WO2019047826A1 - 芳香类衍生物、其制备方法及其在医药上的应用 - Google Patents

芳香类衍生物、其制备方法及其在医药上的应用 Download PDF

Info

Publication number
WO2019047826A1
WO2019047826A1 PCT/CN2018/104007 CN2018104007W WO2019047826A1 WO 2019047826 A1 WO2019047826 A1 WO 2019047826A1 CN 2018104007 W CN2018104007 W CN 2018104007W WO 2019047826 A1 WO2019047826 A1 WO 2019047826A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
independently selected
haloalkyl
mmol
Prior art date
Application number
PCT/CN2018/104007
Other languages
English (en)
French (fr)
Inventor
邵宁
王丁
袁宏斌
凯泽⋅弗兰克
Original Assignee
博奥阿迪斯生物科技公司
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 博奥阿迪斯生物科技公司 filed Critical 博奥阿迪斯生物科技公司
Priority to CN201880034411.9A priority Critical patent/CN110997637B/zh
Priority to EP18854268.2A priority patent/EP3680236A4/en
Priority to US16/644,919 priority patent/US11279697B2/en
Priority to KR1020207009736A priority patent/KR102677016B1/ko
Priority to SG11202001979PA priority patent/SG11202001979PA/en
Priority to CA3074885A priority patent/CA3074885A1/en
Priority to JP2020533340A priority patent/JP7346420B2/ja
Publication of WO2019047826A1 publication Critical patent/WO2019047826A1/zh
Priority to PH12020500436A priority patent/PH12020500436A1/en
Priority to JP2023144389A priority patent/JP2023158113A/ja

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/12Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/4161,2-Diazoles condensed with carbocyclic ring systems, e.g. indazole
    • 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
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/02Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having nitrogen atoms of carboxamide groups bound to hydrogen atoms or to carbon atoms of unsubstituted hydrocarbon radicals
    • C07C233/09Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having nitrogen atoms of carboxamide groups bound to hydrogen atoms or to carbon atoms of unsubstituted hydrocarbon radicals with carbon atoms of carboxamide groups bound to carbon atoms of an acyclic unsaturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/38Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • C07D231/56Benzopyrazoles; Hydrogenated benzopyrazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D403/10Heterocyclic 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 carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention relates to a novel aromatic derivative or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer, diastereomer, prodrug, hydrate or solvent thereof And mixtures thereof, or pharmaceutical compositions containing the same, and methods for their preparation.
  • the present invention also relates to a method for treating and/or preventing a FGFR4 tyrosine kinase-mediated disease using the aromatic ether derivative, and the preparation of the FGFR4 tyrosine kinase inhibitor and the medicament by the aromatic ether derivative Use in.
  • Fibroblast growth factor is a polypeptide secreted by the pituitary and hypothalamus. It promotes mitosis of fibroblasts, growth of mesoderm cells, stimulates angiogenesis, and plays an important role in wound healing and limb regeneration.
  • the FGF receptor (FGFR) signaling system is critical for normal development and physiological processes.
  • FGFR has four subtypes of FGFR1-4, and the amino acid sequences of these four subtypes are highly conserved.
  • FGFR1-4 has different binding forces for different growth factors, and its distribution in tissues is also different.
  • a complete FGFR receptor protein includes an extracellular portion, a hydrophobic single-stranded cell membrane portion, and an intracellular tyrosine kinase portion.
  • FGFR4 is a protein encoded by the FGFR-4 gene.
  • the FGFR4 gene has 18 exons.
  • FGF-FGFR signal imbalance is associated with tumorigenesis and evolution. It has been found that in mice, the FGFR4–FGF19 signal axis is closely related to hepatocellular carcinoma (HCCs). In several types of cancers such as liver cancer, FGFR4 expression is significantly increased. At the same time, the occurrence of liver cancer requires FGFR4.
  • the progeny of FGF19 transgenic mice can develop into liver cancer, however, the offspring proliferated with FGFR4 knockout mice do not develop liver cancer.
  • FGF19 is neutralized by FGF19-specific antibodies, tumor growth is inhibited.
  • FGFR4 overexpression also occurs in other types of tumors, including breast cancer, colorectal cancer, pancreatic cancer, prostate cancer, lung cancer, and thyroid cancer.
  • FGFR4 mutations occur in rhabdomyosarcoma. Small molecule targeted inhibition of FGFR4 can be used in the treatment of cancer.
  • mice were treated with FGFR-1 inhibitors they were found to cause side effects such as calcium phosphate deposition in soft tissues (Brown, AP et al. (2005), Toxicol. Pathol., p. 449-455). These indicate that selective inhibition of the FGFR4 receptor, rather than extensive inhibition of the FGFR1-4 receptor, would avoid such side effects.
  • the invention provides a compound of formula (I), or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer, diastereomer thereof, Medicine, hydrate or solvate, and mixtures thereof:
  • Ring A is absent or is selected from a 6-14 membered arylene group, a 5-10 membered heteroarylene group, a C 3 -C 8 cycloalkylene group or a 3-10 membered heterocyclylene group;
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 Alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, -OR 13 , -NR 11 R 12 , -C(O)R 13 , -C(O)OR 13 , -C(O )NR 11 R 12 , -NR 11 C(O)R 13 , -NR 11 C(O)OR 13 , -NR 11 C(O)NR 11 R 12 , -OC(O)R 13 , -OC(O )OR 13 , -OC(O)NR 11 R 12 , -S(O) p R 13 , -S(O) p OR 13 , -S(O) p NR 11 R 12 , -
  • R 6 is each independently selected from H or -CH 2 CH 2 -, and when R 6 is -CH 2 CH 2 -, the other end thereof is bonded to X, optionally one of -CH 2 CH 2 - The base is replaced by -O- or -NH-;
  • R 7 is each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3- C 8 cycloalkyl, -OR 13 , or -NR a R b ; wherein R a and R b are each independently selected from H or C 1 -C 6 alkyl optionally substituted by -NR c R d ; R c and R d are each independently selected from H or C 1 -C 6 alkyl;
  • R 8 is selected from an optionally substituted C 2 -C 6 alkenyl group or a C 2 -C 6 alkynyl group;
  • R 11 , R 12 and R 13 are each independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 8 cycloalkyl, 3-10 membered heterocyclyl, 5 -10 membered heteroaryl or 6-14 membered aryl;
  • X is selected from -O-, -NH- or -CH 2 -; Y is selected from -O-, -NH- or -CH 2 -; with the proviso that at least one of X and Y is CH 2 ;
  • W is selected from a chemical bond, -NH- or -CH 2 -;
  • n 0, 1, 2, 3 or 4;
  • p 1 or 2.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer, diastereomer thereof , prodrugs, hydrates or solvates, and mixtures thereof, and pharmaceutically acceptable excipients.
  • a compound of the invention is provided in the pharmaceutical composition in an effective amount.
  • the compounds of the invention are provided in a therapeutically effective amount.
  • the compounds of the invention are provided in a prophylactically effective amount.
  • the pharmaceutical compositions of the invention also contain other therapeutic agents.
  • the invention provides a kit comprising: a first container comprising a compound of the invention or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer thereof a diastereomer, a prodrug, a hydrate or a solvate, and mixtures thereof; and, optionally, a second container containing other therapeutic agents; and optionally, a third container containing A pharmaceutically acceptable excipient that dilutes or suspends the compound and/or other therapeutic agent.
  • the invention provides a compound of the invention, or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer, diastereomer, prodrug, hydrate thereof, or a pharmaceutically acceptable salt thereof Or a solvate, and mixtures thereof, or the use of a pharmaceutical composition of the invention in the preparation of a FGFR4 tyrosine kinase inhibitor.
  • the invention provides a compound of the invention, or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer, diastereomer, prodrug, hydrate thereof, or a pharmaceutically acceptable salt thereof Or a solvate, and mixtures thereof, or the use of a pharmaceutical composition of the invention in the manufacture of a medicament for the treatment and/or prevention of a FGFR4 tyrosine kinase mediated disease.
  • the invention provides a compound of the invention, or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer, diastereomer, prodrug, hydrate thereof, or a pharmaceutically acceptable salt thereof Or a solvate, and mixtures thereof, or a pharmaceutical composition of the invention for use in the treatment and/or prevention of a FGFR4 tyrosine kinase mediated disease.
  • the invention provides a method of treating and/or preventing a FGFR4 tyrosine kinase mediated disease in a subject, comprising administering to the subject a compound of the invention or Pharmaceutically acceptable salts, tautomers, racemates, enantiomers, diastereomers, prodrugs, hydrates or solvates, and mixtures thereof, or pharmaceutical compositions of the invention .
  • the disease is a tumor, such as gastric cancer, thyroid cancer, prostate cancer, breast cancer, sarcoma (such as rhabdomyosarcoma), skin cancer (such as melanoma), liver cancer (such as hepatocellular carcinoma and bile duct).
  • Epithelial cancer eg, pancreatic intraepithelial neoplasia and pancreatic ductal adenocarcinoma
  • lung cancer eg, non-small cell lung cancer and lung adenocarcinoma
  • renal cancer eg, renal cell carcinoma
  • colorectal cancer ovarian cancer.
  • C 1 -C 6 alkyl includes C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1 -C 6 , C 1 -C 5 , C 1 -C 4 , C 1 - C 3 , C 1 -C 2 , C 2 -C 6 , C 2 -C 5 , C 2 -C 4 , C 2 -C 3 , C 3 -C 6 , C 3 -C 5 , C 3 -C 4 C 4 -C 6 , C 4 -C 5 and C 5 -C 6 alkyl.
  • C 1 -C 6 alkyl group means a straight or branched saturated hydrocarbon group having 1 to 6 carbon atoms, which is also referred to as "lower alkyl group”. In some embodiments, a C 1 -C 4 alkyl group is particularly preferred.
  • alkenyl group examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, sec-butyl, n-pentyl, 1,1-di Methylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1 -ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethyl Butyl, 1,3-dimethylbutyl, 2-ethylbutyl, and various branched isomers thereof.
  • the alkyl group can be optionally substituted or unsubstituted.
  • C 2 -C 6 alkenyl refers to a straight or branched chain hydrocarbon radical having from 2 to 6 carbon atoms and one or more carbon-carbon double bonds (eg, 1, 2 or 3 carbon-carbon double bonds) group.
  • One or more carbon-carbon double bonds may be internal (eg, in 2-butenyl) or end (eg, in 1-butenyl).
  • C 2 -C 4 alkenyl is particularly preferred.
  • alkenyl group examples include, but are not limited to, ethenyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, butadienyl, pentenyl, pentadienyl, Hexenyl, its various branched isomers, and the like.
  • the alkenyl group can be optionally substituted or unsubstituted.
  • C 2 -C 6 alkynyl means having from 2 to 6 carbon atoms, one or more carbon-carbon triple bonds (eg, 1, 2 or 3 carbon-carbon triple bonds), and optionally one or more A linear or branched hydrocarbon group of a carbon-carbon double bond (for example, 1, 2 or 3 carbon-carbon double bonds).
  • a C 2 -C 4 alkynyl group is particularly preferred.
  • an alkynyl group does not contain any double bonds.
  • the one or more carbon oxime bonds may be internal (eg, in 2-butynyl) or end (eg, in 1-butynyl).
  • alkynyl group examples include, but are not limited to, ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, pentynyl, hexynyl, and various Branched isomers, etc.
  • An alkynyl group can be optionally substituted or unsubstituted.
  • C 1 -C 6 heteroalkyl refers to an alkyl group, as defined herein, which further contains one or more (eg, 1, 2, 3 or 4) heteroatoms (eg, oxygen, in the parent chain). Sulfur, nitrogen, boron, silicon, phosphorus), wherein one or more heteroatoms are between adjacent carbon atoms in the parent carbon chain, and/or one or more heteroatoms are in the carbon atom and the parent molecule Between, that is, between the connection points. Heteroalkyl groups can be optionally substituted or unsubstituted.
  • the C 1 -C 6 heteroalkyl group includes a C 1 -C 6 alkoxy group, a C 1 -C 6 alkylthio group, a C 1 -C 6 alkylamino group, and the like, which are defined in detail as follows .
