CN112409331B - Heterocyclic derivative inhibitor, preparation method and application thereof - Google Patents

Heterocyclic derivative inhibitor, preparation method and application thereof Download PDF

Info

Publication number
CN112409331B
CN112409331B CN202010852140.7A CN202010852140A CN112409331B CN 112409331 B CN112409331 B CN 112409331B CN 202010852140 A CN202010852140 A CN 202010852140A CN 112409331 B CN112409331 B CN 112409331B
Authority
CN
China
Prior art keywords
mmol
esi
added
preparation
give
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.)
Active
Application number
CN202010852140.7A
Other languages
Chinese (zh)
Other versions
CN112409331A (en
Inventor
肖华玲
蔡加强
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.)
Jiangsu Hansoh Pharmaceutical Group Co Ltd
Shanghai Hansoh Biomedical Co Ltd
Original Assignee
Jiangsu Hansoh Pharmaceutical Group Co Ltd
Shanghai Hansoh Biomedical 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 Jiangsu Hansoh Pharmaceutical Group Co Ltd, Shanghai Hansoh Biomedical Co Ltd filed Critical Jiangsu Hansoh Pharmaceutical Group Co Ltd
Publication of CN112409331A publication Critical patent/CN112409331A/en
Application granted granted Critical
Publication of CN112409331B publication Critical patent/CN112409331B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D471/12Heterocyclic 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 three hetero rings
    • C07D471/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/18Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 one oxygen and one nitrogen atom, e.g. guanine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains three hetero rings
    • C07D513/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention relates to a heterocyclic derivative inhibitor, a preparation method and application thereof. In particular, the invention relates to compounds of general formula (I), to processes for their preparation, to pharmaceutical compositions containing them and to their use as P2X3 inhibitors in the treatment of P2X3 receptor dysfunction disorders, in particular in the treatment of neurogenic disorders.

