WO2023160527A1 - 嘧啶并芳香环类化合物及其在药物中的应用 - Google Patents

嘧啶并芳香环类化合物及其在药物中的应用 Download PDF

Info

Publication number
WO2023160527A1
WO2023160527A1 PCT/CN2023/077368 CN2023077368W WO2023160527A1 WO 2023160527 A1 WO2023160527 A1 WO 2023160527A1 CN 2023077368 W CN2023077368 W CN 2023077368W WO 2023160527 A1 WO2023160527 A1 WO 2023160527A1
Authority
WO
WIPO (PCT)
Prior art keywords
butyl
methyl
amino
cancer
alkyl
Prior art date
Application number
PCT/CN2023/077368
Other languages
English (en)
French (fr)
Inventor
任青云
刘为顺
张英俊
颜光华
Original Assignee
广东东阳光药业股份有限公司
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 广东东阳光药业股份有限公司 filed Critical 广东东阳光药业股份有限公司
Publication of WO2023160527A1 publication Critical patent/WO2023160527A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • 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
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • 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

Definitions

  • the invention belongs to the field of medicine. It specifically relates to a pyrimido aromatic ring compound and its use as a medicine, especially the use as a TLR8 agonist.
  • the present invention also relates to the composition of these pyrimidoaromatic ring compounds and other therapeutic agents, and their use as medicine, especially the use as TLR8 agonist.
  • TLRs Toll-like receptors
  • PAMPs such as lipopolysaccharide, flagellin, single/double-stranded RNA, etc.
  • different TLRs can induce gene expression in specific functional regions, thereby stimulating the body's antigens Specific acquired immune response.
  • TLR8 is a member of a subgroup of TLRs (TLRs 3, 7, 8, and 9) that is restricted to the endosomal compartment of cells that specifically recognize non-self nucleic acids. TLR8 is mainly expressed by monocytes, NK cells and myeloid dendritic cells (mDC) in humans. TLR8 agonists can lead to the release of various pro-inflammatory cytokines, such as IL-6, IL-12, TNF- ⁇ and IFN- ⁇ .
  • TLR8 After TLR8 activation, it mediates inflammatory immunity and promotes the clearance of virus-infected cells and tumor cells in the body. Its agonists can be used as independent immunotherapy drugs or immune adjuvants, showing important clinical application prospects in immunotherapy.
  • the activation of TLR8 is closely related to the anti-infection natural immune response, and can mediate the occurrence and development of HBV, HCV, HIV, herpes virus and other viral infection diseases, tumors, autoimmune diseases, and metabolic diseases.
  • TLR8 and TLR7 have been reported in many patents.
  • TLR8 agonist drugs In view of the extensive therapeutic potential of TLR8 agonist drugs, there is still a need for new TLR8 agonist drugs, especially for the treatment and/or prevention of hepatitis B virus. Agonist drugs with high selectivity for TLR8.
  • the present invention relates to novel pyrimidoaromatic ring compounds and pharmaceutically acceptable compositions thereof, which have a better activation effect on TLR8 and a better selective activation effect on TLR8;
  • the drug enzyme has basically no induction effect, basically no inhibitory effect on the liver drug enzyme, and basically no toxicity to the heart. In addition, they have good solubility, good stability and very good pharmacokinetic properties.
  • the compound of the present invention can treat and/or prevent various diseases related to TLR8 activity, and especially has a good application prospect in anti-hepatitis B virus (HBV).
  • HBV anti-hepatitis B virus
  • the present invention relates to a compound as shown in formula (I) or stereoisomers, tautomers, nitrogen oxides, solvates, metabolites, pharmaceutical acceptable salt or its prodrugs,
  • X is N or CR 4 ;
  • Each of R 1 , R 2 , R 4 , R 5 , R 6 , R 7 and R 8 is independently hydrogen, deuterium, F, Cl, Br, I, hydroxyl, cyano, amino, C 1-4 alkylamino, C 1-6 alkoxy or C 1-6 alkyl, wherein the C 1-4 alkylamino, C 1-6 alkoxy and C 1-6 alkyl are each independently unsubstituted or replaced by 1 , 2, 3 or 4 substituents independently selected from F, Cl, Br, I, hydroxyl, cyano, amino and C 1-4 alkyl are substituted;
  • Y is O or S
  • R 3 is -C 1-6 alkylene-R 10 , wherein the -C 1-6 alkylene- in the -C 1-6 alkylene-R 10 is unsubstituted or replaced by 1, 2 , 3, 4 or 5 substituents of R w1 are substituted;
  • Each R c is independently hydrogen, deuterium or C 1-4 alkyl, wherein said C 1-4 alkyl is unsubstituted or is independently selected from F, Cl, Br by 1, 2, 3 or 4 , I, hydroxyl, cyano, amino and C 1-4 alkyl are substituted by substituents;
  • Each R 10a and R 10b is independently a C 1-6 alkyl group, a C 3-7 cycloalkyl group, a heteroaryl group consisting of 5-12 ring atoms or a heterocyclyl group consisting of 3-12 ring atoms, wherein, The C 1-6 alkyl group, C 3-7 cycloalkyl group, heteroaryl group consisting of 5-12 ring atoms and heterocyclic group consisting of 3-12 ring atoms are each independently unsubstituted or replaced by 1 , 2, 3, 4 or 5 substituents of R w2 are substituted;
  • R is H, deuterium, C 1-6 alkyl or a heterocyclic group consisting of 3-6 ring atoms, wherein the C 1-6 alkyl and 3-6 ring atoms are independently Unsubstituted or substituted by 1, 2, 3, 4 or 5 R substituents;
  • R 9 is hydrogen, deuterium, -LR 11 , F, Cl, Br, I, hydroxyl, cyano, amino, C 1-4 alkylamino, C 1-6 alkoxy or C 1-6 alkyl, wherein, The C 1-4 alkylamino, C 1-6 alkoxy and C 1-6 alkyl are each independently unsubstituted or 1, 2, 3 or 4 independently selected from F, Cl, Br, 1, hydroxyl, cyano, amino and C 1-4 alkyl substituents are substituted;
  • the aryl group and the heterocyclic group consisting of 3-12 ring atoms are each independently unsubstituted or substituted by 1, 2, 3, 4 or 5 R substituents;
  • R a , R b and the N atoms connected to them together form a heterocyclic group composed of 3-8 ring atoms, wherein the heterocyclic group composed of 3-8 ring atoms is unsubstituted or is 1, 2, 3 or 4 R w4 substitutions;
  • Each of R 9a , R 9b , R 9c and R 9d is independently C 1-12 alkyl, C 2-12 alkenyl, C 2-12 alkynyl or -C 1-6 alkylene-R d2 , wherein, The -C 1-6 alkylene in the C 1-12 alkyl, C 2-12 alkenyl, C 2-12 alkynyl and -C 1-6 alkylene-R d2 are independently not is substituted or substituted by 1, 2, 3, 4 or 5 R substituents ;
  • Each of R d1 and R d2 is independently hydroxyl, C 1-6 alkoxy, C 6-10 aryl, heteroaryl consisting of 5-12 ring atoms, C 3-6 cycloalkyl or C 1-6 Alkylamino, wherein, the C 1-6 alkoxyl group, C 6-10 aryl group, heteroaryl group consisting of 5-12 ring atoms, C 3-6 cycloalkyl group and C 1-6 alkylamino group respectively independently unsubstituted or substituted by 1, 2, 3, 4 or 5 R substituents ;
  • a heterocyclic group consisting of 5-6 ring atoms, a heteroaryl group or a C 6-10 aryl group, wherein, the amino group, C 1-6 alkylamino group, C 1-6 alkoxy group, C 1-6 6 alkyl, C 1-6 haloalkyl, -C( O)-C 1-4 alkyl, C 2-12 alkenyl, C 2-12 alkynyl, C 3-6 cycloalkyl, 3-6 The heterocyclic group consisting of 5-6 ring atoms,
  • t 0, 1 or 2.
  • each of R 1 , R 2 , R 4 , R 5 , R 6 , R 7 and R 8 is independently hydrogen, deuterium, F, Cl, Br, I, hydroxyl, cyano, amino, N-methylamino, N-ethylamino, N,N-dimethylamino, N,N-diethylamino, methoxy, ethoxy, 1-propoxy, 2-propoxy, 1-butoxy base, 2-methyl-l-propoxy, 2-butoxy, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n- Pentyl or n-hexyl, wherein, the N-methylamino, N-ethylamino, N,N-dimethylamino, N,N-diethylamino, methoxy, ethoxy, 1-propoxy , 2-propoxy,
  • R is H, deuterium, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, aziridine, nitrogen Heterocyclobutyl, oxetanyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrofuryl, tetrahydrothiophenyl, tetrahydropyranyl, tetrahydrothiopyranyl, Piperidinyl, morpholinyl, thiomorpholinyl or piperazinyl, wherein, the methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert Butyl, n-pentyl
  • each R w3 has the meaning as described in the present invention.
  • R 3 is -CH 2 -R 10 , -(CH 2 ) 2 -R 10 , -(CH 2 ) 3 -R 10 , -CH(CH 3 )CH 2 -R 10 , - CH 2 CH(CH 3 )-R 10 or -(CH 2 ) 4 -R 10 , wherein -CH 2 - in -CH 2 -R 10 and -(CH 2 ) 2 -R 10 in -(CH 2 ) 2 -R 10 -(CH 2 ) 2 -, -(CH 2 ) 3 - in -(CH 2 ) 3 -R 10 , -CH(CH 3 )CH 2 - in -CH(CH 3 )CH 2 -R 10 , -CH 2 CH(CH 3 )- in -CH 2 CH(CH 3 )-R 10 and -( CH 2 ) 4 -(CH 2 ) 4 in -R 10 are each independently unsubstituted or
  • Each R 10a and R 10b is independently a C 1-4 alkyl group, a C 3-6 cycloalkyl group, a heteroaryl group consisting of 5-6 ring atoms or a heterocyclyl group consisting of 3-6 ring atoms, wherein, The C 1-4 alkyl group, C 3-6 cycloalkyl group, heteroaryl group consisting of 5-6 ring atoms and heterocyclic group consisting of 3-6 ring atoms are each independently unsubstituted or replaced by 1 , 2, 3, 4 or 5 substituents of R w2 are substituted;
  • R c is hydrogen, deuterium, methyl, ethyl, n-propyl or isopropyl, wherein said methyl, ethyl, n-propyl and isopropyl are unsubstituted or 1, 2, 3 or 4 substituents independently selected from F, Cl, Br, I, hydroxyl, cyano, amino, methyl, ethyl, n-propyl and isopropyl are substituted;
  • each of R w1 and R w2 has the meaning described in the present invention.
  • each R 10a and R 10b is independently methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, cyclopropyl, Cyclobutyl, cyclopentyl, cyclohexyl, furyl, pyrrolyl, pyridyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, 1 ,3,5-Triazinyl, Thiazolyl, Thienyl, Pyrazinyl, Pyridazinyl, Pyrimidinyl, Aziridyl, Azetidinyl, Oxetanyl, Thietanyl, Pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrofuryl
  • each R w2 has the meaning as described in the present invention.
  • R 9 is hydrogen, deuterium, -LR 11 , F, Cl, Br, I, hydroxyl, cyano, amino, C 1-4 alkylamino, C 1-4 alkoxy or C 1 -4 alkyl, wherein the C 1-4 alkylamino, C 1-4 alkoxy and C 1-4 alkyl are each independently unsubstituted or are independently selected by 1, 2, 3 or 4 Substituents from F, Cl, Br, I, hydroxyl, cyano, amino and C 1-4 alkyl;
  • the aryl group and the heterocyclic group composed of 3-6 ring atoms are each independently unsubstituted or substituted by 1, 2, 3, 4 or 5 R substituents;
  • each of R a , R b , R 9a , R 9b , t, R 9c and R w4 has the meaning as described in the present invention.
  • R 9 is hydrogen, deuterium, -LR 11 , F, Cl, Br, I, hydroxyl, cyano, amino, N-methylamino, N-ethylamino, N,N-dimethylamino , N,N-diethylamino, methoxy, ethoxy, 1-propoxy, 2-propoxy, 1-butoxy, 2-methyl-l-propoxy, 2-butoxy base, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl or n-hexyl, wherein the N-methylamino, N -Ethylamino, N,N-dimethylamino, N,N-diethylamino, methoxy, ethoxy, 1-propoxy, 2-propoxy, 1-butoxy, 2-methyl -l-propoxy, 2-butoxy, methyl,
  • each of R a , R b , R 9a , R 9b , L, t, R 9c and R w4 has the meanings described in the present invention.
  • the -C 1-4 alkylene- each independently unsubstituted or substituted by 1, 2, 3, 4 or 5 substituents of R W5 ;
  • R a , R b and the N atoms connected to them together form a heterocyclic group composed of 3-6 ring atoms, wherein the heterocyclic group composed of 3-6 ring atoms is unsubstituted or replaced by 1, 2, 3 or 4 R w4 substitutions;
  • each of R 9d , R d1 , R w5 and R w4 has the meaning as described in the present invention.
  • R a , R b and the N atoms they are connected together form aziridinyl, azetidinyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, piperidinyl, morpholinyl, Thiomorpholinyl or piperazinyl, wherein, the aziridinyl, azetidinyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, piperidinyl, morpholinyl, thio Morpholinyl and piperazinyl are each independently unsubstituted or substituted by 1, 2, 3 or 4 R;
  • each of R 9d , R d1 , R w5 and R w4 has the meaning as described in the present invention.
  • each of R 9a , R 9b , R 9c and R 9d is independently C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl or -C 1-4 alkylene Group -R d2 , wherein the -C 1-4 alkylene in the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl and -C 1-4 alkylene -R d2 Group-each independently unsubstituted or substituted by 1, 2, 3, 4 or 5 R substituents ;
  • Each R d1 and R d2 are independently hydroxyl, C 1-4 alkoxy, phenyl, heteroaryl consisting of 5-6 ring atoms, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or C 1-4 alkylamino, wherein, the C 1-4 alkoxy, phenyl, heteroaryl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and C 1-4 alkylamino groups are each independently unsubstituted or substituted by 1, 2, 3, 4 or 5 R substituents ;
  • each of R d2 , R w6 and R w7 has the meaning as described in the present invention.
  • Each R d1 and R d2 are independently hydroxyl, methoxy, ethoxy, 1-propoxy, 2-propoxy, 1-butoxy, 2-methyl-l-propoxy, 2- Butoxy, phenyl, furyl, pyrrolyl, pyridyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, 1,3,5 -Triazinyl, thiazolyl, thienyl, pyrazinyl, pyridazinyl, pyrimidinyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, N-methylamino, N-ethylamino, N,N -Dimethylamino or N,N-diethylamino, wherein, the methoxy, ethoxy, 1-propoxy, 2-propoxy, 1-but
  • each of R d2 , R w6 and R w7 has the meaning as described in the present invention.
  • the present invention also provides a pharmaceutical composition, comprising the compound described in the present invention and pharmaceutically acceptable auxiliary materials.
  • the pharmaceutical composition of the present invention further comprises other one or more therapeutic agents, wherein the therapeutic agents are HBV DNA polymerase inhibitors, toll-like receptor 7 modulators, Toll-like receptor 8 modulators, toll-like receptor 7 and 8 modulators, toll-like receptor 3 modulators, interferon ⁇ ligands, HBsAg inhibitors, compounds targeting HbcAg, cyclophilin inhibitors , HBV therapeutic vaccine, HBV preventive vaccine, HBV virus entry inhibitor, NTCP inhibitor, antisense oligonucleotide targeting viral mRNA, short interfering RNA (siRNA), hepatitis B virus E antigen inhibitor, HBx Inhibitors, cccDNA inhibitors, HBV antibodies, thymosin agonists, cytokines, nucleoprotein inhibitors, stimulators of retinoic acid-inducible gene 1, NOD2 stimulators, recombinant thymosin ⁇ -1, hepatitis B virus replication inhibition agents,
  • the pharmaceutical composition of the present invention wherein the other one or more therapeutic agents are lamivudine, telbivudine, tenofovir, entecavir, adefovir dipivoxil , Tenofovir Alafenamide, Tenofovir Disoproxil, Tenofovir Alafenamide Fumarate, Tenofovir Alafenamide Hemifumarate, Alfaferone, Alloferon, Simox interleukin, clavudine, emtricitabine, famciclovir, interferon, Bao Ganling CP, interferon, interleukin-2, milvotidate, nitazoxanide, ribavirin, roxime Interferon-A, sizoran, Euforavac, Ampligen, Phosphazid, Heplisav, recombinant human interleukin-2, levamisole, or propagermanium.
  • the other one or more therapeutic agents are
  • the pharmaceutical composition of the present invention wherein the other one or more therapeutic agents are lamivudine, telbivudine, tenofovir, entecavir, adefovir dipivoxil , Tenofovir Alafenamide, Tenofovir Disoproxil, Tenofovir Alafenamide Fumarate, Tenofovir Alafenamide Hemifumarate, Alfaferone, Alloferon, Simoleukin, Clavudine, Emtricitabine, Famciclovir, Bao Ganling CP, Interferon, Interferon ⁇ -1b, Interferon ⁇ , Interferon ⁇ -2a, Interferon ⁇ -1a, Interferon ⁇ -2, interleukin-2, milfutilate, nitazoxanide, pegylated interferon ⁇ -2a, ribavirin, rosiferin-A, sizoran, Euforavac, ampligen , P
  • the present invention also provides the use of the compound or the pharmaceutical composition in activating TLR8.
  • the present invention also provides the use of the compound or the pharmaceutical composition in preparing a kit for activating TLR8.
  • the present invention also provides the use of the compound or the pharmaceutical composition in preparing medicines for preventing, treating, treating or alleviating diseases mediated by TLR8 in patients.
  • the disease mediated by TLR8 is hepatitis B virus infection, hepatitis C virus infection, influenza virus infection, herpes virus infection, HIV infection, allergic disease, rheumatoid arthritis, allergic asthma, chronic fatigue, type 2 diabetes, Hay fever, lupus erythematosus, multiple sclerosis, melanoma, lung cancer, liver cancer, basal cell carcinoma, kidney cancer, myeloma, biliary tract cancer, brain cancer, breast cancer, cervical cancer, choriocarcinoma, colon cancer, rectal cancer , head and neck cancer, peritoneal tumor, fallopian tube cancer, endometrial cancer, esophageal cancer, gastric cancer, leukemia, lymphoma, sarcoma, neuroblastoma, oral cancer, ovarian cancer, pancreatic cancer, prostate cancer, testicular cancer, skin cancer or Thyroid cancer.
  • the present invention also provides the use of the compound or pharmaceutical composition in the preparation of medicines for treating or preventing diseases of the immune regulatory system.
  • the present invention also provides the use of the compound or pharmaceutical composition in the preparation of medicines for treating or preventing viral infection or tumors.
  • the present invention also provides the preparation of the compound or the pharmaceutical composition for the treatment or prevention of hepatitis B virus infection, hepatitis C virus infection, influenza virus infection, herpes virus infection, HIV infection , allergic diseases, rheumatoid arthritis, allergic asthma, chronic fatigue, type II diabetes, hay fever, lupus erythematosus, multiple sclerosis, melanoma, lung cancer, liver cancer, basal cell carcinoma, kidney cancer, myeloma, Biliary tract cancer, brain cancer, breast cancer, cervical cancer, choriocarcinoma, colon cancer, rectal cancer, head cancer, peritoneal tumor, fallopian tube cancer, endometrial cancer, esophageal cancer, gastric cancer, leukemia, lymphoma, sarcoma, adult Use in medicine for neurocytoma, oral cancer, ovarian cancer, pancreatic cancer, prostate cancer, testicular cancer, skin cancer and thyroid cancer.
  • Another aspect of the present invention relates to methods for the preparation, isolation and purification of compounds represented by formula (I).
  • the compounds of the present invention can be optionally substituted by one or more substituents, such as the above general formula compounds, or as specific examples in the embodiments, subclasses, and included in the present invention A class of compounds.
  • C 1-6 alkyl specifically refers to independently disclosed methyl, ethyl, C 3 alkyl, C 4 alkyl, C 5 alkyl and C 6 alkyl.
  • alkyl used in the present invention includes a saturated linear or branched monovalent hydrocarbon group of 1-20 carbon atoms, wherein the alkyl group can be independently and optionally substituted by one or more substituents described in the present invention.
  • the alkyl group contains 1-12 carbon atoms; in other embodiments, the alkyl group contains 1-10 carbon atoms; in other embodiments, the alkyl group contains 1-8 carbon atoms; some other embodiments are that the alkyl group contains 1-6 carbon atoms; some other embodiments are that the alkyl group contains 1-4 carbon atoms; some other embodiments are that the alkyl group Contains 1-3 carbon atoms.
  • alkyl groups include, but are not limited to, methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), n-propyl (n-Pr, -CH 2 CH 2 CH 3 ), isopropyl (i-Pr, -CH(CH 3 ) 2 ), n-butyl (n-Bu, -CH 2 CH 2 CH 2 CH 3 ), 2-methylpropyl or isobutyl (i-Bu, -CH 2 CH(CH 3 ) 2 ), 1-methylpropyl or sec-butyl (s-Bu, -CH(CH 3 )CH 2 CH 3 ), tert-butyl (t-Bu , -C(CH 3 ) 3 ), n-pentyl (-CH 2 CH 2 CH 2 CH 3 ), 2-pentyl (-CH(CH 3 )CH 2 CH 2 CH 3 ), 3-pentyl ( -CH(CH 2 CH 3 ) 2 ),
  • alkylene means a saturated divalent or multivalent hydrocarbyl group obtained by removing two or more hydrogen atoms from a saturated linear or branched chain hydrocarbyl group. Unless otherwise specified, an alkylene group contains 1-12 carbon atoms. In some embodiments, an alkylene group contains 1-6 carbon atoms; in other embodiments, an alkylene group contains 1-4 carbon atoms; in still other embodiments, an alkylene group The group contains 1-3 carbon atoms; in still other embodiments, the alkylene group contains 1-2 carbon atoms.
  • alkylene groups include, but are not limited to , methylene ( -CH2- ), ethylene ( -CH2CH2- ) , n-propylene ( -CH2CH2CH2- ), isopropylene group (-CH(CH 3 )CH 2 -) and the like.
  • alkynyl means a linear or branched monovalent hydrocarbon group containing 2-12 carbon atoms, or 2-8 carbon atoms, or 2-6 carbon atoms, or 2-4 carbon atoms, wherein at least CC in one position is an sp triple bond, wherein the alkynyl group can be independently unsubstituted or substituted by one or more substituents described herein, specific examples include, but are not limited to, ethynyl ( -C ⁇ CH), propargyl (-CH 2 C ⁇ CH), propynyl (-C ⁇ C-CH 3 ), 1-ynylbutyl (-CH 2 CH 2 C ⁇ CH), 2-alkyne Butyl (-CH 2 C ⁇ CCH 3 ), 3-alkynyl butyl (-C ⁇ CCH 2 CH 3 ), etc., wherein the alkynyl groups may independently be unsubstituted or replaced by one or more of the present invention The described substituents are substituted.
  • haloalkyl denotes an alkyl, alkenyl or alkoxy group substituted with one or more halogen atoms, wherein alkyl, alkenyl and alkoxy base has the meaning stated in the present invention.
  • alkoxy denotes an alkyl group attached to the rest of the molecule through an oxygen atom, wherein the alkyl group has the meaning described herein. Unless specified otherwise, the alkoxy groups contain 1-12 carbon atoms. In some embodiments, alkoxy groups contain 1-8 carbon atoms; in other embodiments, alkoxy groups contain 1-6 carbon atoms; in other embodiments, alkoxy groups The group contains 1-4 carbon atoms; in yet other embodiments, the alkoxy group contains 1-3 carbon atoms. The alkoxy groups may be optionally substituted with one or more substituents described herein.
  • alkoxy groups include, but are not limited to, methoxy (MeO, -OCH 3 ), ethoxy (EtO, -OCH 2 CH 3 ), 1-propoxy (n-PrO, n- Propoxy, -OCH 2 CH 2 CH 3 ), 2-propoxy (i-PrO, i-propoxy, -OCH(CH 3 ) 2 ), 1-butoxy (n-BuO,n- Butoxy, -OCH 2 CH 2 CH 2 CH 3 ), 2-methyl-l-propoxy (i-BuO, i-butoxy, -OCH 2 CH(CH 3 ) 2 ), 2-Butyl Oxygen (s-BuO, s-butoxy, -OCH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propoxy (t-BuO, t-butoxy, -OC(CH 3 ) 3 ), 1-pentyloxy (n-pentyloxy, -OCH 2 CH 2 CH 2 CH 3 ),
  • aryl may be used alone or as part of “arylalkyl", “aralkoxy” or “aryloxyalkyl” and means 6-14 carbon atoms, or 6-12 carbon atoms, or monocyclic, bicyclic, and tricyclic carbocyclic ring systems of 6 to 10 carbon atoms, of which at least one ring system is aromatic, wherein each ring system contains rings of 3 to 7 carbon atoms , with one or more attachment points connected to the rest of the molecule.
  • aryl may be used interchangeably with the terms “aromatic ring” or “aromatic ring”, eg aryl may include phenyl, naphthyl and anthracenyl.
  • the aryl groups can be independently unsubstituted or substituted with one or more substituents described herein.
  • heteroaryl means a monocyclic, bicyclic or tricyclic ring system containing 5-16 ring atoms, at least one of which is aromatic and at least one of which contains one or more heteroatoms, wherein Each ring system contains rings composed of 5-7 ring atoms, and there is one or Multiple attachment points connect to the rest of the molecule.
  • heteroaryl may be used interchangeably with the term “heteroaryl ring” or “heteroaromatic”, which includes monocyclic heteroaryls, fused bicyclic heteroaryls or polycyclic fused heteroaryls.
  • the heteroaryl group is a heteroaryl group consisting of 5-14 ring atoms comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. In other embodiments, the heteroaryl group is a heteroaryl group consisting of 5-12 ring atoms comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. In other embodiments, the heteroaryl group is a heteroaryl group consisting of 5-10 ring atoms comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. In other embodiments, the heteroaryl group is a fused bicyclic heteroaryl group consisting of 8-10 ring atoms comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N.
  • the heteroaryl group is a heteroaryl group consisting of 5-8 ring atoms comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. In other embodiments, the heteroaryl group is a heteroaryl group consisting of 5-7 ring atoms comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. In other embodiments, the heteroaryl group is a monocyclic heteroaryl group consisting of 5-6 ring atoms comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. In other embodiments, the heteroaryl group is a 5 ring atom heteroaryl group comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. In other embodiments, the heteroaryl group is a 6 ring atom heteroaryl group comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N.
  • heteroaryl includes, but is not limited to, the following monocyclic groups: 2-furyl, 3-furyl, N-imidazolyl, 2-imidazolyl, 4-imidazole Base, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl , 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (such as 3-pyridazine base), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (also known as tetrazolyl, such as 5H-tetrazolyl, 2H-tetrazolyl), triazolyl (such as
  • MM 1 ring atom means that the cyclic group is composed of MM 1 ring atom, and the ring atom includes carbon atom and/or O, N, S, P and other heteroatoms.
  • heteroaryl consisting of 5-10 ring atoms means that it includes heteroaryl consisting of 5, 6, 7, 8, 9 or 10 ring atoms.
  • heterocyclyl and “heterocycle” are used interchangeably herein to refer to a saturated or partially unsaturated, non-aromatic monocyclic, bicyclic or tricyclic ring system containing 3-12 ring atoms, wherein At least one ring atom is selected from nitrogen, sulfur and oxygen atoms, and the ring system has one or more points of attachment to the remainder of the molecule.
  • heterocyclic group includes monocyclic heterocyclic groups, bicyclic fused heterocyclic groups or polycyclic fused heterocyclic groups, spirocyclic heterocyclic groups or bridged heterocyclic groups, and also includes heterocyclic groups in which the heterocyclic group can be combined with one or more A non-aromatic carbocyclic or heterocyclic ring or a polycyclic ring system fused by one or more aromatic rings or a combination thereof, wherein the connecting atomic group or point is on the heterocyclic ring.
  • Bicyclic heterocyclyls include bridged bicyclic heterocyclyls, fused bicyclic heterocyclyls and spirobicyclic heterocyclyls.
  • Ring sulfur atoms can optionally be oxidized to S-oxides.
  • Ring nitrogen atoms can optionally be oxidized to N-oxygen compounds.
  • the heterocyclyl is a ring system consisting of 3-12 ring atoms; in some embodiments, the heterocyclyl is a monocyclic heterocyclyl consisting of 4-7 ring atoms; in some embodiments , the heterocyclyl is a monocyclic heterocyclyl consisting of 3-7 ring atoms; in some embodiments, the heterocyclyl is a monocyclic heterocyclyl consisting of 4-6 ring atoms; in some embodiments, the heterocyclyl The cyclic group is a monocyclic heterocyclic group consisting of 3-6 ring atoms; in some embodiments, the heterocyclic group is a monocyclic heterocyclic group composed of 5-6 ring atoms; in some embodiments, the heterocyclic group is a bicyclic heterocyclyl consisting of 7-12 ring atoms; in some embodiments, the heterocyclyl is a fused bicyclic heterocyclyl consisting of 7-12 ring atoms; in some embodiments
  • heterocyclic rings include, but are not limited to, pyrrolidinyl, tetrahydrofuryl, dihydrofuryl, tetrahydrothiophenyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, Linyl, thiomorpholinyl, thioxanyl, piperazinyl, homopiperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxirane Base, azepanyl, oxepinyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 2-pyrrolinyl, 3-pyrrolinyl, Indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolyl, pyrazolin
  • fused bicyclic ring means a monovalent or multivalent, saturated, partially unsaturated or fully unsaturated non-aromatic or aromatic ring system in which two rings share two adjacent rings atom.
  • spirocyclyl spirocycle
  • spirobicyclyl spirobicyclo
  • rings B and B' are called “fused bicyclic rings", while ring A' and ring B sharing a carbon atom are called “spiro rings” or “spirobicyclic rings”.
  • Each ring of the fused bicyclyl and spirobicyclyl can be carbocyclyl or heterocyclyl, and each ring is optionally substituted with one or more substituents described herein.
  • fused bicyclic heterocyclyl and "bicyclic fused heterocyclyl” are used interchangeably to denote monovalent saturated or partially unsaturated non-aromatic amalgamated ring systems. Such a system may contain independent or conjugated unsaturation, but its core structure does not contain aromatic rings or aromatic heterocycles (although aromatics may serve as substituents thereon).
  • Each ring in the ring system contains 3-7 atoms, and at least one ring contains one or more heteroatoms, that is, contains 1-6 carbon atoms and 1-3 atoms selected from N, O, P, S heteroatoms, where S or P is optionally substituted with one or more oxygen atoms to give groups like SO, SO2 , PO, PO2 , in some embodiments, the fused bicyclic heterocyclyl is 7 - a fused bicyclic heterocyclyl of 10 ring atoms; in some embodiments, a fused bicyclic heterocyclyl of 8-10 ring atoms; such examples include, but do not Limited to, 3-azafused[3.1.0]hexane, 3-azabicyclo[3.3.0]octane, hexahydro-furo[3,4-c]pyrrolyl, hexahydro-thiophene[3, 4-c]pyrrolyl, 3,4,5,6-tetrahydro-cyclopentane[c]thi
  • bridged bicyclic group or "bridged bicyclic ring” means a saturated or partially unsaturated non-aromatic bridged ring system, as shown in formula b, that is, ring A1 and ring A2 share an alkane chain, a heteroatom or a heteroalkane chain , wherein j is 1, 2, 3 or 4, and X 3 is an alkane chain, a heteroatom or a heteroalkane chain.
  • Such a system may contain independent or conjugated unsaturation, but its core structure does not contain aromatic rings or aromatic heterocycles (although aromatics may serve as substituents thereon).
  • each ring such as A1 or A2 contains 3-7 atoms
  • such examples include, but are not limited to, bicyclo[2.2.1]heptyl, 2-methyl-diazabicyclo[2.2. 1] Heptyl, etc.
  • the bridged bicyclyl is optionally substituted with one or more substituents described herein.
  • bridged bicyclic heterocyclic group means a saturated or partially unsaturated non-aromatic bridged bicyclic ring system, wherein each ring contains 3-7 atoms, and at least one ring contains one or more heteroatoms, that is, contains 1- 6 carbon atoms and 1-3 heteroatoms selected from N, O, P, S, where S or P is optionally substituted by one or more oxygen atoms to obtain like SO, SO 2 , PO, PO 2
  • the bridged bicyclic heterocyclyl is a bridged bicyclic heterocyclyl consisting of 6-10 ring atoms, examples of which include, but are not limited to, 2-oxo-5-azabicyclo[ 2.2.1] Heptyl, 2-thio-5-azabicyclo[2.2.1]heptyl, 2-oxo-5-azabicyclo[2.2.1]heptanyl, 2,5-diazabicyclo[2.2.1 ]heptyl, 2-methyl-2,5-diazabicyclo[[2.2.1 ]h
  • cycloalkyl means a monocyclic, bicyclic or tricyclic ring system having one or more points of attachment to the rest of the molecule, saturated, containing from 3 to 12 ring carbon atoms, including monocyclic, bicyclic or polycyclic Ring fused, spiro or bridged ring systems.
  • the cycloalkyl group is a spirobicycloalkyl group consisting of 6-10 atoms; in other embodiments, the cycloalkyl group is a fused bicycloalkyl group consisting of 6-10 atoms;
  • the base is a ring system containing 3-10 ring carbon atoms; in some other embodiments, the cycloalkyl group is a ring system containing 3-8 ring carbon atoms; in some other embodiments, the cycloalkyl group is a ring system containing 3-7 ring carbon atoms; A ring system of carbon atoms; some other embodiments, cycloalkyl is a ring system containing 5-8 ring carbon atoms; some other embodiments, cycloalkyl is a ring system containing 3-6 ring carbon atoms; some other By way of example, cycloalkyl is a ring system containing 5-6 ring carbon atoms; examples of cycloalkyl
  • alkylamino and alkylamino are used interchangeably and include “N-alkylamino” and “N,N-dialkylamino", wherein the hydrogen atoms in the amino group are independently replaced by one or replaced by two alkyl groups.
  • the alkylamino group is a lower alkylamino group formed by connecting one or two C 1-12 alkyl groups to a nitrogen atom.
  • the alkylamino group is a lower alkylamino group formed by one or two C 1-6 alkyl groups attached to a nitrogen atom.
  • the alkylamino group is a lower alkylamino group formed by one or two C 1-4 alkyl groups attached to a nitrogen atom. In still other embodiments, the alkylamino group is a lower alkylamino group formed by one or two C 1-3 alkyl groups attached to a nitrogen atom.
  • Suitable alkylamino groups may be mono- or dialkylamino, examples of alkylamino include, but are not limited to, N-methylamino (-NHCH 3 ), N-ethylamino (-NHCH 2 CH 3 ) , N,N-dimethylamino (-N(CH 3 ) 2 ), N,N-diethylamino (-N(CH 2 CH 3 ) 2 ) and the like.
  • the structural formulas described in the present invention include all isomeric forms (such as enantiomers, diastereomers, and geometric isomers (or conformational isomers)): for example, those containing asymmetric centers R, S configuration, double bond (Z), (E) isomers, and (Z), (E) conformational isomers. Accordingly, individual stereochemical isomers of the compounds of the present invention or mixtures thereof as enantiomers, diastereomers, or geometric isomers (or conformational isomers) are within the scope of the present invention.
  • prodrug used in the present invention means that a compound is transformed into a compound represented by formula (I) in vivo. Such conversion is effected by prodrug hydrolysis in blood or enzymatic conversion in blood or tissue to the parent structure.
  • the prodrug compound of the present invention can be an ester.
  • the ester can be used as a prodrug with phenyl esters, aliphatic (C 1-24 ) esters, acyloxymethyl esters, and carbonates. , carbamates and amino acid esters.
  • a compound of the present invention that contains a hydroxyl group can be acylated to give a prodrug form of the compound.
  • prodrug forms include phosphate esters, eg, phosphorylated parent hydroxyl groups.
  • phosphate esters eg, phosphorylated parent hydroxyl groups.
  • a complete discussion of prodrugs can be found in the following literature: T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the ACSSymposium Series, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, J. Rautio et al, Prodrugs: Design and Clinical Applications, Nature Review Drug Discovery, 2008, 7, 255-270, and SJHecker et al, Prodrugs of Phosphates and Phosphonates, Journal of Medicinal Chem istry, 2008 , 51, 2328-2345.
  • Metal refers to a product obtained through metabolism of a specific compound or its salt in vivo. Metabolites of a compound can be identified by techniques known in the art, and their activity can be characterized using assays as described herein. Such products can be obtained by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, degreasing, enzymatic cleavage and the like of the administered compound. Accordingly, the invention includes metabolites of the compounds, including metabolites produced by contacting a compound of the invention with a mammal for a substantial period of time.
  • Stereochemistry and usage of conventions in the present invention are generally referred to in the following documents: SP Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S. , "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., New York, 1994.
  • the compounds of the present invention may contain asymmetric centers or chiral centers and thus exist in different stereoisomers. All stereoisomeric forms of the compounds of the present invention, including but not limited to, diastereomers, enantiomers, atropisomers, and mixtures thereof, such as racemic mixtures, constitute the present invention part.
  • optically active compounds that is, they have the ability to rotate the plane of plane-polarized light.
  • the prefixes D, L or R and S are used to represent the absolute configuration of molecular chiral centers.
  • the prefixes d, l or (+), (-) are used to name the symbol of the compound's plane polarized light rotation, (-) or l means that the compound is left-handed, and the prefix (+) or d means that the compound is right-handed.
  • the chemical structures of these stereoisomers are the same, but their three-dimensional structures are different.
  • a particular stereoisomer may be an enantiomer, and a mixture of isomers is often referred to as an enantiomeric mixture.
  • racemic mixture A 50:50 mixture of enantiomers is known as a racemic mixture or racemate, which can result in no stereoselectivity or stereospecificity during a chemical reaction.
  • racemic mixture and “racemate” refer to an equimolar mixture of two enantiomers, devoid of optical activity.
  • tautomer or "tautomeric form” means that isomers of structures of different energies can be interconverted through a low energy barrier.
  • proton tautomers ie, prototropic tautomers
  • Atomic (valency) tautomers include interconversions of rearranged bonding electrons. Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
  • the "pharmaceutically acceptable salt” used in the present invention refers to organic and inorganic salts of the compounds of the present invention.
  • Pharmaceutically acceptable salts are well known in the art, as described in the literature: SM Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66:1-19, 1977.
  • Pharmaceutically acceptable non-toxic acid salts include, but are not limited to, inorganic acid salts formed by reaction with amino groups such as hydrochloride, hydrobromide, phosphate, sulfate, perchlorate, And organic acid salts such as acetate, oxalate, maleate, tartrate, citrate, succinate, malonate, or other methods such as ion exchange methods recorded in books and literature these salts.
  • salts include adipate, malate, 2-hydroxypropionate, alginate, ascorbate, aspartate, besylate, benzoate, bisulfate , borate, butyrate, camphorate, camphorsulfonate, cyclopentyl propionate, digluconate, lauryl sulfate, ethanesulfonate, formate, trans-butene di Glucoheptonate, Glycerophosphate, Gluconate, Hemisulfate, Heptanoate, Hexanoate, Hydroiodide, 2-Hydroxy-ethanesulfonate, Lacturonate, Lactic Acid Salt, laurate, lauryl sulfate, malate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, palmitate, pamoate , pectate, persulfate
  • Salts obtained with appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (C 1-4 alkyl) 4 salts.
  • the present invention also contemplates the quaternary ammonium salts of any compound containing an N group.
  • Water-soluble or oil-soluble or dispersed products can be obtained by quaternization.
  • Alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and amine cations formed as counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C1 -8 sulfonates and aromatic sulfonates.
  • a “solvate” of the present invention refers to an association of one or more solvent molecules with a compound of the present invention.
  • Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethylsulfoxide, ethyl acetate, acetic acid, aminoethanol.
  • hydrate refers to an association of solvent molecules with water.
  • protecting group refers to a substituent that reacts with another functional group, usually to block or protect specific functionality.
  • amino protecting group refers to a substituent attached to the amino group to block or protect the functionality of the amino group in the compound. Suitable amino protecting groups include acetyl, trifluoroacetyl, tert-butoxycarbonyl (BOC), benzyloxycarbonyl (CBZ) and 9-fluorenylmethyleneoxycarbonyl (Fmoc).
  • a “hydroxyl protecting group” refers to a substituent of a hydroxy group used to block or protect the functionality of the hydroxy group, suitable protecting groups include acetyl and silyl groups.
  • Carboxyl protecting group refers to the substituent of the carboxyl group used to block or protect the functionality of the carboxyl group.
  • the general carboxyl protecting group includes -CH 2 CH 2 SO 2 Ph, cyanoethyl, 2-(trimethylsilane base) ethyl, 2-(trimethylsilyl)ethoxymethyl, 2-(p-toluenesulfonyl)ethyl, 2-(p-nitrobenzenesulfonyl)ethyl, 2-(diphenyl phosphino)ethyl, nitroethyl, etc.
  • a general description of protecting groups can be found in: T W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991; and PJ Kocienski, Protecting Groups, Thieme, Stuttgart, 2005.
  • the compound involved in the present invention can effectively activate TLR8 and inhibit HBV infection.
  • the present invention relates to a compound as shown in formula (I) or stereoisomers, tautomers, nitrogen oxides, solvates, metabolites, pharmaceutical acceptable salt or its prodrugs,
  • X is N or CR 4 ;
  • Each of R 1 , R 2 , R 4 , R 5 , R 6 , R 7 and R 8 is independently hydrogen, deuterium, F, Cl, Br, I, hydroxyl, cyano, amino, C 1-4 alkylamino, C 1-6 alkoxy or C 1-6 alkyl, wherein the C 1-4 alkylamino, C 1-6 alkoxy and C 1-6 alkyl are each independently unsubstituted or replaced by 1 , 2, 3 or 4 substituents independently selected from F, Cl, Br, I, hydroxyl, cyano, amino and C 1-4 alkyl are substituted;
  • Y is O or S
  • R 3 is -C 1-6 alkylene-R 10 , wherein the -C 1-6 alkylene- in the -C 1-6 alkylene-R 10 is unsubstituted or replaced by 1, 2 , 3, 4 or 5 substituents of R w1 are substituted;
  • Each R c is independently hydrogen, deuterium or C 1-4 alkyl, wherein said C 1-4 alkyl is unsubstituted or is independently selected from F, Cl, Br by 1, 2, 3 or 4 , I, hydroxyl, cyano, amino and C 1-4 alkyl are substituted by substituents;
  • Each R 10a and R 10b is independently a C 1-6 alkyl group, a C 3-7 cycloalkyl group, a heteroaryl group consisting of 5-12 ring atoms or a heterocyclyl group consisting of 3-12 ring atoms, wherein, The C 1-6 alkyl group, C 3-7 cycloalkyl group, heteroaryl group consisting of 5-12 ring atoms and heterocyclic group consisting of 3-12 ring atoms are each independently unsubstituted or replaced by 1 , 2, 3, 4 or 5 substituents of R w2 are substituted;
  • R is H, deuterium, C 1-6 alkyl or a heterocyclic group consisting of 3-6 ring atoms, wherein the C 1-6 alkyl and 3-6 ring atoms are independently Unsubstituted or substituted by 1, 2, 3, 4 or 5 R substituents;
  • R 9 is hydrogen, deuterium, -LR 11 , F, Cl, Br, I, hydroxyl, cyano, amino, C 1-4 alkylamino, C 1-6 alkoxy or C 1-6 alkyl, wherein, The C 1-4 alkylamino, C 1-6 alkoxy and C 1-6 alkyl are each independently unsubstituted or 1, 2, 3 or 4 independently selected from F, Cl, Br, 1, hydroxyl, cyano, amino and C 1-4 alkyl substituents are substituted;
  • the aryl group and the heterocyclic group consisting of 3-12 ring atoms are each independently unsubstituted or substituted by 1, 2, 3, 4 or 5 R substituents;
  • R a , R b and the N atoms connected to them together form a heterocyclic group composed of 3-8 ring atoms, wherein the heterocyclic group composed of 3-8 ring atoms is unsubstituted or is 1, 2, 3 or 4 R w4 substitutions;
  • Each of R 9a , R 9b , R 9c and R 9d is independently C 1-12 alkyl, C 2-12 alkenyl, C 2-12 alkynyl or -C 1-6 alkylene-R d2 , wherein, The -C 1-6 alkylene in the C 1-12 alkyl, C 2-12 alkenyl, C 2-12 alkynyl and -C 1-6 alkylene-R d2 are independently not is substituted or substituted by 1, 2, 3, 4 or 5 R substituents ;
  • Each of R d1 and R d2 is independently hydroxyl, C 1-6 alkoxy, C 6-10 aryl, heteroaryl consisting of 5-12 ring atoms, C 3-6 cycloalkyl or C 1-6 Alkylamino, wherein, the C 1-6 alkoxyl group, C 6-10 aryl group, heteroaryl group consisting of 5-12 ring atoms, C 3-6 cycloalkyl group and C 1-6 alkylamino group respectively independently unsubstituted or substituted by 1, 2, 3, 4 or 5 R substituents ;
  • a heterocyclic group consisting of 5-6 ring atoms, a heteroaryl group or a C 6-10 aryl group, wherein, the amino group, C 1-6 alkylamino group, C 1-6 alkoxy group, C 1-6 6 alkyl, C 1-6 haloalkyl, -C( O)-C 1-4 alkyl, C 2-12 alkenyl, C 2-12 alkynyl, C 3-6 cycloalkyl, 3-6 The heterocyclic group consisting of 5-6 ring atoms,
  • t 0, 1 or 2.
  • each of R 1 , R 2 , R 4 , R 5 , R 6 , R 7 and R 8 is independently hydrogen, deuterium, F, Cl, Br, I, hydroxyl, cyano, amino, N-methylamino, N-ethylamino, N,N-dimethylamino, N,N-diethylamino, methoxy, ethoxy, 1-propoxy, 2-propoxy, 1-butoxy base, 2-methyl-l-propoxy, 2-butoxy, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n- Pentyl or n-hexyl, wherein, the N-methylamino, N-ethylamino, N,N-dimethylamino, N,N-diethylamino, methoxy, ethoxy, 1-propoxy , 2-propoxy,
  • R is H, deuterium, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, Aziridyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrofuryl, tetrahydrothiophenyl, tetrahydropyranyl , tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl or piperazinyl, wherein the methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl Base, sec-butyl, tert-butyl,
  • each R w3 has the meaning as described in the present invention.
  • R 3 is -CH 2 -R 10 , -(CH 2 ) 2 -R 10 , -(CH 2 ) 3 -R 10 , -CH(CH 3 )CH 2 -R 10 , - CH 2 CH(CH 3 )-R 10 or -(CH 2 ) 4 -R 10 , wherein -CH 2 - in -CH 2 -R 10 and -(CH 2 ) 2 -R 10 in -(CH 2 ) 2 -R 10 -(CH 2 ) 2 -, -(CH 2 ) 3 - in -(CH 2 ) 3 -R 10 , -CH(CH 3 )CH 2 - in -CH(CH 3 )CH 2 -R 10 , -CH 2 CH(CH 3 )- in -CH 2 CH(CH 3 )-R 10 and -( CH 2 ) 4 -(CH 2 ) 4 in -R 10 are each independently unsubstituted or
  • each of R 10 and R w1 has the meaning as described in the present invention.
  • each of R c , R 10a and R 10b has the meaning as described in the present invention.
  • each R 10a and R 10b is independently C 1-4 alkyl, C 3-6 cycloalkyl, heteroaryl consisting of 5-6 ring atoms or 3-6 ring atoms
  • the heterocyclic group wherein, the C 1-4 alkyl, C 3-6 cycloalkyl, heteroaryl composed of 5-6 ring atoms and heterocyclic group composed of 3-6 ring atoms are independently Unsubstituted or substituted by 1, 2, 3, 4 or 5 R w2 substituents;
  • each R w2 has the meaning as described in the present invention.
  • R c is hydrogen, deuterium, methyl, ethyl, n-propyl or isopropyl, wherein said methyl, ethyl, n-propyl and isopropyl are unsubstituted or Substituted by 1, 2, 3 or 4 substituents independently selected from F, Cl, Br, I, hydroxy, cyano, amino, methyl, ethyl, n-propyl and isopropyl.
  • each R 10a and R 10b is independently methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl Base, tert-butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, furyl, pyrrolyl, pyridyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, iso Oxazolyl, oxadiazolyl, 1,3,5-triazinyl, thiazolyl, thienyl, pyrazinyl, pyridazinyl, pyrimidinyl, aziridinyl, azetidinyl, oxa Cyclobutyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolidin
  • each R w2 has the meaning as described in the present invention.
  • R 9 is hydrogen, deuterium, -LR 11 , F, Cl, Br, I, hydroxyl, cyano, amino, C 1-4 alkylamino, C 1-4 alkoxy or C 1 -4 alkyl, wherein the C 1-4 alkylamino, C 1-4 alkoxy and C 1-4 alkyl are each independently unsubstituted or are independently selected by 1, 2, 3 or 4 Substituents from F, Cl, Br, I, hydroxyl, cyano, amino and C 1-4 alkyl;
  • each of L and R has the meaning as described in the present invention.
  • each of R a , R b , R 9a , R 9b , t, R 9c and R w4 has the meaning as described in the present invention.
  • R 9 is hydrogen, deuterium, -LR 11 , F, Cl, Br, I, hydroxyl, cyano, amino, N-methylamino, N-ethylamino, N,N-dimethylamino , N,N-diethylamino, methoxy, ethoxy, 1-propoxy, 2-propoxy, 1-butoxy, 2-methyl-l-propoxy, 2-butoxy base, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl or n-hexyl, wherein the N-methylamino, N -Ethylamino, N,N-dimethylamino, N,N-diethylamino, methoxy, ethoxy, 1-propoxy, 2-propoxy, 1-butoxy, 2-methyl -l-propoxy, 2-butoxy, methyl,
  • each of L and R has the meaning as described in the present invention.
  • each of R a , R b , R 9a , R 9b , t, R 9c and R w4 has the meaning as described in the present invention.
  • the -C 1-4 alkylene- each independently unsubstituted or substituted by 1, 2, 3, 4 or 5 substituents of R W5 ;
  • R a , R b and the N atoms connected to them together form a heterocyclic group consisting of 3-6 ring atoms, wherein the 3-6 ring
  • the heterocyclic group consisting of atoms is unsubstituted or substituted by 1, 2, 3 or 4 R w4 ;
  • each of R 9d , R d1 , R w5 and R w4 has the meaning as described in the present invention.
  • R a , R b and the N atoms they are connected together form aziridinyl, azetidinyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, piperidinyl, morpholinyl, Thiomorpholinyl or piperazinyl, wherein, the aziridinyl, azetidinyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, piperidinyl, morpholinyl, thio Morpholinyl and piperazinyl are each independently unsubstituted or substituted by 1, 2, 3 or 4 R;
  • each of R 9d , R d1 , R w5 and R w4 has the meaning as described in the present invention.
  • each of R 9a , R 9b , R 9c and R 9d is independently C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl or -C 1-4 alkylene Group -R d2 , wherein the -C 1-4 alkylene in the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl and -C 1-4 alkylene -R d2 Group-each independently unsubstituted or substituted by 1, 2, 3, 4 or 5 R substituents ;
  • each of R d2 and R w6 has the meaning as described in the present invention.
  • each R d1 and R d2 are independently hydroxyl, C 1-4 alkoxy, phenyl, heteroaryl consisting of 5-6 ring atoms, cyclopropyl, cyclobutyl, ring Pentyl, cyclohexyl or C 1-4 alkylamino, wherein, the C 1-4 alkoxy, phenyl, heteroaryl, cyclopropyl, cyclobutyl, ring Pentyl, cyclohexyl and C 1-4 alkylamino are each independently unsubstituted or substituted by 1, 2, 3, 4 or 5 R substituents;
  • each R w7 has the meaning as described in the present invention.
  • each of R d2 and R w6 has the meaning as described in the present invention.
