CN116354935A - P38MAPK/MK2 pathway regulator, and composition, preparation method and application thereof - Google Patents

P38MAPK/MK2 pathway regulator, and composition, preparation method and application thereof Download PDF

Info

Publication number
CN116354935A
CN116354935A CN202211699892.XA CN202211699892A CN116354935A CN 116354935 A CN116354935 A CN 116354935A CN 202211699892 A CN202211699892 A CN 202211699892A CN 116354935 A CN116354935 A CN 116354935A
Authority
CN
China
Prior art keywords
compound
alkyl
reaction
mmol
halogen
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.)
Pending
Application number
CN202211699892.XA
Other languages
Chinese (zh)
Inventor
栾林波
姚元山
陈永凯
王朝东
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Meiyue Biotech Development Co Ltd
Original Assignee
Shanghai Meiyue Biotech Development Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Meiyue Biotech Development Co Ltd filed Critical Shanghai Meiyue Biotech Development Co Ltd
Publication of CN116354935A publication Critical patent/CN116354935A/en
Pending legal-status Critical Current

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
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers
    • 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 present disclosure discloses a compound as shown in formula I, its racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt or prodrug thereof, and its composition, preparation method and application. The compound has good regulation effect on the p38MAPK/MK2 pathway, good selectivity, good pharmacokinetics and other properties. Also, it is useful for treating diseases associated with the mediation of p38 kinase inhibitors, and for preparing medicaments for such conditions or diseases.