Description

Heterocyclic derivative inhibitor, preparation method and application thereof
Technical Field
The invention belongs to the field of medicine synthesis, and particularly relates to a heterocyclic derivative inhibitor, and a preparation method and application thereof.
Background
P2X receptors (P2X receptors), or P2X purinerceptors (P2X purinerceptors), are a family of cation-permeable ATP ligand-gated ion channels that bind extracellular ATP. The P2X receptor has seven subunits, present as homotrimers or heterotrimers, expressed predominantly in nerve endings (presynaptic and postsynaptic) of the nervous system, regulating synaptic transmission. The P2X3 receptor is one of the P2X family members, is a key sensory receptor that senses upper respiratory tract stimulation and triggers cough reflex, is believed to play a key role in sensitization (sensory) of specific sensory nerves, is involved in pain and cough, and is involved in perception of bone cancer pain, and blocking P2X3 can inhibit signal stimulation of cough.
Cough is defensive nerve reflex of organism, is favorable for clearing away secretion of respiratory tract and harmful factors, but frequent and severe cough can cause serious influence on work, life and social activities of patients and the like. Cough is classified as acute, subacute, chronic cough. Chronic cough (Chronic cough) is a cough with a period of >8 weeks, and chest imaging examined lung for no apparent lesions with cough as the primary or sole symptom. Chronic cough has been recognized as a consequence of various diseases such as asthma/eosinophilic bronchitis, rhinitis and gastroesophageal reflux disease. Recent evidence, however, suggests that chronic cough is a clinical symptom with unique neuroallergies that are inherent pathophysiological in nature. Chronic cough or idiopathic cough of unknown cause is mainly manifested by chronic stimulating dry cough, which is more sensitive to external stimulus, and cough high sensitivity is commonly existed, which is the physiological and pathological mechanism. Cough-related afferent nerve abnormalities may be the cause of intractable or unexplained chronic cough. Chronic cough can cause complications of multiple systems such as cardiovascular, digestive, neurological, urinary, musculoskeletal, etc., such as urinary incontinence, syncope, insomnia, anxiety, etc.
Based on the pathophysiological characteristics of the cough hypersensitive syndrome, treatment should be aimed at reducing cough sensitivity. Currently, there are limited treatment options, including drug therapies and non-drug therapies. Clinical research results show that the medicine gabapentin of the neuromodulation factor type is effective in treatment, and other medicines such as amitriptyline, baclofen, carbamazepine, pregabalin and the like can also be selected. The severe cough can be properly treated by the anti-cough drugs, which are mainly divided into central anti-cough drugs and peripheral anti-cough drugs. The central antitussive is classified into a dependent antitussive (morphine alkaloid and its derivative) and a non-dependent antitussive (artificially synthesized dextromethorphan and pentofyllin), the former having side effects such as addiction and narcosis, and the latter being widely used in clinical applications. Peripheral antitussives, also known as peripheral antitussives, act as antitussives by inhibiting a link in the reflex arc of cough, including local anesthetics (narcotine, benzonatate) and mucoprotective agents (benproperine and moguisteine).
At present, there is no approved P2X3 receptor antagonist small molecule drug on the market. The existing P2X3 receptor antagonist drugs in clinical stages are MK-7264 of Merck (Merck & Co) which is used for treating diseases such as pain, pulmonary fibrosis and the like, has low selectivity on P2X3/P2X2/3, good safety and side effects such as taste loss and the like, and chronic cough of the current indication has been studied in clinical stage III. Bei Lesi Health (Bacillus Health) BLU5937 has high selectivity, and no side effects such as taste sensation in phase I clinical trial; the main study results of the RELIEF trial at stage 2 of BLU-5937 in refractory chronic cough patients were published by health at Bei Lesi, 7/2020: in phase II clinical studies, the RELIEF test did not reach statistical significance of the primary endpoint of cough frequency reduction after placebo adjustment at any dose. But are well tolerated, taste effects including taste changes and partial taste loss are rare at all dose levels, and are mostly mild in nature, with no patient reporting complete loss of taste. In addition, bayer (Bayer) BAY-1817080 and Shionogi S-600918, the indication chronic cough is currently in clinical stage I/II. Therefore, there is an urgent need to develop P2X3 receptor inhibitor drugs with good safety, non-addictive narcotics and high selectivity for treating chronic cough and other diseases so as to meet the huge market demand.
The compounds of the present examples have better liver microsomal stability and less taste side effects than Bei Lesi healthy BLU 5937.
Disclosure of Invention
The invention aims to provide a compound shown in a general formula (I), a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the compound shown in the general formula (I) has the following structure:
wherein:
L 1 and L 2 Each independently selected from the group consisting of bond, - (CH) 2 ) n1 -、-(CH 2 ) n1 NR aa (CR bb R cc ) n2 -、-(CH 2 ) n1 C(O)NR aa -、-(CR aa R bb ) n1 O(CH 2 ) n2 -、-(CR aa R bb ) n1 (CH 2 ) n2 O-、-(CH 2 ) n1 NR aa C(O)-、-(CH 2 ) n1 (CR aa R bb ) n2 -、-(CH 2 ) n1 O(CR aa R bb ) n2 -、-(CR aa R bb ) n1 S(CH 2 ) n2 -、-(CH 2 ) n1 S(CR aa R bb ) n2 -、-(CR aa R bb ) n1 (CH 2 ) n2 NR cc -、-(CH 2 ) n1 C(O)(CR aa R bb ) n2 -、-(CH 2 ) n1 P(O)R aa -、-(CH 2 ) n1 S(O) n2 -、-(CH 2 ) n1 S(O) n2 NR aa -or- (CH) 2 ) n1 NR aa S(O) n2 -;
Ring a is selected from cycloalkyl, heterocyclyl, aryl or heteroaryl;
ring B is selected from cycloalkyl, heterocyclyl, aryl or heteroaryl;
ring C is selected from cycloalkyl, heterocyclyl, aryl or heteroaryl;
R a selected from the group consisting of hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, oxo, thio, alkyl, deuteroalkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, heterocyclylalkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, - (CH) 2 ) n1 R aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 C(O)OR aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 SR aa 、-(CH 2 ) n1 NR aa C(O)R bb 、-(CH 2 ) n1 NR aa C(O)OR bb 、-(CH 2 ) n1 NR aa C(O)NR bb R cc 、-(CH 2 ) n1 NR aa R bb 、-NR aa (CH 2 ) n1 R bb 、-(CH 2 ) n1 C(O)NR aa R bb 、-(CH 2 ) n1 C(O)R aa 、-OC(R aa R bb ) n1 (CH 2 ) n2 R cc 、-C(R aa R bb ) n1 (CH 2 ) n2 OR cc 、-(CH 2 ) n1 S(O) n2 R aa 、-(CH 2 ) n1 NR aa S(O) n2 R bb 、-CH=CH(CH 2 ) n1 R aa 、-CH=CH(CH 2 ) n1 NR aa R bb 、-CH=CH(CH 2 ) n1 NR aa C(O)R bb Or-ch=ch (CH 2 ) n1 NR aa C(O)NR bb R cc Said amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, heterocyclylalkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl groups, optionally further substituted;
R b selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, oxo, thio, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy A group, haloalkoxy, alkenyl, alkynyl, heterocyclylalkyl, cycloalkyl, heterocyclyl, aryl, aryloxy, heteroaryl, heteroaryloxy, - (CH) 2 ) n1 R aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 C(O)OR aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 SR aa 、-(CH 2 ) n1 NR aa C(O)R bb 、-(CH 2 ) n1 NR aa C(O)OR bb 、-(CH 2 ) n1 NR aa C(O)NR bb R cc 、-(CH 2 ) n1 NR aa R bb 、-NR aa (CH 2 ) n1 R bb 、-(CH 2 ) n1 C(O)NR aa R bb 、-(CH 2 ) n1 C(O)R aa 、-OC(R aa R bb ) n1 (CH 2 ) n2 R cc 、-(CH 2 ) n1 S(O) n2 R aa 、-(CH 2 ) n1 NR aa S(O) n2 R bb 、-CH=CH(CH 2 ) n1 R aa 、-CH=CH(CH 2 ) n1 NR aa R bb 、-CH=CH(CH 2 ) n1 NR aa C(O)R bb Or-ch=ch (CH 2 ) n1 NR aa C(O)NR bb R cc Said amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, heterocyclylalkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl groups, optionally further substituted;
R c selected from the group consisting of hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, oxo, thio, alkyl, deuteroalkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, heterocyclylalkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, - (CH) 2 ) n1 R aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 C(O)OR aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 SR aa 、-(CH 2 ) n1 NR aa C(O)R bb 、-(CH 2 ) n1 NR aa C(O)OR bb 、-(CH 2 ) n1 NR aa C(O)NR bb R cc 、-(CH 2 ) n1 NR aa R bb 、-NR aa (CH 2 ) n1 R bb 、-(CH 2 ) n1 C(O)NR aa R bb 、-(CH 2 ) n1 C(O)R aa 、-OC(R aa R bb ) n1 (CH 2 ) n2 R cc 、-(CH 2 ) n1 S(O) n2 R aa 、-(CH 2 ) n1 NR aa S(O) n2 R bb -CH=CH(CH 2 ) n1 R aa 、-CH=CH(CH 2 ) n1 NR aa R bb 、-CH=CH(CH 2 ) n1 NR aa C(O)R bb Or-ch=ch (CH 2 ) n1 NR aa C(O)NR bb R cc Said amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, heterocyclylalkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl groups, optionally further substituted;
R aa 、R bb and R is cc Each independently selected from the group consisting of hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, heterocyclylalkyl, cycloalkyl, heterocyclyl, aryl, aryloxy, heteroaryl, or heteroaryloxy, said amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, heterocyclylalkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, optionally further substituted;
Alternatively, R aa 、R bb And R is cc Any two of which are linked to form cycloalkyl, heterocyclyl, aryl or heteroaryl, said cycloalkyl, heterocyclyl, aryl and heteroaryl optionallyMay be further substituted;
x is an integer of 0 to 6;
y is an integer of 0 to 6;
z is an integer of 0 to 6;
n1 is 0, 1, 2 or 3; and is also provided with
n2 is 0, 1, 2 or 3.
In a preferred embodiment of the invention, L 1 Selected from bond, - (CH) 2 ) n1 -、-(CH 2 ) n1 C(O)NR aa -、-(CH 2 ) n1 (CR aa R bb ) n2 -、-(CR aa R bb ) n1 (CH 2 ) n2 O-、-(CH 2 ) n1 O(CR aa R bb ) n2 -、-(CR aa R bb ) n1 S(CH 2 ) n2 -、-(CH 2 ) n1 S(CR aa R bb ) n2 -、-(CR aa R bb ) n1 (CH 2 ) n2 NR cc -、-(CH 2 ) n1 NR aa (CR bb R cc ) n2 -、-(CH 2 ) n1 C(O)(CR aa R bb ) n2 -、-(CH 2 ) n1 NR aa C(O)-、-(CH 2 ) n1 S(O) n2 -、-(CH 2 ) n1 S(O) n2 NR aa -or- (CH) 2 ) n1 NR aa S(O) n2 -。
In a further preferred embodiment of the invention, L 1 Selected from bond, - (CH) 2 ) n1 -、-(CH 2 ) n1 O-、-(CH 2 ) n1 S-、-(CH 2 ) n1 NR aa -、-(CH 2 ) n1 C(O)NR aa -or- (CH) 2 ) n1 NR aa C(O)-。
In a further preferred embodiment of the invention, L 1 Selected from bond, -CH 2 -, -O-, -S-; -NH- -C (O) NH-or-NHC (O) -.
The invention also provides a preferable scheme, L 2 Selected from the group consisting of a bond- (-) aCH 2 ) n1 -、-(CH 2 ) n1 C(O)NR aa -、-(CH 2 ) n1 (CR aa R bb ) n2 -、-(CR aa R bb ) n1 (CH 2 ) n2 O-、-(CH 2 ) n1 O(CR aa R bb ) n2 -、-(CR aa R bb ) n1 S(CH 2 ) n2 -、-(CH 2 ) n1 S(CR aa R bb ) n2 -、-(CR aa R bb ) n1 (CH 2 ) n2 NR cc -、-(CH 2 ) n1 NR aa (CR bb R cc ) n2 -、-(CH 2 ) n1 C(O)(CR aa R bb ) n2 -、-(CH 2 ) n1 NR aa C(O)-、-(CH 2 ) n1 S(O) n2 -、-(CH 2 ) n1 S(O) n2 NR aa -or- (CH) 2 ) n1 NR aa S(O) n2 -。
In a further preferred embodiment of the invention, L 2 Selected from bond, - (CH) 2 ) n1 -、-(CH 2 ) n1 (CR aa R bb ) n2 -、-(CH 2 ) n1 O-、-(CH 2 ) n1 S-、-(CH 2 ) n1 NR aa -、-(CH 2 ) n1 C(O)(CR aa R bb ) n2 -、-(CH 2 ) n1 C(O)NR aa -or- (CH) 2 ) n1 NR aa C(O)-。
In a further preferred embodiment of the invention, L 2 Selected from bond, -CH 2 -、-O-、-S-、-NH-、-CH 2 C(O)-、-CH 2 C (O) NH-or
In a preferred embodiment of the invention, ring B is selected from C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl;
in a further preferred embodiment of the invention, ring B is selected from C 6-14 Aryl or 5-14 membered heteroaryl, more preferably phenyl, benzo 5-8 membered nitrogen containing heterocyclyl, benzo 5-8 membered nitrogen containing heteroaryl, further selected from the group consisting of:
In a preferred embodiment of the invention, ring C is selected from C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl;
in a further preferred embodiment of the invention, ring C is selected from C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl, more preferably cyclobutyl, piperidinyl, phenyl, pyridinyl or morpholinyl, further selected from the group consisting of:
in a preferred embodiment of the invention, R a Selected from hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, oxo, thioxo, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, halo C 1-6 Alkoxy, C 1-6 Hydroxyalkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl, 5-14 membered heteroaryl, - (CH) 2 ) n1 R aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 C(O)OR aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 SR aa 、-(CH 2 ) n1 NR aa C(O)R bb 、-C(R aa R bb ) n1 (CH 2 ) n2 OR cc 、-(CH 2 ) n1 NR aa C(O)OR bb 、-(CH 2 ) n1 NR aa C(O)NR bb R cc 、-(CH 2 ) n1 NR aa R bb 、-NR aa (CH 2 ) n1 R bb 、-(CH 2 ) n1 C(O)NR aa R bb 、-(CH 2 ) n1 C(O)R aa 、-OC(R aa R bb ) n1 (CH 2 ) n2 R cc 、-(CH 2 ) n1 S(O) n2 R aa Or- (CH) 2 ) n1 NR aa S(O) n2 R bb The amino group, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally substituted with hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 One or more substituents in aryl and 5-14 membered heteroaryl;
In a further preferred embodiment of the invention, R a Selected from hydrogen, oxo, C 1-6 Alkyl, C 2-6 Alkenyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, 5-10 membered heteroaryl, - (CH) 2 ) n1 NR aa R bb 、-(CH 2 ) n1 SR aa 、-(CH 2 ) n1 C(O)NR aa R bb 、-(CH 2 ) n1 NR aa C(O)R bb or-C (R) aa R bb ) n1 (CH 2 ) n2 OR cc The C is 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, 3-8 membered heterocyclyl and 5-10 membered heteroaryl, optionally substituted with hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl group,C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl and 5-10 membered heteroaryl;
in a further preferred embodiment of the invention, R a Selected from hydrogen, halogen, oxo, C 1-6 Alkyl, C 2-6 Alkenyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, 5-10 membered heteroaryl, - (CH) 2 ) n1 NR aa R bb 、-(CH 2 ) n1 SR aa 、-(CH 2 ) n1 C(O)NR aa R bb 、-(CH 2 ) n1 NR aa C(O)R bb or-C (R) aa R bb ) n1 (CH 2 ) n2 OR cc The C is 1-6 Alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, 3-8 membered heterocyclyl and 5-10 membered heteroaryl, optionally substituted with hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl and 5-10 membered heteroaryl;
in a further preferred embodiment of the invention, R a Selected from hydrogen, oxo, methyl, ethyl, isopropyl, difluoro isobutyl, methyl dioxazolyl, vinyl, cyclopropyl, cyclobutyl, -NR aa R bb 、-SR aa 、-C(O)NR aa R bb 、-NR aa C(O)R bb or-C (R) aa R bb ) n1 (CH 2 ) n2 OR cc
In a preferred embodiment of the invention, R b Selected from hydrogen, deuterium, halogen, amino, hydroxyA group, cyano group, nitro group, oxo group, thio group, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl, C 6-14 Aryloxy, 5-14 membered heteroaryl, 5-14 membered heteroaryloxy, - (CH) 2 ) n1 R aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 C(O)OR aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 SR aa 、-(CH 2 ) n1 NR aa C(O)R bb 、-(CH 2 ) n1 NR aa C(O)OR bb 、-(CH 2 ) n1 NR aa C(O)NR bb R cc 、-(CH 2 ) n1 NR aa R bb 、-NR aa (CH 2 ) n1 R bb 、-(CH 2 ) n1 C(O)NR aa R bb 、-(CH 2 ) n1 C(O)R aa 、-OC(R aa R bb ) n1 (CH 2 ) n2 R cc 、-(CH 2 ) n1 S(O) n2 R aa Or- (CH) 2 ) n1 NR aa S(O) n2 R bb The amino group, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl, C 6-14 Aryloxy, 5-14 membered heteroaryl and 5-14 membered heteroaryloxy, optionally interrupted by hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-12 One or more substituents in aryl and 5-12 membered heteroaryl;
in a further preferred embodiment of the invention, R b Selected from hydrogen, halogen, oxo, C 1-3 Alkyl, -OR aa or-C (O) NR aa R bb
In a further preferred embodiment of the invention, R b Selected from hydrogen, halogen, oxo, C 1-6 Alkyl, - (CH) 2 ) n1 OR aa 、-(CH 2 ) n1 C(O)NR aa R bb 、-(CH 2 ) n1 NR aa C(O)R bb 、-(CH 2 ) n1 NR aa C(O)OR bb 、-(CH 2 ) n1 S(O) n2 NR aa R bb 、-(CH 2 ) n1 NR aa S(O) n2 R bb Or- (CH) 2 ) n1 NR aa S(O) n2 NR bb R cc The method comprises the steps of carrying out a first treatment on the surface of the Preferably hydrogen, halogen, oxo, C 1-3 Alkyl, -OR aa 、-C(O)NR aa R bb 、-NR aa C(O)R bb 、-NR aa C(O)OR bb 、-S(O) 2 NR aa R bb 、-NR aa S(O) 2 R bb or-NR aa S(O) 2 NR bb R cc
In a further preferred embodiment of the invention, R b Selected from hydrogen, halogen, oxo, C 1-6 Alkyl, - (CH) 2 ) n1 OR aa 、-(CH 2 ) n1 C(O)NR aa R bb 、-(CH 2 ) n1 NR aa C(O)R bb 、-(CH 2 ) n1 NR aa C(O)OR bb 、-(CH 2 ) n1 S(O) n2 R aa 、-(CH 2 ) n1 S(O) n2 NR aa R bb 、-(CH 2 ) n1 NR aa S(O) n2 R bb 、-(CH 2 ) n1 S(O)(=NR aa )R bb 、-(CH 2 ) n1 NR aa S(O)(=NR bb )R cc Or- (CH) 2 ) n1 NR aa S(O) n2 NR bb R cc The method comprises the steps of carrying out a first treatment on the surface of the Preferably hydrogen, halogen, oxo, C 1-3 Alkyl, -OR aa 、-C(O)NR aa R bb 、-NR aa C(O)R bb 、-NR aa C(O)OR bb 、-S(O) 2 R aa 、-S(O) 2 NR aa R bb 、-NR aa S(O) 2 R bb 、-S(O)(=NR aa )R bb 、-NR aa S(O)(=NR bb )R cc or-NR aa S(O) 2 NR bb R cc
In a further preferred embodiment of the invention, R b Selected from hydrogen, fluorine, chlorine, bromine, oxo, methyl, ethyl, isopropyl, -C (O) NHCH 3 、-C(O)NHC(CH 3 )CH 2 OCH 3 、-NHC(O)OCH 3 、-NHC(O)CHF 2 、-NHS(O) 2 CH 3 、-NHS(O) 2 N(CH 3 ) 2 、-S(O) 2 CH 3 、-S(O) 2 NHCH 3 、-S(O) 2 N(CH 3 ) 2 、-S(O) 2 NHCH(CH 3 ) 2 、-S(O)(=NH)CH 3 、-NHS(O)(=NH)CH 3
In a preferred embodiment of the invention, R c Selected from hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl, 5-14 membered heterogeniesAryl, - (CH) 2 ) n1 C(O)R aa 、-(CH 2 ) n1 C(O)OR aa Or- (CH) 2 ) n1 C(O)NR aa R bb The amino group, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally substituted with hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-12 One or more substituents in aryl and 5-12 membered heteroaryl;
in a further preferred embodiment of the invention, R c Selected from hydrogen, halogen, C 1-3 Alkyl, halophenyl, -C (O) R aa OR-C (O) OR aa
In a further preferred embodiment of the invention, R c Selected from hydrogen, halogen, C 1-6 Alkyl, halophenyl, - (CH) 2 ) n1 C(O)R aa 、-(CH 2 ) n1 C(O)OR aa 、-(CH 2 ) n1 S(O) n2 R aa Or- (CH) 2 ) n1 S(O) n2 NR aa R bb The method comprises the steps of carrying out a first treatment on the surface of the Preferably hydrogen, halogen, C 1-6 Alkyl, halophenyl, -C (O) R aa 、-C(O)OR aa 、-S(O) 2 R aa or-S (O) 2 NR aa R bb
In a further preferred embodiment of the invention, R c Selected from hydrogen, fluorine, chlorine, bromine, C 1-6 Alkyl, methyl, ethyl, isopropyl, chlorophenyl, -C (O) CH 3 、-C(O)CHF 2 、-C(O)CH 2 CH 3 、-C(O)OCH 3 、-C(O)OC(CH 3 ) 3 、-S(O) 2 CH 3 、-S(O) 2 CH 2 CH 3 、-S(O) 2 N(CH 3 ) 2
In a preferred embodiment of the invention, R aa 、R bb And R is cc Each independently selected from hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl, said amino, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally substituted with hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl, 5-14 membered heteroaryl, said amino, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 One or more substituents in aryl and 5-14 membered heteroaryl;
alternatively, R aa 、R bb And R is cc Any two links forming C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-12 membered heteroaryl, said C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally substituted with hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 One or more substituents in aryl and 5-14 membered heteroaryl;
in a further preferred embodiment of the present invention, there is provided a compound of formula (II), a stereoisomer thereof or a pharmaceutically acceptable salt thereof, having the specific structure:
in a further preferred embodiment of the present invention, there is provided a compound of formula (III), a stereoisomer thereof or a pharmaceutically acceptable salt thereof, having the specific structure:
wherein:
M 1 selected from N or CR 9 The method comprises the steps of carrying out a first treatment on the surface of the Preferably N, CH or CF;
R 9 、R 10 and R is 13 Each independently selected from hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl groups、C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl, said amino, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally substituted with hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 One or more substituents in aryl and 5-14 membered heteroaryl;
preferably hydrogen, fluorine, chlorine, bromine or iodine;
R 11 and R is 12 Each independently selected from hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl, 5-14 membered heteroaryl, - (CH) 2 ) n1 R aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 C(O)OR aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 SR aa 、-(CH 2 ) n1 NR aa C(O)R bb 、-C(R aa R bb ) n1 (CH 2 ) n2 OR cc 、-(CH 2 ) n1 NR aa C(O)OR bb 、-(CH 2 ) n1 NR aa C(O)NR bb R cc 、-(CH 2 ) n1 NR aa R bb 、-NR aa (CH 2 ) n1 R bb 、-(CH 2 ) n1 C(O)NR aa R bb 、-(CH 2 ) n1 C(O)R aa 、-OC(R aa R bb ) n1 (CH 2 ) n2 R cc 、-(CH 2 ) n1 S(O) n2 R aa Or- (CH) 2 ) n1 NR aa S(O) n2 R bb The amino group, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally substituted with hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 One or more substituents in aryl and 5-14 membered heteroaryl; preferably hydrogen, methyl, ethyl, isopropyl, vinyl, methoxy, methylthio, cyclopropyl, cyclobutyl or-NHCH 3
Or R is 11 And R is 12 To carbon atoms to which it is attached to form C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl, said C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally further substituted with hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl groups、C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 One or more substituents in aryl and 5-14 membered heteroaryl;
R 11 and R is 12 A ring linked to the carbon atom to which it is attached, preferably a 5-6 membered nitrogen containing heterocyclyl or heteroaryl, a 5-6 membered oxygen containing heterocyclyl or heteroaryl, or a 5-6 membered sulfur containing heterocyclyl or heteroaryl;
further preferred are the following groups:
R 14 selected from hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl, 5-14 membered heteroaryl, - (CH) 2 ) n1 R aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 C(O)OR aa 、-(CH 2 ) n1 OR aa 、-(CH 2 ) n1 SR aa 、-(CH 2 ) n1 NR aa C(O)R bb 、-C(R aa R bb ) n1 (CH 2 ) n2 OR cc 、-(CH 2 ) n1 NR aa C(O)OR bb 、-(CH 2 ) n1 NR aa C(O)NR bb R cc 、-(CH 2 ) n1 NR aa R bb 、-NR aa (CH 2 ) n1 R bb 、-(CH 2 ) n1 C(O)NR aa R bb 、-(CH 2 ) n1 C(O)R aa 、-OC(R aa R bb ) n1 (CH 2 ) n2 R cc 、-(CH 2 ) n1 S(O) n2 R aa Or- (CH) 2 ) n1 NR aa S(O) n2 R bb The amino group, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally substituted with hydrogen, deuterium, halogen, amino, hydroxy, cyano, nitro, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl-cyano-substituted C 1-6 Alkyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 One or more substituents in aryl and 5-14 membered heteroaryl; preferably hydrogen, halogen, methyl, ethyl, isopropyl, -C (O) OCH 3 、-C(O)CH 2 CH 3 、-C(O)OCH 2 CH 3
In a further preferred embodiment of the invention, R 14 Selected from hydrogen, halogen, C 1-6 Alkyl, halophenyl, - (CH) 2 ) n1 C(O)R aa 、-(CH 2 ) n1 C(O)OR aa 、-(CH 2 ) n1 S(O) n2 R aa Or- (CH) 2 ) n1 S(O) n2 NR aa R bb The method comprises the steps of carrying out a first treatment on the surface of the Preferably hydrogen, halogen, C 1-6 Alkyl, halophenyl, -C (O) R aa 、-C(O)OR aa 、-S(O) 2 R aa or-S (O) 2 NR aa R bb The method comprises the steps of carrying out a first treatment on the surface of the More preferably hydrogen, fluorine, chlorine, bromine, methyl, ethyl, isopropyl, -C (O) OCH 3 、-C(O)CH 2 CH 3 、-C(O)CHF 2 、-C(O)OCH 2 CH 3 、-S(O) 2 CH 3 、-S(O) 2 CH 2 CH 3 、-S(O) 2 N(CH 3 ) 2
In a further preferred embodiment of the present invention, there is provided a compound of formula (IV), a stereoisomer thereof or a pharmaceutically acceptable salt thereof, having the specific structure:
wherein:
R 14 is-C (O) R aa
R aa Selected from C 1-6 Alkyl, C 1-6 Haloalkyl or C 1-6 An alkoxy group;
preferably C 1-3 Alkyl, C 1-3 Haloalkyl or C 1-3 An alkoxy group;
more preferably methyl, ethyl, difluoromethyl, methoxy or ethoxy.
The invention also relates to a method for preparing the compound shown in the general formula (IV) or the stereoisomer and the pharmaceutically acceptable salt thereof, which comprises the following steps,
the compound shown in the general formula (IV-1) or salt thereof and the compound shown in the general formula (IV-2) undergo condensation reaction to obtain the compound shown in the general formula (IV) or stereoisomer and pharmaceutically acceptable salt thereof;
Wherein,
x is selected from hydroxyl, chlorine or bromine;
R 14 is-C (O) R aa
R aa Selected from C 1-6 Alkyl, C 1-6 Haloalkyl or C 1-6 An alkoxy group;
preferably C 1-3 Alkyl, C 1-3 Haloalkyl or C 1-3 An alkoxy group;
more preferably methyl, ethyl, difluoromethyl, methoxy or ethoxy.
The invention further relates to an intermediate of the compound shown in the preparation (IV) or a stereoisomer and a pharmaceutically acceptable salt thereof, wherein the compound shown in the formula (IV-1) or the salt thereof has the following specific structure:
the invention further relates to a method for preparing the compound shown in the general formula (IV-1) or the salt thereof, which specifically comprises the following steps,
deprotection of the general formula (IV-3) to give a compound of the general formula (IV-1) or a salt thereof;
wherein:
Pg 1 is an amino protecting group selected from allyloxycarbonyl, trifluoroacetyl, 2, 4-dimethoxybenzyl, nitrobenzenesulfonyl, trityl, benzyloxycarbonyl, p-toluenesulfonyl, formate, acetyl, benzyloxycarbonyl, t-butoxycarbonyl, benzyl or p-methoxyphenyl; t-butoxycarbonyl is preferred.
The invention also relates to a method for preparing the compound shown in the general formula (IV) or the stereoisomer and the pharmaceutically acceptable salt thereof, which comprises the following steps,
the invention further relates to a pharmaceutical composition comprising a therapeutically effective dose of any of the compounds of formula (I), stereoisomers or pharmaceutically acceptable salts thereof, as shown in any of the formulae shown herein, together with one or more pharmaceutically acceptable carriers, diluents or excipients.
The invention further relates to application of any compound shown in the general formula (I), stereoisomer or pharmaceutically acceptable salt thereof, or the pharmaceutical composition in preparation of P2X3 receptor inhibitor drugs.
The invention further relates to application of a compound shown in the general formula (I), a stereoisomer or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof in preparing medicines for treating neurogenic diseases, wherein the neurogenic diseases are selected from gynecological diseases, urinary tract disease states, respiratory disorder diseases or pain related diseases or symptoms and the like.
The invention further relates to a method for preparing the compound shown in the general formula (I), the stereoisomer or the pharmaceutically acceptable salt thereof or the pharmaceutical composition thereof for treating the neurogenic disease.
The invention also relates to a method for the therapeutic prophylaxis and/or treatment of pre-prepared neurogenic diseases, which comprises administering to a patient a therapeutically effective dose of a compound of formula (I), a stereoisomer thereof or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
The invention also provides methods of treating disease conditions, including but not limited to conditions associated with P2X3 receptor dysfunction, using the compounds or pharmaceutical compositions of the invention.
The invention also relates to a method of treating a neurogenic disease in a mammal comprising administering to said mammal a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
In some embodiments, the present methods relate to a disorder or condition such as a gynecological disease, a urinary tract disease state, a respiratory disorder disease, or pain related disease or condition, and the like.
In some embodiments, the present methods relate to the treatment of conditions such as endometriosis, overactive bladder, pulmonary fibrosis, or chronic cough.
In some embodiments, the present methods relate to neuropathic pain or uterine fibroid-related pain and discomfort.
Chronic cough and neuropathic pain are preferred;
more preferably chronic cough.
Detailed description of the invention
Unless stated to the contrary, the terms used in the specification and claims have the following meanings.
The term "alkyl" refers to a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing from 1 to 20 carbon atoms, preferably an alkyl group containing from 1 to 8 carbon atoms, more preferably an alkyl group containing from 1 to 6 carbon atoms, most preferably an alkyl group containing from 1 to 3 carbon atoms. Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl 5-methylhexyl, 2, 3-dimethylpentyl, 2, 4-dimethylpentyl, 2-dimethylpentyl, 3-dimethylpentyl, 2-ethylpentyl, 3-ethylpentyl, n-octyl, 2, 3-dimethylhexyl, 2, 4-dimethylhexyl, 2, 5-dimethylhexyl, 2-dimethylhexyl, 3-dimethylhexyl 4, 4-dimethylhexyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl, n-nonyl, 2-methyl-2-ethylhexyl, 2-methyl-3-ethylhexyl, 2-diethylpentyl, n-decyl, 3-diethylhexyl, 2-diethylhexyl, and various branched isomers thereof. More preferred are lower alkyl groups containing 1 to 6 carbon atoms, and non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, and the like. The alkyl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxy or carboxylate, with methyl, ethyl, isopropyl, t-butyl, haloalkyl, deuteroalkyl, alkoxy-substituted alkyl and hydroxy-substituted alkyl being preferred.
The term "alkylene" means that one hydrogen atom of the alkyl group is further substituted, for example: "methylene" means-CH 2 - "ethylene" means- (CH) 2 ) 2 - "propylene" means- (CH) 2 ) 3 "butylene" means- (CH) 2 ) 4 -and the like. The term "alkenyl" refers to an alkyl group as defined above consisting of at least two carbon atoms and at least one carbon-carbon double bond, such as vinyl, 1-propenyl, 2-propenyl, 1-, 2-, or 3-butenyl, and the like. Alkenyl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio.
The term "cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring containing from 3 to 20 carbon atoms, preferably from 3 to 12 carbon atoms, more preferably from 3 to 6 carbon atoms. Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, and the like; polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups, preferably cyclopropyl, cyclobutyl, cyclohexyl, cyclopentyl and cycloheptyl.
The term "spirocycloalkyl" refers to a polycyclic group sharing one carbon atom (referred to as a spiro atom) between 5-to 20-membered monocyclic rings, which may contain one or more double bonds, but no ring has a fully conjugated pi-electron system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The spirocycloalkyl group is classified into a single spirocycloalkyl group, a double spirocycloalkyl group or a multiple spirocycloalkyl group according to the number of common spiro atoms between rings, and preferably a single spirocycloalkyl group and a double spirocycloalkyl group. More preferably 3-membered/6-membered, 3-membered/5-membered, 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered, mono-spirocycloalkyl. Non-limiting examples of spirocycloalkyl groups include:
etc.;
spirocycloalkyl groups also containing a spiro atom common to both the monocyclocycloalkyl and heterocycloalkyl groups, non-limiting examples include:
etc.
