55(m,2H),3.46(br s,1H),3.11-2.93(m,3H),2.62-2.51(m,3H),2.37(s,3H),2.34-2.02(m,5H),0.91(t,J=7.4Hz,3H).
Example 63
(S) -2- ((2- (2, 6-difluoro-4- (2-carbonylpyrrolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -N-methylmorpholine-4-carboxamide (63)
Figure BDA0003034737920000672
57c (30.0mg,0.065mmol) was dissolved in dichloromethane (1mL) under ice-water bath and nitrogen blanket, and methylaminocarbonyl chloride (9.1mg,0.097mmol) and N, N-diisopropylethylamine (32. mu.L, 0.194mmol) were added sequentially. After the completion of the dropwise addition, the reaction was carried out at room temperature for 2 hours. The reaction was concentrated, and the crude product was directly purified by C18 reverse phase chromatography (acetonitrile/water (containing 0.05% ammonia) ═ 10-75%) and lyophilized to give the title compound 63(13.1mg, yield: 42%).
MS(ESI)m/z=484.4[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.37(d,J=7.0Hz,1H),7.60(d,J=10.3Hz,2H),7.33(s,1H),6.81(dd,J=1.4,7.2Hz,1H),6.39(br d,J=4.3Hz,1H),3.88(t,J=7.0Hz,2H),3.73(br d,J=12.5Hz,1H),3.66(br dd,J=2.1,11.4Hz,1H),3.56(br d,J=13.1Hz,1H),3.22-3.12(m,1H),3.07-2.98(m,1H),2.97-2.89(m,1H),2.67(dt,J=3.3,12.3Hz,1H),2.57(t,J=8.0Hz,2H),2.52(s,2H),2.51(br s,3H),2.37(s,3H),2.08(q,J=7.5Hz,2H).
Example 64
Tert-butyl (S) -2- ((2- (2, 6-difluoro-4- (2-carbonylpyrrolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (64)
Figure BDA0003034737920000681
Example 64 is 57b from example 57.
1H NMR(400MHz,CDCl3)δ8.19(br d,J=7.2Hz,1H),7.44(br d,J=10.0Hz,3H),6.68(br d,J=5.6Hz,1H),3.88(t,J=6.8Hz,2H),3.78(br s,3H),3.56(br s,1H),3.47-3.31(m,1H),3.05-2.92(m,2H),2.83(br s,1H),2.68(t,J=7.6Hz,2H),2.53(dd,J=10.8,13.2Hz,1H),2.42(s,3H),2.26-2.17(m,2H),1.43(s,9H).
Example 65
3-methyloxetan-3-yl (S) -2- ((2- (2, 6-difluoro-4- (2-carbonylpyrrolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (65)
Figure BDA0003034737920000682
The method comprises the following steps: 3-Methyloxabutan-3-yl (4-nitrophenyl) carbonate (65a)
Under the ice bath condition, 3-methyloxetan-3-ol (176.0mg, 2.00mmol) is dissolved in anhydrous tetrahydrofuran (3mL), sodium hydride (120.0mg, 3.00mmol) is added in batches, 4-nitrophenyl chloroformate (443.4mg, 2.20mmol) is added in batches, the ice bath is removed, and the reaction is carried out at room temperature for 2 hours; saturated aqueous ammonium chloride solution was added dropwise, extraction was performed with dichloromethane, the phases were separated, the organic phase was concentrated, and the crude product was purified by silica gel column chromatography (12g, ethyl acetate/petroleum ether ═ 0 to 25%, flow rate: 35mL/min) via the crude product and lyophilized to give the title compound 65a (257.0mg, 50.8%).
Step two: 3-methyloxetan-3-yl (S) -2- ((2- (2, 6-difluoro-4- (2-carbonylpyrrolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (65)
A solution of 65a (50.0mg, 0.20mmol), 57C (64.8mg, 0.16mmol) and triethylamine (76.9mg, 0.76mmol) in dichloromethane (5mL) was stirred at room temperature for 3h, the reaction was concentrated and the crude product was directly subjected to preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3.H2O), 30% -65%; flow rate: 25mL/min) and lyophilized to give the title compound 65(60.0mg, yield: 59.3%).
Example 66
(S) -1- (4- (3- ((4- (ethylsulfonyl) morpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluorophenyl) pyrrolidin-2-one (66)
Figure BDA0003034737920000691
57C (35.1mg, 0.10mmol), N-diisopropylethylamine (32. mu.L, 0.19mmol) were dissolved in dichloromethane (3mL) under ice-bath conditions, ethylsulfonyl chloride (12.5mg,0.097mmol) was stirred at room temperature for 2 hours, the reaction was concentrated, and the crude product was directly subjected to preparative HPLC (column: Boston Prime C18150X 30mm X5. mu.m; mobile phase: acetonitrile/water (0.05%)
NH3·H2O), 30% -65%; flow rate: 25mL/min) and lyophilized to give the title compound 66(15.6mg, yield: 46.4%).
1H NMR(400MHz,DMSO-d6)δ8.42(d,J=7.0Hz,1H),7.60(d,J=10.3Hz,2H),7.34(s,1H),6.83(dd,J=1.5,7.0Hz,1H),6.85-6.80(m,1H),3.89(t,J=7.0Hz,2H),3.79-3.72(m,1H),3.61-3.53(m,1H),3.33-3.22(m,2H),3.05(br d,J=6.3Hz,2H),3.02-2.90(m,2H),2.84-2.75(m,1H),2.60-2.55(m,2H),2.51(d,J=1.8Hz,2H),2.38(s,3H),2.09(q,J=7.5Hz,2H),1.16(t,J=7.4Hz,3H).
Example 67
Methyl (S) -2- ((2- (2-chloro-4- (2-carbonylpyrrolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (67)
Figure BDA0003034737920000692
The method comprises the following steps: (S) -1- (3-chloro-4- (7-methyl-3- (morpholin-2-ylmethyl) imidazo [1,2-a ] pyridin-2-yl) phenyl) pyrrolidin-2-one (67a)
See example 10, step three, for the synthesis of compound 67 a. Wherein 4-bromo-2-chlorobenzaldehyde is used for replacing 5-formyl-N, 4-dimethylpyridine-2-formamide, and (2S) -2-ethynylmorpholine-4-carboxylic acid methyl ester is used for replacing (2S) -2-ethynylmorpholine-4-carboxylic acid tert-butyl ester.
MS(ESI)m/z=478.1[M+H]+.
1H NMR(400MHz,CDCl3)δ8.11(d,J=7.2Hz,1H),7.67(d,J=1.6Hz,1H),7.48(dd,J=2.0,8.0Hz,1H),7.40-7.33(m,2H),6.68(br d,J=6.8Hz,1H),4.37(dd,J=2.8,6.5Hz,1H),3.67(s,3H),3.60-3.50(m,2H),3.46-3.33(m,2H),3.04-2.96(m,2H),2.88(br s,1H),2.60-2.48(m,1H),2.42(s,3H).
Step two: methyl (S) -2- ((2- (2-chloro-4- (2-carbonylpyrrolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (67)
See example 40 for synthetic procedure for example 67. Wherein compound 67a is substituted for compound 13 a.
MS(ESI)m/z=483.2[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.38(d,J=7.0Hz,1H),7.99(d,J=2.3Hz,1H),7.66(dd,J=2.0,8.5Hz,1H),7.48(d,J=8.3Hz,1H),7.33(s,1H),6.80(dd,J=1.5,7.0Hz,1H),3.90(t,J=7.0Hz,2H),3.72-3.60(m,3H),3.54(s,3H),3.49(br d,J=7.5Hz,1H),3.24(dt,J=2.4,11.6Hz,1H),3.02(br d,J=6.3Hz,2H),2.81(br s,1H),2.58-2.51(m,3H),2.37(s,3H),2.09(q,J=7.5Hz,2H).
Example 68
(S) -1- (4- (3- ((4-acetylmorpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3-methylphenyl) pyrrolidin-2-one (68)
Figure BDA0003034737920000701
68 for synthesis see example 67, where 4-bromo-2-chlorobenzaldehyde is replaced with 4-bromo-2-methylbenzaldehyde and methoxycarbonyl chloride is replaced with acetyl chloride.
MS(ESI)m/z=447.3[M+H]+.
Example 69
Methyl (S) -2- ((7-methyl-2- (4- (2-carbonylpyrrolidin-1-yl) -2- (trifluoromethyl) phenyl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (69)
Figure BDA0003034737920000711
Synthesis of example 69 see example 67 wherein 4-bromo-2-chlorobenzaldehyde is replaced with 4-bromo-2-trifluoromethylbenzaldehyde.
MS(ESI)m/z=517.2[M+H]+.
Example 70
Methyl (S) -2- ((2- (2-cyano-4- (2-carbonylpyrrolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (70)
Figure BDA0003034737920000712
67(50.0mg,0.11mmol) and cuprous cyanide (13.9mg,0.16mmol) were dissolved in DMF (3mL) and then heated to 140 ℃ for 12 hours. The reaction solution was directly subjected to preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 30% -65%; flow rate: 25mL/min) and lyophilized to give the title compound 66(8.5mg, yield: 17.3%).
MS(ESI)m/z=474.2[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.44(d,J=7.3Hz,1H),8.23(d,J=2.3Hz,1H),8.06(dd,J=2.3,8.8Hz,1H),7.78(d,J=8.6Hz,1H),7.38(s,1H),6.85(d,J=7.1Hz,1H),3.93(t,J=7.0Hz,2H),3.82(br d,J=12.1Hz,1H),3.74-3.62(m,2H),3.57(s,3H),3.32-3.05(m,4H),2.85(br s,1H),2.56(t,J=8.1Hz,3H),2.38(s,3H),2.15-2.05(m,2H)。
Example 71
Methyl (2S) -2- ((2- (2, 6-difluoro-4- (4-methyl-2-carbonylimidazolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (71)
Figure BDA0003034737920000721
The method comprises the following steps: methyl (2S) -2- ((2- (2, 6-difluoro-4- (3- (1-hydroxypropan-2-yl) ureido) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (71a)
