WO2020168939A1 - 杂环化合物、包含其的药物组合物及其制备方法和用途 - Google Patents

杂环化合物、包含其的药物组合物及其制备方法和用途 Download PDF

Info

Publication number
WO2020168939A1
WO2020168939A1 PCT/CN2020/074696 CN2020074696W WO2020168939A1 WO 2020168939 A1 WO2020168939 A1 WO 2020168939A1 CN 2020074696 W CN2020074696 W CN 2020074696W WO 2020168939 A1 WO2020168939 A1 WO 2020168939A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
group
cycloalkyl
alkoxy
Prior art date
Application number
PCT/CN2020/074696
Other languages
English (en)
French (fr)
Inventor
陈忠辉
段霜霜
李桂英
韩润丰
孙启正
景连栋
韩晓军
田强
宋宏梅
薛彤彤
王晶翼
Original Assignee
四川科伦博泰生物医药股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to SG11202107848TA priority Critical patent/SG11202107848TA/en
Priority to MX2021009971A priority patent/MX2021009971A/es
Priority to CN202080009954.2A priority patent/CN113316578B/zh
Priority to JP2021571055A priority patent/JP2022521859A/ja
Application filed by 四川科伦博泰生物医药股份有限公司 filed Critical 四川科伦博泰生物医药股份有限公司
Priority to CA3130245A priority patent/CA3130245A1/en
Priority to BR112021016299-2A priority patent/BR112021016299A2/pt
Priority to CN202311264080.7A priority patent/CN117327078A/zh
Priority to AU2020226422A priority patent/AU2020226422A1/en
Priority to US17/431,930 priority patent/US20220144847A1/en
Priority to EP20760123.8A priority patent/EP3929198A4/en
Priority to CN202311221283.8A priority patent/CN117263945A/zh
Priority to KR1020217022806A priority patent/KR20210131314A/ko
Publication of WO2020168939A1 publication Critical patent/WO2020168939A1/zh
Priority to IL285378A priority patent/IL285378A/en
Priority to CONC2021/0011023A priority patent/CO2021011023A2/es

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • 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
    • C07D487/08Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/504Pyridazines; Hydrogenated pyridazines forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • 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
    • C07D417/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/08Bridged systems