  • each Rd1 and Rd2 are independently hydroxyl, methoxy, ethoxy, 1-propoxy, 2-propoxy, 1-butoxy, 2-methyl-1 - Propoxy, 2-butoxy, phenyl, furyl, pyrrolyl, pyridyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazole base, 1,3,5-triazinyl, thiazolyl, thienyl, pyrazinyl, pyridazinyl, pyrimidinyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, N-methylamino, N -Ethylamino, N,N-dimethylamino or N,N-diethylamino, wherein the methoxy, ethoxy, 1-propoxy, 2-propoxy, 1-butoxy, 2-methyl
  • each R w7 has the meaning as described in the present invention.
  • the present invention comprises a compound of one of the following structures or a stereoisomer, tautomer, nitrogen oxide, solvate, metabolite, pharmaceutically acceptable compound of a compound comprising one of the following structures Salt or its prodrugs:
  • the present invention also provides a pharmaceutical composition, comprising the compound described in the present invention and pharmaceutically acceptable auxiliary materials.
  • the pharmaceutical composition of the present invention further comprises other one or more therapeutic agents, wherein the therapeutic agents are HBV DNA polymerase inhibitors, toll-like receptor 7 modulators, Toll-like receptor 8 modulators, toll-like receptor 7 and 8 modulators, toll-like receptor 3 modulators, interferon ⁇ ligands, HBsAg inhibitors, compounds targeting HbcAg, cyclophilin inhibitors , HBV therapeutic vaccine, HBV preventive vaccine, HBV virus entry inhibitor, NTCP inhibitor, antisense oligonucleotide targeting viral mRNA, short interfering RNA (siRNA), hepatitis B virus E antigen inhibitor, HBx Inhibitors, cccDNA inhibitors, HBV antibodies, thymosin agonists, cytokines, nucleoprotein inhibitors, stimulators of retinoic acid-inducible gene 1, NOD2 stimulators, recombinant thymosin ⁇ -1, hepatitis B virus replication inhibition agents,
  • the pharmaceutical composition of the present invention wherein the other one or more therapeutic agents are lamivudine, telbivudine, tenofovir, entecavir, adefovir dipivoxil , Tenofovir Alafenamide, Tenofovir Disoproxil, Tenofovir Alafenamide Fumarate, Tenofovir Alafenamide Hemifumarate, Alfaferone, Alloferon, Simox interleukin, clavudine, emtricitabine, famciclovir, interferon, Bao Ganling CP, interferon, interleukin-2, milvotidate, nitazoxanide, ribavirin, roxime Interferon-A, sizoran, Euforavac, Ampligen, Phosphazid, Heplisav, recombinant human interleukin-2, levamisole, or propagermanium.
  • the other one or more therapeutic agents are
  • the interferon of the present invention is interferon ⁇ -1b, interferon ⁇ , interferon ⁇ -2a, interferon ⁇ -1a, interferon ⁇ -2, pegylated interferon ⁇ - 2a or interferon alpha-2b.
  • the present invention proposes the use of the compound or pharmaceutical composition described in the present invention in activating TLR8.
  • the present invention proposes the use of the compound or pharmaceutical composition of the present invention in the preparation of a kit for activating TLR8.
  • the present invention also provides the preparation of the compound or the pharmaceutical composition for preventing, treating, treating or alleviating the TLR8 Use in medicines for mediated diseases.
  • the disease mediated by TLR8 is hepatitis B virus infection, hepatitis C virus infection, influenza virus infection, herpes virus infection, HIV infection, allergic disease, rheumatoid arthritis, allergic Asthma, chronic fatigue, type 2 diabetes, hay fever, lupus erythematosus, multiple sclerosis, melanoma, lung cancer, liver cancer, basal cell carcinoma, kidney cancer, myeloma, biliary tract cancer, brain cancer, breast cancer, cervical cancer, Choriocarcinoma, colon cancer, rectal cancer, head and neck cancer, peritoneal tumor, fallopian tube cancer, endometrial cancer, esophageal cancer, gastric cancer, leukemia, lymphoma, sarcoma, neuroblastoma, oral cavity cancer, ovarian cancer, pancreatic cancer, Prostate, testicular, skin, or thyroid cancer.
  • the present invention also provides the use of the compound or pharmaceutical composition in the preparation of medicines for treating or preventing diseases of the immune regulatory system.
  • the present invention also provides the use of the compound or pharmaceutical composition in the preparation of medicines for treating or preventing viral infection or tumors.
  • the present invention also provides the preparation of the compound or the pharmaceutical composition for the treatment or prevention of hepatitis B virus infection, hepatitis C virus infection, influenza virus infection, herpes virus infection, HIV infection , allergic diseases, rheumatoid arthritis, allergic asthma, chronic fatigue, type II diabetes, hay fever, lupus erythematosus, multiple sclerosis, melanoma, lung cancer, liver cancer, basal cell carcinoma, kidney cancer, myeloma, Biliary tract cancer, brain cancer, breast cancer, cervical cancer, choriocarcinoma, colon cancer, rectal cancer, head and neck cancer, peritoneal tumor, fallopian tube cancer, endometrial cancer, esophageal cancer, gastric cancer, leukemia, lymphoma, sarcoma, neuroblast Cell tumor, oral cancer, ovarian cancer, pancreatic cancer, prostate cancer, testicular cancer, skin cancer and thyroid cancer.
  • the present invention also provides the use of said compound or pharmaceutical composition in the preparation of medicines for preventing, treating or alleviating immune regulatory system diseases, comprising administering an effective therapeutic dose of said compound or said present invention.
  • the pharmaceutical composition described in the invention is administered to patients.
  • the present invention also provides a method for preventing, treating or alleviating a disease mediated by TLR8 in a patient with the compound or pharmaceutical composition, the method comprising using a pharmaceutically acceptable effective dose of the compound of the present invention for the patient Dosing.
  • the present invention also provides a method for treating or preventing viral infection or tumor with the compound or the pharmaceutical composition, the method comprises using a pharmaceutically acceptable effective dose of the pharmaceutical composition containing the compound of the present invention on The patient administers the drug.
  • the present invention also provides the compound or the pharmaceutical composition for treating or preventing hepatitis B virus infection, hepatitis C virus infection, influenza virus infection, herpes virus infection, HIV infection, allergic disease , rheumatoid arthritis, allergic asthma, chronic fatigue, type II diabetes, hay fever, lupus erythematosus, multiple sclerosis, melanoma, lung cancer, liver cancer, basal cell carcinoma, kidney cancer, myeloma, biliary tract cancer, brain cancer Cancer, breast cancer, cervical cancer, choriocarcinoma, colon cancer, rectal cancer, head and neck cancer, peritoneal cancer, fallopian tube cancer, endometrial cancer, esophageal cancer, gastric cancer, leukemia, lymphoma, sarcoma, neuroblastoma, oral cavity Cancer, ovarian cancer, pancreatic cancer, prostate cancer, testicular cancer, skin cancer and thyroid cancer, the method comprising administering to a patient a pharmaceutical composition
  • the present invention also provides a method for preventing, treating or alleviating a patient's immunomodulatory system disease with the compound or the pharmaceutical composition, comprising administering an effective therapeutic dose of the compound or the pharmaceutical combination of the present invention Drugs are administered to patients.
  • the present invention also provides the use of the compound or the pharmaceutical composition of the present invention as a drug for preventing, treating or alleviating the diseases mediated by TLR8 in patients.
  • the present invention also provides the use of the compound or pharmaceutical composition described in the present invention as a drug for preventing, treating or alleviating diseases of the patient's immune regulatory system.
  • the present invention also provides the use of the compound or pharmaceutical composition described in the present invention as a drug for treating or preventing viral infection or tumor.
  • the present invention also provides the use of the compound of the present invention or the pharmaceutical composition as a method for treating or preventing hepatitis B virus infection, hepatitis C virus infection, influenza virus infection, herpes virus infection, HIV infection , allergic diseases, rheumatoid arthritis, allergic asthma, chronic fatigue, type II diabetes, hay fever, lupus erythematosus, multiple sclerosis, melanoma, lung cancer, liver cancer, basal cell carcinoma, kidney cancer, myeloma, Biliary tract cancer, brain cancer, breast cancer, cervical cancer, choriocarcinoma, colon cancer, rectal cancer, head and neck cancer, peritoneal tumor, fallopian tube cancer, endometrial cancer, esophageal cancer, gastric cancer, leukemia, lymphoma, sarcoma, neuroblast Cell tumor, oral cancer, ovarian cancer, pancreatic cancer, prostate cancer, testicular cancer, skin cancer and thyroid cancer.
  • the patient is a mammal, and in other embodiments, the patient is a human.
  • the use further comprises contacting the cells with a therapeutic agent.
  • Another aspect of the present invention relates to methods for the preparation, isolation and purification of compounds represented by formula (I).
  • the invention also relates to the use of the compounds of the invention and their pharmaceutically acceptable salts for the manufacture of medicinal products effective in the treatment of the diseases mentioned in the invention.
  • the compounds of the present invention are also useful in the manufacture of a medicament for alleviating, preventing, controlling or treating the symptoms of the diseases of the present invention in patients.
  • pharmaceutically acceptable includes that the substance or composition must be chemically or toxicologically appropriate in relation to the other ingredients making up the formulation and the mammal being used for treatment.
  • the salts of the compounds of the present invention also include the salts of the intermediates of the compounds shown in the formula (I) or the separated enantiomers used in the preparation or purification of the compounds shown in the formula (I), but not necessarily pharmaceutically acceptable Salt.
  • the desired salts may be prepared by any suitable method provided in the literature, for example, using inorganic acids such as hydrochloric, hydrobromic, sulfuric, nitric and phosphoric acids and the like.
  • organic acids such as acetic, maleic, succinic, mandelic, fumaric, malonic, pyruvic, malic, 2-hydroxypropionic, citric, oxalic, glycolic, and salicylic ; pyranonic acids, such as glucuronic acid and galacturonic acid; alpha-hydroxy acids, such as citric acid and tartaric acid; amino acids, such as aspartic acid and glutamic acid; aromatic acids, such as benzoic acid and cinnamic acid; Sulfonic acids, such as p-toluenesulfonic acid, benzenesulfonic acid, methanesulfonic acid, ethanesulfonic acid, trifluoromethanesulfonic acid,
  • the desired salts can be prepared by suitable methods, e.g., using inorganic or organic bases, such as ammonia (primary, secondary, tertiary), alkali metal hydroxides, ammonium , N + (R 14 ) 4 salts and alkaline earth metal hydroxides, etc.
  • inorganic or organic bases such as ammonia (primary, secondary, tertiary), alkali metal hydroxides, ammonium , N + (R 14 ) 4 salts and alkaline earth metal hydroxides, etc.
  • Suitable salts include, but are not limited to, organic salts derived from amino acids such as glycine and arginine, ammonia such as primary, secondary and tertiary ammonia, salts of N + (R 14 ) 4 such as R 14 is H, C 1-4 alkyl, C 6-10 aryl, C 6-10 aryl C 1-4 alkyl, etc., and cyclic ammonia, such as piperidine, morpholine, and piperazine, etc., and from sodium, Calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium give inorganic salts.
  • nontoxic ammonium, quaternary ammonium salts and amine cations formed by counterions such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C 1-8 sulfonates and Aromatic sulfonates.
  • the pharmaceutical composition of the present invention includes the compound represented by formula (I), the compounds listed in the present invention, or the compounds of the examples, and pharmaceutically acceptable auxiliary materials.
  • the compounds in the pharmaceutical composition of the present invention can be effectively used for the treatment of diseases mediated by TLR8.
  • Areas of disease treatment that may be mentioned for the compounds or pharmaceutical compositions of the present invention are, for example: immune diseases, diseases caused by viral infections and tumors, for example, hepatitis B virus infection, hepatitis C virus infection, Influenza virus infection, herpes virus infection, HIV infection, allergic disease, rheumatoid arthritis, allergic asthma, chronic fatigue, type 2 diabetes, hay fever, lupus erythematosus, multiple sclerosis, melanoma, lung cancer, liver cancer , basal cell carcinoma, kidney cancer, myeloma, biliary tract cancer, brain cancer, breast cancer, cervical cancer, choriocarcinoma, colon cancer, rectal cancer, head and neck cancer, peritoneal tumor, fallopian tube cancer, endometrial cancer, esophageal cancer, Gastric cancer, leukemia, lymphoma, sarcoma,
  • the present invention includes pharmaceutical preparations, which contain one or more compounds represented by formula (I) or pharmaceutical compositions thereof in addition to non-toxic and inert pharmaceutically suitable auxiliary materials.
  • the above-mentioned pharmaceutical preparation may also contain other active pharmaceutical ingredients other than the compound represented by formula (I).
  • compositions of the present invention exist in free form, or suitably, as pharmaceutically acceptable derivatives.
  • pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable prodrugs, salts, esters, salts of esters, or any other compounds that can be administered directly or indirectly according to the needs of patients.
  • the pharmaceutical composition of the present invention comprises any compound represented by formula (I) of the present invention, and further comprises pharmaceutically acceptable adjuvants, and these adjuvants, for example, as used in the present invention, include Any solvent, solid excipient, diluent, binder, disintegrant, or other liquid excipient, dispersant, flavoring or suspending agent, surfactant, isotonic agent, thickener, emulsifier , preservatives, solid binders or lubricants, etc., suitable for specific target dosage forms. As described in: In Remington: The Science and Practice of Pharmacy, 21st edition, 2005, ed.
  • Substances which may be used as pharmaceutically acceptable excipients include, but are not limited to, ion exchangers; aluminum; aluminum stearate; lecithin; serum proteins, such as human serum albumin; buffer substances such as phosphate; glycine; sorbic acid; Potassium phosphate; partial glyceride mixture of saturated vegetable fatty acids; water; salts or electrolytes such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts; colloidal silicon; magnesium trisilicate; polyethylene Pyrrolidone; polyacrylates; waxes; polyethylene-polyoxypropylene-blocking polymers; lanolin; sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as carboxymethyl sodium cellulose, ethyl cellulose, and cellulose acetate; gum powder; malt; gelatin; talc; excipients such as
  • the pharmaceutical composition of the compound of the present invention can be administered in any of the following ways: oral administration, spray inhalation, topical administration, rectal administration, nasal administration, topical administration, vaginal administration, parenteral administration Drugs such as subcutaneous, intravenous, intramuscular, intraperitoneal, intrathecal, intraventricular, intrasternal, or intracranial injection or infusion, or by means of an explanted reservoir.
  • oral administration intramuscular injection, intraperitoneal administration or intravenous injection.
  • Dosage forms for administration may be liquid dosage forms or other dosage forms.
  • the liquid dosage forms can be true solutions, colloids, particulate dosage forms, and suspension dosage forms.
  • Other dosage forms such as tablets, capsules, drop pills, aerosols, pills, powders, solutions, suspensions, emulsions, granules, suppositories, lyophilized powder injections, clathrates, implants, patches, wipes agent etc.
  • Oral tablets and capsules may contain excipients such as binders such as syrup, acacia, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers such as lactose, sucrose, corn starch, calcium phosphate, sorbitol, amino acetic acid; lubricants such as magnesium stearate, talc, polyethylene glycol, silica; disintegrants such as potato starch; or acceptable wetting agents such as sodium lauryl sulfate. Tablets can be coated by methods known in pharmacy.
  • binders such as syrup, acacia, sorbitol, tragacanth, or polyvinylpyrrolidone
  • fillers such as lactose, sucrose, corn starch, calcium phosphate, sorbitol, amino acetic acid
  • lubricants such as magnesium stearate, talc, polyethylene glycol, silica
  • disintegrants such as potato starch
  • Oral solutions can be made into hydrated oil suspensions, solutions, emulsions, syrups or elixirs, and can also be made into dry products, which are supplemented with water or other suitable media before use.
  • This liquid preparation may contain conventional additives such as suspending agent, sorbitol, cellulose methyl ether, glucose syrup, gelatin, hydroxyethyl cellulose, carboxymethyl cellulose, aluminum stearate gel, hydrogenated edible Fats, emulsifiers, such as lecithin, sorbitan monooleate, gum arabic; or non-aqueous excipients (which may contain edible oils), such as almond oil, fats such as glycerin, glycol, or ethanol; preservatives, Such as methyl or propyl paraben, sorbic acid. Flavoring or coloring agents may be added if desired.
  • Suppositories may contain conventional suppository bases, such as cocoa butter or other glycerides.
  • liquid dosage forms are generally made up of the compound and a sterile excipient.
  • the preferred auxiliary material is water.
  • the compound can be either dissolved in the excipients or made into a suspension solution.
  • the compound is first dissolved in water, filtered and sterilized, and put into a sealed bottle or ampoule.
  • the compounds of the present invention may be prepared in the form of a suitable ointment, lotion, or cream, wherein the active ingredient is suspended or dissolved in one or more excipients, wherein ointment formulations may use excipients including but Not limited to: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene, emulsifying wax and water; excipients that can be used in lotions and creams include but are not limited to: mineral oil, sorbitan mono Stearate, Tween 60, cetyl esters wax, cetyl aryl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • excipients including but Not limited to: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene, emulsifying wax and water
  • excipients that can be used in lotions and creams include but are not limited to: mineral oil,
  • dosage forms for oral administration to humans may contain from about 1 to 1000 mg of active substance formulated with suitable and convenient amounts of pharmaceutically acceptable excipients.
  • the pharmaceutically acceptable excipients vary from about 5% to about 95% (weight:weight) of the total composition.
  • the compounds disclosed in the present invention can be administered to individuals for a desired period of time or duration according to an effective dosage regimen, such as at least about one month, at least about 2 months, at least about 3 months months, at least about 6 months, or at least about 12 months or longer.
  • an effective dosage regimen such as at least about one month, at least about 2 months, at least about 3 months months, at least about 6 months, or at least about 12 months or longer.
  • the compound is administered on a daily or intermittent schedule over the life of the individual.
  • the dose or frequency of administration of the disclosed compounds may be adjusted during the course of treatment based on the judgment of the administering physician.
  • Compounds can be administered to an individual (eg, a human) in an effective amount. In certain embodiments, the compound is administered once daily.
  • a method for treating or preventing a human disease or condition comprising a therapeutically effective amount of one or more (e.g., one, two, three, four, one or two, one to three, or one to four) additional therapeutic agent combinations
  • Administration of a therapeutically effective amount of a compound disclosed herein or its pharmaceutically acceptable salt since agonists of TLR-8 are useful in the treatment of a variety of diseases or conditions, the particular identity of the additional therapeutic agent will depend on the particular disease or condition being treated.
  • Compounds represented by formula (I) may be administered by any useful route and manner, such as orally or parenterally (eg intravenously).
  • the therapeutically effective amount of the compound represented by formula (I) is about 0.00001 mg/kg body weight/day to about 10 mg/kg body weight/day, such as about 0.0001 mg/kg body weight/day to about 10 mg/kg/day or about 0.001 mg /kg body weight/day to about 1 mg/kg body weight/day, or for example about 0.01 mg/kg body weight/day to about 1 mg/g body weight/day or for example about 0.05 mg/kg body weight/day to about 0.5 mg/kg body weight/day sky.
  • the therapeutically effective amount of the compound represented by formula (I) is about 0.01 mg/dose to about 1000 mg/dose, such as about 0.01 mg/dose to about 100 mg/dose, or for example about 0.1 mg/dose to about 100 mg/dose, or For example about 1 mg/dose to about 100 mg/dose, or for example about 1 mg/dose to about 10 mg/dose.
  • Other therapeutically effective doses of compounds represented by formula (I) are about 1 mg/dose, or about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40 , 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 100 mg/dose.
  • Other therapeutically effective amounts of compounds represented by formula (I) are about 100 mg/dose, or about 125, 150, 175, 200, 225, 250, 275, 300, 350, 400, 450 or 500 mg/dose.
  • Single doses can be administered hourly, daily or weekly. For example, a single dose may be administered every 1 hour, every 2, 3, 4, 6, 8, 12, 16 hours, or every 24 hours. A single dose can also be administered every 1 day, every 2, 3, 4, 5, 6 days or every 7 days. Single doses can also be administered every 1 week, every 2, 3 weeks or every 4 weeks. In certain embodiments, a single dose may be administered weekly. A single dose can also be administered once a month.
  • the frequency of administration of the compound represented by formula (I) will be determined by the needs of the individual patient and may be, for example, once a day or two or more times a day.
  • administration of the compound can be administered to a human infected with HBV or HCV for a period of time ranging from 20 days to 180 days, or for example 20 days to 90 days, or for example a period of 30 days to 60 days.
  • Administration may be intermittent, wherein the patient receives daily doses of a compound of formula (I) over a period of several days or more, followed by several days or more in which the patient receives no daily doses of the compound.
  • multi-day time period For example, a patient may receive doses of the compound every other day or three times a week. Again by way of example, a patient may receive daily doses of the compound for a period of 1 to 14 days, followed by a period of 7 to 21 days during which the patient receives no doses of the compound, followed by a period of each day in which the patient receives again the compound. Subsequent periods of daily dosage (eg, from 1 to 14 days). Administration of the compound, followed by periods of non-administration of the compound, may be repeated as clinically required to treat the patient.
  • a pharmaceutical composition comprising one or more (e.g., one, two, three, four, one or two, one to three, or One to four) additional therapeutic agents in combination of a compound disclosed in the present invention or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.
  • kits comprising one or more (e.g., one, two, three, four, one or two, one to three or one to four) additional therapeutic agents in combination with a compound disclosed herein or a pharmaceutically acceptable salt thereof.
  • a compound disclosed herein, or a pharmaceutically acceptable salt thereof is combined with one, two, three, four or more additional therapeutic agents. In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with two additional therapeutic agents. In other embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with three additional therapeutic agents. In additional embodiments, a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is combined with four additional therapeutic agents.
  • the one, two, three, four or more additional therapeutic agents may be different therapeutic agents selected from the same class of therapeutic agents, and/or they may be selected from different classes of therapeutic agents.
  • the components of the composition are administered in a simultaneous or sequential schedule.
  • the combination may be administered in two or more administrations.
  • a compound disclosed herein is combined with one or more additional therapeutic agents in a single dosage form for simultaneous administration to a patient, eg, as a solid dosage form for oral administration.
  • compounds disclosed herein are administered with one or more additional therapeutic agents.
  • Co-administration of a compound disclosed herein with one or more additional therapeutic agents generally refers to simultaneous or sequential administration of a compound disclosed herein and one or more additional therapeutic agents such that a therapeutically effective amount of the compound disclosed herein Both the compound and one or more additional therapeutic agents are present in the patient.
  • Co-administration includes administering a unit dose of a compound disclosed herein before or after administration of a unit dose of one or more additional therapeutic agents, for example within seconds, minutes or hours of administration of the one or more additional therapeutic agents
  • Compounds disclosed herein are administered internally.
  • a unit dose of a compound disclosed herein is administered first, followed by administration of a unit dose of one or more additional therapeutic agents within seconds or minutes.
  • a unit dose of one or more additional therapeutic agents is administered first, followed by administration of a unit dose of a compound disclosed herein within seconds or minutes.
  • a unit dose of a compound disclosed herein is administered first followed by administration of a unit dose of one or more additional therapeutic agents over a period of several hours (eg, 1-12 hours). In other embodiments, a unit dose of one or more additional therapeutic agents is administered first followed by administration of a unit dose of a compound disclosed herein over a period of several hours (eg, 1-12 hours).
  • the pharmaceutical composition provided by the present invention comprises the compound represented by the formula (I) of the present invention or a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable excipient, and one or more other therapeutic agents.
  • said therapeutic agent is HBV DNA polymerase inhibitor, toll-like receptor 7 modulator, toll-like receptor 8 modulator, toll-like receptor 7 and 8 modulator, toll-like receptor 3 modulator , Interferon ⁇ ligands, HBsAg inhibitors, compounds targeting HbcAg, cyclophilin inhibitors, HBV therapeutic vaccines, HBV preventive vaccines, HBV viral entry inhibitors, NTCP inhibitors, antisense targeting viral mRNA Oligonucleotides, short interfering RNA (siRNA), hepatitis B virus E antigen inhibitors, HBx inhibitors, cccDNA inhibitors, HBV antibodies, thymosin agonists, cytokines, nuclear protein inhibitors, retinoic acid-induced genes 1, NOD2 stimulators, re
  • the one or more therapeutic agents are lamivudine, telbivudine, tenofovir, entecavir, adefovir dipivoxil, tenofovir alafenamide, tenofovir disoproxil , tenofovir alafenamide fumarate, tenofovir alafenamide hemifumarate, Alfaferone, Alloferon, simoleukin, clavudine, emtricitabine, famciclovir, Interferon, Bao Ganling CP, Interferon, Interferon ⁇ -1b, Interferon ⁇ , Interferon ⁇ -2a, Interferon ⁇ -1a, Interferon ⁇ -2, Interleukin-2, Mivotidate , Nitazoxanide, Pegylated Interferon ⁇ -2a, Ribavirin, Rosferon-A, Sizoran, Euforavac, Ampligen, Phosphazid, He
  • Hepatitis B disease refers to liver diseases caused by hepatitis B virus infection or hepatitis B infection, including acute hepatitis, chronic hepatitis, cirrhosis and hepatocellular carcinoma.
  • Acute HBV infection can be asymptomatic or present with symptoms of acute hepatitis.
  • Patients with chronic viral infection have active disease and can develop cirrhosis and hepatocellular carcinoma.
  • the compounds of the present invention can be prepared by the methods described in the present invention, and unless otherwise specified, the definitions of the substituents are as shown in formula (I).
  • the following synthetic schemes and examples are used to further illustrate the content of the present invention.
  • the chromatographic column used a silica gel column, and the silica gel (200-300 mesh) was purchased from Qingdao Ocean Chemical Factory.
  • the nuclear magnetic resonance spectrum uses CDC1 3 , DMSO-d 6 , CD 3 OD or acetone-d 6 as the solvent (reported in ppm), and TMS (0 ppm) or chloroform (7.25 ppm) as the reference standard.
  • MS data is determined by the spectrometer of Agilent6320 series LC-MS equipped with G1312A binary pump and a G1316A TCC (column temperature is kept at 30 °C), and G1329A automatic sampler and G1315B DAD detector are used for analysis, ESI sources are applied to LC-MS spectrometers.
  • the low-resolution mass spectrometry (MS) data was also passed through an Agilent equipped with a G1311A quaternary pump and a G1316A TCC (column temperature maintained at 30°C). 6120 series LC-MS spectrometer is used for determination, G1329A automatic sampler and G1315D DAD detector are used for analysis, and ESI source is used for LC-MS spectrometer.
  • each R 1 , R 2 , R 3 , R 5 , R 6 , X, R 7 , R 8 , R 9 , R and Y have the meanings as described in the present invention, and each X 1 and X 2 are independently Cl, Br or I.
  • Compound C and/or the intermediate represented by compound C-1 can be synthesized by the method disclosed in Synthesis Scheme 1. Under basic conditions (such as N,N-diisopropylethylamine, etc.), compound C1 reacts with compound B or its salt to obtain compound C2 . Then, under the action of base (such as potassium carbonate, sodium carbonate, etc.), compound C2 reacts with 2,4-dimethoxybenzylamine to obtain compound C3 .
  • base such as potassium carbonate, sodium carbonate, etc.
  • Compound (I) can be synthesized by the method disclosed in Synthesis Scheme 2. First, compound C or C-1 is reacted with a base (such as potassium carbonate, etc.) Next, react with compound A to generate compound D1 . Then, compound D1 removes the 2,4-dimethoxybenzyl protecting group on the amino group under acidic conditions (such as trifluoroacetic acid, etc.) to obtain compound (I).
  • a base such as potassium carbonate, etc.
  • compound D1 removes the 2,4-dimethoxybenzyl protecting group on the amino group under acidic conditions (such as trifluoroacetic acid, etc.) to obtain compound (I).
  • Step 1 tert-butyl (R)-(1-hydroxy-2-methylhexan-2-yl)carbamate
  • Step 4 (R)-2-((7-Bromo-2-((2,4-dimethoxybenzyl)amino)pyrido[3,2-d]pyrimidin-4-yl)amino)- 2-Methylhexan-1-ol
  • Step 5 (R)-(2-((2,4-dimethoxybenzyl)amino)-4-((1-hydroxy-2-methylhexan-2-yl)amino)pyrido[ 3,2-d]pyrimidin-7-yl)boronic acid
  • Step 7 N-((4,6-dichloropyridin-3-yl)methyl)-N-methyl-2-phenylethylamine
  • Step 8 4-Chloro-5-((methyl(phenethyl)amino)methyl)pyridin-2(1H)-one
  • N-((4,6-dichloropyridin-3-yl)methyl)-N-methyl-2-phenylethylamine (0.35g, 1.19mmol)
  • potassium tert-butyrate (0.54g, 4.78 mmol)
  • tert-butanol (30 mL)
  • Step 9 4-Chloro-1-methyl-5-((methyl(phenethyl)amino)methyl)pyridin-2(1H)-one
  • Step 10 (R)-4-(2-((2,4-dimethoxybenzyl)amino)-4-((1-hydroxy-2-methylhexan-2-yl)amino)pyridine And[3,2-d]pyrimidin-7-yl)-1-methyl Base-5-((methyl(phenethyl)amino)methyl)pyridin-2(1H)-one
  • Step 11 (R)-4-(2-amino-4-((1-hydroxy-2-methylhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl) -1-methyl-5-((methyl(phenethyl) Amino)methyl)pyridin-2(1H)-one
  • Examples 2-8 can be obtained by referring to the synthesis method of Example 1, and the structures and characterization data of Examples 2-8 are shown in Table 1 below.
  • N-((6-fluoro-4-iodopyridin-3-yl)methyl)-N-methylbut-3-ene-1-methylamine 10.46g, 32.68mmol
  • potassium tert-butyrate 11.00g, 98.04mmol
  • tert-butanol 30mL
  • Step 5 (R)-4-(2-amino-4-((1-hydroxy-2-methylhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl) -5-((but-3-en-1-yl(methyl)amino) methyl)pyridin-2(1H)-one
  • Examples 10 to 27 can be obtained by referring to the synthesis method of Example 9, and the structures and characterization data of Examples 10-27 are shown in Table 2 below
  • Step 1 (R)-5-((but-3-en-1-yl(methyl)amino)methyl)-4-(2-((2,4-dimethoxybenzyl)amino) -4-((1-hydroxy-2-methylhexan-2-yl) amino)pyrido[3,2-d]pyrimidin-7-yl)pyridin-2(1H)-one
  • Step 2 (R)-5-((Butyl(methyl)amino)methyl)-4-(2-((2,4-dimethoxybenzyl)amino)-4-((1- Hydroxy-2-methylhexan-2-yl)amino)pyrido [3,2-d]pyrimidin-7-yl)pyridin-2(1H)-one
  • Step 3 (R)-4-(2-Amino-4-((1-hydroxy-2-methylhex-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl)- 5-((Butyl(methyl)amino)methyl) pyridin -2(1H)-one
  • Step 1 (R)-(2-((2,4-dimethoxybenzyl)amino)-4-((1-hydroxy-2-methylhexan-2-yl)amino)quinazoline -7-yl)boronic acid
  • Step 2 (R)-4-(2-amino-4-((1-hydroxy-2-methylhexan-2-yl)amino)quinazolin-7-yl)-5-((methyl (Phenylethyl)amino)methyl)pyridin -2(1H)-one
  • Examples 30 to 35 can be obtained by referring to the synthesis method of Example 29, and the characterization data of Examples 30-35 are shown in Table 3 below
  • Step 1 5-((But-3-en-1-yl(methyl)amino)methyl)-4-iodo-1-(1-methylpiperidin-4-yl)pyridine-2(1H) -ketone
  • Step 2 (R)-4-(2-amino-4-((1-hydroxy-2-methylhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl) -5-((but-3-en-1-yl(methyl)amino)methyl )-1-(1-methylpiperidin-4-yl)pyridin-2(1H)-one
  • Step 4 (R)-4-(2-amino-4-((1-hydroxy-2-methylhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl) -N-(but-3-en-1-yl)-N-methyl-6-oxo- 1,6-dihydropyridine-3-carboxamide
  • Step 3 (R)-4-(2-amino-4-((1-hydroxy-2-methylhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl) -5-(piperidin-1-carbonyl)pyridin-2(1H) -one
  • Step 4 (R)-N-((4-(2-amino-4-((1-hydroxy-2-methylhexan-2-yl)amino)pyrido[3,2-d]pyrimidine- 7-yl)-6-oxo-1,6-dihydropyridin-3- yl)methyl)-N-methylbut-3-enamide
  • N-((6-fluoro-4-iodopyridin-3-yl)methyl)-N-methylbut-3-enamide was used as the starting material to prepare N- ((4-iodo-6-oxo-1,6-dihydropyridin-3-yl)methyl)-N-methylbut-3-enamide, and then N-((4-iodo-6 -Oxo-1,6-dihydropyridin-3-yl)methyl)-N-methylbut-3-enamide and (R)-(2-((2,4-dimethoxybenzyl )amino)-4-((1-hydroxyl-2-methylhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl)boronic acid as starting material, referring to Example 38 Prepared by the synthetic method of Synthesis Step 4, the title compound was obtained as a white solid with a yield of 28%.
  • Step 5 (R)-4-(2-amino-4-((1-hydroxy-2-methylhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl) -5-((but-3-en-1-yloxy)methyl) pyridin-2(1H)-one
  • Step 1 (R)-(2-((2,4-dimethoxybenzyl)amino)-4-((1-hydroxyhexan-2-yl)amino)pyrido[3,2-d ]pyrimidin-7-yl)boronic acid
  • Step 2 (R)-4-(2-amino-4-((1-hydroxyhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl)-5-(( But-3-en-1-yl(methyl)amino)methyl) pyridin-2(1H)-one
  • Step 1 (R)-5-((but-3-en-1-yl(methyl)amino)methyl)-4-(2-((2,4-dimethoxybenzyl)amino) -4-((1-Hydroxyhexan-2-yl)amino)pyrido [3,2-d]pyrimidin-7-yl)pyridin-2(1H)-one
  • Example 1 step Prepared by the synthetic method of 10, the title compound can be obtained as a brown-black solid with a yield of 11%.
  • Step 2 (R)-4-(2-amino-4-((1-hydroxyhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl)-5-(( Butyl(methyl)amino)methyl)pyridin -2(1H)-one
  • Step 2 tert-butyl (R)-(1-amino-2-methyl-1-oxohexan-2-yl)carbamate
  • Step 4 tert-butyl (R)-(2-methyl-1-(1-methyl-1H-pyrazole-4-carboxamido)hexane-2-yl)carbamate
  • Step 7 (R)-N-(2-((2-Amino-7-(5-((but-3-en-1-yl(methyl)amino)methyl)-2-oxo-1 ,2-dihydropyridin-4-yl)pyrido[3,2-d]pyrimidinb4- yl)amino)-2-methylhexyl)-1-methyl-1H-pyrazole-4-carboxamide
  • Examples 45 to 46 can be obtained by referring to the synthesis method of Example 44, and the structures and characterization data of Examples 45-46 are shown in Table 4 below
  • Step 2 (R)-2-((2-Amino-7-(5-((but-3-en-1-yl(methyl)amino)methyl)-2-oxo-1,2- Dihydropyridin-4-yl)pyrido[3,2-d]pyrimidin-4- yl)amino)-2-methylhexyl 1-methyl-1H-pyrazole-4-carboxylate
  • Example 48 can be obtained by referring to the synthesis method of Example 47. The structure and characterization data of Example 48 are shown in Table 5 below
  • 4-iodo-5-((methylamino)methyl)pyridine can be obtained by referring to step 1 to step 3 of Example 56 -2(1H)-one, followed by 4-iodo-5-((methylamino)methyl)pyridin-2(1H)-one and (R)-(2-((2,4-dimethyl Oxybenzyl)amino)-4-((1-hydroxy-2-methylhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl)boronic acid as starting material reference
  • the synthesis method in Step 5 of Example 56 can obtain the title compound as a yellow solid with a total yield of 16%.
  • Example 56 Using 2-fluoro-4-iodo-5-methylpyridine and dimethylamine as starting materials, refer to the synthesis method from step 1 to step 3 of Example 56 to obtain 5-((dimethylamino)methyl)-4-iodo pyridin-2(1H)-one, followed by 5-((dimethylamino)methyl)-4-iodopyridin-2(1H)-one and (R)-(2-((2,4-dimethyl Oxybenzyl)amino)-4-((1-hydroxy-2-methylhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl)boronic acid as starting material reference
  • Example 56 was prepared by the synthesis method in Step 5, and the title compound could be obtained as a yellow solid with a total yield of 33%.
  • Step 3 (R)-4-(2-amino-4-((1-hydroxy-2-methylhexyl-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl)- 5-(methoxymethyl)pyridin-2(1H) -one
  • Step 5 (R)-4-(2-amino-4-((1-hydroxy-2-methylhexyl-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl)- 5-((Methylthio)methyl)pyridin -2(1H)-one
  • Step 1 tert-butyl (R)-(1-methoxy-2-methylhex-2-yl)carbamate
  • Step 2 (R)-4-(2-amino-4-((1-methoxy-2-methylhexyl-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl )-5-((Butyl(methyl)amino)methyl) pyridin-2(1H)-one
  • the starting material can be obtained by referring to the method from step 2 to step 5 of Example 1 to obtain the compound (R)-(2-((2,4-dimethoxybenzyl)amino)-4-((1-methoxy-2 -Methylhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl)boronic acid, followed by (R)-(2-((2,4-dimethoxybenzyl base)amino)-4-((1-methoxy-2-methylhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl)boronic acid and 5-((butyl Base (methyl)amino)methyl)-4-iodopyridin-2(
  • Step 3 4-iodo-5-(((2-methoxyethyl)(methyl)amino)methyl)pyridin-2(1H)-one
  • N-((6-fluoro-4-iodopyridin-3-yl)methyl)-2-methoxy-N-methylethylamine (0.65g, 2.01mmol) and potassium tert-butoxide (0.90 g, 8.02mmol) were mixed with tert-butanol (30mL), placed in an oil bath at 90°C and heated for about 18 hours; 4M hydrochloric acid solution in 1,4-dioxane was added to the system to adjust the pH to 2- 3.
  • Step 4 (R)-4-(2-Amino-4-((1-hydroxy-2-methylhexan-2-yl)amino)quinazolin-7-yl)-5-(((2 -Methoxyethyl)(methyl)amino)methyl) pyridin-2(1H)-one
  • N-((6-fluoro-4-iodopyridin-3-yl)methyl)-N-methylbutan-1-amine (6.50 g, 20.18 mmol) was weighed and dissolved in tert-butanol (50 mL), followed by Potassium tert-butoxide (9.05 g, 80.69 mmol) was added and stirred at room temperature for about 47 hours.
  • Step 5 (R)-4-(2-amino-4-((1-hydroxy-2-methylhexan-2-yl)amino)quinazolin-7-yl)-5-((butyl (Methyl)amino)methyl)-1-methylpyridin -2(1H)-one
  • the method for synthesis from step 2 to step 3 of the examples can be used to obtain 5-( (2-Hydroxyethyl)(methyl)amino)methyl)-4-iodopyridin-2(1H)-one, followed by 5-((2-hydroxyethyl)(methyl)amino)methyl) -4-iodopyridin-2(1H)-one and (R)-(2-((2,4-dimethoxybenzyl)amino)-4-((1-hydroxy-2-methylhexane -2-yl)amino)quinazolin-7-yl)boronic acid as the starting material was synthesized by referring to step 5 of Example 56 to obtain the title compound as a white solid with a yield of 19%.
  • Step 2 N-((4-iodo-6-((4-methoxybenzyl)thio)pyridin-3-yl)methyl)-2-methoxy-N-methylethylamine
  • Step 3 2-((2-((2,4-dimethoxybenzyl)amino)-7-(2-((4-methoxybenzyl)thio)-5-(((2- Methoxyethyl)(methyl)amino)methyl)pyridin-4- yl)quinazolin-4-yl)amino)-2-methylhexan-1-ol
  • Step 4 (R)-4-(2-Amino-4-((1-hydroxy-2-methylhexan-2-yl)amino)quinazolin-7-yl)-5-(((2 -Methoxyethyl)(methyl)amino)methyl) pyridine-2(1H)-thione
  • Step 1 4-iodo-6-methoxynicotine nitrile
  • 6-Methoxynicotinonitrile (1.0g, 7.45mmol) was dissolved in THF (30mL), under nitrogen protection, a tetrahydrofuran solution (4.86mL, 9.71mmol) of lithium diisopropylamide was slowly added dropwise at -78°C, After the drop, the reaction was stirred at -78°C for 1 h. Under this condition, a solution of iodine (3.03 g, 11.92 mmol) in THF (5 mL) was added, and the reaction was continued for 1 h after the drop.
  • Step 6 (R)-4-(2-amino-4-((1-hydroxy-2-methylhexan-2-yl)amino)pyrido[3,2-d]pyrimidin-7-yl) -1-Methyl-5-(piperidin-1-ylmethyl) pyridin-2(1H)-one
  • Test 1 Test for agonistic activity of human TLR7 and TLR8
  • HEK-Blue TM hTLR7 and HEK-Blue TM hTLR8 cells were used to detect the agonistic and cytotoxic activities of compounds on hTLR7 and hTLR8.
  • Compound preparation the compound was first prepared into a 20 mM stock solution with DMSO, and then the compound was diluted 3-fold into a 96-well plate, with a total of 10 concentrations, and each concentration was duplicated. Add 0.5 ⁇ L DMSO to each negative control well. The final concentration of DMSO was 0.5%.
  • QUANTI-Blue detection solution Take 1mL of QB reagent and 1mL of QB buffer, add 98mL of sterile water, mix and dissolve, and place at room temperature for 10min.
  • Compound activity test Take 20 ⁇ L of the culture supernatant in step 2) from each well and add it to a 96-well plate containing 180 ⁇ L of QUANTI-Blue detection solution. Absorbance at 650 nm (OD 650 ) was detected.
  • Detection of cell viability operate according to the instructions of Celltiter-Glo, and detect the chemiluminescent signal (RLU) with a multifunctional microplate reader Flextation III.
  • Compound activity The OD 650 value was analyzed by GraphPad Prism software, and the dose-response curve of the compound was fitted to calculate the EC 50 value of the compound.
  • Cell Viability Detection The calculation formula of cell viability % is as follows. The cell activity % value was analyzed with GraphPad Prism software, and the dose-effect curve of the compound was fitted to calculate the CC 50 value of the compound on the cells.
  • the compound of the present invention has better agonistic activity to hTLR8, and has better selective activation to hTLR8, the test results of the agonistic activity of the compound of the present invention to human TLR7 and TLR8 are shown in Table A, and the compound of the present invention has a positive effect on cells The toxicity is small, and the toxicity results of the compounds of the present invention to cells are shown in Table A-1.
  • Table A Agonistic activity of compounds of the present invention on human TLR7 and TLR8
  • Test 2 Automatic patch clamp detection of the effect of compounds on the hERG channel current stably expressed in HEK293 cells
  • the hERG cells were clamped by the patch clamp technique, and after forming a high-resistance seal, the membrane was ruptured to record the hERG current in the whole-cell recording mode.
  • Each drug concentration was set to be administered twice, and the time was at least 5 minutes.
  • the test concentration was 0.3 ⁇ M, 1 ⁇ M, 3 ⁇ M, 10 ⁇ M and 30 ⁇ M, and the changes of the test compound on the hERG current were observed. Refer to Table B for the test results.
  • the comparative compound is (R)-2-((2-amino-7-fluoropyrido[3,2-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol
  • Test 3 Inhibitory effect of compounds of the present invention on human liver microsomal CYP enzymes
  • the human liver microsome system was used to evaluate the inhibitory effect of test compounds on the main metabolic enzymes CYP1A2, CYP2C19, CYP2D6 and CYP3A4 in microsomes.
  • the concentration of the test substance in the final incubation system is 10 ⁇ M, and it is incubated with the suspension of human liver microsomes containing cytochrome P450 enzymes CYP1A2, CYP2C19, CYP2D6 and CYP3A4 respectively.
  • the known substrates are metabolized by corresponding single enzymes to generate specific metabolites, and the metabolites are determined by UPLC-MS/MS method.
  • the relative inhibition rate (Relative Inhibition (%)) was calculated by the reduction percentage of the metabolite generation of the test compound and the solvent DMSO in the same time.
  • Relative inhibition rate% (1-(N +inh /N veh )) ⁇ 100
  • N is the concentration value of the metabolite of the probe substrate and it is assumed that the bloody product with or without the inhibitor group is 0 at 0 min.
  • N +inh is the concentration of metabolites in the inhibitor group, and N veh is the concentration of metabolites in the group without inhibitors. See Table C for the results of the inhibition test of the compounds of the present invention on human liver microsomal CYP enzymes.
  • Test 4 Induction effect of the compounds of the present invention on human liver microsomal CYP enzymes
  • Experimental purpose Using frozen human hepatocytes as a test system, evaluate the inducing effect of the test substance on CYP1A2, CYP2B6 and CYP3A4 from two aspects of enzyme activity and mRNA level.
  • Experimental method 24 hours after cell plating, add medium containing the compound to be tested to start induction (test concentration is 10, 1 and 0.1 ⁇ M), change a fresh drug-containing medium every 24 hours, and after 72 hours of induction, culture
  • the base was replaced with a medium containing CYP1A2, CYP2B6 and CYP3A4 specific substrates for 30 minutes of incubation, 100 ⁇ L was taken for treatment, and the labeled metabolites of the substrates were detected for enzyme activity level evaluation.
  • the cells were lysed, reverse transcribed, and the expression levels of CYP1A2, CYP2B6 and CYP3A4 genes in the cells were evaluated by real-time quantitative PCR.
  • the induction fold of the test compound Compared with the blank control, the induction fold of the test compound, and compared with the positive control, the induction percentage of the test compound is used as the basis for evaluating the induction potential of the test compound.
  • the positive control of CYP1A2 is: omeprazole; the positive control of CYP2B6 is: phenobarbital; the positive control of CYP3A4 is: rifampicin. See Table D for the experimental results of the induction test of human liver microsomal CYP enzymes.
  • Table D Experimental data of the induction test of the compounds of the present invention on human liver microsomal CYP enzymes
  • the comparative compound is (R)-2-((2-amino-7-fluoropyrido[3,2-d]pyrimidin-4-yl)amino)-2-methylhexan-1-ol
  • the experimental data show that the compound of the present invention has basically no induction effect on liver drug enzymes CYP1A2, CYP2B6 and CYP3A4.
  • Test 5 Pharmacokinetic experiments of compounds of the present invention in beagle dogs, mice, rats or cynomolgus monkeys
  • the PK of the compounds of the present invention in Beagle dogs (purchased from Hunan Slake Jingda Experimental Animal Co., Ltd., body weight 10-12kg, male, age 10-12 months, 3 for oral administration, 3 for intravenous injection) Determination of experimental methods:
  • Beagle dogs were given 2.5 mg/kg or 5 mg/kg orally or 0.5 mg/kg or 1 mg/kg or 2 mg/kg of the test compound intravenously.
  • ICR mice were given test compound at 10 mg/kg by oral gavage or by tail vein injection at 2 mg/kg or 10 mg/kg.
  • Plasma samples were quantitatively analyzed by multiple reaction monitoring (MRM) on a triple quadrupole tandem mass spectrometer. Pharmacokinetic parameters were calculated using WinNonlin 6.3 software with non-compartmental model method.
  • the PK of the compound of the present invention in SD rats (purchased from Hunan Slake Jingda Experimental Animal Co., Ltd., body weight 200-250g, male, age 2-3 months, 3 for oral administration, 3 for intravenous injection) Determination of experimental methods:
  • Plasma samples were quantitatively analyzed by multiple reaction monitoring (MRM) on a triple quadrupole tandem mass spectrometer. Pharmacokinetic parameters were calculated using WinNonlin 6.3 software with non-compartmental model method.
  • the compound of the present invention is effective in cynomolgus monkeys (purchased from Guangdong Chunsheng Biotechnology Development Co., Ltd., body weight 3-6kg, male, age 4-6 years old, oral 3 animals in each group, 3 mice in each group for intravenous injection) PK determination test method in vivo:
  • Cynomolgus monkeys were orally administered 2.5 mg/kg or 5 mg/kg or intravenously with 0.5 mg/kg or 1 mg/kg of the test compound.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • Pulmonology (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • Epidemiology (AREA)
  • Rheumatology (AREA)
  • Dermatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Diabetes (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Obesity (AREA)
  • AIDS & HIV (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Child & Adolescent Psychology (AREA)
  • Pain & Pain Management (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

