CN114075212A - Fused tricyclic derivative, preparation method and application thereof in medicine - Google Patents

Fused tricyclic derivative, preparation method and application thereof in medicine Download PDF

Info

Publication number
CN114075212A
CN114075212A CN202110928898.9A CN202110928898A CN114075212A CN 114075212 A CN114075212 A CN 114075212A CN 202110928898 A CN202110928898 A CN 202110928898A CN 114075212 A CN114075212 A CN 114075212A
Authority
CN
China
Prior art keywords
group
compound
groups
pharmaceutically acceptable
enantiomer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202110928898.9A
Other languages
Chinese (zh)
Other versions
CN114075212B (en
Inventor
张晓敏
胡伟民
贺峰
陶维康
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Jiangsu Hengrui Medicine Co Ltd
Shanghai Hengrui Pharmaceutical Co Ltd
Original Assignee
Jiangsu Hengrui Medicine Co Ltd
Shanghai Hengrui Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Jiangsu Hengrui Medicine Co Ltd, Shanghai Hengrui Pharmaceutical Co Ltd filed Critical Jiangsu Hengrui Medicine Co Ltd
Publication of CN114075212A publication Critical patent/CN114075212A/en
Application granted granted Critical
Publication of CN114075212B publication Critical patent/CN114075212B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/06Peri-condensed systems
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • 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
    • 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
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • 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
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Diabetes (AREA)
  • Virology (AREA)
  • Pulmonology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Molecular Biology (AREA)
  • Cardiology (AREA)
  • Vascular Medicine (AREA)
  • Dermatology (AREA)
  • AIDS & HIV (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Endocrinology (AREA)
  • Transplantation (AREA)
  • Emergency Medicine (AREA)
  • Hospice & Palliative Care (AREA)

Abstract

The disclosure relates to fused tricyclic derivatives, processes for their preparation and their use in medicine. In particular to a fused tricyclic derivative shown in a general formula (I), a preparation method thereof, a pharmaceutical composition containing the derivative and application of the derivative as a therapeutic agent, in particular application of the derivative as a TLR7/8/9 inhibitor and application of the derivative in preparing medicines for treating and/or preventing inflammatory and autoimmune diseases.