Definitions

  • the present invention relates to a new heterocyclic compound, a pharmaceutical composition containing the same, a preparation method thereof, and its use in the prevention or treatment of diseases or conditions related to RET (Rearranged during transfection) activity.
  • Protein kinases are a class of enzymes that catalyze protein phosphorylation reactions. By mediating the process of cell signal transduction, protein phosphorylation regulates the physiological activities of cells, such as cell survival, proliferation, differentiation, apoptosis and metabolism.
  • the dysfunction of protein kinases is closely related to many diseases, including tumors, autoimmune diseases, inflammatory reactions, central nervous system diseases, cardiovascular diseases and diabetes.
  • RET is a proto-oncogene.
  • the RET protein encoded by it is a transmembrane receptor tyrosine protein kinase. It consists of a cysteine-rich cadherin-like extracellular domain (used to bind ligands), The transmembrane region and the intracellular structural region with tyrosine kinase activity are composed of three parts.
  • the activated RET protein can activate multiple downstream signaling pathways, including the RAS/RAF/ERK pathway, PI3K/Akt pathway and JNK pathway, leading to cell proliferation, migration and differentiation.
  • RET gene changes have a more significant effect on the downstream cascade.
  • RET gene mutations are mainly related to medullary thyroid cancer and papillary thyroid cancer
  • RET gene fusion is mainly related to non-small cell lung cancer and chronic myeloid leukemia. Therefore, inhibiting RET activity has great medical value (Nature Reviews Cancer, 2014, 14(3): 173-86).
  • RET inhibitors have great potential to treat and prevent many diseases (such as tumors, irritable bowel syndrome, etc.). There are currently five compounds in the clinical trial stage, and compounds from many companies are in the pre-clinical research stage. However, no inhibitors with RET as the main target are currently on the market. Therefore, it is necessary to develop new, high-efficiency and low-toxic RET inhibitors to meet clinical needs.
  • the present invention provides a new heterocyclic compound, which has a good inhibitory effect on RET, and has good pharmacokinetics, safety and other properties.
  • One aspect of the present invention provides compounds of formula I, stereoisomers, tautomers or mixtures of said compounds, N-oxides of said compounds, pharmaceutically acceptable salts, co- Crystals, polymorphs or solvates, or stable isotope derivatives, metabolites or prodrugs of said compounds:
  • Ring A is selected from C 6-10 aromatic ring and 5-6 membered heteroaromatic ring;
  • Ring B is selected from C 3-8 cycloalkyl and 4-11 membered heterocyclic group
  • X 1 is selected from CH and N;
  • R 1 is selected from H, halogen, hydroxyl, cyano, C 1-6 alkyl, C 1-6 heteroalkyl (e.g. C 1-6 alkoxy), C 3-8 cycloalkyl, 4-10 member Heterocyclyl and -NR 20a R 20b , the alkyl, heteroalkyl (e.g. alkoxy), cycloalkyl and heterocyclyl are each optionally substituted with one or more substituents selected from the following: hydroxy , Halogen, CN, NO 2 , C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 hydroxyalkyl, C 1-4 haloalkoxy and C 1-4 heteroalkyl (e.g. C 1- 4 alkoxy);
  • R 3 and R 4 are absent or are each independently selected from hydroxyl, halogen, CN, C 1-6 alkyl, C 1-6 heteroalkyl (e.g. C 1-6 alkoxy) and C 3-6 cycloalkyl, each of said alkyl, heteroalkyl (eg alkoxy) and cycloalkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, CN, C1-4 Alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy; when m is greater than 1, two R 3 optionally together with the atoms to which they are attached form C 3-6 Cycloalkyl or 4-10 membered heterocyclic group; and/or when n is greater than 1, two R 4 optionally together with the atoms to which they are attached form a C 3-6 cycloalkyl group or 4-10 membered heterocyclic group ;
  • the alkylene, heteroalkylene, alkenylene and alkynylene groups are each optionally substituted with one or more substituents selected from the group consisting of hydroxyl, halogen, CN, NO 2 , C 1-6 alkyl , C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 haloalkoxy, C 1-6 heteroalkyl (e.g. C 1-6 alkoxy) and C 3-8 cycloalkyl;
  • L is -N(R 23a )-;
  • R 20a , R 20b , R 23a , R 23b , R 23c , R 24a , R 25a and R 25b are each independently selected from H, OH, C 1-6 alkyl, C 1-6 alkoxy and C 3- 8 cycloalkyl; or R 20a and R 20b , R 23a and R 23b or R 25a and R 25b together with the atoms to which they are connected form a 3-8 membered cycloalkyl or heterocyclic group, the alkyl, alkoxy Group, cycloalkyl and heterocyclic group are each optionally substituted with one or more substituents selected from the group consisting of OH, CN, halogen, NO 2 , C 1-4 alkyl, C 1-4 alkoxy, C 1-4 hydroxyalkyl, C 1-4 haloalkyl and C 1-4 haloalkoxy;
  • R 30a , R 30b , R 33a , R 33b , R 34a , R 35a and R 35b are each independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy and C 1-6 haloalkoxy;
  • R 21 , R 22 , R 31 and R 32 are each independently selected from C 1-6 alkyl, C 1-6 alkoxy, C 3-8 cycloalkyl, 4-10 membered heterocyclyl, C 6- 12 aryl groups and 5-10 membered heteroaryl groups, each of the alkyl group, alkoxy group, cycloalkyl group, heterocyclic group, aryl group and heteroaryl group is optionally substituted by one or more substituents selected from the following Substitution: OH, halogen, CN, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 haloalkyl, C 1-4 haloalkoxy, C 3-6 cycloalkyl and 4-10 members Heterocyclic group;
  • n 0, 1, 2, 3 or 4;
  • n 0, 1, 2, 3 or 4;
  • t 0, 1, 2, 3 or 4;
  • u 0, 1, 2, 3 or 4;
  • the proviso is that when ring B is a piperazine ring and X 1 is CH, R 2 is not 4-CF 3 -pyridin-2-yl or 4-CN-pyridin-2-yl.
  • compositions which comprises a preventive or therapeutically effective amount of a compound of the present invention, a stereoisomer, a tautomer of the compound, or a mixture thereof, and an N-oxide of the compound , A pharmaceutically acceptable salt, co-crystal, polymorph or solvate of the compound, or a stable isotope derivative, metabolite or prodrug of the compound.
  • the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers.
  • Another aspect of the present invention provides a compound of the present invention, a stereoisomer, a tautomer of the compound, or a mixture thereof, an N-oxide of the compound, a pharmaceutically acceptable salt of the compound, Co-crystals, polymorphs or solvates, or stable isotope derivatives, metabolites or prodrugs of the compounds, or pharmaceutical compositions as described above are being prepared for the prevention or treatment of diseases related to RET activity or Use in medicine for medical conditions.
  • Another aspect of the present invention provides a compound of the present invention, a stereoisomer, a tautomer of the compound, or a mixture thereof, an N-oxide of the compound, a pharmaceutically acceptable salt of the compound, Co-crystals, polymorphs or solvates, or stable isotope derivatives, metabolites or prodrugs of the compounds, or pharmaceutical compositions as described above, which are used for the prevention or treatment of diseases related to RET activity or Condition.
  • Another aspect of the present invention provides a method for preventing or treating diseases or conditions associated with RET activity, the method comprising administering to an individual in need thereof an effective amount of a compound of the present invention, stereoisomers, mutual Tautomers or mixtures thereof, N-oxides of the compounds, pharmaceutically acceptable salts, co-crystals, polymorphs or solvates of the compounds, or stable isotopic derivatives of the compounds, Metabolites or prodrugs, or pharmaceutical compositions as described above.
  • Another aspect of the invention provides a method of preparing the compound of the invention.
  • Figure 1 shows the in vivo efficacy test results of compound 17 and the control compound BLU-667 in a subcutaneous transplantation tumor model of medullary thyroid carcinoma TT cells.
  • alkyl is defined as a linear or branched saturated aliphatic hydrocarbon.
  • the alkyl group has 1 to 12, for example 1 to 6 carbon atoms.
  • C 1-6 alkyl and C 1-4 alkyl refer to linear or branched groups having 1-6 carbon atoms and 1-4 carbon atoms, respectively (Such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl or n-hexyl), any Optionally substituted by one or more (such as 1 to 3) suitable substituents such as halogen (in this case the group is referred to as "haloalkyl”) (e.g.
  • C 1-4 alkyl refers to a linear or branched aliphatic hydrocarbon chain of 1 to 4 carbon atoms (ie methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, Sec-butyl or tert-butyl).
  • alkylene means a corresponding divalent group, including, for example, “C 1-8 alkylene", “C 1-6 alkylene", “C 1-4 alkylene” and the like.
  • alkylene group is optionally substituted with one or more (such as 1 to 3) substituents which are the same or different.
  • heteroalkyl refers to an optionally substituted alkyl group having one or more backbone chain atoms selected from atoms other than carbon, such as oxygen, nitrogen, sulfur, phosphorus, or combinations thereof .
  • Numerical ranges that can be given e.g. C1-6 heteroalkyl refer to the number of carbons in the chain, including 1-6 carbon atoms in this example.
  • the -CH 2 OCH 2 CH 3 group is called a C 3 heteroalkyl group.
  • the connection to the rest of the molecule can be through heteroatoms or carbon atoms in the heteroalkyl chain.
  • heteroalkylene means a corresponding divalent group, including, for example, “C 1-6 heteroalkylene", “C 1-4 heteroalkylene” and the like.
  • haloalkyl refers to an alkyl group substituted with one or more (such as 1 to 3) identical or different halogen atoms
  • C 1-8 haloalkyl refers to haloalkyl groups having 1 to 8 carbon atoms, 1 to 6 carbon atoms and 1-4 carbon atoms, respectively, such as -CF 3 , -C 2 F 5 , -CHF 2 , -CH 2 F, -CH 2 CF 3 , -CH 2 Cl or -CH 2 CH 2 CF 3 and so on.
  • hydroxyalkyl refers to a group formed by replacing the hydrogen atom in an alkyl group with one or more hydroxy groups, such as C 1-4 hydroxyalkyl or C 1-3 hydroxyalkyl, Examples thereof include, but are not limited to, hydroxymethyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, -CH(OH)CH 3 , -C(CH 3 ) 2 OH and the like.
  • alkoxy means a group with an oxygen atom inserted at any reasonable position of an alkyl group (as defined above), preferably a C 1-8 alkoxy group, a C 1-6 alkoxy group Group, C 1-4 alkoxy or C 1-3 alkoxy.
  • C 1-6 alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy Group, tert-butoxy, pentyloxy, hexyloxy, -CH 2 -OCH 3, etc., the alkoxy is optionally substituted by one or more (such as 1 to 3) substituents which are the same or different .
  • alkyleneoxy refers to a divalent alkoxy group, such as -OCH 2 -, -OCH(CH 3 )CH 2 -, -OCH 2 CH 2 O-, -CH 2 CH 2 O- etc., the alkyleneoxy group is optionally substituted with one or more (such as 1 to 3) substituents which are the same or different.
  • alkenyl means a linear or branched monovalent hydrocarbon group, which contains one or more double bonds and has 2-6 carbon atoms (“C 2-6 alkenyl”).
  • alkynyl means a monovalent hydrocarbon group containing one or more triple bonds, which preferably has 2, 3, 4, 5 or 6 carbon atoms, such as ethynyl, 2-propynyl, 2 -Butynyl, 1,3-butadiynyl, etc.
  • the alkynyl group is optionally substituted with one or more (such as 1 to 3) substituents which are the same or different.
  • alkynylene is a corresponding divalent group, including, for example, “C 2-8 alkynylene", “C 2-6 alkynylene", “C 2-4 alkynylene” and the like. Examples include but are not limited to Etc., the alkynylene group is optionally substituted with one or more (such as 1 to 3) substituents which are the same or different.
  • fused ring or “fused ring” refers to a ring system formed by two or more ring structures sharing two adjacent atoms with each other.
  • spirocyclic ring refers to a ring system formed by two or more ring structures sharing one ring atom with each other.
  • bridged ring refers to a ring system formed by two or more ring structures sharing two atoms that are not directly connected to each other.
  • cycloalkyl refers to a saturated or unsaturated non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring group, including but not limited to monocyclic alkyl (such as cyclopropyl, cyclobutyl Group, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, etc.) and bicycloalkyl, including spiro ring, fused ring (fused ring) or bridged ring system (ie, spirocycloalkyl, and Cyclo (fused ring) alkyl groups and bridged cycloalkyl groups, such as bicyclo[1.1.1]pentyl, bicyclo[2.2.1]heptyl, etc.).
  • monocyclic alkyl such as cyclopropyl, cyclobutyl Group, cyclopentyl, cyclohexyl, cyclo
  • the cycloalkyl group is optionally substituted with one or more (such as 1 to 3) substituents which are the same or different.
  • C 3-8 cycloalkyl refers to a cycloalkyl group having 3 to 8 ring-forming carbon atoms, such as a C 3-6 cycloalkyl group, which may be a monocyclic alkyl group, such as cyclopropyl, cyclobutyl , Cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl, it can also be a bicyclic alkyl group, such as C 5-8 spirocycloalkyl, C 5-8 bridged cycloalkyl, C 5-8 fused cycloalkyl , C 5-6 spirocycloalkyl, C 5-6 bridged cycloalkyl or C 5-6 condensed cycloalkyl.
  • cycloalkoxy means -O-cycloalkyl, where cycloalkyl is as defined above.
  • Representative examples of cycloalkoxy include, but are not limited to, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy, and the like.
  • 4-11 membered heterocyclic group means a heterocyclic group containing 4-11 ring atoms, including but not limited to 4-10 membered heterocyclic group, 4-9 membered heterocyclic group, 4-8 membered heterocyclic group, 4-7 membered heterocyclic group, 5-6 membered heterocyclic group, 3-8 membered heterocyclic group, 3-7 membered heterocyclic group, 4-7 membered nitrogen-containing heterocyclic group, 4-7 membered oxygen-containing heterocyclic group, 4-7 membered sulfur-containing heterocyclic group, 5-6 membered nitrogen-containing heterocyclic group, 5-6 membered oxygen-containing heterocyclic group, 5-6 membered sulfur-containing heterocyclic group, etc.
  • Each of the "nitrogen-containing heterocyclic group", “oxygen-containing heterocyclic group” and “sulfur-containing heterocyclic group” optionally further contains one or more other heteroatoms selected from oxygen, nitrogen and sulfur.
  • 4-11 membered heterocyclic groups include, but are not limited to, oxirane, aziridinyl, azetidinyl, oxetanyl, tetrahydrofuryl, pyrrolidinyl, pyrrolidinone (e.g.
  • imidazolidinyl imidazolidinyl, pyrazolidinyl, tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, trithianyl.
  • heterocyclic group encompasses a fused ring structure, and the point of connection between the fused ring structure and other groups can be on any ring in the fused ring structure. Therefore, the heterocyclic group of the present invention also includes but is not limited to heterocyclic group and heterocyclic group, heterocyclic group and cycloalkyl group, single heterocyclic group and single heterocyclic group, single heterocyclic group and monocyclic alkyl group, for example 3-7 membered (mono) heterocyclyl and 3-7 membered (mono) heterocyclyl, 3-7 membered (mono) heterocyclyl and (mono) cycloalkyl, 3-7 membered (mono) heterocyclyl And C 4-6 (mono)cycloalkyl, examples of which include but are not limited to pyrrolidinocyclopropyl, cyclopentylazacyclopropyl, pyrrolidinocyclobutyl, pyrrolidin
  • heterocyclic group encompasses bridged heterocyclic groups and spiro heterocyclic groups.
  • bridged heterocycle refers to two saturated rings that share two ring atoms that are not directly connected and contain one or more (for example, 1, 2, 3, or 4) heteroatoms.
  • cyclic structure including but not limited to 7-10 membered heterocyclic ring, 8-10 membered heterocyclic ring, 7-10 membered nitrogen-containing bridged heterocyclic ring, 7- 10-membered oxygen-containing bridged heterocyclic ring, 7-10 membered sulfur-containing bridged heterocyclic ring, etc., for example Wait.
  • the "nitrogen-containing heterocyclic ring”, "oxygen-containing heterocyclic ring” and “sulfur-containing heterocyclic ring” optionally further contain one or more other heteroatoms selected from oxygen, nitrogen and sulfur.
  • spiro heterocyclic ring refers to two or more saturated rings that share one ring atom and contain one or more (for example, 1, 2, 3, or 4) heteroatoms.
  • oxygen atom, nitrogen atom, sulfur atom cyclic structure, including but not limited to 5-10 membered spiro heterocyclic ring, 6-10 membered spiro heterocyclic ring, 6-10 membered nitrogen-containing spiro heterocyclic ring, 6-10 membered Oxygen-containing spiro heterocycles, 6-10 membered sulfur-containing spiro heterocycles, etc., for example
  • the "nitrogen-containing spiro heterocyclic ring", “oxygen-containing spiro heterocyclic ring”, and “sulfur-containing spiro heterocyclic ring” optionally further contain one or more other heteroatoms selected from oxygen, nitrogen, and sulfur.
  • the term "6-10 membered nitrogen-containing spiroheterocyclic group" refers to two or more saturated
  • a heterocyclic group may be fused with an aryl group to form a fused ring structure.
  • the fused ring structure include but are not limited to:
  • aryl refers to an all-carbon monocyclic or fused polycyclic aromatic group having a conjugated ⁇ -electron system.
  • C 6-12 aryl (aromatic ring) means an aryl group (aromatic ring) containing 6 to 12 carbon atoms, preferably a C 6-10 aryl group (aromatic ring), preferably It is phenyl or naphthyl.
  • the aryl group is optionally substituted with one or more (such as 1 to 3) identical or different substituents (eg halogen, OH, CN, NO 2 , C 1 -C 6 alkyl, etc.).
  • heteroaryl or “heteroaromatic ring” refers to a monocyclic or polycyclic aromatic group containing one or more identical or different heteroatoms, including monocyclic heteroaryl and containing at least A bicyclic or polycyclic ring system of a heteroaromatic ring (an aromatic ring system containing at least one heteroatom), which can have 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 ring atoms, For example, 5, 6, 7, 8, 9 or 10 ring atoms.
  • the heteroatom may be oxygen, nitrogen or sulfur.
  • the term "5-10 membered heteroaryl” or “5-10 membered heteroaromatic ring” means a heteroaryl group containing 5 to 10 (e.g. 5 to 6) ring atoms (heteroaryl ring ), including 5-10 member nitrogen-containing heteroaryl group, 5-10 member oxygen-containing heteroaryl group, 5-10 member sulfur-containing heteroaryl group, 5-6 member nitrogen-containing heteroaryl group, 5-6 member oxygen-containing heteroaryl group Aryl, 5-6 membered sulfur-containing heteroaryl, etc.
  • the "nitrogen-containing heteroaryl group”, “oxygen-containing heteroaryl group” and “sulfur-containing heteroaryl group” each optionally contain one or more other heteroatoms selected from oxygen, nitrogen and sulfur.
  • Examples include, but are not limited to, thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, triazolyl, tetrazolyl, oxadiazolyl , Thiadiazolyl, etc., or pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, etc., and 5-10 membered cyclic groups containing these groups.
  • heteroaryl encompasses a fused ring structure, and the point of connection between the fused ring structure and other groups can be on any ring in the fused ring structure. Therefore, the heteroaryl group of the present invention also includes but is not limited to (mono)heteroaryl and (mono)heteroaryl, (mono)heteroaryl and (monocyclic)aryl, (mono)heteroaryl and (mono) ) Heterocyclyl and (mono)heteroaryl and (mono)cycloalkyl, for example 5-6 membered (mono)heteroaryl and 5-6 membered (mono)heteroaryl, 5-6 membered (mono)heteroaryl, 5-6 membered (mono)hetero Aryl phenyl, 5-6 membered (mono)heteroaryl and 5-6 membered (mono)heterocyclyl or 5-6 membered (mono)heteroaryl
  • halo or halogen group is defined to include F, Cl, Br, or I.
  • substituted means that one or more (eg, one, two, three, or four) hydrogens on the specified atom are replaced by a selection from the indicated group, provided that no more than the specified atom is present In the case of normal valence and the substitution forms a stable compound. Combinations of substituents and/or variables are only allowed when such combinations form stable compounds.
  • substituent can be (1) unsubstituted or (2) substituted. If the carbon of a substituent is described as being optionally substituted with one or more of the list of substituents, then one or more hydrogens on the carbon (to the extent of any hydrogens present) may be independently and/or together independently Optional substitution of selected substituents. If the nitrogen of a substituent is described as being optionally substituted with one or more of the list of substituents, then one or more hydrogens on the nitrogen (to the extent of any hydrogens present) may each be independently selected optionally Substituent replacement.
  • each substituent is selected independently of the other. Therefore, each substituent may be the same or different from another (other) substituent.
  • one or more means 1 or more than 1, such as 2, 3, 4, 5, or 10 under reasonable conditions.
  • the point of attachment of a substituent can be from any suitable position of the substituent.
  • the present invention also includes all pharmaceutically acceptable isotope-labeled compounds, which are the same as the compounds of the present invention, except that one or more atoms have the same atomic number but the atomic mass or mass number is different from the predominant atomic mass in nature. Or atomic substitution of mass number.
  • isotopes suitable for inclusion in the compounds of the present invention include, but are not limited to, isotopes of hydrogen (such as deuterium ( 2 H), tritium ( 3 H)); isotopes of carbon (such as 11 C, 13 C, and 14 C) ; Isotopes of chlorine (such as 36 Cl); isotopes of fluorine (such as 18 F); isotopes of iodine (such as 123 I and 125 I); isotopes of nitrogen (such as 13 N and 15 N); isotopes of oxygen (such as 15 O , 17 O and 18 O); phosphorus isotopes (such as 32 P); and sulfur isotopes (such as 35 S).
  • isotopes of hydrogen such as deuterium ( 2 H), tritium ( 3 H)
  • isotopes of carbon such as 11 C, 13 C, and 14 C
  • Isotopes of chlorine such as 36 Cl
  • isotopes of fluorine
  • Certain isotope-labeled compounds of the invention can be used in drug and/or substrate tissue distribution studies (such as analysis).
  • the radioisotopes tritium (i.e. 3 H) and carbon-14 (i.e. 14 C) are particularly useful for this purpose because they are easy to incorporate and easy to detect.
  • Substitution with positron emission isotopes (such as 11 C, 18 F, 15 O, and 13 N) can be used to test substrate receptor occupancy in positron emission tomography (PET) studies.
  • the isotopically-labeled compounds of the present invention can be prepared by methods similar to those described in the attached routes and/or examples and preparations by using appropriate isotopically-labeled reagents instead of previously used non-labeled reagents.
  • the pharmaceutically acceptable solvates of the present invention include those in which the crystallization solvent can be replaced by an isotope, for example, D 2 O, acetone-d 6 or DMSO-d 6 .
  • stereoisomer means an isomer due to at least one asymmetric center. In compounds with one or more (for example, one, two, three or four) asymmetric centers, it can produce exo/meso mixtures, single enantiomers, and diastereoisomer mixtures And individual diastereomers. Certain individual molecules can also exist as geometric isomers (cis/trans). Similarly, the compounds of the present invention may exist in mixtures of two or more structurally different forms (commonly referred to as tautomers) in rapid equilibrium. Representative examples of tautomers include keto-enol tautomers, phenol-ketone tautomers, nitroso-oxime tautomers, imine-enamine tautomers Wait. For example, nitroso-oximes can exist in equilibrium in the following tautomeric forms in solution:
  • Solid lines can be used in this article Solid wedge Virtual wedge Depicts the chemical bonds of the compounds of the invention.
  • the use of a solid line to depict the bond to an asymmetric carbon atom is intended to indicate that all possible stereoisomers at that carbon atom (e.g., specific enantiomers, racemic mixtures, etc.) are included.
  • the use of real or imaginary wedges to depict bonds to asymmetric carbon atoms is intended to indicate that the stereoisomers shown exist. When present in a racemic mixture, use real and imaginary wedges to define relative stereochemistry, rather than absolute stereochemistry.
  • the compounds of the present invention are intended to be stereoisomers (which include cis and trans isomers, optical isomers (such as R and S enantiomers), diastereomers, Geometric isomers, rotamers, conformational isomers, atropisomers and mixtures thereof) exist in the form of.
  • the compounds of the present invention can exhibit more than one type of isomerism, and are composed of mixtures thereof (for example, racemic mixtures and diastereomeric pairs).
  • the present invention covers all possible crystalline forms or polymorphs of the compounds of the present invention, which can be a single polymorph or a mixture of more than one polymorph in any ratio.
  • Eutectic refers to the fact that the active molecules of the drug and other physiologically acceptable molecules of acids, bases, salts, and non-ionic compounds are connected by hydrogen bonds, ⁇ - ⁇ stacking, van der Waals forces and other non-covalent bonds to be combined in the same crystal lattice. .
  • compositions of the present invention may exist in free form for treatment, or, when appropriate, in the form of pharmaceutically acceptable derivatives thereof.
  • pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable salts, esters, solvates, N-oxides, metabolites, or prodrugs, which can be administered to patients in need thereof. After medication, the compound of the present invention or its metabolites or residues can be directly or indirectly provided. Therefore, when “the compound of the present invention” is referred to herein, it is also intended to cover the above-mentioned various derivative forms of the compound.
  • the pharmaceutically acceptable salts of the compounds of the present invention include acid addition salts and base addition salts thereof.
  • acid addition salts for example, hexafluorophosphate, meglumine salt, etc.
  • suitable salts see “Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by Stahl and Wermuth (Wiley-VCH, 2002).
  • ester means an ester derived from each of the compounds of the general formula in this application, which includes physiologically hydrolyzable esters (which can be hydrolyzed under physiological conditions to release the free acid or alcohol form of the present invention Compound).
  • the compound of the present invention may itself be an ester.
  • the compound of the present invention may exist in the form of a solvate (preferably a hydrate), wherein the compound of the present invention contains a polar solvent as a structural element of the crystal lattice of the compound, particularly, for example, water, methanol or ethanol.
  • a polar solvent as a structural element of the crystal lattice of the compound, particularly, for example, water, methanol or ethanol.
  • the amount of polar solvent, especially water can be present in a stoichiometric or non-stoichiometric ratio.
  • metabolites of the compounds of the present invention that is, substances formed in the body when the compounds of the present invention are administered. Such products can be produced by, for example, oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymatic hydrolysis, etc. of the administered compound. Therefore, the present invention includes metabolites of the compounds of the present invention, including compounds prepared by contacting the compound of the present invention with a mammal for a time sufficient to produce its metabolites.
  • the present invention further includes within its scope the prodrugs of the compounds of the present invention, which are certain derivatives of the compounds of the present invention that may themselves have less or no pharmacological activity when administered to or on the body It can be converted into the compound of the present invention having the desired activity by, for example, hydrolytic cleavage.
  • prodrugs will be functional group derivatives of the compound, which are easily converted into the desired therapeutically active compound in vivo.
  • prodrugs please refer to "Pro-drugs as Novel Delivery Systems", Volume 14, ACS Symposium Series (T. Higuchi and V. Stella).
  • prodrugs of the present invention can be used, for example, by using certain parts known to those skilled in the art as “pro-moiety (e.g., “Design of Prodrugs", described in H. Bundgaard (Elsevier, 1985))" It is prepared by substituting appropriate functional groups present in the compounds of the present invention.
  • the present invention also encompasses compounds of the present invention containing protecting groups.
  • protecting groups In any process of preparing the compounds of the present invention, protection of sensitive groups or reactive groups on any relevant molecule may be necessary and/or desirable, thereby forming a chemically protected form of the compounds of the present invention. This can be achieved by conventional protecting groups, such as those described in T.W. Greene & P. G. M. Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991, these references are incorporated herein by reference. Using methods known in the art, the protecting group can be removed at an appropriate subsequent stage.
  • One aspect of the present invention provides compounds of formula I, stereoisomers, tautomers or mixtures of said compounds, N-oxides of said compounds, pharmaceutically acceptable salts, co- Crystals, polymorphs or solvates, or stable isotope derivatives, metabolites or prodrugs of said compounds:
  • Ring A is selected from C 6-10 aromatic ring and 5-6 membered heteroaromatic ring;
  • Ring B is selected from C 3-8 cycloalkyl and 4-11 membered heterocyclic group
  • X 1 is selected from CH and N;
  • R 1 is selected from H, halogen, hydroxyl, cyano, C 1-6 alkyl, C 1-6 heteroalkyl (e.g. C 1-6 alkoxy), C 3-8 cycloalkyl, 4-10 member Heterocyclyl and -NR 20a R 20b , the alkyl, heteroalkyl (e.g. alkoxy), cycloalkyl and heterocyclyl are each optionally substituted with one or more substituents selected from the following: hydroxy , Halogen, CN, NO 2 , C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 hydroxyalkyl, C 1-4 haloalkoxy and C 1-4 heteroalkyl (e.g. C 1- 4 alkoxy);
  • R 3 and R 4 are absent or are each independently selected from hydroxyl, halogen, CN, C 1-6 alkyl, C 1-6 heteroalkyl (e.g. C 1-6 alkoxy) and C 3-6 cycloalkyl, each of said alkyl, heteroalkyl (eg alkoxy) and cycloalkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, CN, C1-4 Alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy; when m is greater than 1, two R 3 optionally together with the atoms to which they are attached form C 3-6 Cycloalkyl or 4-10 membered heterocyclic group; and/or when n is greater than 1, two R 4 optionally together with the atoms to which they are attached form a C 3-6 cycloalkyl group or 4-10 membered heterocyclic group ;
  • the alkylene, heteroalkylene, alkenylene and alkynylene groups are each optionally substituted with one or more substituents selected from the group consisting of hydroxyl, halogen, CN, NO 2 , C 1-6 alkyl , C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 haloalkoxy, C 1-6 heteroalkyl (e.g. C 1-6 alkoxy) and C 3-8 cycloalkyl;
  • L is -N(R 23a )-;
  • R 20a , R 20b , R 23a , R 23b , R 23c , R 24a , R 25a and R 25b are each independently selected from H, OH, C 1-6 alkyl, C 1-6 alkoxy and C 3- 8 cycloalkyl; or R 20a and R 20b , R 23a and R 23b or R 25a and R 25b together with the atoms to which they are connected form a 3-8 membered cycloalkyl or heterocyclic group, the alkyl, alkoxy Group, cycloalkyl and heterocyclic group are each optionally substituted with one or more substituents selected from the group consisting of OH, CN, halogen, NO 2 , C 1-4 alkyl, C 1-4 alkoxy, C 1-4 hydroxyalkyl, C 1-4 haloalkyl and C 1-4 haloalkoxy;
  • R 30a , R 30b , R 33a , R 33b , R 34a , R 35a and R 35b are each independently selected from H, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy and C 1-6 haloalkoxy;
  • R 21 , R 22 , R 31 and R 32 are each independently selected from C 1-6 alkyl, C 1-6 alkoxy, C 3-8 cycloalkyl, 4-10 membered heterocyclyl, C 6- 12 aryl groups and 5-10 membered heteroaryl groups, each of the alkyl group, alkoxy group, cycloalkyl group, heterocyclic group, aryl group and heteroaryl group is optionally substituted by one or more substituents selected from the following Substitution: OH, halogen, CN, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 haloalkyl, C 1-4 haloalkoxy, C 3-6 cycloalkyl and 4-10 members Heterocyclic group;
  • n 0, 1, 2, 3 or 4;
  • n 0, 1, 2, 3 or 4;
  • t 0, 1, 2, 3 or 4;
  • u 0, 1, 2, 3 or 4;
  • the proviso is that when ring B is a piperazine ring and X 1 is CH, R 2 is not 4-CF 3 -pyridin-2-yl or 4-CN-pyridin-2-yl.
  • ring A is a benzene ring or a 5-6 membered heteroaromatic ring; preferably, ring A is a benzene ring, a thiazole ring, a pyridine ring, a pyrazine ring or a pyrimidine ring; more preferably, ring A is It is connected to the ring where X 1 is located through the position marked with *, and is connected to ring B through the position marked with **.
  • ring B is C 3-6 cycloalkyl or 5-7 membered heterocyclyl; preferably, ring B is piperidine ring, piperazine ring, azepane bridged ring or diazide A heterocycloheptane bridged ring; more preferably, ring B is It is connected to ring A by the position marked with *, and connected to L by the position marked by **.
  • X 1 is CH or N; preferably, X 1 is N.
  • R 1 is selected from H, halogen, hydroxy, cyano, C 1-4 alkyl, C 1-4 heteroalkyl (e.g. C 1-4 alkoxy), C 3-6 ring Alkyl and 4-10 membered heterocyclyl, each of said alkyl, heteroalkyl (e.g. alkoxy), cycloalkyl and heterocyclyl is optionally substituted with one or more substituents selected from the following: Hydroxyl, halogen, CN, NO 2 , C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 hydroxyalkyl, C 1-4 haloalkoxy and C 1-4 heteroalkyl (e.g. C 1 -4 alkoxy).
  • R 1 is selected from C 1-4 alkyl, 5-membered nitrogen-containing heterocyclic group and C 1-4 heteroalkyl (eg C 1-4 alkoxy), the alkyl, hetero
  • the cyclic group and the heteroalkyl group are each optionally substituted with one or more substituents selected from the group consisting of hydroxy, halogen, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 hydroxyalkyl, C 1-3 haloalkoxy, and C 1-3 heteroalkyl (e.g. C 1-4 alkoxy).
  • R 1 is selected from C 1-3 alkyl (e.g. methyl), pyrrolidinyl (e.g. pyrrolidin-1-yl), and C 1-3 alkoxy (e.g. ethoxy).
  • R 2 is methyl-substituted pyrazolyl (e.g. 5-methyl-1H-pyrazol-3-yl or 1-methyl-1H-pyrazol-4-yl), cyclopropyl-substituted pyrazolyl (e.g. 5-cyclopropyl-1H-pyrazol-3-yl) or -C(O)CH 3 .
  • R 3 and R 4 are absent or are independently selected from hydroxyl, halogen, CN, C 1-4 alkyl, and C 1-4 alkoxy at each occurrence, the alkyl and The alkoxy groups are each optionally substituted with one or more substituents selected from the group consisting of halogen, CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy and C 1-4 Haloalkoxy; when m is greater than 1, two R 3 optionally together with the atoms to which they are attached form a C 3-6 cycloalkyl or 4-10 membered heterocyclic group; and/or when n is greater than 1, two Each R 4 optionally together with the atom to which it is attached forms a C 3-6 cycloalkyl group or a 4-10 membered heterocyclic group.
  • R 3 and R 4 are absent or are independently selected from hydroxyl, halogen, CN, C 1-3 alkyl, and C 1-3 alkoxy at each occurrence, said alkyl and Each alkoxy group is optionally substituted with one or more substituents selected from the group consisting of halogen, CN and C 1-3 alkyl; when m is greater than 1, two R 3 are optionally together with the atoms to which they are attached A C 3-6 cycloalkyl group or a 4-10 membered heterocyclic group is formed; and/or when n is greater than 1, two R 4 optionally together with the atoms to which they are attached form a C 3-6 cycloalkyl group or a 4- 10-membered heterocyclic group.
  • R 3 and R 4 are absent or are independently selected from: F, Cl, CN, OH, C 1-3 alkyl, and C 1-3 alkoxy at each occurrence; preferably , R 3 and R 4 do not exist.
  • L is selected from -O-, -S-, -C(O)-, -N(R 23a )-C(O)-, -C(O)-N(R 23c )- , C 1-4 alkylene, C 1- 4 alkylene heteroalkyl
  • the alkylene and heteroalkylene groups are each optionally substituted with one or more substituents selected from the group consisting of hydroxyl, halogen, CN, NO 2 , C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 hydroxyalkyl, C 1-4 haloalkoxy, C 1-4 heteroalkyl (e.g. C 1-4 alkoxy) and C 3-6 cycloalkyl.
  • L is selected from -O-, -C(O)-, -NHC(O)-, -C(O)NH-, C 1-3 alkylene, C 1-3 hetero alkyl
  • the alkylene and heteroalkylene groups are each optionally substituted with one or more substituents selected from the group consisting of hydroxy, halogen, CN, NO 2 , C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 hydroxyalkyl, C 1-3 haloalkoxy, C 1-3 heteroalkyl (e.g. C 1-3 alkoxy) and C 3-6 cycloalkyl, wherein R 23a and R 23b are preferably H or C 1-3 alkyl.
  • L is selected from -O-, -C(O)-, -NHC(O)-, -C(O)NH-, C 1-3 alkylene,
  • the alkylene group is optionally substituted with one or more substituents selected from the group consisting of hydroxy, halogen, CN, C 1-3 alkyl, and C 1-3 haloalkyl.
  • L is -CH 2 -, -CH(CH 3 )-, -O-, -C(O)-, -C(O)NH-or
  • R 5 is selected from C 3-6 cycloalkyl, 4-10 membered heterocyclyl, C 6-12 aryl, and 5-10 membered heteroaryl, the cycloalkyl, heterocyclic Group, aryl group, and heteroaryl group are each optionally substituted with one or more substituents selected from the group consisting of hydroxyl, halogen, CN, NO 2 , C 1-4 alkyl, C 1-4 haloalkyl, C 1 -4 hydroxyalkyl, C 1-4 haloalkoxy, C 1-4 heteroalkyl (e.g.
  • R 5 is selected from phenyl and 5-6 membered heteroaryl (e.g., pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrazolyl, oxazolyl, imidazolyl or thiazolyl ), the phenyl and heteroaryl groups are each optionally substituted by one or more substituents selected from the group consisting of hydroxy, halogen, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1- 3 hydroxyalkyl, C 1-3 haloalkoxy, C 1-3 heteroalkyl (e.g., phenyl and 5-6 membered heteroaryl (e.g., pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyrazolyl, oxazolyl, imidazolyl or thiazolyl ), the phenyl and heteroaryl groups
  • the heterocyclic group and the heteroaryl group are each optionally substituted with one or more substituents selected from the group consisting of hydroxy, halogen, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 hydroxyalkane Group, C 1-3 haloalk
  • R 5 is selected from the group consisting of phenyl, pyridyl, pyrazolyl and thiazolyl, each of which is optionally selected by one or more of the following Substituent substitution of: halogen, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 hydroxyalkyl, C 1-3 haloalkoxy, C 1-3 alkoxy, C 3- 6 cycloalkyl, C 3-6 cycloalkoxy, 4-6 membered heterocyclic group, 5-8 membered heteroaryl (e.g.
  • heterocyclic group and heteroaryl group are each optionally substituted with one or more substituents selected from the following: Halogen, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 hydroxyalkyl, C 1-3 haloalkoxy, C 1-3 alkoxy, C 3-6 cycloalkyl, C 3-6 cycloalkoxy and 4-6 membered heterocyclic group.
  • R 5 is optionally substituted with one or more substituents selected from halogen (e.g. fluorine or chlorine), CN, C 1-3 alkyl (e.g. methyl or ethyl), C 1- 3 haloalkyl (e.g. three Fluoromethyl), C 1-3 alkoxy (e.g. methoxy or ethoxy), C 3-6 cycloalkyl (e.g. cyclopropyl), C 3-6 cycloalkoxy (e.g. cyclopropoxy Group) and 5-6 membered heteroaryl (e.g.
  • halogen e.g. fluorine or chlorine
  • CN C 1-3 alkyl (e.g. methyl or ethyl), C 1- 3 haloalkyl (e.g. three Fluoromethyl), C 1-3 alkoxy (e.g. methoxy or ethoxy), C 3-6 cycloalkyl (e.g. cyclopropyl), C 3-6 cycl
  • R 5 is selected from the group consisting of phenyl, pyridyl, pyrazolyl and thiazolyl, each of which is optionally selected by one or more of the following Substituent substitution of: halogen, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 hydroxyalkyl, C 1-3 haloalkoxy, C 1-3 alkoxy, C 3- 6 cycloalkyl, C 3-6 cycloalkoxy, 4-6 membered heterocyclic group, 5-6 membered heteroaryl (e.g.
  • heterocyclic group and heteroaryl group are each optionally substituted with one or more substituents selected from the group consisting of halogen, CN, C 1-3 Alkyl, C 1-3 haloalkyl, C 1-3 haloalkoxy, C 1-3 alkoxy, C 3-6 cycloalkyl, and 4-6 membered heterocyclic group.
  • R 5 is optionally selected from halogen (e.g. fluorine or chlorine), CN, C 1-3 alkyl (e.g.
  • methyl or ethyl C 1-3 haloalkyl (e.g. tri Fluoromethyl), C 1-3 alkoxy (e.g. methoxy or ethoxy), C 3-6 cycloalkyl (e.g. cyclopropyl), C 3-6 cycloalkoxy (e.g. cyclopropoxy Phenyl, pyridyl, pyrazolyl or thiazolyl substituted with substituents of five-membered heteroaryl (e.g. pyrazolyl, imidazolyl or thiazolyl), wherein the five-membered heteroaryl is optionally further It is substituted with one or more substituents selected from halogen (e.g. fluorine or chlorine), C 1-3 alkyl (e.g. methyl), C 1-3 hydroxyalkyl (e.g. hydroxymethyl or hydroxypropyl).
  • halogen e.g. fluorine or chlorine
  • C 1-3 alkyl e.
  • R 20a , R 20b , R 23a , R 23b , R 23c , R 24a , R 25a and R 25b are each independently selected from H, C 1-4 alkyl, C 1-4 alkoxy group and a C 3-8 cycloalkyl group; or R 20a and R 20b, R 23a and R 23b, or R 25a and R 25b together with the atoms to which they are attached form a 3-8 membered cycloalkyl or heterocyclyl, said Alkyl, alkoxy, cycloalkyl and heterocyclyl are each optionally substituted with one or more substituents selected from the group consisting of OH, CN, halogen, NO 2 , C 1-4 alkyl, C 1- 4 Alkoxy, C 1-4 hydroxyalkyl, C 1-4 haloalkyl and C 1-4 haloalkoxy.
  • R 20a , R 20b , R 23a , R 23b , R 23c , R 24a , R 25a and R 25b are each independently H, C 1-4 alkyl, or C 1-4 alkoxy .
  • R 23a and R 23b are each independently selected from H, C 1-3 alkyl, C 1-3 alkoxy, and C 3-6 cycloalkyl; or R 23a and R 23b together with them
  • R 21 , R 22 , R 31 and R 32 are each independently selected from C 1-4 alkyl, C 1-4 alkoxy, C 3-8 cycloalkyl, and 4-10 membered Heterocyclyl, the alkyl, alkoxy, cycloalkyl and heterocyclyl are each optionally substituted with one or more substituents selected from the group consisting of OH, halogen, CN, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 haloalkyl, C 1-4 haloalkoxy, C 3-6 cycloalkyl and 4-10 membered heterocyclic group.
  • R 21 , R 22 , R 31 and R 32 are each independently selected from C 1-4 alkyl.
  • R 30a, R 30b, R 33a, R 33b, R 34a, R 35a and R 35b are each independently selected from H, C 1-4 alkyl, C 1- 4 haloalkyl -alkyl, C 1 -4 hydroxyalkyl, C 1-4 alkoxy and C 1-4 haloalkoxy;
  • R 30a , R 30b , R 33a , R 33b , R 34a , R 35a and R 35b are each independently selected from H and C 1-4 alkyl.
  • n is zero.
  • n 0, 1, or 2.
  • t is 0 or 1.
  • u is 0 or 1.
  • the compound of the invention has the structure shown in Formula I-A:
  • R 1 and R 2 are as defined in formula I above;
  • R 23a , R 30a , R 30b , R 31 , R 32 , R 33a , R 33b , R 34a , R 35a and R 35b are as defined in Formula I above, and R 23a is preferably H or C 1-3 alkyl.
  • the 5-10 membered heteroaryl group is optionally substituted by one or more substituents selected from the group consisting of hydroxyl, halogen, CN, NO 2 , C 1-4 alkyl, C 1-4 Haloalkyl, C 1-4 hydroxyalkyl, C 1-4 haloalkoxy, C 1-4 heteroalkyl (e.g.
  • R 23a , R 30a , R 30b , R 31 , R 32 , R 33a , R 33b , R 34a , R 35a and R 35b are as defined in Formula I above, and R 23a is preferably H or C 1-3 alkyl.
  • the compound of the present invention has the structure shown in Formula I-B:
  • R 1 , R 2 , R 5 and R 23a are as defined in Formula I above, and R 23a is preferably H or C 1-3 alkyl.
  • the compound of the present invention has the structure shown in Formula I-C:
  • R 1 , R 2 , R 5 and R 23a are as defined in Formula I above, and R 23a is preferably H or C 1-3 alkyl; and when X 1 is N, R 1 , R 2 , R 5 and R 23a are as defined in Formula IA above.
  • the compound of the present invention has the structure shown in Formula I-D:
  • R 1 , R 2 , R 23a , R 23b and t are as defined in formula I above;
  • R 5 is as defined in Formula I above;
  • R 5 is C 6-12 aryl or 5-10 membered heteroaryl, wherein
  • the C 6-12 aryl group and 5-10 membered heteroaryl group are each optionally substituted with one or more substituents selected from the group consisting of hydroxyl, halogen, CN, NO 2 , C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 hydroxyalkyl, C 1-4 haloalkoxy, C 1-4 heteroalkyl (e.g.
  • the C 6-12 aryl group is optionally substituted with one or more substituents selected from the group consisting of hydroxyl, halogen, CN, NO 2 , C 1-4 alkane Group, C 1-4 haloalkyl, C 1-4 hydroxyalkyl, C 1-4 haloalkoxy, C 1-4 heteroalkyl (e.g.
  • R 30a , R 30b , R 31 , R 32 , R 33a , R 33b , R 34a , R 35a and R 35b are as defined in Formula I above.
  • the compound of the present invention has the structure shown in Formula I-E:
  • R 1 , R 2 , R 5 , R 23a , R 23b , X 1 and t are as defined in formula ID above.
  • the compounds of the invention have the structure shown in Formula I-F:
  • R 1 , R 2 , R 5 , R 23a , R 23b and X 1 are as defined in formula ID above;
  • R 4 is as defined in Formula I above, and is preferably C 1-3 alkyl or C 1-3 alkoxy;
  • R 23c is H, C 1-3 alkyl or C 1-3 alkoxy, each of said alkyl and alkoxy is optionally substituted by one or more substituents selected from the group consisting of OH, CN, halogen , C 1-4 alkoxy and C 1-4 hydroxyalkyl;
  • u is 0 or 1
  • n 0 or 1.
  • the compound of the present invention has the structure shown in Formula I-G:
  • X 1 is CH or N
  • R 1 , R 2 and R 4 are as defined in formula I above, and R 4 is preferably C 1-3 alkyl or C 1-3 alkoxy;
  • n 0 or 1
  • R 30a , R 30b , R 31 , R 32 , R 33a , R 33b , R 34a , R 35a and R 35b are as defined in Formula I above.
  • the compound of the present invention has the structure shown in Formula I, wherein:
  • Ring A is It is connected to the ring where X 1 is located through the position marked by *, and is connected to ring B through the position marked by **;
  • Ring B is It is connected to ring A through the position marked by *, and connected to L through the position marked by **;
  • X 1 is N
  • R 1 is selected from C 1-3 alkyl (e.g. methyl), pyrrolidinyl (e.g. pyrrolidin-1-yl) and C 1-3 alkoxy (e.g. ethoxy);
  • R 2 is a methyl-substituted pyrazolyl (e.g. 5-methyl-1H-pyrazol-3-yl or 1-methyl-1H-pyrazol-4-yl), a cyclopropyl-substituted pyrazolyl ( For example, 5-cyclopropyl-1H-pyrazol-3-yl) or -C(O)CH 3 ;
  • R 3 and R 4 do not exist
  • L is -CH 2 -, -CH(CH 3 )-, -O-, -C(O)-, -C(O)NH-or
  • R 5 is optionally substituted by one or more selected from halogen (e.g. fluorine or chlorine), CN, C 1-3 alkyl (e.g. methyl or ethyl), C 1-3 haloalkyl (e.g. trifluoromethyl) ), C 1-3 alkoxy (e.g. methoxy or ethoxy), C 3-6 cycloalkyl (e.g. cyclopropyl), C 3-6 cycloalkoxy (e.g.
  • halogen e.g. fluorine or chlorine
  • CN C 1-3 alkyl (e.g. methyl or ethyl), C 1-3 haloalkyl (e.g. trifluoromethyl) ), C 1-3 alkoxy (e.g. methoxy or ethoxy), C 3-6 cycloalkyl (e.g. cyclopropyl), C 3-6 cycloalkoxy (e.g.
  • 5-6 membered heteroaryl for example pyridyl, pyrrolyl, pyrazolyl, furyl, oxazolyl, imidazolyl or thiazolyl
  • the 5-6 membered heteroaryl group is optionally further substituted by one or more selected from halogen (e.g. fluorine or chlorine), C 1-3 alkyl (e.g. methyl, ethyl or isopropyl), C 1 -3 haloalkyl (e.g. fluoromethyl), C 1-3 hydroxyalkyl (e.g.
  • the present invention covers any combination of the above embodiments.
  • the compounds of the invention include, but are not limited to:
  • the compound of formula I-A can be synthesized by the method shown in Route A as follows:
  • Hal 1 and Hal 2 are each independently F, Cl, Br or I; preferably, Hal 1 is F, Cl, Br or I, and Hal 2 is Cl, Br or I;
  • R 1 is selected from H, cyano, C 1-6 alkyl, C 1-6 heteroalkyl (e.g. C 1-6 alkoxy), C 3-8 cycloalkyl, 4-6 membered heterocyclyl and -NR 20a R 20b , the alkyl, heteroalkyl (for example, alkoxy), cycloalkyl and heterocyclic groups are each optionally substituted with one or more substituents selected from the group consisting of halogen, CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 haloalkoxy and C 1-4 heteroalkyl (e.g. C 1-4 alkoxy);
  • R 2 is selected from the group consisting of C 1-6 alkyl, C 1-6 heteroalkyl, C 3-8 cycloalkyl, 4-6 membered heterocyclyl and 5-6 membered heteroaryl, said alkyl, heteroalkyl Group, cycloalkyl, heterocyclyl and heteroaryl are each optionally substituted with one or more substituents selected from the group consisting of hydroxy, halogen, CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 hydroxyalkyl, C 1-4 haloalkoxy, C 1-4 heteroalkyl and C 3-6 cycloalkyl;
  • R 23a is selected from H, C 1-6 alkyl, C 1-6 alkoxy and C 3-8 cycloalkyl, each of which is optionally substituted by one or more Substitution selected from the following substituents: OH, CN, halogen, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 hydroxyalkyl, C 1-4 haloalkyl, and C 1-4 haloalkoxy base;
  • R 20a and R 20b are as defined in Formula I above;
  • R 5 is as defined in Formula IA above.
  • the first step Compound IA-1 and R 2 -NH 2 undergo a substitution or coupling reaction (such as Buchwald reaction, Suzuki reaction or Ullmann reaction) in the presence of a base to produce compound IA-2.
  • a substitution or coupling reaction such as Buchwald reaction, Suzuki reaction or Ullmann reaction
  • usable bases are, for example, t BuONa, t BuOK, t BuOLi, Cs 2 CO 3 , DIPEA, LiHMDS, LDA, NaHMDS, KHMDS, K 3 PO 4 , Na 2 CO 3 , KOAc, NaHCO 3 or K 2 CO 3
  • usable solvents are, for example, tert-butanol, toluene, xylene, THF, DME, 1,4-dioxane, DMF, DMSO or NMP, and the reaction temperature is 40°C to 140°C.
  • usable catalysts are, for example, Pd(OAc) 2 , Pd 2 (dba) 3 , Pd(dba) 2 , PdCl 2 , Pd(PPh 3 ) 4 , Pd(dppf)Cl 2 , Pd(acac) 2 or Pd(allyl) 2
  • the usable ligands are PPh 3 , XPhos, SPhos, RuPhos, XantPhos, Dppf, BINOL, BINAP or Pcy 3, etc.
  • the usable bases are for example t BuONa, t BuOK, t BuOLi, Cs 2 CO 3 , LiHMDS, LDA, NaHMDS, KHMDS, K 3 PO 4 , Na 2 CO 3 , KOAc, NaHCO 3 or K 2 CO 3
  • usable solvents are for example toluene, xylene, THF, DME, 1, 4-Dioxane, DMF, DMSO or NMP, and the reaction temperature is 40°C to 140°C.
  • the usable catalyst is, for example, Pd(PPh 3 ) 4 or Pd(dppf)Cl 2
  • the usable base is, for example, Cs 2 CO 3 , K 3 PO 4 , Na 2 CO 3 , AcOK, NaHCO 3 or K 2 CO 3
  • the usable solvent is, for example, 1,4-dioxane/H 2 O, DMF/H 2 O, DMSO/H 2 O or CH 3 CN/H 2 O, and the reaction temperature is 60° C. To 120°C;
  • the catalyst that can be used is, for example, CuCl, CuBr, CuI, or Cu 2 O
  • the ligand that can be used is, for example, salicylaldoxime, cyclohexanediamine, N,N'-dimethylethylenediamine, TMEDA or ethyl Diamine
  • usable bases are, for example, t BuONa, t BuOK, t BuOLi, Cs 2 CO 3 , LiHMDS, LDA, NaHMDS, KHMDS, K 3 PO 4 , Na 2 CO 3 , KOAc, NaHCO 3 or K 2 CO 3
  • the usable solvent is, for example, toluene, xylene, THF, DME, 1,4-dioxane, DMF, DMSO or NMP, and the reaction temperature is 40°C to 140°C.
  • Usable bases are, for example, t BuONa, t BuOK, t BuOLi, Cs 2 CO 3 , DIPEA, LiHMDS, LDA, NaHMDS, KHMDS, K 3 PO 4 , Na 2 CO 3 , KOAc, NaHCO 3 or K 2 CO 3 .
  • Usable solvents are, for example, tert-butanol, toluene, xylene, THF, DME, 1,4-dioxane, DMF, DMSO or NMP.
  • the reaction temperature is 40°C to 140°C.
  • the third step Compound I-A-5 reacts with boron-containing reagent to produce compound I-A-6.
  • the boron-containing reagent that can be used is, for example, B 2 (pin) 2 .
  • Usable catalysts are, for example, Pd(PPh 3 ) 4 , Pd(dppf)Cl 2 or Pd(dppf) 2 Cl 2 ⁇ DCM.
  • Usable bases are, for example, Cs 2 CO 3 , K 3 PO 4 , Na 2 CO 3 , KOAc, NaHCO 3 or K 2 CO 3.
  • Usable solvents are, for example, 1,4-dioxane, DMF, DMSO, or CH 3 CN.
  • the reaction temperature is 50°C to 120°C.
  • the fourth step Compound I-A-2 and I-A-6 undergo a coupling reaction (such as Suzuki reaction) to produce compound I-A-7.
  • a coupling reaction such as Suzuki reaction
  • Usable catalysts are, for example, Pd(PPh 3 ) 4 , Pd(dppf)Cl 2 or Pd(dppf) 2 Cl 2 ⁇ DCM.
  • Usable bases are, for example, Cs 2 CO 3 , K 3 PO 4 , Na 2 CO 3 , KOAc, NaHCO 3 or K 2 CO 3 .
  • the usable solvent is, for example, 1,4-dioxane, DMF, DMSO, or CH 3 CN, or a mixture of any of the above solvents and H 2 O.
  • the reaction temperature is 50°C to 120°C.
  • Step 5 Compound I-A-7 is deprotected under acidic conditions to produce compound I-A-8.
  • Usable acids are, for example, a 1,4-dioxane solution of HCl, an EA solution of HCl, or a DCM solution of TFA.
  • the reaction temperature is 0°C to 80°C.
  • Step 6 Compound I-A-8 and I-A-9 undergo reductive amination reaction to produce compound I-A.
  • the usable solvent is, for example, methanol, ethanol, THF, DCM, DCE, DMA, or a mixture of them and acetic acid in any ratio.
  • the reducing agent that can be used is, for example, NaBH 4 , NaBH 3 CN, or NaBH(OAc) 3 .
  • the reaction temperature is 0°C to 80°C.
  • the reaction may be performed in the presence of a base or an acid, such as TEA or DIPEA, and the acid, such as AcOH, HCl, or Ti(O i Pr) 4 .
  • the compound of Formula I-A can be synthesized by the method shown in Route B as follows:
  • Hal 2 , R 1 , R 2 , R 5 and R 23a are as defined in Route A above.
  • the first step Compound I-A-5 is deprotected under acidic conditions to produce compound I-A-10.
  • Usable acids are, for example, a 1,4-dioxane solution of HCl, an EA solution of HCl, or a DCM solution of TFA.
  • the reaction temperature is 0°C to 80°C.
  • the second step Compound I-A-10 and I-A-9 undergo reductive amination to produce compound I-A-11.
  • Usable bases are, for example, DIPEA or TEA.
  • the reducing agent that can be used is, for example, NaBH 3 CN or NaBH(OAc) 3 .
  • Usable solvents are, for example, MeOH, EtOH or DCE.
  • the reaction temperature is 0°C to 80°C.
  • the usable solvent is, for example, methanol, ethanol, THF, DCM, DCE, DMA, or a mixture of them and acetic acid in any ratio.
  • the reducing agent that can be used is, for example, NaBH 4 , NaBH 3 CN, or NaBH(OAc) 3 .
  • the reaction temperature is 0°C to 80°C.
  • the reaction may be performed in the presence of a base or an acid, such as TEA or DIPEA, and the acid, such as AcOH, HCl, or Ti(O i Pr) 4 .
  • the third step Compound I-A-11 reacts with boron-containing reagent to produce compound I-A-12.
  • the boron-containing reagent that can be used is, for example, B 2 (pin) 2 .
  • Usable catalysts are, for example, Pd(PPh 3 ) 4 , Pd(dppf)Cl 2 or Pd(dppf) 2 Cl 2 ⁇ DCM.
  • Usable bases are, for example, Cs 2 CO 3 , K 3 PO 4 , Na 2 CO 3 , KOAc, NaHCO 3 or K 2 CO 3 .
  • Usable solvents are, for example, 1,4-dioxane, DMF, DMSO, or CH 3 CN.
  • the reaction temperature is 50°C to 120°C.
  • the fourth step Compound I-A-12 and I-A-2 undergo a coupling reaction (such as Suzuki reaction) to produce compound I-A.
  • a coupling reaction such as Suzuki reaction
  • Usable catalysts are, for example, Pd(PPh 3 ) 4 , Pd(dppf)Cl 2 or Pd(dppf) 2 Cl 2 ⁇ DCM.
  • Usable bases are, for example, Cs 2 CO 3 , K 3 PO 4 , Na 2 CO 3 , KOAc, NaHCO 3 or K 2 CO 3 .
  • the usable solvent is, for example, 1,4-dioxane, DMF, DMSO, or CH 3 CN, or a mixture of any of the above solvents and H 2 O.
  • the reaction temperature is 50°C to 120°C.
  • the compound of formula I-B can be synthesized by the method shown in Route C as follows:
  • Hal 1 , Hal 2 , R 1 , R 2 , R 5 , and R 23a are as defined in Route A above.
  • the first step Compound IB-1 and R 2 -NH 2 undergo a substitution or coupling reaction (for example, Buchwald reaction, Suzuki reaction, Ullmann reaction) in the presence of a base to produce compound IB-2.
  • a substitution or coupling reaction for example, Buchwald reaction, Suzuki reaction, Ullmann reaction
  • reaction conditions are as described in the first step in Route A for the preparation of compounds of formula I-A.
  • Second step Compound I-B-2 and I-A-12 undergo a coupling reaction (such as Suzuki reaction) to produce compound I-B.
  • a coupling reaction such as Suzuki reaction
  • reaction conditions are as described in the fourth step of Route A for the preparation of compounds of formula I-A.
  • compounds of formula I-C can be synthesized by the method shown in Route D as follows:
  • Hal 1 , Hal 2 , R 1 , R 2 , R 5 and R 23a are as defined in Route A above;
  • X 1 is selected from CH and N.
  • the first step Compound IC-1 and R 2 -NH 2 undergo a substitution or coupling reaction (such as Buchwald reaction, Suzuki reaction or Ullmann reaction) in the presence of a base to produce compound IC-2.
  • a substitution or coupling reaction such as Buchwald reaction, Suzuki reaction or Ullmann reaction
  • reaction conditions are as described in the first step in Route A for the preparation of compounds of formula I-A.
  • the second step Compound I-C-3 reacts with boron-containing reagent to produce compound I-C-4.
  • reaction conditions are as described in the third step of Route A for the preparation of compounds of formula I-A.
  • the third step Compound I-C-2 and I-C-4 undergo a coupling reaction (such as Suzuki reaction) to produce compound I-C-5.
  • a coupling reaction such as Suzuki reaction
  • reaction conditions are as described in the fourth step of Route A for the preparation of compounds of formula I-A.
  • Step 4 Compound I-C-5 is deprotected under acidic conditions to produce compound I-C-6.
  • reaction conditions are as described in the fifth step in Route A for the preparation of compounds of formula I-A.
  • Step 5 Compound I-C-6 and I-A-9 undergo reductive amination to produce compound I-C.
  • reaction conditions are as described in the sixth step of Route A for the preparation of compounds of formula I-A.
  • compounds of formula I-D can be synthesized by the method shown in Route E as follows:
  • R 1 and R 2 are as defined in Route A above;
  • R 5 is defined as the above formula ID
  • R 23a and R 23b are each independently selected from H, C 1-6 alkyl, C 1-6 alkoxy and C 3-8 cycloalkyl; or R 23a and R 23b together with the C atom to which they are attached form 3-8 membered cycloalkyl or heterocyclic group, each of said alkyl, alkoxy, cycloalkyl and heterocyclic group is optionally substituted with one or more substituents selected from the following: CN, halogen, C 1-4 alkyl, C 1-4 alkoxy, C 1-4 hydroxyalkyl, C 1-4 haloalkyl and C 1-4 haloalkoxy;
  • X 1 is selected from CH and N;
  • t 0 or 1.
  • Compound I-C-6 and I-D-1 undergo condensation reaction to produce compound I-D.
  • the condensing agent that can be used is, for example, HATU, CDI, HOBt, DMAP, DCC, DIC, EDC, HBTU, HCTU or PyBOP.
  • Usable bases are, for example, TEA, DIPEA, t BuOK, t BuONa, t BuOLi, NaH, NaOH, Cs 2 CO 3 , K 3 PO 4 or Na 2 CO 3 .
  • Usable solvents are, for example, THF, DCM, DCE, MeOH, EtOH, DMF, DMSO, acetone, CH 3 CN, 1,4-dioxane or toluene.
  • the reaction temperature is 0°C to 120°C, such as room temperature.
  • compound ID-1 first reacts with an acylating reagent to form an acid halide, and then reacts with compound IC-6 optionally in the presence of a base to form a compound of formula ID.
  • the acylating agent that can be used is, for example, thionyl chloride or oxalyl chloride.
  • the reaction can also be carried out under a small amount of DMF catalysis.
  • the base that can be used is, for example, TEA or DIPEA.
  • Usable solvents are, for example, THF, DCM, DCE, CH 3 CN, 1,4-dioxane, or toluene.
  • the reaction temperature is 0°C to 100°C.
  • compounds of formula I-E can be synthesized by the method shown in Route F as follows:
  • R 1 , R 2 , R 5 , R 23a , R 23b and t are as defined in Route E above;
  • X 1 is selected from CH and N;
  • Hal 2 is F, Cl, Br or I; preferably, Hal 2 is Cl, Br or I.
  • the first step Compound I-C-2 and I-A-6 undergo a coupling reaction (such as Suzuki reaction) to produce compound I-E-1.
  • a coupling reaction such as Suzuki reaction
  • reaction conditions are as described in the fourth step of Route A for the preparation of compounds of formula I-A.
  • reaction conditions are as described in the fifth step in Route A for the preparation of compounds of formula I-A.
  • the third step Compound I-E-2 and I-D-1 undergo condensation reaction to produce compound I-E.
  • compounds of formula I-F can be synthesized by the method shown in Route G as follows:
  • R 1 , R 2 , R 23a , R 23b and R 5 are as defined in Route E above;
  • X 1 is selected from CH and N;
  • R 4 is absent or selected from hydroxy, CN, C 1-6 alkyl, C 1-6 haloalkyl, and C 1-6 heteroalkyl (for example, C 1-6 alkoxy);
  • R 23c is H, C 1-3 alkyl or C 1-3 alkoxy, each of said alkyl and alkoxy is optionally substituted by one or more substituents selected from the group consisting of OH, CN, halogen , C 1-4 alkoxy and C 1-4 hydroxyalkyl;
  • Hal 2 is F, Cl, Br or I; preferably, Hal 2 is Cl, Br or I;
  • u is 0 or 1
  • n 0 or 1.
  • the first step Compound I-C-2 and I-F-1 undergo a coupling reaction (such as Suzuki reaction) to produce compound I-F-2.
  • a coupling reaction such as Suzuki reaction
  • reaction conditions are as described in the fourth step of Route A for the preparation of compounds of formula I-A.
  • the second step Compound I-F-3 and amine undergo condensation reaction to produce compound I-F-4.
  • reaction conditions are as described in the fifth step in Route A for the preparation of compounds of formula I-A.
  • the fourth step Compound I-F-2 and I-F-5 undergo nucleophilic substitution reaction in the presence of a base to form compound I-F.
  • reaction conditions are as described in the second step of Route A for the preparation of compounds of formula I-A.
  • compounds of formula I-G can be synthesized by the method shown in Route H as follows:
  • R 1 and R 2 are as defined in Route A above;
  • X 1 is selected from CH and N;
  • R 4 is selected from H, C 1-6 alkyl, C 1-6 haloalkyl and C 1-6 heteroalkyl;
  • R 5 is as defined in formula IG above.
  • n 0 or 1.
  • the first step compound IG-1 and R 5 -OH react with Mitsunobu to produce compound IG-2.
  • the reaction reagent that can be used is, for example, PPh 3 , PMe 3 , DIAD, DEAD or DBAD.
  • Solvents that can be used are aprotic solvents such as THF, ether, DCM, DMF or toluene.
  • the reaction temperature is -20°C to 100°C, such as room temperature.
  • the acid that can be used is, for example, a 1,4-dioxane solution of HCl, an EA solution of HCl, or a DCM solution of TFA, or a reaction between the aforementioned acid solution and any solvent selected from the group consisting of THF, MeOH, and EtOH. In a mixture.
  • the reaction temperature is 0°C to 80°C.
  • the third step Compound I-F-2 and I-G-3 undergo a nucleophilic substitution reaction in the presence of a base to form compound I-G.
  • reaction conditions are as described in the second step of Route A for the preparation of compounds of formula I-A.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a preventive or therapeutically effective amount of a compound of the present invention, a stereoisomer, a tautomer of the compound, or a mixture thereof, and N- Oxides, pharmaceutically acceptable salts, co-crystals, polymorphs or solvates of the compounds, or stable isotopic derivatives, metabolites or prodrugs of the compounds.
  • the pharmaceutical composition further comprises one or more pharmaceutically acceptable carriers.
  • the present invention provides pharmaceutical preparations, which are preferably solid preparations, semi-solid preparations, liquid preparations or gaseous preparations.
  • the pharmaceutical composition may also include one or more other therapeutic agents.
  • the pharmaceutical composition or pharmaceutical formulation is preferably administered by oral, intravenous, intraarterial, subcutaneous, intraperitoneal, intramuscular or transdermal route.
  • the present invention provides a compound of the present invention, a stereoisomer, a tautomer or a mixture of the compound, an N-oxide of the compound, and a pharmaceutically acceptable compound of the compound Salts, co-crystals, polymorphs or solvates, or stable isotope derivatives, metabolites or prodrugs of said compounds, or pharmaceutical compositions as described above, or pharmaceutical preparations of the present invention are prepared for prevention Or use in drugs for treating diseases or conditions related to RET activity.
  • the present invention provides a compound of the present invention, a stereoisomer, a tautomer or a mixture of the compound, an N-oxide of the compound, and a pharmaceutically acceptable compound of the compound Salts, co-crystals, polymorphs or solvates, or stable isotope derivatives, metabolites or prodrugs of said compounds, or pharmaceutical compositions as described above, or pharmaceutical preparations of the present invention are prepared for regulation (For example, to reduce or inhibit) the use of RET activity in medicine.
  • the present invention provides a compound of the present invention, a stereoisomer, a tautomer or a mixture of the compound, an N-oxide of the compound, and a pharmaceutically acceptable compound of the compound Salts, co-crystals, polymorphs or solvates, or stable isotope derivatives, metabolites or prodrugs of said compounds, or pharmaceutical compositions as described above, or pharmaceutical preparations of the present invention, which are used for prevention Or to treat diseases or conditions related to RET activity.
  • the present invention provides a method of preventing or treating diseases or conditions associated with RET activity, the method comprising administering to an individual in need thereof an effective amount of a compound of the present invention, a stereoisomer of the compound , Tautomers or mixtures thereof, N-oxides of the compounds, pharmaceutically acceptable salts, co-crystals, polymorphs or solvates of the compounds, or stable isotope derivatives of the compounds A substance, metabolite or prodrug, or a pharmaceutical composition as described above, or a pharmaceutical preparation of the present invention.
  • the disease or condition associated with RET activity is preferably cancer or tumor, or irritable bowel syndrome.
  • the cancer or tumor is further preferably lung cancer (such as non-small cell lung cancer), breast cancer, head and neck cancer, rectal cancer, liver cancer, lymphoma, thyroid cancer (such as medullary thyroid cancer or papillary thyroid cancer) ), colon cancer, multiple myeloma, melanoma, glioma, brain tumor or sarcoma.
  • lung cancer such as non-small cell lung cancer
  • breast cancer head and neck cancer
  • rectal cancer liver cancer
  • lymphoma such as medullary thyroid cancer or papillary thyroid cancer
  • thyroid cancer such as medullary thyroid cancer or papillary thyroid cancer
  • colon cancer multiple myeloma, melanoma, glioma, brain tumor or sarcoma.
  • pharmaceutically acceptable carrier refers to a diluent, adjuvant, excipient or vehicle administered with the therapeutic agent, and it is suitable for contact with humans and/or within the scope of reasonable medical judgment Tissues of other animals without excessive toxicity, irritation, allergic reactions, or other problems or complications corresponding to a reasonable benefit/risk ratio.
  • the pharmaceutically acceptable carrier that can be used in the pharmaceutical composition of the present invention includes, but is not limited to, sterile liquid. Examples of suitable pharmaceutically acceptable carriers are described in Remington's Pharmaceutical Sciences (1990).
  • the pharmaceutical composition of the present invention can act systemically and/or locally. For this purpose, they can be administered by a suitable route.
  • the pharmaceutical composition of the present invention can be administered in a suitable dosage form.
  • an effective amount refers to the amount of a compound that will relieve one or more symptoms of the condition being treated to a certain extent after being administered.
  • the dosage regimen can be adjusted to provide the best desired response. For example, a single bolus can be administered, several divided doses can be administered over time, or the dose can be proportionally reduced or increased as indicated by the urgent need for the treatment situation. It should be noted that the dose value may vary with the type and severity of the condition to be alleviated, and may include single or multiple doses. It should be further understood that for any particular individual, the specific dosing regimen should be adjusted over time according to the needs of the individual and the professional judgment of the person administering the composition or supervising the administration of the composition.
  • the amount of the compound of the present invention administered will depend on the individual being treated, the severity of the disorder or condition, the rate of administration, the treatment of the compound, and the judgment of the prescribing physician. Generally, the effective dose is about 0.0001 to about 50 mg per kg body weight per day. In some cases, a dose level not higher than the lower limit of the aforementioned range may be sufficient, while in other cases, a larger dose can still be used without causing any harmful side effects, provided that the larger The dose is divided into several smaller doses to be administered throughout the day.
  • the content or amount of the compound of the present invention in the pharmaceutical composition may be about 0.01 mg to about 1000 mg.
  • treating means reversing, alleviating, or inhibiting the disease or condition to which such term is applied or the progression of one or more symptoms of such disease or condition, or Preventing such a disorder or condition or one or more symptoms of such a disorder or condition.
  • “Individual” as used herein includes human or non-human animals.
  • Exemplary human individuals include human individuals (referred to as patients) or normal individuals suffering from diseases such as the diseases described herein.
  • “non-human animals” include all vertebrates, such as non-mammals (such as birds, amphibians, reptiles) and mammals, such as non-human primates, livestock and/or domesticated animals (such as sheep, dogs). , Cats, cows, pigs, etc.).
  • the pharmaceutical composition of the present invention may also include one or more additional therapeutic or preventive agents (for example, other drugs used to treat cancer or tumor diseases).
  • the methods of the present invention may also include the administration of one or more additional therapeutic or preventive agents (e.g., other drugs used to treat cancer or tumor diseases).
  • the compound of the present invention is separated and purified by preparative TLC, silica gel column chromatography, Prep-HPLC and/or flash column chromatography (Flash column chromatography), and its structure is confirmed by 1 H NMR and/or MS.
  • the reaction monitoring is carried out by TLC or LC-MS.
  • the 1 H NMR spectroscopy method uses a Bruker superconducting nuclear magnetic resonance spectrometer (model AVACE III HD 400MHz).
  • LC/MS adopts Aglient 1260Infinity/Aglient 6120Quadrupole.
  • TLC uses silica gel GF 254 as the stationary phase.
  • Fast column chromatography uses Biotage fast column chromatography.
  • Prep-HPLC adopts Agilent 1260, Waters 2489 and GeLai 3500 chromatographs.
  • the microwave reaction was performed using a BiotageInitiator microwave reactor.
  • reaction temperature is room temperature (15-30°C).
  • the reagents used in this application are purchased from companies such as Acros Organics, Aldrich Chemical Company, or Terbo Chemical.
  • the fifth step 2-(6-(6-benzyl-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-( Preparation of 5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 1)
  • the second step 2-(6-(6-((6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo[3.1 .1]Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 17)
  • the fourth step 2-(6-(4-((6-methoxypyridin-3-yl)methyl)piperazin-1-yl)pyridin-3-yl)-6-methyl-N-( Preparation of 5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 49)
  • Example 18 6-methyl-2-(6-(6-((6-(4-methyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6- Diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 62)
  • Step 1 Preparation of 1-(4-(1,3-dioxolane-2-yl)phenyl)-4-methyl-1H-pyrazole (Compound 88c)
  • Example 32 2-(6-(6-((6-(3,4-Dimethyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-diazide Heterobicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 121)
  • Step 1 Preparation of 2-(3,4-Dimethyl-1H-pyrazol-1-yl)-5-(1,3-dioxolane-2-yl)pyridine (Compound 121c)
  • the third step 2-(6-(6-((6-(3,4-dimethyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-diazide Heterobicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 121) Preparation
  • the first step Preparation of 5-(1,3-dioxolane-2-yl)-2-(4-fluoro-1H-imidazol-1-yl)pyridine (compound 64b)
  • the third step 2-(6-(6-((6-(4-fluoro-1H-imidazol-1-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo[3.1. 1) Preparation of heptane-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 64)
  • Example 36 2-(6-(6-(1-(6-(4-Fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-3,6-diazabicyclo [3.1.1]Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 52)
  • Step 1 Preparation of tert-butyl 4-((6-methylpyridin-3-yl)oxy)piperidine-1-carboxylate (Compound 119b)
  • Step 1 Preparation of 1-(4-(1,3-dioxolane-2-yl)phenyl)-3-fluoro-1H-pyrazole (compound 89b)
  • the third step 2-(6-(6-(4-(3-fluoro-1H-pyrazol-1-yl)benzyl)-3,6-diazabicyclo[3.1.1]heptane-3 -Yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 89)
  • Example 40 6'-(6-((6-(4-Fluoro-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1 ]Heptan-3-yl)-4-methyl-N-(5-methyl-1H-pyrazol-3-yl)-[2,3'-bipyridine]-6-amine (Compound 6)
  • Step 2 Preparation of 3-((6-bromo-4-methylpyridin-2-yl)amino)-5-methyl-1H-pyrazole-1-carboxylic acid tert-butyl ester (Compound 6d)
  • Example 41 2-(6-(6-((6'-methoxy-[2,3'-bipyridyl]-5-yl)methyl)-3,6-diazabicyclo[3.1. 1)Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 7)
  • Step 1 Preparation of 5-(1,3-dioxolane-2-yl)-6'-methoxy-2,3'-bipyridine (compound 7b)
  • the third step 2-(6-(6-((6'-methoxy-[2,3'-bipyridyl]-5-yl)methyl)-3,6-diazabicyclo[3.1. 1) Preparation of heptane-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 7)
  • Example 43 6-methyl-2-(6-(6-((6-(5-methyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6- Diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 9)
  • the first step preparation of 2-chloro-N-(5-cyclopropyl-1H-pyrazol-3-yl)-6-methylpyrimidin-4-amine (compound 10c)
  • the fourth step 6-((6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3-(5-(4,4,5,5-tetramethyl Preparation of yl-1,3,2-dioxaborolan-2-yl)pyridin-2-yl)-3,6-diazabicyclo[3.1.1]heptane (compound 10f)
  • the first step preparation of 2-(3,5-dimethyl-1H-pyrazol-1-yl)-5-(1,3-dioxolane-2-yl)pyridine (compound 11b)
  • the third step 2-(6-(6-((6-(3,5-dimethyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-diazide Heterobicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 11)
  • Step 1 Preparation of 5-(1,3-dioxolane-2-yl)-2-(5-isopropyl-1H-pyrazol-1-yl)pyridine (Compound 12b)
  • the third step 2-(6-(6-((6-(5-isopropyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo [3.1.1]Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 12)
  • Instrument model Biotage Isolera Prime 2.3.1, Chromatographic Column: Agela Technologies C18 spherical 20-35um 100A, 12g; Chromatographic column temperature: 25°C; Flow rate: 15.0mL/min; Detection wavelength: 254nm; Elution gradient: (0min: 0% A, 100% B; 3.0 min: 0% A, 100% B; 20 min: 80% A, 20% B); mobile phase A: 100% acetonitrile; mobile phase B: 0.5% ammonium bicarbonate aqueous solution; compound Collection time: 10.4min-11.8min.
  • Instrument model Biotage Isolera Prime 2.3.1, Chromatographic Column: Agela Technologies C18 spherical 20-35um 100A, 12g; Chromatographic column temperature: 25°C; Flow rate: 15.0mL/min; Detection wavelength: 254nm; Elution gradient: (0min: 0% A, 100% B; 3.0 min: 0% A, 100% B; 20 min: 80% A, 20% B); mobile phase A: 100% acetonitrile; mobile phase B: 0.5% ammonium bicarbonate aqueous solution; compound Collection time: 11.4min-12.8min.
  • Example 48 (6-(4-Fluoro-1H-pyrazol-1-yl)pyridin-3-yl)(3-(5-(4-methyl-6-((5-methyl-1H- (Pyrazol-3-yl)amino)pyrimidin-2-yl)pyridin-2-yl)-3,6-diazabicyclo[3.1.1]heptane-6-yl)methanone (Compound 26)
  • the third step (6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)(3-(5-(4-methyl-6-((5-methyl-1H- (Pyrazol-3-yl)amino)pyrimidin-2-yl)pyridin-2-yl)-3,6-diazabicyclo[3.1.1]heptane-6-yl)methanone (Compound 26)
  • Step 4 Preparation of 2-(5-cyclobutoxy-1H-pyrazol-1-yl)-5-(1,3-dioxolane-2-yl)pyridine (compound 27f)
  • Example 50 2-(6-(6-((6-(4-chloro-3-methyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-di Azabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine and 2-(6-(6-((6-(4-chloro-5-methyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo[ 3.1.1]Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 28/compound 28')
  • the first step 2-(4-chloro-3-methyl-1H-pyrazol-1-yl)-5-(1,3-dioxolane-2-yl)pyridine and 2-(4-chloro Preparation of -5-methyl-1H-pyrazol-1-yl)-5-(1,3-dioxolan-2-yl)pyridine (compound 28b/compound 28b')
  • the second step 6-(4-chloro-3-methyl-1H-pyrazol-1-yl)nicotinaldehyde and 6-(4-chloro-5-methyl-1H-pyrazol-1-yl) Preparation of nicotine aldehyde (compound 28c/compound 28c')
  • the third step 2-(6-(6-((6-(4-chloro-3-methyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-di Azabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine and 2-(6-(6-((6-(4-chloro-5-methyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo[ 3.1.1]Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 28/compound 28') Preparation
  • Example 52 2-(6-(6-((R)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-3,6- Diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine And 2-(6-(6-((S)-1-(6-(4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-3,6-diazepine Bicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 52 -1/Compound 52-2)
  • the third step Preparation of 2-(5-cyclopropoxy-1H-pyrazol-1-yl)-5-(1,3-dioxolane-2-yl)pyridine (compound 30d)
  • Example 54 2-(6-(6-((6-(3-(fluoromethyl)-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-diazide Heterobicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 31)
  • the third step Preparation of 5-(1,3-dioxolane-2-yl)-2-(3-(fluoromethyl)-1H-pyrazol-1-yl)pyridine (compound 31d)
  • the fifth step 2-(6-(6-((6-(3-(fluoromethyl)-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-diazide Heterobicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 31)
  • Step 4 Preparation of 5-(1,3-dioxolane-2-yl)-2-(5-ethyl-1H-pyrazol-1-yl)pyridine (Compound 46d)
  • the sixth step 2-(6-(6-((6-(5-ethyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo[ 3.1.1]Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 46) preparation
  • Example 56 2-(6-(6-((6-(4-Fluoro-5-methyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-di Azabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine ( Compound 122)
  • the first step the preparation of 4-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole (compound 122a)
  • Step 4 Preparation of 5-(1,3-dioxolane-2-yl)-2-(4-fluoro-5-methyl-1H-pyrazol-1-yl)pyridine (Compound 122d)
  • the sixth step 2-(6-(6-((6-(4-fluoro-5-methyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-di Azabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine ( Compound 122) Preparation
  • the third step 2-(6-(6-((6-(1H-pyrrol-2-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptane -3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 123)
  • Example 58 2-(6-(6-((6-(3-cyclopropyl-4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6- Diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 124)
  • Step 2 Preparation of 5-(1,3-dioxolan-2-yl)-2-(4-fluoro-3-iodo-1H-pyrazol-1-yl)pyridine (Compound 124b)
  • Step 4 Preparation of 6-(3-cyclopropyl-4-fluoro-1H-pyrazol-1-yl)nicotinaldehyde (Compound 124d)
  • the fifth step 2-(6-(6-((6-(3-cyclopropyl-4-fluoro-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6- Diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 124)
  • Example 59 2-(6-(6-((R)-1-(6-(1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-3,6-diazabicyclo [3.1.1]Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine and 2-( 6-(6-((S)-1-(6-(1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-3,6-diazabicyclo[3.1.1]heptane -3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 125-1/Compound 125-2)
  • the third step 2-(6-(6-((R)-1-(6-(1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-3,6-diazabicyclo [3.1.1]Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine and 2-( 6-(6-((S)-1-(6-(1H-pyrazol-1-yl)pyridin-3-yl)ethyl)-3,6-diazabicyclo[3.1.1]heptane -3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 125-1/Compound 125-2) Preparation
  • Compound 125 (250 mg) was resolved by chiral Prep-HPLC to obtain compound 125-1 (retention time 7.647 min) and compound 125-2 (retention time 11.638 min). The two compounds were differentiated and defined according to the retention time of chiral resolution, and the stereo structure was not identified. Thus, compound 125-1 (101 mg, ee: 100%), MS m/z (ESI): 533.9 [M+H] + ; compound 125-2 (100 mg, ee: 99.93%), MS m/z ( ESI): 533.9 [M+H] + .
  • Example 60 2-(6-(6-((6-(4-fluoromethyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo [3.1.1]Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 126) And (1-(5-((3-(5-(4-methyl-6-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-2-yl)pyridine-2- (Yl)-3,6-diazabicyclo[3.1.1]heptane-6-yl)methyl)pyridin-2-yl)-1H-pyrazol-4-yl)methanol (Compound 127)
  • the first step Preparation of 1-(5-(1,3-dioxolane-2-yl)pyridin-2-yl)-1H-pyrazole-4-carboxylic acid ethyl ester (compound 126b)
  • Step 2 Preparation of 1-(5-(1,3-dioxolane-2-yl)pyridin-2-yl)-1H-pyrazol-4-yl)methanol (Compound 126c)
  • the fifth step 2-(6-(6-((6-(4-fluoromethyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo [3.1.1]Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 126) And (1-(5-((3-(5-(4-methyl-6-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-2-yl)pyridine-2- (Yl)-3,6-diazabicyclo[3.1.1]heptane-6-yl)methyl)pyridin-2-yl)-1H-pyrazol-4-yl)methanol (Compound 127)
  • Example 61 6-Methyl-N-(5-methyl-1H-pyrazol-3-yl)-2-(6-(6-((6-(1-methyl-1H-pyrrole-2 -Yl)pyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrimidin-4-amine (compound 128)
  • the fifth step 6-methyl-N-(5-methyl-1H-pyrazol-3-yl)-2-(6-(6-((6-(1-methyl-1H-pyrrole-2 -Yl)pyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrimidin-4-amine (compound 128)
  • Example 62 6-Methyl-N-(5-methyl-1H-pyrazol-3-yl)-2-(6-(6-((6-(thiazol-4-yl)pyridine-3- (Yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrimidin-4-amine (compound 129)
  • Step 1 Preparation of 4-(5-(1,3-dioxolane-2-yl)pyridin-2-yl)thiazole (compound 129b)
  • the third step 6-methyl-N-(5-methyl-1H-pyrazol-3-yl)-2-(6-(6-((6-(thiazol-4-yl)pyridine-3- (Yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrimidin-4-amine (compound 129)
  • Step 1 Preparation of 5-(1,3-dioxolane-2-yl)-2,2'-bipyridine (compound 130b)
  • Example 64 2-(6-(6-((6-(5-Fluoro-1H-pyrrol-2-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo[3.1. 1)Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 131)
  • Step 1 Preparation of 6-(5-fluoro-1H-pyrrol-2-yl)nicotinaldehyde (compound 131a)
  • the second step 2-(6-(6-((6-(5-fluoro-1H-pyrrol-2-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo[3.1. 1) Preparation of heptane-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 131)
  • Example 65 2-(6-(6-((6-(1H-pyrazol-5-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]hepta Alkyl-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 132)
  • Step 1 Preparation of 6-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-5-yl)nicotinaldehyde (compound 132b)
  • the third step 2-(6-(6-((6-(1H-pyrazol-5-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptan Alkyl-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (compound 132)
  • Example 66 6-Methyl-N-(5-methyl-1H-pyrazol-3-yl)-2-(6-(6-((6-(5-methylfuran-2-yl) (Pyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrimidin-4-amine (compound 133)
  • Step 1 Preparation of 2-(5-(1,3-dioxolane-2-yl)pyridin-2-yl)oxazole (compound 134b)
  • Example 68 2-(6-(6-((6-(1-isopropyl-1H-pyrazol-4-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo [3.1.1]Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 135)
  • Step 1 Preparation of 5-(1,3-dioxolane-2-yl)-2-(1H-pyrazol-4-yl)pyridine (compound 135b)
  • Step 2 Preparation of 5-(1,3-dioxolane-2-yl)-2-(1-isopropyl-1H-pyrazol-4-yl)pyridine (Compound 135d)
  • the fourth step 2-(6-(6-((6-(1-isopropyl-1H-pyrazol-4-yl)pyridin-3-yl)methyl)-3,6-diazabicyclo [3.1.1]Heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine (Compound 135)
  • Example 69 2-(1-(5-((3-(5-(4-methyl-6-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-2-yl )Pyridin-2-yl)-3,6-diazabicyclo[3.1.1]heptane-6-yl)methyl)pyridin-2-yl)-1H-pyrazol-5-yl)propan-2 -Alcohol (Compound 136)
  • Step 2 Preparation of 6-(3-(2-hydroxyprop-2-yl)-1H-pyrazol-1-yl)nicotinaldehyde (Compound 136b)
  • the third step 2-(1-(5-((3-(5-(4-methyl-6-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-2-yl )Pyridin-2-yl)-3,6-diazabicyclo[3.1.1]heptane-6-yl)methyl)pyridin-2-yl)-1H-pyrazol-5-yl)propan-2 -Preparation of alcohol (compound 136)
  • Example 70 (1-(5-((3-(5-(4-methyl-6-((5-methyl-1H-pyrazol-3-yl)amino)pyrimidin-2-yl)pyridine -2-yl)-3,6-diazabicyclo[3.1.1]heptane-6-yl)methyl)pyridin-2-yl)-1H-pyrazol-3-yl)methanol (compound 137)
  • Step 1 Preparation of 6-(3-(hydroxymethyl)-1H-pyrazol-1-yl)nicotinaldehyde (compound 137a)
  • Example 71 2-(6-(6-((6-(4-Fluoro-3-methyl-1H-pyrazol-1-yl)pyridin-3-yl)methyl)-3,6-di Azabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)-6-methyl-N-(5-methyl-1H-pyrazol-3-yl)pyrimidin-4-amine ( Compound 138)
  • Relative inhibitory activity percentage 1-(compound groups of different concentrations-blank control) / (negative control-blank control) * 100%
  • y is the relative inhibitory activity percentage
  • max and min are the maximum and minimum values of the fitted curve
  • x is the logarithmic concentration of the compound
  • Hillslope is the slope of the curve.
  • the inhibitory effect of the compound of the present invention on the enzyme activity of VEGFR2 was determined according to the instructions of HTRF KinEASE-TK kit (Cisbio). After pre-incubating the VEGFR2 enzyme with test compounds of different concentrations for 30 min at room temperature, the substrate and adenosine triphosphate (ATP) were added to initiate the reaction. After incubating at room temperature for 40 min, TK antibody-cryptate and streptavidin-XL665 were added, and the detection was performed after incubating at room temperature for 45 min. With the vehicle group (DMSO) as the negative control and the buffer group (without VEGFR2 enzyme) as the blank control, the relative inhibitory activity percentages (ie, the inhibition rate) of different concentrations of compounds were calculated according to the following formula:
  • Relative inhibitory activity percentage 1-(compound groups of different concentrations-blank control) / (negative control-blank control) * 100%
  • y is the relative inhibitory activity percentage
  • max and min are the maximum and minimum values of the fitted curve
  • x is the logarithmic concentration of the compound
  • Hillslope is the slope of the curve.
  • N/A means not tested.
  • Compound number RET-V804M inhibition rate Compound number RET-V804M inhibition rate 6(10nM) 83% 84(100nM) 72% 7(10nM) 46% 85(100nM) 77% 8(10nM) 46% 86(10nM) 64% 15(100nM) 91% 88(10nM) 62% 16(100nM) 92% 89(10nM) 70% 18(100nM) 70% 91(10nM) 70% 21(100nM) 49% 96(100nM) 84% 23(10nM) 47% 98(100nM) 63% 26(10nM) 83% 110(100nM) 62% 27(10nM) 87% 111(100nM) 46% 28'(10nM) 34% 117(100nM) 76% 29(10nM) 52% 118(100nM) 74% 30(10nM) 88% 125-2(10nM) 82% 31(10nM) 81% 126(100n
  • Compound number VEGFR2 inhibition rate Compound number VEGFR2 inhibition rate 1(100nM) 38% 70(100nM) 14% 2(100nM) 29% 80(100nM) -10% 3(100nM) 14% 82(100nM) 15% 4(1000nM) 52% 83(100nM) 18% 6(100nM) -11% 86(100nM) 52% 7(100nM) 16% 88(100nM) 32% 8(300nM) 5% 89(100nM) 16% 9(300nM) 69% 91(100nM) 26% 10(300nM) 69% 96(100nM) 4% 12(300nM) 62% 119(100nM) -7% 15(100nM) 29% 120(100nM) 28% 23(100nM) -10% 121(100nM) 37%
  • BLU-667 prepared according to Example 5 of WO2017/079140A1
  • compound 17 were administered to male SD rats by intragastric (PO), respectively, and the plasma concentration of BLU-667 and compound 17 in rats and the concentration of brain, lung and thyroid tissues were measured , To investigate the pharmacokinetic characteristics.
  • the dose of PO administration is 5 mg/kg, and the vehicle is 0.5% MC (methyl cellulose).
  • PO was administered, and blood samples were collected at different time points (0h before administration, 0.25h, 0.5h, 1h, 2h, 4h, 6h, 8h and 24h after administration).
  • the blood samples were anticoagulated with dipotassium ethylenediaminetetraacetate, and the plasma samples were obtained after centrifugation and stored at -80°C.
  • Rats were sacrificed by bleeding from the abdominal aorta at 0.5h, 2h, 8h and 24h after PO administration, and brain, lung and thyroid were collected. After washing, homogenized with saline in a certain proportion to obtain tissue samples, which were stored in- 80°C. Each plasma sample and tissue sample were processed with precipitated protein and then analyzed by LC-MS/MS. Using WinNonlin 6.3 software, the non-compartmental model was used to calculate the pharmacokinetic parameters. The results are shown in Table 5.
  • composition Compound 17 and BLU-667 were dissolved in 0.1M H 3 PO 4 aqueous solution and 0.1M HOAc aqueous solution, respectively, to prepare a clear solution (pH about 4.0).
  • the vehicle control group used an aqueous H 3 PO 4 solution with a pH of about 4.0.
  • the mode of administration is PO, BID.
  • Tumor measurement Measure the diameter of the tumor with a vernier caliper twice a week.
  • TGI tumor growth inhibition rate