一种嘧啶并芳香环类化合物及其在药物中的应用,尤其是作为TLR8激动剂的应用。具体地说,涉及通式(I)所示的化合物或其立体异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐或它的前药,以及它们作为药物的用途,尤其是作为TLR8激动剂的应用。

Description

嘧啶并芳香环类化合物及其在药物中的应用 技术领域
本发明属于医药领域。具体涉及一种嘧啶并芳香环类化合物及其作为药物的用途,尤其是作为TLR8激动剂的用途。本发明还涉及这些嘧啶并芳香环类化合物同其他治疗剂组成的组合物,及其作为药物的用途,尤其是作为TLR8激动剂的用途。
背景技术
Toll样受体(toll-like receptors,TLRs)是先天免疫应答的一个重要模式识别受体,广泛分布于哺乳动物的髓样树突状细胞,单核细胞以及单核巨噬细胞中。一方面,TLRs能识别特定微生物PAMPs(例如:脂多糖、鞭毛蛋白、单/双链RNA等),从而激发机体先天免疫;另一方面,不同TLRs能够诱导特定功能区域基因表达,从而激发机体抗原特异性获得性免疫应答。
在哺乳动物中,已发现13个TLRs成员,其中TLR1-TLR9、TLR11为人类和老鼠共有,TLR10、TLR12和TLR13为老鼠特有。TLR8是TLRs亚组(TLRs 3、7、8和9)的成员,局限于专门识别非己核酸的细胞的内涵体隔室。TLR8在人身上主要通过单核细胞、NK细胞和髓样树突细胞(mDC)表达。TLR8激动剂可以导致各种不同的促炎细胞因子的释放,如IL-6、IL-12、TNF-α和IFN-γ。
TLR8激活后介导炎症免疫促进体内病毒感染的细胞和肿瘤细胞的清除,其激动剂可作为独立的免疫治疗药物或者免疫佐剂,在免疫治疗中展现出重要的临床应用前景。TLR8激活与抗感染天然免疫反应密切相关,可介导HBV、HCV、HIV、疱疹病毒等病毒感染疾病、肿瘤、自身免疫性疾病,以及代谢性疾病等的发生发展。
目前TLR8和TLR7的双重激动剂在很多专利里都有报道,鉴于TLR8激动剂药物的广泛治疗潜力,仍然需要有新的TLR8激动剂药物,尤其是用作治疗和/或预防乙型肝炎病毒的对TLR8有较高选择性的激动剂药物。
发明内容
本发明涉及新型的嘧啶并芳香环类化合物,及其药学上可接受的组合物,其对TLR8具有较好的激活作用,并对TLR8具有较好的选择性激活作用,同时它们还具有对肝药酶基本无诱导作用、对肝药酶基本无抑制作用和对心脏基本无毒性等优点。另外,它们还具有溶解性好、稳定性好和非常好的药代动力学性质。本发明化合物可治疗和/或预防与TLR8活性相关的各种疾病,尤其在抗乙型肝炎病毒(HBV)方面具有很好的应用前景。
一方面,本发明涉及一种如式(I)所示的化合物或如式(I)所示的化合物的立体异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐或它的前药,
其中,X为N或CR4
各R1、R2、R4、R5、R6、R7和R8独立地为氢、氘、F、Cl、Br、I、羟基、氰基、氨基、C1-4烷氨基、C1-6烷氧基或C1-6烷基,其中,所述的C1-4烷氨基、C1-6烷氧基和C1-6烷基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基取代;
Y为O或S;
R3为-C1-6亚烷基-R10,其中,所述的-C1-6亚烷基-R10中的-C1-6亚烷基-未被取代或被1、2、3、4或5个Rw1的取代基取代;
各R10独立地为C1-6烷氧基、-OH、-NH2、-O-C(=O)-R10a或-NRc-C(=O)-R10b,其中所述C1-6烷氧基未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基取代;
各Rc独立地为氢、氘或C1-4烷基,其中,所述的C1-4烷基未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基取代;
各R10a和R10b独立地为C1-6烷基、C3-7环烷基、5-12个环原子组成的杂芳基或3-12个环原子组成的杂环基,其中,所述的C1-6烷基、C3-7环烷基、5-12个环原子组成的杂芳基和3-12个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw2的取代基取代;
R为H、氘、C1-6烷基或3-6个环原子组成的杂环基,其中,所述的C1-6烷基和3-6个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw3的取代基取代;
R9为氢、氘、-L-R11、F、Cl、Br、I、羟基、氰基、氨基、C1-4烷氨基、C1-6烷氧基或C1-6烷基,其中,所述的C1-4烷氨基、C1-6烷氧基和C1-6烷基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基取代;
L为-C1-6亚烷基-或-C(=O)-,其中,所述的-C1-6亚烷基-未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基或异丙基的取代基取代;
R11为-NRaRb、C1-12烷基、-C(=O)R9a、-OR9b、-S(=O)tR9c、C3-7环烷基、5-12个环原子组成的杂芳基或3-12个环原子组成的杂环基,其中,所述的C1-12烷基、C3-7环烷基、5-12个环原子组成的杂芳基和3-12个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw4的取代基取代;
各Ra和Rb独立地为氢、氘、-C(=O)R9d、C1-12烷基、C2-12烯基、C2-12炔基或-C1-6亚烷基-Rd1,其中,所述的C1-12烷基、C2-12烯基、C2-12炔基和-C1-6亚烷基-Rd1中的-C1-6亚烷基-各自独立地未被取代或被1、2、3、4或5个Rw5的取代基取代;
或,Ra、Rb以及与它们所连接的N原子一起形成3-8个环原子组成的杂环基,其中,所述的3-8个环原子组成的杂环基未被取代或被1、2、3或4个Rw4取代;
各R9a、R9b、R9c和R9d独立地为C1-12烷基、C2-12烯基、C2-12炔基或-C1-6亚烷基-Rd2,其中,所述的C1-12烷基、C2-12烯基、C2-12炔基和-C1-6亚烷基-Rd2中的-C1-6亚烷基-各自独立地未被取代或被1、2、3、4或5个Rw6的取代基取代;
各Rd1和Rd2独立地为羟基、C1-6烷氧基、C6-10芳基、5-12个环原子组成的杂芳基、C3-6环烷基或C1-6烷氨基,其中,所述的C1-6烷氧基、C6-10芳基、5-12个环原子组成的杂芳基、C3-6环烷基和C1-6烷氨基各自独立地未被取代或被1、2、3、4或5个Rw7的取代基取代;
各Rw1、Rw2、Rw3、Rw4和Rw7独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、C1-6烷氨基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基、-C(=O)-C1-4烷基、C3-6环烷基、3-6个环原子组成的杂环基或C6-10芳基,其中,所述的C1-6烷氨基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基、-C(=O)-C1-4烷基、C3-6环烷基、3-6个环原子组成的杂环基和C6-10芳基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、C1-4烷基和C1-4烷氨基的取代基取代;
各Rw5和Rw6独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、C1-6烷氨基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基、-C(=O)-C1-4烷基、C2-12烯基、C2-12炔基、C3-6环烷基、3-6个环原子组成的杂环基、5-6个环原子组成的杂芳基或C6-10芳基,其中,所述的氨基、C1-6烷氨基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基、-C(=O)-C1-4烷基、C2-12烯基、C2-12炔基、C3-6环烷基、3-6个环原子组成的杂环基、5-6个环原子组成的杂芳基和C6-10芳基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、C1-4烷基和C1-4烷氨基的取代基取代;
t为0、1或2。
在一些实施例方案中,各R1、R2、R4、R5、R6、R7和R8独立地为氢、氘、F、Cl、Br、I、羟基、氰基、氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基或正己基,其中,所述的N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基和正己基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基和仲丁基的取代基取代;
R为H、氘、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、正己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基或哌嗪基,其中,所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基和哌嗪基各自独立地未被取代或被1、2、3、4或5个Rw3的取代基取代;
其中,各Rw3具有如本发明所述的含义。
在一些实施例方案中,R3为-CH2-R10、-(CH2)2-R10、-(CH2)3-R10、-CH(CH3)CH2-R10、-CH2CH(CH3)-R10或-(CH2)4-R10,其中,所述的-CH2-R10中的-CH2-、-(CH2)2-R10中的-(CH2)2-、-(CH2)3-R10中的-(CH2)3-、-CH(CH3)CH2-R10中的-CH(CH3)CH2-、-CH2CH(CH3)-R10中的-CH2CH(CH3)-和-(CH2)4-R10中的-(CH2)4-各自独立地未被取代或被1、2、3、4或5个Rw1的取代基取代;
各R10独立地为甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、-OH、-NH2、-O-C(=O)-R10a或-NRc-C(=O)-R10b,其中,所述的甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基和2-丁氧基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基和仲丁基的取代基取代;
各R10a和R10b独立地为C1-4烷基、C3-6环烷基、5-6个环原子组成的杂芳基或3-6个环原子组成的杂环基,其中,所述的C1-4烷基、C3-6环烷基、5-6个环原子组成的杂芳基和3-6个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw2的取代基取代;
Rc为氢、氘、甲基、乙基、正丙基或异丙基,其中,所述的甲基、乙基、正丙基和异丙基未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基和异丙基的取代基取代;
其中,各Rw1和Rw2具有如本发明所述的含义。
在一些实施例方案中,各R10a和R10b独立地为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、环丙基、环丁基、环戊基、环己基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基或哌嗪基,其中,所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、环丙基、环丁基、环戊基、环己基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基和哌嗪基各自独立地未被取代或被1、2、3、4或5个Rw2的取代基取代;
其中,各Rw2具有如本发明所述的含义。
在一些实施例方案中,R9为氢、氘、-L-R11、F、Cl、Br、I、羟基、氰基、氨基、C1-4烷氨基、C1-4烷氧基或C1-4烷基,其中,所述的C1-4烷氨基、C1-4烷氧基和C1-4烷基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基所取代;
L为-CH2-、-(CH2)2-、-(CH2)3-、-CH2CH(CH3)-、-CH(CH3)CH2-、-(CH2)4-或-C(=O)-,其中,所述的-CH2-、-(CH2)2-、-(CH2)3-、-CH2CH(CH3)-、-CH(CH3)CH2-和-(CH2)4-各自独立地未被取代或被1、2、3或4个独 立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基或异丙基的取代基取代;
R11为-NRaRb、C1-10烷基、-C(=O)R9a、-OR9b、-S(=O)tR9c、C3-6环烷基、5-6个环原子组成的杂芳基或3-6个环原子组成的杂环基,其中,所述的C1-10烷基、C3-6环烷基、5-6个环原子组成的杂芳基和3-6个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw4的取代基取代;
其中,各Ra、Rb、R9a、R9b、t、R9c和Rw4具有如本发明所述的含义。
在一些实施例方案中,R9为氢、氘、-L-R11、F、Cl、Br、I、羟基、氰基、氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基或正己基,其中,所述的N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基和正己基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基和仲丁基的取代基所取代;
R11为-NRaRb、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、正己基、-C(=O)R9a、-OR9b、-S(=O)tR9c、环丙基、环丁基、环戊基、环己基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基或哌嗪基,其中,所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、环丙基、环丁基、环戊基、环己基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基和哌嗪基各自独立地未被取代或被1、2、3、4或5个Rw4的取代基取代;
其中,各Ra、Rb、R9a、R9b、L、t、R9c和Rw4具有如本发明所述的含义。
在一些实施例方案中,各Ra和Rb独立地为氢、氘、-C(=O)R9d、C1-6烷基、C2-6烯基、C2-6炔基或-C1-4亚烷基-Rd1,其中,所述的C1-6烷基、C2-6烯基、C2-6炔基和-C1-4亚烷基-Rd1中的-C1-4亚烷基-各自独立地未被取代或被1、2、3、4或5个Rw5的取代基取代;
或,Ra、Rb以及与它们所连接的N原子一起形成3-6个环原子组成的杂环基,其中,所述的3-6个环原子组成的杂环基未被取代或被1、2、3或4个Rw4取代;
其中,各R9d、Rd1、Rw5和Rw4具有如本发明所述的含义。
在一些实施例方案中,各Ra和Rb独立地为氢、氘、-C(=O)R9d、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、正己基、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、-CH2-Rd1、-(CH2)2-Rd1、-(CH2)3-Rd1、-CH2CH(CH3)-Rd1、-CH(CH3)CH2-Rd1或-(CH2)4-Rd1,其中,所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、-CH2-Rd1中的-CH2-、-(CH2)2-Rd1中的-(CH2)2-、-(CH2)3-Rd1中的-(CH2)3-、-CH2CH(CH3)-Rd1中的-CH2CH(CH3)-、-CH(CH3)CH2-Rd1中的-CH(CH3)CH2-和-(CH2)4-Rd1中的-(CH2)4-各自独立地未被取代或被1、2、3、4或5个Rw5的取代基取代;
或,Ra、Rb以及与它们所连接的N原子一起形成氮杂环丙基、氮杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、哌啶基、吗啉基、硫代吗啉基或哌嗪基,其中,所述的氮杂环丙基、氮杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、哌啶基、吗啉基、硫代吗啉基和哌嗪基各自独立地未被取代或被1、2、3或4个Rw4取代;
其中,各R9d、Rd1、Rw5和Rw4具有如本发明所述的含义。
在一些实施例方案中,各R9a、R9b、R9c和R9d独立地为C1-6烷基、C2-6烯基、C2-6炔基或-C1-4亚烷基-Rd2,其中所述的C1-6烷基、C2-6烯基、C2-6炔基和-C1-4亚烷基-Rd2中的-C1-4亚烷基-各自独立地未被取代或被1、2、3、4或5个Rw6的取代基取代;
各Rd1和Rd2独立地为羟基、C1-4烷氧基、苯基、5-6个环原子组成的杂芳基、环丙基、环丁基、环戊基、环己基或C1-4烷氨基,其中,所述的C1-4烷氧基、苯基、5-6个环原子组成的杂芳基、环丙基、环丁基、环戊基、环己基和C1-4烷氨基各自独立地未被取代或被1、2、3、4或5个Rw7的取代基取代;
其中,各Rd2、Rw6和Rw7具有如本发明所述的含义。
在一些实施例方案中,各R9a、R9b、R9c和R9d独立地为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、(-C≡CH)、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、-CH2-Rd2、-(CH2)2-Rd2、-(CH2)3-Rd2、-CH2CH(CH3)-Rd2、-CH(CH3)CH2-Rd2或-(CH2)4-Rd2,其中所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、-CH2-Rd2中的-CH2-、-(CH2)2-Rd2中的-(CH2)2-、-(CH2)3-Rd2中的-(CH2)3-、-CH2CH(CH3)-Rd2中的-CH2CH(CH3)-、-CH(CH3)CH2-Rd2中的-CH(CH3)CH2-和-(CH2)4-Rd2中的-(CH2)4-各自独立地未被取代或被1、2、3、4或5个Rw6的取代基取代;
各Rd1和Rd2独立地为羟基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、环丙基、环丁基、环戊基、环己基、N-甲氨基、N-乙氨基、N,N-二甲氨基或N,N-二乙氨基,其中,所述的甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、环丙基、环丁基、环戊基、环己基、N-甲氨基、N-乙氨基、N,N-二甲氨基和N,N-二乙氨基各自独立地未被取代或被1、2、3、4或5个Rw7的取代基取代;
其中,各Rd2、Rw6和Rw7具有如本发明所述的含义。
在一些实施例方案中,各Rw1、Rw2、Rw3、Rw4和Rw7独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、-CH2F、-CH2Cl、-CF3、-CHF2、-CHCl2、-CH2CH2F、-CH2CH2Cl、-CH2CHF2、-CH2CHCl2、-CHFCH2F、-CHClCH2Cl、-CH2CF3、-CH(CF3)2、-CF2CH2CH3、-CH2CH2CH2F、-CH2CH2CHF2、-CH2CH2CF3、-C(=O)-CH3、-C(=O)-CH2CH3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基或苯基,其中,所述的N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、-CH2F、-CH2Cl、-CHF2、-CHCl2、-CH2CH2F、-CH2CH2Cl、-CH2CHF2、-CH2CHCl2、-CHFCH2F、-CHClCH2Cl、-CH2CF3、-CH(CF3)2、-CF2CH2CH3、-CH2CH2CH2F、-CH2CH2CHF2、-CH2CH2CF3、-C(=O)-CH3、-C(=O)-CH2CH3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基和苯基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、-N(CH3)2、-NHCH3、-N(CH2CH3)2、-N(CH3)CH2CH3和-NHCH2CH3的取代基取代;
各Rw5和Rw6独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、C1-4烷氨基、C1-4烷氧基、C1-4烷基、C1-4卤代烷基、-C(=O)-C1-4烷基、C2-6烯基、C2-6炔基、环丙基、环丁基、环戊基、环己基、3-6个环原子组成的杂环基、5-6个环原子组成的杂芳基或苯基,其中,所述的氨基、C1-4烷氨基、C1-4烷氧基、C1-4烷基、C1-4卤代烷基、-C(=O)-C1-4烷基、C2-6烯基、C2-6炔基、环丙基、环丁基、环戊基、环己基、3-6个环原子组成的杂环基、5-6个环原子组成的杂芳基和苯基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、C1-4烷基和C1-4烷氨基的取代基取代。
在一些实施例方案中,各Rw5和Rw6独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l- 丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、-CH2F、-CH2Cl、-CF3、-CHF2、-CHCl2、-CH2CH2F、-CH2CH2Cl、-CH2CHF2、-CH2CHCl2、-CHFCH2F、-CHClCH2Cl、-CH2CF3、-CH(CF3)2、-CF2CH2CH3、-CH2CH2CH2F、-CH2CH2CHF2、-CH2CH2CF3、-C(=O)-CH3、-C(=O)-CH2CH3、-C(=O)-CH2CH2CH3、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基或苯基,其中,所述的氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、-CH2F、-CH2Cl、-CHF2、-CHCl2、-CH2CH2F、-CH2CH2Cl、-CH2CHF2、-CH2CHCl2、-CHFCH2F、-CHClCH2Cl、-CH2CF3、-CH(CF3)2、-CF2CH2CH3、-CH2CH2CH2F、-CH2CH2CHF2、-CH2CH2CF3、-C(=O)-CH3、-C(=O)-CH2CH3、-C(=O)-CH2CH2CH3、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基和苯基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、-N(CH3)2、-NHCH3、-N(CH2CH3)2、-N(CH3)CH2CH3和-NHCH2CH3的取代基取代。
另一方面,本发明还提供了一种药物组合物,包含本发明所述的化合物,及药学上可接受的辅料。
在一些实施方案中,本发明所述的药物组合物,其进一步地包含其它一种或多种治疗剂,其中所述治疗剂为HBV DNA聚合酶抑制剂、toll-样受体7调节剂、toll-样受体8调节剂、toll-样受体7和8调节剂、toll-样受体3调节剂、干扰素ɑ配体、HBsAg抑制剂、靶向HbcAg的化合物、亲环蛋白抑制剂、HBV治疗性疫苗、HBV预防性疫苗、HBV病毒进入抑制剂、NTCP抑制剂、靶向病毒mRNA的反义寡核苷酸、短干扰RNA(siRNA)、乙型肝炎病毒E抗原抑制剂、HBx抑制剂、cccDNA抑制剂、HBV抗体、胸腺素激动剂、细胞因子、核蛋白抑制剂、视黄酸诱导基因1的刺激剂、NOD2刺激剂、重组胸腺素ɑ-1、乙型肝炎病毒复制抑制剂、乙型肝炎表面抗原(HBsAg)分泌或组装抑制剂、IDO抑制剂或其组合。
在一些实施方案中,本发明所述的药物组合物,其中所述的其他一种或多种治疗剂为拉米夫定、替比夫定、替诺福韦、恩替卡韦、阿德福韦酯、替诺福韦艾拉酚胺、替诺福韦二吡呋酯、替诺福韦艾拉酚胺富马酸盐、替诺福韦艾拉酚胺半富马酸盐、Alfaferone、Alloferon、西莫白介素、克拉夫定、恩曲他滨、法昔洛韦、干扰素、宝甘灵CP、因特芬、白细胞介素-2、米伏替酯、硝唑尼特、病毒唑、罗扰素-A、西佐喃、Euforavac、安普利近、Phosphazid、Heplisav、重组人白细胞介素-2、左旋咪唑或丙帕锗。
在一些实施方案中,本发明所述的药物组合物,其中所述的其他一种或多种治疗剂为拉米夫定、替比夫定、替诺福韦,恩替卡韦、阿德福韦酯、替诺福韦艾拉酚胺、替诺福韦二吡呋酯、替诺福韦艾拉酚胺富马酸盐、替诺福韦艾拉酚胺半富马酸盐、Alfaferone、Alloferon、西莫白介素、克拉夫定、恩曲他滨、法昔洛韦、宝甘灵CP、因特芬、干扰素α-1b、干扰素α、干扰素α-2a、干扰素β-1a、干扰素α-2、白细胞介素-2、米伏替酯、硝唑尼特、聚乙二醇干扰素α-2a、病毒唑、罗扰素-A、西佐喃、Euforavac、安普利近、Phosphazid、Heplisav、干扰素α-2b、重组人白细胞介素-2、左旋咪唑或丙帕锗。
另一方面,本发明还提供了所述的化合物或的药物组合物在激活TLR8中的用途。
另一方面,本发明还提供了所述的化合物或的药物组合物在制备试剂盒中的用途,所述试剂盒用于激活TLR8。
另一方面,本发明还提供了所述的化合物或的药物组合物在制备预防、处理、治疗或减轻患者由TLR8介导的疾病的药物中的用途。
在一些实施方案中,所述由TLR8介导的疾病为乙型肝炎病毒感染、丙型肝炎病毒感染、流感病毒感染、疱疹病毒感染、艾滋病毒感染、过敏性疾病、风湿性关节炎、过敏性哮喘、慢性疲劳、II型糖尿病、 枯草热、红瘢狼疮、多发性硬化症、黑色素瘤、肺癌、肝癌、基底细胞癌、肾癌、骨髓瘤、胆道癌、脑癌、乳腺癌、宫颈癌、绒毛膜癌、结肠癌、直肠癌、头颈癌、腹膜肿瘤、输卵管癌、子宫内膜癌、食道癌、胃癌、白血病、淋巴瘤、肉瘤、成神经细胞瘤、口腔癌、卵巢癌、胰腺癌、前列腺癌、睾丸癌、皮肤癌或甲状腺癌。
另一方面,本发明还提供了所述的化合物或药物组合物在制备用于治疗或预防免疫调节系统疾病的药物中的用途。
另一方面,本发明还提供了所述的化合物或药物组合物在制备用于治疗或预防病毒感染或肿瘤的药物中的用途。
另一方面,本发明还提供了所述的化合物或所述的药物组合物在制备用于治疗或预防乙型肝炎病毒感染、丙型肝炎病毒感染、流感病毒感染、疱疹病毒感染、艾滋病毒感染、过敏性疾病、风湿性关节炎、过敏性哮喘、慢性疲劳、II型糖尿病、枯草热、红瘢狼疮、多发性硬化症、黑色素瘤、肺癌、肝癌、基底细胞癌、肾癌、骨髓瘤、胆道癌、脑癌、乳腺癌、宫颈癌、绒毛膜癌、结肠癌、直肠癌、头劲癌、腹膜肿瘤、输卵管癌、子宫内膜癌、食道癌、胃癌、白血病、淋巴瘤、肉瘤、成神经细胞瘤、口腔癌、卵巢癌、胰腺癌、前列腺癌、睾丸癌、皮肤癌和甲状腺癌的药物中的用途。
本发明另一方面涉及式(I)所示的化合物的制备、分离和纯化的方法。
前面所述内容只概述了本发明的某些方面,但并不限于这些方面。这些方面及其他方面的内容将在下面作更加具体完整的描述。
本发明的详细说明书
定义和一般术语
本发明将会把确定的具体化的内容所对应的文献详细列出,实施例都伴随有结构式和化学式的图解。本发明有预期地涵盖所有的选择余地、变体和同等物,这些可能如权利要求所定义的那样包含在现有发明领域。所属领域的技术人员将识别许多类似或等同于在此所描述的方法和物质,这些可以应用于本发明的实践中。本发明绝非限于方法和物质的描述。有很多文献和相似的物质与本发明申请相区别或抵触,其中包括但绝不限于术语的定义,术语的用法,描述的技术,或如本发明申请所控制的范围。
本发明将应用以下定义除非其他方面表明。根据本发明的目的,化学元素根据元素周期表,CAS版本和化学药品手册,75,thEd,1994来定义。另外,有机化学一般原理见"Organic Chemistry,"Thomas Sorrell,University Science Books,Sausalito:1999,and"March's Advanced Organic Chemistry,"by Michael B.Smith and Jerry March,John Wiley&Sons,New York:2007,因此所有的内容通过引用并入本发明。
如本发明所描述的,本发明的化合物可以任选地被一个或多个取代基所取代,如上面的通式化合物,或者如实施例中特殊的例子、子类、和本发明所包含的一类化合物。
另外,需要说明的是,除非以其他方式明确指出,在本文中通篇采用的描述方式“各…和…独立地为”、“…和…各自独立地为”和“…和…分别独立地为”可以互换,应做广义理解,其既可以是指在不同基团中,相同符号之间所表达的具体选项之间互相不影响,也可以表示在相同的基团中,相同符号之间所表达的具体选项之间互相不影响。
在本说明书的各部分,本发明化合物的取代基按照基团种类或范围公开。特别指出,本发明包括这些基团种类和范围的各个成员的每一个独立的次级组合。例如,术语“C1-6烷基”特别指独立公开的甲基、乙基、C3烷基、C4烷基、C5烷基和C6烷基。
本发明使用的术语“烷基”包括1-20个碳原子饱和直链或支链的单价烃基,其中烷基可以独立任选地被一个或多个本发明所描述的取代基所取代。其中一些实施例是,烷基基团含有1-12个碳原子;另外一些实施例是,烷基基团含有1-10个碳原子;另外一些实施例是,烷基基团含有1-8个碳原子;另外一些实施例是,烷基基团含有1-6个碳原子;另外一些实施例是,烷基基团含有1-4个碳原子;另外一些实施例是,烷基基团含有1-3个碳原子。烷基基团更进一步的实例包括,但并不限于,甲基(Me,-CH3)、乙基(Et,-CH2CH3)、正丙基(n-Pr,-CH2CH2CH3)、异丙基(i-Pr,-CH(CH3)2)、正丁基(n-Bu,-CH2CH2CH2CH3)、2-甲基丙基或异丁基(i-Bu,-CH2CH(CH3)2)、1-甲基丙基或仲丁基(s-Bu,-CH(CH3)CH2CH3)、叔丁基(t-Bu, -C(CH3)3)、正戊基(-CH2CH2CH2CH2CH3)、2-戊基(-CH(CH3)CH2CH2CH3)、3-戊基(-CH(CH2CH3)2)、2-甲基-2-丁基(-C(CH3)2CH2CH3)、3-甲基-2-丁基(-CH(CH3)CH(CH3)2)、3-甲基-1-丁基(-CH2CH2CH(CH3)2)、2-甲基-1-丁基(-CH2CH(CH3)CH2CH3)、正己基(-CH2CH2CH2CH2CH2CH3)、2-己基(-CH(CH3)CH2CH2CH2CH3)、3-己基(-CH(CH2CH3)(CH2CH2CH3))、2-甲基-2-戊基(-C(CH3)2CH2CH2CH3)、3-甲基-2-戊基(-CH(CH3)CH(CH3)CH2CH3)、4-甲基-2-戊基(-CH(CH3)CH2CH(CH3)2)、3-甲基-3-戊基(-C(CH3)(CH2CH3)2)、2-甲基-3-戊基(-CH(CH2CH3)CH(CH3)2)、2,3-二甲基-2-丁基(-C(CH3)2CH(CH3)2)、3,3-二甲基-2-丁基(-CH(CH3)C(CH3)3)、正庚基、正辛基等等。
术语“亚烷基”表示从饱和的直链或支链烃基中去掉两个或多个氢原子所得到的饱和的二价或多价烃基基团。除非另外详细说明,亚烷基基团含有1-12个碳原子。在一些实施方案中,亚烷基基团含有1-6个碳原子;在另一些实施方案中,亚烷基基团含有1-4个碳原子;还在一些实施方案中,亚烷基基团含有1-3个碳原子;还在另一些实施方案中,亚烷基基团含有1-2个碳原子。亚烷基的实例包括,但不限于亚甲基(-CH2-)、亚乙基(-CH2CH2-)、亚正丙基(-CH2CH2CH2-)、亚异丙基(-CH(CH3)CH2-)等等。
术语“烯基”表示含有2-12个碳原子,或2-8个碳原子,或2-6个碳原子,或2-4个碳原子的直链或支链的一价烃基,其中至少有一个位置的C-C为sp2双键,其中烯基的基团可以独立地未被取代或被一个或多个本发明所描述的取代基所取代,包括有“顺”、“反”或"Z"、"E"异构体,其中具体的实例包括,但并不限于,乙烯基(-CH=CH2),丙烯基(-CH=CHCH3)、烯丙基(-CH2CH=CH2)、-CH2CH2CH=CH2等等,其中所述烯基基团可以独立地未被取代或被一个或多个本发明所描述的取代基所取代。
术语“炔基”表示含有2-12个碳原子,或2-8个碳原子,或2-6个碳原子,或2-4个碳原子的直链或支链的一价烃基,其中至少有一个位置的C-C为sp三键,其中炔基基团可以独立地未被取代或被一个或多个本发明所描述的取代基所取代,具体的实例包括,但并不限于,乙炔基(-C≡CH)、炔丙基(-CH2C≡CH)、丙炔基(-C≡C-CH3)、1-炔丁基(-CH2CH2C≡CH)、2-炔丁基(-CH2C≡CCH3)、3-炔丁基(-C≡CCH2CH3)等等,其中所述炔基基团可以独立地未被取代或被一个或多个本发明所描述的取代基所取代。
术语“卤代烷基”、“卤代烯基”或“卤代烷氧基”表示烷基、烯基或烷氧基基团被一个或多个卤素原子所取代,其中,烷基、烯基和烷氧基具有本发明所述的含义。这样的实例包含,但并不限于,二氟乙基(-CH2CHF2,-CF2CH3,-CHFCH2F)、三氟乙基(-CH2CF3,-CF2CH2F,-CFHCHF2)、三氟甲基(-CF3)、三氟甲氧基(-OCF3)、氟乙烯基(-CH=CHF,-CF=CH2)等等。
术语“烷氧基”表示烷基基团通过氧原子与分子其余部分相连,其中烷基基团具有如本发明所述的含义。除非另外详细说明,所述烷氧基基团含有1-12个碳原子。在一些实施方案中,烷氧基基团含有1-8个碳原子;在另一些实施方案中,烷氧基基团含有1-6个碳原子;在另一些实施方案中,烷氧基基团含有1-4个碳原子;在又一些实施方案中,烷氧基基团含有1-3个碳原子。所述烷氧基基团可以任选地被一个或多个本发明描述的取代基所取代。
烷氧基基团的实例包括,但并不限于,甲氧基(MeO、-OCH3)、乙氧基(EtO,-OCH2CH3)、1-丙氧基(n-PrO,n-丙氧基,-OCH2CH2CH3)、2-丙氧基(i-PrO,i-丙氧基,-OCH(CH3)2)、1-丁氧基(n-BuO,n-丁氧基,-OCH2CH2CH2CH3)、2-甲基-l-丙氧基(i-BuO,i-丁氧基,-OCH2CH(CH3)2)、2-丁氧基(s-BuO,s-丁氧基,-OCH(CH3)CH2CH3)、2-甲基-2-丙氧基(t-BuO,t-丁氧基,-OC(CH3)3)、1-戊氧基(n-戊氧基,-OCH2CH2CH2CH2CH3)、2-戊氧基(-OCH(CH3)CH2CH2CH3)、3-戊氧基(-OCH(CH2CH3)2)、2-甲基-2-丁氧基(-OC(CH3)2CH2CH3),3-甲基-2-丁氧基(-OCH(CH3)CH(CH3)2)、3-甲基-l-丁氧基(-OCH2CH2CH(CH3)2)、2-甲基-l-丁氧基(-OCH2CH(CH3)CH2CH3)等等。
术语“芳基”可以单独使用或作为“芳烷基”,“芳烷氧基”或“芳氧基烷基”的一大部分,表示含有6-14个碳原子,或6-12个碳原子,或6-10个碳原子的单环,双环,和三环的碳环体系,其中,至少有一个环体系是芳香族的,其中每一个环体系包含3-7个碳原子组成的环,且有一个或多个附着点与分子的其余部分相连。术语“芳基”可以和术语“芳环”或“芳香环”交换使用,如芳基可以包括苯基,萘基和蒽基。所述芳基基团可以独立地未被取代或被一个或多个本发明所描述的取代基所取代。
术语“杂芳基”表示含有5-16个环原子的单环,双环或三环体系,其中至少有一个环体系是芳香族的,且至少有一个环体系包含一个或多个杂原子,其中每一个环体系包含5-7个环原子组成的环,且有一个或 多个附着点与分子其余部分相连。术语“杂芳基”可以与术语“杂芳环”或“杂芳族化合物”交换使用,其包括单环杂芳基、稠合双环杂芳基或多环稠合杂芳基。在一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5-14个环原子组成的杂芳基。在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5-12个环原子组成的杂芳基。在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5-10个环原子组成的杂芳基。在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的8-10个环原子组成的稠合双环杂芳基。在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5-8个环原子组成的杂芳基。在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5-7个环原子组成的杂芳基。在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5-6个环原子组成的单环杂芳基。在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5个环原子组成的杂芳基。在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的6个环原子组成的杂芳基。