Under the protection of nitrogen and in an ice-water bath, 32b (200.0mg,0.48mmol) and diisopropylethylamine (0.40mL,2.40mmol) were dissolved in dichloromethane (5mL), and then triphosgene (171.1mg,0.58mmol) was added in portions, the ice-water bath was maintained, and the reaction was continued for 1 hour with stirring2-aminopropane-1-ol (0.12mL,1.45mmol) was added dropwise, and after the addition was completed, the reaction was heated to 15 ℃ to continue the reaction for 2 hours. After the reaction was complete, water was added for extraction, the phases were separated and concentrated, and the crude product was purified by preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 30% -65%; flow rate: 25mL/min) and lyophilized to give the title compound 71a (148.0mg,0.29mmol, 59.5%).
Step two: methyl (2S) -2- ((2- (2, 6-difluoro-4- (4-methyl-2-carbonylimidazolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (71)
71a (100.0mg,0.20mmol) is dissolved in tetrahydrofuran (3mL) under the protection of nitrogen and ice-water bath, then potassium tert-butoxide (52.0mg,0.47mmol) is added in portions, then a solution of p-methylbenzenesulfonyl chloride (44.2mg,0.24mmol) in tetrahydrofuran (1mL) is slowly added dropwise, and after the dropwise addition is finished, the reaction is continued for 10 minutes under ice bath stirring. The reaction solution was extracted with ethyl acetate and water, the organic phase was concentrated and the crude product was purified by preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 35% -65%; flow rate: 25mL/min) and lyophilized to give the title compound 71(6.1mg,0.01mmol, 6.0%).
MS(ESI)m/z=500.2[M+H]+.
1H NMR(400MHz,CD3OD)δ8.38(d,J=7.2Hz,1H),7.41(d,J=10.4Hz,2H),7.33(s,1H),6.89-6.83(m,1H),4.15-4.08(m,1H),3.97(td,J=6.0,8.8Hz,1H),3.81(br d,J=11.2Hz,3H),3.67(s,3H),3.54(br dd,J=6.0,9.2Hz,2H),3.43-3.37(m,1H),3.14-3.01(m,2H),2.92(br s,1H),2.63(br s,1H),2.46(s,3H),1.35(d,J=6.0Hz,3H).
Example 72
Methyl (S) -2- ((2- (4- (2, 4-dicarbonylimidazolidin-1-yl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (72)
Figure BDA0003034737920000731
Oriented foodTo a solution of compound 13a (100.0mg,0.21mmol) in 1, 4-dioxane (5mL) were added imidazolidinedione (31.3mg,0.32mmol), Xantphos (0.20g,0.42mmol), and Cs in that order2CO3(135.7mg,0.42mmol) and Pd2(dba)3(24.1mg,0.04 mmol). Stirring was carried out at 110 ℃ for 12 hours under nitrogen. The reaction was concentrated and the crude product was purified by preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 30% -60%; flow rate: 25mL/min) and lyophilized to give the title compound 72(8.1mg,0.02mmol, 7.8%) as a white solid.
MS(ESI)m/z=500.2[M+H]+.
1H NMR:(400MHz,CD3OD)δ8.39(d,J=7.2Hz,1H),7.54-7.48(m,2H),7.36-7.32(m,1H),6.87(d,J=7.2Hz,1H),4.50(s,2H),3.81(br d,J=11.6Hz,3H),3.67(s,3H),3.58(br s,1H),3.42-3.35(m,1H),3.14-3.02(m,2H),2.97-2.78(m,1H),2.62(br s,1H),2.46(s,3H).
Example 73
Methyl (S) -2- ((2- (2, 6-difluoro-4- (2-carbonyl-2, 3-dihydro-1H-imidazol-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (73)
Figure BDA0003034737920000732
See example 37 for synthetic procedure for example 73. Wherein 1, 3-dihydroimidazole-2-ketone is used for replacing 2-piperidone.
MS(ESI)m/z=484.3[M+H]+.
Example 74
Methyl (S) -2- ((2- (2, 6-difluoro-4- (5-carbonyl-4, 5-dihydro-1H-1, 2, 4-triazol-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (74)
Figure BDA0003034737920000741
The method comprises the following steps: (E) -2- (2- (4-bromo-3, 5-difluorophenyl) hydrazinoylidene) acetic acid (74a)
3, 5-difluoro-4-bromophenylhydrazine hydrochloride (2.0g,8.97mmol) was dissolved in water (20mL) under an ice-water bath, concentrated hydrochloric acid (2mL) was added, glyoxylic acid (0.8mL,13.45mmol) was added, the mixed solution was warmed to 20 ℃ and stirred for 1 hour, the reaction solution was cooled to room temperature, a large amount of solid was precipitated, filtered, and the filter cake was washed and dried to obtain the title compound 74a (1.4g,4.52mmol, 50.35%) as a yellow solid.
MS(ESI)m/z=280.9[M+H]+.
1H NMR(400MHz,DMSO-d6)δ12.57(br s,1H),11.51(s,1H),7.18(s,1H),6.92(d,J=8.8Hz,2H).
Step two: 2- (4-bromo-3, 5-difluorophenyl) -2, 4-dihydro-3H-1, 2, 4-triazol-3-one (74b)
74a (1.4g,5.02mmol) was dissolved in toluene (20mL), triethylamine (2.1mL,15.05mmol) and biphenylphosphoramidate (1.7mL,7.53mmol) were added, and the reaction mixture was heated to 120 ℃ for reaction for 2 hours. The reaction was cooled to room temperature, extracted with 2M aqueous sodium hydroxide and dichloromethane, and the aqueous phase was adjusted to pH 2-3 with dilute hydrochloric acid to precipitate a large amount of solid, which was filtered, washed and dried to give the title compound 74b (370.0mg,1.21mmol, 24.04%) as a yellow powdery solid.
MS(ESI)m/z=276.0[M+H]+.
1H NMR(400MHz,DMSO-d6)δ12.25(br s,1H),8.23(s,1H),7.77(br d,J=8.8Hz,2H).
Step three: 2, 6-difluoro-4- (5-carbonyl-4, 5-dihydro-1H-1, 2, 4-triazol-1-yl) benzaldehyde (74c)
Under the condition of ice-water bath, adding sodium hydride (108.7mg,2.72mmol) into a tetrahydrofuran (5mL) solution of 74b (250mg,0.91mmol) in batches, and continuing to react for 1 hour under ice bath after the addition is finished; the reaction mixture was further cooled to-78 ℃, n-butyllithium (0.91mL,2.27mmol) was added dropwise, the reaction was further continued at-78 ℃ for 1 hour after the completion of the addition, saturated aqueous solution of ammonium chloride was added dropwise to quench the reaction mixture, the reaction mixture was extracted with ethyl acetate, the organic phase was dried and concentrated, and the crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether: 0-50%, flow rate: 18mL/min) to give the title compound 74c (100mg,0.40mmol, 44.13%) as a white powdery solid.
MS(ESI)m/z=226.0[M+H]+.
1H NMR(400MHz,DMSO-d6)δ12.35(br s,1H),10.14(s,1H),8.30(s,1H),7.73(br d,J=11.2Hz,2H).
Step four: methyl (S) -2- ((2- (2, 6-difluoro-4- (5-carbonyl-4, 5-dihydro-1H-1, 2, 4-triazol-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (74)
74 Synthesis see example 54, step four, where N- (2,3, 5-trifluoro-4-formylphenyl) acetamide is replaced with 74c
MS(ESI)m/z=485.2[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.42(br s,1H),8.23(br s,1H),7.75(br s,2H),7.35(br s,1H),6.83(br d,J=7.2Hz,1H),3.73-3.59(m,4H),3.55(s,3H),3.04(br s,2H),2.80(br s,1H),2.55(br s,2H),2.38(s,3H).
Example 75
Methyl (S) -2- ((2- (4- (2, 5-dicarbonylimidazolidin-1-yl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (75)
Figure BDA0003034737920000751
The method comprises the following steps: methyl (S) -2- ((2- (2, 6-difluoro-4- (3- (2-methoxy-2-carbonylethyl) ureido) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (75a)
Under the ice-water bath condition, triphosgene (86.0mg,0.29mmol) was added to 13a (100.0mg,0.24mmol) and diisopropylethylamine (0.2mL,1.20mmol) in portions in dichloromethane (4mL), the reaction mixture was reacted for 1 hour under the ice-water bath condition, then glycine methyl ester (60.3mg,0.48mmol) was added, the temperature of the reaction mixture was raised to 15 ℃ and the reaction was continued for 2 hours. After the reaction was complete, water and dichloromethane were added for extraction, the organic phase was concentrated by drying and the crude product was purified by preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 20% -60%; flow ofFast: 25mL/min) and lyophilized to give the title compound 75a (100.0mg,0.15mmol, 62.7%).
MS(ESI)m/z=532.1[M+H]+.
1H NMR(400MHz,CDCl3)δ9.48(br s,1H),8.30(br s,1H),7.49(br s,1H),7.09(br d,J=9.6Hz,2H),6.81(br s,1H),4.04(br s,2H),3.84-3.80(m,2H),3.95-3.78(m,4H),3.75-3.74(m,3H),3.69(s,3H),3.60-3.59(m,1H),3.41-3.40(m,1H),2.97-2.96(m,3H),2.92-2.91(m,1H),2.65-2.63(m,1H),2.47-2.46(m,3H),2.44-2.43(m,1H).
Step two: methyl (S) -2- ((2- (4- (2, 5-dicarbonylimidazolidin-1-yl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (75)