Abstract

本发明涉及杂环化合物、包含其的药物组合物及其制备方法和用途。具体而言,本发明的化合物由式(I)表示,其用于预防或治疗与RET活性相关的疾病或病况。

Description

杂环化合物、包含其的药物组合物及其制备方法和用途 发明领域
本发明涉及新的杂环化合物、包含其的药物组合物、其制备方法及其用于预防或治疗与RET(Rearranged during transfection)活性相关的疾病或病况。
发明背景
蛋白激酶是一类催化蛋白质磷酸化反应的酶。通过介导细胞信号传导过程,蛋白质磷酸化调控细胞的生理活动,例如细胞的存活、增殖、分化、凋亡与代谢等。蛋白激酶的功能失调与很多疾病密切相关,包括肿瘤、自身免疫病、炎性反应、中枢神经系统疾病、心血管疾病及糖尿病等。
RET是一种原癌基因,其编码的RET蛋白是一种跨膜受体型酪氨酸蛋白激酶,由富含半胱氨酸的钙黏素样胞外区(用于结合配体)、跨膜区以及具有酪氨酸激酶活性的胞内结构区三部分构成。活化后的RET蛋白可激活多个下游信号通路,包括RAS/RAF/ERK通路、PI3K/Akt通路和JNK通路,从而导致细胞增殖、迁移和分化。RET基因改变(突变或融合)和野生型RET基因的异常表达导致RET蛋白异常激活,从而使得信号通路过度活跃,这是致癌的主要机制之一。异常激活的RET蛋白通过多种信号通路参与不同肿瘤细胞的增殖和侵袭,从而影响肿瘤的发生发展。RET基因改变对下游的级联反应作用更为显著,其中RET基因突变主要与甲状腺髓样癌和乳头状甲状腺癌相关,RET基因融合主要与非小细胞肺癌和慢性髓细胞白血病相关。因此,抑制RET活性具有重大的医疗价值(Nature Reviews Cancer,2014,14(3):173-86)。
RET抑制剂具有治疗和预防多种疾病(例如肿瘤、肠易激综合征等)的巨大潜能。目前有五个化合物处于临床试验阶段,并且多家公司的化合物处于临床前研究阶段。然而,目前尚未有以RET为主要靶点的抑制剂上市。因此,需要开发新的、高效低毒的RET抑制剂来满足临床需求。
发明概述
本发明提供新的杂环化合物,其对RET具有良好的抑制作用,并且具有良好的药物代谢动力学、安全性等性质。
本发明的一个方面提供式I的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药:
Figure PCTCN2020074696-appb-000001
其中:
环A选自C 6-10芳环和5-6元杂芳环;
环B选自C 3-8环烷基和4-11元杂环基;
X 1选自CH和N;
R 1选自H、卤素、羟基、氰基、C 1-6烷基、C 1-6杂烷基(例如C 1-6烷氧基)、C 3-8环烷基、4-10元杂环基和-NR 20aR 20b,所述烷基、杂烷基(例如烷氧基)、环烷基和杂环基各自任选地被一个或多个选 自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基和C 1-4杂烷基(例如C 1-4烷氧基);
R 2选自C 1-6烷基、C 1-6杂烷基、C 3-8环烷基、4-10元杂环基、5-10元杂芳基和-C(=O)R 21,所述烷基、杂烷基、环烷基、杂环基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基和C 3-6环烷基;
R 3和R 4不存在或者在每次出现时各自独立地选自羟基、卤素、CN、C 1-6烷基、C 1-6杂烷基(例如C 1-6烷氧基)和C 3-6环烷基,所述烷基、杂烷基(例如烷氧基)和环烷基各自任选地被一个或多个选自下列的取代基取代:卤素、CN、C 1-4烷基、C 1-4卤代烷基、C 1-4烷氧基和C 1-4卤代烷氧基;当m大于1时,两个R 3任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基;和/或当n大于1时,两个R 4任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基;
L选自-O-、-S-、-S(O)-、-S(O) 2-、-N=CR 21-、-N(R 23a)-C(O)-、C 1-6亚烷基、C 1-6亚杂烷基、C 2-6亚烯基、C 2-6亚炔基、
Figure PCTCN2020074696-appb-000002
Figure PCTCN2020074696-appb-000003
Figure PCTCN2020074696-appb-000004
所述亚烷基、亚杂烷基、亚烯基和亚炔基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6卤代烷氧基、C 1-6杂烷基(例如C 1-6烷氧基)和C 3-8环烷基;或者L为-N(R 23a)-;
R 5选自羟基、卤素、CN、NO 2、C 1-6烷基、C 1-6杂烷基(例如C 1-6烷氧基)、C 2-6烯基、C 2-6炔基、C 3-8环烷基、C 3-8环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 20aR 20b、-OR 21、-SR 21、-S(=O)R 22、-S(=O) 2R 22、-S(=O)NR 20aR 20b、-S(=O) 2NR 20aR 20b、-NR 20aS(=O)R 20b、-NR 20aS(=O) 2R 20b、-C(=O)R 21、-C(=O)NR 23aR 23b、-NR 23aC(=O)R 23b、-OC(=O)NR 23aR 23b和-NR 24aC(=O)NR 25aR 25b,所述烷基、杂烷基(例如烷氧基)、烯基、炔基、环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基、C 3-6环烷氧基和4-10元杂环基;
R 20a、R 20b、R 23a、R 23b、R 23c、R 24a、R 25a和R 25b各自独立地选自H、OH、C 1-6烷基、C 1-6烷氧基和C 3-8环烷基;或者R 20a与R 20b、R 23a与R 23b或R 25a与R 25b连同其所连接的原子一起形成3-8元环烷基或杂环基,所述烷基、烷氧基、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:OH、CN、卤素、NO 2、C 1-4烷基、C 1-4烷氧基、C 1-4羟烷基、C 1-4卤代烷基和C 1-4卤代烷氧基;
R 30a、R 30b、R 33a、R 33b、R 34a、R 35a和R 35b各自独立地选自H、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基和C 1-6卤代烷氧基;
R 21、R 22、R 31和R 32各自独立地选自C 1-6烷基、C 1-6烷氧基、C 3-8环烷基、4-10元杂环基、C 6-12芳基和5-10元杂芳基,所述烷基、烷氧基、环烷基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:OH、卤素、CN、C 1-4烷基、C 1-4烷氧基、C 1-4卤代烷基、C 1-4卤代烷氧基、C 3-6环烷基和4-10元杂环基;
m为0、1、2、3或4;
n为0、1、2、3或4;
t为0、1、2、3或4;且
u为0、1、2、3或4;
条件是,当环B为哌嗪环且X 1为CH时,R 2不是4-CF 3-吡啶-2-基或4-CN-吡啶-2-基。
本发明的另一方面提供药物组合物,其包含预防或治疗有效量的本发明的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药。任选地,所述药物组合物还包含一种或多种药学上可接受的载体。
本发明的另一方面提供本发明的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,或者如上所述的药物组合物在制备用于预防或治疗与RET活性相关的疾病或病况的药物中的用途。
本发明的另一方面提供本发明的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,或者如上所述的药物组合物,其用于预防或治疗与RET活性相关的疾病或病况。
本发明的另一方面提供预防或治疗与RET活性相关的疾病或病况的方法,所述方法包括向需要其的个体给药有效量的本发明的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,或者如上所述的药物组合物。
本发明的另一方面提供制备本发明的化合物的方法。
附图简述
图1显示化合物17及对照化合物BLU-667在髓样甲状腺癌TT细胞皮下移植瘤模型中进行的体内药效测试结果。
发明详述
定义
除非在下文中另有定义,本文中所用的所有技术术语和科学术语的含义意图与本领域技术人员通常所理解的相同。提及本文中使用的技术意图指在本领域中通常所理解的技术,包括那些对本领域技术人员显而易见的技术的变化或等效技术的替换。虽然相信以下术语对于本领域技术人员很好理解,但仍然阐述以下定义以更好地解释本发明。
术语“包括”、“包含”、“具有”、“含有”或“涉及”及其在本文中的其它变体形式为包含性的(inclusive)或开放式的,其不排除其它未列举的元素或方法步骤,尽管其它未列举的元素或方法步骤不一定存在(即,这些术语也涵盖术语“基本上由……组成”和“由……组成”)。
如本文中所使用,术语“烷基”定义为线性或支化饱和脂肪族烃。在一些实施方案中,烷基具有1至12个,例如1至6个碳原子。例如,如本文中所使用,术语“C 1-6烷基”和“C 1-4烷基”分别指具有1-6个碳原子和1-4个碳原子的线性或支化的基团(例如甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、异戊基、新戊基或正己基),其任选地被一个或多个(诸如1至3个)适合的取代基如卤素取代(此时该基团被称作“卤代烷基”)(例如CH 2F、CHF 2、CF 3、CCl 3、C 2F 5、C 2Cl 5、CH 2CF 3、CH 2Cl或-CH 2CH 2CF 3等)。术语“C 1-4烷基”指1至4个碳原子的线性或支化的脂肪族烃链(即甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基或叔丁基)。术语“亚烷基”表示相应的二价基团,包括例如“C 1-8亚烷基”、“C 1-6亚烷基”、“C 1-4亚烷基”等,具体实例包括但不限于:亚甲基(-CH 2-)、亚乙基(-CH 2CH 2-或-CH(CH 3)-)、亚丙基(-CH 2CH 2CH 2-)、亚异丙基(-CH(CH 3)CH 2-)、亚丁基、亚戊基、亚己基等。所述亚烷基任选地被一个或多个(诸如1至3个)相同或不同的取代基取代。
如本文中所使用,术语“杂烷基”指任选被取代的烷基,其具有一个或多个选自除碳以外的原子的骨架链原子,例如氧、氮、硫、磷或其组合。可以给出数值范围(例如C 1-6杂烷基)是指链中的碳数目,在此实例中包括1-6个碳原子。例如,-CH 2OCH 2CH 3基团被称为C 3杂烷基。与分子其余部分的连接可以通过杂烷基链中的杂原子或碳原子进行。术语“亚杂烷基”表示相应的二价基团,包括例如“C 1-6亚杂烷基”、“C 1-4亚杂烷基”等。
如本文中所使用,术语“卤代烷基”是指被一个或多个(诸如1至3个)相同或不同的卤素原子取代的烷基,术语“C 1-8卤代烷基”、“C 1-6卤代烷基”和“C 1-4卤代烷基”分别指具有1至8个碳原子、1至6个碳原子和1-4个碳原子的卤代烷基,例如-CF 3、-C 2F 5、-CHF 2、-CH 2F、-CH 2CF 3、-CH 2Cl或-CH 2CH 2CF 3等。
如本文中所使用,术语“羟烷基”是指烷基中的氢原子被一个或多个羟基取代所形成的基团,例 如C 1-4羟烷基或C 1-3羟烷基,其实例包括但不限于羟甲基、羟乙基、羟丙基、羟丁基、-CH(OH)CH 3、-C(CH 3) 2OH等。
如本文中所使用,术语“烷氧基”意指在烷基(如上文所定义)任意合理的位置插入氧原子的基团,优选为C 1-8烷氧基、C 1-6烷氧基、C 1-4烷氧基或C 1-3烷氧基。C 1-6烷氧基的代表性实例包括但不限于甲氧基、乙氧基、丙氧基、异丙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基、叔丁氧基、戊氧基、己氧基、-CH 2-OCH 3等,所述烷氧基任选地被一个或多个(诸如1至3个)相同或不同的取代基取代。
如本文中所使用,术语“亚烷氧基”指二价的烷氧基,例如-OCH 2-、-OCH(CH 3)CH 2-、-OCH 2CH 2O-、-CH 2CH 2O-等,所述亚烷氧基任选地被一个或多个(诸如1至3个)相同或不同的取代基取代。
如本文中所使用,术语“烯基”意指线性的或支化的单价烃基,其包含一个或多个双键,且具有2-6个碳原子(“C 2-6烯基”)。所述烯基为例如-CH=CH 2、-CH 2CH=CH 2、-C(CH 3)=CH 2、-CH 2-CH=CH-CH 3、2-戊烯基、3-戊烯基、4-戊烯基、2-己烯基、3-己烯基、4-己烯基、5-己烯基、2-甲基-2-丙烯基和4-甲基-3-戊烯基。当本发明的化合物含有烯基时,所述化合物可以纯E(异侧(entgegen))形式、纯Z(同侧(zusammen))形式或其任意混合物形式存在。术语“亚烯基”为相应的二价基团,包括例如“C 2-6亚烯基”、“C 2-4亚烯基”等,其具体实例包括但不限于:-CH=CH-、-CH 2CH=CH-、-C(CH 3)=CH-、亚丁烯基、亚戊烯基、亚己烯基、亚环戊烯基、亚环己烯基等。
如本文中所使用,术语“炔基”表示包含一个或多个三键的单价烃基,其优选具有2、3、4、5或6个碳原子,例如乙炔基、2-丙炔基、2-丁炔基、1,3-丁二炔基等。所述炔基任选地被一个或多个(诸如1至3个)相同或不同的取代基取代。术语“亚炔基”为相应的二价基团,包括例如“C 2-8亚炔基”、“C 2-6亚炔基”、“C 2-4亚炔基”等。其实例包括但不限于
Figure PCTCN2020074696-appb-000005
Figure PCTCN2020074696-appb-000006
等,所述亚炔基任选地被一个或多个(诸如1至3个)相同或不同的取代基取代。
如本文中所使用,术语“并环”或“稠环”指由两个或两个以上环状结构彼此共用两个相邻的原子所形成的环系。
如本文中所使用,术语“螺环”指由两个或两个以上环状结构彼此共用一个环原子所形成的环系。
如本文中所使用,术语“桥环”指由两个或两个以上环状结构彼此共用两个不直接相连的原子所形成的环系。
如本文中所使用,术语“环烷基”指饱和或不饱和的非芳族单环或多环(诸如双环)烃环基,包括但不限于单环烷基(诸如环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环壬基等)和双环烷基,包括螺环、并环(稠环)或桥环系统(即,螺环烷基、并环(稠环)烷基和桥环烷基,诸如双环[1.1.1]戊基、双环[2.2.1]庚基等)。本发明中,环烷基任选地被一个或多个(诸如1至3个)相同或不同的取代基取代。环烷基上的碳原子任选地被氧代(oxo)基团取代(即形成C=O)。术语“C 3-8环烷基”指具有3至8个成环碳原子的环烷基,例如C 3-6环烷基,其可以是单环烷基,例如环丙基、环丁基、环戊基、环己基、环庚基或环辛基,也可以是双环烷基,例如C 5-8螺环烷基、C 5-8桥环烷基、C 5-8稠环烷基、C 5-6螺环烷基、C 5-6桥环烷基或C 5-6稠环烷基。
如本文中所使用,术语“环烷氧基”意指-O-环烷基,其中环烷基如上文所定义。环烷氧基的代表性实例包括但不限于环丙氧基、环丁氧基、环戊氧基、环己氧基等。
如本文中所使用,术语“杂环基”或“杂环”指具有2个或2个以上(例如3、4、5、6、7、8、9、10、11、12、13或14个)碳原子,以及一个或多个(例如1个、2个、3个或4个)杂原子的单环或多环(例如并环、螺环或桥环)基团,所述杂原子包括但不限于氧原子、氮原子和硫原子,所述杂环基上的碳原子和杂原子任选地被氧代基团取代(例如形成C=O、S(=O)或S(=O) 2)。
如本文中所使用,术语“4-11元杂环基”意指含有4-11个环原子的杂环基,包括但不限于4-10元杂环基、4-9元杂环基、4-8元杂环基、4-7元杂环基、5-6元杂环基、3-8元杂环基、3-7元杂环基、4-7元含氮杂环基、4-7元含氧杂环基、4-7元含硫杂环基、5-6元含氮杂环基、5-6元含氧杂环基、5-6元含硫杂环基等,所述“含氮杂环基”、“含氧杂环基”和“含硫杂环基”各自任选地还含有一个或多个选自氧、氮和硫的其他杂原子。4-11元杂环基的实例包括但不限于环氧乙烷基、氮丙啶基、氮杂环丁基、氧杂环丁基、四氢呋喃基、吡咯烷基、吡咯烷酮基(如
Figure PCTCN2020074696-appb-000007
)、咪唑烷基、吡唑烷基、四氢吡喃基、哌啶基、吗啉基、二噻烷基(dithianyl)、硫吗啉基、哌嗪基、三噻烷基(trithianyl)。
如本文中所使用,术语“杂环基”涵盖并环结构,所述并环结构与其他基团的连接点可以在并环结构中的任一环上。因此,本发明的杂环基还包括但不限于杂环基并杂环基、杂环基并环烷基、单杂环基并单杂环基、单杂环基并单环烷基,例如3-7元(单)杂环基并3-7元(单)杂环基、3-7元(单)杂环基并(单)环烷基、3-7元(单)杂环基并C 4-6(单)环烷基,其实例包括但不限于吡咯烷基并环丙基、环戊基并氮杂环丙基、吡咯烷基并环丁基、吡咯烷基并吡咯烷基、吡咯烷基并哌啶基、吡咯烷基并哌嗪基、哌啶基并吗啉基、
Figure PCTCN2020074696-appb-000008
如本文中所使用,术语“杂环基”涵盖桥杂环基和螺杂环基。
如本文中所使用,术语“桥杂环”是指两个饱和环共用两个不直接相连的环原子形成的含有一个或多个(例如1个、2个、3个或4个)杂原子(例如氧原子、氮原子和/或硫原子)的环状结构,包括但不限于7-10元桥杂环、8-10元桥杂环、7-10元含氮桥杂环、7-10元含氧桥杂环、7-10元含硫桥杂环等,例如
Figure PCTCN2020074696-appb-000009
Figure PCTCN2020074696-appb-000010
等。所述“含氮桥杂环”、“含氧桥杂环”、“含硫桥杂环”任选地还含有一个或多个选自氧、氮和硫的其他杂原子。
如本文中所使用,术语“螺杂环”是指由两个或两个以上饱和环共用一个环原子形成的含有一个或多个(例如1个、2个、3个或4个)杂原子(例如氧原子、氮原子、硫原子)的环状结构,包括但不限于5-10元螺杂环、6-10元螺杂环、6-10元含氮螺杂环、6-10元含氧螺杂环、6-10元含硫螺杂环等,例如
Figure PCTCN2020074696-appb-000011
Figure PCTCN2020074696-appb-000012
Figure PCTCN2020074696-appb-000013
所述“含氮螺杂环”、“含氧螺杂环”、“含硫螺杂环”任选地还含有一个或多个选自氧、氮、硫的其他杂原子。术语“6-10元含氮螺杂环基”是指含有共计6-10个环原子并且其中至少一个环原子为氮原子的螺杂环基。
在本发明中,杂环基可以与芳基稠合形成稠环结构,所述稠环结构的实例包括但不限于:
Figure PCTCN2020074696-appb-000014
如本文中所使用,术语“芳基”或“芳环”指具有共轭π电子系统的全碳单环或稠合多环芳族基团。如本文中所使用,术语“C 6-12芳基(芳环)”意指含有6至12个碳原子的芳基(芳环),优选为C 6-10芳基(芳环),优选为苯基或萘基。芳基任选地被一个或多个(诸如1至3个)相同或不同的取代基(例如卤素、OH、CN、NO 2、C 1-C 6烷基等)取代。
如本文中所使用,术语“杂芳基”或“杂芳环”指含有一个或多个相同或不同杂原子的单环或多环芳族基团,包括单环的杂芳基和含有至少一个杂芳环(至少含有一个杂原子的芳族环系)的双环或多环环系,其可以具有5、6、7、8、9、10、11、12、13或14个环原子,例如5、6、7、8、9或10个环原子。所述杂原子可以是氧、氮或硫。所述杂芳基上的碳原子和杂原子任选地被氧代基团取代(例如形成C=O、S(=O)或S(=O) 2)。
如本文中所使用,术语“5-10元杂芳基”或“5-10元杂芳环”意指含有5至10个(例如5至6个)环原子的杂芳基(杂芳环),包括5-10元含氮杂芳基、5-10元含氧杂芳基、5-10元含硫杂芳基、5-6元含 氮杂芳基、5-6元含氧杂芳基、5-6元含硫杂芳基等。所述“含氮杂芳基”、“含氧杂芳基”和“含硫杂芳基”各自任选地含有一个或多个选自氧、氮和硫的其他杂原子。其实例包括但不限于噻吩基、呋喃基、吡咯基、噁唑基、噻唑基、咪唑基、吡唑基、异噁唑基、异噻唑基、三唑基、四唑基、噁二唑基、噻二唑基等,或吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基等,以及包含这些基团的5-10元并环基团。
如本文中所使用,术语“杂芳基”涵盖并环结构,所述并环结构与其他基团的连接点可以在并环结构中的任一环上。因此,本发明的杂芳基还包括但不限于(单)杂芳基并(单)杂芳基、(单)杂芳基并(单环)芳基、(单)杂芳基并(单)杂环基和(单)杂芳基并(单)环烷基,例如5-6元(单)杂芳基并5-6元(单)杂芳基、5-6元(单)杂芳基并苯基、5-6元(单)杂芳基并5-6元(单)杂环基或5-6元(单)杂芳基并C 4-6(单)环烷基(例如5-6元杂芳基并环丁基、5-6元杂芳基并环戊基或5-6元杂芳基并环己基),其实例包括但不限于吲哚基、异吲哚基、吲唑基、苯并咪唑、喹啉基、异喹啉基、
Figure PCTCN2020074696-appb-000015
Figure PCTCN2020074696-appb-000016
等。
如本文中所使用,术语“卤代”或“卤素”基团定义为包括F、Cl、Br或I。
术语“取代”指所指定的原子上的一个或多个(例如一个、两个、三个或四个)氢被从所指出的基团的选择代替,条件是未超过所指定的原子在当前情况下的正常原子价并且所述取代形成稳定的化合物。取代基和/或变量的组合仅仅当这种组合形成稳定的化合物时才是允许的。
如果取代基被描述为“任选地……被取代”,则取代基可(1)未被取代或(2)被取代。如果取代基的碳被描述为任选地被取代基列表中的一个或多个取代,则碳上的一个或多个氢(至存在的任何氢的程度)可单独和/或一起被独立地选择的任选的取代基替代。如果取代基的氮被描述为任选地被取代基列表中的一个或多个取代,则氮上的一个或多个氢(至存在的任何氢的程度)可各自被独立地选择的任选的取代基替代。
如果取代基被描述为“独立地选自”一组,则各取代基独立于另一者被选择。因此,各取代基可与另一(其他)取代基相同或不同。
如本文中所使用,术语“一个或多个”意指在合理条件下的1个或超过1个,例如2个、3个、4个、5个或10个。
除非指明,否则如本文中所使用,取代基的连接点可来自取代基的任意适宜位置。
当取代基的键显示为穿过环中连接两个原子的键时,则这样的取代基可键连至该可取代的环中的任一成环原子。
本发明还包括所有药学上可接受的同位素标记的化合物,其与本发明的化合物相同,除了一个或多个原子被具有相同原子序数但原子质量或质量数不同于在自然界中占优势的原子质量或质量数的原子替代。适合包含于本发明的化合物中的同位素的实例包括(但不限于)氢的同位素(例如氘( 2H)、氚( 3H));碳的同位素(例如 11C、 13C及 14C);氯的同位素(例如 36Cl);氟的同位素(例如 18F);碘的同位素(例如 123I及 125I);氮的同位素(例如 13N及 15N);氧的同位素(例如 15O、 17O及 18O);磷的同位素(例如 32P);及硫的同位素(例如 35S)。某些同位素标记的本发明的化合物(例如掺入放射性同位素的那些)可用于药物和/或底物组织分布研究(例如分析)中。放射性同位素氚(即 3H)及碳-14(即 14C)因易于掺入且容易检测而特别可用于该目的。用正电子发射同位素(例如 11C、 18F、 15O及 13N)进行取代可在正电子发射断层显像术(PET)研究中用于检验底物受体占据情况。被同位素标记的本发明的化合物可通过与描述于随附路线和/或实施例及制备中的那些类似的方法通过使用适当的被同位素标记的试剂代替之前采用的非标记的试剂来制备。本发明的药学上可接受的溶剂合物包括其中结晶溶剂可被同位素取代的那些,例如,D 2O、丙酮-d 6或DMSO-d 6
术语“立体异构体”表示由于至少一个不对称中心形成的异构体。在具有一个或多个(例如一个、两个、三个或四个)不对称中心的化合物中,其可产生外/内消旋混合物、单一对映异构体、非对映异构体混合物和单独的非对映异构体。特定个别分子也可以几何异构体(顺式/反式)存在。类似地,本发明的化合物可以两种或更多种处于快速平衡的结构不同的形式的混合物(通常称作互变异构体)存在。互变异构体的代表性实例包括酮-烯醇互变异构体、苯酚-酮互变异构体、亚硝基-肟互变异构体、亚胺-烯胺互变异构体等。例如,亚硝基-肟在溶液中可以下列互变异构形式平衡存在:
Figure PCTCN2020074696-appb-000017
要理解,本申请的范围涵盖所有这样的以任意比例(例如60%、65%、70%、75%、80%、85%、90%、95%、96%、97%、98%、99%)的异构体或其混合物。
本文中可使用实线
Figure PCTCN2020074696-appb-000018
实楔形
Figure PCTCN2020074696-appb-000019
或虚楔形
Figure PCTCN2020074696-appb-000020
描绘本发明的化合物的化学键。使用实线以描绘键连至不对称碳原子的键欲表明,包括该碳原子处的所有可能的立体异构体(例如,特定的对映异构体、外消旋混合物等)。使用实或虚楔形以描绘键连至不对称碳原子的键欲表明,存在所示的立体异构体。当存在于外消旋混合物中时,使用实及虚楔形以定义相对立体化学,而非绝对立体化学。除非另外指明,否则本发明的化合物意欲可以立体异构体(其包括顺式及反式异构体、光学异构体(例如R及S对映异构体)、非对映异构体、几何异构体、旋转异构体、构象异构体、阻转异构体及其混合物)的形式存在。本发明的化合物可表现一种以上类型的异构现象,且由其混合物(例如外消旋混合物及非对映异构体对)组成。
本发明涵盖本发明的化合物的所有可能的结晶形式或多晶型物,其可为单一多晶型物或多于一种多晶型物的任意比例的混合物。
共晶是指药物活性分子与其它生理上可接受的酸、碱、盐、非离子化合物分子以氢键、π-π堆积作用、范德华力和其它非共价键相连而结合在同一晶格中。
还应当理解,本发明的某些化合物可以游离形式存在用于治疗,或适当时,以其药学上可接受的衍生物形式存在。在本发明中,药学上可接受的衍生物包括但不限于,药学上可接受的盐、酯、溶剂合物、N-氧化物、代谢物或前药,在将它们向需要其的患者给药后,能够直接或间接提供本发明的化合物或其代谢物或残余物。因此,当在本文中提及“本发明的化合物”时,也意在涵盖化合物的上述各种衍生物形式。
本发明的化合物的药学上可接受的盐包括其酸加成盐及碱加成盐。例如六氟磷酸盐、葡甲胺盐等。适合的盐的综述参见Stahl及Wermuth的“Handbook of Pharmaceutical Salts:Properties,Selection,and Use”(Wiley-VCH,2002)。
如本文中所使用,术语“酯”意指衍生自本申请中各个通式化合物的酯,其包括生理上可水解的酯(可在生理条件下水解以释放游离酸或醇形式的本发明的化合物)。本发明的化合物本身也可以是酯。
本发明的化合物可以溶剂合物(优选水合物)的形式存在,其中本发明的化合物包含作为所述化合物晶格的结构要素的极性溶剂,特别是例如水、甲醇或乙醇。极性溶剂特别是水的量可以化学计量比或非化学计量比存在。
本领域技术人员会理解,由于氮需要可用的孤对电子来氧化成氧化物,因此并非所有的含氮杂环都能够形成N-氧化物。本领域技术人员会识别能够形成N-氧化物的含氮杂环。本领域技术人员还会认识到叔胺能够形成N-氧化物。用于制备杂环和叔胺的N-氧化物的合成方法是本领域技术人员熟知的,包括但不限于用过氧酸如过氧乙酸和间氯过氧苯甲酸(MCPBA)、过氧化氢、烷基过氧化氢如叔丁基过氧化氢、过硼酸钠和双环氧乙烷(dioxirane)如二甲基双环氧乙烷来氧化杂环和叔胺。这些用于制备N-氧化物的方法已在文献中得到广泛描述和综述,参见例如:T.L.Gilchrist,Comprehensive Organic Synthesis,vol.7,pp 748-750;A.R.Katritzky和A.J.Boulton,Eds.,Academic Press;以及G.W.H.Cheeseman和E.S.G.Werstiuk,Advances in Heterocyclic Chemistry,vol.22,pp 390-392,A.R.Katritzky和A.J.Boulton,Eds.,Academic Press。
在本发明的范围内还包括本发明的化合物的代谢物,即在给药本发明的化合物时体内形成的物质。这样的产物可由例如被给药的化合物的氧化、还原、水解、酰胺化、脱酰胺化、酯化、酶解等产生。因此,本发明包括本发明的化合物的代谢物,包括通过使本发明的化合物与哺乳动物接触足以产生其代谢产物的时间的方法制得的化合物。
本发明在其范围内进一步包括本发明的化合物的前药,其为自身可具有较小药理学活性或无药理学活性的本发明的化合物的某些衍生物当被给药至身体中或其上时可通过例如水解裂解转化成具有期望活性的本发明的化合物。通常这样的前药会是所述化合物的官能团衍生物,其易于在体内转化成期望的治疗活性化合物。关于前药的使用的其他信息可参见“Pro-drugs as Novel Delivery Systems”,第14卷,ACS Symposium Series(T.Higuchi及V.Stella)。本发明的前药可例如通过用本领域技术人员已知作为“前-部分(pro-moiety)(例如“Design of Prodrugs”,H.Bundgaard(Elsevier,1985)中所述)”的某些部分替代本发明的化合物中存在的适当官能团来制备。
本发明还涵盖含有保护基的本发明的化合物。在制备本发明的化合物的任何过程中,保护在任 何有关分子上的敏感基团或反应基团可能是必需的和/或期望的,由此形成本发明的化合物的化学保护的形式。这可以通过常规的保护基实现,例如,在T.W.Greene&P.G.M.Wuts,Protective Groups in Organic Synthesis,John Wiley&Sons,1991中所述的那些保护基,这些参考文献通过援引加入本文。使用本领域已知的方法,在适当的后续阶段可以移除保护基。
术语“约”是指在所述数值的±10%范围内,优选±5%范围内,更优选±2%范围内。
化合物
本发明的一个方面提供式I的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药:
Figure PCTCN2020074696-appb-000021
其中:
环A选自C 6-10芳环和5-6元杂芳环;
环B选自C 3-8环烷基和4-11元杂环基;
X 1选自CH和N;
R 1选自H、卤素、羟基、氰基、C 1-6烷基、C 1-6杂烷基(例如C 1-6烷氧基)、C 3-8环烷基、4-10元杂环基和-NR 20aR 20b,所述烷基、杂烷基(例如烷氧基)、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基和C 1-4杂烷基(例如C 1-4烷氧基);
R 2选自C 1-6烷基、C 1-6杂烷基、C 3-8环烷基、4-10元杂环基、5-10元杂芳基和-C(=O)R 21,所述烷基、杂烷基、环烷基、杂环基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基和C 3-6环烷基;
R 3和R 4不存在或者在每次出现时各自独立地选自羟基、卤素、CN、C 1-6烷基、C 1-6杂烷基(例如C 1-6烷氧基)和C 3-6环烷基,所述烷基、杂烷基(例如烷氧基)和环烷基各自任选地被一个或多个选自下列的取代基取代:卤素、CN、C 1-4烷基、C 1-4卤代烷基、C 1-4烷氧基和C 1-4卤代烷氧基;当m大于1时,两个R 3任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基;和/或当n大于1时,两个R 4任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基;
L选自-O-、-S-、-S(O)-、-S(O) 2-、-N=CR 21-、-N(R 23a)-C(O)-、C 1-6亚烷基、C 1-6亚杂烷基、C 2-6亚烯基、C 2-6亚炔基、
Figure PCTCN2020074696-appb-000022
Figure PCTCN2020074696-appb-000023
Figure PCTCN2020074696-appb-000024
所述亚烷基、亚杂烷基、亚烯基和亚炔基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6卤代烷氧基、C 1-6杂烷基(例如C 1-6烷氧基)和C 3-8环烷基;或者L为-N(R 23a)-;
R 5选自羟基、卤素、CN、NO 2、C 1-6烷基、C 1-6杂烷基(例如C 1-6烷氧基)、C 2-6烯基、C 2-6炔基、C 3-8环烷基、C 3-8环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 20aR 20b、-OR 21、-SR 21、-S(=O)R 22、-S(=O) 2R 22、-S(=O)NR 20aR 20b、-S(=O) 2NR 20aR 20b、-NR 20aS(=O)R 20b、-NR 20aS(=O) 2R 20b、-C(=O)R 21、-C(=O)NR 23aR 23b、-NR 23aC(=O)R 23b、-OC(=O)NR 23aR 23b和-NR 24aC(=O)NR 25aR 25b,所述烷基、杂烷基(例如烷氧基)、烯基、炔基、环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基、C 3-6环烷氧基和4-10元杂环基;
R 20a、R 20b、R 23a、R 23b、R 23c、R 24a、R 25a和R 25b各自独立地选自H、OH、C 1-6烷基、C 1-6烷氧基和C 3-8环烷基;或者R 20a与R 20b、R 23a与R 23b或R 25a与R 25b连同其所连接的原子一起形成3-8元环烷基或杂环基,所述烷基、烷氧基、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:OH、CN、卤素、NO 2、C 1-4烷基、C 1-4烷氧基、C 1-4羟烷基、C 1-4卤代烷基和C 1-4卤代烷氧基;
R 30a、R 30b、R 33a、R 33b、R 34a、R 35a和R 35b各自独立地选自H、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基和C 1-6卤代烷氧基;
R 21、R 22、R 31和R 32各自独立地选自C 1-6烷基、C 1-6烷氧基、C 3-8环烷基、4-10元杂环基、C 6-12芳基和5-10元杂芳基,所述烷基、烷氧基、环烷基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:OH、卤素、CN、C 1-4烷基、C 1-4烷氧基、C 1-4卤代烷基、C 1-4卤代烷氧基、C 3-6环烷基和4-10元杂环基;
m为0、1、2、3或4;
n为0、1、2、3或4;
t为0、1、2、3或4;且
u为0、1、2、3或4;
条件是,当环B为哌嗪环且X 1为CH时,R 2不是4-CF 3-吡啶-2-基或4-CN-吡啶-2-基。
在某些实施方案中,环A为苯环或5-6元杂芳环;优选地,环A为苯环、噻唑环、吡啶环、吡嗪环或嘧啶环;更优选地,环A为
Figure PCTCN2020074696-appb-000025
其通过*标记的位置与X 1所在环连接,并且通过**标记的位置与环B连接。
在某些实施方案中,环B为C 3-6环烷基或5-7元杂环基;优选地,环B为哌啶环、哌嗪环、氮杂环庚烷桥环或二氮杂环庚烷桥环;更优选地,环B为
Figure PCTCN2020074696-appb-000026
其通过*标记的位置与环A连接,并且通过**标记的位置与L连接。
在某些实施方案中,X 1为CH或N;优选地,X 1为N。
在某些实施方案中,R 1选自H、卤素、羟基、氰基、C 1-4烷基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基,所述烷基、杂烷基(例如烷氧基)、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基和C 1-4杂烷基(例如C 1-4烷氧基)。
在某些实施方案中,R 1选自C 1-4烷基、5元含氮杂环基和C 1-4杂烷基(例如C 1-4烷氧基),所述烷 基、杂环基和杂烷基(例如烷氧基)各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基和C 1-3杂烷基(例如C 1-4烷氧基)。
在某些实施方案中,R 1选自C 1-3烷基(例如甲基)、吡咯烷基(例如吡咯烷-1-基)和C 1-3烷氧基(例如乙氧基)。
在某些实施方案中,R 2选自C 1-4烷基、C 1-4杂烷基、C 3-6环烷基、4-6元杂环基、5-6元杂芳基和-C(=O)R 21,所述烷基、杂烷基、环烷基、杂环基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基和C 3-6环烷基。
在某些实施方案中,R 2选自C 1-3烷基、5-6元杂芳基和-C(=O)CH 3,所述烷基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3杂烷基和C 3-6环烷基。
在某些实施方案中,R 2选自C 1-3烷基(例如甲基)、-C(=O)CH 3、噻吩基、吡咯基、吡唑基、咪唑基、噻唑基、噻二唑基、异噻唑基、噁唑基、噁二唑基、异噁唑基和吡啶基,所述烷基、噻吩基、吡咯基、吡唑基、咪唑基、噻唑基、噻二唑基、异噻唑基、噁唑基、噁二唑基、异噁唑基和吡啶基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、C 1-3烷基(例如甲基)、C 1-3卤代烷基、C 1-3卤代烷氧基、C 1-3杂烷基(例如C 1-3烷氧基)和C 3-6环烷基;优选地,R 2为甲基取代的吡唑基(例如5-甲基-1H-吡唑-3-基或1-甲基-1H-吡唑-4-基)、环丙基取代的吡唑基(例如5-环丙基-1H-吡唑-3-基)或-C(O)CH 3
在某些实施方案中,R 3和R 4不存在或者在每次出现时独立地选自羟基、卤素、CN、C 1-4烷基和C 1-4烷氧基,所述烷基和烷氧基各自任选地被一个或多个选自下列的取代基取代:卤素、CN、C 1-4烷基、C 1-4卤代烷基、C 1-4烷氧基和C 1-4卤代烷氧基;当m大于1时,两个R 3任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基;和/或当n大于1时,两个R 4任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基。
在某些实施方案中,R 3和R 4不存在或者在每次出现时独立地选自羟基、卤素、CN、C 1-3烷基和C 1-3烷氧基,所述烷基和烷氧基各自任选地被一个或多个选自下列的取代基取代:卤素、CN和C 1-3烷基;当m大于1时,两个R 3任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基;和/或当n大于1时,两个R 4任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基。
在某些实施方案中,R 3和R 4不存在或者在每次出现时独立地选自:F、Cl、CN、OH、C 1-3烷基和C 1-3烷氧基;优选地,R 3和R 4不存在。
在某些实施方案中,L选自-O-、-S-、-C(O)-、-N(R 23a)-C(O)-、-C(O)-N(R 23c)-、C 1-4亚烷基、C 1- 4亚杂烷基、
Figure PCTCN2020074696-appb-000027
Figure PCTCN2020074696-appb-000028
所述亚烷基和亚杂烷基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)和C 3-6环烷基。
在某些实施方案中,L选自-O-、-C(O)-、-NHC(O)-、-C(O)NH-、C 1-3亚烷基、C 1-3亚杂烷基、
Figure PCTCN2020074696-appb-000029
所述亚烷基和亚杂烷基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3杂烷基(例如C 1-3烷氧基)和C 3-6环烷基,其中R 23a和R 23b优选为H或C 1-3烷基。
在某些实施方案中,L选自-O-、-C(O)-、-NHC(O)-、-C(O)NH-、C 1-3亚烷基、
Figure PCTCN2020074696-appb-000030
Figure PCTCN2020074696-appb-000031
所述亚烷基任选地被一个或多个选自 下列的取代基取代:羟基、卤素、CN、C 1-3烷基和C 1-3卤代烷基。优选地,L为-CH 2-、-CH(CH 3)-、-O-、-C(O)-、
Figure PCTCN2020074696-appb-000032
-C(O)NH-或
Figure PCTCN2020074696-appb-000033
在某些实施方案中,R 5选自羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 20aR 20b、-OR 21、-SR 21、-S(=O)R 22、-S(=O) 2R 22、-S(=O)NR 20aR 20b、-S(=O) 2NR 20aR 20b、-NR 20aS(=O)R 20b、-NR 20aS(=O) 2R 20b、-C(=O)R 21、-C(=O)NR 23aR 23b、-NR 23aC(=O)R 23b、-OC(=O)NR 23aR 23b和-NR 24aC(=O)NR 25aR 25b,所述烷基、杂烷基(例如烷氧基)、烯基、炔基、环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1- 4烷氧基)、C 3-6环烷基、C 3-6环烷氧基和4-10元杂环基。
在某些实施方案中,R 5选自C 3-6环烷基、4-10元杂环基、C 6-12芳基和5-10元杂芳基,所述环烷基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1- 4烷氧基)、C 3-6环烷基、C 3-6环烷氧基和4-10元杂环基。
在某些实施方案中,R 5选自C 6-10芳基和5-6元杂芳基,所述芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3杂烷基(例如C 1-3烷氧基)、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-C(=O)R 31、-C(=O)NR 33aR 33b和-NR 33aC(=O)R 33b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基、C 3-6环烷氧基和4-6元杂环基。
在某些实施方案中,R 5选自苯基和5-6元杂芳基(例如吡啶基、嘧啶基、吡嗪基、哒嗪基、吡唑基、噁唑基、咪唑基或噻唑基),所述苯基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3杂烷基(例如C 1-3烷氧基)、C 3-6环烷基、C 3-6环烷氧基、4-6元杂环基、5-8元杂芳基(例如吡啶基、吡咯基、吡唑基、呋喃基、噁唑基、咪唑基、噻唑基或环戊基并吡唑基)、-NR 30aR 30b、-OR 31、-C(=O)R 31、-C(=O)NR 33aR 33b和-NR 33aC(=O)R 33b,其中所述环烷基、环烷氧基、杂环基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3杂烷基(例如C 1-3烷氧基)、C 3-6环烷基、C 3-6环烷氧基和4-6元杂环基。
在某些实施方案中,R 5选自苯基、吡啶基、吡唑基和噻唑基,所述苯基、吡啶基、吡唑基和噻唑基各自任选地被一个或多个选自下列的取代基取代:卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3烷氧基、C 3-6环烷基、C 3-6环烷氧基、4-6元杂环基、5-8元杂芳基(例如吡啶基、吡咯基、吡唑基、呋喃基、噁唑基、咪唑基、噻唑基或环戊基并吡唑基)、-NR 30aR 30b和-OR 31,其中所述杂环基和杂芳基各自任选地被一个或多个选自下列的取代基取代:卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3烷氧基、C 3-6环烷基、C 3-6环烷氧基和4-6元杂环基。优选地,R 5为任选地被一个或多个选自卤素(例如氟或氯)、CN、C 1-3烷基(例如甲基或乙基)、C 1- 3卤代烷基(例如三氟甲基)、C 1-3烷氧基(例如甲氧基或乙氧基)、C 3-6环烷基(例如环丙基)、C 3-6环烷氧基(例如环丙氧基)和5-6元杂芳基(例如吡啶基、吡咯基、吡唑基、呋喃基、噁唑基、咪唑基或噻唑基)的取代基取代的苯基、吡啶基、吡唑基或噻唑基,其中所述5-6元杂芳基任选地进一步被一个或多个选自卤素(例如氟或氯)、C 1-3烷基(例如甲基、乙基或异丙基)、C 1-3卤代烷基(例如氟甲基)、C 1-3羟烷基(例如羟甲基或羟丙基)、C 1-3烷氧基(例如甲氧基)、C 3-6环烷基(例如环丙基)、C 3-6环烷氧基(例如 环丙氧基或环丁氧基)的取代基取代。
在某些实施方案中,R 5选自苯基、吡啶基、吡唑基和噻唑基,所述苯基、吡啶基、吡唑基和噻唑基各自任选地被一个或多个选自下列的取代基取代:卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3烷氧基、C 3-6环烷基、C 3-6环烷氧基、4-6元杂环基、5-6元杂芳基(例如吡啶基、吡咯基、呋喃基、吡唑基、噁唑基、咪唑基或噻唑基)、-NR 30aR 30b和-OR 31,其中所述杂环基和杂芳基各自任选地被一个或多个选自下列的取代基取代:卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3卤代烷氧基、C 1-3烷氧基、C 3-6环烷基和4-6元杂环基。优选地,R 5为任选地被一个或多个选自卤素(例如氟或氯)、CN、C 1-3烷基(例如甲基或乙基)、C 1-3卤代烷基(例如三氟甲基)、C 1-3烷氧基(例如甲氧基或乙氧基)、C 3-6环烷基(例如环丙基)、C 3-6环烷氧基(例如环丙氧基)和五元杂芳基(例如吡唑基、咪唑基或噻唑基)的取代基取代的苯基、吡啶基、吡唑基或噻唑基,其中所述五元杂芳基任选地进一步被一个或多个选自卤素(例如氟或氯)、C 1-3烷基(例如甲基)、C 1-3羟烷基(例如羟甲基或羟丙基)的取代基取代。
在某些实施方案中,R 20a、R 20b、R 23a、R 23b、R 23c、R 24a、R 25a和R 25b各自独立地选自H、C 1-4烷基、C 1-4烷氧基和C 3-8环烷基;或者R 20a与R 20b、R 23a与R 23b或R 25a与R 25b连同其所连接的原子一起形成3-8元环烷基或杂环基,所述烷基、烷氧基、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:OH、CN、卤素、NO 2、C 1-4烷基、C 1-4烷氧基、C 1-4羟烷基、C 1-4卤代烷基和C 1-4卤代烷氧基。
在某些实施方案中,R 20a、R 20b、R 23a、R 23b、R 23c、R 24a、R 25a和R 25b各自独立地为H、C 1-4烷基或C 1-4烷氧基。
在某些实施方案中,R 23a和R 23b各自独立地选自H、C 1-3烷基、C 1-3烷氧基和C 3-6环烷基;或者R 23a与R 23b连同其所连接的C原子一起形成C 3-6环烷基或杂环基,所述烷基、烷氧基、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:卤素、C 1-3烷基、C 1-3烷氧基、C 1-3羟烷基、C 1-3卤代烷基和C 1-3卤代烷氧基。
在某些实施方案中,R 21、R 22、R 31和R 32各自独立地选自C 1-4烷基、C 1-4烷氧基、C 3-8环烷基和4-10元杂环基,所述烷基、烷氧基、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:OH、卤素、CN、C 1-4烷基、C 1-4烷氧基、C 1-4卤代烷基、C 1-4卤代烷氧基、C 3-6环烷基和4-10元杂环基。
在某些实施方案中,R 21、R 22、R 31和R 32各自独立地选自C 1-4烷基。
在某些实施方案中,R 30a、R 30b、R 33a、R 33b、R 34a、R 35a和R 35b各自独立地选自H、C 1-4烷基、C 1- 4卤代烷基、C 1-4羟烷基、C 1-4烷氧基和C 1-4卤代烷氧基;
在某些实施方案中,R 30a、R 30b、R 33a、R 33b、R 34a、R 35a和R 35b各自独立地选自H和C 1-4烷基。
在某些实施方案中,m为0。
在某些实施方案中,n为0、1或2。
在某些实施方案中,t为0或1。
在某些实施方案中,u为0或1。
在一些实施方案中,本发明的化合物具有式I-A所示的结构:
Figure PCTCN2020074696-appb-000034
其中:
R 1和R 2如上文式I所定义;
R 5选自C 6-12芳基和5-10元杂芳基,其中(1)所述C 6-12芳基任选地被一个或多个选自下列的取代 基取代:C 3-6环烷氧基、C 6-12芳基、5-10元杂芳基、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷氧基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基,且(2)所述5-10元杂芳基任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基C 3-6环烷氧基和4-10元杂环基;且
R 23a、R 30a、R 30b、R 31、R 32、R 33a、R 33b、R 34a、R 35a和R 35b如上文式I所定义,且R 23a优选为H或C 1-3烷基。
在一些实施方案中,R 5选自C 6-12芳基和5-10元杂芳基,其中(1)所述C 6-12芳基任选地被一个或多个选自下列的取代基取代:C 3-6环烷氧基、C 6-12芳基、5-10元杂芳基、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷氧基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基,且(2)所述5-10元杂芳基任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1- 4烷氧基)、C 3-6环烷基和4-10元杂环基;且
R 23a、R 30a、R 30b、R 31、R 32、R 33a、R 33b、R 34a、R 35a和R 35b如上文式I所定义,且R 23a优选为H或C 1-3烷基。
在一些实施方案中,本发明的化合物具有式I-B所示的结构:
Figure PCTCN2020074696-appb-000035
其中:
R 1、R 2、R 5和R 23a如上文式I所定义,且R 23a优选为H或C 1-3烷基。
在一些实施方案中,本发明的化合物具有式I-C所示的结构:
Figure PCTCN2020074696-appb-000036
其中:
当X 1为CH时,R 1、R 2、R 5和R 23a如上文式I所定义,且R 23a优选为H或C 1-3烷基;而当X 1为N时,R 1、R 2、R 5和R 23a如上文式I-A所定义。
在一些实施方案中,本发明的化合物具有式I-D所示的结构:
Figure PCTCN2020074696-appb-000037
其中:
R 1、R 2、R 23a、R 23b和t如上文式I所定义;
当X 1为CH时,R 5如上文式I所定义;且
当X 1为N时,R 5为C 6-12芳基或5-10元杂芳基,其中
(i)当t为0时,所述C 6-12芳基和5-10元杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基,
(ii)当t为1时,(1)所述C 6-12芳基任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基,且(2)所述5-10元杂芳基任选地被一个或多个选自下列的取代基取代:NO 2、C 2-6烯基、C 2-6炔基、C 3-6环烷氧基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基;且
R 30a、R 30b、R 31、R 32、R 33a、R 33b、R 34a、R 35a和R 35b如上文式I所定义。
在一些实施方案中,本发明的化合物具有式I-E所示的结构:
Figure PCTCN2020074696-appb-000038
其中:
R 1、R 2、R 5、R 23a、R 23b、X 1和t如上文式I-D所定义。
在一些实施方案中,本发明的化合物具有式I-F所示的结构:
Figure PCTCN2020074696-appb-000039
其中:
R 1、R 2、R 5、R 23a、R 23b和X 1如上文式I-D所定义;
R 4如上文式I所定义,且优选为C 1-3烷基或C 1-3烷氧基;
R 23c为H、C 1-3烷基或C 1-3烷氧基,所述烷基和烷氧基各自任选地被一个或多个选自下列的取代基取代:OH、CN、卤素、C 1-4烷氧基和C 1-4羟烷基;
u为0或1;且
n为0或1。
在一些实施方案中,本发明的化合物具有式I-G所示的结构:
Figure PCTCN2020074696-appb-000040
其中:
X 1为CH或N;
R 1、R 2和R 4如上文式I所定义,且R 4优选为C 1-3烷基或C 1-3烷氧基;
n为0或1;
R 5选自C 6-12芳基和5-10元杂芳基,其中(1)所述C 6-12芳基任选地被一个或多个选自下列的取代基取代:C 3-6环烷氧基、C 6-12芳基、5-10元杂芳基、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷氧基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤 代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基;且(2)所述5-10元杂芳基任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基;且
R 30a、R 30b、R 31、R 32、R 33a、R 33b、R 34a、R 35a和R 35b如上文式I所定义。
在一些实施方案中,本发明的化合物具有式I所示的结构,其中:
环A为
Figure PCTCN2020074696-appb-000041
其通过*标记的位置与X 1所在环连接,并且通过**标记的位置与环B连接;
环B为
Figure PCTCN2020074696-appb-000042
其通过*标记的位置与环A连接,并且通过**标记的位置与L连接;
X 1为N;
R 1选自C 1-3烷基(例如甲基)、吡咯烷基(例如吡咯烷-1-基)和C 1-3烷氧基(例如乙氧基);
R 2为甲基取代的吡唑基(例如5-甲基-1H-吡唑-3-基或1-甲基-1H-吡唑-4-基)、环丙基取代的吡唑基(例如5-环丙基-1H-吡唑-3-基)或-C(O)CH 3
R 3和R 4不存在;
L为-CH 2-、-CH(CH 3)-、-O-、-C(O)-、
Figure PCTCN2020074696-appb-000043
-C(O)NH-或
Figure PCTCN2020074696-appb-000044
R 5为任选地被一个或多个选自卤素(例如氟或氯)、CN、C 1-3烷基(例如甲基或乙基)、C 1-3卤代烷基(例如三氟甲基)、C 1-3烷氧基(例如甲氧基或乙氧基)、C 3-6环烷基(例如环丙基)、C 3-6环烷氧基(例如环丙氧基)和5-6元杂芳基(例如吡啶基、吡咯基、吡唑基、呋喃基、噁唑基、咪唑基或噻唑基)的取代基取代的苯基、吡啶基、吡唑基或噻唑基,其中所述5-6元杂芳基任选地进一步被一个或多个选自卤素(例如氟或氯)、C 1-3烷基(例如甲基、乙基或异丙基)、C 1-3卤代烷基(例如氟甲基)、C 1-3羟烷基(例如羟甲基或羟丙基)、C 1-3烷氧基(例如甲氧基)、C 3-6环烷基(例如环丙基)和C 3-6环烷氧基(例如环丙氧基或环丁氧基)的取代基取代。
本发明涵盖以上各实施方案的任意组合。
在一些实施方案中,本发明的化合物包括,但不限于:
Figure PCTCN2020074696-appb-000045
Figure PCTCN2020074696-appb-000046
Figure PCTCN2020074696-appb-000047
Figure PCTCN2020074696-appb-000048
Figure PCTCN2020074696-appb-000049
制备方法
在某些实施方案中,式I-A的化合物可由如下路线A所示的方法合成:
路线A
Figure PCTCN2020074696-appb-000050
其中:
Hal 1和Hal 2各自独立地为F、Cl、Br或I;优选地,Hal 1为F、Cl、Br或I,且Hal 2为Cl、Br或I;
R 1选自H、氰基、C 1-6烷基、C 1-6杂烷基(例如C 1-6烷氧基)、C 3-8环烷基、4-6元杂环基和-NR 20aR 20b,所述烷基、杂烷基(例如烷氧基)、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:卤素、CN、C 1-4烷基、C 1-4卤代烷基、C 1-4卤代烷氧基和C 1-4杂烷基(例如C 1-4烷氧基);
R 2选自C 1-6烷基、C 1-6杂烷基、C 3-8环烷基、4-6元杂环基和5-6元杂芳基,所述烷基、杂烷基、环烷基、杂环基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基和C 3-6环烷基;
R 23a选自H、C 1-6烷基、C 1-6烷氧基和C 3-8环烷基,所述烷基、烷氧基和环烷基各自任选地被一个或多个选自下列的取代基取代:OH、CN、卤素、C 1-4烷基、C 1-4烷氧基、C 1-4羟烷基、C 1-4卤代烷基和C 1-4卤代烷氧基;
R 20a和R 20b如上文式I所定义;且
R 5如上文式I-A所定义。
第一步:化合物I-A-1与R 2-NH 2在碱存在下经取代或偶联反应(例如Buchwald反应、Suzuki反应或Ullmann反应)生成化合物I-A-2。
对于取代反应,可使用的碱例如为 tBuONa、 tBuOK、 tBuOLi、Cs 2CO 3、DIPEA、LiHMDS、LDA、NaHMDS、KHMDS、K 3PO 4、Na 2CO 3、KOAc、NaHCO 3或K 2CO 3,可使用的溶剂例如为叔丁醇、甲苯、二甲苯、THF、DME、1,4-二氧六环、DMF、DMSO或NMP,且反应温度为40℃至140℃。
对于Buchwald反应,可使用的催化剂例如为Pd(OAc) 2、Pd 2(dba) 3、Pd(dba) 2、PdCl 2、Pd(PPh 3) 4、Pd(dppf)Cl 2、Pd(acac) 2或Pd(allyl) 2,可使用的配体为PPh 3、XPhos、SPhos、RuPhos、XantPhos、Dppf、BINOL、BINAP或Pcy 3等,可使用的碱例如为 tBuONa、 tBuOK、 tBuOLi、Cs 2CO 3、LiHMDS、LDA、NaHMDS、KHMDS、K 3PO 4、Na 2CO 3、KOAc、NaHCO 3或K 2CO 3,可使用的溶剂例如为甲苯、二甲苯、THF、DME、1,4-二氧六环、DMF、DMSO或NMP,且反应温度为40℃至140℃。
对于Suzuki反应,可使用的催化剂例如为Pd(PPh 3) 4或Pd(dppf)Cl 2,可使用的碱例如为Cs 2CO 3、K 3PO 4、Na 2CO 3、AcOK、NaHCO 3或K 2CO 3,可使用的溶剂例如为1,4-二氧六环/H 2O、DMF/H 2O、DMSO/H 2O或CH 3CN/H 2O,且反应温度为60℃至120℃;
对于Ullmann反应,可使用的催化剂例如为CuCl、CuBr、CuI或Cu 2O,可使用的配体例如为水杨醛肟、环己二胺、N,N’-二甲基乙二胺、TMEDA或乙二胺,可使用的碱例如为 tBuONa、 tBuOK、 tBuOLi、Cs 2CO 3、LiHMDS、LDA、NaHMDS、KHMDS、K 3PO 4、Na 2CO 3、KOAc、NaHCO 3或K 2CO 3,可使用的溶剂例如为甲苯、二甲苯、THF、DME、1,4-二氧六环、DMF、DMSO或NMP,且反应温度为40℃至140℃。
第二步:化合物I-A-3与I-A-4在碱存在下反应生成化合物I-A-5。
可使用的碱例如为 tBuONa、 tBuOK、 tBuOLi、Cs 2CO 3、DIPEA、LiHMDS、LDA、NaHMDS、KHMDS、K 3PO 4、Na 2CO 3、KOAc、NaHCO 3或K 2CO 3。可使用的溶剂例如为叔丁醇、甲苯、二甲苯、THF、DME、1,4-二氧六环、DMF、DMSO或NMP。反应温度为40℃至140℃。
第三步:化合物I-A-5与含硼试剂反应生成化合物I-A-6。
可使用的含硼试剂例如为B 2(pin) 2。可使用的催化剂例如为Pd(PPh 3) 4、Pd(dppf)Cl 2或Pd(dppf) 2Cl 2·DCM。可使用的碱例如为Cs 2CO 3、K 3PO 4、Na 2CO 3、KOAc、NaHCO 3或K 2CO 3。可使用的溶剂例如为1,4-二氧六环、DMF、DMSO或CH 3CN。反应温度为50℃至120℃。
第四步:化合物I-A-2与I-A-6经偶联反应(例如Suzuki反应)生成化合物I-A-7。
可使用的催化剂例如为Pd(PPh 3) 4、Pd(dppf)Cl 2或Pd(dppf) 2Cl 2·DCM。可使用的碱例如为Cs 2CO 3、 K 3PO 4、Na 2CO 3、KOAc、NaHCO 3或K 2CO 3。可使用的溶剂例如为1,4-二氧六环、DMF、DMSO或CH 3CN,或者为任意上述溶剂与H 2O的混合物。反应温度为50℃至120℃。
第五步:化合物I-A-7在酸性条件下脱除保护基生成化合物I-A-8。
可使用的酸例如为HCl的1,4-二氧六环溶液、HCl的EA溶液或TFA的DCM溶液。反应温度为0℃至80℃。
第六步:化合物I-A-8与I-A-9经还原胺化反应生成化合物I-A。
可使用的溶剂例如为甲醇、乙醇、THF、DCM、DCE、DMA或它们与乙酸的任意配比的混合物。可使用的还原剂例如为NaBH 4、NaBH 3CN或NaBH(OAc) 3。反应温度为0℃至80℃。在一些实施方案中,所述反应可在碱或酸的存在下进行,所述碱例如为TEA或DIPEA,并且所述酸例如为AcOH、HCl或Ti(O iPr) 4
在某些实施方案中,式I-A的化合物可由如下路线B所示的方法合成:
路线B
Figure PCTCN2020074696-appb-000051
其中:
Hal 2、R 1、R 2、R 5和R 23a如上文路线A所定义。
第一步:化合物I-A-5在酸性条件下脱除保护基生成化合物I-A-10。
可使用的酸例如为HCl的1,4-二氧六环溶液、HCl的EA溶液或TFA的DCM溶液。反应温度为0℃至80℃。
第二步:化合物I-A-10与I-A-9经还原胺化反应生成化合物I-A-11。
可使用的碱例如为DIPEA或TEA。可使用的还原剂例如为NaBH 3CN或NaBH(OAc) 3。可使用的溶剂例如为MeOH、EtOH或DCE。反应温度为0℃至80℃。
可使用的溶剂例如为甲醇、乙醇、THF、DCM、DCE、DMA或它们与乙酸的任意配比的混合物。可使用的还原剂例如为NaBH 4、NaBH 3CN或NaBH(OAc) 3。反应温度为0℃至80℃。在一些实施方案中,所述反应可在碱或酸的存在下进行,所述碱例如为TEA或DIPEA,并且所述酸例如为AcOH、HCl或Ti(O iPr) 4
第三步:化合物I-A-11与含硼试剂反应生成化合物I-A-12。
可使用的含硼试剂例如为B 2(pin) 2。可使用的催化剂例如为Pd(PPh 3) 4、Pd(dppf)Cl 2或Pd(dppf) 2Cl 2·DCM。可使用的碱例如为Cs 2CO 3、K 3PO 4、Na 2CO 3、KOAc、NaHCO 3或K 2CO 3。可使用的溶剂例如为1,4-二氧六环、DMF、DMSO或CH 3CN。反应温度为50℃至120℃。
第四步:化合物I-A-12与I-A-2经偶联反应(例如Suzuki反应)生成化合物I-A。
可使用的催化剂例如为Pd(PPh 3) 4、Pd(dppf)Cl 2或Pd(dppf) 2Cl 2·DCM。可使用的碱例如为Cs 2CO 3、K 3PO 4、Na 2CO 3、KOAc、NaHCO 3或K 2CO 3。可使用的溶剂例如为1,4-二氧六环、DMF、DMSO或CH 3CN,或者为任意上述溶剂与H 2O的混合物。反应温度为50℃至120℃。
在某些实施方案中,式I-B的化合物可由如下路线C所示的方法合成:
路线C
Figure PCTCN2020074696-appb-000052
其中:
Hal 1、Hal 2、R 1、R 2、R 5、R 23a如上文路线A所定义。
第一步:化合物I-B-1与R 2-NH 2在碱存在下经取代或偶联反应(例如Buchwald反应、Suzuki反应、Ullmann反应)生成化合物I-B-2。
反应条件如制备式I-A的化合物的路线A中第一步所述。
第二步:化合物I-B-2与I-A-12经偶联反应(例如Suzuki反应)生成化合物I-B。
反应条件如制备式I-A的化合物的路线A中第四步所述。
在某些实施方案中,式I-C的化合物可由如下路线D所示的方法合成:
路线D
Figure PCTCN2020074696-appb-000053
其中:
Hal 1、Hal 2、R 1、R 2、R 5和R 23a如上文路线A所定义;且
X 1选自CH和N。
第一步:化合物I-C-1与R 2-NH 2在碱存在下经取代或偶联反应(例如Buchwald反应、Suzuki反应或Ullmann反应)生成化合物I-C-2。
反应条件如制备式I-A的化合物的路线A中第一步所述。
第二步:化合物I-C-3与含硼试剂反应生成化合物I-C-4。
反应条件如制备式I-A的化合物的路线A中第三步所述。
第三步:化合物I-C-2与I-C-4经偶联反应(例如Suzuki反应)生成化合物I-C-5。
反应条件如制备式I-A的化合物的路线A中第四步所述。
第四步:化合物I-C-5在酸性条件下脱除保护基生成化合物I-C-6。
反应条件如制备式I-A的化合物的路线A中第五步所述。
第五步:化合物I-C-6与I-A-9经还原胺化反应生成化合物I-C。
反应条件如制备式I-A的化合物的路线A中第六步所述。
在某些实施方案中,式I-D化合物可由如下路线E所示的方法合成:
路线E
Figure PCTCN2020074696-appb-000054