另外一些实施例是,杂芳基包括以下的单环基团,但并不限于这些单环基团:2-呋喃基、3-呋喃基、N-咪唑基、2-咪唑基、4-咪唑基、5-咪唑基、3-异噁唑基、4-异噁唑基、5-异噁唑基、2-噁唑基、4-噁唑基、5-噁唑基、N-吡咯基、2-吡咯基、3-吡咯基、2-吡啶基、3-吡啶基、4-吡啶基、2-嘧啶基、4-嘧啶基、5-嘧啶基、哒嗪基(如3-哒嗪基)、2-噻唑基、4-噻唑基、5-噻唑基、四唑基(也称四氮唑基,如5H-四唑基,2H-四唑基)、三唑基(如2-三唑基,5-三唑基,4H-1,2,4-三唑基,1H-1,2,4-三唑基,1,2,3-三唑基)、2-噻吩基、3-噻吩基、吡唑基(如,2-吡唑基和3-吡唑基)、异噻唑基、1,2,3-噁二唑基、1,2,5-噁二唑基、1,2,4-噁二唑基、1,3,4-噁二唑基、1,2,3-硫代二唑基、1,3,4-硫代二唑基、1,2,5-硫代二唑基、吡嗪基、1,3,5-三嗪基;也包括以下的双或者三环基团,但绝不限于这些基团:苯并咪唑基、苯并呋喃基、苯并噻吩基、吲哚基(如2-吲哚基)、嘌呤基、喹啉基(如2-喹啉基,3-喹啉基,4-喹啉基)、异喹啉基(如1-异喹啉基,3-异喹啉基或4-异喹啉基)、吩噁噻基、二苯并咪唑基、二苯并呋喃基、二苯并噻吩基等。所述杂芳基基团任选地被一个或多个本发明所描述的取代基所取代。
术语“M-M1个环原子组成的”表示所述环状基团由M-M1个环原子所组成,所述的环原子包括碳原子和/或O、N、S、P等杂原子。例如,“5-10个环原子组成的杂芳基”代表其包括5、6、7、8、9或10个环原子组成的杂芳基。
术语“杂环基”和“杂环”在此处可交换使用,都是指包含3-12个环原子的饱和或部分不饱和的,非芳香性的单环、双环或三环体系,其中至少有一个环原子选自氮,硫和氧原子,且此环体系有一个或多个连接点与分子的其余部分相连。术语“杂环基”包括单环杂环基、双环稠合杂环基或多环稠合杂环基、螺环杂环基或桥连杂环基,还包括其中杂环可与一个或多个非芳香族碳环或杂环或一个或多个芳环或其组合稠合的多环环系,其中连接的原子团或点在杂环上。双环杂环基包括桥双环杂环基、稠合双环杂环基和螺双环杂环基。除非另外说明,杂环基的-CH2-基团可以任选地被-C(=O)-替代。环的硫原子可以任选地被氧化成S-氧化物。环的氮原子可以任选地被氧化成N-氧化合物。在一些实施方案中,杂环基为3-12个环原子组成的环体系;在一些实施方案中,杂环基为4-7个环原子组成的单环杂环基;在一些实施方案中,杂环基为3-7个环原子组成的单环杂环基;在一些实施方案中,杂环基为4-6个环原子组成的单环杂环基;在一些实施方案中,杂环基为3-6个环原子组成的单环杂环基;在一些实施方案中,杂环基为5-6个环原子组成的单环杂环基;在一些实施方案中,杂环基为7-12个环原子组成的双环杂环基;在一些实施方案中,杂环基为7-12个环原子组成的稠合双环杂环基;在一些实施方案中,杂环基为7-10个环原子组成的稠合双环杂环基;在一些实施方案中,杂环基为8-10个环原子组成的稠合双环杂环基;在一些实施方案中,杂环基为6-10个环原子组成的桥双环杂环基;在其它一些实施方案中,杂环基为3-8个环原子组成的环体系;在其它一些实施方案中,杂环基为3-6个环原子组成的环体系;在其他一些实施方案中,杂环基为5-7个环原子组成的环体系;在其他一些实施方案中,杂环基为5-8个环原子组成的环体系;在其他一些实施方案中,杂环基为6-8个环原子组成的环体系;杂环基为3个环原子组成的环体系;在其他一些实施方案中,杂环基为4个环原子组成的环体系;在其他一些实施方案中,杂环基为5个环原子组成的环体系;在其他一些实施方案中,杂环基为6个环原子组成的环体系;在其他一些实施方案中,杂环基为7个环原子组成的环体系;在其他一些实施方案中,杂环基为8个环原子组成的环体系。
杂环的实例包括,但并不限于吡咯烷基、四氢呋喃基、二氢呋喃基、四氢噻吩基、四氢吡喃基、二氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、噻噁烷基、哌嗪基、高哌嗪基、氮杂环丁基、氧杂环丁基、硫杂环丁基、高哌啶基、氧杂环丙基、氮杂环庚基、氧杂环庚基、硫杂环庚基、氧氮杂卓基、二氮杂卓基、硫氮杂卓基、2-吡咯啉基、3-吡咯啉基、二氢吲哚基、2H-吡喃基、4H-吡喃基、二氧杂环己基、1,3-二氧环戊基、吡唑啉基、二噻烷基、二噻茂烷基、二氢噻吩基、吡唑烷基、咪唑啉基、咪唑烷基、1,2,3,4-四氢异喹啉基、3-氮杂双环[3.1.0]己基、3-氮杂双环[4.1.0]庚基、氮杂双环[2.2.2]己基、3H-吲哚基喹嗪基、和N-吡啶基尿素。杂环基团的实例还包括,1,1-二氧硫代吗啉基;其中,环上碳原子被氧代(=O)基团所取代的实例包括,但不限于嘧啶二酮基、1,2,4-噻二唑-5(4H)-酮基、1,2,4-噁二唑-5(4H)-酮基、1H-1,2,4-三唑-5(4H)-酮基等;其中环上碳原子被=S基团所取代的实例包括,但不限于1,2,4-噁二唑-5(4H)-硫酮基、1,3,4-噁二唑-2(3H)-硫酮基等。所述的杂环基基团可以任选地被一个或多个本发明所描述的取代基所取代。
术语“稠合双环”是指单价或多价的,饱和、部分不饱和或完全不饱和的非芳香性或芳香性的环体系,所述环体系中的两个环共用两个相邻的环原子。
术语“螺环基”,“螺环”,“螺双环基”或“螺双环”在此处可交换使用,是指单价或多价的,饱和或部分不饱和的非芳香性的环体系,其中一个环起源于另一个环上特定的环碳原子,并且两个环只共用一个原子。
例如,像下面式a所描述的,环B和B′被称为“稠合双环”,而环A′和环B共用一个碳原子,被称为“螺环”或“螺双环”。稠合双环基和螺双环基的每个环都可以是碳环基或杂环基,并且每个环任选地被一个或多个本发明所描述的取代基所取代。
术语“稠合双环杂环基”和“双环稠合杂环基”可以交换使用,表示单价的饱和或部分不饱和的非芳香性并环体系。这样的体系可以包含独立的或共轭的不饱和状态,但其核心结构不包含芳香环或芳杂环(但是芳香族可以作为其上的取代基)。所述环体系中的每一个环包含3-7个原子,且至少一个环包含一个或多个杂原子,即包含1-6个碳原子和选自N,O,P,S的1-3个杂原子,在此S或P任选地被一个或多个氧原子所取代得到像SO,SO2,PO,PO2的基团,在一些实施方案中,稠合双环杂环基为7-10个环原子组成的稠合双环杂环基;在一些实施方案中,稠合双杂环基为8-10个环原子组成的稠合双环杂环基;这样的实例包括,但并不限于,3-氮杂稠合[3.1.0]己烷、3-氮杂双环[3.3.0]辛烷、六氢-呋喃[3,4-c]吡咯基、六氢-噻吩[3,4-c]吡咯基、3,4,5,6-四氢-环戊烷[c]噻吩基等。所述稠合杂双环基任选地被一个或多个本发明所描述的取代基所取代。
术语“桥双环基”或“桥双环”表示饱和或部分不饱和的非芳香性的桥环体系,如式b所示,即环A1与环A2共有一个烷链、杂原子或一个杂烷链,其中j为1、2、3或4,X3为烷链、杂原子或杂烷链。这样的体系可以包含独立的或共轭的不饱和状态,但其核心结构不包含芳香环或芳杂环(但是芳香族可以作为其上的取代基)。其中每一个环,如A1或A2,包含3-7个原子,这样的实例包括,但并不限于,双环[2.2.1]庚烷基,2-甲基-二氮杂二环[2.2.1]庚烷基等。所述桥双环基任选地被一个或多个本发明所描述的取代基所取代。
术语“桥双环杂环基”表示饱和或部分不饱和的非芳香性的桥双环体系,其中每一个环包含3-7个原子,且至少一个环包含一个或多个杂原子,即包含1-6个碳原子和选自N,O,P,S的1-3个杂原子,在此S或P任选地被一个或多个氧原子所取代得到像SO,SO2,PO,PO2的基团,在一些实施方案中,桥双环杂环基为6-10个环原子组成的桥双环杂环基,这样的实例包括,但并不限于2-氧代-5-氮杂双环[2.2.1]庚烷基、 2-硫代-5-氮杂双环[2.2.1]庚烷基、2-氧代-5-氮杂双环[2.2.1]庚烷基,2,5-二氮杂双环[2.2.1]庚烷基、2-甲基-2,5-二氮杂双环[2.2.1]庚烷基。所述桥双环杂环基任选地被一个或多个本发明所描述的取代基所取代。
术语“环烷基”是指有一个或多个连接点连接到分子的其余部分,饱和的,含有3-12个环碳原子的单环,双环或三环体系,包括单环、双环或多环稠合、螺式或桥连环环系。其中一些实施例,环烷基是6-10个原子组成的螺双环烷基;另外一些实施例,环烷基是6-10个原子组成的稠合双环烷基;另外一些实施例,环烷基是含3-10个环碳原子的环体系;另外一些实施例,环烷基是含3-8个环碳原子的环体系;另外一些实施例,环烷基是含3-7个环碳原子的环体系;另外一些实施例,环烷基是含5-8个环碳原子的环体系;另外一些实施例,环烷基是含3-6个环碳原子的环体系;另外一些实施例,环烷基是含5-6个环碳原子的环体系;环烷基基团的实例包含,但并不限于,环丙基,环丁基,环戊基,环己基等等,且所述环烷基基团可以独立地未被取代或被一个或多个本发明所描述的取代基所取代。
术语“烷基氨基”和“烷氨基”可以相互交换使用,其包括“N-烷氨基”和“N,N-二烷氨基”,其中,氨基基团中的氢原子分别独立地被一个或两个烷基基团所取代。其中,一些实施方案是,烷氨基是一个或两个C1-12烷基连接到氮原子上形成的较低级的烷基氨基基团。在另一些实施方案中,烷氨基是一个或两个C1-6烷基连接到氮原子上形成的较低级的烷基氨基基团。在另一些实施方案中,烷氨基是一个或两个C1-4烷基连接到氮原子上形成的较低级的烷基氨基基团。还在另外一些实施方案中,烷氨基是一个或两个C1-3烷基连接到氮原子上形成的较低级的烷基氨基基团。合适的烷氨基基团可以是单烷基氨基或二烷基氨基,烷氨基的实例包括,但并不限于,N-甲氨基(-NHCH3)、N-乙氨基(-NHCH2CH3),N,N-二甲氨基(-N(CH3)2)、N,N-二乙氨基(-N(CH2CH3)2)等等。
除非其他方面表明,本发明所描述的结构式包括所有的同分异构形式(如对映异构,非对映异构,和几何异构(或构象异构)):例如含有不对称中心的R、S构型,双键的(Z)、(E)异构体,和(Z)、(E)的构象异构体。因此,本发明的化合物的单个立体化学异构体或其对映异构体,非对映异构体,或几何异构体(或构象异构体)的混合物都属于本发明的范围。
本发明所使用的术语“前药”,代表一个化合物在体内转化为式(I)所示的化合物。这样的转化受前体药物在血液中水解或在血液或组织中经酶转化为母体结构的影响。本发明前体药物类化合物可以是酯,在现有的发明中酯可以作为前体药物的有苯酯类,脂肪族(C1-24)酯类,酰氧基甲基酯类,碳酸酯,氨基甲酸酯类和氨基酸酯类。例如本发明里的一个化合物包含羟基,即可以将其酰化得到前体药物形式的化合物。其他的前体药物形式包括磷酸酯,如这些磷酸酯类化合物是经母体上的羟基磷酸化得到的。关于前体药物完整的讨论可以参考以下文献:T.Higuchi and V.Stella,Pro-drugs as Novel Delivery Systems,Vol.14 of the A.C.S.Symposium Series,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987,J.Rautio et al,Prodrugs:Design and Clinical Applications,Nature Review Drug Discovery,2008,7,255-270,and S.J.Hecker et al,Prodrugs of Phosphates and Phosphonates,Journal of Medicinal Chemistry,2008,51,2328-2345。
除非其他方面表明,本发明的化合物的所有互变异构形式都包含在本发明的范围之内。另外,除非其他方面表明,本发明所描述的化合物的结构式包括一个或多个不同的原子的富集同位素。
“代谢产物”是指具体的化合物或其盐在体内通过代谢作用所得到的产物。一个化合物的代谢产物可以通过所属领域公知的技术来进行鉴定,其活性可以通过如本发明所描述的那样采用试验的方法进行表征。这样的产物可以是通过给药化合物经过氧化、还原、水解、酰氨化、脱酰氨作用、酯化、脱脂作用、酶裂解等等方法得到。相应地,本发明包括化合物的代谢产物,包括将本发明的化合物与哺乳动物充分接触一段时间所产生的代谢产物。
本发明中立体化学的定义和惯例的使用通常参考以下文献:S.P.Parker,Ed.,McGraw-Hill Dictionary of Chemical Terms(1984)McGraw-Hill Book Company,New York;and Eliel,E.and Wilen,S.,"Stereochemistry of Organic Compounds",John Wiley&Sons,Inc.,New York,1994.本发明的化合物可以包含不对称中心或手性中心,因此存在不同的立体异构体。本发明的化合物所有的立体异构形式,包括但绝不限于,非对映体,对映异构体,阻转异构体,和它们的混合物,如外消旋混合物,组成了本发明的一部分。很多有机化合物都以光学活性形式存在,即它们有能力旋转平面偏振光的平面。在描述光学活性化合物时,前缀D、L或 R、S用来表示分子手性中心的绝对构型。前缀d、l或(+)、(-)用来命名化合物平面偏振光旋转的符号,(-)或l是指化合物是左旋的,前缀(+)或d是指化合物是右旋的。这些立体异构体的化学结构是相同的,但是它们的立体结构不一样。特定的立体异构体可以是对映体,异构体的混合物通常称为对映异构体混合物。50:50的对映体混合物被称为外消旋混合物或外消旋体,这可能导致化学反应过程中没有立体选择性或立体定向性。术语“外消旋混合物”和“外消旋体”是指等摩尔的两个对映异构体的混合物,缺乏光学活性。
术语“互变异构体”或“互变异构的形式”是指不同能量的结构的同分异构体可以通过低能垒互相转化。例如质子互变异构体(即质子移变的互变异构体)包括通过质子迁移的互变,如酮式-烯醇式和亚胺-烯胺的同分异构化作用。原子价(化合价)互变异构体包括重组成键电子的互变。除非另外指出,本发明化合物的所有互变异构体形式都在本发明的范围之内。
本发明所使用的“药学上可接受的盐”是指本发明的化合物的有机盐和无机盐。药学上可接受的盐在所属领域是为我们所熟知的,如文献:S.M.Berge et al.,describe pharmaceutically acceptable salts in detail in J.Pharmaceutical Sciences,66:1-19,1977.所记载的。药学上可接受的无毒的酸形成的盐包括,但并不限于,与氨基基团反应形成的无机酸盐有盐酸盐、氢溴酸盐、磷酸盐、硫酸盐、高氯酸盐、和有机酸盐如乙酸盐、草酸盐、马来酸盐、酒石酸盐、柠檬酸盐、琥珀酸盐、丙二酸盐、或通过书籍文献上所记载的其他方法如离子交换法来得到这些盐。其他药学上可接受的盐包括己二酸盐、苹果酸盐、2-羟基丙酸盐、藻酸盐、抗坏血酸盐、天冬氨酸盐、苯磺酸盐、苯甲酸盐、重硫酸盐、硼酸盐、丁酸盐、樟脑酸盐、樟脑磺酸盐、环戊基丙酸盐、二葡萄糖酸盐、十二烷基硫酸盐、乙磺酸盐、甲酸盐、反丁烯二酸盐、葡庚糖酸盐、甘油磷酸盐、葡萄糖酸盐、半硫酸盐、庚酸盐、己酸盐、氢碘酸盐、2-羟基-乙磺酸盐、乳糖醛酸盐、乳酸盐、月桂酸盐、月桂基硫酸盐、苹果酸盐、丙二酸盐、甲磺酸盐、2-萘磺酸盐、烟酸盐、硝酸盐、油酸盐、棕榈酸盐、扑酸盐、果胶酸盐、过硫酸盐、3-苯基丙酸盐、苦味酸盐、特戊酸盐、丙酸盐、硬脂酸盐、硫氰酸盐、对甲苯磺酸盐、十一酸盐、戊酸盐,等等。通过适当的碱得到的盐包括碱金属,碱土金属,铵和N+(C1-4烷基)4的盐。本发明也拟构思了任何所包含N的基团的化合物所形成的季铵盐。水溶性或油溶性或分散产物可以通过季铵化作用得到。碱金属或碱土金属盐包括钠、锂、钾、钙、镁,等等。药学上可接受的盐进一步包括适当的、无毒的铵,季铵盐和抗平衡离子形成的胺阳离子,如卤化物、氢氧化物、羧化物、硫酸化物、磷酸化物、硝酸化物、C1-8磺酸化物和芳香磺酸化物。
本发明的“溶剂化物”是指一个或多个溶剂分子与本发明的化合物所形成的缔合物。形成溶剂化物的溶剂包括,但并不限于,水、异丙醇、乙醇、甲醇、二甲亚砜、乙酸乙酯、乙酸、氨基乙醇。术语“水合物”是指溶剂分子是水所形成的缔合物。
术语“保护基团”或“Pg”是指一个取代基与别的官能团起反应的时候,通常用来阻断或保护特殊的功能性。例如,“氨基的保护基团”是指一个取代基与氨基基团相连来阻断或保护化合物中氨基的功能性,合适的氨基保护基团包括乙酰基、三氟乙酰基、叔丁氧羰基(BOC)、苄氧羰基(CBZ)和9-芴亚甲氧羰基(Fmoc)。相似地,“羟基保护基团”是指羟基的取代基用来阻断或保护羟基的功能性,合适的保护基团包括乙酰基和甲硅烷基。“羧基保护基团”是指羧基的取代基用来阻断或保护羧基的功能性,一般的羧基保护基包括-CH2CH2SO2Ph、氰基乙基、2-(三甲基硅烷基)乙基、2-(三甲基硅烷基)乙氧基甲基、2-(对甲苯磺酰基)乙基、2-(对硝基苯磺酰基)乙基、2-(二苯基膦基)乙基、硝基乙基,等等。对于保护基团一般的描述可参考文献:T W.Greene,Protective Groups in Organic Synthesis,John Wiley&Sons,New York,1991;and P.J.Kocienski,Protecting Groups,Thieme,Stuttgart,2005。
本发明化合物的描述
本发明所涉及的化合物,及其药学上可接受的组合物,都可以有效激活TLR8,抑制HBV感染。
一方面,本发明涉及一种如式(I)所示的化合物或如式(I)所示的化合物的立体异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐或它的前药,
其中,X为N或CR4
各R1、R2、R4、R5、R6、R7和R8独立地为氢、氘、F、Cl、Br、I、羟基、氰基、氨基、C1-4烷氨基、C1-6烷氧基或C1-6烷基,其中,所述的C1-4烷氨基、C1-6烷氧基和C1-6烷基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基取代;
Y为O或S;
R3为-C1-6亚烷基-R10,其中,所述的-C1-6亚烷基-R10中的-C1-6亚烷基-未被取代或被1、2、3、4或5个Rw1的取代基取代;
各R10独立地为C1-6烷氧基、-OH、-NH2、-O-C(=O)-R10a或-NRc-C(=O)-R10b,其中所述C1-6烷氧基未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基取代;
各Rc独立地为氢、氘或C1-4烷基,其中,所述的C1-4烷基未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基取代;
各R10a和R10b独立地为C1-6烷基、C3-7环烷基、5-12个环原子组成的杂芳基或3-12个环原子组成的杂环基,其中,所述的C1-6烷基、C3-7环烷基、5-12个环原子组成的杂芳基和3-12个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw2的取代基取代;
R为H、氘、C1-6烷基或3-6个环原子组成的杂环基,其中,所述的C1-6烷基和3-6个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw3的取代基取代;
R9为氢、氘、-L-R11、F、Cl、Br、I、羟基、氰基、氨基、C1-4烷氨基、C1-6烷氧基或C1-6烷基,其中,所述的C1-4烷氨基、C1-6烷氧基和C1-6烷基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基取代;
L为-C1-6亚烷基-或-C(=O)-,其中,所述的-C1-6亚烷基-未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基或异丙基的取代基取代;
R11为-NRaRb、C1-12烷基、-C(=O)R9a、-OR9b、-S(=O)tR9c、C3-7环烷基、5-12个环原子组成的杂芳基或3-12个环原子组成的杂环基,其中,所述的C1-12烷基、C3-7环烷基、5-12个环原子组成的杂芳基和3-12个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw4的取代基取代;
各Ra和Rb独立地为氢、氘、-C(=O)R9d、C1-12烷基、C2-12烯基、C2-12炔基或-C1-6亚烷基-Rd1,其中,所述的C1-12烷基、C2-12烯基、C2-12炔基和-C1-6亚烷基-Rd1中的-C1-6亚烷基-各自独立地未被取代或被1、2、3、4或5个Rw5的取代基取代;
或,Ra、Rb以及与它们所连接的N原子一起形成3-8个环原子组成的杂环基,其中,所述的3-8个环原子组成的杂环基未被取代或被1、2、3或4个Rw4取代;
各R9a、R9b、R9c和R9d独立地为C1-12烷基、C2-12烯基、C2-12炔基或-C1-6亚烷基-Rd2,其中,所述的C1-12烷基、C2-12烯基、C2-12炔基和-C1-6亚烷基-Rd2中的-C1-6亚烷基-各自独立地未被取代或被1、2、3、4或5个Rw6的取代基取代;
各Rd1和Rd2独立地为羟基、C1-6烷氧基、C6-10芳基、5-12个环原子组成的杂芳基、C3-6环烷基或C1-6烷氨基,其中,所述的C1-6烷氧基、C6-10芳基、5-12个环原子组成的杂芳基、C3-6环烷基和C1-6烷氨基各自独立地未被取代或被1、2、3、4或5个Rw7的取代基取代;
各Rw1、Rw2、Rw3、Rw4和Rw7独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、C1-6烷氨基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基、-C(=O)-C1-4烷基、C3-6环烷基、3-6个环原子组成的杂环基或C6-10芳基,其中,所述的C1-6烷氨基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基、-C(=O)-C1-4烷基、C3-6环烷基、3-6个环原子组成的杂环基和C6-10芳基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、C1-4烷基和C1-4烷氨基的取代基取代;
各Rw5和Rw6独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、C1-6烷氨基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基、-C(=O)-C1-4烷基、C2-12烯基、C2-12炔基、C3-6环烷基、3-6个环原子组成的杂环基、5-6个环原子组成的杂芳基或C6-10芳基,其中,所述的氨基、C1-6烷氨基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基、-C(=O)-C1-4烷基、C2-12烯基、C2-12炔基、C3-6环烷基、3-6个环原子组成的杂环基、5-6个环原子组成的杂芳基和C6-10芳基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、C1-4烷基和C1-4烷氨基的取代基取代;
t为0、1或2。
在一些实施例方案中,各R1、R2、R4、R5、R6、R7和R8独立地为氢、氘、F、Cl、Br、I、羟基、氰基、氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基或正己基,其中,所述的N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基和正己基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基和仲丁基的取代基取代。
在一些实施例方案中,R为H、氘、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、正己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基或哌嗪基,其中,所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基和哌嗪基各自独立地未被取代或被1、2、3、4或5个Rw3的取代基取代;
其中,各Rw3具有如本发明所述的含义。
在一些实施例方案中,R3为-CH2-R10、-(CH2)2-R10、-(CH2)3-R10、-CH(CH3)CH2-R10、-CH2CH(CH3)-R10或-(CH2)4-R10,其中,所述的-CH2-R10中的-CH2-、-(CH2)2-R10中的-(CH2)2-、-(CH2)3-R10中的-(CH2)3-、-CH(CH3)CH2-R10中的-CH(CH3)CH2-、-CH2CH(CH3)-R10中的-CH2CH(CH3)-和-(CH2)4-R10中的-(CH2)4-各自独立地未被取代或被1、2、3、4或5个Rw1的取代基取代;
其中,各R10和Rw1具有如本发明所述的含义。
在一些实施例方案中,各R10独立地为甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、-OH、-NH2、-O-C(=O)-R10a或-NRc-C(=O)-R10b,其中,所述的甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基和2-丁氧基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基和仲丁基的取代基取代;
其中,各Rc、R10a和R10b具有如本发明所述的含义。
在一些实施例方案中,各R10a和R10b独立地为C1-4烷基、C3-6环烷基、5-6个环原子组成的杂芳基或3-6个环原子组成的杂环基,其中,所述的C1-4烷基、C3-6环烷基、5-6个环原子组成的杂芳基和3-6个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw2的取代基取代;
其中,各Rw2具有如本发明所述的含义。
在一些实施例方案中,Rc为氢、氘、甲基、乙基、正丙基或异丙基,其中,所述的甲基、乙基、正丙基和异丙基未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基和异丙基的取代基取代。
在一些实施例方案中,各R10a和R10b独立地为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁 基、叔丁基、环丙基、环丁基、环戊基、环己基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基或哌嗪基,其中,所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、环丙基、环丁基、环戊基、环己基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基和哌嗪基各自独立地未被取代或被1、2、3、4或5个Rw2的取代基取代;
其中,各Rw2具有如本发明所述的含义。
在一些实施例方案中,R9为氢、氘、-L-R11、F、Cl、Br、I、羟基、氰基、氨基、C1-4烷氨基、C1-4烷氧基或C1-4烷基,其中,所述的C1-4烷氨基、C1-4烷氧基和C1-4烷基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基所取代;
其中,各L和R11具有如本发明所述的含义。
在一些实施例方案中,L为-CH2-、-(CH2)2-、-(CH2)3-、-CH2CH(CH3)-、-CH(CH3)CH2-、-(CH2)4-或-C(=O)-,其中,所述的-CH2-、-(CH2)2-、-(CH2)3-、-CH2CH(CH3)-、-CH(CH3)CH2-和-(CH2)4-各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基或异丙基的取代基取代。
在一些实施例方案中,R11为-NRaRb、C1-10烷基、-C(=O)R9a、-OR9b、-S(=O)tR9c、C3-6环烷基、5-6个环原子组成的杂芳基或3-6个环原子组成的杂环基,其中,所述的C1-10烷基、C3-6环烷基、5-6个环原子组成的杂芳基和3-6个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw4的取代基取代;
其中,各Ra、Rb、R9a、R9b、t、R9c和Rw4具有如本发明所述的含义。
在一些实施例方案中,R9为氢、氘、-L-R11、F、Cl、Br、I、羟基、氰基、氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基或正己基,其中,所述的N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基和正己基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基和仲丁基的取代基所取代;
其中,各L和R11具有如本发明所述的含义。
在一些实施例方案中,R11为-NRaRb、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、正己基、-C(=O)R9a、-OR9b、-S(=O)tR9c、环丙基、环丁基、环戊基、环己基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基或哌嗪基,其中,所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、环丙基、环丁基、环戊基、环己基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基和哌嗪基各自独立地未被取代或被1、2、3、4或5个Rw4的取代基取代;
其中,各Ra、Rb、R9a、R9b、t、R9c和Rw4具有如本发明所述的含义。
在一些实施例方案中,各Ra和Rb独立地为氢、氘、-C(=O)R9d、C1-6烷基、C2-6烯基、C2-6炔基或-C1-4亚烷基-Rd1,其中,所述的C1-6烷基、C2-6烯基、C2-6炔基和-C1-4亚烷基-Rd1中的-C1-4亚烷基-各自独立地未被取代或被1、2、3、4或5个Rw5的取代基取代;
或,Ra、Rb以及与它们所连接的N原子一起形成3-6个环原子组成的杂环基,其中,所述的3-6个环 原子组成的杂环基未被取代或被1、2、3或4个Rw4取代;
其中,各R9d、Rd1、Rw5和Rw4具有如本发明所述的含义。
在一些实施例方案中,各Ra和Rb独立地为氢、氘、-C(=O)R9d、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、正己基、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、-CH2-Rd1、-(CH2)2-Rd1、-(CH2)3-Rd1、-CH2CH(CH3)-Rd1、-CH(CH3)CH2-Rd1或-(CH2)4-Rd1,其中,所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、-CH2-Rd1中的-CH2-、-(CH2)2-Rd1中的-(CH2)2-、-(CH2)3-Rd1中的-(CH2)3-、-CH2CH(CH3)-Rd1中的-CH2CH(CH3)-、-CH(CH3)CH2-Rd1中的-CH(CH3)CH2-和-(CH2)4-Rd1中的-(CH2)4-各自独立地未被取代或被1、2、3、4或5个Rw5的取代基取代;
或,Ra、Rb以及与它们所连接的N原子一起形成氮杂环丙基、氮杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、哌啶基、吗啉基、硫代吗啉基或哌嗪基,其中,所述的氮杂环丙基、氮杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、哌啶基、吗啉基、硫代吗啉基和哌嗪基各自独立地未被取代或被1、2、3或4个Rw4取代;
其中,各R9d、Rd1、Rw5和Rw4具有如本发明所述的含义。
在一些实施例方案中,各R9a、R9b、R9c和R9d独立地为C1-6烷基、C2-6烯基、C2-6炔基或-C1-4亚烷基-Rd2,其中所述的C1-6烷基、C2-6烯基、C2-6炔基和-C1-4亚烷基-Rd2中的-C1-4亚烷基-各自独立地未被取代或被1、2、3、4或5个Rw6的取代基取代;
其中,各Rd2和Rw6具有如本发明所述的含义。
在一些实施例方案中,各Rd1和Rd2独立地为羟基、C1-4烷氧基、苯基、5-6个环原子组成的杂芳基、环丙基、环丁基、环戊基、环己基或C1-4烷氨基,其中,所述的C1-4烷氧基、苯基、5-6个环原子组成的杂芳基、环丙基、环丁基、环戊基、环己基和C1-4烷氨基各自独立地未被取代或被1、2、3、4或5个Rw7的取代基取代;
其中,各Rw7具有如本发明所述的含义。
在一些实施例方案中,各R9a、R9b、R9c和R9d独立地为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、(-C≡CH)、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、-CH2-Rd2、-(CH2)2-Rd2、-(CH2)3-Rd2、-CH2CH(CH3)-Rd2、-CH(CH3)CH2-Rd2或-(CH2)4-Rd2,其中所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、-CH2-Rd2中的-CH2-、-(CH2)2-Rd2中的-(CH2)2-、-(CH2)3-Rd2中的-(CH2)3-、-CH2CH(CH3)-Rd2中的-CH2CH(CH3)-、-CH(CH3)CH2-Rd2中的-CH(CH3)CH2-和-(CH2)4-Rd2中的-(CH2)4-各自独立地未被取代或被1、2、3、4或5个Rw6的取代基取代;
其中,各Rd2和Rw6具有如本发明所述的含义。
在一些实施例方案中,各Rd1和Rd2独立地为羟基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、环丙基、环丁基、环戊基、环己基、N-甲氨基、N-乙氨基、N,N-二甲氨基或N,N-二乙氨基,其中,所述的甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、环丙基、环丁基、环戊基、环己基、N-甲氨基、N-乙氨基、N,N-二甲氨基和N,N-二乙氨基各自独立地未被取代或被1、2、3、4或5个Rw7的取代基取代;
其中,各Rw7具有如本发明所述的含义。
在一些实施例方案中,各Rw1、Rw2、Rw3、Rw4和Rw7独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1- 丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、-CH2F、-CH2Cl、-CF3、-CHF2、-CHCl2、-CH2CH2F、-CH2CH2Cl、-CH2CHF2、-CH2CHCl2、-CHFCH2F、-CHClCH2Cl、-CH2CF3、-CH(CF3)2、-CF2CH2CH3、-CH2CH2CH2F、-CH2CH2CHF2、-CH2CH2CF3、-C(=O)-CH3、-C(=O)-CH2CH3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基或苯基,其中,所述的N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、-CH2F、-CH2Cl、-CHF2、-CHCl2、-CH2CH2F、-CH2CH2Cl、-CH2CHF2、-CH2CHCl2、-CHFCH2F、-CHClCH2Cl、-CH2CF3、-CH(CF3)2、-CF2CH2CH3、-CH2CH2CH2F、-CH2CH2CHF2、-CH2CH2CF3、-C(=O)-CH3、-C(=O)-CH2CH3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基和苯基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、-N(CH3)2、-NHCH3、-N(CH2CH3)2、-N(CH3)CH2CH3和-NHCH2CH3的取代基取代。
在一些实施例方案中,各Rw5和Rw6独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、C1-4烷氨基、C1-4烷氧基、C1-4烷基、C1-4卤代烷基、-C(=O)-C1-4烷基、C2-6烯基、C2-6炔基、环丙基、环丁基、环戊基、环己基、3-6个环原子组成的杂环基、5-6个环原子组成的杂芳基或苯基,其中,所述的氨基、C1-4烷氨基、C1-4烷氧基、C1-4烷基、C1-4卤代烷基、-C(=O)-C1-4烷基、C2-6烯基、C2-6炔基、环丙基、环丁基、环戊基、环己基、3-6个环原子组成的杂环基、5-6个环原子组成的杂芳基和苯基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、C1-4烷基和C1-4烷氨基的取代基取代。
在一些实施例方案中,各Rw5和Rw6独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、-CH2F、-CH2Cl、-CF3、-CHF2、-CHCl2、-CH2CH2F、-CH2CH2Cl、-CH2CHF2、-CH2CHCl2、-CHFCH2F、-CHClCH2Cl、-CH2CF3、-CH(CF3)2、-CF2CH2CH3、-CH2CH2CH2F、-CH2CH2CHF2、-CH2CH2CF3、-C(=O)-CH3、-C(=O)-CH2CH3、-C(=O)-CH2CH2CH3、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基或苯基,其中,所述的氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、-CH2F、-CH2Cl、-CHF2、-CHCl2、-CH2CH2F、-CH2CH2Cl、-CH2CHF2、-CH2CHCl2、-CHFCH2F、-CHClCH2Cl、-CH2CF3、-CH(CF3)2、-CF2CH2CH3、-CH2CH2CH2F、-CH2CH2CHF2、-CH2CH2CF3、-C(=O)-CH3、-C(=O)-CH2CH3、-C(=O)-CH2CH2CH3、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基和苯基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、-N(CH3)2、-NHCH3、-N(CH2CH3)2、-N(CH3)CH2CH3和-NHCH2CH3的取代基取代。
在一实施方案中,本发明包含下列之一结构的化合物或包含下列之一结构的化合物的立体异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐或它的前药:






