WO2018040885A1 - 成纤维细胞生长因子受体抑制剂及其用途 - Google Patents

成纤维细胞生长因子受体抑制剂及其用途 Download PDF

Info

Publication number
WO2018040885A1
WO2018040885A1 PCT/CN2017/096848 CN2017096848W WO2018040885A1 WO 2018040885 A1 WO2018040885 A1 WO 2018040885A1 CN 2017096848 W CN2017096848 W CN 2017096848W WO 2018040885 A1 WO2018040885 A1 WO 2018040885A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
amino
alkoxy
cancer
Prior art date
Application number
PCT/CN2017/096848
Other languages
English (en)
French (fr)
Inventor
吴永谦
李进
Original Assignee
南京药捷安康生物科技有限公司
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 南京药捷安康生物科技有限公司 filed Critical 南京药捷安康生物科技有限公司
Priority to KR1020197007909A priority Critical patent/KR102270620B1/ko
Priority to US16/328,817 priority patent/US10744139B2/en
Priority to RU2019107910A priority patent/RU2732127C1/ru
Priority to AU2017317864A priority patent/AU2017317864B2/en
Priority to ES17845187T priority patent/ES2859637T3/es
Priority to EP17845187.8A priority patent/EP3508482B1/en
Priority to JP2019511715A priority patent/JP6751977B2/ja
Publication of WO2018040885A1 publication Critical patent/WO2018040885A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • 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
    • C07D403/12Heterocyclic 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 linked by a chain containing hetero atoms as chain links
    • 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
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 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
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Definitions