其中:
R 1和R 2如上文路线A所定义;
R 5如上文式I-D所定义;
R 23a和R 23b各自独立地选自H、C 1-6烷基、C 1-6烷氧基和C 3-8环烷基;或者R 23a与R 23b连同其所连接的C原子一起形成3-8元环烷基或杂环基,所述烷基、烷氧基、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:CN、卤素、C 1-4烷基、C 1-4烷氧基、C 1-4羟烷基、C 1-4卤代烷基和C 1-4卤代烷氧基;
X 1选自CH和N;且
t为0或1。
化合物I-C-6与I-D-1经缩合反应生成化合物I-D。
可使用的缩合剂例如为HATU、CDI、HOBt、DMAP、DCC、DIC、EDC、HBTU、HCTU或PyBOP。可使用的碱例如为TEA、DIPEA、 tBuOK、 tBuONa、 tBuOLi、NaH、NaOH、Cs 2CO 3、K 3PO 4或Na 2CO 3。可使用的溶剂例如为THF、DCM、DCE、MeOH、EtOH、DMF、DMSO、丙酮、CH 3CN、1,4-二氧六环或甲苯。反应温度为0℃至120℃,例如室温。
或者,化合物I-D-1首先与酰化试剂反应生成酰基卤化物,然后与化合物I-C-6任选地在碱存在下反应生成式I-D的化合物。可使用的酰化试剂例如为氯化亚砜或草酰氯。该反应也可在少量DMF催化下进行。可使用的碱例如为TEA或DIPEA。可使用的溶剂例如为THF、DCM、DCE、CH 3CN、1,4-二氧六环或甲苯。反应温度为0℃至100℃。
在某些实施方案中,式I-E的化合物可由如下路线F所示的方法合成:
路线F
Figure PCTCN2020074696-appb-000055
其中:
R 1、R 2、R 5、R 23a、R 23b和t如上文路线E所定义;
X 1选自CH和N;且
Hal 2为F、Cl、Br或I;优选地,Hal 2为Cl、Br或I。
第一步:化合物I-C-2与I-A-6经偶联反应(例如Suzuki反应)生成化合物I-E-1。
反应条件如制备式I-A的化合物的路线A中第四步所述。
第二步:化合物I-E-1在酸性条件下脱除保护基生成化合物I-E-2。
反应条件如制备式I-A的化合物的路线A中第五步所述。
第三步:化合物I-E-2与I-D-1经缩合反应生成化合物I-E。
反应条件如制备式I-D的化合物的路线E所述。
在某些实施方案中,式I-F的化合物可由如下路线G所示的方法合成:
路线G
Figure PCTCN2020074696-appb-000056
其中:
R 1、R 2、R 23a、R 23b和R 5如上文路线E所定义;
X 1选自CH和N;
R 4不存在或者选自羟基、CN、C 1-6烷基、C 1-6卤代烷基和C 1-6杂烷基(例如C 1-6烷氧基);
R 23c为H、C 1-3烷基或C 1-3烷氧基,所述烷基和烷氧基各自任选地被一个或多个选自下列的取代基取代:OH、CN、卤素、C 1-4烷氧基和C 1-4羟烷基;
Hal 2为F、Cl、Br或I;优选地,Hal 2为Cl、Br或I;
u为0或1;且
n为0或1。
第一步:化合物I-C-2与I-F-1经偶联反应(例如Suzuki反应)生成化合物I-F-2。
反应条件如制备式I-A的化合物的路线A中第四步所述。
第二步:化合物I-F-3与胺经缩合反应生成化合物I-F-4。
反应条件如制备式I-D的化合物的路线E所述。
第三步:化合物I-F-4在酸性条件下脱除保护基生成化合物I-F-5。
反应条件如制备式I-A的化合物的路线A中第五步所述。
第四步:化合物I-F-2与I-F-5在碱存在下经亲核取代反应生成化合物I-F。
反应条件如制备式I-A的化合物的路线A中第二步所述。
在某些实施方案中,式I-G的化合物可由如下路线H所示的方法合成:
路线H
Figure PCTCN2020074696-appb-000057
其中:
R 1和R 2如上文路线A所定义;
X 1选自CH和N;
R 4选自H、C 1-6烷基、C 1-6卤代烷基和C 1-6杂烷基;
R 5如上文式I-G所定义;且
n为0或1。
第一步:化合物I-G-1与R 5-OH经Mitsunobu反应生成化合物I-G-2。
可使用的反应试剂例如为PPh 3、PMe 3、DIAD、DEAD或DBAD。可使用的溶剂为非质子性溶剂例如THF、乙醚、DCM、DMF或甲苯。反应温度为-20℃至100℃,例如室温。
第二步:化合物I-G-2在酸性条件下脱除保护基生成化合物I-G-3。
可使用的酸例如为HCl的1,4-二氧六环溶液、HCl的EA溶液或TFA的DCM溶液,或者反应在前述酸的溶液与例如选自THF、MeOH和EtOH的任一种溶剂的混合物中进行。反应温度为0℃至80℃。
第三步:化合物I-F-2与I-G-3在碱存在下经亲核取代反应生成化合物I-G。
反应条件如制备式I-A的化合物的路线A中第二步所述。
药物组合物、制剂和治疗方法
在一些实施方案中,本发明提供药物组合物,其包含预防或治疗有效量的本发明的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药。任选地,所述药物组合物还包含一种或多种药学上可接受的载体。
在一些实施方案中,本发明提供药物制剂,所述药物制剂优选为固体制剂、半固体制剂、液体制剂或气态制剂。在一些实施方案中,所述药物组合物还可包含一种或多种其它治疗剂。
在一些实施方案中,所述药物组合物或药物制剂优选通过口服、静脉内、动脉内、皮下、腹膜内、肌内或经皮途径给药。
在一些实施方案中,本发明提供本发明的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,或者如上所述的药物组合物、或者本发明的药物制剂在制备用于预防或治疗与RET活性相关的疾病或病况的药物中的用途。
在一些实施方案中,本发明提供本发明的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,或者如上所述的药物组合物、或者本发明的药物制剂在制备用于调节(例如降低或抑制)RET的活性的药物中的用途。
在一些实施方案中,本发明提供本发明的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,或者如上所述的药物组合物、或者本发明的药物制剂,其用于预防或治疗与RET活性相关的疾病或病况。
在一些实施方案中,本发明提供预防或治疗与RET活性相关的疾病或病况的方法,所述方法包括向需要其的个体给药有效量的本发明的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,或者如上所述的药物组合物、或者本发明的药物制剂。
在一些实施方案中,所述与RET活性相关的疾病或病况优选为癌症或肿瘤,或肠易激综合征。
在一些实施方案中,所述癌症或肿瘤进一步优选为肺癌(例如非小细胞肺癌)、乳腺癌、头颈癌、直肠癌、肝癌、淋巴瘤、甲状腺癌(例如甲状腺髓样癌或乳头状甲状腺癌)、结肠癌、多发性骨髓瘤、黑色素瘤、胶质瘤、脑瘤或肉瘤。
本发明中“药学上可接受的载体”是指与治疗剂一同给药的稀释剂、辅剂、赋形剂或媒介物,并且其在合理的医学判断的范围内适于接触人类和/或其它动物的组织而没有过度的毒性、刺激、过敏反应或与合理的益处/风险比相应的其它问题或并发症。
在本发明的药物组合物中可使用的药学上可接受的载体包括但不限于无菌液体。适合的药学上可接受的载体的实例如在Remington’s Pharmaceutical Sciences(1990)中所述。
本发明的药物组合物可以系统地作用和/或局部地作用。为此目的,它们可以适合的途径给药。
对于这些给药途径,可以适合的剂型给药本发明的药物组合物。
如本文中所使用的术语“有效量”指被给药后会在一定程度上缓解所治疗病症的一或多种症状的化合物的量。
可调整给药方案以提供最佳所需响应。例如,可给药单次推注,可随时间给药数个分剂量,或可如治疗情况的急需所表明而按比例减少或增加剂量。要注意,剂量值可随要减轻的病况的类型及严重性而变化,且可包括单次或多次剂量。要进一步理解,对于任何特定个体,具体的给药方案应根据个体需要及给药组合物或监督组合物的给药的人员的专业判断来随时间调整。
所给药的本发明的化合物的量会取决于所治疗的个体、病症或病况的严重性、给药的速率、化合物的处置及处方医师的判断。一般而言,有效剂量在每日每kg体重约0.0001至约50mg。在一些情 况下,不高于前述范围的下限的剂量水平可以是足够的,而在其它情况下,仍可在不引起任何有害副作用的情况下采用较大剂量,条件是首先将所述较大剂量分成数个较小剂量以在一整天中给药。
本发明的化合物在药物组合物中的含量或用量可以是约0.01mg至约1000mg。
除非另外说明,否则如本文中所使用,术语“治疗(treating)”意指逆转、减轻、抑制这样的术语所应用的病症或病况或者这样的病症或病况的一或多种症状的进展,或预防这样的病症或病况或者这样的病症或病况的一或多种症状。
如本文所使用的“个体”包括人或非人动物。示例性人个体包括患有疾病(例如本文所述的疾病)的人个体(称为患者)或正常个体。本发明中“非人动物”包括所有脊椎动物,例如非哺乳动物(例如鸟类、两栖动物、爬行动物)和哺乳动物,例如非人灵长类、家畜和/或驯化动物(例如绵羊、犬、猫、奶牛、猪等)。
在一些实施方案中,本发明的药物组合物还可以包含一种或多种另外的治疗剂或预防剂(例如其它用于治疗癌症或肿瘤疾病的药物)。在一些实施方案中,本发明的方法还可以包括给药一种或多种另外的治疗剂或预防剂(例如其它用于治疗癌症或肿瘤疾病的药物)。
实施例
以下结合实施例进一步描述本发明,但提供这些实施例并非意图限制本发明的范围。
本文中所用的缩写具有以下含义:
Figure PCTCN2020074696-appb-000058
Figure PCTCN2020074696-appb-000059
本发明的化合物通过制备TLC、硅胶柱层析、Prep-HPLC和/或快速柱层析(Flash柱层析)来分离纯化,其结构通过 1H NMR和/或MS来确证。反应监测采用TLC或LC-MS进行。
1H NMR波谱法采用Bruker超导核磁共振波谱仪(型号AVACE III HD 400MHz)。
LC/MS采用Aglient 1260Infinity/Aglient 6120Quadrupole。
TLC采用硅胶GF 254作为固定相。
柱层析一般使用200~300目硅胶(青岛海洋)作为固定相。
快速柱层析使用Biotage快速柱色谱仪。
Prep-HPLC采用Agilent 1260型、Waters 2489型和GeLai 3500型色谱仪。
微波反应使用BiotageInitiator微波反应器进行。
在以下实施例中,如无特殊说明,反应的温度为室温(15-30℃)。
本申请中所使用的试剂购自Acros Organics、Aldrich Chemical Company或特伯化学等公司。
实施例1:2-(6-(6-苄基-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物1)
Figure PCTCN2020074696-appb-000060
第一步:3-(5-溴吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-羧酸叔丁酯(化合物1c)的制备
在100mL单口瓶中依次加入化合物1a(1.50g)和1b(1.77g),再依次加入DMSO(20.0mL)和 K 2CO 3(5.83g),并于氮气保护下加热至90℃搅拌20h。反应结束后,将反应液冷却至室温,加入水(100mL)稀释,并用EA萃取。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩,并经硅胶快速柱层析分离纯化(PE:EA=5:1),得到化合物1c(2.03g)。MSm/z(ESI):354.1[M+H] +
第二步:3-(5-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-羧酸叔丁酯(化合物1d)的制备
在100mL单口瓶中依次加入化合物1c(2.03g)、B 2(pin) 2(4.01g)、KOAc(1.55g)、1,4-二氧六环(15.0mL)和Pd(dppf)Cl 2·DCM(644.67mg),并于氮气保护下加热至90℃反应。反应结束后,将反应液冷却至室温,加入水(30mL)稀释,并用EA萃取(40mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩至干,并经硅胶快速柱层析分离纯化(DCM:MeOH=15:1),得到化合物1d(2.11g)。MS m/z(ESI):402.3[M+H] +
第三步:3-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-羧酸叔丁酯(化合物1f)的制备
将化合物1e(950mg)溶解于1,4-二氧六环(50.0mL)中,依次加入化合物1d(2.11g)、Cs 2CO 3(3.15g)和水(5.0mL),再加入Pd(dppf)Cl 2·DCM(477.83mg),并于氮气保护下加热至90℃反应14h。反应结束后将反应液冷却至室温,加入水(100mL)稀释,并用EA萃取(60mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩至干,得到化合物1f(587.0mg)。MS m/z(ESI):463.3[M+H] +
第四步:2-(6-(3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物1g)的制备
将化合物1f(1.36g)溶解于DCM(20.0mL)中,再加入TFA(20.0mL),并于氮气保护下于室温反应。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物1g的三氟乙酸盐(587.0mg)。MS m/z(ESI):363.3[M+H] +
第五步:2-(6-(6-苄基-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物1)的制备
将化合物1g的三氟乙酸盐(31.58mg)和化合物1h(27.74mg)溶解于MeOH(0.5mL)中,依次加入TEA(8.46mg)和氰基硼氢化钠(26.27mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物1(11.0mg)。MS m/z(ESI):453.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.15(br,1H),9.67(s,1H),9.12(d,J=2.4Hz,1H),8.44(dd,J=8.8,2.4Hz,1H),7.43-7.21(m,5H),6.78(d,J=8.8Hz,2H),6.30(br,1H),3.98-3.57(m,8H),2.72-2.61(m,1H),2.33(s,3H),2.24(s,3H),1.72-1.64(m,1H).
实施例2:2-(6-(6-(4-甲氧基苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物2)
Figure PCTCN2020074696-appb-000061
将化合物1g的三氟乙酸盐(30.0mg)和化合物2a(33.81mg)溶解于MeOH(0.5mL)中,依次加入TEA(8.12mg)和氰基硼氢化钠(25.21mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物2(15.0mg)。MS m/z(ESI):483.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.14(br,1H),9.66(s,1H),9.12(d,J=2.4Hz,1H),8.43(dd,J=8.8,2.4Hz,1H),7.26(d,J=8.8Hz,2H),6.88-6.76(m,4H),6.30(br,1H),3.79-3.72(m,7H),3.58-3.53(m,4H),2.59-2.55(m,1H),2.33(s,3H),2.26(s,3H),1.60(d,J=8.4Hz,1H).
实施例3:2-(6-(6-((6-氯吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物3)
Figure PCTCN2020074696-appb-000062
将化合物1g的三氟乙酸盐(30.0mg)和化合物3a(35.15mg)溶解于MeOH(0.5mL)中,依次加入TEA(8.12mg)和氰基硼氢化钠(25.21mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物3(20.0mg)。MS m/z(ESI):488.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.12(br,1H),9.67(s,1H),9.11(d,J=2.0Hz,1H),8.46-8.42(m,2H),7.87-7.80(m,1H),7.50-7.47(m,1H),6.80-6.77(m,2H),6.31(br,1H),4.01-3.52(m,8H),2.66-2.57(m,1H),2.33(s,3H),2.26(s,3H),1.69-1.62(m,1H).
实施例4:2-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物4)
Figure PCTCN2020074696-appb-000063
第一步:3-(5-溴吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷(化合物4a)的制备
氮气保护下将化合物1c(460.0mg)溶解于DCM(5.0mL)中,加入TFA(5.0mL),并于室温反应2h至原料完全转化。反应结束后,将反应液减压浓缩至干,用饱和碳酸钠溶液洗涤,DCM萃取(30mL x 3),无水硫酸钠干燥,过滤后减压浓缩,得到化合物4a(250.0mg)。MS m/z(ESI):254.0[M+H] +
第二步:3-(5-溴吡啶-2-基)-6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷(化合物4c)的制备
将化合物4a(250.0mg)和4b(275.0mg)溶解于DCE(5.0mL)中,加入NaBH(OAc) 3(1.06g),并于室温反应10h。反应结束后,向反应液中加入水(100mL)稀释,并用EA萃取(50mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩至干,并经硅胶快速柱层析分离纯化(PE:EA=10:1-1:3),得到化合物4c(320.0mg)。MS m/z(ESI):375.1[M+H] +
第三步:6-((6-甲氧基吡啶-3-基)甲基)-3-(5-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷(化合物4d)的制备
将化合物4c(332.0mg)溶解于1,4-二氧六环(5.0mL)中,依次加入B 2(pin) 2(619.76mg)和KOAc(237.11mg),于氮气保护下加入Pd(dppf)Cl 2·DCM(99.65mg),并加热至90℃反应4h。反应结束后,将反应液冷却至室温,加入水(100mL)稀释,并用EA萃取(60mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩至干,并经硅胶快速柱层析分离纯化(DCM:MeOH=10:1),得到化合物4d(320.0mg)。MS m/z(ESI):423.3[M+H] +
第四步:2-(6-(6-((6-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物4)的制备
将化合物4d(129.8mg)溶解于1,4-二氧六环(5.0mL)中,依次加入化合物1e(55mg)、Cs 2CO 3(149mg)和水(5.0mL),于氮气保护下加入Pd(dppf)Cl 2·DCM(27.93mg),并加热至90℃反应5h。反应结束后,将反应液冷却至室温,加入水(100mL)稀释,并用EA萃取(60mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物4(18.0mg)。MS m/z(ESI):484.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.65(s,1H),9.12(d,J=2.4Hz,1H),8.43(dd,J=8.8,2.4Hz,1H),8.07(d,J=2.0Hz,1H),7.68(dd,J=8.4,2.4Hz,1H),6.78-6.75(m,3H),6.30(br,1H),3.82(s,3H),3.74(d,J=11.6Hz,2H),3.66(d,J=6.0Hz,2H),3.61-3.45(m,4H),2.53-2.51(m,1H),2.33(s, 3H),2.26(s,3H),1.57(d,J=8.0Hz,1H).
实施例5:2-(6-(6-((5-甲氧基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物18)
Figure PCTCN2020074696-appb-000064
将化合物1g的三氟乙酸盐(35.00mg)和化合物18a(27.75mg)加入到甲醇(1.0mL)中,然后依次加入三乙胺(6.83mg)和氰基硼氢化钠(17.00mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物18(6.0mg)。MS m/z(ESI):484.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.13(br,1H),9.65(s,1H),9.12(d,J=2.2Hz,1H),8.43(dd,J=8.92,2.32Hz,1H),8.21(s,1H),8.16-8.12(m,2H),7.35-7.31(m,1H),6.77(d,J=9.0Hz,1H),6.31(br,1H),3.81(s,3H),3.78-3.69(m,4H),3.59(br,4H),2.59-2.52(m,1H),2.33(s,3H),2.25(s,3H),1.59(d,J=8.36Hz,1H).
实施例6:2-(6-(6-((6-(4-氟-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物17)
Figure PCTCN2020074696-appb-000065
第一步:6-(4-氟-1H-吡唑-1-基)烟碱醛(化合物17a)的制备
将化合物8c(2.0g)、91a的盐酸盐(1.58g)和碳酸钾(4.45g)依次加入到DMF(15mL)中,并加热至80℃搅拌14h。将反应液降至室温,加入水(100mL)稀释,并用DCM萃取(50mL x 2)。合并有机相,用水及饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩,并经硅胶快速柱层析分离纯化(PE:EA=10:1),得到化合物17a(0.81g)。MS m/z(ESI):192.1[M+H] +
第二步:2-(6-(6-((6-(4-氟-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物17)的制备
将化合物1g的三氟乙酸盐(22.82mg)和化合物17a(27.47mg)加入到甲醇(1.0mL)中,然后依次加入三乙胺(4.45mg)和氰基硼氢化钠(13.86mg),并于室温反应14h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物17(7.0mg)。MS m/z(ESI):538.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.98(s,1H),9.66(s,1H),9.12(d,J=2.16Hz,1H),8.67(dd,J=4.54,0.64Hz,1H),8.43(dd,J=8.94,2.28Hz,1H),8.41(d,J=1.68,1H),7.98(dd,J=8.48Hz,2.12 1H),7.92(d,J=4.28,1H),7.87(d,J=8.4,1H),6.78(d,J=9.0Hz,2H),6.31(br,1H),3.78-3.71(m,4H),3.68-3.52(m,4H),2.59-2.52(m,1H),2.33(s,3H),2.25(s,3H),1.60(d,J=8.36Hz,1H).
实施例7:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-((6-甲基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物16)
Figure PCTCN2020074696-appb-000066
将化合物1g的三氟乙酸盐(35.0mg)和化合物16a(35.1mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(9.5mg)和氰基硼氢化钠(29.4mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物16(15.0mg)。MS m/z(ESI):468.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.66(s,1H),9.12(d,J=2.4Hz,1H),8.43(dd,J=8.8,2.4Hz,1H),8.38(s,1H),7.63(dd,J=7.6,1.6Hz,1H),7.18(d,J=8.0Hz,1H),6.77(d,J=8.8Hz,2H),6.29(br,1H),3.83-3.64(m,4H),3.63-3.45(m,4H),2.57-2.52(m,1H),2.43(s,3H),2.33(s,3H),2.26(s,3H),1.58(d,J=8.0Hz,1H).
实施例8:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-((2-甲基噻唑-5-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物15)
Figure PCTCN2020074696-appb-000067
将化合物1g的三氟乙酸盐(35.0mg)和化合物15a(36.8mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(9.5mg)和氰基硼氢化钠(29.4mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物15(16.0mg)。MS m/z(ESI):474.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.19(br,1H),9.65(s,1H),9.11(d,J=2.0Hz,1H),8.43(dd,J=8.8,2.4Hz,1H),8.17(s,1H),7.46(s,1H),6.76(d,J=8.8Hz,1H),6.29(br,1H),3.78-3.66(m,6H),3.64-3.51(m,2H),2.59(s,3H),2.49-2.44(m,1H),2.33(s,3H),2.25(s,3H),1.57(d,J=8.4Hz,1H).
实施例9:2-(6-(4-((6-甲氧基吡啶-3-基)甲基)哌嗪-1-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物49)
Figure PCTCN2020074696-appb-000068
第一步:4-(5-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)吡啶-2-基)哌嗪-1-羧酸叔丁酯(化合物49b)的制备
在反应瓶中依次加入化合物49a(2.00g)、B 2(pin) 2(4.09g)、KOAc(1.58g)、1,4-二氧六环(15.0mL)和Pd(dppf)Cl 2·DCM(657.43mg),并于氮气保护下加热至90℃反应3h。反应结束后,将反应液冷却至室温,加入水(30mL)稀释,并用EA萃取(40mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩至干,并经硅胶快速柱层析分离纯化(DCM:MeOH=15:1),得到化合物49b(2.77g)。MS m/z(ESI):390.3[M+H] +
第二步:4-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)哌嗪-1-羧酸叔丁酯(化合物49c)的制备
将化合物1e(1.30g)溶解于1,4-二氧六环(50.0mL)中,依次加入化合物49b(2.80g)、Cs 2CO 3(3.44g)和水(2.0mL),再加入Pd(dppf)Cl 2·DCM(647.3mg),并于氮气保护下加热至90℃反应4小时。反应结束后,将反应液冷却至室温,加入水(100mL)稀释,并用EA萃取(60mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩至干,得到化合物49c(587.0mg)。MS m/z(ESI):451.3[M+H] +
第三步:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(哌嗪-1-基)吡啶-3-基)嘧啶-4-胺(化合物49d)的制备
将化合物49c(2.7g)溶解于DCM(20.0mL)中,再加入TFA(20.0mL),并于氮气保护下于室温反应4h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物49d的三氟乙酸盐(761.0mg)。MS m/z(ESI):351.2[M+H] +
第四步:2-(6-(4-((6-甲氧基吡啶-3-基)甲基)哌嗪-1-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物49)的制备
将化合物49d的三氟乙酸盐(35.0mg)和化合物4b(38.9mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(9.3mg)和氰基硼氢化钠(28.8mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物49(25.0mg)。MS m/z(ESI):472.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.20(br,1H),9.65(s,1H),9.03(d,J=2.4Hz,1H),8.36(dd,J=8.8,2.0Hz,1H),8.09(d,J=2.0Hz,1H),7.68(dd,J=8.4,2.4Hz,1H),6.94-6.78(m,3H),6.29(br,1H),3.84(s,3H),3.64-3.56(m,4H),3.49(s,2H),2.49-2.44(m,4H),2.31(s,3H),2.24(s,3H).
实施例10:2-(6-(4-(4-甲氧基苄基)哌嗪-1-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物23)
Figure PCTCN2020074696-appb-000069
将化合物49d的三氟乙酸盐(35.0mg)和化合物2a(38.9mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(9.3mg)和氰基硼氢化钠(28.8mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物23(18.0mg)。MS m/z(ESI):471.3[M+H] +
1H NMR(CD 3OD,400MHz)δ9.07(d,J=2.4Hz,1H),8.45(dd,J=8.8,2.4Hz,1H),8.28(s,1H),7.42-7.35(m,2H),7.02-6.92(m,3H),6.76(s,1H),6.26(s,1H),3.99(s,2H),3.82(s,7H),3.08-3.00(m,4H),2.40(s,3H),2.31(s,3H).
实施例11:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-((4-甲基吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物21)
Figure PCTCN2020074696-appb-000070
将化合物1g的三氟乙酸盐(35.0mg)和化合物21a(27.52mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(7.4mg)和氰基硼氢化钠(23.08mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物21(10.0mg)。MS m/z(ESI):468.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.93(s,1H),9.61(s,1H),9.07(d,J=1.2Hz,1H),8.39(dd,J=8.8,2.2Hz,1H),8.35(s,1H),8.25(d,J=4.8Hz,1H),7.11(d,J=4.8Hz,1H),7.01-6.63(m,2H),6.27(br,1H),3.82-3.69(m,2H),3.63(d,J=6.4Hz,2H),3.59-3.42(m,4H),2.52-2.46(m,1H),2.28(s,3H),2.25(s,3H),2.21(s,3H),1.53(d,J=8.4Hz,1H).
实施例12:2-(6-(6-(2-氟-5-甲氧基苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物96)
Figure PCTCN2020074696-appb-000071
将化合物1g的三氟乙酸盐(30.0mg)和化合物96a(19.41mg)溶解于MeOH(0.5mL)中,依次加入TEA(6.37mg)和氰基硼氢化钠(19.78mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物96(10.0mg)。MS m/z(ESI):501.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(br,1H),9.65(s,1H),9.11(d,J=2.0Hz,1H),8.43(dd,J=9.2,2.4Hz,1H),7.06(t,J=9.2Hz,1H),7.03-7.00(m,1H),6.83-6.78(m,1H),7.03-6.64(m,2H),6.32(br,1H),3.77-3.71(m,7H),3.58-3.53(m,4H),2.58-2.53(m,1H),2.33(s,3H),2.25(s,3H),1.58(d,J=8.4Hz, 1H).
实施例13:(6-甲氧基吡啶-3-基)(4-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)哌嗪-1-基)甲酮(化合物110)
Figure PCTCN2020074696-appb-000072
将化合物110a(16.5mg)、HATU(53.2mg)和DIPEA(41.7mg)加入到DMF(3.0mL)中,并于室温反应5min。然后加入化合物49d的三氟乙酸盐(50.0mg),并于室温反应0.5小时。反应结束后,将反应液用EA稀释,饱和氯化钠溶液洗涤3次,无水硫酸钠干燥,过滤后浓缩,并经Prep-HPLC分离纯化,得到化合物110(19.0mg)。MS m/z(ESI):486.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.01(br,1H),9.69(s,1H),9.08(d,J=2.2Hz,1H),8.41(dd,J=9.0,2.3Hz,1H),8.34(d,J=2.1Hz,1H),7.84(dd,J=8.5,2.3Hz,1H),7.12-6.63(m,3H),6.30(s,1H),3.92(s,3H),3.72(s,8H),2.34(s,3H),2.26(s,3H).
实施例14:2-(6-甲氧基吡啶-3-基)-1-(4-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)哌嗪-1-基)乙-1-酮(化合物111)
Figure PCTCN2020074696-appb-000073
将化合物111a(18.0mg)、HATU(53.2mg)和DIPEA(41.7mg)加入到DMF(3.0mL)中,并于室温反应5min。然后加入化合物49d的三氟乙酸盐(50.0mg),并于室温反应0.5小时。反应结束后,将反应液用EA稀释,饱和氯化钠溶液洗涤3次,无水硫酸钠干燥,过滤后浓缩,并经Prep-HPLC分离纯化,得到化合物111(6.0mg)。MS m/z(ESI):500.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.02(br,1H),9.68(s,1H),9.06(d,J=2.3Hz,1H),8.39(dd,J=9.0,2.3Hz,1H),8.02(d,J=2.2Hz,1H),7.56(dd,J=8.5,2.4Hz,1H),6.94(d,J=9.0Hz,1H),6.90-6.69(m,2H),6.27(s,1H),3.83(s,3H),3.74(s,2H),3.70-3.59(m,8H),2.32(s,3H),2.25(s,3H).
实施例15:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-(4-甲基苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物80)
Figure PCTCN2020074696-appb-000074
将化合物1g的三氟乙酸盐(30mg)和化合物80a(22.70mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(6.37mg)和氰基硼氢化钠(19.78mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物80(10.0mg)。MS m/z(ESI):467.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.16(s,1H),9.66(s,1H),9.11(d,J=2.2Hz,1H),8.43(dd,J=8.9, 2.3Hz,1H),7.22(d,J=7.9Hz,2H),7.11(d,J=7.9Hz,2H),6.76(d,J=9.0Hz,2H),6.31(br,1H),3.78-3.65(m,4H),3.64-3.49(m,4H),2.60-2.49(m,1H),2.33(s,3H),2.27(s,3H),2.25(s,3H),1.59(d,J=8.4Hz,1H).
实施例16:5-((3-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚-6-基)甲基)-2-氰基吡啶(化合物117)
Figure PCTCN2020074696-appb-000075
将化合物1g的三氟乙酸盐(30mg)和化合物117a(24.96mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(6.37mg)和氰基硼氢化钠(19.78mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物117(2.0mg)。MS m/z(ESI):479.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.65(s,1H),9.11(d,J=2.2Hz,1H),8.74(d,J=1.2Hz,1H),8.43(dd,J=8.9,2.3Hz,1H),8.01(dt,J=17.2,5.0Hz,2H),6.77(d,J=9.0Hz,2H),6.32(br,1H),3.81-3.66(m,6H),3.65-3.52(m,2H),2.61-2.54(m,1H),2.33(s,3H),2.25(s,3H),1.60(d,J=8.4Hz,1H).
实施例17:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-((6-(三氟甲基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物118)
Figure PCTCN2020074696-appb-000076
将化合物1g的三氟乙酸盐(30mg)和化合物118a(43.48mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(8.38mg)和氰基硼氢化钠(26.01mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物118(6.0mg)。MS m/z(ESI):522.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.65(s,1H),9.12(d,J=2.1Hz,1H),8.74(s,1H),8.43(dd,J=8.9,2.3Hz,1H),8.06(d,J=7.1Hz,1H),7.84(d,J=8.1Hz,1H),6.77(d,J=9.0Hz,2H),6.30(br,1H),3.84-3.65(m,6H),3.60-3.48(m,2H),2.61-2.54(m,1H),2.33(s,3H),2.25(s,3H),1.61(d,J=8.4Hz,1H).
实施例18:6-甲基-2-(6-(6-((6-(4-甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物62)
Figure PCTCN2020074696-appb-000077
将化合物1g的三氟乙酸盐(30mg)和化合物62a(46.49mg,参考实施例6化合物17a合成方法制备,除将4-氟吡唑替换为4-甲基吡唑)加入到甲醇(0.5mL)中,然后依次加入三乙胺(8.38mg)和氰基硼氢化钠(26.01mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物62(6.0mg)。MS m/z(ESI):534.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.65(s,1H),9.12(d,J=2.2Hz,1H),8.44(dd,J=8.9,2.3Hz,1H),8.37(m,2H),7.93(dd,J=8.5,2.2Hz,1H),7.82(d,J=8.4Hz,1H),7.62(s,1H),6.78(d,J=9.0Hz,2H),6.30(br,1H),3.83-3.67(m,4H),3.66-3.50(m,4H),2.60-2.52(m,1H),2.33(s,3H),2.26(s,3H),2.11(s,3H),1.59(d,J=8.4Hz,1H).
实施例19:2-(6-(6-((6-(1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物60)
Figure PCTCN2020074696-appb-000078
将化合物1g的三氟乙酸盐(30mg)和化合物60a(43.00mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(8.38mg)和氰基硼氢化钠(26.01mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物60(11.0mg)。MS m/z(ESI):520.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.98(s,1H),9.65(s,1H),9.13(d,J=2.2Hz,1H),8.59(dd,J=2.6,0.5Hz,1H),8.46-8.42(dd,J=8.8,2.4Hz,1H),8.41-8.39(d,J=2.0Hz,1H),7.97(dd,J=8.4,2.2Hz,1H),7.88(d,J=8.4Hz,1H),7.81(d,J=1.0Hz,1H),6.78(d,J=9.0Hz,2H),6.56(dd,J=2.5,1.7Hz,1H),6.29(br,1H),3.82-3.68(m,4H),3.67-3.52(m,4H),2.59-2.52(m,1H),2.33(s,3H),2.26(s,3H),1.60(d,J=8.4Hz,1H).
实施例20:2-(6-(6-((5-氟吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物98)
Figure PCTCN2020074696-appb-000079
将化合物1g的三氟乙酸盐(35.0mg)和化合物98a(36.24mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(9.77mg)和氰基硼氢化钠(30.34mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物98(15.0mg)。MS m/z(ESI):472.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.98(br,1H),9.66(s,1H),9.12(d,J=2.2Hz,1H),8.45-8.41(m,3H),7.73-7.65(m,1H),6.77(d,J=9.0Hz,2H),6.29(br,1H),3.74(t,J=8.7Hz,4H),3.64(s,2H),3.58(d,J=6.5Hz,2H),2.55-2.51(m,1H),2.33(s,3H),2.26(s,3H),1.59(d,J=8.4Hz,1H).
实施例21:2-(6-(6-((5-氯吡啶-2-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物69)
Figure PCTCN2020074696-appb-000080
将化合物1g的三氟乙酸盐(35.0mg)和化合物69a(41.0mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(9.77mg)和氰基硼氢化钠(30.34mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物69(16.0mg)。MS m/z(ESI):488.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.01(br,1H),9.66(s,1H),9.11(d,J=2.2Hz,1H),8.50(d,J=2.3Hz,1H),8.43(dd,J=8.9,2.3Hz,1H),7.89(dd,J=8.4,2.5Hz,1H),7.52(d,J=8.4Hz,1H),6.76(d,J=9.0Hz,2H),6.30(s,1H),3.79(d,J=11.7Hz,2H),3.73(d,J=5.9Hz,2H),3.65(s,2H),3.57(d,J=10.0Hz,3H),2.55-2.51(m,1H),2.33(s,3H),2.26(s,3H),1.60(d,J=8.4Hz,1H).
实施例22:2-(6-(6-((5-甲氧基吡啶-2-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物67)
Figure PCTCN2020074696-appb-000081
将化合物1g的三氟乙酸盐(30mg)和化合物67a(25.90mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(6.37mg)和氰基硼氢化钠(19.78mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物67(10.0mg)。MS m/z(ESI):484.3[M+H] +
1H NMR(400MHz,DMSO-d 6))δ11.97(br,1H),9.65(s,1H),9.11(d,J=2.1Hz,1H),8.43(dd,J=8.9,2.3Hz,1H),8.16(dd,J=2.8,0.6Hz,1H),7.39(d,J=8.3Hz,1H),7.35(dd,J=8.6,2.9Hz,1H),6.76(d,J=9.0Hz,2H),6.30(s,1H),3.85-3.77(m,5H),3.69(d,J=5.9Hz,2H),3.60-3.47(m,4H),2.55-2.51(m,1H),2.33(s,3H),2.26(s,3H),1.58(d,J=8.4Hz,1H).
实施例23:2-(6-(6-(4-乙氧基苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物41)
Figure PCTCN2020074696-appb-000082
将化合物1g的三氟乙酸盐(30mg)和化合物41a(28.37mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(6.37mg)和氰基硼氢化钠(19.78mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物41(15.0mg)。MS m/z(ESI):497.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(br,1H),9.65(s,1H),9.11(d,J=2.4Hz,1H),8.43(dd,J=8.8,2.4Hz,1H),7.22(d,J=8.8Hz,2H),6.88-6.76(m,4H),6.32(br,1H),3.98(q,J=6.9Hz,2H),3.68(dd,J=30.7,8.6Hz,4H),3.51(d,J=30.4Hz,4H),2.59-2.55(m,1H),2.33(s,3H),2.26(s,3H),1.56(d,J=8.3Hz,1H),1.31(t,J=7.0Hz,3H).
实施例24:2-(6-(6-(1-(4-甲氧基苯基)乙基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物50)
Figure PCTCN2020074696-appb-000083
将化合物1g的三氟乙酸盐(30mg)和化合物50a(28.37mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(6.37mg)和氰基硼氢化钠(19.78mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物50(15.0mg)。MS m/z(ESI):497.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.63(s,1H),9.11(d,J=2.1Hz,1H),8.42(dd,J=8.9,2.2Hz,1H),7.25(d,J=8.6Hz,2H),7.02-6.57(m,4H),6.30(br,1H),3.97-3.77(m,2H),3.72(s,3H),3.67-3.50(m,2H),3.50-3.36(m,2H),2.49-2.39(m,2H),2.33(s,3H),2.26(s,3H),1.51(d,J=8.3Hz,1H),1.12(d,J=6.2Hz,3H).
实施例25:2-(6-(6-(4-氟苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物83)
Figure PCTCN2020074696-appb-000084
将化合物1g的三氟乙酸盐(35.0mg)和化合物83a(18.2mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(7.4mg)和氰基硼氢化钠(23.1mg),并于室温反应20h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物83(26.0mg)。MS m/z(ESI):471.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.64(s,1H),9.11(d,J=2.4Hz,1H),8.42(dd,J=9.2,2.4Hz,1H),7.93-7.35(m,2H),7.13-7.09(m,2H),6.83(br,1H),6.76(d,J=9.2Hz,1H),6.30(br,1H), 3.74-3.66(m,4H),3.62-3.53(m,4H),2.55-2.52(m,1H),2.32(s,3H),2.25(s,3H),1.57(d,J=8.4Hz,1H).
实施例26:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-(4-(三氟甲基)苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物84)
Figure PCTCN2020074696-appb-000085
将化合物1g的三氟乙酸盐(35.0mg)和化合物84a(25.6mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(7.4mg)和氰基硼氢化钠(23.1mg),并于室温反应20h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物84(17.0mg)。MS m/z(ESI):521.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.90(br,1H),9.65(s,1H),9.11(d,J=2.4Hz,1H),8.42(dd,J=8.8,2.4Hz,1H),7.66(d,J=8.0,2H),7.58(d,J=8.4,2H),6.94(br,1H),6.76(d,J=9.2Hz,1H),6.30(br,1H),3.74-3.57(m,8H),2.57-2.55(m,1H),2.33(s,3H),2.25(s,3H),1.59(d,J=8.4Hz,1H).
实施例27:4-((3-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-基)甲基)苯甲腈(化合物85)
Figure PCTCN2020074696-appb-000086
将化合物1g的三氟乙酸盐(35.0mg)和化合物85a(19.3mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(7.4mg)和氰基硼氢化钠(23.1mg),并于室温反应20h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物85(12.0mg)。MS m/z(ESI):478.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.08(br,1H),9.65(s,1H),9.11(d,J=2.4Hz,1H),8.42(dd,J=9.2,2.4Hz,1H),7.77(d,J=8.4,2H),7.56(d,J=8.0,2H),6.89(br,1H),6.76(d,J=8.8Hz,1H),6.30(br,1H),3.74-3.56(m,8H),2.59-2.58(m,1H),2.33(s,3H),2.25(s,3H),1.61(d,J=8.4Hz,1H).
实施例28:2-(6-(6-(4-(1H-吡唑-1-基)苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物86)
Figure PCTCN2020074696-appb-000087
第一步:4-(1H-吡唑-1-基)苯甲醛(化合物86b)的制备
将化合物83a(1.0g)和86a(0.8g)溶解于DMF(10.0mL)中,加入无水碳酸钾(2.2g),并加热至 120℃反应16h,直至原料完全转化。反应结束后,将反应液用饱和碳酸钠溶液洗涤,用EA萃取(30mL x 3),无水硫酸钠干燥,过滤后减压浓缩,得到化合物86b(1.1g)。MS m/z(ESI):173.1[M+H] +
第二步:2-(6-(6-(4-(1H-吡唑-1-基)苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物86)的制备
将化合物1g的三氟乙酸盐(35.0mg)和化合物86b(25.3mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(7.4mg)和氰基硼氢化钠(23.1mg),并于室温反应20h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物86(12.0mg)。MS m/z(ESI):519.2[M+H] +
1H NMR(400MHz,DMSO-d 6)11.97(s,1H),9.65(s,1H),9.12(d,J=2.4Hz,1H),8.46-8.42(m,2H),7.77-7.72(m,3H),7.45(d,J=8.8Hz,2H),6.89(br,1H),6.77(d,J=9.2Hz,1H),6.52(t,J=2.0Hz,1H),6.30(br,1H),3.77-3.59(m,8H),2.56-2.54(m,1H),2.33(s,3H),2.25(s,3H),1.59(d,J=8.4Hz,1H).
实施例29:2-(6-(6-(4-氯苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物82)
Figure PCTCN2020074696-appb-000088
将化合物1g的三氟乙酸盐(35.0mg)和化合物82a(20.6mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(7.4mg)和氰基硼氢化钠(23.1mg),并于室温反应20h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物82(26.0mg)。MS m/z(ESI):487.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.21(br,1H),9.65(s,1H),9.11(d,J=2.4Hz,1H),8.43(dd,J=8.8,2.4Hz,1H),7.41-7.36(m,4H),6.86(br,1H),6.76(d,J=9.2Hz,1H),6.30(br,1H),3.75-3.63(m,8H),2.60-2.56(m,1H),2.33(s,3H),2.25(s,3H),1.63(d,J=8.4Hz,1H).
实施例30:2-(6-(6-(4-(4-氟-1H-吡唑-1-基)苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物91)
Figure PCTCN2020074696-appb-000089
第一步:4-(4-氟-1H-吡唑-1-基)苯甲醛(化合物91b)的制备
将化合物83a(0.1g)和91a(0.1g)溶解于DMF(5.0mL)中,加入叔丁醇钾(0.3g),并加热至120℃反应16h,直至原料完全转化。反应结束后,将反应液用饱和碳酸钠溶液洗涤,用EA萃取(30mL x 3),无水硫酸钠干燥,过滤后减压浓缩,得到化合物91b(60mg)。MS m/z(ESI):190.1[M+H] +
第二步:2-(6-(6-(4-(4-氟-1H-吡唑-1-基)苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物91)的制备
将化合物1g的三氟乙酸盐(35.0mg)和化合物91b(27.9mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(7.4mg)和氰基硼氢化钠(23.1mg),并于室温反应20h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物91(23.0mg)。MS m/z(ESI):537.3[M+H] +
1H NMR(400MHz,DMSO-d 6)11.97(s,1H),9.63(s,1H),9.12(d,J=2.4Hz,1H),8.61(d,J=4.8 Hz,1H),8.43(dd,J=8.8,2.0Hz,1H),7.80(d,J=4.0Hz,1H),7.71(d,J=8.8Hz,2H),7.46(d,J=6.8Hz,2H),6.89(br,1H),6.77(d,J=8.8Hz,1H),6.30(br,1H),3.76-3.59(m,8H),2.56-2.54(m,1H),2.33(s,3H),2.25(s,3H),1.59(d,J=8.4Hz,1H).
实施例31:6-甲基-2-(6-(6-(4-(4-甲基-1H-吡唑-1-基)苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物88)
Figure PCTCN2020074696-appb-000090
第一步:1-(4-(1,3-二氧戊环-2-基)苯基)-4-甲基-1H-吡唑(化合物88c)的制备
将化合物88a(300mg)、88b(161.3mg)、反式-N,N’-二甲基-1,2-环己二胺(74.5mg)、CuI(50.0mg)和碳酸铯(1.71g)加入到DMF(5.0mL)中,并于氮气保护下加热至115℃反应7h。向反应液中加入H 2O(10mL)淬灭反应,并用EA萃取(10mL x 3)。合并有机相,用水(10mL x 3)洗涤,无水硫酸钠干燥,过滤后减压浓缩。通过硅胶柱层析(PE:EA=3:1)分离纯化,得到化合物88c(120mg)。MS m/z(ESI):231.2[M+H] +
第二步:4-(4-甲基-1H-吡唑-1-基)苯甲醛(化合物88d)的制备
将浓盐酸(1.5mL)滴加到化合物88c(120mg)的THF(10mL)和H 2O(8mL)溶液中,并加热至65℃反应1.5h。反应结束后,将反应液在冰浴下冷却,然后缓慢滴加饱和碳酸氢钠溶液,将反应液pH值调至约8。减压除去THF溶剂,然后用DCM萃取(10mL x 3)。合并有机相,用无水硫酸钠干燥,过滤后减压浓缩。通过硅胶柱层析(PE:EA=5:1)分离纯化,得到化合物88d(90mg)。MS m/z(ESI):187.1[M+H] +
第三步:6-甲基-2-(6-(6-(4-(4-氟-1H-吡唑-1-基)苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物88)的制备
将化合物1g的三氟乙酸盐(35.0mg)和化合物88d(54.0mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(9.8mg)和氰基硼氢化钠(30.3mg),并于室温反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物88(10.0mg)。MS m/z(ESI):533.3[M+H] +
1H NMR(400MHz,DMSO)δ11.98(s,1H),9.66(s,1H),9.14(s,1H),8.45(d,J=8.4Hz,1H),8.23(s,1H),7.71(d,J=8.0Hz,2H),7.54(s,1H),7.44(d,J=8.0Hz,2H),6.97-6.65(m,2H),6.32(br,1H),3.82-3.67(m,4H),3.65-3.50(m,4H),2.62-2.55(m,1H),2.35(s,3H),2.27(s,3H),2.11(s,3H),1.60(d,J=8.0Hz,1H).
实施例32:2-(6-(6-((6-(3,4-二甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物121)
Figure PCTCN2020074696-appb-000091
第一步:2-(3,4-二甲基-1H-吡唑-1-基)-5-(1,3-二氧戊环-2-基)吡啶(化合物121c)的制备
将化合物121a(300mg)、121b(188.0mg)、反式-N,N’-二甲基-1,2-环己二胺(74.2mg)、CuI(49.7mg)和碳酸铯(1.70g)加入到DMF(5.0mL)中,并于氮气保护下加热至120℃反应5h。向反应液中加入H 2O(10mL)淬灭反应,并用EA萃取(10mL x 3)。合并有机相,用水(10mL x 3)洗涤,无水硫酸钠干燥,过滤后减压浓缩。通过硅胶柱层析(PE:EA=4:1)分离纯化,得到化合物121c(305mg)。MSm/z(ESI):246.1[M+H] +
第二步:6-(3,4-二甲基-1H-吡唑-1-基)烟碱醛(化合物121d)的制备
将浓盐酸(2.0mL)滴加到化合物121c(305mg)的THF(10mL)溶液中,并加热至65℃反应1.5h。反应结束后,将反应在冰浴下冷却,然后缓慢加入碳酸钾,将pH值调至约8,然后用EA萃取(10mL x 3)。合并有机相,用无水硫酸钠干燥,过滤后减压浓缩。通过硅胶柱层析(PE:EA=4:1)分离纯化,得到化合物121d(200mg)。MS m/z(ESI):202.2[M+H] +
第三步:2-(6-(6-((6-(3,4-二甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物121)的制备
将化合物1g的三氟乙酸盐(40.4mg)和化合物121d(53.4mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(8.7mg)和氰基硼氢化钠(26.9mg),并于20℃反应16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物121(3.0mg)。MS m/z(ESI):548.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.96(s,1H),9.63(s,1H),9.12(d,J=2.4Hz,1H),8.43(dd,J=8.8,2.4Hz,1H),8.32(d,J=1.6Hz,1H),8.27(s,1H),7.88(dd,J=8.4,2.4Hz,1H),7.79-7.70(m,1H),7.03-6.60(m,2H),6.29(br,1H),3.83-3.70(m,4H),3.66-3.47(m,4H),2.58-2.52(m,1H),2.33(s,3H),2.25(s,3H),2.19(s,3H),2.02(s,3H),1.58(d,J=8.4Hz,1H).
实施例33:2-(6-(6-((5-氟吡啶-2-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物70)