Description

P38MAPK/MK2 pathway regulator, and composition, preparation method and application thereof
The present disclosure claims priority from a prior application entitled "a p38MAPK/MK2 pathway modulator and compositions, methods of preparation and uses thereof", filed on day 29 12 of 2021 to the China national intellectual property agency, patent application No. 202111640112. X. The entirety of this prior application is incorporated by reference into this disclosure.
Technical Field
The present disclosure relates to the field of medicine, and in particular to a p38MAPK/MK2 pathway modulator, a composition, a preparation method and an application thereof.
Background
Biosignal transduction involves specific protein-protein interactions and post-translational modifications, regulating genetic and epigenetic processes to cope with the effects of the internal and external environment. Mitogen-activated protein kinase MAPK (mitogen-activated protein kinase) is a group of serine-threonine protein kinases that can be activated by different intracellular and external stresses, and is an important transmitter of signal transduction from the cell surface to the inside of the nucleus. Stress factors include cytokines, neurotransmitters, hormones, cellular stress, cell adhesion, and the like.
As a subfamily of the MAPK family, p38 MAPK, when activated, phosphorylates and activates downstream various protein kinases and transcription factors in response to the action of cells on external signals and inflammatory cytokines, thus exerting a complex biological effect. The p38 MAPK includes four members, p38α, p38β, p38γ, and p38δ. Among them, p38α is thought to play an important role in the signaling pathway of inflammatory processes, while the biological functions of other isoforms have not been completely discovered, but they are pleiotropic. Studies have shown that p38β plays an important role in the cytoprotective mechanism, whereas mitogen-activated protein kinase MKK3 (MAP Kinase Kinase 3) mediates p38 δ to have an effect on proliferation and survival of advanced colorectal cancer (CRC) cells. As an attractive target in the field of drug development, p38 MAPK has several inhibitor drugs into clinical studies, and no drug has been approved for marketing until now. According to published information, some candidate compounds fail during the clinical study phase, and the primary reasons for clinical failure include dose limiting to avoid toxicity ranging from insufficient exposure of drug molecules at the target site to down-regulation of anti-inflammatory pathways, redundancy of signal networks, or inhibition of key proteins involved in feedback regulation of other MAPK pathways, which may up-regulate other pro-inflammatory pathways, leading to increased inflammation. Thus, the development of a safe and effective p38 MAPK inhibitor is currently a major challenge in the art of drug development.
p38MAPK can regulate over 60 substrates and perform different physiological functions [ Cell 2013 (152), 924], so selectively inhibiting activation of p38MAPK downstream effectors is a major strategy to avoid side effects/insufficient efficacy due to overall inhibition of p38 MAPK. MAPK-activated protein kinase 2 (MAPK-activated protein kinase, MK 2) is a direct substrate downstream of p38MAPK and is activated by p38α and p38β. As the first discovered p38MAPK substrate, MK2 can regulate the expression of inflammatory factors at transcriptional and posttranscriptional levels, thereby playing an important role in the regulation of a number of inflammatory diseases. Studies have shown that MK2 can be used to increase expression of inflammatory factors such as TNF- α, IL-6, IL-8 and COX-2 by stabilizing AU-rich elements of mRNA. In a post-operative ileus model of mice [ The Journal of surgical research2013 (185), 102], MK2 inhibitors can reduce the expression of inflammatory factors MIP-1α, TNF- α, IL-6, IL-1β, and the like, while finding a reduction in polymorphonuclear leukocyte, mast cell, mononuclear macrophage infiltration and an improvement in intestinal smooth muscle contractility. In a mouse collagen-induced arthritis (CIA) model [ Journal ofimmunology 2006 (177), 1913], knock-out of the MK2 gene reduced the incidence of collagen-induced arthritis, decreased the incidence and severity of MK 2-/-and MK2 +/-mouse collagen-induced arthritis, and decreased the expression of inflammatory factors TNF-. Alpha.and IL-6 to varying degrees, as compared to wild-type mice. In the MK2 knockout hypercholesterolemia mouse model [ Circ Res 2007 (101), 1104], lipid deposition and macrophage reduction of the rat's aorta, and reduced expression of inflammatory factors such as VCAM-1 and MCP-1. In addition, studies have shown that inhibition of MK2 can be used in the development of antitumor drugs [ Cancer cell 2007 (11), 175].
Many diseases are associated with the p38MAPK/MK2 pathway, including, but not limited to, autoimmune and inflammatory diseases (e.g., rheumatoid arthritis, felicitous sweat gland, psoriasis, inflammatory bowel disease, atopic dermatitis, systemic lupus erythematosus, etc.), bone diseases, metabolic diseases, neurological and neurodegenerative diseases, cancer, cardiovascular diseases, allergies and asthma, alzheimer's disease, hormone-related diseases, and the like. The selective inhibition of the p38MAPK/MK2 pathway reduces the influence on other downstream pathways of the p38MAPK, thereby reducing the problems of potential toxic and side effects and insufficient drug effects in drug development; meets the unmet clinical need in the field of diseases associated with the p38MAPK/MK2 pathway.
Disclosure of Invention
The present disclosure provides a compound, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt or prodrug thereof, as shown in formula I:
Figure BDA0004023656070000021
wherein W is CH or N;
m is an integer of 0 to 5;
n is an integer of 0 to 3;
ring A is C 3-20 Cycloalkyl, 3-20 membered heterocyclyl, the carbon atom in ring a being attached to the parent nucleus, said 3-20 membered heterocyclyl containing 1, 2 or more O, N or S atoms;
R 1 Selected from H, halogen, CN and C 1-6 An alkyl group;
R 2 selected from-OR 81 、-NH-C(O)R 82 、-NHR 83 and-C (O) NHR 84
R 3 Selected from H, C 1-10 Alkyl and C 3-20 Cycloalkyl;
R 4 selected from H, halogen and C 1-10 An alkyl group;
R 5 are each independently selected from H, halogen, -OH, -C 1-6 Alkyl, -C 1-6 Alkoxy, oxo (=o), -C (O) C 1-6 Alkyl, -C (O) OH, -C (O) NR 91a R 91b 、-S(O) 2 R 92 and-S (O) 2 NR 93a R 93b
R 6 Selected from H, halogen and methyl;
R 7 independently selected from H, halogen, C unsubstituted or substituted by Ra 1-10 Alkyl and C 3-20 Cycloalkyl; ra is halogen or C 3-20 Cycloalkyl;
R 81 、R 82 、R 83 、R 84 identical or different, independently of one another, are selected from C which is unsubstituted or optionally substituted by 1, 2, 3, 4 or 5 Rb 6-14 aryl-C 1-10 Alkyl group5-14 membered heteroaryl-C 1-10 Alkyl, C 6-14 Aryl and 5-14 membered heteroaryl; each Rb is the same or different and is independently selected from halogen, halogenated C 1-10 Alkyl, C 1-10 Alkyl and C 1-10 An alkoxy group;
R 91a 、R 91b 、R 92 、R 93a 、R 93b identical or different, independently of one another, from H, C 1-6 Alkyl and C 3-20 Cycloalkyl groups.
According to an embodiment of the disclosure, W is CH or N; m is an integer of 0 to 5; n is an integer of 0 to 3;
ring A is C 3-20 Cycloalkyl, 3-20 membered heterocyclyl, the carbon atom in ring a being attached to the parent nucleus, said 3-20 membered heterocyclyl containing 1, 2 or more O, N or S atoms;
R 1 selected from H, halogen, CN and C 1-6 An alkyl group;
R 2 selected from-OR 81 、-NH-C(O)R 82 、-NHR 83 and-C (O) NHR 84
R 3 Selected from H, C 1-10 Alkyl and C 3-20 Cycloalkyl;
R 4 selected from H, halogen and C 1-10 An alkyl group;
R 5 are each independently selected from H, halogen, OH, C 1-6 Alkyl, C 1-6 Alkoxy, oxo (=o), -C (O) C 1-6 Alkyl, -C (O) OH, -C (O) NR 91a R 91b 、-S(O) 2 R 92 and-S (O) 2 NR 93a R 93b
R 6 Selected from H, halogen and methyl;
R 7 are independently selected from H, halogen, C 1-10 Alkyl and C 3-20 Cycloalkyl;
R 81 、R 82 、R 83 、R 84 identical or different, independently of one another, from C 6-14 aryl-C 1-10 Alkyl, 5-14 membered heteroaryl-C 1-10 Alkyl, C 6-14 Aryl and 5-14 membered heteroaryl; wherein C is 6-14 Aryl group5-to 14-membered heteroaryl groups are unsubstituted or optionally substituted with 1, 2, 3, 4 or 5 groups independently of one another selected from halogen, halogenated C 1-10 Alkyl, C 1-10 Alkyl and C 1-6 Alkoxy substitution;
R 91a 、R 91b 、R 92 、R 93a 、R 93b identical or different, independently of one another, from H, C 1-6 Alkyl and C 3-20 Cycloalkyl groups.
In accordance with an embodiment of the present disclosure,
w is CH or N;
m is 0, 1, 2, 3, 4 or 5;
n is 0, 1, 2 or 3;
ring A is C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, the carbon atom in ring a being attached to the parent nucleus, said 3-12 membered heterocyclyl containing 1, 2 or more O, N or S atoms;
R 1 selected from H, halogen, CN and-C 1-6 An alkyl group;
R 2 selected from-OR 81 、-NH-C(O)R 82 、-NHR 83 and-C (O) NHR 84
R 3 Selected from H, C 1-6 Alkyl and C 3-12 Cycloalkyl;
R 4 selected from H, halogen and C 1-6 An alkyl group;
R 5 are each independently selected from halogen, -OH, -C 1-6 Alkyl, -C 1-6 Alkoxy, oxo (=o), -C (O) C 1-6 Alkyl, -C (O) OH, -C (O) NR 91a R 91b 、-S(O) 2 R 92 and-S (O) 2 NR 93a R 93b
R 6 Selected from H, halogen and methyl;
R 7 are independently selected from H, halogen, C 1-6 Alkyl and C 3-12 Cycloalkyl;
R 81 、R 82 、R 83 、R 84 identical or different, independently of one another, from C 6-14 aryl-C 1-6 Alkyl, 5-14 membered heteroaryl-C 1-6 Alkyl, C 6-14 Aryl and 5-14 membered heteroaryl; wherein C is 6-14 Aryl, 5-14 membered heteroaryl is unsubstituted or optionally substituted with 1, 2, 3, 4 or 5 substituents independently of one another selected from halogen, halo C 1-6 Alkyl, C 1-6 Alkyl and C 1-3 Alkoxy substitution;
R 91a 、R 91b 、R 92 、R 93a 、R 93b identical or different, independently of one another, from H, C 1-3 Alkyl and C 3-10 Cycloalkyl groups.
In accordance with an embodiment of the present disclosure,
w is CH or N;
m is 0, 1, 2, 3, 4 or 5;
n is 0, 1, 2 or 3;
ring A is C 3-9 Cycloalkyl, 3-9 membered heterocyclyl, the carbon atom in ring a being attached to the parent, said 3-9 membered heterocyclyl containing 1, 2 or more O, N or S atoms;
R 1 is halogen;
R 2 selected from-OR 81 、-NH-C(O)R 82 、-NHR 83 and-C (O) NHR 84
R 3 Is C 1-3 Alkyl and C 3-6 Cycloalkyl;
R 4 is C 1-3 An alkyl group;
R 5 are each independently selected from halogen, -OH, -C 1-3 Alkyl, -C 1-3 Alkoxy, oxo (=o), -C (O) C 1-3 Alkyl, -C (O) OH, -C (O) NR 91a R 91b 、-S(O) 2 R 92 and-S (O) 2 NR 93a R 93b
R 6 Selected from H, halogen and methyl;
R 7 are each independently selected from H, halogen and C 1-3 An alkyl group;
R 81 、R 82 、R 83 、R 84 identical or different, independently of one another, from C 6-8 aryl-C 1-3 Alkyl, 5-6 membered heteroaryl-C 1-3 Alkyl, C 6-14 Aryl and 5-14 membered heteroaryl; which is a kind ofIn C 6-14 Aryl, 5-14 membered heteroaryl is unsubstituted or optionally substituted with 1, 2, 3, 4 or 5 substituents independently of one another selected from halogen, halo C 1-3 Alkyl, C 1-3 Alkyl and C 1-3 Alkoxy substitution;
R 91a 、R 91b 、R 92 、R 93a 、R 93b identical or different, independently of one another, from H, C 1-3 Alkyl and C 3-6 Cycloalkyl groups.
In accordance with an embodiment of the present disclosure,
w is CH or N;
m is 0, 1, 2 or 3;
n is 0 or 1;
ring A is selected from the group consisting of piperidinyl, tetrahydro-2H-pyranyl, tetrahydrofuranyl, oxetanyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 2-oxaspiro [3.3] heptyl, 2-oxaspiro [3.5] nonyl, 2-azaspiro [3.3] heptyl, 2-azaspiro [3.5] nonyl, azetidinyl, tetrahydropyrrolyl, thietanyl, tetrahydro-2H-thiopyranyl;
R 1 is Cl or Br;
R 2 selected from-OR 81 、-NH-C(O)R 82 、-NHR 83 and-C (O) NHR 84
R 3 Methyl or cyclopropyl;
R 4 is methyl;
R 5 are each independently selected from F, -OH, methyl, methoxy, oxo (= O), -C (O) C 1-3 Alkyl, -C (O) OH, -C (O) NH 2 、-C(O)NHCH 3 、-S(O) 2 CH 3 、-S(O) 2 CH 2 CH 3 and-S (O) 2 -cyclopropane;
R 6 selected from H, F and Cl;
R 7 is H;
R 81 、R 82 、R 83 、R 84 the same or different, independently of one another, are selected from phenylmethyl, pyridylmethyl, pyridylethyl, phenyl and pyridinyl, which are unsubstituted or optionally substituted by 1, 2 or 3 Rb; each Rb is identical to Or different from each other, independently selected from F, cl and CF 3
According to embodiments of the present disclosure, ring a may be selected from:
Figure BDA0004023656070000051
according to embodiments of the present disclosure, R 5 The structure formed with ring a may be selected from:
Figure BDA0004023656070000052
Figure BDA0004023656070000061
in a preferred embodiment, the compounds of formula I have the structure shown in formula Ia or Ib:
Figure BDA0004023656070000062
wherein R is 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 A, W, m, n have the definition set forth above, bolded chemical bonds indicate the presence of axial chirality in the compounds.
In a preferred embodiment, the compound of formula I has the structure of formula II:
Figure BDA0004023656070000071
wherein R is 1 、R 3 、R 4 、R 5 、R 6 、R 7 W, m, n and ring A independently of one another have the meanings indicated above;
R 10 selected from H, halogen, unsubstituted or optionally substituted with 1, 2 or more halogen, OH, NH 2 Substituted with the following groups: c (C) 1-10 Alkyl, C 1-10 Alkoxy, halo C 1-10 Alkyl, halogenated C 1-10 Alkoxy, C 2-10 Alkenyl, C 2-10 Alkenyloxy, C 2-10 Alkynyl, C 2-10 Alkynyl oxy;
each R 11 The same or different, independently of one another, from H, halogen, C 1-6 Alkyl, halogenated C 1-10 An alkyl group;
p is an integer of 0 to 4.
According to embodiments of the present disclosure, R 10 Selected from H, halogen, unsubstituted or optionally substituted with 1, 2 or more halogen, OH, NH 2 Substituted with the following groups: c (C) 1-6 Alkyl, C 1-6 Alkoxy, halo C 1-6 Alkyl, halogenated C 1-6 Alkoxy, C 2-6 Alkenyl, C 2-6 Alkenyloxy, C 2-6 Alkynyl, C 2-6 Alkynyl oxy;
each R 11 The same or different, independently of one another, from H, halogen, C 1-6 Alkyl, halogenated C 1-6 An alkyl group;
p is 0, 1, 2, 3 or 4.
According to embodiments of the present disclosure, R 10 Selected from H, halogen, C 1-3 Alkyl, halogenated C 1-3 An alkyl group; p is 0, 1 or 2;
each R 11 The same or different, independently of one another, from H, halogen, C 1-3 Alkyl, halogenated C 1-3 An alkyl group.
According to embodiments of the present disclosure, R 10 Selected from H and methyl; p is 0, 1 or 2;
each R 11 The same or different, are independently selected from F, cl, CF 3
In a more preferred embodiment, the compound of formula ii has formula la or formula lb:
Figure BDA0004023656070000081
wherein, the liquid crystal display device comprises a liquid crystal display device,R 1 、R 3 、R 4 、R 5 、R 6 、R 7 、R 10 、R 11 a, W, m, n, p have the definition set forth above, bolded chemical bonds indicate the presence of axial chirality of the compound.
In some embodiments, a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, or formula IIb, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt, or prodrug thereof, wherein W is N.
In some embodiments, a compound of formula I, formula Ia, or formula Ib, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt, or prodrug thereof, wherein R 2 is-OR 81 or-NHR 83 ;R 81 And R is 83 Identical or different, independently of one another, from C 6-8 aryl-C 1-3 Alkyl, 5-6 membered heteroaryl-C 1-3 Alkyl, C 6-8 Aryl and 5-6 membered heteroaryl; wherein C is 6-8 Aryl, 5-6 membered heteroaryl is unsubstituted or optionally substituted with 1, 2, 3, 4 or 5 substituents independently of one another selected from halogen, halo C 1-3 Alkyl, C 1-3 Alkyl and C 1-3 Alkoxy substitution; c (C) 1-3 The alkyl moiety is attached to O or NH.
In some embodiments, a compound of formula I, formula Ia, or formula Ib, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt, or prodrug thereof, wherein R 2 is-OR 81 ;R 81 Is C 6 aryl-C 1-3 Alkyl or 6-membered heteroaryl-C 1-3 An alkyl group; wherein C is 6 Aryl or 6 membered heteroaryl is unsubstituted or optionally substituted with 1, 2, 3, 4 or 5 substituents independently of one another selected from halogen, halo C 1-3 Alkyl, C 1-3 Alkyl and C 1-3 Alkoxy substitution; c (C) 1-3 The alkyl moiety is attached to O.
In some embodiments, a compound of formula I, formula Ia, formula Ib, formula II, formula IIa or formula IIb, racemates, stereoisomers, tautomers, isotopic labels, and,A solvate, pharmaceutically acceptable salt or prodrug thereof, wherein
Figure BDA0004023656070000082
Is->
Figure BDA0004023656070000083
R 5 Selected from halogen, -OH, -C 1-3 Alkyl, -C 1-3 Alkoxy, oxo (=o), -C (O) C 1-3 Alkyl, -C (O) OH, -C (O) NR 91a R 91b 、-S(O) 2 R 92 and-S (O) 2 NR 93a R 93b ,R 91a 、R 91b 、R 92 、R 93a 、R 93b Identical or different, independently of one another, from H, C 1-3 Alkyl and C 3-6 Cycloalkyl groups.
In some embodiments, a compound of formula I, formula Ia, formula Ib, formula II, formula IIa or formula IIb, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt or prodrug thereof, wherein
Figure BDA0004023656070000084
Is->
Figure BDA0004023656070000085
R 5 Selected from F, -OH, methyl, methoxy, -C (O) C 1-3 Alkyl, -C (O) OH, -C (O) NH 2 、-C(O)NHCH 3 、-S(O) 2 CH 3 、-S(O) 2 CH 2 CH 3 and-S (O) 2 -cyclopropane.
In some embodiments, the compound of formula I, formula Ia, formula Ib, formula II, formula IIa or formula IIb, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt or prodrug thereof does not include
Figure BDA0004023656070000091
In some embodiments, a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, or formula IIb, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt, or prodrug thereof, wherein R 1 Is halogen.
In some embodiments, a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, or formula IIb, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt, or prodrug thereof, wherein R 3 Is C 1-3 An alkyl group.
In some embodiments, a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, or formula IIb, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt, or prodrug thereof, wherein R 4 Is halogen or C 1-3 An alkyl group.
In some embodiments, a compound of formula I, formula Ia, formula Ib, formula II, formula IIa, or formula IIb, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt, or prodrug thereof, wherein R 6 Is H or halogen.
According to an embodiment of the present disclosure, the compounds of formula i have the following structure:
Figure BDA0004023656070000092
Figure BDA0004023656070000101
Figure BDA0004023656070000111
/>
Figure BDA0004023656070000121
/>
Figure BDA0004023656070000131
/>
Figure BDA0004023656070000141
/>
Figure BDA0004023656070000151
/>
Figure BDA0004023656070000161
/>
Figure BDA0004023656070000171
/>
Figure BDA0004023656070000181
/>
Figure BDA0004023656070000191
wherein the structural formulae of compounds 10 and 63 indicate the presence of a cis-trans structure therein and one of cis-or trans-form therein. According to an embodiment of the present disclosure, the compound of formula I has the following structure:
Figure BDA0004023656070000201
/>
Figure BDA0004023656070000211
wherein the compounds 10-P1 or 10-P2 and 63-P1 or 63-P2 are represented by the formula wherein a cis-trans structure is present and wherein cis or trans is present.
The present disclosure also provides a process for preparing a compound of formula i comprising:
scheme one: the compound a1 and the compound a2 undergo a coupling reaction to obtain the compound shown in the formula I.
The reaction formula is as follows:
Figure BDA0004023656070000221
wherein Y is Cl or Br; w, R 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 M, n and ring A have the above-described definitions independently of one another.
Scheme II: when W is N, R 7 In case of H, the compound b1 reacts with the compound b2 to obtain a compound of formula I;
the reaction formula is as follows:
Figure BDA0004023656070000222
wherein R is 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 M, n and ring A independently of one another have the definitions described above;
according to an embodiment of the present disclosure, the reaction is carried out in the presence of an inorganic base; the inorganic base is selected from one of sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide and potassium hydroxide.
According to embodiments of the present disclosure, when R 5 In the case of OH, OH in the compound b2 may be protected by a silicon protecting group, which may be tert-butyldiphenylsilyl; the silicon protecting group is removed during the reaction to give deprotected OH.
The present disclosure also provides the use of at least one of a compound of formula I, a racemate, a stereoisomer, a tautomer, an isotopic label, a solvate, a pharmaceutically acceptable salt, or a prodrug compound thereof, in the manufacture of a medicament.
According to embodiments of the present disclosure, the drug may be a drug for treating and/or preventing a disease associated with a p38 kinase inhibitor, for example may be an MK2 inhibitor or a p38 MAPK/MK2 pathway modulator.
The present disclosure also provides a pharmaceutical composition comprising a therapeutically effective amount of at least one of a compound of formula I, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt, or prodrug compound thereof.
According to an embodiment of the present disclosure, the pharmaceutical composition further comprises at least one pharmaceutically acceptable carrier.
According to embodiments of the present disclosure, the pharmaceutical composition may further contain one or more additional therapeutic agents.
The carrier includes disintegrants such as methylcellulose, sodium carboxymethylcellulose, calcium carboxymethylcellulose, croscarmellose sodium, polyvinylpyrrolidone, carboxypropylcellulose, starch and the like; lubricants including calcium stearate, zinc stearate, magnesium stearate, sodium stearyl fumarate, and the like; binders including gelatin, polyethylene glycol, sugars, gums, starches, hydroxypropyl cellulose, and the like; diluents including mannitol, xylitol, lactose, dextrose, sucrose, sorbitol and starch; surfactants include polysorbate 80, sodium lauryl sulfate, talc, and silica. The compositions of the present disclosure may be formulated so as to provide immediate, delayed or prolonged release of the active ingredient after administration to the patient by employing methods known in the art.
The present disclosure also provides the use of a compound of formula I, racemates, stereoisomers, tautomers, isotopic labels, solvates, pharmaceutically acceptable salts, or prodrug compounds thereof, in the treatment and/or prevention of a disease associated with the mediation of a p38 kinase inhibitor.
The present disclosure also provides methods of treating and/or preventing a disease associated with the mediation of a p38 kinase inhibitor comprising administering to a patient a therapeutically or prophylactically effective amount of at least one of a compound represented by formula I, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt, or prodrug compound thereof.
According to embodiments of the present disclosure, the disease may be a disease associated with the p38 MAPK/MK2 pathway, for example, autoimmune and inflammatory diseases (such as rheumatoid arthritis, felicitis, psoriasis, inflammatory bowel disease, atopic dermatitis, systemic lupus erythematosus, etc.), bone diseases, metabolic diseases, neurological and neurodegenerative diseases, cancer, cardiovascular diseases, allergies and asthma, alzheimer's disease, and hormone-related diseases.
Advantageous effects
The compounds of the present disclosure have good modulation of the p38 MAPK/MK2 pathway and have good selectivity. In addition, the compounds of the present disclosure have good pharmacokinetic properties. Also, the compounds of the present disclosure are useful in the treatment of diseases associated with the mediation of p38 kinase inhibitors, and in the preparation of medicaments for such conditions or diseases.
Definition and description of terms
Unless otherwise indicated, the radical and term definitions recited in the specification and claims of this disclosure, including as examples, exemplary definitions, preferred definitions, definitions recited in tables, definitions of specific compounds in the examples, and the like, may be arbitrarily combined and coupled with each other. Such combinations and combinations of radical definitions and compound structures should be understood to be within the scope of the disclosure and/or claims.
The numerical ranges recited in the specification and claims are equivalent to at least each specific integer number recited therein unless otherwise stated. For example, a numerical range "1-20" corresponds to each of the integer numbers recited in the numerical range "1-20," i.e., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20. Furthermore, when certain numerical ranges are defined as "numbers," it is to be understood that both endpoints of the range, each integer within the range, and each fraction within the range are delineated. For example, a "number of 0 to 10" should be understood to describe not only each integer of 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 and 10, but also at least the sum of each integer with 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, respectively.
It should be understood that in describing one, two or more herein, "more" shall mean an integer greater than 2, such as greater than or equal to 3, such as 3, 4, 5, 6, 7, 8, 9 or 10.
The term "halogen" means fluorine, chlorine, bromine and iodine.
The term "C 1-10 Alkyl "is understood to mean a straight-chain or branched saturated monovalent hydrocarbon radical having from 1 to 10 carbon atoms. For example, straight-chain and branched alkyl radicals having 1,2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms are denoted, "C 1-6 Alkyl "means straight and branched alkyl groups having 1,2, 3, 4, 5 or 6 carbon atoms. The alkyl group is, for example, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, 2-methylbutyl, 1-ethylpropyl, 1, 2-dimethylpropyl, neopentyl, 1-dimethylpropyl, 4-methylpentyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 2-ethylbutyl, 1-ethylbutyl, 3-dimethylbutyl, 2-dimethylbutyl, 1-dimethylbutyl, 2, 3-dimethylbutyl, 1, 3-dimethylbutyl, or 1, 2-dimethylbutyl, or the like, or an isomer thereof.
The term "alkoxy" refers to-O- (alkyl), wherein alkyl is as defined herein. Alkoxy groups (C) having preferably 1 to 12 (e.g. 1,2, 3, 4, 5, 6, 7, 8, 9, 10, 11 and 12) carbon atoms 1-12 Alkoxy), more preferably an alkoxy group having 1 to 6 carbon atoms (C 1-6 An alkoxy group). Non-limiting examples of alkoxy groups include: methoxy, ethoxy, propoxy and butoxy. Alkoxy groups may be substituted or unsubstituted.
The term "C 2-10 Alkenyl "is understood to mean preferably a straight-chain or branched monovalent hydrocarbon radical which contains one or more double bonds and has 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms, more preferably" C 2-8 Alkenyl groups). "C 2-10 Alkenyl "is understood to mean preferably a straight-chain or branched monovalent hydrocarbon radical which contains one or more double bonds and has 2, 3, 4, 5, 6, 7 or 8 carbon atoms, for example 2, 3, 4, 5 or 6 carbon atoms (i.e.C 2-6 Alkenyl) having 2 or 3 carbon atoms (i.e., C 2-3 Alkenyl). It will be appreciated that where the alkenyl group comprises more than one double bond, the double bonds may be separated from each other or conjugated. The alkenyl is, for example, vinyl, allyl, (E) -2-methylvinyl, (Z) -2-methylvinyl, (E) -but-2-enyl, (Z) -but-2-enyl, (E) -but-1-enyl, (Z) -but-1-enyl, pent-4-enyl, (E) -pent-3-enyl, (Z) -pent-3-enyl, (E) -pent-2-enyl, (E) -pent-1-enyl, (Z) -pent-1-enyl, hex-5-enyl, (E) -hex-4-enyl, (Z) -hex-4-enyl, (E) -hex-3-enyl, (Z) -hex-3-enyl, (E) -hex-2-enyl, (Z) -hex-1-enyl, isopropenyl, 2-methylprop-2-enyl, 1-methylprop-2-enyl, 2-methylprop-1-enyl, (E) -1-methylprop-1-enyl, (Z) -1-methylbut-1-enyl, 3-methylbut-3-enyl, 2-methylbut-3-enyl, 1-methylbut-3-enyl, 3-methylbut-2-enyl, (E) -2-methylbut-2-enyl, (Z) -2-methylbut-2-enyl, (E) -1-methylbut-2-enyl, (Z) -1-methylbut-2-enyl, (E) -3-methylbut-1-enyl, (Z) -3-methylbut-1-enyl, (E) -2-methylbut-1-enyl, (Z) -2-methylbut-1-enyl, (E) -1-methylbut-1-enyl, (Z) -1-methylbut-1-enyl, 1-dimethylprop-2-enyl, 1-ethylprop-1-enyl, 1-propylvinyl, 1-isopropylvinyl.
The term "C 2-10 Alkynyl "is understood to mean preferably a straight-or branched-chain monovalent hydrocarbon radical which contains one or more triple bonds and has 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms, for example 2, 3, 4, 5, 6, 7 or 8 carbon atoms (i.e." C 2-8 Alkynyl ") having 2, 3, 4, 5, or 6 carbon atoms (i.e.," C 2-6 Alkynyl ") having 2 or 3 carbon atoms (" C 2-3 Alkynyl "). The alkynyl group is, for example, ethynyl, prop-1-ynyl, prop-2-ynyl, but-1-ynyl, but-2-ynyl, but-3-ynyl, pent-1-ynyl, pent-2-ynylAlkynyl, pent-3-ynyl, pent-4-ynyl, hex-1-ynyl, hex-2-ynyl, hex-3-ynyl, hex-4-ynyl, hex-5-ynyl, 1-methylpropan-2-ynyl, 2-methylbutan-3-ynyl, 1-ethylprop-2-ynyl, 3-methylpent-4-ynyl, 2-methylpent-4-ynyl, 1-methylpent-4-ynyl, 2-methylpent-3-ynyl, 1-methylpent-3-ynyl, 4-methylpent-2-ynyl, 1-methylpent-2-ynyl, 4-methylpent-1-ynyl, 3-methylpent-1-ynyl, 2-ethylbut-3-ynyl, 1-ethylbut-2-ynyl, 1-prop-2-ynyl, 1-isopropyl-2-alkynyl, 2-dimethyl-but-3-ynyl, 1-dimethyl-but-3-ynyl, dimethyl-1-but-3-dimethyl-but, 1-dimethyl-3-but. In particular, the alkynyl group is ethynyl, prop-1-ynyl or prop-2-ynyl.
