WO2019201131A1 - 用于抑制蛋白激酶活性的二(杂)芳基大环化合物 - Google Patents

用于抑制蛋白激酶活性的二(杂)芳基大环化合物 Download PDF

Info

Publication number
WO2019201131A1
WO2019201131A1 PCT/CN2019/082051 CN2019082051W WO2019201131A1 WO 2019201131 A1 WO2019201131 A1 WO 2019201131A1 CN 2019082051 W CN2019082051 W CN 2019082051W WO 2019201131 A1 WO2019201131 A1 WO 2019201131A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkylene
group
cycloalkyl
alkyl
membered heterocyclic
Prior art date
Application number
PCT/CN2019/082051
Other languages
English (en)
French (fr)
Inventor
王义汉
赵九洋
艾义新
Original Assignee
深圳市塔吉瑞生物医药有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 深圳市塔吉瑞生物医药有限公司 filed Critical 深圳市塔吉瑞生物医药有限公司
Priority to EP19787700.4A priority Critical patent/EP3770161B1/en
Priority to JP2020556951A priority patent/JP7092405B2/ja
Priority to US17/047,877 priority patent/US11358973B2/en
Publication of WO2019201131A1 publication Critical patent/WO2019201131A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged 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
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61K31/529Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C215/00Compounds containing amino and hydroxy groups bound to the same carbon skeleton
    • C07C215/46Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton
    • C07C215/48Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton with amino groups linked to the six-membered aromatic ring, or to the condensed ring system containing that ring, by carbon chains not further substituted by hydroxy groups
    • C07C215/50Compounds containing amino and hydroxy groups bound to the same carbon skeleton having hydroxy groups bound to carbon atoms of at least one six-membered aromatic ring and amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings of the same carbon skeleton with amino groups linked to the six-membered aromatic ring, or to the condensed ring system containing that ring, by carbon chains not further substituted by hydroxy groups with amino groups and the six-membered aromatic ring, or the condensed ring system containing that ring, bound to the same carbon atom of the carbon chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/18Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms to carbon atoms of hydrocarbon radicals substituted by doubly-bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C313/00Sulfinic acids; Sulfenic acids; Halides, esters or anhydrides thereof; Amides of sulfinic or sulfenic acids, i.e. compounds having singly-bound oxygen atoms of sulfinic or sulfenic groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C313/02Sulfinic acids; Derivatives thereof
    • C07C313/06Sulfinamides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/08Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon radicals, substituted by hetero atoms, attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/18Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D207/20Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • 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
    • C07D471/22Heterocyclic 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 systems contains four or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D487/12Heterocyclic 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 three hetero rings
    • C07D487/18Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/22Heterocyclic 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 four or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/22Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains four or more hetero rings