75a (75mg,0.1411mmol) and p-toluenesulfonic acid (72.9mg,0.43mmol) in toluene (20mL) were reacted at 120 ℃ for 12 h, the reaction was complete and concentrated under reduced pressure to give the crude product which was purified by preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 25% -65%; flow rate: 20mL/min) and lyophilized to give the title compound 75(22.0mg,0.11mmol, 31.0%).
Example 76
Methyl (S) -2- ((2- (2, 6-difluoro-4- (2-carbonyltetrahydropyrimidin-1 (2H) -yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (76)
Figure BDA0003034737920000761
Example 76 the synthesis of example 72 was seen with propylene urea in place of imidazolidinyl dione.
MS(ESI)m/z=500.3[M+H]+.
1H NMR(400MHz,CD3OD)δ8.37(d,J=7.0Hz,1H),7.31(s,1H),7.18(d,J=9.6Hz,2H),6.84(d,J=6.8Hz,1H),3.90-3.73(m,5H),3.65(s,3H),3.57(br.s,1H),3.39(t,J=6.0Hz,2H),3.36(br.s,1H),3.13-2.98(m,2H),2.88(br.s,1H),2.63(br.s,1H),2.44(s,3H),2.12(q,J=6.0Hz,2H).
Example 77
Methyl (S) -2- ((2- (4- (2, 4-dicarbonyltetrahydropyrimidin-1 (2H) -yl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (77)
Figure BDA0003034737920000762
Example 77 the synthesis of example 72 was seen with dihydro uracil replacing imidazolidinyl dione.
MS(ESI)m/z=514.2[M+H]+.
1H NMR(400MHz,CD3OD)δ8.39(d,J=7.0Hz,1H),7.32(s,1H),7.28-7.21(m,2H),6.85(dd,J=1.5,7.0Hz,1H),3.98(t,J=6.7Hz,2H),3.87-3.74(m,3H),3.65(s,3H),3.57(br s,1H),3.40-3.34(m,1H),3.14-3.00(m,2H),2.96-2.87(m,1H),2.85(t,J=6.7Hz,2H),2.63(br s,1H),2.45(s,3H).
Example 78
(S) -1- (4- (3- ((4-acetylmorpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluorophenyl) imidazolidin-2-one (78)
Figure BDA0003034737920000771
Example 78 the synthesis of example 57 was followed, substituting 2-imidazolidinone for 2-pyrrolidone and acetic anhydride for methyl chloroformate.
MS(ESI)m/z=470.3[M+H]+.
Example 79
(S) -1- (4- (3- ((4-acetylmorpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3-chlorophenyl) imidazolidin-2-one (79)
Figure BDA0003034737920000772
Example 79 the synthesis of example 57 was followed, replacing 4-bromo-2, 6-difluorobenzaldehyde with 4-bromo-2-chlorobenzaldehyde and 2-imidazolidinone with 2-pyrrolidone.
MS(ESI)m/z=468.2[M+H]+.
Example 80
(S) -1- (4- (3- ((4-acetylmorpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3-methylphenyl) imidazolidin-2-one (80)
Figure BDA0003034737920000781
Example 80 the synthesis of example 79, wherein 4-bromo-2-chlorobenzene (formaldehyde) aldehyde is replaced by 4-bromo-2-methylbenzene (formaldehyde).
MS(ESI)m/z=448.3[M+H]+.
Example 81
Methyl (S) -2- ((2- (2, 6-difluoro-4- (2-carbonylpyridin-1 (2H) -yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (81)
Figure BDA0003034737920000782
Example 81 the synthesis of example 37 was followed, replacing 2-piperidone with 2-hydroxypyridine.
MS(ESI)m/z=495.2[M+H]+.
1H NMR(400MHz,CD3OD)δ8.42(d,J=6.8Hz,1H),7.73(d,J=6.4Hz,1H),7.65(t,J=8.0Hz,1H),7.40-7.29(m,3H),6.87(d,J=7.6Hz,1H),6.67(d,J=9.2Hz,1H),6.53(t,J=6.8Hz,1H),3.92-3.74(m,3H),3.65(s,3H),3.62-3.45(m,1H),3.42-3.35(m,1H),3.15-3.07(m,2H),2.91(s,1H),2.66(s,1H),2.46(s,3H).
Example 82
(S) -1- (2- ((2- (2, 6-difluoro-4- (3-hydroxyoxetan-3-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholino) ethan-1-one (82)
Figure BDA0003034737920000783
The method comprises the following steps: (S) -1- (2- ((2- (4-bromo-2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholino) ethan-1-one (82a)
57a (52.0mg, 0.10mmol) in dichloromethane (5mL) was added trifluoroacetic acid (0.5mL), the reaction was stirred at room temperature for 1h, and the reaction was concentrated under reduced pressure to give a crude product; under the ice-water bath condition, the crude product was dissolved in methanol (5mL), triethylamine (0.9mL) was added, acetic anhydride (0.6mL) was added dropwise, and the reaction was carried out for 30 minutes in an ice bath. After the reaction was complete, the reaction was concentrated and the crude product was purified by preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 35% -55%; flow rate: 20mL/min) and lyophilized to give the title compound 82a (11.0mg,0.08mmol, 29.0%).
MS(ESI)m/z=464.2[M+H]+.
Step two: (S) -1- (2- ((2- (2, 6-difluoro-4- (3-hydroxyoxetan-3-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholino) ethan-1-one (82)
To 82a (36.0mg,0.08mmol) of anhydrous tetrahydrofuran (5mL) was added dropwise a solution of n-butyllithium in n-hexane (80.0. mu.L, 0.12mmol) at-78 ℃, 3-oxetanone (7.0. mu.L, 0.12mmol) was added dropwise immediately after completion of the addition, and the reaction was terminated for 1 hour. After the reaction is complete, the reaction is brought to room temperature, quenched by addition of saturated aqueous ammonium chloride solution, extracted with dichloromethane, the organic phase is concentrated and the crude product is purified by preparative HPLC (column: Boston Prime C18150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 30% -65%; flow rate:30mL/min) and lyophilized to give the title compound 82(36.0mg, 29.0%).
MS(ESI)m/z=458.3[M+H]+.
1H NMR(400MHz,CD3OD)δ8.41-8.34(m,1H),8.07(s,2H),7.84(s,2H),7.27(s,2H),7.11(br d,J=8.0Hz,1H),6.83(s,1H),6.57(s,2H),5.45(br dd,J=7.3,13.8Hz,2H),4.31-4.16(m,1H),3.63(s,2H),3.19-2.98(m,4H),2.66(s,1H),2.47-2.36(m,1H),2.44(s,2H),2.29(s,3H),2.04(s,2H),1.97(s,1H).
Example 83
Methyl (S) -2- ((2- (2, 6-difluoro-4- (methylsulfonylamino) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (83)
Figure BDA0003034737920000791
Under ice-water bath conditions, 13a (20.0mg, 0.05mmol) and N, N-diisopropylethylamine (22.0. mu.L, 0.14mmol)
Adding dropwise methylsulfonyl chloride (5.0 μ L,0.06mmol) into dichloromethane (2mL), heating to 40 deg.C for 3 hr, concentrating the reaction solution under reduced pressure, and subjecting the crude product to preparative HPLC (column: Boston Prime C18150 × 30mm × 5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 35% -65%; flow rate:30mL/min) and lyophilized to give the title compound 83(3.0mg, 13.7%).
MS(ESI)m/z=495.3[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.38(d,J=7.0Hz,1H),7.32(s,1H),6.96(d,J=9.0Hz,2H),6.81(dd,J=1.3,7.0Hz,1H),3.73-3.61(m,4H),3.55(s,3H),3.47(br dd,J=5.6,10.7Hz,2H),3.29-3.22(m,1H),3.18(s,3H),3.01(br d,J=6.3Hz,2H),2.37(s,3H).
Example 84
Methyl 5- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -3, 3-difluoropiperidine-1-carboxylate (84)
Figure BDA0003034737920000801
The method comprises the following steps: benzyl 3, 3-difluoro-5- (hydroxymethyl) piperidine-1-carboxylate (84a)
To a mixed solution of 1-benzyl 3-methyl 5, 5-difluoropiperidine-1, 3-dicarboxylate (300.0mg, 0.96mmol) in tetrahydrofuran (8mL) and ethanol (8mL) was added sodium borohydride (97.2mg, 2.88mmol) and calcium chloride (159.4mg, 1.44mmol) in an ice-water bath, followed by warming and stirring at 20 ℃ for 12 hours. After the reaction was completed, the reaction mixture was concentrated, and the crude product was extracted with ethyl acetate and water, the organic phase was dried and concentrated, and the crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether 0 to 15%, flow rate:30mL/min) to obtain compound 84a (260.0mg, 85.7%).
MS(ESI)m/z=286.2[M+H]+.
1H NMR(400MHz,CDCl3)δ7.37-7.20(m,5H),5.20-5.00(m,2H),4.24-3.81(m,2H),3.51(br s,2H),3.41-2.75(m,2H),2.21-1.96(m,2H),1.78(br s,1H).
Step two: benzyl 3, 3-difluoro-5-formylpiperidine-1-carboxylic acid ester (84b)
To 84a (260.0mg, 0.92mmol) in dichloromethane (10mL) was added dropwise a deslimatin reagent (579.8mg, 1.37mmol), the reaction was reacted at 20 ℃ for 2 hours, after completion of the reaction, quenched with aqueous sodium sulfite solution, extracted with dichloromethane, dried and concentrated, and the crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether: 0-35%, flow rate:30mL/min) to give compound 84b (250.0mg,0.89mmol, 96.8%).