The term "C 3-20 Cycloalkyl "is understood to mean a saturated monovalent monocyclic, bicyclic (e.g. fused, spiro, bridged) hydrocarbon ring or tricyclic hydrocarbon ring having 3 to 20 carbon atoms, preferably" C ", a 3-12 Cycloalkyl ", more preferably" C 3-8 Cycloalkyl groups). The term "C 3-12 Cycloalkyl "is understood to mean a saturated monovalent monocyclic, bicyclic (e.g. bridged, spiro) hydrocarbon ring or tricyclic hydrocarbon ring having 3,4, 5, 6, 7, 8, 9, 10, 11 or 12 carbon atoms. The C is 3-12 Cycloalkyl can be a monocyclic hydrocarbon group such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl or cyclodecyl, or a bicyclic hydrocarbon group such as campholyl, indolyl, hexahydroindolyl, tetrahydronaphthyl, decahydronaphthyl, bicyclo [2.1.1 ]]Hexyl, bicyclo [2.2.1]Heptyl, bicyclo [2.2.1]Heptenyl, 6-dimethylbicyclo [3.1.1]Heptyl, 2, 6-trimethylbicyclo [3.1.1]Heptyl, bicyclo [2.2.2]Octyl, 2, 7-diazaspiro [3,5 ]]Nonylalkyl, 2, 6-diazaspiro [3,4 ]]Octyl, or tricyclic hydrocarbon groups such as adamantyl.
The term "3-20 membered heterocyclyl" refers to a saturated or unsaturated, non-aromatic ring or ring system, e.g., which is a 4-, 5-, 6-, or 7-membered monocyclic, 7-, 8-, 9-, 10-, 11-, 12-, 13-, or 14-membered bicyclic (e.g., parallel, spiro, bridged) or tricyclic ring system, and which contains up to One less, for example 1, 2, 3, 4, 5 or more heteroatoms selected from O, S and N, where N and S can also optionally be oxidized to various oxidation states to form nitrogen oxides, -S (O) -or-S (O) 2 -a state of the device. Preferably, the heterocyclic group may be selected from "3-12 membered heterocyclic groups". The term "3-12 membered heterocyclyl" means a saturated or unsaturated, non-aromatic ring or ring system and contains at least one heteroatom selected from O, S and N. The heterocyclic group may be attached to the remainder of the molecule through any of the carbon atoms or a nitrogen atom, if present. The heterocyclic group may include fused or bridged rings as well as spiro rings. In particular, the heterocyclic groups may include, but are not limited to: 3-membered rings, such as ethylene oxide; 4-membered rings such as azetidinyl, oxetanyl; a 5-membered ring such as tetrahydrofuranyl, dioxolyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl; or a 6 membered ring such as tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl or trithianyl; or a 7-membered ring such as diazepanyl. Optionally, the heterocyclyl may be benzo-fused. The heterocyclic group may be bicyclic, such as, but not limited to, a 5,5 membered ring, such as hexahydrocyclopenta [ c ] ]Pyrrol-2 (1H) -yl ring, or 5,6 membered bicyclic ring, e.g. hexahydropyrrolo [1,2-a ]]Pyrazin-2 (1H) -yl ring. The heterocyclic group may be partially unsaturated, i.e., it may contain one or more double bonds, such as, but not limited to, dihydrofuranyl, dihydropyranyl, 2, 5-dihydro-1H-pyrrolyl, 4H- [1,3,4]Thiadiazinyl, 1,2,3, 5-tetrahydrooxazolyl or 4H- [1,4]Thiazinyl, or it may be benzo-fused, such as, but not limited to, dihydroisoquinolinyl. When the 3-12 membered heterocyclic group is connected with other groups to form the compound disclosed by the invention, the carbon atom on the 3-12 membered heterocyclic group can be connected with other groups, or the heterocyclic atom on the 3-12 membered heterocyclic group ring can be connected with other groups. For example, when the 3-12 membered heterocyclic group is selected from piperazinyl, it may be that the nitrogen atom on the piperazinyl group is attached to other groups. Or when the 3-12 membered heterocyclic group is selected from piperidyl, it may be that the nitrogen atom on the piperidyl ring and the carbon atom at the para position thereof are attached to other groups.
The term "spiro" refers to a ring system in which two rings share 1 ring-forming atom.
The term "fused ring" refers to a ring system in which two rings share 2 ring-forming atoms.
The term "bridged ring" refers to a ring system in which two rings share more than 3 ring members.
The term "C 6-14 aryl-C 1-10 Alkyl "means C 6-14 Aryl substituted C 1-10 Alkyl, attachment site at C 1-10 An alkyl group.
The term "5-14 membered heteroaryl-C 1-10 Alkyl "refers to C substituted with a 5-14 membered heteroaryl group 1-10 Alkyl, attachment site at C 1-10 An alkyl group.
The term "aryl" refers to a 6 to 14 membered all-carbon monocyclic or fused polycyclic (fused polycyclic being a ring sharing adjacent pairs of carbon atoms) group having a conjugated pi-electron system, preferably 6 to 10 membered, such as phenyl and naphthyl. Such aryl rings include aryl rings fused to heteroaryl, heterocyclyl, or cycloalkyl rings as described herein, wherein the ring attached to the parent structure is an aryl ring, non-limiting examples of which include:
Figure BDA0004023656070000261
Figure BDA0004023656070000262
aryl groups may be substituted or unsubstituted.
The term "heteroaryl" refers to a heteroaromatic system containing 1 to 4 (e.g., 1, 2, 3, and 4) heteroatoms, 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur, and nitrogen. Heteroaryl is preferably 5 to 10 membered (e.g., 5, 6, 7, 8, 9, or 10 membered), more preferably 5 or 6 membered, such as furyl, thienyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, and the like. Such heteroaryl rings include heteroaryl fused to an aryl, heterocyclyl, or cycloalkyl ring as described herein, wherein the ring attached to the parent structure is a heteroaryl ring, non-limiting examples of which include:
Figure BDA0004023656070000263
Figure BDA0004023656070000264
Heteroaryl groups may be substituted or unsubstituted.
The terms "alkyl", "alkoxy", "cycloalkyl", "heterocyclyl", "aryl" and "heteroaryl" and the like herein may be substituted or unsubstituted; when substituted, it may be substituted at any available point of attachment, preferably independently optionally with one or more substituents selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl, the same or different.
Pharmaceutically acceptable salts of the compounds described in this disclosure may be inorganic salts or organic salts, which may form acid addition salts if the compounds have a basic center; if these compounds have an acidic center, they may form base addition salts; these compounds may also form internal salts if they contain both acidic (e.g., carboxyl) and basic (e.g., amino) centers.
The compounds of the present disclosure may exist in particular geometric or stereoisomeric forms. Such as cis and trans isomers, (-) -and (+) -pairs of enantiomers, (R) -and (S) -enantiomers, diastereomers, (D) -isomers, (L) -isomers, racemic and other mixtures, and enantiomerically or diastereomerically enriched mixtures, all of which are within the scope of the disclosure. Additional asymmetric carbon atoms may be present in substituents such as alkyl groups. All such isomers and mixtures thereof are included within the scope of the present disclosure.
In the chemical structure of the compounds of the present disclosure, the bond
Figure BDA0004023656070000265
Indicating no indicationFixed form, ->
Figure BDA0004023656070000266
Represents absolute configuration, i.e. bond +.>
Figure BDA0004023656070000267
Can be +.>
Figure BDA0004023656070000268
Or at the same time contain
Figure BDA0004023656070000269
Two configurations, I/O>
Figure BDA00040236560700002610
Indicating the presence of axial chirality.
Key with a key
Figure BDA00040236560700002611
Indicating unspecified configurations including cis (E) or trans (Z) configurations.
In addition, the compounds and intermediates of the present disclosure may also exist in different tautomeric forms, and all such forms are included within the scope of the disclosure. "tautomer" refers to structural isomers of different energies that can interconvert via a low energy barrier. For example, proton tautomers (also known as proton transfer tautomers) include tautomers via proton transfer, such as keto-enol isomerisation, imine-enamine isomerisation and lactam-lactam isomerisation. All tautomeric forms of all compounds in the present disclosure are within the scope of the present disclosure. The names of compounds named in a singular manner do not exclude any tautomers.
The present disclosure also includes some isotopically-labeled compounds of the present disclosure having the same structure as recited herein, but wherein one or more atoms are replaced by an atom having an atomic weight or mass number different from the atomic weight or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the present disclosure include hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, Isotopes of iodine and chlorine, such as 2 H、 3 H、 11 C、 13 C、 14 C、 13 N、 15 N、 15 O、 17 O、 18 O、 31 P、 32 P、 35 S、 18 F、 123 I、 125 I and 36 cl, and the like. All isotopic variations of the compounds of the present disclosure, whether radioactive or not, are intended to be encompassed within the scope of the present disclosure.
Unless otherwise indicated, when a position is specifically designated as deuterium (D), that position is understood to be deuterium (i.e., at least 10% deuterium incorporation) having an abundance that is at least 1000 times greater than the natural abundance of deuterium (which is 0.015%). The natural abundance of a compound in an example can be at least 1000 times greater than the abundance of deuterium, at least 2000 times greater than the abundance of deuterium, at least 3000 times greater than the abundance of deuterium, at least 4000 times greater than the abundance of deuterium, at least 5000 times greater than the abundance of deuterium, at least 6000 times greater than the abundance of deuterium, or higher than the abundance of deuterium. Each available hydrogen atom attached to a carbon atom may be independently replaced with a deuterium atom. Those skilled in the art are able to refer to the relevant literature for the synthesis of deuterated forms of the compounds. Commercially available deuterated starting materials may be used in preparing the deuterated form of the compound or they may be synthesized using conventional techniques using deuterated reagents including, but not limited to, deuterated borane, tridentate borane tetrahydrofuran solution, deuterated lithium aluminum hydride, deuterated iodoethane, deuterated iodomethane, and the like.
The term "therapeutically effective amount" of the present disclosure refers to that amount of active compound or drug substance that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, medical doctor or other clinician, which includes one or more of the following: (1) prevention of disease: for example, preventing a disease, disorder or condition in an individual who is susceptible to the disease, disorder or condition but has not experienced or developed a pathology or symptomatology of the disease. (2) inhibition of disease: for example, inhibiting a disease, disorder or condition (i.e., preventing further development of pathology and/or symptoms) in an individual experiencing or presenting with the pathology or symptoms of the disease, disorder or condition. (3) alleviation of disease: for example, alleviating a disease, disorder or condition (i.e., reversing the pathology and/or symptoms) in an individual experiencing or presenting with the pathology or symptoms of the disease, disorder or condition. For a drug or pharmacologically active agent, a "therapeutically effective amount" refers to a sufficient amount of the drug or agent that is non-toxic but achieves the desired effect. Determination of an effective amount varies from person to person, depending on the age and general condition of the recipient, and also on the particular active substance, a suitable effective amount in an individual case can be determined by one skilled in the art according to routine experimentation.
The present disclosure "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio, and are effective for the intended use.
The term "patient" in this disclosure refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, pigs, cattle, sheep, horses or primates, most preferably humans.
Detailed Description
The technical scheme of the present disclosure will be described in further detail below with reference to specific embodiments. It should be understood that the following examples are illustrative only and are not to be construed as limiting the scope of the present disclosure. All techniques implemented based on the foregoing disclosure are intended to be within the scope of the disclosure.
Unless otherwise indicated, the starting materials and reagents used in the following examples were either commercially available or may be prepared by known methods.
The structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) or/and Mass Spectrometry (MS). NMR shift (. Delta.) of 10 -6 Units of (ppm) are given. NMR was determined using Bruker ASCEND TM -400 nuclear magnetic resonance apparatus, the measuring solvent is deuterated dimethyl sulfoxide (DMSO-d 6 ) Deuterated chloroform (CDCl) 3 ) Deuterated methanol (CD) 3 OD), internal standard is Tetramethylsilane (TMS).
MS was measured using an Agilent 6110,Agilent 1100,Agilent 6120,AgilentG6125B liquid mass spectrometer.
HPLC was performed using an Shimadzu HPLC-2010C high performance liquid chromatograph (XBRIDGE 2.150 mm,3.5um column).
Chiral HPLC analysis assay using THARSFC X5.
The thin layer chromatography silica gel plate uses a Qingdao GF254 silica gel plate, the specification of the silica gel plate used by the Thin Layer Chromatography (TLC) is 0.15 mm-0.2 mm, and the specification of the thin layer chromatography separation and purification product is 0.4 mm-0.5 mm.
Column chromatography generally uses Qingdao ocean silica gel 200-300 mesh silica gel as a carrier.
High performance liquid phase preparation using Waters 2767, waters 2545, and innovative constant-pass LC3000 preparative chromatograph.
Chiral preparative column chromatography was used, shimadzu LC-20AP, THARSFC PREP.
The CombiFlash flash rapid prep instrument used CombiFlash Rf200 (teldyne ISCO).
The pressurized hydrogenation reaction uses a GCD-500G type hydrogen generator of Beijing Jia Wei Kokai technology.
The microwave reaction uses a Biotage initiator + type microwave reactor.
In the experimental examples, unless otherwise specified, the reaction was carried out under an argon atmosphere or a nitrogen atmosphere.
The argon or nitrogen atmosphere means that the reactor flask is connected to a balloon of argon or nitrogen of about 1 liter volume.
The hydrogen atmosphere is defined as the reaction flask being connected to a balloon of hydrogen gas having a volume of about 1 liter.
In the experimental example, if no special description exists, the reaction temperature is room temperature, and the temperature range is 20-30 ℃.
Asterisks in chemical formulas in the schemes indicate the presence of cis-trans isomerism at specific ring structure positions (as will be appreciated by those skilled in the art for substitution of the cycloalkane structure), exemplary cis-trans isomerism are as follows:
Figure BDA0004023656070000281
* In cis or trans formOne of the two.
It will be appreciated by those skilled in the art that resolved chiral compounds can be distinguished by the order of retention times in the chiral chromatographic column, and therefore chiral compounds resolved sequentially for retention times are correspondingly distinguished by the numbered suffixes P1, P2. Namely, the suffix P1 corresponds to the chiral structure which is split first, and the suffix P2 corresponds to the chiral structure which is split later. If absolute configurations of the compounds are listed in the reaction formula, the absolute configurations do not mean that the absolute configurations are represented by the compounds with the numbers P1 and P2, and the absolute configurations are represented by the two existing forms. The absolute configuration of the compounds numbered suffixes P1, P2 is subject to the absolute configuration that corresponds objectively to a particular retention time.
Synthesis of intermediate compound A-5
Figure BDA0004023656070000291
The first step: synthesis of Compound A-5b
Thionyl chloride (22.43 g,188.5 mmol) was slowly added dropwise to a solution of compound A-5a (20 g,125 mol) in ethanol (60 mL) and the reaction mixture was run at 60℃for 3 hours. After the reaction, the reaction mixture was directly concentrated under reduced pressure to remove the solvent, thereby obtaining a crude product of the compound A-5b (20 g), which was directly used for the next reaction. MS m/z (ESI) 187.9[ M+1 ]] +
And a second step of: synthesis of Compound A-5c
Compound A-5b (16 g,85.6 mmol) was dissolved in ethanol (60 mL) and sodium borohydride (6.48 g,171.2 mmol) was added slowly in portions to the solution at 0deg.C. The resulting mixture was stirred at room temperature for 3 hours. After the completion of the reaction, the solvent was removed by concentration under reduced pressure, and the obtained residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/5) to give compound a-5c (16 g, yield: 73.5%). MS m/z (ESI) 146.1[ M+1 ]] +
Third step, the synthesis of the compound A-5
Thionyl chloride (1.77 g,0.015 mol) was slowly added to dichloromethane (50 m) of compound A-5c (1.8 g,0.0124 mol) and N, N-dimethylformamide (5 drops) at room temperatureL) in solution, the reaction mixture was carried out at room temperature for 1 hour. After the completion of the reaction, an ammonium chloride solution (100 mL,4 m) was added to the reaction mixture to adjust the pH to neutrality, then water (20 mL) was added and extracted with methylene chloride (10 ml×3), the combined organic phases were dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure to remove the solvent, and the obtained residue was purified by column chromatography (petroleum ether/ethyl acetate=1/0 to 5/1) to give compound a-5 (1.8 g, yield: 84.68%). 1 H NMR(400MHz,CDCl 3 )δ8.35(d,J=2.4Hz,1H),7.26(ddd,J=9.1,8.0,2.6Hz,1H),4.72(d,J=2.1Hz,2H).
Synthesis of intermediate compound a:
Figure BDA0004023656070000292
the first step: synthesis of Compound A-2
A solution of lithium bis (trimethylsilyl) amide in tetrahydrofuran (141 mL,141 mmol) was slowly added to a solution of Compound A-1 (20 g,141 mmol) in tetrahydrofuran (500 mL) at-78deg.C, and acetyl chloride (6.6 g,844 mmol) was slowly added dropwise after stirring the reaction solution at-78deg.C for 1 hour, and the resulting mixture was further stirred at-78deg.C for 1 hour. After completion of the reaction, the reaction mixture was slowly poured into a saturated aqueous ammonium chloride solution (500 mL) and extracted with ethyl acetate (300 mL. Times.3). The combined organic phases were washed with saturated brine (300 ml×2), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (petroleum ether/ethyl acetate=2/1) to give compound a-2 (6.6 g, yield: 30%). MS m/z (ESI): 185.1[ M+1 ]] +
And a second step of: synthesis of Compound A-4
A solution of compound A-2 (8.98 g,48.7 mmol) and compound A-3 (4.63 g,32.5 mmol) in 1, 4-dioxane (150 mL) was heated to 90℃and stirred for 3.5 h. After the reaction mixture was cooled to room temperature, methanesulfonic acid (3.12 g,32.5 mmol) was added, and the reaction was heated to 50℃and stirred for 3 hours. After the completion of the reaction, the reaction mixture was cooled to room temperature and filtered, and the cake was collected and dried to give Compound A-4 (5.6 g, yield: 69%). MS m/z (ESI): 251.0[ M+1 ] ] +
And a third step of: synthesis of Compound A-6
Compound A-5 (4.01 g,24.6 mmol) was added to a mixture of compound A-4 (5.6 g,22.3 mmol), potassium carbonate (7.69 g,55.7 mmol) and 18-crown-6 (1.18 g,4.4 mmol) in N, N-dimethylformamide (80 mL), and the reaction was stirred at room temperature for 16 hours. After completion of the reaction, the reaction mixture was poured into water (100 mL) and extracted with ethyl acetate (80 mL. Times.3). The combined organic phases were washed with brine (20 mL. Times.3), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure to give crude compound A-6 (8.4 g, yield 80%) which was used directly in the next reaction. MS m/z (ESI): 378.0[ M+1 ]] +
Fourth step: synthesis of Compound A-8
Bis triphenylphosphine palladium dichloride (1.56 g,2.22 mmol) was added to a solution of compound A-6 (8.4 g,22.2 mmol) and tributyl (1-ethoxyethylene) tin (compound A-7) (10.21 g,24.2 mmol) in 1, 4-dioxane (100 mL), and the reaction was heated to 130℃and stirred for 4 hours. The reaction solution was then filtered, the filtrate was concentrated under reduced pressure, tetrahydrofuran (100 mL) was added to the residue to dissolve, and 5mL of concentrated hydrochloric acid was added dropwise thereto and stirred for 1 hour. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (ethyl acetate) to give Compound A-8 (5 g, yield: 60%). MS m/z (ESI): 386.0[ M+1 ] ] +
Fifth step: synthesis of Compound A-9
Acetic acid (2 mL) was added dropwise to a solution of Compound A-8 (5 g,13 mmol) and N-chlorosuccinimide (1.9 g,14.3 mmol) in isopropanol (100 mL), and the reaction was stirred at 60℃for 16 hours. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (ethyl acetate) to give Compound A-9 (3.6 g, yield: 70%). MS m/z (ESI) 420.0[ M+1 ]] +
Sixth step: synthesis of Compound A
N, N-dimethylformamide dimethyl acetal (0.85 g,7.2 mmol) was added to a solution of Compound A-9 (1.3 g,3.1 mmol) in N, N-dimethylformamide (15 mL), and the reaction mixture was stirred at 100deg.C for 3 hours. After completion of the reaction, the reaction mixture was poured into water (50 mL) and extracted with ethyl acetate(30 mL. Times.3). The combined organic phases were washed with saturated brine (30 ml×3), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (dichloromethane/methanol=50/1) to give compound a (900 mg, yield: 78%). MS m/z (ESI) 474.9[ M+H ]] +
Synthesis of intermediate compound B:
Figure BDA0004023656070000311
the first step: synthesis of Compound B-2
Diphenyl azide phosphate (23.5 g,0.085 mol) was added to a mixed solution of compound B-1 (10 g,0.057 mol) and triethylamine (17.3 g,0.17 mol) in t-butanol/toluene (50 mL/50 mL) and the reaction mixture was run at 110 ℃ for 16 hours. After the completion of the reaction, the reaction mixture was poured into water and extracted with methylene chloride (200 mL. Times.3). The combined organic phases were washed with water (30 mL), dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure to remove the solvent, and the resulting residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give compound B-2 (3.6 g, yield: 25%). MS m/z (ESI): 247.0[ M+1 ] ] +
And a second step of: synthesis of Compound B-3
Compound B-2 (3.6 g,14.5 mol) was added to a mixed solution of trifluoroacetic acid/dichloromethane (15 mL/30 mL), and the reaction mixture was stirred at room temperature for 16 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to remove the solvent, and the obtained residue was purified by column chromatography (dichloromethane/methanol=20/1) to give a crude product (3.1 g) of compound B-3. MS m/z (ESI): 147.0[ M+1 ]] +
And a third step of: synthesis of Compound B-4
Silver sulfate (6.61 g,0.02 mol) and iodine (5.38 g,0.02 mol) were added to a solution of compound B-3 (3.1 g,0.02 mol) in ethanol (50 mL), and the reaction was stirred at 50℃for 16 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to remove the solvent, and the obtained residue was purified by silica gel column chromatography (dichloromethane/methanol=10/1) to give compound B-4 (4.9 g, yield: 65%). MS m/z (ESI): 272.7[M+1] +
Fourth step: synthesis of Compound B-6
Methyl boric acid (530 mg,8.8 mmol), cesium carbonate (8.96 g,27.5 mmol) and [1,1' -bis (diphenylphosphino) ferrocene were reacted under nitrogen]Palladium dichloride (450 mg,0.55 mmol) was added sequentially to a solution of compound B-4 (1.5 g,5.5 mmol) in 1, 4-dioxane (30 mL) and the reaction mixture was allowed to proceed at 100deg.C for 1.5 hours. After the completion of the reaction, an aqueous sodium hydrogencarbonate solution was added to the reaction mixture to dilute it, extraction was performed with ethyl acetate (50 ml×3), the combined organic phases were washed once with saturated brine, dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure to remove the solvent, and the obtained residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=2/3) to give compound B-6 (0.43 g, yield: 48%). MS m/z (ESI): 161.0[ M+1 ] ] +
Fifth step: synthesis of Compound B-7
Compound B-6 (1.2 g,6.88 mmol) was added to a solution of compound A-5 (850 mg,5.29 mmol) in anhydrous 1, 4-dioxane (8 mL) and the reaction mixture was heated to 110℃and stirred at that temperature for 1 hour. After the reaction mixture was naturally cooled to 50℃and methanesulfonic acid (284 mg,2.96 mmol) was added thereto, the reaction was continued at 50℃for 1 hour. After completion of the reaction, water (50 mL) was added to the reaction mixture to dilute the mixture, and the mixture was extracted with ethyl acetate (100 mL. Times.3). The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate and filtered, the solvent was removed by concentrating the filtrate under reduced pressure, and the resulting residue was purified by silica gel column chromatography (dichloromethane) to give compound B-7 (530 mg, yield: 66%). MS m/z (ESI): 269.0[ M+1 ]] +
Sixth step: synthesis of Compound B-8
Potassium carbonate (1.14 g,8.28 mmol), 18-crown-6 (175 mg,0.66 mmol) was added to a solution of compound A-5 (702 mg,4.30 mmol) and compound B-7 (890 mg,3.31 mmol) in N, N-dimethylformamide (15 mL), and the reaction mixture was heated to 40℃and stirred at that temperature for 16 hours. After the completion of the reaction, the reaction mixture was cooled to room temperature, diluted with water (50 mL), extracted with ethyl acetate (50 mL. Times.3), and the combined organic phases were washed with saturated brine (10 mL) and dried over sulfur Sodium acid is dried and filtered, and the filtrate is concentrated under reduced pressure to give a crude product of compound B-8 (1.5 g, purity: 84%, yield: 96%), which is directly used for the next reaction. MS m/z (ESI): 395.8[ M+1 ]] +
Seventh step, synthesis of Compound B-9
Compound A-7 (2.01 g,5.55 mmol) was reacted with [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride (257 mg,0.37 mmol) was added to a solution of compound B-8 (1.45 g,3.70 mmol) in 1, 4-dioxane (20 mL) and the reaction mixture stirred at 130℃for 1.5 h. After the reaction was completed, the reaction solution was directly concentrated under reduced pressure to obtain a crude product of Compound B-9 (3.9 g), which was directly used in the next step. MS m/z (ESI): 431.9[ M+1 ]] +
Eighth step, the synthesis of the compound B-10
Compound B-9 (1.45 g,3.40 mmol) was added to a solution of tetrahydrofuran (15 mL) and concentrated hydrochloric acid (0.5 mL), and the reaction mixture was stirred at room temperature for 1 hour. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to remove the solvent, and the obtained residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=3/2) to give B-10 (910 mg, yield: 66%). MS m/z (ESI): 403.9[ M+1 ]] +
Ninth step: synthesis of Compound B-11
N-chlorosuccinimide (330 mg,2.48 mmol) and glacial acetic acid (0.2 mL) were added sequentially to a solution of compound B-10 (910 mg,2.25 mmol) in isopropanol (12 mL) and the reaction mixture was stirred at 60℃for 3 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to remove the solvent, and the obtained residue was purified by silica gel column chromatography (ethyl acetate) to give compound B-11 (1.13 g). MS m/z (ESI): 437.8[ M+1 ] ] +
Tenth step: synthesis of Compound B
N, N-dimethylformamide dimethyl acetal (600 mg,5.0 mmol) was added to a solution of Compound B-11 (1.08 g,2.5 mmol) in N, N-dimethylformamide (15 mL), and the reaction mixture was stirred at 100deg.C for 3 hours. After the completion of the reaction, the reaction mixture was cooled to room temperature, diluted with water (50 mL), and extracted with ethyl acetate (50 mL. Times.3). The combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate and filteredThe filtrate was concentrated under reduced pressure to remove the solvent, and the obtained residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to give compound B (900 mg, yield: 72%). MS m/z (ESI): 492.7[ M+1 ]] +
EXAMPLE 1 Synthesis of Compound 1
Figure BDA0004023656070000331
The first step: synthesis of Compounds 1-2
Compound A (200 mg,0.42 mmol) was added to a solution of compound 1-1 (191.5 mg,0.84 mmol) and potassium carbonate (232.9 mg,1.684 mmol) in N, N-dimethylformamide (2 mL). The reaction mixture was heated to 60 ℃ and stirred at that temperature for 18 hours. After completion of the reaction, the reaction mixture was cooled to room temperature, diluted with ethyl acetate (40 mL), and washed with saturated brine (20 mL. Times.5). The organic phase was dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (methanol/dichloromethane=1/10) to give compound 1-2 (200 mg, yield: 71%). MS m/z (ESI): 661.0[ M+23 ] ] +
And a second step of: synthesis of Compounds 1-3
Compounds 1-2 (280 mg) were added to a mixed solution of trifluoroacetic acid/dichloromethane (3 mL/6 mL), and the reaction mixture was stirred at room temperature for 16 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to remove the solvent, and the obtained residue was purified by column chromatography (methanol/dichloromethane=1:10) to give compound 1-3 (220 mg, yield: 84%). MS m/z (ESI): 538.7[ M+1 ]] +
And a third step of: synthesis of Compound 1
Paraformaldehyde (55 mg) and glacial acetic acid (0.1 mL) are added to a solution of compounds 1-3 (110 mg) in methanol (5 mL), the reaction is stirred at room temperature for 0.5 h, sodium cyanoborohydride (39 mg) is added and the reaction is continued at room temperature for 16 h. After the reaction, the reaction mixture was concentrated under reduced pressure to remove the solvent, and the residue was purified by high performance liquid chromatography (column: xbridge-C18; 150X 21.2mm,5 μm; column temperature: 25 ℃ C.; flow rate:20mL/min; wavelength: 214nm; column pressure: 80bar; mobile phase: acetonitrile-water (0.1% formic acid); gradient: 15-40%) to give compound 1 (25.5 mg, yield: 22%). MS m/z (ESI): 553.0[ M+1 ]] +1 H NMR(400MHz,CD 3 OD):δ8.90(d,J=5.2Hz,1H),8.85(s,1H),8.49(d,J=2.3Hz,1H),8.38(s,1H),8.31(d,J=5.2Hz,1H),7.80-7.72(m,1H),6.86(s,1H),5.54(d,J=1.6Hz,2H),3.42-3.33(m,2H),3.22-3.10(m,1H),2.84(t,J=11.2Hz,2H),2.70(s,3H),2.32-2.22(m,3H),2.20(s,3H),2.19-2.12(m,1H),2.08(s,3H)。
