WO2019161803A1 - 肽酰精氨酸脱亚胺酶抑制剂及其用途 - Google Patents

肽酰精氨酸脱亚胺酶抑制剂及其用途 Download PDF

Info

Publication number
WO2019161803A1
WO2019161803A1 PCT/CN2019/076129 CN2019076129W WO2019161803A1 WO 2019161803 A1 WO2019161803 A1 WO 2019161803A1 CN 2019076129 W CN2019076129 W CN 2019076129W WO 2019161803 A1 WO2019161803 A1 WO 2019161803A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
group
hydrogen
amino
alkoxy
Prior art date
Application number
PCT/CN2019/076129
Other languages
English (en)
French (fr)
Inventor
吴永谦
李琳
Original Assignee
南京药捷安康生物科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 南京药捷安康生物科技有限公司 filed Critical 南京药捷安康生物科技有限公司
Priority to EP19757537.6A priority Critical patent/EP3760628A4/en
Priority to US16/975,920 priority patent/US20200407370A1/en
Priority to JP2020568018A priority patent/JP2021515043A/ja
Publication of WO2019161803A1 publication Critical patent/WO2019161803A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs 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
    • 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
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P27/00Drugs for disorders of the senses
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • the present invention belongs to the field of medical technology, and particularly relates to a peptidyl arginine deiminase PAD4 inhibitor compound represented by the general formula (I), and pharmaceutically acceptable salts, stereoisomers and tautomers thereof, And their pharmaceutical compositions, pharmaceutical preparations and uses.
  • the compound of the present invention has an inhibitory effect on peptidyl arginine deiminase PAD4 and can be used for treating various diseases such as rheumatoid arthritis, vasculitis, systemic lupus erythematosus, ulcerative colitis, and multiple Sclerosing, cystic fibrosis, cancer, cutaneous lupus erythematosus, asthma and psoriasis.
  • PAD4 is a member of the peptidylarginine deiminase (PAD) family, has a homodimeric structure, is a Ca 2+ -dependent enzyme, and contains 5 calcium-binding sites per monomer. It consists of 663 amino acid residues and has a molecular weight of 74 kDa.
  • the arginine residue in the protein polypeptide is catalyzed to form citrulline in the presence of Ca 2+ .
  • the citrullinated protein often changes its original molecular conformation, which leads to changes in the biochemical activity of the protein and participates in a variety of physiological and pathological reactions. For example, rheumatoid arthritis, systemic lupus erythematosus, ulcerative colitis, vasculitis, cancer, and the like.
  • Rheumatoid arthritis is a chronic, inflammatory, systemic autoimmune disease.
  • the main clinical manifestations are joint swelling and pain caused by synovial membrane of the facet joint, followed by cartilage destruction and narrowing of joint space. Functional disorders such as joint stiffness and deformity.
  • the worldwide incidence rate is 0.4%-1%, of which there are about 5 million rheumatoid arthritis patients in China.
  • the disease is more prevalent in the female population, with a female to male incidence ratio of 3:1 and an onset age of 25-55 years.
  • PAD4 inhibitors can also be used to treat multiple sclerosis.
  • histone citrullination is involved in the formation of neutrophil extracellular Trap (NET) (innate immune response mechanism), while PAD4 inhibitors can reduce neutrophil activity in a variety of disease pathologies. Therefore, PAD4 inhibitors can be used for tissue damage diseases caused by NET formation. These diseases include, but are not limited to, systemic lupus erythematosus, ulcerative colitis, vasculitis, cystic fibrosis, and asthma.
  • PAD4 inhibitors can also be used to treat skin diseases caused by NET formation.
  • PAD4 inhibitors can also be used to treat cancer, and overexpression of PAD4 has been demonstrated in numerous cancers.
  • the inhibitors targeting PAD are in the preclinical research stage, which are directed to their catalytic active sites, which are mainly divided into two major categories: irreversible inhibitors and reversible inhibitors: the first type is mainly PAD enzyme substrate analogs, ie Peptidomimetics, which are non-selective inhibitors, include guanidines, O-haloxines, and tripeptides.
  • the second class is mainly a selective reversible inhibitor of PAD4, and a compound of the following structure (GSK199) is disclosed in WO 2014/015905 A1.
  • GSK199 is currently in preclinical research, the main indication is rheumatoid arthritis (RA), but its in vitro enzymatic activity has not reached the desired level.
  • PAD4 inhibitors At present, there are few varieties of PAD4 inhibitors under development. In order to better meet the huge clinical needs, we aim to develop PAD4 inhibitors with higher activity and better drug-forming properties.
  • the compound of the present invention has a good peptidyl arginine deiminase PAD4 kinase inhibitory activity and can be used for treating or preventing a disease mediated by peptidyl arginine deiminase PAD4.
  • X and Y are each independently CR 6 or N;
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen, C 1-6 alkyl, 3-6 membered cycloalkyl, C 1-6 alkoxy, halo C 1-6 alkyl or halo C 1-6 alkoxy;
  • R 3 is hydrogen, cyano, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkoxy C 1-6 alkyl, -L 1 -Cy 1 , or unsubstituted or Halogen, cyano, amino, hydroxy-substituted C 1-6 alkyl, C 1-6 alkylcarbonyl, C 1-6 alkoxycarbonyl, C 1-6 alkylsulfonyl, C 1-6 alkoxy C a 1-6 alkyl group, a C 1-6 alkylamino group, a (C 1-6 alkyl) 2 amino group, L 1 is absent or a C 1-6 alkylene group, and Cy 1 is a 3-12 membered cycloalkyl group, 3-12 membered cycloalkenyl, 3-12 membered heterocyclic, aryl, 5-10 membered heteroaryl, Cy 1 may optionally be hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl , C
  • R 4 is hydrogen, cyano, 3-6 membered cycloalkyl, or C 1-6 alkyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy, 3-6 membered cycloalkyl, C 1- 6 alkylcarbonyl, C 1-6 alkoxycarbonyl, C 1-6 alkylsulfonyl, C 1-6 alkoxy C 1-6 alkyl, C 1-6 alkylamino, (C 1-6 alkane Base 2 amino group;
  • R 5 is hydrogen, halogen, cyano, amino, hydroxy, -L 2 -Cy 2 , or C 1-6 alkyl, C 2-8 alkenyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy , C 1-6 alkylcarbonyl, C 1-6 alkylcarbonylamino, C 1-6 alkoxy, C 1-6 alkoxycarbonyl, C 1-6 alkylsulfonyl, C 1-6 alkyl sulphide , C 1-6 alkylsulfonylamino, C 1-6 alkylamino, (C 1-6 alkyl) 2 amino, L 2 is absent or is C 1-6 alkylene, C 1-6 Alkoxy, C 2-6 alkenylene, C 1-6 alkyleneamino, Cy 2 is a 3-12 membered cycloalkyl group, a 3-12 membered cycloalkenyl group, a 3-12 membered heterocyclic group, an
  • R 6 is hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl
  • R 7 is hydrogen or C 1-6 alkyl
  • R 8 is hydrogen, halogen, cyano, amino, hydroxy, -L 3 -Cy 3 , or C 1-6 alkyl, C 2-8 alkenyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy , C 1-6 alkylcarbonyl, C 1-6 alkylcarbonylamino, C 1-6 alkoxy, C 1-6 alkoxycarbonyl, L 3 absent or C 1-6 alkylene, C 2 -6 alkenylene group, Cy 3 is a 3-12 membered cycloalkyl group, a 3-12 membered cycloalkenyl group, a 3-12 membered heterocyclic group, an aryl group, a 5-10 membered heteroaryl group, and Cy 3 may optionally be Hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy;
  • R 9 is hydrogen or C 1-6 alkyl
  • R 8 and R 9 and the attached N form a 4-7 membered heterocyclic group which is unsubstituted or substituted with a substituent, a 6-11 membered heterocyclic group, a 7-12 membered spiroheterocyclyl group, 6- a 10-membered heterocyclic group selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkylamino, (C 1 -6 alkyl) 2 amino, -L 4 -Cy 4 , L 4 is absent or is C 1-6 alkylene, Cy 4 is 3-12 membered cycloalkyl, 3-12 membered cycloalkenyl, 3- 12-membered heterocyclic group, aryl group, 5-10 membered heteroaryl group;
  • Ring B is Or a 5-membered heteroaryl
  • ring B and Condensed, * end up, # end down, not specified * end and # end can be fused in any direction;
  • n is an integer from 0-4.
  • X is CR 6 and Y is N.
  • Y is CR 6 and X is N.
  • X and Y are each independently CR 6.
  • X and Y are each independently N.
  • R 1 is hydrogen
  • R 2 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy, halo C 1-6 alkyl or halo C 1-6 alkoxy. In another embodiment, R 2 is hydrogen, C 1-6 alkyl or C 1-6 alkoxy. In another embodiment, R 2 is C 1-6 alkoxy. In another embodiment, R 2 is methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy or tert-butoxy. In another embodiment, R 2 is methoxy.
  • R 3 is hydrogen, cyano, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkoxy C 1-6 alkyl, or unsubstituted or halogenated a cyano group, an amino group, a hydroxy-substituted C 1-6 alkyl group.
  • R 3 is hydrogen or C 1-6 alkyl.
  • R 3 is C 1-6 alkyl.
  • R 3 is methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl or tert-butyl.
  • R 3 is methyl.
  • R 4 is hydrogen, cyano, 3-6 membered cycloalkyl, or C 1-6 unsubstituted or substituted by halogen, cyano, amino, hydroxy, 3-6 membered cycloalkyl. alkyl.
  • R 4 is hydrogen, halogen or substituted or unsubstituted, 3-6 membered cycloalkyl substituted by C 1-6 alkyl.
  • R 4 is hydrogen, or unsubstituted or substituted with halogen, cyclopropyl or cyclobutyl substituted C 1-6 alkyl group.
  • R 4 is C 1-6 alkyl which is unsubstituted or substituted with halogen, cyclopropyl or cyclobutyl.
  • R 4 is methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl unsubstituted or substituted by halogen, cyclopropyl or cyclobutyl. Base or tert-butyl.
  • R 4 is unsubstituted or substituted with halo, cyclopropyl or cyclobutyl substituted methyl or ethyl.
  • R 4 is ethyl, i-butyl, trifluoroethyl, cyclopropylmethyl or cyclobutylmethyl. In another embodiment, R 4 is cyclobutylmethyl or cyclopropylmethyl.
  • R 5 is hydrogen, halogen, cyano, amino, hydroxy, or unsubstituted or substituted by halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy .
  • R 5 is hydrogen, halo, cyano, amino, hydroxy, or substituted or unsubstituted halo, cyano, amino, hydroxy-substituted C 1-6 alkyl.
  • R 5 is hydrogen, halogen, or C 1-6 alkyl, C 1-6 alkoxy, unsubstituted or substituted with halogen.
  • R 5 is hydrogen, halogen, C 1-6 alkyl, C 1-6 alkoxy.
  • R 5 is hydrogen, fluoro, chloro, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, methoxy, Ethoxy, n-propoxy, isopropoxy, n-butoxy or tert-butoxy.
  • R 5 is hydrogen, fluoro, methyl or methoxy.
  • R 5 is hydrogen, fluoro, methyl or methoxy.
  • R 5 is hydrogen.
  • R 6 is hydrogen or C 1-6 alkyl. In another embodiment, R 6 is hydrogen.
  • R 7 is hydrogen
  • R 8 is hydrogen, halogen, cyano, amino, hydroxy, -L 3 -Cy 3 , or C 1-6 alkyl unsubstituted or substituted by halogen, cyano, amino, hydroxy, L 3 is absent or is a C 1-6 alkylene group
  • Cy 3 is a 3-6 membered cycloalkyl group, a 3-12 membered heterocyclic group, an aryl group, a 5-10 membered heteroaryl group
  • Cy 3 may optionally be Hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy are substituted.
  • R 8 is hydrogen, or a substituted or unsubstituted amino group substituted with C 1-6 alkyl.
  • R 8 is -L 3 -Cy 3 , L 3 is absent, Cy 3 is a 3-6 membered cycloalkyl group, and Cy 3 is substituted with an amino group.
  • R 8 is -L 3 -Cy 3 , L 3 is absent, Cy 3 is cyclobutyl, and Cy 3 is substituted with an amino group.
  • R 9 is C 1-6 alkyl. In another embodiment, R 9 is methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl or tert-butyl. In another embodiment, R 9 is methyl.
  • R 8 and R 9 form a 5-6 membered heterocyclyl, 7-10 membered heterocyclyl, 7-11 membered spiroheterocyclic ring, which is unsubstituted or substituted with a N attached.
  • a 7-10 membered bridged heterocyclic group selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • R 8 and R 9 form, with the attached N, a 5-6 membered heterocyclyl group which is unsubstituted or substituted with a substituent selected from the group consisting of amino groups.
  • R 8 and R 9 form, with the attached N, a 5-6 membered saturated nitrogen-containing heterocyclic group which is unsubstituted or substituted with a substituent selected from the group consisting of amino groups.
  • R 8 and R 9 form, with the attached N, an azetidinyl, azepanyl, pyrrolidinyl, piperidinyl, piperazinyl group which is unsubstituted or substituted with a substituent.
  • morpholinyl the substituent is selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • R 8 and R 9 form, with the attached N, a pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl group which is unsubstituted or substituted with a substituent selected from hydrogen. Halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • R 8 and R 9 form, with the attached N, a pyrrolidinyl, piperidinyl, azepanyl group substituted with an amino group.
  • R 8 and R 9 form, with the attached N, a piperidinyl group substituted with an amino group.
  • R 8 and R 9 are bonded to the attached N to form an unsubstituted or substituted group.
  • the substituent is selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • R 8 and R 9 form a substituted amino group with the attached N.
  • R 8 and R 9 are bonded to the attached N to form an unsubstituted or substituted group.
  • the substituent is selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • R 8 and R 9 form a substituted amino group with the attached N.
  • R 8 and R 9 form a substituted amino group with the attached N.
  • R 8 and R 9 are bonded to the attached N to form an unsubstituted or substituted group.
  • the substituent is selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • R 8 and R 9 form a substituted amino group with the attached N.
  • ring B is Or a 5-membered heteroaryl group. In another embodiment, Ring B is a 5-membered heteroaryl. In another embodiment, Ring B is In another embodiment, Ring B is In another embodiment, Ring B is When ring B is a 5-membered heteroaryl group, it is preferred to use a hetero atom in an upward direction Fused.
  • m is 0, 1, 2, 3 or 4. In another embodiment, m is 0, 1, 2 or 3. In another embodiment, m is 0, 1 or 2. In another embodiment, m is 0 or 1. In another embodiment, m is zero.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • X and Y are each independently CR 6 or N;
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen, C 1-6 alkyl, 3-6 membered cycloalkyl, C 1-6 alkoxy, halo C 1-6 alkyl or halo C 1-6 alkoxy;
  • R 3 is hydrogen, cyano, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkoxy C 1-6 alkyl, -L 1 -Cy 1 , or unsubstituted or Halogen, cyano, amino, hydroxy-substituted C 1-6 alkyl, C 1-6 alkylcarbonyl, C 1-6 alkoxycarbonyl, C 1-6 alkylsulfonyl, C 1-6 alkoxy C a 1-6 alkyl group, a C 1-6 alkylamino group, a (C 1-6 alkyl) 2 amino group, L 1 is absent or a C 1-6 alkylene group, and Cy 1 is a 3-12 membered cycloalkyl group, 3-12 membered cycloalkenyl, 3-12 membered heterocyclic, aryl, 5-10 membered heteroaryl, Cy 1 may optionally be hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl , C
  • R 4 is hydrogen, cyano, 3-6 membered cycloalkyl, or C 1-6 alkyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy, 3-6 membered cycloalkyl, C 1- 6 alkylcarbonyl, C 1-6 alkoxycarbonyl, C 1-6 alkylsulfonyl, C 1-6 alkoxy C 1-6 alkyl, C 1-6 alkylamino, (C 1-6 alkane Base 2 amino group;
  • R 5 is hydrogen, halogen, cyano, amino, hydroxy, -L 2 -Cy 2 , or C 1-6 alkyl, C 2-8 alkenyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy , C 1-6 alkylcarbonyl, C 1-6 alkylcarbonylamino, C 1-6 alkoxy, C 1-6 alkoxycarbonyl, C 1-6 alkylsulfonyl, C 1-6 alkyl sulphide , C 1-6 alkylsulfonylamino, C 1-6 alkylamino, (C 1-6 alkyl) 2 amino, L 2 is absent or is C 1-6 alkylene, C 1-6 Alkoxy, C 2-6 alkenylene, C 1-6 alkyleneamino, Cy 2 is a 3-12 membered cycloalkyl group, a 3-12 membered cycloalkenyl group, a 3-12 membered heterocyclic group, an
  • R 6 is hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl
  • R 7 is hydrogen or C 1-6 alkyl
  • R 8 is hydrogen, halogen, cyano, amino, hydroxy, -L 3 -Cy 3 , or C 1-6 alkyl, C 2-8 alkenyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy , C 1-6 alkylcarbonyl, C 1-6 alkylcarbonylamino, C 1-6 alkoxy, C 1-6 alkoxycarbonyl, L 3 absent or C 1-6 alkylene, C 2 -6 alkenylene group, Cy 3 is a 3-12 membered cycloalkyl group, a 3-12 membered cycloalkenyl group, a 3-12 membered heterocyclic group, an aryl group, a 5-10 membered heteroaryl group, and Cy 3 may optionally be Hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy;
  • R 9 is hydrogen or C 1-6 alkyl
  • R 8 and R 9 and the attached N form a 4-7 membered heterocyclic group which is unsubstituted or substituted with a substituent, a 6-11 membered heterocyclic group, a 7-12 membered spiroheterocyclyl group, 6- a 10-membered heterocyclic group selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkylamino, (C 1 -6 alkyl) 2 amino, -L 4 -Cy 4 , L 4 is absent or is C 1-6 alkylene, Cy 4 is 3-12 membered cycloalkyl, 3-12 membered cycloalkenyl, 3- 12-membered heterocyclic group, aryl group, 5-10 membered heteroaryl group;
  • Ring B is a 5-membered heteroaryl; preferably, ring B is More preferably, ring B is Further preferably, ring B is
  • X is CR 6 and Y is N.
  • Y is CR 6 and X is N.
  • X and Y are each independently CR 6.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • X and Y are each independently CR 6 or N;
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy, halo C 1-6 alkyl or halo C 1-6 alkoxy;
  • R 3 is hydrogen, cyano, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkoxy C 1-6 alkyl, or unsubstituted or halogen, cyano, amino, a hydroxy-substituted C 1-6 alkyl group;
  • R 4 is hydrogen, cyano, 3-6 membered cycloalkyl, or C 1-6 alkyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy, 3-6 membered cycloalkyl;
  • R 5 is hydrogen, halogen, cyano, amino, hydroxy, or C 1-6 alkyl, C 1-6 alkoxy which is unsubstituted or substituted by halogen, cyano, amino, hydroxy;
  • R 6 is hydrogen or C 1-6 alkyl
  • R 7 is hydrogen or C 1-6 alkyl
  • R 8 is hydrogen, halogen, cyano, amino, hydroxy, -L 3 -Cy 3 , or C 1-6 alkyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy, and L 3 is absent or C 1-6 alkylene, Cy 3 is a 3-6 membered cycloalkyl group, a 3-12 membered heterocyclic group, an aryl group, a 5-10 membered heteroaryl group, and Cy 3 may be optionally hydrogen, halogen or cyano. , amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy substituted;
  • R 9 is hydrogen or C 1-6 alkyl
  • R 8 and R 9 and the attached N form a 5-6 membered heterocyclic group which is unsubstituted or substituted with a substituent, a 7-10 membered heterocyclic group, a 7-11 membered spiroheterocyclyl group, 7- a 10-membered heterocyclic group selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy;
  • Ring B is a 5-membered heteroaryl; preferably, ring B is More preferably, ring B is Further preferably, ring B is
  • n is an integer from 0-4.
  • X is CR 6 and Y is N.
  • Y is CR 6 and X is N.
  • X and Y are each independently CR 6.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • X and Y are each independently CR 6 or N;
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 3 is hydrogen, C 1-6 alkyl
  • R 4 is hydrogen, or a C 1-6 alkyl group which is unsubstituted or substituted by a halogen, a 3-6 membered cycloalkyl group;
  • R 5 is hydrogen, halogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 6 is hydrogen or C 1-6 alkyl
  • R 7 is hydrogen or C 1-6 alkyl
  • R 8 is hydrogen, -L 3 -Cy 3 , or a C 1-6 alkyl group which is unsubstituted or substituted with an amino group, L 3 is absent, and Cy 3 is a 3-6 membered cycloalkyl group optionally substituted with an amino group;
  • R 9 is hydrogen or C 1-6 alkyl
  • R 8 and R 9 and the attached N form a 4-7 membered heterocyclic group, a 7-12 membered spiroheterocyclyl group which is unsubstituted or substituted, and the substituent is an amino group;
  • Ring B is Or a 5-membered heteroaryl
  • ring B and Condensed, * end up, # end down, not specified * end and # end can be fused in any direction;
  • m 0 or 1.
  • R 8 and R 9 form, with the attached N, a piperidinyl group substituted with an amino group.
  • ring B is a 5-membered heteroaryl; more preferably, ring B is Further preferably, ring B is Still further preferably, ring B is
  • X is CR 6 and Y is N.
  • Y is CR 6 and X is N.
  • X and Y are each independently CR 6.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • X and Y are each independently CR 6 or N;
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 3 is hydrogen, C 1-6 alkyl
  • R 4 is hydrogen, or a C 1-6 alkyl group which is unsubstituted or substituted by a halogen, a 3-6 membered cycloalkyl group;
  • R 5 is hydrogen, halogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 6 is hydrogen or C 1-6 alkyl
  • R 7 is hydrogen or C 1-6 alkyl
  • R 8 and R 9 and the attached N form a 5-6 membered heterocyclic group which is unsubstituted or substituted with a substituent selected from an amino group;
  • Ring B is a 5-membered heteroaryl group
  • n is an integer from 0-4.
  • R 8 and R 9 form, with the attached N, a piperidinyl group substituted with an amino group.
  • ring B is More preferably, ring B is Further preferably, ring B is
  • X is CR 6 and Y is N.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • X and Y are each independently CR 6 or N;
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 3 is hydrogen, C 1-6 alkyl
  • R 4 is hydrogen, or a C 1-6 alkyl group which is unsubstituted or substituted by halogen, cyclopropyl or cyclobutyl;
  • R 5 is hydrogen, C 1-6 alkyl
  • R 6 is hydrogen or C 1-6 alkyl
  • R 7 is hydrogen or C 1-6 alkyl
  • R 8 and R 9 and the attached N form a 5-6 membered heterocyclic group which is unsubstituted or substituted with a substituent selected from an amino group;
  • n is an integer from 0-4.
  • R 8 and R 9 form, with the attached N, a piperidinyl group substituted with an amino group.
  • ring B is More preferably, ring B is
  • X is CR 6 and Y is N.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • R 8 and R 9 form, with the attached N, a pyrrolidinyl group, a piperidinyl group, a piperazinyl group, a morpholinyl group which is unsubstituted or substituted with a substituent, and the substituent is an amino group.
  • X is CR 6 and Y is N.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • R 8 and R 9 form an unsubstituted or substituted group with the attached N
  • the substituent is selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • X is CR 6 and Y is N.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • R 8 and R 9 form an unsubstituted or substituted group with the attached N
  • the substituent is selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • X is CR 6 and Y is N.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • R 8 and R 9 form an unsubstituted or substituted group with the attached N
  • the substituent is selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • X is CR 6 and Y is N.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • R 8 is hydrogen, halogen, cyano, amino, hydroxy, -L 3 -Cy 3 , or C 1-6 alkyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy, and L 3 is absent or C 1-6 alkylene group, Cy 3 is a 3-6 membered cycloalkyl group, a 3-8 membered heterocyclic group, and Cy 3 may be optionally halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy substitution;
  • R 9 is hydrogen or C 1-6 alkyl.
  • X is CR 6 and Y is N.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • X and Y are each independently CR 6 or N;
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 3 is hydrogen, C 1-6 alkyl
  • R 4 is hydrogen, or a C 1-6 alkyl group which is unsubstituted or substituted by a halogen, a 3-6 membered cycloalkyl group;
  • R 5 is hydrogen, halogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 6 is hydrogen or C 1-6 alkyl
  • R 7 is hydrogen or C 1-6 alkyl
  • R 8 is hydrogen, or a C 1-6 alkyl group which is unsubstituted or substituted with an amino group
  • R 9 is hydrogen or C 1-6 alkyl
  • R 8 and R 9 and the attached N form a 5-6 membered saturated nitrogen-containing heterocyclic group which is unsubstituted or substituted with a substituent selected from hydrogen, amino, C 1-6 alkyl;
  • the 5-6 membered saturated nitrogen-containing heterocyclic group is piperidinyl;
  • Ring B is Preferably, ring B is
  • n is an integer from 0-4.
  • X is CR 6 and Y is N.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • X and Y are each independently CR 6 or N;
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 3 is hydrogen, C 1-6 alkyl
  • R 4 is hydrogen, or a C 1-6 alkyl group which is unsubstituted or substituted by a halogen, a 3-6 membered cycloalkyl group;
  • R 5 is hydrogen, halogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 6 is hydrogen or C 1-6 alkyl
  • R 7 is hydrogen or C 1-6 alkyl
  • R 8 and R 9 and the attached N form a 5-6 membered saturated nitrogen-containing heterocyclic group which is unsubstituted or substituted with a substituent selected from hydrogen, amino, C 1-6 alkyl; preferably The 5-6 membered saturated nitrogen-containing heterocyclic group is piperidinyl;
  • Ring B is Preferably, ring B is
  • n is an integer from 0-4.
  • X is CR 6 and Y is N.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • X and Y are each independently CR 6 or N;
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 3 is hydrogen, C 1-6 alkyl
  • R 4 is hydrogen, or a C 1-6 alkyl group which is unsubstituted or substituted by halogen, cyclopropyl or cyclobutyl;
  • R 5 is hydrogen, halogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 6 is hydrogen or C 1-6 alkyl
  • R 7 is hydrogen or C 1-6 alkyl
  • R 8 and R 9 and the attached N form a 5-6 membered saturated nitrogen-containing heterocyclic group which is unsubstituted or substituted with a substituent selected from an amino group; preferably, the 5-6 member saturated a nitrogen heterocyclic group is piperidinyl;
  • Ring B is Preferably, ring B is
  • n 0 or 1, preferably m is 0.
  • X is CR 6 and Y is N.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • X and Y are each independently CR 6 or N;
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 3 is hydrogen, C 1-6 alkyl
  • R 4 is ethyl, isobutyl, trifluoroethyl, cyclopropylmethyl or cyclobutylmethyl.
  • R 5 is hydrogen, halogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 6 is hydrogen or C 1-6 alkyl
  • R 7 is hydrogen or C 1-6 alkyl
  • R 8 and R 9 and the attached N form a 5-6 membered saturated nitrogen-containing heterocyclic group which is unsubstituted or substituted with a substituent selected from an amino group; preferably, the 5-6 member saturated a nitrogen heterocyclic group is piperidinyl;
  • Ring B is Preferably, ring B is
  • n 0 or 1, preferably m is 0.
  • X is CR 6 and Y is N.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • X and Y are each independently CR 6 or N;
  • R 1 is hydrogen
  • R 2 is a methoxy group
  • R 3 is a methyl group
  • R 4 is ethyl, isobutyl, trifluoroethyl, cyclopropylmethyl or cyclobutylmethyl;
  • R 5 is hydrogen, halogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 6 is hydrogen
  • R 7 is hydrogen
  • R 8 is hydrogen, -L 3 -Cy 3 , or a C 1-6 alkyl group which is unsubstituted or substituted with an amino group;
  • L 3 is absent, Cy 3 is a cyclobutyl group, and Cy 3 is substituted with an amino group;
  • R 9 is a methyl group
  • R 8 and R 9 form an amino-substituted pyrrolidinyl, piperidinyl or azepanyl group with the attached N, substituted by an amino group. Or substituted by an amino group
  • m 0 or 1.
  • X is CR 6 and Y is N.
  • X and Y are each independently N.
  • the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof in another preferred embodiment, the compound of the formula (I) or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof,
  • X and Y are each independently CR 6 or N;
  • R 1 is hydrogen
  • R 2 is a methoxy group
  • R 3 is a methyl group
  • R 4 is ethyl, isobutyl, trifluoroethyl, cyclopropylmethyl or cyclobutylmethyl, preferably R 4 is cyclopropylmethyl or cyclobutylmethyl;
  • R 6 is hydrogen
  • R 7 is hydrogen
  • R 8 and R 9 form a piperidinyl group substituted with an amino group with the attached N;
  • X is CR 6 and Y is N.
  • X and Y are each independently N.
  • the invention also claims a pharmaceutical composition
  • a pharmaceutical composition comprising one or more compounds of the invention, or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof, which may optionally contain one or more drugs
  • the carrier is used and optionally formulated into any pharmaceutically acceptable dosage form.
  • the pharmaceutical carrier of the present invention may be one or more solid or liquid carriers suitable for human use.
  • the pharmaceutical carrier preferably has sufficient purity and sufficiently low toxicity and is compatible with the active ingredients of the present invention and does not significantly reduce the efficacy of the active ingredient.
  • the pharmaceutically acceptable carrier can be selected from the group consisting of a filler, a binder, a disintegrant, a lubricant, an aqueous solvent or a non-aqueous solvent, and the like.
  • the pharmaceutical composition of the present invention may optionally be formulated into any pharmaceutically acceptable dosage form, and administered in any suitable manner, for example, by oral, parenteral, rectal or pulmonary administration.
  • This treated patient or subject it can be formulated into tablets, capsules, pills, granules and the like.
  • parenteral administration it can be prepared as an injection solution, a sterile powder for injection, or the like.
  • rectal administration it can be made into a suppository or the like.
  • pulmonary administration it can be formulated into an aerosol, a spray, a powder, and the like.
  • the present invention relates to a compound of the present invention or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof or a pharmaceutical composition thereof for use in the preparation or treatment of peptidyl arginine deiminase PAD4
  • a medicament for mediated diseases including diseases caused by abnormal expression of peptidyl arginine deiminase PAD4, such as rheumatoid arthritis, vasculitis, systemic lupus erythematosus, ulcerative Colitis, multiple sclerosis, cystic fibrosis, cancer, cutaneous lupus erythematosus, asthma, and psoriasis.
  • the invention further relates to a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof, or a pharmaceutical composition thereof, for use in the treatment or prophylaxis of peptidyl arginine deiminase PAD4
  • diseases including diseases caused by abnormal expression of peptidyl arginine deiminase PAD4
  • diseases such as rheumatoid arthritis, vasculitis, systemic lupus erythematosus, ulcerative colitis, multiple Sclerosing, cystic fibrosis, cancer, cutaneous lupus erythematosus, asthma and psoriasis.
  • the invention further relates to a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof, or a pharmaceutical composition thereof, for use in the treatment or prevention of a disease.
  • the disease is a disease mediated by peptidyl arginine deiminase PAD4 (including diseases caused by aberrant expression of peptidyl arginine deiminase PAD4), such as rheumatoid Arthritis, vasculitis, systemic lupus erythematosus, ulcerative colitis, multiple sclerosis, cystic fibrosis, cancer, cutaneous lupus erythematosus, asthma, and psoriasis.
  • the invention further relates to a method of treating or preventing a disease, the method comprising administering to a patient in need thereof a compound of the invention or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof or a medicament thereof combination.
  • the disease is a disease mediated by peptidyl arginine deiminase PAD4 (including diseases caused by aberrant expression of peptidyl arginine deiminase PAD4), such as rheumatoid Arthritis, vasculitis, systemic lupus erythematosus, ulcerative colitis, multiple sclerosis, cystic fibrosis, cancer, cutaneous lupus erythematosus, asthma, and psoriasis.
  • the patient is a mammal, preferably a human.
  • X and Y are respectively CR 6 or N;
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen, C 1-6 alkyl, 3-6 membered cycloalkyl, C 1-6 alkoxy, halo C 1-6 alkyl or halo C 1-6 alkoxy;
  • R 3 is hydrogen, cyano, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkoxy C 1-6 alkyl, -L 1 -Cy 1 , or unsubstituted or Halogen, cyano, amino, hydroxy-substituted C 1-6 alkyl, C 1-6 alkylcarbonyl, C 1-6 alkoxycarbonyl, C 1-6 alkylsulfonyl, C 1-6 alkoxy C a 1-6 alkyl group, a C 1-6 alkylamino group, a (C 1-6 alkyl) 2 amino group, L 1 is absent or a C 1-6 alkylene group, and Cy 1 is a 3-12 membered cycloalkyl group, 3-12 membered cycloalkenyl, 3-12 membered heterocyclic, aryl, 5-10 membered heteroaryl, Cy 1 may optionally be hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl , C
  • R 4 is hydrogen, cyano, 3-6 membered cycloalkyl, or C 1-6 alkyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy, 3-6 membered cycloalkyl, C 1- 6 alkylcarbonyl, C 1-6 alkoxycarbonyl, C 1-6 alkylsulfonyl, C 1-6 alkoxy C 1-6 alkyl, C 1-6 alkylamino, (C 1-6 alkane Base 2 amino group;
  • R 5 is hydrogen, halogen, cyano, amino, hydroxy, -L 2 -Cy 2 , or C 1-6 alkyl, C 2-8 alkenyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy , C 1-6 alkylcarbonyl, C 1-6 alkylcarbonylamino, C 1-6 alkoxy, C 1-6 alkoxycarbonyl, C 1-6 alkylsulfonyl, C 1-6 alkyl sulphide , C 1-6 alkylsulfonylamino, C 1-6 alkylamino, (C 1-6 alkyl) 2 amino, L 2 is absent or is C 1-6 alkylene, C 1-6 Alkoxy, C 2-6 alkenylene, C 1-6 alkyleneamino, Cy 2 is a 3-12 membered cycloalkyl group, a 3-12 membered cycloalkenyl group, a 3-12 membered heterocyclic group, an
  • R 6 is hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl
  • R 7 is hydrogen or C 1-6 alkyl
  • R 8 is hydrogen, halogen, cyano, amino, hydroxy, -L 3 -Cy 3 , or C 1-6 alkyl, C 2-8 alkenyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy , C 1-6 alkylcarbonyl, C 1-6 alkylcarbonylamino, C 1-6 alkoxy, C 1-6 alkoxycarbonyl, L 3 absent or C 1-6 alkylene, C 2 -6 alkenylene group, Cy 3 is a 3-12 membered cycloalkyl group, a 3-12 membered cycloalkenyl group, a 3-12 membered heterocyclic group, an aryl group, a 5-10 membered heteroaryl group, and Cy 3 may optionally be Hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy;
  • R 9 is hydrogen or C 1-6 alkyl
  • R 8 and R 9 and the attached N form a 4-7 membered heterocyclic group which is unsubstituted or substituted with a substituent, a 6-11 membered heterocyclic group, a 7-12 membered spiroheterocyclyl group, 6- a 10-membered heterocyclic group selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkylamino, (C 1 -6 alkyl) 2 amino, -L 4 -Cy 4 , L 4 is absent or is C 1-6 alkylene, Cy 4 is 3-12 membered cycloalkyl, 3-12 membered cycloalkenyl, 3- 12-membered heterocyclic group, aryl group, 5-10 membered heteroaryl group;
  • n is an integer from 0-4.
  • X and Y are respectively CR 6 or N;
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy, halo C 1-6 alkyl or halo C 1-6 alkoxy;
  • R 3 is hydrogen, cyano, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 alkoxy C 1-6 alkyl, or unsubstituted or halogen, cyano, amino, a hydroxy-substituted C 1-6 alkyl group;
  • R 4 is hydrogen, cyano, 3-6 membered cycloalkyl, or C 1-6 alkyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy, 3-6 membered cycloalkyl;
  • R 5 is hydrogen, halogen, cyano, amino, hydroxy, or C 1-6 alkyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy;
  • R 6 is hydrogen or C 1-6 alkyl
  • R 7 is hydrogen or C 1-6 alkyl
  • R 8 is hydrogen, halogen, cyano, amino, hydroxy, -L 3 -Cy 3 , or C 1-6 alkyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy, and L 3 is absent or C 1-6 alkylene, Cy 3 is a 3-6 membered cycloalkyl group, a 3-12 membered heterocyclic group, an aryl group, a 5-10 membered heteroaryl group, and Cy 3 may be optionally hydrogen, halogen or cyano. , amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy substituted;
  • R 9 is hydrogen or C 1-6 alkyl
  • R 8 and R 9 and the attached N form a 5-6 membered heterocyclic group which is unsubstituted or substituted with a substituent, a 7-10 membered heterocyclic group, a 7-11 membered spiroheterocyclyl group, 7- a 10-membered heterocyclic group selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy;
  • n is an integer from 0-4.
  • R 8 and R 9 form a pyrrolidinyl group, a piperidinyl group, a piperazinyl group or a morpholinyl group which is unsubstituted or substituted with a substituent selected from the group consisting of hydrogen, halogen, cyano group, amino group. , hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • R 8 and R 9 form an unsubstituted or substituted group with the attached N
  • the substituent is selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • R 8 and R 9 form an unsubstituted or substituted group with the attached N
  • the substituent is selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • R 8 and R 9 form an unsubstituted or substituted group with the attached N
  • the substituent is selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • R 8 is hydrogen, halogen, cyano, amino, hydroxy, -L 3 -Cy 3 , or C 1-6 alkyl which is unsubstituted or substituted by halogen, cyano, amino, hydroxy, and L 3 is absent or C 1-6 alkylene group, Cy 3 is a 3-6 membered cycloalkyl group, a 3-8 membered heterocyclic group, and Cy 3 may be optionally halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy substitution;
  • R 9 is hydrogen or C 1-6 alkyl.
  • a pharmaceutical composition comprising a compound of any one of embodiments 1-8, or a pharmaceutically acceptable salt, stereoisomer and tautomer thereof, optionally containing one or more pharmaceuticals
  • the carrier is formulated into any of the pharmaceutically acceptable dosage forms.
  • composition according to embodiment 9, which may further comprise one or more second active therapeutic agents, said second therapeutically active agent being an antimetabolite, a growth factor inhibitor, a silk classification inhibitor Anti-tumor hormones, alkylating agents, metals, topoisomerase inhibitors, hormonal drugs, immunomodulators, tumor suppressor genes, cancer vaccines, immunological checkpoints or antibodies or small molecule drugs related to tumor immunotherapy.
  • second active therapeutic agents being an antimetabolite, a growth factor inhibitor, a silk classification inhibitor
  • peptidyl arginine deiminase PAD4 mediated disease is rheumatoid arthritis, vasculitis, systemic lupus erythematosus, ulcerative colitis, multiple sclerosis, pouch Sexual fibrosis, cancer, cutaneous lupus erythematosus, asthma and psoriasis.
  • X and Y are respectively CR 6 or N;
  • R 1 is hydrogen or C 1-6 alkyl
  • R 2 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy
  • R 3 is hydrogen, C 1-6 alkyl
  • R 4 is hydrogen, or a C 1-6 alkyl group which is unsubstituted or substituted by a halogen, a 3-6 membered cycloalkyl group;
  • R 5 is hydrogen, halogen, or C 1-6 alkyl, C 1-6 alkoxy which is unsubstituted or substituted by halogen;
  • R 6 is hydrogen or C 1-6 alkyl
  • R 7 is hydrogen or C 1-6 alkyl
  • R 8 is hydrogen, or a C 1-6 alkyl group which is unsubstituted or substituted with an amino group
  • R 9 is hydrogen or C 1-6 alkyl
  • R 8 and R 9 and the attached N form a 5-6 membered heterocyclic group which is unsubstituted or substituted with a substituent selected from an amino group;
  • n is an integer from 0-4.
  • R 8 and R 9 and the N to be bonded form an azetidinyl group, an azepanyl group, a pyrrolidinyl group, a piperidinyl group, a piperazinyl group, a morpholinyl group which is unsubstituted or substituted with a substituent
  • the substituent is selected from the group consisting of hydrogen, halogen, cyano, amino, hydroxy, C 1-6 alkyl, C 1-6 alkoxy.
  • halogen as used in the present invention means fluorine, chlorine, bromine, iodine or the like, and preferably a fluorine atom or a chlorine atom.
  • halo means that one or more hydrogen atoms in the substituent are substituted by one or more of the same or different halogen atoms.
  • Halogen is as defined above.