Step three: benzyl 5-ethynyl-3, 3-difluoropiperidine-1-carboxylic acid ester (84c)
To a solution of 84b (215.0mg,0.76mmol) in methanol (10mL) were added potassium carbonate (420.0mg, 3.04mmol) and dimethyl (1-diazo-2-oxo-propanol) -phosphonate (175.0mg,0.91mmol), respectively, and the reaction was reacted at 20 ℃ for 12 hours.
The reaction solution was concentrated, extracted with ethyl acetate and water, the organic phase was dried and concentrated, and the crude product was purified by silica gel column chromatography (ethyl acetate/petroleum ether: 5-35%, flow rate:30mL/min) to give compound 84c (125mg,0.43mmol, 56.0%).
MS(ESI)m/z=280.2[M+H]+.
1H NMR(400MHz,CDCl3)δ7.44-7.31(m,5H),5.18(br s,2H),4.53-4.22(m,2H),3.20-2.70(m,3H),2.51(br s,1H),2.05-1.83(m,1H).
Step four: benzyl 5- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -3, 3-difluoropiperidine-1-carboxylate (84d)
Synthesis of 84d see synthesis of 4a in example 4, where tert-butyl (2S) -2-ethynylmorpholine-4-carboxylate is replaced with 84c, 4- (trifluoromethyl) pyridin-2-amine is replaced with 4-methylpyridin-2-amine, and 3, 5-difluoro-4-formyl-N-methylbenzamide is replaced with 3, 5-difluoro-4-formyl-N-methylbenzamide.
MS(ESI)m/z=569.5[M+H]+.
Step five: 4- (3- ((5, 5-difluoropiperidin-3-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluoro-N-methylbenzamide (84e)
Compound 4d (48.0mg,0.084mmol) was dissolved in trifluoroacetic acid (3mL) and the reaction stirred at 60 ℃ for 3h, LCMS indicated complete reaction. The filtrate was concentrated under reduced pressure to give the title compound 84e (46.0mg, yield: 99.4%).
MS(ESI)m/z=435.2[M+H]+.
Step six: methyl 5- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -3, 3-difluoropiperidine-1-carboxylate (84)
Synthesis of 84 see synthesis of 63 in example 63, where 57c was replaced with 84e and methyl chloroformate was substituted for methylcarbamoyl chloride.
MS(ESI)m/z=493.4[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.70(br d,J=4.4Hz,1H),8.46(br d,J=6.8Hz,1H),7.67(br d,J=8.0Hz,2H),7.39(s,1H),6.88(br d,J=6.6Hz,1H),3.96(br s,1H),3.76(br s,1H),3.52(s,3H),3.28-3.05(m,3H),2.98-2.86(m,2H),2.82(br d,J=4.4Hz,3H),2.39(s,3H),1.94(br s,2H).
Example 85
(S) -1- (4- (3- ((4-acetylmorpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3-chlorophenyl) pyrrolidin-2-one (85)
Figure BDA0003034737920000821
The method comprises the following steps: tert-butyl (S) -2- ((2- (4-bromo-2-chlorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (85a)
See example 10, step three, for the synthetic procedure for compound 85 a. Wherein 4-bromo-2-chlorobenzaldehyde is used for replacing 5-formyl-N, 4-dimethylpyridine-2-formamide.
MS(ESI)m/z=520.1[M+H]+.
Step two: tert-butyl (S) -2- ((2- (2-chloro-4- (2-carbonylpyrrolidin-1-yl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (85b)
See example 82 for synthetic procedure for example 85 b. Wherein compound 85a is substituted for compound 57 a.
MS(ESI)m/z=462.0[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.39(dd,J=7.2,14.9Hz,1H),8.02-7.96(m,1H),7.65(dd,J=2.0,8.5Hz,1H),7.55-7.44(m,1H),7.32(s,1H),6.80(t,J=6.1Hz,1H),4.05(br t,J=11.5Hz,1H),3.89(t,J=7.0Hz,2H),3.74-3.51(m,3H),3.07-3.02(m,2H),2.82-2.70(m,1H),2.37(s,3H),1.91(d,J=8.3Hz,3H).
Step three: (S) -1- (4- (3- ((4-acetylmorpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3-chlorophenyl) pyrrolidin-2-one (85)
See example 40 for synthetic procedure for example 85. Wherein compound 85b is substituted for compound 13 a.
MS(ESI)m/z=467.2[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.46-8.35(m,1H),7.82(br s,1H),7.70(br d,J=11.8Hz,1H),7.33(s,1H),6.82(t,J=5.9Hz,1H),4.16-4.00(m,1H),3.89(t,J=7.0Hz,2H),3.73-3.39(m,3H),3.29-3.10(m,1H),3.06-2.86(m,3H),2.61-2.52(m,3H),2.37(s,3H),2.08(quin,J=7.5Hz,2H),1.90(d,J=16.1Hz,3H).
Example 86
(S) -1- (4- (3- ((4-acetylmorpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3-chloro-5-fluorophenyl) pyrrolidin-2-one (86)
Figure BDA0003034737920000831
See example 85 for a synthetic procedure for compound 86 wherein 4-bromo-2-chloro-6-fluorobenzaldehyde is substituted for 4-bromo-2-chlorobenzaldehyde in step one.
MS(ESI)m/z=485.4[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.46-8.35(m,1H),7.82(br s,1H),7.70(br d,J=11.8Hz,1H),7.33(s,1H),6.82(t,J=5.9Hz,1H),4.16-4.00(m,1H),3.89(t,J=7.0Hz,2H),3.73-3.39(m,3H),3.29-3.10(m,1H),3.06-2.86(m,3H),2.61-2.52(m,3H),2.37(s,3H),2.08(q,J=7.5Hz,2H),1.90(d,J=16.1Hz,3H).
Example 87
(S) -1- (4- (3- ((4-acetylmorpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3, 5-dichlorophenyl) pyrrolidin-2-one (87)
Figure BDA0003034737920000832
See example 85 for a synthetic procedure for compound 87 wherein 4-bromo-2, 6-dichlorobenzaldehyde is substituted for 4-bromo-2-chlorobenzaldehyde in step one.
MS(ESI)m/z=500.5[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.46-8.35(m,1H),7.82(br s,1H),7.70(br d,J=11.8Hz,1H),7.33(s,1H),6.82(t,J=5.9Hz,1H),4.16-4.00(m,1H),3.89(t,J=7.0Hz,2H),3.73-3.39(m,3H),3.29-3.10(m,1H),3.06-2.86(m,3H),2.61-2.52(m,3H),2.37(s,3H),2.08(q,J=7.5Hz,2H),1.90(d,J=16.1Hz,3H).
Example 88
1- (4- (3- (((2S) -4-acetylmorpholin-2-yl) methyl) -7-chloro-6-fluoroimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluorophenyl) pyrrolidin-2-one (88)
Figure BDA0003034737920000841
The method comprises the following steps: 1- ((2S) -2- ((2- (4-bromo-2, 6-difluorophenyl) -7-chloro-6-fluoroimidazo [1,2-a ] pyridin-3-yl) methyl) morpholin-4-yl) ethan-1-one (88a)
The synthetic procedure for example 88a is as described in example 4, step one. Wherein 4-chloro-5-fluoropyridin-2-amine is substituted for 4- (trifluoromethyl) pyridin-2-amine, 1- ((2S) -2-ethynylmorpholin-4-yl) ethan-1-one is substituted for (2S) -2-ethynylmorpholine-4-carboxylic acid tert-butyl ester, 4-bromo-2, 6-difluorobenzaldehyde is substituted for 5-formyl-N, 4-dimethylpyridine-2-carboxamide.
1H NMR(400MHz,CDCl3)δ8.49-8.41(m,1H),7.71-7.67(m,1H),7.26-7.21(m,2H),4.46-4.36(m,1H),3.95-3.86(m,1H),3.61-3.49(m,2H),3.40(dt,J=2.4,11.8Hz,1H),3.22(dt,J=3.4,12.6Hz,1H),2.99-2.93(m,2H),2.50-2.39(m,1H),2.08(s,2H),2.03(s,1H).
Step two: 1- (4- (3- (((2S) -4-acetylmorpholin-2-yl) methyl) -7-chloro-6-fluoroimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluorophenyl) pyrrolidin-2-one (88)
To a solution of 88a (100.0mg,0.199mmol) and pyrrolidin-2-one (18.6mg,0.219mmol) in 1, 4-dioxane (5mL) under nitrogen was added Cs2CO3(129.6mg,0.398mmol)、Pd(dba)2(22.9mg,0.040mmol) and Xantphos (23.0mg,0.040 mmol). The reaction mixture was then stirred at 95 ℃ for 2.5 hours. LCMS showed reaction complete. The mixture was filtered and concentrated and subjected to preparative HPLC (column: Phenomenex Gemini-NX 75X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O), 21% -61%; flow rate: 25mL/min) and lyophilized to give the title compound 88(32.0mg, yield: 29.7%).
MS(ESI)m/z=507.1[M+H]+.
1H NMR(400MHz,MeOH-d4)δ8.78(dd,J=5.1,6.9Hz,1H),7.74(dd,J=2.7,6.8Hz,1H),7.58(dd,J=2.7,10.3Hz,2H),4.31-4.19(m,1H),3.96(t,J=7.1Hz,2H),3.83(dd,J=2.7,11.7Hz,1H),3.76-3.52(m,2H),3.40(dt,J=2.7,11.9Hz,1H),3.19(dd,J=2.8,12.7Hz,1H),3.15-3.08(m,2H),3.15-3.08(m,1H),2.93(dd,J=10.6,13.0Hz,1H),2.65(t,J=8.1Hz,2H),2.46(dd,J=10.9,13.1Hz,1H),2.21(quin,J=7.5Hz,2H),2.03(d,J=14.2Hz,3H).
Example 89
Methyl (2S) -2- ((7-chloro-2- (2, 6-difluoro-4-methanesulfonylaminophenyl) -6-fluoroimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (89)
Figure BDA0003034737920000851
The method comprises the following steps: methyl (2S) -2- ((2- (4-bromo-2, 6-difluorophenyl) -7-chloro-6-fluoroimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (89a)