另一方面,本发明还提供了一种药物组合物,包含本发明所述的化合物,及药学上可接受的辅料。
在一些实施方案中,本发明所述的药物组合物,其进一步地包含其它一种或多种治疗剂,其中所述治疗剂为HBV DNA聚合酶抑制剂、toll-样受体7调节剂、toll-样受体8调节剂、toll-样受体7和8调节剂、toll-样受体3调节剂、干扰素ɑ配体、HBsAg抑制剂、靶向HbcAg的化合物、亲环蛋白抑制剂、HBV治疗性疫苗、HBV预防性疫苗、HBV病毒进入抑制剂、NTCP抑制剂、靶向病毒mRNA的反义寡核苷酸、短干扰RNA(siRNA)、乙型肝炎病毒E抗原抑制剂、HBx抑制剂、cccDNA抑制剂、HBV抗体、胸腺素激动剂、细胞因子、核蛋白抑制剂、视黄酸诱导基因1的刺激剂、NOD2刺激剂、重组胸腺素ɑ-1、乙型肝炎病毒复制抑制剂、乙型肝炎表面抗原(HBsAg)分泌或组装抑制剂、IDO抑制剂或其组合。
在一些实施方案中,本发明所述的药物组合物,其中所述的其他一种或多种治疗剂为拉米夫定、替比夫定、替诺福韦、恩替卡韦、阿德福韦酯、替诺福韦艾拉酚胺、替诺福韦二吡呋酯、替诺福韦艾拉酚胺富马酸盐、替诺福韦艾拉酚胺半富马酸盐、Alfaferone、Alloferon、西莫白介素、克拉夫定、恩曲他滨、法昔洛韦、干扰素、宝甘灵CP、因特芬、白细胞介素-2、米伏替酯、硝唑尼特、病毒唑、罗扰素-A、西佐喃、Euforavac、安普利近、Phosphazid、Heplisav、重组人白细胞介素-2、左旋咪唑或丙帕锗。
在一些实施方案中,本发明所述的干扰素为干扰素α-1b、干扰素α、干扰素α-2a、干扰素β-1a、干扰素α-2、聚乙二醇干扰素α-2a或干扰素α-2b。
另一方面,本发明提出了本发明所述的化合物或药物组合物在激活TLR8中的用途。
另一方面,本发明提出了本发明所述的化合物或药物组合物在制备试剂盒中的用途,所述试剂盒用于激活TLR8。
另一方面,本发明还提供了所述的化合物或的药物组合物在制备预防、处理、治疗或减轻患者由TLR8 介导的疾病的药物中的用途。
在一些实施方案中,所述由TLR8介导的疾病为乙型肝炎病毒感染、丙型肝炎病毒感染、流感病毒感染、疱疹病毒感染、艾滋病毒感染、过敏性疾病、风湿性关节炎、过敏性哮喘、慢性疲劳、II型糖尿病、枯草热、红瘢狼疮、多发性硬化症、黑色素瘤、肺癌、肝癌、基底细胞癌、肾癌、骨髓瘤、胆道癌、脑癌、乳腺癌、宫颈癌、绒毛膜癌、结肠癌、直肠癌、头颈癌、腹膜肿瘤、输卵管癌、子宫内膜癌、食道癌、胃癌、白血病、淋巴瘤、肉瘤、成神经细胞瘤、口腔癌、卵巢癌、胰腺癌、前列腺癌、睾丸癌、皮肤癌或甲状腺癌。
另一方面,本发明还提供了所述的化合物或药物组合物在制备用于治疗或预防免疫调节系统疾病的药物中的用途。
另一方面,本发明还提供了所述的化合物或药物组合物在制备用于治疗或预防病毒感染或肿瘤的药物中的用途。
另一方面,本发明还提供了所述的化合物或所述的药物组合物在制备用于治疗或预防乙型肝炎病毒感染、丙型肝炎病毒感染、流感病毒感染、疱疹病毒感染、艾滋病毒感染、过敏性疾病、风湿性关节炎、过敏性哮喘、慢性疲劳、II型糖尿病、枯草热、红瘢狼疮、多发性硬化症、黑色素瘤、肺癌、肝癌、基底细胞癌、肾癌、骨髓瘤、胆道癌、脑癌、乳腺癌、宫颈癌、绒毛膜癌、结肠癌、直肠癌、头颈癌、腹膜肿瘤、输卵管癌、子宫内膜癌、食道癌、胃癌、白血病、淋巴瘤、肉瘤、成神经细胞瘤、口腔癌、卵巢癌、胰腺癌、前列腺癌、睾丸癌、皮肤癌和甲状腺癌的药物中的用途。
另一方面,本发明还提供了所述的化合物或药物组合物在制备用于预防、治疗或减轻免疫调节系统疾病的药物中的用途,包括给予有效治疗剂量的本发明所述的化合物或本发明所述的药物组合物对患者进行给药。
另一方面,本发明还提供了所述的化合物或药物组合物预防、治疗或减轻患者TLR8介导的疾病的方法,所述方法包含使用药学上可接受的有效剂量的本发明的化合物对患者进行给药。
另一方面,本发明还提供了所述的化合物或药物组合物治疗或预防病毒感染或肿瘤的方法,所述方法包含使用药学上可接受的有效剂量的含有本发明的化合物的药物组合物对患者进行给药。
另一方面,本发明还提供了所述的化合物或所述的药物组合物治疗或预防乙型肝炎病毒感染、丙型肝炎病毒感染、流感病毒感染、疱疹病毒感染、艾滋病毒感染、过敏性疾病、风湿性关节炎、过敏性哮喘、慢性疲劳、II型糖尿病、枯草热、红瘢狼疮、多发性硬化症、黑色素瘤、肺癌、肝癌、基底细胞癌、肾癌、骨髓瘤、胆道癌、脑癌、乳腺癌、宫颈癌、绒毛膜癌、结肠癌、直肠癌、头颈癌、腹膜肿瘤、输卵管癌、子宫内膜癌、食道癌、胃癌、白血病、淋巴瘤、肉瘤、成神经细胞瘤、口腔癌、卵巢癌、胰腺癌、前列腺癌、睾丸癌、皮肤癌和甲状腺癌的方法,所述方法包含使用药学上可接受的有效剂量的含有本发明的化合物的药物组合物对患者进行给药。
另一方面,本发明还提供了所述的化合物或药物组合物预防、治疗或减轻患者免疫调节系统疾病的方法,包括给予有效治疗剂量的本发明所述的化合物或本发明所述的药物组合物对患者进行给药。
另一方面,本发明还提供了使用本发明所述的化合物或的药物组合物作为预防、治疗或减轻患者TLR8介导的疾病的药物。
另一方面,本发明还提供了使用本发明所述的化合物或药物组合物作为预防、治疗或减轻患者免疫调节系统疾病的药物。
另一方面,本发明还提供了使用本发明所述的化合物或药物组合物作为治疗或预防病毒感染或肿瘤的药物。
另一方面,本发明还提供了使用本发明所述的化合物或所述的药物组合物作为治疗或预防乙型肝炎病毒感染、丙型肝炎病毒感染、流感病毒感染、疱疹病毒感染、艾滋病毒感染、过敏性疾病、风湿性关节炎、过敏性哮喘、慢性疲劳、II型糖尿病、枯草热、红瘢狼疮、多发性硬化症、黑色素瘤、肺癌、肝癌、基底细胞癌、肾癌、骨髓瘤、胆道癌、脑癌、乳腺癌、宫颈癌、绒毛膜癌、结肠癌、直肠癌、头颈癌、腹膜肿瘤、输卵管癌、子宫内膜癌、食道癌、胃癌、白血病、淋巴瘤、肉瘤、成神经细胞瘤、口腔癌、卵巢癌、胰腺癌、前列腺癌、睾丸癌、皮肤癌和甲状腺癌的药物。
其中一些实施方案是,所述患者是哺乳动物,另外一些实施例是,所述患者是人类。另外一些实施例 是,所述用途更进一步地包含细胞与治疗剂的接触。
本发明另一方面涉及式(I)所示的化合物的制备、分离和纯化的方法。
本发明还涉及本发明的化合物及其药学上可接受的盐用于生产医药产品来有效治疗本发明所述疾病的应用。本发明的化合物还用于生产一种医药品用来减轻、阻止、控制或治疗患者本发明所述疾病的病症。
除非其他方面表明,本发明的化合物所有的立体异构体,互变异构体,氮氧化物,溶剂化物,代谢产物,药学上可接受的盐和前药都属于本发明的范围。
术语“药学上可接受的”包括物质或组合物必须是适合化学或毒理学地,与组成制剂的其他组分和用于治疗的哺乳动物有关。
本发明的化合物的盐还包括用于制备或纯化式(I)所示化合物的中间体或式(I)所示化合物分离的对映异构体的盐,但不一定是药学上可接受的盐。
如果本发明的化合物是碱性的,则想得到的盐可以通过文献上提供的任何合适的方法制备得到,例如,使用无机酸,如盐酸、氢溴酸、硫酸、硝酸和磷酸等等。或者使用有机酸、如乙酸、马来酸、琥珀酸、扁桃酸、富马酸、丙二酸、丙酮酸、苹果酸、2-羟基丙酸、枸橼酸、草酸、羟乙酸和水杨酸;吡喃糖酸,如葡萄糖醛酸和半乳糖醛酸;α-羟酸,如柠檬酸和酒石酸;氨基酸,如天门冬氨酸和谷氨酸;芳香族酸,如苯甲酸和肉桂酸;磺酸,如对甲苯磺酸、苯磺酸、甲磺酸、乙磺酸、三氟甲磺酸等等或它们的组合。
如果本发明的化合物是酸性的,则想得到的盐可以通过合适的方法制备得到,如,使用无机碱或有机碱,如氨(伯氨,仲氨,叔氨),碱金属氢氧化物,铵,N+(R14)4的盐和碱土金属氢氧化物,等等。合适的盐包括,但并不限于,从氨基酸得到的有机盐,如甘氨酸和精氨酸,氨,如伯氨、仲氨和叔氨,N+(R14)4的盐,如R14是H、C1-4烷基、C6-10芳基、C6-10芳基C1-4烷基等,和环状氨,如哌啶、吗啉和哌嗪等,和从钠、钙、钾、镁、锰、铁、铜、锌、铝和锂得到无机盐。也包括适当的,无毒的铵,季铵盐和抗平衡离子形成的胺阳离子,如卤化物、氢氧化物、羧化物、硫酸化物、磷酸化物、硝酸化物、C1-8磺酸化物和芳香磺酸化物。
本发明的化合物的药物组合物,制剂,给药和化合物及药物组合物的用途
根据另一方面,本发明的药物组合物包括式(I)所示的化合物,本发明所列出的化合物,或实施例化合物,和药学上可接受的辅料。
本发明的药物组合物中化合物能有效地用于TLR8介导的疾病的治疗。对本发明的化合物或药物组合物来说,可能被提及的疾病治疗的区域是,例如:免疫性疾病、病毒感染引起的疾病和肿瘤,例如,乙型肝炎病毒感染、丙型肝炎病毒感染、流感病毒感染、疱疹病毒感染、艾滋病毒感染、过敏性疾病、风湿性关节炎、过敏性哮喘、慢性疲劳、II型糖尿病、枯草热、红瘢狼疮、多发性硬化症、黑色素瘤、肺癌、肝癌、基底细胞癌、肾癌、骨髓瘤、胆道癌、脑癌、乳腺癌、宫颈癌、绒毛膜癌、结肠癌、直肠癌、头颈癌、腹膜肿瘤、输卵管癌、子宫内膜癌、食道癌、胃癌、白血病、淋巴瘤、肉瘤、成神经细胞瘤、口腔癌、卵巢癌、胰腺癌、前列腺癌、睾丸癌、皮肤癌和甲状腺癌。
本发明包括药物制剂,除了无毒,惰性的制药学上合适的辅料外,还含有一种或多种本发明的式(I)所示的化合物或其药物组合物。
上述药物制剂也可以包含式(I)所示的化合物以外的其他活性药物成分。
本发明的化合物存在自由形态,或合适的、作为药学上可接受的衍生物。根据本发明,药学上可接受的衍生物包括,但并不限于,药学上可接受的前药,盐,酯,酯类的盐,或能直接或间接地根据患者的需要给药的其他任何加合物或衍生物,本发明其他方面所描述的化合物,其代谢产物或它的残留物。
像本发明所描述的,本发明的药物组合物包含任何一种本发明的式(I)所示的化合物,进一步包含药学上可接受的辅料,这些辅料,例如像本发明所应用的,包括任何溶剂、固体赋形剂、稀释剂、粘合剂、崩解剂、或其他液体赋形剂、分散剂、矫味剂或悬浮剂、表面活性剂、等渗剂、增稠剂、乳化剂、防腐剂,固体粘合剂或润滑剂,等等,适合于特有的目标剂型。如以下文献所描述的:In Remington:The Science and Practice of Pharmacy,21st edition,2005,ed.D.B.Troy,Lippincott Williams&Wilkins,Philadelphia,and Encyclopedia of Pharmaceutical Technology,eds.J.Swarbrick and J.C.Boylan,1988-1999,Marcel Dekker,New York,综合此处文献的内容,表明不同的辅料可应用于药学上可接受的组合物的制剂和它们公知的制备方法。除了任何常规的辅料与本发明的化合物不相容的范围,例如所产生的任何不良的生物效应或与药 学上可接受的组合物的任何其他组分以有害的方式产生的相互作用,它们的用途也是本发明所考虑的范围。
可作为药学上可接受辅料的物质包括,但并不限于,离子交换剂;铝;硬脂酸铝;卵磷脂;血清蛋白,如人血清蛋白;缓冲物质如磷酸盐;甘氨酸;山梨酸;山梨酸钾;饱和植物脂肪酸的部分甘油酯混合物;水;盐或电解质,如硫酸鱼精蛋白,磷酸氢二钠,磷酸氢钾,氯化钠,锌盐;胶体硅;三硅酸镁;聚乙烯吡咯烷酮;聚丙烯酸脂;蜡;聚乙烯-聚氧丙烯-阻断聚合体;羊毛脂;糖,如乳糖,葡萄糖和蔗糖;淀粉如玉米淀粉和土豆淀粉;纤维素和它的衍生物如羧甲基纤维素钠,乙基纤维素和乙酸纤维素;树胶粉;麦芽;明胶;滑石粉;辅料如可可豆脂和栓剂蜡状物;油如花生油,棉子油,红花油,麻油,橄榄油,玉米油和豆油;二醇类化合物,如丙二醇和聚乙二醇;酯类如乙基油酸酯和乙基月桂酸酯;琼脂;缓冲剂如氢氧化镁和氢氧化铝;海藻酸;无热原的水;等渗盐;林格(氏)溶液;乙醇;磷酸缓冲溶液;和其他无毒的合适的润滑剂如月桂硫酸钠和硬脂酸镁;着色剂;释放剂;包衣衣料;甜味剂;调味剂;香料;防腐剂和抗氧化剂。
本发明化合物的药物组合物,可以以下方面的任意方式施用:口服给药,喷雾吸入法,局部给药,经直肠给药,经鼻给药,局部给药,阴道给药,非肠道给药如皮下,静脉,肌内,腹腔内,鞘内,心室内,胸骨内,或颅内注射或输液,或借助一种外植的储器用药。优选的方式为口服给药,肌注,向腹膜内给药或静脉注射。
本发明化合物或其药物组合物可以是以单位剂量形式给药。给药剂型可以是液体剂型或其他剂型。液体剂型可以是真溶液类、胶体类、微粒剂型、混悬剂型。其他剂型例如片剂、胶囊、滴丸、气雾剂、丸剂、粉剂、溶液剂、混悬剂、乳剂、颗粒剂、栓剂、冻干粉针剂、包合物、埋植剂、贴剂、擦剂等。
口服片剂和胶囊可以含有赋形剂如粘合剂,如糖浆,阿拉伯胶,山梨醇,黄芪胶,或聚乙烯吡咯烷酮;填充剂,如乳糖,蔗糖,玉米淀粉,磷酸钙,山梨醇,氨基乙酸;润滑剂,如硬脂酸镁,滑石,聚乙二醇,硅土;崩解剂,如马铃薯淀粉;或可接受的增润剂如月桂醇钠硫酸盐。片剂可以用制药学上公知的方法包衣。
口服液可以制成水合油的悬浮液,溶液,乳浊液,糖浆或酏剂,也可以制成干品,用前补充水或其它合适的媒质。这种液体制剂可以包含常规的添加剂,如悬浮剂,山梨醇,纤维素甲醚,葡萄糖糖浆,凝胶,羟乙基纤维素,羧甲基纤维素,硬脂酸铝凝胶,氢化的食用油脂,乳化剂,如卵磷脂,山梨聚糖单油酸盐,阿拉伯胶;或非水辅料(可能包含可食用油),如杏仁油,油脂如甘油,乙二醇,或乙醇;防腐剂,如对羟基苯甲酸甲酯或丙酯,山梨酸。如需要可添加调味剂或着色剂。
栓剂可包含常规的栓剂基质,如可可黄油或其他甘油酯。
对胃外投药,液态剂型通常由化合物和一种消毒的辅料制成。辅料首选水。依照所选辅料和药物浓度的不同,化合物既可溶于辅料中也可制成悬浮溶液,在制成注射用溶液时先将化合物溶于水中,过滤消毒后装入封口瓶或安瓿中。
当皮肤局部施用时,本发明化合物可以制成适当的软膏,洗剂,或霜剂的形式,其中活性成分悬浮或溶解于一种或多种的辅料中,其中软膏制剂可以使用的辅料包括但不局限于:矿物油,液体凡士林,白凡士林,丙二醇,聚氧化乙烯,聚氧化丙烯,乳化蜡和水;洗剂和霜剂可使用的辅料包括但不限于:矿物油,脱水山梨糖醇单硬脂酸酯,吐温60,十六烷酯蜡,十六碳烯芳醇,2-辛基十二烷醇,苄醇和水。
可以与非活性成分组合以产生剂型的活性成分的量可以根据预期的治疗对象和特定施用方式而变化。例如,在一些实施方式中,用于口服施用于人的剂型可以含有用适宜和方便量的药学上可接受的赋形剂配制的约1至1000mg的活性物质。在某些实施方式中,药学上可接受的赋形剂在总组合物的约5%至约95%(重量:重量)之间变化。
本发明公开的化合物,例如式(I)所示的化合物,可以根据有效的给药方案向个体施用所需的时间段或持续时间,例如至少约一个月,至少约2个月,至少约3个月,至少约6个月,或至少约12个月或更长。在一个变型中,化合物在个体的寿命期间以每日或间歇的时间表施用。
基于施用医师的判断,可以在治疗过程中调节本发明公开化合物的剂量或给药频率。
化合物可以以有效量施用于个体(例如,人)。在某些实施方式中,化合物每天施用一次。
在某些实施方式中,提供了用于治疗或预防人类疾病或病症的方法,包括与治疗有效量的一种或多种 (例如,一种、二种、三种、四种、一种或两种、一种至三种或一种至四种)另外的治疗剂组合向人施用治疗有效量的本发明公开化合物或其药学上可接受的盐。由于TLR-8的激动剂可用于治疗各种疾病或症状,所以所述另外的治疗剂的特定身份将取决于所治疗的特定疾病或病症。
式(I)所示的化合物可以通过任何有用的途径和方式施用,例如通过口服或肠胃外(例如静脉内)施用。式(I)所示的化合物的治疗有效量为约0.00001mg/kg体重/天至约10mg/kg体重/天,例如约0.0001mg/kg体重/天至约10mg/kg/天或约0.001mg/kg体重/天至约1mg/kg体重/天,或例如约0.01mg/kg体重/天至约1mg/g体重/天或例如约0.05mg/kg体重/天至约0.5mg/kg体重/天。
式(I)所示的化合物的治疗有效量为约0.01mg/剂至约1000mg/剂,例如约0.01mg/剂至约100mg/剂,或例如约0.1mg/剂至约100mg/剂,或例如约1mg/剂至约100mg/剂,或例如约1mg/剂至约10mg/剂。式(I)所示的化合物的其他治疗有效量为约1mg/剂,或约2、3、4、5、6、7、8、9、10、15、20、25、30、35、40、45、50、55、60、65、70、75、80、85、90、95或100mg/剂。式(I)所示的化合物的其他治疗有效量为约100mg/剂,或约125、150、175、200、225、250、275、300、350、400、450或500mg/剂。单剂量可以每小时、每天或每周施用。例如,单剂量可以每1小时施用一次,每2、3、4、6、8、12、16小时或每24小时施用一次。单剂量也可以每1天施用一次,每2、3、4、5、6天或每7天施用一次。单剂量也可以每1周施用一次,每2、3周或每4周施用一次。在某些实施方式中,单剂量可以每周施用一次。单剂量也可以每月施用一次。
式(I)所示的化合物的给药频率将由个体患者的需要决定,例如可以是每天一次或每天两次或更多次。例如,化合物的施用根据持续治疗HBV或HCV感染所需的时间的长度,可以将式(I)所示的化合物施用于感染HBV或HCV的人达20天至180天的时间段,或者例如20天至90天的时间段,或者例如30天到60天的时间段。
施用可以是间歇性的,其中患者在若干天或更多天的时间段内接受每日剂量的式(I)所示的化合物,随后是其中患者不接受每日剂量的化合物的若干天或更多天的时间段。例如,患者可以每隔一天或者每周三次接受化合物的剂量。再次举例来说,患者可以在1至14天的时间段内每天接受化合物的剂量,随后是7至21天的时间段,在此期间患者不接受化合物的剂量,随后是患者再次接受化合物的每日剂量的后续时间段(例如,从1至14天)。可以根据治疗患者的临床要求来重复化合物的施用、随后化合物的不施用的时间段。
在一个实施方式中,提供了一种药学组合物,其包含与一种或多种(例如,一种,两种,三种,四种,一种或两种,一种至三种,或一种至四种)另外的治疗剂组合的本发明公开的化合物或其药学上可接受的盐和药学上可接受的赋形剂。
在一个实施方式中,提供了一种试剂盒,其包含与一种或多种(例如,一种,两种,三种,四种,一种或两种,一种至三种或一种至四种)另外的治疗剂组合的本发明公开的化合物或其药学上可接受的盐。
在某些实施方式中,将本发明公开的化合物或其药学上可接受的盐与一种,两种,三种,四种或更多种另外的治疗剂组合。在某些实施方式中,将本发明公开的化合物或其药学上可接受的盐与两种另外的治疗剂组合。在其它实施方式中,将本发明公开的化合物或其药学上可接受的盐与三种另外的治疗剂组合。在另外的实施方式中,将本发明公开的化合物或其药学上可接受的盐与四种另外的治疗剂组合。所述一种,两种,三种,四种或更多种另外的治疗剂可以是选自同一类别治疗剂的不同治疗剂,和/或它们可以选自不同类别的治疗剂。
在某些实施方式中,当将本发明公开的化合物与本文所述的一种或多种另外的治疗剂组合时,组合物的组分以同时或依序的方案施用。当依序施用时,组合可以以两次或更多次施用来进行施用。
在某些实施方式中,本发明公开的化合物与一种或多种另外的治疗剂在单一剂型中组合以同时施用于患者,例如作为口服施用的固体剂型。
在某些实施方式中,本发明公开的化合物与一种或多种另外的治疗剂一起施用。将本发明公开的化合物与一种或多种另外的治疗剂共同施用通常是指同时或依序施用本发明公开的化合物和一种或多种另外的治疗剂,使得治疗有效量的本发明公开的化合物和一个或多个另外的治疗剂二者都存在于患者体内。
共同施用包括在施用一种或多种另外的治疗剂的单位剂量之前或之后施用本发明公开的化合物的单位剂量,例如在施用一种或多种另外的治疗剂的数秒、数分钟或数小时内施用本发明公开的化合物。例如, 在一些实施方式中,首先施用本发明公开化合物的单位剂量,在随后的数秒或数分钟内施用单位剂量的一种或多种另外的治疗剂。或者,在其它实施方式中,首先施用一种或多种另外的治疗剂的单位剂量,然后在数秒或数分钟内施用单位剂量的本发明公开化合物。在一些实施方式中,首先施用本发明公开化合物的单位剂量,随后在若干小时(例如1-12小时)的时间段之后,施用单位剂量的一种或多种另外的治疗剂。在其它实施方式中,首先施用一种或多种另外的治疗剂的单位剂量,随后在若干小时(例如1-12小时)的时间段之后,施用单位剂量的本发明公开化合物。
本发明提供的药物组合物包含了本发明式(I)所示的化合物或其药学上可接受的盐以及药学上可接受的赋形剂,还包含其它一种或多种治疗剂。其中所述治疗剂为HBV DNA聚合酶抑制剂、toll-样受体7调节剂、toll-样受体8调节剂、toll-样受体7和8调节剂、toll-样受体3调节剂、干扰素α配体、HBsAg抑制剂、靶向HbcAg的化合物、亲环蛋白抑制剂、HBV治疗性疫苗、HBV预防性疫苗、HBV病毒进入抑制剂、NTCP抑制剂、靶向病毒mRNA的反义寡核苷酸、短干扰RNA(siRNA)、乙型肝炎病毒E抗原抑制剂、HBx抑制剂、cccDNA抑制剂、HBV抗体、胸腺素激动剂、细胞因子、核蛋白抑制剂、视黄酸诱导基因1的刺激剂、NOD2刺激剂、重组胸腺素α-1和乙型肝炎病毒复制抑制剂、乙型肝炎表面抗原(HBsAg)分泌或组装抑制剂、IDO抑制剂或其组合。
所述一种或多种治疗剂为拉米夫定、替比夫定、替诺福韦、恩替卡韦、阿德福韦酯、替诺福韦艾拉酚胺、替诺福韦二吡呋酯、替诺福韦艾拉酚胺富马酸盐、替诺福韦艾拉酚胺半富马酸盐、Alfaferone、Alloferon、西莫白介素、克拉夫定、恩曲他滨、法昔洛韦、干扰素、宝甘灵CP、因特芬、干扰素α-1b、干扰素α、干扰素α-2a、干扰素β-1a、干扰素α-2、白细胞介素-2、米伏替酯、硝唑尼特、聚乙二醇干扰素α-2a、病毒唑、罗扰素-A、西佐喃、Euforavac、安普利近、Phosphazid、Heplisav、干扰素α-2b、重组人白细胞介素-2、左旋咪唑或丙帕锗。
本发明另一方面涉及一种本发明的化合物或药物组合物来制备用于免疫性疾病、病毒感染引起的疾病和肿瘤的药品的用途,尤其是预防、治疗或减轻患者乙型肝炎疾病的药品的用途,包括给予患者药学上可接受的有效剂量对患者进行给药。乙型肝炎疾病是指由乙肝病毒感染或乙型肝炎感染导致引起的肝脏疾病,包括急性肝炎、慢性肝炎、肝硬化和肝细胞癌。急性乙型肝炎病毒感染可以是无症状或表现为急性肝炎症状。慢性病毒感染患者患有活动性疾病,可发展为肝硬化和肝癌。
一般合成方法
一般地,本发明的化合物可以通过本发明所描述的方法制备得到,除非有进一步的说明,其中取代基的定义如式(I)所示。下面的合成方案和实施例用于进一步举例说明本发明的内容。
所属领域的技术人员将认识到:本发明所描述的化学反应可以用来合适地制备许多本发明的其他化合物,且用于制备本发明的化合物的其它方法都被认为是在本发明的范围之内。例如,根据本发明那些非例证的化合物的合成可以成功地被所属领域的技术人员通过修饰方法完成,如适当的保护干扰基团,通过利用其他已知的试剂除了本发明所描述的,或将反应条件做一些常规的修改。另外,本发明所公开的反应或已知的反应条件也公认地适用于本发明其他化合物的制备。
下面所描述的实施例,除非其他方面表明所有的温度定为摄氏度(℃)。试剂购买于商品供应商如Aldrich Chemical Company,Arco Chemical Company and Alfa Chemical Company,使用时都没有经过进一步纯化,除非其他方面表明。一般的试剂从汕头西陇化工厂,广东光华化学试剂厂,广州化学试剂厂,天津好寓宇化学品有限公司,青岛腾龙化学试剂有限公司,和青岛海洋化工厂购买得到。
色谱柱使用硅胶柱,硅胶(200-300目)购于青岛海洋化工厂。核磁共振光谱以CDC13,DMSO-d6,CD3OD或丙酮-d6为溶剂(报导以ppm为单位),用TMS(0ppm)或氯仿(7.25ppm)作为参照标准。当出现多重峰的时候,将使用下面的缩写:s(singlet,单峰),q(四重峰),d(doublet,双峰),t(triplet,三重峰),m(multiplet,多重峰),br(broadened,宽峰),dd(doublet of doublets,双二重峰),dt(doublet of triplets,双三重峰),ddd(双双二重峰),tt(三三重峰),br.s(broadened singlet,宽单峰)。偶合常数J,单位用赫兹(Hz)表示。
低分辨率质谱(MS)数据通过配备G1312A二元泵和a G1316A TCC(柱温保持在30℃)的Agilent6320系列LC-MS的光谱仪来测定,G1329A自动采样器和G1315B DAD检测器应用于分析,ESI源应用于LC-MS光谱仪。
低分辨率质谱(MS)数据还通过配备G1311A四元泵和G1316A TCC(柱温保持在30℃)的Agilent 6120系列LC-MS的光谱仪来测定,G1329A自动采样器和G1315D DAD检测器应用于分析,ESI源应用于LC-MS光谱仪。
以上两种光谱仪都配备了Agilent Zorbax SB-C18柱,规格为2.1×30mm,5μm。注射体积是通过样品浓度来确定;流速为0.6mL/min;HPLC的峰值是通过在210nm和254nm处的UV-Vis波长来记录读取的。流动相为0.1%的甲酸乙腈溶液(相A)和0.1%的甲酸超纯水溶液(相B)。梯度洗脱条件如表a所示:表a:梯度洗脱条件
化合物纯化是通过Agilent 1100系列高效液相色谱(HPLC)来评价的,其中UV检测在210nm和254nm处,Zorbax SB-C18柱,规格为2.1×30mm,4μm,10分钟,流速为0.6mL/min,5-95%的(0.1%甲酸乙腈溶液)的(0.1%甲酸水溶液),柱温保持在40℃。
下面简写词的使用贯穿本发明:
MeOH         甲醇                              t-BuOH     叔丁醇
MeOH-d4        氘代甲醇                        DMBNH2 2,4-二甲氧基苄胺
MsCl          甲基磺酰氯                       HCl/EA,HCl/EtOAc 氯化氢的乙酸乙酯溶液
MeI        碘甲烷                              HAUT 2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六
DIAD    偶氮二甲酸二异丙酯                     氟磷酸酯
DCM,CH2Cl2 二氯甲烷                             NBS N-溴代丁二酰亚胺
DCE        二氯乙烷                            ACN 乙腈
PdCl2(dppf) [1,1’-双(二苯基膦)二茂铁]二氯化钯 DIPEA     N,N-二异丙基乙胺
CDC13 氘代氯仿                                 DMF      N,N-二甲基甲酰胺
Boc 叔丁氧羰基                                 EDCI 1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐
(Boc)2O     二碳酸二叔丁酯                     DMAP     4-二甲氨基吡啶
PE 石油醚                                      AIBN     偶氮二异丁腈
EtOAc,EA 乙酸乙酯                              EtOH 乙醇
1atm 101.325kPa                                Et3N,TEA 三乙胺
h      小时                                    mL,ml 毫升
RT,rt 室温                                     THF      四氢呋喃
DME 乙二醇二甲醚                               DMSO      二甲基亚砜
B2Pin2 联硼酸频那醇酯                           Ac2O 乙酸酐
TFA    三氟乙酸                                t1/2       半衰期
KOAc      醋酸钾                               Rt 保留时间
MeNH2      甲胺
t-BuOK     叔丁醇钾
合成方法
以下合成方案列出了制备本发明中公开化合物的实验步骤。其中,各R1、R2、R3、R5、R6、X、R7、R8、R9、R和Y具有如本发明所述的含义,各X1和X2独立地为Cl、Br或I。
合成方案1
化合物C和/或化合物C-1所示中间体可以通过合成方案1所公开的方法合成。在碱性(如N,N-二异丙基乙胺等)条件下,化合物C1与化合物B或其盐反应,得到化合物C2。然后,在碱(如碳酸钾、碳酸钠等)作用下,化合物C2与2,4-二甲氧基苄胺反应,得到化合物C3。最后,在碱性(如醋酸钾)和催化剂(如[1,1’-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物等)条件下,联硼酸频那醇酯与化合物C3反应,得到化合物C和/或C-1
合成方案2
化合物(I)可以通过合成方案2所公开的方法合成。首先,化合物CC-1在碱(如碳酸钾等)和催化剂(如[1,1’-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物等)作用下,与化合物A反应,生成化合物D1。然后,化合物D1在酸性条件下(如三氟乙酸等)脱去氨基上的2,4-二甲氧基苄基保护基,得到化合物(I)。
具体实施方式
以下实施例用于说明本发明,但不用来限制本发明的范围。
制备实施例
在以下制备实施例中,发明人以本发明的部分化合物为例,详细描述了本发明化合物的制备过程。另外,在以下制备实施例中,当化合物名称和化合物结构存在不对应的情况下,以化合物结构为准。
实施例1:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-1-甲基-5-((甲基(苯乙基)氨基)甲基)吡啶-2(1H)-酮
步骤1:(R)-(1-羟基-2-甲基己烷-2-基)氨基甲酸叔丁基酯
称量(2R)-2-氨基-2-甲基己酸盐酸盐(11.86g,65.29mmol)溶解于四氢呋喃(100mL)中,氮气保护,置于0℃并搅拌下缓慢滴加硼烷四氢呋喃(196.00mL,195.87mmol,1M),滴加完毕之后转移到室温下搅拌约3小时后转移到60℃下继续搅拌约2小时;然后转移到0℃下,接着搅拌下缓慢滴加氢氧化钠(13.06 g,326.45mmol),滴加完毕转移到60℃油浴锅中加热搅拌约3小时;然后滴加Boc酸酐(15.68g,71.82mmol)并继续搅拌反应约3小时;停止反应,加入水(200mL)稀释,再用乙酸乙酯(120mL×3)萃取,合并有机相,有机相用饱和食盐水(150mL)洗涤,无水硫酸钠干燥,减压浓缩,残留物经硅胶柱层析(PE/EA(V/V)=10/1)纯化,得到标题化合物为无色油状液体(15.10g,产率99.98%)。
MS(ESI,pos.ion)m/z:254.2[M+Na]+.
步骤2:(R)-2-氨基-2-甲基己烷-1-醇
称量(R)-(1-羟基-2-甲基己烷-2-基)氨基甲酸叔丁基酯(1.44g,6.22mmol)加入到盐酸的1,4-二氧六环溶液(20mL,4M)中,置于室温下搅拌约1小时;减压浓缩除去溶剂,得到标题化合物为油状液体(0.82g,收率100.47%)直接用于下一步反应。
步骤3:(R)-2-((7-溴-2-氯吡啶并[3,2-d]嘧啶-4-基)氨基)-2-甲基己烷-1-醇
分别将7-溴-2,4-二氯吡啶并[3,2-d]嘧啶(1.73g,6.20mmol)和(R)-2-氨基-2-甲基己烷-1-醇(0.81g,6.2mmol)混合于四氢呋喃(30mL)中,接着加入DIPEA(4.01g,31.00mmol),反应混合物置于60℃油浴锅中加热反应过夜;降至室温,再减压浓缩除去溶剂,残留物经硅胶柱层析(PE/EA(V/V)=5/1)纯化,得到标题化合物为黄色固体(1.63g,产率70.36%)。
MS(ESI,pos.ion)m/z:373.0、375.0[M+H]+.
步骤4:(R)-2-((7-溴-2-((2,4-二甲氧基苄基)氨基)吡啶并[3,2-d]嘧啶-4-基)氨基)-2-甲基己烷-1-醇
分别称量(R)-2-((7-溴-2-氯吡啶并[3,2-d]嘧啶-4-基)氨基)-2-甲基己烷-1-醇(1.53g,4.09mmol)、2,4-二甲氧基苄胺(0.79g,4.70mmol)和碳酸钾(1.13g,8.18mmol)混合于1,4-二氧六环(50mL)中,反应混合物置于100℃油浴锅中加热反应约25小时,降至室温,再减压浓缩,残留物经硅胶柱层析(PE/EA(V/V)=3/1)纯化,得到标题化合物为淡黄色固体(2.02g,产率97.91%)。
步骤5:(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸
分别称量(R)-2-((7-溴-2-((2,4-二甲氧基苄基)氨基)吡啶并[3,2-d]嘧啶-4-基)氨基)-2-甲基己烷-1-醇(1.15g,2.28mmol)、联硼酸频那醇酯(0.75g,2.96mmol)、醋酸钾(0.67g,6.84mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯钯(II)(0.17g,0.23mmol)混合于DME(30mL)中,氮气保护,反应混合物置于80℃油浴锅中加热搅拌反应约4小时,硅藻土过滤,二氯甲烷(20mL)洗涤滤饼,接着减压浓缩滤液,浓缩残留物用二氯甲烷(30mL)溶解,所得溶液用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,减压浓缩,得到标题化合物为棕色泡沫状固体(2.09g,产率97.65%),直接用于下一步反应。
MS(ESI,pos.ion)m/z:470.4[M+H]+.
步骤6:1-(4,6-二氯吡啶-3-基)-N-甲基甲胺
称量4,6-二氯吡啶-3-甲醛(5.77g,32.78mmol)溶于无水甲醇(80mL)中,氮气保护,置于0℃下搅拌;接着缓慢滴加30%甲胺的甲醇溶液(33.94g,327.80mmol),继续搅拌反应;接着分批加入硼氢化钠(1.86g,49.17mmol),加入完毕转移到室温下搅拌反应约20小时;停止反应,向反应液中加入水(100mL)稀释,然后用乙酸乙酯(40mL×3)萃取,合并有机相,饱和食盐水(50mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(PE/EA(V/V)=5/1)纯化,得到标题化合物为淡黄色油状液体(4.0g,产率63.87%)。
MS(ESI,pos.ion)m/z:191.0[M+H]+.
步骤7:N-((4,6-二氯吡啶-3-基)甲基)-N-甲基-2-苯基乙胺
分别称量1-(4,6-二氯吡啶-3-基)-N-甲基甲胺(0.67g,3.51mmol)和2-苯基乙醛(0.51g,4.21mmol)加入到DCM(20mL)中,接着加入三乙酰氧基硼氢化钠(2.98g,14.08mmol)。反应混合物于室温下搅拌反应约20小时,向反应液中加入饱和碳酸氢钠溶液(30mL)稀释,然后用二氯甲烷(30mL×3)萃取,合并有机相,合并的有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(PE/EA(V/V)=10/1)纯化,得到标题化合物为无色油状液体(0.35g,产率33.78%)。
MS(ESI,pos.ion)m/z:295.0[M+H]+
步骤8:4-氯-5-((甲基(苯乙基)氨基)甲基)吡啶-2(1H)-酮
分别将N-((4,6-二氯吡啶-3-基)甲基)-N-甲基-2-苯基乙胺(0.35g,1.19mmol)、叔丁酸钾(0.54g,4.78mmol)和叔丁醇(30mL)加入单口瓶中,氮气保护,置于100℃油浴锅中加热回流反应约48小时。停止搅拌,加4M盐酸的1,4-二氧六环溶液调节pH=5-6,减压浓缩,所得残留物经硅胶柱层析(DCM/MeOH(V/V)=15/1)纯化,得到标题化合物为白色固体(0.17g,产率51.62%)。
MS(ESI,pos.ion)m/z:277.1[M+H]+.
步骤9:4-氯-1-甲基-5-((甲基(苯乙基)氨基)甲基)吡啶-2(1H)-酮
称量4-氯-5-((甲基(苯乙基)氨基)甲基)吡啶-2(1H)-酮(1.00g,2.72mmol)溶解于四氢呋喃(50mL)中,置于0℃下搅拌;接着缓慢加入氢化钠(0.22g,5.44mmol),继续搅拌约1小时后加入碘甲烷(0.77g,5.42mmol),移到室温下继续搅拌反应约4小时;停止搅拌,加水(30mL)稀释,乙酸乙酯(30mL×3)萃取,合并有机相,合并的有机相用饱和食盐水(40mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(DCM/MeOH(V/V)=15/1)进行分离提纯,得到标题化合物为棕黄色油状液体(0.51g,产率49.05%)。
MS(ESI,pos.ion)m/z:291.0[M+H]+.
步骤10:(R)-4-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-1-甲 基-5-((甲基(苯乙基)氨基)甲基)吡啶-2(1H)-酮
分别称量4-氯-1-甲基-5-((甲基(苯乙基)氨基)甲基)吡啶-2(1H)-酮(0.35g,0.92mmol)、(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸(0.47g,1.01mmol)、[1,1'-二(二氯苯基)二茂铁钯(II)(0.067g,0.092mmol)和碳酸钾(0.38g,2.75mmol)混合于1,4-二氧六环/水(V/V=10/1,30mL)中,氮气保护,置于65℃油浴锅中加热反应约18小时;降至室温,再减压浓缩,所得残留物经硅胶柱层析(DCM/MeOH(V/V)=15/1)纯化,得到标题化合物为淡黄色固体(0.29g,产率46.37%)。
MS(ESI,pos.ion)m/z:680.4[M+H]+
步骤11:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-1-甲基-5-((甲基(苯乙基) 氨基)甲基)吡啶-2(1H)-酮
称量(R)-4-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-1-甲基-5-((甲基(苯乙基)氨基)甲基)吡啶-2(1H)-酮(0.29g,0.43mmol)加入到TFA(5mL)中,置于室温下搅拌约2小时;氮气吹干溶剂,向反应体系中加入饱和碳酸氢钠(10mL)溶液,二氯甲烷(10mL×3)萃取,合并有机相,饱和食盐水(20mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析 (DCM/MeOH(V/V)=15/1)纯化,得到标题化合物为淡黄色固体(0.11g,产率48.30%)。
MS(ESI,pos.ion)m/z:530.5[M+H]+
1H NMR(400MHz,DMSO-d6):δ(ppm)13.38(s,1H),9.84(s,1H),8.60(s,3H),8.32(s,1H),8.20(s,1H),7.77(s,1H),7.23(dd,J=17.3,7.0Hz,3H),7.09(d,J=6.9Hz,2H),6.47(s,1H),3.74(d,J=10.6Hz,2H),3.51(s,4H),3.06(s,2H),2.79(s,2H),2.53(s,3H),2.08-1.86(m,2H),1.47(s,3H),1.25(s,4H),0.85(s,3H);
HR-MS(ESI):530.3245[M+H]+.
实施例2-8可参考实施例1的合成方法得到,实施例2-8的结构和表征数据见下表1。
表1:实施例2-8的结构和表征数据

