WO2018041122A1 - 氧代吡啶酰胺类衍生物、其制备方法及其在医药上的应用 - Google Patents

氧代吡啶酰胺类衍生物、其制备方法及其在医药上的应用 Download PDF

Info

Publication number
WO2018041122A1
WO2018041122A1 PCT/CN2017/099579 CN2017099579W WO2018041122A1 WO 2018041122 A1 WO2018041122 A1 WO 2018041122A1 CN 2017099579 W CN2017099579 W CN 2017099579W WO 2018041122 A1 WO2018041122 A1 WO 2018041122A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound
formula
cycloalkyl
alkyl
Prior art date
Application number
PCT/CN2017/099579
Other languages
English (en)
French (fr)
Inventor
杨方龙
王伟民
李晓东
陈刚
贺峰
陶维康
Original Assignee
江苏恒瑞医药股份有限公司
上海恒瑞医药有限公司
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
Priority to DK17845424.5T priority Critical patent/DK3486242T3/da
Priority to US16/328,128 priority patent/US10633375B2/en
Priority to CN202111196664.6A priority patent/CN113929618B/zh
Priority to UAA201902875A priority patent/UA125520C2/uk
Priority to CA3031592A priority patent/CA3031592A1/en
Priority to AU2017317988A priority patent/AU2017317988B2/en
Priority to MX2019001481A priority patent/MX2019001481A/es
Application filed by 江苏恒瑞医药股份有限公司, 上海恒瑞医药有限公司 filed Critical 江苏恒瑞医药股份有限公司
Priority to JP2019511336A priority patent/JP7032814B2/ja
Priority to PL17845424T priority patent/PL3486242T3/pl
Priority to BR112019003557-5A priority patent/BR112019003557A2/pt
Priority to CN201780004214.8A priority patent/CN108495850B/zh
Priority to ES17845424T priority patent/ES2903283T3/es
Priority to RU2019107938A priority patent/RU2742771C2/ru
Priority to CN202111197415.9A priority patent/CN113912586B/zh
Priority to EP17845424.5A priority patent/EP3486242B1/en
Priority to KR1020197007965A priority patent/KR102416718B1/ko
Publication of WO2018041122A1 publication Critical patent/WO2018041122A1/zh
Priority to HK18114604.9A priority patent/HK1255566A1/zh
Priority to US16/804,180 priority patent/US11084808B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4418Non condensed pyridines; Hydrogenated derivatives thereof having a carbocyclic group directly attached to the heterocyclic ring, e.g. cyproheptadine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/63One oxygen atom
    • C07D213/64One oxygen atom attached in position 2 or 6
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/69Two or more oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/89Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members with hetero atoms directly attached to the ring nitrogen atom
    • 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
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/06Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/10Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the present invention relates to a novel class of oxopyridamide derivatives, a process for the preparation thereof, and a pharmaceutical composition containing the same, and as a therapeutic agent, in particular as an inhibitor of Factor XIa (Factor XIa, abbreviated as FXIa) and in preparation Use in medicines for the treatment and prevention of diseases such as thromboembolism.
  • a novel class of oxopyridamide derivatives a process for the preparation thereof, and a pharmaceutical composition containing the same, and as a therapeutic agent, in particular as an inhibitor of Factor XIa (Factor XIa, abbreviated as FXIa) and in preparation Use in medicines for the treatment and prevention of diseases such as thromboembolism.
  • Factor XIa Factor XIa
  • cardiovascular and cerebrovascular diseases such as cerebrovascular, cerebral infarction, myocardial infarction, coronary heart disease and arteriosclerosis take nearly 12 million lives, which is close to 1/4 of the world's total death toll, becoming the number one enemy of human health.
  • the number of people dying from cardiovascular disease in China each year is more than 2.6 million, and 75% of surviving patients are disabled, and more than 40% of them are severely disabled.
  • the problem of thrombosis caused by cardiovascular and cerebrovascular diseases and diabetes and its complications has become an urgent problem to be solved today.
  • Human blood coagulation is composed of an intrinsic pathway, an extrinsic pathway, and a common pathway (Annu. Rev. Med. 2011. 62: 41–57), which is sequentially activated by multiple zymogens. The process continues to be a chain reaction that is strengthened and amplified.
  • the coagulation cascade is initiated by the endogenous pathway (also known as the contact activation pathway) and the exogenous pathway (also known as the tissue factor pathway) to produce FXa, which is then produced by a common pathway to form thrombin (FIIa), which ultimately forms fibrin.
  • endogenous pathway also known as the contact activation pathway
  • exogenous pathway also known as the tissue factor pathway
  • the endogenous pathway refers to the process by which the XII factor activates the XIa-VIIIa-Ca2 ⁇ P L complex and activates the X factor, and the exogenous coagulation pathway is released from the tissue factor (TF) to the TF-VIIaCa2+ complex.
  • the process of forming and activating factor X refers to the process in which the two pathways are combined into one after the formation of factor Xa, which activates prothrombin and finally produces fibrin, where FXI is necessary for maintaining the endogenous pathway and is amplified during the coagulation cascade. Play a key role.
  • thrombin In the coagulation cascade, thrombin can feedbackly activate FXI, and activated FXI (FXIa) in turn promotes the production of thrombin, which amplifies the coagulation cascade. Therefore, antagonists of FXI have been extensively developed for the treatment of various thrombi.
  • FXI activity ⁇ 15U/dL Human FXI deficiency
  • type C hemophilia Human FXI deficiency
  • This type of patient has a mild bleed phenotype and spontaneous spontaneous bleeding. Even in the event of injury or surgery, the body's hemostatic function is not affected.
  • Patients with Friend C can be pregnant normally (Arterioscler Thromb Vasc Biol. 2010; 30: 388-392). This shows that FXIa security is significantly better than FXa. Therefore, the target FXIa has become a hot research topic among major companies and research institutions.
  • inhibition of FXIa factor can effectively inhibit thrombus formation, but in the case of more severe thrombosis, FXIa has little effect (Blood.
  • FXIa is an emerging target, and patents which disclose compounds having FXIa inhibitory activity are disclosed in WO9630396, WO9941276, WO2013093484, WO2004002405, WO2013056060, WO2017005725, and US20050171148. Among them, only Bayer's antisense oligonucleotides (ASO) BAY-2306001 entered the clinical phase II study and achieved good results.
  • ASO antisense oligonucleotides
  • the present invention devises a novel small molecule FXIa antagonist having the general structure (AI) wherein R 1 is C(O)R 7 , which has a significant improvement in anticoagulant action and pharmacogen absorption activity of the entire compound.
  • the compounds of the invention have higher activity than the disclosed patented compounds having a similar core structure.
  • the compound of the present invention exhibits excellent anticoagulant action against human blood and has good pharmacokinetic activity, and can be used for effectively treating and/or preventing cardiovascular and cerebrovascular diseases and thrombotic symptoms.
  • Ring A is an aryl or heteroaryl group
  • R 1 is -C(O)R 7 ;
  • R 2 are the same or different and are each independently selected from the group consisting of a hydrogen atom, a halogen, an alkyl group, a halogenated alkyl group, an alkoxy group, a halogenated alkoxy group, an amino group, a nitro group, a cyano group, a hydroxyl group, a hydroxyalkyl group, a cycloalkyl group, and a heterocyclic group. a cyclic group, an aryl group and a heteroaryl group;
  • R 3 is selected from the group consisting of halogen, alkyl, haloalkyl, alkoxy, cycloalkyloxy, haloalkoxy, amino, nitro, cyano, hydroxy, hydroxyalkyl and alkylthio, wherein said alkyl group Alkoxy, haloalkyl, haloalkoxy, hydroxyalkyl and alkylthio are optionally further selected from the group consisting of a halogen atom, a halogen, an alkoxy group, a halogenated alkoxy group, an amino group, a nitro group, a cyano group, a hydroxyl group and a hydroxyalkane group. Substituted by one or more substituents in the group;
  • R 4 is selected from the group consisting of a hydrogen atom, an alkyl group, an alkoxy group, a halogenated alkyl group, a hydroxyalkyl group, a cycloalkyl group, a cycloalkyloxy group, a heterocyclic group, an aryl group, and a heteroaryl group, wherein the alkyl group and the ring are The alkyl, heterocyclyl, aryl and heteroaryl are optionally further substituted with one or more R 9 substituents;
  • R 5 are the same or different and are each independently selected from the group consisting of a hydrogen atom, an alkyl group, an alkoxy group, an oxo group, a halogen, a halogenated alkyl group, a halogenated alkoxy group, an amino group, a nitro group, a cyano group, a hydroxyl group, a hydroxyalkyl group, and a ring.
  • alkyl Alkyl, heterocyclic, aryl, heteroaryl, -C(O)R 8 , -C(O)OR 8 , -NHC(O)R 8 , -NHC(O)OR 8 , -NR 10 R 11 , -C(O)NR 10 R 11 , -CH 2 NHC(O)OR 8 , -CH 2 NR 10 R 11 , -C(O)OCH(R 10 )R 11 and -S(O) m R 8 wherein the alkyl group is optionally further selected from the group consisting of a halogen atom, a halogen, an alkoxy group, a halogenated alkoxy group, an amino group, a nitro group, a cyano group, a hydroxyl group, a hydroxyalkyl group, -NR 10 R 11 and -NHC ( O) substituted with one or more substituents in OR 8 ;
  • R 7 is selected from the group consisting of a hydrogen atom, an alkyl group, a halogenated alkyl group, an alkoxy group, a halogenated alkoxy group, an amino group, a hydroxyl group, a hydroxyalkyl group, a cycloalkyl group, a heterocyclic group, an aryl group and a heteroaryl group, wherein said alkyl group Alkoxy, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally further selected from the group consisting of a hydrogen atom, a halogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkoxy group, an amino group, a nitro group, and a cyano group.
  • R 8 is selected from the group consisting of a hydrogen atom, an alkyl group, a halogenated alkyl group, a hydroxyl group, an amino group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group;
  • R 9 is selected from the group consisting of a hydrogen atom, a halogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, an amino group, a nitro group, a cyano group, a hydroxyl group, a hydroxyalkyl group, a cycloalkyl group, a cycloalkyloxy group, a heterocyclic group, Aryl and heteroaryl, wherein said cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally further selected from the group consisting of a halogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, an amino group, a nitro group Substituting one or more substituents of cyano, hydroxy, hydroxyalkyl, cycloalkyl, cycloalkyloxy, heterocyclyl, -NHC(O)R 12
  • R 10 and R 11 are the same or different and are each independently selected from a hydrogen atom, an alkyl group, a halogenated alkyl group, a cycloalkyl group, a heterocyclic group, -C(O)OR 8 and -OC(O)OR 12 , wherein The cycloalkyl and heterocyclic groups are optionally further selected from the group consisting of a halogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, an oxo group, an amino group, a nitro group, a cyano group, a hydroxyl group, and a hydroxyalkyl group. Substituted by a plurality of substituents;
  • R 12 is selected from the group consisting of a hydrogen atom, an alkyl group, an alkoxy group, a halogenated alkyl group, a halogenated alkoxy group, an amino group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group;
  • R 13 is an aryl or heteroaryl group, wherein the aryl and heteroaryl are optionally further selected from the group consisting of a hydrogen atom, a halogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, an amino group, a nitro group, and a cyano group. Substituted by one or more substituents of a hydroxy group, a hydroxyalkyl group, a cycloalkyl group, a cycloalkyloxy group, and a heterocyclic group;
  • n 0, 1, 2, 3 or 4;
  • n 0, 1 or 2;
  • s 0, 1, 2, 3 or 4.
  • the compound of the formula (AI) is a compound of the formula (I):
  • L 1 is an alkylene group, wherein the alkylene group is optionally further substituted with one or more substituents of a halogen or a halogen atom;
  • R 6 is selected from the group consisting of a hydrogen atom, a halogen atom, a halogen, an alkyl group, an alkoxy group, an amino group, a nitro group, a cyano group, a hydroxyl group, a hydroxyalkyl group, a cycloalkyl group, a cycloalkyloxy group, a heterocyclic group, an aryl group, and a heteroaryl group, wherein said cycloalkyl group, heterocyclic group, aryl group and heteroaryl group are further optionally selected from the group consisting of a halogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, an amino group, a nitro group, and a cyano group.
  • R 12 is selected from the group consisting of a hydrogen atom, an alkyl group, a halogenated alkyl group, an amino group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group;
  • R 13 is an aryl or heteroaryl group, wherein the aryl and heteroaryl are optionally further selected from the group consisting of a hydrogen atom, a halogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, an amino group, a nitro group, and a cyano group. Substituted by one or more substituents of a hydroxy group, a hydroxyalkyl group, a cycloalkyl group, a cycloalkyloxy group, and a heterocyclic group;
  • Ring A, R 1 to R 3 , R 5 , n and s are as defined in the formula (AI).
  • the compound of the formula (AI) is a compound of the formula (Iaa):
  • Rings A, L 1 , R 1 to R 3 , R 5 to R 6 , n and s are as defined in the formula (I).
  • R 5 and s are as defined in the formula (AI).
  • the compound of the formula (AI) is a compound of the formula (II):
  • R 7 is selected from the group consisting of alkyl, cycloalkyl and haloalkyl, wherein said alkyl or cycloalkyl is optionally further substituted with one or more substituents selected from the group consisting of a halogen atom, a halogen, an alkyl group and a cycloalkyl group.
  • L 1 , R 2 , R 3 , R 5 , R 6 and n are as defined in the formula (I).
  • the compound of the formula (AI) wherein R 5 is the same or different and each independently selected from alkyl, alkoxy, oxo, halogen, haloalkane Base, cyano, hydroxy, -C(O)OR 8 , -NHC(O)OR 8 , -C(O)NR 10 R 11 , -CH 2 NHC(O)OR 8 , -CH 2 NR 10 R 11 , -C(O)OCH(R 10 )R 11 and -S(O) m R 8 ;
  • R 8 is selected from the group consisting of a hydrogen atom, an alkyl group, a hydroxyl group, and an amino group;
  • R 10 and R 11 are the same or different and are each independently Selected from a hydrogen atom, an alkyl group, a halogenated alkyl group, a cycloalkyl group, a heterocyclic group, -C(O)OR 8 and -OC(O)OR
  • the compound of the formula (AI) is a compound of the formula (III):
  • R 5 is the same or different and is each independently selected from the group consisting of alkyl, alkoxy, oxo, halogen, haloalkyl, cyano, hydroxy, -C(O)OR 8 , -NHC(O)OR 8 ,- C(O)NR 10 R 11 , -CH 2 NHC(O)OR 8 , -CH 2 NR 10 R 11 , -C(O)OCH(R 10 )R 11 and -S(O) m R 8 ;
  • R 8 is selected from the group consisting of a hydrogen atom, an alkyl group, a hydroxyl group, and an amino group; and
  • R 10 and R 11 are the same or different and are each independently selected from a hydrogen atom, an alkyl group, a halogenated alkyl group, a cycloalkyl group, a heterocyclic group, and -C(O).
  • cycloalkyl and heterocyclic groups are optionally further selected from the group consisting of a halogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, an oxo group, an amino group, a nitrate Substituted by one or more substituents of a cyano group, a cyano group, a hydroxy group and a hydroxyalkyl group; R 12 is an alkyl group;
  • R 7 is selected from the group consisting of alkyl, cycloalkyl and haloalkyl, wherein said alkyl or cycloalkyl is optionally further substituted with one or more substituents selected from the group consisting of a halogen atom, a halogen, an alkyl group and a cycloalkyl group.
  • L 1 , R 2 , R 3 , R 6 and n are as defined in the formula (I).
  • the compound of the formula (AI) is a compound of the formula (IV):
  • L 1 , R 2 , R 3 , R 6 , R 7 and n are as defined in the formula (III).
  • R 14 is a hydrogen atom or a halogen atom;
  • R 6 is selected from the group consisting of a hydrogen atom, a halogen atom, a halogen, an alkyl group, an alkoxy group, a cycloalkyl group, a cycloalkyloxy group, a heterocyclic group, an aryl group, and a heteroaryl group; wherein the cycloalkyl group is And a heterocyclic group, an aryl group and a heteroaryl group, optionally further selected from the group consisting of a halogen atom, a halogen, an alkyl group, an alkoxy group, a cycloalkyl group, a heterocyclic group, -NHC(O)R 12 and R 13 Or substituted with a plurality of substituent
  • a heterocyclic group, an aryl group and a heteroaryl group wherein the cycloalkyl group, heterocyclic group, aryl group and heteroaryl group are further selected from a halogen atom, a halogen, an alkyl group, an alkoxy group, a cycloalkane Substituted with one or more substituents of -NHC(O)R 12 and R 13 ;
  • R 12 is alkyl or cycloalkyl;
  • R 13 is aryl or heteroaryl, wherein The aryl and heteroaryl groups are optionally further substituted with one or more substituents selected from the group consisting of a halogen atom, a halogen, an alkyl group, an alkoxy group, a halogenated alkyl group, an amino group, a nitro group, a cyano group, and a hydroxyl group.
  • Typical compounds of the general formula (AI) include, but are not limited to:
  • a tautomer a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a prodrug thereof.
  • a method of preparing a compound of the formula (AI), the method comprising:
  • a compound of the formula (AI-A) and a compound of the formula (AI-B) or a hydrochloride thereof are subjected to a condensation reaction under basic conditions, optionally under basic conditions, to give a compound of the formula (AI). ;
  • Ring A, R 1 to R 5 , n and s are as defined in the formula (AI).
  • a compound of the formula (I-A) and a compound of the formula (AI-B) or a hydrochloride thereof are subjected to a condensation reaction under basic conditions, optionally under basic conditions, to give a compound of the formula (I);
  • Rings A, L 1 , R 1 to R 3 , R 5 to R 6 , n and s are as defined in the formula (I).
  • Another aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of the formula (AI) or a tautomer thereof, a mesogen, a racemate, an enantiomer a form, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent and excipient.
  • Another aspect of the invention relates to a compound of the formula (AI) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof Or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, which is used as a medicament.
  • Another aspect of the invention relates to a compound of the formula (AI) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof Or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same, for use in the manufacture of a medicament for inhibiting factor XIa.
  • Another aspect of the invention relates to a compound of the formula (AI) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof Or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, which is used as a XIa inhibitor.
  • Another aspect of the invention relates to a compound of the formula (AI) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof Or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same, for use in the manufacture of a medicament for preventing and/or treating a XIa factor mediated disease.
  • Another aspect of the invention relates to a compound of the formula (AI) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof Or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for use in the preparation of a prophylactic and/or therapeutic cardio-cerebral vascular disease, wherein the cardiovascular disease is preferably a thromboembolic disease, more preferably a myocardial infarction Re-occlusion and restenosis after angina pectoris, angioplasty or aortic coronary shunt, disseminated intravascular coagulation, stroke, transient ischemic attack, peripheral arterial occlusive disease, pulmonary embolism or deep vein thrombosis.
  • the cardiovascular disease is preferably a thromboembolic disease, more preferably a myocardial infarction Re-occlusion and restenosis after angina pectoris, angioplasty or
  • Another aspect of the invention relates to a method of preventing and/or treating a XIa factor mediated disease comprising administering to a patient a therapeutically effective amount of a compound of the formula (AI) or a tautomer thereof, A racemate, a racemate, an enantiomer, a diastereomer or a mixture thereof or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same.
  • a compound of the formula (AI) or a tautomer thereof A racemate, a racemate, an enantiomer, a diastereomer or a mixture thereof or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same.
  • Another aspect of the invention relates to a method for the treatment of prevention and/or treatment of cardiovascular and cerebrovascular diseases, comprising administering to a patient a therapeutically effective amount of a compound of the formula (AI) or a tautomer thereof, internal elimination a rotator, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, wherein the cardiovascular and cerebrovascular disease is selected from the group consisting of myocardial infarction Re-obstruction and restenosis, stroke, transient ischemic attack, peripheral arterial occlusive disease, pulmonary embolism, or deep vein thrombosis after angina pectoris, angioplasty, or aortic coronary shunt.
  • a compound of the formula (AI) or a tautomer thereof internal elimination a rotator, a racemate, an enantiomer, a diastereomer or a mixture thereof,
  • Another aspect of the invention relates to a medicament for inhibiting factor XIa, which comprises a compound of the formula (AI) or a tautomer thereof, a mesogen, a racemate, an enantiomer a form, a diastereomer or a mixture thereof or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same.
  • a compound of the formula (AI) or a tautomer thereof a mesogen, a racemate, an enantiomer a form, a diastereomer or a mixture thereof or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same.
  • the active ingredient-containing pharmaceutical composition may be in a form suitable for oral administration, such as tablets, dragees, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or Tincture.
  • Oral compositions can be prepared according to any method known in the art for preparing pharmaceutical compositions, such compositions may contain one or more ingredients selected from the group consisting of sweeteners, flavoring agents, coloring agents, and preservatives, To provide a pleasing and tasty pharmaceutical preparation. Tablet containing the active ingredient and a non-toxic pharmaceutically acceptable tablet for mixing Excipients used.
  • excipients can be inert excipients, granulating agents, disintegrating agents, binders, and lubricants. These tablets may be uncoated or may be coated by masking the taste of the drug or delaying disintegration and absorption in the gastrointestinal tract, thus providing a sustained release effect over a longer period of time.
  • Oral formulations can also be provided in soft gelatine capsules in which the active ingredient is mixed with an inert solid diluent or the active ingredient in admixture with a water-soluble vehicle or an oil vehicle.
  • the aqueous suspension contains the active substance and excipients suitable for the preparation of the aqueous suspension for mixing. Such excipients are suspending, dispersing or wetting agents.
  • the aqueous suspensions may also contain one or more preservatives, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents.
  • the oil suspension can be formulated by suspending the active ingredient in vegetable oil, or mineral oil.
  • the oil suspension may contain a thickening agent.
  • the above sweeteners and flavoring agents may be added to provide a palatable preparation. These compositions can be preserved by the addition of an antioxidant.
  • the dispersible powders and granules suitable for the preparation of aqueous suspensions can be provided by the addition of water to provide the active ingredient and dispersing or wetting agents, suspending agents or one or more preservatives. Suitable dispersing or wetting agents and suspending agents can be used to illustrate the above examples. Other excipients such as sweetening, flavoring, and coloring agents may also be added. These compositions are preserved by the addition of an anti-oxidant such as ascorbic acid.
  • compositions of the invention may also be in the form of an oil-in-water emulsion.
  • the oil phase can be a vegetable oil, or a mineral oil or a mixture thereof.
  • Suitable emulsifiers can be naturally occurring phospholipids, and emulsions can also contain sweeteners, flavoring agents, preservatives, and antioxidants.
  • Such formulations may also contain a demulcent, a preservative, a colorant, and an antioxidant.
  • the pharmaceutical compositions of the invention may be in the form of a sterile injectable aqueous solution.
  • acceptable vehicles or solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • the sterile injectable preparation may be a sterile injectable oil-in-water microemulsion in which the active ingredient is dissolved in the oily phase.
  • the injection or microemulsion is injected into the bloodstream of the patient by topical injection.
  • the solution and microemulsion are preferably administered in a manner that maintains a constant circulating concentration of the compound of the invention.
  • a continuous intravenous delivery device can be used.
  • An example of such a device is the Deltec CADD-PLUS.TM.5400 intravenous pump.
  • compositions of the invention may be in the form of a sterile injectable aqueous or oily suspension for intramuscular and subcutaneous administration.
  • the suspension may be formulated according to known techniques using those suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension prepared in a parenterally acceptable non-toxic diluent or solvent.
  • sterile fixed oils may conveniently be employed as a solvent or suspension medium. Any blended fixed oil can be used for this purpose.
  • fatty acids can also be prepared as injections.
  • the compounds of the invention may be administered in the form of a suppository for rectal administration.
  • These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid in the rectum and thus dissolves in the rectum to release the drug.
  • the dosage of the drug to be administered depends on a variety of factors including, but not limited to, the following factors: the activity of the particular compound used, the age of the patient, the weight of the patient, the health of the patient, the behavior of the patient. , patient's diet, time of administration, mode of administration, rate of excretion, drug Combinations and the like; in addition, the optimal treatment mode such as the mode of treatment, the daily dose of the compound of the formula (I) or the kind of the pharmaceutically acceptable salt can be verified according to a conventional treatment regimen.
  • alkyl refers to a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing from 1 to 20 carbon atoms, preferably an alkyl group having from 1 to 12 carbon atoms, more preferably from 1 to 6 carbons.
  • the alkyl group of the atom is a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing from 1 to 20 carbon atoms, preferably an alkyl group having from 1 to 12 carbon atoms, more preferably from 1 to 6 carbons.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1 ,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2- Methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3 - dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2 -methylhexyl, 3-methylhexyl, 4-methylhexyl,
  • lower alkyl groups having from 1 to 6 carbon atoms, non-limiting examples including methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl Base, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethyl Butyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl Base, 2,3-dimethylbutyl and the like.
  • the alkyl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, preferably one or more of the following groups, independently selected from hydrazine.
  • alkylene refers to a saturated straight or branched aliphatic hydrocarbon radical having two residues derived from the removal of two hydrogen atoms from the same carbon atom of the parent alkane or two different carbon atoms.
  • Non-limiting examples of alkylene include, but are not limited to, methylene (-CH 2 -), 1,1-ethylene (-CH(CH 3 )-), 1,2-ethylene (-CH 2 ) CH 2 )-, 1,1-propylene (-CH(CH 2 CH 3 )-), 1,2-propylene (-CH 2 CH(CH 3 )-), 1,3-propylene (-CH 2 CH 2 CH 2 -), 1,4-butylene (-CH 2 CH 2 CH 2 CH 2 -), and the like.
  • the alkylene group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, which is preferably independently independently selected from the group consisting of a ruthenium atom, an alkyl group, and an alkenyl group.
  • cycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, -C(O)R 8 , -C(O)OR 8 and -S(O) m R 8 Substituted by a substituent.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 carbon atoms, preferably from 3 to 12 carbon atoms, more preferably from 3 to 8 carbon atoms. One carbon atom, most preferably containing from 3 to 5 carbon atoms.
  • Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene
  • a cycloalkyl group or the like is preferably a cycloalkyl group;
  • a polycyclic cycloalkyl group includes a spiro ring, a fused ring, and a bridged cycloalkyl group.
  • spirocycloalkyl refers to a polycyclic group that shares a carbon atom (referred to as a spiro atom) between 5 to 20 members of a single ring, which may contain one or more double bonds, but none of the rings have a fully conjugated ⁇ electronic system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • the spirocycloalkyl group is classified into a monospirocycloalkyl group, a bispirocycloalkyl group or a polyspirocycloalkyl group, preferably a monospirocycloalkyl group and a bispirocycloalkyl group, depending on the number of common spiro atoms between the rings.
  • spirocycloalkyl groups include:
  • fused cycloalkyl refers to 5 to 20 members, and each ring in the system shares an all-carbon polycyclic group of an adjacent pair of carbon atoms with other rings in the system, wherein one or more of the rings may contain one or Multiple double bonds, but none of the rings have a fully conjugated ⁇ -electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • fused cycloalkyl groups include:
  • bridged cycloalkyl refers to an all-carbon polycyclic group of 5 to 20 members, any two rings sharing two carbon atoms which are not directly bonded, which may contain one or more double bonds, but none of the rings have complete Conjugate ⁇ -electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members. Depending on the number of constituent rings, it may be classified into a bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl group, preferably a bicyclic ring, a tricyclic ring or a tetracyclic ring, and more preferably a bicyclic ring or a tricyclic ring.
  • bridged cycloalkyl groups include:
  • the cycloalkyl ring may be fused to an aryl, heteroaryl or heterocycloalkyl ring, wherein the ring to which the parent structure is attached is a cycloalkyl group, non-limiting examples include indanyl, tetrahydronaphthalene A benzocycloheptyl group or the like; preferably a phenylcyclopentyl group or a tetrahydronaphthyl group.
  • the cycloalkyl group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio Substituted by one or more substituents of a heterocycloalkylthio group, an oxo group, -C(O)R 8 , -C(O)OR 8 and -S(O) m R 8 .
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 ring atoms wherein one or more ring atoms are selected from nitrogen, oxygen or S(O).
  • a hetero atom of m (where m is an integer of 0 to 2), but excluding the ring moiety of -OO-, -OS- or -SS-, the remaining ring atoms being carbon.
  • Non-limiting examples of monocyclic heterocyclic groups include pyrrolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiophenyl, dihydroimidazolyl, dihydrofuranyl, m-dioxole , dihydropyrazolyl, dihydropyrrolyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl and the like.
  • Polycyclic heterocyclic groups include spiro, fused, and bridged heterocyclic groups.
  • spiroheterocyclyl refers to a polycyclic heterocyclic group in which one atom (called a spiro atom) is shared between 5 to 20 members of a single ring, wherein one or more ring atoms are selected from nitrogen, oxygen or S (O). ) m (where m is an integer 0 to 2) heteroatoms, and the remaining ring atoms are carbon. It may contain one or more double bonds, but none of the rings have a fully conjugated pi-electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • the spiroheterocyclyl group is classified into a monospiroheterocyclic group, a dispiroheterocyclic group or a polyspirocyclic group according to the number of shared spiro atoms between the ring and the ring, and is preferably a monospiroheterocyclic group and a dispiroheterocyclic group. More preferably, it is 4 yuan / 4 yuan, 4 yuan / 5 yuan, 4 yuan / 6 yuan, 5 yuan / 5 yuan or 5 yuan / 6-membered monospiroheterocyclic group.
  • Non-limiting examples of spiroheterocyclyl groups include:
  • fused heterocyclyl refers to 5 to 20 members, and each ring in the system shares an adjacent pair of atomic polycyclic heterocyclic groups with other rings in the system, and one or more rings may contain one or more Double bond, but none of the rings have a fully conjugated ⁇ -electron system in which one or more ring atoms are heteroatoms selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), and the remaining rings
  • the atom is carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • fused heterocyclic groups include:
  • bridge heterocyclyl refers to a polycyclic heterocyclic group of 5 to 14 members, any two rings sharing two atoms which are not directly bonded, which may contain one or more double bonds, but none of the rings have a total A ⁇ -electron system of a yoke in which one or more ring atoms are heteroatoms selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), the remaining ring atoms being carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • bridge heterocyclic groups include:
  • the heterocyclyl ring may be fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring to which the parent structure is attached is a heterocyclic group, non-limiting examples of which include:
  • the heterocyclic group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio Substituted by one or more substituents of a heterocycloalkylthio group, an oxo group, -C(O)R 8 , -C(O)OR 8 and -S(O) m R 8 .
  • aryl refers to a 6 to 20 membered all-carbon monocyclic or fused polycyclic ring (i.e., a ring sharing a pair of adjacent carbon atoms) having a conjugated ⁇ -electron system, preferably 6 to 10 members, more preferably 6 yuan, such as phenyl and naphthyl.
  • the aryl ring may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring to which the parent structure is attached is an aryl ring, non-limiting examples of which include:
  • the aryl group may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of a halogen atom, an alkyl group, an alkenyl group, an alkynyl group, an alkoxy group, and an alkane group.
  • heteroaryl refers to a heteroaromatic system containing from 1 to 4 heteroatoms, from 5 to 20 ring atoms, wherein the heteroatoms are selected from the group consisting of oxygen, sulfur and nitrogen.
  • the heteroaryl group is preferably 5 to 10 members, and has 1 to 3 hetero atoms; more preferably 5 or 6 members, and 1 to 2 hetero atoms; preferably, for example, imidazolyl, furyl, thienyl, thiazolyl, pyridyl Azolyl, oxazolyl, pyrrolyl, triazolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, pyrazinyl and the like.
  • the heteroaryl ring may be fused to an aryl, heterocyclic or cycloalkyl ring, wherein the ring to which the parent structure is attached is a heteroaryl ring, non-limiting examples of which include:
  • the heteroaryl group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio Substituted by one or more substituents of a heterocycloalkylthio group, -C(O)R 8 , -C(O)OR 8 and -S(O) m R 8 .
  • alkoxy refers to -O-(alkyl) and -O-(unsubstituted cycloalkyl), wherein alkyl is as defined above.
  • alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy.
  • the alkoxy group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio Substituted by one or more substituents of a heterocycloalkylthio group, -C(O)R 8 , -C(O)OR 8 and -S(O) m R 8 .
  • alkylthio refers to -S-(alkyl) and -S-(unsubstituted cycloalkyl), wherein alkyl is as defined above.
  • alkylthio groups include: methylthio, ethylthio, propylthio, butylthio, cyclopropylthio, cyclobutylthio, cyclopentylthio, cyclohexylthio.
  • the alkylthio group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane.
  • cycloalkyloxy refers to -O-cycloalkyl, wherein cycloalkyl is as defined above.
  • haloalkyl refers to an alkyl group substituted by a halogen wherein alkyl is as defined above.
  • haloalkoxy refers to an alkoxy group substituted by a halogen wherein alkoxy is as defined above.
  • hydroxyalkyl refers to an alkyl group substituted with a hydroxy group, wherein alkyl is as defined above.
  • hydroxy refers to an -OH group.
  • halogen means fluoro, chloro, bromo or iodo.
  • amino means -NH 2.
  • cyano refers to -CN.
  • nitro refers to -NO 2 .
  • carboxylate group refers to -C(O)O(alkyl) or -C(O)O(cycloalkyl), wherein alkyl, cycloalkyl are as defined above.
  • heterocyclic group optionally substituted by an alkyl group means that an alkyl group may be, but not necessarily, present, and the description includes the case where the heterocyclic group is substituted with an alkyl group and the case where the heterocyclic group is not substituted with an alkyl group.
  • Substituted refers to one or more hydrogen atoms in the group, preferably up to 5, more preferably 1 to 3, hydrogen atoms, independently of each other, substituted by a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and those skilled in the art will be able to determine without much effort (by experimentation). Or theory) may or may not be replaced. For example, an amino group or a hydroxyl group having a free hydrogen may be unstable when combined with a carbon atom having an unsaturated (e.g., olefinic) bond.
  • “Pharmaceutical composition” means a mixture comprising one or more of the compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, as well as other components such as physiological/pharmaceutically acceptable carriers. And excipients.
  • the purpose of the pharmaceutical composition is to promote the administration of the organism, which facilitates the absorption of the active ingredient and thereby exerts biological activity.
  • “Pharmaceutically acceptable salt” refers to a salt of a compound of the invention which is safe and effective for use in a mammal and which possesses the desired biological activity.
  • R 8 and m are as defined by the formula (AI).
  • the first step is a reaction of a compound of the formula (AI-1) with a compound of the formula (AI-2) under basic conditions in an organic solvent to obtain a compound of the formula (AI-3); or a formula (AI) -1) the compound is subjected to nucleophilic substitution reaction with a general formula (AI-2') in an organic solvent under basic conditions to obtain a compound of the formula (AI-A);
  • the second step reaction is a compound of the general formula (AI-3), which is hydrolyzed under acidic conditions to obtain a formula (AI-A);
  • the compound of the formula (AI-A) and the compound of the formula (AI-B) or a hydrochloride thereof are subjected to a condensation reaction under basic conditions, optionally under basic conditions, to obtain a pass.
  • a compound of formula (AI) is subjected to a condensation reaction under basic conditions, optionally under basic conditions, to obtain a pass.
  • the reagents providing basic conditions include organic bases including, but not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, and inorganic bases. , bis-trimethylsilylamino lithium, potassium acetate, sodium t-butoxide or potassium t-butoxide, said inorganic bases including but not limited to sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, cesium carbonate, hydrogen Sodium oxide and lithium hydroxide.
  • Conditions which provide acidity include, but are not limited to, pyridine hydrobromide, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid or methanesulfonic acid, preferably pyridine hydrobromide or hydrochloric acid.
  • the condensing agent includes, but is not limited to, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, N,N'-dicyclohexylcarbodiimide, N,N'- Diisopropylcarbodiimide, O-benzotriazole-N,N,N',N'-tetramethyluronium tetrafluoroborate, 1-hydroxybenzotriazole, 1-hydroxy- 7-azobenzotriazole, O-benzotriazole-N,N,N',N'-tetramethyluron hexafluorophosphate, 2-(7-oxobenzotriazole)-N ,N,N',N'-tetramethylurea hexafluorophosphate, 2-(7-azobenzotriazole)-N,N,N',N'-tetramethyluron hexafluorophosphate , benzotriazol-1-
  • the above reaction is preferably carried out in a solvent including, but not limited to, acetic acid, methanol, ethanol, toluene, tetrahydrofuran, dichloromethane, dimethyl sulfoxide, 1,4-dioxane, water or N,N- dimethylformamide.
  • a solvent including, but not limited to, acetic acid, methanol, ethanol, toluene, tetrahydrofuran, dichloromethane, dimethyl sulfoxide, 1,4-dioxane, water or N,N- dimethylformamide.
  • X is a halogen, preferably bromine
  • R a is an alkyl group, preferably a methyl group
  • Ring A, R 1 to R 5 , n and s are as defined in the formula (AI).
  • the first step is a compound of the formula (I-1) which is subjected to nucleophilic substitution reaction with (I-2) under basic conditions in an organic solvent to obtain a compound of the formula (I-3); or a compound of the formula (I-1).
  • the nucleophilic substitution reaction with the general formula (I-2') under basic conditions in an organic solvent gives a compound of the formula (I-A);
  • the second step reaction is a compound of the formula (I-3), which is hydrolyzed under acidic conditions to obtain a formula (I-A);
  • the compound of the formula (IA) and the compound of (AI-B) or a hydrochloride thereof are subjected to a condensation reaction under basic conditions, optionally under basic conditions, to give a compound of the formula (I). ;
  • the reagents providing basic conditions include organic bases including, but not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, and inorganic bases. , bis-trimethylsilylamino lithium, potassium acetate, sodium t-butoxide or potassium t-butoxide, said inorganic bases including but not limited to sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, cesium carbonate, hydrogen Sodium oxide and lithium hydroxide.
  • Conditions which provide acidity include, but are not limited to, pyridine hydrobromide, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid or methanesulfonic acid, preferably pyridine hydrobromide or hydrochloric acid.
  • Condensing agents include, but are not limited to, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, N,N'-dicyclohexylcarbodiimide, N,N'-di Propylcarbodiimide, O-benzotriazole-N,N,N',N'-tetramethyluronium tetrafluoroborate, 1-hydroxybenzotriazole, 1-hydroxy-7- Azobenzotriazole, O-benzotriazole-N,N,N',N'-tetramethyluron hexafluorophosphate, 2-(7-oxobenzotriazole)-N,N , N', N'-tetramethylurea hexafluorophosphate, 2-(7-azobenzotriazole)-N,N,N',N'-tetramethylurea hexafluorophosphate, benzene And triazol-1-
  • the above reaction is preferably carried out in a solvent including, but not limited to, acetic acid, methanol, ethanol, toluene, tetrahydrofuran, dichloromethane, dimethyl sulfoxide, 1,4-dioxane, water or N,N- dimethylformamide.
  • a solvent including, but not limited to, acetic acid, methanol, ethanol, toluene, tetrahydrofuran, dichloromethane, dimethyl sulfoxide, 1,4-dioxane, water or N,N- dimethylformamide.
  • X is a halogen, preferably bromine
  • R a is an alkyl group, preferably a methyl group
  • Rings A, L 1 , R 1 to R 3 , R 5 to R 6 , n and s are as defined in the formula (I).
  • the first step of the reaction is a compound of the formula (I-1-a) in an organic solvent, and nucleophilic substitution reaction with (I-2-a) under basic conditions to obtain a compound of the formula (I-3);
  • the second step reaction is a compound of the formula (I-3), which is hydrolyzed under acidic conditions to obtain a formula (I-A);
  • the compound of the formula (IA) and the compound of (AI-B) or a hydrochloride thereof are subjected to a condensation reaction under basic conditions, optionally under basic conditions, to give a compound of the formula (I). ;
  • the reagents providing basic conditions include organic bases including, but not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, and inorganic bases. , bis-trimethylsilylamino lithium, potassium acetate, sodium t-butoxide or potassium t-butoxide, said inorganic bases including but not limited to sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, cesium carbonate, hydrogen Sodium oxide and lithium hydroxide.
  • Conditions which provide acidity include, but are not limited to, pyridine hydrobromide, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid or methanesulfonic acid, preferably pyridine hydrobromide or hydrochloric acid.
  • Condensing agents include, but are not limited to, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, N,N'-dicyclohexylcarbodiimide, N,N'-di Propylcarbodiimide, O-benzotriazole-N,N,N',N'-tetramethyluronium tetrafluoroborate, 1-hydroxybenzotriazole, 1-hydroxy-7- Azobenzotriazole, O-benzotriazole-N,N,N',N'-tetramethyluron hexafluorophosphate, 2-(7-oxobenzotriazole)-N,N , N', N'-tetramethylurea hexafluorophosphate, 2-(7-azobenzotriazole)-N,N,N',N'-tetramethylurea hexafluorophosphate, benzene Triazol-1-y
  • the above reaction is preferably carried out in a solvent including, but not limited to, acetic acid, methanol, ethanol, toluene, tetrahydrofuran, dichloromethane, dimethyl sulfoxide, 1,4-dioxane, water or N,N- dimethylformamide.
  • a solvent including, but not limited to, acetic acid, methanol, ethanol, toluene, tetrahydrofuran, dichloromethane, dimethyl sulfoxide, 1,4-dioxane, water or N,N- dimethylformamide.
  • R a is an alkyl group, preferably a methyl group
  • R b is a leaving group, preferably methanesulfonyloxy or trifluoromethanesulfonyloxy;
  • Rings A, L 1 , R 1 to R 3 , R 5 to R 6 , n and s are as defined in the formula (I).
  • the first step is a compound of the formula (II-1) which is subjected to nucleophilic substitution reaction with (I-2) under basic conditions in an organic solvent to obtain a compound of the formula (II-2); or a compound of the formula (II-1).
  • the nucleophilic substitution reaction with the general formula (I-2') under basic conditions in an organic solvent gives a compound of the formula (II-A);
  • the second step is a reaction of the compound of the formula (II-2) under acidic conditions to obtain the formula (II-A);
  • the compound of the formula (II-A) and the compound of the formula (II-B) or a hydrochloride thereof are subjected to a condensation reaction under basic conditions, optionally under basic conditions, to obtain a formula (II).
  • a condensation reaction under basic conditions, optionally under basic conditions, to obtain a formula (II).
  • the reagents providing basic conditions include organic bases including, but not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, and inorganic bases. , bis-trimethylsilylamino lithium, potassium acetate, sodium t-butoxide or potassium t-butoxide, preferably lithium bis trimethylsilylamine, said inorganic base package
  • organic bases including, but not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, and inorganic bases.
  • bis-trimethylsilylamino lithium, potassium acetate, sodium t-butoxide or potassium t-butoxide preferably lithium bis trimethylsilylamine
  • said inorganic base package include, but are not limited to, sodium hydride, potassium phosphate, sodium carbonate, potassium carbonate, cesium carbon
  • Conditions which provide acidity include, but are not limited to, pyridine hydrobromide, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid or methanesulfonic acid, preferably pyridine hydrobromide or hydrochloric acid.
  • Condensing agents include, but are not limited to, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, N,N'-dicyclohexylcarbodiimide, N,N'-di Propylcarbodiimide, O-benzotriazole-N,N,N',N'-tetramethyluronium tetrafluoroborate, 1-hydroxybenzotriazole, 1-hydroxy-7- Azobenzotriazole, O-benzotriazole-N,N,N',N'-tetramethyluron hexafluorophosphate, 2-(7-oxobenzotriazole)-N,N , N', N'-tetramethylurea hexafluorophosphate, 2-(7-azobenzotriazole)-N,N,N',N'-tetramethylurea hexafluorophosphate, benzene And triazol-1-
  • the above reaction is preferably carried out in a solvent including, but not limited to, acetic acid, methanol, ethanol, toluene, tetrahydrofuran, dichloromethane, dimethyl sulfoxide, 1,4-dioxane, water or N,N- dimethylformamide.
  • a solvent including, but not limited to, acetic acid, methanol, ethanol, toluene, tetrahydrofuran, dichloromethane, dimethyl sulfoxide, 1,4-dioxane, water or N,N- dimethylformamide.
  • X is a halogen, preferably bromine
  • R a is an alkyl group, preferably a methyl group
  • L 1 , R 2 , R 3 , R 5 to R 7 and n are as defined in the formula (II);
  • the first step is a compound of the formula (II-A) and a compound of the formula (IV-1) or a hydrochloride thereof is subjected to a condensation reaction under basic conditions to obtain a compound of the formula (IV-2);
  • the second step is a reaction of the compound of the formula (IV-2) under acidic conditions to give a compound of the formula (IV-3);
  • the third step is a chiral preparation of a compound of formula (IV-3) to give a compound of formula (IV);
  • the reagents providing basic conditions include organic bases including, but not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, and inorganic bases. , bis-trimethylsilylamino lithium, potassium acetate, sodium t-butoxide or potassium t-butoxide, preferably lithium bis trimethylsilylamine, said inorganic bases including but not limited to sodium hydride, phosphoric acid Potassium, sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide or lithium hydroxide is preferably potassium carbonate, sodium hydride or lithium hydroxide.
  • Conditions which provide acidity include, but are not limited to, pyridine hydrobromide, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid or methanesulfonic acid, preferably pyridine hydrobromide or hydrochloric acid.
  • Conditions for chiral preparation include, but are not limited to, the column being Superchiral S-AD (Chiralway), the mobile phase being carbon dioxide, ethanol and diethylamine.
  • Condensing agents include, but are not limited to, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, N,N'-dicyclohexylcarbodiimide, N,N'-di Propylcarbodiimide, O-benzotriazole-N,N,N',N'-tetramethyluronium tetrafluoroborate, 1-hydroxybenzotriazole, 1-hydroxy-7- Azobenzotriazole, O-benzotriazole-N,N,N',N'-tetramethyluron hexafluorophosphate, 2-(7-oxobenzotriazole)-N,N , N', N'-tetramethylurea hexafluorophosphate, 2-(7-azobenzotriazole)-N,N,N',N'-tetramethylurea hexafluorophosphate, benzene And triazol-1-
  • the above reaction is preferably carried out in a solvent including, but not limited to, acetic acid, methanol, ethanol, toluene, tetrahydrofuran, dichloromethane, dimethyl sulfoxide, 1,4-dioxane, water or N,N- dimethylformamide.
  • a solvent including, but not limited to, acetic acid, methanol, ethanol, toluene, tetrahydrofuran, dichloromethane, dimethyl sulfoxide, 1,4-dioxane, water or N,N- dimethylformamide.
  • R c is an alkyl group, preferably a methyl group
  • L 1 , R 2 , R 3 , R 6 , R 7 and n are as defined in the formula (IV).
  • the compounds of the general formula involved in the present invention such as the form of the compound salt obtained during the synthesis, can be further obtained by conventional experimental means; if the free compound form is obtained during the synthesis, the compound can be further obtained by conventional experimental means. Salt.
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) or/and mass spectrometry (MS).
  • NMR shift ( ⁇ ) is given in units of 10 -6 (ppm).
  • NMR was measured using a Bruker AVANCE-400 nuclear magnetic apparatus, and the solvent was deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD), internal standard was four.
  • DMSO-d 6 dimethyl sulfoxide
  • CDCl 3 deuterated chloroform
  • CD 3 OD deuterated methanol
  • TMS Methyl silane
  • the measurement of the MS was carried out using a FINNIGAN LCQAd (ESI) mass spectrometer (manufacturer: Thermo, model: Finnigan LCQ advantage MAX).
  • ESI FINNIGAN LCQAd
  • HPLC High performance liquid chromatography
  • Chiral HPLC analysis was performed using an Agilent 1260 DAD high performance liquid chromatograph.
  • the CombiFlash Rapid Preparer uses the Combiflash Rf200 (TELEDYNE ISCO).
  • Thin layer chromatography silica gel plate uses Yantai Yellow Sea HSGF254 or Qingdao GF254 silica gel plate.
  • the specification of silica gel plate used for thin layer chromatography (TLC) is 0.15mm ⁇ 0.2mm.
  • the specification for thin layer chromatography separation and purification is 0.4mm. ⁇ 0.5mm.
  • Silica gel column chromatography generally uses Yantai Huanghai silica gel 200-300 mesh silica gel as a carrier.
  • the known starting materials of the present invention may be synthesized by or according to methods known in the art, or may be purchased from ABCR GmbH & Co. KG, Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc, Companies such as Dare Chemicals.
  • the reactions can all be carried out under an argon atmosphere or a nitrogen atmosphere.
  • An argon atmosphere or a nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon having a volume of about 1 L.
  • the hydrogen atmosphere means that the reaction flask is connected to a hydrogen balloon of about 1 L volume.
  • the pressurized hydrogenation reaction was carried out using a Parr Model 3916EKX hydrogenation apparatus and a clear blue QL-500 type hydrogen generator or a HC2-SS type hydrogenation apparatus.
  • the hydrogenation reaction is usually evacuated, charged with hydrogen, and operated three times.
  • the microwave reaction used a CEM Discover-S Model 908860 microwave reactor.
  • the solution means an aqueous solution.
  • reaction temperature is room temperature and is 20 ° C to 30 ° C.
  • TLC thin layer chromatography
  • the developing agent used for the reaction the column chromatography eluent system used for the purification of the compound
  • the thin layer chromatography developing solvent system including: A: Methylene chloride/methanol system, B: n-hexane/ethyl acetate system, C: petroleum ether/ethyl acetate system, D: acetone, E: dichloromethane/acetone system, F: ethyl acetate/dichloromethane system , G: ethyl acetate / dichloromethane / n-hexane, H: ethyl acetate / dichloromethane / acetone, I: petroleum ether / ethyl acetate / dichloromethane, the volume ratio of the solvent is different depending on the polarity of the compound Adjustment can also be carried out by adding a small amount of an alkaline or acidic acidic acidic acidic acidic acidic acid
  • Methyl 4-methoxybutyrate 1a (1.6 g, 12.1 mmol) was added to 50 mL of tetrahydrofuran, cooled to -78 ° C with a dry ice-acetone bath, and di-trimethylsilylamine lithium (12.7 mL, 12.7 mmol), after the addition was completed, the reaction was stirred for 1 hour, trimethylchlorosilane (1.31 g, 12.1 mmol) was added, and the reaction was further stirred for 20 minutes, and then N-bromosuccinimide (2.15 g, 12.1 mmol) was added. The reaction was stirred for 2 hours.
  • reaction solution was naturally cooled to room temperature, and 50 mL of water was added to the reaction mixture, and the mixture was combined with ethyl acetate (50 mL ⁇ 3), and the organic phase was combined, and the organic phase was washed with water (50 mL) The organic layer was dried (MgSO4), evaporated, evaporated,
  • 2-Bromo-4-chloro-1-iodobenzene 8a (1.0 g, 3.15 mmol, prepared by a known method "Angewandte Chemie, International Edition, 2010, 49 (46), 8729-8732”) was dissolved in 1 mL The mixture was cooled to -20 ° C in tetrahydrofuran, and isopropylmagnesium chloride (421.15 mg, 4.10 mmol) was added, and the mixture was reacted for 1 hour.
  • Propionyl chloride 8b (378.89 mg, 4.10 mmol), lithium chloride (11.42 mg, 189.00 ⁇ mol), cuprous chloride (9.36 mg, 94.50 ⁇ mol) and aluminum trichloride (12.61 mg, 94.50 ⁇ mol) were added to 1 mL of tetrahydrofuran. The mixture was stirred at room temperature, and the reaction liquid which had been reacted for 1 hour in advance was added to the above mixture, and reacted at room temperature for 2 hours. The reaction mixture was quenched by the addition of 20 mL of EtOAc EtOAc (EtOAc) The obtained residue was purified to give the title compound 8c (640 mg, yield: 82.0%).
  • reaction solution was cooled to room temperature, poured into 100 mL of ice-water, and then evaporated with ethyl acetate (60 mL ⁇ 3), and the organic phase was combined, washed with 25 mL of saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, The obtained residue was purified to silica gel elut372 elut elut elut elut elut elut
  • Chiral HPLC analysis retention time 5.297 minutes, (column: CHIRAL PAK IE, 4.6*150 mm, 5 um; flow rate: 1 mL/min; mobile phase: ethanol).
  • Chiral HPLC analysis retention time 8.442 minutes, (column: CHIRAL PAK IE, 4.6*150 mm, 5 um; flow rate: 1 mL/min; mobile phase: ethanol).
  • the crude compound 16d (70 mg, 115.89 ⁇ mol) was dissolved in 8 mL of tetrahydrofuran, and platinum chloride (5.26 mg, 23.18 ⁇ mol) was added thereto, and the mixture was replaced with hydrogen twice, and the mixture was stirred at room temperature for 2.5 hours. The reaction mixture was filtered, and the filtrate was evaporated.
  • the reaction was quenched by slowly adding 1 mL of water to the reaction mixture at -78 ° C, and the mixture was warmed to room temperature, then 10 mL of water was added, and the mixture was extracted with ethyl acetate (20 mL ⁇ 2), and the organic phase was combined and washed with saturated sodium chloride solution (20 mL ⁇ 2) Dry with anhydrous sodium sulfate. Filtration, the filtrate was concentrated under reduced pressure.
  • the compound 17f (200 mg, 0.297 mmol) was dissolved in a mixed solvent of 2 mL of methanol and 2 mL of tetrahydrofuran, and lithium hydroxide monohydrate (29.9 mg, 0.71 mmol) was added to the reaction mixture, and the reaction was stirred for 60 hours. 15 mL of water was added to the reaction mixture, and 3 M hydrochloric acid was added dropwise to pH 4 to 5, and extracted with ethyl acetate (20 mL ⁇ 2). The organic phase was combined and washed with saturated sodium chloride solution (20 mL ⁇ 2), anhydrous sodium sulfate Dry, filter, and concentrate the filtrate under reduced pressure. Purified by high-performance liquid chromatography (Waters 2767-SQ detecor 2, elution system: acetonitrile, water) The residue was obtained to give the title compound 17 (10 mg, yield: 16.2%).
  • Example 23 Using the synthetic route of Example 23, the starting compound 23a was replaced with 4-aminobenzamide (prepared by a known method "Chemical Communications (Cambridge, United Kingdom), 2017, 53 (35), 4807-4810") The title compound 24 (150 mg) was obtained.
  • Example 23 Using the synthetic route of Example 23, the starting compound 23a was replaced with 6-aminocarbazole (prepared by a known method "Tetrahedron Letters, 2010, 51 (5), 786-789") to give the title compound 27 ( 45mg).
  • Example 18 Using the synthetic route of Example 18, the starting compound 18a was replaced with 6-amino-1H-indole-3-carbonitrile (prepared by the method disclosed in the patent application "WO 20160271105”) to obtain the title compound 28 (30 mg). ).
  • Example 8 Using the synthetic route of Example 8, the starting compound 8c was replaced by 1-(2-bromo-4-chlorophenyl)-2,2,2-trifluoroethyl ketone (prepared by the method disclosed in the patent application "WO2011100285"). The title compound 35 (10 mg) was obtained.
  • the title compound 37 (40 mg) was obtained from the title compound.
  • Example 8 Using the synthetic route of Example 8, the starting compound 8b was replaced with cyclopropylacetyl chloride (prepared by the method disclosed in the patent application "WO 2015110435"), and the raw material cuprous chloride was replaced with cuprous iodide to obtain the title compound. 39 (30 mg).
  • the title compound 40 (80 mg) was obtained from the title compound 40a.
  • Example 8 Using the synthetic route of Example 8, the starting compound 8b was replaced with cyclopropanoyl chloride (prepared by the method disclosed in the patent application "WO 2015143380”) to give the title compound 41 (60 mg).
  • Example 18 Using the synthetic route of Example 18, the starting compound 18a was replaced with 6-aminocarbazole (prepared by the known method "Tetrahedron Letters, 2010, 51 (5), 786-789") to give the title compound 42 ( 83mg).
  • the title compound 43 (40 mg) was obtained from the title compound.
  • Example 8 Using the synthetic route of Example 8, the starting compound 8b was replaced with isobutyryl chloride (prepared by the known method "Organic Letters, 2017, 19(7), 1768-1771") to give the title compound 44 (200 mg). .
  • Example 48 Using the synthetic route of Example 48, the starting compound 48a was replaced with 2-methyl-1H-benzo[d]imidazole-5-amine (prepared by the method disclosed in the patent application "WO2012044090") to obtain the title compound. 49 (40 mg).
  • the title compound 50 (40 mg) was obtained from the title compound.
  • Example 38 Using the synthetic route of Example 38, the starting compound 38a was replaced with 6-amino-1H-indole-2-carbonitrile (prepared by the method disclosed in the patent application "WO20160271105”) to give the title compound 54 (30 mg). .
  • Example 18 Using the synthetic route of Example 18, the starting compound 18a was replaced with 4-amino-2-(trifluoromethyl)benzonitrile (prepared by a known method "Medicinal Chemistry Research, 2016, 25(4), 539-552"). The title compound 55 (40 mg) was obtained.
  • Example 18 Using the synthetic route of Example 18, the starting compound 18a was replaced with 4-(aminosulfonyl)aniline (prepared by a known method "Journal of Organic Chemistry, 2014, 79 (19), 9433-9439”). The title compound 58 (40 mg) was obtained.
  • Example 18 Using the synthetic route of Example 18, the starting compound 18a was replaced with 4-amino-3-fluorobenzonitrile (prepared by the well-known method "Journal of Medicinal Chemistry, 2005, 48 (18), 5823-5836"), The title compound 59 (20 mg) was obtained.
  • Example 18 Using the synthetic route of Example 18, the starting compound 18a was replaced with 6-amino-2H-benzo[b][1,4]oxazin-3(4H)-one (prepared by the method disclosed in the patent application "WO20100216783"). The title compound 60 (50 mg) was obtained.
  • Example 4 Using the synthetic route of Example 4, the starting compound 4a was replaced with 2-bromo-3-(4-fluorophenyl)propionic acid (prepared by the method disclosed in the patent application "US5981529A") to give the title compound 63 ( 64mg).
  • Example 7 Using the synthetic route of Example 7, the starting compound 7c was replaced with 1-bromo-2-(bromomethyl)benzene (prepared by a known method "Bioorganic & Medicinal Chemistry Letters, 2014, 24(21), 5127-5133"). The title compound 64 (8 mg) was obtained.
  • Example 7 Using the synthetic route of Example 7, the starting compound 7c was replaced with 1-(bromomethyl)-2-methylbenzene (prepared by a known method "Journal of Organic Chemistry, 2014, 79(1), 223-229”). The title compound 66 (60 mg) was obtained.
  • the starting compound 7c was replaced with 1-(bromomethyl)-3-methylbenzene by the synthetic route of Example 7 (using a known method "Chemical Communications (Cambridge, United Kingdom), 2014, 50 (28), 3692
  • the title compound 67 (80 mg) was obtained.
  • Example 7 Using the synthetic route of Example 7, the starting compound 7c was replaced by 1-(bromomethyl)-2-fluorobenzene (prepared by the known method "Tetrahedron Letters, 2000, 41 (27), 5161-5164") The title compound 68 (55 mg) was obtained.
  • Example 4 Using the synthetic route of Example 4, the starting compound 4a was replaced with 2-bromo-3-(4-chlorophenyl)propionic acid (prepared by the method disclosed in the patent application "WO2012118216") to obtain the title compound 69 ( 15mg).
  • Example 7 Using the synthetic route of Example 7, the starting compound 7c was replaced by 1-(bromomethyl)-2-chlorobenzene (prepared by the known method "Tetrahedron Letters, 2016, 57(2), 168-171") The title compound 70 (25 mg) was obtained.
  • Example 7 Using the synthetic route of Example 7, the starting compound 7c was replaced with 1-(bromomethyl)-3-methoxybenzene (prepared by the method disclosed in the patent application "WO2014135095") to give the title compound 71 (48 mg). ).
  • Example 74 Using the synthetic route of Example 74, the starting compound 74a was replaced with 2-(bromomethyl)-1,3-dichlorobenzene (prepared by the known method "Organic Letters, 2017, 19(7), 1634-1637". The title compound 76 (20 mg) was obtained.
  • Example 74 Using the synthetic route of Example 74, the starting compound 74a was replaced with 1-(bromomethyl)-2-methoxybenzene (using a known method "Journal of the American Chemical Society, 2013, 135(30), 10934- The title compound 78 (60 mg) was obtained.
  • Example 74 Using the synthetic route of Example 74, the starting compound 74a was replaced with 2-(bromomethyl)pyridine (prepared by the well-known method "Journal of the American Chemical Society, 2016, 138(26), 8253-8258"). The title compound 79 (370 mg) was obtained.
  • Example 74 Using the synthetic route of Example 74, the starting compound 74a was replaced with 3-(bromomethyl)pyridine (prepared by the known method "Chemical Communications (Cambridge, United Kingdom), 2016, 52(82), 12159-12162”). The title compound 82 (30 mg) was obtained.
  • Example 74 Using the synthetic route of Example 74, the starting compound 74a was replaced with 4-(bromomethyl)pyridine hydrobromide (using a known method "Chemical Communications (Cambridge, United Kingdom), 2011, 47(5), 1482- The title compound 86 (20 mg) was obtained.
  • Example 7 Using the synthetic route of Example 7, the starting compound 7c was replaced by 2-(bromomethyl)benzonitrile (known in the art) The title compound (manufactured by Journal of Organic Chemistry, 2014, 79(23), 11592-11608) was prepared to give the title compound 87 (15 mg).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Epidemiology (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Pulmonology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pyridine Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