The term "fused ring alkyl" refers to a 5 to 20 membered, all carbon polycyclic group wherein each ring in the system shares an adjacent pair of carbon atoms with the other rings in the system, wherein one or more of the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi electron system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The number of constituent rings may be classified as a bicyclic, tricyclic, tetracyclic or polycyclic fused ring alkyl group, preferably a bicyclic or tricyclic, more preferably a 5-membered/5-membered or 5-membered/6-membered bicycloalkyl group. Non-limiting examples of fused ring alkyl groups include:
Etc.
The term "bridged cycloalkyl" refers to an all-carbon polycyclic group of 5 to 20 members, any two rings sharing two carbon atoms not directly attached, which may contain one or more double bonds, but no ring has a fully conjugated pi-electron system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. Cycloalkyl groups which may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged according to the number of constituent rings are preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of bridged cycloalkyl groups include:
/>
the cycloalkyl ring may be fused to an aryl, heteroaryl, or heterocycloalkyl ring, where the ring attached to the parent structure is cycloalkyl, non-limiting examples include indanyl, tetrahydronaphthyl, benzocycloheptyl, and the like. Cycloalkyl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxyl, or carboxylate groups.
The term "heterocyclyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 ring atoms in which one or more ring atoms are selected from nitrogen, oxygen or S (O) m (wherein m is an integer from 0 to 2), but does not include a ring moiety of-O-O-, -O-S-, or-S-S-, and the remaining ring atoms are carbon. Preferably containing 3 to 12 ring atoms, of which 1 to 4 are heteroatoms; more preferably 3 to 8 ring atoms; most preferably containing 3 to 8 ring atoms; further preferred is a 3-8 membered heterocyclic group containing 1 to 3 nitrogen atoms, optionally substituted with 1 to 2 oxygen atoms, sulfur atoms, oxo groups, including a nitrogen-containing monocyclic heterocyclic group, a nitrogen-containing spiro heterocyclic group or a nitrogen-containing condensed heterocyclic group.
Non-limiting examples of monocyclic heterocyclyl groups include azetidinyl, pyrrolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dioxazolyl, dihydroimidazolyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrrolyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, azepinyl, 1, 4-diazepinyl, pyranyl, and the like, with azetidinyl, pyrrolidinyl, tetrahydropyranyl, dioxazolyl, morpholinyl, piperidinyl, azepinyl, 1, 4-diazepinyl, and piperazinyl being preferred. Polycyclic heterocyclyl groups include spiro, fused and bridged heterocyclic groups; the heterocyclic groups of the spiro ring, the condensed ring and the bridged ring are optionally connected with other groups through single bonds, or are further connected with other cycloalkyl groups, heterocyclic groups, aryl groups and heteroaryl groups through any two or more atoms on the ring in a parallel ring mode.
The term "spiroheterocyclyl" refers to a polycyclic heterocyclic group having a single ring of 5 to 20 members sharing one atom (referred to as the spiro atom) between them, wherein one or more of the ring atoms is selected from nitrogen, oxygen or S (O) m (wherein m is an integer from 0 to 2) and the remaining ring atoms are carbon. Which may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The spiroheterocyclyl groups are classified into a single spiroheterocyclyl group, a double spiroheterocyclyl group or a multiple spiroheterocyclyl group according to the number of common spiro atoms between rings, and preferably a single spiroheterocyclyl group and a double spiroheterocyclyl group. More preferably 3-membered/5-membered, 3-membered/6-membered, 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered single spiro heterocyclyl. Non-limiting examples of spiroheterocyclyl groups include:
etc.
The term "fused heterocyclyl" refers to 5 to 20 membered, polycyclic heterocyclic groups in which each ring in the system shares an adjacent pair of atoms with the other rings in the system, one or more of which may contain one or moreDouble bonds, but none of the rings have a fully conjugated pi-electron system in which one or more ring atoms are selected from nitrogen, oxygen or S (O) m (wherein m is an integer from 0 to 2) and the remaining ring atoms are carbon. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The number of constituent rings may be classified as a bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic group, preferably a bicyclic or tricyclic, more preferably a 5-membered/5-membered or 5-membered/6-membered bicyclic fused heterocyclic group. Non-limiting examples of fused heterocyclyl groups include:
Etc.
The term "bridged heterocyclyl" refers to a 5 to 14 membered, polycyclic heterocyclic group in which any two rings share two atoms not directly attached, which may contain one or more double bonds, but none of the rings has a fully conjugated pi electron system in which one or more ring atoms are selected from nitrogen, oxygen, or S (O) m (wherein m is an integer from 0 to 2) and the remaining ring atoms are carbon. Preferably 6 to 14 membered, more preferably 7 to 10 membered. Heterocyclic groups which may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged according to the number of constituent rings are preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of bridged heterocyclyl groups include:
etc.
The heterocyclyl ring may be fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring attached to the parent structure is heterocyclyl, non-limiting examples of which include:
etc.
The heterocyclic group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxyl, or carboxylate groups.
The term "aryl" refers to a 6 to 14 membered all-carbon monocyclic or fused polycyclic (i.e., rings sharing adjacent pairs of carbon atoms) group having a conjugated pi-electron system, preferably 6 to 12 membered, such as phenyl and naphthyl. More preferably phenyl. The aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, including benzo 5-10 membered heteroaryl, benzo 3-8 membered cycloalkyl and benzo 3-8 membered heteroalkyl, preferably benzo 5-6 membered heteroaryl, benzo 3-6 membered cycloalkyl and benzo 3-6 membered heteroalkyl, wherein heterocyclyl is a heterocyclyl containing 1-3 nitrogen, oxygen, sulfur atoms; or further comprises a ternary nitrogen-containing fused ring containing a benzene ring.
Wherein the ring attached to the parent structure is an aryl ring, non-limiting examples of which include:
etc.
Aryl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl, or carboxylate groups.
The term "heteroaryl" refers to a heteroaromatic system containing from 1 to 4 heteroatoms, from 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur and nitrogen. Heteroaryl is preferably 5 to 12 membered, more preferably 5 or 6 membered, such as imidazolyl, furyl, thienyl, thiazolyl, pyrazolyl, oxazolyl, pyrrolyl, triazolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazolyl, pyrazinyl, and the like, preferably pyridyl, triazolyl, thienyl, imidazolyl, pyrazolyl, oxazolyl, oxadiazolyl, pyrimidinyl, or thiazolyl; more preferably pyridyl, thiazolyl, oxadiazolyl, pyrazolyl, pyrrolyl and oxazolyl. The heteroaryl ring may be fused to an aryl, heterocyclyl, or cycloalkyl ring, wherein the ring attached to the parent structure is a heteroaryl ring, non-limiting examples of which include:
Heteroaryl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl, or carboxylate groups.
The term "alkoxy" refers to-O- (alkyl) and-O- (unsubstituted cycloalkyl), wherein alkyl is as defined above. Non-limiting examples of alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentoxy, cyclohexyloxy. The alkoxy groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl, or carboxylate groups.
"haloalkyl" refers to an alkyl group substituted with one or more halogens, where alkyl is as defined above.
"haloalkoxy" refers to an alkoxy group substituted with one or more halogens, wherein the alkoxy group is as defined above.
"hydroxyalkyl" refers to an alkyl group substituted with a hydroxy group, wherein alkyl is as defined above.
"alkenyl" refers to alkenyl groups, also known as alkenyl groups, wherein the alkenyl groups may be further substituted with other related groups, such as: alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl or carboxylate groups.
"alkynyl" refers to (CH≡C-), wherein the alkynyl group may be further substituted with other related groups, such as: alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl or carboxylate groups.
The term "alkenylcarbonyl" refers to-C (O) - (alkenyl), wherein alkenyl is as defined above. Non-limiting examples of alkenylcarbonyl groups include: vinylcarbonyl, propenylcarbonyl, butenylcarbonyl. The alkenylcarbonyl group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl, or carboxylate.
"hydroxy" refers to an-OH group.
"halogen" means fluorine, chlorine, bromine or iodine.
"amino" means-NH 2
"cyano" refers to-CN.
"nitro" means-NO 2
"carbonyl" means-C (O) -.
"carboxy" means-C (O) OH.
"THF" refers to tetrahydrofuran.
"EtOAc" refers to ethyl acetate.
"MeOH" refers to methanol.
"DMF" refers to N, N-dimethylformamide.
"DIPEA" refers to diisopropylethylamine.
"TFA" refers to trifluoroacetic acid.
"MeCN" refers to acetonitrile.
"DMA" refers to N, N-dimethylacetamide.
“Et 2 O "refers to diethyl ether.
"DCE" refers to 1,2 dichloroethane.
"DIPEA" refers to N, N-diisopropylethylamine.
"NBS" refers to N-bromosuccinimide.
"NIS" refers to N-iodosuccinimide.
"Cbz-Cl" refers to benzyl chloroformate.
“Pd 2 (dba) 3 "means tris (dibenzylideneacetone) dipalladium.
"Dppf" refers to 1,1' -bis-diphenylphosphino ferrocene.
"HATU" refers to 2- (7-oxo-benzotriazol) -N, N' -tetramethylurea hexafluorophosphate.
"KHMDS" refers to potassium hexamethyldisilazide.
"LiHMDS" refers to lithium bis (trimethylsilylamide).
"MeLi" refers to lithium-based.
"n-BuLi" refers to n-butyllithium.
“NaBH(OAc) 3 "means sodium triacetoxyborohydride.
The terms "X is selected from A, B or C", "X is selected from A, B and C", "X is A, B or C", "X is A, B and C", etc. all express the same meaning, that is, X may be any one or several of A, B, C.
The hydrogen atoms of the invention can be replaced by the isotope deuterium thereof, and any hydrogen atom in the compound of the embodiment of the invention can be replaced by deuterium atoms.
"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, "a heterocyclic group optionally substituted with an alkyl group" means that an alkyl group may be, but is not necessarily, present, and the description includes cases where the heterocyclic group is substituted with an alkyl group and cases where the heterocyclic group is not substituted with an alkyl group.
"substituted" means that one or more hydrogen atoms, preferably up to 5, more preferably 1 to 3 hydrogen atoms in the group are independently substituted with a corresponding number of substituents. It goes without saying that substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (by experiment or theory) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups having free hydrogen may be unstable when bound to carbon atoms having unsaturated (e.g., olefinic) bonds.
"pharmaceutical composition" means a mixture comprising one or more of the compounds described herein or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to promote the administration to organisms, facilitate the absorption of active ingredients and thus exert biological activity.
By "pharmaceutically acceptable salts" is meant salts of the compounds of the present invention which are safe and effective when used in a mammal, and which possess the desired biological activity.
Detailed Description
The invention is further described below in connection with examples, which are not intended to limit the scope of the invention.
Examples
The structure of the compounds of the present invention is determined by Nuclear Magnetic Resonance (NMR) or/and liquid chromatography-mass spectrometry (LC-MS). NMR chemical shifts (δ) are given in parts per million (ppm). NMR was performed using Bruker AVANCE-400 nuclear magnetic resonance apparatus with deuterated dimethyl sulfoxide (DMSO-d) 6 ) Deuterated methanol (CD) 3 OD) and deuterated chloroform (CDCl) 3 ) The internal standard is Tetramethylsilane (TMS).
An Agilent 1200 affinity Series mass spectrometer was used for LC-MS measurement. HPLC was performed using Agilent 1200DAD high pressure liquid chromatograph (Sunfire C18X 4.6mm column) and Waters 2695-2996 high pressure liquid chromatograph (Gimini C) 18 150X 4.6mm column).
The thin layer chromatography silica gel plate uses a smoke table yellow sea HSGF254 or Qingdao GF254 silica gel plate, the specification adopted by TLC is 0.15 mm-0.20 mm, and the specification adopted by the thin layer chromatography separation and purification product is 0.4 mm-0.5 mm. Column chromatography generally uses tobacco stand yellow sea silica gel 200-300 mesh silica gel as a carrier.
The starting materials in the examples of the present invention are known and commercially available or may be synthesized using or according to methods known in the art.
All reactions of the invention were carried out under continuous magnetic stirring under dry nitrogen or argon atmosphere, with the solvent being a dry solvent and the reaction temperature being in degrees celsius, without specific explanation.
Example 1
4- (1- (2- ((5-chloropyridin-2-yl) amino) -2-oxoethyl) -5-methyl-1H-benzo [ d ] imidazol-2-yl) -3-fluoro-N-methylbenzamide
The first step: preparation of 2-bromo-N- (5-fluoropyridin-2-yl) acetamide
To a solution of (1 g,7.78 mmol) 5-fluoro-pyridin-2-ylamine and 0.3mL pyridine in 20mL toluene was added dropwise bromoacetyl bromide dissolved in 5mL toluene (1.9 g,7.92 mmol) under ice-cooling. After 2 hours, a white solid was precipitated, and the precipitate was separated by filtration and recrystallized from toluene to obtain example 1-1 (1.8 g, yield: 70%) as a white solid.
MS m/z(ESI):249.23[M+H] + .
1 H NMR(400MHz,MeOD)δ8.29(d,J=2.7Hz,1H),8.12(d,J=8.9Hz,1H),7.80(dd,J=8.9,2.7Hz,1H),4.04(s,2H).
And a second step of: preparation of 4- (bromomethyl) -3-fluorobenzoic acid
3-fluoro-4-methyl-benzoic acid (7 g,45.4 mmol) and NBS (8.08 g,45.4 mmol) in CHCl 3 The suspension in (150 mL) was heated to reflux until the solution was homogeneous. AIBN (100 mg,0.61 mmol) was added to the reaction mixture and the resulting mixture was stirred under reflux overnight. The reaction was cooled to room temperature and concentrated in vacuo to give an orange solid. The material was slurried with a mixture of DCM (50 mL) and n-hexane (50 mL) and the byproducts filtered off. The filtrate was concentrated in vacuo and purified by column chromatography eluting with petroleum ether in ethyl acetate (0-100%). The title compound example 1-2 (5.22 g,13.4mmol, yield: 30%) was isolated as a white solid.
MS m/z(ESI):233.23[M+H] + .
And a third step of: preparation of 4- (bromomethyl) -3-fluoro-N-methylbenzamide
Examples 1-2 (1.12 g,5.20 mmol) were dissolved in toluene (30 mL), thionyl chloride (7.59 mL,104 mmol) was added, and stirred under reflux with heating for 5 hours. The reaction mixture was concentrated under reduced pressure, and the resulting residue was dissolved in THF (20 mL). To this was added methylammonium hydrochloride (0.67 g,4.11 mmol), triethylamine (0.86 mL,6.17 mmol), and then stirred at room temperature for 1 hour. The precipitated solid was filtered, and the precipitated solid was collected and then washed with diethyl ether to give examples 1-3 (1.28 g, 60%).
MS m/z(ESI):246.13[M+H] + .
Fourth step: preparation of 3-fluoro-N-methyl-4- (5-methyl-1H-benzo [ d ] imidazol-2-yl) benzamide
Examples 1-3 (2 g,8.19 mmol), pyridine-N-oxide (1.95 g,20.5 mmol) and 4-methylbenzene-1, 2-diamine (1 g,8.19 mmol) were thoroughly mixed in a 100mL round bottom flask and placed under reflux with heating for 4h using a magnetic stirrer. After the reaction was complete (TLC, ethyl acetate: petroleum ether=1:1), the organic mixture was washed with dilute NaOH and filtered off. Toluene recrystallization gave examples 1-4 (2 g, yield: 86%).
MS m/z(ESI):284.32[M+H] + .
Fifth step: preparation of 4- (1- (2- ((5-fluoropyridin-2-yl) amino) -2-oxoethyl) -5-methyl-1H-benzo [ d ] imidazol-2-yl) -3-fluoro-N-methylbenzamide
Examples 1-14 (100 mg,0.35 mmol) and examples 1-1 (88 mg,0.35 mmol), K 2 CO 3 (97.6 mg,0.7 mmol) in DMF (5 mL) was heated to 50deg.C and reacted for 2h, after completion of the reaction by LCMS detection, 10mL of water and 20mL of ethyl acetate were added to extract, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product, which was eluted with ethyl acetate solution of petroleum ether (0-100%) by silica gel chromatography to give example 1 (50 mg, yield: 30%).
MS m/z(ESI):452.23[M+H] + .
Example 2
(S) -6- (3- ((4-Acetylmorpholin-2-yl) methyl) -7-methylimidazole [1,2-a ] pyridin-2-yl) -5, 7-difluoro-2-methyl-3, 4-dihydroisoquinolin-1 (2H) -one
The first step: preparation of 5, 7-difluoro-2-methyl-3, 4-dihydroisoquinolin-1 (2H) -one
Example 2-1 (800 mg,5 mmol) was dissolved in acetonitrile (10 ml), 1-chloromethyl-4-fluoro-1, 4-diazobicyclo [2.2.2] octane bis (tetrafluoroborate) (3.54 g,10 mmol) was added and the mixture was heated at 80℃to reflux overnight. After the reaction solution was cooled to room temperature, it was concentrated, and then purified by a silica gel column to give example 2-2 (460 mg,47% yield).
MS m/z(ESI):198.0[M+H] +
And a second step of: preparation of 5, 7-difluoro-2-methyl-1-oxo-1, 2,3, 4-tetrahydroquinoline-6-carbaldehyde
To a solution of example 2-2 (460 mg,2.33 mmol) in 2-methyltetrahydrofuran (6 mL) was added TMEDA (595 mg,5.13 mmol) at room temperature. The resulting solution was cooled to-78 ℃ and n-butyllithium (1 m,2.6 ml) was added dropwise. After stirring the mixture at-78 ℃ for 1.5 hours, anhydrous methyl formate (290 mg,4.8 mmol) was added, stirring was continued for 45 minutes at-78 ℃ and then the dry ice bath was removed. The mixture was stirred at room temperature for 18 hours, then the mixture was cooled to 0-5 ℃ and the excess base was quenched with 6M water HCI and the aqueous layer extracted 3 times with ethyl acetate. The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated. The residue was slurried with minimal ethyl acetate, filtered, washed with hexane and dried under vacuum to give examples 2-3 (180 mg,34% yield).
MS m/z(ESI):226.1[M+H] +
And a third step of: preparation of (S) -6- (3- ((4-acetylmorpholin-2-yl) methyl) -7-methylimidazole [1,2-a ] pyridin-2-yl) -5, 7-difluoro-2-methyl-3, 4-dihydroisoquinolin-1 (2H) -one
At N 2 A mixture of 4-methylpyridin-2-amine (54 mg,0.50 mmo1), examples 2-3 (100 mg,0.50 mmo1), (S) -1- (2-ethynylmorpholine) ethan-1-one (75 mg,0.50 mmol), cuprous chloride (15 mg,0.15 mmol), copper bis (trifluoromethylsulfonyloxy) (54 mg,0.15 mmo1) and toluene (2 mL) was added to the reactor, heated to 85℃and then N, N-dimethylacetamide (0.1 mL) was added, and the mixture was stirred at 85℃for 5 hours, followed by overnight at room temperature. The reaction solution was concentrated under reduced pressure and purified by preparative HPLC to give example 2 (16 mg,7% yield).
MS m/z(ESI):469.0[M+H] + .
Example 3
(S) -6- (3- ((4-Acetylmorpholin-2-yl) methyl) -7-methylimidazole [1,2-a ] pyridin-2-yl) -5, 7-difluoro-2-methylisoquinolin-1 (2H) -one
Synthetic method of example 3 referring to example 2, substituting 2-methylisoquinolin-1 (2H) -one for example 1-1 gave example 3 (23 mg,10% yield).
MS m/z(ESI):467.1[M+H] + .
Example 4
((S) -5- (3- ((4-Acetylmorpholin-2-yl) methyl) -7-methylimidazole [1,2-a ] pyridin-2-yl) -4, 6-difluoro-2-methylisoindol-1-one
Synthetic method for example 4 referring to example 2, substituting 2-methylisoindolin-1-one for example 1-1 gave example 4 (18 mg,7% yield).
MS m/z(ESI):455.0[M+H] + .
Example 5
N- (4- (4-chlorobenzyl) -5-oxo-2- (4- (pyridin-2-yloxy) phenyl) -4, 5-dihydropyrazolo [1,5-a ] pyrimidin-6-yl) tetrahydro-2H-pyran-4-carboxamide
The first step: preparation of methyl 4- (pyridin-2-yloxy) benzoate
Example 5-1 (2 g,9.29 mmol) was dissolved in toluene (20 mL), thionyl chloride (7.5 mL,93 mmol) was added, and the mixture was stirred under reflux with heating for 3 hours. The reaction mixture was concentrated under reduced pressure and the resulting residue was dissolved in MeOH (50 mL). Spin-drying and collecting the precipitated solid, which was then washed with diethyl ether to give example 5-2 (2 g,93% yield).
MS m/z(ESI):230.0[M+H] + .
And a second step of: preparation of 4- (bromomethyl) -3-fluorobenzoic acid
A1M solution of LHMDS in dry THF (26.3 mL,26.3 mmol) was cooled to-78deg.C and acetonitrile (1.43 mL,27.5 mmol) was added dropwise over 2 minutes. The mixture was stirred at-78deg.C for 1 hour, and a solution of example 5-2 (5.75 g,25.0 mmol) in dry THF (50 mL) was added. The mixture was stirred for 3 hours and the dry ice bath was removed to bring the mixture to ambient temperature. TLC detection reaction was complete, 1M HCl was added dropwise to the solution to pH=3%Initial ph=12). The mixture was extracted with ethyl acetate (2 x 80 ml) and the combined extracts were subjected to Na 2 SO 4 And (5) drying. The crude product was eluted with ethyl acetate solution (0-50%) of petroleum ether by silica gel chromatography. Example 5-3 was obtained as a colorless oil (4 g,67% yield).
MS m/z(ESI):239.1[M+H] + .
And a third step of: preparation of 3- (4- (pyridin-2-yloxy) phenyl) -1H-pyrazol-5-amine
Examples 5-3 (2 g,8.39 mmol) were dissolved in ethanol (25 mL) at room temperature, and glacial acetic acid (1 mL) and hydrazine hydrate (1.26 g,25.18 mmol) were added. The mixture was added to hot 80℃and the reaction stirred for 2 hours. After the mixture was cooled to room temperature, naHCO was added 3 The solution was extracted three times with ethyl acetate. The organic phase was washed with brine, dried and concentrated to give examples 5-4 (1.5 g,70% yield).
MS m/z(ESI):253.1[M+H] + .
Fourth step: preparation of ethyl 5-oxo-2- (4- (pyridin-2-yloxy) phenyl) -4, 5-dihydropyrazolo [1,5-a ] pyrimidine-6-carboxylate
Examples 5-4 (1.5 g,5.95 mmol) were dissolved in ethanol (35 mL) and diethyl 2-ethoxymethylenemalonate (1.46 g,6.54 mmol) and sodium ethoxide (0.607 g,8.92 mmol) were added to the mixture at room temperature. The mixture was added to hot 100℃and the reaction stirred for 5 hours. After cooling, the precipitate was filtered and washed with ethanol to give examples 5-5 (1.6 g,71% yield) as a white powder.
MS m/z(ESI):377.1[M+H] + .
Fifth step: preparation of ethyl 4- (4-chlorobenzyl) -5-oxo-2- (4- (pyridin-2-yloxy) phenyl) -4, 5-dihydropyrazolo [1,5-a ] pyrimidine-6-carboxylate
Examples 5-5 (1 g,2.66 mmol) were dissolved in DMF (15 mL) and 1- (bromomethyl) -4-chlorobenzene (0.82 g,3.99 mmol) and potassium carbonate (0.73 g,5.31 mmol) were added to the mixture at room temperature. The mixture was added to heat at 50℃and the reaction was stirred for 2 hours. After cooling, 30mL of water was added, the mixture was extracted with ethyl acetate (2 x 40 mL) and the combined extracts were taken over Na 2 SO 4 And (5) drying. The crude spin-dried product was slurried with petroleum ether (30 mL) to give examples 5-6 (1.1 g,83% yield) as a white powder.
MS m/z(ESI):501.2[M+H] + .
Sixth step: preparation of 4- (4-chlorobenzyl) -5-oxo-2- (4- (pyridin-2-yloxy) phenyl) -4, 5-dihydropyrazolo [1,5-a ] pyrimidine-6-carboxylic acid
Examples 5-6 (0.2 g,0.4 mmol) were dissolved in THF (6 mL) and water (1.5 mL) at room temperature, and lithium hydroxide (20 mg,0.48 mmol) was added to the mixture. The mixture was stirred at room temperature for 2 hours. 1M HCl was added to ph=3, the mixture was extracted with ethyl acetate (2×40 ml), and the combined extracts were taken up over Na 2 SO 4 And (5) drying. The crude spin-dried product was slurried with petroleum ether (10 mL) to give examples 5-7 (0.15 g,80% yield) as a white powder.
MS m/z(ESI):473.0[M+H] + .
Seventh step: preparation of tert-butyl (4- (4-chlorobenzyl) -5-oxo-2- (4- (pyridin-2-yloxy) phenyl) -4, 5-dihydropyrazol [1,5-a ] pyrimidin-6-yl) carbamate
Examples 5-7 (140 mg,0.3 mmol) were dissolved in t-butanol (5 ml), triethylamine (50 mg,0.5 mmol) was added and stirred for 30 min, followed by DPPA (138 mg,0.5 mmol). The reaction mixture was heated to 100 ℃ and stirred overnight. After cooling, the solvent was removed by concentration and TBME was slurried to give examples 5-8 (102 mg,63% yield)
MS m/z(ESI):544.2[M+H] + .
Eighth step: preparation of 6-amino-4- (4-chlorobenzyl) -2- (4- (pyridin-2-yloxy) phenyl) pyrazolo [1,5-a ] pyrimidin-5 (4 h) -one
Examples 5-8 (102 mg,0.19 mmol) were dissolved in DCM (6 mL) and a mixture of TFA (3 mL) was added and stirred at room temperature for 2 hours. The reaction solution was concentrated to give examples 5 to 9 (82 mg,63% yield)
MS m/z(ESI):444.1[M+H] + .
Step 9: preparation of N- (4- (4-chlorobenzyl) -5-oxo-2- (4- (pyridin-2-yloxy) phenyl) -4, 5-dihydropyrazolo [1,5-a ] pyrimidin-6-yl) tetrahydro-2H-pyran-4-carboxamide
Examples 5-9 (20 mg,0.042 mmol) were dissolved in DMF (1 mL) at room temperature, and HATU (16 mg,0.084 mmol) and DIEA (11 mg,0.084 mmol) were added to the mixture. The mixture was stirred at room temperature for 10min. Then, tetrahydro-2H-pyran-4-carboxylic acid (5.13 mg,0.05 mmol) was added thereto, and the mixture was stirred at room temperature for 16H. 10mL of water was added to the reaction, the mixture was extracted with ethyl acetate (2 x 10 mL), and the combined extracts were taken up in Na 2 SO 4 Drying, spin-drying, and p-TLC (dichloro: methanol=20:1) gave example 5 (11 mg, yield: 43%) as a white powder.
MS m/z(ESI):557.0[M+H] + .
Example 6
4- (4-chlorobenzyl) -5-oxo-2- (4- (pyridin-2-yloxy) phenyl) -N- (tetrahydro-2H-pyran-4-yl) -4, 5-dihydropyrazolo [1,5-a ] pyrimidine-6-carboxamide
The first step: preparation of methyl 4- (pyridin-2-yloxy) benzoate
4- (pyridin-2-yloxy) benzoic acid (2 g,9.29 mmol) was dissolved in toluene (20 mL), thionyl chloride (7.5 mL,93 mmol) was added and stirred under reflux with heating for 3 hours. The reaction mixture was concentrated under reduced pressure and the resulting residue was dissolved in MeOH (50 mL). Spin-drying, collecting the precipitated solid, and washing with diethyl ether gave example 6-1 (2 g, yield: 93%).
MS m/z(ESI):230.12[M+H] + .
And a second step of: preparation of 4- (bromomethyl) -3-fluorobenzoic acid
A1M solution of LHMDS in dry THF (26.3 mL,26.3 mmol) was cooled to-78deg.C and acetonitrile (1.43 mL,27.5 mmol) was added dropwise over 2 minutes. The mixture was stirred at-78deg.C for 1 hour, and a solution of example 6-1 (5.75 g,25.0 mmol) in dry THF (50 mL) was added. The mixture was stirred for 3 hours and the dry ice bath was removed to bring the mixture to ambient temperature. TLC detects completion of reaction, 1M HCl was added dropwise to the solution to ph=3 (starting ph=12). The mixture was extracted with ethyl acetate (2 x 80 ml) and the combined extracts were subjected to Na 2 SO 4 And (5) drying. The crude product was eluted with ethyl acetate solution (0-50%) of petroleum ether by silica gel chromatography. Example 6-2 was obtained as a colorless oil (4 g, 67%).
MS m/z(ESI):239.12[M+H] + .
And a third step of: preparation of 3- (4- (pyridin-2-yloxy) phenyl) -1H-pyrazol-5-amine
Example 6-2 (2 g,8.39 mmol) was dissolved in ethanol (25 mL) at room temperature, followed by glacial acetic acid (1 mL) and hydrazine hydrate (1.26 g,25.18 mmol). The mixture was added to heat 80℃and the reaction was stirred for 2h. After the mixture was cooled to room temperature, naHCO was added 3 The solution was extracted three times with ethyl acetate. The organic phase was washed with brine, dried and concentrated to give example 6-3 (1.5 g, yield: 70%).
MS m/z(ESI):253.10[M+H] + .
Fourth step: preparation of ethyl 5-oxo-2- (4- (pyridin-2-yloxy) phenyl) -4, 5-dihydropyrazolo [1,5-a ] pyrimidine-6-carboxylate
Example 6-3 (1.5 g,5.95 mmol) was dissolved in ethanol (35 mL) and diethyl 2-ethoxymethylenemalonate (1.46 g,6.54 mmol) and sodium ethoxide (0.607 g,8.92 mmol) were added to the mixture at room temperature. The mixture was added to heat at 100℃and the reaction was stirred for 5h. After cooling, the precipitate was filtered and washed with ethanol to give examples 6-4 (1.6 g, 71%) as a white powder.
MS m/z(ESI):377.12[M+H] + .
Fifth step: preparation of ethyl 4- (4-chlorobenzyl) -5-oxo-2- (4- (pyridin-2-yloxy) phenyl) -4, 5-dihydropyrazolo [1,5-a ] pyrimidine-6-carboxylate
Examples 6-4 (1 g,2.66 mmol) were dissolved in DMF (15 mL) and 1- (bromomethyl) -4-chlorobenzene (0.82 g,3.99 mmol) and potassium carbonate (0.73 g,5.31 mmol) were added to the mixture at room temperature. The mixture was added to heat at 50℃and the reaction was stirred for 2h. After cooling, 30mL of water was added, the mixture was extracted with ethyl acetate (2 x 40 mL) and the combined extracts were taken over Na 2 SO 4 And (5) drying. Rotary screwThe dried crude product was slurried with petroleum ether (30 mL) to give examples 6-5 (1.1 g, yield: 83%) as a white powder.
MS m/z(ESI):501.13[M+H] + .
Sixth step: preparation of 4- (4-chlorobenzyl) -5-oxo-2- (4- (pyridin-2-yloxy) phenyl) -4, 5-dihydropyrazolo [1,5-a ] pyrimidine-6-carboxylic acid
Examples 6-5 (0.2 g,0.4 mmol) were dissolved in THF (6 mL) and water (1.5 mL) at room temperature, and lithium hydroxide (20 mg,0.48 mmol) was added to the mixture. The mixture was stirred at room temperature for 2h. 1M HCl was added to ph=3, the mixture was extracted with ethyl acetate (2×40 ml), and the combined extracts were taken up over Na 2 SO 4 And (5) drying. The crude product was dried by spinning and slurried with petroleum ether (10 mL) to give examples 6-6 (0.15 g, yield: 80%) as a white powder.
MS m/z(ESI):473.09[M+H] + .
Seventh step: preparation of 4- (4-chlorobenzyl) -5-oxo-2- (4- (pyridin-2-yloxy) phenyl) -N- (tetrahydro-2H-pyran-4-yl) -4, 5-dihydropyrazolo [1,5-a ] pyrimidine-6-carboxamide
Examples 6-6 (20 mg,0.042 mmol) were dissolved in DMF (1 mL) and HATU (16 mg,0.084 mmol) and DIEA (11 mg,0.084 mmol) were added to the mixture at room temperature. The mixture was stirred at room temperature for 10min. Then, tetrahydro-2H-pyran-4-amine (5.13 mg,0.05 mmol) was added thereto, and the mixture was stirred at room temperature for 16H. 10mL of water was added to the reaction, the mixture was extracted with ethyl acetate (2 x 10 mL), and the combined extracts were taken up in Na 2 SO 4 Drying, spin-drying, crude product was obtained by p-TLC (dichloro: methanol=20:1) as a white powder of example 6 (10 mg, yield: 43%).
MS m/z(ESI):556.17[M+H] + .
Example 7
4- (4-chlorobenzyl) -6- (3-methyl-1, 2, 4-oxadiazol-5-yl) -2- (4- (pyridin-2-yloxy) phenyl) pyrazolo [1,5-a ] pyrimidin-5 (4H) -one
The first step: preparation of N-hydroxyacetamidine
To a solution of (0.57 g,14.2 mmol) NaOH in 5mL of water was added (1.00 g,14.2 mmol) hydroxylamine. 15mL of acetonitrile was added dropwise and the mixture was stirred at 37℃overnight at room temperature. Acetonitrile and water were removed in vacuo and ethanol was added to the crude product. The product was filtered off and washed with ethanol to give example 7-1 (0.6 g, yield: 60%).
1 H NMR(400MHz,DMSO-d6)δ8.65(s,1H),5.33(br,2H),1.60(s,3H).
And a second step of: preparation of 4- (4-chlorobenzyl) -6- (3-methyl-1, 2, 4-oxadiazol-5-yl) -2- (4- (pyridin-2-yloxy) phenyl) pyrazolo [1,5-a ] pyrimidin-5 (4H) -one
Examples 6-5 (50 mg,0.1 mmol) were added to toluene (5 mL) followed by N-hydroxyacetimidate (15 mg,0.2 mmol). The reaction mixture was heated to reflux, stirred for 16 hours and cooled to room temperature. The reaction mixture was concentrated, and then, example 7 (25 mg, yield: 50%) was obtained by preparation.
MS m/z(ESI):499.13[M+H] + .
1 HNMR(400MHz,DMSO-d6)δ7.80(s,1H),7.39–7.34(m,2H),7.29(dt,J=7.5,1.1Hz,2H),5.72(s,1H),5.30(d,J=1.1Hz,2H),3.84–3.77(m,2H),3.79–3.71(m,2H),3.01–2.92(m,1H),2.44(s,2H),2.31(qd,J=7.1,3.4Hz,4H).
Example 8
1- (4-chlorobenzyl) -6- (4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) -3-ethyl-1, 7-dihydro-2H-pyrrolo [2,3-D ] pyrimidine-2, 4 (3H) -dione
The first step: preparation of 5-bromo-2- ((4-chlorobenzyl) amino) -N-ethyl-1H-pyrrole-3-carboxamide