Definitions

  • the present invention relates to the field of medical technology, and in particular to specific di(hetero)aryl macrocycles, pharmaceutical compositions comprising the same, and methods for their preparation and use for the treatment of cancer, pain, neurological diseases, autoimmune diseases and inflammation .
  • Protein kinases are key regulators of cell growth, proliferation and survival. Genetic and epigenetic changes accumulate in cancer cells, leading to aberrant activation of signal transduction pathways that drive malignant processes. Inhibition of these signaling pathways represents a promising intervention opportunity for targeted cancer therapies.
  • ALK belongs to the insulin receptor (IR) superfamily of receptor tyrosine kinases. ALK is an attractive molecular target for cancer therapeutic intervention due to its important role in hematopoiesis, solid and stromal tumors.
  • IR insulin receptor
  • TRK The promyosin-related receptor tyrosine kinase
  • NT neurotrophic protein
  • ROS1 kinase is a receptor tyrosine kinase with an unknown ligand.
  • ROS1 kinase has been reported to undergo gene rearrangement to produce constitutively active fusion proteins in many human cancers, including glioblastoma, non-small cell lung cancer (NSCLC), cholangiocarcinoma, ovarian cancer, gastric adenocarcinoma, colorectal Cancer, inflammatory myofibroblastic tumor, angiosarcoma, and epithelioid hemangioendothelioma.
  • NSCLC non-small cell lung cancer
  • cholangiocarcinoma cholangiocarcinoma
  • ovarian cancer gastric adenocarcinoma
  • colorectal Cancer colorectal Cancer
  • inflammatory myofibroblastic tumor angiosarcoma
  • epithelioid hemangioendothelioma epithelioid hemangioendot
  • the di(hetero)aryl macrocyclic compound of the present invention can effectively bind to the ATP binding site of ALK, ROS1, and TRK kinase, and has an inhibitory effect on these proteins. More critically, it binds to mutants of these proteins, such as ALK G1202R, ALK L1196M, ROS1 G2032R or TRKA G595R.
  • the compounds of the invention are inhibitors of wild and mutant ALK, ROSl, TRK, etc. and will be useful in the treatment of subjects with abnormal signaling in one or more of ALK, ROSl or TRK.
  • the present invention provides a novel di(hetero)aryl macrocyclic compound and a composition comprising the same and use thereof for better inhibition of wild and mutant kinases such as ALK, ROS1 and TRK.
  • the invention provides a compound of formula (I):
  • a 1 is selected from CR 1 or N;
  • a 2 is selected from CR 2 or N;
  • a 3 is selected from CR 3 or N;
  • a 4 is selected from CR 4 or N;
  • R 1 , R 2 , R 3 and R 4 are independently selected from the group consisting of H, D, halogen, -CN, -NO 2 , -OR a , -SR a , -NR b R c , -C(O)R a , -C(O)OR a , -C(O)NR b R c , -OC(O)R a , -NR b C(O)R a , -S(O)R a , -S(O) 2 R a , C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, 3 to 7 membered heterocyclic, C 6-10 aryl or 5 to 10 membered heteroaryl;
  • the above groups are optionally substituted by one or more D until completely deuterated;
  • each R a , R b and R c are independently selected from the group consisting of H, D, C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, 3 to 7 membered heterocyclic, C 6 a -10 aryl or a 5 to 10 membered heteroaryl; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • L 1 is selected from C(R 1a )(R 2a ), O, S, N(R 1a ), C(O), S(O) or S(O) 2 ;
  • L 2 is selected from C(R 1b )(R 2b ), O, S, N(R 1b ), C(O), S(O) or S(O) 2 ;
  • X is selected from O, S, N(R 1c ) or C(R 1c )(R 2c );
  • Y is selected from O, S, N(R 1d ) or C(R 1d )(R 2d );
  • W is selected from O, S, N(R 1e ) or C(R 1e )(R 2e );
  • R is selected from H, D, C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, 3 to 7 membered heterocyclic, C 6-10 aryl or 5 to 10 membered heteroaryl a group; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • n is selected from 1, 2, 3, 4 or 5;
  • n is selected from 1, 2 or 3;
  • R 1a and R 2a is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0-6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 Alkyl-C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 An alkyl-5 to 10 membered heteroary
  • R 1b and R 2b is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0-6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 Alkyl-C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 An alkyl-5 to 10 membered heteroary
  • R 1c and R 2c are each independently selected from H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membered heteroaryl group
  • R 1d and R 2d are each independently selected from H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membered heteroaryl group
  • R 1e and R 2e are each independently selected from H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membered heteroaryl group
  • Substituents present on different atoms in -X-(L 1 ) m -Y- may be bonded to form a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, a C 6-14 aryl group or a 3 to 10 membered hetero An aryl group; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • the substituents present on different atoms in -(L 2 ) n -W- may be bonded to form a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, a C 6-14 aryl group or a 3 to 10 membered heteroaryl group. Wherein the above group is optionally substituted by one or more D until completely deuterated;
  • the invention provides a compound of the invention, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, polymorph thereof, A pharmaceutical composition of a prodrug or active metabolite and a pharmaceutically acceptable excipient.
  • a compound of the invention is provided in the pharmaceutical composition in an effective amount.
  • the compounds of the invention are provided in a therapeutically effective amount.
  • the compounds of the invention are provided in a prophylactically effective amount.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable excipient, further comprising other therapeutic agents.
  • the invention provides a compound comprising a compound of the invention, or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, polymorph thereof, , a prodrug or active metabolite, optionally other therapeutic agents, and a pharmaceutically acceptable carrier, adjuvant or vehicle kit.
  • the present invention provides a process for the preparation of a pharmaceutical composition as described above, comprising the steps of: mapping a pharmaceutically acceptable excipient with a compound of the present invention or a pharmaceutically acceptable salt thereof, Isomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or active metabolites are combined to form a pharmaceutical composition.
  • the invention provides a method of treating cancer, pain, neurological disease, autoimmune disease, and inflammation in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of the invention or a pharmaceutical thereof Acceptable salts, enantiomers, diastereomers, racemates, solvates, hydrates, polymorphs, prodrugs or active metabolites or pharmaceutical compositions of the invention.
  • the compound is administered orally, subcutaneously, intravenously or intramuscularly.
  • the compound is administered chronically.
  • the invention in another aspect, relates to a method of inhibiting a protein or tyrosine kinase (including one or more of ALK, ROS1, TRK) comprising: causing one or more of said kinases with at least one effective amount a compound of formula (I) or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, polymorph, prodrug or active metabolism thereof And/or in contact with at least one pharmaceutical composition of the invention, wherein the contacting is in vitro, ex vivo or in vivo.
  • a protein or tyrosine kinase including one or more of ALK, ROS1, TRK
  • C 1-6 alkyl includes C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1-6 , C 1-5 , C 1-4 , C 1-3 , C 1 -2 , C 2-6 , C 2-5 , C 2-4 , C 2-3 , C 3-6 , C 3-5 , C 3-4 , C 4-6 , C 4-5 and C 5 -6 alkyl.
  • C 1-6 alkyl refers to a straight or branched saturated hydrocarbon group having from 1 to 6 carbon atoms, also referred to herein as “lower alkyl.” In some embodiments, a C 1-4 alkyl group is particularly preferred.
  • alkyl group examples include, but are not limited to, methyl (C 1 ), ethyl (C 2 ), n-propyl (C 3 ), isopropyl (C 3 ), n-butyl (C 4 ), uncle Butyl (C 4 ), sec-butyl (C 4 ), isobutyl (C 4 ), n-pentyl (C 5 ), 3-pentyl (C 5 ), pentyl (C 5 ), neopentyl (C 5 ), 3-methyl-2-butyl (C 5 ), tert-amyl (C 5 ) and n-hexyl (C 6 ).
  • each of the alkyl groups is independently optionally substituted, for example, 1 to 5 substituents, 1 to 3 substituents or 1 substituent, and the appropriate substituents are as follows definition.
  • C 2-6 alkenyl means a straight or branched hydrocarbon group having 2 to 6 carbon atoms and one or more carbon-carbon double bonds (eg, 1, 2 or 3 carbon-carbon double bonds) .
  • One or more carbon-carbon double bonds may be internal (eg, in 2-butenyl) or end (eg, in 1-butenyl).
  • C2-4 alkenyl is particularly preferred.
  • alkenyl group examples include, but are not limited to, vinyl (C 2 ), 1-propenyl (C 3 ), 2-propenyl (C 3 ), 1-butenyl (C 4 ), 2-butene A group (C 4 ), a butadienyl group (C 4 ), a pentenyl group (C 5 ), a pentadienyl group (C 5 ), a hexenyl group (C 6 ), and the like.
  • each of the alkenyl groups is independently optionally substituted, for example, 1 to 5 substituents, 1 to 3 substituents or 1 substituent, and the appropriate substituents are as follows definition.
  • C 2-6 alkynyl means having from 2 to 6 carbon atoms, one or more carbon-carbon triple bonds (eg, 1, 2 or 3 carbon-carbon triple bonds) and optionally one or more carbons a linear or branched hydrocarbon group of a carbon double bond (for example, 1, 2 or 3 carbon-carbon double bonds).
  • a C2-4 alkynyl group is particularly preferred.
  • an alkynyl group does not contain any double bonds.
  • the one or more carbon oxime bonds may be internal (eg, in 2-butynyl) or end (eg, in 1-butynyl).
  • alkynyl group examples include, but are not limited to, ethynyl (C 2 ), 1-propynyl (C 3 ), 2-propynyl (C 3 ), 1-butynyl (C 4 ), 2- Butynyl (C 4 ), pentynyl (C 5 ), hexynyl (C 6 ), and the like.
  • each of the alkynyl groups is independently optionally substituted, for example, 1 to 5 substituents, 1 to 3 substituents or 1 substituent, and the appropriate substituents are as follows definition.
  • C 1-6 alkylene group means a divalent group formed by removing another hydrogen of a C 1-6 alkyl group, and may be a substituted or unsubstituted alkylene group. In some embodiments, C 1-4 alkylene is particularly preferred.
  • the unsubstituted alkylene group includes, but is not limited to, methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), propylene (-CH 2 CH 2 CH 2 -), arylene (-CH 2 CH 2 CH 2 CH 2 -), pentylene (-CH 2 CH 2 CH 2 CH 2 CH 2 -), hexylene (-CH 2 CH 2 CH 2 CH 2 CH 2 CH 2 -) ,and many more.
  • alkylene groups substituted with one or more alkyl groups (methyl groups) include, but are not limited to, substituted methylene groups (-CH(CH 3 )- , -C(CH 3 ) 2 -), substituted ethylene (-CH(CH 3 )CH 2 -, -CH 2 CH(CH 3 )-, -C(CH 3 ) 2 CH 2 -, -CH 2 C(CH 3 ) 2- ), substituted propylene (-CH(CH 3 )CH 2 CH 2 -, -CH 2 CH(CH 3 )CH 2 -, -CH 2 CH 2 CH(CH 3 ) -, -C(CH 3 ) 2 CH 2 CH 2 -, -CH 2 C(CH 3 ) 2 CH 2 -, -CH 2 CH 2 C(CH 3 ) 2 -), and the like.
  • substituted methylene groups -CH(CH 3 )- , -C(CH 3 ) 2 -
  • substituted ethylene -CH(CH
  • C 0-6 alkylene includes a chemical bond and a C 1-6 alkylene group as defined above.
  • C 1-6 alkoxy refers to the group -OR wherein R is a substituted or unsubstituted C 1-6 alkyl group. In some embodiments, a C 1-4 alkoxy group is particularly preferred. Specific alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentyloxy, N-Hexyloxy and 1,2-dimethylbutoxy.
  • Halo or halogen refers to fluorine (F), chlorine (Cl), bromine (Br), and iodine (I).
  • the halo group is F, Cl or Br.
  • the halogen group is F or Cl.
  • the halogen group is F.
  • C 1-6 haloalkyl and “C 1-6 haloalkoxy” mean the above-mentioned “C 1-6 alkyl” and “C 1-6 alkoxy” which are bonded to one or more halogen groups. Replaced by the regiment.
  • a C 1-4 haloalkyl group is particularly preferred, more preferably a C 1-2 haloalkyl group.
  • a C 1-4 haloalkoxy group is particularly preferred, more preferably a C 1-2 haloalkoxy group.
  • Exemplary haloalkyl groups include, but the are not limited to: -CF 3, -CH 2 F, -CHF 2, -CHFCH 2 F, -CH 2 CHF 2, -CF 2 CF 3, -CCl 3, -CH 2 Cl , -CHCl 2 , 2,2,2-trifluoro-1,1-dimethyl-ethyl, and the like.
  • Exemplary haloalkoxy groups include, but are not limited to, -OCH 2 F, -OCHF 2 , -OCF 3 , and the like.
  • C 3-10 cycloalkyl refers to a non-aromatic cyclic hydrocarbon group having from 3 to 10 ring carbon atoms and zero heteroatoms. In some embodiments, a C 3-7 cycloalkyl group is preferred, and a C 3-6 cycloalkyl group is particularly preferred, more preferably a C 5-6 cycloalkyl group.
  • the cycloalkyl group also includes a ring system in which the above cycloalkyl ring is fused to one or more aryl or heteroaryl groups, wherein the point of attachment is on the cycloalkyl ring, and in such a case, the number of carbons continues to be represented The number of carbons in the cycloalkyl system.
  • Exemplary cycloalkyl groups include, but are not limited to, cyclopropyl (C 3 ), cyclopropenyl (C 3 ), cyclobutyl (C 4 ), cyclobutenyl (C 4 ), cyclopentyl ( C 5 ), cyclopentenyl (C 5 ), cyclohexyl (C 6 ), cyclohexenyl (C 6 ), cyclohexadienyl (C 6 ), cycloheptyl (C 7 ), cycloheptene (C 7 ), cycloheptadienyl (C 7 ), cycloheptatrienyl (C 7 ), cyclooctyl (C 8 ), cyclooctenyl (C 8 ), bicyclo [2.2.1] Heptyl (C 7 ), bicyclo [2.2.2] octyl (C 8 ), cyclodecyl (C 9 ), cyclode
  • each of the cycloalkyl groups is independently optionally substituted, for example, 1 to 5 substituents, 1 to 3 substituents or 1 substituent, and appropriate substitution.
  • the basis is defined as follows.
  • heterocyclic group or a group of a 3 to 10 membered non-aromatic ring system having a ring carbon atom and 1 to 4 ring hetero atoms, wherein each hetero atom is independently selected from nitrogen and oxygen. , sulfur, boron, phosphorus and silicon.
  • the point of attachment may be a carbon or nitrogen atom as long as the valence permits.
  • a 3- to 7-membered heterocyclic group is preferred, which is a 3- to 7-membered non-aromatic ring system having a ring carbon atom and 1 to 3 ring heteroatoms; in some embodiments, 3 to 6 A heterocyclic group is particularly preferred, which is a 3- to 6-membered non-aromatic ring system having a ring carbon atom and 1 to 3 ring hetero atoms; more preferably a 5- to 6-membered heterocyclic group having a ring carbon atom and A 5- to 6-membered non-aromatic ring system of 1 to 3 ring heteroatoms.
  • Heterocyclyl also includes ring systems wherein the above heterocyclyl ring is fused to one or more cycloalkyl, aryl or heteroaryl groups, wherein the point of attachment is on the heterocyclyl ring; and in such cases, the ring The number of members continues to indicate the number of ring members in the heterocyclyl ring system. Regardless of whether or not the heterocyclic group is previously modified with a "substituted" group, each of the heterocyclic groups is independently optionally substituted, for example, 1 to 5 substituents, 1 to 3 substituents or 1 substituent, and appropriate substitution. The basis is defined as follows.
  • Exemplary 3-membered heterocyclic groups containing one hetero atom include, but are not limited to, aziridine, oxacyclopropane, thiorenyl.
  • Exemplary 4-membered heterocyclic groups containing one hetero atom include, but are not limited to, azetidinyl, oxetanyl and thietane.
  • Exemplary 5-membered heterocyclic groups containing one hetero atom include, but are not limited to, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothiophenyl, dihydrothienyl, pyrrolidinyl, dihydropyrrolyl and pyrrolyl-2, 5-dione.
  • Exemplary 5-membered heterocyclic groups containing two heteroatoms include, but are not limited to, dioxalanyl, oxasulfuranyl, disulfuranyl, and Azolidin-2-one.
  • Exemplary 5-membered heterocyclic groups containing three heteroatoms include, but are not limited to, triazolinyl, oxadiazolyl, and thiadiazolyl.
  • Exemplary 6-membered heterocyclic groups containing one hetero atom include, but are not limited to, piperidinyl, tetrahydropyranyl, dihydropyridyl, and thianyl.
  • Exemplary 6-membered heterocyclic groups containing two heteroatoms include, but are not limited to, piperazinyl, morpholinyl, dithianyl, dioxoalkyl.
  • Exemplary 6-membered heterocyclic groups containing three heteroatoms include, but are not limited to, triazinanyl.
  • Exemplary 7-membered heterocyclic groups containing one hetero atom include, but are not limited to, azepanyl, oxaheptyl, and thiaheptanyl.
  • Exemplary 8-membered heterocyclic groups containing one hetero atom include, but are not limited to, azacyclooctyl, oxacyclooctyl, and thicyclooctyl.
  • Exemplary 5-membered heterocyclic groups (also referred to herein as 5,6-bicyclic heterocyclyl) fused to a C6 aryl ring include, but are not limited to, indanyl, isoindoline , dihydrobenzofuranyl, dihydrobenzothiophenyl, benzoxazolinone, and the like.
  • Exemplary 6-membered heterocyclic groups fused to a C 6 aryl ring include, but are not limited to, tetrahydroquinolyl, tetrahydroisoquinolinyl, and many more.
  • C 6-14 aryl refers to a monocyclic or polycyclic (eg, bicyclic or tricyclic) 4n+2 aromatic ring system having 6 to 14 ring carbon atoms and zero heteroatoms (eg, having A group of 6, 10 or 14 ⁇ electrons shared in a ring.
  • an aryl group having six ring carbon atoms ( “C 6 aryl”; e.g., phenyl).
  • an aryl group has ten ring carbon atoms (" C10 aryl”; for example, naphthyl, eg, 1-naphthyl and 2-naphthyl).
  • an aryl group has fourteen ring carbon atoms (" C14 aryl"; for example, fluorenyl).
  • C14 aryl for example, fluorenyl
  • a C 6-10 aryl group is particularly preferred, more preferably a C 6 aryl group.
  • the aryl group also includes a ring system in which the above aryl ring is fused to one or more cycloalkyl or heterocyclic groups, and the point of attachment is on the aryl ring, in which case the number of carbon atoms continues to be represented. The number of carbon atoms in the aryl ring system.
  • each of the aryl groups is independently optionally substituted, for example, 1 to 5 substituents, 1 to 3 substituents or 1 substituent, and the appropriate substituents are as follows definition.
  • 5 to 10 membered heteroaryl means a 5-10 membered monocyclic or bicyclic 4n+2 aromatic ring system having a ring carbon atom and 1 to 4 ring heteroatoms (for example, having a ring-like arrangement) a group of 6 or 10 ⁇ electrons, wherein each heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur.
  • the point of attachment may be a carbon or nitrogen atom as long as the valence permits.
  • Heteroaryl bicyclic systems may include one or more heteroatoms in one or both rings.
  • Heteroaryl also includes ring systems wherein the above heteroaryl ring is fused to one or more cycloalkyl or heterocyclic groups, and the point of attachment is on the heteroaryl ring, in this case a carbon atom The number continues to indicate the number of carbon atoms in the heteroaryl ring system.
  • a 5 to 6 membered heteroaryl group is particularly preferred, which is a 5-6 membered monocyclic or bicyclic 4n+2 aromatic ring system having a ring carbon atom and 1-4 ring heteroatoms.
  • each of the heteroaryl groups is independently optionally substituted, for example, 1 to 5 substituents, 1 to 3 substituents or 1 substituent, suitably substituted
  • the basis is defined as follows.
  • Exemplary 5-membered heteroaryl groups containing one hetero atom include, but are not limited to, pyrrolyl, furyl and thienyl.
  • Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl.
  • Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to, triazolyl, oxadiazolyl, and thiadiazolyl.
  • Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to, tetrazolyl.
  • Exemplary 6-membered heteroaryl groups containing one hetero atom include, but are not limited to, pyridyl.
  • Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to, pyridazinyl, pyrimidinyl, and pyrazinyl.
  • Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to, triazinyl and tetrazinyl, respectively.
  • Exemplary 7-membered heteroaryl groups containing one hetero atom include, but are not limited to, azepandinyl, oxepanethylene, and thiephenylene.
  • Exemplary 5,6-bicyclic heteroaryl groups include, but are not limited to, mercapto, isodecyl, oxazolyl, benzotriazolyl, benzothienyl, isobenzothienyl, benzofuranyl , benzoisofuranyl, benzimidazolyl, benzoxazolyl, benzoisoxazolyl, benzooxadiazolyl, benzothiazolyl, benzisothiazolyl, benzothiadiazolyl, Pyridazinyl and fluorenyl.
  • Exemplary 6,6-bicyclic heteroaryl groups include, but are not limited to, naphthyridinyl, acridinyl, quinolyl, isoquinolinyl, fluorenyl, quinoxalinyl, pyridazinyl and quinazolinyl .
  • Each of R aa is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two R aa groups are bonded to form a heterocyclyl or a heteroaryl ring wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl group is independently 0, 1, 2, 3, 4 or 5 R dd groups Substitute
  • Each of R cc is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two R cc groups are bonded to form a heterocycle a hetero or heteroaryl ring wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl group is independently 0, 1, 2, 3, 4 or 5 R Substituting dd group;
  • Each of R ee is independently selected from the group consisting of alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, aryl, heterocyclyl, and heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbon The cyclo, heterocyclyl, aryl and heteroaryl are independently substituted by 0, 1, 2, 3, 4 or 5 R gg groups;
  • Each of R ff is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl, or two R ff groups are bonded to form a heterocyclic group Or a heteroaryl ring wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl group is independently 0, 1, 2, 3, 4 or 5 R gg Group substitution
  • Deuterated or “D” means that one or more hydrogens in the compound or group are replaced by deuterium; deuteration may be monosubstituted, disubstituted, polysubstituted or fully substituted.
  • deuteration may be monosubstituted, disubstituted, polysubstituted or fully substituted.
  • deuterated are used interchangeably with “one or more deuterated”.
  • Non-deuterated compound means a compound containing a proportion of germanium atoms not higher than the natural helium isotope content (0.015%).
  • the strontium isotope content of the cerium in the deuterated position is at least 0.015%, preferably more than 30%, more preferably more than 50%, more preferably more than 75%, more preferably more than 95%, more preferably more than 95%, more preferably more than 95%, more preferably More than 99%.
  • pharmaceutically acceptable salt means that, within the scope of sound medical judgment, it is suitable for contact with tissues of humans and lower animals without excessive toxicity, irritation, allergies, etc., and with reasonable benefits/dangers. Those salts that are proportionate.
  • Pharmaceutically acceptable salts are well known in the art. For example, Berge et al., pharmaceutically acceptable salts as described in detail in J. Pharmaceutical Sciences (1977) 66: 1-19.
  • Pharmaceutically acceptable salts of the compounds of the invention include those derived from suitable inorganic and organic acids and inorganic and organic bases.
  • non-toxic acid addition salts examples include salts with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid, or salts with organic acids such as acetic acid, oxalic acid, Maleic acid, tartaric acid, citric acid, succinic acid or malonic acid. Also included are salts formed using conventional methods in the art, for example, ion exchange methods.
  • adipic acid salts alginate, ascorbate, aspartate, besylate, benzoate, disulfate, borate, butyrate, camphor Acid salt, camphor sulfonate, citrate, cyclopentanoate, digluconate, lauryl sulfate, ethanesulfonate, formate, fumarate, gluconate, glycerol Phosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate , malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate Salt, pectin
  • Pharmaceutically acceptable salts derived from suitable bases include the alkali metal, alkaline earth metal, ammonium and N + (C 1-4 alkyl) 4 salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium salts, and the like.
  • other pharmaceutically acceptable salts include non-toxic ammonium salts, quaternary ammonium salts and amine cations formed with counterions, counterions such as halides, hydroxides, carboxylates, sulfates, phosphates, Nitrate, lower alkyl sulfonate and aryl sulfonate.
  • Active metabolite refers to a pharmacologically active product of the metabolism of a compound of formula (I) or a salt thereof in vivo. Prodrugs and active metabolites of the compounds can be determined using conventional techniques known or available in the art.
  • Subjects for administration include, but are not limited to, humans (ie, males or females of any age group, eg, pediatric subjects (eg, infants, children, adolescents) or adult subjects (eg, young Adults, middle-aged adults or older adults) and/or non-human animals, for example, mammals, for example, primates (eg, cynomolgus monkeys, rhesus monkeys), cattle, pigs, horses, sheep , goats, rodents, cats and/or dogs.
  • the subject is a human.
  • the subject is a non-human animal.
  • the terms "person,” “patient,” and “subject” are used interchangeably herein.
  • treatment includes the effect of a subject having a particular disease, disorder, or condition that reduces the severity of the disease, disorder, or condition, or delays or slows the disease, disorder. Or the development of a condition ("therapeutic treatment"), but also the effect that occurs before the subject begins to have a particular disease, disorder or condition (“prophylactic treatment”).
  • Combination and related terms mean the simultaneous or sequential administration of a therapeutic agent of the invention.
  • a compound of the invention may be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms, or together with another therapeutic agent in a single unit dosage form.
  • a compound of the invention refers to a compound of formula (I) - a compound of formula (VI) and a compound of formula (I') - a compound of formula (V') (including a subset of the formulae, eg, formula (III) -1) a compound), or a pharmaceutically acceptable salt, enantiomer, diastereomer, racemate, solvate, hydrate, polymorph, prodrug or active metabolite thereof .
  • the invention relates to a compound of formula (I):
  • a 1 is selected from CR 1 or N;
  • a 2 is selected from CR 2 or N;
  • a 3 is selected from CR 3 or N;
  • a 4 is selected from CR 4 or N;
  • R 1 , R 2 , R 3 and R 4 are independently selected from the group consisting of H, D, halogen, -CN, -NO 2 , -OR a , -SR a , -NR b R c , -C(O)R a , -C(O)OR a , -C(O)NR b R c , -OC(O)R a , -NR b C(O)R a , -S(O)R a , -S(O) 2 R a , C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, 3 to 7 membered heterocyclic, C 6-10 aryl or 5 to 10 membered heteroaryl;
  • the above groups are optionally substituted by one or more D until completely deuterated;
  • each R a , R b and R c are independently selected from the group consisting of H, D, C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, 3 to 7 membered heterocyclic, C 6 a -10 aryl or a 5 to 10 membered heteroaryl; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • L 1 is selected from C(R 1a )(R 2a ), O, S, N(R 1a ), C(O), S(O) or S(O) 2 ;
  • L 2 is selected from C(R 1b )(R 2b ), O, S, N(R 1b ), C(O), S(O) or S(O) 2 ;
  • X is selected from O, S, N(R 1c ) or C(R 1c )(R 2c );
  • Y is selected from O, S, N(R 1d ) or C(R 1d )(R 2d );
  • W is selected from O, S, N(R 1e ) or C(R 1e )(R 2e );
  • R is selected from H, D, C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, 3 to 7 membered heterocyclic, C 6-10 aryl or 5 to 10 membered heteroaryl a group; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • n is selected from 1, 2, 3, 4 or 5;
  • n is selected from 1, 2 or 3;
  • R 1a and R 2a is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1b and R 2b is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1c and R 2c is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1d and R 2d is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1e and R 2e is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • Substituents present on different atoms in -X-(L 1 ) m -Y- may be bonded to form a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, a C 6-14 aryl group or a 3 to 10 membered hetero An aryl group; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • the substituents present on different atoms in -(L 2 ) n -W- may be bonded to form a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, a C 6-14 aryl group or a 3 to 10 membered heteroaryl group. Wherein the above group is optionally substituted by one or more D until completely deuterated;
  • the invention relates to the above compound, which is of formula (II):
  • a 1 is selected from CR 1 or N;
  • a 4 is selected from CR 4 or N;
  • R 1 , R 3 and R 4 are independently selected from H, D, halogen, -CN, -NO 2 , -OR a , -SR a , -NR b R c , -C(O)R a , -C (O)OR a , -C(O)NR b R c , -OC(O)R a , -NR b C(O)R a , -S(O)R a , -S(O) 2 R a a C 1-6 alkyl group, a C 1-6 haloalkyl group, a C 3-7 cycloalkyl group, a 3 to 7 membered heterocyclic group, a C 6-10 aryl group or a 5 to 10 membered heteroaryl group; wherein the above group Optionally substituted by one or more D until completely deuterated;
  • each R a , R b and R c are independently selected from the group consisting of H, D, C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, 3 to 7 membered heterocyclic, C 6 a -10 aryl or a 5 to 10 membered heteroaryl; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • X is selected from O or C(R 1c )(R 2c );
  • Y is selected from N(R 1d ) or C(R 1d )(R 2d );
  • W is selected from O or NH
  • n is selected from 1, 2, 3, 4 or 5;
  • R 1a and R 2a is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1b and R 2b is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1c and R 2c is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1d and R 2d is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • m -Y- may be bonded to form a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, and a C 6-14 aryl group. Or a 3 to 10 membered heteroaryl; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • the invention relates to the above compound, which is of formula (III-1) or (III-2):
  • a 1 is selected from CR 1 or N;
  • a 4 is selected from CR 4 or N;
  • R 1 , R 3 and R 4 are independently selected from the group consisting of H, D, halogen, -CN, -NO 2 , -OR a , -SR a , -NR b R c , C 1-6 alkyl, C 1- 6 haloalkyl, C 3-7 cycloalkyl, 3 to 7 membered heterocyclyl, C 6-10 aryl or 5 to 10 membered heteroaryl; wherein said group is optionally substituted by one or more D, Until complete era;
  • each R a , R b and R c are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; wherein the above groups are optionally substituted by one or more D until completely deuterated generation;
  • X is selected from O or C(R 1c )(R 2c );
  • Y is selected from N(R 1d ) or C(R 1d )(R 2d );
  • W is selected from O or NH
  • n is selected from 1, 2 or 3;
  • R 1a and R 2a is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1b is selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 Alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0-6 alkylene- C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene-C 3-6 Cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene-5 to 10 a heteroaryl group; wherein the above group is optional
  • R 1c and R 2c is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1d and R 2d is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • m -Y- may be bonded to form a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, and a C 6-14 aryl group. Or a 3 to 10 membered heteroaryl; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • the invention relates to the above compound, which is of formula (IV-1) or (IV-2):
  • R 3 is selected from the group consisting of H, D, halogen, -CN, -NO 2 , -OR a , -SR a or -NR b R c ;
  • each R a , R b and R c are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; wherein the above groups are optionally substituted by one or more D until completely deuterated generation;
  • X is selected from O or C(R 1c )(R 2c );
  • Y is selected from NH, CH 2 or C(Me)(Me); wherein the above groups are optionally substituted by one or more D until completely deuterated;
  • n is selected from 1, 2 or 3;
  • R 1a and R 2a is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1b is selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 Alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0-6 alkylene- C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene-C 3-6 Cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene-5 to 10 a heteroaryl group; wherein the above group is optional
  • each of R 1c and R 2c is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a ,- C 0-6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 Alkylene-C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 An alkylene-5 to 10-membered
  • R 3 is selected from the group consisting of H, D, halogen, -CN or -NO 2 ;
  • X is O
  • Y is selected from NH, CH 2 or C(Me)(Me); wherein the above groups are optionally substituted by one or more D until completely deuterated;
  • n is selected from 1, 2 or 3;
  • R 1a and R 2a are, independently, selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; wherein the above groups are optionally substituted by one or more D until fully deuterated;
  • R 1b is selected from C 1-6 alkyl or C 1-6 haloalkyl; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • R 3 is selected from the group consisting of H, D, halogen, -CN or -NO 2 ;
  • X is O
  • Y is selected from CH 2 or C(Me)(Me); wherein the above groups are optionally substituted by one or more D until completely deuterated;
  • n is selected from 1, 2 or 3;
  • R 1a and R 2a are, independently, selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; wherein the above groups are optionally substituted by one or more D until fully deuterated;
  • R 1b is selected from C 1-6 alkyl or C 1-6 haloalkyl; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • the invention relates to the above compound, which is of formula (V-1) or (V-2):
  • R 3 is selected from the group consisting of H, D, halogen, -CN or -NO 2 ;
  • Y is selected from NH, CH 2 , CHD or CD 2 ;
  • n is selected from 1, 2 or 3;
  • R 1a and R 2a is independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; wherein the above groups are optionally substituted with one or more D until fully deuterated;
  • R 1b is selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; wherein the above groups are optionally substituted by one or more D until completely deuterated;
  • the invention relates to the above compound, which is of formula (VI-1) or (VI-2):
  • R 3 is selected from the group consisting of H, D, halogen, -CN or -NO 2 ;
  • R 1a and R 2a are, independently, selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; wherein the above groups are optionally substituted by one or more D until fully deuterated;
  • R 1b is selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; wherein the above groups are optionally substituted by one or more D until completely deuterated;
  • the invention relates to the above compound, which is a compound of formula (I'):
  • a 1 is selected from CR 1 or N;
  • a 2 is selected from CR 2 or N;
  • a 3 is selected from CR 3 or N;
  • a 4 is selected from CR 4 or N;
  • R 1 , R 2 , R 3 and R 4 are independently selected from the group consisting of H, D, halogen, -CN, -NO 2 , -OR a , -SR a , -NR b R c , -C(O)R a , -C(O)OR a , -C(O)NR b R c , -OC(O)R a , -NR b C(O)R a , -S(O)R a , -S(O) 2 R a , C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, 3 to 7 membered heterocyclic, C 6-10 aryl or 5 to 10 membered heteroaryl;
  • the above groups are optionally substituted by one or more D until completely deuterated;
  • each R a , R b and R c are independently selected from the group consisting of H, D, C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, 3 to 7 membered heterocyclic, C 6 a -10 aryl or a 5 to 10 membered heteroaryl; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • L 1 is selected from C(R 1a )(R 2a ), O, S, N(R 1a ), C(O), S(O) or S(O) 2 ;
  • X is selected from O, S, N(R 1c ) or C(R 1c )(R 2c );
  • Y is selected from O, S, N(R 1d ) or C(R 1d )(R 2d );
  • L 3 is selected from C(R 1f )(R 2f ), O, S, N(R 1f ), C(O), S(O) or S(O) 2 ;
  • L 4 is selected from C(R 1g )(R 2g ), O, S, N(R 1g ), C(O), S(O) or S(O) 2 ;
  • L 5 is selected from C(R 1h )(R 2h ), O, S, N(R 1h ), C(O), S(O) or S(O) 2 ;
  • R is selected from H, D, C 1-6 alkyl, C 1-6 haloalkyl, C 3-7 cycloalkyl, 3 to 7 membered heterocyclic, C 6-10 aryl or 5 to 10 membered heteroaryl a group; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • n is selected from 1, 2, 3, 4 or 5;
  • n is selected from 1, 2 or 3;
  • R 1a and R 2a is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1c and R 2c is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1d and R 2d is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1f and R 2f is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1g and R 2g is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1h and R 2h is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • Substituents present on different atoms in -X-(L 1 ) m -Y- may be bonded to form a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, a C 6-14 aryl group or a 3 to 10 membered hetero An aryl group; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • the invention relates to the above compound, which is of formula (II'):
  • a 1 is selected from CR 1 or N;
  • R 1 and R 3 are independently selected from the group consisting of H, D, halogen, -CN, -NO 2 , -OR a , -SR a , -NR b R c , C 1-6 alkyl, C 1-6 haloalkyl a C 3-7 cycloalkyl group, a 3 to 7 membered heterocyclic group, a C 6-10 aryl group or a 5 to 10 membered heteroaryl group; wherein the above group is optionally substituted by one or more D until it is completely deuterated generation;
  • each R a , R b and R c are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; wherein the above groups are optionally substituted by one or more D until completely deuterated generation;
  • L 1 is selected from C(R 1a )(R 2a ), O, S, N(R 1a ), C(O), S(O) or S(O) 2 ;
  • X is selected from O or C(R 1c )(R 2c );
  • Y is selected from CH 2 , CHD, CD 2 , CH(R 1d ), CD(R 1d ) or C(R 1d )(R 2d );
  • R 1a and R 2a is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1c and R 2c are each independently selected from H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membered heteroaryl group
  • R 1d and R 2d is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1f and R 2f is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • n is selected from 1, 2 or 3;
  • Substituents present on different atoms in -X-(L 1 ) m -Y- may be bonded to form a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, a C 6-14 aryl group or a 3 to 10 membered hetero An aryl group; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • the invention relates to the above compounds, which are of formula (III'-1) or (III'-2):
  • a 1 is selected from CR 1 or N;
  • R 1 and R 3 are independently selected from the group consisting of H, D, halogen, -CN, -NO 2 , -OR a , -SR a , -NR b R c , C 1-6 alkyl, C 1-6 haloalkyl a C 3-7 cycloalkyl group, a 3 to 7 membered heterocyclic group, a C 6-10 aryl group or a 5 to 10 membered heteroaryl group; wherein the above group is optionally substituted by one or more D until it is completely deuterated generation;
  • each R a , R b and R c are independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; wherein the above groups are optionally substituted by one or more D until completely deuterated generation;
  • X is selected from O or C(R 1c )(R 2c );
  • Y is selected from CH 2 , CHD, CD 2 , CH(R 1d ), CD(R 1d ) or C(R 1d )(R 2d );
  • R 1a and R 2a is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1c and R 2c is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1d and R 2d is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • R 1f and R 2f is independently selected from the group consisting of H, D, halogen, -C 0-6 alkylene-CN, -C 0-6 alkylene-OR a , -C 0-6 alkylene-SR a , -C 0-6 alkylene-NR b R c , -C 0-6 alkylene-C(O)R a , -C 0-6 alkylene-C(O)OR a , -C 0 -6 alkylene-C(O)NR b R c , C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -C 0-6 alkylene -C 3-6 cycloalkyl, -C 0-6 alkylene-3 to 7 membered heterocyclic group, -C 0-6 alkylene-C 6-10 aryl or -C 0-6 alkylene a 5- to 10-membere
  • n is selected from 1, 2 or 3;
  • m -Y- may be bonded to form a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, and a C 6-14 aryl group. Or a 3 to 10 membered heteroaryl; wherein the above group is optionally substituted with one or more D until completely deuterated;
  • the invention relates to the above compounds, which are of formula (IV'-1) or (IV'-2):
  • R 3 is selected from the group consisting of H, D, halogen, -CN or -NO 2 ;
  • R 1a and R 2a are each independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; wherein the above groups are optionally substituted by one or more D until fully deuterated;
  • Y is selected from CH 2 or C(Me)(Me); wherein the above groups are optionally substituted by one or more D until completely deuterated;
  • the present invention relates to the above compound, which is of the formula (V'-1) or (V'-2):
  • R 3 is selected from the group consisting of H, D, halogen, -CN or -NO 2 ;
  • R 1a and R 2a are each independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; wherein the above groups are optionally substituted by one or more D until fully deuterated;
  • R 3 is selected from the group consisting of H, D, halogen, -CN or -NO 2 ;
  • R 1a and R 2a are each independently selected from H, D, C 1-6 alkyl or C 1-6 haloalkyl; and R 1a and R 2a are not simultaneously H or D; wherein the above group is optionally one Or multiple D substitutions until complete degeneration;
  • a 1 is CR 1 ; in another specific embodiment, A 1 is CH; in another specific embodiment, A 1 is CD; in another specific embodiment, A 1 is N.
  • a 2 is CR 2 ; in another specific embodiment, A 2 is CH; in another specific embodiment, A 2 is CD; in another specific embodiment, A 2 is N.
  • a 3 is CR 3 ; in another specific embodiment, A 3 is CH; in another specific embodiment, A 3 is CD; in another specific embodiment, A 3 is CF; In another specific embodiment, A 3 is N.
  • a 4 is CR 4 ; in another specific embodiment, A 4 is CH; in another specific embodiment, A 4 is CD; in another specific embodiment, A 4 is N.
  • R 1 , R 2 , R 3 and R 4 are independently H; in another specific embodiment, R 1 , R 2 , R 3 and R 4 are independently D; in another In a particular embodiment, R 1 , R 2 , R 3 and R 4 are independently halo; in another specific embodiment, R 1 , R 2 , R 3 and R 4 are independently -CN; In an embodiment, R 1 , R 2 , R 3 and R 4 are independently -NO 2 ; in another specific embodiment, R 1 , R 2 , R 3 and R 4 are independently -OR a ; In a specific embodiment, R 1 , R 2 , R 3 and R 4 are independently -SR a ; in another specific embodiment, R 1 , R 2 , R 3 and R 4 are independently -NR b R c ; In another specific embodiment, R 1 , R 2 , R 3 and R 4 are independently -C(O)R a ; in another specific embodiment, R 1 , R 2 , R 3 and R 4
  • L 1 is C(R 1a )(R 2a ); in another specific embodiment, L 1 is O; in another specific embodiment, L 1 is S; In an embodiment, L 1 is N(R 1a ); in another specific embodiment, L 1 is C(O); in another specific embodiment, L 1 is S(O); in another embodiment In the scheme, L 1 is S(O) 2 .
  • L 2 is C(R 1b )(R 2b ); in another specific embodiment, L 2 is O; in another specific embodiment, L 2 is S; In an embodiment, L 2 is N(R 1b ); in another specific embodiment, L 2 is C(O); in another specific embodiment, L 2 is S(O); in another embodiment In the scheme, L 2 is S(O) 2 .
  • L 3 is C(R 1f )(R 2f ); in another specific embodiment, L 3 is O; in another specific embodiment, L 3 is S; In an embodiment, L 3 is N(R 1f ); in another specific embodiment, L 3 is C(O); in another specific embodiment, L 3 is S(O); in another embodiment In the scheme, L 3 is S(O) 2 .
  • L 4 is C(R 1g )(R 2g ); in another specific embodiment, L 4 is O; in another specific embodiment, L 4 is S; In an embodiment, L 4 is N(R 1g ); in another specific embodiment, L 4 is C(O); in another specific embodiment, L 4 is S(O); in another embodiment In the scheme, L 4 is S(O) 2 .
  • L 5 is C(R 1h )(R 2h ); in another specific embodiment, L 5 is O; in another specific embodiment, L 5 is S; In an embodiment, L 5 is N(R 1h ); in another specific embodiment, L 5 is C(O); in another specific embodiment, L 5 is S(O); in another embodiment In the scheme, L 5 is S(O) 2 .
  • X is O; in another specific embodiment, X is S; in another specific embodiment, X is N(R 1c ); in another specific embodiment, X is C (R 1c )(R 2c ).
  • Y is O; in another specific embodiment, Y is S; in another specific embodiment, Y is N(R 1d ); in another specific embodiment, Y is C (R 1d ) (R 2d ).
  • W is O; in another specific embodiment, W is S; in another specific embodiment, W is N(R 1e ); in another specific embodiment, W is C (R 1e )(R 2e ).
  • R is H; in another specific embodiment, R is D; in another specific embodiment, R is C1-6 alkyl; in another specific embodiment, R is C 1-6 haloalkyl; in another specific embodiment, R is C 3-7 cycloalkyl; in another specific embodiment, R is 3 to 7 membered heterocyclyl; in another embodiment R is a C 6-10 aryl group; in another specific embodiment, R is a 5 to 10 membered heteroaryl group; in another specific embodiment, each group defined in R is optionally one Replace with multiple D until completely deuterated.
  • m is 1; in another specific embodiment, m is 2; in another specific embodiment, m is 3; in another specific embodiment, m is 4; In a specific embodiment, m is 5.
  • n is 1; in another specific embodiment, n is 2; in another specific embodiment, n is 3.
  • R 1a and R 2a /R 1b and R 2b /R 1c and R 2c /R 1d and R 2d /R 1e and R 2e /R 1f and R 2f /R 1g and R 2g /R 1h and R 2h are independently H; in another specific embodiment, R 1a and R 2a /R 1b and R 2b /R 1c and R 2c /R 1d and R 2d /R 1e and R 2e /R 1f and R 2f /R 1g and R 2g /R 1h and R 2h are independently D; in another specific embodiment, R 1a and R 2a /R 1b and R 2b /R 1c and R 2c /R 1d and R 2d /R 1e and R 2e /R 1f and R 2f /R 1g and R 2g /R 1h and R 2h are independently halogen; in another specific embodiment, R 1a and R 2a /R
  • Substituents present on different atoms in -X-(L 1 ) m -Y- may be bonded to form a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, a C 6-14 aryl group or a 3 to 10 membered hetero Aryl.
  • a C 3-10 cycloalkyl group is formed; in another specific embodiment, a 3 to 10 membered heterocyclyl group is formed; in another specific embodiment, a C 6-14 aryl group is formed; In another specific embodiment, a 3 to 10 membered heteroaryl group is formed; in another specific embodiment, a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, a C 6-14 aryl group or 3 to 3 is formed.
  • the 10-membered heteroaryl is optionally substituted with one or more D until it is completely substituted.
  • the substituents present on different atoms in -(L 2 ) n -W- may be bonded to form a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, a C 6-14 aryl group or a 3 to 10 membered heteroaryl group. .
  • a C 3-10 cycloalkyl group is formed; in another specific embodiment, a 3 to 10 membered heterocyclyl group is formed; in another specific embodiment, a C 6-14 aryl group is formed; In another specific embodiment, a 3 to 10 membered heteroaryl group is formed; in another specific embodiment, a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, a C 6-14 aryl group or 3 to 3 is formed.
  • the 10-membered heteroaryl is optionally substituted with one or more D until it is completely substituted.
  • the substituents present on different atoms in -L 5 -(L 3 ) n -L 4 - may be bonded to form a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, a C 6-14 aryl group or 3 to 10 Meta-heteroaryl.
  • a C 3-10 cycloalkyl group is formed; in another specific embodiment, a 3 to 10 membered heterocyclyl group is formed; in another specific embodiment, a C 6-14 aryl group is formed; In another specific embodiment, a 3 to 10 membered heteroaryl group is formed; in another specific embodiment, a C 3-10 cycloalkyl group, a 3 to 10 membered heterocyclic group, a C 6-14 aryl group or 3 to 3 is formed.
  • the 10-membered heteroaryl is optionally substituted with one or more D until it is completely substituted.
  • first R 1c or R 2c on one carbon atom may be ringed with R 1a or R 2a on the second carbon atom, or R 1c or R 2c on the first carbon atom may be bonded to R on the third carbon atom 1d or R 2d may form a ring, or R 1a or R 2a on the second carbon atom may form a ring with R 1d or R 2d on the third carbon atom.
  • R 1b or R 2b may be ringed with R 1b or R 2b on the second carbon atom, or R 1b or R 2b on the first carbon atom may be ringed with R 1e on the third nitrogen atom, or R 1b or R 2b on the second carbon atom may form a ring with R 1e on the third nitrogen atom.
  • any one of the above specific embodiments, or any combination thereof, may be combined with any one of the other specific embodiments or any combination thereof.
  • any of the technical solutions of A 1 or any combination thereof may be combined with any of A 1 -A 4 , R 1 -R 4 , X, Y, W, R, L 1 -L 5 , m and n.
  • the present invention is intended to include a combination of all such technical solutions, which are limited in scope and are not listed one by one.
  • the compounds of the invention may be selected from the group consisting of:
  • the compounds of the invention may include one or more asymmetric centers, and thus may exist in a variety of stereoisomeric forms, for example, enantiomeric and/or diastereomeric forms.
  • the compounds of the invention may be in the form of individual enantiomers, diastereomers or geometric isomers (e.g., cis and trans isomers), or may be in the form of a mixture of stereoisomers, A mixture of a racemate and a mixture rich in one or more stereoisomers is included.
  • the isomers can be separated from the mixture by methods known to those skilled in the art, including: chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of a chiral salt; or preferred isomers can be passed Prepared by asymmetric synthesis.
  • HPLC high pressure liquid chromatography
  • the organic compound can form a complex with the solvent in which it reacts or precipitates or crystallizes from the solvent. These complexes are referred to as "solvates.” When the solvent is water, the complex is referred to as a "hydrate.”
  • the present invention encompasses all solvates of the compounds of the invention.
  • solvate means a form of a compound or a salt thereof which is usually combined with a solvent which is formed by a solvolysis reaction. This physical association can include hydrogen bonding.
  • Conventional solvents include water, methanol, ethanol, acetic acid, DMSO, THF, diethyl ether, and the like.
  • the compounds described herein can be prepared, for example, in crystalline form, and can be solvated.
  • Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric solvates and non-stoichiometric solvates. In some cases, the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated into the crystal lattice of the crystalline solid.
  • “Solvate” includes solvates and isolatable solvates in solution. Representative solvates include hydrates, ethanolates, and methanolates.
  • hydrate refers to a compound that binds to water. Generally, the ratio of the number of water molecules contained in the hydrate of the compound to the number of molecules of the compound in the hydrate is determined.
  • a hydrate of a compound can be represented, for example, by the formula R.x H 2 O, wherein R is the compound, and x is a number greater than zero.
  • a given compound can form more than one hydrate type, including, for example, a monohydrate (x is 1), a lower hydrate (x is a number greater than 0 and less than 1, for example, a hemihydrate (R ⁇ 0.5 H 2 ) O)) and polyhydrate (x is a number greater than 1, for example, dihydrate (R ⁇ 2 H 2 O) and hexahydrate (R ⁇ 6 H 2 O)).
  • the compounds of the invention may be in amorphous or crystalline form (polymorph). Furthermore, the compounds of the invention may exist in one or more crystalline forms. Accordingly, the invention includes within its scope all amorphous or crystalline forms of the compounds of the invention.
  • polymorph refers to a crystalline form (or a salt, hydrate or solvate thereof) of a compound in a particular crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms typically have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optoelectronic properties, stability, and solubility. Recrystallization solvents, crystallization rates, storage temperatures, and other factors can result in a crystalline form that predominates. Various polymorphs of the compounds can be prepared by crystallization under different conditions.
  • the invention also includes isotopically-labeled compounds which are equivalent to those described for formula (I), but one or more of the atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number common in nature.
  • isotopes which may be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, for example 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, 18, respectively. O, 17 O, 31 P, 32 P, 35 S, 18 F and 36 Cl.
  • Compounds of the invention, prodrugs thereof and pharmaceutically acceptable salts of said compounds or of said prodrugs containing such isotopes and/or other isotopes of other atoms are within the scope of the invention.
  • Certain isotopically-labeled compounds of the invention, such as those incorporating radioisotopes (e.g., 3 H and 14 C), are useful in drug and/or substrate tissue distribution assays. Ruthenium, i.e., 3 H and carbon-14, i.e., 14 C isotopes, are particularly preferred because they are easy to prepare and detect.
  • isotopically labeled compounds of the formula (I) of the present invention and prodrugs thereof can generally be prepared by substituting readily available isotopically labeled reagents for non-isotopes in the following schemes and/or the procedures disclosed in the examples and preparations. Labeled reagents.
  • prodrugs are also included within the context of the present invention.
  • the term "prodrug” as used herein refers to a compound which is converted in vivo to an active form thereof having a medical effect by, for example, hydrolysis in blood.
  • Pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems, ACSSymposium Series Vol. 14, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and D. Fleisher, S. Ramon, and H. Barbra "Improved oral drug delivery: Solubility limitations overcome by the use of prodrugs", Advanced Drug Delivery Reviews (1996) 19(2) 115-130, each introduction This article serves as a reference.
  • a prodrug is any covalently bonded compound of the invention which, when administered to a patient, releases the parent compound in vivo.
  • Prodrugs are typically prepared by modifying functional groups in such a way that the modifications can be cleaved by routine manipulation or in vivo to yield the parent compound.
  • Prodrugs include, for example, a compound of the invention wherein a hydroxy, amino or thiol group is bonded to any group which, when administered to a patient, can be cleaved to form a hydroxy, amino or thiol group.
  • representative examples of prodrugs include, but are not limited to, the hydroxy, thiol and amino functional acetate/amide, formate/amide and benzoate/amide derivatives of the compounds of formula (I).
  • an ester such as a methyl ester, an ethyl ester or the like can be used.
  • the ester itself may be active and/or may hydrolyze under conditions in humans.
  • Suitable pharmaceutically acceptable in vivo hydrolysable ester groups include those groups which readily decompose in the human body to release the parent acid or a salt thereof.
  • compositions, formulations and kits are provided.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention (also referred to as "active ingredient") and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition comprises an effective amount of the active component.
  • the pharmaceutical composition comprises a therapeutically effective amount of the active component.
  • the pharmaceutical composition comprises a prophylactically effective amount of the active component.
  • a pharmaceutically acceptable excipient for use in the present invention refers to a non-toxic carrier, adjuvant or vehicle which does not destroy the pharmacological activity of the compound formulated together.
  • Pharmaceutically acceptable carriers, adjuvants, or vehicles that can be used in the compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (eg, human serum albumin) ), buffer substances (such as phosphate), glycine, sorbic acid, potassium sorbate, a mixture of partial glycerides of saturated plant fatty acids, water, salt or electrolyte (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate, Sodium chloride, zinc salt, silica gel, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based material, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylate, wax, polyethylene-polyoxypropylene-e
  • kits e.g., pharmaceutical packs.
  • Kits provided may include a compound of the invention, other therapeutic agents, and first and second containers (eg, vials, ampoules, bottles, syringes, and/or dispersible packages or other materials containing the compounds of the invention, other therapeutic agents) Suitable container).
  • first and second containers eg, vials, ampoules, bottles, syringes, and/or dispersible packages or other materials containing the compounds of the invention, other therapeutic agents
  • kits can also optionally include a third container containing a pharmaceutically acceptable excipient for diluting or suspending a compound of the invention and/or other therapeutic agent.
  • a compound of the invention provided in a first container and a second container is combined with other therapeutic agents to form a unit dosage form.
  • the pharmaceutical composition provided by the present invention can be administered by a variety of routes including, but not limited to, oral administration, parenteral administration, inhalation administration, topical administration, rectal administration, nasal administration, oral administration, vaginal administration.
  • parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intra-articular administration, intra-arterial administration, intrasynovial administration, intrasternal administration. , intracerebroventricular administration, intralesional administration, and intracranial injection or infusion techniques.
  • an effective amount of a compound provided herein is administered.
  • the amount of compound actually administered can be determined by the physician. .
  • the compound provided herein is administered to a subject at risk of developing the condition, typically based on a physician's recommendation and administered under the supervision of a physician, at the dosage level as described above.
  • Subjects at risk of developing a particular condition typically include subjects with a family history of the condition, or those subjects that are particularly susceptible to developing the condition by genetic testing or screening.
  • long-term administration can also be administered chronically.
  • Long-term administration refers to administration of a compound or a pharmaceutical composition thereof for a long period of time, for example, 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or can be continuously administered indefinitely, For example, the rest of the subject.
  • chronic administration is intended to provide a constant level of the compound in the blood over a prolonged period of time, for example, within a therapeutic window.
  • a pharmaceutical composition of the present invention can be further delivered using various methods of administration.
  • a pharmaceutical composition can be administered by bolus injection, for example, to rapidly increase the concentration of a compound in the blood to an effective level.
  • the bolus dose will depend on the target systemic level of the active ingredient, for example, an intramuscular or subcutaneous bolus dose will allow the active component to be slowly released, while a bolus delivered directly to the vein (eg, via IV IV drip) can be more Delivered rapidly so that the concentration of the active ingredient in the blood is rapidly increased to an effective level.
  • the pharmaceutical composition can be administered in a continuous infusion form, for example, by IV intravenous drip to provide a steady state concentration of the active ingredient in the subject's body.
  • a bolus dose of the pharmaceutical composition can be administered first, followed by continued infusion.
  • Oral compositions can be in the form of a bulk liquid solution or suspension or bulk powder. More generally, however, the composition is provided in unit dosage form for ease of precise dosing.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active ingredient suitable to produce the desired therapeutic effect with a suitable pharmaceutical excipient.
  • Typical unit dosage forms include prefilled, pre-measured ampoules or syringes of the liquid compositions, or pills, tablets, capsules and the like in the case of solid compositions.
  • the compound will generally be a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), with the remainder being useful for forming the desired form of administration.
  • a carrier or excipient and a processing aid is provided in unit dosage form for ease of precise dosing.
  • a representative regimen is one to five oral doses per day, especially two to four oral doses, typically three oral doses.
  • each dose provides from about 0.01 to about 20 mg/kg of a compound of the invention, each preferably providing from about 0.1 to about 10 mg/kg, especially from about 1 to about 5 mg/kg.
  • a transdermal dose is generally selected in an amount of from about 0.01 to about 20% by weight, preferably from about 0.1 to about 20% by weight, preferably about 0.1. To about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
  • the injection dose level ranges from about 1 mg/kg/hr to at least 10 mg/kg/hr from about 1 to about 120 hours, especially 24 to 96 hours.
  • a preload bolus of about 0.1 mg/kg to about 10 mg/kg or more can also be administered.
  • the maximum total dose cannot exceed about 2 g/day.
  • Liquid forms suitable for oral administration may include suitable aqueous or nonaqueous vehicles as well as buffers, suspending and dispersing agents, coloring agents, flavoring agents, and the like.
  • the solid form may include, for example, any of the following components, or a compound having similar properties: a binder, for example, microcrystalline cellulose, tragacanth or gelatin; an excipient such as starch or lactose, a disintegrant, For example, alginic acid, Primogel or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silica; a sweetener such as sucrose or saccharin; or a flavoring agent such as mint, water Methyl salicylate or orange flavoring.
  • a binder for example, microcrystalline cellulose, tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrant, For example, alginic acid, Primogel or corn star
  • Injectable compositions are typically based on injectable sterile saline or phosphate buffered saline, or other injectable excipients known in the art.
  • the active compound will typically be a minor component, often from about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
  • transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient.
  • the active component When formulated as an ointment, the active component is typically combined with a paraffin or water miscible ointment base.
  • the active ingredient can be formulated as a cream with, for example, an oil-in-water cream base.
  • Such transdermal formulations are well known in the art and generally include other ingredients for enhancing stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and components are included within the scope of the invention.
  • transdermal administration can be accomplished using a reservoir or a porous membrane type, or a patch of a plurality of solid matrices.
  • compositions for oral administration, injection or topical administration are merely representative.
  • Other materials, processing techniques, and the like are set forth in Section 8 of Remington's Pharmaceutical Sciences, 17th edition, 1985, Mack Publishing Company, Easton, Pennsylvania, which is incorporated herein by reference.
  • the compounds of the invention may also be administered in sustained release form or from a sustained release delivery system.
  • sustained release materials can be found in Remington's Pharmaceutical Sciences.
  • the invention further relates to pharmaceutically acceptable formulations of the compounds of the invention.
  • the formulation comprises water.
  • the formulation comprises a cyclodextrin derivative.
  • the most common cyclodextrins are alpha-, beta- and gamma-cyclodextrins consisting of 6, 7 and 8 alpha-1,4-linked glucose units, respectively, optionally including one on the attached sugar moiety. Or a plurality of substituents including, but not limited to, methylated, hydroxyalkylated, acylated, and sulfoalkyl ether substituted.
  • the cyclodextrin is a sulfoalkyl ether beta-cyclodextrin, eg, sulfobutylether beta-cyclodextrin, also known as Captisol. See, for example, U.S. 5,376,645.
  • the formulation comprises hexapropyl- ⁇ -cyclodextrin (eg, 10-50% in water).
  • Exemplary diseases include cancer, pain, neurological diseases, autoimmune diseases, and inflammation.
  • Cancer includes, for example, lung cancer, colon cancer, breast cancer, prostate cancer, hepatocellular carcinoma, renal cell carcinoma, gastric and esophageal cancer, glioblastoma, head and neck cancer, inflammatory myofibroblastic tumors, and anaplastic large cell lymphocytes.
  • Pain includes, for example, pain from any source or cause, including cancer pain, chemotherapy pain, neuropathic pain, injury pain, or other sources.
  • Autoimmune diseases include, for example, rheumatoid arthritis, Sjogren syndrome, type I diabetes, and lupus.
  • Exemplary neurological diseases include Alzheimer's Disease, Parkinson's Disease, amyotrophic lateral sclerosis, and Huntington's disease.
  • Exemplary inflammatory diseases include atherosclerosis, allergies, and inflammation due to infection or injury.
  • the compounds and pharmaceutical compositions of the invention specifically target tyrosine receptor kinases, specifically ALK, ROS1 and TRK.
  • the compounds and pharmaceutical compositions are useful for preventing, reversing, slowing or inhibiting the activity of one or more of the kinases.
  • the method of treatment targets cancer.
  • the methods are for treating lung cancer or non-small cell lung cancer.
  • the effective amount is intended to mean an amount effective to inhibit a target protein.
  • the target modulation measurements can be performed by conventional analytical methods, such as those described below.
  • the adjustments can be used in a variety of settings, including in vitro assays.
  • the cells are preferably cancer cells that have abnormal signaling due to up-regulation of ALK, ROS1 and TRK.
  • an "effective amount” is intended to mean an amount or dose sufficient to produce the desired therapeutic benefit in an individual in need of such treatment.
  • An effective amount or dose of a compound of the invention may be by conventional methods (e.g., modelling, dose escalation, or clinical trials) as well as conventional factors (e.g., mode or route of drug delivery, pharmacokinetics of the agent, severity and course of infection, individual The health status and weight, and the judgment of the treating physician) are determined.
  • Exemplary dosages range from about 0.1 mg to 1 g per day, or from about 1 mg to 50 mg per day, or from about 50 mg to 250 mg per day or from about 250 mg to 1 g per day.
  • the total dose can be in single or divided dosage units (eg, BID, TID, QID).
  • the dose can be adjusted for prophylactic or maintenance treatment.
  • the dosage or frequency of administration, or both can be reduced depending on the symptoms to an amount that maintains the desired therapeutic or prophylactic effect.
  • treatment can be stopped.
  • the patient may need long-term intermittent treatment. Patients may also require long-term slow treatment.
  • the compounds of the invention described herein can be used in combination with one or more other active ingredients in a pharmaceutical composition or method to treat the diseases and conditions described herein.
  • Other additional active ingredients include other therapeutic agents or agents that alleviate the adverse effects of the therapeutic agent against the intended disease target.
  • the combination can be used to increase efficacy, to ameliorate other disease symptoms, to reduce one or more negative effects, or to reduce the desired dose of a compound of the invention.
  • Additional active ingredients may be formulated as separate pharmaceutical compositions from the compounds of the invention or may be included in a single pharmaceutical composition with the compounds of the invention. Additional active ingredients can be administered simultaneously with, prior to, or subsequent to administration of the compounds of the invention.
  • Combination agents include those additional active ingredients known or observed to be effective in the treatment of the diseases and conditions described herein, including those effective against another target associated with the disease.
  • the compositions and formulations, and methods of treatment of the invention may further comprise other drugs or medicaments, such as other active agents useful for treating or ameliorating a target disease or associated condition or condition.
  • kinase inhibitors such as EGFR inhibitors (eg, erlotinib, gefitinib); Raf inhibitors (eg, Vero) Vemirfenib), VEGFR inhibitors (eg, sunitinib); standard chemotherapeutic agents such as alkylating agents, antimetabolites, antitumor antibiotics, topoisomerase inhibitors, platinum drugs , mitotic inhibitors, antibodies, hormone therapy or corticosteroids.
  • suitable combination agents include anti-inflammatory agents such as NSAIDs.
  • the pharmaceutical compositions of the present invention may additionally comprise one or more of the active agents, and the method of treatment may additionally comprise administering an effective amount of one or more of the active agents.
  • each reaction is carried out in an inert solvent at room temperature to reflux temperature (e.g., 0 ° C to 100 ° C, preferably 0 ° C to 80 ° C).
  • the reaction time is usually from 0.1 to 60 hours, preferably from 0.5 to 24 hours.
  • Example 2-1 (7S,13R)-11-fluoro-7,13-dimethyl-6,7,13,14-tetrahydro-1,15-vinyl bridylpyrazolo[4,3-f ][1,4,8,10]Benzene Preparation of oxatriazatridecane-5(6H)-one (Compound T-2-A).
  • Chiral preparative column CHIRALPAK IC (trade name), 4.6 mm ⁇ 250 mm (inner diameter ⁇ length), 5 ⁇ m (filler particle size)
  • UV detection wavelength 254nm
  • Example 8 (14S) -9- fluoro-14-methyl-13-oxa-pentaazabenz -2,17,20,21,24- pentacyclo [16.5.2.0 2,6 .0 7,12 .0 21,25 ] twenty-five -1(24),7,9,11,18(25),19,22-heptenene-16-one (compound 13a),
  • Chiral preparative column CHIRALPAK IC (trade name), 4.6 mm ⁇ 250 mm (inner diameter ⁇ length), 5 ⁇ m (filler particle size)
  • UV detection wavelength 254nm
  • Chiral preparative column CHIRALPAK IC (trade name), 4.6 mm ⁇ 250 mm (inner diameter ⁇ length), 5 ⁇ m (filler particle size)
  • UV detection wavelength 254nm
  • the compound 20a (606 mg, 1.46 mmol) was dissolved in dichloromethane (20 ml), and then the mixture was added to the Dess-Martin oxidant (928 mg, 2.19 mmol) in an ice bath, and the mixture was gradually stirred at room temperature for 1 hour, and the reaction was monitored by TLC. Diluted with dichloromethane and washed 2-3 times with saturated sodium bicarbonate solution. The organic phase was concentrated and directly poured into the next reaction.
  • Chiral preparative column CHIRALPAK IC (trade name), 4.6 mm ⁇ 250 mm (inner diameter ⁇ length), 5 ⁇ m (filler particle size)
  • UV detection wavelength 254nm
  • the racemic compound 23a was isolated to obtain the target product L-4-A (retention time: 6.28 min, relative content: 22.3%), L-4-B ( Retention time: 18.46min, relative content: 22.4%), L-4-C (retention time: 30.45min, relative content: 22.2%) and L-4-D (retention time: 37.26min, relative content: 22.4%) .
  • the compound 25a (577 mg, 1.46 mmol) was dissolved in dichloromethane (20 ml), and then the mixture was added to the mixture of Dess-Martin oxidant (928 mg, 2.19 mmol), and the mixture was gradually stirred at room temperature for 1 hour, and the reaction was monitored by TLC. Diluted with dichloromethane and washed 2-3 times with saturated sodium bicarbonate solution. The organic phase was concentrated and directly poured into the next reaction.
  • Chiral preparative column CHIRALPAK IC (trade name), 4.6 mm ⁇ 250 mm (inner diameter ⁇ length), 5 ⁇ m (filler particle size)
  • UV detection wavelength 254nm
  • EXAMPLE 12 9-fluoro-15-methylerythromycin -2,17,20,21,24- pentaazabenz pentacyclo [16.5.2.0 2,6 .0 7,12 .0 21,25] pentacosane -1(24),7,9,11,18(25),19,22-heptenene-16-one (compound 32a),
  • the compound 29a (597 mg, 1.46 mmol) was dissolved in dichloromethane (20 ml), and the mixture was evaporated. Diluted with dichloromethane, washed with saturated sodium bicarbonate solution 2-3 times, and the organic phase was concentrated and directly poured into the next reaction.
  • Chiral preparative column CHIRALPAK IC (trade name), 4.6 mm ⁇ 250 mm (inner diameter ⁇ length), 5 ⁇ m (filler particle size)
  • UV detection wavelength 254nm
  • the racemic compound 32a was isolated to obtain the target product L-6-A (retention time: 7.14 min, relative content: 21.6%), L-6-B ( Retention time: 15.69min, relative content: 22.8%), L-6-C (retention time: 29.54min, relative content: 17.6%) and L-6-D (retention time: 34.26min, relative content: 16.9%) .
  • EXAMPLE 13 9-fluoro-15-methylerythromycin -2,11,17,20,21,24- hexaazatetracyclo pentacyclo [16.5.2.0 2,6 .0 7,12 .0 21,25] twenty-five alkyl -1(24),7,9,11,18(25),19,22-heptenene-16-one (compound 41a),
  • the compound 38a (599 mg, 1.46 mmol) was dissolved in dichloromethane (20 ml), and then the mixture was added to the Dess-Martin oxidant (928 mg, 2.19 mmol), and the mixture was gradually stirred at room temperature for 1 hour, and the reaction was monitored by TLC. Diluted with dichloromethane and washed 2-3 times with saturated sodium bicarbonate solution. The organic phase was concentrated and directly poured into the next reaction.
  • Chiral preparative column CHIRALPAK IC (trade name), 4.6 mm ⁇ 250 mm (inner diameter ⁇ length), 5 ⁇ m (filler particle size)
  • UV detection wavelength 254nm
  • the racemic compound 41a was isolated to obtain the target product L-7-A (retention time: 4.77 min, relative content: 30.21%), L-7-B ( Retention time: 15.68min, relative content: 22.5%), L-7-C (retention time: 26.31min, relative content: 10.66%) and L-7-D (retention time: 29.67min, relative content: 23.4%) .
  • Example 14 9-Fluoro embodiment -2,11,17,20,21,24- hexaazatetracyclo pentacyclo [16.5.2.0 2,6 .0 7,12 .0 21,25] pentacosane -1(24),7,9,11,18(25),19,22-heptenene-16-one (compound 45a),
  • the compound 42a (578 mg, 1.46 mmol) was dissolved in dichloromethane (20 ml), and then the mixture was added to the Dess-Martin oxidant (928 mg, 2.19 mmol), and the mixture was gradually stirred at room temperature for 1 hour, and the reaction was monitored by TLC. Diluted with dichloromethane and washed 2-3 times with saturated sodium bicarbonate solution. The organic phase was concentrated and directly poured into the next reaction.
  • Chiral preparative column CHIRALPAK IC (trade name), 4.6 mm ⁇ 250 mm (inner diameter ⁇ length), 5 ⁇ m (filler particle size)
  • UV detection wavelength 254nm
  • the racemic compound 45a was isolated to obtain the target product L-8-A (retention time: 20.15 min, relative content: 43.7%) and L-8-B ( Retention time: 8.25min, relative content: 44.0%).
  • Example 15 (4R) -9- fluoro-4-hydroxy-hexaazatetracyclo -2,11,17,20,21,24- embodiment pentacyclo [16.5.2.0 2,6 .0 7,12 .0 21,25] T-pentadecane -1(24),7,9,11,18(25),19,22-heptenene-16-one (compound 56a),
  • the compound 53a (601 mg, 1.46 mmol) was dissolved in dichloromethane (20 ml), and then the mixture was added to the mixture of Dess-Martin oxidant (928 mg, 2.19 mmol), and the mixture was gradually stirred at room temperature for 1 hour, and the reaction was monitored by TLC. Diluted with dichloromethane and washed 2-3 times with saturated sodium bicarbonate solution. The organic phase was concentrated and directly poured into the next reaction.
  • Chiral preparative column CHIRALPAK IC (trade name), 4.6 mm ⁇ 250 mm (inner diameter ⁇ length), 5 ⁇ m (filler particle size)
  • UV detection wavelength 254nm
  • Example 16 9-fluoro-13-oxa-hexaazatetracyclo -2,11,18,21,22,25- pentacyclo [17.5.2.0 2,6 .0 7,12 .0 22,26] twenty-six alkyl -1(25),7,9,11,19(26),20,23-hepten-17-one (compound 64a),
  • Chiral preparative column CHIRALPAK IC (trade name), 4.6 mm ⁇ 250 mm (inner diameter ⁇ length), 5 ⁇ m (filler particle size)
  • UV detection wavelength 254nm
  • the racemic compound 64a was isolated to obtain the target product L-10-A (retention time: 27.54 min, relative content: 49.4%) and L-10-B ( Retention time: 38.22min, relative content: 49.3%).
  • TRKA, TRKB, and TRKC kinase inhibition can be measured by the HTRF (High Fluorescence Resonance Energy Transfer) method.
  • the reaction was carried out in a 384-well plate at a volume of 20 ⁇ L at 23 °C.
  • TRKA, TRKB or TRKC kinases were mixed with different concentrations of pre-diluted compounds (11 doses, 3 fold concentration gradient, initial concentration 300 nM, 2% DMSO final concentration) for ten minutes at each concentration.
  • the corresponding substrate and ATP were added and reacted at room temperature for 20 minutes (where a positive control was set: negative for blank control and positive for Entrectinib).
  • the detection reagent (reagent in the HTRF Kinase TK kit) was added, and after incubation at room temperature for 30 minutes, the enzyme activity in the presence of the compound of the present invention at each concentration was measured by a PerkinElmer Envision plate reader, and different concentrations of the compound were calculated. The inhibitory activity on the enzyme activity was then fitted to the inhibitory activity of the enzyme activity at different concentrations of the compound according to the four-parameter equation according to Graphpad 5.0 software, and the IC 50 value was calculated. The data for the compounds tested in this analysis are presented in Table 1.
  • the anti-proliferative activity of the compounds of the present invention against tumor cells cultured in vitro was examined by the CGT method.
  • the KM12 cell line was maintained in RPMI-1640 medium containing 10% fetal bovine serum and antibiotics, and the cells grown in log phase were harvested and seeded in 96-well plates, and cultured in a 5% carbon dioxide incubator at 37 ° C overnight. .
  • After the test compound was dissolved in DMSO, it was diluted in a 3-fold concentration gradient with a concentration of 9 compounds. Different concentrations of pre-formulated compounds were transferred to the cell plates, three wells per concentration, and incubation continued for 72 h. The final concentration of DMSO in the system was 0.1% and the starting concentration of the test compound was 300 nM.
  • the Ba/F3 parental cells, Ba/F3 LMNA-NTRK1 and Ba/F3 LMNA-NTRK1-G595R cells were maintained in RPMI-1640 medium containing 10% fetal bovine serum and antibiotics, respectively, and the cells growing in log phase were harvested. They were separately planted in 96-well plates and cultured overnight at 37 ° C in a 5% carbon dioxide incubator. Among them, 8 ng/ml IL-3 was added to the Ba/F3 parental cells. After the test compound was dissolved in DMSO, it was diluted by a 3.16-fold concentration gradient to a concentration of 9 compounds. Different concentrations of pre-formulated compounds were transferred to the cell plates, three wells per concentration, and incubation continued for 72 h.
  • the final concentration of DMSO in the system was 0.1%, the starting concentration of the test compound in the Ba/F3 parental cells was 10 ⁇ M, and the starting concentration of the test compound in Ba/F3 LMNA-NTRK1 and Ba/F3 LMNA-NTRK1-G595R cells was 1 ⁇ M.
  • CellTiter-Glo reagent was added to the cell plate, incubated for 30 minutes at room temperature to stabilize the luminescence signal, and the inhibitory activity of the compound of the present invention on cell proliferation at each concentration was measured by a PerkinElmer Envision plate reader. The inhibitory activity of cell proliferation under different concentrations of compounds was fitted according to Graphpad 5.0 software, and the IC 50 value was calculated. The results showed that the compound of the present invention had almost no inhibitory effect on Ba/F3 parental cells, but had an inhibitory effect on Ba/F3 LMNA-NTRK1 and Ba/F3 LMNA-NTRK1-G595R cells.
  • Rats were fed a standard diet and given water. Fasting began 16 hours before the test.
  • the drug was dissolved with PEG400 and dimethyl sulfoxide. Blood was collected from the eyelids at a time point of 0.083 hours, 0.25 hours, 0.5 hours, 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, and 24 hours after administration.
  • Rats were briefly anesthetized after inhalation of ether, and 300 ⁇ L of blood samples were collected from the eyelids in test tubes. There was 30 ⁇ L of 1% heparin salt solution in the test tube. The tubes were dried overnight at 60 ° C before use. After the blood sample collection was completed at the last time point, the rats were anesthetized with ether and sacrificed.
  • Plasma samples were centrifuged at 5000 rpm for 5 minutes at 4 ° C to separate plasma from red blood cells. Pipette 100 ⁇ L of plasma into a clean plastic centrifuge tube, indicating the name and time of the compound. Plasma was stored at -80 °C prior to analysis. The concentration of the compound of the invention in plasma was determined by LC-MS/MS. Pharmacokinetic parameters were calculated based on the plasma concentration of each animal at different time points.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Pain & Pain Management (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Pyridine Compounds (AREA)

Abstract

一种对蛋白激酶活性具有抑制作用的二(杂)芳基大环化合物,以及它们的制备和用途。具体地,公开了式(I)所示的二(杂)芳基大环化合物、或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物,以及含有该化合物及其衍生物的药物组合物和使用其的方法,包括治疗癌症、疼痛、神经疾病、自身免疫疾病和炎症的方法。

Description

用于抑制蛋白激酶活性的二(杂)芳基大环化合物 技术领域
本发明属于医药技术领域,尤其涉及具体二(杂)芳基大环化合物,包含其的药物组合物,以及它们的制备方法和用于治疗癌症、疼痛、神经疾病、自身免疫疾病和炎症的用途。
背景技术
蛋白激酶是细胞生长、增殖和存活的关键调控剂。遗传和表观遗传改变在癌症细胞中累积,导致驱动恶性过程的信号转导途径的异常活化。这些信号传导途径的抑制代表了靶向癌症疗法的有前景的干预机会。
ALK属于受体酪氨酸激酶的胰岛素受体(IR)超家族。由于造血、实体和间质肿瘤中的重要作用,ALK是癌症治疗干预的有吸引力分子靶标。
原肌凝蛋白相关受体酪氨酸激酶(TRK)是神经滋养蛋白(NT)的高亲和力受体。TRK家族的成员高度表达于神经起源的细胞中。由于TRK在疼痛感觉以及肿瘤细胞生长和存活信号传导中起重要作用,因此TRK受体激酶的抑制剂作为疼痛和癌症的治疗剂可提供益处。
ROS1激酶是具有未知配体的受体酪氨酸激酶。已报告ROS1激酶经历基因重排以在许多人类癌症中产生组成型活性融合蛋白,所述癌症包括胶质母细胞瘤、非小细胞肺癌(NSCLC)、胆管癌、卵巢癌、胃腺癌、结肠直肠癌、炎症性肌纤维母细胞肿瘤、血管肉瘤和上皮样血管内皮瘤。
业内仍需要具有所需医药性质的这些多种蛋白质或酪氨酸激酶靶标的小分子抑制剂,以解决激酶抑制剂不断变化的突变抗性。在本发明所述的二(杂)芳基大环化合物能有效结合ALK、ROS1、TRK激酶的ATP结合位点,对这些蛋白起到抑制的作用。更关键的是,它能结合这些蛋白的突变型,如ALK G1202R、ALK L1196M、ROS1 G2032R或TRKA G595R等。本发明的化合物是野生和突变ALK、ROS1、TRK等的抑制剂且将可用于治疗具有ALK、ROS1或TRK中的一或多者信号传导异常的受试者。
发明概述
针对以上技术问题,本发明提供了一种新的二(杂)芳基大环化合物及包含该化合物的组合物及其用途,其对野生和突变ALK、ROS1、TRK等激酶具有更好的抑制活性、更低的副作用和/或更好的药效学/药代动力学性能,治疗ALK、ROS1或TRK中的一或多者介导的疾病。
对此,本发明采用以下技术方案:
在一个方面中,本发明提供了式(I)化合物:
Figure PCTCN2019082051-appb-000001
其中,
A 1选自CR 1或N;
A 2选自CR 2或N;
A 3选自CR 3或N;
A 4选自CR 4或N;
其中R 1、R 2、R 3和R 4独立地选自H、D、卤素、-CN、-NO 2、-OR a、-SR a、-NR bR c、-C(O)R a、-C(O)OR a、-C(O)NR bR c、-OC(O)R a、-NR bC(O)R a、-S(O)R a、-S(O) 2R a、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
其中各个R a、R b和R c独立地选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
L 1选自C(R 1a)(R 2a)、O、S、N(R 1a)、C(O)、S(O)或S(O) 2
L 2选自C(R 1b)(R 2b)、O、S、N(R 1b)、C(O)、S(O)或S(O) 2
X选自O、S、N(R 1c)或C(R 1c)(R 2c);
Y选自O、S、N(R 1d)或C(R 1d)(R 2d);
W选自O、S、N(R 1e)或C(R 1e)(R 2e);
R选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
m选自1、2、3、4或5;
n选自1、2或3;
其中,
各个R 1a和R 2a各自独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1a、R 2a和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1b和R 2b各自独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1b、R 2b和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
R 1c和R 2c各自独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1c、R 2c和它们所连接的碳原子一起形成C 3-7 环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
R 1d和R 2d各自独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1d、R 2d和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
R 1e和R 2e各自独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1e、R 2e和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
-X-(L 1) m-Y-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
-(L 2) n-W-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
在另一方面,本发明提供了含有本发明化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物和药学上可接受的赋形剂的药物组合物。在具体实施方案中,本发明化合物以有效量提供在所述药物组合物中。在具体实施方案中,本发明化合物以治疗有效量提供。在具体实施方案中,本发明化合物以预防有效量提供。
在另一方面,本发明提供了含有本发明化合物和药学上可接受的赋形剂的药物组合物,其还含有其它治疗剂。
在另一方面,本发明提供了包含本发明化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物,任选地其它治疗剂,以及药学上可接受的载剂、佐剂或媒剂的试剂盒。
在另一方面,本发明提供了一种如上所述的药物组合物的制备方法,包括以下步骤:将药学上可接受的赋形剂与本发明化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物进行混合,从而形成药物组合物。
在另一方面,本发明提供了在需要其的受试者中治疗癌症、疼痛、神经疾病、自身免疫疾病和炎症的方法,所述方法包括给予受试者有效量的本发明化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物或本发明的药物组合物。在具体实施方案中,口服、皮下、静脉内或肌肉内给药所述化合物。在具体实施方案中,长期给药所述化合物。
在另一方面,本发明涉及抑制蛋白质或酪氨酸激酶(包括ALK、ROS1、TRK中的一或多者)的 方法,其包括使所述激酶中的一或多者与有效量的至少一种式(I)的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物,和/或与至少一种本发明的药物组合物接触,其中所述接触是在活体外、离体或活体内。
由随后的具体实施方式、实施例和权利要求,本发明的其他目的和优点将对于本领域技术人员显而易见。
定义
化学定义
下面更详细地描述具体官能团和化学术语的定义。
当列出数值范围时,既定包括每个值和在所述范围内的子范围。例如“C 1-6烷基”包括C 1、C 2、C 3、C 4、C 5、C 6、C 1-6、C 1-5、C 1-4、C 1-3、C 1-2、C 2-6、C 2-5、C 2-4、C 2-3、C 3-6、C 3-5、C 3-4、C 4-6、C 4-5和C 5-6烷基。
“C 1-6烷基”是指具有1至6个碳原子的直链或支链饱和烃基团,本文也称为“低级烷基”。在一些实施方案中,C 1-4烷基是特别优选的。所述烷基的实例包括但不限于:甲基(C 1)、乙基(C 2)、正丙基(C 3)、异丙基(C 3)、正丁基(C 4)、叔丁基(C 4)、仲丁基(C 4)、异丁基(C 4)、正戊基(C 5)、3-戊基(C 5)、戊基(C 5)、新戊基(C 5)、3-甲基-2-丁基(C 5)、叔戊基(C 5)和正己基(C 6)。不论烷基前是否修饰有“取代的”,烷基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
“C 2-6烯基”是指具有2至6个碳原子和一个或多个碳-碳双键(例如,1、2或3个碳-碳双键)的直链或支链烃基团。一个或多个碳-碳双键可以在内部(例如,在2-丁烯基中)或端部(例如,在1-丁烯基中)。在一些实施方案中,C 2-4烯基是特别优选的。所述烯基的实例包括但不限于:乙烯基(C 2)、1-丙烯基(C 3)、2-丙烯基(C 3)、1-丁烯基(C 4)、2-丁烯基(C 4)、丁二烯基(C 4)、戊烯基(C 5)、戊二烯基(C 5)、己烯基(C 6),等等。不论烯基前是否修饰有“取代的”,烯基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
“C 2-6炔基”是指具有2至6个碳原子、一个或多个碳-碳叁键(例如,1、2或3个碳-碳叁键)以及任选一个或多个碳-碳双键(例如,1、2或3个碳-碳双键)的直链或支链烃基团。在一些实施方案中,C 2-4炔基是特别优选的。在一些实施方案中,炔基不含有任何双键。一个或多个碳叁键可以在内部(例如,在2-丁炔基中)或端部(例如,在1-丁炔基中)。所述炔基的实例包括但不限于:乙炔基(C 2)、1-丙炔基(C 3)、2-丙炔基(C 3)、1-丁炔基(C 4)、2-丁炔基(C 4)、戊炔基(C 5)、己炔基(C 6),等等。不论炔基前是否修饰有“取代的”,炔基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
“C 1-6亚烷基”是指除去C 1-6烷基的另一个氢而形成的二价基团,并且可以是取代或未取代的亚烷基。在一些实施方案中,C 1-4亚烷基是特别优选的。未取代的所述亚烷基包括但不限于:亚甲基(-CH 2-)、亚乙基(-CH 2CH 2-)、亚丙基(-CH 2CH 2CH 2-)、亚丁基(-CH 2CH 2CH 2CH 2-)、亚戊基(-CH 2CH 2CH 2CH 2CH 2-)、亚己基(-CH 2CH 2CH 2CH 2CH 2CH 2-),等等。示例性的取代的所述亚烷基,例如,被一个或多个烷基(甲 基)取代的所述亚烷基,包括但不限于:取代的亚甲基(-CH(CH 3)-、-C(CH 3) 2-)、取代的亚乙基(-CH(CH 3)CH 2-、-CH 2CH(CH 3)-、-C(CH 3) 2CH 2-、-CH 2C(CH 3) 2-)、取代的亚丙基(-CH(CH 3)CH 2CH 2-、-CH 2CH(CH 3)CH 2-、-CH 2CH 2CH(CH 3)-、-C(CH 3) 2CH 2CH 2-、-CH 2C(CH 3) 2CH 2-、-CH 2CH 2C(CH 3) 2-),等等。
“C 0-6亚烷基”包括化学键和如上定义的C 1-6亚烷基。
“C 1-6烷氧基”是指基团-OR,其中,R为取代或未取代的C 1-6烷基。在一些实施方案中,C 1-4烷氧基是特别优选的。具体的所述烷氧基包括但不限于:甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、叔丁氧基、仲丁氧基、正戊氧基、正己氧基和1,2-二甲基丁氧基。
“卤代”或“卤素”是指氟(F)、氯(Cl)、溴(Br)和碘(I)。在一些实施方案中,卤素基团是F、Cl或Br。在一些实施方案中,卤素基团是F或Cl。在一些实施方案中,卤素基团是F。
因此,“C 1-6卤代烷基”和“C 1-6卤代烷氧基”是指上述“C 1-6烷基”和“C 1-6烷氧基”,其被一个或多个卤素基团取代。在一些实施方案中,C 1-4卤代烷基是特别优选的,更优选C 1-2卤代烷基。在一些实施方案中,C 1-4卤代烷氧基是特别优选的,更优选C 1-2卤代烷氧基。示例性的所述卤代烷基包括但不限于:-CF 3、-CH 2F、-CHF 2、-CHFCH 2F、-CH 2CHF 2、-CF 2CF 3、-CCl 3、-CH 2Cl、-CHCl 2、2,2,2-三氟-1,1-二甲基-乙基,等等。示例性的所述卤代烷氧基包括但不限于:-OCH 2F、-OCHF 2、-OCF 3,等等。
“C 3-10环烷基”是指具有3至10个环碳原子和零个杂原子的非芳香环烃基团。在一些实施方案中,C 3-7环烷基是优选的,C 3-6环烷基是特别优选的,更优选C 5-6环烷基。环烷基还包括其中上述环烷基环与一个或多个芳基或杂芳基稠合的环体系,其中连接点在环烷基环上,且在这样的情况中,碳的数目继续表示环烷基体系中的碳的数目。示例性的所述环烷基包括但不限于:环丙基(C 3)、环丙烯基(C 3)、环丁基(C 4)、环丁烯基(C 4)、环戊基(C 5)、环戊烯基(C 5)、环己基(C 6)、环己烯基(C 6)、环已二烯基(C 6)、环庚基(C 7)、环庚烯基(C 7)、环庚二烯基(C 7)、环庚三烯基(C 7)、环辛基(C 8)、环辛烯基(C 8)、二环[2.2.1]庚基(C 7)、二环[2.2.2]辛基(C 8)、环壬基(C 9)、环壬烯基(C 9)、环癸基(C 10)、环癸烯基(C 10)、八氢-1H-茚基(C 9)、十氢萘基(C 10)、螺[4.5]癸基(C 10),等等。不论环烷基前是否修饰有“取代的”,环烷基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
“3至10元杂环基”或是指具有环碳原子和1至4个环杂原子的3至10元非芳香环系的基团,其中,每个杂原子独立地选自氮、氧、硫、硼、磷和硅。在包含一个或多个氮原子的杂环基中,只要化合价允许,连接点可为碳或氮原子。在一些实施方案中,3至7元杂环基是优选的,其为具有环碳原子和1至3个环杂原子的3至7元非芳香环系;在一些实施方案中,3至6元杂环基是特别优选的,其为具有环碳原子和1至3个环杂原子的3至6元非芳香环系;更优选5至6元杂环基,其为具有环碳原子和1至3个环杂原子的5至6元非芳香环系。杂环基还包括其中上述杂环基环与一个或多个环烷基、芳基或杂芳基稠合的环体系,其中连接点在杂环基环上;且在这样的情况下,环成员的数目继续表示在杂环基环体系中环成员的数目。不论杂环基前是否修饰有“取代的”,杂环基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
示例性的包含一个杂原子的3元杂环基包括但不限于:氮杂环丙烷基、氧杂环丙烷基、硫杂环丙 烷基(thiorenyl)。示例性的含有一个杂原子的4元杂环基包括但不限于:氮杂环丁烷基、氧杂环丁烷基和硫杂环丁烷基。示例性的含有一个杂原子的5元杂环基包括但不限于:四氢呋喃基、二氢呋喃基、四氢噻吩基、二氢噻吩基、吡咯烷基、二氢吡咯基和吡咯基-2,5-二酮。示例性的包含两个杂原子的5元杂环基包括但不限于:二氧杂环戊烷基、氧硫杂环戊烷基(oxasulfuranyl)、二硫杂环戊烷基(disulfuranyl)和噁唑烷-2-酮。示例性的包含三个杂原子的5元杂环基包括但不限于:三唑啉基、噁二唑啉基和噻二唑啉基。示例性的包含一个杂原子的6元杂环基包括但不限于:哌啶基、四氢吡喃基、二氢吡啶基和硫杂环己烷基(thianyl)。示例性的包含两个杂原子的6元杂环基包括但不限于:哌嗪基、吗啉基、二硫杂环己烷基、二噁烷基。示例性的包含三个杂原子的6元杂环基包括但不限于:六氢三嗪基(triazinanyl)。示例性的含有一个杂原子的7元杂环基包括但不限于:氮杂环庚烷基、氧杂环庚烷基和硫杂环庚烷基。示例性的包含一个杂原子的8元杂环基包括但不限于:氮杂环辛烷基、氧杂环辛烷基和硫杂环辛烷基。示例性的与C 6芳基环稠合的5元杂环基(在本文中也称作5,6-双环杂环基)包括但不限于:二氢吲哚基、异二氢吲哚基、二氢苯并呋喃基、二氢苯并噻吩基、苯并噁唑啉酮基,等等。示例性的与C 6芳基环稠合的6元杂环基(本文还指的是6,6-双环杂环基)包括但不限于:四氢喹啉基、四氢异喹啉基,等等。
“C 6-14芳基”是指具有6-14个环碳原子和零个杂原子的单环或多环的(例如,双环或三环)4n+2芳族环体系(例如,具有以环状排列共享的6、10或14个π电子)的基团。在一些实施方案中,芳基具有六个环碳原子(“C 6芳基”;例如,苯基)。在一些实施方案中,芳基具有十个环碳原子(“C 10芳基”;例如,萘基,例如,1-萘基和2-萘基)。在一些实施方案中,芳基具有十四个环碳原子(“C 14芳基”;例如,蒽基)。在一些实施方案中,C 6-10芳基是特别优选的,更优选C 6芳基。芳基还包括其中上述芳基环与一个或多个环烷基或杂环基稠合的环系统,而且连接点在所述芳基环上,在这种情况下,碳原子的数目继续表示所述芳基环系统中的碳原子数目。不论芳基前是否修饰有“取代的”,芳基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
“5至10元杂芳基”是指具有环碳原子和1-4个环杂原子的5-10元单环或双环的4n+2芳族环体系(例如,具有以环状排列共享的6或10个π电子)的基团,其中每个杂原子独立地选自氮、氧和硫。在含有一个或多个氮原子的杂芳基中,只要化合价允许,连接点可以是碳或氮原子。杂芳基双环系统在一个或两个环中可以包括一个或多个杂原子。杂芳基还包括其中上述杂芳基环与一个或多个环烷基或杂环基稠合的环系统,而且连接点在所述杂芳基环上,在这种情况下,碳原子的数目继续表示所述杂芳基环系统中的碳原子数目。在一些实施方案中,5至6元杂芳基是特别优选的,其为具有环碳原子和1-4个环杂原子的5-6元单环或双环的4n+2芳族环体系。不论杂芳基前是否修饰有“取代的”,杂芳基的每个独立地任选被取代,例如,1至5个取代基、1至3个取代基或1个取代基,适当的取代基如下定义。
示例性的含有一个杂原子的5元杂芳基包括但不限于:吡咯基、呋喃基和噻吩基。示例性的含有两个杂原子的5元杂芳基包括但不限于:咪唑基、吡唑基、噁唑基、异噁唑基、噻唑基和异噻唑基。示例性的含有三个杂原子的5元杂芳基包括但不限于:三唑基、噁二唑基和噻二唑基。示例性的含有四个杂原子的5元杂芳基包括但不限于:四唑基。示例性的含有一个杂原子的6元杂芳基包括但不限 于:吡啶基。示例性的含有两个杂原子的6元杂芳基包括但不限于:哒嗪基、嘧啶基和吡嗪基。示例性的含有三个或四个杂原子的6元杂芳基分别包括但不限于:三嗪基和四嗪基。示例性的含有一个杂原子的7元杂芳基包括但不限于:氮杂环庚三烯基、氧杂环庚三烯基和硫杂环庚三烯基。示例性的5,6-双环杂芳基包括但不限于:吲哚基、异吲哚基、吲唑基、苯并三唑基、苯并噻吩基、异苯并噻吩基、苯并呋喃基、苯并异呋喃基、苯并咪唑基、苯并噁唑基、苯并异噁唑基、苯并噁二唑基、苯并噻唑基、苯并异噻唑基、苯并噻二唑基、茚嗪基和嘌呤基。示例性的6,6-双环杂芳基包括但不限于:萘啶基、喋啶基、喹啉基、异喹啉基、噌琳基、喹喔啉基、酞嗪基和喹唑啉基。
示例性的碳原子上的取代基包括但不局限于:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OR aa、-ON(R bb) 2、-N(R bb) 2、-N(R bb) 3 +X -、-N(OR cc)R bb、-SH、-SR aa、-SSR cc、-C(=O)R aa、-CO 2H、-CHO、-C(OR cc) 2、-CO 2R aa、-OC(=O)R aa、-OCO 2R aa、-C(=O)N(R bb) 2、-OC(=O)N(R bb) 2、-NR bbC(=O)R aa、-NR bbCO 2R aa、-NR bbC(=O)N(R bb) 2、-C(=NR bb)R aa、-C(=NR bb)OR aa、-OC(=NR bb)R aa、-OC(=NR bb)OR aa、-C(=NR bb)N(R bb) 2、-OC(=NR bb)N(R bb) 2、-NR bbC(=NR bb)N(R bb) 2、-C(=O)NR bbSO 2R aa、-NR bbSO 2R aa、-SO 2N(R bb) 2、-SO 2R aa、-SO 2OR aa、-OSO 2R aa、-S(=O)R aa、-OS(=O)R aa、-Si(R aa) 3、-OSi(R aa) 3、-C(=S)N(R bb) 2、-C(=O)SR aa、-C(=S)SR aa、-SC(=S)SR aa、-SC(=O)SR aa、-OC(=O)SR aa、-SC(=O)OR aa、-SC(=O)R aa、-P(=O) 2R aa、-OP(=O) 2R aa、-P(=O)(R aa) 2、-OP(=O)(R aa) 2、-OP(=O)(OR cc) 2、-P(=O) 2N(R bb) 2、-OP(=O) 2N(R bb) 2、-P(=O)(NR bb) 2、-OP(=O)(NR bb) 2、-NR bbP(=O)(OR cc) 2、-NR bbP(=O)(NR bb) 2、-P(R cc) 2、-P(R cc) 3、-OP(R cc) 2、-OP(R cc) 3、-B(R aa) 2、-B(OR cc) 2、-BR aa(OR cc)、烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
或者在碳原子上的两个偕氢被基团=O、=S、=NN(R bb) 2、=NNR bbC(=O)R aa、=NNR bbC(=O)OR aa、=NNR bbS(=O) 2R aa、=NR bb或=NOR cc取代;
R aa的每个独立地选自烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基,或者两个R aa基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R bb的每个独立地选自:氢、-OH、-OR aa、-N(R cc) 2、-CN、-C(=O)R aa、-C(=O)N(R cc) 2、-CO 2R aa、-SO 2R aa、-C(=NR cc)OR aa、-C(=NR cc)N(R cc) 2、-SO 2N(R cc) 2、-SO 2R cc、-SO 2OR cc、-SOR aa、-C(=S)N(R cc) 2、-C(=O)SR cc、-C(=S)SR cc、-P(=O) 2R aa、-P(=O)(R aa) 2、-P(=O) 2N(R cc) 2、-P(=O)(NR cc) 2、烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基,或者两个R bb基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R cc的每个独立地选自氢、烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基,或者两个R cc基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R dd的每个独立地选自:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OR ee、-ON(R ff) 2、-N(R ff) 2,、-N(R ff) 3 +X -、-N(OR ee)R ff、-SH、-SR ee、-SSR ee、-C(=O)R ee、-CO 2H、-CO 2R ee、-OC(=O)R ee、-OCO 2R ee、 -C(=O)N(R ff) 2、-OC(=O)N(R ff) 2、-NR ffC(=O)R ee、-NR ffCO 2R ee、-NR ffC(=O)N(R ff) 2、-C(=NR ff)OR ee、-OC(=NR ff)R ee、-OC(=NR ff)OR ee、-C(=NR ff)N(R ff) 2、-OC(=NR ff)N(R ff) 2、-NR ffC(=NR ff)N(R ff) 2、-NR ffSO 2R ee、-SO 2N(R ff) 2、-SO 2R ee、-SO 2OR ee、-OSO 2R ee、-S(=O)R ee、-Si(R ee) 3、-OSi(R ee) 3、-C(=S)N(R ff) 2、-C(=O)SR ee、-C(=S)SR ee、-SC(=S)SR ee、-P(=O) 2R ee、-P(=O)(R ee) 2、-OP(=O)(R ee) 2、-OP(=O)(OR ee) 2、烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基、杂芳基,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代,或者两个偕R dd取代基可结合以形成=O或=S;
R ee的每个独立地选自烷基、卤代烷基、烯基、炔基、碳环基、芳基、杂环基和杂芳基,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代;
R ff的每个独立地选自氢、烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基,或者两个R ff基团结合形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代;
R gg的每个独立地是:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OC 1-6烷基、-ON(C 1-6烷基) 2、-N(C 1-6烷基) 2、-N(C 1-6烷基) 3 +X -、-NH(C 1-6烷基) 2 +X -、-NH 2(C 1-6烷基) +X -、-NH 3 +X -、-N(OC 1-6烷基)(C 1-6烷基)、-N(OH)(C 1-6烷基)、-NH(OH)、-SH、-SC 1-6烷基、-SS(C 1-6烷基)、-C(=O)(C 1-6烷基)、-CO 2H、-CO 2(C 1-6烷基)、-OC(=O)(C 1-6烷基)、-OCO 2(C 1-6烷基)、-C(=O)NH 2、-C(=O)N(C 1-6烷基) 2、-OC(=O)NH(C 1-6烷基)、-NHC(=O)(C 1-6烷基)、-N(C 1-6烷基)C(=O)(C 1-6烷基)、-NHCO 2(C 1-6烷基)、-NHC(=O)N(C 1-6烷基) 2、-NHC(=O)NH(C 1-6烷基)、-NHC(=O)NH 2、-C(=NH)O(C 1-6烷基)、-OC(=NH)(C 1-6烷基)、-OC(=NH)OC 1-6烷基、-C(=NH)N(C 1-6烷基) 2、-C(=NH)NH(C 1-6烷基)、-C(=NH)NH 2、-OC(=NH)N(C 1-6烷基) 2、-OC(NH)NH(C 1-6烷基)、-OC(NH)NH 2、-NHC(NH)N(C 1-6烷基) 2、-NHC(=NH)NH 2、-NHSO 2(C 1-6烷基)、-SO 2N(C 1-6烷基) 2、-SO 2NH(C 1-6烷基)、-SO 2NH 2、-SO 2C 1-6烷基、-SO 2OC 1-6烷基、-OSO 2C 1-6烷基、-SOC 1-6烷基、-Si(C 1-6烷基) 3、-OSi(C 1-6烷基) 3、-C(=S)N(C 1-6烷基) 2、C(=S)NH(C 1-6烷基)、C(=S)NH 2、-C(=O)S(C 1-6烷基)、-C(=S)SC 1-6烷基、-SC(=S)SC 1-6烷基、-P(=O) 2(C 1-6烷基)、-P(=O)(C 1-6烷基) 2、-OP(=O)(C 1-6烷基) 2、-OP(=O)(OC 1-6烷基) 2、C 1-6烷基、C 1-6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 7碳环基、C 6-C 10芳基、C 3-C 7杂环基、C 5-C 10杂芳基;或者两个偕R gg取代基可结合形成=O或=S;其中,X -为反离子。
示例性的氮原子上取代基包括但不局限于:氢、-OH、-OR aa、-N(R cc) 2、-CN、-C(=O)R aa、-C(=O)N(R cc) 2、-CO 2R aa、-SO 2R aa、-C(=NR bb)R aa、-C(=NR cc)OR aa、-C(=NR cc)N(R cc) 2、-SO 2N(R cc) 2、-SO 2R cc、-SO 2OR cc、-SOR aa、-C(=S)N(R cc) 2、-C(=O)SR cc、-C(=S)SR cc、-P(=O) 2R aa、-P(=O)(R aa) 2、-P(=O) 2N(R cc) 2、-P(=O)(NR cc) 2、烷基、卤代烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基,或者连接至氮原子的两个R cc基团结合形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代,且其中R aa、R bb、R cc和R dd如上所述。
“氘代”或“D”指化合物或基团中的一个或多个氢被氘所取代;氘代可以是一取代、二取代、多取代或完全取代。术语“一个或多个氘代的”与“一次或多次氘代”可互换使用。
“非氘代的化合物”是指含氘原子比例不高于天然氘同位素含量(0.015%)的化合物。
氘在氘代位置的氘同位素含量至少是大于天然氘同位素含量0.015%,较佳地大于30%,更佳地大于50%,更佳地大于75%,更佳地大于95%,更佳地大于99%。
术语“药学上可接受的盐”是指,在可靠的医学判断范围内,适合与人和低等动物的组织接触而没有过度毒性、刺激性、变态反应等等,并且与合理的益处/危险比例相称的那些盐。药学上可接受的盐在本领域是众所周知的。例如,Berge等人在J.Pharmaceutical Sciences(1977)66:1-19中详细描述的药学上可接受的盐。本发明化合物的药学上可接受的盐包括衍生自合适的无机和有机酸和无机和有机碱的盐。药学上可接受的无毒的酸加成盐的实例是与无机酸形成的盐,例如盐酸、氢溴酸、磷酸、硫酸和高氯酸,或与有机酸形成的盐,例如乙酸、草酸、马来酸、酒石酸、枸橼酸、琥珀酸或丙二酸。也包括使用本领域常规方法形成的盐,例如,离子交换方法。其它药学上可接受的盐包括:已二酸盐、海藻酸盐、抗坏血酸盐、天冬氨酸盐、苯磺酸盐、苯甲酸盐、重硫酸盐、硼酸盐、丁酸盐、樟脑酸盐、樟脑磺酸盐、柠檬酸盐、环戊丙酸盐、二葡糖酸盐、十二烷基硫酸盐、乙磺酸盐、甲酸盐、富马酸盐、葡萄糖酸盐、甘油磷酸盐、葡糖酸盐、半硫酸盐、庚酸盐、己酸盐、氢碘酸盐、2-羟基-乙磺酸盐、乳糖酸盐、乳酸盐、月桂酸盐、月桂基硫酸盐、苹果酸盐、马来酸盐、丙二酸盐、甲磺酸盐、2-萘磺酸盐、烟酸盐、硝酸盐、油酸盐、草酸盐、棕榈酸盐、双羟萘酸盐、果胶酯酸盐、过硫酸盐、3-苯丙酸盐、磷酸盐、苦味酸盐、特戊酸盐、丙酸盐、硬脂酸盐、琥珀酸盐、硫酸盐、酒石酸盐、硫氰酸盐、对甲苯磺酸盐、十一烷酸盐、戊酸盐,等等。衍生自合适的碱的药学上可接受的盐包括碱金属、碱土金属、铵和N +(C 1-4烷基) 4盐。代表性的碱金属或碱土金属盐包括钠、锂、钾、钙、镁盐,等等。如果合适的话,其它的药学上可接受的盐包括与反离子形成的无毒的铵盐、季铵盐和胺阳离子,反离子例如卤离子、氢氧根、羧酸根、硫酸根、磷酸根、硝酸根、低级烷基磺酸根和芳基磺酸根。
“活性代谢物“指式(I)化合物或其盐在体内代谢的药理学活性产物。化合物的前药和活性代谢物可使用业内已知或可获得的常规技术来测定。
给药的“受试者”包括但不限于:人(即,任何年龄组的男性或女性,例如,儿科受试者(例如,婴儿、儿童、青少年)或成人受试者(例如,年轻的成人、中年的成人或年长的成人))和/或非人的动物,例如,哺乳动物,例如,灵长类(例如,食蟹猴、恒河猴)、牛、猪、马、绵羊、山羊、啮齿动物、猫和/或狗。在一些实施方案中,受试者是人。在一些实施方案中,受试者是非人动物。本文可互换使用术语“人”、“患者”和“受试者”。
“疾病”、“障碍”和“病症”在本文中可互换地使用。
除非另作说明,否则,本文使用的术语“治疗”包括受试者患有具体疾病、障碍或病症时所发生的作用,它降低疾病、障碍或病症的严重程度,或延迟或减缓疾病、障碍或病症的发展(“治疗性治疗”),还包括受试者开始患有具体疾病、障碍或病症之前发生的作用(“预防性治疗”)。
“组合”以及相关术语是指同时或依次给药本发明的治疗剂。例如,本发明化合物可以与另一治疗剂以分开的单位剂型同时或依次给药,或与另一治疗剂一起呈单一单位剂型同时给药。
具体实施方式
化合物
本文中,“本发明化合物”指的是以下的式(I)化合物-式(VI)化合物和式(I’)化合物-式(V’)化合物(包括各式的子集,例如式(III-1)化合物),或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
在一个实施方案中,本发明涉及式(I)化合物:
Figure PCTCN2019082051-appb-000002
其中,
A 1选自CR 1或N;
A 2选自CR 2或N;
A 3选自CR 3或N;
A 4选自CR 4或N;
其中R 1、R 2、R 3和R 4独立地选自H、D、卤素、-CN、-NO 2、-OR a、-SR a、-NR bR c、-C(O)R a、-C(O)OR a、-C(O)NR bR c、-OC(O)R a、-NR bC(O)R a、-S(O)R a、-S(O) 2R a、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
其中各个R a、R b和R c独立地选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
L 1选自C(R 1a)(R 2a)、O、S、N(R 1a)、C(O)、S(O)或S(O) 2
L 2选自C(R 1b)(R 2b)、O、S、N(R 1b)、C(O)、S(O)或S(O) 2
X选自O、S、N(R 1c)或C(R 1c)(R 2c);
Y选自O、S、N(R 1d)或C(R 1d)(R 2d);
W选自O、S、N(R 1e)或C(R 1e)(R 2e);
R选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
m选自1、2、3、4或5;
n选自1、2或3;
其中,
各个R 1a和R 2a独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1a、R 2a和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1b和R 2b独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1b、R 2b和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1c和R 2c独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1c、R 2c和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1d和R 2d独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1d、R 2d和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1e和R 2e独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1e、R 2e和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
-X-(L 1) m-Y-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
-(L 2) n-W-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
在另一个实施方案中,本发明涉及上述的化合物,其为式(II):
Figure PCTCN2019082051-appb-000003
其中,
A 1选自CR 1或N;
A 4选自CR 4或N;
其中R 1、R 3和R 4独立地选自H、D、卤素、-CN、-NO 2、-OR a、-SR a、-NR bR c、-C(O)R a、-C(O)OR a、-C(O)NR bR c、-OC(O)R a、-NR bC(O)R a、-S(O)R a、-S(O) 2R a、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
其中各个R a、R b和R c独立地选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
X选自O或C(R 1c)(R 2c);
Y选自N(R 1d)或C(R 1d)(R 2d);
W选自O或NH;
m选自1、2、3、4或5;
各个R 1a和R 2a独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1a、R 2a和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1b和R 2b独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1b、R 2b和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
其中,
各个R 1c和R 2c独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1c、R 2c和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1d和R 2d独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1d、R 2d和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
-X-(C(R 1a)(R 2a)) m-Y-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
在另一个实施方案中,本发明涉及上述化合物,其为式(III-1)或(III-2):
Figure PCTCN2019082051-appb-000004
其中,
A 1选自CR 1或N;
A 4选自CR 4或N;
其中R 1、R 3和R 4独立地选自H、D、卤素、-CN、-NO 2、-OR a、-SR a、-NR bR c、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
其中各个R a、R b和R c独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
X选自O或C(R 1c)(R 2c);
Y选自N(R 1d)或C(R 1d)(R 2d);
W选自O或NH;
m选自1、2或3;
各个R 1a和R 2a独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1a、R 2a和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
R 1b选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
其中,
各个R 1c和R 2c独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1c、R 2c和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1d和R 2d独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、 C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1d、R 2d和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
-X-(C(R 1a)(R 2a)) m-Y-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
在另一个实施方案中,本发明涉及上述化合物,其为式(IV-1)或(IV-2):
Figure PCTCN2019082051-appb-000005
其中,
R 3选自H、D、卤素、-CN、-NO 2、-OR a、-SR a或-NR bR c
其中各个R a、R b和R c独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
X选自O或C(R 1c)(R 2c);
Y选自NH、CH 2或C(Me)(Me);其中上述基团任选地被一个或多个D取代,直至完全氘代;
m选自1、2或3;
各个R 1a和R 2a独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1a、R 2a和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
R 1b选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
其中,各个R 1c和R 2c独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1c、R 2c和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
优选地,
R 3选自H、D、卤素、-CN或-NO 2
X为O;
Y选自NH、CH 2或C(Me)(Me);其中上述基团任选地被一个或多个D取代,直至完全氘代;
m选自1、2或3;
R 1a和R 2a独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
R 1b选自C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
优选地,
R 3选自H、D、卤素、-CN或-NO 2
X为O;
Y选自CH 2或C(Me)(Me);其中上述基团任选地被一个或多个D取代,直至完全氘代;
m选自1、2或3;
R 1a和R 2a独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
R 1b选自C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
在另一个实施方案中,本发明涉及上述化合物,其为式(V-1)或(V-2):
Figure PCTCN2019082051-appb-000006
其中,
R 3选自H、D、卤素、-CN或-NO 2
Y选自NH、CH 2、CHD或CD 2
m选自1、2或3;
各个R 1a和R 2a独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
R 1b选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
在另一个实施方案中,本发明涉及上述化合物,其为式(VI-1)或(VI-2):
Figure PCTCN2019082051-appb-000007
其中,
R 3选自H、D、卤素、-CN或-NO 2
R 1a和R 2a独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
R 1b选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
在另一个实施方案中,本发明涉及上述化合物,其为式(I’)化合物:
Figure PCTCN2019082051-appb-000008
其中,
A 1选自CR 1或N;
A 2选自CR 2或N;
A 3选自CR 3或N;
A 4选自CR 4或N;
其中R 1、R 2、R 3和R 4独立地选自H、D、卤素、-CN、-NO 2、-OR a、-SR a、-NR bR c、-C(O)R a、-C(O)OR a、-C(O)NR bR c、-OC(O)R a、-NR bC(O)R a、-S(O)R a、-S(O) 2R a、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
其中各个R a、R b和R c独立地选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
L 1选自C(R 1a)(R 2a)、O、S、N(R 1a)、C(O)、S(O)或S(O) 2
X选自O、S、N(R 1c)或C(R 1c)(R 2c);
Y选自O、S、N(R 1d)或C(R 1d)(R 2d);
L 3选自C(R 1f)(R 2f)、O、S、N(R 1f)、C(O)、S(O)或S(O) 2
L 4选自C(R 1g)(R 2g)、O、S、N(R 1g)、C(O)、S(O)或S(O) 2
L 5选自C(R 1h)(R 2h)、O、S、N(R 1h)、C(O)、S(O)或S(O) 2
R选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
m选自1、2、3、4或5;
n选自1、2或3;
其中,
各个R 1a和R 2a独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1a、R 2a和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1c和R 2c独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1c、R 2c和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1d和R 2d独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1d、R 2d和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1f和R 2f独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1f、R 2f和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1g和R 2g独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1g、R 2g和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1h和R 2h独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1h、R 2h和它们所连接的碳原子一起形成C 3-7 环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
-X-(L 1) m-Y-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
在另一个实施方案中,本发明涉及上述化合物,其为式(II’):
Figure PCTCN2019082051-appb-000009
其中,
A 1选自CR 1或N;
其中R 1和R 3独立地选自H、D、卤素、-CN、-NO 2、-OR a、-SR a、-NR bR c、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
其中各个R a、R b和R c独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
L 1选自C(R 1a)(R 2a)、O、S、N(R 1a)、C(O)、S(O)或S(O) 2
X选自O或C(R 1c)(R 2c);
Y选自CH 2、CHD、CD 2、CH(R 1d)、CD(R 1d)或C(R 1d)(R 2d);
各个R 1a和R 2a独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1a、R 2a和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
R 1c和R 2c各自独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1c、R 2c和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1d和R 2d独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1d、R 2d和它们所连接的碳原子一起形成C 3-7 环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1f和R 2f独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1f、R 2f和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
m选自1、2或3;
-X-(L 1) m-Y-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
在另一个实施方案中,本发明涉及上述化合物,其为式(III’-1)或(III’-2):
Figure PCTCN2019082051-appb-000010
其中,
A 1选自CR 1或N;
其中R 1和R 3独立地选自H、D、卤素、-CN、-NO 2、-OR a、-SR a、-NR bR c、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
其中各个R a、R b和R c独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
X选自O或C(R 1c)(R 2c);
Y选自CH 2、CHD、CD 2、CH(R 1d)、CD(R 1d)或C(R 1d)(R 2d);
各个R 1a和R 2a独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1a、R 2a和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1c和R 2c独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1c、R 2c和它们所连接的碳原子一起形成C 3-7 环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1d和R 2d独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1d、R 2d和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
各个R 1f和R 2f独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1f、R 2f和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
m选自1、2或3;
-X-(C(R 1a)(R 2a)) m-Y-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
在另一个实施方案中,本发明涉及上述化合物,其为式(IV’-1)或(IV’-2):
Figure PCTCN2019082051-appb-000011
其中,
R 3选自H、D、卤素、-CN或-NO 2
R 1a和R 2a各自独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
Y选自CH 2或C(Me)(Me);其中上述基团任选地被一个或多个D取代,直至完全氘代;
或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
在另一个实施方案中,本发明涉及上述化合物,其为式(V’-1)或(V’-2):
Figure PCTCN2019082051-appb-000012
其中,
R 3选自H、D、卤素、-CN或-NO 2
R 1a和R 2a各自独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
优选地,
R 3选自H、D、卤素、-CN或-NO 2
R 1a和R 2a各自独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;且R 1a和R 2a不同时为H或D;其中上述基团任选地被一个或多个D取代,直至完全氘代;
或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
A 1、A 2、A 3和A 4
在一个具体实施方案中,A 1为CR 1;在另一个具体实施方案中,A 1为CH;在另一个具体实施方案中,A 1为CD;在另一个具体实施方案中,A 1为N。
在一个具体实施方案中,A 2为CR 2;在另一个具体实施方案中,A 2为CH;在另一个具体实施方案中,A 2为CD;在另一个具体实施方案中,A 2为N。
在一个具体实施方案中,A 3为CR 3;在另一个具体实施方案中,A 3为CH;在另一个具体实施方案中,A 3为CD;在另一个具体实施方案中,A 3为CF;在另一个具体实施方案中,A 3为N。
在一个具体实施方案中,A 4为CR 4;在另一个具体实施方案中,A 4为CH;在另一个具体实施方案中,A 4为CD;在另一个具体实施方案中,A 4为N。
R 1、R 2、R 3和R 4
在一个具体实施方案中,R 1、R 2、R 3和R 4独立地为H;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为D;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为卤素;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为-CN;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为-NO 2;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为-OR a;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为-SR a;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为-NR bR c;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为-C(O)R a;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为-C(O)OR a;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为-C(O)NR bR c;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为-OC(O)R a;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为-NR bC(O)R a;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为-S(O)R a;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为-S(O) 2R a;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为C 1-6烷基;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为C 1-6卤代烷基;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为C 3-7环烷基;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为3至7元杂环基;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为C 6-10芳基;在另一个具体实施方案中,R 1、R 2、R 3和R 4独立地为5至10元杂芳基;在另一个具体实施方案中,R 1、R 2、R 3和R 4 中所定义的每个基团任选地被一个或多个D所取代,直至完全氘代。
L 1
在一个具体实施方案中,L 1为C(R 1a)(R 2a);在另一个具体实施方案中,L 1为O;在另一个具体实施方案中,L 1为S;在另一个具体实施方案中,L 1为N(R 1a);在另一个具体实施方案中,L 1为C(O);在另一个具体实施方案中,L 1为S(O);在另一个具体实施方案中,L 1为S(O) 2
L 2
在一个具体实施方案中,L 2为C(R 1b)(R 2b);在另一个具体实施方案中,L 2为O;在另一个具体实施方案中,L 2为S;在另一个具体实施方案中,L 2为N(R 1b);在另一个具体实施方案中,L 2为C(O);在另一个具体实施方案中,L 2为S(O);在另一个具体实施方案中,L 2为S(O) 2
L 3
在一个具体实施方案中,L 3为C(R 1f)(R 2f);在另一个具体实施方案中,L 3为O;在另一个具体实施方案中,L 3为S;在另一个具体实施方案中,L 3为N(R 1f);在另一个具体实施方案中,L 3为C(O);在另一个具体实施方案中,L 3为S(O);在另一个具体实施方案中,L 3为S(O) 2
L 4
在一个具体实施方案中,L 4为C(R 1g)(R 2g);在另一个具体实施方案中,L 4为O;在另一个具体实施方案中,L 4为S;在另一个具体实施方案中,L 4为N(R 1g);在另一个具体实施方案中,L 4为C(O);在另一个具体实施方案中,L 4为S(O);在另一个具体实施方案中,L 4为S(O) 2
L 5
在一个具体实施方案中,L 5为C(R 1h)(R 2h);在另一个具体实施方案中,L 5为O;在另一个具体实施方案中,L 5为S;在另一个具体实施方案中,L 5为N(R 1h);在另一个具体实施方案中,L 5为C(O);在另一个具体实施方案中,L 5为S(O);在另一个具体实施方案中,L 5为S(O) 2
X
在一个具体实施方案中,X为O;在另一个具体实施方案中,X为S;在另一个具体实施方案中,X为N(R 1c);在另一个具体实施方案中,X为C(R 1c)(R 2c)。
Y
在一个具体实施方案中,Y为O;在另一个具体实施方案中,Y为S;在另一个具体实施方案中,Y为N(R 1d);在另一个具体实施方案中,Y为C(R 1d)(R 2d)。
W
在一个具体实施方案中,W为O;在另一个具体实施方案中,W为S;在另一个具体实施方案中, W为N(R 1e);在另一个具体实施方案中,W为C(R 1e)(R 2e)。
R
在一个具体实施方案中,R为H;在另一个具体实施方案中,R为D;在另一个具体实施方案中,R为C 1-6烷基;在另一个具体实施方案中,R为C 1-6卤代烷基;在另一个具体实施方案中,R为C 3-7环烷基;在另一个具体实施方案中,R为3至7元杂环基;在另一个具体实施方案中,R为C 6-10芳基;在另一个具体实施方案中,R为5至10元杂芳基;在另一个具体实施方案中,R中所定义的每个基团任选地被一个或多个D所取代,直至完全氘代。
m
在一个具体实施方案中,m为1;在另一个具体实施方案中,m为2;在另一个具体实施方案中,m为3;在另一个具体实施方案中,m为4;在另一个具体实施方案中,m为5。
n
在一个具体实施方案中,n为1;在另一个具体实施方案中,n为2;在另一个具体实施方案中,n为3。
R 1a和R 2a、R 1b和R 2b、R 1c和R 2c、R 1d和R 2d、R 1e和R 2e、R 1f和R 2f、R 1g和R 2g、R 1h和R 2h
在一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为H;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为D;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为卤素;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为-C 0-6亚烷基-CN;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为-C 0-6亚烷基-OR a;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为-C 0-6亚烷基-SR a;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为-C 0-6亚烷基-NR bR c;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为-C 0-6亚烷基-C(O)R a;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为-C 0-6亚烷基-C(O)OR a;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为-C 0-6亚烷基-C(O)NR bR c;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为C 1-6烷基;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为C 1-6卤代烷基;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为C 2-6烯基;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h 独立地为C 2-6炔基;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为-C 0-6亚烷基-C 3-6环烷基;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为-C 0-6亚烷基-3至7元杂环基;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h独立地为-C 0-6亚烷基-C 6-10芳基;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 1g/R 1h和R 2h和R 2d独立地为-C 0-6亚烷基-5至10元杂芳基;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h,连同它们所连接的碳原子一起形成C 3-7环烷基;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h,连同它们所连接的碳原子一起形成3至7元杂环基;在另一个具体实施方案中,R 1a和R 2a/R 1b和R 2b/R 1c和R 2c/R 1d和R 2d/R 1e和R 2e/R 1f和R 2f/R 1g和R 2g/R 1h和R 2h所定义的每个基团任选地被一个或多个D所取代,直至完全取代。
-X-(L 1) m-Y-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基。在一个具体实施方案中,形成C 3-10环烷基;在另一个具体实施方案中,形成3至10元杂环基;在另一个具体实施方案中,形成C 6-14芳基;在另一个具体实施方案中,形成3至10元杂芳基;在另一个具体实施方案中,形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基任选地被一个或多个D所取代,直至完全取代。
-(L 2) n-W-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基。在一个具体实施方案中,形成C 3-10环烷基;在另一个具体实施方案中,形成3至10元杂环基;在另一个具体实施方案中,形成C 6-14芳基;在另一个具体实施方案中,形成3至10元杂芳基;在另一个具体实施方案中,形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基任选地被一个或多个D所取代,直至完全取代。
-L 5-(L 3) n-L 4-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基。在一个具体实施方案中,形成C 3-10环烷基;在另一个具体实施方案中,形成3至10元杂环基;在另一个具体实施方案中,形成C 6-14芳基;在另一个具体实施方案中,形成3至10元杂芳基;在另一个具体实施方案中,形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基任选地被一个或多个D所取代,直至完全取代。
例如,当-X-(L 1) m-Y-表示-C(R 1c)(R 2c)-C(R 1a)(R 2a)-C(R 1d)(R 2d)-时,第一个碳原子上的R 1c或R 2c可以与第二个碳原子上的R 1a或R 2a成环,或者第一个碳原子上的R 1c或R 2c可以与第三个碳原子上的R 1d或R 2d成环,或者第二个碳原子上的R 1a或R 2a可以与第三个碳原子上的R 1d或R 2d成环。又如,当-(L 2) n-W-表示-C(R 1b)(R 2b)-C(R 1b)(R 2b)-N(R 1e)-时,第一个碳原子上的R 1b或R 2b可以与第二个碳原子上的R 1b或R 2b成环,或者第一个碳原子上的R 1b或R 2b可以与第三个氮原子上的R 1e成环,或者第二个碳原子上的R 1b或R 2b可以与第三个氮原子上的R 1e成环。
以上任一具体实施方案中的任一技术方案或其任意组合,可以与其它具体实施方案中的任一技术方案或其任意组合进行组合。例如,A 1的任一技术方案或其任意组合,可以与A 1-A 4、R 1-R 4、X、Y、 W、R、L 1-L 5、m和n的任一技术方案或其任意组合进行组合。本发明旨在包括所有这些技术方案的组合,限于篇幅,不再一一列出。
在另一个实施方案中,本发明化合物可选自下述化合物:
Figure PCTCN2019082051-appb-000013
Figure PCTCN2019082051-appb-000014
Figure PCTCN2019082051-appb-000015
或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
本发明化合物可包括一个或多个不对称中心,且因此可以存在多种立体异构体形式,例如,对映异构体和/或非对映异构体形式。例如,本发明化合物可为单独的对映异构体、非对映异构体或几何异构体(例如顺式和反式异构体),或者可为立体异构体的混合物的形式,包括外消旋体混合物和富含一种或多种立体异构体的混合物。异构体可通过本领域技术人员已知的方法从混合物中分离,所述方法包括:手性高压液相色谱法(HPLC)以及手性盐的形成和结晶;或者优选的异构体可通过不对称合成来制备。
本领域技术人员将理解,有机化合物可以与溶剂形成复合物,其在该溶剂中发生反应或从该溶剂中沉淀或结晶出来。这些复合物称为“溶剂合物”。当溶剂是水时,复合物称为“水合物”。本发明涵盖了本发明化合物的所有溶剂合物。
术语“溶剂合物”是指通常由溶剂分解反应形成的与溶剂相结合的化合物或其盐的形式。这个物理缔合可包括氢键键合。常规溶剂包括包括水、甲醇、乙醇、乙酸、DMSO、THF、乙醚等。本文所述的化合物可制备成,例如,结晶形式,且可被溶剂化。合适的溶剂合物包括药学上可接受的溶剂合物且进一步包括化学计量的溶剂合物和非化学计量的溶剂合物。在一些情况下,所述溶剂合物将能够分离,例如,当一或多个溶剂分子掺入结晶固体的晶格中时。“溶剂合物”包括溶液状态的溶剂合物和可分离的溶剂合物。代表性的溶剂合物包括水合物、乙醇合物和甲醇合物。
术语“水合物”是指与水相结合的化合物。通常,包含在化合物的水合物中的水分子数与该水合物 中该化合物分子数的比率确定。因此,化合物的水合物可用例如通式R·x H 2O代表,其中R是该化合物,和x是大于0的数。给定化合物可形成超过一种水合物类型,包括,例如,单水合物(x为1)、低级水合物(x是大于0且小于1的数,例如,半水合物(R·0.5 H 2O))和多水合物(x为大于1的数,例如,二水合物(R·2 H 2O)和六水合物(R·6 H 2O))。
本发明化合物可以是无定形或结晶形式(多晶型)。此外,本发明化合物可以以一种或多种结晶形式存在。因此,本发明在其范围内包括本发明化合物的所有无定形或结晶形式。术语“多晶型物”是指特定晶体堆积排列的化合物的结晶形式(或其盐、水合物或溶剂合物)。所有的多晶型物具有相同的元素组成。不同的结晶形式通常具有不同的X射线衍射图、红外光谱、熔点、密度、硬度、晶体形状、光电性质、稳定性和溶解度。重结晶溶剂、结晶速率、贮存温度和其他因素可导致一种结晶形式占优。化合物的各种多晶型物可在不同的条件下通过结晶制备。
本发明还包括同位素标记的化合物,它们等同于式(I)所述的那些,但一个或多个原子被原子质量或质量数不同于自然界常见的原子质量或质量数的原子所代替。可以引入本发明化合物中的同位素的实例包括氢、碳、氮、氧、磷、硫、氟和氯的同位素,分别例如 2H、 3H、 13C、 11C、 14C、 15N、 18O、 17O、 31P、 32P、 35S、 18F和 36Cl。含有上述同位素和/或其它原子的其它同位素的本发明化合物、其前体药物和所述化合物或所述前体药物的药学上可接受的盐都属于本发明的范围。某些同位素标记的本发明化合物、例如引入放射性同位素(例如 3H和 14C)的那些可用于药物和/或底物组织分布测定。氚、即 3H和碳-14、即 14C同位素是特别优选的,因为它们容易制备和检测。进而,被更重的同位素取代,例如氘、即 2H,由于代谢稳定性更高可以提供治疗上的益处,例如延长体内半衰期或减少剂量需求,因而在有些情况下可能是优选的。同位素标记的本发明式(I)化合物及其前体药物一般可以这样制备,在进行下述流程和/或实施例与制备例所公开的工艺时,用容易得到的同位素标记的试剂代替非同位素标记的试剂。
此外,前药也包括在本发明的上下文内。本文所用的术语“前药”是指在体内通过例如在血液中水解转变成其具有医学效应的活性形式的化合物。药学上可接受的前药描述于T.Higuchi和V.Stella,Prodrugs as Novel Delivery Systems,A.C.S.Symposium Series的Vol.14,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987,以及D.Fleisher、S.Ramon和H.Barbra“Improved oral drug delivery:solubility limitations overcome by the use of prodrugs”,Advanced Drug Delivery Reviews(1996)19(2)115-130,每篇引入本文作为参考。
前药为任何共价键合的本发明化合物,当将这种前药给予患者时,其在体内释放母体化合物。通常通过修饰官能团来制备前药,修饰是以使得该修饰可以通过常规操作或在体内裂解产生母体化合物的方式进行的。前药包括,例如,其中羟基、氨基或巯基与任意基团键合的本发明化合物,当将其给予患者时,可以裂解形成羟基、氨基或巯基。因此,前药的代表性实例包括(但不限于)式(I)化合物的羟基、巯基和氨基官能团的乙酸酯/酰胺、甲酸酯/酰胺和苯甲酸酯/酰胺衍生物。另外,在羧酸(-COOH)的情况下,可以使用酯,例如甲酯、乙酯等。酯本身可以是有活性的和/或可以在人体体内条件下水解。合适的药学上可接受的体内可水解的酯基包括容易在人体中分解而释放母体酸或其盐的那些基团。
药物组合物、制剂和试剂盒
在另一方面,本发明提供了药物组合物,其包含本发明化合物(还称为“活性组分”)和药学上可接受的赋形剂。在一些实施方案中,所述药物组合物包含有效量的活性组分。在一些实施方案中,所述药物组合物包含治疗有效量的活性组分。在一些实施方案中,所述药物组合物包含预防有效量的活性组分。
用于本发明的药学上可接受的赋形剂是指不会破坏一起配制的化合物的药理学活性的无毒载剂、佐剂或媒剂。可以用于本发明组合物中的药学上可接受的载剂、佐剂或媒剂包括但不限于,离子交换剂、氧化铝、硬脂酸铝、卵磷脂、血清蛋白(如人类血清白蛋白)、缓冲物质(如磷酸盐)、甘氨酸、山梨酸、山梨酸钾、饱和植物脂肪酸的偏甘油酯混合物、水、盐或电解质(如硫酸鱼精蛋白)、磷酸氢二钠、磷酸氢钾、氯化钠、锌盐、硅胶、三硅酸镁、聚乙烯吡咯烷酮、基于纤维素的物质、聚乙二醇、羧甲基纤维素钠、聚丙烯酸酯、蜡、聚乙烯-聚氧丙烯-嵌段聚合物、聚乙二醇以及羊毛脂。
本发明还包括试剂盒(例如,药物包装)。所提供的试剂盒可以包括本发明化合物、其它治疗剂,以及含有本发明化合物、其它治疗剂的第一和第二容器(例如,小瓶、安瓿瓶、瓶、注射器和/或可分散包装或其它合适的容器)。在一些实施方案中,提供的试剂盒还可以任选包括第三容器,其含有用于稀释或悬浮本发明化合物和/或其它治疗剂的药用赋形剂。在一些实施方案中,提供在第一容器和第二容器中的本发明化合物和其它治疗剂组合形成一个单位剂型。
本发明提供的药物组合物可以通过许多途径给药,包括但不限于:口服给药、肠胃外给药、吸入给药、局部给药、直肠给药、鼻腔给药、口腔给药、阴道给药、通过植入剂给药或其它给药方式。例如,本文使用的肠胃外给药包括皮下给药、皮内给药、静脉内给药、肌肉内给药、关节内给药、动脉内给药、滑膜腔内给药、胸骨内给药、脑脊髓膜内给药、病灶内给药、和颅内的注射或输液技术。
通常,给予有效量的本文所提供的化合物。按照有关情况,包括所治疗的病症、选择的给药途径、实际给予的化合物、个体患者的年龄、体重和响应、患者症状的严重程度,等等,可以由医生确定实际上给予的化合物的量。
当用于预防本发明所述病症时,给予处于形成所述病症危险之中的受试者本文所提供的化合物,典型地基于医生的建议并在医生监督下给药,剂量水平如上所述。处于形成具体病症的危险之中的受试者,通常包括具有所述病症的家族史的受试者,或通过遗传试验或筛选确定尤其对形成所述病症敏感的那些受试者。
还可以长期给予本文所提供的药物组合物(“长期给药”)。长期给药是指在长时间内给予化合物或其药物组合物,例如,3个月、6个月、1年、2年、3年、5年等等,或者可无限期地持续给药,例如,受试者的余生。在一些实施方案中,长期给药意欲在长时间内在血液中提供所述化合物的恒定水平,例如,在治疗窗内。
可以使用各种给药方法,进一步递送本发明的药物组合物。例如,在一些实施方案中,可以推注给药药物组合物,例如,为了使化合物在血液中的浓度快速提高至有效水平。推注剂量取决于活性组分的目标全身性水平,例如,肌内或皮下的推注剂量使活性组分缓慢释放,而直接递送至静脉的推注 (例如,通过IV静脉滴注)能够更加快速地递送,使得活性组分在血液中的浓度快速升高至有效水平。在其它实施方案中,可以以持续输液形式给予药物组合物,例如,通过IV静脉滴注,从而在受试者身体中提供稳态浓度的活性组分。此外,在其它实施方案中,可以首先给予推注剂量的药物组合物,而后持续输液。
口服组合物可以采用散装液体溶液或混悬剂或散装粉剂形式。然而,更通常,为了便于精确地剂量给药,以单位剂量形式提供所述组合物。术语“单位剂型”是指适合作为人类患者及其它哺乳动物的单元剂量的物理离散单位,每个单位包含预定数量的、适于产生所需要的治疗效果的活性物质与合适药学赋形剂。典型的单位剂量形式包括液体组合物的预装填的、预先测量的安瓿或注射器,或者在固体组合物情况下的丸剂、片剂、胶囊剂等。在这种组合物中,所述化合物通常为较少的组分(约0.1至约50重量%,或优选约1至约40重量%),剩余部分为对于形成所需给药形式有用的各种载体或赋形剂以及加工助剂。
对于口服剂量,代表性的方案是,每天一个至五个口服剂量,尤其是两个至四个口服剂量,典型地是三个口服剂量。使用这些剂量给药模式,每个剂量提供大约0.01至大约20mg/kg的本发明化合物,优选的剂量各自提供大约0.1至大约10mg/kg,尤其是大约1至大约5mg/kg。
为了提供与使用注射剂量类似的血液水平,或比使用注射剂量更低的血液水平,通常选择透皮剂量,数量为大约0.01至大约20%重量,优选大约0.1至大约20%重量,优选大约0.1至大约10%重量,且更优选大约0.5至大约15%重量。
从大约1至大约120小时,尤其是24至96小时,注射剂量水平在大约0.1mg/kg/小时至至少10mg/kg/小时的范围。为了获得足够的稳定状态水平,还可以给予大约0.1mg/kg至大约10mg/kg或更多的预载推注。对于40至80kg的人类患者来说,最大总剂量不能超过大约2g/天。
适于口服给药的液体形式可包括合适的水性或非水载体以及缓冲剂、悬浮剂和分散剂、着色剂、调味剂,等等。固体形式可包括,例如,任何下列组份,或具有类似性质的化合物:粘合剂,例如,微晶纤维素、黄蓍胶或明胶;赋形剂,例如,淀粉或乳糖,崩解剂,例如,褐藻酸、Primogel或玉米淀粉;润滑剂,例如,硬脂酸镁;助流剂,例如,胶体二氧化硅;甜味剂,例如,蔗糖或糖精;或调味剂,例如,薄荷、水杨酸甲酯或橙味调味剂。
可注射的组合物典型地基于可注射用的无菌盐水或磷酸盐缓冲盐水,或本领域中已知的其它可注射的赋形剂。如前所述,在这种组合物中,活性化合物典型地为较少的组分,经常为约0.05至10%重量,剩余部分为可注射的赋形剂等。
典型地将透皮组合物配制为含有活性组分的局部软膏剂或乳膏剂。当配制为软膏剂时,活性组分典型地与石蜡或可与水混溶的软膏基质组合。或者,活性组分可与例如水包油型乳膏基质一起配制为乳膏剂。这种透皮制剂是本领域中公知的,且通常包括用于提升活性组分或制剂的稳定的皮肤渗透的其它组份。所有这种已知的透皮制剂和组份包括在本发明提供的范围内。
本发明化合物还可通过经皮装置给予。因此,经皮给药可使用贮存器(reservoir)或多孔膜类型、或者多种固体基质的贴剂实现。
用于口服给予、注射或局部给予的组合物的上述组份仅仅是代表性的。其它材料以及加工技术等 阐述于Remington′s Pharmaceutical Sciences,17th edition,1985,Mack Publishing Company,Easton,Pennsylvania的第8部分中,本文以引用的方式引入该文献。
本发明化合物还可以以持续释放形式给予,或从持续释放给药系统中给予。代表性的持续释放材料的描述可在Remington′s Pharmaceutical Sciences中找到。
本发明还涉及本发明化合物的药学上可接受的制剂。在一个实施方案中,所述制剂包含水。在另一个实施方案中,所述制剂包含环糊精衍生物。最常见的环糊精为分别由6、7和8个α-1,4-连接的葡萄糖单元组成的α-、β-和γ-环糊精,其在连接的糖部分上任选包括一个或多个取代基,其包括但不限于:甲基化的、羟基烷基化的、酰化的和磺烷基醚取代。在一些实施方案中,所述环糊精为磺烷基醚β-环糊精,例如,磺丁基醚β-环糊精,也称作Captisol。参见,例如,U.S.5,376,645。在一些实施方案中,所述制剂包括六丙基-β-环糊精(例如,在水中,10-50%)。
适应症
示例性疾病包括癌症、疼痛、神经疾病、自身免疫疾病和炎症。癌症包括(例如)肺癌、结肠癌、乳腺癌、前列腺癌、肝细胞癌、肾细胞癌、胃和食管癌、胶质母细胞瘤、头颈癌、炎症性肌纤维母细胞肿瘤和间变性大细胞淋巴瘤。疼痛包括(例如)任一源或病因的疼痛,包括癌症疼痛、化学治疗的疼痛、神经疼痛、损伤疼痛或其它源。自身免疫疾病包括(例如)类风湿性关节炎、休格伦氏综合症(Sjogren syndrome)、I型糖尿病和狼疮。示例性神经疾病包括阿尔茨海默病(Alzheimer’s Disease)、帕金森氏病(Parkinson’s Disease)、肌萎缩性侧索硬化症和亨廷顿氏病(Huntington’s disease)。示例性炎症疾病包括动脉粥样硬化、过敏和因感染或损伤而炎症。
在一个方面中,本发明的化合物和药物组合物特定靶向酪氨酸受体激酶、具体来说ALK、ROS1和TRK。因此,所述化合物和药物组合物可用于阻止、逆转、减缓或抑制所述激酶中的一或多者的活性。在优选实施方案中,治疗方法靶向癌症。在其它实施方案中,方法用于治疗肺癌或非小细胞肺癌。
在本发明的抑制方法中,“有效量”打算指有效抑制靶标蛋白质的量。所述靶标调节测量可通过常规分析方法(例如那些下文所述者)实施。所述调节可用于各种设置中,包括活体外分析。在所述方法中,细胞优选是由于ALK、ROS1和TRK上调具有异常信号传导的癌症细胞。
在本发明的治疗方法中,“有效量”打算指足以在需要所述治疗的个体中产生所需治疗益处的量或剂量。本发明化合物的有效量或剂量可通过常规方法(例如模型化、剂量递增或临床试验)以及常规因素(例如药物递送的模式或途径、药剂的药代动力学、感染的严重程度和过程、个体的健康状况和体重、和治疗医师的判断)来确定。示例性剂量是在每天约0.1mg到1g、或每天约1mg到50mg、或每天约50mg到250mg或每天约250mg到1g的范围内。总剂量可以单一或分开剂量单位(例如,BID、TID、QID)。
在患者的疾病发生改善后,可调整剂量以便预防性或维持性治疗。例如,可根据症状将给药剂量或给药频率或二者降低到维持所需治疗或预防效应的量。当然,如果症状已减轻到适当程度,那么可停止治疗。然而,任一症状复发时,患者可能需要长期间歇治疗。患者还可需要长期缓慢治疗。
药物组合
本文所述的本发明化合物可与一或多种其它活性成份组合用于药物组合物或方法中以治疗本文所述的疾病和病症。其它额外活性成份包括缓和治疗剂针对预期疾病靶标的不利效应的其它治疗剂或药剂。所述组合可用于增加功效,改善其它疾病症状,降低一或多种负效应,或降低本发明化合物的所需剂量。额外活性成份可调配成与本发明化合物分开的药物组合物或可与本发明化合物包括在单一药物组合物中。额外活性成份可与本发明化合物的给药同时、在其之前或在其之后给药。
组合药剂包括那些已知或观察到在治疗本文所述疾病和病症中有效的额外活性成份,包括那些有效针对与疾病相关的另一靶标。举例来说,本发明的组合物和制剂、以及治疗方法可进一步包含其它药物或医药,例如其它可用于治疗或缓解目标疾病或相关症状或状况的活性剂。对于癌症适应症来说,其它所述药剂包括(但不限于)激酶抑制剂,例如EGFR抑制剂(例如,埃罗替尼、吉非替尼(gefitinib));Raf抑制剂(例如,维罗非尼(vemurafenib))、VEGFR抑制剂(例如,舒尼替尼(sunitinib));标准化学治疗剂,例如烷基化剂、抗代谢物、抗肿瘤抗生素、拓扑异构酶抑制剂、铂药物、有丝分裂抑制剂、抗体、激素疗法或皮质类固醇。对于疼痛适应症来说,适宜组合药剂包括消炎剂,例如NSAID。本发明的药物组合物可另外包含一或多种所述活性剂,并且治疗方法可另外包含给药有效量的一或多种所述活性剂。
实施例
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则份数和百分比为重量份和重量百分比。
通常,在制备流程中,各反应在惰性溶剂中,在室温至回流温度(如0℃~100℃,优选0℃~80℃)下进行。反应时间通常为0.1-60小时,优选地为0.5-24小时。
实施例1(13R)-11-氟-13-甲基-6,7,13,14-四氢-1,15-乙烯桥基吡唑并[4,3-f][1,4,8,10]苯并氧杂三氮 杂十三烷-5(6H)-酮(化合物T-1)的制备。
Figure PCTCN2019082051-appb-000016
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000017
步骤1化合物1的合成
向反应瓶中加入5-氟-2-羟基苯甲醛(1.4g,10.0mmol),R-叔丁基亚磺酰胺(1.21g,10.0mmol),用20ml二氯甲烷溶解,加入碳酸铯(5.21g,16.0mmol),氮气保护下室温搅拌反应18小时,加入过量水淬灭反应,二氯甲烷萃取3-4遍,合并有机相,饱和食盐水洗涤,浓缩,柱层析纯化,真空干燥得到产物2.11g,收率:86.8%。
步骤2化合物2和3的合成
向反应瓶中加入化合物1(1.86g,7.67mmol),氮气保护下加入25ml无水四氢呋喃溶解,-65℃下缓慢滴加3M的甲基溴化镁的无水四氢呋喃溶液(12.8ml,38.4mmol),加毕,升至室温搅拌反应过夜,TLC监测反应完毕,冰浴下滴加20ml水淬灭反应,分出有机相,水相用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析(石油醚∶乙酸乙酯=2∶1)纯化得到0.98g化合物2和1.1g化合物3,收率:100%。
步骤3化合物4的合成
在反应瓶中加入化合物3(1.14g,4.4mmol),加入4N的氯化氢的二氧六环溶液(13.7ml,54.77mmol),室温下搅拌反应4小时,析出白色固体,过滤得到798mg产物,不用纯化直接投入下一步,收率:95%。
步骤4化合物5的合成
向反应瓶中加入化合物4(382mg,2.0mmol)、5-氯-3-硝基吡唑并[1,5-α]嘧啶(396mg,2.0mmol)和DIPEA(N,N-二异丙基乙胺,1.29g,10mmol),加入8ml无水乙醇,加热至80℃反应1小时,TLC检测反应完毕,浓缩除去溶剂,硅胶柱层析纯化得到538mg产物,收率:85%。LC-MS(APCI): m/z=318.3(M+1) +
步骤5化合物6的合成
向反应瓶中加入化合物5(100mg,0.315mmol)、3-羟基丙酸乙酯(55.8mg,0.473mmol)和三苯基膦(124.1mg,0.473mmol),氮气保护下加入5ml无水四氢呋喃,0℃下滴加DIAD(偶氮二甲酸二异丙酯,95.6mg,0.473mmol),加毕,升至室温搅拌反应15小时,浓缩除去溶剂,硅胶柱层析纯化得到51.6mg产物,收率:38%。LC-MS(APCI):m/z=418.1(M+1) +
步骤6化合物7的合成
向反应瓶中加入化合物6(192mg,0.46mmol)、用8ml甲醇溶解,加入一水合氢氧化锂(96.6mg,2.3mmol)的4ml水溶液,加热至50℃反应4-5小时,TLC监测反应完毕,降温至0℃,用1N稀盐酸调PH至弱酸性,乙酸乙酯萃取3-4次,合并有机相,无水硫酸钠干燥,过滤浓缩得到174mg产物,收率:97%。LC-MS(APCI):m/z=390.5(M+1)+。
步骤7化合物8的合成
向反应瓶中加入化合物7(174mg,0.45mmol)、用5ml甲醇溶解,加入催化量的Pd/C,充氢气,室温下搅拌反应5-7小时,TLC监测反应完毕后,过滤除去催化剂,滤液浓缩后得到化合物8粗品,不需纯化直接投入到下一步反应。LC-MS(APCI):m/z=360.4(M+1) +
步骤8化合物T-1的合成
向反应瓶中加入化合物8(161mg,0.45mmol)、用15ml无水DMF溶解,加入FDPP(五氟苯基二苯基磷酸酯,207.5mg,0.54mmol)和DIPEA(290.8mg,2.25mmol),氮气保护下室温搅拌反应过夜,TLC监测反应完毕后,加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析纯化得到目标产物106mg,收率69%。LC-MS(APCI):m/z=342.5(M+1) +1H NMR(400MHz,DMSO-d 6)δ10.11(s,1H),8.56(s,1H),8.12(d,J=2.3Hz,1H),7.35(t,J=4.4Hz,1H),6.98(m,2H),6.52(d,J=2.3Hz,1H),4.25(t,J=3.3Hz,2H),4.01(m,1H),2.66(t,J=3.3Hz,2H),1.21(d,J=5.5Hz,3H).
实施例2-1(7S,13R)-11-氟-7,13-二甲基-6,7,13,14-四氢-1,15-乙烯桥基吡唑并[4,3-f][1,4,8,10]苯并 氧杂三氮杂十三烷-5(6H)-酮(化合物T-2-A)的制备。
Figure PCTCN2019082051-appb-000018
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000019
步骤1化合物9的合成
向反应瓶中加入化合物5(200mg,0.63mmol)、(R)-3-羟基丁酸乙酯(125mg,0.946mmol)和三苯基膦(248.2mg,0.946mmol),氮气保护下加入10ml无水四氢呋喃,0℃下滴加DIAD(191.2mg,0.946mmol),加毕,升至室温搅拌反应15小时,浓缩除去溶剂,硅胶柱层析纯化得到78.8mg产物,收率:29%。LC-MS(APCI):m/z=432.1(M+1) +
步骤2化合物10的合成
向反应瓶中加入化合物9(200mg,0.46mmol)、用8ml甲醇溶解,加入一水合氢氧化锂(96.6mg,2.3mmol)的4ml水溶液,加热至50℃反应4-5小时,TLC监测反应完毕,降温至0℃,用1N稀盐酸调PH至弱酸性,乙酸乙酯萃取3-4次,合并有机相,无水硫酸钠干燥,过滤浓缩得到182mg产物,收率:98%。LC-MS(APCI):m/z=404.5(M+1) +
步骤3化合物11的合成
向反应瓶中加入化合物10(209mg,0.52mmol)、用8ml甲醇溶解,加入催化量的Pd/C,充氢气,室温下搅拌反应5-7小时,TLC监测反应完毕后,过滤除去催化剂,滤液浓缩后得到化合物11粗品,不需纯化直接投入到下一步反应。LC-MS(APCI):m/z=374.6(M+1) +
步骤4化合物T-2-A的合成
向反应瓶中加入化合物11(168mg,0.45mmol)、用15ml无水DMF溶解,加入FDPP(207.5mg,0.54mmol)和DIPEA(290.8mg,2.25mmol),氮气保护下室温搅拌反应过夜,TLC监测反应完毕后,加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析纯化得到目标产物81mg,收率51%。LC-MS(APCI):m/z=356.3(M+1) +1H NMR(400MHz,DMSO-d 6)δ10.13(s,1H),8.53(s,1H),8.11(d,J=2.7Hz,1H),7.35(t,J=4.4Hz,1H),6.99(m,2H),6.50(d,J=2.7Hz,1H),4.06(m,1H),3.99(m,1H),2.66(t,J=3.3Hz,2H),1.26-1.21(m,6H).
实施例2-2(7S,13S)-11-氟-7,13-二甲基-6,7,13,14-四氢-1,15-乙烯桥基吡唑并[4,3-f][1,4,8,10]苯并氧 杂三氮杂十三烷-5(6H)-酮(化合物T-2-B)的制备。
Figure PCTCN2019082051-appb-000020
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000021
步骤1化合物12的合成
在反应瓶中加入化合物2(1.14g,4.4mmol),加入4N的氯化氢的二氧六环溶液(13.7ml,54.77mmol),室温下搅拌反应4小时,析出白色固体,过滤得到798mg产物,不用纯化直接投入下一步,收率:95%。
步骤2化合物13的合成
向反应瓶中加入化合物12(382mg,2.0mmol)、5-氯-3-硝基吡唑并[1,5-α]嘧啶(396mg,2.0mmol)和DIPEA(N,N-二异丙基乙胺,1.29g,10mmol),加入8ml无水乙醇,加热至80℃反应1小时,TLC检测反应完毕,浓缩除去溶剂,硅胶柱层析纯化得到538mg产物,收率:85%。LC-MS(APCI):m/z=318.3(M+1) +
步骤3化合物14的合成
向反应瓶中加入化合物13(200mg,0.63mmol)、(R)-3-羟基丁酸乙酯(125mg,0.946mmol)和三苯基膦(248.2mg,0.946mmol),氮气保护下加入10ml无水四氢呋喃,0℃下滴加DIAD(191.2mg,0.946mmol),加毕,升至室温搅拌反应15小时,浓缩除去溶剂,硅胶柱层析纯化得到78.8mg产物,收率:29%。LC-MS(APCI):m/z=432.1(M+1) +
步骤4化合物15的合成
向反应瓶中加入化合物14(200mg,0.46mmol)、用8ml甲醇溶解,加入一水合氢氧化锂(96.6mg,2.3mmol)的4ml水溶液,加热至50℃反应4-5小时,TLC监测反应完毕,降温至0℃,用1N稀盐酸调PH至弱酸性,乙酸乙酯萃取3-4次,合并有机相,无水硫酸钠干燥,过滤浓缩得到182mg产物, 收率:98%。LC-MS(APCI):m/z=404.5(M+1) +
步骤5化合物16的合成
向反应瓶中加入化合物15(209mg,0.52mmol)、用8ml甲醇溶解,加入催化量的Pd/C,充氢气,室温下搅拌反应5-7小时,TLC监测反应完毕后,过滤除去催化剂,滤液浓缩后得到化合物16粗品,不需纯化直接投入到下一步反应。LC-MS(APCI):m/z=374.6(M+1) +
步骤6化合物T-2-B的合成
向反应瓶中加入化合物16(168mg,0.45mmol)、用15ml无水DMF溶解,加入FDPP(207.5mg,0.54mmol)和DIPEA(290.8mg,2.25mmol),氮气保护下室温搅拌反应过夜,TLC监测反应完毕后,加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析纯化得到目标产物81mg,收率51%。LC-MS(APCI):m/z=356.3(M+1) +1H NMR(400MHz,DMSO-d 6)δ10.13(s,1H),8.53(s,1H),8.11(d,J=2.7Hz,1H),7.35(t,J=4.4Hz,1H),6.99(m,2H),6.50(d,J=2.7Hz,1H),4.06(m,1H),3.99(m,1H),2.66(t,J=3.3Hz,2H),1.26-1.21(m,6H).
实施例3((13R)-11-氟-6,6,13-三甲基-6,7,13,14-四氢-1,15-乙烯桥基吡唑并[4,3-f][1,4,8,10]苯并氧 杂三氮杂十三烷-5(6H)-酮(化合物T-3)的制备。
Figure PCTCN2019082051-appb-000022
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000023
步骤1化合物17的合成
向反应瓶中加入异丁酸乙酯(2.5g,21.5mmol),氮气保护下加入40ml无水四氢呋喃,降温至-40℃,缓慢滴加LDA(二异丙基氨基锂,11.8ml,23.6mmol),加毕升至室温搅拌反应半小时,再降温至-40℃,缓慢滴加二碘甲烷(5.76g,21.5mmol)的10ml无水四氢呋喃溶液,加毕,升至室温反应过 夜,加水淬灭反应,分出有机相,水相用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得到4.62g产物,收率:84%。
步骤2化合物18的合成
向反应瓶中加入化合物5(300mg,0.95mmol),化合物17(291mg,1.14mmol)和碳酸钾(525.2mg,3.8mmol),加入DMF,升温至80℃搅拌反应过夜,TLC监测反应完毕后,降至室温,加入过量水稀释,乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析纯化得到目标产物321.3mg,收率:76%,LC-MS(APCI):m/z=446.1(M+1) +
步骤3化合物19的合成
向反应瓶中加入化合物18(321mg,0.72mmol),用10ml甲醇溶解,加入一水合氢氧化锂(151.5mg,3.5mmol)的3ml水溶液,加热至50℃反应4-5小时,TLC监测反应完毕,降温至0℃,用1N稀盐酸调PH至弱酸性,乙酸乙酯萃取3-4次,合并有机相,无水硫酸钠干燥,过滤浓缩得到279mg产物,收率:93%。LC-MS(APCI):m/z=418.3(M+1) +
步骤4化合物20的合成
向反应瓶中加入化合物19(279mg,0.67mmol),用10ml甲醇溶解,加入催化量的Pd/C,充氢气,室温下搅拌反应5-7小时,TLC监测反应完毕后,过滤除去催化剂,滤液浓缩后得到化合物20粗品,不需纯化直接投入到下一步反应。LC-MS(APCI):m/z=387.9(M+1) +
步骤5化合物T-3的合成
向反应瓶中加入化合物20(259mg,0.67mmol)、用20ml无水DMF溶解,加入FDPP(309mg,0.80mmol)和DIPEA(433mg,3.35mmol),氮气保护下室温搅拌反应过夜,TLC监测反应完毕后,加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析纯化得到目标产物136mg,收率55%。LC-MS(APCI):m/z=370.3(M+1) +1H NMR(400MHz,DMSO-d 6)δ10.15(s,1H),8.56(s,1H),8.12(d,J=2.3Hz,1H),7.37(t,J=4.4Hz,1H),6.99(m,2H),6.52(d,J=2.3Hz,1H),4.23(s,2H),4.00(m,1H),1.21(d,J=5.5Hz,3H).
实施例4(14R)-12-氟-14-甲基-5,6,7,8,14,15-六氢-1,16-乙烯桥基吡唑并[4,3-g][1,4,9,11]苯并氧杂四 氮杂十四烷-5(6H)-酮(化合物T-4)的制备。
Figure PCTCN2019082051-appb-000024
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000025
步骤1化合物21的合成
向反应瓶中加入化合物5(300mg,0.95mmol)、N-叔丁氧羰基-2-氯乙胺(203.3mg,1.13mmol)和碳酸钾(525.2mg,3.8mmol),加入20ml无水DMF溶解,加热至80℃反应过夜,TLC检测反应完毕后加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析纯化得到产物345mg,收率:79%。LC-MS(APCI):m/z=461.2(M+1) +
步骤2化合物22的合成
向反应瓶中加入化合物21(346mg,0.75mmol),加入12ml无水甲醇溶解,加入催化量的Pd/C,充氢气,室温反应1-2小时,TLC检测反应完毕后过滤,滤液浓缩得到产物346mg,不需纯化直接投入下一步反应。LC-MS(APCI):m/z=431.3(M+1) +
步骤3化合物23的合成
向反应瓶中加入化合物22(346mg,0.75mmol),加入4M的氯化氢二氧六环(10ml,40mmol),室温下搅拌反应2-3小时,MS监测反应完毕后浓缩除去溶剂,直接投入到下一步反应。LC-MS(APCI):m/z=331.6(M+1) +
步骤4化合物T-4的合成
向反应瓶中加入化合物23(200mg,0.61mmol),加入15ml二氯甲烷溶剂,加入CDI(N,N′-羰基二咪唑,196mg,1.21mmol),室温下搅拌反应6小时,TLC监测反应完毕,浓缩除去溶剂,硅胶柱层析纯化得到产物134mg,收率:62%。LC-MS(APCI):m/z=357.5(M+1) +1H NMR(400MHz,DMSO-d 6)δ10.11(s,1H),9.85(s,1H),8.56(s,1H),8.12(d,J=2.3Hz,1H),7.35(t,J=4.4Hz,1H),6.98(m,2H),6.51(d,J=2.3Hz,1H),4.35(t,J=3.5Hz,2H),4.01(m,1H),3.85(t,J=3.5Hz,2H),1.24(d,J=5.5Hz,3H).
实施例5(14R)-12-氟-14-甲基-5,6,7,8,14,15-六氢-1,16-乙烯桥基吡唑并[4,3-g][1,4,9,11]苯并四氮 杂十四烷-5(6H)-酮(化合物T-5)的制备。
Figure PCTCN2019082051-appb-000026
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000027
步骤1化合物24的合成
向反应瓶中加入5-氟-2-碘苯乙酮(3.5g,13.26mmol)、(R)-叔丁基亚磺酰胺(2.41g,19.88mmol)和钛酸四乙酯(7.54g,26.52mmol),加入50ml无水四氢呋喃,氮气保护下加热至回流搅拌反应12小时,TLC检测反应完毕,加入水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析纯化得到产物3.94g,收率:81%。LC-MS(APCI):m/z=368.4(M+1) +
步骤2化合物26的合成
向反应瓶中加入化合物24(3.5g,9.54mmol),加入25ml四氢呋喃和0.5ml水溶解,0℃下分批加入硼氢化钠(0.54g,14.3mmol),加毕,升至室温搅拌反应12小时,TLC监测反应完成后,加入水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析纯化得到产物3.27g,收率:93%。LC-MS(APCI):m/z=370.6(M+1) +
步骤3化合物27的合成
向反应瓶中加入化合物26(3.27g,8.87mmol),加入4M的氯化氢二氧六环(20ml,80mmol),室温下搅拌反应4-5小时,洗出白色固体,过滤记得产物,真空干燥后直接投入下一步反应。LC-MS(APCI):m/z=266.4(M+1) +
步骤4化合物28的合成
向反应瓶中加入化合物27(300mg,1.13mmol),5-氯-3-硝基吡唑并[1,5-α]嘧啶(224mg,1.13mmol),加入10ml乙醇溶解,加入DIPEA(584mg,4.52mmol),加热至80℃搅拌反应1小时,TLC检测反应完毕,降至室温,浓缩后硅胶柱层析纯化得到产物371mg,收率:77%。LC-MS(APCI):m/z=428.1(M+1) +
步骤5化合物29的合成
向反应瓶中加入化合物28(371mg,0.87mmol),3-酞酰亚胺基-1-丙炔(241mg,1.3mmol),双三苯基膦二氯化钯(30.5mg,0.043mmol)和碘化亚铜(16.4mg,0.086mmol),加入10ml无水四氢呋喃,加入DIPEA(337.3mg,2.61mmol),氮气保护室温下搅拌反应过夜,TLC检测反应完毕,浓缩后硅胶柱层析纯化得到产物274mg,收率:65%。LC-MS(APCI):m/z=485.5(M+1) +
步骤6化合物30的合成
向反应瓶中加入化合物29(274mg,0.57mmol),加入5ml甲醇溶解,再加入催化量的Pd/C,充氢气,室温下搅拌反应过夜,TLC检测反应完毕,过滤得到产物粗品258mg,直接投入到下一步反应。LC-MS(APCI):m/z=459.4(M+1) +
步骤7化合物31的合成
向反应瓶中加入化合物30(258mg,0.56mmol),加入10ml甲醇溶解,再加入过量水合肼,加热至回流反应4-6小时,TLC检测反应完毕,浓缩除去溶剂,加入水稀释,加入氯仿异丙醇(3∶1)萃取4-5次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化得到产物71.6mg。收率:39%,LC-MS(APCI):m/z=329.6(M+1) +
步骤8化合物T-5的合成
向反应瓶中加入化合物31(71mg,0.22mmol),加入10ml二氯甲烷溶剂,加入CDI(54mg,0.33mmol),室温下搅拌反应6小时,TLC监测反应完毕,浓缩除去溶剂,硅胶柱层析纯化得到产物51mg,收率:66%。LC-MS(APCI):m/z=355.1(M+1) +1H NMR(400MHz,DMSO-d 6)δ10.11(s,1H),8.56(s,1H),8.12(d,J=2.3Hz,1H),7.35(t,J=4.4Hz,1H),6.98(m,2H),6.52(d,J=2.3Hz,1H),4.01(m,1H),2.66(t,J=3.8Hz,2H),2.45(t,J=5.2Hz,2H),1.75(m,2H),1.21(d,J=5.5Hz,3H).
实施例6(13R)-11-氟-13-甲基-6,7,13,14-四氢-1,15-乙烯桥基吡唑并[4,3-f][1,4,8,10]苯并二氧杂 二氮杂十三烷-5(6H)-酮(化合物T-6)的制备。
Figure PCTCN2019082051-appb-000028
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000029
步骤1化合物32的合成
向反应瓶中加入5-氟-2-羟基苯乙酮(4.5g,29.2mmol),加入40ml无水THF溶解,氮气保护下0℃滴加R-CBS(1.46ml,1.46mmol),加毕,搅拌反应20分钟,再缓慢滴加硼烷四氢呋喃溶液(29.2ml,29.2mmol),加毕,低温下搅拌反应半小时,TLC检测反应完毕后,加入少量甲醇淬灭反应,浓缩除去溶剂,加入乙酸乙酯和水,分出有机相,水相用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化得到产物4.33g,收率:95%。
步骤2化合物33的合成
向反应瓶中加入化合物32(1.0g,6.41mmol),5-氯-3-硝基吡唑并[1,5-α]嘧啶(1.27g,6.41mmol),加入20ml乙醇溶解,加入NaH(2.48g,19.23mmol),加热至80℃搅拌反应1小时,TLC检测反应完毕,降至室温,浓缩后硅胶柱层析纯化得到产物1.79g,收率:88%。LC-MS(APCI):m/z=319.2(M+1) +
步骤3化合物34的合成
向反应瓶中加入化合物33(1.2g,3.77mmol)、3-羟基丙酸乙酯(0.67g,5.66mmol)和三苯基膦(1.48g,5.66mmol),氮气保护下加入5ml无水四氢呋喃,0℃下滴加DIAD(1.14g,5.66mmol),加毕,升至室温搅拌反应15小时,浓缩除去溶剂,硅胶柱层析纯化得到488.5mg产物,收率:31%。LC-MS(APCI):m/z=419.7(M+1) +
步骤4化合物35的合成
向反应瓶中加入化合物34(488mg,1.17mmol),用10ml甲醇溶解,加入一水合氢氧化锂(245mg,5.84mmol)的4ml水溶液,加热至50℃反应4-5小时,TLC监测反应完毕,降温至0℃,用1N稀盐酸调PH至弱酸性,乙酸乙酯萃取3-4次,合并有机相,无水硫酸钠干燥,过滤浓缩得到433.4mg产物,收率:95%。LC-MS(APCI):m/z=391.5(M+1) +
步骤5化合物36的合成
向反应瓶中加入化合物35(433mg,1.11mmol),用10ml甲醇溶解,加入催化量的Pd/C,充氢 气,室温下搅拌反应5-7小时,TLC监测反应完毕后,过滤除去催化剂,滤液浓缩后得到化合物36粗品,不需纯化直接投入到下一步反应。LC-MS(APCI):m/z=361.4(M+1) +
步骤6化合物T-6的合成
向反应瓶中加入化合物36(399mg,1.11mmol),用25ml无水DMF溶解,加入FDPP(511.8mg,1.33mmol)和DIPEA(717.3mg,5.55mmol),氮气保护下室温搅拌反应过夜,TLC监测反应完毕后,加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析纯化得到目标产物221mg,收率58%。LC-MS(APCI):m/z=343.5(M+1) +。LC-MS(APCI):m/z=342.5(M+1) +1H NMR(400MHz,DMSO-d 6)δ10.13(s,1H),8.59(s,1H),8.10(d,J=2.3Hz,1H),7.35(t,J=4.4Hz,1H),6.98(m,2H),6.52(d,J=2.3Hz,1H),4.25(t,J=3.3Hz,2H),4.12(m,1H),2.68(t,J=3.3Hz,2H),1.23(d,J=5.5Hz,3H).
实施例7 9-氟-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物9a),
(6R)-9-氟-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-1-A),
和(6S)-9-氟-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-1-B)的制备。
Figure PCTCN2019082051-appb-000030
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000031
步骤1化合物1a的合成
取2-溴-4-氟苯甲醚(18.95g,93mmol)溶于无水THF(100mL)中,-40℃下缓慢滴加异丙基氯化镁溶液(43.4mL,86.8mmol),滴毕自然升温至0℃搅拌1h,再于-40℃下缓慢滴加N-叔丁氧羰基-2-吡咯烷酮(11.46g,62.0mmol)的无水四氢呋喃(30mL)溶液,滴毕室温搅拌30min。反应液倒入100mL饱和氯化铵溶液中搅拌10min,静置分液,水相用40mL乙酸乙酯萃取三次,合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥。过滤浓缩,柱层析得17.68g浅黄色液体即为化合物1a,收率:61.1%。LC-MS(APCI):m/z=312.1(M+1) +
步骤2化合物2a的合成
取化合物1a(1.2g,3.85mmol)溶于甲苯(10mL)中,加入0.7mL浓盐酸,升温至65℃搅拌反应过夜,降至室温,用2M氢氧化钠调PH至14,继续搅拌1h,TLC检测反应完毕。分出有机相,水相用乙酸乙酯萃取3次,合并有机相,用饱和食盐水洗涤,浓缩,柱层析得到557mg黄色液体即为化合物2a,收率:75%。LC-MS(APCI):m/z=194.3(M+1) +
步骤3化合物3a的合成
取化合物2a(557mg,2.89mmol)溶于无水甲醇(10mL)中,加入Pd/C(50mg),室温氢化过夜。过滤,滤渣用20mL乙酸乙酯洗涤,滤液浓缩,得556mg无色油状液体即为化合物3a,收率:98.5%,直接投入下一步。LC-MS(APCI):m/z=196.3(M+1) +
步骤4化合物4a的合成
取化合物3a(722mg,3.7mmol)和5-氯-3-硝基吡唑并[1,5-a]嘧啶(733mg,3.7mmol)溶于无水乙醇(10mL)中,室温下加入DIPEA(N,N-二异丙基乙胺,1.91g,14.8mmol),加热回流30min。浓缩反应液,柱层析(PE/EA,30%~50%)得1.15g淡黄色固体粉末即为化合物4a,收率:81%。LC-MS(APCI):m/z=385.5(M+1) +
步骤5化合物5a的合成
取化合物4a(1.57g,4.1mmol)溶于10ml无水二氯甲烷中,0℃下滴加1M的三溴化硼溶液(20.5ml,20.5mmol),加毕升至室温反应1-2小时,TLC检测反应完毕后,冰浴下滴加少量水淬灭反应,分出有机相,水相用二氯甲烷萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析纯化得到目标产物880mg,收率:58%。LC-MS(APCI):m/z=371.6(M+1) +
步骤6化合物6a的合成
向反应瓶中加入化合物5a(116mg,0.315mmol)、3-羟基丙酸乙酯(55.8mg,0.473mmol)和三苯基膦(124.1mg,0.473mmol),氮气保护下加入5ml无水四氢呋喃,0℃下滴加DIAD(偶氮二甲酸二异丙酯,95.6mg,0.473mmol),加毕,升至室温搅拌反应15小时,浓缩除去溶剂,硅胶柱层析纯化得到53mg产物,收率:38%。LC-MS(APCI):m/z=444.1(M+1) +
步骤7化合物7a的合成
向反应瓶中加入化合物6a(204mg,0.46mmol),用8ml甲醇溶解,加入一水合氢氧化锂(96.6mg,2.3mmol)的4ml水溶液,加热至50℃反应4-5小时,TLC监测反应完毕,降温至0℃,用1N稀盐酸调PH至弱酸性,乙酸乙酯萃取3-4次,合并有机相,无水硫酸钠干燥,过滤浓缩得到181mg产物,收率:95%。LC-MS(APCI):m/z=416.5(M+1) +
步骤8化合物8a的合成
向反应瓶中加入化合物7a(186mg,0.45mmol),用5ml甲醇溶解,加入催化量的Pd/C,充氢气,室温下搅拌反应5-7小时,TLC监测反应完毕后,过滤除去催化剂,滤液浓缩后得到化合物8a粗品,不需纯化直接投入到下一步反应。LC-MS(APCI):m/z=386.4(M+1) +
步骤9化合物9a的合成
将上步所得产物溶于20ml无水DMF中,室温下加入FDPP(五氟苯基二苯基磷酸酯,240mg,0.62mmol)和DIPEA(336mg,2.6mmol),氮气保护下搅拌反应过夜,TLC检测反应完毕后,加入水稀释,乙酸乙酯萃取3-4次,合并有机相,用饱和食盐水洗涤,浓缩后柱层析纯化得到71mg类白色固体即为化合物9a,收率:43%。LC-MS(APCI):m/z=368.3(M+1) +1H NMR(400MHz,DMSO-d 6)δ10.11(s,1H),8.87(s,1H),8.04(d,J=2.3Hz,1H),7.52(s,1H),7.44(d,J=3.3Hz,1H),7.28(d,J=3.3Hz,1H),6.74(d,J=2.3Hz,1H),4.38(t,J=4.4Hz,2H),4.12(m,1H),3.61(dd,J=17.0,9.3Hz,2H),2.36(t,J=4.4Hz,2H),2.01-1.65(m,2H),1.22(d,J=4.5Hz,2H)。
步骤10化合物L-1-A和L-1-B的合成
手性制备色谱柱:CHIRALPAK IC(商品名),4.6mm×250mm(内径×长度),5μm(填料粒径)
柱温:30℃
流速:1.0mL/min
紫外检测波长:254nm
流动相:甲基叔丁醚∶甲醇=70∶30
在上述手性制备色谱柱和手性拆分条件下,消旋体化合物9a进行分离得到目标产物L-1-A(保留时间:20.33min,相对含量:43.4%)和L-1-B(保留时间:6.31min,相对含量:44.0%)。
实施例8(14S)-9-氟-14-甲基-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物13a),
(6R,14S)-9-氟-14-甲基-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-2-A),
和(6S,14S)-9-氟-14-甲基-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-2-B)的制备。
Figure PCTCN2019082051-appb-000032
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000033
步骤1化合物10a的合成
向反应瓶中加入化合物5a(216mg,0.63mmol)、(R)-3-羟基丁酸乙酯(125mg,0.946mmol)和三苯基膦(248.2mg,0.946mmol),氮气保护下加入10ml无水四氢呋喃,0℃下滴加DIAD(191.2mg,0.946mmol),加毕,升至室温搅拌反应15小时,浓缩除去溶剂,硅胶柱层析纯化得到77.7mg产物,收率:27%。LC-MS(APCI):m/z=458.1(M+1) +
步骤2化合物11a的合成
向反应瓶中加入化合物10a(210mg,0.46mmol)、用8ml甲醇溶解,加入一水合氢氧化锂(96.6mg,2.3mmol)的4ml水溶液,加热至50℃反应4-5小时,TLC监测反应完毕,降温至0℃,用1N稀盐酸调PH至弱酸性,乙酸乙酯萃取3-4次,合并有机相,无水硫酸钠干燥,过滤浓缩得到193mg产物,收率:98%。LC-MS(APCI):m/z=430.5(M+1) +
步骤3化合物12a的合成
向反应瓶中加入化合物11a(223mg,0.52mmol)、用8ml甲醇溶解,加入催化量的Pd/C,充氢 气,室温下搅拌反应5-7小时,TLC监测反应完毕后,过滤除去催化剂,滤液浓缩后得到化合物12a粗品,不需纯化直接投入到下一步反应。LC-MS(APCI):m/z=400.6(M+1) +
步骤4化合物13a的合成
向反应瓶中加入化合物12a(180mg,0.45mmol)、用15ml无水DMF溶解,加入FDPP(207.5mg,0.54mmol)和DIPEA(290.8mg,2.25mmol),氮气保护下室温搅拌反应过夜,TLC监测反应完毕后,加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析纯化得到目标产物103mg,收率60%。LC-MS(APCI):m/z=382.3(M+1) +1H NMR(400MHz,DMSO-d 6)δ10.08(s,1H),8.83(s,1H),8.04(d,J=2.3Hz,1H),7.52(s,1H),7.42(d,J=3.3Hz,1H),7.28(d,J=3.3Hz,1H),6.74(d,J=2.3Hz,1H),4.31(m,1H),4.12(m,1H),3.55(dd,J=17.0,9.3Hz,2H),2.32(t,J=4.4Hz,2H),1.95-1.61(m,2H),1.22(d,J=4.5Hz,2H),1.15(d,J=4.4Hz,3H)。
步骤5化合物L-2-A和L-2-B的合成
手性制备色谱柱:CHIRALPAK IC(商品名),4.6mm×250mm(内径×长度),5μm(填料粒径)
柱温:30℃
流速:1.0mL/min
紫外检测波长:254nm
流动相:甲基叔丁醚∶甲醇=70∶30
在上述手性制备色谱柱和手性拆分条件下,消旋体化合物13a进行分离得到目标产物L-2-A(保留时间:27.06min,相对含量:27.7%)和L-2-B(保留时间:7.02min,相对含量:67.8%)。
实施例9 9-氟-15,15-二甲基-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物18a)
(6R)-9-氟-15,15-二甲基-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-3-A),
和(6S)-9-氟-15,15-二甲基-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-3-B)的制备。
Figure PCTCN2019082051-appb-000034
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000035
步骤1化合物14a的合成
向反应瓶中加入异丁酸乙酯(2.5g,21.5mmol),氮气保护下加入40ml无水四氢呋喃,降温至-40℃,缓慢滴加LDA(11.8ml,23.6mmol),加毕升至室温搅拌反应半小时,再降温至-40℃,缓慢滴加二碘甲烷(5.76g,21.5mmol)的10ml无水四氢呋喃溶液,加毕,升至室温反应过夜,加水淬灭反应,分出有机相,水相用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得到4.62g产物,收率:84%。
步骤2化合物15a的合成
向反应瓶中加入化合物5a(326mg,0.95mmol),化合物14a(291mg,1.14mmol)和碳酸钾(525.2mg,3.8mmol),加入DMF,升温至80℃搅拌反应过夜,TLC监测反应完毕后,降至室温,加入过量水稀释,乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析纯化得到目标产物300mg,收率:67%,LC-MS(APCI):m/z=472.1(M+1) +
步骤3化合物16a的合成
向反应瓶中加入化合物15a(339mg,0.72mmol)、用10ml甲醇溶解,加入一水合氢氧化锂(151.5mg,3.5mmol)的3ml水溶液,加热至50℃反应4-5小时,TLC监测反应完毕,降温至0℃,用1N稀盐酸调PH至弱酸性,乙酸乙酯萃取3-4次,合并有机相,无水硫酸钠干燥,过滤浓缩得到296mg产物,收率:93%。LC-MS(APCI):m/z=444.3(M+1) +
步骤4化合物17a的合成
向反应瓶中加入化合物16a(297mg,0.67mmol)、用10ml甲醇溶解,加入催化量的Pd/C,充氢气,室温下搅拌反应5-7小时,TLC监测反应完毕后,过滤除去催化剂,滤液浓缩后得到化合物17a粗品,不需纯化直接投入到下一步反应。LC-MS(APCI):m/z=414.9(M+1) +
步骤5化合物18a的制备
向反应瓶中加入化合物17a(277mg,0.67mmol)、用20ml无水DMF溶解,加入FDPP(309mg,0.80mmol)和DIPEA(433mg,3.35mmol),氮气保护下室温搅拌反应过夜,TLC监测反应完毕后,加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后硅胶柱层析纯化得到目标产物111mg,收率42%。LC-MS(APCI):m/z=396.3(M+1) +1H NMR(400MHz,DMSO-d 6)δ10.10(s,1H),8.85(s,1H),8.04(d,J=2.3Hz,1H),7.52(s,1H),7.42(d,J=3.3Hz,1H),7.28(d,J=3.3Hz,1H),6.74(d,J=2.3Hz,1H),4.32(t,J=4.4Hz,2H),4.12(m,1H),3.61(dd,J=17.0,9.3Hz,2H), 2.01-1.65(m,2H),1.22(d,J=4.5Hz,2H),1.05(s,6H)。
步骤6化合物L-3-A和L-3-B的制备
手性制备色谱柱:CHIRALPAK IC(商品名),4.6mm×250mm(内径×长度),5μm(填料粒径)
柱温:30℃
流速:1.0mL/min
紫外检测波长:254nm
流动相:甲基叔丁醚∶甲醇=70∶30
在上述手性制备色谱柱和手性拆分条件下,消旋体化合物18a进行分离得到目标产物L-3-A(保留时间:24.2min,相对含量:33.1%)和L-3-B(保留时间:8.34min,相对含量:61.0%)。
实施例10 9-氟-15-甲基-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物23a),
(6R,15R)-9-氟-15-甲基-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-4-A),
(6R,15S)-9-氟-15-甲基-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-4-B),
(6S,15R)-9-氟-15-甲基-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-4-C),和
(6S,15S)-9-氟-15-甲基-13-氧杂-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-4-D)的制备。
Figure PCTCN2019082051-appb-000036
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000037
步骤1化合物19a的合成
取2-甲基-1,3-丙二醇(2.0g,22.2mmol)溶于二氯甲烷(30ml)中,冰浴下加入三乙胺(3.37g,33.3mmol),分批加入对甲苯磺酰氯(TsCl,4.23g,22.2mmol),保持温度不超过5℃,加毕,低温下搅拌反应2-3小时,TLC监测反应完毕后,加入20ml水,分出有机相,饱和食盐水洗涤,浓缩后柱层析纯化得到3.52g无色油状液体,即为化合物19a,收率:65%。LC-MS(APCI):m/z=245.6(M+1) +
步骤2化合物20a的合成
取化合物5a(800mg,2.33mmol)溶于DMF(20ml)中,加入碳酸钾(805mg,5.82mmol)和化合物19a(854mg,3.49mmol),升温至80℃搅拌反应4-6小时,TLC监测反应完毕后,降至室温,加入过量水稀释,乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化得到淡黄色固体691mg,收率:71.4%。LC-MS(APCI):m/z=416.1(M+1) +
步骤3化合物21a的合成
取化合物20a(606mg,1.46mmol)溶于二氯甲烷(20ml)中,冰浴下分批加入Dess-Martin氧化剂(928mg,2.19mmol),逐渐升至室温搅拌反应1小时,TLC监测反应完毕后,加入二氯甲烷稀释,用饱和碳酸氢钠溶液洗涤2-3次,有机相浓缩后直接投入到下一步反应。
将上步所得中间体溶于乙腈(20ml)中,加入磷酸二氢钠(876mg,7.3mmol),冰浴下加入亚氯酸钠(264mg,2.92mmol)的5ml水溶液,逐渐升至室温搅拌反应1小时,TLC监测反应完成后,加入过量水稀释,乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化得到552mg化合物21a,收率:88%。LC-MS(APCI):m/z=430.4(M+1) +
步骤4化合物22a的合成
取化合物21a(540mg,1.26mmol)溶于甲醇(15ml)中,加入催化量的Pd/C,充氢气球,室温下搅拌反应过夜,TLC监测反应完全后,过滤除去催化剂,滤液浓缩得化合物22a粗品478mg,直接投入到下一步反应,收率:95%。LC-MS(APCI):m/z=400.8(M+1) +
步骤5化合物23a的合成
取化合物22a(447mg,1.12mmol)溶于无水DMF(20ml)中,加入DIPEA(580mg,4.48mmol)和FDPP(516.4mg,1.34mmol),氮气保护下室温搅拌反应过夜,TLC监测反应完毕后,加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析得到淡黄色固体184mg,即为化合物23a。收率:43%。LC-MS(APCI):m/z=382.3(M+1) +1H NMR(500MHz,DMSO-d 6)δ8.58(s,1H),8.54(d,J=7.6Hz,1H),7.76(s,1H),7.09(d,J=7.2Hz,1H),7.01-6.87(m,2H),6.44(d,J=7.6Hz,1H),5.54(s,1H),4.35-4.25(m,1H),4.18(t,J=10.6Hz,1H),3.90(d,J=9.2Hz,1H),3.61(s,1H),2.96(s,1H),2.31(s,2H),1.97(d,J=7.0Hz,1H),1.83(d,J=5.0Hz,1H),1.23(d,J=7.1Hz,3H).
步骤6化合物L-4-A、L-4-B、L-4-C和L-4-D的合成
手性制备色谱柱:CHIRALPAK IC(商品名),4.6mm×250mm(内径×长度),5μm(填料粒径)
柱温:30℃
流速:1.0mL/min
紫外检测波长:254nm
流动相:甲基叔丁醚∶甲醇=70∶30
在上述手性制备色谱柱和手性拆分条件下,消旋体化合物23a进行分离得到目标产物L-4-A(保留时间:6.28min,相对含量:22.3%)、L-4-B(保留时间:18.46min,相对含量:22.4%)、L-4-C(保留时间:30.45min,相对含量:22.2%)和L-4-D(保留时间:37.26min,相对含量:22.4%)。
实施例11 9-氟-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷-1(24),7,9,11,18(25),19,22- 七烯-16-酮(化合物28a),
(6R)-9-氟-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷-1(24),7,9,11,18(25),19,22-七烯 -16-酮(化合物L-5-A),和
(6S)-9-氟-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷-1(24),7,9,11,18(25),19,22-七烯 -16-酮(化合物L-5-B)的制备。
Figure PCTCN2019082051-appb-000038
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000039
步骤1化合物24a的合成
取化合物5a(1.0g,2.91mmol)溶于二氯甲烷(30ml)中,加入三乙胺(589.5mg,5.82mmol),加入N,N-二三氟甲磺酰基苯胺(1.25g,3.49mmol),室温下,搅拌反应过夜,TLC监测反应完毕后,浓缩除去溶剂,硅胶柱层析纯化得到1.02g淡黄色固体,即为化合物24a,收率:74%。LC-MS(APCI):m/z=476.4(M+1) +
步骤2化合物25a的合成
取化合物24a(1.42g,3mmol)和3-丁炔-1-醇(315mg,4.5mmol)溶于二氧六环(30ml)中,氮气保护下加入双三苯基膦二氯化钯(21mg,0.03mmol)和碘化亚铜(57mg,0.3mmol),加热至100℃搅拌反应5小时,TLC检测反应完毕后,降至室温,加入水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化,得到854mg淡黄色固体即为化合物25a,收率:72%。LC-MS(APCI):m/z=396.8(M+1) +
步骤3化合物26a的合成
取化合物25a(577mg,1.46mmol)溶于二氯甲烷(20ml)中,冰浴下分批加入Dess-Martin氧化剂(928mg,2.19mmol),渐升至室温搅拌反应1小时,TLC监测反应完毕后,加入二氯甲烷稀释,用饱和碳酸氢钠溶液洗涤2-3次,有机相浓缩后直接投入到下一步反应。
将上步所得中间体溶于乙腈(20ml)中,加入磷酸二氢钠(876mg,7.3mmol),冰浴下加入亚氯酸钠(264mg,2.92mmol)的5ml水溶液,渐升至室温搅拌反应1小时,TLC监测反应完成后,加入过量水稀释,乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化得到466mg化合物26a,收率:78%。LC-MS(APCI):m/z=410.2(M+1) +
步骤4化合物27a的合成
取化合物26a(515mg,1.26mmol)溶于甲醇(15ml)中,加入催化量的Pd/C,充氢气球,室温下搅拌反应过夜,TLC监测反应完全后,过滤除去催化剂,滤液浓缩得化合物27a粗品468.6mg, 直接投入到下一步反应,收率:97%。LC-MS(APCI):m/z=384.6(M+1) +
步骤5化合物28a的合成
取化合物27a(429mg,1.12mmol)溶于无水DMF(20ml)中,加入DIPEA(580mg,4.48mmol)和FDPP(516.4mg,1.34mmol),氮气保护下室温搅拌反应过夜,TLC监测反应完毕后,加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析得到淡黄色固体160mg,即为化合物28a。收率:39%。LC-MS(APCI):m/z=366.1(M+1) +1H NMR(500MHz,DMSO-d 6)δ8.76(d,J=7.7Hz,1H),8.42(d,J=2.4Hz,1H),7.62(d,J=6.6Hz,1H),7.59(d,J=6.4Hz,1H),7.57(dd,J=9.9,2.4Hz,1H),7.22(d,J=6.4Hz,1H),6.60(d,J=7.7Hz,1H),5.36(t,J=6.5Hz,1H),4.05(dd,J=16.9,7.0Hz,2H),3.75(dd,J=14.0,8.6Hz,1H),3.67-3.51(m,1H),2.95(d,J=16.9Hz,1H),2.55(dd,J=14.0,10.9Hz,1H),2.42(dd,J=13.4,6.5Hz,2H),2.23(dd,J=12.0,6.2Hz,2H),2.12-1.96(m,1H),1.79-1.66(m,1H).
步骤6化合物L-5-A和L-5-B的合成
手性制备色谱柱:CHIRALPAK IC(商品名),4.6mm×250mm(内径×长度),5μm(填料粒径)
柱温:30℃
流速:1.0mL/min
紫外检测波长:254nm
流动相:甲基叔丁醚∶甲醇=70∶30
在上述手性制备色谱柱和手性拆分条件下,消旋体化合物28a进行分离得到目标产物L-5-A(保留时间:6.28min,相对含量:44.6%)和L-5-B(保留时间:18.46min,相对含量:44.8%)。
实施例12 9-氟-15-甲基-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物32a),
(6R,15R)-9-氟-15-甲基-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-6-A),
(6R,15S)-9-氟-15-甲基-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-6-B),
(6S,15R)-9-氟-15-甲基-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-6-C),和
(6S,15S)-9-氟-15-甲基-2,17,20,21,24-五氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-6-D)的制备。
Figure PCTCN2019082051-appb-000040
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000041
步骤1化合物29a的合成
取化合物24a(1.42g,3mmol)和2-甲基-3-丁炔-1-醇(364mg,4.5mmol)溶于二氧六环(30ml)中,氮气保护下加入双三苯基膦二氯化钯(21mg,0.03mmol)和碘化亚铜(57mg,0.3mmol),加热至100℃搅拌反应5小时,TLC检测反应完毕后,降至室温,加入水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化,得到773mg淡黄色固体即为化合物29a,收率:63%。LC-MS(APCI):m/z=410.1(M+1) +
步骤2化合物30a的合成
取化合物29a(597mg,1.46mmol)溶于二氯甲烷(20ml)中,冰浴下分批加入Dess-Martin氧化剂(928mg,2.19mmol),渐升至室温搅拌反应1小时,TLC监测反应完毕后,加入二氯甲烷稀释, 用饱和碳酸氢钠溶液洗涤2-3次,有机相浓缩后直接投入到下一步反应。
将上步所得中间体溶于乙腈(20ml)中,加入磷酸二氢钠(876mg,7.3mmol),冰浴下加入亚氯酸钠(264mg,2.92mmol)的5ml水溶液,渐升至室温搅拌反应1小时,TLC监测反应完成后,加入过量水稀释,乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化得到550mg化合物30a,收率:89%。LC-MS(APCI):m/z=424.2(M+1) +
步骤3化合物31a的合成
取化合物30a(533mg,1.26mmol)溶于甲醇(15ml)中,加入催化量的Pd/C,充氢气球,室温下搅拌反应过夜,TLC监测反应完全后,过滤除去催化剂,滤液浓缩得化合物31a粗品485.7mg,直接投入到下一步反应,收率:97%。LC-MS(APCI):m/z=398.9(M+1)+。
步骤4化合物32a的合成
取化合物31a(445mg,1.12mmol)溶于无水DMF(20ml)中,加入DIPEA(580mg,4.48mmol)和FDPP(516.4mg,1.34mmol),氮气保护下室温搅拌反应过夜,TLC监测反应完毕后,加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析得到淡黄色固体187mg,即为化合物32a。收率:44%。LC-MS(APCI):m/z=380.1(M+1) +1H NMR(500MHz,DMSO-d 6)δ8.76(d,J=7.7Hz,1H),8.42(d,J=2.4Hz,1H),7.62(d,J=6.6Hz,1H),7.59(d,J=6.4Hz,1H),7.57(dd,J=9.9,2.4Hz,1H),7.22(d,J=6.4Hz,1H),6.60(d,J=7.7Hz,1H),5.36(t,J=6.5Hz,1H),4.05(dd,J=16.9,7.0Hz,2H),3.75(dd,J=14.0,8.6Hz,1H),3.67-3.51(m,1H),2.95(d,J=16.9Hz,1H),2.42(dd,J=13.4,6.5Hz,2H),2.23(dd,J=12.0,6.2Hz,2H),2.12-1.96(m,1H),1.88(d,J=6.9Hz,3H),1.79-1.66(m,1H).
步骤5化合物L-6-A、L-6-B、L-6-C和L-4-D的合成
手性制备色谱柱:CHIRALPAK IC(商品名),4.6mm×250mm(内径×长度),5μm(填料粒径)
柱温:30℃
流速:1.0mL/min
紫外检测波长:254nm
流动相:甲基叔丁醚∶甲醇=70∶30
在上述手性制备色谱柱和手性拆分条件下,消旋体化合物32a进行分离得到目标产物L-6-A(保留时间:7.14min,相对含量:21.6%)、L-6-B(保留时间:15.69min,相对含量:22.8%)、L-6-C(保留时间:29.54min,相对含量:17.6%)和L-6-D(保留时间:34.26min,相对含量:16.9%)。
实施例13 9-氟-15-甲基-2,11,17,20,21,24-六氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物41a),
(6R,15R)-9-氟-15-甲基-2,11,17,20,21,24-六氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-7-A),
(6R,15S)-9-氟-15-甲基-2,11,17,20,21,24-六氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-7-B),
(6S,15R)-9-氟-15-甲基-2,11,17,20,21,24-六氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-7-C),和
(6S,15S)-9-氟-15-甲基-2,11,17,20,21,24-六氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-7-D)的制备。
Figure PCTCN2019082051-appb-000042
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000043
步骤1化合物33a的合成
取2-氯-3-溴-5-氟吡啶(19.53g,93mmol)溶于无水THF(100mL)中,-40℃下缓慢滴加异丙基氯化镁溶液(43.4mL,86.8mmol),滴毕自然升温至0℃搅拌1h,再于-40℃下缓慢滴加N-叔丁氧羰基-2-吡咯烷酮(11.46g,62.0mmol)的无水四氢呋喃(30mL)溶液,滴毕室温搅拌30min。反应液倒入100mL饱和氯化铵溶液中搅拌10min,静置分液,水相用40mL乙酸乙酯萃取三次,合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥。过滤浓缩,柱层析得19.5g浅黄色液体即为化合物33a,收率:66.3%。LC-MS(APCI):m/z=317.2(M+1) +
步骤2化合物34a的合成
取化合物33a(1.22g,3.85mmol)溶于二氯甲烷(10mL)中,加入2mL三氟乙酸,室温下搅拌1h,TLC检测反应完毕。加入饱和碳酸氢钠水溶液洗涤,分出有机相,水相用二氯甲烷萃取3次,合并有机相,用饱和食盐水洗涤,浓缩,即为化合物34a粗品,直接投入到下一步反应。LC-MS(APCI):m/z=199.3(M+1) +
步骤3化合物35a的合成
取化合物34a(764mg,3.85mmol)溶于无水甲醇(10mL)中,加入Pd/C(50mg),室温氢 化过夜。过滤,滤渣用20mL乙酸乙酯洗涤,滤液浓缩,得726mg无色油状液体即为化合物35a,收率:95%,直接投入下一步。LC-MS(APCI):m/z=201.3(M+1) +
步骤4化合物36a的合成
取化合物35a(742mg,3.7mmol)和5-氯-3-硝基吡唑并[1,5-a]嘧啶(733mg,3.7mmol)溶于无水乙醇(10mL)中,室温下加入DIPEA(1.91g,14.8mmol),加热回流30min。浓缩反应液,柱层析(PE/EA,30%~50%)得1.09g淡黄色固体粉末即为化合物36a,收率:81%。LC-MS(APCI):m/z=363.5(M+1) +
步骤5化合物37a的合成
取化合物36a(1.09g,3mmol)和4-((叔丁基二甲基硅基)氧代)-3-甲基丁炔(893mg,4.5mmol)溶于二氧六环(30ml)中,氮气保护下加入双三苯基膦二氯化钯(21mg,0.03mmol)和碘化亚铜(57mg,0.3mmol),加热至100℃搅拌反应5小时,TLC检测反应完毕后,降至室温,加入水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化,得到936mg淡黄色固体即为化合物37a,收率:59.5%。LC-MS(APCI):m/z=525.8(M+1) +
步骤6化合物38a的合成
取化合物37a(936mg,1.78mmol)溶于无水四氢呋喃(20ml)中,缓慢滴加四丁基氟化铵的1M四氢呋喃溶液(3.6ml,3.6mmol),加毕室温搅拌反应半小时,TLC检测反应完毕后,浓缩除去溶剂,硅胶柱层析纯化得到599mg化合物38a,收率:82%。LC-MS(APCI):m/z=411.1(M+1) +
步骤7化合物39a的合成
取化合物38a(599mg,1.46mmol)溶于二氯甲烷(20ml)中,冰浴下分批加入Dess-Martin氧化剂(928mg,2.19mmol),渐升至室温搅拌反应1小时,TLC监测反应完毕后,加入二氯甲烷稀释,用饱和碳酸氢钠溶液洗涤2-3次,有机相浓缩后直接投入到下一步反应。
将上步所得中间体溶于乙腈(20ml)中,加入磷酸二氢钠(876mg,7.3mmol),冰浴下加入亚氯酸钠(264mg,2.92mmol)的5ml水溶液,渐升至室温搅拌反应1小时,TLC监测反应完成后,加入过量水稀释,乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化得到533mg化合物39a,收率:86.1%。LC-MS(APCI):m/z=425.7(M+1) +
步骤8化合物40a的合成
取化合物39a(533mg,1.26mmol)溶于甲醇(15ml)中,加入催化量的Pd/C,充氢气球,室温下搅拌反应过夜,TLC监测反应完全后,过滤除去催化剂,滤液浓缩得化合物40a粗品446.7mg,直接投入到下一步反应,收率:89%。LC-MS(APCI):m/z=399.7(M+1) +
步骤9化合物41a的合成
取化合物40a(446.7mg,1.12mmol)溶于无水DMF(20ml)中,加入DIPEA(580mg,4.48mmol)和FDPP(516.4mg,1.34mmol),氮气保护下室温搅拌反应过夜,TLC监测反应完毕后,加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析得到淡黄色固体200.2mg,即为化合物41a。收率:47%。LC-MS(APCI):m/z=381.4(M+1) +1H NMR(500MHz,DMSO-d 6)δ8.70(d,J=7.7Hz,1H),8.39(d,J=2.4Hz,1H),8.01(s,1H),7.66(d,J=6.6Hz,1H),7.57(dd,J=9.9, 2.4Hz,1H),6.60(d,J=7.7Hz,1H),5.36(t,J=6.5Hz,1H),4.03(dd,J=16.9,7.0Hz,2H),3.75(dd,J=14.0,8.6Hz,1H),3.67-3.51(m,1H),2.91(d,J=16.9Hz,1H),2.54(dd,J=14.0,10.9Hz,1H),2.44(dd,J=13.4,6.5Hz,1H),2.23(dd,J=12.0,6.2Hz,2H),2.12-1.96(m,1H),1.79-1.66(m,1H),1.13(d,J=6.6Hz,3H).
步骤10化合物L-7-A、L-7-B、L-7-C和L-7-D的合成
手性制备色谱柱:CHIRALPAK IC(商品名),4.6mm×250mm(内径×长度),5μm(填料粒径)
柱温:30℃
流速:1.0mL/min
紫外检测波长:254nm
流动相:甲基叔丁醚∶甲醇=70∶30
在上述手性制备色谱柱和手性拆分条件下,消旋体化合物41a进行分离得到目标产物L-7-A(保留时间:4.77min,相对含量:30.21%)、L-7-B(保留时间:15.68min,相对含量:22.5%)、L-7-C(保留时间:26.31min,相对含量:10.66%)和L-7-D(保留时间:29.67min,相对含量:23.4%)。
实施例14 9-氟-2,11,17,20,21,24-六氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物45a),
(6R)-9-氟-2,11,17,20,21,24-六氮杂五环[16.5.2.0 2,6.0 7,12.0 21,,5]二十五烷-1(24),7,9,11,18(25),19,22-七 烯-16-酮(化合物L-8-A),
(6S)-9-氟-2,11,17,20,21,24-六氮杂五环[16.5.2.0 2,6.0 7,12.0 2125]二十五烷-1(24),7,9,11,18(25),19,22-七 烯-16-酮(化合物L-8-B)的制备。
Figure PCTCN2019082051-appb-000044
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000045
步骤1化合物42a的合成
取化合物36a(1.09g,3mmol)和3-丁炔-1-醇(315mg,4.5mmol)溶于二氧六环(30ml)中,氮气保护下加入双三苯基膦二氯化钯(21mg,0.03mmol)和碘化亚铜(57mg,0.3mmol),加热至100℃搅拌反应5小时,TLC检测反应完毕后,降至室温,加入水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化,得到845mg淡黄色固体即为化合物42a,收率:66%。LC-MS(APCI):m/z=397.1(M+1) +
步骤2化合物43a的合成
取化合物42a(578mg,1.46mmol)溶于二氯甲烷(20ml)中,冰浴下分批加入Dess-Martin氧化剂(928mg,2.19mmol),渐升至室温搅拌反应1小时,TLC监测反应完毕后,加入二氯甲烷稀释,用饱和碳酸氢钠溶液洗涤2-3次,有机相浓缩后直接投入到下一步反应。
将上步所得中间体溶于乙腈(20ml)中,加入磷酸二氢钠(876mg,7.3mmol),冰浴下加入亚氯酸钠(264mg,2.92mmol)的5ml水溶液,渐升至室温搅拌反应1小时,TLC监测反应完成后,加入过量水稀释,乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化得到485mg化合物43a,收率:81%。LC-MS(APCI):m/z=411.4(M+1) +
步骤3化合物44a的合成
取化合物43a(517mg,1.26mmol)溶于甲醇(15ml)中,加入催化量的Pd/C,充氢气球,室温下搅拌反应过夜,TLC监测反应完全后,过滤除去催化剂,滤液浓缩得化合物44a粗品450.4mg,直接投入到下一步反应,收率:93%。LC-MS(APCI):m/z=385.7(M+1) +
步骤4化合物45a的合成
取化合物44a(430.5mg,1.12mmol)溶于无水DMF(20ml)中,加入DIPEA(580mg,4.48mmol)和FDPP(516.4mg,1.34mmol),氮气保护下室温搅拌反应过夜,TLC监测反应完毕后,加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析得到淡黄色固体232.7mg,即为化合物45a。收率:56.7%。LC-MS(APCI):m/z=367.6(M+1) +1H NMR(500MHz,DMSO-d 6)δ8.76(d,J=7.7Hz,1H),8.42(d,J=2.4Hz,1H),8.01(s,1H),7.62(d,J=6.6Hz,1H),7.57(dd,J=9.9, 2.4Hz,1H),6.60(d,J=7.7Hz,1H),5.36(t,J=6.5Hz,1H),4.05(dd,J=16.9,7.0Hz,2H),3.75(dd,J=14.0,8.6Hz,1H),3.67-3.51(m,1H),2.91(d,J=16.9Hz,1H),2.54(dd,J=14.0,10.9Hz,1H),2.44(dd,J=13.4,6.5Hz,2H),2.23(dd,J=12.0,6.2Hz,2H),2.12-1.96(m,1H),1.79-1.66(m,1H).
步骤5化合物L-8-A和L-8-B合成
手性制备色谱柱:CHIRALPAK IC(商品名),4.6mm×250mm(内径×长度),5μm(填料粒径)
柱温:30℃
流速:1.0mL/min
紫外检测波长:254nm
流动相:甲基叔丁醚∶甲醇=70∶30
在上述手性制备色谱柱和手性拆分条件下,消旋体化合物45a进行分离得到目标产物L-8-A(保留时间:20.15min,相对含量:43.7%)和L-8-B(保留时间:8.25min,相对含量:44.0%)。
实施例15(4R)-9-氟-4-羟基-2,11,17,20,21,24-六氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物56a),
(4R,6R)-9-氟-4-羟基-2,11,17,20,21,24-六氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-9-A),
(4R,6S)-9-氟-4-羟基-2,11,17,20,21,24-六氮杂五环[16.5.2.0 2,6.0 7,12.0 21,25]二十五烷 -1(24),7,9,11,18(25),19,22-七烯-16-酮(化合物L-9-B)的制备。
Figure PCTCN2019082051-appb-000046
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000047
步骤1化合物46a的合成
取(R)-4-羟基吡咯烷-2-酮(5.0g,49.4mmol)溶于二氯甲烷(60ml)中,加入咪唑(4.04g,59.3mmol),冰浴下,加入三异丙基氯硅烷(TISCl,10.5g,54.3mmol),加毕室温下搅拌反应1-2小时,TLC监测反应完毕后,加入二氯甲烷稀释,用水洗涤3次,有机相浓缩后柱层析纯化得到10.3g无色油状液体,即为化合物46a,收率:81%。LC-MS(APCI):m/z=258.3(M+1) +
步骤2化合物47a的合成
取化合物46a(10.3g,40mmol)溶于二氯甲烷(100ml)中,加入三乙胺(6.07g,60mmol),冰浴下缓慢滴加二碳酸二叔丁酯(10.5g,48mmol),加毕升至室温反应3-4小时,TLC监测反应完全后,浓缩除去溶剂,柱层析纯化得到12.73g无色油状液体即为化合物47a,收率:89%。LC-MS(APCI):m/z=358.3(M+1) +
步骤3化合物48a的合成
取2-氯-3-溴-5-氟吡啶(19.53g,93mmol)溶于无水THF(100mL)中,-40℃下缓慢滴加异丙基氯化镁溶液(43.4mL,86.8mmol),滴毕自然升温至0℃搅拌1h,再于-40℃下缓慢滴加化合物47a(22.1g,62.0mmol)的无水四氢呋喃(30mL)溶液,滴毕室温搅拌30min。反应液倒入100mL饱和氯化铵溶液中搅拌10min,静置分液,水相用40mL乙酸乙酯萃取三次,合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥。过滤浓缩,柱层析得18.5g浅黄色液体即为化合物48a,收率:61.1%。LC-MS(APCI):m/z=490.2(M+1) +
步骤4化合物49a的合成
取化合物48a(1.88g,3.85mmol)溶于二氯甲烷(10mL)中,加入2mL三氟乙酸,室温下搅拌1h,TLC检测反应完毕。加入饱和碳酸氢钠水溶液洗涤,分出有机相,水相用二氯甲烷萃取3次, 合并有机相,用饱和食盐水洗涤,浓缩,即为化合物49a粗品,直接投入到下一步反应。LC-MS(APCI):m/z=372.3(M+1) +
步骤5化合物50a的合成
取化合物49a(1.43g,3.85mmol)溶于无水甲醇(20mL)中,加入Pd/C(100mg),室温氢化过夜。过滤,滤渣用20mL乙酸乙酯洗涤,滤液浓缩,得1.36g无色油状液体即为化合物50a,收率:95%,直接投入下一步。LC-MS(APCI):m/z=374.3(M+1) +
步骤6化合物51a的合成
取化合物50a(1.38g,3.7mmol)和5-氯-3-硝基吡唑并[1,5-a]嘧啶(733mg,3.7mmol)溶于无水乙醇(10mL)中,室温下加入DIPEA(1.91g,14.8mmol),加热回流30min。浓缩反应液,柱层析(PE/EA,30%~50%)得1.74g淡黄色固体粉末即为化合物51a,收率:88%。LC-MS(APCI):m/z=535.5(M+1) +
步骤7化合物52a的合成
取化合物51a(1.6g,3mmol)和3-羟基丁炔-1-醇(893mg,4.5mmol)溶于二氧六环(30ml)中,氮气保护下加入双三苯基膦二氯化钯(21mg,0.03mmol)和碘化亚铜(57mg,0.3mmol),加热至100℃搅拌反应5小时,TLC检测反应完毕后,降至室温,加入水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化,得到938mg淡黄色固体即为化合物52a,收率:55%。LC-MS(APCI):m/z=569.8(M+1) +
步骤8化合物53a的合成
取化合物52a(1.01g,1.78mmol)溶于无水四氢呋喃(20ml)中,缓慢滴加四丁基氟化铵的1M四氢呋喃溶液(3.6ml,3.6mmol),加毕室温搅拌反应半小时,TLC检测反应完毕后,浓缩除去溶剂,硅胶柱层析纯化得到565mg化合物53a,收率:77%。LC-MS(APCI):m/z=413.1(M+1) +
步骤9化合物54a的合成
取化合物53a(601mg,1.46mmol)溶于二氯甲烷(20ml)中,冰浴下分批加入Dess-Martin氧化剂(928mg,2.19mmol),渐升至室温搅拌反应1小时,TLC监测反应完毕后,加入二氯甲烷稀释,用饱和碳酸氢钠溶液洗涤2-3次,有机相浓缩后直接投入到下一步反应。
将上步所得中间体溶于乙腈(20ml)中,加入磷酸二氢钠(876mg,7.3mmol),冰浴下加入亚氯酸钠(264mg,2.92mmol)的5ml水溶液,渐升至室温搅拌反应1小时,TLC监测反应完成后,加入过量水稀释,乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析纯化得到553mg化合物54a,收率:89%。LC-MS(APCI):m/z=427.7(M+1) +
步骤10化合物55a的合成
取化合物54a(536mg,1.26mmol)溶于甲醇(15ml)中,加入催化量的Pd/C,充氢气球,室温下搅拌反应过夜,TLC监测反应完全后,过滤除去催化剂,滤液浓缩得化合物55a粗品449mg,直接投入到下一步反应,收率:89%。LC-MS(APCI):m/z=401.7(M+1) +
步骤11化合物56a的合成
取化合物55a(448mg,1.12mmol)溶于无水DMF(20ml)中,加入DIPEA(580mg,4.48mmol) 和FDPP(516.4mg,1.34mmol),氮气保护下室温搅拌反应过夜,TLC监测反应完毕后,加入过量水稀释,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤,浓缩后柱层析得到淡黄色固体248mg,即为化合物56a。收率:58%。LC-MS(APCI):m/z=383.4(M+1) +1H NMR(500MHz,DMSO-d 6)δ8.73(d,J=7.7Hz,1H),8.41(d,J=2.4Hz,1H),8.00(s,1H),7.62(d,J=6.6Hz,1H),7.57(dd,J=9.9,2.4Hz,1H),6.62(d,J=7.7Hz,1H),5.36(t,J=6.5Hz,1H),4.16(m,1H),4.05(dd,J=16.9,7.0Hz,2H),3.75(dd,J=14.0,8.6Hz,1H),3.64-3.51(m,1H),2.91(d,J=16.9Hz,1H),2.54(dd,J=14.0,10.9Hz,1H),2.44(dd,J=13.4,6.5Hz,2H),2.23(dd,J=12.0,6.2Hz,1H),2.12-1.96(m,1H),1.79-1.66(m,1H).
步骤12化合物L-9-A和L-9-B合成
手性制备色谱柱:CHIRALPAK IC(商品名),4.6mm×250mm(内径×长度),5μm(填料粒径)
柱温:30℃
流速:1.0mL/min
紫外检测波长:254nm
流动相:甲基叔丁醚∶甲醇=70∶30
在上述手性制备色谱柱和手性拆分条件下,消旋体化合物56a进行分离得到目标产物L-9-A(保留时间:14.26min,相对含量:45.0%)和L-9-B(保留时间:5.38min,相对含量:42.9%)。
实施例16 9-氟-13-氧杂-2,11,18,21,22,25-六氮杂五环[17.5.2.0 2,6.0 7,12.0 22,26]二十六烷 -1(25),7,9,11,19(26),20,23-七烯-17-酮(化合物64a),
(6R)-9-氟-13-氧杂-2,11,18,21,22,25-六氮杂五环[17.5.2.0 2,6.0 7,12.0 22,26]二十六烷 -1(25),7,9,11,19(26),20,23-七烯-17-酮(化合物L-10-A),
(6S)-9-氟-13-氧杂-2,11,18,21,22,25-六氮杂五环[17.5.2.0 2,6.0 7,12.0 22,26]二十六烷 -1(25),7,9,11,19(26),20,23-七烯-17-酮(化合物L-10-B)的制备。
Figure PCTCN2019082051-appb-000048
采用以下路线进行合成:
Figure PCTCN2019082051-appb-000049
步骤1化合物57a的合成
取2-甲氧基-3-溴-5-氟吡啶(19.16g,93mmol)溶于无水THF(100mL)中,-40℃下缓慢滴加异丙基氯化镁溶液(43.4mL,86.8mmol),滴毕自然升温至0℃搅拌1h,再于-40℃下缓慢滴加N-叔丁氧羰基-2-吡咯烷酮(11.46g,62.0mmol)的无水四氢呋喃(30mL)溶液,滴毕室温搅拌30min。反应液倒入100mL饱和氯化铵溶液中搅拌10min,静置分液,水相用40mL乙酸乙酯萃取三次,合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥。过滤浓缩,柱层析得16.2g浅黄色液体即为化合物57a,收率:56%。LC-MS(APCI):m/z=313.2(M+1) +
步骤2化合物58a的合成
取化合物57a(1.2g,3.85mmol)溶于二氯甲烷(10mL)中,加入2mL三氟乙酸,室温下搅拌1h,TLC检测反应完毕。加入饱和碳酸氢钠水溶液洗涤,分出有机相,水相用二氯甲烷萃取3次,合并有机相,用饱和食盐水洗涤,浓缩,即为化合物58a粗品,直接投入到下一步反应。LC-MS(APCI):m/z=195.2(M+1) +
步骤3化合物59a的合成
取化合物58a(747mg,3.85mmol)溶于无水甲醇(10mL)中,加入Pd/C(50mg),室温氢化过夜。过滤,滤渣用20mL乙酸乙酯洗涤,滤液浓缩,得731mg无色油状液体即为化合物59a,收率:97%,直接投入下一步。LC-MS(APCI):m/z=197.3(M+1) +
步骤4化合物60a的合成
取化合物59a(725mg,3.7mmol)和5-氯-3-硝基吡唑并[1,5-a]嘧啶(733mg,3.7mmol)溶于无水乙醇(10mL)中,室温下加入DIPEA(1.91g,14.8mmol),加热回流30min。浓缩反应液, 柱层析(PE/EA,30%~50%)得1.01g淡黄色固体粉末即为化合60a,收率:76%。LC-MS(APCI):m/z=359.5(M+1) +
步骤5化合物61a的合成
取化合物60a(1.01g,2.80mmol)溶于甲醇(20ml)中,加入催化量Pd/C,充氢气球,室温下搅拌反应4-5小时,TLC监测反应完成后,过滤除去催化剂,滤液浓缩得到目标产物粗品920mg,直接投入到下一步反应。LC-MS(APCI):m/z=329.6(M+1) +
步骤6化合物62a的合成
取化合物61a(920mg,2.8mmol)溶于无水DMF(15ml)中,加入4-氯丁酸(360mg,2.94mmol)和HATU(1.28g,3.36mmol),再加入DIPEA(1.08g,8.4mmol),室温下搅拌反应5小时,TLC监测反应完毕后,加入过量水稀释,乙酸乙酯萃取,合并有机相,用饱和食盐水洗涤,浓缩后柱层析纯化得到679mg淡黄色固体,即为化合物62a,收率:56%。LC-MS(APCI):m/z=433.6(M+1) +
步骤7化合物63a的合成
取化合物62a(679mg,1.57mmol),加入4N的氯化氢二氧六环溶液(10ml),加热至回流搅拌反应过夜,TLC监测反应完全后,浓缩除去溶剂,直接投入到下一步反应。LC-MS(APCI):m/z=419.2(M+1) +
步骤8化合物64a的合成
取化合物63a(655mg,1.57mmol)溶于无水DMF(20ml)中,加入碳酸钾(434mg,3.14mmol),加热至80℃搅拌反应过夜,TLC监测反应完全后,加入过量水,用乙酸乙酯萃取3-4次,合并有机相,饱和食盐水洗涤后,浓缩除去溶剂,硅胶柱层析纯化得到白色固体252mg,即为化合物64a,收率:41%。LC-MS(APCI):m/z=383.3(M+1) +1H NMR(500MHz,DMSO-d 6)δ8.76(d,J=7.7Hz,1H),8.42(d,J=2.4Hz,1H),8.01(s,1H),7.62(d,J=6.6Hz,1H),7.57(dd,J=9.9,2.4Hz,1H),6.60(d,J=7.7Hz,1H),5.36(t,J=6.5Hz,1H),4.05(dd,J=16.9,7.0Hz,2H),3.75(dd,J=14.0,8.6Hz,1H),3.67-3.51(m,1H),2.91(d,J=16.9Hz,1H),2.54(dd,J=14.0,10.9Hz,1H),2.44(dd,J=13.4,6.5Hz,2H),2.23(dd,J=12.0,6.2Hz,2H),2.12-1.96(m,1H),1.79-1.66(m,1H).
步骤9化合物L-10-A和L-10-B合成
手性制备色谱柱:CHIRALPAK IC(商品名),4.6mm×250mm(内径×长度),5μm(填料粒径)
柱温:30℃
流速:1.0mL/min
紫外检测波长:254nm
流动相:甲基叔丁醚∶甲醇=70∶30
在上述手性制备色谱柱和手性拆分条件下,消旋体化合物64a进行分离得到目标产物L-10-A(保留时间:27.54min,相对含量:49.4%)和L-10-B(保留时间:38.22min,相对含量:49.3%)。
生物活性测试。
生物实施例1:生物化学激酶分析
TRKA、TRKB、TRKC激酶抑制可通过HTRF(强荧光共振能量转移(High Fluorescence Resonance Energy Transfer))方法来测量。反应是在23℃下于384孔板中以20μL体积实施。将TRKA、TRKB或TRKC激酶与预先稀释配制的不同浓度的化合物(11个剂量,3倍浓度梯度,起始浓度300nM,2%DMSO终浓度)混合十分钟,每个浓度双复孔。加入对应底物及ATP,室温反应20分钟(其中设置阴阳性对照:阴性为空白对照,阳性为Entrectinib)。反应完毕加入检测试剂(HTRF Kinase TK试剂盒内的试剂),室温孵育30分钟后,通过PerkinElmer Envision酶标仪检测,测定在各浓度的本发明化合物存在下的酶活力,并计算不同浓度的化合物对酶活力的抑制活性,之后根据四参数方程,根据Graphpad 5.0软件对不同浓度化合物下酶活力的抑制活性进行拟合,计算出IC 50值。此分析中所测试化合物的数据呈现于表1中。
生物实施例2:KM12细胞增殖分析
采用CGT方法检测了本发明化合物对体外培养的肿瘤细胞的体外抗增殖活性。将KM12细胞株维持于含有10%胎牛血清和抗生素的RPMI-1640培养基中,收获对数期生长的细胞并种于96孔板中,置于5%二氧化碳培养箱中于37℃培养过夜。将受试化合物溶于DMSO后,3倍浓度梯度稀释,9个化合物浓度。将预先配制的不同浓度的化合物转移到细胞板中,每个浓度三复孔,继续培养72h。体系中DMSO终浓度为0.1%,受试化合物起始浓度300nM。
向细胞板中加入CellTiter-Glo试剂,室温孵育30分钟使发光信号稳定,通过PerkinElmer Envision酶标仪检测,测定在各浓度的本发明化合物对细胞增殖的抑制活性。根据Graphpad 5.0软件对不同浓度化合物下细胞增殖的抑制活性进行拟合,计算出IC 50值。此分析中所测试化合物的数据呈现于表1中。
表1:
Figure PCTCN2019082051-appb-000050
生物实施例3:Ba/F3细胞增殖分析
采用CGT方法检测了本发明化合物对体外培养的三株细胞的体外抗增殖活性。
将Ba/F3亲本细胞,Ba/F3 LMNA-NTRK1和Ba/F3 LMNA-NTRK1-G595R细胞分别维持于含有 10%胎牛血清和抗生素的RPMI-1640培养基中,收获对数期生长的细胞,并分别种于96孔板中,置于5%二氧化碳培养箱中于37℃培养过夜。其中Ba/F3亲本细胞中加入8ng/ml IL-3。将受试化合物溶于DMSO后,3.16倍浓度梯度稀释,9个化合物浓度。将预先配制的不同浓度的化合物转移到细胞板中,每个浓度三复孔,继续培养72h。体系中DMSO终浓度为0.1%,Ba/F3亲本细胞中受试化合物起始浓度10μM,Ba/F3 LMNA-NTRK1和Ba/F3 LMNA-NTRK1-G595R细胞中受试化合物起始浓度1μM。向细胞板中加入CellTiter-Glo试剂,室温孵育30分钟使发光信号稳定,通过PerkinElmer Envision酶标仪检测,测定在各浓度的本发明化合物对细胞增殖的抑制活性。根据Graphpad5.0软件对不同浓度化合物下细胞增殖的抑制活性进行拟合,计算出IC 50值。结果表明,本发明化合物对Ba/F3亲本细胞几乎没有抑制作用,但是对Ba/F3 LMNA-NTRK1和Ba/F3 LMNA-NTRK1-G595R细胞具有抑制作用。
生物实施例4:大鼠药代动力学实验
6只雄性Sprague-Dawley大鼠,7-8周龄,体重约210g,分成2组,每组3只,经静脉或口服单个剂量的化合物(静脉3mg/kg,口服10mg/kg),比较其药代动力学差异。
大鼠采用标准饲料饲养,给予水。试验前16小时开始禁食。药物用PEG400和二甲亚砜溶解。眼眶采血,采血的时间点为给药后0.083小时,0.25小时、0.5小时、1小时、2小时、4小时、6小时、8小时、12小时和24小时。
大鼠吸入乙醚后短暂麻醉,眼眶采集300μL血样于试管。试管内有30μL 1%肝素盐溶液。使用前,试管于60℃烘干过夜。在最后一个时间点血样采集完成之后,大鼠乙醚麻醉后处死。
血样采集后,立即温和地颠倒试管至少5次,保证混合充分后放置于冰上。血样在4℃ 5000rpm离心5分钟,将血浆与红细胞分离。用移液器吸出100μL血浆到干净的塑料离心管中,标明化合物的名称和时间点。血浆在进行分析前保存在-80℃。用LC-MS/MS测定血浆中本发明化合物的浓度。药代动力学参数基于每只动物在不同时间点的血药浓度进行计算。
实验表明,本发明化合物在动物体内具有更好的药代动力学性质,因此具有更好的药效学和治疗效果。代表性实施例化合物的大鼠药代动力学实验结果归纳于下表2和表3中。
表2:
Figure PCTCN2019082051-appb-000051
表3:
Figure PCTCN2019082051-appb-000052
以上内容是结合具体的优选实施方式对本发明所作的进一步详细说明,不能认定本发明的具体实施只局限于这些说明。对于本发明所属技术领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干简单推演或替换,都应当视为属于本发明的保护范围。

Claims (12)

  1. 式(I)化合物:
    Figure PCTCN2019082051-appb-100001
    其中,
    A 1选自CR 1或N;
    A 2选自CR 2或N;
    A 3选自CR 3或N;
    A 4选自CR 4或N;
    其中R 1、R 2、R 3和R 4独立地选自H、D、卤素、-CN、-NO 2、-OR a、-SR a、-NR bR c、-C(O)R a、-C(O)OR a、-C(O)NR bR c、-OC(O)R a、-NR bC(O)R a、-S(O)R a、-S(O) 2R a、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    其中各个R a、R b和R c独立地选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    L 1选自C(R 1a)(R 2a)、O、S、N(R 1a)、C(O)、S(O)或S(O) 2
    L 2选自C(R 1b)(R 2b)、O、S、N(R 1b)、C(O)、S(O)或S(O) 2
    X选自O、S、N(R 1c)或C(R 1c)(R 2c);
    Y选自O、S、N(R 1d)或C(R 1d)(R 2d);
    W选自O、S、N(R 1e)或C(R 1e)(R 2e);
    R选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    m选自1、2、3、4或5;
    n选自1、2或3;
    其中,
    各个R 1a和R 2a独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1a、R 2a和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1b和R 2b独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、 -C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1b、R 2b和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1c和R 2c独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1c、R 2c和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1d和R 2d独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1d、R 2d和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1e和R 2e独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1e、R 2e和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    -X-(L 1) m-Y-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    -(L 2) n-W-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
  2. 权利要求1的化合物,其为式(III-1)或(III-2):
    Figure PCTCN2019082051-appb-100002
    其中,
    A 1选自CR 1或N;
    A 4选自CR 4或N;
    其中R 1、R 3和R 4独立地选自H、D、卤素、-CN、-NO 2、-OR a、-SR a、-NR bR c、C 1-6烷基、C 1-6 卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    其中各个R a、Rb和R c独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    X选自O或C(R 1c)(R 2c);
    Y选自N(R 1d)或C(R 1d)(R 2d);
    W选自O或NH;
    m选自1、2或3;
    各个R 1a和R 2a独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1a、R 2a和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    R 1b选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    其中,
    各个R 1c和R 2c独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1c、R 2c和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1d和R 2d独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1d、R 2d和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    -X-(C(R 1a)(R 2a)) m-Y-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
  3. 权利要求1或2的化合物,其为式(IV-1)或(IV-2):
    Figure PCTCN2019082051-appb-100003
    其中,
    R 3选自H、D、卤素、-CN、-NO 2、-OR a、-SR a或-NR bR c
    其中各个R a、R b和R c独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    X选自O或C(R 1c)(R 2c);
    Y选自NH、CH 2或C(Me)(Me);其中上述基团任选地被一个或多个D取代,直至完全氘代;
    m选自1、2或3;
    各个R 1a和R 2a独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1a、R 2a和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    R 1b选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    其中,各个R 1c和R 2c独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1c、R 2c和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    优选地,
    R 3选自H、D、卤素、-CN或-NO 2
    X为O;
    Y选自NH、CH 2或C(Me)(Me);其中上述基团任选地被一个或多个D取代,直至完全氘代;
    m选自1、2或3;
    R 1a和R 2a独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    R 1b选自C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    优选地,
    R 3选自H、D、卤素、-CN或-NO 2
    X为O;
    Y选自CH 2或C(Me)(Me);其中上述基团任选地被一个或多个D取代,直至完全氘代;
    m选自1、2或3;
    R 1a和R 2a独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    R 1b选自C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
  4. 权利要求1-3中任一项的化合物,其为式(VI-1)或(VI-2):
    Figure PCTCN2019082051-appb-100004
    其中,
    R 3选自H、D、卤素、-CN或-NO 2
    R 1a和R 2a独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    R 1b选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
  5. 权利要求1的化合物,其为式(I’)化合物:
    Figure PCTCN2019082051-appb-100005
    其中,
    A 1选自CR 1或N;
    A 2选自CR 2或N;
    A 3选自CR 3或N;
    A 4选自CR 4或N;
    其中R 1、R 2、R 3和R 4独立地选自H、D、卤素、-CN、-NO 2、-OR a、-SR a、-NR bR c、-C(O)R a、 -C(O)OR a、-C(O)NR bR c、-OC(O)R a、-NR bC(O)R a、-S(O)R a、-S(O) 2R a、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    其中各个R a、R b和R c独立地选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    L 1选自C(R 1a)(R 2a)、O、S、N(R 1a)、C(O)、S(O)或S(O) 2
    X选自O、S、N(R 1c)或C(R 1c)(R 2c);
    Y选自O、S、N(R 1d)或C(R 1d)(R 2d);
    L 3选自C(R 1f)(R 2f)、O、S、N(R 1f)、C(O)、S(O)或S(O) 2
    L 4选自C(R 1g)(R 2g)、O、S、N(R 1g)、C(O)、S(O)或S(O) 2
    L 5选自C(R 1h)(R 2h)、O、S、N(R 1h)、C(O)、S(O)或S(O) 2
    R选自H、D、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    m选自1、2、3、4或5;
    n选自1、2或3;
    其中,
    各个R 1a和R 2a独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1a、R 2a和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1c和R 2c独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1c、R 2c和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1d和R 2d独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1d、R 2d和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1f和R 2f独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1f、R 2f和它们所连接的碳原子一起形成C 3-7 环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1g和R 2g独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1g、R 2g和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1h和R 2h独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1h、R 2h和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    -X-(L 1) m-Y-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
  6. 权利要求5的化合物,其为式(III’-1)或(III’-2):
    Figure PCTCN2019082051-appb-100006
    其中,
    A 1选自CR 1或N;
    其中R 1和R 3独立地选自H、D、卤素、-CN、-NO 2、-OR a、-SR a、-NR bR c、C 1-6烷基、C 1-6卤代烷基、C 3-7环烷基、3至7元杂环基、C 6-10芳基或5至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    其中各个R a、R b和R c独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    X选自O或C(R 1c)(R 2c);
    Y选自CH 2、CH(R 1d)或C(R 1d)(R 2d);其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1a和R 2a独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1a、R 2a和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1c和R 2c独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1c、R 2c和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1d和R 2d独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1d、R 2d和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    各个R 1f和R 2f独立地选自H、D、卤素、-C 0-6亚烷基-CN、-C 0-6亚烷基-OR a、-C 0-6亚烷基-SR a、-C 0-6亚烷基-NR bR c、-C 0-6亚烷基-C(O)R a、-C 0-6亚烷基-C(O)OR a、-C 0-6亚烷基-C(O)NR bR c、C 1-6烷基、C 1-6卤代烷基、C 2-6烯基、C 2-6炔基、-C 0-6亚烷基-C 3-6环烷基、-C 0-6亚烷基-3至7元杂环基、-C 0-6亚烷基-C 6-10芳基或-C 0-6亚烷基-5至10元杂芳基;或者,R 1f、R 2f和它们所连接的碳原子一起形成C 3-7环烷基或3至7元杂环基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    m选自1、2或3;
    -X-(C(R 1a)(R 2a)) m-Y-中不同原子上存在的取代基可以连接形成C 3-10环烷基、3至10元杂环基、C 6-14芳基或3至10元杂芳基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
  7. 权利要求5或6的化合物,其为式(V’-1)或(V’-2):
    Figure PCTCN2019082051-appb-100007
    其中,
    R 3选自H、D、卤素、-CN或-NO 2
    R 1a和R 2a各自独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    优选地,
    R 3选自H、D、卤素、-CN或-NO 2
    R 1a和R 2a各自独立地选自H、D、C 1-6烷基或C 1-6卤代烷基;且R 1a和R 2a不同时为H或D;其中上述基团任选地被一个或多个D取代,直至完全氘代;
    或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
  8. 化合物,其选自:
    Figure PCTCN2019082051-appb-100008
    Figure PCTCN2019082051-appb-100009
    Figure PCTCN2019082051-appb-100010
    或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物。
  9. 药物组合物,其含有权利要求1-8中任一项的化合物,或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物,和药学上可接受的赋形剂。
  10. 权利要求1-8中任一项的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物在制备用于治疗癌症、疼痛、神经疾病、自身免疫疾病和炎症的药物中的用途。
  11. 一种在受试者中治疗癌症、疼痛、神经疾病、自身免疫疾病和炎症的方法,包括给药有效量的权利要求1-8中任一项所述的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物,或权利要求9的药物组合物。
  12. 权利要求1-8中任一项所述的化合物或其药学上可接受的盐、对映异构体、非对映异构体、外消旋体、溶剂合物、水合物、多晶型、前药或活性代谢物,其用于治疗癌症、疼痛、神经疾病、自身免疫疾病和炎症。
PCT/CN2019/082051 2018-04-16 2019-04-10 用于抑制蛋白激酶活性的二(杂)芳基大环化合物 WO2019201131A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP19787700.4A EP3770161B1 (en) 2018-04-16 2019-04-10 Di(hetero)aryl macrocyclic compound for inhibiting protein kinase activity
JP2020556951A JP7092405B2 (ja) 2018-04-16 2019-04-10 キナーゼ活性を阻害するためのジ(ヘテロ)アリール大環状化合物
US17/047,877 US11358973B2 (en) 2018-04-16 2019-04-10 Di(hetero)aryl macrocyclic compound for inhibiting protein kinase activity

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201810337944.6 2018-04-16
CN201810337944 2018-04-16
CN201810338480.0 2018-04-16
CN201810338480 2018-04-16

Publications (1)

Publication Number Publication Date
WO2019201131A1 true WO2019201131A1 (zh) 2019-10-24

Family

ID=68240438

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/082051 WO2019201131A1 (zh) 2018-04-16 2019-04-10 用于抑制蛋白激酶活性的二(杂)芳基大环化合物

Country Status (5)

Country Link
US (1) US11358973B2 (zh)
EP (1) EP3770161B1 (zh)
JP (1) JP7092405B2 (zh)
CN (2) CN113956269A (zh)
WO (1) WO2019201131A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021042890A1 (zh) * 2019-09-04 2021-03-11 罗欣药业(上海)有限公司 杂环化合物及其作为Trk激酶抑制剂的应用
JP2021529182A (ja) * 2018-06-25 2021-10-28 北京諾誠健華医薬科技有限公司 Trk阻害剤としてのヘテロ環化合物

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112110938B (zh) * 2019-06-21 2021-11-09 成都海博为药业有限公司 一种作为蛋白质激酶抑制剂的化合物及其制备方法和用途
CN112979654B (zh) * 2019-12-16 2024-03-19 赛诺哈勃药业(成都)有限公司 杂芳基稠环化合物、其制备方法及应用
CN114230553B (zh) * 2020-09-09 2023-05-05 凯特立斯(深圳)科技有限公司 一种左旋烟碱的不对称合成方法
CN114907315B (zh) * 2022-05-17 2023-09-12 重庆医科大学 Selitrectinib中间体的合成方法
CN115746023A (zh) * 2022-10-27 2023-03-07 复旦大学 一种作为蛋白激酶抑制剂的含吲唑结构的杂环大环化合物及其制备方法

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
CN102971322A (zh) * 2010-05-20 2013-03-13 阵列生物制药公司 作为trk激酶抑制剂的大环化合物
CN106170289A (zh) * 2014-01-24 2016-11-30 Tp生物医药公司 作为蛋白质激酶的调节剂的二芳基巨环
WO2017007759A1 (en) * 2015-07-06 2017-01-12 Tp Therapeutics, Inc. Diaryl macrocycle polymorph
WO2017015367A1 (en) * 2015-07-21 2017-01-26 Tp Therapeutics, Inc. Chiral diaryl macrocycles and uses thereof
WO2017075107A1 (en) * 2015-10-26 2017-05-04 Nanda Nisha Point mutations in trk inhibitor-resistant cancer and methods relating to the same
WO2017148325A1 (zh) * 2016-03-03 2017-09-08 深圳市塔吉瑞生物医药有限公司 一种大环化合物及包含该化合物的组合物

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2016510793A (ja) * 2013-03-15 2016-04-11 イプセン ファルマ ソシエテ パール アクシオン サンプリフィエIpsen Pharma S.A.S. 大環状lrrk2キナーゼ阻害剤
MX2015012526A (es) * 2013-03-15 2016-04-26 Oncodesign Sa Inhibodores macrociclicos de cinasa rip2.
CN110945000B (zh) 2017-08-23 2021-09-03 正大天晴药业集团股份有限公司 含有氨基吡唑并嘧啶的大环化合物及其药物组合物和用途
AU2018364938B2 (en) * 2017-11-10 2021-11-11 Angex Pharmaceutical, Inc. Macrocyclic compounds as TRK kinase inhibitors and uses thereof
WO2019149131A1 (zh) 2018-01-30 2019-08-08 上海吉倍生物技术有限公司 具有大环分子结构的化合物及其用途
CN110156813B (zh) 2018-02-13 2023-07-25 北京诺诚健华医药科技有限公司 作为trk抑制剂的杂环化合物
CN110627812B (zh) 2018-06-25 2022-10-11 北京诺诚健华医药科技有限公司 作为trk抑制剂的杂环化合物

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
CN102971322A (zh) * 2010-05-20 2013-03-13 阵列生物制药公司 作为trk激酶抑制剂的大环化合物
CN106170289A (zh) * 2014-01-24 2016-11-30 Tp生物医药公司 作为蛋白质激酶的调节剂的二芳基巨环
WO2017007759A1 (en) * 2015-07-06 2017-01-12 Tp Therapeutics, Inc. Diaryl macrocycle polymorph
WO2017015367A1 (en) * 2015-07-21 2017-01-26 Tp Therapeutics, Inc. Chiral diaryl macrocycles and uses thereof
WO2017075107A1 (en) * 2015-10-26 2017-05-04 Nanda Nisha Point mutations in trk inhibitor-resistant cancer and methods relating to the same
WO2017148325A1 (zh) * 2016-03-03 2017-09-08 深圳市塔吉瑞生物医药有限公司 一种大环化合物及包含该化合物的组合物

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
BERGE, J. PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
D. FLEISHERS. RAMONH. BARBRA: "Improved oral drug delivery: solubility limitations overcome by the use of prodrugs", ADVANCED DRUG DELIVERY REVIEWS, vol. 19, no. 2, 1996, pages 115 - 130
See also references of EP3770161A4
T. HIGUCHIV. STELLA: "Prodrugs as Novel Delivery Systems, A.C.S. Symposium Series", vol. 14, 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS, article "Bioreversible Carriers in Drug Design"

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2021529182A (ja) * 2018-06-25 2021-10-28 北京諾誠健華医薬科技有限公司 Trk阻害剤としてのヘテロ環化合物
JP7372686B2 (ja) 2018-06-25 2023-11-01 北京諾誠健華医薬科技有限公司 Trk阻害剤としてのヘテロ環化合物
US11878987B2 (en) 2018-06-25 2024-01-23 Beijing Innocare Pharma Tech Co., Ltd. Heterocyclic compound as TRK inhibitor
WO2021042890A1 (zh) * 2019-09-04 2021-03-11 罗欣药业(上海)有限公司 杂环化合物及其作为Trk激酶抑制剂的应用

Also Published As

Publication number Publication date
US11358973B2 (en) 2022-06-14
EP3770161A1 (en) 2021-01-27
EP3770161B1 (en) 2022-08-24
CN110386944A (zh) 2019-10-29
US20210171542A1 (en) 2021-06-10
JP2021521239A (ja) 2021-08-26
EP3770161A4 (en) 2021-04-28
JP7092405B2 (ja) 2022-06-28
CN110386944B (zh) 2021-10-29
CN113956269A (zh) 2022-01-21

Similar Documents

Publication Publication Date Title
WO2019201131A1 (zh) 用于抑制蛋白激酶活性的二(杂)芳基大环化合物
WO2021219090A1 (zh) 喹喔啉二酮衍生物作为kras g12c突变蛋白的不可逆抑制剂
WO2018177403A1 (zh) 1H-咪唑[4,5-h]喹唑啉类化合物作为蛋白激酶抑制剂
WO2021057877A1 (zh) 取代的芳香稠合环衍生物及其组合物及用途
CN108699039B (zh) 用于抑制蛋白激酶活性的(杂)芳基酰胺类化合物
EP3715343B1 (en) Diphenylaminopyrimidine compound for inhibiting kinase activity
CN109851638B (zh) 取代的二氨基嘧啶化合物
WO2023134266A1 (zh) 2-哌啶基或2-吡唑基取代的嘧啶化合物作为egfr抑制剂
EP3715350B1 (en) Arylphosphine oxides for inhibiting kinase activity
WO2018214846A1 (zh) 咪唑并[1',2':1,6]吡啶并[2,3-d]嘧啶类化合物作为蛋白激酶抑制剂
CN109111439B (zh) 一种酰胺类化合物及包含该化合物的组合物及其用途
WO2019228330A1 (zh) 取代的苯并[d]咪唑类化合物及其药物组合物
WO2021057796A1 (zh) 取代的稠合三环衍生物及其组合物及用途
JP7323218B2 (ja) 置換された縮合芳香環誘導体、その組成物、およびそれらの使用
WO2021164697A1 (zh) 取代的酰胺衍生物及其组合物及用途
CN114874189B (zh) 取代的杂芳基衍生物及其组合物及用途
WO2021185348A1 (zh) 取代的丙烯酰胺衍生物及其组合物及用途
EP3885346A1 (en) Aminopyrimidine compound used for inhibiting activity of protein kinase
WO2023216910A1 (zh) 取代的双环杂芳基化合物作为usp1抑制剂
TW202334095A (zh) Sos1抑制劑
JP2023538774A (ja) 新規な選択的flt3阻害剤としての1h-イミダゾ[4,5-h]キナゾリン化合物

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: 19787700

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2020556951

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019787700

Country of ref document: EP

Effective date: 20201023