本发明涉及氧代吡啶酰胺类衍生物、其制备方法及其在医药上的应用。具体而言,本发明涉及一种通式(AI)所示的氧代吡啶酰胺类衍生物、其制备方法及含有该衍生物的药物组合物以及其作为治疗剂,特别是作为凝血因子XIa(Factor XIa,简称FXIa)的抑制剂和在制备治疗血栓栓塞等疾病的药物中的用途,其中通式(AI)中的各取代基的定义与说明书中的定义相同。

Description

氧代吡啶酰胺类衍生物、其制备方法及其在医药上的应用 技术领域
本发明涉及一类新的氧代吡啶酰胺类衍生物、其制备方法及含有该衍生物的药物组合物以及其作为治疗剂特别是作为凝血XIa(Factor XIa,简称FXIa)的抑制剂和在制备治疗和预防血栓栓塞等疾病的药物中的用途。
背景技术
全球每年脑血管、脑梗塞、心肌梗塞、冠心病、动脉硬化等心脑血管疾病夺走近1200万人的生命,接近世界总死亡人数的1/4,成为人类健康的头号大敌。中国每年死于心血管疾病的人数达到260万人以上,存活的患者75%致残,其中40%以上重残。由心脑血管疾病和糖尿病及其并发症引起的血栓问题,成为当今要解决的刻不容缓的问题。
据独立市场分析机构Datamonitor 2011年数据预计,随着仿制药的生产,心血管及代谢疾病在七大主要市场的份额将在2011年达到顶峰,之后逐渐降低,其销售额将从2010年的1090亿美元将至2019年的1010亿美元。其中血栓市场保持基本稳定,由2010年的195亿美元微降至2019年的189亿美元(Datamonitor:HC00034-001、HC00139-001)。广州标点2011年的调研报告也显示,2011年中国抗血栓药物市场规模可达81.35亿元,同比增长20.52%,具有巨大的市场潜力(抗血栓药物市场研究报告:广州标点(2011))。
人体血液凝固过程由内源性途径(intrinsic pathway)、外源性途径(extrinsic pathway)和共同通路组成(Annu.Rev.Med.2011.62:41–57),是通过多种酶原被顺序激活而过程不断得到加强和放大的一种连锁反应。凝血级联反应由内源性途径(又称接触激活途径)及外源性途径(又称组织因子途径)启动生成FXa,再经共同途径生成凝血酶(FIIa),最终形成纤维蛋白。
内源性途径是指由XII因子被激活形XIa-VIIIa-Ca2±P L复合物、并激活X因子的过程,外源性凝血途径则是从组织因子(TF)释放到TF-VIIaCa2+复合物形成并激活因子Ⅹ的过程。共同通路是指因子Xa形成后,两条途径合二为一,激活凝血酶原并最终生成纤维蛋白的过程,其中FXI是维持内源性途径所必需的,而且在凝血级联反应放大过程中发挥关键作用。在凝血级联反应中,凝血酶可反馈激活FXI,活化的FXI(FXIa)又促使凝血酶的大量产生,从而使凝血级联反应放大。因此,FXI的拮抗剂被广泛开发,用于各种血栓的治疗。
传统的抗凝药物,如华法林、肝素、低分子量肝素(LMWH),以及近年上市的新药,如FXa抑制剂(利伐沙班、阿哌沙班等)和凝血酶抑制剂(达比加群酯、水蛭素等),对减少血栓形成均具有较好效果,以其显著有效性占据广大心脑血管市场,然而其副作用也越来越显著,其中“出血风险(bleeding risk)”是首当其冲 最为严峻的问题之一(N Engl J Med 1991;325:153-8、Blood.2003;101:4783-4788)。
人类FXI缺陷症(FXI活性<15U/dL)又称C型血友病,该类病人出血表型温和,少许发生自发性出血,即使在受伤或手术中机体的止血功能也不受影响,血友病C患者可以正常怀孕分娩(Arterioscler Thromb Vasc Biol.2010;30:388-392)。由此可知FXIa安全性显著优于FXa。因此,靶点FXIa成为各大公司及研究机构的研究热门。研究发现,在血栓模型中,抑制FXIa因子可以有效抑制血栓的形成,但在更为严重的血栓情况下,FXIa的作用微乎其微(Blood.2010;116(19):3981-3989)。临床统计显示,提高FXIa的量会增加VTE的患病率(Blood 2009;114:2878-2883),而FXIa严重不足者其患有DVT的风险性减少(Thromb Haemost 2011;105:269–273)。
FXIa作为新兴靶点,公开具有FXIa抑制活性的化合物的专利申请有WO9630396、WO9941276、WO2013093484、WO2004002405、WO2013056060、WO2017005725和US20050171148。其中,仅拜耳公司的反义寡核苷酸(antisense oligonucleotides,ASO)BAY-2306001进入了临床二期研究,并取得良好效果。在该药的临床I期试验中,受试者FXI活性出现持续的、剂量依赖地减少,并伴随着aPTT延长,即使体内FXI降至不能被检测的水平时,也不会出现与药物相关的出血症状,显示出FXIa作为新兴靶点的潜力(Arterioscler Thromb Vasc Biol,2013,33(7)1670-1678)。但FXI ASO需注射给药,且起效较慢,需要数周时间才能形成抗栓效果,在作为防治药物时可能会受到一定限制。在小分子抑制剂方面,仅有BMS公司的FXIa抑制剂于2014年进入临床一期,到目前为止尚无临床结果报道,因此新型FXIa抑制剂的研究具有重要意义。
本发明设计了具有通式(AI)结构的新的小分子FXIa拮抗剂,其中通式中R1为C(O)R7,对整个化合物的抗凝血作用和药代吸收活性具有显著改善,与已公开的具有类似母核结构的专利化合物相比,本发明化合物具有更高的活性。特别是本发明化合物表现出优异的对人血液的抗凝血作用,并具有良好的药代活性,可用于有效治疗和/或预防心脑血管疾病及血栓症状。
发明内容
本发明的目的在于提供一种通式(AI)所示的化合物:
Figure PCTCN2017099579-appb-000001
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用的盐,或其前药,
其中:
环A为芳基或杂芳基;
R1为-C(O)R7
R2相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、氨基、硝基、氰基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
R3选自卤素、烷基、卤代烷基、烷氧基、环烷基氧基、卤代烷氧基、氨基、硝基、氰基、羟基、羟烷基和烷硫基,其中所述的烷基、烷氧基、卤代烷基、卤代烷氧基、羟烷基和烷硫基任选进一步被选自氘原子、卤素、烷氧基、卤代烷氧基、氨基、硝基、氰基、羟基和羟烷基中的一个或多个取代基所取代;
R4选自氢原子、烷基、烷氧基、卤代烷基、羟烷基、环烷基、环烷基氧基、杂环基、芳基和杂芳基,其中所述的烷基、环烷基、杂环基、芳基和杂芳基任选进一步被一个或多个R9取代基所取代;
R5相同或不同,且各自独立地选自氢原子、烷基、烷氧基、氧代基、卤素、卤代烷基、卤代烷氧基、氨基、硝基、氰基、羟基、羟烷基、环烷基、杂环基、芳基、杂芳基、-C(O)R8、-C(O)OR8、-NHC(O)R8、-NHC(O)OR8、-NR10R11、-C(O)NR10R11、-CH2NHC(O)OR8、-CH2NR10R11、-C(O)OCH(R10)R11和-S(O)mR8,其中所述的烷基任选进一步被选自氘原子、卤素、烷氧基、卤代烷氧基、氨基、硝基、氰基、羟基、羟烷基、-NR10R11和-NHC(O)OR8中的一个或多个取代基所取代;
R7选自氢原子、烷基、卤代烷基、烷氧基、卤代烷氧基、氨基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、烷氧基、环烷基、杂环基、芳基和杂芳基任选进一步被选自氢原子、氘原子、卤素、烷基、烷氧基、卤代烷氧基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
R8选自氢原子、烷基、卤代烷基、羟基、氨基、环烷基、杂环基、芳基和杂芳基;
R9选自氢原子、氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基、杂环基、芳基和杂芳基,其中所述的环烷基、杂环基、芳基和杂芳基任选进一步被选自氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基、杂环基、-NHC(O)R12和R13中的一个或多个取代基所取代;
R10和R11相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、环烷基、杂环基、-C(O)OR8和-OC(O)OR12,其中所述的环烷基和杂环基任选进一步被选自氘原子、卤素、烷基、烷氧基、卤代烷基、氧代基、氨基、硝基、氰基、羟基和羟烷基中的一个或多个取代基所取代;
R12选自氢原子、烷基、烷氧基、卤代烷基、卤代烷氧基、氨基、环烷基、杂环基、芳基和杂芳基;
R13为芳基或杂芳基,其中所述的芳基和杂芳基任选进一步被选自氢原子、氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基和杂环基中的一个或多个取代基所取代;
n为0、1、2、3或4;
m为0、1或2;且
s为0、1、2、3或4。
在本发明一个优选的实施方案中,所述的通式(AI)所示的化合物为通式(I)所示的化合物:
Figure PCTCN2017099579-appb-000002
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用的盐、或其前药,
其中:
L1为亚烷基,其中所述的亚烷基任选进一步被卤素或氘原子中的一个或多个取代基所取代;
R6选自氢原子、氘原子、卤素、烷基、烷氧基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基、杂环基、芳基和杂芳基,其中所述的环烷基、杂环基、芳基和杂芳基任选进一步被选自氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基、杂环基、-NHC(O)R12和R13中的一个或多个取代基所取代;
R12选自氢原子、烷基、卤代烷基、氨基、环烷基、杂环基、芳基和杂芳基;
R13为芳基或杂芳基,其中所述的芳基和杂芳基任选进一步被选自氢原子、氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基和杂环基中的一个或多个取代基所取代;
环A、R1~R3、R5、n和s如通式(AI)中所定义。
在本发明一个优选的实施方案中,所述的通式(AI)所示的化合物为通式(Iaa)所示的化合物:
Figure PCTCN2017099579-appb-000003
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用的盐、或其前药,
其中:
环A、L1、R1~R3、R5~R6、n和s如通式(I)中所定义。
在本发明一个优选的实施方案中,所述的通式(AI)所示的化合物,其中
Figure PCTCN2017099579-appb-000004
选自:
Figure PCTCN2017099579-appb-000005
Figure PCTCN2017099579-appb-000006
其中R5和s如通式(AI)中所定义。
在本发明一个优选的实施方案中,所述的通式(AI)所示的化合物为通式(II)所示的化合物:
Figure PCTCN2017099579-appb-000007
其中:
R7选自烷基、环烷基和卤代烷基,其中所述的烷基或环烷基任选进一步被选自氘原子、卤素、烷基和环烷基中的一个或多个取代基所取代;
L1、R2、R3、R5、R6和n如通式(I)中所定义。
在本发明一个优选的实施方案中,所述的通式(AI)所示的化合物,其中R5相同或不同,且各自独立地选自烷基、烷氧基、氧代基、卤素、卤代烷基、氰基、羟基、-C(O)OR8、-NHC(O)OR8、-C(O)NR10R11、-CH2NHC(O)OR8、-CH2NR10R11、-C(O)OCH(R10)R11和-S(O)mR8;R8选自氢原子、烷基、羟基和氨基;R10和R11相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、环烷基、杂环基、-C(O)OR8和-OC(O)OR12,其中所述的环烷基和杂环基任选进一步被选自氘原子、卤素、烷基、烷氧基、卤代烷基、氧代基、氨基、硝基、氰基、羟基和羟烷基中的一个或多个取代基所取代;R12为烷基。
在本发明一个优选的实施方案中,所述的通式(AI)所示的化合物,其中R1为-C(O)R7;R7选自烷基、环烷基和卤代烷基,其中所述的烷基或环烷基任选进一步被选自氘原子、卤素、烷基和环烷基中的一个或多个取代基所取代。在本发明一个优选的实施方案中,所述的通式(AI)所示的化合物为通式(III)所示的化合物:
Figure PCTCN2017099579-appb-000008
其中:
R5相同或不同,且各自独立地选自烷基、烷氧基、氧代基、卤素、卤代烷基、氰基、羟基、-C(O)OR8、-NHC(O)OR8、-C(O)NR10R11、-CH2NHC(O)OR8、-CH2NR10R11、-C(O)OCH(R10)R11和-S(O)mR8;R8选自氢原子、烷基、羟基和氨基;R10和R11相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、环烷基、杂环基、-C(O)OR8和-OC(O)OR12,其中所述的环烷基和杂环基任选进一步被选自氘原子、卤素、烷基、烷氧基、卤代烷基、氧代基、氨基、硝基、氰基、羟基和羟烷基中的一个或多个取代基所取代;R12为烷基;
R7选自烷基、环烷基和卤代烷基,其中所述的烷基或环烷基任选进一步被选自氘原子、卤素、烷基和环烷基中的一个或多个取代基所取代;
L1、R2、R3、R6和n如通式(I)中所定义。
在本发明一个优选的实施方案中,所述的通式(AI)所示的化合物,其为通式(IV)所示的化合物:
Figure PCTCN2017099579-appb-000009
其中:
L1、R2、R3、R6、R7和n如通式(III)中所定义。
在本发明一个优选的实施方案中,所述的通式(AI)所示的化合物,其中R2为卤素;n为0、1或2。
在本发明一个优选的实施方案中,所述的通式(AI)所示的化合物,其中R3为烷氧基,其中所述的烷氧基任选进一步被氘原子和卤素中的一个或多个所取代。
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物,其中L1为-(CR14 2)x-,x为1~4的整数;R14为氢原子或氘原子;R6选自氢原子、氘原子、卤素、烷基、烷氧基、环烷基、环烷基氧基、杂环基、芳基和杂芳基;其中所述的环烷基、杂环基、芳基和杂芳基任选进一步被选自氘原子、卤素、烷基、烷氧基、环烷基、杂环基、-NHC(O)R12和R13中的一个或多个取代基所取代;R12为烷基或环烷基;R13为芳基或杂芳基,其中所述的芳基和杂芳基任选进一步被选自氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基和羟基中的一个或多个取代基所取代。。
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物,其中L1为-CH2-或-CD2-,其中D为氘原子;R6选自环烷基、杂环基、芳基和杂芳基;其中所述的环烷基、杂环基、芳基和杂芳基任选进一步被选自氘原子、卤素、烷基、烷氧基、环烷基、杂环基、-NHC(O)R12和R13中的一个或多个取代基所取代;R12为烷基或环烷基;R13为芳基或杂芳基,其中所述的芳基和杂芳基任选进一步被选 自氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基和羟基中的一个或多个取代基所取代。
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物,其中L1为-CH2CH2-;R6为烷基、烷氧基或环烷基氧基。
通式(AI)的典型化合物,包括但不限于:
Figure PCTCN2017099579-appb-000010
Figure PCTCN2017099579-appb-000011
Figure PCTCN2017099579-appb-000012
Figure PCTCN2017099579-appb-000013
Figure PCTCN2017099579-appb-000014
Figure PCTCN2017099579-appb-000015
Figure PCTCN2017099579-appb-000016
Figure PCTCN2017099579-appb-000017
Figure PCTCN2017099579-appb-000018
Figure PCTCN2017099579-appb-000019
Figure PCTCN2017099579-appb-000020
Figure PCTCN2017099579-appb-000021
Figure PCTCN2017099579-appb-000022
Figure PCTCN2017099579-appb-000023
Figure PCTCN2017099579-appb-000024
Figure PCTCN2017099579-appb-000025
Figure PCTCN2017099579-appb-000026
Figure PCTCN2017099579-appb-000027
Figure PCTCN2017099579-appb-000028
Figure PCTCN2017099579-appb-000029
Figure PCTCN2017099579-appb-000030
Figure PCTCN2017099579-appb-000031
Figure PCTCN2017099579-appb-000032
Figure PCTCN2017099579-appb-000033
Figure PCTCN2017099579-appb-000034
Figure PCTCN2017099579-appb-000035
Figure PCTCN2017099579-appb-000036
Figure PCTCN2017099579-appb-000037
Figure PCTCN2017099579-appb-000038
Figure PCTCN2017099579-appb-000039
Figure PCTCN2017099579-appb-000040
Figure PCTCN2017099579-appb-000041
Figure PCTCN2017099579-appb-000042
Figure PCTCN2017099579-appb-000043
Figure PCTCN2017099579-appb-000044
Figure PCTCN2017099579-appb-000045
Figure PCTCN2017099579-appb-000046
Figure PCTCN2017099579-appb-000047
Figure PCTCN2017099579-appb-000048
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用盐、或其前药。
在本发明另一个优选的实施方案中,一种制备通式(AI)所示的化合物的方法,该方法包括:
Figure PCTCN2017099579-appb-000049
通式(AI-A)的化合和通式(AI-B)的化合物或其盐酸盐在碱性条件下,进行缩合反应,任选在碱性条件下水解,得到通式(AI)化合物;
其中:
环A、R1~R5、n和s如通式(AI)中所定义。
在本发明另一个优选的实施方案中,一种制备通式(I)所示的化合物的方法,该方法包括:
Figure PCTCN2017099579-appb-000050
通式(I-A)的化合和通式(AI-B)的化合物或其盐酸盐在碱性条件下,进行缩合反应,任选在碱性条件下水解,得到通式(I)化合物;
其中:
环A、L1、R1~R3、R5~R6、n和s如通式(I)中所定义。
本发明的另一方面涉及一种药物组合物,其含有治疗有效剂量的通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐以及药学上可接受的载体、稀释剂和赋形剂。
本发明的另一方面涉及通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物,其用作药物。
本发明的另一方面涉及通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式或其可药用盐或包含其的药物组合物,在制备用于抑制XIa因子的药物中的用途。
本发明的另一方面涉及通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物,其用作XIa抑制剂。
本发明的另一方面涉及通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式或其可药用盐或包含其的药物组合物,在制备预防和/或治疗XIa因子介导的疾病的药物中的用途。
本发明的另一方面涉及通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物在制备预防和/或治疗心脑血管疾病中的用途,其中所述的心血管疾病,优选为血栓栓塞性疾病,更优选为心肌梗塞、心绞痛、血管成型术或主动脉冠状动脉分流术后的再阻塞和再狭窄、弥散性血管内凝血、中风、短暂的局部缺血发作、周围动脉闭塞性疾病、肺栓塞或深部静脉血栓形成。
本发明的另一方面涉及一种预防和/或治疗XIa因子介导的疾病的方法,其包括向患者施用治疗有效剂量的通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式或其可药用盐或包含其的药物组合物。
本发明的另一方面涉及一种治疗预防和/或治疗心脑血管疾病的方法,其包括向患者施用治疗有效剂量的通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式或其可药用盐或包含其的药物组合物,其中所述的心脑血管疾病选自心肌梗塞、心绞痛、血管成型术或主动脉冠状动脉分流术后的再阻塞和再狭窄、中风、短暂的局部缺血发作、周围动脉闭塞性疾病、肺栓塞或深部静脉血栓形成。
本发明的另一方面涉及一种用于抑制XIa因子的药物,其包括通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式或其可药用盐或包含其的药物组合物。
含活性成分的药物组合物可以是适用于口服的形式,例如片剂、糖锭剂、锭剂、水或油混悬液、可分散粉末或颗粒、乳液、硬或软胶囊,或糖浆剂或酏剂。可按照本领域任何已知制备药用组合物的方法制备口服组合物,此类组合物可含有一种或多种选自以下的成分:甜味剂、矫味剂、着色剂和防腐剂,以提供悦目和可口的药用制剂。片剂含有活性成分和用于混合的适宜制备片剂的无毒的可药 用的赋形剂。这些赋形剂可以是惰性赋形剂,造粒剂、崩解剂,粘合剂,和润滑剂,。这些片剂可以不包衣或可通过掩盖药物的味道或在胃肠道中延迟崩解和吸收,因而在较长时间内提供缓释作用的已知技术将其包衣。
也可用其中活性成分与惰性固体稀释剂或其中活性成分与水溶性载体或油溶媒混合的软明胶胶囊提供口服制剂。
水悬浮液含有活性物质和用于混合的适宜制备水悬浮液的赋形剂。此类赋形剂是悬浮剂,分散剂或湿润剂。水混悬液也可以含有一种或多种防腐剂、一种或多种着色剂、一种或多种矫味剂和一种或多种甜味剂。
油混悬液可通过使活性成分悬浮于植物油,或矿物油配制而成。油悬浮液可含有增稠剂。可加入上述的甜味剂和矫味剂,以提供可口的制剂。可通过加入抗氧化剂保存这些组合物。
通过加入水可使适用于制备水混悬的可分散粉末和颗粒提供活性成分和用于混合的分散剂或湿润剂、悬浮剂或一种或多种防腐剂。适宜的分散剂或湿润剂和悬浮剂可说明上述的例子。也可加入其他赋形剂例如甜味剂、矫味剂和着色剂。通过加入抗氧化剂例如抗坏血酸保存这些组合物。
本发明的药物组合物也可以是水包油乳剂的形式。油相可以是植物油,或矿物油或其混合物。适宜的乳化剂可以是天然产生的磷脂,乳剂也可以含有甜味剂、矫味剂、防腐剂和抗氧剂。此类制剂也可含有缓和剂、防腐剂、着色剂和抗氧剂。
本发明的药物组合物可以是无菌注射水溶液形式。可以使用的可接受的溶媒或溶剂有水、林格氏液和等渗氯化钠溶液。无菌注射制剂可以是其中活性成分溶于油相的无菌注射水包油微乳可通过局部大量注射,将注射液或微乳注入患者的血流中。或者,最好按可保持本发明化合物恒定循环浓度的方式给予溶液和微乳。为保持这种恒定浓度,可使用连续静脉内递药装置。这种装置的实例是Deltec CADD-PLUS.TM.5400型静脉注射泵。
本发明的药物组合物可以是用于肌内和皮下给药的无菌注射水或油混悬液的形式。可按已知技术,用上述那些适宜的分散剂或湿润剂和悬浮剂配制该混悬液。无菌注射制剂也可以是在肠胃外可接受的无毒稀释剂或溶剂中制备的无菌注射溶液或混悬液。此外,可方便地用无菌固定油作为溶剂或悬浮介质。为此目的,可使用任何调和固定油。此外,脂肪酸也可以制备注射剂。
可按用于直肠给药的栓剂形式给予本发明化合物。可通过将药物与在普通温度下为固体但在直肠中为液体,因而在直肠中会溶化而释放药物的适宜的无刺激性赋形剂混合来制备这些药物组合物。
如本领域技术人员所熟知的,药物的给药剂量依赖于多种因素,包括但并非限定于以下因素:所用具体化合物的活性、患者的年龄、患者的体重、患者的健康状况、患者的行为、患者的饮食、给药时间、给药方式、排泄的速率、药物的 组合等;另外,最佳的治疗方式如治疗的模式、通式化合物(I)的日用量或可药用的盐的种类可以根据传统的治疗方案来验证。
发明的详细说明
除非有相反陈述,在说明书和权利要求书中使用的术语具有下述含义。
术语“烷基”指饱和脂肪族烃基团,其为包含1至20个碳原子的直链或支链基团,优选含有1至12个碳原子的烷基,更优选含有1至6个碳原子的烷基。非限制性实例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基、正壬基、2-甲基-2-乙基己基、2-甲基-3-乙基己基、2,2-二乙基戊基、正癸基、3,3-二乙基己基、2,2-二乙基己基,及其各种支链异构体等。更优选的是含有1至6个碳原子的低级烷基,非限制性实施例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基等。烷基可以是取代的或非取代的,当被取代时,取代基可以在任何可使用的连接点上被取代,所述取代基优选为一个或多个以下基团,其独立地选自氘原子、烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、-C(O)R8、-C(O)OR8和-S(O)mR8中的一个或多个取代基所取代。
术语“亚烷基”指饱和的直链或支链脂肪族烃基,其具有2个从母体烷的相同碳原子或两个不同的碳原子上除去两个氢原子所衍生的残基,其为包含1至20个碳原子的直链或支链基团,优选含有1至12个碳原子,更优选含有1至6个碳原子的亚烷基。亚烷基的非限制性实例包括但不限于亚甲基(-CH2-)、1,1-亚乙基(-CH(CH3)-)、1,2-亚乙基(-CH2CH2)-、1,1-亚丙基(-CH(CH2CH3)-)、1,2-亚丙基(-CH2CH(CH3)-)、1,3-亚丙基(-CH2CH2CH2-)、1,4-亚丁基(-CH2CH2CH2CH2-)等。亚烷基可以是取代的或非取代的,当被取代时,取代基可以在任何可使用的连接点上被取代,所述取代基优选独立地任选选自氘原子、烷基、烯基、炔基、烷氧基、 烷硫基、烷基氨基、卤素、硫醇、羟基、硝基、氰基、环烷基、杂环基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、-C(O)R8、-C(O)OR8和-S(O)mR8中的一个或多个取代基所取代。
术语“环烷基”指饱和或部分不饱和单环或多环环状烃取代基,环烷基环包含3至20个碳原子,优选包含3至12个碳原子,更优选包含3至8个碳原子,最优选包含3至5个碳原子。单环环烷基的非限制性实例包括环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等,优选环烷基;多环环烷基包括螺环、稠环和桥环的环烷基。
术语“螺环烷基”指5至20元的单环之间共用一个碳原子(称螺原子)的多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺环烷基分为单螺环烷基、双螺环烷基或多螺环烷基,优选为单螺环烷基和双螺环烷基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺环烷基。螺环烷基的非限制性实例包括:
Figure PCTCN2017099579-appb-000051
术语“稠环烷基”指5至20元,系统中的每个环与体系中的其他环共享毗邻的一对碳原子的全碳多环基团,其中一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠环烷基,优选为双环或三环,更优选为5元/5元或5元/6元双环烷基。稠环烷基的非限制性实例包括:
Figure PCTCN2017099579-appb-000052
术语“桥环烷基”指5至20元,任意两个环共用两个不直接连接的碳原子的全碳多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥环烷基,优选为双环、三环或四环,更有选为双环或三环。桥环烷基的非限制性实例包括:
Figure PCTCN2017099579-appb-000053
所述环烷基环可以稠合于芳基、杂芳基或杂环烷基环上,其中与母体结构连接在一起的环为环烷基,非限制性实例包括茚满基、四氢萘基、苯并环庚烷基等;优选苯基并环戊基、四氢萘基。环烷基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、-C(O)R8、-C(O)OR8和-S(O)mR8中的一个或多个取代基所取代。
术语“杂环基”指饱和或部分不饱和单环或多环环状烃取代基,其包含3至20个环原子,其中一个或多个环原子为选自氮、氧或S(O)m(其中m是整数0至2)的杂原子,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包含3至12个环原子,其中1~4个是杂原子;最优选包含3至8个环原子,其中1~3个是杂原子;最优选包含3至5个环原子,其中1~2或1~3个是杂原子。单环杂环基的非限制性实例包括吡咯烷基、咪唑烷基、四氢呋喃基、四氢吡喃基、四氢噻吩基、二氢咪唑基、二氢呋喃基、间二氧杂环戊烯、二氢吡唑基、二氢吡咯基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基等。多环杂环基包括螺环、稠环和桥环的杂环基。
术语“螺杂环基”指5至20元的单环之间共用一个原子(称螺原子)的多环杂环基团,其中一个或多个环原子为选自氮、氧或S(O)m(其中m是整数0至2)的杂原子,其余环原子为碳。其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺杂环基分为单螺杂环基、双螺杂环基或多螺杂环基,优选为单螺杂环基和双螺杂环基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺杂环基。螺杂环基的非限制性实例包括:
Figure PCTCN2017099579-appb-000054
术语“稠杂环基”指5至20元,系统中的每个环与体系中的其他环共享毗邻的一对原子的多环杂环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统,其中一个或多个环原子为选自氮、氧或S(O)m(其 中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠杂环基,优选为双环或三环,更优选为5元/5元或5元/6元双环稠杂环基。稠杂环基的非限制性实例包括:
Figure PCTCN2017099579-appb-000055
术语“桥杂环基”指5至14元,任意两个环共用两个不直接连接的原子的多环杂环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统,其中一个或多个环原子为选自氮、氧或S(O)m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥杂环基,优选为双环、三环或四环,更有选为双环或三环。桥杂环基的非限制性实例包括:
Figure PCTCN2017099579-appb-000056
所述杂环基环可以稠合于芳基、杂芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基,其非限制性实例包括:
Figure PCTCN2017099579-appb-000057
等。
杂环基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、-C(O)R8、-C(O)OR8和-S(O)mR8中的一个或多个取代基所取代。
术语“芳基”指具有共轭的π电子体系的6至20元全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,优选为6至10元,更优选6元,例如苯基和萘基。 所述芳基环可以稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为芳基环,其非限制性实例包括:
Figure PCTCN2017099579-appb-000058
芳基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自氘原子、烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、-C(O)R8、-C(O)OR8和-S(O)mR8中的一个或多个取代基所取代。
术语“杂芳基”指包含1至4个杂原子、5至20个环原子的杂芳族体系,其中杂原子选自氧、硫和氮。杂芳基优选为5至10元,含1至3个杂原子;更优选为5元或6元,含1至2个杂原子;优选例如咪唑基、呋喃基、噻吩基、噻唑基、吡唑基、噁唑基、吡咯基、三唑基、四唑基、吡啶基、嘧啶基、噻二唑、吡嗪基等。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,其非限制性实例包括:
Figure PCTCN2017099579-appb-000059
杂芳基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、 杂环烷氧基、环烷硫基、杂环烷硫基、-C(O)R8、-C(O)OR8和-S(O)mR8中的一个或多个取代基所取代。
术语“烷氧基”指-O-(烷基)和-O-(非取代的环烷基),其中烷基的定义如上所述。烷氧基的非限制性实例包括:甲氧基、乙氧基、丙氧基、丁氧基、环丙氧基、环丁氧基、环戊氧基、环己氧基。烷氧基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、-C(O)R8、-C(O)OR8和-S(O)mR8中的一个或多个取代基所取代。
术语“烷硫基”指-S-(烷基)和-S-(非取代的环烷基),其中烷基的定义如上所述。烷硫基的非限制性实例包括:甲硫基、乙硫基、丙硫基、丁硫基、环丙硫基、环丁硫基、环戊硫基、环己硫基。烷硫基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、-C(O)R8、-C(O)OR8和-S(O)mR8中的一个或多个取代基所取代。
术语“环烷基氧基”指-O-环烷基,其中环烷基如上所定义。
术语“卤代烷基”指被卤素取代的烷基,其中烷基如上所定义。
术语“卤代烷氧基”指被卤素取代的烷氧基,其中烷氧基如上所定义。
术语“羟烷基”指被羟基取代的烷基,其中烷基如上所定义。
术语“羟基”指-OH基团。
术语“卤素”指氟、氯、溴或碘。
术语“氨基”指-NH2
术语“氰基”指-CN。
术语“硝基”指-NO2
术语“羧基”指-C(O)OH。
术语“羧酸酯基”指-C(O)O(烷基)或-C(O)O(环烷基),其中烷基、环烷基如上所定义。
“任选”或“任选地”意味着随后所描述的事件或环境可以但不必发生,该说明包括该事件或环境发生或不发生的场合。例如,“任选被烷基取代的杂环基团”意味着烷基可以但不必须存在,该说明包括杂环基团被烷基取代的情形和杂环基团不被烷基取代的情形。
“取代的”指基团中的一个或多个氢原子,优选为最多5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验 或理论)可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。
“药物组合物”表示含有一种或多种本文所述化合物或其生理学上/可药用的盐或前体药物与其他化学组分的混合物,以及其他组分例如生理学/可药用的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
“可药用盐”是指本发明化合物的盐,这类盐用于哺乳动物体内时具有安全性和有效性,且具有应有的生物活性。
R8和m如通式(AI)所定义。
本发明化合物的合成方法
为了完成本发明的目的,本发明采用如下技术方案:
方案一
本发明通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐制备方法,包括以下步骤:
Figure PCTCN2017099579-appb-000060
第一步反应为通式(AI-1)化合物在有机溶剂中,碱性条件下与通式(AI-2)进行亲核取代反应得到通式(AI–3)化合物;或者通式(AI-1)化合物在有机溶剂中,碱性条件下与通式(AI-2’)进行亲核取代反应得到通式(AI–A)化合物;
第二步反应为通式(AI-3)化合物在酸性条件下,水解得到通式(AI-A);
第三步反应为通式(AI-A)的化合物和通式(AI-B)的化合物或其盐酸盐在碱性条件下,进行缩合反应,任选在碱性条件下水解,得到通式(AI)化合物;
提供碱性条件的试剂包括有机碱和无机碱类,所述的有机碱类包括但不限于三乙胺、N,N-二异丙基乙胺、正丁基锂、二异丙基氨基锂、双三甲基硅基胺基锂、醋酸钾、叔丁醇钠或叔丁醇钾,所述的无机碱类包括但不限于氢化钠、磷酸钾、碳酸钠、碳酸钾、碳酸铯、氢氧化钠和氢氧化锂。
提供酸性的条件包括但不限于吡啶氢溴酸盐、三氟乙酸、甲酸、乙酸、盐酸、硫酸或甲磺酸,优选为吡啶氢溴酸盐或盐酸。
所述缩合剂包括但不限于1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐、N,N'-二环己基碳化二亚胺、N,N'-二异丙基碳二酰亚胺、O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸酯、1-羟基苯并三唑、1-羟基-7-偶氮苯并三氮唑、O-苯并三氮唑-N,N,N',N'-四甲脲六氟磷酸酯、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、苯并三氮唑-1-基氧基三(二甲基氨基)磷鎓六氟磷酸盐或六氟磷酸苯并三唑-1-基-氧基三吡咯烷基磷,优选为2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯。
上述反应优选在溶剂中进行,所用溶剂包括但不限于:醋酸、甲醇、乙醇、甲苯、四氢呋喃、二氯甲烷、二甲基亚砜、1,4-二氧六环、水或N,N-二甲基甲酰胺。
其中:
X为卤素,优选为溴;
Ra为烷基,优选为甲基;
环A、R1~R5、n和s如通式(AI)中所定义。
方案二
本发明通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐制备方法,包括以下步骤:
Figure PCTCN2017099579-appb-000061
第一步反应为式(I-1)化合物在有机溶剂中,碱性条件下与(I-2)进行亲核取代反应得到式(I-3)化合物;或者通式(I-1)化合物在有机溶剂中,碱性条件下与通式(I-2’)进行亲核取代反应得到通式(I–A)化合物;
第二步反应为通式(I-3)化合物在酸性条件下,水解得到通式(I-A);
第三步反应为式(I-A)的化合物和(AI-B)的化合物或其盐酸盐在碱性条件下,进行缩合反应,任选在碱性条件下水解,得到通式(I)化合物;
提供碱性条件的试剂包括有机碱和无机碱类,所述的有机碱类包括但不限于三乙胺、N,N-二异丙基乙胺、正丁基锂、二异丙基氨基锂、双三甲基硅基胺基锂、醋酸钾、叔丁醇钠或叔丁醇钾,所述的无机碱类包括但不限于氢化钠、磷酸钾、碳酸钠、碳酸钾、碳酸铯、氢氧化钠和氢氧化锂。
提供酸性的条件包括但不限于吡啶氢溴酸盐、三氟乙酸、甲酸、乙酸、盐酸、硫酸或甲磺酸,优选为吡啶氢溴酸盐或盐酸。
缩合剂包括但不限于1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐、N,N'-二环己基碳化二亚胺、N,N'-二异丙基碳二酰亚胺、O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸酯、1-羟基苯并三唑、1-羟基-7-偶氮苯并三氮唑、O-苯并三氮唑-N,N,N',N'-四甲脲六氟磷酸酯、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、苯并三氮唑-1-基氧基三(二甲基氨基)磷鎓六氟磷酸盐或六氟磷酸苯并三唑-1-基-氧基三吡咯烷基磷,优选为2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯。
上述反应优选在溶剂中进行,所用溶剂包括但不限于:醋酸、甲醇、乙醇、甲苯、四氢呋喃、二氯甲烷、二甲基亚砜、1,4-二氧六环、水或N,N-二甲基甲酰胺。
其中:
X为卤素,优选为溴;
Ra为烷基,优选为甲基;
环A、L1、R1~R3、R5~R6、n和s如通式(I)中所定义。
方案三
本发明通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐制备方法,包括以下步骤:
Figure PCTCN2017099579-appb-000062
第一步反应为式(I-1-a)化合物在有机溶剂中,碱性条件下与(I-2-a)进行亲核取代反应得到式(I-3)化合物;
第二步反应为通式(I-3)化合物在酸性条件下,水解得到通式(I-A);
第三步反应为式(I-A)的化合物和(AI-B)的化合物或其盐酸盐在碱性条件下,进行缩合反应,任选在碱性条件下水解,得到通式(I)化合物;
提供碱性条件的试剂包括有机碱和无机碱类,所述的有机碱类包括但不限于三乙胺、N,N-二异丙基乙胺、正丁基锂、二异丙基氨基锂、双三甲基硅基胺基锂、醋酸钾、叔丁醇钠或叔丁醇钾,所述的无机碱类包括但不限于氢化钠、磷酸钾、碳酸钠、碳酸钾、碳酸铯、氢氧化钠和氢氧化锂。
提供酸性的条件包括但不限于吡啶氢溴酸盐、三氟乙酸、甲酸、乙酸、盐酸、硫酸或甲磺酸,优选为吡啶氢溴酸盐或盐酸。
缩合剂包括但不限于1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐、N,N'-二环己基碳化二亚胺、N,N'-二异丙基碳二酰亚胺、O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸酯、1-羟基苯并三唑、1-羟基-7-偶氮苯并三氮唑、O-苯并三氮唑-N,N,N',N'-四甲脲六氟磷酸酯、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、苯并三氮唑-1-基氧基三(二甲基氨基) 磷鎓六氟磷酸盐或六氟磷酸苯并三唑-1-基-氧基三吡咯烷基磷,优选为2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯。
上述反应优选在溶剂中进行,所用溶剂包括但不限于:醋酸、甲醇、乙醇、甲苯、四氢呋喃、二氯甲烷、二甲基亚砜、1,4-二氧六环、水或N,N-二甲基甲酰胺。
其中:
Ra为烷基,优选为甲基;
Rb为离去基团,优选为甲磺酰氧基或三氟甲磺酰氧基;
环A、L1、R1~R3、R5~R6、n和s如通式(I)中所定义。
方案四
本发明通式(II)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐制备方法,包括以下步骤:
Figure PCTCN2017099579-appb-000063
第一步反应为式(II-1)化合物在有机溶剂中,碱性条件下与(I-2)进行亲核取代反应得到式(II-2)化合物;或者通式(II-1)化合物在有机溶剂中,碱性条件下与通式(I-2’)进行亲核取代反应得到通式(II–A)化合物;
第二步反应为通式(II-2)化合物在酸性条件下,水解得到通式(II-A);
第三步反应为式(II-A)的化合物和(II-B)的化合物或其盐酸盐在碱性条件下,进行缩合反应,任选在碱性条件下水解,得到通式(II)化合物;
提供碱性条件的试剂包括有机碱和无机碱类,所述的有机碱类包括但不限于三乙胺、N,N-二异丙基乙胺、正丁基锂、二异丙基氨基锂、双三甲基硅基胺基锂、醋酸钾、叔丁醇钠或叔丁醇钾,优选为双三甲基硅基胺基锂,所述的无机碱类包 括但不限于氢化钠、磷酸钾、碳酸钠、碳酸钾、碳酸铯、氢氧化钠或氢氧化锂,优选为碳酸钾、氢化钠或氢氧化锂。
提供酸性的条件包括但不限于吡啶氢溴酸盐、三氟乙酸、甲酸、乙酸、盐酸、硫酸或甲磺酸,优选为吡啶氢溴酸盐或盐酸。
缩合剂包括但不限于1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐、N,N'-二环己基碳化二亚胺、N,N'-二异丙基碳二酰亚胺、O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸酯、1-羟基苯并三唑、1-羟基-7-偶氮苯并三氮唑、O-苯并三氮唑-N,N,N',N'-四甲脲六氟磷酸酯、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、苯并三氮唑-1-基氧基三(二甲基氨基)磷鎓六氟磷酸盐或六氟磷酸苯并三唑-1-基-氧基三吡咯烷基磷,优选为2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯。
上述反应优选在溶剂中进行,所用溶剂包括但不限于:醋酸、甲醇、乙醇、甲苯、四氢呋喃、二氯甲烷、二甲基亚砜、1,4-二氧六环、水或N,N-二甲基甲酰胺。
其中:
X为卤素,优选为溴;
Ra为烷基,优选为甲基;
L1、R2、R3、R5~R7和n如通式(II)中所定义;
方案五
本发明通式(IV)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用的盐制备方法,包括以下步骤:
Figure PCTCN2017099579-appb-000064
第一步反应为式(II-A)的化合和(IV-1)的化合物或其盐酸盐在碱性条件下,进行缩合反应,得到通式(IV-2)化合物;
第二步反应为式(IV-2)化合物在酸性条件下,水解得到式(IV-3)化合物;
第三步反应为式(IV-3)化合物进行手性制备得到通式(IV)化合物;
提供碱性条件的试剂包括有机碱和无机碱类,所述的有机碱类包括但不限于三乙胺、N,N-二异丙基乙胺、正丁基锂、二异丙基氨基锂、双三甲基硅基胺基锂、醋酸钾、叔丁醇钠或叔丁醇钾,优选为双三甲基硅基胺基锂,所述的无机碱类包括但不限于氢化钠、磷酸钾、碳酸钠、碳酸钾、碳酸铯、氢氧化钠或氢氧化锂,优选为碳酸钾、氢化钠或氢氧化锂。
提供酸性的条件包括但不限于吡啶氢溴酸盐、三氟乙酸、甲酸、乙酸、盐酸、硫酸或甲磺酸,优选为吡啶氢溴酸盐或盐酸。
手性制备的条件包括但不限于色谱柱为Superchiral S-AD(Chiralway),流动相为二氧化碳,乙醇和二乙胺。
缩合剂包括但不限于1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐、N,N'-二环己基碳化二亚胺、N,N'-二异丙基碳二酰亚胺、O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸酯、1-羟基苯并三唑、1-羟基-7-偶氮苯并三氮唑、O-苯并三氮唑-N,N,N',N'-四甲脲六氟磷酸酯、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、苯并三氮唑-1-基氧基三(二甲基氨基)磷鎓六氟磷酸盐或六氟磷酸苯并三唑-1-基-氧基三吡咯烷基磷,优选为2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯。
上述反应优选在溶剂中进行,所用溶剂包括但不限于:醋酸、甲醇、乙醇、甲苯、四氢呋喃、二氯甲烷、二甲基亚砜、1,4-二氧六环、水或N,N-二甲基甲酰胺。
其中:
Rc为烷基,优选为甲基;
L1、R2、R3、R6、R7和n如通式(IV)中所定义。
本发明所涉及的各通式化合物,如在合成过程中得到化合物盐的形式,可进一步采用常规的实验手段得到游离态化合物;如在合成过程中得到游离态化合物形式,可进一步采用常规实验手段得到化合物的盐。
具体实施方式
以下结合实施例进一步描述本发明,但这些实施例并非限制着本发明的范围。
实施例
化合物的结构是通过核磁共振(NMR)或/和质谱(MS)来确定的。NMR位移(δ)以10-6(ppm)的单位给出。NMR的测定是用Bruker AVANCE-400核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6),氘代氯仿(CDCl3),氘代甲醇(CD3OD),内标为四甲基硅烷(TMS)。
MS的测定用FINNIGAN LCQAd(ESI)质谱仪(生产商:Thermo,型号:Finnigan LCQ advantage MAX)。
高效液相色谱法(HPLC)分析使用Agilent HPLC 1200DAD、Agilent HPLC 1200VWD和Waters HPLC e2695-2489高压液相色谱仪。
手性HPLC分析测定使用Agilent 1260DAD高效液相色谱仪。
CombiFlash快速制备仪使用Combiflash Rf200(TELEDYNE ISCO)。
薄层层析硅胶板使用烟台黄海HSGF254或青岛GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm~0.2mm,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm。
硅胶柱色谱法一般使用烟台黄海硅胶200~300目硅胶为载体。
激酶平均抑制率及IC50值的测定用NovoStar酶标仪(德国BMG公司)。
本发明的已知的起始原料可以采用或按照本领域已知的方法来合成,或可购买自ABCR GmbH & Co.KG,Acros Organics,Aldrich Chemical Company,韶远化学科技(Accela ChemBio Inc)、达瑞化学品等公司。
实施例中无特殊说明,反应能够均在氩气氛或氮气氛下进行。
氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
氢气氛是指反应瓶连接一个约1L容积的氢气气球。
加压氢化反应使用Parr 3916EKX型氢化仪和清蓝QL-500型氢气发生器或HC2-SS型氢化仪。
氢化反应通常抽真空,充入氢气,反复操作3次。
微波反应使用CEM Discover-S 908860型微波反应器。
实施例中无特殊说明,溶液是指水溶液。
实施例中无特殊说明,反应的温度为室温,为20℃~30℃。
实施例中的反应进程的监测采用薄层色谱法(TLC),反应所使用的展开剂,纯化化合物采用的柱层析的洗脱剂的体系和薄层色谱法的展开剂体系包括:A:二氯甲烷/甲醇体系,B:正己烷/乙酸乙酯体系,C:石油醚/乙酸乙酯体系,D:丙酮,E:二氯甲烷/丙酮体系,F:乙酸乙酯/二氯甲烷体系,G:乙酸乙酯/二氯甲烷/正己烷,H:乙酸乙酯/二氯甲烷/丙酮,I:石油醚/乙酸乙酯/二氯甲烷,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的三乙胺和醋酸等碱性或酸性试剂进行调节。
实施例1
5-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酰基氨基)-1H-吲哚-2-羧酸
Figure PCTCN2017099579-appb-000065
Figure PCTCN2017099579-appb-000066
第一步
2-溴-4-甲氧基丁酸甲酯1b
将4-甲氧基丁酸甲酯1a(1.6g,12.1mmol)加入到50mL四氢呋喃中,用干冰-丙酮浴降温至-78℃,缓慢加入双三甲基硅基胺基锂(12.7mL,12.7mmol),加毕,搅拌反应1小时,加入三甲基氯硅烷(1.31g,12.1mmol),继续搅拌反应20分钟,再加入N-溴代丁二酰亚胺(2.15g,12.1mmol),搅拌反应2小时。撤去干冰-丙酮浴,反应液温度自然升至室温,用饱和氯化铵溶液淬灭反应,反应液用乙酸乙酯萃取(50mL×3),合并有机相,有机相分别用水(50mL),饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题产物1b(900mg,产率:35.3%)。
第二步
1-(4-氯-2-(2,5-二甲氧基吡啶-4-基)苯基)乙酮1e
将2-溴-4-氯苯乙酮1c(1.27g,5.46mmol)、(2,5-二甲氧基吡啶-4-基)硼酸1d(1.0g,5.46mmol,采用专利申请“WO2015063093”公开的方法制备而得)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(230mg,0.32mmol)和碳酸钾(2.2g,16.38mmol)加入到25mL的1,4-二氧六环中,加毕,反应液加热至110℃,搅拌反应8小时。反应液自然冷却至室温,向反应液中加入50mL水,用乙酸乙酯萃取(50mL×3),合并有机相,有机相分别用水(50mL),饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题产物1e(1.0g,产率:63.3%)。
MS m/z(ESI):292.3[M+1]
第三步
4-(2-乙酰基-5-氯苯基)-5-甲氧基吡啶-2(1H)-酮1f
将1e(1.0g,3.43mmol)加入到10mL的N,N-二甲基甲酰胺中,加入吡啶氢溴酸盐(3.30g,20.6mmol),加毕,加热至105℃,搅拌反应3小时。向反应液中加入50mL水,用乙酸乙酯萃取(50mL×3),合并有机相,有机相分别用水(50mL),饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩得到标题产物1f(550mg,产率:57.8%)。
MS m/z(ESI):276.3[M-1]
第四步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酸1g
将1f(350mg,1.28mmol)加入到10mL N,N-二甲基甲酰胺中,冰水浴冷却下缓慢加入氢化钠(153mg,3.84mmol),加毕,撤去冰浴,反应液温度自然升至室温,搅拌反应30分钟,加入1b(350mg,1.66mmol),继续搅拌反应24小时。向反应液中加入50mL水,滴加1M的盐酸至反应液pH至3~4,用乙酸乙酯(50mL×3)萃取,合并有机相,用水(50mL),饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤除去干燥剂,滤液减压浓缩得到粗品标题产物1g(360mg),产物不经纯化直接用于下一步反应。
MS m/z(ESI):394.4[M+1]
第五步
5-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酰氨基)-1H-吲哚-2-羧酸乙酯1i
将粗品1g(180mg,0.45mmol)加入到10mL N,N-二甲基甲酰胺中,再加入2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(313mg,0.82mmol)、N,N-二异丙基乙胺(0.22mL,1.35mmol)和5-氨基-1H-吲哚-2-羧酸乙酯盐酸盐1h(129mg,0.54mmol,采用公知的方法“Journal of Organic Chemistry,2012,55(2),766-782”制备而得),加毕,加热至50℃,搅拌反应16小时。向反应液中加入50mL水,用乙酸乙酯(50mL×3)萃取,合并有机相,用水(50mL),饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤除去干燥剂,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题产物1i(60mg,产率23.1%)。
MS m/z(ESI):580.4[M+1]
第六步
5-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酰基氨基)-1H-吲哚-2-羧酸1
将1i(60mg,0.103mmol)加入到4mL四氢呋喃和甲醇的混合溶剂(V:V=3:1)中,加入1M的氢氧化锂溶液(0.83mL,0.83mmol),加毕,搅拌反应16小时。反应液减压蒸除溶剂,向所得残余物中加入10mL水,搅拌均匀,滴加1M的盐 酸至反应液pH至3~4,乙酸乙酯萃取(50mL×3),合并有机相,有机相分别用水(50mL),饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤除去干燥剂,滤液减压浓缩,用高效液相制备(Gilson-281,洗脱体系:乙腈,水)纯化所得残余物,制得标题产物1(4mg,产率:7.0%)。
MS m/z(ESI):552.4[M+1]
1H NMR(400MHz,DMSO-d6)δ11.60(s,1H),11.33(s,1H),8.01(s,1H),7.89-7.87(d,1H),7.64-7.61(dd,1H),7.47-7.46(d,1H),7.37-7.32(m,3H),6.97(s,1H),6.40(s,1H),5.79-5.75(m,1H),3.54(s,3H),3.22(s,3H),2.42-2.31(m,2H)
实施例2,3
(S)-5-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酰氨基)-1H-吲哚-2-羧酸2
(R)-5-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酰氨基)-1H-吲哚-2-羧酸3
Figure PCTCN2017099579-appb-000067
将1(100mg,0.18mmol)进行手性制备(分离条件:手性制备柱CHIRAL PAK IE,20*250mm,5um;流动相:流动相:乙醇(含0.01%三氟乙酸)=100,流速:8mL/min),收集其相应组分,减压浓缩,得到标题产物3(10mg)和4(15mg)。
化合物2:
MS m/z(ESI):552.5[M+1]
手性HPLC分析:保留时间8.907分钟,手性纯度:98%(色谱柱:CHIRAL PAK IE,4.6*150mm,5um;流动相:正己烷/乙醇/三氟乙酸=40/60/0.06(v/v/v))。
1H NMR(400MHz,DMSO-d6)δ11.60(s,1H),11.33(s,1H),8.01(s,1H),7.89-7.87(d,1H),7.64-7.61(dd,1H),7.47-7.46(d,1H),7.37-7.32(m,3H),6.97(s,1H),6.40(s,1H),5.79-5.75(m,1H),3.54(s,3H),3.22(s,3H),2.42-2.31(m,2H)
化合物3:
MS m/z(ESI):552.4[M+1]
手性HPLC分析:保留时间6.720分钟,手性纯度:98%(色谱柱:CHIRAL PAK IE,4.6*150mm,5um;流动相:正己烷/乙醇/三氟乙酸=40/60/0.06(v/v/v))。
1H NMR(400MHz,DMSO-d6)δ11.60(s,1H),11.33(s,1H),8.01(s,1H),7.89-7.87(d,1H),7.64-7.61(dd,1H),7.47-7.46(d,1H),7.37-7.32(m,3H),6.97(s,1H),6.40(s,1H),5.79-5.75(m,1H),3.54(s,3H),3.22(s,3H),2.42-2.31(m,2H)
实施例4
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸4
Figure PCTCN2017099579-appb-000068
第一步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯丙酸4b
将叔丁醇镁(701.62mg,7.2mmol)溶于250mL四氢呋喃中,再加入(R)-2-溴-3-苯丙酸(1649.77mg,7.2mmol,采用公知方法“Chemical Communications(Cambridge,United Kingdom),2014,50(88),13489-13491”制备而得)、叔丁醇钾(404.07mg,3.6mmol)和粗品化合物1f(1000mg,3.6mmol),60℃条件下,反应16小时。反应液冷却至室温,滴加1M盐酸至反应液pH至2~3,用乙酸乙酯萃取(50mL×3),合并有机相,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相制备(Gilson-281,洗脱体系:乙腈,水)纯化所得残余物,制得到标题化合物4b(350mg,产率:20.5%)。
MS m/z(ESI):426.4[M+1]
第二步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸甲酯4d
依次将化合物4b(350mg,0.82mmol)、对氨基苯甲酸甲酯4c(39.23mg,0.26mmol,采用公知的方法“Chemical Communications(Cambridge,United Kingdom),2015,51(58),11705-11708”制备而得)和N,N-二异丙基乙胺(0.57mL,3.29mmol)溶于30mL乙酸乙酯中,再滴加2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,1569mg,2.47mmol),加毕,反应升温至60℃,搅拌反应2小时。反应液冷却至室温,减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题化合物4d(140mg,产率:28.9%)。
MS m/z(ESI):559.5[M+1]
第三步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸4
将化合物4d(120mg,0.21mmol)溶于4mL四氢呋喃和甲醇的混合溶剂(V/V=3:1),加入1.28mL 1M的氢氧化锂溶液,加毕,反应16小时。滴加10%盐酸至反应液pH至3~4,用乙酸乙酯萃取(50mL×2),合并有机相,有机相减压浓缩,用高效液相制备(Gilson-281,洗脱体系:乙腈,水)纯化所得残余物,制得到标题化合物4(50mg,产率:42.7%)。
MS m/z(ESI):545.4[M+1]
1H NMR(400MHz,DMSO-d6)δ10.81(s,1H),7.92(s,1H),7.90(s,1H),7.83-7.81(d,1H),7.74(s,1H),7.72(s,1H),7.62-7.59(dd,1H),7.43(s,1H)7.38(s,1H),7.30-7.25(m,4H),7.21-7.17(m,1H),6.31(s,1H),6.05-6.01(m,1H),3.54(s,3H),3.49-3.44(m,2H),2.37(s,3H).
实施例5,6
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸5
(R)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸6
Figure PCTCN2017099579-appb-000069
将4(900mg,1.65mmol)进行手性制备(分离条件:手性制备柱Superchiral S-AD(Chiralway),2cm I.D.*25cm Length,5um;流动相:二氧化碳:乙醇:二乙胺=60:40:0.05,流速:50g/min),收集其相应组分,减压浓缩,得到标题产物5(421mg)和6(405mg)。
化合物5:
MS m/z(ESI):545.4[M+1];
手性HPLC分析:保留时间4.138分钟,手性纯度:98%(色谱柱:Superchiral S-AD(Chiralway),2cm I.D.*25cm Length,5um;流动相:乙醇/正己烷/三氟乙酸=50/50/0.05(v/v/v))。
1H NMR(400MHz,DMSO-d6)δ10.81(s,1H),7.92(s,1H),7.90(s,1H),7.83-7.81(d,1H),7.74(s,1H),7.72(s,1H),7.62-7.59(dd,1H),7.43(s,1H)7.38(s,1H),7.30-7.25(m,4H),7.21-7.17(m,1H),8.31(s,1H),6.05-6.01(m,1H),3.54(s,3H), 3.49-3.44(m,2H),2.37(s,3H)
化合物6:
MS m/z(ESI):545.4[M+1];
手性HPLC分析:保留时间1.475分钟,(色谱柱:Superchiral S-AD(Chiralway),2cm I.D.*25cm Length,5um;流动相:乙醇/正己烷/三氟乙酸=50/50/0.05(v/v/v))。
1H NMR(400MHz,DMSO-d6)δ10.81(s,1H),7.92(s,1H),7.90(s,1H),7.83-7.81(d,1H),7.74(s,1H),7.72(s,1H),7.62-7.59(dd,1H),7.43(s,1H)7.38(s,1H),7.30-7.25(m,4H),7.21-7.17(m,1H),8.31(s,1H),6.05-6.01(m,1H),3.54(s,3H),3.49-3.44(m,2H),2.37(s,3H)
实施例7
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(噻吩-3-基)丙酰氨基)苯甲酸7
Figure PCTCN2017099579-appb-000070
第一步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)乙酸叔丁酯7b
依次将1f(3.4g,12.24mmol)、碳酸铯(11937.34mg,36.73mmol)和2-溴代乙酸叔丁酯7a(3.58g,18.37mmol,采用公知的方法“Chemical Communications(Cambridge,United Kingdom),2012,48(22),2803-2805”制备而得)溶于40mL N,N-二甲基甲酰胺,加毕,反应液温度升至65℃,搅拌反应2小时。冷却反应液温度至室温,反应液中加入50mL水,用乙酸乙酯萃取(50mL×3),有机相用饱和氯化钠溶液洗涤(50mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系I纯化所得残余物得标题产物7b(3.2g,产率:65.4%)。
MS m/z(ESI):392.1[M+1];
第二步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(噻吩-3-基)丙酸叔丁酯7d
依次将7b(100mg,0.26mmol)和3-溴甲基噻吩7c(90.37mg,0.51mmol,采用公知的方法“Journal of Organic Chemistry,2016,81(22),11035-11042”制备而得)溶于10mL四氢呋喃中,反应液冷却至-78℃,滴加二异丙基氨基锂溶液(1.53mL,1.02mmol),反应2小时,缓慢加入1mL水,反应液温度自然升至室温。向反应液中加10mL水,乙酸乙酯萃取(20mL×3),有机相用饱和氯化钠溶液洗涤(20mL×2),过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物得标题产物7d(84mg,产率:64.1%)。
MS m/z(ESI):488.4[M+1];
第三步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(噻吩-3-基)丙酸7e
将7d(80mg,0.16mmol)溶于4mL二氯甲烷中,滴加三氟乙酸(0.5mL,0.78mmol),搅拌反应5小时。反应液减压蒸馏,得粗品标题产物7e(68mg),产物不经纯化直接投入下一步反应。
MS m/z(ESI):432.3[M+1];
第四步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(噻吩-3-基)丙酰氨基)苯甲酸甲酯7f