Figure PCTCN2020074696-appb-000092
将化合物1g的三氟乙酸盐(30mg)和化合物70a(23.63mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(6.37mg)和氰基硼氢化钠(19.78mg),并于室温反应20h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物70(12.0mg)。MS m/z(ESI):472.3[M+H] +
1H NMR(400MHz,DMSO-d 6))δ11.97(br,1H),9.64(s,1H),9.11(d,J=2.2Hz,1H),8.43(dd,J=9.6,2.7Hz,2H),7.69(td,J=8.8,3.0Hz,1H),7.54(dd,J=8.7,4.7Hz,1H),6.77(d,J=9.0Hz,2H),6.31(s,1H),3.80(d,J=11.9Hz,2H),3.72(d,J=5.9Hz,2H),3.64(s,2H),3.56(d,J=10.2Hz,2H),2.57-2.51(m,1H),2.33(s,3H),2.26(s,3H),1.59(d,J=8.4Hz,1H).
实施例34:2-(6-(6-((6-(3-氟-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物63)
Figure PCTCN2020074696-appb-000093
将化合物1g的三氟乙酸盐(30mg)和化合物63a(47.47mg,参考实施例6化合物17a合成方法制备,除将4-氟吡唑替换为3-氟吡唑)加入到甲醇(0.5mL)中,然后依次加入三乙胺(8.38mg)和氰基硼氢化钠(26.01mg),并于20℃搅拌16h。反应结束后,将反应液减压浓缩至干,经Prep-HPLC分离纯化,得到化合物63(10.0mg)。MS m/z(ESI):538.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.65(s,1H),9.12(d,J=2.1Hz,1H),8.55(m,1H),8.47-8.38(m,2H),7.98(d,J=8.6Hz,1H),7.70(d,J=8.4Hz,1H),6.80(d,J=8.0Hz,2H),6.56(dd,J=8,4Hz,1H),6.29(br,1H),3.84-3.68(m,4H),3.67-3.47(m,4H),2.60-2.54(m,1H),2.33(s,3H),2.25(s,3H),1.60(d,J=8.2Hz,1H).
实施例35:2-(6-(6-((6-(4-氟-1H-咪唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物64)
Figure PCTCN2020074696-appb-000094
第一步:5-(1,3-二氧戊环-2-基)-2-(4-氟-1H-咪唑-1-基)吡啶(化合物64b)的制备
将化合物64a(230mg)、121a(86mg)、N,N’-二甲基乙二胺(38mg)、CuI(83mg)和碳酸铯(425mg)加入到DMF(1mL)中,并于氮气保护下加热至115℃反应3h。向反应液中加入水淬灭反应,并用EA萃取。有机相用水洗涤,无水硫酸钠干燥,过滤后减压浓缩,得到化合物64b(120mg)粗产物,其不经纯化直接用于下一步反应。MS m/z(ESI):236.1[M+H] +
第二步:6-(4-氟-1H-咪唑-1-基)烟碱醛(化合物64c)的制备
将浓盐酸(12N,3.0mL)滴加到化合物64b(120mg)的THF(10mL)和水(10mL)溶液中,并于室温反应18h。反应结束后,用饱和碳酸氢钠溶液将反应液pH值调至约8,然后用EA萃取,用无水硫酸钠干燥。过滤后减压浓缩,得到化合物64c(60mg)粗产物,其不经纯化直接用于下一步反应。MS m/z(ESI):192.1[M+H] +
第三步:2-(6-(6-((6-(4-氟-1H-咪唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物64)的制备
将化合物1g的三氟乙酸盐(30mg)和化合物64c(47.47mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(8.38mg)和氰基硼氢化钠(26.01mg),并于20℃搅拌16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物64(10.0mg)。MS m/z(ESI):538.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.64(s,1H),9.12(d,J=2.2Hz,1H),8.44(dd,J=9.1,2.4 Hz,2H),8.28(m,1H),7.99(d,J=8.4Hz,1H),7.76-7.74(d,J=8.0Hz,1H),7.69-7.67(dd,J=8.4,1.6Hz,1H),6.86(br,1H),6.77(d,J=9.2Hz,1H),6.30(br,1H),3.78-3.71(m,4H),3.67-3.51(m,4H),2.59-2.53(m,1H),2.33(s,3H),2.26(s,3H),1.60(d,J=8.4Hz,1H).
实施例36:2-(6-(6-(1-(6-(4-氟-1H-吡唑-1-基)吡啶-3-基)乙基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物52)
Figure PCTCN2020074696-appb-000095
将化合物1g(320mg)、化合物52a(181.17mg,参考实施例6化合物17a合成方法制备,除将6-溴烟碱醛替换为1-(6-溴吡啶-3-基)乙酮)和钛酸四异丙酯(752.81mg)加入到干燥的THF(25mL)中,氮气置换三次后,于75℃搅拌24h。然后向体系分批加入三乙酰氧基硼氢化钠(935.64mg),并补加干燥的THF(15mL),继续于75℃搅拌16h。反应结束后,将反应液减压浓缩至干,经快速柱层析(MeOH:DCM=1:9)分离纯化,得到化合物52粗品,再经Prep-HPLC分离纯化,得到化合物52(90.0mg)。MS m/z(ESI):552.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.96(s,1H),9.63(s,1H),9.12(d,J=2.1Hz,1H),8.66(d,J=4.4Hz,1H),8.46-8.39(m,2H),8.01(dd,J=8.5,2.0Hz,1H),7.90(dd,J=14.7,6.2Hz,2H),6.82(br,1H),6.75(d,J=12.0Hz,1H),6.27(br,1H),3.96-3.82(m,2H),3.77(q,J=6.1Hz,1H),3.63(m,1H),3.49-3.37(m,3H),2.55-2.51(m,1H),2.33(s,3H),2.25(s,3H),1.55(d,J=8.4Hz,1H),1.23(d,J=6.2Hz,3H).
实施例37:2-(6-(6-((6-(3-环丙基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物120)
Figure PCTCN2020074696-appb-000096
将化合物1g的三氟乙酸盐(30mg)和化合物120a(22.95mg,参考实施例35化合物64c合成方法制备,除将起始原料4-氟咪唑替换为3-环丙基吡唑))加入到甲醇(0.5mL)中,然后依次加入三乙胺(8.38mg)和氰基硼氢化钠(26.01mg),并于20℃搅拌16h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物120(13.0mg)。MS m/z(ESI):560.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.64(s,1H),9.12(d,J=2.2Hz,1H),8.44(dd,J=8.9,2.4Hz,2H),8.35(d,J=1.3Hz,1H),7.92(dd,J=8.5,1.8Hz,1H),7.78(d,J=8.4Hz,1H),6.8(br,1H),6.78(d,J=9.0Hz,1H),6.28(br,1H),6.26(d,J=2.5Hz,1H),3.81-3.66(m,4H),3.67-3.52(m,4H),2.58-2.53(m,1H),2.33(s,3H),2.26(s,3H),2.03-1.95(m,1H),1.59(d,J=8.4Hz,1H),0.98-0.91(m,2H),0.79-0.73(m,2H).
实施例38:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(4-((6-甲基吡啶-3-基)氧基)哌啶-1-基)吡啶-3-基)嘧啶-4-胺(化合物119)
Figure PCTCN2020074696-appb-000097
第一步:4-((6-甲基吡啶-3-基)氧基)哌啶-1-羧酸叔丁基酯(化合物119b)的制备
将化合物119a(100mg)、N-Boc-4-羟基哌啶(276.65mg)和三苯基膦(480.18mg)加入到干燥的THF(5mL)中,并降温至0℃。向体系内滴加DIAD(370.21mg),并于室温下搅拌16h。反应结束后,将体系减压浓缩,并经快速柱层析纯化(PE:EA=1:1),得到化合物119b(65mg)。MS m/z(ESI):293.2[M+H] +
第二步:2-甲基-5-(哌啶-4-基氧基)吡啶(化合物119c)的制备
将化合物119b(65mg)加入到氯化氢的1,4-二氧六环溶液(4N,2mL)和THF(1mL)的混合溶液中,并于室温搅拌2h。减压浓缩至干,得到化合物119c的盐酸盐(54mg)。MS m/z(ESI):193.2[M+H] +
第三步:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(4-((6-甲基吡啶-3-基)氧基)哌啶-1-基)吡啶-3-基)嘧啶-4-胺(化合物119)的制备
将化合物119c的盐酸盐(50mg)、化合物119d(50mg)和碳酸钾(73mg)加入到DMF(2mL)中,并加热至100℃搅拌16h。反应结束后,将反应液降至室温。加入水(30mL)稀释,并用EA萃取(30mL x 2)。合并有机相,用无水硫酸钠干燥,过滤后浓缩。通过Prep-HPLC分离纯化,得到化合物119(13mg)。MS m/z(ESI):457.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.96(s,1H),9.63(s,1H),9.04(d,J=2.4Hz,1H),8.37(dd,J=9.0,2.4Hz,1H),8.19(d,J=2.9Hz,1H),7.38(dd,J=8.5,3.0Hz,1H),7.18(d,J=8.5Hz,1H),6.96(d,J=9.1Hz,1H),6.82(s,1H),6.28(s,1H),4.72-4.65(m,1H),4.13-4.01(m,2H),3.49-3.40(m,2H),2.40(s,3H),2.32(s,3H),2.25(s,3H),2.06-1.98(m,2H),1.69-1.58(m,2H).
实施例39:2-(6-(6-(4-(3-氟-1H-吡唑-1-基)苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物89)
Figure PCTCN2020074696-appb-000098
第一步:1-(4-(1,3-二氧戊环-2-基)苯基)-3-氟-1H-吡唑(化合物89b)的制备
将化合物89a(80mg)、88a(212.92mg)、N,N'-二甲基乙二胺(81.94mg)、碳酸铯(908.55mg)和CuI(177.02mg)依次加入到DMF(6mL)中,并加热至110℃搅拌12h。将反应液降至室温,加入水(50mL)稀释,并用DCM萃取(50mL x 2)。合并有机相,用水及饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩,并经快速柱层析分离纯化(PE:EA=63:37),得到化合物89b(35mg)。MS m/z(ESI):235.1[M+H] +
第二步:4-(3-氟-1H-吡唑-1-基)苯甲醛(化合物89c)的制备
将化合物89b(35mg)加入到氯化氢的1,4-二氧六环溶液(4N,2mL)和DCM(1mL)的混合溶液中,并于室温搅拌2h。将反应液减压浓缩,得到化合物89c(28mg)。MS m/z(ESI):191.1[M+H] +
第三步:2-(6-(6-(4-(3-氟-1H-吡唑-1-基)苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5- 甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物89)的制备
将化合物89c(14.37mg)、化合物1g的三氟乙酸盐(30mg)、三乙胺(6.37mg)和氰基硼氢化钠(19.78mg)依次加入到甲醇(0.5mL)中,并于室温搅拌36h。加入饱和氯化铵水溶液(0.1mL)淬灭反应。将反应液浓缩,用制备TLC预纯化(DCM:MeOH=10:1),得到粗产物5mg(R f=0.15-0.25),再经Prep-HPLC分离纯化,得到化合物89(4mg)。MS m/z(ESI):537.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.64(s,1H),9.12(d,J=2.3Hz,1H),8.49-8.39(m,2H),7.68(d,J=8.6Hz,2H),7.46(d,J=8.5Hz,2H),6.78(d,J=9.0Hz,2H),6.31(dd,J=5.8,2.6Hz,2H),3.80-3.67(m,4H),3.64-3.48(m,4H),2.59-2.54(m,1H),2.33(s,3H),2.25(s,3H),1.59(d,J=8.4Hz,1H).
实施例40:6'-(6-((6-(4-氟-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)-4-甲基-N-(5-甲基-1H-吡唑-3-基)-[2,3'-联吡啶]-6-胺(化合物6)
Figure PCTCN2020074696-appb-000099
第一步:3-氨基-5-甲基-1H-吡唑-1-羧酸叔丁酯(化合物6b)的制备
将化合物6a(2.4g)溶解于干燥的THF(50mL)中,分批加入NaH(988.39mg,纯度60%),然后搅拌10min。滴入二碳酸二叔丁酯(5.39g),并于氮气保护下25℃搅拌2h。向反应液中加入水淬灭反应,并用EA萃取。有机相用无水硫酸钠干燥,过滤后浓缩。通过硅胶柱层析(PE:EA=5:1)分离纯化,得到化合物6b(3.95g)。
第二步:3-((6-溴-4-甲基吡啶-2-基)氨基)-5-甲基-1H-吡唑-1-羧酸叔丁酯(化合物6d)的制备
将化合物6c(1.0g)、6b(864.65mg)、醋酸钯(89.47mg)、Xantphos(461.20mg)、碳酸铯(2.60g)依次加入到1,4-二氧六环(10mL)中,并于氮气保护下95℃搅拌2h。LC-MS监测原料反应完全,有目标产物生成。将反应液降至室温,过滤后浓缩。通过硅胶柱层析(PE:EA=3:1)分离纯化,得到化合物6d(415mg)。MS m/z(ESI):367[M+H] +
第三步:6-溴-4-甲基-N-(5-甲基-1H-吡唑-3-基)吡啶-2-胺(化合物6e)的制备
将化合物6d(300mg)加入到氯化氢的1,4-二氧六环溶液(4N,2mL)中,并于25℃搅拌1h。直接浓缩至干,加入甲醇(5mL)溶解粗产物。然后加入三乙胺(1mL),并于室温搅拌15min。将反应液浓缩,并经硅胶快速柱层析(DCM:MeOH=97:3)纯化,得到化合物6e(145mg)。MS m/z(ESI):267[M+H] +
第四步:6'-(6-((6-(4-氟-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)-4-甲基-N-(5-甲基-1H-吡唑-3-基)-[2,3'-联吡啶]-6-胺(化合物6)的制备
向反应瓶中依次加入化合物6e(30mg)、10f(85mg)、Na 2CO 3(27.85mg)、Pd(PPh 3) 4(12.98mg)、H 2O(1mL)、1,4-二氧六环(5mL),氮气置换三次后,于95℃搅拌5h。反应结束后,加水稀释,并用EA萃取。收集有机层,用水及饱和食盐水洗涤,无水硫酸钠干燥,抽滤后浓缩,用制备TLC预纯化(DCM:MeOH=9:1),再经Prep-HPLC分离纯化,得到化合物6(6mg)。MS m/z(ESI):537.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.63(s,1H),9.02(s,1H),8.86(d,J=2.3Hz,1H),8.67(dd,J=4.5,0.6Hz,1H),8.41(d,J=1.7Hz,1H),8.21(dd,J=8.9,2.4Hz,1H),7.98(dd,J=8.5,2.2Hz,1H),7.92(d,J=4.3Hz,1H),7.86(d,J=8.4Hz,1H),7.03(s,1H),6.88(s,1H),6.77(d,J=9.0Hz,1H),6.22(s,1H),3.81-3.69(m,4H),3.66-3.53(m,4H),2.58-2.53(m,1H),2.26(s,3H),2.22(s,3H),1.59(d,J=8.4Hz,1H).
实施例41:2-(6-(6-((6'-甲氧基-[2,3'-联吡啶]-5-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)- 6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物7)
Figure PCTCN2020074696-appb-000100
第一步:5-(1,3-二氧戊环-2-基)-6'-甲氧基-2,3'-联吡啶(化合物7b)的制备
向反应瓶中依次加入化合物7a(499mg)、121a(500mg)、Na 2CO 3(691mg)、Pd(PPh 3) 4(126mg)、H 2O(3mL)、1,4-二氧六环(17mL),氮气置换三次后,于95℃搅拌5h。反应结束后,浓缩至干。通过硅胶柱层析(PE:EA=1:1)分离纯化,得到化合物7b(350mg)。
第二步:6'-甲氧基-[2,3'-联吡啶]-5-甲醛(化合物7c)的制备
将浓盐酸(12N,3.0mL)滴加到化合物7b(200mg)的THF(11mL)和水(9mL)溶液中,并于室温反应18h。反应结束后,用碳酸钾溶液将反应液pH值调至约10,然后用EA萃取。有机相用无水硫酸钠干燥,过滤后减压浓缩。通过硅胶柱层析(PE:EA=2:1)分离纯化,得到化合物7c(60mg)。MSm/z(ESI):215.1[M+H] +
第三步:2-(6-(6-((6'-甲氧基-[2,3'-联吡啶]-5-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物7)的制备
将化合物1g的三氟乙酸盐(50mg)和化合物7c(67.44mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(10.62mg)和氰基硼氢化钠(32.97mg),并于室温反应20h。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物7(10.0mg)。MS m/z(ESI):561.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.98(br,1H),9.65(s,1H),9.13(d,J=2.2Hz,1H),8.86(d,J=2.2Hz,1H),8.60(d,J=1.5Hz,1H),8.44(dd,J=8.9,2.3Hz,1H),8.36(dd,J=8.7,2.5Hz,1H),7.90(d,J=8.1Hz,1H),7.83(dd,J=8.2,2.1Hz,1H),6.85(dd,J=56.9,8.8Hz,3H),6.33(s,1H),3.91(s,3H),3.75(dd,J=20.2,8.9Hz,4H),3.65-3.49(m,4H),2.59-2.53(m,1H),2.33(s,3H),2.26(s,3H),1.60(d,J=8.4Hz,1H).
实施例42:2-(6-(6-((5'-氟-2'-甲基-[2,3'-联吡啶]-5-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物8)
Figure PCTCN2020074696-appb-000101
第一步:5-氟-2-甲基吡啶-3-硼酸频那醇酯(化合物8b)的制备
向反应瓶中依次加入联硼酸频那醇酯(1.20g)、化合物8a(300mg)、Pd(dppf)Cl 2·DCM(128.93mg)、醋酸钾(464.85mg)和干燥的1,4-二氧六环(10mL),并于氮气保护下加热至100℃搅拌4h。反应结束后,过滤。加水稀释滤液,并用EA萃取。将有机相用水洗涤两次,用无水硫酸钠干燥。过滤,浓缩滤液,并经硅胶快速柱层析(PE:EA=1:1)分离纯化,得到化合物8b(300mg)。MS m/z(ESI):238.2[M+H] +
第二步:2-(6-(6-((6-溴吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基- 1H-吡唑-3-基)嘧啶-4-胺(化合物8d)的制备
将化合物1g的三氟乙酸盐(200mg)和化合物8c(195.20mg)加入到甲醇(10mL)中,于室温搅拌下加入三乙胺(42.48mg)。搅拌30min后,加入氰基硼氢化钠(131.89mg),于20℃搅拌16h。LC-MS监测显示产物峰明显。经硅胶快速柱层析(DCM:MeOH=10:1)分离纯化,得到化合物8d(220mg)。MSm/z(ESI):532.1[M+H] +
第三步:2-(6-(6-((5'-氟-2'-甲基-[2,3'-联吡啶]-5-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物8)的制备
向反应瓶中依次加入化合物8b(222.63mg)、8d(100mg)、Na 2CO 3(69.87mg)、Pd(PPh 3) 4(32.55mg)、水(1mL)和1,4-二氧六环(5mL),并于氮气保护下95℃搅拌5h。反应结束后,旋转蒸发除去溶剂,经制备TLC预纯化(DCM:MeOH=9:1),再经Prep-HPLC分离纯化,得到化合物8(25mg)。MS m/z(ESI):563.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.66(s,1H),9.13(d,J=2.2Hz,1H),8.66(d,J=1.5Hz,1H),8.51(d,J=2.9Hz,1H),8.44(dd,J=8.9,2.3Hz,1H),7.90(dd,J=8.1,2.1Hz,1H),7.76(dd,J=9.5,2.8Hz,1H),7.62(d,J=8.0Hz,1H),6.81(br,1H),6.79(d,J=9.0Hz,1H),6.32(br,1H),3.85-3.71(m,4H),3.70-3.51(m,4H),2.61-2.53(m,1H),2.51(s,3H),2.33(s,3H),2.26(s,3H),1.61(d,J=8.4Hz,1H).
实施例43:6-甲基-2-(6-(6-((6-(5-甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物9)
Figure PCTCN2020074696-appb-000102
将化合物1g的三氟乙酸盐(30mg)和化合物9a(17.68mg,参考实施例35化合物64c合成方法制备,除将起始原料4-氟咪唑替换为5-甲基吡唑)加入到甲醇(0.5mL)中,然后加入三乙胺(8.38mg)。搅拌30min后,加入氰基硼氢化钠(19.78mg),于20℃搅拌16h。LC-MS检测显示原料反应完全,产物峰明显。经Prep-HPLC分离纯化,得到化合物9(7mg)。MS m/z(ESI):534.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.99(s,1H),9.67(s,1H),9.14(d,J=2.1Hz,1H),8.49(d,J=2.4Hz,1H),8.45(dd,J=9.2,2.4Hz,1H),8.38(d,J=1.6Hz,1H),7.94(dd,J=8.5,2.1Hz,1H),7.81(d,J=8.4Hz,1H),6.83(br,1H),6.79(d,J=9.2Hz,1H),6.38(d,J=2.4Hz,1H),6.33(br,1H),3.85-3.71(m,4H),3.69-3.51(m,4H),2.61-2.55(m,1H),2.35(s,3H),2.30(s,3H),2.27(s,3H),1.61(d,J=8.4Hz,1H).
实施例44:N-(5-环丙基-1H-吡唑-3-基)-2-(6-(6-((6-(4-氟-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基嘧啶-4-胺(化合物10)
Figure PCTCN2020074696-appb-000103
第一步:2-氯-N-(5-环丙基-1H-吡唑-3-基)-6-甲基嘧啶-4-胺(化合物10c)的制备
将化合物10b(1.0g)和10a(755.53mg)溶解于乙醇(20.00mL)中,然后加入N,N-二异丙基乙胺(1590.00mg),并于氮气保护下加热至70℃搅拌48h。将反应液减压浓缩,并用EA(300.00mL)稀释。有机相用水洗涤三次,再用饱和氯化钠水溶液洗涤,无水硫酸钠干燥。过滤后浓缩,经硅胶柱层析(DCM:MeOH=9:1)分离纯化,得到化合物10c(820.00mg)。MS m/z(ESI):250.1[M+H] +
第二步:3-(5-溴吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷(化合物10d)的制备
将化合物1c(1180.00mg)溶解于甲醇(5.00mL)和DCM(10.00mL)混合溶剂中,然后在冰浴下缓慢滴加氯化氢的1,4-二氧六环溶液(4N,6.00mL),并于25℃搅拌16h。将反应液减压浓缩,得到化合物10d的盐酸盐(1040.00mg)。MS m/z(ESI):254.0[M+H] +
第三步:3-(5-溴吡啶-2-基)-6-((6-(4-氟-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷(化合物10e)的制备
将化合物10d的盐酸盐(300.00mg)和化合物17a(297.04mg)溶解于1,2-二氯乙烷(10.00mL)中,缓慢滴加三乙胺(104.82mg)。搅拌10min后滴加醋酸(31.10mg),并继续搅拌30min。然后加入三乙酰氧基硼氢化钠(878.21mg),并于25℃搅拌20h。向反应液中加入饱和氯化铵水溶液淬灭反应,加水稀释,并用DCM萃取。有机层用无水硫酸钠干燥,过滤后浓缩。经硅胶柱层析(PE:EA=1:1)分离纯化,得到化合物10e(363.00mg)。MS m/z(ESI):429.1[M+H] +
第四步:6-((6-(4-氟-1H-吡唑-1-基)吡啶-3-基)甲基)-3-(5-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷(化合物10f)的制备
将化合物10e(187.00mg)和联硼酸频那醇酯(221.23mg)溶解于1,4-二氧六环(15.00mL)中,然后加入醋酸钾(106.88mg)、Pd(dppf)Cl 2·DCM(35.57mg),并于氮气保护下加热至95℃搅拌5h。向反应液中加入EA(100.00mL)稀释,并用水洗涤三次。有机相用无水硫酸钠干燥,过滤后浓缩,经硅胶柱层析(PE:EA=3:1)分离纯化,得到化合物10f(100.00mg)。MS m/z(ESI):477.3[M+H] +
第五步:N-(5-环丙基-1H-吡唑-3-基)-2-(6-(6-((6-(4-氟-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基嘧啶-4-胺(化合物10)的制备
将化合物10c(26.21mg)和10f(50.00mg)溶解于1,4-二氧六环(4.00mL)中,依次加入Pd(PPh 3) 4(18.19mg)和碳酸钠水溶液(33.38mg溶解于1.00mL水),并于氮气保护下加热至95℃搅拌12h。反应结束后,向反应液中加入EA(20.00mL)稀释,并用水洗涤三次。有机相用无水硫酸钠干燥,过滤 后浓缩,得到粗产物。粗产物经Prep-HPLC分离纯化,得到化合物10(9.00mg)。MS m/z(ESI):564.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.02(s,1H),9.64(s,1H),9.11(d,J=2.4Hz,1H),8.67(dd,J=4.8,0.8Hz,1H),8.42(dd,J=9.2,2.0Hz,2H),7.99(dd,J=8.4,2.0Hz,1H),7.92(dd,J=4.4,0.8Hz,1H),7.87(d,J=8.4Hz,1H),6.78(d,J=9.2Hz,2H),6.18(br,1H),3.79-3.71(m,4H),3.64-3.53(m,4H),2.56-2.51(m,1H),2.33(s,3H),1.95-1.89(m,1H),1.60(d,J=8.4Hz,1H),0.97-0.92(m,2H),0.75-0.69(m,2H).
实施例45:2-(6-(6-((6-(3,5-二甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物11)
Figure PCTCN2020074696-appb-000104
第一步:2-(3,5-二甲基-1H-吡唑-1-基)-5-(1,3-二氧戊环-2-基)吡啶(化合物11b)的制备
将化合物11a(1.0g)、121a(1.74g)、N,N’-二甲基乙二胺(664.83mg)、CuI(1.44g)和碳酸铯(7.37g)加入到DMF(30mL)中,并于氮气保护下加热至98℃反应16h。向反应液中加入水淬灭反应,并用EA萃取。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩。通过硅胶柱层析(PE:EA=3:1)分离纯化,得到化合物11b(0.69g)。MS m/z(ESI):246.2[M+H] +
第二步:6-(3,5-二甲基-1H-吡唑-1-基)烟碱醛(化合物11c)的制备
将盐酸(2N,3.0mL)滴加到化合物11b(250mg)的THF(6mL)溶液中,并于室温反应3h。反应结束后,将反应液在冰浴下冷却。缓慢加入碳酸钾,将pH值调至约8,然后用EA萃取(10mL x 3)。合并有机相,用无水硫酸钠干燥,过滤后减压浓缩。通过硅胶柱层析(PE:EA=3:1)分离纯化,得到化合物11c(150mg)。
第三步:2-(6-(6-((6-(3,5-二甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物11)的制备
将化合物1g(30mg)和化合物11c(25mg)加入到甲醇(1mL)中,然后加入乙酸(5mg),并于室温搅拌0.5h。加入氰基硼氢化钠(15.6mg),并于室温反应20h。反应结束后,将反应液减压浓缩至干,经制备TLC预纯化(DCM:MeOH=96:4),得到粗产物,再经Prep-HPLC分离纯化,得到化合物11(7mg)。MS m/z(ESI):548.3[M+H] +
1H NMR(400MHz,DMSO-d 6))δ11.97(s,1H),9.65(s,1H),9.12(d,J=2.1Hz,1H),8.44(dd,J=8.9,2.2Hz,1H),8.37(d,J=1.8Hz,1H),7.91(dd,J=8.5,2.2Hz,1H),7.74(d,J=8.4Hz,1H),6.96-6.67(m,2H),6.31(s,1H),6.09(s,1H),3.85-3.68(m,4H),3.66-3.49(m,4H),2.56(s,3H),2.54(s,1H),2.33(s,3H),2.26(s,3H),2.19(s,3H),1.60(d,J=8.5Hz,1H).
实施例46:2-(6-(6-((6-(5-异丙基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物12)
Figure PCTCN2020074696-appb-000105
第一步:5-(1,3-二氧戊环-2-基)-2-(5-异丙基-1H-吡唑-1-基)吡啶(化合物12b)的制备
将化合物12a(210.68mg)、121a(400mg)、N,N'-二甲基乙二胺(153.27mg)、碳酸铯(1.13g)和CuI (331.13mg)依次加入到DMF(10mL)中,并加热至110℃搅拌3h。将反应液降至室温,加入水(30mL)稀释,并用DCM萃取(50mL x 2)。合并有机相,用水及饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩,并经硅胶快速柱层析分离纯化(PE:EA=55:45),得到化合物12b(300mg)。MS m/z(ESI):260.2[M+H] +
第二步:6-(5-异丙基-1H-吡唑-1-基)烟碱醛(化合物12c)的制备
将化合物12b(300.00mg)加入到氯化氢的1,4-二氧六环溶液(4N,6mL)和DCM(4mL)的混合溶液中,并于室温搅拌2h。将反应液减压浓缩,并经快速柱层析纯化(PE:EA=80:20),得到化合物12c(120mg)。MS m/z(ESI):216.2[M+H] +
第三步:2-(6-(6-((6-(5-异丙基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物12)的制备
将化合物12c(26.73mg)、1g(30mg)和氰基硼氢化钠(26.01mg)依次加入到甲醇(1mL)和乙酸(0.1mL)的混合溶剂中,并升温至40℃搅拌16h。加入饱和氯化铵水溶液(0.1mL)淬灭反应。将反应液浓缩,经Prep-HPLC分离纯化,得到化合物12的三氟乙酸盐(15mg),再经MPLC分离,得到游离态化合物12(8mg)。MS m/z(ESI):562.3[M+H] +.
MPLC条件:
仪器型号:Biotage Isolera Prime 2.3.1,色谱柱:Agela Technologies C18 spherical 20-35um 100A,12g;色谱柱温:25℃;流速:15.0mL/min;检测波长:254nm;洗脱梯度:(0min:0%A,100%B;3.0min:0%A,100%B;20min:80%A,20%B);流动相A:100%乙腈;流动相B:0.5%碳酸氢铵水溶液;化合物收集时间:10.4min-11.8min。
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.65(s,1H),9.16-9.09(m,1H),8.49-8.40(m,2H),8.36(d,J=1.9Hz,1H),7.93(dd,J=8.5,2.3Hz,1H),7.81(dd,J=8.4,0.7Hz,1H),6.78(d,J=9.0Hz,2H),6.42(d,J=2.5Hz,1H),6.33(s,1H),3.83-3.68(m,4H),3.66-3.50(m,4H),2.99(p,J=6.9Hz,1H),2.59-2.53(m,1H),2.33(s,3H),2.26(s,3H),1.60(d,J=8.4Hz,1H),1.27(s,3H),1.25(s,3H).
实施例47:2-(6-(6-(4-(5,6-二氢环戊烯并[c]吡唑-2(4H)-基)苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物25)
Figure PCTCN2020074696-appb-000106
第一步:2-(5-(1,3-二氧戊环-2-基)吡啶-2-基)-2,4,5,6-四氢环戊烯并[c]吡唑(化合物25b)的制备
将化合物25a(206.83mg)、化合物121a(400mg)、N,N'-二甲基乙二胺(153.27mg)、碳酸铯(1.13g)和CuI(331.13mg)依次加入到DMF(10mL)中,并加热至110℃搅拌12h。将反应液降至室温,加入水(50mL)稀释,并用DCM萃取(50mL x 2)。合并有机相,用水及饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩,得到粗品化合物25b(525mg)。MS(ESI,m/z):258.2[M+H] +
第二步:6-(5,6-二氢环戊烯并[c]吡唑-2(4H)-基)烟碱醛(化合物25c)的制备
将化合物25b(315mg)加入到氯化氢的1,4-二氧六环溶液(4N,10mL)和DCM(10mL)的混合溶液中,并于室温搅拌2h。将反应液减压浓缩,并经快速柱层析纯化(PE:EA=80:20),得到化合物25c(200mg)。MS m/z(ESI):214.2[M+H] +
第三步:2-(6-(6-(4-(5,6-二氢环戊烯并[c]吡唑-2(4H)-基)苄基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物25)的制备
将化合物25c(29.42mg)、1g(50mg)和氰基硼氢化钠(43.35mg)依次加入到甲醇(0.5mL)和乙酸(0.05mL)的混合溶剂中,并升温至40℃搅拌16h。加入饱和氯化铵水溶液(0.1mL)淬灭反应。将反应液浓缩,经Prep-HPLC分离纯化,得到化合物25的三氟乙酸盐(25mg),再经MPLC分离,得到游离态化合物25(15mg)。MS m/z(ESI):560.3[M+H] +
MPLC条件:
仪器型号:Biotage Isolera Prime 2.3.1,色谱柱:Agela Technologies C18 spherical 20-35um 100A,12g;色谱柱温:25℃;流速:15.0mL/min;检测波长:254nm;洗脱梯度:(0min:0%A,100% B;3.0min:0%A,100%B;20min:80%A,20%B);流动相A:100%乙腈;流动相B:0.5%碳酸氢铵水溶液;化合物收集时间:11.4min-12.8min。
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.65(s,1H),9.15-9.10(m,1H),8.44(dd,J=8.9,2.3Hz,1H),8.35-8.30(m,1H),8.19(d,J=1.2Hz,1H),7.89(dd,J=8.5,2.3Hz,1H),7.76(dd,J=8.4,0.7Hz,1H),6.78(d,J=9.0Hz,2H),6.31(s,1H),3.82-3.69(m,4H),3.66-3.50(m,4H),2.74-2.62(m,4H),2.57-2.54(m,2H),2.38(q,J=7.4Hz,2H),2.33(s,3H),2.26(s,3H),1.59(d,J=8.4Hz,1H).
实施例48:(6-(4-氟-1H-吡唑-1-基)吡啶-3-基)(3-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-基)甲酮(化合物26)
Figure PCTCN2020074696-appb-000107
第一步:6-(4-氟-1H-吡唑-1-基)烟酸甲酯(化合物26b)的制备
将化合物26a(1g)和91a(478.08mg)加入到乙腈(20mL)中,然后依次加入水杨醛肟(129.96mg)、碳酸铯(3.77g)和氧化亚铜(132.47mg),并于氮气保护下加热至85℃搅拌16h。反应结束后,向反应液中加入稀盐酸,调节至约pH=5。加入硅胶拌样,并经硅胶柱层析(DCM:MeOH=20:1)分离纯化,得到化合物26b(602mg)。MS m/z(ESI):221.9[M+H] +
第二步:6-(4-氟-1H-吡唑-1-基)烟酸(化合物26c)的制备
将化合物26b(580mg)加入到THF(10mL)和H 2O(5mL)中,然后加入NaOH(314.66mg),并于25℃搅拌14h。反应结束后,向反应液中加入稀盐酸,调节至约pH=5,并用EA萃取(80mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,得到化合物26c(312mg)。MS m/z(ESI):208.1[M+H] +
第三步:(6-(4-氟-1H-吡唑-1-基)吡啶-3-基)(3-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-基)甲酮(化合物26)的制备
将化合物26c(20mg)加入到DMF(5mL)中,然后依次加入HBTU(29.41mg)、DIPEA(37.43)和1g的三氟乙酸盐(50.60mg),于25℃搅拌1h。向反应液中加入水(25mL)淬灭反应,并用EA萃取(80mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,经Prep-HPLC分离纯化,得到化合物26(25mg)。MS m/z(ESI):551.8[M+H] +
1H NMR(400MHz,DMSO)δ11.97(s,1H),9.65(s,1H),9.04(s,1H),8.77-8.73(m,2H),8.38(dd,J=8.8,2.0Hz,1H),8.25(dd,J=8.4,2.0Hz,1H),8.03(d,J=4.4Hz,1H),7.97(d,J=8.8Hz,1H),7.05-6.47(m,2H),6.27(br,1H),4.96(s,1H),4.67(s,1H),4.17(d,J=9.6Hz,1H),3.80-3.65(m,2H),3.63-3.50(m,1H),2.92-2.82(m,1H),2.31(s,3H),2.24(s,3H),1.72(d,J=8.4Hz,1H).
实施例49:2-(6-(6-((6-(5-环丁氧基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物27)
Figure PCTCN2020074696-appb-000108
第一步:3-羟基-1H-吡唑-1-羧酸叔丁酯(化合物27b)的制备
在反应瓶中依次加入化合物27a(500mg)、Boc 2O(1.30g)、TEA(1.81g)和DMAP(145.31mg),然后加入THF(20mL),并于室温下搅拌16h,使得反应液由黄色浑浊逐渐变澄清。反应结束后,直接将反应液减压浓缩,并经硅胶快速柱层析分离纯化(DCM:MeOH=20:1),得到化合物27b(425mg)。
第二步:3-环丁氧基-1H-吡唑-1-羧酸叔丁酯(化合物27d)的制备
将化合物27b(360mg)、27c(215.71mg)和PPh 3(776.71mg)溶解于甲苯(10mL)中,并用冰水浴降温。加入DIAD(628.74mg),并于氮气保护下加热至110℃反应6h。反应结束后,将反应液冷却至室温,加入水(30mL)稀释,并用EA萃取。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩至干,并经硅胶快速柱层析分离纯化(EA:PE=1:20),得到化合物27d(374mg)。
第三步:3-环丁氧基-1H-吡唑(化合物27e)的制备
将化合物27d(374mg)溶解于甲醇(3mL)中,再加入氯化氢的1,4-二氧六环溶液(4N,3mL),并于氮气保护下室温反应。反应结束后,将反应液减压浓缩至干,得到化合物27e的盐酸盐(280mg)。MS m/z(ESI):139.1[M+H] +
第四步:2-(5-环丁氧基-1H-吡唑-1-基)-5-(1,3-二氧戊环-2-基)吡啶(化合物27f)的制备
在反应瓶中加入化合物121a(315mg)、化合物27e的盐酸盐(239mg)、Cs 2CO 3(675.94mg)和DMF(10mL),搅拌均匀后加入N,N'-二甲基乙二胺(48.77mg)及CuI(52.68mg,273.84μmol),并于氮气保护下加热至100℃反应14h。反应结束后,将反应液冷却至室温,加入水(30mL)稀释,并用EA萃取(40mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩至干,并经硅胶快速柱层析分离纯化(EA:PE=1:1),得到化合物27f(300mg)。MS m/z(ESI):287.9[M+H] +
第五步:6-(5-环丁氧基-1H-吡唑-1-基)烟碱醛(化合物27g)的制备
将化合物27f(300mg)溶解于THF(5mL)中,向溶液中加入盐酸(2N,5mL),并于室温反应8h。反应结束后,向反应液中加入水(20mL)稀释,用饱和NaHCO 3水溶液调节pH至7-8,并用EA萃取(40mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩至干,并经硅胶快速柱层析分离纯化(EA:PE=1:15),得到化合物27g(140mg)。MS m/z(ESI):243.9[M+H] +
第六步:2-(6-(6-((6-(5-环丁氧基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物27)的制备
将化合物1g(65mg)、27g(50.66mg)、钛酸四异丙酯(195.65mg)溶解于干燥的THF(3mL)中,并加热至75℃反应1h。向反应瓶中加入三乙酰氧基硼氢化钠(182.37mg),并保持75℃反应18h。反应结束后,将反应液冷却至室温,加入水(30mL)稀释,并用EA萃取(20mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩至干,并经Prep-HPLC纯化,得到化合物27(20mg)。MS m/z(ESI):589.9[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.98(s,1H),9.66(s,1H),9.13(d,J=2.4Hz,1H),8.44(dd,J=8.8,2.4Hz,1H),8.40(d,J=2.4Hz,1H),8.33(d,J=2.0Hz,1H),7.91(dd,J=8.4,2.4Hz,1H),7.66(d,J=8.4Hz,1H),7.12-6.69(m,2H),6.49-6.14(m,1H),6.02(d,J=2.8Hz,1H),4.90(p,J=7.6Hz,1H),3.83-3.73(m,2H),3.73-3.66(m,2H),3.65-3.47(m,4H),2.60-2.53(m,1H),2.46-2.38(m,2H),2.33(s,3H),2.26(s,3H),2.13-2.03(m,2H),1.82-1.73(m,1H),1.67-1.56(m,2H).
实施例50:2-(6-(6-((6-(4-氯-3-甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺和2-(6-(6-((6-(4-氯-5-甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物28/化合物28')
Figure PCTCN2020074696-appb-000109
第一步:2-(4-氯-3-甲基-1H-吡唑-1-基)-5-(1,3-二氧戊环-2-基)吡啶和2-(4-氯-5-甲基-1H-吡唑-1-基)-5-(1,3-二氧戊环-2-基)吡啶(化合物28b/化合物28b')的制备
于氮气保护下将化合物121a(202.0mg)、28a(102.3mg)、CuI(167.2mg)、N,N’-二甲基乙二胺(77.3mg)和Cs 2CO 3(856.1mg)溶解于DMF(5.0mL)中,并加热至120℃反应,直至原料完全转化。反应结束后,将反应液用饱和碳酸钠水溶液洗涤,并用EA萃取(30mL x 3)。合并有机相,用无水硫酸钠干燥,过滤后浓缩,经制备TLC分离纯化,得到化合物28b和28b'的混合物(230.0mg)。MS m/z(ESI):266.0[M+H] +
第二步:6-(4-氯-3-甲基-1H-吡唑-1-基)烟碱醛和6-(4-氯-5-甲基-1H-吡唑-1-基)烟碱醛(化合物28c/化合物28c')的制备
将化合物28b和28b'的混合物(230.0mg)加入到浓盐酸(3mL)、THF(10mL)和水(10mL)的混合溶剂中,并于25℃反应,直至原料完全转化。将反应液减压浓缩至干,得到化合物28c和28c'的混合物(39.0mg)。MS m/z(ESI):222.1[M+H] +
第三步:2-(6-(6-((6-(4-氯-3-甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺和2-(6-(6-((6-(4-氯-5-甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物28/化合物28')的制备
将化合物28c和28c'的混合物(29.0mg)和化合物1g的三氟乙酸盐(30.0mg)加入到甲醇(0.5mL)中,然后依次加入三乙胺(6.4mg)和氰基硼氢化钠(19.8mg),并于室温反应,直至原料全部转化。反应结束后,将反应液减压浓缩至干,并经Prep-HPLC分离纯化,得到化合物28(2.0mg,收集时间5.6-6.0分钟),MS m/z(ESI):568.2[M+H] +,和化合物28'(1.0mg,收集时间5.0-5.4分钟),MS m/z(ESI):568.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.98(s,1H),9.64(s,1H),9.13(d,J=2.0Hz,1H),8.69(s,1H),8.45(d,J=8.0Hz,1H),8.40(s,1H),7.98(d,J=8.8Hz,1H),7.82(d,J=8.4Hz,1H),6.89(d,J=8.8Hz,1H),6.76-6.67(m,1H),6.38-6.23(m,1H),3.80-3.55(m,8H),2.62-2.58(m,1H),2.34(s,3H),2.28(s,3H),2.27(s,3H),1.61(d,J=8.0Hz,1H)(化合物28).
1H NMR(400MHz,DMSO-d 6)δ12.03(s,1H),9.68(s,1H),9.18(d,J=2.4Hz,1H),8.51(d,J=2.4Hz,1H),8.48(d,J=2.4Hz,1H),8.05(dd,J=8.4,2.4Hz,1H),7.90(s,1H),7.83(d,J=8.4Hz,1H),6.85(d,J=9.2Hz,1H),6.82-6.69(m,1H),6.43-6.29(m,1H),3.86-3.56(m,8H),2.63(s,3H),2.62-2.58(m,1H),2.39(s,3H),2.31(s,3H),1.66(d,J=8.4Hz,1H)(化合物28').
实施例51:2-(6-(4-((5-氟吡啶-3-基)氧基)哌啶-1-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶- 4-胺(化合物29)
Figure PCTCN2020074696-appb-000110
第一步:4-((5-氟吡啶-3-基)氧基)哌啶-1-羧酸叔丁酯(化合物29b)的制备
于氮气保护下将化合物29a(425.6mg)、N-Boc-4-羟基哌啶(505.0mg)和PPh 3(1.3g)溶解于干燥的THF(5.0mL)中,降温至0℃,滴加DIAD(1.0g),然后缓慢升温至25℃反应,直至原料完全转化。反应结束后,将反应液减压浓缩至干,经柱层析分离纯化,得到化合物29b(736.2mg)。MS m/z(ESI):297.1[M+H] +
第二步:3-氟-5-(哌啶-4-基氧基)吡啶(化合物29c)盐酸盐的制备
将化合物29b(555.5mg)加入到氯化氢的1,4-二氧六环溶液(4N,20mL)中,于25℃反应,直至原料完全转化。将反应液减压浓缩至干,得到化合物29c的盐酸盐(390.0mg)。MS m/z(ESI):197.1[M+H] +
第三步:2-(6-(4-((5-氟吡啶-3-基)氧基)哌啶-1-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物29)的制备
将化合物29c的盐酸盐(86.0mg)、化合物119d(55.5mg)和K 2CO 3(122.7mg)加入到DMF(5mL)中,并加热至125℃反应,直至原料完全转化。反应结束后,将反应液用饱和碳酸钠水溶液洗涤,并用EA萃取(10mL x 3)。合并有机层,用无水硫酸钠干燥,过滤后浓缩,并经Prep-HPLC分离纯化,得到化合物29(17.0mg)。MS m/z(ESI):460.9[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.41(s,1H),11.12(s,1H),8.97(d,J=2.4Hz,1H),8.31(dd,J=9.2,2.4Hz,1H),8.25(t,J=1.6Hz,1H),8.20(d,J=2.4Hz,1H),7.59(dt,J=11.6,2.4Hz,1H),7.14(d,J=9.2Hz,1H),6.90-6.80(m,2H),4.87-4.81(m,1H),4.19-4.13(m,2H),3.85-3.73(m,2H),2.47(s,3H),2.28(s,3H),2.11-2.05(m,2H),1.72-1.63(m,2H).
实施例52:2-(6-(6-((R)-1-(6-(4-氟-1H-吡唑-1-基)吡啶-3-基)乙基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺和2-(6-(6-((S)-1-(6-(4-氟-1H-吡唑-1-基)吡啶-3-基)乙基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物52-1/化合物52-2)
Figure PCTCN2020074696-appb-000111
化合物52(100mg)经过手性Prep-HPLC拆分,得到化合物52-1(保留时间6.788min)和化合物52-2(保留时间10.115min)。两个化合物根据手性拆分保留时间先后进行区分定义。由此得到化合物52-1(14mg,ee:98.85%),MS m/z(ESI):552.3[M+H] +;化合物52-2(20mg,ee:99.22%),MS m/z(ESI):552.3[M+H] +
手性HPLC拆分条件:
仪器型号:Shimadzu LC-20AD;色谱柱:CHIRALPAK IE(IE00CD-RH008),0.46cm I.D.×15cm L;色谱柱温:35℃;流速:1.0mL/min;检测波长:254nm;流动相:MeOH:CAN:DEA=80:20:0.1(V/V/V);化合物52-1出峰时间为6.788min,化合物52-2出峰时间为10.115min。
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.65(s,1H),9.11(d,J=2.0Hz,1H),8.66(d,J=4.6Hz,1H),8.43(d,J=2Hz,1H),8.41(d,J=2.4Hz,1H),8.01(dd,J=8.5,1.9Hz,1H),7.90(dd,J=16.8, 6.4Hz,2H),6.82(br,1H),6.75(d,J=8.8Hz,1H),6.29(br,1H),3.95-3.81(m,2H),3.77(q,J=6.1Hz,1H),3.63(s,1H),3.53-3.38(m,3H),2.55-2.53(m,1H),2.32(s,3H),2.25(s,3H),1.55(d,J=8.4Hz,1H),1.22(d,J=6.2Hz,3H).(化合物52-1)
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.65(s,1H),9.11(d,J=2.0Hz,1H),8.66(d,J=4.6Hz,1H),8.43(d,J=2Hz,1H),8.41(d,J=2.4Hz,1H),8.00(dd,J=8.5,1.9Hz,1H),7.89(dd,J=16.8,6.4Hz,2H),6.82(br,1H),6.75(d,J=8.8Hz,1H),6.27(br,1H),3.95-3.81(m,2H),3.76(q,J=6.1Hz,1H),3.62(s,1H),3.53-3.38(m,3H),2.55-2.51(m,1H),2.32(s,3H),2.25(s,3H),1.54(d,J=8.4Hz,1H),1.21(d,J=6.2Hz,3H).(化合物52-2)
实施例53:2-(6-(6-((6-(5-环丙氧基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物30)
Figure PCTCN2020074696-appb-000112
第一步:3-环丙氧基-1H-吡唑-1-羧酸叔丁酯(化合物30b)的制备
将化合物27b(300mg)、30a(141.89mg)和三苯基膦(640.09mg)加入甲苯(10mL)中,并降温至0℃。滴加DIAD(493.51mg),并加热至110℃反应6h。将反应液降温至室温,减压浓缩,并经硅胶快速柱层析纯化(PE:EA=23:77),得到化合物30b(60mg)。
第二步:3-环丙氧基-1H-吡唑(化合物30c)的制备
将化合物30b(102mg)加入到氯化氢的1,4-二氧六环溶液(4N,2mL)和THF(2mL)的混合溶液中,并于室温搅拌16h。将反应液减压浓缩,得到化合物30c的盐酸盐(73mg)。MS m/z(ESI):125.1[M+H] +
第三步:2-(5-环丙氧基-1H-吡唑-1-基)-5-(1,3-二氧戊环-2-基)吡啶(化合物30d)的制备
将化合物30c的盐酸盐(69.81mg)、化合物121a(100mg)、N,N'-二甲基乙二胺(38.32mg)、碳酸铯(424.87mg)和CuI(82.78mg)依次加入到DMF(10mL)中,并加热至110℃搅拌3h。将反应液降至室温,加入水(30mL)稀释,并用DCM萃取(50mL x 2)。合并有机相,用水及饱和食盐水洗涤,无水硫酸钠干燥,过滤并减压浓缩,并用Prep-HPLC分离纯化,得到化合物30d(80mg)。MS m/z(ESI):273.9[M+H] +
第四步:6-(5-环丙氧基-1H-吡唑-1-基)烟碱醛(化合物30e)的制备
将化合物30d(80mg)加入到氯化氢的1,4-二氧六环溶液(4N,3mL)和EA(2mL)的混合溶液中,并于室温搅拌2h。将反应液减压浓缩,用DCM(30mL)稀释。用饱和碳酸氢钠水溶液及饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩,得到化合物30e(55mg)。MS m/z(ESI):229.9[M+H] +
第五步:2-(6-(6-((6-(5-环丙氧基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物30)的制备
将化合物30e(40mg)、1g(40mg)和钛酸四异丙酯(26.01mg)置于5mL反应瓶内,加入干燥的THF(2mL),并加热至75℃搅拌16h。然后加入三乙酰氧基硼氢化钠(26.01mg),并于75℃搅拌8h。将反应液降至室温,加入饱和氯化铵水溶液(0.1mL)淬灭反应。将反应液浓缩,并经制备TLC预 纯化(DCM:MeOH=10:1),得到粗产物25mg(R f=0.35-0.45),再经Prep-HPLC分离纯化,得到化合物30(10mg)。MS m/z(ESI):575.9[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.95(s,1H),9.62(s,1H),9.12(d,J=2.3Hz,1H),8.46-8.41(m,2H),8.34(d,J=2.2Hz,1H),7.92(dd,J=8.4,2.2Hz,1H),7.67(d,J=8.4Hz,1H),6.78(d,J=9.0Hz,2H),6.29(s,1H),6.17(d,J=2.7Hz,1H),4.10(tt,J=6.0,3.2Hz,1H),3.84-3.68(m,4H),3.66-3.48(m,4H),2.60-2.54(m,1H),2.33(s,3H),2.26(s,3H),1.59(d,J=8.4Hz,1H),0.79-0.68(m,4H).
实施例54:2-(6-(6-((6-(3-(氟甲基)-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物31)
Figure PCTCN2020074696-appb-000113
第一步:1-(5-(1,3-二氧戊环-2-基)吡啶-2-基)-1H-吡唑-3-羧酸甲酯(化合物31b)的制备
向反应瓶中加入化合物121a(2.0g)、31a(1.12g)、Cs 2CO 3(5.72g)、反式-N,N'-二甲基环己二胺(504.72mg)、CuI(337.89mg)和DMF(10mL),并于氮气保护下加热至90℃反应2h。反应结束后,将反应液冷却至室温,加入水(20mL)稀释,并用EA萃取(30mL x 3)。合并有机相,用水洗涤,无水硫酸钠干燥,过滤后减压浓缩至干,并经硅胶快速柱层析分离纯化(EA:PE=20:80),得到化合物31b(2.2g)。MS m/z(ESI):276.0[M+H] +
第二步:(1-(5-(1,3-二氧戊环-2-基)吡啶-2-基)-1H-吡唑-3-基)甲醇(化合物31c)的制备
将化合物31b(2.2g)溶解于THF(20mL)中,并降温至-20℃。向反应液中分批缓慢加入LiAlH 4(464.31mg),保持在该温度下反应15min。反应结束后,缓慢滴加EA(1mL)消耗过量的LiAlH 4。然后滴加水(1mL)淬灭反应,加水(20mL)稀释,并用EA萃取(30mL x 3)。合并有机相,用无水硫酸钠干燥,过滤后减压浓缩至干,并经硅胶快速柱层析分离纯化(EA:PE=50:50),得到化合物31c(1.24g)。MS m/z(ESI):248.0[M+H] +
第三步:5-(1,3-二氧戊环-2-基)-2-(3-(氟甲基)-1H-吡唑-1-基)吡啶(化合物31d)的制备
于氮气保护下将干燥的DCM(20mL)降温至-40℃,然后缓慢滴加双(2-甲氧基乙基)氨基三氟化硫(2.86g)。将化合物31c(0.8g)的DCM溶液(20mL)滴加到反应液中,然后将反应液缓慢升温至25℃反应20h。反应结束后,将反应液倒入饱和碳酸氢钠水溶液中,待气泡释放完成后,用DCM萃取(20mL x 3)。合并有机相,用无水硫酸钠干燥,过滤后减压浓缩至干,并经硅胶快速柱层析分离纯化(EA:PE=20:80),得到化合物31d(200mg)。MS m/z(ESI):249.9[M+H] +
第四步:6-(3-(氟甲基)-1H-吡唑-1-基)烟碱醛(化合物31e)的制备
将化合物31d(200mg)溶解于THF(5mL)中,向溶液中加入盐酸(2N,2.3mL),并于室温反应16h。反应结束后,向反应液中加入水(20mL)稀释,并用饱和NaHCO 3水溶液调节pH至7-8。直接浓缩,并经硅胶快速柱层析分离纯化(EA:PE=15:85),得到化合物31e(110mg)。MS m/z(ESI):206.0[M+H] +
第五步:2-(6-(6-((6-(3-(氟甲基)-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物31)的制备
将化合物1g(87mg)、31e(110mg)、钛酸四异丙酯(278.46mg)溶解于干燥的THF(10mL)中,并加热至75℃反应8h。将反应液降温至25℃,向反应瓶中加入三乙酰氧基硼氢化钠(259.57mg),并保持25℃反应12h。滴加水(1mL)淬灭反应,浓缩后经硅胶快速柱层析预纯化(DCM:MeOH=90:10),再经Prep-HPLC分离纯化,得到化合物31(25mg)。MS m/z(ESI):551.8[M+H] +
1H NMR(400MHz,DMSO-d 6)δ9.63(s,1H),9.13(d,J=2.0Hz,1H),8.62(d,J=2.4Hz,1H),8.49-8.39(m,2H),8.15(s,1H),7.99(dd,J=8.4,1.6Hz,1H),7.90-7.84(m,1H),7.07-6.74(m,2H),6.70(s,1H),6.30(br,1H),5.53(s,1H),5.41(s,1H),3.81-3.73(m,4H),3.67-3.58(m,4H),2.63-2.53(m,1H),2.34(s,3H),2.26(s,3H),1.61(d,J=8.4Hz,1H).
实施例55:2-(6-(6-((6-(5-乙基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物46)
Figure PCTCN2020074696-appb-000114
第一步:1-(四氢-2H-吡喃-2-基)-1H-吡唑(化合物46a)的制备
将化合物86a(2g)加入到3,4-二氢吡喃(6mL)中,然后加入TFA(334.97mg),并加热至100℃搅拌16h。向反应液中加入水(100mL)淬灭反应,并用EA萃取(50mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,并经硅胶柱层析(PE:EA=60:1~10:1)分离纯化,得到化合物46a(2.4g)。MS m/z(ESI):153.2[M+H] +
第二步:5-乙基-1-(四氢-2H-吡喃-2-基)-1H-吡唑(化合物46b)的制备
将化合物46a(2g)加入到干燥的THF(30mL)中,然后于-78℃加入正丁基锂(2.5M,6.31mL),并搅拌1h。然后于-78℃缓慢加入碘乙烷(3.07g),缓慢升温至室温搅拌5h。向反应液中加入甲醇(50mL)淬灭反应,直接硅胶拌样,并经柱层析(PE:EA=40:1~5:1)分离纯化,得到化合物46b(721mg)。MS m/z(ESI):181.0[M+H] +
第三步:5-乙基-1H-吡唑(化合物46c)的制备
将化合物46b(721mg)加入到MeOH(7mL)中,然后加入氯化氢的1,4-二氧六环溶液(4N,1mL),并于25℃搅拌1h。向反应液中加入饱和碳酸氢钠水溶液(50mL)淬灭反应,并用EA萃取(80mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后旋干有机相,得到化合物46c(185mg)。MS m/z(ESI):97.1[M+H] +
第四步:5-(1,3-二氧戊环-2-基)-2-(5-乙基-1H-吡唑-1-基)吡啶(化合物46d)的制备