Example 8-1 (1.2 g,5.2 mmol) was dissolved in methanol (15 ml) at room temperature, 4-chlorobenzaldehyde (0.73 g,5.2 mmol) was added to the mixture, and the mixture was stirred at room temperature for 5 hours, and NaBH was added to the reaction mixture 4 (0.4 g,10.38 mmol). The mixture was added to heat at 50℃and the reaction was stirred for 2 hours. After cooling, 20 ml of water was added, extracted with ethyl acetate (2 x 30 ml), the organic phase was suspended and the crude material was purified by column chromatography to give example 15-4 as a white solid (1.4 g,75% yield).
MS m/z(ESI):356.1[M+H] + .
And a second step of: preparation of 6-bromo-1- (4-chlorobenzyl) -3-ethyl-1, 7-dihydro-2H-pyrrolo [2,3-D ] pyrimidine-2, 4 (3H) -dione
Example 8-2 (1.28 g,3.6 mmol) was dissolved in DMF (15 mL) and NaH (0.17 g,7.18 mmol) was added to the mixture. The reaction was stirred at 0 ℃ for 1 hour under nitrogen atmosphere. CDI (0.87 g,5.38 mmol) was added and the reaction solution was heated to 75℃for 2 hours. The reaction solution was cooled to room temperature, water (80 mL) was added to terminate the reaction, and the filter cake was dried to give example 8-3 (1.07 g,78% yield).
MS m/z(ESI):382.0[M+H] + .
And a third step of: preparation of 1- (4-chlorobenzyl) -3-ethyl-6- (4-hydroxyphenyl) -1, 7-dihydro-2H-pyrrolo [2,3-D ] pyrimidine-2, 4 (3H) -dione
To a solution containing example 8-3 (1 g,2.6 mmol), parahydroxyphenylboronic acid (720 mg,5.2 mmol), pd (dppf) Cl at room temperature 2 A round bottom flask of (95 mg,0.13 mmol) and potassium carbonate (1.07 g,7.8 mmol) was charged with the solvent Dioxane (10 mL) and water (2 mL), and after displacing nitrogen, the mixture was heated to 80℃and stirred for 3 hours. The mixture was cooled to room temperature and concentrated, and purified by flash column chromatography to give example 8-4 (1.65 g,82% yield).
MS m/z(ESI):777.1[M+H] + .
Fourth step: preparation of 1- (4-chlorobenzyl) -6- (4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) -3-ethyl-1, 7-dihydro-2H-pyrrolo [2,3-D ] pyrimidine-2, 4 (3H) -dione
Examples 8-4 (100 mg,0.13 mmol) were dissolved in DMF (2 mL) at room temperature, and 2,4, 6-trifluoropyridine (26 mg,0.2 mmol) and cesium carbonate (100 mg,0.3 mmol) were added to the mixture. The reaction solution was heated to 80℃and reacted for 2 hours. The mixture was cooled to room temperature and concentrated, and purified by preparative HPLC to give example 8 (37 mg,56% yield).
MS m/z(ESI):509.0[M+H] + .
Example 9
(S) -N- (5-chloropyridin-2-yl) -2- (6- (1-methoxypropane-2-yl) -4, 7-dioxo-2- ((4- (pyridin-2-yloxy) phenyl) amino) -4,5,6, 7-tetrahydro-1H-pyrrolo [3,4-D ] pyrimidin-1-yl) acetamide
The first step: preparation of 2-amino-N- (5-fluoropyridin-2-yl) acetamide
Example 9-1 (1 g,4.29 mmol) and ammonium carbonate (3.3 g,34.33 mmol) were dissolved in acetonitrile (30 mL) and heated to 50deg.C. After 10 hours, a white solid was precipitated, and the precipitate was separated by filtration and recrystallized from toluene to obtain example 9-2 (0.6 g,83% yield).
MS m/z(ESI):170.2[M+H] + .
And a second step of: preparation of N- (5-fluoropyridin-2-yl) thiourea acetamide
Example 9-2 (0.5 g,2.96 mmol) and benzoyl isothiocyanate (0.53 g,3.25 mmol) were reacted in DCM (10 mL) for 2 hours at room temperature. After the reaction solution was dried by spinning, K2CO3 (817 mg,5.91 mmol) was added to the reaction mixture, and the resulting mixture was stirred under reflux overnight. The reaction was cooled to room temperature and concentrated in vacuo to give an orange solid. The material was washed with water (50 mL) to give the title compound example 9-3 (0.4 g,60% yield).
MS m/z(ESI):229.0[M+H] + .
And a third step of: preparation of (S) -N- (4-chlorophenyl) -2- (2-mercapto-6- (1-methoxypropane-2-yl) -4, 7-dioxo-4, 5,6, 7-tetrahydro-1H-pyrrolo [3,4-D ] pyrimidin-1-yl) acetamide
Example 9-3 (0.4 g,1.75 mmol) was dissolved in EtOH (20 mL), naOEt (143 mg,2.1 mmol) was added and ethyl (S) -4-hydroxy-1- (1-methoxypropane-2-yl) -5-oxo-2, 5-dihydro-1H-pyrrole-3-carboxylate (425 mg,1.75m mol) was stirred under reflux with heating for 5 hours. The reaction mixture was concentrated under reduced pressure, and the resulting residue was dissolved in dichloro (20 mL). The precipitated solid was filtered, and the precipitated solid was collected and then washed with diethyl ether to give example 10-3 (350 mg,46% yield).
MS m/z(ESI):423.1[M+H] + .
Fourth step: preparation of (S) -N- (4-chlorophenyl) -2- (6- (1-methoxypropane-2-yl) -2- (methylthio) -4, 7-dioxo-4, 5,6, 7-tetrahydro-1H-pyrrolo [3,4-D ] pyrimidin-1-yl) acetamide
Examples 9-4 (350 mg,0.8 mmol), methyl iodide (184 mg,0.88 mmol) and K 2 CO 3 (220 mg,1.6 mmol) DMF (10 mL) was thoroughly mixed in a 100mL round bottom flask and reacted for 4h at room temperature with a magnetic stirrer. Purification by silica gel column after completion of the reaction gave examples 9-5 (180 mg,50% yield).
MS m/z(ESI):437.0[M+H] + .
Fifth step: preparation of (S) -N- (5-chloropyridin-2-yl) -2- (6- (1-methoxypropane-2-yl) -4, 7-dioxo-2- ((4- (pyridin-2-yloxy) phenyl) amino) -4,5,6, 7-tetrahydro-1H-pyrrolo [3,4-D ] pyrimidin-1-yl) acetamide
Examples 9-5 (50 mg,0.11 mmol), 4- (pyridin-2-yloxy) aniline (50 mg,0.26 mmol), acOH (0.25 mL) and t-BuOH (2 mL) were stirred at 100deg.C for 2 hours. After cooling the reaction, it was concentrated and purified by preparative HPLC to give example 9 (23 mg,37% yield).
MS m/z(ESI):576.2[M+H] + .
Example 10
2- (2- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -6-isopropyl-4, 7-dioxo-4, 5,6, 7-tetrahydro-1H-pyrrolo [3,4-d ] pyrimidin-1-yl) -N- (5-fluoropyridin-2-yl) acetamide
The first step: preparation of 2-amino-N- (5-fluoropyridin-2-yl) acetamide
(1 g,4.29 mmol) 2-bromo-N- (5-fluoropyridin-2-yl) acetamide and (3.3 g,34.33 mmol) ammonium carbonate were dissolved in acetonitrile (30 mL) and heated to 50deg.C. After 10 hours, a white solid precipitated, and the precipitate was isolated by filtration and recrystallized from toluene to give example 10-1 (0.6 g, 83%) as a white solid.
MS m/z(ESI):170.23[M+H] + .
And a second step of: preparation of N- (5-fluoropyridin-2-yl) thiourea acetamide
2-amino-N- (5-fluoropyridin-2-yl) acetamide (0.5 g,2.96 mmol) and benzoyl isothiocyanate (0.53 g,3.25 mmol) were combined in CH 2 Cl 2 (10 mL) was reacted at room temperature for 2 hours. After the reaction solution was dried by spin drying, K was added to the reaction mixture 2 CO 3 (817 mg,5.91 mmol) and the resulting mixture was stirred at reflux overnight. The reaction was cooled to room temperature and concentrated in vacuo to give an orange solid. The material was washed with water (50 mL). The title compound example 10-2 (0.4 g,60% yield) was obtained as a yellow solid.
MS m/z(ESI):229.05[M+H] + .
And a third step of: preparation of N- (5-fluoropyridin-2-yl) -2- (6-isopropyl-2-mercapto-4, 7-dioxo-4, 5,6, 7-tetrahydro-1H-pyrrolo [3,4-d ] pyrimidin-1-yl) acetamide
Example 10-2 (0.4 g,1.75 mmol) was dissolved in EtOH (20 mL), naOEt (143 mg,2.1 mmol) was added, and 4-hydroxy-1-isopropyl-5-oxo-2, 5-dihydro-1H-pyrrole-3-carboxylic acid ethyl ester (264 mg,1.75 mmol) was stirred under reflux with heating for 5 hours. The reaction mixture was concentrated under reduced pressure, and the resulting residue was dissolved in dichloro (20 mL). The precipitated solid was filtered, and the precipitated solid was collected and then washed with diethyl ether to give example 10-3 (0.3 g, 45%).
MS m/z(ESI):378.13[M+H] + .
Fourth step: preparation of N- (5-fluoropyridin-2-yl) -2- (6-isopropyl-2- (methylsulfanyl) -4, 7-dioxo-4, 5,6, 7-tetrahydro-1H-pyrrolo [3,4-d ] pyrimidin-1-yl) acetamide
Example 10-3 (300 mg,0.8 mmol), methyl iodide (184 mg,0.88 mmol) and K 2 CO 3 (220 mg,1.6mmol, DMF (10 mL) was thoroughly mixed in a 100mL round bottom flask and placed in a magnetic stirrer for reaction at room temperature for 4h after completion of the reaction (TLC, DCM: meOH=10:1), example 10-4 (0.2 g, 51%) was obtained via climbing plates.
MS m/z(ESI):392.12[M+H] + .
Fifth step: preparation of 2- (2- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -6-isopropyl-4, 7-dioxo-4, 5,6, 7-tetrahydro-1H-pyrrolo [3,4-d ] pyrimidin-1-yl) -N- (5-fluoropyridin-2-yl) acetamide
Example 10-4 (50 mg,0.13 mmol) and 4- ((2, 6-difluoropyridin-4-yl) oxy) aniline (28.4 mg,0.13 mmol), cs 2 CO 3 (83.2 mg,0.26 mmol) in DMF (5 mL) was heated to 80℃and reacted for 2h, after completion of the reaction by LCMS detection, 10mL of water and 20mL of ethyl acetate were added, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was eluted with silica gel chromatography in dichloro methanol (0-10%) to give example 10 (20 mg, 28%).
MS m/z(ESI):566.17[M+H] + .
Example 11
N- (4- (4-chlorobenzyl) -2- (cyclopentylamine) -5-oxo-4, 5-dihydropyrazol [1,5-a ] pyrimidin-6-yl) tetrahydro-2H-pyran-4-carboxamide
/>
The first step: preparation of N- (4- (4-chlorobenzyl) -2- (cyclopentylamine) -5-oxo-4, 5-dihydropyrazol [1,5-a ] pyrimidin-6-yl) tetrahydro-2H-pyran-4-carboxamide
Example 11-1 (220 mg,0.47 mmol), ruPhos Pd G2 (35 mg,0.047 mmol) and RuPhos ((21 mg,0.047 mmol)), liHMDS (1M in THF) (2.35 mL,2.35 mmol) were dissolved in anhydrous dioxane (5 mL), followed by addition of lithium chloride (198 mg,4.7 mmol), cyclopentylamine (80 mg,0.94 mmol). The reaction solution was stirred for 2 hours at 100℃with microwaves. Saturated NaCl solution was added and extracted three times with ethyl acetate. The organic phase was washed with brine, dried and concentrated, and purified by column to give example 11 (20 mg,9% yield).
MS m/z(ESI):470.1[M+H] + .
Example 12
3- (4-chlorobenzyl) -6-ethyl-2- ((4- (isoxazol-3-yloxy) phenyl) amino) -5, 6-dihydro-3H-pyrrolo [3,4-d ] pyrimidine-4, 7-dione
The first step: preparation of 3-chloro-1-isopropyl-4, 5-dioxopyrrolidine-3-carboxylic acid ethyl ester
The compound 1-isopropyl-4, 5-dioxopyrrolidine-3-carboxylic acid ethyl ester (4 g,18.7 mmol) was suspended in a mixed solution of acetic acid (5 mL) and water (5 mL), cooled to 0deg.C, and sodium hypochlorite (30 mL,21mmol, 0.7M) was added dropwise. The mixture was slowly warmed to room temperature and reacted for 1h. Excess sodium chloride was added, extracted with dichloromethane (30 ml x 5), and the combined organic phases dried over anhydrous sodium sulfate and spun dry under high vacuum. Diethyl ether (20 mL) was added and stirred for 10min, filtered, the filter cake was washed with diethyl ether and dried to give example 12-1 (3.7 g, 75%) as a white solid.
MS m/z(ESI):248.1[M+H] + .
1 HNMR:(400MHz,CDCl3)δ4.67-4.61(m,1H),4.38-4.28(m,3H),3.77(d,J=11.5Hz,1H),1.37-1.27(m,9H).
And a second step of: preparation of N- (6-isopropyl-4, 7-dioxo-4, 5,6, 7-tetrahydro-3H-pyrrolo [3,4-D ] pyrimidin-2-yl) acetamide
Acetylguanidine (2.54 g,25.2 mmol) was dissolved in ethanol (15 mL), cooled to 0deg.C, 3-chloro-1-isopropyl-4, 5-dioxopyrrolidine-3-carboxylic acid ethyl ester (2.5 g,10.1 mmol) and 4A molecular sieve (3 g) were added and reacted for 2h with stirring. Then, a freshly prepared chromium chloride solution (126 mmol) was added dropwise, and after the completion of the addition, the ice bath was removed, and the reaction was stirred at room temperature for 3 hours. Water (100 mL) was added for dilution, dichloromethane extraction (50 mL. Times.4), the combined organic phases dried over anhydrous sodium sulfate, and spin-dried to afford example 12-3 (1.8 g, 71%) as a yellow solid by column chromatography (methanol/dichloromethane=0-4%).
MS m/z(ESI):251.1[M+H] + .
1 HNMR:(400MHz,CDCl 3 )δ10.8(s,1H),4.65–4.61(m,1H),4.12(s,2H),1.28(d,J=6.8Hz,6H).
And a third step of: preparation of N- (3- (4-chlorobenzyl) -6-ethyl-4, 7-dioxo-4, 5,6, 7-tetrahydro-3H-pyrrolo [3,4-d ] pyrimidin-2-yl) acetamide
N- (6-isopropyl-4, 7-dioxo-4, 5,6, 7-tetrahydro-3H-pyrrolo [3,4-D ] pyrimidin-2-yl) acetamide (1.8 g,7.2 mmol) was dissolved in N, N-dimethylformamide (30 mL), cooled to 0deg.C, sodium hydride (432mg,10.8mmol,60%in oil) was added in portions, stirred for 30min, 4-chlorobenzyl bromide (1.77 g,8.64 mmol) was added, and the mixture was slowly warmed to room temperature for 4H. The reaction mixture was quenched with 100mL of water, extracted with ethyl acetate (60 mL x 3), the combined organic phases were washed with water (50 mL x 2), saturated brine (50 mL), dried over anhydrous sodium sulfate, and dried by spin-drying, and column chromatography (methanol/dichloromethane=0 to 5%) gave example 12-4 (1.1 g, 42%) as a yellow solid.
MS m/z(ESI):375.1[M+H] + .
1 HNMR:(400MHz,CDCl 3 )δ7.42(d,J=8.4Hz,2H),7.13(d,J=8.4Hz,2H),5.76(s,2H),4.65–4.61(m,1H),4.12(s,2H),1.28(d,J=6.8Hz,6H).
Fourth step: preparation of 2-amino-3- (4-chlorobenzyl) -6-isopropyl-5, 6-dihydro-3H-pyrrolo [3,4-d ] pyrimidine-4, 7-dione hydrochloride
N- (3- (4-chlorobenzyl) -6-ethyl-4, 7-dioxo-4, 5,6, 7-tetrahydro-3H-pyrrolo [3,4-d ] pyrimidin-2-yl) acetamide (1.1 g,2.94 mmol) was dissolved in 4M hydrochloric acid and reacted at 110℃under reflux for 4H. The reaction was dried by spin-drying to give example 12-5 (1.0 g, 100%).
MS m/z(ESI):333.1[M+H] + .
1 HNMR:(400MHz,CDCl 3 )δ10.8(s,1H),4.65–4.61(m,1H),4.12(s,2H),1.28(d,J=6.8Hz,6H).
Fifth step: preparation of 3- (4-chlorobenzyl) -2- ((4-hydroxyphenyl) amino) -6-isopropyl-5, 6-dihydro-3H-pyrrolo [3,4-D ] pyrimidine-4, 7-dione
2-amino-3- (4-chlorobenzyl) -6-isopropyl-5, 6-dihydro-3H-pyrrolo [3,4-d ] pyrimidine-4, 7-dione hydrochloride (200 mg,0.601 mmol) was dissolved in 1, 4-dioxane (6 mL), 4-bromophenol (208 mg,1.20 mmol), palladium acetate (7 mg,0.03 mmol), 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (17 mg,0.03 mmol) and cesium carbonate (293 mg,0.901 mmol) were added and reacted at 100℃for 2H under microwave. Cooled to room temperature, filtered, and the filtrate was dried by spin-drying and column chromatography (methanol/dichloromethane=0 to 10%) to give example 12-6 (85 mg, 34%).
MS m/z(ESI):411.1[M+H] + .
Sixth step: preparation of 3- (4-chlorobenzyl) -6-ethyl-2- ((4- (isoxazol-3-yloxy) phenyl) amino) -5, 6-dihydro-3H-pyrrolo [3,4-D ] pyrimidine-4, 7-dione
Referring to examples 12-6, starting from 3- (4-chlorobenzyl) -2- ((4-hydroxyphenyl) amino) -6-isopropyl-5, 6-dihydro-3H-pyrrolo [3,4-D ] pyrimidine-4, 7-dione (50 mg,0.122 mmol) and 3-bromoisoxazole (36 mg,0.243 mmol), example 12 (11 mg, 20%) was obtained.
MS m/z(ESI):478.1[M+H] + .
Example 13
4- (4-chlorobenzyl) -7-oxo-2- (tetrahydro-2H-pyran-4-yl) -N- (2, 2-trifluoromethyl) -4, 7-dihydropyrazolo [1,5-a ] pyrimidine-6-carboxamide
The first step: preparation of N- (4-chlorobenzyl) -2-fluoro-4-iodocarboxamide
2-fluoro-4-iodonicotinic acid (5 g,18.7 mmol) was dissolved in N, N-dimethylformamide (30 mL), O- (7-azobenzotriazole-1-oxy) -N, N' -tetramethylurea hexafluorophosphate (7.8 g,20.6 mmol) was added, and stirred for 15min, p-4-chlorobenzylamine (2.65 g,18.7 mmol) was added and stirred at room temperature for 2h. Poured into water (100 mL), extracted with ethyl acetate (60 mL x 3), the combined organic phases washed with water (60 mL), saturated brine (60 mL), dried over anhydrous sodium sulfate, spin-dried and column chromatographed (methanol/dichloromethane=0-5%) to give example 13-2 (6.6 g, 91%).
MS m/z(ESI):391.0[M+H] + .
And a second step of: preparation of 2-amino-N- (4-chlorobenzyl) -4-iodoformamide
N- (4-chlorobenzyl) -2-fluoro-4-iodoformamide (1.5 g,16.9 mmol) was dissolved in dimethyl sulfoxide (10 mL), aqueous ammonia (2 mL) was added, and the mixture was reacted at 110℃for 3h under microwave. The reaction mixture was poured into water (40 mL), extracted with ethyl acetate (30 ml×3), washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and spin-dried, and column chromatographed (methanol/dichloromethane=0 to 10%) to give example 13-3 (1.3 g, 87%).
MS m/z(ESI):388.0[M+H] + .
And a third step of: preparation of N- (4-chlorobenzyl) -4-iodo-2- ((triphenyl-l 5-phosphono) amino) nicotinamide
2-amino-N- (4-chlorobenzyl) -4-iodoformamide (1.3 g,3.36 mmol) was dissolved in toluene (20 mL), triphenylphosphine (0.88 g,3.36 mmol) and hexachloroethane (0.795 g,3.36 mmol) were added and reacted under reflux under nitrogen for 2h. Cooled to room temperature, filtered, and the filtrate was dried by spin-drying, followed by column chromatography (ethyl acetate/petroleum ether=50% -100%) to give example 13-4 (1.8 g, 83%).
MS m/z(ESI):648.0[M+H] + .
Fourth step: preparation of 3- (4-chlorobenzyl) -5-iodo-2- ((4-methoxyphenyl) amino) pyridin [2,3-d ] pyrimidin-4 (3 h) -one
N- (4-chlorobenzyl) -4-iodo-2- ((triphenyl-l 5-phosphono) amino) nicotinamide (1.8 g,2.78 mmol) was dissolved in xylene (50 mL), p-methoxyisocyanate (1.24 g,8.33 mmol) was added and reacted at 150℃under reflux for 3h. Cooled to room temperature, dried by spin-drying, and subjected to column chromatography (methanol/dichloromethane=0 to 10%) to give example 13-5 (920 mg, 64%).
MS m/z(ESI):519.0[M+H] + .
Fifth step: preparation of 3- (4-chlorobenzyl) -2- ((4-methoxyphenyl) amino) -5- ((tetrahydro-2H-pyran-4-yl) amino) pyridin [2,3-D ] pyrimidin-4 (3H) -one
Referring to examples 12-7, examples 13-6 (162 mg, 57%) were obtained starting from 3- (4-chlorobenzyl) -2- ((4-methoxyphenyl) amino) -5- ((tetrahydro-2H-pyran-4-yl) amino) pyridin [2,3-D ] pyrimidin-4 (3H) -one (300 mg,0.579 mmol) and 4-aminotetrahydropyran (88 mg,0.869 mmol), respectively.
MS m/z(ESI):492.2[M+H] + .
Sixth step: preparation of 3- (4-chlorobenzyl) -2- ((4-methoxyphenyl) amino) -5- ((tetrahydro-2H-pyran-4-yl) amino) pyridin [2,3-D ] pyrimidin-4 (3H) -one
3- (4-chlorobenzyl) -2- ((4-methoxyphenyl) amino) -5- ((tetrahydro-2H-pyran-4-yl) amino) pyridin [2,3-D ] pyrimidin-4 (3H) -one (162 mg,0.330 mmol) was dissolved in dichloromethane (10 mL), cooled to 0℃and a solution of boron tribromide in dichloromethane (1 mL,1mmol, 1M) was added dropwise and the mixture was allowed to react slowly to room temperature for 1H. Poured into an ice water solution of sodium bicarbonate (50 mL), extracted with dichloromethane (30 mL x 2), the combined organic phases were washed with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, spin-dried and column chromatographed (methanol/dichloromethane=0-10%), giving examples 13-7 (90 mg, 58%).
MS m/z(ESI):478.2[M+H] + .
Seventh step: preparation of 3- (4-chlorobenzyl) -2- ((4- (isoxazol-3-oxy) phenyl) amino) -5- ((tetrahydro-2H-pyran-4-yl) amino) pyridin [2,3-D ] pyrimidin-4 (3H) -one
Referring to examples 12-5, starting with 3- (4-chlorobenzyl) -2- ((4-methoxyphenyl) amino) -5- ((tetrahydro-2H-pyran-4-yl) amino) pyridin [2,3-D ] pyrimidin-4 (3H) -one (30 mg,0.063 mmol) gave example 13 (17 mg, 50%).
MS m/z(ESI):545.2[M+H] + .
Example 14
4- (4-chlorobenzyl) -6- ((2, 6-difluoropyridin-4-yl) oxy) -1-ethyl-1, 4-dihydro-9 h-imidazo [4,5-b ] [1,8] naphthyridin-9-one
The first step: preparation of 1-ethyl-4-nitroimidazole
Example 14-1 (1.13 g,10 mmol) and DMF (10 mL) were added to a round bottom flask, potassium carbonate (2.07 g,15 mmol) and ethyl iodide (2.34 g,15 mmol) were added. The mixture was added to hot 80℃and the reaction was stirred for 2 hours. After the mixture was cooled to room temperature, water (50 ml) was added and filtered to give example 14-2 (1.1 g, 78%) as a yellow solid.
MS m/z(ESI):142.1[M+H] + .
And a second step of: preparation of 1-ethyl-4-aminoimidazole
Example 14-2 (1.1 g,7.8 mmol), methanol (20 mL) and palladium on carbon (200 mg) were added to a round bottom flask and the mixture was stirred at room temperature for 12 hours. After the reaction was completed, the mixture was filtered and concentrated to give example 14-3 (830 mg, 96%) as a white solid.
MS m/z(ESI):112.2[M+H] + .
And a third step of: preparation of 6-chloro-2- ((1-ethyl-1H-imidazol-4-yl) amino) nicotinic acid
Example 14-3 (830 mg,7.5 mmol) was dissolved in THF (15 ml) and lithium bis (trimethylsilyl) amide (1M, 11 mL) was added dropwise under nitrogen at-75℃and stirred at this temperature for 1 hour. A solution of 2, 6-dichloronicotinic acid (720 mg,3.8 mmol) in THF (6 ml) was then added dropwise to the mixture, and the mixture was stirred at room temperature for 12 hours. After the reaction was completed, it was quenched with water, then cooled in an ice bath, and acidified to pH 2-3 with 5N HCl solution. The aqueous phase was then extracted multiple times with ethyl acetate, the organic phase was dried and then concentrated to give example 14-4 (608 mg, 61%) as a yellow solid.
MS m/z(ESI):267.1[M+H] + .
Fourth step: preparation of 6-chloro-1-ethyl-1, 4-dihydro-9H-imidazo [4,5-B ] [1,8] naphthyridin-9-one
Examples 14-4 (608 mg,2.3 mmol) and (8 mL) were added to a round bottom flask and the mixture was stirred at 120deg.C for 5 hours. After the completion of the reaction, it was quenched with water and filtered to give example 14-5 (319 mg, 56%) as a pale yellow solid.
MS m/z(ESI):249.1[M+H] + .
Fifth step: preparation of 6-chloro-4- (4-chlorobenzyl) -1-ethyl-1, 4-dihydro-9H-imidazo [4,5-B ] [1,8] naphthyridin-9-one
Examples 14-5 (319 mg,1.3 mmol) and DMF (5 mL) were added to a round bottom flask, potassium carbonate (418 mg,3 mmol) and p-chlorobenzyl bromide (612 mg,3 mmol) were added. The mixture was added to heat at 60℃and the reaction was stirred for 2 hours. After the mixture was cooled to room temperature, water (30 ml) was added and filtered to give example 14-6 (435 mg, 78%) as a white solid.
MS m/z(ESI):373.2[M+H] + .
Sixth step: preparation of 4- (4-chlorobenzyl) -6- ((2, 6-difluoropyridin-4-yl) oxy) -1-ethyl-1, 4-dihydro-9 h-imidazo [4,5-b ] [1,8] naphthyridin-9-one
Examples 14-6 (100 mg,0.27 mmol) and DMF (2 mL) were added to a round bottom flask, potassium carbonate (418 mg,3 mmol) and 2, 6-difluoropyridin-4-ol (35 mg,0.27 mmol) were added. The mixture was heated to 100deg.C and stirred for 2 hours. The mixture was cooled to room temperature and filtered and purified by preparative HPLC to give example 14 (45 mg, 36%) as a white solid.
MS m/z(ESI):468.1[M+H] + .
Example 15
4- (4-chlorobenzyl) -5- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -1-ethyl-1, 4-dihydro-7H-pyrazolo [4,3-d ] pyrimidin-7-one
The first step: preparation of 4-nitro-1H-pyrazole-3-carboxamide
The compound 4-nitro-1H-pyrazole-3-carboxylic acid (15.7 g,99.95 mmol) was placed in a 500mL three-necked round bottom flask, followed by addition of THF (250 mL) and oxalyl chloride (13.9 g,109.51 mmol) in DMF as a drop. The reaction was stirred at 25℃for 0.5 h. The reaction was then cooled in an ice bath, aqueous ammonium hydroxide (36 ml,5.00 eq.) was added and the reaction was stirred at 25 ℃ for 2 hours. The resulting mixture was concentrated in vacuo. The crude material was treated with H 2 O-washing gave example 15-1 (15 g, 93%) as a white solid.
MS m/z(ESI):157.03[M+H] + .
1 H NMR(400MHz,DMSO-d6)δ14.10(s,6H),8.72(s,6H),8.03(d,J=33.5Hz,8H),7.77(d,J=58.7Hz,8H),1.25(d,J=8.9Hz,1H).
And a second step of: preparation of 1-ethyl-4-nitro-1H-pyrazole-5-carboxamide
Example 15-1 (5.0 g,32.03mmol,1.00 eq.) was placed in a 500mL 3-neck round bottom flask. Then K is added 2 CO 3 (8.8 g,63.2mmol,2.0 eq.) DMF (150 mL) and iodoethane (10 g,64.12mmol,2.00 eq.). The reaction was stirred at 25℃for 5 hours. The resulting solution was treated with H 2 O was diluted and extracted with EtOAc. The organic layers were combined, washed with brine, dried over anhydrous Na 2 SO 4 Dried and concentrated in vacuo. The crude material was purified by column chromatography to give example 15-2 as a white solid (1.8 g, 31%).
MS m/z(ESI):185.06[M+H] + .
1 H NMR(400MHz,DMSO-d6)δ8.50(s,1H),8.30(d,J=9.3Hz,2H),4.16(q,J=7.2Hz,2H),1.37(t,J=7.2Hz,3H).
And a third step of: preparation of 4-amino-1-ethyl-1H-pyrazole-5-carboxamide
Example 15-2 (1.5 g,8.15mmol,1.00 eq.) was placed in a 100mL round bottom flask and dissolved in MeOH (20 mL). Pd-C (0.15 g) was then added and the resulting suspension stirred under a hydrogen atmosphere at 25℃for 3 hours. The solid was filtered off. The resulting mixture was concentrated in vacuo to afford example 15-3 (0.9 g, 72%) as a violet solid.
MS m/z(ESI):155.10[M+H] + .
Fourth step: preparation of 4- ((4-chlorophenyl) amino) -1-ethyl-1H-pyrazole-5-carboxamide
Example 15-3 (0.8 g,5.19 mmol) was dissolved in methanol (15 ml) at room temperature, 4-chlorobenzaldehyde (0.73 g,5.19 mmol) was added to the mixture, and stirred at room temperature for 5 hours, and NaBH was added to the reaction mixture 4 (0.4 g,10.38 mmol). The mixture was added to heat at 50℃and the reaction was stirred for 2h. After cooling, 20ml of water was added, extracted with ethyl acetate (2 x 30 ml), the organic phase was suspended and the crude material was purified by column chromatography to give example 15-4 as a white solid (1 g, 69.4%).
MS m/z(ESI):279.09[M+H] + .
Fifth step: preparation of 4- (4-chlorobenzyl) -1-ethyl-1, 4-dihydro-5H-pyrazolo [4,3-d ] pyrimidine-5, 7 (6H) -dione
Examples 15-4 (1 g,3.59 mmol) were dissolved in DMF (15 ml) and NaH (0.17 g,7.18 mmol) was added to the mixture. The reaction was stirred at 0 ℃ for 1 hour under nitrogen atmosphere. Bis (1H-imidazol-1-yl) methane thioketone (0.89 g,5.38 mmol) was added, and the reaction solution was heated to 75℃for 2 hours. The reaction solution was cooled to room temperature, water (80 mL) was added to terminate the reaction, and the filter cake was dried to give example 15-5 (0.9 g, 81%) as a white powder.
MS m/z(ESI):321.07[M+H] + .
Sixth step: preparation of 5-chloro-4- (4-chlorobenzyl) -1-ethyl-1, 4-dihydro-7H-pyrazolo [4,3-d ] pyrimidin-7-one
Examples 15-5 (0.9 g,2.81 mmol) were dissolved in DMF (6 ml) and methyl iodide (0.8 g,5.61 mmol) was added and reacted at room temperature for 2h. The reaction mixture was added to water (20 ml), and the insoluble matter was filtered off to give example 15-6 (0.8 g, 85%) as a white powder.
MS m/z(ESI):335.04[M+H] + .
Seventh step: preparation of 4- (4-chlorobenzyl) -1-ethyl-5- ((4-hydroxyphenyl) amino) -1, 4-dihydro-7H-pyrazolo [4,3-d ] pyrimidin-7-one
Examples 15-6 (30 mg,0.09 mmol) were dissolved in t-BuOH (2 mL), CH at room temperature 3 COOH (0.5 mL) to the solution was added 4-hydroxyaniline (30 mg,0.27 mmol). The reaction solution was heated to 100℃and reacted for 6 hours. To the reaction was added 10ml of water, the mixture was extracted with EtOAc (2 x 10 ml) and the combined extracts were taken over Na 2 SO 4 Drying, spin-drying, and p-TLC (dichloro: methanol=20:1) gave examples 15-7 (20 mg, 56%) as a white powder.
MS m/z(ESI):396.11[M+H] + .
Eighth step: preparation of 4- (4-chlorobenzyl) -5- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -1-ethyl-1, 4-dihydro-7H-pyrazolo [4,3-d ] pyrimidin-7-one
Examples 15-7 (30 mg,0.075 mmol) were dissolved in DMF (1 mL) at room temperature, and 2,4, 6-trifluoropyridine (15 mg,0.114 mmol) and cesium carbonate (49 mg,0.15 mmol) were added to the mixture. The reaction solution was heated to 80℃and reacted for 2 hours. Addition of 10 to the reaction mass The mixture was extracted with EtOAc (2 x 10 ml) and the combined extracts were taken up in Na 2 SO 4 Drying, spin-drying, crude product using p-TLC (dichloro: methanol=20:1) gave example 15 (8 mg, 21%) as a white powder.
MS m/z(ESI):509.12[M+H] + .
Example 16
4- (4-chlorobenzyl) -5- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -1-ethyl-1, 4-dihydro-7 h- [1,2,3] triazolo [4,5-d ] pyrimidin-7-one
The first step: preparation of N- ((4-chlorobenzyl) carbamic acid thio) benzamide
Example 16-1 (1.63 g,10 mmol) was dissolved in THF, cooled to 0deg.C and p-chlorobenzylamine (1.55 g,11 mmol) was added dropwise. After the mixture was stirred at room temperature for 2 hours, the reaction solution was concentrated, and the solid was slurried with petroleum ether and filtered to give example 16-2 (2.7 g, 90%) as a white solid.
And a second step of: preparation of 1- (4-chlorobenzyl) thiourea
Example 16-2 (2.7 g,9 mmol) was dissolved in ethanol (20 mL) and water (20 mL) at room temperature, and potassium carbonate (2.5 g,18 mmol) was added. The mixture was added to hot 100℃and the reaction stirred for 3 hours. After the mixture was cooled to room temperature, ethanol was removed by concentration and filtration, and the solid was washed with water to give example 16-3 (1.7 g, 94%) as a white solid.
MS m/z(ESI):201.1[M+H] + .
And a third step of: preparation of 6-amino-1- (4-chlorobenzyl) -2-mercapto-5-nitrosopyrimidin-4 (1H) -one
To a sodium ethoxide solution prepared from sodium (460 mg,20 mmol) and 10ml ethanol were added example 16-3 (1.2 g,6 mmol) and ethyl cyanoacetate 2-oxime (1.4 g,10 mmol). The orange mixture was stirred under reflux for 2 hours. The mixture was cooled to 0deg.C, water (15 mL) and 1N HCl (20 mL) were added, the mixture was filtered and the solid was washed three times with water to give example 16-4 (1.2 g, 63%) as a cyan solid.
MS m/z(ESI):297.1[M+H] + .
Fourth step: preparation of (3- (4-chlorobenzyl) -2-mercapto-5-nitroso-6-oxo-3, 6-dihydropyrimidin-4-yl) carbamic acid tert-butyl ester
To a solution of example 16-4 (1 g,3.37 mmol) in DCM (20 mL) was added (Boc) 2 O (0.88 g,4.04 mmol) and triethylamine (0.68 g,6.74 mmol) were added and the mixture was stirred at room temperature for 3h. DCM was removed in vacuo and ethyl acetate was added to the crude product. The product was filtered off and washed with ethyl acetate to give example 16-5 (1.1 g, 82%).
MS m/z(ESI):397.07[M+H] + .
Fifth step: preparation of (5-amino-3- (4-chlorobenzyl) -2-mercapto-6-oxo-3, 6-dihydropyrimidin-4-yl) carbamic acid tert-butyl ester
Examples 16-5 (1 g,2.52 mmol) were added to dioxane (20 mL) and water (5 mL), followed by Na 2 S 2 O 4 (877 mg,5.04 mmol). After stirring the reaction mixture at room temperature for 2 hours. Water (20 mL) was added, extracted with ethyl acetate (20 mL. Times.2), the organic phases combined, dried over anhydrous Na 2 SO 4 Drying and concentration gave example 16-6 (850 mg,88%)。
MS m/z(ESI):383.10[M+H] + .
sixth step: preparation of tert-butyl (3- (4-chlorobenzyl) -5- (ethylamino) -2-mercapto-6-oxo-3, 6-dihydropyrimidin-4-yl) carbamate
Examples 16-6 (1 g,2.61 mmol) were placed in a 100mL round bottom flask and dissolved in MeOH (20 mL). Then acetaldehyde (0.13 g,2.87 mmol) was added and stirred for 5 minutes before NaBH was added 3 CN (0.25 g,3.92 mmol) was stirred at room temperature for 3 hours. Water (20 mL) was added, extracted with ethyl acetate (20 mL. Times.2), the organic phases were combined, dried and concentrated, and the crude product was purified by column on silica gel to give examples 16-7 (800 mg, 75%).
MS m/z(ESI):411.12[M+H] + .
Seventh step: preparation of 6-amino-1- (4-chlorobenzyl) -5- (ethylamino) -2-mercaptopyrimidin-4-one
Dioxane (10 ml, 6M) of example 16-7 (0.8 g,1.95 mmol) dissolved in HCl was stirred at room temperature for 1 hour and suspended directly to give example 16-8 (0.58 g, 95%) as a white solid.
MS m/z(ESI):311.09[M+H] + .
Eighth step: preparation of 4- (4-chlorobenzyl) -1-ethyl-5-mercapto-1, 4-dihydro-7H- [1,2,3] triazolo [4,5-d ] pyrimidin-7-one
Examples 16-8 (0.5 g,1.61 mmol) were dissolved in concentrated HCl (5 ml) and NaNO2 (0.17 g,2.41 mmol) was added to the mixture. The reaction was stirred at 0 ℃ for 2 hours under nitrogen. Addition of NaHCO 3 Ph=7, filtered,the filter cake was dried to give example 16-9 (0.3 g, 58%) as a white powder.
MS m/z(ESI):322.07[M+H] + .
Ninth step: preparation of 4- (4-chlorobenzyl) -1-ethyl-5- (methylthio) -1, 4-dihydro-7H- [1,2,3] triazolo [4,5-d ] pyrimidin-7-one
Examples 16-9 (0.2 g,0.62 mmol) were dissolved in DMF (6 ml) and water (1 ml) at room temperature, naOH (106 mg,0.75 mmol) and MeI (30 mg,0.75 mmol) were added and the reaction stirred at room temperature for 2h. Water (20 mL) was added, extracted with ethyl acetate (2X 40 mL), the combined organic phases dried and concentrated, and the crude product was purified by column on silica gel to give examples 16-10 (120 mg, 57%).
MS m/z(ESI):336.06[M+H] + .
Tenth step: preparation of 4- (4-chlorobenzyl) -5- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -1-ethyl-1, 4-dihydro-7H- [1,2,3] triazolo [4,5-d ] pyrimidin-7-one