EXAMPLE 2 Synthesis of Compounds 2, 2-P1 and 2-P2
Figure BDA0004023656070000341
Acetic anhydride (51.15 mg,0.50 mmol) was added to a solution of compounds 1-3 (90 mg,0.16 mmol) and triethylamine (67.59, 0.67 mmol) in dichloromethane (8 mL) and the reaction stirred at room temperature for 12 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, diluted with water (20 mL), extracted with ethyl acetate (20 ml×3), the combined organic phases were washed with saturated brine (20 ml×3), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the resulting residue was purified by column chromatography (dichloromethane/methanol=10/1) and preparative HPLC (column: xbridge-Gemini-C18, 150×21.2mm,5 μm; mobile phase: acetonitrile-water (0.1% formic acid) gradient: 30-60%) to give compound 2 (43.8 mg, yield: 43.77%). MS m/z (ESI): 580.8[ M+H ] ] +1 H NMR(400MHz,CD 3 OD):δ8.88(d,J=5.3Hz,1H),8.83(s,1H),8.49(d,J=2.3Hz,1H),8.42(s,1H),8.29(dd,J=5.3,2.3Hz,1H),7.81-7.72(m,1H),6.84(s,1H),5.53(d,J=1.8Hz,2H),4.69-4.57(m,1H),4.06(d,J=11.8Hz,1H),3.31-3.16(m,2H),2.84(t,J=12.7Hz,1H),2.19(s,3H),2.17-2.05(m,8H),2.04-1.83(m,2H)。
Compound 2 was separated by supercritical fluid chromatography (apparatus: SFC Thar prep 80; column: CHIRALPAK AD-H,250mm x20 mm,5 μm; mobile phase: 40% methanol (methanol/carbon dioxide, 0.2% ammonia; total flow: 12.5 g/min)) to give compound 2-P1 (19.4 mg) and compound 2-P2 (18.6 mg).
Compound 2-P1:
MS m/z(ESI):581.1[M+H] + the method comprises the steps of carrying out a first treatment on the surface of the SFC: retention time = 4.08min, uv = 214nm; 1 H NMR(400MHz,CD 3 OD)δ8.83(d,J=5.2Hz,1H),8.79(s,1H),8.44(d,J=2.4Hz,1H),8.37(d,J=1.6Hz,1H),8.25(dd,J=5.3,2.3Hz,1H),7.71(ddd,J=9.6,8.6,2.4Hz,1H),6.80(s,1H),5.49(d,J=1.9Hz,2H),4.64-4.50(m,1H),4.06-3.97(m,1H),3.21(ddd,J=15.4,11.1,7.8Hz,2H),2.80(t,J=12.8Hz,1H),2.15(s,3H),2.11-2.05(m,5H),2.04(s,3H),2.01-1.86(m,2H)。
compound 2-P2:
MS m/z(ESI):581.1[M+H] + the method comprises the steps of carrying out a first treatment on the surface of the SFC: retention time = 5.51min, uv = 214nm; 1 H NMR(400MHz,CD 3 OD)δ8.83(dd,J=5.3,0.6Hz,1H),8.79(s,1H),8.44(d,J=2.4Hz,1H),8.36(s,1H),8.25(dd,J=5.3,2.2Hz,1H),7.76–7.66(m,1H),6.81(s,1H),5.49(d,J=2.0Hz,2H),4.58(ddd,J=13.0,5.4,3.2Hz,1H),4.00(dd,J=18.6,12.5Hz,1H),3.26-3.15(m,2H),2.87 -2.74(m,1H),2.15(s,3H),2.10(m,5H),2.04(m,3H),1.99-1.84(m,2H)。
EXAMPLE 3 Synthesis of Compounds 3 and 3-P1 and 3-P2
Figure BDA0004023656070000351
Compound 3-1 (134 mg,1.05 mmol) and potassium carbonate (218 mg,1.57 mmol) were added to a solution of compound A (250 mg,0.52 mmol) in N, N-dimethylformamide (5 mL). The reaction solution was heated to 60℃and stirred at that temperature for 16 hours. After completion of the reaction, the reaction mixture was poured into water and extracted with ethyl acetate (50 mL. Times.3). The combined organic phases were washed with saturated brine (50 ml×3), dried over anhydrous sodium sulfate and filtered, the solvent was removed by concentrating the filtrate under reduced pressure, and the residue was purified by high performance liquid chromatography (column: xbridge-C18, 150×21.2mm,5 μm; column temperature: 25 ℃ C.; flow rate: 20mL/min; wavelength: 214nm; column pressure: 80bar; mobile phase: acetonitrile-water (0.1% formic acid; gradient: 30-60%) to give compound 3 (121.2 mg, yield: 47%). MS m/z (ESI): 540.2[M+1] +1 H NMR(400MHz,DMSO-d6):δ8.90(d,J=5.2Hz,1H),8.83(s,1H),8.58(d,J=2.4Hz,1H),8.34(s,1H),8.17(d,J=5.2Hz,1H),8.11-8.04(m,1H),6.79(s,1H),5.45(d,J=1.5Hz,2H),3.96-3.87(m,2H),3.43(td,J=11.6,2.4Hz,2H),3.15-3.04(m,1H),2.05(s,3H),1.93(s,3H),1.91-1.79(m,4H)。
Compound 3 was resolved by supercritical fluid chromatography (apparatus: SFC Thar prep 80; column: chiralpak-AD; mobile phase: 40% isopropyl alcohol (isopropyl alcohol/carbon dioxide, 0.2% ammonia water), flow rate: 12.5 g/min) to give compound 3-P1 (62.1 mg, yield 21%) and compound 3-P2 (59.1 mg, yield 20%).
Compound 3-P1:
MS m/z(ESI):540.2[M+1] + the method comprises the steps of carrying out a first treatment on the surface of the SFC: retention time = 3.42min, uv = 214nm; 1 H NMR(400MHz,CD 3 OD):δ8.88(d,J=5.2Hz,1H),8.84(s,1H),8.49(d,J=2.2Hz,1H),8.41(s,1H),8.29(d,J=5.3Hz,1H),7.80-7.71(m,1H),6.86(s,1H),5.54(d,J=1.4Hz,2H),4.08(d,J=11.1Hz,2H),3.62(dd,J=17.9,6.7Hz,2H),3.28 -3.17(m,1H),2.20(s,3H),2.13-1.91(m,7H)。
compound 3-P2:
MS m/z(ESI):540.2[M+1] + the method comprises the steps of carrying out a first treatment on the surface of the SFC: retention time = 4.64min, uv = 214nm; 1 H NMR(400MHz,CD 3 OD):δ8.88(d,J=5.3Hz,1H),8.84(s,1H),8.49(d,J=2.3Hz,1H),8.41(s,1H),8.29(d,J=5.3Hz,1H),7.80-7.71(m,1H),6.85(s,1H),5.54(d,J=1.8Hz,2H),4.12-4.03(m,2H),3.62(dt,J=13.0,6.7Hz,2H),3.27-3.17(m,1H),2.20(s,3H),2.12-1.95(m,7H)。
EXAMPLE 4 Synthesis of Compound 4
Figure BDA0004023656070000352
Potassium carbonate (87.32 mg,0.63 mmol) and compound 4-1 (63.44 mg,0.42 mmol) were added to a solution of compound A (100 mg,0.21 mmol) in N, N-dimethylformamide (10 mL), and the reaction mixture was stirred at 80℃for 12 hours. After the reaction, the reaction solution was poured into water, acetic acidEthyl ester extraction (50 mL x 3), drying of the combined organic phases with saturated brine (50 mL x 3), drying over anhydrous sodium sulfate, concentration under reduced pressure to remove the solvent, and purification of the residue by high performance liquid chromatography (column: xbridge-C18, 150 x 21.2mm,5 μm; column temperature: 25 ℃, flow rate: 20mL/min; wavelength: 214nm; column pressure: 80bar; mobile phase: acetonitrile-water (0.1% formic acid; gradient: 35-60%) gave compound 4 (36.6 mg, yield: 31.7%). MS m/z (ESI): 525.8[ M+1 ] ] +1 H NMR(400MHz,CD 3 OD):δ8.83(dd,J=5.2,0.9Hz,1H),8.80(s,1H),8.44(d,J=2.4Hz,1H),8.37(d,J=1.5Hz,1H),8.26(dd,J=5.2,0.9Hz,1H),7.71(ddd,J=9.6,8.6,2.4Hz,1H),6.81(d,J=0.5Hz,1H),5.49(d,J=1.9Hz,2H),4.23-4.14(m,1H),4.133.99(m,2H),3.94-3.86(m,1H),3.85-3.76(m,1H),2.39(ddd,J=13.5,6.7,1.4Hz,2H),2.15(s,3H),2.04(d,J=0.7Hz,3H)。
EXAMPLE 5 Synthesis of Compound 6
Figure BDA0004023656070000361
Potassium carbonate (116 mg,0.84 mmol) and Compound 6-1 (62 mg,0.42 mmol) were added to a solution of Compound A (100 mg,0.21 mmol) in N, N-dimethylformamide (3 mL), and the reaction mixture was heated to 60℃and stirred at that temperature for 16 hours. After the reaction was completed, the reaction mixture was poured into water, extracted with ethyl acetate (50 mL. Times.3), the combined organic phases were washed with saturated brine (50 mL. Times.3), dried over anhydrous sodium sulfate and filtered, the solvent was removed from the filtrate by concentration under reduced pressure, and the residue was purified by high performance liquid chromatography (column: xbridge-C18, 150X 21.2mm,5 μm; column temperature: 25 ℃ C.; flow rate: 20mL/min; wavelength: 214nm; column pressure: 80bar; mobile phase: acetonitrile-water (0.1% formic acid); gradient: 50-70%) to give Compound 6 (35.3 mg; yield: 32%). MS m/z (ESI): 524.0[ M+1 ]] +1 H NMR(400MHz,DMSO-d 6 ):δ8.90(d,J=5.2Hz,1H),8.86(s,1H),8.61(d,J=2.4Hz,1H),8.31(s,1H),8.17(d,J=5.2Hz,1H),8.14-8.07(m,1H),6.83(s,1H),5.49(s,2H),3.40-3.33(m,1H),2.10(s,3H),2.08-2.00(m,2H),1.96(s,3H),1.94-1.86(m,2H),1.83-1.72(m,2H),1.71-1.59(m,2H)。
EXAMPLE 6 Synthesis of Compound 7
Figure BDA0004023656070000362
Potassium carbonate (186.25 mg,1.35 mmol) and compound 7-1 (81.2 mg,0.67 mmol) were added to a solution of compound A (160 mg,0.34 mmol) in N, N-dimethylformamide (2 mL), and the reaction mixture was heated to 60℃and stirred at that temperature for 18 hours. After the completion of the reaction, ethyl acetate (30 mL) was added to the reaction mixture to dilute it, which was washed with saturated brine (30 mL. Times.5), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the resulting residue was purified by high performance liquid chromatography (Gemini-C18, 150X 21.2mm,5 μm; mobile phase: acetonitrile-water (0.1% formic acid) =45-60%, UV:214 nm) to give compound 7 (43 mg, yield: 25.2%). MS m/z (ESI): 496.0[ M+1 ] ] +1 H NMR(400MHz,CD 3 OD)δ8.85(s,1H),8.80(d,J=5.2Hz,1H),8.61(d,J=2.4Hz,1H),8.32(s,1H),8.14-8.08(m,2H),6.83(s,1H),5.50(s,2H),2.33-2.24(m,1H),2.09(s,3H),1.96(s,3H),1.18-1.15(m,1H),1.12-1.06(m,3H)。
EXAMPLE 7 Synthesis of Compound 8
Figure BDA0004023656070000371
Potassium carbonate (87.32 mg,0.63 mmol) and Compound 8-1 (56.7 mg,0.42 mmol) were added to a solution of Compound A (100 mg,0.21 mmol) in N, N-dimethylformamide (10 mL), and the reaction was heated to 80℃and stirred at that temperature for 12 hours. After the reaction, the reaction mixture was poured into water, extracted with ethyl acetate (50 mL. Times.3), the combined organic phases were washed with saturated brine (50 mL. Times.3), dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure to remove the solvent, and the residue was purified by high performance liquid chromatography (column: xbridge-C18, 150X 21.2mm,5um; column temperature: 25 ℃ C.; flow rate: 20mL/min; wavelength: 214nm; column pressure: 80bar;mobile phase: acetonitrile-water (0.1% formic acid); gradient: 35-60%) to give compound 8 (52.9 mg, yield: 47.8%). MS m/z (ESI): 509.8[ M+1 ]] +1 H NMR(400MHz,CD 3 OD)δ8.83-8.78(m,2H),8.44(d,J=2.4Hz,1H),8.38(s,1H),8.22(d,J=5.3Hz,1H),7.71(ddd,J=9.6,8.6,2.4Hz,1H),6.80(d,J=0.5Hz,1H),5.49(d,J=2.0Hz,2H),3.91-3.79(m,1H),2.57-2.44(m,2H),2.41-2.31(m,2H),2.15(s,3H),2.13-2.06(m,1H),2.06-2.03(m,3H),1.99-1.89(m,1H)。
EXAMPLE 8 Synthesis of Compound 9
Figure BDA0004023656070000381
The first step: synthesis of Compound 9-2
Sodium borohydride (200 mg,5 mmol) was slowly added to a solution of compound 9-1 (1 g,10 mmol) in methanol (15 mL) under ice-bath. The reaction was stirred at room temperature for 3 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (petroleum ether/ethyl acetate=10/1) to give compound 9-2 (560 mg, yield: 57%). 1 H NMR(400MHz,CDCl 3 ):δ4.28-4.18(m,1H),2.79-2.69(m,2H),2.62-2.51(m,1H),2.37 -2.26(m,2H)。
And a second step of: synthesis of Compound 9-4
Compound 9-3 (1.56 g,5.68 mmol) was added to a solution of compound 9-2 (460 mg,4.74 mmol) and imidazole (640 mg,9.47 mmol) in dichloromethane (15 mL), and the reaction was stirred at room temperature for 16 hours. After the completion of the reaction, the reaction mixture was diluted with water (30 mL), extracted with methylene chloride (20 ml×3), the combined organic phases were washed with water (20 mL), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (petroleum ether/ethyl acetate=10/1) to give compound 9-4 (1.2 g, yield: 80%). MS m/z (ESI) 358.0[ M+23 ]] +
And a third step of: synthesis of Compound 9-5
Ethyl hydrochloride solution (2.0M, 5 mL) was added dropwise to methanol (27 mg,0.89 m) of Compound 9-4 (100 mg,0.29 mmol)mol) solution, the reaction mixture was stirred at room temperature for 16 hours, the reaction mixture was concentrated under reduced pressure, and an methanolic ammonia solution (7.0M, 5 mL) was added, and the reaction was continued at room temperature for 3 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to give crude compound 9-5 (41 mg, yield 39%) which was used directly in the next step. MS m/z (ESI) 353.0[ M+1 ]] +
Fourth step: synthesis of Compound 9
Potassium carbonate (30 mg,0.221 mmol) and Compound 9-5 (39 mg,0.110 mmol) were added to a solution of Compound A (35 mg,0.073 mmol) in N, N-dimethylformamide (5 mL), and the reaction was heated to 100deg.C and stirred at that temperature for 16 hours. After the completion of the reaction, the reaction mixture was poured into water (20 mL), extracted with ethyl acetate (20 ml×3), and the combined organic phases were washed with saturated brine (20 ml×3), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by thin plate chromatography (dichloromethane/methanol=10/1) to give compound 9 (8.7 mg, yield: 15%). MS m/z (ESI): 525.7[ M+1 ] ] +1 H NMR(400MHz,CD 3 OD):δ8.82-8.77(m,2H),8.46-8.41(m,2H),8.24(d,J=5.2Hz,1H),7.74-7.67(m,1H),6.82(s,1H),5.49(d,J=1.9Hz,2H),4.25-4.21(m,1H),3.26-3.24(m,1H),2.75-2.62(m,2H),2.51-2.31(m,2H),2.16(s,3H),2.05(s,3H)。
EXAMPLE 9 Compounds 10, 10-P1&Synthesis of Compound 10-P2
Figure BDA0004023656070000391
Figure BDA0004023656070000392
The first step: synthesis of Compound 10-2
Titanium tetrachloride (6.3 mL,6.3 mmol) and a methyllithium lithium chloride complex (2.0M, 3.2mL,6.3 mmol) were slowly added dropwise to a toluene (15 mL) solution of compound 10-1 (500 mg,5.2 mmol) at-5℃and the reaction system was warmed to room temperature naturally and stirred at room temperature for 3 hours. After the completion of the reaction, the reaction mixture was poured into a saturated aqueous ammonium chloride solution (30 mL) and quenched, followed by extraction with ethyl acetate (20 mL. Times.3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure to give crude compound 10-2 (230 mg), which was used directly in the next reaction.
And a second step of: synthesis of Compound 10-3
Aqueous hydroxylamine (50%, 2 mL) was added to a solution of compound 10-2 (160 mg,1.44 mmol) in ethanol (5 mL), the reaction was heated to 75 ℃ and stirred at that temperature for 12 hours, and after completion of the reaction, the reaction solution was directly concentrated under reduced pressure to give a crude product of compound 10-3 (300 mg), which was directly used in the next reaction. MS m/z (ESI): 145.1[ M+H ]] +
And a third step of: synthesis of Compound 10-4
Raney nickel (400 mg) was added to a solution of compound 10-3 (200 mg,1.39 mmol) in methanol (8 mL) at room temperature under nitrogen. The reaction was carried out under a hydrogen atmosphere at room temperature for 12 hours at normal pressure. After the reaction was completed, the reaction solution was directly filtered, and the filtrate was concentrated under reduced pressure to give a crude product of Compound 10-4 (160 mg), which was directly used for the next reaction. MS m/z (ESI): 129.1[ M+H ] ] +
Fourth step: synthesis of Compounds 10-P1 and 10-P2
Compound 10-4 (160 mg,1.11 mmol) and potassium carbonate (262 mg,1.89 mmol) were added to a solution of compound A (300 mg,0.63 mmol) in N, N-dimethylformamide (15 mL), and the reaction was heated to 90℃and stirred at that temperature for 12 hours. After the completion of the reaction, the reaction mixture was poured into water (30 mL), extracted with methylene chloride (20 mL. Times.3), the combined organic phases were washed with water (20 mL. Times.2), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by high performance liquid chromatography (column: gemini-C18, 150X 21.2mm,5 μm; mobile phase: acetonitrile-water (0.1% formic acid; gradient: 35-55%, column temperature: 25 ℃ C., flow rate: 14mL/min; wavelength: 214nm; column pressure: 80 bar) to give compound 10 (70 mg).
The compound 10 was subjected to chiral resolution by supercritical fluid chromatography (apparatus: SFC tharprep 80; column: CHIRALPAK AD-H250 mm. Times.20 mm,5 μm; mobile phase: 40% ethanol (ethanol/carbon dioxide, 0.2% ammonia water; flow rate: 12.5 g/min) to give compound 10-P1 (15.7 mg, yield: 4.6%) and 10-P2 (17.2 mg, yield: 5.1%).
Compound 10-P1:
MS m/z(ESI):539.8[M+1] + the method comprises the steps of carrying out a first treatment on the surface of the SFC: retention time = 2.66min, uv = 214nm; 1 H NMR(400MHz,CD 3 OD):δ8.79(d,J=5.1Hz,2H),8.48–8.42(m,2H),8.23(d,J=5.3Hz,1H),7.75–7.67(m,1H),6.81(s,1H),5.49(d,J=1.9Hz,2H),3.41–3.30(m,1H),2.63–2.50(m,2H),2.39-2.44(m,2H),2.15(s,3H),2.05(s,3H),1.44(s,3H)。
Compound 10-P2:
MS m/z(ESI):539.8[M+1] + the method comprises the steps of carrying out a first treatment on the surface of the SFC: retention time = 3.22min, uv = 214nm; 1 H NMR(400MHz,CD 3 OD):δ8.79(d,J=5.2Hz,2H),8.49–8.42(m,2H),8.23(d,J=5.3Hz,1H),7.68-7.72(m,1H),6.82(d,J=0.6Hz,1H),5.49(d,J=1.9Hz,2H),3.38–3.31(m,1H),2.55(dd,J=10.7,9.7Hz,2H),2.47–2.36(m,2H),2.15(s,3H),2.05(s,3H),1.44(s,3H)。
EXAMPLE 10 Synthesis of Compounds 14, 14-G1 and 14-G2
Figure BDA0004023656070000401
The first step: synthesis of Compound 14-2
Titanium tetrachloride (19.4 mL,19.40mmol, 1M) and methyllithium (9.7 mL,19.40mmol, 2M) were slowly added dropwise to a solution of compound 14-1 (2.0 g,16.20 mmol) in toluene (50 mL) at-5℃and after the dropwise addition the reaction was allowed to warm naturally to room temperature and stirred at room temperature for 3 hours. After the completion of the reaction, the reaction mixture was quenched with saturated ammonium chloride solution (20 mL), extracted with ethyl acetate (50 ml×3), the combined organic phases were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (ethyl acetate/petroleum ether=1/2) to give compound 14-2 (1.0 g, yield: 40%). 1 H NMR(400MHz,CDCl 3 )δ2.87–2.33(m,1H),2.05–1.80(m,2H),1.79–1.53(m,3H),1.39(m,3H),1.28–1.19(m,3H)。
Second step, synthesis of Compound 14-3
Aqueous hydroxylamine (50%, 0.5 mL) was added to a solution of compound 14-2 (200 mg,1.4 mmol) in ethanol (5 mL), the reaction was heated to 75 ℃ and stirred at that temperature for 16 hours, and after the completion of the reaction, the reaction solution was directly concentrated under reduced pressure to give crude compound 14-3 (300 mg), which was directly used in the next reaction. MS m/z (ESI): 173.1[ M+H ] ] +
And a third step of: synthesis of Compound 14-4
Raney nickel (40 mg) was added to a solution of compound 14-3 (300 mg,1.70 mmol) in methanol (10 mL) under nitrogen and the reaction was carried out at room temperature under an atmospheric hydrogen atmosphere for 16 hours. After the completion of the reaction, the reaction solution was filtered, and the filtrate was directly concentrated under reduced pressure to give a crude product of Compound 14-4 (300 mg), which was directly used in the next step. MS m/z (ESI): 157.1[ M+H ]] +
Fourth step: synthesis of Compounds 14-G1 and 14-G2
Compound 14-4 (200 mg,1.2 mmol) and potassium carbonate (34 mg,2.50 mmol) were added to a solution of compound A (304 mg,0.64 mmol) in N, N-dimethylformamide (10 mL), and the reaction was heated to 90℃and stirred at that temperature for 12 hours. After the completion of the reaction, the reaction mixture was poured into water (50 mL), extracted with methylene chloride (30 mL. Times.3), the combined organic phases were washed with water (20 mL. Times.2), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the resulting residue was purified by high performance liquid chromatography (column: gemini-C18, 150X 21.2mm,5um; mobile phase: acetonitrile-water (0.1% trifluoroacetic acid); gradient: 35-45%, column temperature: 25 ℃ C.; flow rate: 14mL/min; wavelength: 214nm; column pressure: 80 bar) to give two groups of compound 14-G1 (10 mg, yield: 1.20%) and compound 14-G2 (20 mg, yield: 2.34%). .
Compound 14-G1:
MS m/z(ESI):568.7[M+H] + the method comprises the steps of carrying out a first treatment on the surface of the HPLC: retention time = 5.00min, uv = 214nm; 1 H NMR(400MHz,CD 3 OD)δ8.92–8.81(m,2H),8.49(d,J=2.3Hz,1H),8.41(s,1H),8.30(d,J=5.3Hz,1H),7.81–7.70(m,1H),6.85(s,1H),5.53(d,J=1.8Hz,2H),3.02–2.93(m,1H),2.20(s,3H),2.11–2.01(m,5H),1.94–1.78(m,4H),1.67(dd,J=12.6,3.9Hz,2H),1.31(s,3H)。
compound 14-G2:
MS m/z(ESI):568.1[M+H] + the method comprises the steps of carrying out a first treatment on the surface of the HPLC: retention time = 5.31min, uv = 214nm; 1 H NMR(400MHz,CD 3 OD):δ8.91–8.82(m,2H),8.52–8.44(m,2H),8.33(d,J=5.4Hz,1H),7.81–7.70(m,1H),6.86(s,1H),5.54(d,J=1.8Hz,2H),2.92(ddd,J=12.6,7.8,3.3Hz,1H),2.23–2.04(m,8H),1.83(t,J=13.6Hz,4H),1.63–1.52(m,2H),1.26(s,3H)。
EXAMPLE 11 Synthesis of Compounds 16, 16-P1 and 16-P2
Figure BDA0004023656070000411
The first step: synthesis of Compound 16-2
Trimethylcyanosilane (11.89 g,119.8 mmol) and zinc iodide (0.82 g,2.56 mmol) were slowly added in sequence to a solution of compound 16-1 (6 g,85.6 mmol) in tetrahydrofuran (100 mL) under an ice bath. The reaction was naturally warmed to room temperature and stirred at room temperature for 40 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (petroleum ether/ethyl acetate=4/1) to give compound 16-2 (5.2 g, yield: 59.5%). 1 H NMR(400MHz,CDCl 3 )δ3.55(s,1H),2.71-2.57(m,2H),2.42-2.28(m,2H),2.05-1.88(m,2H)。
And a second step of: synthesis of Compound 16-3
Compound 16-2 (5.2 g,53.5 mmol) was added to ethanol hydrochloride (4M, 25 mL) and the reaction stirred at room temperature for 12 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the obtained residue was slurried with diethyl ether (20 mL), filtered, and the cake was collected and dried to give Compound 16-3 (2.8 g, yield: 32.9%). MS m/z (ESI): 144.0[ M+1 ]] +11 H NMR(400MHz,DMSO-d 6 ):δ11.13(d,J=111.0Hz,2H),4.54(q,J=7.0Hz,2H),2.43-2.20(m,2H),1.98-1.65(m,2H),1.49-1.31(m,3H)。
And a third step of: synthesis of Compound 16-4
Compound 16-3 (2.8 g,19.6 mmol) was added to an ethanol solution of ammonia (2M, 30 mL) and the reaction stirred at room temperature for 12 hours. After the completion of the reaction, the reaction mixture was filtered, the filtrate was concentrated under reduced pressure, and the obtained residue was slurried with diethyl ether (15 mL), filtered, and the cake was collected and dried to give Compound 16-4 (1.4 g, yield: 56.1%). MS m/z (ESI) 115.1[ M+1 ] ] +
Fourth step: synthesis of Compound 16
Compound 16-4 (144.21 mg,1.26 mmol) and potassium carbonate (261.92 mg,1.895 mmol) were added to a solution of compound A (300 mg,0.63 mmol) in N, N-dimethylformamide (5 mL) and the reaction stirred under microwave (90 ℃ C.) for 2 hours. After the completion of the reaction, the reaction mixture was poured into water (20 mL), extracted with ethyl acetate (20 ml×3), and the combined organic phases were washed with saturated brine (20 ml×3), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (dichloromethane/methanol=10/1) and high performance liquid chromatography (column: gemini-C18, 150×21.2mm,5um; mobile phase: acetonitrile-water (0.1% formic acid), gradient: 40-60%) to obtain compound 16 (60 mg). MS m/z (ESI) 525.8[ M+1 ]] +1 H NMR(400MHz,CD 3 OD):δ8.92(d,J=5.2Hz,1H),8.80(s,1H),8.51(s,1H),8.44(d,J=2.3Hz,1H),8.28(d,J=5.2Hz,1H),7.76-7.67(m,1H),6.81(s,1H),5.49(d,J=1.9Hz,2H),2.77-2.66(m,2H),2.37(dd,J=10.5,9.1Hz,2H),2.16(s,3H),2.04(d,J=5.9Hz,3H),2.02-1.94(m,2H)。
Compound 16 was purified by supercritical fluid chiral preparative chromatography (apparatus: SFC Thar prep 80;Column:CHIRALPAK AD-H250 mm. Times.20 mm,5 μm; mobile phase: 40% ethanol (ethanol/carbon dioxide, 0.2% ammonia; flow rate: 12.5 g/min)) to give compound 16-P1 (21.8 mg) and 16-P2 (22 mg).
Compound 16-P1:
MS m/z(ESI):525.8[M+1] +1 . Chiral HPLC: retention time = 6.36min, uv = 214nm. 1 H NMR(400MHz,CD 3 OD)δ8.92(d,J=5.2Hz,1H),8.80(s,1H),8.51(s,1H),8.44(d,J=2.4Hz,1H),8.28(d,J=5.2Hz,1H),7.71(ddd,J=9.7,8.6,2.4Hz,1H),6.81(d,J=0.6Hz,1H),5.49(d,J=2.0Hz,2H),2.77–2.67(m,2H),2.38(dt,J=11.8,8.8Hz,2H),2.16(s,3H),2.05(d,J=0.5Hz,3H),2.03–1.94(m,2H).
Compound 16-P2:
MS m/z(ESI):525.8[M+1] +1 . Chiral HPLC: retention time = 16.05min, uv = 214nm. 1 H NMR(400MHz,CD 3 OD)δ8.92(d,J=5.2Hz,1H),8.80(s,1H),8.51(s,1H),8.44(d,J=2.4Hz,1H),8.28(d,J=5.2Hz,1H),7.71(ddd,J=9.6,8.6,2.4Hz,1H),6.81(d,J=0.5Hz,1H),5.49(d,J=2.0Hz,2H),2.71(ddd,J=9.8,7.2,5.1Hz,2H),2.38(dd,J=9.3,3.4Hz,2H),2.16(s,3H),2.05(d,J=0.5Hz,3H),2.03–1.94(m,2H).
EXAMPLE 12 Synthesis of Compound 33, 33-P1 or 33-P2
Figure BDA0004023656070000431
The first step: synthesis of Compound 33-2
Trimethylnitrile silane (3.31 g,33.3 mmol) and zinc iodide (0.23 g,0.71 mmol) were added sequentially to a solution of compound 33-1 (2.0 g,23.8 mmol) in tetrahydrofuran (25 mL) under an ice bath. The reaction was naturally warmed to room temperature and stirred at room temperature for 24 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the obtained residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give compound 33-2 (1.5 g, yield: 32.7%). 1 H NMR(400MHz,CDCl 3 ):δ2.11-2.05(m,2H),2.04–1.96(m,2H),1.89–1.73(m,4H),0.25–0.22(m,9H).
And a second step of: synthesis of Compound 33-3
Compound 33-2 (1.5 g,8.2 mmol) was added to ethanol hydrochloride (4M, 15 mL) and the reaction stirred at room temperature for 12 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the obtained residue was slurried with diethyl ether (15 mL), filtered, and the cake was collected and dried to give compound 33-3 (0.8 g, yield: 58.5%). MS m/z (ESI): 158.1[ M+H ]] +
And a third step of: synthesis of Compound 33-4
Compound 33-3 (0.5 g,3.2 mmol) was added to an ethanol solution of ammonia (2M, 15 mL),the reaction was stirred at room temperature for 12 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the obtained residue was slurried with diethyl ether (15 mL), filtered, and the cake was collected and dried to give compound 33-4 (0.23 g, yield: 50.8%). MS m/z (ESI): 129.1[ M+H ] ] +
Fourth step, synthesis of Compound 33-P1 and Compound-P2
Compound 33-4 (162 mg,1.26 mmol) and potassium carbonate (262 mg,1.9 mmol) were added to a solution of compound A (300 mg,0.63 mmol) in N, N-dimethylformamide (15 mL), and the reaction was heated to 90℃and stirred at that temperature for 12 hours. After the completion of the reaction, the reaction mixture was poured into water (20 mL), extracted with ethyl acetate (20 ml×3), and the combined organic phases were washed with saturated brine (20 ml×3), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the resulting residue was purified by column chromatography (dichloromethane/methanol=10/1) and high performance liquid chromatography (column: gemini-c18×21.2mm,5um; mobile phase: acetonitrile-water (0.1% formic acid; gradient: 40-60%) to give compound 33.
Compound 33 was subjected to supercritical fluid chromatography (apparatus: SFC tharprep 80; column: CHIRALPAK AD-H,250 mm. Times.20 mm,5 μm; mobile phase: 40% isopropyl alcohol (isopropyl alcohol/carbon dioxide, 0.2% ammonia water; flow rate: 15 g/min)) to give compound 33-P1 (11.3 mg) and compound 33-P2 (14.2 mg).
Compound 33-P1:
MS m/z(ESI):539.8[M+H] + the method comprises the steps of carrying out a first treatment on the surface of the SFC: retention time = 6.67min, uv = 214nm; 1 H NMR(400MHz,CD 3 OD)δ8.92(d,J=5.2Hz,1H),8.84(s,1H),8.56(s,1H),8.48(d,J=2.4Hz,1H),8.30(d,J=5.2Hz,1H),7.81–7.71(m,1H),6.86(s,1H),5.53(d,J=1.9Hz,2H),2.42–2.26(m,2H),2.20(s,3H),2.10(s,3H),2.06–1.83(m,6H).
compound 33-P2:
MS m/z(ESI):539.8[M+H] + the method comprises the steps of carrying out a first treatment on the surface of the SFC: retention time = 11.99min, uv = 214nm; 1 H NMR(400MHz,CD 3 OD)δ8.80(d,J=5.2Hz,1H),8.72(s,1H),8.44(s,1H),8.36(d,J=2.3Hz,1H),8.17(d,J=5.2Hz,1H),7.64–7.61(m,1H),6.74(s,1H),5.41(d,J=1.9Hz,2H),2.42–2.23(m,2H),2.08(s,3H),1.98(s,3H),1.93–1.72(m,6H)。
EXAMPLE 13 Synthesis of Compounds 41, 41-P1 and 41-P2
Figure BDA0004023656070000441
The first step: synthesis of Compound 41-2
Aqueous hydroxylamine (50%, 5 mL) was added to a solution of 41-1 (5.0 g,1.4 mmol) in ethanol (5 mL), the reaction was heated to 75 ℃ and stirred at that temperature for 12 hours, and after the completion of the reaction, the reaction solution was directly concentrated under reduced pressure to give crude compound 41-2 (5.8 g), which was directly used in the next reaction. MS m/z (ESI) 160.1[ M+H ]] +
And a second step of: synthesis of Compound 41-3
Raney nickel (3.0 g) was added to a solution of compound 41-2 (5.8 g,27.9 mmol) in methanol (50 mL) under nitrogen and the reaction was carried out at room temperature under an atmospheric pressure of hydrogen for 12 hours. After the completion of the reaction, the reaction solution was filtered, and the filtrate was directly concentrated under reduced pressure to give a crude product of compound 41-3 (5.2 g), which was directly used for the next reaction. MS m/z (ESI) 200.1[ M+H ]] +
And a third step of: synthesis of Compound 41-4
Compound 41-3 (193 mg,0.96 mmol) and potassium carbonate (201 mg,1.45 mmol) were added to a solution of compound A (230 mg,0.48 mmol) in N, N-dimethylformamide (10 mL), and the reaction was heated to 90℃and stirred at that temperature for 12 hours. After the completion of the reaction, the reaction mixture was poured into water (50 mL), extracted with ethyl acetate (50 ml×3), and the combined organic phases were washed with saturated brine (20 ml×3), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by column chromatography (dichloromethane/methanol=10/1) to give compound 41-4 (90 mg, yield: 28%). MS m/z (ESI): 510.8[ M-Boc ] ] +
Fourth step: synthesis of Compound 41-5
Trifluoroacetic acid (84 mg,0.74 mmol) was slowly added to a dichloromethane solution (10 mL) of compound 41-4 (90 mg,0.15 mmol) and the reaction was carried out at room temperature for 12 hours. ReactionAfter completion, the reaction mixture was directly concentrated under reduced pressure to give crude compound 41-5 (70 mg), which was directly used for the next reaction. MS m/z (ESI): 510.8[ M+H ]] +
Fifth step: synthesis of Compound 41, compound 41-P1 and Compound 41-P2
Acetic anhydride (60 mg,0.59 mmol) was slowly added to a solution of compound 41-5 (100 mg,0.19 mmol) and triethylamine (59 mg,0.59 mmol) in dichloromethane (8 mL) and the reaction stirred at room temperature for 12 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, then poured into water (20 mL), extracted with ethyl acetate (20 mL. Times.3), the combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was subjected to high performance liquid chromatography (column: gemini-C18; 150X 21.2mm,5 μm; mobile phase: acetonitrile-water (0.1% formic acid), gradient: 30-60%, column temperature: 25 ℃ C., flow rate: 14mL/min, wavelength: 214nm, column pressure: 80 bar) to give compound 41 (50 mg).
Compound 41 was purified by supercritical fluid chromatography (apparatus: SFC Thar prep 80; column: CHIRALPAK AD-H250 mm. Times.20 mm,5 μm; mobile phase: 40% methanol (methanol/carbon dioxide, 0.2% ammonia; flow rate: 12.5 g/min)) to give compound 41-P1 (19 mg yield: 17.6%) and compound 41-P2 (17 mg yield: 15.7%).
Compound 41-P1