  • C 1 -C 6 alkoxy refers to the group -OR wherein R is a substituted or unsubstituted C 1 -C 6 alkyl group. In some embodiments, a C 1 -C 4 alkoxy group is particularly preferred. Specific alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentyloxy, n-Hexyloxy and 1,2-dimethylbutoxy, and various branched isomers thereof and the like. The alkoxy group can be optionally substituted or unsubstituted.
  • C 1 -C 6 alkylthio refers to the group -SR wherein R is optionally substituted C 1 -C 6 alkyl.
  • a C 1 -C 4 alkylthio group is particularly preferred.
  • the C 1 -C 6 alkylthio group includes, but is not limited to, methylthio, ethylthio, n-propylthio, isopropylthio, n-butylthio, tert-butylthio, sec-butylthio, N-pentylthio, n-hexylthio and 1,2-dimethylbutylthio, and various branched isomers thereof.
  • the alkylthio group can be optionally substituted or unsubstituted.
  • C 1 -C 6 alkylamino refers to the group -NHR or -NR 2 wherein R is optionally substituted C 1 -C 6 alkyl.
  • a C 1 -C 4 alkylamino group is particularly preferred.
  • the C 1 -C 6 alkylamino group includes, but is not limited to, methylamino, ethylamino, n-propylamino, isopropylamino, n-butylamino, t-butylamino, dimethylamino, methylethylamino and diethylamino, and Its various branched isomers and the like.
  • the alkylamino group can be optionally substituted or unsubstituted.
  • Halogen means fluorine (F), chlorine (Cl), bromine (Br) and iodine (I).
  • C 1 -C 6 haloalkyl and “C 1 -C 6 haloalkoxy” mean the above-mentioned "C 1 -C 6 alkyl” and "C 1 -C 6 alkoxy", which are one or more Halogen group substitution.
  • a C 1 -C 4 haloalkyl group is particularly preferred, more preferably a C 1 -C 2 haloalkyl group.
  • a C 1 -C 4 haloalkoxy group is particularly preferred, more preferably a C 1 -C 2 haloalkoxy group.
  • haloalkyl groups include, but the are not limited to: -CF 3, -CH 2 F, -CHF 2, -CHFCH 2 F, -CH 2 CHF 2, -CF 2 CF 3, -CCl 3, -CH 2 Cl , -CHCl 2 , 2,2,2-trifluoro-1,1-dimethyl-ethyl, and the like.
  • exemplary haloalkoxy groups include, but are not limited to, -OCH 2 F, -OCHF 2 , -OCF 3, and the like.
  • the haloalkyl and haloalkoxy groups can be optionally substituted or unsubstituted.
  • C 3 -C 8 cycloalkyl refers to a non-aromatic cyclic hydrocarbon group having from 3 to 8 ring carbon atoms and zero heteroatoms. In some embodiments, a C 3 -C 6 cycloalkyl group is particularly preferred, more preferably a C 5 -C 6 cycloalkyl group.
  • Cycloalkyl also includes ring systems wherein the cycloalkyl group is fused, bridged or spirocyclic to one or more cycloalkyl, heterocyclyl, aryl or heteroaryl groups, wherein the point of attachment is on the cycloalkyl ring And in such cases, the number of carbons continues to indicate the number of carbons in the cycloalkyl system.
  • cycloalkyl groups include, but are not limited to, cyclopropyl, cyclopropenyl, cyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexane Alkenyl, cycloheptyl, cycloheptenyl, cycloheptadienyl, cycloheptatrienyl, cyclooctyl, cyclooctenyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.2] Octyl, its various branched isomers, and the like.
  • the cycloalkyl group can be optionally substituted or unsubstituted.
  • heterocyclic group or a group of a 3 to 10 membered non-aromatic ring system having a ring carbon atom and 1 to 4 ring hetero atoms, wherein each hetero atom is independently selected from nitrogen and oxygen. , sulfur, boron, phosphorus and silicon.
  • the point of attachment may be a carbon or nitrogen atom as long as the valence permits.
  • a 3-8 membered heterocyclyl is preferred, which is a 3 to 8 membered non-aromatic ring system having a ring carbon atom and 1 to 3 ring heteroatoms; in some embodiments, 4-7 A heterocyclic group is particularly preferred, which is a 4- to 7-membered non-aromatic ring system having a ring carbon atom and 1 to 3 ring hetero atoms; more preferably a 5-6 membered heterocyclic group having a ring carbon atom and A 5- to 6-membered non-aromatic ring system of 1 to 3 ring heteroatoms.
  • Exemplary 3-membered heterocyclic groups containing one hetero atom include, but are not limited to, aziridine, oxacyclopropane, thicyclopropyl.
  • Exemplary 4-membered heterocyclic groups containing one hetero atom include, but are not limited to, azetidinyl, oxetanyl and thietane.
  • Exemplary 5-membered heterocyclic groups containing one hetero atom include, but are not limited to, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothiophenyl, dihydrothienyl, pyrrolidinyl, dihydropyrrolyl and pyrrolyl-2, 5-dione.
  • Exemplary 5-membered heterocyclic groups containing two heteroatoms include, but are not limited to, dioxolyl, oxathiolan, dithiolane, and oxazolidin-2-one .
  • Exemplary 5-membered heterocyclic groups containing three heteroatoms include, but are not limited to, triazolinyl, oxadiazolyl, and thiadiazolyl.
  • Exemplary 6-membered heterocyclic groups containing one hetero atom include, but are not limited to, piperidinyl, dihydropyranyl, tetrahydropyranyl, dihydropyridyl and thiacyclohexane.
  • Exemplary 6-membered heterocyclic groups containing two heteroatoms include, but are not limited to, piperazinyl, morpholinyl, dithianyl, dioxoalkyl.
  • Exemplary 6-membered heterocyclic groups containing three heteroatoms include, but are not limited to, hexahydrotriazinyl.
  • Exemplary 7-membered heterocyclic groups containing one hetero atom include, but are not limited to, azepanyl, oxaheptyl, and thiaheptanyl.
  • Exemplary 8-membered heterocyclic groups containing one hetero atom include, but are not limited to, azacyclooctyl, oxacyclooctyl, and thicyclooctyl.
  • Exemplary 5-membered heterocyclic groups (also referred to herein as 5,6-bicyclic heterocyclyl) fused to a 6-membered aryl ring include, but are not limited to, indanyl, isoindoline , dihydrobenzofuranyl, dihydrobenzothiophenyl, benzoxazolinone, and the like.
  • Exemplary 6-membered heterocyclic groups fused to a 6-membered aryl ring include, but are not limited to, tetrahydroquinolyl, tetrahydroisoquinolinyl, and many more.
  • the heterocyclic group includes a spiro ring, a fused ring, and a heterocyclic group of a bridged ring.
  • “Spiroheterocyclyl” means a polycyclic heterocyclic group of 5 to 20 members in which one atom (referred to as a spiro atom) is shared between monocyclic rings, wherein one or more ring atoms are selected from nitrogen, oxygen or S(O) m
  • the hetero atom (where m is an integer from 0 to 2) and the remaining ring atoms are carbon. These may contain one or more double bonds, but none of the rings are aromatic. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • the spirocycloalkyl group is classified into a monospiroheterocyclic group, a dispiroheterocyclic group or a polyspirocyclic group according to the number of common spiro atoms between the ring and the ring, and is preferably a monospirocycloalkyl group and a bispirocycloalkyl group. More preferably, it is 4 yuan / 4 yuan, 4 yuan / 5 yuan, 4 yuan / 6 yuan, 5 yuan / 5 yuan or 5 yuan / 6-membered monospirocycloalkyl group.
  • Non-limiting examples of spirocycloalkyl groups include:
  • “Fused heterocyclyl” refers to 5 to 20 members, each ring of the system sharing an adjacent pair of atoms of a polycyclic heterocyclic group with other rings in the system, and one or more rings may contain one or more A bond, but none of the rings are aromatic in which one or more ring atoms are selected from the group consisting of nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), and the remaining ring atoms are carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • fused heterocyclic groups include:
  • the heterocyclyl ring may be fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring to which the parent structure is attached is a heterocyclic group, non-limiting examples include:
  • the heterocyclic group may be optionally substituted or unsubstituted.
  • 6-14 membered aryl refers to a monocyclic or polycyclic (eg, bicyclic or tricyclic) 4n+2 aromatic ring system having 6 to 14 ring carbon atoms and zero heteroatoms (eg, having A group of 6, 10 or 14 ⁇ electrons shared in a ring.
  • an aryl group has six ring carbon atoms ("6-membered aryl”; for example, phenyl).
  • an aryl group has ten ring carbon atoms ("10 membered aryl”; for example, naphthyl, for example, 1-naphthyl and 2-naphthyl).
  • an aryl group has fourteen ring carbon atoms ("14-membered aryl"; for example, fluorenyl).
  • 14-membered aryl for example, fluorenyl
  • a 6-10 membered aryl group is particularly preferred, more preferably a 6 membered aryl group.
  • the aryl group may be fused to a heteroaryl, heterocyclic or cycloalkyl ring, wherein the ring to which the parent structure is attached is an aryl ring, non-limiting examples include:
  • the aryl group can be substituted or unsubstituted.
  • 5-10 membered heteroaryl means a 5-10 membered monocyclic or bicyclic 4n+2 aromatic ring system having a ring carbon atom and 1-4 ring heteroatoms (eg, having a ring-like arrangement) a group of 6 or 10 ⁇ electrons, wherein each heteroatom is independently selected from the group consisting of nitrogen, oxygen and sulfur.
  • a heteroaryl group containing one or more nitrogen atoms the point of attachment may be a carbon or nitrogen atom as long as the valence permits.
  • a 5-6 membered heteroaryl group is particularly preferred, which is a 5-6 membered monocyclic 4n+2 aromatic ring system having a ring carbon atom and 1-4 ring heteroatoms.
  • Exemplary 5-membered heteroaryl groups containing one hetero atom include, but are not limited to, pyrrolyl, furyl and thienyl.
  • Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl.
  • Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to, triazolyl, oxadiazolyl, and thiadiazolyl.
  • Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to, tetrazolyl.
  • Exemplary 6-membered heteroaryl groups containing one hetero atom include, but are not limited to, pyridyl.
  • Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to, pyridazinyl, pyrimidinyl, and pyrazinyl.
  • Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to, triazinyl and tetrazinyl, respectively.
  • Exemplary 7-membered heteroaryl groups containing one hetero atom include, but are not limited to, azepandinyl, oxepanethylene, and thiephenylene.
  • Exemplary 5,6-bicyclic heteroaryl groups include, but are not limited to, mercapto, isodecyl, oxazolyl, benzotriazolyl, benzothienyl, isobenzothienyl, benzofuranyl , benzoisofuranyl, benzimidazolyl, benzoxazolyl, benzoisoxazolyl, benzooxadiazolyl, benzothiazolyl, benzisothiazolyl, benzothiadiazolyl, Pyridazinyl and fluorenyl.
  • Exemplary 6,6-bicyclic heteroaryl groups include, but are not limited to, naphthyridinyl, acridinyl, quinolyl, isoquinolinyl, fluorenyl, quinoxalinyl, pyridazinyl and quinazolinyl .
  • the heteroaryl ring may be fused to an aryl, heterocyclic or cycloalkyl ring, wherein the ring to which the parent structure is attached is a heteroaryl ring, non-limiting examples include:
  • the heteroaryl group can be optionally substituted or unsubstituted.
  • alkylene alkenylene, alkynylene, arylene, “heteroarylene”, “cycloalkylene” and “heterocyclylene” refer to It is a divalent group of the above alkyl group, alkenyl group, alkynyl group, aryl group, heteroaryl group, cycloalkyl group and heterocyclic group. The group may be optionally substituted or unsubstituted.
  • C 1 -C 6 alkylene group means a divalent alkylene group formed by removing one hydrogen of a C 1 -C 6 alkyl group, and may be a substituted or unsubstituted alkylene group. In some embodiments, a C 1 -C 4 alkylene group is particularly preferred.
  • the unsubstituted alkylene group includes, but is not limited to, methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), propylene (-CH 2 CH 2 CH 2 -), arylene (-CH 2 CH 2 CH 2 CH 2 -), pentylene (-CH 2 CH 2 CH 2 CH 2 CH 2 -), hexylene (-CH 2 CH 2 CH 2 CH 2 CH 2 CH 2 -) ,and many more.