Examples 16-10 (30 mg,0.089 mmol) were dissolved in t-butanol (3 mL) and acetic acid (1 mL) at room temperature, and 4- ((2, 6-difluoropyridin-4-yl) oxy) aniline (20 mg,0.089 mmol) was added to the mixture. The reaction solution was heated to 100℃and reacted for 16 hours. To the reaction was added 10ml of water, the mixture was extracted with EtOAc (2 x 10 ml) and the combined extracts were taken over Na 2 SO 4 Drying, spin-drying, crude product was purified by p-TLC (dichloro: methanol=20:1) to give example 16 (10 mg, 22%) as a white powder.
MS m/z(ESI):510.12[M+H] + .
Example 17
7- (4-chlorobenzyl) -6- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -3-ethylisoxazol [5,4-d ] pyrimidin-4 (7 h) -one
The first step: preparation of 2- (1-ethoxypropyl) malononitrile
To malononitrile (1 g,15 mmol) was added example 17-1 (2.7 g,15 mmol) and the mixture was stirred at 90℃for 1.5 h. After cooling to room temperature, concentration gave example 17-2 (2 g, 91%) as a yellow oil.
And a second step of: preparation of 3-ethyl-4-cyano-5-aminoisoxazole
A solution of example 17-2 (2 g,13.3 mmol) in ethanol (10 mL) was added to a mixture of hydroxylamine hydrochloride (1.39 g,20 mmol), naOH (800 mg,20 mmol) and water (10 mL), and the reaction was stirred at room temperature for 6 hours. After completion of the reaction, the mixture was concentrated and washed with water to obtain example 17-3 (1.3 g, 73%) as a white solid.
MS m/z(ESI):138.0[M+H] + .
And a third step of: preparation of 5-amino-3-ethylisoxazole-4-carboxamide
Example 17-3 (1.3 g,9.5 mmol) and concentrated sulfuric acid (8 mL) were added to a round bottom flask and the mixture was stirred at room temperature for 5 hours. After the completion of the reaction, the reaction mixture was poured into ice water (30 mL) and filtered to give example 17-4 (1.3 g, 90%) as a white solid.
MS m/z(ESI):156.1[M+H] + .
Fourth step: preparation of 5- ((4-chlorobenzyl) amino) -3-ethylisoxazole-4-carboxamide
Examples 17-4 (1.3 g,8.3 mmol), p-chlorobenzaldehyde (1.16 g,8.3 mmol) and methanol (15 mL) were added to a round bottom flask, acetic acid (1 mL) was added dropwise, sodium cyanoborohydride (1.04 g,16.6 mmol) was added in portions, and the mixture was stirred at room temperature for 2 hours. After the reaction was completed, it was quenched with water, then the aqueous phase was extracted with ethyl acetate several times, and after drying the organic phase was concentrated, it was purified by flash column chromatography to give example 17-5 (1.8 g, 80%) as pale yellow solid.
MS m/z(ESI):280.3[M+H] + .
Fifth step: preparation of 7- (4-chlorobenzyl) -3-ethylisoxazole [5,4-d ] pyrimidine-4, 6 (5 h,7 h) -dione
Examples 17-5 (1.4 g,5 mmol) and DCM (30 mL) were added to a round bottom flask and CDI (810 mg,5 mmol) and p-chlorobenzyl bromide (612 mg,3 mmol) were added. The mixture was stirred at room temperature for 5 hours. After the reaction was completed, the mixture was concentrated and purified by flash silica gel column to give example 17-6 (1.1 g, 73%) as a white solid.
MS m/z(ESI):306.0[M+H] + .
Sixth step: preparation of 4-chloro-7- (4-chlorobenzyl) -3-ethylisoxazole [5,4-d ] pyrimidin-6 (7 h) -one
Examples 17-6 (1.1 g,3.6 mmol) and phosphorus oxychloride (10 mL) were added to a round bottom flask. The mixture was added to heat 110℃and the reaction was stirred for 2 hours. The mixture was cooled to room temperature and concentrated to give example 17-7 (1.1 g, 95%) as an oil.
MS m/z(ESI):324.0[M+H] + .
Seventh step: preparation of 7- (4-chlorobenzyl) -6- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -3-ethylisoxazol [5,4-d ] pyrimidin-4 (7 h) -one
Examples 17-7 (100 mg,0.3 mmol), 4- ((2, 6-difluoropyridin-4-yl) oxy) aniline (23 mg,0.1 mmol) and ethanol (2 mL) were added to a round bottom flask. The mixture was heated to 100deg.C and stirred for 2 hours. The mixture was cooled to room temperature and concentrated, and purified by preparative HPLC to give example 17 (15 mg, 30%) as a white solid.
MS m/z(ESI):510.1[M+H] + .
Example 18
7- (4-chlorobenzyl) -6- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -3-ethylisoxazole [3,4-d ] pyrimidine-4 (7 h) -1
Example 18 Synthesis procedure, as reference to the synthesis of example 17, substituting 3-amino-5-ethylisoxazole-4-carbonitrile for example 17-3, gave example 18 via a 5-step reaction.
MS m/z(ESI):510.2[M+H] + .
Example 19
N- (3- (4-chlorobenzyl) -7-ethyl-6-oxo-6, 7-dihydro-3H-purin-2-yl) -4- ((2, 6-difluoropyridin-4-yl) oxy) benzamide
The first step: preparation of 5, 6-diamino-1- (4-chlorobenzyl) -2-mercaptopyrimidin-4 (1H) -one
Example 16-4 (600 mg,2 mmol) was dissolved in Dioxane (8 mL) and water (8 mL) and added1N aqueous NaOH solution (8 mL) and Na 2 S 2 O 4 (1.2 g,7 mmol). The mixture was stirred at room temperature for 1 hour, and filtered to give example 19-1 (450 mg, 80%) as a yellow solid.
MS m/z(ESI):283.2[M+H] + .
And a second step of: preparation of 3- (4-chlorobenzyl) -2-mercapto-3, 7-dihydro-6 h-purin-6-one
Example 19-1 (450 mg,1.6 mmol) and formic acid (6 mL) were added to a round bottom flask and the mixture was heated to 100deg.C and allowed to react for 5 hours with stirring. After the mixture was cooled to room temperature, it was filtered to give example 19-2 (400 mg, 85%) as a pale yellow solid.
MS m/z(ESI):293.1[M+H] + .
And a third step of: preparation of 3- (4-chlorobenzyl) -2- (methylthio) -3, 7-dihydro-6 h-purin-6-one
/>
A mixture of example 19-2 (292 mg,1 mmol), sodium hydroxide (40 mg,1 mmol), methyl iodide (0.12 mL,2 mmol) and water (5 mL) was stirred at room temperature overnight and then filtered to give example 19-3 (245 mg, 80%) as a yellow solid.
MS m/z(ESI):307.2[M+H] + .
Fourth step: preparation of 3- (4-chlorobenzyl) -7-ethyl-2- (methylthio) -3, 7-dihydro-6 h-purin-6-one
Example 19-3 (150 mg,0.5 mmol) and DMF (5 mL) were added to a round bottom flask, potassium carbonate (276 mg,2 mmol) and ethyl iodide (312 mg,2 mmol) were added. The mixture was added to hot 80℃and the reaction was stirred for 2 hours. After the mixture was cooled to room temperature, water (10 ml) was added and filtered to give example 19-4 (108 mg, 65%) as a white solid.
MS m/z(ESI):334.1[M+H] + .
Fifth step: preparation of 2-amino-3- (4-chlorobenzyl) -7-ethyl-3, 7-dihydro-6 h-purin-6-one
Examples 19-4 (100 mg,0.3 mmol) and aminomethyl alcohol (6 mL) were added to a round bottom flask and the mixture was heated to 80 ℃ and stirred for 12 hours. After the mixture was cooled to room temperature, it was concentrated to give example 19-5 (90 mg, 95%) as a pale yellow solid.
MS m/z(ESI):304.3[M+H] + .
Sixth step: preparation of N- (3- (4-chlorobenzyl) -7-ethyl-6-oxo-6, 7-dihydro-3H-purin-2-yl) -4-hydroxybenzoamide
Examples 19-5 (41 mg,0.3 mmol) and HATU (114 mg,0.3 mmol) were dissolved in N, N-dimethylformamide (2 mL), DIPEA (129 mg,1 mmol) was added to the above mixture and stirred at room temperature for 2 hours. After completion of the reaction, water (5 ml) was added and filtered to give examples 19-6 (95 mg, 80%).
MS m/z(ESI):424.1[M+H] + .
Seventh step: preparation of N- (3- (4-chlorobenzyl) -7-ethyl-6-oxo-6, 7-dihydro-3H-purin-2-yl) -4- ((2, 6-difluoropyridin-4-yl) oxy) benzamide
To DMF (2 ml) of example 19-6 (85 mg,0.2 mmol) was added 2,4, 6-trifluoropyridine (66 mg,0.5 mmol) and potassium carbonate (69 mg,0.5 mmol). The mixture was added to hot 80℃and the reaction was stirred for 3 hours. After completion of the reaction, filtration and purification by preparative HPLC gave example 19 (25 mg, 23%).
MS m/z(ESI):537.2[M+H] + .
Example 20
3- (4-chlorobenzyl) -2- (3- ((2, 6-difluoropyridin-4-yl) oxy) azepin-1-yl) -7-ethyl-3, 7-dihydro-6 h-purin-6-one
The first step: preparation of 1- (tert-Butoxycarbonyl) -3- ((2, 6-difluoropyridin-4-yl) oxy) azetidine
To DMF (15 ml) of example 20-1 (1.7 g,10 mmol) was added 2,4, 6-trifluoropyridine (1.3 g,10 mmol) and potassium carbonate (1.4 g,10 mmol). The mixture was added to hot 80℃and the reaction was stirred for 3 hours. After the reaction was completed, water quenching (80 mL) was added, extraction was performed with ethyl acetate (300 ml×2), and the combined organic phases were dried over anhydrous sodium sulfate, concentrated, and purified by silica gel column (petroleum ether: ethyl acetate=5:1) to obtain example 20-2 (2.5 g, 87%).
MS m/z(ESI):287.2[M+H] + .
And a second step of: preparation of 3- ((2, 6-difluoropyridin-4-yl) oxy) azetidine
Trifluoroacetic acid (8 mL) was added to DCM (15 mL) of example 20-2 (1.43 g,5 mmol) under ice-bath, and the mixture was stirred at room temperature for 2 hours. After completion of the reaction, the mixture was concentrated to give example 20-3 (960 mg, 87%).
MS m/z(ESI):187.2[M+H] + .
And a third step of: preparation of 3- (4-chlorobenzyl) -2- (3- ((2, 6-difluoropyridin-4-yl) oxy) azepin-1-yl) -7-ethyl-3, 7-dihydro-6 h-purin-6-one
Example 19-4 (100 mg,0.3 mmol), ethanol (5 mL) and example 20-3 (399mg, 2 mmol) were added to a round bottom flask and the mixture was heated to 100deg.C and allowed to react for 12 hours with stirring. The mixture was cooled to room temperature and filtered and purified by preparative HPLC to give example 20 (36 mg, 26%).
MS m/z(ESI):473.3[M+H] + .
Example 21
1- (4-chlorobenzyl) -2- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -6-isopropyl-5, 6-dihydro-1H-pyrrole [3,4-d ] pyrimidine-4, 7-dione
The synthesis method of example 21 was referred to the synthesis method of example 10.
MS m/z(ESI):538.1[M+H] + .
Example 22
3- (4-chlorobenzyl) -7- (2, 2-difluoroethyl) -2- ((2-ethyl-3-oxoisoindol-5-yl) amino) -3, 7-dihydro-6 h-purin-6-one
The first step: preparation of 3- (4-chlorobenzyl) -7- (2, 2-difluoroethyl) -2- (methylthio) -3, 7-dihydro-6 h-purin-6-one
The synthesis of example 22-1 was carried out by substituting 1, 1-difluoro-2-iodoethane for iodoethane with reference to the synthesis of example 19-4 to give example 22-1 (200 mg, 75%).
MS m/z(ESI):371.0[M+H] + .
And a second step of: preparation of 3- (4-chlorobenzyl) -7- (2, 2-difluoroethyl) -2- ((2-ethyl-3-oxoisoindol-5-yl) amino) -3, 7-dihydro-6 h-purin-6-one
The procedure of example 22 was followed, and the synthetic method of example 20 was followed, substituting example 22-1 for example 19-4 and substituting 6-amino-2-ethylisoindol-1-one for example 20-3 to give example 22 (16 mg, 31%).
MS m/z(ESI):499.1[M+H] + .
Example 23
3- (4-chlorobenzyl) -7-ethyl-2- ((2-ethyl-3-oxoisoindol-5-yl) amino) -3, 7-dihydro-6 h-purin-6-one
The first step: preparation of 3- (4-chlorobenzyl) -7-ethyl-2- ((2-ethyl-3-oxoisoindol-5-yl) amino) -3, 7-dihydro-6 h-purin-6-one
The procedure of example 23 was followed, and the synthetic method of example 20 was followed, substituting 6-amino-2-ethylisoindol 1-one for example 20-3, to give example 23 (18 mg, 30%).
MS m/z(ESI):463.2[M+H] + .
Example 24
3- (4-chlorobenzyl) -2- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -7-ethyl-3, 7-dihydro-6H-purin-6-one
The first step: preparation of 2- ((5-bromothiazol-2-yl) amino) -3- (4-chlorobenzyl) -7-ethyl-3, 7-dihydro-6H-purin-6-one
Example 24-1 (50 mg,0.15 mmol) was dissolved in t-butanol (3 mL) and acetic acid (1 mL) at room temperature, and 5-bromothiazol-2-amine (32 mg,0.18 mmol) was added to the mixture. The reaction solution was heated to 100℃and reacted for 6 hours. To the reaction was added 10ml of water, the mixture was extracted with EtOAc (2 x 10 ml) and the combined extracts were taken over Na 2 SO 4 Drying, spin-drying, and p-TLC (dichloro: methanol=20:1) gave example 24-2 (30 mg, 43%) as a white powder.
MS m/z(ESI):465.12[M+H]+.
And a second step of: preparation of 3- (4-chlorobenzyl) -2- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -7-ethyl-3, 7-dihydro-6H-purin-6-one
Example 24-2 (30 mg,0.064 mmol) was added to DMF (2 mL) followed by 2, 6-difluoropyridin-4-ol (13 mg,0.096 mmol), cs 2 CO 3 (31.5 mg,0.096 mmol). The reaction solution was heated to 80℃and reacted for 2 hours. To the reaction was added 10ml of water, the mixture was extracted with EtOAc (2 x 10 ml) and the combined extracts were taken over Na 2 SO 4 Drying, spin-drying, and p-TLC (dichloro: methanol=20:1) gave example 24 (10 mg, 31%) as a white powder.
MS m/z(ESI):509.12[M+H] + .
Example 25
3- (1- (4-chlorophenyl) cyclopropyl) -2- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -7-ethyl-3, 7-dihydro-6 h-purin-6-one
The first step: preparation of N- ((1- (4-chlorophenyl) cyclopropyl) carbamoyl) benzamide
The synthesis of example 25-1, reference to the synthesis of example 16-2, replaced p-chlorobenzylamine with 1- (4-chlorophenyl) cyclopropane-1-amine, gave example 25-1 (3.8 g, 92%).
And a second step of: preparation of 1- (1- (4-chlorophenyl) cyclopropyl) thiourea
The synthesis method of example 25-2 was referred to as the synthesis method of example 16-3, and example 25-1 was used in place of example 16-2 to obtain example 25-2 (2.7 g, 90%).
MS m/z(ESI):227.0[M+H] + .
And a third step of: preparation of 6-amino-1- (1- (4-chlorophenyl) cyclopropyl) -2-mercapto-5-nitrosopyrimidin-4 (1H) -one
The synthesis method of example 25-3 was referred to as the synthesis method of example 16-4, and example 25-2 was used in place of example 16-3 to obtain example 25-3 (2.1 g, 80%).
MS m/z(ESI):323.2[M+H] + .
Fourth step: preparation of 5, 6-diamino-1- (1- (4-chlorophenyl) cyclopropyl) -2-mercaptopyrimidin-4 (1H) -one
The synthesis of example 25-4 was performed by referring to the synthesis of example 19-1 and substituting example 25-3 for example 16-4 to give example 25-4 (1.6 g, 80%).
MS m/z(ESI):309.0[M+H] + .
Fifth step: preparation of 3- (1- (4-chlorophenyl) cyclopropyl) -2-mercapto-3, 7-dihydro-6 h-purin-6-one
The synthesis of example 25-5, reference to the synthesis of example 19-2, substituting example 25-4 for example 19-1, gave example 25-5 (1.6 g, 80%).
MS m/z(ESI):319.1[M+H] + .
Sixth step: preparation of 3- (1- (4-chlorophenyl) cyclopropyl) -2- (methylthio) -3, 7-dihydro-6 h-purin-6-one
The synthesis of example 25-6 was performed by referring to the synthesis of example 19-3 and substituting example 25-5 for example 19-2 to give example 25-6 (1.6 g, 80%).
MS m/z(ESI):333.2[M+H] + .
Seventh step: preparation of 3- (1- (4-chlorophenyl) cyclopropyl) -7-ethyl-2- (methylthio) -3, 7-dihydro-6 h-purin-6-one
The synthesis of example 25-7, reference to the synthesis of example 19-4, substituting example 25-6 for example 19-3, gave example 25-7 (1.6 g, 80%).
MS m/z(ESI):361.1[M+H] + .
Eighth step: preparation of 3- (1- (4-chlorophenyl) cyclopropyl) -2- (3- ((2, 6-difluoropyridin-4-yl) oxy) azepin-1-yl) -7-ethyl-3, 7-dihydro-6 h-purin-6-one
The synthesis of example 25, reference to the synthesis of example 20, substituting examples 25-7 for examples 19-4, gave example 25 (1.6 g, 80%).
MS m/z(ESI):535.1[M+H] + .
Example 26
3- (2- (4-chlorophenyl) -2-oxoethyl) -2- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -7-ethyl-3, 7-dihydro-6H-purin-6-one
The procedure of 8 steps of the synthesis of example 26, reference to the synthesis of example 25, was performed to obtain example 26 by substituting 2-amino-1- (4-chlorophenyl) ethan-1-one for 1- (4-chlorophenyl) cyclopropan-1-amine.
MS m/z(ESI):537.2[M+H] + .
Example 27
3- (3- (4-chlorophenyl) cyclobutyl) -2- ((4- ((2, 6-difluoropyridin-4-yl) oxy) phenyl) amino) -7-ethyl-3, 7-dihydro-6 h-purin-6-one
Synthesis of example 27 referring to the Synthesis of example 25, 8 steps were taken to obtain example 27 with 3- (4-chlorophenyl) cyclobutane-1-amine instead of 1- (4-chlorophenyl) cyclopropane-1-amine.
MS m/z(ESI):549.1[M+H] + .
Example 28
(S) -2- (((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7) -vinylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The first step: preparation of tert-butyl (S) -2- ((7-bromo-2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate
4-bromopyridin-2-amine (0.86 g,0.005 mol), 3, 5-difluoro-4-formyl-N-methylbenzamide (1 g,0.005 mol), (5) -tert-butyl 2-ethynylmorpholine) were heated by charging a 250mL jacketed reactor with N2 at a temperature of 110℃for 5min, using a mixture of 4-carboxylate (1.057 g,0.005 mol), copper (I) chloride (0.15 g,0.0015 mol), bis (trifluoromethylsulfonyloxy) copper (0.54 g,0.0015 mol) and toluene (60 mL). N, N-dimethylacetamide (0.1 mL) was then added and the resulting mixture was stirred at 110℃for 16 h. The toluene layer was separated from the solid residue by decantation and then concentrated under reduced pressure. The crude material obtained was loaded onto a dry silica gel column (25 g) and eluted with ethyl acetate-petroleum ether 1:1-3:1 to give example 28-1 as a grey solid (0.4 g, 15%).
MS m/z(ESI):565.13[M+H] + .
And a second step of: preparation of tert-butyl (S) -2- (((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-vinylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate
Example 28-1 (0.2 g,0.354 mmol), potassium vinylfluoroborate (95 mg,0.71 mol), K 2 CO 3 (147mg,1.06mmol),Pd(dppf)Cl 2 (26 mg,0.0354 mmol) was dissolved in dioxane (10 mL) and water (1 mL), and the resulting mixture was stirred at 100deg.C for 2 hours. Concentrated under reduced pressure to dryness. The crude material obtained was loaded onto a dry silica gel column (25 g) and purified using CH 2 Cl 2 : meoh=20: 1, eluting the mixture by using a washing machine,example 28-2 was obtained as a yellow solid (0.1 g, 55%).
MS m/z(ESI):513.22[M+H] + .
1 H NMR(400MHz,CDCl 3 )δ7.73–7.68(m,2H),7.62–7.58(m,1H),6.83(q,J=3.7Hz,1H),6.78(dt,J=7.5,1.4Hz,1H),6.66(t,J=16.7,9.9,1.3Hz,1H),5.87(dd,J=10.1,1.5Hz,1H),5.83(dd,J=16.7,1.4Hz,1H),4.27(t,J=7.0Hz,1H),3.99–3.90(m,2H),3.79(dd,J=12.5,7.0Hz,1H),3.72(td,J=7.2,1.4Hz,2H),3.56(dd,J=12.4,7.0Hz,1H),3.37(dd,J=12.3,7.0Hz,1H),3.22(dd,J=12.3,7.0Hz,1H),2.91(d,J=3.7Hz,3H),1.46(s,9H).
And a third step of: preparation of (S) -3, 5-difluoro-N-methyl-4- (3- (morpholin-2-ylmethyl) -7-vinylimidazo [1,2-a ] pyridin-2-yl) benzamide
Example 28-2 (0.1 g,0.2 mol) was dissolved in ethanol solution of hydrochloric acid (5 mL), and the resulting mixture was stirred at room temperature for 1 hour. The solution was suspended to give example 28-3 as a white solid (50 mg, 62%).
MS m/z(ESI):413.23[M+H] + .
Fourth step: preparation of methyl (S) -2- (((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7) -vinylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate
Example 28-3 (50 mg,0.12 mmol), DIEA (31 mg,0.24 mmol) and methyl chloroformate (23 mg,24 mmol) were dissolved in DCM (6 mL), mixed thoroughly in a 50mL round bottom flask and reacted for 0.5h at room temperature with a magnetic stirrer. After completion of the reaction (TLC, DCM: meoh=20:1), example 28 (20 mg, 35%) was obtained via climbing plate.
MS m/z(ESI):471.18[M+H] + .
1 H NMR(400MHz,CDCl 3 )δ7.73–7.68(m,2H),7.62–7.58(m,1H),6.83(q,J=3.7Hz,1H),6.78(dt,J=7.5,1.4Hz,1H),6.66(t,J=16.7,9.9,1.3Hz,1H),5.87(dd,J=10.1,1.5Hz,1H),5.83(dd,J=16.7,1.4Hz,1H),4.27(t,J=7.0Hz,1H),3.99–3.90(m,2H),3.79(dd,J=12.5,7.0Hz,1H),3.72(td,J=7.2,1.4Hz,2H),3.70(s,3H)3.56(dd,J=12.4,7.0Hz,1H),3.37(dd,J=12.3,7.0Hz,1H),3.22(dd,J=12.3,7.0Hz,1H),2.91(d,J=3.7Hz,3H).
Example 29
(S) -2- (((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7) - (methylthio) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 29 was performed according to the method of example 28 to give the title compound (15 mg,48% yield).
MS m/z(ESI):491.15[M+H] + .
Example 30
(S) -2- (((7-cyclopropyl-2- (2, 6-difluoro-4- (methylcarbamoyl)) phenyl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 30 was performed according to the method of example 28 to give the objective compound (18 mg,58% yield).
MS m/z(ESI):485.15[M+H] + .
Example 31
(S) -2- (((7-cyclobutyl-2- (2, 6-difluoro-4- (methylcarbamoyl)) phenyl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 31 was performed according to the method of example 28 to give the title compound (14 mg,48% yield).
MS m/z(ESI):499.21[M+H] + .
Example 32
(S) -2- ((2- (4- (cyclopropylcarbamoyl) -2, 6-difluorophenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 32 was performed according to the method of example 28, and the specific synthetic route is as follows:
the first step: preparation of 3, 5-difluoro-4-formylbenzoic acid
3, 5-Difluorobenzoic acid (2.0 g,12.6 mmol) was dissolved in tetrahydrofuran (30 mL), N, N, N ', N' -tetramethyl ethylenediamine (3.2 g,27.8 mmol) was added thereto, and the mixture was cooled to-78℃under nitrogen, and an N-hexane solution of N-butyllithium (1.6M, 17.4mL,27.8 mmol) was added dropwise thereto, followed by reaction at that temperature for 1.5 hours. Ethyl formate (1.86 g,25.2 mmol) was added and the reaction was allowed to slowly warm to room temperature for 4 hours. The reaction was slowly poured into ice water solution of dilute hydrochloric acid and extracted with ethyl acetate (50 ml x 3). The combined organic phases were washed with water (50 mL), saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and spun-dried to give the crude product as a white solid (2.3 g, 97%).
1 HNMR(400MHz,DMSO-d 6 )δ13.95(s,1H),10.22(s,1H),7.65(d,J=9.2Hz,2H).
MS m/z(ESI):185.1[M-H] -
And a second step of: preparation of N-cyclopropyl-3, 5-difluoro-4-formylbenzamide
3, 5-difluoro-4-formylbenzoic acid (2.3 g,12.4 mmol) was dissolved in dichloromethane (30 mL), N-dimethylformamide (92 mg,1.24 mmol) was added, cooled to 0deg.C, and oxalyl chloride (1.88 g,14.8 mmol) was added dropwise. After 4 hours of reaction at room temperature, spin-drying was performed. Triethylamine (3.13 g,62.0 mmol) and cyclopropylamine (1.41 g,24.8 mmol) were dissolved in methylene chloride (30 mL) and added thereto under ice bath, and reacted at room temperature for 0.5 hours. Poured into 100mL of water and extracted with dichloromethane (60 mL x 3). The organic phases were combined and washed with water (60 mL), saturated brine (60 mL), dried over anhydrous sodium sulfate, filtered, and spin-dried to give a white solid (1.6 g, 66%) by column chromatography (ethyl acetate/petroleum ether=0-35%).
1 HNMR(400MHz,DMSO-d 6 )δ8.65(s,1H),7.47(d,J=9.2Hz,2H),2.87-2.82(m,1H),0.82-0.78(m,2H),0.66-0.62(m,2H).
MS m/z(ESI):226.1[M+H] +
And a third step of: preparation of tert-butyl (S) -2- ((2- (4- (cyclopropylcarbamoyl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate
The compound 4-methylpyridin-2-amine (500 mg,4.63 mmol), tert-butyl (S) -2-ethynylmorpholine-4-carboxylate (977 mg,4.63 mmol), N-cyclopropyl-3, 5-difluoro-4-formylbenzamide (1.04 g,4.63 mmol), cuprous chloride (138 mg,1.39 mmol), copper triflate (503 mg,1.39 mmol) was dissolved in toluene (20 mL), nitrogen protected, and heated to 85℃with stirring. N, N-dimethylacetamide (0.2 mL) was added and reacted overnight at 85 ℃. Cooled to room temperature, poured into 100mL of water, filtered and extracted with ethyl acetate (50 mL x 3). The polar phases were combined and washed with water (50 mL), saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and spin-dried to give a yellow solid (390 mg, 16.0%) by column chromatography (methanol/dichloromethane=0 to 10%).
1 HNMR(400MHz,DMSO-d 6 )δ8.32(d,J=8.0Hz,1H),7.67(s,1H),7.51(d,J=7.2Hz,2H),6.87(d,J=6.0Hz,1H),3.89-3.70(m,3H),3.59-3.51(m,1H),3.41-3.30(m,1H),3.02-2.91(m,3H),2.87-2.75(m,1H),2.59-2.54(m,1H),2.48(s,3H),1.42(s,9H),0.90-0.86(m,2H),0.76-0.69(m,2H).
MS m/z(ESI):527.2[M+H] +
Fourth step: preparation of (S) -N-cyclopropyl-3, 5-difluoro-4- (7-methyl-3- (morpholin-2-ylmethyl) imidazo [1,2-a ] pyridin-2-yl) benzamide hydrochloride
Tert-butyl (S) -2- ((2- (4- (cyclopropylcarbamoyl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate (360 mg,0.575 mmol) was dissolved in dichloromethane (10 mL), 1M dioxane hydrochloride solution (5 mL) was added and the reaction was carried out at room temperature for 4 hours. Spin-dry to give a yellow solid (320 mg, 100%).
1 HNMR(400MHz,DMSO-d 6 )δ8.68(d,J=4.0Hz,1H),8.38(d,J=7.2Hz,1H),7.65(d,J=8.0Hz,2H),7.35(s,1H),6.83(d,J=7.2Hz,1H),3.48–3.44(m,1H),3.28–3.22(m,1H),3.01–2.95(m,1H),2.91–2.86(m,2H),2.63–2.57(m,2H),2.38(s,3H),2.29–2.21(m,1H),2.06–1.97(m,1H),0.76–0.71(m,2H),0.63–0.59(m,2H).
MS m/z(ESI):427.2[M+H] +
Fifth step: preparation of methyl (S) -2- ((2- (4- (cyclopropylcarbamoyl) -2, 6-difluorophenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate
(S) -N-cyclopropyl-3, 5-difluoro-4- (7-methyl-3- (morpholin-2-ylmethyl) imidazo [1,2-a ] pyridin-2-yl) benzamide hydrochloride (35 mg,0.076 mmol) was dissolved in dichloromethane (2 mL), triethylamine (23 mg,0.227 mmol) was added, methyl chloroformate (14 mg,0.152 mmol) was added dropwise, and the reaction was carried out at room temperature for 1 hour. Poured into 20mL of water and extracted with ethyl acetate (20 mL x 2). The organic phases were combined and washed with water (20 mL), saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and spun-dried to give the title compound (14 mg,45% yield) in reverse.
MS m/z(ESI):485.5[M+H] + .
1 H NMR(400MHz,DMSO-d6):8.67(d,J=4.0Hz,1H),8.43(d,J=7.2Hz,1H),7.64(d,J=8.4Hz,1H),7.36(s,1H),6.84(d,J=6.8HZ,1H),3.77–3.61(m,4H),3.55(s,3H),3.49–3.43(m,1H),3.25–3.19(m,1H),3.06–3.00(m,2H),2.91–2.85(m,1H),2.83–2.74(m,1H),2.38(s,3H),0.76–0.71(m,2H),0.63–0.59(m,2H).
Example 33
(2S) -2- ((2- (2, 6-difluoro-4- ((1-methoxypropane-2-yl) carbamoyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 33 was performed according to the method of example 28 to give the objective compound (10 mg,41% yield).
MS m/z(ESI):517.6[M+H] + .
Example 34
(S) -2- ((2- (2, 6-difluoro-4- ((tetrahydro-2H-pyran-4-yl) carbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 34 was performed according to the method of example 28 to give the objective compound (10 mg,41% yield).
MS m/z(ESI):529.6[M+H] + .
Example 35
(S) -2- ((2- (2, 6-difluoro-4- (3-hydroxyazepine-1-carbonyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 35 was performed according to the method of example 28 to give the objective compound (12 mg,41% yield).
MS m/z(ESI):501.5[M+H] + .
Example 36
(S) -3- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) -4, 4-difluoropiperidine-1-carboxylic acid methyl ester
The synthesis of example 36 was performed according to the method of example 28 to give the objective compound (12 mg,41% yield).
MS m/z(ESI):493.5[M+H] + .
Example 37
(S) -4- (3- ((4, 4-difluoro-1-propionylpiperidin-3-yl) methyl) -7-methylimidazole [1,2-a ] pyridin-2-yl) -3, 5-difluoro-N-methylbenzamide
The synthesis of example 37 was performed according to the method of example 28 to give the objective compound (12 mg,41% yield).
MS m/z(ESI):491.5[M+H] + .
Example 38
(S) -4- (3- ((1-ethyl-4, 4-difluoropiperidin-3-yl) methyl) -7-methylimidazole [1,2-a ] pyridin-2-yl) -3, 5-difluoro-N-methylbenzamide
The synthesis of example 38 was performed according to the method of example 28 to give the objective compound (12 mg,41% yield).
MS m/z(ESI):463.5[M+H] + .
Example 39
1- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazol [1,2-a ] pyridin-3-yl) methyl) -2-oxo-5-azabicyclo [2.2.1] heptane-5-carboxylic acid methyl ester
The synthesis of example 39 was performed according to the method of example 28 to give the objective compound (12 mg,41% yield).
MS m/z(ESI):471.5[M+H] + .
Example 40
(S) -2- (((7-methyl-2- (4-methyl-6- (methylcarbamoyl) pyridin-3-yl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The first step: preparation of 5-formyl-N, 4-dimethylpyridine carboxamide
6-bromo-4-methylnicotine (2 g,10 mmol) was dissolved in DMSO (30 mL), and methylammonium hydrochloride (1.5 g,15 mmol), triethylamine (2.02 g,20 mmol), pd (dppf) Cl was added 2 (0.7 g,1 mmol) was heated to 100℃under a CO balloon and stirred for 5 hours. 50ml of water was added to the reaction, the mixture was extracted with EtOAc (2×50 ml), and the combined extracts were taken over Na 2 SO 4 Drying, spin drying, and chromatography of the crude product on a column (dichloro: methanol=50:1) gave example 40-1 (0.9 g, 50.5%) as a white solid.
MS m/z(ESI):179.03[M+H] + .
The synthesis of example 40 was performed according to the method of example 31 to give the target compound (17 mg,49% yield).
MS m/z(ESI):438.21[M+H] + .
Example 41
(S) -2- (((7-methyl-2- (3-methyl-5- (methylcarbamoyl) thiophen-2-yl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The first step: preparation of N, 4-dimethylthiophene-2-carboxamide
4-methylthiophene-2-carboxylic acid (5 g,35.17 mmol) was dissolved in toluene (80 mL), thionyl chloride (12.55 g,106 mmol) was added, and the mixture was stirred under reflux with heating for 3 hours. The reaction mixture was concentrated under reduced pressure, and the resulting residue was dissolved in THF (80 mL). To this was added methylammonium hydrochloride (4.5 g,70.34 mmol), triethylamine (14.4 mL,105.5 mmol) and then stirred at room temperature for 1 hour. The precipitated solid was filtered, and the precipitated solid was collected and then washed with diethyl ether to give example 41-1 (4.5 g, 82%).
MS m/z(ESI):156.03[M+H] + .
And a second step of: preparation of 5-formyl-N, 4-dimethylthiophene-2-carboxamide
Example 41-1 (1 g,6.44 mmol) was added to DMF (20 mL) followed by POCl 3 (2.96 g,19.33 mmol). The reaction solution was heated to 80℃and reacted for 5 hours. 50ml of water was added to the reaction, the mixture was extracted with EtOAc (2×50 ml), and the combined extracts were taken over Na 2 SO 4 Drying, spin drying, and chromatography of the crude product on a column (dichloro: methanol=20:1) gave example 41-2 (0.8 g, 68%) as a white solid.
MS m/z(ESI):184.04[M+H] + .
The synthesis of example 41 was followed by the procedure of example 31 to give the title compound (15 mg,47% yield).
MS m/z(ESI):443.18[M+H] + .
Example 42
(R) -5- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -3, 3-difluoropiperidine-1-carboxylic acid methyl ester
The first step: preparation of 1- (tert-butyl) 3-methyl 5-oxopiperidine-1, 3-dicarboxylic acid ester
The compound 4-nitro-1H-pyrazole-3-carboxylic acid (5 g,19.28 mmol) was placed in a 250mL round bottom flask, then DCM (100 mL) and Dess Martin (9.8 g,23.14 mmol) were added the reaction was stirred at 25℃for 16H, then water (80 mL) was added, extracted with DCM, the organic layers were combined, washed with brine, and dried over anhydrous Na 2 SO 4 Dried and concentrated in vacuo. The crude material was purified by column chromatography to give example 42-1 as a white solid (4 g, 80%).
MS m/z(ESI):258.13[M+H] + .
The synthesis of example 42 was followed by the procedure of example 36 to give the title compound (14 mg,48% yield).
1 H NMR(400MHz,CDCl 3 )δ7.99–7.88(m,1H),7.56–7.48(m,3H),6.87(d,J=6.8Hz,1H),4.24–3.98(m,2H),3.66(s,3H),3.02(s,3H),2.91–2.71(m,2H),2.48(s,3H),2.23-1.94(m,5H).
MS m/z(ESI):493.18[M+H] + .
Example 42A
(S) -5- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazo [1,2-a ] pyridin-3-yl) methyl) -3, 3-difluoropiperidine-1-carboxylic acid methyl ester
The synthesis of example 42A was performed according to the method of example 36 to give the target compound (9.6 mg,23% yield).
1 H NMR(400MHz,CDCl 3 )δ7.99–7.88(m,1H),7.56–7.48(m,3H),6.87(d,J=6.8Hz,1H),4.24–3.98(m,2H),3.66(s,3H),3.02(s,3H),2.91–2.71(m,2H),2.48(s,3H),2.23-1.94(m,5H).
MS m/z(ESI):493.18[M+H] + .
Example 43
(R) -4- (3- (((5, 5-difluoro-1-propionylpiperidin-3-yl) methyl) -7) -methylimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluoro-N-methylbenzamide
The synthesis of example 43 was performed according to the method of example 36 to give the objective compound (18 mg,53% yield).
MS m/z(ESI):491.20[M+H] + .
Example 44
((S) -N-cyclopropyl-3, 5-difluoro-4- (7-methyl-3- ((4-propionylmorpholin-2-yl) methyl) imidazo [1,2-a ] pyridin-2-yl) benzamide
The synthesis of example 44 was performed according to the method of example 28 to give the title compound (18.1 mg,54% yield).
1 H NMR(400MHz,DMSO-d 6 )δ8.66(d,J=3.6Hz,1H),8.45–8.41(m,1H),7.68–7.61(m,2H),7.35(s,1H),6.86–6.81(m,1H),4.14–4.04(m,1H),3.71–3.60(m,2H),3.49–3.39(m,2H),3.24–3.20(m,1H),3.08–2.99(m,2H),2.91–2.83(m,1H),2.75–2.68(m,1H),2.38(s,3H),2.30–2.16(m,2H),0.92(t,J=7.2Hz,3H),0.76–0.71(m,2H),0.63-0.59(m,2H).
MS m/z(ESI):483.5[M+H] + .
Example 45
(S) -2- ((2- (4- (cyclopropylcarbamoyl) -2, 6-difluorophenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid ethyl ester
/>
The synthesis of example 45 was performed according to the method of example 28 to give the title compound (18.1 mg,54% yield).
MS m/z(ESI):499.5[M+H] + .