  • C 1-6 alkyl group as used in the present invention means a straight or branched alkyl group derived from a hydrocarbon having 1 to 6 carbon atoms and removed by a hydrogen atom, such as a methyl group, an ethyl group, a n-propyl group, Isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, 2-methylbutyl, neopentyl, 1-ethylpropyl, n-hexyl, iso-hexyl Base, 4-methylpentyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 3,3-dimethylbutyl, 2,2-dimethylbutyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 1,3-dimethylbutyl, 2,3-dimethylbutyl, 2-eth
  • C 1-4 alkyl group means the above examples having 1 to 4 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert. Butyl.
  • the "C 2-8 alkenyl group” as used in the present invention means an olefin moiety having 2 to 8 carbon atoms containing at least one carbon-carbon double bond, and a linear or branched olefin group derived by removing one hydrogen atom, such as a vinyl group.
  • the "C 2-8 alkenyl group” contains a carbon-carbon double bond.
  • C 1-6 alkoxy refers to the present invention as hereinbefore defined "C 1-6 alkyl” group linked to the parent molecule through an oxygen atom, i.e., "C 1-6 alkyl -O- "Groups such as methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, n-pentyloxy, neopentyloxy and n-hexyloxy.
  • C 1-4 alkoxy group refers to the above-mentioned example having 1 to 4 carbon atoms, that is, a "C 1-4 alkyl-O-" group such as a methoxy group, an ethoxy group, or a n-propyl group. Oxyl, isopropoxy, n-butoxy, tert-butoxy.
  • C 1-6 alkoxy C 1-6 alkyl group as used in the present invention means a group formed by a C 1-6 alkoxy group substituted with a C 1-6 alkyl group.
  • C 1-6 alkoxycarbonyl group as used in the present invention means a group formed by a C 1-6 alkoxy-C(O)-form.
  • C 1-6 alkylamino means C 1-6 alkyl-NH-, (C 1-6 Alkyl)(C 1-6 alkyl)N-, C 1-6 alkyl-C(O)-NH-, C 1-6 alkyl-S(O) 2 -NH 2 -, C 1-6 a group formed by an alkyl-S(O) 2 -, C 1-6 alkyl-S-, C 1-6 alkyl-C(O)-form.
  • the "sub” of "C 1-6 alkylene, C 2-6 alkenylene, C 1-6 alkyleneoxy, C 1-6 alkyleneamino" as used in the present invention means C 1-6 , respectively.
  • the alkyl group, the C 2-6 alkenyl group, the C 1-6 alkoxy group, and the C 1-6 alkylamino group remove a divalent group derived from two hydrogen atoms.
  • the "fused ring” as used in the present invention means a polycyclic ring structure formed by joining two, two or more cyclic structures in a snail, a snail, or a bridge.
  • the parallel ring refers to a fused ring structure formed by two or more ring structures sharing two adjacent ring atoms with each other (ie, sharing one bond).
  • the bridged ring refers to a fused ring structure formed by two or more ring structures sharing two non-adjacent ring atoms with each other.
  • the spiro ring refers to a fused ring structure formed by two or more ring structures sharing one ring atom with each other.
  • the "3-12 membered cycloalkyl group" as used in the present invention means a monovalent group derived from a 3-12 membered cycloalkane or (as needed) a divalent group which may be monocyclic or bicyclic. Or a polycyclic cycloalkyl system. Unless otherwise specified, all monocyclic, fused rings (including fused in the form of snails, snails, bridges) that may be formed are included. Monocyclic systems are typically cyclic hydrocarbon groups containing from 3 to 12 carbon atoms, such as from 3 to 8 or from 3 to 6 carbon atoms.
  • cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutane, cyclopentyl, cyclohexane, cycloheptyl, cyclooctyl, cyclopentane-1,3-diyl, ring Hexane-1,4-diyl, cycloheptane-1,4-diyl and the like.
  • the fused ring cycloalkyl group includes a cycloalkyl group, a bridged cycloalkyl group, a spirocycloalkyl group.
  • a cycloalkyl group may be 6-11 members and a cycloalkyl group such as 7-10 members and a cycloalkyl group, and representative examples thereof include, but are not limited to, bicyclo [3.1.1] heptane, bicyclo [2.2.1] heptane, Bicyclo [2.2.2] octane, bicyclo [3.2.2] decane, bicyclo [3.3.1] decane and bicyclo [4.2.1] nonyl.
  • the spirocycloalkyl group can be a 7-12 membered spirocycloalkyl group such as a 7-11 membered spirocycloalkyl group, examples of which include, but are not limited to: base.
  • the bridged cycloalkyl group can be a 6-10 membered bridged cycloalkyl group such as a 7-10 membered bridged cycloalkyl group, examples of which include, but are not limited to: base.
  • the "3-12 membered cycloalkenyl group” as used in the present invention means a group having at least one double bond in the group of the above 3-12 membered cycloalkyl group, such as a cyclobutenyl group or a cyclopentene group.
  • Base cyclohexenyl, cycloheptenyl, cyclooctenyl and the like. Therefore, the "3-12 membered cycloalkenyl group” of the present invention includes all monocyclic, fused rings (including fused in the form of a snail, a snail, or a bridge) which may be formed, unless otherwise specified.
  • a 3-8 membered cycloalkenyl group a 7-11 membered spirocycloalkenyl group, a 7-11 membered cycloalkenyl group, a 6-11 membered bridged cycloalkenyl group, and the like.
  • the spirocycloalkenyl group may be a 7-12 membered spirocycloalkenyl group such as a 7-11 membered spirocycloalkenyl group, examples of which include, but are not limited to: base.
  • the bridged cycloalkenyl group may be a 6-10 membered bridged cycloalkenyl group such as a 7-10 membered bridged cycloalkenyl group, examples of which include, but are not limited to: base.
  • the "3-12 membered heterocyclic group” as used in the present invention means a monovalent group derived from a 3-12 membered heterocycloalkane or (as needed) a divalent group, that is, at least 3-12 members. a non-aromatic cyclic group in which a ring carbon atom is replaced by a hetero atom selected from O, S, S(O), S(O) 2 , C(O), N, which preferably contains 1-3 impurities atom.
  • "3-12 membered heterocyclyl” includes monocyclic heterocyclyl, bicyclic heterocyclyl systems or polycyclic heterocyclyl systems wherein one or more of the rings may be saturated or partially saturated, but does not include an aromatic ring. Unless otherwise specified, all single rings, fused rings (including fused in the form of snails, snails, bridges), saturated and partially saturated may be included.
  • the monocyclic heterocyclic group may be a 3-8 membered heterocyclic group such as a 3-6 membered heterocyclic group, a 4-7 membered heterocyclic group or a 5-6 membered heterocyclic group, and a 3-8 membered nitrogen-containing heterocyclic group such as 4 a -7 membered nitrogen-containing heterocyclic group or a 5-6 membered nitrogen-containing heterocyclic group, and a 3-8 membered saturated heterocyclic group such as a 5-6 membered saturated heterocyclic group.
  • Examples thereof include, but are not limited to, aziridine, oxacyclopropane, thietyl, azetidinyl, oxetanyl, thietane, tetrahydrofuranyl, tetrahydrogen Pyrrolyl, tetrahydrothiophenyl, imidazolidinyl, pyrazolidinyl, 1,2-oxazolidinyl, 1,3-oxazolidinyl, 1,2-thiazolidinyl, 1,3-thiazolidinyl , tetrahydro-2H-pyranyl, tetrahydro-2H-thiopyranyl, piperidinyl, piperazinyl, morpholinyl, 1,4-dioxanyl, 1,4-oxothio Cyclohexane, 4,5-dihydroisoxazolyl, 4,5-dihydrooxazolyl, 2,5-dihydrooxazolyl
  • the fused heterocyclic group includes a heterocyclic group, a spiroheterocyclic group and a bridged heterocyclic group which may be saturated, partially saturated or unsaturated, but not aromatic.
  • the fused heterocyclic group may be fused to a benzene ring, a 5-6 membered monocyclic cycloalkyl group, a 5-6 membered monocyclic cycloalkenyl group, a 5-6 membered monocyclic heterocyclic group or a 5-6 membered monocyclic heteroaryl group.
  • a 5-6 membered monocyclic heterocyclic ring of the group may be fused to a benzene ring, a 5-6 membered monocyclic cycloalkyl group, a 5-6 membered monocyclic cycloalkenyl group, a 5-6 membered monocyclic heterocyclic group or a 5-6 membered monocyclic heteroaryl group.
  • the heterocyclic group may be 6-12 members and a heterocyclic group such as 6-11 membered heterocyclic group or 7-10 membered heterocyclic group, 6-11 membered saturated heterocyclic group, 6-11 yuan
  • nitrogen-containing heterocyclic groups include, but are not limited to, 3-azabicyclo[3.1.0]hexane, 3,6-diazabicyclo[3.2.0]heptanyl, 3,8 -diazabicyclo[4.2.0]octyl, 3,7-diazabicyclo[4.2.0]octyl, octahydropyrrolo[3,4-c]pyrrolyl, octahydropyrrole[ 3,4-b]pyrrolyl, octahydropyrrolo[3,4-b][1,4]oxazinyl, octahydro-1H-pyrrolo[3,4-c]pyridyl, 2,3- Dihydrobenzofuran-2-yl, 2,
  • the spiroheterocyclyl group may be a 6-12 membered spiroheterocyclyl group such as a 7-12 membered spiroheterocyclyl group, a 7-12 membered saturated spiroheterocyclyl group, and a 7-12 membered nitrogen-containing spiroheterocyclyl group. Examples thereof including but not limited to:
  • the bridged heterocyclic group may be a 6-12 membered bridged heterocyclic group such as a 6-10 membered bridged heterocyclic group, a 7-10 membered bridged heterocyclic group, and examples thereof include, but are not limited to:
  • aryl group as used in the present invention means a monovalent or, if desired, divalent cyclic aromatic group derived from an aromatic carbocyclic hydrocarbon having 6 to 14 carbon atoms, including phenyl, naphthalene. Base, Fickey, etc.
  • the "5-10 membered heteroaryl group” of the present invention means an aromatic 5-10 membered cyclic group in which at least one ring carbon atom is replaced by a hetero atom selected from O, S, N, preferably containing 1-3.
  • a hetero atom including a carbon atom, a sulfur atom by oxo, or a nitrogen atom, for example, a carbon atom is replaced by C(O), and a sulfur atom is replaced by S(O), S(O) 2 .
  • the heteroaryl group includes a monoheteroaryl group and a fused heteroaryl group, and unless otherwise specified, a certain heteroaryl group includes all monocyclic, fused ring, wholly aromatic, partially aromatic forms which may be formed.
  • the monoheteroaryl group may be a 5-7 membered heteroaryl group such as a 5-6 membered heteroaryl group, examples of which include, but are not limited to, furyl, imidazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl , oxazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyrazolyl, pyrrolyl, tetrazolyl, thiadiazolyl, thienyl, triazolyl and triazinyl.
  • a fused heteroaryl refers to a group formed by condensing a monocyclic heteroaryl ring to a phenyl, cycloalkenyl, heteroaryl, cycloalkyl or heterocyclyl group.
  • a fused heteroaryl group can be 8-12 members and a heteroaryl group such as a 9-10 membered heteroaryl group, examples including, but not limited to, benzimidazolyl, benzofuranyl, benzothienyl , benzooxadiazolyl, benzothiadiazolyl, benzothiazolyl, porphyrinyl, 5,6-dihydroquinolin-2-yl, 5,6-dihydroisoquinolin-1-yl , furopyridinyl, oxazolyl, fluorenyl, isodecyl, isoquinolyl, naphthyridinyl, fluorenyl, quinolinyl, 5,6,7,8-tetrahydroquinolin-2- , 5,6,7,8-tetrahydroquinolinyl, 5,6,7,8-tetrahydroquinolin-4-yl, 5,6,7,8-tetrahydroiso
  • the "pharmaceutically acceptable salt” as used in the present invention means a pharmaceutically acceptable acid or base addition salt or a solvate thereof.
  • Such pharmaceutically acceptable salts include addition salts of acids such as hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, sulfurous acid, formic acid, toluenesulfonic acid, methanesulfonic acid, nitric acid, benzoic acid, citric acid, tartaric acid. , maleic acid, hydroiodic acid, and alkanoic acid (such as acetic acid, HOOC-(CH 2 ) n-COOH (where n is 0 to 4)), and the like.
  • Such pharmaceutically acceptable salts also include salts of the bases such as the sodium, potassium, calcium and ammonium salts and the like.
  • a variety of non-toxic pharmaceutically acceptable addition salts are known to those skilled in the art.
  • a stereoisomer refers to an isomer produced by the arrangement of atoms in a molecule in a spatially different manner.
  • an enantiomer is produced; when the compound has a carbon-carbon double bond or a cyclic structure, a cis-trans isomer is produced; all enantiomers of the compound of formula (I) , diastereomers, racemates, cis and trans isomers, geometric isomers, epimers, and mixtures thereof, are included within the scope of the invention.
  • Tautomer refers to a specific functional group isomer resulting from the rapid movement of an atom in a molecule at two locations. Examples thereof include tautomerization of a carbonyl compound containing ⁇ -H, specifically as (T, T 1 , and T 2 are each independently a group that conforms to the bonding law of the compound) and other protons are tautomeric, such as phenol-keto tautomerism and nitroso-oxime mutual variation. Structure, imine-enamine tautomerism.
  • the compounds of the invention can be prepared by a variety of methods, including standard chemical methods. Unless otherwise stated, any previously defined variables will continue to have the previously defined meaning. Exemplary general synthetic methods are set forth in the schemes below and can be readily modified to prepare other compounds of the invention. Particular compounds of the invention are prepared in the Examples section.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 7 , R 8 , R 9 , ring B, X, Y and m are as defined above, and PG is a suitable protecting group for the amine, such as tert-butyl Oxycarbonyl (Boc) or benzyloxycarbonyl (Cbz), after which a salt of the compound thus formed is prepared, if necessary.
  • PG is a suitable protecting group for the amine, such as tert-butyl Oxycarbonyl (Boc) or benzyloxycarbonyl (Cbz)
  • a suitable reagent such as trifluoroacetic acid, hydrogen chloride in ethanol, trimethylsilyl iodide or Trimethylsilyl triflate, in some embodiments, is added to the phenol) and stirred at a suitable temperature (0 ° C or ambient temperature) for a suitable period of time (eg, 10 minutes to 10 hours).
  • a suitable base such as saturated aqueous sodium hydrogencarbonate solution or aqueous ammonia
  • a suitable base such as saturated aqueous sodium bicarbonate or aqueous ammonia
  • extract with a suitable extractant such as dichloromethane or ethyl acetate
  • a suitable purification method such as silica gel column chromatography
  • R 1 , R 2 , R 3 , R 7 , R 8 and R 9 are as defined above, and PG is a suitable protecting group for the amine, such as t-butoxycarbonyl (Boc) or benzyloxycarbonyl (Cbz), and The compound of IV) is prepared by reaction.
  • R 4 , ring B, R 5 and m are as defined above.
  • a compound of formula (III) and a compound of formula (IV) are added to a suitable solvent (such as ethanol and water), and a suitable reducing agent (such as sodium dithionite) is added at a suitable temperature (eg, 90 ° C to 100 ° C). Heating or microwave stirring for a suitable period of time (eg 4 to 8 hours). After the reaction is completed, the reaction solution is poured into an appropriate amount of water, extracted with a suitable extractant (such as ethyl acetate), concentrated under reduced pressure, and then separated by a suitable purification method (such as silica gel column chromatography, preparative thin layer chromatography, etc.). a compound of formula (II).
  • a suitable solvent such as ethanol and water
  • a suitable reducing agent such as sodium dithionite
  • R 1 , R 2 , R 3 and R 7 are as defined above, reacted with a compound of formula (VIII) to prepare.
  • R 8 and R 9 are as defined above, and PG is a suitable protecting group for the amine, such as t-butoxycarbonyl (Boc) or benzyloxycarbonyl (Cbz).
  • a compound of formula (VII) and a compound of formula (VIII) are added to a suitable solvent (such as tetrahydrofuran, DMF) and a suitable peptide coupling agent (such as 2-(7-benzotriazole)- N,N,N',N'-tetramethyluronium hexafluorophosphate), then add a suitable base (such as N, N-diisopropylethylamine), stir at the appropriate temperature (such as ambient temperature) The appropriate time period (such as 2 to 10 hours).
  • a suitable solvent such as tetrahydrofuran, DMF
  • a suitable peptide coupling agent such as 2-(7-benzotriazole)- N,N,N',N'-tetramethyluronium hexafluorophosphate
  • a suitable base such as N, N-diisopropylethylamine
  • the reaction solution is concentrated under reduced pressure, and an appropriate amount of water is added, and extracted with a suitable extracting agent (such as ethyl acetate), the extract is concentrated, and the crude product is isolated by a conventional purification method (such as silica gel column chromatography) to obtain a compound of formula (III). .
  • a suitable extracting agent such as ethyl acetate
  • the extract is concentrated, and the crude product is isolated by a conventional purification method (such as silica gel column chromatography) to obtain a compound of formula (III).
  • the compound of formula (IV) can be a compound of formula (IX)
  • R 4 , X, ring B, R 5 and m are as defined above, prepared by oxidation.
  • a compound of formula (IX) is added to a suitable solvent (such as dichloromethane), a suitable oxidizing agent (such as manganese dioxide) is added, and stirred at a suitable temperature (such as reflux) for a suitable period of time (eg, 2 hours). .
  • a suitable solvent such as dichloromethane
  • a suitable oxidizing agent such as manganese dioxide
  • a suitable temperature such as reflux
  • a suitable period of time eg, 2 hours.
  • the reaction was filtered, the cake was rinsed with an appropriate solvent, and the filtrate was concentrated under reduced pressure to give compound of formula (IV).
  • R 4 , X, ring B, R 5 and m are as defined above, and R 10 is methyl or ethyl, prepared by reduction.
  • a compound of formula (X) is added to a suitable solvent (such as tetrahydrofuran), and a suitable reducing agent (such as lithium aluminum hydride or diisobutylaluminum hydride) is added and stirred at a suitable temperature (e.g., ambient temperature). Time period (such as 2 to 10 hours). Add a suitable quenching reagent (such as water), filter, dry with a desiccant (such as anhydrous sodium sulfate), and concentrate the filtrate under reduced pressure to give the compound of formula (IX).
  • a suitable solvent such as tetrahydrofuran
  • a suitable reducing agent such as lithium aluminum hydride or diisobutylaluminum hydride
  • R 4 L wherein R 4 , X, ring B, R 5 , R 10 and m are as defined above, and L is a leaving group (eg, halogen, triflate) .
  • a compound of the formula (XI) and R 4 L are added to a suitable solvent (such as acetonitrile), a suitable base (such as potassium carbonate or cesium carbonate) is added, and the mixture is suitably stirred at a suitable temperature (e.g., ambient temperature to 90 ° C). Time period (such as 1 to 35 hours). After the reaction is completed, it is filtered, and the filtrate is concentrated under reduced pressure, and the crude product is isolated by a conventional purification method (e.g., silica gel column chromatography) to give the compound of formula (X).
  • a suitable solvent such as acetonitrile
  • a suitable base such as potassium carbonate or cesium carbonate
  • the compound of formula (IV) can be a compound of formula (XII)
  • a compound of the formula (XII) and R 4 L are added to a suitable solvent (such as DMF), and a suitable base (such as sodium hydride or potassium carbonate) is added and stirred at a suitable temperature (e.g., -20 ° C to 40 ° C). The appropriate time period (such as 1 to 2 hours). After the reaction is completed, water is added, and the mixture is extracted with a suitable extracting agent (e.g., ethyl acetate), and the extract is concentrated under reduced pressure. The crude product is purified by a conventional purification method (e.g., silica gel column chromatography) to give the compound of formula (IV).
  • a suitable solvent such as DMF
  • a suitable base such as sodium hydride or potassium carbonate
  • the appropriate time period such as 1 to 2 hours.
  • water is added, and the mixture is extracted with a suitable extracting agent (e.g., ethyl acetate), and the extract is concentrated under reduced pressure.
  • R 6 , ring B, R 5 , R 10 and m are as defined above.
  • R 4 , ring B, R 5 and m are as defined above.
  • DMF dimethylformamide
  • THF tetrahydrofuran
  • EA ethyl acetate
  • DCM dichloromethane
  • TMEDA tetramethyl Ethylenediamine
  • LDA lithium diisopropylamide
  • PE petroleum ether
  • DIPEA N,N-diisopropylethylamine
  • HATU 2-(7- Oxidized benzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate
  • DMSO means dimethyl sulfoxide.
  • Methyl 4-hydroxy-3-methoxybenzoate (25.4 g, 0.139 mol) was dissolved in glacial acetic acid (100 mL), and a mixed solution of nitric acid (10.2 mL) and acetic acid (52 mL) was slowly added dropwise on an ice bath, about 1 The hour is added. The reaction solution was slowly warmed to room temperature and stirred for 4 hours. After filtration, the cake was washed with EtOAc EtOAc (EtOAc)
  • Methyl 4-chloro-3-methoxy-5-nitrobenzoate (14 g, 0.057 mol) was dissolved in DMF (140 mL), cooled to 0 ° C, and a solution of methylamine (2 mol/L) The reaction mixture was heated to 80 ° C for 3 hours, then cooled to room temperature, diluted with water and filtered to give an orange solid product (11.1 g, yield 81%).
  • diisopropylamine (1.48 g, 14.5 mmol, 2.5 eq) was added to dry THF (15 mL), cooled to -45 ° C, and n-butyl lithium (2.4 mol/L, 6 mL, 14.5 mmol) was added dropwise.
  • reaction mixture was poured into water (300 mL), EA (500 mL) was added, filtered, and the filter cake was washed with EA (50 mL ⁇ 2). The filtrate was separated and the organic phase was washed with saturated brine. The mixture was dried, filtered, filtered, and evaporated tolululuu
  • diisopropylamine (1.48 g, 14.5 mmol, 2.5 eq) was added to dry THF (15 mL), cooled to -45 ° C, and n-butyl lithium (2.4 mol/L, 6 mL, 14.5 mmol) was added dropwise.
  • Step 1 Synthesis of 2-azidoacetic acid ethyl acetate
  • Step 3 Synthesis of ethyl 6-ethyl-6H-furo[2,3-b]pyrrole-5-carboxylate
  • Lithium aluminum hydride (0.83 g, 21.9 mmol, 1.5 eq) was added to THF (30 mL) under nitrogen, and 6-ethyl-6H-furo[2,3-b]pyrrole-5- was added dropwise at 0 °C.
  • Step 2 Synthesis of 6-ethyl-6H-thieno[2,3-b]pyrrole-5-carboxylic acid ethyl ester
  • Ethyl 6-ethyl-6H-thieno[2,3-b]pyrrole-5-carboxylate (2.0 g, 8.96 mmol, 1.0 eq) was added to THF (20 mL).
  • Diisobutylaluminum hydride (2M in toluene, 16.2 mL, 22.4 mmol, 2.5 eq) was added dropwise, and the mixture was stirred at 0 ° C for 2 h.
  • the reaction was detected by TLC, and water (1 mL), 15 w/w% aqueous sodium hydroxide (1 mL) and water (2.4 mL) were successively added at 0 °C.
  • Step 2 Synthesis of 2-methyl-6H-thieno[2,3-b]pyrrole-5-carboxylic acid ethyl ester
  • Step 3 Synthesis of ethyl 6-ethyl-2-methyl-6H-thieno[2,3-b]pyrrole-5-carboxylate
  • Ethyl 6-ethyl-2-methyl-6H-thieno[2,3-b]pyrrole-5-carboxylate (4.25 g, 17.91 mmol, 1.0 eq) was added to THF (45 mL). The temperature was lowered to 0 ° C, and diisobutylaluminum hydride (1.5 mol/L in toluene, 29.85 mL, 44.77 mmol, 2.5 eq) was added dropwise. After completion of the dropwise addition, the reaction was stirred at 0 ° C for 2 h.
  • 3-Bromo-4-methylthiophene (30 g, 169.43 mmol, 1.0 eq) was added to diethyl ether (200 mL), cooled to -78 ° C, and n-butyllithium (2.5 mol/L n-hexane solution, 102 mL, 254.15 mmol, 1.5 eq).
  • Step 2 Synthesis of 3-methyl-6H-thieno[2,3-b]pyrrole-5-carboxylic acid ethyl ester
  • Step 3 Synthesis of ethyl 6-ethyl-3-methyl-6H-thieno[2,3-b]pyrrole-5-carboxylate
  • Ethyl 6-ethyl-3-methyl-6H-thieno[2,3-b]pyrrole-5-carboxylate (9.8 g, 41.29 mmol, 1.0 eq) was added to THF (100 mL). The temperature was lowered to 0 ° C, and diisobutylaluminum hydride (1.5 mol/L in toluene, 69 mL, 103.24 mmol, 2.5 eq) was added dropwise. After completion of the dropwise addition, the reaction was stirred at 0 ° C for 2 h.
  • Step 2 Synthesis of (R)-(1-(3-methoxy-4-(methylamino)-5-nitrobenzoyl)piperidin-3-yl)carbamic acid tert-butyl ester
  • Step 4 (R)-(3-Aminopiperidin-1-yl)(2-(5-ethyl-5H-pyrrolo[2,3-b]pyrazine-6-yl)-7-methoxy Synthesis of keto-1-methyl-1H-benzo[d]imidazol-5-yl)methanone
  • Step 1 (R)-(1-(2-(1-ethyl-1H-pyrrolo[3,2-b]pyridin-2-yl)-7-methoxy-1-methyl-1H- Synthesis of tert-butyl benzo[d]imidazol-5-carbonyl)piperidin-3-yl)carbamate
  • Step 1 (R)-(1-(2-(6-ethyl-6H-thieno[2,3-b]pyrrol-5-yl)-7-methoxy-1-methyl-1H- Synthesis of tert-butyl benzo[d]imidazol-5-carbonyl)piperidin-3-yl)carbamate
  • Step 1 (R)-(1-(2-(6-ethyl-6H-furo[2,3-b]pyrrol-5-yl)-7-methoxy-1-methyl-1H- Synthesis of tert-butyl benzo[d]imidazol-5-carbonyl)piperidin-3-yl)carbamate
  • Step 1 Synthesis of (R)-(1-(3-Amino-5-methoxy-4-(methylamino)benzoyl)piperidin-3-yl)carbamic acid tert-butyl ester
  • Step 2 (R)-(1-(2-(3-ethyl-3H-thieno[2,3-d]imidazol-2-yl)-7-methoxy-1-methyl-1H- Synthesis of tert-butyl benzo[d]imidazol-5-carbonyl)piperidin-3-yl)carbamate
  • Step 2 Synthesis of (R)-(1-(3-methoxy-4-(methylamino)-5-nitrobenzoyl)piperidin-3-yl)carbamic acid tert-butyl ester
  • Step 3 (R)-(1-(2-(6-ethyl-2-methyl-6H-thieno[2,3-b]pyrrol-5-yl)-7-methoxy-1- Synthesis of tert-butyl methyl-1H-benzo[d]imidazol-5-carbonyl)piperidin-3-yl)carbamate
  • Step 4 (R)-(3-Aminopiperidin-1-yl)(2-(6-ethyl-2-methyl-6H-thieno[2,3-b]pyrrole-5-yl)- Synthesis of 7-methoxy-1-methyl-1H-benzo[d]imidazol-5-yl)methanone
  • Step 1 (R)-(1-(2-(6-ethyl-3-methyl-6H-thieno[2,3-b]pyrrol-5-yl)-7-methoxy-1- Synthesis of tert-butyl methyl-1H-benzo[d]imidazol-5-carbonyl)piperidin-3-yl)carbamate
  • Step 1 Synthesis of tert-butyl (3-((4-methoxybenzyl)(methyl)amino)cyclobutyl)carbamate
  • tert-Butyl (3-oxocyclobutyl)carbamate (2.69 g, 14.5 mmol, 1.1 eq) was dissolved in dichloromethane (10.0 mL) and 1-(4-methoxyphenyl)-N- Methylmethylamine (2.0 g, 13.2 mmol, 1.0 eq) and glacial acetic acid (0.2 mL) were stirred at room temperature for 2 hr, and sodium triacetoxyborohydride (5.6 g, 26.4 mmol, 2.0 eq. overnight. The reaction was completed by TLC. EtOAc EtOAc (EtOAc) ,
  • Step 2 Synthesis of tert-butyl (3-(methylamino)cyclobutyl)carbamate
  • Step 3 Synthesis of tert-butyl (3-(3-methoxy-N-methyl-4-(methylamino)-5-nitrobenzamide)cyclobutyl)carbamate
  • Step 4 (3-(2-(6-Ethyl-6H-thieno[2,3-b]pyrrole-5-yl)-7-methoxy-N,1-dimethyl-1H-benzene Synthesis of [d]imidazole-5-formylamino)cyclobutyl)carbamic acid tert-butyl ester
  • Step 5 N-(3-Aminocyclobutyl)-2-(6-ethyl-6H-thieno[2,3-b]pyrrole-5-yl)-7-methoxy-N,1- Synthesis of Dimethyl-1H-benzo[d]imidazole-5-carboxamide
  • Methyl 3-methoxy-4-(methylamino)-5-nitrobenzoate (3.0 g, 12.4 mmol, 1.0 eq) was dissolved in tetrahydrofuran (30 mL), and lithium hydroxide monohydrate (2.0 g) was added dropwise. , 49.9 mmol, 4.0 eq) of aqueous solution (6 mL), and stirred at 45 ° C overnight, the reaction was completed by TLC, and concentrated under reduced pressure. Water (60 mL) was added to the reaction mixture, and the pH was adjusted to 2 with 2 mol/L hydrochloric acid in an ice bath. The mixture was extracted with dichloromethane (50 mL).
  • Step 2 (6-(3-Methoxy-4-(methylamino)-5-nitrobenzoyl)-6-azaspiro[2.5]octane-4-yl)carbamic acid tert-butyl ester synthesis
  • Step 3 (6-(2-(6-ethyl-6H-thieno[2,3-b]pyrrole-5-yl)-7-methoxy-1-methyl-1H-benzo[d] Synthesis of tert-butyl iodide-5-carbonyl)-6-azaspiro[2.5]octane-4-yl)carbamate
  • Step 3 (1-(2-(1-Ethyl-1H-pyrrolo[2,3-b]pyridin-2-yl)-7-methoxy-1-methyl-1H-benzo[d] Synthesis of tert-butyl ester of imidazole-5-carbonyl)-1,2,3,4-tetrahydroquinolin-3-yl)carbamate
  • Step 4 (3-Amino-3,4-dihydroquinolin-1(2H)-yl)(2-(1-ethyl-1H-pyrrolo[2,3-b]pyridin-2-yl) Synthesis of -7-methoxy-1-methyl-1H-benzo[d]imidazol-5-yl)methanone
  • Step 2 (6-(2-(1-ethyl-1H-pyrrolo[2,3-b]pyridin-2-yl)-7-methoxy-1-methyl-1H-benzo[d] Synthesis of tert-butyl iodide-5-carbonyl)-6-azaspiro[2.5]octane-4-yl)carbamate
  • Step 1 Synthesis of (R)-(1-(3-methoxy-4-methylamino-5-nitrobenzoyl)pyrrolidin-3-yl)carbamic acid tert-butyl ester
  • Step 2 (R)-(1-(2-(6-ethyl-6H-thieno[2,3-b]pyrrol-5-yl)-7-methoxy-1-methyl-1H- Synthesis of tert-butyl benzo[d]imidazol-5-carbonyl)pyrrolidin-3-yl)carbamate
  • Step 1 Synthesis of 6H-thieno[2,3-b]pyrrole-5-carboxylic acid ethyl ester
  • Step 2 Synthesis of 6-(cyclopropylmethyl)-6H-thieno[2,3-b]pyrrole-5-carboxylic acid ethyl ester
  • Step 5 (R)-(1-(2-(6-(cyclopropylmethyl)-6H-thieno[2,3-b]pyrrol-5-yl)-7-methoxy-1- Synthesis of tert-butyl methyl-1H-benzo[d]imidazol-5-carbonyl)piperidin-3-yl)carbamate
  • Step 6 (R)-(3-Aminopiperidin-1-yl)(2-(6-(cyclopropylmethyl)-6H-thieno[2,3-b]pyrrole-5-yl)- Synthesis of 7-methoxy-1-methyl-1H-benzo[d]imidazol-5-yl)methanone
  • Step 1 Synthesis of (R)-3-(((benzyloxy)carbonyl)amino)azepan-1-carboxylic acid tert-butyl ester
  • Step 3 Synthesis of (R)-(1-(3-methoxy-4-(methylamino)-5-nitrobenzoyl)azetidin-3-yl)carbamic acid benzyl ester
  • Step 4 (R)-(1-(2-(6-ethyl-6H-thieno[2,3-b]pyrrol-5-yl)-7-methoxy-1-methyl-1H- Synthesis of Benzo[d]imidazol-5-carbonyl)azetidin-3-yl)carbamate
  • Step 5 (R)-(3-Aminoazetidin-1-yl)(2-(6-ethyl-6H-thieno[2,3-b]pyrrole-5-yl)-7- Synthesis of methoxy-1-methyl-1H-benzo[d]imidazol-5-yl)methanone
  • Step 1 Synthesis of the intermediate 6-isobutyl-6H-thieno[2,3-b]pyrrole-5-carboxylic acid ethyl ester
  • Step 4 Intermediate (R)-(1-(2-(6-isobutyl-6H-thieno[2,3-b]pyrrol-5-yl)-7-methoxy-1-methyl Synthesis of -1H-benzo[d]imidazol-5-carbonyl)piperidin-3-yl)carbamic acid tert-butyl ester
  • Step 2 Synthesis of ethyl 6-(2,2,2-trifluoroethyl)-6H-thieno[2,3-b]pyrrole-5-carboxylate
  • Lithium tetrahydrogen aluminum (79.39 mg, 2.09 mmol, 1.0 eq) was taken in tetrahydrofuran (20 mL) and cooled to 0.
  • Ethyl 6-(2,2,2-trifluoroethyl)-6H-thieno[2,3-b]pyrrole-5-carboxylate (580 mg, 2.09 mmol, 1.0 eq) was dissolved in tetrahydrofuran (4 mL) It was slowly added to a suspension of lithium tetrahydrogenate in tetrahydrofuran to maintain the temperature of the reaction system at around 0 °C. After the addition, stirring was continued for 2 hours, and TLC showed the reaction was completed.
  • Step 5 (R)-(1-(7-Methoxy-1-methyl-2-(6-(2,2,2-trifluoroethyl)-6H-thieno[2,3-b Synthesis of tert-butyl pyrrol-5-yl)-1H-benzo[d]imidazol-5-carbonyl)piperidin-3-yl)carbamate
  • Step 6 Compound (R)-(3-Aminopiperidin-1-yl)(7-methoxy-1-methyl-2-(6-(2,2,2-trifluoroethyl)-6H Synthesis of thieno[2,3-b]pyrrole-5-yl)-1H-benzo[d]imidazol-5-yl)methanone
  • Step 1 Synthesis of the intermediate 6-(cyclobutylmethyl)-6H-thieno[2,3-b]pyrrole-5-carboxylic acid ethyl ester
  • Step 4 Intermediate (R)-(1-(2-(6-(cyclobutylmethyl)-6H-thieno[2,3-b]pyrrol-5-yl)-7-methoxy-1- Synthesis of tert-butyl methyl-1H-benzo[d]imidazol-5-carbonyl)piperidin-3-yl)carbamate
  • Step 5 Compound (R)-(3-Aminopiperidin-1-yl)(2-(6-(cyclobutylmethyl)-6H-thieno[2,3-b]pyrrole-5-yl)-7 Synthesis of 1-methoxy-1-methyl-1H-benzo[d]imidazol-5-yl)methanone
  • Step 2 Synthesis of ethyl 2-azido-3-(5-methylthiophen-3-yl)acrylate
  • Step 4 Synthesis of 6-(cyclopropylmethyl)-2-methyl-6H-thieno[2,3-b]pyrrole-5-carboxylic acid ethyl ester
  • Lithium tetrahydrogen aluminum (0.78 g, 20.5 mmol, 1.5 eq) was suspended in tetrahydrofuran (40 mL), cooled to 0 ° C, 6-(cyclopropylmethyl)-2-methyl-6H-thiophene [2]
  • Ethyl 3-b-pyrrole-5-carboxylate (3.6 g, 13.67 mmol, 1.0 eq) was dissolved in tetrahydrofuran (20 mL) and slowly added to lithium tetrahydrochloride to maintain the temperature of the reaction system at around 0 °C. After the addition, stirring was continued for 2 hours, and TLC showed the reaction was completed.
  • Step 7 (R)-(1-(2-(6-(cyclopropylmethyl)-2-methyl-6H-thieno[2,3-b]pyrrole-5-yl)-7-A Synthesis of tert-butyl oxy-1-methyl-1H-benzo[d]imidazol-5-carbonyl)piperidin-3-yl)carbamate
  • Step 8 (R)-(3-Aminopiperidin-1-yl)(2-(6-(cyclopropylmethyl)-2-methyl-6H-thieno[2,3-b]pyrrole- Synthesis of 5-yl)-7-methoxy-1-methyl-1H-benzo[d]imidazol-5-yl)methanone
  • Step 3 Synthesis of 2-azido-3-(furan-3-yl)acrylic acid ethyl ester
  • Step 5 Synthesis of 6-(cyclopropylmethyl)-6H-furo[2,3-b]pyrrole-5-carboxylic acid ethyl ester
  • Lithium tetrahydrogen aluminum (1.3 g, 34.1 mmol, 1.5 eq) was taken in tetrahydrofuran (100 mL), cooled to 0 ° C, 6-(cyclopropylmethyl)-6H-furo[2,3-b] Ethyl pyrrol-5-carboxylate (5.3 g of crude product) was dissolved in tetrahydrofuran (20 mL) and slowly added to the lithium tetrahydrogen aluminum suspension to maintain the temperature of the reaction system ⁇ -5 °C. After the addition, stirring was continued for 2 hours, and TLC showed the reaction was completed.
  • Step 8 (R)-(1-(2-(6-(cyclopropylmethyl)-6H-furo[2,3-b]pyrrol-5-yl)-7-methoxy-1- Synthesis of tert-butyl methyl-1H-benzo[d]imidazol-5-carbonyl)piperidin-3-yl)carbamate
  • Step 9 (R)-(3-Aminopiperidin-1-yl)(2-(6-(cyclopropylmethyl)-6H-furo[2,3-b]pyrrole-5-yl)- Synthesis of 7-methoxy-1-methyl-1H-benzo[d]imidazol-5-yl)methanone
  • Test substance a compound of the invention, prepared according to the method of the examples
  • Peptidyl arginine deiminase 4 Cayman, Cat. No. 10500, Lot. No. 0519694;
  • DMSO dimethyl sulfoxide
  • Ethylenediaminetetraacetic acid Sigma, Cat. No. E5134, CAS No. 60-00-4;
  • 96-well plate Corning, Cat. No. 3365, Lot. No. 22008026;
  • 384-well plate Corning, Cat. No. 3573, Lot. No. 12608008.
  • test compound Take the appropriate amount of the test compound and dissolve the DMSO to 10 mM.
  • An appropriate amount of 10 mM test compound mother liquor was diluted with DMSO to 1.5 mM solution, and then subjected to 3-fold gradient dilution with DMSO to obtain a 50 ⁇ concentration compound gradient solution.
  • a 10-fold dilution was carried out with a reaction buffer (100 mM HEPES (4-hydroxyethylpiperazine ethanesulfuric acid) containing 0.01% v/v Triton, pH 8.0) to obtain a 5 ⁇ concentration compound gradient solution ( 150, 50, 16.667, 5.556, 1.852, 0.617, 0.206, 0.069, 0.023, and 0.008 ⁇ M).
  • reaction buffer 100 mM HEPES (4-hydroxyethylpiperazine ethanesulfuric acid) containing 0.01% v/v Triton, pH 8.0
  • Reaction buffer an aqueous solution of 100 mM HEPES (4-hydroxyethylpiperazine ethanesulfuric acid) containing 0.01% v/v Triton, pH 8.0;
  • Stop solution 100 mM HEPES (4-hydroxyethylpiperazine) containing 0.015% w/v Brij-35, 0.2% v/v Coating Reagent #3 (PE, part number 760050) and 50 mM EDTA An aqueous solution of ethanesulfonic acid) having a pH of 8.0;
  • 2.5 ⁇ polypeptide solution Take appropriate amount of carboxyfluorescein-labeled peptide (FAM-AcH4(1-5) peptide) and CaCl 2 solution, and dilute to 2.5 ⁇ with reaction buffer (final concentration of peptide 5 ⁇ M, final concentration of Ca 2+ 0.1 mM).
  • Test object PAD4 IC 50 ( ⁇ M) Compound GSK199 0.14 Compound 3 0.20 Compound 4 0.35 Compound 9 0.11 Compound 18 0.036 Compound 20 0.058
  • the compound of the present invention has a good inhibitory activity against the PAD4 enzyme, and has a good clinical application potential for treating diseases mediated by PAD4 enzyme abnormality.
  • composition of the incubation system is the composition of the incubation system:
  • liver microsomes (20 mg protein/mL) were taken out from the -80 °C refrigerator, placed in a 37 ° C water bath thermostat shaker for 3 min, and thawed for use.
  • Control group (excluding ⁇ -NADPH): 30 ⁇ L of water and 30 ⁇ L of compound working solution (10 ⁇ M) were added to 240 ⁇ L of the mixed solution of step (2), vortexed for 30 s, mixed, total reaction The volume was 300 ⁇ L, and 2 samples were parallel. Incubate into a 37 ° C water bath thermostat and start timing. The sampling time points are 0 min and 60 min.
  • Sample group Take 70 ⁇ L ⁇ -NADPH solution (10 mM) and 70 ⁇ L compound working solution (10 ⁇ M), respectively, and add 560 ⁇ L of the incubation solution of step (2) to the incubation solution.
  • the total volume of the reaction is 700 ⁇ L, vortex for 30 s, and mix. , 2 samples in parallel.
  • the cells were incubated in a 37 ° C water bath thermostat and started to be timed. The sampling time points were 0 min, 5 min, 10 min, 20 min, 30 min and 60 min after the time.
  • V d 1/protein content in liver microsomes
  • k is the slope of the logarithm of the remaining amount of the compound versus time
  • V d is the apparent volume of distribution
  • C 0 is the concentration of 0h drug
  • Ct is the drug concentration at time t
  • t is time
  • e is a natural number
  • Cl int is Inherent clearance rate.
  • the compound of the present invention has good stability in human and canine liver microsomes, and the removal rate is low.
  • the test compound was prepared by dissolving 5v/v% DMSO+20v/v% (30v/v%solutol (polyethylene glycol-15 hydroxystearate, available from BASF, batch number 84773247G0)) +75v/v% saline.
  • the compound solution was administered orally to the SD rats at a dose of 5.0 mg/kg, and the blood collection time points were: 15 min, 30 min, 1 h, 2 h, 4 h, 6 h, 8 h, and 24 h.
  • test compound was prepared by dissolving 5v/v% DMSO+20v/v% (30v/v%solutol)+75v/v% saline, and the solution of the compound was intravenously injected into the SD rat at a dose of 1 mg/kg.
  • the time points of blood collection were: 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 6 h, 8 h and 24 h.
  • the animals were fixed, and the tail was heated in a water bath 10 min before each time point, and about 100 ⁇ L of blood was collected through the tail vein, and the blood was collected and placed in an anticoagulation tube containing EDTA-K 2 . Blood samples were centrifuged at 8000 rpm for 6 min at 4 ° C to obtain plasma samples, and plasma was prepared within 30 min after blood collection. Store in a -80 ° C freezer before plasma testing.
  • the sample to be tested was taken out from the -80 ° C refrigerator, vortexed at room temperature for 5 min, and 20 ⁇ L of plasma sample was accurately aspirated into a 1.5 mL centrifuge tube; 200 ⁇ L of an internal standard working solution (toluene butyrate methanol) at a concentration of 100 ng/mL was added. Solution), mix; vortex 5 min, centrifuge at 12000 rpm for 5 min; accurately pipet 50 ⁇ L of supernatant into 96-well plates pre-filled with 150 ⁇ L water/well; vortex for 5 min for LC-MS/MS analysis.
  • an internal standard working solution toluene butyrate methanol
  • the concentration of the test substance was output using AB's Analyst 1.6.3.
  • the mean, standard deviation, coefficient of variation and other parameters were calculated using Microsoft Excel (Analyst 1.6.3 direct output is not calculated), and PK parameters are calculated using Pharsight Phoenix 6.1 software NCA (T max is the median).
  • t z1/2 terminal elimination half-life
  • Cl _obs clearance rate
  • V z_obs apparent volume of distribution
  • T max blood concentration peak time
  • AUC last area under the drug-time curve 0 to 24 hours
  • F% Absolute bioavailability
  • - Not tested.
  • the compound of the present invention has higher exposure and lower clearance than the control drug GSK199.
  • OBJECTIVE To evaluate the stability of mouse/rat liver microsomes of the compounds of the invention.
  • composition of the incubation system is the composition of the incubation system:
  • liver microsomes (20 mg protein/mL) were taken out from the -80 °C refrigerator, placed in a 37 ° C water bath thermostat shaker for 3 min, and thawed for use.
  • Control group (excluding ⁇ -NADPH): 30 ⁇ L of water and 30 ⁇ L of compound working solution (10 ⁇ M) were added to 240 ⁇ L of the mixed solution of step (2), vortexed for 30 s, mixed, total reaction The volume was 300 ⁇ L, and 2 samples were parallel. Incubate into a 37 ° C water bath thermostat and start timing. The sampling time points are 0 min and 60 min.
  • Sample group Take 70 ⁇ L ⁇ -NADPH solution (10 mM) and 70 ⁇ L compound working solution (10 ⁇ M), respectively, and add 560 ⁇ L of the incubation solution of step (2) to the incubation solution.
  • the total volume of the reaction is 700 ⁇ L, vortex for 30 s, and mix. , 2 samples in parallel.
  • the cells were incubated in a 37 ° C water bath thermostat and started to be timed. The sampling time points were 0 min, 5 min, 10 min, 20 min, 30 min and 60 min after the time.
  • k is the slope of the logarithm of the remaining amount of the compound versus time
  • V d is the apparent volume of distribution
  • C 0 is the concentration of 0h drug
  • Ct is the drug concentration at time t
  • t is time
  • e is a natural number
  • Cl int is Inherent clearance rate.
  • the compound of the present invention has good stability in mouse and rat liver microsomes, and the clearance rate is low.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pulmonology (AREA)
  • Rheumatology (AREA)
  • Dermatology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Immunology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)