将化合物121a(401mg)和46c(184.31mg)加入到DMF(15mL)中,然后依次加入N,N-二甲基乙二胺(153.39mg)、碳酸铯(1.7g)和CuI(331.96mg),并于氮气保护下加热至110℃搅拌10h。向反应液中加入水(100mL)淬灭反应,并用EA萃取(50mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,并经硅胶柱层析(PE:EA=40:1~3:1)分离纯化,得到化合物46d(199mg)。MS m/z(ESI):245.9[M+H] +
第五步:6-(5-乙基-1H-吡唑-1-基)烟碱醛(化合物46e)的制备
将化合物46d(200mg)加入到THF(4mL)和H 2O(2mL)中,然后加入氯化氢的1,4-二氧六环溶液(4N,1mL),并于25℃搅拌5h。向反应液中加入饱和碳酸氢钠水溶液(50mL)淬灭反应,并用EA萃取(50mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,并经硅胶柱层析(PE:EA=40:1~3:1)分离纯化,得到化合物46c(91mg)。MS m/z(ESI):202.1[M+H] +
第六步:2-(6-(6-((6-(5-乙基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物46)的制备
将化合物1g(40.93mg)、46e(25mg)和钛酸四异丙酯(128.41mg)加入到干燥的THF(10mL)中,在氮气置换三次后,于75℃搅拌10h。然后分批加入三乙酰氧基硼氢化钠(119.69mg),继续于75℃搅拌6h。反应结束后,将反应液减压浓缩至干,经Prep-HPLC分离纯化,得到化合物46(8mg)。MS m/z(ESI):547.9[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.98(s,1H),9.65(s,1H),9.12(d,J=2.0Hz,1H),8.47(d,J=2.4Hz,1H),8.44(dd,J=9.20,2.4Hz,1H),8.36(d,J=1.6Hz,1H),7.93(dd,J=8.4,2.0Hz,1H),7.81(d,J=8.4Hz,1H),7.20-6.69(m,2H),6.40(d,J=2.8Hz,1H),6.32(s,1H),3.83-3.68(m,4H),3.66-3.53(m,4H),2.66(q,J=7.6Hz,2H),2.61-2.53(m,1H),2.33(s,3H),2.25(s,3H),1.60(d,J=8.4Hz,1H),1.23(t,J=7.6Hz,3H).
实施例56:2-(6-(6-((6-(4-氟-5-甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物122)
Figure PCTCN2020074696-appb-000115
第一步:4-氟-1-(四氢-2H-吡喃-2-基)-1H-吡唑(化合物122a)的制备
将化合物91a(500mg)加入到3,4-二氢吡喃(2.5mL)中,然后加入TFA(129.96mg),并加热至100℃搅拌16h。向反应液中加入水(100mL)淬灭反应,并用EA萃取(50mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,并经硅胶柱层析(PE:EA=50:1~10:1)分离纯化,得到化合物122a(901mg)。MS m/z(ESI):171.1[M+H] +
第二步:4-氟-5-甲基-1-(四氢-2H-吡喃-2-基)-1H吡唑(化合物122b)的制备
将化合物122a(900mg)加入到干燥的THF(10mL)中,然后于-78℃加入正丁基锂(2.5M,2.33mL),并搅拌1h。然后于-78℃缓慢加入碘甲烷(1.13g),缓慢升温至室温搅拌3h。向反应液中加入甲醇淬灭反应,并用EA萃取(80 x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,并经硅胶柱层析(DCM:MeOH=60:1~10:1)分离纯化,得到化合物122b(845mg),为淡黄色液体。MS m/z(ESI):185.0[M+H] +
第三步:4-氟-5-甲基-1H-吡唑(化合物122c)的制备
将化合物122b(800mg)加入到MeOH(7mL)中,然后加入氯化氢的1,4-二氧六环溶液(4N,7mL),并于25℃搅拌1h。向反应液中加入饱和碳酸氢钠水溶液(50mL)淬灭反应,并用EA萃取(80mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,得到化合物122c(300mg)。MS m/z(ESI):101.2[M+H] +
第四步:5-(1,3-二氧戊环-2-基)-2-(4-氟-5-甲基-1H-吡唑-1-基)吡啶(化合物122d)的制备
将化合物121a(300mg)和122c(143.58mg)加入到DMF(15mL)中,然后依次加入N,N-二甲基乙二胺(114.75mg)、碳酸铯(1.27g)和CuI(248.35mg),并于氮气保护下加热至110℃搅拌10h。向反应液中加入水(100mL)淬灭反应,并用EA萃取(50mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,并经硅胶柱层析(PE:EA=50:1~10:1)分离纯化,得到化合物122d(254mg)。MS m/z(ESI):249.9[M+H] +
第五步:6-(4-氟-5-甲基-1H-吡唑-1-基)烟碱醛(化合物122e)的制备
将化合物122d(240mg)加入到THF(5mL)和H 2O(2mL)中,然后加入氯化氢的1,4-二氧六环溶 液(4N,991.74uL),并于25℃搅拌5h。向反应液中加入饱和碳酸氢钠水溶液(50mL)淬灭反应,并用EA萃取(50mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,得到化合物122e(55mg)。MS m/z(ESI):205.9[M+H] +
第六步:2-(6-(6-((6-(4-氟-5-甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物122)的制备
将化合物1g(101mg)、122e(51.98mg)和钛酸四异丙酯(288.01mg)加入到干燥的THF(25mL)中,在氮气置换三次后,于75℃搅拌10h。然后分批加入三乙酰氧基硼氢化钠(268.46mg),继续于75℃搅拌6h。反应结束后,将反应液减压浓缩至干,经Prep-HPLC分离纯化,得到化合物122(22mg)。MS m/z(ESI):551.8[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.99(s,1H),9.65(s,1H),9.13(d,J=4.0Hz,1H),8.57(d,J=4.8Hz,1H),8.44(dd,J=8.8,2.0Hz,1H),8.37(s,1H),7.94(dd,J=8.4,1.6Hz,1H),7.80(d,J=8.4Hz,1H),6.97-6.74(m,2H),6.31(s,1H),3.81-3.69(m,4H),3.66-3.52(m,4H),2.59-2.53(m,1H),2.33(s,3H),2.28(s,3H),2.24(s,3H),1.59(d,J=8.4Hz,1H).
实施例57:2-(6-(6-((6-(1H-吡咯-2-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物123)
Figure PCTCN2020074696-appb-000116
第一步:2-(5-甲酰基吡啶-2-基)-1H-吡咯-1-羧酸叔丁酯(化合物123b)的制备
将化合物123a(1.13g)、8c(1.0g)、四三苯基磷钯(310.62mg)和碳酸钠(1.71g)加入到1,4-二氧六环(60mL)和水(15mL)中,并于氮气保护下加热至95℃反应14h。反应结束后,浓缩除去部分有机溶剂,并用EA萃取。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩,并经硅胶柱层析(PE:EA=5:1)分离纯化,得到化合物123b(1.25g)。
第二步:2-(5-((3-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-基)甲基)吡啶-2-基)-1H-吡咯-1-羧酸叔丁酯(化合物123c)的制备
将化合物1g(50mg)、123b(37.57mg)和钛酸四异丙酯(156.84mg)加入到干燥的THF(5mL)中,并于72℃搅拌18h。然后加入三乙酰氧基硼氢化钠(146.19mg),并于72℃反应4h。反应结束后,直接硅胶柱层析(DCM:MeOH=93:7)纯化,得到化合物123c(40mg)。MS m/z(ESI):618.9[M+H] +
第三步:2-(6-(6-((6-(1H-吡咯-2-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物123)的制备
将氯化氢的1,4-二氧六环溶液(4N,2.0mL)滴加到化合物123c(40mg)的甲醇(2mL)溶液中,并于室温反应2h。反应结束后,将反应液浓缩至干。将粗产物溶解于甲醇中,加入过量碳酸钾,并于室温搅拌0.5h,将盐酸盐游离。过滤后减压浓缩,经Prep-HPLC分离纯化,得到化合物123(22mg)。MS m/z(ESI):518.9[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.98(s,1H),11.43(s,1H),9.66(s,1H),9.12(d,J=2.1Hz,1H),8.49-8.35(m,2H),7.70(d,J=7.9Hz,1H),7.61(d,J=8.2Hz,1H),6.93-6.59(m,4H),6.31(s,1H),6.12(dd,J=5.7,2.5Hz,1H),3.85-3.68(m,4H),3.65-3.49(m,4H),2.59-2.54(m,1H),2.33(s,3H),2.26(s,3H),1.59(d,J=8.2Hz,1H).
实施例58:2-(6-(6-((6-(3-环丙基-4-氟-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物124)
Figure PCTCN2020074696-appb-000117
第一步:4-氟-3-碘-1H-吡唑(化合物124a)的制备
向反应瓶中依次加入4-氟吡唑(1.5g)、NIS(4.31g)和氯仿(30mL),并于氮气保护下80℃搅拌7h。反应结束后,将反应液降至室温,硅胶直接拌样,并经快速硅胶柱层析(PE:EA=4:1)分离纯化,得到化合物124a(1.2g)。MS m/z(ESI):213[M+H] +
第二步:5-(1,3-二氧戊环-2-基)-2-(4-氟-3-碘-1H-吡唑-1-基)吡啶(化合物124b)的制备
向反应瓶中依次加入化合物121a(576.33mg)、124a(354mg)、碳酸铯(1.63g)和DMF(15mL),并于90℃搅拌12h。反应结束后,将反应液降至室温,加水(20mL)稀释,并用EA萃取。将有机相用水洗涤三次,无水硫酸钠干燥,过滤后浓缩,并经C 18柱分离纯化(乙腈:0.05%碳酸氢铵水溶液=68:32),得到化合物124b(300mg)。MS m/z(ESI):362[M+H] +
第三步:2-(3-环丙基-4-氟-1H-吡唑-1-基)-5-(1,3-二氧戊环-2-基)吡啶(化合物124c)的制备
向反应瓶中依次加入环丙基硼酸(89.20mg)、化合物124b(125mg)、醋酸钯(15.54mg)、磷酸钾(257.17mg)、三环己基膦(9.71mg)、甲苯(5mL)和水(1mL),并于氮气保护下90℃搅拌12h。反应结束后,将反应液降至室温,直接硅胶拌样,并经硅胶柱层析(PE:EA=3:1)分离纯化,得到化合物124c(70mg)。MS m/z(ESI):276[M+H] +
第四步:6-(3-环丙基-4-氟-1H-吡唑-1-基)烟碱醛(化合物124d)的制备
将化合物124c(70mg)溶解于THF(8mL)和水(8mL)的混合溶液中,滴入浓盐酸(5mL,37%),并于25℃搅拌18h。反应结束后,旋出部分溶剂,用饱和碳酸氢钠水溶液调节反应液pH至约9,并用EA萃取。有机层用无水硫酸钠干燥,过滤后浓缩,并经硅胶快速柱层析(PE:EA=3:1)分离纯化,得到化合物124d(18mg)。MS m/z(ESI):232[M+H] +
第五步:2-(6-(6-((6-(3-环丙基-4-氟-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物124)的制备
向反应瓶中依次加入化合物124d(18mg)、1g(31.04mg)、钛酸异丙酯(88.50mg)和干燥的THF(5mL),并于氮气保护下75℃搅拌18h。然后分批加入三乙酰氧基硼氢化钠(82.49mg),并继续于75℃搅拌6h。反应结束后,旋出部分溶剂,用饱和碳酸氢钠溶液调节反应液pH至约9,并用EA萃取。有机层用无水硫酸钠干燥,过滤后浓缩,并经制备TLC预纯化(DCM:MeOH=9:1),再经Prep-HPLC分离纯化,得到化合物124(15mg)。MS m/z(ESI):577.9[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.98(s,1H),9.65(s,1H),9.12(d,J=2.1Hz,1H),8.55(d,J=4.6Hz,1H),8.44(dd,J=8.9,2.2Hz,1H),8.35(d,J=1.5Hz,1H),7.93(dd,J=8.5,2.0Hz,1H),7.76(d,J=8.5Hz,1H),6.82(br,1H),6.79(d,J=12Hz,1H),6.30(br,1H),3.84-3.75(m,4H),3.73-3.49(m,4H),2.59-2.53(m,1H),2.33(s,3H),2.26(s,3H),2.02-1.93(m,1H),1.59(d,J=8.4Hz,1H),1.03-0.95(m,2H),0.94-0.86(m,2H).
实施例59:2-(6-(6-((R)-1-(6-(1H-吡唑-1-基)吡啶-3-基)乙基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺和2-(6-(6-((S)-1-(6-(1H-吡唑-1-基)吡啶-3-基)乙基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物125-1/化合物125-2)
Figure PCTCN2020074696-appb-000118
第一步:1-(6-(1H-吡唑-1-基)吡啶-3-基)乙酮(化合物125b)的制备
将化合物86a(328.18mg)、125a(500mg)和碳酸铯(1.57g)依次加入DMSO(5mL)中,并加热至100℃搅拌2h。将反应液降至室温,加入水稀释,并用EA萃取。有机相用水及饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩,并经硅胶快速柱层析纯化(PE:EA=50:50),得到化合物125b(364mg)。MS m/z(ESI):188.1[M+H] +
第二步:2-(6-(6-(1-(6-(1H-吡唑-1-基)吡啶-3-基)乙基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物125)的制备
将化合物125b(309.90mg)、1g(400mg)和钛酸四异丙酯(1.25g)置于250mL反应瓶内,加入干燥的THF(20mL),并加热至75℃搅拌16h。然后加入三乙酰氧基硼氢化钠(1.17g),并于75℃搅拌2h。将反应液降至室温,加入饱和氯化铵水溶液(3mL)淬灭反应。将反应液浓缩,并经硅胶快速柱层析分离纯化(DCM:MeOH=9:1),得到化合物125(270mg)。MS m/z(ESI):533.9[M+H] +
第三步:2-(6-(6-((R)-1-(6-(1H-吡唑-1-基)吡啶-3-基)乙基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺和2-(6-(6-((S)-1-(6-(1H-吡唑-1-基)吡啶-3-基)乙基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物125-1/化合物125-2)的制备
化合物125(250mg)经过手性Prep-HPLC拆分,得到化合物125-1(保留时间7.647min)和化合物125-2(保留时间11.638min)。两个化合物根据手性拆分保留时间先后进行区分定义,未进行立体结构鉴定。由此得到化合物125-1(101mg,ee:100%),MS m/z(ESI):533.9[M+H] +;化合物125-2(100mg,ee:99.93%),MS m/z(ESI):533.9[M+H] +
手性HPLC拆分条件:
仪器型号:Shimadzu LC-20AD;色谱柱:CHIRALPAK IE-3(IE30CD-UL006),0.46cm I.D.×15cm L;色谱柱温:25℃;流速:1.0mL/min;检测波长:254nm;流动相:MeOH:ACN:DEA=70:30:0.1(V/V/V);化合物125-1出峰时间为7.647min,化合物125-2出峰时间为11.638min。
1H NMR(400MHz,DMSO-d 6)δ11.98(s,1H),9.66(s,1H),9.12(d,J=2.3Hz,1H),8.59(s,1H),8.47-8.39(m,2H),7.99(d,J=6.4Hz,1H),7.89(d,J=7.4Hz,1H),7.81(s,1H),6.75(d,J=9.0Hz,2H),6.56(s,1H),6.31(s,1H),3.96-3.83(m,2H),3.75(d,J=3.3Hz,1H),3.63(s,1H),3.54-3.36(m,3H),2.57-2.53(m,1H),2.33(s,3H),2.25(s,3H),1.54(d,J=6.9Hz,1H),1.22(d,J=2.5Hz,3H).(化合物125-1)
1H NMR(400MHz,DMSO-d 6)δ11.98(s,1H),9.66(s,1H),9.12(d,J=2.3Hz,1H),8.59(d,J=2.6Hz,1H),8.47-8.39(m,2H),8.00(dd,J=8.5,2.2Hz,1H),7.90(d,J=8.4Hz,1H),7.81(d,J=1.6Hz,1H),6.75(d,J=9.0Hz,2H),6.57(t,J=2.1Hz,1H),6.31(s,1H),3.96-3.83(m,2H),3.76(q,J=6.2Hz,1H),3.63(s,1H),3.54-3.36(m,3H),2.57-2.53(m,1H),2.33(s,3H),2.25(s,3H),1.55(d,J=8.4Hz,1H),1.23(d,J=6.1Hz,3H).(化合物125-2)
实施例60:2-(6-(6-((6-(4-氟甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物126)和(1-(5-((3-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-基)甲基)吡啶-2-基)-1H-吡唑-4-基)甲醇(化合物127)
Figure PCTCN2020074696-appb-000119
第一步:1-(5-(1,3-二氧戊环-2-基)吡啶-2-基)-1H-吡唑-4-羧酸乙酯(化合物126b)的制备
在反应瓶中加入化合物121a(3.0g)、126a(1.86g)、Cs 2CO 3(8.67g)、反式-N,N'-二甲基环己二胺(757.08mg)、CuI(506.84mg)和DMF(15mL),并于氮气保护下加热至90℃反应2h。反应结束后,将反应液冷却至室温,加入水(30mL)稀释,并用EA萃取(30mL x 3)。合并有机相,用水洗涤,无水硫酸钠干燥,过滤后减压浓缩至干,并经硅胶快速柱层析分离纯化(EA:PE=20:80),得到化合物126b(3.31g)。MS m/z(ESI):290.0[M+H] +
第二步:1-(5-(1,3-二氧戊环-2-基)吡啶-2-基)-1H-吡唑-4-基)甲醇(化合物126c)的制备
将化合物126b(3.31g)溶解于THF(30mL)中,并降温至-20℃。向反应液中分批缓慢加入LiAlH 4(651.40mg,16.99mmol),保持在该温度下反应15min。反应结束后,缓慢滴加EA(2mL)消耗过量的LiAlH 4。然后滴加水(1mL)淬灭反应,加水(20mL)稀释,并用EA萃取(30mL x 3)。合并有机相,用无水硫酸钠干燥,过滤后减压浓缩至干,并经硅胶快速柱层析分离纯化(EA:PE=50:50),得到化合物126c(2.30g)。MS m/z(ESI):248.0[M+H] +
第三步:5-(1,3-二氧戊环-2-基)-2-(4-(氟甲基)-1H-吡唑-1-基)吡啶(化合物126d)的制备
于氮气保护下将干燥的DCM(30mL)降温至-40℃,然后缓慢滴加二乙胺基三氟化硫(6.06g)。将化合物126c(2.30g)的DCM溶液(30mL)滴加到反应液中,然后将反应液缓慢升温至25℃反应20h。反应结束后,将反应液倒入饱和碳酸氢钠水溶液中,待气泡释放完成后,用DCM萃取(30mL x 3)。合并有机相,用无水硫酸钠干燥,过滤后减压浓缩至干,并经硅胶快速柱层析分离纯化(EA:PE=20:80),得到化合物126d(684mg)。MS m/z(ESI):249.9[M+H] +
第四步:6-(4-(氟甲基)-1H-吡唑-1-基)烟碱醛(化合物126e)的制备
将化合物126d(684mg)溶解于THF(10mL)中,向溶液中加入盐酸(1N,5mL),并于25℃反应16h。反应结束后,向反应液中加入水(20mL)稀释,并用饱和NaHCO 3水溶液调节pH至7-8。直接浓缩除去THF,然后加入DCM萃取(30mL x 3)。合并有机相,用无水硫酸钠干燥,抽滤并浓缩,得到化合物126e(460mg)。MS m/z(ESI):206.0[M+H] +
第五步:2-(6-(6-((6-(4-氟甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物126)和(1-(5-((3-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-基)甲基)吡啶-2-基)-1H-吡唑-4-基)甲醇(化合物127)的制备
将化合物1g(200mg)、126e(182mg)、钛酸四异丙酯(640mg)溶解于干燥的THF(40mL)中,并加热至75℃反应8h。将反应液降温至25℃,向反应瓶中加入三乙酰氧基硼氢化钠(597mg),并保持25℃反应12h。滴加水(1mL)淬灭反应,浓缩后经硅胶快速柱层析分离预纯化(DCM:MeOH=90:10),得到粗产物(化合物126和127的混合物),再经Prep-HPLC分离纯化,得到化合物126(21mg),MSm/z(ESI):552.3[M+H] +;化合物127(19mg),MS m/z(ESI):550.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.00(br,1H),9.65(s,1H),9.13(d,J=2.2Hz,1H),8.77(d,J=3.2Hz,1H),8.44(dd,J=8.8,2.2Hz,2H),7.99(dd,J=8.4,2.0Hz,1H),7.94(s,1H),7.91-7.96(m,1H),6.95- 6.69(m,2H),6.30(br,1H),5.46(s,1H),5.34(s,1H),3.82-3.70(m,4H),3.68-3.52(m,4H),2.59-2.54(m,1H),2.33(s,3H),2.26(s,3H),1.60(d,J=8.4Hz,1H).(化合物126)
1H NMR(400MHz,DMSO-d 6)δ12.04(br,1H),9.66(s,1H),9.13(d,J=2.2Hz,1H),8.52-8.35(m,3H),7.95(dd,J=8.4,1.6Hz,1H),7.88-7.81(m,1H),7.73(s,1H),6.99-6.64(m,2H),6.31(br,1H),5.05(br,1H),4.45(s,2H),3.93-3.70(m,4H),3.67-3.51(m,4H),2.59-2.53(m,1H),2.33(s,3H),2.26(s,3H),1.60(d,J=8.4Hz,1H).(化合物127)
实施例61:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-((6-(1-甲基-1H-吡咯-2-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物128)
Figure PCTCN2020074696-appb-000120
第一步:6-(1H-吡咯-2-基)烟碱醛(化合物128a)的制备
将氯化氢的1,4-二氧六环溶液(4N,6.0mL)滴加到化合物123b(500mg)的甲醇(10mL)溶液中,并于室温反应2h。反应结束后,将反应液浓缩至干。将粗产物溶解于甲醇中,加入过量碳酸钾,并于室温搅拌0.5h,将盐酸盐游离。过滤后减压浓缩,经硅胶柱层析分离纯化(PE:EA=4:1),得到化合物128a(250mg)。MS m/z(ESI):173.0[M+H] +
第二步:5-(1,3-二氧戊环-2-基)-2-(1H-吡咯-2-基)吡啶(化合物128b)的制备
将化合物128a(250mg)、乙二醇(180mg)和对甲苯磺酸(27.6mg)加入到甲苯(15mL)中,回流分水18h。反应结束后,将反应液用EA稀释,饱和碳酸氢钠水溶液洗涤。有机相用无水硫酸钠干燥,过滤后减压浓缩,得到化合物128b(290mg)。MS m/z(ESI):217.0[M+H] +
第三步:5-(1,3-二氧戊环-2-基)-2-(1-甲基-1H-吡咯-2-基)吡啶(化合物128c)的制备
将化合物128b(250mg)溶解于干燥的DMF(5mL)中,于氮气保护下0℃加入NaH(138.7mg,纯度60%),然后搅拌15min。加入碘甲烷(820mg),并于室温反应5h。反应结束后,将反应液倒入饱和食盐水中,用EA萃取,无水硫酸钠干燥,过滤后减压浓缩,并经硅胶柱层析(PE:EA=5:1)分离纯化,得到化合物128c(125mg)。MS m/z(ESI):231.0[M+H] +
第四步:6-(1-甲基-1H-吡咯-2-基)烟碱醛(化合物128d)的制备
将稀盐酸(2N,2.0mL)滴加到化合物128c(110mg)的THF(4.0mL)溶液中,并于室温反应3h。反应结束后,将反应液浓缩至干。将粗产物溶解于甲醇中,加入过量碳酸钾,并于室温搅拌0.5h,将盐酸盐游离。过滤后减压浓缩,经硅胶柱层析(PE:EA=3:1)分离纯化,得到化合物128d(75mg)。
第五步:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-((6-(1-甲基-1H-吡咯-2-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物128)的制备
将化合物1g(40mg)、128d(25mg)和钛酸四异丙酯(125mg)加入到干燥的THF(5mL)中,并于72℃搅拌10h。然后加入三乙酰氧基硼氢化钠(117mg),并于72℃反应6h。反应结束后,直接经硅胶柱层析(DCM:MeOH=93:7)预纯化,再经Prep-HPLC分离纯化,得到化合物128(34mg)。MS m/z(ESI):532.9[M+H] +
1H NMR(400MHz,CD 3OD)δ9.14(d,J=2.1Hz,1H),8.59-8.43(m,2H),7.78(dd,J=8.2,2.2Hz,1H),7.55(d,J=8.2Hz,1H),6.85(d,J=9.0Hz,1H),6.81-6.65(m,2H),6.50(dd,J=3.7,1.8Hz,1H),6.35(s,1H),6.10(dd,J=3.7,2.7Hz,1H),4.02-3.77(m,7H),3.77-3.59(m,4H),2.73(d,J=6.3Hz,1H),2.41(s,3H),2.32(s,3H),1.71(d,J=8.8Hz,1H).
实施例62:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-((6-(噻唑-4-基)吡啶-3-基)甲基)-3,6-二氮杂双环 [3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物129)
Figure PCTCN2020074696-appb-000121
第一步:4-(5-(1,3-二氧戊环-2-基)吡啶-2-基)噻唑(化合物129b)的制备
将化合物129a(325mg)、121a(200mg)和四三苯基磷钯(50mg)加入到甲苯(10mL)中,并于120℃下反应6h。反应结束后,将反应液浓缩至干,通过硅胶柱层析(PE:EA=2:1)分离纯化,得到化合物129b(115mg)。MS m/z(ESI):235.1[M+H] +
第二步:6-(噻唑-4-基)烟碱醛(化合物129c)的制备
将稀盐酸(2.0mL,3N)滴加到化合物129b(115mg)的THF(3.0mL)溶液中,并于室温反应15h。反应结束后,向反应液中缓慢滴加饱和碳酸氢钠溶液,调节溶液pH至约10,然后用EA萃取。有机相用无水硫酸钠干燥,过滤后浓缩,得到化合物129c(83mg),其不经纯化直接用于下一步反应。MS m/z(ESI):191.1[M+H] +
第三步:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-((6-(噻唑-4-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物129)的制备
将化合物1g(50mg)、129c(26.2mg)和钛酸四异丙酯(156.8mg)加入到干燥的THF(5mL)中,于72℃搅拌反应10h,然后再加入三乙酰氧基硼氢化钠(146.2mg)并于72℃反应6h。反应结束后,直接硅胶柱层析(DCM:MeOH=8:1)粗纯化,再经Prep-HPLC分离纯化,得到化合物129(51mg)。MSm/z(ESI):537.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.98(s,1H),9.66(s,1H),9.22(d,J=2.1Hz,1H),9.13(d,J=2.1Hz,1H),8.58(d,J=1.6Hz,1H),8.44(dd,J=8.9,2.3Hz,1H),8.29(d,J=2.0Hz,1H),8.06(d,J=8.0Hz,1H),7.88(dd,J=8.1,2.2Hz,1H),7.01–6.64(m,2H),6.33(s,1H),3.87–3.49(m,8H),2.61–2.54(m,1H),2.33(s,3H),2.26(s,3H),1.60(d,J=8.4Hz,1H).
实施例63:2-(6-(6-([[2,2'-联吡啶]-5-基甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物130)
Figure PCTCN2020074696-appb-000122
第一步:5-(1,3-二氧戊环-2-基)-2,2'-联吡啶(化合物130b)的制备
将化合物130a(320mg)、121a(200mg)和四三苯基磷钯(50mg)加入到甲苯(10mL)中,在120℃下微波反应4h。反应结束后,将反应液浓缩至干,通过硅胶柱层析(PE:EA=2:1)分离纯化,得到化合物130b(100mg)。MS m/z(ESI):229.1[M+H] +
第二步:[2,2'-联吡啶]-5-甲醛(化合物130c)的制备
将稀盐酸(5.0mL,3N)滴加到化合物130b(100mg)的THF(5.0mL)溶液中,并于室温反应15h。反应结束后,向反应液中缓慢滴加饱和碳酸氢钠溶液,调节溶液pH至约10,然后用EA萃取。有机相用无水硫酸钠干燥,过滤后浓缩,得到化合物130c(45mg),其不经纯化直接用于下一步反应。MS m/z(ESI):185.2[M+H] +
第三步:2-(6-(6-([[2,2'-联吡啶]-5-基甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物130)的制备
将化合物1g(50mg)、130c(25.4mg)和钛酸四异丙酯(156.8mg)加入到干燥的THF(5mL)中,于72℃搅拌反应10h,然后再加入三乙酰氧基硼氢化钠(146.2mg)并于72℃反应6h。反应结束后,直接硅胶柱层析(DCM:MeOH=10:1)粗纯化,再经Prep-HPLC分离纯化,得到化合物130(40mg)。MS m/z(ESI):531.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.97(s,1H),9.65(s,1H),9.13(d,J=2.2Hz,1H),8.75–8.57(m,2H),8.44(dd,J=8.9,2.3Hz,1H),8.35(dd,J=9.7,8.2Hz,2H),7.93(ddd,J=8.3,5.9,1.9Hz,2H),7.44(ddd,J=7.5,4.8,1.1Hz,1H),7.08–6.57(m,2H),6.31(s,1H),3.82–3.57(m,8H),2.63–2.56(m,1H),2.33(s,3H),2.26(s,3H),1.61(d,J=8.4Hz,1H).
实施例64:2-(6-(6-((6-(5-氟-1H-吡咯-2-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物131)
Figure PCTCN2020074696-appb-000123
第一步:6-(5-氟-1H-吡咯-2-基)烟碱醛(化合物131a)的制备
将化合物128a(200mg)和1-氯甲基-4-氟-1,4-二氮杂双环[2.2.2]辛烷二(四氟硼酸)盐(432mg,选择性氟试剂)加入到乙腈(15mL)中,在70℃下微波反应10min。反应结束后,将反应液浓缩至干,通过硅胶柱层析(PE:EA=5:1)分离纯化,得到化合物131a(60mg)。MS m/z(ESI):191.1[M+H] +
第二步:2-(6-(6-((6-(5-氟-1H-吡咯-2-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物131)的制备
将化合物1g(50mg)、131a(26.2mg)和钛酸四异丙酯(156.8mg)加入到干燥的THF(5mL)中,于72℃搅拌反应10h,然后再加入三乙酰氧基硼氢化钠(146.2mg)并于72℃反应6h。反应结束后,直接硅胶柱层析(DCM:MeOH=8:1)粗纯化,再经Prep-HPLC分离纯化,得到化合物131(9mg)。MS m/z(ESI):537.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.06(s,1H),11.98(s,1H),9.65(s,1H),9.12(d,J=2.1Hz,1H),8.43(dd,J=8.9,2.3Hz,1H),8.39(d,J=1.3Hz,1H),7.74–7.65(m,1H),7.57(d,J=8.2Hz,1H),6.92–6.72(m,2H),6.57(t,J=4.2Hz,1H),6.31(s,1H),5.57(t,J=3.8Hz,1H),3.76(d,J=11.2Hz,2H),3.70(d,J=5.7Hz,2H),3.63–3.52(m,4H),2.60-2.55(m,1H),2.33(s,3H),2.26(s,3H),1.59(d,J=8.2Hz,1H).
实施例65:2-(6-(6-((6-(1H-吡唑-5-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物132)
Figure PCTCN2020074696-appb-000124
第一步:6-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-5-基)烟碱醛(化合物132b)的制备
向反应瓶中依次加入化合物132a(2.24g)、8c(1g)、四(三苯基膦)钯(310.6mg)和1,4-二氧六环(20mL),再加入碳酸钠(1.71g)的水(5mL)溶液,并于氮气保护下于95℃搅拌反应5h。反应结束后,加水稀释,并用EA萃取(20mL×3)。合并有机相,依次用水和饱和食盐水洗涤一次,无水硫酸钠干燥,过滤后浓缩,经快速柱层析(DCM:MeOH=96:4)分离纯化,得到化合物132b(1.14g)。
第二步:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-((6-(1-(四氢-2H-吡喃-2-基)-1H-吡唑-5-基)吡啶-3-基)甲 基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物132c)的制备
将化合物1g(500mg)和132b(477.8mg)溶于DMA(10mL)中,并于25℃搅拌反应2h。然后加入三乙酰氧基硼氢化钠(1.17g),并于25℃继续搅拌反应16h。反应结束后,加水淬灭,然后用EA萃取。有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,得到化合物132c(697mg)。MS m/z(ESI):604.3[M+H] +
第三步:2-(6-(6-((6-(1H-吡唑-5-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物132)的制备
将化合物132c(697mg)溶于MeOH(15mL)中,再滴入TFA(0.86mL),并于25℃反应16h。反应结束后,加入饱和碳酸氢钠溶液淬灭反应,将溶液pH调至约9。浓缩除去甲醇,加水稀释,并用二氯甲烷萃取。有机相用无水硫酸钠干燥,过滤后浓缩,经Prep-HPLC分离纯化,得到化合物132(279mg)。MS m/z(ESI):521.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ13.48(s,0.3H,互变异构体1),13.01(s,0.7H,互变异构体2),11.98(s,1H),9.66(s,1H),9.13(d,J=2.2Hz,1H),8.52(s,1H),8.44(dd,J=8.9,2.3Hz,1H),8.01-7.64(m,3H),7.01–6.59(m,3H),6.32(br,1H),3.85-3.68(m,4H),3.68–3.45(m,4H),2.60–2.53(m,1H),2.33(s,3H),2.26(s,3H),1.60(d,J=8.4Hz,1H).
实施例66:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-((6-(5-甲基呋喃-2-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物133)
Figure PCTCN2020074696-appb-000125
第一步:6-(5-甲基呋喃-2-基)烟碱醛(化合物133b)的制备
依次向反应瓶中加入化合物133a(203.1mg)、8c(200mg)、Na 2CO 3(341.9mg)、四(三苯基膦)钯(62.1mg)、水(2.5mL)和1,4-二氧六环(10mL),并于95℃搅拌反应2h。反应结束后,将反应液降至室温,直接经硅胶柱层析(PE:EA=87:13)分离纯化,得到化合物133b(124mg)。MS m/z(ESI):188.1[M+H] +
第二步:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-((6-(5-甲基呋喃-2-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物133)的制备
向反应瓶中依次加入化合物133b(23.2mg)、1g(30mg)和DMA(1mL),并于25℃搅拌反应2h。然后再加入三乙酰氧基硼氢化钠(70.2mg),并于25℃继续搅拌反应16h。反应结束后,反应液经Prep-HPLC分离纯化,得到化合物133(9mg)。MS m/z(ESI):534.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.98(s,1H),9.66(s,1H),9.12(d,J=2.2Hz,1H),8.48(d,J=1.5Hz,1H),8.44(dd,J=8.9,2.3Hz,1H),7.78(dd,J=8.2,2.1Hz,1H),7.60(d,J=8.1Hz,1H),6.96(d,J=4.5Hz,1H),6.84(br,1H),6.78(d,J=9.0Hz,1H),6.31(br,1H),6.25(dd,J=3.2,1.0Hz,1H),3.82-3.68(m,4H),3.68-3.45(m,4H),2.60–2.53(m,1H),2.36(s,3H),2.33(s,3H),2.26(s,3H),1.59(d,J=8.4Hz,1H).
实施例67:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-((6-(噁唑-2-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物134)
Figure PCTCN2020074696-appb-000126
第一步:2-(5-(1,3-二氧戊环-2-基)吡啶-2-基)噁唑(化合物134b)的制备
将化合物121a(300.0mg)和134a(467.0mg)溶于甲苯(15.0mL)中,然后加入四(三苯基膦)钯(75.3mg),并于氮气保护下于120℃搅拌12h。反应液经减压浓缩后用EA(200.0mL)稀释,有机相用水洗涤3次,再用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤后浓缩,经硅胶柱层析(PE:EA=7:3)分离纯化,得到化合物134b(78.0mg)。MS m/z(ESI):219.1[M+1] +
第二步:6-(噁唑-2-基)烟碱醛(化合物134c)的制备
将化合物134b(78.0mg)溶于THF(2.0mL)与水(2.0mL)的混合溶剂中,然后缓慢滴加浓盐酸(1.0mL,12N),并于25℃搅拌8h。反应液用碳酸钾水溶液调节至碱性,并用EA(100.0mL)萃取。有机相水洗3次,再用饱和氯化钠溶液洗,无水硫酸钠干燥,过滤后浓缩,经硅胶柱层析(DCM:MeOH=9:1)分离纯化,得到化合物134c(51.0mg)。MS m/z(ESI):175.1[M+1] +
第三步:6-甲基-N-(5-甲基-1H-吡唑-3-基)-2-(6-(6-((6-(噁唑-2-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)嘧啶-4-胺(化合物134)的制备
将化合物134c(28.8mg)和1g(30.0mg)溶于N,N-二甲基乙酰胺(1.0mL)中,并于25℃下搅拌1h。然后向反应体系中加入三乙酰氧基硼氢化钠(70.2mg),并于25℃下搅拌12h。反应液经EA(20mL)稀释,水洗3次,再用饱和氯化钠溶液洗,经无水硫酸钠干燥,过滤后浓缩,得到粗产物,其经Prep-HPLC分离纯化,得到化合物134(17.0mg)。MS m/z(ESI):521.3[M+1] +
1H NMR(400MHz,DMSO-d 6)δ12.00(s,1H),9.68(s,1H),9.14(d,J=2.4Hz,1H),8.66(s,1H),8.45(dd,J=9.2,2.4Hz,1H),8.29(s,1H),8.06(d,J=8.4Hz,1H),7.95(d,J=8.0Hz,1H),7.45(s,1H),6.79(br,1H),6.78(d,J=8.8Hz,1H),6.32(br,1H),3.79-3.75(m,4H),3.67-3.60(m,4H),2.58-2.56(m,1H),2.34(s,3H),2.26(s,3H),1.61(d,J=8.0Hz,1H).
实施例68:2-(6-(6-((6-(1-异丙基-1H-吡唑-4-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物135)
Figure PCTCN2020074696-appb-000127
第一步:5-(1,3-二氧戊环-2-基)-2-(1H-吡唑-4-基)吡啶(化合物135b)的制备
将化合物121a(1.0g)和135a(1.0g)溶于1,4-二氧六环(30mL)和H 2O(6mL)中,然后依次加入Pd(dppf)Cl 2(130mg)和K 2CO 3(1.5g),并于氮气保护下于95℃搅拌2h。反应结束后,用冰水浴降温,并经硅藻土过滤。将滤液浓缩至干,经MPLC分离纯化,得到化合物135b(452mg)。MS m/z(ESI): 218.2[M+H] +
MPLC条件:
仪器型号:Biotage Isolera Prime 2.3.1;色谱柱:Agela Technologies C18 spherical 20-35um 100A,120g;色谱柱温:25℃;流速:30.0mL/min;检测波长:254nm;洗脱梯度:(0min:20%A,80%B;3.0min:20%A,80%B;25min:90%A,10%B);流动相A:乙腈,流动相B:0.05%TFA水溶液。
第二步:5-(1,3-二氧戊环-2-基)-2-(1-异丙基-1H-吡唑-4-基)吡啶(化合物135d)的制备
将化合物135b(100mg)和Cs 2CO 3(374.9mg)加入到干燥的DMF(10mL)中,然后加入化合物135c(195.6mg),并于80℃搅拌12h。反应结束后,向反应液中加入水(100mL)淬灭反应,并用EA萃取。有机相经饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,经硅胶柱层析(PE:EA=1:1)分离纯化,得到化合物135d(52mg)。MS m/z(ESI):260.0[M+H] +
第三步:6-(1-异丙基-1H-吡唑-4-基)烟碱醛(化合物135e)的制备
将化合物135d(240mg)加入到THF(4mL)和H 2O(2mL)中,然后加入HCl的1,4-二氧六环溶液(4N,1mL),并于25℃搅拌5h。反应结束后,向反应液中加入饱和碳酸氢钠水溶液(50mL)淬灭反应,并用EA萃取(50mL×3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩至干,得到化合物135e(40mg)。MS m/z(ESI):216.1[M+H]。
第四步:2-(6-(6-((6-(1-异丙基-1H-吡唑-4-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物135)的制备
将化合物1g(40mg)、135e(26.1mg)和钛酸四异丙酯(31.4mg)加入到干燥的THF(25mL)中,并于氮气保护下于75℃搅拌10h。然后再向反应体系中加入三乙酰氧基硼氢化钠(23.4mg),并于75℃下继续搅拌6h。反应结束后,将反应液减压浓缩至干,经Prep-HPLC分离纯化,得到化合物135(2.1mg)。MS m/z(ESI):562.1[M+H] +
1H NMR(400MHz,DMSO-d 6)δ11.98(s,1H),9.67(s,1H),9.12(d,J=2.0Hz,1H),8.56-8.39(m,2H),8.32(s,1H),7.97(s,1H),7.71(dd,J=8.0,2.0Hz,1H),7.59(d,J=8.0Hz,1H),7.05-6.65(m,2H),6.33(s,1H),4.64-4.36(m,1H),3.94-3.63(m,4H),3.64-3.45(m,4H),2.62-2.50(m,1H),2.31(s,3H),2.29(s,3H),1.56(d,J=8.0Hz,1H),1.45(d,J=6.4Hz,6H).
实施例69:2-(1-(5-((3-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-基)甲基)吡啶-2-基)-1H-吡唑-5-基)丙-2-醇(化合物136)
Figure PCTCN2020074696-appb-000128
第一步:2-(1-(5-(1,3-二氧戊环-2-基)吡啶-2-基)-1H-吡唑-3-基)丙-2-醇(化合物136a)的制备
将化合物31b(200mg)加入到干燥的THF(10mL)中,并置于干冰乙醇浴中降温15min。然后缓慢滴加甲基溴化镁的乙醚溶液(3N,0.65mL),并保持温度反应15min,然后升至室温继续反应4h。向反应液中加入饱和氯化铵水溶液(1mL)淬灭反应,再加入水(30mL)稀释,并用EA萃取(30mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,得到化合物136a(200mg)。MS m/z(ESI):276.1[M+H] +
第二步:6-(3-(2-羟基丙-2-基)-1H-吡唑-1-基)烟碱醛(化合物136b)的制备
将化合物136a(200mg)加入到THF(5mL)中,然后加入盐酸(2N,4.5mL),并于25℃搅拌12h。向反应液中加入饱和碳酸氢钠水溶液调节pH至7-8,并用EA萃取(30mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,得到化合物136b(165mg)。MS m/z(ESI):232.1[M+H] +
第三步:2-(1-(5-((3-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-基)甲基)吡啶-2-基)-1H-吡唑-5-基)丙-2-醇(化合物136)的制备
将化合物136b(50.5mg)和化合物1g(30mg)加入到DMA(3mL)中,并于氮气保护下于室温搅 拌1h。然后加入三乙酰氧基硼氢化钠(101mg),保持室温反应过夜。反应完毕后,向反应液中加入水(60mL)淬灭反应,并用EA萃取(30mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,经Pre-HPLC分离纯化,得到化合物136(12mg)。MS m/z(ESI):578.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.14(br,1H),9.67(s,1H),9.12(d,J=2.0Hz,1H),8.50-8.41(m,2H),8.41-8.35(m,1H),7.99-7.92(m,1H),7.82(d,J=8.4Hz,1H),6.79(d,J=9.2Hz,2H),6.52(d,J=2.4Hz,1H),6.31(br,1H),5.10(s,1H),3.82-3.70(m,4H),3.68-3.55(m,4H),2.61-2.54(m,1H),2.33(s,3H),2.26(s,3H),1.60(d,J=8.4Hz,1H),1.49(s,6H).
实施例70:(1-(5-((3-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-基)甲基)吡啶-2-基)-1H-吡唑-3-基)甲醇(化合物137)
Figure PCTCN2020074696-appb-000129
第一步:6-(3-(羟甲基)-1H-吡唑-1-基)烟碱醛(化合物137a)的制备
将化合物31c(189mg)加入到THF(5mL)中,然后加入稀盐酸(2N,5mL),并于25℃搅拌2h。向反应液中加入饱和碳酸氢钠水溶液调节pH至7-8,并用EA萃取(30mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,得到化合物137a(157mg)。MS m/z(ESI):204.1[M+H] +
第二步:(1-(5-((3-(5-(4-甲基-6-((5-甲基-1H-吡唑-3-基)氨基)嘧啶-2-基)吡啶-2-基)-3,6-二氮杂双环[3.1.1]庚烷-6-基)甲基)吡啶-2-基)-1H-吡唑-3-基)甲醇(化合物137)的制备
将化合物137a(50.5mg)和化合物1g(50mg)加入到DMA(2mL)中,并于氮气保护下于室温搅拌1h。然后加入三乙酰氧基硼氢化钠(159mg),保持室温反应过夜。反应完毕后,向反应液中加入水(60mL)淬灭反应,并用EA萃取(30mL x 3)。合并有机相,用饱和食盐水洗涤,无水硫酸钠干燥,过滤后浓缩,并经Pre-HPLC分离纯化,得到化合物137(10mg)。MS m/z(ESI):550.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.00(s,1H),9.68(s,1H),9.13(d,J=2.0Hz,1H),8.52(d,J=2.4Hz,1H),8.44(dd,J=8.8,2.4Hz,1H),8.38(d,J=2.4Hz,1H),7.94(dd,J=8.4,2.0Hz,1H),7.82(d,J=8.4Hz,1H),7.06-6.65(m,2H),6.51(d,J=2.4Hz,1H),6.29(br,1H),5.26(t,J=6.0Hz,1H),4.53(d,J=5.6Hz,2H),3.81-3.69(m,4H),3.67-3.49(m,4H),2.59-2.53(m,1H),2.34(s,3H),2.26(s,3H),1.59(d,J=8.4Hz,1H).
实施例71:2-(6-(6-((6-(4-氟-3-甲基-1H-吡唑-1-基)吡啶-3-基)甲基)-3,6-二氮杂双环[3.1.1]庚烷-3-基)吡啶-3-基)-6-甲基-N-(5-甲基-1H-吡唑-3-基)嘧啶-4-胺(化合物138)
Figure PCTCN2020074696-appb-000130
化合物138参考实施例56的合成方法制备。MS m/z(ESI):551.8[M+H] +
分离方法:
实施例1-51、53-58、60-71的化合物的Prep-HPLC纯化均采用Aglient 1260型、Waters 2489型或GeLai 3500型HPLC进行,柱温均为25℃,检测波长为214nm、254nm或280nm,其它分离条件如下表所示:
Figure PCTCN2020074696-appb-000131
Figure PCTCN2020074696-appb-000132
Figure PCTCN2020074696-appb-000133
Figure PCTCN2020074696-appb-000134
Figure PCTCN2020074696-appb-000135
Figure PCTCN2020074696-appb-000136
Figure PCTCN2020074696-appb-000137
Figure PCTCN2020074696-appb-000138
实施例中的以下中间体化合物采用GeLai 3500型HPLC纯化,柱温为25℃,检测波长为214nm、254nm或280nm,其它分离条件如下表所示:
Figure PCTCN2020074696-appb-000139
生物学评价
实验例1:RET抑制实验
实验方法:根据HTRF KinEASE-TK试剂盒(Cisbio)的说明测定本发明化合物对野生型RET酶、突变型RET酶(RET-V804M、RET-V804L和RET-M918T)和融合型RET酶(RET-CCDC6)活性的抑制作用。将不同的RET酶与不同浓度的测试化合物在室温下预孵育30min后,加入底物和三磷酸腺苷(ATP)启动反应。在室温下孵育40min后加入TK抗体-穴状化合物以及抗生蛋白链菌素-XL665,并于室温下孵育45min后进行检测。以溶媒组(DMSO)为阴性对照,缓冲液组(不含RET酶)为空白对照,按照下式计算不同浓度化合物的相对抑制活性百分比(即抑制率):
相对抑制活性百分比=1-(不同浓度化合物组-空白对照)/(阴性对照-空白对照)*100%
将不同浓度的化合物的相对抑制活性百分比相对于化合物浓度作图,按照四参数模型拟合曲线,通过下式计算IC 50值:
y=min+(max-min)/(1+(x/IC 50)^(-Hillslope))
其中y为相对抑制活性百分比,max和min分别为拟合曲线的最大值与最小值,x为化合物的对数浓度,Hillslope为曲线斜率。
实验例2:VEGFR2抑制实验
实验方法:根据HTRF KinEASE-TK试剂盒(Cisbio)的说明测定本发明化合物对VEGFR2酶活性的抑制作用。将VEGFR2酶与不同浓度的测试化合物在室温下预孵育30min后,加入底物和三磷酸腺苷(ATP)启动反应。在室温下孵育40min后加入TK抗体-穴状化合物以及抗生蛋白链菌素-XL665,并于室温下孵育45min后进行检测。以溶媒组(DMSO)为阴性对照,缓冲液组(不含VEGFR2酶)为空白对照,按照下式计算不同浓度化合物的相对抑制活性百分比(即抑制率):
相对抑制活性百分比=1-(不同浓度化合物组-空白对照)/(阴性对照-空白对照)*100%
将不同浓度的化合物的相对抑制活性百分比相对于化合物浓度作图,按照四参数模型拟合曲线,通过下式计算IC 50值:
y=min+(max-min)/(1+(x/IC 50)^(-Hillslope))
其中y为相对抑制活性百分比,max和min分别为拟合曲线的最大值与最小值,x为化合物的对数浓度,Hillslope为曲线斜率。
实验结果:
实验结果如表1-4所示。
表1.本发明化合物在100nM浓度下对突变型RET酶活性的抑制率
化合物编号 对RET-V804M的抑制率 对RET-M918T的抑制率
1 86% 33%
2 93% 51%
3 86% 34%
4 N/A 69%
注:N/A表示未测试。
由表1可以看出,本发明化合物对突变型RET酶具有明显的抑制作用。表2.本发明化合物对RET-V804M酶抑制率
化合物编号 RET-V804M抑制率 化合物编号 RET-V804M抑制率
6(10nM) 83% 84(100nM) 72%
7(10nM) 46% 85(100nM) 77%
8(10nM) 46% 86(10nM) 64%
15(100nM) 91% 88(10nM) 62%
16(100nM) 92% 89(10nM) 70%
18(100nM) 70% 91(10nM) 70%
21(100nM) 49% 96(100nM) 84%
23(10nM) 47% 98(100nM) 63%
26(10nM) 83% 110(100nM) 62%
27(10nM) 87% 111(100nM) 46%
28'(10nM) 34% 117(100nM) 76%
29(10nM) 52% 118(100nM) 74%
30(10nM) 88% 125-2(10nM) 82%
31(10nM) 81% 126(100nM) 82%
41(10nM) 64% 127(100nM) 84%
46(10nM) 92% 128(10nM) 48%
49(100nM) 75% 129(10nM) 84%
50(100nM) 50% 130(10nM) 44%
63(10nM) 65% 131(10nM) 72%
64(10nM) 79% 132(10nM) 84%
67(100nM) 82% 133(10nM) 82%
69(10nM) 56% 134(10nM) 50%
80(10nM) 51% 135(10nM) 65%
82(10nM) 43% 137(10nM) 93%
83(10nM) 56% - -
由表2可以看出,本发明化合物对RET-V804M酶具有明显的抑制作用。表3-1.本发明化合物对RET-WT酶的抑制IC 50(nM)
化合物编号 对RET-WT的抑制IC 50(nM)
17 1.32±0.31
60 2.70±0.69
120 7.33±1.19
122 2.35±0.24
表3-2.本发明化合物对RET-CCDC6酶的抑制IC 50(nM)
化合物编号 对RET-CCDC6的抑制IC 50(nM)
17 2.50±0.55
60 3.99±0.79
69 10.97±9.65
120 18.66±7.27
122 2.91±0.41
表3-3.本发明化合物对RET-V804L酶的抑制IC 50(nM)
化合物编号 对RET-V804L抑制IC 50(nM)
17 6.54±2.43
60 5.07±0.40
120 10.09±1.05
122 4.79±1.68
表3-4.本发明化合物对RET-V804M酶的抑制IC 50(nM)
化合物编号 对V804M抑制IC 50(nM)
9 5.43±0.91
10 3.97±0.30
11 7.22±0.58
12 6.94±0.78
17 1.03±0.47
25 6.29±0.90
28 10.41±1.38
52-2 12.95±2.34
60 3.77±0.52
62 4.53±1.23
70 41.36±3.73
119 14.21±1.27
120 9.12±1.85
121 6.22±1.39
122 3.33±0.75
123 1.88±0.11
124 10.68±0.98
136 2.35±0.13
表3-5.本发明化合物对RET-M918T酶的抑制IC 50(nM)
化合物编号 对RET-M918T酶的抑制IC 50(nM)
17 1.47±0.29
60 1.29±0.27
69 8.60±1.46
120 15.81±3.65
122 4.03±0.66
由表3-1至3-5可以看出,本发明化合物对RET-CCDC6、RET-M918T、RET-V804M、RET-V804L和RET-WT酶均具有明显的抑制作用。
表4.本发明化合物对VEGFR2的抑制率
化合物编号 VEGFR2抑制率 化合物编号 VEGFR2抑制率
1(100nM) 38% 70(100nM) 14%
2(100nM) 29% 80(100nM) -10%
3(100nM) 14% 82(100nM) 15%
4(1000nM) 52% 83(100nM) 18%
6(100nM) -11% 86(100nM) 52%
7(100nM) 16% 88(100nM) 32%
8(300nM) 5% 89(100nM) 16%
9(300nM) 69% 91(100nM) 26%
10(300nM) 69% 96(100nM) 4%
12(300nM) 62% 119(100nM) -7%
15(100nM) 29% 120(100nM) 28%
23(100nM) -10% 121(100nM) 37%
25(300nM) 53% 122(300nM) 67%
26(300nM) 64% 123(30nM) 56%
27(30nM) 23% 124(30nM) 41%
28(300nM) 23% 125(300nM) 44%
29(300nM) 47% 125-2(300nM) 63%
30(30nM) 51% 128(100nM) 20%
31(30nM) 17% 129(30nM) 52%
41(100nM) 22% 130(100nM) 63%
46(300nM) 79% 131(30nM) 50%
52(100nM) -5% 132(30nM) 31%
52-2(300nM) 12% 133(50nM) 28%
60(100nM) 65% 134(100nM) 11%
62(100nM) 62% 135(100nM) 34%
63(100nM) 38% 136(30nM) 18%
64(100nM) 38% 137(30nM) 26%
69(100nM) 2% - -
另外,经测定,化合物11对VEGFR2的抑制IC 50为158.02±25.08nM,并且化合物17对VEGFR2的抑制IC 50为62.97±11.77nM。上述结果结合表4的抑制率数据表明,本发明的化合物对VEGFR2的抑制较弱,与VEGFR2相比,对RET酶具有更好的选择性抑制作用。
实验例3:化合物在大鼠体内药代动力学及组织分布
分别通过灌胃(PO)给予雄性SD大鼠BLU-667(根据WO2017/079140A1实施例5制备)和化合物17,测定大鼠体内BLU-667和化合物17的血浆浓度和脑、肺及甲状腺组织浓度,考察药代动力学特点。PO给药剂量为5mg/kg,溶媒为0.5%MC(甲基纤维素)。PO给药,并在不同时间点(给药前0h,给药后0.25h、0.5h、1h、2h、4h、6h、8h和24h)采集血液样品。血液样品采用乙二胺四乙酸二钾抗凝,离心后得到血浆样品,保存于-80℃。分别于PO给药后0.5h、2h、8h和24h经腹主动脉放血处死大鼠,采集脑、肺和甲状腺,洗净后,按一定比例用生理盐水匀浆,得到组织样品,保存于-80℃。各血浆样品和组织样品经沉淀蛋白处理后进行LC-MS/MS分析。应用WinNonlin 6.3软件,采用非房室模型计算药代动力学参数,结果见表5。
表5.通过PO给药的化合物在大鼠体内血浆及各组织中的药代动力学参数
Figure PCTCN2020074696-appb-000140
表5的数据显示,SD大鼠分别灌胃给予5mg/kg的BLU-667和化合物17后,化合物17在各靶器官组织脑、肺和甲状腺的暴露量均优于BLU-667。
实验例4:化合物在小鼠体内的药效测试
实验目的:评价化合物17和BLU-667在带有人髓样甲状腺癌TT细胞皮下异种移植瘤的Balb/c-nu小鼠模型中的体内药效。
药物配制:化合物17和BLU-667分别用0.1M H 3PO 4水溶液和0.1M HOAc水溶液溶解,配制成澄清溶液(pH为约4.0)。溶媒对照组采用pH为约4.0的H 3PO 4水溶液。给药方式为PO,BID。
肿瘤测量:每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:V=0.5×a×b 2,其中a和b分别表示肿瘤的长径和短径。化合物的抑瘤疗效用肿瘤生长抑制率TGI(%)评价:TGI(%)=[(1-(处理组给药结束时平均瘤体积-处理组开始给药时平均瘤体积))/(溶媒对照组治疗结束时平均瘤体积-溶媒对照组开始治疗时平均瘤体积)]×100%。结果见图1。
统计分析:基于试验结束时相对肿瘤体积,运用Prism Graphpad5.0软件进行统计分析。多组间比较用two-way ANOVA进行分析,p<0.05认为有显著性差异。
实验结果:在人髓样甲状腺癌TT裸鼠移植瘤模型中,化合物17在5mg/kg的给药剂量下即具有显著抗肿瘤作用,并且抗肿瘤作用具有量效依赖的趋势。化合物17在5mg/kg给药剂量下的抗肿瘤效果(T/C=17.44%,TGI=131.36%,p<0.05)优于BLU-667 5mg/kg剂量组(T/C=33.62%,TGI=88.82%,p<0.05)。相对肿瘤增值率T/C(%)=T RTV/C RTV×100%,其中T RTV为受试化合物组平均相对肿瘤体积,C RTV为溶媒对照组相对肿瘤体积;并且相对肿瘤体积RTV=V t/V 0,其中V 0为给药开始时平均肿瘤体积,V t为给药后t天测量的平均肿瘤体积。
上述实施例不以任何方式限定本申请的方案。除本文中描述的那些外,根据前述描述,本发明的多种修改对本领域技术人员而言会是显而易见的。这样的修改也意图落入所附权利要求书的范围内。本申请中所引用的各参考文献(包括所有专利、专利申请、期刊文章、书籍及任何其它公开)均以其整体援引加入本文。