Description

Fused tricyclic derivative, preparation method and application thereof in medicine
Technical Field
The disclosure belongs to the field of medicines, and relates to a fused tricyclic derivative, a preparation method and an application thereof in medicines. In particular, the disclosure relates to fused tricyclic derivatives represented by general formula (I), a preparation method thereof, a pharmaceutical composition containing the derivatives, and use of the fused tricyclic derivatives as a TLR7/8/9 inhibitor in treating inflammatory and autoimmune diseases.
Background
Toll-like receptors (TLRs) are a class of evolutionarily conserved transmembrane innate immune receptors that are involved in the first-line defense for protection of human health and play an important role in the recognition of pathogen-associated molecular patterns (PAMPs) (Kawai, t. et al, Nature immunol.,11,2010, 373-. TLRs are expressed in various immune cells and can be classified into two types according to their expression sites: TLR expressed in cell membranes (TLR1/2/4/5/6) and endosomal membranes (TLR3/7/8/9) recognize different components and molecules in PAMPs, respectively. Among them, TLR7/8/9 is mainly highly expressed in DC cells and B cells, TLR7/8 mainly recognizes ssRNA, and TLR9 mainly recognizes CpG-DNA. TLR7/8/9 is activated after binding its ligand, binds to adaptor protein MyD88 in cytoplasm, initiates NF-kappa B and IRF pathways, activates DC cells, and produces type I interferon and other various inflammatory cytokines. In B cells, TLR7/8/9 and nucleic acid substances are combined to play an important role in the process of producing antinuclear antibodies by the B cells, and the type I interferon secreted by DC cells can promote the further proliferation and activation of the autoimmune B cells so as to cause a series of inflammatory reactions.
Systemic Lupus Erythematosus (SLE) belongs to an autoimmune connective tissue disease, and there are three major classes of first-line clinical drugs for SLE: hormones, immunosuppressants and antimalarial drugs. Only one new drug, belimumab, was approved by the FDA in this century, butIt has only modest and delayed efficacy in a small fraction of SLE patients (Navarra, s.v. et al, Lancet 2011,377,721), with very limited treatment options. Therefore, there is an urgent need for new therapies that improve a larger proportion of the patient population and that can be used safely for long periods of time. A phenomenon in which TLR7/9 and type I interferon expression were significantly upregulated was found in PBMCs of Systemic Lupus Erythematosus (SLE) patients (Beverly D.LC et al, Mol Immunol.,2014,61: 38-43). Mice overexpressing TLR7 have been reported to exacerbate autoimmune disease and autoinflammation (Santiago-Raber ML et al, J Immunol.,2008,181: 1556-Oak 1562), while functional inhibition of TLR7/9 may alleviate B6-FaslprAnd pathological manifestations in lupus mice such as BXSB (Dlight H.Kono et al, PNAS,2009,106(29): 12061-12066). Given the close relationship of TLR7/8/9 to antinuclear antibodies and type I interferons, small molecule inhibitors targeting TLR7/8/9 are likely to have potential for the treatment of SLE.
Patent applications for published TLR7/8/9 inhibitors include WO2019233941a1, WO2020020800a1, WO2018049089a1, WO2019238616a1, WO2017106607a1, CN109923108A, and WO2020048605a1, among others.
Disclosure of Invention
The purpose of the present disclosure is to provide a compound represented by the general formula (I), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure BDA0003210363510000021
wherein:
x is selected from O atom, S atom and NH;
L1is selected from CH2、CR4aR4bAnd C (O);
L2selected from the group consisting of CR4cR4dC (O), O atom, S atom and NH;
g is CR2aOr an N atom;
ring a is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
each R is1Is the same as orDifferent and each independently selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, - (CH)2)rNR6aR6bNitro, hydroxy, hydroxyalkyl, -C (O) NR6aR6b、-C(O)R7、-NR6cC(O)R7、-C(O)OR8Cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R2aand R2The same or different, and each is independently selected from the group consisting of hydrogen atom, halogen, alkyl group, alkoxy group, haloalkyl group, haloalkoxy group, cyano group, amino group, nitro group, hydroxyl group and hydroxyalkyl group;
ring B is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
each R is3The same or different, and each is independently selected from the group consisting of hydrogen atom, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, oxo, cyano, amino, nitro, hydroxy, and hydroxyalkyl;
R0selected from the group consisting of hydrogen atom, deuterium atom, halogen, alkyl, alkenyl, alkynyl, heteroalkyl, alkoxy, haloalkyl, deuterated alkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, - (CH)2)rC(O)NR6aR6bAnd
Figure BDA0003210363510000031
L3selected from the group consisting of a bond, alkylene, and heteroalkylene, wherein said alkylene and heteroalkylene are each independently optionally selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl, and heteroalkyleneAryl is substituted by one or more substituents in the aryl;
ring C is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
each R is5Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, alkyl, heteroalkyl, alkenyl, alkynyl, alkoxy, oxo, haloalkyl, haloalkoxy, cyano, amino, - (CH)2)rNR6aR6bNitro, hydroxy, hydroxyalkyl, -C (O) NR6aR6b、-C(O)OR8、-S(O)mR9、-S(O)mNR6aR6bCycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, heteroalkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R4aand R4bThe same or different and each is independently selected from the group consisting of hydrogen atom, alkyl group, halogen, alkoxy group, haloalkyl group, haloalkoxy group, cyano group, hydroxyl group and hydroxyalkyl group;
R4cand R4dThe same or different and each is independently selected from the group consisting of hydrogen atom, alkyl group, halogen, alkoxy group, haloalkyl group, haloalkoxy group, cyano group, hydroxyl group and hydroxyalkyl group; or R4cAnd R4dTogether with the carbon atom to which they are attached form a cycloalkyl or heterocyclyl group;
R6aand R6bAre the same or different and are each independently selected from the group consisting of a hydrogen atom, an alkyl group, an alkenyl group, an alkynyl group, a haloalkyl group, a hydroxyalkyl group, a cycloalkyl group, a cycloalkylalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group; or R6aAnd R6bTogether with the nitrogen atom to which they are attached form a heterocyclic group optionally substituted with one member selected from the group consisting of halogen, alkyl, oxo, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclic, aryl and heteroarylSubstituted by one or more substituents;
R6cselected from the group consisting of hydrogen atoms, alkyl groups, alkenyl groups, alkynyl groups, haloalkyl groups, hydroxyalkyl groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups;
R7selected from the group consisting of hydrogen atoms, alkyl groups, haloalkyl groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups; wherein said alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy and hydroxyalkyl;
R8selected from the group consisting of hydrogen atoms, alkyl groups, alkenyl groups, alkynyl groups, haloalkyl groups, hydroxyalkyl groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups;
R9selected from the group consisting of hydrogen atoms, alkyl groups, alkenyl groups, alkynyl groups, haloalkyl groups, hydroxyalkyl groups, amino groups, hydroxyl groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups;
u is 0, 1,2 or 3;
v is 0, 1,2 or 3;
n is 0 or 1;
p is 0, 1,2, 3,4,5 or 6;
r is 0, 1,2, 3,4,5 or 6;
m is 0, 1 or 2;
s is 0, 1,2, 3,4,5 or 6; and is
t is 0, 1,2, 3,4,5 or 6.
In some preferred embodiments of the present disclosure, the compound of formula (I), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein
X is selected from O atom, S atom and NH;
L1is selected from CH2、CR4aR4bAnd C (O);
L2selected from the group consisting of C (O), O atoms, S atoms, and NH;
g is CR2aOr an N atom;
ring a is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
each R is1Are the same or different and are each independently selected from the group consisting of a hydrogen atom, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, - (CH)2)rNR6aR6bNitro, hydroxy, hydroxyalkyl, -C (O) NR6aR6b、-C(O)R7、-NR6cC(O)R7、-C(O)OR8Cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R2aand R2The same or different, and each is independently selected from the group consisting of hydrogen atom, halogen, alkyl group, alkoxy group, haloalkyl group, haloalkoxy group, cyano group, amino group, nitro group, hydroxyl group and hydroxyalkyl group;
ring B is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
each R is3The same or different, and each is independently selected from the group consisting of hydrogen atom, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, oxo, cyano, amino, nitro, hydroxy, and hydroxyalkyl;
R0selected from the group consisting of hydrogen atom, deuterium atom, halogen, alkyl, alkenyl, alkynyl, heteroalkyl, alkoxy, haloalkyl, deuterated alkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl and
Figure BDA0003210363510000041
L3selected from the group consisting of a bond, alkylene and heteroalkylene, wherein said alkylene and heteroalkylene are each independently optionally selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, ammoniaSubstituted with one or more substituents selected from the group consisting of nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
ring C is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
each R is5Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, alkyl, heteroalkyl, alkenyl, alkynyl, alkoxy, oxo, haloalkyl, haloalkoxy, cyano, amino, - (CH)2)rNR6aR6bNitro, hydroxy, hydroxyalkyl, -C (O) NR6aR6b、-C(O)OR8、-S(O)mR9、-S(O)mNR6aR6bCycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, heteroalkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R4aand R4bThe same or different and each is independently selected from the group consisting of hydrogen atom, alkyl group, halogen, alkoxy group, haloalkyl group, haloalkoxy group, cyano group, hydroxyl group and hydroxyalkyl group;
R6aand R6bAre the same or different and are each independently selected from the group consisting of a hydrogen atom, an alkyl group, an alkenyl group, an alkynyl group, a haloalkyl group, a hydroxyalkyl group, a cycloalkyl group, a cycloalkylalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group; or R6aAnd R6bTogether with the nitrogen atom to which they are attached form a heterocyclyl group, which heterocyclyl group is optionally substituted by one or more substituents selected from the group consisting of halogen, alkyl, oxo, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R6cselected from the group consisting of hydrogen atoms, alkyl groups, alkenyl groups, alkynyl groups, haloalkyl groups, hydroxyalkyl groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups;
R7selected from the group consisting of hydrogen atoms, alkyl groups, haloalkyl groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups; wherein said alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy and hydroxyalkyl;
R8selected from the group consisting of hydrogen atoms, alkyl groups, alkenyl groups, alkynyl groups, haloalkyl groups, hydroxyalkyl groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups;
R9selected from the group consisting of hydrogen atoms, alkyl groups, alkenyl groups, alkynyl groups, haloalkyl groups, hydroxyalkyl groups, amino groups, hydroxyl groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups;
u is 0, 1,2 or 3;
v is 0, 1,2 or 3;
n is 0 or 1;
p is 0, 1,2, 3,4,5 or 6;
r is 0, 1,2, 3,4,5 or 6;
m is 0, 1 or 2;
s is 0, 1,2, 3,4,5 or 6; and is
t is 0, 1,2, 3,4,5 or 6.
In some preferred embodiments of the present disclosure, the compound of formula (I), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein X is an O atom.
In some preferred embodiments of the present disclosure, the compound of formula (I), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein L is1Is CH2. In some preferred embodiments of the present disclosure, the compound of formula (I), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is0Selected from hydrogen atoms, C1-6Alkyl, - (CH)2)rC(O)NR6aR6bAnd
Figure BDA0003210363510000061
ring C, L3、R5、R6a、R6bR and t are as defined in formula (I); preferably, R0Is composed of
Figure BDA0003210363510000062
Ring C, L3、R5And t is as defined in formula (I).
In some preferred embodiments of the present disclosure, the compound of formula (I), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein L is2Is CR4cR4d,R4cAnd R4dAs defined in formula (I).
In some preferred embodiments of the present disclosure, the compound of formula (I), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is4cAnd R4dAre the same or different and are each independently selected from the group consisting of a hydrogen atom, C1-6Alkyl and halogen; or R4cAnd R4dTogether with the carbon atom to which they are attached form a 3-to 6-membered cycloalkyl group or a 3-to 6-membered heterocyclyl group; preferably, R4cAnd R4dTogether with the carbon atom to which they are attached form a 3 to 6 membered cycloalkyl group; more preferably, R4cAnd R4dTogether with the attached carbon atom form a cyclopropyl group.
In some preferred embodiments of the present disclosure, the compound represented by the general formula (I), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, is a compound represented by the general formula (II), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure BDA0003210363510000063
wherein:
ring A, ring B, ring C, G, L2、L3、R1To R3、R5P, s, t, n, u and v are as defined in formula (I).
In some preferred embodiments of the present disclosure, the compound of formula (I) or formula (II), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein G is CR2a(ii) a And R is2aIs a hydrogen atom.
In some preferred embodiments of the present disclosure, the compound represented by formula (I) or formula (II), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein n is 0.
In some preferred embodiments of the present disclosure, the compound of formula (I) or formula (II), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring a is selected from the group consisting of 3-to 8-membered cycloalkyl, 3-to 12-membered heterocyclyl, 6-to 10-membered aryl, and 5-to 10-membered heteroaryl; preferably, ring a is 5 to 10 membered heteroaryl; more preferably, ring A is pyridyl or
Figure BDA0003210363510000071
Most preferably, ring a is pyridyl.
In some preferred embodiments of the present disclosure, the compound of formula (I) or formula (II), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring B is selected from the group consisting of 3-to 8-membered cycloalkyl, 3-to 12-membered heterocyclyl, 6-to 10-membered aryl, and 5-to 10-membered heteroaryl; preferably, ring B is a 3 to 12 membered heterocyclic group containing at least one nitrogen atom; more preferably, ring B is piperidinyl.
In some preferred embodiments of the present disclosure, the compound represented by formula (I) or formula (II), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, is a compound represented by formula (III), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure BDA0003210363510000072
wherein:
ring B is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
ring C, R1To R3、R5、R2a、L3S, t and v are as defined in formula (I).
In some preferred embodiments of the present disclosure, the compound of formula (III), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring B is piperidinyl.
In some preferred embodiments of the present disclosure, the compound of formula (I), formula (II) or formula (III), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring C is selected from the group consisting of 3-to 8-membered cycloalkyl, 3-to 12-membered heterocyclyl, 6-to 10-membered aryl, and 5-to 10-membered heteroaryl; preferably, ring C is a 3 to 12 membered heterocyclic group containing at least one nitrogen atom or a 5 to 10 membered heteroaryl group containing at least one nitrogen atom; more preferably, ring C is piperidinyl or 5,6,7, 8-tetrahydro-2, 6-naphthyridin-3-yl.
In some preferred embodiments of the present disclosure, the compound represented by formula (I), formula (II) or formula (III), or a tautomer, racemate, enantiomer or mixture thereofIn the form of a isomer, diastereoisomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R2Selected from hydrogen atoms, halogens, C1-6Alkyl and C1-6An alkoxy group; preferably, R2Is a hydrogen atom.
In some preferred embodiments of the present disclosure, the compound represented by formula (I), formula (II) or formula (III), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, is a compound represented by formula (IV), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure BDA0003210363510000081
wherein:
ring C is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom or
Figure BDA0003210363510000082
Ring D is a 4-to 6-membered heterocyclic group containing at least one nitrogen atom;
Z1、Z2、Z3and Z4One is a carbon atom and the remaining three are the same or different and are each independently CR5Or an N atom;
R10selected from the group consisting of hydrogen atoms, alkyl groups, heteroalkyl groups, haloalkyl groups, and hydroxyalkyl groups;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
t is 1,2, 3,4,5 or 6;
R1、R3、R5、L3s and v are as defined in formula (I).
In some preferred embodiments of the present disclosure, the compound of formula (IV), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring C is piperidinyl or 5,6,7, 8-tetrahydro-2, 6-naphthyridin-3-yl.
In some preferred embodiments of the present disclosure, the compound of formula (IV), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is10Is a hydrogen atom or C1-6An alkyl group.
In some preferred embodiments of the present disclosure, the compound of formula (I), (II), (III) or (IV), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein each R is1Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Haloalkyl, C1-6Haloalkoxy, cyano, amino, nitro, hydroxy and C1-6A hydroxyalkyl group; preferably, each R1Are the same or different and are each independently a hydrogen atom or C1-6An alkyl group.
In some preferred embodiments of the present disclosure, the compound of formula (I), (II), (III) or (IV), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein each R is3Are the same or different and are each independently selected from the group consisting of hydrogen, halogen and C1-6An alkyl group; preferably, R3Is a hydrogen atom.
In some preferred embodiments of the present disclosure, the compound of formula (I), formula (II), formula (III) or formula (IV), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein L is3Is a bond.
In some preferred embodiments of the present disclosure, the compound of formula (I), formula (II), formula (III) or formula (IV), or a tautomer thereofIn the form of a solid, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein each R is5Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C1-6Alkyl radical, C1-6Heteroalkyl group, C1-6Alkoxy, oxo, C1-6Haloalkyl, C1-6Haloalkoxy, cyano, amino, nitro, hydroxy and C1-6A hydroxyalkyl group; preferably, each R5Are the same or different and are each independently a hydrogen atom or C1-6An alkyl group.
In some preferred embodiments of the present disclosure, the compound of formula (I), (II), (III) or (IV), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein v is 1,2, or 3.
In some preferred embodiments of the present disclosure, the compound of formula (I) or formula (II), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein u is 1.
In some preferred embodiments of the present disclosure, the compound of formula (I) or formula (II), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein p is 0, 1,2, 3, or 4; preferably, p is 2.
In some preferred embodiments of the present disclosure, the compound of formula (I) or formula (II), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein s is 0 or 1; preferably, s is 0.
In some preferred embodiments of the present disclosure, the compound of formula (I), (II) or (III), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein t is 0, 1 or 2; preferably, t is 0 or 1.
In some preferred embodiments of the present disclosure, the compound of formula (IV), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein t is 1 or 2.
In some preferred embodiments of the present disclosure, the compound of formula (I), (II), (III) or (IV), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein r is 0, 1 or 2; preferably, r is 1.
In some preferred embodiments of the present disclosure, the compound of formula (I), (II), (III) or (IV), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is6aAnd R6bAre the same or different and are each independently selected from the group consisting of a hydrogen atom, C1-6Alkyl radical, C1-6Haloalkyl and C1-6A hydroxyalkyl group; or R6aAnd R6bTogether with the nitrogen atom to which they are attached form a 3-to 12-membered heterocyclic group, said 3-to 12-membered heterocyclic group being optionally selected from halogen, C1-6Alkyl, oxo, C1-6Alkoxy radical, C1-6Haloalkyl, C1-6Haloalkoxy, cyano, amino, hydroxy and C1-6Substituted with one or more substituents in hydroxyalkyl; preferably, R6aAnd R6bAre the same or different and are each independently a hydrogen atom or C1-6An alkyl group.
In some preferred embodiments of the present disclosure, the compound represented by formula (I), formula (II), formula (III) or formula (IV), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein the pharmaceutically acceptable salt is hydrochloride or trifluoroacetate; preferably a trifluoroacetate salt.
In some preferred embodiments of the present disclosure, the compound of formula (I), or a tautomer, racemate, enantiomer, or mixture thereof,Diastereoisomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein R1Is C1-6Alkyl, and p is 0, 1,2, 3 or 4.
In some preferred embodiments of the present disclosure, the compound of formula (II), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is1Is C1-6Alkyl, and p is 2.
In some preferred embodiments of the present disclosure, the compound of formula (I), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is5Is C1-6Alkyl, and t is 0, 1 or 2.
In some preferred embodiments of the present disclosure, the compound of formula (II) or formula (III), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is5Is C1-6Alkyl, and t is 0 or 1.
In some preferred embodiments of the present disclosure, the compound of formula (IV), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is5Is C1-6Alkyl, and t is 1 or 2.
In some preferred embodiments of the present disclosure, the compound of formula (I), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring a is selected from the group consisting of a 3-to 8-membered cycloalkyl group, a 3-to 12-membered heterocyclyl group, a 6-to 10-membered aryl group, and a 5-to 10-membered heteroaryl group; ring B is selected from 3-to 8-membered cycloalkyl, 3-to 12-membered heterocyclyl, 6-to 10-membered aryl, and 5-to 10-membered heteroaryl; x is an O atom; l is1Is CH2;L2Is CR4cR4d;R4cAnd R4dTogether with the carbon atom to which they are attached form a 3 to 6 membered cycloalkyl group; g is CR2a;R2aIs a hydrogen atom;R0selected from hydrogen atoms, C1-6Alkyl, - (CH)2)rC(O)NR6aR6bAnd
Figure BDA0003210363510000111
ring C is selected from 3-to 8-membered cycloalkyl, 3-to 12-membered heterocyclyl, 6-to 10-membered aryl, and 5-to 10-membered heteroaryl; l is3Is a bond; r1Is C1-6An alkyl group; r2Is a hydrogen atom; r5Is C1-6An alkyl group; p is 0, 1,2, 3 or 4; t is 0, 1 or 2; n is 0 or 1; u is 1; v is 1,2 or 3; r6aAnd R6bAre the same or different and are each independently a hydrogen atom or C1-6An alkyl group; r is 0, 1 or 2; s is 0.
In some preferred embodiments of the present disclosure, the compound of formula (II), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring a is a 5-to 10-membered heteroaryl; ring B is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom; ring C is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom or a 5-to 10-membered heteroaryl group containing at least one nitrogen atom; g is CR2a;R2aIs a hydrogen atom; l is3Is a bond; r1Is C1-6An alkyl group; r2Is a hydrogen atom; r5Is C1-6An alkyl group; p is 2; t is 0 or 1; n is 0; u is 1; v is 1,2 or 3; s is 0.
In some preferred embodiments of the present disclosure, the compound of formula (III), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring B is piperidinyl; w1、W2、W3、W4And W5Are the same or different and are each independently CR1Or an N atom; ring C is piperidinyl or 5,6,7, 8-tetrahydro-2, 6-naphthyridin-3-yl; r1Is a hydrogen atom or C1-6An alkyl group; r2Is a hydrogen atom; r2aIs a hydrogen atom; r5Is C1-6An alkyl group; l is3Is a bond; t is 0 or 1; v is 1,2 or 3; s is 0.
In some preferred embodiments of the present disclosure, the compound of formula (IV), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring C is piperidinyl or 5,6,7, 8-tetrahydro-2, 6-naphthyridin-3-yl; r10Is a hydrogen atom or C1-6An alkyl group; w1、W2、W3、W4And W5Are the same or different and are each independently CR1Or an N atom; r1Is a hydrogen atom or C1-6An alkyl group; r5Is C1-6An alkyl group; l is3Is a bond; t is 1 or 2; v is 1,2 or 3; s is 0.
Table a typical compounds of the present disclosure include, but are not limited to:
Figure BDA0003210363510000112
Figure BDA0003210363510000121
Figure BDA0003210363510000131
Figure BDA0003210363510000141
another aspect of the present disclosure relates to a compound of formula (IIIa), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure BDA0003210363510000142
wherein:
ring B is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
R1to R3、R2aS and v are as defined in formula (III).
Another aspect of the present disclosure relates to a compound of formula (IVa), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure BDA0003210363510000143
wherein:
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
R1、R3s and v are as defined in formula (IV).
Table B typical compounds of the present disclosure include, but are not limited to:
Figure BDA0003210363510000151
Figure BDA0003210363510000161
another aspect of the present disclosure relates to a compound represented by general formula (IIIaa), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure BDA0003210363510000162