The synthetic procedure for example 89a is as described in example 4, step one. Wherein 4-chloro-5-fluoropyridin-2-amine is substituted for 4- (trifluoromethyl) pyridin-2-amine, (2S) -2-ethynylmorpholine-4-carboxylic acid methyl ester is substituted for (2S) -2-ethynylmorpholine-4-carboxylic acid tert-butyl ester, and 4-bromo-2, 6-difluorobenzaldehyde is substituted for 5-formyl-N, 4-dimethylpyridine-2-carboxamide.
MS(ESI)m/z=518.0[M+H]+.
1H NMR(400MHz,CDCl3)δ8.44(d,J=4.8Hz,1H),7.69(d,J=6.9Hz,1H),7.25(d,J=6.9Hz,2H),4.02-3.77(m,3H),3.70(s,3H),3.59(dt,J=2.9,5.5Hz,1H),3.53-3.48(m,1H),3.40(br t,J=11.4Hz,1H),2.93(br d,J=5.8Hz,2H),2.64(br t,J=12.3Hz,1H).
Step two: methyl (2S) -2- ((7-chloro-2- (2, 6-difluoro-4-methanesulfonylaminophenyl) -6-fluoroimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (89)
To a solution of 89a (100.0mg,0.19mmol) and methanesulfonamide (18.3mg,0.19mmol) in 1, 4-dioxane (4mL) under nitrogen was added Cs2CO3(125.6mg,0.38mmol)、Pd(dba)2(11.1mg,0.019mmol) and Xantphos (22.3mg,0.038 mmol). The reaction mixture was then stirred at 100 ℃ for 4 hours. LCMS showed reaction complete. The mixture was filtered and concentrated and subjected to preparative HPLC (column: Phenomenex Gemini-NX 150X 30mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O+10mM NH4HCO3) 28% -48%; flow rate: 25mL/min) and lyophilized to give the title compound 89(15.7mg, yield: 15.3%).
MS(ESI)m/z=533.1[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.77(d,J=5.0Hz,1H),7.74(d,J=6.8Hz,1H),7.02(d,J=9.3Hz,2H),3.93-3.76(m,3H),3.68(s,3H),3.61(br d,J=7.8Hz,1H),3.38(dt,J=2.5,11.8Hz,1H),3.14(s,3H),3.10(br d,J=5.8Hz,2H),2.95(br d,J=15.3Hz,1H),2.68(br s,1H).
Example 90
Methyl (S) -2- ((2- (2, 6-difluoro-4- (N-methylsulfamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (90)
Figure BDA0003034737920000861
The method comprises the following steps: methyl (S) -2- ((2- (4- (chlorosulfonyl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (90a)
Compound 32b (200.0mg,0.48mmol) was dissolved in a mixture of HCl (1mL,12.00mmol) and acetic acid (3mL) and cooled to 0 deg.C, a solution of sodium nitrite (49.7mg,0.72mmol) in water (1mL) was added dropwise with stirring and stirred at 0 deg.C for 0.5 h. The reaction mixture was then added dropwise to cuprous chloride (16.6mg,0.17mmol) and SO at 0 deg.C2Acetic acid saturated solution (5mL) in the suspension mixture. The reaction mixture was stirred at 25 ℃ for 0.5 h and LCMS showed complete consumption of starting material. The reaction mixture was poured into ice water, the resulting mixture was extracted with ethyl acetate (20 mL. times.3), and the combined organic phases were washed with NaSO4Drying and filtration, the filtrate was concentrated to give the title compound 90a (200.0mg, 83.3%).
MS(ESI)m/z=500.0[M+H]+.
Step two: methyl (S) -2- ((2- (2, 6-difluoro-4- (N-methylsulfamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (90)
To a solution of 90a (180.0mg,0.36mmol) in DCM (3mL) was added methylamine (52. mu.L, 0.43mmol, 33% in EtOH) and diisopropylethylamine (119. mu.L, 0.72 mmol). The reaction mixture was stirred at 25 ℃ for 0.5 hour. LCMS showed complete consumption of starting material. The residue obtained is concentrated under reduced pressure by preparative HPLC (column: Boston Prime C18150X 25mm X5 μm; mobile phase: acetonitrile/water (0.05% NH)3·H2O),37%-60%;Flow rate:30mL/min) and lyophilized to give the title compound 90(38.7mg, yield: 21.6%).
MS(ESI)m/z=495.2[M+H]+.
1H NMR(400MHz,DMSO-d6)δ8.44(d,J=7.1Hz,1H),7.79(br d,J=4.0Hz,1H),7.59(d,J=6.5Hz,2H),7.36(s,1H),6.85(dd,J=1.6,7.1Hz,1H),3.77(br d,J=12.3Hz,1H),3.62(br d,J=11.7Hz,3H),3.55(s,3H),3.26-3.16(m,1H),3.14-2.97(m,2H),2.78(br s,1H),2.53(br d,J=3.2Hz,4H),2.38(s,3H).
Biological assay
Test example A, evaluation of in vitro biological Activity
The FLIPR assay screens compounds for antagonistic activity at hP2X3 and hP2X2/3 receptors (changes in calcium flux signaling indicate the effect of the compound on ion channels).
1. Laboratory instruments and materials
Figure BDA0003034737920000871
2. Experimental procedure
1321N1 cells (adherent cells) stably transfected with hP2X3 and hP2X2/3 receptor were digested, centrifuged, resuspended and counted in plating medium (DMEM + 10% DFBS), and cells were adjusted to 2X 105cells/mL, 50. mu.L of cells per well in 384-well Assay Plate, placed in 5% CO2And culturing in an incubator at 37 ℃ for 16-24 hours. 180-fold higher concentration of test Compound (20mM DMSO stock) was made up in DMSO, 500nL per well was added to 384-well Compound Plate, and 30. mu.L of FLIPR buffer (containing 1.26mM Ca) was supplemented2+1 HBSS +2mM CaCl2+20mM HEPES), shaken for 20-40min to mix. 3-fold desired concentration of agonist α, β -meATP (500 nM final hP2X3 cells and 1000nM final hP2X2/3 cells) was formulated with a FLIPR buffer, adding 35. mu.L of agonist per well to another 384-well Compound Plate. The cell plates, which were plated for 16-24 hours, were removed, the cell supernatants were aspirated, and 30. mu.L of Dye (E) was added to each well
Figure BDA0003034737920000882
Calcium 4Assay Kit, FLIPR buffer dilution), incubated for 1 hour. mu.L of compound was added to each well (sample was added to the FLIPR instrument) and after 15 minutes, 22.5. mu.L of agonist was added to each well and the fluorescence signal was detected (excitation wavelength 470nm-495nm, emission wavelength 515nm-575 nm). Taking the difference value between the peak value and the valley value of the signal as basic data, taking the data of the highest concentration of the positive drug as the 100% inhibition rate and taking DMSO data as the 0% inhibition rate, fitting an inhibition effect curve of the compound on software Graphpad Prism 6 and calculating IC50The value is obtained.
TABLE 1 half Inhibitory Concentration (IC) of the compounds of the invention at hP2X3 and hP2X2/3 receptors50)
Figure BDA0003034737920000881
Figure BDA0003034737920000891
Figure BDA0003034737920000901
Figure BDA0003034737920000911
Test example B, CYP inhibition experiment
Representative substrate metabolic responses of human major 5 CYP subtypes (CYP1a2, CYP2C9, CYP2C19, CYP2D6, CYP3a4/5) were evaluated using 150 donor pooled human liver microsomes (purchased from Corning, cat # 452117). The influence of different concentrations of the compounds to be tested on metabolic reactions of phenacetin (CYP1A2), diclofenac sodium (CYP2C9), S-metfungin (CYP2C19), bufuralol hydrochloride (CYP2D6) and midazolam (CYP3A4/5) is determined by liquid chromatography-tandem mass spectrometry (LC/MS/MS).
mu.M phenacetin, 10. mu.M diclofenac sodium, 35. mu. M S-metin, 5. mu.M bufuralol hydrochloride, 3. mu.M midazolam, 1mM NADPH, test compounds (concentrations of 0.1, 0.3, 1,3, 10, 30. mu. mol/L, respectively), or positive compounds or blank controls were incubated with 200. mu.L (100mmol/L phosphate buffer, pH 7.4, containing DMSO in a volume ratio of 0.3%, 0.6% acetonitrile, 0.1% methanol, respectively) of a reaction system mixed with human liver microsomes (0.2mg/mL) at 37 ℃ for 5 minutes. Then 200. mu.L of acetonitrile containing 3% formic acid and 40nM internal standard verapamil was added and centrifuged at 4000rpm for 50 minutes. The mixture was cooled on ice for 20 minutes and centrifuged at 4000rpm for 20 minutes to precipitate the protein. 200 μ L of the supernatant was analyzed by LC/MS/MS.
The peak area was calculated from the chromatogram. The residual activity ratio (%) was calculated by the following formula:
peak area ratio metabolite peak area/internal standard peak area
Residual activity ratio (%) -ratio of peak area of test compound group/peak area of blank group
Half Inhibition of CYP (IC)50) Calculated by Excel XLfit 5.3.1.3.
Measured half Inhibition Concentration (IC) of CYP50) The values are shown in Table 2.