  • alkylene groups substituted with one or more alkyl groups (methyl groups) include, but are not limited to, substituted methylene groups (-CH(CH 3 )- , -C(CH 3 ) 2 -), substituted ethylene (-CH(CH 3 )CH 2 -, -CH 2 CH(CH 3 )-, -C(CH 3 ) 2 CH 2 -, -CH 2 C(CH 3 ) 2- ), substituted propylene (-CH(CH 3 )CH 2 CH 2 -, -CH 2 CH(CH 3 )CH 2 -, -CH 2 CH 2 CH(CH 3 ) -, -C(CH 3 ) 2 CH 2 CH 2 -, -CH 2 C(CH 3 ) 2 CH 2 -, -CH 2 CH 2 C(CH 3 ) 2 -), and the like.
  • substituted methylene groups -CH(CH 3 )- , -C(CH 3 ) 2 -
  • substituted ethylene -CH(CH
  • C 2 -C 6 alkenylene group means a divalent alkenylene group formed by removing one hydrogen of a C 2 -C 6 alkenyl group, and may be a substituted or unsubstituted alkenylene group. In some embodiments, C 2 -C 4 alkenylene is particularly preferred.
  • C 2 -C 6 alkynylene group means a divalent alkynylene group formed by removing one hydrogen of a C 2 -C 6 alkynyl group, and may be a substituted or unsubstituted alkynylene group. In some embodiments, a C 2 -C 4 alkynylene group is particularly preferred. Exemplary such alkynylene groups include, but are not limited to, ethynylene (-C ⁇ C-), substituted or unsubstituted propynylene (-C ⁇ CCH 2 -), and the like.
  • 6-14 membered arylene means the removal of the above 6 a divalent group formed by a hydrogen of a 14-membered aryl group, a 5-10 membered heteroaryl group, a C 3 -C 8 cycloalkyl group, and a 3-10 membered heterocyclic group.
  • heterocyclic group optionally substituted by an alkyl group means that an alkyl group may be, but is not necessarily, present, and the description includes the case where the heterocyclic group is substituted with an alkyl group and the case where the heterocyclic group is not substituted with an alkyl group.
  • Substituted refers to one or more hydrogen atoms in the group, preferably up to 5, more preferably 1 to 3, hydrogen atoms, independently of each other, substituted by a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and those skilled in the art will be able to determine (by experiment or theory) substitutions that may or may not be possible without undue effort. For example, an amino group or a hydroxyl group having a free hydrogen may be unstable when combined with a carbon atom having an unsaturated (e.g., olefinic) bond.
  • Each of R aa is independently selected from alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, or two R aa groups are bonded to form a heterocyclic group or a heteroaryl ring wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl group is independently 0, 1, 2, 3, 4 or 5 R dd groups Substitute
  • Each of R cc is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, or two R cc groups are bonded to form a heterocyclic ring a hetero or heteroaryl ring wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl group is independently 0, 1, 2, 3, 4 or 5 R Substituting dd group;
  • Each of R ee is independently selected from the group consisting of alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocyclyl and heteroaryl, wherein each alkyl, alkenyl, alkynyl, ring
  • An alkyl group, a heterocyclic group, an aryl group and a heteroaryl group are independently substituted by 0, 1, 2, 3, 4 or 5 R gg groups;
  • Each of R ff is independently selected from hydrogen, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, or two R ff groups are bonded to form a heterocyclic group Or a heteroaryl ring wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl group is independently 0, 1, 2, 3, 4 or 5 R gg Group substitution
  • pharmaceutically acceptable salt means that, within the scope of sound medical judgment, it is suitable for contact with tissues of humans and lower animals without excessive toxicity, irritation, allergies, etc., and with reasonable benefits/risk ratios. Proportionate of those salts.
  • Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts as described in detail by Berge et al., J. Pharmaceutical Sciences (1977) 66: 1-19.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and inorganic and organic bases.
  • non-toxic acid addition salts examples include salts with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid, or salts with organic acids such as acetic acid, oxalic acid, Maleic acid, tartaric acid, citric acid, succinic acid or malonic acid. Also included are salts formed using conventional methods in the art, for example, ion exchange methods.
  • adipic acid salts alginate, ascorbate, aspartate, besylate, benzoate, disulfate, borate, butyrate, camphoric acid Salt, camphor sulfonate, citrate, cyclopentanoate, digluconate, lauryl sulfate, ethanesulfonate, formate, fumarate, gluconate, glycerophosphate Salt, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, Malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate , pectic acid ester
  • Pharmaceutically acceptable salts derived from suitable bases include the alkali metal, alkaline earth metal, ammonium and N + (C 1-4 alkyl) 4 salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium salts, and the like.
  • other pharmaceutically acceptable salts include non-toxic ammonium salts, quaternary ammonium salts and amine cations formed with counterions, counterions such as halides, hydroxides, carboxylates, sulfates, phosphates, nitric acid Root, lower alkyl sulfonate and aryl sulfonate.
  • Subjects for administration include, but are not limited to, humans (ie, males or females of any age group, eg, pediatric subjects (eg, infants, children, adolescents) or adult subjects (eg, young Adults, middle-aged adults or older adults) and/or non-human animals, for example, mammals, for example, primates (eg, cynomolgus monkeys, rhesus monkeys), cattle, pigs, horses, sheep , goats, rodents, cats and/or dogs.
  • the subject is a human.
  • the subject is a non-human animal.
  • treatment includes the effect of a subject having a particular disease, disorder, or condition that reduces the severity of the disease, disorder, or condition, or delays or slows the disease, disorder. Or the development of a condition ("therapeutic treatment"), which also includes the effect that occurs before the subject begins to have a particular disease, disorder or condition (“prophylactic treatment").
  • an "effective amount" of a compound refers to an amount sufficient to cause a target biological response.
  • an effective amount of a compound of the invention can vary depending on, for example, the biological target, the pharmacokinetics of the compound, the condition being treated, the mode of administration, and the subject's Age health conditions and symptoms. Effective amounts include therapeutically effective amounts and prophylactically effective amounts.
  • a "therapeutically effective amount" of a compound as used herein is an amount sufficient to provide a therapeutic benefit in the course of treating a disease, disorder or condition, or one or more symptoms associated with a disease, disorder or condition. The amount of delay or minimization.
  • a therapeutically effective amount of a compound refers to the amount of a therapeutic agent that, when used alone or in combination with other therapies, provides a therapeutic benefit in the treatment of a disease, disorder or condition.
  • the term "therapeutically effective amount” can include an amount that improves overall treatment, reduces or avoids the symptoms or causes of the disease or condition, or enhances the therapeutic effect of other therapeutic agents.
  • a “prophylactically effective amount” of a compound is an amount sufficient to prevent a disease, disorder or condition, or an amount sufficient to prevent one or more symptoms associated with a disease, disorder or condition, or to prevent disease, unless otherwise stated. The amount of recurrence of a disorder or condition.
  • a prophylactically effective amount of a compound refers to the amount of a therapeutic agent that, when used alone or in combination with other agents, provides a prophylactic benefit in the prevention of a disease, disorder or condition.
  • the term “prophylactically effective amount” can include an amount that improves the overall prevention, or an amount that enhances the prophylactic effect of other prophylactic agents.
  • Combination and related terms mean the simultaneous or sequential administration of a compound of the invention and other therapeutic agents.
  • the compounds of the invention may be administered simultaneously or sequentially with other therapeutic agents in separate unit dosage forms, or together with other therapeutic agents, in a single unit dosage form.
  • Prodrug means a compound that is converted in vivo to an active form thereof having a medical effect by, for example, hydrolysis in blood.
  • Pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems, ACSSymposium Series, Vol. 14, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and D. Fleisher, S. Ramon, and H. Barbra "Improved oral drug delivery: Solubility limitations overcome by the use of prodrugs", Advanced Drug Delivery Reviews (1996) 19(2) 115-130, each This article is incorporated herein by reference.
  • a prodrug is any covalently bonded carrier which, when administered to a patient, releases the compound of the invention in vivo.
  • Prodrugs are typically prepared by modifying functional groups that cleave the prodrug in vivo to yield the parent compound.
  • Prodrugs include, for example, a compound of the invention wherein a hydroxy, amino or thiol group is bonded to any group which, when administered to a patient, can be cleaved to form a hydroxy, amino or thiol group.
  • representative examples of prodrugs include, but are not limited to, covalent derivatives of the compounds of the invention formed by the hydroxyl, amino or thiol functional groups thereof with acetic acid, formic acid or benzoic acid.
  • an ester such as a methyl ester, an ethyl ester or the like can be used.
  • the ester itself may be active and/or may hydrolyze under conditions in humans.
  • Suitable pharmaceutically acceptable in vivo hydrolysable esters include those which readily decompose in the human body to release the parent acid or its salt.
  • the compounds of the invention may include one or more asymmetric centers, and thus may exist in a variety of stereoisomeric forms, for example, enantiomers, diastereomers, and racemate forms.
  • the compounds of the invention may be in the form of individual enantiomers, diastereomers or geometric isomers (e.g., cis and trans isomers), or may be in the form of a mixture of stereoisomers, A racemic mixture and a mixture rich in one or more stereoisomers are included.
  • the isomers can be separated from the mixture by methods known to those skilled in the art, including: chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of a chiral salt; or preferred isomers can be passed Prepared by asymmetric synthesis.
  • HPLC high pressure liquid chromatography
  • the compounds of the invention may also exist as “tautomers".
  • “Tautomer” refers to an isomer which is produced by the phenomenon that the structure of an organic compound produces a balance between two functional groups.
  • the present invention provides a compound of the formula (I) or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer, diastereomer thereof, or a diastereomer thereof Constructs, prodrugs, hydrates or solvates, and mixtures thereof:
  • Ring A is absent or is selected from a 6-14 membered arylene group, a 5-10 membered heteroarylene group, a C 3 -C 8 cycloalkylene group or a 3-10 membered heterocyclylene group;
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 Alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, -OR 13 , -NR 11 R 12 , -C(O)R 13 , -C(O)OR 13 , -C(O )NR 11 R 12 , -NR 11 C(O)R 13 , -NR 11 C(O)OR 13 , -NR 11 C(O)NR 11 R 12 , -OC(O)R 13 , -OC(O )OR 13 , -OC(O)NR 11 R 12 , -S(O) p R 13 , -S(O) p OR 13 , -S(O) p NR 11 R 12 , -
  • R 6 is each independently selected from H or -CH 2 CH 2 -, and when R 6 is -CH 2 CH 2 -, the other end thereof is bonded to X, optionally one of -CH 2 CH 2 - The base is replaced by -O- or -NH-;
  • R 7 is each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3- C 8 cycloalkyl, -OR 13 , or -NR a R b ; wherein R a and R b are each independently selected from H or C 1 -C 6 alkyl optionally substituted by -NR c R d ; R c and R d are each independently selected from H or C 1 -C 6 alkyl; or, R a , R b and the N atom to which they are attached together form a 3-10 membered heterocyclic group (eg, 6-membered heterocyclic ring). a group optionally substituted by a C 1 -C 6 alkyl group or a C 1 -C 6 haloalkyl group;
  • R 8 is selected from an optionally substituted C 2 -C 6 alkenyl group or a C 2 -C 6 alkynyl group;
  • R 11 , R 12 and R 13 are each independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 8 cycloalkyl, 3-10 membered heterocyclyl, 5 -10 membered heteroaryl or 6-14 membered aryl;
  • X is selected from -O-, -NH- or -CH 2 -; Y is selected from -O-, -NH- or -CH 2 -; with the proviso that at least one of X and Y is CH 2 ;
  • W is selected from a chemical bond, -NH- or -CH 2 -;
  • n 0, 1, 2, 3 or 4;
  • p 1 or 2.
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from H, halo, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 Alkoxy or C 1 -C 6 haloalkoxy.
  • R 1 and R 5 are each independently selected from halogen, preferably Cl or F.
  • R 2 and R 4 are each independently selected from C 1 -C 6 alkoxy, preferably methoxy.
  • R 3 is H.
  • R 6 is selected from H.