MS m/z(ESI):552.8[M+H] + The method comprises the steps of carrying out a first treatment on the surface of the SFC: retention time = 6.33min, uv = 214nm; 1 H NMR(400MHz,CD 3 OD)δ8.90(dd,J=5.2,4.3Hz,1H),8.81(s,1H),8.44(d,J=2.4Hz,1H),8.39(d,J=2.3Hz,1H),8.30(dd,J=5.3,1.1Hz,1H),7.71(ddd,J=9.6,8.6,2.4Hz,1H),6.80(s,1H),5.48(d,J=1.9Hz,2H),4.57(ddd,J=14.7,8.7,4.3Hz,2H),4.40–4.30(m,2H),4.16(ddd,J=8.8,5.9,2.8Hz,1H),2.15(s,3H),2.03(s,3H),1.87(s,3H)。
compound 41-P2
MS m/z(ESI):552.8[M+H] + The method comprises the steps of carrying out a first treatment on the surface of the SFC: retention time = 13.32min, uv = 214nm; 1 H NMR(400MHz,CD 3 OD)δ8.90(dd,J=5.2,4.4Hz,1H),8.81(s,1H),8.44(d,J=2.4Hz,1H),8.38(s,1H),8.31(dd,J=5.2,1.2Hz,1H),7.71(ddd,J=9.6,8.6,2.4Hz,1H),6.81(s,1H),5.49(d,J=1.9Hz,2H),4.57(ddd,J=14.7,8.8,4.1Hz,2H),4.40–4.30(m,2H),4.17(ddd,J=8.9,5.9,2.9Hz,1H),2.15(s,3H),2.04(s,3H),1.87(s,3H)。
example 14: synthesis of Compound 63, 63-P1 or 63-P2
Figure BDA0004023656070000461
The first step: synthesis of Compound 63
Compound 10-4 (104 mg,0.81 mmol) and potassium carbonate (168 mg,1.2 mmol) were added to a solution of compound B (200 mg,0.41 mmol) in N, N-dimethylformamide (15 mL), and the reaction was heated to 90℃and stirred at that temperature for 12 hours. After the completion of the reaction, the reaction mixture was poured into water (30 mL) and extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were washed with saturated brine (20 mL x 2), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the resulting residue was purified by high performance liquid chromatography (column: gemini-C18, 150X 21.2mm,5um; mobile phase: acetonitrile-water (0.1% formic acid; gradient: 35-65%, column temperature: 25 ℃ C.; flow rate: 14mL/min; wavelength: 214nm; column pressure: 80 bar) to give compound 63 (65 mg).
Compound 63 was purified by supercritical fluid preparation chromatography (apparatus: SFC Thar prep 80, column: CHIRALPAK AD-H250 mm. Times.20 mm,5 μm; mobile phase: 40% ethanol (ethanol/carbon dioxide, 0.2% ammonia water), total flow rate: 40 g/min) to give compound 63-P1 (25.3 mg, yield: 11.2%) and compound 63-P2 (26.7 mg, yield: 11.8%).
Compound 63-P1:
MS m/z(ESI):557.8[M+1] + the method comprises the steps of carrying out a first treatment on the surface of the SFC: retention time = 2.90min, uv = 220nm; 1 H NMR(400MHz,CD 3 OD)δ8.89(d,J=5.3Hz,1H),8.73(d,J=0.5Hz,1H),8.48(d,J=2.3Hz,1H),7.98(d,J=4.8Hz,1H),7.75(ddd,J=9.6,8.6,2.4Hz,1H),6.91(s,1H),5.55(d,J=1.9Hz,2H),3.42(dd,J=17.0,8.6Hz,1H),2.59(dd,J=15.0,5.7Hz,2H),2.53–2.44(m,2H),2.25(s,3H),2.16(s,3H),1.48(s,3H).
compound 63-P2:
MS m/z(ESI):557.8[M+1] + the method comprises the steps of carrying out a first treatment on the surface of the SFC: retention time = 5.56min, uv = 220nm; 1 H NMR(400
MHz,CD 3 OD)δ8.77(d,J=5.3Hz,1H),8.61(d,J=0.6Hz,1H),8.36(d,J=2.4Hz,1H),7.86(d,J=4.8Hz,1H),7.63(ddd,J=9.6,8.6,2.4Hz,1H),6.78(d,J=0.6Hz,1H),5.42(d,J=1.9Hz,2H),3.31(t,J=8.3Hz,1H),2.53–2.42(m,2H),2.41–2.32(m,2H),2.13(s,3H),2.04(s,3H),1.36(s,3H).
EXAMPLE 15 Synthesis of Compound 58
Figure BDA0004023656070000471
The first step: synthesis of Compound 58-1
Trifluoromethanesulfonic anhydride (2.37 g,8.4 mmol) is slowly added to a solution of compound A-4 (1.2 g,4.2 mmol) and triethylamine (1.27 g,12.6 mmol) in dichloromethane (20 mL). The reaction was stirred at room temperature for 12 hours. After the completion of the reaction, the solvent was removed by concentration under reduced pressure, and the obtained residue was purified by column chromatography (petroleum ether/ethyl acetate=1/5) to give compound 58-1 (0.35 g, yield: 64%). MS m/z (ESI) 416.9[ M+1 ]] +
And a second step of: synthesis of Compound 58-4
Compound 58-2 (205 mg,1.45 mmol) was added to a solution of compound 58-1 (500 mg,1.2 mmol) in 1, 4-dioxane (10 mL), the reaction was heated to 90℃and stirred at that temperature for 12 hours. After the completion of the reaction, the solvent was removed by concentration under reduced pressure, and the obtained residue was purified by column chromatography (petroleum ether/ethyl acetate=2/3) to give compound 58-3 (1.3 g, yield: 71.4%). MS m/z (ESI): 409.9[ M+1 ]] +
Third step, synthesis of Compound 58-4
Bis triphenylphosphine palladium dichloride (68 mg,0.1 mmol) was added to a solution of compound 58-3 (200 mg,0.48 mmol) and compound A-7 (704 mg,1.95 mmol) in 1, 4-dioxane/water (10 mL/1 mL), the reaction was heated to 110℃and stirred at that temperature for 3 hours. After the completion of the reaction, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to remove the solvent, and the obtained residue was purified by column chromatography (dichloromethane Methanol=5/1) to give compound 58-4 (0.11 g, yield: 48%). MS m/z (ESI): 446.1[ M+1 ]] +
Fourth step: synthesis of Compound 58-5
Compound 58-4 (120 mg,0.67 mmol) was dissolved in tetrahydrofuran/hydrochloric acid (5 mL/1 mL), and the reaction was stirred at room temperature for 3 hours. After the completion of the reaction, the solvent was removed by concentration under reduced pressure, and the obtained residue was purified by column chromatography (dichloromethane/methanol=10/1) to give compound 58-5 (0.1 g, yield: 80.4%). MS m/z (ESI) 418.1[ M+1 ]] +
Fifth step: synthesis of Compound 58-6
Compound 58-5 (100 mg,0.24 mmol) was added to a solution of N, N-dimethylformamide dimethyl acetal (42 mg,0.36 mmol) in N, N-dimethylformamide (10 mL), and the reaction was heated to 100deg.C and stirred at that temperature for 2 hours. After the completion of the reaction, the solvent was removed by concentration under reduced pressure, and the obtained residue was purified by column chromatography (dichloromethane/methanol=10/1) to give compound 58-6 (0.1 g, yield: 80%). MS m/z (ESI): 472.8[ M+1 ]] +
Sixth step: synthesis of Compound 71
Compound 58-7 (108 mg,0.84 mmol) and potassium carbonate (58 mg,0.42 mmol) were added to a solution of compound 58-6 (100 mg,0.21 mmol) in N, N-dimethylformamide (10 mL), and the reaction was heated to 90℃and stirred at that temperature for 16 hours. After the completion of the reaction, the solvent was removed by concentration under reduced pressure, and the obtained residue was purified by high performance liquid chromatography (column: gemini-C18, 150X121.2 mm,5um; mobile phase: acetonitrile-water (0.1% formic acid; gradient: 35-70%) to give compound 71 (9.1 mg, yield: 7.9%). MS m/z (ESI): 538.1[ M+1 ] ] +1 H NMR(400MHz,CD 3 OD):δ8.86(d,J=5.3Hz,1H),8.80(s,1H),8.34(s,1H),8.27(d,J=5.3Hz,1H),7.48-7.39(m,1H),7.07-6.98(m,2H),6.23(s,1H),4.67(s,2H),4.11-4.04(m,2H),3.66-3.57(m,2H),3.25-3.14(m,1H),2.19(s,3H),2.10-1.96(m,4H),1.94(s,3H)。
EXAMPLE 16 Synthesis of Compound 12
Figure BDA0004023656070000481
The first step: synthesis of Compound 12-2
Aqueous hydroxylamine (1 mL) was added to a solution of compound 12-1 (234 mg,2.0 mmol) in ethanol (5 mL), the reaction mixture was heated to 75℃and stirred at this temperature for 16 hours, and after completion of the reaction, the reaction solution was concentrated under reduced pressure to give compound 12-2 (260 mg, crude product). MS m/z (ESI) 151.0[ M+1 ]] +
And a second step of: synthesis of Compound 12-3
Raney nickel (508 mg) was added to a solution of compound 12-2 (260 mg,1.73 mmol) in methanol (10 mL), and the reaction mixture was stirred under hydrogen atmosphere at atmospheric pressure for 12 hours. After the reaction, the reaction solution was filtered, and the filtrate was concentrated under reduced pressure to give Compound 12-3 (180 mg, crude product): MS m/z (ESI): 135.1[ M+H ]] +
And a third step of: synthesis of Compound 12
Potassium carbonate (104 mg,0.75 mmol) was added to a solution of compound A (120 mg,0.25 mmol) and compound 12-3 (68 mg,0.50 mmol) in N, N-dimethylformamide (5 mL), and the reaction mixture was heated to 90℃and stirred for 12 hours. After completion of the reaction, the reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were washed with saturated brine (20 mL. Times.2), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the resulting residue was purified by high performance liquid chromatography (column: gemini-C18; 150X 21.2mm,5 μm; mobile phase: acetonitrile-water (0.1% formic acid); gradient: 40-50%; column temperature: 25 ℃ C.; flow rate: 20mL/min; wavelength: 214nm; column pressure: 80 bar) to give compound 12 (17.1 mg, yield 12.4%). MS m/z (ESI): 545.8[ M+1 ] ] +1 H NMR(400MHz,CD 3 OD)δ8.86(d,J=5.3Hz,1H),8.80(s,1H),8.44(d,J=2.4Hz,1H),8.38(s,1H),8.27(d,J=5.3Hz,1H),7.71(ddd,J=9.6,8.6,2.4Hz,1H),6.80(d,J=0.5Hz,1H),5.49(d,J=2.0Hz,2H),3.70–3.59(m,1H),3.08–2.91(m,4H),2.15(s,3H),2.04(d,J=0.5Hz,3H)。
EXAMPLE 17 Synthesis of Compound 37
Figure BDA0004023656070000491
Potassium carbonate (252 mg,1.82 mmol) was added to a solution of intermediate B (300 mg,0.61 mmol) and intermediate 16-4 (139 mg,1.2 mmol) in N, N-dimethylformamide (15 mL), and the reaction mixture was heated to 90℃under microwave conditions and stirred for 2 hours. After completion of the reaction, the reaction mixture was poured into water (20 mL) and extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were washed with saturated brine (20 ml×3), dried over anhydrous sodium sulfate and filtered, the filtrate concentrated under reduced pressure, and the residue was purified by column chromatography on silica gel (dichloromethane: methanol=1:0 to 10:1) to give crude product, which was purified by high performance liquid chromatography (column: gemini-C18;150×21.2mm,5 μm; mobile phase: acetonitrile-water (0.1% formic acid); gradient: 30-60%; column temperature: 25 ℃ C.; flow rate: 14mL/min; wavelength: 214nm; column pressure: 80 bar) to give compound 37 (17.7 mg, yield: 5.1%). MS m/z (ESI): 543.7[ M+1 ]] +1 H NMR(400MHz,CD 3 OD)δ8.96(d,J=5.2Hz,1H),8.70(s,1H),8.44(d,J=2.4Hz,1H),8.06(d,J=5.2Hz,1H),7.71(ddd,J=9.6,8.6,2.4Hz,1H),6.87(s,1H),5.50(d,J=1.9Hz,2H),2.75–2.66(m,2H),2.46–2.35(m,2H),2.21(s,3H),2.12(s,3H),2.03-1.90(m,2H)。
EXAMPLE 18 Synthesis of Compound 38
Figure BDA0004023656070000492
Potassium carbonate (405 mg3 mmol) was added to a solution of compound B (200 mg,0.4 mmol) and compound 33-4 (400 mg3 mmol) in N, N-dimethylformamide (15 mL), and the reaction mixture was heated to 90℃and stirred for 12 hours. After completion of the reaction, the reaction mixture was poured into water (20 mL) and extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were washed with saturated brine (20 ml×3), dried over anhydrous sodium sulfate and filtered, the filtrate concentrated under reduced pressure, and the residue purified by column chromatography on silica gel (dichloromethane: methanol=1:0-10:1) to give crude product, which was purified by high performance liquid chromatography (column: gemini-C18;150×21.2mm,5 μm; mobile phase: acetonitrile-water (0.1% formic acid; gradient: 40-60%; column temperature: 25 ℃ C.; flow rate: 14mL/min; wavelength: 214nm; column pressure: purification at 80bar afforded compound 38 (22 mg, 15% yield). MS m/z (ESI): 558.1[ M+1 ]] +1 H NMR(400MHz,CD 3 OD)δ8.98(d,J=5.3Hz,1H),8.75(s,1H),8.49(d,J=2.4Hz,1H),8.13(d,J=5.3Hz,1H),7.76(ddd,J=9.6,8.6,2.4Hz,1H),6.91(s,1H),5.55(d,J=1.9Hz,2H),2.41–2.30(m,2H),2.26(s,3H),2.16(s,3H),2.04–1.89(m,6H)。
EXAMPLE 19 Synthesis of Compound 42
Figure BDA0004023656070000501
Propionyl chloride (13 mg,0.14 mmol) was added dropwise to a solution of compound 41-5 (35 mg,0.068 mmol) and triethylamine (21 mg,0.21 mmol) in dichloromethane (5 mL) at 0deg.C and the reaction mixture was stirred at room temperature for 12 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to remove the solvent, and the residue was diluted with water (20 mL) and extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by high performance liquid chromatography (column: gemini-C18; 150X 21.2mm,5 μm; mobile phase: acetonitrile-water (0.1% formic acid); gradient: 35-36%; column temperature: 25 ℃ C.; flow rate: 14mL/min; wavelength: 214nm; column pressure: 80 bar) to give compound 42 (16.7 mg, yield: 42%). MS m/z (ESI): 556.8[ M+H ]] +1 H NMR(400MHz,CD 3 OD)δ9.01–8.89(m,1H),8.85(s,1H),8.49(d,J=2.4Hz,1H),8.43(d,J=1.7Hz,1H),8.35(dd,J=5.2,1.4Hz,1H),7.80–7.72(m,1H),6.84(d,J=1.9Hz,1H),5.53(d,J=1.8Hz,2H),4.60(dt,J=14.4,8.7Hz,2H),4.46–4.32(m,2H),4.26–4.14(m,1H),2.26–2.12(m,5H),2.07(d,J=1.7Hz,3H),1.10(dt,J=9.2,7.6Hz,3H)。
EXAMPLE 20 Synthesis of Compound 43
Figure BDA0004023656070000502
Isobutyryl chloride (25 mg,0.23 mmol) was added dropwise to compound 41-5 (60 mg,0.12 m) at 0deg.Cmol) and triethylamine (36 mg,0.35 mmol) in dichloromethane (5 mL), the reaction mixture was stirred at room temperature for 12 hours. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was diluted with water (20 mL) and extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by high performance liquid chromatography (column: gemini-C18; 150X 21.2mm,5 μm; mobile phase: acetonitrile-water (0.1% formic acid); gradient: 25-70%; column temperature: 25 ℃ C.; flow rate: 14mL/min; wavelength: 214nm; column pressure: 80 bar) to give compound 43 (16.7 mg, yield: 12.5%). MS m/z (ESI): 580.8[ M+H ] ] +1 H NMR(400MHz,CD3OD)δ8.94(dd,J=6.7,5.3Hz,1H),8.85(s,1H),8.49(d,J=2.2Hz,1H),8.41(d,J=4.1Hz,1H),8.35(d,J=5.2Hz,1H),7.81–7.72(m,1H),6.85(d,J=2.6Hz,1H),5.53(s,2H),4.68(t,J=8.6Hz,1H),4.60(dd,J=8.6,5.8Hz,1H),4.41(t,J=9.5Hz,1H),4.36–4.27(m,1H),4.25–4.13(m,1H),2.60(dd,J=13.6,6.8Hz,1H),2.20(d,J=1.8Hz,3H),2.07(d,J=2.0Hz,3H),1.11(dd,J=6.8,3.2Hz,3H),1.05(dd,J=14.0,6.8 3.2Hz,3H)。
EXAMPLE 21 Synthesis of Compound 44
Figure BDA0004023656070000511
The first step: synthesis of Compound 44-2
Aqueous hydroxylamine (0.5 mL) was added to an ethanol solution of compound 44-1 (200 mg,1.0 mmol), and the reaction mixture was heated to 80℃and stirred for 12 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to give Compound 44-2 (300 mg, crude product). MS m/z (ESI) 230.1[ M+H ]] +
And a second step of: synthesis of Compound 44-3
Raney nickel (30 mg) was added to a solution of compound 44-2 (300 mg,1.3 mmol) in methanol (10 mL), and the reaction mixture was stirred under hydrogen atmosphere at atmospheric pressure for 12 hours. After the completion of the reaction, the reaction solution was filtered, and the filtrate was concentrated under reduced pressure to give Compound 44-3 (300 mg, crude product). MS m/z (ESI) 214.1[ M+H ]] +
And a third step of: synthesis of Compound 44-4
Potassium carbonate (260 mg,1.9 mmol) was added to a solution of compound 44-3 (200 mg,0.93 mmol) and compound A (356 mg,0.75 mmol) in N, N-dimethylformamide (10 mL), and the reaction mixture was heated to 90℃and stirred for 12 hours. After completion of the reaction, the reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 mL. Times.3). The combined organic phases were washed with saturated brine (20 ml×3), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the obtained residue was purified by silica gel column chromatography (dichloromethane: methanol=1:0 to 10:1) to give compound 44-4 (90 mg, yield: 13%). MS m/z (ESI) 525.0[ M-100 ] ] +
Fourth step: synthesis of Compound 44-5
1, 4-Dioxahexacyclic solution of hydrogen chloride (4M, 2 mL) was added to a solution of compound 44-4 (70 mg,0.11 mmol) in dichloromethane (10 mL) at room temperature, and the reaction mixture was stirred at room temperature for 12 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to give Compound 44-5 (100 mg, crude product). MS m/z (ESI): 525.2[ M+H ]] +
Fifth step: synthesis of Compound 44
Acetic anhydride (20 mg,0.19 mmol) was added to a solution of compound 44-5 (70 mg,0.13 mmol) and triethylamine (27 mg,0.26 mmol) in dichloromethane (5 mL) at room temperature, and the reaction mixture was stirred at room temperature for 2 hours. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was diluted with water (20 mL) and extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the resulting residue was subjected to high performance liquid chromatography (column: gemini-C18; 150X 21.2mm,5 μm; mobile phase: acetonitrile-water (0.1% trifluoroacetic acid); gradient: 15-50%; column temperature: 25 ℃ C.; flow rate: 20mL/min; wavelength: 214nm; column pressure: 80 bar) to give compound 44 (10 mg, yield: 12.6%). MS M/z (ESI) [ M+H ]] +1 H NMR(400MHz,CD 3 OD)δ8.91(d,J=5.2Hz,1H),8.85(s,1H),8.49(dd,J=2.2,1.1Hz,1H),8.43–8.30(m,2H),7.80–7.71(m,1H),6.85(s,1H),5.54(s,2H),4.14–3.63(m,5H),2.56–2.30(m,2H),2.20(s,3H),2.08(dd,J=11.2,5.0Hz,6H)。
Synthesis of Compounds 66, 66-P1, 66-P2, 66-P3 and 66-P4 from example 22
Figure BDA0004023656070000521
The first step: synthesis of Compound 66-2
Compound 66-1 (3 g,23 mmol) was added to a solution of ammonia in ethanol (150 mL) and the reaction mixture stirred at 90℃for 12 h. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to give Compound 66-2 (3 g, yield: 90%). MS m/z (ESI) 130.3[ M+1 ]] +
And a second step of: synthesis of Compound 66-3
Para-toluenesulfonyl chloride (5.3 g,27.8 mmol) was added to a solution of compound 66-2 (3 g,23.2 mmol) and triethylamine (4.7 g,46 mmol) in dichloromethane (20 mL) and the reaction mixture was stirred at room temperature for 5 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane: methanol=1:0 to 10:1) to give compound 66-3 (5.2 g, yield: 71%). MS m/z (ESI) 284.0[ M+1 ]] +
And a third step of: synthesis of Compound 66-4
A solution of compound 66-3 (5.78 g,20.4 mmol) and sodium cyanide (2 g,40.8 mmol) in dimethyl sulfoxide (40 mL) was heated to 110℃and stirred at that temperature for 16 hours. After the completion of the reaction, the reaction mixture was quenched with water (150 mL), extracted with ethyl acetate (100 ml×3), the combined organic phases were dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure and purified by silica gel column chromatography (dichloromethane: methanol=1:0 to 10:1) to give compound 66-4 (400 mg, yield: 13%). 1 H NMR(400MHz,CD 3 OD)δ3.30–3.21(m,2H),2.68(s,3H),2.64–2.51(m,2H),2.50–2.42(m,2H)。
Fourth step: synthesis of Compound 66-5
Aqueous hydroxylamine (1 mL) was added to a solution of compound 66-4 (400 mg,2.89 mmol) in ethanol (10 mL) and the reaction mixture was stirred at 75deg.C for 16 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to give compound 66-5 (350 mg, yield: 56%). MS m/z (ESI): 172.1[ M+1 ]] +
Fifth step: synthesis of Compound 66-6
Raney nickel (1.2 g,20 mmol) was added to a solution of compound 66-5 (350 mg,2 mmol) and acetic acid (122 mg,2 mmol) in methanol (15 mL) and the reaction mixture was stirred at room temperature under a hydrogen atmosphere for 16 hours. After the completion of the reaction, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give compound 66-6 (300 mg, yield: 89%). MS m/z (ESI): 156.2[ M+1 ]] +
Sixth step: synthesis of Compound 66
Potassium carbonate (232 mg,1.68 mmol) was added to a solution of compound 66-6 (196 mg,1.26 mmol) and compound A (200 mg,0.42 mmol) in N, N-dimethylformamide (10 mL), and the reaction mixture was stirred at 90℃for 16 hours. After the reaction is finished, the reaction solution is filtered, the filtrate is concentrated under reduced pressure, the residue is purified by silica gel column chromatography (dichloromethane: methanol=1:0-10:1) to obtain crude product compound, the crude product is purified by supercritical fluid chiral chromatography (equipment: SFC Thar prep 80, column: CHIRALPAK AD-H250 mm x 20mm,5 μm, mobile phase: 40% EtOH/CO 2 (NH 4 OH 0.2%), total flow rate: 40 g/min) to give compound 66-P1 (16.3 mg, yield 6.8%), compound 66-P2 (7.4 mg, yield 3.1%), compound 66-P3 (14.7 mg, yield 6.1%) and compound 66-P4 (8.2 mg, yield 3.4%).
Compound 66-P1:
MS m/z(ESI):566.8[M+1] + . Supercritical fluid chromatography SFC: retention time = 7.2min, uv = 214nm. 1 HNMR(400MHz,CD 3 OD)δ8.73(d,J=5.3Hz,2H),8.40–8.34(m,2H),8.17(d,J=5.3Hz,1H),7.68–7.59(m,1H),6.74(s,1H),5.42(d,J=1.8Hz,2H),3.69–3.59(m,1H),3.01(ddd,J=17.8,9.6,8.3Hz,1H),2.67–2.54(m,7H),2.08(s,3H),1.98(s,3H)。
Compound 66-P2:
MS m/z(ESI):566.8[M+1] + . Supercritical fluid chromatography SFC: retention time = 7.9min, uv = 214nm. 1 H NMR(400MHz,CD 3 OD)δ8.88(d,J=5.3Hz,1H),8.85(s,1H),8.49(d,J=2.3Hz,1H),8.43(s,1H),8.29(d,J=5.3Hz,1H),7.81–7.72(m,1H),6.85(s,1H),5.53(d,J=1.8Hz,2H),3.91(dt,J=
14.8,7.2Hz,1H),3.31–3.24(m,1H),2.75–2.58(m,7H),2.20(s,3H),2.09(s,3H)。
Compound 66-P3:
MS m/z(ESI):566.8[M+1] + . Supercritical fluid chromatography SFC: retention time=8.56 min, uv=214 nm. 1 HNMR(400MHz,CD 3 OD)δ8.85(d,J=5.3Hz,2H),8.49(d,J=2.4Hz,1H),8.47(s,1H),8.29(d,J=5.3Hz,1H),7.78–7.72(m,1H),6.86(s,1H),5.54(d,J=1.9Hz,2H),3.82–3.72(m,1H),3.18–3.09(m,1H),2.73–2.58(m,7H),2.20(s,3H),2.10(s,3H)。
Compound 66-P4:
MS m/z(ESI):566.8[M+1] + . Supercritical fluid chromatography SFC: retention time = 9.8min, uv = 214nm.1HNMR (400 MHz, CD) 3 OD)δ8.76(d,J=5.2Hz,1H),8.73(s,1H),8.37(d,J=2.4Hz,1H),8.31(s,1H),8.17(d,J=5.3Hz,1H),7.68–7.60(m,1H),6.73(s,1H),5.41(d,J=1.8Hz,2H),3.86–3.73(m,1H),3.18–3.10(m,1H),2.65–2.55(m,7H),2.08(s,3H),1.97(s,3H)。
EXAMPLE 23 Synthesis of Compound 69
Figure BDA0004023656070000541
The first step: synthesis of Compound 69-2
Aqueous hydroxylamine (1 mL) was added to a solution of compound 69-1 (500 mg,3.93 mmol) in ethanol (10 mL), and the reaction mixture was stirred at 75deg.C for 16 hours. After completion of the reaction, the reaction mixture was concentrated under reduced pressure to give compound 69-2 (610 mg, yield: 82.3%). MS m/z (ESI): 161.1[ M+1 ]] +
And a second step of: synthesis of Compound 69-3
Raney nickel (640 mg,11.42 mmol) was added to a mixed solution of compound 69-2 (610 mg,3.81 mmol) in methanol and acetic acid (15 mL/1 mL) and the reaction mixture was stirred at room temperature for 16 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to give compound 69-3 (500 mg, crude product). MS M/z (ESI): 145.1.[ M+1 ] ] +
And a third step of: synthesis of Compound 69-4
Potassium carbonate (174 mg,1.26 mmol) was added to a solution of compound 69-3 (182 mg,1.26 mmol) and compound A (150 mg,0.32 mmol) in acetonitrile (5 mL), and the reaction mixture was stirred at 75℃for 12 hours. After the completion of the reaction, the reaction mixture was diluted with water (30 mL), extracted with ethyl acetate (30 ml×3), the combined organic phases were dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane/methanol=1/0 to 10/1) to give compound 69-4 (100 mg, yield: 54%). MS M/z (ESI): 555.7 [ M+1 ]] +
Fourth step: synthesis of Compound 69
Potassium hydrogen persulfate (331.6 mg,0.54 mmol) was added to a solution of compound 69-4 (100 mg,0.18 mmol) in methanol (5 mL), and the reaction mixture was stirred at room temperature for 16 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, the residue was diluted with water (20 mL), extracted with diethyl ether (20 mL. Times.3), the combined organic phases were dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/0 to 1/2) to give crude compound 69, which was purified by high performance liquid chromatography (column: gemini-C18; 150X 21.2mm,5 μm; mobile phase: acetonitrile-water (0.1% formic acid); gradient: 40-60%; column temperature: 25 ℃ C.; flow rate: 20mL/min; wavelength: 214nm; column pressure: 80 bar) to give compound 69 (40 mg; yield: 35.9%). MS m/z (ESI): 587.7[ M+1 ] ] +1 H NMR(400MHz,DMSO-d 6 )δ8.92(d,J=5.2Hz,1H),8.83(s,1H),8.57(d,J=2.4Hz,1H),8.29(s,1H),8.19(d,J=5.2Hz,1H),8.07(ddd,J=10.0,9.0,2.4Hz,1H),6.79(s,1H),5.46(d,J=1.7Hz,2H),3.35–3.32(m,1H),3.27(ddd,J=9.5,3.8,1.7Hz,2H),3.09(dt,J=6.9,6.2Hz,2H),2.45–2.34(m,2H),2.32–2.20(m,2H),2.06(s,3H),1.93(s,3H)。
EXAMPLE 24 Synthesis of Compounds 70, 70-P1, 70-P2, 70-P3 and 70-P4
Figure BDA0004023656070000551
The first step: synthesis of Compound 70-2
Sodium borohydride (1.53 g,40.4 mmol) was added to a solution of compound 70-1 (3.5 g,36.8 mmol) in methanol (150 mL) and the reaction mixture stirred at 0deg.C for 1 hr. After the completion of the reaction, the reaction mixture was quenched with water (100 mL), extracted with ethyl acetate (100 mL. Times.3), and the combined organic phases were dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure to give compound 70-2 (3.4 g, yield: 76%).
And a second step of: synthesis of Compound 70-3
Paralylsulfonyl chloride (7.34 g,38.5 mmol) was added dropwise to a solution of compound 70-2 (3.4 g,35 mmol), 4-dimethylaminopyridine (0.86 g,7 mmol) and triethylamine (4.25 g,42 mmol) in dichloromethane (50 mL) at 0deg.C and the reaction mixture was stirred at room temperature for 2 hours. After the completion of the reaction, the reaction mixture was quenched with hydrochloric acid (1M), extracted with dichloromethane (30 ml×3), the combined organic phases were dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/0 to 10/1) to give compound 70-3 (5 g, yield: 54%). 1 H NMR(400MHz,CDCl 3 )δ7.77(d,J=8.3Hz,2H),7.37(d,J=8.0Hz,2H),4.81–4.69(m,1H),2.74–2.59(m,3H),2.59–2.48(m,2H),2.46(s,3H)。
And a third step of: synthesis of Compound 70-4
Potassium thioacetate (3.64 g,31.8 mmol) was added to a solution of compound 70-3 (4 g,15.91 mmol) in N, N-dimethylformamide (40 mL), and the reaction mixture was stirred at 80℃for 2 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/0 to 3/1) to give compound 70-4 (2 g, yield: 72%). 1 H NMR(400MHz,CDCl 3 )δ4.31–4.18(m,1H),3.29–3.25(m,1H),2.93–2.86(m,2H),2.50–2.42(m,2H),2.31(s,3H)。
Fourth step: synthesis of Compound 70-5
Potassium carbonate (890 mg,6.44 mmol) was added to a solution of compound 70-4 (500 mg,3.22 mmol) in methanol (10 mL), and the reaction mixture was stirred at 50℃for 3 hours. After the completion of the reaction, the reaction mixture was diluted with water (10 mL), ph=4 was adjusted with 1N hydrochloric acid, extracted with ethyl acetate (15 ml×3), the combined organic phases were dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetateEthyl acetate=1/0 to 3/1) to give compound 70-5 (600 mg, yield: 74%). 1 H NMR(400MHz,CDCl 3 )δ3.81–3.73(m,2H),3.31–3.11(m,2H),2.83–2.62(m,4H),2.55–2.35(m,4H)。
Fifth step: synthesis of Compound 70-6
Hydrochloric acid (31.2 mL, 2M) was added to a solution of compound 70-5 (700 mg,3.12 mmol) in tetrahydrofuran (15 mL), zinc powder (2.04 g,3.12 mmol) was added with stirring, and the reaction mixture was stirred at 45℃for 1 hour. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, the residue was diluted with water (10 mL), extracted with ethyl acetate (15 mL. Times.3), and the combined organic phases were dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure to give compound 70-6 (320 mg, yield: 81%). 1 H NMR(400MHz,CDCl 3 )δ3.80–3.74(m,1H),3.22–3.30(m,1H),2.95–2.79(m,2H),2.45–2.30(m,2H),1.90(d,J=7.2Hz,1H)。
Sixth step: synthesis of Compound 70-7
Methyl iodide (1.2 g,8.5 mmol) was added to a solution of compound 70-7 (320 mg,2.83 mmol) and potassium carbonate (781 mg,5.6 mmol) in N, N-dimethylformamide (15 mL), and the reaction mixture was stirred at room temperature for 12 hours. After the completion of the reaction, the reaction mixture was diluted with water (10 mL), extracted with ethyl acetate (15 ml×3), the combined organic phases were dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/0 to 4/1) to give compound 70-7 (350 mg, yield: 87%). 1 H NMR(400MHz,CDCl 3 )δ3.67–3.56(m,1H),3.36–3.24(m,1H),2.81–2.69(m,2H),2.40–2.29(m,2H),2.07(s,3H)。
Seventh step: synthesis of Compound 70-8
Hydroxylamine (649 mg,19.65 mmol) was added to a solution of compound 70-7 (500 mg,3.93 mmol) in ethanol (10 mL) and the reaction mixture was stirred at 70℃for 16 h. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to give compound 70-8 (600 mg, yield: 66%). MS m/z (ESI): 161.2[ M+1 ]] +
Eighth step: synthesis of Compound 70-9
Aluminum nickel alloy (2.650 g,31.2 mmol) was added to compound 70-8 (500 mg,3.12 m)mol) and acetic acid (2 mL) in methanol (10 mL), the reaction mixture was stirred at room temperature under a hydrogen atmosphere for 16 hours. After the completion of the reaction, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give compound 70-9 (500 mg, yield: 77%). MS m/z (ESI) 145.2[ M+1 ]] +
Ninth step: synthesis of Compound 70-10
Compound 70-9 (45 mg,3.15 mmol) was added to a solution of compound A (500 mg,1.05 mmol) and potassium carbonate (436 mg,3.15 mmol) in N, N-dimethylformamide (15 mL), and the reaction mixture was stirred at 90℃for 12 hours. After the completion of the reaction, the reaction solution was filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/0 to 5/4) to give compound 70-10 (350 mg, yield: 50%). MS m/z (ESI): 555.8[ M+1 ]] +
Tenth step: synthesis of Compound 70
Potassium hydrogen persulfate (1127 mg,1.83 mmol) was added to a solution of compound 70-10 (340 mg,0.61 mmol) in methanol (20 mL), the reaction mixture was stirred at room temperature for 12 hours, after the completion of the reaction, the reaction solution was filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane/methanol=1/0 to 10/1) to give compound 70, and compound 70 was purified by supercritical fluid chiral chromatography (apparatus: SFC tharprep 80, column: CHIRALPAK AD-H250 mm. Times.20 mm,5 μm, mobile phase: 40% EtOH/CO) 2 (NH 4 OH 0.2%), total flow rate: 40 g/min) to give compound 70-P1 (16.5 mg, yield 4.6%), compound 70-P2 (6.3 mg, yield 1.7%), compound 70-P3 (23.0 mg, yield 6.4%) and compound 70-P4 (6.9 mg, yield 1.9%).
Compound 70-P1:
MS m/z(ESI):587.7[M+1] + . Supercritical fluid chromatography SFC: retention time=7.59 min, uv=214 nm. 1 HNMR(400MHz,CD 3 OD)δ8.87–8.79(m,2H),8.44(s,2H),8.25(d,J=5.3Hz,1H),7.73–7.67(m,1H),6.81(s,1H),5.49(s,2H),4.09–3.98(m,1H),3.90–3.79(m,1H),2.99(dt,J=12.6,9.4Hz,1H),2.92–2.81(m,4H),2.78–2.66(m,2H),2.15(s,3H),2.06(d,J=0.4Hz,3H)。
Compound 70-P2:
MS m/z(ESI):587.7[M+1] + . Supercritical fluid chromatography SFC: retention time = 7.96min, uv = 214nm. 1 HNMR(400MHz,CD 3 OD)δ8.90–8.78(m,2H),8.47–8.39(m,2H),8.27(d,J=5.3Hz,1H),7.73–7.67(m,1H),6.80(s,1H),5.49(d,J=1.9Hz,2H),4.13–3.91(m,2H),2.96–2.88(m,5H),2.88–2.78(m,2H),2.15(s,3H),2.05(s,3H)。
Compound 70-P3:
MS m/z(ESI):587.7[M+1] + . Supercritical fluid chromatography SFC: retention time=9.71 min, uv=214 nm. 1 HNMR(400MHz,CD 3 OD)δ8.87–8.77(m,2H),8.44(d,J=2.7Hz,2H),8.25(d,J=5.3Hz,1H),7.73–7.67(m,1H),6.80(d,J=0.7Hz,1H),5.49(d,J=1.9Hz,2H),4.07–3.95(m,1H),3.89–3.80(m,1H),3.03–2.96(m,1H),2.92–2.82(m,4H),2.77–2.66(m,2H),2.15(s,3H),2.05(d,J=0.5Hz,3H)。
Compound 70-P4:
MS m/z(ESI):587.7[M+1] + . Supercritical fluid chromatography SFC: retention time = 10.98min, uv = 214nm.1H NMR (400 MHz, CD) 3 OD)δ8.86(d,J=5.3Hz,1H),8.80(s,1H),8.48–8.38(m,2H),8.27(d,J=5.3Hz,1H),7.73–7.67(m,1H),6.80(s,1H),5.49(d,J=1.8Hz,2H),4.12–3.90(m,2H),2.99–2.78(m,7H),2.15(s,3H),2.04(s,3H)。