1H NMR(400MHz,DMSO-d6):8.70–8.67(m,1H),8.42(d,J=6.8Hz,1H),7.65(d,J=8.4Hz,1H),7.36(s,1H),6.87–6.83(m,1H),4.04–3.93(m,2H),3.71–3.59(m,4H),3.52–3.42(m,1H),3.25–3.19(m,1H),3.09–3.01(m,2H),2.91–2.85(m,1H),2.82–2.73(m,1H),2.38(s,3H),1.13(t,J=7.2Hz,3H),0.77–0.70(m,1H),0.63–0.57(m,2H).
Example 46
(S) -2- ((2- (3-fluoro-5- (methylcarbamoyl) pyridin-2-yl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 46 was performed according to the method of example 28 to give the title compound (10 mg,42% yield).
MS m/z(ESI):442.5[M+H] + .
Example 47
(S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -8-fluoro-7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 47 was performed according to the method of example 28 to give the title compound (10 mg,42% yield).
MS m/z(ESI):477.5[M+H] + .
Example 48
Methyl (S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methoxyimidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
/>
The synthesis of example 48 was performed according to the method of example 28 to give the objective compound (10 mg,42% yield).
MS m/z(ESI):475.5[M+H] + .
Example 49
(S) -2- ((8-chloro-2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 49 was performed according to the method of example 28 to give the title compound (10 mg,42% yield).
MS m/z(ESI):493.9[M+H] + .
Example 50
(S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methoxy-6- (methylamino) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 50 was performed according to the method of example 28 to give the target compound (10 mg,42% yield).
MS m/z(ESI):504.5[M+H] + .
Example 51
(S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) imidazo [1,2-a ] thiophen [3,2-d ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 51 was performed according to the method of example 28 to give the objective compound (10 mg,42% yield).
MS m/z(ESI):501.5[M+H] + .
Example 52
(S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -6-methyl-6H-imidazo [1,2-a ] pyrrolo [3,2-d ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 52 was performed according to the method of example 28 to give the title compound (10 mg,42% yield).
MS m/z(ESI):498.5[M+H] + .
Example 53
(S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) furan [3,2-d ] imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 53 was performed according to the method of example 28 to give the title compound (10 mg,42% yield).
MS m/z(ESI):485.5[M+H] + .
Example 54
(S) -2- ((7- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) imidazo [1,2-a ] thiazol [5,4-d ] pyridin-6-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 54 was performed according to the method of example 28 to give the title compound (10 mg,42% yield).
MS m/z(ESI):502.5[M+H] + .
Example 55
(S) -2- ((6- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) imidazo [1,2-a ] thiazol [4,5-d ] pyridin-7-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 55 was performed according to the method of example 28 to give the title compound (10 mg,42% yield).
MS m/z(ESI):502.5[M+H] + .
Example 56
(S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) imidazo [2,1-g ] [1,7] naphthyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 56 was performed according to the method of example 28 to give the title compound (10 mg,42% yield).
MS m/z(ESI):496.5[M+H] + .
Example 57
(S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazole [2,1-g ] [1,7] naphthyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 57 was performed according to the method of example 28 to give the title compound (10 mg,42% yield).
MS m/z(ESI):510.5[M+H] + .
Example 58
(S) -N-cyclopropyl-4- (3- ((4- (2, 2-difluoroacetyl) morpholin-2-yl) methyl) -7-methylimidazo [1,2-a ] pyridin-2-yl) -3, 5-difluorobenzamide
The synthesis of example 58 was performed according to the method of example 28 to give the title compound (30 mg,81% yield).
1 H NMR(400MHz,Chloroform-d)δ8.42(s,1H),7.98(s,1H),7.57(s,2H),7.03(d,J=25.7Hz,1H),6.07(t,J=53.5Hz,1H),4.30(d,J=13.2Hz,1H),4.04–3.73(m,2H),3.67(s,1H),3.44(m,1H),3.24(s,1H),3.00(d,J=51.7Hz,3H),2.55(m,4H),0.89(d,J=7.2Hz,2H),0.74(d,J=7.2Hz,2H).
MS m/z(ESI):505.5[M+H] + .
Example 59
(S) -2- ((2- (2, 6-difluoro-4- (N-methylsulfamoyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 59 was performed according to the method of example 28 to give the objective compound (70 mg,60% yield).
1 HNMR:(400MHz,DMSO-d6)δ8.44(d,J=7.2Hz,1H),7.82(s,1H),7.59(d,J=6.1Hz,2H),7.36(s,1H),6.85(d,J=7.0Hz,1H),3.77(d,J=13.1Hz,1H),3.63(d,J=12.2Hz,2H),3.55(s,3H),3.49–3.45(m,3H),3.23(d,J=11.6Hz,1H),3.13–2.98(m,2H),2.53(s,3H),2.38(s,3H).
MS m/z(ESI):495.1[M+H] + .
Example 60
(S) -2- ((2- (2, 6-difluoro-4- (methylcarbamoyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid cyclopropyl ester
The synthesis of example 60 was performed according to the method of example 28 to give the objective compound (25 mg,86% yield).
1 H NMR(400MHz,DMSO-d6)δ8.70(d,J=5.2Hz,1H),8.42(d,J=7.1Hz,1H),8.35(s,1H),7.65(d,J=8.1Hz,1H),7.36(s,1H),6.84(d,J=7.1Hz,1H),3.93(s,1H),3.62(s,2H),3.03(d,J=6.4Hz,2H),2.83(d,J=4.4Hz,3H),2.72-2.67(m,1H),2.38(s,5H),2.00(q,J=7.2,6.6Hz,2H),0.91–0.82(m,1H),0.64–0.49(m,3H).
MS m/z(ESI):485.5[M+H] + .
Example 61
(S) -2- ((7-methyl-2- (2, 3, 6-trifluoro-4- (methylcarbamoyl) phenyl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 61 was performed according to the method of example 28 to give the objective compound (25 mg,86% yield).
MS m/z(ESI):477.5[M+H] + .
Example 62
(S) -2- ((7-methyl-2- (2, 3, 6-trifluoro-4- (methylcarbamoyl) phenyl) imidazo [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid cyclopropyl ester
The synthesis of example 62 was performed according to the method of example 28 to give the title compound (18 mg,86% yield).
MS m/z(ESI):503.5[M+H] + .
Example 63
(S) -2- ((2- (2, 6-difluoro-4- ((2, 2-trifluoroethyl) carbamoyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 63 was performed according to the method of example 28 to give the objective compound (21 mg,86% yield).
MS m/z(ESI):527.5[M+H] + .
Example 64
(S) -2- ((2- (2, 6-difluoro-4- ((2, 2-trifluoroethyl) carbamoyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid cyclopropyl ester
The synthesis of example 64 was performed according to the method of example 28 to give the objective compound (11 mg,45% yield).
MS m/z(ESI):553.5[M+H] + .
Example 65
(S) -3, 5-difluoro-4- (7-methyl-3- ((4-methylsulfonyl) morpholin-2-yl) methyl) imidazo [1,2-a ] pyridin-2-yl) -N- (2, 2-trifluoroethyl) benzamide
The synthesis of example 65 was performed according to the method of example 28 to give the title compound (15 mg,75% yield).
MS m/z(ESI):547.5[M+H] + .
Example 66
(S) -4- (3- ((4- (cyclopropylsulfonyl) morpholin-2-yl) methyl) -7-methylimidazol [1,2-a ] pyridin-2-yl) -3, 5-difluoro-N- (2, 2-trifluoroethyl) benzamide
The synthesis of example 66 was performed according to the method of example 28 to give the title compound (15 mg,72% yield).
MS m/z(ESI):573.6[M+H] + .
Example 67
(S) -4- (3- ((4- (ethylsulfonyl) morpholin-2-yl) methyl) -7-methylimidazole [1,2-a ] pyridin-2-yl) -3, 5-difluoro-N-methylbenzamide
The synthesis of example 67 was performed according to the method of example 28 to give the objective compound (11 mg,72% yield).
MS m/z(ESI):493.5[M+H] + .
Example 68
(S) -4- (3- ((4- (N, N-dimethyl sulfamoyl) morpholin-2-yl) methyl) -7-methylimidazole [1,2-a ] pyridin-2-yl) -3, 5-difluoro-N- (2, 2-trifluoroethyl) benzamide
The synthesis of example 68 was performed according to the method of example 28 to give the objective compound (11 mg,61% yield).
MS m/z(ESI):576.6[M+H] + .
Example 69
(S) -4- (3- ((4- ((cyclopropylmethyl) sulfonyl) morpholin-2-yl) methyl) -7-methylimidazole [1,2-a ] pyridin-2-yl) -3, 5-difluoro-N- (2, 2-trifluoroethyl) benzamide
The synthesis of example 69 was performed according to the method of example 28 to give the title compound (14 mg,58% yield).
MS m/z(ESI):587.6[M+H] + .
Example 70
(S) -4- (3- ((4- (2, 2-difluoroacetyl) morpholin-2-yl) methyl) -7-methylimidazole [1,2-a ] pyridin-2-yl) -3, 5-difluoro-N-methylbenzenesulfonamide
The synthesis of example 70 was performed according to the method of example 59 to give the objective compound (14 mg,58% yield).
MS m/z(ESI):515.5[M+H] + .
Example 71
(S) -2- ((2- (4- (2, 2-difluoroacetamide) -2, 6-difluorophenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 71 was performed according to the method of example 28 to give the title compound (14 mg,58% yield).
MS m/z(ESI):587.6[M+H] + .
Example 72
(S) -2- ((2- (2, 6-difluoro-4- (methylsulfonamino) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 72 was performed according to the method of example 71 to give the objective compound (14 mg,30% yield).
MS m/z(ESI):495.5[M+H] + .
Example 73
(S) -2- ((2- (4- ((N, N-dimethyl-sulfamoyl) amino) -2, 6-difluorophenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 73 was performed according to the method of example 71 to give the title compound (18 mg,28% yield).
MS m/z(ESI):524.6[M+H] + .
Example 74
(S) -2- ((2- (2, 6-difluoro-4- ((methoxycarbonyl) amino) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 74 was performed according to the method of example 71 to give the objective compound (21 mg,50% yield).
MS m/z(ESI):475.5[M+H] + .
Example 75
(S) -2- ((2- (2, 6-difluoro-4- (N-methylsulfamoyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid cyclopropyl ester
The synthesis of example 75 was performed according to the method of example 59 to give the objective compound (24 mg,50% yield).
MS m/z(ESI):521.6[M+H] + .
Example 76
(S) -3, 5-difluoro-N-methyl-4- (7-methyl-3- ((4-methylsulfonyl) morpholin-2-yl) methyl) imidazo [1,2-a ] pyridin-2-yl) benzenesulfonamide
The synthesis of example 76 was performed according to the method of example 59 to give the objective compound (20 mg,40% yield).
MS m/z(ESI):515.6[M+H] + .
Example 77
(S) -2- ((2- (2, 6-difluoro-4- (N-isopropylsulfamoyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 77 was performed according to the method of example 59 to give the title compound (46 mg,41% yield).
MS m/z(ESI):523.6[M+H] + .
Example 78
(S) -2- ((2- (2, 6-difluoro-4- (N-cyclopropylsulfamoyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 78 was performed according to the method of example 59 to give the objective compound (42 mg,41% yield).
MS m/z(ESI):521.6[M+H] + .
Example 79
(S) -2- ((2- (4- (N, N-Dimethylsulfamoyl) -2, 6-difluorophenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 79 was performed according to the method of example 59 to give the objective compound (31 mg,41% yield).
MS m/z(ESI):509.5[M+H] + .
Example 80
(S) -2- ((2- (4- (cyclopropanesulfonamide) -2, 6-difluorophenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 80 was performed according to the method of example 71 to give the objective compound (33 mg,47% yield).
MS m/z(ESI):521.6[M+H] + .
Example 81
(S) -2- ((2- (4- (cyclopropylamide) -2, 6-difluorophenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 81 was performed according to the method of example 71 to give the title compound (30 mg,47% yield).
MS m/z(ESI):485.5[M+H] + .
Example 82
(S) -2- ((2- (2, 6-difluoro-4- (methylsulfonyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 82 was performed according to the method of example 59 to give the title compound (30 mg,47% yield).
MS m/z(ESI):480.5[M+H] + .
Example 83
(S) -2- ((2- (2, 6-difluoro-4- (cyclopropylsulfonyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 83 was performed according to the method of example 59 to give the title compound (30 mg,47% yield).
MS m/z(ESI):506.5[M+H] + .
Example 84
(2S) -2- ((2- (2, 6-difluoro-4- (S-methylsulfonyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The first step: preparation of methyl (2S) -2- ((2- (2, 6-difluoro-4- (methylsulfonyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate
The synthesis of example 84-1 was performed according to the method of example 59 to obtain the objective compound (100 mg,54% yield).
MS m/z(ESI):464.5[M+H] + .
And a second step of: preparation of methyl (2S) -2- ((2- (2, 6-difluoro-4- (methylsulfonyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate
Example 84-1 (100 mg) and trifluoroacetamide (41.2 mg) were dissolved in methylene chloride (4 mL), and then MgO (37.5 mg) and Rh were added 2 (OAc) 4 (2 mg) and iodobenzene acetate (90.1 mg) were added thereto, stirred at room temperature overnight, filtered, and the solid was washed with methylene chloride, concentrated in solution, and purified by column chromatography to give example 84-2 (30 mg,20% yield).
MS m/z(ESI):575.5[M+H] + .
And a third step of: preparation of methyl (2S) -2- ((2- (2, 6-difluoro-4- (S-methylsulfonyl) phenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylate
Example 84-2 (30 mg) was dissolved in methanol (2 mL), potassium carbonate (120 mg) was added and stirred at room temperature for 2 hours, water and ethyl acetate were added, the pH was adjusted to 6 with 3N hydrochloric acid solution, the solution was separated, concentrated and dried, and example 84 (13 mg, yield 52%) was obtained by separation and purification.
MS m/z(ESI):479.5[M+H] + .
Example 85
(2S) -2- ((2- (4- (cyclopropanesulfonylimino) -2, 6-difluorophenyl) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 85 was performed according to the method of example 84 to give the target compound (21 mg,47% yield).
MS m/z(ESI):505.6[M+H] + .
Example 86
(2S) -2- ((2- (4- (methylsulfonylimino) -2, 6-difluorophenylamino) -7-methylimidazole [1,2-a ] pyridin-3-yl) methyl) morpholine-4-carboxylic acid methyl ester
The synthesis of example 86 was performed according to the method of example 84 to give the objective compound (11 mg,30% yield).
MS m/z(ESI):494.5[M+H] + .
Biological test evaluation
The invention is further illustrated below in conjunction with test examples, which are not meant to limit the scope of the invention.
1. Cell function experiment
Test example 1 determination of the influence of the Compounds of the invention on the ability of calcium ion to flow in cells stably expressing the HEK293-hP2X3 receptor
1. The purpose of the experiment is as follows:
the inhibition of HEK293-hP2X3 receptor by the compounds was measured.
2. Laboratory instruments and reagents:
2.1 laboratory apparatus:
384 well-assay plates (Corning; 3712);
pipette (Axygen);
FLIPR(Molecular Devices)。
2.2 experimental reagents:
DMEM(Invitrogen;11965);
fetal bovine serum (Biowest; S1810-500);
dialyzing serum (S-FBS-AU-065; seran);
penicillin and streptomycin (Biowest; L0022-100);
hygromycin B (CABIOCHEM, 400052);
Matrigel(BD;354230);
DMSO(Sigma;D2650);
HBSS(Invitrogen;14065);
HEPES(Invitrogen;15630080);
Probenecid(Sigma;P8761);
BSA(renview;FA016);
trypsin (HDB; 0458);
stable transformed cells: HEK293-hP2X3 (offered by Kang Long chemical (beijing) new technologies inc.). 3. The experimental method comprises the following steps:
1. buffer preparation: 1x HBSS,20mM HEPES,2.5mM probenecid (probenecid 400mM stock in 1M NaOH), 0.1% BSA. Probenicid and BSA were added fresh on the day of the experiment. The assay buffer includes dye buffer and compound dilution buffer.
2. After cells were digested with trypsin, the cells were digested with trypsin at 1X 10 4 The density of individual cells/well was seeded into 384 well-assay plates and incubated for 16-24 hours (at least overnight).
3. The broth was discarded and 20 μl of dye was added. Incubated at 37 ℃ for 60min in dark place, and the calcium signal is read.
4. Antagonists were prepared prior to the experiment. 5. Mu.L/well of 5x antagonist compound was added to 384 well-assay plates and incubated at room temperature for 15min in the absence of light. The assay plate was transferred to the FLIPR and agonist compound was added at a concentration of 5 μl/well 6 x. The values were read using the FLIPR and the data saved. The total assay volume was 30 μl, including 20 μl/well dye buffer, 5 μl/well 5x concentration of test compound, and 5 μl/well 6x concentration of agonist compound. 4. The experimental data processing method comprises the following steps:
the calcium signal values were read by FLIPR. The calculated output for each sampling time point in the experiment is the ratio of 340/510nm to 380/510nm wavelength signal. The maximum minus the minimum results from the ratio signal curve.
IC of the compound was calculated using GraphPad prism fit percent inhibition and ten-point concentration data to parametric nonlinear logistic equation 50 Values.
5. Experimental results:
the results of the compounds of the present invention in the functional calcium flux assay of HEK293-hP2X3 receptor cells are shown in Table 1:
TABLE 1
6. Conclusion of experiment:
according to the scheme, the compound disclosed by the invention shows good inhibition effect in HEK293-hP2X3 receptor cell functional calcium flux test.
Test example 2 determination of the influence of the Compounds of the invention on the ability of calcium ion to flow in cells stably expressing the 1321N1-hP2X3 receptor
1. The purpose of the experiment is as follows:
measurement of inhibition of 1321N1-hP2X3 cell Activity by Compounds 2. Laboratory instruments and reagents:
2.1 laboratory apparatus:
384 well-assay plates (Corning; 3712);
384 well-compound plates (Corning; 3657);
384 well-assay plates (LABCYTE; P-05525);
FLIPR(Molecular Devices)。
2.2 experimental reagents:
DMEM(Gibco;11965);
FBS(Gibco;10099-141);
dialyzing serum (S-FBS-AU-065; seran);
penicillin and streptomycin (Biowest; L0022-100);
hygromycin B (Invitrogen, 10687010);
Matrix(Thermo;5416);
DMSO(Sigma;D2650);
HBSS(Invitrogen;14025);
HEPES(Invitrogen;15630080);
Probenecid(Sigma;P8761);
Versene(Gibco;15040066);
G418(Sigma;G5013);
Calcium 4 Assay Kit(Molecular Devices;R8141);
α,β-meATP;
stable transformed cells: 1321N1-hP2X3 (available from Shanghai Ming's chemical research Co., ltd.).
3. The experimental method comprises the following steps:
1. buffer preparation: assay buffer was prepared and Dye was diluted 1X.
2. After cells were digested with trypsin, the cells were digested with trypsin at 1X 10 4 The density of individual cells/well was seeded into 384 well-assay plates and incubated for 16-24 hours (at least overnight).
3. The culture broth was discarded, 30. Mu.L of dye was added and incubated at 37℃for 60min in the dark.
4. Preparation of 3X compound plates: 500nl 180X compound was transferred to a compound plate (source plate for FLIPR) with ECHO, 30ul assay buffer per well and gently shaken for 20-40 min. And (3) detecting: 15uL of 3X compound was added to the cell plate per well (FLIPR instrument loading) and assayed for calcium signaling. After 15 minutes, 22.5ul 3x agonist (EC 80 Concentration), calcium signal was detected.
4. The experimental data processing method comprises the following steps:
the calcium signal values were read by FLIPR. The calculated output for each sampling time point in the experiment is the ratio of 340/510nm to 380/510nm wavelength signal. The maximum minus the minimum results from the ratio signal curve.
IC of the compound was calculated using GraphPad prism fit percent inhibition and ten-point concentration data to parametric nonlinear logistic equation 50 Values.
5. Experimental results:
the results of the compounds of the examples of the invention in the 1321N1-hP2X3 receptor cell functional calcium flux assay are shown in Table 2:
TABLE 2
Names of Compounds 1321N1-hP2X3 IC 50 (nM)
Example 32 75.03
Example 42 39.00
Example 42A 50.10
Example 44 31.68
Example 58 40.84
Example 59 57.50
Example 60 54.40
6. Conclusion of experiment:
the scheme shows that the compound disclosed by the invention has a better inhibition effect in a 1321N1-hP2X3 receptor cell functional calcium flux test.
Test example 3 determination of the influence of the Compounds of the invention on the ability of calcium ion to flow in cells stably expressing the P2X2/3 receptor
1. The purpose of the experiment is as follows:
the inhibition of the P2X2/3 receptor by the compounds was measured.
2. Laboratory instruments and reagents:
2.1 laboratory apparatus:
384 well-assay plates (Corning; 3712);
384 well-compound plates (Corning; 3657);
384 well-assay plates (LABCYTE; P-05525);
pipette (Axygen);
FLIPR(Molecular Devices)。
2.2 experimental reagents:
DMEM(Invitrogen;11965);
fetal bovine serum (Biowest; S1810-500);
dialyzing serum (S-FBS-AU-065; seran);
penicillin and streptomycin (Biowest; L0022-100);
hygromycin B (CABIOCHEM, 400052);
Matrigel(BD;354230);
DMSO(Sigma;D2650);
HBSS(Invitrogen;14065);
HEPES(Invitrogen;15630080);
Probenecid(Sigma;P8761);
BSA(renview;FA016);
trypsin (HDB; 0458);
FBS(Gibco;10099-141);
Matrix(Thermo;5416);
Versene(Gibco;15040066);
G418(Sigma;G5013);
Calcium 4Assay Kit(Molecular Devices;R8141);
α,β-meATP;
stable transformed cells: 1321N1-hP2X2/3 (available from Shanghai Ming's research GmbH).
3. The experimental steps are as follows:
3.1 Experimental method one:
1. buffer preparation: 1x HBSS,20mM HEPES,2.5mM probenecid (probenecid 400mM stock in 1M NaOH), 0.1% BSA. Probenicid and BSA were added fresh on the day of the experiment. The experiment buffer comprises dye buffer, compound dilution buffer and the like.
2. After cells were digested with trypsin, the cells were digested with trypsin at 1X 10 4 The density of individual cells/well was seeded into 384 well-assay plates and incubated for 16-24 hours (at least overnight).
3. The broth was discarded and 20 μl of dye was added. Incubated at 37 ℃ for 60min in dark place, and the calcium signal is read.
4. Antagonists were prepared prior to the experiment. 5uL/well of 5x antagonist compound was added to 384 well-assay plates and incubated at room temperature in the absence of light for 15min. The assay plate was transferred to the FLIPR and agonist compound was added at a concentration of 5 μl/well 6 x. The values were read using the FLIPR and the data saved. The total assay volume was 30uL, including 20 uL/well dye buffer, 5uL/well 5x concentration of test compound, and 5uL/well 6x concentration of agonist compound. 3.2 Experimental methods two:
1. Buffer preparation: assay buffer was prepared and Dye was diluted 1X.
2. After cells were digested with trypsin, the cells were digested with trypsin at 1X 10 4 Individual cellsDensity of/well was seeded to 384 well-assay plates for 16-24 hours (at least overnight).
3. The broth was discarded and 30 μl of dye was added. Incubate at 37℃for 60min in the dark.
4. Preparation of 3X compound plates: 500nl 180X compound was transferred to a compound plate (source plate for FLIPR) with ECHO, 30ul assay buffer per well and gently shaken for 20-40 min. And (3) detecting: 15uL of 3X compound was added to the cell plate per well (FLIPR instrument loading) and assayed for calcium signaling. After 15 minutes, 22.5ul 3x agonist (EC 80 Concentration), calcium signal was detected.
4. The experimental data processing method comprises the following steps:
the calcium signal values were read by FLIPR. The calculated output for each sampling time point in the experiment is the ratio of 340/510nm to 380/510nm wavelength signal. The maximum minus the minimum results from the ratio signal curve.
IC of the compound was calculated using GraphPad prism fit percent inhibition and ten-point concentration data to parametric nonlinear logistic equation 50 Values.
5. Experimental results:
the inhibition of P2X2/3 receptor by compounds can be measured by both of the above protocols, and the invention uses the second experimental method to test the functional calcium flux of the compounds of the examples on 1321N1-hP2X2/3 receptor cells, and the results are shown in Table 3:
TABLE 3 Table 3
Names of Compounds 1321N1-hP2X2/3 IC 50 (nM)
BLU-5937 3381
Example 28 16890
Example 32 13420
Example 42 27100
Example 42A 15960
Example 44 11610
Example 58 13060
Example 60 9615
6. Conclusion of experiment:
from the above scheme, the compounds shown in the invention show smaller inhibition effect in the P2X2/3 receptor cell function calcium flow test.
Test example 4, balb/C mouse pharmacokinetic assay
1. Study purposes:
the following examples of compounds were studied using Balb/C mice as test animals, orally administered plasma pharmacokinetic behavior in mice at a dose of 5 mg/kg.
2. Test protocol
2.1 test drug:
according to the embodiment of the invention, the home-made product is prepared.
2.2 test animals:
Balb/C Mouse 6/example, male, shanghai Jieshike laboratory animal Co., ltd., animal production license number (SCXK (Shanghai) 2013-0006N0.311620400001794).
2.3 preparation of medicine:
5g of hydroxyethylcellulose (HEC, CMC-Na, viscosity: 800-1200 Cps) was weighed, dissolved in 1000mL of purified water, and 10g of Tween80 was added. Uniformly mixing to obtain a clear solution.
2.4 administration:
Balb/C mice, males; after one night of fasting, the dosages were 5mg/kg, respectively, and the dosing volumes were 10mL/kg.
2.5 sample collection:
mice were bled with 0.1mL of orbital blood at 0, 0.5, 1, 2, 4, 6, 8 and 24 hours, and placed in EDTA-K before and after dosing 2 In a test tube, plasma was separated by centrifugation at 6000rpm at 4℃for 6min and stored at-80 ℃.
2.6 sample treatment:
1) The plasma sample 40uL was precipitated by adding 160uL acetonitrile, and centrifuged at 3500 Xg for 5-20 minutes after mixing.
2) The concentration of the test compound was analyzed by LC/MS/MS by taking 100uL of the supernatant solution after the treatment.
2.6 liquid phase analysis
Liquid phase conditions: shimadzu LC-20AD pump
Mass spectrometry conditions AB Sciex API 4000 Mass spectrometer
Chromatographic column: phenomenex Gemiu 5um C18.times.4.6 mm
Mobile phase: solution A is 0.1% formic acid water solution, solution B is acetonitrile
Flow rate: 0.8mL/min
Elution time: 0-4.0 minutes, the eluent is as follows:
3. test results and analysis:
the main pharmacokinetic parameters were calculated using WinNonlin 6.1 and the results of the mouse drug substitution experiments are shown in table 4 below:
TABLE 4 results of mouse drug substitution experiments
4. Conclusion of experiment:
as can be seen from the results of the mouse drug generation experiments in the table, the compound of the embodiment of the invention shows good metabolic properties, exposure AUC and maximum blood concentration C max All perform well.
Test example 5 test of metabolic stability of liver microsomes
1. The purpose of the experiment is as follows:
the purpose of this experiment was to examine the stability of the compounds of the examples in mouse, rat, canine and human liver microsomes.
2. The experimental steps are as follows:
2.1 preparing working solution of Compound
Working solution preparation of the compound: the compound stock solution was added to phosphate buffer at a final concentration of 20. Mu.M.
2.2 preparation of liver microsome working solution
Diluted with 100mM phosphate buffer to a final concentration of 0.625mg/mL.
2.3 preparation of NADPH and UDPGA
NADPH (reduced nicotinamide adenine dinucleotide phosphate) and UDPGA (uridine diphosphate glucuronic acid) were weighed and added to 100mM phosphate buffer, at a final concentration of 20mM.
2.4 preparation of the perforating agent
1mg of Alamethacin (Propofol) was weighed into 200. Mu.L of DMSO to prepare a 5mg/mL solution. And diluted with phosphate buffer to a final concentration of 50. Mu.g/mL.
2.5 preparation of reaction termination solution
Stop solution: contains 100ng/mL labetalol hydrochloride and 400ng/mL tolbutamide as internal standard cold acetonitrile.
2.6 incubation procedure
400. Mu.L of the prepared liver microsomes, 25. Mu.L of the compound working solution and 25. Mu.L of Alamethacin were sequentially added to a 96-well plate, and pre-incubated at 37℃for 10min. Then 50. Mu.L of the prepared NADPH/UDPGA was added to start the reaction, and incubated at 37℃with a total volume of 500. Mu.L, and the final contents of the respective components were as follows:
composition of the components Content of
Liver microsome 0.5mg/mL
Compounds of formula (I) 1μM
NADPH 2mM
UDPGA 2mM
Alamethicin 2.5μg/mL
2.7 sample analysis
2.7.1 chromatographic conditions:
Instrument: shimadzu LC-30AD;
chromatographic column:c18 (50 x 4.6mm,5 μm particle size);
mobile phase: a:0.1% formic acid solution, B: methanol
Flushing gradient: 0.2-1.6 min 5% A to 95% A, 3.0-3.1 min 95% A to 5%A
Run time: 4.0min.
2.7.2 Mass Spectrometry conditions:
instrument: API5500 type liquid chromatograph-mass spectrometer, AB Sciex company;
ion source: electrospray ionization source (ESI);
drying gas: n (N) 2 The temperature is 500 ℃;
electrospray voltage: 5000V;
the detection mode is as follows: detecting positive ions;
scanning mode: reaction Monitoring (MRM) mode.
3. Experimental results:
table 5: example Compounds liver microsomal metabolic stability results
4. Conclusion of experiment:
the above data shows that the compound of example 32 of the present invention has good metabolic stability in liver microsomes of mice, rats, dogs and humans, and better stability than the reference compound BLU-5937.
Test example 6, BALB/c mouse taste sensitivity test
1. The purpose of the experiment is as follows:
the compound with small toxic and side effects on animal taste is screened out by quinine bitter water experiments.
2. Experimental main instrument and materials
2.1 instrument:
1. ultra clean bench (CJ-2F, von Willebrand laboratory animal Co., ltd.);
2. Electronic balance (CPA 2202D, cerdolis);
3. electronic balance (BSA 2202S-CW, sidoris);
4. pure water instrument (Pacific TII, thermo).
2.2 reagents:
quinine monohydrochloride dihydrate (6119-47-7, adamas).
2.3 animals:
BALB/c mice, 6-8 weeks,purchased from Shanghai Sipuler-BiKai laboratory animals Co.
3. The experimental steps are as follows:
3.1 animal screening
1 day prior to the experiment, all BALB/c mice were weighed and animals with either over-high or under-body weight were removed.
3.2 grouping and Water Disable
BALB/c mice were randomized on demand according to body weight and were kept off water and not fasted 12-16 hours prior to dosing.
3.3 preparation of quinine aqueous solution
Weighing a proper amount of quinine monohydrochloride dihydrate, and preparing quinine hydrochloric acid aqueous solution with concentration of 3mmol/L by using ultrapure water for later use.
3.4 preparation of test Compounds
Weighing a proper amount of compound to be tested, and preparing the compound to be tested into target concentration by using a corresponding solvent according to an experimental design for later use. 3.5 dosing and animal quinine Water intake test
Dosing and fasting: on the day of the experiment, animals were weighed, fasted, and the pads were replaced and dosed as designed.
Quinine water intake test:
1. rinsing corresponding clean mouse drinking bottle with ultrapure water and prepared quinine hydrochloride water solution of 3mmol/L for 2-3 times, respectively, and weighing and recording the weight as Wi 0
2. According to experimental design, after administration for a certain period of time, the filled water bottle is gently placed on the corresponding mouse cage, timing is started, after 30min, the water bottle is gently taken out, and the weight is recorded as Wi 30
3. Water intake calculation for each group of animals: Δww (g) =wi 30 -Wi 0 The method comprises the steps of carrying out a first treatment on the surface of the Water intake calculation for single mice: Δpww (g) =Δww/N, N being the number of animals per group.
4. Gustatory disorder ratio= (drinking water is quinine hydrochloride aqueous solution and simultaneously administration of a solvent control group delta pWW with a test drug group delta pWW-drinking water is quinine hydrochloride aqueous solution)/(drinking water is ultrapure water and simultaneously administration of a solvent control group delta pWW-drinking water is quinine hydrochloride aqueous solution and simultaneously administration of a solvent control group delta pWW) ×100%. Data processing was performed using Excel et al software.
5. Animals were euthanized after the end of the experiment.
4. Test results:
table 6: example Compound taste results
5. Conclusion of experiment:
from the above results, it can be seen that the compound of example 32 of this patent has a significantly smaller toxic and side effect on the taste of mice at the same dose as the reference compound BLU-5937.