  • each R 7 is independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or -NR a R b ; wherein R a and R b are each independently selected from H Or a C 1 -C 6 alkyl group optionally substituted by -NR c R d ; R c and R d are each independently selected from H or C 1 -C 6 alkyl, or R a , R b and they are attached The N atoms together form a 3-10 membered heterocyclyl (eg 6-membered heterocyclyl) which is optionally substituted by C 1 -C 6 alkyl or C 1 -C 6 haloalkyl; preferably R 7 is selected from H or C 1- C 6 alkyl or -NR a R b , preferably methyl or -N(CH 3 )CH 2 CH 2 N(CH 3 ) 2 .
  • R 8 is selected from C 2 -C 6 alkenyl or C 2 -C 6 alkynyl, preferably selected from vinyl or propynyl.
  • Ring A is selected from the group consisting of 6-14 membered arylene groups, preferably 6 membered arylene groups.
  • Ring A is selected from a 5-10 membered heteroarylene, preferably a 5-6 membered heteroarylene, preferably a 5 membered heteroarylene.
  • Ring A is selected from C 3 -C 8 cycloalkylene.
  • Ring A is selected from the group consisting of phenyl, naphthyl, pyrrolyl, furyl, thienyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazole a divalent group of a oxadiazolyl, thiadiazolyl, tetrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, tetrazinyl or cyclohexyl group, preferably selected from the group consisting of Phenyl, pyrazolyl or cyclohexylene.
  • W is selected from a chemical bond.
  • m is one.
  • the present invention provides a compound of the formula (II) or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer, diastereomer thereof Body, prodrug, hydrate or solvate, and mixtures thereof:
  • Ring A is absent or is selected from a 6-14 membered arylene group, a 5-10 membered heteroarylene group, a C 3 -C 8 cycloalkylene group or a 3-10 membered heterocyclylene group;
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 Alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, -OR 13 , -NR 11 R 12 , -C(O)R 13 , -C(O)OR 13 , -C(O )NR 11 R 12 , -NR 11 C(O)R 13 , -NR 11 C(O)OR 13 , -NR 11 C(O)NR 11 R 12 , -OC(O)R 13 , -OC(O )OR 13 , -OC(O)NR 11 R 12 , -S(O) p R 13 , -S(O) p OR 13 , -S(O) p NR 11 R 12 , -
  • R 6 is each independently selected from H or -CH 2 CH 2 -, and when R 6 is -CH 2 CH 2 -, the other end thereof is bonded to X, optionally one of -CH 2 CH 2 - The base is replaced by -O- or -NH-;
  • R 7 is each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3- C 8 cycloalkyl, -OR 13 , or -NR a R b ; wherein R a and R b are each independently selected from H or C 1 -C 6 alkyl optionally substituted by -NR c R d ; R c and R d are each independently selected from H or C 1 -C 6 alkyl;
  • R 8 is selected from an optionally substituted C 2 -C 6 alkenyl group or a C 2 -C 6 alkynyl group;
  • R 11 , R 12 and R 13 are each independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 8 cycloalkyl, 3-10 membered heterocyclyl, 5 -10 membered heteroaryl or 6-14 membered aryl;
  • X is selected from -O-, -NH- or -CH 2 -; Y is selected from -O-, -NH- or -CH 2 -; with the proviso that at least one of X and Y is CH 2 ;
  • n 0, 1, 2, 3 or 4;
  • p 1 or 2.
  • Ring A is selected from the group consisting of 6-14 membered arylene groups, preferably 6 membered arylene groups.
  • Ring A is selected from a 5-10 membered heteroarylene, preferably a 5-6 membered heteroarylene, preferably a 5 membered heteroarylene.
  • Ring A is selected from C 3 -C 8 cycloalkylene.
  • Ring A is selected from the group consisting of phenyl, naphthyl, pyrrolyl, furyl, thienyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazole a divalent group of a oxadiazolyl, thiadiazolyl, tetrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, tetrazinyl or cyclohexyl group, preferably selected from the group consisting of Phenyl, pyrazolyl or cyclohexylene.
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from H, halo, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 Alkoxy or C 1 -C 6 haloalkoxy.
  • R 1 and R 5 are each independently selected from halogen, preferably Cl or F.
  • R 2 and R 4 are each independently selected from C 1 -C 6 alkoxy, preferably methoxy.
  • R 3 is H.
  • each of R 6 is independently selected from H or —CH 2 CH 2 —, and when R 6 is —CH 2 CH 2 —, the other end is attached to X. In a particular embodiment, R 6 is H.
  • R 6 is selected from H.
  • each R 7 is independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or -NR a R b ; wherein R a and R b are each independently selected from H Or C 1 -C 6 alkyl optionally substituted by -NR c R d ; R c and R d are each independently selected from H or C 1 -C 6 alkyl, preferably R 7 is selected from H or C 1 -C 6 alkyl or -NR a R b , preferably methyl or -N(CH 3 )CH 2 CH 2 N(CH 3 ) 2 , preferably H.
  • R 8 is selected from C 2 -C 6 alkenyl or C 2 -C 6 alkynyl, preferably selected from vinyl or propynyl.
  • X is selected from -O -, - NH- or -CH 2 -; Y is selected from -O -, - NH- or -CH 2 -; with the proviso that X and Y is at least one is CH 2 .
  • X is -CH 2 -;
  • Y is selected from -O -, - NH- or -CH 2 -;
  • m is one.
  • the present invention provides a compound of the formula (III) or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer, diastereomer thereof Body, prodrug, hydrate or solvate, and mixtures thereof:
  • Ring A is absent or is selected from a 6-14 membered arylene group, a 5-10 membered heteroarylene group, a C 3 -C 8 cycloalkylene group or a 3-10 membered heterocyclylene group;
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 Alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, -OR 13 , -NR 11 R 12 , -C(O)R 13 , -C(O)OR 13 , -C(O )NR 11 R 12 , -NR 11 C(O)R 13 , -NR 11 C(O)OR 13 , -NR 11 C(O)NR 11 R 12 , -OC(O)R 13 , -OC(O )OR 13 , -OC(O)NR 11 R 12 , -S(O) p R 13 , -S(O) p OR 13 , -S(O) p NR 11 R 12 , -
  • R 7 is each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3- C 8 cycloalkyl, -OR 13 , or -NR a R b ; wherein R a and R b are each independently selected from H or C 1 -C 6 alkyl optionally substituted by -NR c R d ; R c and R d are each independently selected from H or C 1 -C 6 alkyl; or, R a , R b and the N atom to which they are attached together form a 3-10 membered heterocyclic group (eg, 6-membered heterocyclic ring). a group optionally substituted by a C 1 -C 6 alkyl group or a C 1 -C 6 haloalkyl group;
  • R 8 is selected from an optionally substituted C 2 -C 6 alkenyl group or a C 2 -C 6 alkynyl group;
  • R 11 , R 12 and R 13 are each independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 8 cycloalkyl, 3-10 membered heterocyclyl, 5 -10 membered heteroaryl or 6-14 membered aryl;
  • n 0, 1, 2, 3 or 4;
  • p 1 or 2.
  • Ring A is selected from the group consisting of 6-14 membered arylene groups, preferably 6 membered arylene groups.
  • Ring A is selected from a 5-10 membered heteroarylene, preferably a 5-6 membered heteroarylene, preferably a 5 membered heteroarylene.
  • Ring A is selected from C 3 -C 8 cycloalkylene.
  • Ring A is selected from the group consisting of phenyl, naphthyl, pyrrolyl, furyl, thienyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazole a divalent group of a oxadiazolyl, thiadiazolyl, tetrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, tetrazinyl or cyclohexyl group, preferably selected from the group consisting of Phenyl, pyrazolyl or cyclohexylene.
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from H, halo, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 Alkoxy or C 1 -C 6 haloalkoxy.
  • R 1 and R 5 are each independently selected from H or halogen, preferably H, Cl or F.
  • R 1 and R 5 are each independently selected from halogen, preferably Cl or F.
  • R 2 and R 4 are each independently selected from C 1 -C 6 alkoxy, preferably methoxy.
  • R 3 is H.
  • each R 7 is independently selected from H, halo, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or -NR a R b ; wherein R a and R b are each independently selected From C or a C 1 -C 6 alkyl group optionally substituted by -NR c R d ; R c and R d are each independently selected from H or C 1 -C 6 alkyl, or R a , R b and they The attached N atoms together form a 3-10 membered heterocyclyl (eg 6-membered heterocyclyl) which is optionally substituted by a C 1 -C 6 alkyl or a C 1 -C 6 haloalkyl; preferably R 7 is selected from H , halogen, methyl, -N(CH 3 )CH 2 CH 2 N(CH 3 ) 2 , -N(CH 3 ) 2 , 4-methyl-piperazinyl or morph
  • R 8 is selected from C 2 -C 6 alkenyl or C 2 -C 6 alkynyl, preferably selected from vinyl or propynyl, preferably vinyl.
  • m is one.
  • the present invention provides a compound of the formula (IV): or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer, diastereomer thereof a compound, a prodrug, a hydrate or a solvate, and mixtures thereof, which are compounds of the formula (IV):
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 Alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, -OR 13 , -NR 11 R 12 , -C(O)R 13 , -C(O)OR 13 , -C(O )NR 11 R 12 , -NR 11 C(O)R 13 , -NR 11 C(O)OR 13 , -NR 11 C(O)NR 11 R 12 , -OC(O)R 13 , -OC(O )OR 13 , -OC(O)NR 11 R 12 , -S(O) p R 13 , -S(O) p OR 13 , -S(O) p NR 11 R 12 , -
  • R 7 is each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3- C 8 cycloalkyl, -OR 13 , or -NR a R b ; wherein R a and R b are each independently selected from H or C 1 -C 6 alkyl optionally substituted by -NR c R d ; R c and R d are each independently selected from H or C 1 -C 6 alkyl;
  • R 8 is selected from an optionally substituted C 2 -C 6 alkenyl group or a C 2 -C 6 alkynyl group;
  • R 11 , R 12 and R 13 are each independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 8 cycloalkyl, 3-10 membered heterocyclyl, 5 -10 membered heteroaryl or 6-14 membered aryl;
  • X is selected from -O-, -NH- or -CH 2 -; Y is selected from -O-, -NH- or -CH 2 -; with the proviso that at least one of X and Y is CH 2 ;
  • n 0, 1, 2, 3 or 4;
  • p 1 or 2.
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from H, halo, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 Alkoxy or C 1 -C 6 haloalkoxy.
  • R 1 and R 5 are each independently selected from halogen, preferably Cl or F.
  • R 2 and R 4 are each independently selected from C 1 -C 6 alkoxy, preferably methoxy.
  • R 3 is H.
  • each R 7 is independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or -NR a R b ; wherein R a and R b are each independently selected from H Or C 1 -C 6 alkyl optionally substituted by -NR c R d ; R c and R d are each independently selected from H or C 1 -C 6 alkyl, preferably R 7 is selected from H or C 1 -C 6 alkyl or -NR a R b , preferably methyl or -N(CH 3 )CH 2 CH 2 N(CH 3 ) 2 .
  • R 8 is selected from C 2 -C 6 alkenyl or C 2 -C 6 alkynyl, preferably selected from vinyl or propynyl.
  • X is selected from -NH-
  • Y is selected from -CH 2 -.
  • X is selected from -CH 2 -
  • Y is selected from -NH-.
  • m is one.
  • the present invention provides a compound of the formula (V) or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer, diastereomer thereof Body, prodrug, hydrate or solvate, and mixtures thereof:
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 Alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, -OR 13 , -NR 11 R 12 , -C(O)R 13 , -C(O)OR 13 , -C(O )NR 11 R 12 , -NR 11 C(O)R 13 , -NR 11 C(O)OR 13 , -NR 11 C(O)NR 11 R 12 , -OC(O)R 13 , -OC(O )OR 13 , -OC(O)NR 11 R 12 , -S(O) p R 13 , -S(O) p OR 13 , -S(O) p NR 11 R 12 , -
  • R 7 is each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3- C 8 cycloalkyl, -OR 13 , or -NR a R b ; wherein R a and R b are each independently selected from H or C 1 -C 6 alkyl optionally substituted by -NR c R d ; R c and R d are each independently selected from H or C 1 -C 6 alkyl;
  • R 8 is selected from an optionally substituted C 2 -C 6 alkenyl group or a C 2 -C 6 alkynyl group;
  • R 11 , R 12 and R 13 are each independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 8 cycloalkyl, 3-10 membered heterocyclyl, 5 -10 membered heteroaryl or 6-14 membered aryl;
  • n 0, 1 or 2;
  • p 1 or 2.