实施例9:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁-3-烯-1-基(甲基)氨基)甲基)吡啶-2(1H)-酮
步骤1:5-(溴甲基)-2-氟-4-碘吡啶
分别称量2-氟-4-碘-5-甲基吡啶(20.00g,84.38mmol)、N-溴代琥珀酰亚胺(16.52g,92.82mmol)和2,2'-偶氮二(2-甲基丙腈)(1.39g,8.44mmol)混合于四氯化碳(200mL)中,氮气保护,置于80℃油浴锅中回流反应约14小时;停止搅拌,向反应体系中加入饱和硫代硫酸钠溶液(30mL),然后用二氯甲烷(100mL×3)萃取,合并有机相,饱和食盐水(50mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(PE/EA(V/V)=20/1)纯化,得到标题化合物为白色固体(19.56g,产率73.38%)。
1HNMR(400MHz,CDCl3):δ(ppm)8.25(s,1H),7.51(d,J=3.1Hz,1H),4.58(s,2H).
步骤2:1-(6-氟-4-碘吡啶-3-基)-N-甲基甲胺
往烧瓶加入5-(溴甲基)-2-氟-4-碘吡啶(6.01g,19.02mmol)、甲胺(5.97g,192.3mmol)和甲醇(120mL),于室温搅拌约1.5小时。结束反应,减压蒸干溶剂,往残余物加入饱和碳酸氢钠水溶液(200mL),然后用二氯甲烷(200mL×3)萃取,有机相用无水硫酸钠干燥,减压蒸干,得到标题化合物为黄色油状物(5.04g,产率99.60%)。
MS(ESI,pos.ion)m/z:266.9[M+H]+.
步骤3:N-((6-氟-4-碘吡啶-3-基)甲基)-N-甲基丁-3-烯-1-甲胺
分别称量1-(6-氟-4-碘吡啶-3-基)-N-甲基甲胺(11.28g,42.40mmol)、4-溴-1-丁烯(22.90g,169.6mmol)、碘化钠(0.64g,4.27mmol)和碳酸钾(29.30g,212.04mmol)混合于乙腈(30mL)中,氮气保护,置于70℃油浴锅中加热搅拌约17小时;停止搅拌,减压浓缩,得到标题化合物为无色油状液体(11.00g,产率81.04%)。
MS(ESI,pos.ion)m/z:321.0[M+H]+.
步骤4:5-((丁-3-烯-1-基(甲基)氨基)甲基)-4-碘代吡啶-2(1H)-酮
分别将N-((6-氟-4-碘吡啶-3-基)甲基)-N-甲基丁-3-烯-1-甲胺(10.46g,32.68mmol)、叔丁酸钾(11.00g,98.04mmol)和叔丁醇(30mL)加入于单口瓶中,氮气保护,置于100℃油浴锅中加热回流反应约27小时。停止搅拌,用4M盐酸的1,4-二氧六环溶液将反应液调节pH至5-6,然后减压浓缩,所得残留物经硅胶柱层析(DCM/MeOH(V/V)=15/1)纯化,得到标题化合物为白色固体(6.20g,产率59.63%)。
MS(ESI,pos.ion)m/z:319.1[M+H]+.
步骤5:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁-3-烯-1-基(甲基)氨基) 甲基)吡啶-2(1H)-酮
以5-((丁-3-烯-1-基(甲基)氨基)甲基)-4-碘代吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料,参照实施例1步骤10和步骤11得到标题化合物为白色固体,产率为51%。
MS(ESI,pos.ion)m/z:466.1[M+H]+
1H NMR(400MHz,CDCl3):δ(ppm)8.35(s,1H),7.70(s,1H),7.49(s,1H),7.37(s,1H),6.53(s,1H),5.73-5.63(m,1H),5.42(s,2H),4.92(t,J=14.1Hz,2H),3.88-3.79(m,2H),3.16(s,2H),2.29(t,J=7.3Hz,2H),2.10-1.96(m,6H),1.81(t,J=10.6Hz,1H),1.48-1.31(m,8H),0.93(d,J=7.0Hz,3H).
实施例10至实施例27可参照实施例9的合成方法得到,实施例10-27的结构和表征数据见下表2




实施例28:(R)-4-(2-氨基-4-((1-羟基-2-甲基己-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁基(甲基)氨基)甲基)吡啶-2(1H)-酮
步骤1:(R)-5-((丁-3-烯-1-基(甲基)氨基)甲基)-4-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基) 氨基)吡啶并[3,2-d]嘧啶-7-基)吡啶-2(1H)-酮
以5-((丁-3-烯-1-基(甲基)氨基)甲基)-4-碘代吡啶-2(1H)-酮为原料和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为原料,参考实施例1步骤10的合成方法制备得到。
步骤2:(R)-5-((丁基(甲基)氨基)甲基)-4-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶 并[3,2-d]嘧啶-7-基)吡啶-2(1H)-酮
称量(R)-5-((丁-3-烯-1-基(甲基)氨基)甲基)-4-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)吡啶-2(1H)-酮(0.10g,0.16mmol)和10%钯/碳(20mg,0.16mmol)溶解于THF/MeOH(V/V=1/1)(20mL)中,置于氢气球氛围中反应约20小时;停止搅拌,过滤,得到标题化合物为淡黄色固体(99mg,产率100%)。
MS(ESI,pos.ion)m/z:618.2[M+H]+
步骤3:(R)-4-(2-氨基-4-((1-羟基-2-甲基己-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁基(甲基)氨基)甲基)吡 啶-2(1H)-酮
称量(R)-5-((丁基(甲基)氨基)甲基)-4-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)吡啶-2(1H)-酮(99mg,0.16mmol)加入到TFA(15mL)中,置于室温下搅拌反应约2小时;停止搅拌,氮气吹干溶剂,向反应体系中加入饱和碳酸氢钠(10mL),然后用二氯甲烷(10mL×3)萃取,合并有机相,有机相用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(DCM/MeOH(V/V)=15/1)纯化,得到标题化合物为淡黄色固体(0.060g,产率80.20%)。
MS(ESI,pos.ion)m/z:468.2[M+H]+
1H NMR(400MHz,DMSO-d6):δ(ppm)13.31(s,1H),12.31(s,1H),9.23(s,1H),8.63(s,2H),8.47(s,1H),8.37(s,1H),7.88(s,1H),7.82(s,1H),6.42(s,1H),4.19(d,J=13.4Hz,1H),3.95(dd,J=13.7,6.0Hz,1H),3.76(d,J=10.6Hz,1H),3.50(d,J=10.8Hz,1H),2.86(s,1H),2.72(s,1H),2.47(d,J=3.8Hz,2H),2.08-1.88(m,2H),1.49(s,3H),1.28(s,6H),1.18-1.08(m,2H),0.87(t,J=6.8Hz,3H),0.75(t,J=7.2Hz,3H).
实施例29:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-((甲基(苯乙基)氨基)甲基)吡啶 -2(1H)-酮
步骤1:(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸
以(R)-2-氨基-2-甲基己烷-1-醇和7-溴-2,4-二氯喹唑啉为起始物料参考实施例1的步骤3至步骤5的合成方法,可得到标题化合物为棕黑色固体,总收率42%。
MS(ESI,pos.ion)m/z:469.1[M+H]+.
步骤2:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-((甲基(苯乙基)氨基)甲基)吡啶 -2(1H)-酮
以(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸和4-氯-5-((甲基(苯乙基)氨基)甲基)吡啶-2(1H)-酮为起始物料参照实施例1步骤10至步骤11的合成方法,可得标题化合物为白色固体,总收率25%。
MS(ESI,pos.ion)m/z:515.1[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm):12.82(s,1H),12.28(s,1H),9.45(s,1H),8.53(d,J=8.4Hz,1H),8.26(s,2H),7.96(s,1H),7.88(s,1H),7.41(d,J=8.4Hz,1H),7.35-7.17(m,4H),7.06(d,J=7.1Hz,2H),6.33(s,1H),4.89(s,1H),4.29(s,1H),4.05(s,1H),3.95(d,J=10.9Hz,1H),3.58(d,J=10.8Hz,1H),2.99(s,2H),2.73(s,2H),2.16(s,1H),1.81-1.68(m,1H),1.45(s,3H),1.27(d,J=7.1Hz,4H),0.85(t,J=6.8Hz,3H).
实施例30至实施例35可参照实施例29的合成方法得到,实施例30-35的表征数据见下表3


实施例36:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-((丁基(甲基)氨基)甲基)吡啶-2(1H)-酮
以5-((丁基(甲基)氨基)甲基)-4-碘吡啶-2(1H)-酮(参照实施例56步骤1至步骤3的合成方法制备得到)和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例56步骤5的合成方法,可得到标题化合物为黄色固体,总收率为54%。
MS(ESI,pos.ion)m/z:467.3[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.42(d,J=8.4Hz,1H),7.94(s,1H),7.51(s,1H),7.45(d,J=8.6Hz,1H),6.57(s,1H),4.36(s,1H),4.20(d,J=11.2Hz,2H),3.72(d,J=11.2Hz,1H),2.99–2.79(m,2H),2.62(s,3H),2.35–2.19(m,1H),1.29–1.77(m,1H),1.57(s,3H),1.47–1.27(m,8H),0.94(t,J=6.7Hz,3H),0.84(t,J=7.2Hz,3H).
实施例37:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁-3-烯-1-基(甲基)氨基)甲基)-1-(1-甲基哌啶-4-基)吡啶-2(1H)-酮
步骤1:5-((丁-3-烯-1-基(甲基)氨基)甲基)-4-碘-1-(1-甲基哌啶-4-基)吡啶-2(1H)-酮
将5-((丁-3-烯-1-基(甲基)氨基)甲基)-4-碘代吡啶-2(1H)-酮(0.55g,1.74mmol,参照实施例9步骤1-4的合成方法制备得到)、1-甲基哌啶-4-醇(0.20g,1.74mmol)和三苯基膦(0.68g,2.61mmol)溶在无水四氢呋喃(10mL)中,氮气置换三次,将反应将至0℃,再加入DIAD(0.53g,2.61mmol),加毕,移至室温搅 拌反应约13小时。后处理:加入正己烷(5mL),有固体析出,过滤,滤液减压浓缩,所得残留物经硅胶柱层析纯化(DCM/MeOH(V/V)=10/1),得黄油状化合物(0.52g,产率71.27%)。
MS(ESI,pos.ion)m/z:208.7[(M+H)/2]+.
步骤2:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁-3-烯-1-基(甲基)氨基) 甲基)-1-(1-甲基哌啶-4-基)吡啶-2(1H)-酮
以5-((丁-3-烯-1-基(甲基)氨基)甲基)-4-碘-1-(1-甲基哌啶-4-基)吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料,参照实施例1合成步骤10和合成步骤11的合成方法,得到标题化合物为白色固体,总收率31%。
MS(ESI,pos.ion)m/z:563.6[M+H]+
1H NMR(400MHz,DMSO-d6):δ(ppm)8.63(s,2H),8.36(s,1H),7.83(s,1H),6.94(d,J=3.9Hz,1H),5.68–5.56(m,1H),5.10-4.97(m,2H),4.42(s,1H),4.19(s,1H),3.77(d,J=10.9Hz,1H),3.39(d,J=11.7Hz,2H),3.19(s,2H),3.11–2.87(m,2H),2.81(s,3H),2.47(s,2H),2.29(s,3H),2.19–2.06(m,2H),2.04-1.83(m,3H),1.49(s,3H),1.28(s,4H),1.20(d,J=6.3Hz,1H),0.87(t,J=6.7Hz,3H).
实施例38:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-N-(丁-3-烯-1-基)-N-甲基-6-氧代-1,6-二氢吡啶-3-甲酰胺
步骤1:N-(丁-3-烯-1-基)-4,6-二氯吡啶-3-甲酰胺
分别称量4,6-二氯烟酸(1.00g,5.21mmol)、丁-3-烯-1-胺盐酸盐(0.84g,7.81mmol)、HATU(3.96g,10.42mmol)和DIPEA(4.04g,31.26mmol)混合于二氯甲烷(40mL)中,置于室温下搅拌约12小时;停止搅拌;减压浓缩,所得残留物经硅胶柱层析(DCM/MeOH(V/V)=15/1)纯化,得到标题化合物为棕黑色固体(1.28g,产率23%)。
MS(ESI,pos.ion)m/z:245.0[M+H]+.
步骤2:N-(丁-3-烯-1-基)-4,6-二氯-N-甲基吡啶-3-甲酰胺
称量N-(丁-3-烯-1-基)-4,6-二氯吡啶-3-甲酰胺(1.28g,5.22mmol)溶解于THF(40mL)中,置于0℃下搅拌,接着加入60%的氢化钠(0.42g,10.5mmol)搅拌约15分钟;再加入碘甲烷(3.70g,26.07mmol)到其中,加毕,转移到室温下反应约15小时;停止反应,加水(30mL)稀释,然后用乙酸乙酯(20mL×3)萃取,合并有机相,有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(PE/EA(V/V)=10/1)纯化,得到标题化合物为淡黄色油状液体(0.90g,产率66.54%)。
MS(ESI,pos.ion)m/z:259.1[M+H]+.
步骤3:N-(丁-3-烯-1-基)-4-氯-N-甲基-6-氧代-1,6-二氢吡啶-3-甲酰胺
称量N-(丁-3-烯-1-基)-4,6-二氯-N-甲基吡啶-3-甲酰胺(0.40g,1.54mmol)加入到叔丁醇(30mL)中,接着加入叔丁醇钾(0.69g,6.16mmol),置于室温下搅拌反应约15小时;原料未反应完,补加叔丁醇钾(0.23g,2.03mmol),继续在该温度下搅拌反应约12小时;停止搅拌,用4M盐酸的1,4-二氧六环溶液调节pH至2-3,搅拌约30分钟,接着加入饱和碳酸氢钠溶液调节pH至8-9,再用EA(20mL×3)萃取,合并有机相,饱和食盐水(30mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(DCM/MeOH(V/V)=15/1)纯化,得到标题化合物为淡黄色固体(0.17g,产率45.86%)。
MS(ESI,pos.ion)m/z:241.1[M+H]+.
步骤4:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-N-(丁-3-烯-1-基)-N-甲基-6- 氧代-1,6-二氢吡啶-3-甲酰胺
以N-(丁-3-烯-1-基)-4-氯-N-甲基-6-氧代-1,6-二氢吡啶-3-甲酰胺和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料,参照实施例1合成步骤10和合成步骤11的合成方法,得到标题化合物为白色固体,总收率30%。
MS(ESI,pos.ion)m/z:480.3[M+H]+
1H NMR(400MHz,DMSO-d6):δ(ppm)13.08(s,1H),9.12(s,1H),8.57(s,1H),8.35(s,2H),8.30(s,1H),7.50(s,1H),5.87(d,J=7.0Hz,1H),5.63(d,J=7.7Hz,1H),5.16(d,J=17.0Hz,1H),5.11–4.97(m,2H),3.79(d,J=10.9Hz,1H),3.61-3.47(m,3H),3.25(s,1H),2.99(s,1H),2.88(s,1H),2.36(d,J=7.4Hz,1H),2.27(d,J=7.2Hz,1H),1.97(s,2H),1.50(s,3H),1.28(s,4H),0.88(t,J=6.7Hz,3H).
实施例39:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-(哌啶-1-羰基)吡啶-2(1H)-酮
步骤1:(6-溴-4-氯吡啶-3-基)(哌啶-1-基)甲酮
分别称量6-溴-4-氯吡啶-3-羧酸(2.46g,10.42mmol)、哌啶(1.77g,20.84mmol)、HATU(7.92g,20.84mmol)和DIPEA(8.08g,62.52mmol)混合于DMF(30mL)中,置于室温下搅拌反应约24小时;加水(30mL)稀释,然后用乙酸乙酯(30mL×3)萃取,合并有机相,饱和食盐水(25mL×3)洗涤,无水硫酸钠干燥,减压浓缩,硅胶柱层析(PE/EA(V/V)=10/1)纯化,得到标题化合物为淡黄色油状液体(2.70g,85.35%)。
MS(ESI,pos.ion)m/z:303.0[M+H]+.
步骤2:4-氯-5-(哌啶-1-羰基)吡啶-2(1H)-酮
分别称量(6-溴-4-氯吡啶-3-基)(哌啶-1-基)甲酮(1.50g,4.94mmol)和叔丁醇钾(2.22g,19.76mmol)加入到叔丁醇(50mL)中,置于90℃油浴锅中加热反应过夜;停止搅拌,2M盐酸的1,4-二氧六环溶液调节pH=1-3,然后向体系中加入饱和碳酸氢钠溶液(50mL),乙酸乙酯(20mL×3)洗涤,饱和食盐水(30mL)洗涤,无水硫酸钠干燥,减压浓缩,硅胶柱层析法(DCM/MeOH(V/V)=15/1)分离提纯,得到标题化合物为棕黑色油状液体(0.31g,收率26.07%)。
MS(ESI,pos.ion)m/z:241.0[M+H]+.
步骤3:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-(哌啶-1-羰基)吡啶-2(1H)-
以4-氯-5-(哌啶-1-羰基)吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料参照实施例38步骤4的合成方法,可得到标题化合物为白色固体,总收率44%。
MS(ESI,pos.ion)m/z:480.3[M+H]+
1H NMR(400MHz,DMSO-d6):δ(ppm)13.16(s,1H),8.59(d,J=1.6Hz,1H),8.27(s,1H),7.71(d,J=1.6Hz,1H),7.63(s,1H),6.58(s,1H),3.79(d,J=11.0Hz,2H),3.51(d,J=11.0Hz,2H),3.40-3.19(m,4H),1.96(d,J=9.3Hz,2H),1.48(s,5H),1.29(d,J=15.4Hz,8H),0.86(t,J=6.7Hz,3H).
实施例40:(R)-N-((4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-6-氧代-1,6-二氢吡啶-3-基)甲基)-N-甲基丁-3-烯酰胺
步骤1:(6-氟-4-碘吡啶-3-基)甲胺
称量5-(溴甲基)-2-氟-4-碘吡啶(1.01g,3.19mmol)加入到氨的甲醇溶液(1.08g,63.77mmol,7M)中,置于室温下搅拌反应约16小时;减压浓缩,所得残留物经硅胶柱层析(DCM/MeOH(V/V)=15/1)纯化,得到标题化合物为无色油状液体(0.71g,产率88.31%)。
MS(ESI,pos.ion)m/z:253.1[M+H]+.
步骤2:N-((6-氟-4-碘吡啶-3-基)甲基)丁-3-烯酰胺
分别称量(6-氟-4-碘吡啶-3-基)甲胺(0.70g,2.78mmol)、乙烯基乙酸(0.29g,3.36mmol)和HATU(1.59g,4.17mmol)混合于二氯甲烷(50mL)中,接着加入DIPEA(1.08g,8.34mmol),置于室温下搅拌反 应约42小时;停止搅拌,减压浓缩,所得残留物经硅胶柱层析(PE/EA(V/V)=5/1)纯化,得到标题化合物为无色油状液体(0.81g,产率91.02%)。
MS(ESI,pos.ion)m/z:321.2[M+H]+.
步骤3:N-((6-氟-4-碘吡啶-3-基)甲基)-N-甲基丁-3-烯酰胺
称量N-((6-氟-4-碘吡啶-3-基)甲基)丁-3-烯酰胺(0.70g,7.06mmol)加入到四氢呋喃(50mL)中,置于0℃下搅拌;接着加入氢化钠(0.20g,5.06mmol,纯度:60%),搅拌约15分钟;接着加入碘甲烷(1.80g,12.68mmol),加入完毕转移到室温下搅拌反应约28小时;减压浓缩,所得残留物经硅胶柱层析(PE/EA(V/V)=5/1)纯化,得到标题化合物为淡黄色固体(0.55g,产率65.06%)。
MS(ESI,pos.ion)m/z:334.9[M+H]+.
步骤4:(R)-N-((4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-6-氧代-1,6-二氢吡啶-3- 基)甲基)-N-甲基丁-3-烯酰胺
以N-((6-氟-4-碘吡啶-3-基)甲基)-N-甲基丁-3-烯酰胺为起始物料参照实施例38合成步骤3的合成方法制备得到N-((4-碘-6-氧代-1,6-二氢吡啶-3-基)甲基)-N-甲基丁-3-烯酰胺,然后再以N-((4-碘-6-氧代-1,6-二氢吡啶-3-基)甲基)-N-甲基丁-3-烯酰胺和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料,参照实施例38合成步骤4的合成方法制备,得到标题化合物为白色固体,产率为28%。
MS(ESI,pos.ion)m/z:480.2[M+H]+
1H NMR(400MHz,DMSO-d6):δ(ppm)13.00(s,1H),8.52(d,J=17.9Hz,2H),8.29(s,2H),7.78-7.66(m,1H),7.51-7.36(m,1H),6.32(s,1H),6.30–5.94(m,2H),4.32(d,J=13.8Hz,3H),3.76(d,J=10.9Hz,1H),3.50(d,J=10.9Hz,1H),2.70(s,2H),2.61(s,1H),2.08-1.88(m,2H),1.70(d,J=6.1Hz,2H),1.60(d,J=5.0Hz,1H),1.49(s,3H),1.35–1.18(m,4H),0.87(t,J=6.8Hz,3H).
实施例41:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁-3-烯-1-基氧基)甲基)吡啶-2(1H)-酮
步骤1:4-碘-2-甲氧基-5-甲基吡啶
称量2-氟-4-碘-5-甲基吡啶(2.00g,8.44mmol)溶解于甲醇(20mL)中,接着加入7M甲醇钠的甲醇溶液(1.82g,33.76mmol),置于60℃油浴锅中加热反应约12小时;加水(30mL)稀释,然后用乙酸乙酯(20mL×3)萃取,合并有机相,有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,减压浓缩,得到标题化合物为淡黄色油状液体(2.10g,产率99.91%)。
MS(ESI,pos.ion)m/z:250.1[M+H]+.
步骤2:5-(溴甲基)-4-碘-2-甲氧基吡啶
分别称量4-碘-2-甲氧基-5-甲基吡啶(2.10g,8.43mmol)、偶氮二异丁腈(1.38g,8.43mmol)和NBS(1.50g,8.43mmol)加入到四氯化碳(40mL)中,氮气保护,置于78℃油浴锅中加热反应约2小时;停止搅拌,过滤,向滤液中加入饱和硫代硫酸钠溶液(20mL),二氯甲烷(30mL×3)萃取,合并有机相,有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(PE/EA(V/V)=20/1)纯化,得到标题化合物为类白色固体(2.20g,产率79.58%)。
MS(ESI,pos.ion)m/z:327.8[M+H]+.
步骤3:5-((丁-3-烯-1-丙氧基)甲基)-4-碘-2-甲氧基吡啶
向丁-3-烯-1-醇(9.68g,134.20mmol)中加入氢氧化钾(1.88g,33.55mmol),置于90℃油浴锅中加热搅拌约1小时;降低温度至室温,接着加入5-(溴甲基)-4-碘-2-甲氧基吡啶(2.20g,6.71mmol)的四氢呋喃(20mL)溶液,继续在室温下搅拌反应约17小时;加水(50mL)稀释,然后用乙酸乙酯(40mL×3)萃取,合并有机相,有机相用饱和食盐水(50mL×3)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(PE/EA(V/V)=20/1)纯化,得到标题化合物为淡黄色油状液体(1.80g,产率84.06%)。
MS(ESI,pos.ion)m/z:319.9[M+H]+.
步骤4:5-((丁-3-烯-1-丙氧基)甲基)-4-碘代-1,2-二氢吡啶-2-酮
分别称量5-((丁-3-烯-1-丙氧基)甲基)-4-碘-2-甲氧基吡啶(1.80g,5.64mmol)和吡啶氢溴酸盐(18.05g,112.8mmol)混合于DMF(30mL)中,置于80℃油浴锅中加热反应约18小时;停止搅拌,向反应体系中加入饱和碳酸氢钠溶液(30mL),然后用乙酸乙酯(20mL×3)萃取,合并有机相,饱和食盐水(30mL×3)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(DCM/MeOH(V/V)=15/1)纯化,得到标题为淡黄色固体(0.40g,产率23.24%)。
MS(ESI,pos.ion)m/z:306.0[M+H]+.
步骤5:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁-3-烯-1-基氧基)甲基) 吡啶-2(1H)-酮
以5-((丁-3-烯-1-丙氧基)甲基)-4-碘代-1,2-二氢吡啶-2-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料参照实施例38步骤4的合成可得到标题化合物为白色固体,收率为46%。
MS(ESI,pos.ion)m/z:453.2[M+H]+
1H NMR(400MHz,DMSO-d6):δ(ppm)13.34(s,1H),11.93(s,1H),8.62(s,1H),8.27(s,1H),7.87(s,1H),7.58(s,1H),6.38(s,1H),5.64–5.50(m,1H),5.04-4.84(m,2H),4.10(s,2H),3.78(d,J=10.8Hz,1H),3.51(d,J=10.8Hz,1H),3.26(t,J=6.6Hz,2H),2.51(s,2H),2.13(q,J=6.6Hz,2H),1.97(d,J=11.9Hz,2H),1.48(s,3H),1.27(d,J=3.5Hz,4H),0.86(t,J=6.7Hz,3H).
实施例42:(R)-4-(2-氨基-4-((1-羟基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁-3-烯-1-基(甲基)氨基)甲基)吡啶-2(1H)-酮
步骤1:(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸
以7-溴-2,4-二氯吡啶[3,2-d]嘧啶和(R)-2-氨基己酸为起始物料参照实施例1步骤1至步骤5的合成方法,可得到标题化合物为棕黑色固体,总产率为99%。
MS(ESI,pos.ion)m/z:456.5[M+H]+.
步骤2:(R)-4-(2-氨基-4-((1-羟基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁-3-烯-1-基(甲基)氨基)甲基) 吡啶-2(1H)-酮
以5-((丁-3-烯-1-基(甲基)氨基)甲基)-4-碘代吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料参照实施例38步骤4的合成方法,可得到标题化合物为白色固体,总收率为77%。
MS(ESI,pos.ion)m/z:452.2[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.67(d,J=1.2Hz,1H),7.97(s,2H),6.62(s,1H),5.75-5.62(m,1H),5.18-5.08(m,2H),4.64-4.55(m,1H),4.27(s,1H),3.77(d,J=5.3Hz,2H),3.33(s,2H),3.03(s,2H),2.63(s,3H),2.35(q,J=7.4Hz,2H),1.85-1.67(m,2H),1.42(s,4H),0.93(t,J=6.8Hz,3H).
实施例43:(R)-4-(2-氨基-4-((1-羟基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁基(甲基)氨基)甲基)吡啶-2(1H)-酮
步骤1:(R)-5-((丁-3-烯-1-基(甲基)氨基)甲基)-4-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基己烷-2-基)氨基)吡啶 并[3,2-d]嘧啶-7-基)吡啶-2(1H)-酮
以5-((丁-3-烯-1-基(甲基)氨基)甲基)-4-碘代吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为原料,参照实施例1步骤10的合成方法制备,可得到标题化合物为棕黑色固体,产率为11%。
MS(ESI,pos.ion)m/z:602.3[M+H]+.
步骤2:(R)-4-(2-氨基-4-((1-羟基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁基(甲基)氨基)甲基)吡啶 -2(1H)-酮
以(R)-5-((丁-3-烯-1-基(甲基)氨基)甲基)-4-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)吡啶-2(1H)-酮为起始物料参照实施例28步骤2至步骤3的合成可得到标题化合物为白色固体,产率为26%。
MS(ESI,pos.ion)m/z:454.3[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.68(s,1H),7.99(d,J=2.5Hz,2H),6.62(s,1H),4.71-4.53(m,1H),4.36(s,1H),4.16(s,1H),3.77(d,J=5.3Hz,2H),3.33(s,3H),2.93(s,2H),2.63(s,3H),1.88-1.69(m,2H),1.43(s,6H),1.29–1.20(m,2H),0.94(t,J=6.4Hz,3H),0.87(t,J=7.3Hz,3H).
实施例44:(R)-N-(2-((2-氨基-7-(5-((丁-3-烯-1-基(甲基)氨基)甲基)-2-氧代-1,2-二氢吡啶-4-基)吡啶并[3,2-d]嘧啶-4-基)氨基)-2-甲基己基)-1-甲基-1H-吡唑-4-甲酰胺
步骤1:(R)-2-((叔丁氧羰基)氨基)-2-甲基己酸
称量(2R)-2-氨基-2-甲基己酸盐酸盐(20.00g,110.10mmol)加入到水(400mL)中,接着加入氢氧化钠(14.53g,363.33mmol),置于室温下搅拌至澄清状态;再缓慢滴加Boc酸酐(28.83g,132.11mmol)的四氢 呋喃(100mL)溶液;并继续室温搅拌反应过夜;停止搅拌,用饱和的柠檬酸溶液调节反应液pH至4-5,乙酸乙酯(100mL×3)萃取,合并有机相,饱和食盐水(80mL)洗涤,无水硫酸钠干燥,减压浓缩,得到标题化合物为淡黄色膏状固体(22.50g,产率83.30%)。
MS(ESI,pos.ion)m/z:146.4[M+H-Boc]+.
步骤2:(R)-(1-氨基-2-甲基-1-氧代己烷-2-基)氨基甲酸叔丁基酯
于两口瓶中加入(R)-2-((叔丁氧羰基)氨基)-2-甲基己酸(19.40g,79.08mmol)和三乙胺(10.40g,102.80mmol)的四氢呋喃(130mL)溶液,置于-10℃并搅拌下,缓慢滴加氯甲酸乙酯(10.30g,94.90mmol),滴加完毕继续在该温度下搅拌反应约2小时;将温度降低至-20℃下,将上述反应液置于氨气氛围下搅拌约2小时;转移到室温下继续搅拌反应约5小时;停止搅拌,然后用乙酸乙酯(20mL×3)萃取,合并有机相,有机相用饱和食盐水(30mL)洗涤,无水硫酸钠干燥,减压浓缩,得到无色油状液体为标题化合物(19.32g,产率99.99%)。
MS(ESI,pos.ion)m/z:145.1[M+H-Boc]+.
步骤3:(R)-(1-氨基-2-甲基己烷-2-基)氨基甲酸叔丁基酯
0℃下,向氢化锂铝的四氢呋喃溶液(9.00g,237.21mmol,1M)中缓慢滴加(R)-(1-氨基-2-甲基-1-氧代己烷-2-基)氨基甲酸叔丁基酯(19.32g,79.07mmol)的四氢呋喃(100mL)溶液;约1小时后滴加完毕,室温搅拌约3小时;转移到60℃油浴锅中加热搅拌约5小时;停止搅拌,向反应体系中加入15%氢氧化钠溶液(20mL),然后用乙酸乙酯(10mL×3)萃取,合并有机相,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,减压浓缩,得到标题产物为无色油状液体(12.65g,产率69.45%)。
MS(ESI,pos.ion)m/z:231.2[M+H]+.
步骤4:(R)-(2-甲基-1-(1-甲基-1H-吡唑-4-甲酰胺基)己烷-2-基)氨基甲酸叔丁基酯
分别称量(R)-(1-氨基-2-甲基己烷-2-基)氨基甲酸叔丁基酯(11.00g,47.75mmol)、1-甲基-1H-吡唑-4-羧酸(6.02g,47.75mmol)、EDCI(18.31g,95.50mmol)和DIPEA(18.51g,143.25mmol)混合于二氯甲烷(150mL)中,置于39℃下搅拌反应;停止搅拌,加水(100mL),分液,水相用DCM(80mL×3)萃取,合并有机相,有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(DCM/MeOH(V/V)=15/1)纯化,得到标题化合物为无色油状液体(12.12g,产率75.00%)。
MS(ESI,pos.ion)m/z:239.2[M-Boc+H]+.
步骤5:(R)-N-(2-氨基-2-甲基己基)-1-甲基-1H-吡唑-4-甲酰胺
称量(R)-(2-甲基-1-(1-甲基-1H-吡唑-4-甲酰胺基)己烷-2-基)氨基甲酸叔丁基酯(3.17g,9.37mmol)加入到盐酸的1,4-二氧六环溶液(4M,30mL)中,置于室温下搅拌约1小时;减压浓缩,除去溶剂,二氯甲烷(30mL)溶解,再减压浓缩,重复两次,得到标题化合物为白色固体(2.23g,产率99.86%)。
b骤6:(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((2-甲基-1-(1-甲基-1H-吡唑-4-甲酰胺基)己烷-2-基)氨基)吡啶并 [3,2-d]嘧啶-7-基)硼酸
以(R)-N-(2-氨基-2-甲基己基)-1-甲基-1H-吡唑-4-甲酰胺和7-溴-2,4-二氯吡啶并[3,2-d]嘧啶为起始物料参照实施例1步骤3至步骤5的合成方法,可得到标题化合物为棕黑色固体,收率为27%。
MS(ESI,pos.ion)m/z:577.5[M+H]+.
步骤7:(R)-N-(2-((2-氨基-7-(5-((丁-3-烯-1-基(甲基)氨基)甲基)-2-氧代-1,2-二氢吡啶-4-基)吡啶并[3,2-d]嘧啶b4-基)氨基)-2-甲基己基)-1-甲基-1H-吡唑-4-甲酰胺
以(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((2-甲基-1-(1-甲基-1H-吡唑-4-甲酰胺基)己烷-2-基)氨基)吡啶基[3,2-d]嘧啶-7-基)硼酸和5-((丁-3-烯-1-基(甲基)氨基)甲基)-4-碘吡啶-2(1H)-酮(参照实施例9步骤1至步骤4即可得到)为起始物料参照实施例1步骤10至步骤11的合成方法制备,可得标题化合物为白色固体,产率为46%。
MS(ESI,pos.ion)m/z:573.3[M+H]+
1H NMR(400MHz,DMSO-d6):δ(ppm)13.19(s,1H),12.31(s,1H),9.32(s,1H),8.94(s,1H),8.60(s,1H),8.36(s,1H),8.15(s,1H),7.87(s,1H),7.84(s,1H),7.79(s,1H),6.40(s,1H),5.71–5.52(m,1H),5.08-4.97(m,2H),4.20(s,1H),4.03-3.88(m,2H),3.84(s,3H),2.99(s,1H),2.84(s,1H),2.47(s,3H),2.22(s,2H),2.08(s,2H),1.54(s,3H),1.39-1.21(m,4H),0.87(t,J=6.3Hz,3H).
实施例45至实施例46可参照实施例44的合成方法得到,实施例45-46的结构和表征数据见下表4

实施例47:(R)-2-((2-氨基-7-(5-((丁-3-烯-1-基(甲基)氨基)甲基)-2-氧代-1,2-二氢吡啶-4-基)吡啶并[3,2-d]嘧啶-4-基)氨基)-2-甲基己基1-甲基-1H-吡唑-4-羧酸酯
步骤1:(R)-2-((叔丁氧羰基)氨基)-2-甲基己基1-甲基-1H-吡唑-4-羧酸酯
分别称取N-((2R)-1-羟基-2-甲基己烷-2-基)氨基甲酸叔丁基酯(1.00g,4.32mmol)和1-甲基-1H-吡唑-4-羧酸(0.82g,6.48mmol)溶解在1,2-二氯乙烷(10mL)中,然后依次加入EDCI(1.24g,6.48mmol)和DMAP(0.053g,0.43mmol),升温至60℃下搅拌反应约18小时。停反应,降至室温,加入水(100mL)然后用乙酸乙酯(100mL×3)萃取,收集有机相,有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,旋干除去溶剂,所得残留物经硅胶柱层析(PE/EA(V/V)=1/1)纯化,得到标题化合物为无色油状物产物(0.64g,产率43.65%)。
MS(ESI,pos.ion)m/z:240.4[M+H-Boc]+.
步骤2:(R)-2-((2-氨基-7-(5-((丁-3-烯-1-基(甲基)氨基)甲基)-2-氧代-1,2-二氢吡啶-4-基)吡啶并[3,2-d]嘧啶-4- 基)氨基)-2-甲基己基1-甲基-1H-吡唑-4-羧酸酯
以(R)-2-((叔丁氧羰基)氨基)-2-甲基己基1-甲基-1H-吡唑-4-羧酸酯为原料参考实施例44步骤5的合成方法制备,得到(R)-2-氨基-2-甲基己基1-甲基-1H-吡唑-4-甲酸酯,再以(R)-2-氨基-2-甲基己基1-甲基-1H-吡唑-4-甲酸酯和7-溴-2,4-二氯吡啶并[3,2-d]嘧啶为起始物料参照实施例44步骤6至步骤7的合成方法制 备,可得到标题化合物为白色固体,产率为8.7%。
MS(ESI,pos.ion)m/z:574.5[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.66(s,1H),8.17(s,1H),7.97(s,2H),7.85(s,1H),6.61(s,1H),5.75-5.59(m,1H),5.17-5.06(m,2H),4.61(d,J=11.3Hz,1H),4.26(s,2H),3.92(s,3H),3.03(s,2H),2.62(s,3H),2.34(d,J=7.5Hz,3H),1.99(t,J=10.3Hz,1H),1.70(s,3H),1.53-1.36(m,5H),0.96(t,J=6.7Hz,3H).
实施例48:实施例48可参考实施例47的合成方法得到,实施例48的结构和表征数据见下表5
实施例49:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((甲氨基)甲基)-吡啶-2(1H)-酮
以2-氟-4-碘-5-甲基吡啶、Boc甲胺为起始物料参照实施例56步骤1至步骤3方法可得到4-碘-5-((甲胺基)甲基)吡啶-2(1H)-酮,然后再以4-碘-5-((甲胺基)甲基)吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料参照实施例56步骤5的合成方法,可得到标题化合物为黄色固体,总产率为16%。
MS(ESI,pos.ion)m/z:412.2[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.64(d,J=1.4Hz,1H),7.94(d,J=1.4Hz,1H),7.88(s,1H),6.60(s,1H),4.09(s,2H),4.01(d,J=11.1Hz,1H),3.75(d,J=11.2Hz,1H),2.59(s,3H),2.19-2.08(m,1H),2.06-1.93(m,1H),1.59(s,3H),1.38(s,4H),0.95(t,J=6.5Hz,3H).
实施例50:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((二甲氨基)甲基)吡啶-2(1H)-酮
以2-氟-4-碘-5-甲基吡啶、二甲胺为起始物料参照实施例56步骤1至步骤3合成方法可得到5-((二甲氨基)甲基)-4-碘吡啶-2(1H)-酮,然后以5-((二甲氨基)甲基)-4-碘吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料参照实施例56步骤5的合成方法制备,可得到标题化合物为黄色固体,总产率为33%。
MS(ESI,pos.ion)m/z:426.3[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.64(d,J=1.6Hz,1H),7.99-7.92(m,2H),6.61(s,1H),4.23(s,2H),4.00(d,J=11.2Hz,1H),3.75(d,J=11.2Hz,1H),2.68(s,6H),2.18-2.08(m,1H),2.03-1.96(m,1H),1.59(s,3H),1.44-1.35(m,4H),0.95(t,J=6.7Hz,3H).
实施例51:(R)-4-(2-氨基-4-((1-羟基-2-甲基己基-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-(甲氧基甲基)吡啶-2(1H)-酮
步骤1:4-碘-2-甲氧基-5-(甲氧基甲基)吡啶
将5-(溴甲基)-2-氟-4-碘代吡啶(1.10g,3.48mmol)溶在甲醇钠的甲醇溶液(6.57mL,34.8mmol)中,室温搅拌反应约3小时。停止反应,浓缩溶剂,加入水(20mL)和乙酸乙酯(20mL),搅拌分液,水相用乙酸乙酯(20mL×2)萃取,合并有机相,有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩溶剂,得标题化合物为黄色固体(0.88g,产率90.30%)。
步骤2:4-碘-5-(甲氧基甲基)吡啶-2(1H)-酮
将碘化钠(2.35g,15.70mmol)悬浮在ACN(20mL)中,置于0℃条件下,加入三甲基氯硅烷(1.71g,15.70mmol),加毕,移至室温搅拌15min,将4-碘-2-甲氧基-5-(甲氧基甲基)吡啶(0.877g,3.14mmol)加入到反应体系中,加毕,继续搅拌反应约17小时。停止反应,浓缩溶剂,加入水(10mL)和二氯甲烷(10mL)搅拌分液,水相用二氯甲烷(10mL×3)萃取,合并有机相,有机相用饱和食盐水(10mL×3)洗涤,无水硫酸钠干燥,浓缩溶剂,所得残留物经硅胶柱层析(DCM/MeOH(V/V)=15/1)纯化,得到标题化合物为淡黄色固体(0.38g,产率45.78%)。
步骤3:(R)-4-(2-氨基-4-((1-羟基-2-甲基己基-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-(甲氧基甲基)吡啶-2(1H)-
以4-碘-5-(甲氧基甲基)吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料参照实施例1步骤10至步骤11的合成方法,可得到标题化合物为白色固体,总产率为46%。
MS(ESI,pos.ion)m/z:413.2[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.67(s,1H),7.95(s,1H),7.64(s,1H),6.57(s,1H),4.12(s,2H),4.00(d,J=11.2Hz,1H),3.74(d,J=11.2Hz,1H),3.25(s,3H),2.19-2.07(m,1H),2.02-1.88(m,1H),1.57(s,3H),1.37(s,4H),0.93(t,J=6.3Hz,3H).
实施例52:(R)-4-(2-氨基-4-((1-羟基-2-甲基己基-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((甲硫基)甲基)吡啶-2(1H)-酮
步骤1:4-碘-2-甲氧基-5-甲基吡啶
将2-氟-4-碘-5-甲基吡啶(4.0g,16.88mmol)溶在甲醇钠的甲醇溶液(9.54mL,50.47mmol)中,室温搅拌反应约22小时。停止反应,浓缩溶剂,加入水(20mL)和乙酸乙酯(20mL),搅拌分液,水相用乙酸乙酯(20mL×2)萃取,合并有机相,有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,浓缩溶剂,得标题化合物为黄色固体(3.22g,产率76.55%)。
MS(ESI,pos.ion)m/z:250.0[M+H]+.
步骤2:5-(溴甲基)-4-碘-2-甲氧基吡啶
将4-碘-2-甲氧基-5-甲基吡啶(3.20g,12.85mmol)、NBS(2.29g,12.85mmol)和AIBN(2.11g,12.85mmol)溶在四氯化碳(30mL)中,氮气置换三次,置于70℃条件下搅拌反应约4小时。停止搅拌,关闭加热,冷却至室温,过滤,滤液加入饱和硫代硫酸钠溶液(20mL),水相用二氯甲烷(20mL×3)萃取,合并有机层并用饱和食盐水溶液(20mL)洗涤,有机层合并用无水硫酸钠干燥,过滤并减压浓缩,残留物经硅胶柱层析纯化(PE/EA(V/V)=20/1),得标题化合物为白色固体(3.59g,产率85.19%)。
步骤3:4-碘-2-甲氧基-5-((甲硫基)甲基)吡啶
将5-(溴甲基)-4-碘-2-甲氧基吡啶(3.20g,9.76mmol)和甲硫醇钠(0.75g,10.74mmol)加入到DMF(16L)中,室温下搅拌反应约3小时。停止反应,加入乙酸乙酯(20mL)和水(20mL)搅拌分液,水相用乙酸乙酯(20mL×2)萃取,合并有机相,合并的有机相用饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,过滤,浓缩溶剂,所得残留物经硅胶柱层析纯化(PE/EA(V/V)=10/1),得标题化合物为白色固体(0.54g,产率18.75%)。
MS(ESI,pos.ion)m/z:296.00[M+H]+.
步骤4:4-碘-5-((甲硫基)甲基)吡啶-2(1H)-酮的合成
将碘化钠(1.37g,9.15mmol)悬浮在乙腈(10mL)中,然后于0℃条件下,加入三甲基氯硅烷(0.99g,9.15mmol),加毕,移至室温搅拌15分钟,将4-碘-2-甲氧基-5-((甲硫基)甲基)吡啶(0.54g,1.83mmol)加入到反应体系中,加毕,继续搅拌反应约23小时。停止反应,浓缩除去溶剂,加入水(10mL)和二氯甲烷(10mL)搅拌分液,水相用二氯甲烷(10mL×2)萃取,合并有机相,饱和食盐水(10mL×3)洗涤,无水硫酸钠干燥,过滤,浓缩溶剂,所得残留物经硅胶柱层析纯化(DCM/MeOH(V/V)=10/1),得标题化合物为白色固体(0.11g,产率22.16%)。
MS(ESI,pos.ion)m/z:282.1[M+H]+.
步骤5:(R)-4-(2-氨基-4-((1-羟基-2-甲基己基-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((甲硫基)甲基)吡啶 -2(1H)-酮
以4-碘-5-((甲硫基)甲基)吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料参照实施例1步骤10至步骤11的合成方法,可得到标题化 合物为白色固体,总产率为13%。
MS(ESI,pos.ion)m/z:429.4[M+H]+
1H NMR(400MHz,CD3OD)δ(ppm)8.62(d,J=1.4Hz,1H),7.89(d,J=1.3Hz,1H),7.50(s,1H),6.50(s,1H),3.96(d,J=11.3Hz,1H),3.71(d,J=11.3Hz,1H),3.44(s,2H),3.27(d,J=1.4Hz,2H),2.09(d,J=14.4Hz,1H),1.90(s,3H),1.53(s,3H),1.33(s,4H),0.90(t,J=6.5Hz,3H).
实施例53:(R)-4-(2-氨基-4-((1-甲氧基-2-甲基己基-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁基(甲基)氨基)甲基)吡啶-2(1H)-酮
步骤1:(R)-(1-甲氧基-2-甲基己-2-基)氨基甲酸叔丁基酯
将氢化钠(0.10g,4.30mmol)溶在DMF(5mL)中,氮气保护,置于0℃条件搅拌反应,慢慢滴加溶在DMF(5mL)中的(R)-(1-羟基-2-甲基己烷-2-基)氨基甲酸叔丁酯(0.50g,2.14mmol),在该条件下继续搅拌约30分钟,加入碘甲烷(0.36g,2.57mmol),移至室温搅拌反应约1小时。停止反应,加入乙酸乙酯(20mL)和水(20mL)搅拌分液,水相用乙酸乙酯(20mL×2)萃取,合并有机相,饱和食盐水(20mL×3)洗涤,无水硫酸钠干燥,过滤,浓缩溶剂,得标题化合物为黄色油状(0.46g,产率88.18%)。
MS(ESI,pos.ion)m/z:146.3[M+H-Boc]+.
步骤2:(R)-4-(2-氨基-4-((1-甲氧基-2-甲基己基-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁基(甲基)氨基)甲基) 吡啶-2(1H)-酮
以7-溴-2,4-二氯吡啶并[3,2-d]嘧啶和(R)-(1-甲氧基-2-甲基己-2-基)氨基甲酸叔丁基酯为起始物料参照实施例1步骤2至步骤5方法可得到化合物(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-甲氧基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸,然后再以(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-甲氧基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸和5-((丁基(甲基)氨基)甲基)-4-碘吡啶-2(1H)-酮为起始物料参照实施例1步骤10至步骤11的合成方法,可得到标题化合物为白色固体,总产率为10.3%。
MS(ESI,pos.ion)m/z:482.5[M+H]+
1H NMR(400MHz,CD3OD)δ(ppm)8.64(d,J=1.5Hz,1H),7.95(s,2H),6.61(s,1H),3.82(d,J=9.3Hz,1H),3.58(d,J=9.4Hz,1H),3.43(s,3H),3.32(s,2H),3.31(s,2H),2.62(s,3H),2.08(t,J=7.4Hz,2H),1.60(s,3H),1.48-1.32(m,6H),1.28-1.18(m,2H),0.93(t,J=6.6Hz,3H),0.86(t,J=7.3Hz,3H).
实施例54:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-(((2-甲氧基乙基)(甲基)氨基)甲基)吡啶-2(1H)-酮
步骤1:5-(溴甲基)-2-氟-4-碘吡啶
分别称量2-氟-4-碘-5-甲基吡啶(20.00g,84.38mmol)、N-溴代琥珀酰亚胺(16.52g,92.82mmol)和2,2'-偶氮二(2-甲基丙腈)(1.39g,8.44mmol)混合于四氯化碳(200mL)中,氮气保护,置于80℃油浴锅中回流反应约14小时;停止搅拌,向反应体系中加入饱和硫代硫酸钠溶液(30mL),然后用二氯甲烷(100mL×3)萃取,合并有机相,饱和食盐水(50mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(PE/EA(V/V)=20/1)纯化,得到标题化合物为白色固体(19.56g,产率73.38%)。
1HNMR(400MHz,CDCl3)δ(ppm)8.25(s,1H),7.51(d,J=3.1Hz,1H),4.58(s,2H).
步骤2:N-((6-氟-4-碘吡啶-3-基)甲基)-2-甲氧基-N-甲基乙胺
分别称量5-(溴甲基)-2-氟-4-碘吡啶(1.00g,3.17mmol)、N-(2-甲氧基乙基)甲基胺(0.93g,10.46mmol)和DIPEA(4.10g,31.69mmol)混合于二氯甲烷(50mL)中,置于室温下搅拌反应约2小时;减压浓缩,所得残留物经硅胶硅胶柱层析(PE/EA(V/V)=3/1)纯化,得到标题化合物为无色油状液体(0.76g,产率73.97%)。
MS(ESI,pos.ion)m/z:325.2[M+H]+.
步骤3:4-碘-5-(((2-甲氧基乙基)(甲基)氨基)甲基)吡啶-2(1H)-酮
分别称量N-((6-氟-4-碘吡啶-3-基)甲基)-2-甲氧基-N-甲基乙胺(0.65g,2.01mmol)和叔丁醇钾(0.90g,8.02mmol)混合于叔丁醇(30mL)中,置于90℃油浴锅中加热反应约18小时;向体系中加入4M盐酸的1,4-二氧六环溶液调节pH至2-3,继续搅拌约15分钟;接着向体系里面加入饱和碳酸氢钠溶液调节pH至7-8,二氯甲烷(30mL×3)萃取,合并有机相,饱和食盐水(30mL)洗涤,无水硫酸钠干燥,减压浓缩,得到标题化合物为淡黄色固体(0.63g,产率97.30%)。
MS(ESI,pos.ion)m/z:323.0[M+H]+.
步骤4:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-(((2-甲氧基乙基)(甲基)氨基)甲基) 吡啶-2(1H)-酮
以4-碘-5-(((2-甲氧基乙基)(甲基)氨基)甲基)吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例1步骤10至步骤11的合成方法,可得到标题化合物为白色固体,产率为39%。
MS(ESI,pos.ion)m/z:469.3[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.41(d,J=8.4Hz,1H),7.92(s,1H),7.50(s,1H),7.44(d,J=8.4Hz,1H),6.55(s,1H),4.30(s,1H),4.20(d,J=11.2Hz,1H),3.73(d,J=11.2Hz,1H),3.55(t,J=4.9Hz,2H),3.39-3.31(m,3H),3.26(s,3H),3.17(s,2H),2.63(s,3H),2.34-2.23(m,1H),1.91-1.78(m,1H),1.57(s,3H),1.44-1.27(m,4H),0.93(t,J=6.9Hz,3H).
实施例55:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-((2-甲氧基乙基)(甲基)氨基)甲基)-1-甲基吡啶-2(1H)-酮
以以4,6-二氯烟醛、2-甲氧基乙烷-1-胺为起始物料参照实施例1步骤6至步骤9的合成方法,可得到4-碘-5-(((2-甲氧基乙基)(甲基)氨基)甲基)-1-甲基吡啶-2(1H)-酮,然后再以4-碘-5-(((2-甲氧基乙基)(甲基)氨 基)甲基)-1-甲基吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例1步骤10至步骤11的合成方法,可得到标题化合物为白色固体,产率为24%。
MS(ESI,pos.ion)m/z:483.3[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.41(d,J=8.4Hz,1H),8.12(s,1H),7.50(s,1H),7.43(d,J=8.4Hz,1H),6.57(s,1H),4.30(s,1H),4.20(d,J=11.3Hz,1H),3.73(d,J=11.2Hz,1H),3.69(s,3H),3.55(t,J=4.9Hz,2H),3.33(s,2H),3.26(s,3H),3.18(s,2H),2.64(s,3H),2.35-2.20(m,1H),1.91-1.79(m,1H),1.56(s,3H),1.44-1.25(m,4H),0.93(t,J=6.8Hz,3H).
实施例56:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-((丁基(甲基)氨基)甲基)-1-甲基吡啶-2(1H)-酮
步骤1:5-(溴甲基)-2-氟-4-碘吡啶
分别称量2-氟-4-碘-5-甲基吡啶(20.00g,84.38mmol)、N-溴代琥珀酰亚胺(16.52g,92.82mmol)、2,2'-偶氮二(2-甲基丙腈)(1.39g,8.44mmol)混合于四氯化碳(200mL)中,氮气保护,置于80℃油浴锅中回流反应约14小时;停止搅拌,降至室温,向反应体系中加入饱和硫代硫酸钠溶液(30mL),然后用二氯甲烷(100mL×3)萃取,合并有机相,饱和食盐水(50mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(PE/EA(V/V)=20/1)纯化,得到标题化合物为白色固体(19.56g,产率73.38%)。
1HNMR(400MHz,CDCl3):δ(ppm)8.25(s,1H),7.51(d,J=3.1Hz,1H),4.58(s,2H).
步骤2:N-((6-氟-4-碘吡啶-3-基)甲基)-N-甲基丁-1-胺
分分别称量5-(溴甲基)-2-氟-4-碘吡啶(6.90g,21.84mmol)、N-甲基-1-丁胺(5.71g,65.52mmol)和DIPEA(14.11g,109.18mmol)混合于二氯甲烷(100mL)中,置于室温下搅拌反应约30分钟;减压浓缩,所得残留物经硅胶柱层析(PE/EA(V/V)=20/1)纯化,得到标题化合物为淡黄色油状液体(6.52g,产率92.67%)。
MS(ESI,pos.ion)m/z:323.1[M+H]+.
步骤3:5-((丁基(甲基)氨基)甲基)-4-碘吡啶-2(1H)-酮
称量N-((6-氟-4-碘吡啶-3-基)甲基)-N-甲基丁-1-胺(6.50g,20.18mmol)溶解于叔丁醇(50mL)中,接着加入叔丁醇钾(9.05g,80.69mmol),置于室温下搅拌约47小时。停止反应,用6M盐酸的1,4-二氧六环溶液调节pH至1-3,在室温下搅拌约30分钟;然后向里面加入饱和碳酸氢钠溶液调节pH至7-9,乙酸乙酯(30mL×3)萃取,合并有机相,有机相用饱和食盐水(35mL)洗涤,无水硫酸钠干燥,减压浓缩,得到标题化合物为类白色固体(5.14g,产率79.55%)。
MS(ESI,pos.ion)m/z:321.1[M+H]+.
步骤4:5-((丁基(甲基)氨基)甲基)-4-碘-1-甲基吡啶-2(1H)-酮
分别称量5-((丁基(甲基)氨基)甲基)-4-碘吡啶-2(1H)-酮(0.39g,1.22mmol)、碳酸铯(0.60g,1.83mmol)和碘甲烷(0.21g,1.46mmol)混合于DMF(30mL)中,置于室温下搅拌反应约4小时;加水(30mL)稀释,然后用乙酸乙酯(30mL×3)萃取,合并有机相,饱和食盐水(30mL×3)洗涤,无水硫酸钠干燥,减压浓缩,得到标题化合物为淡黄色油状液体(0.23g,产率56.41%)。
MS(ESI,pos.ion)m/z:335.1[M+H]+.
步骤5:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-((丁基(甲基)氨基)甲基)-1-甲基吡啶 -2(1H)-酮
以5-((丁基(甲基)氨基)甲基)-4-碘-1-甲基吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例1步骤10至步骤11的合成方法,可得到标题化合物为白色固体,产率为16%。
MS(ESI,pos.ion)m/z:481.4[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.42(d,J=8.4Hz,1H),8.15(s,1H),7.51(d,J=0.9Hz,1H),7.45(d,J=8.4Hz,1H),6.57(s,1H),4.36(s,1H),4.19(d,J=11.2Hz,1H),3.72(d,J=11.3Hz,1H),3.68(s,3H),3.33(t,J=1.6Hz,2H),2.99-2.78(m,2H),2.64(s,3H),2.34-2.23(m,1H),1.91-1.80(m,1H),1.57(s,3H),1.45-1.18(m,8H),0.93(t,J=6.9Hz,3H),0.84(t,J=7.3Hz,3H).
实施例57:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-(((2-甲氧基乙基)氨基)甲基)吡啶-2(1H)-酮
以5-(溴甲基)-2-氟-4-碘吡啶和2-甲氧基乙烷-1-胺为起始物料参照实施例56步骤2至步骤3的合成方法可得到4-碘-5-((2-甲氧基乙基)氨基)甲基)吡啶-2(1H)-酮,然后以4-碘-5-((2-甲氧基乙基)氨基)甲基)吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例56步骤5的合成方法,可得到标题化合物为黄色固体,产率为6.4%。
MS(ESI,pos.ion)m/z:455.6[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.39(d,J=8.4Hz,1H),7.84(s,1H),7.49-7.39(m,2H),6.53(s,1H),4.19(d,J=11.2Hz,1H),4.08(s,2H),3.71(d,J=11.2Hz,1H),3.49(t,J=5.0Hz,2H),3.24(s,3H),3.06(t,J=4.8Hz,2H),2.26(t,J=10.5Hz,1H),1.87-1.77(m,1H),1.55(s,3H),1.40-1.29(m,4H),0.92(t,J=6.9Hz,3H).
实施例58:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-((丁胺基)甲基)吡啶-2(1H)-酮
以5-(溴甲基)-2-氟-4-碘吡啶和和正丁胺为起始物料参照实施例56步骤2至步骤3的合成方法可得5-((丁胺基)甲基)-4-碘吡啶-2(1H)-酮,然后以5-((丁胺基)甲基)-4-碘吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例56步骤5的合成方法,可得到标题化合物为黄色固体,总产率为3.8%。
MS(ESI,pos.ion)m/z:453.5[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.38(d,J=8.4Hz,1H),7.85(s,1H),7.52-7.37(m,2H),6.53(s,1H),4.18(d,J=11.2Hz,1H),4.05(s,2H),3.71(d,J=11.3Hz,1H),2.82(t,J=8.0Hz,2H),2.32-2.20(m,1H),1.89-1.76(m,1H),1.55(s,3H),1.50-1.42(m,2H),1.41-1.20(m,8H),0.92(t,J=6.8Hz,3H),0.86(t,J=7.3Hz,3H).
实施例59:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-((甲氨基)甲基)吡啶-2(1H)-酮
以5-(溴甲基)-2-氟-4-碘吡啶和Boc甲胺为起始物料,参照实施例56步骤2至步骤3的合成方法可得 到((4-碘代-6-氧代-1,6-二氢吡啶-3-基)甲基)(甲基)氨基甲酸叔丁酯,然后以((4-碘代-6-氧代-1,6-二氢吡啶-3-基)甲基)(甲基)氨基甲酸叔丁酯和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例56步骤5的合成方法,可得到标题化合物为黄色固体,总产率为77%。MS(ESI,pos.ion)m/z:411.5[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.40(d,J=8.4Hz,1H),7.85(s,1H),7.47(s,1H),7.43(d,J=8.3Hz,1H),6.54(s,1H),4.20(d,J=11.3Hz,1H),4.07(s,2H),3.73(d,J=11.2Hz,1H),2.56(s,3H),2.30(d,J=12.8Hz,1H),1.84(t,J=10.7Hz,1H),1.57(s,3H),1.44-1.32(m,4H),0.94(t,J=6.8Hz,3H).
实施例60:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-(((2-羟乙基)(甲基)氨基)甲基)吡啶-2(1H)-酮
以5-(溴甲基)-2-氟-4-碘吡啶和2-(甲氨基)乙烷-1-醇为起始物料参照实施例步骤2至步骤3的方法合成可得到5-((2-羟乙基)(甲基)氨基)甲基)-4-碘吡啶-2(1H)-酮,然后以5-((2-羟乙基)(甲基)氨基)甲基)-4-碘吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例56步骤5的方法合成可得到标题化合物为白色固体,产率为19%。
MS(ESI,pos.ion)m/z:455.3[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.40(d,J=8.4Hz,1H),7.95(s,1H),7.51(s,1H),7.43(d,J=8.4Hz,1H),6.55(s,1H),4.32(s,2H),4.19(d,J=11.3Hz,1H),3.67-3.79(m,3H),3.08(s,2H),2.66(s,3H),2.34-2.22(m,1H),1.91-1.78(m,1H),1.57(s,3H),1.43-1.27(m,4H),0.94(t,J=6.8Hz,3H).
实施例61:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-(((2-甲氧基乙基)氨基)甲基)-1-甲基吡啶-2(1H)-酮
以5-(溴甲基)-2-氟-4-碘吡啶和2-甲氧基乙烷-1-胺和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例56步骤2至步骤5的合成方法,可得到标题化合物为白色固体,总产率为24%。
MS(ESI,pos.ion)m/z:469.5[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.38(d,J=8.4Hz,1H),8.05(s,1H),7.49(s,1H),7.41(d,J=8.4Hz,1H),6.53(s,1H),4.18(d,J=11.3Hz,1H),4.09(s,2H),3.71(d,J=11.3Hz,1H),3.65(s,3H),3.49(t,J=5.0Hz,2H),3.24(s,3H),3.06(t,J=11.3Hz,2H),2.32-2.20(m,1H),1.87-1.76(m,1H),1.55(s,3H),1.42-1.23(m,4H),0.91(t,J=6.9Hz,3H).
实施例62:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-((丁胺基)甲基)-1-甲基吡啶-2(1H)-酮
以5-(溴甲基)-2-氟-4-碘吡啶和正丁胺和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例56步骤2至步骤5的合成方法,可得到标题化合物为白色固体,总产率为11%。
MS(ESI,pos.ion)m/z:467.5[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.38(d,J=8.4Hz,1H),8.06(s,1H),7.49(s,1H),7.41(dd,J=8.4,1.2Hz,1H),6.53(s,1H),4.18(d,J=11.3Hz,1H),4.06(s,2H),3.71(d,J=11.3Hz,1H),3.64(s,3H),3.35(s,2H),2.87-2.78(m,2H),2.31-2.19(m,1H),1.90-1.76(m,1H),1.53(s,3H),1.50-1.42(m,2H),1.42-1.21(m,7H),0.91(t,J=6.8Hz,3H),0.85(t,J=7.3Hz,3H).
实施例63:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-1-甲基-5-((甲基氨基)甲基)吡啶-2(1H)-酮
以5-(溴甲基)-2-氟-4-碘吡啶和Boc甲胺为起始物料,参照实施例56步骤2至步骤3的合成方法可得到((4-碘代-6-氧代-1,6-二氢吡啶-3-基)甲基)(甲基)氨基甲酸叔丁酯,然后以((4-碘代-6-氧代-1,6-二氢吡啶-3-基)甲基)(甲基)氨基甲酸叔丁酯和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例56步骤4-5的合成方法,可得到标题化合物为白色固体,总产率为57%。MS(ESI,pos.ion)m/z:425.5[M+H]+.
实施例64:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-(((2-羟乙基)(甲基)氨基)甲基)-1-甲基吡啶-2(1H)-酮
以5-(溴甲基)-2-氟-4-碘吡啶和2-(甲氨基)乙烷-1-醇和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例56步骤2至步骤5的合成方法,可得到标题化 合物为白色固体,总产率为16%。
MS(ESI,pos.ion)m/z:469.3[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.40(d,J=8.4Hz,1H),8.14(s,1H),7.50(s,1H),7.43(d,J=8.4Hz,1H),6.57(s,1H),4.31(s,2H),4.19(d,J=11.3Hz,1H),3.78-3.70(m,3H),3.69(s,3H),3.09(s,2H),2.66(s,3H),2.34-2.21(m,1H),1.91-1.79(m,1H),1.57(s,3H),1.44-1.25(m,4H),0.94(t,J=6.8Hz,3H).
实施例65:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-(((2-甲氧基乙基)(甲基)氨基)甲基)吡啶-2(1H)-硫酮
步骤1:N-((6-氟-4-碘吡啶-3-基)甲基)-2-甲氧基-N-甲基乙胺
以5-(溴甲基)-2-氟-4-碘吡啶和2-甲氧基乙烷-1-胺为起始物料参照实施例56步骤2的合成方法,即可得到标题化合物为淡黄色油状液体,产率为90%。
MS(ESI,pos.ion)m/z:325.1[M+H]+.
步骤2:N-((4-碘-6-((4-甲氧基苄基)硫)吡啶-3-基)甲基)-2-甲氧基-N-甲基乙胺
称量N-((6-氟-4-碘吡啶-3-基)甲基)-2-甲氧基-N-甲基乙胺(1.00g,3.09mmol)溶解于四氢呋喃(20mL)中,置于0℃下缓慢加入氢化钠(0.15g,3.60mmol,60%),并搅拌约10分钟,然后向其中加入4-甲氧基苄胺(0.48g,3.09mmol),于该温度下继续搅拌反应;再自然升温至室温反应约2小时,加水(20mL)淬灭反应,然后用EA(15mL×3)萃取,合并有机相,饱和食盐水(30mL)洗涤,无水硫酸钠干燥,减压浓缩,所得残留物经硅胶柱层析(PE/EA(V/V)=5/1)纯化,得到标题化合物为无色油状液体(0.76g,产率53.66%)
MS(ESI,pos.ion)m/z:459.1[M+H]+.
步骤3:2-((2-((2,4-二甲氧基苄基)氨基)-7-(2-((4-甲氧基苄基)硫)-5-(((2-甲氧基乙基)(甲基)氨基)甲基)吡啶-4- 基)喹唑啉-4-基)氨基)-2-甲基己烷-1-醇
分别称量(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸(0.69g,0.72mmol)、N-((4-碘-6-((4-甲氧基苄基)硫)吡啶-3-基)甲基)-2-甲氧基-N-甲基乙胺(0.36g,0.79mmol)、碳酸钾(0.30g,2.16mmol)和[1,1'-双(二苯基膦)二茂铁]二氯化钯(0.053g,0.072mmol)混合于1,4-二氧六环/水(V/V)=5/1(30mL)中,氮气保护,置于80℃油浴锅中加热反应约29小时;降至室温,减压浓缩,所得残留物经硅胶柱层析(DCM/MeOH(V/V)=15/1)纯化,得到标题化合物为棕黑色固体(0.54g,产率99.34%)。
MS(ESI,pos.ion)m/z:755.4[M+H]+.
步骤4:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-(((2-甲氧基乙基)(甲基)氨基)甲基) 吡啶-2(1H)-硫酮
称量2-((2-((2,4-二甲氧基苄基)氨基)-7-(2-((4-甲氧基苄基)硫)-5-(((2-甲氧基乙基)(甲基)氨基)甲基)吡啶-4-基)喹唑啉-4-基)氨基)-2-甲基己烷-1-醇(0.60g,0.79mmol)加入到三氟乙酸(3mL)中,置于60℃下搅拌反应约18小时。停止搅拌,降至室温,减压浓缩,接着依次加入碳酸钾(0.44g,3.18mmol)和甲醇(5mL),反应混合物在室温下搅拌约6小时,过滤,减压浓缩,残留物中加入乙腈(2mL)、水(2mL)和三氟乙酸(0.5mL)稀释,然后过滤,滤液经制备色谱纯化(15%-50%乙腈/水洗脱30分钟,含千分之一三氟乙酸),减压浓缩除掉乙腈,接着冻干机上面冻干,得到标题化合物为黄色固体(0.13g,产率33.95%)。MS(ESI,pos.ion)m/z:485.3[M+H]+
1H NMR(400MHz,CD3OD)δ(ppm)8.42(d,J=8.4Hz,1H),8.04(s,1H),7.51(s,1H),7.46-7.40(m,2H),4.34(s,2H),4.20(d,J=11.3Hz,1H),3.72(d,J=11.2Hz,1H),3.56(t,J=4.9Hz,2H),3.27(s,3H),3.24-3.14(m,2H),2.64(s,3H),2.32-2.21(m,1H),1.92-1.77(m,1H),1.56(s,3H),1.42-1.30(m,4H),0.93(t,J=6.8Hz,3H).
实施例66:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((2-甲氧基乙基)(甲基)氨基)甲基)吡啶-2(1H)-硫酮
以N-((4-碘-6-((4-甲氧基苄基)硫)吡啶-3-基)甲基)-2-甲氧基-N-甲基乙胺和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料参照实施例65步骤3至步骤4的合成方法,可得到标题化合物为白色固体,总产率为8%。
MS(ESI,pos.ion)m/z:486.5[M+H]+
1H NMR(400MHz,CD3OD):δ(ppm)8.65(d,J=1.6Hz,1H),8.40(s,1H),8.06(s,1H),7.99(d,J=1.7Hz,1H),7.46(s,1H),4.33(s,2H),4.00(d,J=11.2Hz,1H),3.74(d,J=11.2Hz,1H),3.63-3.53(m,2H),3.38-3.31(m,3H),3.28(s,3H),3.21(s,2H),2.68(s,3H),2.13(d,J=14.5Hz,1H),2.00(dd,J=15.2,9.5Hz,1H),1.58(s,3H),1.35(d,J=25.3Hz,5H),0.95(t,J=6.7Hz,3H).
实施例67:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-((二甲基氨基)甲基)吡啶-2(1H)-
以5-((二甲氨基)甲基)-4-碘吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例56步骤5的合成方法制备,可得到标题化合物为淡黄色黄色固体,产率为54.27%。
MS(ESI,pos.ion)m/z:425.5[M+H]+
1H NMR(400MHz,CD3OD)δ(ppm)7.99(d,J=8.4Hz,1H),7.50(s,1H),7.28(d,J=1.4Hz,1H),7.14(dd,J=8.4,1.7Hz,1H),6.48(s,1H),3.94(d,J=11.4Hz,1H),3.75(d,J=11.4Hz,1H),3.23(s,2H),2.24-2.13(m,1H),2.13(s,6H),1.92-1.78(m,1H),1.45(s,3H),1.40-1.26(m,4H),0.92(t,J=7.0Hz,3H).
实施例68:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)-5-(甲氧基甲基)吡啶-2-酮
以4-碘-5-(甲氧基甲基)吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)喹唑啉-7-基)硼酸为起始物料参照实施例56步骤5的合成方法制备,可得到标题化合物为淡黄色黄色固 体,产率为83.88%。
MS(ESI,pos.ion)m/z:412.5[M+H]+
1H NMR(400MHz,CD3OD)δ(ppm)8.21(d,J=8.4Hz,1H),7.60(s,1H),7.41(s,1H),7.37(d,J=8.4Hz,1H),6.51(s,1H),4.14(s,2H),4.11(d,J=7.9Hz,1H),3.75(d,J=11.3Hz,1H),3.26(s,3H),1.53(s,3H),2.32–2.21(m,1H),1.87-1.76(m,1H),1.45-1.31(m,4H),0.93(t,J=6.9Hz,3H).
实施例69:4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-5-((丁基(甲基)氨基)甲基) 吡啶-2(1H)-酮
以5-((丁基(甲基)氨基)甲基)-4-碘吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料参照实施例56步骤5的合成方法,可得到标题化合物为黄色固体,产率为59.03%。
MS(ESI,pos.ion)m/z:468.50[M+H]+
1H NMR(400MHz,CD3OD)δ(ppm)8.38(d,J=1.6Hz,1H),7.63(d,J=1.6Hz,1H),7.49(s,1H),6.52(s,1H),3.96(d,J=11.2Hz,1H),3.77(d,J=11.3Hz,1H),3.25(s,2H),2.17(t,J=7.1Hz,2H),2.14-2.06(m,1H),2.03(s,3H),1.97-1.82(m,1H),1.50(s,3H),1.42-1.33(m,4H),1.19-1.09(m,2H),1.08-0.98(m,2H),0.93(t,J=6.9Hz,3H),0.73(t,J=7.2Hz,3H).
实施例70:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-1-甲基-5-(哌啶-1-基甲 基)吡啶-2(1H)-酮
步骤1:4-碘-6-甲氧基烟碱腈
将6-甲氧基烟腈(1.0g,7.45mmol)溶在THF(30mL)中,氮气保护,-78℃下缓慢滴加二异丙基氨基锂的四氢呋喃溶液(4.86mL,9.71mmol),滴毕,-78℃下搅拌反应1h,在该条件下,加入溶碘(3.03g,11.92mmol)的THF(5mL)溶液,滴毕,继续反应1h。加入饱和氯化铵溶液(20mL)和乙酸乙酯(30mL),水相用乙酸乙酯萃取(20mL×2),合并有机相,合并的有机相用无水硫酸钠干燥,过滤,减压浓缩,所得残留物经硅胶柱层析纯化(PE/EA(V/V)=5/1)得白色固体化合物0.919g,收率47.44%。
MS(ESI,pos.ion)m/z:260.9[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm)8.34(s,1H),7.34(s,1H),3.97(s,3H).
步骤2:4-溴-6-氧-1,6-二氢吡啶-3-腈
将4-碘-6-甲氧基烟碱腈(7.0g,26.92mmol)溶在DMF(105mL)中,加入吡啶氢溴酸盐(86.15g,538.40mmol),加热到100℃搅拌反应23h。冷却至室温,浓缩除去溶剂得到黄色固体,加入水(50mL),置于冰浴下搅拌反应液,产物析出,抽滤,用水(50mL)洗涤滤饼,干燥滤饼,得到白色固体化合物3.15g,收率58.80%。
MS(ESI,pos.ion)m/z:198.9[M+H]+.
步骤3:4-溴-6-氧代-1,6-二氢吡啶-3-甲醛
将4-溴-6-氧-1,6-二氢吡啶-3-腈(0.400g,2.01mmol)溶在甲酸(6mL)中,加入雷尼镍(0.35g,6.03mmol)加热回流反应3h,经硅藻土过滤,二氯甲烷(20mL)洗涤滤饼,浓缩溶剂,得绿色固体,加入乙醇(10mL)打浆得白色固体化合物0.270g,收率66.50%。
MS(ESI,pos.ion)m/z:202.0[M+H]+.
步骤4:4-溴-5-(哌啶-1-基甲基)吡啶-2(1H)-酮
将4-溴-6-氧-1,6-二氢吡啶-3-甲醛(0.248g,1.23mmol)溶在DCE(10mL)中,加入哌啶(0.19mL,1.88mmol)和三乙胺(0.34mL,2.47mmol),反应混合物于室温下搅拌反应1h,然后冰浴下加入三乙酰氧基硼氢化钠(1.04g,4.91mmol),加毕,室温搅拌反应12h。反应完后,加入饱和碳酸氢钠(20mL)和二氯甲烷(10mL),分液,水相用二氯甲烷(20mL×2)萃取,合并有机相,合并的有机相用无水硫酸钠干燥,浓缩溶剂,所得残留物经硅胶柱层析纯化(DCM/MeOH(V/V)=10/1)得黄色固体化合物0.250g,收率74.96%。
MS(ESI,pos.ion)m/z:271.0[M+H]+.
步骤5:4-溴-1-甲基-5-(哌啶-1-基甲基)吡啶-2(1H)-酮
以4-溴-5-(哌啶-1-基甲基)吡啶-2(1H)-酮和碘甲烷为原料,参考实施1例步骤9的合成方法得标题化合物为白色固体0.18g,收率78.53%。
MS(ESI,pos.ion)m/z:285.1[M+H]+.
步骤6:(R)-4-(2-氨基-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)-1-甲基-5-(哌啶-1-基甲基) 吡啶-2(1H)-酮
以4-溴-1-甲基-5-(哌啶-1-基甲基)吡啶-2(1H)-酮和(R)-(2-((2,4-二甲氧基苄基)氨基)-4-((1-羟基-2-甲基己烷-2-基)氨基)吡啶并[3,2-d]嘧啶-7-基)硼酸为起始物料参照实施例1步骤10至步骤11的合成方法,可得到标题化合物为类黄色固体,产率为40.75%。
MS(ESI,pos.ion)m/z:480.3[M+H]+
1H NMR(400MHz,CD3OD)δ(ppm)8.40(d,J=1.7Hz,1H),7.70(s,1H),7.64(d,J=1.7Hz,1H),6.52(s,1H),3.98(d,J=11.2Hz,1H),3.78(d,J=11.3Hz,1H),3.65(s,3H),3.19(s,2H),2.28–2.11(m,5H),1.91–1.80(m,1H),1.51(s,3H),1.43–1.32(m,10H),0.93(t,J=6.8Hz,3H).
生物学测试
测试1:对人源TLR7和TLR8的激动活性的测试
实验目的:应用HEK-BlueTMhTLR7和HEK-BlueTMhTLR8细胞检测化合物对hTLR7和hTLR8的激动活性和细胞毒活性。
实验步骤如下:
1)化合物配制:化合物先用DMSO配制成20mM的母液,然后将化合物3倍梯度稀释加入到96孔板中,共10个浓度,每个浓度双复孔。阴性对照孔每孔加入0.5μL DMSO。DMSO终浓度为0.5%。
2)将HEK-BlueTMhTLR7或HEK-BlueTMhTLR8细胞悬浮在培养液中,然后种到含有化合物的96孔板中,每孔细胞数为50,000。化合物和细胞在37℃,5%CO2条件下共孵育24h。
3)QUANTI-Blue检测溶液:取1mL的QB reagent和1mL的QB buffer,加入98mL的无菌水,混匀溶解,室温放置10min。
4)化合物活性测试:每孔取20μL的步骤2)中的培养上清液加入含有180μL QUANTI-Blue检测溶液的96孔板中,37℃条件下孵育1h后,用多功能酶标仪Flextation III检测650nm的吸光值(OD650)。
5)细胞活性检测:按照Celltiter-Glo说明书方法操作,化学发光信号(RLU)用多功能酶标仪Flextation III检测。
6)数据分析
化合物活性:OD650值用GraphPad Prism软件分析,并拟合化合物剂量效应曲线,计算化合物的EC50值。
细胞活性检测:细胞活性%计算公式如下。细胞活性%值用GraphPad Prism软件分析,并拟合化合物剂量效应曲线,计算化合物对细胞的CC50值。
细胞活性%=RLU化合物/RLUDMSO Control*100%
本发明化合物对hTLR8具有较好的激动活性,且对hTLR8有较好的选择性激活作用,本发明化合物对人源TLR7和TLR8的激动活性的测试结果见表A,并且本发明化合物对细胞的毒性小,本发明化合物对细胞的毒性结果见表A-1。
表A:本发明化合物对人源TLR7和TLR8的激动活性


注明:N/A表示未测
结论:实验数据表明,本发明化合物大部分对hTLR8具有较好的激动活性,且对hTLR8具有较好的选择性激活作用。
表A-1:本发明化合物对细胞的毒性

结论:实验数据表明,本发明化合物对细胞的毒性小。
测试2:自动膜片钳检测化合物对HEK293细胞稳定性表达的hERG通道电流的影响
实验目的:利用膜片钳技术和稳定表达hERG通道的HEK-293细胞系检测化合物对hERG通道的阻断作用浓度效应关系,从而评价其对心脏hERG钾通道抑制作用的风险。
实验方法:利用膜片钳技术将hERG细胞进行钳制,形成高阻封接后破膜进行全细胞记录模式开始记录hERG电流,每个药物浓度设定为两次给药,时间至少5分钟。测试浓度为0.3μM,1μΜ,3μΜ,10μΜ和30μΜ,观察测试化合物对hERG电流的变化,测试结果参考表B。
表B:本发明化合物hERG IC50
注明:对比化合物为(R)-2-((2-氨基-7-氟吡啶并[3,2-d]嘧啶-4-基)氨基)-2-甲基己-1-醇
结论:hERG测试实验数据表明,本发明化合物对心脏的毒性小。
测试3:本发明化合物对人肝微粒体CYP酶的抑制作用
实验目的:利用人肝微粒体体系评价测试化合物对微粒体中主要代谢酶CYP1A2、CYP2C19、CYP2D6和CYP3A4的抑制作用。
实验方法:受试物在最终孵育体系中的浓度为10μM,分别与含有细胞色素P450酶CYP1A2、CYP2C19、CYP2D6和CYP3A4的人肝微粒体混悬液一起进行孵育。已知底物被相应的单酶代谢转化生成特定的代谢产物,且代谢产物由UPLC-MS/MS方法测定。通过测试化合物与溶剂DMSO在同等时间内代谢产物生成减少百分比来计算相对抑制率%(Relative Inhibition(%))。
相对抑制率%=(1-(N+inh/Nveh))×100
N为探针底物代谢产物的浓度值并假定0min时有或无抑制剂组的带血产物均为0。N+inh为加抑制剂组代谢物浓度,Nveh为不加抑制剂组代谢物浓度。本发明化合物对人肝微粒体CYP酶的抑制测试结果见表C。
表C:本发明化合物对人肝微粒体CYP酶的抑制测试实验数据
结论:实验数据表明,本发明化合物对主要人肝微粒体CYP酶基本无抑制作用。
测试4:本发明化合物对人肝微粒体CYP酶的诱导作用
实验目的:利用冷冻人肝细胞作为测试体系,分别从酶活性和mRNA水平两方面评价受试物对CYP1A2、CYP2B6和CYP3A4的诱导作用。
实验方法:细胞铺板后24小时,加含待测试化合物的培养基开始诱导(测试浓度为10、1和0.1μM),每24小时换一次新鲜的含药培养基,诱导72小时后,将培养基换为含有CYP1A2、CYP2B6和CYP3A4特异性底物的培养基进行30分钟孵育,取100μL进行处理,检测底物的标记代谢产物进行酶活水平评价。最后将细胞裂解、反转录,用实时定量PCR的方法评估细胞中CYP1A2、CYP2B6和CYP3A4基因的表达量。
以与空白对照相比,测试化合物诱导倍数,和与阳性对照相比,测试化合物诱导百分数作为评价待测化合物具有诱导潜力的依据。其中,CYP1A2阳性对照为:奥美拉唑;CYP2B6阳性对照为:苯巴比妥;CYP3A4阳性对照为:利福平。对人肝微粒体CYP酶的诱导测试实验结果见表D。
表D:本发明化合物对人肝微粒体CYP酶的诱导测试实验数据

注明:对比化合物为(R)-2-((2-氨基-7-氟吡啶并[3,2-d]嘧啶-4-基)氨基)-2-甲基己-1-醇
结论:实验数据表明,本发明化合物对肝药酶CYP1A2、CYP2B6和CYP3A4基本无诱导作用。
测试5:本发明化合物在比格犬、小鼠、大鼠或食蟹猴中的药代动力学实验
(1)比格犬PK测试实验
本发明化合物在比格犬(购自湖南斯莱克景达实验动物有限公司,体重10-12kg,雄性,年龄10-12个月,口服每组3只,静脉注射每组3只)体内的PK测定实验方法:
比格犬经口灌胃给予2.5mg/kg或5mg/kg或经静脉注射0.5mg/kg或1mg/kg或2mg/kg的测试化合物。
给药后按时间点(0.083、0.25、0.5、1、2、4、6、8和24小时)静脉采血,收集于加EDTA-K2的抗凝管内。血浆样品经液液萃取后,在三重四极杆串联质谱仪上,以多重反应离子监测(MRM)方式进行定量分析。采用WinNonlin 6.3软件用非房室模型法计算药动学参数。
结论:药代实验数据表明,本发明化合物在比格犬体内具有较好的药代动力学性质,在抗HBV方面具有很好的应用前景。
(2)小鼠PK测试实验:
本发明化合物在ICR小鼠(购自湖南斯莱克景达实验动物有限公司,体重20-25g,雄性,年龄45-60天,口服每组3只,静脉注射每组3只)体内的PK测定实验方法:
ICR小鼠经口灌胃给予10mg/kg或经尾静脉注射2mg/kg或10mg/kg的测试化合物。
给药后按时间点(0.083,0.25,0.5,1,2,4,6,8和24小时)眼眶静脉采血,收集于加EDTA-K2的抗凝管内。血浆样品经液液萃取后,在三重四极杆串联质谱仪上,以多重反应离子监测(MRM)方式进行定量分析。采用WinNonlin 6.3软件用非房室模型法计算药动学参数。
结论:药代实验数据表明,本发明化合物在小鼠体内具有较好的药代动力学性质,在抗HBV方面具有很好的应用前景。
(3)SD大鼠PK测试实验:
本发明化合物在SD大鼠(购自湖南斯莱克景达实验动物有限公司,体重200-250g,雄性,年龄2-3个月,口服每组3只,静脉注射每组3只)体内的PK测定实验方法:
大鼠经口灌胃给予2.5mg/kg或5mg/kg或经静脉注射1mg/kg的测试化合物。
给药后按时间点(0.083、0.25、0.5、1、2、5、7和24小时)静脉采血,收集于加EDTA-K2的抗凝管内。血浆样品经液液萃取后,在三重四极杆串联质谱仪上,以多重反应离子监测(MRM)方式进行定量分析。采用WinNonlin 6.3软件用非房室模型法计算药动学参数。
结论:药代实验数据表明,本发明化合物在SD大鼠体内暴露量较大,说明本发明化合物在SD大鼠体内吸收好,并且具有很好的生物利用度,在抗HBV方面具有很好的应用前景。
(4)食蟹猴PK测试实验:
本发明化合物在食蟹猴(购自广东春盛生物科技发展有限公司,体重3-6kg,雄性,年龄4-6岁,口服 每组3只,静脉注射每组3只)体内的PK测定实验方法:
食蟹猴经口灌胃给予2.5mg/kg或5mg/kg或经静脉注射0.5mg/kg或1mg/kg的测试化合物。
给药后按时间点(0.083、0.25、0.5、1、2、4、6、8和24小时)静脉采血,收集于加EDTA-K2的抗凝管内。血浆样品经液液萃取后,在三重四极杆串联质谱仪上,以多重反应离子监测(MRM)方式进行定量分析。采用WinNonlin 6.3软件用非房室模型法计算药动学参数。
结论:药代实验数据表明,本发明化合物在食蟹猴体内具有较好的药代动力学性质,在抗HBV方面具有很好的应用前景。
虽然,上文中已经用一般性说明、具体实施方式及试验,对本发明作了详尽的描述,但在本发明基础上,可以对之作一些修改或改进,这对本领域技术人员而言是显而易见的。因此,在不偏离本发明精神的基础上所做的这些修改或改进,均属于本发明要求保护的范围。

Claims (22)

  1. 一种化合物,其为如式(I)所示的化合物或如式(I)所示的化合物的立体异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐或它的前药,
    其中,X为N或CR4
    各R1、R2、R4、R5、R6、R7和R8独立地为氢、氘、F、Cl、Br、I、羟基、氰基、氨基、C1-4烷氨基、C1-6烷氧基或C1-6烷基,其中,所述的C1-4烷氨基、C1-6烷氧基和C1-6烷基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基取代;
    Y为O或S;
    R3为-C1-6亚烷基-R10,其中,所述的-C1-6亚烷基-R10中的-C1-6亚烷基-未被取代或被1、2、3、4或5个Rw1的取代基取代;
    各R10独立地为C1-6烷氧基、-OH、-NH2、-O-C(=O)-R10a或-NRc-C(=O)-R10b,其中所述C1-6烷氧基未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基取代;
    各Rc独立地为氢、氘或C1-4烷基,其中,所述的C1-4烷基未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基取代;
    各R10a和R10b独立地为C1-6烷基、C3-7环烷基、5-12个环原子组成的杂芳基或3-12个环原子组成的杂环基,其中,所述的C1-6烷基、C3-7环烷基、5-12个环原子组成的杂芳基和3-12个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw2的取代基取代;
    R为H、氘、C1-6烷基或3-6个环原子组成的杂环基,其中,所述的C1-6烷基和3-6个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw3的取代基取代;
    R9为氢、氘、-L-R11、F、Cl、Br、I、羟基、氰基、氨基、C1-4烷氨基、C1-6烷氧基或C1-6烷基,其中,所述的C1-4烷氨基、C1-6烷氧基和C1-6烷基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基取代;
    L为-C1-6亚烷基-或-C(=O)-,其中,所述的-C1-6亚烷基-未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基或异丙基的取代基取代;
    R11为-NRaRb、C1-12烷基、-C(=O)R9a、-OR9b、-S(=O)tR9c、C3-7环烷基、5-12个环原子组成的杂芳基或3-12个环原子组成的杂环基,其中,所述的C1-12烷基、C3-7环烷基、5-12个环原子组成的杂芳基和3-12个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw4的取代基取代;
    各Ra和Rb独立地为氢、氘、-C(=O)R9d、C1-12烷基、C2-12烯基、C2-12炔基或-C1-6亚烷基-Rd1,其中,所述的C1-12烷基、C2-12烯基、C2-12炔基和-C1-6亚烷基-Rd1中的-C1-6亚烷基-各自独立地未被取代或被1、2、3、4或5个Rw5的取代基取代;
    或,Ra、Rb以及与它们所连接的N原子一起形成3-8个环原子组成的杂环基,其中,所述的3-8个环原子组成的杂环基未被取代或被1、2、3或4个Rw4取代;
    各R9a、R9b、R9c和R9d独立地为C1-12烷基、C2-12烯基、C2-12炔基或-C1-6亚烷基-Rd2,其中,所述的C1-12烷基、C2-12烯基、C2-12炔基和-C1-6亚烷基-Rd2中的-C1-6亚烷基-各自独立地未被取代或被1、2、3、4 或5个Rw6的取代基取代;
    各Rd1和Rd2独立地为羟基、C1-6烷氧基、C6-10芳基、5-12个环原子组成的杂芳基、C3-6环烷基或C1-6烷氨基,其中,所述的C1-6烷氧基、C6-10芳基、5-12个环原子组成的杂芳基、C3-6环烷基和C1-6烷氨基各自独立地未被取代或被1、2、3、4或5个Rw7的取代基取代;
    各Rw1、Rw2、Rw3、Rw4和Rw7独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、C1-6烷氨基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基、-C(=O)-C1-4烷基、C3-6环烷基、3-6个环原子组成的杂环基或C6-10芳基,其中,所述的C1-6烷氨基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基、-C(=O)-C1-4烷基、C3-6环烷基、3-6个环原子组成的杂环基和C6-10芳基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、C1-4烷基和C1-4烷氨基的取代基取代;
    各Rw5和Rw6独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、C1-6烷氨基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基、-C(=O)-C1-4烷基、C2-12烯基、C2-12炔基、C3-6环烷基、3-6个环原子组成的杂环基、5-6个环原子组成的杂芳基或C6-10芳基,其中,所述的氨基、C1-6烷氨基、C1-6烷氧基、C1-6烷基、C1-6卤代烷基、-C(=O)-C1-4烷基、C2-12烯基、C2-12炔基、C3-6环烷基、3-6个环原子组成的杂环基、5-6个环原子组成的杂芳基和C6-10芳基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、C1-4烷基和C1-4烷氨基的取代基取代;
    t为0、1或2。
  2. 根据权利要求1所述的化合物,其中,各R1、R2、R4、R5、R6、R7和R8独立地为氢、氘、F、Cl、Br、I、羟基、氰基、氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基或正己基,其中,所述的N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基和正己基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基和仲丁基的取代基取代;
    R为H、氘、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、正己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基或哌嗪基,其中,所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基和哌嗪基各自独立地未被取代或被1、2、3、4或5个Rw3的取代基取代。
  3. 根据权利要求1或2所述的化合物,其中,R3为-CH2-R10、-(CH2)2-R10、-(CH2)3-R10、-CH(CH3)CH2-R10、-CH2CH(CH3)-R10或-(CH2)4-R10,其中,所述的-CH2-R10中的-CH2-、-(CH2)2-R10中的-(CH2)2-、-(CH2)3-R10中的-(CH2)3-、-CH(CH3)CH2-R10中的-CH(CH3)CH2-、-CH2CH(CH3)-R10中的-CH2CH(CH3)-和-(CH2)4-R10中的-(CH2)4-各自独立地未被取代或被1、2、3、4或5个Rw1的取代基取代;
    各R10独立地为甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、-OH、-NH2、-O-C(=O)-R10a或-NRc-C(=O)-R10b,其中,所述的甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基和2-丁氧基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基和仲丁基的取代基取代;
    各R10a和R10b独立地为C1-4烷基、C3-6环烷基、5-6个环原子组成的杂芳基或3-6个环原子组成的杂环基,其中,所述的C1-4烷基、C3-6环烷基、5-6个环原子组成的杂芳基和3-6个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw2的取代基取代;
    Rc为氢、氘、甲基、乙基、正丙基或异丙基,其中,所述的甲基、乙基、正丙基和异丙基未被取代或 被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基和异丙基的取代基取代。
  4. 根据权利要求1-3任意一项所述的化合物,其中,各R10a和R10b独立地为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、环丙基、环丁基、环戊基、环己基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基或哌嗪基,其中,所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、环丙基、环丁基、环戊基、环己基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基和哌嗪基各自独立地未被取代或被1、2、3、4或5个Rw2的取代基取代。
  5. 根据权利要求1-4任意一项所述的化合物,其中,R9为氢、氘、-L-R11、F、Cl、Br、I、羟基、氰基、氨基、C1-4烷氨基、C1-4烷氧基或C1-4烷基,其中,所述的C1-4烷氨基、C1-4烷氧基和C1-4烷基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基和C1-4烷基的取代基所取代;
    L为-CH2-、-(CH2)2-、-(CH2)3-、-CH2CH(CH3)-、-CH(CH3)CH2-、-(CH2)4-或-C(=O)-,其中,所述的-CH2-、-(CH2)2-、-(CH2)3-、-CH2CH(CH3)-、-CH(CH3)CH2-和-(CH2)4-各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基或异丙基的取代基取代;
    R11为-NRaRb、C1-10烷基、-C(=O)R9a、-OR9b、-S(=O)tR9c、C3-6环烷基、5-6个环原子组成的杂芳基或3-6个环原子组成的杂环基,其中,所述的C1-10烷基、C3-6环烷基、5-6个环原子组成的杂芳基和3-6个环原子组成的杂环基各自独立地未被取代或被1、2、3、4或5个Rw4的取代基取代。
  6. 根据权利要求1-5任意一项所述的化合物,其中,R9为氢、氘、-L-R11、F、Cl、Br、I、羟基、氰基、氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基或正己基,其中,所述的N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基和正己基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基和仲丁基的取代基所取代;
    R11为-NRaRb、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、正己基、-C(=O)R9a、-OR9b、-S(=O)tR9c、环丙基、环丁基、环戊基、环己基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基或哌嗪基,其中,所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、环丙基、环丁基、环戊基、环己基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基和哌嗪基各自独立地未被取代或被1、2、3、4或5个Rw4的取代基取代。
  7. 根据权利要求1-6任意一项所述的化合物,其中,各Ra和Rb独立地为氢、氘、-C(=O)R9d、C1-6烷基、C2-6烯基、C2-6炔基或-C1-4亚烷基-Rd1,其中,所述的C1-6烷基、C2-6烯基、C2-6炔基和-C1-4亚烷基-Rd1 中的-C1-4亚烷基-各自独立地未被取代或被1、2、3、4或5个Rw5的取代基取代;
    或,Ra、Rb以及与它们所连接的N原子一起形成3-6个环原子组成的杂环基,其中,所述的3-6个环原子组成的杂环基未被取代或被1、2、3或4个Rw4取代。
  8. 根据权利要求1-7任意一项所述的化合物,其中,各Ra和Rb独立地为氢、氘、-C(=O)R9d、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、正己基、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、-CH2-Rd1、-(CH2)2-Rd1、-(CH2)3-Rd1、-CH2CH(CH3)-Rd1、-CH(CH3)CH2-Rd1或-(CH2)4-Rd1,其中,所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、-CH2-Rd1中的-CH2-、-(CH2)2-Rd1中的-(CH2)2-、-(CH2)3-Rd1中的-(CH2)3-、-CH2CH(CH3)-Rd1中的-CH2CH(CH3)-、-CH(CH3)CH2-Rd1中的-CH(CH3)CH2-和-(CH2)4-Rd1中的-(CH2)4-各自独立地未被取代或被1、2、3、4或5个Rw5的取代基取代;
    或,Ra、Rb以及与它们所连接的N原子一起形成氮杂环丙基、氮杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、哌啶基、吗啉基、硫代吗啉基或哌嗪基,其中,所述的氮杂环丙基、氮杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、哌啶基、吗啉基、硫代吗啉基和哌嗪基各自独立地未被取代或被1、2、3或4个Rw4取代。
  9. 根据权利要求1-8任意一项所述的化合物,其中,各R9a、R9b、R9c和R9d独立地为C1-6烷基、C2-6烯基、C2-6炔基或-C1-4亚烷基-Rd2,其中所述的C1-6烷基、C2-6烯基、C2-6炔基和-C1-4亚烷基-Rd2中的-C1-4亚烷基-各自独立地未被取代或被1、2、3、4或5个Rw6的取代基取代;
    各Rd1和Rd2独立地为羟基、C1-4烷氧基、苯基、5-6个环原子组成的杂芳基、环丙基、环丁基、环戊基、环己基或C1-4烷氨基,其中,所述的C1-4烷氧基、苯基、5-6个环原子组成的杂芳基、环丙基、环丁基、环戊基、环己基和C1-4烷氨基各自独立地未被取代或被1、2、3、4或5个Rw7的取代基取代。
  10. 根据权利要求1-9任意一项所述的化合物,其中,各R9a、R9b、R9c和R9d独立地为甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、-CH2-Rd2、-(CH2)2-Rd2、-(CH2)3-Rd2、-CH2CH(CH3)-Rd2、-CH(CH3)CH2-Rd2或-(CH2)4-Rd2,其中所述的甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、正己基、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、-CH2-Rd2中的-CH2-、-(CH2)2-Rd2中的-(CH2)2-、-(CH2)3-Rd2中的-(CH2)3-、-CH2CH(CH3)-Rd2中的-CH2CH(CH3)-、-CH(CH3)CH2-Rd2中的-CH(CH3)CH2-和-(CH2)4-Rd2中的-(CH2)4-各自独立地未被取代或被1、2、3、4或5个Rw6的取代基取代;
    各Rd1和Rd2独立地为羟基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、环丙基、环丁基、环戊基、环己基、N-甲氨基、N-乙氨基、N,N-二甲氨基或N,N-二乙氨基,其中,所述的甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基、环丙基、环丁基、环戊基、环己基、N-甲氨基、N-乙氨基、N,N-二甲氨基和N,N-二乙氨基各自独立地未被取代或被1、2、3、4或5个Rw7的取代基取代。
  11. 根据权利要求1-10任意一项所述的化合物,其中,各Rw1、Rw2、Rw3、Rw4和Rw7独立地为氘、F、 Cl、Br、I、=O、羟基、氰基、氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、-CH2F、-CH2Cl、-CF3、-CHF2、-CHCl2、-CH2CH2F、-CH2CH2Cl、-CH2CHF2、-CH2CHCl2、-CHFCH2F、-CHClCH2Cl、-CH2CF3、-CH(CF3)2、-CF2CH2CH3、-CH2CH2CH2F、-CH2CH2CHF2、-CH2CH2CF3、-C(=O)-CH3、-C(=O)-CH2CH3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基或苯基,其中,所述的N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、-CH2F、-CH2Cl、-CHF2、-CHCl2、-CH2CH2F、-CH2CH2Cl、-CH2CHF2、-CH2CHCl2、-CHFCH2F、-CHClCH2Cl、-CH2CF3、-CH(CF3)2、-CF2CH2CH3、-CH2CH2CH2F、-CH2CH2CHF2、-CH2CH2CF3、-C(=O)-CH3、-C(=O)-CH2CH3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基和苯基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、-N(CH3)2、-NHCH3、-N(CH2CH3)2、-N(CH3)CH2CH3和-NHCH2CH3的取代基取代;
    各Rw5和Rw6独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、C1-4烷氨基、C1-4烷氧基、C1-4烷基、C1-4卤代烷基、-C(=O)-C1-4烷基、C2-6烯基、C2-6炔基、环丙基、环丁基、环戊基、环己基、3-6个环原子组成的杂环基、5-6个环原子组成的杂芳基或苯基,其中,所述的氨基、C1-4烷氨基、C1-4烷氧基、C1-4烷基、C1-4卤代烷基、-C(=O)-C1-4烷基、C2-6烯基、C2-6炔基、环丙基、环丁基、环戊基、环己基、3-6个环原子组成的杂环基、5-6个环原子组成的杂芳基和苯基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、C1-4烷基和C1-4烷氨基的取代基取代。
  12. 根据权利要求1-11任意一项所述的化合物,其中,各Rw5和Rw6独立地为氘、F、Cl、Br、I、=O、羟基、氰基、氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、-CH2F、-CH2Cl、-CF3、-CHF2、-CHCl2、-CH2CH2F、-CH2CH2Cl、-CH2CHF2、-CH2CHCl2、-CHFCH2F、-CHClCH2Cl、-CH2CF3、-CH(CF3)2、-CF2CH2CH3、-CH2CH2CH2F、-CH2CH2CHF2、-CH2CH2CF3、-C(=O)-CH3、-C(=O)-CH2CH3、-C(=O)-CH2CH2CH3、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基或苯基,其中,所述的氨基、N-甲氨基、N-乙氨基、N,N-二甲氨基、N,N-二乙氨基、甲氧基、乙氧基、1-丙氧基、2-丙氧基、1-丁氧基、2-甲基-l-丙氧基、2-丁氧基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、-CH2F、-CH2Cl、-CHF2、-CHCl2、-CH2CH2F、-CH2CH2Cl、-CH2CHF2、-CH2CHCl2、-CHFCH2F、-CHClCH2Cl、-CH2CF3、-CH(CF3)2、-CF2CH2CH3、-CH2CH2CH2F、-CH2CH2CHF2、-CH2CH2CF3、-C(=O)-CH3、-C(=O)-CH2CH3、-C(=O)-CH2CH2CH3、-CH=CH2、-CH=CHCH3、-CH2CH=CH2、-CH2CH2CH=CH2、-C≡CH、-CH2C≡CH、-C≡C-CH3、-CH2CH2C≡CH、-CH2C≡CCH3、-C≡CCH2CH3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、吡唑烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、三唑基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基和苯基各自独立地未被取代或被1、2、3或4个独立地选自F、Cl、Br、I、羟基、氰基、氨基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、-N(CH3)2、-NHCH3、-N(CH2CH3)2、-N(CH3)CH2CH3和-NHCH2CH3的取代基取代。
  13. 根据权利要求1-12任意一项所述的化合物,其包含以下其中之一的结构:






    或其立体异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐或它的前药。
  14. 一种药物组合物,包含权利要求1-13任意一项所述的化合物,及其药学上可接受的辅料。
  15. 根据权利要求14所述的药物组合物,其更进一步地包含其它一种或多种治疗剂,其中所述治疗剂为HBV DNA聚合酶抑制剂、toll-样受体7调节剂、toll-样受体8调节剂、toll-样受体7和8调节剂、toll-样受体3调节剂、干扰素α配体、HBsAg抑制剂、靶向HbcAg的化合物、亲环蛋白抑制剂、HBV治疗性疫苗、HBV预防性疫苗、HBV病毒进入抑制剂、NTCP抑制剂、靶向病毒mRNA的反义寡核苷酸、短干扰RNA、乙型肝炎病毒E抗原抑制剂、HBx抑制剂、cccDNA抑制剂、HBV抗体、胸腺素激动剂、细胞因子、核蛋白抑制剂、视黄酸诱导基因1的刺激剂、NOD2刺激剂、重组胸腺素α-1、乙型肝炎病毒复制抑制剂、乙型肝炎表面抗原分泌或组装抑制剂、IDO抑制剂或其组合。
  16. 根据权利要求14所述的药物组合物,其中所述治疗剂为拉米夫定、替比夫定、替诺福韦、恩替卡韦、阿德福韦酯、替诺福韦艾拉酚胺、替诺福韦二吡呋酯、替诺福韦艾拉酚胺富马酸盐、替诺福韦艾拉酚胺半富马酸盐、Alfaferone、Alloferon、西莫白介素、克拉夫定、恩曲他滨、法昔洛韦、干扰素、宝甘灵CP、因特芬、白细胞介素-2、米伏替酯、硝唑尼特、病毒唑、罗扰素-A、西佐喃、Euforavac、安普利近、Phosphazid、Heplisav、重组人白细胞介素-2、左旋咪唑或丙帕锗。
  17. 权利要求1-13任意一项所述的化合物或权利要求14-16任意一项所述的药物组合物在制备预防、处理、治疗或减轻患者由TLR8介导的疾病的药物中的用途。
  18. 根据权利要求17所述的用途,其中,所述由TLR8介导的疾病为乙型肝炎病毒感染、丙型肝炎病毒感染、流感病毒感染、疱疹病毒感染、艾滋病毒感染、过敏性疾病、风湿性关节炎、过敏性哮喘、慢性疲劳、II型糖尿病、枯草热、红瘢狼疮、多发性硬化症、黑色素瘤、肺癌、肝癌、基底细胞癌、肾癌、骨髓瘤、胆道癌、脑癌、乳腺癌、宫颈癌、绒毛膜癌、结肠癌、直肠癌、头颈癌、腹膜肿瘤、输卵管癌、子宫内膜癌、食道癌、胃癌、白血病、淋巴瘤、肉瘤、成神经细胞瘤、口腔癌、卵巢癌、胰腺癌、前列腺癌、睾丸癌、皮肤癌或甲状腺癌。
  19. 权利要求1-13任意一项所述的化合物或权利要求14-16任意一项所述的药物组合物用于预防、处理、治疗或减轻患者由TLR8介导的疾病。
  20. 根据权利要求19所述化合物或药物组合物,其中,所述由TLR8介导的疾病为乙型肝炎病毒感染、丙型肝炎病毒感染、流感病毒感染、疱疹病毒感染、艾滋病毒感染、过敏性疾病、风湿性关节炎、过敏性哮喘、慢性疲劳、II型糖尿病、枯草热、红瘢狼疮、多发性硬化症、黑色素瘤、肺癌、肝癌、基底细胞癌、肾癌、骨髓瘤、胆道癌、脑癌、乳腺癌、宫颈癌、绒毛膜癌、结肠癌、直肠癌、头颈癌、腹膜肿瘤、输卵管癌、子宫内膜癌、食道癌、胃癌、白血病、淋巴瘤、肉瘤、成神经细胞瘤、口腔癌、卵巢癌、胰腺癌、前列腺癌、睾丸癌、皮肤癌或甲状腺癌。
  21. 一种预防、处理、治疗或减轻患者由TLR8介导的疾病的方法,包括给与患者有效治疗量的权利要求1-13任意一项所述的化合物或权利要求14-16任意一项所述的药物组合物。
  22. 根据权利要求21所述化合物或药物组合物,其中,所述由TLR8介导的疾病为乙型肝炎病毒感染、丙型肝炎病毒感染、流感病毒感染、疱疹病毒感染、艾滋病毒感染、过敏性疾病、风湿性关节炎、过敏性哮喘、慢性疲劳、II型糖尿病、枯草热、红瘢狼疮、多发性硬化症、黑色素瘤、肺癌、肝癌、基底细胞癌、肾癌、骨髓瘤、胆道癌、脑癌、乳腺癌、宫颈癌、绒毛膜癌、结肠癌、直肠癌、头颈癌、腹膜肿瘤、输卵管癌、子宫内膜癌、食道癌、胃癌、白血病、淋巴瘤、肉瘤、成神经细胞瘤、口腔癌、卵巢癌、胰腺癌、前列腺癌、睾丸癌、皮肤癌或甲状腺癌。
PCT/CN2023/077368 2022-02-24 2023-02-21 嘧啶并芳香环类化合物及其在药物中的应用 WO2023160527A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210172494.6 2022-02-24
CN202210172494 2022-02-24

Publications (1)

Publication Number Publication Date
WO2023160527A1 true WO2023160527A1 (zh) 2023-08-31

Family

ID=87764763

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/077368 WO2023160527A1 (zh) 2022-02-24 2023-02-21 嘧啶并芳香环类化合物及其在药物中的应用

Country Status (2)

Country Link
CN (1) CN116693527A (zh)
WO (1) WO2023160527A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101010299A (zh) * 2004-06-15 2007-08-01 布里斯托尔-迈尔斯斯奎布公司 用作丝氨酸蛋白酶抑制剂的6元杂环
CN108069963A (zh) * 2017-11-17 2018-05-25 清华大学 吡啶并嘧啶衍生物或其盐及其制法、药物组合物和用途
WO2020007275A1 (zh) * 2018-07-03 2020-01-09 江苏恒瑞医药股份有限公司 吡啶并嘧啶类衍生物、其制备方法及其在医药上的应用
CN112225738A (zh) * 2020-12-18 2021-01-15 北京鑫开元医药科技有限公司 作为tlr8激动剂的化合物、其制备方法、组合物及其用途
CN112390795A (zh) * 2019-08-19 2021-02-23 上海挚盟医药科技有限公司 一类2-氨基嘧啶类化合物及其药物组合物和用途

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101010299A (zh) * 2004-06-15 2007-08-01 布里斯托尔-迈尔斯斯奎布公司 用作丝氨酸蛋白酶抑制剂的6元杂环
CN108069963A (zh) * 2017-11-17 2018-05-25 清华大学 吡啶并嘧啶衍生物或其盐及其制法、药物组合物和用途
WO2020007275A1 (zh) * 2018-07-03 2020-01-09 江苏恒瑞医药股份有限公司 吡啶并嘧啶类衍生物、其制备方法及其在医药上的应用
CN112390795A (zh) * 2019-08-19 2021-02-23 上海挚盟医药科技有限公司 一类2-氨基嘧啶类化合物及其药物组合物和用途
CN112225738A (zh) * 2020-12-18 2021-01-15 北京鑫开元医药科技有限公司 作为tlr8激动剂的化合物、其制备方法、组合物及其用途

Also Published As

Publication number Publication date
CN116693527A (zh) 2023-09-05

Similar Documents

Publication Publication Date Title
CN110036005B (zh) 酰胺衍生物及其在药物中的应用
JP2020537677A (ja) ジヒドロピリミジン化合物、及び医薬におけるその使用
CN109251166B (zh) 抑制ssao/vap-1的胺类化合物及其在医药上的应用
CN107759571B (zh) 流感病毒复制抑制剂及其使用方法和用途
CN108218873B (zh) 流感病毒复制抑制剂及其用途
JP2022507724A (ja) 抗ウイルス剤としての官能化複素環
AU2018290511B2 (en) Dihydropyrimidine compounds and uses thereof in medicine
CN108976223B (zh) 稠合三环类化合物及其在药物中的应用
CN113316576A (zh) 用于治疗癌症的作为HPK1抑制剂的2,3-二氢-1H-吡咯并[3,4-c]吡啶-1-酮衍生物
CN109988106B (zh) 抑制ssao/vap-1的胺类化合物及其在医药上的应用
CN110862390B (zh) 稠合三环类化合物及其在药物中的应用
TW202136254A (zh) 可作為t細胞活化劑之經取代雙環化合物
CN112789277B (zh) 胍类衍生物及其用途
WO2018127096A1 (en) Inhibitors of influenza virus replication and uses thereof
CN110914234A (zh) 抑制ssao/vap-1的胺类化合物及其在医药上的应用
CN110964023B (zh) 稠合四环类化合物及其在药物中的应用
KR20240024935A (ko) Pd-l1을 표적화하기 위한 방법 및 조성물
CN110446711B (zh) 流感病毒复制抑制剂及其用途
WO2022143610A1 (zh) 新型酰胺吡咯类化合物及其在药物中的应用
WO2023160527A1 (zh) 嘧啶并芳香环类化合物及其在药物中的应用
CN111217811B (zh) 稠合三环类化合物及其在药物中的应用
CN113072476B (zh) RORγt抑制剂及其制备方法和用途
WO2022156758A1 (zh) 新型酰胺吡咯类化合物及其在药物中的用途
CN110903284B (zh) 稠合三环类化合物及其在药物中的应用
WO2021197486A1 (zh) 新型螺环类化合物及其在药物中的应用

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: 23759152

Country of ref document: EP

Kind code of ref document: A1