Abstract

本发明提供了一种式(I)所示的肽酰精氨酸脱亚胺酶PAD4抑制剂化合物或其药学上可接受的盐、立体异构体和互变异构体,以及它们的药物组合物、药物制剂及用途。X、Y、R 1、R 2、R 3、R 4、R 5、R 7、R 8、R 9、环B和m如说明书中所定义。本发明提供的化合物对肽酰精氨酸脱亚胺酶PAD4具有抑制作用,可以用于治疗多种病症,例如类风湿性关节炎、血管炎、系统性红斑狼疮、溃疡性结肠炎、多发性硬化症、囊性纤维化、癌症、皮肤型红斑狼疮、哮喘和银屑病。

Description

肽酰精氨酸脱亚胺酶抑制剂及其用途 技术领域
本发明属于医药技术领域,具体涉及通式(I)所表示的肽酰精氨酸脱亚胺酶PAD4抑制剂化合物及其药学上可接受的盐、立体异构体和互变异构体,以及它们的药物组合物、药物制剂及用途。本发明所涉及的化合物对肽酰精氨酸脱亚胺酶PAD4具有抑制作用,可以用于治疗多种病症,例如类风湿性关节炎、血管炎、系统性红斑狼疮、溃疡性结肠炎、多发性硬化症、囊性纤维化、癌症、皮肤型红斑狼疮、哮喘和银屑病。
背景技术
PAD4是肽酰精氨酸脱亚胺酶(peptidylarginine deiminase,PAD)家族的一员,具有同型二聚体的结构,是Ca 2+依赖性酶,每个单体含有5个钙离子结合位点,由663个氨基酸残基组成,分子量为74kDa,在Ca 2+的存在下将蛋白多肽中的精氨酸残基催化生成瓜氨酸。瓜氨酸化后的蛋白质往往改变其原有分子构象,从而导致蛋白质的生化活性亦发生改变,并参与多种生理和病理反应。比如,类风湿性关节炎、系统性红斑狼疮、溃疡性结肠炎、血管炎、癌症等。
类风湿性关节炎(rheumatoid arthritis,RA)是一种慢性、炎性、系统性的自体免疫疾病,主要临床表现为小关节滑膜所致的关节肿痛,继而软骨破坏、关节间隙变窄、关节僵直、畸形等功能性障碍。世界范围内的发病率为0.4%-1%,其中,中国约有500万名类风湿性关节炎患者。该疾病在女性人群中更加高发,女性与男性发病率比值为3∶1,发病年龄主要集中在25-55周岁。研究发现RA患者血清中PAD4含量明显升高,患者机体可产生自身抗PAD4抗体,并且PAD4瓜氨酸化多种蛋白质,引起机体自身的免疫反应,参与RA的发生与发展。如波形蛋白发生瓜氨酸化后,瓜氨酸肽段被HLA-DR分子识别,引发T细胞免疫反应。
另外,多发性硬化症(multiple sclerosis,MS)患者脑中PAD4表达升高,因此,PAD4抑制剂也可以用于治疗多发性硬化症。同时组蛋白瓜氨酸化与嗜中性粒细胞细胞外Trap(NET)的形成(先天性免疫反 应机制)有关,而PAD4抑制剂可以降低多种疾病病理学下的嗜中性粒细胞活性。因此,PAD4抑制剂可以用于因NET形成所导致的组织损伤疾病。这些疾病包括,但不限于系统性红斑狼疮、溃疡性结肠炎、血管炎、囊性纤维化和哮喘等。另外,NET形成与皮肤疾病的病理学相关,比如银屑病和皮肤型红斑狼疮等。因此,PAD4抑制剂也可以用于治疗由于NET形成引起的皮肤疾病。除此之外,PAD4抑制剂也可以用于治疗癌症,目前已在众多癌症中证实了PAD4的过表达。
目前靶向PAD的抑制剂均处于临床前研究阶段,都是针对其催化活性位点,主要分为不可逆抑制剂和可逆抑制剂两大类:第一类主要是PAD酶底物类似物,即模拟肽,为非选择性抑制剂,包括卤脒类、O-卤脒类和三肽类。第二类主要为PAD4选择性可逆抑制剂,WO2014/015905A1公开了如下结构的化合物(GSK199)。GSK199目前处于临床前研究阶段,主要的适应症是类风湿性关节炎(RA),但其体外酶学活性仍未达到理想水平。
Figure PCTCN2019076129-appb-000001
目前在研的PAD4抑制剂品种较少,为了更好的满足巨大的临床需求,我们旨在开发出具有较高活性、成药性更好的PAD4抑制剂。
发明内容
本发明的目的是提供一类肽酰精氨酸脱亚胺酶PAD4抑制剂的化合物及其药学上可接受的盐、立体异构体和互变异构体。本发明化合物具有良好的肽酰精氨酸脱亚胺酶PAD4激酶抑制活性,可用于治疗或者预防由肽酰精氨酸脱亚胺酶PAD4介导的疾病。
本发明采用的技术方案如下:
通式(I)表示的化合物或其药学上可接受的盐、立体异构体和互变异构体:
Figure PCTCN2019076129-appb-000002
其中,X和Y分别独立地为CR 6或者N;
R 1为氢或C 1-6烷基;
R 2为氢、C 1-6烷基、3-6元环烷基、C 1-6烷氧基、卤代C 1-6烷基或卤代C 1-6烷氧基;
R 3为氢、氰基、C 1-6烷基、C 1-6烷氧基、C 1-6烷氧基C 1-6烷基、-L 1-Cy 1,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 1-6烷基羰基、C 1-6烷氧羰基、C 1-6烷基磺酰基、C 1-6烷氧基C 1-6烷基、C 1-6烷基氨基、(C 1-6烷基) 2氨基,L 1不存在或者为C 1-6亚烷基,Cy 1为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基,Cy 1可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
R 4为氢、氰基、3-6元环烷基,或未被取代或被卤素、氰基、氨基、羟基、3-6元环烷基取代的C 1-6烷基、C 1-6烷基羰基、C 1-6烷氧羰基、C 1-6烷基磺酰基、C 1-6烷氧基C 1-6烷基、C 1-6烷基氨基、(C 1-6烷基) 2氨基;
R 5为氢、卤素、氰基、氨基、羟基、-L 2-Cy 2,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 2-8烯基、C 1-6烷基羰基、C 1-6烷基羰基氨基、C 1-6烷氧基、C 1-6烷氧羰基、C 1-6烷基磺酰基、C 1-6烷基硫基、C 1-6烷基磺酰氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基,L 2不存在或者为C 1-6亚烷基、C 1-6亚烷氧基、C 2-6亚烯基、C 1-6亚烷氨基,Cy 2为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基,Cy 2可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
R 6为氢、C 1-6烷基或C 3-6环烷基;
R 7为氢或C 1-6烷基;
R 8为氢、卤素、氰基、氨基、羟基、-L 3-Cy 3,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 2-8烯基、C 1-6烷基羰基、C 1-6烷基羰基氨基、C 1-6烷氧基、C 1-6烷氧羰基,L 3不存在或者为C 1-6亚烷基、C 2-6亚烯基,Cy 3为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳 基、5-10元杂芳基,Cy 3可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
R 9为氢或C 1-6烷基;
或者,R 8和R 9与所连接的N形成未被取代或被取代基取代的4-7元杂环基、6-11元并杂环基、7-12元螺杂环基、6-10元桥杂环基,所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、-L 4-Cy 4,L 4不存在或者为C 1-6亚烷基,Cy 4为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基;
环B为
Figure PCTCN2019076129-appb-000003
或5元杂芳基;
其中,环B与
Figure PCTCN2019076129-appb-000004
的稠合,*端向上,#端向下,未指明*端和#端的,可以任意方向稠合;
条件是,
当环B为
Figure PCTCN2019076129-appb-000005
且R 8和R 9与所连接的N形成
Figure PCTCN2019076129-appb-000006
时,X为N;
当环B为
Figure PCTCN2019076129-appb-000007
且X为CR 6时,R 8和R 9与所连接的N形成
Figure PCTCN2019076129-appb-000008
m为0-4的整数。
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,Y为CR 6,X为N。
在一个实施方案中,X和Y分别独立地为CR 6
在一个实施方案中,X和Y分别独立地为N。
在一个实施方案中,R 1为氢。
在一个实施方案中,R 2为氢、C 1-6烷基、C 1-6烷氧基、卤代C 1-6烷 基或卤代C 1-6烷氧基。在另一个实施方案中,R 2为氢、C 1-6烷基或C 1-6烷氧基。在另一个实施方案中,R 2为C 1-6烷氧基。在另一个实施方案中,R 2为甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基或叔丁氧基。在另一个实施方案中,R 2为甲氧基。
在一个实施方案中,R 3为氢、氰基、C 1-6烷基、C 1-6烷氧基、C 1-6烷氧基C 1-6烷基,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基。在另一个实施方案中,R 3为氢或C 1-6烷基。在另一个实施方案中,R 3为C 1-6烷基。在另一个实施方案中,R 3为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基。在另一个实施方案中,R 3为甲基。
在一个实施方案中,R 4为氢、氰基、3-6元环烷基,或未被取代或被卤素、氰基、氨基、羟基、3-6元环烷基取代的C 1-6烷基。在另一个实施方案中,R 4为氢,或未被取代或被卤素、3-6元环烷基取代的C 1-6烷基。在另一个实施方案中,R 4为氢,或未被取代或被卤素、环丙基或环丁基取代的C 1-6烷基。在另一个实施方案中,R 4为未被取代或被卤素、环丙基或环丁基取代的C 1-6烷基。在另一个实施方案中,R 4为未被取代或被卤素、环丙基或环丁基取代的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基。在另一个实施方案中,R 4为未被取代或被卤素、环丙基或环丁基取代的甲基或乙基。在另一个实施方案中,R 4为乙基、异丁基、三氟乙基、环丙基甲基或环丁基甲基。在另一个实施方案中,R 4为环丁基甲基或环丙基甲基。
在一个实施方案中,R 5为氢、卤素、氰基、氨基、羟基,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 1-6烷氧基。在另一个实施方案中,R 5为氢、卤素、氰基、氨基、羟基,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基。在另一个实施方案中,R 5为氢、卤素,或未被取代或被卤素取代的C 1-6烷基、C 1-6烷氧基。在另一个实施方案中,R 5为氢、卤素、C 1-6烷基、C 1-6烷氧基。在另一个实施方案中,R 5为氢、氟、氯、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基或叔丁氧基。在另一个实施方案中,R 5为氢、氟、甲基或甲氧基。在另一个实施方案中,R 5为氢、氟、甲基或甲氧基。在另一个实施方案中,R 5为氢。
在一个实施方案中,R 6为氢或C 1-6烷基。在另一个实施方案中,R 6为氢。
在一个实施方案中,R 7为氢。
在一个实施方案中,R 8为氢、卤素、氰基、氨基、羟基、-L 3-Cy 3,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基,L 3不存在或者为C 1-6亚烷基,Cy 3为3-6元环烷基、3-12元杂环基、芳基、5-10元杂芳基,Cy 3可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代。在另一个实施方案中,R 8为氢,或未被取代或被氨基取代的C 1-6烷基。在另一个实施方案中,R 8为-L 3-Cy 3,L 3不存在,Cy 3为3-6元环烷基,Cy 3被氨基取代。在另一个实施方案中,R 8为-L 3-Cy 3,L 3不存在,Cy 3为环丁基,Cy 3被氨基取代。
在一个实施方案中,R 9为C 1-6烷基。在另一个实施方案中,R 9为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基。在另一个实施方案中,R 9为甲基。
在一个实施方案中,R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元杂环基、7-10元并杂环基、7-11元螺杂环基、7-10元桥杂环基,所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。在一个实施方案中,R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元杂环基,所述取代基选自氨基。在一个实施方案中,R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元饱和含氮杂环基,所述取代基选自氨基。在另一个实施方案中,R 8和R 9与所连接的N形成未被取代或被取代基取代的氮杂环丁基、氮杂环庚基、吡咯烷基、哌啶基、哌嗪基、吗啉基,所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。在另一个实施方案中,R 8和R 9与所连接的N形成未被取代或被取代基取代的吡咯烷基、哌啶基、哌嗪基、吗啉基,所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。在另一个实施方案中,R 8和R 9与所连接的N形成被氨基取代的吡咯烷基、哌啶基、氮杂环庚基。在另一个实施方案中,R 8和R 9与所连接的N形成被氨基取代的哌啶基。在另一个实施方案中,R 8和R 9与所连接的N形成未被取代或被取代基取代的
Figure PCTCN2019076129-appb-000009
Figure PCTCN2019076129-appb-000010
Figure PCTCN2019076129-appb-000011
所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。在另一个实施方案中,R 8和R 9与所连接的N形成被氨基取代的
Figure PCTCN2019076129-appb-000012
在另一个实施方案中,R 8和R 9与所连接的N形成未被取代或被取代基取代的
Figure PCTCN2019076129-appb-000013
Figure PCTCN2019076129-appb-000014
Figure PCTCN2019076129-appb-000015
所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。在另一个实施方案中,R 8和R 9与所连接的N形成被氨基取代的
Figure PCTCN2019076129-appb-000016
Figure PCTCN2019076129-appb-000017
Figure PCTCN2019076129-appb-000018
在另一个实施方案中,R 8和R 9与所连接的N形成被氨基取代的
Figure PCTCN2019076129-appb-000019
在另一个实施方案中,R 8和R 9与所连接的N形成未被取代或被取代基取代的
Figure PCTCN2019076129-appb-000020
Figure PCTCN2019076129-appb-000021
所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。在另一个实施方案中,R 8和R 9与所连接的N形成被氨基取代的
Figure PCTCN2019076129-appb-000022
在一个实施方案中,环B为
Figure PCTCN2019076129-appb-000023
或5元杂芳基。在另一个实施方案中,环B为5元杂芳基。在另一个实施方案中,环B为
Figure PCTCN2019076129-appb-000024
在另一个实施方案中,环B为
Figure PCTCN2019076129-appb-000025
Figure PCTCN2019076129-appb-000026
在另一个实施方案中,环B为
Figure PCTCN2019076129-appb-000027
当环B为5元杂芳基时,优选以杂原子向上的方向与
Figure PCTCN2019076129-appb-000028
稠合。
在一个实施方案中,m为0、1、2、3或4。在另一个实施方案中,m为0、1、2或3。在另一个实施方案中,m为0、1或2。在另一个实施方案中,m为0或1。在另一个实施方案中,m为0。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
其中,X和Y分别独立地为CR 6或者N;
R 1为氢或C 1-6烷基;
R 2为氢、C 1-6烷基、3-6元环烷基、C 1-6烷氧基、卤代C 1-6烷基或卤代C 1-6烷氧基;
R 3为氢、氰基、C 1-6烷基、C 1-6烷氧基、C 1-6烷氧基C 1-6烷基、-L 1-Cy 1,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 1-6烷基羰基、C 1-6烷氧羰基、C 1-6烷基磺酰基、C 1-6烷氧基C 1-6烷基、C 1-6烷基氨基、(C 1-6烷基) 2氨基,L 1不存在或者为C 1-6亚烷基,Cy 1为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基,Cy 1可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
R 4为氢、氰基、3-6元环烷基,或未被取代或被卤素、氰基、氨基、羟基、3-6元环烷基取代的C 1-6烷基、C 1-6烷基羰基、C 1-6烷氧羰基、C 1-6烷基磺酰基、C 1-6烷氧基C 1-6烷基、C 1-6烷基氨基、(C 1-6烷基) 2氨基;
R 5为氢、卤素、氰基、氨基、羟基、-L 2-Cy 2,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 2-8烯基、C 1-6烷基羰基、C 1-6烷基 羰基氨基、C 1-6烷氧基、C 1-6烷氧羰基、C 1-6烷基磺酰基、C 1-6烷基硫基、C 1-6烷基磺酰氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基,L 2不存在或者为C 1-6亚烷基、C 1-6亚烷氧基、C 2-6亚烯基、C 1-6亚烷氨基,Cy 2为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基,Cy 2可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
R 6为氢、C 1-6烷基或C 3-6环烷基;
R 7为氢或C 1-6烷基;
R 8为氢、卤素、氰基、氨基、羟基、-L 3-Cy 3,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 2-8烯基、C 1-6烷基羰基、C 1-6烷基羰基氨基、C 1-6烷氧基、C 1-6烷氧羰基,L 3不存在或者为C 1-6亚烷基、C 2-6亚烯基,Cy 3为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基,Cy 3可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
R 9为氢或C 1-6烷基;
或者,R 8和R 9与所连接的N形成未被取代或被取代基取代的4-7元杂环基、6-11元并杂环基、7-12元螺杂环基、6-10元桥杂环基,所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、-L 4-Cy 4,L 4不存在或者为C 1-6亚烷基,Cy 4为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基;
环B为5元杂芳基;优选地,环B为
Figure PCTCN2019076129-appb-000029
更优选地,环B为
Figure PCTCN2019076129-appb-000030
进一步优选地,环B为
Figure PCTCN2019076129-appb-000031
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,Y为CR 6,X为N。
在一个实施方案中,X和Y分别独立地为CR 6
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
其中,X和Y分别独立地为CR 6或者N;
R 1为氢或C 1-6烷基;
R 2为氢、C 1-6烷基、C 1-6烷氧基、卤代C 1-6烷基或卤代C 1-6烷氧基;
R 3为氢、氰基、C 1-6烷基、C 1-6烷氧基、C 1-6烷氧基C 1-6烷基,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基;
R 4为氢、氰基、3-6元环烷基,或未被取代或被卤素、氰基、氨基、羟基、3-6元环烷基取代的C 1-6烷基;
R 5为氢、卤素、氰基、氨基、羟基,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 1-6烷氧基;
R 6为氢或C 1-6烷基;
R 7为氢或C 1-6烷基;
R 8为氢、卤素、氰基、氨基、羟基、-L 3-Cy 3,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基,L 3不存在或者为C 1-6亚烷基,Cy 3为3-6元环烷基、3-12元杂环基、芳基、5-10元杂芳基,Cy 3可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
R 9为氢或C 1-6烷基;
或者,R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元杂环基、7-10元并杂环基、7-11元螺杂环基、7-10元桥杂环基,所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基;
环B为5元杂芳基;优选地,环B为
Figure PCTCN2019076129-appb-000032
更优选地,环B为
Figure PCTCN2019076129-appb-000033
进一步优选地,环B为
Figure PCTCN2019076129-appb-000034
m为0-4的整数。
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,Y为CR 6,X为N。
在一个实施方案中,X和Y分别独立地为CR 6
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
其中,X和Y分别独立地为CR 6或者N;
R 1为氢或C 1-6烷基;
R 2为氢、C 1-6烷基、C 1-6烷氧基;
R 3为氢、C 1-6烷基;
R 4为氢,或未被取代或被卤素、3-6元环烷基取代的C 1-6烷基;
R 5为氢、卤素、C 1-6烷基、C 1-6烷氧基;
R 6为氢或C 1-6烷基;
R 7为氢或C 1-6烷基;
R 8为氢、-L 3-Cy 3,或未被取代或被氨基取代的C 1-6烷基,L 3不存在,Cy 3为任选被氨基取代的3-6元环烷基;
R 9为氢或C 1-6烷基;
或者,R 8和R 9与所连接的N形成未被取代或被取代基取代的4-7元杂环基、7-12元螺杂环基,所述取代基为氨基;
环B为
Figure PCTCN2019076129-appb-000035
或5元杂芳基;
其中,环B与
Figure PCTCN2019076129-appb-000036
的稠合,*端向上,#端向下,未指明*端和#端的,可以任意方向稠合;
条件是,
当环B为
Figure PCTCN2019076129-appb-000037
且R 8和R 9与所连接的N形成
Figure PCTCN2019076129-appb-000038
时,X为N;
当环B为
Figure PCTCN2019076129-appb-000039
且X为CR 6时,R 8和R 9与所连接的N形成
Figure PCTCN2019076129-appb-000040
m为0或1。
优选地,R 8和R 9与所连接的N形成被氨基取代的哌啶基。
优选地,环B为5元杂芳基;更优选地,环B为
Figure PCTCN2019076129-appb-000041
Figure PCTCN2019076129-appb-000042
进一步优选地,环B为
Figure PCTCN2019076129-appb-000043
再进一步优选地,环B为
Figure PCTCN2019076129-appb-000044
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,Y为CR 6,X为N。
在一个实施方案中,X和Y分别独立地为CR 6
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
其中,X和Y分别独立地为CR 6或者N;
R 1为氢或C 1-6烷基;
R 2为氢、C 1-6烷基、C 1-6烷氧基;
R 3为氢、C 1-6烷基;
R 4为氢,或未被取代或被卤素、3-6元环烷基取代的C 1-6烷基;
R 5为氢、卤素、C 1-6烷基、C 1-6烷氧基;
R 6为氢或C 1-6烷基;
R 7为氢或C 1-6烷基;
R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元杂环基,所述取代基选自氨基;
环B为5元杂芳基;
m为0-4的整数。
优选地,R 8和R 9与所连接的N形成被氨基取代的哌啶基。
优选地,环B为
Figure PCTCN2019076129-appb-000045
更优选地,环B为
Figure PCTCN2019076129-appb-000046
Figure PCTCN2019076129-appb-000047
进一步优选地,环B为
Figure PCTCN2019076129-appb-000048
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
其中,X和Y分别独立地为CR 6或者N;
R 1为氢或C 1-6烷基;
R 2为氢、C 1-6烷基、C 1-6烷氧基;
R 3为氢、C 1-6烷基;
R 4为氢,或未被取代或被卤素、环丙基或环丁基取代的C 1-6烷基;
R 5为氢、C 1-6烷基;
R 6为氢或C 1-6烷基;
R 7为氢或C 1-6烷基;
R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元杂环基,所述取代基选自氨基;
环B为
Figure PCTCN2019076129-appb-000049
m为0-4的整数。
优选地,R 8和R 9与所连接的N形成被氨基取代的哌啶基。
优选地,环B为
Figure PCTCN2019076129-appb-000050
更优选地,环B为
Figure PCTCN2019076129-appb-000051
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
R 8和R 9与所连接的N形成未被取代或被取代基取代的吡咯烷基、哌啶基、哌嗪基、吗啉基,所述取代基为氨基。
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
R 8和R 9与所连接的N形成未被取代或被取代基取代的
Figure PCTCN2019076129-appb-000052
Figure PCTCN2019076129-appb-000053
Figure PCTCN2019076129-appb-000054
所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
R 8和R 9与所连接的N形成未被取代或被取代基取代的
Figure PCTCN2019076129-appb-000055
Figure PCTCN2019076129-appb-000056
Figure PCTCN2019076129-appb-000057
所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
R 8和R 9与所连接的N形成未被取代或被取代基取代的
Figure PCTCN2019076129-appb-000058
Figure PCTCN2019076129-appb-000059
所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
R 8为氢、卤素、氰基、氨基、羟基、-L 3-Cy 3,或未被取代或被卤素、 氰基、氨基、羟基取代的C 1-6烷基,L 3不存在或者为C 1-6亚烷基,Cy 3为3-6元环烷基、3-8元杂环基,Cy 3可任选被卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
R 9为氢或C 1-6烷基。
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
其中,X和Y分别独立地为CR 6或者N;
R 1为氢或C 1-6烷基;
R 2为氢、C 1-6烷基、C 1-6烷氧基;
R 3为氢、C 1-6烷基;
R 4为氢,或未被取代或被卤素、3-6元环烷基取代的C 1-6烷基;
R 5为氢、卤素、C 1-6烷基、C 1-6烷氧基;
R 6为氢或C 1-6烷基;
R 7为氢或C 1-6烷基;
R 8为氢,或未被取代或被氨基取代的C 1-6烷基;
R 9为氢或C 1-6烷基;
或者,R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元饱和含氮杂环基,所述取代基选自氢、氨基、C 1-6烷基;优选地,所述5-6元饱和含氮杂环基为哌啶基;
环B为
Figure PCTCN2019076129-appb-000060
优选地,环B为
Figure PCTCN2019076129-appb-000061
m为0-4的整数。
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
其中,X和Y分别独立地为CR 6或者N;
R 1为氢或C 1-6烷基;
R 2为氢、C 1-6烷基、C 1-6烷氧基;
R 3为氢、C 1-6烷基;
R 4为氢,或未被取代或被卤素、3-6元环烷基取代的C 1-6烷基;
R 5为氢、卤素、C 1-6烷基、C 1-6烷氧基;
R 6为氢或C 1-6烷基;
R 7为氢或C 1-6烷基;
R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元饱和含氮杂环基,所述取代基选自氢、氨基、C 1-6烷基;优选地,所述5-6元饱和含氮杂环基为哌啶基;
环B为
Figure PCTCN2019076129-appb-000062
优选地,环B为
Figure PCTCN2019076129-appb-000063
m为0-4的整数。
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
其中,X和Y分别独立地为CR 6或者N;
R 1为氢或C 1-6烷基;
R 2为氢、C 1-6烷基、C 1-6烷氧基;
R 3为氢、C 1-6烷基;
R 4为氢,或未被取代或被卤素、环丙基、环丁基取代的C 1-6烷基;
R 5为氢、卤素、C 1-6烷基、C 1-6烷氧基;
R 6为氢或C 1-6烷基;
R 7为氢或C 1-6烷基;
R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元饱和含氮杂环基,所述取代基选自氨基;优选地,所述5-6元饱和含氮杂环基为哌啶基;
环B为
Figure PCTCN2019076129-appb-000064
优选地,环B为
Figure PCTCN2019076129-appb-000065
m为0或1,优选地,m为0。
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
其中,X和Y分别独立地为CR 6或者N;
R 1为氢或C 1-6烷基;
R 2为氢、C 1-6烷基、C 1-6烷氧基;
R 3为氢、C 1-6烷基;
R 4为乙基、异丁基、三氟乙基、环丙基甲基或环丁基甲基。
R 5为氢、卤素、C 1-6烷基、C 1-6烷氧基;
R 6为氢或C 1-6烷基;
R 7为氢或C 1-6烷基;
R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元饱和含氮杂环基,所述取代基选自氨基;优选地,所述5-6元饱和含氮杂环基为哌啶基;
环B为
Figure PCTCN2019076129-appb-000066
优选地,环B为
Figure PCTCN2019076129-appb-000067
m为0或1,优选地,m为0。
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
其中,X和Y分别独立地为CR 6或者N;
R 1为氢;
R 2为甲氧基;
R 3为甲基;
R 4为乙基、异丁基、三氟乙基、环丙基甲基或环丁基甲基;
R 5为氢、卤素、C 1-6烷基、C 1-6烷氧基;
R 6为氢;
R 7为氢;
R 8为氢、-L 3-Cy 3,或未被取代或被氨基取代的C 1-6烷基;
L 3不存在,Cy 3为环丁基,Cy 3被氨基取代;
R 9为甲基;
或者,R 8和R 9与所连接的N形成被氨基取代的吡咯烷基、哌啶基或氮杂环庚基,被氨基取代的
Figure PCTCN2019076129-appb-000068
或被氨基取代的
Figure PCTCN2019076129-appb-000069
环B为
Figure PCTCN2019076129-appb-000070
m为0或1。
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,X和Y分别独立地为N。
在另一优选例中,式(I)所示的化合物或其药学上可接受的盐、立体异构体和互变异构体,
其中,X和Y分别独立地为CR 6或者N;
R 1为氢;
R 2为甲氧基;
R 3为甲基;
R 4为乙基、异丁基、三氟乙基、环丙基甲基或环丁基甲基,优选地,R 4为环丙基甲基或环丁基甲基;
R 6为氢;
R 7为氢;
R 8和R 9与所连接的N形成被氨基取代的哌啶基;
环B为
Figure PCTCN2019076129-appb-000071
m为0。
在一个实施方案中,X为CR 6,Y为N。
在一个实施方案中,X和Y分别独立地为N。
本发明的部分化合物如下表所示:
Figure PCTCN2019076129-appb-000072
Figure PCTCN2019076129-appb-000073
本发明还要求保护含有本发明的一种或多种化合物或其药学上可接受的盐、立体异构体和互变异构体的药物组合物,其可以任选含有一种或多种药用载体并任选被制成药学上可接受的任一剂型。
本发明所述的药用载体可以是一种或多种适合于人使用的固体或液体载体。所述药用载体优选具有足够的纯度和足够低的毒性,并且与本发明活性成分具有相容性且不明显减低活性成分的药效。例如,药用载体可以选自填充剂、粘合剂、崩解剂、润滑剂、水性溶剂或非水性溶剂等。
本发明所述的药物组合物,可以任选被制成药学上可接受的任一剂 型,以任何合适的给药方式,例如通过口服、肠胃外、直肠或经肺给药等方式施用于需要这种治疗的患者或受试者。用于口服给药时,可以制成片剂、胶囊剂、丸剂、颗粒剂等。用于肠胃外给药时,可以制成注射液、注射用无菌粉末等。用于直肠给药时,可以制成栓剂等。用于经肺给药时,可以制成气雾剂、喷雾剂和粉雾剂等。
本发明涉及本发明的化合物或其药学上可接受的盐、立体异构体和互变异构体或者它们的药物组合物在制备用于治疗或者预防由肽酰精氨酸脱亚胺酶PAD4介导的疾病(包括由肽酰精氨酸脱亚胺酶PAD4异常表达所导致的疾病)的药物中的用途,所述疾病例如类风湿性关节炎、血管炎、系统性红斑狼疮、溃疡性结肠炎、多发性硬化症、囊性纤维化、癌症、皮肤型红斑狼疮、哮喘和银屑病。
本发明进一步涉及本发明的化合物或其药学上可接受的盐、立体异构体和互变异构体或者它们的药物组合物用于治疗或者预防由肽酰精氨酸脱亚胺酶PAD4介导的疾病(包括由肽酰精氨酸脱亚胺酶PAD4异常表达所导致的疾病)的用途,所述疾病例如类风湿性关节炎、血管炎、系统性红斑狼疮、溃疡性结肠炎、多发性硬化症、囊性纤维化、癌症、皮肤型红斑狼疮、哮喘和银屑病。
本发明进一步涉及本发明的化合物或其药学上可接受的盐、立体异构体和互变异构体或者它们的药物组合物,其用于治疗或者预防疾病。在一个实施方案中,所述疾病为由肽酰精氨酸脱亚胺酶PAD4介导的疾病(包括由肽酰精氨酸脱亚胺酶PAD4异常表达所导致的疾病),例如类风湿性关节炎、血管炎、系统性红斑狼疮、溃疡性结肠炎、多发性硬化症、囊性纤维化、癌症、皮肤型红斑狼疮、哮喘和银屑病。
本发明进一步涉及一种治疗或者预防疾病的方法,所述方法包括给需要其的患者施用本发明的化合物或其药学上可接受的盐、立体异构体和互变异构体或者它们的药物组合物。在一个实施方案中,所述疾病为由肽酰精氨酸脱亚胺酶PAD4介导的疾病(包括由肽酰精氨酸脱亚胺酶PAD4异常表达所导致的疾病),例如类风湿性关节炎、血管炎、系统性红斑狼疮、溃疡性结肠炎、多发性硬化症、囊性纤维化、癌症、皮肤型红斑狼疮、哮喘和银屑病。在一个实施方案中,所述患者为哺乳动物,优选为人。
本发明的其它实施方案
本发明还涉及以下实施方案:
1.通式(I)表示的化合物或其药学上可接受的盐、立体异构体和互变异构体:
Figure PCTCN2019076129-appb-000074
其中,X和Y分别为CR 6或者N;
R 1为氢或C 1-6烷基;
R 2为氢、C 1-6烷基、3-6元环烷基、C 1-6烷氧基、卤代C 1-6烷基或卤代C 1-6烷氧基;
R 3为氢、氰基、C 1-6烷基、C 1-6烷氧基、C 1-6烷氧基C 1-6烷基、-L 1-Cy 1,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 1-6烷基羰基、C 1-6烷氧羰基、C 1-6烷基磺酰基、C 1-6烷氧基C 1-6烷基、C 1-6烷基氨基、(C 1-6烷基) 2氨基,L 1不存在或者为C 1-6亚烷基,Cy 1为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基,Cy 1可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
R 4为氢、氰基、3-6元环烷基,或未被取代或被卤素、氰基、氨基、羟基、3-6元环烷基取代的C 1-6烷基、C 1-6烷基羰基、C 1-6烷氧羰基、C 1-6烷基磺酰基、C 1-6烷氧基C 1-6烷基、C 1-6烷基氨基、(C 1-6烷基) 2氨基;
R 5为氢、卤素、氰基、氨基、羟基、-L 2-Cy 2,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 2-8烯基、C 1-6烷基羰基、C 1-6烷基羰基氨基、C 1-6烷氧基、C 1-6烷氧羰基、C 1-6烷基磺酰基、C 1-6烷基硫基、C 1-6烷基磺酰氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基,L 2不存在或者为C 1-6亚烷基、C 1-6亚烷氧基、C 2-6亚烯基、C 1-6亚烷氨基,Cy 2为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基,Cy 2可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
R 6为氢、C 1-6烷基或C 3-6环烷基;
R 7为氢或C 1-6烷基;
R 8为氢、卤素、氰基、氨基、羟基、-L 3-Cy 3,或未被取代或被卤素、 氰基、氨基、羟基取代的C 1-6烷基、C 2-8烯基、C 1-6烷基羰基、C 1-6烷基羰基氨基、C 1-6烷氧基、C 1-6烷氧羰基,L 3不存在或者为C 1-6亚烷基、C 2-6亚烯基,Cy 3为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基,Cy 3可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
R 9为氢或C 1-6烷基;
或者,R 8和R 9与所连接的N形成未被取代或被取代基取代的4-7元杂环基、6-11元并杂环基、7-12元螺杂环基、6-10元桥杂环基,所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、-L 4-Cy 4,L 4不存在或者为C 1-6亚烷基,Cy 4为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基;
环B为
Figure PCTCN2019076129-appb-000075
m为0-4的整数。
2.如实施方案1所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
其中,X和Y分别为CR 6或者N;
R 1为氢或C 1-6烷基;
R 2为氢、C 1-6烷基、C 1-6烷氧基、卤代C 1-6烷基或卤代C 1-6烷氧基;
R 3为氢、氰基、C 1-6烷基、C 1-6烷氧基、C 1-6烷氧基C 1-6烷基,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基;
R 4为氢、氰基、3-6元环烷基,或未被取代或被卤素、氰基、氨基、羟基、3-6元环烷基取代的C 1-6烷基;
R 5为氢、卤素、氰基、氨基、羟基,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基;
R 6为氢或C 1-6烷基;
R 7为氢或C 1-6烷基;
R 8为氢、卤素、氰基、氨基、羟基、-L 3-Cy 3,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基,L 3不存在或者为C 1-6亚烷基,Cy 3为3-6元环烷基、3-12元杂环基、芳基、5-10元杂芳基,Cy 3可任选被 氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
R 9为氢或C 1-6烷基;
或者,R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元杂环基、7-10元并杂环基、7-11元螺杂环基、7-10元桥杂环基,所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基;
环B为
Figure PCTCN2019076129-appb-000076
m为0-4的整数。
3.如实施方案2所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
R 8和R 9与所连接的N形成未被取代或被取代基取代的吡咯烷基、哌啶基、哌嗪基、吗啉基,所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。
4.如实施方案2所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
R 8和R 9与所连接的N形成未被取代或被取代基取代的
Figure PCTCN2019076129-appb-000077
Figure PCTCN2019076129-appb-000078
Figure PCTCN2019076129-appb-000079
所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。
5.如实施方案2所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
R 8和R 9与所连接的N形成未被取代或被取代基取代的
Figure PCTCN2019076129-appb-000080
Figure PCTCN2019076129-appb-000081
Figure PCTCN2019076129-appb-000082
所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。
6.如实施方案2所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
R 8和R 9与所连接的N形成未被取代或被取代基取代的
Figure PCTCN2019076129-appb-000083
Figure PCTCN2019076129-appb-000084
所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。
7.如实施方案2所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
R 8为氢、卤素、氰基、氨基、羟基、-L 3-Cy 3,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基,L 3不存在或者为C 1-6亚烷基,Cy 3为3-6元环烷基、3-8元杂环基,Cy 3可任选被卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
R 9为氢或C 1-6烷基。
8.如实施方案1所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,选自如下结构的化合物:
Figure PCTCN2019076129-appb-000085
Figure PCTCN2019076129-appb-000086
9.含有实施方案1-8任一项所述的化合物或其药学上可接受的盐、立体异构体和互变异构体的药物组合物,可以任选含有一种或多种药用载体,制成药学上可接受的任一剂型。
10.如实施方案9所述的药物组合物,还可以包括一种或多种第二活性治疗剂,所述的第二治疗活性剂为抗代谢物、生长因子抑制剂、有丝分类抑制剂、抗肿瘤激素类、烷化剂类、金属类、拓扑异构酶抑制剂、激素药、免疫调节剂、肿瘤抑制基因、癌疫苗、免疫检查点或肿瘤免疫治疗相关的抗体或小分子药物。
11.实施方案1-8任一项所述的化合物或其药学上可接受的盐、立体异构体和互变异构体和实施方案9所述的药物组合物在制备治疗或者预防由肽酰精氨酸脱亚胺酶PAD4介导的疾病的药物中的用途。
12.实施方案11所述的用途,肽酰精氨酸脱亚胺酶PAD4介导的疾病为类风湿性关节炎、血管炎、系统性红斑狼疮、溃疡性结肠炎、多发性硬化症、囊性纤维化、癌症、皮肤型红斑狼疮、哮喘和银屑病。
13.如实施方案2所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
其中,X和Y分别为CR 6或者N;
R 1为氢或C 1-6烷基;
R 2为氢、C 1-6烷基、C 1-6烷氧基;
R 3为氢、C 1-6烷基;
R 4为氢,或未被取代或被卤素、3-6元环烷基取代的C 1-6烷基;
R 5为氢、卤素,或未被取代或被卤素取代的C 1-6烷基、C 1-6烷氧基;
R 6为氢或C 1-6烷基;
R 7为氢或C 1-6烷基;
R 8为氢,或未被取代或被氨基取代的C 1-6烷基;
R 9为氢或C 1-6烷基;
或者,R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元杂环基,所述取代基选自氨基;
环B为
Figure PCTCN2019076129-appb-000087
m为0-4的整数。
14.如实施方案2所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
R 8和R 9与所连接的N形成未被取代或被取代基取代的氮杂环丁基、氮杂环庚基、吡咯烷基、哌啶基、哌嗪基、吗啉基,所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。
发明详述
本发明所述的“卤素”是指氟、氯、溴、碘等,优选氟原子、氯原子。