TABLE 2 half Inhibitory Concentration (IC) of a portion of the compounds of the invention against CYP50)
Figure BDA0003034737920000912
Figure BDA0003034737920000921
Test example C detection of human hepatocyte metabolic stability in vitro
The intrinsic clearance of the test compound was calculated by measuring the concentration of the compound in the reaction system using LC/MS/MS, and the in vitro metabolic stability in human hepatocytes was evaluated.
247.5. mu.L of 1X 106cells/mL of a mixture of human hepatocytes (purchased from BiorecamationIVT, cat # S01205) and 2.5. mu.L of 100. mu.M test compound or positive control were added to the incubation plates to initiate the reaction. At 37 ℃ and 600rpmIncubation was performed. Transfer 20 μ L of incubation system to stop plates at 0.5, 5, 15, 30, 45, 60, 80, 100 and 120 minutes, respectively. Then mix by vortexing for 2 minutes. The stop plates were centrifuged at 4000rpm for 20 minutes. Transfer 40. mu.L of the supernatant of each compound to 96 wells into the template, after which 160. mu.L of pure water was added to dilute the sample.
The resulting samples were quantified by ion chromatogram. The residual rate was calculated from the peak area of the test compound or the positive control. Slope k was determined by linear regression of the natural pair values of residual rate versus incubation time using Microsoft Excel.
Intrinsic clearance (in vitro CL)int,μL/min/106Cells) were calculated from the slope values according to the following equation:
in vitro CLint=kV/N
v-incubation volume (0.25 mL);
n-number of cells per well (0.25 × 10)6Cell)
The values of intrinsic clearance of human hepatocytes measured are shown in table 3.
TABLE 3 intrinsic human hepatocyte clearance of some of the compounds of the invention
Figure BDA0003034737920000922
Test example D, rat hepatocyte in vitro metabolic stability assay
The intrinsic clearance of the test compound was calculated by measuring the concentration of the compound in the reaction system using LC/MS/MS and evaluating the metabolic stability in vitro in rat hepatocytes.
247.5. mu.L of 1X 106cells/mL of rat liver cell (purchased from BiorecamationIVT, cat # M00065) mixture and 2.5. mu.L of 100. mu.M test compound or positive control were added to the incubation plates to initiate the reaction. Incubations were performed at 37 ℃ and 900 rpm. Transfer 20 μ L of incubation system to stop plates at 0.5, 5, 15, 30, 45, 60, 80, 100 and 120 minutes, respectively. Then mix by vortexing for 2 minutes. The stop plates were centrifuged at 4000rpm for 20 minutes. Transfer 30. mu.L of supernatant of each compound to 96-well platesAfter that, 120. mu.L of purified water was added to dilute the sample. The resulting samples were quantified by ion chromatogram. The residual rate was calculated from the peak area of the test compound or the positive control. Slope k was determined by linear regression of the natural pair values of residual rate versus incubation time using Microsoft Excel.
Intrinsic clearance (in vitro CL)int,μL/min/106Cells) were calculated from the slope values according to the following equation:
in vitro CLint=kV/N
v-incubation volume (0.25 mL);
n-number of cells per well (0.25 × 10)6Cell)
The intrinsic clearance values of rat hepatocytes are shown in table 4.
TABLE 4 intrinsic clearance of rat hepatocytes, a partial compound of the invention
Figure BDA0003034737920000931
Example E, Caco-2 Permeability test
Determination of apparent permeability coefficient (P) of analytical drug by liquid chromatography tandem mass spectrometry (LC/MS/MS) through Caco-2 cell modelapp)。
The density in Caco-2 cells (purchased from ATCC) was 7.92X 105cells/cm2mu.L of HBSS (25mM HEPES, pH 7.4) containing 10. mu.M of the compound to be tested, 30. mu.M of benzbromone, 20. mu.M of sulfasalazine, was added to the tip of the Transwell chamber of (Transwell, Corning Co.), and 10. mu.L of the sample was immediately taken out into a 96-well plate to which 90. mu.L of HBSS (25mM HEPES, pH 7.4 containing 50. mu.M quinidine, 30. mu.M benzbromone, 20. mu.M of sulfasalazine) had been added as an initial-addition sample, and 800. mu.L of HBSS (25mM HEPES, pH 7.4 containing 50. mu.M quinidine, 30. mu.M of benzbromone, 20. mu.M of sulfasalazine) was added to the base end. Incubate at 37 ℃ for 2 hours, aspirate 10 μ L of sample from the tip into 96-well deep-well plates containing 90 μ L of HBSS (25mM HEPES, pH 7.4, containing 50 μ M quinidine,30 μ M benzbromorone, 20 μ M sulfasalazine) at 45 min and 2 hour time points, respectively. Suction from the substrate end at 45 min and 2 hr time points, respectively100 μ L of sample was placed in a 96-well deep-well plate. Then 3 volumes of pre-cooled internal standard were added per well. Vortex at 1000rmp for 10 min and centrifuge at 4000rpm for 20 min. Samples were taken at 100. mu.L per well and 3 samples were mixed together with 100. mu.L of pure water for LC/MS/MS analysis.
Data were calculated using Microsoft Excel and peak areas were calculated from the chromatograms. The apparent permeability coefficient (Papp) is in cm/s and is calculated using the following formula:
Figure BDA0003034737920000941
CRthe concentration of the compound to be measured at the substrate end (the superscript "120" or "45" is the sampling time in minutes), CDThe concentration of the compound to be measured at the top (superscript "120" or "45" is the sampling time in minutes) and the Area is the membrane surface Area (0.33 cm)2) Time is the total transit time (75 × 60 seconds).
The values of the measured apparent permeability coefficients of Caco-2 cells are shown in Table 5.
TABLE 5 cell apparent permeability coefficient of partial compound of the invention Caco-2
Example numbering Papp(A-B)(10-6,cm/s)
5 4.19
40 21.81
57 4.86
58 12.41
59 14.16
61 16.66
62 0.50
85 3.27
86 4.15
88 12.62
Test example E, in vivo pharmacokinetic experiment in rat
The drug concentration in the plasma of rats at different times after gavage administration of the compound of the present invention was determined by LC/MS/MS method using rats as test animals. The pharmacokinetic behavior of the compounds of the invention in rats was studied and their pharmacokinetic profile was evaluated.
2. Test protocol
2.1 test drugs
Example 5 and example 86.
2.2 test animals
3 male SD rats were healthy 6-8 weeks per group.
2.3 pharmaceutical formulation
Intragastric administration: weighing a certain amount of medicine, adding 0.5% of hydroxypropyl methylcellulose, 0.1% of Tween 80 and 99.4% of water to prepare 1mg/mL white suspension.
2.4 administration
SD rats were fasted overnight and then gavaged with 5mg/kg for example 5 and example 86.
3. Operation of
The compound of the present invention is administered to rats by gavage, and 0.2mL of blood is collected from jugular vein at 0.25, 0.5, 1,2,4, 8, 24 hours after administration, and placed in a test tube containing EDTA-K2, and plasma is separated by centrifugation at 4000rpm for 5 minutes at 4 ℃ and stored at-75 ℃.
Determining the content of the compound to be tested in rat plasma after the drug with different concentrations is administered by gastric lavage: rat plasma at each time point after administration was taken at 50. mu.L, added with 200. mu.L of an acetonitrile solution of internal standard dexamethasone (50ng/mL), vortex mixed for 30 seconds, centrifuged at 4 ℃ at 4700 rpm for 15 minutes, plasma samples were taken supernatant diluted three times with water, and 2.0. mu.L was taken for LC/MS/MS analysis.
4. Pharmacokinetic parameter results
The rat pharmacokinetic parameters for some of the compounds of the invention are as follows:
Figure BDA0003034737920000951