wherein:
Rwfor protection of amino groupsA group; preferably, RwIs tert-butyloxycarbonyl;
ring B is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
R1、R2、R2a、R3s and v are as defined in formula (IIIa).
Another aspect of the present disclosure relates to a compound of formula (iva), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure BDA0003210363510000171
wherein:
Rwis an amino protecting group; preferably, RwIs tert-butyloxycarbonyl;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom; r1、R3S and v are as defined in formula (IVa).
Table C typical compounds of the present disclosure include, but are not limited to:
Figure BDA0003210363510000172
Figure BDA0003210363510000181
another aspect of the present disclosure relates to a method of preparing a compound of formula (III), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, comprising:
Figure BDA0003210363510000182
method 1
Subjecting a compound of formula (IIIa) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, to reductive amination reaction with a compound of formula (V) to obtain a compound of formula (III) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof,
or
Method 2
Carrying out coupling reaction on the compound of the general formula (IIIa) or a tautomer, a racemate, an enantiomer, a diastereomer or a mixture form thereof or a pharmaceutically acceptable salt thereof and the compound of the general formula (V') to obtain the compound of the general formula (III) or the tautomer, the racemate, the enantiomer, the diastereomer or a mixture form thereof or a pharmaceutically acceptable salt thereof;
wherein:
L3is a bond;
ring B is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom;
y is halogen; preferably, Y is chloro;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
ring C, R1To R3、R5、R2aS, t and v are as defined in formula (III).
Another aspect of the present disclosure relates to a method of preparing a compound of formula (IV), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, comprising:
Figure BDA0003210363510000191
method 1
The compound of the general formula (IVa) or the tautomer, racemate, enantiomer, diastereomer or mixture form thereof, or the pharmaceutically acceptable salt thereof, and the compound of the general formula (VI) are subjected to reductive amination reaction to obtain R10Selected from the group consisting of hydrogen atoms, alkyl groups, heteroalkyl groups, haloalkyl groups, hydroxyalkyl groups and RwA compound of general formula (IV) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
or
Method 2
The compound of the general formula (IVa) or the tautomer, racemate, enantiomer, diastereomer or mixture form thereof, or the pharmaceutically acceptable salt thereof and the compound of the general formula (VI') are subjected to coupling reaction to obtain R10Selected from the group consisting of hydrogen atoms, alkyl groups, heteroalkyl groups, haloalkyl groups, hydroxyalkyl groups and RwA compound of general formula (IV) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
and
optionally, when R10Is RwWhen it further comprises the step of reacting R10Is RwIn the form of a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or removing the protecting group RwTo obtain R10A compound of formula (IV) being a hydrogen atom or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
wherein:
L3is a bond;
ring C is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom or
Figure BDA0003210363510000201
Ring D is a 4-to 6-membered heterocyclic group containing at least one nitrogen atom;
Z1、Z2、Z3and Z4One is a carbon atom and the remaining three are the same or different and are each independently CR5Or an N atom;
y is halogen; preferably, Y is chloro;
RWis an amino protecting group; preferably, RWIs tert-butyloxycarbonyl;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
R1、R3、R5s, t and v are as defined in formula (IV).
Another aspect of the present disclosure relates to a method of preparing a compound of formula (IIIa), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, comprising:
Figure BDA0003210363510000211
removing the protecting group R from the compound of formula (IIIaa) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereofwTo obtain a compound of formula (IIIa) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
wherein:
RWis an amino protecting group; preferably, RWIs tert-butyloxycarbonyl;
ring B is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
R1、R2、R2a、R3s and v are as defined in formula (IIIa).
Another aspect of the present disclosure relates to a method of preparing a compound of formula (IVa), or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, comprising:
Figure BDA0003210363510000212
removing the protecting group R from the compound of formula (IVaa) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereofwTo obtain a compound of general formula (IVa) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
wherein:
RWis an amino protecting group; preferably, RWIs tert-butyloxycarbonyl;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
R1、R3s and v are as defined in formula (IVa).
Another aspect of the present disclosure relates to a pharmaceutical composition comprising a compound of the present disclosure represented by formula (I), formula (II), formula (III), formula (IV), and table a, or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, or excipients.
The disclosure further relates to the use of compounds of formula (I), formula (II), formula (III), formula (IV) and table a or tautomers, racemates, enantiomers, diastereomers or mixtures thereof, or pharmaceutically acceptable salts thereof, or pharmaceutical compositions comprising the same, in the manufacture of a medicament for inhibiting TLR7, TLR8 and TLR 9.
The disclosure further relates to the use of a compound of formula (I), formula (II), formula (III), formula (IV) and table a, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, in the manufacture of a medicament for inhibiting TLR7, TLR8, or TLR 9; preferably for the manufacture of a medicament for inhibiting TLR7 and TLR8, or for the manufacture of a medicament for inhibiting TLR7 and TLR 9.
The present disclosure further relates to the use of a compound of formula (I), formula (II), formula (III), formula (IV) and table a, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for the preparation of a medicament for the treatment and/or prevention of inflammatory or autoimmune diseases. Wherein said inflammatory or autoimmune disease is preferably selected from the group consisting of Systemic Lupus Erythematosus (SLE), rheumatoid arthritis, Multiple Sclerosis (MS) and Sjogren's syndrome.
The present disclosure further relates to a method of inhibiting TLR7, TLR8, and TLR9 comprising administering to a patient in need thereof an effective inhibiting amount of a compound of formula (I), formula (II), formula (III), formula (IV), and table a, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
The present disclosure further relates to a method of inhibiting TLR7, TLR8 or TLR9, preferably TLR7 and TLR8, or TLR7 and TLR9, comprising administering to a patient in need thereof an effective inhibiting amount of a compound of formula (I), formula (II), formula (III), formula (IV) and table a or a tautomer, racemate, enantiomer, diastereomer or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
The present disclosure further relates to a method of treating and/or preventing inflammatory or autoimmune diseases comprising administering to a patient in need thereof a therapeutically effective amount of a compound of formula (I), formula (II), formula (III), formula (IV) and table a or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same. Wherein said inflammatory or autoimmune disease is preferably selected from the group consisting of Systemic Lupus Erythematosus (SLE), rheumatoid arthritis, Multiple Sclerosis (MS) and Sjogren's syndrome.
The present disclosure further relates to compounds of formula (I), formula (II), formula (III), formula (IV) and table a or tautomers, racemates, enantiomers, diastereomers or mixtures thereof, or pharmaceutically acceptable salts thereof, or pharmaceutical compositions comprising the same, for use as a medicament.
The disclosure further relates to compounds of formula (I), formula (II), formula (III), formula (IV) and table a or tautomers, racemates, enantiomers, diastereomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, or pharmaceutical compositions comprising the same, for use in inhibiting TLR7, TLR8 and TLR 9.
The present disclosure further relates to compounds of formula (I), formula (II), formula (III), formula (IV) and table a or tautomers, racemates, enantiomers, diastereomers or mixtures thereof, or pharmaceutically acceptable salts thereof, or pharmaceutical compositions comprising the same, for use in inhibiting TLR7, TLR8 or TLR 9; preferably for inhibiting TLR7 and TLR8, or TLR7 and TLR 9.
The present disclosure further relates to compounds of formula (I), formula (II), formula (III), formula (IV) and table a or tautomers, racemates, enantiomers, diastereomers or mixtures thereof, or pharmaceutically acceptable salts thereof, or pharmaceutical compositions comprising the same, for use in the treatment and/or prevention of inflammatory or autoimmune diseases. Wherein said inflammatory or autoimmune disease is preferably selected from the group consisting of Systemic Lupus Erythematosus (SLE), rheumatoid arthritis, Multiple Sclerosis (MS) and Sjogren's syndrome.
In view of their activity as selective inhibitors of TLR7, TLR8, or TLR9, compounds of formula (I), formula (II), formula (III), formula (IV), and table a are useful for treating TLR7, TLR8, or TLR9 family receptor-related diseases, respectively, including but not limited to: inflammatory diseases (such as crohn's disease, ulcerative colitis, asthma, graft versus host disease, allograft rejection, chronic obstructive pulmonary disease); autoimmune diseases (such as graves' disease, rheumatoid arthritis, systemic lupus erythematosus, lupus nephritis, cutaneous lupus, psoriasis); autoinflammatory diseases (including Cryopyrin-associated periodic syndrome (CAPS), TNF receptor-associated periodic syndrome (TRAPS), Familial Mediterranean Fever (FMF), adult still's disease, systemic onset juvenile idiopathic arthritis, gout, gouty arthritis); metabolic disorders (including type 2 diabetes, atherosclerosis, myocardial infarction); destructive bone disorders (such as bone resorption diseases, osteoarthritis, osteoporosis, multiple myeloma-related bone disorders); proliferative disorders (such as acute myeloid leukemia, chronic myeloid leukemia); angiogenic disorders (such as angiogenic disorders including solid tumors, ocular neovascularisation and infantile hemangiomas); infectious diseases (such as sepsis, septic shock, and shigellosis); neurodegenerative diseases (such as alzheimer's disease, parkinson's disease, cerebral ischemia caused by traumatic injury or neurodegenerative diseases), neoplastic diseases (such as metastatic melanoma, kaposi's sarcoma, multiple myeloma) and viral diseases (such as HIV infection, CMV retinitis and AIDS).
More specifically, specific conditions or diseases that may be treated with the compounds of the disclosure include, but are not limited to, pancreatitis (acute or chronic), asthma, allergy, adult respiratory distress syndrome, chronic obstructive pulmonary disease, glomerulonephritis, rheumatoid arthritis, systemic lupus erythematosus, scleroderma, chronic thyroiditis, graves ' disease, autoimmune gastritis, diabetes, autoimmune hemolytic anemia, autoimmune neutropenia, thrombocytopenia, atopic dermatitis, chronic active hepatitis, myasthenia gravis, multiple sclerosis, inflammatory bowel disease, ulcerative colitis, crohn's disease, psoriasis, graft-versus-host disease, endotoxin-induced inflammatory responses, tuberculosis, atherosclerosis, muscle degeneration, cachexia, psoriatic arthritis, Reiter's syndrome, gout, Traumatic arthritis, rubella arthritis, acute synovitis, pancreatic beta cell disease; diseases characterized by massive neutrophil infiltration; rheumatoid spondylitis, gouty arthritis and other arthritic conditions, cerebral malaria, chronic pulmonary inflammatory disease, silicosis, pulmonary sarcoidosis, bone resorption disease, allograft rejection, fever and myalgia from infection, cachexia secondary to infection, keloid formation, scar tissue formation, ulcerative colitis, pyretic disease (pyresis), influenza, osteoporosis, osteoarthritis, acute myelogenous leukemia, chronic myelogenous leukemia, metastatic melanoma, kaposi's sarcoma, multiple myeloma, sepsis, septic shock and shigellasis; alzheimer's disease, Parkinson's disease, cerebral ischemia or neurodegenerative diseases caused by traumatic injury; angiogenic disorders including solid tumors, ocular neovascularization, and infantile hemangiomas; viral diseases including acute hepatitis infections (including hepatitis a, hepatitis b and hepatitis c), HIV infection and CMV retinitis, AIDS, ARC or malignancy and herpes; stroke, myocardial ischemia, ischemia in stroke heart attack, organ hypoxia, vascular proliferation, cardiac and renal reperfusion injury, thrombosis, cardiac hypertrophy, thrombin-induced platelet aggregation, endotoxemia and/or toxic shock syndrome, prostaglandin endoperoxidase synthase-2 related conditions, and pemphigus vulgaris. In a preferred method of treatment, the condition is selected from those of crohn's disease, ulcerative colitis, allograft rejection, rheumatoid arthritis, psoriasis, ankylosing spondylitis, psoriatic arthritis and pemphigus vulgaris. Alternatively preferred methods of treatment, the disorder is ischemia reperfusion injury, including cerebral ischemia reperfusion injury caused by stroke and myocardial ischemia reperfusion injury caused by myocardial infarction. In another preferred method of treatment, the disorder is multiple myeloma.
The active compounds may be formulated in a form suitable for administration by any suitable route, using one or more pharmaceutically acceptable carriers to formulate compositions of the disclosure by conventional methods. Thus, the active compounds of the present disclosure may be formulated in a variety of dosage forms for oral administration, injection (e.g., intravenous, intramuscular, or subcutaneous), inhalation, or insufflation. The compounds of the present disclosure may also be formulated in sustained release dosage forms, such as tablets, hard or soft capsules, aqueous or oily suspensions, emulsions, injections, dispersible powders or granules, suppositories, lozenges, or syrups.
As a general guide, the active compound is preferably administered in a unit dose or in a manner such that the patient can self-administer it in a single dose. The unit dose of a compound or composition of the present disclosure may be expressed in the form of a tablet, capsule, cachet, bottled liquid, powder, granule, lozenge, suppository, reconstituted powder, or liquid. A suitable unit dose may be 0.1 to 1000 mg.
The pharmaceutical compositions of the present disclosure may contain, in addition to the active compound, one or more excipients selected from the following: fillers (diluents), binders, wetting agents, disintegrants, excipients, and the like. Depending on the method of administration, the compositions may contain from 0.1 to 99% by weight of active compound.
Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be inert excipients, granulating agents, disintegrating agents, binding agents and lubricating agents. These tablets may be uncoated or they may be coated by known techniques which mask the taste of the drug or delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
Oral formulations may also be provided in soft gelatin capsules wherein the active ingredient is mixed with an inert solid diluent or wherein the active ingredient is mixed with a water soluble carrier or an oil vehicle.
Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending, dispersing or wetting agents. Aqueous suspensions may also contain one or more preservatives, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents.
Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, or in a mineral oil. The oil suspension may contain a thickener. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable preparation. These compositions can be preserved by the addition of antioxidants.
The pharmaceutical compositions of the present disclosure may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, or a mineral oil or a mixture thereof. Suitable emulsifiers may be naturally occurring phospholipids, and the emulsions may also contain sweetening, flavoring, preservative and antioxidant agents. Such formulations may also contain a demulcent, a preservative, a colorant and an antioxidant.
The pharmaceutical compositions of the present disclosure may be in the form of a sterile injectable aqueous solution. Among the acceptable vehicles or solvents that may be employed are water, ringer's solution and isotonic sodium chloride solution. The sterile injectable preparation may be a sterile injectable oil-in-water microemulsion in which the active ingredient is dissolved in an oil phase, and the injection or microemulsion may be injected into the bloodstream of a patient by local mass injection. Alternatively, it may be desirable to administer the solution and microemulsion in a manner that maintains a constant circulating concentration of the disclosed compounds. To maintain such a constant concentration, a continuous intravenous delivery device may be used. An example of such a device is an intravenous pump model Deltec CADD-PLUS. TM.5400.
The pharmaceutical compositions of the present disclosure may be in the form of sterile injectable aqueous or oleaginous suspensions for intramuscular and subcutaneous administration. The suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a parenterally-acceptable, non-toxic diluent or solvent. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. Any blend fixed oil may be used for this purpose. In addition, fatty acids can also be prepared into injections.
The compounds of the present disclosure may be administered in the form of suppositories for rectal administration. These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid in the rectum and therefore will melt in the rectum to release the drug.
Dispersible powders and granules of the compounds of the present disclosure can be administered by the addition of water to prepare an aqueous suspension. These pharmaceutical compositions may be prepared by mixing the active ingredient with dispersing or wetting agents, suspending agents, or one or more preservatives.
As is well known to those skilled in the art, the dosage of a drug administered depends on a variety of factors, including, but not limited to: the activity of the particular compound used, the age of the patient, the weight of the patient, the health of the patient, the behavior of the patient, the diet of the patient, the time of administration, the mode of administration, the rate of excretion, the combination of drugs, the severity of the disease, and the like; in addition, the optimal treatment regimen, such as mode of treatment, daily amount of compound or type of pharmaceutically acceptable salt, can be verified according to conventional treatment protocols.
Description of the terms
Unless stated to the contrary, terms used in the specification and claims have the following meanings.
The term "alkyl" refers to a saturated straight or branched chain aliphatic hydrocarbon group, which is a straight or branched chain group containing 1 to 20 carbon atoms, preferably an alkyl (i.e., C) group containing 1 to 12 (e.g., 1,2, 3,4,5, 6,7,8, 9,10, 11, and 12) carbon atoms1-12Alkyl), more preferably an alkyl group having 1 to 6 carbon atoms (i.e., C)1-6Alkyl groups). Non-limiting examples of alkyl groups include: methyl group, ethyl group, n-propyl group, isopropyl group, n-butyl group, isobutyl group, tert-butyl group, sec-butyl group, n-pentyl group, 1-dimethylpropyl group, 1, 2-dimethylpropyl group, 2-dimethylpropyl group, 1-ethylpropyl group, 2-methylbutyl group, 3-methylbutyl group, n-hexyl group, 1-ethyl-2-methylpropyl group, 1, 2-trimethylpropyl group, 1-dimethylbutyl group, 1, 2-dimethylbutyl group, 2-dimethylbutyl group, 1, 3-dimethylbutyl group, 2-ethylbutyl group, 2-methylpentyl group, 3-methylpentyl group, 4-methylpentyl group, 2, 3-dimethylbutyl group, n-heptyl group, 2-methylhexyl group, 3-methylhexyl group, 4-methylhexyl group, 2-methylpentyl group, 3-methylhexyl group, 4-methylhexyl group, 2-dimethylpropyl group, 2-pentyl group, 2-methylpropyl group, 2-methyl-pentyl group, 3-pentyl group, 2-methyl-pentyl group, 2-pentyl group, and 3-pentyl group, 5-methylhexyl, 2, 3-dimethylpentyl, 2, 4-dimethylpentyl,2, 2-dimethylpentyl group, 3-dimethylpentyl group, 2-ethylpentyl group, 3-ethylpentyl group, n-octyl group, 2, 3-dimethylhexyl group, 2, 4-dimethylhexyl group, 2, 5-dimethylhexyl group, 2-dimethylhexyl group, 3-dimethylhexyl group, 4-dimethylhexyl group, 2-ethylhexyl group, 3-ethylhexyl group, 4-ethylhexyl group, 2-methyl-2-ethylpentyl group, 2-methyl-3-ethylpentyl group, n-nonyl group, 2-methyl-2-ethylhexyl group, 2-methyl-3-ethylhexyl group, 2-diethylpentyl group, n-decyl group, 3-diethylhexyl group, 2-diethylhexyl group, and various branched chain isomers thereof, and the like. Most preferably lower alkyl having 1 to 6 carbon atoms, non-limiting examples of which include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl and the like. The alkyl group may be substituted or unsubstituted, and when substituted, may be substituted at any available point of attachment, the substituents preferably being selected from one or more of H atoms, D atoms, halogens, alkyl groups, alkoxy groups, haloalkyl groups, haloalkoxy groups, cycloalkyloxy groups, heterocyclyloxy groups, hydroxy groups, hydroxyalkyl groups, oxo groups, cyano groups, amino groups, nitro groups, cycloalkyl groups, heterocyclyl groups, aryl groups and heteroaryl groups.
The term "heteroalkyl" refers to one or more-CH's in an alkyl group2-is selected from N, O, S, S (O) and S (O)2One or more of; wherein said alkyl is as defined above; heteroalkyl groups may be substituted or unsubstituted, and when substituted, may be substituted at any available point of attachment, with the substituents preferably being selected from one or more of D atoms, halogen, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, and heteroaryl.
The term "alkylene" refers to saturatedIs a residue derived from the parent alkane by removal of two hydrogen atoms from the same carbon atom or two different carbon atoms, and is a straight or branched chain group containing 1 to 20 carbon atoms, preferably 1 to 12 (e.g., 1,2, 3,4,5, 6,7,8, 9,10, 11, and 12) carbon atoms (i.e., C)1-12Alkylene) groups, more preferably alkylene groups having 1 to 6 carbon atoms (i.e., C)1-6Alkylene). Non-limiting examples of alkylene groups include, but are not limited to, methylene (-CH)2-), 1-ethylidene (-CH (CH)3) -), 1, 2-ethylene (-CH)2CH2) -, 1-propylene (-CH (CH)2CH3) -), 1, 2-propylene (-CH)2CH(CH3) -), 1, 3-propylene (-CH)2CH2CH2-) 1, 4-butylene (-CH2CH2CH2CH2-) and the like. The alkylene group may be substituted or unsubstituted and when substituted, may be substituted at any available point of attachment, the substituents preferably being selected from one or more of alkyl, alkenyl, alkynyl, alkoxy, haloalkoxy, cycloalkyloxy, heterocyclyloxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkoxy, heterocyclyloxy, cycloalkylthio, heterocyclylthio and oxo.
The term "heteroalkylene" refers to one or more-CH's in an alkylene group2-is selected from N, O, S, S (O) and S (O)2One or more of; wherein said alkyl is as defined above; the heteroalkylene group can be substituted or unsubstituted, and when substituted, can be substituted at any available point of attachment, with the substituents preferably being selected from one or more of a D atom, halogen, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, and heteroaryl.
The term "alkenyl" refers to an alkyl compound containing at least one carbon-carbon double bond in the molecule, wherein alkyl is as defined above. Preferably 2 to 12 (e.g. 2,3, 4,5, 6,7,8, 9,10, 1)Alkenyl of 1 and 12) carbon atoms (i.e. C)2-12Alkenyl), more preferably alkenyl having 2 to 6 carbon atoms (i.e., C)2-6Alkenyl). The alkenyl group may be substituted or unsubstituted, and when substituted, the substituent is preferably selected from one or more of a hydrogen atom, an alkyl group, an alkoxy group, a halogen, a haloalkyl group, a haloalkoxy group, a cycloalkyloxy group, a heterocyclyloxy group, a hydroxyl group, a hydroxyalkyl group, an oxo group, a cyano group, an amino group, a nitro group, a cycloalkyl group, a heterocyclyl group, an aryl group and a heteroaryl group.
The term "alkynyl" refers to an alkyl compound containing at least one carbon-carbon triple bond in the molecule, wherein alkyl is as defined above. Alkynyl (i.e., C) groups containing 2 to 12 (e.g., 2,3, 4,5, 6,7,8, 9,10, 11, and 12) carbon atoms are preferred2-12Alkynyl), more preferably an alkynyl group having 2 to 6 carbon atoms (i.e., C)2-6Alkynyl). The alkynyl group may be substituted or unsubstituted, and when substituted, the substituent is preferably selected from one or more of a hydrogen atom, an alkyl group, an alkoxy group, a halogen, a haloalkyl group, a haloalkoxy group, a cycloalkyloxy group, a heterocyclyloxy group, a hydroxyl group, a hydroxyalkyl group, a cyano group, an amino group, a nitro group, a cycloalkyl group, a heterocyclyl group, an aryl group and a heteroaryl group.
The term "cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring containing from 3 to 20 carbon atoms, preferably 3 to 12 (e.g., 3,4,5, 6,7,8, 9,10, 11, and 12) carbon atoms (i.e., 3 to 12 membered cycloalkyl groups), preferably 3 to 8 carbon atoms (i.e., 3 to 8 membered cycloalkyl groups), and more preferably 3 to 6 carbon atoms (i.e., 3 to 6 membered cycloalkyl groups). Non-limiting examples of monocyclic cycloalkyl groups include: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, and cyclooctyl, and the like; polycyclic cycloalkyl groups include spirocycloalkyl, fused ring alkyl, and bridged cycloalkyl groups.