Claims (9)

1. A compound as shown below or a pharmaceutically acceptable salt thereof:
2. an intermediate compound represented by the formula (IV-1) and a pharmaceutically acceptable salt thereof,
3. A process for preparing a compound of formula (IV-1) as defined in claim 2 or a salt thereof, comprising the steps of,
deprotection of the general formula (IV-3) to give a compound of the formula (IV-1) or a salt thereof;
wherein:
Pg 1 is an amino protecting group selected from allyloxycarbonyl, trifluoroacetyl, 2, 4-dimethoxybenzyl, nitrobenzenesulfonyl, trityl, benzyloxycarbonyl, p-toluenesulfonyl, formate, acetyl, benzyloxycarbonyl, t-butoxycarbonyl, benzyl or p-methoxyphenyl.
4. A process for preparing a compound of formula (IV-1) as defined in claim 2 or a salt thereof according to claim 3,
Pg 1 is tert-butyloxycarbonyl.
5. A pharmaceutical composition comprising a therapeutically effective amount of a compound of claim 1 or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, or excipients.
6. Use of a compound according to claim 1 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 5, for the preparation of a P2X3 inhibitor drug.
7. Use of a compound according to claim 1 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 5, for the manufacture of a medicament for the treatment of a neurogenic disease; wherein the neurogenic disease is selected from a gynaecological disease, a urinary tract disease state, a respiratory disorder disease or a pain related disease or condition.
8. The use according to claim 7, wherein the disease is selected from endometriosis, overactive bladder, pulmonary fibrosis or chronic cough.
9. The use according to claim 7, wherein the pain-related disease or disorder is selected from neuropathic pain or uterine fibroid-related pain and discomfort.
CN202010852140.7A 2019-08-21 2020-08-21 Heterocyclic derivative inhibitor, preparation method and application thereof Active CN112409331B (en)