Claims (12)

  1. 化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,其中所述化合物具有式I的结构:
    Figure PCTCN2020074696-appb-100001
    其中:
    环A选自C 6-10芳环和5-6元杂芳环;
    环B选自C 3-8环烷基和4-11元杂环基;
    X 1选自CH和N;
    R 1选自H、卤素、羟基、氰基、C 1-6烷基、C 1-6杂烷基(例如C 1-6烷氧基)、C 3-8环烷基、4-10元杂环基和-NR 20aR 20b,所述烷基、杂烷基(例如烷氧基)、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基和C 1-4杂烷基(例如C 1-4烷氧基);
    R 2选自C 1-6烷基、C 1-6杂烷基、C 3-8环烷基、4-10元杂环基、5-10元杂芳基和-C(=O)R 21,所述烷基、杂烷基、环烷基、杂环基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基和C 3-6环烷基;
    R 3和R 4不存在或者在每次出现时各自独立地选自羟基、卤素、CN、C 1-6烷基、C 1-6杂烷基(例如C 1-6烷氧基)和C 3-6环烷基,所述烷基、杂烷基(例如烷氧基)和环烷基各自任选地被一个或多个选自下列的取代基取代:卤素、CN、C 1-4烷基、C 1-4卤代烷基、C 1-4烷氧基和C 1-4卤代烷氧基;当m大于1时,两个R 3任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基;和/或当n大于1时,两个R 4任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基;
    L选自-O-、-S-、-S(O)-、-S(O) 2-、-N=CR 21-、-N(R 23a)-C(O)-、C 1-6亚烷基、C 1-6亚杂烷基、C 2-6亚烯基、C 2-6亚炔基、
    Figure PCTCN2020074696-appb-100002
    Figure PCTCN2020074696-appb-100003
    Figure PCTCN2020074696-appb-100004
    所述亚烷基、亚杂烷基、亚烯基和亚炔基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6卤代烷氧基、C 1-6杂烷基(例如C 1-6烷氧基)和C 3-8环烷基;或者L为-N(R 23a)-;
    R 5选自羟基、卤素、CN、NO 2、C 1-6烷基、C 1-6杂烷基(例如C 1-6烷氧基)、C 2-6烯基、C 2-6炔基、C 3-8环烷基、C 3-8环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 20aR 20b、-OR 21、-SR 21、 -S(=O)R 22、-S(=O) 2R 22、-S(=O)NR 20aR 20b、-S(=O) 2NR 20aR 20b、-NR 20aS(=O)R 20b、-NR 20aS(=O) 2R 20b、-C(=O)R 21、-C(=O)NR 23aR 23b、-NR 23aC(=O)R 23b、-OC(=O)NR 23aR 23b和-NR 24aC(=O)NR 25aR 25b,所述烷基、杂烷基(例如烷氧基)、烯基、炔基、环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基、C 3-6环烷氧基和4-10元杂环基;
    R 20a、R 20b、R 23a、R 23b、R 23c、R 24a、R 25a和R 25b各自独立地选自H、OH、C 1-6烷基、C 1-6烷氧基和C 3-8环烷基;或者R 20a与R 20b、R 23a与R 23b或R 25a与R 25b连同其所连接的原子一起形成3-8元环烷基或杂环基,所述烷基、烷氧基、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:OH、CN、卤素、NO 2、C 1-4烷基、C 1-4烷氧基、C 1-4羟烷基、C 1-4卤代烷基和C 1-4卤代烷氧基;
    R 30a、R 30b、R 33a、R 33b、R 34a、R 35a和R 35b各自独立地选自H、C 1-6烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基和C 1-6卤代烷氧基;
    R 21、R 22、R 31和R 32各自独立地选自C 1-6烷基、C 1-6烷氧基、C 3-8环烷基、4-10元杂环基、C 6-12芳基和5-10元杂芳基,所述烷基、烷氧基、环烷基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:OH、卤素、CN、C 1-4烷基、C 1-4烷氧基、C 1-4卤代烷基、C 1-4卤代烷氧基、C 3-6环烷基和4-10元杂环基;
    m为0、1、2、3或4,优选为0;
    n为0、1、2、3或4,优选为0、1或2;
    t为0、1、2、3或4,优选为0或1;且
    u为0、1、2、3或4,优选为0或1;
    条件是,当环B为哌嗪环且X 1为CH时,R 2不是4-CF 3-吡啶-2-基或4-CN-吡啶-2-基。
  2. 权利要求1所述的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,其中:
    环A为苯环或5-6元杂芳环;
    优选地,环A为苯环、噻唑环、吡啶环、吡嗪环或嘧啶环;
    更优选地,环A为
    Figure PCTCN2020074696-appb-100005
    其通过*标记的位置与X 1所在环连接,并且通过**标记的位置与环B连接;
    和/或
    环B为C 3-6环烷基或5-7元杂环基;
    优选地,环B为哌啶环、哌嗪环、氮杂环庚烷桥环或二氮杂环庚烷桥环;
    更优选地,环B为
    Figure PCTCN2020074696-appb-100006
    其通过*标记的位置与环A连接,并且通过**标记的位置与L连接;
    和/或
    X 1为CH或N,优选为N。
  3. 权利要求1或2所述的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,其中:
    R 1选自H、卤素、羟基、氰基、C 1-4烷基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元 杂环基,所述烷基、杂烷基(例如烷氧基)、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基和C 1-4杂烷基(例如C 1-4烷氧基);
    优选地,R 1选自C 1-4烷基、5元含氮杂环基和C 1-4杂烷基(例如C 1-4烷氧基),所述烷基、杂环基和杂烷基(例如烷氧基)各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基和C 1-3杂烷基(例如C 1-3烷氧基);
    更优选地,R 1选自C 1-3烷基(例如甲基)、吡咯烷基(例如吡咯烷-1-基)和C 1-3烷氧基(例如乙氧基);
    和/或
    R 2选自C 1-4烷基、C 1-4杂烷基、C 3-6环烷基、4-6元杂环基、5-6元杂芳基和-C(=O)R 21,所述烷基、杂烷基、环烷基、杂环基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基和C 3-6环烷基;
    优选地,R 2选自C 1-3烷基、5-6元杂芳基和-C(=O)CH 3,所述烷基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3杂烷基和C 3-6环烷基;
    更优选地,R 2选自C 1-3烷基(例如甲基)、-C(=O)CH 3、噻吩基、吡咯基、吡唑基、咪唑基、噻唑基、噻二唑基、异噻唑基、噁唑基、噁二唑基、异噁唑基和吡啶基,所述烷基、噻吩基、吡咯基、吡唑基、咪唑基、噻唑基、噻二唑基、异噻唑基、噁唑基、噁二唑基、异噁唑基和吡啶基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、C 1-3烷基(例如甲基)、C 1-3卤代烷基、C 1-3卤代烷氧基、C 1-3杂烷基(例如C 1-3烷氧基)和C 3-6环烷基;
    进一步优选地,R 2为甲基取代的吡唑基(例如5-甲基-1H-吡唑-3-基或1-甲基-1H-吡唑-4-基)、环丙基取代的吡唑基(例如5-环丙基-1H-吡唑-3-基)或-C(O)CH 3
  4. 权利要求1至3中任意一项所述的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,其中:
    R 3和R 4不存在或者在每次出现时独立地选自羟基、卤素、CN、C 1-4烷基和C 1-4烷氧基,所述烷基和烷氧基各自任选地被一个或多个选自下列的取代基取代:卤素、CN、C 1-4烷基、C 1-4卤代烷基、C 1-4烷氧基和C 1-4卤代烷氧基;当m大于1时,两个R 3任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基;和/或当n大于1时,两个R 4任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基;
    优选地,R 3和R 4不存在或者在每次出现时独立地选自羟基、卤素、CN、C 1-3烷基和C 1-3烷氧基,所述烷基和烷氧基各自任选地被一个或多个选自下列的取代基取代:卤素、CN和C 1-3烷基;当m大于1时,两个R 3任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基;和/或当n大于1时,两个R 4任选地与其所连接的原子一起形成C 3-6环烷基或4-10元杂环基;
    更优选地,R 3和R 4不存在或者在每次出现时独立地选自:F、Cl、CN、OH、C 1-3烷基和C 1-3烷氧基;
    进一步优选地,R 3和R 4不存在。
  5. 权利要求1至4中任意一项所述的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,其中:
    L选自-O-、-S-、-C(O)-、-N(R 23a)-C(O)-、-C(O)-N(R 23c)-、C 1-4亚烷基、C 1-4亚杂烷基、
    Figure PCTCN2020074696-appb-100007
    Figure PCTCN2020074696-appb-100008
    所述亚烷基和亚杂烷基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)和C 3-6环烷基;
    优选地,L选自-O-、-C(O)-、-NHC(O)-、-C(O)NH-、C 1-3亚烷基、C 1-3亚杂烷基、
    Figure PCTCN2020074696-appb-100009
    Figure PCTCN2020074696-appb-100010
    所述亚烷基和亚杂烷基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3杂烷基(例如C 1-3烷氧基)和C 3-6环烷基,其中R 23a和R 23b优选为H或C 1-3烷基;
    更优选地,L选自-O-、-C(O)-、-NHC(O)-、-C(O)NH-、C 1-3亚烷基、
    Figure PCTCN2020074696-appb-100011
    Figure PCTCN2020074696-appb-100012
    所述亚烷基任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、C 1-3烷基和C 1-3卤代烷基;
    进一步优选地,L为-CH 2-、-CH(CH 3)-、-O-、-C(O)-、
    Figure PCTCN2020074696-appb-100013
    -C(O)NH-或
    Figure PCTCN2020074696-appb-100014
  6. 权利要求1至5中任意一项所述的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,其中:
    R 5选自羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 20aR 20b、-OR 21、-SR 21、-S(=O)R 22、-S(=O) 2R 22、-S(=O)NR 20aR 20b、-S(=O) 2NR 20aR 20b、-NR 20aS(=O)R 20b、-NR 20aS(=O) 2R 20b、-C(=O)R 21、-C(=O)NR 23aR 23b、-NR 23aC(=O)R 23b、-OC(=O)NR 23aR 23b和-NR 24aC(=O)NR 25aR 25b,所述烷基、杂烷基(例如烷氧基)、烯基、炔基、环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基、C 3-6环烷氧基和4-10元杂环基;
    优选地,R 5选自C 3-6环烷基、4-10元杂环基、C 6-12芳基和5-10元杂芳基,所述环烷基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1- 4烷氧基)、C 3-6环烷基、C 3-6环烷氧基和4-10元杂环基;
    更优选地,R 5选自C 6-10芳基和5-6元杂芳基,所述芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3杂烷基(例如C 1-3烷氧基)、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-C(=O)R 31、-C(=O)NR 33aR 33b和-NR 33aC(=O)R 33b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基、C 3-6环烷氧基和4-6元杂环基;
    进一步优选地,R 5选自苯基和5-6元杂芳基(例如吡啶基、嘧啶基、吡嗪基、哒嗪基、吡唑基、 噁唑基、咪唑基或噻唑基),所述苯基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3杂烷基(例如C 1-3烷氧基)、C 3-6环烷基、C 3-6环烷氧基、4-6元杂环基、5-8元杂芳基(例如吡啶基、吡咯基、吡唑基、呋喃基、噁唑基、咪唑基、噻唑基或环戊基并吡唑基)、-NR 30aR 30b、-OR 31、-C(=O)R 31、-C(=O)NR 33aR 33b和-NR 33aC(=O)R 33b,其中所述环烷基、环烷氧基、杂环基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3杂烷基(例如C 1-3烷氧基)、C 3-6环烷基、C 3-6环烷氧基和4-6元杂环基;
    进一步更优选地,R 5选自苯基、吡啶基、吡唑基和噻唑基,所述苯基、吡啶基、吡唑基和噻唑基各自任选地被一个或多个选自下列的取代基取代:卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3烷氧基、C 3-6环烷基、C 3-6环烷氧基、4-6元杂环基、5-8元杂芳基(例如吡啶基、吡咯基、吡唑基、呋喃基、噁唑基、咪唑基、噻唑基或环戊基并吡唑基)、-NR 30aR 30b和-OR 31,其中所述杂环基和杂芳基各自任选地被一个或多个选自下列的取代基取代:卤素、CN、C 1-3烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3卤代烷氧基、C 1-3烷氧基、C 3-6环烷基、C 3-6环烷氧基和4-6元杂环基;
    最优选地,R 5为任选地被一个或多个选自卤素(例如氟或氯)、CN、C 1-3烷基(例如甲基或乙基)、C 1-3卤代烷基(例如三氟甲基)、C 1-3烷氧基(例如甲氧基或乙氧基)、C 3-6环烷基(例如环丙基)、C 3-6环烷氧基(例如环丙氧基)和5-6元杂芳基(例如吡啶基、吡咯基、吡唑基、呋喃基、噁唑基、咪唑基或噻唑基)的取代基取代的苯基、吡啶基、吡唑基或噻唑基,其中所述5-6元杂芳基任选地进一步被一个或多个选自卤素(例如氟或氯)、C 1-3烷基(例如甲基、乙基或异丙基)、C 1-3卤代烷基(例如氟甲基)、C 1- 3羟烷基(例如羟甲基或羟丙基)、C 1-3烷氧基(例如甲氧基)、C 3-6环烷基(例如环丙基)和C 3-6环烷氧基(例如环丙氧基或环丁氧基)的取代基取代。
  7. 权利要求1至6中任意一项所述的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,其中:
    R 20a、R 20b、R 23a、R 23b、R 23c、R 24a、R 25a和R 25b各自独立地选自H、C 1-4烷基、C 1-4烷氧基和C 3- 8环烷基;或者R 20a与R 20b、R 23a与R 23b或R 25a与R 25b连同其所连接的原子一起形成3-8元环烷基或杂环基,所述烷基、烷氧基、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:OH、CN、卤素、NO 2、C 1-4烷基、C 1-4烷氧基、C 1-4羟烷基、C 1-4卤代烷基和C 1-4卤代烷氧基;
    优选地,R 20a、R 20b、R 23a、R 23b、R 23c、R 24a、R 25a和R 25b各自独立地为H、C 1-4烷基或C 1-4烷氧基;
    特别地,R 23a和R 23b各自独立地选自H、C 1-3烷基、C 1-3烷氧基和C 3-6环烷基;或者R 23a与R 23b连同其所连接的C原子一起形成C 3-6环烷基或杂环基,所述烷基、烷氧基、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:卤素、C 1-3烷基、C 1-3烷氧基、C 1-3羟烷基、C 1-3卤代烷基和C 1-3卤代烷氧基;
    和/或
    R 21、R 22、R 31和R 32各自独立地选自C 1-4烷基、C 1-4烷氧基、C 3-8环烷基和4-10元杂环基,所述烷基、烷氧基、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:OH、卤素、CN、C 1-4烷基、C 1-4烷氧基、C 1-4卤代烷基、C 1-4卤代烷氧基、C 3-6环烷基和4-10元杂环基;
    优选地,R 21、R 22、R 31和R 32各自独立地选自C 1-4烷基;
    和/或
    R 30a、R 30b、R 33a、R 33b、R 34a、R 35a和R 35b各自独立地选自H、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4烷氧基和C 1-4卤代烷氧基;
    优选地,R 30a、R 30b、R 33a、R 33b、R 34a、R 35a和R 35b各自独立地选自H和C 1-4烷基。
  8. 权利要求1所述的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药,
    其中所述化合物具有式I-A至式I-G之一所示的结构:
    Figure PCTCN2020074696-appb-100015
    其中:
    R 5选自C 6-12芳基和5-10元杂芳基,其中(1)所述C 6-12芳基任选地被一个或多个选自下列的取代基取代:C 3-6环烷氧基、C 6-12芳基、5-10元杂芳基、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷氧基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基,且(2)所述5-10元杂芳基任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基C 3-6环烷氧基和4-10元杂环基;且
    R 1、R 2、R 23a、R 30a、R 30b、R 31、R 32、R 33a、R 33b、R 34a、R 35a和R 35b如权利要求1-7中任意一项所定义,且R 23a优选为H或C 1-3烷基;
    Figure PCTCN2020074696-appb-100016
    其中:
    R 1、R 2、R 5和R 23a如权利要求1-7中任一项所定义,且R 23a优选为H或C 1-3烷基;
    Figure PCTCN2020074696-appb-100017
    其中:
    当X 1为CH时,R 1、R 2、R 5和R 23a如权利要求1-7中任意一项所定义,且R 23a优选为H或C 1- 3烷基;而当X 1为N时,R 1、R 2、R 5和R 23a如上文式I-A所定义;
    Figure PCTCN2020074696-appb-100018
    其中:
    R 1、R 2、R 23a、R 23b和t如权利要求1-7中任意一项所定义;
    当X 1为CH时,R 5如权利要求1-7中任意一项所定义;且
    当X 1为N时,R 5为C 6-12芳基或5-10元杂芳基,其中
    (i)当t为0时,所述C 6-12芳基和5-10元杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基,
    (ii)当t为1时,(1)所述C 6-12芳基任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基,且(2)所述5-10元杂芳基任选地被一个或多个选自下列的取代基取代:NO 2、C 2-6烯基、C 2-6炔基、C 3-6环烷氧基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基;且
    R 30a、R 30b、R 31、R 32、R 33a、R 33b、R 34a、R 35a和R 35b如权利要求1-7中任意一项所定义;
    Figure PCTCN2020074696-appb-100019
    其中:
    R 1、R 2、R 5、R 23a、R 23b、X 1和t如上文式I-D所定义;
    Figure PCTCN2020074696-appb-100020
    其中:
    R 1、R 2、R 5、R 23a、R 23b和X 1如上文式I-D所定义;
    R 4如权利要求1-7中任意一项所定义,且优选为C 1-3烷基或C 1-3烷氧基;
    R 23c为H、C 1-3烷基或C 1-3烷氧基,所述烷基和烷氧基各自任选地被一个或多个选自下列的取代基取代:OH、CN、卤素、C 1-4烷氧基和C 1-4羟烷基;
    u为0或1;且
    n为0或1;
    Figure PCTCN2020074696-appb-100021
    其中:
    X 1为CH或N;
    R 1、R 2和R 4如权利要求1-7中任意一项所定义,且R 4优选为C 1-3烷基或C 1-3烷氧基;
    n为0或1;
    R 5选自C 6-12芳基和5-10元杂芳基,其中(1)所述C 6-12芳基任选地被一个或多个选自下列的取代基取代:C 3-6环烷氧基、C 6-12芳基、5-10元杂芳基、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷氧基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基;且(2)所述5-10元杂芳基任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 2-6烯基、C 2-6炔基、C 3-6环烷基、C 3-6环烷氧基、4-10元杂环基、C 6-12芳基、5-10元杂芳基、-NR 30aR 30b、-OR 31、-SR 31、-S(=O)R 32、-S(=O) 2R 32、-S(=O)NR 30aR 30b、-S(=O) 2NR 30aR 30b、-NR 30aS(=O)R 30b、-NR 30aS(=O) 2R 30b、-C(=O)R 31、-C(=O)NR 33aR 33b、-NR 33aC(=O)R 33b、-OC(=O)NR 33aR 33b和-NR 34aC(=O)NR 35aR 35b,其中所述环烷基、环烷氧基、杂环基、芳基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、NO 2、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基(例如C 1-4烷氧基)、C 3-6环烷基和4-10元杂环基;且
    R 30a、R 30b、R 31、R 32、R 33a、R 33b、R 34a、R 35a和R 35b如权利要求1-7中任意一项所定义。
  9. 权利要求1所述的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳 定同位素衍生物、代谢物或前药,
    其中所述化合物选自:
    Figure PCTCN2020074696-appb-100022
    Figure PCTCN2020074696-appb-100023
    Figure PCTCN2020074696-appb-100024
    Figure PCTCN2020074696-appb-100025
  10. 制备权利要求8的化合物的方法,所述方法包括以下步骤:
    Figure PCTCN2020074696-appb-100026
    其中:
    Hal 1和Hal 2各自独立地为F、Cl、Br或I;优选地,Hal 1为F、Cl、Br或I,且Hal 2为Cl、Br或I;
    R 1选自H、氰基、C 1-6烷基、C 1-6杂烷基(例如C 1-6烷氧基)、C 3-8环烷基、4-6元杂环基和-NR 20aR 20b,所述烷基、杂烷基(例如烷氧基)、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:卤素、CN、C 1-4烷基、C 1-4卤代烷基、C 1-4卤代烷氧基和C 1-4杂烷基(例如C 1-4烷氧基);
    R 2选自C 1-6烷基、C 1-6杂烷基、C 3-8环烷基、4-6元杂环基和5-6元杂芳基,所述烷基、杂烷基、环烷基、杂环基和杂芳基各自任选地被一个或多个选自下列的取代基取代:羟基、卤素、CN、C 1-4烷基、C 1-4卤代烷基、C 1-4羟烷基、C 1-4卤代烷氧基、C 1-4杂烷基和C 3-6环烷基;
    R 23a选自H、C 1-6烷基、C 1-6烷氧基和C 3-8环烷基,所述烷基、烷氧基和环烷基各自任选地被一个或多个选自下列的取代基取代:OH、CN、卤素、C 1-4烷基、C 1-4烷氧基、C 1-4羟烷基、C 1-4卤代烷基和C 1-4卤代烷氧基;且
    R 20a、R 20b和R 5如权利要求8式I-A所定义;
    第一步:化合物I-A-1与R 2-NH 2在碱存在下反应生成化合物I-A-2;
    第二步:化合物I-A-3与I-A-4在碱存在下反应生成化合物I-A-5;
    第三步:化合物I-A-5与含硼试剂反应生成化合物I-A-6;
    第四步:化合物I-A-2与I-A-6反应生成化合物I-A-7;
    第五步:化合物I-A-7在酸性条件下脱除保护基生成化合物I-A-8;
    第六步:化合物I-A-8与I-A-9反应生成化合物I-A;
    或者,
    所述方法包括以下步骤:
    Figure PCTCN2020074696-appb-100027
    其中:
    各基团如上文路线A所定义;
    第一步:化合物I-A-5在酸性条件下脱除保护基生成化合物I-A-10;
    第二步:化合物I-A-10与I-A-9反应生成化合物I-A-11;
    第三步:化合物I-A-11与含硼试剂反应生成化合物I-A-12;
    第四步:化合物I-A-12与I-A-2反应生成化合物I-A;
    或者,
    所述方法包括以下步骤:
    Figure PCTCN2020074696-appb-100028
    其中:
    各基团如上文路线A所定义;
    第一步:化合物I-B-1与R 2-NH 2在碱存在下反应生成化合物I-B-2;
    第二步:化合物I-B-2与I-A-12反应生成化合物I-B;
    或者,
    所述方法包括以下步骤:
    Figure PCTCN2020074696-appb-100029
    其中:
    各基团如上文路线A所定义;且X 1选自CH和N;
    第一步:化合物I-C-1与R 2-NH 2在碱存在下反应生成化合物I-C-2;
    第二步:化合物I-C-3与含硼试剂反应生成化合物I-C-4;
    第三步:化合物I-C-2与I-C-4反应生成化合物I-C-5;
    第四步:化合物I-C-5在酸性条件下脱除保护基生成化合物I-C-6;
    第五步:化合物I-C-6与I-A-9反应生成化合物I-C;
    或者,
    所述方法包括以下步骤:
    Figure PCTCN2020074696-appb-100030
    其中:
    R 1和R 2如路线A所定义;
    R 5如权利要求8式I-D所定义;
    R 23a和R 23b各自独立地选自H、C 1-6烷基、C 1-6烷氧基和C 3-8环烷基;或者R 23a与R 23b连同其所连接的C原子一起形成3-8元环烷基或杂环基,所述烷基、烷氧基、环烷基和杂环基各自任选地被一个或多个选自下列的取代基取代:CN、卤素、C 1-4烷基、C 1-4烷氧基、C 1-4羟烷基、C 1-4卤代烷基和C 1-4卤代烷氧基;
    X 1选自CH和N;且
    t为0或1;
    或者,
    所述方法包括以下步骤:
    Figure PCTCN2020074696-appb-100031
    其中:
    R 1、R 2、R 5、R 23a、R 23b和t如路线E所定义;
    X 1选自CH和N;且
    Hal 2为F、Cl、Br或I;优选地,Hal 2为Cl、Br或I;
    第一步:化合物I-C-2与I-A-6反应生成化合物I-E-1;
    第二步:化合物I-E-1在酸性条件下脱除保护基生成化合物I-E-2;
    第三步:化合物I-E-2与I-D-1经缩合反应生成化合物I-E;
    或者,
    所述方法包括以下步骤:
    Figure PCTCN2020074696-appb-100032
    Figure PCTCN2020074696-appb-100033
    其中:
    R 1、R 2、R 23a、R 23b和R 5如路线E所定义;
    X 1选自CH和N;
    R 4不存在或者选自羟基、CN、C 1-6烷基、C 1-6卤代烷基和C 1-6杂烷基(例如C 1-6烷氧基);
    R 23c为H、C 1-3烷基或C 1-3烷氧基,所述烷基和烷氧基各自任选地被一个或多个选自下列的取代基取代:OH、CN、卤素、C 1-4烷氧基和C 1-4羟烷基;
    Hal 2为F、Cl、Br或I;优选地,Hal 2为Cl、Br或I;
    u为0或1;且
    n为0或1;
    第一步:化合物I-C-2与I-F-1反应生成化合物I-F-2;
    第二步:化合物I-F-3与胺反应生成化合物I-F-4;
    第三步:化合物I-F-4在酸性条件下脱除保护基生成化合物I-F-5;
    第四步:化合物I-F-2与I-F-5在碱存在下反应生成化合物I-F;
    或者,
    所述方法包括以下步骤:
    Figure PCTCN2020074696-appb-100034
    其中:
    R 1和R 2如路线A所定义;
    X 1选自CH和N;
    R 4选自H、C 1-6烷基、C 1-6卤代烷基和C 1-6杂烷基;
    R 5如上文式I-G所定义;且
    n为0或1;
    第一步:化合物I-G-1与R 5-OH反应生成化合物I-G-2;
    第二步:化合物I-G-2在酸性条件下脱除保护基生成化合物I-G-3;
    第三步:化合物I-F-2与I-G-3在碱存在下经亲核取代反应生成化合物I-G。
  11. 药物组合物,其包含预防或治疗有效量的权利要求1-9中任意一项所述的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药;
    任选地,所述药物组合物还包含一种或多种药学上可接受的载体。
  12. 权利要求1-9中任意一项所述的化合物,所述化合物的立体异构体、互变异构体或其混合物,所述化合物的N-氧化物,所述化合物的药学上可接受的盐、共晶、多晶型物或溶剂合物,或者所述化合物的稳定同位素衍生物、代谢物或前药、或权利要求11所述的药物组合物在制备用于预防或治疗与RET活性相关的疾病或病况的药物中的用途;
    优选地,所述与RET活性相关的疾病或病况为癌症或肿瘤,或肠易激综合征;
    所述癌症或肿瘤进一步优选为肺癌(例如非小细胞肺癌)、乳腺癌、头颈癌、直肠癌、肝癌、淋巴瘤、甲状腺癌(例如甲状腺髓样癌或乳头状甲状腺癌)、结肠癌、多发性骨髓瘤、黑色素瘤、胶质瘤、脑瘤或肉瘤。
PCT/CN2020/074696 2019-02-19 2020-02-11 杂环化合物、包含其的药物组合物及其制备方法和用途 WO2020168939A1 (zh)