The term "spirocycloalkyl" refers to a polycyclic group having 5 to 20 membered rings sharing one carbon atom (referred to as a spiro atom) between them, which may contain one or more double bonds. Preferably 6 to 14, more preferably 7 to 10 (e.g.7, 8, 9 or 10). Spirocycloalkyl groups are classified as mono-or polyspirocycloalkyl (e.g., a bispyridyl cycloalkyl group), preferably mono-and bispyridyl, depending on the number of spiro atoms shared between rings. More preferably 3-membered/5-membered, 3-membered/6-membered, 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered, 5-membered/6-membered, 6-membered/4-membered, 6-membered/5-membered or 6-membered/6-membered spirocycloalkyl. Non-limiting examples of spirocycloalkyl groups include:
Figure BDA0003210363510000281
the term "fused cyclic alkyl" refers to an all-carbon polycyclic group of 5 to 20 members sharing an adjacent pair of carbon atoms between the rings, wherein one or more of the rings may contain one or more double bonds. Preferably 6 to 14, more preferably 7 to 10 (e.g.7, 8, 9 or 10). They may be divided into polycyclic fused-ring alkyls such as bicyclic, tricyclic, tetracyclic, etc., according to the number of constituent rings, preferably bicyclic or tricyclic, more preferably 3-membered/4-membered, 3-membered/5-membered, 3-membered/6-membered, 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/3-membered, 5-membered/4-membered, 5-membered/5-membered, 5-membered/7-membered, 6-membered/3-membered, 6-membered/4-membered, 6-membered/5-membered, 6-membered/6-membered, 6-membered/7-membered, 7-membered/5-membered or 7-membered/6-membered bicycloalkyl. Non-limiting examples of fused ring alkyl groups include:
Figure BDA0003210363510000282
the term "bridged cycloalkyl" refers to a 5 to 20 membered all carbon polycyclic group in which any two rings share two carbon atoms not directly attached, which may contain one or more double bonds. Preferably 6 to 14, more preferably 7 to 10 (e.g.7, 8, 9 or 10). They may be classified into bicyclic, tricyclic, tetracyclic, etc. polycyclic bridged cycloalkyl groups according to the number of constituent rings, preferably bicyclic, tricyclic or tetracyclic bridged cycloalkyl groups, and more preferably bicyclic or tricyclic bridged cycloalkyl groups. Non-limiting examples of bridged cycloalkyl groups include:
Figure BDA0003210363510000283
said cycloalkyl ring includes cycloalkyl as described above(including monocyclic, spiro, fused and bridged rings) are fused to an aryl, heteroaryl or heterocycloalkyl ring wherein the ring(s) attached to the parent structure are cycloalkyl, non-limiting examples of which include
Figure BDA0003210363510000284
Etc.; preference is given to
Figure BDA0003210363510000285
Figure BDA0003210363510000286
Cycloalkyl groups may be substituted or unsubstituted, and when substituted, may be substituted at any available point of attachment, with the substituents preferably being selected from one or more of hydrogen, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, oxo, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
The term "alkoxy" refers to-O- (alkyl), wherein alkyl is as defined above. Non-limiting examples of alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, and the like. The alkoxy group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably selected from one or more of H atom, D atom, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, oxo, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
The term "heterocyclyl" refers to a saturated or partially unsaturated mono-or polycyclic cyclic substituent comprising from 3 to 20 ring atoms, one or more of which is a heteroatom selected from nitrogen, oxygen and sulfur, which may optionally be oxo (i.e., form a sulfoxide or sulfone), but does not include the ring portion of-O-, -O-S-or-S-, the remaining ring atoms being carbon. Preferably 3 to 12 (e.g., 3,4,5, 6,7,8, 9,10, 11 and 12) ring atoms (i.e., 3 to 12 membered heterocyclyl groups), of which 1-4 (e.g., 1,2, 3 and 4) are heteroatoms; more preferably 3 to 8 ring atoms (e.g., 3,4,5, 6,7, and 8), wherein 1-3 are heteroatoms (e.g., 1,2, and 3) (i.e., 3-to 8-membered heterocyclyl); more preferably 3 to 6 ring atoms, of which 1-3 are heteroatoms (i.e. 3 to 6 membered heterocyclyl); most preferably 5 or 6 ring atoms, of which 1 to 3 are heteroatoms (i.e. a 5 or 6 membered heterocyclyl group). Non-limiting examples of monocyclic heterocyclyl groups include pyrrolidinyl, tetrahydropyranyl, 1, 2.3.6-tetrahydropyridinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, and the like. Polycyclic heterocyclic groups include spiro, fused and bridged heterocyclic groups.
The term "spiroheterocyclyl" refers to a 5 to 20 membered polycyclic heterocyclic group in which one atom (referred to as the spiro atom) is shared between monocyclic rings, wherein one or more of the ring atoms is a heteroatom selected from nitrogen, oxygen, and sulfur, which may optionally be oxo (i.e., to form a sulfoxide or sulfone), with the remaining ring atoms being carbon. It may contain one or more double bonds. Preferably 6 to 14, more preferably 7 to 10 (e.g.7, 8, 9 or 10). Spiro heterocyclic groups are classified into a mono-spiro heterocyclic group or a multi-spiro heterocyclic group (e.g., a bis-spiro heterocyclic group), preferably a mono-spiro heterocyclic group and a bis-spiro heterocyclic group, according to the number of spiro atoms shared between rings. More preferably 3-membered/5-membered, 3-membered/6-membered, 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered, 5-membered/6-membered or 6-membered/6-membered mono spiroheterocyclyl. Non-limiting examples of spiro heterocyclic groups include:
Figure BDA0003210363510000291
the term "fused heterocyclyl" refers to a 5 to 20 membered polycyclic heterocyclic group in which the rings share an adjacent pair of atoms between them, one or more of the rings may contain one or more double bonds in which one or more of the ring atoms is a heteroatom selected from nitrogen, oxygen and sulfur, which may optionally be oxo (i.e., to form a sulfoxide or sulfone), and the remaining ring atoms are carbon. Preferably 6 to 14, more preferably 7 to 10 (e.g.7, 8, 9 or 10). They may be classified into bicyclic, tricyclic, tetracyclic, etc. polycyclic fused heterocyclic groups according to the number of constituting rings, preferably bicyclic or tricyclic, more preferably 3-membered/4-membered, 3-membered/5-membered, 3-membered/6-membered, 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/3-membered, 5-membered/4-membered, 5-membered/6-membered, 5-membered/7-membered, 6-membered/3-membered, 6-membered/4-membered, 6-membered/5-membered, 6-membered/6-membered, 6-membered/7-membered, 7-membered/5-membered or 7-membered/6-membered bicyclic fused heterocyclic groups. Non-limiting examples of fused heterocyclic groups include:
Figure BDA0003210363510000301
the term "bridged heterocyclyl" refers to a 5 to 14 membered polycyclic heterocyclic group in which any two rings share two atoms not directly attached, which may contain one or more double bonds, wherein one or more of the ring atoms is a heteroatom selected from nitrogen, oxygen and sulfur, which may optionally be oxo (i.e., to form a sulfoxide or sulfone), and the remaining ring atoms are carbon. Preferably 6 to 14, more preferably 7 to 10 (e.g.7, 8, 9 or 10). They may be classified into bicyclic, tricyclic, tetracyclic, etc. polycyclic bridged heterocyclic groups according to the number of constituent rings, preferably bicyclic, tricyclic, or tetracyclic bridged heterocyclic groups, and more preferably bicyclic or tricyclic bridged heterocyclic groups. Non-limiting examples of bridged heterocyclic groups include:
Figure BDA0003210363510000302
the heterocyclyl ring includes a heterocyclyl (including monocyclic, spiroheterocyclic, fused heterocyclic and bridged heterocyclic) fused to an aryl, heteroaryl or cycloalkyl ring as described above, wherein the ring to which the parent structure is attached is a heterocyclyl, non-limiting examples of which include:
Figure BDA0003210363510000303
and the like.
The heterocyclyl group may be substituted or unsubstituted and when substituted may be substituted at any available point of attachment, the substituents preferably being selected from one or more of hydrogen, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, oxo, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
The term "aryl" refers to a 6 to 14 membered all carbon monocyclic or fused polycyclic (fused polycyclic is 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 those wherein the aryl ring as described above is fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring attached to the parent structure is an aryl ring, non-limiting examples of which include:
Figure BDA0003210363510000311
aryl groups may be substituted or unsubstituted, and when substituted, may be substituted at any available point of attachment, with the substituents preferably being selected from one or more of hydrogen, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, oxo, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
The term "heteroaryl" refers to a heteroaromatic system comprising 1 to 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, for example furyl, thienyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl and the like. The heteroaryl ring includes a heteroaryl fused to an aryl, heterocyclyl or cycloalkyl ring as described above, wherein the ring joined together with the parent structure is a heteroaryl ring, non-limiting examples of which include:
Figure BDA0003210363510000312
heteroaryl groups may be substituted or unsubstituted, and when substituted, may be substituted at any available point of attachment, with the substituents preferably being selected from one or more of hydrogen, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, oxo, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
The above-mentioned cycloalkyl, heterocyclyl, aryl and heteroaryl groups include those derived by removal of one hydrogen atom from the parent ring atom, or those derived by removal of two hydrogen atoms from the parent ring atom or two different ring atoms, i.e., "divalent cycloalkyl", "divalent heterocyclyl", "arylene" and "heteroarylene".
The term "amino protecting group" is intended to protect an amino group with a group that can be easily removed in order to keep the amino group unchanged when the rest of the molecule is subjected to a reaction. Non-limiting examples include: (trimethylsilyl) ethoxymethyl, tetrahydropyranyl, t-butoxycarbonyl, acetyl, benzyl, allyl, and p-methoxybenzyl, and the like. These groups may be optionally substituted with 1 to 3 substituents selected from halogen, alkoxy or nitro.
The term "hydroxyl-protecting group" is a suitable group for hydroxyl protection known in the art, see the literature ("Protective Groups in Organic Synthesis", 5)Th Ed.T.W.Greene&P.g.m.wuts). By way of example, the hydroxyl protecting group may preferably be (C)1-10Alkyl or aryl)3Silane groups, for example: triethylsilyl, triisopropylsilyl, tert-butyldimethylsilyl, tert-butyldiphenylsilyl and the like; may be C1-10Alkyl or substituted alkyl, preferably alkoxy or aryl substituted alkyl, more preferably C1-6Alkoxy-substituted C1-6Alkyl or phenyl substituted C1-6Alkyl, most preferably C1-4Alkoxy-substituted C1-4Alkyl groups, for example: methyl, t-butyl, benzyl, methoxymethyl (MOM), ethoxyethyl, and the like; may be (C)1-10Alkyl or aryl) acyl groups, such as: formyl, acetyl, benzoyl, p-nitrobenzoyl and the like; may be (C)1-6Alkyl or C6-10Aryl) sulfonyl; or (C)1-6Alkoxy or C6-10Aryloxy) carbonyl; it may also be allyl, 2-Tetrahydropyranyl (THP).
The term "cycloalkyloxy" refers to cycloalkyl-O-wherein cycloalkyl is as defined above.
The term "heterocyclyloxy" refers to heterocyclyl-O-, wherein heterocyclyl is as defined above.
The term "aryloxy" refers to aryl-O-wherein aryl is as defined above.
The term "heteroaryloxy" refers to heteroaryl-O-, wherein heteroaryl is as defined above.
The term "alkylthio" refers to an alkyl-S-group wherein alkyl is as defined above.
The term "haloalkyl" refers to an alkyl group substituted with one or more halogens, wherein alkyl is as defined above.
The term "haloalkoxy" refers to an alkoxy group substituted with one or more halogens, wherein the alkoxy group is as defined above.
The term "deuterated alkyl" refers to an alkyl group substituted with one or more deuterium atoms, wherein alkyl is as defined above.
The term "hydroxyalkyl" refers to an alkyl group substituted with one or more hydroxyl groups, wherein alkyl is as defined above.
The term "halogen" refers to fluorine, chlorine, bromine or iodine.
The term "hydroxy" refers to-OH.
The term "mercapto" refers to-SH.
The term "amino" refers to the group-NH2
The term "cyano" refers to — CN.
The term "nitro" means-NO2
The term "oxo" or "oxo" refers to "═ O".
The term "carbonyl" refers to C ═ O.
The term "aldehyde group" refers to-C (O) H;
the term "carboxy" refers to-C (O) OH.
The term "carboxylate" refers to-C (O) O (alkyl), -C (O) O (cycloalkyl), (alkyl) C (O) O-or (cycloalkyl) C (O) O-, wherein alkyl and cycloalkyl are as defined above.
The compounds of the present disclosure may also comprise isotopic derivations thereofA compound (I) is provided. The term "isotopic derivative" refers to a compound that differs in structure only in the presence of one or more isotopically enriched atoms. For example, having the structure of the present disclosure except that "deuterium" or "tritium" is substituted for hydrogen, or18F-fluorine labeling: (18Isotope of F) instead of fluorine, or with11C-、13C-, or14C-enriched carbon (C11C-、13C-, or14C-carbon labeling;11C-、13c-, or14C-isotopes) instead of carbon atoms are within the scope of the present disclosure. Such compounds are useful as analytical tools or probes in, for example, biological assays, or as tracers for in vivo diagnostic imaging of disease, or as tracers for pharmacodynamic, pharmacokinetic or receptor studies. The disclosure also includes various deuterated forms of the compounds. Each available hydrogen atom attached to a carbon atom may be independently replaced by a deuterium atom. The person skilled in the art is able to synthesize the deuterated forms of the compounds with reference to the relevant literature. Commercially available deuterated starting materials can be used in preparing the deuterated forms of the compounds, or they can be synthesized using conventional techniques using deuterated reagents including, but not limited to, deuterated boranes, trideuteroborane in tetrahydrofuran, deuterated lithium aluminum hydrides, deuterated iodoethanes, and deuterated iodomethanes, among others. Deuterations can generally retain activity comparable to non-deuterated compounds and can achieve better metabolic stability when deuterated at certain specific sites, thereby achieving certain therapeutic advantages.
In the chemical structure of the compounds described in the present disclosure, a bond
Figure BDA0003210363510000331
Denotes an unspecified configuration, i.e. a bond if a chiral isomer is present in the chemical structure
Figure BDA0003210363510000334
Can be that
Figure BDA0003210363510000335
Or
Figure BDA0003210363510000336
Or at the same time contain
Figure BDA0003210363510000332
And
Figure BDA0003210363510000333
two configurations.
"optional" or "optionally" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs or does not. For example, "a heterocyclic group optionally substituted with an alkyl" means that an alkyl may, but need not, be present, and the description includes the case where the heterocyclic group is substituted with an alkyl and the heterocyclic group is not substituted with an alkyl.
"substituted" means that one or more, preferably 1 to 5, more preferably 1 to 3, hydrogen atoms in the group are independently substituted with a corresponding number of substituents. Those skilled in the art are able to ascertain (by experiment or theory) without undue effort, substitutions that are possible or impossible. For example, amino or hydroxyl groups having free hydrogen may be unstable in combination with carbon atoms having unsaturated (e.g., olefinic) bonds.
"pharmaceutical composition" means a mixture containing one or more compounds described herein or a physiologically/pharmaceutically acceptable salt or prodrug thereof in admixture with other chemical components, as well as other components such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of the active ingredient and exert biological activity.
"pharmaceutically acceptable salts" refers to salts of the disclosed compounds which are safe and effective for use in a mammalian body and which possess the requisite biological activity. Salts may be prepared separately during the final isolation and purification of the compound, or by reacting the appropriate group with an appropriate base or acid. Bases commonly used to form pharmaceutically acceptable salts include inorganic bases such as sodium hydroxide and potassium hydroxide, and organic bases such as ammonia. Acids commonly used to form pharmaceutically acceptable salts include inorganic acids as well as organic acids.
The term "therapeutically effective amount" with respect to a drug or pharmacologically active agent refers to a sufficient amount of the drug or agent that is non-toxic but achieves the desired effect. The 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, and an appropriate effective amount in an individual case can be determined by a person skilled in the art according to routine tests.
The term "solvate" as used herein refers to a physical association of a compound of the present disclosure with one or more, preferably 1-3, solvent molecules, whether organic or inorganic. The physical bonding includes hydrogen bonding. In some cases, for example, when one or more, preferably 1-3, solvent molecules are incorporated into the crystal lattice of a crystalline solid, the solvate will be isolated. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Solvation methods are well known in the art.
By "prodrug" is meant a compound that can be converted in vivo under physiological conditions, for example, by hydrolysis in blood, to yield the active parent compound.
The term "pharmaceutically acceptable" as used herein 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 effective for the intended use.
As used herein, the singular forms "a", "an" and "the" include plural references and vice versa unless the context clearly dictates otherwise.
When the term "about" is applied to a parameter such as pH, concentration, temperature, etc., it is meant that the parameter may vary by ± 10%, and sometimes more preferably within ± 5%. As will be appreciated by those skilled in the art, when the parameters are not critical, the numbers are generally given for illustrative purposes only and are not limiting.
Synthesis of the Compounds of the disclosure
In order to achieve the purpose of the present disclosure, the present disclosure adopts the following technical solutions:
scheme one
The preparation method of the compound shown in the general formula (III) or the tautomer, the racemate, the enantiomer, the diastereoisomer or the mixture form thereof or the pharmaceutically acceptable salt thereof comprises the following steps:
Figure BDA0003210363510000351
(a) a compound of the general formula (IIIaa) or a tautomer, racemate, enantiomer, diastereomer or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein the protecting group R is removed in the presence of an acidWTo obtain a compound of formula (IIIa) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
(b) carrying out reductive amination reaction on the compound of the general formula (IIIa) or a tautomer, a racemate, an enantiomer, a diastereomer or a mixture thereof or a pharmaceutically acceptable salt thereof and the compound of the general formula (V) or the pharmaceutically acceptable salt thereof in the presence of a reducing agent under alkaline conditions to obtain the compound of the general formula (III) or the tautomer, the racemate, the enantiomer, the diastereomer or a mixture thereof or the pharmaceutically acceptable salt thereof;
or
Carrying out coupling reaction on the compound of the general formula (IIIa) or a tautomer, a racemate, an enantiomer, a diastereomer or a mixture form thereof or a pharmaceutically acceptable salt thereof and the compound of the general formula (V') to obtain the compound of the general formula (III) or the tautomer, the racemate, the enantiomer, the diastereomer or a mixture form thereof or a pharmaceutically acceptable salt thereof;
wherein:
RWis an amino protecting group; preferably, RWIs tert-butyloxycarbonyl;
L3is a bond;
ring B is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom;
y is halogen; preferably, Y is chloro;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
ring C, R1To R3、R5、R2aS, t and v are as defined in formula (III).
Scheme two
The preparation method of the compound shown in the general formula (IV) or a tautomer, a racemate, an enantiomer, a diastereoisomer or a mixture form thereof, or a pharmaceutically acceptable salt thereof comprises the following steps:
Figure BDA0003210363510000361
(a) a compound of the general formula (IVaa) or a tautomer, racemate, enantiomer, diastereomer or mixture thereof, or a pharmaceutically acceptable salt thereof, in the presence of an acid to remove the protecting group RWTo obtain a compound of general formula (IVa) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
(b) the compound of the general formula (IVa) or the tautomer, the racemate, the enantiomer, the diastereomer or the mixture form or the pharmaceutically acceptable salt thereof and the compound of the general formula (VI) or the pharmaceutically acceptable salt thereof are subjected to reductive amination reaction under the alkaline condition and in the presence of a reducing agent to obtain R10Selected from the group consisting of hydrogen atoms, alkyl groups, heteroalkyl groups, haloalkyl groups, hydroxyalkyl groups and RWA compound of general formula (IV) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
or
A compound of formula (IVa) or a tautomer, thereofRacemic modification, enantiomer, diastereoisomer or mixture thereof, or pharmaceutically acceptable salt thereof, with a compound of formula (VI') to give R10Selected from the group consisting of hydrogen atoms, alkyl groups, heteroalkyl groups, haloalkyl groups, hydroxyalkyl groups and RWA compound of general formula (IV) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
and
optionally, when R10Is RWWhen R is the same as R, the method further comprises the step (c) of10Is RWIn the form of a compound of the general formula (IV) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, in the presence of an acid to remove the protecting group RWTo obtain R10A compound of formula (IV) being a hydrogen atom or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
wherein:
RWis an amino protecting group; preferably, RWIs tert-butyloxycarbonyl;
L3is a bond;
ring C is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom or
Figure BDA0003210363510000371
Ring D is a 4-to 6-membered heterocyclic group containing at least one nitrogen atom;
Z1、Z2、Z3and Z4One is a carbon atom and the remaining three are the same or different and are each independently CR5Or an N atom;
y is halogen; preferably, Y is chloro;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
R1、R3、R5s, t and v are as in formula (IV)As defined.
In the above reaction of steps (a) and (c), the acid includes organic acid and inorganic acid, and the organic acid includes, but is not limited to, trifluoroacetic acid, formic acid, acetic acid, methanesulfonic acid, p-toluenesulfonic acid, Me3SiCl and TMSOTf, preferably trifluoroacetic acid; such inorganic acids include, but are not limited to, hydrogen chloride, 1, 4-dioxane solution of hydrogen chloride, hydrochloric acid, sulfuric acid, nitric acid, and phosphoric acid.
In the reaction of step (b), the reducing agent used in the reductive amination reaction includes, but is not limited to, sodium acetate borohydride, sodium triacetoxyborohydride, sodium borohydride, lithium borohydride, sodium cyanoborohydride, sodium acetylborohydride, and the like, and preferably sodium acetate borohydride. The base includes organic bases including but not limited to triethylamine, N-diisopropylethylamine, N-butyllithium, lithium diisopropylamide, sodium acetate, potassium acetate, sodium ethoxide, sodium tert-butoxide, and potassium tert-butoxide, and inorganic bases including but not limited to sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide monohydrate, lithium hydroxide, and potassium hydroxide; sodium acetate is preferred.
In the above reaction of step (b), the coupling reaction is well known in the art and includes, but is not limited to, Buchwald-Hartwig coupling reaction, Negishi coupling reaction, etc. The coupling reaction conditions include, but are not limited to, in the presence of a metal catalyst (preferably in the presence of a palladium or nickel containing catalyst) including, but not limited to, 1, 3-bis (2, 6-di-3-pentylphenyl) imidazol-2-ylidene](3-Chloropyridyl) palladium (II) dichloride, chlorine (2-dicyclohexylphosphino-2 ',6' -di-isopropoxy-1, 1 '-biphenyl) (2-amino-1, 1' -biphenyl-2-yl) palladium (II), palladium acetate, methanesulfonic acid (2-dicyclohexylphosphino-2 ',6' -diisopropoxy-1, 1 '-biphenyl) (2-amino-1, 1' -biphenyl-2-yl) palladium (II) (RuPhos Pd G3), methanesulfonic acid (2-dicyclohexylphosphino-2 ',4',6 '-tri-isopropyl-1, 1' -biphenyl) (2 '-amino-1, 1' -biphenyl-2-yl) palladium (II) (XPhos Pd G3), Chloro (2-dicyclohexylphosphino-2 ',6' -di-isopropoxy-1, 1 '-biphenyl) (2-amino-1, 1' -biphenyl-2-yl) palladium (II) (RuPhos Pd G2), organozinc reagent, bis (triphenylphosphine) chloridePalladium (II) (Pd (PPh)3)Cl2) Etc., preferably 1, 3-bis (2, 6-di-3-pentylphenyl) imidazol-2-ylidene](3-Chloropyridyl) palladium (II) dichloride. The metal catalyst may also be used in combination with a ligand including, but not limited to, S- (-) -1,1' -binaphthyl-2, 2' -bisdiphenylphosphine (S- (-) -BINAP), R- (-) -1,1' -binaphthyl-2, 2' -bisdiphenylphosphine (R- (-) -BINAP), 1' -binaphthyl-2, 2' -Bisdiphenylphosphine (BINAP), 1' -bis (diphenylphosphino) ferrocene (DPPF), 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (XANTPHOS), 2' -bis (diphenylphosphino) benzophenone (DPBP), 2' -diallylbisphenol a (dba), and the like, and a base. The base includes organic bases including but not limited to triethylamine, N-diisopropylethylamine, N-butyllithium, lithium diisopropylamide, sodium acetate, potassium acetate, sodium ethoxide, sodium tert-butoxide, and potassium tert-butoxide, and inorganic bases including but not limited to sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide monohydrate, lithium hydroxide, and potassium hydroxide; cesium carbonate is preferred.