EXAMPLE 25 Synthesis of Compound 76
Figure BDA0004023656070000581
The first step: synthesis of Compound 76-2
Thionyl chloride (813 g,6.8 mmol) was added to a solution of compound 76-1 (800 mg,4.6 mmol) in ethanol (20 mL) and the reaction mixture stirred at 60℃for 3 h. After the reaction was completed, the reaction solution was cooled to room temperature, quenched by addition of saturated sodium bicarbonate solution (10 mL), concentrated under reduced pressure to remove ethanol, the residue was extracted with water (20 mL), ethyl acetate (20 ml×2), and the combined organic phases were concentrated under reduced pressure to give compound 76-2 (900 mg, crude product). MS m/z (ESI): 204.1[ M+H ]] +
And a second step of: synthesis of Compound 76-3
Sodium borohydride (334 mg,8.8 mmol) was added to a solution of compound 76-2 (900 mg,4.4 mmol) in ethanol (20 mL) and the reaction mixture stirred at 25℃for 12 h. After the completion of the reaction, the reaction mixture was quenched by addition of saturated ammonium chloride solution (5 mL), ethanol was removed by concentration under reduced pressure, the residue was diluted with (20 mL) of water, extracted with ethyl acetate (30 mL. Times.3), and the combined organic phases were concentrated under reduced pressure to give compound 76-3 (600 mg, crude product). MS m/z (ESI): 162.1[ M+H ]] +
And a third step of: synthesis of Compound 76-4
To a solution of compound 76-3 (100 mg,0.62 mmol) in dichloromethane (10 mL) was added dropwise 2 drops of N, N-dimethylformamide, and then 0.1mL of a solution of thionyl chloride was added, and the reaction mixture was stirred at room temperature for 2 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to give Compound 76-4 (100 mg, crude product).
Fourth step: synthesis of Compound 76-5
Compound A-4 (143 mg,0.55 mmol) was added to a solution of compound 76-4 (100 mg,0.55 mmol) and potassium carbonate (154 mg,1.11 mmol) in N, N-dimethylformamide (5 mL) and the reaction mixture was stirred at 65℃for 3 hours. After completion of the reaction, the reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (30 mL. Times.3). The combined organic phases were washed with saturated brine (20 ml×3), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane/methanol=1/0 to 2/1) to give compound 76-5 (100 mg, yield: 45%). MS m/z (ESI) 402.0[ M+H ]] +
Fifth step: synthesis of Compound 76-6
N-chlorosuccinimide (50 mg,0.37 mmol) was added to a solution of compound 76-5 (100 mg,0.25 mmol) in isopropanol (10 mL), and the reaction was stirred at 60℃for 16 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane/methanol=1/0 to 5/1) to give compound 76-6 (100 mg, yield: 92%). MS m/z (ESI): 435.9[ M+H ]] +
Sixth step: synthesis of Compound 76-7
N, N-dimethylformamide dimethyl acetal (55 mg,0.46 mmol) was added to a solution of compound 76-6 (100 mg,0.23 mmol) in N, N-dimethylformamide (10 mL) and the reaction mixture was stirred at 100deg.C for 16 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane/methanol=1/0 to 5/1) to give compound 76-7 (80 mg, yield: 71%). MS m/z (ESI): 491.0[ M+H ] ] +
Seventh step: synthesis of Compound 76
Tetrahydropyran-4-carboxamidine (42 mg,0.33 mmol) was added to a solution of compound 76-7 (80 mg,0.16 mmol) and potassium carbonate (68 mg,0.49 mmol) in N, N-dimethylformamide (10 mL), and the reaction mixture was stirred at 90℃for 16 hours. After completion of the reaction, the reaction mixture was diluted with water (50 mL) and extracted with methylene chloride (30 mL. Times.3). The combined organic phases were washed with saturated brine (20 mL. Times.2), dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by high pressure liquid chromatography (column: gemini-C18, 150X 21.2mm,5um; mobile: acetonitrile-water (0.1% formic acid); gradient: 40-70%, column temperature: 25 ℃ C.; flow rate: 14mL/min; wavelength: 214nm; column pressure: 80 bar) to give compound 76 (13.4 mg, yield: 14.7%). MS m/z (ESI): 556.1 558.1[ M+H ]] +1 H NMR(400MHz,CDCl 3 )δ8.82(d,J=3.9Hz,1H),8.74(s,1H),8.48(d,J=1.5Hz,1H),8.31(s,1H),8.21(d,J=3.5Hz,1H),7.59(dd,J=8.8,1.8Hz,1H),6.42(s,1H),5.43(s,2H),4.11(dd,J=12.5,6.0Hz,2H),3.58(t,J=10.8Hz,2H),3.19(t,J=10.6Hz,1H),2.20(s,3H),2.13-2.07(m,2H),2.01-1.96(m,5H)。
EXAMPLE 26 Synthesis of Compound 77
Figure BDA0004023656070000591
The first step: synthesis of Compound 77-2
Thionyl chloride (813 mg,6.83 mmol) was slowly added dropwise to a solution of compound 77-1 (800 mg,4.56 mmol) in ethanol (15 mL) and the reaction mixture stirred at 60℃for 3 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to give Compound 77-2 (910 mg, crude product). MS m/z (ESI) 203.9[ M+H ]] +
And a second step of: synthesis of Compound 77-3
Sodium borohydride (353.07 mg,9.3 mmol) was added in portions to a solution of compound 77-2 (910 mg,4.6 mmol) in ethanol (15 mL) and the reaction mixture was stirred at room temperature for 16 hours. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was diluted with water (50 mL) and extracted with ethyl acetate (80 mL. Times.3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, and filtered, and the filtrate was concentrated under reduced pressure to give compound 77-3 (700 mg, crude). MS m/z (ESI): 162.0[ M+H ]] +
And a third step of: synthesis of Compound 77-4
To a solution of compound 77-3 (120 mg,0.74 mmol) in dichloromethane (5 mL) was added dropwise 2 drops of N, N-dimethylformamide, and then 0.5mL of a solution of thionyl chloride was added, and the reaction mixture was stirred at room temperature for 2 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure to give compound 77-4 (60 mg, crude product). MS m/z (ESI) 180.7[ M+H ]] +
Fourth step: synthesis of Compound 77-5
Compound A-4 (110 mg,0.42 mmol) was added to a solution of compound 77-4 (76 mg,0.42 mmol) and potassium carbonate (117 mg,0.85 mmol) in N, N-dimethylformamide (20 mL), and the reaction mixture was stirred at 60℃for 3 hours. After completion of the reaction, the reaction mixture was diluted with water (100 mL) and extracted with ethyl acetate (80 mL. Times.3). The combined organic phases were washed with saturated brine (20 mL. Times.3), dried over anhydrous sodium sulfate, and filtered, and the filtrate was concentrated under reduced pressure to give compound 77-5 (80 mg, yield 42%). MS m/z (ESI): 401.9[ M+H ] ] +
Fifth step: synthesis of Compound 77-6
N-chlorosuccinimide (26 mg,0.19 mmol) was added to a solution of compound 77-5 (80 mg,0.91 mmol) in isopropanol (15 mL), and the reaction mixture was stirred at 60℃for 3 hours. After the completion of the reaction, the reaction mixture was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (ethyl acetate/petroleum ether=0/1 to 1/0) to give compound 77-6 (80 mg, yield: 73%). MS m/z (ESI): 437.0[ M+H ]] +
Sixth step: synthesis of Compound 77-7
N, N-dimethylDimethylformamide dimethyl acetal (87 mg,0.73 mmol) was added to a solution of compound 77-6 (80 mg,0.18 mmol) in N, N-dimethylformamide (10 mL) and the reaction mixture stirred at 100deg.C for 3 hours. After completion of the reaction, the reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (30 mL. Times.3). The combined organic phases were washed with saturated brine (30 ml×3), dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane/methanol=1/0 to 50/1) to give compound 77-7 (35 mg, yield: 34%). MS m/z (ESI): 491.0[ M+H ]] +
Seventh step: synthesis of Compound 77
Tetrahydropyran-4-carboxamidine (18 mg,0.14 mmol) was added to a solution of compound 77-7 (35 mg,0.07 mmol) and potassium carbonate (29 mg,0.21 mmol) in N, N-dimethylformamide (10 mL), and the reaction mixture was stirred at 90℃for 12 hours. After completion of the reaction, the reaction mixture was diluted with water (50 mL) and extracted with methylene chloride (30 mL. Times.3). The combined organic phases were washed with saturated brine (20 mL. Times.2), dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by high performance liquid chromatography (column: gemini-C18, 150X 21.2mm,5um; mobile: acetonitrile-water (0.1% formic acid); gradient: 40-70%, column temperature: 25 ℃ C.; flow rate: 14mL/min; wavelength: 214nm; column pressure: 80 bar) to give compound 77 (6.6 mg, yield: 14.2%). MS m/z (ESI): 556.0 558.0[ M+H ] ] +1 H NMR(400MHz,CD 3 OD)δ8.88(d,J=5.3Hz,1H),8.84(s,1H),8.55(d,J=2.5Hz,1H),8.41(s,1H),8.29(d,J=5.3Hz,1H),7.97(dd,J=8.2,2.5Hz,1H),6.81(s,1H),5.58(s,2H),4.12–4.03(m,2H),3.62(dd,J=17.8,6.6Hz,2H),3.27–3.14(m,1H),2.20(s,3H),2.09–1.96(m,7H)。
EXAMPLE 27 Synthesis of Compounds 78, 78-P1 and 78-P2
Figure BDA0004023656070000611
The first step: synthesis of Compound 78-1
Compound 69-3 (878 mg,6.09 mmol) was added to acetonitrile of compound B (500 mg,1.01 mmol) and potassium carbonate (981 mg,7.10 mmol)10 mL) solution, the reaction mixture was stirred at 75 ℃ for 12 hours. After the completion of the reaction, the reaction mixture was filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane/methanol=1/0 to 20/1) to give compound 78-1 (100 mg, yield: 16%). MS m/z (ESI): 574.0[ M+1 ]] +
And a second step of: synthesis of Compound 78
Potassium hydrogen persulfate (540 mg,0.89 mmol) was added to a solution of Compound 78-1 (170 mg,0.30 mmol) in methanol (10 mL), the reaction mixture was stirred at room temperature for 16 hours, after the completion of the reaction, the reaction solution was filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (dichloromethane/methanol=1/0 to 10/1) to give Compound 78, compound 78 was purified by supercritical fluid chiral chromatography (apparatus: SFC Thprep 80, column: CHIRALPAK AD-H250 mm. Times.20 mm,5 μm, mobile phase: 40% EtOH/CO) 2 (NH 4 OH 0.2%), total flow rate: resolution of 40g/min gave compound 78-P1 (15.6 mg, yield 8.26%) and compound 78-P2 (19.0 mg, yield 10%).
Compound 78-P1:
MS m/z(ESI):606.0[M+1] + . Supercritical fluid chromatography SFC: retention time = 4.58min, uv = 214nm. 1 HNMR(400MHz,CD 3 OD)δ8.89–8.84(m,1H),8.66–8.59(m,1H),8.41(d,J=2.4Hz,1H),7.95(d,J=5.2Hz,1H),7.54–7.52(m,1H),6.58(d,J=0.6Hz,1H),5.41(d,J=2.0Hz,2H),3.30–
3.19(m,1H),3.15–3.04(m,4H),2.41(dd,J=12.0,5.9Hz,4H),2.15(d,J=0.4Hz,3H),1.99(s,3H)。
Compound 78-P2:
MS m/z(ESI):606.0[M+1] + . Supercritical fluid chromatography SFC: retention time = 6.39min, uv = 214nm. 1 H NMR(400MHz,CD 3 OD)δ8.86(d,J=5.2Hz,1H),8.62(s,1H),8.41(d,J=2.4Hz,1H),7.95(d,J=5.2Hz,1H),7.54–7.52(m,1H),6.58(d,J=0.5Hz,1H),5.41(d,J=2.0Hz,2H),3.30–3.20(m,1H),3.16–3.04(m,4H),2.41(dd,J=12.0,5.9Hz,4H),2.15(s,3H),1.99(s,3H)。
EXAMPLE 28 Synthesis of Compounds 82, 82-P1 and 82-P2
Figure BDA0004023656070000621
The first step: synthesis of Compound 82-2
Methylamine (2.38 g,35.2 mmol) was added to a solution of compound 82-1 (3 g,17.6 mmol), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (6.75 g,35.2 mmol), 1-hydroxybenzotriazole (4.76 g,35.2 mmol) and triethylamine (5.34 g,52.8 mmol) in dichloromethane (50 mL) and the reaction mixture stirred at room temperature for 12 hours. After the completion of the reaction, the reaction mixture was diluted with water (150 mL), extracted with methylene chloride (100 ml×3), the combined organic phases were dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/0 to 5/2) to give compound 82-2 (1.1 g, yield: 30%). MS m/z (ESI): 184.1[ M+1 ]] +
And a second step of: synthesis of Compound 82-3
Compound 82-2 (1.1 g,6 mmol) was added to an ethanol solution of ammonia (8.6 mL,60 mmol) and the reaction mixture was placed in a sealed tube, heated to 90℃and stirred at that temperature for 12 hours. After completion of the reaction, the reaction mixture was concentrated under reduced pressure to give compound 82-3 (1 g, yield: 90%). MS m/z (ESI): 168.9[ M+1 ] ] +
And a third step of: synthesis of Compound 82-4
Trifluoroacetic anhydride (1.86 g,8.8 mmol) was added to a solution of compound 82-3 (1 g,5.9 mmol) and triethylamine (3 g,29.5 mmol) in dichloromethane (20 mL) and the reaction mixture was stirred at room temperature for 2 hours. After the completion of the reaction, the reaction mixture was diluted with water (50 mL), extracted with methylene chloride (50 ml×3), the combined organic phases were dried over anhydrous sodium sulfate and filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (methylene chloride/methanol=1/0 to 10/1) to give compound 82-4 (280 mg, yield: 28%). MS m/z (ESI) 150.9[ M+1 ]] +
Fourth step: synthesis of Compound 82-5
Aqueous hydroxylamine (2 mL,3.73 mmol) was added to a solution of compound 82-4 (280 mg,1.86 mmol) in ethanol (10 mL) and the reaction mixture stirred at 75deg.C for 12 h. After the completion of the reaction, the reaction mixture,the reaction solution was concentrated under reduced pressure to give compound 82-5 (300 mg, yield: 79%). MS m/z (ESI): 184.0[ M+1 ]] +
Fifth step: synthesis of Compound 82-6
Raney nickel (14 mg,0.24 mmol) was added to a solution of compound 82-5 (300 mg,1.6 mmol) and acetic acid (1 mL) in methanol (10 mL), and the reaction mixture was stirred at room temperature under a hydrogen atmosphere for 12 hours. After the completion of the reaction, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give compound 82-6 (300 mg, yield: 98%). MS m/z (ESI): 168.2[ M+1 ] ] +
Sixth step: synthesis of Compound 82
Adding 82-6 (300 mg,0.6 mmol) to a solution of A (211 mg,1.2 mmol) and potassium carbonate (175 mg,1.2 mmol) in N, N-dimethylformamide (10 mL), stirring the reaction mixture at 90deg.C for 12 hr, filtering the reaction solution after the completion of the reaction, concentrating the filtrate under reduced pressure, purifying the residue by silica gel column chromatography (dichloromethane/methanol=1/0-10/1) to obtain 82, purifying 82 by supercritical fluid chiral chromatography (equipment: SFC tharprep 80, column: CHIRALPAK AD-H250 mm. Times.20 mm,5 μm, mobile phase: 40% EtOH/CO) 2 (NH 4 OH 0.2%), total flow rate: resolution of 40g/min gave compound 82-P1 (35.2 mg, yield 9.61%) and compound 82-P2 (36.6 mg, yield 10.61%).
Compound 82-P1:
MS m/z(ESI):578.8[M+1] + . Supercritical fluid chromatography SFC: retention time = 2.37min, uv = 254nm. 1 HNMR(400MHz,CD 3 OD)δ8.89–8.83(m,2H),8.49(d,J=2.3Hz,1H),8.39–8.31(m,2H),7.79–7.71(m,1H),6.85(s,1H),5.54(d,J=1.8Hz,2H),2.77(s,3H),2.43(d,J=12.0Hz,6H),2.19(s,3H),2.08(s,3H)。
Compound 82-P2:
MS m/z(ESI):578.8[M+1] + . Supercritical fluid chromatography SFC: retention time = 7.9min, uv = 254nm. 1 H NMR(400MHz,CD 3 OD)δ8.88–8.84(m,2H),8.49(d,J=2.3Hz,1H),8.38–8.32(m,2H),7.79–7.71(m,1H),6.85(s,1H),5.53(d,J=1.8Hz,2H),2.77(s,3H),2.45(s,6H),2.19(s,3H),2.08(s,3H)。
Following the procedure of examples 1-28 above, the following compounds were prepared:
Figure BDA0004023656070000631
/>
Figure BDA0004023656070000641
/>
Figure BDA0004023656070000651
/>
Figure BDA0004023656070000661
/>
Figure BDA0004023656070000671
/>
Figure BDA0004023656070000681
/>
Figure BDA0004023656070000691
/>
Figure BDA0004023656070000701
Figure BDA0004023656070000711
/>
compound C:
Figure BDA0004023656070000712
prepared by the method of example 1 with reference to WO2021195475A 1.
Biological evaluation
Test example 1 measurement of in vitro Activity of P38 MAPK/MK2
The inhibition of p38 MAPK/MK2 by the compounds was detected using the Z-LYTE kinase assay kit (Thermo, PV 3177). The test compounds were dissolved in DMSO to 10mM stock solution and stored at-20℃until use. The initial concentration of the compound is 10 mu M,1% DMSO, 5-fold dilution, 8 concentrations, double-well; 50mM HEPES pH 7.5,10mM MgCl 2 0.01% Brij-35,1mM EGTA as reaction buffer was used to prepare a 2x active p38a/inactive MK2/Ser/Thr 4 mixture, and the final 10. Mu.L reaction was performed in 384 well plates (Corning, 4514) containing 500ng/mL inactive MK2 (abcam, 79910), 8ng/mL active p38a (Cana, 04-152), 2. Mu.M Ser/Thr 4; after 1 hour of reaction at 20℃ Development Reagent A diluted 2048 times was added to each well, and after 1 hour of incubation at room temperature, 5. Mu.L of stop buffer solution was added to terminate the reaction, and the reaction was detected by a microplate reader (Ex.400 nm, em.445nm; ex.400nm, em.520 nm). Concentration-effect curves were fitted with GraphPad Prism 8 software and compound concentrations with 50% inhibition, i.e. IC, were calculated 50 . The results are shown in Table 1.
TABLE 1
Figure BDA0004023656070000713
/>
Figure BDA0004023656070000721
From Table 1, it can be seen that the compounds of the present disclosure have good inhibitory activity against p38 MAPK/MK 2.
Test example 2. In vitro Activity assay of p38 MAPK/MK5
The inhibition of p38 MAPK/MK5 by the compounds was detected using the Z-LYTE kinase assay kit (Thermo, PV 3177). The test compounds were dissolved in DMSO to 10mM stock solution and stored at-20℃until use. The initial concentration of the compound is 10 mu M,1% DMSO, 5-fold dilution, 8 concentrations, double-well; 50mM HEPES pH 7.5,10mM MgCl 2 0.01% Brij-35,1mM EGTA as reaction buffer for preparation The mixture of 2x active p38a/inactive MK5/Ser/Thr 4 was placed and the final 10. Mu.L reaction was performed in 384 well plates (Corning, 4514) containing 10. Mu.g/mL of inactive MK5 (abcam, 217826), 1ng/mL of active p38a (Carna, 04-152), 2. Mu.M Ser/Thr 4; after 4 hours of reaction at 20℃ Development Reagent A diluted 2048 times was added to each well, and after 1 hour of incubation at room temperature, 5. Mu.L of stop buffer solution was added to terminate the reaction, and the reaction was detected by a microplate reader (Ex. 400nm, em.445nm; ex.400nm, em.520 nm). Concentration-effect curves were fitted with GraphPad Prism 8 software and compound concentrations with 50% inhibition, i.e. IC, were calculated 50 . The results are shown in Table 2.
TABLE 2
Compounds of formula (I) p38 MAPK/MK5 IC 50 (nM)
Compound 3 9353
Compound 3-P1 3155
Compound 4 5729
Compound 6 3933
Compound 7 9321
Compound 8 4621
Compound 9 864
Compound 10 2894
Compound 10-P1 1980
Compound 14-G1 773.2
Compound 14-G2 2674
Compound 16-P1 8940
Compound 33-P1 679.5
Compound 42 2091
Compound 58 873.7
Compound 63-P1 5449
As can be seen from Table 2, the compounds of the present disclosure all have greater than 0.6. Mu.M inhibitory activity against p38 MAPK/MK 5. This further demonstrates that the compounds of the present disclosure have good selectivity for p38 MAPK/MK 2.
Test example 3 measurement of in vitro Activity of P38 MAPK/ATF2
Compound p38a catalyzed ATF2Inhibition was detected using HTRF method. The test compounds were dissolved in DMSO to 10mM stock solution and stored at-20℃until use. Compound initial concentration 10 μm,0.25% DMSO,5 fold dilution, 8 concentrations, double-multiplexed wells; 40mM Tris pH 7.5,20mM MgCl2,0.1mg/mL BSA, 50. Mu.M DTT as reaction buffer was used to prepare 3.5Xp38 a (MAPK 14, carna Biosciences, 04-152) Protein working solution, 3.5x Human ATF2 Protein (Sino Biological, 11599-H20B) working solution and 3.5 XATP working solution, 10mM EDTA was used to terminate the reaction, and finally 14. Mu.L of the reaction was performed in 96-well plates (cisbio, 66PL 96025) containing 0.29 ng/. Mu. L p38a, 0.29. Mu.M Human ATF2 Protein; 25. Mu.M ATP. After 35min of reaction at 20℃a pre-formulated antibody solution (cibio, 63ADK015PEG, phospho-ATF2 Eu Cryptate antibody, phospho-ATF2 d2 anti-body) was added to each well, incubated overnight at room temperature, diluted 40-fold with Detection buffer, and detected by an ELISA reader (HTRF compatible reader). Concentration-effect curves were fitted with GraphPad Prism 8 software and compound concentrations with 50% inhibition, i.e. IC, were calculated 50 . The results are shown in Table 3.
TABLE 3 Table 3
Compounds of formula (I) p38 MAPK/ATF2 IC 50 (nM)
Compound 2-P1 902.7
Compound 3-P1 4715
Compound 4 28021
Compound 6 854.7
Compound 7 5389
Compound 8 1450
Compound 9 6976
Compound 10 4657
Compound 10-P1 1173
Compound 14-G1 15915
Compound 14-G2 8262
Compound 16-P1 8634
Compound 33-P1 >20000
Compound 41 >20000
Compound 63-P1 1250
As can be seen from Table 3, the compounds of the present disclosure all have greater than 0.8. Mu.M inhibitory activity against p38 MAPK/ATF 2. This further demonstrates that the compounds of the present disclosure have good selectivity for p38 MAPK/MK 2.
Test example 4 in vitro Activity assay of human PBMC cell supernatant TNF-alpha
Inhibition of human PBMC cell supernatant TNF- α assay protocol was tested using the Elisa assay kit (Biyun, PI 518). The test compounds were dissolved in DMSO to 10mM stock solution and stored at-20℃until use. The initial concentration of the compound is 2 mu M, the dilution is 5 times, the concentration is 6, the cells are plated into double wells, the Elisa is detected as a single well, the final concentration of the DMSO is 0.4%, and the initial concentration, the dilution times, the gradient concentration number and the double well number of the compound can be changed according to the actual condition of compound screening.
Fresh human Peripheral Blood Mononuclear Cells (PBMC) (Eriosema chinense) were plated in 96-well plates (Corning, 3599) at a rate of 2X 10A 5, each well containing 100. Mu.L of RPMI-1640 (Gibco #A 1049101) +10% FBS (Gibco, 10099141C), 37℃C, 5% CO 2 Culturing overnight; the compound to be tested is added into 96-well culture plate with the volume of 25 mu L/well, after 1h, 5 mu L of LPS is added to make the final concentration of the compound to be 100ng/mL, no LPS and compound are added into the negative control well, no compound is added into the positive control well, and the temperature is 37 ℃ and the concentration is 5% of CO 2 After further incubation for 24h, the cell culture supernatants were collected by centrifugation at 500rcf for 8min, and assayed for TNF- α concentration according to the protocol in the Elisaa kit. Concentration-effect curves were fitted with GraphPad Prism 8 software and compound concentrations with 50% inhibition, i.e. IC, were calculated 50 . The results are shown in Table 4.
TABLE 4 Table 4
Compounds of formula (I) TNF-αIC50(nM)
Compound 2 45.38
Compound 2-P1 10.39
Compound 3 41.40
Compound 3-P1 6.39
Compound 6 14.96
Compound 7 41.48
Compound 9 37.91
Compound 10 17.68
Compound 10-P1 7.58
Compound 14-G1 7.96
Compound 14-G2 4.84
Compound 16 29.15
Compound 16-P1 9.27
Compound 33-P1 22.79
Compound 37 0.61
Compound 38 0.21
Compound 41 98.84
Compound 63-P1 1.73
Compound 66-P2 24.62
Compound 70-P2 9.92
Compound 82-P2 17.27
From table 4, it can be seen that the compounds of the present disclosure have good inhibitory effect on TNF- α of human PBMC cells.
Test example 5 evaluation of in vitro CYP enzyme inhibition
The experiment adopts a cocktail method to conduct inhibition research on the 1A2,2B6,2C8,2C19,2C9,2D6 and 3A4 subtype enzyme activities of the CYP enzyme by the compound to be tested, and IC50 values of the compound to be tested on the activities of several CYP enzyme subtypes are measured. The control compounds were: fluvoxamine (1 A2), ketoconazole (2B 6), montelukast sodium Montelukast (2C 8), tranylcypromine (2C 19), sulfafenpyrazol Sulfaphenazole (2C 9), quinidine (2D 6) and Ketoconazole (3 A4/5); the CYP enzyme probe substrates used are: phenacetin (1A 2), bupropion buprofion (2B 6), amodiaquine (2C 8), mefenone Mephenytoin (2C 19), diclofenac sodium Diclofenac (2C 9), dextromethorphan dexteromethorphan (2D 6) and Testosterone tester (3A 4/5). PBS Buffer was 50mM K2HPO4 Buffer. The concentrations of the test compounds were 50. Mu.M, 12.5. Mu.M, 3.125. Mu.M, 0.781. Mu.M, 0.195. Mu.M, 0.0488. Mu.M, respectively. Adding the corresponding probe substrate and microsomes into PBS, uniformly mixing, then adding a control compound/a compound to be tested/DMSO solution into a corresponding reaction system, pre-incubating for 5min in a 37 ℃ water bath, adding a 10mM NADPH solution, placing in the 37 ℃ water bath for reaction for 10min, and adding an internal standard acetonitrile solution to terminate the reaction. Centrifuging at 4000rpm, adding equal volume of pure water into the supernatant solution, mixing, analyzing the amount of each probe substrate reaction product by LC-MS/MS (AB Triple Quard 5500), and calculating IC50 by using the measured product yield by GraphPad Prism 5. The results are shown in Table 5:
TABLE 5
Figure BDA0004023656070000761
As can be seen from Table 5, compounds 10-P1 and 63-P1 of the present disclosure have no inhibitory effect on CYP enzymes in vitro.
Test example 6 evaluation of in vitro time dependent CYP3A4 enzyme inhibition
The test compares the IC50 value change condition of the test compound for inhibiting CYP3A4/5 subtype enzyme activity under the two conditions under the test conditions that the test compound is pre-reacted with a microsome reaction system and no pre-reaction is carried out, and calculates the corresponding IC50shift value. The control compounds were: verapamil (CYP 3 A4/5), probe substrates were: testosterone (CYP 3A 4/5). PBS Buffer was 50mM K2HPO4 Buffer. The concentrations of the test compounds were 50. Mu.M, 10. Mu.M, 2. Mu.M, 0.4. Mu.M, 0.08. Mu.M, and 0.016. Mu.M, respectively. The probe substrate was prepared in advance with PBS solution to a substrate solution (1990. Mu.L of PBS+10. Mu.L of substrate). Adding microsomes into PBS, pre-incubating a sample without pre-reaction group in a water bath at 37 ℃ for 30min, adding a control compound/test compound/DMSO solution, a 10mM NADPH solution and a probe substrate solution into a corresponding reaction system, placing the mixture into the water bath at 37 ℃ for reaction for 10min, and adding an internal standard acetonitrile solution to terminate the reaction; adding a control compound/compound to be tested/DMSO solution and a 10mM NADPH solution into a corresponding system of the pre-reaction group sample, uniformly mixing, placing in a 37 ℃ water bath for reaction for 30min, then adding a probe substrate solution, placing in a 37 ℃ water bath for reaction for 10min, and adding an internal standard acetonitrile solution to terminate the reaction. After centrifugation at 4000rpm, the supernatant solution was mixed with an equal volume of pure water, the amounts of the reaction products of the respective probe substrates were analyzed by LC-MS/MS (AB Triple Quard 5500) respectively, and IC50 calculation was performed using the measured amounts of the products by using GraphPad Prism 5. The results are shown in Table 6:
TABLE 6
Figure BDA0004023656070000762
Figure BDA0004023656070000771
As can be seen from Table 6, compounds 10-P1 and 63-P1 of the present disclosure have no time-dependent CYP3A4 enzyme inhibition.
Test example 7 evaluation of in vitro liver microsome stability
The control compounds used in this experiment were: ketansertin, the final concentration of microsomes in the experimental system was 0.5mg/mL. PBS Buffer was 50mM K2HPO4 Buffer. The concentration of the test compound was 1. Mu.M. Adding microsomes into PBS, respectively adding a control compound/a compound to be detected into a corresponding reaction system, uniformly mixing, placing in a 37 ℃ water bath for pre-incubation for 5min, adding 20mM of NADPH solution, and starting the reaction under the 37 ℃ water bath condition (for a No NADPH sample, replacing 20mM of NADPH solution with the same volume of PBS solution). At the reaction time points of 0min,10min,30min,60min and 90min, respectively, 30. Mu.L of the reaction sample (for the No NADPH sample, 0min,90min were taken out respectively) was taken out of each reaction system, and the reaction was terminated by immediately adding 300. Mu.L of the internal standard acetonitrile solution. Centrifuging at 4000rpm, collecting supernatant, adding equal volume of pure water, mixing, and LC-MS/MS (AB Triple Quard 5500) separately measured the amount of compound in each time point sample and calculated T 1/2 . The results are shown in Table 7:
TABLE 7
Figure BDA0004023656070000772
As can be seen from table 7, the compound 10-P1 of the present disclosure has good in vitro liver microsomal stability (human/mouse/rat).
Test example 8 in vivo pharmacokinetic evaluation in mice
The compound was weighed and dissolved in DMAC: solutol: in pbs=1:1:8 mixed solvent, 30 μl of whole blood was collected at 0.083, 0.25, 0.5, 1, 2, 4, 7 and 24 hours after venous/gastric lavage of mice, and immediately after anticoagulation with EDTA-K2, centrifugation was performed at 4000rpm for 5min at 4 ℃, and the supernatant was frozen in a-80 ℃ refrigerator. Treatment of plasma samples: after precipitation with CH3CN precipitant containing internal standard, 12700rpm is used for centrifugation for 10min, supernatant is taken for analysis by LC-MS/MS (AB Triple Quard 5500), blood concentration is obtained, and parameter calculation is carried out through non-atrioventricular model version 8.1 of Winnolin. The results are shown in Table 8:
TABLE 8
Figure BDA0004023656070000773
Figure BDA0004023656070000781
As can be seen from table 8, both the compound 10-P1 and the compound 63-P1 of the present disclosure have good exposure, in vivo clearance and bioavailability over the concentration dose and detection time range of administration. In intravenous administration, compound 10-P1 had an in vivo clearance CL of 1241.95mL/hr/kg, which was significantly better than compound C (2313.22 mL/hr/kg); in the gastric lavage administration, the blood drug exposure of the compound 10-P1 is 2120.68hr ng/mL, which is doubled compared with the blood drug exposure of the compound C; compound 63-P1 had an in vivo clearance CL of 1492.93mL/hr/kg, which was significantly better than that of compound C (2313.22 mL/hr/kg); in the administration by gastric lavage, the blood drug exposure of compound 63-P1 was 2596.40hr ng/mL, which is doubled over the blood drug exposure of compound C. Thus, this suggests that the disclosed compounds have good in vivo pharmacokinetic properties and are significantly superior to control compound C.
The embodiments of the present disclosure have been described above. However, the present disclosure is not limited to the above embodiments. Any modification, equivalent replacement, improvement, etc. made within the spirit and principle of the present disclosure should be included in the protection scope of the present disclosure.