  • the invention belongs to the technical field of medicine and relates to an irreversible inhibitor of fibroblast growth factor receptor (FGFR), or a pharmaceutically acceptable salt thereof, a stereoisomer thereof and use thereof.
  • FGFR fibroblast growth factor receptor
  • Tyrosine kinase receptors play an important role in tumor angiogenesis, tumor cell proliferation, migration and infiltration. More than 100 tyrosine kinase inhibitor drugs have been marketed or entered clinical trials. These small molecule tyrosine kinase inhibitors (TKI) play a role in reversible inhibition, which brings some disadvantages: 1 the selectivity is not good enough, 2 the efficacy is not strong enough and lasting, 3 It is easy to cause drug resistance. Therefore, scientists are encouraged to focus their research on the development of irreversible TKI.
  • TKI small tyrosine kinase inhibitors
  • the irreversible TKI is usually modeled by the backbone structure of the reversible TKI, and an electrophilic functional group is attached at a suitable position, and the electrophilic functional group can be associated with a cysteine residue near the ATP binding domain of the tyrosine kinase. (Electron-rich nucleophilic structure) undergoes an electrophilic reaction to form a covalent bond, thereby irreversibly inhibiting kinase activity.
  • irreversible TKI has many unique advantages: 1 irreversible TKI works in a permanent inactivation manner, this way of inhibiting enzyme activity makes its effect stronger and longer, even drug molecules It is completely removed from the circulatory system and its efficacy is maintained.
  • Fibroblast growth factor receptor belongs to An important member of the tyrosine kinase receptor family, FGFR contains four members, namely FGFR-1, FGFR-2, FGFR-3 and FGFR-4. They are mostly single-chain glycoprotein molecules with molecular masses ranging from 110 to 150 kd. The structure is divided into extracellular regions, transmembrane regions and intracellular regions.
  • FGFR binds to its ligand fibroblast growth factor (FGF), which dimerizes FGFR and phosphorylates itself, activating downstream signaling pathways such as JAK/STAT pathway, phospholipase The C pathway, phosphatidylinositol-3-kinase PI3K and MAPK signaling pathways play an important role in tumor growth and angiogenesis.
  • FGF ligand fibroblast growth factor
  • JAK/STAT pathway phospholipase
  • PI3K phosphatidylinositol-3-kinase PI3K and MAPK signaling pathways play an important role in tumor growth and angiogenesis.
  • Abnormally high expression of FGFR is closely related to the development of various tumors such as lung cancer, liver cancer, glioma, rhabdomyosarcoma and melanoma.
  • pan-FGFR inhibitors which have potent inhibitory activity against pan-FGFR and which offer possibilities for the treatment of diseases mediated by pan-FGFR abnormalities.
  • the invention also provides the use of the above FGFR inhibitors.
  • R 1 and R 2 are each independently selected from the group consisting of hydrogen, hydroxy, amino, cyano, nitro, halogen atom, carboxyl group, C 1-6 alkyl group, C 1-6 alkoxy group, C 1-6 alkylamino group, (C 1-6 alkyl) 2 amino, halo C 1-6 alkyl, halo C 1-6 alkoxy, C 2-8 alkenyl, C 2-8 alkynyl, C 1-6 alkyl a sulfonyl group, a C 1-6 alkylcarbonylamino group, a C 1-6 alkyl-substituted 3-8-membered cycloalkyl group, a C 1-6 alkyl-substituted 3-8-membered heterocyclic group, optionally, R 1 and R 2 may form a 3- to 8-membered cycloalkyl group, a 3- to 8-membered heterocyclic group, a 6- to 14-membered aryl group or
  • R 3 and R 4 are each independently selected from the group consisting of hydrogen, hydroxy, amino, cyano, nitro, halogen atom, carboxyl group, C 1-6 alkyl group, C 1-6 alkoxy group, C 1-6 alkylamino group, (C 1-6 alkyl) 2 amino, halo C 1-6 alkyl, halo C 1-6 alkoxy, C 2-8 alkenyl, C 2-8 alkynyl, C 1-6 alkyl Sulfonyl group, C 1-6 alkylcarbonylamino group, C 1-6 alkylaminocarbonyl group, 3- to 8-membered cycloalkyl group, 3- to 8-membered heterocyclic group, 6 to 14-membered aryl group or 5- to 10-membered heteroaryl group a C 1-6 alkyl-substituted 3-8-membered cycloalkyl group, a C 1-6 alkyl-substituted 3-8-membered heterocyclic
  • Ar is selected from the group consisting of a 6 to 14 membered aromatic ring group or a 5 to 10 membered heteroaryl group containing 0 to 3 O, S and/or N atoms;
  • Ring A is selected from a 3- to 8-membered cycloalkyl group having from 0 to 3 O, S and/or N atoms, optionally substituted by 1 to 3 R 5 , a 3 to 8 membered heterocyclic group, and a 6 to 14 membered aromatic group.
  • a 5- to 10-membered heteroaryl group wherein the S atom in any ring can be optionally oxidized to S(O) or S(O) 2 , and the carbon atom in any ring can be optionally oxidized to C (O);
  • Ring B is selected from a 3- to 10-membered saturated or unsaturated heterocyclic group or a 5- to 6-membered N-containing heteroaryl group containing at least one N hetero atom, optionally substituted by 1 to 3 R 6 groups, and ring B
  • the upper N atom is directly bonded to the Warhead bond, wherein the S atom in any ring B can be optionally oxidized to S(O) or S(O) 2 , and any carbon atom in ring B can optionally be Oxidation to C(O);
  • X is selected from CR 7 and N;
  • R 5 , R 6 and R 7 are each independently selected from
  • n 1 , m 2 represents 1, 2 or 3, and m 1 and m 2 are added to be less than or equal to 5;
  • Warhead refers to a moiety that is capable of forming a covalent bond with a nucleophile.
  • R 1 is independently selected from the group consisting of hydrogen, halogen, and hydroxyl
  • R 2 is independently selected from the group consisting of hydrogen, halogen, hydroxy, C 1-4 alkyl, C 1-4 alkoxy, cyano, halo C 1-4 alkyl, halo C 1-4 alkoxy;
  • Ar is selected from the group consisting of a 6- to 14-membered aromatic ring group or a 5- to 6-membered heteroaryl group containing 0 to 3 O, S and/or N atoms;
  • n 1 , m 2 represents 1, 2 or 3, and m 1 and m 2 are added to be less than or equal to 5.
  • R 1 is independently selected from the group consisting of hydrogen, halogen, and hydroxyl
  • R 2 is independently selected from the group consisting of hydrogen, halogen, hydroxy, C 1-4 alkyl, C 1-4 alkoxy, cyano, halo C 1-4 alkyl, halo C 1-4 alkoxy;
  • R 3 and R 4 are each independently selected from the group consisting of hydrogen, hydroxy, amino, cyano, nitro, halogen atom, carboxyl group, C 1-4 alkyl group, C 1-4 alkoxy group, C 1-4 alkylamino group, (C 1-4 alkyl) 2 amino, halo C 1-4 alkyl, halo C 1-4 alkoxy, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 alkyl Sulfonyl, C 1-4 alkylcarbonylamino;
  • Ar is a phenyl group
  • Ring A is selected from phenyl optionally substituted by 1 to 3 R 5 ;
  • ring B is selected from 4 to 10 membered saturated or unsaturated, optionally substituted by 1 to 3 R 6 and containing at least 1 N hetero atom. a heterocyclic group, and the N atom on ring B is directly bonded to the Warhead bond;
  • X is selected from CR 7 and N;
  • R 5 and R 7 are each independently selected from the group consisting of hydrogen, hydroxy, amino, carboxy, cyano, nitro, halogen atom, C 1-4 alkyl, C 1-4 alkoxy;
  • R 6 is selected from
  • n 1 , m 2 represents 1, 2 or 3, and m 1 and m 2 are added to be less than or equal to 5;
  • Warhead refers to a moiety that is capable of forming a covalent bond with a nucleophile.
  • the compound of the formula (I), or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, according to any one of the schemes 1 to 3, has a structure as shown in the formula (II):
  • R 4 is selected from hydrogen or C 1-4 alkyl
  • Ring A is a phenyl group
  • Ring B is selected from a 4- to 6-membered saturated or unsaturated monoheterocyclic group or a 6- to 10-membered saturated or unsaturated fused heterocyclic group optionally substituted by 1 to 3 R 6 and having at least one N hetero atom. And, the N atom on ring B is directly connected to the Warhead bond;
  • X is N
  • R 6 is selected from
  • amino-carbonyl cyano-carbonyl, C 1-4 alkyl-carbonyl, C 1-4 alkylamino-carbonyl, (C 1-4 alkyl) 2 amino-carbonyl, C 1-4 alkoxy a carbonyl group, a 3 to 8 membered cycloalkyl-carbonyl group, a 3 to 8 membered heterocyclyl-carbonyl group;
  • Warhead is selected from
  • Z refers to the leaving group or the activated hydroxyl moiety
  • R 11 , R 12 , R 13 are independently selected from the group consisting of hydrogen, halogen, cyano, C 1-4 alkyl optionally substituted by a substituent, halogenated C 1-4 alkyl, 3- to 8-membered cycloalkyl, a 3- to 8-membered heterocyclic group, a 5- to 8-membered aryl group, and a 5- to 10-membered heteroaryl group, the substituent being selected from the group consisting of a hydroxyl group, an amino group, a carboxyl group, a cyano group, a nitro group, a halogen group, and a C 1-4 alkyl group.
  • R 11 , R 12 and R 13 are preferably hydrogen.
  • Warhead is directly connected to the N atom on ring B as follows:
  • ring B is selected from the group consisting of:
  • Warhead is selected from the following structures:
  • Z refers to the leaving group or the activated hydroxyl moiety
  • R 11 , R 12 and R 13 are each independently selected from hydrogen or C 1-4 alkyl.
  • Ring B is selected from a 5- to 6-membered saturated monoheterocyclic group containing at least one N hetero atom optionally substituted by 1 to 3 R 6 , and the N atom on ring B is directly bonded to the Warhead bond.
  • the compound of the formula 8, or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, has the structure shown in the formula (III):
  • R 4 is selected from hydrogen or C 1-4 alkyl
  • X is N
  • R 6 is selected from
  • amino-carbonyl cyano-carbonyl, C 1-4 alkyl-carbonyl, C 1-4 alkylamino-carbonyl, (C 1-4 alkyl) 2 amino-carbonyl, C 1-4 alkoxy a carbonyl group, a 3 to 8 membered cycloalkyl-carbonyl group, a 3 to 8 membered heterocyclyl-carbonyl group;
  • n is an integer from 1 to 3;
  • Warhead is selected from
  • Z refers to a leaving group or an activated hydroxyl moiety
  • R 11 , R 12 , R 13 are independently selected from the group consisting of hydrogen, halogen, cyano, C 1-4 alkyl optionally substituted by a substituent, halogenated C 1-4 alkyl, 3- to 8-membered cycloalkyl, a 3- to 8-membered heterocyclic group, a 5- to 8-membered aryl group, and a 5- to 10-membered heteroaryl group, the substituent being selected from the group consisting of a hydroxyl group, an amino group, a carboxyl group, a cyano group, a nitro group, a halogen group, and a C 1-4 alkyl group.
  • R 11 , R 12 and R 13 are preferably hydrogen.
  • R 6 is selected from
  • amino-carbonyl cyano-carbonyl, C 1-4 alkyl-carbonyl, C 1-4 alkylamino-carbonyl, (C 1-4 alkyl) 2 amino-carbonyl, C 1-4 alkoxy a carbonyl group, a 3 to 8 membered cycloalkyl-carbonyl group, a 3 to 8 membered heterocyclyl-carbonyl group;
  • Warhead is selected from
  • R 11 , R 12 , R 13 are independently selected from hydrogen or C 1-4 alkyl, preferably
  • the compound of claim 10 may be selected from the group consisting of the following structures:
  • the compound of the present invention a pharmaceutically acceptable salt thereof or a stereoisomer thereof is:
  • the invention also claims a pharmaceutical formulation of any of the compounds of the invention, or a pharmaceutically acceptable salt or stereoisomer thereof, which formulation further comprises one or more pharmaceutically acceptable carriers.
  • the pharmaceutical carrier of the present invention may be one or more solid or liquid filler or gel materials suitable for human use.
  • the pharmaceutical carrier preferably has sufficient purity and sufficiently low toxicity and is compatible with the active ingredients of the present invention and does not significantly reduce the efficacy of the active ingredient.
  • the pharmaceutically acceptable carrier can be a filler, a binder, a disintegrant, a lubricant, an aqueous solvent or a non-aqueous solvent, and the like.
  • the pharmaceutical preparation of the present invention can be formulated into any pharmaceutically acceptable dosage form, and administered to any patient in need of such treatment by any suitable administration means, for example, by oral, parenteral, rectal or pulmonary administration. Or subject.
  • oral administration it can be formulated into tablets, capsules, pills, granules and the like.
  • parenteral administration it can be prepared as an injection solution, a sterile powder for injection, or the like.
  • the pharmaceutical preparation of the present invention further comprises one or more second therapeutically active agents, wherein the second therapeutically active agent is an antimetabolite, a growth factor inhibitor, a mitotic inhibitor, and an antitumor Hormones, alkylating agents, metals, topoisomerase inhibitors, hormonal drugs, immunomodulators, tumor suppressor genes, cancer vaccines, immunological checkpoints or antibodies and small molecule drugs related to tumor immunotherapy.
  • the second therapeutically active agent is an antimetabolite, a growth factor inhibitor, a mitotic inhibitor, and an antitumor Hormones, alkylating agents, metals, topoisomerase inhibitors, hormonal drugs, immunomodulators, tumor suppressor genes, cancer vaccines, immunological checkpoints or antibodies and small molecule drugs related to tumor immunotherapy.
  • the invention also claims the use of any of the compounds of the invention, or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, or a pharmaceutical formulation according to the invention, for the manufacture of a medicament for the treatment of a disease mediated by FGF/FGFR abnormalities.
  • the FGF/FGFR abnormality-mediated disease is cancer; the cancer includes lung cancer, squamous cell carcinoma, bladder cancer, gastric cancer, ovarian cancer, peritoneal cancer, breast cancer, breast ductal carcinoma, head and neck cancer, Endometrial cancer, uterine body cancer, rectal cancer, liver cancer, kidney cancer, renal pelvic cancer, esophageal cancer, esophageal adenocarcinoma, glioma, prostate cancer, thyroid cancer, female reproductive system cancer, carcinoma in situ, lymphoma, Neurofibromatosis, bone cancer, skin cancer, brain cancer, colon cancer, testicular cancer, gastrointestinal stromal tumor, oral cancer, pharyngeal cancer, multiple myeloma, leukemia, non-Hodgkin's lymphoma, large intestine villus Tumor, melanoma, cell tumor and sarcoma, myelodysplastic syndrome.
  • the invention also provides a method for treating a disease mediated by FGF/FGFR abnormality, the method comprising administering to a subject in need thereof a compound of any of the invention, or a pharmaceutically acceptable salt, stereoisomer thereof or Pharmaceutical preparation according to the invention.
  • the FGF/FGFR abnormality-mediated disease is cancer; the cancer includes lung cancer, squamous cell carcinoma, bladder cancer, gastric cancer, ovarian cancer, peritoneal cancer, breast cancer, breast ductal carcinoma, head and neck cancer, Endometrial cancer, uterine body cancer, rectal cancer, liver cancer, kidney cancer, renal pelvic cancer, esophageal cancer, esophageal adenocarcinoma, glioma, prostate cancer, thyroid cancer, female reproductive system cancer, carcinoma in situ, lymphoma, Neurofibromatosis, bone cancer, skin cancer, brain cancer, colon cancer, testicular cancer, gastrointestinal stromal tumor, oral cancer, pharyngeal cancer, multiple myeloma, leukemia, non-Hodgkin's lymphoma, large intestine villus Tumor, melanoma, cell tumor and sarcoma, myelodysplastic syndrome.
  • the invention further relates to any of the compounds of the invention, or a pharmaceutically acceptable salt, stereoisomer or pharmaceutical formulation thereof according to the invention for use as a medicament.
  • halogen as used in the present invention means fluorine, chlorine, bromine, iodine or the like, preferably a fluorine atom or a chlorine atom.
  • oxo means that any C in the substituent structure may be replaced by "-C(O)-"; if a hetero atom is contained, the hetero atom may form an oxide, such as Can be Alternatively, S can be oxidized to S(O) or S(O) 2 .
  • halo means that any of the hydrogen atoms in the substituent may be substituted by one or more of the same or different halogen atoms.
  • Halogen is as defined above.
  • C 1-6 alkyl group as used in the present invention means a straight or branched alkyl group derived from a hydrocarbon moiety having 1 to 6 carbon atoms, which is removed by a hydrogen atom, such as methyl, ethyl or n-propyl groups.
  • C 2-8 alkenyl group as used in the present invention means an olefin moiety having 2 to 8 carbon atoms containing a carbon-carbon double bond, and a linear or branched or cyclic alkene group derived from a hydrogen atom, such as a vinyl group.
  • C 2-8 alkynyl group as used in the present invention means an alkyne moiety having 2 to 8 carbon atoms containing a carbon-carbon oxime bond, and a straight or branched alkyne group derived by removing one hydrogen atom, such as ethynyl group, C. Alkynyl, 2-butynyl, 2-pentynyl, 3-pentynyl, 4-methyl-2-pentynyl, 2-hexynyl, 3-hexynyl, and the like.
  • C 1-6 alkylamino group "(C 1-6 alkyl) 2 amino group”, “C 1-6 alkylcarbonylamino group”, and “C 1-6 alkylsulfonylamino group” according to the present invention "C 1-6 alkylaminocarbonyl”", “(C 1-6 alkyl) 2 amino-carbonyl”, “C 1-6 alkoxy-carbonyl”, “C 1-6 alkylsulfonyl””””,”C 1-6 alkylthio", "C 1-6 alkyl-carbonyl", “3- to 8-membered cycloalkyl-carbonyl", “3- to 8-membered heterocyclyl-carbonyl”, respectively C 1-6 alkyl-NH-, (C 1-6 alkyl)(C 1-6 alkyl)N-, C 1-6 alkyl-C(O)-NH-, C 1-6 alkyl -S(O) 2 -NH 2 -, C 1-6 alkyl-NH-C
  • C 1-6 alkoxy refers to the present invention as hereinbefore defined "C 1-6 alkyl” group linked to the parent molecule through an oxygen atom, i.e., "C 1-6 alkyl -O- "Groups such as methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, n-pentyloxy, neopentyloxy and n-hexyloxy.
  • the "C 1-4 alkoxy group” means the above-mentioned example having 1 to 4 carbon atoms, that is, a "C 1-4 alkyl-O-" group.
  • the "fused ring” as used in the present invention means a polycyclic ring structure formed by joining two, two or more cyclic structures in a snail, a snail, or a bridge.
  • the parallel ring refers to a fused ring structure formed by two or more ring structures sharing two adjacent ring atoms with each other (ie, sharing one bond).
  • the bridged ring refers to a fused ring structure formed by two or more ring-shaped structures sharing two non-adjacent ring atoms with each other.
  • the spiro ring refers to a fused ring structure formed by two or more ring structures sharing one ring atom with each other.
  • cycloalkyl refers to a monocyclic cycloalkyl, bicyclic cycloalkyl system or polycyclic cycloalkyl system (also known as a fused ring system).
  • the monocyclic system is a cyclic hydrocarbon group having 3 to 8 carbon atoms.
  • 3- to 8-membered cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutane, cyclopentyl, cyclohexane, cycloheptyl, cyclooctyl, and the like.
  • the fused ring cycloalkyl group includes a cyclocycloalkyl group, a bridged cycloalkyl group, a spirocycloalkyl group.
  • the cyclocycloalkyl group may be a 6-12 membered cyclocycloalkyl group, a 7-10 membered cyclocycloalkyl group, and representative examples thereof include, but are not limited to, bicyclo [3.1.1] heptane, bicyclo [2.2.1 Heptane, bicyclo [2.2.2] octane, bicyclo [3.2.2] decane, bicyclo [3.3.1] decane and bicyclo [4.2.1] decane.
  • the spiro group may be a 6-12 membered spiro group, a 7-11 membered spiro group, examples of which include, but are not limited to:
  • the bridged ring group may be a 6-12 membered bridged ring group and a 7-11 membered bridged ring group, and examples thereof include, but are not limited to:
  • heterocyclic group as used in the present invention means a non-aromatic cyclic group in which at least one ring carbon atom is replaced by a hetero atom selected from O, S, N, preferably 1 to 3 hetero atoms, including carbon. Atoms, nitrogen atoms and sulfur atoms can be replaced by oxo.
  • Heterocyclyl means a monocyclic heterocyclic ring, a bicyclic heterocyclic ring system or a polycyclic heterocyclic ring system (also known as a fused ring system), including saturated, partially saturated heterocyclic groups, but excluding aromatic rings.
  • the monoheterocyclic group may be a 3- to 8-membered heterocyclic group, a 3- to 8-membered saturated heterocyclic group, a 3- to 6-membered heterocyclic group, a 4- to 7-membered heterocyclic group, a 5- to 7-membered heterocyclic group, or a 5- to 5-membered heterocyclic group.
  • "3-8"-membered saturated heterocyclic group examples of which include, but are not limited to, aziridine groups, oxygen rings Propane, thietyl, azetidinyl, oxetanyl, thietane, tetrahydrofuranyl, tetrahydropyrrolyl, tetrahydrothiophenyl, imidazolidinyl, pyrazolidine Base, 1,2-oxazolidinyl, 1,3-oxazolidinyl, 1,2-thiazolidinyl, 1,3-thiazolidinyl, tetrahydro-2H-pyranyl, tetrahydro-2H- Thiantyl, piperidinyl, piperazinyl, morpholinyl
  • the fused heterocyclic ring includes a heterocyclic group, a spiroheterocyclic group, a bridged heterocyclic group, and may be saturated, partially saturated or unsaturated, but not aromatic.
  • the fused heterocyclic group is a monocyclic cycloalkyl group, a 5-6 membered monocyclic cycloalkenyl group, a 5-6 membered monocyclic heterocyclic group or a 5-6 membered monocyclic heteroaryl group fused to a benzene ring, 5-6 members.
  • a 5-6 membered monocyclic heterocyclic ring of the group is a monocyclic cycloalkyl group, a 5-6 membered monocyclic cycloalkenyl group, a 5-6 membered monocyclic heterocyclic group or a 5-6 membered monocyclic heteroaryl group fused to a benzene ring, 5-6 members.
  • the heterocyclic group may be 6-12 members and a cyclic group, a 7-10 membered ring group, a 6-10 membered ring group, a 6-12 membered saturated ring group, and representative examples include, but are not limited to: 3-azabicyclo[3.1.0]hexane, 3,6-diazabicyclo[3.2.0]heptyl, 3,8-diazabicyclo[4.2.0]octyl, 3, 7-diazabicyclo[4.2.0]octyl, octahydropyrrolo[3,4-c]pyrrolyl, octahydropyrrolo[3,4-b]pyrrolyl, octahydropyrrolo[3, 4-b][1,4]oxazinyl, octahydro-1H-pyrrolo[3,4-c]pyridyl, 2,3-dihydrobenzofuran-2-yl, 2,3-dihydro Benzofuran
  • the spiroheterocyclyl group may be a 6-12 membered spiroheterocyclyl group, a 7-11 membered spiroheterocyclyl group, a 6-12 membered saturated spirocyclic group, and examples thereof include, but are not limited to:
  • the bridged heterocyclic group may be a 6-12 membered bridged heterocyclic group, a 7-11 membered bridged heterocyclic group, and a 6-12 membered saturated bridged heterocyclic group, and examples thereof include, but are not limited to:
  • the "6- to 14-membered aryl group” as used in the present invention means a cyclic aromatic group having 6 to 14 carbon atoms, and includes a "6-8 membered monocyclic aryl group” such as a phenyl group; ⁇ 14-membered fused ring aryl group, such as pentylene, naphthalene, phenanthrene, and the like.
  • heteroaryl of the present invention may be a 5-10 membered heteroaryl group, and means an aromatic cyclic group in which at least one ring carbon atom is replaced by a hetero atom selected from O, S, N, preferably 1 to 3 heteroatoms, including carbon atoms, sulfur atoms by oxo, such as carbon atoms replaced by C (O), sulfur atoms replaced by S (O), S (O) 2 .
  • Heteroaryl groups include monoheteroaryl and fused heteroaryl.
  • the monoheteroaryl group may be a 5- to 7-membered heteroaryl group or a 5- to 6-membered heteroaryl group, and examples thereof include, but are not limited to, furyl, imidazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl , oxazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyrazolyl, pyrrolyl, tetrazolyl, thiadiazolyl, thienyl, triazolyl and triazinyl.
  • X is a 5- to 7-membered heteroaryl group or a 5- to 6-membered heteroaryl group, and examples thereof include, but are not limited to, furyl, imidazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl , oxazolyl
  • One or two groups are optionally substituted.
  • the fused heteroaryl group may be a 8-12 membered heteroaryl group, a 9-10 membered heteroaryl group, and examples include, but are not limited to, benzimidazolyl, benzofuranyl, benzothienyl, benzooxadiazolyl.
  • Benzothiadiazolyl benzothiazolyl, porphyrinyl, 5,6-dihydroquinolin-2-yl, 5,6-dihydroisoquinolin-1-yl, furopyridine Furopyridinyl, carbazolyl, fluorenyl, isoquinolyl, naphthyridinyl, fluorenyl, quinolyl, 5,6,7,8-tetrahydroquinolin-2-yl, 5,6, 7,8-tetrahydroquinolinyl, 5,6,7,8-tetrahydroquinolin-4-yl, 5,6,7,8-tetrahydroisoquinolin-1-yl, thienopyridyl ( Thienopyridinyl), 4,5,6,7-tetrahydro[c][1,2,5]oxadiazolyl and 6,7-dihydro-[c][1,2,5]oxadiazole- 4(5H) ketone group
  • the "pharmaceutically acceptable salt” as used in the present invention means a pharmaceutically acceptable acid or base addition salt or a solvate thereof.
  • Such pharmaceutically acceptable salts include salts of the following acids: hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, sulfurous acid, formic acid, toluenesulfonic acid, methanesulfonic acid, nitric acid, benzoic acid, citric acid, tartaric acid, maleic acid. Hydroiodic acid, alkanoic acid (such as acetic acid, HOOC-(CH 2 ) n-COOH (where n is 0 to 4)), and the like.
  • Salts of bases sodium salts, potassium salts, calcium salts, ammonium salts, and the like. Those skilled in the art are aware of a variety of non-toxic pharmaceutically acceptable addition salts.
  • the "stereoisomer" of the compound of the formula (I) of the present invention means that when the compound of the formula (I), (II), (III) has an asymmetric carbon atom, an enantiomer is produced; when the compound has carbon and carbon In the case of a double bond or a cyclic structure, a cis-trans isomer is produced; when a compound has a ketone or a oxime, a tautomer is produced, and all of the compounds of the formula (I), (II), (III) are enantiomerically Constructs, diastereomers, racemic isomers, cis-trans isomers, tautomers, geometric isomers, epimers, and mixtures thereof are included within the scope of the invention.
  • Warhead refers to a moiety capable of forming a covalent bond with a nucleophile.
  • a "nucleophile” is a substance that is directed to an electrophile to supply an electron pair to form a chemical bond in the reaction.
  • the pro-test nucleating agent can be an oxygen nucleophile, eg, water or a hydroxyl group; a nitrogen nucleophile, eg, an amine; or a sulfur nucleophile, eg, a thiol group, such as a cystine residue side A thiol group in the chain.
  • warhead refers to a moiety that is reversibly or irreversibly involved in the reaction of a donor (eg, a protein) with a substrate.
  • Warhead can, for example, form a covalent bond with a protein, or can form a stable transition state, or a reversible irreversible alkylating agent.
  • warhead can be a functional group on an inhibitor that can participate in a bond formation reaction, wherein a new covalent bond is formed between a portion of the warhead and a donor (eg, an amino acid residue of a protein).
  • Warhead is an electrophile and the "donor” is a nucleophile such as a side chain of a cysteine residue. Suitable for the warhead part include but are not limited to the following structure:
  • Z refers to a leaving group (such as a halogen) or an activated hydroxyl moiety (such as a triflate);
  • R 11 , R 12 , R 13 are independently selected from the group consisting of hydrogen, halogen, cyano, C 1-4 alkyl optionally substituted by a substituent, halogenated C 1-4 alkyl, 3- to 8-membered cycloalkyl, a 3- to 8-membered heterocyclic group, a 5- to 8-membered aryl group, and a 5- to 10-membered heteroaryl group, the substituent being selected from the group consisting of a hydroxyl group, an amino group, a carboxyl group, a cyano group, a nitro group, a halogen group, and a C 1-4 alkyl group.
  • R 11 , R 12 and R 13 are preferably hydrogen.
  • NMP N-methylpyrrolidone
  • DIPEA N,N-diisopropylethylamine
  • TLC thin layer chromatography
  • PE:EA petroleum ether Ethyl acetate
  • THF tetrahydrofuran
  • EA ethyl acetate
  • DCM: MeOH means dichloromethane: methanol
  • DCM means dichloro Methane
  • MTBE means methyl tert-butyl ether
  • TFAA means trifluoroacetic anhydride.
  • the synthetic route is as follows:
  • SM1 (5.00 g, 20.5 mmol) and SM2 (3.73 g, 20.5 mmol) were dissolved in tetrahydrofuran (30 ml), a solution of cesium carbonate (20.00 g, 61.5 mmol) in water (30 ml) was added, and a catalytic amount of Pd (PPh) was added. 3 ) Cl 2 , the resulting mixture was heated under reflux for 4 hours under a nitrogen atmosphere.
  • reaction solution was concentrated to dryness and evaporated with ethyl acetate.
  • organic phase was washed once with saturated sodium chloride and dried over anhydrous sodium sulfate. 1 (3.80 g, 62% yield) as a pale yellow solid.
  • SM3 (81 mg, 0.40 mmol) and I-2 (100 mg, 0.35 mmol) were added to N-methylpyrrolidone (3 ml), and N,N-diisopropylethylamine (80 mg, 0.80 mmol) was obtained. The mixture was heated to 100 ° C for 4 hours.
  • Step 2 (S)-6-(2,6-Dichloro-3,5-dimethoxyphenyl)-N-(pyrrolidin-3-yl)quinazolin-2-amine trifluoroacetic acid Salt synthesis
  • Step 3 (S)-1-(3-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)pyrrolidine Synthesis of -1-yl)prop-2-en-1-one
  • Step 1 (R)-3-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)pyrrolidine-1-carboxylic acid tert-butyl Ester synthesis
  • the starting material 2-chloro-6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazoline (100.0 mg, 0.27 mol, 1.0 eq) was dissolved in NMP (3 mL) and added ( R)-3-Aminopyrrolidine-1-carboxylic acid tert-butyl ester (57.4 mg, 0.31 Methyl, 1.5 eq) and DIPEA (80.2 mg, 0.62 mol, 2.3 eq), heated at 100 ° C for 4 h, the reaction was completely monitored by TLC, the reaction mixture was cooled to room temperature, the reaction mixture was poured into ice water, filtered, and the filter cake was collected.
  • Step 2 (R)-6-(2,6-Dichloro-3,5-dimethoxyphenyl)-N-(pyrrolidin-3-yl)quinazolin-2-amine trifluoroacetic acid Salt synthesis
  • Step 3 (R)-1-(3-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)pyrrolidine-1- Synthesis of propyl-2-en-1-one:
  • Step 1 Synthesis of tert-butyl 4-((6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)piperidine-1-carboxylate:
  • the starting material 2-chloro-6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazoline (100.0 mg, 0.27 mol, 1.0 eq) was dissolved in NMP (3 mL) and added 4 -aminopiperidine-1-carboxylic acid tert-butyl ester (61.7 mg, 0.31 mmol, 1.2 eq) and DIPEA (80.0 mg, 0.62 mol, 2.3 eq), heated at 100 ° C for 4 h, the reaction was completely monitored by TLC, and the reaction mixture was cooled to room temperature. The reaction solution was poured into ice water, suction filtered, and the filter cake was collected.
  • Step 2 Synthesis of 6-(2,6-dichloro-3,5-dimethoxyphenyl)-N-(piperidin-4-yl)quinazolin-2-amine trifluoroacetate
  • Step 3 1-(4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)piperidin-1-yl)-propene- Synthesis of 2-en-1-one:
  • EtOAc EtOAc: EtOAc (EtOAc) :1 to 40:1), 1-(4-((6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)piperidine- 1-yl)prop-2-en-1-one (40.0 mg, yield: 70%).
  • Step 1 trans-5-((6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2-azabicyclo[2.1. 1] Synthesis of tert-butyl hexane-2-carboxylate
  • Step 3 1-(trans-5-((6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2-azabicyclo [2.1.1] Synthesis of hexane-2-yl)prop-2-en-1-one (Compound 11)
  • Step 2 6-(2,6-Dichloro-3,5-dimethoxyphenyl)-N-((3S,4R)-4-methylpyrrolidin-3-yl)quinazoline-2 - Synthesis of amino hydrochloride
  • Step 3 1-((3S,4R)-3-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-4- Synthesis of methylpyrrolidin-1-yl)propyl-2-en-1-one (Compound 20)
  • Step 1 (2R,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2-methylpyrrole Synthesis of alkane-1-carboxylic acid tert-butyl ester
  • Step 2 6-(2,6-Dichloro-3,5-dimethoxyphenyl)-N-((3S,5R)-5-methylpyrrolidin-3-yl)quinazoline-2 -Amine synthesis
  • Step 3 1-((2R,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2- Synthesis of methylpyrrolidin-1-yl)prop-2-en-1-one (Compound 21)
  • Step 2 Synthesis of (2S,4S)-4-((((benzyloxy)carbonyl)amino)-2-(hydroxymethyl)pyrrolidine-1-carboxylic acid tert-butyl ester
  • Lithium aluminum hydride (0.7 g, 19.0 mmol, 2.0 eq) was dissolved in anhydrous tetrahydrofuran (20 mL) at 0 ° C, the reaction was stirred for half an hour, and 1-tert-butyl 2-methyl (2S) was slowly added dropwise.
  • 4S) 4-((benzyloxy)carbonyl)amino)pyrrolidine-1,2-dicarboxylate (3.6 g, 9.5 mmol, 1.0 eq) in tetrahydrofuran, gradually warmed to room temperature for 2 h. The reaction was completely detected by TLC.
  • reaction solution was cooled to 0 ° C, and water (0.7 mL) and 10% sodium hydroxide solution (0.7 mL) were slowly added dropwise to the reaction solution, and water (2.1 mL) was added thereto, and the mixture was stirred for half an hour to react.
  • the solution was filtered, and the filtrate was concentrated.
  • EtOAcjjjjjjjjjjjjjj Carbonyl)amino)-2-(hydroxymethyl)pyrrolidine-1-carboxylic acid tert-butyl ester (1.05 g, yield 30.0%).
  • Step 3 Synthesis of (2S,4S)-4-amino-2-(hydroxymethyl)pyrrolidine-1-carboxylic acid tert-butyl ester
  • Step 4 (2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin)-2-amino)-2-(hydroxymethyl) Synthesis of tert-butyl pyrrolidine-1-carboxylate
  • Step 5 ((2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin)-2-amino)pyrrolidine)-2- Synthesis of methanol hydrochloride
  • Step 6 1-((2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2- Synthesis of (hydroxymethyl)pyrrolidine)-1-propyl-2-en-1-one (Compound 22)
  • EtOAc EtOAc
  • EtOAc EtOAc
  • EtOAc EtOAc
  • ethyl acetate 8 mL ⁇ 2
  • EtOAc EtOAc m. 1 ⁇ 50:1
  • a pale yellow solid 1-((2S,4S)-4-((6-(2,6-dichloro-3,5-dimethoxy) Phenylphenyl)quinazolin-2-yl)amino)-2-(hydroxymethyl)pyrrolidin-1-yl)propyl-2-en-1-one (75.0 mg, yield 42.5%).
  • Step 2 Synthesis of (2S,4R)-2-(hydroxymethyl)-4-((tetrahydro-2H-pyran-2-yl)oxy)pyrrolidine-1-carboxylic acid tert-butyl ester
  • Lithium tetrahydrogen aluminum (3.1 g, 81.6 mmol) was added to tetrahydrofuran (100.0 mL), ethanol, water, cooled to -20 ° C on dry ice, and (2S, 4R)-4-((tetrahydro-2H-pyridyl) was added dropwise.
  • a solution of 1-(tert-butyl)-2-methyl ester (13.43 g, 40.8 mmol) in tetrahydrofuran (100.0 mL). Nitrogen protection The reaction was stirred at -20 ° C for three hours, and the reaction was completely monitored by TLC.
  • Step 4 Synthesis of (2S,4R)-2-(fluoromethyl)-4-((tetrahydro-2H-pyran-2-yl)oxy)pyrrolidine-1-carboxylic acid tert-butyl ester
  • Step 5 Synthesis of (2S,4R)-2-(fluoromethyl)-4-hydroxypyrrolidine-1-carboxylic acid tert-butyl ester
  • the starting material ((2S,4R)-2-(fluoromethyl)-4-((tetrahydro-2H-pyran-2-yl)oxy)pyrrolidine-1-carboxylic acid tert-butyl ester (3.2 g, 10.55) mmoL) was dissolved in a mixed solvent of acetic acid (33.0 mL), tetrahydrofuran (33.0 mL), water (33.0 mL), and was refluxed under reflux. The reaction was monitored by TLC. The pH was adjusted with saturated sodium bicarbonate (8-9). Tetrahydrofuran was spun off, and the aqueous phase was combined with ethyl acetate (3*50 mL).
  • Step 7 Synthesis of (2S,4S)-4-amino-2-(fluoromethyl)pyrrolidine-1-carboxylic acid tert-butyl ester
  • Step 8 (2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2-(fluoromethyl) Synthesis of tert-butyl pyrrolidine-1-carboxylate
  • N-diisopropylethylamine (178.21 mg, 1.38 mmol)
  • 2-chloro-6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazoline (203.1 mg, 0.552 MmoL)
  • TLC monitoring reaction was complete, the reaction solution was cooled to room temperature, 10 mL of water was added, filtered, the filter cake was washed with a small amount of ice water, and the filter cake was dissolved in dichloromethane (10 mL).
  • Step 9 6-(2,6-Dichloro-3,5-dimethoxyphenyl)-N-((3S,5S)-5-(fluoromethyl)pyrrolidin-3-yl)quinazole Synthesis of oxa-2-amine
  • Step 10 1-((2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2- (fluoromethyl)pyrrolidin-1-yl)prop-2-en-1-one
  • the starting material is 6-(2,6-dichloro-3,5-dimethoxyphenyl)-N-((3S,5S)-5-(fluoromethyl)pyrrolidin-3-yl)quinazoline 2-Amine (37.0 mg, 0.082 mmol) was dissolved in tetrahydrofuran (1.0 mL) and reacted to 0 ° C in an ice bath. Triethylamine (24.0 mg, 0.246 mmol) was added, and acryloyl chloride (11.13 mg, 0.123 mmol) was added at 0 ° C, and the mixture was slowly warmed to room temperature for two hours, and the reaction was monitored by TLC.
  • Step 2 (2S,4S)-2-(cyanomethyl)-4-((6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl Synthesis of amino)pyrrolidine-1-carboxylic acid tert-butyl ester
  • Step 3 2-((2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)pyrrolidine- Synthesis of 2-yl)acetonitrile
  • Step 4 2-((2S,4S)-1-acryloyl-4-((6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl) Synthesis of Amino)pyrrolidin-2-yl)acetonitrile
  • Step 3 (2S,4S)-2-carbamoyl-4-((6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino) Synthesis of tert-butyl pyrrolidine-1-carboxylate
  • Step 4 (2S,4S)-2-cyano-4-((6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)pyridinium Synthesis of tert-butyl 1 -carboxylic acid
  • Step 5 (2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)pyrrolidine-2-yl Nitrile synthesis
  • Step 3 (2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2-(dimethyl Base Synthesis of tert-butyl carbamoyl pyrrolidine-1-carboxylate
  • reaction was completed by TLC, and the reaction mixture was added to a dimethylamine/tetrahydrofuran solution (5 mL) cooled to 0 ° C. After 4 h of reaction, the reaction was completed by TLC. Saturated potassium carbonate solution (10 mL), saturated brine (10 mL), EA (20 mL), and the mixture was stirred. The aqueous phase was extracted with EA (10 mL ⁇ 2), and the organic phase was combined, dried and concentrated. .
  • Step 1 (2R,4R)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)pyrrolidine-1,2 -Synthesis of 1-(tert-butyl)-2-methyl dicarboxylate
  • Step 2 (2R,4R)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2-(2- Synthesis of tert-butyl hydroxypropyl-2-yl)-pyrrolidine-1-carboxylate
  • Step 3 2-((2R,4R)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-pyrrolidine Synthesis of 2-yl)propan-2-ol
  • Step 4 1-((2R,4R)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2- Synthesis of (2-hydroxypropyl-2-yl)pyrrolidin-1-yl)prop-2-en-1-one:
  • Step 1 (2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2-((1) Synthesis of 3-(2-oxoisoindoline-2-yl)amino)methyl)pyrrolidine-1-carboxylic acid tert-butyl ester
  • Step 3 (2S,4S)-2-(Acetylaminomethyl)-4-((6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl Synthesis of amino)pyrrolidine-1-carboxylic acid tert-butyl ester
  • Step 4 N-(((2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)pyrrolidine Synthesis of 2-yl)methyl)acetamide
  • Step 5 N-(((2S,4S)-1-acryloyl-4-((6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl) Synthesis of amino)pyrrolidin-2-yl)methyl)acetamide (Compound 32)
  • Step 1 (2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2-(methyl Synthesis of tert-butyl sulfonylaminomethyl)pyrrolidine-1-carboxylate
  • Step 2 N-(((2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)pyrrolidine Synthesis of 2-yl)methyl)methanesulfonamide
  • Step 3 N-(((2S,4S)-1-acryloyl-4-((6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl) Synthesis of amino)pyrrolidin-2-yl)methyl)methanesulfonamide (Compound 33)
  • Step 2 tert-Butyl (2S,4S)-2-(azetidin-1-carbonyl)-4-((6-(2,6-dichloro-3,5-dimethoxyphenyl) Synthesis of quinazolin-2-yl)amino)pyrrolidine-1-carboxylate
  • Step 3 Azetidin-1-yl ((2S,4S)-4-(6-(2,6-dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl) Synthesis of pyrrolidin-2-yl)methanone
  • Step 4 1-((2S,4S)-2-(azetidin-1-carbonyl)-4-((6-(2,6-dichloro-3,5-dimethoxyphenyl) Synthesis of quinazolin-2-yl)amino)pyrrolidin-1-yl)prop-2-en-1-one (Compound 35)
  • Step 1 (2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2-(methoxy Synthesis of tert-butyl ester of methyl)pyrrolidine-1-carboxylate
  • Step 3 1-((2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2- Synthesis of (methoxymethyl)pyrrolidin-1-yl)prop-2-en-1-one:
  • Step 1 (2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)pyrrolidine-1,2 -Synthesis of 1-(tert-butyl)-2-methyl dicarboxylate
  • Step 4 ((2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)pyrrolidine-2- Synthesis of ()pyrrolidin-1-yl)methanone
  • Step 5 1-((2S,4S)-4-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2- Synthesis of (pyrrolidine-1-carbonyl)pyrrolidin-1-yl)prop-2-en-1-one
  • EtOAc EtOAc
  • EtOAc m EtOAc m.
  • the ether (10 ml) was stirred for 2 hours and then suction filtered to give the product 1-((2,,,,,,,,,,,,, 2-yl)amino)-2-(pyrrolidin-1-carbonyl)pyrrolidin-1-yl)prop-2-en-1-one (162.8 mg, yield 66.1%).
  • Step 1 (3S,4R)-3-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin)-2-yl)amino)-4-methyl Synthesis of tert-butyl pyrrolidine-1-carboxylate
  • Step 2 (6-(2,6-Dichloro-3,5-dimethoxyphenyl)-N-((3S,4R)-4-methylpyrrolidin-3-yl)quinazoline- Synthesis of 2-amine hydrochloride:
  • Step 3 1-((3S,4R)-3-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-4- Preparation of methylpyrrolidin-1-yl)prop-2-yn-1-one
  • Step 1 6-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2-azaspiro[3.3]heptane- Synthesis of 2-carboxylic acid tert-butyl ester
  • Step 3 1-(6-((6-(2,6-Dichloro-3,5-dimethoxyphenyl)quinazolin-2-yl)amino)-2-azaspiro[3.3] Synthesis of heptane-2-yl)prop-2-en-1-one:
  • Test substance The compound of the present invention, the structure of which is shown above.
  • Test Instruments Use the LabChip EZ Reader II drug screening platform.
  • FGFR1-4(h) was reacted with 2.5 times enzymatic solution at room temperature for 10 min in 1% kinase buffer, and then FAM-labeled polypeptide substrate was added to initiate reaction with ATP. After incubation for 30 minutes, 25 ⁇ L of stop solution (100 mM HEPES, pH 7.5; 0.015% Brij-35; 0.2% Coating Reagent #3; 50 mM EDTA) Stop the reaction Caliper read the final data.
  • stop solution 100 mM HEPES, pH 7.5; 0.015% Brij-35; 0.2% Coating Reagent #3; 50 mM EDTA
  • Hep3B is an abnormal cell of hepatocellular carcinoma FGFR
  • RT112/84 is an abnormal cell of bladder cancer FGFR
  • DMS114 is a small cell lung cancer FGFR abnormal cell
  • AN3CA is an abnormal cell of endometrial cancer FGFR
  • SNU-16 is an abnormal cell of gastric cancer FGFR
  • Test substance The compound of the present invention, the structure of which is shown above.
  • Test instrument Use the Espire multi-function microplate reader.
  • Each strain of cells was inoculated in a 96-well plate and cultured overnight. Different concentrations of the compound (12 dose groups, 3 times DMSO serial dilution) were added to give a final concentration of 0.17-30000 nM, wherein the final DMSO content was 5 ⁇ .
  • the negative control wells were medium containing 5 ⁇ DMSO. After incubation at 37 ° C, 5% CO 2 , 95% humidity for 72 h, test. Add 30 ⁇ L of Cell titer-Glo reagent to each well and incubate for 30 min at room temperature. Espire reads the final data.
  • the compound of the present invention has good inhibitory activity against cells abnormal in FGFR such as Hep3B, RT112/84, DMS114, AN3CA, SNU-16, etc., indicating that the compound of the present invention can be used for the treatment of abnormalities by FGF/FGFR.
  • Guided cancers such as liver cancer, gastric cancer, small cell carcinoma, bladder cancer, and endometrial cancer have very good clinical value.

Abstract

提供了一种式I所示的成纤维细胞生长因子受体(FGFR)不可逆抑制剂,其药学上可接受的盐、立体异构体,药物制剂、药物组合物及其应用。R 1、R 2、R 3、R 4、环A、Ar、环B以及warhead如说明书中所定义。所述化合物对成纤维细胞生长因子受体具有高效的和高选择性的抑制作用,可以应用于FGF/FGFR异常介导的相关疾病的治疗,尤其是在癌症疾病方面的治疗。

Description

成纤维细胞生长因子受体抑制剂及其用途
本申请要求于2016年9月1日提交中国专利局的申请号为201610802100.5发明名称为“一种新型的用于成纤维细胞生长因子受体的抑制剂及其用途”和于2017年5月18日提交中国专利局的申请号为201710351160.4发明名称为“成纤维细胞生长因子受体抑制剂及其用途”的中国专利申请的优先权,其全部内容通过引用并入本申请中。
技术领域
本发明属于医药技术领域,涉及成纤维细胞生长因子受体(FGFR)不可逆抑制剂,或其药学上可接受的盐、立体异构体及其应用。
背景技术
酪氨酸激酶受体在肿瘤血管的生成、肿瘤细胞的增殖、迁移以及浸润方面发挥着重要的作用,目前已经相继有100多个酪氨酸激酶抑制剂药物上市或进入临床试验阶段。这些小分子酪氨酸激酶抑制剂(tyrosine kinase inhibitor,TKI)多以可逆性抑制的方式来发挥作用,由此带来了一些缺点:①选择性不够好,②药效不够强烈和持久,③易引发耐药性。因此,促使科学家将研究方向集中在不可逆的TKI的开发上。
不可逆性TKI通常以可逆性TKI的骨架结构为原型,在合适的位置连接上亲电的功能团,该亲电的功能团可以与酪氨酸激酶的ATP结合域附近的半胱氨酸残基(富电子的亲核结构)发生亲电反应形成共价键,从而不可逆的抑制激酶活性。与可逆性TKI相比,不可逆性TKI具有诸多独特的优势:①不可逆性TKI以永久性灭活的方式来发挥作用,这种抑制酶活性的方式使得其作用更为强烈而持久,即使药物分子从循环系统中被完全清除掉,其药效也仍能维持。②因为其与激酶的结合并不存在ATP竞争性,也使得激酶突变的可能性降低而减轻或规避了耐药性的产生。③因其分子结构上的亲电功能团可选择性地与半胱氨酸残基上的巯基反应,因此不可逆性TKI的选择性非常高。基于以上特点,开发不可逆的TKI正逐渐成为研发的热点方向。
成纤维细胞生长因子受体(fibroblast growth factor receptor,FGFR)是属于 酪氨酸激酶受体家族中的重要一员,FGFR包含4个成员,即FGFR-1、FGFR-2、FGFR-3和FGFR-4。它们多为单链的糖蛋白分子,分子质量在110~150kd,结构分为胞外区,跨膜区和胞内区组成。在正常生理条件下,FGFR与其配体成纤维细胞生长因子(fibroblast growth factor,FGF)结合,FGFR发生二聚体化以及自身的磷酸化,激活下游的信号通路,如JAK/STAT通路、磷脂酶C通路、磷酸酰肌醇-3-激酶PI3K以及MAPK信号通路,以上信号通路在肿瘤生长和血管发生过程中发挥着重要的作用。FGFR的异常高表达,与多种肿瘤,如肺癌、肝癌、脑胶质瘤、横纹肌肉瘤以及黑色素瘤的发生发展密切相关。
目前尚无不可逆FGFR抑制剂药物问世,尤其是对pan-FGFR具有高选择性的不可逆抑制剂上市。
发明内容
本发明的一个目的是提供一类新型高选择性不可逆pan-FGFR抑制剂,此类化合物对pan-FGFR具有很好的抑制活性,为由pan-FGFR异常介导的疾病的治疗提供了可能性。本发明还提供上述FGFR抑制剂的应用。
本发明采用的技术方案如下:
方案1:通式(I)所示的成纤维细胞生长因子受体(FGFR)不可逆抑制剂,或其药学上可接受的盐、立体异构体:
Figure PCTCN2017096848-appb-000001
其中,
R1、R2分别独立地选自氢、羟基、氨基、氰基、硝基、卤素原子、羧基、C1-6烷基、C1-6烷氧基、C1-6烷基氨基、(C1-6烷基)2氨基、卤代C1-6烷基、卤代C1-6烷氧基、C2-8烯基、C2-8炔基、C1-6烷基磺酰基、C1-6烷基羰基氨基、C1-6烷基取代的3~8元环烷基、C1-6烷基取代的3~8元杂环基,可供选择的,R1和R2可与它们分别连接的芳环或杂芳环上的两个原子一起形成3~8元环烷基、3~8元杂环基、6~14元芳基或5~10元杂芳基,并且任意环中的S原子可任选 被氧化为S(O)或S(O)2,任意环中的碳原子可任选地被氧化为C(O);
R3、R4分别独立地选自氢、羟基、氨基、氰基、硝基、卤素原子、羧基、C1-6烷基、C1-6烷氧基、C1-6烷基氨基、(C1-6烷基)2氨基、卤代C1-6烷基、卤代C1-6烷氧基、C2-8烯基、C2-8炔基、C1-6烷基磺酰基、C1-6烷基羰基氨基、C1-6烷基氨基羰基、3~8元环烷基、3~8元杂环基、6~14元芳基或5~10元杂芳基,C1-6烷基取代的3~8元环烷基、C1-6烷基取代的3~8元杂环基、C1-6烷基取代的6~14元芳基或C1-6烷基取代的5~10元杂芳基;
Ar选自任选的含0~3个O、S和/或N原子的6~14元芳环基或5~10元杂芳基;
环A选自任选被1~3个R5取代的含0~3个O、S和/或N原子的3~8元环烷基、3~8元杂环基、6~14元芳基或5~10元杂芳基,其中,任意环中的S原子可任选地被氧化为S(O)或S(O)2,任意环中的碳原子可任选地被氧化为C(O);
环B选自任选被1~3个R6取代的含至少1个N杂原子的3~10元饱和或不饱和的杂环基或5~6元含N杂芳基,并且,环B上的N原子与Warhead键直接相连,其中,任意环B中的S原子可任选被氧化为S(O)或S(O)2,并且任意的环B中的碳原子可任选地被氧化为C(O);
X选自CR7、N;
R5、R6、R7分别独立的选自
(i)氢,
(ii)羟基、氨基、羧基、氰基、硝基、卤素原子,
(iii)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~6烷基、C1-6烷氧基、C1-6烷氧C1-6烷氧基、C1-6烷基氨基、(C1-6烷基)2氨基、C1-6烷基羰基氨基、C1-6烷基磺酰氨基、3~8元杂环基取代的C1~6烷基、C1-6烷氧基、C1-6烷基氨基、(C1-6烷基)2氨基、卤代C1-6烷基、卤代C1-6烷氧基、C2-8烯基、C2-8炔基、C1-6烷基磺酰基、C1-6烷基硫基,所述的3~8元杂环基可任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~6烷基、C1-6烷氧基、C1-6烷基氨基、(C1-6烷基)2氨基取代,
(iv)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1-6烷基、C1-6烷氧基、C1-6烷基氨基、(C1-6烷基)2氨基取代的3~8元环烷基、3~8元杂环基,
(v)氨基-羰基、氰基-羰基、C1-6烷基-羰基、C1-6烷基氨基-羰基、(C1-6烷基)2氨基-羰基、C1-6烷氧基-羰基、3~8元环烷基-羰基、3~8元杂环基-羰基;
m1、m2代表1、2或3,且m1与m2相加小于等于5;
Warhead指的是能够与亲核试剂形成共价键的部分。
方案2:如方案1所述的化合物或其药学上可接受的盐、立体异构体:
其中,
R1分别独立的选自氢、卤素、羟基;
R2分别独立的选自氢、卤素、羟基、C1-4烷基、C1-4烷氧基、氰基、卤代C1-4烷基、卤代C1-4烷氧基;
Ar选自任选的含0~3个O、S和/或N原子的6~14元芳环基或5~6元杂芳基;
m1、m2代表1、2或3,且m1与m2相加小于等于5。
方案3:如方案2所述的化合物或其药学上可接受的盐、立体异构体:
其中,
R1分别独立的选自氢、卤素、羟基;
R2分别独立的选自氢、卤素、羟基、C1-4烷基、C1-4烷氧基、氰基、卤代C1-4烷基、卤代C1-4烷氧基;
R3、R4分别独立的选自氢、羟基、氨基、氰基、硝基、卤素原子、羧基、C1-4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、卤代C1-4烷基、卤代C1-4烷氧基、C2-4烯基、C2-4炔基、C1-4烷基磺酰基、C1-4烷基羰基氨基;
Ar为苯基;
环A选自任选被1~3个R5取代的苯基;环B选自任选被1~3个R6取代的含至少1个N杂原子的4~10元饱和或不饱和的杂环基,并且,环B上的N原子与Warhead键直接相连;
X选自CR7、N;
R5、R7分别独立的选自氢、羟基、氨基、羧基、氰基、硝基、卤素原子、C1-4烷基、C1-4烷氧基;
R6选自
(i)氢,
(ii)羟基、氨基、羧基、氰基、硝基、卤素原子,
(iii)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷氧C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、C1-4烷基羰基氨基、C1-4烷基磺酰氨基、3~8元杂环基取代的C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、卤代C1-4烷基、卤代C1-4烷氧基、C2-4烯基、C2-4炔基、C1-4烷基磺酰基、C1-4烷基硫基,所述的3~8元杂环基可任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基取代,
(iv)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1-4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基取代的3~8元环烷基、3~8元杂环基,
(v)氨基-羰基、氰基-羰基、C1-4烷基-羰基、C1-4烷基氨基-羰基、(C1-4烷基)2氨基-羰基、C1-4烷氧基-羰基、3~8元环烷基-羰基、3~8元杂环基-羰基;
m1、m2代表1、2或3,且m1与m2相加小于等于5;
Warhead指的是能够与亲核试剂形成共价键的部分。
方案4.如方案1~3任一方案所述的式(I)化合物或其药学上可接受的盐、立体异构体,结构如通式(II)所示:
Figure PCTCN2017096848-appb-000002
R4选自氢或C1-4烷基;
环A为苯基;
环B选自任选被1~3个R6取代的含至少1个N杂原子的4~6元饱和或不饱和的单杂环基或6~10元饱和或不饱和的稠杂环基,并且,环B上的N原子与Warhead键直接相连;
X为N;
R6选自
(i)氢,
(ii)羟基、氨基、羧基、氰基、硝基、卤素原子,
(iii)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷氧C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、C1-4烷基羰基氨基、C1-4烷基磺酰氨基、3~8元杂环基取代的C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、卤代C1-4烷基、卤代C1-4烷氧基、C2-4烯基、C2-4炔基、C1-4烷基磺酰基、C1-4烷基硫基,所述的3~8元杂环基可任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基取代,
(iv)氨基-羰基、氰基-羰基、C1-4烷基-羰基、C1-4烷基氨基-羰基、(C1-4烷基)2氨基-羰基、C1-4烷氧基-羰基、3~8元环烷基-羰基、3~8元杂环基-羰基;
Warhead选自
Figure PCTCN2017096848-appb-000003
Z指离去基团或活化羟基部分,
R11,R12,R13独立地选自氢,卤素,氰基,任选被取代基取代的C1-4烷基、卤代C1-4烷基、3~8元环烷基、3~8元杂环基、5~8元芳基、5~10元杂芳基,所述取代基选自:羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷氧C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、C1-4烷基羰基氨基、C1-4烷基磺酰氨基、3~8元杂环基;R11,R12,R13优选为氢。
方案5:如方案1-4任一方案所述的化合物或其药学上可接受的盐、立体异构体,结构如通式(II)所示:
Figure PCTCN2017096848-appb-000004
Figure PCTCN2017096848-appb-000005
Warhead与环B上的N原子直接相连,如下所示:
Figure PCTCN2017096848-appb-000006
其中,环B选自如下基团:
Figure PCTCN2017096848-appb-000007
方案6:根据方案1-5任一方案所述的化合物或其药学上可接受的盐、立体异构体,
warhead选自如下结构:
Figure PCTCN2017096848-appb-000008
Z指离去基团或活化羟基部分,
R11,R12,R13分别独立地选自氢或C1-4烷基。
方案7:本发明的化合物、其药学上可接受的盐或其立体异构体为:
Figure PCTCN2017096848-appb-000009
Figure PCTCN2017096848-appb-000010
方案8.如方案4所述的化合物或其药学上可接受的盐、立体异构体:
环B选自任选被1~3个R6取代的含至少1个N杂原子的5~6元饱和单杂环基,并且,环B上的N原子与Warhead键直接相连。
方案9.如方案8所述的化合物或其药学上可接受的盐、立体异构体,结构如通式(III)所示:
Figure PCTCN2017096848-appb-000011
R4选自氢或C1-4烷基;
X为N;
R6选自
(i)氢,
(ii)羟基、氨基、羧基、氰基、硝基、卤素原子,
(iii)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷氧C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、C1-4烷基羰基氨基、C1-4烷基磺酰氨基、3~8元杂环基取代的C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、卤代C1-4烷基、卤代C1-4烷氧基、C2-4烯基、C2-4炔基、C1-4烷基磺酰基、C1-4烷基硫基,所述的3~8元杂环基可任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基取代,
(iv)氨基-羰基、氰基-羰基、C1-4烷基-羰基、C1-4烷基氨基-羰基、(C1-4烷基)2氨基-羰基、C1-4烷氧基-羰基、3~8元环烷基-羰基、3~8元杂环基-羰基;
m为1~3的整数;
Warhead选自
Figure PCTCN2017096848-appb-000012
Z指离去基团或活化羟基部分;
R11,R12,R13独立地选自氢,卤素,氰基,任选被取代基取代的C1-4烷基、卤代C1-4烷基、3~8元环烷基、3~8元杂环基、5~8元芳基、5~10元杂芳基,所述取代基选自:羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷氧C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、C1-4烷基羰基氨基、C1-4烷基磺酰氨基、3~8元杂环基;R11,R12,R13优选为氢。
方案10.如方案9所述的化合物或其药学上可接受的盐、立体异构体:
R6选自
(i)氢,
(ii)羟基、氨基、羧基、氰基、硝基、卤素原子,
(iii)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷氧C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、C1-4烷基羰基氨基、C1-4烷基磺酰氨基、3~8元杂环基取代的C1~4烷基、C1-4烷氧基,所述的3~8元杂环基可任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基取代,所述3~8元杂环基优选是4~6元饱和杂环基,更优选氮杂环丁烷基、吡咯烷基、哌啶基、吗啉基;
(iv)氨基-羰基、氰基-羰基、C1-4烷基-羰基、C1-4烷基氨基-羰基、(C1-4烷基)2氨基-羰基、C1-4烷氧基-羰基、3~8元环烷基-羰基、3~8元杂环基-羰基;
Warhead选自
Figure PCTCN2017096848-appb-000013
R11,R12,R13独立地选自氢或C1-4烷基,优选为
Figure PCTCN2017096848-appb-000014
方案11.如方案10所述的化合物或其药学上可接受的盐、立体异构体,可选自如下结构的化合物:
Figure PCTCN2017096848-appb-000015
Figure PCTCN2017096848-appb-000016
Figure PCTCN2017096848-appb-000017
Figure PCTCN2017096848-appb-000018
本发明的化合物、其药学上可接受的盐或其立体异构体为:
Figure PCTCN2017096848-appb-000019
Figure PCTCN2017096848-appb-000020
本发明还要求保护本发明任一化合物或其药学上可接受的盐、立体异构体的药物制剂,所述药物制剂还包含一种或多种药用载体。
本发明所述的药用载体可以是一种或多种适合于人使用的固体或液体填料或凝胶物质。所述药用载体优选具有足够的纯度和足够低的毒性,并且与本发明活性成分具有相容性且不明显减低活性成分的药效。例如,药用载体可以填充剂、粘合剂、崩解剂、润滑剂、水性溶剂或非水性溶剂等。
本发明所述的药物制剂,可以制成药学上可接受的任意剂型,以任何合适的给药方式,例如通过口服、肠胃外、直肠或经肺给药等方式施用于需要这种治疗的患者或受试者。用于口服给药时,可以制成片剂、胶囊剂、丸剂、颗粒剂等。用于肠胃外给药时,可以制成注射液、注射用无菌粉末等。
本发明所述的药物制剂,进一步包含一种或多种第二治疗活性剂,所述的第二治疗活性剂为抗代谢物、生长因子抑制剂、有丝分裂抑制剂、抗肿瘤 激素类、烷化剂类、金属类、拓扑异构酶抑制剂、激素药、免疫调节剂、肿瘤抑制基因、癌疫苗、免疫检查点或肿瘤免疫治疗相关的抗体和小分子药物。
本发明还要求保护本发明任一化合物或其药学上可接受的盐、立体异构体或者根据本发明的药物制剂在制备治疗FGF/FGFR异常介导的疾病的药物中的应用。本发明所述的FGF/FGFR异常介导的疾病为癌症;所述的癌症包括肺癌、鳞状上皮细胞癌、膀胱癌、胃癌、卵巢癌、腹膜癌、乳腺癌、乳腺导管癌、头颈癌、子宫内膜癌、宫体癌、直肠癌、肝癌、肾癌、肾盂癌、食管癌、食管腺癌、神经胶质瘤、前列腺癌、甲状腺癌、女性生殖系统癌症、原位癌、淋巴瘤、神经纤维瘤病、骨癌、皮肤癌、脑癌、结肠癌、睾丸癌、胃肠道间质瘤、口腔癌、咽癌、多发性骨髓瘤、白血病、非霍奇金淋巴瘤、大肠绒毛腺瘤、黑色素瘤、细胞瘤和肉瘤,骨髓增生异常综合症。
本发明还提供一种用于治疗FGF/FGFR异常介导的疾病的方法,所述方法包括向有此需要的对象施用本发明任一化合物或其药学上可接受的盐、立体异构体或者根据本发明的药物制剂。本发明所述的FGF/FGFR异常介导的疾病为癌症;所述的癌症包括肺癌、鳞状上皮细胞癌、膀胱癌、胃癌、卵巢癌、腹膜癌、乳腺癌、乳腺导管癌、头颈癌、子宫内膜癌、宫体癌、直肠癌、肝癌、肾癌、肾盂癌、食管癌、食管腺癌、神经胶质瘤、前列腺癌、甲状腺癌、女性生殖系统癌症、原位癌、淋巴瘤、神经纤维瘤病、骨癌、皮肤癌、脑癌、结肠癌、睾丸癌、胃肠道间质瘤、口腔癌、咽癌、多发性骨髓瘤、白血病、非霍奇金淋巴瘤、大肠绒毛腺瘤、黑色素瘤、细胞瘤和肉瘤、骨髓增生异常综合症。
本发明还涉及本发明任一化合物或其药学上可接受的盐、立体异构体或者根据本发明的药物制剂用作药物。
发明详述
本发明所述的“卤素”是指氟、氯、溴、碘等,优选氟原子,氯原子。
本发明所述的“氧代”是指取代基结构中的任一C可被“-C(O)-”替换;若含有杂原子,其杂原子可形成氧化物,如
Figure PCTCN2017096848-appb-000021
可被
Figure PCTCN2017096848-appb-000022
替换,如S可被 氧化为S(O)或S(O)2
本发明所述的“卤代”是指取代基中的任一氢原子可被一个或多个相同或不同的卤素原子取代。“卤素”如前文所定义。
本发明所述的“C1-6烷基”指含有1~6个碳原子的烃部分去除一个氢原子衍生的直链或支链的烷基,如甲基、乙基、正丙基、异丙基、正丁基、异丁基、仲丁基、叔丁基、正戊基、异戊基、2-甲基丁基、新戊基、1-乙基丙基、正己基、异己基、4-甲基戊基、3-甲基戊基、2-甲基戊基、1-甲基戊基、3,3-二甲基丁基、2,2-二甲基丁基、1,1-二甲基丁基、1,2-二甲基丁基、1,3-二甲基丁基、2,3-二甲基丁基、2-乙基丁基和1-甲基-2-甲基丙基等。所述“C1-4烷基”指含有1~4个碳原子的上述实例。
本发明所述的“C2-8烯基”指含有碳碳双键的2~8个碳原子的烯烃部分去除一个氢原子衍生的直链或支链或环状的烯烃基,如乙烯基、1-丙烯基、2-丙烯基、1-丁烯基、2-丁烯基、1,3-丁二烯基、1-戊烯基、2-戊烯基、3-戊烯基、1,3-戊二烯基、1,4-戊二烯基、1-己烯基、1,4-己二烯基、环丁烯基、环戊烯基、环己烯基、1,4-环己二烯基、环庚烯基、1,4-环庚二烯基、环辛烯基、1,5-环辛二烯基等,还包括可能形成的多环系统,例如螺环烯、并环烯、桥环烯等
本发明所述的“C2-8炔基”指含有碳碳叁键的2~8个碳原子的炔烃部分去除一个氢原子衍生的直链或支链的炔烃基,如乙炔基、丙炔基、2-丁炔基、2-戊炔基、3-戊炔基、4-甲基-2-戊炔基、2-己炔基、3-己炔基等。
本发明所述的“C1-6烷基氨基”、“(C1-6烷基)2氨基”、“C1-6烷基羰基氨基””、“C1-6烷基磺酰氨基”、“C1-6烷基氨基羰基””、“(C1-6烷基)2氨基-羰基”、“C1-6烷氧基-羰基”、“C1-6烷基磺酰基””、“C1-6烷基硫基”、“C1-6烷基-羰基”、“3~8元环烷基-羰基”、“3~8元杂环基-羰基”分别指C1-6烷基-NH-、(C1-6烷基)(C1-6烷基)N-、C1-6烷基-C(O)-NH-、C1-6烷基-S(O)2-NH2-、C1-6烷基-NH-C(O)-、(C1-6烷基)(C1-6烷基)N-C(O)-、C1-6烷基-O-C(O)-、C1-6烷基-S(O)2-、C1-6烷基-S-、C1-6烷基-C(O)-、3~8元环烷基-C(O)-、3~8元杂环基-C(O)-;所述“C1-6烷基”如前文所定义,优选为“C1-4烷基”。
本发明所述的“C1-6烷氧基”是指前文所定义的“C1-6烷基”通过氧原子与母体分子连接的基团,即“C1-6烷基-O-”基团,如甲氧基、乙氧基、正丙氧基、 异丙氧基、正丁氧基、叔丁氧基、正戊氧基、新戊氧基和正己氧基等。所述的“C1-4烷氧基”指含有1~4个碳原子的上述实例,即“C1-4烷基-O-”基团。
本发明所述的“稠环”是指由两个或两个以上环状结构以并、螺、桥的连接方式所形成的多环系结构。所述的并环是指由两个或两个以上环状结构彼此公用两个相邻的环原子(即共用一个键)所形成的稠环结构。所述的桥环是指有两个或两个以上环装结构彼此共用两个非相邻的环原子所形成的稠环结构。所述的螺环是指由两个或两个以上环状结构彼此共用一个环原子所形成的稠环结构。
本发明所述的“环烷基”,是指单环环烷基,双环环烷基系统或者是多环环烷基系统(也称为稠环系统)。单环系统是含3至8个碳原子的环烃基基团。3~8元环烷基实例包括但不限于:环丙烷基、环丁烷基、环戊烷基、环己烷基、环庚烷基、环辛烷基等。稠环环烷基包括并环环烷基、桥环烷基、螺环烷基。
并环环烷基可以为6-12元并环环烷基、7-10元并环环烷基,其的代表性例子包括但不限于双环[3.1.1]庚烷、双环[2.2.1]庚烷、双环[2.2.2]辛烷、双环[3.2.2]壬烷、双环[3.3.1]壬烷和双环[4.2.1]壬烷。
所述的螺环基可以为6-12元螺环基、7-11元螺环基,其实例包括但不限于:
Figure PCTCN2017096848-appb-000023
所述的桥环基可以为6-12元桥环基、7-11元桥环基,其实例包括但不限于:
Figure PCTCN2017096848-appb-000024
本发明所述的“杂环基”是指至少一个环碳原子被选自O、S、N的杂原子替代的非芳香性的环状基团,优选1~3个杂原子,同时包括碳原子、氮原子和硫原子可以被氧代。
“杂环基”,是指单环杂环基、双环杂环基系统或多环杂环基系统(也称为稠环系统),包括饱和、部分饱和的杂环基,但不包括芳环。单杂环基可以为3~8元杂环基、3~8元饱和杂环基、3~6元杂环基、4~7元元杂环基、5~7元杂环基、5~6元杂环基、5~6元含氧杂环基、5~6元含氮杂环基、5~6元饱和杂环基等。“3-8”元饱和杂环基,其实例包括但不限于氮杂环丙烷基、氧杂环 丙烷基、硫杂环丙烷基、氮杂环丁烷基、氧杂环丁烷基、硫杂环丁烷基、四氢呋喃基、四氢吡咯基、四氢噻吩基、咪唑烷基、吡唑烷基、1,2-噁唑烷基、1,3-噁唑烷基、1,2-噻唑烷基、1,3-噻唑烷基、四氢-2H-吡喃基、四氢-2H-噻喃基、哌啶基、哌嗪基、吗啉基、1,4-二氧杂环己烷基、1,4-氧硫杂环己烷基;“3-8”元部分饱和杂环基,其实例包括但不限于4,5-二氢异噁唑基、4,5-二氢噁唑基、2,5-二氢噁唑基、2,3-二氢噁唑基、3,4-二氢-2H-吡咯基、2,3-二氢-1H-吡咯基、2,5-二氢-1H-咪唑基、4,5-二氢-1H-咪唑基、4,5-二氢-1H-吡唑基、4,5-二氢-3H-吡唑基、4,5-二氢噻唑基、2,5-二氢噻唑基、2H-吡喃基、4H-吡喃基、2H-噻喃基、4H-噻喃基、2,3,4,5-四氢吡啶基、1,2-异噁嗪基、1,4-异噁嗪基或6H-1,3-噁嗪基等。稠杂环包括并杂环基、螺杂环基、桥杂环基,可以是饱和的、部分饱和的或不饱和的,但不是芳香性的。稠杂环基是稠合到苯环、5-6元的单环环烷基、5-6元单环环烯基、5-6元单环杂环基或5-6元单环杂芳基的5-6元单环杂环基环。所述的并杂环基可以为6-12元并环基、7-10元并环基、6-10元并环基、6-12元饱和并环基,代表性实例包括但不限于:3-氮杂双环[3.1.0]己烷基、3,6-二氮杂双环[3.2.0]庚烷基、3,8-二氮杂双环[4.2.0]辛烷基、3,7-二氮杂双环[4.2.0]辛烷基、八氢吡咯并[3,4-c]吡咯基、八氢吡咯并[3,4-b]吡咯基、八氢吡咯并[3,4-b][1,4]噁嗪基、八氢-1H-吡咯并[3,4-c]吡啶基、2,3-二氢苯并呋喃-2-基、2,3-二氢苯并呋喃-3-基、二氢吲哚-1-基、二氢吲哚-2-基、二氢吲哚3-基、2,3-二氢苯并噻吩-2基、八氢-1H-吲哚基、八氢苯并呋喃基。
所述的螺杂环基可以为6-12元螺杂环基、7-11元螺杂环基、6-12元饱和螺环基,其实例包括但不限于:
Figure PCTCN2017096848-appb-000025
Figure PCTCN2017096848-appb-000026
所述的桥杂环基可以为6-12元桥杂环基、7-11元桥杂环基、6-12元饱和 桥杂环基,其实例包括但不限于:
Figure PCTCN2017096848-appb-000027
Figure PCTCN2017096848-appb-000028
本发明所述“6~14元芳基”,是指含有6~14个碳原子的环状芳香性基团,包括“6-8元单环芳基”,例如苯基、;包括“8~14元稠环芳基”,例如戊搭烯、萘、菲等。
本发明所述“杂芳基”可以是5-10元杂芳基,是指至少一个环碳原子被选自O、S、N的杂原子替代的芳香性的环状基团,优选1~3个杂原子,同时包括碳原子、硫原子被氧代的情况,例如碳原子被C(O)替代,硫原子被S(O)、S(O)2替代。杂芳基包括单杂芳基和稠杂芳基。单杂芳基可以为5~7元杂芳基、5~6元杂芳基,其实例包括但不仅限于呋喃基、咪唑基、异噁唑基、噻唑基、异噻唑基、噁二唑基、噁唑基、吡啶基、哒嗪基、嘧啶基、吡嗪基、吡唑基、吡咯基、四唑基、噻二唑基、噻吩基、三唑基和三嗪基。在某些实施例中,稠杂芳基是稠合到苯基环、5元或6元单环环烷基、5元或6元单环环烯基、5元或6元单环杂环基、或5元或6元单环杂芳基的5元或6元单环杂芳环,其中稠合的环烷基、环烯基和杂环基被作为独立氧代基或硫代基的一个或两个基团选择性取代。稠杂芳基可以为8-12元并杂芳基、9~10元并杂芳基,例子包括但不限于苯并咪唑基、苯并呋喃基、苯并噻吩基、苯并噁二唑基、苯并噻二唑基(benzthiadiazolyl)、苯并噻唑基、噌啉基、5,6-二氢喹啉-2-基、5,6-二氢异喹啉-1-基、呋喃并吡啶基(furopyridinyl)、吲唑基、吲哚基、异喹啉基、萘啶基、嘌呤基、喹啉基、5,6,7,8-四氢喹啉-2-基、5,6,7,8-四氢喹啉基、5,6,7,8-四氢喹啉-4-基、5,6,7,8-四氢异喹啉-1-基、噻吩并吡啶基(thienopyridinyl)、4,5,6,7-四氢并[c][1,2,5]噁二唑基和6,7-二氢并[c][1,2,5]噁二唑-4(5H)酮基。
本发明所述的“药学上可接受的盐”是指可药用的酸和碱的加成盐或其溶剂化物。这样的可药用盐包括诸如以下的酸的盐:盐酸、磷酸、氢溴酸、硫酸、亚硫酸、甲酸、甲苯磺酸、甲磺酸、硝酸、苯甲酸、柠檬酸、酒石酸、马来酸、氢碘酸、链烷酸(诸如乙酸、HOOC-(CH2)n-COOH(其中n是0~4))等。碱的盐:钠盐、钾盐、钙盐、铵盐等。本领域技术人员知晓多种无毒的 可药用加成盐。
本发明式(I)化合物的“立体异构体”是指当式(I)、(II)、(III)化合物存在不对称碳原子时,会产生对映异构体;当化合物存在碳碳双键或环状结构时,会产生顺反异构体;当化合物存在酮或肟时,会产生互变异构体,所有式(I)、(II)、(III)化合物的对映异构体、非对映异构体、消旋异构体、顺反异构体、互变异构体、几何异构体、差向异构体及其混合物,均包括在本发明范围中。
本发明所述的“Warhead”指的是能够与亲核试剂形成共价键的部分。“亲核试剂”是指向亲电体供给电子对以在反应中形成化学键的物质。在一些实施方案中,亲试核剂可为氧亲核试剂,例如,水或羟基;氮亲核试剂,例如,胺;或硫亲核试剂,例如,巯基,诸如,胱氨酸残基侧链中的巯基。
本发明所述的“warhead”是指抑制剂中可逆地或不可逆地参与供体(例如,蛋白质)与底物的反应的部分。warhead可(例如)与蛋白质形成共价键,或可生成稳定过渡态,或是可逆不可逆烷基化剂。例如,warhead可为抑制剂上可参与键形成反应的官能基,其中在warhead的一部分与供体(例如蛋白质的氨基酸残基)之间形成新的共价键。warhead是亲电体且“供体”是亲核试剂,诸如半胱氨酸残基侧链。适宜做warhead部分的包括但不限于以下结构:
Figure PCTCN2017096848-appb-000029
其中,
Z指离去基团(诸如卤素)或活化羟基部分(例如三氟甲磺酸酯);
R11,R12,R13独立地选自氢,卤素,氰基,任选被取代基取代的C1-4烷基、卤代C1-4烷基、3~8元环烷基、3~8元杂环基、5~8元芳基、5~10元杂芳基,所述取代基选自:羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、 C1-4烷氧基、C1-4烷氧C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、C1-4烷基羰基氨基、C1-4烷基磺酰氨基、3~8元杂环基;R11,R12,R13优选为氢。
本文中使用的缩写“NMP”是指N-甲基吡咯烷酮;“DIPEA”是指N,N-二异丙基乙胺;“TLC”是指薄层色谱;“PE:EA”是指石油醚:乙酸乙酯;“TFA”是指三氟乙酸;“THF”是指四氢呋喃;“EA”是指乙酸乙酯;“DCM:MeOH”是指二氯甲烷:甲醇;“DCM”是指二氯甲烷;“MTBE”是指甲基叔丁基醚;“TFAA”是指三氟乙酸酐。
具体实施方式
实施例1:化合物1的合成
Figure PCTCN2017096848-appb-000030
合成路线如下所示:
Figure PCTCN2017096848-appb-000031
中间体I-1的合成
将SM1(5.00g,20.5mmol)和SM2(3.73g,20.5mmol)溶于四氢呋喃(30ml)中,加入碳酸铯(20.00g,61.5mmol)的水(30ml)溶液,加入催化量的Pd(PPh3)Cl2,所得到的混合物在氮气氛围下加热回流反应4个小时。
将反应液浓缩至干,用乙酸乙酯萃取。有机相用饱和氯化钠洗涤一次,用无水硫酸钠干燥,减压浓缩得到的粗品进行柱层析(200-300目硅胶,石油醚/乙酸乙酯=10/1)得到中间体I-1(3.80g,62%收率),为淡黄色固体。
中间体I-2的合成
将I-1(3.80g,12.6mmol)溶于四氢呋喃(100ml)中,在氮气保护下降温到-20~30℃,滴加磺酰氯(5.11g,37.9mmol),所得到的混合物在相同温度下反应2个小时。
将反应液缓慢升到环境温度,加入乙腈(100ml),搅拌10分钟,过滤收集生成的固体。烘干,得到中间体I-2(2.80g,60%收率),为淡黄色固体。
中间体I-3的合成
将SM3(81mg,0.40mmol)和I-2(100mg,0.35mmol)加入到N-甲基吡咯烷酮(3ml)中,加入N,N-二异丙基乙胺(80mg,0.80mmol),所得到的混合物加热到100℃反应4个小时。
将反应液倒入冰水中,过滤收集析出的固体。固体进行柱层析(200-300目硅胶,石油醚/乙酸乙酯=3/1)得到中间体I-3(100mg,54%收率),为淡黄色固体。
中间体I-4的合成
将I-3(100mg,0.19mmol)溶于二氯甲烷(5ml)中,加入三氟乙酸(1ml),所得到的混合物在环境温度下反应4个小时。
将反应液浓缩至干,加入甲苯,再次减压浓缩,除去残留的三氟乙酸。得到的粗品I-4(81mg,100%收率),为黄色油状物,直接进行下一步反应。
化合物1的合成
将I-4(81mg,0.18mmol)溶于四氢呋喃(10ml)中,加入三乙胺(0.1ml,0.54mmol),加入丙烯酰氯(25mg,0.21mmol),所得到的混合物在环境温度下反应4个小时。加入少量的甲醇,淬灭剩余的丙烯酰氯,所得到的混合物减压浓缩,得到的粗品进行柱层析(200-300目硅胶,石油醚/乙酸乙酯=1/1)得到化合物1(11mg,15%收率),为淡黄色固体。
1H NMR(400MHz,DMSO-d6)δ(ppm)9.18(s,1H),7.51-7.69(m,4H),7.01(s,1H),6.79-6.86(m,1H),6.04-6.09(m,1H),5.66-5.87(m,1H),3.86-4.40(m,9H),3.07-3.17(m,1H),2.90-2.95(m,1H),1.99-2.01(m,1H),1.36-1.84(m,3H);LC-MS[M+H]=488。
实施例2:(S)-1-(3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-基)丙-2-烯-1-酮(化合物5)的合成
Figure PCTCN2017096848-appb-000032
步骤1:(S)-3-(6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000033
将原料2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(100.0mg,0.27mol,1.0eq)溶解在NMP(3mL)中,加入(S)-3-氨基吡咯烷-1-甲酸叔丁酯(55.4mg,0.30mmol,1.1eq)和DIPEA(104.9mg,0.81mol,3.0eq),100℃加热反应4h,TLC监测反应完全,反应液冷却至室温,将反应液倒入冰水中,抽滤,收集滤饼,粗品经硅胶柱层析(PE:EA=10:1~2:1),得到黄色液体(S)-3-(6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(78.7mg,收率:47%)。
步骤2:(S)-6-(2,6-二氯-3,5-二甲氧基苯基)-N-(吡咯烷-3-基)喹唑啉-2-胺三氟乙酸盐的合成
Figure PCTCN2017096848-appb-000034
将(S)-3-(6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(78.7mg,0.15mmol,1.0eq)溶于二氯甲烷(8mL)中,加入TFA(1mL),室温反应8h,TLC监测反应完全,浓缩得到黄色液体(S)-6-(2,6-二氯-3,5-二甲氧基苯基)-N-(吡咯烷-3-基)喹唑啉-2-胺三氟乙酸盐(77.9mg,收率:以100%计)。
步骤3:(S)-1-(3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷 -1-基)丙-2-烯-1-酮的合成
Figure PCTCN2017096848-appb-000035
将(S)-6-(2,6-二氯-3,5-二甲氧基苯基)-N-(吡咯烷-3-基)喹唑啉-2-胺三氟乙酸盐(77.9mg,0.15mmol,1.0eq)溶于THF(10mL)中,加入三乙胺(45.5mg,0.45mmol,2.0eq),搅拌反应30min,加入丙烯酰氯(27.0mg,0.30mmol,2.0eq),反应8h,TLC监测反应完全。加入饱和碳酸氢钠溶液(20mL),EA萃取(3×20mL),合并有机相,无水硫酸镁干燥,过滤,浓缩,粗品经硅胶柱层析(200-300目硅胶,DCM:MeOH=100:1~20:1)纯化得到(S)-1-(3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-基)丙-2-烯-1-酮(28.4mg,收率:40%)。
1HNMR(400MHz,DMSO-d6)δ(ppm):9.19(s,1H),7.84-7.89(m,1H),7.69(s,1H),7.50-7.59(m,2H),7.01(s,1H),6.52-6.63(m,1H),6.10-6.17(m,1H),5.61-5.70(m,1H),4.40-4.60(m,1H),3.93(s,6H),3.49-3.78(m,4H),2.20-2.35(m,3H)。
分子式:C23H22Cl2N4O3  分子量:473.35  LC-MS(m/z)=473.25[M+H+]。
实施例3:(R)-1-(3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-基)丙-2-烯-1-酮(化合物6)的合成
Figure PCTCN2017096848-appb-000036
步骤1:(R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000037
将原料2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(100.0mg,0.27mol,1.0eq)溶解在NMP(3mL)中,加入(R)-3-氨基吡咯烷-1-甲酸叔丁酯(57.4mg,0.31 mmol,1.5eq)和DIPEA(80.2mg,0.62mol,2.3eq),100℃加热反应4h,TLC监测反应完全,反应液冷却至室温,将反应液倒入冰水中,抽滤,收集滤饼,粗品经硅胶柱层析(PE:EA=10:1~2:1),得到(R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(88mg,收率:67%)。
步骤2:(R)-6-(2,6-二氯-3,5-二甲氧基苯基)-N-(吡咯烷-3-基)喹唑啉-2-胺三氟乙酸盐的合成
Figure PCTCN2017096848-appb-000038
将中间体(R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯((88.0mg,0.17mmol,1.0eq)溶于DCM(5mL)中,加入三氟乙酸(2.5mL),室温反应8h,TLC监测反应完全,直接浓缩溶液至干,得到(R)-6-(2,6-二氯-3,5-二甲氧基苯基)-N-(吡咯烷-3-基)喹唑啉-2-胺三氟乙酸盐(90.4mg,收率:以100%计)。
步骤3:(R)-1-(3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-基)丙-2-烯-1-酮的合成:
Figure PCTCN2017096848-appb-000039
将中间体(R)-6-(2,6-二氯-3,5-二甲氧基苯基)-N-(吡咯烷-3-基)喹唑啉-2-胺三氟乙酸盐(90.4mg,0.17mmol,1.0eq)溶于THF(8mL)中,加入三乙胺(68.6mg,0.68mmol,4.0eq),搅拌反应30min,加入丙烯酰氯(18.4mg,0.20mmol,1.2eq),反应8h,TLC监测反应完全,加入饱和碳酸氢钠溶液(20mL),EA萃取(3×15mL),合并有机相,无水硫酸镁干燥,过滤,浓缩,粗品经硅胶柱层析(DCM:MeOH=100:1~40:1),得到(R)-1-(3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-基)丙-2-烯-1-酮(56.0mg,收率:70%)。
1HNMR(400MHz,DMSO-d6)δ(ppm):9.19(m,1H),7.82(m,2H),7.51-7.69(m,2H),7.02(s,1H),6.53-6.63(m,1H),6.11-6.17(m,1H),5.62-5.69(m,1H), 4.53-5.61(d,1H),3.97(s,6H),3.52-3.78(m,3H),2.07-2.29(m,3H)。
分子式:C23H22Cl2N4O3  分子量:473.35。
实施例4:1-(4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)哌啶-1-基)丙-2-烯-1-酮(化合物10)的合成
Figure PCTCN2017096848-appb-000040
步骤1:4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)哌啶-1-甲酸叔丁酯的合成:
Figure PCTCN2017096848-appb-000041
将原料2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(100.0mg,0.27mol,1.0eq)溶解在NMP(3mL)中,加入4-氨基哌啶-1-甲酸叔丁酯(61.7mg,0.31mmol,1.2eq)和DIPEA(80.0mg,0.62mol,2.3eq),100℃加热反应4h,TLC监测反应完全,反应液冷却至室温,将反应液倒入冰水中,抽滤,收集滤饼,粗品经硅胶柱层析(PE:EA=10:1~2:1),得到4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)哌啶-1-甲酸叔丁酯(62.6mg,收率:43%)。
步骤2:6-(2,6-二氯-3,5-二甲氧基苯基)-N-(哌啶-4-基)喹唑啉-2-胺三氟乙酸盐的合成
Figure PCTCN2017096848-appb-000042
将中间体4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)哌啶-1-甲酸叔丁酯(62.6mg,0.12mmol,1.0eq)溶于乙醇(5mL)中,加入三氟乙酸(2.5mL),室温反应8h,TLC监测反应完全,直接浓缩溶液至干,得到6-(2,6-二氯-3,5-二甲氧基苯基)-N-(哌啶-4-基)喹唑啉-2-胺三氟乙酸盐(64.2mg,收率以 100%计)。
步骤3:1-(4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)哌啶-1-基)丙-2-烯-1-酮的合成:
Figure PCTCN2017096848-appb-000043
将中间体6-(2,6-二氯-3,5-二甲氧基苯基)-N-(哌啶-4-基)喹唑啉-2-胺三氟乙酸盐(64.2mg,0.12mmol,1.0eq)溶于THF(8mL)中,加入三乙胺(48.5mg,0.48mmol,4.0eq),搅拌反应30min,加入丙烯酰氯(12.7mg,0.14mmol,1.2eq),反应8h,TLC监测反应完全,加入饱和碳酸氢钠溶液(20mL),EA萃取(3×15mL),合并有机相,无水硫酸镁干燥,过滤,浓缩,粗品经硅胶柱层析(DCM:MeOH=100:1~40:1),得到1-(4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)哌啶-1-基)丙-2-烯-1-酮(40.0mg,收率:70%)。
1HNMR(400MHz,DMSO)δ(ppm):9.15(m,1H),7.47-7.66(m,3H),7.01(s,1H),6.82-6.89(m,1H),6.09-6.14(m,1H),5.66-5.70(m,1H),4.05-4.36(m,8H),2.90-2.96(m,1H),1.99(m,2H),1.44-1.47(m,2H)。
分子式:C24H24Cl2N4O3  分子量:487.38  LC-MS(m/z)=487.28[M+H+]。
实施例5:1-(反式-5-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-氮杂双环[2.1.1]己烷-2-基)丙-2-烯-1-酮(化合物11)的合成
Figure PCTCN2017096848-appb-000044
步骤1:反式-5-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-氮杂双环[2.1.1]己烷-2-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000045
将原料2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(500.0mg,1.35mol,1.0eq)溶解在NMP(5mL)中,加入反式-5-氨基-2-氮杂双环[2.1.1]己烷-2-羧酸叔丁酯(402.3mg,2.03mmol,1.5eq)和DIPEA(524.6mg,4.06mol,3.0eq),100℃加热反应4h,TLC监测反应完全,反应液冷却至室温,倒入冰水中,抽滤,收集滤饼,粗品经硅胶柱层析(200-300目硅胶,PE:EA=10:1~2:1),得到淡黄色固体反式-5-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-氮杂双环[2.1.1]己烷-2-甲酸叔丁酯(520mg,收率:72%)。
步骤2:反式-N-(6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)-2-氮杂双环[2.1.1]己烷-5-胺盐酸盐的合成
Figure PCTCN2017096848-appb-000046
将反式-5-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-氮杂双环[2.1.1]己烷-2-甲酸叔丁酯(510mg,0.96mmol,1.0eq)溶于乙醇(10mL)中,加入盐酸乙醇(10mL),室温反应8h,TLC监测反应完全,浓缩得到黄色固体反式-N-(6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)-2-氮杂双环[2.1.1]己烷-5-胺盐酸盐(480mg,收率:100%)。
步骤3:1-(反式-5-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-氮杂双环[2.1.1]己烷-2-基)丙-2-烯-1-酮(化合物11)的合成
Figure PCTCN2017096848-appb-000047
将反式-N-(6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)-2-氮杂双环[2.1.1]己烷-5-胺盐酸盐(480mg,1.03mmol,1.0eq)溶于THF(22mL)中,加入三乙胺 (415.3mg,4.10mmol,4.0eq),搅拌反应30min,加入丙烯酰氯(138.6mg,1.54mmol,1.5eq),反应8h,TLC监测反应完全。加入饱和碳酸氢钠溶液(50mL),EA萃取(3×30mL),合并有机相,无水硫酸镁干燥,过滤,浓缩,粗品经硅胶柱层析(200-300目硅胶,DCM:MeOH=100:1~40:1),得到白色固体1-(反式-5-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-氮杂双环[2.1.1]己烷-2-基)丙-2-烯-1-酮(350mg,收率:70%)。
1HNMR(400MHz,DMSO-d6)δ(ppm):9.15-9.17(d,1H),7.51-7.69(m,4H),7.00(s,1H),6.43-6.55(m,1H),6.07-6.12(m,1H),5.92-5.97(m,1H),5.65-5.68(m,1H),4.77-4.79(m,1H),4.12(m,1H),3.92(s,6H),3.83-3.85(m,1H),3.26-3.59(m,2H),2.96-3.01(m,1H),1.85-1.87(m,1H),1.21-1.23(m,1H)。
分子式:C24H22Cl2N4O3  分子量:485.37  LC-MS(m/z)=485.00[M+H+]。
实施例6:1-((3S,4R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-4-甲基吡咯烷-1-基)丙基-2-烯-1-酮(化合物20)的合成
Figure PCTCN2017096848-appb-000048
步骤1:(3S,4R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-4-甲基吡咯烷-1-羧酸叔丁酯的合成
Figure PCTCN2017096848-appb-000049
将原料2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(400.0mg,1.1mmol,1.0eq)和(3S,4R)-3-氨基-4-甲基吡咯烷-1-羧酸叔丁酯(440.6mg,2.2mmol,2.0eq)溶解在N-甲基吡咯烷酮(3.0mL)中,加入N,N-二异丙基乙胺(568.7mg,4.4mmol,4.0eq),逐渐升温至110℃反应过夜,TLC监测反应完全,将反应液降温至室温,加入15mL冰水,过滤,滤饼少量冰水洗涤,用二氯甲烷(5mL)溶解滤饼,加入无水硫酸镁干燥,过滤,浓缩,粗品硅胶柱层析(200-300目硅胶,PE:EA=10:1~3:1)得黄色固体(3S,4R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基) 喹唑啉-2-基)氨基)-4-甲基吡咯烷-1-羧酸叔丁酯(346.0mg,收率:59.0%)。
步骤2:6-(2,6-二氯-3,5-二甲氧基苯基)-N-((3S,4R)-4-甲基吡咯烷-3-基)喹唑啉-2-氨基盐酸盐的合成
Figure PCTCN2017096848-appb-000050
将(3S,4R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-4-甲基吡咯烷-1-羧酸叔丁酯(346.0mg,0.65mmol,1.0eq)溶解在乙醇(5.0mL)中,冰浴降温至0℃,加入氯化氢乙醇溶液(5.0mL)逐渐升温至室温,反应过夜,TLC监测反应完全。将反应液直接浓缩得到黄色固体6-(2,6-二氯-3,5-二甲氧基苯基)-N-((3S,4R)-4-甲基吡咯烷-3-基)喹唑啉-2-氨基盐酸盐(313.0mg粗品,收率:100%)。
步骤3:1-((3S,4R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-4-甲基吡咯烷-1-基)丙基-2-烯-1-酮(化合物20)的合成
Figure PCTCN2017096848-appb-000051
将6-(2,6-二氯-3,5-二甲氧基苯基)-N-((3S,4R)-4-甲基吡咯烷-3-基)喹唑啉-2-氨基盐酸盐(313.0mg,0.67mmol,1.0eq)溶解在四氢呋喃(10.0mL)中,加入三乙胺(203.0mg,2.0mmol,3.0eq),室温搅拌反应半小时,降温至0℃,缓慢滴加丙烯酰氯(122.4mg,1.35mmol,2.0eq),逐步升温至室温反应1小时,TLC监测反应完全,向反应液中加入饱和碳酸氢钠溶液(20mL),加入乙酸乙酯(8.0mL)分液,水相乙酸乙酯萃取(8.0mL×2),合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩,粗品硅胶柱层析(DCM:MeOH=125:1~80:1)得黄色固体1-((3S,4R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-4-甲基吡咯烷-1-基)丙基-2-烯-1-酮(160.5mg,收率49.2%)。
1HNMR(400MHz,DMSO)δ(ppm):9.18(s,1H),7.82(s,1H),7.79(s,1H),7.49-7.54(m,2H),7.02(s,1H),6.60-6.71(m,1H),6.16(d,1H),5.67(m,1H),4.12-4.20(m, 2H),4.03(s,6H),3.97(m,1H),3.24(m,1H),3.05(m,1H),2.48(m,1H),1.23(s,3H)。分子式:C24H24Cl2N4O3  分子量:487.39  LC-MS(Pos,m/z)=489.07[M+H+]。
实施例7:1-((2R,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-甲基吡咯烷-1-基)丙-2-烯-1-酮(化合物21)的合成
Figure PCTCN2017096848-appb-000052
步骤1:(2R,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-甲基吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000053
将2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(200mg,0.514mmol,1.0eq)、(2R,4S)-4-氨基-2-甲基吡咯烷-1-羧酸叔丁酯(162.63mg,0.812mmol,1.5eq)和N,N-二异丙基乙胺(139.85mg,1.082mmol,2.0eq)溶解在N-甲基吡咯烷酮(2mL)中,120℃加热反应过夜。TLC监测原料少量剩余,将反应液倒入冷水中(20mL),有褐色固体析出,抽滤,滤液用乙酸乙酯萃取(10mL×3),褐色固体用DCM溶解,合并有机相,用水洗涤(20mL×3),分液,有机相用无水硫酸钠干燥,过滤,滤饼用DCM淋洗,母液减压浓缩,粗品经硅胶柱层析(PE:EA=5:1~3:1)得固体(2R,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-甲基吡咯烷-1-甲酸叔丁酯(100mg,收率:35%)。
步骤2:6-(2,6-二氯-3,5-二甲氧基苯基)-N-((3S,5R)-5-甲基吡咯烷-3-基)喹唑啉-2-胺的合成
Figure PCTCN2017096848-appb-000054
将(2R,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-甲基吡咯烷-1-甲酸叔丁酯(100mg,0.187mmol,1.0eq)溶于盐酸乙醇(15mL)中,45℃加热反应3h。TLC监测反应完成,浓缩,粗品加入THF溶解,浓缩,重复三次,粗品不经纯化直接投入下一步。
步骤3:1-((2R,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-甲基吡咯烷-1-基)丙-2-烯-1-酮(化合物21)的合成
Figure PCTCN2017096848-appb-000055
将6-(2,6-二氯-3,5-二甲氧基苯基)-N-((3S,5R)-5-甲基吡咯烷-3-基)喹唑啉-2-胺(88mg,0.203mmol,1.0eq)溶于THF(15mL)中,加入三乙胺(102.71mg,1.015mmol,5.0eq)。降温至0℃,缓慢加入丙烯酰氯(18.37mg,0.203mmol,1.0eq),反应过夜。TLC监测反应完成,加入饱和碳酸氢钠溶液(10mL),分液,水相用EA萃取(10mL×3),分液,有机相用无水硫酸钠干燥,过滤,滤饼用EA淋洗,母液减压浓缩,粗品经制备薄层色谱分离(DCM:MeOH=15:1)得固体1-((2R,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-甲基吡咯烷-1-基)丙-2-烯-1-酮(21mg,收率:21%)。
1HNMR(400MHz,DMSO)δ(ppm):9.18(s,1H),7.82(s,1H),7.68(s,1H),7.49-7.57(m,2H),7.00(s,1H),6.54-6.58(m,1H),6.11-6.18(m,1H),5.63-5.67(m,1H),4.45-4.47(m,3H),4.11-4.13(m,6H),3.40-3.44(m,1H)。
分子式:C24H24Cl2N4O3  分子量:487.38  LC-MS(Neg,m/z)=487.1[M-H+]。
实施例8:1-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(羟甲基)吡咯烷)-1-丙基-2-烯-1-酮(化合物22)的合成
Figure PCTCN2017096848-appb-000056
步骤1:1-叔丁基2-甲基(2S,4S)-4-(((苄氧基)羰基)氨基)吡咯烷-1,2-二甲酸酯的合成
Figure PCTCN2017096848-appb-000057
将原料1-(叔丁基)2-甲基(2S,4S)-4-氨基吡咯烷-1,2-二甲酸酯盐酸盐(10.5g,36.8mmol,1.0eq)溶解在二氯甲烷(100mL)中,加入三乙胺(11.7g,115.8mmol,3.0eq),室温搅拌反应0.5h,冰浴降温至0℃,用恒压滴液漏斗缓慢滴加二氯甲烷溶解的氯甲酸苄酯(7.9g,46.3mmol,1.2eq),逐渐升温至室温反应过夜,TLC监测反应完全,向反应液中加入饱和碳酸氢钠溶液(100mL)分液,水相二氯甲烷(50mL×2)萃取,合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩,粗品经硅胶柱层析(200-300目硅胶,PE:EA=10:1~3:1)得无色油状物1-叔丁基2-甲基(2S,4S)-4-(((苄氧基)羰基)氨基)吡咯烷-1,2-二甲酸酯(3.6g,收率24.7%)。
步骤2:(2S,4S)-4-((((苄氧基)羰基)氨基)-2-(羟甲基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000058
将氢化铝锂(0.7g,19.0mmol,2.0eq)在0℃条件下溶于无水四氢呋喃(20mL)中,搅拌反应半小时,缓慢滴加溶有1-叔丁基2-甲基(2S,4S)-4-(((苄氧基)羰基)氨基)吡咯烷-1,2-二甲酸酯(3.6g,9.5mmol,1.0eq)的四氢呋喃溶液,逐渐升温至室温反应2小时,TLC检测反应完全,将反应液降温至0℃,向反应液中缓慢滴加水(0.7mL)和10%的氢氧化钠溶液(0.7mL),补加水(2.1mL),搅拌半小时,将反应液过滤,滤液浓缩,粗品经硅胶柱层析(200-300目,PE:EA=10:1~2:1)得无色油状物(2S,4S)-4-((((苄氧基)羰基)氨基)-2-(羟甲基)吡咯烷-1-羧酸叔丁酯(1.05g,收率30.0%)。
步骤3:(2S,4S)-4-氨基-2-(羟甲基)吡咯烷-1-羧酸叔丁酯的合成
Figure PCTCN2017096848-appb-000059
将(2S,4S)-4-((((苄氧基)羰基)氨基)-2-(羟甲基)吡咯烷-1-甲酸叔丁酯(250mg,0.71mmol,1.0eq)溶解在甲醇(5.0mL)中,加入钯碳(75.2mg,7.2mmoL,0.01eq)。置换氢气4次,室温搅拌反应过夜,TLC监测反应完全,将反应液垫上硅藻土抽滤,滤液浓缩得无色透明油状物(2S,4S)-4-氨基-2-(羟甲基)吡咯烷-1-甲酸叔丁酯(123.0mg,收率80.2%)。
步骤4:(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉)-2-氨基)-2-(羟甲基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000060
将原料(2S,4S)-4-氨基-2-(羟甲基)吡咯烷-1-羧酸叔丁酯(123.0mg,0.57mmol,1.0eq)和2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(231.5mg,0.63mmol,1.1eq)溶解在N-甲基吡咯烷酮(4.0mL)中,加入N,N-二异丙基乙胺(147.3mg,1.14mmoL,2.0eq),逐渐升温至110℃,搅拌反应5小时,TLC监测反应完全,将反应液降温至室温,向反应液中加入20mL冰水,搅拌10分 钟,过滤,滤饼少量冰水洗涤后二氯甲烷(10mL)溶解,无水硫酸钠干燥,过滤,浓缩,粗品经硅胶柱层析(200-300目,DCM:MeOH=100:1~50:1)得淡棕色固体(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(羟甲基)吡咯烷-1-羧酸叔丁酯(113.0mg,收率36.1%)。
步骤5:((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉)-2-氨基)吡咯烷)-2-甲醇盐酸盐的合成
Figure PCTCN2017096848-appb-000061
将原料(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(羟甲基)吡咯烷-1-羧酸叔丁酯(113.0mg,0.21mmol,1.0eq)溶解在乙醇(5.0mL)中,冰浴降温至0℃,加入氯化氢乙醇溶液(5.0mL),逐渐升温至室温反应过夜,TLC监测反应完全。将反应液直接浓缩得到黄色固体((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉)-2-氨基)吡咯烷)-2-甲醇盐酸盐(180.0mg粗品,收率:以100%计)。
步骤6:1-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(羟甲基)吡咯烷)-1-丙基-2-烯-1-酮(化合物22)的合成
Figure PCTCN2017096848-appb-000062
将原料((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉)-2-氨基)吡咯烷)-2-甲醇盐酸盐(160.0mg,0.35mmol,1.0eq)溶解在四氢呋喃(10.0mL)中,加入三乙胺(106.0mg,1.1mmol,3.0eq),室温搅拌反应半小时,降温至0℃,缓慢滴加丙烯酰氯(38.0mg,0.42mmol,1.2eq)逐步升温至室温反应1小时,TLC监测反应完全,向反应液中加入饱和碳酸氢钠溶液(15mL),加入乙酸乙酯(8mL),分液,水相乙酸乙酯萃取(8mL×2),合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩,粗品硅胶柱层析(200-300目硅胶,DCM:MeOH=100:1~50:1)得淡黄色固体1-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧 基苯基)喹唑啉-2-基)氨基)-2-(羟甲基)吡咯烷-1-基)丙基-2-烯-1-酮(75.0mg,收率42.5%)。
1HNMR(400MHz,DMSO)δ(ppm):9.18(s,1H),7.89(s,1H),7.80(s,1H),7.55(d,1H),7.52(d,1H),7.00(s,1H),6.60-6.71(m,1H),6.16(d,1H),5.67(d,1H),5.09(s,1H),4.55(m,1H),4.12-4.20(m,2H),4.03(s,6H),3.97(m,1H),3.74(m,1H),3.52(m,1H),1.98(m,2H)。
分子式:C24H24Cl2N4O4  分子量:503.39  LC-MS(Pos,m/z)=505.40[M+H+]。
实施例9:1-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基-2-(氟甲基)吡咯烷-1-基)丙-2-烯-1-酮(化合物24)的合成
Figure PCTCN2017096848-appb-000063
步骤1:(2S,4R)-4-((四氢-2H-吡喃-2-基)氧基)吡咯烷-1,2-二羧酸1-(叔丁基)-2-甲酯的合成
Figure PCTCN2017096848-appb-000064
将原料(2S,4R)-4-羟基吡咯烷-1,2-二羧酸1-(叔丁基)-2-甲酯(10.0g,40.8eq)溶解在叔丁基甲醚(100mL)中,加入对甲基苯磺酸(234.2mg,1.36mmol),冰浴冷却至0℃,缓慢滴加3,4-二氢-2H-吡喃(10.3g,122.4mmol),缓慢升至室温反应3h,TLC监测反应完全,减压浓缩得(2S,4R)-4-((四氢-2H-吡喃-2-基)氧基)吡咯烷-1,2-二羧酸-1-(叔丁基)-2-甲酯(14g)。
步骤2:(2S,4R)-2-(羟基甲基)-4–((四氢-2H-吡喃-2-基)氧基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000065
将四氢铝锂(3.1g,81.6mmoL)加入到四氢呋喃(100.0mL)中,乙醇,水,干冰冷却至-20℃,滴加(2S,4R)-4-((四氢-2H-吡喃-2-基)氧基)吡咯烷-1,2-二羧酸1-(叔丁基)-2-甲酯(13.43g,40.8mmoL)的四氢呋喃溶液(100.0mL)。氮气保 护下,-20℃搅拌反应三小时,TLC监测反应完全。冰浴下向反应液中加入水(3.1mL),10%NaOH(3.1mL),水(9.3mL),搅拌十分钟,硅藻土抽滤,滤液直接浓缩至干(2S,4R)-2-(羟基甲基)-4-((四氢-2H-吡喃-2-基)氧基)吡咯烷-1-甲酸叔丁酯(13.1g粗品,收率100%)。
步骤3:(2S,4R)-2-(((甲基磺酰基)氧基)甲基)-4-((四氢-2H-吡喃-2-基)氧基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000066
将原料(2S,4R)-2-(羟基甲基)-4-((四氢-2H-吡喃-2-基)氧基)吡咯烷-1-甲酸叔丁酯(2.0g,6.65mmoL)溶解在二氯甲烷(10.0mL)中,冰浴冷却至0℃,加入三乙胺(1.35g,13.3mmoL),0℃下缓慢滴加甲基磺酰氯(1.15g,9.975mmoL)。缓慢升至室温1小时,TLC监测.加入二氯甲烷(10mL),加入饱和氯化铵(10mL)分层,合并有机相,有机相用水(3*5mL)洗,饱和食盐水洗涤,无水硫酸钠干燥,过柱(PE:EA=15:1,10:1,DCM:MeOH=100:1),得(2S,4R)-2-(((甲基磺酰基)氧基)甲基)-4-((四氢-2H-吡喃-2-基)氧基)吡咯烷-1-甲酸叔丁酯(2.0g,收率79.3%)。
步骤4:(2S,4R)-2-(氟甲基)-4-((四氢-2H-吡喃-2-基)氧基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000067
将原料(2S,4R)-2-(((甲基磺酰基)氧基)甲基)-4-((四氢-2H-吡喃-2-基)氧基)吡咯烷-1-甲酸叔丁酯(7.5g,19.8mmoL)溶解在四氢呋喃(45.0mL)中,加入四甲基氟化铵(1.5g,4.75mmoL)加热至80℃,回流监测。TLC监测反应完全.将反应液减压旋干,加入乙酸乙酯(10mL),用水洗(5*10mL),合并有机相,无水硫酸钠干燥,过柱(DCM:MeOH=200:1,150:1,100:1),得(2S,4R)-2-(((甲基磺酰基)氧基)甲基)-4-((四氢-2H-吡喃-2-基)氧基)吡咯烷-1-甲酸叔丁酯(3.2g,收率 53.3%)。
步骤5:(2S,4R)-2-(氟甲基)-4-羟基吡咯烷-1-羧酸叔丁酯的合成
Figure PCTCN2017096848-appb-000068
将原料((2S,4R)-2-(氟甲基)-4-((四氢-2H-吡喃-2-基)氧基)吡咯烷-1-甲酸叔丁酯(3.2g,10.55mmoL)溶解在乙酸(33.0mL),四氢呋喃(33.0mL),水(33.0mL)的混合溶剂中,加热回流监测,TLC监测反应完全。用饱和碳酸氢钠调节PH值(8~9),将四氢呋喃旋去,水相乙酸乙酯(3*50mL),合并有机相,用水洗(1*10mL),饱和食盐水洗,无水硫酸钠干燥,过柱(DCM:MeOH=200:1,150:1,100:1,50:1),得(2S,4R)-2-(氟甲基)-4-羟基吡咯烷-1-甲酸叔丁酯(2.0g,收率87%)。步骤6:(2S,4S)-4-叠氮基-2-(氟甲基)吡咯烷-1-羧酸叔丁酯的合成
Figure PCTCN2017096848-appb-000069
将原料(2S,4R)-2-(氟甲基)-4-羟基吡咯烷-1-羧酸叔丁酯(100.0mg,0.46mmoL)溶解在四氢呋喃(1.5mL),冰浴至0℃,加入三苯基膦(144.8mg,0.552mmoL),0℃下加入偶氮二甲酸二异丙酯(111.6mg,0.552mmoL),0℃下搅拌20分钟,0℃下缓慢滴加入DDPA(151.9mg,0.552mmoL),缓慢升至室温过夜.TLC监测反应完全.将四氢呋喃旋去,过柱(PE:EA=5:1,2:1),得(2S,4S)-4-叠氮基-2-(氟甲基)吡咯烷-1-甲酸叔丁酯(170.0mg,收率100%)。
步骤7:(2S,4S)-4-氨基-2-(氟甲基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000070
将原料(2S,4S)-4-叠氮基-2-(氟甲基)吡咯烷-1-甲酸叔丁酯(112.3mg,0.46mmoL)溶解在甲醇(2mL),加入钯碳(5.62mg,5%wt),置换氢气三次。加氢过夜.TLC监测反应完全.用硅藻土抽滤,滤液旋干得(2S,4S)-4-氨基-2-(氟甲基)吡咯烷-1甲酸叔丁酯(105.0mg).
步骤8:(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(氟甲 基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000071
将原料(2S,4S)-4-氨基-2-(氟甲基)吡咯烷-1-甲酸叔丁酯(100.3mg,0.46mmol)溶解在N-甲基吡咯烷酮(2.0mL)中,加入N,N-二异丙基乙胺(178.21mg,1.38mmol),2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(203.1mg,0.552mmoL),逐渐升温至110℃加热回流过夜,TLC监测反应完全,将反应液降温至室温,加入10mL水,过滤,滤饼少量冰水洗涤,用二氯甲烷(10mL)溶解滤饼,加入无水硫酸钠干燥,制备板提纯(DCM:MeOH=20:1)得(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(氟甲基)吡咯烷-1-甲酸叔丁酯(50.0mg,19.8%)。
步骤9:6-(2,6-二氯-3,5-二甲氧基苯基)-N-((3S,5S)-5-(氟甲基)吡咯烷-3-基)喹唑啉-2-胺的合成
Figure PCTCN2017096848-appb-000072
将原料(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(氟甲基)吡咯烷-1-甲酸叔丁酯(45.0mg,0.082mmoL)溶解二氯甲烷(2.0mL)中,冰浴降温至0℃,加入氯化氢乙醇溶液(1.0mL),缓慢升至室温反应2小时,TLC监测反应完全。将反应液直接浓缩至干得6-(2,6-二氯-3,5-二甲氧基苯基)-N-((3S,5S)-5-(氟甲基)吡咯烷-3-基喹唑啉-2-胺(40.0mg)。
步骤10:1-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(氟甲基)吡咯烷-1-基)丙-2-烯-1-酮
Figure PCTCN2017096848-appb-000073
将原料6-(2,6-二氯-3,5-二甲氧基苯基)-N-((3S,5S)-5-(氟甲基)吡咯烷-3-基)喹唑啉-2-胺(37.0mg,0.082mmoL)溶解在四氢呋喃(1.0mL)中,冰浴反应至0℃, 加入三乙胺(24.0mg,0.246mmol),0℃下加入丙烯酰氯(11.13mg,0.123mmoL),缓慢升至室温反应两小时,TLC监测反应完全。加入水(5mL),水相用乙酸乙酯萃取(8.0mL*3),合并有机相,有机相用饱和食盐水洗涤,无水硫酸钠干燥,过柱(DCM:MeOH=250:1,200:1,150:1,100:1)得1-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(氟甲基)吡咯烷-1-基)丙-2-烯-1-酮(11.5mg,收率27.7%)。
1HNMR(400MHz,DMSO-d6)δ(ppm):11.93(s,1H),9.19(s,1H),7.85-7.83(d,1H),7.69(s,1H),7.58-7.50(m,2H),7.01(s,1H),6.59(s,2H),6.19-6.15(d,1H),5.71-5.69(d,1H),5.31(s,1H),4.65-4.54(d,4H),4.44(s,1H),4.27-4.19(d,3H),3.97(s,6H),3.81-3.75(d,2H),2.19-2.16(t,2H),2.10-1.98(m,3H)。
分子式:C24H22Cl2N4O3分子量:485.37LC-MS(Pos,m/z)=485.43[M+H+]。
实施例10:2-((2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)乙腈(化合物27)的合成
Figure PCTCN2017096848-appb-000074
步骤1:(2S,4S)-2-(溴甲基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000075
将原料(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(羟甲基)吡咯烷-1-甲酸叔丁酯(1571mg,2.859mmol,1.0eq)溶解在DCM(20mL)中,搅拌,加入四溴化碳(1185mg,3.574mmol,1.25eq),冷却至0℃,分批加入三苯基膦(2250mg,8.577mmol,3eq),氮气保护,反应体系缓慢升至室温。反应过夜。TLC监测反应完全,反应液直接加入硅胶并旋去溶剂,柱层析分离(洗脱剂PE:EA=3:1)得到(2S,4S)-2-(溴甲基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(800mg,收率45.7%)。
步骤2:(2S,4S)-2-(氰基甲基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000076
将中间体(2S,4S)-2-(溴甲基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(800mg,1.3064mmol,1eq)溶于乙腈(10mL)中,室温下加入TMSCN(259mg,2.6128mmol,2eq),适量四乙基氟化铵,升至60℃反应过夜,TLC监测反应完全,冷却反应液,加入100ml水,DCM(200ml×3)萃取反应液,分液,有机相用无水硫酸钠干燥,过滤,减压浓缩至干,柱层析分离(洗脱剂PE:EA=5:1-4:1),得到(2S,4S)-2-(氰基甲基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(215mg,收率29.5%)。
步骤3:2-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)乙腈的合成
Figure PCTCN2017096848-appb-000077
将中间体(2S,4S)-2-(氰基甲基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(215mg)溶于DCM(4mL)中,冷却至0℃,加入TFA(3ml)。之后缓慢升至室温,TLC监测反应完毕,室温减压浓缩至干,加入DCM减压浓缩,将体系TFA尽量除去。得到2-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)乙腈(理论产量176mg),直接用于下步反应。
步骤4:2-((2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)乙腈的合成
Figure PCTCN2017096848-appb-000078
将中间体2-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)乙腈(按176mg计,0.3840mmol)溶于THF(4mL)中,加入TEA,冷却至-10℃,将丙烯酰氯(41mg,0.4608mmol,1.2eq,溶于1ml DCM)溶液缓慢滴加到反应体系,TLC监测反应完全,冷却下加入饱和碳酸氢钠溶液,使反应体系呈碱性,DCM萃取(100mL×3),分液,合并有机相,无水硫酸钠干燥,过滤,滤液浓缩,柱层析(DCM:MeOH=200:1-50:1)分离,得到2-((2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)乙腈(138mg,收率:70.4%)。
1HNMR(400MHz,DMSO)δ(ppm):9.21(s,1H),7.98-8.00(d,1H),7.70-7.71(d,1H),7.52-7.61(m,2H),7.02(s,1H),6.60-6.67(q,1H),6.18-6.23(q,1H),5.72-5.76(t,1H),4.57-4.59(t,1H),4.51-4.53(d,1H),4.24-4.25(d,2H),3.98(s,3H),3.48-3.50(d,3H),3.42-3.43(d,2H),3.13-3.22(m,1H),3.01-3.10(m,1H),1.98-2.05(q,1H)。
分子式:C25H23Cl2N5O3,分子量:512.39,LC-MS(Pos,m/z)=512.1[M+H+]。
实施例11:(2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲腈(化合物28)的合成
Figure PCTCN2017096848-appb-000079
步骤1:1-叔丁基2-甲基(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1,2-二羧酸酯的合成
Figure PCTCN2017096848-appb-000080
将原料2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(500mg,1.353mmol,1.0eq)、1-叔丁基2-甲基(2S,4S)-4-氨基吡咯烷-1,2-二羧酸酯盐酸盐(644mg,2.030mmol,1.5eq)、N,N-二异丙基乙胺(874.4mg,6.765mmol,5.0eq)溶解在N-甲基吡咯烷酮(5mL)中,120℃加热反应过夜,第二天早上TLC监测反应完全,将反应液倒入冷水中(20mL),有褐色固体析出,抽滤,滤液用乙酸乙酯萃取(10mL×3),褐色固体用乙酸乙酯溶解,合并有机相,用水洗涤(20mL×3),分液,有机相用无水硫酸钠干燥,过滤,滤饼用乙酸乙酯淋洗,母液减压浓缩,粗品经硅胶柱层析(PE:EA=5:1)得固体1-叔丁基2-甲基(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1,2-二羧酸酯(384mg,收率:49%)。
步骤2:(2S,4S)-1-(叔丁氧羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲酸的合成
Figure PCTCN2017096848-appb-000081
将1-叔丁基2-甲基(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1,2-二羧酸酯(300mg,0.5195mmol,1.0eq)溶于甲醇(5mL)中,搅拌溶解,降温至0℃,滴加一水合氢氧化锂(65mg,1.558mmol,3.0eq)的水(1mL)溶液,反应3h后,TLC监测原料剩余,补加一水合氢氧化锂(65mg,1.558mmol,3.0eq)的水(1mL)溶液,加甲醇至体系澄清,反应过夜。TLC监测反应完成,减压浓缩,加水(10mL)和甲基叔丁基醚(10mL),搅拌,分液,水相加乙酸乙酯(20mL),降温至0℃,用柠檬酸调pH至5-6,分出有机相,水相用乙酸乙酯(10mL)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤饼用乙酸乙酯淋洗,母液减压浓缩得淡黄色固体(2S,4S)-1-(叔丁氧羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲酸(260mg,收率:89%)。
步骤3:(2S,4S)-2-氨基甲酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000082
将(2S,4S)-1-(叔丁氧羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲酸(260mg,0.462mmol,1.0eq)溶于THF(10mL)中,加入三乙胺(140.1mg,1.384mmol,3.0eq),降温至-15℃左右,缓慢加入氯甲酸异丁酯(75.64mg,0.554mmol,1.2eq)。反应2h后,TLC监测反应完成,将反应液滴加至降温至0℃的氨水溶液(5mL)中,反应3h后,TLC监测反应完成。加入饱和碳酸钾溶液(10mL)、饱和食盐水(10mL)、MTBE(20mL),搅拌分液,水相用MTBE萃取(10mL),合并有机相,干燥,浓缩得淡黄色固体(2S,4S)-2-氨基甲酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(350mg粗品)。
步骤4:(2S,4S)-2-氰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡 咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000083
将(2S,4S)-2-氨基甲酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(250mg,0.445mmol,1.0eq)溶于THF(10mL)中,加入三乙胺(112.6mg,1.113mmol,2.5eq),降温至0℃左右,缓慢加入TFAA(186.9mg,0.89mmol,2.0eq),0.5h后升温至40℃,反应过夜,TLC监测原料有剩余,补加TFAA(93.46mg,0.445mmol,1.0eq)继续反应1h,TLC监测反应完成。反应液降温,加DCM(20mL)饱和碳酸氢钠溶液(10mL),搅拌,分液,分出有机相,用无水硫酸钠干燥,过滤,滤饼用DCM淋洗,母液减压浓缩得固体(2S,4S)-2-氰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(350mg粗品)。
步骤5:(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲腈的合成
Figure PCTCN2017096848-appb-000084
将中间体(2S,4S)-2-氰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(350mg,0.445mmol,1.0eq)溶于DCM(5mL)中,加入三氟乙酸(5mL),加热至30℃反应4h。TLC监测反应完成,浓缩,粗品加EA,浓缩,重复三次。加入MTBE(20mL),有固体析出,抽滤得固体(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲腈(143mg,收率:50%)。
步骤6:(2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲腈(化合物28)的合成
Figure PCTCN2017096848-appb-000085
将(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲腈(143mg,0.322mmol,1.0eq)溶于THF(10mL)中,加入三乙胺(162.81mg,1.61mmol,5.0eq),降温至0℃,缓慢加入丙烯酰氯(29.12mg,0.322mmol,1.0eq),4h后TLC监测反应完成,加入饱和碳酸氢钠溶液(10mL),分液,水相用EA萃取(10mL×3),合并有机相,浓缩,粗品通过厚制备硅胶板分离(PE:EA=2:1)得固体(2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲腈(10mg,收率:6%)。
1HNMR(400MHz,DMSO)δ(ppm):9.04(s,1H),7.72(m,1H),7.60-7.70(m,2H),6.65(s,1H),6.51-6.55(m,1H),6.42-6.51(m,1H),5.81-5.84(m,1H),5.65(s,1H),5.30-5.36(m,1H),4.90(s,2H),4.13-4.16(m,1H),4.01(s,6H),3.98(m,1H),2.74(m,1H),2.50(m,1H),2.20(m,1H),2.00(s,2H),1.63(m,1H)。
分子式:C24H21Cl2N5O3  分子量:498.36  LC-MS(Neg,m/z)=498.1[M-H+]。
实施例12:(2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-N,N-二甲基吡咯烷-2-甲酰胺(化合物30)的合成
Figure PCTCN2017096848-appb-000086
步骤1:1-叔丁基2-甲基(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1,2-二羧酸酯的合成
Figure PCTCN2017096848-appb-000087
将原料2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(500mg,1.353mmol,1.0eq)、1-叔丁基2-甲基(2S,4S)-4-氨基吡咯烷-1,2-二羧酸酯盐酸盐(644mg,2.030mmol,1.5eq)和N,N-二异丙基乙胺(874.4mg,6.765mmol,5.0eq)溶解在N-甲基吡咯烷酮(5mL)中,120℃加热反应过夜,TLC监测反应完全,将反应液倒入冷水中(20mL),有褐色固体析出,抽滤,滤液用乙酸乙酯萃取(10mL×3),褐色固体用乙酸乙酯溶解,合并有机相,用水洗涤(20mL×3),用无水硫酸钠干燥,过滤,滤饼用乙酸乙酯淋洗,母液减压浓缩,粗品经硅胶柱层析(PE:EA=5:1)得固体1-叔丁基2-甲基(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1,2-二羧酸酯(384mg,收率:49%)。
步骤2:(2S,4S)-1-(叔丁氧基羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲酸的合成
Figure PCTCN2017096848-appb-000088
将中间体1-叔丁基2-甲基(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1,2-二羧酸酯(300mg,0.520mmol,1.0eq)溶于甲醇(5mL)中,搅拌溶解,降温至0℃,滴加一水合氢氧化锂(65mg,1.558mmol,3.0eq)的水(1mL)溶液,反应3h后,TLC监测原料剩余,补加一水合氢氧化锂(65mg,1.558mmol,3.0eq)的水(1mL)溶液,加甲醇至体系澄清,反应过夜。TLC监测反应完全,减压浓缩,加水(10mL)和甲基叔丁基醚(10mL)搅拌分液,水相加乙酸乙酯(20mL),降温至0℃,用柠檬酸调pH至5-6,分出有机相,水相用乙酸乙酯(10mL)萃取,合并有机相,减压浓缩得淡黄色固体(2S,4S)-1-(叔丁氧基羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲酸(260mg,收率:89%)。
步骤3:(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(二甲基 氨基甲酰基)吡咯烷-1-羧酸叔丁酯的合成
Figure PCTCN2017096848-appb-000089
将中间体(2S,4S)-1-(叔丁氧羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲酸(200mg,0.355mmol,1.0eq)溶于THF(15mL)中,加入三乙胺(107.8mg,1.065mmol,3.0eq)。降温至-15℃左右缓慢加入THF稀释的氯甲酸异丁酯(48.49mg,0.355mmol,1.0eq)。反应1h后,TLC监测反应完成,将反应液滴加至降温至0℃的二甲胺/四氢呋喃溶液(5mL)中,反应4h后,TLC监测反应完成。加饱和碳酸钾溶液(10mL)、饱和食盐水(10mL)、EA(20mL),搅拌分液,水相用EA萃取(10mL×2),合并有机相,干燥浓缩,粗品不经纯化投入下一步。
步骤4:(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-N,N-二甲基吡咯烷-2-甲酰胺的合成
Figure PCTCN2017096848-appb-000090
将中间体(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(二甲基氨基甲酰基)吡咯烷-1-羧酸叔丁酯(上一步所得粗品)溶于DCM(10mL)中,加入三氟乙酸(10mL)加热至30℃,反应过夜。第二天早上TLC监测反应完成,减压浓缩。粗品加入乙酸乙酯,浓缩,重复三次,加入MTBE(10mL),有固体析出,抽滤得固体(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-N,N-二甲基吡咯烷-2-甲酰胺(60mg,收率:34%)。
步骤5:(2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-N,N-二甲基吡咯烷-2-甲酰胺(化合物30)的合成
Figure PCTCN2017096848-appb-000091
将中间体(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨 基)-N,N-二甲基吡咯烷-2-甲酰胺(60mg,0.122mmol,1.0eq)溶于THF(10mL)中,加入三乙胺(61.73mg,0.61mmol,5.0eq),降温至0℃,缓慢加入THF稀释的丙烯酰氯(11.04mg,0.122mmol,1.0eq),2h后TLC监测反应完成,加入饱和碳酸氢钠溶液(10mL),分液,水相用EA萃取(10mL×3),合并有机相,浓缩,通过厚制备硅胶板(DCM:MeOH=15:1)分离得固体(2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-N,N-二甲基吡咯烷-2-甲酰胺(7mg,收率:10%)。
1HNMR(400MHz,DMSO)δ(ppm):9.17(s,1H),7.81(s,1H),7.69(s,1H),7.50-7.58(m,2H),7.00(s,1H),6.62-6.67(m,1H),6.10-6.14(m,1H),5.66-5.69(m,1H),5.32(s,1H),4.91(m,1H),4.75(m,1H),4.14-4.16(s,1H),3.97(s,6H),3.11(s,1H),3.11-3.32(m,2H),2.85(s,3H),2.00(m,2H),1.23(s,6H),1.10(s,2H)。
分子式:C26H27Cl2N5O4  分子量:544.43  LC-MS(Neg,m/z)=544.2[M-H+]。
实施例13:1-((2R,4R)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(2-羟丙基-2-基)吡咯烷-1-基)丙-2-烯-1-酮(化合物31)的合成
Figure PCTCN2017096848-appb-000092
步骤1:(2R,4R)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1,2-二羧酸1-(叔丁基)-2-甲酯的合成
Figure PCTCN2017096848-appb-000093
将原料2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(500mg,1.353mmol,1.0eq),(2R,4R)-4-氨基吡咯烷-1,2-二羧酸-1-(叔丁基)-2-甲酯盐酸盐(644mg,2.030mmol,1.5eq),N,N-二异丙基乙胺(874.4mg,6.765mmol,5.0eq)溶解在N-甲基吡咯烷酮(5mL)中,120℃加热反应16h,TLC监测反应完全,将反应液倒入冷水中(20mL),有褐色固体析出,抽滤,滤液用乙酸乙酯萃取(10mL×3),褐色固体用乙酸乙酯溶解,合并有机相,用水洗涤(20mL×3),用无水硫酸钠 干燥后,减压浓缩,柱层析(PE:EA=5:1)得(2R,4R)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1,2-二羧酸-1-(叔丁基)-2-甲酯(384mg,收率:49%)。
步骤2:(2R,4R)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(2-羟丙基-2-基)-吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000094
将(2R,4R)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1,2-二羧酸1-(叔丁基)-2-甲酯(300mg,0.5195mmol,1.0eq)溶于THF(5mL)中,搅拌溶清,降温至0℃,滴加3M甲基氯化镁(0.8mL,2.5975mmol,5.0eq)的THF溶液,0℃反应1h后,TLC监测无原料,加饱和氯化铵溶液(5mL),用二氯甲烷(20mL×3)萃取,分液,水相弃置,合并有机相,用无水硫酸干燥,过滤,母液减压浓缩得粗品,过柱,洗脱剂甲醇:二氯甲烷=1:50,收集所要点,减压浓缩得(2R,4R)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(2-羟丙基-2-基)-吡咯烷-1-甲酸叔丁酯(150mg,收率:50%)。
步骤3:2-((2R,4R)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-吡咯烷-2-基)丙-2-醇的合成
Figure PCTCN2017096848-appb-000095
将(2R,4R)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(2-羟丙基-2-基)-吡咯烷-1-甲酸叔丁酯(150mg,0.2597mmol,1.0eq)溶于甲醇(5mL)中,搅拌溶清降温0℃,滴加入35%盐酸乙醇溶液(5mL),室温搅拌2h,TLC监测无原料,浓缩,加入THF(50mL)溶解,浓缩,重复三次,得到2-((2R,4R)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-吡咯烷-2-基)丙-2-醇。按理论量直接进行下一步反应。
步骤4:1-((2R,4R)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(2-羟丙基-2-基)吡咯烷-1-基)丙-2-烯-1-酮的合成:
Figure PCTCN2017096848-appb-000096
将2-((2R,4R)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-吡咯烷-2-基)丙-2-醇(123mg,0.2597mmol,1.0eq)溶于THF(10mL)中,加入三乙胺(131mg,1.2985mmol,5.0eq)降温至0℃,缓慢加入丙烯酰氯(15.7mg,0.2597mmol,1.0eq),4h后TLC监测反应完成,加入饱和碳酸氢钠溶液(10mL),分液,水相用EA萃取(10mL×3),合并有机相浓缩。通过厚制备板分离(PE:EA=2:1),得1-((2R,4R)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(2-羟丙基-2-基)吡咯烷-1-基)丙-2-烯-1-酮(8mg,收率:6%)。
1HNMR(400MHz,DMSO)δ(ppm):9.18(s,1H),7.69(s,1H),7.50-7.57(d,3H),7.01(s,1H),6.21(m,1H),6.17(m,1H),5.76(m,1H),4.44(s,6H),4.24(m,1H),3.51(m,1H),3.22-3.41(m,2H),2.67-2.70(m,2H),1.24(s,6H)。
分子式:C26H28Cl2N4O4分子量:531.43LC-MS(Pos,m/z)=531.1[M-H+]。
实施例14:N-(((2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)甲基)乙酰胺(化合物32)的合成
Figure PCTCN2017096848-appb-000097
步骤1:(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-((1,3-二氧代异吲哚啉-2-基)氨基)甲基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000098
将(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(羟甲基)吡咯烷-1-甲酸叔丁酯(550.0mg,1.0mol,1.0eq)溶解在四氢呋喃(15mL)中,加入三苯基膦(918.0mg,3.5mmol,1.5eq)和邻苯二甲酰亚胺(206.1mg,1.4mmol,1.4eq),冰浴降温至0℃。0℃下加入偶氮二甲酸二乙酯(523.0mg,3.0mmol),缓慢升至室温过夜。TLC监测反应完全,用H2O(2mL)淬灭反应,将溶剂旋去,加入H2O(5mL)和乙酸乙酯(20mL),分出有机相,浓缩,粗品经硅胶柱层析(DCM:MeOH=100:1~40:1),得黄色胶状固体(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-((1,3-二氧代异吲哚啉-2-基)氨基)甲基)吡咯烷-1-甲酸叔丁酯(600.0mg,收率:88.6%)。
步骤2:(2S,4S)-2-(氨基甲基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000099
将(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-((1,3-二氧代异吲哚啉-2-基)氨基)甲基)吡咯烷-1-甲酸叔丁酯(600mg,0.88mmol,1.0eq)溶于乙醇(12mL)中,加入水合肼(6mL),室温反应2h,TLC监测反应完全,加入H2O(6mL),将反应液浓缩,水相用乙酸乙酯萃取,合并有机相,无水硫酸钠干燥,粗品经硅胶柱层析(DCM:MeOH=100:1~40:1)得黄色胶状固体(2S,4S)-2-(氨基甲基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(500.0mg,收率:100%)。
步骤3:(2S,4S)-2-(乙酰氨基甲基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000100
将(2S,4S)-2-(氨基甲基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(150.0mg,0.274mmol,1.0eq)溶于DCM(3mL)中,冰 浴降温至0℃,加入三乙胺(55.7mg,0.55mmol)和乙酸酐(30.61mg,0.3mmol),缓慢升至室温过夜,TLC监测反应完全,向反应液中加入二氯甲烷(5mL),有机相用饱和氯化铵洗,水洗,饱和食盐水洗,无水硫酸镁干燥,过滤,浓缩得到白色胶状固体(2S,4S)-2-(乙酰氨基甲基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(100.0mg,收率:62.1%)。
步骤4:N-(((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)甲基)乙酰胺的合成
Figure PCTCN2017096848-appb-000101
将(2S,4S)-2-(乙酰氨基甲基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(100.0mg,0.17mmol,1.0eq)溶于DCM(4mL)中,冰浴降温至0℃,0℃下加入三氟乙酸(2mL),缓慢升至室温过夜,搅拌2小时,TLC监测反应完全,浓缩至干得到黄色胶体N-(((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)甲基)乙酰胺(136.0mg粗品)。
步骤5:N-(((2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)甲基)乙酰胺(化合物32)的合成
Figure PCTCN2017096848-appb-000102
将N-(((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)甲基)乙酰胺(134.0mg粗品,0.17mmol,1.0eq)溶于DCM(2mL)中,冰浴降温至0℃,0℃下加入三乙胺(83.2mg,0.822mmol),0℃下加入丙烯酰氯(37.2mg,0.41mmol),缓慢升至室温,搅拌2小时,TLC监测反应完全,浓缩,粗品经硅胶柱层析(DCM:MeOH=200:1~100:1)得白色固体N-(((2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)甲基)乙酰胺(20.0mg,收率:13.3%)。
1HNMR(400MHz,CDCl3)δ(ppm):9.19(s,1H),8.14-7.91(m,2H),7.89(s,1H), 7.70-7.51(m,2H),7.01(s,1H),6.80-6.55(m,2H),6.23-6.17(t,1H),5.72-5.66(t,1H),4.48-4.45(t,1H),4.18-4.10(m,2H),4.08(s,6H),3.53-3.49(m,1H),3.21-3.11(m,1H),2.33-3.32(d,1H),2.01-1.99(d,1H),1.85-1.81(d,3H)。
分子式:C26H27Cl2N5O4  分子量:544.43  LC-MS(Pos,m/z)=544.2[M+H+]。
实施例15:N-(((2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)甲基)甲磺酰胺(化合物33)的合成
Figure PCTCN2017096848-appb-000103
步骤1:(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(甲基磺酰氨基甲基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000104
将中间体(2S,4S)-2-(氨基甲基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸叔丁酯(150.0mg,0.274mmol,1.0eq)溶于DCM(3mL)中,冰浴降温至0℃,加入三乙胺(55.7mg,0.55mmol),0℃下加入甲磺酰氯(36.9mg,0.324mmol),缓慢升至室温过夜,TLC监测反应完全,向反应液中加入二氯甲烷(5mL),有机相依次用饱和氯化铵洗,水洗,饱和食盐水洗,无水硫酸镁干燥,过滤,浓缩得到黄色胶状固体(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(甲基磺酰氨基甲基)吡咯烷-1-甲酸叔丁酯(90.0mg,收率:53.3%)。
步骤2:N-(((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)甲基)甲磺酰胺的合成
Figure PCTCN2017096848-appb-000105
将(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(甲基磺酰氨基甲基)吡咯烷-1-甲酸叔丁酯(90.0mg,0.27mmol,1.0eq)溶于DCM(4mL)中,冰浴降温至0℃,加入三氟乙酸(2mL),缓慢升至室温过夜,搅拌2小时,TLC监测反应完全,真空浓缩得到N-(((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)甲基)甲磺酰胺(142.0mg,收率:100%)。
步骤3:N-(((2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)甲基)甲磺酰胺(化合物33)的合成
Figure PCTCN2017096848-appb-000106
将N-(((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)甲基)甲磺酰胺(141.8mg,0.27mmol,1.0eq)溶于四氢呋喃(2mL)中,冰浴降温至0℃,依次加入三乙胺(83.2mg,0.822mmol)和丙烯酰氯(37.2mg,0.41mmol),缓慢升至室温,搅拌2小时,TLC监测反应完全,浓缩,粗品经硅胶柱层析(DCM:MeOH=200:1~100:1)得白色固体N-(((2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)甲基)甲磺酰胺(化合物2)(21.0mg,收率:13.3%)。
1HNMR(400MHz,CDCl3)δ(ppm):9.19(s,1H),7.81-7.80(d,1H),7.70(s,1H),7.59-7.51(m,2H),7.30-7.17(m,1H),7.01(s,1H),6.80-6.55(m,2H),6.23-6.17(t,1H),5.72-5.66(t,1H),4.48-4.45(t,1H),4.18-4.10(m,2H),4.08(s,6H),3.53-3.49(m,1H),3.21-3.11(m,1H),2.73(s,3H),2.33-3.32(d,1H),2.01-1.99(d,1H)。
分子式:C25H27Cl2N5O5S  分子量:580.48  LC-MS(Pos,m/z)=580.1[M+H+]。
实施例16:1-((2S,4S)-2-(氮杂环丁烷-1-羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-基)丙-2-烯-1-酮(化合物35)的合成
Figure PCTCN2017096848-appb-000107
步骤1:(2S,4S)-1-(叔丁氧羰基)-4-(6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲酸的合成
Figure PCTCN2017096848-appb-000108
将原料1-叔丁基2-甲基(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1,2-二甲酸酯(400.7mg,0.69mmol,1.0eq)溶解在甲醇(6mL)中,降温至0℃,加入一水合氢氧化锂(87.34mg,2.08mmol,3.0eq)水溶液(3mL),加完缓慢升至室温搅拌过夜,TLC监测反应完全,反应液浓缩,用甲基叔丁基醚(5mL)打浆,30分钟后抽滤,滤饼用水(10mL)溶解,用饱和碳酸氢钠水溶液将pH调节至5~6,用二氯甲烷(20mL×1)萃取,有机相用无水硫酸钠干燥,抽滤,浓缩得到化合物(2S,4S)-1-(叔丁氧羰基)-4-(6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲酸(288.7mg,收率:73.8%)。
步骤2:叔丁基(2S,4S)-2-(氮杂环丁烷-1-羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸酯的合成
Figure PCTCN2017096848-appb-000109
将(2S,4S)-1-(叔丁氧羰基)-4-(6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲酸(421.3mg,0.74mmol,1.0eq)溶于四氢呋喃(6mL)和乙腈(1 mL)的混合溶剂中,搅拌下降温至0℃,缓慢分批加入2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(312.7mg,0.82mmol,1.1eq),加完后滴加N,N-二异丙基乙胺(193.1mg,1.49mmol,2.0eq),滴加完毕0℃反应1小时。滴加氮杂环丁烷(42.68mg,0.74mmol,1.0eq)的四氢呋喃溶液(0.5mL),滴加完毕,缓慢升至室温,氮气保护下搅拌过夜。TLC显示反应完全,将体系浓缩,加入饱和氯化钠水溶液(30mL),用乙酸乙酯(3×10mL)萃取,有机相用无水硫酸钠干燥,抽滤,浓缩,粗品经柱层析(200-300目硅胶,二氯甲烷:甲醇=200:1~100:1)分离得到叔丁基(2S,4S)-2-(氮杂环丁烷-1-羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸酯(194.3mg,收率:43.1%)。
步骤3:氮杂环丁烷-1-基((2S,4S)-4-(6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-氨基)吡咯烷-2-基)甲酮的合成
Figure PCTCN2017096848-appb-000110
将叔丁基(2S,4S)-2-(氮杂环丁烷-1-羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-甲酸酯(194.3mg,0.32mmol,1.0eq)溶于二氯甲烷(5mL)中,降温至0℃,滴加三氟乙酸(5mL),滴加完毕后升到室温后反应2.5小时。TLC显示反应完全,将体系温度降至0℃,用饱和碳酸钠水溶液将体系pH调节至7~8,加入二氯甲烷萃取(3×10mL),有机相用无水硫酸钠干燥,抽滤,浓缩得到氮杂环丁烷-1-基((2S,4S)-4-(6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-氨基)吡咯烷-2-基)甲酮(151.1mg,收率:93.2%)。
步骤4:1-((2S,4S)-2-(氮杂环丁烷-1-羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-基)丙-2-烯-1-酮(化合物35)的合成
Figure PCTCN2017096848-appb-000111
将氮杂环丁烷-1-基((2S,4S)-4-(6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-氨 基)吡咯烷-2-基)甲酮(150mg,0.32mmol,1.0eq)溶于四氢呋喃(3mL)中,降温至0℃,依次滴加三乙胺(65.2mg,0.64mmol,2.0eq)和丙烯酰氯(35.0mg,0.39mmol,1.2eq),滴加完毕后缓慢升至室温搅拌4小时。TLC显示反应完全,加入饱和氯化铵水溶液,用乙酸乙酯(3×10mL)萃取,有机相合并,用无水硫酸钠干燥,抽滤,浓缩,粗品经柱层析分离(200-300目硅胶,二氯甲烷:甲醇=100:1)得产物1-((2S,4S)-2-(氮杂环丁烷-1-羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1-基)丙-2-烯-1-酮(41.0mg,收率:22.3%)。
1HNMR(400MHz,DMSO)δ(ppm):9.18(s,1H),7.95(s,1H),7.51-7.70(m,3H),7.01(s,1H),6.61-6.65(m,1H),6.14-6.19(m,1H),5.69-5.76(m,1H),4.74(m,1H),4.41-4.48(m,4H),1.11-1.12(s,6H),3.84-3.92(m,1H),3.50-3.54(t,1H),1.90-1.91(m,1H),2.55-2.89(m,1H)。
分子式:C27H27Cl2N5O4  分子量:555.14  LC-MS(Neg,m/z)=556.2[M+H+]。
实施例17:(2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-羧酸乙酯(化合物36)的合成
Figure PCTCN2017096848-appb-000112
步骤1:(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-羧酸乙酯的合成
Figure PCTCN2017096848-appb-000113
将中间体(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(二甲基氨基甲酰基)吡咯烷-1-羧酸叔丁酯(183mg,0.310mmol,1.0eq)溶于盐酸乙醇(15mL)中,45℃反应3h,TLC监测反应完成。浓缩,粗品用THF溶解,浓缩,重复三次,粗品不经纯化直接投入下一步(理论量:170mg)。
步骤2:(2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-羧酸乙酯(化合物36)的合成
Figure PCTCN2017096848-appb-000114
将中间体(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-羧酸乙酯(170mg,0.347mmol,1.0eq)溶于THF(15mL)中,加入三乙胺(175.6mg,1.735mmol,5.0eq)。降温至0℃,缓慢加入THF稀释的丙烯酰氯(31.4mg,0.347mmol,1.0eq),2h后TLC监测反应完成,加入饱和碳酸氢钠溶液(10mL),分液,水相用EA萃取(10mL×3),合并有机相浓缩。通过制备薄层色谱(DCM:MeOH=15:1)分离得固体(2S,4S)-1-丙烯酰基-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-羧酸乙酯(21mg,收率:11%)。
1HNMR(400MHz,DMSO)δ(ppm):9.02(s,1H),7.70(m,1H),7.68(s,2H),6.66(s,1H),6.45-6.47(m,2H),6.11-6.13(m,1H),5.73-5.76(m,1H),5.00(s,1H),4.66(m,1H),4.28(m,2H),4.12-4.14(m,1H),3.99(s,6H),3.77-3.80(m,1H),2.69(m,1H),2.15(m,1H),1.23-1.25(m,2H),1.25-1.27(m,3H)。
分子式:C26H26Cl2N4O5  分子量:545.42  LC-MS(Pos,m/z)=545.1[M-H+]。
实施例18:1-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(甲氧基甲基)吡咯烷-1-基)丙-2-烯-1-酮(化合物53)的合成
Figure PCTCN2017096848-appb-000115
步骤1:(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(甲氧基甲基)吡咯烷-1-甲酸叔丁酯的合成
Figure PCTCN2017096848-appb-000116
将原料(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(羟甲基)吡咯烷-1-甲酸叔丁酯(210mg,0.382mmol,1.0eq)、叔丁醇钠(91.78mg, 0.955mmol,2.5eq)溶解在THF(10mL)中,降温至0℃,搅拌1h后,用针头注入THF(1mL)稀释的碘甲烷(108.44mg,0.764mmol,2.0eq),反应过夜,第二天早上TLC监测原料剩余少许,加饱和氯化铵溶液(10mL)、DCM(20mL),分液,水相TLC监测无产品,有机相干燥浓缩,柱层析(PE:EA=5:1)得(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(甲氧基甲基)吡咯烷-1-甲酸叔丁酯(200mg,收率:93%)。
步骤2:6-(2,6-二氯-3,5-二甲氧基苯基)-N-((3S,5S)-5-(甲氧基甲基)吡咯烷-3-基)喹唑啉-2-胺的合成
Figure PCTCN2017096848-appb-000117
将中间体(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(甲氧基甲基)吡咯烷-1-甲酸叔丁酯(200mg,0.355mmol,1.0eq)溶于DCM(5mL)中,搅拌溶清,降温至0℃,加入三氟乙酸(5mL),反应1.5h后,TLC监测反应完成,浓缩,用加入THF(50mL)溶解,浓缩,重复三次,粗品投入下一步。
步骤3:1-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(甲氧基甲基)吡咯烷-1-基)丙-2-烯-1-酮的合成:
Figure PCTCN2017096848-appb-000118
将中间体6-(2,6-二氯-3,5-二甲氧基苯基)-N-((3S,5S)-5-(甲氧基甲基)吡咯烷-3-基)喹唑啉-2-胺(160mg,0.345mmol,1.0eq)、三乙胺(174.55mg,1.725mmol,5.0eq)溶于THF(10mL)中,降温至0℃,用针头注入THF(1mL)稀释的丙烯酰氯(31.22mg,0.345mmol,1.0eq),反应过夜。第二天早上TLC监测反应完成。加饱和碳酸氢钠溶液(10mL)、EA(20mL),分液,水相用EA萃取。TLC显示水相无产品,有机相合并,干燥浓缩,柱层析(PE:EA=5:1→3:1),得1-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(甲氧基甲基) 吡咯烷-1-基)丙-2-烯-1-酮(34mg,收率:19%)。
1HNMR(400MHz,DMSO)δ(ppm):9.19(s,1H),7.80(s,1H),7.69(s,1H),7.51-7.59(m,2H),7.02(s,1H),6.70-6.75(m,1H),6.57-6.70(m,1H),6.18(m,1H),5.67-5.76(m,1H),4.44-4.52(m,2H),4.22-4.36(m,2H),3.98(s,6H),3.61(s,1H),3.28-3.32(m,1H),3.22(s,3H),2.02(s,1H),1.23(s,1H)。
分子式:C25H26Cl2N4O4分子量:517.41LC-MS(Neg,m/z)=517.2[M-H+]。
实施例19:1-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(吡咯烷-1-羰基)吡咯烷-1-基)丙-2-烯-1-酮(化合物55)的合成
Figure PCTCN2017096848-appb-000119
步骤1:(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1,2-二羧酸1-(叔丁基)-2-甲酯的合成
Figure PCTCN2017096848-appb-000120
将原料2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(3.28g,8.87mmol,1.0eq)溶解在N-甲基吡咯烷酮(8mL)中,室温下向体系中加入N,N-二异丙基乙胺(3.44g,26.6mmol,3.0eq)和(2S,4S)-4-氨基吡咯烷-1,2-二羧酸1-(叔丁基)-2-甲酯盐酸盐(3.73g,13.3mmol,1.5eq),加完升至120℃回流搅拌过夜。第二天,TLC监测反应完全,反应液冷却后加入水(32mL),有固体析出,搅拌2小时后抽滤得到固体,固体用二氯甲烷(150×3ml)萃取,有机相用无水硫酸钠干燥后柱层析(石油醚:乙酸乙酯=5:1)分离,得到(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1,2-二羧酸1-(叔丁基)-2-甲酯(1.91g,收率:38.1%)。
步骤2:(2S,4S)-1-(叔丁氧基羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲酸的合成
Figure PCTCN2017096848-appb-000121
将中间体(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-1,2-二羧酸1-(叔丁基)-2-甲酯(1.05g,1.81mmol,1.0eq)溶于甲醇(14mL)中,室温搅拌下温度降至0℃左右滴加一水合氢氧化锂(229.5mg,5.46mmol,3.0eq)水溶液(5.5ml),滴毕,缓慢升至室温搅拌过夜。第二天TLC显示反应完全,将体系浓缩至干,加入甲基叔丁基醚(30ml)搅拌半小时后抽滤,滤饼用水(20ml)溶解后将其pH调至6~7,再加入二氯甲烷(40ml)萃取,有机相无水硫酸钠干燥后浓缩得到产物(2S,4S)-1-(叔丁氧基羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲酸(900.8mg,收率:89.8%)。步骤3:(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(吡咯烷-1-羰基)羧酸的合成
Figure PCTCN2017096848-appb-000122
将(2S,4S)-1-(叔丁氧基羰基)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-甲酸(287.3mg,0.50mmol,1.0eq)溶于四氢呋喃(11mL)和乙腈(1.5ml)的混合溶液中,搅拌下温度降至0℃左右滴加7-偶氮苯并三氮唑(213.27mg,0.56mmol,1.1eq)和N,N-二异丙基乙胺(131.7mg,1.01mmol,2.0eq),滴毕,0℃反应1小时;1小时后同样温度滴加吡咯烷,反应3小时,3小时后TLC显示反应完全。将体系浓缩至干后用乙酸乙酯(30ml)和饱和氯化钠水溶液(15ml)洗涤3次,合并有机相且用无水硫酸钠干燥后浓缩得到(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(吡咯烷-1-羰基)羧酸(471.8mg,收率:100%)。
步骤4:((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)(吡咯烷-1-基)甲酮的合成
Figure PCTCN2017096848-appb-000123
将(2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(吡咯烷-1-羰基)羧酸(374.1mg,0.60mmol,1.0eq)溶于二氯甲烷(6ml)中,搅拌下温度降至0℃左右滴加三氟乙酸(4ml),滴毕,缓慢升至室温搅拌3小时;3小时后TLC显示反应完全。将体系冷却至0℃左右后滴加饱和碳酸氢钠水溶液将体系pH调节至6~7,再加入二氯甲烷(15ml)萃取,有机相无水硫酸钠干燥后浓缩得到((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)(吡咯烷-1-基)甲酮(226.0mg,收率:72.2%)。
步骤5:1-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(吡咯烷-1-羰基)吡咯烷-1-基)丙-2-烯-1-酮的合成
Figure PCTCN2017096848-appb-000124
将((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)吡咯烷-2-基)(吡咯烷-1-基)甲酮溶解于四氢呋喃(5ml)中,搅拌下降温至0℃后依次滴加三乙胺(132.8mg,1.31mmol,3.0eq)和丙烯酰氯(55.4mg,0.61mmol,1.4eq),滴毕,缓慢升至室温搅拌过夜。第二天,TLC显示反应完全,向体系中加入饱和氯化铵水溶液(10ml)和二氯甲烷(20ml)分层,有机相无水硫酸钠干燥后浓缩至干,然后加入甲基叔丁基醚(10ml)搅拌2小时后抽滤得到产物1-((2S,4S)-4-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-(吡咯烷-1-羰基)吡咯烷-1-基)丙-2-烯-1-酮(162.8mg,收率66.1%)。
1HNMR(400MHz,DMSO)δ(ppm):9.18(s,1H),7.95(s,1H),7.70(s,1H),7.51-7.59(m,2H),7.01(s,1H),6.63-6.67(m,1H),6.12-6.16(m,1H),5.67-5.70(m,1H),4.70-4.71(m,1H),4.68(s,1H),4.13(s,5H),4.21(m,1H),3.74-3.76(m,1H),3.49-3.54(m,1H),1.89-1.94(m,3H),1.78-1.83(m,2H)。
分子式:C28H29Cl2N5O4  分子量:569.16  LC-MS(Neg,m/z)=570.2[M+H+]。
实施例20:1-((3S,4R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-4- 甲基吡咯烷-1-基)丙-2-炔-1-酮(化合物56)的合成
Figure PCTCN2017096848-appb-000125
步骤1:(3S,4R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉)-2-基)氨基)-4-甲基吡咯烷-1-羧酸叔丁酯的合成
Figure PCTCN2017096848-appb-000126
将原料2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(400.0mg,1.1mmol,1.0eq)和(3S,4R)-3-氨基-4-甲基吡咯烷-1-甲酸叔丁酯(440.6mg,2.2mmol,2.0eq)溶解在N-甲基吡咯烷酮(3.0mL)中,加入N,N-二异丙基乙胺(568.7mg,4.4mmol,4.0eq),逐渐升温至110℃反应过夜。TLC监测反应完全,将反应液降温至室温,加入冰水(15mL),过滤,滤饼用少量冰水洗涤。用乙酸乙酯(20mL)溶解滤饼,乙酸乙酯相用饱和食盐水(10mL)洗涤,加入无水硫酸钠干燥,过滤,浓缩。粗品通过硅胶柱层析(DCM:MeOH=200:1~100:1)得黄色固体(3S,4R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉)-2-基)氨基)-4-甲基吡咯烷-1-羧酸叔丁酯(320.6mg,54.7%)。
步骤2:(6-(2,6-二氯-3,5-二甲氧基苯基)-N-((3S,4R)-4-甲基吡咯烷-3-基)喹唑啉-2-胺盐酸盐的合成:
Figure PCTCN2017096848-appb-000127
将原料(3S,4R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉)-2-基)氨基)-4-甲基吡咯烷-1-羧酸叔丁酯(320.6mg,1.0eq)溶解在乙醇(8.0mL)中,冰浴降温至0℃,加入氯化氢乙醇溶液(8.0mL),搅拌反应两小时,TLC监测反应完全。将反应液直接浓缩得到淡黄色固体(6-(2,6-二氯-3,5-二甲氧基苯基)-N-((3S,4R)-4-甲基吡咯烷-3-基)喹唑啉-2-胺盐酸盐(339.2mg粗品,收率 100%)。
步骤3:1-((3S,4R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-4-甲基吡咯烷-1-基)丙-2-炔-1-酮的制备
Figure PCTCN2017096848-appb-000128
将(6-(2,6-二氯-3,5-二甲氧基苯基)-N-((3S,4R)-4-甲基吡咯烷-3-基)喹唑啉-2-胺盐酸盐(339.2mg)溶解在四氢呋喃(10.0mL)中,加入三乙胺(203.0mg,2.0mmol,3.0eq),超声5分钟。加入水(15mL)和乙酸乙酯(10mL)。分液,水相用乙酸乙酯萃取(8.0mL×2),合并有机相。有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤,浓缩得淡黄色固体(216.0mg)。取处理后的固体(108.0mg,0.25mmol)溶于二氯甲烷(2mL)中,加入丙炔酸(37.8mg,0.54mmol),4-二甲氨基吡啶(3.1mg,0.025mmol)和N,N'-二环己基碳酰亚胺(56.5mg,0.27mmol),在微波、40℃下反应30min,TLC监测反应完全。将反应液水洗(5mL×3)后浓缩至干并用制备薄层色谱板分离,得黄色固体1-((3S,4R)-3-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-4-甲基吡咯烷-1-基)丙-2-炔-1-酮(24.2mg,收率20%)。
1HNMR(400MHz,DMSO-d6)δ(ppm):9.20-9.19(d,1H),7.96(s,1H),7.86-7.81(t,1H),7.69(s,1H),7.56-7.52(m,2H),7.01(s,1H),4.49-4.42(d,1H),4.25-4.15(t,3H),3.97(s,6H),4.03(s,6H),3.77-3.72(m,1H),3.45-3.41(m,1H),3.20-3.15(m,1H),3.06-3.01(m,1H),2.89(s,2H),2.73(s,2H),2.40-2.33(d,2H),1.24(s,2H),1.10(s,3H)。
分子式:C24H22Cl2N4O3  分子量:485.37  LC-MS(Pos,m/z)=485.43[M+H+]。
实施例21:1-(6-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-氮杂螺[3.3]庚烷-2-基)丙-2-烯-1酮(化合物65)的合成
Figure PCTCN2017096848-appb-000129
步骤1:6-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-氮杂螺[3.3]庚烷-2-羧酸叔丁酯的合成
Figure PCTCN2017096848-appb-000130
将原料2-氯-6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉(400mg,1.082mmol,1.0eq)、6-氨基-2-氮杂螺[3.3]庚烷-2-羧酸叔丁酯(248.53mg,1.082mmol,1.0eq)溶解在NMP(1.5mL)中,加入DIPEA(699.24mg,5.41mmol,5.0eq),升温至110℃,反应过夜。第二天早上TLC监测反应完成,降温后将反应液缓慢倒入水中(50mL),抽滤,滤饼用DCM溶解,水相用EA(100mL×2)萃取,分液,水相TLC显示无产品,合并有机相,干燥浓缩,柱层析(PE:EA=5:1)得稠状液体6-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-氮杂螺[3.3]庚烷-2-羧酸叔丁酯(812mg),直接进行下一步反应。
步骤2:6-(2,6-二氯-3,5-二甲氧基苯基)-N-(2-氮杂螺[3.3]庚烷-6-基)喹唑啉-2-胺的合成
Figure PCTCN2017096848-appb-000131
将中间体6-((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-氮杂螺[3.3]庚烷-2-羧酸叔丁酯(590.3mg,0.355mmol,1.0eq)溶于DCM(5mL)中,搅拌溶清,降温至0℃,加入三氟乙酸(5mL),反应1h后,TLC监测反应完成,浓缩,加入THF(50mL)溶解,浓缩,重复三次,粗品投入下一步。
步骤3:1-(6–((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-氮杂螺[3.3]庚烷-2-基)丙-2-烯-1-酮的合成:
Figure PCTCN2017096848-appb-000132
将中间体6-(2,6-二氯-3,5-二甲氧基苯基)-N-(2-氮杂螺[3.3]庚烷-6-基)喹唑 啉-2-胺(480mg,1.08mmol,1.0eq)、三乙胺(546.4mg,5.4mmol,5.0eq)溶于THF(10mL)中,体系pH调节至8左右,降温至0℃,用针头注入THF(1mL)稀释的丙烯酰氯(97.74mg,1.08mmol,1.0eq),1h后TLC监测显示原料剩余少许,补加一滴丙烯酰氯,0.5h后TLC显示原料剩余少许,加饱和碳酸氢钠溶液(10mL)、EA(20mL),搅拌分液,水相用EA萃取(10mL×2)。TLC显示水相无产品,有机相合并,干燥浓缩,柱层析(PE:EA=2:1→1:1),得1-(6–((6-(2,6-二氯-3,5-二甲氧基苯基)喹唑啉-2-基)氨基)-2-氮杂螺[3.3]庚烷-2-基)丙-2-烯-1-酮(337mg,收率:62%)
1HNMR(400MHz,DMSO)δ(ppm):9.14(s,1H),7.81(s,1H),7.66(s,1H),7.48-7.52(m,2H),7.01(s,1H),6.31-6.34(m,1H),6.07-6.11(m,1H),5.64-5.68(m,1H),4.54(s,1H),4.40(s,1H),4.26(m,1H),4.20(s,1H),3.98(s,6H),3.80(s,1H),2.62(s,2H),2.26(s,2H).
分子式:C25H24Cl2N4O3分子量:499.39LC-MS(Neg,m/z)=499.1[M-H+].
参照上述制备方法,可以制备如下化合物:
Figure PCTCN2017096848-appb-000133
Figure PCTCN2017096848-appb-000134
Figure PCTCN2017096848-appb-000135
Figure PCTCN2017096848-appb-000136
Figure PCTCN2017096848-appb-000137
Figure PCTCN2017096848-appb-000138
根据下述实验例,可以更好地理解本发明。然而,本领域的技术人员容易理解,实施例所描述的内容仅用于说明本发明,而不应当也不会限制权利要求书中所详细描述的本发明。
实验例1:本发明化合物的酶学活性测试
测试物:本发明中的化合物,其结构见前文所示。
测试仪器:使用LabChip EZ ReaderⅡ药物筛选平台。
试验方法:
1.化合物板准备
a)96孔板,10个剂量组,3倍系列稀释,每孔加入DMSO,最高浓度为500μM
b)384孔板,通过1倍激酶缓冲液(50mM HEPES,Ph 7.5;0.0015%Brij-35;2mM DTT)稀释,使每孔含有5μL 10%DMSO溶解的5倍化合物。阴性对照孔为5μL含有10%DMSO的1倍激酶缓冲液。
2.实验步骤
FGFR1-4(h)在1倍激酶缓冲液中,与2.5倍酶液在室温中反应10min后,加入FAM标记的多肽底物与ATP启动反应后,孵育30分钟加入25μL终止液(100mM HEPES,pH 7.5;0.015%Brij-35;0.2%Coating Reagent#3;50mM EDTA)终止反应Caliper读取终数据。
测试结果如表1所示:
表1本发明化合物对FGFR的抑制活性(IC50)
Figure PCTCN2017096848-appb-000139
Figure PCTCN2017096848-appb-000140
-表示未测试。
由表1实验结果可见,本发明的化合物对FGFR具有良好的抑制活性,说明本发明化合物在治疗由FGFR异常介导的疾病方面具有较好的临床应用潜力。
实验例2:本发明化合物的细胞活性测试
Hep3B为肝细胞癌FGFR异常细胞
RT112/84为膀胱癌FGFR异常细胞
DMS114为小细胞肺癌FGFR异常细胞
AN3CA为子宫内膜癌FGFR异常细胞
SNU-16为胃癌FGFR异常细胞
测试物:本发明中的化合物,其结构见前文所示。
测试仪器:使用Espire多功能酶标仪。
试验方法:
各株细胞接种于96孔板中贴壁培养过夜后,加入不同浓度的化合物(12个剂量组,3倍DMSO系列稀释)使终浓度为0.17-30000nM,其中DMSO终含量均为5‰。阴性对照孔为含有5‰DMSO的培养基。37℃,5%CO2,95%湿度孵育72h后待测。每孔加入30μL Cell titer-Glo试剂,室温孵育30min后,Espire读取终数据。
测试结果如表2所示:
表2本发明化合物对细胞的抑制活性(IC50)
Figure PCTCN2017096848-appb-000141
Figure PCTCN2017096848-appb-000142
-表示未测试。
由表2实验结果可见,本发明的化合物对Hep3B、RT112/84、DMS114、AN3CA、SNU-16等FGFR异常的细胞具有良好的抑制活性,说明本发明化合物可以用来治疗由FGF/FGFR异常介导的癌症如肝癌、胃癌、小细胞癌、膀胱癌、子宫内膜癌,具有非常好的临床使用价值。

Claims (18)

  1. 通式(I)表示的成纤维细胞生长因子受体(FGFR)不可逆抑制剂,或其药学上可接受的盐、立体异构体:
    Figure PCTCN2017096848-appb-100001
    其中,
    R1、R2分别独立地选自氢、羟基、氨基、氰基、硝基、卤素原子、羧基、C1-6烷基、C1-6烷氧基、C1-6烷基氨基、(C1-6烷基)2氨基、卤代C1-6烷基、卤代C1-6烷氧基、C2-8烯基、C2-8炔基、C1-6烷基磺酰基、C1-6烷基羰基氨基,C1-6烷基取代的3~8元环烷基、C1-6烷基取代的3~8元杂环基,可供选择的,R1和R2可与它们分别连接的芳环或杂芳环上的两个原子一起形成3~8元环烷基、3~8元杂环基、6~14元芳基或5~10元杂芳基,并且任意环中的S原子可任选被氧化为S(O)或S(O)2,任意环中的碳原子可任选地被氧化为C(O);
    R3、R4分别独立地选自氢、羟基、氨基、氰基、硝基、卤素原子、羧基、C1-6烷基、C1-6烷氧基、C1-6烷基氨基、(C1-6烷基)2氨基、(C1-6烷基)2氨基C1-6烷基、卤代C1-6烷基、卤代C1-6烷氧基、C2-8烯基、C2-8炔基、C1-6烷基磺酰基、C1-6烷基羰基氨基、C1-6烷基氨基羰基、3~8元环烷基、3~8元杂环基、6~14元芳基或5~10元杂芳基、C1-6烷基取代的3~8元环烷基、C1-6烷基取代的3~8元杂环基、C1-6烷基取代的6~14元芳基或C1-6烷基取代的5~10元杂芳基;
    Ar选自任选的含0~3个O、S和/或N原子的6~14元芳环基或5~10元杂芳基;
    环A选自任选被1~3个R5取代的含0~3个O、S和/或N原子的3~8元环烷基、3~8元杂环基、6~14元芳基或5~10元杂芳基,其中,任意环中的S原子可任选地被氧化为S(O)或S(O)2,任意环中的碳原子可任选地被氧化为C(O);
    环B选自任选被1~3个R6取代的含至少1个N杂原子的3~10元饱和或不饱和的杂环基或5~6元含N杂芳基,并且,环B上的N原子与Warhead键直接相连,其中,任意环B中的S原子可任选被氧化为S(O)或S(O)2,并且任意的环B中的碳原子可任选地被氧化为C(O);
    X选自CR7、N;
    R5、R6、R7分别独立的选自
    (i)氢,
    (ii)羟基、氨基、羧基、氰基、硝基、卤素原子、C2-4烯基羰基氨基、=CH2
    (iii)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~6烷基、C1-6烷氧基、C1-6烷氧C1-6烷氧基、C1-6烷基氨基、(C1-6烷基)2氨基、C1-6烷基羰基氨基、C1-6烷基磺酰氨基、3~8元杂环基取代的C1~6烷基、C1-6烷氧基、C1-6烷基氨基、(C1-6烷基)2氨基、卤代C1-6烷基、卤代C1-6烷氧基、C2-8烯基、C2-8炔基、C1-6烷基磺酰基、C1-6烷基硫基,所述的3~8元杂环基可任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~6烷基、C1-6烷氧基、C1-6烷基氨基、(C1-6烷基)2氨基取代,
    (iv)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1-6烷基、C1-6烷氧基、C1-6烷基氨基、(C1-6烷基)2氨基取代的3~8元环烷基、3~8元杂环基,
    (v)氨基-羰基、氰基-羰基、C1-6烷基-羰基、C1-6烷基氨基-羰基、(C1-6烷基)2氨基-羰基、C1-6烷氧基-羰基、3~8元环烷基-羰基、3~8元杂环基-羰基;
    m1、m2代表1、2或3,且m1与m2相加小于等于5;
    Warhead指的是能够与亲核试剂形成共价键的部分。
  2. 如权利要求1所述的化合物或其药学上可接受的盐、立体异构体:
    其中,
    R1选自氢、卤素、羟基;
    R2选自氢、卤素、羟基、C1-4烷基、C1-4烷氧基、氰基、卤代C1-4烷基、卤代C1-4烷氧基;
    Ar选自任选的含0~3个O、S和/或N原子的6~14元芳环基或5~6元杂芳基;
    m1、m2代表1、2或3,且m1与m2相加小于等于5。
  3. 如权利要求2所述的化合物或其药学上可接受的盐、立体异构体:
    其中,
    R1选自氢、卤素、羟基;
    R2选自氢、卤素、羟基、C1-4烷基、C1-4烷氧基、氰基、卤代C1-4烷基、卤代C1-4烷氧基;
    R3、R4分别独立的选自氢、羟基、氨基、氰基、硝基、卤素原子、羧基、C1-4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、(C1-4烷基)2氨基C1-4烷基、卤代C1-4烷基、卤代C1-4烷氧基、C2-4烯基、C2-4炔基、C1-4烷基磺酰基、C1-4烷基羰基氨基;
    Ar为苯基;
    环A选自任选被1~3个R5取代的苯基;
    环B选自任选被1~3个R6取代的含至少1个N杂原子的4~10元饱和或不饱和的杂环基,并且,环B上的N原子与Warhead键直接相连;
    X选自CR7、N;
    R5、R7分别独立的选自氢、羟基、氨基、羧基、氰基、硝基、卤素原子、C1-4烷基、C1-4烷氧基;
    R6选自
    (i)氢,
    (ii)羟基、氨基、羧基、氰基、硝基、卤素原子、C2-4烯基羰基氨基、=CH2
    (iii)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷氧C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、C1-4烷基羰基氨基、C1-4烷基磺酰氨基、3~8元杂环基取代的C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、卤代C1-4烷基、卤代C1-4烷氧基、C2-4烯基、C2-4炔基、C1-4烷基磺酰基、C1-4烷基硫基,所述的3~8元杂环基可任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基取代,
    (iv)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1-4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基取代的3~8元环烷基、3~8元杂环基,
    (v)氨基-羰基、氰基-羰基、C1-4烷基-羰基、C1-4烷基氨基-羰基、(C1-4烷基)2 氨基-羰基、C1-4烷氧基-羰基、3~8元环烷基-羰基、3~8元杂环基-羰基;
    m1、m2代表1、2或3,且m1与m2相加小于等于5;
    Warhead指的是能够与亲核试剂形成共价键的部分。
  4. 如权利要求1~3任意一项所述的式(I)化合物或其药学上可接受的盐、立体异构体,结构如通式(II)所示:
    Figure PCTCN2017096848-appb-100002
    R4选自氢、C1-4烷基、(C1-6烷基)2氨基C1-6烷基;
    环A为苯基;
    环B选自任选被1~3个R6取代的含至少1个N杂原子的4~6元饱和或不饱和的单杂环基或6~10元饱和或不饱和的稠杂环基,并且,环B上的N原子与Warhead键直接相连;
    X为N;
    R6选自
    (i)氢,
    (ii)羟基、氨基、羧基、氰基、硝基、卤素原子、C2-4烯基羰基氨基、=CH2
    (iii)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷氧C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、C1-4烷基羰基氨基、C1-4烷基磺酰氨基、3~8元杂环基取代的C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、卤代C1-4烷基、卤代C1-4烷氧基、C2-4烯基、C2-4炔基、C1-4烷基磺酰基、C1-4烷基硫基,所述的3~8元杂环基可任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基取代,
    (iv)氨基-羰基、氰基-羰基、C1-4烷基-羰基、C1-4烷基氨基-羰基、(C1-4烷基)2氨基-羰基、C1-4烷氧基-羰基、3~8元环烷基-羰基、3~8元杂环基-羰基;
    Warhead选自
    Figure PCTCN2017096848-appb-100003
    Z指离去基团或活化羟基部分,
    R11,R12,R13独立地选自氢,卤素,氰基,任选被取代基取代的C1-4烷基、卤代C1-4烷基、3~8元环烷基、3~8元杂环基、5~8元芳基、5~10元杂芳基,所述取代基选自:羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷氧C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、C1-4烷基羰基氨基、C1-4烷基磺酰氨基、3~8元杂环基;R11,R12,R13优选为氢。
  5. 如权利要求1~4中任一项所述的化合物或其药学上可接受的盐、立体异构体,结构如通式(II)所示:
    Figure PCTCN2017096848-appb-100004
    Warhead与环B上的N原子直接相连,如下所示:
    Figure PCTCN2017096848-appb-100005
    环B选自如下基团:
    Figure PCTCN2017096848-appb-100006
    Figure PCTCN2017096848-appb-100007
  6. 如权利要求1~5任一项所述的化合物或其药学上可接受的盐、立体异构体:
    warhead选自如下结构,
    Figure PCTCN2017096848-appb-100008
    Z指离去基团或活化羟基部分,
    R11,R12,R13分别独立地选自氢或C1-4烷基。
  7. 如权利要求1~6任一项所述的化合物或其药学上可接受的盐、立体异构体,所述化合物选自如下结构:
    Figure PCTCN2017096848-appb-100009
    Figure PCTCN2017096848-appb-100010
  8. 如权利要求4所述的化合物或其药学上可接受的盐、立体异构体:
    环B选自任选被1~3个R6取代的含至少1个N杂原子的5~6元饱和单杂环基,并且,环B上的N原子与Warhead键直接相连。
  9. 如权利要求8所述的化合物或其药学上可接受的盐、立体异构体,结构如通式(III)所示:
    Figure PCTCN2017096848-appb-100011
    R4选自氢或C1-4烷基;
    X为N;
    R6选自
    (i)氢,
    (ii)羟基、氨基、羧基、氰基、硝基、卤素原子,
    (iii)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷氧C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、C1-4烷基羰基氨基、C1-4烷基磺酰氨基、3~8元杂环基取代的C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、卤代C1-4烷基、卤代C1-4烷氧基、C2-4烯基、C2-4炔基、C1-4烷基磺酰基、C1-4烷基硫基,所述的3~8元杂环基可任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基取代,
    (iv)氨基-羰基、氰基-羰基、C1-4烷基-羰基、C1-4烷基氨基-羰基、(C1-4烷基)2氨基-羰基、C1-4烷氧基-羰基、3~8元环烷基-羰基、3~8元杂环基-羰基;
    m为1~3的整数;
    Warhead选自
    Figure PCTCN2017096848-appb-100012
    Z指离去基团或活化羟基部分;
    R11,R12,R13独立地选自氢,卤素,氰基,任选被取代基取代的C1-4烷基、卤代C1-4烷基、3~8元环烷基、3~8元杂环基、5~8元芳基、5~10元杂芳基,所述取代基选自:羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷氧C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、C1-4烷基羰基氨基、C1-4烷基磺酰氨基、3~8元杂环基;R11,R12,R13优选为氢。
  10. 如权利要求9所述的化合物或其药学上可接受的盐、立体异构体:
    R6选自
    (i)氢,
    (ii)羟基、氨基、羧基、氰基、硝基、卤素原子,
    (iii)任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷氧C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基、C1-4烷基羰基氨基、C1-4烷基磺酰氨基、3~8元杂环基取代的C1~4烷基、C1-4烷氧基,所述的3~8元杂环基可任选被羟基、氨基、羧基、氰基、硝基、卤素、C1~4烷基、C1-4烷氧基、C1-4烷基氨基、(C1-4烷基)2氨基取代;
    (iv)氨基-羰基、氰基-羰基、C1-4烷基-羰基、C1-4烷基氨基-羰基、(C1-4烷基)2氨基-羰基、C1-4烷氧基-羰基、3~8元环烷基-羰基、3~8元杂环基-羰基;
    m为1~3的整数;
    Warhead选自
    Figure PCTCN2017096848-appb-100013
    R11,R12,R13独立地选自氢或C1-4烷基,warhead优选为
    Figure PCTCN2017096848-appb-100014
  11. 如权利要求10所述的化合物或其药学上可接受的盐、立体异构体,选自如下结构的化合物:
    Figure PCTCN2017096848-appb-100015
    Figure PCTCN2017096848-appb-100016
    Figure PCTCN2017096848-appb-100017
  12. 含有权利要求1~11任一项所述的化合物或其药学上可接受的盐、立体异构体的药物制剂,其特征在于还包含一种或多种药用载体。
  13. 权利要求12所述的药物制剂,其特征在于进一步包含一种或多种第二治疗活性剂,所述的第二治疗活性剂为抗代谢物、生长因子抑制剂、有丝分裂抑制剂、抗肿瘤激素类、烷化剂类、金属类、拓扑异构酶抑制剂、激素药、免疫调节剂、肿瘤抑制基因、癌疫苗、免疫检查点或肿瘤免疫治疗相关 的抗体和小分子药物。
  14. 权利要求1~11任一项所述的化合物或其药学上可接受的盐、立体异构体或者权利要求12~13任一项所述的药物制剂在制备治疗FGF/FGFR异常介导的疾病的药物中的应用。
  15. 根据权利要求14所述的应用,所述的FGF/FGFR异常介导的疾病为癌症;所述的癌症包括肺癌、鳞状上皮细胞癌、膀胱癌、胃癌、卵巢癌、腹膜癌、乳腺癌、乳腺导管癌、头颈癌、子宫内膜癌、宫体癌、直肠癌、肝癌、肾癌、肾盂癌、食管癌、食管腺癌、神经胶质瘤、前列腺癌、甲状腺癌、女性生殖系统癌症、原位癌、淋巴瘤、神经纤维瘤病、骨癌、皮肤癌、脑癌、结肠癌、睾丸癌、胃肠道间质瘤、口腔癌、咽癌、多发性骨髓瘤、白血病、非霍奇金淋巴瘤、大肠绒毛腺瘤、黑色素瘤、细胞瘤和肉瘤、骨髓增生异常综合症。
  16. 一种用于治疗FGF/FGFR异常介导的疾病的方法,所述方法包括向有此需要的对象施用权利要求1~11任一项所述的化合物或其药学上可接受的盐、立体异构体或者权利要求12~13任一项所述的药物制剂。
  17. 根据权利要求16所述的方法,所述的FGF/FGFR异常介导的疾病为癌症;所述的癌症包括肺癌、鳞状上皮细胞癌、膀胱癌、胃癌、卵巢癌、腹膜癌、乳腺癌、乳腺导管癌、头颈癌、子宫内膜癌、宫体癌、直肠癌、肝癌、肾癌、肾盂癌、食管癌、食管腺癌、神经胶质瘤、前列腺癌、甲状腺癌、女性生殖系统癌症、原位癌、淋巴瘤、神经纤维瘤病、骨癌、皮肤癌、脑癌、结肠癌、睾丸癌、胃肠道间质瘤、口腔癌、咽癌、多发性骨髓瘤、白血病、非霍奇金淋巴瘤、大肠绒毛腺瘤、黑色素瘤、细胞瘤和肉瘤、骨髓增生异常综合症。
  18. 权利要求1~11任一项所述的化合物或其药学上可接受的盐、立体异构体或者权利要求12~13任一项所述的药物制剂用作药物。
PCT/CN2017/096848 2016-09-01 2017-08-10 成纤维细胞生长因子受体抑制剂及其用途 WO2018040885A1 (zh)

Priority Applications (7)

Application Number Priority Date Filing Date Title
KR1020197007909A KR102270620B1 (ko) 2016-09-01 2017-08-10 섬유아세포 성장 인자 수용체의 억제제 및 이의 용도
US16/328,817 US10744139B2 (en) 2016-09-01 2017-08-10 Inhibitors of fibroblast growth factor receptor and use thereof
RU2019107910A RU2732127C1 (ru) 2016-09-01 2017-08-10 Ингибиторы рецептора фактора роста фибробластов и их применение
AU2017317864A AU2017317864B2 (en) 2016-09-01 2017-08-10 Inhibitors of fibroblast growth factor receptor and use thereof
ES17845187T ES2859637T3 (es) 2016-09-01 2017-08-10 Inhibidores del receptor de factor de crecimiento de los fibroblastos y uso de los mismos
EP17845187.8A EP3508482B1 (en) 2016-09-01 2017-08-10 Inhibitors of fibroblast growth factor receptor and use thereof
JP2019511715A JP6751977B2 (ja) 2016-09-01 2017-08-10 線維芽細胞増殖因子受容体の阻害剤及びそれらの使用

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201610802100.5 2016-09-01
CN201610802100 2016-09-01
CN201710351160 2017-05-18
CN201710351160.4 2017-05-18

Publications (1)

Publication Number Publication Date
WO2018040885A1 true WO2018040885A1 (zh) 2018-03-08

Family

ID=61300040

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/096848 WO2018040885A1 (zh) 2016-09-01 2017-08-10 成纤维细胞生长因子受体抑制剂及其用途

Country Status (9)

Country Link
US (1) US10744139B2 (zh)
EP (1) EP3508482B1 (zh)
JP (1) JP6751977B2 (zh)
KR (1) KR102270620B1 (zh)
CN (1) CN107793395B (zh)
AU (1) AU2017317864B2 (zh)
ES (1) ES2859637T3 (zh)
RU (1) RU2732127C1 (zh)
WO (1) WO2018040885A1 (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020131627A1 (en) * 2018-12-19 2020-06-25 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as inhibitors of fgfr tyrosine kinases
EP3694528A4 (en) * 2017-10-13 2021-07-28 The Regents of the University of California MTORC1 MODULATORS
WO2022022735A1 (zh) 2020-07-31 2022-02-03 药捷安康(南京)科技股份有限公司 成纤维细胞生长因子受体抑制剂的晶型及其制备方法

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110386921A (zh) * 2018-04-23 2019-10-29 南京药捷安康生物科技有限公司 成纤维细胞生长因子受体抑制剂化合物
CN113318113A (zh) * 2020-02-28 2021-08-31 南京药捷安康生物科技有限公司 成纤维细胞生长因子受体抑制剂的用途
AR125588A1 (es) 2021-03-04 2023-08-02 Lilly Co Eli Compuestos inhibidores de fgfr3

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1878727A1 (en) * 2005-04-28 2008-01-16 Kyowa Hakko Kogyo Co., Ltd. 2-aminoquinazoline derivatives
CN104540809A (zh) * 2012-07-11 2015-04-22 蓝印药品公司 成纤维细胞生长因子受体的抑制剂
WO2015108992A1 (en) * 2014-01-15 2015-07-23 Blueprint Medicines Corporation Heterobicyclic compounds and their use as fgfr4 receptor inhibitors
CN105658642A (zh) * 2013-10-25 2016-06-08 蓝图药品公司 纤维母细胞生长因子受体抑制剂

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2335535T3 (es) * 2005-11-15 2010-03-29 Array Biopharma Inc. Derivados de n4-fenil-quinazolin-4-amina ycompuestos afines como inhibidores de la tirosina quinasa receptora erbb tipo i para el tratamiento de enfermedades hiperproliferativas.
GB201307577D0 (en) 2013-04-26 2013-06-12 Astex Therapeutics Ltd New compounds
WO2017070256A2 (en) * 2015-10-19 2017-04-27 Araxes Pharma Llc Method for screening inhibitors of ras

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1878727A1 (en) * 2005-04-28 2008-01-16 Kyowa Hakko Kogyo Co., Ltd. 2-aminoquinazoline derivatives
CN104540809A (zh) * 2012-07-11 2015-04-22 蓝印药品公司 成纤维细胞生长因子受体的抑制剂
CN105658642A (zh) * 2013-10-25 2016-06-08 蓝图药品公司 纤维母细胞生长因子受体抑制剂
WO2015108992A1 (en) * 2014-01-15 2015-07-23 Blueprint Medicines Corporation Heterobicyclic compounds and their use as fgfr4 receptor inhibitors

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3694528A4 (en) * 2017-10-13 2021-07-28 The Regents of the University of California MTORC1 MODULATORS
WO2020131627A1 (en) * 2018-12-19 2020-06-25 Array Biopharma Inc. Substituted pyrazolo[1,5-a]pyridine compounds as inhibitors of fgfr tyrosine kinases
CN113490666A (zh) * 2018-12-19 2021-10-08 奥瑞生物药品公司 作为fgfr酪氨酸激酶的抑制剂的取代的吡唑并[1,5-a]吡啶化合物
WO2022022735A1 (zh) 2020-07-31 2022-02-03 药捷安康(南京)科技股份有限公司 成纤维细胞生长因子受体抑制剂的晶型及其制备方法

Also Published As

Publication number Publication date
KR102270620B1 (ko) 2021-06-30
CN107793395B (zh) 2021-01-05
KR20190039583A (ko) 2019-04-12
EP3508482A4 (en) 2019-07-31
EP3508482A1 (en) 2019-07-10
ES2859637T3 (es) 2021-10-04
JP2019529374A (ja) 2019-10-17
AU2017317864B2 (en) 2020-01-16
JP6751977B2 (ja) 2020-09-09
EP3508482B1 (en) 2020-12-16
RU2732127C1 (ru) 2020-09-11
CN107793395A (zh) 2018-03-13
US20190209564A1 (en) 2019-07-11
US20200108070A2 (en) 2020-04-09
AU2017317864A1 (en) 2019-03-28
US10744139B2 (en) 2020-08-18

Similar Documents

Publication Publication Date Title
WO2018040885A1 (zh) 成纤维细胞生长因子受体抑制剂及其用途
CN109843873B (zh) 炔代杂环化合物、其制备方法及其在医药学上的应用
CN115873020A (zh) Ras抑制剂
RU2677667C2 (ru) Соединения n-пирролидинилмочевины, n'-пиразолилмочевины, тиомочевины, гуанидина и цианогуанидина как ингибиторы киназы trka
TW202016109A (zh) 作為hpk1抑制劑的吡咯並[2,3-b]吡啶或吡咯並[2,3-b]吡嗪及其用途
TWI677499B (zh) Pde9抑制劑及其用途
CN105566321B (zh) 杂芳化合物及其在药物中的应用
TWI669300B (zh) 嘧啶類衍生物、其製備方法、其藥物組合物以及其在醫藥上的用途
TW201311685A (zh) 咪唑並吡啶化合物
WO2020143763A1 (zh) 卤代烯丙基胺类化合物及其应用
JP2019502714A (ja) Nik阻害剤としての新たな6員のヘテロ芳香族置換シアノインドリン誘導体
WO2023025320A1 (zh) 含氮杂环类衍生物抑制剂、其制备方法和应用
TW201910331A (zh) 成纖維細胞生長因子受體抑制劑、含有其的藥物製劑及其用途
JP2020537692A (ja) ヘテロ環式化合物、ヘテロ環式化合物を含む組成物、及びその使用方法
TW202321219A (zh) 雜環衍生物及其組合物和藥學上的應用
KR20230146639A (ko) 이미다졸 함유 축합고리계 유도체, 이의 제조방법 및 이의 의약적 용도
WO2022166642A1 (zh) 含氮多环稠环类化合物,其药物组合物、制备方法和用途
WO2021080015A1 (en) Modulators of trek (twik related k+ channels) channel function
TWI727152B (zh) 炔代雜環化合物、其製備方法及其在醫藥學上的應用
CN117430606A (zh) 磺酰胺类parp7抑制剂
WO2023236960A1 (zh) 一种具有rsk抑制作用的羧酰胺衍生物、包含其的药物组合物及其用途
CN113087724A (zh) 异噻唑并嘧啶酮类化合物,包含其的药物组合物及其用途
WO2024097953A1 (en) Naphthyridine compounds for inhibition of raf kinases

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: 17845187

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2019511715

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: 20197007909

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017317864

Country of ref document: AU

Date of ref document: 20170810

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017845187

Country of ref document: EP

Effective date: 20190401