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
CN2019107749269 2019-08-21
CN201910774926 2019-08-21
CN201911061711 2019-11-01
CN2019110617119 2019-11-01
CN201911237094 2019-12-05
CN2019112370943 2019-12-05
CN202010280272 2020-04-10
CN2020102802727 2020-04-10
CN2020103286966 2020-04-23
CN202010328696 2020-04-23

Publications (2)

Publication Number Publication Date
CN112409331A CN112409331A (en) 2021-02-26
CN112409331B true CN112409331B (en) 2024-02-20

Family

ID=74854102

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010852140.7A Active CN112409331B (en) 2019-08-21 2020-08-21 Heterocyclic derivative inhibitor, preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN112409331B (en)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20180058741A (en) 2015-09-14 2018-06-01 인피니티 파마슈티칼스, 인코포레이티드 Solid form of isoquinolines, a process for their preparation, compositions comprising them and methods for using them
AU2020369568B2 (en) 2019-10-25 2024-03-21 Gilead Sciences, Inc. GLP-1R modulating compounds
CA3164134A1 (en) 2019-12-06 2021-06-10 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
MX2022007969A (en) 2019-12-23 2022-12-13 Blueprint Medicines Corp Inhibitors of mutant forms of egfr.
BR112022015858A2 (en) * 2020-02-14 2022-10-25 Bellus Health Cough Inc COMPOUND, PHARMACEUTICAL COMPOSITION, AND, METHODS TO TREAT A DISORDER ASSOCIATED WITH P2X3 ACTIVITY, TO TREAT PAIN, TO TREAT A URINARY TRACT DISORDER, TO REDUCE OR PREVENT UNCONTROLLED URINE LOSS, TO TREAT COUGH, TO TREAT ITCH, AND TO TREAT Endometriosis, pain associated with endometriosis and symptoms associated with endometriosis
WO2021244634A1 (en) * 2020-06-05 2021-12-09 武汉人福创新药物研发中心有限公司 Imidazopyridine compound and use thereof
CN113912601B (en) * 2020-07-10 2023-06-06 上海拓界生物医药科技有限公司 Novel imidazo [1,2-a ] pyridine derivatives, preparation method and medical application thereof
US11851419B2 (en) 2020-11-20 2023-12-26 Gilead Sciences, Inc. GLP-1R modulating compounds
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
IL305165A (en) 2021-02-19 2023-10-01 Sudo Biosciences Ltd Tyk2 inhibitors and uses thereof
CR20230495A (en) 2021-04-21 2023-11-30 Gilead Sciences Inc CARBOXYBENZIMIDAZOLIC LPG-IR MODULATING COMPOUNDS.
AR125966A1 (en) 2021-05-26 2023-08-30 Novartis Ag NEW BICYCLIC COMPOUNDS AND THEIR USES
IL308953A (en) 2021-06-04 2024-01-01 Vertex Pharma N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamides as modulators of sodium channels
CN115463133B (en) * 2021-06-10 2024-03-01 武汉朗来科技发展有限公司 Pharmaceutical composition, preparation method and application thereof
WO2022268218A1 (en) * 2021-06-24 2022-12-29 武汉朗来科技发展有限公司 Preparation method for heterocycloalkyl compound, and intermediate and application thereof heterocycloalkyl compound
CN115043836B (en) * 2021-08-20 2023-07-18 苏州璞正医药有限公司 P2X3 receptor selective modulator of imidazopyridine derivative and pharmaceutical application thereof
WO2023154766A1 (en) 2022-02-09 2023-08-17 Quanta Therapeutics, Inc. Kras modulators and uses thereof

Citations (4)

* 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
CN111377917A (en) * 2018-12-29 2020-07-07 武汉朗来科技发展有限公司 Heterocyclic compound, intermediate, preparation method and application thereof
CN111601601A (en) * 2017-09-18 2020-08-28 贝卢斯医疗咳嗽病公司 Selective P2X3 modulators
CN113164490A (en) * 2018-10-10 2021-07-23 贝卢斯医疗咳嗽病公司 Treatment of pruritis with P2X3 antagonists

Patent Citations (4)

* 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
CN111601601A (en) * 2017-09-18 2020-08-28 贝卢斯医疗咳嗽病公司 Selective P2X3 modulators
CN113164490A (en) * 2018-10-10 2021-07-23 贝卢斯医疗咳嗽病公司 Treatment of pruritis with P2X3 antagonists
CN111377917A (en) * 2018-12-29 2020-07-07 武汉朗来科技发展有限公司 Heterocyclic compound, intermediate, preparation method and application thereof

Also Published As

Publication number Publication date
CN112409331A (en) 2021-02-26

Similar Documents

Publication Publication Date Title
CN112409331B (en) Heterocyclic derivative inhibitor, preparation method and application thereof
US10556908B2 (en) Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
CN111484480B (en) Polycyclic derivative inhibitor, preparation method and application thereof
CN111936499B (en) Heterobicyclic inhibitors of MAT2A and methods for treating cancer
KR102447084B1 (en) Macrocyclic compounds and their use as TRK kinase inhibitors
TWI741423B (en) Heteroaromatic derivative regulator, its preparation method and application
WO2020211798A1 (en) Inhibitor containing bicyclic ring derivative, and preparation method and uses therefor
AU2007260851B2 (en) Purinone derivatives as HM74a agonists
AU2009211296B2 (en) (Dihydro)pyrrolo(2,1-a)isoquinolines
AU2014249192A1 (en) BET bromodomain inhibitors and therapeutic methods using the same
KR20020053094A (en) Fused heterotricyclic compounds, process for preparing the compounds and drugs containing the same
AU3406397A (en) Tricyclic benzazepine vasopressin antagonists
CA2713324A1 (en) Pyrrolopyrazine kinase inhibitors
WO2015095492A1 (en) Tricyclic heterocycles as bet protein inhibitors
CN112778311A (en) Nitrogen-containing fused ring derivative inhibitor, preparation method and application thereof
CN107690434B (en) Fused tricyclic imidazopyrazine derivatives as modulators of TNF activity
WO2022022680A1 (en) Nitrogen-containing fused ring derivative inhibitor, preparation method therefor and use thereof
CN113527299A (en) Nitrogen-containing condensed ring compounds, preparation method and application
WO2021115225A1 (en) Pyrazole-containing polycyclic derivative inhibitor, preparation method therefor and application thereof
WO1997049708A9 (en) Tricyclic benzazepine vasopressin antagonists
WO1997049708A1 (en) Tricyclic benzazepine vasopressin antagonists
JP2019512533A (en) Fused pentacyclic imidazole derivatives as modulators of TNF activity
WO2023036252A1 (en) Pyrrolopyrimidine or pyrrolopyridine derivative and medical use thereof
TW202214634A (en) Heterocyclic compound and derivative thereof
CN117412974A (en) Salt and crystal form containing pyrazole polycyclic derivative as well as preparation method and application thereof

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