The above-described reactions of steps (a) - (c) are preferably carried out in a solvent, including but not limited to: ethylene glycol dimethyl ether, acetic acid, methanol, ethanol, acetonitrile, N-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, N-hexane, dimethyl sulfoxide, 1, 4-dioxane, water, N-dimethylformamide, N-dimethylacetamide, 1, 2-dibromoethane, and mixtures thereof.
Detailed Description
The present disclosure is further described below with reference to examples, but these examples do not limit the scope of the present disclosure.
Examples
The structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) or/and Mass Spectrometry (MS). NMR shift (. delta.) of 10-6The units in (ppm) are given. NMR was measured using Bruker AVANCE NEO 500M in deuterated dimethyl sulfoxide (DMSO-d)6) Deuterated chloroform (CDCl)3) Deuterated methanol (CD)3OD), internal standard Tetramethylsilane (TMS).
MS was measured using an Agilent 1200/1290DAD-6110/6120Quadrupole MS LC MS (manufacturer: Agilent, MS model: 6110/6120Quadrupole MS), waters ACQuity UPLC-QD/SQD (manufacturer: waters, MS model: waters ACQuity Qda Detector/waters SQ Detector), THERMO Ultratate 3000-Q active (manufacturer: THERMO, MS model: THERMO Q active).
High Performance Liquid Chromatography (HPLC) analysis was performed using Agilent HPLC1200 DAD, Agilent HPLC1200VWD and Waters HPLC e2695-2489 HPLC.
Chiral HPLC assay using Agilent 1260DAD HPLC.
High Performance liquid preparation A preparative chromatograph was used from Waters 2767, Waters 2767-SQ Detector 2, Shimadzu LC-20AP and Gilson-281.
Chiral preparation was performed using Shimadzu LC-20AP preparative chromatograph.
The CombiFlash rapid preparation instrument uses CombiFlash Rf200(TELEDYNE ISCO).
The thin layer chromatography silica gel plate adopts HSGF254 of tobacco yellow sea or GF254 of Qingdao, the specification of the silica gel plate used by Thin Layer Chromatography (TLC) is 0.15 mm-0.2 mm, and the specification of the thin layer chromatography separation and purification product is 0.4 mm-0.5 mm.
Silica gel column chromatography generally uses 200-300 mesh silica gel of the Tibet Huanghai silica gel as a carrier.
Average inhibition rate of kinase and IC50The values were determined with a NovoStar microplate reader (BMG, Germany).
Known starting materials of the present disclosure may be synthesized using or according to methods known in the art, or may be purchased from companies such as ABCR GmbH & co.kg, Acros Organics, Aldrich Chemical Company, nephelo Chemical science and technology (Accela ChemBio Inc), dare chemicals, and the like.
In the examples, the reaction can be carried out in an argon atmosphere or a nitrogen atmosphere, unless otherwise specified.
An argon atmosphere or nitrogen atmosphere means that the reaction flask is connected to a balloon of argon or nitrogen with a volume of about 1L.
The hydrogen atmosphere refers to a reaction flask connected with a hydrogen balloon with a volume of about 1L.
The pressure hydrogenation reaction used a hydrogenation apparatus of Parr 3916EKX type and a hydrogen generator of Qinglan QL-500 type or a hydrogenation apparatus of HC2-SS type.
The hydrogenation reaction was usually evacuated and charged with hydrogen and repeated 3 times.
The microwave reaction was carried out using a CEM Discover-S908860 type microwave reactor.
In the examples, the solution means an aqueous solution unless otherwise specified.
In the examples, the reaction temperature is, unless otherwise specified, from 20 ℃ to 30 ℃ at room temperature.
The monitoring of the progress of the reaction in the examples employed Thin Layer Chromatography (TLC), a developing solvent used for the reaction, a system of eluents for column chromatography used for purifying compounds and a developing solvent system for thin layer chromatography including: a: n-hexane/ethyl acetate system, B: the volume ratio of the solvent in the dichloromethane/methanol system is adjusted according to the polarity of the compound, and a small amount of basic or acidic reagents such as triethylamine, acetic acid and the like can be added for adjustment.
Example 1
5- (2, 6-dimethylpyridin-4-yl) -9- (1 '-isobutyl- [1,4' -dipiperidine ] -4-yl) -1,3,4, 6-tetrahydroxepin-o [5,4,3-cd ] indole bistrifluoroacetate 1
Figure BDA0003210363510000391
Figure BDA0003210363510000401
First step of
Triethyl (4- ((tetrahydro-2H-pyran-2-yl) oxy) but-1-yn-1-yl) silane 1b
2- (but-3-yn-1-yloxy) tetrahydro-2H-pyran 1a (11.3g, 73.3mmol, Toshibara) was dissolved in anhydrous tetrahydrofuran (130mL), cooled to-70 ℃ and a 2.5M n-hexane solution of n-butyllithium (31mL, 77.7mmol, Toshitanan) was added dropwise. The reaction was stirred at-70 ℃ for 1 hour under nitrogen. Triethylchlorosilane (12.3g, 82.2mmol, obtained from Shanghai) was added. Stirred at room temperature for 4 hours. Jia Jian FuAnd aqueous ammonium chloride (80mL), and the organic phase was separated. The aqueous phase was extracted with dichloromethane (80 mL. times.2), the organic phases were combined, concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 1b (19.0g, yield: 96.6%).1H NMR(500MHz,CDCl3)δ4.66(t,1H),3.91-3.79(m,2H),3.56-3.48(m,2H),2.53(t,2H),1.86-1.79(m,1H),1.72-1.66(m,1H),1.61-1.47(m,4H),0.96(t,9H),0.56(q,6H)。
Second step of
4- (triethylsilyl) but-3-yn-1-ol 1c
Compound 1b (19.0g, 70.8mmol) was dissolved in methanol (170mL) and 4-toluenesulfonic acid monohydrate (1.4g, 7.1mmol, Shaoshima science) was added. Stirred at room temperature for 16 hours. Concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 1c (10.9g, yield: 83.9%).
1H NMR(500MHz,CDCl3)δ3.70(t,2H),2.51(t,2H),1.98(s,1H),0.97(t,9H),0.58(q,6H)。
The third step
Triethyl (4- ((2-iodo-3-nitrobenzyl) oxy) but-1-yn-1-yl) silane 1e
Sodium hydride (663.1mg, 16.6mmol, 60%, Shanghai Tatan) was suspended in anhydrous tetrahydrofuran (60 mL). At 0 deg.C, compound 1c (3.1g, 16.6mmol) was added and stirred at room temperature for 20 minutes. 1- (bromomethyl) -2-iodo-3-nitrobenzene 1d (4.4g, 12.8mmol, prepared using methods known in the literature "Heterocycles, 2018, vol.97, p.1175-1190") was added. The reaction was stirred at room temperature for 16 hours. Saturated aqueous ammonia chloride (50mL), ethyl acetate (50 mL. times.3) were added, the mixture was extracted, concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 1e (3.8g, yield: 66.9%).
MS m/z(ESI):445.9[M+1]。
The fourth step
2-iodo-3- (((4- (triethylsilyl) but-3-yn-1-yl) oxy) methyl) aniline 1f
Compound 1e (3.8g, 8.5mmol) was dissolved in ethanol (60mL) and water (12mL), and iron powder (3.4g, 59.8mmol, Shanghai medicine) and ammonium chloride (3.4g, 59.8mmol, Shanghai medicine) were added. The reaction was stirred at 80 ℃ for 1.5 hours. Concentrated under reduced pressure, extracted with ethyl acetate (50 mL. times.3), and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 1f (3.1g, yield: 87.5%).
MS m/z(ESI):416.0[M+1]。
The fifth step
4-bromo-2-iodo-3- (((4-triethylsilyl) but-3-yn-1-yl) oxy) methyl) aniline 1g
Compound 1f (4.6g, 11.0mmol) was dissolved in N, N-dimethylformamide (70mL) and N-bromosuccinimide (2.0g, 11.0mmol, Shanghai nationality drug) was added. Stirred at room temperature for 1 hour. Aqueous sodium thiosulfate (140mL) was added and extracted with ethyl acetate (50 mL. times.3). The organic phases were combined and washed with water (30 mL. times.3). The organic phase was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to obtain 1g (5.1g, yield: 95.2%) of the title product.
MS m/z(ESI):493.9[M+1]。
The sixth step
9-bromo-5- (triethylsilyl) -1,3,4, 6-tetrahydroxyhepto [5,4,3-cd ] indole for 1h
1g (4.4g, 8.9mmol) of the compound was dissolved in N, N-dimethylformamide (540mL), and palladium acetate (997.1mg, 4.5mmol, Shanghai Tantan), triphenylphosphine (2.4g, 9.0mmol, Shanghai nationality medicine), lithium chloride (1.2g, 26.9mmol, Shanghai nationality medicine) and potassium carbonate (3.7g, 26.7mmol, Shanghai nationality test) were added. The nitrogen was replaced three times. Stirring is carried out for 16 hours at 80 ℃ under the protection of nitrogen. Water (1L) was added, and extraction was performed with ethyl acetate (300 mL. times.3). The organic phases were combined and washed with water (100 mL. times.3). The organic phase was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 1h (830mg, yield: 25.2%).
MS m/z(ESI):366.0[M+1]。
Seventh step
4- (5- (triethylsilyl) -1,3,4, 6-tetrahydroxyhepto [5,4,3-cd ] indol-9-yl) -3, 6-dihydropyridine-1 (2H) carboxylic acid tert-butyl ester 1j
Compound 1h (830.0mg, 2.3mmol) and compound 1i (1.0g, 3.4mmol, Shaoshima science) were dissolved in 1, 4-dioxane (20mL) and water (4 mL). Bis [ di-tert-butyl- (4-dimethylaminophenyl) phosphine ] palladium dichloride (322.0mg, 0.45mmol, Shanghai Biao) and cesium carbonate (1.5g, 4.6mmol, Shaoshima science) were added. The nitrogen was replaced three times, and the reaction was stirred at 80 ℃ for 3 hours under nitrogen protection. Water (20mL) was added, extraction was performed with ethyl acetate (20 mL. times.3), concentration was performed under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 1j (920.0mg, yield: 88.6%).
MS m/z(ESI):469.2[M+1]。
Eighth step
4- (5- (triethylsilyl) -1,3,4, 6-tetrahydroxyhepto [5,4,3-cd ] indol-9-yl) piperidine-1-carboxylic acid tert-butyl ester 1k
Compound 1j (820.0mg, 1.75mmol) was dissolved in methanol (50 mL). 10% palladium on carbon (800.0mg, water content < 1%, Shanghai Wolka) was added. The hydrogen was replaced three times. The reaction was stirred at 40 ℃ for 1 hour under hydrogen. Suction filtration is carried out, 10 percent palladium carbon (800.0mg, the water content is less than 1 percent, and Shanghai Wolka) is added into the filtrate. The hydrogen was replaced three times. The reaction was stirred at 40 ℃ for 3 hours under hydrogen. Suction filtration and concentration of the filtrate under reduced pressure, and purification of the resulting residue by silica gel column chromatography with eluent system A gave the title product 1k (335.0mg, yield: 40.7%).
MS m/z(ESI):471.2[M+1]。
The ninth step
4- (5-bromo-1, 3,4, 6-tetrahydroxepin [5,4,3-cd ] indol-9-yl) piperidine-1-carboxylic acid tert-butyl ester 1l
Compound 1k (275.0mg, 0.6mmol) was dissolved in 1, 2-dichloroethane (28 mL). A solution of N-bromosuccinimide (2.0g, 11.0mmol, Shanghai nationality drug) in 1, 2-dichloroethane (10mL) was slowly added dropwise at-30 ℃. The reaction was stirred for 10 minutes at-30 ℃. Aqueous sodium thiosulfate (30mL) was added and extracted with dichloromethane (30 mL. times.3). The organic phase was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 1l (145.0mg, yield: 57.0%).
MS m/z(ESI):457.0[M+23]。
The tenth step
4- (5- (2, 6-dimethylpyridin-4-yl) -1,3,4, 6-tetrahydroxepin [5,4,3-cd ] indol-9-yl) piperidine-1-carboxylic acid tert-butyl ester 1n
Compound 1l (195.0mg, 0.45mmol) and 2, 6-dimethyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine 1m (88.0mg, 0.58mmol, Shanghai Yam kang) were dissolved in 1, 4-dioxane (8mL) and water (2 mL). [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (73.3mg, 0.09mmol, Beijing carbofuran) and potassium phosphate (284.9mg, 1.3mmol, Beijing carbofuran) were added. The nitrogen was replaced three times, and the reaction was stirred at 90 ℃ for 3 hours. Water (10mL) was added, extraction was performed with ethyl acetate (20 mL. times.3), concentration was performed under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 1n (150.0mg, yield: 72.6%).
MS m/z(ESI):462.2[M+1]。
The eleventh step
5- (2, 6-dimethylpyridin-4-yl) -9- (piperidin-4-yl) -1,3,4, 6-tetrahydroxepin [5,4,3-cd ] indole 1o
Compound 1n (75.0mg, 0.16mmol) was dissolved in methanol (2mL) and 4M hydrogen chloride/1, 4-dioxane solution (3mL) was added. Stirred at room temperature for 1 hour. The reaction mixture was concentrated under reduced pressure, and 7M ammonia/methanol solution (5mL) was added to the residue. Concentration under reduced pressure gave the title product 1o (58.0mg, yield: 100%).
MS m/z(ESI):362.1[M+1]。
The twelfth step
5- (2, 6-dimethylpyridin-4-yl) -9- (1 '-isobutyl- [1,4' -dipiperidine ] -4-yl) -1,3,4, 6-tetrahydroxepin-o [5,4,3-cd ] indole bistrifluoroacetate 1
Compound 1o (58.0mg, 0.16mmol) and 1-isobutylpiperidin-4-one 1p (155.2mg, 0.17mmol) were dissolved in anhydrous N, N-dimethylformamide (5mL), and sodium acetate (223.0mg, 2.7mmol, Shanghaitant) was added. Stirred at room temperature for 1 hour. Sodium borohydride acetate (223.0mg, 2.7mmol, Shaoshima technologies Shanghai, Inc.) was added. Stirred at room temperature for 16 hours. The residue was isolated and purified by high performance liquid chromatography (column: Sharpsil-T C18150 x 30mm,5 μm; mobile phase 1: water (containing 0.1% trifluoroacetic acid), mobile phase 2: acetonitrile; 15 min gradient: 10% -95%, flow rate: 30mL/min) to give the title product 1(30.0mg, yield: 30.5%). MS M/z (ESI) 501.3[ M-227 ].
1H NMR(500MHz,CD3OD)δ7.86(s,2H),7.41(d,1H),7.25(d,1H),5.30(s,2H),4.17(t,2H),3.83-3.62(m,6H),3.42(t,2H),3.11-3.01(m,5H),2.81-2.72(m,6H),2.46(d,2H),2.25-2.06(m,8H),1.07(d,6H)。
Example 2
2- (2, 6-Dimethylpyridin-4-yl) -6- (1 '-isobutyl- [1,4' -dipiperidine ] -4-yl) -1, 5-dihydro-3H-pyrano [3,4,5-cd ] indole bistrifluoroacetate 2
Figure BDA0003210363510000431
Figure BDA0003210363510000441
First step of
Triethyl (3- ((2-iodo-3-nitrobenzyl) oxy) prop-1-yn-1-yl) silane 2b
1d (2.2g, 6.4mmol, prepared by methods known in the literature "Heterocycles, 2018, vol.97, p.1175-1190") and 3- (triethylsilyl) prop-2-yn-1-ol 2a (1.7g, 9.7mmol, prepared by methods known in the literature "Tetrahedron, 1996, vol.52, p.7487-7510") were dissolved in acetonitrile (25mL) and potassium carbonate (4.5g, 32.2mmol, Shanghai medicine) was added. The reaction was stirred at 80 ℃ for 16 hours. Concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 2b (1.6g, yield: 57.7%).
MS m/z(ESI):432.1[M+1]。
Second step of
2-iodo-3- (((3- (triethylsilyl) prop-2-yn-1-yl) oxy) methyl) aniline 2c
Compound 2b (1.6g, 3.7mmol) was dissolved in ethanol (25mL) and water (5mL), and iron powder (1.5g, 26.8mmol, Shanghai medicine) and ammonium chloride (1.5g, 26.8mmol, Shanghai medicine) were added. The reaction was stirred at 80 ℃ for 1.5 hours. Concentrated under reduced pressure, extracted with ethyl acetate (30 mL. times.3), and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 2c (1.1g, yield: 73.9%).
MS m/z(ESI):402.0[M+1]。
The third step
4-bromo-2-iodo-3- (((3- (triethylsilyl) prop-2-yn-1-yl) oxy) methyl) aniline 2d
Compound 2c (1.1g, 2.7mmol) was dissolved in N, N-dimethylformamide (18mL) and N-bromosuccinimide (487.8g, 2.7mmol, Shanghai nationality) was added. Stirred at room temperature for 1 hour. Aqueous sodium thiosulfate (40mL) was added and extracted with ethyl acetate (50 mL. times.3). The organic phases were combined and washed with water (20 mL. times.3). The organic phase was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 2d (1.1g, yield: 85.0%).
MS m/z(ESI):479.9[M+1]。
The fourth step
6-bromo-2- (triethylsilyl) -1, 5-dihydro-3H-pyrano [3,4,5-cd ] indole 2e
Compound 2d (650.0mg, 1.4mmol) was dissolved in N, N-dimethylformamide (90mL), and palladium acetate (151.6mg, 0.7mmol, Shanghai Tantan), triphenylphosphine (354.7mg, 1.4mmol, Shanghai nationality), lithium chloride (57.4mg, 1.4mmol, Shanghai nationality) and potassium carbonate (186.8mg, 1.4mmol, Shanghai nationality). The nitrogen was replaced three times. Stirring is carried out for 16 hours at 80 ℃ under the protection of nitrogen. Water (180mL) was added, and extraction was performed with ethyl acetate (100 mL. times.3). The organic phases were combined and washed with water (50 mL. times.3). The organic phase was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 2e (400.0mg, yield: 83.9%). MS M/z (ESI) 352.0[ M +1 ].
The fifth step
4- (2- (triethylsilyl) -1, 5-dihydro-3H-pyrano [3,4,5-cd ] indol-6-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester 2f
Compound 2e (400.0mg, 1.1mmol) and compound 1i (526.6g, 1.7mmol, Shaoshima science) were dissolved in 1, 4-dioxane (10mL) and water (2 mL). Bis [ di-tert-butyl- (4-dimethylaminophenyl) phosphine ] palladium dichloride (160.8mg, 0.23mmol, Shanghai Biao) and cesium carbonate (738.0mg, 2.3mmol, Shaoshima science) were added. The nitrogen was replaced three times. The reaction was stirred at 80 ℃ for 3 hours under nitrogen. Water (20mL) was added, extraction was performed with ethyl acetate (20 mL. times.3), concentration was performed under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 2f (500.0mg, yield: 96.9%).
MS m/z(ESI):455.2[M+1]。
The sixth step
4- (2- (triethylsilyl) -1, 5-dihydro-3H-pyrano [3,4,5-cd ] indol-6-yl) piperidine-1-carboxylic acid tert-butyl ester 2g
Compound 2f (1.0g, 2.2mmol) was dissolved in methanol (60mL), 10% palladium on carbon (1.0g, water content < 1%, Shanghai Wolka) was added, and hydrogen was replaced three times. The reaction was stirred at 40 ℃ for 1 hour under hydrogen. Suction filtration is carried out, 10% palladium carbon (1.0g, water content is less than 1%, Shanghai Wolka) is added into the filtrate. The hydrogen was replaced three times. The reaction was stirred at 40 ℃ for 3 hours under hydrogen. Suction filtration and concentration of the filtrate under reduced pressure, and purification of the resulting residue by silica gel column chromatography with eluent system A gave 2g (760.0mg, yield: 75.7%) of the title product.
MS m/z(ESI):457.2[M+1]。
Seventh step
4- (2-bromo-1, 5-dihydro-3H-pyrano [3,4,5-cd ] indol-6-yl) piperidine-1-carboxylic acid tert-butyl ester 2H
Compound 2g (200.0mg, 0.44mmol) was dissolved in 1, 2-dichloroethane (20 mL). A solution of N-bromosuccinimide (78.0g, 0.44mmol, Shanghai nationality drug) in 1, 2-dichloroethane (7mL) was slowly added dropwise at-30 ℃. The reaction was stirred for 10 minutes at-30 ℃. Aqueous sodium thiosulfate (20mL) was added and extracted with dichloromethane (20 mL. times.3). The organic phase was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 2h (115.0mg, yield: 62.3%).
MS m/z(ESI):365.0[M-55]。
Eighth step
4- (2- (2, 6-dimethylpyridin-4-yl) -1, 5-dihydro-3H-pyrano [3,4,5-cd ] indol-6-yl) piperidine-1-carboxylic acid tert-butyl ester 2i
Compound 2h (420.0mg, 1.0mmol) and compound 1m (200.7mg, 1.2mmol, Shanghai Yao Ming kang) were dissolved in 1, 4-dioxane (16mL) and water (4 mL). [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (163.1mg, 0.2mmol, Beijing carbofuran) and potassium phosphate (634.0mg, 3.0mmol, Beijing carbofuran) were added. The nitrogen was replaced three times, and the reaction was stirred at 90 ℃ for 3 hours. Water (10mL) was added, extraction was performed with ethyl acetate (20 mL. times.3), concentration was performed under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 2i (380.0mg, yield: 85.2%).
MS m/z(ESI):448.3[M+1]。
The ninth step
2- (2, 6-dimethylpyridin-4-yl) -6- (piperidin-4-yl) -1, 5-dihydro-3H-pyrano [3,4,5-cd ] indole 2j
Compound 2i (340.0mg, 0.76mmol) was dissolved in methanol (5mL) and 4M hydrogen chloride/1, 4-dioxane solution (10mL) was added. Stirred at room temperature for 1 hour. The reaction mixture was concentrated under reduced pressure, and 7M ammonia/methanol solution (10mL) was added to the residue. Concentration under reduced pressure gave the title product 2j (268.0mg, yield: 100%).
MS m/z(ESI):348.0[M+1]。
The tenth step
2- (2, 6-Dimethylpyridin-4-yl) -6- (1 '-isobutyl- [1,4' -dipiperidine ] -4-yl) -1, 5-dihydro-3H-pyrano [3,4,5-cd ] indole bistrifluoroacetate 2
Compound 2j (130.0mg, 0.37mmol) and compound 1p (406.6mg, 2.6mmol) were dissolved in anhydrous N, N-dimethylformamide (7mL), and sodium acetate (521.7mg, 6.4mmol, shanghaitant) was added. Stirred at room temperature for 1 hour. Sodium borohydride acetate (555.4mg, 2.6mmol, Shaoshima science) was added. Stirred at room temperature for 16 hours. The residue was isolated and purified by high performance liquid chromatography (column: Sharpsil-T C18150 x 30mm,5 μm; mobile phase 1: water (containing 0.1% trifluoroacetic acid), mobile phase 2: acetonitrile; 30 min gradient: 5% -95%, flow rate: 30mL/min) to give the title product 2(100.0mg, yield: 44.5%).
MS m/z(ESI):487.3[M-227]。
1H NMR(500MHz,CD3OD)δ7.64(s,2H),7.34(d,1H),7.26(d,1H),5.32(s,2H),5.07(s,2H),3.80(s,2H),3.71(d,2H),3.60(s,2H),3.11-3.08(m,3H),3.00(s,2H),2.74(s,6H),2.46(s,2H),2.23-2.07(m,8H),1.07(d,6H)。
Example 3
2- (2, 6-dimethylpyridin-4-yl) -7- (1- (5,6,7, 8-tetrahydro-2, 6-naphthyridin-3-yl) piperidin-4-yl) -1,3,4, 6-tetrahydroxepin-no [5,4,3-cd ] indole tetratrifluoroacetate 3
Figure BDA0003210363510000471
First step of
5- (2, 6-dimethylpyridin-4-yl) -9- (piperidin-4-yl) -1,3,4, 6-tetrahydroxepin [5,4,3-cd ] indole hydrochloride 3a
Compound 1n (65.0mg, 0.14mmol) was dissolved in dichloromethane (2mL) and 4M hydrogen chloride/1, 4-dioxane solution (2mL) was added. Stirred at room temperature for 1 hour. The reaction solution was concentrated under reduced pressure to give the title product 3a (56.0mg, yield: 99.9%).
MS m/z(ESI):362.1[M-35]。
Second step of
7- (4- (2- (2, 6-dimethylpyridin-4-yl) -1,3,4, 6-tetrahydroxepin [5,4,3-cd ] indol-7-yl) piperidin-1-yl) -3, 4-dihydro-2, 6-naphthyridine-2 (1H) -carboxylic acid tert-butyl ester 3c
Compound 3a (56.0mg, 0.14mmol) and tert-butyl 7-chloro-3, 4-dihydro-2, 6-naphthyridine-2 (1H) -carboxylate 3b (50.0mg, 0.19mmol, Shaoshima Tech.) were dissolved in 1, 4-dioxane (4.0 mL). 1, 3-bis (2, 6-di-3-pentylphenyl) imidazol-2-ylidene ] (3-chloropyridyl) palladium (II) dichloride (12.0mg, 0.015mmol, Sigma-Aldrich) and cesium carbonate (200.0mg, 0.62mmol, Shao Yuan technology) were added. The nitrogen gas was replaced three times, and the reaction was stirred at 105 ℃ for 16 hours. The reaction solution was cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 3c (20.0mg, yield: 23.9%).
MS m/z(ESI):594.1[M+1]。
The third step
2- (2, 6-dimethylpyridin-4-yl) -7- (1- (5,6,7, 8-tetrahydro-2, 6-naphthyridin-3-yl) piperidin-4-yl) -1,3,4, 6-tetrahydroxepin-no [5,4,3-cd ] indole trifluoroacetate 3
Compound 3c (20.0mg, 0.034mmol) was dissolved in dichloromethane (3.0mL) and trifluoroacetic acid (1.0 mL). The reaction was stirred at room temperature for 1.0 hour. Concentrated under reduced pressure and the residue was isolated and purified by high performance liquid chromatography (column: Sharpsil-TC 18150 x 30mm,5 μm; mobile phase 1: water (containing 0.1% trifluoroacetic acid), mobile phase 2: acetonitrile; 15 min gradient: 10% -95%, flow rate: 30mL/min) to give the title product 3(5.0mg, yield: 6.1%). MS M/z (ESI) 494.3[ M-455 ].
1H NMR(500MHz,DMSO-d6)δ11.90(s,1H),9.26(brs,2H),8.03(s,1H),7.89(s,2H),7.33(d,1H),7.28(s,1H),7.18(s,1H),7.15(d,1H),7.08(s,1H),6.96(s,1H),5.25(s,2H),4.36(d,2H),4.28(s,2H),4.08(t,2H),3.40-3.33(m,4H),3.04(t,2H),2.96(m,1H),2.91-2.88(m,2H),2.71(s,6H),1.78-1.67(m,4H)。
Example 4
5- (2, 6-dimethylpyridin-4-yl) -9- (1-isopropylpiperidin-4-yl) -1,3,4, 6-tetrahydroxepin [5,4,3-cd ] indole 4
Figure BDA0003210363510000481
Compound 1o (105mg, 0.29mmol) was dissolved in acetonitrile (5mL), and potassium carbonate (200mg, 1.45mmol, Shanghai drug) and 2-iodopropane (200mg, 1.18mmol, adamas) were added. Stirred at 80 ℃ for 2 hours. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by high performance liquid chromatography (column: Boston Phlex Prep C185. mu.m 30 x 150 mM; mobile phase 1: water (containing 10mM ammonium bicarbonate), mobile phase 2: acetonitrile; 20 min gradient: 30% -60%, flow rate: 30mL/min) to give the title product 4(57mg, yield: 48.6%).
MS m/z(ESI):404.0[M+1]。
1H NMR(500MHz,CD3OD)δ7.38(s,2H),7.32(d,1H),7.17(d,1H),5.24(s,2H),4.11-4.08(m,2H),3.30-3.28(m,2H),3.09-3.07(m,2H),2.85(m,1H),2.68(m,1H),2.57(s,6H),2.45-2.40(m,2H),1.92-1.85(m,2H),1.80-1.78(m,2H),1.17(d,6H)。
Example 5
5'- (2, 6-dimethylpyridin-4-yl) -9' - (piperidin-4-yl) -1',6' -dihydro-3 'H-spiro [ cyclopropane-1, 4' -oxepino [5,4,3-cd ] indole ]5
Figure BDA0003210363510000491
First step of
(1- ((2, 6-Dimethylpyridin-4-yl) ethynyl) cyclopropyl) methanol 5c
(1-ethynylcyclopropyl) methanol 5a (1.0g, 10.4mmol, Nanjing Yam), 4-bromo-2, 6-dimethylpyridine 5b (1.9g, 10.2mmol, Bidayama), bis (triphenylphosphine) palladium dichloride (720mg, 1.03mmol, carbofuran), cuprous iodide (200mg, 1.05mmol, Alfa Aesar) and N, N-diisopropylethylamine (5.0g, 38.7mmol, adamas) were dissolved in N, N-dimethylformamide (25 mL). Stirring at 90 ℃ for 1.5 hours under the protection of nitrogen. Concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 5c (2.0g, yield: 97.3%).
MS m/z(ESI):202.2[M+1]。
Second step of
4- ((1- (((2-iodo-3-nitrobenzyl) oxy) methyl) cyclopropyl) ethynyl) -2, 6-lutidine 5d
Sodium hydride (820mg, 20.5mmol, 60%, Shanghai Tatan) was suspended in anhydrous tetrahydrofuran (30 mL). At 0 deg.C, compound 5c (2.1g, 10.4mmol) was added and stirred at room temperature for 30 minutes. 1- (bromomethyl) -2-iodo-3-nitrobenzene 1d (3.5g, 10.2mmol) was added. The reaction was stirred at room temperature for 16 hours. Saturated aqueous ammonia chloride (100mL), dichloromethane (80 mL. times.4) were added and the mixture was extracted, concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography using eluent system B to give the title product 5d (4.2g, yield: 88.8%).
MS m/z(ESI):463.0[M+1]。
The third step
3- (((1- ((2, 6-dimethylpyridin-4-yl) ethynyl) cyclopropyl) methoxy) methyl) -2-iodoaniline 5e
Compound 5d (4.2g, 9.1mmol) was dissolved in ethanol (75mL) and water (15mL), and iron powder (4.0g, 71.6mmol, Shanghai medicine) and ammonium chloride (4.0g, 74.1mmol, Shanghai medicine) were added. The reaction was stirred at 80 ℃ for 2.0 hours. Concentrated under reduced pressure, extracted with ethyl acetate (80 mL. times.4), and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 5e (2.5g, yield: 63.7%).
MS m/z(ESI):432.8[M+1]。
The fourth step
4-bromo-3- (((1- ((2, 6-dimethylpyridin-4-yl) ethynyl) cyclopropyl) methoxy) methyl) -2-iodoaniline 5f
Compound 5e (2.45g, 5.7mmol) was dissolved in N, N-dimethylformamide (70mL) and N-bromosuccinimide (1.05g, 5.9mmol, Shanghai nationality drug) was added. Stirred at room temperature for 1 hour. Water (50mL) was added, and extraction was performed with ethyl acetate (50 mL. times.3). The organic phase was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 5f (2.8g, yield: 96.6%).