Priority Applications (14)

Application Number Priority Date Filing Date Title
BR112021016299-2A BR112021016299A2 (pt) 2019-02-19 2020-02-11 Composto heterocíclico, composição farmacêutica compreendendo o mesmo, método de preparação do mesmo e uso do mesmo
CN202080009954.2A CN113316578B (zh) 2019-02-19 2020-02-11 杂环化合物、包含其的药物组合物及其制备方法和用途
JP2021571055A JP2022521859A (ja) 2019-02-19 2020-02-11 複素環化合物、これを含む薬物組成物、その製造方法および使用
AU2020226422A AU2020226422A1 (en) 2019-02-19 2020-02-11 Heterocyclic compound, pharmaceutical composition comprising same, preparation method therefor, and use thereof
CA3130245A CA3130245A1 (en) 2019-02-19 2020-02-11 Heterocyclic compound, pharmaceutical composition comprising same, preparation method therefor, and use thereof
MX2021009971A MX2021009971A (es) 2019-02-19 2020-02-11 Compuesto heterociclico, composicion farmaceutica que lo comprende, metodo para su preparacion, y uso de estos.
CN202311264080.7A CN117327078A (zh) 2019-02-19 2020-02-11 杂环化合物、包含其的药物组合物及其制备方法和用途
SG11202107848TA SG11202107848TA (en) 2019-02-19 2020-02-11 Heterocyclic compound, pharmaceutical composition comprising same, preparation method therefor, and use thereof
US17/431,930 US20220144847A1 (en) 2019-02-19 2020-02-11 Heterocyclic compound, pharmaceutical composition comprising same, preparation method therefor, and use thereof
EP20760123.8A EP3929198A4 (en) 2019-02-19 2020-02-11 HETEROCYCLIC COMPOUND, PHARMACEUTICAL COMPOSITION COMPRISING IT, METHOD FOR PREPARING IT AND ITS USE
CN202311221283.8A CN117263945A (zh) 2019-02-19 2020-02-11 杂环化合物、包含其的药物组合物及其制备方法和用途
KR1020217022806A KR20210131314A (ko) 2019-02-19 2020-02-11 헤테로시클릴 화합물, 이를 포함하는 약제학적 조성물, 이의 제조 방법, 및 이의 용도
IL285378A IL285378A (en) 2019-02-19 2021-08-04 A heterocyclic compound, a medical preparation containing it, a method for its preparation, and its use
CONC2021/0011023A CO2021011023A2 (es) 2019-02-19 2021-08-20 Compuesto heterocíclico, composición farmacéutica que lo comprende, método para su preparación, y uso de estos

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN201910124584 2019-02-19
CN201910124584.6 2019-02-19
CN201910437878 2019-05-24
CN201910437878.4 2019-05-24
CN201910932095.3 2019-09-29
CN201910932095 2019-09-29

Publications (1)

Publication Number Publication Date
WO2020168939A1 true WO2020168939A1 (zh) 2020-08-27

Family

ID=72143544

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/074696 WO2020168939A1 (zh) 2019-02-19 2020-02-11 杂环化合物、包含其的药物组合物及其制备方法和用途

Country Status (14)

Country Link
US (1) US20220144847A1 (zh)
EP (1) EP3929198A4 (zh)
JP (1) JP2022521859A (zh)
KR (1) KR20210131314A (zh)
CN (3) CN117327078A (zh)
AU (1) AU2020226422A1 (zh)
BR (1) BR112021016299A2 (zh)
CA (1) CA3130245A1 (zh)
CL (1) CL2021002205A1 (zh)
CO (1) CO2021011023A2 (zh)
IL (1) IL285378A (zh)
MX (1) MX2021009971A (zh)
SG (1) SG11202107848TA (zh)
WO (1) WO2020168939A1 (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022022398A1 (zh) * 2020-07-28 2022-02-03 四川科伦博泰生物医药股份有限公司 一种嘧啶类化合物的盐和晶型及其制备方法
CN114235976A (zh) * 2021-11-09 2022-03-25 暨南大学 一种含氮杂环有机化合物中间产物的合成和分析方法
WO2022199503A1 (zh) 2021-03-24 2022-09-29 四川科伦博泰生物医药股份有限公司 杂环化合物治疗与激酶耐药突变相关的疾病的用途和方法

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101166734A (zh) * 2005-02-28 2008-04-23 日本烟草产业株式会社 具有Syk抑制活性的新型的氨基吡啶化合物
WO2012154518A1 (en) * 2011-05-10 2012-11-15 Merck Sharp & Dohme Corp. Bipyridylaminopyridines as syk inhibitors
CN103298788A (zh) * 2010-10-28 2013-09-11 日本新药株式会社 吡啶衍生物及医药
WO2017079140A1 (en) 2015-11-02 2017-05-11 Blueprint Medicines Corporation Inhibitors of ret

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010208945A (ja) * 2007-06-01 2010-09-24 Mitsubishi Tanabe Pharma Corp 複素環化合物
JP2013107824A (ja) * 2010-03-17 2013-06-06 Dainippon Sumitomo Pharma Co Ltd 新規単環ピリミジン誘導体

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101166734A (zh) * 2005-02-28 2008-04-23 日本烟草产业株式会社 具有Syk抑制活性的新型的氨基吡啶化合物
CN103298788A (zh) * 2010-10-28 2013-09-11 日本新药株式会社 吡啶衍生物及医药
WO2012154518A1 (en) * 2011-05-10 2012-11-15 Merck Sharp & Dohme Corp. Bipyridylaminopyridines as syk inhibitors
WO2017079140A1 (en) 2015-11-02 2017-05-11 Blueprint Medicines Corporation Inhibitors of ret

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
DATABASE REGISTRY 2 October 2006 (2006-10-02), retrieved from STN Database accession no. 909343-99-3 *
DATABASE REGISTRY 2 October 2006 (2006-10-02), retrieved from STN Database accession no. 909344-03-2 *
DATABASE REGISTRY 2 October 2006 (2006-10-02), retrieved from STN Database accession no. 909344-04-3 *
DATABASE REGISTRY 2 October 2006 (2006-10-02), retrieved from STN Database accession no. 909344-08-7 *
DATABASE REGISTRY 22 May 2012 (2012-05-22), retrieved from STN Database accession no. 1374201-58-7 *
DATABASE REGISTRY 5 December 2011 (2011-12-05), retrieved from STN Database accession no. 1349069-14-2 *
DATABASE REGISTRY 5 December 2011 (2011-12-05), retrieved from STN Database accession no. 1349252-44-3 *
G. W. H. CHEESEMANE. S. G. WERSTIUK: "Pro-drugs as Novel Delivery Systems", vol. 14, 1985, ACS SYMPOSIUM SERIES, pages: 390 - 392
LI, BINGKE ET AL.: "In Silico Prediction of Spleen Tyrosine Kinase Inhibitors Using Machine Learning Approaches and an Optimized Molecular Descriptor Subset Generated by Recursive Feature Elimination Method", COMPUTERS IN BIOLOGY AND MEDICINE, vol. 43, no. 4, 1 May 2013 (2013-05-01), XP055143456, ISSN: 0010-4825, DOI: 10.1016/j.compbiomed.2013.01.015 *
NATURE REVIEWS CANCER, vol. 14, no. 3, 2014, pages 173 - 86
STAHLWERMUTH: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY-VCH
T. W. GREENEP. G. M. WUTS: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022022398A1 (zh) * 2020-07-28 2022-02-03 四川科伦博泰生物医药股份有限公司 一种嘧啶类化合物的盐和晶型及其制备方法
WO2022199503A1 (zh) 2021-03-24 2022-09-29 四川科伦博泰生物医药股份有限公司 杂环化合物治疗与激酶耐药突变相关的疾病的用途和方法
CN114235976A (zh) * 2021-11-09 2022-03-25 暨南大学 一种含氮杂环有机化合物中间产物的合成和分析方法
CN114235976B (zh) * 2021-11-09 2023-11-03 暨南大学 一种含氮杂环有机化合物中间产物的合成和分析方法

Also Published As

Publication number Publication date
EP3929198A4 (en) 2022-11-16
CA3130245A1 (en) 2020-08-27
CO2021011023A2 (es) 2021-09-09
AU2020226422A1 (en) 2021-09-09
CN117327078A (zh) 2024-01-02
IL285378A (en) 2021-09-30
JP2022521859A (ja) 2022-04-12
CN117263945A (zh) 2023-12-22
BR112021016299A2 (pt) 2021-10-13
SG11202107848TA (en) 2021-08-30
EP3929198A1 (en) 2021-12-29
US20220144847A1 (en) 2022-05-12
KR20210131314A (ko) 2021-11-02
CL2021002205A1 (es) 2022-04-22
CN113316578B (zh) 2023-10-31
MX2021009971A (es) 2021-10-13
CN113316578A (zh) 2021-08-27

Similar Documents

Publication Publication Date Title
JP6190883B2 (ja) 1,4−二置換ピリダジン類似体およびsmn欠損に関連する状態を処置するための方法
WO2021043077A1 (zh) 一种取代吡嗪化合物、其制备方法和用途
WO2020168939A1 (zh) 杂环化合物、包含其的药物组合物及其制备方法和用途
JP2017518276A (ja) ブルトン型チロシンキナーゼ(btk)インヒビターとしての多フルオロ置換化合物
JP2020525525A (ja) Rho−関連プロテインキナーゼ阻害剤、rho−関連プロテインキナーゼ阻害剤を含む医薬組成物、当該医薬組成物の調製方法及び使用
CN114867720A (zh) 杂芳基类衍生物及其制备方法和用途
AU2017359023A1 (en) 3-substituted propionic acids as alpha v integrin inhibitors
CN114846005A (zh) Shp2抑制剂及其应用
CN113950481A (zh) 大环化合物
CN114423753A (zh) 作为cd38抑制剂的杂双环酰胺
TW202130631A (zh) 3—(5—甲氧基—1—側氧基異吲哚啉—2—基)哌啶—2,6—二酮衍生物及其用途
WO2022028506A1 (zh) Sos1抑制剂、包含其的药物组合物及其用途
WO2020182018A1 (zh) 氮杂环化合物、其制备方法及用途
WO2022237178A1 (zh) 双环杂芳基类衍生物及其制备方法和用途
TW202214233A (zh) 可做為tlr9抑制劑之經取代雜芳基化合物
CN111484479A (zh) 氮杂环化合物、包含其的药物组合物及其制备方法和用途
WO2022199503A1 (zh) 杂环化合物治疗与激酶耐药突变相关的疾病的用途和方法
CN112574201B (zh) 芳基胺类化合物、包含其的药物组合物及其制备方法和用途
WO2022188792A1 (zh) 具有蛋白激酶抑制活性的杂环化合物、包含其的药物组合物及其制备方法和用途
WO2024022186A1 (zh) 甲基吡唑化合物、包含其的药物组合物及其制备方法和用途
WO2022089219A1 (zh) 芳基酰胺化合物、包含其的药物组合物及其制备方法和用途
WO2022166796A1 (zh) 嘧啶或吡啶并杂环类腺苷受体抑制剂及其制备方法和用途
WO2023165581A1 (zh) 一种吡啶类衍生物及其用途
WO2024032589A1 (zh) TGF-β抑制剂化合物及其用途
TW202104208A (zh) 吡唑基-氨基-嘧啶基衍生物的苯醚和苯胺及其組合物和方法

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: 20760123

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3130245

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021571055

Country of ref document: JP

Kind code of ref document: A

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: 112021016299

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2020226422

Country of ref document: AU

Date of ref document: 20200211

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2021124417

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 112021016299

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20210817

ENP Entry into the national phase

Ref document number: 2020760123

Country of ref document: EP

Effective date: 20210920

WWE Wipo information: entry into national phase

Ref document number: 521430084

Country of ref document: SA