Claims (11)

1. A compound of formula I, racemates, stereoisomers, tautomers, isotopic labels, solvates, pharmaceutically acceptable salts or prodrugs thereof:
Figure FDA0004023656060000011
wherein W is CH or N;
m is an integer of 0 to 5;
n is an integer of 0 to 3;
ring A is C 3-20 Cycloalkyl, 3-20 membered heterocyclyl, the carbon atom in ring a being attached to the parent nucleus, said 3-20 membered heterocyclyl containing 1, 2 or more O, N or S atoms;
R 1 selected from H, halogen, CN and C 1-6 An alkyl group;
R 2 selected from-OR 81 、-NH-C(O)R 82 、-NHR 83 and-C (O) NHR 84
R 3 Selected from H, C 1-10 Alkyl and C 3-20 Cycloalkyl;
R 4 selected from H, halogen and C 1-10 An alkyl group;
R 5 are each independently selected from H, halogen, -OH, -C 1-6 Alkyl, -C 1-6 Alkoxy, oxo (=o), -C (O) C 1-6 Alkyl, -C (O) OH, -C (O) NR 91a R 91b 、-S(O) 2 R 92 and-S (O) 2 NR 93a R 93b
R 6 Selected from H, halogen and methyl;
R 7 independently selected from H, halogen, C unsubstituted or substituted by Ra 1-10 Alkyl and C 3-20 Cycloalkyl; ra is selected from halogen, C 3-20 Cycloalkyl;
R 81 、R 82 、R 83 、R 84 identical or different, independently of one another, are selected from C which is unsubstituted or optionally substituted by 1, 2, 3, 4 or 5 Rb 6-14 aryl-C 1-10 Alkyl, 5-14 membered heteroaryl-C 1-10 Alkyl, C 6-14 Aryl and 5-14 membered heteroaryl; each Rb is the same or different and is independently selected from halogen, halogenated C 1-10 Alkyl, C 1-10 Alkyl and C 1-10 An alkoxy group;
R 91a 、R 91b 、R 92 、R 93a 、R 93b identical or different, independently of one another, from H, C 1-6 Alkyl and C 3-20 Cycloalkyl groups.
2. The compound of claim 1, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt or prodrug thereof,
w is CH or N;
m is an integer of 0 to 5;
n is an integer of 0 to 3;
ring A is C 3-20 Cycloalkyl, 3-20 membered heterocyclyl, the carbon atom in ring a being attached to the parent nucleus, said 3-20 membered heterocyclyl containing 1, 2 or more O, N or S atoms;
R 1 selected from H, halogen, CN and C 1-6 An alkyl group;
R 2 selected from-OR 81 、-NH-C(O)R 82 、-NHR 83 and-C (O) NHR 84
R 3 Selected from H, C 1-10 Alkyl groupC 3-20 Cycloalkyl;
R 4 selected from H, halogen and C 1-10 An alkyl group;
R 5 are each independently selected from H, halogen, OH, C 1-6 Alkyl, C 1-6 Alkoxy, oxo (=o), -C (O) C 1-6 Alkyl, -C (O) OH, -C (O) NR 91a R 91b 、-S(O) 2 R 92 and-S (O) 2 NR 93a R 93b
R 6 Selected from H, halogen and methyl;
R 7 are independently selected from H, halogen, C 1-10 Alkyl and C 3-20 Cycloalkyl;
R 81 、R 82 、R 83 、R 84 identical or different, independently of one another, from C 6-14 aryl-C 1-10 Alkyl, 5-14 membered heteroaryl-C 1-10 Alkyl, C 6-14 Aryl and 5-14 membered heteroaryl; wherein C is 6-14 Aryl, 5-14 membered heteroaryl is unsubstituted or optionally substituted with 1, 2, 3, 4 or 5 substituents independently of one another selected from halogen, halo C 1-10 Alkyl, C 1-10 Alkyl and C 1-6 Alkoxy substitution;
R 91a 、R 91b 、R 92 、R 93a 、R 93b identical or different, independently of one another, from H, C 1-6 Alkyl and C 3-20 Cycloalkyl groups.
3. The compound, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt or prodrug thereof according to claim 1 or 2,
w is CH or N;
m is 0, 1, 2, 3, 4 or 5;
n is 0, 1, 2, 3;
ring A is C 3-9 Cycloalkyl, 3-9 membered heterocyclyl, the carbon atom in ring a being attached to the parent, said 3-9 membered heterocyclyl containing 1, 2 or more O, N or S atoms;
R 1 is halogen;
R 2 selected from-OR 81 、-NH-C(O)R 82 、-NHR 83 and-C (O) NHR 84
R 3 Is C 1-3 Alkyl or C 3-6 Cycloalkyl;
R 4 is C 1-3 An alkyl group;
R 5 are each independently selected from halogen, -OH, -C 1-3 Alkyl, -C 1-3 Alkoxy, oxo (=o), -C (O) C 1-3 Alkyl, -C (O) OH, -C (O) NR 91a R 91b 、-S(O) 2 R 92 and-S (O) 2 NR 93a R 93b
R 6 Selected from H, halogen and methyl;
R 7 are each independently selected from H, halogen and C 1-3 An alkyl group;
R 81 、R 82 、R 83 、R 84 identical or different, independently of one another, from C 6-8 aryl-C 1-3 Alkyl, 5-6 membered heteroaryl-C 1-3 Alkyl, C 6-14 Aryl and 5-14 membered heteroaryl; wherein C is 6-14 Aryl, 5-14 membered heteroaryl is unsubstituted or optionally substituted with 1, 2, 3, 4 or 5 substituents independently of one another selected from halogen, halo C 1-3 Alkyl, C 1-3 Alkyl and C 1-3 Alkoxy substitution;
R 91a 、R 91b 、R 92 、R 93a 、R 93b identical or different, independently of one another, from H, C 1-3 Alkyl and C 3-6 Cycloalkyl groups.
4. A compound, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt or prodrug thereof according to claim 1 to 3,
w is CH or N;
m is 0, 1, 2 or 3;
n is 0 or 1;
ring A is selected from the group consisting of piperidinyl, tetrahydro-2H-pyranyl, tetrahydrofuranyl, oxetanyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 2-oxaspiro [3.3] heptyl, 2-oxaspiro [3.5] nonyl, 2-azaspiro [3.3] heptyl, 2-azaspiro [3.5] nonyl, azetidinyl, tetrahydropyrrolyl, thietanyl, tetrahydro-2H-thiopyranyl;
R 1 selected from Cl, br;
R 2 selected from-OR 81 、-NH-C(O)R 82 、-NHR 83 、-C(O)NHR 84
R 3 Selected from methyl, cyclopropyl;
R 4 selected from methyl;
R 5 are each independently selected from F, -OH, methyl, methoxy, oxo (= O), -C (O) C 1-3 Alkyl, -C (O) OH, -C (O) NH 2 、-C(O)NHCH 3 、-S(O) 2 CH 3 、-S(O) 2 CH 2 CH 3 、-S(O) 2 -cyclopropane;
R 6 selected from H, F, cl;
R 7 Selected from H;
R 81 、R 82 、R 83 、R 84 the same or different, independently of one another, are selected from phenylmethyl, pyridylmethyl, pyridylethyl, phenyl, pyridinyl, unsubstituted or optionally substituted by 1, 2 or 3 Rb; each Rb is the same or different and is selected from F, cl, CF independently of the others 3
5. The compound, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt or prodrug thereof of any one of claims 1 to 4, wherein ring a is selected from the group consisting of the structures:
Figure FDA0004023656060000031
preferably, R 5 And ring a is selected from the group consisting of:
Figure FDA0004023656060000041
preferably, the compound of formula I has the structure of formula Ia or Ib:
Figure FDA0004023656060000051
wherein R is 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 A, W, m, n have the definition of any one of claims 1 to 4.
6. The compound, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt or prodrug thereof of any one of claims 1 to 5, wherein formula I has the structure shown in formula ii:
Figure FDA0004023656060000052
wherein R is 1 、R 3 、R 4 、R 5 、R 6 、R 7 W, m, n and ring a independently of each other have the definition as defined in any one of claims 1 to 5;
R 10 selected from H, halogen, unsubstituted or optionally substituted with 1, 2 or more halogen, OH, NH 2 Substituted with the following groups: c (C) 1-10 Alkyl, C 1-10 Alkoxy, halo C 1-10 Alkyl, halogenated C 1-10 Alkoxy, C 2-10 Alkenyl, C 2-10 Alkenyloxy, C 2-10 Alkynyl, C 2-10 Alkynyl oxy;
each R 11 The same or different, independently of one another, from H, halogen, C 1-6 Alkyl, halogenated C 1-10 An alkyl group;
p is an integer of 0 to 4;
preferably, R 10 Selected from H, armorA base; p is 0, 1 or 2;
each R 11 The same or different, are independently selected from F, cl, CF 3
Preferably, the compound of formula III has the structure of formula IIa or IIb:
Figure FDA0004023656060000061
wherein R is 1 、R 3 、R 4 、R 5 、R 6 、R 7 、R 10 、R 11 A, W, m, n, p have the definition as set forth above or in any of claims 1 to 5.
7. The compound according to claim 1 or 6, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt or prodrug thereof, wherein
Figure FDA0004023656060000062
Is->
Figure FDA0004023656060000063
R 5 Selected from halogen, -OH, -C 1-3 Alkyl, -C 1-3 Alkoxy, oxo (=o), -C (O) C 1-3 Alkyl, -C (O) OH, -C (O) NR 91a R 91b 、-S(O) 2 R 92 and-S (O) 2 NR 93a R 93b ,R 91a 、R 91b 、R 92 、R 93a 、R 93b Identical or different, independently of one another, from H, C 1-3 Alkyl and C 3-6 Cycloalkyl; preferably, R 5 Selected from F, -OH, methyl, methoxy, -C (O) C 1-3 Alkyl, -C (O) OH, -C (O) NH 2 、-C(O)NHCH 3 、-S(O) 2 CH 3 、-S(O) 2 CH 2 CH 3 and-S (O) 2 -cyclopropane.
8. The compound, racemate, stereoisomer, tautomer, isotopic label, solvate, pharmaceutically acceptable salt or prodrug thereof according to any one of claims 1 to 7, wherein the compound has the following structure:
Figure FDA0004023656060000064
Figure FDA0004023656060000071
Figure FDA0004023656060000081
Figure FDA0004023656060000091
Figure FDA0004023656060000101
/>
Figure FDA0004023656060000111
/>
Figure FDA0004023656060000121
/>
Figure FDA0004023656060000131
/>
Figure FDA0004023656060000141
/>
Figure FDA0004023656060000151
/>
Figure FDA0004023656060000161
Wherein the x in the formulae of compounds 10 and 63 represent the presence of a cis-trans structure therein, and the x is one of cis or trans; preferably, the compound of formula I has the following structure:
Figure FDA0004023656060000171
/>
Figure FDA0004023656060000181
wherein the compounds 10-P1 or 10-P2 and 63-P1 or 63-P2 are represented by the formula wherein a cis-trans structure is present and wherein cis or trans is present.
9. A process for the preparation of a compound according to any one of claims 1 to 8, comprising the steps of:
scheme one: the compound a1 and the compound a2 undergo a coupling reaction to obtain a compound of the formula I
The reaction formula is as follows:
Figure FDA0004023656060000191
wherein Y is Cl or Br; w, R 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 M, n and ring A have the above-described definitions independently of one another.
Scheme II: when W is N, R 7 In case of H, the compound b1 is reacted with the compound b2 to obtain the compound of formula I
The reaction formula is as follows:
Figure FDA0004023656060000192
wherein R is 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 M, n, p and ring a independently of each other have the definition as defined in any one of claims 1 to 7;
preferably, the reaction is carried out in the presence of an inorganic base; the inorganic base is selected from one of sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide and potassium hydroxide;
preferably, when R 5 In the case of OH, OH in the compound b2 may be protected by a silicon protecting group, which may be tert-butyldiphenylsilyl; the silicon protecting group is removed during the reaction to give deprotected OH.
10. A pharmaceutical composition comprising a therapeutically effective amount of at least one of the compounds of any one of claims 1-7, racemates, stereoisomers, tautomers, isotopic labels, solvates, pharmaceutically acceptable salts, or prodrug compounds thereof, and one or more pharmaceutically acceptable carriers.
11. Use of a compound according to any one of claims 1-8, at least one of its racemates, stereoisomers, tautomers, isotopic labels, solvates, pharmaceutically acceptable salts or prodrug compounds thereof, or a pharmaceutical composition according to claim 10, for the preparation of a medicament;
preferably, the medicament is a medicament for the treatment and/or prophylaxis of a disease associated with a p38 kinase inhibitor, for example may be an MK2 inhibitor or a p38 MAPK/MK2 pathway modulator;
preferably, the disease is a disease associated with the p38 MAPK/MK2 pathway, such as autoimmune and inflammatory diseases (e.g., rheumatoid arthritis, felicitous sweat gland, psoriasis, inflammatory bowel disease, atopic dermatitis, systemic lupus erythematosus, etc.), bone diseases, metabolic diseases, neurological and neurodegenerative diseases, cancer, cardiovascular diseases, allergies and asthma, alzheimer's disease, and hormone-related diseases.
CN202211699892.XA 2021-12-29 2022-12-28 P38MAPK/MK2 pathway regulator, and composition, preparation method and application thereof Pending CN116354935A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111640112X 2021-12-29
CN202111640112 2021-12-29