MS m/z(ESI):510.7[M+1]。
The fifth step
9 '-bromo-5' - (2, 6-dimethylpyridin-4-yl) -1',6' -dihydro-3 'H-spiro [ cyclopropane-1, 4' -oxepino [5,4,3-cd ] indole ]5g
Compound 5f (800mg, 1.6mmol) was dissolved in N, N-dimethylformamide (80mL), and [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (130mg, 0.16mmol, shanghai tatan), lithium chloride (70mg, 1.7mmol, shanghai national drug) and potassium carbonate (650mg, 4.7mmol, shanghai test) were added. The nitrogen was replaced three times. Stirring was carried out at 105 ℃ for 16 hours under nitrogen. Concentrated under reduced pressure, and extracted with water (60mL) and ethyl acetate (50 mL. times.4). The organic phase was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to obtain 5g (530mg, yield: 88.4%) of the title product.
MS m/z(ESI):383.8[M+1]。
The sixth step
4- (5'- (2, 6-dimethylpyridin-4-yl) -1',6 '-dihydro-3' H-spiro [ cyclopropane-1, 4 '-oxepino [5,4,3-cd ] indol ] -9' -yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester 5H
Compound 5g (530.0mg, 1.4mmol) and compound 1i (600mg, 1.9mmol, Shaoyuan technology) were dissolved in 1, 4-dioxane (8.0mL) and water (2.0 mL). Bis [ di-tert-butyl- (4-dimethylaminophenyl) phosphine ] palladium dichloride (100mg, 0.14mmol, obtained from Shanghai Biao) and cesium carbonate (1.35g, 4.2mmol, Shaoshima science) were added. The nitrogen was replaced three times and the reaction was stirred at 80 ℃ for 16 hours under nitrogen protection. Water (20mL) was added, extraction was performed with ethyl acetate (20 mL. times.3), concentration was performed under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 5h (620.0mg, yield: 92.3%).
MS m/z(ESI):486.0[M+1]。
Seventh step
4- (5'- (2, 6-dimethylpyridin-4-yl) -1',6 '-dihydro-3' H-spiro [ cyclopropane-1, 4 '-oxepino [5,4,3-cd ] indol ] -9' -yl) piperidine-1-carboxylic acid tert-butyl ester 5i
Compound 5h (620.0mg, 1.3mmol) was dissolved in methanol (20 mL). 10% palladium on carbon (300.0mg, water content < 1%, Shanghai Wolka) was added. The hydrogen was replaced three times. The reaction was stirred at room temperature under hydrogen for 16 hours. Suction filtration is carried out, 10% palladium carbon (300.0mg, water content is less than 1%, Shanghai Wolka) is added into the filtrate. The hydrogen was replaced three times. The reaction was stirred at room temperature under hydrogen for 16 hours. Suction was then carried out, and the filtrate was concentrated under reduced pressure to give the crude title product 5i (460.0mg, yield: 73.9%).
MS m/z(ESI):488.0[M+1]。
Eighth step
5'- (2, 6-dimethylpyridin-4-yl) -9' - (piperidin-4-yl) -1',6' -dihydro-3 'H-spiro [ cyclopropane-1, 4' -oxepino [5,4,3-cd ] indole ]5
Crude compound 5i (50.0mg, 0.10mmol) was dissolved in dichloromethane (4.0mL) and trifluoroacetic acid (2.0 mL). The reaction was stirred at room temperature for 1.0 hour. Concentrated under reduced pressure and the residue dissolved in 7M ammonia/methanol (5.0 mL). Stir at room temperature for 10 minutes. Concentrated under reduced pressure and purified by high performance liquid chromatography (column: Boston Phlex Prep C185. mu.m 30 x 150 mM; mobile phase 1: water (containing 10mM ammonium bicarbonate), mobile phase 2: acetonitrile; 20 min gradient: 10% -95%, flow rate: 30mL/min) to give the title product 5(20mg, yield: 50.3%). MS M/z (ESI) 388.2[ M +1 ].
1H NMR(500MHz,CD3OD)δ7.29(s,2H),7.25(d,1H),7.11(d,1H),5.39(s,2H),4.09(s,2H),3.37-3.34(m,2H),3.10-2.98(m,3H),2.58(s,6H),1.95-1.88(m,4H),0.92-0.89(m,2H),0.69-0.67(m,2H)。
Example 6
2- (4- (5'- (2, 6-dimethylpyridin-4-yl) -1',6 '-dihydro-3' H-spiro [ cyclopropane-1, 4 '-oxepino [5,4,3-cd ] indol ] -9' -yl) piperidin-1-yl) acetamide 6
Figure BDA0003210363510000511
Compound 5(352.0mg, 0.78mmol) and 2-bromoacetamide 6a (161.5mg, 1.2mmol) were dissolved in N, N-dimethylformamide (5mL) and cesium carbonate (1.3g, 3.9mmol, Shanghai Shao Yuan) was added. Stirred at room temperature for 2 hours. Water (10mL) was added and extraction was performed with ethyl acetate (30 mL). The organic phase was concentrated under reduced pressure and the residue was purified by high performance liquid chromatography (column: Welch Xitinate C18,5 μm,30 mM. about.150 mM; mobile phase 1: water (containing 10mM NH)4HCO3) (ii) a Mobile phase 2: acetonitrile; gradient for 18 minutes: 30% -95%, flow rate: 30mL/min) was separated and purified to give the title product 6(165.5mg, yield: 47.7%).
MS m/z(ESI):445.2[M+1]。
1H NMR(500MHz,DMSO-d6)δ10.98(s,1H),7.22-7.13(m,5H),7.04(d,1H),5.20(s,2H),3.92(s,2H),2.92(d,2H),2.88(s,2H),2.72(t,1H),2.48(m,6H),2.22(t,2H),1.85-1.78(m,2H),1.62(d,2H),0.81(s,2H),0.54(s,2H)。
Example 7
6- (2, 6-dimethylpyridin-4-yl) -10- (piperidin-4-yl) -3,4,5, 7-tetrahydro-1H-oxa-octano [5,4,3-cd ] indole 7
Figure BDA0003210363510000521
First step of
5- (2, 6-Dimethylpyridin-4-yl) pent-4-yn-1-ol 7b
Pent-4-yn-1-ol 7a (1.0g, 11.9mmol, Pebamedicine), 4-bromo-2, 6-dimethylpyridine 5b (1.5g, 8.1mmol, Pebamedicine), bis (triphenylphosphine) palladium dichloride (565mg, 0.80mmol, carbofuran), cuprous iodide (160mg, 0.84mmol, Alfa Aesar) and N, N-diisopropylethylamine (4.0g, 30.9mmol, adamas) were dissolved in N, N-dimethylformamide (20 mL). Stirring at 90 ℃ for 1.5 hours under the protection of nitrogen. Concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title product 7B (1.35g, yield: 88.5%).
MS m/z(ESI):190.2[M+1]。
Second step of
4- (5- ((2-iodo-3-nitrobenzyl) oxy) pent-1-yn-1-yl) -2, 6-dimethylpyridine 7c
Sodium hydride (350mg, 8.75mmol, 60%, shanghaitant) was suspended in anhydrous tetrahydrofuran (20 mL). At 0 deg.C, compound 7b (900mg, 4.76mmol) was added and stirred at room temperature for 30 minutes. 1- (bromomethyl) -2-iodo-3-nitrobenzene 1d (1.5g, 4.39mmol) was added. The reaction was stirred at room temperature for 16 hours. Saturated aqueous ammonia chloride (50mL), ethyl acetate (50 mL. times.4) were added, the mixture was extracted, concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 7c (1.4g, yield: 70.9%).
MS m/z(ESI):451.0[M+1]。
The third step
3- (((5- (2, 6-dimethylpyridin-4-yl) pent-4-yn-1-yl) oxy) methyl) -2-iodoaniline 7d
Compound 7c (1.6g, 3.55mmol) was dissolved in ethanol (25mL) and water (5mL), and iron powder (1.5g, 26.9mmol, Shanghai medicine) and ammonium chloride (1.5g, 27.8mmol, Shanghai medicine) were added. The reaction was stirred at 80 ℃ for 2.0 hours. Concentrated under reduced pressure, extracted with ethyl acetate (50 mL. times.4), and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 7d (1.2g, yield: 80.4%).
MS m/z(ESI):421.1[M+1]。
The fourth step
4-bromo-3- (((5- (2, 6-dimethylpyridin-4-yl) pent-4-yn-1-yl) oxy) methyl) -2-iodoaniline 7e
Compound 7d (1.2g, 2.86mmol) was dissolved in N, N-dimethylformamide (15mL) and N-bromosuccinimide (510mg, 5.87mmol, Shanghai nationality medicine) was added. Stirred at room temperature for 1 hour. Water (50mL) was added, and extraction was performed with ethyl acetate (50 mL. times.3). The organic phase was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 7e (1.7g, yield: 98.2%).
MS m/z(ESI):499.0[M+1]。
The fifth step
10-bromo-6- (2, 6-dimethylpyridin-4-yl) -3,4,5, 7-tetrahydro-1H-oxa-octa [5,4,3-cd ] indole 7f
Compound 7e (500mg, 1.0mmol) was dissolved in N, N-dimethylformamide (80mL), and [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (80mg, 0.098mmol, shanghai taitan), lithium chloride (50mg, 1.18mmol, shanghai national drug) and potassium carbonate (415mg, 3.0mmol, shanghai Hu test) were added. The nitrogen was replaced three times. Stirring was carried out at 105 ℃ for 16 hours under nitrogen. Concentrated under reduced pressure, and extracted with water (20mL) and ethyl acetate (20 mL. times.4). The organic phase was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title product 7f (130mg, yield: 35%).
MS m/z(ESI):373.1[M+1]。
The sixth step
4- (6- (2, 6-dimethylpyridin-4-yl) -3,4,5, 7-tetrahydro-1H-oxaocta-lo [5,4,3-cd ] indol-10-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester 7g
Compound 7f (130.0mg, 0.35mmol) and compound 1i (150mg, 0.49mmol, Shaoyou Tech) were dissolved in 1, 4-dioxane (4.0mL) and water (1.0 mL). Bis [ di-tert-butyl- (4-dimethylaminophenyl) phosphine ] palladium dichloride (30mg, 0.042mmol, obtained from Shanghai) and cesium carbonate (345mg, 1.06mmol, Shaoshima science) were added. The nitrogen was replaced three times and the reaction was stirred at 80 ℃ for 16 hours under nitrogen protection. Water (10mL) was added, extraction was performed with ethyl acetate (15 mL. times.3), concentration was performed under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system A to obtain 7g of the title product (150.0mg, yield: 90.5%).
MS m/z(ESI):474.3[M+1]。
Seventh step
4- (6- (2, 6-dimethylpyridin-4-yl) -3,4,5, 7-tetrahydro-1H-oxaocta-cyclo [5,4,3-cd ] indol-10-yl) piperidine-1-carboxylic acid tert-butyl ester 7H
Compound 7g (250.0mg, 0.5mmol) was dissolved in methanol (20 mL). 10% palladium on carbon (200.0mg, water content < 1%, Shanghai Wolka) was added. The hydrogen was replaced three times. The reaction was stirred at room temperature under hydrogen for 16 hours. Suction filtration is carried out, 10% palladium carbon (200.0mg, water content is less than 1%, Shanghai Wolka) is added into the filtrate. The hydrogen was replaced three times. The reaction was stirred at room temperature under hydrogen for 16 hours. Suction filtration and concentration of the filtrate under reduced pressure and purification of the resulting residue by silica gel column chromatography with eluent system A gave the title product 7h (150.0mg, yield: 59.7%).
MS m/z(ESI):476.0[M+1]。
Eighth step
6- (2, 6-dimethylpyridin-4-yl) -10- (piperidin-4-yl) -3,4,5, 7-tetrahydro-1H-oxa-octano [5,4,3-cd ] indole 7
Compound 7h (30.0mg, 0.063mmol) was dissolved in dichloromethane (3.0mL) and trifluoroacetic acid (1.0 mL). The reaction was stirred at room temperature for 1.0 hour. Concentrated under reduced pressure and the residue dissolved in 7M ammonia/methanol (5.0 mL). Stir at room temperature for 10 minutes. Concentrated under reduced pressure and purified by high performance liquid chromatography (column: Boston Phlex Prep C185. mu.m 30 x 150 mM; mobile phase 1: water (containing 10mM ammonium bicarbonate), mobile phase 2: acetonitrile; 20 min gradient: 10% -95%, flow rate: 30mL/min) to give the title product 7(12mg, yield: 50.7%). MS M/z (ESI) 376.0[ M +1 ].
1H NMR(500MHz,CD3OD)δ7.37(d,1H),7.23(s,2H),7.19(d,1H),5.23(s,2H),3.83-3.81(m,2H),3.33-3.19(m,5H),2.85(m,2H),2.58(s,6H),2.00-1.96(m,2H),1.84-1.80(m,4H)。
Example 8
2- (4- (5- (7, 8-dimethyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) -1,3,4, 6-tetrahydroxepin-o [5,4,3-cd ] indol-9-yl) piperidin-1-yl) acetamide 8
Figure BDA0003210363510000551
First step of
7, 8-dimethyl-6- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) - [1,2,4] triazolo [1,5-a ] pyridine 8b
The compound 6-bromo-7, 8-dimethyl- [1,2,4] triazolo [1,5-a ] pyridine 8a (500.0mg, 2.2mmol, prepared using the reference "preparation of intermediate F-4 on page 140 of WO2018005586a 1") and pinacol diboride diborate (842.5mg, 3.3mmol, shanghai shao) were dissolved in 1, 4-dioxane (10 mL). [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (161.7mg, 0.22mmol, Shanghai Tantan) and potassium acetate (650.3mg, 6.6mmol, Shanghai Hu test) were added. The nitrogen was replaced three times, and the reaction was stirred at 80 ℃ for 16 hours. Water (20mL) was added, extraction was performed with ethyl acetate (20 mL. times.3), and the organic phase was concentrated under reduced pressure to give crude compound 8b (1.3 g). The product was used in the next step without purification.
MS m/z(ESI):274.1[M+1]。
Second step of
4- (2- (7, 8-dimethyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) -1,3,4, 6-tetrahydroxepin-o [5,4,3-cd ] indol-7-yl) piperidine-1-carboxylic acid tert-butyl ester 8c
The crude compound 1l (300.0mg, 0.7mmol) and compound 8b (613.8mg, 1.0mmol, 46% purity) were dissolved in 1, 4-dioxane (10mL) and water (2 mL). Dichlorobis [ di-tert-butyl- (4-dimethylaminophenyl) phosphine ] palladium (97.6mg, 0.14mmol, Shanghai Biao) and potassium phosphate (438.3mg, 2.1mmol, Beijing carbofuran) were added. The nitrogen gas was replaced three times, and the reaction was stirred at 90 ℃ for 12 hours. Water (10mL) was added, extraction was performed with ethyl acetate (20 mL. times.3), concentration was performed under reduced pressure, and the residue was purified by silica gel column chromatography with eluent system A to give the title product 8c (320.0mg, yield: 92.6%).
MS m/z(ESI):502.1[M+1]。
The third step
2- (7, 8-dimethyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) -7- (piperidin-4-yl) -1,3,4, 6-tetrahydroxepin-o [5,4,3-cd ] indole hydrochloride salt 8d
Compound 8c (320.0mg, 0.64mmol) was dissolved in methanol (2mL) and 4M hydrogen chloride/1, 4-dioxane solution (3mL) was added. Stirred at room temperature for 2 hours. The reaction solution was concentrated under reduced pressure to give the title product 8d (280.0mg, yield: 100%).
MS m/z(ESI):402.1[M+1]。
The fourth step
2- (4- (5- (7, 8-dimethyl- [1,2,4] triazolo [1,5-a ] pyridin-6-yl) -1,3,4, 6-tetrahydroxepin-o [5,4,3-cd ] indol-9-yl) piperidin-1-yl) acetamide 8
Compound 8d (150.0mg, 0.34mmol) and 6a (70.9mg, 0.5mmol) were dissolved in N, N-dimethylformamide (5mL) and cesium carbonate (556.6g, 1.7mmol, Shanghai Shao Yuan) was added. Stirred at room temperature for 2 hours. Water (20mL) was added for dilution, and the mixture was filtered under reduced pressure. The obtained solid was purified by high performance liquid chromatography (column: Welch Xtimate C18,5 μm,30 mM. about.150 mM; mobile phase 1: water (containing 10mM NH)4HCO3) (ii) a Mobile phase 2: acetonitrile; gradient for 23 min: 20% -95%, flow rate: 30mL/min) was separated and purified to give the title product 8(37.8mg, yield: 24.1%).
MS m/z(ESI):459.1[M+1]。
1H NMR(500MHz,DMSO-d6)δ11.17(s,1H),8.82(s,1H),8.46(s,1H),7.24(d,2H),7.14(s,1H),7.10(d,1H),5.16(s,2H),3.96(t,2H),2.92(d,2H),2.88(s,2H),2.83(s,2H),2.63(d,1H),2.58(s,3H),2.22(d,2H),2.19(s,3H),1.86-1.79(m,2H),1.65(d,2H)。
Biological evaluation
The present disclosure is further described and explained below in conjunction with test examples, but these examples are not meant to limit the scope of the present disclosure.
Test example 1: inhibition of human TLR7 activation pathway by disclosed compounds
First, experimental material and instrument
1.HEK-BlueTMhTLR7 cell (Invivogen)
2. Racemote (R848/Resiquimod, Invivogen)
3. Alkaline phosphatase Detection Medium (Quanti-Blue Detection, Invivogen)
4. Blasticidin (Invivogen)
5. Bleomycin (Zeocin, Invivogen)
6. Neomycin (Normocin, Invivogen)
DMEM HIGH sugar medium (DMEM/HIGH Glucose, GE Healthcare)
8. Fetal bovine serum (FBS, Gibco)
9. Phosphate buffer (Shanghai Yuan culture Biotech Co., Ltd.)
10. Sterile pure water (homemade Hengrui Shanghai)
11.15ml centrifuge tube (Corning)
12.96 hole dispensing plate (Corning)
13.96 well Flat bottom cell culture plate (Corning)
14. Constant temperature cell culture box (Thermo scientific)
15. Constant temperature box (Shanghai-Heng scientific instruments Co., Ltd.)
PHERAstar FS microplate reader (BMG Labtech)
Second, the experimental procedure
HEK-Blue was purchased from InvivogenTMhTLR7 cell, which is characterized by that it utilizes human Toll-like receptor 7(TLR7) gene and secretory alkaline phosphatase reporter gene (SEAP) which is placed under the control of IFN-beta minimal promoter containing 5 NF-kB and AP-1 binding sites to cotransfect HEK293 cell, when the TLR7 is activated by agonist, the SEAP secretion can be induced by downstream NF-kB and AP-1, after the antagonist compound is added, the above-mentioned pathway can be inhibited, the SEAP secretion is reduced, and the OD620 can be measured by SEAP substrate so as to evaluate the activity of said compound on TLR7 pathway.
20mM test compound in 100% DMSO was serially diluted with 100% DMSO to 2000, 400, 80, 16, 3.2, 0.64, 0.128, 0.0256. mu.M in blank wells with 100% DMSO, and further diluted 20-fold in DMEM high-sugar medium (complete medium, the same below) containing 10% inactivated FBS. R848 was diluted to 10. mu.M with sterile water. Add 20. mu.L/well 10. mu.MR 848 diluted in sterile water to 96-well cell culture plates, and add the above compound diluted in complete medium and 100% DMSO at 20. mu.L per well to the well containing R848; negative control wells were filled with 20. mu.L of sterile water and 20. mu.L of 100% DMSO diluted in complete medium.
HEK-BlueTMhTLR7 cells were cultured in DMEM high-glucose medium containing 10% inactivated FBS, 100. mu.g/mL neomycin, 10. mu.g/mL blasticidin and 100. mu.g/mL bleomycin. Collecting cells which grow well and grow to 70% -80%, discarding the growth medium, adding 5-10mL of 37 deg.C preheated PBS to wash the cells once, adding 2-5mL of preheated PBS to placeCulturing at 37 deg.C for 1-2 min, blowing off cells with a pipette, transferring cells to a 15mL centrifuge tube, counting cells, adjusting cell density to 4.8 × 10 with complete medium5and/mL. 160 μ L of the cell suspension after density adjustment was added to the above 96 well cell culture plate to give 76500 cells/well per well, R848 was 1 μ M at final concentration, and the test compounds were 10000, 2000, 400, 80, 16, 3.2, 0.64, and 0.128nM, respectively. The cells were incubated at 37 ℃ with 5% CO2Culturing for 20 hours in an incubator, then taking 20 mu L of supernatant, adding 180 mu L of prepared Quanti-Blue, incubating for 120 minutes in a constant temperature box at 37 ℃ in the dark, and reading OD620 light absorption value by a microplate reader. The inhibition rate was calculated using the following formula: the inhibition rate is {1- (OD test compound-OD negative control well)/(OD blank well-OD negative control well) } × 100%, an inhibition curve is drawn by Graphpad Prism software according to each concentration of the compound and the corresponding inhibition rate, and the concentration of the compound at which the inhibition rate reaches 50%, i.e., IC, is calculated50The values are shown in Table 1.
Table 1 IC of compounds of the present disclosure by measurement of human TLR7 pathway50Value of
Example numbering IC50(nM)
1 37.8
2 124.2
3 6.0
4 7.3
5 72.3
6 111.1
7 2.5
8 1.5
And (4) conclusion: the disclosed compounds have inhibitory effects on the TLR7 pathway.
Test example 2: inhibition of human TLR8 activation pathway by disclosed compounds
First, experimental material and instrument
1.HEK-BlueTMhTLR8 cell (Invivogen)
2. Racemote (R848/Resiquimod, Invivogen)
3. Alkaline phosphatase Detection Medium (Quanti-Blue Detection, Invivogen)
4. Blasticidin (Invivogen)
5. Bleomycin (Zeocin, Invivogen)
6. Neomycin (Normocin, Invivogen)
DMEM HIGH sugar medium (DMEM/HIGH Glucose, GE Healthcare)
8. Fetal bovine serum (FBS, Gibco)
9. Phosphate buffer (Shanghai Yuan culture Biotech Co., Ltd.)
10. Sterile pure water (homemade Hengrui Shanghai)
11.15ml centrifuge tube (Corning)
12.96 hole dispensing plate (Corning)
13.96 well Flat bottom cell culture plate (Corning)
14. Constant temperature cell culture box (Thermo scientific)
15. Constant temperature box (Shanghai-Heng scientific instruments Co., Ltd.)
PHERAstar FS microplate reader (BMG Labtech)
Second, the experimental procedure
HEK-Blue was purchased from InvivogenTMhTLR8 cell, which is characterized by that it utilizes human Toll-like receptor 8(TLR8) gene and secretory alkaline phosphatase reporter gene (SEAP) which is placed under the control of IFN-beta minimal promoter containing 5 NF-kB and AP-1 binding sites to cotransfect HEK293 cell, when the TLR8 is activated by agonist, the SEAP secretion can be induced by downstream NF-kB and AP-1, after the antagonist compound is added, the above-mentioned pathway can be inhibited, the SEAP secretion is reduced, and the OD620 can be measured by SEAP substrate so as to evaluate the activity of said compound on TLR8 pathway.
20mM test compound in 100% DMSO was serially diluted with 100% DMSO to 2000, 400, 80, 16, 3.2, 0.64, 0.128, 0.0256. mu.M in blank wells with 100% DMSO, and further diluted 20-fold in DMEM high-sugar medium (complete medium, the same below) containing 10% inactivated FBS. R848 was diluted to 60 μ M with sterile water. mu.L/well of 60. mu. M R848 diluted in sterile water was added to a 96 well cell culture plate, and the above compound diluted in complete medium and 100% DMSO were added to wells containing R848 at 20. mu.L per well. Negative control wells were filled with 20. mu.L of sterile water and 20. mu.L of 100% DMSO diluted in complete medium.
HEK-BlueTMhTLR8 cells were cultured in DMEM high-glucose medium containing 10% inactivated FBS, 100. mu.g/mL neomycin, 10. mu.g/mL blasticidin and 100. mu.g/mL bleomycin. Collecting cells which grow well and grow to 70% -80%, discarding the growth medium, adding 5-10mL of PBS preheated at 37 deg.C for washing cells once, adding 2-5mL of preheated PBS for culturing at 37 deg.C for 1-2 min, blowing off cells with a pipette, transferring cells to a 15mL centrifuge tube, counting cells, adjusting cell density to 4.8 × 10 with the whole medium5and/mL. 160. mu.L of the cell suspension adjusted to the density was added to the above 96-well cell culture plate to give 76500 cells/well and 6. mu.M R848 as a final concentration, and 10000, 2000, 400, 80, 16, 3.2, 0.64, and test compound as final concentrations,0.128 nM. The cells were incubated at 37 ℃ with 5% CO2Culturing for 20 hours in an incubator, then taking 20 mu L of supernatant, adding 180 mu L of prepared Quanti-Blue, incubating for 120 minutes in a constant temperature box at 37 ℃ in the dark, and reading OD620 light absorption value by a microplate reader. The inhibition rate was calculated using the following formula: the inhibition rate is {1- (OD test compound-OD negative control well)/(OD blank well-OD negative control well) } × 100%, an inhibition curve is drawn by Graphpad Prism software according to each concentration of the compound and the corresponding inhibition rate, and the concentration of the compound at which the inhibition rate reaches 50%, i.e., IC, is calculated50The values are shown in Table 2.
Table 2 IC of compounds of the present disclosure by measurement of human TLR8 pathway50Value of
Example numbering IC50(nM)
1 26.5
2 238.9
3 44.3
4 26.3
5 50.7
6 73.3
7 3.7
8 1.5
And (4) conclusion: the disclosed compounds have inhibitory effects on the TLR8 pathway.
Test example 3: inhibition of human TLR9 activation pathway by disclosed compounds
First, experimental material and instrument
1.HEK-BlueTMhTLR9 cell (Invivogen)
2.CpG ODN2006(Invivogen)
3. Alkaline phosphatase Detection Medium (Quanti-Blue Detection, Invivogen)
4. Blasticidin (Invivogen)
5. Bleomycin (Zeocin, Invivogen)
6. Neomycin (Normocin, Invivogen)
DMEM HIGH sugar medium (DMEM/HIGH Glucose, GE Healthcare)
8. Fetal bovine serum (FBS, Gibco)
9. Phosphate buffer (Shanghai Yuan culture Biotech Co., Ltd.)
10. Sterile pure water (homemade Hengrui Shanghai)
11.15ml centrifuge tube (Corning)
12.96 hole dispensing plate (Corning)
13.96 well Flat bottom cell culture plate (Corning)
14. Constant temperature cell culture box (Thermo scientific)
15. Constant temperature box (Shanghai-Heng scientific instruments Co., Ltd.)
PHERAstar FS microplate reader (BMG Labtech)
Second, the experimental procedure
HEK-Blue was purchased from InvivogenTMhTLR9 cell, which is formed by human Toll-like receptor 9(TLR9) gene and secretory alkaline phosphatase-containing reportGenes (SEAP) were co-transfected into HEK293 cells and alkaline phosphatase reporter genes (SEAP) under the control of an IFN- β minimal promoter containing 5 NF-kB and AP-1 binding sites, SEAP secretion was induced by downstream NF-kB and AP-1 when TLR9 was activated with agonist, the pathway was inhibited by addition of antagonist compounds, SEAP secretion was decreased, and OD620 was measured by SEAP substrate to assess the activity of compounds on the TLR9 pathway.
20mM test compound in 100% DMSO was serially diluted with 100% DMSO to 2000, 400, 80, 16, 3.2, 0.64, 0.128, 0.0256. mu.M in blank wells with 100% DMSO, and further diluted 20-fold in DMEM high-sugar medium (complete medium, the same below) containing 10% inactivated FBS. ODN2006 was diluted to 10 μ M with sterile water. To a 96-well cell culture plate, 20 μ L/well of 10 μ MODN2006 diluted in sterile water was added, and the above-described compound diluted in complete medium and 100% DMSO were added to wells containing ODN2006 at 20 μ L per well. Negative control wells were filled with 20. mu.L of sterile water and 20. mu.L of 100% DMSO diluted in complete medium.
HEK-BlueTMhTLR9 cells were cultured in DMEM/high glucose medium containing 10% FBS, 100. mu.g/mL neomycin, 10. mu.g/mL blasticidin and 100. mu.g/mL bleomycin. Collecting cells which grow well and grow to 70% -80%, discarding the growth medium, adding 5-10mL of PBS preheated at 37 deg.C for washing cells once, adding 2-5mL of preheated PBS for culturing at 37 deg.C for 1-2 min, blowing off cells with a pipette, transferring cells to a 15mL centrifuge tube, counting cells, adjusting cell density to 4.8 × 10 with the whole medium5and/mL. 160. mu.L of the cell suspension after density adjustment was added to the above 96-well cell culture plate, the final cell count per well was 76500/well, the final ODN2006 concentration was 1. mu.M, and the final test compound concentrations were 10000, 2000, 400, 80, 16, 3.2, 0.64, and 0.128nM, respectively. The cells were incubated at 37 ℃ with 5% CO2Culturing for 20 hours in an incubator, then taking 20 mu L of supernatant, adding 180 mu L of prepared Quanti-Blue, incubating for 15 minutes in a constant temperature box at 37 ℃ in the dark, and reading OD620 light absorption value by a microplate reader. The inhibition rate was calculated using the following formula: inhibition rate {1- (OD test compound-OD negative control well)/(OD blank well-OD negative control well) } × 100%, according to each compound, using Graphpad Prism softwareThe concentration and the corresponding inhibition rate are plotted to obtain an inhibition curve, and the concentration of the compound when the inhibition rate reaches 50 percent, namely IC is calculated50The values are shown in Table 3.
Table 3 IC of compounds of the present disclosure by measurement of human TLR9 pathway50Value of
Example numbering IC50(nM)
1 64.3
2 34.6
3 175.4
4 135.1
And (4) conclusion: the disclosed compounds have inhibitory effects on the TLR9 pathway.

Claims (29)

1. A compound of formula (I), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure FDA0003210363500000011
wherein:
x is selected from O atom, S atom and NH;
L1is selected from CH2、CR4aR4bAnd C (O);
L2selected from the group consisting of CR4cR4dC (O), O atom, S atom and NH;
g is CR2aOr an N atom;
ring a is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
each R is1Are the same or different and are each independently selected from the group consisting of a hydrogen atom, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, - (CH)2)rNR6aR6bNitro, hydroxy, hydroxyalkyl, -C (O) NR6aR6b、-C(O)R7、-NR6cC(O)R7、-C(O)OR8Cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R2aand R2The same or different, and each is independently selected from the group consisting of hydrogen atom, halogen, alkyl group, alkoxy group, haloalkyl group, haloalkoxy group, cyano group, amino group, nitro group, hydroxyl group and hydroxyalkyl group;
ring B is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
each R is3The same or different, and each is independently selected from the group consisting of hydrogen atom, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, oxo, cyano, amino, nitro, hydroxy, and hydroxyalkyl;
R0selected from the group consisting of hydrogen atom, deuterium atom, halogen, alkyl, alkenyl, alkynyl, heteroalkyl, alkoxy, haloalkyl, deuterated alkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, - (CH)2)rC(O)NR6aR6bAnd
Figure FDA0003210363500000012
L3selected from the group consisting of a bond, alkylene, and heteroalkylene, wherein said alkylene and heteroalkylene are each independently optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
ring C is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
each R is5Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, alkyl, heteroalkyl, alkenyl, alkynyl, alkoxy, oxo, haloalkyl, haloalkoxy, cyano, amino, - (CH)2)rNR6aR6bNitro, hydroxy, hydroxyalkyl, -C (O) NR6aR6b、-C(O)OR8、-S(O)mR9、-S(O)mNR6aR6bCycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, heteroalkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R4aand R4bThe same or different and each is independently selected from the group consisting of hydrogen atom, alkyl group, halogen, alkoxy group, haloalkyl group, haloalkoxy group, cyano group, hydroxyl group and hydroxyalkyl group;
R4cand R4dThe same or different and each is independently selected from the group consisting of hydrogen atom, alkyl group, halogen, alkoxy group, haloalkyl group, haloalkoxy group, cyano group, hydroxyl group and hydroxyalkyl group; or R4cAnd R4dTogether with the carbon atom to which they are attached form a cycloalkyl or heterocyclyl group;
R6aand R6bAre the same or different and are each independently selected from a hydrogen atom,Alkyl, alkenyl, alkynyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, aryl, and heteroaryl; or R6aAnd R6bTogether with the nitrogen atom to which they are attached form a heterocyclyl group, which heterocyclyl group is optionally substituted by one or more substituents selected from the group consisting of halogen, alkyl, oxo, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R6cselected from the group consisting of hydrogen atoms, alkyl groups, alkenyl groups, alkynyl groups, haloalkyl groups, hydroxyalkyl groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups;
R7selected from the group consisting of hydrogen atoms, alkyl groups, haloalkyl groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups; wherein said alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are each independently optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy and hydroxyalkyl;
R8selected from the group consisting of hydrogen atoms, alkyl groups, alkenyl groups, alkynyl groups, haloalkyl groups, hydroxyalkyl groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups;
R9selected from the group consisting of hydrogen atoms, alkyl groups, alkenyl groups, alkynyl groups, haloalkyl groups, hydroxyalkyl groups, amino groups, hydroxyl groups, cycloalkyl groups, heterocyclic groups, aryl groups, and heteroaryl groups;
u is 0, 1,2 or 3;
v is 0, 1,2 or 3;
n is 0 or 1;
p is 0, 1,2, 3,4,5 or 6;
r is 0, 1,2, 3,4,5 or 6;
m is 0, 1 or 2;
s is 0, 1,2, 3,4,5 or 6; and is
t is 0, 1,2, 3,4,5 or 6.
2. The compound of formula (I) according to claim 1, or a tautomer, racemate, enantiomer, or non-enantiomer thereofEnantiomers, or mixtures thereof, or pharmaceutically acceptable salts thereof, wherein L2Selected from the group consisting of C (O), O atoms, S atoms, and NH; and/or R0Selected from the group consisting of hydrogen atom, deuterium atom, halogen, alkyl, alkenyl, alkynyl, heteroalkyl, alkoxy, haloalkyl, deuterated alkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl and
Figure FDA0003210363500000031
ring C, L3、R5And t is as defined in claim 1.
3. The compound of the general formula (I) according to claim 1 or2, wherein X is an O atom, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof.
4. A compound of general formula (I) according to any one of claims 1 to 3, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein L1Is CH2
5. A compound of general formula (I) according to any one of claims 1,3 and 4, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is0Selected from hydrogen atoms, C1-6Alkyl, - (CH)2)rC(O)NR6aR6bAnd
Figure FDA0003210363500000032
ring C, L3、R5、R6a、R6bR and t are as defined in claim 1; preferably, R0Is composed of
Figure FDA0003210363500000033
Ring C, L3、R5And t is as defined in claim 1.
6. The compound of general formula (I), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 5, which is a compound of general formula (II), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure FDA0003210363500000041
wherein:
ring A, ring B, ring C, G, L2、L3、R1To R3、R5P, s, t, n, u and v are as defined in claim 1.
7. The compound of general formula (I) according to any one of claims 1 to 6, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein n is 0.
8. A compound of general formula (I) according to any one of claims 1 to 7, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring a is selected from the group consisting of 3-to 8-membered cycloalkyl, 3-to 12-membered heterocyclyl, 6-to 10-membered aryl, and 5-to 10-membered heteroaryl; preferably, ring a is 5 to 10 membered heteroaryl; more preferably, ring A is pyridyl or
Figure FDA0003210363500000042
Most preferably, ring a is pyridyl.
9. A compound of general formula (I) according to any one of claims 1 to 8, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring B is selected from the group consisting of 3-to 8-membered cycloalkyl, 3-to 12-membered heterocyclyl, 6-to 10-membered aryl, and 5-to 10-membered heteroaryl; preferably, ring B is a 3 to 12 membered heterocyclic group containing at least one nitrogen atom; more preferably, ring B is piperidinyl.
10. The compound of general formula (I), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 9, which is a compound of general formula (III), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure FDA0003210363500000051
wherein:
ring B is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
ring C, R1To R3、R5、R2a、L3S, t and v are as defined in claim 1.
11. A compound of general formula (I) according to any one of claims 1 to 10, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring C is selected from the group consisting of 3-to 8-membered cycloalkyl, 3-to 12-membered heterocyclyl, 6-to 10-membered aryl, and 5-to 10-membered heteroaryl; preferably, ring C is a 3 to 12 membered heterocyclic group containing at least one nitrogen atom or a 5 to 10 membered heteroaryl group containing at least one nitrogen atom; more preferably, ring C is piperidinyl or 5,6,7, 8-tetrahydro-2, 6-naphthyridin-3-yl.
12. The compound of general formula (I) according to any one of claims 1 to 11, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R2Is a hydrogen atom.
13. The compound of general formula (I), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 12, which is a compound of general formula (IV), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure FDA0003210363500000052
wherein:
ring C is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom or
Figure FDA0003210363500000061
Ring D is a 4-to 6-membered heterocyclic group containing at least one nitrogen atom;
Z1、Z2、Z3and Z4One is a carbon atom and the remaining three are the same or different and are each independently CR5Or an N atom;
R10selected from the group consisting of hydrogen atoms, alkyl groups, heteroalkyl groups, haloalkyl groups, and hydroxyalkyl groups;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
t is 1,2, 3,4,5 or 6;
R1、R3、R5、L3s and v are as defined in claim 1.
14. The compound of general formula (I) according to any one of claims 1 to 13, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R1Is a hydrogen atom or C1-6An alkyl group.
15. The compound of general formula (I) according to any one of claims 1 to 14, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R3Is a hydrogen atom.
16. A compound of general formula (I) according to any one of claims 1 to 15, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein L3Is a bond.
17. The compound of general formula (I) according to any one of claims 1 to 16, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R5Is a hydrogen atom or C1-6An alkyl group.
18. A compound of general formula (I), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, according to any one of claims 1 to 17, selected from the group consisting of:
Figure FDA0003210363500000062
Figure FDA0003210363500000071
Figure FDA0003210363500000081
19. a compound of formula (IIIa), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure FDA0003210363500000082
wherein:
ring B is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
R1to R3、R2aS and v are as defined in claim 10.
20. The compound of formula (IIIa) according to claim 19, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, selected from the group consisting of:
Figure FDA0003210363500000083
Figure FDA0003210363500000091
21. a compound of formula (IIIaa), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure FDA0003210363500000092
wherein:
RWis an amino protecting group; preferably, RWIs tert-butyloxycarbonyl;
ring B is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
R1、R2、R2a、R3s and v are as defined in claim 10.
22. A compound, or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, selected from the group consisting of:
Figure FDA0003210363500000093
Figure FDA0003210363500000101
23. a process for preparing a compound of formula (III), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, comprising:
Figure FDA0003210363500000102
method 1
Subjecting a compound of the general formula (IIIa) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, to a reductive amination reaction with a compound of the general formula (V) to obtain a compound of the general formula (III) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
or
Method 2
Carrying out coupling reaction on the compound of the general formula (IIIa) or a tautomer, a racemate, an enantiomer, a diastereomer or a mixture form thereof or a pharmaceutically acceptable salt thereof and the compound of the general formula (V') to obtain the compound of the general formula (III) or the tautomer, the racemate, the enantiomer, the diastereomer or a mixture form thereof or a pharmaceutically acceptable salt thereof;
wherein:
L3is a bond;
ring B is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom;
y is halogen; preferably, Y is chloro;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
ring C, R1To R3、R5、R2aS, t and v are as defined in claim 10.
24. A process for preparing a compound of formula (IIIa), or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, comprising:
Figure FDA0003210363500000111
a compound of formula (IIIaa) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereofWith salts, removing protecting groups RWTo obtain a compound of formula (IIIa) or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
wherein:
RWis an amino protecting group; preferably, RWIs tert-butyloxycarbonyl;
ring B is a 3-to 12-membered heterocyclic group containing at least one nitrogen atom;
W1、W2、W3、W4and W5Are the same or different and are each independently CR1Or an N atom;
R1、R2、R2a、R3s and v are as defined in claim 10.
25. A pharmaceutical composition comprising a compound of formula (I) according to any one of claims 1 to 18, or a tautomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, or excipients.
26. Use of a compound of general formula (I) according to any one of claims 1 to 18 or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 25, for the preparation of a medicament for inhibiting TLR7, TLR8, and TLR 9.
27. Use of a compound of general formula (I) according to any one of claims 1 to 18 or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 25, for the manufacture of a medicament for inhibiting TLR7, TLR8, or TLR 9; preferably for the manufacture of a medicament for inhibiting TLR7 and TLR8, or for the manufacture of a medicament for inhibiting TLR7 and TLR 9.
28. Use of a compound of general formula (I) according to any one of claims 1 to 18 or a tautomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 25, for the preparation of a medicament for the treatment and/or prevention of inflammatory or autoimmune diseases.
29. The use according to claim 28, wherein the inflammatory or autoimmune disease is selected from the group consisting of Systemic Lupus Erythematosus (SLE), rheumatoid arthritis, Multiple Sclerosis (MS) and schungren's syndrome.
CN202110928898.9A 2020-08-14 2021-08-13 Fused tricyclic derivatives, preparation method thereof and application thereof in medicines Active CN114075212B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010819202 2020-08-14
CN2020108192024 2020-08-14

Publications (2)

Publication Number Publication Date
CN114075212A true CN114075212A (en) 2022-02-22
CN114075212B CN114075212B (en) 2023-05-12

Family

ID=80283232

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110928898.9A Active CN114075212B (en) 2020-08-14 2021-08-13 Fused tricyclic derivatives, preparation method thereof and application thereof in medicines

Country Status (1)

Country Link
CN (1) CN114075212B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11661431B2 (en) 2021-04-16 2023-05-30 Gilead Sciences, Inc. Thienopyrrole compounds

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030236295A1 (en) * 2000-09-22 2003-12-25 Yoshikazu Fukui Tricylic indole compounds having affinity for serotonin receptor
WO2016194806A1 (en) * 2015-05-29 2016-12-08 塩野義製薬株式会社 Nitrogen-containing tricyclic derivative having hiv replication inhibitory activity
CN108794486A (en) * 2017-05-05 2018-11-13 江苏恒瑞医药股份有限公司 Condensed ring radical ketones derivant, preparation method and its application in medicine
CN109661394A (en) * 2016-12-19 2019-04-19 上海和誉生物医药科技有限公司 FGFR4 inhibitor, preparation method and application pharmaceutically

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030236295A1 (en) * 2000-09-22 2003-12-25 Yoshikazu Fukui Tricylic indole compounds having affinity for serotonin receptor
WO2016194806A1 (en) * 2015-05-29 2016-12-08 塩野義製薬株式会社 Nitrogen-containing tricyclic derivative having hiv replication inhibitory activity
CN109661394A (en) * 2016-12-19 2019-04-19 上海和誉生物医药科技有限公司 FGFR4 inhibitor, preparation method and application pharmaceutically
CN108794486A (en) * 2017-05-05 2018-11-13 江苏恒瑞医药股份有限公司 Condensed ring radical ketones derivant, preparation method and its application in medicine

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
PENGYU TAO ET AL.: "Rhodium-catalyzed intramolecular annulation via C–H activation leading to fused tricyclic indole scaffolds" *
QING DONG ET AL.: "Reductive Cleavage of TROC Groups Under Neutral Conditions with Cadmium-Lead Couple", 《TETRAHEDRON LETTERS》 *
YAN GAO 等: "Intramolecular Larock indole synthesis for the preparation of tricyclic indoles and its application in the synthesis of tetrahydropyrroloquinoline and fargesine" *
张晓敏 等: "PD-1/PD-L1受体抑制剂与肾癌免疫治疗的研究进展", 《临床肿瘤学杂志》 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11661431B2 (en) 2021-04-16 2023-05-30 Gilead Sciences, Inc. Thienopyrrole compounds

Also Published As

Publication number Publication date
CN114075212B (en) 2023-05-12

Similar Documents

Publication Publication Date Title
CN110177793B (en) Heteroaryl pyrazole derivative, preparation method and application thereof in medicine
CN107556244B (en) Fused ring compound, pharmaceutical composition and application thereof
KR20170129810A (en) Substituted tricyclic heterocyclic compounds
CN111094289A (en) Pyridopyrimidine derivative, preparation method thereof and application thereof in medicine
WO2022247816A1 (en) Nitrogen-containing heterocyclic compound, preparation method therefor, and application thereof in medicines
WO2022017365A1 (en) Sulfur-containing isoindoline derivative, and preparation method therefor and medical use thereof
CN114075212B (en) Fused tricyclic derivatives, preparation method thereof and application thereof in medicines
TW202023572A (en) Cyclic dinucleotide analogues, pharmaceutical composition of analogues and applications of analogues and pharmaceutical composition
CN115557968A (en) Fused tetracyclic compound, preparation method and medical application thereof
CN114057754B (en) Nitrogen-containing bridged ring derivatives, preparation method thereof and application thereof in medicines
TW202214632A (en) Indole fused ring derivatives, their preparation method and medical use
WO2022111636A1 (en) Fused tricyclic compound, preparation method therefor and application thereof in medicine
CN114075219B (en) Quinoline condensed ring derivative, preparation method and medical application thereof
WO2022028389A1 (en) Fused tricyclic derivative, preparation method therefor, and pharmaceutical use thereof
CN112574212B (en) Pyrimido five-membered nitrogen heterocyclic derivative, preparation method and medical application thereof
CN111433201B (en) Benzepine derivative, process for preparing the same and use thereof in medicine
CN114057759B (en) Fused tetracyclic derivative, preparation method thereof and application thereof in medicines
CN112996783B (en) 2-aminopyrimidine derivatives, preparation method and application thereof in medicines
CN114929685B (en) Fused pyridine ring derivatives, preparation method thereof and medical application thereof
CN114057734B (en) Fused tricyclic derivatives, preparation method thereof and application thereof in medicines
CN114276351B (en) Nitrogen-containing heterocyclic derivative, preparation method and medical application thereof
CN109694351B (en) Benzazepine derivatives, process for their preparation and their use in medicine
CN114621230B (en) Nitrogen-containing heterocyclic compound, preparation method thereof and application thereof in medicine
CN113912608B (en) Pyrimidopyrimidinone derivatives, preparation method thereof and application thereof in medicines
CN114456173B (en) Condensed ring group substituted cyclohexanediimide derivative, preparation method and medical application thereof

Legal Events

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