将粗品7e(67mg,0.16mmol)和4(30.48mg,0.20mmol)溶于6mL乙酸乙酯,依次滴加0.5mL吡啶和2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物(197.44mg,0.62mmol),加毕,反应升温至70℃,搅拌反应1.5小时。反应液中加入15mL水,乙酸乙酯萃取(15mL×2),用饱和氯化钠溶液洗涤(15mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物得标题产物7f(80mg),产率:86.7%。
MS m/z(ESI):565.5[M+1];
第五步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(噻吩-3-基)丙酰氨基)苯甲酸7
将7f(80mg,0.13mmol)溶于3mL四氢呋喃,再向反应液中加入氢氧化钠溶液(1N,0.67mL),加毕,反应12小时。再加入氢氧化钠溶液(1N,0.67mL),反应液升温至35℃,搅拌16小时。减压蒸馏旋除有机溶剂,加入15mL水,滴加3N盐酸至反应液pH至4~5,过滤。收集滤饼,滤饼用硅胶柱色谱法以洗脱剂体系A纯化所得残余物得标题产物7(50mg,产率:64.8%)。
MS m/z(ESI):551.1[M+1]
1H NMR(400MHz,DMSO-d6)δ12.76(s,1H),10.81(s,1H),7.92(d,2H),7.84(d,1H),7.75(d,2H),7.62(dd,1H),7.45(dd,1H),7.40(d,2H),7.22(d,1H),7.01(d,1H),6.34(s,1H),5.99-5.95(m,1H),3.58-3.52(m,1H),3.53(s,3H),3.46-3.41(m,1H),2.41(s,3H)
实施例8
4-(2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸8
Figure PCTCN2017099579-appb-000071
第一步
1-(2-溴-4-氯苯基)丙烷-1-酮8c
将2-溴-4-氯-1-碘苯8a(1.0g,3.15mmol,采用公知的方法“Angewandte Chemie,International Edition,2010,49(46),8729-8732”制备而得)溶于1mL四氢呋喃中,冷却至-20℃,加入异丙基氯化镁(421.15mg,4.10mmol),预先反应1小时。将丙酰氯8b(378.89mg,4.10mmol)、氯化锂(11.42mg,189.00μmol)、氯化亚铜(9.36mg,94.50μmol)和三氯化铝(12.61mg,94.50μmol)加入1mL四氢呋喃中,室温搅拌均匀,将预先反应了1小时的反应液滴加到以上混合液中,室温反应2小时。反应液中加入20mL饱和氯化铵溶液淬灭反应,用二氯甲烷萃取(20mL×3),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用CombiFlash快速制备仪以洗脱剂B纯化所得残余物,制得标题化合物8c(640mg,产率:82.0%)
第二步
1-(4-氯-2-(2,5-二甲氧基吡啶-4-基)苯基)丙烷-1-酮8d
将化合物8c(640mg,2.59mmol)、化合物1d(520.41mg,2.84mmol)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(191.8mg,0.259mmol)和碳酸钠(822.16mg,7.76mmol)加入到20mL 1,4-二氧六环和4mL水的混合溶剂中,加毕,反应液加热至85℃,搅拌反应16小时。反应液自然冷却至室温,加入20mL水,用乙酸乙酯萃取(20mL×3),合并有机相,有机相分别用水(30mL),饱和氯化钠溶液(30mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物8d(600mg,产率:75.9%)。
MS m/z(ESI):306.0[M+1]
第三步
4-(5-氯-2-丙酰基苯基)-5-甲氧基吡啶-2(1H)-酮8e
将化合物8d(600mg,1.96mmol)加入到10mL N,N-二甲基甲酰胺中,加入吡啶氢溴酸盐(1.51g,9.81mmol),加毕,加热至100℃,搅拌反应3小时。反应液冷却至室温,减压浓缩除去有机溶剂,所得残余物中加入30mL水,用二氯甲烷萃取(20mL×3),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品标题化合物8e(550mg),产物不经纯化直接下一步反应。
MS m/z(ESI):292.0[M+1]
第四步
2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)乙酸叔丁酯8f
将粗品化合物8e(550mg,1.89mmol)、碳酸铯(1.84g,5.67mmol)和化合物7a(551.61mg,2.83mmol)溶于10mL N,N-二甲基甲酰胺,加毕,反应液温度升至65℃,搅拌反应2小时。冷却反应液温度至室温,加入30mL水,用乙酸乙酯萃取(30mL×3),合并有机相,用饱和氯化钠溶液洗涤(30mL×3)无水硫酸钠干燥,过滤,滤液减压浓缩,用CombiFlash快速制备仪以洗脱剂B纯化所得残余物,得到标题化合物8f(350mg,产率:51.0%)。
MS m/z(ESI):405.4[M+1]
第五步
2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酸叔丁酯8h
将化合物8f(122mg,302.37μmol)溶于10mL四氢呋喃中,冷却至-78℃,加入苄溴8g(103.43mg,604.74μmol),再滴加入二(三甲基硅基)氨基锂的四氢呋喃溶液(1.21mL,1.21mmol),反应2小时。反应液温度自然升至室温,向反应液中加入10mL水,乙酸乙酯萃取(20mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物8h(75mg,产率:50.0%)。
MS m/z(ESI):496.2[M+1]
第六步
2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯丙酸8i
将化合物8h(75mg,0.15mmol)溶于4mL二氯甲烷中,滴加三氟乙酸(0.5mL), 搅拌反应5小时。反应液减压浓缩,得粗品标题化合物8i(70mg),产物不经纯化直接投入下一步反应。
MS m/z(ESI):439.9[M+1]
第七步
4-(2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸8
将粗品化合物8i(70mg,159.13μmol)和4-氨基苯甲酸8j(32.73mg,237.70μmol,采用公知的方法“Angewandte Chemie-International Edition,2012,51(34),8564-8567”制备而得)溶于20mL乙酸乙酯中,依次滴加三乙胺(64.41mg,636.53μmol)和2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,303.80mg,477.39μmol),加毕,反应升温至60℃,搅拌反应2小时。反应液冷却至室温,加入15mL水,用乙酸乙酯萃取(15mL×2),用饱和氯化钠溶液洗涤(15mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,制得标题化合物8(30mg,产率:35.7%)。
MS m/z(ESI):559.4[M+1]
1H NMR(400MHz,CDCl3)δ9.97(s,1H),8.10(d,2H),7.86(d,2H),7.70(d,1H),7.51-7.24(m,8H),6.64(s,1H),6.26(s,1H),3.67-3.62(m,4H),3.33-3.29(m,1H),2.86(s,2H),1.18-0.92(m,3H).
实施例9,10
(S)-4-(2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸9
(R)-4-(2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸10
Figure PCTCN2017099579-appb-000072
将化合物8(1g,1.79mmol)进行手性制备(分离条件:手性制备柱CHIRAL PAK IE 20*250mm 5um;流动相:正己烷:乙醇=55:45,流速:7mL/min),收集其相应组分,减压浓缩,得到标题化合物9(300mg)和化合物10(400mg)。
化合物9:
MS m/z(ESI):559.5[M+1]
手性HPLC分析:保留时间11.267分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:乙醇/正己烷=50/50(v/v))。
1H NMR(400MHz,CDCl3)δ9.88(s,1H),8.10(d,2H),7.85(d,2H),7.69(d,1H),7.48(d,1H),7.40(s,7H),6.62(s,1H),6.22(s,1H),3.65(s,3H),3.60(s,1H),3.31(s,1H),2.85(s,2H),1.15(s,3H).
化合物10:
MS m/z(ESI):559.5[M+1]
手性HPLC分析:保留时间4.836分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:乙醇/正己烷=50/50(v/v))。
1H NMR(400MHz,CDCl3)δ9.88(s,1H),8.10(d,2H),7.85(d,2H),7.69(d,1H),7.48(d,1H),7.40(s,7H),6.62(s,1H),6.22(s,1H),3.65(s,3H),3.60(s,1H),3.31(s,1H),2.85(s,2H),1.15(s,3H).
实施例11
(S)-4-(2-(4-(2-乙酰-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酰胺11
Figure PCTCN2017099579-appb-000073
将化合物5(54mg,99.09μmol)溶于5mL N,N-二甲基甲酰胺中,加入碳酸铵(95.21mg,990.86μmol)、N,N-二异丙基乙胺(64.03mg,495.43μmol)和O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(112.96mg,297.26μmol),室温搅拌反应16小时。反应液中加入20mL饱和碳酸氢钠溶液,用乙酸乙酯萃取(50mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,制得标题化合物11(40mg,产率:74.2%)。
MS m/z(ESI):544.2[M+1]
1H NMR(400MHz,CD3OD)δ7.87-7.82(m,3H),7.70-7.69(m,1H),7.68-7.66(m,1H),7.56-7.54(dd,1H),7.36(s,1H),7.32-7.31(d,1H),7.29-7.25(m,4H),7.24-7.19(m,1H),6.41(s,1H),5.89-5.85(m,1H),3.65-3.60(m,1H),3.59(s,3H),3.50-3.45(m,1H),2.46(s,3H).
实施例12
(S)-(4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯基)氨基甲酸甲酯12
Figure PCTCN2017099579-appb-000074
化合物5(100mg,183.83μmol)溶于8mL甲苯中,依次加入三乙胺(65.1mg,643.39μmol)、叠氮磷酸二苯酯(60.71mg,220.59μmol)和甲醇(58.9mg,1.84mmol),升温至100℃,搅拌反应2小时。冷却反应液至室温,减压浓缩除去溶剂,所得残余物中加入15mL饱和碳酸氢钠溶液,用乙酸乙酯萃取(50mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A 纯化所得残余物,制得标题化合物12(75mg,产率:71.2%)。
MS m/z(ESI):574.2[M+1]
1H NMR(400MHz,CDCl3)δ7.68-7.66(d,1H),7.48-7.46(d,3H),7.29-7.21(m,8H),7.15-7.10(m,1H),6.61-6.50(m,2H),5.95-5.85(m,1H),3.76(s,3H),3.65-3.60(m,1H),3.59(s,3H),3.30-3.20(m,1H),2.42(s,3H).
实施例13
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)-N-甲基苯甲酰胺13
Figure PCTCN2017099579-appb-000075
将化合物5(70mg,128.44μmol)溶于5mLN,N-二甲基甲酰胺中,依次加入甲胺(11.97mg,385.33μmol)、N,N-二异丙基乙胺(66.4mg,513.78μmol)和O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(97.62mg,256.89μmol),室温搅拌反应16小时。反应液中加入50mL乙酸乙酯,用水洗涤(30mL×2),有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相制备(Waters 2767-SQ detecor2,洗脱体系:乙腈,水)纯化所得残余物,制得标题化合物13(45mg,产率:62.8%)。
MS m/z(ESI):558.1[M+1]
1H NMR(400MHz,DMSO-d6)δ10.75(s,1H),8.39-8.34(m,1H),7.84-7.82(m,3H),7.71(d,2H),7.62(dd,1H),7.44(s,1H),7.38(s,1H),7.31-7.26(m,4H),7.22-7.18(m,1H),6.31(s,1H),6.04-6.01(m,1H),3.54(s,3H),3.49-3.39(m,2H),2.77(d,3H),2.38(s,3H).
实施例14,15
(R)-((乙氧羰基)氧基)甲基4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸酯14
(S)-((乙氧羰基)氧基)甲基4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸酯15
Figure PCTCN2017099579-appb-000076
Figure PCTCN2017099579-appb-000077
第一步
((乙氧羰基)氧基)甲基4-((叔丁氧羰基)氨基)苯甲酸酯14c
将4-((叔丁氧羰基)氨基)苯甲酸14a(4g,16.86mmol,采用公知的方法“Journal of Medicinal Chemistry,2016,59(22),10299-10314”制备而得)、碘化钾(2.24g,13.49mmol)和碳酸钾(2.33g,16.86mmol)溶于50mL N,N-二甲基甲酰胺中,氩气氛下,加入氯甲基乙基碳酸酯14b(3.5g,25.29mmol,采用公知的方法“Tetrahedron Letters,2007,48(1),109-112”制备而得),加毕,反应升温至50℃,搅拌反应16小时。反应液冷却至室温,倒入100mL冰水中,用乙酸乙酯萃取(60mL×3),合并有机相,用25mL饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系C纯化所得残余物,制得标题化合物14c(5.3g,产率:88.0%)。
MS m/z(ESI):340.5[M+1]
第二步
((乙氧基羰基)氧基)甲基4-氨基苯甲酸酯盐酸盐14d
将氯化氢的1,4-二氧六环溶液(13.3mL,66.52mmol)加入到13mL四氢呋喃中,加入化合物14c(2.7g,7.56mmol),加毕,反应升温至50℃,搅拌反应5小时。反应液冷却至室温,减压浓缩除去溶剂,残余物中加入20mL乙酸乙酯和正己烷(V/V=1:9)的混合溶剂,搅拌,过滤,收集滤饼,得粗品标题化合物14d(2g),产物不经纯化直接下一步反应。
MS m/z(ESI):240.4[M+1]
第三步
((乙氧羰基)氧基)甲基4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸酯14e
将化合物4b(250mg,0.59mmol)溶于50mL乙酸乙酯中,加入N,N-二异丙基 乙胺(303.48mg,2.35mmol)、粗品化合物14d(178.03mg,0.65mmol)和2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,747.14mg,1.17mmol),加毕,反应升温至60℃,搅拌反应2小时。反应液冷却至室温,加入25mL饱和碳酸氢钠溶液,用乙酸乙酯萃取(50mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相制备(Gilson-281,洗脱体系:乙腈,水)纯化所得残余物,制得标题化合物14e(230mg,产率:60.6%)。
MS m/z(ESI):647.5[M+1]
第四步
(R)-((乙氧羰基)氧基)甲基4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸酯14
(S)-((乙氧羰基)氧基)甲基4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸酯15
将化合物14e(230mg,0.36mmol)进行手性制备(分离条件:色谱柱:Superchiral S-AD(Chiralway),2cm I.D.*25cm Length,5um;流动相:二氧化碳:异丙醇=60:40,流速:50g/min),收集其相应组分,减压浓缩,得到标题化合物14(84mg)和化合物15(76mg)。
化合物14:
MS m/z(ESI):647.5[M+1]
手性HPLC分析:保留时间5.297分钟,(色谱柱:CHIRAL PAK IE,4.6*150mm,5um;流速:1mL/min;流动相:乙醇)。
1H NMR(400MHz,CDCl3)δ9.70(s,1H),8.03(s,1H),8.01(s,1H),7.70-7.69(d,1H),7.64(s,1H),7.62(s,1H),7.48-7.46(dd,1H),7.30-7.27(m,4H),7.26-7.22(m,2H),7.05-7.02(m,1H),6.57(s,1H),5.99(s,2H),5.95-5.85(m,1H),4.29-4.24(m,2H),3.75-3.65(m,1H),3.59(s,3H),3.35-3.25(m,1H),2.44(s,3H),1.35-1.31(m,3H).
化合物15:
MS m/z(ESI):647.5[M+1]
手性HPLC分析:保留时间8.442分钟,(色谱柱:CHIRAL PAK IE,4.6*150mm,5um;流速:1mL/min;流动相:乙醇)。
1H NMR(400MHz,CDCl3)δ9.70(s,1H),8.03(s,1H),8.01(s,1H),7.70-7.69(d,1H),7.64(s,1H),7.62(s,1H),7.48-7.46(dd,1H),7.30-7.27(m,4H),7.26-7.22(m,2H),7.05-7.02(m,1H),6.57(s,1H),5.99(s,2H),5.95-5.85(m,1H),4.29-4.24(m,2H),3.75-3.65(m,1H),3.59(s,3H),3.35-3.25(m,1H),2.44(s,3H),1.35-1.31(m,3H).
实施例16
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-(环丙烷基甲酰氨基)苯基)丙酰氨基)苯甲酸16
Figure PCTCN2017099579-appb-000078
第一步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-硝基苯基)丙酸叔丁酯16b
依次将化合物7b(150mg,0.38mmol)和1-(溴甲基)-4-硝基苯16a(165.39mg,0.77mmol,采用公知的方法“Angewandte Chemie-International Edition,2014,53(50),13835-13839”制备而得)溶于10mL四氢呋喃中,反应液冷却至-78℃,滴加二(三甲基硅基)氨基锂溶液(1.53mL,1.53mmol),反应2小时。-78℃下向反应液中缓慢加入1mL水淬灭反应,自然升至室温,加10mL水,用乙酸乙酯萃取(20mL×2),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物16b(180mg,产率:80.3%)。
MS m/z(ESI):527.4[M+1]
第二步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-硝基苯基)丙酸16c
将化合物16b(180mg,0.34mmol)溶于5mL二氯甲烷中,滴加三氟乙酸(0.5mL),搅拌反应5小时。反应液减压浓缩,得粗品标题化合物16c(166mg),产物不经纯化直接投入下一步反应。
MS m/z(ESI):471.4[M+1]
第三步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-硝基苯基)丙酰氨基)苯甲酸甲酯16d
将粗品化合物16c(161mg,0.34mmol)和化合物4c(67.19mg,0.44mmol)溶于6mL乙酸乙酯,依次滴加0.5mL吡啶和2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,870.36mg,1.37mmol),加毕,反应升温至70℃,搅拌反应1.5小时。反应液冷却至室温,加入15mL水,用乙酸乙酯萃取(15mL×2),合并有机相,用饱和氯化钠溶液洗涤(15mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题化合物16d(130mg,产率:53.5%)。
MS m/z(ESI):604.4[M+1]
第四步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-氨基苯基)丙酰氨基)苯甲酸甲酯16e
将粗品化合物16d(70mg,115.89μmol)溶于8mL四氢呋喃中,加入二氧化铂(5.26mg,23.18μmol),氢气置换2次,室温搅拌反应2.5小时。反应液过滤,滤液减压浓缩,得粗品标题化合物16e(66mg),产物不经纯化直接投入下一步反应。
第五步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-(环丙烷基甲酰氨基)苯基)丙酰氨基)苯甲酸甲酯16g
将粗品化合物16e(66mg,114.98μmol)和三乙胺(290.87mg,2.87mmol)溶于8mL四氢呋喃中,加入环丙甲酰氯(240.38mg,2.30mmol),搅拌反应16小时。反应液减压浓缩,所得残余物中加入15mL水,用乙酸乙酯萃取(15mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题化合物16g(40mg,产率:54.2%)。
MS m/z(ESI):642.1[M+1]
第六步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-(环丙烷基甲酰氨基)苯基)丙酰氨基)苯甲酸16
将化合物16g(40mg,62.3μmol)溶于3mL甲醇中,加入氢氧化钠(12.46mg,311.48μmol),升温至50℃搅拌反应5小时。反应液冷却至室温,加入15mL水,滴加3M盐酸至pH为5,用乙酸乙酯萃取(20mL×2),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相制备(Waters 2767-SQ detecor2,洗脱体系:乙腈,水)纯化所得残余物,制得标题化合物16(16mg,产率:40.9%)。
MS m/z(ESI):628.5[M+1]
1H NMR(400MHz,DMSO-d6)δ10.84(s,1H),10.12(s,1H),7.92(d,2H),7.83(d,1H),7.76(d,2H),7.61(dd,1H),7.48(d,2H),7.41(d,2H),7.18(d,2H),6.30(s,1H),6.00-5.96(m,1H),3.55(s,3H),3.42-3.36(m,2H),2.39(s,3H),1.76-1.70(m,1H),0.76-0.74(m,4H).
实施例17
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4'-氰基-2'-甲基-[1,1'-联苯]-4-基)丙酰氨基)苯甲酸17
Figure PCTCN2017099579-appb-000079
第一步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-溴苯基)丙酸叔丁酯17b
依次将化合物7b(150mg,0.38mmol)和1-溴-4-(溴甲基)苯17a(191.35mg,0.77mmol,采用公知的方法“Tetrahedron Letters,2016,57(2),168-171”制备而得)溶于10mL四氢呋喃中,冷却至-78℃,滴加二(三甲基硅基)氨基锂溶液(1.53mL,1.53mmol),反应2小时。-78℃下向反应液中缓慢加入1mL水淬灭反应,自然升至室温,加入10mL水,用乙酸乙酯萃取(20mL×2),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物17b(180mg,产率:79.6%)。
MS m/z(ESI):562.3[M+1]
第二步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-溴苯基)丙酸17c
将化合物17b(180mg,0.30mmol)溶于5mL二氯甲烷中,滴加三氟乙酸(0.5mL),搅拌反应5小时。反应液减压浓缩,得粗品标题化合物17c(160mg),产物不经纯化直接投入下一步反应。
MS m/z(ESI):506.3[M+1]
第三步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-溴苯基)丙酰氨基)苯甲酸甲酯17d
将粗品化合物17c(154mg,0.31mmol)和化合物4c(59.95mg,0.40mmol)溶于6mL乙酸乙酯中,依次滴加0.5mL吡啶和2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,776.60mg,1.22mmol),加毕,升温至70℃,搅拌反应1.5小时。反应液中加入15mL水,乙酸乙酯萃取(15mL×2),合并有机相,用饱和氯化钠溶液洗涤(15mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题化合物17d(180mg,产率:90.6%)。
MS m/z(ESI):639.3[M+1]
第四步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4'-氰基-2'-甲基-[1,1'-联苯]-4-基)丙酰氨基)苯甲酸甲酯17f
将化合物17d(180mg,0.28mmol)、(4-氰基-2-甲基苯基)硼酸17e(90.84mg,0.56mmol,采用公知的方法“Tetrahedron,2011,67(52),10082-10088”制备而得)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(20.65mg,0.03mmol)和碳酸钠(89.73mg,0.85mmol)加入到甲苯(8mL)、乙醇(3mL)和水(1mL)的混合溶剂中,加毕,升温至85℃,搅拌反应16小时。反应液自然冷却至室温,加入15mL水,用乙酸乙酯萃取(20mL×2),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物17f(200mg,产率:31.5%)。
MS m/z(ESI):674.5[M+1]
第五步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4'-氰基-2'-甲基-[1,1'-联苯]-4-基)丙酰氨基)苯甲酸17
将化合物17f(200mg,0.297mmol)溶于2mL甲醇和2mL四氢呋喃的混合溶剂中,向反应液中加入一水合氢氧化锂(29.9mg,0.71mmol),搅拌反应60小时。反应液中加入15mL水,滴加3M盐酸至pH为4~5,用乙酸乙酯萃取(20mL×2),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相制备(Waters 2767-SQ detecor2,洗脱体系:乙腈,水)纯化所 得残余物,制得标题化合物17(10mg,产率:16.2%)。
MS m/z(ESI):660.5[M+1]
1H NMR(400MHz,DMSO-d6)δ10.81(s,1H),7.92(d,2H),7.83(d,1H),7.78-7.74(m,3H),7.70(dd,1H),7.61(dd,1H),7.43-7.35(m,5H),7.29(d,2H),6.33(s,1H),6.06-6.02(m,1H),3.61-3.50(m,2H),3.54(s,3H),2.38(s,3H),2.22(s,3H).
实施例18
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(2-甲基-2H-吲唑-5-基)-3-苯基丙酰胺18
Figure PCTCN2017099579-appb-000080
将化合物4b(90mg,211.34μmol)、2-甲基-5-氨基-2H-吲唑18a(34.21mg,232.47μmol,采用公知的方法“Journal of the American Chemical Society,2016,138(14),4730-4738”制备而得)和N,N-二异丙基乙胺(273.14mg,2.11mmol)加入15mL乙酸乙酯中,再滴加2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,537.94mg,845.36μmol),加毕,升温至75℃,搅拌反应2小时。反应液冷却至室温,加入30mL水,滴加3M盐酸至pH为5,分液,水相用乙酸乙酯萃取(30mL×2),合并有机相,用饱和氯化钠溶液洗涤(35mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题化合物18(80mg,产率:68.2%)。
MS m/z(ESI):555.0[M+1]
1H NMR(400MHz,DMSO-d6)δ10.49(s,1H),8.27(s,1H),8.16(s,1H),7.83(d,1H),7.61(dd,1H),7.56(d,1H),7.49(s,1H),7.38(s,1H),7.31-7.25(m,5H),7.21-7.17(m,1H),6.31(s,1H),6.07-6.03(m,1H),4.13(s,3H),3.55(s,3H),3.46(d,2H),2.38(s,3H).
实施例19,20
(R)-2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(2-甲基-2H-吲唑-5-基)-3-苯基丙酰胺19
(S)-2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(2-甲基-2H-吲唑-5-基)-3-苯基丙酰胺20
Figure PCTCN2017099579-appb-000081
将化合物18(75mg,135.13μmol)进行手性制备(分离条件:手性制备柱Lux Cellulose-1OD 21.2*250mm 5um;流动相:正己烷:乙醇=60:40,流速:8mL/min),收集其相应组分,减压浓缩,得到标题化合物19(28mg)和化合物20(27mg)。
化合物19:
MS m/z(ESI):555.5[M+1]
手性HPLC分析:保留时间5.816分钟,手性纯度:100%(色谱柱:Lux Cellulose-1 OD 4.6*150mm 5um(带保护柱);流动相:乙醇/正己烷=30/70(v/v))。
1H NMR(400MHz,DMSO-d6)δ10.48(s,1H),8.27(s,1H),8.15(s,1H),7.82(d,1H),7.61(d,1H),7.56(d,1H),7.48(s,1H),7.38(s,1H),7.31-7.25(m,5H),7.21-7.17(m,1H),6.31(s,1H),6.07-6.03(m,1H),4.13(s,3H),3.55(s,3H),3.45(d,2H),2.38(s,3H).
化合物20:
MS m/z(ESI):555.5[M+1]
手性HPLC分析:保留时间10.287分钟,手性纯度:100%(色谱柱:Lux Cellulose-1 OD 4.6*150mm 5um(带保护柱);流动相:乙醇/正己烷=30/70(v/v))。
1H NMR(400MHz,DMSO-d6)δ10.48(s,1H),8.27(s,1H),8.15(s,1H),7.83(d,1H),7.61(d,1H),7.56(d,1H),7.48(s,1H),7.38(s,1H),7.31-7.25(m,5H),7.21-7.17(m,1H),6.31(s,1H),6.07-6.03(m,1H),4.13(s,3H),3.55(s,3H),3.45(d,2H),2.38(s,3H).
实施例21
4-(2-(4-(2-丁酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸21
Figure PCTCN2017099579-appb-000082
采用实施例8的合成路线,将原料化合物8b替换为正丁酰氯,制得标题化合物21(95mg)。
MS m/z(ESI):573.2[M+1]
1H NMR(400MHz,CDCl3)δ10.00(s,1H),8.10-8.00(d,2H),7.83-7.80(d,2H),7.69-7.67(d,1H),7.50-7.45(dd,1H),7.34-7.25(m,7H),6.65(s,1H),6.29-6.19(s,3H),3.64-3.58(m,4H),3.30-3.22(m,1H),2.80-2.70(m,2H),1.70-1.60(m,2H),0.94-0.89(m,3H).
实施例22
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)-2-氟苯甲酰胺22
Figure PCTCN2017099579-appb-000083
采用实施例18的合成路线,将原料化合物18a替换为4-氨基-2-氟苯甲酰胺(采用专利申请“WO 2013146963”公开的方法制备而得),制得标题化合物22(30mg)。
MS m/z(ESI):562.5[M+1]
1H NMR(400MHz,DMSO-d6)δ10.86(s,1H),7.80(d,1H),7.67-7.52(m,5H),7.38-7.37(m,3H),7.26-7.25(m,4H),7.05-7.04(m,1H),6.29(s,1H),5.96-5.93(m,1H),3.51(s,3H),3.46-3.41(m,2H),2.36(s,3H).
实施例23
2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基-N-(喹唑啉-6-基)丙酰胺23
Figure PCTCN2017099579-appb-000084
将化合物8i(90mg,204.6μmol)、6-氨基喹唑啉23a(32.67mg,225.06μmol,采用公知的方法“Bioorganic&Medicinal Chemistry Letters,2015,25(4),803-806”制备而得)和N,N-二异丙基乙胺(264.42mg,2.05mmol)加入15mL乙酸乙酯中,再滴加2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,537.94mg,845.36μmol),加毕,反应升温至75℃,搅拌反应2小时。反应液冷却至室温,加入30mL水,滴加3M盐酸至反应液pH至5,分液,水相用乙酸乙酯萃取(30mL×2),合并有机相,用饱和氯化钠溶液洗涤(35mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题化合物23(50mg,产率:43.1%)。
MS m/z(ESI):567.5[M+1]
1H NMR(400MHz,DMSO-d6)δ11.04(s,1H),9.59(s,1H),9.22(s,1H),8.63(s,1H),8.04(q,2H),7.81(d,1H),7.60(d,1H),7.43(s,1H),7.37(s,1H),7.31-7.27(m,4H),7.22-7.20(m,1H),6.32(s,1H),6.07-6.03(m,1H),3.53(s,3H),3.51-3.48(m,2H),2.85-2.67(m,2H),0.98(t,3H).
实施例24
4-(2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酰胺24
Figure PCTCN2017099579-appb-000085
采用实施例23的合成路线,将原料化合物23a替换为4-氨基苯甲酰胺(采用公知的方法“Chemical Communications(Cambridge,United Kingdom),2017,53(35),4807-4810”制备而得),制得标题化合物24(150mg)。
MS m/z(ESI):558.1[M+1]
1H NMR(400MHz,CD3OD)δ7.89-7.86(m,2H),7.82-7.81(m,1H),7.77-7.70(m,2H),7.58-7.55(m,1H),7.34-7.29(m,7H),7.25-7.23(m,1H),6.43(s,1H),5.92-5.89(m,1H),3.63-3.58(m,1H),3.57(s,3H),3.47-3.41(m,2H),1.12-1.09(m,3H).
实施例25,26
(R)-4-(2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酰胺25
(S)-4-(2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酰胺26
Figure PCTCN2017099579-appb-000086
将化合物24(150mg,268.81μmol)进行手性制备(分离条件:手性制备柱CHIRALPAK IF 250*20mm;流动相:A正己烷:B乙醇=60:40,流速:7.0mL/min),收集其相应组分,减压浓缩,得到标题化合物25(50mg)和化合物26(50mg)。
化合物25:
MS m/z(ESI):558.5[M+1]
手性HPLC分析:保留时间6.587分钟,(色谱柱:Lux Amylose-2(AY)4.6*150mm5um(带保护柱);流动相:乙醇/正己烷=20/80(v/v))。
1H NMR(400MHz,MeOH-d4)δ7.89-7.86(m,2H),7.82-7.81(m,1H),7.77-7.70(m,2H),7.58-7.55(m,1H),7.34-7.29(m,7H),7.25-7.23(m,1H),6.43(s,1H),5.92-5.89(m,1H),3.63-3.58(m,1H),3.57(s,3H),3.47-3.41(m,2H),1.12-1.09(m,3H).
化合物26:
MS m/z(ESI):558.4[M+1]
手性HPLC分析:保留时间8.966分钟,(色谱柱:Lux Amylose-2(AY)4.6*150mm5um(带保护柱);流动相:乙醇/正己烷=20/80(v/v))。
1H NMR(400MHz,CD3OD)δ7.89-7.86(m,2H),7.82-7.81(m,1H),7.77-7.70(m,2H),7.58-7.55(m,1H),7.34-7.29(m,7H),7.25-7.23(m,1H),6.43(s,1H),5.92-5.89(m,1H),3.63-3.58(m,1H),3.57(s,3H),3.47-3.41(m,2H),1.12-1.09(m,3H).
实施例27
2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(1H-吲唑-6-基)-3-苯基丙酰胺27
采用实施例23的合成路线,将原料化合物23a替换为6-氨基吲唑(采用公知的方法“Tetrahedron Letters,2010,51(5),786-789”制备而得),制得标题化合物27(45mg)。
MS m/z(ESI):555.5[M+1]
1H NMR(400MHz,DMSO-d6)δ12.95(s,1H),10.68(s,1H),8.14(s,1H),7.99(s,1H),7.80(d,1H),7.70(d,1H)7.60(d,1H),7.45(s,1H),7.38(s,1H),7.30-7.26(m,4H),7.21-7.16(m,2H),6.30(s,1H),6.07-6.03(m,1H),3.53(s,3H),3.50-3.47(m,2H),2.85-2.67(m,2H),0.97(t,3H).
实施例28
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(3-氰基-1H-吲哚-6-基)-3-苯基丙酰胺28
Figure PCTCN2017099579-appb-000088
采用实施例18的合成路线,将原料化合物18a替换为6-氨基-1H-吲哚-3-甲腈(采用专利申请“WO 20160271105”公开的方法制备而得),制得标题化合物28(30mg)。
MS m/z(ESI):565.0[M+1]
1H NMR(400MHz,CD3OD)δ8.03(s,1H),7.91(s,1H),7.82(d,1H),7.55(t,2H),7.41(s,1H),7.31-7.15(m,7H),6.42(s,1H),5.96-5.93(m,1H),3.58(s,3H),3.43-3.38(m,2H),2.44(s,3H).
实施例29
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)-3-氟 苯甲酸29
Figure PCTCN2017099579-appb-000089
采用实施例4的合成路线,将第二步原料化合物4c替换为4-氨基-3-氟苯甲酸甲酯(采用专利申请“WO 2012087519”公开的方法制备而得),制得标题化合物29(20mg)。
MS m/z(ESI):563.4[M+1]
1H NMR(400MHz,DMSO-d6)δ10.65(s,1H),8.15-8.11(m,1H),7.85-7.80(d,1H),7.79-7.72(m,2H),7.61-7.59(dd,1H),7.38-7.37(d,2H),7.34-7.32(d,2H),7.29-7.25(m,2H),7.20-7.17(m,1H),6.31(s,1H),6.23-6.19(m,1H),3.57-3.45(m,5H),2.36(s,3H).
实施例30
4-(2-(4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸30
Figure PCTCN2017099579-appb-000090
第一步
1-(4-氯-2-(2,5-二甲氧基吡啶-4-基)-3-氟苯基)乙酮30b
将1-(2-溴-4-氯-3-氟苯基)乙酮30a(630mg,2.51mmol,采用专利申请“WO2013056034”公开的方法制备而得)、化合物1d(550.05mg,3.01mmol)、四(三苯基膦)钯(868.46mg,0.75mmol)和碳酸钠(796.57mg,7.52mmol)加入到3mL1,4-二氧六环和1mL水的混合溶剂中,加毕,加热至95℃,搅拌反应16小时。反应液自然冷却至室温,加入50mL水,用乙酸乙酯萃取(50mL×2),合并有机相,用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物30b(650mg,产率:83.7%)。
MS m/z(ESI):310.3[M+1]
第二步
4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基吡啶-2(1H)-酮30c
将化合物30b(650mg,2.1mmol)溶于20mL1,4-二氧六环中,加入浓盐酸(20mL,240mmol),加毕,加热至110℃,搅拌反应16小时。反应液自然冷却至室温,减压浓缩除去有机溶剂,所得残余物中加入20mL水,用饱和碳酸氢钠溶液中和,用乙酸乙酯萃取(30mL×3),合并有机相,用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物30c(418mg,产率:67.4%)。
MS m/z(ESI):296.1[M+1]
第三步
2-(4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯丙酸30d
将化合物30c(243mg,0.82mmol)溶于50mL四氢呋喃中,加入化合物4a(282.38mg,1.23mmol)、叔丁醇钾(404.07mg,3.6mmol)和叔丁醇镁(280.29mg,1.64mmol),加热至65℃搅拌反应16小时。反应液自然冷却至室温,滴加1M盐酸至pH为3,用乙酸乙酯萃取(150mL×2),合并有机相,用无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物30d(200mg,产率:54.8%)。
MS m/z(ESI):444.4[M+1]
第四步
4-(2-(4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸甲酯30e
将化合物30d(200mg,0.45mmol)、化合物4c(68.11mg,0.45mmol)和N,N-二异丙基乙胺(58.24mg,0.45mmol)溶于5mL乙酸乙酯中,冰浴下,滴加2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,286.74mg,0.45mmol),加毕,升温至65℃,搅拌反应1小时。反应液自然冷却至室温,加入饱和碳酸氢钠溶液淬灭反应,用乙酸乙酯萃取(150mL×2),合并有机相,用无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余 物,得到标题化合物30e(120mg,产率:46.2%)。
MS m/z(ESI):575.4[M-1]
第五步
4-(2-(4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸30
将化合物30e(120mg,0.21mmol)溶于8mL1,2-二氯乙烷中,加入三甲基氢氧化锡(564.08mg,3.12mmol),升温至90℃,搅拌反应48小时。反应液自然冷却至室温,过滤,滤液减压浓缩,用高效液相制备(Waters 2767-SQ detecor2,洗脱体系:乙腈,水)纯化所得残余物,制得到标题化合物30(40mg,产率:32.8%)。
MS m/z(ESI):563.2[M+1]
1H NMR(400MHz,CD3OD)δ8.00(d,2H),7.75-7.67(m,4H),7.45(d,1H),7.31-7.29(m,4H),7.26-7.22(m,1H),6.40(s,1H),5.97-5.91(m,1H),3.63(d,3H),3.61-3.40(m,2H),2.45(d,3H).
实施例31,32
(S)-4-(2-(4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸31
(R)-4-(2-(4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸32
Figure PCTCN2017099579-appb-000091
将化合物30(35mg,0.06mmol)进行手性制备(分离条件:手性制备柱CHIRALPAK ID,5.0cm I.D.*25cm L;流动相:乙醇/二氯甲烷/醋酸=90/10/0.1(V/V/V),流速:60mL/min),收集其相应组分,减压浓缩,得到标题化合物31(11mg)和化合物32(11mg)。
化合物31:
MS m/z(ESI):563.2[M+1]
手性HPLC分析:保留时间8.000分钟,手性纯度:98%(色谱柱:CHIRAL PAK IE4.6*150mm 5um(带保护柱);流动相:乙醇(含0.1%三氟乙酸)/正己烷=50/50(V/V),流速:1.0mL/min)。
1H NMR(400MHz,CD3OD)δ8.00(d,2H),7.75-7.67(m,4H),7.45(d,1H),7.31-7.29(m,4H),7.26-7.22(m,1H),6.40(s,1H),5.97-5.91(m,1H),3.63(d,3H),3.61-3.40(m,2H),2.45(d,3H).
化合物32:
MS m/z(ESI):563.2[M+1]
手性HPLC分析:保留时间3.777分钟,手性纯度:100%(色谱柱:CHIRAL PAK  IE 4.6*150mm 5um(带保护柱);流动相:乙醇(含0.1%三氟乙酸)/正己烷=50/50(V/V),流速:1.0mL/min)。
1H NMR(400MHz,CD3OD)δ8.00(d,2H),7.75-7.67(m,4H),7.45(d,1H),7.31-7.29(m,4H),7.26-7.22(m,1H),6.40(s,1H),5.97-5.91(m,1H),3.63(d,3H),3.61-3.40(m,2H),2.45(d,3H).
实施例33
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(1H-咪唑并[4,5-b]吡啶-5-基)-3-苯基丙酰胺33
Figure PCTCN2017099579-appb-000092
采用实施例18的合成路线,将原料化合物18a替换为1H-咪唑并[4,5-b]吡啶-5-胺,制得标题化合物33(40mg)。
MS m/z(ESI):542.5[M+1]
1H NMR(400MHz,CDCl3)δ10.55(s,1H),8.18(s,1H),8.08(s,2H),7.71(d,1H),7.52-7.50(dd,1H),7.34(d,1H),7.12(m,5H),6.65(s,1H),6.26(s,1H),3.69(s,3H),3.68-3.64(m,1H),3.34-3.29(m,1H),2.49(s,3H).
实施例34
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸甲酯34
Figure PCTCN2017099579-appb-000093
将化合物5(60mg,110.10μmol)溶于5mL二氯甲烷中,加入甲醇(35.27mg,1.1mmol)、4-二甲氨基吡啶(20.34mg,165.14μmol)和1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐(31.54mg,165.14μmol),搅拌反应16小时。反应液中加入20mL饱和碳酸氢钠溶液,用二氯甲烷萃取(50mL×2),合并有机相,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,制得标题化合物34(35mg,产率:56.9%)。
MS m/z(ESI):559.2[M+1]
1H NMR(400MHz,DMSO-d6)δ10.88(s,1H),7.97-7.96(m,1H),7.95-7.94(m,1H),7.83-7.81(d,1H),7.80-7.79(m,1H),7.78-7.77(m,1H),7.62-7.59(m,1H),7.43(s,1H),7.38(s,1H),7.30-7.25(m,4H),7.21-7.17(m,1H),6.31(s,1H),6.05-6.01(m,1H),3.83(s,3H),3.53(s,3H),3.52-3.42(m,2H),2.37(s,3H).
实施例35
4-(2-(4-(5-氯-2-(2,2,2-三氟乙酰基)苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸35
Figure PCTCN2017099579-appb-000094
采用实施例8的合成路线,将原料化合物8c替换为1-(2-溴-4-氯苯基)-2,2,2-三氟乙酮(采用专利申请“WO2011100285”公开的方法制备而得),制得标题化合物35(10mg)。
MS m/z(ESI):599.4[M+1]
1H NMR(400MHz,CDCl3)δ9.77(s,1H),8.10-8.08(m,2H),7.83-7.80(m,3H),7.60-7.58(m,1H),7.43(s,1H),7.36-7.26(m,5H),6.70(s,1H),6.14(br,1H),3.68-3.64(m,1H),3.63(s,3H),3.33-3.28(m,1H).
实施例36
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(4-氰基-3-甲氧基苯基)-3-苯基丙酰胺36
Figure PCTCN2017099579-appb-000095
采用实施例18的合成路线,将原料化合物18a替换为4-氨基-2-甲氧基苯腈(采用专利申请“WO 2013042782”公开的方法制备而得),制得标题化合物36(40mg)。
MS m/z(ESI):556.5[M+1]
1H NMR(400MHz,CDCl3)δ9.89(s,1H),7.73(d,1H),7.54-7.51(dd,1H),7.48(d,1H),7.41(s,1H),7.29(s,4H),7.21(s,1H),7.09-7.06(dd,2H),6.59(s,1H),6.00(s,1H),3.90(s,3H),3.69-3.75(m,1H),3.65(s,3H),3.33-3.28(m,1H),2.50(s,3H).
实施例37
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)-N-乙基苯甲酰胺37
Figure PCTCN2017099579-appb-000096
采用实施例13的合成路线,将原料甲胺替换为乙胺,制得标题化合物37(40mg)。
MS m/z(ESI):572.1[M+1]
1H NMR(400MHz,CDCl3)δ9.61(br,1H),7.73-7.64(m,5H),7.51(d,1H),7.36-7.30(m,4H),7.29-7.24(m,1H),7.13(s,1H),6.64(s,1H),6.11(s,1H),5.99-5.96(m,1H),3.76-3.73(m,1H),3.64(s,3H),3.53-3.51(m,2H),3.34-3.31(m,1H),2.47(s,3H),1.28(t,3H).
实施例38
N-(1H-苯并[d]咪唑-5-基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰胺38
Figure PCTCN2017099579-appb-000097
将化合物8i(80mg,181.86μmol)、1H-苯并[d]咪唑-5-胺38a(24.22mg,181.86μmol,采用公知的方法“Chemical Communications(Cambridge,United Kingdom),2011,47(39),10972-10974”制备而得)和N,N-二异丙基乙胺(70.51mg,545.59μmol)加入到10mL四氢呋喃中,再滴加2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,231.34mg,363.73μmol),加毕,升温至50℃,搅拌反应1.5小时。反应液冷却至室温,减压浓缩,所得残余物中加入25mL饱和碳酸氢钠溶液,用乙酸乙酯萃取(50mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题化合物38(75mg,产率:74.3%)。
MS m/z(ESI):555.5[M+1]
1H NMR(400MHz,DMSO-d6)δ10.84(s,1H),9.05(s,1H),9.14(s,1H),8.27(s,1H),7.79-7.77(d,1H),7.72-7.69(d,1H),7.59-7.52(m,2H),7.41(s,1H),7.34(s,1H),7.30-7.24(m,4H),7.19-7.17(m,1H),6.30(s,1H),6.03-5.99(m,1H),3.52-3.50(m,4H),3.48-3.44(m,1H),2.85-2.75(m,2H),1.25-1.18(m,3H).
实施例39
4-(2-(4-(5-氯-2-(2-环丙基乙酰基)苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸
Figure PCTCN2017099579-appb-000098
采用实施例8的合成路线,将原料化合物8b替换为环丙基乙酰氯(采用专利申请“WO 2015110435”公开的方法制备而得),原料氯化亚铜替换为碘化亚铜制得标题化合物39(30mg)。
MS m/z(ESI):585.2[M+1]
1H NMR(400MHz,CDCl3)δ9.93(br,1H),8.10-8.08(m,2H),7.85-7.83(m,2H),7.69-7.67(m,1H),7.51-7.48(m,1H),7.35-7.26(m,6H),6.63(s,1H),6.22(br,1H),3.69-3.64(m,4H),3.62-3.29(m,1H),2.74-2.72(m,2H),1.07-1.05(m,1H),0.61-0.59(m,2H),0.15-0.14(m,2H).
实施例40
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基-N-(喹唑啉-6-基)丙酰胺40
Figure PCTCN2017099579-appb-000099
采用实施例18的合成路线,将原料化合物18a替换为化合物23a,制得标题化合物40(80mg)。
MS m/z(ESI):553.1[M+1]
1H NMR(400MHz,DMSO-d6)δ11.05(s,1H),9.59(s,1H),9.22(s,1H),8.63(s,1H),8.07-8.01(m,2H),7.83(d,1H),7.61(dd,1H),7.46(s,1H),7.38(s,1H),7.32-7.26(m,4H),7.22-7.18(m,1H),6.33(s,1H),6.10-6.06(m,1H),3.56(s,3H),3.52(d,2H),2.40(s,3H).
实施例41
4-(2-(4-(5-氯-2-(环丙甲酰基)苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸41
Figure PCTCN2017099579-appb-000100
采用实施例8的合成路线,将原料化合物8b替换为环丙甲酰氯(采用专利申请“WO 2015143380”公开的方法制备而得),制得标题化合物41(60mg)。
MS m/z(ESI):571.2[M+1]
1H NMR(400MHz,DMSO-d6)δ10.84(s,1H),7.92(d,2H),7.75-7.72(m,3H),7.61(dd,1H),7.38(d,2H),7.31-7.24(m,4H),7.20-7.17(m,1H),6.34(s,1H),6.05-6.01(m,1H),3.57-3.49(m,2H),3.52(s,3H),2.18-2.11(m,1H),0.85-0.75(m,4H).
实施例42
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(1H-吲唑-6-基)-3-苯基丙酰胺42
Figure PCTCN2017099579-appb-000101
采用实施例18的合成路线,将原料化合物18a替换为6-氨基吲唑(采用公知的方法“Tetrahedron Letters,2010,51(5),786-789”制备而得),制得标题化合物42(83mg)。
MS m/z(ESI):541.4[M+1]
1H NMR(400MHz,DMSO-d6)δ12.94(s,1H),10.67(s,1H),8.13(s,1H),7.99(s,1H),7.82(d,1H),7.70(d,1H),7.61(d,1H),7.48(s,1H),7.38(s,1H),7.31-7.25(m,4H),7.21-7.16(m,2H),6.31(s,1H),6.08-6.04(m,1H),3.56(s,3H),3.47(d,2H),2.38(s,3H).
实施例43
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)-N-环丙基苯甲酰胺43
Figure PCTCN2017099579-appb-000102
采用实施例13的合成路线,将原料甲胺替换为环丙胺,制得标题化合物43(40mg)。
MS m/z(ESI):584.1[M+1]
1H NMR(400MHz,DMSO-d6)δ10.75(s,1H),8.37(d,1H),7.84-7.80(m,3H),7.70(d,2H),7.61(dd,1H),7.43(s,1H),7.38(s,1H),7.31-7.26(m,4H),7.22-7.18(m,1H),6.31(s,1H),6.04-6.00(m,1H),3.55(s,3H),3.50-3.42(m,2H),2.85-2.80(m,1H),2.38(s,3H),0.71-0.54(m,4H).
实施例44
4-(2-(4-(5-氯-2-异丁酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酸44
Figure PCTCN2017099579-appb-000103
采用实施例8的合成路线,将原料化合物8b替换为异丁酰氯(采用公知的方法“Organic Letters,2017,19(7),1768-1771”制备而得),制得标题化合物44(200mg)。
MS m/z(ESI):573.5[M+1]
1H NMR(400MHz,CDCl3)δ9.99(s,1H),8.10(d,2H),7.86(d,2H),7.72(d,1H),7.52-7.29(m,8H),6.59(s,1H),6.28(s,1H),3.67-3.62(m,4H),3.33-3.23(m,2H),1.15-1.12(m,6H).
实施例45
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基-N-(喹喔啉-6-基)丙酰胺45
Figure PCTCN2017099579-appb-000104
采用实施例18的合成路线,将原料化合物18a替换为6-氨基喹喔啉(采用专利申请“WO2013006792”公开的方法制备而得),制得标题化合物45(45mg)。
MS m/z(ESI):553.0[M+1]
1H NMR(400MHz,CD3OD)δ8.85-8.83(d,1H),8.83-8.80(d,1H),8.61-8.57(m,1H),8.08-8.04(d,1H),8.02-7.94(dd,1H),7.85-7.83(d,1H),7.58-7.55(dd,1H),7.39(s,1H),7.33-7.27(m,5H),7.23-7.20(m,1H),6.43(s,1H),6.00-5.95(m,1H),3.65-3.60(m,1H),3.59(s,3H),3.50-3.45(m,1H),2.46(s,3H).
实施例46
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(异喹啉-6-基)-3-苯基丙酰胺46
Figure PCTCN2017099579-appb-000105
采用实施例18的合成路线,将原料化合物18a替换为6-氨基异喹啉(采用专利申请“WO 2010146881”公开的方法制备而得),制得标题化合物46(88mg)。
MS m/z(ESI):552.5[M+1]
1H NMR(400MHz,DMSO-d6)δ10.95(s,1H),9.20(s,1H),8.44-8.43(m,2H),8.10(d,1H),7.83(d,1H),7.77-7.73(m,2H),7.61(d,1H),7.45(s,1H),7.38(s,1H), 7.32-7.26(m,4H),7.21-7.18(m,1H),6.32(s,1H),6.11-6.06(m,1H),3.56(s,3H),3.51(d,2H),2.39(s,3H).
实施例47
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(1H-苯并[d]咪唑-5-基)-3-苯基丙酰胺47
Figure PCTCN2017099579-appb-000106
采用实施例18的合成路线,将原料化合物18a替换化合物38a,制得标题化合物47(10mg)。
MS m/z(ESI):541.2[M+1]
1H NMR(400MHz,MeOH-d4)δ8.18(s,1H),8.03(s,1H),7.84-7.82(d,1H),7.56-7.50(d,2H),7.43(s,1H),7.30-7.22(m,7H),6.43(s,1H),5.89-5.85(m,1H),3.65-3.60(m,1H),3.59(s,3H),3.50-3.45(m,1H),2.46(s,3H).
实施例48
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基-N-(2-(三氟甲基)-1H-苯并[d]咪唑-5-基)丙酰胺48
Figure PCTCN2017099579-appb-000107
将化合物4b(43mg,100.97μmol)、2-(三氟甲基)-1H-苯并[d]咪唑-5-胺48a(20.31mg,100.97μmol,采用公知的方法“International Journal of PharmTech Research,2009,1(2),277-281”制备而得)和N,N-二异丙基乙胺(39.15mg,302.92μmol)加入到15mL四氢呋喃中,再滴加2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物(64.22mg,201.94μmol),加毕,升温至60℃,搅拌反应1小时。反应液冷却至室温,减压浓缩,所得残余物加入15mL饱和碳酸氢钠溶液,用乙酸乙酯萃取(50mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,制得标题化合物48(50mg)。
MS m/z(ESI):609.2[M+1]
1H NMR(400MHz,DMSO-d6)δ13.93-13.83(d,1H),10.74-10.65(d,1H),8.25-8.15 (m,1H),7.84-7.82(d,1H),7.78-7.74(d,1H),7.62-7.60(dd,1H),7.50-7.45(m,1H),7.44-7.36(m,2H),7.35-7.25(m,4H),7.22-7.15(m,1H),6.50(s,1H),6.10-6.00(m,1H),3.55(s,3H),3.51-3.48(m,2H),2.38(s,3H).
实施例49
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(2-甲基-1H-苯并[d]咪唑-5-基)-3-苯基丙酰胺49
Figure PCTCN2017099579-appb-000108
采用实施例48的合成路线,将原料化合物48a替换为2-甲基-1H-苯并[d]咪唑-5-胺(采用专利申请“WO2012044090”公开的方法制备而得),制得标题化合物49(40mg)。
MS m/z(ESI):555.2[M+1]