Publications (1)

Publication Number Publication Date
CN116354935A true CN116354935A (en) 2023-06-30

Family

ID=86930069

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211699892.XA Pending CN116354935A (en) 2021-12-29 2022-12-28 P38MAPK/MK2 pathway regulator, and composition, preparation method and application thereof

Country Status (3)

Country Link
CN (1) CN116354935A (en)
TW (1) TW202332439A (en)
WO (1) WO2023125708A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024046327A1 (en) * 2022-08-30 2024-03-07 长春金赛药业有限责任公司 P38α-MK2 INHIBITOR COMPOUNDS, PHARMACEUTICAL COMPOSITIONS, AND USE THEREOF

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI2648516T1 (en) * 2010-12-06 2019-01-31 Aclaris Therapeutics, Inc. Substituted pyridinone-pyridinyl compounds
SI3003039T1 (en) * 2013-06-07 2021-08-31 Aclaris Therapeutics, Inc. Methyl/fluoro-pyridinyl-methoxy substituted pyridinone-pyridinyl compounds and fluoro-pyrimidinyl-methoxy substituted pyridinone-pyridinyl compounds
WO2021195475A1 (en) * 2020-03-27 2021-09-30 Aclaris Therapeutics, Inc. Process, compositions, and crystalline forms of substituted pyridinone-pyridinyl compounds
CR20230468A (en) * 2021-03-31 2023-12-07 Xinthera Inc Mk2 inhibitors and uses thereof
CN115636814A (en) * 2022-02-28 2023-01-24 瑞石生物医药有限公司 Pyrimidine derivative and application thereof

Also Published As

Publication number Publication date
TW202332439A (en) 2023-08-16
WO2023125708A1 (en) 2023-07-06

Similar Documents

Publication Publication Date Title
TWI713952B (en) Compounds and compositions as inhibitors of bromodomain proteins
RU2677699C1 (en) Imidazotriazine derivatives as modulators of tnf activity
EP3144308B1 (en) Nitrogen-containing heterocyclic compound
EP3728207B1 (en) Quinazolinones as parp14 inhibitors
EP3317289A1 (en) Novel substituted aminothiazolopyrimidinedione for the treatment and prophylaxis of virus infection
BR112016011851B1 (en) Fused Imidazole and Pyrazole Derivatives as Modulators of TNF Activity
RU2677697C1 (en) Triazolopyridine derivatives as modulators of tnf activity
US20120309739A1 (en) Akt / pkb inhibitors
EA025881B1 (en) Macrocyclic flt3 kinase inhibitors
EP3426243B1 (en) 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
MX2008001020A (en) A pyridin quinolin substituted pyrrolo [1,2-b] pyrazole monohydrate as tgf-beta inhibitor.
EP3426244B1 (en) 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
TW200936140A (en) Substituted arylamide oxazepinopyrimidone derivatives
WO2023003002A1 (en) Annulated pyridazine compound
CN113518779B (en) Thieno heterocyclic derivative, preparation method and medical application thereof
CN116354935A (en) P38MAPK/MK2 pathway regulator, and composition, preparation method and application thereof
TW202328130A (en) Sik inhibitor and its composition, preparation method and application
CN116444492A (en) P38MAPK/MK2 pathway regulator, and composition, preparation method and application thereof
WO2017156177A1 (en) 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
JP2023542969A (en) Compounds as Akt Kinase Inhibitors
KR20140069204A (en) Compounds useful as inhibitors of choline kinase
CN116655638B (en) Deuterated PRMT5 inhibitors
CN117800948A (en) Substituted pyridine or pyrimidine derivative, pharmaceutical composition, preparation method and application thereof
WO2022171126A1 (en) Fused ring compound used as wee-1 inhibitor
WO2022135390A1 (en) Ketohexokinase inhibitor and use 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
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40087937

Country of ref document: HK