Claims (20)

1. a compound of formula (I) or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure FDA0003034737910000011
wherein the content of the first and second substances,
R1selected from hydrogen, deuterium, alkyl optionally substituted with halogen or deuterium, halogen, alkoxy optionally substituted with halogen or deuterium, hydroxyalkyl;
R2selected from hydrogen, deuterium, halogen, cyano, cyclopropyl, alkyl optionally substituted with halogen or deuterium;
R3selected from the group consisting of alkyl optionally substituted with halogen or cyano, cycloalkyl, heterocyclyl, alkoxy optionally substituted with halogen, amino optionally substituted with alkyl;
X1is selected from N or CH;
X2oxygen, nitrogen, methylene groups, the methylene groups optionally substituted with halogen, cyclic hydrocarbon groups, alkyl groups;
z is selected from:
a) selected from hydrogen, deuterium, halogen, cyano, amino, hydroxy, optionally halogen-substituted alkyl, sulfone, sulfoxide, sulfonamide, C1-3Optionally halogen-substituted alkoxy; or
b)
Figure FDA0003034737910000012
m is selected from 0, 1 or 2, R4、R5Independently selected from the group consisting of: hydrogen, alkyl, alkoxy and cycloalkyl, or R4And R5Together with the nitrogen to which they are attached form a 4-6 membered heterocyclyl, wherein: said heterocyclyl is optionally substituted with halogen, hydroxy, cyano, haloalkyl or alkyl; and when m is 0, R4And R5A combination of (a) is other than hydrogen and methyl; or
c)
Figure FDA0003034737910000013
Heterocyclyl or heteroaryl, said heterocyclyl or heteroaryl optionally substituted with oxo, halogen, hydroxy, carbonyl, alkyl or cyano, said alkyl optionally substituted with halogen; or
d)
Figure FDA0003034737910000014
Wherein R is6Selected from the group consisting of alkyl, alkoxy, aryl, heteroaryl, cycloalkyl, heterocyclyl, cyanoalkyl, cycloalkyloxy, amino optionally substituted with alkyl;
n is an integer selected from 1 to 4.
2. A compound of formula (I) according to claim 1, wherein R1Selected from halogen, C1-3Alkyl radical, C1-3Alkoxy radical, aSaid alkyl or alkoxy being optionally substituted by one or more halogen or deuterium atoms, R2、R3、X1、X2Z, n is as defined in claim 1.
3. A compound of formula (I) according to claim 1, wherein R1、R2、R3、X1、X2N is as defined in claim 1, and Z is selected from:
a) selected from the group consisting of halogen, cyano, amino, sulfone, sulfonamide, alkyl substituted with one or more halogen atoms; or
b)
Figure FDA0003034737910000021
m is selected from 0, 1 or 2, R4、R5Independently selected from the group consisting of: hydrogen, alkyl, alkoxy and cycloalkyl, or R4And R5Together with the nitrogen to which they are attached form a 4-6 membered heterocyclyl, which heterocyclyl is optionally substituted by halogen, hydroxy or alkyl; and when m is 0, R4And R5A combination of (a) is other than hydrogen and methyl; or
c) A 4-6 membered heterocyclyl or heteroaryl group, which heterocyclyl or heteroaryl group is optionally substituted by oxo, halogen, hydroxy, carbonyl, alkyl or cyano, which alkyl group may be optionally substituted by halogen; or
d)
Figure FDA0003034737910000022
Or
e)
Figure FDA0003034737910000023
Wherein R is6Can be selected from C1-3Alkyl radical, C1-3Alkoxy, 5-6 membered aryl or heteroaryl, 3-8 membered cycloalkyl, 3-8 membered heterocyclyl, cyanoalkyl, cycloalkyloxy, amino optionally substituted with alkyl.
4. A compound of formula (I) according to claim 1, which is represented by formula (I-1):
Figure FDA0003034737910000024
wherein R is2aAnd R2bIndependently selected from hydrogen, halogen, C optionally substituted by halogen1-3Alkyl radical, R1、R3、X2Z is defined as in claim 1.
5. The compound of formula (I-1) according to claim 4, wherein R1Selected from halogen, C optionally substituted by halogen or deuterium1-3Alkyl or alkoxy; r2aAnd R2bIs as defined in claim 4, R3、X2Z is defined as in claim 1.
6. A compound of formula (I) according to claim 1, which is represented by formula (I-2):
Figure FDA0003034737910000031
wherein R is3Selected from C optionally substituted by halogen or cyano1-6Alkyl radical, C3-8Cycloalkyl, 4-to 8-membered heterocyclic group having 1 to 3 hetero atoms, alkoxy optionally substituted with halogen or amino optionally substituted with alkyl, R1、R2、X1Z, n are as defined in claim 1.
7. The compound of formula (I-2) according to claim 6, wherein R1Selected from halogen, C1-3Alkyl radical, C1-3Alkoxy, said alkyl or alkoxy being optionally substituted by one or more halogen or deuterium atoms, R3As defined in claim 6, R2、X1Z, n are as defined in claim 1.
8. A compound of formula (I) according to claim 1, which is of formula (I-3):
Figure FDA0003034737910000032
wherein R is2aAnd R2bIndependently selected from hydrogen, halogen, C optionally substituted by halogen1-3An alkyl group; r3Selected from C optionally substituted by halogen or cyano1-6Alkyl radical, C3-8Cycloalkyl, 4-to 8-membered heterocyclic group containing 1 to 3 hetero atoms, alkoxy optionally substituted with halogen or amino optionally substituted with alkyl; r1And Z is as defined in claim 1.
9. A compound of formula (I) according to claim 1, which is of formula (I-3):
Figure FDA0003034737910000033
wherein R is1Selected from methyl, chlorine, wherein methyl may be substituted by one or more fluorine or deuterium atoms;
R2aand R2bIndependently selected from hydrogen, fluorine atom, chlorine atom, methyl;
R3is selected from C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Fluoroalkyl, cyanomethyl, methylamino, or dimethylamino;
z is selected from:
a) selected from the group consisting of halogen, cyano, amino, sulfone, sulfonamide, alkyl substituted with one or more halogen atoms; or
b)
Figure FDA0003034737910000041
m is selected from 0, 1 or 2, R4、R5Independently selected from the group consisting of: hydrogen, alkyl, alkoxy and cycloalkyl, or R4And R5Together with the nitrogen to which they are attached form a 4-6 membered heterocyclyl, wherein: said heterocyclyl is optionally substituted with halogen, hydroxy or alkyl; and when m is 0, R4And R5A combination of (a) is other than hydrogen and methyl; or
c) A 4-6 membered heterocyclyl or heteroaryl group, which heterocyclyl or heteroaryl group is optionally substituted by oxo, halogen, hydroxy, carbonyl, alkyl or cyano, which alkyl group may be optionally substituted by halogen; or
d)
Figure FDA0003034737910000042
Or
e)
Figure FDA0003034737910000043
Wherein R is6Can be selected from C1-3Alkyl, C1-3 alkoxy, 5-6 membered aryl or heteroaryl, 3-8 membered cycloalkyl, 3-8 membered heterocyclyl, cyanoalkyl, cycloalkyloxy, amino optionally substituted with alkyl.
10. A compound of formula (I) according to claim 1, which is of formula (I-3)
Figure FDA0003034737910000044
Wherein R is1Selected from methyl, chlorine atoms, wherein methyl is optionally substituted with one or more fluorine or deuterium atoms;
R3is selected from C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Fluoroalkyl, cyanomethyl, methylamino, or dimethylamino;
R2aand R2bIndependently selected from hydrogen, deuterium, halogen, cyano, cyclopropyl, alkyl optionally substituted with halogen or deuterium;
z is selected from the following groups optionally substituted by one or more methyl, fluorine, chlorine, halomethyl, cyano groups:
Figure FDA0003034737910000051
11. a compound of formula (II), or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure FDA0003034737910000052
wherein Y is selected from
Figure FDA0003034737910000053
R7 is selected from alkyl optionally substituted with halogen or cyano, cycloalkyl, heterocyclyl, alkoxy optionally substituted with halogen, amino optionally substituted with alkyl, R8 is selected from alkyl, aryl optionally substituted with alkyl;
Z1selected from heterocyclic, heteroaryl or
Figure FDA0003034737910000054
Said heterocyclyl or heteroaryl optionally substituted with oxo, halo, hydroxy, carbonyl, alkyl or cyano, said alkyl optionally substituted with halo;
p is selected from 0, 1 or 2;
and the compound of formula (II) does not include Y being
Figure FDA0003034737910000055
And Z is1Is composed of
Figure FDA0003034737910000056
And p is 1.
12. A compound of formula (I) according to claim 1 or a compound of formula (II) according to claim 11, or a pharmaceutically acceptable salt thereof, or an isomer thereof, wherein Z is the following optionally substituted with one or more methyl, fluorine, chlorine, halomethyl, cyano groups:
Figure FDA0003034737910000057
Figure FDA0003034737910000061
13. the present disclosure also provides a compound of formula (III), or a pharmaceutically acceptable salt thereof, or an isomer thereof:
Figure FDA0003034737910000062
wherein R is1Selected from methyl, methoxy, methyl substituted with one or more fluorine atoms or deuterium atoms;
R2aand R2bIndependently selected from hydrogen, fluorine atom, chlorine atom, methyl;
z is selected from the following optionally substituted with one or more methyl, halomethyl, cyano groups:
Figure FDA0003034737910000063
l is a direct bond, an oxygen atom, -NH-
R3aIs selected from C1-6Alkyl radical, C3-6Cycloalkyl radical, C1-6alkyl-C3-6Cycloalkyl, 4-6 membered heterocyclyl, 5-6 membered heteroaryl, phenyl, 5-12 membered bridged cyclic group, 5-12 membered bridged spiro cyclic group, C as defined above1-6Alkyl radical, C3-6Cycloalkyl radical, C1-6alkyl-C3-6Cycloalkyl, 4-6 membered heterocyclyl, 5-6 membered heteroaryl, phenyl, 5-12 membered bridged cyclic group, 5-12 membered bridged spiro cyclic group may optionally be oxo, or optionally be substituted with one or more methyl, fluoromethyl groupsFluorine atom, cyano group, hydroxy group, C1-3Alkoxy radical, C1-3Fluoroalkoxy substitution.
14. A compound represented by the following formula, or a pharmaceutically acceptable salt thereof, or an isomer thereof,
Figure FDA0003034737910000071
Figure FDA0003034737910000081
Figure FDA0003034737910000091
Figure FDA0003034737910000101
15. a process for preparing a compound of formula (I) as claimed in claim 1 or a pharmaceutically acceptable salt thereof, or an isomer thereof, comprising the steps of:
Figure FDA0003034737910000102
carrying out cyclization reaction on the compound shown in the formula (I-a) and the compounds shown in the formula (I-b) and the formula (I-c) under the condition of a catalyst to obtain the compound shown in the formula (I);
the catalyst is selected from palladium/carbon, raney nickel, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride, 1' -bis (dibenzylphosphine) dichloropentairon palladium, tris (dibenzylideneacetone) dipalladium or 2-dicyclohexylphosphine-2 ',6' -dimethoxybiphenyl, [1,1' -bis (di-tert-butylphosphino) ferrocene ] dichloropalladium (II), cuprous iodide, cuprous bromide, cuprous chloride and copper trifluoromethanesulfonate;
R1、R2、R3z, n is as defined in claim 1.
16. A process for preparing a compound of formula (I) as claimed in claim 1 or a pharmaceutically acceptable salt thereof, or an isomer thereof, comprising the steps of:
Figure FDA0003034737910000111
carrying out cyclization reaction on the compound shown in the formula (I-d) and the compound shown in the formula (I-a) to obtain a compound shown in the formula (I-e); halogenating the compound shown in the formula (I-e) to obtain a compound shown in a formula (I-f); carrying out C-C coupling reaction on the compound of the formula (I-f) under an alkaline condition to obtain a compound shown in the formula (I);
the catalyst is selected from palladium/carbon, raney nickel, tetrakis-triphenylphosphine palladium, palladium dichloride, palladium acetate, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride, 1' -bis (dibenzylideophosphonium) dichlorodipentaerythritol palladium, tris (dibenzylideneacetone) dipalladium or 2-dicyclohexylphosphine-2 ',6' -dimethoxybiphenyl, [1,1' -bis (di-tert-butylphosphino) ferrocene ] dichloropalladium (II);
R1、R2、R3z, n is as defined in claim 1.
17. A process for preparing a compound of formula (I) as claimed in claim 1 or a pharmaceutically acceptable salt thereof, or an isomer thereof, comprising the steps of:
Figure FDA0003034737910000112
the compound shown as the formula (I-a) and the compound shown as the formula (I-g) are subjected to cyclization reaction to obtain the compound shown as the formula (I)
R1, R2, R3, Z, n are as defined in claim 1;
z' is selected from the group consisting of halogen, sulfonyl and sulfinyl.
18. A pharmaceutical composition comprising a compound of claims 1-14, or a pharmaceutically acceptable salt thereof, or an isomer thereof, and at least one pharmaceutically acceptable carrier, diluent, or excipient.
19. Use of a compound according to claims 1-14, or a pharmaceutically acceptable salt, or isomer thereof, or a pharmaceutical composition of claim 18, for the manufacture of a medicament for the treatment of a disease associated with P2X3 activity.
20. The use according to claim 19, wherein the disease is selected from pain, urinary tract diseases and cough.
CN202110463162.9A 2020-04-24 2021-04-23 Novel imidazopyridine compound, preparation method and medical application thereof Active CN113549068B (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202010333060 2020-04-24
CN2020103330600 2020-04-24
CN202010783728 2020-08-06
CN2020107837281 2020-08-06