1H NMR(400MHz,DMSO-d6)δ10.58(s,1H),7.86(s,1H),7.88-7.81(d,1H),7.61-7.59(dd,1H),7.47(s,1H),7.45-7.43(d,1H),7.37(s,1H),7.31-7.24(m,5H),7.21-7.17(m,1H),6.30(s,1H),6.05-6.01(m,1H),3.55(s,3H),3.51-3.48(m,2H),2.50(s,3H),2.38(s,3H).
实施例50
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)-N,N-二甲基苯甲酰胺50
Figure PCTCN2017099579-appb-000109
采用实施例13的合成路线,将原料甲胺替换为二甲胺,制得标题化合物50(40mg)。
MS m/z(ESI):572.1[M+1]
1H NMR(400MHz,DMSO-d6)δ10.72(s,1H),7.84(d,1H),7.82(d,2H),7.70-7.68(m,1H),7.44-7.40(m,4H),7.31-7.28(m,4H),7.21-7.18(m,1H),6.31(s,1H),6.04-6.00(m,1H),3.55(s,3H),3.52-3.47(m,2H),2.96(s,6H),2.38(s,3H).
实施例51
5-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)-2-吡啶酰胺51
Figure PCTCN2017099579-appb-000110
采用实施例18的合成路线,将原料化合物18a替换为5-氨基-2-吡啶酰胺(采用专利申请“WO2013146963”公开的方法制备而得),制得标题化合物51(70mg)。
MS m/z(ESI):545.1[M+1]
1H NMR(400MHz,CD3OD)δ8.83-8.82(d,1H),8.23-8.20(dd,1H),8.08-8.06(d,1H),7.85-7.83(d,1H),7.57-7.54(dd,1H),7.32-7.29(m,1H),7.28-7.25(m,5H),7.23-7.20(m,1H),6.41(s,1H),5.89-5.85(m,1H),3.65-3.60(m,1H),3.59(s,3H),3.50-3.45(m,1H),2.46(s,3H).
实施例52
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基-N-(1H-吡咯并[3,2-b]吡啶-6-基)丙酰胺52
Figure PCTCN2017099579-appb-000111
采用实施例18的合成路线,将原料化合物18a替换为1H-吡咯并[3,2-b]吡啶-6-胺(韶远),制得标题化合物52(23mg)。
MS m/z(ESI):541.4[M+1]
1H NMR(400MHz,DMSO-d6)δ8.94(s,1H),8.69(s,1H),8.08(d,1H),7.86(d,1H),7.58-7.55(m,1H),7.32-7.18(m,7H),6.82(d,1H),6.45(s,1H),5.76-5.73(m,1H),3.63-3.61(m,1H),3.51(s,3H),3.50-3.48(m,1H),2.48(s,3H).
实施例53
3-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苄基氨基甲酸甲酯53
Figure PCTCN2017099579-appb-000112
Figure PCTCN2017099579-appb-000113
第一步
3-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苄基氨基甲酸叔丁酯53b
将化合物4b(90mg,211.34μmol)、3-氨基苄基甲酸叔丁酯53a(51.68mg,232.47μmol,采用公知的方法“Chemical Communications(Cambridge,United Kingdom),2014,50(97),15305-15308”制备而得)和N,N-二异丙基乙胺(273.14mg,2.11mmol)溶于15mL乙酸乙酯中,再滴加2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物(268.97mg,845.36μmol),加毕,升温至75℃,搅拌反应2小时。反应液冷却至室温,加入30mL水,滴加3M盐酸至pH为5,分液,水相用乙酸乙酯萃取(30mL×2),合并有机相,用饱和氯化钠溶液洗涤(35mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题化合物53b(105mg,产率:78.85%)。
MS m/z(ESI):630.1[M+1]
第二步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(3-(氨甲基)苯基)-3-苯基丙酰胺53c
将化合物53b(105mg,166.63μmol)溶于7mL二氯甲烷中,滴加三氟乙酸(1mL),搅拌反应1小时。反应液减压浓缩,得粗品标题化合物53c(80mg),产物不经纯化直接投入下一步反应。
MS m/z(ESI):530.1[M+1]
第三步
3-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苄基氨基甲酸甲酯53
将粗品化合物53c(80mg,105.94μmol)溶于10mL二氯甲烷中,滴加三乙胺(61.096mg,603.76μmol),冰浴冷却下,滴加氯甲酸甲酯(21.40mg,226.41μmol),室温搅拌反应2小时。反应液中加入25mL二氯甲烷,依次用0.5M盐酸洗涤(15mL)、饱和碳酸氢钠溶液(15mL)、饱和食盐水(15mL)洗涤,无水硫酸钠干燥。过滤, 滤液减压浓缩,用高效液相制备(Waters 2767-SQ detecor2,洗脱体系:乙腈,水)纯化所得残余物,制得标题化合物53(35mg,39.4%)。
MS m/z(ESI):588.3[M+1]
1H NMR(400MHz,DMSO-d6)δ9.24(s,1H),7.66(d,1H),7.47(dd,1H),7.43-7.41(m,2H),7.28-7.20(m,7H),7.12(s,1H),7.04(d,1H),6.55(s,1H),6.00-5.91(m,1H),5.08(s,1H),4.31(d,2H),3.72-3.65(m,1H),3.68(s,3H),3.60(s,3H),3.28-3.23(m,1H),2.43(s,3H).
实施例54
2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(2-氰基-1H-吲哚-6-基)-3-苯基丙酰胺54
Figure PCTCN2017099579-appb-000114
采用实施例38的合成路线,将原料化合物38a替换为6-氨基-1H-吲哚-2-甲腈(采用专利申请“WO20160271105”公开的方法制备而得),制得标题化合物54(30mg)。
MS m/z(ESI):579.1[M+1]
1H NMR(400MHz,CD3OD)δ8.01(s,1H),7.78-7.79(d,1H),7.59-7.57(d,1H),7.56-7.53(dd,1H),7.38(s,1H),7.33-7.25(m,5H),7.25-7.18(m,1H),7.16-7.15(d,1H),7.14-7.12(dd,1H),6.42(s,1H),5.95-5.90(m,1H),3.60-3.56(m,1H),3.54(s,3H),3.45-3.35(m,1H),3.00-2.95(m,2H),1.10-1.00(m,3H).
实施例55
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(4-氰基-3-(三氟甲基)苯基)-3-苯基丙酰胺55
Figure PCTCN2017099579-appb-000115
采用实施例18的合成路线,将原料化合物18a替换为4-氨基-2-(三氟甲基)苯腈(采用公知的方法“Medicinal Chemistry Research,2016,25(4),539-552”制备而得),制得标题化合物55(40mg)。
MS m/z(ESI):594.4[M+1]
1H NMR(400MHz,CDCl3)δ10.31(s,1H),7.75-7.72(m,2H),7.55-7.51(m,2H)7.29-7.26(m,5H),7.24-7.16(m,2H),6.55(s,1H),5.98(s,1H),3.71-3.67(m,1H),3.65(s,3H),3.33-3.27(m,1H),2.51(m,3H).
实施例56
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(4-(甲基磺酰基)苯基)-3-苯基丙酰胺56
Figure PCTCN2017099579-appb-000116
采用实施例18的合成路线,将原料化合物18a替换为4-(甲基磺酰基)苯胺(采用专利申请“WO2014100833”公开的方法制备而得),制得标题化合物56(110mg)。
MS m/z(ESI):579.0[M+1]
1H NMR(400MHz,DMSO-d6)δ10.96(s,1H),7.91-7.86(m,4H),7.83(d,1H),7.61(dd,1H),7.39(d,2H),7.30-7.25(m,4H),7.21-7.18(m,1H),6.32(s,1H),6.03-5.99(m,1H),3.53(s,3H),3.50-3.45(m,2H),3.18(s,3H),2.38(s,3H).
实施例57
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(1,6-萘啶-3-基)-3-苯基丙酰胺57
Figure PCTCN2017099579-appb-000117
采用实施例48的合成路线,将原料化合物48a替换为1,6-萘啶-3-胺(采用专利申请“WO2007048070”公开的方法制备而得),制得标题化合物57(40mg)。
MS m/z(ESI):553.2[M+1]
1H NMR(400MHz,DMSO-d6)δ11.18(s,1H),9.40(s,1H),9.14(s,1H),8.95(s,1H),8.67-8.65(d,1H),7.87-7.82(m,2H),7.62-7.60(d,1H),7.48(s,1H),7.38(s,1H),7.30-7.26(m,4H),7.21-7.20(m,1H),6.36(s,1H),6.05-6.03(m,1H),3.64-3.58(m,4H),3.30-3.22(m,1H),2.40(s,3H).
实施例58
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基-N-(4-氨基磺酰基苯基)丙酰胺58
Figure PCTCN2017099579-appb-000118
采用实施例18的合成路线,将原料化合物18a替换为4-(氨基磺酰基)苯胺(采用公知的方法“Journal of Organic Chemistry,2014,79(19),9433-9439”制备而得),制得标题化合物58(40mg)。
MS m/z(ESI):580.2[M+1]
1H NMR(400MHz,CD3OD)δ7.85-7.82(m,3H),7.76-7.74(m,2H),7.56-7.54(dd,1H),7.33-7.32(m,2H),7.28-7.25(m,4H),7.22-7.19(m,1H),6.41(s,1H),5.89-5.85(m,1H),3.65-3.60(m,1H),3.59(s,3H),3.50-3.45(m,1H),2.46(s,3H).
实施例59
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(4-氰基-2-氟苯基)-3-苯基丙酰胺59
Figure PCTCN2017099579-appb-000119
采用实施例18的合成路线,将原料化合物18a替换为4-氨基-3-氟苯腈(采用公知的方法“Journal of Medicinal Chemistry,2005,48(18),5823-5836”制备而得),制得标题化合物59(20mg)。
MS m/z(ESI):544.4[M+1]
1H NMR(400MHz,CDCl3)δ9.83(s,1H),8.53(t,1H),7.72(d,1H),7.53-7.50(dd,1H),7.47(d,2H),7.42-7.29(dd,1H),7.37-7.32(m,4H),7.30-7.26(m,1H),6.92(s,1H),6.65(s,1H),5.86(s,1H),3.82-3.74(m,1H),3.61(s,3H),3.37-3.32(m,1H),2.49(m,3H).
实施例60
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(3-氧代-3,4-二氢-2H-苯并[b][1,4]噁嗪-6-基)-3-苯基丙酰胺60
Figure PCTCN2017099579-appb-000120
采用实施例18的合成路线,将原料化合物18a替换为6-氨基-2H-苯并[b][1,4]噁嗪-3(4H)-酮(采用专利申请“WO20100216783”公开的方法制备而得),制得标题化合物60(50mg)。
MS m/z(ESI):569.8[M-1]
1H NMR(400MHz,CD3OD)δ10.10(s,1H),7.85-7.83(d,1H),7.58-7.55(dd,1H),7.39(s,1H),7.38-7.36(m,2H),7.29-7.25(m,4H),7.23-7.20(m,1H),7.00-6.99(dd,1H),6.91-6.81(d,1H),6.43(s,1H),5.89-5.85(m,1H),4.54(s,2H),3.65-3.60(m,1H),3.59(s,3H),3.50-3.45(m,1H),2.46(s,3H).
实施例61,62
(S)-(5-甲基-2-氧代-1,3-二氧杂环戊烯-4-基)甲基4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酯61
(R)-(5-甲基-2-氧代-1,3-二氧杂环戊烯-4-基)甲基4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲 氧基-2-氧代吡啶-1(2H)-基)-3-苯基丙酰氨基)苯甲酯62
Figure PCTCN2017099579-appb-000121
采用实施例14,15的合成路线,将原料化合物14b替换为4-(氯甲基)-5-甲基-1,3-二氧杂环戊烯-2-酮(采用专利申请“CN103450146”公开的方法制备而得),经手性制备(分离条件:色谱柱:Superchiral S-AD(Chiralway),2cm I.D.*25cm Length,5um;流动相:二氧化碳:异丙醇=60:40,流速:50g/min),收集其相应组分,减压浓缩,制得标题化合物61(600mg)和化合物62(600mg)。
化合物61
MS m/z(ESI):657.5[M+1]
手性HPLC分析:保留时间7.283分钟,手性纯度:99.8%(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:乙醇/甲醇=50/50(V/V),流速:1.0mL/min)。
1H NMR(400MHz,DMSO-d6)δ10.91(s,1H),7.98-7.97(m,1H),7.96-7.95(m,1H),7.83-7.79(m,3H),7.62-7.59(dd,1H),7.41(s,1H),7.37(s,1H),7.30-7.26(m,4H),7.20-7.17(m,1H),6.30(s,1H),6.04-5.95(m,1H),5.20(s,2H),3.51(s,3H),3.49-3.42(m,2H),2.37(s,3H),2.22(s,3H).
化合物62
MS m/z(ESI):657.2[M+1]
手性HPLC分析:保留时间5.342分钟,手性纯度:99.8%(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:乙醇/甲醇=50/50(V/V),流速:1.0mL/min)。
1H NMR(400MHz,DMSO-d6)δ10.92(s,1H),7.98(d,2H),7.84-7.80(m,3H),7.63-7.60(m,1H),7.40(d,2H),7.30-7.26(m,4H),7.22-7.18(m,1H),6.31(s,1H),6.04-6.01(m,1H),5.20(s,2H),3.54(s,3H),3.51-3.43(m,2H),2.38(s,3H),2.22(s,3H).
实施例63
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-氟苯基)丙酰氨基)苯甲酸63
Figure PCTCN2017099579-appb-000122
采用实施例4的合成路线,将原料化合物4a替换为2-溴-3-(4-氟苯基)丙酸(采用专利申请“US5981529A”公开的方法制备而得),制得标题化合物63(64mg)。
MS m/z(ESI):563.4[M+1]
1H NMR(400MHz,DMSO-d6)δ10.79(s,1H),7.90(d,2H),7.84(d,1H),7.71(d,2H), 7.61(dd,1H),7.39(s,2H),7.33-7.29(m,2H),7.10(t,2H),6.30(s,1H),6.03-5.99(m,1H),3.55(s,3H),3.50-3.41(m,2H),2.40(s,3H).
实施例64
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(2-溴苯基)丙酰氨基)苯甲酸64
Figure PCTCN2017099579-appb-000123
采用实施例7的合成路线,将原料化合物7c替换为1-溴-2-(溴甲基)苯(采用公知的方法“Bioorganic & Medicinal Chemistry Letters,2014,24(21),5127-5133”制备而得),制得标题化合物64(8mg)。
MS m/z(ESI):625.3[M+1]
1H NMR(400MHz,CD3OD)δ8.02-7.98(m,2H),7.87(d,1H),7.74-7.72(m,2H),7.62-7.57(m,2H),7.34(d,1H),7.29-7.26(m,3H),7.19-7.15(m,1H),6.43(s,1H),5.93-5.89(m,1H),3.79-3.74(m,1H),3.63-3.60(m,1H),3.58(s,3H),2.51(s,3H).
实施例65
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(2,4-二氟苯基)丙酰氨基)苯甲酸65
Figure PCTCN2017099579-appb-000124
采用实施例7的合成路线,将原料化合物7c替换为1-(溴甲基)-2,4-二氟苯(采用专利申请“WO2012177638”公开的方法制备而得),制得标题化合物65(8mg)。
MS m/z(ESI):581.4[M+1]
1H NMR(400MHz,CD3OD)δ8.02-7.99(m,2H),7.87(d,1H),7.74-7.72(m,2H),7.58(dd,1H),7.36-7.28(m,3H),6.98-6.90(m,2H),6.41(s,1H),5.91(br,1H),3.63-3.59(m,4H),3.52-3.46(m,1H),2.52(s,3H).
实施例66
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(邻甲基苯基)丙酰氨基)苯甲酸66
Figure PCTCN2017099579-appb-000125
采用实施例7的合成路线,将原料化合物7c替换为1-(溴甲基)-2-甲基苯(采用公知的方法“Journal of Organic Chemistry,2014,79(1),223-229”制备而得),制得标题化合物66(60mg)。
MS m/z(ESI):559.2[M+1]
1H NMR(400MHz,CDCl3)δ9.95(s,1H),8.09(d,2H),7.86(d,2H),7.72(d,1H),7.52-7.50(m,2H),7.30(s,1H),7.19-7.13(m,4H),6.63(s,1H),6.30(s,1H),3.69-3.62(m,4H),3.28-3.24(m,1H),2.52-2.46(m,6H).
实施例67
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(间甲基苯基)丙酰胺基)苯甲酸67
Figure PCTCN2017099579-appb-000126
采用实施例7的合成路线,将原料化合物7c替换为1-(溴甲基)-3-甲基苯(采用公知的方法“Chemical Communications(Cambridge,United Kingdom),2014,50(28),3692-3694”制备而得),制得标题化合物67(80mg)。
MS m/z(ESI):559.2[M+1]
1H NMR(400MHz,CDCl3)δ9.87(s,1H),8.10(d,2H),7.84-7.71(m,3H),7.52-7.50(m,1H),7.31(s,2H),7.21-7.07(m,4H),6.65(s,1H),6.18(s,1H),3.67-3.59(m,4H),3.27-3.22(m,1H),2.66(s,3H),2.33(s,3H).
实施例68
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(2-氟苯基)丙酰氨基)苯甲酸68
Figure PCTCN2017099579-appb-000127
采用实施例7的合成路线,将原料化合物7c替换为1-(溴甲基)-2-氟苯(采用公知的方法“Tetrahedron Letters,2000,41(27),5161-5164”制备而得),制得标题化合物68(55mg)。
MS m/z(ESI):563.2[M+1]
1H NMR(400MHz,DMSO-d6)δ10.73(s,1H),7.90-7.89(m,1H),7.90-7.89(m,1H),7.84-7.82(d,1H),7.73-7.72(m,1H),7.71-7.70(m,1H),7.62-7.60(dd,1H),7.40-7.38(d,2H),7.33-7.29(m,1H),7.28-7.24(m,1H),7.16-7.12(m,1H),7.10-7.08(m,1H),6.33(s,1H),6.04-5.95(m,1H),3.51(s,3H),3.49-3.42(m,2H),2.39(s,3H).
实施例69
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-氯苯基)丙酰氨基)苯甲酸69
Figure PCTCN2017099579-appb-000128
采用实施例4的合成路线,将原料化合物4a替换为2-溴-3-(4-氯苯基)丙酸(采用专利申请“WO2012118216”公开的方法制备而得),制得标题化合物69(15mg)。
MS m/z(ESI):579.5[M+1]
1H NMR(400MHz,DMSO-d6)δ10.83(s,1H),7.95-7.94(m,1H),7.93-7.92(m,1H),7.85-7.83(d,1H),7.78-7.77(m,1H),7.76-7.75(m,1H),7.63-7.61(m,1H),7.43-7.42(m,1H),7.41-7.40(m,1H),7.36-7.34(m,2H),7.31-7.29(m,2H),6.33(s,1H),6.04-5.95(m,1H),3.51(s,3H),3.49-3.42(m,2H),2.39(s,3H).
实施例70
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(2-氯苯基)丙酰氨基)苯甲酸70
Figure PCTCN2017099579-appb-000129
采用实施例7的合成路线,将原料化合物7c替换为1-(溴甲基)-2-氯苯(采用公知的方法“Tetrahedron Letters,2016,57(2),168-171”制备而得),制得标题化合物70(25mg)。
MS m/z(ESI):579.2[M+1]
1H NMR(400MHz,CDCl3)δ9.81(s,1H),8.10(d,2H),7.83(d,2H),7.72(d,1H),7.51(d,1H),7.35-7.30(m,1H),7.30-7.29(m,1H),7.29-7.28(m,2H),7.27-7.23(m,2H),6.62(s,1H),6.30(s,1H),3.77-3.71(m,1H),3.69(s,3H),3.52-3.49(m,1H),2.51(s,3H).
实施例71
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(3-甲氧基苯基)丙酰氨基)苯甲酸71
Figure PCTCN2017099579-appb-000130
采用实施例7的合成路线,将原料化合物7c替换为1-(溴甲基)-3-甲氧基苯(采用专利申请“WO2014135095”公开的方法制备而得),制得标题化合物71(48mg)。
MS m/z(ESI):575.4[M+1]
1H NMR(400MHz,DMSO-d6)δ10.83(s,1H),7.91(d,2H),7.82(d,1H),7.73(d,2H),7.61(dd,1H),7.43(s,1H),7.38(s,1H),7.18(t,1H),6.89-6.85(m,2H),6.76(dd,1H),6.32(s,1H),6.06-6.02(m,1H),3.70(s,3H),3.54(s,3H),3.47-3.44(m,2H),2.37(s,3H).
实施例72
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(3-甲氧基苯基)丙酰氨基)苯甲酸72
Figure PCTCN2017099579-appb-000131
将化合物71(48mg,83.48μmol)进行手性制备(分离条件:手性制备柱CHIRAL PAK IF,20*250mm,5um;流动相:乙醇(含0.01%三氟醋酸)=100,流速:7mL/min),收集其相应组分,减压浓缩,得到标题化合物72(18mg)。
MS m/z(ESI):575.4[M+1]
手性HPLC分析:保留时间8.546分钟,手性纯度:98%(色谱柱:Lux Amylose-1(AD)4.6*150mm 5um(带保护柱);流动相:乙醇(含0.1%三氟醋酸)/正己烷=50/50(v/v))。1H NMR(400MHz,DMSO-d6)δ12.69(s,1H),10.88(s,1H),7.92(d,2H),7.82(d,1H),7.76(d,2H),7.61(dd,1H),7.42(s,1H),7.38(s,1H),7.18(t,1H),6.90-6.86(m,2H),6.76(dd,1H),6.32(s,1H),6.05-6.01(m,1H),3.70(s,3H),3.54(s,3H),3.48-3.42(m,2H),2.37(s,3H).
实施例73
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(2-氯苯基)丙酰氨基)苯甲酸甲酯73
Figure PCTCN2017099579-appb-000132
采用化合物7f的合成路线,将原料化合物7c替换为1-(溴甲基)-2-氯苯(采用 公知的方法“Tetrahedron Letters,2016,57(2),168-171”制备而得),制得标题化合物73(70mg)。
MS m/z(ESI):593.4[M+1]
1H NMR(400MHz,CDCl3)δ10.17(s,1H),8.00(d,2H),7.69-7.62(m,3H),7.49(d,1H),7.31(d,2H),7.25-7.16(m,4H),6.47(s,1H),6.23(s,1H),3.93(s,3H),3.76-3.74(m,1H),3.65(s,3H),3.55-3.52(m,1H),2.48(s,3H).
实施例74
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(对甲基苯基)丙酰氨基)苯甲酸74
Figure PCTCN2017099579-appb-000133
第一步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(对甲基苯基)丙酸叔丁酯74b
将化合物7b(100mg,0.26mmol)和1-(溴甲基)-4-甲基苯74a(94.45mg,0.51mmol,采用公知的方法“Tetrahedron Letters,2016,57(22),2430-2433”制备而得)溶于6mL四氢呋喃中,反应液冷却至-78℃,滴加二(三甲基硅基)氨基锂溶液(1.02mL,1.02mmol),搅拌反应2小时。-78℃下,向反应液中缓慢加入15mL饱和氯化铵溶液淬灭反应,自然升至室温,用乙酸乙酯萃取(50mL×2),合并有机相,用无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物74b(120mg,产率:94.8%)。
MS m/z(ESI):496.2[M+1]
第二步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(对甲基苯基)丙酸74c
将化合物74b(100mg,0.20mmol)溶于4mL二氯甲烷中,滴加三氟乙酸(0.5mL),搅拌反应5小时。反应液减压浓缩,得粗品标题化合物74c(80mg),产物不 经纯化直接投入下一步反应。
MS m/z(ESI):440.0[M+1]
第三步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(对甲基苯基)丙酰氨基)苯甲酸74
将粗品化合物74c(80mg,0.18mmol)和化合物8j(68.52mg,0.27mmol)溶于10mL乙酸乙酯中,滴加N,N-二异丙基乙胺(112.43mg,0.87mmol)和2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,415.18mg,0.65mmol),加毕,升温至60℃,搅拌反应2小时。反应液中加入15mL水,用乙酸乙酯萃取(15mL×2),合并有机相,用饱和氯化钠溶液洗涤(15mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相制备(Waters 2767-SQ detecor2,洗脱体系:乙腈,水)纯化所得残余物,制得标题化合物74(40mg,产率:39.4%)。
MS m/z(ESI):559.1[M+1]
1H NMR(400MHz,CDCl3)δ9.92(s,1H),8.10(d,2H),7.84(d,2H),7.71(d,1H),7.56-7.50(m,2H),7.30(s,1H),7.23-7.11(m,4H),6.64(s,1H),6.20(s,1H),3.67-3.59(m,4H),3.27-3.22(m,1H),2.49(s,3H),2.31(s,3H).
实施例75
4-(3-(4-乙酰氨基苯基)-2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)丙酰氨基)苯甲酸75
Figure PCTCN2017099579-appb-000134
采用实施例16的合成路线,将原料化合物环丙酰氯替换为乙酰氯,制得标题化合物75(10mg)。
MS m/z(ESI):602.4[M+1]
1H NMR(400MHz,DMSO-d6)δ10.88(s,1H),9.89(s,1H),7.92(d,2H),7.83(d,1H),7.76(d,2H),7.61(dd,1H),7.46(d,2H),7.41(d,2H),7.18(d,2H),6.30(s,1H),6.00-5.96(m,1H),3.55(s,3H),3.42-3.39(m,2H),2.39(s,3H),2.00(s,3H).
实施例76
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(2,6-二氯苯基)丙酰氨基)苯甲酸76
Figure PCTCN2017099579-appb-000135
采用实施例74的合成路线,将原料化合物74a替换为2-(溴甲基)-1,3-二氯苯(采用公知的方法“Organic Letters,2017,19(7),1634-1637”制备而得)制得标题化合物76(20mg)。
MS m/z(ESI):613.1[M+1]
1H NMR(400MHz,CD3OD)δ8.00(dd,2H),8.86-7.88(m,1H),7.74-7.70(m,2H),7.52-7.50(m,1H),7.42-7.40(m,2H),7.34(d,2H),7.25(t,1H),6.87(s,1H),6.42(s,1H),3.92-3.82(m,1H),3.64-3.52(m,4H),2.51(d,3H).
实施例77
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(5-氟-2-甲基苯基)丙酰氨基)苯甲酸77
Figure PCTCN2017099579-appb-000136
采用实施例7的合成路线,将原料化合物7c替换为2-(溴甲基)-4-氟-1-甲基苯(Adamas)制得标题化合物77(30mg)。
MS m/z(ESI):577.2[M+1]
1H NMR(400MHz,CDCl3)δ9.94(s,1H),8.11(d,2H),7.87(d,2H),7.73(d,1H),7.53-7.50(m,2H),7.31(s,1H),7.15-7.14(m,1H),6.87-6.66(m,2H),6.66(s,1H),6.28(s,1H),3.70-3.59(m,4H),3.23-3.19(m,1H),2.53(s,3H),2.46(s,3H).
实施例78
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(2-甲氧基苯基)丙酰氨基)苯甲酸78
Figure PCTCN2017099579-appb-000137
采用实施例74的合成路线,将原料化合物74a替换为1-(溴甲基)-2-甲氧基苯(采用公知的方法“Journal of the American Chemical Society,2013,135(30),10934-10937”制备而得),制得标题化合物78(60mg)。
MS m/z(ESI):575.0[M+1]
1H NMR(400MHz,DMSO-d6)δ7.90(d,2H),7.82(d,1H),7.73(d,2H),7.60(dd,1H),7.37(s,1H),7.25(s,1H),7.21-7.14(m,2H),6.94(d,1H),6.82(t,1H),6.31(s,1H),5.91-5.87(m,1H),3.75(s,3H),3.48(s,3H),3.38(d,2H),2.39(s,3H).
实施例79
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(吡啶-2-基)丙酰氨 基)苯甲酸79
Figure PCTCN2017099579-appb-000138
采用实施例74的合成路线,将原料化合物74a替换为2-(溴甲基)吡啶(采用公知的方法“Journal of the American Chemical Society,2016,138(26),8253-8258”制备而得)制得标题化合物79(370mg)。
MS m/z(ESI):546.4[M+1]
1H NMR(400MHz,CD3OD)δ8.72(d,1H),8.29-8.25(m,1H),8.00(d,2H),7.91(d,1H),7.75-7.71(m,4H),7.59(dd,1H),7.35(d,1H),7.25(s,1H),6.40(s,1H),6.09-5.87(m,1H),3.98-3.94(m,1H),3.80-3.76(m,1H),3.59(s,3H),2.56(s,3H).
实施例80,81
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(吡啶-2-基)丙酰氨基)苯甲酸80
(R)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(吡啶-2-基)丙酰氨基)苯甲酸81
Figure PCTCN2017099579-appb-000139
将化合物79(370mg,677.69μmol)进行手性制备(分离条件:手性制备柱CHIRAL PAK IF,20*250mm,5um;流动相:正己烷:乙醇=50:50,流速:10.0mL/min),收集其相应组分,减压浓缩,得到标题化合物80(120mg)和化合物81(120mg)。
化合物80:
MS m/z(ESI):546.2[M+1]
手性HPLC分析:保留时间9.971分钟,(色谱柱:Lux Amylose-1(AD)4.6*150mm,5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
化合物81:
MS m/z(ESI):546.2[M+1]
手性HPLC分析:保留时间6.219分钟,(色谱柱:Lux Amylose-1(AD)4.6*150mm,5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
实施例82
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(吡啶-3-基)丙酰氨 基)苯甲酸82
Figure PCTCN2017099579-appb-000140
采用实施例74的合成路线,将原料化合物74a替换为3-(溴甲基)吡啶(采用公知的方法“Chemical Communications(Cambridge,United Kingdom),2016,52(82),12159-12162”制备而得),制得标题化合物82(30mg)。
MS m/z(ESI):546.2[M+1]
1H NMR(400MHz,CD3OD)δ8.76-8.72(m,2H),8.42(d,1H),8.03-7.92(m,5H),7.76(d,2H),7.62-7.59(m,1H),7.37(s,2H),6.36(s,1H),3.84-3.63(m,5H),2.59(s,3H).
实施例83,84
(R)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(吡啶-2-基)丙酰氨基)苯甲酸83
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(吡啶-2-基)丙酰氨基)苯甲酸84
Figure PCTCN2017099579-appb-000141
将化合物82(300mg,549.48μmol)进行手性制备(分离条件:手性制备柱CHIRAL PAK IF,20*250mm,5um;流动相:正己烷:乙醇(含0.01%三氟乙酸)=50:50,流速:12.0mL/min),收集其相应组分,减压浓缩,得到标题化合物83(120mg)和化合物84(120mg)。
化合物83:
MS m/z(ESI):546.1[M+1]
手性HPLC分析:保留时间3.723分钟,(色谱柱:CHIRAL PAK IF 4.6*150mm,5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,CD3OD)δ8.76-8.72(m,2H),8.42(d,1H),8.03-7.92(m,5H),7.76(d,2H),7.62-7.59(m,1H),7.37(s,2H),6.36(s,1H),3.84-3.63(m,5H),2.59(s,3H).
化合物84:
MS m/z(ESI):546.1[M+1]
手性HPLC分析:保留时间7.315分钟,(色谱柱:CHIRAL PAK IF 4.6*150mm,5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,CD3OD)δ8.76-8.72(m,2H),8.42(d,1H),8.03-7.92(m,5H),7.76(d,2H),7.62-7.59(m,1H),7.37(s,2H),6.36(s,1H),3.84-3.63(m,5H),2.59(s,3H).
实施例85
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(吡啶-2-基)丙酰氨基)苯甲酰胺85
Figure PCTCN2017099579-appb-000142
采用实施例11的合成路线,将原料化合物5替换为化合物80制得标题化合物85(18mg)。
MS m/z(ESI):545.1[M+1]
1H NMR(400MHz,CD3OD)δ8.51(d,1H),7.88-7.84(m,3H),7.78-7.74(dd,1H),7.72(d,2H),7.57-7.55(dd,1H),7.35-7.32(m,3H),7.30-7.27(m,1H),6.39(s,1H),6.06(t,1H),3.79-3.74(dd,1H),3.64-3.58(m,4H),2.49(m,3H).
实施例86
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(吡啶-4-基)丙酰氨基)苯甲酸86
Figure PCTCN2017099579-appb-000143
采用实施例74的合成路线,将原料化合物74a替换为4-(溴甲基)吡啶氢溴酸盐(采用公知的方法“Chemical Communications(Cambridge,United Kingdom),2011,47(5),1482-1484”制备而得)制得标题化合物86(20mg)。
MS m/z(ESI):546.2[M+1]
实施例87
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(2-氰基苯基)丙酰氨基)苯甲酸87
Figure PCTCN2017099579-appb-000144
采用实施例7的合成路线,将原料化合物7c替换为2-(溴甲基)苯腈(采用公知 的方法“Journal of Organic Chemistry,2014,79(23),11592-11608”制备而得)制得标题化合物87(15mg)。
MS m/z(ESI):570.1[M+1]
1H NMR(400MHz,DMSO-d6)δ12.75(s,1H),10.69(s,1H),7.92(s,1H),7.90(s,1H),7.86-7.84(d,1H),7.83(m,1H),7.74(s,1H),7.72(s,1H),7.64-7.60(m,2H),7.45-7.41(m,2H),7.39-7.38(d,1H),7.30(s,1H),6.33(s,1H),6.04-5.95(m,1H),3.76-3.70(m,1H),3.59-3.54(m,1H),3.51(s,3H),2.43(s,3H).
实施例88
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(3-氰基苯基)丙酰氨基)苯甲酸88
Figure PCTCN2017099579-appb-000145
采用实施例7的合成路线,将原料化合物7c替换为3-(溴甲基)苯腈(采用公知的方法“ChemMedChem,2015,10(4),688-714”制备而得)制得标题化合物88(25mg)。
MS m/z(ESI):570.4[M+1]
1H NMR(400MHz,DMSO-d6)δ10.78(s,1H),7.94-7.93(m,1H),7.92-7.91(m,1H),7.86-7.84(d,1H),7.77-7.68(m,4H),7.62-7.60(dd,1H),7.59-7.57(d,1H),7.52-7.48(m,1H),7.44(s,1H),7.38(s,1H),6.30(s,1H),6.04-6.00(m,1H),3.62-3.50(m,5H),2.41(s,3H).
实施例89
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(3-氰基苯基)丙酰氨基)苯甲酸89
Figure PCTCN2017099579-appb-000146
将化合物88(350mg,614.04μmol)进行手性制备(分离条件:手性制备柱CHIRAL PAK IF,20*250mm;流动相:正己烷:乙醇:三氟乙酸=50:50:0.06,流速:10.0mL/min),收集其相应组分,减压浓缩,得到标题化合物89(60mg)。
MS m/z(ESI):570.1[M+1]
手性HPLC分析:保留时间12.723分钟,(色谱柱:CHIRALPAK IE 150*4.6mm,5um(带保护柱);流动相:正己烷/乙醇(含0.01%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,CD3OD)δ8.01-8.00(m,1H),7.98-7.97(m,1H),7.87-7.85(d,1H),7.73-7.71(m,1H),7.70-7.69(m,2H),7.61-7.55(m,3H),7.50-7.46(m,1H),7.38(s,1H),7.34-7.33(d,1H),6.39(s,1H),5.95-5.85(m,1H),3.65-3.60(m,1H)3.59(s,3H),3.50-3.45(m,1H),2.46(s,3H).
实施例90
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-氰基苯基)丙酰氨基)苯甲酸90
Figure PCTCN2017099579-appb-000147
采用实施例74的合成路线,将原料化合物74a替换为4-(溴甲基)苯腈(采用公知的方法“Organic & Biomolecular Chemistry,2017,15(12),2551-2561”制备而得)制得标题化合物90(15mg)。
MS m/z(ESI):570.5[M+1]
1H NMR(400MHz,DMSO-d6)δ10.82(s,1H),7.94-7.93(m,1H),7.92-7.91(m,1H),7.85-7.83(d,1H),7.77-7.74(m,4H),7.62-7.59(dd,1H),7.48(s,1H),7.45(s,1H),7.40-7.39(m,1H),7.38-7.36(d,1H),6.29(s,1H),6.04-6.00(m,1H),3.67-3.65(m,1H),3.64-3.54(m,4H),2.39(s,3H).
实施例91
4-(2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)丙酰氨基)苯甲酸91
Figure PCTCN2017099579-appb-000148
第一步
(1-环丙基-1H-吡唑-3-基)甲醇91b
将1-环丙基-1H-吡唑-3-甲酸乙酯91a(500mg,2.77mmol,采用专利申请“WO20140349990”公开的方法制备而得)溶于15mL四氢呋喃中,0℃下加入氢化 锂铝(527.18mg,13.87mmol),0℃搅拌反应1小时。反应液中加入3mL碳酸氢钠溶液淬灭反应,搅拌至无灰色固体,过滤,滤液用无水硫酸钠干燥,减压浓缩,得粗品标题化合物91b(300mg),产物不经纯化直接下一步反应。
MS m/z(ESI):139.2[M+1]
第二步
3-(溴甲基)-1-环丙基-1H-吡唑91c
将粗品化合物91b(350mg,2.53mmol)溶于5mL二氯甲烷中,滴加入三溴化磷(2.06g,7.60mmol),搅拌反应16小时。加入20mL饱和碳酸氢钠溶液淬灭反应,用二氯甲烷萃取(20mL×3),合并有机相,用无水硫酸钠干燥。过滤,滤液减压浓缩得粗品标题化合物91c(400mg),产物不经纯化直接下一步反应。
第三步
2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)丙酸叔丁酯91d
将化合物8f(100mg,246.38μmol)和粗品化合物91c(99.08mg,492.77μmol)溶于10mL四氢呋喃中,冷却至-78℃,滴加二(三甲基硅基)氨基锂溶液(0.985mL,985.53μmol),搅拌反应6小时。-78℃下,向反应液中缓慢加入2mL饱和氯化铵溶液淬灭反应,反应液温度自然升至室温。向反应液中加10mL水,用乙酸乙酯萃取(20mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物91d(90mg,产率:69.4%)。
MS m/z(ESI):526.2[M+1]
第四步
2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)丙酸91e
将化合物91d(90mg,171.1μmol)溶于5mL二氯甲烷中,滴加三氟乙酸(195.09mg,1.71mmol),搅拌反应2小时。反应液减压浓缩,得粗品标题化合物91e(80mg),产物不经纯化直接投入下一步反应。
MS m/z(ESI):470.4[M+1]
第五步
44-(2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)丙酰氨基)苯甲酸91
将粗品化合物91e(90mg,191.52μmol)和化合物8j(31.52mg,229.83μmol)溶于5mL乙酸乙酯中,依次滴加N,N-二异丙基乙胺(123.76mg,957.62μmol)和2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,365.64mg,574.57μmol),加毕,升温至60℃,搅拌反应2小时。反应液中加入15mL水,用二氯甲烷萃取(15mL×2),合并有机相,用饱和氯化钠溶液洗涤(15mL×2), 无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相制备(Waters 2767,洗脱体系:乙腈,水)纯化所得残余物,制得标题化合物91(50mg,产率:44.3%)。
MS m/z(ESI):589.3[M+1]
1H NMR(400MHz,CDCl3)δ9.96(s,1H),8.13(d,2H),7.85(d,2H),7.70(d,1H),7.50(d,1H),7.37(d,2H),6.65(s,1H),6.28(s,1H),6.15(s,1H),3.77-3.42(m,6H),2.91(s,2H),1.18-1.15(m,3H),1.10-1.05(m,4H).
实施例92
(S)-4-(2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)丙酰氨基)苯甲酸92
Figure PCTCN2017099579-appb-000149
将化合物91(32mg,54.33μmol)进行手性制备(分离条件:手性制备柱CHIRAL PAK IE,20*250mm,5um;流动相:正己烷:乙醇(含0.01%三氟乙酸)=40:60,流速:10.0mL/min),收集其相应组分,减压浓缩,得到标题化合物92(10mg)。
MS m/z(ESI):589.2[M+1]
手性HPLC分析:保留时间13.016分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,CDCl3)δ10.07(s,1H),8.12(d,2H),7.87(d,2H),7.70(d,1H),7.50(d,1H),7.37(d,1H),7.30(d,1H),6.60(s,1H),6.25(s,1H),6.12(s,1H),3.72-3.68(m,4H),3.59-3.58(m,1H),3.44-3.41(m,1H),2.88-2.86(m,2H),1.18-1.15(m,3H),1.10-1.05(m,4H).
实施例93
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)丙酰氨基)苯甲酸93
Figure PCTCN2017099579-appb-000150
Figure PCTCN2017099579-appb-000151
第一步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)丙酸叔丁酯93a
将化合物7b(100mg,255.20μmol)和粗品化合物91c(102.62mg,510.40μmol)溶于10mL四氢呋喃中,反应液冷却至-78℃,滴加二(三甲基硅基)氨基锂溶液(1.02mL,1.02mmol),搅拌反应6小时。-78℃下,向反应液中缓慢加入2mL饱和氯化铵溶液淬灭反应,自然升至室温,加10mL水,用乙酸乙酯萃取(20mL×3),合并有机相,有机相用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物93a(50mg,产率:38.3%)。
MS m/z(ESI):512.3[M+1]
第二步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)丙酸93b
将化合物93a(50mg,97.66μmol)溶于5mL二氯甲烷中,滴加三氟乙酸(111.35mg,976.57μmol),搅拌反应16小时。反应液减压浓缩,得粗品标题化合物93b(45mg),产物不经纯化直接投入下一步反应。
MS m/z(ESI):456.2[M+1]
第三步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)丙酰氨基)苯甲酸93
将粗品化合物93b(45mg,98.71μmol)和化合物8j(17.60mg,128.32μmol)溶于5mL乙酸乙酯中,依次滴加N,N-二异丙基乙胺(63.79mg,493.54μmol)和2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,188.44mg,296.12μmol),加毕,反应升温至60℃,搅拌反应2小时。反应液中冷却至室温,加入15mL水,用二氯甲烷萃取(15mL×2),合并有机相,用饱和氯化钠溶液洗涤(15mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相制备(Waters 2767-SQ  detecor2,洗脱体系:乙腈,水)纯化所得残余物,制得标题化合物93(50mg,产率:88.2%)。
MS m/z(ESI):575.1[M+1]
1H NMR(400MHz,CD3OD)δ8.00(d,2H),7.86(d,1H),7.73(d,2H),7.59-7.55(m,2H),7.37(d,2H),6.44(s,1H),6.13(s,1H),5.97-5.93(m,1H),3.64-3.47(m,5H),3.16(s,1H),2.51(s,3H),1.02-0.98(m,4H).
实施例94,95
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)丙酰氨基)苯甲酸94
(R)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)丙酰氨基)苯甲酸95
Figure PCTCN2017099579-appb-000152
将化合物93(60mg,104.35μmol)进行手性制备(分离条件:手性制备柱CHIRAL PAK IE,20*250mm,5um;流动相:正己烷:乙醇(含0.01%三氟乙酸)=30:70,流速:7.0mL/min),收集其相应组分,减压浓缩,得到标题化合物94(15mg)和化合物95(15mg)。
化合物94:
MS m/z(ESI):575.2[M+1]
手性HPLC分析:保留时间15.655分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,CD3OD)δ8.00(d,2H),7.86(d,1H),7.73(d,2H),7.59-7.55(m,2H),7.37(d,2H),6.44(s,1H),6.13(s,1H),5.97-5.93(m,1H),3.64-3.47(m,5H),3.16(s,1H),2.51(s,3H),1.02-0.98(m,4H).
化合物95:
MS m/z(ESI):575.2[M+1]
手性HPLC分析:保留时间8.787分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,CD3OD)δ8.00(d,2H),7.86(d,1H),7.73(d,2H),7.59-7.55(m,2H),7.37(d,2H),6.44(s,1H),6.13(s,1H),5.97-5.93(m,1H),3.64-3.47(m,5H),3.16(s,1H),2.51(s,3H),1.02-0.98(m,4H).
实施例96
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)-N-(喹喔啉-6-基)丙酰胺96
Figure PCTCN2017099579-appb-000153
采用实施例93的合成路线,将原料化合物8j替换为6-氨基喹喔啉(采用专利申请“WO2013006792”公开的方法制备而得),制得标题化合物96(35mg)。
MS m/z(ESI):583.4[M+1]
1H NMR(400MHz,CD3OD)δ8.80(d,2H),8.55(s,1H),8.06-7.98(m,2H),7.76(dd,1H),7.58-7.53(m,1H),7.51(d,1H),7.43(s,1H),7.30(s,1H),6.48(s,1H),6.08(s,1H),6.06-6.02(m,1H),3.65(s,3H),3.60-3.53(m,2H),3.44-3.41(m,1H),2.51(s,3H),1.02-0.98(m,4H).
实施例97
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)-N-(2-甲基-2H-吲唑-5-基)丙酰胺97
Figure PCTCN2017099579-appb-000154
采用实施例93的合成路线,将原料化合物8j替换为化合物18a,制得标题化合物97(35mg)。
MS m/z(ESI):585.3[M+1]
1H NMR(400MHz,CD3OD)δ8.16(s,1H),8.11(s,1H),7.86(d,1H),7.59-7.55(m,3H),7.42-7.33(m,3H),6.46(s,1H),6.15(d,1H),6.00-5.96(m,1H),4.21(s,3H),3.64(s,3H),3.58-3.53(m,3H),2.52(s,3H),1.02-0.98(m,4H).
实施例98
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-环丙基-1H-吡唑-3-基)-N-(喹唑啉-6-基)丙酰胺98
Figure PCTCN2017099579-appb-000155
采用实施例93的合成路线,将原料化合物8j替换为化合物23a,制得标题化合物98(30mg)。
MS m/z(ESI):583.2[M+1]
1H NMR(400MHz,MeOH-d4)δ9.50(s,1H),8.19(s,1H),8.62(d,1H),8.12(d,1H),8.03(d,1H),7.87(d,1H),7.60-7.56(m,2H),7.39-7.37(m,2H),6.46(s,1H),6.15(d,1H),6.00-5.96(m,1H),3.64(s,3H),3.63-3.54(m,3H),2.52(s,3H),1.02-0.98(m, 4H).
实施例99
4-(2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-甲基-1H-吡唑-3-基)丙酰氨基)苯甲酸99
Figure PCTCN2017099579-appb-000156
采用实施例8的合成路线,将原料化合物8g替换为3-(溴甲基)-1-甲基-1H-吡唑(采用专利申请“WO2016045125”公开的方法制备而得),制得标题化合物99(35mg)。
MS m/z(ESI):563.2[M+1]
1H NMR(400MHz,CDCl3)δ9.96(s,1H),8.13(d,2H),7.87(d,2H),7.71-7.69(m,1H),7.52-7.49(m,1H),7.35(d,1H),7.31(s,1H),6.64(s,1H),6.29-6.27(m,1H),6.17(d,1H),3.93(s,3H),3.79-3.73(m,4H),3.47-3.45(m,1H),2.91-2.89(s,2H),1.19-1.15(m,3H).
实施例100
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-甲基-1H-吡唑-3-基)丙酰氨基)苯甲酸100
Figure PCTCN2017099579-appb-000157
采用实施例74的合成路线,将原料化合物74a替换为3-(溴甲基)-1-甲基-1H-吡唑(采用专利申请“WO2016045125”公开的方法制备而得),制得标题化合物100(40mg)。
MS m/z(ESI):549.2[M+1]
1H NMR(400MHz,CD3OD)δ8.00(d,2H),7.87(d,1H),7.76-7.73(m,2H),7.60-7.59(m,1H),7.58-7.57(m,1H),7.39-7.37(m,2H),6.46(s,1H),6.15(d,1H),5.96-5.94(m,1H),3.64(s,3H),3.63(s,3H),3.55-3.51(m,1H),3.49-3.46(m,1H),2.52(s,3H).
实施例101,102
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-甲基-1H-吡唑-3-基)丙酰氨基)苯甲酸101
(R)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-甲基-1H-吡唑-3-基)丙酰氨基)苯甲酸102
Figure PCTCN2017099579-appb-000158
将化合物100(40mg,72.86μmol)进行手性制备(分离条件:手性制备柱CHIRAL PAK IE,20*250mm,5um;流动相:正己烷:乙醇(含0.01%三氟乙酸)=30:70,流速:7.0mL/min),收集其相应组分,减压浓缩,得到标题化合物101(15mg)和化合物102(15mg)。
化合物101:
MS m/z(ESI):549.2[M+1]
手性HPLC分析:保留时间16.341分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,CDCl3)δ10.03(s,1H),8.12(d,2H),7.87(d,2H),7.71(d,1H),7.50(d,1H),7.30-7.28(m,2H),6.61(s,1H),6.28-6.27(m,1H),6.13(s,1H),3.88(s,3H),3.72-3.68(m,1H),3.67(s,3H),3.43-3.41(m,1H),2.57(s,3H).
化合物102:
MS m/z(ESI):549.2[M+1]
手性HPLC分析:保留时间9.904分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,CDCl3)δ10.03(s,1H),8.12(d,2H),7.87(d,2H),7.71(d,1H),7.50(d,1H),7.30-7.28(m,2H),6.61(s,1H),6.28-6.27(m,1H),6.13(s,1H),3.88(s,3H),3.72-3.68(m,1H),3.67(s,3H),3.43-3.41(m,1H),2.57(s,3H).
实施例103
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-甲基-1H-吡唑-4-基)丙酰氨基)苯甲酸103
Figure PCTCN2017099579-appb-000159
采用实施例74的合成路线,将原料化合物74a替换为4-(溴甲基)-1-甲基-1H-吡唑(采用专利申请“WO 2015090599”公开的方法制备而得),制得标题化合物103(40mg)。
MS m/z(ESI):549.2[M+1]
1H NMR(400MHz,CDCl3)δ9.95(s,1H),8.12(d,2H),7.90(d,2H),7.75(d,1H),7.54-7.52(m,2H),7.33-7.27(m,3H),6.68(s,1H),6.08(s,1H),3.92(s,3H),3.65(s,3H),3.48-3.43(m,1H),3.24(s,1H),2.57(s,3H).
实施例104,105
(R)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-甲基-1H-吡唑-4-基)丙酰氨基)苯甲酸104
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-甲基-1H-吡唑-4-基)丙酰氨基)苯甲酸105
Figure PCTCN2017099579-appb-000160
将化合物103(300mg,546.47μmol)进行手性制备(分离条件:手性制备柱CHIRAL PAK IE,20*250mm,5um;流动相:乙醇(含0.01%)=100,流速:7.0mL/min),收集其相应组分,减压浓缩,得到标题化合物104(120mg)和化合物105(120mg)。
化合物104:
MS m/z(ESI):549.2[M+1]
手性HPLC分析:保留时间3.778分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:乙醇(含0.1%三氟乙酸)=100)。
1H NMR(400MHz,DMSO-d6)δ10.82(s,1H),7.93-7.88(m,3H),7.76(d,2H),7.65(d,1H),7.48-7.45(m,3H),7.25(s,1H),6.37(s,1H),5.82-5.80(m,1H),3.97(s,3H),3.76(s,3H),3.45-3.24(m,2H),2.49(s,3H).
化合物105:
MS m/z(ESI):549.2[M+1]
手性HPLC分析:保留时间5.535分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:乙醇(含0.1%三氟乙酸)=100)。
1H NMR(400MHz,DMSO-d6)δ10.82(s,1H),7.93-7.88(m,3H),7.76(d,2H),7.65(d,1H),7.48-7.45(m,3H),7.25(s,1H),6.37(s,1H),5.82-5.80(m,1H),3.97(s,3H),3.76(s,3H),3.45-3.24(m,2H),2.49(s,3H).
实施例106
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(异噁唑-5-基)丙酰氨基)苯甲酸106
Figure PCTCN2017099579-appb-000161
采用实施例74的合成路线,将原料化合物74a替换为5-溴甲基异噁唑(采用公知的方法“Journal of Medicinal Chemistry,2016,59(7),3471-3488”制备而得),制 得标题化合物106(55mg)。
MS m/z(ESI):536.4[M+1]
1H NMR(400MHz,DMSO-d6)δ10.84(s,1H),8.47-8.46(s,1H),7.93-7.94(m,1H),7.92-7.91(m,1H),7.86-7.85(d,1H),7.77-7.76(m,1H),7.75-7.74(m,1H),7.64-7.61(dd,1H),7.44-7.43(d,1H),7.38(s,1H),6.39(s,1H),6.23-6.22(d,1H),6.05-6.01(m,1H),3.89-3.82(m,1H),3.73-3.71(m,1H),3.52(s,3H),2.45(s,3H)
实施例107
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(噻唑-2-基)丙酰氨基)苯甲酸107
Figure PCTCN2017099579-appb-000162
采用实施例74的合成路线,将原料化合物74a替换为2-溴甲基噻唑(采用专利申请“WO2014065413”公开的方法制备而得),制得标题化合物107(20mg)。