本发明所述的“卤代”是指取代基中的一个或多个氢原子被一个或多个相同或不同的卤素原子取代。“卤素”如前文所定义。
本发明所述的“C 1-6烷基”指含有1-6个碳原子的烃部分去除一个氢原子衍生的直链或支链的烷基,如甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、异戊基、2-甲基丁基、新戊基、1-乙基丙基、正己基、异己基、4-甲基戊基、3-甲基戊基、2-甲基戊基、1-甲基戊基、3,3-二甲基丁基、2,2-二甲基丁基、1,1-二甲基丁基、1,2-二甲基丁基、1,3-二甲基丁基、2,3-二甲基丁基、2-乙基丁基和1-甲基-2-甲基丙基等。所述“C 1-4烷基”指含有1-4个碳原子的上述实例,如甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基。
本发明所述的“C 2-8烯基”指含有至少一个碳碳双键的2-8个碳原子 的烯烃部分去除一个氢原子衍生的直链或支链的烯烃基,如乙烯基、1-丙烯基、2-丙烯基、1-丁烯基、2-丁烯基、1,3-丁二-1-烯基、1-戊烯-3-基、2-戊烯-1-基、3-戊烯-1-基、3-戊烯-2-基、1,3-戊二烯-1-基、1,4-戊二烯-3-基、1-己烯-3-基、1,4-己二烯-1-基。优选地,“C 2-8烯基”中含有一个碳碳双键。
本发明所述的“C 1-6烷氧基”是指前文所定义的“C 1-6烷基”通过氧原子与母体分子连接的基团,即“C 1-6烷基-O-”基团,如甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、叔丁氧基、正戊氧基、新戊氧基和正己氧基等。所述的“C 1-4烷氧基”指含有1-4个碳原子的上述实例,即“C 1-4烷基-O-”基团,如甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、叔丁氧基。
本发明所述的“C 1-6烷氧基C 1-6烷基”是指C 1-6烷氧基取代C 1-6烷基所形成的基团。
本发明所述的“C 1-6烷氧基羰基”是指以C 1-6烷氧基-C(O)-方式所形成的基团。
本发明所述的“C 1-6烷基氨基”、“(C 1-6烷基) 2氨基”、“C 1-6烷基羰基氨基”、“C 1-6烷基磺酰氨基”、“C 1-6烷基磺酰基”、“C 1-6烷基硫基”、“C 1-6烷基羰基”分别指以C 1-6烷基-NH-、(C 1-6烷基)(C 1-6烷基)N-、C 1-6烷基-C(O)-NH-、C 1-6烷基-S(O) 2-NH 2-、C 1-6烷基-S(O) 2-、C 1-6烷基-S-、C 1-6烷基-C(O)-方式所形成的基团。
本发明所述的“C 1-6亚烷基、C 2-6亚烯基、C 1-6亚烷氧基、C 1-6亚烷氨基”的“亚”分别是指C 1-6烷基、C 2-6烯基、C 1-6烷氧基、C 1-6烷氨基基团去除两个氢原子衍生的二价基团。
本发明所述的“稠环”是指由两个或两个以上环状结构以并、螺、桥的连接方式所形成的多环系结构。所述的并环是指由两个或两个以上环状结构彼此共用两个相邻的环原子(即共用一个键)所形成的稠环结构。所述的桥环是指由两个或两个以上环状结构彼此共用两个非相邻的环原子所形成的稠环结构。所述的螺环是指由两个或两个以上环状结构彼此共用一个环原子所形成的稠环结构。
本发明所述的“3-12元环烷基”,是指从3-12元环烷烃衍生得到的一价基团或(根据需要的)二价基团,其可以是单环、双环、或者多环环烷基系统。在不特别指明的情况下,包括可能形成的所有单环、稠环(包 括以并、螺、桥的形式稠合)的情形。单环系统通常是含3-12个碳原子如3-8个或3-6个碳原子的环烃基团。环烷基实例包括但不限于:环丙烷基、环丁烷基、环戊烷基、环己烷基、环庚烷基、环辛烷基、环戊烷-1,3-二基、环己烷-1,4-二基、环庚烷-1,4-二基等。稠环环烷基包括并环烷基、桥环烷基、螺环烷基。并环烷基可以为6-11元并环烷基如7-10元并环烷基,其代表性例子包括但不限于双环[3.1.1]庚烷、双环[2.2.1]庚烷、双环[2.2.2]辛烷、双环[3.2.2]壬烷、双环[3.3.1]壬烷和双环[4.2.1]壬烷基。螺环烷基可以为7-12元螺环烷基如7-11元螺环烷基,其实例包括但不限于:
Figure PCTCN2019076129-appb-000088
基。桥环烷基可以是6-10元桥环烷基如7-10元桥环烷基,其实例包括但不限于:
Figure PCTCN2019076129-appb-000089
基。
本发明所述的“3-12元环烯基”,是指在上述3-12元环烷基的基团中具有至少一个双键而得的基团,如环丁烯基、环戊烯基、环己烯基、环庚烯基、环辛烯基等。因此,本发明所述的“3-12元环烯基”,在不特别指明的情况下,包括可能形成的所有单环、稠环(包括以并、螺、桥的形式稠合)的情形,例如3-8元环烯基、7-11元螺环烯基、7-11元并环烯基、6-11元桥环烯基等。螺环烯基可以为7-12元螺环烯基如7-11元螺环烯基,其实例包括但不限于:
Figure PCTCN2019076129-appb-000090
基。桥环烯基可以是6-10元桥环烯基如7-10元桥环烯基,其实例包括但不限于:
Figure PCTCN2019076129-appb-000091
基。
本发明所述的“3-12元杂环基”,是指从3-12元杂环烷烃衍生得到的一价基团或(根据需要的)二价基团,即3-12元的至少一个环碳原子被选自O、S、S(O)、S(O) 2、C(O)、N的杂原子替代的非芳香性的环状基团,其优选含有1-3个杂原子。“3-12元杂环基”包括单环杂环基、双环杂环基系统或多环杂环基系统,其中一个或多个环可以是饱和或部分饱和的,但不包括芳环。在不特别指明的情况下,包括可能形成的所有单环、稠环(包括以并、螺、桥的形式稠合)、饱和和部分饱和的情形。
单环杂环基可以为3-8元杂环基如3-6元杂环基、4-7元杂环基或 5-6元杂环基,3-8元含氮杂环基如4-7元含氮杂环基或5-6元含氮杂环基,3-8元饱和杂环基如5-6元饱和杂环基等。其实例包括但不限于氮杂环丙烷基、氧杂环丙烷基、硫杂环丙烷基、氮杂环丁烷基、氧杂环丁烷基、硫杂环丁烷基、四氢呋喃基、四氢吡咯基、四氢噻吩基、咪唑烷基、吡唑烷基、1,2-噁唑烷基、1,3-噁唑烷基、1,2-噻唑烷基、1,3-噻唑烷基、四氢-2H-吡喃基、四氢-2H-噻喃基、哌啶基、哌嗪基、吗啉基、1,4-二氧杂环己烷基、1,4-氧硫杂环己烷基,4,5-二氢异噁唑基、4,5-二氢噁唑基、2,5-二氢噁唑基、2,3-二氢噁唑基、3,4-二氢-2H-吡咯基、2,3-二氢-1H-吡咯基、2,5-二氢-1H-咪唑基、4,5-二氢-1H-咪唑基、4,5-二氢-1H-吡唑基、4,5-二氢-3H-吡唑基、4,5-二氢噻唑基、2,5-二氢噻唑基、2H-吡喃基、4H-吡喃基、2H-噻喃基、4H-噻喃基、2,3,4,5-四氢吡啶基、1,2-异噁嗪基、1,4-异噁嗪基或6H-1,3-噁嗪基等。
稠杂环基包括并杂环基、螺杂环基和桥杂环基,其可以是饱和的、部分饱和的或不饱和的,但不是芳香性的。稠杂环基可以是稠合到苯环、5-6元单环环烷基、5-6元单环环烯基、5-6元单环杂环基或5-6元单环杂芳基的5-6元单环杂环基环。
所述的并杂环基可以为6-12元并杂环基如6-11元并杂环基或7-10元并杂环基,6-11元饱和并杂环基,6-11元含氮并杂环基,其代表性实例包括但不限于:3-氮杂双环[3.1.0]己烷基、3,6-二氮杂双环[3.2.0]庚烷基、3,8-二氮杂双环[4.2.0]辛烷基、3,7-二氮杂双环[4.2.0]辛烷基、八氢吡咯并[3,4-c]吡咯基、八氢吡咯并[3,4-b]吡咯基、八氢吡咯并[3,4-b][1,4]噁嗪基、八氢-1H-吡咯并[3,4-c]吡啶基、2,3-二氢苯并呋喃-2-基、2,3-二氢苯并呋喃-3-基、二氢吲哚-1-基、二氢吲哚-2-基、二氢吲哚3-基、2,3-二氢苯并噻吩-2基、八氢-1H-吲哚基、八氢苯并呋喃基。
所述的螺杂环基可以为6-12元螺杂环基如7-12元螺杂环基、7-12元饱和螺杂环基、7-12元含氮螺杂环基,其实例包括但不限于:
Figure PCTCN2019076129-appb-000092
所述的桥杂环基可以为6-12元桥杂环基如6-10元桥杂环基、7-10元桥杂环基,其实例包括但不限于:
Figure PCTCN2019076129-appb-000093
Figure PCTCN2019076129-appb-000094
本发明所述“芳基”,是指含有6-14个碳原子的从芳香性的碳环烃衍生得到的一价或根据需要的二价环状芳香性基团,包括,苯基、萘基、菲基等。
本发明所述“5-10元杂芳基”是指至少一个环碳原子被选自O、S、N的杂原子替代的芳香性的5-10元环状基团,优选含有1-3个杂原子,同时包括碳原子、硫原子被氧代、氮代的情况,例如碳原子被C(O)替代,硫原子被S(O)、S(O) 2替代。杂芳基包括单杂芳基和稠杂芳基,在不特别指明的情况下,某元杂芳基,包括可能形成的所有单环、稠环、全部芳香、部分芳香的情形。单杂芳基可以为5-7元杂芳基如5-6元杂芳基,其实例包括但不仅限于呋喃基、咪唑基、异噁唑基、噻唑基、异噻唑基、噁二唑基、噁唑基、吡啶基、哒嗪基、嘧啶基、吡嗪基、吡唑基、吡咯基、四唑基、噻二唑基、噻吩基、三唑基和三嗪基。在某些实施方案中,稠杂芳基是指单环杂芳环稠合到苯基、环烯基、杂芳基、环烷基或杂环基所形成的基团。在某些实施方案中,稠杂芳基可以为8-12元并杂芳基如9-10元并杂芳基,例子包括但不限于苯并咪唑基、苯并呋喃基、苯并噻吩基、苯并噁二唑基、苯并噻二唑基、苯并噻唑基、噌啉基、5,6-二氢喹啉-2-基、5,6-二氢异喹啉-1-基、呋喃并吡啶基、吲唑基、吲哚基、异吲哚基、异喹啉基、萘啶基、嘌呤基、喹啉基、5,6,7,8-四氢喹啉-2-基、5,6,7,8-四氢喹啉基、5,6,7,8-四氢喹啉-4-基、5,6,7,8-四氢异喹啉-1-基、噻吩并吡啶基、4,5,6,7-四氢并[c][1,2,5]噁二唑基和6,7-二氢并[c][1,2,5]噁二唑-4(5H)酮基。
本发明所述的“药学上可接受的盐”是指药学上可接受的酸或碱的加成盐或其溶剂化物。这样的药学上可接受的盐包括诸如以下的酸的加成盐:盐酸、磷酸、氢溴酸、硫酸、亚硫酸、甲酸、甲苯磺酸、甲磺酸、 硝酸、苯甲酸、柠檬酸、酒石酸、马来酸、氢碘酸和链烷酸(诸如乙酸、HOOC-(CH 2)n-COOH(其中n是0~4))等。这样的药学上可接受的盐还包括诸如以下的碱的盐:钠盐、钾盐、钙盐和铵盐等。本领域技术人员知晓多种无毒的药学上可接受的加成盐。
立体异构体是指由分子中原子在空间上排列方式不同所产生的异构体。当化合物存在不对称碳原子时,会产生对映异构体;当化合物存在碳碳双键或环状结构时,会产生顺反异构体;所有式(I)化合物的对映异构体、非对映异构体、外消旋体、顺反异构体、几何异构体、差向异构体及其混合物,均包括在本发明范围中。
“互变异构体”是指因分子中某一原子在两个位置迅速移动而产生的一种特殊的官能团异构体。其实例包括含有α-H的羰基化合物的互变异构,具体如
Figure PCTCN2019076129-appb-000095
(T、T 1、T 2分别独立地为任意符合化合物成键规律的基团)和其它其它质子迁移互变异构,具体如酚-酮互变异构、亚硝基-肟互变异构、亚胺-烯胺互变异构。
本发明中提及的所有数值范围,均表示包括该范围的两个端点、该范围之内的所有整数以及由这些整数形成的子范围。例如“3-12元”包括3、4、5、6、7、8、9、10、11或12元,以及“5-10元”、“10元”等等。
说明书中给出的本发明的通式化合物中所涉及的所有变量,包括X、Y、R 1、R 2、R 3、R 4、R 5、R 7、R 8、R 9、环B和m都是独立地选择的。说明书中所列举的这些变量的各种不同层次的定义之间可以相互组合,并且本发明包括由这些组合中的任意一种所构成的通式化合物的定义。
化合物制备
通过标准化学方法在内的多种方法,可以制备本发明的化合物。除非另外指出,否则任何前面定义的变量将继续具有前面定义的含义。示例性的一般合成方法阐述在下述方案中,并且可以很容易的改进以制备本发明的其它化合物。在实施例部分中制备本发明的具体化合物。
式(I)的化合物可以通过式(II)的化合物去保护来制备。下述提 供了由式(II)的化合物去保护来制备式(I)的化合物的方法:
Figure PCTCN2019076129-appb-000096
其中,R 1、R 2、R 3、R 4、R 5、R 7、R 8、R 9、环B、X、Y和m如上文中所定义,PG为胺的合适保护基,如叔丁氧羰基(Boc)或苄氧羰基(Cbz),此后,如果需要的话,制备如此形成的化合物的盐。
例如,向式(II)的化合物在合适溶剂(如二氯甲烷、乙醇、甲醇或乙腈)中的溶液中,加入合适的反应试剂(如三氟乙酸、氯化氢乙醇溶液、三甲基碘硅烷或三氟甲磺酸三甲基硅酯,某些实施方案加入苯酚),在合适的温度(0℃或环境温度)搅拌合适的时间段(如10分钟到10小时)。反应结束后,向反应液中加入合适的碱(如饱和碳酸氢钠水溶液或氨水)将反应混合物调节pH值至中性,或将反应混合物在减压下浓缩,粗品用水溶解后加入合适的碱(如饱和碳酸氢钠水溶液或氨水)调节pH值至中性,用合适的萃取剂(如二氯甲烷或乙酸乙酯)萃取,减压浓缩后,通过合适的纯化方法(如硅胶柱色谱、制备薄层色谱或反相柱色谱等)分离得到式(I)的化合物。
当Y为N时,式(II)的化合物可以通过式(III)的化合物
Figure PCTCN2019076129-appb-000097
其中R 1、R 2、R 3、R 7、R 8和R 9如上文中所定义,PG为胺的合适保护基,如叔丁氧羰基(Boc)或苄氧羰基(Cbz),与式(IV)的化合物反应来制备。
Figure PCTCN2019076129-appb-000098
其中R 4、环B、R 5和m如上文中所定义。
下面提供了由式(III)的化合物和式(IV)的化合物反应来制备式(II)的化合物的方法:
例如,将式(III)的化合物和式(IV)的化合物加入合适溶剂(如乙醇和水)中,加入合适的还原剂(如连二亚硫酸钠),在合适的温度(如90℃~100℃)加热或微波搅拌合适的时间段(如4~8小时)。反应结束后,将反应液倒入适量的水中,用合适的萃取剂(如乙酸乙酯)萃取,减压浓缩后,通过合适的纯化方法(如硅胶柱色谱、制备薄层色谱等)分离得到式(II)的化合物。
其中式(III)的化合物可由式(VII)的化合物
Figure PCTCN2019076129-appb-000099
其中R 1、R 2、R 3和R 7如上文中所定义,与式(VIII)的化合物反应来制备。
Figure PCTCN2019076129-appb-000100
其中R 8和R 9如上文中所定义,PG为胺的合适保护基,如叔丁氧羰基(Boc)或苄氧羰基(Cbz)。
下面提供了由式(VII)的化合物和式(VIII)的化合物反应来制备式(III)的化合物的方法:
例如,将式(VII)的化合物和式(VIII)的化合物加入合适的溶剂(如四氢呋喃、DMF)中,加入合适的肽偶联剂(如2-(7-氧化苯并三氮唑)-N,N,N′,N′-四甲基脲鎓六氟磷酸盐),再加入合适的碱(如N,N-二异丙基乙胺),在适当的温度(如环境温度)搅拌合适的时间段(如2~10小时)。将反应液减压浓缩,加入适量的水,用合适的萃取剂(如乙酸乙酯)萃取,将萃取液浓缩,粗品经常规的纯化方法(如硅胶柱色谱)分离得到式(III)的化合物。
在某些实施方案中,式(IV)的化合物可由式(IX)的化合物
Figure PCTCN2019076129-appb-000101
其中R 4、X、环B、R 5和m如上文中所定义,通过氧化制备。
下面提供了由式(IX)的化合物通过氧化来制备式(IV)的化合物的方法:
例如,将式(IX)的化合物加入合适的溶剂(如二氯甲烷)中,加入合适的氧化剂(如二氧化锰),在适当的温度(如回流)搅拌合适的时间段(如2小时)。将反应物过滤,滤饼用适量的溶剂淋洗,滤液减压浓缩,得到式(IV)的化合物。
其中,式(IX)的化合物可由式(X)的化合物
Figure PCTCN2019076129-appb-000102
其中R 4、X、环B、R 5和m如上文中所定义,R 10为甲基或乙基,通过还原制备。
下面提供了由式(X)的化合物通过还原来制备式(IX)的化合物的方法:
例如,将式(X)的化合物加入合适的溶剂(如四氢呋喃)中,加入合适的还原剂(如四氢铝锂或二异丁基氢化铝),在适当的温度(如环境温度)搅拌合适的时间段(如2~10小时)。加入合适的淬灭试剂(如水),过滤,加入干燥剂(如无水硫酸钠)干燥,滤液减压浓缩,得到式(IX)的化合物。
其中,式(X)的化合物可由式(XI)的化合物
Figure PCTCN2019076129-appb-000103
与R 4L通过烷基化反应制备,其中,R 4、X、环B、R 5、R 10和m如上文中所定义,L为离去基团(如卤素、三氟甲磺酸酯)。
下面提供了由式(XI)的化合物通过烷基化来制备式(X)的化合物的方法:
例如,将式(XI)的化合物和R 4L加入合适的溶剂(如乙腈)中,加入合适的碱(如碳酸钾或碳酸铯),在适当的温度(如环境温度至90℃)搅拌合适的时间段(如1~35小时)。反应完全后,过滤,滤液减压浓缩,粗品经常规的纯化方法(如硅胶柱色谱)分离得到式(X)的化合物。
在某些实施方案中,式(IV)的化合物可由式(XII)的化合物
Figure PCTCN2019076129-appb-000104
与R 4L通过烷基化反应制备,其中X、环B、R 5和m如上文中所定义,L为离去基团(如卤素、三氟甲磺酸酯)。
下面提供了由式(XII)的化合物通过烷基化来制备式(IV)的化合物的方法:
例如,将式(XII)的化合物和R 4L加入合适的溶剂(如DMF)中,加入合适的碱(如氢化钠或碳酸钾),在适当的温度(如-20℃~40℃)搅拌合适的时间段(如1~2小时)。反应完全后,加入水,用合适的萃取剂(如乙酸乙酯)萃取,萃取液减压浓缩,粗品经常规的纯化方法(如硅胶柱色谱)分离得到式(IV)的化合物。
当X为CR 6时,式(XI)的化合物可由以下方案来制备
Figure PCTCN2019076129-appb-000105
其中R 6、环B、R 5、R 10和m如上文中所定义。
当X为N时,式(IV)的化合物可由以下方案来制备
Figure PCTCN2019076129-appb-000106
其中R 4、环B、R 5和m如上文中所定义。
式(XII)的化合物可由以下方案来制备
Figure PCTCN2019076129-appb-000107
其中X、环B、R 5和m如上文中所定义。
具体实施方式
本文中使用的缩写,“DMF”是指二甲基甲酰胺;“THF”是指四氢呋喃;“EA”是指乙酸乙酯;“DCM”是指二氯甲烷;“TMEDA”是指四甲基乙二胺;“LDA”是指二异丙基氨基锂;“PE”是指石油醚;“DIPEA”是指N,N-二异丙基乙胺;“HATU”是指2-(7-氧化苯并三氮唑)-N,N,N′,N′-四甲基脲鎓六氟磷酸盐;“DMSO”是指二甲基亚砜。
实施例1:中间体3-甲氧基-4-(甲基氨基)-5-硝基苯甲酸甲酯的合成
Figure PCTCN2019076129-appb-000108
步骤1:4-羟基-3-甲氧基-5-硝基苯甲酸甲酯的合成
Figure PCTCN2019076129-appb-000109
将4-羟基-3-甲氧基苯甲酸甲酯(25.4g,0.139mol)溶于冰醋酸(100mL),冰浴下缓慢滴加硝酸(10.2mL)和醋酸(52mL)混合溶液,约1小时滴加完。反应液缓慢升至室温搅拌4小时。过滤,滤饼依次用水、正己烷和乙醚洗涤,干燥得到黄色固体状产物(21.0g,产率:66%)。
步骤2:4-氯-3-甲氧基-5-硝基苯甲酸甲酯的合成
Figure PCTCN2019076129-appb-000110
将4-羟基-3-甲氧基-5-硝基苯甲酸甲酯(21g,0.095mol)溶于DMF(200mL),冷却到-20℃,缓慢滴加草酰氯(23.7mL),滴完升温至80℃搅拌3小时。反应液冷却至室温,倒入冰水,搅拌过夜。过滤,滤饼溶于二氯甲烷,依次用饱和碳酸氢钠水溶液和食盐水洗涤,无水硫酸钠干燥,浓缩,粗品溶于乙酸乙酯,用食盐水洗涤,无水硫酸钠干燥,减压浓缩,粗品经甲醇重结晶得到目标产物(14.5g,产率:63.9%)。
步骤3:3-甲氧基-4-(甲基氨基)-5-硝基苯甲酸甲酯的合成
Figure PCTCN2019076129-appb-000111
将4-氯-3-甲氧基-5-硝基苯甲酸甲酯(14g,0.057mol)溶于DMF(140mL),冷却至0℃,氮气保护下,滴加甲胺溶液(2mol/L四氢呋喃溶液,114mL),加完后反应液加热至80℃搅拌3小时,降至室温,加水稀释,过滤得到橘色固体状产品(11.1g,产率81%)。
1HNMR(400MHz,DMSO-d 6)δ(ppm):8.07(d,J=1.6Hz,1H),7.80-7.76(m,1H),7.38(s,1H),3.91(s,3H),3.83(s,3H),2.94(t,J=5.6Hz,3H)。
实施例2:中间体1-乙基-1H-吡咯并[3,2-b]吡啶-2-甲醛的合成
Figure PCTCN2019076129-appb-000112
步骤1:1-(苯基磺酰基)-1H-吡咯并[3,2-b]吡啶的合成
Figure PCTCN2019076129-appb-000113
氮气保护下,室温下将氢化钠(质量分数60%,0.8g,20mmol,2.0eq)分批加入到无水THF(30mL)中,将1H-吡咯并[3,2-b]吡啶(1.19g,10mmol,1.0eq)溶于无水THF(10mL),缓慢滴加至体系中,滴毕,保温反应30分钟。将苯磺酰氯(3.5g,20mmol,2.0eq)溶于THF(10mL)中,缓慢滴加至体系中,15分钟滴加完毕,室温下反应4小时。LC-MS检测反应完全,缓慢滴加0.1mol/L HCl水溶液(20mL)淬灭反应,加水(30mL),EA萃取(50mL×2),有机相合并,食盐水洗,干燥,浓缩,粗品经硅胶柱色谱纯化得类白色固体状产物(1.5g,产率:57.7%)。
步骤2:1-(苯基磺酰基)-1H-吡咯并[3,2-b]吡啶-2-甲醛的合成
Figure PCTCN2019076129-appb-000114
氮气保护下,将二异丙胺(1.48g,14.5mmol,2.5eq)加入到无水THF(15mL)中,降温至-45℃,滴加正丁基锂(2.4mol/L,6mL,14.5mmol,2.5eq),滴毕保温30分钟,降温至-78℃,滴加1-(苯基磺酰基)-1H-吡咯并[3,2-b]吡啶(1.5g,5.8mmol,1.0eq)的THF(15mL)溶液,滴毕保温1小时,滴加DMF(0.85g,11.6mmol,2.0eq),滴毕保温30分钟,滴加饱和氯化铵水溶液(20mL)淬灭反应,EA(50mL×3)萃取,有机相合并,干燥,浓缩,粗品经硅胶柱色谱纯化得类白色固体状产物(1.2g,产率:72.3%)。
步骤3:1H-吡咯并[3,2-b]吡啶-2-甲醛的合成
Figure PCTCN2019076129-appb-000115
室温下将1-(苯基磺酰基)-1H-吡咯并[3,2-b]吡啶-2-甲醛(1.2g,4.1mmol,1.0eq)溶解在无水THF(15mL)中,降温至0℃,滴加氢氧化钠(1.67g,41mmol,10eq)的甲醇/水混合溶液(1∶1v/v,30mL)。滴加完毕后,缓慢恢复至室温反应1小时。LC-MS和TLC检测反应完全,滴加饱和氯化铵水溶液(20mL)淬灭反应,DCM萃取(50mL×2),有机相合并,食盐水洗,干燥,浓缩,粗品经硅胶柱色谱纯化得黄色固体状产物(300mg,产率:48.9%)。
步骤4:1-乙基-1H-吡咯并[3,2-b]吡啶-2-甲醛的合成
Figure PCTCN2019076129-appb-000116
室温下将1H-吡咯并[3,2-b]吡啶-2-甲醛(294mg,2mmol,1.0eq)溶解在无水DMF(5mL)中,加入碳酸钾(552.8mg,4mmol,2.0eq)和碘乙烷(375mg,2.4mmol,1.2eq),加热至40℃反应2小时。LC-MS和TLC检测反应完全,倒入冰水(10mL)中,EA萃取(20mL×2),食盐水洗,干燥,浓缩,粗品经硅胶柱色谱纯化得白色固体状产物(150mg,产率:42.8%)。
1HNMR(300MHz,DMSO-d 6)δ(ppm):10.01(s,1H),8.54(d,J=4.2Hz,1H),8.14(d,J=8.1Hz,1H),7.58(s,1H),7.42-7.37(m,1H),4.58(q,J=7.2Hz,2H),1.27(t,J=6.9Hz,3H).
LC-MS:175.1.[M+H] +
实施例3:中间体5-乙基-5H-吡咯并[2,3-b]吡嗪-6-甲醛的合成
Figure PCTCN2019076129-appb-000117
步骤1:3-((三甲基甲硅烷基)乙炔基)吡嗪-2-胺的合成
Figure PCTCN2019076129-appb-000118
氮气保护下,将3-氯吡嗪-2-胺(12.9g,0.1mol,1.0eq)溶于DMF(150mL)中,依次加入CuI(9.5g,0.05mol,0.5eq)、四(三苯基膦)钯(11.55g,0.01mol,0.1eq)和三乙胺(30.3g,0.3mol,3.0eq),降温至0℃,缓慢滴加三甲基硅乙炔(14.7g,0.15mol,1.5eq),滴加完毕后加热至70℃搅拌反应12小时。TLC和LC-MS检测反应完全,将反应液倒入水(300mL)中,加入EA(500mL),抽滤,滤饼用EA洗(50mL×2),滤液分液,有 机相用饱和食盐水洗,干燥,抽滤,滤液浓缩,粗品经硅胶柱色谱纯化得灰白色固体状产物(9.2g,收率:48%)。
步骤2:5H-吡咯并[2,3-b]吡嗪的合成
Figure PCTCN2019076129-appb-000119
氮气保护下,将3-((三甲基甲硅烷基)乙炔基)吡嗪-2-胺(9.55g,0.05mol,1.0eq)溶于THF(100mL)中,加入叔丁醇钾(11.2g,0.1mol,2.0eq),加热至回流,搅拌反应8小时。TLC和LC-MS检测反应完全,降至室温,加水(100mL),EA萃取(200mL×3),有机相合并,干燥,浓缩,粗品经硅胶柱色谱纯化得类白色固体状产物(3.5g,收率:59%)。
步骤3:5-(苯基磺酰基)-5H-吡咯并[2,3-b]吡嗪的合成
Figure PCTCN2019076129-appb-000120
氮气保护下,室温下将氢化钠(质量分数60%,0.8g,20mmol,2.0eq)分批加入到无水THF(30mL)中,将5H-吡咯并[2,3-b]吡嗪(1.19g,10mmol,1.0eq)溶于无水THF(10mL),缓慢滴加至体系中,滴完室温反应30分钟。将苯磺酰氯(3.5g,20mmol,2.0eq)溶于THF(10mL)中,缓慢滴加至体系中,20分钟滴加完毕,室温下反应4小时。LC-MS检测反应完全,缓慢滴加0.1mol/L HCl水溶液(20mL)淬灭反应,加水(30mL),EA萃取(50mL×2),有机相合并,食盐水洗,干燥,浓缩,粗品经硅胶柱色谱纯化得类白色固体状产物(1.5g,产率:58%)。
步骤4:5-(苯基磺酰基)-5H-吡咯并[2,3-b]吡嗪-6-甲醛的合成
Figure PCTCN2019076129-appb-000121
氮气保护下,将二异丙胺(1.48g,14.5mmol,2.5eq)加入到无水THF(15mL)中,降温至-45℃,滴加正丁基锂(2.4mol/L,6mL,14.5mmol,2.5eq),滴毕保温30分钟,降温至-78℃,滴加5-(苯基磺酰基)-5H-吡咯并[2,3-b]吡嗪(1.5g,5.8mmol,1.0eq)的THF(15mL)溶液,滴毕保温1小时,滴加DMF(0.85g,11.6mmol,2.0eq),滴毕保温30分钟,滴加饱和氯化铵水溶液(20mL)淬灭反应,EA(50mL×3)萃取,有机相合并,干燥,浓缩,粗品经硅胶柱色谱纯化得类白色固体状产物(1.2g,产率:73%)。
步骤5:5H-吡咯并[2,3-b]吡嗪-6-甲醛的合成
Figure PCTCN2019076129-appb-000122
室温下将5-(苯基磺酰基)-5H-吡咯并[2,3-b]吡嗪-6-甲醛(1.2g,4.1mmol,1.0eq)溶解在无水THF(15mL)中,降温至0℃,滴加氢氧化钠(1.67g,41mmol,10eq)的甲醇/水混合溶液(1∶1v/v,30mL)。滴加完毕后,缓慢升至室温反应1小时。LC-MS和TLC检测反应完全,滴加饱和氯化铵水溶液(20mL)淬灭反应,DCM萃取(50mL×2),有机相合并,食盐水洗,干燥,浓缩,粗品经硅胶柱色谱纯化得黄色固体状产物(300mg,产率:49%)。
步骤6:5-乙基-5H-吡咯并[2,3-b]吡嗪-6-甲醛的合成
Figure PCTCN2019076129-appb-000123
室温下将5H-吡咯并[2,3-b]吡嗪-6-甲醛(294mg,2mmol,1.0eq)溶解在无水DMF(5mL)中,加入碳酸钾(552.8mg,4mmol,2.0eq)和碘乙烷(375mg,2.4mmol,1.2eq),加热至40℃反应2小时。LC-MS和TLC检测反应完全,倒入冰水(10mL)中,EA萃取(20mL×2),食盐水洗,干燥, 浓缩,粗品经硅胶柱色谱纯化得白色固体状产物(150mg,产率:43%)。
1HNMR(300MHz,DMSO-d 6)δ(ppm):10.00(s,1H),8.53(d,J=1.2Hz,1H),8.41(s,1H),7.37(s,1H),6.25(d,J=2.7Hz,1H),4.69(q,J=7.2Hz,2H),1.35(t,J=6.9Hz,3H).
LC-MS:176.1[M+H] +.
实施例4:中间体1-乙基-1H-吡咯并[2,3-b]吡啶-2-甲醛的合成
Figure PCTCN2019076129-appb-000124
步骤1:1-(苯基磺酰基)-1H-吡咯并[2,3-b]吡啶的合成
Figure PCTCN2019076129-appb-000125
将1H-吡咯并[2,3-b]吡啶(10g,85mmol,1.0eq)溶于无水THF(150mL),冰浴下将氢化钠(质量分数60%,5g,125mmol,1.5eq)分批加入,反应30分钟。缓慢滴加苯磺酰氯,滴加完毕,升至室温反应2小时。TLC检测反应完全,加水(100mL),EA萃取,有机相合并,食盐水洗,干燥,浓缩,粗品经硅胶柱色谱纯化得产物(14g,产率:65%)。
步骤2:1-(苯基磺酰基)-1H-吡咯并[2,3-b]吡啶-2-甲醛的合成
Figure PCTCN2019076129-appb-000126
氮气保护下,将二异丙胺(4.13mL,0.029mol)加入到无水THF(50mL)中,降温至-78℃,滴加正丁基锂(2.4mol/L,10.42mL,0.026mol),升至室温反应1小时。将1-(苯基磺酰基)-1H-吡咯并[2,3-b]吡啶(5g,0.019mol)和TMEDA(4.38mL)溶于无水THF(50mL)溶液,降温至-30℃,氮气保护下,滴加上面制备的LDA溶液,滴毕反应2.5小时,缓慢滴加DMF(3mL),滴毕反应2小时。TLC和LCMS监控反应完全,加入饱和氯化铵水溶液(100mL)和EA(100mL),分液,有机相用无水硫酸钠干燥,浓缩,粗品经硅胶柱色谱纯化得黄色固体状产物(4g,产率:72%)。
步骤3:1H-吡咯并[2,3-b]吡啶-2-甲醛的合成
Figure PCTCN2019076129-appb-000127
将1-(苯基磺酰基)-1H-吡咯并[2,3-b]吡啶-2-甲醛(2.5g,8.73mmol)溶解在甲醇(50mL)中,氮气保护下,滴加氢氧化钾(1.46g,26mmol)的水溶液(5mL),搅拌反应0.5小时。TLC检测反应完全,加入水(150mL),DCM萃取,有机相合并,食盐水洗,无水硫酸钠干燥,减压浓缩,粗品经硅胶柱色谱纯化得黄色固体状产物(1.0g,产率:55%)。
步骤4:1-乙基-1H-吡咯并[2,3-b]吡啶-2-甲醛的合成
Figure PCTCN2019076129-appb-000128
室温下将1H-吡咯并[2,3-b]吡啶-2-甲醛(1.0g,6.8mmol)和碳酸钾(2.8g,20mmol)溶解在DMF(20mL)中,氮气保护下,-20℃滴加碘乙烷(1.2mL),反应液缓慢升至室温反应1小时。TLC检测反应完全,加入水(50mL),DCM萃取(50mL×3),有机相合并,食盐水洗,无水硫酸钠干燥,浓缩,粗品经硅胶柱色谱纯化得白色固体状产物(380mg,产率:32%)。
1HNMR(400MHz,DMSO-d 6)δ(ppm):9.96(s,1H),8.54(dd,J1=4.8Hz, J2=1.6Hz,1H),8.24(dd,J1=8.0Hz,J2=1.6Hz,1H),7.49(s,1H),7.26-7.23(m,1H),4.64(q,J=7.2Hz,2H),1.28(t,J=7.2Hz,3H).
实施例5:中间体6-乙基-6H-呋喃并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000129
步骤1:2-叠氮基乙酸乙酯的合成
Figure PCTCN2019076129-appb-000130
将2-溴乙酸乙酯(200g,1.2mol,1.0eq)加入到丙酮(1250mL)中,降温至0℃,滴加叠氮化钠(194.5g,3.0mol,2.5eq)的水(1000mL)溶液。滴毕加热至回流反应1h,冷却至室温,DCM萃取(1000mL×3),有机相合并,用饱和碳酸氢钠水溶液洗(800mL×2),水洗(800mL×2),干燥,低温减压浓缩得到产品(145g,产率:93%),直接用于下一步反应。
步骤2:6H-呋喃并[2,3-b]吡咯-5-甲酸乙酯的合成
Figure PCTCN2019076129-appb-000131
氮气保护下,将钠屑(7.64g,0.33mol,4.0eq)加入到无水乙醇(400mL)中,搅拌至反应完全,降温至0℃,滴加2-叠氮基乙酸乙酯(43g,0.33mol,4.0eq)和呋喃-3-甲醛(8g,0.083mol,1.0eq)的乙醇(40mL)溶液。滴毕0℃下反应1h,然后升至室温反应1h。TLC检测反应完全,滴加饱和氯化铵水溶液(200mL)淬灭反应,甲基叔丁基醚萃取(300mL×3),有 机相合并,饱和食盐水洗(300mL×3),干燥,浓缩得到粗品。将所得粗品加入到二甲苯(100mL)中,加热至回流搅拌反应4h。TLC和LC-MS检测反应完全,浓缩,粗品经硅胶柱色谱纯化得黄色固体状产物(5.14g,收率:33%)。
步骤3:6-乙基-6H-呋喃并[2,3-b]吡咯-5-甲酸乙酯的合成
Figure PCTCN2019076129-appb-000132
氮气保护下,将氢化钠(质量分数60%,1.5g,60.2mmol,2.0eq)加入到DMF(126mL)中,然后分批加入6H-呋喃并[2,3-b]吡咯-5-甲酸乙酯(5.4g,30.1mmol,1.0eq)。加完室温搅拌20分钟,滴加碘乙烷(14.1g,90.3mmol,3.0eq)。滴毕室温搅拌30分钟,TLC检测反应完全,降温至0℃,滴加饱和氯化铵水溶液(100mL)淬灭反应,甲基叔丁基醚萃取(100mL×3),有机相合并,饱和食盐水洗涤(100mL×3),干燥,浓缩得到黄色油状产物(5.4g粗品),不经纯化直接用于下一步。
步骤4:(6-乙基-6H-呋喃并[2,3-b]吡咯-5-基)甲醇的合成
Figure PCTCN2019076129-appb-000133
氮气保护下,将氢化铝锂(0.83g,21.9mmol,1.5eq)加入到THF(30mL)中,0℃滴加6-乙基-6H-呋喃并[2,3-b]吡咯-5-甲酸乙酯(3.01g,14.5mmol,1.0eq)的THF(30mL)溶液,滴完0℃搅拌反应2h。TLC检测反应完全,0℃下滴加水(1.04mL)、15w/w%的氢氧化钠水溶液(1.04mL)和水(3.12mL),加入无水硫酸钠搅拌15分钟,抽滤,滤液浓缩得到产物(2.4g粗品),直接用于下一步反应。
步骤5:6-乙基-6H-呋喃并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000134
将(6-乙基-6H-呋喃并[2,3-b]吡咯-5-基)甲醇(2.4g粗品)溶于 DCM(120mL)中,加入二氧化锰(12.63g,10eq),加热至回流反应1h。HPLC检测反应完全,加入硅藻土,抽滤,滤饼用DCM(20mL×3)洗,滤液浓缩得到产品(1.2g)。
1HNMR(300MHz,CDCl 3)δ(ppm):9.48(s,1H),7.34(s,1H),6.79(s,1H),6.56(s,1H),4.51(q,J=7.2Hz,2H),1.47(t,J=7.2Hz,3H).
LC-MS:164.1[M+H] +.
实施例6:中间体6-乙基-6H-噻吩并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000135
步骤:
Figure PCTCN2019076129-appb-000136
步骤1:6H-呋喃并[2,3-b]吡咯-5-甲酸乙酯的合成
Figure PCTCN2019076129-appb-000137
氮气保护下,将钠屑(7.64g,0.33mol,4.0eq)加入到无水乙醇(400mL)中,搅拌至反应完全,降温至0℃,滴加2-叠氮基乙酸乙酯(43g,0.33mol,4.6eq)和噻吩-3-甲醛(8g,0.071mol,1.0eq)的乙醇(40mL)溶液。滴毕0℃下反应1h,然后升至室温反应1h。TLC检测反应完全,滴加饱和氯化铵水溶液(200mL)淬灭反应,甲基叔丁基醚萃取(300mL×3),有机相合并,饱和食盐水洗(300mL×3),干燥,浓缩得到粗品。将所得粗品加入到二甲苯(100mL)中,加热至回流搅拌反应4h。TLC和LC-MS 检测反应完全,浓缩,粗品经硅胶柱色谱纯化得黄色固体状产物(2.0g,收率:15%)。
步骤2:6-乙基-6H-噻吩并[2,3-b]吡咯-5-甲酸乙酯的合成
Figure PCTCN2019076129-appb-000138
氮气保护下,将氢化钠(质量分数60%,0.83g,20.6mmol,2.0eq)加入到DMF(47mL)中,然后分批加入6H-噻吩并[2,3-b]吡咯-5-甲酸乙酯(2.0g,10.3mmol,1.0eq)。加完室温搅拌20分钟,滴加碘乙烷(4.8g,30.9mmol,3.0eq)。滴毕室温搅拌30分钟,TLC检测反应完全,降温至0℃,滴加饱和氯化铵水溶液(50mL)淬灭反应,甲基叔丁基醚萃取(50mL×3),有机相合并,饱和食盐水洗涤(50mL×3),干燥,浓缩得到黄色油状产物(2.0g粗品),不经纯化直接用于下一步。
步骤3:(6-乙基-6H-呋喃并[2,3-b]吡咯-5-基)甲醇的合成
Figure PCTCN2019076129-appb-000139
氮气保护下,将6-乙基-6H-噻吩并[2,3-b]吡咯-5-甲酸乙酯(2.0g,8.96mmol,1.0eq)加入到THF(20mL)中,降温至0℃,滴加二异丁基氢化铝(2M的甲苯溶液,16.2mL,22.4mmol,2.5eq),滴毕0℃搅拌反应2h。TLC检测反应全,0℃下依次滴加水(1mL)、15w/w%的氢氧化钠水溶液(1mL)和水(2.4mL)。滴完加入无水硫酸钠,搅拌15分钟,抽滤,固体用乙酸乙酯洗,滤液浓缩得到粗品(1.68g),直接用于下一步反应。
步骤4:6-乙基-6H-噻吩并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000140
将(6-乙基-6H-呋喃并[2,3-b]吡咯-5-基)甲醇(1.68g粗品)溶于DCM(84mL)中,加入二氧化锰(8.06g,10eq),加热至回流反应1h。HPLC检测反应完全,加入硅藻土,抽滤,滤饼用DCM(20mL×3)洗, 滤液浓缩得到产品(1.2g)。
1HNMR(300MHz,CDCl 3)δ(ppm):9.56(s,1H),7.04(d,J=7.2Hz,1H),6.96(d,J=7.2Hz,1H),4.52(q,J=7.2Hz,2H),1.46(t,J=7.2Hz,3H).
LC-MS:180.1[M+H] +.
实施例7:中间体6-乙基-2-甲基-6H-噻吩并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000141
步骤1:2-甲基噻吩-4-甲醛的合成
Figure PCTCN2019076129-appb-000142
将4-溴-2-甲基噻吩(25g,141.2mmol,1.0eq)加入到乙醚(170mL)中,然后降温至-78℃,滴加正丁基锂(2.5mol/L的正己烷溶液,84.72mL,211.8mmol,1.5eq)。滴毕,保温反应1h,滴加DMF(15.48g,211.8mmol,1.5eq)的乙醚(56mL)溶液。滴毕,保温反应1h。HPLC检测反应完毕后,-78℃下滴加饱和氯化铵(300mL)淬灭反应,甲基叔丁基醚萃取(200mL×3),有机相饱和食盐水洗(300mL×2),无水硫酸钠干燥,过滤,浓缩得粗品(23.28g),用油泵减压蒸馏得产品(10.9g,收率:61%)。
步骤2:2-甲基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯的合成
Figure PCTCN2019076129-appb-000143
氮气保护下,将钠屑(5g,217.4mmol,5.5eq)加入到无水乙醇(250mL)中,搅拌至溶解,滴加叠氮乙酸乙酯(20.47g,158.50mmol,4.0eq)和2-甲基噻吩-4-甲醛(5g,39.63mmol,1.0eq)的乙醇(25mL)溶液。滴毕,0℃下搅拌反应1h,然后升至室温搅拌反应1h。TLC检测反应完毕,滴加饱和氯化铵水溶液(300mL)淬灭反应,甲基叔丁基醚萃取(300mL×3),有机相合并,饱和食盐水洗(300mL×3),无水硫酸钠干燥,过滤,减压浓缩得到粗品,加入至甲基叔丁基醚(30mL)中,饱和食盐水洗 (30mL×3),干燥,减压浓缩,将得到的中间体加入到二甲苯(65mL)中,加热至回流反应2h。TLC、LC-MS检测反应完毕,浓缩,粗品经硅胶柱色谱纯化得到产品(4.1g,收率:49%)。
步骤3:6-乙基-2-甲基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯的合成
Figure PCTCN2019076129-appb-000144
氮气保护下,将氢化钠(0.94g,39.19mmol,2.0eq)加入到DMF(96mL)中,分批加入2-甲基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(4.1g,19.59mmol,1.0eq)。加毕,室温搅拌20分钟,滴加碘乙烷(9.17g,58.78mmol,3.0eq),滴毕,室温搅拌反应30分钟,TLC检测反应完毕,降温至0℃,滴加饱和氯化铵水溶液(50mL)淬灭反应,甲基叔丁基醚萃取(50mL×3),有机相合并,用饱和食盐水洗涤(100mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩得产品(4.25g粗品)。
步骤4:(6-乙基-2-甲基-6H-噻吩并[2,3-b]吡咯-5-基)甲醇的合成
Figure PCTCN2019076129-appb-000145
氮气保护下,6-乙基-2-甲基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(4.25g,17.91mmol,1.0eq)加入到THF(45mL)中,降温至0℃,滴加二异丁基氢化铝(1.5mol/L的甲苯溶液,29.85mL,44.77mmol,2.5eq)。滴毕,0℃搅拌反应2h。TLC检测反应完毕,0℃下依次滴加水(1.8mL)、15w/w%的氢氧化钠水溶液(1.8mL)和水(4.5mL)。滴毕,加入无水硫酸钠,搅拌15分钟,抽滤,滤饼用乙酸乙酯洗,滤液浓缩得到粗品(3.25g),用PE(30mL)浆洗,抽滤得产品(2.3g,收率:65.9%)。
步骤5:6-乙基-2-甲基-6H-噻吩并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000146
将(6-乙基-2-甲基-6H-噻吩并[2,3-b]吡咯-5-基)甲醇(2.3g,11.78mmol,1.0eq)溶于DCM(115mL)中,加入二氧化锰(10.24g,117.78mmol,10eq),加热至回流反应2h。HPLC检测反应完毕,加硅藻土抽滤,固体用DCM(50mL×3)洗,滤液减压浓缩得到产品(1.95g,收率:85%)。
1H NMR(400MHz,CDCl 3)δ(ppm):9.49(s,1H),6.92(s,1H),6.67(d,J=1.2Hz,1H),4.49(q,J=7.2Hz,2H),2.52(d,J=1.2Hz,3H),1.42(t,J=7.2Hz,3H).
分子式:C 10H 11NOS  分子量:193.26  LC-MS(Pos,m/z)=194.1[M+H] +
实施例8:中间体6-乙基-3-甲基-6H-噻吩并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000147
步骤1:4-甲基噻吩-3-甲醛的合成
Figure PCTCN2019076129-appb-000148
将3-溴-4-甲基噻吩(30g,169.43mmol,1.0eq)加入到乙醚(200mL)中,降温至-78℃,滴加正丁基锂(2.5mol/L正己烷溶液,102mL,254.15mmol,1.5eq)。滴毕,保温反应1h,滴加DMF(18.58g,254.15mmol,1.5eq)的乙醚(67mL)溶液,滴毕,保温反应1h,HPLC检测反应完毕,-78℃下滴加饱和氯化铵水溶液(400mL)淬灭反应,甲基叔丁基醚萃取(300mL×3),有机相合并,饱和食盐水洗(300mL×2),无水硫酸钠干燥,减压浓缩得到粗品(28.5g),用油泵减压蒸馏得产品(9.4g,收率:44%)。
步骤2:3-甲基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯的合成
Figure PCTCN2019076129-appb-000149
氮气保护下,将钠屑(6.86g,297.99mmol,4.0eq)加入到无水乙醇 (470mL)中,搅拌至溶解,滴加叠氮乙酸乙酯(38.48g,297.99mmol,4.0eq)和4-甲基噻吩-3-甲醛(9.4g,74.50mmol,1.0eq)的乙醇(47mL)溶液。滴毕,0℃下搅拌反应1h,升至室温搅拌反应1h。TLC检测反应完毕,滴加饱和氯化铵水溶液(500mL)淬灭反应,甲基叔丁基醚萃取(500mL×3),合并有机相,饱和食盐水洗(500mL×3),无水硫酸钠干燥,减压浓缩得到粗品,加入至甲基叔丁基醚(100mL)中,饱和食盐水洗(50mL×3),无水硫酸钠干燥,减压浓缩,将中间体加入到二甲苯(120mL)中,加热至回流反应2h。TLC、LC-MS检测反应完毕,浓缩,粗品经硅胶柱色谱纯化得产品(7.7g,收率:49.3%)。
步骤3:6-乙基-3-甲基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯的合成
Figure PCTCN2019076129-appb-000150
氮气保护下,将氢化钠(1.77g,73.59mmol,2.0eq)加入DMF(180mL)中,然后分批加入3-甲基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(7.7g,36.80mmol,1.0eq)。加毕,室温搅拌20分钟,然后滴加碘乙烷(17.22g,110.39mmol,3.0eq),滴毕,室温搅拌30分钟,TLC检测反应完毕,降至0℃,滴加饱和氯化铵水溶液(50mL)淬灭反应,甲基叔丁基醚萃取(200mL×3),合并有机相,饱和食盐水洗涤(200mL×5),干燥,减压浓缩得到产品(9.8g,粗品)。
步骤4:(6-乙基-3-甲基-6H-噻吩并[2,3-b]吡咯-5-基)甲醇的合成
Figure PCTCN2019076129-appb-000151
氮气保护下,将6-乙基-3-甲基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(9.8g,41.29mmol,1.0eq)加入到THF(100mL)中,降温至0℃,滴加二异丁基氢化铝(1.5mol/L的甲苯溶液,69mL,103.24mmol,2.5eq)。滴毕,0℃搅拌反应2h。TLC检测反应完毕,0℃下依次滴加水(4.2mL)、15w/w%的氢氧化钠水溶液(4.2mL)和水(10.3mL)。滴毕,加入无水硫酸 钠,搅拌15分钟,抽滤,固体用乙酸乙酯洗,滤液减压浓缩得到粗品(13.1g),用PE(30mL)浆洗,抽滤得产品(6.1g,收率:75.6%)。
步骤5:6-乙基-3-甲基-6H-噻吩并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000152