Publications (2)

Publication Number Publication Date
CN113549068A true CN113549068A (en) 2021-10-26
CN113549068B CN113549068B (en) 2022-12-02

Family

ID=78130146

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110463162.9A Active CN113549068B (en) 2020-04-24 2021-04-23 Novel imidazopyridine compound, preparation method and medical application thereof

Country Status (1)

Country Link
CN (1) CN113549068B (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114085220A (en) * 2020-06-22 2022-02-25 上海海雁医药科技有限公司 Substituted morpholine-4-carboxylic ester derivatives, their compositions and their medical use
WO2022156784A1 (en) * 2021-01-22 2022-07-28 武汉人福创新药物研发中心有限公司 Preparation method for heterocyclic compound, and intermediate of heterocyclic compound
WO2022156783A1 (en) * 2021-01-22 2022-07-28 武汉人福创新药物研发中心有限公司 Preparation method for imidazopyridine compound and intermediate thereof
WO2022161462A1 (en) * 2021-01-29 2022-08-04 上海海雁医药科技有限公司 Morpholine derivative, and pharmaceutical composition and use thereof
CN115043836A (en) * 2021-08-20 2022-09-13 苏州璞正医药有限公司 P2X3 receptor selective regulator of imidazopyridine derivative and pharmaceutical application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105246888A (en) * 2013-01-31 2016-01-13 尼奥迈德研究所 Imidazopyridine compounds and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105246888A (en) * 2013-01-31 2016-01-13 尼奥迈德研究所 Imidazopyridine compounds and uses thereof

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114085220A (en) * 2020-06-22 2022-02-25 上海海雁医药科技有限公司 Substituted morpholine-4-carboxylic ester derivatives, their compositions and their medical use
CN114085220B (en) * 2020-06-22 2023-06-16 上海海雁医药科技有限公司 Substituted morpholine-4-carboxylic acid ester derivatives, compositions and pharmaceutical uses thereof
WO2022156784A1 (en) * 2021-01-22 2022-07-28 武汉人福创新药物研发中心有限公司 Preparation method for heterocyclic compound, and intermediate of heterocyclic compound
WO2022156783A1 (en) * 2021-01-22 2022-07-28 武汉人福创新药物研发中心有限公司 Preparation method for imidazopyridine compound and intermediate thereof
WO2022161462A1 (en) * 2021-01-29 2022-08-04 上海海雁医药科技有限公司 Morpholine derivative, and pharmaceutical composition and use thereof
CN115043836A (en) * 2021-08-20 2022-09-13 苏州璞正医药有限公司 P2X3 receptor selective regulator of imidazopyridine derivative and pharmaceutical application thereof

Also Published As

Publication number Publication date
CN113549068B (en) 2022-12-02

Similar Documents

Publication Publication Date Title
CN113549068B (en) Novel imidazopyridine compound, preparation method and medical application thereof
JP6742452B2 (en) Novel tetrahydropyridopyrimidines for treatment and prevention of HBV infection
KR101864561B1 (en) JAK inhibitors
JP2023538408A (en) Compounds as GLP-1R agonists
AU2012323085B2 (en) PDE9i with imidazo pyrazinone backbone
JP5780954B2 (en) Azetidines and cyclobutanes as histamine H3 receptor antagonists
JP7219902B2 (en) Bruton's Tyrosine Kinase Inhibitor
CN105228995B (en) Substituted piperidine compounds and their purposes as orexin receptor regulator
KR20180103683A (en) CCR2 modulator
TW201350476A (en) Compounds and compositions for modulating EGFR activity
KR20130057971A (en) New enzyme inhibitor compounds
CN107474050A (en) 6,7 dihydro-pyrazolos [1,5 a] pyrazine 4 (5H) assimilation compound
JP7168149B2 (en) Pyrazin-2(1H)-one compounds as FGFR inhibitors
WO2022007824A1 (en) Compound having btk kinase degrading activity, and preparation method and pharmaceutical use therefor
CN110494431B (en) Azacyclo derivative, preparation method and medical application thereof
CN109415355A (en) Benzdioxan derivative and its medicinal usage
CN115835867A (en) Benzimidazole derivative, preparation method and medical application thereof
JP2023554506A (en) FGFR inhibitor compounds and their uses
JP2023536891A (en) JAK inhibitor compounds for treating severe pneumonia
KR20230038530A (en) Compounds acting as BTK inhibitors, methods for their preparation and uses thereof
JP2022543690A (en) Bridged Heterocyclyl-Substituted Pyrimidine Compounds, Methods for Their Preparation, and Pharmaceutical Uses Therefor
WO2017018475A1 (en) Condensed pyrazole derivative having new linker site and medicinal use of same
WO2022028154A1 (en) N-carboxamidopyrazoline derivative serving as p2x3 receptor antagonist and applications
WO2019013562A1 (en) Novel 1h-pyrazolopyridine derivative and pharmaceutical composition containing same
JP2020522542A (en) Carboxylic acid derivatives as AT2R receptor antagonists

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant