WO2018019204A1 - 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物 - Google Patents

用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物 Download PDF

Info

Publication number
WO2018019204A1
WO2018019204A1 PCT/CN2017/094102 CN2017094102W WO2018019204A1 WO 2018019204 A1 WO2018019204 A1 WO 2018019204A1 CN 2017094102 W CN2017094102 W CN 2017094102W WO 2018019204 A1 WO2018019204 A1 WO 2018019204A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
group
alkyl
mmol
compound
Prior art date
Application number
PCT/CN2017/094102
Other languages
English (en)
French (fr)
Inventor
王义汉
任兴业
金剑
李焕银
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 JP2019503547A priority Critical patent/JP6740452B2/ja
Priority to EP17833507.1A priority patent/EP3492462B1/en
Priority to CN201780009980.3A priority patent/CN108602802B/zh
Publication of WO2018019204A1 publication Critical patent/WO2018019204A1/zh
Priority to US16/256,203 priority patent/US11111233B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the invention belongs to the field of medicine.
  • the present invention relates to aminopyrimidine compounds having an inhibitory effect on protein tyrosine kinases, pharmaceutical compositions containing the same, and methods for their preparation and use.
  • the epidermal growth factor receptor (ie, EGFR, ErbB-1 or HER1) is a member of the ErbB receptor family, and the ErbB receptor family includes four closely related receptor tyrosine kinase members: EGFR (ErbB-1), HER2/c-neu (ErbB-2), Her3 (ErbB-3) and Her4 (ErbB-4).
  • EGFR is a cell surface receptor for members of the epidermal growth factor family (EGF family) of extracellular protein ligands. Mutations that affect EGFR expression or activity may result in cancer. It has been reported that EGFR is in an dysregulated state in most solid tumors such as lung cancer, breast cancer and brain tumors. It is estimated that 30% of epithelial cancers are associated with mutations, amplification or disorders of EGFR or family members.
  • EGFR-TKI EGFR kinase inhibitors
  • gefitinib and erlotinib EGFR kinase inhibitors
  • EGFR-TKI was subsequently found to treat non-small cells.
  • Primary or secondary drug resistance in lung cancer is a new challenge for us in the treatment of advanced non-small cell lung cancer. It is necessary to carry out new explorations and find countermeasures.
  • Third-generation and subsequent EGFR inhibitors include compounds such as AZD9291 and CO-1686, which irreversibly inhibit EGFR and are more efficient in patients with T790M-resistant mutations, but still have an inhibitory effect on wild-type EGFR.
  • the present invention provides a novel aminopyrimidine compound and a composition comprising the same, which has better EGFR kinase inhibitory activity and high selectivity for the resistant mutations T790M, L858R and both, It can be used to treat, prevent, and alleviate EGFR kinase-mediated diseases.
  • R 1 and R 2 are independently selected from H, halogen, -CN, -NO 2 , optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally substituted C 1 -C 6 alkoxy, optionally substituted C 1 -C 6 haloalkoxy, optionally substituted C 1 -C 6 alkylthio, optionally substituted C 1 -C 6 alkylamino or optionally substituted C 1- C 6 alkanoyl;
  • L is selected from the group consisting of a bond, NR 7 , O, C(R 7 ) 2 or S; wherein each R 7 is independently selected from H, optionally substituted C 1 -C 6 alkyl, for example by C 1 -C 6 alkoxy Substituted C 1 -C 6 alkyl, or optionally substituted C 1 -C 6 alkoxy;
  • Y is H, or Y is selected from the following structures:
  • R 3 , R 4 and R 5 are independently selected from H, halogen, -CN, optionally substituted C 1 -C 6 alkyl or optionally substituted C 1 -C 6 haloalkyl;
  • R 6 is selected from H, C 1 -C 6 alkyl, C 2 -C 6 alkynyl or 3 to 8 membered heterocyclic, wherein the above group is optionally substituted with from 1 to 3 substituents: halogen, -OH , -CN, -NO 2 , optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 alkoxy, -C(O)R 8 or NR 8 R 9 ; wherein R 8 and R 9 are each independently selected from H, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 alkoxy, optionally substituted C 1 -C 6 haloalkyl or optionally substituted a C 3 -C 6 carbocyclic group, or R 8 and R 9 together with the nitrogen atom to which they are attached form an optionally substituted 4 to 6 membered heterocyclic group, for example, substituted by C 1 -C 6 alkyl;
  • W is selected from the group consisting of a bond, C(R 10 ) 2 , NR 10 , O or S, wherein R 10 is selected from H, halogen, -CN, -NO 2 , optionally substituted C 1 -C 6 alkyl, optionally substituted a C 1 -C 6 haloalkyl group or an optionally substituted C 1 -C 6 alkoxy group, as long as it is chemically acceptable;
  • n 0, 1 or 2;
  • n 0, 1 or 2;
  • Ring A is selected from the following structures:
  • each X 1 is independently selected from C(R 11 ) 2 , NR 11 , O or S
  • each of X 2 and X 3 is independently selected from C(R 11 ) 2 or NR 11 , wherein X 1 and X 2 are The location to connect with the mother core;
  • Ring B is selected from a 5-membered carbocyclic group, a 5-membered heterocyclic group or a 5-membered heteroaryl group, which is optionally substituted with from 1 to 3 R 11 substituents;
  • R 11 is independently selected from H, halogen, -CN, -NO 2 , -OH, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally substituted C 2 a -C 6 alkenyl group, an optionally substituted C 2 -C 6 alkynyl group, an optionally substituted C 1 -C 6 alkoxy group or an optionally substituted C 1 -C 6 haloalkoxy group, as long as it is chemically acceptable; Wherein two adjacent R 11 may together form an optionally substituted C 5 -C 8 carbocyclic group, an optionally substituted 5 to 8 membered heterocyclic group, an optionally substituted C 6 -C 14 aryl group or any a substituted 5 to 10 membered heteroaryl group;
  • a preferred embodiment of this aspect is the above compound which is a compound of formula (II):
  • R 1 , R 2 , L, R 6 , W, A, m and n are as defined above,
  • a preferred embodiment of this aspect is the above compound which is a compound of formula (III):
  • R 1 , R 2 , L, Y, R 6 , A and n are as defined above,
  • a preferred embodiment of this aspect is the above compound which is a compound of formula (IV):
  • R 1 , R 2 , L, R 6 , A and n are as defined above,
  • a preferred embodiment of this aspect is the compound described above, wherein
  • Ring A is selected from the following structures:
  • ring A is optionally substituted with from 1 to 6 R 11 substituents;
  • ring A is selected from the following structures:
  • ring A is optionally substituted with from 1 to 6 R 11 substituents;
  • ring A is selected from the following structures:
  • ring A is optionally substituted with from 1 to 6 R 11 substituents, wherein each X 4 is independently selected from C(R 11 ) 2 , NR 11 , O, S, C(O), S(O) or S ( O) 2 , each X 5 is independently selected from C(R 11 ) 2 or NR 11 ;
  • ring A is selected from the following structures:
  • ring A is optionally substituted with from 1 to 6 R 11 substituents, wherein each X 4 is independently selected from C(R 11 ) 2 , NR 11 , O, S, C(O), S(O) or S ( O) 2 , each X 5 is independently selected from C(R 11 ) 2 or NR 11 atoms;
  • ring A is selected from the group consisting of:
  • ring A is selected from the group consisting of:
  • ring A is selected from the group consisting of:
  • R 11 is independently selected from H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally substituted C 2-6 olefin a optionally substituted C 1 -C 6 alkoxy group or an optionally substituted C 1 -C 6 haloalkoxy group; preferably, R 11 is independently selected from H, halogen, optionally substituted C 1 -C 6 An alkyl group or an optionally substituted C 2-6 alkenyl group; more preferably, R 11 is independently selected from H, -F, methyl, isopropyl or 1-propen-2-yl; or, wherein two phases The adjacent R 11 may together form an optionally substituted C 5 -C 8 carbocyclic group, an optionally substituted 5 to 8 membered heterocyclic group, an optionally substituted C 6 -C 14 aryl group or an optionally substituted 5 to 10 yuan
  • ring A is selected from the group consisting of:
  • a preferred embodiment of this aspect is the compound described above, wherein
  • Y is selected from the following structures:
  • Y is selected from the following structures:
  • Y is selected from the following structures:
  • R 3 , R 4 and R 5 are independently selected from H, halogen, optionally substituted C 1 -C 6 alkyl or optionally substituted C 1 -C 6 haloalkyl; preferably, R 3 , R 4 and R 5 is independently selected from H or halogen; more preferably, R 3 , R 4 and R 5 are all H.
  • a preferred embodiment of this aspect is the above compound, wherein m is 1 or 2; preferably, m is 1.
  • R 1 is selected from the group consisting of H, halogen, -CN, -NO 2 , optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl , optionally substituted C 1 -C 6 alkoxy, optionally substituted C 1 -C 6 haloalkoxy, optionally substituted C 1 -C 6 alkylthio, optionally substituted C 1 -C 6 alkane
  • R 1 is selected from the group consisting of H, halogen, -CN, -NO 2 , optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 a -C 6 haloalkyl group, an optionally substituted C 1 -C 6 alkoxy group or an optionally substituted C 1 -C 6 haloalkoxy group; preferably, R
  • a preferred embodiment of this aspect is the above compound, wherein R 2 is selected from H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally substituted C 1 -C 6 alkoxy, optionally substituted C 1 -C 6 haloalkoxy, optionally substituted C 1 -C 6 alkylthio, optionally substituted C 1 -C 6 alkylamino or optionally substituted C 1 -C 6 alkanoyl; preferably, R 2 is selected from H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally substituted C 1 -C 6 alkoxy or optionally substituted C 1 -C 6 haloalkoxy; preferably R 2 is selected from H, halogen, optionally substituted C 1 -C 6 alkyl or optionally substituted C 1 -C 6 Alkoxy; more preferably
  • a preferred embodiment of this aspect is the compound described above, wherein
  • R 6 is selected from a C 1 -C 6 alkyl group or a 4 to 7 membered heterocyclic group; more preferably, R 6 is selected from a C 1 -C 6 alkyl group or a 5 to 6 membered heterocyclic group; more preferably, R 6 Either selected from ethyl or 6-membered heterocyclic groups; more preferably, R 6 is selected from ethyl or 6-membered heterocyclic group containing 2 N heteroatoms; more preferably, R 6 is selected from ethyl or piperazinyl;
  • R 6 is optionally substituted by a halogen, -OH, -CN, -NO 2 , an optionally substituted C 1 -C 6 alkyl group, an optionally substituted C 1 -C 6 alkoxy group or NR 8 R 9 ;
  • R 6 is optionally substituted by a halogen, an optionally substituted C 1 -C 6 alkyl group, an optionally substituted C 1 -C 6 alkoxy group or NR 8 R 9 ; more preferably , R 6 is optionally substituted with the following substituent: optionally substituted C 1 -C 6 alkyl or NR 8 R 9 ;
  • R 8 and R 9 are each independently selected from an optionally substituted C 1 -C 6 alkyl group, an optionally substituted C 1 -C 6 alkoxy group or an optionally substituted C 3 -C 6 carbocyclic group; R 8 and R 9 are each independently selected from optionally substituted C 1 -C 6 alkyl; more preferably, R 8 and R 9 are both methyl;
  • R 8 and R 9 together with the nitrogen atom to which they are attached form an optionally substituted 4 to 6 membered heterocyclic group, for example, substituted by a C 1 -C 6 alkyl group.
  • a preferred embodiment of this aspect is the above compound, wherein n is 0 or 1.
  • a preferred embodiment of this aspect is the above compound, wherein L is selected from NR 7 , O, C(R 7 ) 2 or S; preferably, L is selected from NR 7 , O or S; more preferably, L is NR 7
  • R 7 is selected from H, optionally substituted C 1 -C 6 alkyl or optionally substituted C 1 -C 6 alkoxy; preferably, R 7 is selected from H or optionally substituted C 1 -C 6 alkyl; more preferably, R 7 is an optionally substituted C 1 -C 6 alkyl; more preferably, R 7 is methyl.
  • a preferred embodiment of this aspect is the above compound, wherein the group -(L) n -R 6 is N(Me)CH 2 CH 2 N(Me) 2 or is selected from the following structures:
  • a preferred embodiment of this aspect is the compound described above, wherein
  • W is selected from C(R 10 ) 2 , NR 10 , O or S; preferably, W is selected from NR 10 , O or S; more preferably, W is NR 10 ;
  • R 10 is selected from H, halogen, -CN, -NO 2 , optionally substituted C 1-6 alkyl, optionally substituted C 1-6 haloalkyl or optionally substituted C 1-6 , as long as it is chemically acceptable. Alkoxy; preferably, R 10 is selected from H, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 haloalkyl or optionally substituted C 1-6 alkoxy; more preferably R 10 is selected from H or an optionally substituted C 1-6 alkyl group; more preferably, R 10 is H.
  • a preferred embodiment of this aspect is the above compound, wherein R 1 is selected from H or F; R 2 is methoxy; -(L) n -R 6 is selected from N(Me)CH 2 CH 2 N(Me) 2 Or 4-methylpiperazin-1-yl; and if present, L is
  • the invention provides a pharmaceutical composition comprising a compound of the invention 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, a pharmaceutically acceptable salt, stereoisomer, solvate, hydrate, polymorph, prodrug or isotope derivative thereof, and other therapeutic agents, and A kit of pharmaceutically acceptable carriers, adjuvants or vehicles.
  • the invention provides a method of treating cancer caused by EGFR (including cancer caused by EGFR mutation, eg, a cancer with a T790M mutation, a L858R mutation, and a L858R/T790M double mutation) in a subject in need thereof
  • a method comprising: administering to a subject an effective amount of a compound of the invention.
  • the EGFR-induced cancer is selected from the group consisting of: non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cancer, Gastrointestinal stromal tumors, leukemia, histiocytic lymphoma, nasopharyngeal carcinoma, etc.
  • the compound is administered orally, subcutaneously, intravenously or intramuscularly. In a specific embodiment, the compound is administered chronically.
  • C 1 -C 6 alkyl includes C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1 -C 6 , C 1 -C 5 , C 1 -C 4 , C 1 - C 3 , C 1 -C 2 , C 2 -C 6 , C 2 -C 5 , C 2 -C 4 , C 2 -C 3 , C 3 -C 6 , C 3 -C 5 , C 3 -C 4 C 4 -C 6 , C 4 -C 5 and C 5 -C 6 alkyl.
  • C 1 -C 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 -C 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, ie, unsubstituted ("unsubstituted alkyl") or substituted with one or more substituents ("substituted alkyl"); For example, 1 to 5 substituents, 1 to 3 substituents or 1 substituent.
  • the alkyl group is unsubstituted C 1 -C 6 alkyl (e.g., -CH 3). In some embodiments, the alkyl group is a substituted C 1 -C 6 alkyl.
  • C 2 -C 6 alkenyl refers to a straight or branched chain hydrocarbon radical having from 2 to 6 carbon atoms and one or more carbon-carbon double bonds (eg, 1, 2 or 3 carbon-carbon double bonds) group.
  • 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-propen-2-yl (C 3 ), 1- Butenyl (C 4 ), 2-butenyl (C 4 ), butadienyl (C 4 ), pentenyl (C 5 ), pentadienyl (C 5 ), hexenyl (C 6 ),and many more.
  • each of the alkenyl groups is independently optionally substituted, ie, unsubstituted ("unsubstituted alkenyl") or substituted with one or more substituents ("substituted alkenyl"); For example, 1 to 5 substituents, 1 to 3 substituents or 1 substituent.
  • an alkenyl group is an unsubstituted C 2-6 alkenyl group.
  • an alkenyl group is a substituted C 2-6 alkenyl group.
  • C 2 -C 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 A linear or branched hydrocarbon group of a carbon-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 ), 3-methylbut-1-ynyl (C 5 ), hexynyl (C 6 ), and the like.
  • each of the alkynyl groups is independently optionally substituted, ie, unsubstituted ("unsubstituted alkynyl") or substituted with one or more substituents ("substituted alkynyl"); For example, 1 to 5 substituents, 1 to 3 substituents or 1 substituent.
  • an alkynyl group is an unsubstituted C 2-6 alkynyl group.
  • an alkynyl group is a substituted C 2-6 alkynyl group.
  • C 1 -C 6 alkoxy refers to the group -OR wherein R is a substituted or unsubstituted C 1 -C 6 alkyl group. In some embodiments, a C 1 -C 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.
  • C 1 -C 6 alkylthio refers to the group -SR wherein R is optionally substituted C 1 -C 6 alkyl.
  • a C 1 -C 4 alkylthio group is particularly preferred.
  • the C 1 -C 6 alkylthio group includes, but is not limited to, methylthio, ethylthio, n-propylthio, isopropylthio, n-butylthio, tert-butylthio, sec-butylthio, N-pentylthio, n-hexylthio and 1,2-dimethylbutylthio.
  • C 1 -C 6 alkylamino refers to the group -NHR or -NR 2 wherein R is optionally substituted C 1 -C 6 alkyl.
  • a C 1 -C 4 alkylamino group is particularly preferred.
  • the C 1 -C 6 alkylamino group includes, but is not limited to, methylamino, ethylamino, n-propylamino, isopropylamino, n-butylamino, t-butylamino, dimethylamino, methylethylamino and diethylamino.
  • 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 -C 6 haloalkyl and “C 1 -C 6 haloalkoxy” mean the above-mentioned “C 1 -C 6 alkyl” and "C 1 -C 6 alkoxy", which are one or Multiple halogen groups are substituted.
  • a C 1 -C 4 haloalkyl group is particularly preferred, more preferably a C 1 -C 2 haloalkyl group.
  • a C 1 -C 4 haloalkoxy group is particularly preferred, more preferably a C 1 -C 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 -C 8 carbocyclic group means a non-aromatic cyclic hydrocarbon group having 3 to 8 ring carbon atoms and zero hetero atoms.
  • a C 5 -C 8 carbocyclic group, a non-aromatic cyclic hydrocarbon group having 5 to 8 ring carbon atoms and zero heteroatoms is preferred.
  • a 5-membered carbocyclic group is preferred, which is a non-aromatic cyclic hydrocarbon group having 5 ring carbon atoms and zero heteroatoms.
  • Carbocyclyl also includes ring systems wherein the above carbocyclyl ring is fused to one or more aryl or heteroaryl groups, wherein the point of attachment is on the carbocyclic ring, and in such cases, the number of carbons continues to be represented The number of carbons in the carbocyclic system.
  • carbocyclic 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 ), cyclopentadienyl (C 5 ), cyclohexyl (C 6 ), cyclohexenyl (C 6 ), cyclohexadienyl (C 6 ), and Cycloheptyl (C 7 ), cycloheptenyl (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 ), and the
  • each of the carbocyclic groups is independently optionally substituted, ie, unsubstituted ("unsubstituted carbocyclyl") or substituted with one or more substituents ("substituted carbocyclic group"").
  • the carbocyclic group is unsubstituted C 3 -C 8 carbocyclyl.
  • the carbocyclyl is substituted with C 3 -C 8 carbocyclyl.
  • 3 to 8 membered heterocyclic group means a 3 to 8 membered non-aromatic ring system having a ring carbon atom and 1 to 3 ring heteroatoms, wherein each hetero atom is independently selected from the group consisting of nitrogen, oxygen, sulfur, boron, Phosphorus and silicon, wherein carbon, nitrogen, sulfur and phosphorus atoms may also exist in their oxidation state, such as C(O), S(O), S(O) 2 , P(O) and the like.
  • the point of attachment may be a carbon or nitrogen atom as long as the valence permits.
  • a 4- to 7-membered heterocyclic group is preferred, which is a 4 to 7 membered non-aromatic ring system having a ring carbon atom and 1 to 3 ring heteroatoms.
  • a 5- to 8-membered heterocyclic group is preferred, which is a 5 to 8 membered non-aromatic ring system having a ring carbon atom and 1 to 3 ring heteroatoms.
  • a 4 to 6 membered heterocyclyl group is preferred, which is a 4 to 6 membered non-aromatic ring system having a ring carbon atom and 1 to 3 ring heteroatoms.
  • a 5- to 6-membered heterocyclic group is preferred, which is a 5 to 6 membered non-aromatic ring system having a ring carbon atom and 1 to 3 ring heteroatoms.
  • a 5-membered heterocyclic group is more preferred, which is a 5-membered non-aromatic ring system having a ring carbon atom and 1 to 3 ring heteroatoms.
  • the cyclic group contains 1 to 3 (more preferably 1 or 2) ring heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, preferably nitrogen or oxygen.
  • each of the heterocyclic groups is independently optionally substituted, that is, unsubstituted ("unsubstituted heterocyclic group") or substituted with one or more substituents ("substituted heterocyclic ring"base").
  • the heterocyclyl is an unsubstituted 3-8 membered heterocyclyl.
  • a heterocyclic group is a substituted 3-8 membered heterocyclyl.
  • the heterocyclic group further includes a ring system in which the above heterocyclic ring is fused to one or more carbocyclic groups, wherein the point of attachment is on the carbocyclic ring, or wherein the above heterocyclic ring is bonded to one or more aryl groups or A heteroaryl fused ring system wherein the point of attachment is on a heterocyclyl ring; and in such cases, the number of ring members continues to indicate the number of ring members in the heterocyclyl ring system.
  • Exemplary 3-membered heterocyclic groups containing one hetero atom include, but are not limited to, aziridine, oxacyclopropane, thicyclopropyl.
  • 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, dioxolyl, oxathiolanyl, oxathiolanyl (1,2-oxothio) Cyclopentenyl, 1,3-oxathiolanyl, dithiolanyl, dihydropyrazolyl, dihydroimidazolyl, dihydrothiazolyl, dihydroisothiazolyl, dihydrogen Oxazolyl, dihydroisoxazolyl, dihydrooxadiazolyl and oxazolidin-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, tetrahydropyridyl and thiacyclohexane.
  • Exemplary 6-membered heterocyclic groups containing two heteroatoms include, but are not limited to, dihydropyrazinyl, piperazinyl, morpholinyl, dithianyl, dioxoalkyl.
  • Exemplary 6-membered heterocyclic groups containing three heteroatoms include, but are not limited to, hexahydrotriazinyl.
  • Exemplary 7-membered heterocyclic groups containing one or two heteroatoms include, but are not limited to, azepanyl, diazepanyl, oxacycloheptyl, and thienyl.
  • Exemplary 8-membered heterocyclic groups containing one or two heteroatoms include, but are not limited to, azacyclooctyl, oxacyclooctyl, thietane, octahydrocyclopentadienyl [ c] pyrrolyl and octahydropyrrolo[3,4-c]pyrrolyl.
  • Exemplary 5-membered heterocyclic groups (also referred to herein as 5,6-bicyclic heterocyclic groups) fused to a C 6 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 -C 14 aryl refers to a monocyclic or polycyclic (eg, bicyclic or tricyclic) 4n+2 having 6 to 14 ring carbon atoms and zero heteroatoms provided in an aromatic ring system.
  • An aromatic ring system for example, a group having 6, 10 or 14 ⁇ electrons shared in a cyclic arrangement
  • C 6-14 aryl an aryl group having six ring carbon atoms
  • C 6 aryl an aryl group having six ring carbon atoms
  • 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).
  • C 6 -C 14 aryl also includes ring systems in which the above aryl ring is fused to one or more carbocyclic or heterocyclic groups, wherein the atomic group or point of attachment is On the aryl ring, in this case, the number of carbon atoms continues to indicate the number of carbon atoms in the aryl ring system.
  • Typical aryl groups include, but are not limited to, those derived from acetaminophen, decene, acephenanthrylene, hydrazine, hydrazine, benzene, quinone, acetophenone, fluoranthene, anthracene, hexacene, and hexyl Fen, hexene, asymmetric introduction, symmetric introduction, indole, anthraquinone, naphthalene, octabenzene, octene, octene, egg benzene, penta-2,4-diene, pentacene , pentylene, penfen, hydrazine, phenalene, phenanthrene, anthracene, heptarene, anthracene, quercetin, jade red, benzophenanthrene and quinone.
  • the aryl group includes a phenyl group, a naphthyl group, an anthracenyl group, and a tetrahydronaphthyl group.
  • each of the aryl groups is independently optionally substituted, that is, unsubstituted ("unsubstituted aryl") or substituted with one or more substituents ("substituted aryl").
  • the aryl group is an unsubstituted C 6-14 aryl group.
  • the aryl group is a substituted C 6-14 aryl group.
  • 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.
  • a 5-membered heteroaryl is preferred, which is a 5-membered monocyclic 4n+2 aromatic ring system having a ring carbon atom and 1-4 ring heteroatoms (eg, having 6 shared in a cyclic arrangement)
  • 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 carbocyclic 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.
  • each of the heteroaryl groups is independently optionally substituted, ie, unsubstituted ("unsubstituted heteroaryl") or substituted with one or more substituents ("substituted heteroaryl" base").
  • the heteroaryl is an unsubstituted 5 to 10 membered heteroaryl.
  • the heteroaryl is a substituted 5 to 10 membered heteroaryl.
  • Exemplary 5-membered heteroaryl groups containing one hetero atom include, but are not limited to, pyrrolyl, furyl and thienyl.
  • An exemplary 5-membered heteroaryl group containing two heteroatoms includes However, it is 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 .
  • Alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl as defined herein are optionally substituted groups.
  • substituted whether or not preceded by the term “optionally”, means that at least one hydrogen present on a group (eg, a carbon or nitrogen atom) is substituted with an allowable substituent, for example, when substituted.
  • Substituents that result in stable compounds for example, compounds that do not spontaneously undergo a transition (eg, by rearrangement, cyclization, elimination, or other reaction).
  • a “substituted” group has a substituent at one or more substitutable positions of the group, and when more than one position in any given structure is substituted, The substituents at one position are the same or different.
  • substituted includes substitutions made with all of the permissible substituents of the organic compound that result in the formation of any of the substituents described herein.
  • a hetero atom such as nitrogen can have a hydrogen substituent and/or any suitable substituent described herein that satisfies the valence of the hetero atom and results in the formation of a stable moiety.
  • 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
  • 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 bases.
  • non-toxic acid addition salts are salts of amino and 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, or a salt formed using methods used in the art, for example, an ion exchange method.
  • amino and inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, Maleic acid, tartaric acid, citric acid, succinic acid or malonic acid, or a salt formed using methods used in the art, for example, an ion exchange method.
  • 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.
  • Further pharmaceutically acceptable salts include non-toxic ammonium salts, quaternary ammonium salts and amine cations formed using counterions, counterions such as halides, hydroxides, carboxylates, sulfates, phosphates, Nitrate, lower alkyl sulfonate and aryl sulfonate.
  • 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.
  • 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”).
  • an "effective amount" of a compound refers to an amount sufficient to cause a target biological response.
  • an effective amount of a compound of the invention can vary depending on, for example, the biological target, the pharmacokinetics of the compound, the condition being treated, the mode of administration, and the age of the subject. Health conditions and symptoms. Effective amounts include therapeutically and prophylactically effective amounts.
  • a "therapeutically effective amount" of a compound as used herein is an amount sufficient to provide a therapeutic benefit in the course of treating a disease, disorder or condition, or one or more conditions associated with a disease, disorder or condition. Delay or minimize.
  • a therapeutically effective amount of a compound refers to the amount of a therapeutic agent used alone or in combination with other therapies that provides a therapeutic benefit in the treatment of a disease, disorder or condition.
  • the term "therapeutically effective amount” can include an amount that improves overall treatment, reduces or avoids the symptoms or causes of a disease or condition, or enhances the therapeutic efficacy of other therapeutic agents.
  • a “prophylactically effective amount” of a compound is an amount sufficient to prevent a disease, disorder, or condition, or a quantity sufficient to prevent one or more symptoms associated with a disease, disorder, or condition, or to prevent disease, unless otherwise stated. The number of relapses of a disorder or condition.
  • a prophylactically effective amount of a compound refers to the amount of a therapeutic agent used alone or in combination with other agents that provides a prophylactic benefit in the prevention of a disease, disorder or condition.
  • the term “prophylactically effective amount” can include an amount that improves the overall amount of prevention, or enhances the prophylactic efficacy of other prophylactic agents.
  • 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.
  • Cancer-induced cancer refers to a cancer characterized by an unsuitable high expression of the EGFR gene or a mutation of the EGFR gene that alters the biological activity of the EGFR nucleic acid molecule or polypeptide. Cancers caused by EGFR can occur in any tissue including brain, blood, connective tissue, liver, mouth, muscle, spleen, stomach, testes, and trachea.
  • Cancers caused by EGFR include, but are not limited to, non-small cell lung cancer, small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell carcinoma, gastrointestinal stromal tumor, leukemia , tissue cell lymphoma, nasopharyngeal cancer.
  • an "EGFR mutation” or “EGFR mutant” includes one or more deletions, substitutions or additions in the amino acid or nucleotide sequence of the EGFR protein or EGFR coding sequence.
  • the EGFR mutation may also include one or more deletions, substitutions or additions, or fragments thereof, so long as the mutant retains or increases tyrosine kinase activity relative to wild-type EGFR.
  • the kinase or phosphorylation activity can be increased or decreased relative to wild-type EGFR (eg, at least 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70). %, 80%, 90% or even 100%).
  • Exemplary EGFR mutations include, but are not limited to, the T790M mutation, the L858R mutation, and the L858R/T790M double mutation.
  • a compound of the invention refers to a pharmaceutically acceptable salt of a compound of the following formula (I), (II), (III), (III-a), (III-b) and (IV).
  • the pharmaceutically acceptable salts of the compounds of (I), (II), (III), (III-a), (III-b), (IV) and (IV-a) also include any of their possible Stereoisomers, solvates, hydrates, polymorphs, prodrugs or isotopic derivatives.
  • the invention relates to a compound of formula (I):
  • R 1 is selected from the group consisting of H, halogen, -CN, -NO 2 , optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally substituted C 1 -C 6 alkoxy , optionally substituted C 1 -C 6 haloalkoxy, optionally substituted C 3 -C 6 carbocyclyl, optionally substituted C 1 -C 6 alkylthio, optionally substituted C 1 -C 6 An alkylamino group or an optionally substituted C 1 -C 6 alkanoyl group;
  • R 2 is selected from the group consisting of H, halogen, -CN, -NO 2 , optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally substituted C 1 -C 6 alkoxy a optionally substituted C 1 -C 6 haloalkoxy group, an optionally substituted C 1 -C 6 alkylthio group, an optionally substituted C 1 -C 6 alkylamino group or an optionally substituted C 1 -C 6 alkane Acyl group
  • L is selected from the group consisting of a bond, NR 7 , O, C(R 7 ) 2 or S; wherein each R 7 is independently selected from H, optionally substituted C 1 -C 6 alkyl, for example by C 1 -C 6 alkoxy Substituted C 1 -C 6 alkyl, or optionally substituted C 1 -C 6 alkoxy;
  • Y is H, or Y is selected from the following structures:
  • R 3 , R 4 and R 5 are independently selected from H, halogen, -CN, optionally substituted C 1 -C 6 alkyl or optionally substituted C 1 -C 6 haloalkyl;
  • R 6 is selected from H, C 1 -C 6 alkyl, C 2 -C 6 alkynyl or 3 to 8 membered heterocyclic, wherein the above group is optionally substituted with from 1 to 3 substituents: halogen, -OH , -CN, -NO 2 , optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 alkoxy, -C(O)R 8 or NR 8 R 9 ; wherein R 8 and R 9 are each independently selected from H, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 alkoxy, optionally substituted C 1 -C 6 haloalkyl or optionally substituted a C 3 -C 6 carbocyclic group, or R 8 and R 9 together with the nitrogen atom to which they are attached form an optionally substituted 4 to 6 membered heterocyclic group, for example, substituted by C 1 -C 6 alkyl;
  • W is selected from the group consisting of a bond, C(R 10 ) 2 , NR 10 , O or S, wherein R 10 is selected from H, halogen, -CN, -NO 2 , optionally substituted C 1 -C 6 alkyl, optionally substituted a C 1 -C 6 haloalkyl group or an optionally substituted C 1 -C 6 alkoxy group, as long as it is chemically acceptable;
  • n 0, 1 or 2;
  • n 0, 1 or 2;
  • Ring A is selected from the following structures:
  • each X 1 is independently selected from C(R 11 ) 2 , NR 11 , O or S
  • each of X 2 and X 3 is independently selected from C(R 11 ) 2 or NR 11 , wherein X 1 and X 2 are The location to connect with the mother core;
  • Ring B is selected from a 5-membered carbocyclic group, a 5-membered heterocyclic group or a 5-membered heteroaryl group, which is optionally substituted with from 1 to 3 R 11 substituents;
  • R 11 is independently selected from H, halogen, -CN, -NO 2 , -OH, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 2 -C 6 alkynyl, optionally substituted C 1 -C 6 alkoxy, optionally substituted C 3 -C 6 carbocyclyl or optionally substituted C 1 a -C 6 haloalkoxy group, as long as it is chemically acceptable; or wherein two adjacent R 11 groups may together form an optionally substituted C 5 -C 8 carbocyclic group, an optionally substituted 5 to 8 membered heterocyclic group, An optionally substituted C 6 -C 14 aryl or an optionally substituted 5 to 10 membered heteroaryl;
  • R 1 is selected from the group consisting of H, halogen, -CN, -NO 2 , optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally a substituted C 1 -C 6 alkoxy group, an optionally substituted C 1 -C 6 haloalkoxy group, an optionally substituted C 3 -C 6 carbocyclic group, an optionally substituted C 1 -C 6 alkylthio group, An optionally substituted C 1 -C 6 alkylamino group or an optionally substituted C 1 -C 6 alkanoyl group; preferably, R 1 is selected from H, halogen, -CN, -NO 2 , optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally substituted C 1 -C 6 alkoxy, optionally substituted C 1 -C 6 haloalk
  • R 2 is selected from H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally substituted C 1 -C 6 Alkoxy, optionally substituted C 1 -C 6 haloalkoxy, optionally substituted C 3 -C 6 carbocyclyl, optionally substituted C 1 -C 6 alkylthio, optionally substituted C 1 - C 6 alkylamino or optionally substituted C 1 -C 6 alkanoyl; preferably, R 2 is selected from H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkane , optionally substituted C 1 -C 6 alkoxy, optionally substituted C 1 -C 6 haloalkoxy, optionally substituted C 1 -C 6 alkylthio, optionally substituted C 1 -C 6 An alkylamino group or
  • R 6 is selected from a C 1 -C 6 alkyl group, a C 2 -C 6 alkynyl group or a 3 to 8 membered heterocyclic group; preferably, R 6 is selected from a C 1 -C 6 alkane Or a 4- to 7-membered heterocyclic group; more preferably, R 6 is selected from a C 1 -C 6 alkyl group or a 5- to 6-membered heterocyclic group; more preferably, R 6 is selected from an ethyl group or a 6-membered heterocyclic group.
  • R 6 is selected from ethyl or a 6-membered heterocyclic group containing 1 or 2 N heteroatoms; more preferably, R 6 is selected from ethyl or 6-membered heterocyclic ring containing 2 N heteroatoms More preferably, R 6 is selected from ethyl, piperidinyl, morpholinyl or piperazinyl; more preferably, R 6 is selected from ethyl or piperazinyl.
  • R 6, R 6 is optionally substituted with the following substituents: halogen, -OH, -CN, -NO 2, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 - C 6 alkoxy or NR 8 R 9 ; preferably, R 6 is optionally substituted by a halogen, an optionally substituted C 1 -C 6 alkyl group, an optionally substituted C 1 -C 6 alkoxy group Or NR 8 R 9 ; more preferably, R 6 is optionally substituted by an optionally substituted C 1 -C 6 alkyl group, an optionally substituted C 1 -C 6 alkoxy group or NR 8 R 9 ; More preferably, R 6 is optionally substituted by an optionally substituted C 1 -C 6 alkyl group or NR 8 R 9 .
  • R 8 and R 9 are each independently selected from an optionally substituted C 1 -C 6 alkyl group, an optionally substituted C 1 -C 6 alkoxy group or an optionally substituted C 3 -C 6 carbocyclic group; R 8 and R 9 are each independently selected from an optionally substituted C 1 -C 6 alkyl group; more preferably, both R 8 and R 9 are methyl groups.
  • R 8 and R 9 together with the nitrogen atom to which they are attached form an optionally substituted 4 to 6 membered heterocyclic group, for example substituted by C 1 -C 6 alkyl or C 1 -C 6 alkoxy; or R 8 and R 9 together with the nitrogen atom to which they are attached form an optionally substituted 4 to 6 membered heterocyclic group, for example, substituted by a C 1 -C 6 alkyl group; more preferably, R 8 and R 9 are taken together with the nitrogen atom to which they are attached a 4 to 6 membered heterocyclic group containing 1 N hetero atom, or 2 N hetero atoms, or 1 N hetero atom and 1 O hetero atom, optionally C 1 -C 6 alkyl or C 1 -C 6 alkoxy substituted; more preferably, R 8 and R 9 together with the nitrogen atom to which they are attached form one N hetero atom, or two N hetero atoms, or one N hetero atom and one O hetero A 4- to 6-membered heterocyclic group
  • L is selected from NR 7 , O, C(R 7 ) 2 or S; preferably, L is selected from NR 7 , O or S; more preferably, L is NR 7 .
  • R 7 is selected from H, optionally substituted C 1 -C 6 alkyl or optionally substituted C 1 -C 6 alkoxy; preferably, R 7 is selected from H or optionally Substituted C 1 -C 6 alkyl; more preferably, R 7 is an optionally substituted C 1 -C 6 alkyl; more preferably, R 7 is methyl.
  • ring A is selected from the following structures:
  • ring A is optionally substituted with from 1 to 6 R 11 substituents;
  • ring A is selected from the following structures:
  • ring A is optionally substituted with from 1 to 6 R 11 substituents;
  • ring A is selected from the following structures:
  • ring A is optionally substituted with from 1 to 6 R 11 substituents;
  • ring A is selected from the following structures:
  • ring A is optionally substituted with from 1 to 6 R 11 substituents, wherein each X 4 is independently selected from C(R 11 ) 2 , NR 11 , O, S, C(O), S(O) or S ( O) 2 , each X 5 is independently selected from C(R 11 ) 2 or NR 11 ;
  • ring A is selected from the following structures:
  • ring A is optionally substituted with from 1 to 6 R 11 substituents, wherein each X 4 is independently selected from C(R 11 ) 2 , NR 11 , O, S, C(O), S(O) or S ( O) 2 , each X 5 is independently selected from C(R 11 ) 2 or NR 11 ;
  • ring A is selected from the following structures:
  • ring A is optionally substituted with from 1 to 6 R 11 substituents, wherein each X 4 is independently selected from C(R 11 ) 2 , NR 11 , O, S, C(O), S(O) or S ( O) 2 , each X 5 is independently selected from C(R 11 ) 2 or NR 11 ;
  • ring A is selected from the group consisting of:
  • ring A is selected from the group consisting of:
  • ring A is selected from the group consisting of:
  • ring A is selected from the group consisting of:
  • ring A is selected from the group consisting of:
  • R 11 is independently selected from H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally as long as it is chemically acceptable a substituted C 2-6 alkenyl group, an optionally substituted C 1 -C 6 alkoxy group, an optionally substituted C 3 -C 6 carbocyclic group or an optionally substituted C 1 -C 6 haloalkoxy group; preferably R 11 is independently selected from H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally substituted C 2-6 alkenyl, optionally substituted C 1 -C 6 alkoxy or optionally substituted C 1 -C 6 haloalkoxy; preferably, R 11 is independently selected from H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally
  • ring A is selected from the group consisting of:
  • ring A is selected from the group consisting of:
  • Y is H; preferably, Y is selected from the following structures:
  • Y is selected from the following structures:
  • R 3 , R 4 and R 5 are independently selected from H, halogen, optionally substituted C 1 -C 6 alkyl or optionally substituted C 1 -C 6 haloalkyl; preferably R 3 , R 4 and R 5 are independently selected from H or halogen; more preferably, R 3 , R 4 and R 5 are both H.
  • W is selected from C(R 10 ) 2 , NR 10 , O or S; preferably, W is selected from NR 10 , O or S; more preferably, W is NR 10 .
  • R 10 is selected from H, halogen, -CN, -NO 2 , optionally substituted C 1-6 alkyl, optionally substituted C 1-6 haloalkyl, or Optionally substituted C 1-6 alkoxy; preferably, R 10 is selected from H, optionally substituted C 1-6 alkyl, optionally substituted C 1-6 haloalkyl or optionally substituted C 1- 6 alkoxy; more preferably, R 10 is selected from H or optionally substituted C 1-6 alkyl; more preferably, R 10 is H.
  • n is 1 or 2; preferably, m is 1; more preferably, m is 0.
  • n is 0; preferably, n is 1.
  • L is selected from the group consisting of a bond, NR 7 , O or S; wherein R 7 is selected from H or an optionally substituted C 1 -C 6 alkyl group;
  • n 0, 1 or 2;
  • R 6 is selected from H, C 1 -C 6 alkyl or a 3- to 8-membered heterocyclic group wherein the above group is optionally substituted with from 1 to 3 substituents: halogen, -OH, -CN, -NO 2 , An optionally substituted C 1 -C 6 alkyl group, an optionally substituted C 1 -C 6 alkoxy group or NR 8 R 9 , wherein R 8 and R 9 are each independently selected from H or optionally substituted C 1 - C 6 alkyl; or R 8 and R 9 together with the nitrogen atom to which they are attached form an optionally substituted 4 to 6 membered heterocyclic group;
  • L is selected from a bond or NR 7 ; wherein R 7 is selected from H or an optionally substituted C 1 -C 6 alkyl;
  • n 0 or 1
  • R 6 is selected from H, C 1 -C 6 alkyl or a 3- to 8-membered heterocyclic group wherein the above group is optionally substituted with from 1 to 3 substituents: halogen, -OH, -CN, -NO 2 , An optionally substituted C 1 -C 6 alkyl group, an optionally substituted C 1 -C 6 alkoxy group or NR 8 R 9 , wherein R 8 and R 9 are each independently selected from H or optionally substituted C 1 - C 6 alkyl; or R 8 and R 9 together with the nitrogen atom to which they are attached form an optionally substituted 4 to 6 membered heterocyclic group;
  • -(L) n -R 6 is selected from:
  • -(L) n -R 6 is selected from:
  • -(L) n -R 6 is selected from:
  • -(L) n -R 6 is selected from:
  • the compound of formula (I) is a compound of formula (II):
  • R 1 , R 2 , L, R 6 , W, A, m and n are as defined above.
  • the compound of formula (I) is a compound of formula (III):
  • Y is selected from the following structures:
  • Y is selected from the following structures:
  • Y is the following structure:
  • Y is the following structure:
  • R 1 -R 6 , L, A and n are as defined above.
  • the compound of formula (I) is a compound of formula (IV):
  • R 1 , R 2 , L, R 6 , A and n are as defined above.
  • the present invention relates to the above compounds, wherein
  • R 1 is selected from H, halogen, -CN, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl, optionally substituted C 1 -C 6 alkoxy or optionally Substituted C 3 -C 6 carbocyclic group;
  • R 2 is selected from an optionally substituted C 1 -C 6 alkoxy group or an optionally substituted C 1 -C 6 haloalkoxy group;
  • -(L) n -R 6 is selected from:
  • Ring A is selected from:
  • R 11 is independently selected from H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 1 -C 6 haloalkyl, optionally a substituted C 1 -C 6 alkoxy group or an optionally substituted C 3 -C 6 carbocyclic group; or wherein two adjacent R 11 groups may together form an optionally substituted 5 to 8 membered heterocyclic group;
  • the present invention relates to the above compounds, wherein
  • -(L) n -R 6 is selected from:
  • the present invention relates to the above compounds, wherein
  • Ring A is selected from:
  • the invention relates to a compound of formula (III-a):
  • R 1 is selected from H, halogen, -CN, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl or optionally substituted C 1 -C 6 alkoxy; preferably R 1 is selected from H, halogen or optionally substituted C 1 -C 6 alkyl; preferably, R 1 is selected from H, -F, -Cl, -Br, -CN, -CH 3 , -CF 3 or -OCH 3 ; preferably, R 1 is selected from H, -Cl, -Br or -CH 3 ;
  • R 2 is selected from an optionally substituted C 1 -C 6 alkoxy group or an optionally substituted C 1 -C 6 haloalkoxy group; preferably, R 2 is selected from -OCH 3 , -OCH 2 CH 3 , -OCHF 2 Or -OCH 2 CF 3 ;
  • R 11 is selected from H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 1 -C 6 haloalkyl or optionally substituted C 1 -C 6 alkoxy; preferably, R 11 is selected from H, halogen or optionally substituted C 1 -C 6 alkoxy; preferably, R 11 is selected from H, -F, -Cl, -Br, - Me, -iPr, 1-propen-2-yl, -CH 2 F, CF 3 , -OMe or -OC(CH 3 ) 3 ; preferably, R 11 is selected from the group consisting of H, -F, -Cl, -Br, -OMe or -OC(CH 3 ) 3 ;
  • R 12 is selected from H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 1 -C 6 haloalkyl or optionally substituted C 1 -C 6 alkoxy; preferably, R 12 is selected from an optionally substituted C 1 -C 6 alkyl group or an optionally substituted C 1 -C 6 haloalkyl group; preferably, R 12 is selected from H, -F, -Cl, -Br, -Me, -iPr, 1-propen-2-yl, -CH 2 F, CF 3 , -OMe or -OC(CH 3 ) 3 ; preferably, R 12 is selected from -Me, - iPr or -CH 2 F;
  • R 13 is selected from H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 1 -C 6 haloalkyl or optionally substituted C 1 -C 6 alkoxy; preferably, R 13 is an optionally substituted C 1 -C 6 alkyl group; preferably, R 13 is selected from the group consisting of H, -F, -Cl, -Br, -Me, -iPr, 1 - propen-2-yl, -CH 2 F, CF 3 , -OMe or -OC(CH 3 ) 3 ; preferably, R 13 is selected from -Me or -iPr;
  • R 1 is H and R 2 and R 11 -R 13 are as defined above.
  • R 1 is halogen (e.g., -F, -Cl or -Br, preferably -Cl or -Br), and R 2 and R 11 -R 13 are as defined above.
  • R 1 is -CN, and R 2 and R 11 -R 13 are as defined above.
  • R 1 is an optionally substituted C 1 -C 6 alkyl group (e.g., -CH 3 ), and R 2 and R 11 -R 13 are as defined above.
  • R 1 is an optionally substituted C 1 -C 6 haloalkyl group (e.g., -CF 3 ), and R 2 and R 11 -R 13 are as defined above.
  • R 1 is an optionally substituted C 1 -C 6 alkoxy group (e.g., -OCH 3 ), and R 2 and R 11 -R 13 are as defined above.
  • the invention relates to a compound of formula (III-a):
  • R 1 is an optionally substituted C 3 -C 6 carbocyclic group; preferably, R 1 is a cyclopropyl group;
  • R 2 and R 11 -R 13 are as defined above.
  • R 2 is an optionally substituted C 1 -C 6 alkoxy group (e.g., -OCH 3 or -OCH 2 CH 3 ), and R 1 and R 11 -R 13 are as defined above.
  • R 2 is an optionally substituted C 1 -C 6 haloalkoxy group (e.g., -OCHF 2 or -OCH 2 CF 3 ), and R 1 and R 11 -R 13 are as defined above.
  • R 11 is H, and R 1 , R 2 , R 12 and R 13 are as defined above.
  • R 11 is halogen (e.g., -F, -Cl or -Br), and R 1 , R 2 , R 12 and R 13 are as defined above.
  • R 11 is an optionally substituted C 1 -C 6 alkyl group (e.g., -Me or -iPr), and R 1 , R 2 , R 12 and R 13 are as defined above.
  • R 11 is an optionally substituted C 2 -C 6 alkenyl group (e.g., 1-propen-2-yl), and R 1 , R 2 , R 12 and R 13 are as defined above.
  • R 11 is an optionally substituted C 1 -C 6 haloalkyl group (e.g., -CH 2 F or -CF 3 ), and R 1 , R 2 , R 12 and R 13 are as defined above.
  • R 11 is optionally substituted C 1 -C 6 alkoxy (eg, -OMe or -OC(CH 3 ) 3 ), and R 1 , R 2 , R 12 and R 13 As defined above.
  • R 12 is H, and R 1 , R 2 , R 11 and R 13 are as defined above.
  • R 12 is halogen (e.g., -F, -Cl or -Br), and R 1 , R 2 , R 11 and R 13 are as defined above.
  • R 12 is an optionally substituted C 1 -C 6 alkyl group (e.g., -Me or -iPr), and R 1 , R 2 , R 11 and R 13 are as defined above.
  • R 12 is an optionally substituted C 2 -C 6 alkenyl group (e.g., 1-propen-2-yl), and R 1 , R 2 , R 11 and R 13 are as defined above.
  • R 12 is optionally substituted C 1 -C 6 haloalkyl (eg, -CH 2 F or -CF 3 , preferably -CH 2 F), and R 1 , R 2 , R 11 And R 13 is as defined above.
  • C 1 -C 6 haloalkyl eg, -CH 2 F or -CF 3 , preferably -CH 2 F
  • R 12 is optionally substituted C 1 -C 6 alkoxy (eg, -OMe or -OC(CH 3 ) 3 ), and R 1 , R 2 , R 11 and R 13 As defined above.
  • R 13 is H, and R 1 , R 2 , R 11 and R 12 are as defined above.
  • R 13 is halo (e.g., -F, -Cl or -Br), and R 1 , R 2 , R 11 and R 12 are as defined above.
  • R 13 is an optionally substituted C 1 -C 6 alkyl group (e.g., -Me or -iPr), and R 1 , R 2 , R 11 and R 12 are as defined above.
  • R 13 is an optionally substituted C 2 -C 6 alkenyl group (e.g., 1-propen-2-yl), and R 1 , R 2 , R 11 and R 12 are as defined above.
  • R 13 is an optionally substituted C 1 -C 6 haloalkyl group (e.g., -CH 2 F or -CF 3 ), and R 1 , R 2 , R 11 and R 12 are as defined above.
  • R 13 is an optionally substituted C 1 -C 6 alkoxy group (eg, -OMe or -OC(CH 3 ) 3 ), and R 1 , R 2 , R 11 and R 12 As defined above.
  • the invention relates to a compound of formula (III-b) above:
  • R 1 is selected from H, halogen, -CN, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl or optionally substituted C 1 -C 6 alkoxy; preferably R 1 is selected from H, halogen or optionally substituted C 1 -C 6 alkyl; preferably, R 1 is selected from H, -F, -Cl, -Br, -CN, -CH 3 , -CF 3 or -OCH 3 ; preferably, R 1 is selected from H, -Cl, -Br or -CH 3 ;
  • R 2 is selected from an optionally substituted C 1 -C 6 alkoxy group or an optionally substituted C 1 -C 6 haloalkoxy group; preferably, R 2 is selected from -OCH 3 , -OCH 2 CH 3 , -OCHF 2 Or -OCH 2 CF 3 ;
  • R 11 is selected from H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 2 -C 6 alkenyl, optionally substituted C 1 -C 6 haloalkyl or optionally substituted C 1 -C 6 alkoxy; preferably, R 11 is selected from H, halogen or optionally substituted C 1 -C 6 alkoxy; preferably, R 11 is selected from H, -F, -Cl, -Br, - Me, -iPr, 1-propen-2-yl, -CH 2 F, CF 3 , -OMe or -OC(CH 3 ) 3 ; preferably, R 11 is selected from the group consisting of H, -F, -Cl, -Br, -OMe or -OC(CH 3 ) 3 ;
  • the invention relates to a compound of formula (III-b) above:
  • R 1 is an optionally substituted C 3 -C 6 carbocyclic group; preferably, R 1 is a cyclopropyl group;
  • R 2 and R 11 are as defined above;
  • the invention relates to a compound of formula (IV-a) above:
  • R 1 is selected from H, halogen, -CN, optionally substituted C 1 -C 6 alkyl, optionally substituted C 1 -C 6 haloalkyl or optionally substituted C 1 -C 6 alkoxy; preferably , R 1 is selected from H or -F;
  • R 2 is selected from an optionally substituted C 1 -C 6 alkoxy group or an optionally substituted C 1 -C 6 haloalkoxy group; preferably, R 2 is selected from H or -OCH 3 ;
  • the compounds of the invention are the following compounds:
  • 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 racemic mixture and a mixture rich in one or more stereoisomers are 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 invention also includes all suitable isotopic derivatives of the compounds of the invention.
  • An isotope derivative of a compound of the invention is defined as wherein at least one atom is replaced by an atom having the same atomic number but differing in atomic mass from the atomic mass typically found in nature.
  • isotopes which may be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine and chlorine, for example 2 H, 3 H, 13 C, 14 C, 15 N, 17 O, 18 O, respectively. 18 F, 31 P, 32 P, 35 S and 36 Cl.
  • isotopic derivatives of the compounds of the invention are useful in drug and/or substrate tissue distribution studies.
  • Deuterated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes are particularly preferred for their ease of preparation and detectability.
  • substitution with isotopes e.g., hydrazine, i.e., 2 H
  • Isotopic derivatives of the compounds of the invention can generally be prepared by conventional procedures, for example by descriptive methods or by the preparations described in the Examples below, using appropriate isotopic derivatives of the appropriate reagents.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof may be in an amorphous or crystalline form.
  • 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.
  • the term "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.
  • 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 carrier which, when administered to a patient, releases the compound of formula (I) 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, compounds of the invention wherein a hydroxy, amine or sulfhydryl group is bonded to any group which, when administered to a patient, can be cleaved to form a hydroxy, amine or sulfhydryl group.
  • prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol, mercapto and amine functional groups 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 white) Protein), 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 materials, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylate, wax, polyethylene-polyoxypropylene - Block
  • 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.
  • formulation examples illustrate representative pharmaceutical compositions that can be prepared in accordance with the present invention.
  • the invention is not limited to the following pharmaceutical compositions.
  • Exemplary Formulation 1 - Tablet The compound of the present invention in dry powder form can be mixed with the dried gel binder in a weight ratio of about 1:2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped into 0.3-30 mg tablets (each tablet contains 0.1-10 mg of active compound per tablet) in a tablet press.
  • Exemplary Formulation 2 - Tablet The compound of the present invention in dry powder form can be mixed with the dried gel binder in a weight ratio of about 1:2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is formed into a 30-90 mg tablet (each tablet contains 10-30 mg of active compound per tablet) in a tablet press.
  • Exemplary Formulation 3 - Tablet The compound of the invention in dry powder form can be combined with the dried gel binder in a weight ratio of about 1:2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped into 90-150 mg tablets (30-50 mg of active compound per tablet) in a tablet press.
  • Exemplary Formulation 4-Tablet The compound of the invention in dry powder form can be combined with the dried gel binder in a weight ratio of about 1:2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is formed into a 150-240 mg tablet (each tablet contains 50-80 mg of active compound per tablet) in a tablet press.
  • Exemplary Formulation 5 - Tablet The compound of the invention in dry powder form can be combined with the dried gel binder in a weight ratio of about 1:2. A smaller amount of magnesium stearate was added as a lubricant. Forming the mixture into a tablet press 240-270 mg tablets (each tablet contains 80-90 mg of active compound).
  • Exemplary Formulation 6-Tablet The compound of the invention in dry powder form can be combined with the dried gel binder in a weight ratio of about 1:2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped into a 270-450 mg tablet (each tablet contains 90-150 mg of active compound) in a tablet press.
  • Exemplary Formulation 7-Tablet The compound of the invention in dry powder form can be combined with the dried gel binder in a weight ratio of about 1:2. A smaller amount of magnesium stearate was added as a lubricant. The mixture is shaped into 450-900 mg tablets (each tablet contains 150-300 mg of active compound per tablet) in a tablet press.
  • Exemplary Formulation 8-Capsule The compound of the present invention in dry powder form can be mixed with a starch diluent in a weight ratio of about 1:1. The mixture was filled into 250 mg capsules (each capsule containing 125 mg of active compound).
  • Exemplary Formulation 9-Liquid The compound of the present invention (125 mg) can be mixed with sucrose (1.75 g) and xanthan gum (4 mg), and the resulting mixture can be blended, passed through a No. 10 mesh U.S. sieve, and then It was mixed with an aqueous solution of microcrystalline cellulose and sodium carboxymethylcellulose (11:89, 50 mg) prepared in advance. Sodium benzoate (10 mg), flavor and color are diluted with water and added with stirring. Then, sufficient water can be added to give a total volume of 5 mL.
  • Exemplary Formulation 10 - Injection The compound of the invention may be dissolved or suspended in a buffered sterile saline injectable aqueous medium to a concentration of about 5 mg/mL.
  • 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.
  • long-term administration is intended to be A constant level of the compound is provided in the blood for 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 increase the concentration of the compound in the blood to an effective level.
  • the bolus dose depends on the target systemic level of the active ingredient through the body, for example, an intramuscular or subcutaneous bolus dose that causes a slow release of the active ingredient, while a bolus that is delivered directly to the vein (eg, via IV IV drip) ) can be delivered more quickly, 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; lubricants, for example, magnesium stearate; A flow agent, for example, colloidal silica; a sweetener such as sucrose or saccharin; or a flavoring agent such as peppermint, 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 starch
  • 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 as well as processing techniques and the like are set forth in Remington's Pharmaceutical Sciences, 17 th edition , 1985, Mack Publishing Company, Easton, in Section 8 Pennsylvania, herein incorporated by reference in this document.
  • 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).
  • the compounds of the invention or compositions thereof may be administered in combination with other therapeutic agents to treat the disease.
  • therapeutic agents include, but are not limited to, Adriamycin, dexamethasone, vincristine, cyclophosphamide, fluorouracil, topotecan ( Topotecan), taxol, interferon, platinum derivatives, taxanes (eg, paclitaxel), vinca alkaloids (eg, vinblastine), anthracycline ( Anthracycline) (eg, doxorubicin), epipodophyllotoxin (eg, etoposide), cisplatin, mTOR inhibitor (eg, rapamycin), Methotrexate, actinomycin D, dolastatin 10, colchicine, emetine, trimetrexate, chlorobenzene Metoprine, cyclosporine, daunorubicin, teniposide, amphotericin, alkylating agent (eg chlorambucil
  • a compound of the invention or a composition thereof can be administered in combination with any one or more anti-proliferative or chemotherapeutic agents selected from the group consisting of: abarelix, aldileukin (aldesleukin), alemtuzumab, alitretinoin, allopurinol, altretamine, amifostine, anastrozole, arsenic trioxide, day Asparaginase, azacitidine, BCG Live, bevacuzimab, fluorouracil, bexarotene, bleomycin, bortezomib, white Busulfan, calbuterone, capecitabine, camptothecin, carboplatin, carmustine, celecoxib, cetuximab Antibiotic (cetuximab), chlorambucil, cladribine, clofarabine, cyclophosphamide, cytarabine, actinomycin D, darbepo
  • therapeutic agents to which the compounds of the invention may also be combined include, but are not limited to, therapeutic agents for Alzheimer's Disease, such as donepezil hydrochloride and rivastigmine. ; therapeutic agents for Parkinson's Disease, such as L-DOPA/carbidopa, entacapone, ropinrole, pramipexole ( Pramipexole), bromocriptine, pergolide, trihexephendyl, and amantadine; a therapeutic agent for multiple sclerosis (MS), Such as beta interferon, glatiramer acetate and mitoxantrone; therapeutic agents for asthma, such as albuterol and montelukast; therapeutic agents for schizophrenia, such as Zyprexa, risperdal, seroquel, and haloperidol; anti-inflammatory agents such as corticosteroids, TNF blockers, IL-1RA, azathioprine , cyclophosphamide and sulfasala
  • those other active agents can be administered separately from the compositions containing the compounds of the invention as part of a multiple dosing regimen.
  • those active agents may be part of a single dosage form, mixed with a compound of the invention in a single composition. If administered as part of a multiple dosing regimen, the two active agents can be provided simultaneously, sequentially, or at intervals from one another (usually within 5 hours of each other).
  • the invention provides a method for inhibiting a protein tyrosine kinase (such as EGFR kinase) or treating a disease (such as cancer, cell proliferative disease, inflammation, infection, immune disease, organ transplantation, viral disease, cardiovascular disease or A method of metabolic disease comprising the steps of administering a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, hydrate, crystal form, prodrug thereof, to a subject in need of treatment Or an isotope derivative, or a pharmaceutical composition of the invention.
  • a protein tyrosine kinase such as EGFR kinase
  • a disease such as cancer, cell proliferative disease, inflammation, infection, immune disease, organ transplantation, viral disease, cardiovascular disease or A method of metabolic disease comprising the steps of administering a compound of the invention, or a pharmaceutically acceptable salt, stereoisomer, solvate, hydrate, crystal form, prodrug thereof, to a subject in need of treatment Or
  • the compounds of the invention are useful in the treatment of cancer caused by EGFR.
  • the compounds are useful for treating cancer caused by EGFR expressing an EGFR mutant and for treating cancer caused by EGFR that is refractory to RTKI therapy (eg, erlotinib or gefitinib).
  • the compounds of the invention are inhibitors of at least one mutant of EGFR and are therefore suitable for treatment with one or more EGFR mutants (eg, deletion mutations, activating mutations, resistance mutations, or combinations thereof, specific examples include T790M mutations, One or more disorders associated with the activity of the L858R mutation and the L858R/T790M double mutation.
  • the invention provides a method of treating a mutant EGFR mediated disorder comprising administering a compound of the invention, or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, to a patient in need thereof a solvate, hydrate, crystal form, prodrug or isotope derivative, or a step of administering a pharmaceutical composition of the invention.
  • Cancers treatable by the compounds of the invention include, but are not limited to, non-small cell lung cancer (NSCLS), small cell lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell carcinoma, Hyperproliferative diseases such as gastrointestinal stromal tumors, leukemia, histiocytic lymphoma, and nasopharyngeal carcinoma.
  • the compounds of the invention may also be used to maintain the maintenance of cancer recurrence in patients in need of such treatment.
  • an effective amount of a compound of the invention will generally be administered in a single or multiple doses at an average daily dose of from 0.01 mg to 50 mg of compound per kilogram of patient body weight, preferably from 0.1 mg to 25 mg of compound per kilogram of patient body weight.
  • the compounds of the invention may be administered to a patient in need of such treatment in a daily dosage range of from about 1 mg to about 3500 mg per patient, preferably from 10 mg to 1000 mg.
  • the daily dose per patient can be 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, 400, 500, 600, 700, 800, 900 or 1000 mg. It can be administered one or more times daily, weekly (or several days apart) or on an intermittent schedule.
  • the compound can be administered one or more times per day on a weekly basis (e.g., every Monday), continually or for several weeks, such as 4-10 weeks.
  • the administration may be continued for several days (e.g., 2-10 days), followed by a few days (e.g., 1-30 days) without administration of the compound, and the cycle may be repeated indefinitely or repeated for a given number of times, such as 4-10 Cycles.
  • the compounds of the invention may be administered daily for 5 days, then intermittently for 9 days, then administered daily for 5 days, then intermittent for 9 days, and so on, optionally repeating the cycle or repeating 4-10 times.
  • EGFR-TKI eg, erlotinib or gefitinib
  • the combination therapy can be administered at their dosage level and regimen for monotherapy.
  • erlotinib has been orally administered at a dose of 150 mg per day for the treatment of non-small cell lung cancer, and has been orally administered at a dose of 100 mg per day for pancreatic cancer.
  • gefitinib has been administered orally at 250 mg per day for the treatment of non-small cell lung cancer.
  • EGFR-TKI e.g., erlotinib or gefitinib
  • the dosage level of one or both components is reduced compared to when used alone.
  • the compounds of the present invention can be prepared according to conventional methods in the art and using suitable reagents, starting materials, and purification methods known to those skilled in the art.
  • each reaction is usually 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.
  • Step 4 Compound 4-Fluoro-1-isopropyl-2-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- Synthesis of -1H-benzo[d]imidazole.
  • Step 1 Synthesis of the compound (4-fluoro-2-methoxy-5-nitrophenyl)carbamic acid tert-butyl ester.
  • Boc anhydride (7.05 g, 32.20 mmol) was slowly added dropwise to 4-fluoro-2-methoxy-5-nitroaniline (4.0 g, 21.50 mmol) and triethylamine (TEA, 3.60 g).
  • TEA triethylamine
  • Petroleum ether / ethyl acetate (v / v) 2: 1) gave 3.6 g of a yellow solid, yield: 58.8%.
  • LC-MS (APCI): m/z 287.2 (M+1).
  • Step 2 Synthesis of the compound (4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxy-5-nitrophenyl)carbamic acid tert-butyl ester.
  • Step 3 Synthesis of the compound (5-amino-4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxyphenyl)carbamic acid tert-butyl ester.
  • Step 4 Compound (5-Acrylamide-4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxyphenyl) Synthesis of tert-butyl carbamate.
  • Step 5 Synthesis of the compound N-(5-amino-2-((2-(dimethylamino)ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide.
  • 2,4-dichloro-5-fluoropyrimidine (517 mg, 3.1 mmol)
  • sodium carbonate (583 mg, 5.5 mmol) was dissolved in a mixed solution of 1 mL of water and 5 mL of ethylene glycol dimethyl ether under nitrogen atmosphere, and PdCl was added.
  • Step 1 Synthesis of the compound 5-bromo-3-fluoro-N 1 -isopropylbenzene-1,2-diamine.
  • Ethanol-water (4/1, v/v, 30 mL) was added to the next step, and reduced iron powder (5.74 g) and ammonium chloride (1.80 g) were added, and the mixture was heated under reflux at 90 ° C for 2.5 hours. After cooling to room temperature, the insoluble solid was removed by suction filtration, and most of the ethanol was evaporated under reduced pressure. 20 mL of a saturated aqueous solution of sodium hydrogencarbonate and 20 mL of EA. The organic phase was dried over anhydrous sodium sulfate and evaporated to dryness.
  • reaction solution was cooled to room temperature, and 50 mL of water was added, PE-EA (v/ v, 1/1, 10 mL * 2) extraction, combined organic phase evaporation column chromatography (PE / EA, v / v, 4 / 1) to give compound 51, a total of 0.49 g.
  • Phosphorus oxychloride (6.70 mL) was added to 4-bromo-2,6-difluoroaniline (10.0 g, 48.00 mmol) and N-isocyclopropylacetamide (9.50 g, 96.00 mmol) at room temperature, triethylamine
  • a toluene solution 150 mL of (10 mL)
  • the reaction mixture was stirred under reflux for 3 hours. After cooling to room temperature, the reaction solution was removed under reduced pressure.
  • the crude product was dissolved in 150 mL of dichloromethane and saturated.
  • Step 5 Synthesis of the intermediate compound 6-(2-chloropyrimidin-4-yl)-1-cyclopropyl-4-fluoro-2-methyl-1H-benzo[d]imidazole.
  • Step 2 Synthesis of the compound 5-bromo-3-fluoro-N 1 -isopropylamine benzene-1,2-diamine.
  • Step 5 Synthesis of the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • Glacial acetic acid (0.86 mL, 15.00 mmol) was added to 5-bromopyridine-2,3-diamine (2.82 g, 15.00 mmol) and acetone (870 mg, 15.00 mmol) in anhydrous dichloromethane (30 mL) In the solution, the reaction was stirred at room temperature for 5 hrs. After cooling to an ice bath, sodium triacetoxyborohydride (4.77 g, 22.50 mmol) was added portionwise, and the reaction mixture was warmed and allowed to react overnight at room temperature.
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 6-(2-chloropyrimidin-4-yl)-1-isopropyl-2-methyl-1H-imidazo[4,5-b]pyridine 510 mg of a white solid was obtained. The yield was 91.1%.
  • Step 4 Synthesis of the intermediate compound 6-(2-chloropyrimidin-4-yl)-1-isopropyl-2-methoxy-1H-imidazo[4,5-b]pyridine.
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 5-(2-chloropyrimidin-4-yl)-3-methylbenzo[d]oxazole-2(3H)-one (340 mg, white solid) was obtained. The yield was 75.9%.
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 6-(2-chloropyrimidin-4-yl)-4-fluoro-1-isopropyl-1H-indazole (447 mg, white solid) was obtained. The yield was 76.8%.
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 6-(2-chloropyrimidin-4-yl)-1-isopropyl-1H-indazole (290 mg, white solid) was obtained. The yield was 63.9%.
  • Pd(dppf)Cl 2 ⁇ CH 2 Cl 2 (250 mg, 0.31 mmol) was added to 6-bromo-8-fluoro-3-iodo-2-methylimidazo[1,2-a]pyridine under nitrogen. 1.17 g, 3.32 mmol), isopropenylboronic acid pinacol ester (557 mg, 3.31 mmol) and cesium carbonate (2.16 g, 6.64 mmol) in 1,4-dioxane (20 mL) and water (5 mL) mixed solvent The reaction was carried out at 80 ° C for 1 hour under a nitrogen atmosphere. The mixture was cooled to room temperature, filtered over Celite, and filtered, and the filtrate was washed with dichloromethane.
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 6-(2-chloropyrimidin-4-yl)-8-fluoro-2-methyl-3-(prop-1-en-2-yl)imidazo[1,2-a]pyridine 800 mg, White solid). The yield was 100%.
  • LC-MS (APCI): m/z 303.0 (M+1).
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 6-(2-chloropyrimidin-4-yl)-8-fluoro-2-methyl-imidazo[1,2-a]pyridine 630 mg, white solid was obtained. The yield was 80.00%.
  • LC-MS (APCI): m/z 263.0 (M-1).
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 5-(2-chloropyrimidin-4-yl)-7-fluoro-2-methylbenzo[d]oxazole 700 mg, white solid was obtained. The yield was 98.20%.
  • LC-MS (APCI): m/z 262.
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • 6-(2-Chloropyrimidin-4-yl)-4-fluoro-2-methylbenzo[d]oxazole 700 mg of a white solid was obtained. The yield was 98.20%.
  • LC-MS (APCI): m/z 266.
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 5-(2-chloropyrimidin-4-yl)-7-fluoro-benzo[d]oxazole 500 mg of a yellow-white solid was obtained. The yield was 56.2%.
  • LC-MS (APCI): m/z 250.1 (M+1).
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 6-(2-chloropyrimidin-4-yl)-1-isopropyl-2-(trifluoromethyl)-1H-benzo[d]imidazole (576 mg white solid) was obtained. The yield was 100%.
  • LC-MS (APCI): m/z 34.
  • Step 5 Synthesis of the intermediate compound 5-(2-chloropyrimidin-4-yl)-7-fluoro-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole.
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole. 600 mg of a yellowish white solid were obtained. The yield was 90.36%.
  • LC-MS (APCI): m/z 353.
  • Step 1 Synthesis of 5-bromo-7-fluoro-2-methyl-2H-indazole and 5-bromo-7-fluoro-1-methyl-1H-indazole.
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 5-(2-chloropyrimidin-4-yl)-7-fluoro-1-methyl-1H-indazole (295 mg of a white solid) was obtained. The yield was 75.6%.
  • LC-MS (APCI): m/z 263.0 (M-1).
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 5-(2-chloropyrimidin-4-yl)-7-fluoro-2-methyl-2H-indazole 180 mg yellow white solid was obtained. The yield was 83.5%.
  • LC-MS (APCI): m/z 266.
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 5-(2-chloropyrimidin-4-yl)-7-fluoro-2-methyl-3-(prop-1-en-2-yl)-2H-indazole 190 mg of a white solid was obtained. The yield was 95.3%.
  • LC-MS (APCI): m/z 302.9 (M+1).
  • Step 4 Synthesis of the intermediate compound 5-(2-chloropyrimidin-4-yl)-2-methyl-3-(prop-1-en-2-yl)-2H-indazole.
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 5-(2-chloropyrimidin-4-yl)-2-methyl-3-(prop-1-en-2-yl)-2H-indazole (460 mg of a white solid) was obtained. The yield was 92.0%.
  • LC-MS (APCI): m/z 285.0 (M+1).
  • Pd(dppf)Cl 2 .CH 2 Cl 2 (240 mg) was added to 6-chloro-3-iodoimidazo[1,2-b]pyridazine (1.0 g, 3.57 mmol), isopropenylboronic acid under nitrogen.
  • the mixture of pinacol ester (602 mg, 3.57 mmol) and sodium carbonate (800 mg, 7.16 mmol) in dioxane (16 mL) and water (4 mL) was reacted for one hour at 80 ° C under nitrogen atmosphere.
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 6-(2-chloropyrimidin-4-yl)imidazo[1,2-b]pyridazine (220 mg of a white solid) was obtained. The yield was 27.6%.
  • LC-MS (APCI): m/z 21.
  • the reduced iron powder (860 mg, 15.33 mmol) was added to 6-bromo-8-nitro-3,4-dihydroquinolin-1(2H)-carboxylic acid methyl ester (690 mg, 2.20 mmol) in glacial acetic acid (8 mL) ) in solution.
  • the reaction mixture was reacted at 80 ° C for 2.5 hrs, cooled to room temperature, and filtered with celite. The filtrate was diluted with water and evaporated with ethyl acetate (50 mL ⁇ 3).
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 8-(2-chloropyrimidin-4-yl)-1-isopropyl-5,6-dihydro-4H-imidazo[4,5,1-ij]quinolin-2(1H)-one was obtained. (300 mg white solid). The yield was 67.3%.
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 8-(2-chloropyrimidin-4-yl)-2-methoxy-5,6-dihydro-4H-imidazo[4,5,1-ij]quinoline 300 mg of a yellow solid) was obtained. The yield was 66.4%.
  • LC-MS (APCI): m/z 301.0 (M+1).
  • the synthesis step is the same as the intermediate compound 6-(2-chloropyrimidin-4-yl)-4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d]imidazole.
  • the product 6-(2-chloropyrimidin-4-yl)benzo[d]thiophene 120 mg of a yellow solid was obtained. The yield was 66.4%.
  • LC-MS (APCI): m/z 248.0 (M + 1).
  • Step 1 Synthesis of the compound (4-fluoro-2-methoxy-5-nitrophenyl)carbamic acid tert-butyl ester.
  • Boc anhydride (7.05 g, 32.20 mmol) was slowly added dropwise to 4-fluoro-2-methoxy-5-nitroaniline (4.0 g, 21.50 mmol) and triethylamine (TEA, 3.60 g).
  • TEA triethylamine
  • Petroleum ether / ethyl acetate (v / v) 2: 1) gave 3.6 g of a yellow solid, yield: 58.8%.
  • LC-MS (APCI): m/z 287.2 (M+1).
  • Step 2 Synthesis of the compound (4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxy-5-nitrophenyl)carbamic acid tert-butyl ester.
  • Step 3 Synthesis of the compound (5-amino-4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxyphenyl)carbamic acid tert-butyl ester.
  • Step 4 Compound (5-Acrylamide-4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxyphenyl) Synthesis of tert-butyl carbamate.
  • Step 5 Synthesis of the intermediate compound N-(5-amino-2-((2-(dimethylamino)ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide.
  • Step 4 Synthesis of intermediate compound N 1 -(2-(dimethylamino)ethyl)-5-ethoxy-N 1 -methyl-2-nitrobenzene-1,4-diamine
  • N-(6-chloro-2-methoxy-5-nitropyridin-3-yl)acetamide (1.00 g, 4.10 mmol) and N,N,N'-trimethylethylenediamine (630 mg, 6.20)
  • LC-MS (APCI): m/z 3121.
  • reaction solution was cooled to room temperature, poured into 50 mL of water, and extracted with ethyl acetate (5 mL*2). The organic phase was combined and evaporated to dryness and purified by preparative chromatography (PE/EA, V/V, 1.5/1) gave a white solid, a total of 118 mg, yield 31%.
  • Step 2 Compound 5-Chloro-4-(4-fluoro-1-isopropyl-2-methyl-1H-benzo[d]imidazol-6-yl)-N-(4-fluoro-2-methoxy Synthesis of 5--5-nitrophenyl)pyrimidine-2-amine.
  • Step 3 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((5-chloro-4-(4-fluoro-1-isopropyl-2) Synthesis of -methyl-1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • Step 1 Synthesis of the compound 6-(2-chloro-5-methylpyrimidin-4-yl)-4-fluoro-1-isopropyl-2-methyl-1H-benzo[d]imidazole.
  • reaction mixture was cooled to room temperature, poured into 50 mL of water, extracted with ethyl acetate (5 mL*2), and the organic phase was combined, and the solvent was evaporated to dryness under reduced pressure to purified by chromatography (PE/EA, V/V, 2 /1), a white solid was obtained in a total of 210 mg, yield 53.6%.
  • Step 2 Compound 4-(4-Fluoro-1-isopropyl-2-methyl-1H-benzo[d]imidazol-6-yl)-N-(4-fluoro-2-methoxy-5- Synthesis of nitrophenyl)-5-methylpyrimidin-2-amine.
  • Step 3 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((5-methyl-4-(4-fluoro-1-isopropyl)- Synthesis of 2-methyl-1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • the red oil was dissolved in ethanol-water (4 mL +1 mL), and reduced iron powder (106 mg) and ammonium chloride (34 mg) were added, and the mixture was heated under reflux at 90 ° C for 5 hours.
  • the reaction mixture was cooled to room temperature, and the insoluble solid was removed by filtration. The filtrate was evaporated to dryness.
  • the obtained oil was dissolved in 10 liters of EtOAc.
  • 0.15 M of acryloyl chloride (3 mL) was added dropwise, and the mixture was reacted for 30 minutes in an ice bath. The layers were separated, and the organic layer was evaporated.
  • Step 1 Synthesis of the compound 6-(2-chloro-5-methoxypyrimidin-4-yl)-4-fluoro-1-isopropyl-2-methyl-1H-benzo[d]imidazole.
  • the mixture was heated to dryness, and the reaction mixture was cooled to room temperature, poured into 50 mL of water, ethyl acetate (5mL*2), and the organic phase was combined, dried over anhydrous sodium sulfate and evaporated to dryness.
  • Step 2 Compound 4-(4-Fluoro-1-isopropyl-2-methyl-1H-benzo[d]imidazol-6-yl)-N-(4-fluoro-2-methoxy-5- Synthesis of nitrophenyl)-5-methoxypyrimidin-2-amine.
  • Step 3 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((5-methoxy-4-(4-fluoro-1-isopropyl) Synthesis of 2-methyl-1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • the red oil was dissolved in ethanol-water (4 mL +1 mL), reduced iron powder (178 mg) and ammonium chloride (57 mg) were added, and the mixture was refluxed at 90 ° C for 4.5 hours.
  • the reaction mixture was cooled to room temperature, and the insoluble solid was removed by filtration. The filtrate was evaporated to dryness.
  • the obtained oil was dissolved in 10 mL of DCM, and 2 mL of aq. To the above two-phase system, 1 M of acryloyl chloride (1 mL) was added dropwise, and the mixture was reacted for 30 minutes in an ice bath.
  • Step 1 Synthesis of the compound 4-chloro-N-(4-fluoro-2-methoxy-5-nitrophenyl)-5-(trifluoromethyl)pyrimidin-2-amine.
  • Step 2 Compound 4-(4-Fluoro-1-isopropyl-2-methyl-1H-benzo[d]imidazol-6-yl)-N-(4-fluoro-2-methoxy-5- Synthesis of nitrophenyl)-5-(trifluoromethyl)pyrimidin-2-amine.
  • Step 3 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((5-trifluoromethyl-4-(4-fluoro-1-isopropyl) Synthesis of benzyl-2-methyl-1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • the red oil was dissolved in ethanol-water (4 mL +1 mL), and reduced iron powder (50 mg) and ammonium chloride (17 mg) were added, and the mixture was refluxed at 90 ° C for 3 hours.
  • the reaction mixture was cooled to room temperature, and the insoluble solid was removed by filtration. The filtrate was evaporated to dryness.
  • the obtained oil was dissolved in 10 mL of DCM, and 2 mL of aq. 1 M acryloyl chloride (0.4 mL) was added dropwise to the above two-phase system, and the mixture was reacted for 30 minutes in an ice bath.
  • Step 4 Compound 5-Fluoro-1,1-dimethyl-7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- Synthesis of 2,3-dihydro-1H-benzo[d]pyrrolo[1,2-a]imidazole.
  • Step 5 Compound 7-(2-Chloropyrimidin-4-yl)-5-fluoro-1,1-dimethyl-2,3-dihydro-1H-benzo[d]pyrrolo[1,2-a Synthesis of imidazole.
  • the reaction system was cooled to room temperature, poured into 50 mL of water and extracted with EA (10 mL*2). The organic phases were combined, washed with water and brine, and dried over anhydrous sodium sulfate. The mixture was filtered under suction, and the filtrate was evaporated to dryness crystals crystals
  • Step 6 Compound 4-(5-Fluoro-1,1-dimethyl-2,3-dihydro-1H-benzo[d]pyrrolo[1,2-d]imidazol-7-yl)-N- Synthesis of (4-fluoro-2-methoxy-5-nitrophenyl)pyrimidine-2-amine.
  • the dioxane was evaporated to dryness, and 20 mL of DCM and 20 mL of saturated aqueous sodium hydrogen carbonate solution were added, and the mixture was stirred for 10 minutes, filtered, and separated. The organic phase was evaporated to dryness (mjjjjjjjjjj 414 mg).
  • Step 7 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-(5-fluoro-1,1-dimethyl-2,3) Synthesis of dihydro-1H-benzo[d]imidazo[1,2-a]pyrrolidin-6-ylpyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • the red oil was dissolved in ethanol-water (8 mL + 2 mL), and reduced iron powder (199 mg) and ammonium chloride (95 mg) were added, and the mixture was refluxed at 100 ° C for 4.5 hours.
  • the reaction mixture was cooled to room temperature, and the insoluble solid was removed by filtration. The filtrate was evaporated to dryness.
  • the obtained oil was dissolved in 10 mL of DCM, To the above two-phase system, 1 M of acryloyl chloride (1.7 mL) was added dropwise, and the mixture was reacted for 30 minutes in an ice bath. The mixture was separated, EtOAc (EtOAc m.
  • Step 1 Compound 4-(4-Fluoro-1-isopropyl-2-methoxy-1H-benzo[d]imidazol-6-yl)-N-(4-fluoro-2-methoxy-5 Synthesis of -nitrophenyl)pyrimidine-2-amine.
  • Step 2 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-(4-fluoro-1-isopropyl-2-methoxy) Synthesis of -1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • the red oil was dissolved in ethanol-water (8 mL + 2 mL), and reduced iron powder (112 mg) and ammonium chloride (37 mg) were added, and the mixture was refluxed at 100 ° C overnight. The reaction was cooled to room temperature, and the insoluble solid was removed by filtration. The filtrate was evaporated to dryness. The obtained oil was dissolved in 5 mL of DCM. 1 M acryloyl chloride (0.7 mL) was added dropwise to the above two-phase system, and the mixture was reacted for 30 minutes in an ice bath.
  • 2-Dimethylaminoethanol 130 mg, 1.456 mmol was added to dry dioxane (2.5 mL) to dissolve, then sodium hydrogen (58 mg, 1.456 mmol), 4-(4-fluoro-1-isopropyl) 2-methoxy-1H-benzo[d]imidazol-6-yl)-N-(4-fluoro-2-methoxy-5-nitrophenyl)pyrimidine-2-amine (137 mg, 0.291 Methyl), stirred at room temperature for 75 minutes. The reaction mixture was diluted with aq. EtOAc (EtOAc)EtOAc.
  • the red oil was dissolved in ethanol-water (10 mL + 2.5 mL), and reduced iron powder (81 mg) and ammonium chloride (32 mg) were added, and the mixture was refluxed at 80 ° C overnight. The reaction was cooled to room temperature, and the insoluble solid was removed by filtration. The filtrate was evaporated to dryness. The obtained oil was dissolved in 5 mL of DCM. 1 M acryloyl chloride (0.45 mL) was added dropwise to the above two-phase system, and the mixture was reacted for 30 minutes in an ice bath.
  • Step 1 Synthesis of the compound 6-(5-bromo-2-chloropyrimidin-4-yl)-4-fluoro-1-isopropyl-2-methyl-1H-benzo[d]imidazole.
  • Step 2 Compound 5-Bromo-4-(4-fluoro-1-isopropyl-2-methyl-1H-benzo[d]imidazol-6-yl)-N-(4-fluoro-2-methoxy Synthesis of 5--5-nitrophenyl)pyrimidine-2-amine.
  • Step 3 Compound N 1 -(5-Bromo-4-(4-fluoro-1-isopropyl-2-methyl-1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)-N 2-methoxy-4 -N ((dimethylamino) ethyl 2) - - synthesis of methyl-5-nitrobenzene-1,4-diamine 4.
  • Step 4 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((5-bromo-4-(4-fluoro-1-isopropyl-2) Synthesis of -methyl-1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • the obtained oil was dissolved in 10 mL of DCM, and 5 mL of aq. To the above two-phase system, 0.15 M of acryloyl chloride (2.5 mL) was added dropwise, and the mixture was reacted for 30 minutes in an ice bath.
  • Step 1 Compound 4-(4-Fluoro-1-isopropyl-2-methyl-1H-benzo[d]imidazol-6-yl)-N-(4-fluoro-2-methoxy-5- Synthesis of nitrophenyl)pyrimidine-2-amine.
  • Step 2 Compound N-(2-(2-Methoxy-ethyl-oxy)-5-((4-(4-fluoro-1-isopropyl-2-methyl-1H-benzo[d] Synthesis of imidazolium-6-ylpyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • the mixture was cooled to room temperature, filtered, and the filtrate was evaporated, evaporated, mjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjj
  • the mixture was cooled to room temperature, filtered, and the filtrate was evaporated, evaporated, mjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjj
  • Step 2 Compound N-(2-(4-Ethoxypiperidine)-1-yl)-5-((4-(4-fluoro-1-isopropyl-2-methyl-1H-benzo[ d] Synthesis of imidazolium-6-ylpyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • Step 2 Compound N-(2-(4-Methoxypiperidine)-1-yl)-5-((4-(4-fluoro-1-isopropyl-2-methyl-1H-benzo[ d] Synthesis of imidazolium-6-ylpyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • Step 2 Compound 4-(4-Fluoro-1-isopropyl-2-methyl-1H-benzo[d]imidazol-6-yl)-2-(methylsulfonyl)pyrimidine-5-carbonitrile synthesis.
  • Step 3 Compound 2 - ((4-((2-(Dimethylamino)ethyl)(methyl)amino)-2-methoxy-5-nitrophenyl)amino)-4-(4- Synthesis of fluoro-1-isopropyl-2-methyl-1H-benzo[d]imidazol-6-ylpyrimidine-5-carbonitrile.
  • Step 4 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((5-cyano-4-(4-fluoro-1-isopropyl)- Synthesis of 2-methyl-1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • Step 1 Compound N 1 -(4-(1-cyclopropyl-4-fluoro-2-methyl-1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)-N 4 -(2 Synthesis of -(dimethylamino)ethyl)-2-methoxy-N 4 -methyl-5-nitrobenzene-1,4-diamine.
  • Step 2 Compound N 4 -(4-(1-cyclopropyl-4-fluoro-2-methyl-1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)-N 1 -(2 Synthesis of -(dimethylamino)ethyl)-5-methoxy-N 1 -methylbenzene-1,2,4-triamine
  • Step 3 Compound N-(5-((4-(1-cyclopropyl-4-fluoro-2-methyl-1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)amino)- Synthesis of 2-((2-(Dimethylamino)ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide
  • Step 1 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d Synthesis of imidazolium-6-yl)pyrimidin-2-yl)-5-methoxy-N 1 -methyl-2-nitrobenzene-1,4-diamine.
  • Step 2 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(4-fluoro-2-(fluoromethyl)-1-isopropyl-1H-benzo[d] Synthesis of imidazolium-6-ylpyrimidin-2-yl)-5-methoxy-N 1 -methylbenzene-1,2,4-triamine
  • Step 3 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-(4-fluoro-2-(fluoromethyl))-1-) Synthesis of propyl-1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide
  • Step 1 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(1-isopropyl-2-methyl-1H-imidazo[4,5-b]pyridine- Synthesis of 6-yl)pyrimidin-2-yl)-5-methoxy-N 1 -methyl-2-nitrobenzene-1,4-diamine.
  • Step 2 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(1-isopropyl-2-methyl-1H-imidazo[4,5-b]pyridine- Synthesis of 6-yl)pyrimidin-2-yl)-5-methoxy-N 1 -methylbenzene-1,2,4-triamine.
  • Step 3 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-(1-isopropyl-2-methyl-1H-imidazo) Synthesis of [4,5-b]pyridin-6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • Step 1 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(1-isopropyl-2-methoxy-1H-imidazo[4,5-b]pyridine Synthesis of -6-ylpyrimidin-2-yl)-5-methoxy-N 1 -methyl-2-nitrobenzene-1,4-diamine.
  • Step 2 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(1-isopropyl-2-methoxy-1H-imidazo[4,5-b]pyridine Synthesis of -6-ylpyrimidin-2-yl)-5-methoxy-N 1 -methylbenzene-1,2,4-triamine
  • Step 3 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-(1-isopropyl-2-methoxy-1H-imidazole) Synthesis of [4,5-b]pyridin-6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide
  • Step 1 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(1-isopropyl-2-methoxy-1H-benzo[4,5-b]imidazole Synthesis of -6-ylpyrimidin-2-yl)-5-methoxy-N 1 -methyl-2-nitrobenzene-1,4-diamine.
  • Step 2 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(1-isopropyl-2-methoxy-1H-benzo[4,5-b]imidazole Synthesis of -6-ylpyrimidin-2-yl)-5-methoxy-N 1 -methylbenzene-1,2,4-triamine
  • Step 3 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-(1-isopropyl-2-methoxy-1H-benzene) And [d]imidazole-6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide
  • Step 1 Compound 5-(2-((4-(2-(Dimethylamino)ethyl)(methyl)amino)-2-methoxy-5-nitrophenyl)amino)pyrimidine-4- Synthesis of 3-methylbenzo[d]oxazole-2(3H)-one.
  • Step 2 Compound 5-(2-((5-Amino-4-(4-(dimethylamino)ethyl)(methyl)amino)-2-methoxyphenyl)amino)pyrimidine-4- Synthesis of 3-methylbenzo[d]oxazole-2(3H)-one
  • Step 3 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-4-methoxy-5-((4-(3-methyl-2-oxo)) Synthesis of -2,3-dihydrobenzo[d]oxazol-5-yl)pyrimidin-2-yl)amino)phenyl)acrylamide
  • Step 1 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(4-fluoro-1-isopropyl-1H-indazol-6-yl)pyrimidin-2-yl Synthesis of 5-methoxy-N 1 -methyl-2-nitrobenzene-1,4-diamine.
  • Step 2 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(4-fluoro-1-isopropyl-1H-indazol-6-yl)pyrimidin-2-yl Synthesis of 5-methoxy-N 1 -methylbenzene-1,2,4-triamine
  • Step 3 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-(4-fluoro-1-isopropyl-1H-indazole-) Synthesis of 6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • Step 1 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(1-isopropyl-1H-indazol-6-yl)pyrimidin-2-yl)-5- Synthesis of methoxy-N 1 -methyl-2-nitrobenzene-1,4-diamine.
  • Step 2 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(4-fluoro-1-isopropyl-1H-indazol-6-yl)pyrimidin-2-yl Synthesis of 5-methoxy-N 1 -methylbenzene-1,2,4-triamine.
  • Step 3 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-(1-isopropyl-1H-indazol-6-yl)) Synthesis of pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • Step 1 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(8-fluoro-2-methyl-3-(prop-1-en-2-yl)imidazolium Synthesis of [1,2-a]pyridin-6-yl)pyrimidin-2-yl)-5-methoxy-N 1 -methyl-2-nitrobenzene-1,4-diamine.
  • Step 2 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(8-fluoro-2-methyl-3-(prop-1-en-2-yl)imidazolium Synthesis of [1,2-a]pyridin-6-yl)pyrimidin-2-yl)-5-methoxy-N 1 -methylbenzene-1,2,4-triamine
  • Step 3 Compound N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-(8-fluoro-3-isopropyl-2-methylimidazole) Synthesis of [1,2-a]pyridin-6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide
  • Step 1 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(8-fluoro-2-methyl-imidazo[1,2-a]pyridine-6-yl) Synthesis of pyrimidin-2-yl)-5-methoxy-N 1 -methyl-2-nitrobenzene-1,4-diamine.
  • Step 2 Compound N 1 -(2-(Dimethylamino)ethyl)-N 4 -(4-(8-fluoro-2-methyl-imidazo[1,2-a]pyridin-6-yl) Synthesis of pyrimidin-2-yl)-5-methoxy-N 1 -methylbenzene-1,2,4-triamine.
  • Step 3 Compound N-(2-((2-(Dimethylamino)ethyl))(methyl)amino)-5-((4-(8-fluoro-2-methylimidazo[1,2- Synthesis of a]pyridin-6-yl)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide.
  • Step 1 Compound N 1 -(4-(4-(tert-Butoxy)-1-isopropyl-2-methyl-1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)- Synthesis of N 4 -(2-(dimethylamino)ethyl)-2-methoxy-N 4 -methyl-5-nitrobenzene-1,4-diamine.
  • Step 2 Compound N 4 -(4-(4-tert-Butoxy)-1-isopropyl-2-methyl-1H-benzo[d]imidazol-6-yl)pyrimidin-2-yl)-N Synthesis of 1- (2-(dimethylamino)ethyl)-5-methoxy-N 1 -methylbenzene 1,2,4-triamine.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

一种对蛋白酪氨酸激酶具有抑制作用的氨基嘧啶类化合物,包含它们的药物组合物,以及它们的制备和用途。具体地,一种式(I)所示的氨基嘧啶类化合物,其中R1、R2、L、Y、R6、W、A、m 和n 如说明书中所定义,及其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素变体。所述化合物可用于治疗和/或预防蛋白酪氨酸激酶相关性疾病,例如细胞增殖性疾病、癌症、免疫性疾病。

Description

用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物 技术领域
本发明属于医药领域。具体地,本发明涉及对蛋白酪氨酸激酶具有抑制作用的氨基嘧啶类化合物,包含它们的药物组合物,以及它们的制备方法和用途。
背景技术
表皮生长因子受体(即EGFR、ErbB-1或HER1)是ErbB受体家族的成员之一,ErbB受体家族包括四种密切相关的受体酪氨酸激酶成员:EGFR(ErbB-1)、HER2/c-neu(ErbB-2)、Her3(ErbB-3)和Her4(ErbB-4)。EGFR是胞外蛋白配体的表皮生长因子家族(EGF家族)成员的细胞表面受体。影响EGFR表达或活性的突变可能导致癌症。据报道,在大多数实体瘤如肺癌、乳腺癌和脑瘤中,EGFR处于失调状态。据估计,30%的上皮癌与EGFR或家族成员的突变、扩增或失调有关联。
已经研发出基于通过抗体药或小分子抑制剂药物(例如吉非替尼和厄洛替尼)抑制EGFR的治疗方法。在非小细胞肺癌(NSCLC)的情况下,吉非替尼和厄洛替尼对10%~40%的病人有益处。然而,治疗一段时间后,对吉非替尼或厄洛替尼的获得性耐药性成为主要的临床问题。研究证实,产生耐药性的一个主要原因是由于T790M的新突变,其为EGFR的“门卫”。然后,研发人员又研发了针对T790M的抑制剂,如BIBW2992,并在临床试验中表现出优势。但是,这些以EGFR的T790M突变为靶标的抑制剂对野生型EGFR也具有相当的抑制活性,这导致的严重的毒副作用限制了其临床应用。所以,有必要进一步研发出更多仅靶向突变型而非野生型的EGFR的选择性抑制剂的有效类型。
针对EGFR突变型的晚期非小细胞肺癌,虽然吉非替尼、厄洛替尼等EGFR激酶抑制剂(EGFR-TKI)取得了令人瞩目的疗效,但是随后发现EGFR-TKI在治疗非小细胞肺癌时的原发性耐药或继发性耐药,是我们在治疗晚期非小细胞肺癌面临新的挑战,继而有必要开展新的探索,寻找对策。
第三代及后续的EGFR抑制剂包括AZD9291和CO-1686等化合物,其不可逆抑制EGFR,并且对具有T790M耐药突变的患者有较高的有效率,但是对野生型EGFR仍存在抑制作用。
因此,有必要进一步研发新的EGFR抑制剂,其不仅能有效抑制T790M突变,而且相对于野生型而言对T790M突变具有高选择性。
发明内容
本发明提供了一种新的氨基嘧啶类化合物及包含该化合物的组合物及其用途,其具有更好的EGFR激酶抑制活性,以及对于耐药突变T790M、L858R及其二者的高选择性,可用于治疗、预防以及缓解EGFR激酶介导的疾病。
对此,本发明采用的技术方案如下:
本发明的第一方面中,提供了式(I)化合物:
Figure PCTCN2017094102-appb-000001
其中,
R1和R2独立地选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷氧基、任选取代的C1-C6烷硫基、任选取代的C1-C6烷氨基或任选取代的C1-C6烷酰基;
L选自键、NR7、O、C(R7)2或S;其中各R7独立地选自H、任选取代的C1-C6烷基例如被C1-C6烷氧基取代的C1-C6烷基,或任选取代的C1-C6烷氧基;
Y为H,或者Y选自以下结构:
Figure PCTCN2017094102-appb-000002
其中R3、R4和R5独立地选自H、卤素、-CN、任选取代的C1-C6烷基或任选取代的C1-C6卤代烷基;
R6选自H、C1-C6烷基、C2-C6炔基或3至8元杂环基,其中上述基团任选被1-3个以下取代基取代:卤素、-OH、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基、-C(O)R8或NR8R9;其中R8和R9各自独立地选自H、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷基或任选取代的C3-C6碳环基,或者R8和R9与它们相连的氮原子一起形成任选取代的4至6元杂环基,例如被C1-C6烷基取代;
W选自键、C(R10)2、NR10、O或S,其中R10选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基或任选取代的C1-C6烷氧基,只要化学上允许;
m为0、1或2;
n为0、1或2;
环A选自以下结构:
Figure PCTCN2017094102-appb-000003
其中各X1独立地选自C(R11)2、NR11、O或S,各X2和X3独立地选自C(R11)2或NR11,其中X1和X2为可与母核连接的位置;
环B选自5元碳环基、5元杂环基或5元杂芳基,其任选地被1-3个R11取代基取代;
R11独立地选自H、卤素、-CN、-NO2、-OH、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C2-C6烯基、任选取代的C2-C6炔基、任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基,只要化学上允许;或者,其中两个相邻的R11可一起形成任选取代的C5-C8碳环基、任选取代的5至8元杂环基、任选取代的C6-C14芳基或任选取代的5至10元杂芳基;
或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
该方面的优选实施方案为上述的化合物,其为式(II)化合物:
Figure PCTCN2017094102-appb-000004
其中R1、R2、L、R6、W、A、m和n如上所定义,
或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
该方面的优选实施方案为上述的化合物,其为式(III)化合物:
Figure PCTCN2017094102-appb-000005
其中R1、R2、L、Y、R6、A和n如上所定义,
或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
该方面的优选实施方案为上述的化合物,其为式(IV)化合物:
Figure PCTCN2017094102-appb-000006
其中R1、R2、L、R6、A和n如上所定义,
或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
该方面的优选实施方案为上述的化合物,其中
环A选自以下结构:
Figure PCTCN2017094102-appb-000007
其中环A任选被1-6个R11取代基取代;
优选地,环A选自以下结构:
Figure PCTCN2017094102-appb-000008
其中环A任选被1-6个R11取代基取代;
优选地,环A选自以下结构:
Figure PCTCN2017094102-appb-000009
其中环A任选被1-6个R11取代基取代,其中各X4独立地选自C(R11)2、NR11、O、S、C(O)、S(O)或S(O)2,各X5独立地选自C(R11)2或NR11
优选地,环A选自以下结构:
Figure PCTCN2017094102-appb-000010
其中环A任选被1-6个R11取代基取代,其中各X4独立地选自C(R11)2、NR11、O、S、C(O)、S(O)或S(O)2,各X5独立地选自C(R11)2或NR11原子;
优选地,环A选自以下基团:
Figure PCTCN2017094102-appb-000011
Figure PCTCN2017094102-appb-000012
Figure PCTCN2017094102-appb-000013
优选地,环A选自以下基团:
Figure PCTCN2017094102-appb-000014
Figure PCTCN2017094102-appb-000015
更优选地,环A选自以下基团:
Figure PCTCN2017094102-appb-000016
其中,只要化学上允许,R11独立地选自H、卤素、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C2-6烯基、任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;优选地,R11独立地选自H、卤素、任选取代的C1-C6烷基或任选取代的C2-6烯基;更优选地,R11独立地选自H、-F、甲基、异丙基或1-丙烯-2-基;或者,其中两个相邻的R11可一起形成任选取代的C5-C8碳环基、任选取代的5至8元杂环基、任选取代的C6-C14芳基或任选取代的5至10元杂芳基;
最优选地,环A选自以下基团:
Figure PCTCN2017094102-appb-000017
该方面的优选实施方案为上述的化合物,其中
Y选自以下结构:
Figure PCTCN2017094102-appb-000018
Y选自以下结构:
Figure PCTCN2017094102-appb-000019
优选地,Y选自以下结构:
Figure PCTCN2017094102-appb-000020
其中R3、R4和R5独立地选自H、卤素、任选取代的C1-C6烷基或任选取代的C1-C6卤代烷基;优选地,R3、R4和R5独立地选自H或卤素;更优选地,R3、R4和R5均为 H。
该方面的优选实施方案为上述的化合物,其中m为1或2;优选地,m为1。
该方面的优选实施方案为上述的化合物,其中R1选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷氧基、任选取代的C1-C6烷硫基、任选取代的C1-C6烷氨基或任选取代的C1-C6烷酰基;优选地,R1选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;优选地,R1选自H或卤素;更优选地,R1选自H或F。
该方面的优选实施方案为上述的化合物,其中R2选自H、卤素、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷氧基、任选取代的C1-C6烷硫基、任选取代的C1-C6烷氨基或任选取代的C1-C6烷酰基;优选地,R2选自H、卤素、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;优选地,R2选自H、卤素、任选取代的C1-C6烷基或任选取代的C1-C6烷氧基;更优选地,R2选自H或任选取代的C1-C6烷氧基;更优选地,R2选自甲氧基。
该方面的优选实施方案为上述的化合物,其中
R6选自C1-C6烷基或4至7元杂环基;更优选地,R6选自C1-C6烷基或5至6元杂环基;更优选地,R6选自乙基或6元杂环基;更优选地,R6选自乙基或含有2个N杂原子的6元杂环基;更优选地,R6选自乙基或哌嗪基;
其中R6任选被以下取代基取代:卤素、-OH、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基或NR8R9;优选地,R6任选被以下取代基取代:卤素、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基或NR8R9;更优选地,R6任选被以下取代基取代:任选取代的C1-C6烷基或NR8R9
其中R8和R9各自独立地选自任选取代的C1-C6烷基、任选取代的C1-C6烷氧基或任选取代的C3-C6碳环基;优选地,R8和R9各自独立地选自任选取代的C1-C6烷基;更优选地,R8和R9均为甲基;
或者R8和R9与它们相连的氮原子一起形成任选取代的4至6元杂环基,例如被C1-C6烷基取代。
该方面的优选实施方案为上述的化合物,其中n为0或1。
该方面的优选实施方案为上述的化合物,其中L选自NR7、O、C(R7)2或S;优选地,L选自NR7、O或S;更优选地,L为NR7;其中R7选自H、任选取代的C1-C6烷基或任选取代的C1-C6烷氧基;优选地,R7选自H或任选取代的C1-C6烷基;更优选地, R7为任选取代的C1-C6烷基;更优选地,R7为甲基。
该方面的优选实施方案为上述的化合物,其中基团-(L)n-R6为N(Me)CH2CH2N(Me)2或选自以下结构:
Figure PCTCN2017094102-appb-000021
该方面的优选实施方案为上述的化合物,其中
W选自C(R10)2、NR10、O或S;优选地,W选自NR10、O或S;更优选地,W为NR10
只要化学上允许,R10选自H、卤素、-CN、-NO2、任选取代的C1-6烷基、任选取代的C1-6卤代烷基或任选取代的C1-6烷氧基;优选地,R10选自H、任选取代的C1-6烷基、任选取代的C1-6卤代烷基或任选取代的C1-6烷氧基;更优选地,R10选自H或任选取代的C1-6烷基;更优选地,R10为H。
该方面的优选实施方案为上述的化合物,其中R1选自H或F;R2为甲氧基;-(L)n-R6 选自N(Me)CH2CH2N(Me)2或4-甲基哌嗪-1-基;且若存在时L为
Figure PCTCN2017094102-appb-000022
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。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、其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物,和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。
在另一个方面,本发明提供了在需要其的受试者中治疗EGFR导致的癌症(包括EGFR突变导致的癌症,例如,带有T790M突变、L858R突变和L858R/T790M双突变的癌症)相关病症的方法,所述方法包括:给予受试者有效量的本发明化合物。在具体实施方案中,所述EGFR导致的癌症选自:非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、乳腺癌、***癌、肝癌、皮肤癌、上皮细胞癌、胃肠间质瘤、白血病、组织细胞性淋巴癌、鼻咽癌等。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。
由随后的具体实施方式、实施例和权利要求,本发明的其它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“C1-C6烷基”包括C1、C2、C3、C4、C5、C6、C1-C6、C1-C5、C1-C4、C1-C3、C1-C2、C2-C6、C2-C5、C2-C4、C2-C3、C3-C6、C3-C5、C3-C4、C4-C6、C4-C5和C5-C6烷基。
应该理解,当本文描述时,任何下面所定义的基团可以被许多取代基取代,而且相应的定义在下面列出的它们的范围内,包括取代的基团。除非另作说明,否则,术语“取代”如下面所定义。
“C1-C6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,C1-C4烷基是特别优选的。所述烷基的实例包括但不限于: 甲基(C1)、乙基(C2)、正丙基(C3)、异丙基(C3)、正丁基(C4)、叔丁基(C4)、仲丁基(C4)、异丁基(C4)、正戊基(C5)、3-戊基(C5)、戊基(C5)、新戊基(C5)、3-甲基-2-丁基(C5)、叔戊基(C5)和正己基(C6)。除非另作说明,否则,烷基的每个独立地任选被取代,即,未取代(“未取代的烷基”)或被一个或多个取代基取代(“取代的烷基”);例如,1至5个取代基、1至3个取代基或1个取代基。在一些实施方案中,烷基是未取代的C1-C6烷基(例如,-CH3)。在一些实施方案中,烷基是取代的C1-C6烷基。
“C2-C6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,C2-4烯基是特别优选的。所述烯基的实例包括但不限于:乙烯基(C2)、1-丙烯基(C3)、2-丙烯基(C3)、1-丙烯-2-基(C3)、1-丁烯基(C4)、2-丁烯基(C4)、丁二烯基(C4)、戊烯基(C5)、戊二烯基(C5)、己烯基(C6),等等。除非另作说明,否则,烯基的每个独立地任选被取代,即,未取代(“未取代的烯基”)或被一个或多个取代基取代(“取代的烯基”);例如,1至5个取代基、1至3个取代基或1个取代基。在一些实施方案中,烯基是未取代的C2-6烯基。在一些实施方案中,烯基是取代的C2-6烯基。
“C2-C6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,C2-4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(C2)、1-丙炔基(C3)、2-丙炔基(C3)、1-丁炔基(C4)、2-丁炔基(C4)、戊炔基(C5)、3-甲基丁-1-炔基(C5)、己炔基(C6),等等。除非另作说明,否则,炔基的每个独立地任选被取代,即,未取代(“未取代的炔基”)或被一个或多个取代基取代(“取代的炔基”);例如,1至5个取代基、1至3个取代基或1个取代基。在一些实施方案中,炔基是未取代的C2-6炔基。在一些实施方案中,炔基是取代的C2-6炔基。
“C1-C6烷氧基”是指基团-OR,其中,R为取代或未取代的C1-C6烷基。在一些实施方案中,C1-C4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。
“C1-C6烷硫基”是指基团-SR,其中,R为任选取代的C1-C6烷基。在一些实施方案中,C1-C4烷硫基是特别优选的。具体的所述C1-C6烷硫基包括但不限于:甲硫基、乙硫基、正丙硫基、异丙硫基、正丁硫基、叔丁硫基、仲丁硫基、正戊硫基、正己硫基 和1,2-二甲基丁硫基。
“C1-C6烷氨基”是指基团-NHR或者-NR2,其中,R为任选取代的C1-C6烷基。在一些实施方案中,C1-C4烷氨基是特别优选的。具体的所述C1-C6烷氨基包括但不限于:甲氨基、乙氨基、正丙氨基、异丙氨基、正丁氨基、叔丁氨基、二甲氨基、甲乙氨基和二乙氨基。
“C1-C6烷酰基”是指基团-(=O)R,其中,R为任选取代的C1-C6烷基。在一些实施方案中,C1-C4烷酰基是特别优选的。示例性的所述C1-C6烷酰基包括但不限于:-(=O)CH3、-(=O)CH2CH3、-(=O)CH2CH2CH3和-(=O)CH(CH3)2
“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,“C1-C6卤代烷基”和“C1-C6卤代烷氧基”是指上述“C1-C6烷基”和“C1-C6烷氧基”,其被一个或多个卤素基团取代。在一些实施方案中,C1-C4卤代烷基是特别优选的,更优选C1-C2卤代烷基。在一些实施方案中,C1-C4卤代烷氧基是特别优选的,更优选C1-C2卤代烷氧基。示例性的所述卤代烷基包括但不限于:-CF3、-CH2F、-CHF2、-CHFCH2F、-CH2CHF2、-CF2CF3、-CCl3、-CH2Cl、-CHCl2、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:-OCH2F、-OCHF2、-OCF3,等等。
“C3-C8碳环基”是指具有3至8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,优选C5-C8碳环基,具有5至8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,优选C3-C6碳环基,具有3至6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,优选5元碳环基,其为具有5个环碳原子和零个杂原子的非芳香环烃基团。碳环基还包括其中上述碳环基环与一个或多个芳基或杂芳基稠合的环体系,其中连接点在碳环基环上,且在这样的情况中,碳的数目继续表示碳环基体系中的碳的数目。示例性的所述碳环基包括但不限于:环丙基(C3)、环丙烯基(C3)、环丁基(C4)、环丁烯基(C4)、环戊基(C5)、环戊烯基(C5)、环戊二烯基(C5)、环己基(C6)、环己烯基(C6)、环已二烯基(C6),以及环庚基(C7)、环庚烯基(C7)、环庚二烯基(C7)、环庚三烯基(C7)、环辛基(C8)、环辛烯基(C8)、二环[2.2.1]庚基(C7)、二环[2.2.2]辛基(C8),等等。除非另作说明,否则碳环基的每个独立地为任选取代的,即,未取代(“未取代的碳环基”)或被一个或多个取代基取代(“取代的碳环基”)。在一些实施方案中,碳环基是未取代的C3-C8碳环基。在一些实施方案中,碳环基是取代的C3-C8碳环基。
“3至8元杂环基”是指具有环碳原子和1至3个环杂原子的3至8元非芳香环系,其中每个杂原子独立地选自氮、氧、硫、硼、磷和硅,其中碳、氮、硫和磷原子还可 以其氧化态存在,如C(O)、S(O)、S(O)2、P(O)等。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,优选4至7元杂环基,其为具有环碳原子和1至3个环杂原子的4至7元非芳香环系。在一些实施方案中,优选5至8元杂环基,其为具有环碳原子和1至3个环杂原子的5至8元非芳香环系。在一些实施方案中,优选4至6元杂环基,其为具有环碳原子和1至3个环杂原子的4至6元非芳香环系。在一些实施方案中,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。在一些实施方案中,更优选5元杂环基,其为具有环碳原子和1至3个环杂原子的5元非芳香环系。在一些实施方案中,上述3至8元杂环基、4至7元杂环基、5至8元杂环基、4至6元杂环基、5至6元杂环基和5元杂环基含有1至3个(更优选1或2个)选自氮、氧和硫(优选氮或氧)的环杂原子。除非另作说明,否则,杂环基的每个独立地为任选取代的,即,未取代(“未取代的杂环基”)或被一个或多个取代基取代(“取代的杂环基”)。在一些实施方案中,杂环基是未取代的3-8元杂环基。在一些实施方案中,杂环基是取代的3-8元杂环基。杂环基还包括其中上述杂环基环与一个或多个碳环基稠合的环体系,其中连接点在碳环基环上,或其中上述杂环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。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。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、氧硫杂环戊烯基(1,2-氧硫杂环戊烯基、1,3-氧硫杂环戊烯基)、二硫杂环戊烷基、二氢吡唑基、二氢咪唑基、二氢噻唑基、二氢异噻唑基、二氢噁唑基、二氢异噁唑基、二氢噁二唑基和噁唑烷-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:***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、四氢吡啶基和硫杂环己烷基。示例性的包含两个杂原子的6元杂环基包括但不限于:二氢吡嗪基、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。示例性的含有一个或两个杂原子的7元杂环基包括但不限于:氮杂环庚烷基、二氮杂环庚烷基、氧杂环庚烷基和硫杂环庚烷基。示例性的包含一个或两个杂原子的8元杂环基包括但不限于:氮杂环辛烷基、氧杂环辛烷基、硫杂环辛烷基、八氢环戊二烯并[c]吡咯基和八氢吡咯并[3,4-c]吡咯基。示例性的与 C6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C6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。
“C6-C14芳基”是指具有提供在芳族环系中的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(“C6-14芳基”)。在一些实施方案中,芳基具有六个环碳原子(“C6芳基”;例如,苯基)。在一些实施方案中,芳基具有十个环碳原子(“C10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(“C14芳基”;例如,蒽基)。“C6-C14芳基”还包括这样的环***,在这种环***中,上述芳基环与一个或多个碳环基或杂环基稠合,其中,原子团或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***中的碳原子数目。典型的芳基包括但不限于衍生自以下的基团:醋蒽烯、苊烯、醋菲烯(acephenanthrylene)、蒽、薁、苯、屈、晕苯、荧蒽、芴、并六苯、己芬、己搭烯、不对称引达省、对称引达省、茚满、茚、萘、并八苯、辛芬、辛搭烯、卵苯、戊-2,4-二烯、并五苯、戊搭烯、戊芬、苝、非那烯、菲、苉、七曜烯、芘、皮蒽、玉红省、苯并菲和联三萘。具体地说,芳基包括苯基、萘基、茚基和四氢萘基。除非另作说明,否则,芳基的每个独立地任选被取代,即,未取代(“未取代的芳基”)或被一个或多个取代基取代(“取代的芳基”)。在一些实施方案中,芳基是未取代的C6-14芳基。在一些实施方案中,芳基是取代的C6-14芳基。
“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。在一些实施方案中,优选5元杂芳基,其为具有环碳原子和1-4个环杂原子的5元单环4n+2芳族环体系(例如,具有以环状排列共享的6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***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碳环基或杂环基稠合的环***,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***中的碳原子数目。除非另作说明,否则,杂芳基的每个独立地任选被取代的,即,未取代(“未取代的杂芳基”)或被一个或多个取代基取代(“取代的杂芳基”)。在一些实施方案中,杂芳基是未取代的5至10元杂芳基。在一些实施方案中,杂芳基是取代的5至10元杂芳基。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:***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***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。
本文定义的烷基、烯基、炔基、碳环基、杂环基、芳基和杂芳基等为任选取代的基团。通常,术语“取代的”,不论前面是否有术语“任选”,是指存在于基团(例如,碳或氮原子)上的至少一个氢被可允许的取代基取代,例如,在取代时产生稳定的化合物的取代基,例如,不自发地进行转变(例如通过重排、环化、消除或其它反应)的化合物。除非另外说明,否则,“取代的”基团在所述基团的一个或多个可取代的位置处具有取代基,且当在任何给定结构中的一个以上的位置被取代时,在每个位置处的取代基是相同或不同的。术语“取代的”包括用有机化合物的所有可允许的取代基(导致形成稳定化合物的本文描述的任何取代基)进行的取代。对于本发明,杂原子例如氮可具有氢取代基和/或本文描述的任何合适的取代基,其满足杂原子的化合价且导致形成稳定的部分。
示例性的碳原子上的取代基包括但不局限于:卤素、-CN、-NO2、-N3、-SO2H、-SO3H、-OH、-ORaa、-ON(Rbb)2、-N(Rbb)2、-N(Rbb)3 +X-、-N(ORcc)Rbb、-SH、-SRaa、-SSRcc、-C(=O)Raa、-CO2H、-CHO、-C(ORcc)2、-CO2Raa、-OC(=O)Raa、-OCO2Raa、-C(=O)N(Rbb)2、-OC(=O)N(Rbb)2、-NRbbC(=O)Raa、-NRbbCO2Raa、-NRbbC(=O)N(Rbb)2、-C(=NRbb)Raa、-C(=NRbb)ORaa、-OC(=NRbb)Raa、-OC(=NRbb)ORaa、-C(=NRbb)N(Rbb)2、-OC(=NRbb)N(Rbb)2、-NRbbC(=NRbb)N(Rbb)2、-C(=O)NRbbSO2Raa、-NRbbSO2Raa、-SO2N(Rbb)2、-SO2Raa、-SO2ORaa、-OSO2Raa、-S(=O)Raa、-OS(=O)Raa、-Si(Raa)3、-OSi(Raa)3、-C(=S)N(Rbb)2、-C(=O)SRaa、-C(=S)SRaa、-SC(=S)SRaa、-SC(=O)SRaa、-OC(=O)SRaa、-SC(=O)ORaa、-SC(=O)Raa、-P(=O)2Raa、-OP(=O)2Raa、-P(=O)(Raa)2、-OP(=O)(Raa)2、-OP(=O)(ORcc)2、-P(=O)2N(Rbb)2、-OP(=O)2N(Rbb)2、-P(=O)(NRbb)2、-OP(=O)(NRbb)2、-NRbbP(=O)(ORcc)2、 -NRbbP(=O)(NRbb)2、-P(Rcc)2、-P(Rcc)3、-OP(Rcc)2、-OP(Rcc)3、-B(Raa)2、-B(ORcc)2、-BRaa(ORcc)、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Rdd基团取代;
或者在碳原子上的两个偕氢被基团=O、=S、=NN(Rbb)2、=NNRbbC(=O)Raa、=NNRbbC(=O)ORaa、=NNRbbS(=O)2Raa、=NRbb或=NORcc取代;
Raa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Raa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Rdd基团取代;
Rbb的每个独立地选自:氢、-OH、-ORaa、-N(Rcc)2、-CN、-C(=O)Raa、-C(=O)N(Rcc)2、-CO2Raa、-SO2Raa、-C(=NRcc)ORaa、-C(=NRcc)N(Rcc)2、-SO2N(Rcc)2、-SO2Rcc、-SO2ORcc、-SORaa、-C(=S)N(Rcc)2、-C(=O)SRcc、-C(=S)SRcc、-P(=O)2Raa、-P(=O)(Raa)2、-P(=O)2N(Rcc)2、-P(=O)(NRcc)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Rbb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Rdd基团取代;
Rcc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Rcc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Rdd基团取代;
Rdd的每个独立地选自:卤素、-CN、-NO2、-N3、-SO2H、-SO3H、-OH、-ORee、-ON(Rff)2、-N(Rff)2,、-N(Rff)3 +X-、-N(ORee)Rff、-SH、-SRee、-SSRee、-C(=O)Ree、-CO2H、-CO2Ree、-OC(=O)Ree、-OCO2Ree、-C(=O)N(Rff)2、-OC(=O)N(Rff)2、-NRffC(=O)Ree、-NRffCO2Ree、-NRffC(=O)N(Rff)2、-C(=NRff)ORee、-OC(=NRff)Ree、-OC(=NRff)ORee、-C(=NRff)N(Rff)2、-OC(=NRff)N(Rff)2、-NRffC(=NRff)N(Rff)2、-NRffSO2Ree、-SO2N(Rff)2、-SO2Ree、-SO2ORee、-OSO2Ree、-S(=O)Ree、-Si(Ree)3、-OSi(Ree)3、-C(=S)N(Rff)2、-C(=O)SRee、-C(=S)SRee、-SC(=S)SRee、-P(=O)2Ree、-P(=O)(Ree)2、-OP(=O)(Ree)2、-OP(=O)(ORee)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Rgg基团取代,或者两个偕Rdd取代基可结合以形成=O或=S;
Ree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Rgg基团取代;
Rff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Rff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Rgg基团取代;
Rgg的每个独立地是:卤素、-CN、-NO2、-N3、-SO2H、-SO3H、-OH、-OC1-6烷基、-ON(C1-6烷基)2、-N(C1-6烷基)2、-N(C1-6烷基)3 +X-、-NH(C1-6烷基)2 +X-、-NH2(C1-6烷基)+X-、-NH3 +X-、-N(OC1-6烷基)(C1-6烷基)、-N(OH)(C1-6烷基)、-NH(OH)、-SH、-SC1-6烷基、-SS(C1-6烷基)、-C(=O)(C1-6烷基)、-CO2H、-CO2(C1-6烷基)、-OC(=O)(C1-6烷基)、-OCO2(C1-6烷基)、-C(=O)NH2、-C(=O)N(C1-6烷基)2、-OC(=O)NH(C1-6烷基)、-NHC(=O)(C1-6烷基)、-N(C1-6烷基)C(=O)(C1-6烷基)、-NHCO2(C1-6烷基)、-NHC(=O)N(C1-6烷基)2、-NHC(=O)NH(C1-6烷基)、-NHC(=O)NH2、-C(=NH)O(C1-6烷基)、-OC(=NH)(C1-6烷基)、-OC(=NH)OC1-6烷基、-C(=NH)N(C1-6烷基)2、-C(=NH)NH(C1-6烷基)、-C(=NH)NH2、-OC(=NH)N(C1-6烷基)2、-OC(NH)NH(C1-6烷基)、-OC(NH)NH2、-NHC(NH)N(C1-6烷基)2、-NHC(=NH)NH2、-NHSO2(C1-6烷基)、-SO2N(C1-6烷基)2、-SO2NH(C1-6烷基)、-SO2NH2、-SO2C1-6烷基、-SO2OC1-6烷基、-OSO2C1-6烷基、-SOC1-6烷基、-Si(C1-6烷基)3、-OSi(C1-6烷基)3、-C(=S)N(C1-6烷基)2、C(=S)NH(C1-6烷基)、C(=S)NH2、-C(=O)S(C1-6烷基)、-C(=S)SC1-6烷基、-SC(=S)SC1-6烷基、-P(=O)2(C1-6烷基)、-P(=O)(C1-6烷基)2、-OP(=O)(C1-6烷基)2、-OP(=O)(OC1-6烷基)2、C1-6烷基、C1-6全卤代烷基、C2-6烯基、C2-6炔基、C3-10碳环基、C6-10芳基、3-10元杂环基、5-10元杂芳基;或者两个偕Rgg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、-ORaa、-N(Rcc)2、-CN、-C(=O)Raa、-C(=O)N(Rcc)2、-CO2Raa、-SO2Raa、-C(=NRbb)Raa、-C(=NRcc)ORaa、-C(=NRcc)N(Rcc)2、-SO2N(Rcc)2、-SO2Rcc、-SO2ORcc、-SORaa、-C(=S)N(Rcc)2、-C(=O)SRcc、-C(=S)SRcc、-P(=O)2Raa、-P(=O)(Raa)2、-P(=O)2N(Rcc)2、-P(=O)(NRcc)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Rcc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Rdd基团取代,且其中Raa、Rbb、Rcc和Rdd如上所述。
其它定义
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、***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无机和有机酸和碱的盐。药学上可接受的无毒酸加成盐的实例是氨基与无机酸形成的盐,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,或使用本领域使用的方法形成的盐,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+(C1-4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,进一步的药学上可接受的盐包括使用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。
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。
“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”)。
通常,化合物的“有效量”是指足以引起目标生物反应的数量。正如本领域普通技术人员所理解的那样,本发明化合物的有效量可以根据下列因素而改变:例如,生物学目标、化合物的药物动力学、所治疗的疾病、给药模式以及受试者的年龄健康情况和症状。有效量包括治疗和预防性治疗有效量。
除非另作说明,否则,本文使用的化合物的“治疗有效量”是在治疗疾病、障碍或病症的过程中足以提供治疗益处的数量,或使与疾病、障碍或病症有关的一或多种症 状延迟或最小化。化合物的治疗有效量是指单独使用或与其它疗法联用的治疗剂的数量,它在治疗疾病、障碍或病症的过程中提供治疗益处。术语“治疗有效量”可以包括改善总体治疗、降低或避免疾病或病症的症状或病因、或增强其它治疗剂的治疗效能的数量。
除非另作说明,否则,本文使用的化合物的“预防有效量”是足以预防疾病、障碍或病症的数量,或足以预防与疾病、障碍或病症有关的一或多种症状的数量,或防止疾病、障碍或病症复发的数量。化合物的预防有效量是指单独使用或与其它药剂联用的治疗剂的数量,它在预防疾病、障碍或病症的过程中提供预防益处。术语“预防有效量”可以包括改善总体预防的数量,或增强其它预防药剂的预防效能的数量。
“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。
“EGFR导致的癌症”是指以EGFR基因的不适合的高表达为特征或以改变EGFR核酸分子或多肽的生物活性的EGFR基因突变为特征的癌症。EGFR导致的癌症可出现在任何组织(包括脑、血液、***、肝、口、肌肉、脾、胃、睾丸和气管)中。EGFR导致的癌症包括但不限于非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、乳腺癌、***癌、肝癌、皮肤癌、上皮细胞癌、胃肠间质瘤、白血病、组织细胞性淋巴癌、鼻咽癌。
“EGFR突变”或“EGFR突变体”包括EGFR蛋白或EGFR编码序列的氨基酸或核苷酸序列中一个或多个缺失、置换或添加。EGFR突变还可以包括一个或多个缺失、置换或添加,或其片段,只要该突变体相对于野生型EGFR保留或增加了酪氨酸激酶活性。在具体的EGFR突变中,激酶或磷酸化活性相对于野生型EGFR可以增加或减少(例如,至少5%、10%、15%、20%、30%、40%、50%、60%、70%、80%、90%或甚至100%)。示例性的EGFR突变包括但不限于T790M突变、L858R突变和L858R/T790M双突变。
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)、(II)、(III)、(III-a)、(III-b)和(IV)的化合物其药学上可接受的盐。所述(I)、(II)、(III)、(III-a)、(III-b)、(IV)和(IV-a)的化合物其药学上可接受的盐还包括其任何可能的立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
在一个实施方案中,本发明涉及式(I)化合物:
Figure PCTCN2017094102-appb-000023
其中,
R1选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷氧基、任选取代的C3-C6碳环基、任选取代的C1-C6烷硫基、任选取代的C1-C6烷氨基或任选取代的C1-C6烷酰基;
R2选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷氧基、任选取代的C1-C6烷硫基、任选取代的C1-C6烷氨基或任选取代的C1-C6烷酰基;
L选自键、NR7、O、C(R7)2或S;其中各R7独立地选自H、任选取代的C1-C6烷基例如被C1-C6烷氧基取代的C1-C6烷基,或任选取代的C1-C6烷氧基;
Y为H,或者Y选自以下结构:
Figure PCTCN2017094102-appb-000024
其中R3、R4和R5独立地选自H、卤素、-CN、任选取代的C1-C6烷基或任选取代的C1-C6卤代烷基;
R6选自H、C1-C6烷基、C2-C6炔基或3至8元杂环基,其中上述基团任选被1-3个以下取代基取代:卤素、-OH、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基、-C(O)R8或NR8R9;其中R8和R9各自独立地选自H、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷基或任选取代的C3-C6碳环基,或者R8和R9与它们相连的氮原子一起形成任选取代的4至6元杂环基,例如被C1-C6烷基取代;
W选自键、C(R10)2、NR10、O或S,其中R10选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基或任选取代的C1-C6烷氧基,只要化学上允许;
m为0、1或2;
n为0、1或2;
环A选自以下结构:
Figure PCTCN2017094102-appb-000025
其中各X1独立地选自C(R11)2、NR11、O或S,各X2和X3独立地选自C(R11)2或NR11,其中X1和X2为可与母核连接的位置;
环B选自5元碳环基、5元杂环基或5元杂芳基,其任选地被1-3个R11取代基取代;
R11独立地选自H、卤素、-CN、-NO2、-OH、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C2-C6烯基、任选取代的C2-C6炔基、任选取代的C1-C6烷氧基、任选取代的C3-C6碳环基或任选取代的C1-C6卤代烷氧基,只要化学上允许;或者,其中两个相邻的R11可一起形成任选取代的C5-C8碳环基、任选取代的5至8元杂环基、任选取代的C6-C14芳基或任选取代的5至10元杂芳基;
或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
在该实施方案中,优选地,R1选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷氧基、任选取代的C3-C6碳环基、任选取代的C1-C6烷硫基、任选取代的C1-C6烷氨基或任选取代的C1-C6烷酰基;优选地,R1选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷氧基、任选取代的C1-C6烷硫基、任选取代的C1-C6烷氨基或任选取代的C1-C6烷酰基;优选地,R1选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷氧基或任选取代的C3-C6碳环基;优选地,R1选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;更优选地,R1选自H、卤素、-CN、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基或任选取代的C3-C6碳环基;更优选地,R1选自H或卤素;更优选地,R1选自H、-F、-Br、-Cl、-CN、甲基、三氟甲基、甲氧基或环丙基;更优选地,R1选自H、-Br、-Cl、甲基或环丙基;更优选地,R1选自H或F。
在该实施方案中,优选地,R2选自H、卤素、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷氧基、任选取代的C3-C6碳环基、任选取代的C1-C6烷硫基、任选取代的C1-C6烷氨基或任选取代的C1-C6烷酰基;优选地,R2选自H、卤素、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷氧基、任选取代的C1-C6 烷硫基、任选取代的C1-C6烷氨基或任选取代的C1-C6烷酰基;优选地,R2选自H、卤素、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;优选地,R2选自H、卤素、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;优选地,R2选自H、卤素、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;优选地,R2选自H、卤素、任选取代的C1-C6烷基或任选取代的C1-C6烷氧基;更优选地,R2选自H、任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;更优选地,R2选自H或任选取代的C1-C6烷氧基;更优选地,R2选自-OCH3、-OCH2CH3、-OCHF2、-OCH2CF3;更优选地,R2选自甲氧基。
在该实施方案中,优选地,R6选自C1-C6烷基、C2-C6炔基或3至8元杂环基;优选地,R6选自C1-C6烷基或4至7元杂环基;更优选地,R6选自C1-C6烷基或5至6元杂环基;更优选地,R6选自乙基或6元杂环基;更优选地,R6选自乙基或含有1个或2个N杂原子的6元杂环基;更优选地,R6选自乙基或含有2个N杂原子的6元杂环基;更优选地,R6选自乙基、哌啶基、吗啉基或哌嗪基;更优选地,R6选自乙基或哌嗪基。
在R6的上述实施方案中,R6任选被以下取代基取代:卤素、-OH、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基或NR8R9;优选地,R6任选被以下取代基取代:卤素、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基或NR8R9;更优选地,R6任选被以下取代基取代:任选取代的C1-C6烷基、任选取代的C1-C6烷氧基或NR8R9;更优选地,R6任选被以下取代基取代:任选取代的C1-C6烷基或NR8R9
其中R8和R9各自独立地选自任选取代的C1-C6烷基、任选取代的C1-C6烷氧基或任选取代的C3-C6碳环基;优选地,R8和R9各自独立地选自任选取代的C1-C6烷基;更优选地,R8和R9均为甲基。或者R8和R9与它们相连的氮原子一起形成任选取代的4至6元杂环基,例如被C1-C6烷基或C1-C6烷氧基取代;或者R8和R9与它们相连的氮原子一起形成任选取代的4至6元杂环基,例如被C1-C6烷基取代;更优选地,R8和R9与它们相连的氮原子一起形成含有1个N杂原子,或2个N杂原子,或1个N杂原子和1个O杂原子的4至6元杂环基,其任选地被C1-C6烷基或C1-C6烷氧基取代;更优选地,R8和R9与它们相连的氮原子一起形成含有1个N杂原子,或2个N杂原子,或1个N杂原子和1个O杂原子的4至6元杂环基,其任选地被C1-C6烷基取代。
在该实施方案中,优选地,L选自NR7、O、C(R7)2或S;优选地,L选自NR7、O或S;更优选地,L为NR7
在L的上述实施方案中,R7选自H、任选取代的C1-C6烷基或任选取代的C1-C6 烷氧基;优选地,R7选自H或任选取代的C1-C6烷基;更优选地,R7为任选取代的C1-C6烷基;更优选地,R7为甲基。
在该实施方案中,优选地,环A选自以下结构:
Figure PCTCN2017094102-appb-000026
其中环A任选被1-6个R11取代基取代;
优选地,环A选自以下结构:
Figure PCTCN2017094102-appb-000027
Figure PCTCN2017094102-appb-000028
其中环A任选被1-6个R11取代基取代;
优选地,环A选自以下结构:
Figure PCTCN2017094102-appb-000029
其中环A任选被1-6个R11取代基取代;
优选地,环A选自以下结构:
Figure PCTCN2017094102-appb-000030
Figure PCTCN2017094102-appb-000031
其中环A任选被1-6个R11取代基取代,其中各X4独立地选自C(R11)2、NR11、O、S、C(O)、S(O)或S(O)2,各X5独立地选自C(R11)2或NR11
优选地,环A选自以下结构:
Figure PCTCN2017094102-appb-000032
Figure PCTCN2017094102-appb-000033
其中环A任选被1-6个R11取代基取代,其中各X4独立地选自C(R11)2、NR11、O、S、C(O)、S(O)或S(O)2,各X5独立地选自C(R11)2或NR11
优选地,环A选自以下结构:
Figure PCTCN2017094102-appb-000034
Figure PCTCN2017094102-appb-000035
其中环A任选被1-6个R11取代基取代,其中各X4独立 地选自C(R11)2、NR11、O、S、C(O)、S(O)或S(O)2,各X5独立地选自C(R11)2或NR11
优选地,环A选自以下基团:
Figure PCTCN2017094102-appb-000036
Figure PCTCN2017094102-appb-000037
Figure PCTCN2017094102-appb-000038
优选地,环A选自以下基团:
Figure PCTCN2017094102-appb-000039
Figure PCTCN2017094102-appb-000040
优选地,环A选自以下基团:
Figure PCTCN2017094102-appb-000041
优选地,环A选自以下基团:
Figure PCTCN2017094102-appb-000042
更优选地,环A选自以下基团:
Figure PCTCN2017094102-appb-000043
在环A的上述实施方案中,只要化学上允许,R11独立地选自H、卤素、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C2-6烯基、任选取代的C1-C6 烷氧基、任选取代的C3-C6碳环基或任选取代的C1-C6卤代烷氧基;优选地,R11独立地选自H、卤素、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C2-6烯基、任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;优选地,R11独立地选自H、卤素、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C2-C6烯基、任选取代的C1-C6烷氧基或任选取代的C3-C6碳环基;优选地,R11独立地选自H、卤素、任选取代的C1-C6烷基或任选取代的C2-6烯基;更优选地,R11独立地选自H、-F、甲基、异丙基、三氟甲基、1-丙烯-2-基、甲氧基、叔丁氧基、环丙基;更优选地,R11独立地选自H、-F、甲基、异丙基或1-丙烯-2-基;或者,其中两个相邻的R11可一起形成任选取代的C5-C8碳环基、任选取代的5至8元杂环基、任选取代的C6-C14芳基或任选取代的5至10元杂芳基。
最优选地,环A选自以下基团:
Figure PCTCN2017094102-appb-000044
Figure PCTCN2017094102-appb-000045
最优选地,环A选自以下基团:
Figure PCTCN2017094102-appb-000046
在该实施方案中,优选地,Y为H;优选地,Y选自以下结构:
Figure PCTCN2017094102-appb-000047
更优选地,Y选自以下结构:
Figure PCTCN2017094102-appb-000048
在Y的上述实施方案中,R3、R4和R5独立地选自H、卤素、任选取代的C1-C6烷基或任选取代的C1-C6卤代烷基;优选地,R3、R4和R5独立地选自H或卤素;更优选地,R3、R4和R5均为H。
在该实施方案中,优选地,W选自C(R10)2、NR10、O或S;优选地,W选自NR10、O或S;更优选地,W为NR10
在W的上述实施方案中,只要化学上允许,R10选自H、卤素、-CN、-NO2、任选取代的C1-6烷基、任选取代的C1-6卤代烷基或任选取代的C1-6烷氧基;优选地,R10选自H、任选取代的C1-6烷基、任选取代的C1-6卤代烷基或任选取代的C1-6烷氧基;更优选地,R10选自H或任选取代的C1-6烷基;更优选地,R10为H。
在该实施方案中,优选地,m为1或2;优选地,m为1;更优选地,m为0。
在该实施方案中,优选地,n为0;优选地,n为1。
在该实施方案中,优选地,
L选自键、NR7、O或S;其中R7选自H或任选取代的C1-C6烷基;
n为0、1或2;
R6选自H、C1-C6烷基或3至8元杂环基,其中上述基团任选被1-3个以下取代基取代:卤素、-OH、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基或NR8R9,其中R8和R9各自独立地选自H或任选取代的C1-C6烷基;或者R8和R9与它们相连的氮原子一起形成任选取代的4至6元杂环基;
在该实施方案中,优选地,
L选自键或NR7;其中R7选自H或任选取代的C1-C6烷基;
n为0或1;
R6选自H、C1-C6烷基或3至8元杂环基,其中上述基团任选被1-3个以下取代基取代:卤素、-OH、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基或NR8R9,其中R8和R9各自独立地选自H或任选取代的C1-C6烷基;或者R8和R9与它们相连的氮原子一起形成任选取代的4至6元杂环基;
在该实施方案中,优选地,
-(L)n-R6选自:
Figure PCTCN2017094102-appb-000049
优选地,-(L)n-R6选自:
Figure PCTCN2017094102-appb-000050
优选地,-(L)n-R6选自:
Figure PCTCN2017094102-appb-000051
Figure PCTCN2017094102-appb-000052
优选地,-(L)n-R6选自:
Figure PCTCN2017094102-appb-000053
在另一个实施方案中,式(I)化合物为以下式(II)化合物:
Figure PCTCN2017094102-appb-000054
其中R1、R2、L、R6、W、A、m和n如上文所定义。
在另一个实施方案中,式(I)化合物为以下式(III)化合物:
Figure PCTCN2017094102-appb-000055
其中
Y选自以下结构:
Figure PCTCN2017094102-appb-000056
优选地,Y选自以下结构:
Figure PCTCN2017094102-appb-000057
优选地,Y为以下结构:
Figure PCTCN2017094102-appb-000058
优选地,Y为以下结构:
Figure PCTCN2017094102-appb-000059
并且R1-R6、L、A和n如上文所定义。
在另一个实施方案中,式(I)化合物为以下式(IV)化合物:
Figure PCTCN2017094102-appb-000060
其中R1、R2、L、R6、A和n如上文所定义。
在另一个具体实施方案中,本发明涉及上述化合物,其中
R1选自H、卤素、-CN、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基或任选取代的C3-C6碳环基;
R2选自任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;
-(L)n-R6选自:
Figure PCTCN2017094102-appb-000061
Figure PCTCN2017094102-appb-000062
环A选自:
Figure PCTCN2017094102-appb-000063
其中,R11独立地选自H、卤素、任选取代的C1-C6烷基、任选取代的C2-C6烯基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基或任选取代的C3-C6碳环基;或者,其中两个相邻的R11可一起形成任选取代的5至8元杂环基;
或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
在另一个具体实施方案中,本发明涉及上述化合物,其中
环A为
Figure PCTCN2017094102-appb-000064
-(L)n-R6选自:
Figure PCTCN2017094102-appb-000065
在另一个具体实施方案中,本发明涉及上述化合物,其中
-(L)n-R6
Figure PCTCN2017094102-appb-000066
环A选自:
Figure PCTCN2017094102-appb-000067
在另一个具体实施方案中,本发明涉及式(III-a)化合物:
Figure PCTCN2017094102-appb-000068
其中,
R1选自H、卤素、-CN、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基或任选取代的C1-C6烷氧基;优选地,R1选自H、卤素或任选取代的C1-C6烷基;优选地,R1选自H、-F、-Cl、-Br、-CN、-CH3、-CF3或-OCH3;优选地,R1选自H、-Cl、-Br或-CH3
R2选自任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;优选地,R2选自-OCH3、-OCH2CH3、-OCHF2或-OCH2CF3
R11选自H、卤素、任选取代的C1-C6烷基、任选取代的C2-C6烯基、任选取代的C1-C6卤代烷基或任选取代的C1-C6烷氧基;优选地,R11选自H、卤素或任选取代的C1-C6烷氧基;优选地,R11选自H、-F、-Cl、-Br、-Me、-iPr、1-丙烯-2-基、-CH2F、CF3、-OMe或-OC(CH3)3;优选地,R11选自H、-F、-Cl、-Br、-OMe或-OC(CH3)3
R12选自H、卤素、任选取代的C1-C6烷基、任选取代的C2-C6烯基、任选取代的 C1-C6卤代烷基或任选取代的C1-C6烷氧基;优选地,R12选自任选取代的C1-C6烷基或任选取代的C1-C6卤代烷基;优选地,R12选自H、-F、-Cl、-Br、-Me、-iPr、1-丙烯-2-基、-CH2F、CF3、-OMe或-OC(CH3)3;优选地,R12选自-Me、-iPr或-CH2F;
R13选自H、卤素、任选取代的C1-C6烷基、任选取代的C2-C6烯基、任选取代的C1-C6卤代烷基或任选取代的C1-C6烷氧基;优选地,R13为任选取代的C1-C6烷基;优选地,R13选自H、-F、-Cl、-Br、-Me、-iPr、1-丙烯-2-基、-CH2F、CF3、-OMe或-OC(CH3)3;优选地,R13选自-Me或-iPr;
或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
在优选的上述具体实施方案中,R1为H,且R2和R11-R13如上定义。
在优选的上述具体实施方案中,R1为卤素(例如-F、-Cl或-Br,优选-Cl或-Br),且R2和R11-R13如上定义。
在优选的上述具体实施方案中,R1为-CN,且R2和R11-R13如上定义。
在优选的上述具体实施方案中,R1为任选取代的C1-C6烷基(例如-CH3),且R2和R11-R13如上定义。
在优选的上述具体实施方案中,R1为任选取代的C1-C6卤代烷基(例如-CF3),且R2和R11-R13如上定义。
在优选的上述具体实施方案中,R1为任选取代的C1-C6烷氧基(例如-OCH3),且R2和R11-R13如上定义。
在另一个具体实施方案中,本发明涉及式(III-a)化合物:
Figure PCTCN2017094102-appb-000069
其中,
R1为任选取代的C3-C6碳环基;优选地,R1为-环丙烷基;
且R2和R11-R13如上定义。
或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
在优选的上述具体实施方案中,R2为任选取代的C1-C6烷氧基(例如-OCH3或-OCH2CH3),且R1和R11-R13如上定义。
在优选的上述具体实施方案中,R2为任选取代的C1-C6卤代烷氧基(例如-OCHF2或-OCH2CF3),且R1和R11-R13如上定义。
在优选的上述具体实施方案中,R11为H,且R1、R2、R12和R13如上定义。
在优选的上述具体实施方案中,R11为卤素(例如-F、-Cl或-Br),且R1、R2、R12和R13如上定义。
在优选的上述具体实施方案中,R11为任选取代的C1-C6烷基(例如-Me或-iPr),且R1、R2、R12和R13如上定义。
在优选的上述具体实施方案中,R11为任选取代的C2-C6烯基(例如1-丙烯-2-基),且R1、R2、R12和R13如上定义。
在优选的上述具体实施方案中,R11为任选取代的C1-C6卤代烷基(例如-CH2F或-CF3),且R1、R2、R12和R13如上定义。
在优选的上述具体实施方案中,R11为任选取代的C1-C6烷氧基(例如-OMe或-OC(CH3)3),且R1、R2、R12和R13如上定义。
在优选的上述具体实施方案中,R12为H,且R1、R2、R11和R13如上定义。
在优选的上述具体实施方案中,R12为卤素(例如-F、-Cl或-Br),且R1、R2、R11和R13如上定义。
在优选的上述具体实施方案中,R12为任选取代的C1-C6烷基(例如-Me或-iPr),且R1、R2、R11和R13如上定义。
在优选的上述具体实施方案中,R12为任选取代的C2-C6烯基(例如1-丙烯-2-基),且R1、R2、R11和R13如上定义。
在优选的上述具体实施方案中,R12为任选取代的C1-C6卤代烷基(例如-CH2F或-CF3,优选-CH2F),且R1、R2、R11和R13如上定义。
在优选的上述具体实施方案中,R12为任选取代的C1-C6烷氧基(例如-OMe或-OC(CH3)3),且R1、R2、R11和R13如上定义。
在优选的上述具体实施方案中,R13为H,且R1、R2、R11和R12如上定义。
在优选的上述具体实施方案中,R13为卤素(例如-F、-Cl或-Br),且R1、R2、R11和R12如上定义。
在优选的上述具体实施方案中,R13为任选取代的C1-C6烷基(例如-Me或-iPr),且R1、R2、R11和R12如上定义。
在优选的上述具体实施方案中,R13为任选取代的C2-C6烯基(例如1-丙烯-2-基), 且R1、R2、R11和R12如上定义。
在优选的上述具体实施方案中,R13为任选取代的C1-C6卤代烷基(例如-CH2F或-CF3),且R1、R2、R11和R12如上定义。
在优选的上述具体实施方案中,R13为任选取代的C1-C6烷氧基(例如-OMe或-OC(CH3)3),且R1、R2、R11和R12如上定义。
在另一个具体实施方案中,本发明涉及上述式(III-b)化合物:
Figure PCTCN2017094102-appb-000070
其中,
R1选自H、卤素、-CN、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基或任选取代的C1-C6烷氧基;优选地,R1选自H、卤素或任选取代的C1-C6烷基;优选地,R1选自H、-F、-Cl、-Br、-CN、-CH3、-CF3或-OCH3;优选地,R1选自H、-Cl、-Br或-CH3
R2选自任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;优选地,R2选自-OCH3、-OCH2CH3、-OCHF2或-OCH2CF3
R11选自H、卤素、任选取代的C1-C6烷基、任选取代的C2-C6烯基、任选取代的C1-C6卤代烷基或任选取代的C1-C6烷氧基;优选地,R11选自H、卤素或任选取代的C1-C6烷氧基;优选地,R11选自H、-F、-Cl、-Br、-Me、-iPr、1-丙烯-2-基、-CH2F、CF3、-OMe或-OC(CH3)3;优选地,R11选自H、-F、-Cl、-Br、-OMe或-OC(CH3)3
在另一个具体实施方案中,本发明涉及上述式(III-b)化合物:
Figure PCTCN2017094102-appb-000071
其中,
R1为任选取代的C3-C6碳环基;优选地,R1为-环丙烷基;
R2和R11如上所定义;
或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
在另一个具体实施方案中,本发明涉及上述式(IV-a)化合物:
Figure PCTCN2017094102-appb-000072
其中,
R1选自H、卤素、-CN、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基或任选取代的C1-C6烷氧基;优选地,R1选自H或-F;
R2选自任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;优选地,R2选自H或-OCH3
A选自
Figure PCTCN2017094102-appb-000073
或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
在最优选的实施方案中,本发明化合物为以下化合物:
Figure PCTCN2017094102-appb-000074
Figure PCTCN2017094102-appb-000075
Figure PCTCN2017094102-appb-000076
Figure PCTCN2017094102-appb-000077
Figure PCTCN2017094102-appb-000078
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。
本发明也包括本发明化合物的所有合适的同位素衍生物。本发明化合物的同位素衍生物的定义为其中至少一个原子被具有相同原子数但原子质量与自然界中通常发现的原子质量不同的原子代替。可以掺入本发明化合物中的同位素的实例包括氢、碳、氮、氧、氟和氯的同位素,例如分别为2H、3H、13C、14C、15N、17O、18O、18F、31P、32P、35S和36Cl。本发明化合物的一些同位素衍生物,例如其中掺入放射性同位素如3H或14C的那些,可用于药物和/或底物组织分布研究。氚化(即3H)和碳-14(即14C)同位素因其易于制备和可检测性而为特别优选的。此外,用同位素(例如氘,即2H)取代可以提供由于更大的代谢稳定性而得到的一些治疗优点,例如增加的体内半衰期或减少 的剂量需要,并且因此在一些情况下可以是优选的。本发明化合物的同位素衍生物通常可以通过如下制备:常规程序例如通过说明性方法或通过下文实施例中描述的制备,其使用合适试剂的适当同位素衍生物。
本发明化合物或其药学上可接受的盐可以是无定形或结晶形式。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”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。
本领域技术人员将理解,许多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。
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载体,当将这种前药给予患者时,其在体内释放式(I)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、胺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、胺或巯基。因此,前药的代表性实例包括(但不限于)式(I)化合物的醇、巯基和胺官能团的乙酸酯、甲酸酯和苯甲酸酯衍生物。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调配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。
下列制剂实施例说明可根据本发明制备的代表性的药物组合物。然而,本发明不限于下列药物组合物。
示例性的制剂1-片剂:可以将干粉形式的本发明化合物与干燥的凝胶粘合剂以约1:2的重量比混合。加入较少量的硬脂酸镁作为润滑剂。使该混合物在压片机中成型为0.3-30mg片剂(每个片剂含有0.1-10mg活性化合物)。
示例性的制剂2-片剂:可以将干粉形式的本发明化合物与干燥的凝胶粘合剂以约1:2的重量比混合。加入较少量的硬脂酸镁作为润滑剂。使该混合物在压片机中成型为30-90mg片剂(每个片剂含有10-30mg活性化合物)。
示例性的制剂3-片剂:可以将干粉形式的本发明化合物与干燥的凝胶粘合剂以约1:2的重量比混合。加入较少量的硬脂酸镁作为润滑剂。使该混合物在压片机中成型为90-150mg片剂(每个片剂含有30-50mg活性化合物)。
示例性的制剂4-片剂:可以将干粉形式的本发明化合物与干燥的凝胶粘合剂以约1:2的重量比混合。加入较少量的硬脂酸镁作为润滑剂。使该混合物在压片机中成型为150-240mg片剂(每个片剂含有50-80mg活性化合物)。
示例性的制剂5-片剂:可以将干粉形式的本发明化合物与干燥的凝胶粘合剂以约1:2的重量比混合。加入较少量的硬脂酸镁作为润滑剂。使该混合物在压片机中成型为 240-270mg片剂(每个片剂含有80-90mg活性化合物)。
示例性的制剂6-片剂:可以将干粉形式的本发明化合物与干燥的凝胶粘合剂以约1:2的重量比混合。加入较少量的硬脂酸镁作为润滑剂。使该混合物在压片机中成型为270-450mg片剂(每个片剂含有90-150mg活性化合物)。
示例性的制剂7-片剂:可以将干粉形式的本发明化合物与干燥的凝胶粘合剂以约1:2的重量比混合。加入较少量的硬脂酸镁作为润滑剂。使该混合物在压片机中成型为450-900mg片剂(每个片剂含有150-300mg活性化合物)。
示例性的制剂8-胶囊剂:可以将干粉形式的本发明化合物与淀粉稀释剂以约1:1的重量比混合。将该混合物填充到250mg胶囊中(每个胶囊含有125mg活性化合物)。
示例性的制剂9-液体:可以将本发明化合物(125mg)与蔗糖(1.75g)和黄原胶(4mg)混合,且可将得到的混合物共混,通过No.10筛目美国筛,然后与预先制备的微晶纤维素和羧甲基纤维素钠(11:89,50mg)的水溶液混合。将苯甲酸钠(10mg)、调味剂和着色剂用水稀释,并在搅拌下加入。然后,可以加入充足的水,得到5mL的总体积。
示例性的制剂10-注射剂:可以将本发明化合物溶解或悬浮在缓冲无菌盐水可注射的水性介质中,达到约5mg/mL的浓度。
给药
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、***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提高至有效水平。推注剂量取决于通过身体的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***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%)。
组合
本发明化合物或其组合物可以与其它治疗剂组合给药,以治疗所述疾病。已知治疗剂的实例包括但不限于阿德力霉素(Adriamycin)、***(dexamethasone)、长春新碱(vincristine)、环磷酰胺(cyclophosphamide)、氟尿嘧啶(fluorouracil)、拓朴替康(topotecan)、紫杉酚(taxol)、干扰素、铂衍生物、紫杉烷(taxane)(例如太平洋紫杉醇(paclitaxel))、长春花生物碱(例如长春碱(vinblastine))、蒽环霉素(anthracycline)(例如多柔比星(doxorubicin))、表鬼臼毒素(epipodophyllotoxin)(例如依托泊苷(etoposide))、顺铂 (cisplatin)、mTOR抑制剂(例如雷帕霉素(rapamycin))、甲氨蝶呤(methotrexate)、放线菌素D(actinomycin D)、海兔毒素10(dolastatin 10)、秋水仙碱(colchicine)、吐根素(emetine)、三甲曲沙(trimetrexate)、氯苯氨啶(metoprine)、环孢霉素(cyclosporine)、道诺霉素(daunorubicin)、替尼泊苷(teniposide)、两性霉素(amphotericin)、烷化剂(例如苯丁酸氮芥(chlorambucil))、5-氟尿嘧啶、喜树碱(camptothecin)、顺铂、甲硝哒唑(metronidazole)以及GleevecTM。在其它实施例中,本发明化合物与如阿瓦斯汀(Avastin)或维克替比(VECTIBIX)等生物剂组合给药。
在一些实施方案中,本发明化合物或其组合物可以与选自以下中的任一种或一种以上的抗增殖剂或化学治疗剂组合给药:阿巴瑞克(abarelix)、阿地白介素(aldesleukin)、阿仑单抗(alemtuzumab)、阿利维甲酸(alitretinoin)、别嘌呤醇(allopurinol)、六甲蜜胺(altretamine)、氨磷汀(amifostine)、阿那曲唑(anastrozole)、三氧化二砷、天冬酰胺酶、阿扎胞苷(azacitidine)、BCG Live、贝伐单抗(bevacuzimab)、氟尿嘧啶、贝瑟罗汀(bexarotene)、博莱霉素(bleomycin)、硼替佐米(bortezomib)、白消安(busulfan)、二***(calusterone)、卡培他滨(capecitabine)、喜树碱、卡铂(carboplatin)、卡莫司汀(carmustine)、塞内昔布(celecoxib)、西妥昔单抗(cetuximab)、苯丁酸氮芥(chlorambucil)、克拉屈滨(cladribine)、氯法拉滨(clofarabine)、环磷酰胺、阿糖胞苷(cytarabine)、放线菌素D、达贝泊汀α(darbepoetin alfa)、道诺霉素、地尼白介素(denileukin)、右雷佐生(dexrazoxane)、多烯紫杉醇(docetaxel)、多柔比星、盐酸多柔比星、丙酸屈他雄酮(dromostanolone propionate)、表柔比星(epirubicin)、依伯汀α(epoetin alfa)、埃罗替尼(erlotinib)、雌莫司汀(estramustine)、磷酸依托泊苷、依托泊苷、依西美坦(exemestane)、非格司亭(filgrastim)、氟尿苷(floxuridine)、氟达拉宾(fludarabine)、氟维司群(fulvestrant)、吉非替尼、吉西他滨(gemcitabine)、吉妥珠单抗(gemtuzumab)、乙酸戈舍瑞林(goserelin acetate)、乙酸组氨瑞林(histrelin acetate)、羟基脲(hydroxyurea)、替伊莫单抗(ibritumomab)、艾达霉素(idarubicin)、异环磷酰胺(ifosfamide)、甲磺酸伊马替尼(imatinib mesylate)、干扰素α-2a、干扰素α-2b、伊立替康(irinotecan)、来那度胺(lenalidomide)、来曲唑(letrozole)、甲酰四氢叶酸(leucovorin)、乙酸亮丙立德(leuprolide acetate)、左旋咪唑(levamisole)、洛莫司汀(lomustine)、乙酸甲地孕酮(megestrol acetate)、美法仑(melphalan)、巯嘌呤(mercaptopurine)、6-MP、巯乙磺酸钠(mesna)、甲氨蝶呤、补骨脂素(methoxsalen)、丝裂霉素C(mitomycin C)、米托坦(mitotane)、米托蒽醌(mitoxantrone)、诺龙(nandrolone)、奈拉滨(nelarabine)、诺非单抗(nofetumomab)、奥普瑞白介素(oprelvekin)、奥沙利铂(oxaliplatin)、太平洋紫杉醇、帕利夫明(palifermin)、帕米膦酸盐(pamidronate)、培加酶(pegademase)、培门冬酶(pegaspargase)、乙二醇化非格司亭 (pegfilgrastim)、培美曲塞二钠(pemetrexed disodium)、喷司他汀(pentostatin)、哌泊溴烷(pipobroman)、普卡霉素(plicamycin)、卟吩姆钠(porfimer sodium)、丙卡巴肼(procarbazine)、奎纳克林(quinacrine)、拉布立酶(rasburicase)、利妥昔单抗(rituximab)、沙格司亭(sargramostim)、索拉非尼(sorafenib)、链脲霉素(streptozocin)、顺丁烯二酸舒尼替尼(sunitinib maleate)、滑石、他莫西芬(tamoxifen)、替莫唑胺(temozolomide)、替尼泊苷、VM-26、睾内酯(testolactone)、硫鸟嘌呤(thioguanine)、6-TG、噻替派(thiotepa)、拓朴替康、托瑞米芬(toremifene)、托西莫单抗(tositumomab)、曲妥珠单抗(trastuzumab)、维甲酸(tretinoin)、ATRA、尿嘧啶氮芥(uracil mustard)、伐柔比星(valrubicin)、长春碱、长春新碱(vincristine)、长春瑞滨(vinorelbine)、唑来膦酸盐(zoledronate)或唑来膦酸(zoledronic acid)。
本发明化合物还可以组合的治疗剂的其它实例包括但不限于:用于阿尔茨海默氏病(Alzheimer′s Disease)的治疗剂,如盐酸多奈哌齐(donepezil hydrochloride)和雷斯替明(rivastigmine);用于帕金森氏病(Parkinson′s Disease)的治疗剂,如L-DOPA/卡比多巴(carbidopa)、恩他卡朋(entacapone)、罗匹尼洛(ropinrole)、普拉克索(pramipexole)、溴麦角环肽(bromocriptine)、培高利特(pergolide)、三己芬迪(trihexephendyl)以及三环癸胺(amantadine);用于多发性硬化症(Multiple Sclerosis;MS)的治疗剂,如β干扰素、乙酸格拉默(glatiramer acetate)和米托蒽醌;用于哮喘的治疗剂,如舒喘宁(albuterol)和孟鲁司特;用于精神***症的治疗剂,如再普乐(zyprexa)、理斯必妥(risperdal)、思瑞康(seroquel)以及氟哌啶醇(haloperidol);消炎剂,如皮质类固醇、TNF阻断剂、IL-1RA、硫唑嘌呤(azathioprine)、环磷酰胺以及柳氮磺胺吡啶(sulfasalazine);免疫调节剂和免疫抑制剂,如环孢素(51yclosporine)、他克莫司(tacrolimus)、雷帕霉素、霉酚酸吗啉乙酯(mycophenolate mofetil)、干扰素、皮质类固醇、环磷酰胺、硫唑嘌呤以及柳氮磺胺吡啶;神经营养因子,如乙酰胆碱酯酶抑制剂、MAO抑制剂、干扰素、抗惊厥剂、离子通道阻断剂、利鲁唑(riluzole)以及抗帕金森氏病剂;用于心血管疾病的治疗剂,如β阻断剂、ACE抑制剂、利尿剂、硝酸盐、钙离子通道阻断剂以及他汀类(statin);用于肝病的治疗剂,如皮质类固醇、消胆胺(cholestyramine)、干扰素以及抗病毒剂;用于血液病症的治疗剂,如皮质类固醇、抗白血病剂以及生长因子;以及用于免疫缺陷病症的治疗剂,如γ球蛋白。
那些其它活性剂可以与含有本发明化合物的组合物分开给药,作为多次给药方案的一部分。或者,那些活性剂可以是单一剂型的一部分,与本发明化合物一起混合在单一组合物中。如果作为多次给药方案的一部分给药,那么两种活性剂可以同时、依次或彼此间隔一段时间(通常彼此间隔在5小时以内)提供。
治疗
本发明提供了一种抑制蛋白酪氨酸激酶(如EGFR激酶)的方法或治疗疾病(如癌症、细胞增殖性疾病、炎症、感染、免疫性疾病、器官移植、病毒性疾病、心血管疾病或代谢性疾病)的方法,它包括步骤:给需要治疗的受试者给药本发明化合物,或其药学上可接受的盐、立体异构体、溶剂合物、水合物、晶型、前药或同位素衍生物,或给药本发明所述的药物组合物。
本发明化合物可用于治疗EGFR导致的癌症。尤其是,所述化合物可用于治疗表达EGFR突变体的EGFR导致的癌症和用于治疗对RTKI疗法(例如,厄洛替尼或吉非替尼)难治的EGFR导致的癌症。
本发明化合物是EGFR的至少一种突变体的抑制剂并且因此适用于治疗与一种或一种以上EGFR突变体(例如缺失突变、活化突变、抗性突变或其组合,具体实例包括T790M突变、L858R突变和L858R/T790M双突变)的活性相关的一种或一种以上病症。因此,在具体实施方案中,本发明提供一种治疗突变EGFR介导的病症的方法,其包含向有需要的患者给药本发明化合物,或其药学上可接受的盐、立体异构体、溶剂合物、水合物、晶型、前药或同位素衍生物,或给药本发明所述的药物组合物的步骤。
本发明化合物可治疗的癌症包括但不限于:非小细胞肺癌(NSCLS)、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、乳腺癌、***癌、肝癌、皮肤癌、上皮细胞癌、胃肠间质瘤、白血病、组织细胞性淋巴癌、鼻咽癌等过度增殖性疾病。此外,本发明化合物也可用于在需要此类治疗的患者中起到预防癌症复发的维持作用。
本发明化合物的有效量通常在平均日剂量为0.01mg至50mg化合物/千克患者体重,优选0.1mg至25mg化合物/千克患者体重,以单次或多次给药。通常,本发明化合物可向该有此治疗需要的患者以每位患者约1mg至约3500mg的日剂量范围给药,优选10mg至1000mg。例如,每位患者的日剂量可为10、20、30、40、50、60、70、80、90、100、150、200、250、300、350、400、500、600、700、800、900或1000mg。可每天、每周(或间隔数天)或以间歇时间表,给药一次或多次。例如,可在每周的基础上(例如每周一),每天给予所述化合物一次或多次,不定地或持续几周,例如4-10周。或者,可每天给药持续几天(例如2-10天),然后几天(例如1-30天)不给药所述化合物,不定地重复该循环或重复给定的次数,例如4-10个循环。例如,本发明化合物可每天给药持续5天,然后间断9天,然后再每天给药持续5天,然后间断9天,以此类推,不定地重复该循环或共重复4-10次。
当EGFR-TKI(例如,厄洛替尼或吉非替尼)与本发明化合物组合使用时,该组合疗 法的各个成分可以以它们单一疗法的剂量水平和方案给药。例如,厄洛替尼,对于治疗非小细胞肺癌,已经以每天150mg口服给药,对于胰腺癌,已经以每天100mg口服给药。在另一实例中,吉非替尼对于治疗非小细胞肺癌已经以每天250mg口服给药。
优选地,当EGFR-TKI(例如,厄洛替尼或吉非替尼)与本发明化合物组合使用,其一种或两种成分的剂量水平相比于单独使用时降低。
实施例
提供以下实施例以便为本领域技术人员提供如何进行、制备和评估本文请求保护的方法和化合物的完整公开和说明,旨在仅仅示例本发明而非限制本发明的范围。
合成方法
本发明化合物可按照本领域常规方法,并使用合适的试剂、原料和本领域技术人员已知的纯化方法制备。
下面更具体地描述本发明式I结构化合物的制备方法,但这些具体方法不对本发明构成任何限制。本发明化合物还可以任选将在本说明书中描述的或本领域已知的各种合成方法组合起来而方便地制得,这样的组合可由本发明所属领域的技术人员容易地进行。
通常,在制备中,各反应通常在惰性溶剂中,在室温至回流温度(如0℃~100℃,优选0℃~80℃)进行。反应时间通常为0.1小时-60小时,优选0.5-24小时。
中间体化合物的合成
中间体化合物4-氟-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑的合成。
Figure PCTCN2017094102-appb-000079
步骤1化合物N-异丙基乙酰胺的合成。
冰浴下,将乙酸酐(16.15mL)缓慢滴加到三乙胺(TEA,23.58mL)和异丙胺(10g)的二氯甲烷(100mL)溶液中,滴加完毕过,室温下搅拌反应过夜。减压除去二氯甲烷。加入***,再加入大量的碳酸钾,搅拌过夜。固体过滤,滤液减压浓缩得到16.0g无色透明的油状物,收率:100%。LC-MS(APCI):m/z=102.1(M+1);1H NMR(500MHz,CDCl3)(δ/ppm)4.14–3.99(m,1H),1.94(s,3H),1.13(d,J=6.6Hz,6H)。
步骤2化合物(E)-N’-(4-溴-2,6-二氟苯基)-N-异丙基乙脒的合成。
室温下三氯氧磷(6.5mL)加入到4-溴-2,6-二氟苯胺(9.00g,43.27mmol)和N-异丙基乙酰胺(8.74g,86.54mmol),三乙胺(10mL)的甲苯溶液(130mL)中,反应液回流搅拌反应3小时。冷却至室温,减压除去反应液。粗品溶入150mL二氯甲烷中,用饱和的碳酸氢钠溶液洗涤3-4次,无水硫酸钠干燥,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=5:1),得到12.0g淡黄色固体。收率:95.6%。LC-MS(APCI):m/z=291.1(M+1)。
步骤3化合物6-溴-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑的合成。
氮气保护下,将叔丁醇钾(3.65g,32.6mmol)加入到(E)-N’-(4-溴-2,6-二氟苯基)-N-异丙基乙脒(4.50g,15.5mmol)的无水四氢呋喃中,反应在70℃反应2小时。冷却至室温,硅藻土过滤,二氯甲烷洗涤滤饼,滤液用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=5:1),得到4.0g淡黄色固体。收率:95.6%。LC-MS(APCI):m/z=271.1(M+1)。
步骤4化合物4-氟-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑的合成。
氮气保护下将醋酸钯(28mg)和三环己基磷(54.3mg)加入到6-溴-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑(300mg,1.10mmol),联硼酸频那醇酯(422mg,1.70mmol)和醋酸钾(326mg,3.3mmol)的无水二甲亚砜(DMSO,5mL)溶液中,反应在氮气保护下100℃反应2小时。冷却至室温,硅藻土过滤,乙酸乙酯洗涤滤饼,滤液用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1),得到260mg淡黄色固体。收率:74.3%。LC-MS(APCI):m/z=319.3(M+1)。
中间体化合物N-(5-氨基-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
Figure PCTCN2017094102-appb-000080
步骤1化合物(4-氟-2-甲氧基-5-硝基苯基)氨基甲酸叔丁酯的合成。
在冰浴下,将Boc酸酐(7.05g,32.20mmol)缓慢滴加到4-氟-2-甲氧基-5-硝基苯胺(4.0g,21.50mmol)和三乙胺(TEA,3.60g,35.64mmol)的二氯甲烷(65mL)混合液中,滴加完毕后,反应液在40℃反应过夜。冷却至室温,加水(100mL)稀释,用二氯甲烷(100mLx3)萃取,合并有机相,饱和食盐水(100mL)洗涤,无水硫酸钠干燥,除去溶剂,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=2:1),得到黄固体3.6g,收率:58.8%。LC-MS(APCI):m/z=287.2(M+1)。
步骤2化合物(4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基-5-硝基苯基)氨基甲酸叔丁酯的合成。
将N1,N1,N2-三甲基乙烷-1,2-二胺(2.14g,20.40mmol)加入到(4-氟-2-甲氧基-5-硝基苯基)氨基甲酸叔丁酯(4.0g,21.50mmol)和N,N-二异丙基乙胺(DIPEA,4.92mL,27.20mmol)的N,N-二甲基甲酰胺(28mL)混合液中,反应液在90℃反应5小时。冷却至室温,加水(100mL)稀释,用乙酸乙酯(100mLx3)萃取,合并有机相,饱和食盐水(100mLx3)洗涤,无水硫酸钠干燥,除去溶剂得到5.0g棕色油状物,不经纯化直接用于下一步反应。LC-MS(APCI):m/z=369.3(M+1)。
步骤3化合物(5-氨基-4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基苯基)氨基甲酸叔丁酯的合成。
将Pd/C(500mg)加入到(4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基-5-硝基苯基)氨基甲酸叔丁酯(5.00g,13.6mmol)的甲醇(65mL)溶液中,氢气球下搅拌反应过夜。硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=20:1),得到棕黄色油状物4.0g,两步收率:86.8%。LC-MS(APCI):m/z=339.3(M+1)。
步骤4化合物(5-丙烯酰胺基-4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基苯基) 氨基甲酸叔丁酯的合成。
室温下将3-氯丙酰氯(1.80g,14.20mmol)加入到(5-氨基-4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基苯基)氨基甲酸叔丁酯(4.00g,11.80mmol)的四氢呋喃(95mL)和水(9.5mL)的混合溶液中,室温下反应1小时。将氢氧化钠(2.40g)加入到反应液中,反应在65℃反应过夜。冷却至室温,减压浓缩反应液。残渣加水(100mL)稀释,用二氯甲烷(100mLx3)萃取,合并有机相,饱和食盐水(100mL)洗涤,无水硫酸钠干燥,除去溶剂,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=20:1),得到棕黄色固体3.20g,收率:69.2%。LC-MS(APCI):m/z=393.3(M+1)。
步骤5化合物N-(5-氨基-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
室温下将氯化氢的甲醇溶液(4M,5mL)加入到((5-丙烯酰胺基-4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基苯基)氨基甲酸叔丁酯(392mg,1.00mmol)的甲醇(2mL)中,反应搅拌反应过夜,减压除去甲醇,加入饱和的碳酸氢钠(30mL),二氯甲烷(30mLx3)萃取,合并有机相,饱和食盐水(30mL)洗涤,无水硫酸减压浓缩得到黄固体292mg,收率:95%。LC-MS(APCI):m/z=293.3(M+1)。
中间体化合物2-甲氧基-4-(4-甲基哌嗪-1-基)苯胺的合成。
Figure PCTCN2017094102-appb-000081
将5-氟-2-硝基苯甲醚(1.5g,8.7mmol)、1-甲基哌嗪(1.0g,10.5mmol)和碳酸钾(2.4g,17.4mmol)的N,N-二甲基甲酰胺(DMF,5mL)反应液加热至80℃并反应18小时后,冷却至室温,倒入50mL水,过滤得沉淀直接用于下一步。在氢气氛围下,将沉淀溶于30mL甲醇中,加入兰尼镍(200mg),氢气氛围下室温反应5小时,过滤后收集滤液,通过柱色谱分离纯化得到产物2-甲氧基-4-(4-甲基哌嗪-1-基)苯胺1.6g,收率为85%。LC-MS(APCI):m/z=221.3(M+1)。
中间体化合物6-(2-氯-5-氟嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑的合成。
Figure PCTCN2017094102-appb-000082
氮气氛围下,将2,4-二氯-5-氟嘧啶(517mg,3.1mmol)、碳酸钠(583mg,5.5mmol)溶于1mL水和5mL乙二醇二甲醚的混合溶液中,加入PdCl2(PPh3)2(4.7mg,66μmol),加热至80℃后,加入4-氟-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基-1H-苯并[d]咪唑(700mg,2.2mmol)的乙二醇二甲醚溶液(40mL),在该温度下搅拌4小时后,加入30mL水,继续搅拌25分钟,过滤在80℃烘干滤饼后用异丙醇洗涤滤饼后,干燥得产物6-(2-氯-5-氟嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑425mg,收率为60%。LC-MS(APCI):m/z=322.7(M+1)。
中间体化合物4-氟-1-异丙基-2-甲氧基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑的合成。
Figure PCTCN2017094102-appb-000083
步骤1化合物5-溴-3-氟-N1-异丙基苯-1,2-二胺的合成。
化合物4-溴-2,6-二氟-2硝基苯胺(4.08g,17.1mmol)溶于30mL无水四氢呋喃,冰浴冷却,加入碳酸铯(5.59g,17.1mmol)搅拌均匀。取异丙基胺(1.01g,17.1mmol)缓慢滴加,滴加完毕自然升温反应过夜。抽滤去除不溶性固体,合并滤液蒸干得白色固体产物直接用于下一步。向上一步产物中加乙醇-水(4/1,v/v,30mL)溶解,加入还原铁粉(5.74g)和氯化铵(1.80g),90摄氏度加热回流反应2.5小时。冷却至室温,抽滤去除不溶性固体,减压蒸去大部分乙醇,加入饱和碳酸氢钠水溶液20mL,EA 20mL萃取分液。有机相无水硫酸钠干燥减压蒸干得到粗品共3.93g。
步骤2化合物6-溴-4-氟-1-异丙基-1,3-二氢-2H-苯并[d]咪唑-2-酮的合成。
取化合物5-溴-3-氟-N1-异丙基苯-1,2-二胺的粗品1.0g,室温下加入4mL无水DMF搅拌溶解,之后加入CDI(N,N’-羰基二咪唑,0.66g,4.05mmol)室温反应过夜。倒入20mL水中,EA(5mL*3)萃取,合并有机相经无水硫酸钠干燥蒸干得到粗品,无需纯化,直接用于下一步。
步骤3化合物6-溴-2-氯-4-氟-1-异丙基-1H-苯并[d]咪唑的合成。
向化合物6-溴-4-氟-1-异丙基-1,3-二氢-2H-苯并[d]咪唑-2-酮中加入三氯氧磷5mL, 甲苯5mL,氮气保护下90摄氏度加热搅拌反应72小时,冷却反应液,之后倒入冰浴冷却的饱和碳酸氢钠-EA(20mL-10mL)搅拌10分钟,分液。有机相蒸干柱层析(PE/EA,v/v,20/1)得到产物0.57g。
步骤4中间体化合物4-氟-1-异丙基-2-甲氧基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑的合成。
将化合物6-溴-2-氯-4-氟-1-异丙基-1H-苯并[d]咪唑(0.57g,1.96mmol)溶于5mL无水DMF,一次性加入甲醇钠(0.65g,11.75mmol)室温搅拌反应过夜,LC-MS监控显示反应完毕,滴加饱和氯化铵水溶液0.5mL淬灭反应,加水50mL,EA(10mL*2)萃取,合并有机相无水硫酸钠干燥蒸干得到白色固体产物,无需纯化直接用于下一步。氮气保护下,将上述白色固体,连硼酸频哪醇酯(597mg,2.35mmol),三环己基膦(56mg,0.2mmol),醋酸钾(577mg,5.88mmol)加入到无水DMSO(5mL)中,搅拌均匀氮气置换5次,之后加入醋酸钯(22mg,0.1mmol)氮气置换3次,于90摄氏度油浴中加热搅拌反应1.5小时,冷却反应液至室温,加水50mL,PE-EA(v/v,1/1,10mL*2)萃取,合并有机相蒸干柱层析(PE/EA,v/v,4/1)得到化合物51,共0.49g。
中间体化合物4-氟-1-环丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑和中间体化合物6-(2-氯嘧啶-4-基)-1-环丙基-4-氟-2-甲基-1H-苯并[d]咪唑的合成。
Figure PCTCN2017094102-appb-000084
步骤1化合物N-环丙基乙酰胺的合成。
冰浴下,将乙酸酐(16mL)缓慢滴加到三乙胺(23mL)和异丙胺(9g)的二氯甲烷(100mL)溶液中,滴加完毕过,室温下搅拌反应过夜。减压除去二氯甲烷。加入***,再加入大量的碳酸钾,搅拌过夜。固体过滤,滤液减压浓缩得到17g无色透明的油状物,收率:100%。LC-MS(APCI):m/z=100.1(M+1)。
步骤2化合物(E)-N’-(4-溴-2,6-二氟苯基)-N-环丙基乙脒的合成。
室温下三氯氧磷(6.70mL)加入到4-溴-2,6-二氟苯胺(10.0g,48.00mmol)和N-异环丙基乙酰胺(9.50g,96.00mmol),三乙胺(10mL)的甲苯溶液(150mL)中,反应液回流搅拌反应3小时。冷却至室温,减压除去反应液。粗品溶入150mL二氯甲烷中,用饱和 的碳酸氢钠溶液洗涤3-4次,无水硫酸钠干燥,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=5:1),得到12g白色固体。收率:86.5%。LC-MS(APCI):m/z=289.1(M+1)。
步骤3化合物(6-溴-4-氟-1-环丙基-2-甲基)-1H苯并[d]咪唑的合成。
氮气保护下,将叔丁醇钾(7.00g,62.5mmol)加入到(E)-N’-(4-溴-2,6-二氟苯基)-N-环丙基乙脒(12g,41.7mmol)的无水四氢呋喃(100mL)中,反应在80℃反应2小时。冷却至室温,硅藻土过滤,二氯甲烷洗涤滤饼,滤液用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1),得到10.0g淡黄色固体。收率:81.1%。LC-MS(APCI):m/z=269.1(M+1)。
步骤4中间体化合物4-氟-1-环丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑的合成。
氮气保护下将醋酸钯(800mg)和三环己基磷(1.60g)加入到6-溴-4-氟-1-环丙基-2-甲基-1H苯并[d]咪唑(10g,37.17mmol),联硼酸频那醇酯(14g,55.76mmol)和醋酸钾(10.9g,111.51mmol)的无水二甲亚砜(DMSO,100mL)溶液中,反应在氮气保护下100℃反应2小时。冷却至室温,硅藻土过滤,乙酸乙酯洗涤滤饼,滤液用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1),得到8.60g白色固体。收率:73.2%。LC-MS(APCI):m/z=317.3(M+1)。
步骤5中间体化合物6-(2-氯嘧啶-4-基)-1-环丙基-4-氟-2-甲基-1H-苯并[d]咪唑的合成。
氮气保护下将乙腈(9mL)和水(3mL)加入到4-氟-1-环丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(1.00g,3.16mmol)和2,4-二氯嘧啶(566mg,3.82mmol)、碳酸钠(839mg,7.91mmol)、双三苯基磷二氯化钯(111mg,0.16mmol)的混合物中,反应在氮气保护下,80℃搅拌反应2小时。冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼,用饱和食盐水洗涤滤液,无水硫酸钠干燥,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1)得到900mg白色固体。收率为94.3%。LC-MS(APCI):m/z=303.1(M+1)。
中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑的合成。
Figure PCTCN2017094102-appb-000085
步骤1化合物5-溴-3-氟-N-异丙基-2-硝基苯胺的合成。
冰浴下,将碳酸铯(5.86g,18.00mmol)和异丙胺(1.06g,18.00mmol)依次加入到5-溴-1,3-二氟-2-硝基苯(4.28g,18.00mmol)的无水四氢呋喃(50mL)溶液中,反应液自然升温至室温,搅拌过夜,硅藻土过滤,滤液减压浓缩得到粗品,不纯化,直接用于下一步反应。收率为100%。
步骤2化合物5-溴-3-氟-N1-异丙胺苯-1,2-二胺的合成。
将上述的粗品溶入乙醇和水的混合溶剂中(30mL,v/v=3/1),加入还原铁粉(6.05g,108mmol)和氯化铵(2.91g,54mmol),反应液升温至回流2.5hrs,冷却至室温,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=50:1),得到4.296g棕色液体。收率为96.6%。LC-MS(APCI):m/z=247.1(M+1)。
步骤3化合物(6-溴-4-氟-1-异丙基-1H-苯并[d]咪唑-2-基)甲醇的合成。
将6N的稀盐酸(15mL)和水(23mL)加入到5-溴-3-氟-N1-异丙胺苯-1,2-二胺(4.00g,16.26mmol)和乙醇酸(6.18g,81.30mmol)混合物中,反应回流反应2hrs,冷却至室温,用浓氨水调pH至碱性,二氯甲烷(100mLx3)萃取,合并有机相,饱和食盐水(100mL)洗涤无水硫酸钠干燥,除去溶剂,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=1:1),得到淡黄固体3.7g,收率为79.8%。LC-MS(APCI):m/z=287.2(M+1);1H NMR(400MHz,CDCl3):δ7.45(d,J=1.5Hz,1H),7.07(dd,J=9.6,1.5Hz,1H),5.47(br,1H),4.97–4.85(m,3H),1.63(d,J=7.0Hz,6H).
步骤4化合物6-溴-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑的合成。
冰浴下,将DAST氟化试剂(0.64mL,4.80mmol)缓慢加入到(6-溴-4-氟-1-异丙基-1H-苯并[d]咪唑-2-基)甲醇(1.14g,4.00mmol)的无水二氯甲烷(30mL)溶液中,反应液在是冰浴下反应30min,用饱和碳酸氢钠溶液淬灭反应,二氯甲烷(30mLx3)萃取,合并有机相,饱和食盐水(50mL)洗涤无水硫酸钠干燥,除去溶剂,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1),得到淡黄固体860mg,收率为75.4%。LC-MS(APCI):m/z=289.2(M+1);1H NMR(400MHz,CDCl3):δ7.52(s,1H),7.15(dd,J=9.5,1.4Hz,1H),5.70(s,1H),5.58(s,1H),4.84(dt,J=13.6,6.8Hz,1H),1.67(d,J=6.9Hz,6H).
步骤5中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑的合成。
氮气保护下,将pd(dppf)Cl2.DCM(81mg,0.1mmol)加入到6-溴-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑(340mg,1.18mmol),联硼酸频那醇酯(360mg,1.42mmol)和乙酸钾(230mg,2.36mmol)的无水二氧六环(16mL)混合物中,氮气保护下,反应在100℃下反应过夜。
反应液冷却至室温,氮气保护下将2,4-二氯嘧啶(213mg,1.42mmol),碳酸钠(250mmol,2.36mmol),pd(dppf)Cl2.DCM(81mg,0.1mmol)和水(4mL)加入到反应液中,氮气保护下,反应升温至80℃反应2.5hrs。冷却至室温,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=1:1),得到290mg黄色固体。收率为76.1%。LC-MS(APCI):m/z=323.2(M+1)。
中间体化合物6-(2-氯嘧啶-4-基)-1-异丙基-2-甲基-1H-咪唑并[4,5-b]吡啶的合成。
Figure PCTCN2017094102-appb-000086
步骤1 5-溴-N3-异丙基吡啶-2,3-二胺的合成。
室温下,将冰醋酸(0.86mL,15.00mmol)加入到5-溴吡啶-2,3-二胺(2.82g,15.00mmol)和丙酮(870mg,15.00mmol)的无水二氯甲烷(30mL)溶液中,反应在室温下搅拌5hrs。冷却至冰浴,分批加入三乙酰氧基硼氢化钠(4.77g,22.50mmol),反应液自然升温,室温下反应过夜。用饱和碳酸氢钠溶液调pH至8,二氯甲烷(50mLx3)萃取,有机相分别用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=35:1),得到1.5固体。收率为47.8%。LC-MS(APCI):m/z=230(M+1)。1H NMR(400MHz,DMSO-d6):δ7.25(d,J=2.1Hz,1H),6.61(d,J=2.0Hz,1H),5.74(Br,2H),4.80(d,J=7.3Hz,1H),3.60–3.44(m,1H),1.14(d,J=6.3Hz,6H).
步骤2 6-溴-1-异丙基-2-甲基-1H-咪唑并[4,5-b]吡啶的合成。
将乙酸酐(2mL)加入到5-溴-N3-异丙基吡啶-2,3-二胺(1.50g,7.16mmol)的乙酸(15mL)溶液中,反应液在125℃下回流反应2hrs。冷却至室温。减压除去反应液,残渣用饱和碳酸氢钠溶液调pH至8,二氯甲烷(50mLx3)萃取,有机相分别用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=25:1),得到1.4固体。收率为47.8%。LC-MS(APCI):m/z=254.0(M+1)。1H NMR(400MHz,DMSO-d6):δ8.39(dd,J=5.7,2.1Hz,2H),4.76(hept,J=6.9Hz,1H),2.62(s,3H),1.54(d,J=6.9Hz,6H).
步骤3中间体化合物6-(2-氯嘧啶-4-基)-1-异丙基-2-甲基-1H-咪唑并[4,5-b]吡啶的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物6-(2-氯嘧啶-4-基)-1-异丙基-2-甲基-1H-咪唑并[4,5-b]吡啶(510mg白色固体)。收率为91.1%。LC-MS(APCI):m/z=287.8(M+1);1H NMR(400MHz,DMSO-d6):δ9.16(d,J=2.0Hz,1H),8.83(d,J=5.3Hz,1H),8.72(d,J=2.1Hz,1H),8.36(d,J=5.3Hz,1H),4.87(dt,J=13.8,6.9Hz,1H),2.68(s,3H),1.62(d,J=6.9Hz,6H).
中间体化合物6-(2-氯嘧啶-4-基)-1-异丙基-2-甲氧基-1H-咪唑并[4,5-b]吡啶的合成。
Figure PCTCN2017094102-appb-000087
步骤1 6-溴-1-异丙基-1,3-二氢-2H-咪唑并[4,5-b]吡啶-2-酮的合成。
室温下将CDI(4.5g,627.83mmol)加入到5-溴-N3-异丙基吡啶-2,3-二胺(1.6g,6.96mmol)的无水乙腈(50mL),反应在室温下反应过夜,加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=25:1),得到1.00g白色固体,收率为83.0%。LC-MS(APCI):m/z=256.0(M+1)。
步骤2 6-溴-2-氯-1-异丙基-1H-咪唑并[4,5-b]吡啶的合成。
室温下,将6-溴-1-异丙基-1,3-二氢-2H-咪唑并[4,5-b]吡啶-2-酮(1.00g,3.92mmol)溶入三氯氧磷(20mL)中,反应回流反应过夜,冷却至室温,减压除去反应液,残渣用饱和碳酸氢钠溶液调pH至中性,二氯甲烷(50mLx3)萃取,有机相分别用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=1:1),得到950mg白色固体。收率为88.5%。LC-MS(APCI):m/z=274.0(M+1)。
步骤3 6-溴-2-甲氧基-1-异丙基-1H-咪唑并[4,5-b]吡啶的合成。
冰浴下,将甲醇钠(245mg,4.50mmol)加入到6-溴-2-氯-1-异丙基-1H-咪唑并[4,5-b]吡啶(500mg,1.80mmol)的无水DMF(10mL),自然升温,反应搅拌过夜。加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减 压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=1:1),得到450mg白色固体,收率为93.0%。LC-MS(APCI):m/z=270.0(M+1)。
步骤4中间体化合物6-(2-氯嘧啶-4-基)-1-异丙基-2-甲氧基-1H-咪唑并[4,5-b]吡啶的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到185mg白色固体。收率为75.9%。LC-MS(APCI):m/z=304.1(M+1)。
中间体化合物1-异丙基-2-甲氧基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑和6-(2-氯嘧啶-4-基)-1-异丙基-2-甲氧基-1H-苯并[d]咪唑的合成。
Figure PCTCN2017094102-appb-000088
步骤1 6-溴-1-异丙基-1,3-二氢-2H-苯并[d]咪唑-2-酮的合成。
室温下将CDI(650mg,4.00mmol)加入到5-溴-N1-异丙基苯-1,2-二胺(912mg,4.00mmol)的无水DMF(10mL),反应在室温下反应过夜,加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=25:1),得到850mg棕黄色固体,收率为83.0%。LC-MS(APCI):m/z=256.4(M+1)。
步骤2 6-溴-2-氯-1-异丙基-1H-苯并[d]咪唑的合成。
室温下,将6-溴-1-异丙基-1,3-二氢-2H-苯并[d]咪唑-2-酮(850mg,3.34mmol)溶入三氯氧磷(10mL)中,反应回流反应3天,冷却至室温,减压除去反应液,残渣用饱和碳酸氢钠溶液调pH至中性,二氯甲烷(50mLx3)萃取,有机相分别用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=5:1),得到850mg固体。收率为90.4%。LC-MS(APCI):m/z=272.9(M+1)。
步骤3 6-溴-2-甲氧基-1-异丙基-1H-苯并[d]咪唑的合成。
冰浴下,将甲醇钠(420mg,7.80mmol)加入到6-溴-2-氯-1-异丙基-1H-苯并[d]咪唑(850mg,3.12mmol)的无水DMF(10mL),自然升温,反应搅拌过夜。加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=5:1),得到570mg棕黄色固体,收率为71.0%。LC-MS(APCI):m/z=269.0(M+1)。
步骤4中间体化合物1-异丙基-2-甲氧基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑的合成
氮气保护下,将TCHP(52mg,0.18mmol)和Pd(Oac)2(20mg,0.09mmol)加入到6-溴-2-甲氧基-1-异丙基-1H-苯并[d]咪唑(520mg,1.87mmol),联硼酸频那醇酯(570mg,2.24mmol)和乙酸钾(550mg,5.61mmol)的无水DMSO(13mL)混合物中,氮气保护下,反应在100℃下反应2hrs。冷却室温,加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=4:1),得到562mg棕黄色固体,收率为95.2%。LC-MS(APCI):m/z=317.1(M+1)。
步骤5中间体化合物6-(2-氯嘧啶-4-基)-1-异丙基-2-甲氧基-1H-苯并[d]咪唑的合成
氮气保护下,将Pd(PPh3)2Cl2(35mg,0.05mmol)加入到1-异丙基-2-甲氧基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(562mg,1.78mmol),2,4-二氯嘧啶(320mg,2.13mmol)和碳酸钠(480mg,4.45mmol)的乙腈(12mL)和水(4mL)的混合物中,氮气保护下反应升温至80℃反应2.5hrs。冷却至室温,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=1:1),得到582mg白色固体。收率为100%。LC-MS(APCI):m/z=303.0(M+1)。
中间体化合物5-(2-氯嘧啶-4-基)-3-甲基苯并[d]噁唑-2(3H)-酮的合成。
Figure PCTCN2017094102-appb-000089
步骤1 5-溴苯并[d]噁唑-2(3H)-酮的合成。
室温下将CDI(1.95g,12.00mmol)加入到2-氨基-4-溴苯酚(1.88g,10.00mmol)的无水THF(50mL),反应在85℃下回流反应2hrs,冷却至室温。用2N的稀盐酸淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用饱和碳酸氢钠溶液和食盐水洗涤,无水硫酸钠干燥,减压浓缩得到2.15g棕黄色固体,收率为100%。LC-MS(APCI):m/z=212(M-1)。
步骤2 5-溴-3-甲基苯并[d]噁唑-2(3H)-酮的合成。
室温下将碘甲烷(795mg,4.80mmol)滴加到5-溴苯并[d]噁唑-2(3H)-酮(1.00g,4.67mmol)和碳酸钾(1.29g,9.34mmol)的无水DMSO(20mL),反应在室温下反应过夜,加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用水和食盐水洗涤,无水硫酸钠 干燥,液减压浓缩,浓缩液进行柱分离得到910mg棕黄色固体,收率为85.5%。LC-MS(APCI):m/z=228.0(M-1)。
步骤3中间体化合物5-(2-氯嘧啶-4-基)-3-甲基苯并[d]噁唑-2(3H)-酮的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物5-(2-氯嘧啶-4-基)-3-甲基苯并[d]噁唑-2(3H)-酮(340mg,白色固体)。收率为75.9%。LC-MS(APCI):m/z=262.0(M+1);1H NMR(400MHz,DMSO-d6):δ8.84(d,J=5.3Hz,1H),8.20(d,J=5.4Hz,1H),8.08–8.02(m,2H),7.52(d,J=8.2Hz,1H),3.44(s,3H).
中间体化合物6-(2-氯嘧啶-4-基)-4-氟-1-异丙基-1H-吲唑的合成。
Figure PCTCN2017094102-appb-000090
步骤1 6-溴-4-氟-1H-吲唑的合成。
室温下,将水合肼(10mL)加入到4-溴-2,6-二氟苯甲醛(5.00g,22.60mmok)的1,4-二氧六环(10mL)溶液中,反应液在95℃下反应1.5hrs,冷却至室温,加入到冰水混合物中,用乙酸乙酯(50mLx3)萃取,合并有机层,无水硫酸钠干燥,加压浓缩得到3.5g黄色固体,收率为71.0%。LC-MS(APCI):m/z=215.1(M+1)。
步骤2 6-溴-4-氟-1-异丙基-1H-吲唑的合成。
室温下将2-碘丙烷(1.58g,9.30mmol)滴加到6-溴-4-氟-1H-吲唑(1.60g,7.44mmol)和碳酸钾(1.50g,11.16mmol)的无水DMSO(8mL),反应在室温下反应过夜,加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用水和食盐水洗涤,无水硫酸钠干燥,液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=9:1)得到960mg淡黄色固体,收率为50.2%。LC-MS(APCI):m/z=257.0(M-1)。
步骤3中间体化合物6-(2-氯嘧啶-4-基)-4-氟-1-异丙基-1H-吲唑的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物6-(2-氯嘧啶-4-基)-4-氟-1-异丙基-1H-吲唑(447mg,白色固体)。收率为76.8%。LC-MS(APCI):m/z=291.0(M+1);1H NMR(400MHz,CDCl3):δ8.69(d,J=5.2Hz,1H),8.13(s,2H),7.68(d,J=5.3Hz,1H),7.41(d,J=10.8Hz,1H),4.99(dt,J=13.2,6.6Hz,1H),1.64(d,J=6.7Hz,6H).
中间体化合物6-(2-氯嘧啶-4-基)-1-异丙基-1H-吲唑的合成。
Figure PCTCN2017094102-appb-000091
步骤1 6-溴-1-异丙基-1H-吲唑的合成
室温下将2-碘丙烷(2.00g,12.18mmol)滴加到6-溴-1H-吲唑(2.00g,10.15mmol)和碳酸钾(1.96g,14.21mmol)的无水DMSO(20mL),反应在室温下反应过夜,加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用水和食盐水洗涤,无水硫酸钠干燥,液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=5:1)得到1.17g淡黄油状物,收率为48.03%。LC-MS(APCI):m/z=240.9(M-1)。
步骤2中间体化合物6-(2-氯嘧啶-4-基)-1-异丙基-1H-吲唑的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物6-(2-氯嘧啶-4-基)-1-异丙基-1H-吲唑(290mg,白色固体)。收率为63.9%。LC-MS(APCI):m/z=273.0(M+1)。
中间体化合物6-(2-氯嘧啶-4-基)-8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶的合成。
Figure PCTCN2017094102-appb-000092
步骤1化合物5-溴-3-氟吡啶-2-胺的合成。
氮气保护下将N-溴代丁二酰亚胺(NBS,5.47g,30.57mmol)分批加入到3-氟吡啶-2-胺(3.36g,29.97mmol)的乙腈(150mL)中,反应在氮气保护下室温反应2小时。减压浓缩除去乙腈,加饱和硫代硫酸钠溶液淬灭,用乙酸乙酯萃取(100mLx3)合并有机相用饱和食盐水洗涤滤液,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=5:1),得到5.0g白色固体,收率为87.3%。LC-MS(APCI):m/z=191.1(M+1)。
步骤2化合物6-溴-8-氟-2-甲基咪唑并[1,2-a]吡啶的合成。
将1-溴丙-2-酮(10.00g,73.3mmol)加入到5-溴-3-氟吡啶-2-胺(7.00g,36.6mmol)的乙醇(70mL)中,反应在氮气保护下回流反应过夜。冷却至温室慢慢析出固体,过滤,用冷乙醇洗涤,真空干燥得到3.5g白色固体,收率:41.8%。LC-MS(APCI):m/z=229.1(M+1);1H NMR(400MHz,DMSO-d6):δ9.12(s,1H),8.16–8.09(m,2H),2.48(s,3H)。
步骤3化合物6-溴-8-氟-3-碘-2-甲基咪唑并[1,2-a]吡啶的合成。
将单质碘(4.60g,18.3mmol)加入到6-溴-8-氟-2-甲基咪唑并[1,2-a]吡啶(3.50g,15.30mmol)和乙酸钠(2.01g,24.5mmol)的甲醇(20mL)中,反应在氮气保护下室温反应3小时。减压浓缩除去甲醇,加饱和硫代硫酸钠溶液淬灭,用乙酸乙酯萃取(100mLx3)合并有机相用饱和食盐水洗涤滤液,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1),得到2.8g白色固体,收率为51.7%。LC-MS(APCI):m/z=355.1(M+1)。
步骤4化合物6-溴-8氟-2甲基-3-(丙-1-烯-2基)咪唑并[1,2-a]吡啶的合成。
氮气保护下将Pd(dppf)Cl2■CH2Cl2(250mg,0.31mmol)加入到6-溴-8-氟-3-碘-2-甲基咪唑并[1,2-a]吡啶(1.17g,3.32mmol)、异丙烯基硼酸频哪醇酯(557mg,3.31mmol)和碳酸铯(2.16g,6.64mmol)的1,4-二氧六环(20mL)和水(5mL)混合溶剂中,反应在氮气保护下80℃反应1小时。冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=10:1),得到600mg白色固体。收率为67.4.9%。LC-MS(APCI):m/z=269.1(M+1)。
步骤5中间体化合物6-(2-氯嘧啶-4-基)-8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物6-(2-氯嘧啶-4-基)-8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶(800mg,白色固体)。收率为100%。LC-MS(APCI):m/z=303.0(M+1)。
中间体化合物6-(2-氯嘧啶-4-基)-8-氟-2-甲基-咪唑并[1,2-a]吡啶的合成。
Figure PCTCN2017094102-appb-000093
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物6-(2-氯嘧啶-4-基)-8-氟-2-甲基-咪唑并[1,2-a]吡啶(630mg,白色固体)。收率为80.00%。LC-MS(APCI):m/z=263.0(M+1)。
中间体化合物4-叔丁氧基-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑和6-(2-氯嘧啶-4-基)-4-叔丁氧基-1-异丙基-2-甲基-1H-苯并[d]咪唑的合成。
Figure PCTCN2017094102-appb-000094
步骤1化合物6-溴-4-(叔丁氧基)-1-异丙基-2-甲基-1H-苯并[d]咪唑的合成
氮气保护下,将叔丁醇钾(3.65g,32.6mmol)加入到(E)-N’-(4-溴-2,6-二氟苯基)-N-异丙基乙脒(4.50g,15.5mmol)的无水四氢呋喃中,反应在70℃下反应2hrs。冷却至室温,硅藻土过滤,二氯甲烷洗涤滤饼,滤液用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩滤液,浓缩液进行柱分离(淋洗剂:石油醚烷/乙酸乙酯(v/v)=5:1),得到4.0g淡黄色固体混合物。收率:95.6%。LC-MS(APCI):m/z=271.1(M+1)&324.1(M+1)
步骤2中间体化合物4-叔丁氧基-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑的合成
氮气保护下将醋酸钯(466mg)和三环己基磷(900mg)加入到上述混合物中(5.00mg,联硼酸频那醇酯(7.00g)和醋酸钾(5.40g)的无水DMSO(50mL)溶剂中,反应在氮气保护下100℃反应2hrs。冷却至室温,硅藻土过滤,乙酸乙酯洗涤滤饼,滤液用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩滤液,浓缩液进行柱分离(淋洗剂:石油醚烷/乙酸乙酯(v/v)=3:1),得到产物4-叔丁氧基-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基-1H-苯并[d]咪唑(1.3g)。LC-MS(APCI):m/z=373.3(M+1)。
步骤3中间体化合物6-(2-氯嘧啶-4-基)-4-叔丁氧基-1-异丙基-2-甲基-1H-苯并[d]咪唑的合成
氮气保护下将乙腈(9mL)和水(3mL)加入到4-叔丁基-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(600mg,1.60mmol)和2,4-二氯嘧啶(290mg,1.90mmol)、碳酸钠(430mg,4.00mmol)、双三苯基磷二氯化钯(56mg,0.08mmol)的混合物中,反应在氮气保护下,80℃搅拌反应2hrs。冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼,用饱和食盐水洗涤滤液,无水硫酸钠干燥,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=2:1),得到产物6-(2-氯嘧啶-4-基)-4-叔丁氧基-1-异丙基-2-甲基-1H-苯并[d]咪唑(700mg,黄色固体)。收率:100%。LC-MS(APCI):m/z:359.1(M+1)。
中间体化合物5-(2-氯嘧啶-4-基)-7-氟-2-甲基苯并[d]噁唑的合成。
Figure PCTCN2017094102-appb-000095
步骤1:化合物2-氨基-4-溴-6-氟苯酚的合成
室温下,将SnCl2.2HO(14.3g,63.50mmol)加入到4-溴-2-氟-6-硝基苯酚(3.00g,12.7mmol)的乙醇(100mL)中,反应液回流反应2hrs。冷却至室温,加入到冰水中,继续搅拌0.5hrs。减压除去乙醇,用乙酸乙酯(100mLx3)萃取,合并有机层用水和食盐水洗涤,无水硫酸钠干燥,液减压浓缩得到淡黄色固体2.0g,收率为76.4%,LC-MS(APCI):m/z=206.1(M+1)。
步骤2:化合物5-溴-7-氟-2-甲基苯并[d]噁唑的合成
2-氨基-4-溴-6-氟苯酚(700mg,3.41mmol)的原乙酸三乙酯(20mL)中,反应液150℃下回流反应2hrs。冷却至室温,加入饱和碳酸氢钠溶液淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用水和食盐水洗涤,无水硫酸钠干燥,液减压浓缩,缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=1:1),得到淡黄色固体600mg,收率为76.5%,LC-MS(APCI):m/z=229.9(M+1)。
步骤3中间体化合物5-(2-氯嘧啶-4-基)-7-氟-2-甲基苯并[d]噁唑的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物5-(2-氯嘧啶-4-基)-7-氟-2-甲基苯并[d]噁唑(700mg,白色固体)。收率为98.20%。LC-MS(APCI):m/z=264.1(M+1)。
中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-甲基苯并[d]噁唑的合成。
Figure PCTCN2017094102-appb-000096
步骤1:化合物N-(4-溴-2,6-二氟苯基)乙酰胺的合成
室温下,将醋酸酐(9mL)滴加到4-溴-2,6-二氟苯胺(3.00g,14.42mmol)的冰醋酸(30mL)中,反应液在室温下反应2hrs。加入到冰水中,固体过滤,滤饼用二氯甲烷溶解,用饱和碳酸氢钠溶液和饱和食盐水洗涤,无水硫酸钠干燥,液减压浓缩得到淡黄色固体3.058g,收率为85.28%,LC-MS(APCI):m/z=249.1(M+1)。1H NMR(400MHz,DMSO-d6):δ9.74(s,1H),7.54(d,J=7.2Hz,2H),2.06(s,3H).
步骤2:化合物6-溴-4-氟-2-甲基苯并[d]噁唑的合成
将碳酸铯(8.00g)加入到N-(4-溴-2,6-二氟苯基)乙酰胺(3.00g)的无水NMP(19mL)中,反应液150℃下回流反应2hrs。冷却至室温,加入水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用水和食盐水洗涤,无水硫酸钠干燥,液减压浓缩,缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=9:1),得到淡黄色固体760mg,收率为27.6%, LC-MS(APCI):m/z=230.0(M+1)。
步骤3中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-甲基苯并[d]噁唑的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到6-(2-氯嘧啶-4-基)-4-氟-2-甲基苯并[d]噁唑(700mg白色固体)。收率为98.20%。LC-MS(APCI):m/z=264.0(M+1)。
中间体化合物5-(2-氯嘧啶-4-基)-7-氟-苯并[d]噁唑的合成。
Figure PCTCN2017094102-appb-000097
步骤1化合物5-溴-7-氟苯并[d]噁唑的合成
2-氨基-4-溴-6-氟苯酚(1.20g,5.80mmol)的原乙酸三乙酯(20mL)中,反应液150℃下回流反应2hrs。冷却至室温,加入饱和碳酸氢钠溶液淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用水和食盐水洗涤,无水硫酸钠干燥,液减压浓缩,缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1),得到淡黄色固体800mg,收率为64.2%,LC-MS(APCI):m/z=215..9(M+1)。
步骤2 5-(2-氯嘧啶-4-基)-7-氟-苯并[d]噁唑的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物5-(2-氯嘧啶-4-基)-7-氟-苯并[d]噁唑(500mg黄白色固体)。收率为56.2%。LC-MS(APCI):m/z=250.1(M+1)。
中间体化合物1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑和6-(2-氯嘧啶-4-基)-1-异丙基-2-甲基-1H-苯并[d]咪唑的合成。
Figure PCTCN2017094102-appb-000098
步骤1 5-溴-N-异丙基-2-硝基苯胺的合成。
冰浴下,将碳酸钾(5.02g,36.36mmol)和异丙胺(1.55mL,18.18mmol)依次加入到 4-溴-2-氟-1-硝基苯(4.00g,18.18mmol)的无水DMF(40mL)溶液中,反应液自然升温至室温,搅拌过夜。加水淬灭反应,用乙酸乙酯(100mLx3)萃取,合并有机层用水和食盐水洗涤,无水硫酸钠干燥,液减压浓缩得到4.62g黄色固体,不纯化,直接用于下一步反应。
步骤2 5-溴-N1-异丙基苯-1,2-二胺的合成。
将上述的粗品溶入乙醇和水的混合溶剂中(v/v=200mL/80mL),加入保险粉(18.46g,89.6mmol),反应在室温下反应过夜,硅藻土过滤,乙酸乙酯洗涤滤饼,减压除去乙醇,用乙酸乙酯(100mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩得到4.1g棕色液体。LC-MS(APCI):m/z=229.1(M+1)。
步骤3 6-溴-1-异丙基-2-甲基-1H-苯并[d]咪唑的合成。
将5-溴-N1-异丙基苯-1,2-二胺(2.6g,11.40mmol)加入到冰醋酸(40mL),反应在回流反应2hrs。冷却至室温,减压除去醋酸,残渣用饱和碳酸氢钠溶液调pH大约7,二氯甲烷(50x3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=20:1)得到2.31g棕黄色固体,收率为:80.10%,LC-MS(APCI):m/z=253.1(M+1)。
步骤4中间体化合物1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑的合成
氮气保护下,将TCHP(460mg)和Pd(Oac)2(230mg)加入到6-溴-2-甲基-1-异丙基-1H-苯并[d]咪唑(2.30g,9.10mmol),联硼酸频那醇酯(3.50g,13.6mmol)和乙酸钾(2.70g,27.3mmol)的无水DMSO(25mL)混合物中,氮气保护下,反应在100℃下反应2hrs。冷却室温,加水淬灭反应,用乙酸乙酯(100mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=2:1),得到1.60g棕黄色固体,收率为58.6%。LC-MS(APCI):m/z=301.2(M+1)。
步骤5中间体化合物6-(2-氯嘧啶-4-基)-1-异丙基-2-甲基-1H-苯并[d]咪唑的合成
氮气保护下,将Pd(PPh3)2Cl2(58mg,0.08mmol)加入到1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(500mg,1.67mmol),2,4-二氯嘧啶(298mg,2.00mmol)和碳酸钠(442mg,4.17mmol)的乙腈(9mL)和水(3mL)的混合物中,氮气保护下反应升温至80℃反应2.5hrs。冷却至室温,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=1:1),得到700mg白色固体。收率为100%。LC-MS(APCI):m/z=287.2(M+1)。
中间体化合物1-异丙基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并 [d]咪唑和6-(2-氯嘧啶-4-基)-1-异丙基-1H-苯并[d]咪唑的合成。
Figure PCTCN2017094102-appb-000099
步骤1 6-溴-1-异丙基-1H-苯并[d]咪唑的合成。
将5-溴-N1-异丙基苯-1,2-二胺(1.50g,6.57mmol)加入到甲酸(50mL),反应在回流反应2hrs。冷却至室温,减压除去甲酸,残渣用饱和碳酸氢钠溶液调pH大约7,二氯甲烷(50x3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=50:1)得到1.40g棕黄色固体,收率为:89.5%,LC-MS(APCI):m/z=238.2(M+1)。
步骤2 1-异丙基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑的合成
氮气保护下,将TCHP(280mg)和Pd(Oac)2(140mg)加入到6-溴-1-异丙基-1H-苯并[d]咪唑(1.40g,5.91mmol),联硼酸频那醇酯(2.20g,8.86mmol)和乙酸钾(1.70g,17.72mmol)的无水DMSO(20mL)混合物中,氮气保护下,反应在100℃下反应2hrs。冷却室温,加水淬灭反应,用乙酸乙酯(100mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=2:1),得到1.60g黄色油状物,收率为94.3%。LC-MS(APCI):m/z=287.2(M+1)。
步骤3 6-(2-氯嘧啶-4-基)-1-异丙基-1H-苯并[d]咪唑的合成
氮气保护下,将Pd(PPh3)2Cl2(74mg,0.10mmol)加入到1-异丙基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(600mg,2.10mmol),2,4-二氯嘧啶(375mg,2.52mmol)和碳酸钠(556mg,5.24mmol)的乙腈(9mL)和水(3mL)的混合物中,氮气保护下反应升温至80℃反应2.5hrs。冷却至室温,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=1:1),得到300mg黄色固体。收率为52.52%。LC-MS(APCI):m/z=273.1(M+1)。
中间体化合物6-(2-氯嘧啶-4-基)-1-异丙基-2-(三氟甲基)-1H-苯并[d]咪唑的合成。
Figure PCTCN2017094102-appb-000100
步骤1 6-溴-1-异丙基-2-(三氟甲基)-1H-苯并[d]咪唑的合成。
将5-溴-N1-异丙基苯-1,2-二胺(920mg,4.01mmol)加入到三氟乙酸(20mL),反应在 80℃反应过夜。冷却至室温,减压除去三氟乙酸,残渣用饱和碳酸氢钠溶液调pH大约7,二氯甲烷(50x3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=20:1)得到900mg淡黄色固体,收率为:73.3%,LC-MS(APCI):m/z=306.9(M+1)。
步骤2 6-(2-氯嘧啶-4-基)-1-异丙基-2-(三氟甲基)-1H-苯并[d]咪唑的合成
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物6-(2-氯嘧啶-4-基)-1-异丙基-2-(三氟甲基)-1H-苯并[d]咪唑(576mg白色固体)。收率为100%。LC-MS(APCI):m/z=341.0(M+1)。
中间体化合物5-(2-氯嘧啶-4-基)-7-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑的合成。
Figure PCTCN2017094102-appb-000101
步骤1 5-溴-2,3-二氟苯甲醛的合成。
将NBS(7.50g,42.00mmol)加入到2,3-二氟苯甲醛(5.00g,35.20mmol)的浓硫酸(37.7mL)中,反应液在60℃下搅拌过夜,冷却至室温,加入冰水中,搅拌10min,用石油醚萃取,饱和的食盐水洗涤,无水硫酸钠干燥。减压浓缩,浓缩液进行柱分离(100%石油醚)得到2.3g油状物,收率为25.0%。LC-MS(APCI):m/z=221.1(M+1)。
步骤2(E)-5-溴-2,3-二氟苯甲醛-O-甲基肟的合成。
将碳酸钾(1.73g)加入到5-溴-2,3-二氟苯甲醛(2.3g)和甲氧基胺盐酸盐(0.96g)的DME(26mL)溶液中,反应在45℃下反应过夜,冷去至室温,过滤,用乙酸乙酯洗涤,浓缩液进行柱分离(100%石油醚)得到2.342g油状物,收率为89.1%。LC-MS(APCI):m/z=250.2(M+1)。
步骤3 5-溴-7-氟-1H-吲唑的合成
将水合肼(9.5mL)加入到(E)-5-溴-2,3-二氟苯甲醛-O-甲基肟(2.34g)的无水四氢呋喃溶液中,反应在90℃下反应4天。减压除去四氢呋喃,残渣用乙酸乙酯溶解,水洗涤,无水硫酸钠干燥。减压浓缩,浓缩液进行柱分离(石油醚/乙酸乙酯(v/v)=4:1)得到1.00白色固体,收率为48.9%。LC-MS(APCI):m/z=215.0(M+1)。1H NMR(500MHz,DMSO-d6):δ13.84(s,1H),8.15(s,1H),7.86(d,J=1.3Hz,1H),7.44(d,J=10.0Hz,1H).
步骤4 5-溴-7-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑的合成
室温下,对甲基苯磺酸(40mg)加入到5-溴-7-氟-1H-吲唑(1.0g,4.64mmol)和3,4-二氢-2H-吡喃(0.78g,9.28mmol)的无水二氯甲烷(25mL),反应液在室温下反应过夜。用饱和的碳酸氢钠溶液淬灭反应,二氯甲烷萃取(30mLx3),合并有机层用饱和食盐水洗涤,无水硫酸钠干燥。减压浓缩,浓缩液进行柱分离(石油醚/乙酸乙酯(v/v)=10:1)得到1.20g白色固体,收率为86.8%。LC-MS(APCI):m/z=299.0(M+1)。
步骤5中间体化合物5-(2-氯嘧啶-4-基)-7-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到600mg黄白色固体。收率为90.36%。LC-MS(APCI):m/z=333.0(M+1)。
中间体化合物5-(2-氯嘧啶-4-基)-7-氟-1-甲基-1H-吲唑的合成。
Figure PCTCN2017094102-appb-000102
步骤1 5-溴-7-氟-2-甲基-2H-吲唑和5-溴-7-氟-1-甲基-1H-吲唑的合成。
冰浴下将氢化钠(80mg,3.35mmol)加入到5-溴-7-氟-1H-吲唑(600mg,2.79mmol)的无水四氢呋喃(10mL)溶液中,升温至室温反应1hr。再在冰浴下降温,将碘甲烷(0.26mL,4.18mmol)加入到反应液中,升温至室温下反应2hrs。加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥。浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=4:1-2:1)得到190mg白色固体B(5-溴-7-氟-2-甲基-2H-吲唑),LC-MS(APCI):m/z=229.0(M+1),1H NMR(500MHz,CDCl3):δ7.90(d,J=2.4Hz,1H),7.63(d,J=1.5Hz,1H),7.17(dd,J=11.0,1.5Hz,1H),4.23(d,J=1.0Hz,3H).
步骤2中间体化合物5-(2-氯嘧啶-4-基)-7-氟-1-甲基-1H-吲唑的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物5-(2-氯嘧啶-4-基)-7-氟-1-甲基-1H-吲唑(295mg黄白色固体)。收率为75.6%。LC-MS(APCI):m/z=263.0(M+1)。
中间体化合物5-(2-氯嘧啶-4-基)-7-氟-2-甲基-2H-吲唑的合成。
Figure PCTCN2017094102-appb-000103
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物5-(2-氯嘧啶-4-基)-7-氟-2-甲基-2H-吲唑(180mg黄白色固体)。收率为83.5%。LC-MS(APCI):m/z=263.1(M+1)。
中间体化合物5-(2-氯嘧啶-4-基)-7-氟-2-甲基-3-(丙-1-烯-2-基)-2H-吲唑的合成。
Figure PCTCN2017094102-appb-000104
步骤1 5-溴-7-氟-3-碘-1H-吲唑的合成。
室温下,将氢氧化钠(393mg,7.01mmol)分批加入到5-溴-7-氟-1H-吲唑(500mg,2.34mmol)和单质碘(1.19mg,4.68mmol)的无水DMF(10mL)溶液中,反应在室温下反应搅拌2hrs。用饱和的硫代硫酸钠溶液淬灭反应,乙酸乙酯萃取(30mLx3),合并有机层,用饱和的食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1),得到610mg黄色固体。收率为76.9%。LC-MS(APCI):m/z=340.9(M+1).
步骤2 5-溴-7-氟-3-碘-2-甲基-2H-吲唑的合成。
冰浴下将氢化钠(86mg,2.15mmol)加入到5-溴-7-氟-3-碘-1H-吲唑(610mg,1.79mmol)的无水四氢呋喃(10mL)溶液中,升温至室温反应1hr。再在冰浴下降温,将碘甲烷(0.17mL,2.19mmol)加入到反应液中,升温至室温下反应2hrs。加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥。浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=10:1)得到190mg白色固体,收率为29.9%,LC-MS(APCI):m/z=356.8(M+1).
步骤3 5-溴-7-氟-2-甲基-3-(丙-1-烯-2-基)-2H-吲唑的合成。
氮气保护下将Pd(dppf)Cl2.CH2Cl2(150mg)加入到5-溴-7-氟-3-碘-2-甲基-2H-吲唑(480mg,1.35mmol),异丙烯基硼酸频哪醇酯(215mg,1.28mmol)和碳酸铯(880mg, 2.70mmol)的二氧六环(16mL)和水(4mL)混合溶剂中,反应在氮气保护下80℃反应1hrs。冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼,减压浓缩滤液,浓缩液进行柱分离(淋洗剂:石油醚/乙酸乙酯(v/v)=10:1),得到185mg淡黄色油状物。收率:50.6%,LC-MS(APCI):m/z=271.0(M+1)。
步骤4 5-(2-氯嘧啶-4-基)-7-氟-2-甲基-3-(丙-1-烯-2-基)-2H-吲唑的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物5-(2-氯嘧啶-4-基)-7-氟-2-甲基-3-(丙-1-烯-2-基)-2H-吲唑(190mg黄白色固体)。收率为95.3%。LC-MS(APCI):m/z=302.9(M+1)。
中间体化合物5-(2-氯嘧啶-4-基)-2-甲基-3-(丙-1-烯-2-基)-2H-吲唑的合成。
Figure PCTCN2017094102-appb-000105
步骤1 5-溴-3-碘-1H-吲唑的合成。
室温下,将氢氧化钠(4.27g,76.13mmol)分批加入到5-溴-1H-吲唑(5.00g,25.38mmol)和单质碘(12.9g,50.75mmol)的无水DMF(10mL)溶液中,反应在室温下反应搅拌2hrs。用饱和的硫代硫酸钠溶液淬灭反应,乙酸乙酯萃取(50mLx3),合并有机层,用饱和的食盐水洗涤,无水硫酸钠干燥,减压浓缩得到8g黄色固体。收率为90.0%。LC-MS(APCI):m/z=322.2(M+1)。
步骤2 5-溴-3-碘-2-甲基-2H-吲唑的合成。
冰浴下将氢化钠(500mg,12.00mmol)加入到5-溴-3-碘-1H-吲唑(3.20g,10.00mmol)的无水四氢呋喃(30mL)溶液中,升温至室温反应1hr。再在冰浴下降温,将碘甲烷(1.25mL,20.00mmol)加入到反应液中,升温至室温下反应2hrs。加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥。浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=10:1)得到800mg白色固体,收率为23.9%,LC-MS(APCI):m/z=336.7(M+1)。1H NMR(500MHz,CDCl3):δ7.57(s,1H),7.54(d,J=9.1Hz,1H),7.36(d,J=8.1Hz,1H),4.24(s,3H).
步骤3 5-溴-2-甲基-3-(丙-1-烯-2-基)-2H-吲唑的制备
氮气保护下将Pd(dppf)Cl2.CH2Cl2(200mg,2.40mmol)加入到5-溴-3-碘-2-甲基-2H-吲唑(800mg,2.40mmol),异丙烯基硼酸频哪醇酯(390mg,2.30mmol)和碳酸铯(1.56g, 4.80mmol)的二氧六环(16mL)和水(4mL)混合溶剂中,反应在氮气保护下80℃反应1hr。冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼,减压浓缩滤液,浓缩液进行柱分离(淋洗剂:石油醚/乙酸乙酯(v/v)=5:1),得到440mg淡黄色油状物。收率:73.3%,LC-MS(APCI):m/z=251.0(M+1)。
步骤4中间体化合物5-(2-氯嘧啶-4-基)-2-甲基-3-(丙-1-烯-2-基)-2H-吲唑的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物5-(2-氯嘧啶-4-基)-2-甲基-3-(丙-1-烯-2-基)-2H-吲唑(460mg黄白色固体)。收率为92.0%。LC-MS(APCI):m/z=285.0(M+1)。
中间体化合物6-(2-氯嘧啶-4-基)咪唑并[1,2-b]哒嗪的合成。
Figure PCTCN2017094102-appb-000106
步骤1 6-氯-3-碘咪唑并[1,2-b]哒嗪的合成。
室温下,将NIS(1.50g,6.50mmol)分批加入到6-氯-3-咪唑并[1,2-b]哒嗪(1.0g,6.50mmol)的甲醇(10mL)溶液中,反应回流搅拌过夜。用饱和的硫代硫酸钠溶液淬灭反应,二氯甲烷萃取(50mLx3),合并有机层,用饱和的食盐水洗涤,无水硫酸钠干燥,减压浓缩得到1.13g黄色固体。收率为90.0%。LC-MS(APCI):m/z=290.1(M+1)。
步骤2 6-氯-3-(丙-1-烯-2-基)咪唑并[1,2-b]哒嗪的制备
氮气保护下将Pd(dppf)Cl2.CH2Cl2(240mg)加入到6-氯-3-碘咪唑并[1,2-b]哒嗪(1.0g,3.57mmol),异丙烯基硼酸频哪醇酯(602mg,3.57mmol)和碳酸钠(800mg,7.16mmol)的二氧六环(16mL)和水(4mL)混合溶剂中,反应在氮气保护下80℃反应1hr。冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼,减压浓缩滤液,浓缩液进行柱分离(淋洗剂:石油醚/乙酸乙酯(v/v)=5:1),得到509mg淡黄色油状物。收率:74.1%,LC-MS(APCI):m/z=194.1(M+1)。
步骤3中间体化合物6-(2-氯嘧啶-4-基)咪唑并[1,2-b]哒嗪的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物6-(2-氯嘧啶-4-基)咪唑并[1,2-b]哒嗪(220mg黄白色固体)。收率为27.6%。LC-MS(APCI):m/z=232.0(M+1)。
中间体化合物8-(2-氯嘧啶-4-基)-1-异丙基-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉-2(1H)-酮的合成。
Figure PCTCN2017094102-appb-000107
步骤1 3,4-二氢喹啉-1(2H)-羧酸甲酯的合成。
冰浴下,将氯甲酸甲酯(19.90mL,258mmol)缓慢滴加到1,2,3,4-四氢喹啉(10.8mL,85.8mmol)和碳酸钾(47.6g,344mmol)的无水DMF(100mL)。滴加完毕后,室温下搅拌10min,升温至50℃下反应5hrs。冷却至室温,加水(200mL)淬灭反应,用石油醚(100mLx3)萃取,合并有机层,用饱和食盐水洗涤(150mLx2)洗涤,无水硫酸钠干燥,减压浓缩得到16.3g油状物直接用于下一步反应。LC-MS(APCI):m/z=192.1(M+1)。1HNMR(400MHz,CDCl3):δ7.68(d,J=8.0Hz,1H),7.20–7.14(m,1H),7.10(d,J=6.8Hz,1H),7.03(td,J=7.5,1.0Hz,1H),3.81(s,3H),3.80–3.76(m,2H),2.79(t,J=6.6Hz,2H),1.96(dt,J=12.7,6.5Hz,2H).
步骤2 6-溴-3,4-二氢喹啉-1(2H)-羧酸甲酯的合成。
在冰浴下,将NBS(16.7g,93.83mmol)分批加入到3,4-二氢喹啉-1(2H)-羧酸甲酯(16.3g,85.3mmol)的DMF(100mL)溶液中,反应在室温下搅拌反应过夜。加水(200mL)淬灭反应,用石油醚(100mLx3)萃取,合并有机层,用饱和食盐水洗涤(150mLx2)洗涤,无水硫酸钠干燥,减压浓缩得到22.1g固体,直接用于下一步反应。LC-MS(APCI):m/z=271.1(M+1)。1H NMR(400MHz,CDCl3):δ7.61(d,J=8.0Hz,1H),7.31–7.25(m,1H),7.23(d,J=2.3Hz,1H),3.81(s,3H),3.78–3.72(m,2H),2.76(t,J=6.6Hz,2H),1.94(dt,J=12.6,6.4Hz,2H).
步骤3 6-溴-8-硝基-3,4-二氢喹啉-1(2H)-羧酸甲酯的合成。
在冰浴下,将6-溴-3,4-二氢喹啉-1(2H)-羧酸甲酯(2.69g,10.00mmol)的乙腈(50mL)溶液缓慢滴加到四氟硼酸硝(1.85g,14.00mmol)的乙腈(50mL)溶液中。反应液在是0℃下反应1.5hrs,加水淬灭反应,减压除去乙腈,水层用乙酸乙酯萃取(50mLx3),合并有机层,分别用饱和的碳酸氢钠溶液、饱和食盐水洗涤,无水硫酸钠干燥滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=5:1)得到890mg淡黄色固体,收率为:28.4%。LC-MS(APCI):m/z=315.0(M+1)。
步骤4 8-溴-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉-2(1H)-酮的合成。
将还原铁粉(860mg,15.33mmol)加入到6-溴-8-硝基-3,4-二氢喹啉-1(2H)-羧酸甲酯 (690mg,2.20mmol)的冰醋酸(8mL)溶液中。反应液在是80℃下反应2.5hrs,冷却至室温,硅藻土过滤,滤液加水稀释,用乙酸乙酯萃取(50mLx3),合并有机层,用饱和食盐水洗涤,无水硫酸钠干燥滤液减压浓缩得到粗品,粗品用正己烷打浆纯化得到340mg淡黄色固体,收率为:61.0%。LC-MS(APCI):m/z=253.0(M+1),1H NMR(400MHz,DMSO-d6):δ10.78(s,1H),7.02-6.96(m,1H),6.94(d,J=1.4Hz,1H),3.67(t,J=6.0Hz,2H),2.75(t,J=6.0Hz,2H),2.03–1.91(m,2H).
步骤5 8-溴-1-异丙基-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉-2(1H)-酮的合成。
冰浴下,将氢化钠(80mg,2.00mmol)分批加入到8-溴-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉-2(1H)-酮(340mg,1.34mmol)的无水DMF(10mL)溶液中,反应在室温下反应搅拌过夜。用水淬灭反应,乙酸乙酯萃取(30mLx3),合并有机层,用饱和的食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=1/1)得到300mg淡黄色固体,收率为:76.2%。LC-MS(APCI):m/z=295.0(M+1)。
步骤6中间体化合物8-(2-氯嘧啶-4-基)-1-异丙基-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉-2(1H)-酮的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物8-(2-氯嘧啶-4-基)-1-异丙基-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉-2(1H)-酮(300mg白色固体)。收率为67.3%。LC-MS(APCI):m/z=329.0(M+1);1HNMR(400MHz,CDCl3):δ8.67(d,J=5.3Hz,1H),7.86(s,1H),7.74(s,1H),7.71(d,J=5.4Hz,1H),4.86(dt,J=14.0,7.0Hz,1H),4.00(t,J=6.0Hz,2H),3.04(t,J=6.0Hz,2H),2.32–2.22(m,2H),1.71(d,J=7.0Hz,6H).
中间体化合物8-(2-氯嘧啶-4-基)-2-甲氧基-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉的合成。
Figure PCTCN2017094102-appb-000108
步骤1 8-溴-2-氯-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉的合成。
室温下,将8-溴-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉-2(1H)-酮(2.60g,10.31mmol)溶入三氯氧磷(30mL)和甲苯(10mL)的混合溶液中,反应回流反应过夜天,冷却至室温,减压除去反应液,残渣用饱和碳酸氢钠溶液调pH至中性,乙酸乙酯(50mLx3)萃取,有机相分别用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1),得到1.46g白色固体固体。收率为52.4%。LC-MS(APCI): m/z=270.9(M+1)。
步骤2 8-溴-2-甲氧基-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉的合成。
冰浴下,将甲醇钠(730mg,13.5mmol)加入到8-溴-2-氯-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉(750mg,2.77mmol)的无水DMF(15mL),自然升温,反应搅拌过夜。加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=5:1),得到560mg淡黄色固体,收率为76.0%。LC-MS(APCI):m/z=267.0(M+1)。
步骤3中间体化合物8-(2-氯嘧啶-4-基)-2-甲氧基-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉的合成。
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物8-(2-氯嘧啶-4-基)-2-甲氧基-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉(300mg黄色固体)。收率为66.4%。LC-MS(APCI):m/z=301.0(M+1)。
中间体化合物6-(2-氯嘧啶-4-基)苯并[d]噻吩的合成。
Figure PCTCN2017094102-appb-000109
合成步骤与中间体化合物6-(2-氯嘧啶-4-基)-4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑合成步骤5相同,得到产物6-(2-氯嘧啶-4-基)苯并[d]噻吩(120mg黄色固体)。收率为66.4%。LC-MS(APCI):m/z=248.0(M+1)。
中间体化合物N-(5-氨基-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
Figure PCTCN2017094102-appb-000110
步骤1化合物(4-氟-2-甲氧基-5-硝基苯基)氨基甲酸叔丁酯的合成。
在冰浴下,将Boc酸酐(7.05g,32.20mmol)缓慢滴加到4-氟-2-甲氧基-5-硝基苯胺(4.0g,21.50mmol)和三乙胺(TEA,3.60g,35.64mmol)的二氯甲烷(65mL)混合液中,滴加完毕后,反应液在40℃反应过夜。冷却至室温,加水(100mL)稀释,用二氯甲烷(100mLx3)萃取,合并有机相,饱和食盐水(100mL)洗涤,无水硫酸钠干燥,除去溶剂,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=2:1),得到黄固体3.6g,收率:58.8%。LC-MS(APCI):m/z=287.2(M+1)。
步骤2化合物(4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基-5-硝基苯基)氨基甲酸叔丁酯的合成。
将N1,N1,N2-三甲基乙烷-1,2-二胺(2.14g,20.40mmol)加入到(4-氟-2-甲氧基-5-硝基苯)氨基甲酸叔丁酯(4.0g,21.50mmol)和N,N-二异丙基乙胺(DIPEA,4.92mL,27.20mmol)的N,N-二甲基甲酰胺(28mL)混合液中,反应液在90℃反应5小时。冷却至室温,加水(100mL)稀释,用乙酸乙酯(100mLx3)萃取,合并有机相,饱和食盐水(100mLx3)洗涤,无水硫酸钠干燥,除去溶剂得到5.0g棕色油状物,不经纯化直接用于下一步反应。LC-MS(APCI):m/z=369.3(M+1)。
步骤3化合物(5-氨基-4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基苯基)氨基甲酸叔丁酯的合成。
将Pd/C(500mg)加入到(4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基-5-硝基苯基)氨基甲酸叔丁酯(5.00g,13.6mmol)的甲醇(65mL)溶液中,氢气球下搅拌反应过夜。硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=20:1),得到棕黄色油状物4.0g,两步收率:86.8%。LC-MS(APCI):m/z=339.3(M+1)。
步骤4化合物(5-丙烯酰胺基-4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基苯基) 氨基甲酸叔丁酯的合成。
室温下将3-氯丙酰氯(1.80g,14.20mmol)加入到(5-氨基-4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基苯基)氨基甲酸叔丁酯(4.00g,11.80mmol)的四氢呋喃(95mL)和水(9.5mL)的混合溶液中,室温下反应1小时。将氢氧化钠(2.40g)加入到反应液中,反应在65℃反应过夜。冷却至室温,减压浓缩反应液。残渣加水(100mL)稀释,用二氯甲烷(100mLx3)萃取,合并有机相,饱和食盐水(100mL)洗涤,无水硫酸钠干燥,除去溶剂,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=20:1),得到棕黄色固体3.20g,收率:69.2%。LC-MS(APCI):m/z=393.3(M+1)。
步骤5中间体化合物N-(5-氨基-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
室温下将氯化氢的甲醇溶液(4M,5mL)加入到((5-丙烯酰胺基-4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基苯基)氨基甲酸叔丁酯(392mg,1.00mmol)的甲醇(2mL)中,反应搅拌反应过夜,减压除去甲醇,加入饱和的碳酸氢钠(30mL),二氯甲烷(30mLx3)萃取,合并有机相,饱和食盐水(30mL)洗涤,无水硫酸减压浓缩得到黄固体292mg,收率:95%。LC-MS(APCI):m/z=293.3(M+1)。
中间体化合物5-(二氟甲氧基)-N1-(2-(二甲基氨基)乙基)-N1-甲基-2-硝基苯-1,4-二胺的合成。
Figure PCTCN2017094102-appb-000111
步骤1 2-(二氟甲氧基)-4-氟-1-硝基苯的合成。
室温下,将碳酸钾(6.60g,47.7mmol)加入到5-氟-2-硝基苯酚(5.0g,31.8mmol)和二氟氯乙酸钠(7.30g,47.7mmol)的DMF(100mL)溶液中,反应液在90℃下反应5hrs。减压除去DMF,加水(200mL)稀释,用二氯甲烷(200mLx3)萃取,合并有机层,用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=30:1),得到4.17g黄色油状物。收率为63.3%。
步骤2 2-(二氟甲氧基)-4-氟苯胺的合成
将10%的Pd/C(400mg)加入到2-(二氟甲氧基)-4-氟-1-硝基苯(4.17g,20.1mmol)的甲醇(40mL)溶液中,氢气(气球)下室温反应过夜,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=50:1),得到2.982g淡黄色油状物,收率 为83.7%。LC-MS(APCI):m/z=178.1M+1)。
步骤3 2-(二氟甲氧基)-4-氟-5-硝基苯胺的合成
0℃下,将2-(二氟甲氧基)-4-氟苯胺(1.5g)溶于3ml浓硫酸中,分批加入硝酸钾(1.1g),室温搅拌反应,LC-MS检测反应完毕后,将体系缓慢倒入100ml饱和NaHCO3冰水中,有固体析出,抽滤,滤液加入60ml二氯甲烷萃取3次,滤饼溶于二氯甲烷萃取液后再次过滤,无水硫酸纳干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=30:1),得到1.3g褐色油状物,收率为69.1%。LC-MS(APCI):m/z=223.1M+1)。
步骤4中间体化合物5-(二氟甲氧基)-N1-(2-(二甲基氨基)乙基)-N1-甲基-2-硝基苯-1,4-二胺的合成
室温下,将碳酸钾(2.0g,14.4mmol)加入到2-(二氟甲氧基)-4-氟-5-硝基苯胺(1.6g,7.2mmol)和N,N,N’-三甲基乙二胺(1.1g,10.8mmol)的乙腈(20mL)中。反应在80℃下反应3hrs。冷却至室温。硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1),得到2.0g红褐色油固体,收率为45.5%。LC-MS(APCI):m/z=305.2M+1)。
中间体化合物N1-(2-(二甲基氨基)乙基)-5-乙氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
Figure PCTCN2017094102-appb-000112
步骤1 2-乙氧基-4-氟-1-硝基苯的合成。
将5-氟-2-硝基苯酚(5.0g,31.8mmol),碳酸钾(13.17g,95.4mmol)混悬于150mLDMF中,滴入碘乙烷(5.00mL,63.6mmol),升温至370℃搅拌反应,TLC检测反应完毕后,将体系倒入冰水中,用乙酸乙酯(200mLx3)萃取,合并有机层,用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=20:1),得到4.8g黄色油状物。收率为83.1%。
步骤2 2-乙氧基-4-氟苯胺的合成
将10%的Pd/C(400mg)加入到2-乙氧基-4-氟-1-硝基苯(4.8g,26.0mmol)的甲醇(40mL)溶液中,氢气(气球)下室温反应过夜,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=50:1),得到产物3.8g淡黄色油状物, LC-MS(APCI):m/z=156.1(M+1)。1H NMR(400MHz,CDCl3)δ6.63(dd,J=8.5,5.8Hz,1H),6.56(dd,J=10.4,2.7Hz,1H),6.50(td,J=8.5,2.7Hz,1H),4.04(q,J=7.0Hz,2H),3.42(br,2H),1.45(t,J=7.0Hz,3H).
步骤3 2-乙氧基-4-氟-5-硝基苯胺的合成
0℃下,将2-乙氧基-4-氟苯胺(2.0g.12.90mmol)溶于10ml浓硫酸中,将硝酸钾(1.56g,15.5mmol)分批加入体系,然后自然升至室温搅拌,LC-MS检测反应完毕后处理反应,将体系缓慢倒入100ml冰水中,滴入浓氨水调pH值至7-8,加入乙酸乙酯萃取数次,有机相采用无水硫酸钠干燥,减压浓缩后得到512mg褐色固体,收率为20.0%。LC-MS(APCI):m/z=201.0M+1)。
步骤4中间体化合物N1-(2-(二甲基氨基)乙基)-5-乙氧基-N1-甲基-2-硝基苯-1,4-二胺的合成
室温下,将碳酸钾(983mg,7.12mmol)加入到2-(乙氧基)-4-氟-5-硝基苯胺(512mg,3.56mmol)和N,N,N’-三甲基乙二胺(545mg,5.34mmol)的乙腈(10mL)中。反应在80℃下反应3hrs。冷却至室温。硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1),得到635mg红褐色油油状物,收率为63.3%,LC-MS(APCI):m/z=283.2(M+1)。
中间体化合物N1-(2-(二甲基氨基)乙基)-N1-甲基-2-硝基-5-(2,2,2-三氟乙氧基)苯-1,4-二胺的合成。
Figure PCTCN2017094102-appb-000113
步骤1 4-氟-1-硝基-2-(2,2,2-三氟乙氧基)苯的合成。
将2,4-二氟-1-硝基苯(10g,62.9mmol),碳酸铯(20.5g,62.9mmol)混悬于100mLTHF中,滴入三氟乙醇(6.29g,62.9mmol),反应在室温下反应过夜,TLC检测反应完毕后,将体系倒入冰水中,用乙酸乙酯(200mLx3)萃取,合并有机层,用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=20:1),得到15g黄色固体。收率为99.8%。
步骤2 4-氟-2-(2,2,2-三氟乙氧基)苯胺的合成
将4-氟-1-硝基-2-(2,2,2-三氟乙氧基)苯(15g,62.76mmol)溶入乙醇和水的混合溶剂 中(50mL,v/v=4/1),加入还原铁粉(21g,0.38mol)和氯化铵(10g,0.19mol),反应液升温至回流2.5hrs,冷却至室温,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=20:1),得到产物11.8g黄色油状物。收率为85.9%。LC-MS(APCI):m/z=210.1(M+1).
步骤3 4-氟-5-硝基-2-(2,2,2-三氟乙氧基)苯胺的合成
0℃下,将4-氟-2-(2,2,2-三氟乙氧基)苯胺(3.00g.14.35mmol)溶于10ml浓硫酸中,将硝酸钾(1.70g,17.22mmol)分批加入体系,然后自然升至5℃搅拌,LC-MS检测反应完毕后处理反应,将体系缓慢倒入100ml冰水中,加入乙酸乙酯萃取数次,有机相采用无水硫酸钠干燥,减压浓缩后得到3.2g褐色固体。LC-MS(APCI):m/z=255.0M+1)。
步骤4中间体化合物N1-(2-(二甲基氨基)乙基)-N1-甲基-2-硝基-5-(2,2,2-三氟乙氧基)苯-1,4-二胺的合成
室温下,将碳酸钾(3.5g,25.2mmol)加入到4-氟-5-硝基-2-(2,2,2-三氟乙氧基)苯胺(3.2g,12.6mmol)和N,N,N’-三甲基乙二胺(1.90g,18.9mmol)的乙腈(20mL)中。反应在80℃下反应3hrs。冷却至室温。硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1),得到3.2g红褐色油油状物,收率为75.6%,LC-MS(APCI):m/z=337.1M+1)。
中间体化合物N2-(2-(二甲基氨基)乙基)-6-甲氧基-N2-甲基-3-硝基苯-2,5-二胺的合成。
Figure PCTCN2017094102-appb-000114
步骤1 6-氯-2-甲氧基-3-硝基吡啶的合成。
冰浴下。将氢化钠(1.0g,25mmol,60%)分批加入到2,6-二氯-3-硝基吡啶(4.80g,25mmol)和无水甲醇(800mg)的无水四氢呋喃(60mL),反应缓慢升温至室温下反应过夜,TLC检测反应完毕后,减压浓缩去除四氢呋喃,将体系倒入冰水中,用乙酸乙酯(100mLx3)萃取,合并有机层,用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=30:1),得到3g黄色固体。收率为64%。
步骤2 6-氯-2-甲氧基吡啶-3-胺的合成
将6-氯-2-甲氧基-3-硝基吡啶(1.00g,5.32mmol)溶入乙醇和水的混合溶剂中(40mL,v/v=4/1),加入还原铁粉(1.89g,31.92mol)和氯化铵(846mg,15.96mol),反应液升温至回流1.5hrs,冷却至室温,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=10:1),得到0.8g黄色固体。收率为94.6%。LC-MS(APCI):m/z=159.0(M+1).
步骤3N-(6-氯-2-甲氧基吡啶-3-基)乙酰胺的合成
冰浴下。将乙酰氯(397mg,5.06mmol)的无水二氯甲烷(10mL)滴加到6-氯-2-甲氧基吡啶-3-胺(800mg,5.06mmol)和DIPEA(981mg,7.59mmol)的无水二氯甲烷(30mL)中,反应搅拌1.5hrs。反应液依次用50ml水,50mL 1N稀盐酸及80ml饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,减压蒸干,得到1.0g淡黄色固体。收率为98.8%,LC-MS(APCI):m/z=201.1(M+1)。
步骤4:N-(6-氯-2-甲氧基-5-硝基吡啶-3-基)乙酰胺的合成
向100ml单口瓶中加入N-(6-氯-2-甲氧基吡啶-3-基)乙酰胺(1.10g,5.50mmol)及10ml三氟醋酸酐,冰盐浴下冷却至-10℃,滴加发烟硝酸(0.1ml),控制温度在-5℃以下,滴加完毕,继续在冰盐浴下反应2小时。将反应物缓慢加入碎冰中,析出固体,过滤。得到的粗产品于60℃烘干,乙酸乙酯打浆纯化,得到1.0g黄色固体。收率为73.9%,LC-MS(APCI):m/z=246.1(M+1)。1H NMR(400MHz,DMSO-d6):δ9.92(s,1H),9.18(s,1H),4.07(s,3H),2.17(s,3H).
步骤5N-(6-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基-5-硝基吡啶-3-基)乙酰胺的合成
N-(6-氯-2-甲氧基-5-硝基吡啶-3-基)乙酰胺(1.00g,4.10mmol)和N,N,N’-三甲基乙二胺(630mg,6.20mmol)的乙腈(30mL)溶液在80℃下反应3hrs。冷却至室温。硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1),得到1.2g红褐色油状物,收率为93.8%,LC-MS(APCI):m/z=312.2(M+1)。
步骤6中间体化合物N2-(2-(二甲基氨基)乙基)-6-甲氧基-N2-甲基-3-硝基苯-2,5-二胺的合成
将浓盐酸(0.6mL)加入到N-(6-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基-5-硝基吡啶-3-基)乙酰胺(1.2g,3.85mmol)的甲醇(30mL)中,60℃反应过夜。将反应液减压蒸干,加入200ml二氯甲烷和160ml饱和碳酸氢钠溶液,搅拌至气泡后分出有机层,无水硫酸纳干燥,过滤,减压浓缩。浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1),得到320mg红褐色油状物,收率为30.8%,LC-MS(APCI):270.1(M+1)。
实施例化合物的合成
实施例1 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-氯-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000115
步骤1化合物6-(2,5-二氯嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑的合成
取化合物2,4,5-三氯嘧啶(206mg,1.12mmol)、化合物4-氟-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(357mg,1.12mmol),Pd(PPh3)2Cl2(43mg,0.06mmol)加入到单口反应瓶中,氮气置换三次,在氮气保护下先后加入DMF(5mL)和碳酸钠水溶液(2M,2mL),60摄氏度搅拌反应2小时。关闭加热,反应液冷却至室温,倒入50mL水中,乙酸乙酯(5mL*2)萃取,合并有机相,减压蒸干溶剂,使用制备级薄层色谱纯化(PE/EA,V/V,1.5/1),得到白色固体,共118mg,收率为31%。
步骤2化合物5-氯-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺的合成。
将化合物6-(2,5-二氯嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑(118mg,0.35mmol),4-氟-2-甲氧基-5-硝基苯胺(78mg,0.42mmol)、Xant-phos(20mg,0.035mmol)、Pd2(dba)3(16mg,0.017mmol)以及碳酸铯(283mg,0.87mmol)于氮气保护下加入到单口反应瓶中,之后加入无水二氧六环(2mL),氮气置换三次,90摄氏度加热搅拌反应过夜。减压蒸干溶剂,制备级薄层色谱纯化(PE/EA,V/V,1/1)得到灰色固体,共30mg。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-氯-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
将化合物5-氯-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺(30mg,0.061mmol)加入乙腈(5mL)中分散均匀,先后加入三甲基乙二胺(13mg,0.123mmol)、碳酸钾(17mg,0.123mmol),加热至90摄氏度并回流反应1.5小时。将反应液冷却至室温,过滤去除不溶性固体,滤液蒸干得红色油状物。将红 色油状物溶于乙醇-水(4mL+1mL),加入还原铁粉(42mg)和氯化铵(13mg),90摄氏度加热回流反应5小时。冷却反应至室温,过滤去除不溶性固体,滤液蒸干,所得油状物加入10mL二氯甲烷(DCM)溶解,加入饱和碳酸氢钠水溶液2mL,冰浴冷却。向以上两相体系滴加1M的丙烯酰氯(0.12mL),冰浴下反应30分钟。分液,有机相蒸干,制备级薄层色谱纯化(DCM/MeOH,V/V,30/1),得到灰绿色固体,共10mg。LC-MS(APCI):m/z=596.3(M+1)。
实施例2 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-甲基-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000116
步骤1化合物6-(2-氯-5-甲基嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑的合成。
取化合物2,4–二氯-5-甲基嘧啶(200mg,1.23mmol)、化合物4-氟-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(391mg,1.23mmol),Pd(PPh3)2Cl2(52mg,0.074mmol)加入到单口反应瓶中,氮气置换三次,在氮气保护下先后加入DMF(5mL)和碳酸钠水溶液(2M,2.5mL),80摄氏度搅拌反应3.5小时。关闭加热,反应液冷却至室温,倒入50mL水中,乙酸乙酯(5mL*2)萃取,合并有机相,减压蒸干溶剂,制备级薄层色谱纯化(PE/EA,V/V,2/1),得到白色固体,共210mg,收率为53.6%。
步骤2化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)-5-甲基嘧啶-2-胺的合成。
将化合物6-(2-氯-5-甲基嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑(210mg,0.66mmol),4-氟-2-甲氧基-5-硝基苯胺(147mg,0.79mmol)、pTSA(454mg,2.64mmol)于氮气保护下加入到单口反应瓶中,之后加入无水二氧六环(2mL),氮气置换三次,100摄氏度加热搅拌反应24小时。减压蒸干溶剂,制备级薄层色谱纯化(PE/EA,V/V,1/1),得到灰色固体,共162mg。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-甲基-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
将化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)-5-甲基嘧啶-2-胺(149mg,0.32mmol)加入乙腈(5mL)分散均匀,先后加入三甲基乙二胺(39mg,0.38mmol)、碳酸钾(88mg,0.64mmol),90摄氏度加热回流反应2小时。将反应也冷却至室温,过滤去除不溶性固体,滤液蒸干得红色油状物。将红色油状物溶于乙醇-水(4mL+1mL),加入还原铁粉(106mg)和氯化铵(34mg),90摄氏度加热回流反应5小时。冷却反应至室温,过滤去除不溶性固体,滤液蒸干,所得油状物加入10mLDCM溶解,加入饱和碳酸氢钠水溶液5mL,冰浴冷却。向以上两相体系滴加0.15M的丙烯酰氯(3mL),冰浴下反应30分钟。分液,有机相蒸干,制备级薄层色谱纯化(DCM/MeOH,V/V,20/1)得到白色固体,共30mg。1H NMR(400MHz,CDCl3)δ9.56(s,1H),9.49(s,1H),8.40(s,1H),7.64–7.50(m,2H),7.40–7.27(m,2H),6.91(s,1H),6.69(s,1H),6.43(d,J=16.8Hz,1H),5.69(d,J=10.4Hz,1H),4.71(dt,J=13.8,6.9Hz,1H),3.85(s,3H),3.13(s,2H),2.82(s,2H),2.68(s,3H),2.66(s,3H),2.58(s,6H),2.29(s,3H),1.63(d,J=6.9Hz,6H)。
实施例3 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-甲氧基-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000117
步骤1化合物6-(2-氯-5-甲氧基嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑的合成。
取化合物2,4–二氯-5-甲氧基嘧啶(192mg,1.07mmol)、化合物4-氟-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(310mg,0.97mmol),Pd(PPh3)2Cl2(34mg,0.05mmol)加入到单口反应瓶中,氮气置换三次,在氮气保护下先后加入DMF(5mL)和碳酸钠水溶液(2M,2mL),60摄氏度搅拌反应3小时。关闭加热,反应液冷却至室温,倒入50mL水中,乙酸乙酯(5mL*2)萃取,合并有机相,无水硫酸 钠干燥,减压蒸干溶剂粗品220mg。
步骤2化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)-5-甲氧基嘧啶-2-胺的合成。
将化合物6-(2-氯-5-甲氧基嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑(220mg,0.79mmol),4-氟-2-甲氧基-5-硝基苯胺(145mg,0.79mmol)、Xant-phos(40mg,0.066mmol)、Pd2(dba)3(26mg,0.031mmol)以及碳酸铯(537mg,1.64mmol)于氮气保护下加入到单口反应瓶中,之后加入无水二氧六环(10mL),氮气置换三次,90摄氏度加热搅拌反应过夜。过滤去除不溶性固体,合并滤液减压蒸干溶剂,所得固体加入DCM-PE(V/V,1/4)回流10min,自然降温冷却过滤收集固体得灰色固体,共257mg,收率为67%。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-甲氧基-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
将化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)-5-甲氧基嘧啶-2-胺(257mg,0.53mmol)加入乙腈(5mL)分散均匀,先后加入三甲基乙二胺(65mg,0.64mmol)、碳酸钾(147mg,1.06mmol),90摄氏度加热回流反应1.5小时。将反应液冷却至室温,过滤去除不溶性固体,滤液蒸干得红色油状物。将红色油状物溶于乙醇-水(4mL+1mL),加入还原铁粉(178mg)和氯化铵(57mg),90摄氏度加热回流反应4.5小时。冷却反应至室温,过滤去除不溶性固体,滤液蒸干,所得油状物加入10mL DCM溶解,加入饱和碳酸氢钠水溶液2mL,冰浴冷却。向以上两相体系滴加1M的丙烯酰氯(1mL),冰浴下反应30分钟。分液,有机相蒸干,制备级薄层色谱纯化(DCM/MeOH,V/V,30/1),得到灰白色固体,共80mg。1H NMR(400MHz,CDCl3)(δ/ppm)9.50(s,1H),9.40(s,1H),8.32(s,1H),8.16(s,1H),7.85(d,J=11.9Hz,1H),7.54(s,1H),6.68(s,1H),6.44(d,J=16.8Hz,1H),5.82–5.63(m,1H),4.70(dt,J=13.8,6.9Hz,1H),3.88(s,3H),3.87(s,3H),3.21(t,J=5.5Hz,2H),2.95(t,J=27.9Hz,2H),2.69(s,3H),2.67(d,J=4.3Hz,3H),2.65(s,3H),2.16(d,J=0.5Hz,3H),1.65(d,J=6.9Hz,6H)。
实施例4 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-三氟甲基-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000118
步骤1化合物4-氯-N-(4-氟-2-甲氧基-5-硝基苯基)-5-(三氟甲基)嘧啶-2-胺的合成。
取化合物2,4–二氯-5-三氟甲基嘧啶(700mg,3.23mmol)加15mL无水叔丁醇搅拌溶解,先后加入4-氟-2-甲氧基-5-硝基苯胺(600mg,3.23mmol)和二异丙基乙基胺(417mg,3.23mmol),之后加热至60摄氏度反应过夜。冷却反应至室温,过滤收集固体(244mg)。
步骤2化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)-5-(三氟甲基)嘧啶-2-胺的合成。
取化合物4-氯-N-(4-氟-2-甲氧基-5-硝基苯基)-5-(三氟甲基)嘧啶-2-胺(244mg,0.66mmol)、化合物4-氟-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(233mg,0.73mmol),Pd(PPh3)2Cl2(28mg,0.04mmol)加入到单口反应瓶中,氮气置换三次,在氮气保护下先后加入DMF(6mL)和碳酸钠水溶液(2M,1.4mL),60摄氏度搅拌反应2小时。关闭加热,反应液冷却至室温,倒入50mL水中,乙酸乙酯(5mL*2)萃取,合并有机相,无水硫酸钠干燥,减压蒸干溶剂得粗品,PE/EA(石油醚/乙酸乙酯:3mL/3mL)回流打浆得纯品107mg。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-三氟甲基-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
将化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)-5-(三氟甲基)嘧啶-2-胺(107mg)加入乙腈(5mL)分散均匀,先后加入三甲基乙二胺(25mg,0.25mmol)、碳酸钾(57mg,0.41mmol),90摄氏度加热回流反应0.5小时。将反应液冷却至室温,过滤去除不溶性固体,滤液蒸干得红色油状物。将红色油状物溶于乙醇-水(4mL+1mL),加入还原铁粉(50mg)和氯化铵(17mg),90摄氏度加热回流反应3小时。冷却反应至室温,过滤去除不溶性固体,滤液蒸干,所得油状物加入10mL DCM溶解,加入饱和碳酸氢钠水溶液2mL,冰浴冷却。向以上两相体系滴加1M的丙烯酰氯(0.4mL),冰浴下反应30分钟。分液,有机相蒸干,制备级薄层色谱纯化(DCM/MeOH,V/V,30/1),得灰白色固体(40mg)。1H NMR(400MHz,CDCl3)(δ/ppm)9.62(s,1H),9.50 (s,1H),8.63(s,1H),8.42(d,J=1.2Hz,1H),8.21(d,J=11.8Hz,1H),7.60(s,1H),6.91(s,1H),6.77(s,1H),6.48(dd,J=16.9,1.6Hz,1H),6.40–5.72(m,1H),5.71(dd,J=10.2,1.6Hz,1H),4.72(dt,J=14.0,7.0Hz,1H),3.89(s,3H),3.19(s,2H),2.89(s,2H),2.73(s,3H),2.65(s,3H),2.63(s,6H),1.65(d,J=7.0Hz,6H)。
实施例5 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(5-氟-1,1-二甲基-2,3-二氢-1H-苯并[d]咪唑[1,2-a]吡咯-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000119
步骤1化合物5,5-二甲基吡咯烷-2-酮的合成。
取丙烯酸乙酯(1.24g,12.4mmol)、2-硝基丙烷(1.10g,12.4mmol)和DBU(1,8-二氮杂二环十一碳-7-烯,189mg,1.24mmol)溶于25mL无水四氢呋喃,加热回流反应过夜。冷却反应至室温,加入4N盐酸20mL,EA 20mL,分液,有机相无水硫酸钠干燥。抽滤,滤液减压蒸干得黄绿色油状物。以上油状物溶于30mL无水四氢呋喃,加入5%的Pd/C(234mg),通入4个大气压的氢气室温反应72h。抽滤去除钯碳,滤液减压蒸干得到粗品(1.4g)。
步骤2化合物(Z)-N-(4-溴-2,6-二氟苯基)-5,5-二甲基吡咯烷-2-亚胺的合成。
将化合物5,5-二甲基吡咯烷-2-酮溶于30mL甲苯,先后加入4-溴-2,6-二氟苯胺(2.8g,12.4mmol)、三乙胺(1.3g,12.4mmol)和三氯氧磷(1.9g,12.4mmol),之后于110摄氏度加热回流反应5小时。冰浴冷却反应液,加入EA 10mL,饱和碳酸氢钠水溶液20mL,搅拌10分钟,分液。有机相蒸干柱层析(PE/EA,v/v,2/1),得化合物53(2.27g)。
步骤3化合物7-溴-5-氟-1,1-二甲基-2,3-二氢-1H-苯并[d]吡咯并[1,2-a]咪唑的合成。
将化合物(Z)-N-(4-溴-2,6-二氟苯基)-5,5-二甲基吡咯烷-2-亚胺溶于20mL二甲基亚砜,一次性加入氢氧化钾(0.84g,14.98mmol),氮气保护下加热至100摄氏度搅拌反应过夜。冷却反应液至室温,倒入200mL水中,EA(10mL*2)萃取,有机相蒸干柱层析(PE/EA,v/v,3/1),得到689mg产物。1H NMR(400MHz,CDCl3)δ7.33(s,1H),7.09(d,J=9.8Hz,1H),3.13–3.03(m,2H),2.54(t,J=7.6Hz,2H),1.63(s,6H)。
步骤4化合物5-氟-1,1-二甲基-7-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-2,3-二氢-1H-苯并[d]吡咯并[1,2-a]咪唑的合成。
将化合物7-溴-5-氟-1,1-二甲基-2,3-二氢-1H-苯并[d]吡咯并[1,2-a]咪唑(698mg,2.43mmol)、连硼酸频哪醇酯(680mg,2.68mmol)、三环己基膦(115mg,0.41mmol)和醋酸钾(715mg,7.29mmol)于氮气保护下加入到5mL无水二甲基亚砜中,搅拌均匀,氮气置换三次,加入醋酸钯(60mg,0.27mmol),氮气置换三次,90摄氏度加热搅拌反应45分钟。冷却反应至室温,倒入50mL水中,EA-PE(v/v,1/1,10mL*2)萃取,合并有机相蒸干得到粗品直接用于下一步。
步骤5化合物7-(2-氯嘧啶-4-基)-5-氟-1,1-二甲基-2,3-二氢-1H-苯并[d]吡咯并[1,2-a]咪唑的合成。
氮气保护下,将化合物5-氟-1,1-二甲基-7-(4,4,5,5-四甲基-1,3,2-二氧硼杂环戊烷-2-基)-2,3-二氢-1H-苯并[d]吡咯并[1,2-a]咪唑、2,4-二氯嘧啶(362mg,2.43mmol)、2M的碳酸钠水溶液(4.8mL)加入到8mL DMF中,氮气置换三次,加入Pd(PPh3)2Cl2(84mg,0.12mmol),氮气置换三次,之后60摄氏度加热搅拌反应1小时。冷却反应体系至室温,倒入到50mL水中,EA(10mL*2)萃取。合并有机相,水洗,饱和食盐水洗,之后无水硫酸钠干燥。抽滤,收集滤液减压蒸干得到粗品(793mg),直接用于下一步。
步骤6化合物4-(5-氟-1,1-二甲基-2,3-二氢-1H-苯并[d]吡咯并[1,2-d]咪唑-7-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺的合成。
将化合物7-(2-氯嘧啶-4-基)-5-氟-1,1-二甲基-2,3-二氢-1H-苯并[d]吡咯并[1,2-a]咪唑溶于无水10mL二氧六环,先后加入4-氟-2-甲氧基-5-硝基苯胺(466mg,2.5mmol)、pTSA(对甲苯磺酰胺,1.72g,10mmol),加热至100摄氏度搅拌反应过夜。蒸干二氧六环,加入DCM 20mL,饱和碳酸氢钠水溶液20mL,搅拌10分钟,抽滤,分液。有机相蒸干柱层析(DCM/MeOH,v/v,80/1),浓缩所得固体经PE-EA(v/v,1/1,20mL),回流打浆1小时纯化,得到纯品(414mg)。
步骤7化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(5-氟-1,1-二甲基-2,3-二氢-1H-苯并[d]咪唑[1,2-a]吡咯-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
将化合物4-(5-氟-1,1-二甲基-2,3-二氢-1H-苯并[d]吡咯并[1,2-d]咪唑-7-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺加入乙腈(10mL)分散均匀,先后加入三甲基乙二胺(109mg,1.06mmol)、碳酸钾(246mg,1.78mmol),90摄氏度加热回流反应2小时。将反应液冷却至室温,过滤去除不溶性固体,滤液蒸干得红色油状物。将红色油状物溶于乙醇-水(8mL+2mL),加入还原铁粉(199mg)和氯化铵(95mg),100摄氏度加热回流反应4.5小时。冷却反应至室温,过滤去除不溶性固体,滤液蒸干,所得油状物加入 10mL DCM溶解,加入饱和碳酸氢钠水溶液10mL,冰浴冷却。向以上两相体系滴加1M的丙烯酰氯(1.7mL),冰浴下反应30分钟。分液,有机相蒸干,制备级薄层色谱纯化(DCM/MeOH,V/V,20/1),得灰白色固体(90mg)。1H NMR(400MHz,CDCl3)(δ/ppm)9.49(s,1H),9.39(s,1H),8.51(d,J=5.3Hz,1H),7.95(d,J=1.3Hz,1H),7.82–7.71(m,1H),7.62(d,J=8.0Hz,1H),7.15(d,J=5.3Hz,1H),6.69(s,1H),6.45(dd,J=16.8,1.9Hz,1H),5.72(dd,J=10.2,1.9Hz,1H),3.89(s,3H),3.29(t,J=5.4Hz,2H),3.11(t,J=7.6Hz,4H),2.78(s,6H),2.72(s,3H),2.63–2.48(m,2H),1.69(s,6H)。
实施例6 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-2-甲氧基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000120
步骤1化合物4-(4-氟-1-异丙基-2-甲氧基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺的合成。
氮气保护下,将中间体化合物4-氟-1-异丙基-2-甲氧基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基-1H-苯并[d]咪唑、2,4-二氯嘧啶(478mg,1.43mmol)、2M的碳酸钠水溶液(2.8mL)加入到5mL DMF中,氮气置换三次,加入Pd(PPh3)2Cl2(50mg,0.072mmol),氮气置换三次,之后60摄氏度加热搅拌反应35分钟。冷却反应体系至室温,倒入到50mL水中,EA(10mL*2)萃取。合并有机相,水洗,饱和食盐水洗,之后无水硫酸钠干燥。抽滤,收集滤液减压蒸干,得到灰色固体的粗品,直接用于下一步。氮气保护下将上述灰色固体溶于15mL无水二氧六环,先后加入4-氟-2-甲氧基-5-硝基苯胺(266mg,1.43mmol)、Xant-phos(42mg,0.072mmol)、碳酸铯(1.16g,3.58mmol),氮气置换3次,加入Pd2(dba)3(66mg,0.072mmol),氮气置换3次,90摄氏度搅拌反应过夜。过滤去除不溶性固体,蒸干二氧六环柱层析(DCM)得到纯品(293mg)。
步骤2化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-2-甲氧基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
取化合物4-(4-氟-1-异丙基-2-甲氧基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺(157mg,0.334mmol)加入乙腈(10mL)分散均匀,先后加入三甲基乙二胺(51mg)、碳酸钾(69mg),90摄氏度加热回流反应2.5小时。将反应液冷却至室温,过滤去除不溶性固体,滤液蒸干得红色油状物。将红色油状物溶于乙醇-水(8mL+2mL), 加入还原铁粉(112mg)和氯化铵(37mg),100摄氏度加热回流反应过夜。冷却反应至室温,过滤去除不溶性固体,滤液蒸干,所得油状物加入5mL DCM溶解,加入饱和碳酸氢钠水溶液5mL,冰浴冷却。向以上两相体系滴加1M的丙烯酰氯(0.7mL),冰浴下反应30分钟。分液,有机相蒸干,制备级薄层色谱纯化(DCM/MeOH,V/V,30/1),得产物80mg。1H NMR(400MHz,CDCl3)(δ/ppm)9.59(s,1H),8.49(d,J=5.3Hz,1H),7.90(d,J=1.2Hz,1H),7.83–7.55(m,2H),7.11(t,J=8.4Hz,1H),6.73(s,1H),6.45(dd,J=16.9,1.8Hz,1H),5.71(d,J=11.8Hz,1H),4.74(dt,J=13.7,6.8Hz,1H),4.25(d,J=3.1Hz,3H),3.91(s,3H),3.25–3.08(m,2H),2.72(s,3H),2.59(s,6H),1.56(dd,J=6.9,2.6Hz,6H)。
实施例7 N-(2-((2-(二甲基氨基)乙氧基)-5-((4-(4-氟-1-异丙基-2-甲氧基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000121
取2-二甲基氨基乙醇(130mg,1.456mmol)加入无水二氧六环(2.5mL)溶解,先后加入钠氢(58mg,1.456mmol)、4-(4-氟-1-异丙基-2-甲氧基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺(137mg,0.291mmol),室温搅拌75分钟。滴加饱和氯化铵水溶液0.5mL淬灭反应,加水50mL,EA(10mL*2)萃取,合并有机相无水硫酸钠干燥蒸干得红色油状物。将红色油状物溶于乙醇-水(10mL+2.5mL),加入还原铁粉(81mg)和氯化铵(32mg),80摄氏度加热回流反应过夜。冷却反应至室温,过滤去除不溶性固体,滤液蒸干,所得油状物加入5mL DCM溶解,加入饱和碳酸氢钠水溶液5mL,冰浴冷却。向以上两相体系滴加1M的丙烯酰氯(0.45mL),冰浴下反应30分钟。分液,有机相蒸干,制备级薄层色谱纯化(DCM/MeOH,V/V,30/1),得到产物50mg。1H NMR(400MHz,CDCl3)(δ/ppm)9.83–9.34(m,2H),8.46(t,J=7.3Hz,1H),8.00–7.38(m,3H),7.18–7.00(m,1H),6.62(d,J=3.7Hz,1H),6.51–6.20(m,2H),5.69(dd,J=10.0,1.7Hz,1H),4.72(tt,J=16.1,7.9Hz,1H),4.25(s,3H),4.21–4.06(m,2H),3.87(s,3H),2.67(m,2H),2.40(s,6H),1.54(d,6H)。
实施例8 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-溴-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000122
步骤1化合物6-(5-溴-2-氯嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑的合成。
取化合物5-溴-2,4-二氯嘧啶(200mg,0.88mmol)、化合物4-氟-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(279mg,0.88mmol)、Pd(PPh3)2Cl2(37mg,0.053mmol)置于两口烧瓶中,氮气置换三次,在氮气保护下加入DMF(5mL)和2M Na2CO3(1.8mL,3.51mmol),60摄氏度搅拌反应3小时。关闭加热,反应液冷却至室温,倒入50mL水中,乙酸乙酯(5mL*2)萃取,合并有机相,无水硫酸钠干燥,减压蒸干溶剂得粗品(185mg)。
步骤2化合物5-溴-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺的合成。
将化合物6-(5-溴-2-氯嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑(185mg),4-氟-2-甲氧基-5-硝基苯胺(408mg,0.58mmol)、pTSA(332mg,1.93mmol)于氮气保护下加入到单口反应瓶中,之后加入无水二氧六环(2mL),氮气置换三次,100摄氏度加热搅拌反应24小时。减压蒸干溶剂,制备级薄层色谱纯化(PE/EA,V/V,1/1),得灰色固体(166mg)。
步骤3化合物N1-(5-溴-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-N4-(2-(二甲基氨基)乙基)-2-甲氧基-N4–甲基-5-硝基苯-1,4-二胺的合成。
将化合物5-溴-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺(166mg,0.31mmol)加入乙腈(5mL)分散均匀,先后加入三甲基乙二胺(39mg,0.38mmol)、碳酸钾(86mg,0.62mmol),90摄氏度加热回流反应2小时,将反应液冷却至室温,过滤去除不溶性固体,滤液蒸干得到红色油状物153mg。
步骤4化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-溴-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
将化合物N1-(5-溴-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-N4-(2-(二甲基氨基)乙基)-2-甲氧基-N4–甲基-5-硝基苯-1,4-二胺(153mg)溶于乙醇- 水(4mL+1mL),加入还原铁粉(106mg)和氯化铵(34mg),90摄氏度加热回流反应5小时。冷却反应至室温,过滤去除不溶性固体,滤液蒸干,所得油状物加入10mL DCM溶解,加入饱和碳酸氢钠水溶液5mL,冰浴冷却。向以上两相体系滴加0.15M的丙烯酰氯(2.5mL),冰浴下反应30分钟。分液,有机相蒸干,制备级薄层色谱纯化(DCM/MeOH,V/V,20/1),得白色固体(54mg)。1H NMR(400MHz,CDCl3)(δ/ppm)10.06(s,1H),9.57(s,1H),8.65(s,1H),7.86(d,J=1.4Hz,1H),7.67(s,1H),6.78(s,1H),6.46(dd,J=17.0,1.7Hz,1H),6.29(dd,J=16.9,10.2Hz,1H),5.68(dd,J=10.1,1.7Hz,1H),4.73(dq,J=13.9,6.9Hz,1H),3.86(s,2H),2.86(t,J=5.6Hz,2H),2.69(s,3H),2.66(s,3H),2.28(s,2H),2.25(s,5H),1.66(d,J=6.9Hz,6H)。
实施例9 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-环丙基-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000123
将化合物N1–(5-溴-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-N4-(2-(二甲基氨基)乙基)-2-甲氧基-N4–甲基-5-硝基苯-1,4-二胺(37mg,0.06mmol)、环丙基硼酸(6.4mg,0.075mol)、三环丙基磷(6.7mg,0.024μmol)、Pd(Oac)2(2.8mg,0.013mol),K3PO4(34mg,0.016mol)置于圆底烧瓶,氮气置换三次,氮气保护下加入甲苯(5mL),100摄氏度搅拌2小时。冷却至室温,过滤,取滤液浓缩,柱层析(DCM/MeOH,V/V,20/1)得20mg淡黄色固体。溶于乙醇(10mL)和水(3mL),加入还原铁粉(56mg)和氯化铵固体(34mg),加热回流2小时。加入二氯甲烷(20mL)稀释,过滤,浓缩滤液得18mg棕色油状液体,加入13mL二氯甲烷溶解,3mL饱和碳酸氢钠溶液,冰水浴下滴加0.7L 0.15M的丙烯酰氯的二氯甲烷溶液,冰水浴下搅拌30分钟。分液,水相用二氯甲烷萃取(3mL×3),合并有机相并依次用饱和碳酸氢钠溶液和饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析(DCM/MeOH,V/V,20/1),得16mg白色固体。1H NMR(400MHz,CDCl3)(δ/ppm)8.65(s,1H),8.31(s,1H),7.95(s,1H),7.50(d,J=11.3Hz,1H),6.93(s,1H),6.58(dd,J=16.8,10.5Hz,1H),6.41(d,J=16.2Hz,1H),5.82(d,J=10.9Hz,1H),5.34(t,J=4.7Hz,1H),3.98(s,3H),3.43(d,J=5.9Hz,2H),3.24(t,J=5.6Hz,2H),2.83(s,6H),2.69(s,3H),2.69(s,3H),2.19(t,J=7.5Hz,1H),1.67(d,J =6.9Hz,6H),0.93(dd,J=13.1,6.1Hz,3H),0.66(q,J=5.4Hz,3H)。
实施例10 N-(2-(2-甲氧基-乙基-氧基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000124
步骤1化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺的合成。
将化合物6-(2-氯嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑(740mg)溶于无水10mL二氧六环,先后加入4-氟-2-甲氧基-5-硝基苯胺(452mg,2.43mmol)、pTSA(2.26g,13.16mmol),加热至115摄氏度搅拌反应过夜。蒸干二氧六环,冰浴下加入EA 20mL,1N的氢氧化钠水溶液20mL,搅拌10分钟,抽滤,分液。有机相蒸干柱层析(DCM/MeOH,v/v,80/1),浓缩所得固体经PE-EA(v/v,1/1,20mL)回流打浆1小时纯化得到纯品(200mg)。
步骤2化合物N-(2-(2-甲氧基-乙基-氧基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
取乙二醇单甲醚(167mg,2.20mmol)溶于THF(4mL),分批加入NaH(60%,97mg,2.42mmol),室温搅拌30分钟。缓慢加入化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺(200mg,0.44mmol)的无水二氧六环(4mL)溶液,室温搅拌过夜。浓缩反应液,加入10mLEA和10mL饱和食盐水分液,水相用乙酸乙酯萃取(10mL×3),合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得166mg棕色油状液体,用乙醇(10mL)溶解,加入水(2mL),还原铁粉(123mg,2.20mmol),NH4Cl(47mg,0.88mmol),加热回流2小时。冷却至室温,过滤,滤液浓缩,用二氯甲烷(10mL)溶解,加入饱和碳酸氢钠(10mL),冰水浴下滴加1.0M丙烯酰氯的二氯甲烷溶液(0.53mL),搅拌30分钟。分液,水相用二氯甲烷萃取(3mL×3),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(DCM/MeOH,V/V,20/1),得到白色固体粉末(51mg)。1H NMR(400MHz,CDCl3)(δ/ppm)9.57(s,1H),8.49(d,J=5.2Hz,1H),8.32(s,1H),8.01(d,J=1.3Hz,1H),7.77(d,J=11.5Hz,1H),7.54(s,1H),7.11(d,J=5.3Hz,1H),6.64(s,1H),6.43(dd,J=16.9,1.5Hz,1H),6.27(dd,J=16.9,10.2Hz,1H),5.75–5.66(m,1H),4.74(p,J=7.0Hz,1H),4.21–4.10 (m,2H),3.87(s,3H),3.73–3.63(m,2H),3.46(s,3H),2.66(s,3H),1.65(d,J=7.0Hz,6H)。
实施例11 N-(2-(2-(二甲基氨基)乙基)(硫基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000125
取2-二甲基氨基乙硫醇盐酸盐(234mg,1.65mmol)溶于DMF(3mL),分批加入NaH(60%,80mg,3.30mmol),室温搅拌30分钟。缓慢加入化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺(150mg,0.33mmol)的DMF(3mL)溶液,室温搅拌过夜。浓缩反应液,加入10mL EA和10mL饱和食盐水分液,水相用乙酸乙酯萃取(10mL×3),合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩得173mg棕色油状液体,用乙醇(10mL)溶解,加入水(2mL),还原铁粉(123mg,2.20mmol),NH4Cl(47mg,0.88mmol),加热回流2小时。冷却至室温,过滤,滤液浓缩,用二氯甲烷(10mL)溶解,加入饱和碳酸氢钠(10mL),冰水浴下滴加1.0M丙烯酰氯的二氯甲烷溶液(0.53mL),搅拌30分钟。分液,水相用二氯甲烷萃取(3mL×3),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(DCM/MeOH,V/V,20/1),得到白色固体粉末46mg。1H NMR(400MHz,CDCl3)(δ/ppm)9.64(s,1H),8.54(d,J=5.2Hz,1H),7.98(d,J=1.3Hz,1H),7.85(d,J=18.7Hz,2H),7.17(d,J=5.3Hz,1H),7.10(s,1H),6.48(dd,J=17.0,1.5Hz,1H),6.31(dd,J=17.0,10.2Hz,1H),5.73(d,J=10.2Hz,1H),4.76(p,J=7.0Hz,1H),3.92(s,3H),2.87(t,J=6.3Hz,2H),2.67(s,3H),2.38(m,2H),2.28(s,6H),1.67(d,J=7.0Hz,6H)。
实施例12 N-(2-(2-(二甲基氨基)乙基)(氧基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000126
取2-二甲基氨基乙醇(49mg,0.55mmol)溶于二氧六环(1mL),分批加入NaH(60%,24mg,0.61mmol),室温搅拌30分钟。缓慢加入化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺(50mg,0.11mmol)的二氧六环(1mL)溶液,室温搅拌过夜。浓缩反应液,加入3mLEA和3mL饱和食盐水分液,水相用乙酸乙酯萃取(3mL×3),合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得43mg棕色油状液体,用乙醇(3mL)溶解,加入水(1mL),还原铁粉(41mg,0.73mmol),NH4Cl(16mg,0.30mmol),加热回流2小时。冷却至室温,过滤,滤液浓缩,用二氯甲烷(3mL)溶解,加入饱和碳酸氢钠(10mL),冰水浴下滴加1.0M丙烯酰氯的二氯甲烷溶液(0.18mL),搅拌30分钟。分液,水相用二氯甲烷萃取(3mL×3),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(DCM/MeOH,V/V,20/1),得白色固体粉末(20mg)。1H NMR(400MHz,CDCl3)(δ/ppm)9.66(s,1H),9.48(s,1H),8.49(d,J=5.3Hz,1H),8.02(d,J=1.3Hz,1H),7.76(d,J=11.5Hz,1H),7.54(s,1H),7.10(d,J=5.3Hz,1H),6.62(s,1H),6.42(d,J=2.4Hz,1H),5.68(d,J=11.5Hz,1H),4.74(p,J=6.8Hz,1H),4.13(t,J=5.1Hz,2H),3.87(s,3H),2.69(m,2H),2.66(s,3H),2.45–2.39(m,6H),1.66(d,J=7.0Hz,6H)。
实施例13 N-(2-(4-吗啉哌啶)-1-基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000127
取化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺(150mg,0.33mmol),4-(4-哌啶基)吗啉(67.4mg,0.40mmol)溶于乙腈(10mL),加入K2CO3(183mg,1.32mmol),加热回流过夜。浓缩反应液,加入10mL EA和 10mL饱和食盐水分液,水相用乙酸乙酯萃取(10mL×3),合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得180mg棕色油状液体,用乙醇(10mL)溶解,加入水(2mL),还原铁粉(123mg,2.20mmol),NH4Cl(47mg,0.88mmol),加热回流2小时。冷却至室温,过滤,滤液浓缩,用二氯甲烷(10mL)溶解,加入饱和碳酸氢钠(10mL),冰水浴下滴加1.0M丙烯酰氯的二氯甲烷溶液(0.53mL),冰水浴下搅拌30分钟。分液,水相用二氯甲烷萃取(3mL×3),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(DCM/MeOH,V/V,20/1),得白色固体粉末(90mg)。1H NMR(400MHz,CDCl3)(δ/ppm)9.61(s,1H),8.51(d,J=5.0Hz,2H),8.01(d,J=1.3Hz,1H),7.82(d,J=11.5Hz,1H),7.66(s,1H),7.14(d,J=5.2Hz,1H),6.75(s,1H),6.42(dd,J=16.9,1.4Hz,1H),6.28(dd,J=16.9,10.2Hz,1H),5.74(d,J=10.1Hz,1H),4.77(p,J=7.0Hz,1H),3.89(s,3H),3.81(s,4H),3.07(d,J=11.6Hz,3H),2.74(d,J=11.9Hz,3H),2.68(s,3H),2.38(m,1H),2.09(d,J=12.1Hz,2H),1.99–1.69(m,6H),1.66(d,J=7.0Hz,6H)。
实施例14 N-(2-(4-乙氧基哌啶)-1-基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000128
步骤1化合物4-乙氧基哌啶盐酸盐的合成。
取化合物4-羟基哌啶-1-羧酸叔丁酯(2.01g,10.0mmol)溶于THF(10mL),冰水浴冷却下分批加入NaH(60%,288mg,12.0mmol),冰水浴下搅拌30分钟。一次性加入碘乙烷(0.85mL,10.5mmol),室温搅拌过夜。浓缩反应液,加入10mL EA和10mL饱和食盐水分液,水相用乙酸乙酯萃取(10mL×3),合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得无色油状液体(2.20g)。以上所得油状液体(2.20g)溶于DCM(10mL),冰水浴下加入4M HCl的二氧六环溶液(10mL),室温搅拌5小时。浓缩反应液得白色固体(1.33g)。
步骤2化合物N-(2-(4-乙氧基哌啶)-1-基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
取化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基 苯基)嘧啶-2-胺(150mg,0.33mmol),化合物4-乙氧基哌啶盐酸盐(82mg,0.49mmol)溶于乙腈(10mL),加入K2CO3(274mg,1.98mmol),加热回流过夜。浓缩反应液,加入10mLEA和10mL饱和食盐水分液,水相用乙酸乙酯萃取(10mL×3),合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得156mg棕色油状液体,用乙醇(10mL)溶解,加入水(2mL),还原铁粉(123mg,2.20mmol),NH4Cl(47mg,0.88mmol),加热回流2小时。冷却至室温,过滤,滤液浓缩,用二氯甲烷(10mL)溶解,加入饱和碳酸氢钠(10mL),冰水浴下滴加1.0M丙烯酰氯的二氯甲烷溶液(0.53mL),冰水浴下搅拌30分钟。分液,水相用二氯甲烷萃取(3mL×3),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(DCM/MeOH,V/V,20/1)得白色固体粉末(74mg)。1H NMR(400MHz,CDCl3)(δ/ppm)9.61(s,1H),8.52(d,J=5.2Hz,1H),8.02(d,J=1.3Hz,1H),7.67(d,J=8.3Hz,1H),7.13(d,J=5.3Hz,1H),6.77(s,1H),6.40(dd,J=16.9,1.4Hz,1H),6.26(dd,J=17.0,10.1Hz,1H),5.73(d,J=10.1Hz,1H),4.78(p,J=7.1Hz,1H),3.90(s,3H),3.58(q,J=7.0Hz,2H),3.53–3.41(m,1H),3.10–2.95(m,2H),2.78–2.69(m,2H),2.68(s,3H),2.09(d,J=19.3Hz,2H),1.84–1.69(m,2H),1.67(d,J=6.9Hz,6H),1.29–1.23(m,4H)。
实施例15 N-(2-(4-甲氧基哌啶)-1-基)-5-(4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺制备
Figure PCTCN2017094102-appb-000129
步骤1化合物4-甲氧基哌啶盐酸盐的合成。
取化合物4-羟基哌啶-1-羧酸叔丁酯(2.01g,10.0mmol)溶于THF(10mL),冰水浴冷却下分批加入NaH(60%,288mg,12.0mmol),冰水浴下搅拌30分钟。一次性加入碘甲烷(0.65mL,10.5mmol),室温搅拌过夜。浓缩反应液,加入10mL EA和10mL饱和食盐水分液,水相用乙酸乙酯萃取(10mL×3),合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得无色油状液体(2.12g)。以上所得油状液体(2.12g)溶于DCM(10mL),冰水浴下加入4M HCl的二氧六环溶液(10mL),室温搅拌5小时。浓缩反应液得白色固体(1.42g)。
步骤2化合物N-(2-(4-甲氧基哌啶)-1-基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
取化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺(150mg,0.33mmol),化合物4-甲氧基哌啶盐酸盐(78mg,0.49mmol)溶于乙腈(10mL),加入K2CO3(274mg,1.98mmol),加热回流过夜。浓缩反应液,加入10mLEA和10mL饱和食盐水分液,水相用乙酸乙酯萃取(10mL×3),合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得167mg棕色油状液体,用乙醇(10mL)溶解,加入水(2mL),还原铁粉(123mg,2.20mmol),NH4Cl(47mg,0.88mmol),加热回流2小时。冷却至室温,过滤,滤液浓缩,用二氯甲烷(10mL)溶解,加入饱和碳酸氢钠(10mL),冰水浴下滴加1.0M丙烯酰氯的二氯甲烷溶液(0.53mL),冰水浴下搅拌30分钟。分液,水相用二氯甲烷萃取(3mL×3),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(DCM/MeOH,V/V,20/1),得白色固体粉末(90mg)。1H NMR(400MHz,CDCl3)(δ/ppm)9.61(s,1H),8.60(s,1H),8.51(d,J=5.2Hz,1H),8.02(d,J=1.2Hz,1H),7.81(d,J=11.5Hz,1H),7.66(s,1H),7.13(d,J=5.3Hz,1H),6.77(s,1H),6.40(dd,J=16.8,1.3Hz,1H),6.27(dd,J=17.0,10.1Hz,1H),5.73(d,J=10.1Hz,1H),4.78(p,J=7.1Hz,1H),3.89(s,3H),3.41(d,J=1.0Hz,4H),3.08–2.97(m,2H),2.73(t,J=10.5Hz,1H),2.68(s,3H),2.05(s,2H),1.84–1.69(m,2H),1.66(d,J=7.0Hz,6H)。
实施例16 N-(2-吗啉)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000130
取化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺(150mg,0.33mmol),吗啉(67mg,0.40mmol)溶于乙腈(10mL),加入K2CO3(183mg,1.32mmol),加热回流过夜。浓缩反应液,加入10mLEA和10mL饱和食盐水分液,水相用乙酸乙酯萃取(10mL×3),合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得167mg棕色油状液体,用乙醇(10mL)溶解,加入水(2mL),还原铁粉(123mg,2.20mmol),NH4Cl(47mg,0.88mmol),加热回流2小时。冷却至室温,过滤,滤液浓缩,用二氯甲烷(10mL)溶解,加入饱和碳酸氢钠(10mL),冰水浴下 滴加1.0M丙烯酰氯的二氯甲烷溶液(0.53mL),冰水浴下搅拌30分钟。分液,水相用二氯甲烷萃取(3mL×3),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(DCM/MeOH,V/V,20/1),得白色固体粉末(90mg)。1H NMR(400MHz,CDCl3)(δ/ppm)9.65(s,1H),8.57(s,1H),8.51(d,J=5.2Hz,1H),7.99(d,J=1.4Hz,1H),7.83(d,J=11.7Hz,1H),7.68(s,1H),7.14(d,J=5.2Hz,1H),6.77(s,1H),6.48–6.38(m,1H),6.27(dd,J=17.0,10.1Hz,1H),5.75(d,J=10.2Hz,1H),4.76(p,J=6.9Hz,1H),3.91(s,3H),3.87(t,J=4.6Hz,4H),2.88(t,J=4.6Hz,4H),2.67(s,3H),1.66(d,J=7.0Hz,6H)。
实施例17 N-(2-(4-羟基哌啶-1-基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000131
取化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(4-氟-2-甲氧基-5-硝基苯基)嘧啶-2-胺(150mg,0.330mmol),4-羟基哌啶(50mg,0.495mmol)溶于乙腈(10mL),加入K2CO3(183mg,1.32mmol),加热回流过夜。浓缩反应液,加入10mL EA和10mL饱和食盐水分液,水相用乙酸乙酯萃取(10mL×3),合并有机相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得167mg棕色油状液体,用乙醇(10mL)溶解,加入水(2mL),还原铁粉(123mg,2.20mmol),NH4Cl(47mg,0.880mmol),加热回流2h。冷却至室温,过滤,滤液浓缩,用二氯甲烷(10mL)溶解,加入饱和碳酸氢钠(10mL),冰水浴下滴加1.0M丙烯酰氯的二氯甲烷溶液(0.53mL),冰水浴下搅拌30min。分液,水相用二氯甲烷萃取(3mL×3),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(DCM/MeOH,V/V,20/1),得白色固体粉末(33mg).1H NMR(400MHz,氯仿-d)δ9.63(s,1H),8.60(s,1H),8.53(d,J=5.2Hz,1H),8.03(d,J=1.3Hz,1H),7.84(d,J=11.5Hz,1H),7.69(s,1H),7.15(d,J=5.3Hz,1H),6.79(s,1H),6.45(d,J=1.3Hz,0H),6.41(d,J=1.4Hz,1H),6.29(dd,J=16.9,10.1Hz,1H),5.76(d,J=10.1Hz,1H),4.79(p,J=7.0Hz,1H),3.91(s,3H),3.09–3.01(m,2H),2.77(ddd,J=12.1,9.3,2.9Hz,2H),2.69(s,3H),2.08(d,J=11.6Hz,2H),1.84–1.73(m,2H),1.68(d,J=7.0Hz,6H)
实施例18 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-氰基-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000132
步骤1化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-2-(甲硫基)嘧啶-5-甲腈的合成。
取化合物4-氯-2-(甲硫基)嘧啶-5-甲腈(200mg,1.08mmol)、化合物4-氟-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(343mg,1.08mmol)、Pd(PPh3)2Cl2(45mg,0.06mmol)置于两口烧瓶中,氮气置换三次,在氮气保护下加入DMF(5mL)和2M Na2CO3(2mL),60摄氏度搅拌反应2小时。关闭加热,反应液冷却至室温,倒入50mL水中,乙酸乙酯(5mL*2)萃取,合并有机相,无水硫酸钠干燥,减压蒸干溶剂,制备级薄层色谱纯化(P/E,V/V,1.5/1)得白色固体(227mg,0.67mmol)。
步骤2化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-2-(甲基磺酰基)嘧啶-5-甲腈的合成。
将化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-2-(甲硫基)嘧啶-5-甲腈(461mg,1.35mmol)溶于30mL二氯甲烷,冰水浴冷却下分批加入m-CPBA(582mg,3.38mmol),室温搅拌1h。减压蒸干溶剂,柱层析(P/E,V/V,1/1)得淡黄色油状液体(504mg,1.35mmol)。
步骤3化合物2–((4–((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基-5-硝基苯基)氨基)-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-5-甲腈合成。
将化合物N1-(2-(二甲基氨基)乙基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺(400mg,1.35mmol)溶于10mL DMF,冰水浴下分批加入60%NaH(65mg,1.62mmol),搅拌10min。滴加化合物4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-2-(甲基磺酰基)嘧啶-5-甲腈(504mg,1.35mmol)的THF(20mL)溶液,室温搅拌1h.。加入2mL甲醇,浓缩反应液,所得固体加入15mL水和15mL EA,分液,水相用乙酸乙酯(15mL×2)萃取, 合并EA并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得到化合物(220mg,0.392mmol)
步骤4化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-氰基-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
取化合物2–((4–((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基-5-硝基苯基)氨基)-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-5-甲腈(220mg,0.392mmol)用乙醇(10mL)溶解,加入水(2mL),还原铁粉(109mg,1.96mmol),NH4Cl(42mg,0.783mmol),加热回流2h。冷却至室温,过滤,滤液浓缩,用二氯甲烷(10mL)溶解,加入饱和碳酸氢钠(10mL),冰水浴下滴加1.0M丙烯酰氯的二氯甲烷溶液(0.59mL),冰水浴下搅拌30min。分液,水相用二氯甲烷萃取(3mL×3),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(DCM/MeOH,V/V,20/1),得白色固体粉末(76mg)。
实施例19 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-氟-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000133
氮气保护下,将Pd2(dba)3(60mg)和Xantphos(80mg)加入到6-(2-氯-5-氟嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑(281mg,0.87mmol),N-(5-氨基-2–((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺(170mg,0.58mmol)和碳酸钾(200mg,1.46mmol)的2-甲基-2-丁醇(10mL)中,反应液氮气保护下100℃下反应3hrs,冷却至室温,硅藻土过滤,减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1),得到130mg黄色固体,收率为:38.8%。LC-MS(APCI):m/z=579.3(M+1)+,纯度为92.77%(HPLC),1H NMR(400MHz,DMSO):δ9.85(s,1H),8.71(s,1H),8.58(d,J=3.9Hz,1H),8.39(s,1H),8.11(s,1H),7.74(d,J=11.8Hz,1H),6.99(s,1H),6.25(d,J=17.0Hz,1H),5.79–5.65(m,1H),4.87–4.75(m,1H),3.85(s,3H),3.25–3.04(m,2H),2.70–2.58(m,J=4.0Hz,8H),2.50(s,6H),1.56(d,J=6.9Hz,6H).
实施例20 N-(5-((4-(1-环丙基-4-氟-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨 基)-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000134
步骤1化合物N1-(4-(1-环丙基-4-氟-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-N4-(2-(二甲基氨基)乙基)-2-甲氧基-N4-甲基-5-硝基苯-1,4-二胺的合成。
氮气保护下,将Pd(Oac)2(26mg,0.11mmol)和Xantphos(135mg,0.23mmol)加入到中间体化合物6-(2-氯嘧啶-4-基)-1-环丙基-4-氟-2-甲基-1H-苯并[d]咪唑(350mg,1.16mmol),N1-(2-(二甲基氨基)乙基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺(311mg,1.16mmol)和碳酸铯(760mg,2.32mmol)的无水DMF(20mL)中,反应液氮气保护下100℃下反应过夜,冷却至室温,加水淬灭反应,乙酸乙酯(50mLx3)萃取,合并有机相,饱和食盐水(50mL)洗涤无水硫酸钠干燥,除去溶剂,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1),得到360mg黄色固体。LC-MS(APCI):m/z=535.2(M+1)。
步骤2化合物N4-(4-(1-环丙基-4-氟-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-N1-(2-(二甲基氨基)乙基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成
将10%的Pd/C(50mg)加入到化合物N1-(4-(1-环丙基-4-氟-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-N4-(2-(二甲基氨基)乙基)-2-甲氧基-N4-甲基-5-硝基苯-1,4-二胺(360mg)的甲醇溶液中,氢气(气球)下室温反应过夜,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=8:1),得到270mg黄色固体。两步收率46.7%,LC-MS(APCI):m/z=505.2(M+1)。
步骤3化合物N-(5-((4-(1-环丙基-4-氟-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺的合成
氮气保护下,将化合物N4-(4-(1-环丙基-4-氟-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-N1-(2-(二甲基氨基)乙基)-5-甲氧基-N1-甲基苯-1,2,4-三胺(250mg,0.47mmol)溶于30mL无水二氯甲烷中,逐滴滴加DIPEA(0.16mg,0.94mmol),滴加完成后将反应体系冷却至-20℃,在该温度下缓慢加入丙烯酰氯(0.76mL,0.47mmol,0.618mmol/mL),搅拌30分钟,加水,分离有机相并依次用水和饱和碳酸氢钠溶液洗涤,收集有机相减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1),得到120mg黄色固体,收率为45.7%。HPLC:94.72%。LC-MS(APCI):m/z=558.2(M+1),1H NMR(500MHz,DMSO-d6):δ10.03(s,1H),8.98(s,1H),8.48(d,J=5.2Hz,1H),8.20–8.09(m,2H),7.90(d,J=12.0Hz,1H),7.48(d,J=5.2Hz,1H),7.01(s,1H),6.59–6.38(m,1H),6.22(dd,J =16.9,1.0Hz,1H),5.72(d,J=11.0Hz,1H),3.85(s,3H),3.36–3.34(m,1H),3.04–2.85(m,2H),2.67(s,3H),2.61(s,3H),2.49–2.48(m,2H).2.30(s,6H),1.20–1.15(m,J=5.9Hz,2H),1.07–1.01(m,2H).
实施例21 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000135
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到350mg黄色固体。收率为70.0%。LC-MS(APCI):m/z=555.3(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成
合成步骤与实施例20的步骤2相同,得到210mg黄色固体。LC-MS(APCI):m/z=525.3(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-2-(氟甲基)-1-异丙基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成
合成步骤与实施例20的步骤3相同,得到黄色固体产物120mg,收率为50.1%。HPLC:98.25%。LC-MS(APCI):m/z=579.3(M+1),1H NMR(400MHz,DMSO-d6):δ10.56(s,1H),9.88(s,1H),8.84(s,1H),8.52(d,J=5.2Hz,1H),8.33(s,1H),8.26(s,1H),8.01(d,J=12.1Hz,1H),7.62(d,J=5.3Hz,1H),6.98(s,1H),6.25(dd,J=17.0,2.0Hz,1H),5.84(s,1H),5.74–5.68(m,2H),4.97(dt,J=13.7,6.7Hz,1H),3.87(s,3H),3.22–3.12(m,2H),2.72–2.51(m,9H),2.04–2.01(m,2H),1.62(d,J=6.9Hz,6H).
实施例22 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-甲基-1H-咪唑并[4,5-b]吡啶-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000136
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-甲基-1H-咪唑并[4,5-b]吡啶-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到270mg棕黄色油状物。收率为52.1%。LC-MS(APCI):m/z=520.1(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-甲基-1H-咪唑并[4,5-b]吡啶-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到90mg黄色固体。LC-MS(APCI):m/z=490.2(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-甲基-1H-咪唑并[4,5-b]吡啶-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到黄色固体产物15mg。HPLC:97.19%。LC-MS(APCI):m/z=543.5(M+1),1H NMR(500MHz,DMSO-d6):δ9.98(s,1H),9.11(d,J=1.8Hz,1H),8.78(s,1H),8.63(d,J=1.8Hz,1H),8.48(d,J=5.2Hz,1H),8.29(s,1H),7.56(d,J=5.3Hz,1H),6.99(s,1H),6.78–6.45(m,1H),6.20(dd,J=17.0,1.8Hz,1H),5.71(d,J=11.7Hz,1H),4.86–4.72(m,1H),3.83(s,3H),3.07–2.91(m,2H),2.66(s,3H),2.63(s,3H),2.49–2.47(m,2H),2.36(s,6H),1.54(d,J=6.9Hz,6H).
实施例23 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-甲氧基-1H-咪唑并[4,5-b]吡啶-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000137
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-甲氧基-1H-咪唑并[4,5-b]吡啶-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到100mg棕黄色油状物。LC-MS(APCI):m/z=536.2(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-甲氧基-1H-咪唑并[4,5-b]吡啶-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成
合成步骤与实施例20的步骤2相同,得到35mg黄色固体,LC-MS(APCI):m/z=506.3(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-甲氧基-1H-咪唑并[4,5-b]吡啶-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成
合成步骤与实施例20的步骤3相同,得到8mg固体,收率为:20.4%。收率:93.68%。LC-MS(APCI):m/z=560.2(M+1)。
实施例24 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-甲氧基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000138
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-甲氧基-1H-苯并[4,5-b]咪唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到棕黄色油状物。LC-MS(APCI):m/z=535.1(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-甲氧基-1H-苯并[4,5-b]咪唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成
合成步骤与实施例20的步骤2相同,得到300mg棕黄色固体,两步收率为61.9%。LC-MS(APCI):m/z=505.1(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-甲氧基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成
合成步骤与实施例20的步骤3相同,得到200mg固体,收率为:59.6%。HPLC:94.26%。LC-MS(APCI):m/z=559.1(M+1),1H NMR(500MHz,DMSO-d6):δ10.07(s,1H),8.94(s,1H),8.43(d,J=5.3Hz,1H),8.11(s,1H),8.05(d,J=8.5Hz,1H),7.98(s,1H),7.44(d,J=5.3Hz,1H),7.21(d,J=8.3Hz,1H),7.01(s,1H),6.47–6.35(m,1H),6.24(dd,J=16.9,1.9Hz,1H),5.77–5.70(m,1H),4.69–4.58(m,1H),3.83(s,3H),3.34(s,3H),2.92–2.83(m,2H),2.69(s,3H),2.41–2.28(m,2H),2.22(s,6H),1.44(d,J=6.9Hz,6H).
实施例25 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基-5-((4-(3-甲基-2-氧代-2,3-二氢苯并[d]噁唑-5-基)嘧啶-2-基)氨基)苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000139
步骤1化合物5-(2-((4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基-5-硝基苯)氨基)嘧啶-4-基)-3-甲基苯并[d]噁唑-2(3H)-酮的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12:1),得到棕黄色油状物。LC-MS(APCI):m/z=494.0(M+1)。
步骤2化合物5-(2-((5-氨基-4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基苯基)氨基)嘧啶-4-基)-3-甲基苯并[d]噁唑-2(3H)-酮的合成
合成步骤与实施例20的步骤2相同,得到110mg棕黄色固体,两步收率为20%。LC-MS(APCI):m/z=461.1(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基-5-((4-(3-甲基-2-氧代-2,3-二氢苯并[d]噁唑-5-基)嘧啶-2-基)氨基)苯基)丙烯酰胺的合成
合成步骤与实施例20的步骤3相同,得到60mg,收率为:50.4%。HPLC:97.83%。LC-MS(APCI):m/z=518.1(M+1),1H NMR(400MHz,DMSO-d6):δ10.06(s,1H),9.11(s,1H),8.52(d,J=5.2Hz,1H),8.13–8.07(m,3H),7.47(d,J=5.3Hz,1H),7.42(d,J=8.1Hz,1H),7.02(s,1H),6.63–6.48(m,1H),6.30–6.18(m,1H),5.79–5.75(m,J=10.4Hz,1H),3.88(s,3H),3.39(s,3H),2.98(s,2H),2.68(s,3H),2.59–2.51(m,2H),2.34(s,6H).
实施例26 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-1H-吲唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000140
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(4-氟-1-异丙基-1H-吲唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12:1),得到棕黄色固体。LC-MS(APCI):m/z=523.1(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(4-氟-1-异丙基-1H-吲唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成
合成步骤与实施例20的步骤2相同,得到360mg橘黄色固体,两步收率为47.4%。LC-MS(APCI):m/z=493.2(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-1H-吲唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成.
合成步骤与实施例20的步骤3相同,得到220mg黄色固体,收率为:55.1%。HPLC:97.27%。LC-MS(APCI):m/z=547.2(M+1),1H NMR(400MHz,DMSO-d6):δ10.13(s,1H),9.11(s,1H),8.56(d,J=5.2Hz,1H),8.42(s,1H),8.24(s,1H),8.20(s,1H),7.84(d,J=11.7Hz,1H),7.60(d,J=5.2Hz,1H),7.03(s,1H),6.50–6.38(m,1H),6.27(dd,J=16.8,1.7Hz,1H),5.76(dd,J=10.1,1.6Hz,1H),5.17(dt,J=13.3,6.6Hz,1H),3.86(s,3H),2.95–2.86(m,2H),2.70(s,3H),2.41–2.31(m,2H),2.24(s,6H),1.49(d,J=6.5Hz,6H).
实施例27 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-1H-吲唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000141
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-1H-吲唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12:1),得到棕黄色固体360mg。LC-MS(APCI):m/z=505.1(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(4-氟-1-异丙基-1H-吲唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到258mg黄色固体,两步收率为51.3%。LC-MS(APCI):m/z=475.2(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-1H-吲唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到135mg黄色固体,收率为:46.2%。HPLC:98.60%。LC-MS(APCI):m/z=543.2(M+1),1H NMR(400MHz,DMSO-d6):δ10.12(s,1H),9.11(s,1H),8.63–8.46(m,2H),8.14(d,J=4.6Hz,2H),8.04(d,J=8.3Hz,1H),7.84(d,J=8.5Hz,1H),7.56(d,J=5.1Hz,1H),7.03(s,1H),6.56–6.39(m,1H), 6.27(d,J=15.9Hz,1H),5.77(d,J=10.0Hz,1H),5.23–5.02(m,1H),3.87(s,3H),2.96–2.86(m,2H),2.70(s,3H),2.45–2.32(m,2H),2.25(s,6H),1.48(d,J=6.5Hz,6H).
实施例28 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(8-氟-3-异丙基-2-甲基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000142
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12:1),得到棕黄色固体。LC-MS(APCI):m/z=535.2(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成
合成步骤与实施例20的步骤2相同,得到336mg橘黄色固体,两步收率为47.3%。LC-MS(APCI):m/z=507.2(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(8-氟-3-异丙基-2-甲基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成
合成步骤与实施例20的步骤3相同,得到120mg黄色固体,收率为:55.1%。HPLC:95.47%。LC-MS(APCI):m/z=561.2(M+1),1H NMR(400MHz,DMSO-d6):δ10.10(s,1H),9.03(d,J=4.0Hz,1H),8.87(s,1H),8.54–8.42(m,1H),8.22(s,1H),7.92(d,J=12.2Hz,1H),7.55(M,1H),7.00(s,1H),6.41(dd,J=15.1,11.2Hz,1H),6.24(d,J=17.1Hz,1H),5.73(d,J=9.7Hz,1H),3.83(s,3H),3.60–3.50(m,1H),2.91–2.80(m,2H),2.68(s,3H),2.40(s,3H),2.37–2.28(m,2H),2.21(s,6H),1.31(d,J=6.4Hz,6H).
实施例29 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(8-氟-2-甲基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000143
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(8-氟-2-甲基-咪唑并[1,2-a]吡啶-6- 基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12:1),得到棕黄色固体。LC-MS(APCI):m/z=495.1(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(8-氟-2-甲基-咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到160mg橘黄色固体,两步收率为30.2%。LC-MS(APCI):m/z=465.2(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(8-氟-2-甲基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到108mg黄色固体,收率为:59.6%。HPLC:97.92%。LC-MS(APCI):m/z=519.2(M+1),1H NMR(400MHz,DMSO-d6):1HNMR(500MHz,DMSO-d6):δ10.24(s,1H),9.62(s,1H),9.32(s,1H),8.52(d,J=5.0Hz,1H),8.00(s,2H),7.89(d,J=12.6Hz,1H),7.42(d,J=4.9Hz,1H),7.04(s,1H),6.49(dd,J=15.9,10.1Hz,1H),6.33(d,J=16.9Hz,1H),5.84(d,J=10.4Hz,1H),3.89(s,3H),2.94-2.85(m,2H),2.70(s,3H),2.37(s,3H),2.35-2.28(m,2H),2.23(s,6H).
实施例30 N-(5-((4-(4-(叔丁氧基)-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000144
步骤1化合物N1-(4-(4-(叔丁氧基)-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-N4-(2-(二甲基氨基)乙基)-2-甲氧基-N4-甲基-5-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到棕黄色固体。LC-MS(APCI):m/z=591.2(M+1)。
步骤2化合物N4-(4-(4-叔丁氧基)-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-N1-(2-(二甲基氨基)乙基)-5-甲氧基-N1-甲基苯1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到220mg淡黄色固体,两步收率为30.2%。LC-MS(APCI):m/z=561.3(M+1)。
步骤3化合物N-(5-((4-(4-(叔丁氧基)-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到54mg黄色固体,收率为:23.4%。 HPLC:96.88%。LC-MS(APCI):m/z=615.4(M+1),1H NMR(400MHz,DMSO-d6):δ9.96(s,1H),8.69(s,1H),8.44(d,J=5.2Hz,1H),8.20(s,1H),8.06(s,1H),7.50(s,1H),7.43(d,J=5.3Hz,1H),7.00(s,1H),6.79–6.48(m,1H),6.20(d,J=15.8Hz,1H),5.72(d,J=11.1Hz,1H),4.87-4.68(m,1H),3.84(s,3H),3.12–2.95(m,2H),2.67(s,3H),2.57(s,3H),2.50-2.49(m,2H),2.41(s,6H),1.55(d,J=6.8Hz,6H),1.37(s,9H).
实施例31 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(7-氟-2-甲基苯并[d]噁唑-5-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000145
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(7-氟-2-甲基苯并[d]噁唑-5-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12:1),得到棕黄色固体。LC-MS(APCI):m/z=496.2(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(7-氟-2-甲基苯并[d]噁唑-5-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成
合成步骤与实施例20的步骤2相同,得到125mg橘黄色固体,两步收率为23.5%。LC-MS(APCI):m/z=466.1(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(7-氟-2-甲基苯并[d]噁唑-5-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成
合成步骤与实施例20的步骤3相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12:1)得到55mg淡黄色固体,收率为41.0%。HPLC:91.53%。LC-MS(APCI):m/z=520.2(M+1),1H NMR(500MHz,DMSO-d6):δ10.05(s,1H),9.12(s,1H),8.51(d,J=5.2Hz,1H),8.38(s,1H),8.29(d,J=11.5Hz,1H),8.15(s,1H),7.54(d,J=5.2Hz,1H),7.00(s,1H),6.52-6.41(m,1H),6.38-6.30(m,1H),5.78-5.71(m,1H),3.85(s,3H),2.99-2.86(m,2H),2.67(s,3H),2.66(s,3H),2.46–2.36(m,2H),2.28(s,6H).
实施例32 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-2-甲基苯并[d]噁唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000146
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(4-氟-2-甲基苯并[d]噁唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12:1),得到棕黄色固体。LC-MS(APCI):m/z=496.2(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(4-氟-2-甲基苯并[d]噁唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到125mg橘黄色固体,两步收率为23.5%。LC-MS(APCI):m/z=466.1(M+1)。
步骤6化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-2-甲基苯并[d]噁唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12:1)得到10mg淡黄色固体,收率为HPLC:98.43%。LC-MS(APCI):m/z=520.2(M+1).
实施例33 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(7-氟苯并[d]噁唑-5-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000147
氮气保护下,将Pd2(dba)3(94mg)和Xantphos(120mg)加入到中间体化合物5-(2-氯嘧啶-4-基)-7-氟-苯并[d]噁唑(190mg,0.76mmol),N-(5-氨基-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺(220mg,0.75mmol)和碳酸钾(270mg,1.90mmol)的无水2-甲基-2-丁醇(20mL)中,反应液氮气保护下100℃下反应3hrs过夜,冷却至室温,硅藻土过滤,减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12:1),得到棕黄色固体40mg。收率为10.4%。HPLC:96.01%。LC-MS(APCI):m/z=506.2(M+1),1H NMR(500MHz,DMSO-d6):δ10.13(s,1H),9.19(s,1H),8.93(s,1H),8.57(s,1H),8.54(d,J=5.1Hz,1H),8.41(d,J=11.6Hz,1H),8.18(s,1H),7.59(d,J=5.1Hz, 1H),7.03(s,1H),6.41(d,J=9.2Hz,1H),6.35(d,J=16.3Hz,1H),5.77(d,J=11.2Hz,1H),3.86(s,3H),2.94-2.84(m,2H),2.70(s,3H),2.40–2.29(m,2H),2.24(s,6H)。
实施例34 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-氟-4-(8-氟-3-异丙基-2-甲基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000148
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(5-氟-4-(8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12:1),得到棕黄色固体。LC-MS(APCI):m/z=553.1(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(5-氟-4-(8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到90mg橘黄色固体,两步收率为47.3%。LC-MS(APCI):m/z=525.2(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-氟-4-(8-氟-3-异丙基-2-甲基咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到50mg黄色固体,收率为:50.8%。LC-MS(APCI):m/z=579.3(M+1),1H NMR(400MHz,DMSO-d6):δ9.77(s,1H),8.78(s,1H),8.74(s,1H),8.60(d,J=3.3Hz,1H),8.36(s,1H),7.76(d,J=12.2Hz,1H),7.20(s,1H),6.94(s,1H),6.27–6.21(m,1H),5.68(d,J=11.6Hz,1H),3.85(s,3H),3.50–3.41(m,2H),3.24–3.23(m,1H),3.13–2.99(m,2H),2.68(s,6H),2.58(s,3H),2.41(s,3H),1.35(d,J=7.1Hz,6H).
实施例35 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000149
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-甲基-1H-苯并[4,5-b]咪 唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到棕黄色油状物。LC-MS(APCI):m/z=519.3(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-甲基-1H-苯并[4,5-b]咪唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成。
将还原铁粉(90mg,1.62mmol)和氯化铵(43mg,0.81mmol)加入到化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-甲基-1H-苯并[4,5-b]咪唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的乙醇和水(8mL/2mL)混合溶液中,反应回流反应2hrs,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=8:1),得到120mg棕黄色固体,两步收率为35.2%。LC-MS(APCI):m/z=489.3(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到50mg,收率为:36.9%。HPLC:95.97%。LC-MS(APCI):m/z=543.7(M+1),1H NMR(500MHz,DMSO-d6):δ10.42(br,1H),9.79(s,1H),8.75(s,1H),8.45(d,J=5.2Hz,1H),8.34(s,1H),8.12(s,1H),8.05(d,J=8.5Hz,1H),7.56(d,J=8.3Hz,1H),7.49(d,J=5.3Hz,1H),6.95(s,1H),6.24(dd,J=17.0,2.0Hz,1H),5.73-5.66(m,1H),4.85-4.72(m,1H),3.87(s,3H),3.25(s,2H),2.75-2.65(m,5H),2.59(s,3H),2.58(s,3H),2.49(s,3H),1.56(d,J=6.9Hz,6H).
实施例36 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000150
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-1H-苯并[4,5-b]咪唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到棕黄色油状物。LC-MS(APCI):m/z=505.2(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-1H-苯并[4,5-b]咪唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成
合成步骤与实施例20的步骤2相同,得到280mg淡绿色固体,两步收率为36.8%。LC-MS(APCI):m/z=475.2(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成
合成步骤与实施例20的步骤3相同,得到50mg,收率为:16.9%。HPLC:96.48%。LC-MS(APCI):m/z=529.2(M+1),1H NMR(500MHz,DMSO-d6):δ10.12(s,1H),9.11(s,1H),8.48(d,J=5.2Hz,1H),8.47-8.43(m,2H),8.14(dd,J=8.5,0.8Hz,1H),8.07(s,1H),7.70(d,J=8.5Hz,1H),7.50(d,J=5.2Hz,1H),7.02(s,1H),6.41(dd,J=16.9,10.1Hz,1H),6.25(dd,J=17.0,1.6Hz,1H),5.82-5.71(m,1H),4.93-4.81(m,1H),3.85(s,3H),2.91-2.82(m,2H),2.69(s,3H),2.32-2.25(m,2H),2.20(s,6H),1.52(d,J=6.7Hz,6H).
实施例37 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-(三氟甲基)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000151
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-(三氟甲基)-1H-苯并[4,5-b]咪唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12:1),得到250mg棕黄色固体。LC-MS(APCI):m/z=573.2(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-(三氟甲基)-1H-苯并[4,5-b]咪唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到58mg黄棕色固体,两步收率为6.34%。LC-MS(APCI):m/z=541.9(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-(三氟甲基)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到40mg粗品,制备TLC进一步纯化得到18mg淡黄色固体,收率为:36.9%。HPLC:96.65%。LC-MS(APCI):m/z=597.2(M+1)+,1H NMR(400MHz,DMSO-d6):δ10.08(s,1H),9.77(s,1H),8.74(s,1H),8.61–8.45(m,2H),8.39–8.21(m,2H),7.89(d,J=8.5Hz,1H),7.61(d,J=5.2Hz,1H),6.97(s,1H),6.25(d,J=17.2Hz,1H),5.73(d,J=8.4Hz,1H),5.00–4.85(m,1H),3.86(s,3H),3.23–3.09(m,2H),2.83–2.50(m,11H),1.67(d,J=6.8Hz,6H).
实施例38 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(7-氟-1H-吲唑-5-基)嘧啶 -2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000152
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(7-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=12:1),得到棕黄色固体。LC-MS(APCI):m/z=565.1(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(7-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成
合成步骤与实施例20的步骤2相同,得到570mg黄棕色固体,两步收率为71.0%。LC-MS(APCI):m/z=535.2(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(7-氟-1H-吲唑-5-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成
合成步骤与实施例20的步骤3相同,得到390mg灰黄色固体N-(2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(7-氟-1-(四氢-2H-吡喃-2-基)-1H-吲唑-5-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺,收率为:61.9%。LC-MS(APCI):m/z=589.2(M+1)。
将三氟乙酸(5mL)加入到上述灰黄色固体(390mg,0.66mmol)的无水二氯甲烷(10mL)中,反应在室温下反应2hrs。减压除去反应液,残渣用饱和碳酸氢钠溶液调pH至碱性,用二氯甲烷(100mLx3)萃取,合并有机层,用饱和食盐水洗涤,无水硫酸钠干燥滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1),得到250mg灰黄色固体,收率为:75.2%。LC-MS(APCI):m/z=505.2(M+1),HPLC:95.59%。1H NMR(500MHz,DMSO-d6):δ13.84(s,1H),10.12(s,1H),9.26(s,1H),8.73(s,1H),8.49(d,J=4.7Hz,1H),8.26(s,1H),8.12(d,J=12.3Hz,1H),8.04(s,1H),7.49(d,J=4.4Hz,1H),7.02(s,1H),6.53–6.40(m,1H),6.32(d,J=17.0Hz,1H),5.79(d,J=10.3Hz,1H),3.87(s,3H),2.94–2.83(m,2H),2.69(s,3H),2.39–2.29(m,2H),2.24(s,6H).
实施例39 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(7-氟-1-甲基-吲唑-5-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000153
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(7-氟-1-甲基-1H-吲唑-5-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=11:1),得到棕黄色固体。LC-MS(APCI):m/z=495.2(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(7-氟-1-甲基-1H-吲唑-5-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到217mg黄棕色固体,两步收率为43.7%。LC-MS(APCI):m/z=465.1(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(7-氟-1-甲基-1H-吲唑-5-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到120mg灰黄色固体,收率为:49.3%。LC-MS(APCI):m/z=519.2(M+1),HLPC:93.48%。1H NMR(300MHz,DMSO-d6):δ10.14(s,1H),9.18(s,1H),8.66(s,1H),8.58(d,J=2.5Hz,1H),8.49(d,J=5.2Hz,1H),8.08(s,1H),7.95(d,J=13.5Hz,1H),7.48(d,J=5.3Hz,1H),7.03(s,1H),6.48(dd,J=16.8,10.1Hz,1H),6.33(dd,J=16.9,1.9Hz,1H),5.79(dd,J=10.0,1.9Hz,1H),4.24(s,3H),3.88(s,3H),2.97–2.85(m,2H),2.70(s,3H),2.42–2.31(m,2H),2.26(s,6H).
实施例40 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(7-氟-2-甲基-2H-吲唑-5-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000154
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(7-氟-2-甲基-2H-吲唑-5-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=11:1),得到棕黄色固体。LC-MS(APCI):m/z=495.2(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(7-氟-2-甲基-2H-吲唑-5-基)嘧啶-2- 基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到120mg黄棕色固体,两步收率为37.5%。LC-MS(APCI):m/z=465.2(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(7-氟-2-甲基-2H-吲唑-5-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到120mg灰黄色固体,收率为:49.3%。LC-MS(APCI):m/z=519.3(M+1),HPLC:97.69%,1H NMR(300MHz,DMSO-d6):δ10.11(s,1H),9.23(s,1H),8.73(s,1H),8.51(d,J=5.2Hz,1H),8.21(d,J=2.2Hz,1H),8.14(d,J=13.6Hz,1H),8.08(s,1H),7.51(d,J=5.3Hz,1H),7.04(s,1H),6.55(dd,J=16.7,10.0Hz,1H),6.34(dd,J=16.9,1.9Hz,1H),5.81(dd,J=10.1,1.8Hz,1H),4.21(s,3H),3.89(s,3H),2.99–2.91(m,2H),2.70(s,3H),2.49–2.41(m,2H),2.31(s,6H).
实施例41 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(7-氟-3-异丙基-2-甲基-2H-吲唑-5-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000155
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(7-氟-2-甲基-3-(丙-1-烯-2-基)-2H-吲唑-5-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=11:1),得到棕黄色固体。LC-MS(APCI):m/z=535.2(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(7-氟-3-异丙基-2-甲基-2H-吲唑-5-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成
合成步骤与实施例20的步骤2相同,得到80mg黄棕色固体,两步收率为25.1%。LC-MS(APCI):m/z=507.2(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(7-氟-3-异丙基-2-甲基-2H-吲唑-5-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成
合成步骤与实施例20的步骤3相同,得到15mg淡黄色固体,收率为:17.0%。LC-MS(APCI):m/z=561.2(M+1),HPLC:98.82%。
实施例42 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(3-异丙基-2-甲基-2H-吲 唑-5-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000156
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(2-甲基-3-(丙-1-烯-2-基)-2H-吲唑-5-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=11:1),得到棕黄色固体。LC-MS(APCI):m/z=517.1(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(3-异丙基-2-甲基-2H-吲唑-5-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到360mg黄棕色固体,两步收率为45.4%。LC-MS(APCI):m/z=489.2(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(-3-异丙基-2-甲基-2H-吲唑-5-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到50mg淡黄色固体,收率为:12.5%。LC-MS(APCI):m/z=543.7(M+1),HPLC:95.93%。1H NMR(500MHz,DMSO-d6):δ10.07(s,1H),8.97(s,1H),8.61(s,1H),8.43(d,J=5.2Hz,1H),8.12(d,J=9.1Hz,1H),8.08(s,1H),7.55(d,J=9.1Hz,1H),7.46(d,J=5.2Hz,1H),7.00(s,1H),6.41(dd,J=16.8,10.1Hz,1H),6.24(d,J=16.6Hz,1H),5.74(d,J=10.2Hz,1H),4.10(s,3H),3.84(s,3H),3.60–3.52(m,1H),2.93–2.84(m,2H),2.69(s,3H),2.38–2.28(m,2H),2.22(s,6H),1.45(d,J=7.0Hz,6H).
实施例43 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(咪唑并[1,2-b]哒嗪-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000157
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(咪唑并[1,2-b]哒嗪-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇 (v/v)=11:1),得到棕黄色油状物。LC-MS(APCI):m/z=464.2(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(咪唑并[1,2-b]哒嗪-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到74mg黄色固体,两步收率为20.9%。LC-MS(APCI):m/z=434.1(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(咪唑并[1,2-b]哒嗪-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到57mg淡黄色固体,收率为:68.9%。LC-MS(APCI):m/z=488.2(M+1),HPLC:97.97%。1H NMR(500MHz,DMSO-d6):δ10.09(s,1H),9.08(s,1H),8.63(d,J=5.0Hz,1H),8.48(d,J=9.3Hz,1H),8.42(s,1H),8.33(s,1H),8.18(d,J=9.6Hz,1H),7.90(s,1H),7.63(d,J=5.0Hz,1H),7.02(s,1H),6.54–6.41(m,1H),6.35(d,J=16.8Hz,1H),5.78(d,J=11.7Hz,1H),3.86(s,3H),2.99–2.84(m,2H),2.69(s,3H),2.48–2.48(m,2H),2.26(s,6H).
实施例44 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-氧代-1,2,5,6-四氢-4H-咪唑并[4,5,1-ij]喹啉-8-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000158
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-氧代-1,2,5,6-四氢-4H-咪唑并[4,5,1-ij]喹啉-8-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=11:1),得到棕黄色固体。LC-MS(APCI):m/z=561.1(M+1)。
步骤2化合物8-(2-((5-氨基-4-((2-(二甲基氨基)乙基)(甲基)氨基)-2-甲氧基苯基)氨基)嘧啶-4-基)-1-异丙基-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉-2(1H)-酮的合成
合成步骤与实施例20的步骤2相同,得到255mg黄棕色固体,两步收率为56.5%。LC-MS(APCI):m/z=530.1(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-氧代-1,2,5,6-四氢-4H-咪唑并[4,5,1-ij]喹啉-8-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成
合成步骤与实施例20的步骤3相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=8:1)得到135mg灰黄色固体,收率为:48.0%。LC-MS(APCI):m/z=585.2(M+1)+, HPLC:99.14%,1H NMR(400MHz,DMSO-d6):δ10.11(s,1H),9.07(s,1H),8.44(d,J=5.3Hz,1H),8.05(s,1H),7.94(s,1H),7.83(s,1H),7.46(d,J=5.3Hz,1H),7.09–7.02(m,1H),6.45(s,1H),6.32–6.19(m,1H),5.80–5.74(m,1H),4.63(dt,J=14.0,6.9Hz,1H),3.88(s,3H),3.77(t,J=5.4Hz,2H),2.94–2.86(m,4H),2.71(s,3H),2.42–2.31(m,2H),2.26(s,6H),2.09–2.00(m,2H),1.48(d,J=6.9Hz,6H).
实施例45 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基-5-((4-(2-甲氧基-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉-8-基)嘧啶-2-基)氨基)苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000159
步骤1化合物N1-(2-(二甲基氨基)乙基)-5-甲氧基-N4-(4-(2-甲氧基-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉-8-基)嘧啶-2-基)-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到棕红色固体。LC-MS(APCI):m/z=533.2(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-5-甲氧基-N4-(4-(2-甲氧基-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉-8-基)嘧啶-2-基)-N1-甲基苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到139mg黄棕色固体,两步收率为27.9%。LC-MS(APCI):m/z=503.2(M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基-5-((4-(2-甲氧基-5,6-二氢-4H-咪唑并[4,5,1-ij]喹啉-8-基)嘧啶-2-基)氨基)苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=9:1)得到70mg黄色固体,收率为:46.7%。LC-MS(APCI):m/z=557.2(M+1)+,HPLC:97.91%,1H NMR(400MHz,DMSO-d6):δ10.06(s,1H),9.13(s,1H),8.41(d,J=5.3Hz,1H),7.93(s,1H),7.87(s,1H),7.83(s,1H),7.38(d,J=5.4Hz,1H),7.00(s,1H),6.48(s,1H),6.21(dd,J=16.9,1.8Hz,1H),5.74(dd,J=10.1,1.6Hz,1H),3.86(s,3H),3.74(t,J=5.7Hz,2H),3.33(s,3H),2.99-2.88(m,2H),2.85(t,J=5.7Hz,2H),2.66(s,3H),2.48-2.47(m,2H),2.28(s,6H),2.07-1.98(m,2H).
实施例46 N-(5-((4-(苯并[d]噻唑-6-基)嘧啶-2-基)氨基)-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000160
步骤1化合物N1-(4-(苯并[d]噻吩-6-基)嘧啶-2-基)-N4-(2-(二甲基氨基)乙基)-2-甲氧基-N4-甲基-5-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到486mg棕红色油状物。LC-MS(APCI):m/z=479.4(M+1)。
步骤2化合物N4-(4-(苯并[d]噻吩-6-基)嘧啶-2-基)-N1-(2-(二甲基氨基)乙基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到184mg黄棕色固体,两步收率为27.0%。LC-MS(APCI):m/z=450.1M+1)。
步骤3化合物N-(5-((4-(苯并[d]噻唑-6-基)嘧啶-2-基)氨基)-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到185mg黄色固体,制备TLC得到50mg淡黄色固体,收率为:24.2%。LC-MS(APCI):m/z=504.1(M+1)+,HPLC:97.92%,1H NMR(400MHz,DMSO-d6):δ10.21(s,1H),9.52(s,1H),9.44(d,J=19.2Hz,2H),8.57(d,J=5.1Hz,1H),8.46–8.33(m,1H),8.17(d,J=8.5Hz,1H),8.05(s,1H),7.58(d,J=5.2Hz,1H),7.06(s,1H),6.56–6.38(m,2H),5.91–5.78(m,1H),3.89(s,3H),2.96–2.82(m,2H),2.70(s,3H),2.35–2.27(m,2H),2.22(s,6H).
实施例47 N-(4-(二氟甲氧基)-2-((2-(二甲基氨基乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑6-基)嘧啶-2-基)氨基)苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000161
步骤1化合物2-(二氟甲氧基)-N4-(2-(二甲基氨基)乙基)-N1-(4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-N4-甲基-5-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到355mg黄色固体。LC-MS(APCI):m/z=573.1(M+1)。
步骤2化合物5-(二氟甲氧基)-N1-(2-(二甲基氨基)乙基)-N4-(4-(4-氟-1-异丙基-2- 甲基-1H-苯并[d]咪唑l-6-基)嘧啶-2-基)-N1-甲基苯-1,2,4-三胺的合成
合成步骤与实施例20的步骤2相同,得到235mg黄棕色固体,两步收率为52.9%。LC-MS(APCI):m/z=543.2M+1)。
步骤3化合物N-(4-(二氟甲氧基)-2-((2-(二甲基氨基乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑6-基)嘧啶-2-基)氨基)苯基)丙烯酰胺的合成
合成步骤与实施例20的步骤3相同,得到80mg黄色固体,收率为:24.2%。LC-MS(APCI):m/z=597.1(M+1),HPLC:95.79%,1H NMR(400MHz,DMSO-d6):δ9.95(s,1H),8.74(s,2H),8.49(d,J=5.3Hz,1H),8.21(s,1H),7.83(d,J=11.9Hz,1H),7.57(d,J=5.3Hz,1H),7.16(s,1H),6.31–6.27(m,1H),6.26–6.22(m,1H),5.88(dd,J=10.3,1.7Hz,1H),5.79–5.76(m,1H),4.88–4.73(m,1H),3.15–3.07(m,2H),2.69–2.52(m,14H),1.58(d,J=6.9Hz,6H).
实施例48 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-乙氧基-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000162
步骤1化合物N1-(2-(二甲基氨基)乙基)-5-乙氧基-N4-(4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到500mg红色固体。LC-MS(APCI):m/z=551.1(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-5-乙氧基-N4-(4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-N1-甲基苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到256mg淡黄色固体,两步收率为46.4%。LC-MS(APCI):m/z=521.3M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-乙氧基-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到150mg黄色固体,收率为:24.2%。LC-MS(APCI):m/z=575.6(M+1),HPLC:97.22%,1H NMR(400MHz,DMSO-d6):δ10.08(s,1H),9.02(s,1H),8.51(s,1H),8.19(d,J=24.3Hz,2H),7.96(d,J=11.8Hz,1H),7.59(s,1H),7.03(s,1H),6.56–6.34(m,1H),6.27(d,J=16.8Hz,1H),5.88–5.63(m, 1H),4.84(s,1H),4.19–4.07(m,2H),2.97–2.86(m,2H),2.70(s,3H),2.63(s,3H),2.45–2.35(m,2H),2.27(s,6H),1.59(d,J=4.8Hz,6H),1.34(t,J=6.0Hz,3H)。
实施例49 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-(2,2,2-三氟乙氧基)苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000163
步骤1化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-N1-甲基-2-硝基-5-(2,2,2-三氟乙氧基)苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到435mg红色固体。LC-MS(APCI):m/z=605.2(M+1)。
步骤2化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(4-氟-1-异丙基-2甲基-1H-苯并[d]嘧啶-6-基)嘧啶-2-基)-N1-甲基-5-(2,2,2-三氟甲氧基)苯-1,2,4-三胺的合成。
合成步骤与实施例20的步骤2相同,得到290mg黄绿色固体,两步收率为33.9%。LC-MS(APCI):m/z=575.6M+1)。
步骤3化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-(2,2,2-三氟乙氧基)苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到140mg黄色固体,收率为:43.7%。LC-MS(APCI):m/z=629.2(M+1),HPLC:94.62%,1H NMR(400MHz,DMSO-d6):δ9.96(s,1H),8.80(s,1H),8.48(d,J=5.2Hz,1H),8.31(s,1H),8.20(s,1H),7.87(d,J=12.2Hz,1H),7.56(d,J=5.3Hz,1H),7.18(s,1H),6.34–6.19(m,1H),5.87(dd,J=10.3,1.7Hz,1H),5.82–5.67(m,1H),4.86–4.75(m,3H),3.10–3.02(m,2H),2.66(s,3H),2.61(s,3H),2.50–2.40(m,8H),1.57(d,J=6.9Hz,6H).
实施例50 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-(甲基-d3)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000164
步骤1化合物6-溴-1-异丙基-2-(甲基-d3)-1H-苯并[d]咪唑的合成。
将5-溴-N1-异丙基苯-1,2-二胺(3.00m,13.10mmol)加入到冰醋酸-d4(15mL)中,反应在回流反应2hrs。冷却至室温,减压除去醋酸,残渣用饱和碳酸氢钠溶液调pH大约7,二氯甲烷(50mlx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=20:1)得到2.68g棕红色油状物,收率为:80.22%,LC-MS(APCI):m/z=256.1(M+1)。
步骤2化合物1-异丙基-2-(甲基-d3)-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑的合成
氮气保护下,将TCHP(500mg)和Pd(Oac)2(250mg)加入到化合物6-溴-1-异丙基-2-(甲基-d3)-1H-苯并[d]咪唑(2.68g,10.47mmol),联硼酸频那醇酯(4.00g,15.70mmol)和乙酸钾(3.00g,31.41mmol)的无水DMSO(25mL)混合物中,氮气保护下,反应在100℃下反应2hrs。冷却至室温,加水淬灭反应,用乙酸乙酯(100mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=2:1),得到2.2g黄色固体,收率为69.3%。LC-MS(APCI):m/z=304.2(M+1)。
步骤3化合物6-(2-氯嘧啶-4-基)-1-异丙基-2-(甲基-d3)-1H-苯并[d]咪唑的合成
氮气保护下,将Pd(PPh3)2Cl2(220mg)加入到1-异丙基-2-(甲基-d3)-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(2.2g,7.26mmol),2,4-二氯嘧啶(1.30g,8.71mmol)和碳酸钠(1.90g,18.15mmol)的乙腈(18mL)和水(6mL)的混合物中,氮气保护下反应升温至80℃反应2.5hrs。冷却至室温,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=1:1),得到2.7g白色固体。收率为100%。LC-MS(APCI):m/z=290.2(M+1)。
步骤4化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-(甲基-d3)-1H-苯并[4,5-b]咪唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到棕黄色油状物。LC-MS(APCI):m/z=522.3(M+1)。
步骤5化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-(甲基-d3)-1H-苯并[4,5-b]咪唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基苯-1,2,4-三胺的合成。
将还原铁粉(1.30g,23.08mmol)和氯化铵(600mg,11.54mmol)加入到化合物化合物N1-(2-(二甲基氨基)乙基)-N4-(4-(1-异丙基-2-(甲基-d3)-1H-苯并[4,5-b]咪唑-6-基)嘧啶-2-基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺的乙醇和水(16mL/4mL)混合溶液中,反应回流反应2hrs,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=8:1),得到1.50g黄色固体,三步收率为42.1%。LC-MS(APCI):m/z=492.2(M+1)。
步骤6化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(1-异丙基-2-(甲基-d3)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到1.13g,收率为:67.7%。HPLC:97.42%。LC-MS(APCI):m/z=546.2(M+1),1H NMR(400MHz,DMSO-d6):δ10.07(s,1H),8.95(s,1H),8.39(d,J=44.1Hz,2H),8.22–7.98(m,2H),7.52(d,J=29.5Hz,2H),7.00(s,1H),6.34(d,J=69.0Hz,2H),5.74(s,1H),4.78(s,1H),3.84(s,3H),2.96–2.81(m,2H),2.69(s,3H),2.40–2.31(m,2H),2.23(s,6H),1.55(s,6H).
实施例51 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基-5-((4-(2-甲基-1-(丙基-2-基-2-d)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000165
步骤1化合物5-溴-N-(4-甲氧基苯基)-2-硝基-N-(异丙基-2-基-2-d)苯胺的合成。
室温下将碳酸钾(248mg,1.80mmol)和N-(4-甲氧基苯基)丙烷-2-d-2-胺(164mg,0.91mmol)依次加入到4-溴-2-氟-1-硝基苯(200mg,0.90mmol)的无水DMF(3mL)溶液中,反应升温至100℃下反应5hrs。加水淬灭反应,用乙酸乙酯(20mLx3)萃取,合并有机层用水和食盐水洗涤,无水硫酸钠干燥,液减压浓缩,浓缩液进行柱分离(洗脱 剂:石油醚/乙酸乙酯(v/v)=25:1)得到130mg淡黄色固体,收率为:38.1%,LC-MS(APCI):m/z=380.1(M+1)。
步骤2化合物5-溴-2-硝基-N-(丙基-2-基-2-d)苯胺的合成。
将化合物5-溴-N-(4-甲氧基苯基)-2-硝基-N-(异丙基-2-基-2-d)苯胺(700mg,1.84mmol)溶于4N氯化氢的二氧六环(20mL)中,反应回流搅拌4.5hrs。减压除去二氧六环,用饱和的碳酸氢钠溶液调pH>7,二氯甲烷(100mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=99:1)得到450mg淡黄色液体,收率为94.4%。LC-MS(APCI):m/z=260.1(M+1)。
步骤3化合物5-溴-N1-(丙基-2-基-2-d)苯基-1,2-二胺的合成。
化合物5-溴-2-硝基-N-(丙基-2-基-2-d)苯胺(450mg,1.73mmol)溶入乙醇和水的混合溶剂中(v/v=20mL/8mL),加入保险粉(900mg),反应在室温下反应过夜,硅藻土过滤,乙酸乙酯洗涤滤饼,减压除去乙醇,用乙酸乙酯(100mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=20:1)得到328mg褐色固体,收率为82.8%。LC-MS(APCI):m/z=230.1(M+1)。
步骤4化合物6-溴-2-甲基-1-(丙基-2-基-2-d)-1H-苯并[d]咪唑的合成。
将化合物5-溴-N1-(丙基-2-基-2-d)苯基-1,2-二胺(885g,3.85mmol)加入到冰醋酸(15mL),反应在回流反应2hrs。冷却至室温,减压除去醋酸,残渣用饱和碳酸氢钠溶液调pH大约7,二氯甲烷(50mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=20:1)得到844mg棕红色油状物,收率为:86.7%,LC-MS(APCI):m/z=254.1(M+1)。
步骤5化合物2-甲基-1-(丙基-2-基-2-d)-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑的合成
氮气保护下,将TCHP(140mg)和Pd(Oac)2(100mg)加入到6-溴-2-甲基-1-丙基-2-基-2-d)-1H-苯并[d]咪唑(840mg),联硼酸频那醇酯(1.27g)和乙酸钾(987mg)的无水DMSO(10mL)混合物中,氮气保护下,反应在100℃下反应2hrs。冷却室温,加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=2:1),得到566mg黄色固体,收率为56.5%。LC-MS(APCI):m/z=302.2(M+1)。
步骤6化合物6-(2-氯嘧啶-4-基)-2-甲基-1-(丙基-2-基-2-d)-1H-苯并[d]咪唑的合成
氮气保护下,将Pd(PPh3)2Cl2(100mg)加入到2-甲基-1-(丙基-2-基-2-d)-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(566mg,1.88mmol),2,4-二氯嘧啶(336mg,2.26mmol)和碳酸钠(498mg,4.70mmol)的乙腈(9mL)和水(3mL)的混合物 中,氮气保护下反应升温至80℃反应2.5hrs。冷却至室温,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=1:1),得到500mg白色固体。收率为92.7%。LC-MS(APCI):m/z=288.2(M+1)。
步骤7化合物N1-(2-(二甲基氨基)乙基)-5-甲氧基-N1-甲基-N4-(4-(2-甲基-1-(丙基-2-基-2-d)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-2-硝基苯-1,4-二胺的合成。
氮气保护下,将Pd(Oac)2(120mg)和Xantphos(200mg)加入到6-(2-氯嘧啶-4-基)-2-甲基-1-(丙基-2-基-2-d)-1H-苯并[d]咪唑(500mg,1.74mmol),N1-(2-(二甲基氨基)乙基)-5-甲氧基-N1-甲基-2-硝基苯-1,4-二胺(467mg,1.74mmol)和碳酸铯(1.40g,4.36mmol)的无水DMF(15mL)中,反应液氮气保护下100℃下反应过夜,冷却至室温,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1),得到棕褐色油状物。LC-MS(APCI):m/z=520.3(M+1)。
步骤8化合物N1-(2-(二甲基氨基)乙基)-5-甲氧基-N1-甲基-N4-(4-(2-甲基-1-(丙基-2-基-2-d)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)苯-1,2,4-三胺的合成。
将还原铁粉(6eq)和氯化铵(3eq)加入到N1-(2-(二甲基氨基)乙基)-5-甲氧基-N1-甲基-N4-(4-(2-甲基-1-(丙基-2-基-2-d)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-2-硝基苯-1,4-二胺的乙醇和水(16mL/4mL)混合溶液中,反应回流反应2hrs,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=8:1),得到300mg红色固体,三步收率为35.3%。LC-MS(APCI):m/z=490.2(M+1)。
步骤9化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基-5-((4-(2-甲基-1-(丙基-2-基-2-d)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到110mg,收率为:33.2%。HPLC:95.14%。LC-MS(APCI):m/z=544.2(M+1),1H NMR(500MHz,DMSO-d6):δ10.56(br,1H),9.82(s,1H),8.76(s,1H),8.45(d,J=5.3Hz,1H),8.33(s,1H),8.12(s,1H),8.04(d,J=8.5Hz,1H),7.55(d,J=8.5Hz,1H),7.49(d,J=5.3Hz,1H),6.94(s,1H),6.23(d,J=16.9Hz,1H),5.69(d,J=10.2Hz,1H),3.86(s,3H),3.28–3.21(m,5H),2.68(s,6H),2.59–2.56(m,5H),1.55(s,6H).
实施例52 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基-5-((4-(2-甲基-1-(丙基-2-基-1,1,1,3,3,3-d6)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)苯基)丙烯酰胺的制备
Figure PCTCN2017094102-appb-000166
步骤1 5-溴-N-(4-甲氧基苯基)-2-硝基-N-(异丙基-2-基-1,1,1,3,3,3-d6)苯胺的合成。
室温下将碳酸钾(4.10g,30.0mmol)和N-(4-甲氧基苯基)丙烷-1,1,1,3,3,3-d6-2-胺(1.85g,10.00mmol)依次加入到4-溴-2-氟-1-硝基苯(2.18g,10.0mmol)的无水DMF(10mL)溶液中,反应升温至100℃下反应5hrs。加水淬灭反应,用乙酸乙酯(100mLx3)萃取,合并有机层用水和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=25:1)得到3.00g淡黄色液体,收率为:80%,LC-MS(APCI):m/z=385.1(M+1)。
步骤2 5-溴-2-硝基-N-(丙基-2-基-1,1,1,3,3,3-d6)苯胺的合成。
将5-溴-N-(4-甲氧基苯基)-2-硝基-N-(异丙基-2-基-1,1,1,3,3,3-d6)苯胺(3.00g,7.79mmol)溶于4N氯化氢的二氧六环(20mL)中,反应回流搅拌4.5hrs。减压除去二氧六环,用饱和的碳酸氢钠溶液调pH>7,二氯甲烷(100mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=99:1)得到1.54g淡黄色液体,收率为75%。LC-MS(APCI):m/z=265.1(M+1)。
步骤3 5-溴-N1-(丙基-2-基-1,1,1,3,3,3-d6)苯基-1,2-二胺的合成。
将5-溴-2-硝基-N-(丙基-2-基-1,1,1,3,3,3-d6)苯胺(1.54g,5.84mmol)溶入乙醇和水的混合溶剂中(v/v=120mL/50mL),加入保险粉(5.93g,28.8mmol),反应在室温下反应过夜,硅藻土过滤,乙酸乙酯洗涤滤饼,减压除去乙醇,用乙酸乙酯(100mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=20:1)得到1.00g淡黄色液体,收率为73.2%。LC-MS(APCI):m/z=235.1(M+1)。
步骤4 6-溴-2-甲基-1-(丙基-2-基-1,1,1,3,3,3-d6)-1H-苯并[d]咪唑的合成。
将5-溴-N1-(丙基-2-基-1,1,1,3,3,3-d6)苯基-1,2-二胺(1.00g,4.27mmol)加入到冰醋酸 (10mL),反应在回流反应2hrs。冷却至室温,减压除去醋酸,残渣用饱和碳酸氢钠溶液调pH大约7,二氯甲烷(50mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=20:1)得到580mg棕红色油状物,收率为:80.22%,LC-MS(APCI):m/z=259.1(M+1).
步骤5 2-甲基-1-(丙基-2-基-1,1,1,3,3,3-d6)-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑的合成。
氮气保护下,将TCHP(40mg,0.12mmol)和Pd(Oac)2(15mg,0.06mmol)加入到6-溴-2-甲基-1-(丙基-2-基-1,1,1,3,3,3-d6)-1H-苯并[d]咪唑(300mg,1.24mmol),联硼酸频那醇酯(380mg,1.50mmol)和乙酸钾(370mg,3.72mmol)的无水DMSO(10mL)混合物中,氮气保护下,反应在100℃下反应2hrs。冷却室温,加水淬灭反应,用乙酸乙酯(50mLx3)萃取,合并有机层用饱和食盐水洗涤,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=2:1),得到266mg黄色固体,收率为69.8%。LC-MS(APCI):m/z=307.2(M+1)。
步骤6 6-(2-氯嘧啶-4-基)-2-甲基-1-(丙基-2-基-1,1,1,3,3,3-d6)-1H-苯并[d]咪唑的合成
氮气保护下,将Pd(PPh3)2Cl2(80mg)加入到2-甲基-1-(丙基-2-基-1,1,1,3,3,3-d6)-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(560mg,1.80mmol),2,4-二氯嘧啶(320mg,.17mmol)和碳酸钠(4.00mg,3.60mmol)的乙腈(12mL)和水(4mL)的混合物中,氮气保护下反应升温至80℃反应2.5hrs。冷却至室温,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=1:1),得到560mg白色固体。收率为100%。LC-MS(APCI):m/z=293.2(M+1)。
步骤7化合物N1-(2-(二甲基氨基)乙基)-5-甲氧基-N1-甲基-N4-(4-(2-甲基-1-(丙基-2-基-1,1,1,3,3,3-d6)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-2-硝基苯-1,4-二胺的合成。
合成步骤与实施例20的步骤1相同,得到400mg棕褐色油状物。LC-MS(APCI):m/z=525.3(M+1)。
步骤8N1-(2-(二甲基氨基)乙基)-5-甲氧基-N1-甲基-N4-(4-(2-甲基-1-(丙基-2-基-1,1,1,3,3,3-d6)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)苯-1,2,4-三胺的合成
将还原铁粉(6eq)和氯化铵(3eq)加入到N1-(2-(二甲基氨基)乙基)-5-甲氧基-N1-甲基-N4-(4-(2-甲基-1-(丙基-2-基-1,1,1,3,3,3-d6)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)-2-硝基苯-1,4-二胺的乙醇和水(16mL/4mL)混合溶液中,反应回流反应2hrs,硅藻土过滤,滤液减压浓缩,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=8:1),得到370mg红色固体,三步收率为41.5%。LC-MS(APCI):m/z=495.2(M+1)。
步骤9化合物N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基-5-((4-(2-甲基-1-(丙基-2-基-1,1,1,3,3,3-d6)-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)苯基)丙烯酰胺的合成。
合成步骤与实施例20的步骤3相同,得到200mg,收率为:48.7%。HPLC:95.55%。LC-MS(APCI):m/z=549.2(M+1),1H NMR(400MHz,DMSO-d6):δ10.01(s,1H),8.90(s,1H),8.46(d,J=5.3Hz,1H),8.34(s,1H),8.14(s,1H),8.09(dd,J=8.5,1.4Hz,1H),7.58(d,J=8.5Hz,1H),7.50(d,J=5.3Hz,1H),7.01(s,1H),6.74–6.44(m,1H),6.26(dd,J=16.9,1.9Hz,1H),5.80–5.70(m,1H),4.84–4.72(m,1H),3.87(s,3H),3.09–2.91(m,2H),2.68(s,3H),2.59(s,3H),2.50–2.27(m,8H).
实施例53 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺(化合物5)的制备
Figure PCTCN2017094102-appb-000167
具体合成步骤如下:
Figure PCTCN2017094102-appb-000168
步骤1化合物3的合成。
氮气保护下将乙二醇二甲醚(2mL)和水(1mL)加入到4-氟-1-异丙基-2-甲基-6-(4,4,5,5-四甲基-1,3,2-二氧杂硼杂环戊烷-2-基)-1H-苯并[d]咪唑(化合物1,260mg,0.82mmol)和2,4-二氯嘧啶(化合物2,122.2mg,0.82mmol)、碳酸钠(217mg,2.05mmol)、双三苯基磷二氯化钯(50mg,0.07mmol)的混合物中,反应在氮气保护下,80℃搅拌反应1小时。冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼,用饱和食盐水洗涤滤液,无水硫酸钠干燥,减压浓缩滤液得到330mg淡黄色固体,直接用于下一步反应。收率为100%。LC-MS(APCI):m/z=305.1(M+1)。
步骤2化合物5的合成。
氮气保护下将无水二氧六环(4mL)加入到6-(2-氯嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑(化合物3,330mg,0.82mmol)和N-(5-氨基-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺(化合物4,290mg,1.00mmol)、碳酸铯(667mg,2.1mmol)、Pd2(dba)3(38mg)和4,5-双二苯基膦-9,9-二甲基氧杂蒽(Xant-Phos,49mg)的混合物中,反应在氮气保护下,110℃搅拌反应过夜。冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1),得到粗品,制备TLC纯化(DCM/MeOH v/v=15/1)得到25mg黄色固体。收率为5.44%。LC-MS(APCI):m/z=561.4(M+1);1H NMR(300MHz,DMSO-d6)(δ/ppm)9.91(br,1H),8.84(s,1H),8.48(d,J=5.3Hz,1H),8.32–8.13(m,2H),7.89(d,J=12.5Hz,1H),7.56(d,J=5.3Hz,1H),6.99(s,1H),6.25(d,J=17.3Hz,1H),5.77–5.70(m,2H),4.90–4.74(m,1H),3.85(s,3H),2.69–2.62(m,4H),2.60(s,3H),2.51–2.48(m,9H),1.57(d,J=6.9Hz,6H)。
实施例54 4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(2-甲氧基-4-(4-甲基哌嗪-1-基)苯基)嘧啶-2-胺(化合物7)的制备
Figure PCTCN2017094102-appb-000169
具体合成步骤如下:
Figure PCTCN2017094102-appb-000170
步骤1化合物7的合成。
将对甲苯磺酸(150mg,0.84mmol)加到6-(2-氯嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑(化合物3,130mg,0.42mmol)和2-甲氧基-4-(4-甲基哌嗪-1-基)苯胺(化 合物6,111mg,0.50mmol)的异丙醇(3mL)溶液中,微波反应180℃反应1小时。减压浓缩反应液,残渣用饱和碳酸氢钠调至碱性,二氯甲烷(30mLx3)萃取,合并有机相,饱和食盐水(30mL)洗涤无水硫酸钠干燥,除去溶剂,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=13:1),得到淡黄固体40mg,收率为19.4%。LC-MS(APCI):m/z=490.3(M+1);1H NMR(500MHz,DMSO-d6)(δ/ppm)8.42(d,J=5.2Hz,1H),8.26(s,1H),8.06(s,1H),7.82(d,J=8.7Hz,1H),7.74(d,J=12.1Hz,1H),7.44(d,J=5.2Hz,1H),6.67(d,J=2.2Hz,1H),6.51(dd,J=8.7,2.3Hz,1H),4.90–4.71(m,1H),3.81(s,3H),3.21–3.14(m,4H),2.67–2.53(m,7H),2.32(s,3H),1.59(d,J=6.9Hz,6H)。
实施例55 5-氟-4-(4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑-6-基)-N-(2-甲氧基-4-(4-甲基哌嗪-1-基)苯基)嘧啶-2-胺(化合物9)
Figure PCTCN2017094102-appb-000171
具体合成步骤如下:
Figure PCTCN2017094102-appb-000172
氮气保护下将叔戊醇(5mL)加入到6-(2-氯-5-氟嘧啶-4-基)-4-氟-1-异丙基-2-甲基-1H-苯并[d]咪唑(化合物8,200mg,0.62mmol)和2-甲氧基-4-(4-甲基哌嗪-1-基)苯胺(化合物6,165mg,0.75mmol)、碳酸钾(214mg,1.55mmol)、Pd2(dba)3(37mg)和Xant-Phos(46mg)的混合物中,反应在氮气保护下,100℃搅拌反应3小时。冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=13:1),得到47mg黄色固体。收率为14.9%。LC-MS(APCI):m/z=508.3(M+1);1H NMR(500MHz,DMSO-d6)(δ/ppm)8.49(d,J=3.7Hz,1H),8.21(s,1H),8.14(s,1H),7.65(d,J=8.6Hz,1H),7.59(d,J=12.2Hz,1H),6.65(d,J=1.1Hz,1H), 6.48(dd,J=8.6,1.8Hz,1H),4.79(M,1H),3.79(s,3H),3.16(M,4H),2.61(s,3H),2.56(M,4H),2.29(s,3H),1.56(d,J=6.8Hz,6H)。
实施例56 N-(2-((2-(二甲基氨基)乙基)(甲基)氨基)-5-((5-氟-4-(8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶-6-基)嘧啶-2-基)氨基)-4-甲氧基苯基)丙烯酰胺(化合物18)的制备
Figure PCTCN2017094102-appb-000173
具体合成步骤如下:
Figure PCTCN2017094102-appb-000174
步骤1化合物11的合成。
氮气保护下将N-溴代丁二酰亚胺(NBS,5.47g,30.57mmol)分批加入到3-氟吡啶-2-胺(化合物10,3.36g,29.97mmol)的乙腈(150mL)中,反应在氮气保护下室温反应2小时。减压浓缩除去乙腈,加饱和硫代硫酸钠溶液淬灭,用乙酸乙酯萃取(100mLx3)合并有机相用饱和食盐水洗涤滤液,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=5:1),得到5.0g白色固体,收率为87.3%。LC-MS(APCI):m/z=191.1(M+1)。
步骤2化合物12的合成。
将1-溴丙-2-酮(10.00g,73.3mmol)加入到5-溴-3-氟吡啶-2-胺(化合物11,7.00g,36.6mmol)的乙醇(70mL)中,反应在氮气保护下回流反应过夜。冷却至温室慢慢析出 固体,过滤,用冷乙醇洗涤,真空干燥得到3.5g白色固体,收率:41.8%。LC-MS(APCI):m/z=229.1(M+1);1H NMR(400MHz,DMSO-d6)(δ/ppm)9.12(s,1H),8.16–8.09(m,2H),2.48(s,3H)。
步骤3化合物13的合成。
将单质碘(4.60g,18.3mmol)加入到6-溴-8-氟-2-甲基咪唑并[1,2-a]吡啶(化合物12,3.50g,15.30mmol)和乙酸钠(2.01g,24.5mmol)的甲醇(20mL)中,反应在氮气保护下室温反应3小时。减压浓缩除去甲醇,加饱和硫代硫酸钠溶液淬灭,用乙酸乙酯萃取(100mLx3)合并有机相用饱和食盐水洗涤滤液,无水硫酸钠干燥,减压浓缩,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1),得到2.8g白色固体,收率为51.7%。LC-MS(APCI):m/z=355.1(M+1)。
步骤4化合物15的合成。
氮气保护下将Pd(dppf)Cl2■CH2Cl2(250mg,0.31mmol)加入到6-溴-8-氟-3-碘-2-甲基咪唑并[1,2-a]吡啶(化合物13,1.17g,3.32mmol)、异丙烯基硼酸频哪醇酯(化合物14,557mg,3.31mmol)和碳酸铯(2.16g,6.64mmol)的1,4-二氧六环(20mL)和水(5mL)混合溶剂中,反应在氮气保护下80℃反应1小时。冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=10:1),得到600mg白色固体。收率为67.4.9%。LC-MS(APCI):m/z=269.1(M+1)。
步骤5化合物17的合成。
氮气保护下将Pd(dppf)Cl2■CH2Cl2(200mg,0.25mmol)加入到6-溴-8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶(化合物15,660mg,2.46mmol)、联硼酸频那醇酯(750mg,3.00mmol)和醋酸钾(490mg,4.92mmol)的无水1,4-二氧六环(16mL)溶剂中,反应在氮气保护下80℃反应2小时。冷却至室温,在氮气保护下将2,4-二氯-5-氟嘧啶(化合物16,410mg,2.46mmol),碳酸钠(520mg,4.92mmol)和水(4mL)加入到反应液中,反应在氮气保护下80℃反应2小时,冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:石油醚/乙酸乙酯(v/v)=3:1),得到600mg白色固体,收率为76.2%。LC-MS(APCI):m/z=321.1(M+1)。
步骤6化合物18的合成。
氮气保护下将叔戊醇(10mL)加入到6-(2-氯-5-氟嘧啶-4-基)-8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶(化合物17,125mg,0.38mmol)和N-(5-氨基-2-((2-(二甲基氨基)乙基)(甲基)氨基)-4-甲氧基苯基)丙烯酰胺(化合物4,125mg,0.43mmol)、碳酸钾(135mg,0.98mmol)、Pd2(dba)3(30mg)和Xant-Phos(40mg)的混合物中,反应在氮气保护下,100℃搅拌反应3小时。冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼,减压浓缩 滤液,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=10:1),得到粗品固体,制备TLC纯化(DCM/MeOH v/v=10/1)得到26mg淡黄色固体,收率为11.9%。LC-MS(APCI):m/z=577.6(M+1);1H NMR(400MHz,CDCl3)(δ/ppm)9.60(s,1H),8.77(s,1H),8.41(d,J=3.7Hz,1H),8.12(d,J=11.8Hz,1H),7.72(s,1H),6.75(s,1H),6.53(d,J=15.5Hz,1H),5.75(d,J=9.5Hz,1H),5.64(s,1H),5.30(s,1H),3.93(s,3H),3.33–3.18(m,2H),3.14–2.90(m,2H),2.86–2.63(m,9H),2.51(s,3H),2.19(s,3H)。
实施例57 5-氟-4-(8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶-6-基)-N-(2-甲氧基-4-(4-甲基哌嗪-1-基)苯基)嘧啶-2-胺(化合物19)
Figure PCTCN2017094102-appb-000175
具体合成步骤如下:
Figure PCTCN2017094102-appb-000176
氮气保护下将叔戊醇(10mL)加入到6-(2-氯-5-氟嘧啶-4-基)-8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶(化合物17,260mg,0.81mmol)和2-甲氧基-4-(4-甲基哌嗪-1-基)苯胺(化合物6,150mg,0.67mmol)、碳酸钾(234mg,1.70mmol)、Pd2(dba)3(70mg)和Xant-Phos(80mg)的混合物中,反应在氮气保护下,100℃搅拌反应3小时。冷却至室温,硅藻土过滤,用二氯甲烷洗涤滤饼,减压浓缩滤液,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=13:1),得到115mg深黄色固体,收率为14.9%。LC-MS(APCI):m/z=506.6(M+1);1H NMR(500MHz,DMSO-d6)(δ/ppm)8.72(d,J=1.2Hz,1H),8.50(d,J=3.8Hz,1H),8.32(s,1H),7.61(d,J=12.4Hz,1H),7.55(d,J=8.7Hz,1H),6.64(d,J=2.5Hz,1H),6.48(dd,J=8.7,2.5Hz,1H),5.68–5.64(m,1H),5.32–5.29(m,1H),3.78(s,3H),3.19–3.14(m,4H),2.61–2.54(m,4H),2.38(s,3H),2.30(s,3H),2.13(s,3H)。
实施例58 5-氟-4-(8-氟-2-甲基咪唑[1,2-a]吡啶-6-基)-N-(2-甲氧基-4-(4-甲基哌嗪-1-基)苯基)嘧啶-2-胺(化合物20)
Figure PCTCN2017094102-appb-000177
具体合成步骤如下:
Figure PCTCN2017094102-appb-000178
将对甲苯磺酸(269mg,1.56mmol)加到6-(2-氯-5-氟嘧啶-4-基)-8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶(化合物17,250mg,0.78mmol)和2-甲氧基-4-(4-甲基哌嗪-1-基)苯胺(化合物6,172mg,0.78mmol)的异丙醇(10mL)溶液中,微波反应180℃反应1.5小时。减压浓缩反应液,残渣用饱和碳酸氢钠调至碱性,二氯甲烷(30mLx3)萃取,合并有机相,饱和食盐水(30mL)洗涤无水硫酸钠干燥,除去溶剂,浓缩液进行柱分离(洗脱剂:二氯甲烷/甲醇(v/v)=13:1),得到淡黄固体50mg粗品,制备TLC纯化(DCM/MeOH v/v=12/1)得到32mg淡黄色固体,收率为8.8%。LC-MS(APCI):m/z=466.5(M+1);1H NMR(400MHz,CDCl3)(δ/ppm)8.77(s,1H),8.32(d,J=3.6Hz,1H),8.18(d,J=8.7Hz,1H),7.75(d,J=12.1Hz,1H),7.52(s,1H),7.50–7.45(m,1H),6.63–6.58(m,1H),6.58–6.55(m,1H),3.90(s,3H),3.24–3.17(m,4H),2.66–2.59(m,4H),2.50(s,3H),2.37(s,3H)。
实施例59 5-氟-4-(8-氟-3-异丙基-2-甲基咪唑并[1,2-a]吡啶-6-基)-N-(2-甲氧基-4-(4-甲基哌嗪-1-基)苯基)嘧啶-2-胺(化合物21)
Figure PCTCN2017094102-appb-000179
具体合成步骤如下:
Figure PCTCN2017094102-appb-000180
室温下将Pd/C(20mg)加入到5-氟-4-(8-氟-2-甲基-3-(丙-1-烯-2-基)咪唑并[1,2-a]吡啶-6-基)-N-(2-甲氧基-4-(4-甲基哌嗪-1-基)苯基)嘧啶-2-胺(化合物19,80mg,0.16mmol)的甲醇(5mL)溶剂中,反应在氢气球下反应搅拌反应48小时。硅藻土过滤,用二氯甲烷洗涤滤饼,减压浓缩滤液,浓缩液进行柱分离得到40mg淡黄色固体,收率为50%。LC-MS(APCI):m/z=508.1(M+1);1H NMR(500MHz,DMSO-d6)(δ/ppm)8.76(s,1H),8.54(d,J=3.7Hz,1H),8.35(s,1H),7.63–7.54(m,2H),6.67(d,J=2.2Hz,1H),6.51(dd,J=8.8,2.3Hz,1H),3.80(s,3H),3.51–3.43(m,1H),3.22–3.12(m,4H),2.63–2.53(m,4H),2.43(s,3H),2.31(s,3H),1.38(d,J=7.1Hz,6H)。
生物活性测试
(1)激酶抑制作用
试剂和耗材:
WT EGFR(Carna,目录号08-115),EGFR[L858R](Carna,目录号08-502),EGFR[L858R/T790M](Carna,目录号08-510),ATP(Sigma,目录号A7699-1G),DMSO(Sigma,目录号D2650),96孔板(Corning,目录号3365),384孔板(Greiner,目录号784076),HTRF Kinase TK试剂盒(Cisbio,目录号62TK0PEJ),厄洛替尼(Selleckchem,目录号S7787),EGFR[d746-750](Life Technologies,目录号PV6178),5x激酶缓冲液 A(Life Technologies,目录号PV3186),激酶示踪剂199(Life Technologies,目录号PV5830),
Figure PCTCN2017094102-appb-000181
 Eu-anti-GST抗体(Life Technologies,目录号PV5594)。
具体实验方法:
化合物配制:将受试化合物溶于DMSO配成20mM母液。然后,在DMSO中等梯度3倍稀释,稀释十次。加药时再用缓冲液稀释10倍。
WT EGFR及EGFR[L858R/T790M]激酶检测:在5x激酶缓冲液A中,将WT EGFR或EGFR[L858R/T790M]激酶与预先稀释配制的不同浓度的化合物混合10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,阳性为厄洛替尼)。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Evnvision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IC50值,其中A表示IC50≤1nM,B表示IC50为1-50nM,C表示IC50为50-100nM,D表示IC50>100nM。
在上述激酶抑制实验中测试了本发明化合物,发现本发明化合物对EGFR[L858R/T790M]具有强效的活性以及优于WT EGFR的优异选择性。代表性实施例化合物的结果归纳于如下表1中。
表1
Figure PCTCN2017094102-appb-000182
Figure PCTCN2017094102-appb-000183
Figure PCTCN2017094102-appb-000184
(2)细胞毒性实验
采用MTS方法检测了本发明化合物对体外培养的3株肿瘤细胞的体外抗增殖活性。实验结果表明本发明化合物对体外培养的癌细胞的体外增殖具有抑制作用;其中对肺癌细胞的体外增殖的抑制作用比皮肤癌细胞的体外增殖的抑制作用强。
细胞系:
皮肤癌细胞A431(购自美国标准生物品收藏中心(ATCC));肺癌细胞NCI-H1975(购自美国标准生物品收藏中心(ATCC))和HCC827(购自美国标准生物品收藏中心(ATCC));均用含10%胎牛血清、100U/ml青霉素、100μg/ml链霉素的RPMI1640培养基培养。
试剂和耗材:
RPMI-1640(GIBCO,目录号A10491-01);胎牛血清(GIBCO,目录号10099141);0.25%胰蛋白酶-EDTA(GIBCO,目录号25200);青霉素-链霉素,液体(GIBCO,目录号15140-122);DMSO(Sigma,目录号D2650);MTS测试试剂盒(Promega,目录号G3581),96孔板(Corning,目录号3365)。
具体实验方法:
化合物配制:受试化合物溶于DMSO配成20mM母液,-20℃保存。使用时用DMSO等梯度3倍稀释,稀释10次。加药时再用细胞培养基RPMI-1640稀释4倍。
MTS细胞活力检测:0.25%胰蛋白酶-EDTA消化对数生长期细胞,按已优化的密度接种150μl于96孔板,24小时后加入培养基稀释4倍的化合物,50μl/孔(一般选择十个浓度:100、33.3、11.1、3.70、1.23、0.412、0.137、0.0457、0.0152、0.00508μM)。以加入同样体积的0.5%DMSO的孔作为对照。细胞继续培养72小时后,MTS检测细胞活力。
具体操作:贴壁细胞,弃去培养基,每孔加入含20μLMTS和100μL培养基的混合液。放入培养箱继续培养1-4小时后检测OD490,以OD650值作为参考。用GraphPad Prism软件制作量效曲线并计算IC50,其中A表示IC50≤10nM,B表示IC50为10-100nM,C表示IC50为100-400nM,D表示IC50为400-1000nM,并且E表示IC50≥1000nM。
在上述细胞毒性实验中测试了本发明化合物,发现本发明化合物对肺癌细胞NCI-H1975和HCC827具有强效的活性以及优于皮肤癌细胞A431的优异选择性。代表性实施例对癌细胞的体外增殖的抑制作用的结果归纳于下表2中。
表2
Figure PCTCN2017094102-appb-000185
Figure PCTCN2017094102-appb-000186
Figure PCTCN2017094102-appb-000187
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,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。
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 (16)

  1. 式(I)化合物:
    Figure PCTCN2017094102-appb-100001
    其中,
    R1选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷氧基、任选取代的C3-C6碳环基、任选取代的C1-C6烷硫基、任选取代的C1-C6烷氨基或任选取代的C1-C6烷酰基;
    R2选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷氧基、任选取代的C1-C6烷硫基、任选取代的C1-C6烷氨基或任选取代的C1-C6烷酰基;
    L选自键、NR7、O、C(R7)2或S;其中各R7独立地选自H、任选取代的C1-C6烷基例如被C1-C6烷氧基取代的C1-C6烷基、或任选取代的C1-C6烷氧基;
    Y为H,或者Y选自以下结构:
    Figure PCTCN2017094102-appb-100002
    其中R3、R4和R5独立地选自H、卤素、-CN、任选取代的C1-C6烷基或任选取代的C1-C6卤代烷基;
    R6选自H、C1-C6烷基、C2-C6炔基或3至8元杂环基,其中上述基团任选被1-3个以下取代基取代:卤素、-OH、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基、-C(O)R8或NR8R9;其中R8和R9各自独立地选自H、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基、任选取代的C1-C6卤代烷基或任选取代的C3-C6碳环基,或者R8和R9与它们相连的氮原子一起形成任选取代的4至6元杂环基,例如被C1-C6烷基取代;
    W选自键、C(R10)2、NR10、O或S,其中R10选自H、卤素、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基或任选取代的C1-C6烷氧基,只要化学上允许;
    m为0、1或2;
    n为0、1或2;
    环A选自以下结构:
    Figure PCTCN2017094102-appb-100003
    其中环A任选被1-6个R11取代基取代,其中各X4独立地选自C(R11)2、NR11、O、S、C(O)、S(O)或S(O)2,各X5独立地选自C(R11)2或NR11;前提是环A不是
    Figure PCTCN2017094102-appb-100004
    R11独立地选自H、卤素、-CN、-NO2、-OH、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C2-C6烯基、任选取代的C2-C6炔基、任选取代的C3-C6碳环基、任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基,只要化学上允许;或者,其中两个相邻的R11可一起形成任选取代的C5-C8碳环基、任选取代的5至8元杂环基、任选取代的C6-C14芳基或任选取代的5至10元杂芳基;
    或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
  2. 根据权利要求1所述的化合物,其为式(III)化合物:
    Figure PCTCN2017094102-appb-100005
    其中,
    Y选自以下结构:
    Figure PCTCN2017094102-appb-100006
    优选地,Y选自以下结构:
    Figure PCTCN2017094102-appb-100007
    优选地,Y为以下结构:
    Figure PCTCN2017094102-appb-100008
    优选地,Y为以下结构:
    Figure PCTCN2017094102-appb-100009
    并且R1-R6、L、A和n如权利要求1所定义,
    或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
  3. 根据权利要求1所述的化合物,其为式(IV)化合物:
    Figure PCTCN2017094102-appb-100010
    其中,R1、R2、L、R6、A和n如权利要求1所定义,
    或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
  4. 根据权利要求1-3中任一项所述的化合物,其中
    环A选自以下基团:
    Figure PCTCN2017094102-appb-100011
    Figure PCTCN2017094102-appb-100012
    Figure PCTCN2017094102-appb-100013
    Figure PCTCN2017094102-appb-100014
    R11独立地选自H、卤素、任选取代的C1-C6烷基、任选取代的C2-C6烯基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基或任选取代的C3-C6碳环基;或者,其中两个相邻的R11可一起形成任选取代的5至8元杂环基;
    或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
  5. 根据权利要求1-4中任一项所述的化合物,其中
    L选自键、NR7、O或S;其中R7选自H或任选取代的C1-C6烷基;
    n为0、1或2;
    R6选自H、C1-C6烷基或3至8元杂环基,其中上述基团任选被1-3个以下取代基取代:卤素、-OH、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基或NR8R9,其中R8和R9各自独立地选自H或任选取代的C1-C6烷基;或者R8和R9与它们相连的氮原子一起形成任选取代的4至6元杂环基;
    优选地,
    L选自键或NR7;其中R7选自H或任选取代的C1-C6烷基;
    n为0或1;
    R6选自H、C1-C6烷基或3至8元杂环基,其中上述基团任选被1-3个以下取代基取代:卤素、-OH、-CN、-NO2、任选取代的C1-C6烷基、任选取代的C1-C6烷氧基或NR8R9,其中R8和R9各自独立地选自H或任选取代的C1-C6烷基;或者R8和R9与它们相连的氮原子一起形成任选取代的4至6元杂环基;
    优选地,-(L)n-R6选自:
    Figure PCTCN2017094102-appb-100015
    Figure PCTCN2017094102-appb-100016
    优选地,-(L)n-R6选自:
    Figure PCTCN2017094102-appb-100017
    优选地,-(L)n-R6选自:
    Figure PCTCN2017094102-appb-100018
    Figure PCTCN2017094102-appb-100019
    优选地,-(L)n-R6选自:
    Figure PCTCN2017094102-appb-100020
    或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
  6. 根据权利要求1-5中任一项的化合物,其中
    R1选自H、卤素、-CN、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基或任选取代的C3-C6碳环基;
    R2选自任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;
    -(L)n-R6选自:
    Figure PCTCN2017094102-appb-100021
    Figure PCTCN2017094102-appb-100022
    环A选自:
    Figure PCTCN2017094102-appb-100023
    其中,R11独立地选自H、卤素、任选取代的C1-C6烷基、任选取代的C2-C6烯基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基或任选取代的C3-C6碳环基;或者,其中两个相邻的R11可一起形成任选取代的5至8元杂环基;
    或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
  7. 根据权利要求1-6中任一项的化合物,其中
    环A为
    Figure PCTCN2017094102-appb-100024
    -(L)n-R6选自:
    Figure PCTCN2017094102-appb-100025
    或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位 素衍生物。
  8. 根据权利要求1-6中任一项的化合物,其中
    -(L)n-R6
    Figure PCTCN2017094102-appb-100026
    环A选自:
    Figure PCTCN2017094102-appb-100027
    或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
  9. 根据权利要求1的化合物,其为式(III-a)化合物:
    Figure PCTCN2017094102-appb-100028
    其中,
    R1选自H、卤素、-CN、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基、任选取代的C1-C6烷氧基或任选取代的C3-C6碳环基;优选地,R1选自H、卤素、任选取代的C1-C6烷基或任选取代的C3-C6碳环基;优选地,R1选自H、-F、-Cl、-Br、-CN、-CH3、-CF3、-OCH3或-环丙烷基;优选地,R1选自H、-Cl、-Br、-CH3或-环丙烷基;
    R2选自任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;优选地,R2选自-OCH3、-OCH2CH3、-OCHF2或-OCH2CF3
    R11选自H、卤素、任选取代的C1-C6烷基、任选取代的C2-C6烯基、任选取代的 C1-C6卤代烷基或任选取代的C1-C6烷氧基;优选地,R11选自H、卤素或任选取代的C1-C6烷氧基;优选地,R11选自H、-F、-Cl、-Br、-Me、-iPr、1-丙烯-2-基、-CH2F、CF3、-OMe或-OC(CH3)3;优选地,R11选自H、-F、-Cl、-Br、-OMe或-OC(CH3)3
    R12选自H、卤素、任选取代的C1-C6烷基、任选取代的C2-C6烯基、任选取代的C1-C6卤代烷基或任选取代的C1-C6烷氧基;优选地,R12选自任选取代的C1-C6烷基或任选取代的C1-C6卤代烷基;优选地,R12选自H、-F、-Cl、-Br、-Me、-iPr、1-丙烯-2-基、-CH2F、CF3、-OMe或-OC(CH3)3;优选地,R12选自-Me、-iPr或-CH2F;
    R13选自H、卤素、任选取代的C1-C6烷基、任选取代的C2-C6烯基、任选取代的C1-C6卤代烷基或任选取代的C1-C6烷氧基;优选地,R13为任选取代的C1-C6烷基;优选地,R13选自H、-F、-Cl、-Br、-Me、-iPr、1-丙烯-2-基、-CH2F、CF3、-OMe或-OC(CH3)3;优选地,R13选自-Me或-iPr;
    或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
  10. 根据权利要求9的化合物,其中
    R1选自H、-Cl、-Br、-CH3或-环丙烷基;
    R2选自-OCH3、-OCH2CH3、-OCHF2或-OCH2CF3
    R11选自H、-F、-Cl、-Br、-OMe或-OC(CH3)3
    R12选自-Me、-iPr或-CH2F;
    R13选自-Me或-iPr;
    或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
  11. 根据权利要求1所述的化合物,其为式(IV-a)化合物:
    Figure PCTCN2017094102-appb-100029
    其中,
    R1选自H、卤素、-CN、任选取代的C1-C6烷基、任选取代的C1-C6卤代烷基或任选取代的C1-C6烷氧基;优选地,R1选自H或-F;
    R2选自任选取代的C1-C6烷氧基或任选取代的C1-C6卤代烷氧基;优选地,R2选自 H或-OCH3
    A选自
    Figure PCTCN2017094102-appb-100030
    或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素衍生物。
  12. 化合物,其选自:
    Figure PCTCN2017094102-appb-100031
    Figure PCTCN2017094102-appb-100032
    Figure PCTCN2017094102-appb-100033
    Figure PCTCN2017094102-appb-100034
    Figure PCTCN2017094102-appb-100035
    或其药学上可接受的盐。
  13. 药物组合物,其含有权利要求1-12中任一项的化合物或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素变体,和药学上可接受的赋形剂。
  14. 权利要求1-12中任一项的化合物或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素变体或权利要求13的药物组合物在制备用于治疗EGFR导致的疾病的药物中的用途;优选地,其中所述EGFR导致的疾病选自癌症、细胞增殖性疾病、炎症、感染、免疫性疾病、器官移植、病毒性疾病、心血管疾病或代谢性疾病;优选地,其中所述癌症选自非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、乳腺癌、***癌、肝癌、皮肤癌、上皮细胞癌、胃肠间质瘤、白血病、组织细胞性淋巴癌和鼻咽癌。
  15. 权利要求1-12中任一项的化合物或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素变体或权利要求13的药物组合物,其用于治疗EGFR导致的疾病;优选地,其中所述EGFR导致的疾病选自癌症、细胞增殖性疾病、炎症、感染、免疫性疾病、器官移植、病毒性疾病、心血管疾病或代谢性疾病;优选地,其中所述癌症选自非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、乳腺癌、***癌、肝癌、皮肤癌、上皮细胞癌、胃肠间质瘤、白血病、组织细胞性淋巴癌和鼻咽癌。
  16. 一种在受试者中治疗EGFR导致的癌症的方法,所述方法包括向所述受试者给药权利要求1-12中任一项的化合物或其药学上可接受的盐、立体异构体、溶剂合物、水合物、多晶型、前药或同位素变体,或者权利要求13的药物组合物;优选地,其中所述EGFR导致的疾病选自癌症、细胞增殖性疾病、炎症、感染、免疫性疾病、器官移植、病毒性疾病、心血管疾病或代谢性疾病;优选地,其中所述癌症选自非小细胞肺癌、小细胞肺癌、肺腺癌、肺鳞癌、胰腺癌、乳腺癌、***癌、肝癌、皮肤癌、上皮细胞癌、胃肠间质瘤、白血病、组织细胞性淋巴癌和鼻咽癌。
PCT/CN2017/094102 2016-07-26 2017-07-24 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物 WO2018019204A1 (zh)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2019503547A JP6740452B2 (ja) 2016-07-26 2017-07-24 プロテインチロシンキナーゼの活性を阻害するためのアミノピリミジン系化合物
EP17833507.1A EP3492462B1 (en) 2016-07-26 2017-07-24 Amino pyrimidine compound for inhibiting protein tyrosine kinase activity
CN201780009980.3A CN108602802B (zh) 2016-07-26 2017-07-24 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物
US16/256,203 US11111233B2 (en) 2016-07-26 2019-01-24 Amino pyrimidine compound for inhibiting protein tyrosine kinase activity

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610594608 2016-07-26
CN201610594608.0 2016-07-26

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/256,203 Continuation-In-Part US11111233B2 (en) 2016-07-26 2019-01-24 Amino pyrimidine compound for inhibiting protein tyrosine kinase activity

Publications (1)

Publication Number Publication Date
WO2018019204A1 true WO2018019204A1 (zh) 2018-02-01

Family

ID=61015656

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2017/094102 WO2018019204A1 (zh) 2016-07-26 2017-07-24 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物
PCT/CN2020/073764 WO2020151742A1 (en) 2016-07-26 2020-01-22 Amino pyrimidine compound for inhibiting protein tyrosine kinase activity

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/073764 WO2020151742A1 (en) 2016-07-26 2020-01-22 Amino pyrimidine compound for inhibiting protein tyrosine kinase activity

Country Status (5)

Country Link
US (1) US11111233B2 (zh)
EP (1) EP3492462B1 (zh)
JP (2) JP6740452B2 (zh)
CN (3) CN108602802B (zh)
WO (2) WO2018019204A1 (zh)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109851638A (zh) * 2018-02-07 2019-06-07 深圳市塔吉瑞生物医药有限公司 取代的二氨基嘧啶化合物
CN110066272A (zh) * 2018-05-29 2019-07-30 深圳市塔吉瑞生物医药有限公司 取代的苯并[d]咪唑类化合物及其药物组合物
CN110156754A (zh) * 2018-02-10 2019-08-23 杭州百诚医药科技股份有限公司 一种三取代嘧啶衍生物对蛋白激酶的抑制作用
US10435388B2 (en) 2016-01-07 2019-10-08 Cs Pharmatech Limited Selective inhibitors of clinically important mutants of the EGFR tyrosine kinase
US10457669B2 (en) 2015-10-21 2019-10-29 Otsuka Pharmaceutical Co., Ltd. Benzolactam compounds as protein kinase inhibitors
CN110698461A (zh) * 2018-07-09 2020-01-17 上海翰森生物医药科技有限公司 第三代egfr抑制剂的制备方法
WO2020135507A1 (zh) * 2018-12-26 2020-07-02 杭州百新生物医药科技有限公司 一种多取代苯氨基嘧啶衍生物及其制备方法和用途
WO2020151742A1 (en) * 2016-07-26 2020-07-30 Shenzhen Targetrx, Inc. Amino pyrimidine compound for inhibiting protein tyrosine kinase activity
WO2021000912A1 (zh) * 2019-07-04 2021-01-07 微境生物医药科技(上海)有限公司 一类抑制egfr激酶的化合物及其制备方法和用途
EP3640248A4 (en) * 2017-06-13 2021-03-31 Beijing Adamadle Biotechnology Limited Liability Company AMINOPYRIMIDINE COMPOUND, PROCESS FOR PREPARATION AND USE
WO2021185297A1 (zh) * 2020-03-18 2021-09-23 南京药石科技股份有限公司 Egfr酪氨酸激酶抑制剂及其用途
US11142518B2 (en) 2017-04-20 2021-10-12 Otsuka Pharmaceutical Co., Ltd. 6-pyrimidin-isoindole derivative as ERK1/2 inhibitor
AU2020255100A8 (en) * 2019-03-29 2021-12-02 Shenzhen Forward Pharmaceuticals Co., Ltd. N-heteroaromatic amide derivatives for treatment of cancer
CN113880813A (zh) * 2020-07-01 2022-01-04 杭州百诚医药科技股份有限公司 一种2-氨基嘧啶类杂环化合物及应用
JP2022515196A (ja) * 2018-12-21 2022-02-17 深▲チェン▼市塔吉瑞生物医薬有限公司 プロテインキナーゼ活性を阻害するためのアミノピリミジン系化合物
WO2022063140A1 (en) * 2020-09-23 2022-03-31 Shenzhen Ionova Life Science Co., Ltd. Pyrimidine and pyridine derivatives as hpk1 modulator and methods using same
WO2022166799A1 (zh) * 2021-02-03 2022-08-11 上海拓界生物医药科技有限公司 稠三环类细胞周期蛋白-依赖性激酶抑制剂及其制备方法和医药用途
US11426412B2 (en) 2017-10-18 2022-08-30 Jubilant Epipad LLC Imidazo-pyridine compounds as PAD inhibitors
US11459338B2 (en) 2017-11-24 2022-10-04 Jubilant Episcribe Llc Heterocyclic compounds as PRMT5 inhibitors
US11529341B2 (en) 2018-03-13 2022-12-20 Jubilant Prodel LLC Bicyclic compounds as inhibitors of PD1/PD-L1 interaction/activation
CN115650974A (zh) * 2022-08-04 2023-01-31 天津大学 N-[5-(嘧啶-2-氨基)-2,4-二取代苯基]-顺式戊二烯酰胺衍生物及应用
US11629135B2 (en) 2017-11-06 2023-04-18 Jubilant Prodell Llc Pyrimidine derivatives as inhibitors of PD1/PD-L1 activation
US11731958B2 (en) 2018-02-20 2023-08-22 Incyte Corporation Carboxamide compounds and uses thereof
US11787784B2 (en) 2019-08-06 2023-10-17 Incyte Corporation Solid forms of an HPK1 inhibitor
US11795166B2 (en) 2016-09-09 2023-10-24 Incyte Corporation Pyrazolopyridine compounds and uses thereof
US11833156B2 (en) 2017-09-22 2023-12-05 Jubilant Epipad LLC Heterocyclic compounds as pad inhibitors
US11866426B2 (en) 2018-08-08 2024-01-09 Incyte Corporation Benzothiazole compounds and uses thereof
US11891388B2 (en) 2016-09-09 2024-02-06 Incyte Corporation Pyrazolopyridine compounds and uses thereof

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20200028966A (ko) * 2017-07-05 2020-03-17 씨에스 파마테크 리미티드 Efgr 티로신 키나제의 임상적으로 중요한 돌연변이체의 선택적 억제제
CN109503573A (zh) * 2017-09-14 2019-03-22 昆明圣加南生物科技有限公司 2-取代苯胺基嘧啶衍生物及其用途
CN107827875B (zh) * 2017-09-25 2021-07-09 文韬创新药物研究(北京)有限责任公司 一种苯并咪唑类衍生物作为周期蛋白依赖性激酶4/6抑制剂的应用
CN109180589A (zh) * 2018-10-16 2019-01-11 武汉工程大学 玻玛西尼中间体的制备方法
TWI778366B (zh) * 2019-06-06 2022-09-21 中國商北京泰德製藥股份有限公司 作為atr激酶抑制劑的2,4,6-三取代的嘧啶化合物
CN111018789A (zh) * 2019-12-02 2020-04-17 睿辰康达生物医药(武汉)有限公司 一种6-溴-4-氟-1-异丙基-2-甲基-1h-苯并[d]咪唑的合成方法
CN111004221B (zh) * 2019-12-13 2021-02-02 长治学院 一种嘧啶/苯并咪唑杂合物、制备方法及医药用途
CN113880816A (zh) * 2020-07-01 2022-01-04 杭州百诚医药科技股份有限公司 一种含哌嗪类的氨基嘧啶衍生物及应用
CN113880814B (zh) * 2020-07-01 2024-01-05 杭州百诚医药科技股份有限公司 一种嘧啶胺类化合物及应用
CN113880809B (zh) * 2020-07-03 2022-10-18 盛世泰科生物医药技术(苏州)有限公司 一种嘧啶类衍生物及其制备方法和应用
TW202221000A (zh) * 2020-09-30 2022-06-01 英屬開曼群島商百濟神州有限公司 用於降解egfr之雙功能化合物及相關使用方法
CN113149978A (zh) * 2021-02-04 2021-07-23 中国药科大学 一种中间体化合物、制备方法及其用途
CN113387935B (zh) * 2021-07-23 2022-06-10 苏州雅深智慧科技有限公司 抑制三突变表皮生长因子受体酪氨酸激酶的化合物及用途
WO2023208130A1 (zh) * 2022-04-29 2023-11-02 江苏亚虹医药科技股份有限公司 嘧啶类化合物、其制备方法及其医药用途
TW202406912A (zh) * 2022-08-01 2024-02-16 大陸商江蘇恒瑞醫藥股份有限公司 稠三環類衍生物的晶型及製備方法
CN117645604A (zh) * 2022-09-05 2024-03-05 浙江同源康医药股份有限公司 用作cdk4激酶抑制剂的化合物及其应用
CN115536699B (zh) * 2022-12-01 2023-05-23 北京鑫开元医药科技有限公司 一种新型EGFR-TKIs、制备方法、药物组合物及其应用

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004074262A1 (en) * 2003-02-20 2004-09-02 Tibotec Pharmaceuticals Ltd. Hiv replication inhibiting pyrimidines and triazines
WO2009158571A1 (en) * 2008-06-27 2009-12-30 Avila Therapeutics And Uses Thereof Heteroaryl compounds and uses thereof
WO2010138578A1 (en) * 2009-05-27 2010-12-02 Abbott Laboratories Pyrimidine inhibitors of kinase activity
CN104803925A (zh) * 2015-04-16 2015-07-29 温州医科大学 一类以 fgfr 为靶点的 2,4,5-三取代嘧啶类化合物及其制备方法和用途
WO2015127872A1 (zh) * 2014-02-25 2015-09-03 上海海雁医药科技有限公司 2,4-二取代苯-1,5-二胺衍生物及其应用以及由其制备的药物组合物和药用组合物
WO2015170266A1 (en) * 2014-05-07 2015-11-12 Lupin Limited Substituted pyrimidine compounds as btk inhibitors
CN105085489A (zh) * 2014-11-05 2015-11-25 上海页岩科技有限公司 嘧啶或吡啶类化合物、其制备方法和医药用途
WO2015195228A1 (en) * 2014-06-19 2015-12-23 Ariad Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
WO2016022460A1 (en) * 2014-08-03 2016-02-11 H. Lee Moffitt Cancer Center And Research Institute, Inc. Potent dual brd4-kinase inhibitors as cancer therapeutics
WO2016054987A1 (zh) * 2014-10-11 2016-04-14 上海翰森生物医药科技有限公司 Egfr抑制剂及其制备和应用
WO2016173438A1 (zh) * 2015-04-29 2016-11-03 南京明德新药研发股份有限公司 稠环或三环芳基嘧啶化合物用作激酶抑制剂
CN106749267A (zh) * 2015-11-23 2017-05-31 南京圣和药业股份有限公司 新的表皮生长因子受体抑制剂及其应用
CN106749193A (zh) * 2015-11-23 2017-05-31 南京圣和药业股份有限公司 吲唑取代的表皮生长因子受体抑制剂及其应用
CN106831730A (zh) * 2017-01-11 2017-06-13 温州医科大学 一种取代的二氨基嘧啶类化合物及其在制备抗恶性肿瘤药物中的用途

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2292753T4 (es) * 2001-03-29 2009-02-16 Vertex Pharmaceuticals Incorporated Inhibidores de quinasas n-terminales c-jun (jnk) y otras proteina quinasas.
WO2008124085A2 (en) * 2007-04-03 2008-10-16 Exelixis, Inc. Methods of using combinations of mek and jak-2 inhibitors
PT2200436E (pt) * 2007-09-04 2015-04-29 Scripps Research Inst Pirimidinilaminas substituídas como inibidoras da proteína quinase
WO2009029998A1 (en) * 2007-09-06 2009-03-12 Cytopia Research Pty Ltd Retrometabolic compounds
EP2440559B1 (en) 2009-05-05 2018-01-10 Dana-Farber Cancer Institute, Inc. Egfr inhibitors and methods of treating disorders
WO2015188747A1 (zh) * 2014-06-12 2015-12-17 南京圣和药业股份有限公司 作为egfr抑制剂的苯基取代的三嗪类化合物及其应用
CN105315259B (zh) * 2014-07-29 2018-03-09 上海艾力斯医药科技有限公司 吡啶胺基嘧啶衍生物、其制备方法及应用
TWI618704B (zh) * 2014-12-11 2018-03-21 貝達醫藥公司 作為egfr調節劑之經取代之2-苯胺嘧啶衍生物
CN104892580B (zh) * 2015-06-17 2018-01-26 镇江圣安医药有限公司 一种新型苯并咪唑‑嘧啶胺衍生物、及其应用
SG10201912959QA (en) 2015-10-21 2020-02-27 Otsuka Pharma Co Ltd Benzolactam compounds as protein kinase inhibitors
US10435388B2 (en) * 2016-01-07 2019-10-08 Cs Pharmatech Limited Selective inhibitors of clinically important mutants of the EGFR tyrosine kinase
DK3405196T3 (en) 2016-01-22 2020-03-09 Janssen Pharmaceutica Nv Nye substituerede cyanoindolinderivater som nik-inhibitorer
WO2018019204A1 (zh) 2016-07-26 2018-02-01 深圳市塔吉瑞生物医药有限公司 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物
CN110066272B (zh) 2018-05-29 2022-09-13 深圳市塔吉瑞生物医药有限公司 取代的苯并[d]咪唑类化合物及其药物组合物

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004074262A1 (en) * 2003-02-20 2004-09-02 Tibotec Pharmaceuticals Ltd. Hiv replication inhibiting pyrimidines and triazines
WO2009158571A1 (en) * 2008-06-27 2009-12-30 Avila Therapeutics And Uses Thereof Heteroaryl compounds and uses thereof
WO2010138578A1 (en) * 2009-05-27 2010-12-02 Abbott Laboratories Pyrimidine inhibitors of kinase activity
WO2015127872A1 (zh) * 2014-02-25 2015-09-03 上海海雁医药科技有限公司 2,4-二取代苯-1,5-二胺衍生物及其应用以及由其制备的药物组合物和药用组合物
WO2015170266A1 (en) * 2014-05-07 2015-11-12 Lupin Limited Substituted pyrimidine compounds as btk inhibitors
WO2015195228A1 (en) * 2014-06-19 2015-12-23 Ariad Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
WO2016022460A1 (en) * 2014-08-03 2016-02-11 H. Lee Moffitt Cancer Center And Research Institute, Inc. Potent dual brd4-kinase inhibitors as cancer therapeutics
WO2016054987A1 (zh) * 2014-10-11 2016-04-14 上海翰森生物医药科技有限公司 Egfr抑制剂及其制备和应用
CN105085489A (zh) * 2014-11-05 2015-11-25 上海页岩科技有限公司 嘧啶或吡啶类化合物、其制备方法和医药用途
CN104803925A (zh) * 2015-04-16 2015-07-29 温州医科大学 一类以 fgfr 为靶点的 2,4,5-三取代嘧啶类化合物及其制备方法和用途
WO2016173438A1 (zh) * 2015-04-29 2016-11-03 南京明德新药研发股份有限公司 稠环或三环芳基嘧啶化合物用作激酶抑制剂
CN106749267A (zh) * 2015-11-23 2017-05-31 南京圣和药业股份有限公司 新的表皮生长因子受体抑制剂及其应用
CN106749193A (zh) * 2015-11-23 2017-05-31 南京圣和药业股份有限公司 吲唑取代的表皮生长因子受体抑制剂及其应用
CN106831730A (zh) * 2017-01-11 2017-06-13 温州医科大学 一种取代的二氨基嘧啶类化合物及其在制备抗恶性肿瘤药物中的用途

Cited By (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11939321B2 (en) 2015-10-21 2024-03-26 Otsuka Pharmaceutical Co., Ltd. Benzolactam compounds as protein kinase inhibitors
US10457669B2 (en) 2015-10-21 2019-10-29 Otsuka Pharmaceutical Co., Ltd. Benzolactam compounds as protein kinase inhibitors
US11001575B1 (en) 2015-10-21 2021-05-11 Otsuka Pharmaceutical Co., Ltd. Benzolactam compounds as protein kinase inhibitors
US10435388B2 (en) 2016-01-07 2019-10-08 Cs Pharmatech Limited Selective inhibitors of clinically important mutants of the EGFR tyrosine kinase
CN113382991B (zh) * 2016-07-26 2022-12-27 深圳市塔吉瑞生物医药有限公司 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物
WO2020151742A1 (en) * 2016-07-26 2020-07-30 Shenzhen Targetrx, Inc. Amino pyrimidine compound for inhibiting protein tyrosine kinase activity
CN113382991A (zh) * 2016-07-26 2021-09-10 深圳市塔吉瑞生物医药有限公司 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物
US11111233B2 (en) 2016-07-26 2021-09-07 Shenzhen Targetrx, Inc. Amino pyrimidine compound for inhibiting protein tyrosine kinase activity
US11795166B2 (en) 2016-09-09 2023-10-24 Incyte Corporation Pyrazolopyridine compounds and uses thereof
US11891388B2 (en) 2016-09-09 2024-02-06 Incyte Corporation Pyrazolopyridine compounds and uses thereof
US11142518B2 (en) 2017-04-20 2021-10-12 Otsuka Pharmaceutical Co., Ltd. 6-pyrimidin-isoindole derivative as ERK1/2 inhibitor
EP3640248A4 (en) * 2017-06-13 2021-03-31 Beijing Adamadle Biotechnology Limited Liability Company AMINOPYRIMIDINE COMPOUND, PROCESS FOR PREPARATION AND USE
US11833156B2 (en) 2017-09-22 2023-12-05 Jubilant Epipad LLC Heterocyclic compounds as pad inhibitors
US11426412B2 (en) 2017-10-18 2022-08-30 Jubilant Epipad LLC Imidazo-pyridine compounds as PAD inhibitors
US11629135B2 (en) 2017-11-06 2023-04-18 Jubilant Prodell Llc Pyrimidine derivatives as inhibitors of PD1/PD-L1 activation
US11459338B2 (en) 2017-11-24 2022-10-04 Jubilant Episcribe Llc Heterocyclic compounds as PRMT5 inhibitors
CN109851638A (zh) * 2018-02-07 2019-06-07 深圳市塔吉瑞生物医药有限公司 取代的二氨基嘧啶化合物
CN110156754A (zh) * 2018-02-10 2019-08-23 杭州百诚医药科技股份有限公司 一种三取代嘧啶衍生物对蛋白激酶的抑制作用
CN110156754B (zh) * 2018-02-10 2023-10-03 杭州百诚医药科技股份有限公司 一种三取代嘧啶衍生物对蛋白激酶的抑制作用
US11731958B2 (en) 2018-02-20 2023-08-22 Incyte Corporation Carboxamide compounds and uses thereof
US11529341B2 (en) 2018-03-13 2022-12-20 Jubilant Prodel LLC Bicyclic compounds as inhibitors of PD1/PD-L1 interaction/activation
CN110066272B (zh) * 2018-05-29 2022-09-13 深圳市塔吉瑞生物医药有限公司 取代的苯并[d]咪唑类化合物及其药物组合物
WO2019228330A1 (zh) * 2018-05-29 2019-12-05 深圳市塔吉瑞生物医药有限公司 取代的苯并[d]咪唑类化合物及其药物组合物
CN110066272A (zh) * 2018-05-29 2019-07-30 深圳市塔吉瑞生物医药有限公司 取代的苯并[d]咪唑类化合物及其药物组合物
CN110698461B (zh) * 2018-07-09 2024-04-05 上海翰森生物医药科技有限公司 第三代egfr抑制剂的制备方法
CN110698461A (zh) * 2018-07-09 2020-01-17 上海翰森生物医药科技有限公司 第三代egfr抑制剂的制备方法
US11866426B2 (en) 2018-08-08 2024-01-09 Incyte Corporation Benzothiazole compounds and uses thereof
JP2022515196A (ja) * 2018-12-21 2022-02-17 深▲チェン▼市塔吉瑞生物医薬有限公司 プロテインキナーゼ活性を阻害するためのアミノピリミジン系化合物
JP7240032B2 (ja) 2018-12-21 2023-03-15 深▲チェン▼市塔吉瑞生物医薬有限公司 プロテインキナーゼ活性を阻害するためのアミノピリミジン系化合物
CN111377907B (zh) * 2018-12-26 2023-03-28 杭州百新生物医药科技有限公司 一种多取代苯氨基嘧啶衍生物及其制备方法和用途
WO2020135507A1 (zh) * 2018-12-26 2020-07-02 杭州百新生物医药科技有限公司 一种多取代苯氨基嘧啶衍生物及其制备方法和用途
CN111377907A (zh) * 2018-12-26 2020-07-07 杭州百新生物医药科技有限公司 一种多取代苯氨基嘧啶衍生物及其制备方法和用途
AU2020255100C1 (en) * 2019-03-29 2023-02-23 Shenzhen Forward Pharmaceuticals Co., Ltd. N-heteroaromatic amide derivatives for treatment of cancer
AU2020255100B2 (en) * 2019-03-29 2022-11-24 Shenzhen Forward Pharmaceuticals Co., Ltd. N-heteroaromatic amide derivatives for treatment of cancer
AU2020255100A8 (en) * 2019-03-29 2021-12-02 Shenzhen Forward Pharmaceuticals Co., Ltd. N-heteroaromatic amide derivatives for treatment of cancer
WO2021000912A1 (zh) * 2019-07-04 2021-01-07 微境生物医药科技(上海)有限公司 一类抑制egfr激酶的化合物及其制备方法和用途
CN114026090B (zh) * 2019-07-04 2023-01-06 微境生物医药科技(上海)有限公司 一类抑制egfr激酶的化合物及其制备方法和用途
US11993598B2 (en) 2019-07-04 2024-05-28 Wigen Biomedicine Technology (shanghai) Co., Ltd. Compounds for inhibiting EGFR kinase, preparation methods and uses thereof
CN114026090A (zh) * 2019-07-04 2022-02-08 微境生物医药科技(上海)有限公司 一类抑制egfr激酶的化合物及其制备方法和用途
US11787784B2 (en) 2019-08-06 2023-10-17 Incyte Corporation Solid forms of an HPK1 inhibitor
WO2021185297A1 (zh) * 2020-03-18 2021-09-23 南京药石科技股份有限公司 Egfr酪氨酸激酶抑制剂及其用途
CN113880813A (zh) * 2020-07-01 2022-01-04 杭州百诚医药科技股份有限公司 一种2-氨基嘧啶类杂环化合物及应用
CN113880813B (zh) * 2020-07-01 2024-05-28 杭州百诚医药科技股份有限公司 一种2-氨基嘧啶类杂环化合物及应用
WO2022063140A1 (en) * 2020-09-23 2022-03-31 Shenzhen Ionova Life Science Co., Ltd. Pyrimidine and pyridine derivatives as hpk1 modulator and methods using same
WO2022166799A1 (zh) * 2021-02-03 2022-08-11 上海拓界生物医药科技有限公司 稠三环类细胞周期蛋白-依赖性激酶抑制剂及其制备方法和医药用途
CN115650974A (zh) * 2022-08-04 2023-01-31 天津大学 N-[5-(嘧啶-2-氨基)-2,4-二取代苯基]-顺式戊二烯酰胺衍生物及应用

Also Published As

Publication number Publication date
JP2020183432A (ja) 2020-11-12
JP7156715B2 (ja) 2022-10-19
EP3492462B1 (en) 2023-08-30
US20190152954A1 (en) 2019-05-23
JP2019522011A (ja) 2019-08-08
CN110818690B (zh) 2021-08-10
US11111233B2 (en) 2021-09-07
EP3492462A1 (en) 2019-06-05
WO2020151742A1 (en) 2020-07-30
EP3492462A4 (en) 2019-07-10
CN113382991A (zh) 2021-09-10
CN108602802A (zh) 2018-09-28
CN113382991B (zh) 2022-12-27
CN110818690A (zh) 2020-02-21
CN108602802B (zh) 2020-01-21
JP6740452B2 (ja) 2020-08-12

Similar Documents

Publication Publication Date Title
CN113382991B (zh) 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物
AU2021200951B2 (en) Benzolactam compounds as protein kinase inhibitors
CN107660205B (zh) 作为lsd1抑制剂的杂环化合物
TWI453207B (zh) 胺基***并吡啶,其組合物及使用其之治療方法
BR112016011851B1 (pt) Derivados de imidazol e pirazol fundidos como moduladores de atividade de tnf
BR112016013632B1 (pt) Carboxamidas, seus usos, seus intermediários e seu processo de preparação, e medicamento
BR122019020716B1 (pt) heterociclilaminas como inibidores de pi3k e composição farmacêutica que as compreende
JP7384536B2 (ja) キナゾリン化合物並びにその調製方法、使用及び医薬組成物
JP6515175B2 (ja) 芳香族ヘテロ環誘導体及びその医薬的適用
MX2014009180A (es) Derivados de isoquinolina y naftiridina.
IL311024A (en) Modified tricyclic compounds as PARP inhibitors and their use
WO2017190637A1 (zh) 用于抑制蛋白酪氨酸激酶活性的稠合嘧啶类化合物
TW202204351A (zh) 具有大環結構的化合物及其用途
CA3117512A1 (en) 5-azaindazole derivatives as adenosine receptor antagonists
WO2019120094A1 (zh) 用于抑制激酶活性的芳基磷氧化物
EP3233839B1 (en) Heterocyclyl linked imidazopyridazine derivatives as pi3kbeta inhibitors
WO2017097113A1 (zh) 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物
WO2019228330A1 (zh) 取代的苯并[d]咪唑类化合物及其药物组合物
CN114599655B (zh) 咪唑烷酮类化合物及其制备方法与应用
EP4310081A1 (en) Ctla-4 small molecule degradation agent and application thereof
WO2023245329A1 (en) Multi-kinase inhibitors, compositions thereof, and methods of using the same
WO2023051495A1 (zh) 异喹啉酮类及喹唑啉酮类化合物及其组合物和用途
WO2022214008A1 (zh) 高活性hpk1激酶抑制剂

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: 17833507

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2019503547

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: 2017833507

Country of ref document: EP

Effective date: 20190226