将(6-乙基-3-甲基-6H-噻吩并[2,3-b]吡咯-5-基)甲醇(6.1g,31.24mmol,1.0eq)溶于DCM(300mL)中,加入二氧化锰(27.16g,312.37mmol,10eq),加热至回流反应2h。HPLC检测反应完毕,加硅藻土抽滤,滤饼用DCM(100mL×3)洗,滤液减压浓缩得到产品(4.5g,收率:74%)。
1HNMR(300MHz,CDCl 3)δ(ppm):δ9.52(s,1H),7.00(s,1H),6.52(s,1H),4.49(q,J=7.2Hz,2H),2.32(s,3H),1.45(t,J=7.2Hz,3H).
分子式:C 10H 11NOS  分子量:193.26  LC-MS(Pos,m/z)=194.1[M+H] +
实施例9:(R)-(3-氨基哌啶-1-基)(2-(5-乙基-5H-吡咯并[2,3-b]吡嗪-6-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物1)的合成
Figure PCTCN2019076129-appb-000153
步骤1:3-甲氧基-4-(甲基氨基)-5-硝基苯甲酸的合成
Figure PCTCN2019076129-appb-000154
将3-甲氧基-4-(甲基氨基)-5-硝基苯甲酸甲酯(1.0g,4.16mmol,1.0eq)溶于甲醇(5mL)和四氢呋喃(5mL)中,加入一水合氢氧化锂(500mg,11.9mmol,2.9eq)的水(5mL)溶液,50℃搅拌2h。TLC监测反应完全,减压浓缩除去大部分有机溶剂,用盐酸(1mol/L)调节pH值至5,过滤得棕黄色固体3-甲氧基-4-(甲基氨基)-5-硝基苯甲酸(600.0mg,收率:63.7%)。
步骤2:(R)-(1-(3-甲氧基-4-(甲基氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000155
将3-甲氧基-4-(甲基氨基)-5-硝基苯甲酸(500.0mg,2.21mmol,1.0eq)溶解于四氢呋喃(30mL)和N,N-二甲基甲酰胺(1mL)的混合溶剂中,冰浴下加入N,N-二异丙基乙胺(570.8mg,4.42mmol,2.0eq)和2-(7-氧化苯并三氮唑)-N,N,N′,N′-四甲基脲鎓六氟磷酸盐(923.9mg,2.43mmol,1.1eq),冰浴下搅拌40分钟,滴加(R)-哌啶-3-基氨基甲酸叔丁酯(530.7mg,2.65mmol,1.2eq)的四氢呋喃溶液(5mL),滴加完毕逐渐升至室温搅拌10小时。TLC显示反应完全,将体系浓缩,加入水(50mL)和乙酸乙酯(100mL),分液,有机相用无水硫酸钠干燥,浓缩,粗品经硅胶柱色谱纯化(200-300目硅胶,二氯甲烷∶甲醇=40∶1v/v)得到产物(960.3mg,收率:100%)。
步骤3:(R)-(1-(2-(5-乙基-5H-吡咯并[2,3-b]吡嗪-6-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000156
将(R)-(1-(3-甲氧基-4-(甲基氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(580.7mg,1.42mmol,1.0eq)溶于乙醇(7mL)中,加入5-乙基-5H-吡咯并[2,3-b]吡嗪-6-甲醛(250.0mg,1.42mmol,1.0eq)、连二亚硫酸钠(742.7mg,4.26mmol,3.0eq)和水(4mL),微波100℃反应5小时。TLC显示反应完全,减压浓缩,加入水(30mL)和二氯甲烷(70mL),分液,有机相用无水硫酸钠干燥,浓缩,粗品经硅胶柱色谱分离(200-300目硅胶,二氯甲烷∶甲醇=60∶1v/v)得到产物(513.7mg,收率:67.8%)。
步骤4:(R)-(3-氨基哌啶-1-基)(2-(5-乙基-5H-吡咯并[2,3-b]吡嗪-6-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000157
将(R)-(1-(2-(5-乙基-5H-吡咯并[2,3-b]吡嗪-6-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯(513.7mg,0.96mmol,1.0eq)溶解在二氯甲烷(4mL)中,滴加三氟乙酸(2mL),滴加完毕室温反应2h,TLC监测反应完全,冰浴下加入饱和碳酸氢钠水溶液(10mL)调节pH值至7~8,用二氯甲烷(15mL)萃取,有机相用无水硫酸钠干燥,粗品经制备薄层色谱纯化得到(R)-(3-氨基哌啶-1-基)(2-(5-乙基-5H-吡咯并[2,3-b]吡嗪-6-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(20.0mg,产率:4.8%)。
1HNMR(400MHz,DMSO-d 6)δ(ppm):8.57(d,J=2.4Hz,1H),8.45(d,J=2.4Hz,1H),7.37(s,1H),7.32(s,1H),6.91(s,1H),4.60-4.65(m,2H),4.17(s,3H),3.99(s,3H),2.97-3.05(m,2H),2.80-2.90(m,2H),1.98-2.02(m,1H),1.90-1.92(m,2H),1.65-1.80(m,2H),1.47-1.55(m,2H),1.25-1.40(m, 3H).
分子式:C 23H 27N 7O 2  分子量:433.52  LC-MS(Pos,m/z)=434.2[M+H] +.
实施例10:(R)-(3-氨基哌啶-1-基)(2-(1-乙基-1H-吡咯并[3,2-b]吡啶-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物2)的合成
Figure PCTCN2019076129-appb-000158
步骤1:(R)-(1-(2-(1-乙基-1H-吡咯并[3,2-b]吡啶-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000159
将(R)-(1-(3-甲氧基-4-(甲基氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(245.0mg,0.60mmol,1.5eq)溶于乙醇(10mL)中,加入1-乙基-1H-吡咯并[3,2-b]吡啶-2-甲醛(70.1mg,0.40mmol,1.0eq)、连二亚硫酸钠(208.9mg,1.20mmol,3.0eq)和水(2mL),微波100℃反应5小时。TLC显示反应完全,减压浓缩,加入水(15mL)和二氯甲烷(30mL),分液,有机相用无水硫酸钠干燥,浓缩,粗品经硅胶柱色谱分离(200-300目硅胶,二氯甲烷∶甲醇=50∶1v/v)得产品(213.6mg,收率:87.2%)。
步骤2:(R)-(3-氨基哌啶-1-基)(2-(1-乙基-1H-吡咯并[3,2-b]吡啶-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000160
将(R)-(1-(2-(1-乙基-1H-吡咯并[3,2-b]吡啶-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯(213.6mg,0.40mmol,1.0eq)溶解在乙醇(3mL)中,滴加氯化氢的乙醇溶液(30w/w%,3mL),滴加完毕室温反应10小时,TLC监测反应完全,浓缩,用饱和碳酸氢钠水溶液调节pH值至7~8,用二氯甲烷(15mL)萃取,有机相用无水硫酸钠干燥,浓缩,粗品经制备薄层色谱纯化得到产品(16.8mg,产率:9.7%)。
1HNMR(400MHz,DMSO-d 6)δ(ppm):8.46-8.47(m,1H),8.07-8.09(m,2H),7.34(s,1H),7.27-7.30(m,1H),7.18(s,1H),6.86-6.88(m,1H),4.52-4.56(m,2H),4.13(s,3H),3.98(s,3H),2.79-2.87(m,2H),1.97-2.00(m,2H),1.89-1.92(m,2H),1.65-1.75(m,2H),1.45-1.48(m,2H),1.20-1.31(m,3H).
分子式:C 24H 28N 6O 2  分子量:432.53  LC-MS(Pos,m/z)=433.1[M+H] +.
实施例11:(R)-(3-氨基哌啶-1-基)(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物3)的合成
Figure PCTCN2019076129-appb-000161
步骤1:(R)-(1-(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000162
将(R)-(1-(3-甲氧基-4-(甲基氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(469.7mg,1.150mmol,1.0eq)溶解于乙醇(16mL)中,加入6-乙基-6H-噻吩并[2,3-b]吡咯-5-甲醛(200.0mg,1.15mmol)、水(2mL)和连二亚硫酸钠(600.65mg,3.45mmol),微波100℃反应5小时。TLC检测反应完全,减压浓缩,加入水(10mL)和乙酸乙酯(20mL),分液,有机相浓缩,粗品经硅胶柱色谱(DCM∶MeOH=60∶1v/v)纯化得白色固体状产物(300.0mg,得率:48.6%)。
步骤2:(R)-(3-氨基哌啶-1-基)(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000163
将(R)-(1-(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯(150.0mg,0.28mmol,1.0eq)溶于二氯甲烷(4mL)中,冰浴下加入氯化氢乙醇溶液(30w/w%,2mL),室温反应2h。TLC监测反应完全,浓缩,用碳酸氢钠水溶液调节pH值至7~8,用乙酸乙酯萃取,有机相合并,无水硫酸钠干燥,过滤,浓缩,粗品经制备薄层色谱纯化(DCM∶MeOH=8∶1v/v)得产物(R)-(3-氨基哌啶-1-基)(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(15.0mg,产率:12.3%)。
1HNMR(400MHz,DMSO-d 6)δ(ppm):7.26(s,1H),7.17-7.16(d,1H),7.12-7.11(d,1H),6.90-6.86(d,1H),6.63(s,1H),4.40-4.38(d,2H),4.09(s,3H),3.96(s,3H),2.02-1.84(m,4H),1.47(s,2H),1.35-1.31(t,3H).
分子式:C 23H 27N 5O 2S  分子量:437.19  LC-MS(Pos, m/z)=438.1[M+H] +.
实施例12:(R)-(3-氨基哌啶-1-基)(2-(6-乙基-6H-呋喃并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物4)的合成
Figure PCTCN2019076129-appb-000164
步骤1:(R)-(1-(2-(6-乙基-6H-呋喃并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000165
将(R)-(1-(3-甲氧基-4-(甲基氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(600.7mg,1.47mmol,1.2eq)溶于乙醇(6mL)中,加入6-乙基-6H-呋喃并[2,3-b]吡咯-5-甲醛(200.0mg,1.22mmol,1.0eq)、连二亚硫酸钠(639.8mg,3.67mmol,3.0eq)和水(2mL),微波100℃反应5小时。TLC显示反应完全,减压浓缩,加入水(25mL)和二氯甲烷(60mL),分液,有机相用无水硫酸钠干燥,浓缩,粗品经硅胶柱色谱分离(200-300目硅胶,二氯甲烷∶甲醇=100∶1v/v)得到产物(60.3mg,收率:1.6%)。
步骤2:(R)-(3-氨基哌啶-1-基)(2-(6-乙基-6H-呋喃并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000166
将(R)-(1-(2-(6-乙基-6H-呋喃并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯(32.5mg,0.06mmol,1.0eq)溶解在二氯甲烷(4mL)中,冰浴下滴加2,6-二甲基吡啶(40.0mg,0.37mmol,6.0eq),滴加完毕冰浴下搅拌30分钟,滴加三氟甲磺酸三甲基硅酯(41.3mg,0.18mmol,2.0eq),冰浴下反应4h。TLC监测反应完全,加入饱和氯化铵水溶液(5mL),用二氯甲烷(15mL)萃取,有机相用无水硫酸钠干燥,浓缩,粗品经制备薄层色谱纯化得产品(R)-(3-氨基哌啶-1-基)(2-(6-乙基-6H-呋喃并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(1.8mg,产率:6.9%)。
1HNMR(400MHz,DMSO-d 6)δ(ppm):8.25(brs,3H),7.62(s,1H),7.33(s,1H),6.86(s,1H),6.72(s,1H),6.63(s,1H),4.40-4.42(m,2H),4.11(s,3H),3.97(s,3H),3.15-3.24(m,2H),1.98-2.07(m,1H),1.75-1.77(m,1H),1.49-1.65(m,2H),1.32-1.35(m,3H).
分子式:C 23H 27N 5O 3  分子量:421.50  LC-MS(Pos,m/z)=422.0[M+H] +.
实施例13:(R)-(3-氨基哌啶-1-基)(2-(3-乙基-3H-噻吩并[2,3-d]咪唑-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物8)的合成
Figure PCTCN2019076129-appb-000167
步骤1:(R)-(1-(3-氨基-5-甲氧基-4-(甲氨基)苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000168
将(R)-(1-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(500.0mg,1.224mmol,1.0eq)和连二亚硫酸钠(639.0mg,3.672 mmol,3.0eq)溶于乙醇(6.0ml)和水(3.0ml),氮气保护下,90℃反应过夜,TLC显示未反应完全,补加连二亚硫酸钠(211.0mg,1.224mmol,1.0eq),1h后TLC显示反应完全。将反应液减压浓缩,粗品经硅胶柱色谱(DCM∶MeOH=200∶1~60∶1v/v)纯化得产品(450.0mg,收率:97.2%)。
步骤2:(R)-(1-(2-(3-乙基-3H-噻吩并[2,3-d]咪唑-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000169
将(R)-(1-(3-氨基-5-甲氧基-4-(甲氨基)苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(250.0mg,0.660mmol,1.0eq)、3-乙基-3H-噻吩并[2,3-d]咪唑-2-甲醛(143.0mg,0.793mmol,1.2eq)和碳酸钾(274.0mg,1.982mmol,3.0eq)溶于1,4-二氧六环(3.0mL)和水(0.5mL),升温至110℃反应过夜,TLC显示反应完全。将反应液减压浓缩,加水(50.0mL),乙酸乙酯(20.0mL)萃取,干燥,减压浓缩,粗品经硅胶柱色谱纯化(DCM∶MeOH=100∶1~20∶1v/v)得产品(125.0mg,收率:35.1%)。
步骤3:(R)-(3-氨基哌啶-1-基)(2-(3-乙基-3H-噻吩并[2,3-d]咪唑-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000170
将(R)-(1-(2-(3-乙基-3H-噻吩并[2,3-d]咪唑-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯(125.0mg,0.232mmol,1.0eq)溶于二氯甲烷中,降温至0℃,加入三氟乙酸(1.5mL),2h后TLC显示反应完全。将反应液减压浓缩,加水(10.0mL),用氨水调pH值至9~10,乙酸乙酯(10.0mL×2)萃取,减压浓缩,粗品经硅胶柱色谱(DCM∶MeOH=60∶1~11∶1v/v)纯化得产品(90.0mg,收率:88.4%)。
1H NMR(400MHz,DMSO-d6)δ(ppm):7.44-7.42(m,1H),7.31-7.29(m,2H),6.87(s,1H),4.76-4.70(m,2H),4.38(s,3H),3.98(s,3H),2.93(m,1H),2.68-2.67(m,2H),1.89-1.85(m,2H),1.75-1.65(m,2H),1.50-1.46(m,3H),1.26-1.24(m,2H).
分子式:C 22H 26N 6O 2S  分子量:438.55  LC-MS(Pos,m/z)=439.36[M+H] +.
实施例14:(R)-(3-氨基哌啶-1-基)(2-(6-乙基-2-甲基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物9)的合成
Figure PCTCN2019076129-appb-000171
步骤1:3-甲氧基-4-(甲氨基)-5-硝基苯甲酸的合成
Figure PCTCN2019076129-appb-000172
将3-甲氧基-4-(甲氨基)-5-硝基苯甲酸甲酯(10.0g,41mmol,1.0eq)溶解于四氢呋喃(200mL)中,滴加一水合氢氧化锂(7.0g,166mmol,4.0eq)的水溶液(40mL),45℃搅拌1小时,TLC监测反应完全,减压浓缩,加水(200mL),冰浴下用2mol/L盐酸调节pH值至2~3,用二氯甲烷(150mL)萃取,有机相用无水硫酸钠干燥,减压浓缩得产品(8.1g,收率:87.9%)。
步骤2:(R)-(1-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000173
将3-甲氧基-4-(甲氨基)-5-硝基苯甲酸(2.23g,9.85mmol,1.0eq)加入到DMF(15mL)和四氢呋喃(60mL)混合溶液中,冰浴下加入DIPEA(2.54g,19.7mmol,2.0eq)和HATU(4.12g,10.83mmol,1.1eq),反应2小时,加入(R)-哌啶-3-基氨基甲酸叔丁酯(2.36g,11.83mmol,1.2eq),加毕,逐渐升至室温搅拌过夜,TLC监测反应完全,减压浓缩,加入乙酸乙酯(150mL)和水(200mL),分液,有机相用无水硫酸钠干燥,减压浓缩得产品(4.7g粗品)。
步骤3:(R)-(1-(2-(6-乙基-2-甲基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000174
将(R)-(1-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(721.6,1.51mmol,1.0eq.)加入到乙醇(9.0mL)中,加入6-乙基-2-甲基-6H-噻吩并[2,3-b]吡咯-5-甲醛(291.82mg,1.51mmol,1.0eq)和连二亚硫酸钠(788.67mg,4.53mmol,3.0eq)的水溶液(3.0mL),微波100℃反应4小时,TLC监测反应完全,减压浓缩,加入二氯甲烷(20mL)和水(15mL),分液,有机相用无水硫酸钠干燥,浓缩,粗品经硅胶柱色谱(二氯甲烷∶甲醇=80∶1v/v)纯化得产品(353.7mg,收率:42.4%)。
步骤4:(R)-(3-氨基哌啶-1-基)(2-(6-乙基-2-甲基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000175
将(R)-(1-(2-(6-乙基-2-甲基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯(300.0mg,0.54mmol,1.0eq)加入到二氯甲烷(6mL)中,冰浴下滴加入三氟乙酸(3 mL),升至室温搅拌2小时。TLC监测反应完全,冰浴下滴加饱和碳酸氢钠水溶液(10mL),调节pH值至7~8,加入二氯甲烷(15mL),萃取,有机相用无水硫酸钠干燥,减压浓缩得产品(177.9mg,收率:72.9%)。
1HNMR(400MHz,DMSO-d 6)δ(ppm):7.24(s,1H),6.81(m,2H),6.77(s,1H),4.33-4.38(m,2H),4.09(s,3H),3.96(s,3H),2.86-2.92(s,1H),2.67-2.69(s,3H),2.50-2.51(s,2H),1.85-1.87(m,1H),1.67(s,1H),1.43-1.49(m,3H),1.31-1.33(m,3H),1.24-1.29(m,1H).
分子式:C 24H 29N 5O 2S 分子量:451.59LC-MS(Pos,m/z)=452.9[M+H] +.
实施例15:(R)-(3-氨基哌啶-1-基)(2-(6-乙基-3-甲基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物10)的合成
Figure PCTCN2019076129-appb-000176
步骤1:(R)-(1-(2-(6-乙基-3-甲基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000177
将(R)-(1-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(347.0mg,0.84mmol,1.0eq)溶解于乙醇(5.0mL),加入6-乙基-3-甲基-6H-噻吩并[2,3-b]吡咯-5-甲醛(164.1mg,0.84mmol,1.0eq)和连二亚硫酸钠(438.7mg,2.52mmol,3.0eq)的水溶液(2.0mL),微波100℃反应5小时,TLC监测反应完全,减压浓缩,粗品经硅胶柱色谱(二氯甲烷∶甲醇=100∶1v/v)纯化得产物(170.0mg,收率:36.7%)。
步骤2:(R)-(3-氨基哌啶-1-基)(2-(6-乙基-3-甲基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000178
将(R)-(1-(2-(6-乙基-2-甲基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯(170.0mg,0.3mmol,1.0eq)溶解于二氯甲烷(3.0mL)中,冰浴下滴加三氟乙酸(1.5mL),升至室温搅拌1小时。TLC监测反应完全,冰浴下滴加饱和碳酸氢钠水溶液(5mL),调节体系pH值至7~8,加入二氯甲烷(10mL),分液,有机相用无水硫酸钠干燥,过滤,减压浓缩得到产品(75.1mg,收率:54.0%)。
1H NMR(400MHz,DMSO-d 6)δ(ppm):7.25(s,1H),6.87(s,1H),6.82(s,1H),6.74-6.75(s,1H),4.34-4.39(m,2H),4.11(s,3H),3.97(s,3H),2.92(s,1H),2.66(s,2H),2.32(s,3H),1.85-1.88(d,1H),1.59(m,3H),1.54(m,1H),1.29-1.32(m,3H).
分子式:C 24H 29N 5O 2S  分子量:451.59  LC-MS(Pos,m/z)=452.4[M+H] +.
实施例16:N-(3-氨基环丁基)-2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-N,1-二甲基-1H-苯并[d]咪唑-5-甲酰胺(化合物13)的合成
Figure PCTCN2019076129-appb-000179
步骤1:(3-((4-甲氧基苄基)(甲基)氨基)环丁基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000180
将(3-氧代环丁基)氨基甲酸叔丁酯(2.69g,14.5mmol,1.1eq)溶解于 二氯甲烷(10.0mL),加入1-(4-甲氧基苯基)-N-甲基甲胺(2.0g,13.2mmol,1.0eq)和冰醋酸(0.2mL),升至室温搅拌2小时,加入三乙酰氧基硼氢化钠(5.6g,26.4mmol,2.0eq),室温反应过夜。TLC监测反应完全,冰浴下滴加饱和碳酸氢钠水溶液(200mL),调节pH值至7~8,加入二氯甲烷(100mL),分液,有机相用无水硫酸钠干燥,过滤,浓缩,粗品经硅胶柱色谱(二氯甲烷∶甲醇=140∶3v/v)纯化得到产物(2.1g,收率:49.6%)。
步骤2:(3-(甲氨基)环丁基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000181
将(3-((4-甲氧基苄基)(甲基)氨基)环丁基)氨基甲酸叔丁酯(2.1g,6.55mmol,1.0eq)溶解于甲醇(20.0mL),加入钯碳(525.0mg),氢气置换3次,室温搅拌过夜,TLC监测反应完全,过滤,滤液减压浓缩得产品(1.83g粗品)。
步骤3:(3-(3-甲氧基-N-甲基-4-(甲氨基)-5-硝基苯甲酰胺基)环丁基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000182
将3-甲氧基-4-(甲氨基)-5-硝基苯甲酸(1.72g,6.15mmol,1.0eq)溶于DMF(10mL),冰浴下加入DIPEA(1.97g,12.31mmol,2.0eq)和HATU(3.19g,6.76mmol,1.1eq),搅拌2小时,加入(3-(甲氨基)环丁基)氨基甲酸叔丁酯(1.53g,6.15mmol,1.0eq)的DMF溶液(10mL),逐渐升至室温搅拌2小时,TLC监测反应完全,减压浓缩,加入乙酸乙酯(100mL)和饱和氯化钠水溶液(150mL),分液,有机相用无水硫酸钠干燥,浓缩,粗品经硅胶柱色谱纯化(二氯甲烷∶甲醇=200∶1v/v)得到产品(1.7g,两步收率:67.7%)。
步骤4:(3-(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-N,1-二甲基-1H-苯并[d]咪唑-5-甲酰氨基)环丁基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000183
将(3-(3-甲氧基-N-甲基-4-(甲氨基)-5-硝基苯甲酰胺基)环丁基)氨基甲酸叔丁酯(1.17g,2.87mmol,1.0eq)溶于乙醇(9.0mL),加入6-乙基-6H-噻吩并[2,3-b]吡咯-5-甲醛(515.1mg,2.87mmol,1.0eq)和连二亚硫酸钠(1.50g,8.61mmol,3.0eq)的水溶液(4.5mL),100℃反应过夜。TLC监测反应完全,减压浓缩,粗品用二氯甲烷(100mL)溶解,过滤,滤液无水硫酸钠干燥,浓缩,粗品经硅胶柱色谱纯化(二氯甲烷∶甲醇=100∶1v/v)得产品(1.01g,收率:65.5%)。
步骤5:N-(3-氨基环丁基)-2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-N,1-二甲基-1H-苯并[d]咪唑-5-甲酰胺的合成
Figure PCTCN2019076129-appb-000184
将(3-(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-N,1-二甲基-1H-苯并[d]咪唑-5-甲酰氨基)环丁基)氨基甲酸叔丁酯(300.0mg,0.55mmol,1.0eq)溶解于二氯甲烷(3.0mL),冰浴下滴加三氟乙酸(1.5mL),升至室温搅拌2小时。TLC监测反应完全,冰浴下滴加饱和碳酸氢钠水溶液(10mL),调节pH值至7~8,加入二氯甲烷(15mL)萃取,有机相用无水硫酸钠干燥,减压浓缩得到产品(109.3mg,收率:44.8%)。
1HNMR(400MHz,DMSO-d 6)δ(ppm):7.21(s,1H),7.16-7.17(m,1H),7.11-7.13(m,1H),6.90(s,1H),6.76(s,1H),4.76(s,1H),4.37-4.42(m,2H),4.10(s,3H),3.97(s,3H),3.42-3.45(m,1H),2.97-2.98(s,3H),2.43-2.46(m,2H),2.41(m,1H),1.90-1.95(m,1H),1.81(m,2H),1.31-1.39 (m,3H).
分子式:C 23H 27N 5O 2S 分子量:437.56LC-MS(Pos,m/z)=338.6[M+H] +.
实施例17:(4-氨基-6-氮杂螺[2.5]辛烷-6-基)(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物14)的合成
Figure PCTCN2019076129-appb-000185
步骤1:3-甲氧基-4-(甲氨基)-5-硝基苯甲酸的合成
Figure PCTCN2019076129-appb-000186
将3-甲氧基-4-(甲氨基)-5-硝基苯甲酸甲酯(3.0g,12.4mmol,1.0eq)溶解于四氢呋喃(30mL)中,滴加一水合氢氧化锂(2.0g,49.9mmol,4.0eq)的水溶液(6mL),45℃加热搅拌过夜,TLC监测反应完全,减压浓缩,向反应液中加水(60mL),冰浴下用2mol/L盐酸调节pH值至2~3,加入二氯甲烷(50mL)萃取,有机相用无水硫酸钠干燥,减压浓缩得到产品(2.6g,收率:92.8%)。
步骤2:(6-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)-6-氮杂螺[2.5]辛烷-4-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000187
将3-甲氧基-4-(甲氨基)-5-硝基苯甲酸(150.0mg,0.66mmol,1.0eq)加入DMF(1.5mL)和四氢呋喃(12mL)的混合溶液中,冰浴下加入DIPEA(171.2mg,1.32mmol,2.0eq)和HATU(277.1mg,0.72mmol,1.1 eq),反应2小时,加入(6-氮杂螺[2.5]辛烷-4-基)氨基甲酸叔丁酯(165.0mg,0.72mmol,1.1eq),加毕,逐渐升至室温搅拌过夜,TLC监测反应完全,减压浓缩,加入乙酸乙酯(15mL)和饱和氯化钠水溶液(20mL),分液,有机相用无水硫酸钠干燥,过滤,浓缩,粗品经硅胶柱色谱纯化(二氯甲烷∶甲醇=160∶1v/v)得产品(200.6mg,收率:69.4%)。
步骤3:(6-(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)-6-氮杂螺[2.5]辛烷-4-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000188
将(6-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)-6-氮杂螺[2.5]辛烷-4-基)氨基甲酸叔丁酯(200.0mg,0.46mmol,1.0eq)溶于乙醇(8.0mL)中,加入6-乙基-6H-噻吩并[2,3-b]吡咯-5-甲醛(82.5mg,0.46mmol,1.0eq)和连二亚硫酸钠(240.2mg,1.38mmol,3.0eq)的水溶液(1.5mL),微波100℃反应5小时,TLC监测反应完全,减压浓缩,加二氯甲烷(20mL)和水(15mL),分液,有机相用无水硫酸钠干燥,过滤,浓缩,粗品经硅胶柱色谱(二氯甲烷∶甲醇=80∶1v/v)纯化得产品(130.1mg,收率:50.1%)。
步骤4:(4-氨基-6-氮杂螺[2.5]辛烷-6-基)(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000189
将(6-(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)-6-氮杂螺[2.5]辛烷-4-基)氨基甲酸叔丁酯(130.1mg,0.23mmol,1.0eq)加入到乙醇(4mL)中,冰浴下滴加氯化氢乙醇溶液(30w/w%,4mL),升至室温搅拌2小时。TLC监测反应完全后,减压浓缩除去乙醇,冰浴下滴加饱和碳酸氢钠水溶液(10mL),调节pH值至 7~8,加入二氯甲烷(15mL),分液,有机相用无水硫酸钠干燥,过滤,浓缩,粗品经制备薄层色谱(二氯甲烷∶甲醇=15∶1v/v)纯化得到产物(20.1mg,收率:18.8%)。
1HNMR(400MHz,DMSO-d 6)δ(ppm):7.35(s,1H),7.16-7.18(m,1H),7.11-7.13(m,1H),6.90(s,2H),4.37-4.42(m,2H),4.10(s,3H),3.97(s,3H),3.54(m,2H),3.51(m,1H),1.74(m,2H),1.32-1.35(m,3H),1.19-1.24(m,2H),0.57(s,2H),0.22-0.30(m,2H).
分子式:C 25H 29N 5O 2S 分子量:463.60LC-MS(Pos,m/z)=464.2[M+H] +.
实施例18:(3-氨基-3,4-二氢喹啉-1(2H)-基)(2-(1-乙基-1H-吡咯并[2,3-b]吡啶-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物15)的合成
Figure PCTCN2019076129-appb-000190
步骤1:3-甲氧基-4-(甲基氨基)-5-硝基苯甲酰氯的合成
Figure PCTCN2019076129-appb-000191
将3-甲氧基-4-(甲基氨基)-5-硝基苯甲酸(600.0mg,2.65mmol,1.0eq)溶于二氯亚砜(10mL)中,常温搅拌0.5h,反应液减压浓缩得黄色固体状产物(540mg粗品),直接用于下一步反应。
步骤2:(1-(3-甲氧基-4-(甲基氨基)-5-硝基苯甲酰基)-1,2,3,4-四氢喹啉-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000192
将3-甲氧基-4-(甲基氨基)-5-硝基苯甲酰氯(300mg,1.23mmol,1.0eq)、(1,2,3,4-四氢喹啉-3-基)氨基甲酸叔丁酯(305mg,1.23mmol,1.0eq)、三乙胺(269mg,2.69mmol,3.0eq)和DMAP(15mg,0.12mmol,0.1eq)溶解于二氯甲烷(10mL)中,常温搅拌1h。加水(5mL)淬灭,用二氯甲烷(20mL×2)萃取,有机相合并,浓缩,粗品经硅胶柱色谱纯化(二氯甲烷∶甲醇=30∶1v/v)得到红色固体状产物(150mg,产率:26.7%)。
步骤3:(1-(2-(1-乙基-1H-吡咯并[2,3-b]吡啶-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)-1,2,3,4-四氢喹啉-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000193
将(1-(3-甲氧基-4-(甲基氨基)-5-硝基苯甲酰基)-1,2,3,4-四氢喹啉-3-基)氨基甲酸叔丁酯(50.0mg,0.1mmol,1.0eq)和1-乙基-1H-吡咯并[2,3-b]吡啶-2-甲醛(17.4mg,0.1mmol,1.0eq)溶于乙醇(2mL)和水(1mL),加入连二亚硫酸钠(53mg,0.3mmol,1.0eq),100℃微波反应5h。反应液浓缩,粗品经硅胶柱色谱纯化(石油醚∶乙酸乙酯=1∶1v/v)得到无色油状产物(35mg,产率:60.3%)。
步骤4:(3-氨基-3,4-二氢喹啉-1(2H)-基)(2-(1-乙基-1H-吡咯并[2,3-b]吡啶-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000194
将(1-(2-(1-乙基-1H-吡咯并[2,3-b]吡啶-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)-1,2,3,4-四氢喹啉-3-基)氨基甲酸叔丁酯(35mg)溶于三氟乙酸(1mL),常温下搅拌1h,反应液浓缩,粗品经反相柱色谱纯化(乙腈∶水∶氨水=30∶100∶0.05v/v)得到白色固体状产物(4.4mg,产率:26.6%)。
1HNMR(400MHz,DMSO-d 6)δ(ppm):8.46(m,1H),8.04(m,1H),7.50(s,1H),7.31(s,2H),7.13-7.26(m,1H),6.91-7.09(m,3H),6.77(s,1H),4.62-4.73(m,2H),4.22(m,1H),4.13(s,3H),3.93(s,3H),3.68(m,1H),3.57(m,1H),3.21(m,1H),2.73(m,1H),1.20-1.41(m,3H).
分子式:C 28H 28N 6O 2  分子量:480.23  LC-MS(Pos,m/z)=481.21[M+H] +.
实施例19:(4-氨基-6-氮杂螺[2.5]辛烷-6-基)(2-(1-乙基-1H-吡咯并[2,3-b]吡啶-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮盐酸盐(化合物16的盐酸盐)的合成
步骤1:(6-(3-甲氧基-4-(甲基氨基)-5-硝基苯甲酰基)-6-氮杂螺[2.5]辛烷-4-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000195
将3-甲氧基-4-(甲基氨基)-5-硝基苯甲酸(150.0mg,0.663mmol,1.0eq)溶解于四氢呋喃(4mL)和N,N-二甲基甲酰胺(2mL)的混合溶剂中,冰浴下加入N,N-二异丙基乙胺(171.2mg,1.326mmol,2.0eq)和2-(7-氧化苯并三氮唑)-N,N,N′,N′-四甲基脲鎓六氟磷酸盐(277.3mg,0.729mmol,1.1eq),冰浴下搅拌40分钟,滴加(6-氮杂螺[2.5]辛烷-4-基)氨基甲酸叔丁酯(165.0mg,0.729mmol,1.1eq)的四氢呋喃溶液(2mL),滴加完毕逐渐升至室温搅拌10小时。TLC显示反应完全,将体系浓缩,加入水(15mL)和二氯甲烷(30mL),分液,有机相用无水硫酸钠干燥,浓缩,粗品经硅胶柱色谱分离(200-300目硅胶,二氯甲烷∶甲醇=40∶1v/v)得到黄色固体状产物(200.3mg,产率:69.4%)。
步骤2:(6-(2-(1-乙基-1H-吡咯并[2,3-b]吡啶-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)-6-氮杂螺[2.5]辛烷-4-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000196
将(6-(3-甲氧基-4-(甲基氨基)-5-硝基苯甲酰基)-6-氮杂螺[2.5]辛烷-4-基)氨基甲酸叔丁酯(200.0mg,0.46mmol,1.0eq)溶于乙醇(8mL)中,加入1-乙基-1H-吡咯并[2,3-b]吡啶-2-甲醛(80.1mg,0.46mmol,1.0eq)、连二亚硫酸钠(240.2mg,1.38mmol,3.0eq)和水(1mL),微波100℃反应5小时。TLC显示反应完全,减压浓缩,加入水(10mL)和二氯甲烷(20mL),分液,有机相用无水硫酸钠干燥,过滤,浓缩,粗品经硅胶柱色谱分离(200-300目硅胶,二氯甲烷∶甲醇=80∶1v/v)得到产物(135.1mg,产率:52.7%)。
步骤3:(4-氨基-6-氮杂螺[2.5]辛烷-6-基)(2-(1-乙基-1H-吡咯并[2,3-b]吡啶-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮盐酸盐的合成
Figure PCTCN2019076129-appb-000197
将(6-(2-(1-乙基-1H-吡咯并[2,3-b]吡啶-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)-6-氮杂螺[2.5]辛烷-4-基)氨基甲酸叔丁酯(121.7mg,0.217mmol,1.0eq)溶解在乙醇(4mL)中,滴加氯化氢的乙醇溶液(30w/w%,4mL),滴加完毕室温反应4h。TLC监测反应完全,浓缩,粗品用石油醚(4ml)和甲基叔丁基醚(2mL)浆洗得到产物(4-氨基-6-氮杂螺[2.5]辛烷-6-基)(2-(1-乙基-1H-吡咯并[2,3-b]吡啶-2-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮盐酸盐(124.4mg,产率:100%)。
1HNMR(400MHz,DMSO-d 6)δ(ppm):8.48(d,J=4.4Hz,1H),8.37(brs,2H),8.21(d,J=7.8Hz,1H),7.59(brs,1H),7.27-7.29(m,1H),7.18(s,2H),4.58-4.63(m,2H),4.16(s,3H),4.03(s,3H),3.16-3.25(m,1H), 3.04-3.06(m,1H),2.67-2.73(m,1H),2.25-2.41(m,1H),1.23-1.26(m,3H),0.82-0.84(m,2H),0.65-0.68(m,1H),0.42-0.52(m,2H).
分子式:C 26H 30N 6O 2  分子量:458.57  LC-MS(Pos,m/z)=459.3[M+H] +.
实施例20:(R)-(3-氨基吡咯烷-1-基)(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物17)的合成
Figure PCTCN2019076129-appb-000198
步骤1:(R)-(1-(3-甲氧基-4-甲氨基-5-硝基苯甲酰基)吡咯烷-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000199
将3-甲氧基-4-甲氨基-5-硝基苯甲酸(500.0mg,2.21mmol,1.0eq)加入到DMF(5mL)中,冰浴降温至0℃,加入HATU(1260.8mg,3.32mmol,1.5eq)和DIPEA(857.1mg,6.63mmol,3.0eq),0℃下搅拌60min后加入(R)-吡咯烷-3-基氨基甲酸叔丁酯(370.5mg,1.99mmol,0.9eq),室温搅拌反应过夜。TLC监测反应完全,减压浓缩,加入乙酸乙酯(10mL),依次用饱和碳酸氢钠水溶液(10mL)、饱和氯化铵水溶液(10mL)、水(10mL×4)和饱和食盐水(10mL)洗涤,无水硫酸钠干燥,抽滤,减压浓缩,粗品经制备薄层色谱纯化(PE∶EA=1∶1,0∶1v/v)得产品(508.0mg,收率:64.7%)。
步骤2:(R)-(1-(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)吡咯烷-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000200
将(R)-(1-(3-甲氧基-4-甲氨基-5-硝基苯甲酰基)吡咯烷-3-基)氨基甲酸叔丁酯(306.0mg,0.78mmol,1.5eq)溶于水(2mL)和乙醇(10mL)的混合液中,加入6-乙基-6H-噻吩并[2,3-b]吡咯-5-甲醛(92.7mg,0.52mmol,1.0eq.)和Na 2S 2O 4(270.2mg,1.55mmol,3.0eq.),微波100℃反应5h。TLC监测反应完全,减压浓缩,过滤,滤液加入二氯甲烷(5mL×3)萃取,合并有机相,无水硫酸钠干燥,浓缩,粗品经硅胶柱色谱(DCM∶MeOH=50∶1~30∶1v/v)纯化得产品(250mg,收率:92.3%)。
步骤3:(R)-(3-氨基吡咯烷-1-基)(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000201
冰浴下,将(R)-(1-(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)吡咯烷-3-基)氨基甲酸叔丁酯(245.0mg,0.47mmol)溶于氯化氢的乙醇溶液(30w/w%,17mL)中,缓慢慢升至室温搅拌反应2h。TLC监测反应完全,减压浓缩,固体加水(10mL)溶解,用10w/w%NaHCO 3水溶液调节pH值至7~8,加入二氯甲烷(5mL×3)萃取,合并有机相,用无水硫酸钠干燥,抽滤,滤液减压浓缩,粗品经制备薄层色谱纯化(DCM∶MeOH=20∶1,10∶1v/v)得产品(108mg,收率:54.2%)。
1H NMR(400MHz,DMSO-d 6)δ(ppm):7.46-7.42(d,1H),7.18-7.12(q,2H),6.96-6.91(d,2H),4.41-4.39(d,2H),4.11(s,3H),3.98(s,3H),3.66-3.64(d,3H),3.51(s,2H),2.06-2.03(d,1H),1.73-1.72(d,1H),1.36-1.32(t,3H).
分子式:C 22H 25N 5O 2S  分子量:423.54  LC-MS(Pos,m/z)=424.38[M+H] +.
实施例21:(R)-(3-氨基哌啶-1-基)(2-(6-(环丙基甲基)-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物18)的合成
步骤1:6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯的合成
Figure PCTCN2019076129-appb-000202
将金属钠(3.0g,130.0mmol,4.86eq.)加入到无水乙醇(60mL)中,搅拌至溶解后滴加噻吩-3-甲醛(3.0g,26.7mmol,1.0eq)和叠氮乙酸乙酯(15.0g,116.0mmol,4.3eq.)的乙醇溶液(40mL),逐渐升至室温搅拌过夜,TLC监测反应完全,冰浴下加饱和氯化铵水溶液(200mL),加入甲基叔丁基醚(200mL×3)萃取,有机相用无水硫酸钠干燥,减压浓缩,加入二甲苯(50mL)回流搅拌过夜,TLC检测反应完全,减压浓缩,粗品经硅胶柱色谱(石油醚∶乙酸乙酯=5∶1v/v)纯化得到产品(435.0mg,收率:8.3%)。
步骤2:6-(环丙基甲基)-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯的合成
Figure PCTCN2019076129-appb-000203
将6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(435.0mg,2.22mmol,1.0eq)溶解于DMF(5mL),加入(溴甲基)环丙烷(449.5mg,3.33mmol,1.5eq)和无水碳酸钾(613.6mg,4.44mmol,2.0eq),50℃反应3小时,TLC监测反应完全,减压浓缩,加入乙酸乙酯(15mL)和水(40mL),分液,有机相用无水硫酸钠干燥,浓缩,粗品经硅胶柱色谱(石油醚∶乙酸乙酯=100∶1v/v)纯化得到产品(507.6mg,收率:91.7%)。
步骤3:(6-(环丙基甲基)-6H-噻吩并[2,3-b]吡咯-5-基)甲醇的合成
Figure PCTCN2019076129-appb-000204
将6-(环丙基甲基)-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(507.6mg,2.03mmol,1.0eq)溶于THF(2.5mL)中,冰浴下加入二异丁基氢化铝的甲苯溶液(1.5mol/L,4.66mL),冰浴反应2小时,TLC监测反应完全,加入饱和硫酸钠水溶液(20mL),搅拌半小时,抽滤,滤液用无水硫酸钠干燥,减压浓缩得产品(430.9mg粗品)。
步骤4:6-(环丙基甲基)-6H-噻吩并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000205
将(6-(环丙基甲基)-6H-噻吩并[2,3-b]吡咯-5-基)甲醇(430.9mg粗品)加入到二氯甲烷(12.5mL)中,加入二氧化锰(1.8g,20.3mmol,10.0eq),40℃反应1小时,TLC监测反应完全,过滤,滤液减压浓缩得到产品(371.7mg,两步收率:89.2%)。