MS m/z(ESI):551.9[M+1]
1H NMR(400MHz,CD3OD)δ7.98(d,2H),7.86(d,1H),7.75-7.71(m,3H),7.57(dd,1H),7.52(d,1H),7.37-7.36(m,2H),6.47(s,1H),6.06-6.02(m,1H),4.06-3.91(m,2H),3.60(s,3H),2.51(s,3H).
实施例108,109
(S)-4-(4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)丁酰氨基)苯甲酸108
(R)-4-(4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)丁酰氨基)苯甲酸109
Figure PCTCN2017099579-appb-000163
Figure PCTCN2017099579-appb-000164
第一步
2-(叔丁氧基)乙基三氟甲磺酸酯108b
将2-叔丁氧基乙醇108a(300mg,2.54mmol)溶于8mL二氯甲烷中,冰浴下加入2,6-二甲基吡啶(299.22mg,2.79mmol),滴加三氟甲磺酸酐(787.87mg,2.79mmol),滴加完后在冰浴下搅拌反应1小时,自然升温至室温搅拌反应1小时。反应液中加入30mL二氯甲烷,用20mL水洗涤,有机相用无水硫酸钠干燥,过滤,减压浓缩得粗品标题化合物108b(550mg),产物不经纯化直接下步反应。
第二步
4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)丁酸叔丁酯108c
将化合物8f(148mg,364.65μmol)和粗品化合物108b(182.50mg,729.30μmol)溶于15mL四氢呋喃中,反应液冷却至-78℃,滴加二(三甲基硅基)氨基锂溶液(1.46mL,1.46mmol),搅拌反应2小时。-78℃下,向反应液中缓慢加入5mL水淬灭反应,反应液温度自然升至室温,加入20mL水,用乙酸乙酯萃取(35mL×3),合并有机相,用饱和氯化钠溶液洗涤(25mL×2),无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题化合物108c(120mg,产率:65.0%)。
MS m/z(ESI):506.5[M+1]
第三步
4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)丁酸108d
将化合物108c(120mg,237.14μmol)溶于8mL乙醇和4mL四氢呋喃的混合溶剂中,加入氢氧化锂(49.75mg,1.19mmol),升温至50℃搅拌反应2小时。反应液冷却至室温,减压浓缩旋去大部分有机溶剂,加入15mL水,用3M盐酸调 节pH至3,乙酸乙酯萃取(20mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥,过滤,滤液浓缩,得到粗品标题化合物108d(106mg),产物不经纯化直接下步反应。
MS m/z(ESI):450.4[M+1]
第四步
4-(4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)丁酰氨基)苯甲酸108e
将粗品化合物108d(106mg,235.59μmol)溶于15mL乙酸乙酯中,依次加入N,N-二异丙基乙胺(304.48mg,2.36mmol)、化合物8j(35.54mg,259.16μmol)和2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,599.70mg,942.38μmol),加毕,反应升温至80℃,搅拌反应2小时。反应液冷却至室温,加入20mL水,用3M盐酸调节pH至5,乙酸乙酯萃取(20mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相制备(Waters 2767-SQ detecor2,洗脱体系:乙腈,水)纯化所得残余物,制得标题化合物108e(60mg,产率:44.8%)。
MS m/z(ESI):569.5[M+1]
第五步
(S)-4-(4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)丁酰氨基)苯甲酸108
(R)-4-(4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)丁酰氨基)苯甲酸109
将化合物108e(60mg,105.44μmol)进行手性制备(分离条件:色谱柱Superchiral S-AD(Chiralway),2cm I.D.*25cm Length,5um;流动相:二氧化碳:乙醇:二乙胺=60:40:0.05,流速:50g/min),收集其相应组分,减压浓缩,得到标题化合物108(22mg)和化合物109(22mg)。
化合物108:
MS m/z(ESI):569.5[M+1]
手性HPLC分析:保留时间8.518分钟,手性纯度100%(色谱柱:CHIRAL PAK IE4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ12.70(s,1H),10.73(s,1H),7.90(d,2H),7.86(d,1H),7.78(d,2H),7.62(dd,1H),7.41(s,1H),7.27(s,1H),6.39(s,1H),5.76-5.72(m,1H),3.52(s,3H),3.39-3.36(m,2H),2.99-2.86(m,2H),2.36-2.27(m,2H),1.06(s,9H),1.00(t,3H).
化合物109:
MS m/z(ESI):569.4[M+1]
手性HPLC分析:保留时间5.172分钟,手性纯度99.7%(色谱柱:CHIRAL PAK IE4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ10.71(s,1H),7.91-7.84(m,3H),7.77(d,2H),7.62(dd,1H),7.41(s,1H),7.27(s,1H),6.39(s,1H),5.76-5.72(m,1H),3.52(s,3H),3.39-3.36(m,2H),2.99-2.86(m,2H),2.36-2.27(m,2H),1.06(s,9H),1.00(t,3H).
实施例110
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)丁酰氨基)苯甲酸110
Figure PCTCN2017099579-appb-000165
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,制得标题化合物110(30mg)。
MS m/z(ESI):555.1[M+1]
1H NMR(400MHz,CD3OD)δ8.01(d,2H),7.88(d,1H),7.75(d,2H),7.58(dd,1H),7.37(d,1H),7.36(s,1H),6.52(s,1H),5.90-5.87(m,1H),3.65(s,3H),3.57-3.43(m,2H),2.55(s,3H),2.49-2.36(m,2H),1.18(s,9H).
实施例111,112
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)丁酰氨基)苯甲酸111
(R)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)丁酰氨基)苯甲酸112
Figure PCTCN2017099579-appb-000166
将化合物110(1.2g,2.16mmol)进行手性制备(分离条件:色谱柱:Superchiral S-AD(Chiralway),2cm I.D.*25cm Length,5um;流动相:二氧化碳:乙醇:二乙胺=60:40:0.05,流速:50g/min),收集其相应组分,减压浓缩,得到标题化合物111(500mg)和化合物112(450mg)。
化合物111:
MS m/z(ESI):555.1[M+1]
手性HPLC分析:保留时间16.803分钟,手性纯度100%(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=70/30(v/v))。
1H NMR(400MHz,CD3OD)δ8.03-7.99(m,2H),7.89(d,1H),7.76-7.74(m,2H),7.60(dd,1H),7.39(d,1H),7.36(s,1H),6.52(s,1H),5.91-5.87(m,1H),3.66(s,3H), 3.60-3.54(m,1H),3.47-3.42(m,1H),2.55(s,3H),2.52-2.45(m,1H),2.42-2.37(m,1H),1.18(s,9H).
化合物112:
MS m/z(ESI):555.1[M+1]
手性HPLC分析:保留时间4.247分钟,手性纯度100%(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=70/30(v/v))。
1H NMR(400MHz,CD3OD)δ8.03-7.99(m,2H),7.89(d,1H),7.76-7.74(m,2H),7.60(dd,1H),7.39(d,1H),7.36(s,1H),6.52(s,1H),5.91-5.87(m,1H),3.66(s,3H),3.60-3.54(m,1H),3.47-3.42(m,1H),2.55(s,3H),2.52-2.45(m,1H),2.42-2.37(m,1H),1.18(s,9H).
实施例113
4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(喹喔啉-6-基)丁酰胺113
Figure PCTCN2017099579-appb-000167
采用实施例108中化合物108e的合成路线,将第四步原料化合物8j替换为6-氨基喹喔啉(采用专利申请“WO2013006792”公开的方法制备而得),制得标题化合物113(35mg)。
MS m/z(ESI):577.3[M+1]
1H NMR(400MHz,CD3OD)δ8.85-8.83(d,1H),8.80-8.79(d,1H),8.61-8.60(m,1H),8.08-8.06(d,1H),8.02-7.97(dd,1H),7.85-7.83(d,1H),7.58-7.55(dd,1H),7.40-7.35(m,2H),6.50(s,1H),5.95-5.85(m,1H),3.65-3.60(m,1H),3.60-3.55(s,3H),3.50-3.40(m,1H),3.00-2.95(m,2H),2.50-2.40(m,1H),2.35-2.25(m,1H),1.17(s,9H),1.10-1.00(m,3H)
实施例114,115
(S)-4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(喹喔啉-6-基)丁酰胺114
(R)-4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(喹喔啉-6-基)丁酰胺115
Figure PCTCN2017099579-appb-000168
将化合物113(65mg,112.64μmol)进行手性制备(分离条件:色谱柱Superchiral  S-AD(Chiralway),2.1cm I.D.*25cm Length,5um;流动相:乙醇:乙腈:二乙胺=15:85:0.05,流速:1.0mL/min),收集其相应组分,减压浓缩,得到标题化合物114(20mg)和化合物115(20mg)。
化合物114:
MS m/z(ESI):577.3[M+1]
手性HPLC分析:保留时间17.031分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=30/70(v/v))。
1H NMR(400MHz,CD3OD)δ10.50(s,1H),8.85-8.83(d,1H),8.80-8.79(d,1H),8.61-8.60(m,1H),8.08-8.06(d,1H),8.02-7.97(dd,1H),7.85-7.83(d,1H),7.58-7.55(dd,1H),7.40-7.35(m,2H),6.50(s,1H),5.95-5.85(m,1H),3.65-3.60(s,3H),3.60-3.55(m,1H),3.50-3.40(m,1H),3.00-2.95(m,2H),2.50-2.40(m,1H),2.35-2.25(m,1H),1.17(s,9H),1.10-1.00(m,3H).
化合物115:
MS m/z(ESI):577.3[M+1]
手性HPLC分析:保留时间7.416分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=30/70(v/v))。
1H NMR(400MHz,CD3OD)δ10.50(s,1H),8.85-8.83(d,1H),8.80-8.79(d,1H),8.61-8.60(m,1H),8.08-8.06(d,1H),8.02-7.97(dd,1H),7.85-7.83(d,1H),7.58-7.55(dd,1H),7.40-7.35(m,2H),6.50(s,1H),5.95-5.85(m,1H),3.65-3.60(m,1H),3.60-3.55(s,3H),3.50-3.40(m,1H),3.00-2.95(m,2H),2.50-2.40(m,1H),2.35-2.25(m,1H),1.17(s,9H),1.10-1.00(m,3H).
实施例116
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(1-氧代异吲哚啉-5-基)丁酰胺116
Figure PCTCN2017099579-appb-000169
采用实施例108中化合物108e的合成路线,将第二步原料化合物8f替换为原料化合物7b,第四步原料化合物8j替换为5-氨基异吲哚啉-1-酮(采用专利申请“WO 2012092880”公开的方法制备而得),制得标题化合物116(35mg)。
MS m/z(ESI):566.1[M+1]
1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.89(d,1H),7.76(d,1H),7.65(d,1H),7.59(d,1H),7.39(s,1H),7.37(s,1H),6.52(m,1H),5.91-5.87(m,1H),4.46(s,2H),3.66(s,3H),3.41-3.58(m,2H),2.55(s,3H),2.30-2.51(m,2H),1.18(s,9H).
实施例117,118
(R)-2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基-N-(1'-氧代 -1'H-螺[环丁烷-1,3'-噁唑并[3,4-a]吲哚]-7'-基)丁酰胺117
(S)-2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基-N-(1'-氧代-1'H-螺[环丁烷-1,3'-噁唑并[3,4-a]吲哚]-7'-基)丁酰胺118
Figure PCTCN2017099579-appb-000170
第一步
(1'-氧代-1'H-螺[环丁烷-1,3'-噁唑并[3,4-a]吲哚]-7'-基)氨基甲酸叔丁酯117b
将5-((叔丁氧基羰基)氨基)-1H-吲哚-2-羧酸117a(4.5g,16.29mmol,采用专利申请“WO2012162482”公开的方法制备而得)溶于160mL四氢呋喃中,冰浴冷却下,加入N,N’-羰基二咪唑(5.82g,32.57mmol),升至室温搅拌反应1.5小时后,冷却至0℃,滴加入环丁酮(2.85g,40.72mmol)和1,8-二氮杂双环[5.4.0]十一碳-7-烯(6.44g,42.35mmol),0℃搅拌反应30分钟后,升至室温搅拌反应2小时。反应液减压浓缩除去大部分四氢呋喃,残余物倒入150mL冰水中,加3M盐酸调节pH至5左右,用乙酸乙酯萃取(50mL×3),合并有机相,用饱和氯化钠溶液洗涤(40mL×2),无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物117b(2.7g,产率:50.5%)。
MS m/z(ESI):329.5[M+1]
第二步
7'-氨基-1'H-螺[环丁烷-1,3'-噁唑并[3,4-a]吲哚]-1'-酮盐酸盐117c
将化合物117b(4.9g,14.92mmol)溶于30mL四氢呋喃中,加入4M氯化氢的1,4-二氧六环溶液(22.38mL,89.54mmol),升温至45℃,搅拌反应5小时。反应液减压浓缩,所得残余物中加入40mL乙酸乙酯和正己烷的混合溶剂(V/V=1:5),搅拌,过滤,收集滤饼,得粗品标题化合物117c(3.9g),产物不经纯化直接下一步反应。
MS m/z(ESI):229.4[M+1]
第三步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基-N-(1'-氧代-1'H-螺[环丁烷-1,3'-噁唑并[3,4-a]吲哚]-7'-基)丁酰胺117d
将化合物1g(400mg,1.02mmol)加入到12mL N,N-二甲基甲酰胺中,再加入O-(7-氮杂苯并三唑-1-基)-N,N,N′,N′-四甲基脲六氟磷酸酯(578.94mg,1.52mmol)、N,N-二异丙基乙胺(0.708mL,4.06mmol)和粗品化合物117c(268.86mg,1.02mmol),加毕,加热至40℃,搅拌反应16小时。向反应液中加入50mL饱和碳酸氢钠溶液,用乙酸乙酯(100mL×2)萃取,合并有机相,用饱和氯化钠溶液(50mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物117d(400mg,产率58.7%)。
MS m/z(ESI):604.5[M+1]
第四步
(R)-2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基-N-(1'-氧代-1'H-螺[环丁烷-1,3'-噁唑并[3,4-a]吲哚]-7'-基)丁酰胺117
(S)-2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基-N-(1'-氧代-1'H-螺[环丁烷-1,3'-噁唑并[3,4-a]吲哚]-7'-基)丁酰胺118
将化合物117d(510mg,0.84mmol)进行手性制备(分离条件:手性制备柱CHIRAL PAK IE,20*250mm,5um;流动相:乙醇=100,流速:8.0mL/min),收集其相应组分,减压浓缩,得到标题化合物117(150mg)和化合物118(135mg)。
化合物117:
MS m/z(ESI):604.6[M+1]
手性HPLC分析:保留时间8.666分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:乙醇)。
1H NMR(400MHz,CDCl3)δ9.55(s,1H),8.20(s,1H),7.70-7.68(d,1H),7.54-7.49(m,2H),7.48-7.47(dd,1H),7.29-7.28(d,1H),7.05(s,1H),7.00(s,1H),6.67(s,1H),5.95-5.80(m,1H),3.60(s,3H),3.57-3.54(m,2H),3.37(s,3H),3.06-3.02(m,2H),2.92-2.87(m,2H),2.72-2.62(m,1H),2.50(s,3H),2.35-2.15(m,3H).
化合物118:
MS m/z(ESI):604.5[M+1]
手性HPLC分析:保留时间11.473分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:乙醇)。
1H NMR(400MHz,CDCl3)δ9.55(s,1H),8.20(s,1H),7.70-7.68(d,1H),7.54-7.49(m,2H),7.48-7.47(dd,1H),7.29-7.28(d,1H),7.05(s,1H),7.00(s,1H),6.67(s,1H),5.95-5.80(m,1H),3.60(s,3H),3.57-3.54(m,2H),3.37(s,3H),3.06-3.02(m,2H),2.92-2.87(m,2H),2.72-2.62(m,1H),2.50(s,3H),2.35-2.15(m,3H).
实施例119
5-(4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)丁酰氨基)-1H-吲哚-2-羧酸119
Figure PCTCN2017099579-appb-000171
采用实施例108中化合物108e的合成路线,将原料化合物8j替换为5-氨基-1H-吲哚-2-羧酸(采用公知的方法“Journal of the American Chemical Society,2006,128(37),12162-12168”制备而得),制得标题化合物119(20mg)。
MS m/z(ESI):608.6[M+1]
1H NMR(400MHz,DMSO-d6)δ11.32(s,1H),10.25(s,1H),7.96(s,1H),7.85(d,1H),7.62(dd,1H),7.41(s,1H),7.35-7.28(m,3H),6.80(s,1H),6.39(s,1H),5.78-5.74(m,1H),3.52(s,3H),3.29-3.25(m,2H),2.98-2.85(m,2H),2.35-2.23(m,2H),1.08(s,9H),1.00(t,3H).
实施例120
44-(2-(4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)丁酰氨基)苯甲酸120
Figure PCTCN2017099579-appb-000172
第一步
2-(4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)乙酸叔丁酯120a
将化合物30c(300mg,1.01mmol)、化合物7a(217.68mg,1.12mmol)和碳酸铯(661.13mg,2.03mmol)溶于10mL N,N-二甲基甲酰胺中,升温至65℃,搅拌反应2小时。反应液冷却至室温,反应液中加入20mL水,用乙酸乙酯萃取(20mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系C纯化所得残余物得标题化合物120a(300 mg,产率:72.1%)。
MS m/z(ESI):410.4[M+1]
第二至四步
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物120a,制得标题化合物120(35mg)。
MS m/z(ESI):573.5[M+1]
1H NMR(400MHz,DMSO-d6)δ10.77(s,1H),7.91-7.89(m,2H),7.83-7.76(m,4H),7.37(d,1H),6.40(d,1H),5.82-5.76(m,1H),3.59(s,3H),3.41-3.36(m,1H),3.29-3.23(m,1H),2.47(d,3H),2.38-2.32(m,2H),1.04(d,9H).
实施例121
4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(喹唑啉-6-基)丁酰胺121
Figure PCTCN2017099579-appb-000173
采用实施例108中化合物108e的合成路线,将原料化合物8j替换为化合物23a,制得标题化合物121(20mg)。
MS m/z(ESI):577.1[M+1]
1H NMR(400MHz,CD3OD)δ8.57(s,1H),7.93(dd,1H),7.87(d,1H),7.80-7.73(m,1H),7.60(dd,1H),7.39-7.38(m,1H),7.34(s,1H),7.31(d,1H),6.51(s,1H),6.21(s,1H),5.83-5.78(m,1H),3.65(s,3H),3.58-3.40(m,2H),3.03-2.97(m,2H),2.54-2.40(m,2H),1.18(s,9H),1.12(t,3H).
实施例122
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)丁酰氨基)苯甲酰胺122
Figure PCTCN2017099579-appb-000174
采用实施例11的合成路线,将原料化合物5替换为化合物111,制得标题化合物122(40mg)。
MS m/z(ESI):554.1[M+1]
1H NMR(400MHz,DMSO-d6)δ10.63(s,1H),7.90-7.83(m,4H),7.73(d,2H),7.63(dd,1H),7.42(s,1H),7.29-7.27(m,2H),6.41(s,1H),5.79-5.76(m,1H),3.55(s,3H),3.30-3.28(m,2H),2.55(s,3H),2.36-2.28(m,2H),1.07(s,9H).
实施例123
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)丁酰氨基)-N-甲基苯甲酰胺123
Figure PCTCN2017099579-appb-000175
采用实施例13的合成路线,将原料化合物5替换为化合物111,制得标题化合物123(40mg)。
MS m/z(ESI):568.1[M+1]
1H NMR(400MHz,CDCl3)δ9.73(br,1H),7.76-7.65(m,4H),7.51(d,1H),7.32-7.31(m,1H),6.98(s,1H),6.66(s,1H),6.26-6.24(m,1H),5.84-5.79(m,1H),3.62(s,3H),3.54-3.52(m,2H),3.03(d,1H),2.66-2.61(m,1H),2.53(s,3H),2.33-2.25(m,2H),1.20(s,9H).
实施例124
4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(2,3-二甲基喹喔啉-6-基)丁酰胺124
Figure PCTCN2017099579-appb-000176
采用实施例108中化合物108e的合成路线,将第四步原料化合物8j替换为2,3-二甲基喹喔啉-6-胺(采用公知的方法“Bioorganic & Medicinal Chemistry Letters,2012,20(7),2227-2234”制备而得),制得标题化合物124(15mg)。
MS m/z(ESI):605.3[M+1]
1H NMR(400MHz,CD3OD)δ8.45-8.42(d,1H),7.92-7.90(d,1H),7.90-7.86(dd,1H),7.85-7.82(d,1H),7.57-7.52(dd,1H),7.37(s,2H),6.50(s,1H),5.95-5.85(m,1H),3.65-3.60(s,3H),3.60-3.55(m,1H),3.50-3.40(m,1H),3.00-2.95(m,2H),2.70(s,6H),2.50-2.40(m,1H),2.35-2.25(m,1H),1.17(s,9H),1.10-1.00(m,3H).
实施例125
4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(2-氰基-1H-吲哚-6-基)丁酰胺125
Figure PCTCN2017099579-appb-000177
采用实施例108中化合物108e的合成路线,将原料化合物8j替换为6-氨基-1H-吲哚-2-甲腈(采用专利申请“WO20160271105”制备而得),制得标题化合物125(30mg)。
MS m/z(ESI):589.5[M+1]
1H NMR(400MHz,CD3OD)δ8.04(s,1H),7.85-7.83(d,1H),7.61-7.59(d,1H),7.58-7.57(dd,1H),7.38-7.37(d,1H),7.36(s,1H),7.25-7.18(dd,1H),7.16-7.15(d,1H),6.50(s,1H),5.95-5.90(m,1H),3.63(s,3H),3.60-3.55(m,1H),3.50-3.40(m,1H),3.00-2.95(m,2H),2.50-2.40(m,1H),2.35-2.25(m,1H),1.17(s,9H),1.10-1.00(m,3H).
实施例126
N-(苯并[d]噻唑-5-基)-4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)丁酰胺126
Figure PCTCN2017099579-appb-000178
采用实施例108中化合物108e的合成路线,将原料化合物8j替换为苯并[d]噻唑-5-胺(采用专利申请“WO2013142266”制备而得),制得标题化合物126(35mg)。
MS m/z(ESI):582.2[M+1]
1H NMR(400MHz,CD3OD)δ9.24(s,1H),8.50(s,1H),8.03-8.00(d,1H),7.84-7.82(d,1H),7.68-7.69(d,1H),7.57-7.54(dd,1H),7.38-7.35(m,2H),6.50(s,1H),5.95-5.85(m,1H),3.62(s,3H),3.60-3.55(m,1H),3.50-3.40(m,1H),3.00-2.80(m,2H),2.50-2.40(m,1H),2.35-2.25(m,1H),1.17(s,9H),1.11-1.09(m,3H).
实施例127
4-(4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)丁酰氨基)噻吩-2-羧酸127
Figure PCTCN2017099579-appb-000179
采用实施例108中化合物108e的合成路线,将原料化合物8j替换为4-氨基噻吩-2-羧酸(采用专利申请“Journal of the American Chemical Society,1999,121(34),7751-7759”制备而得),制得标题化合物127(20mg)。
MS m/z(ESI):575.3[M+1]
1H NMR(400MHz,CD3OD)δ7.85(s,1H),7.85-7.84(d,1H),7.82-7.80(d,1H),7.58-7.55(d,1H),7.38-7.36(d,1H),7.33-7.31(s,1H),6.48(s,1H),5.95-5.85(m,1H),3.65-3.60(m,3H),3.60-3.55(m,1H),3.50-3.40(m,1H),3.00-2.95(m,2H),2.50-2.40(m,1H),2.35-2.25(m,1H),1.17(s,9H),1.10-1.00(m,3H).
实施例128
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(3,4-二氢-2H-苯并[b][1,4]噁嗪-6-基)丁酰胺128
Figure PCTCN2017099579-appb-000180
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,原料化合物8j替换为3,4-二氢-2H-苯并[b][1,4]噁嗪-6-胺(采用公知的方法“Bioorganic & Medicinal Chemistry Letters,2015,25(10),2122-2128”制备而得),制得标题化合物128(23mg)。
MS m/z(ESI):568.1[M+1]
1H NMR(400MHz,CDCl3)δ8.98(s,1H),7.69(d,1H),7.51-7.48(dd,1H),7.32(d,1H),7.07(d,1H),6.95(s,1H),6.75-6.71(m,2H),6.60(s,1H),5.80(s,1H),4.25(t,2H),3.59(s,3H),3.52(t,2H),3.43(t,2H),2.63-2.59(m,1H),2.50(s,3H),2.22-2.18(m,1H),1.22(s,9H).
实施例129
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(3-氟苯基)丁酰胺129
Figure PCTCN2017099579-appb-000181
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,原料化合物8j替换为3-氟苯胺,制得标题化合物129(20mg)。
MS m/z(ESI):529.2[M+1]
1H NMR(400MHz,CDCl3)δ9.67(br,1H),8.01-8.00(m,1H),7.75-7.70(m,2H),7.52-7.45(m,2H),7.40-7.38(m,1H),7.34-7.33(m,1H),6.91(s,1H),6.64(s,1H),5.79(br,1H),3.61(s,3H),3.56-3.53(m,2H),2.70-2.62(m,1H),2.53(s,3H),2.27-2.23(m,1H),1.22(m,9H).
实施例130
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(5-氯吡啶-3-基)丁酰胺130
Figure PCTCN2017099579-appb-000182
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,原料化合物8j替换为5-氯吡啶-3-胺(采用专利申请“WO2006067445”公开的方法制备而得),制得标题化合物130(25mg)。
MS m/z(ESI):546.0[M+1]
1H NMR(400MHz,CD3OD)δ8.68(d,1H),8.33(m,1H),8.31(d,1H),7.89(d,1H),7.58(dd,1H),7.38(d,1H),7.33(s,1H),6.51(s,1H),5.86-5.81(m,1H),3.65(s,3H),3.58-3.40(m,2H),2.56(s,3H),2.53-2.39(m,2H),1.18(s,9H).
实施例131
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(5-氟吡啶-3-基)丁酰胺131
Figure PCTCN2017099579-appb-000183
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合 物7b,原料化合物8j替换为5-氟吡啶-3-胺(采用公知的方法“Journal of Medicinal Chemistry,1999,42(18),3701-3710”制备而得),制得标题化合物131(25mg)。
MS m/z(ESI):530.1[M+1]
1H NMR(400MHz,CD3OD)δ8.59(s,1H),8.24(d,1H),8.18-8.14(m,1H),7.89(d,1H),7.59(dd,1H),7.38(d,1H),7.33(s,1H),6.51(s,1H),5.86-5.81(m,1H),3.65(s,3H),3.58-3.40(m,2H),2.56(s,3H),2.53-2.39(m,2H),1.18(s,9H).
实施例132
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(4-氟苯基)丁酰胺132
Figure PCTCN2017099579-appb-000184
采用实施例108中化合物108e的合成路线,将第二步原料化合物8f替换为原料化合物7b,第四步原料化合物8j替换为4-氟苯胺,制得标题化合物132(20mg)。
MS m/z(ESI):529.2[M+1]
1H NMR(400MHz,CDCl3)δ9.37(br,1H),7.72-7.70(d,1H),7.59-7.56(m,2H),7.52-7.49(m,1H),7.33-7.32(m,1H),7.07-7.02(m,2H),6.95(s,1H),6.62(s,1H),5.78(br,1H),3.60(s,3H),3.54-3.52(m,2H),2.66-2.60(m,1H),2.52(s,3H),2.26-2.21(m,1H),1.22(m,9H).
实施例133
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(3,4-二氢-2H-苯并[b][1,4]噁嗪-7-基)丁酰胺133
Figure PCTCN2017099579-appb-000185
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,原料化合物8j替换为3,4-二氢-2H-苯并[b][1,4]噁嗪-7-胺(采用公知的方法“Journal of Medicinal Chemistry,2005,48(1),71-90”制备而得),制得标题化合物133(19mg)。
MS m/z(ESI):568.5[M+1]
1H NMR(400MHz,CDCl3)δ8.91(s,1H),7.69(d,1H),7.50-7.48(dd,1H),7.32(d,1H),7.09(d,1H),6.97(s,1H),6.94(d,1H),6.60(s,1H),6.56(d,1H),5.78(s,1H),4.26(t,2H),3.59(s,3H),3.42(t,2H),3.51(t,2H),2.60-2.57(m,1H),2.50(s,3H),2.27-2.24(m,1H),1.22(s,9H).
实施例134
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(6-甲氧基吡啶-3-基)丁酰胺134
Figure PCTCN2017099579-appb-000186
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,原料化合物8j替换为6-甲氧基吡啶-3-胺(采用公知的方法“Tetrahedron Letters,2010,51(5),786-789”制备而得),制得标题化合物134(25mg)。
MS m/z(ESI):542.1[M+1]
1H NMR(400MHz,CD3OD)δ8.38(s,1H),7.95-7.91(m,1H),7.89(d,1H),7.59(dd,1H),7.38(d,1H),7.34(s,1H),6.83(d,1H),6.52(s,1H),5.85-5.82(m,1H),3.91(s,3H),3.65(s,3H),3.58-3.40(m,2H),2.55(s,3H),2.51-2.33(m,2H),1.19(s,9H).
实施例135
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(3-(三氟甲基)苯基)丁酰胺135
Figure PCTCN2017099579-appb-000187
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,原料化合物8j替换为3-(三氟甲基)苯胺(采用公知的方法“Journal of Organic Chemistry,2016,81(12),5120-5127”制备而得),制得标题化合物135(15mg)。
MS m/z(ESI):579.2[M+1]
1H NMR(400MHz,CDCl3)δ9.53(br,1H),7.72-7.70(m,1H),7.61-7.58(m,1H),7.52-7.49(m,1H),7.33-7.32(m,1H),7.30-7.21(m,2H),6.92(s,1H),6.86-6.82(m,1H),6.63(s,1H),5.78(br,1H),3.60(s,3H),3.55-3.52(m,2H),2.68-2.60(m,1H),2.52(s,3H),2.26-2.21(m,1H),1.22(s,9H).
实施例136
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(4-(三氟甲基)苯基)丁酰胺136
Figure PCTCN2017099579-appb-000188
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,原料化合物8j替换为4-(三氟甲基)苯胺(采用公知的方法“Journal of Organic Chemistry,2009,74(12),4542-4546”制备而得),制得标题化合物136(20mg)。
MS m/z(ESI):579.2[M+1]
1H NMR(400MHz,CDCl3)δ9.70(br,1H),7.76-7.72(m,3H),7.62-61(m,2H),7.52-7.51(m,1H),7.34-7.32(m,1H),6.92-6.90(m,1H),6.65-6.63(m,1H),5.80-5.77(m,1H),3.61-3.60(m,3H),3.54(br,2H),2.67-2.66(m,1H),2.54-2.52(m,3H),2.28-2.24(m,1H),1.23-1.21(m,9H).
实施例137
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(2,3-二氢苯并[b][1,4]二噁英-5-基)丁酰胺137
Figure PCTCN2017099579-appb-000189
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,原料化合物8j替换为2,3-二氢苯并[b][1,4]二噁英-5-胺(采用专利申请“WO2012092880”公开的方法制备而得),制得标题化合物137(30mg)。
MS m/z(ESI):569.2[M+1]
1H NMR(400MHz,CDCl3)δ8.90(s,1H),7.90(d,1H),7.69(d,1H),7.48(d,1H),7.33(d,1H),7.00(s,1H),6.82(t,1H),6.67(dd,1H),6.61(s,1H),5.89-5.85(m,1H),4.42-4.39(m,2H),4.32-4.30(m,2H),3.60(s,3H),3.52-3.49(m,2H),2.61-2.57(m,1H),2.51(s,3H),2.25-2.12(m,1H),1.21(m,9H).
实施例138
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(3,4-二氢-2H-苯并[b][1,4]噁嗪-5-基)丁酰胺138
Figure PCTCN2017099579-appb-000190
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合 物7b,原料化合物8j替换为3,4-二氢-2H-苯并[b][1,4]噁嗪-5-胺(采用公知的方法“Journal of Medicinal Chemistry,2017,60(6),2401-2410”制备而得),制得标题化合物138(21mg)。
MS m/z(ESI):568.1[M+1]
1H NMR(400MHz,CDCl3)δ8.76(s,1H),7.70(d,1H),7.52-7.49(dd,1H),7.32(d,1H),7.13-7.11(m,1H),6.94(s,1H),6.72-6.70(m,2H),6.61(s,1H),5.70(s,1H),4.23-4.19(m,2H),3.60(s,3H),3.58-3.55(m 2H),3.48(s,2H),2.68-2.62(m,1H),2.52(s,3H),2.28-2.22(m,1H),1.23(s,9H).
实施例139
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(2,3-二氢苯并[b][1,4]二噁英-6-基)丁酰胺139
Figure PCTCN2017099579-appb-000191
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,原料化合物8j替换为2,3-二氢苯并[b][1,4]二噁英-6-胺(采用公知的方法“Chemical Communications,2012,48(64),7982-7984”制备而得),制得标题化合物139(30mg)。
MS m/z(ESI):569.2[M+1]
1H NMR(400MHz,CDCl3)δ9.05(s,1H),7.70(d,1H),7.50(d,1H),7.33(s,1H),7.26(s,1H),6.98(d,1H),6.94(s,1H),6.82(d,1H),6.61(s,1H),5.78-5.77(m,1H),4.27(s,4H),3.60(s,3H),3.51-3.52(m,2H),2.61-2.62(m,1H),2.51(s,3H),2.22-2.26(s,1H),1.22(m,9H).
实施例140
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(苯并[d][1,3]二氧杂环戊烯-5-基)-4-(叔丁氧基)丁酰胺140
Figure PCTCN2017099579-appb-000192
采用实施例108中化合物108e的合成路线,将第二步原料化合物8f替换为原料化合物7b,第四步原料化合物8j替换为苯并[d][1,3]二氧杂环戊烯-5-胺(采用专利申请“CN105348251”公开的方法制备而得),制得标题化合物140(25mg)。
MS m/z(ESI):555.1[M+1]
1H NMR(400MHz,CD3OD)δ7.89(d,1H),7.59(d,1H),7.39(s,1H),7.35(s,1H), 7.06(s,1H),6.97(d,1H),6.79(d,1H),6.52(s,1H),5.85-5.81(m,1H),3.65(s,3H),3.55-3.42(m,2H),2.55(s,3H),2.44-2.34(m,2H),1.19(s,9H).
实施例141
4-(叔丁氧基)-2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(2-甲基-2H-吲唑-5-基)丁酰胺141
Figure PCTCN2017099579-appb-000193
采用实施例108中化合物108e的合成路线,将原料化合物8j替换为化合物18a,制得标题化合物141(25mg)。
MS m/z(ESI):579.1[M+1]
1H NMR(400MHz,CD3OD)δ7.89(d,1H),7.60(d,1H),7.41-7.39(m,2H),7.35(s,1H),7.21(d,1H),7.12(d,1H),6.52(s,1H),5.87-5.84(m,1H),3.66(s,3H),3.62-3.52(m,1H),3.51(s,2H),3.47-3.43(m,1H),2.55(s,3H),2.53-2.37(m,2H),1.35-1.31(m,2H),1.19(s,9H).
实施例142
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(1H-吲哚-4-基)丁酰胺142
Figure PCTCN2017099579-appb-000194
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,原料化合物8j替换为1H-吲哚-4-胺(采用公知的方法“Journal of Medicinal Chemistry,2005,48(9),3417-3427”制备而得),制得标题化合物142(25mg)。
MS m/z(ESI):550.1[M+1]
1H NMR(400MHz,CD3OD)δ7.89(d,1H),7.59(d,1H),7.45(d,1H),7.40(s,1H),7.38(d,2H),7.28-7.26(m,2H),7.10(t,1H),6.63-6.61(m,1H),6.57(s,1H),6.03-5.99(m,1H),3.66(s,3H),3.62-3.47(m,2H),2.55(s,3H),2.54-2.40(m,2H),1.20(s,9H).
实施例143
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(1-氧代-1,2,3,4-四氢异喹啉-6-基)丁酰胺143
Figure PCTCN2017099579-appb-000195
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,原料化合物8j替换为6-氨基-3,4-二氢异喹啉-1(2H)-酮(采用专利申请“CN103804358”公开的方法制备而得),制得标题化合物143(20mg)。
MS m/z(ESI):580.6[M+1]
1H NMR(400MHz,CDCl3)δ9.69(s,1H),8.05(d,1H),7.70(d,1H),7.64(d,1H),7.51-7.47(m,2H),7.32(d,1H),6.91(s,1H),6.62(s,1H),5.91(s,1H),5.79(s,1H),3.60(s,3H),3.59-3.52(m,4H),3.01(t,2H),2.68-2.60(m,1H),2.52(s,3H),2.28-2.20(m,1H),1.22(s,9H).
实施例144
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(2-氧代吲哚啉-6-基)丁酰胺144
Figure PCTCN2017099579-appb-000196
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,原料化合物8j替换为6-氨基吲哚啉-2-酮(采用专利申请“WO2009079767”公开的方法制备而得),制得标题化合物144(25mg)。
MS m/z(ESI):566.1[M+1]
1H NMR(400MHz,CD3OD)δ8.18(s,1H),8.13(s,1H),7.86(d,1H),7.60-7.57(m,2H),7.39-7.37(m,3H),6.52(s,1H),5.90-5.86(m,1H),4.21(s,3H),3.65(s,3H),3.58-3.46(m,2H),2.98(s,2H),2.47-2.32(m,2H),1.20(s,9H).
实施例145
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-(叔丁氧基)-N-(2-氧代吲哚啉-5-基)丁酰胺145
Figure PCTCN2017099579-appb-000197
采用实施例108中化合物108e的合成路线,将原料化合物8f替换为原料化合物7b,原料化合物8j替换为5-氨基吲哚啉-2-酮(采用公知的方法“Bioorganic and  Medicinal Chemistry,2013,21(7),1724-1734”制备而得),制得标题化合物145(25mg)。
MS m/z(ESI):566.1[M+1]
1H NMR(400MHz,CD3OD)δ7.89(d,1H),7.59(d,1H),7.56(s,1H),7.41-7.39(m,2H),7.36(s,1H),6.87(d,1H),6.51(s,1H),5.87-5.82(m,1H),3.65(s,3H),3.55(s,2H),3.54-3.43(m,2H),2.55(s,3H),2.53-2.36(m,2H),1.19(s,9H).
实施例146
4-(2-(4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酰氨基)苯甲酸146
Figure PCTCN2017099579-appb-000198
采用实施例30的合成路线,将原料化合物4a替换为化合物1b,制得标题化合物146(35mg)。
MS m/z(ESI):531.4[M+1]
1H NMR(400MHz,CD3OD)δ8.03-7.99(m,2H),7.78-7.68(m,4H),7.46(d,1H),6.51(s,1H),5.89-5.77(m,1H),3.69(d,3H),3.58-3.53(m,1H),3.47-3.40(m,1H),3.36(s,3H),2.59-2.51(m,4H),2.44-2.38(m,1H).
实施例147
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酰氨基)苯甲酸147
Figure PCTCN2017099579-appb-000199
采用实施例1的合成路线,将原料化合物1h替换为化合物4c制得标题化合物147(20mg)。
MS m/z(ESI):513.4[M+1]
1H NMR(400MHz,DMSO-d6)δ10.73(s,1H),7.89-7.87(m,3H),7.76(s,1H),7.74(s,1H),7.64-7.61(dd,1H),7.46-7.45(d,1H),7.29(s,1H),6.40(s,1H),5.72-5.70(m,1H),3.53(s,3H),3.27-3.25(m,2H),3.22(s,3H),2.50(s,3H),2.33-2.30(m,2H).
实施例148
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酰氨基)-2-氟苯甲酸148
Figure PCTCN2017099579-appb-000200
采用实施例1的合成路线,将原料化合物1h替换为4-氨基-2-氟苯甲酸甲酯(采用专利申请“WO2013068467”公开的方法制备而得)制得标题化合物148(15mg)。
MS m/z(ESI):531.5[M+1]
1H NMR(400MHz,DMSO-d6)δ10.79(s,1H),7.89-7.87(d,1H),7.76-7.72(m,1H),7.64-7.62(dd,1H),7.61-7.58(d,1H),7.47-7.45(d,1H),7.40-7.38(d,1H),7.28(s,1H),6.40(s,1H),5.72-5.70(m,1H),3.53(s,3H),3.27-3.25(m,2H),3.22(s,3H),2.50(s,3H),2.33-2.30(m,2H).
实施例149
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酰氨基)-2-甲氧基苯甲酸149
Figure PCTCN2017099579-appb-000201
采用实施例1的合成路线,将原料化合物1h替换为4-氨基-2-甲氧基苯甲酸甲酯(采用专利申请“WO 2016053794”公开的方法制备而得)制得标题化合物149(42mg)。
MS m/z(ESI):543.5[M+1]
1H NMR(400MHz,DMSO-d6)δ10.67(s,1H),7.89(d,1H),7.63(d,2H),7.53(s,1H),7.46(s,1H),7.28(s,1H),7.23(d,1H),6.41(s,1H),5.74-5.70(m,1H),3.77(s,3H),3.54(s,3H),3.30-3.27(m,2H),3.22(s,3H),2.53(s,3H),2.38-2.36(m,2H)
实施例150
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(2,2-二甲基-3-氧代-3,4-二氢-2H-苯并[b][1,4]噁嗪-6-基)-4-甲氧基丁酰胺150
Figure PCTCN2017099579-appb-000202
采用实施例117的合成路线,将原料化合物117c替换为6-氨基-2,2-二甲基-2H-苯并[b][1,4]噁嗪-3(4H)-酮(采用专利申请“JP 2008013527”公开的方法制备而得)制得标题化合物150(40mg)。
MS m/z(ESI):566.4[M+1]
1H NMR(400MHz,DMSO-d6)δ10.64(s,1H),10.42(s,1H),7.89-7.87(d,1H),7.64-7.62(dd,1H),7.47-7.46(d,1H),7.39-7.38(d,1H),7.29(s,1H),7.12-7.09(dd,1H),6.88-6.86(d,1H),6.40(s,1H),5.72-5.70(m,1H),3.53(s,3H),3.27-3.25(m,2H),3.22(s,3H),2.50(s,3H),2.33-2.30(m,2H),1.37(s,6H)
实施例151
1-((乙氧羰基)氧基)乙基5-((R)-2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酰氨基)-1H-吲哚-2-羧酸酯151
1-((乙氧羰基)氧基)乙基5-((S)-2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酰基氨基)-1H-吲哚-2-羧酸酯152
Figure PCTCN2017099579-appb-000203
采用实施例14,15的合成路线,将原料化合物14a替换为5-硝基-1H-吲哚-2-羧酸(采用专利申请“US20160282369”公开的方法制备而得),原料化合物14b替换为1-氯乙基乙基碳酸酯(采用公知的方法“Tetrahedron Letters,2016,57(14),1619-1621”制备而得),原料化合物4b替换为化合物1g,经手性制备(分离条件:色谱柱Superchiral S-AD(Chiralway),2cm I.D.*25cm Length,5um;流动相:二氧化碳:异丙醇=70:30,流速:50g/min),收集其相应组分,减压浓缩,得到标题化合物151(90mg)和152(96mg)。
化合物151:
MS m/z(ESI):668.6[M+1]
手性HPLC分析:保留时间25.596分钟,手性纯度99.3%(色谱柱: Amylose-1(AD)4.6*150mm 5um(带保护柱);流速:1mL/min;流动相:乙醇/正己烷=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ11.98(s,1H),10.41(s,1H),10.22(s,1H),8.09(s,1H),7.90-7.88(d,1H),7.64-7.61(dd,1H),7.48-7.46(d,1H),7.44-7.40(m,1H),7.34(s,1H),7.21-7.20(d,1H),6.93-6.89(m,1H),6.40(s,1H),5.80-5.70(m,1H),4.19-4.14(m,2H),3.55(s,3H),3.27-3.25(m,2H),3.23(s,3H),2.50(s,3H),2.40-2.30(m,2H),1.60(d,3H),1.24-1.20(m,3H)
化合物152:
MS m/z(ESI):668.5[M+1]
手性HPLC分析:保留时间11.905分钟,手性纯度100%(色谱柱:
Figure PCTCN2017099579-appb-000205
 Amylose-1(AD)4.6*150mm 5um(带保护柱);流速:1mL/min;流动相:乙醇/正己烷=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ11.98(s,1H),10.41(s,1H),10.22(s,1H),8.09(s, 1H),7.90-7.88(d,1H),7.64-7.61(dd,1H),7.48-7.46(d,1H),7.44-7.40(m,1H),7.34(s,1H),7.21-7.20(d,1H),6.93-6.89(m,1H),6.40(s,1H),5.80-5.70(m,1H),4.19-4.14(m,2H),3.55(s,3H),3.27-3.25(m,2H),3.23(s,3H),2.50(s,3H),2.40-2.30(m,2H),1.60(d,3H),1.24-1.20(m,3H)
实施例153,154
(5-甲基-2-氧代-1,3-二氧杂环戊烯-4-基)甲基(R)-5-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酰氨基)-1H-吲哚-2-羧酸酯153
(5-甲基-2-氧代-1,3-二氧杂环戊烯-4-基)甲基(S)-5-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲氧基丁酰基氨基)-1H-吲哚-2-羧酸酯154
Figure PCTCN2017099579-appb-000206
采用实施例14,15的合成路线,将原料化合物14a替换为5-((叔丁氧羰基)氨基)-1H-吲哚-2-羧酸(采用公知的方法“Journal of the American Chemical Society,2007,129(17),5384-5390”制备而得),原料化合物14b替换为4-(氯甲基)-5-甲基-1,3-二氧杂环戊烯-2-酮(采用专利申请“CN103450146”公开的方法制备而得),原料化合物4b替换为化合物1g,经手性制备(分离条件:手性制备柱CHIRAL PAK IE,20*250mm,5um;流动相:甲醇:乙醇=50:50,流速:10mL/min),收集其相应组分,减压浓缩,得到标题化合物153(60mg)和154(25mg)。
化合物153:
MS m/z(ESI):664.5[M+1]
手性HPLC分析:保留时间7.129分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流速:1mL/min;流动相:甲醇/乙醇=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ11.95(s,1H),10.39(s,1H),8.08(s,1H),7.89-7.87(d,1H),7.64-7.61(dd,1H),7.47-7.45(d,1H),7.44-7.41(dd,1H),7.40-7.38(d,1H),7.34(s,1H),7.19-7.18(d,1H),6.40(s,1H),5.80-5.70(m,1H),5.23(s,2H),3.53(s,3H),3.27-3.25(m,2H),3.22(s,3H),2.50(s,3H),2.40-2.30(m,2H),2.23(s,3H)
化合物154:
MS m/z(ESI):664.5[M+1]
手性HPLC分析:保留时间8.597分钟,(色谱柱:CHIRAL PAK IE,4.6*150mm,5um;流速:1mL/min;流动相:甲醇/乙醇=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ11.95(s,1H),10.39(s,1H),8.08(s,1H),7.89-7.87(d,1H),7.64-7.61(dd,1H),7.47-7.457(d,1H),7.44-7.41(dd,1H),7.40-7.38(d,1H),7.34(s,1H),7.19-7.18(d,1H),6.40(s,1H),5.80-5.70(m,1H),5.23(s,2H),3.53(s,3H),3.27-3.25(m,2H),3.22(s,3H),2.50(s,3H),2.40-2.30(m,2H),2.23(s,3H)
实施例155
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)己酰氨基)苯甲酸155
Figure PCTCN2017099579-appb-000207
采用实施例7的合成路线,将原料化合物7c替换为三氟甲烷磺酸丁酯(采用公知的方法“Perkin 1,2000,(4),571-574”制备而得)制得标题化合物155(25mg)。
MS m/z(ESI):511.4[M+1]
1H NMR(400MHz,CD3OD)δ8.01(d,2H),7.88(d,1H),7.75(d,2H),7.58(dd,1H),7.41(d,1H),7.38(s,1H),6.53(s,1H),5.80-5.76(m,1H),3.65(s,3H),2.55(s,3H),2.28-2.14(m,2H),1.50-1.37(m,4H),0.98(t,3H).
实施例156
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)丙酰氨基)苯甲酸156
Figure PCTCN2017099579-appb-000208
采用实施例7的合成路线,将原料化合物7c替换为碘甲烷制得标题化合物156(30mg)。
MS m/z(ESI):469.1[M+1]
1H NMR(400MHz,CDCl3)δ10.03(s,1H),8.13(d,2H),7.90(d,2H),7.73(d,1H),7.53-7.50(m,1H),7.34(s,1H),7.18(s,1H),6.71(s,1H),6.11-6.06(m,1H),3.65(s,3H),2.55(s,3H),1.77-1.75(m,3H).
实施例157
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)丁酰氨基)苯甲酸157
Figure PCTCN2017099579-appb-000209
采用实施例7的合成路线,将原料化合物7c替换为碘乙烷制得标题化合物157(6mg)。
MS m/z(ESI):483.2[M+1]
1H NMR(400MHz,CDCl3)δ10.04(s,1H),8.15(d,2H),7.93(d,2H),7.73(d,1H),7.53-7.50(m,1H),7.34-7.29(m,2H),6.72(s,1H),5.93-5.89(m,1H),3.65(s,3H),2.54(s,3H),2.39-2.30(m,1H),2.08-2.03(m,1H),1.12-1.08(m,3H).
实施例158
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-甲基环丙基)丙酰 氨基)苯甲酸158
Figure PCTCN2017099579-appb-000210
第一步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-甲基戊-4-烯酸叔丁酯158a
将化合物7b(250mg,638.01umol)溶于10mL四氢呋喃中,冷却至-78℃加入3-溴-2-甲基丙烯(172.26mg,1.28mmol)和双三甲基硅基胺基锂(427.02mg,2.55mmol)的四氢呋喃溶液,-78℃搅拌反应1小时。反应液中加入饱和氯化铵溶液淬灭反应,用乙酸乙酯萃取,有机相减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化,得到标题化合物158a(250mg,产率:87.87%)。
第二步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(1-甲基环丙基)丙酸叔丁酯158b
冰浴冷却下,将二乙基锌(2.69mmol,2.69mL)溶于15mL二氯甲烷中,缓慢滴加入三氟乙酸(306.82mg,2.69mmol)的二氯甲烷溶液,再滴加入二碘甲烷(720.72mg,2.69mmol)的二氯甲烷溶液,最后滴加入预制的化合物158a(60mg,134.55μmol)的二氯甲烷溶液,室温搅拌反应24小时。冰浴冷却反应液,滴加入10mL盐酸,用乙酸乙酯萃取(50mL×3),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品标题化合物158b(50mg),产品不经纯化直接进行下一步反应。
采用实施例7的合成路线,将原料化合物7d替换为粗品化合物158b制得标题化合物158(10mg)。
MS m/z(ESI):523.4[M+1]
1H NMR(400MHz,CD3OD)δ10.52(s,1H),8.01-8.00(m,1H),7.98-7.97(m,1H),7.87-7.85(d,1H),7.77-7.76(d,1H),7.75-7.74(d,1H),7.59-7.56(dd,1H),7.40(s,1H),7.39-7.38(d,1H),6.51(s,1H),6.01-5.95(m,1H),3.60(s,3H),2.50(s,3H),2.35-2.25(m,1H),2.00-1.90(m,1H),1.18(s,3H),0.42-0.38(m,1H),0.35-0.31(m,1H),0.30-0.25(m,2H).
实施例159
4-(2-(4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-环丁基丙酰基氨基)苯甲酸159
Figure PCTCN2017099579-appb-000211
将3-环丁基丙酸159a(500mg,3.90mmol,采用公知的方法“Organic Process Research & Development,2008,12(2),183-191”制备而得)溶于5mL四氯化碳中,加入三溴化磷(1.06g,3.90mmol)和溴(1.56g,9.75mmol),升温至85℃搅拌反应12小时。反应液冷却至室温,用饱和亚硫酸氢钠溶液洗涤,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化,得到标题化合物159b(300mg,产率:37.14%)。
采用实施例30的合成路线,将原料化合物4a替换为化合物159b制得标题化合物159(60mg)。
MS m/z(ESI):541.1[M+1]
1H NMR(400MHz,DMSO-d6)δ12.74(br,1H),10.82(s,1H),7.93-7.90(m,2H),7.85-7.75(m,4H),7.42(d,1H),6.41(d,1H),5.72-5.66(m,1H),3.65(d,3H),2.49(s,3H),2.27-2.18(m,3H),2.01-1.91(m,2H),1.79-1.66(m,4H).
实施例160,161
(R)-4-(2-(4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-环丁基丙酰氨基)苯甲酸160
(S)-4-(2-(4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-环丁基丙酰氨基)苯甲酸161
Figure PCTCN2017099579-appb-000212
将化合物159(50mg,92.43umol)进行手性制备(分离条件:色谱柱:Superchiral S-AD(Chiralway),0.46cm I.D.*15cm Length,5um;流动相:二氧化碳:乙醇:二乙胺=60:40:0.05,流速:50g/min),收集其相应组分,减压浓缩,得到标题化合物160(20mg)和化合物161(20mg)。
化合物160:
MS m/z(ESI):541.2[M+1]
手性HPLC分析:保留时间6.264分钟,(色谱柱:Lux Amylose-1(AD)4.6*150mm  5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=70/30(v/v))。
1H NMR(400MHz,CD3OD)δ7.97(d,2H),7.73-7.63(m,4H),7.46(d,1H),6.47(s,1H),5.76-5.70(m,1H),3.65(d,3H),2.34(d,3H),2.29-2.21(m,3H),2.06-1.43(m,6H).
化合物161:
MS m/z(ESI):541.4[M+1]
手性HPLC分析:保留时间9.045分钟,(色谱柱:Lux Amylose-1(AD)4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=70/30(v/v))。
1H NMR(400MHz,CD3OD)δ7.97(d,2H),7.73-7.63(m,4H),7.46(d,1H),6.47(s,1H),5.76-5.70(m,1H),3.65(d,3H),2.34(d,3H),2.29-2.21(m,3H),2.06-1.43(m,6H).
实施例162
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-环丁氧基丁酰氨基)苯甲酸162
Figure PCTCN2017099579-appb-000213
将2-(环丁氧基)乙醇162a(224mg,1.93mmol,采用专利申请“WO 2015120786”公开的方法制备而得)溶于10mL二氯甲烷中,冰浴下加入2,6-二甲基吡啶(206.64mg,1.93mmol),再滴加三氟甲磺酸酐(598.49mg,2.12mmol),搅拌反应2小时。反应液中加15mL水,分液,水相用15mL二氯甲烷萃取,合并有机相,用饱和氯化钠溶液(15mL×2)洗涤,用无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品标题化合物162b(420mg),产品不经纯化直接进行下一步反应。
采用实施例7的合成路线,将原料化合物7c替换为粗品化合物162b制得标题化合物162(35mg)。
MS m/z(ESI):553.4[M+1]
1H NMR(400MHz,DMSO-d6)δ10.75(s,1H),7.91-7.87(m,3H),7.76(d,2H),7.63(dd,1H),7.44(d,1H),7.29(s,1H),6.42(s,1H),5.79-5.75(m,1H),3.87-3.80(m,1H),3.54(s,3H),3.33-3.31(m,1H),3.24-3.22(m,1H),2.49(s,3H),2.41-2.28(m,2H),2.10-2.01(m,2H),1.82-1.68(m,2H),1.60-1.52(m,1H),1.44-1.34(m,1H).
实施例163,164
(R)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-环丁氧基丁酰氨基)苯甲酸163
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-4-环丁氧基丁酰氨基)苯甲酸164
Figure PCTCN2017099579-appb-000214
将化合物162(32mg,57.87μmol)进行手性制备(分离条件:手性制备柱CHIRAL PAK IF,20*250mm,5um;流动相:正己烷:乙醇(含0.01%三氟乙酸)=50:50,流速:6.0mL/min),收集其相应组分,减压浓缩,得到标题化合物163(15mg)和化合物164(15mg)。
化合物163:
MS m/z(ESI):553.4[M+1]
手性HPLC分析:保留时间3.577分钟,(色谱柱:CHIRALPAK IF 150*4.6mm,5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ10.77(s,1H),7.91-7.87(m,3H),7.77(d,2H),7.63(dd,1H),7.44(d,1H),7.29(s,1H),6.42(s,1H),5.79-5.75(m,1H),3.87-3.80(m,1H),3.54(s,3H),3.33-3.31(m,1H),3.24-3.16(m,1H),2.49(s,3H),2.38-2.30(m,2H),2.10-2.02(m,2H),1.79-1.71(m,2H),1.59-1.50(m,1H),1.44-1.36(m,1H).
化合物164:
MS m/z(ESI):553.4[M+1]
手性HPLC分析:保留时间8.134分钟,(色谱柱:CHIRALPAK IF 150*4.6mm,5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ12.77(s,1H),10.79(s,1H),7.91-7.87(m,3H),7.77(d,2H),7.63(dd,1H),7.44(d,1H),7.29(s,1H),6.42(s,1H),5.78-5.75(m,1H),3.87-3.80(m,1H),3.54(s,3H),3.33-3.31(m,1H),3.24-3.18(m,1H),2.49(s,3H),2.40-2.28(m,2H),2.10-2.01(m,2H),1.81-1.68(m,2H),1.59-1.52(m,1H),1.46-1.36(m,1H).
实施例165
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-((1R,4R)-4-羟基环己基)丙酰氨基)苯甲酸165
Figure PCTCN2017099579-appb-000215
Figure PCTCN2017099579-appb-000216
第一步
3-((1R,4R)-4-((叔丁基二甲基硅基)氧基)环己基)丙烯酸乙酯165b
将(1R,4R)-4-((叔丁基二甲基硅基)氧基)环己烷-1-甲醛165a(3.2g,13.2mmol,采用公知的方法“Bioorganic & Medicinal Chemistry Letters,2016,26(14),3213-3215”制备而得)溶于50mL甲苯中,加入乙氧甲酰基亚甲基三苯基膦(5.518g,15.84mmol),升温至100℃搅拌反应16小时。反应液冷却至室温,减压浓缩,所得残余物中加入50mL饱和碳酸氢钠溶液,用乙酸乙酯(100mL×2)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化,得到标题化合物165b(3.2g,产率:73.69%)。
第二步
3-((1R,4R)-4-((叔丁基二甲基硅基)氧基)环己基)丙酸乙酯165c
将化合物165b(1.5g,4.8mmol)溶于30mL乙酸乙酯中,加入钯碳(51.08mg,0.48mmol),氢气置换三次,室温搅拌反应3小时。反应液过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化,得到标题化合物165c(1.509g,产率:94.96%)。
第三步
2-溴-3-((1R,4R)-4-((叔丁基二甲基硅基)氧基)环己基)丙酸乙酯165d
将化合物165c(1.11g,3.53mmol)溶于40mL四氢呋喃中,冷却至-78℃,分批加入双三甲基硅基胺基锂(620.03mg,3.71mmol),搅拌反应60分钟后,加入三甲基氯硅烷(383.39mg,3.53mmol)和N-溴代丁二酰亚胺(628.08mg,3.53mmol),搅拌反应2小时后,升至室温继续搅拌反应1小时。反应液中加入50mL饱和氯化钠溶液,用乙酸乙酯(50mL×3)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化,得到标题化合物165d(260mg,产率:17.79%)。
第四步
2-溴-3-((1R,4R)-4-((叔丁基二甲基硅基)氧基)环己基)丙酸165e
将化合物165d(260mg,0.66mmol)溶于4mL四氢呋喃中,加入一水合氢氧化锂(83.19mg,1.98mmol),搅拌反应2小时。反应液中滴加10%柠檬酸溶液调pH至3~4,用乙酸乙酯(25mL×2)萃取,合并有机相,用50mL饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品标题化合物165e(240mg),产品不经纯化直接进行下一步反应。
第五步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-((1R,4R)-4-((叔丁基二甲基硅基)氧基)环己基)丙酸165f
将叔丁醇镁(171.41mg,1.01mmol)溶于30mL四氢呋喃中,加入粗品化合物165e(239.46mg,0.66mmol)、叔丁醇钾(59.4mg,0.53mmol)和化合物1f(140mg,0.5mmol),60℃搅拌反应16小时。反应液冷却至室温,滴加1M盐酸至pH为3~4,用乙酸乙酯萃取(100mL×2)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化,得到标题化合物165e(283mg,产率:24.96%)。
第六步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-((1R,4R)-4-((叔丁基二甲基硅基)氧基)环己基)丙酰氨基)苯甲酸甲酯165g
将化合物165e(300mg,0.53mmol)溶于20mL乙酸乙酯中,加入化合物4c(80.67mg,0.53mmol)和N,N-二异丙基乙胺(0.28mL,1.6mmol),再加入2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,679.18mg,1.07mmol),升温至60℃,搅拌反应2小时。反应液冷却至室温,减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物165g(60mg,产率:15.36%)。
第七步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-((1R,4R)-4-羟基环己基)丙酰氨基)苯甲酸甲酯165h
将化合物165g(60mg,0.09mmol)溶于10mL四氢呋喃中,加入四丁基氟化铵(180.49mg,0.69mmol),升温至66℃搅拌反应8小时。反应液冷却至室温,加入20mL水,用乙酸乙酯萃取(25mL×4),合并有机相,依次用水(25mL×4),饱和氯化钠溶液(25mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题化合物165h(26mg,产率:49.78%)。
第八步
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-((1R,4R)-4-羟基环己基)丙酰氨基)苯甲酸165
将化合物165h(25mg,0.04mmol)溶于3.63mL四氢呋喃和甲醇混合溶剂 (V/V=10:1)中,加入0.33mL的1M氢氧化锂溶液,搅拌反应16小时。反应液中滴加10%盐酸至pH为3~4,用乙酸乙酯萃取(50mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相制备(Waters 2767-SQ detecor2,洗脱体系:乙腈,水)纯化所得残余物,制得到标题化合物165(8mg,产率:32.46%)。
MS m/z(ESI):567.5[M+1]
1H NMR(400MHz,DMSO-d6)δ10.78(s,1H),7.91-7.87(m,3H),7.75(s,1H),7.73(s,1H),7.64-7.62(dd,1H),7.48-7.47(d,1H),7.28(s,1H),6.42(s,1H),5.95-5.83(m,1H),4.46(m,1H),3.54(s,3H),2.48(s,3H),2.09-2.07(m,1H),2.01-1.99(m,1H),1.90-1.84(m,1H),1.82-1.72(m,4H),1.08-0.96(m,4H).
实施例166
4-(2-(4-(5-氯-2-丙酰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(四氢-2H-吡喃-2-基)丙酰氨基)苯甲酸166
Figure PCTCN2017099579-appb-000217
采用实施例8的合成路线,将原料化合物8g替换为三氟甲磺酸(四氢-2H-吡喃-2-基)甲酯(采用专利申请“WO2016046159”公开的方法制备而得)制得标题化合物166(30mg)。
MS m/z(ESI):567.4[M+1]
1H NMR(400MHz,CD3OD)δ8.01-7.97(m,2H),7.86-7.82(m,1H),7.75-7.70(m,2H),7.58-7.55(m,1H),7.41-7.38(dd,1H),7.36-7.30(m,1H),6.49-6.48(d,1H),5.91-5.60(m,1H),4.00-3.94(m,1H),3.62(s,3H),3.44-3.39(m,1H),3.25-3.21(m,1H),3.00-2.95(m,2H),2.50-2.27(m,2H),1.85-1.74(m,1H),1.71-1.68(m,1H),1.66-1.45(m,3H),1.42-1.38(m,1H),1.13-1.08(m,3H)
实施例167
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-环丙基丙酰氨基)苯甲酸167
Figure PCTCN2017099579-appb-000218
采用实施例7的合成路线,将原料化合物7c替换为溴甲基环丙烷(采用专利申请“CN106242941”公开的方法制备而得)制得标题化合物167(18mg)。
MS m/z(ESI):509.4[M+1]
1H NMR(400MHz,DMSO-d6)δ10.79(s,1H),7.93-7.88(m,3H),7.76(s,1H),7.74(s,1H),7.64-7.62(d,1H),7.46(s,1H),7.33(s,1H),6.42(s,1H),5.81-5.77(m,1H),3.55 (s,3H),2.50(s,3H),2.22-2.14(m,1H),1.91-1.83(m,1H),0.68-0.64(m,1H),0.49-0.42(m,1H),0.40-0.33(m,1H),0.30-0.20(m,2H).
实施例168
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-环丙基丙酰氨基)苯甲酸168
Figure PCTCN2017099579-appb-000219
将化合物167(180mg,353.67umol)进行手性制备(分离条件:手性制备柱:Lux Amylose-1(AD)21.2*250mm 5um;流动相:正己烷:乙醇(含0.01%三氟乙酸)=30:70,流速:10.0mL/min),收集其相应组分,减压浓缩,得到标题化合物168(40mg)。
MS m/z(ESI):509.4[M+1]
手性HPLC分析:保留时间11.482分钟,(色谱柱:Lux Amylose-1(AD)4.6*150mm5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=70/30(v/v))。
1H NMR(400MHz,CD3OD)δ8.01-8.00(m,1H),7.98-7.97(m,1H),7.90-7.85(d,1H),7.75-7.74(m,1H),7.73-7.71(m,1H),7.59-7.56(dd,1H),7.40(s,1H),7.39-7.38(d,1H),6.51(s,1H),5.84-5.80(m,1H),3.60(s,3H),2.50(s,3H),2.20-2.00(m,2H),0.85-0.75(m,1H),0.55-0.45(m,2H),0.35-0.25(m,2H)
实施例169
4-(2-(4-(6-乙酰基-3-氯-2-氟苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-环丙基丙酰基氨基)苯甲酸169
Figure PCTCN2017099579-appb-000220
采用实施例30的合成路线,将原料化合物4a替换为溴甲基环丙烷制得标题化合物169(20mg)。
MS m/z(ESI):527.2[M+1]
1H NMR(400MHz,CD3OD)δ8.01-8.00(d,1H),7.99-7.98(d,1H),7.76-7.67(m,4H),7.49-7.46(d,1H),6.48(s,1H),5.90-5.80(m,1H),3.66(s,3H),2.53-2.48(m,3H),2.15-2.05(m,2H),0.80-0.75(m,1H),0.55-0.45(m,2H),0.25-0.20(m,2H)
实施例170
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-环丁基丙酰氨基)苯甲酸170
Figure PCTCN2017099579-appb-000221
采用实施例4的合成路线,将原料化合物4a替换为化合物159b制得标题化合物170(42mg)。
MS m/z(ESI):523.2[M+1]
1H NMR(400MHz,CD3OD)δ8.02-8.00(m,2H),7.89(d,1H),7.77-7.74(m,2H),7.59(dd,1H),7.41(d,1H),7.38(s,1H),6.51(s,1H),5.76-5.72(m,1H),3.66(s,3H),2.56(s,3H),2.36-2.23(m,3H),2.20-2.10(m,2H),1.96-1.77(m,4H).
实施例171,172
(S)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-环丁基丙酰氨基)苯甲酸171
(R)-4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-环丁基丙酰氨基)苯甲酸172
Figure PCTCN2017099579-appb-000222
将化合物170(38mg,0.07mmol)进行手性制备(分离条件:手性制备柱CHIRAL PAK IE,20*250mm,5um;流动相:乙醇(含0.01%三氟乙酸)=100,流速:6.0mL/min),收集其相应组分,减压浓缩,得到标题化合物171(18mg)和化合物172(18mg)。
化合物171:
MS m/z(ESI):523.2[M+1]
手性HPLC分析:保留时间9.644分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=40/60(v/v))。
1H NMR(400MHz,CD3OD)δ8.02-8.00(m,2H),7.89(d,1H),7.77-7.74(m,2H),7.59(dd,1H),7.41(d,1H),7.38(s,1H),6.51(s,1H),5.76-5.72(m,1H),3.66(s,3H),2.56(s,3H),2.36-2.23(m,3H),2.20-2.10(m,2H),1.96-1.77(m,4H).
化合物172:
MS m/z(ESI):523.2[M+1]
手性HPLC分析:保留时间3.831分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:正己烷/乙醇(含0.1%三氟乙酸)=40/60(v/v))。
1H NMR(400MHz,CD3OD)δ8.02-8.00(m,2H),7.89(d,1H),7.77-7.74(m,2H),7.59(dd,1H),7.41(d,1H),7.38(s,1H),6.51(s,1H),5.76-5.72(m,1H),3.66(s,3H),2.56(s,3H),2.36-2.23(m,3H),2.20-2.10(m,2H),1.96-1.77(m,4H).
实施例173
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(3,3-二甲基环丁基)丙酰氨基)苯甲酸173
Figure PCTCN2017099579-appb-000223
采用实施例165的合成路线,将原料化合物165a替换为3,3-二甲基环丁烷-1-甲醛(采用专利申请“WO2015129926”公开的方法制备而得)制得标题化合物173(25mg)。
MS m/z(ESI):551.2[M+1]
1H NMR(400MHz,CD3OD)δ8.00(s,1H),7.98(s,1H),7.88-7.86(d,1H),7.74(s,1H),7.72(s,1H),7.59-7.26(dd,1H),7.40-7.39(d,1H),7.37(s,1H),6.49(s,1H),5.71-5.69(m,1H),3.64(s,3H),2.53(s,3H),2.30-2.25(m,3H),1.95-1.85(m,2H),1.65-1.60(m,1H),1.55-1.50(m,1H),1.26(s,3H),1.06(s,3H).
实施例174
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(四氢呋喃-2-基)丙酰氨基)苯甲酸174
Figure PCTCN2017099579-appb-000224
采用实施例7的合成路线,将原料化合物7c替换为(四氢呋喃-2-基)甲基三氟甲磺酸酯(采用专利申请“WO2003095438”公开的方法制备而得)制得标题化合物174(15mg)。
MS m/z(ESI):539.1[M+1]
1H NMR(400MHz,CD3OD)δ8.01-8.00(m,1H),7.98-7.97(m,1H),7.90-7.85(m,1H),7.75-7.71(m,2H),7.59-7.56(dt,1H),7.40-7.38(m,1H),7.37-7.34(m,1H),6.52-6.48(m,1H),5.70-5.60(m,1H),3.95-3.85(m,2H),3.75-3.70(m,1H),3.64(s,3H),2.54(s,2H),2.51(s,1H),2.50-2.22(m,2H),2.16-2.10(m,1H),2.00-1.95(m,2H),1.65-1.60(m,1H).
实施例175
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(4-甲氧基环己基)丙酰氨基)苯甲酸175
Figure PCTCN2017099579-appb-000225
采用实施例165的合成路线,将原料化合物165a替换为4-甲氧基环己烷-1-甲醛(采用专利申请“WO2016044626”公开的方法制备而得)制得标题化合物175(8mg)。
MS m/z(ESI):581.2[M+1]
1H NMR(400MHz,CD3OD)δ8.00-7.37(m,8H),6.78-6.53(m,1H),5.95(s,1H),3.89-3.82(m,1H),3.66-3.62(m,3H),3.48-3.16(m,3H),2.68-2.54(m,3H),2.09-1.90(m,5H),1.66-1.14(m,6H).
实施例176
4-(2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(四氢-2H-吡喃-2-基)丙酰氨基)苯甲酸176
Figure PCTCN2017099579-appb-000226
采用实施例7的合成路线,将原料化合物7c替换为(四氢-2H-吡喃-2-基)甲基三氟甲磺酸酯制得标题化合物176(15mg)。
MS m/z(ESI):553.4[M+1]
1H NMR(400MHz,DMSO-d6)δ10.66(s,1H),7.92-7.87(m,3H),7.79-7.75(m,2H),7.64-7.61(dd,1H),7.47-7.45(dd,1H),7.31-7.27(d,1H),6.39(s,1H),5.72-5.65(m,1H),3.90-3.85(m,1H),3.54(s,3H),3.31-3.08(m,2H),2.50(s,3H),2.25-2.35(m,1H),2.22-2.15(m,1H),2.10-2.00(m,1H),1.80-1.70(m,1H),1.65-1.55(m,1H),1.50-1.35(m,3H).
实施例177,178
4-[[(2S)-4-叔丁氧基-2-[4-(5-氯-2-丙酰基-苯基)-2-氧代-5-(三氘代甲氧基)-1-吡啶基]丁酰基]氨基]苯甲酸177
4-[[(2R)-4-叔丁氧基-2-[4-(5-氯-2-丙酰基-苯基)-2-氧代-5-(三氘代甲氧基)-1-吡啶基]丁酰基]氨基]苯甲酸178
Figure PCTCN2017099579-appb-000227
Figure PCTCN2017099579-appb-000228
第一步
2-甲氧基-5-(三氘代甲氧基)吡啶177b
将6-甲氧基吡啶-3-醇177a(4.0g,31.97mmol,采用公知的方法“Medicinal Chemistry Research,2013,22(4),1825-1836”制备而得)溶于50mL N,N-二甲基甲酰胺中,加入碳酸钾(13.25g,95.90mmol),冰浴冷却,滴加入三氘代碘甲烷(6.95g,47.95mmol),滴加过程中控制反应液内温不超过20℃,1小时内滴加完毕,升温至室温搅拌反应3小时。反应液中加入100mL水,用乙酸乙酯萃取(300mL×1),分液,依次用水洗涤(100mL×5)、饱和氯化钠溶液洗涤(100mL),用无水硫酸钠干燥,过滤,滤液减压浓缩,得粗品标题化合物177b(4.4g),产物不经纯化直接用于下一步反应。
第二步
[2-甲氧基-5-(三氘代甲氧基)-4-吡啶基]硼酸177c
将粗品化合物177b(4.40g,30.95mmol)溶于50mL四氢呋喃中,冷却至-78℃,滴加入2M二异丙基氨基锂的四氢呋喃/正庚烷/乙苯溶液(30.95mL,61.90mmol),滴加过程中控制反应液内温不超过-65℃,滴加完毕,-78℃搅拌反应0.5小时,缓慢滴加入硼酸三异丙酯(6.63g,61.90mmol),滴加过程中控制反应液内温不超过-65℃,滴加完毕,-78℃搅拌反应2小时。反应液中加入80mL水淬灭反应,加入80mL乙酸乙酯,分液,向水相中滴加6M盐酸至pH为3-4,析出固体,过滤,收集滤饼,自然干燥,得粗品标题化合物177c(2.5g),产物不经纯化直接用于下一步反应。
第三步
1-[4-氯-2-[2-甲氧基-5-(三氘代甲氧基)-4-吡啶]苯基]丙烷-1-酮177d
将化合物8c(400mg,1.62mmol)溶于13mL 1,4-二氧六环和水的混合溶剂中(V:V=10:3),加入粗品化合物177c(300.57mg,1.62mmol)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(59.94mg,80.80μmol)和碳酸钠(513.91mg,4.85mmol),反应液 升温至85℃,搅拌反应16小时。反应液自然冷却至室温,过滤,滤液中加入30mL水,用乙酸乙酯萃取(80mL×2),合并有机相,有机相用饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物177d(353mg,产率:70.74%)。
第四步
2-[4-(5-氯-2-丙酰基-苯基)-2-氧代-5-(三氘代甲氧基)-1-吡啶基]乙酸叔丁酯177e
将化合物177d(352mg,1.14mmol)和化合物7a(667.08mg,3.42mmol)混合,加热至100℃,搅拌反应2小时。反应液冷却至室温,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物177e(252mg,产率:54.06%)。
第五步
4-叔丁氧基-2-[4-(5-氯-2-丙酰基-苯基)-2-氧代-5-(三氘代甲氧基)-1-吡啶基]丁酸叔丁酯177f
将化合物177e(252mg,616.30μmol)和化合物108b(462.66mg,1.85mmol)溶于15mL四氢呋喃中,反应液冷却至-78℃,滴加二(三甲基硅基)氨基锂溶液(2.47mL,2.47mmol),搅拌反应2小时。-78℃下,向反应液中缓慢加入50mL水淬灭反应,反应液温度自然升至室温,用乙酸乙酯萃取(60mL×2),合并有机相,用饱和氯化钠溶液洗涤(50mL),无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物177f(80mg,产率:25.5%)。
第六步
4-叔丁氧基-2-[4-(5-氯-2-丙酰基-苯基)-2-氧代-5-(三氘代甲氧基)-1-吡啶基]丁酸177g
将化合物177f(80mg,157.16μmol)溶于2mL水、2mL甲醇和10mL四氢呋喃的混合溶剂中,加入一水合氢氧化锂(33mg,785.78μmol),搅拌反应16小时。反应液中滴加1M盐酸至pH为3-4,用乙酸乙酯萃取(50mL×2),合并有机相,用饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品标题化合物177g(60mg),产物不经纯化直接用于下一步反应。
第七步
4-[[4-叔丁氧基-2-[4-(5-氯-2-丙酰基-苯基)-2-氧代-5-(三氘代甲氧基)-1-吡啶基]丁酰基]氨基]苯甲酸177h
将粗品化合物177g(60.09mg,132.66μmol)溶于20mL四氢呋喃中,依次加入N,N-二异丙基乙胺(68.58mg,530.63μmol)和2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,168.74mg,265.31μmol),搅拌反应10分钟,加入化合物8j(19.10mg,139.29μmol),搅拌反应3小时。反应液加入30mL水,乙酸乙酯萃取(50mL×2),合并有机相,用饱和氯化钠溶液洗涤(30mL),无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相制备(Waters 2767,洗脱体系:乙 腈,水,0.05%三氟乙酸)纯化,制得标题化合物177h(30mg,产率:39.53%)。
MS m/z(ESI):572.1[M+1]
第八步
4-[[(2S)-4-叔丁氧基-2-[4-(5-氯-2-丙酰基-苯基)-2-氧代-5-(三氘代甲氧基)-1-吡啶基]丁酰基]氨基]苯甲酸177
4-[[(2R)-4-叔丁氧基-2-[4-(5-氯-2-丙酰基-苯基)-2-氧代-5-(三氘代甲氧基)-1-吡啶基]丁酰基]氨基]苯甲酸178
将化合物177h(30mg,52.44μmol)进行手性制备(分离条件:手性制备柱:Daicel IE 20*250mm 5um;流动相:正己烷/乙醇=50/50(v/v),流速:20mL/min),收集其相应组分,减压浓缩,得到标题化合物177(8mg)和化合物178(8mg)。
化合物177:
MS m/z(ESI):572.1[M+1]
手性HPLC分析:保留时间7.640分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um;流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,CD3OD)δ8.02-8.01(m,2H),7.83-7.76(m,3H),7.57(s,1H),7.36(m,2H),6.52(s,1H),5.88-5.87(m,1H),3.55-3.45(m,2H),3.01-2.97(m,2H),2.48-2.40(m,2H),1.18-1.12(m,12H).
化合物178:
MS m/z(ESI):572.1[M+1]
手性HPLC分析:保留时间4.703分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um;流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,CD3OD)δ8.02-8.01(m,2H),7.83-7.76(m,3H),7.57(s,1H),7.36(m,2H),6.52(s,1H),5.88-5.87(m,1H),3.55-3.45(m,2H),3.01-2.97(m,2H),2.48-2.40(m,2H),1.18-1.12(m,12H)..
实施例179,180
4-[[(2S)-2-[4-(2-乙酰基-5-氯-苯基)-2-氧代-5-(三氘代甲氧基)-1-吡啶基]-3-苯基-丙酰基]氨基]苯甲酸179
4-[[(2R)-2-[4-(2-乙酰基-5-氯-苯基)-2-氧代-5-(三氘代甲氧基)-1-吡啶基]-3-苯基-丙酰基]氨基]苯甲酸180
Figure PCTCN2017099579-appb-000229
Figure PCTCN2017099579-appb-000230
第一至五步
4-[[2-[4-(2-乙酰基-5-氯-苯基)-2-氧代-5-(三氘代甲氧基)-1-吡啶基]-3-苯基-丙酰基]氨基]苯甲酸179e
采用实施例177,178中化合物177h的合成路线,将原料化合物8c替换为化合物1c,化合物108b替换为化合物8g,制得标题化合物179e(200mg)。
第六步
4-[[(2S)-2-[4-(2-乙酰基-5-氯-苯基)-2-氧代-5-(三氘代甲氧基)-1-吡啶基]-3-苯基-丙酰基]氨基]苯甲酸179
4-[[(2R)-2-[4-(2-乙酰基-5-氯-苯基)-2-氧代-5-(三氘代甲氧基)-1-吡啶基]-3-苯基-丙酰基]氨基]苯甲酸180
将化合物179e(200mg,364.96μmol)进行手性制备(分离条件:手性制备柱:Daicel IE 20*250mm 5um;流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v),流速:20mL/min),收集其相应组分,减压浓缩,得到标题化合物179(35mg)和化合物180(35mg)。
化合物179:
MS m/z(ESI):548.0[M+1]
手性HPLC分析:保留时间13.346分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um;流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ10.84(s,1H),7.93(d,2H),7.83(d,1H),7.76(d,2H),7.61(d,1H),7.39(d,2H),7.26-7.30(m,4H),7.18-7.22(m,1H),6.32(s,1H),6.02-6.06(m,1H),3.47-3.50(m,2H),2.38(s,3H).
化合物180:
MS m/z(ESI):548.0[M+1]
手性HPLC分析:保留时间4.909分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um;流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ10.84(s,1H),7.93(d,2H),7.83(d,1H),7.76(d,2H),7.61(d,1H),7.39(d,2H),7.26-7.30(m,4H),7.18-7.22(m,1H),6.32(s,1H),6.02-6.06(m,1H),3.47-3.50(m,2H),2.38(s,3H).
实施例181,182
4-[[(2S)-2-[4-(2-乙酰基-5-氯-苯基)-5-甲氧基-2-氧代-1-吡啶基]-3,3-二氘代-3-(2,3,4,5,6-五氘代苯基)丙酰基]氨基]苯甲酸181
4-[[(2R)-2-[4-(2-乙酰基-5-氯-苯基)-5-甲氧基-2-氧代-1-吡啶基]-3,3-二氘代-3-(2,3,4,5,6-五氘代苯基)丙酰基]氨基]苯甲酸182
Figure PCTCN2017099579-appb-000231
第一步
2-(4-(2-乙酰基-5-氯苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)乙酸乙酯181b
将化合物1e(20.3g,69.59mmol)和2-溴乙酸乙酯181a(34.86g,208.76mmol,采用公知的方法“European Journal of Organic Chemistry,2002,(17),3015-3023”)混合,升温至100℃搅拌反应3小时。反应液冷却至室温,加入50mL异丙醇,搅拌16小时,析出大量固体,过滤,滤饼依次用异丙醇(10mL×2)和正己烷(10mL×2)洗涤,收集滤饼,真空干燥,得粗品标题化合物181b(18.5g),产物不经纯化直接用于下一步反应。
第二步
2-[4-(2-乙酰基-5-氯-苯基)-5-甲氧基-2-氧代-1-吡啶基]-3,3-二氘代-3-(2,3,4,5,6-五氘代苯基)丙酸乙酯181d
将粗品化合物181b(18.5g,50.85mmol)溶于250mL二氯甲烷中,加入1-[溴(二氘代)甲基]-2,3,4,5,6-五氘代-苯181c(22.64g,127.13mmol,采用公知的方法“Angewandte Chemie-International Edition,2015,54(18),5478-5482”制备而得),氩气保护,冷却至-78℃,滴加二(三甲基硅基)氨基锂溶液(254.27mL,254.27mmol),搅拌反应2小时。撤出低温浴,反应液中缓慢滴加100mL饱和氯化铵溶液淬灭反应,温度自然升至室温,加入30mL水,分液,水相用乙酸乙酯萃取(100mL×3),合并有机相,用饱和氯化钠溶液洗涤(100mL),无水硫酸钠干燥。过滤,滤液减压浓缩,得粗品标题化合物181d(30g),产物不经纯化直用于下一步反应。
第三步
2-[4-(2-乙酰基-5-氯-苯基)-5-甲氧基-2-氧代-1-吡啶基]-3,3-二氘代-3-(2,3,4,5,6-五氘代苯基)丙酸181e
将粗品化合物181d(23.44g,50.85mmol)溶于100mL四氢呋喃中,加入1M氢氧化钠溶液(71.19mL,71.19mmol),搅拌反应16小时。反应液减压浓缩除去四氢呋喃,所得残余物用甲基叔丁基醚萃取(100mL×3)除去杂质,水相用浓盐酸调节pH为2~3,用乙酸乙酯萃取(100mL×3),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系C纯化所得残余物,得标题化合物181e(11.7g,产率:53.15%)。
第四步
4-[[2-[4-(2-乙酰基-5-氯-苯基)-5-甲氧基-2-氧代-1-吡啶]-3,3-二氘代-3-(2,3,4,5,6-五氘代苯基)丙酰基]氨基]苯甲酸181f
将化合物181e(11.7g,27.03mmol)溶于60mL四氢呋喃中,冰浴冷却,加入2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,25.8g,40.54mmol),搅拌均匀,滴加入N,N-二异丙基乙胺(10.48g,81.08mmol),冰浴下搅拌反应10分钟,分批加入化合物8j(3.71g,27.03mmol),反应液升温至室温搅拌反应0.5小时。反应液中加入100mL水淬灭反应,搅拌10分钟,分液,水相用乙酸乙酯萃取(50mL×3),合并有机相,减压浓缩除去有机溶剂,加入200mL乙酸乙酯,用饱和氯化钠溶液洗涤(50mL),减压浓缩,所得残余物加入100mL异丙醇,升温至90℃搅拌20分钟,冷却至室温搅拌16小时,过滤,滤饼依次用异丙醇(20mL×2)、甲基叔丁基醚(20mL×2)洗涤,收集滤饼,得粗品标题化合物181f(13.4g),产物不经纯化直接用于下一步反应。
第五步
4-[[(2S)-2-[4-(2-乙酰基-5-氯-苯基)-5-甲氧基-2-氧代-1-吡啶基]-3,3-二氘代-3-(2,3,4,5,6-五氘代苯基)丙酰基]氨基]苯甲酸181
4-[[(2R)-2-[4-(2-乙酰基-5-氯-苯基)-5-甲氧基-2-氧代-1-吡啶基]-3,3-二氘代 -3-(2,3,4,5,6-五氘代苯基)丙酰基]氨基]苯甲酸182
将化合物181f(13.4g,24.27mmol)进行手性制备(分离条件:手性制备柱:CHIRAL PAK AD 5.0*250mm;流动相:二氧化碳/(70%乙醇/30%乙腈/0.1%二乙胺)=60/40(v/v),流速:59mL/min),收集其相应组分,减压浓缩,所得残余物分别用100mL二氯甲烷溶解,冰浴下滴加50mL的0.5M盐酸,室温搅拌反应15分钟,用二氯甲烷萃取(30mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,所得残余物加入100mL甲醇搅拌1小时,过滤,收集滤饼,依次用甲醇(10mL)和甲基叔丁基醚(10mL×2)洗涤,真空干燥,得到标题化合物181(5.5g)和化合物181(4.8g)。
化合物181:
MS m/z(ESI):552.6[M+1]
手性HPLC分析:保留时间12.738分钟,手性纯度99.8%(色谱柱:CHIRAL PAK IE 4.6*150mm 5um;流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ12.77(s,1H),10.83(s,1H),7.92(d,2H),7.82(d,1H),7.76(d,2H),7.60(d,1H),7.42(s,1H),7.37(s,1H),6.30(s,1H),6.01(s,1H),3.54(s,3H),2.37(s,3H).
化合物182:
MS m/z(ESI):552.6[M+1]
手性HPLC分析:保留时间4.902分钟,手性纯度99.1%(色谱柱:CHIRAL PAK IE 4.6*150mm 5um;流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ12.78(s,1H),10.83(s,1H),7.92(d,2H),7.82(d,1H),7.76(d,2H),7.60(d,1H),7.42(s,1H),7.37(s,1H),6.30(s,1H),6.01(s,1H),3.54(s,3H),2.37(s,3H).
实施例183,184
4-[[(2S)-2-[4-[5-氯-2-(2,2,2-三氘代乙酰基)苯基]-5-甲氧基-2-氧代-1-吡啶基]-3-苯基-丙酰基]氨基]苯甲酸183
4-[[(2R)-2-[4-[5-氯-2-(2,2,2-三氘代乙酰基)苯基]-5-甲氧基-2-氧代-1-吡啶基]-3-苯基-丙酰基]氨基]苯甲酸184
Figure PCTCN2017099579-appb-000232
Figure PCTCN2017099579-appb-000233
第一步
1-(2-溴-4-氯-苯)-2,2,2-三氘代-乙酮183b
将化合物8a(3.8g,11.97mmol)溶于50mL四氢呋喃中,冷却至-10℃,慢慢滴加入异丙基氯化镁(1.6g,15.57mmol),预先反应0.5小时。将2,2,2-三氘代乙酰氯183a(1.27g,15.57mmol)、氯化锂(21.70mg,359.23μmol)、氯化亚铜(35.56mg,359.23μmol)和三氯化铝(47.90mg,359.23μmol)加入50mL四氢呋喃中,室温搅拌均匀,将预先反应了0.5小时的反应液滴加到以上混合液中,室温反应0.5小时。反应液中用50mL 3M盐酸洗涤,水相用乙酸乙酯萃取(100mL),有机相用饱和氯化钠溶液洗涤(60mL),无水硫酸钠干燥,过滤,滤液减压浓缩,制得粗品标题化合物183b(2.5g),产物不经纯化直接用于下一步反应。
第二步
1-[4-氯-2-(2,5-二甲氧基-4-吡啶基)苯基]-2,2,2-三氘代-乙酮183c
将化合物183b(400mg,1.69mmol)和化合物1d(309.45mg,1.69mmol)溶于8mL 1,4-二氧六环和1mL重水的混合溶剂中,加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(61.88mg,84.56μmol)和碳酸钠(537.83mg,5.07mmol),反应液加热至85℃,搅拌反应5小时。反应液自然冷却至室温,加入30mL水,用乙酸乙酯萃取(50mL×2),合并有机相,有机相分别用水(40mL)、饱和氯化钠溶液(40mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物183c(400mg,产率:80.24%)。
MS m/z(ESI):295.4[M+1]
第三步
2-[4-[5-氯-2-(2,2,2-三氘代乙酰基)苯基]-5-甲氧基-2-氧代-1-吡啶基]乙酸叔丁酯183d
将化合物183c(400mg,1.36mmol)和化合物7a(794.12mg,4.07mmol)混合, 升温至100℃搅拌反应2小时。反应液冷却至室温,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物183d(480mg,产率:89.57%)。
第四步
2-[4-[5-氯-2-(2,2,2-三氘代乙酰基)苯基]-5-甲氧基-2-氧代-1-吡啶基]-3-苯基-丙酸叔丁酯183e
将化合物183d(480mg,1.22mmol)溶于20mL四氢呋喃中,冷却至-78℃,加入化合物8g(623.73mg,3.65mmol),再滴加入二(三甲基硅基)氨基锂的四氢呋喃溶液(4.86mL,4.86mmol),-78℃搅拌反应1.5小时。反应液中加入4.0mL重水淬灭反应,升温至室温,用乙酸乙酯萃取(50mL×2),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题化合物183e(494mg,产率:83.79%)。
第五步
2-[4-[5-氯-2-(2,2,2-三氘代乙酰基)苯基]-5-甲氧基-2-氧代-1-吡啶基]-3-苯基-丙酸183f
将化合物183e(494mg,1.02mmol)溶于10mL二氯甲烷中,滴加三氟乙酸(2.3g,20.33mmol),搅拌反应2小时。反应液减压浓缩,得粗品标题化合物183f(430mg),产物不经纯化直接用于下一步反应。
第六步
4-[[2-[4-[5-氯-2-(2,2,2-三氘代乙酰基)苯基]-5-甲氧基-2-氧代-1-吡啶]-3-苯基-丙酰基]氨基]苯甲酸183g
将粗品化合物183f(430mg,990.95μmol)溶于10mL四氢呋喃中,冰浴下加入N,N-二异丙基乙胺(512mg,3.96mmol)和2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,1.51g,1.98mmol),冰浴下搅拌反应10分钟,加入化合物8j(136mg,991.72μmol),升温至室温搅拌反应2小时。反应液中加入25mL乙酸乙酯,依次用水(15mL)、饱和氯化钠溶液(15mL)洗涤,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,制得标题化合物183g(512mg,产率:94.28%)。
第七步
4-[[(2S)-2-[4-[5-氯-2-(2,2,2-三氘代乙酰基)苯基]-5-甲氧基-2-氧代-1-吡啶基]-3-苯基-丙酰基]氨基]苯甲酸183
4-[[(2R)-2-[4-[5-氯-2-(2,2,2-三氘代乙酰基)苯基]-5-甲氧基-2-氧代-1-吡啶基]-3-苯基-丙酰基]氨基]苯甲酸184
将化合物183g(512mg,934.31μmol)进行手性制备(分离条件:手性制备柱Daicel IE 20*250mm 5um;流动相:正己烷:乙醇=60:40,流速:20mL/min),收集其相应组分,减压浓缩,得到标题化合物183(200mg)和化合物184(200mg)。
化合物183:
MS m/z(ESI):548.0[M+1]
手性HPLC分析:保留时间12.947分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:乙醇(含0.1%三氟乙酸)/正己烷=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ12.77(br,1H),10.85(s,1H),7.93(d,2H),7.83(d,1H),7.77(d,2H),7.62(d,1H),7.41(d,2H),7.32-7.26(m,4H),7.22-7.18(m,1H),6.32(s,1H),6.06-6.02(m,1H),3.55(s,3H),3.50-3.43(m,2H).
化合物184:
MS m/z(ESI):548.0[M+1]
手性HPLC分析:保留时间4.840分钟,(色谱柱:CHIRAL PAK IE 4.6*150mm 5um(带保护柱);流动相:乙醇(含0.1%三氟乙酸)/正己烷=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ12.77(br,1H),10.85(s,1H),7.93(d,2H),7.83(d,1H),7.77(d,2H),7.62(d,1H),7.41(d,2H),7.32-7.26(m,4H),7.22-7.18(m,1H),6.32(s,1H),6.06-6.02(m,1H),3.55(s,3H),3.50-3.43(m,2H).
对比例1(实施例185)
(S)-4-(2-(4-(5-氯-2-氰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(吡啶-4-基)丙酰氨基)苯甲酸
Figure PCTCN2017099579-appb-000234
第一步
4-氯-2-(2,5-二甲氧基吡啶-4-基)苯甲腈185b
将2-溴-4-氯-苯甲腈185a(5.92g,27.33mmol,采用公知的方法“Angewandte Chemie,International Edition,2017,56(9),2473-2477”)溶于180mL 1,4-二氧六环中,加入化合物1d(5g,27.33mmol)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(2.03g, 2.73mmol)和碳酸钾(11.33g,81.98mmol),氩气保护,反应液升温至110℃,搅拌反应16小时。反应液自然冷却至室温,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物185b(6.5g,产率:86.59%)。
第二步
2-(4-(5-氯-2-氰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)乙酸叔丁酯185c
将化合物185b(5g,18.20mmol)和化合物7a(21.30g,109.21mmol)混合,升温至100℃搅拌反应3小时,降温至90℃搅拌反应4小时。反应液冷却至室温,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物185c(5g,产率:73.29%)。
第三步
2-(4-(5-氯-2-氰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(吡啶-4-基)丙酸叔丁酯185e
将化合物185c(200mg,533.39μmol)和4-(溴甲基)吡啶氢溴酸盐185d(269.92mg,1.07mmol,采用公知的方法“Chemical Communications(Cambridge,United Kingdom),2011,47(5),1482-1484”制备而得)溶于10mL四氢呋喃中,反应液冷却至-78℃,滴加二(三甲基硅基)氨基锂溶液(3.2mL,3.2mmol),搅拌反应2小时。-78℃下,向反应液中缓慢加入10mL水淬灭反应,再加入10mL饱和氯化钠溶液,反应液温度自然升至室温,用乙酸乙酯萃取(20mL×3),合并有机相,用饱和氯化钠溶液洗涤(20mL×2),无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题185e(240mg,产率:96.53%)。
第四步
2-(4-(5-氯-2-氰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(吡啶-4-基)丙酸185f
将化合物185e(240mg,515.10μmol)溶于6mL二氯甲烷中,加入三氟乙酸(1mL,515.1μmol),搅拌反应16小时。反应液减压浓缩,得到粗品标题化合物185f(211.1mg),产物不经纯化直接用于下一步反应。
第五步
4-(2-(4-(5-氯-2-氰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(吡啶-4-基)丙酰氨基)苯甲酸185g
将粗品化合物185f(211mg,514.86μmol)溶于10mL乙酸乙酯中,依次加入化合物8j(70.61mg,514.86μmol)、N,N-二异丙基乙胺(665.40mg,5.15mmol)和2,4,6-三丙基-1,3,5,2,4,6-三氧三磷酸-2,4,6-三氧化物的乙酸乙酯溶液(50%,982.90mg,1.54mmol),升温至68℃搅拌反应1.5小时。反应液冷却至室温,减压浓缩,所得残余物加入20mL水,用3M盐酸调节pH为5,析出固体,过滤,收集滤饼,滤饼用硅胶柱色谱法以洗脱剂体系A纯化,得标题化合物185g(35mg,产率:12.85%)。
第六步
(S)-4-(2-(4-(5-氯-2-氰基苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-3-(吡啶-4-基)丙酰氨基)苯甲酸185
将化合物185g(33mg,62.39μmol)进行手性制备(分离条件:手性制备柱:CHIRAL PAK IG 2.5*250mm;流动相:乙醇/乙酸=100/0.1(v/v),流速:30mL/min),收集其相应组分,减压浓缩,得到标题化合物185(14mg)。
MS m/z(ESI):529.5[M+1]
手性HPLC分析:保留时间9.464分钟,手性纯度97.5%(色谱柱:CHIRAL PAK IE 4.6*150mm 5um;流动相:正己烷/乙醇(含0.1%三氟乙酸)=50/50(v/v))。
1H NMR(400MHz,DMSO-d6)δ10.80(s,1H),8.43-8.32(m,2H),7.98-7.91(m,3H),7.70-7.55(m,5H),7.27-7.16(m,2H),6.41-6.38(m,1H),6.11-6.05(m,1H),3.68-3.59(m,4H),3.56-3.49(m,1H).
对比例2(实施例186)
(S)-4-(叔丁氧基)-2-(4-(5-氯-2-(3-甲基-1,2,4-噁二唑-5-基)苯基)-5-甲氧基-2-氧代吡啶-1(2H)-基)-N-(喹喔啉-6-基)丁酰胺186
Figure PCTCN2017099579-appb-000235
将化合物186a(80mg,132.65μmol,采用专利申请“WO2017005725”公开的方法制备而得)进行手性制备(分离条件:手性制备柱:Daicel IE 20*250mm,5um;流动相:乙醇/正己烷=40/60(v/v),流速:15mL/min),收集其相应组分,减压浓缩,得到标题化合物186(30mg)。
MS m/z(ESI):603.2[M+1]
手性HPLC分析:保留时间9.362分钟(色谱柱:CHIRAL PAK IE 4.6*150mm 5um;流动相:正己烷/乙醇(含0.1%三氟乙酸)=30/70(v/v))。
1H NMR(400MHz,CD3OD)δ10.80(s,1H),8.85-8.83(m,1H),8.80-8.79(m,1H),8.61-8.60(m,1H),8.08-8.07(m,2H),8.06(s,1H),7.70-7.69(m,1H),7.68-7.65(m,1H),7.57-7.50(m,1H),6.61(s,1H),6.00-5.90(m,1H),3.60-3.59(m,1H),3.57-3.47(s,3H),3.50-3.40(m,1H),2.53-2.52(m,1H),2.51-2.49(m,1H),2.43-2.34(m,3H)1.17(s,9H)
生物学评价
以下结合测试例进一步描述解释本发明,但这些实施例并非意味着限制本发明的范围。
本发明测试例中未注明具体条件的实验方法,通常按照常规条件,或按照商品制造厂商所建议的条件。未注明具体来源的试剂,为市场购买的常规试剂。
测试例1吸收光法检测本发明化合物对凝血因子XIa抑制的生物活性
1、实验材料
酶:凝血因子XIa蛋白酶(Abcam,货号ab62411)
底物:凝血因子XIa特异性底物(HYPHEN1310med,货号Biophen cs-21(66))
缓冲液:100mM tris-HCl,200mM NaCl,0.02%吐温20,pH7.4
2、实验步骤
将溶于100%DMSO的20mM受试化合物用100%DMSO稀释至200、20、2、0.2、0.02、0.002μM;在384孔板中每孔加入1μl化合物,空白和对照孔用DMSO替代,离心,将化合物离至底部。每孔加入10μl(2.5μg/ml)的FXIa酶溶液,空白孔加入10μl缓冲液替代,离心,将酶溶液离至底部。
最后每孔加入2mM底物10μl,离心,将底物液离至底部。
在37℃孵育10分钟;然后在405nm处测其吸光值。吸光值用graphpad进行曲线拟合并获得IC50见表一,
表一:
本发明化合物对凝血因子XIa抑制的IC50
化合物编号 IC50(FXIa)/(nM)
1 34
2 19
3 1741
4 39
5 16
6 1817
7 34
8 17
9 14
11 13
12 42
13 46
16 100
18 56
19 5690
20 36
21 36
23 24
24 36
25 1523
26 21
27 66
29 90
37 29
38 30
39 38
40 38
41 39
42 40
43 47
44 53
45 69
46 71
47 82
63 55
64 58
65 43
66 34
67 35
68 47
69 49
70 54
71 46
72 21
74 100
75 100
76 100
78 72
79 45
80 40
81 2678
82 68
83 >10000
84 42
85 92
86 99
87 73
88 74
89 42
90 100
91 50
92 21
93 42
94 24
95 7513
96 32
97 36
98 40
99 29
100 35
101 23
102 8213
103 70
104 8146
105 30
108 12
109 5456
110 53
111 18
112 >10000
113 33
114 27
115 >10000
119 45
120 49
121 67
122 100
123 100
124 100
141 66
146 37
147 56
155 81
158 46
159 33
160 68
161 13110
162 35
163 9114
164 30
166 42
167 92
168 43
171 35
172 8918
173 100
176 71
177 27
178 7956
179 31
180 4834
结论:本发明化合物对FXIa具有明显的抑制活性。
测试例2本发明化合物对人血液体外抗凝血作用的测定
3、实验材料
血浆:人血收集于不含抗凝剂的采血管中,加入3.8%柠檬酸钠(体积比1:9),室温2500rpm离心10min,收集血浆,分装保存于-80℃;
试剂:APTT试剂(活化部分凝血活酶时间测定试剂盒,SIEMENS,货号B4218-1)、氯化钙溶液;
仪器:凝血仪(SYSMEX,CA-500)。
2、实验检测
取分装血浆室温融化后混合均匀。将溶于100%DMSO的10000μM受试化合物用100%DMSO稀释至3000、300、200、150、75、30、10、3、0.3μM,空白为100%DMSO。将试剂、血浆、化合物放入凝血仪中对应位置,进行化合物的APTT检测。
3、数据处理
通过Prism进行曲线拟合,计算CT2,即2倍空白对照的APTT所对应的化合物的浓度,结果见表二,
表二:
本发明化合物对人血液体外抗凝血作用的CT2
化合物编号 抑制血小板凝集CT2(μM)
1 4.4
2 2.9
3 >10000
5 2.0
6 >10000
8 6.4
9 2.4
11 4.2
13 6.0
20 6.5
31 8.5
37 7.8
39 8.3
80 7.9
81 >10000
85 6.5
89 5.7
91 2.1
92 1.2
93 2.9
94 2.2
96 6.7
97 5.4
98 7.6
99 3.5
100 3.3
104 >10000
105 6.9
108 1.4
109 >10000
111 3.2
112 >10000
114 3.6
160 3.8
163 >10000
164 6.4
166 7.2
177 1.5
178 >10000
179 3.3
180 >10000
181 2.3
183 2.6
184 >10000
表三 本发明化合物对人血液体外抗凝血作用的CT2与已公开专利中相似化合物的比较。
化合物编号 抑制血小板凝集CT2(μM)
114 2.9
对比例2 13.1
结论:从表二中可以看出本发明化合物对人血液具有明显的抗凝血作用,从表三中可以看出本发明实施例114的CT2值是对比例2(实施例186)的4.5倍,二者结构上的区别为仅R1位置取代基不同,充分说明通式(AI)中R1为-C(O)R7对整个分子结构的抗凝血作用具有意料不到的影响。
药代动力学评价
测试例3、本发明化合物的药代动力学测试
1、摘要
以大鼠为受试动物,应用LC/MS/MS法测定了大鼠灌胃给予实施例5化合物、实施例9化合物、实施例11化合物、实施例13化合物、实施例29化合物、实施例31化合物、实施例80化合物、实施例84化合物、实施例108化合物、实施例111化合物、实施例114化合物、实施例160化合物、实施例171化合物和对比例1后不同时刻血浆中的药物浓度。研究本发明化合物在大鼠体内的药代动力学行为,评价其药代动力学特征。
2、试验方案
2.1试验药品
实施例5化合物、实施例9化合物、实施例11化合物、实施例13化合物、实施例29化合物、实施例31化合物、实施例80化合物、实施例84化合物、实施例108化合物、实施例111化合物、实施例114化合物、实施例160化合物、实施例171化合物和对比例1。
2.2试验动物
健康成年SD大鼠56只,雌雄各半,平均分为14组,购自上海杰思捷实验动物有限公司,动物生产许可证号:SCXK(沪)2013-0006。
2.3药物配制
大鼠:称取一定量药物,加5%体积的DMSO、5%体积的吐温80和90%的生理盐水配制成0.2mg/mL的无色澄清透明液体。
2.4给药
SD大鼠禁食过夜后灌胃给药,给药剂量均为2.0mg/kg,给药体积均为10.0mL/kg。
3.操作
大鼠灌胃给药实施例5化合物、实施例9化合物、实施例11化合物、实施例13化合物、实施例29化合物、实施例31化合物、实施例80化合物、实施例84化合物、实施例108化合物、实施例111化合物、实施例114化合物、实施例160化合物、实施例171化合物和对比例1,于给药前及给药后0.5,1.0,2.0,4.0,6.0,8.0,11.0,24.0小时由眼眶采血0.2mL,置于肝素化试管中,4℃、3500转/分钟离心10分钟分离血浆,于-20℃保存,给药后2小时进食。
测定不同浓度的药物灌胃给药后大鼠血浆中的待测化合物含量:取给药后各时刻的大鼠血浆25μL,加入内标溶液喜树碱30μL(100ng/mL),乙腈200μL,涡旋混合5分钟,离心10分钟(4000转/分钟),血浆样品取上清液3.0μL进行LC/MS/MS分析。
4、药代动力学参数结果
本发明化合物在大鼠体内的药代动力学参数如下:
Figure PCTCN2017099579-appb-000236
Figure PCTCN2017099579-appb-000237
结论:本发明化合物在大鼠体内的药代吸收良好,特别是实施例84与对比例1(实施例185)相比,两者的Cmax相差6.9倍,AUC相差7.5倍,二者结构上的差异主要为R1位置实施例84为乙酰基,对比例1的相应位置为氰基,充分说明本发明通式(AI)中R1为-C(O)R7对化合物的药代吸收有显著改善,本发明化合物具有药代动力学优势。
测试例4食蟹猴的APTT值的测定及药代动力学测试
1、试验目的
以食蟹猴为受试动物,应用凝血仪测定食蟹猴口服实施例5化合物和实施例108化合物后不同时刻的APTT值,评价其药效特性。
以食蟹猴为受试动物,应用LC/MS/MS法测定了食蟹猴灌胃给予实施例5化合物和实施例108化合物后不同时刻血浆中的药物浓度。研究本发明化合物在食蟹猴体内的药代动力学行为,评价其药代动力学特征。
2、试验动物
雄性食蟹猴6只(101、102、103、201、202和203),购于广西雄森灵长类实验动物养殖开发有限公司。
3、试验药品
实施例5化合物和实施例108化合物。
4、药物配制
称取一定量药物,加2%体积的DMSO、78%体积的PEG400和20%的0.5%CMC-Na配制成3.0mg/mL的无色澄清透明液体。
5、给药
食蟹猴禁食过夜后灌胃给药,给药剂量均为15.0mg/kg,给药体积均为5.0mL/kg。
6、食蟹猴的APTT值测定的试验方案
6.1试验材料
试剂:APTT试剂(活化部分凝血活酶时间测定试剂盒,SIEMENS,货号B4218-1)、PEG-400和CMC-Na;
仪器:凝血仪(SYSMEX,CA-1500)。
6.2APTT血浆样品采集及处理
给药前和给药后1h,2h,4h,8h和12h采血,每只动物每次经股静脉穿刺采约1.8mL血液,枸橼酸钠抗凝,血液样本采集后放置于提前标记好的离心管中,离心分离血浆(离心条件:3500转/分钟,10分钟,2-8℃)。血浆存放于-80℃冰箱。用于APTT测定。
6.3食蟹猴的APTT测定结果
表四 本发明化合物对食蟹猴的APTT值的测定结果
Figure PCTCN2017099579-appb-000238
结论:本发明化合物对食蟹猴的APTT值有明显延长,说明本发明化合具有良好的抗凝血作用。
7、食蟹猴的药代动力学测试的试验方案
7.1试验操作
食蟹猴灌胃给药实施例5化合物和实施例108化合物,于给药前及给药后0.25,0.5,1,2,4,6,8,12,24h由前肢静脉采血1.0ml,置于肝素抗凝试管中,3500rpm离心10min分离血浆,于-80℃保存;给药后2h进食。
测定不同浓度的药物食蟹猴给药后食蟹猴血浆中的待测化合物含量:取给药后各时刻的食蟹猴血浆25μL,加入内标溶液喜树碱30μL(100ng/mL),乙腈225μL,涡旋混合5分钟,离心10分钟(4000转/分钟),血浆样品取上清液1.0μL进行LC/MS/MS分析。
7.2食蟹猴的药代动力学参数结果
本发明化合物的在食蟹猴体内的药代动力学参数如下:
Figure PCTCN2017099579-appb-000239
Figure PCTCN2017099579-appb-000240
结论:本发明化合物在食蟹猴体内的药代吸收良好,具有药代动力学优势。

Claims (24)

  1. 一种通式(AI)所示的化合物:
    Figure PCTCN2017099579-appb-100001
    或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用的盐、或其前药,
    其中:
    环A为芳基或杂芳基;
    R1为-C(O)R7
    R2相同或不同,且各自独立地选自氢原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、氨基、硝基、氰基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    R3选自卤素、烷基、卤代烷基、烷氧基、环烷基氧基、卤代烷氧基、氨基、硝基、氰基、羟基、羟烷基和烷硫基,其中所述的烷基、烷氧基、卤代烷基、卤代烷氧基、羟烷基和烷硫基任选进一步被选自氘原子、卤素、烷氧基、卤代烷氧基、氨基、硝基、氰基、羟基和羟烷基中的一个或多个取代基所取代;
    R4选自氢原子、烷基、烷氧基、卤代烷基、羟烷基、环烷基、环烷基氧基、杂环基、芳基和杂芳基,其中所述的烷基、环烷基、杂环基、芳基和杂芳基任选进一步被一个或多个R9取代基所取代;
    R5相同或不同,且各自独立地选自氢原子、烷基、烷氧基、氧代基、卤素、卤代烷基、卤代烷氧基、氨基、硝基、氰基、羟基、羟烷基、环烷基、杂环基、芳基、杂芳基、-C(O)R8、-C(O)OR8、-NHC(O)R8、-NHC(O)OR8、-NR10R11、-C(O)NR10R11、-CH2NHC(O)OR8、-CH2NR10R11、-C(O)OCH(R10)R11和-S(O)mR8,其中所述的烷基任选进一步被选自氘原子、卤素、烷氧基、卤代烷氧基、氨基、硝基、氰基、羟基、羟烷基、-NR10R11和-NHC(O)OR8中的一个或多个取代基所取代;;
    R7选自氢原子、烷基、卤代烷基、烷氧基、卤代烷氧基、氨基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、烷氧基、环烷基、杂环基、芳基和杂芳基任选进一步被选自氢原子、氘原子、卤素、烷基、烷氧基、卤代烷氧基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基、杂环基、芳基和杂芳基中的一个或多个取代基所取代;
    R8选自氢原子、烷基、卤代烷基、羟基、氨基、环烷基、杂环基、芳基和杂芳基;
    R9选自氢原子、氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基、杂环基、芳基和杂芳基,其中所述的环烷基、杂环基、芳基和杂芳基任选进一步被选自氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基、杂环基、-NHC(O)R12和R13中的一个或多个取代基所取代;
    R10和R11相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、环烷基、杂环基、-C(O)OR8和-OC(O)OR12,其中所述的环烷基和杂环基任选进一步被选自氘原子、卤素、烷基、烷氧基、卤代烷基、氧代基、氨基、硝基、氰基、羟基和羟烷基中的一个或多个取代基所取代;
    R12选自氢原子、烷基、烷氧基、卤代烷基、卤代烷氧基、氨基、环烷基、杂环基、芳基和杂芳基;
    R13为芳基或杂芳基,其中所述的芳基和杂芳基任选进一步被选自氢原子、氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基和杂环基中的一个或多个取代基所取代;
    n为0、1、2、3或4;
    m为0、1或2;且
    s为0、1、2、3或4。
  2. 根据权利要求1所述的通式(AI)所示的化合物,其为通式(I)所示的化合物:
    Figure PCTCN2017099579-appb-100002
    或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用的盐,
    其中:
    L1为亚烷基,其中所述的亚烷基任选进一步被卤素或氘原子中的一个或多个所取代;
    R6选自氢原子、氘原子、卤素、烷基、烷氧基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基、杂环基、芳基和杂芳基,其中所述的环烷基、杂环基、芳基和杂芳基任选进一步被选自氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基、杂环基、-NHC(O)R12 和R13中的一个或多个取代基所取代;
    R12选自氢原子、烷基、卤代烷基、氨基、环烷基、杂环基、芳基和杂芳基;
    R13为芳基或杂芳基,其中所述的芳基和杂芳基任选进一步被选自氢原子、氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基、羟基、羟烷基、环烷基、环烷基氧基和杂环基中的一个或多个取代基所取代;
    环A、R1~R3、R5、n和s如权利要求1中所定义。
  3. 根据权利要求1或2中任一项所述的通式(AI)所示的化合物,其为通式(Iaa)所示的化合物:
    Figure PCTCN2017099579-appb-100003
    或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用的盐,
    其中:
    环A、L1、R1~R3、R5~R6、n和s如权利要求2中所定义。
  4. 根据权利要求1~3中任一项所述的通式(AI)所示的化合物,其中
    Figure PCTCN2017099579-appb-100004
    选自:
    Figure PCTCN2017099579-appb-100005
    Figure PCTCN2017099579-appb-100006
    Figure PCTCN2017099579-appb-100007
    其中R5和s如权利要求1中所定义。
  5. 根据权利要求1、2或4中任一项所述的通式(AI)所示的化合物,其为通式(II)所示的化合物:
    Figure PCTCN2017099579-appb-100008
    其中:
    R7选自烷基、环烷基和卤代烷基,其中所述的烷基或环烷基任选进一步被选自氘原子、卤素、烷基和环烷基中的一个或多个取代基所取代;
    L1、R2、R3、R5、R6和n如权利要求2中所定义。
  6. 根据权利要求1~5中任一项所述的通式(AI)所示的化合物,其中R5相同或不同,且各自独立地选自烷基、烷氧基、氧代基、卤素、卤代烷基、氰基、羟基、-C(O)OR8、-NHC(O)OR8、-C(O)NR10R11、-CH2NHC(O)OR8、-CH2NR10R11、-C(O)OCH(R10)R11和-S(O)mR8;R8选自氢原子、烷基、羟基和氨基;R10和R11相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、环烷基、杂环基、-C(O)OR8和-OC(O)OR12,其中所述的环烷基和杂环基任选进一步被选自氘原子、卤素、烷基、烷氧基、卤代烷基、氧代基、氨基、硝基、氰基、羟基和羟烷基中的一个或多个取代基所取代;R12为烷基。
  7. 根据权利要求1~6中任一项所述的通式(AI)所示的化合物,其中R1为-C(O)R7;R7选自烷基、环烷基和卤代烷基,其中所述的烷基或环烷基任选进一步被选自氘原子、卤素、烷基和环烷基中的一个或多个取代基所取代。
  8. 根据权利要求1~7中任一项所述的通式(AI)所示的化合物,其为通式(III)所示的化合物:
    Figure PCTCN2017099579-appb-100009
    其中:
    R5相同或不同,且各自独立地选自烷基、烷氧基、氧代基、卤素、卤代烷基、氰基、羟基、-C(O)OR8、-NHC(O)OR8、-C(O)NR10R11、-CH2NHC(O)OR8、-CH2NR10R11、-C(O)OCH(R10)R11和-S(O)mR8;R8选自氢原子、烷基、羟基和氨基;R10和R11相同或不同,且各自独立地选自氢原子、烷基、卤代烷基、环烷基、杂环基、-C(O)OR8和-OC(O)OR12,其中所述的环烷基和杂环基任选进一步被选自氘原子、卤素、烷基、烷氧基、卤代烷基、氧代基、氨基、硝基、氰基、羟基和羟烷基中的一个或多个取代基所取代;R12为烷基;
    R7选自烷基、环烷基和卤代烷基,其中所述的烷基或环烷基任选进一步被选自氘原子、卤素、烷基和环烷基中的一个或多个取代基所取代;
    L1、R2、R3、R6和n如权利要求2中所定义。
  9. 根据权利要求1~8中任一项所述的通式(AI)所示的化合物,其为通式(IV)所示的化合物:
    Figure PCTCN2017099579-appb-100010
    其中:
    L1、R2、R3、R6、R7和n如权利要求8中所定义。
  10. 根据权利要求1~9中任一项所述的通式(AI)所示的化合物,其中R2为卤素;n为0、1或2。
  11. 根据权利要求1~10中任一项所述的通式(AI)所示的化合物,其中R3为烷氧基,其中所述的烷氧基进一步任选被氘原子和卤素中的一个或多个所取代。
  12. 根据权利要求2~11中任一项所述的通式(I)所示的化合物,其中L1为-(CR14 2)x-,x为1~4的整数;R14为氢原子或氘原子;R6选自氢原子、氘原子、卤素、烷基、烷氧基、环烷基、环烷基氧基、杂环基、芳基和杂芳基;其中所述的环烷基、杂环基、芳基和杂芳基任选进一步被选自氘原子、卤素、烷基、烷氧基、环烷基、杂环基、-NHC(O)R12和R13中的一个或多个取代基所取代;R12为烷基或环烷基;R13为芳基或杂芳基,其中所述的芳基和杂芳基任选进一步被选自氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基和羟基中的一个或多个取代基所取代。
  13. 根据权利要求2~12中任一项所述的通式(I)所示的化合物,其中L1为-CH2-或-CD2-;R6选自环烷基、杂环基、芳基或杂芳基;其中所述的环烷基、杂环基、芳基和杂芳基任选进一步被选自氘原子、卤素、烷基、烷氧基、环烷基、杂环基、-NHC(O)R12和R13中的一个或多个取代基所取代;R12为烷基或环烷基;R13为芳基或杂芳基,其中所述的芳基和杂芳基任选进一步被选自氘原子、卤素、烷基、烷氧基、卤代烷基、氨基、硝基、氰基和羟基中的一个或多个取代基所取代。
  14. 根据权利要求2~12中任一项所述的通式(I)所示的化合物,其中L1为-CH2CH2-;R6为烷基、烷氧基或环烷基氧基。
  15. 根据权利要求1~12中任一项所述的通式(AI)所示的化合物,其中所述化合物选自:
    Figure PCTCN2017099579-appb-100011
    Figure PCTCN2017099579-appb-100012
    Figure PCTCN2017099579-appb-100013
    Figure PCTCN2017099579-appb-100014
    Figure PCTCN2017099579-appb-100015
    Figure PCTCN2017099579-appb-100016
    Figure PCTCN2017099579-appb-100017
    Figure PCTCN2017099579-appb-100018
    Figure PCTCN2017099579-appb-100019
    Figure PCTCN2017099579-appb-100020
  16. 一种制备根据权利要求1所述的通式(AI)所示的化合物的方法,该方法包括:
    Figure PCTCN2017099579-appb-100021
    通式(AI-A)的化合和通式(AI-B)的化合物或其盐酸盐在碱性条件下,进行缩合反应,任选在碱性条件下水解,得到通式(AI)化合物;
    其中:
    环A、R1~R5、n和s如权利要求1中所定义。
  17. 一种制备根据权利要求2所述的通式(I)所示的化合物的方法,该方法包括:
    Figure PCTCN2017099579-appb-100022
    通式(I-A)的化合和通式(AI-B)的化合物或其盐酸盐在碱性条件下,进行缩合反应,任选在碱性条件下水解,得到通式(I)化合物;
    其中:
    环A、L1、R1~R3、R5~R6、n和s如权利要求2中所定义。
  18. 一种药物组合物,其含有治疗有效量的根据权利要求1~15中任一项所述的通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用的盐以及药学上可接受的载体、稀释剂和赋形剂。
  19. 根据权利要求1~15任一项所述的通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式或其可药用的盐或根据权利要求18所述的药物组合物,在制备用于抑制XIa因子的药物中的用途。
  20. 根据权利要求1~15中任一项所述的通式(AI)所示的化合物或其互变异构体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用的盐或根据权利要求18所述的药物组合物,其用作XIa抑制剂。
  21. 根据权利要求1~15任一项所述的通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式或其可药用的盐或根据权利要求18所述的药物组合物,在制备预防和/或治疗XIa因子介导的疾病的药物中的用途。
  22. 一种预防和/或治疗XIa因子介导的疾病的方法,其包括向患者施用治疗有效剂量的根据权利要求1~15任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式或其可药用盐或根据权利要求18所述的药物组合物。
  23. 根据权利要求1~15任一项所述的通式(AI)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式或其可药用的盐或根据权利要求18所述的药物组合物,在制备预防和/或治疗心脑血管疾病的药物中的用途,其中所述的心血管疾病,优选为血栓栓塞性疾病,更优选为心肌梗塞、心绞痛、血管成型术或主动脉冠状动脉分流术后的再阻塞和再狭窄、弥散性血管内凝血、中风、短暂的局部缺血发作、周围动脉闭塞性疾病、肺栓塞或深部静脉血栓形成。
  24. 一种预防和/或治疗心脑血管疾病的方法,其中所述的心血管疾病,优选为血栓栓塞性疾病,更优选为心肌梗塞、心绞痛、血管成型术或主动脉冠状动脉分流术后的再阻塞和再狭窄、弥散性血管内凝血、中风、短暂的局部缺血发作、周围动脉闭塞性疾病、肺栓塞或深部静脉血栓形成,其包括向患者施用治疗有效剂量的根据权利要求1~15任一项所述的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式或其可药用盐或根据权利要求18所述的药物组合物。
PCT/CN2017/099579 2016-08-31 2017-08-30 氧代吡啶酰胺类衍生物、其制备方法及其在医药上的应用 WO2018041122A1 (zh)

Priority Applications (18)

Application Number Priority Date Filing Date Title
RU2019107938A RU2742771C2 (ru) 2016-08-31 2017-08-30 Производное оксопиколинамида, способ его получения и его фармацевтическое применение
CN202111196664.6A CN113929618B (zh) 2016-08-31 2017-08-30 氧代吡啶酰胺类衍生物、其制备方法及其在医药上的应用
UAA201902875A UA125520C2 (uk) 2016-08-31 2017-08-30 Похідні оксопіколінаміду, спосіб їх отримання та їх фармацевтичне застосування
CA3031592A CA3031592A1 (en) 2016-08-31 2017-08-30 Oxopicolinamide derivative, preparation method therefor and pharmaceutical use thereof
AU2017317988A AU2017317988B2 (en) 2016-08-31 2017-08-30 Oxopicolinamide derivative, preparation method therefor and pharmaceutical use thereof
MX2019001481A MX2019001481A (es) 2016-08-31 2017-08-30 Derivado de oxopicolinamida, metodo de preparacion y uso farmaceutico del mismo.
BR112019003557-5A BR112019003557A2 (pt) 2016-08-31 2017-08-30 derivados de oxopicolinamida, método de preparação dos mesmos e uso farmacêutico dos mesmos
JP2019511336A JP7032814B2 (ja) 2016-08-31 2017-08-30 オキソピコリンアミド誘導体、その製造方法およびその医薬用途
PL17845424T PL3486242T3 (pl) 2016-08-31 2017-08-30 Pochodna oksopikolinamidu, sposób jej wytwarzania i zastosowanie farmaceutyczne
DK17845424.5T DK3486242T3 (da) 2016-08-31 2017-08-30 Oxopicolinamidderivat, fremgangsmåde til fremstilling deraf og farmaceutisk anvendelse deraf
CN201780004214.8A CN108495850B (zh) 2016-08-31 2017-08-30 氧代吡啶酰胺类衍生物、其制备方法及其在医药上的应用
ES17845424T ES2903283T3 (es) 2016-08-31 2017-08-30 Derivado de oxipicolinamida, método de preparación y uso farmacéutico del mismo
US16/328,128 US10633375B2 (en) 2016-08-31 2017-08-30 Oxopicolinamide derivative, preparation method therefor and pharmaceutical use thereof
CN202111197415.9A CN113912586B (zh) 2016-08-31 2017-08-30 氧代吡啶酰胺类衍生物、其制备方法及其在医药上的应用
EP17845424.5A EP3486242B1 (en) 2016-08-31 2017-08-30 Oxopicolinamide derivative, preparation method therefor and pharmaceutical use thereof
KR1020197007965A KR102416718B1 (ko) 2016-08-31 2017-08-30 옥소피콜린아미드 유도체, 이에 대한 제조 방법 및 이의 약학적 용도
HK18114604.9A HK1255566A1 (zh) 2016-08-31 2018-11-15 氧代吡啶酰胺類衍生物、其製備方法及其在醫藥上的應用
US16/804,180 US11084808B2 (en) 2016-08-31 2020-02-28 Oxopicolinamide derivative, preparation method therefor and pharmaceutical use thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201610789384 2016-08-31
CN201610789384.9 2016-08-31
CN201710014133.8 2017-01-09
CN201710014133 2017-01-09

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/328,128 A-371-Of-International US10633375B2 (en) 2016-08-31 2017-08-30 Oxopicolinamide derivative, preparation method therefor and pharmaceutical use thereof
US16/804,180 Continuation US11084808B2 (en) 2016-08-31 2020-02-28 Oxopicolinamide derivative, preparation method therefor and pharmaceutical use thereof

Publications (1)

Publication Number Publication Date
WO2018041122A1 true WO2018041122A1 (zh) 2018-03-08

Family

ID=61300096

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/099579 WO2018041122A1 (zh) 2016-08-31 2017-08-30 氧代吡啶酰胺类衍生物、其制备方法及其在医药上的应用

Country Status (19)

Country Link
US (2) US10633375B2 (zh)
EP (1) EP3486242B1 (zh)
JP (1) JP7032814B2 (zh)
KR (1) KR102416718B1 (zh)
CN (3) CN113929618B (zh)
AU (1) AU2017317988B2 (zh)
BR (1) BR112019003557A2 (zh)
CA (1) CA3031592A1 (zh)
DK (1) DK3486242T3 (zh)
ES (1) ES2903283T3 (zh)
HK (1) HK1255566A1 (zh)
HU (1) HUE057289T2 (zh)
MX (1) MX2019001481A (zh)
PL (1) PL3486242T3 (zh)
PT (1) PT3486242T (zh)
RU (1) RU2742771C2 (zh)
TW (1) TWI756257B (zh)
UA (1) UA125520C2 (zh)
WO (1) WO2018041122A1 (zh)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019197244A1 (en) 2018-04-10 2019-10-17 Bayer Pharma Aktiengesellschaft A substituted oxopyridine derivative
WO2020007256A1 (zh) * 2018-07-02 2020-01-09 江苏恒瑞医药股份有限公司 一种氧代吡啶酰胺类衍生物的晶型及制备方法
WO2020015698A1 (zh) 2018-07-19 2020-01-23 江苏恒瑞医药股份有限公司 一种凝血因子XIa抑制剂及其中间体的制备方法
EP3741746A4 (en) * 2018-02-27 2020-12-16 Jiangsu Hengrui Medicine Co., Ltd. CRYSTAL FORM OF OXOPICOLINAMIDE DERIVATIVE AND MANUFACTURING PROCESS FOR IT
TWI722466B (zh) * 2019-07-01 2021-03-21 大陸商江蘇恒瑞醫藥股份有限公司 氧代吡啶醯胺類衍生物的晶型及製備方法
WO2021057818A1 (zh) 2019-09-27 2021-04-01 深圳信立泰药业股份有限公司 FXIa抑制剂及其制备方法和医药用途
CN112675173A (zh) * 2020-12-25 2021-04-20 华南理工大学 FXIa抑制剂化合物或其盐的医药用途
CN113135929A (zh) * 2020-01-17 2021-07-20 江西济民可信集团有限公司 呋喃并吡啶酮酰胺化合物及其制备方法和用途
TWI749881B (zh) * 2019-11-21 2021-12-11 大陸商深圳信立泰藥業股份有限公司 二氧代哌類衍生物、其製備方法及其在醫藥上的應用
WO2022083706A1 (zh) * 2020-10-23 2022-04-28 深圳信立泰药业股份有限公司 FXIa抑制剂化合物的盐及其制备方法和医药用途
WO2022083707A1 (zh) * 2020-10-23 2022-04-28 深圳信立泰药业股份有限公司 FXIa抑制剂化合物或其盐的医药用途
WO2022122035A1 (zh) * 2020-12-11 2022-06-16 上海领泰生物医药科技有限公司 凝血因子XIa抑制剂及其制备方法和应用
RU2779013C2 (ru) * 2018-07-19 2022-08-30 Цзянсу Хэнжуй Медицин Ко., Лтд. Способ получения ингибитора фактора свертывания крови xia и его интермедиата
WO2022194225A1 (zh) * 2021-03-18 2022-09-22 深圳信立泰药业股份有限公司 FXIa抑制剂化合物杂质及其制备方法和用途FXIa抑制剂化合物杂质及其制备方法和用途

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190119213A1 (en) * 2015-03-19 2019-04-25 Bayer Pharma Aktiengesellschaft Oxo-pyridine-derivatives as factor xia inhibitors for the treatment of thrombosis
WO2018041122A1 (zh) * 2016-08-31 2018-03-08 江苏恒瑞医药股份有限公司 氧代吡啶酰胺类衍生物、其制备方法及其在医药上的应用
US20230083388A1 (en) * 2018-04-09 2023-03-16 Yale University Bifunctional Small Molecules to Target the Selective Degradation of Circulating Proteins
JP2022513919A (ja) * 2018-12-17 2022-02-09 バイエル・アクチエンゲゼルシヤフト 血栓性または血小板塞栓性疾患および/または血栓性または血小板塞栓性合併症の治療および/または予防のための置換オキソピリジン誘導体

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104136431A (zh) * 2011-12-21 2014-11-05 小野药品工业株式会社 作为凝血因子XIa抑制剂的吡啶酮和嘧啶酮衍生物
WO2016046156A1 (de) * 2014-09-24 2016-03-31 Bayer Pharma Aktiengesellschaft Substituierte oxopyridin-derivate

Family Cites Families (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997011695A1 (en) 1995-09-28 1997-04-03 Merck Sharp & Dohme Limited Substituted indolylpropyl-piperazine derivatives as 5-ht1dalpha agonists
WO1996030396A1 (en) 1995-03-24 1996-10-03 Molecumetics Ltd. β-SHEET MIMETICS AND USE THEREOF AS PROTEASE INHIBITORS
ES2192764T3 (es) 1998-02-12 2003-10-16 Molecumetics Ltd Mimeticos de lamina beta y metodos relacionados con el uso de los mismos.
WO2003064413A1 (en) * 2002-02-01 2003-08-07 Astrazeneca Ab Quinazoline compounds
KR100669587B1 (ko) 2002-04-26 2007-01-15 에프. 호프만-라 로슈 아게 치환된 페닐아세트아마이드 및 그의 글루코키나제 활성화제로서의 용도
AU2003243657A1 (en) 2002-06-26 2004-01-19 Bristol-Myers Squibb Company Amino-bicyclic pyrazinones and pyridinones
US7208601B2 (en) 2003-08-08 2007-04-24 Mjalli Adnan M M Aryl and heteroaryl compounds, compositions, and methods of use
US7453002B2 (en) 2004-06-15 2008-11-18 Bristol-Myers Squibb Company Five-membered heterocycles useful as serine protease inhibitors
TW200635899A (en) 2004-12-22 2006-10-16 Astrazeneca Ab Chemical compounds
US8247556B2 (en) 2005-10-21 2012-08-21 Amgen Inc. Method for preparing 6-substituted-7-aza-indoles
US10022352B2 (en) 2006-04-07 2018-07-17 Vertex Pharmaceuticals Incorporated Modulators of ATP-binding cassette transporters
JP2008013527A (ja) 2006-07-10 2008-01-24 Sankyo Co Ltd チエノ[3,2−d]ピリミジン−2,4−ジアミン誘導体
AU2008340991B2 (en) 2007-12-21 2012-02-23 University Health Network Indazolyl, benzimidazolyl, benzotriazolyl substituted indolinone derivatives as kinase inhibitors useful in the treatment of cancer
JP5337259B2 (ja) 2009-02-26 2013-11-06 レビバ ファーマシューティカルズ,インコーポレーテッド アリールピペラジン誘導体の組成物、合成および利用方法
PL2444395T3 (pl) 2009-06-19 2016-06-30 D Western Therapeutics Inst Inc Podstawiona pochodna izochinoliny
JP2013519673A (ja) 2010-02-10 2013-05-30 レクシコン ファーマシューティカルズ インコーポレイテッド 転移性骨疾患の治療のためのトリプトファンヒドロキシラーゼ阻害剤
WO2012044090A2 (ko) 2010-09-29 2012-04-05 크리스탈지노믹스(주) 단백질 키나제 억제 활성을 갖는 신규한 아미노퀴나졸린 화합물
CU24152B1 (es) 2010-12-20 2016-02-29 Irm Llc 1,2 oxazol-8-azabiciclo[3,2,1]octano 8 il como moduladores de fxr
CN102093364B (zh) 2011-01-07 2015-01-28 北京赛林泰医药技术有限公司 作为FAK/Pyk2抑制剂的2,4-二氨基-6,7-二氢-5H-吡咯并[2,3]嘧啶衍生物
US9259000B2 (en) 2011-02-28 2016-02-16 Riken Auxin biosynthesis inhibitor
US8852599B2 (en) 2011-05-26 2014-10-07 Bristol-Myers Squibb Company Immunoconjugates, compositions for making them, and methods of making and use
JP6247632B2 (ja) 2011-06-19 2017-12-13 ヴィアメット ファーマスーティカルズ,インコーポレイテッド メタロ酵素阻害剤化合物
CN102863512B (zh) 2011-07-07 2016-04-20 上海泓博智源医药技术有限公司 抗病毒化合物
WO2013042782A1 (ja) 2011-09-22 2013-03-28 武田薬品工業株式会社 縮合複素環化合物
CN103987696B (zh) 2011-10-14 2016-12-21 百时美施贵宝公司 作为因子xia抑制剂的取代的四氢异喹啉化合物
MY165742A (en) 2011-10-14 2018-04-23 Bristol Myers Squibb Co Substituted tetrahydroisoquinoline compounds as factor xia inhibitors
JP2014532746A (ja) 2011-11-09 2014-12-08 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング バニロイド受容体リガンドとしての、co含有基で置換されたフェニル部分を担持する置換ピラゾリルをベースとするカルボキサミドおよび尿素誘導体
JP6154887B2 (ja) 2012-03-20 2017-06-28 セラゴン ファーマシューティカルズ,インク. エストロゲン受容体モジュレーターおよびその使用
EP2832734A4 (en) 2012-03-28 2015-08-26 Takeda Pharmaceutical HETEROCYCLIC CONNECTION
US20160137639A1 (en) 2012-10-26 2016-05-19 Japan Tobacco Inc. Triazole-isoxazole compound and medical use thereof
WO2014075387A1 (zh) 2012-11-14 2014-05-22 上海医药集团股份有限公司 一类二芳基乙内酰脲衍生物、其制备方法、药物组合物和应用
WO2014100833A1 (en) 2012-12-21 2014-06-26 Northwestern University Benzamide compounds and related methods of use
WO2014135095A1 (en) 2013-03-06 2014-09-12 Syngenta Participations Ag Dihydrobenzofuran derivatives as insecticidal compounds
US9809545B2 (en) * 2013-03-27 2017-11-07 Merck Sharp & Dohme Corp. Factor XIa inhibitors
TWI633089B (zh) * 2013-03-28 2018-08-21 拜耳製藥股份有限公司 經取代的酮基吡啶衍生物
US8975417B2 (en) 2013-05-27 2015-03-10 Novartis Ag Pyrazolopyrrolidine derivatives and their use in the treatment of disease
CN103450146B (zh) 2013-08-27 2015-03-11 合肥工业大学 一种催化合成高纯度4-氯甲基-5-甲基-1,3-二氧杂环戊烯-2-酮的方法
JP6368367B2 (ja) 2013-10-30 2018-08-01 バイエル ファーマ アクチエンゲゼルシャフト 置換オキソピリジン誘導体
MX2016007951A (es) 2013-12-19 2016-09-09 Gruenenthal Gmbh Pirrol carboxamidas iv fluorometil-sustituidas.
KR102502485B1 (ko) 2014-01-21 2023-02-21 얀센 파마슈티카 엔.브이. 대사 조절형 글루탐산 작동성 수용체 제2아형의 양성 알로스테릭 조절제 또는 오르토스테릭 작동제를 포함하는 조합 및 그 용도
CN105829298B (zh) * 2014-02-14 2019-02-01 四川海思科制药有限公司 一种吡啶酮或嘧啶酮衍生物、及其制备方法和应用
BR112016018564B1 (pt) 2014-02-14 2021-08-31 Nanjing Scienx Biological Technology Co. Ltd Formulações ecologicamente favoráveis de benzoato de emamectina e métodos para preparação das mesmas
TWI659019B (zh) 2014-02-28 2019-05-11 日商帝人製藥股份有限公司 吡唑醯胺衍生物
MA39763A (fr) 2014-03-20 2017-01-25 Samumed Llc Indazole-3-carboxamides 5-substitués, et préparation et utilisation de ceux-ci
WO2016044626A1 (en) 2014-09-17 2016-03-24 Epizyme, Inc. Arginine methyltransferase inhibitors and uses thereof
WO2016046159A1 (de) 2014-09-24 2016-03-31 Bayer Pharma Aktiengesellschaft Substituierte oxopyridin-derivate
WO2016045125A1 (en) 2014-09-28 2016-03-31 Merck Sharp & Dohme Corp. Inhibitors of hif prolyl hydroxylase
WO2016053794A1 (en) 2014-09-30 2016-04-07 Bristol-Myers Squibb Company Quinazoline-based kinase inhibitors
CA2980010C (en) 2015-03-27 2023-10-03 The Scripps Research Institute Lipid probes and uses thereof
JO3703B1 (ar) 2015-07-09 2021-01-31 Bayer Pharma AG مشتقات أوكسوبيريدين مستبدلة
CN105348251A (zh) 2015-12-07 2016-02-24 东北制药集团股份有限公司 一种3,4-亚甲二氧基苯胺的制备方法
CN106242941B (zh) 2016-07-08 2019-01-18 山东日兴新材料股份有限公司 一种环丙基溴甲烷的合成方法
WO2018041122A1 (zh) * 2016-08-31 2018-03-08 江苏恒瑞医药股份有限公司 氧代吡啶酰胺类衍生物、其制备方法及其在医药上的应用

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104136431A (zh) * 2011-12-21 2014-11-05 小野药品工业株式会社 作为凝血因子XIa抑制剂的吡啶酮和嘧啶酮衍生物
WO2016046156A1 (de) * 2014-09-24 2016-03-31 Bayer Pharma Aktiengesellschaft Substituierte oxopyridin-derivate

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3486242A4 *

Cited By (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3741746A4 (en) * 2018-02-27 2020-12-16 Jiangsu Hengrui Medicine Co., Ltd. CRYSTAL FORM OF OXOPICOLINAMIDE DERIVATIVE AND MANUFACTURING PROCESS FOR IT
US11299462B2 (en) 2018-02-27 2022-04-12 Jiangsu Hengrui Medicine Co., Ltd. Crystal form of oxopicolinamide derivative and preparation method therefor
US11884660B2 (en) 2018-04-10 2024-01-30 Bayer Pharma Aktiengesellschaft Substituted oxopyridine derivative
JP7333336B2 (ja) 2018-04-10 2023-08-24 バイエル ファーマ アクチエンゲゼルシャフト 置換オキソピリジン誘導体
WO2019197244A1 (en) 2018-04-10 2019-10-17 Bayer Pharma Aktiengesellschaft A substituted oxopyridine derivative
JP2021521132A (ja) * 2018-04-10 2021-08-26 バイエル ファーマ アクチエンゲゼルシャフト 置換オキソピリジン誘導体
JP2021529201A (ja) * 2018-07-02 2021-10-28 江▲蘇▼恒瑞医▲薬▼股▲フン▼有限公司Jiangsu Hengrui Medicine Co., Ltd. オキシピリジンアミド誘導体の結晶形およびその調製方法
WO2020007256A1 (zh) * 2018-07-02 2020-01-09 江苏恒瑞医药股份有限公司 一种氧代吡啶酰胺类衍生物的晶型及制备方法
US11225462B2 (en) 2018-07-02 2022-01-18 Jiangsu Hengrui Medicine Co., Ltd. Crystal forms of oxypyridine amide derivative and preparation method therefor
CN112004810A (zh) * 2018-07-02 2020-11-27 江苏恒瑞医药股份有限公司 一种氧代吡啶酰胺类衍生物的晶型及制备方法
CN112004810B (zh) * 2018-07-02 2022-04-08 江苏恒瑞医药股份有限公司 一种氧代吡啶酰胺类衍生物的晶型及制备方法
CN111094268A (zh) * 2018-07-19 2020-05-01 江苏恒瑞医药股份有限公司 一种凝血因子XIa抑制剂及其中间体的制备方法
RU2779013C2 (ru) * 2018-07-19 2022-08-30 Цзянсу Хэнжуй Медицин Ко., Лтд. Способ получения ингибитора фактора свертывания крови xia и его интермедиата
WO2020015698A1 (zh) 2018-07-19 2020-01-23 江苏恒瑞医药股份有限公司 一种凝血因子XIa抑制剂及其中间体的制备方法
TWI722466B (zh) * 2019-07-01 2021-03-21 大陸商江蘇恒瑞醫藥股份有限公司 氧代吡啶醯胺類衍生物的晶型及製備方法
CN114206854A (zh) * 2019-09-27 2022-03-18 深圳信立泰药业股份有限公司 FXIa抑制剂及其制备方法和医药用途
TWI756848B (zh) * 2019-09-27 2022-03-01 大陸商深圳信立泰藥業股份有限公司 FXIa抑制劑及其製備方法和醫藥用途
JP7450024B2 (ja) 2019-09-27 2024-03-14 シェンヅェン サルブリス ファーマシューティカルズ カンパニー リミテッド FXIa阻害剤及びその調製方法と医薬用途
RU2802878C9 (ru) * 2019-09-27 2023-09-21 Шэньчжень Салюбрис Фармасьютикалс Ко. Лтд Ингибиторы fxia, способ их получения и применение в фармацевтике
RU2802878C1 (ru) * 2019-09-27 2023-09-05 Шэньчжень Салюбрис Фармасьютикалс Ко. Лтд Ингибиторы fxia, способ их получения и применение в фармацевтике
WO2021057818A1 (zh) 2019-09-27 2021-04-01 深圳信立泰药业股份有限公司 FXIa抑制剂及其制备方法和医药用途
CN114206854B (zh) * 2019-09-27 2023-02-24 深圳信立泰药业股份有限公司 FXIa抑制剂及其制备方法和医药用途
CN116120240A (zh) * 2019-09-27 2023-05-16 深圳信立泰药业股份有限公司 FXIa抑制剂化合物及其制备方法和医药用途
JP2022550367A (ja) * 2019-09-27 2022-12-01 シェンヅェン サルブリス ファーマシューティカルズ カンパニー リミテッド FXIa阻害剤及びその調製方法と医薬用途
AU2020355830B2 (en) * 2019-09-27 2023-04-13 Shenzhen Salubris Pharmaceuticals Co. Ltd FXIa inhibitors and preparation method therefor and pharmaceutical use thereof
TWI749881B (zh) * 2019-11-21 2021-12-11 大陸商深圳信立泰藥業股份有限公司 二氧代哌類衍生物、其製備方法及其在醫藥上的應用
CN113135929A (zh) * 2020-01-17 2021-07-20 江西济民可信集团有限公司 呋喃并吡啶酮酰胺化合物及其制备方法和用途
CN113135929B (zh) * 2020-01-17 2024-04-19 江西济民可信集团有限公司 呋喃并吡啶酮酰胺化合物及其制备方法和用途
CN115427043B (zh) * 2020-10-23 2023-04-07 深圳信立泰药业股份有限公司 FXIa抑制剂化合物或其盐的医药用途
CN115427043A (zh) * 2020-10-23 2022-12-02 深圳信立泰药业股份有限公司 FXIa抑制剂化合物或其盐的医药用途
WO2022083707A1 (zh) * 2020-10-23 2022-04-28 深圳信立泰药业股份有限公司 FXIa抑制剂化合物或其盐的医药用途
WO2022083706A1 (zh) * 2020-10-23 2022-04-28 深圳信立泰药业股份有限公司 FXIa抑制剂化合物的盐及其制备方法和医药用途
WO2022122035A1 (zh) * 2020-12-11 2022-06-16 上海领泰生物医药科技有限公司 凝血因子XIa抑制剂及其制备方法和应用
CN117480164A (zh) * 2020-12-11 2024-01-30 上海领泰生物医药科技有限公司 凝血因子XIa抑制剂及其制备方法和应用
CN117480164B (zh) * 2020-12-11 2024-07-05 上海领泰生物医药科技有限公司 凝血因子XIa抑制剂及其制备方法和应用
CN112675173B (zh) * 2020-12-25 2022-04-05 华南理工大学 FXIa抑制剂化合物或其盐的医药用途
CN112675173A (zh) * 2020-12-25 2021-04-20 华南理工大学 FXIa抑制剂化合物或其盐的医药用途
WO2022194225A1 (zh) * 2021-03-18 2022-09-22 深圳信立泰药业股份有限公司 FXIa抑制剂化合物杂质及其制备方法和用途FXIa抑制剂化合物杂质及其制备方法和用途

Also Published As

Publication number Publication date
US10633375B2 (en) 2020-04-28
BR112019003557A2 (pt) 2019-05-21
HK1255566A1 (zh) 2019-08-23
CN108495850B (zh) 2021-11-26
US11084808B2 (en) 2021-08-10
US20190185464A1 (en) 2019-06-20
TW201811748A (zh) 2018-04-01
CN113912586A (zh) 2022-01-11
AU2017317988A1 (en) 2019-02-21
JP2019528299A (ja) 2019-10-10
PL3486242T3 (pl) 2022-03-21
RU2019107938A3 (zh) 2020-10-01
UA125520C2 (uk) 2022-04-13
RU2742771C2 (ru) 2021-02-10
TWI756257B (zh) 2022-03-01
PT3486242T (pt) 2022-01-06
MX2019001481A (es) 2019-05-15
AU2017317988B2 (en) 2020-12-24
EP3486242A1 (en) 2019-05-22
CN113929618A (zh) 2022-01-14
CN108495850A (zh) 2018-09-04
ES2903283T3 (es) 2022-03-31
DK3486242T3 (da) 2022-01-10
JP7032814B2 (ja) 2022-03-09
US20200199115A1 (en) 2020-06-25
CN113929618B (zh) 2023-08-11
HUE057289T2 (hu) 2022-05-28
EP3486242B1 (en) 2021-12-08
RU2019107938A (ru) 2020-10-01
CA3031592A1 (en) 2018-03-08
CN113912586B (zh) 2023-04-07
EP3486242A4 (en) 2019-12-04
KR20190040037A (ko) 2019-04-16
KR102416718B1 (ko) 2022-07-05

Similar Documents

Publication Publication Date Title
TWI756257B (zh) 吡啶酮醯胺類衍生物、其製備方法及其在醫藥上的應用
CN111377917B (zh) 杂环类化合物、中间体、其制备方法及应用
CA3029857C (en) Aromatic acetylene or aromatic ethylene compound, intermediate, preparation method, pharmaceutical composition and use thereof
TWI714600B (zh) 哌啶類衍生物、其製備方法及其在醫藥上的應用
TWI718207B (zh) 苯並呋喃類衍生物、其製備方法及其在醫藥上的應用
KR102057877B1 (ko) 질소함유 헤테로고리 유도체 및 그의 약물에서의 용도
CN108017603B (zh) 三环化合物及其用途
CN107793396B (zh) 环氧基取代的氧代吡啶类衍生物、其制备方法及其在医药上的应用
WO2022135470A1 (zh) 作为kras抑制剂的杂环化合物的制备及其应用方法
WO2019223715A1 (zh) 苯并哌啶或杂芳基并哌啶类衍生物、其制备方法及其在医药上的应用
WO2020192650A1 (zh) 酰胺类化合物制备方法及其在医药领域的应用
WO2022194269A1 (zh) 新型egfr降解剂
CN105658641A (zh) 氮茚-酰胺类衍生物、其制备方法及其在医药上的应用
KR102668958B1 (ko) 브루톤 티로신 키나아제 억제제인 치환된 1-아미노-1h-이미다졸-5-카르복사미드
CN107200726B (zh) 吡咯烷-酰胺类衍生物、其制备方法及其在医药上的应用
CN105218564A (zh) 三环噁唑烷酮类化合物及其制备方法和用途
TW201934558A (zh) 苯并雜芳基類衍生物、其製備方法及其在醫藥上的應用
WO2023093832A1 (zh) 三并环衍生物及其制备方法和应用
KR20200125967A (ko) 수용체 억제제, 이를 포함하는 약학적 조성물 및 그 용도

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: 17845424

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3031592

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2017317988

Country of ref document: AU

Date of ref document: 20170830

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019511336

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017845424

Country of ref document: EP

Effective date: 20190213

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112019003557

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20197007965

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112019003557

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20190221