步骤5:(R)-(1-(2-(6-(环丙基甲基)-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000206
将(R)-(1-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(701.0mg,1.71mmol,1.0eq)加入到乙醇(10.0mL)中,再加入6-(环丙基甲基)-6H-噻吩并[2,3-b]吡咯-5-甲醛(371.7mg,1.81mmol,1.0eq)和连二亚硫酸钠(945.3mg,5.43mmol,3.0eq)的水溶液(5mL),微波 100℃反应5小时,TLC监测反应完全,减压浓缩,加入水(15mL)和二氯甲烷(20mL),分液,有机相用无水硫酸钠干燥,过滤,浓缩,粗品经硅胶柱色谱(二氯甲烷∶甲醇=100∶1~80∶1v/v)纯化得产品(440.0mg,收率:44.0%)。
步骤6:(R)-(3-氨基哌啶-1-基)(2-(6-(环丙基甲基)-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000207
将(R)-(1-(2-(6-(环丙基甲基)-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯(440.0mg,0.78mmol,1.0eq)加入二氯甲烷(2.0mL)搅拌溶解,冰浴下滴加三氟乙酸(2.0mL),升至室温搅拌2小时。TLC监测反应完全,冰浴下滴加饱和碳酸氢钠水溶液(10mL),调节pH值至7~8,加入二氯甲烷(15mL)萃取,有机相用无水硫酸钠干燥,过柱,二氯甲烷∶甲醇=40∶1~30∶1~20∶1v/v分离纯化得到(R)-(3-氨基哌啶-1-基)(2-(6-(环丙基甲基)-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮,减压浓缩得到产品(208.2mg,收率:57.6%)。
1HNMR(400MHz,DMSO-d 6)δ(ppm):7.25-7.26(s,1H),7.15-7.16(m,1H),7.10-7.11(m,1H),6.90(s,1H),6.82(s,1H),4.24-4.26(m,2H),4.09(s,3H),3.97(s,3H),2.92(s,1H),2.67(s,2H),1.85-1.88(m,1H),1.58-1.67(m,3H),1.39-1.46(m,1H),1.19-1.25(m,2H),0.45-0.48(m,2H),0.26(m,2H).
分子式:C 25H 29N 5O 2S 分子量:463.60LC-MS(Pos,m/z)=464.38[M+H] +.
实施例22:(R)-(3-氨基氮杂环庚烷-1-基)(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物19)的合成
步骤1:(R)-3-(((苄氧基)羰基)氨基)氮杂环庚烷-1-羧酸叔丁酯的合成
Figure PCTCN2019076129-appb-000208
将(R)-3-氨基氮杂环庚烷-1-羧酸叔丁酯(500.0mg,2.33mmol,1.0eq)和三乙胺(472.0mg,4.67mmol,2.0eq)溶于二氯甲烷(5.0mL)中,0℃下加入氯甲酸苄酯(517.0mg,3.03mmol,1.3eq),氮气保护下室温反应2h,TLC显示反应完全。将二氯甲烷减压浓缩得产品(1.0g粗品)。
步骤2:(R)-氮杂环庚烷-3-基氨基甲酸苄酯的合成
Figure PCTCN2019076129-appb-000209
将(R)-3-(((苄氧基)羰基)氨基)氮杂环庚烷-1-羧酸叔丁酯(1.0g)溶于乙醇(2.5mL)中,加入氯化氢的乙醇溶液(30w/w%,2.5mL),室温过夜,TLC显示反应完全。将反应液减压浓缩,粗品经硅胶柱色谱纯化(DCM∶MeOH=100∶1~20∶1v/v)得产品(200.0mg,两步收率:34.54%)。
步骤3:(R)-(1-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)氮杂环庚烷-3-基)氨基甲酸苄基酯的合成
Figure PCTCN2019076129-appb-000210
将3-甲氧基-4-(甲氨基)-5-硝基苯甲酸(200.0mg,0.805mmol,1.0eq)和DIPEA(312.0mg,2.42mmol,3.0eq)溶于DMF(2.0mL),0℃氮气保护下,加入HATU(459.0mg,1.21mmol,1.5eq),搅拌0.5h后,加入(R)-氮杂环庚烷-3-基氨基甲酸苄酯(200.0mg,0.805mmol),室温反应2h,TLC显示反应完全。加入水(40.0mL),乙酸乙酯(15.0mL×2)萃取,合并有机相,依次用饱和碳酸钠水溶液(2.0mL)、饱和氯化铵水溶液(2.0mL)和饱和氯化钠溶液(2.0mL)洗,干燥,减压浓缩,粗品经硅胶柱色谱(DCM∶ MeOH=200∶1~100∶1v/v)得产品(323.0mg,收率:87.9%)。
步骤4:(R)-(1-(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)氮杂环庚烷-3-基)氨基甲酸苄酯的合成
Figure PCTCN2019076129-appb-000211
将(R)-(1-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)氮杂环庚烷-3-基)氨基甲酸苄基酯(300.0mg,0.657mmol,1.0eq)、6-乙基-6H-噻吩并[2,3-b]吡咯-5-甲醛(118.0mg,0.657mmol,1.0eq)和连二亚硫酸钠(344.0mg,1.98mmol,3.0eq)溶于乙醇(6.0mL)和水(3.0mL),氮气保护下,微波100℃反应5h,TLC显示反应完全。将反应液减压浓缩,加入乙酸乙酯,抽滤,滤液减压浓缩,粗品经硅胶柱色谱(DCM∶MeOH=100∶1~60∶1v/v)纯化得产品(260.0mg,收率:67.5%)。
步骤5:(R)-(3-氨基氮杂环庚烷-1-基)(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000212
将(R)-(1-(2-(6-乙基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)氮杂环庚烷-3-基)氨基甲酸苄酯(340.0mg,0.580mmol,1.0eq)溶于乙腈(8.0mL)中,氮气保护下,加入三甲基碘硅烷(290.0mg,1.45mmol,2.5eq),反应3h,TLC显示反应完全。加入三乙胺(2.0mL)淬灭,加入水(30.0mL),乙酸乙酯(10.0mL×3)萃取,合并有机相,依次用饱和碳酸氢钠水溶液(2.0mL)和饱和氯化钠溶液(2.0mL)洗,无水硫酸钠干燥,减压浓缩,粗品经硅胶柱色谱(DCM∶MeOH=100∶1~11∶1v/v)纯化得产品(230.0mg,收率:87.8%)。
1H NMR(400MHz,DMSO-d6)δ(ppm):7.24(s,1H),7.18-7.16(m,1H), 7.13-7.11(m,1H),6.90(s,1H),6.80(s,1H),4.43-4.37(m,2H),4.10(s,3H),3.97(s,3H),3.41-3.17(m,4H),2.98-2.81(m,2H),1.85-1.72(m,4H),1.36-1.32(m,4H).
分子式:C 24H 29N 5O 2S  分子量:451.59  LC-MS(Pos,m/z)=452.37[M+H] +.
实施例23:化合物(R)-(3-氨基哌啶-1-基)(2-(6-异丁基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物20)的合成
Figure PCTCN2019076129-appb-000213
步骤1:中间体6-异丁基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯的合成
Figure PCTCN2019076129-appb-000214
将中间体6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(1.50g,7.68mmol,1.0eq)、碳酸铯(3.75g,11.52mmol,1.5eq)和1-溴-2-甲基丙烷(1.58g,11.52mmol,1.5eq)依次加入到乙腈(25mL)中,加热至90℃反应1.5h,TLC显示反应完全,冷却至室温,过滤,滤液浓缩,粗品经硅胶柱色谱纯化(乙酸乙酯∶石油醚=3∶100v/v)得产品(1.52g,产率:78.7%)。
步骤2:中间体(6-异丁基-6H-噻吩并[2,3-b]吡咯-5-基)甲醇的合成
Figure PCTCN2019076129-appb-000215
将6-异丁基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(1.50g,5.97mmol, 1.0eq)溶于THF(25mL)中,冰水浴冷却至0℃左右,将氢化铝锂(227mg,5.97mmol,1.0eq)加入到上述溶液中,搅拌0.5h,然后室温反应2h。TLC显示反应完全,依次加入水(0.23mL)、15w/w%氢氧化钠溶液(0.23mL)和水(0.69mL)到上述反应液中,有大量白色絮状固体析出,加入无水硫酸镁干燥,过滤,滤液浓缩得产品(1.15g,产率:92.1%)。
步骤3:中间体6-异丁基-6H-噻吩并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000216
将(6-异丁基-6H-噻吩并[2,3-b]吡咯-5-基)甲醇(500mg,2.39mmol,1.0eq)加入到二氯甲烷(20mL)中,再加入二氧化锰(2.08g,23.89mmol,10eq),室温搅拌反应16h,TLC显示反应完全,硅藻土过滤,滤液浓缩得棕色油状产物(390mg,产率:78.8%)。
步骤4:中间体(R)-(1-(2-(6-异丁基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000217
将6-异丁基-6H-噻吩并[2,3-b]吡咯-5-甲醛(365mg,1.76mmol,1.2eq)、(R)-(1-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(600mg,1.47mmol,1.0eq)和连二亚硫酸钠(1.28g,7.34mmol,5.0eq)依次加入到乙醇(15mL)和水(7.5mL)的混合溶剂中,加热至90℃反应10h,TLC显示反应完全,将反应液倒入水(50mL)中,乙酸乙酯(30mL)萃取,有机相用依次用水(20mL)和饱和食盐水洗(20mL),干燥 (Na 2SO 4),过滤,滤液浓缩,粗品经硅胶柱色谱纯化(二氯甲烷∶甲醇=50∶1v/v)得产品(550mg,产率:66.2%)。
步骤5:中间体(R)-(3-氨基哌啶-1-基)(2-(6-异丁基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000218
将中间体(R)-(1-(2-(6-异丁基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯(300mg,0.53mmol,1.0eq)和苯酚(149.72mg,1.59mmol,3.0eq)溶于甲醇(5mL)中,加入氯化氢乙醇溶液(5mL),室温搅拌反应2h,TLC显示反应完全,将反应液浓缩,加入二氯甲烷(20mL),依次用饱和碳酸氢钠水溶液(15mL)、水(15mL)和饱和食盐水(15mL)洗,干燥(Na 2SO 4),过滤,滤液浓缩,粗品经硅胶柱色谱纯化(二氯甲烷∶甲醇=50∶1~10∶1v/v)得产品(130mg,产率:52.7%)。
1H NMR(400MHz,DMSO-d 6)δ(ppm):7.27(m,1H),7.16(d,1H),7.11(d,1H),6.91(s,1H),6.82(s,1H),4.10-4.40(m,3H),4.09(s,3H),3.97(s,3H),3.60-3.80(m,1H),2.90-3.00(m,1H),2.60-2.75(m,2H),2.00-2.30(m,3H),1.80-1.95(m,1H),1.60-1.75(m,1H),1.40-1.50(m,1H),1.20-1.35(m,1H),0.71(d,6H).
分子式:C 25H 31N 5O 2S  分子量:465.62  LC-MS(m/z)=465.94[M+H] +.
实施例24:(R)-(3-氨基哌啶-1-基)(7-甲氧基-1-甲基-2-(6-(2,2,2-三氟乙基)-6H-噻吩并[2,3-b]吡咯-5-基)-1H-苯并[d]咪唑-5-基)甲酮(化合物21)的合成
Figure PCTCN2019076129-appb-000219
步骤1:2,2,2-三氟乙基4-甲基苯磺酸酯的合成
Figure PCTCN2019076129-appb-000220
将2,2,2-三氟乙醇(5.77g,57.70mmol,1.1eq)、对甲苯磺酰氯(10.0g,52.45mmol,1.0eq)和三乙胺(10.62g,104.91mmol,2.0eq)溶于二氯甲烷(150mL)中,常温搅拌18小时,反应完毕。水洗(40mL),有机相干燥,浓缩,粗品经硅胶柱色谱纯化(石油醚∶乙酸乙酯=30∶1v/v)得到无色油产物(4.7g,产率:34.8%)。
步骤2:6-(2,2,2-三氟乙基)-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯的合成
Figure PCTCN2019076129-appb-000221
将6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(500mg,2.56mmol,1.0eq)、2,2,2-三氟乙基4-甲基苯磺酸酯(846mg,3.33mmol,1.3eq)和碳酸铯(1.25g,3.84mmol,1.5eq)加入乙腈(15mL)中,加热回流35小时,TLC显示反应完全。过滤,滤液浓缩,粗品经硅胶柱色谱纯化(石油醚∶乙酸乙酯=50∶1v/v)得产品(580mg,产率:81.8%)。
步骤3:(6-(2,2,2-三氟乙基)-6H-噻吩并[2,3-b]吡咯-5-基)甲醇的合成
Figure PCTCN2019076129-appb-000222
将四氢铝锂(79.39mg,2.09mmol,1.0eq)置于四氢呋喃(20mL)中,冷却至0℃。将6-(2,2,2-三氟乙基)-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(580mg,2.09mmol,1.0eq)溶于四氢呋喃(4mL),缓慢加入到四氢铝锂的四氢 呋喃悬浊液中,维持反应体系温度在0℃左右。加毕,继续搅拌2小时,TLC显示反应完毕。依次缓慢加入水(80mg)、15w/w%氢氧化钠水溶液(80mg)和水(240mg),加入无水硫酸钠干燥过夜。过滤,滤液浓缩得产品(490mg,产率:99.6%)。
步骤4:6-(2,2,2-三氟乙基)-6H-噻吩并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000223
将(6-(2,2,2-三氟乙基)-6H-噻吩并[2,3-b]吡咯-5-基)甲醇(250mg,1.06mmol,1.0eq)溶于四氢呋喃(4mL)中,加入二氧化锰(1.0g,10.6mmol,10.0eq),室温搅拌18小时。过滤,滤液浓缩,得到产品(240mg粗品),直接用于下步反应。
步骤5:(R)-(1-(7-甲氧基-1-甲基-2-(6-(2,2,2-三氟乙基)-6H-噻吩并[2,3-b]吡咯-5-基)-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000224
将6-(2,2,2-三氟乙基)-6H-噻吩并[2,3-b]吡咯-5-甲醛(240mg粗品)和(R)-(1-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(324mg,0.79mmol)溶于乙醇(4mL),连二亚硫酸钠(748mg,4.3mmol)溶于水(2mL),两者混合后90℃下加热搅拌4小时,原料反应完毕。浓缩,加入乙酸乙酯(50mL)和水(10mL),将有机相用水洗(10mL×2),无水硫酸钠干燥,过滤,浓缩,粗品经硅胶柱色谱纯化(二氯甲烷∶甲醇=60∶1v/v)得到产品(380mg,产率:80.9%)。
步骤6:化合物(R)-(3-氨基哌啶-1-基)(7-甲氧基-1-甲基-2-(6-(2,2,2-三氟 乙基)-6H-噻吩并[2,3-b]吡咯-5-基)-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000225
将(R)-(1-(7-甲氧基-1-甲基-2-(6-(2,2,2-三氟乙基)-6H-噻吩并[2,3-b]吡咯-5-基)-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯(380mg,0.64mmol,1.0eq)溶于二氯甲烷(2mL)和三氟乙酸(2mL)中,加入苯酚(302mg,3.2mmol,5.0eq),常温搅拌10分钟,TLC显示反应完毕。低温浓缩,加入二氯甲烷(100mL),用15w/w%氢氧化钠水溶液洗(20mL×2),有机相干燥,浓缩,加入乙酸乙酯(0.5mL)和石油醚(1mL),析出固体,过滤得产品(230mg,产率:73.2%)。
1H NMR(400MHz,CDCl 3)δ(ppm):7.41(s,1H),7.08(d,J=5.28Hz,1H),7.01(d,J=5.28Hz,1H),6.86(s,1H),6.83(s,1H),5.23-5.39(m,2H),4.18(s,3H),4.02(s,3H),2.72-3.15(m,4H),2.07(s,1H),1.42-1.89(m,4H).
分子式:C 23H 24F 3N 5O 2S  分子量:491.16  LC-MS(Pos,m/z)=492.23[M+H] +.
实施例25:(R)-(3-氨基哌啶-1-基)(2-(6-(环丁基甲基)-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物22)的合成
Figure PCTCN2019076129-appb-000226
步骤1:中间体6-(环丁基甲基)-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯的合 成
Figure PCTCN2019076129-appb-000227
将6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(1.50g,7.68mmol,1.0eq)、碳酸铯(3.75g,11.52mmol,1.5eq)和溴甲基环丁烷(1.72g,11.52mmol,1.5eq)依次加入到乙腈(25mL)中,加热至90℃反应1.5h,TLC显示反应完全,冷却至室温,过滤,滤液浓缩,粗品经硅胶柱色谱(乙酸乙酯∶石油醚=3∶100v/v)纯化得产品(1.45g,产率:71.7%)。
步骤2:中间体(6-(环丁基甲基)-6H-噻吩并[2,3-b]吡咯-5-基)甲醇的合成
Figure PCTCN2019076129-appb-000228
将6-(环丁基甲基)-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(1.45g,5.51mmol,1.0eq)溶于THF(25mL)中,冰水浴冷却至0℃左右,将氢化铝锂(209mg,5.51mmol,1.0eq)加入到上述溶液中,搅拌反应0.5h,然后室温反应2h。TLC显示反应完全。依次将水(0.21mL)、15w/w%氢氧化钠溶液(0.21mL)、水(0.63mL)加入到上述反应液中,有大量白色絮状固体析出,加入无水硫酸镁干燥,过滤,滤液浓缩得产品(1.2g,产率:98.5%)。
步骤3:中间体6-(环丁基甲基)-6H-噻吩并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000229
将(6-(环丁基甲基)-6H-噻吩并[2,3-b]吡咯-5-基)甲醇(500mg,2.26mmol,1.0eq)加入到二氯甲烷(20mL)中,再加入二氧化锰(1.96g,22.59mmol,10eq),室温搅拌反应15h,TLC显示反应完全,过滤,滤液浓缩得产物(420mg,产率:84.8%)。
步骤4:中间体(R)-(1-(2-(6-(环丁基甲基)-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000230
将6-(环丁基甲基)-6H-噻吩并[2,3-b]吡咯-5-甲醛(387mg,1.76mmol,1.2eq)、(R)-(1-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(600mg,1.47mmol,1.0eq)和连二亚硫酸钠(1.28g,7.34mmol,5.0eq)依次加入到乙醇(15mL)和水(7.5mL)的混合溶剂中,加热至90℃反应8h,TLC显示反应完全,将反应液倒入水(30mL)中,乙酸乙酯(30mL)萃取,有机相用水洗(20mL),饱和食盐水洗(20mL),干燥(Na 2SO 4),过滤,滤液浓缩,粗品经硅胶柱色谱(二氯甲烷∶甲醇=50∶1v/v)得产品(540mg,产率:63.6%)。
步骤5:化合物(R)-(3-氨基哌啶-1-基)(2-(6-(环丁基甲基)-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000231
将(R)-(1-(2-(6-(环丁基甲基)-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯(300mg, 0.52mmol,1.0eq)和苯酚(146.61mg,1.56mmol,3.0eq)溶于甲醇(5mL)中,加入氯化氢的乙醇溶液(30w/w%,5mL),室温搅拌反应2h,TLC显示反应完全,反应液浓缩,加入二氯甲烷(20mL),依次用饱和碳酸氢钠水溶液(15mL)、水(15mL)和饱和食盐水洗(15mL),干燥(Na 2SO 4),过滤,滤液浓缩,粗品经硅胶柱色谱纯化(二氯甲烷∶甲醇=50∶1~10∶1v/v)得产品(135mg,产率:54.4%)。
1H NMR(400MHz,DMSO-d 6)δ(ppm):7.28(m,1H),7.16(d,1H),7.10(d,1H),6.89(s,1H),6.82(s,1H),4.10-4.40(m,3H),4.09(s,3H),3.97(s,3H),3.60-3.80(m,1H),2.90-3.00(m,1H),2.60-2.75(m,2H),2.00-2.30(m,2H),1.60-1.95(m,6H),1.40-1.55(m,3H),1.20-1.30(m,2H).
分子式:C 26H 31N 5O 2S  分子量:477.63  LC-MS(m/z)=477.93[M+H] +.
实施例26:(R)-(3-氨基哌啶-1-基)(2-(6-(环丙基甲基)-2-甲基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物23)的合成
Figure PCTCN2019076129-appb-000232
步骤1:5-甲基噻吩-3-甲醛的合成
Figure PCTCN2019076129-appb-000233
将4-溴-2-甲基噻吩(20.0g,0.11mol,1.0eq)溶于四氢呋喃(200mL),氮气保护,-80℃下缓慢滴加正丁基锂(1.6mol/L,76.0mL,0.12mol,1.1eq),滴加完毕,搅拌10分钟。将DMF(12.4g,0.17mol,1.5eq)溶于四氢呋喃(10mL),-75℃滴加入反应液中,加毕,缓慢升至室温,冰浴下加入氯化铵饱和水溶液(30mL),用乙酸乙酯萃取(200mL×2),有机相合 并,干燥,过滤,浓缩,粗品经硅胶柱色谱纯化(石油醚∶乙酸乙酯=100∶1v/v)得产品(3.5g,收率:24.7%)。
1HNMR(400MHz,CDCl 3)δ(ppm):9.81(s,1H),7.89(s,1H),7.20(s,1H),2.51(s,3H).
分子式:C 6H 6OS  分子量:126.01  LC-MS(Pos,m/z)=127.23[M+H] +.
步骤2:2-叠氮基-3-(5-甲基噻吩-3-基)丙烯酸乙酯的合成
Figure PCTCN2019076129-appb-000234
取钠(2.5g,110mmol,4.0eq)溶解于乙醇(100mL)中制成乙醇钠溶液,降温至-5℃,将5-甲基噻吩-3-甲醛(3.5g,28mmol,1.0eq)和叠氮乙酸乙酯(14.3g,110mmol,4.0eq)溶解于乙醇(20mL)中,缓慢滴加入新制的乙醇钠溶液中,保持温度在-3℃左右,滴毕,保持温度在0℃以下,继续搅拌1小时。加入饱和氯化铵水溶液(30mL),将反应液倒入冰水中,搅拌十分钟,过滤,粗品溶解于乙酸乙酯,干燥,浓缩得到产品(4.0g,产率:60.7%)。
步骤3:2-甲基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯的合成
Figure PCTCN2019076129-appb-000235
将2-叠氮基-3-(5-甲基噻吩-3-基)丙烯酸乙酯(4.0g,17mmol,1.0eq)溶于甲苯(110mL),回流1小时,TLC显示反应完毕。浓缩,粗品经硅胶柱色谱纯化(PE∶EA=30∶1v/v)得到产品(3.5g,产率:99.2%)。
1HNMR(400MHz,CDCl 3)δ(ppm):9.22(s,1H),6.95(s,1H),6.67(s,1H),4.30-4.41(m,2H),2.51(s,3H),1.42-1.37(m,3H).
步骤4:6-(环丙基甲基)-2-甲基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯的合成
Figure PCTCN2019076129-appb-000236
将2-甲基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(3.0g,14.35mmol,1.0eq)、碳酸铯(11.4g,35mmol,2.5eq)和环丙基甲基溴(2.5g,19mmol,1.3eq)溶于乙腈(100mL),回流1小时,TLC显示反应完毕。反应液浓缩,加入水(30mL)和乙酸乙酯(200mL),有机相干燥,浓缩,粗品经硅胶柱色谱纯化(石油醚∶乙酸乙酯=100∶1v/v)得产品(3.5g,产率:92.6%)。
1H NMR(400MHz,CDCl 3)δ(ppm):7.06(s,1H),6.67(s,1H),4.29-4.36(m,4H),2.51(s,3H),1.42-1.37(m,4H),0.55-0.58(m,2H),0.45-0.48(m,2H).
分子式:C 14H 17NO 2S  分子量:263.10  LC-MS(Pos,m/z)=264.23[M+H] +.
步骤5:(6-(环丙基甲基)-2-甲基-6H-噻吩并[2,3-b]吡咯-5-基)甲醇的合成
Figure PCTCN2019076129-appb-000237
将四氢铝锂(0.78g,20.5mmol,1.5eq)悬浮于四氢呋喃(40mL)中,冷却至0℃,将6-(环丙基甲基)-2-甲基-6H-噻吩并[2,3-b]吡咯-5-羧酸乙酯(3.6g,13.67mmol,1.0eq)溶于四氢呋喃(20mL),缓慢加入到四氢铝锂中,维持反应体系温度在0℃左右。加毕,继续搅拌2小时,TLC显示反应完毕。依次缓慢加入水(1.3g)、15w/w%氢氧化钠水溶液(1.3g)和水(3.9g),加入无水硫酸钠干燥,过滤,滤液浓缩得产品(3g,产率:99.1%)。
步骤6:6-(环丙基甲基)-2-甲基-6H-噻吩并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000238
将(6-(环丙基甲基)-2-甲基-6H-噻吩并[2,3-b]吡咯-5-基)甲醇(200mg,0.9mmol,1.0eq)溶于四氢呋喃(3mL),加入二氧化锰(787mg,9.0mmol,10.0eq),室温搅拌18小时,TLC显示反应完毕,过滤,滤液浓缩得产品(280mg粗品),直接用于下步反应。
步骤7:(R)-(1-(2-(6-(环丙基甲基)-2-甲基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000239
将6-(环丙基甲基)-2-甲基-6H-噻吩并[2,3-b]吡咯-5-甲醛(280mg粗品)和(R)-(1-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(401mg,0.98mmol)溶于乙醇(4mL),将连二亚硫酸钠(870mg,5.0mmol)溶于水(2mL),加入反应液中。90℃反应4小时,TLC显示反应完毕。浓缩,加入乙酸乙酯(50mL)和水(10mL),有机相干燥,浓缩,粗品经硅胶柱色谱纯化(二氯甲烷∶甲醇=50∶1v/v)得到产品(450mg,产率:79.6%)。
步骤8:(R)-(3-氨基哌啶-1-基)(2-(6-(环丙基甲基)-2-甲基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000240
将(R)-(1-(2-(6-(环丙基甲基)-2-甲基-6H-噻吩并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-叔丁基-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯(420mg,0.73mmol,1.0eq)溶于二氯甲烷(4mL)和三氟乙酸(4mL),加入苯酚(342mg,3.64mmol,5.0eq),常温搅拌10分钟,TLC显示反应完毕,浓缩,加入二氯甲烷(100mL),用15w/w%氢氧化钠洗(20mL×2),有机相干燥,浓缩,加入乙酸乙酯(0.5mL)和石油醚(1mL),析出固体,过滤得产品(250mg,产率:71.8%)。
1H NMR(400MHz,CDCl 3)δ(ppm):7.40(s,1H),6.85(s,1H),6.72(s,1H),6.60(s,1H),4.22(d,J=7.0Hz,2H),4.14(s,3H),4.00(s,3H),2.61-3.19(m,4H),2.57(s,3H),1.98-2.09(m,1H),1.25-1.83(m,5H),0.42-0.52(m,2H),0.15-0.22(m,2H).
分子式:C 26H 31N 5O 2S  分子量:477.22  LC-MS(Pos,m/z)=478.32[M+H] +.
实施例27:(R)-(3-氨基哌啶-1-基)(2-(6-(环丙基甲基)-6H-呋喃并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮(化合物24)的合成
Figure PCTCN2019076129-appb-000241
步骤1:呋喃-3-基甲醇的合成
Figure PCTCN2019076129-appb-000242
将呋喃-3-羧酸(25g,0.22mmol,1.0eq)溶解于四氢呋喃(330mL),冰浴下缓慢滴加硼烷-四呋喃溶液(1mol/L,330mL),滴加完毕,常温搅拌2小时,TLC显示反应完毕,冰浴下缓慢滴加甲醇(40mL)淬灭反应。减压浓缩,加入乙酸乙酯(500mL),用2mol/L稀盐酸(40mL×2)洗,有机 相干燥,浓缩,粗品经硅胶柱色谱纯化(石油醚∶乙酸乙酯=4∶1v/v)得到产品(12g,产率:55.7%)。
步骤2:呋喃-3-甲醛的合成
Figure PCTCN2019076129-appb-000243
将呋喃-3-基甲醇(12.0g,0.12mol,1.0eq)溶于四氢呋喃(500mL),加入二氧化锰(156.4g,1.2mol,10.0eq),室温搅拌18小时,TLC显示反应完毕。过滤,滤液浓缩得产品(9.0g粗品),直接用于下步反应。
步骤3:2-叠氮基-3-(呋喃-3-基)丙烯酸乙酯的合成
Figure PCTCN2019076129-appb-000244
取钠(8.6g,0.38mol,4.0eq)溶解于乙醇(300mL)中,降温至-5℃。将呋喃-3-甲醛(9.0g,0.1mol,1.0eq)和叠氮乙酸乙酯(48.3g,0.38mol,4.0eq)溶解于乙醇(60mL)中,缓慢滴加至前述新制的乙醇钠溶液中,保持温度在-3℃左右,滴毕,保持温度在0℃以下,继续搅拌1小时。加入饱和氯化铵水溶液(30mL),将反应液倒入冰水中,搅拌十分钟,过滤,滤饼溶于乙酸乙酯,无水硫酸钠干燥,浓缩得到产品(8.4g,产率:40.6%)。
步骤4:6H-呋喃并[2,3-b]吡咯-5-羧酸乙酯的合成
Figure PCTCN2019076129-appb-000245
将2-叠氮基-3-(呋喃-3-基)丙烯酸乙酯(8.4g,0.04mol,1.0eq)溶于甲苯(200mL),回流1小时,TLC显示反应完毕。浓缩,粗品经硅胶柱色谱纯化(PE∶EA=30∶1v/v)得到产品(4.24g,产率:59.2%)。
分子式:C 9H 9NO 3  分子量:179.06  LC-MS(Pos,m/z)=180.23[M+H] +
1H NMR(400MHz,CDCl 3)δ(ppm):9.22(s,1H),6.85(s,1H),6.55(s,1H),4.30-4.41(m,2H),1.36-1.42(m,3H).
步骤5:6-(环丙基甲基)-6H-呋喃并[2,3-b]吡咯-5-羧酸乙酯的合成
Figure PCTCN2019076129-appb-000246
将6H-呋喃并[2,3-b]吡咯-5-羧酸乙酯(4.24g,0.024mol,1.0eq)、碳酸铯(20.0g,0.06mmol,2.5eq)和环丙基甲基溴(4.1g,0.03mmol,1.3eq)溶于乙腈(120mL),80℃回流1小时,TLC显示反应完毕。浓缩,加入水(30mL)和乙酸乙酯(200mL),有机相干燥,浓缩,粗品经硅胶柱色谱纯化(石油醚∶乙酸乙酯=100∶1v/v)得产品(5.3g粗品)。
1H NMR(400MHz,CDCl 3)δ(ppm):7.30(s,1H),6.92(s,1H),6.53(s,1H),4.21-4.36(m,4H),1.35-1.42(m,4H),0.51-0.58(m,2H),0.41-0.52(m,2H).
步骤6:(6-(环丙基甲基)-6H-呋喃并[2,3-b]吡咯-5-基)甲醇的合成
Figure PCTCN2019076129-appb-000247
将四氢铝锂(1.3g,34.1mmol,1.5eq)置于四氢呋喃(100mL)中,冷却至0℃,将6-(环丙基甲基)-6H-呋喃并[2,3-b]吡咯-5-羧酸乙酯(5.3g粗品)溶于四氢呋喃(20mL),缓慢加入到前述四氢铝锂悬浊液中,维持反应体系温度<-5℃。加毕,继续搅拌2小时,TLC显示反应完毕。依次缓慢加入水(1.3g)、15w/w%氢氧化钠水溶液(1.3g)和水(3.9g),加入无水硫酸钠干燥,过滤,滤液浓缩得产品(4.1g,两步产率:90.6%)。
步骤7:6-(环丙基甲基)-6H-呋喃并[2,3-b]吡咯-5-甲醛的合成
Figure PCTCN2019076129-appb-000248
将(6-(环丙基甲基)-6H-呋喃并[2,3-b]吡咯-5-基)甲醇(3g,0.016mol,1.0eq)溶于四氢呋喃(100mL),加入二氧化锰(13.6g,0.16mol,10.0eq),室温搅拌18小时,TLC显示反应完毕。过滤,滤液浓缩,粗品经硅胶 柱色谱纯化(二氯甲烷洗脱)得产品(330mg,产率:11.1%)。
步骤8:(R)-(1-(2-(6-(环丙基甲基)-6H-呋喃并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯的合成
Figure PCTCN2019076129-appb-000249
将6-(环丙基甲基)-6H-呋喃并[2,3-b]吡咯-5-甲醛(310mg,1.64mmol,1.0eq)和(R)-(1-(3-甲氧基-4-(甲氨基)-5-硝基苯甲酰基)哌啶-3-基)氨基甲酸叔丁酯(578mg,1.4mmol,1.0eq)溶于乙醇(2mL),将连二亚硫酸钠(1.2g,7.0mmol,5.0eq)溶于水(2mL)中,加入到反应液中。90℃反应4小时,TLC显示反应完毕。浓缩,加入乙酸乙酯(50mL)和水(10mL),有机相干燥,浓缩,粗品经硅胶柱色谱纯化(二氯甲烷∶甲醇=50∶1v/v)得到产品(130mg,产率:14.5%)。
步骤9:(R)-(3-氨基哌啶-1-基)(2-(6-(环丙基甲基)-6H-呋喃并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-基)甲酮的合成
Figure PCTCN2019076129-appb-000250
将(R)-(1-(2-(6-(环丙基甲基)-6H-呋喃并[2,3-b]吡咯-5-基)-7-甲氧基-1-甲基-1H-苯并[d]咪唑-5-羰基)哌啶-3-基)氨基甲酸叔丁酯(110mg,0.2mmol)溶于二氯甲烷(2mL),冰浴下加入2,6-二甲基吡啶(214.0mg,2.0mmol,10eq)和三氟甲磺酸三甲基硅酯(222mg,1.0mmol,5.0eq),常温搅拌6小时,TLC显示反应完毕。低温浓缩,粗品经硅胶柱色谱纯化(二 氯甲烷∶甲醇=40∶1v/v)得产品(39.0mg,产率:43.6%)。
1H NMR(400MHz,DMSO-d 6)δ(ppm):7.61(d,J=1.96,1H),7.28(s,1H),6.83(s,1H),6.72(d,J=1.96,1H),6.64(s,1H),4.25(d,J=7.0Hz,2H),4.11(s,3H),3.97(s,3H),3.35(s,4H),1.88-1.97(m,1H),1.60-1.79(m,1H),1.30-1.49(m,2H),1.15-1.29(m,2H),0.38-0.49(m,2H),0.19-0.28(m,2H).
分子式:C 25H 29N 5O 3  分子量:447.23  LC-MS(Pos,m/z)=448.32[M+H] +.
根据下述实验例,可以更好地理解本发明。然而,本领域的技术人员容易理解,实验例所描述的内容仅用于说明本发明,而不应当也不会限制权利要求书中所详细描述的本发明。
实验例一:测定化合物的酶抑制活性测试
测试物:本发明化合物,按照实施例方法制备
(1)试剂与耗材
肽酰精氨酸脱亚胺酶4(PAD4):Cayman,Cat.No.10500,Lot.No.0519694;
羧基荧光素标记肽(Peptide FAM-AcH4(1-5)):GL Biochem,Cat.No.0200046,Lot.No.P161110-TL545976;
二甲基亚砜(DMSO):Sigma,Cat.No.D2650,Lot.No.474382;
乙二胺四乙酸(EDTA):Sigma,Cat.No.E5134,CAS No.60-00-4;
96孔板:Corning,Cat.No.3365,Lot.No.22008026;
384孔板:Corning,Cat.No.3573,Lot.No.12608008。
(2)5×浓度化合物配制
取待测化合物适量,DMSO溶解至10mM。取适量10mM待测化合物母液用DMSO稀释至1.5mM溶液,然后用DMSO进行3倍梯度稀释,得到50×浓度化合物梯度溶液。最后用反应缓冲液(含有0.01%v/v Triton的100mM HEPES(4-羟乙基哌嗪乙硫磺酸)的水溶液,其pH值 为8.0)进行10倍稀释,得到5×浓度化合物梯度溶液(150、50、16.667、5.556、1.852、0.617、0.206、0.069、0.023和0.008μM)。
(3)溶液配制
1)反应缓冲液:含有0.01%v/v Triton的100mM HEPES(4-羟乙基哌嗪乙硫磺酸)的水溶液,其pH值为8.0;
2)终止液:含有0.015%w/v Brij-35、0.2%v/v Coating Reagent(包被液)#3(PE,部件号760050)和50mM EDTA的100mM HEPES(4-羟乙基哌嗪乙硫磺酸)的水溶液,其pH值为8.0;
3)2.5×酶溶液:取PAD4酶适量,用反应缓冲液稀释至2.5×(终浓度10nM);
4)2.5×多肽溶液:取羧基荧光素标记肽(FAM-AcH4(1-5)peptide)和CaCl 2溶液适量,用反应缓冲液稀释至2.5×(多肽终浓度5μM,Ca 2+终浓度0.1mM)。
(4)实验方法
首先向384孔板中加入5μL不同浓度的5×化合物溶液或溶剂,然后加入10μL 2.5×酶溶液,室温孵育10min。然后每孔加入10μL 2.5×多肽溶液,28℃孵育一定时间,加入25μL终止液,终止反应。用Caliper进行检测。抑制率按照如下公式计算:
抑制百分比=(max-转化)/(max-min)*100。
“max”表示DMSO对照;“min”表示低对照。
实验结果
表1
测试物 PAD4 IC 50(μM)
化合物GSK199 0.14
化合物3 0.20
化合物4 0.35
化合物9 0.11
化合物18 0.036
化合物20 0.058
化合物21 0.055
化合物22 0.04
化合物23 0.025
化合物24 0.054
由表1实验结果可见,本发明的化合物对PAD4酶具有良好的抑制活性,对于治疗由PAD4酶异常介导的疾病方面具有较好的临床应用潜力。
实验例二:本发明化合物的人/犬肝微粒体稳定性评价
温孵体系的构成:
Figure PCTCN2019076129-appb-000251
受试物配制:
精确称取适量化合物,用DMSO溶解配成5.0mM储备液。将5.0mM的储备液,用DMSO稀释成1.0mM,最后用水稀释成10μM的化合物工作溶液,待用(反应体系中DMSO含量为0.1v/v%)。
试验步骤:
(1).从-80℃冰箱中取出肝微粒体(20mg蛋白/mL),置于37℃水浴恒温振荡器上预温孵3min,融化待用。
(2).按照上面“温孵体系的构成”比例,制备温孵体系混合溶液(不含化合物和β-NADPH),置于37℃水浴恒温振荡器上预孵育2min。
3).对照组(不含β-NADPH):分别取30μL水和30μL化合物工作溶液(10μM)加入到240μL步骤(2)所述温孵体系混合溶液中,涡旋30s,混匀,反应总体积300μL,平行样2份。放入到37℃水浴恒温振荡器中进行孵育,并开始计时,取样时间点为0min和60min。
4).样品组:分别取70μL β-NADPH溶液(10mM)和70μL化合物工作溶液(10μM)加入560μL步骤(2)所述温孵体系混合溶液中,反应总体积700μL,涡旋30s,混匀,平行样2份。放入到37℃水浴恒温振荡器中进行孵育,并开始计时,取样时间点为计时后0min、5min、10min、20min、30min和60min。
(5).涡旋3min后,12000rpm离心5min。
(6).取上清液50μL加入150μL水,涡旋混匀,LC/MS/MS进样分析。
数据分析:
用下列一级动力学公式计算半衰期(t 1/2)和清除率(Cl):
Ct=C 0*e -kt
t 1/2=ln2/k=0.693/k
Cl int=V d*k
V d=1/肝微粒体中蛋白含量
注:k为化合物剩余量的对数与时间作图的斜率,V d为表观分布容积,C 0表示0h药物浓度,Ct为t时刻药物浓度,t为时间,e为自然数,Cl int为固有清除率。
结果:
表2 本发明化合物的人肝微粒体稳定性实验
Figure PCTCN2019076129-appb-000252
表3 本发明化合物的犬肝微粒体稳定性实验
Figure PCTCN2019076129-appb-000253
由表2-3可知,本发明化合物在人和犬肝微粒体中稳定性较好,清 除率较低。
实验例三:化合物的大鼠药代动力学(PK)评价
动物给药及样品采集:
实验用化合物用5v/v%DMSO+20v/v%(30v/v%solutol(聚乙二醇-15羟基硬脂酸酯,购自BASF,批号84773247G0))+75v/v%盐水溶解制备溶液剂,将化合物的溶液剂以5.0mg/kg的剂量灌胃给予SD大鼠,采血时间点为:15min、30min、1h、2h、4h、6h、8h和24h。
实验用化合物用5v/v%DMSO+20v/v%(30v/v%solutol)+75v/v%盐水溶解制备溶液剂,将化合物的溶液剂以1mg/kg的剂量静脉推注给予SD大鼠,采血时间点为:5min、15min、30min、1h、2h、4h、6h、8h和24h。
固定动物,每个时间点前10min用水浴锅加热尾部,通过尾静脉采集100μL左右的血液,血液采集后放置到含有EDTA-K 2的抗凝管中。血液样品在4℃条件下8000rpm离心6min得到血浆样品,血浆在血液采集后的30min内制备。血浆测试前存放在-80℃冰箱内。
样品分析方法:
从-80℃冰箱中取出待测样品,室温自然融化后涡旋5min,精密吸取20μL血浆样品至1.5mL离心管中;加入200μL浓度为100ng/mL的内标工作溶液(甲苯磺丁脲的甲醇溶液),混匀;涡旋5min后,12000rpm离心5min;精密吸取50μL上清液至预先加有150μL水/孔的96孔板中;涡旋混匀5min,进行LC-MS/MS测定分析。
数据处理方法:
受试物浓度使用AB公司的Analyst 1.6.3输出结果。采用Microsoft Excel计算均值、标准差、变异系数等参数(Analyst 1.6.3直接输出的不用计算),PK参数采用Pharsight Phoenix 6.1软件NCA计算(T max为中位数)。
结果:
表4 化合物在SD大鼠体内的PK参数(IV:1mg/kg,PO:5mg/kg, n=3)
Figure PCTCN2019076129-appb-000254
注:t z1/2:末端消除半衰期;Cl _obs:清除率;V z_obs:表观分布容积;T max:血药浓度达峰时间;AUC last:药-时曲线下面积0~24h;F%:绝对生物利用度;-:未测。
由表4可知,本发明化合物和对照药GSK199相比具有较高的暴露量和低清除率。
实验例四:本发明化合物的小鼠/大鼠肝微粒体稳定性评价
目的:评价本发明化合物的小鼠/大鼠肝微粒体稳定性。
温孵体系的构成:
Figure PCTCN2019076129-appb-000255
化合物3、化合物9和化合物18的配制:
精确称取适量化合物,用DMSO溶解配成5.0mM储备液。将5.0mM的储备液,用DMSO稀释成1.0mM,最后用水稀释成10μM的化合物工作溶液,待用(反应体系中DMSO含量为0.1v/v%)。
化合物20、化合物21、化合物22和化合物23的配制:
精确称取适量化合物,用甲醇溶解配成5.0mM储备液。将5.0mM的储备液,用甲醇稀释成1.0mM,最后用10%甲醇-水(v/v)溶液稀释成10μM的化合物工作溶液,待用(反应体系中甲醇含量为1.1v/v%)。
试验步骤:
(1).从-80℃冰箱中取出肝微粒体(20mg蛋白/mL),置于37℃水浴恒温振荡器上预温孵3min,融化待用。
(2).按照上面“温孵体系的构成”比例,制备温孵体系混合溶液(不含化合物和β-NADPH),置于37℃水浴恒温振荡器上预孵育2min。
3).对照组(不含β-NADPH):分别取30μL水和30μL化合物工作溶液(10μM)加入到240μL步骤(2)所述温孵体系混合溶液中,涡旋30s,混匀,反应总体积300μL,平行样2份。放入到37℃水浴恒温振荡器中进行孵育,并开始计时,取样时间点为0min和60min。
4).样品组:分别取70μL β-NADPH溶液(10mM)和70μL化合物工作溶液(10μM)加入560μL步骤(2)所述温孵体系混合溶液中,反应总体积700μL,涡旋30s,混匀,平行样2份。放入到37℃水浴恒温振荡器中进行孵育,并开始计时,取样时间点为计时后0min、5min、10min、20min、30min和60min。
(5).涡旋3min后,12000rpm离心5min。
(6).取上清液50μL加入150μL水,涡旋混匀,LC/MS/MS进样分析。
数据分析:
用下列一级动力学公式计算半衰期(t 1/2)和清除率(Cl):
Ct=C 0*e -kt
t 1/2=ln2/k=0.693/k
Cl int=V d*k
Vd=1/肝微粒体中蛋白含量
注:k为化合物剩余量的对数与时间作图的斜率,V d为表观分布容积,C 0表示0h药物浓度,Ct为t时刻药物浓度,t为时间,e为自然数,Cl int为固有清除率。
结果:
表5 本发明化合物的小鼠和大鼠肝微粒体稳定性实验
Figure PCTCN2019076129-appb-000256
“-”表示未测。
由表5可知,本发明化合物在小鼠和大鼠肝微粒体中稳定性较好,清除率较低。

Claims (14)

  1. 通式(I)表示的化合物或其药学上可接受的盐、立体异构体和互变异构体:
    Figure PCTCN2019076129-appb-100001
    其中,X和Y分别独立地为CR 6或者N;
    R 1为氢或C 1-6烷基;
    R 2为氢、C 1-6烷基、3-6元环烷基、C 1-6烷氧基、卤代C 1-6烷基或卤代C 1-6烷氧基;
    R 3为氢、氰基、C 1-6烷基、C 1-6烷氧基、C 1-6烷氧基C 1-6烷基、-L 1-Cy 1,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 1-6烷基羰基、C 1-6烷氧羰基、C 1-6烷基磺酰基、C 1-6烷氧基C 1-6烷基、C 1-6烷基氨基、(C 1-6烷基) 2氨基,L 1不存在或者为C 1-6亚烷基,Cy 1为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基,Cy 1可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
    R 4为氢、氰基、3-6元环烷基,或未被取代或被卤素、氰基、氨基、羟基、3-6元环烷基取代的C 1-6烷基、C 1-6烷基羰基、C 1-6烷氧羰基、C 1-6烷基磺酰基、C 1-6烷氧基C 1-6烷基、C 1-6烷基氨基、(C 1-6烷基) 2氨基;
    R 5为氢、卤素、氰基、氨基、羟基、-L 2-Cy 2,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 2-8烯基、C 1-6烷基羰基、C 1-6烷基羰基氨基、C 1-6烷氧基、C 1-6烷氧羰基、C 1-6烷基磺酰基、C 1-6烷基硫基、C 1-6烷基磺酰氨基、C 1-6烷基氨基、(C 1-6烷基) 2氨基,L 2不存在或者为C 1-6亚烷基、C 1-6亚烷氧基、C 2-6亚烯基、C 1-6亚烷氨基,Cy 2为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基,Cy 2可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
    R 6为氢、C 1-6烷基或C 3-6环烷基;
    R 7为氢或C 1-6烷基;
    R 8为氢、卤素、氰基、氨基、羟基、-L 3-Cy 3,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 2-8烯基、C 1-6烷基羰基、C 1-6烷基羰基氨基、C 1-6烷氧基、C 1-6烷氧羰基,L 3不存在或者为C 1-6亚烷基、C 2-6亚烯基,Cy 3为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基,Cy 3可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
    R 9为氢或C 1-6烷基;
    或者,R 8和R 9与所连接的N形成未被取代或被取代基取代的4-7元杂环基、6-11元并杂环基、7-12元螺杂环基、6-10元桥杂环基,所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基、C 1-6烷基氨基、(C 1-6烷基) 2氨基、-L 4-Cy 4,L 4不存在或者为C 1-6亚烷基,Cy 4为3-12元环烷基、3-12元环烯基、3-12元杂环基、芳基、5-10元杂芳基;
    环B为
    Figure PCTCN2019076129-appb-100002
    或5元杂芳基;
    其中,环B与
    Figure PCTCN2019076129-appb-100003
    的稠合,*端向上,#端向下,未指明*端和#端的,可以任意方向稠合;
    条件是,
    当环B为
    Figure PCTCN2019076129-appb-100004
    且R 8和R 9与所连接的N形成
    Figure PCTCN2019076129-appb-100005
    时,X为N;
    当环B为
    Figure PCTCN2019076129-appb-100006
    且X为CR 6时,R 8和R 9与所连接的N形成
    Figure PCTCN2019076129-appb-100007
    m为0-4的整数。
  2. 如权利要求1所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
    其中,X和Y分别独立地为CR 6或者N;
    R 1为氢或C 1-6烷基;
    R 2为氢、C 1-6烷基、C 1-6烷氧基、卤代C 1-6烷基或卤代C 1-6烷氧基;
    R 3为氢、氰基、C 1-6烷基、C 1-6烷氧基、C 1-6烷氧基C 1-6烷基,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基;
    R 4为氢、氰基、3-6元环烷基,或未被取代或被卤素、氰基、氨基、羟基、3-6元环烷基取代的C 1-6烷基;
    R 5为氢、卤素、氰基、氨基、羟基,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基、C 1-6烷氧基;
    R 6为氢或C 1-6烷基;
    R 7为氢或C 1-6烷基;
    R 8为氢、卤素、氰基、氨基、羟基、-L 3-Cy 3,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基,L 3不存在或者为C 1-6亚烷基,Cy 3为3-6元环烷基、3-12元杂环基、芳基、5-10元杂芳基,Cy 3可任选被氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
    R 9为氢或C 1-6烷基;
    或者,R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元杂环基、7-10元并杂环基、7-11元螺杂环基、7-10元桥杂环基,所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基;
    环B为5元杂芳基,优选地,环B为
    Figure PCTCN2019076129-appb-100008
    m为0-4的整数。
  3. 如权利要求2所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
    其中,X为CR 6,Y为N;
    R 1为氢或C 1-6烷基;
    R 2为氢、C 1-6烷基、C 1-6烷氧基;
    R 3为氢、C 1-6烷基;
    R 4为氢,或未被取代或被卤素、3-6元环烷基取代的C 1-6烷基;
    R 5为氢、卤素、C 1-6烷基、C 1-6烷氧基;
    R 6为氢或C 1-6烷基;
    R 7为氢或C 1-6烷基;
    R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元杂环基,所述取代基选自氨基;
    环B为5元杂芳基,优选地,
    Figure PCTCN2019076129-appb-100009
    m为0-4的整数。
  4. 如权利要求3所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
    R 8和R 9与所连接的N形成未被取代或被取代基取代的吡咯烷基、哌啶基、哌嗪基、吗啉基,所述取代基为氨基。
  5. 如权利要求2所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
    其中,X为CR 6,Y为N;
    R 8和R 9与所连接的N形成未被取代或被取代基取代的
    Figure PCTCN2019076129-appb-100010
    Figure PCTCN2019076129-appb-100011
    Figure PCTCN2019076129-appb-100012
    所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。
  6. 如权利要求2所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
    其中,X为CR 6,Y为N;
    R 8和R 9与所连接的N形成未被取代或被取代基取代的
    Figure PCTCN2019076129-appb-100013
    Figure PCTCN2019076129-appb-100014
    Figure PCTCN2019076129-appb-100015
    Figure PCTCN2019076129-appb-100016
    所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。
  7. 如权利要求2所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
    其中,X为CR 6,Y为N;
    R 8和R 9与所连接的N形成未被取代或被取代基取代的
    Figure PCTCN2019076129-appb-100017
    Figure PCTCN2019076129-appb-100018
    所述取代基选自氢、卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基。
  8. 如权利要求2所述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
    其中,X为CR 6,Y为N;
    R 8为氢、卤素、氰基、氨基、羟基、-L 3-Cy 3,或未被取代或被卤素、氰基、氨基、羟基取代的C 1-6烷基,L 3不存在或者为C 1-6亚烷基,Cy 3为3-6元环烷基、3-8元杂环基,Cy 3可任选被卤素、氰基、氨基、羟基、C 1-6烷基、C 1-6烷氧基取代;
    R 9为氢或C 1-6烷基。
  9. 如权利要求3述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
    其中,X为CR 6,Y为N;
    R 1为氢或C 1-6烷基;
    R 2为氢、C 1-6烷基、C 1-6烷氧基;
    R 3为氢、C 1-6烷基;
    R 4为氢,或未被取代或被卤素、3-6元环烷基取代的C 1-6烷基;
    R 5为氢、卤素、C 1-6烷基、C 1-6烷氧基;
    R 6为氢或C 1-6烷基;
    R 7为氢或C 1-6烷基;
    R 8为氢,或未被取代或被氨基取代的C 1-6烷基;
    R 9为氢或C 1-6烷基;
    或者,R 8和R 9与所连接的N形成未被取代或被取代基取代的5-6元饱和含氮杂环基,
    所述取代基选自氨基;
    环B为
    Figure PCTCN2019076129-appb-100019
    m为0或1。
  10. 如权利要求1述的化合物或其药学上可接受的盐、立体异构体和互变异构体,
    其中,X和Y分别独立地为CR 6或者N;
    R 1为氢或C 1-6烷基;
    R 2为氢、C 1-6烷基、C 1-6烷氧基;
    R 3为氢、C 1-6烷基;
    R 4为氢,或未被取代或被卤素、3-6元环烷基取代的C 1-6烷基;
    R 5为氢、卤素、C 1-6烷基、C 1-6烷氧基;
    R 6为氢或C 1-6烷基;
    R 7为氢或C 1-6烷基;
    R 8为氢、-L 3-Cy 3,或未被取代或被氨基取代的C 1-6烷基,L 3不存在,Cy 3为任选被氨基取代的3-6元环烷基;
    R 9为氢或C 1-6烷基;
    或者,R 8和R 9与所连接的N形成未被取代或被取代基取代的4-7元杂环基、7-12元螺杂环基,所述取代基为氨基;
    环B为
    Figure PCTCN2019076129-appb-100020
    或5元杂芳基;
    其中,环B与
    Figure PCTCN2019076129-appb-100021
    的稠合,*端向上,#端向下,未指明*端和#端的,可以任意方向稠合;
    条件是,
    当环B为
    Figure PCTCN2019076129-appb-100022
    且R 8和R 9与所连接的N形成
    Figure PCTCN2019076129-appb-100023
    时,X为N;
    当环B为
    Figure PCTCN2019076129-appb-100024
    且X为CR 6时,R 8和R 9与所连接的N形成
    Figure PCTCN2019076129-appb-100025
    m为0或1。
  11. 如下所述的化合物或其药学上可接受的盐、立体异构体和互变异构体:
    Figure PCTCN2019076129-appb-100026
    Figure PCTCN2019076129-appb-100027
  12. 含有权利1-11任一项所述的一种或多种化合物或其药学上可接受的盐、立体异构体和互变异构体的药物组合物,其可以任选含有一种或多种药用载体并任选被制成药学上可接受的任一剂型。
  13. 权利要求1-11任一项所述的化合物或其药学上可接受的盐、立体异构体和互变异构体或权利要求12所述的药物组合物在制备治疗或者预防由肽酰精氨酸脱亚胺酶PAD4介导的疾病的药物中的用途。
  14. 权利要求13所述的用途,肽酰精氨酸脱亚胺酶PAD4介导的疾病选自类风湿性关节炎、血管炎、系统性红斑狼疮、溃疡性结肠炎、多发性硬化症、囊性纤维化、癌症、皮肤型红斑狼疮、哮喘和银屑病。
PCT/CN2019/076129 2018-02-26 2019-02-26 肽酰精氨酸脱亚胺酶抑制剂及其用途 WO2019161803A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP19757537.6A EP3760628A4 (en) 2018-02-26 2019-02-26 PEPTIDYLARGININE DEIMINASE INHIBITOR AND ITS USES
US16/975,920 US20200407370A1 (en) 2018-02-26 2019-02-26 Peptidylarginine deiminase inhibitor and use thereof
JP2020568018A JP2021515043A (ja) 2018-02-26 2019-02-26 ペプチジルアルギニンデイミナーゼ阻害剤及びその使用

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201810158900 2018-02-26
CN201810158900.7 2018-02-26
CN201811512419.X 2018-12-11
CN201811512419 2018-12-11

Publications (1)

Publication Number Publication Date
WO2019161803A1 true WO2019161803A1 (zh) 2019-08-29

Family

ID=67686997

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/076129 WO2019161803A1 (zh) 2018-02-26 2019-02-26 肽酰精氨酸脱亚胺酶抑制剂及其用途

Country Status (5)

Country Link
EP (1) EP3760628A4 (zh)
JP (1) JP2021515043A (zh)
CN (1) CN110194770B (zh)
TW (1) TWI762769B (zh)
WO (1) WO2019161803A1 (zh)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020033490A1 (en) * 2018-08-08 2020-02-13 Bristol-Myers Squibb Company Substituted thienopyrroles as pad4 inhibitors
WO2021057910A1 (zh) * 2019-09-27 2021-04-01 南京药捷安康生物科技有限公司 肽酰精氨酸脱亚胺酶抑制剂及其用途
WO2021174024A1 (en) 2020-02-28 2021-09-02 First Wave Bio, Inc. Methods of treating iatrogenic autoimmune colitis
US11878965B2 (en) 2020-12-22 2024-01-23 Gilead Sciences, Inc. Inhibitors of peptidylarginine deiminases
US11976083B2 (en) 2020-04-30 2024-05-07 Gilead Sciences, Inc. Inhibitors of peptidylarginine deiminases

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111440141A (zh) * 2020-05-20 2020-07-24 安徽泽升科技有限公司 一种羟甲基取代芳杂环类化合物的制备方法

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014015905A1 (en) 2012-07-26 2014-01-30 Glaxo Group Limited 2 - (azaindol- 2 -yl) benz imidazoles as pad4 inhibitors
WO2017100594A1 (en) * 2015-12-09 2017-06-15 Padlock Therapeutics, Inc. Bicyclic inhibitors of pad4

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012061390A2 (en) * 2010-11-01 2012-05-10 The Penn State Research Foundation Therapeutic compositions and methods
CN105884828A (zh) * 2015-02-16 2016-08-24 上海迪诺医药科技有限公司 多环化合物、其药物组合物及应用

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014015905A1 (en) 2012-07-26 2014-01-30 Glaxo Group Limited 2 - (azaindol- 2 -yl) benz imidazoles as pad4 inhibitors
CN104470919A (zh) * 2012-07-26 2015-03-25 葛兰素集团有限公司 作为pad4抑制剂的2-(氮杂吲哚-2-基)苯并咪唑
WO2017100594A1 (en) * 2015-12-09 2017-06-15 Padlock Therapeutics, Inc. Bicyclic inhibitors of pad4

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, 5 July 2017, Columbus, Ohio, US; abstract no. Registry, "STN" *
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 60-00-4
CHEMICAL ABSTRACTS, no. 2101329-03-5, Columbus, Ohio, US; abstract no. Registry, STN *
See also references of EP3760628A4

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020033490A1 (en) * 2018-08-08 2020-02-13 Bristol-Myers Squibb Company Substituted thienopyrroles as pad4 inhibitors
JP2021534106A (ja) * 2018-08-08 2021-12-09 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Pad4阻害剤としての置換チエノピロール
JP7447080B2 (ja) 2018-08-08 2024-03-11 ブリストル-マイヤーズ スクイブ カンパニー Pad4阻害剤としての置換チエノピロール
US11981680B2 (en) 2018-08-08 2024-05-14 Bristol-Myers Squibb Company Substituted thienopyrroles as PAD4 inhibitors
WO2021057910A1 (zh) * 2019-09-27 2021-04-01 南京药捷安康生物科技有限公司 肽酰精氨酸脱亚胺酶抑制剂及其用途
WO2021174024A1 (en) 2020-02-28 2021-09-02 First Wave Bio, Inc. Methods of treating iatrogenic autoimmune colitis
US11976083B2 (en) 2020-04-30 2024-05-07 Gilead Sciences, Inc. Inhibitors of peptidylarginine deiminases
US11878965B2 (en) 2020-12-22 2024-01-23 Gilead Sciences, Inc. Inhibitors of peptidylarginine deiminases

Also Published As

Publication number Publication date
TW201936605A (zh) 2019-09-16
EP3760628A4 (en) 2021-10-13
EP3760628A1 (en) 2021-01-06
TWI762769B (zh) 2022-05-01
CN110194770B (zh) 2022-08-09
CN110194770A (zh) 2019-09-03
JP2021515043A (ja) 2021-06-17

Similar Documents

Publication Publication Date Title
WO2019161803A1 (zh) 肽酰精氨酸脱亚胺酶抑制剂及其用途
TWI363628B (en) Certain chemical entities, compositions, and methods
TW202003511A (zh) 作為免疫調節劑之雜環化合物
JP2020509041A (ja) アミド類誘導体阻害剤及びその製造方法と使用
CN105658643A (zh) Ido抑制剂
WO2019094920A1 (en) Azepin-2-one derivatives as rsv inhibitors
JP7447080B2 (ja) Pad4阻害剤としての置換チエノピロール
WO2019062733A1 (zh) Pde9 抑制剂及其用途
CN105473550A (zh) Ido抑制剂
WO2020011246A1 (zh) 含苯环的化合物、其制备方法及应用
CN111560012B (zh) 一种作为irak抑制剂的化合物
WO2005085245A1 (en) Pyrrolopyridine-2-carboxylic acid hydrazides
KR20140094547A (ko) 1-(5,6-다이클로로-1h-벤조[d]이미다졸-2-일)-1h-피라졸-4-카르복실산의 메글루민 염 제형
JP2021519308A (ja) 転写活性化タンパク質のイミダゾピペラジン阻害剤
CN112292374B (zh) 一种新型磷酸肌醇3-激酶抑制剂及其制备方法和用途
JP2021534108A (ja) Pad酵素のインドールおよびアザインドール阻害剤
CN112574230B (zh) 肽酰精氨酸脱亚胺酶抑制剂及其用途
WO2020143763A1 (zh) 卤代烯丙基胺类化合物及其应用
BR112019026452A2 (pt) compostos químicos como inibidores de h-pgds
WO2019029541A1 (zh) 成纤维细胞生长因子受体抑制剂及其用途
CN117279636A (zh) 用于调节强直性肌营养不良1型的方法和化合物
WO2022073470A1 (zh) 杂环取代的稠合γ-咔啉类衍生物、其制备方法、中间体及应用
BR112021008976A2 (pt) compostos químicos
CN114790164B (zh) 一种取代的异吲哚啉-1,3-二酮类pde4抑制剂及其药物用途
WO2023125121A1 (zh) 一类三并环化合物、其制备方法及用途

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19757537

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2020568018

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019757537

Country of ref document: EP

Effective date: 20200928