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from H, halo, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 Alkoxy or C 1 -C 6 haloalkoxy.
  • R 1 and R 5 are each independently selected from H or halogen, preferably H, Cl or F.
  • R 1 and R 5 are each independently selected from halogen, preferably Cl or F.
  • R 2 and R 4 are each independently selected from C 1 -C 6 alkoxy, preferably methoxy.
  • R 3 is H.
  • each R 7 is independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or -NR a R b ; wherein R a and R b are each independently selected from H Or C 1 -C 6 alkyl optionally substituted by -NR c R d ; R c and R d are each independently selected from H or C 1 -C 6 alkyl, preferably R 7 is selected from H or C 1 -C 6 alkyl or -NR a R b , preferably methyl or -N(CH 3 )CH 2 CH 2 N(CH 3 ) 2 , preferably H or methyl.
  • R 8 is selected from C 2 -C 6 alkenyl or C 2 -C 6 alkynyl, preferably selected from vinyl or propynyl, preferably vinyl.
  • m is one.
  • the present invention provides a compound of the formula (VI) or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer, diastereomer thereof a compound, a prodrug, a hydrate or a solvate, and mixtures thereof, which are compounds of the formula (VI):
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 Alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, -OR 13 , -NR 11 R 12 , -C(O)R 13 , -C(O)OR 13 , -C(O )NR 11 R 12 , -NR 11 C(O)R 13 , -NR 11 C(O)OR 13 , -NR 11 C(O)NR 11 R 12 , -OC(O)R 13 , -OC(O )OR 13 , -OC(O)NR 11 R 12 , -S(O) p R 13 , -S(O) p OR 13 , -S(O) p NR 11 R 12 , -
  • R 7 is each independently selected from H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3- C 8 cycloalkyl, -OR 13 , or -NR a R b ; wherein R a and R b are each independently selected from H or C 1 -C 6 alkyl optionally substituted by -NR c R d ; R c and R d are each independently selected from H or C 1 -C 6 alkyl; or, R a , R b and the N atom to which they are attached together form a 3-10 membered heterocyclic group (eg, 6-membered heterocyclic ring). a group optionally substituted by a C 1 -C 6 alkyl group or a C 1 -C 6 haloalkyl group;
  • R 8 is selected from an optionally substituted C 2 -C 6 alkenyl group or a C 2 -C 6 alkynyl group;
  • R 11 , R 12 and R 13 are each independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 8 cycloalkyl, 3-10 membered heterocyclyl, 5 -10 membered heteroaryl or 6-14 membered aryl;
  • n 0, 1, 2, 3 or 4;
  • p 1 or 2.
  • R 1 , R 2 , R 3 , R 4 and R 5 are each independently selected from H, halo, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 Alkoxy or C 1 -C 6 haloalkoxy.
  • R 1 and R 5 are each independently selected from H or halogen, preferably H, Cl or F.
  • R 1 and R 5 are each independently selected from halogen, preferably Cl or F.
  • R 2 and R 4 are each independently selected from C 1 -C 6 alkoxy, preferably methoxy.
  • R 3 is H.
  • each R 7 is independently selected from H, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or -NR a R b ; wherein R a and R b are each independently selected from H Or a C 1 -C 6 alkyl group optionally substituted by -NR c R d ; R c and R d are each independently selected from H or C 1 -C 6 alkyl, or R a , R b and they are attached The N atoms together form a 3-10 membered heterocyclyl (eg 6-membered heterocyclyl) which is optionally substituted by a C 1 -C 6 alkyl or a C 1 -C 6 haloalkyl; preferably R 7 is selected from H, halo , methyl, -N(CH 3 )CH 2 CH 2 N(CH 3 ) 2 , -N(CH 3 ) 2 , 4-methyl-piperazinyl or morpholinyl
  • R 8 is selected from C 2 -C 6 alkenyl or C 2 -C 6 alkynyl, preferably selected from vinyl or propynyl, preferably vinyl.
  • m is one.
  • the present invention provides a compound of the formula (VII): or a pharmaceutically acceptable salt, tautomer, racemate, enantiomer, diastereomer thereof a compound, a prodrug, a hydrate or a solvate, and mixtures thereof, which are compounds of the formula (VI):
  • R 1 and R 5 are each independently halogen, preferably selected from Cl or F;
  • R 2 and R 4 are each independently C 1 -C 6 alkoxy, preferably -OCH 3 ;
  • R 3 is H
  • R 7 are each independently selected from halogen, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl or -NR a R b ; wherein R a and R b are each independently selected from H or optionally by -NR c R d substituted C 1 -C 6 alkyl; R c and R d are each independently selected from H or C 1 -C 6 alkyl; or, R a , R b together with the N atom to which they are attached form 3 a 10-membered heterocyclic group (for example, a 6-membered heterocyclic group) which is optionally substituted by a C 1 -C 6 alkyl group or a C 1 -C 6 haloalkyl group; preferably, R 7 is selected from the group consisting of halogen, methyl, -N (CH 3 ) 2 or morpholinyl;
  • R 8 is an optionally substituted C 2 -C 6 alkenyl group; preferably, R 8 is a vinyl group;
  • n 0, 1, 2, 3 or 4; preferably, m is 0, 1 or 2; preferably, m is 1.
  • Typical compounds of the invention include, but are not limited to:
  • R 1 is selected from the group consisting of H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl , C 3 -C 8 cycloalkyl, -OR 13 , -NR 11 R 12 , -C(O)R 13 , -C(O)OR 13 , -C(O)NR 11 R 12 , -NR 11 C (O)R 13 , -NR 11 C(O)OR 13 , -NR 11 C(O)NR 11 R 12 , -OC(O)R 13 , -OC(O)OR 13 , -OC(O)NR 11 R 12 , -S(O) p R 13 , -S(O) p OR 13 , -S(O) p NR 11 R 12 , -OS(O) p R 13 or
  • R 2 is selected from the group consisting of H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl , C 3 -C 8 cycloalkyl, -OR 13 , -NR 11 R 12 , -C(O)R 13 , -C(O)OR 13 , -C(O)NR 11 R 12 , -NR 11 C (O) R 13 , -NR 11 C(O)OR 13 , -NR 11 C(O)NR 11 R 12 , -OC(O)R 13 , -OC(O)OR 13 , -OC(O)NR 11 R 12 , -S(O) p R 13 , -S(O) p OR 13 , -S(O) p NR 11 R 12 , -OS(O) p R 13 or
  • R 3 is selected from the group consisting of H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl , C 3 -C 8 cycloalkyl, -OR 13 , -NR 11 R 12 , -C(O)R 13 , -C(O)OR 13 , -C(O)NR 11 R 12 , -NR 11 C (O)R 13 , -NR 11 C(O)OR 13 , -NR 11 C(O)NR 11 R 12 , -OC(O)R 13 , -OC(O)OR 13 , -OC(O)NR 11 R 12 , -S(O) p R 13 , -S(O) p OR 13 , -S(O) p NR 11 R 12 , -OS(O) p R 13 or
  • R 4 is selected from the group consisting of H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl , C 3 -C 8 cycloalkyl, -OR 13 , -NR 11 R 12 , -C(O)R 13 , -C(O)OR 13 , -C(O)NR 11 R 12 , -NR 11 C (O)R 13 , -NR 11 C(O)OR 13 , -NR 11 C(O)NR 11 R 12 , -OC(O)R 13 , -OC(O)OR 13 , -OC(O)NR 11 R 12 , -S(O) p R 13 , -S(O) p OR 13 , -S(O) p NR 11 R 12 , -OS(O) p R 13 or
  • R 5 is selected from the group consisting of H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl , C 3 -C 8 cycloalkyl, -OR 13 , -NR 11 R 12 , -C(O)R 13 , -C(O)OR 13 , -C(O)NR 11 R 12 , -NR 11 C (O)R 13 , -NR 11 C(O)OR 13 , -NR 11 C(O)NR 11 R 12 , -OC(O)R 13 , -OC(O)OR 13 , -OC(O)NR 11 R 12 , -S(O) p R 13 , -S(O) p OR 13 , -S(O) p NR 11 R 12 , -OS(O) p R 13 or
  • R 6 is selected from H or -CH 2 CH 2 -.
  • R 6 is -CH 2 CH 2 -, the other end thereof is bonded to X, and optionally one methylene group in -CH 2 CH 2 - is replaced by -O- or -NH-.
  • R 7 is selected from the group consisting of H, halogen, cyano, NO 2 , C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl , C 3 -C 8 cycloalkyl, -OR 13 , or -NR a R b ; wherein R a and R b are each independently selected from H or C 1 -C 6 optionally substituted by -NR c R d Alkyl; R c and R d are each independently selected from H or C 1 -C 6 alkyl; R 13 is as defined in the specification.
  • R 7 is selected from H or C 1 -C 6 alkyl or -NR a R b .
  • R 7 is methyl or -N(CH 3 )CH 2 CH 2 N(CH 3 ) 2 .
  • R 8 is selected from an optionally substituted C 2 -C 6 alkenyl group or a C 2 -C 6 alkynyl group, wherein the optional substituent is as defined in the specification.
  • R 8 is a vinyl group, an ethynyl group or a propynyl group.
  • R 11 is selected from the group consisting of H, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 8 cycloalkyl, 3-10 membered heterocyclic, 5-10 membered hetero Aryl or 6-14 membered aryl.
  • R 12 is selected from the group consisting of H, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 8 cycloalkyl, 3-10 membered heterocyclic, 5-10 membered hetero Aryl or 6-14 membered aryl.
  • R 13 is selected from the group consisting of H, C 1 -C 6 alkyl, C 1 -C 6 heteroalkyl, C 3 -C 8 cycloalkyl, 3-10 membered heterocyclic, 5-10 membered hetero Aryl or 6-14 membered aryl.
  • Ring A is selected from a 6-14 membered aryl group, a 5-10 membered heteroaryl group, a C 3 -C 8 cycloalkyl group or a 3-10 membered heterocyclylene group.
  • Ring A is a 6-10 membered aryl group, a 5-6 membered heteroaryl group.
  • Ring A is a 6-membered aryl or a 5-membered heteroaryl.
  • ring A is selected from the group consisting of phenyl, naphthyl, pyrrolyl, furyl, thienyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, cacao A oxazolyl, thiadiazolyl, tetrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, tetrazinyl or cyclohexyl group, preferably selected from phenyl, pyrazolyl or cyclohexyl .
  • X is selected from -O-, -NH- or -CH 2 -, and Y is selected from -O-, -NH- or -CH 2 -; with the proviso that at least one of X and Y is CH 2 .
  • X is selected from -O-, Y is selected from -CH 2 -.
  • X is selected from -NH-, Y is selected from -CH 2 -.
  • X is selected from -CH 2 - and Y is selected from -CH 2 -.
  • Y is selected from -O-, X is selected from -CH 2 -.
  • Y is selected from -NH-, X is selected from -CH 2 -.
  • X cannot be -O- at this time.
  • W is selected from a chemical bond, -NH- or -CH 2 -.
  • compositions, formulations and kits are provided.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention (also referred to as "active ingredient") and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition comprises an effective amount of the active component.
  • the pharmaceutical composition comprises a therapeutically effective amount of the active component.
  • the pharmaceutical composition comprises a prophylactically effective amount of the active component.
  • a pharmaceutically acceptable excipient for use in the present invention refers to a non-toxic carrier, adjuvant or vehicle which does not destroy the pharmacological activity of the compound formulated together.
  • Pharmaceutically acceptable carriers, adjuvants, or vehicles that can be used in the compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (eg, human serum white) Protein), buffer substances (such as phosphate), glycine, sorbic acid, potassium sorbate, a mixture of partial glycerides of saturated plant fatty acids, water, salt or electrolyte (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate , sodium chloride, zinc salt, silica gel, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based materials, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylate, wax, polyethylene-polyoxypropylene - Block
  • kits e.g., pharmaceutical packs.
  • Kits provided may include a compound of the invention, other therapeutic agents, and first and second containers (eg, vials, ampoules, bottles, syringes, and/or dispersible packages or other materials containing the compounds of the invention, other therapeutic agents) Suitable container).
  • first and second containers eg, vials, ampoules, bottles, syringes, and/or dispersible packages or other materials containing the compounds of the invention, other therapeutic agents
  • kits can also optionally include a third container containing a pharmaceutically acceptable excipient for diluting or suspending a compound of the invention and/or other therapeutic agent.
  • a compound of the invention provided in a first container and a second container is combined with other therapeutic agents to form a unit dosage form.
  • the pharmaceutical composition provided by the present invention can be administered by a variety of routes including, but not limited to, oral administration, parenteral administration, inhalation administration, topical administration, rectal administration, nasal administration, oral administration, vaginal administration.
  • parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intra-articular administration, intra-arterial administration, intrasynovial administration, intrasternal administration. , intracerebroventricular administration, intralesional administration, and intracranial injection or infusion techniques.
  • an effective amount of a compound provided herein is administered.
  • the amount of compound actually administered can be determined by the physician. .
  • the compound provided herein is administered to a subject at risk of developing the condition, typically based on a physician's recommendation and administered under the supervision of a physician, at the dosage level as described above.
  • Subjects at risk of developing a particular condition typically include subjects with a family history of the condition, or those subjects that are particularly susceptible to developing the condition by genetic testing or screening.
  • long-term administration can also be administered chronically.
  • Long-term administration refers to administration of a compound or a pharmaceutical composition thereof for a long period of time, for example, 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or can be continuously administered indefinitely, For example, the rest of the subject.
  • chronic administration is intended to provide a constant level of the compound in the blood over a prolonged period of time, for example, within a therapeutic window.
  • a pharmaceutical composition of the present invention can be further delivered using various methods of administration.
  • a pharmaceutical composition can be administered by bolus injection, for example, to increase the concentration of the compound in the blood to an effective level.
  • the bolus dose depends on the target systemic level of the active ingredient through the body, for example, an intramuscular or subcutaneous bolus dose that causes a slow release of the active ingredient, while a bolus that is delivered directly to the vein (eg, via IV IV drip) ) can be delivered more quickly, so that the concentration of the active ingredient in the blood is rapidly increased to an effective level.
  • the pharmaceutical composition can be administered in a continuous infusion form, for example, by IV intravenous drip to provide a steady state concentration of the active ingredient in the subject's body.
  • a bolus dose of the pharmaceutical composition can be administered first, followed by continued infusion.
  • Oral compositions can be in the form of a bulk liquid solution or suspension or bulk powder. More generally, however, the compositions are provided in unit dosage form for ease of precise dosage administration.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active ingredient suitable to produce the desired therapeutic effect with a suitable pharmaceutical excipient.
  • Typical unit dosage forms include prefilled, pre-measured ampoules or syringes of the liquid compositions, or pills, tablets, capsules and the like in the case of solid compositions.
  • the compound will generally be a minor component (about 0.1 to about 50% by weight, or preferably about 1 to about 40% by weight), with the remainder being useful for forming the desired form of administration.
  • a carrier or excipient and a processing aid is included in a carrier or excipient and a processing aid.
  • a representative regimen is one to five oral doses per day, especially two to four oral doses, typically three oral doses.
  • each dose provides from about 0.01 to about 20 mg/kg of a compound of the invention, each preferably providing from about 0.1 to about 10 mg/kg, especially from about 1 to about 5 mg/kg.
  • a transdermal dose is generally selected in an amount of from about 0.01 to about 20% by weight, preferably from about 0.1 to about 20% by weight, preferably about 0.1. To about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
  • the injection dose level ranges from about 1 mg/kg/hr to at least 10 mg/kg/hr from about 1 to about 120 hours, especially 24 to 96 hours.
  • a preload bolus of about 0.1 mg/kg to about 10 mg/kg or more can also be administered.
  • the maximum total dose cannot exceed about 2 g/day.
  • Liquid forms suitable for oral administration may include suitable aqueous or nonaqueous vehicles as well as buffers, suspending and dispersing agents, coloring agents, flavoring agents, and the like.
  • the solid form may include, for example, any of the following components, or a compound having similar properties: a binder, for example, microcrystalline cellulose, tragacanth or gelatin; an excipient such as starch or lactose, a disintegrant, For example, alginic acid, Primogel or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silica; a sweetener such as sucrose or saccharin; or a flavoring agent such as mint, water Methyl salicylate or orange flavoring.
  • a binder for example, microcrystalline cellulose, tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrant, For example, alginic acid, Primogel or corn star
  • Injectable compositions are typically based on injectable sterile saline or phosphate buffered saline, or other injectable excipients known in the art.
  • the active compound will typically be a minor component, often from about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
  • transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient.
  • the active component When formulated as an ointment, the active component is typically combined with a paraffin or water miscible ointment base.
  • the active ingredient can be formulated as a cream with, for example, an oil-in-water cream base.
  • Such transdermal formulations are well known in the art and generally include other ingredients for enhancing stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and components are included within the scope of the invention.
  • transdermal administration can be accomplished using a reservoir or a porous membrane type, or a patch of a plurality of solid matrices.
  • compositions for oral administration, injection or topical administration are merely representative.
  • Other materials and processing techniques, etc. are set forth in Remington's Pharmaceutical Sciences, 17th edition, 1985, Mack Publishing Company, Easton, Pennsylvania, part 8 of which is incorporated herein by reference.
  • the compounds of the invention may also be administered in sustained release form or from a sustained release delivery system.
  • sustained release materials can be found in Remington's Pharmaceutical Sciences.
  • the invention further relates to pharmaceutically acceptable formulations of the compounds of the invention.
  • the formulation comprises water.
  • the formulation comprises a cyclodextrin derivative.
  • the most common cyclodextrins are alpha-, beta- and gamma-cyclodextrins consisting of 6, 7 and 8 alpha-1,4-linked glucose units, respectively, optionally including one on the attached sugar moiety. Or a plurality of substituents including, but not limited to, methylated, hydroxyalkylated, acylated, and sulfoalkyl ether substituted.
  • the cyclodextrin is a sulfoalkyl ether beta-cyclodextrin, eg, sulfobutylether beta-cyclodextrin, also known as Captisol. See, for example, U.S. 5,376,645.
  • the formulation comprises hexapropyl- ⁇ -cyclodextrin (eg, 10-50% in water).
  • the compounds of the invention or compositions thereof may be administered in combination with other therapeutic agents to treat the disease.
  • therapeutic agents include, but are not limited to, Adriamycin, dexamethasone, vincristine, cyclophosphamide, fluorouracil, topotecan ( Topotecan), taxol, interferon, platinum derivatives, taxanes (eg, paclitaxel), vinca alkaloids (eg, vinblastine), anthracycline ( Anthracycline) (eg, doxorubicin), epipodophyllotoxin (eg, etoposide), cisplatin, mTOR inhibitor (eg, rapamycin), Methotrexate, actinomycin D, dolastatin 10, colchicine, emetine, trimetrexate, chlorobenzene Metoprine, cyclosporine, daunorubicin, teniposide, amphotericin, alkylating agent (eg chlorambucil
  • a compound of the invention or a composition thereof can be administered in combination with any one or more anti-proliferative or chemotherapeutic agents selected from the group consisting of: abarelix, aldileukin (aldesleukin), alemtuzumab, alitretinoin, allopurinol, altretamine, amifostine, anastrozole, arsenic trioxide, day Asparaginase, azacitidine, BCG Live, bevacuzimab, fluorouracil, bexarotene, bleomycin, bortezomib, white Busulfan, calbuterone, capecitabine, camptothecin, carboplatin, carmustine, celecoxib, cetuximab Antibiotic (cetuximab), chlorambucil, cladribine, clofarabine, cyclophosphamide, cytarabine, actinomycin D, darbepo
  • therapeutic agents to which the compounds of the invention may also be combined include, but are not limited to, therapeutic agents for Alzheimer's Disease, such as donepezil hydrochloride and rivastigmine. ; therapeutic agents for Parkinson's Disease, such as L-DOPA/carbidopa, entacapone, ropinrole, pramipexole ( Pramipexole), bromocriptine, pergolide, trihexephendyl, and amantadine; a therapeutic agent for multiple sclerosis (MS), Such as beta interferon, glatiramer acetate and mitoxantrone; therapeutic agents for asthma, such as albuterol and montelukast; therapeutic agents for schizophrenia, such as Zyprexa, risperdal, seroquel, and haloperidol; anti-inflammatory agents such as corticosteroids, TNF blockers, IL-1RA, azathioprine , cyclophosphamide and sulfasala
  • those other active agents can be administered separately from the compositions containing the compounds of the invention as part of a multiple dosing regimen.
  • those active agents may be part of a single dosage form, mixed with a compound of the invention in a single composition. If administered as part of a multiple dosing regimen, the two active agents can be provided simultaneously, sequentially, or at intervals from one another (usually within 5 hours of each other).
  • FGFR-4 regulates proliferation, survival, and alpha-fetoprotein secretion during progression of hepatocellular carcinoma (HCC); FGFR-4 inhibitors are therefore promising potential therapeutic agents for this unmet medical need (Ho et al., Journal Of Hepatology, 2009, 50: 118-27). HCC afflicts more than 550,000 people worldwide and is one of the worst 1-year survival rates of any cancer type.
  • FGF19 a member of the fibroblast growth factor (FGF) family, which is composed of hormones
  • FGF19 a member of the fibroblast growth factor (FGF) family, which is composed of hormones
  • Increased hepatocyte proliferation and liver tumor formation have been observed in FGF19 gene-transferred mice.
  • FGF19 activates FGFR-4, its major receptor in the liver, and FGFR-4 activation is thought to be a mechanism by which FGF19 can increase hepatocyte proliferation and induce hepatocellular carcinoma formation (Wu et al., J Biol Chem (2010) 285 ( 8): 5165-5170).
  • FGF19 has also been recognized by others as a driver gene in HCC (Sawey et al., Cancer Cell (2011) 19:347-358).
  • the compounds disclosed herein which are potential and selective inhibitors of FGFR-4, are believed to be useful in the treatment of HCC and other liver cancers.
  • FGFR-4 activated fibroblast growth factor receptor 4
  • MDA-MB-453 human breast cancer cell line MDA-MB-453.
  • FGFR-4 may be a driver of tumor growth in breast cancer (Roid et al, Oncogene (2010) 29(10): 1543-1552).
  • the compounds disclosed herein, which are potent and selective inhibitors of FGFR-4 are believed to be useful in the treatment of FGFR-4 regulated breast cancer.
  • FGFR-4 activation/overexpression results in overexpression of FGFR-4.
  • FGFR-4 overexpression by the mechanism is associated with rhabdomyosarcoma (RMS) (Cao et al, Cancer Res (2010) 70(16): 6497-6508).
  • RMS rhabdomyosarcoma
  • Mutations in FGFR-4 itself result in protein overactivation; this mechanism has been associated with the RMS subpopulation (Taylor et al, J Clin Invest (2009) 119: 3395-3407).
  • RMS rhabdomyosarcoma
  • the compounds disclosed herein, which are potent and selective inhibitors of FGFR-4 are believed to be useful in the treatment of FGFR-4 regulated RMS and other sarcomas.
  • FGFR-4 upstream gene Other diseases are associated with changes in the FGFR-4 upstream gene or with mutations in FGFR-4 itself.
  • mutations in the kinase domain of FGFR-4 result in overactivation, which is associated with lung adenocarcinoma (Ding et al, Nature (2008) 455 (7216): 1069-1075).
  • Amplification of FGFR-4 is associated with conditions such as renal cell carcinoma (TCGA interim data).
  • silencing FGFR4 and inhibiting ligand-receptor binding significantly slowed ovarian tumor growth, indicating that inhibitors of FGFR4 can be used to treat ovarian cancer (Zaid et al, Clin. Cancer Res. (2013) 809).
  • FGF19 Increased pathogenicity of bile acid levels is associated with changes in FGF19 levels (Vergnes et al, Cell Metabolism (2013) 17, 916-28). Thus, a decrease in the level of FGF19 may be beneficial in promoting the synthesis of bile acids and thus in the treatment of hyperlipidemia.
  • the compounds of the invention may be used to treat a variety of FGFR-related diseases, including but not limited to: gastric cancer, thyroid cancer, prostate cancer, breast cancer, sarcoma (eg, rhabdomyosarcoma), skin cancer (eg, melanoma) ), liver cancer (such as hepatocellular carcinoma and cholangiocarcinoma), pancreatic cancer (such as pancreatic intraepithelial neoplasia and pancreatic ductal adenocarcinoma), lung cancer (such as non-small cell lung cancer and lung adenocarcinoma), kidney cancer (such as kidney cells) Cancer), colorectal cancer and ovarian cancer.
  • gastric cancer thyroid cancer
  • prostate cancer breast cancer
  • sarcoma eg, rhabdomyosarcoma
  • skin cancer eg, melanoma
  • liver cancer such as hepatocellular carcinoma and cholangiocarcinoma
  • pancreatic cancer such as pancre
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) or mass spectrometry (MS). NMR was measured using a Bruker AVANCE-400 or Varian Oxford-300 nuclear magnetic apparatus, and the solvent was deuterated dimethyl sulfoxide (DMSO-d6), deuterated chloroform (CDC1 3 ), deuterated methanol (CD 3 OD). The internal standard is tetramethylsilane (TMS) and the chemical shift is given in units of 10 -6 (ppm).
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • the MS was measured using an Agilent SQD (ESI) mass spectrometer (manufacturer: Agilent, model: 6110) or a Shimadzu SQD (ESI) mass spectrometer (manufacturer: Shimadzu, model: 2020).
  • ESI Agilent SQD
  • ESI Shimadzu SQD
  • the HPLC was measured using an Agilent 1200 DAD high pressure liquid chromatograph (Sunfire C18, 150 x 4.6 mm, 5 ⁇ m, column) and a Waters 2695-2996 high pressure liquid chromatograph (Gimini C18 150 x 4.6 mm, 5 ⁇ m column).
  • the thin-layer chromatography silica gel plate uses Qingdao Ocean GF254 silica gel plate, and the silica gel plate used for thin-layer chromatography (TLC) has a specification of 0.15 mm to 0.2 mm.
  • the specification for separation and purification of thin layer chromatography is 0.4 mm to 0.5 mm. silicone board.
  • the known starting materials of the present invention may be synthesized by or according to methods known in the art, or may be purchased from ABCR GmbH & Co. KG, Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc, Beijing Coupling Companies such as chemicals.
  • An argon atmosphere or a nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon having a volume of about 1 L.
  • the hydrogen atmosphere means that the reaction flask is connected to a hydrogen balloon of about 1 L volume.
  • the pressurized hydrogenation reaction was carried out using a GCD-500G high purity hydrogen generator and a BLT-2000 medium pressure hydrogenator from Beijing Jiawei Kechuang Technology Co., Ltd.
  • the hydrogenation reaction is usually evacuated, charged with hydrogen, and operated three times.
  • the microwave reaction used a CEM Discover-SP type microwave reactor.
  • the temperature of the reaction is room temperature, and the temperature range is from 20 ° C to 30 ° C.
  • reaction progress in the examples was monitored by thin layer chromatography (TLC), and the system used for the reaction was A: dichloromethane and methanol system; B: petroleum ether and ethyl acetate system, the volume ratio of the solvent was based on The polarity of the compound is adjusted to adjust.
  • TLC thin layer chromatography
  • the system for purifying the compound using the column chromatography eluent and the system for developing the thin layer chromatography include A: dichloromethane and methanol systems; B: petroleum ether and ethyl acetate system, the volume ratio of the solvent according to the compound Adjustments can be made with different polarities and can also be adjusted by adding a small amount of an acidic or alkaline reagent such as triethylamine.
  • Lithium diisopropylamide was slowly added dropwise to the compound 3-(3,5-dimethoxyphenyl)cyclohexane-1-one 10a (9.5 g, 40.6 mmol, 1 eq) in anhydrous tetrahydrofuran at -78 °C. In the solution, stir at -78 ° C for half an hour. Diethyl oxalate (8.9 g, 60.9 mmol, 1.5 eq) was then added dropwise to the reaction mixture, which was warmed to room temperature and stirred for 2 hr. Post-treatment: The reaction was quenched by the addition of 10 mL of aqueous ammonium chloride solution, and the organic layer was evaporated to dryness.
  • EtOAc 3,5-Dimethoxyphenyl)-4,5,6,7-tetrahydro-1H-indazole-3-carboxylic acid ethyl ester 10d (700 mg, yellow liquid), yield: 72%.
  • the solvent is spin-dried, separated by preparative liquid phase, and lyophilized to obtain the target product N-(2-((6-(2,6-dichloro-3,5-dimethoxyphenyl)-4,5, 6,7-Tetrahydro-1H-indazol-3-yl)amino)-3-methylphenyl)acrylamide 10 (5.8 mg, white solid). Yield: 14%.
  • Example 15a was synthesized with reference to the procedure of Example 093d.
  • the first four steps were synthesized with reference to the examples 010d and 027b steps.
  • 6-(2,6-Dichloro-3,5-dimethoxyphenyl)-4,5,6,7-tetrahydro-1H-indazole-3-carboxylic acid 083e (3.92 g, 10.57 mmol, 1 eq) was dissolved in 40 mL of dichloromethane, and oxalyl chloride (2.68 g, 21.13 mmol, 2 eq) was added at 0 ° C, catalyzed by a drop of anhydrous N,N-dimethylformamide, and the reaction was stirred at room temperature for 2 hours, then Ammonia gas was passed through the system for 30 minutes.
  • 6-(2,6-Dichloro-3,5-dimethoxyphenyl)-4,5,6,7-tetrahydro-1H-indazole-3-carboxamide 083f (1.00 g, 2.70 mmol, 1 eq) was dissolved in 60 mL of THF.
  • EtOAc EtOAc
  • EtOAc EtOAc
  • EtOAc Post-treatment: temperature control 0 ° C with water quenching and dilute the reaction solution, adjust the pH value to 3 with hydrochloric acid solution, extract the organic impurities with ethyl acetate, and then adjust the pH of the aqueous phase to 9 with sodium hydroxide solution, Extracted with ethyl acetate and decomposed the organic phase under reduced pressure to give (6-(2,6-dichloro-3,5-dimethoxyphenyl)-4,5,6,7-tetrahydro-1H-carbazole. -3-yl)methylamine 083 g (0.70 g, yellow solid), yield: 73%.
  • n-Butyllithium (3 mL, 7.2 mmol) was mixed with anhydrous tetrahydrofuran (20 mL) at 780 ° C under nitrogen, and acetonitrile (296 mg, 7.2 mmol) was slowly added dropwise to the reaction system, and reacted at -78 ° C for 1 hour.
  • 3-(2,6-Dichloro-3,5-dimethoxyphenyl)acrylic acid ethyl ester (1.1 g, 3.6 mmol) was dissolved in anhydrous tetrahydrofuran (5 mL) and slowly added dropwise to the reaction system. Slowly warm to room temperature and stir for 3 hours.
  • Example 093a was synthesized with reference to example 098d.
  • P1 and P2 were obtained by splitting through a chiral column. Chiral separation conditions: equipment SFC, column: chiralpak-AD mobile phase: CO2-IPA (DEA). The short-lived named P1 is reserved on the SFC machine, and the long-lasting named P2 is retained. One of P1 and P2 is the R-isomer and the other is the S-isomer.
  • Example 96 was synthesized by reference to the procedure of Example 91, but in the eighth step 2-(5-(2,6-dichloro-3,5-dimethoxyphenethyl)-1H-pyrazole-3- Aniline (100 mg, 0.25 mmol), 2-butynoic acid (23 mg, 0.28 mmol) and dichloromethane (5 mL) were mixed, and dicyclohexylcarbodiimide (57 mg, 0.28 mmol) was slowly added dropwise with stirring at 0 °C. 4-Dimethylaminopyridine (6 mg, 0.025 mmol) was stirred for 30 minutes.
  • Cyclohexenone (4 g, 41 mmol), 2-5 dimethoxybenzeneboronic acid (8.5 g, 47 mmol), cesium carbonate (13 g, 41 mmol), palladium acetate (1 g, 4 mmol), triphenylphosphine (2.2 g) , 8 mmol), chloroform (0.5 mL) and toluene (100 mL) were mixed, warmed to 85 ° C under nitrogen atmosphere, and stirred for 24 hours.
  • 6-(2,6-Dichloro-3,5-dimethoxyphenyl)-3-(2-nitrophenyl)-4,5,6,7-tetrahydro-1H-indazole 098e 400 mg, 1 mmol was placed in 10 mL of DMF, zinc powder (310 mg, 5 mmol, 5 eq), ammonium chloride (530 mg, 10 mmol, 10 eq), 1 ml of water, and reacted at 50 ° C for 0.8 h.
  • n-butyllithium (69 mL, 164 mmol) was mixed with anhydrous tetrahydrofuran (200 mL) at 780 ° C, and acetonitrile (6.7 g, 164 mmol) was slowly added dropwise to the reaction system, and reacted at -78 ° C for 1 hour.
  • Methyl 2-nitrobenzoate (15 g, 82 mmol) was slowly added dropwise to anhydrous tetrahydrofuran (100 mL), and the mixture was slowly warmed to room temperature and stirred for 3 hours.
  • N-(2,6-Dichloro-3,5-dimethoxybenzyl)-5-(2-nitrophenyl)-1H-pyrazol-3-amine 200 mg, 0.47 mmol
  • zinc powder (30 mg, 4.7 mmol
  • ammonium chloride (249 mg, 4.7 mmol)
  • ethanol 5 mL
  • Work-up cooling to room temperature, filtration, desolvation under reduced pressure to give the desired product 5-(2-aminophenyl)-N-(2,6-dichloro-3,5-dimethoxybenzyl)-1H- Pyrazole-3-amine 100e (150 mg, white solid), yield 80%.
  • Reference example 101d synthesizes 102a
  • 3-(3,5-Dimethoxyphenyl)cyclohexanone 103b (1.2 g, 5 mmol) was placed in 100 mL of THF and then evaporated. LDA (6 mmol) was added at -78 ° C, and the temperature was raised to -40 ° C for 2 h.
  • 1-hydro-4-nitro-3-formylchloropyrazole (900 mg, 5 mmol) was added and the mixture was warmed to room temperature for 3 h. After work-up: quenched with aq. aq. EtOAc EtOAc (EtOAc:EtOAc.
  • Oxyphenyl)-2-(4-nitro-1H-pyrazole-3-carbonyl)cyclohexanone 103c (630 mg, 2 mmol, yellow solid), yield: 40%.
  • 6-(3,5-Dimethoxyphenyl)-3-(4-nitro-1H-pyrazol-3-yl)-4,5,6,7-tetrahydro-1H-indazole 103d ( 295 mg, 0.8 mmol) was mixed with 20 ml of acetic acid, and NCS (220 mg, 1.8 mmol) was added and reacted at 80 ° C for 2 h.
  • Example 107 was synthesized with reference to Example 103, but in the second step, 1-hydro-4-nitro-3-formylchloropyrazole was replaced with 1-methyl-4-nitro-3-formylchloropyrazole.
  • Example 109 was synthesized by reference to Example 093, but in the fourth step, 2-butynoic acid was used instead of acryloyl chloride: 098f (200 mg, 0.5 mmol) and 20 mL of dichloromethane were combined, and N,N-diisopropylethylamine (190 mg) was added. , 1.5 mmol), HATU (260 mg, 0.7 mmol), then 2-butynoic acid (40 mg, 0.5 mmol) was slowly added, and the mixture was allowed to react to room temperature at 0 ° C for 1 h.
  • 2-butynoic acid was used instead of acryloyl chloride: 098f (200 mg, 0.5 mmol) and 20 mL of dichloromethane were combined, and N,N-diisopropylethylamine (190 mg) was added. , 1.5 mmol), HATU (260 mg, 0.7 mmol), then 2-butynoic acid (40 mg, 0.5 m
  • Example 111 was synthesized by reference to the procedure of Example 100, but in the fifth step 5-(2-aminophenyl)-N-(2,6-dichloro-3,5-dimethoxybenzyl)-1H- Pyrazol-3-amine (150 mg, 0.38 mmol), N,N-diisopropylethylamine (147 mg, 1.14 mmol), methylene chloride (5 mL). 0.40 mmol), stirring was continued for 30 minutes. Post-treatment: 10 mL of water was added to the mixture, and the mixture was extracted with dichloromethane (10 mL ⁇ 3). The organic phase was combined and then evaporated under reduced pressure.
  • Example 112 was synthesized by referring to the procedure of Example 101, but in the sixth step, the butyric acid was replaced with acryloyl chloride to synthesize N-(2-(3-((2,6-dichloro-3,5-dimethoxyphenoxy)). Methyl)-1H-pyrazol-5-yl)phenyl)acrylamide 112.
  • Reference example 102 synthesizes 113a.
  • Synthesis 117a was carried out in accordance with Example 091e.
  • N1-(3-(2,6-Dichloro-3,5-dimethoxyphenethyl)-1H-pyrazol-5-yl)benzene-1,2-diamine 100 mg, 0.24 mmol
  • N,N-Diisopropylethylamine 92 mg, 0.72 mmol
  • acryloyl chloride 26 mg, 0.29 mmol
  • Post-treatment 10 mL of water was added to the mixture, and the mixture was extracted with dichloromethane (10 mL ⁇ 3). The organic phase was combined and then evaporated under reduced pressure.
  • Example 120 was synthesized by reference to the procedure of Example 117, but in the third step, N1-(3-(2,6-dichloro-3,5-dimethoxyphenethyl)-1H-pyrazole-5- Phenyl-1,2-diamine (100 mg, 0.24 mmol), 2-(7-oxobenzotriazole)-N,N,N',N'-tetramethyluron hexafluorophosphate (137 mg , 0.36 mmol), N,N-diisopropylethylamine (87 mg, 0.72 mmol) and dichloromethane (5 mL), and slowly stirred at 0 ° C with 2-butynoic acid (29 mg, 1.2 mmol), stirring was continued.
  • 2-butynoic acid 29 mg, 1.2 mmol
  • N-(5-((1R,2R)-2-Aminocyclohexyl)-1H-pyrazol-3-yl)-2,2,2-trifluoroacetamide 110 mg, 0.39 mmol
  • N, N Diisopropylethylamine 150 mg, 1.17 mmol
  • tetrahydrofuran 3 mL
  • di-tert-butyl dicarbonate 102 mg, 0.47 mmol
  • Lithium diisopropylamide (1.3 mL, 2.5 mmol, 1.1 eq) was slowly added dropwise at -78 °C to the compound 3-(3,5-dimethoxyphenyl)cyclohexane-1-one 10a (540 mg, A solution of 2.3 mmol, 1 eq) in dry tetrahydrofuran (10 mL) was stirred at -30 ° C to -40 ° C for two hours. Then, 4-fluoro-2-nitrobenzoyl chloride 133b (515 mg, 2.5 mmol, 1.1 eq) was added dropwise to the reaction mixture, which was warmed to room temperature and stirred for 2 hr.
  • P1 and P2 were obtained by splitting through a chiral column. Chiral separation conditions: equipment SFC, column: chiralpak-AD mobile phase: CO2-IPA (DEA). The short-lived named P1 is reserved on the SFC machine, and the long-lasting named P2 is retained. One of P1 and P2 is the R-isomer and the other is the S-isomer.
  • Post-treatment quenching with saturated aqueous ammonium chloride solution, adding dichloromethane and aqueous phase to separate layers, the organic phase is desolvated under reduced pressure, and the product is obtained by silica gel chromatography column using petroleum ether/ethyl acetate (3:1) system. (3,5-Dimethoxyphenyl)-2-(5-fluoro-2-nitrobenzoyl)cyclohexanone 137c (5 g, 10 mmol, yellow solid).
  • 6-(3,5-Dimethoxyphenyl)-3-(5-fluoro-2-nitrophenyl)-4,5,6,7-tetrahydro-1H-indazole 137d (1 g, 2.5 Methyl) was mixed with 50 mL of acetic acid, and N-chlorosuccinimide (1 g, 7.5 mmol) was added and reacted at 80 ° C for 2 h.
  • P1 and P2 were obtained by splitting through a chiral column. Chiral separation conditions: equipment SFC, column: chiralpak-AD mobile phase: CO2-IPA (DEA). The short-lived named P1 is reserved on the SFC machine, and the long-lasting named P2 is retained. One of P1 and P2 is the R-isomer and the other is the S-isomer.
  • Example 140 was synthesized by following the procedure of Example 103, but in the first step, 1-methyl-4-nitro-5-formylchloropyrazole was substituted for 1-hydro-4-nitro-3-carbonyl-3-chloropyridyl Oxazole.
  • P1 and P2 were obtained by splitting through a chiral column. Chiral separation conditions: equipment SFC, column: chiralpak-AD mobile phase: CO2-IPA (DEA). The short-lived named P1 is reserved on the SFC machine, and the long-lasting named P2 is retained. One of P1 and P2 is the R-isomer and the other is the S-isomer.
  • Alkaline high performance liquid purity 99.59% (214 nm), 99.70% (254 nm).
  • Alkaline liquid purity 99.59% (214 nm), 99.70% (254 nm).
  • the compound 3-carbonylpiperidine-1-carboxylic acid tert-butyl ester 095a (20 g, 100.5 mmol, 1 eq) was dissolved in 400 mL of tetrahydrofuran, and lithium diisopropylamide (60.3 ml, 120.6) was added at -78 °C. Ment, 1.2 eq), after the addition was completed, the temperature was raised to -40 ° C for 1 hour, then 2-nitrobenzoyl chloride (18.6 g, 100.5 mmol, 1 eq) was added to the system at -40 ° C, and the reaction was allowed to rise to room temperature. hour.
  • the eighth and ninth steps are synthesized with reference to the example 289 steps.
  • the target product N-(2-(6-(2,6-dichloro-3,5-dimethoxyphenyl)-4,5,6,7-tetrahydro-1H-pyrazole[3, 4-c]pyridin-3-yl)phenyl)acrylamide.
  • Example 281 was synthesized with reference to the operational steps of Example 093.
  • the final product N-(5-(6-(2,6-difluoro-3,5-dimethoxyphenyl)-4,5,6,7-tetrahydro-1H-indazol-3-yl) was obtained.
  • P1 and P2 were obtained by splitting through a chiral column.
  • Example 283 was synthesized by the procedure of Example 137, but substituting N-N-dimethyl-N-methylethylenediamine with 1-methylpiperazine in the fifth step.
  • the target product N-(2-(6-(2,6-dichloro-3,5-dimethoxyphenyl)-4,5,6,7-tetrahydro-1H-indazol-3-yl) was finally obtained.
  • Example 284 was synthesized by reference to Example 133, but substituting N-N' dimethyl-N-methylethylenediamine with dimethylamine hydrochloride.
  • Example 285 was synthesized with reference to the operational steps of Example 137. However, in the fifth step, N-N' dimethyl-N-methylethylenediamine was replaced with dimethylamine hydrochloride. P1, P2 were obtained by splitting through a chiral column.

Landscapes

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

Abstract

一种通式(I)所示的芳香类衍生物及其可药用的盐或含有其的药物组合物、及其制备方法,以及上述芳香类衍生物及其可药用的盐或含有其的药物组合物在制备治疗剂,特别是FGFR4酪氨酸激酶抑制剂,和在制备治疗和/或预防肿瘤与炎症等疾病的药物中的用途。其中通式(I)的各取代基与说明书中的定义相同。

Description

芳香类衍生物、其制备方法及其在医药上的应用 发明领域
本发明涉及一种新的芳香类衍生物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,或含有其的药物组合物,及其制备方法。本发明还涉及使用所述芳香族醚类衍生物治疗和/或预防FGFR4酪氨酸激酶介导的疾病的方法,以及所述芳香族醚类衍生物在制备FGFR4酪氨酸激酶抑制剂和药物中的用途。
背景技术
成纤维细胞生长因子(FGF)是由垂体和下丘脑分泌的多肽,具有促进成纤维细胞有丝分裂、中胚层细胞的生长,刺激血管形成和在创伤愈合及肢体再生中发挥重要作用。FGF受体(FGFR)信号系统对正常的发育及生理过程具有至关重要的作用。FGFR有四种亚型FGFR1-4,这四种亚型的氨基酸序列有着高度的保守性。FGFR1-4对不同的生长因子有着不同的结合力,其在组织的分布也相互不同。一条完整的FGFR受体蛋白包括细胞外部分,疏水单链细胞膜部分和细胞内酪氨酸激酶部分。当FGFR细胞外部分和FGF生长因子相互作用,引发下游链式的信号传导(Ras-MAPK、AKT-PI3K、磷脂酶C),最终影响和调节细胞生长、分裂和分化(Eswarakumar,Cytokine&Growth Factor Reviews,2005)。
异常刺激这个信号通路(包括FGF生长因子过度表达、FGFR受体的过度表达和FGFR受体的基因突变)导致肿瘤的生长和对治疗的耐受。对几千种肿瘤样品的DNA测序发现FGFR通路常常发生突变。FGFR4是由FGFR-4基因编码的蛋白。FGFR4基因有着18个外显子。FGF-FGFR信号失调与肿瘤发生及演化相关。已经被发现在老鼠体内,FGFR4–FGF19信号轴与肝细胞癌(HCCs)紧密相关。肝癌等几种类型的癌症中,FGFR4表达被明显提高。同时,肝癌的发生需要FGFR4。FGF19转基因小鼠的后代能够发展为肝癌,然而,与FGFR4敲除的老鼠所繁殖的后代不会发展为肝癌。当FGF19被FGF19特异性抗体中和后,肿瘤的生长受到抑制。另外,FGFR4过度表达还出现在其他种类的肿瘤,包括乳腺癌,结直肠癌,胰腺癌,前列腺癌,肺癌,和甲状腺癌中。FGFR4突变出现在横纹肌肉瘤中。对于FGFR4的小分子靶向抑制可以被用于癌症的治疗中。当老鼠经过FGFR-1抑制剂治疗后,发现可以导致在软组织里发生磷酸钙沉积等副作用(Brown,AP et al.(2005),Toxicol.Pathol.,p.449-455)。这些表明了对FGFR4受体的选择性抑制,而不是广泛抑制FGFR1-4受体,会避免这样的副作用。
尽管目前已公开了一系列的FGFR4抑制剂的专利申请,其中包括WO2014011900、WO2015061572、WO20150197519、WO2015008844、WO2015057938、WO2015059668、WO2012174476、WO2015057963、WO2016064960、WO2016134294、WO2016134314、和WO2016134320等,但仍需开发新的具有更好药效的化合物。经过不断努力,本发明设计具有通式(I)所示的结构的化合物,并发现具有此类结构的化合物表现出优异的效果和作用。
发明内容
本发明涉及以下技术方案:
在一方面,本发明提供了通式(I)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物:
Figure PCTCN2018104007-appb-000001
其中:
环A不存在或者选自6-14元亚芳基、5-10元亚杂芳基、C 3-C 8亚环烷基或3-10元亚杂环基;
R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、-NR 11R 12、-C(O)R 13、-C(O)OR 13、-C(O)NR 11R 12、-NR 11C(O)R 13、-NR 11C(O)OR 13、-NR 11C(O)NR 11R 12、-OC(O)R 13、-OC(O)OR 13、-OC(O)NR 11R 12、-S(O) pR 13、-S(O) pOR 13、-S(O) pNR 11R 12、-OS(O) pR 13或-NR 11S(O) pR 13
R 6各自独立地选自H或-CH 2CH 2-,且当R 6为-CH 2CH 2-时,其另一端连接至X,任选地-CH 2CH 2-中的一个亚甲基被-O-或-NH-替代;
R 7各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;
R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;
R 11、R 12和R 13各自独立地选自H、C 1-C 6烷基、C 1-C 6杂烷基、C 3-C 8环烷基、3-10元杂环基、5-10元杂芳基或6-14元芳基;
X选自-O-、-NH-或-CH 2-;Y选自-O-、-NH-或-CH 2-;条件是X和Y中至少有一个是CH 2
W选自化学键、-NH-或-CH 2-;
m为0、1、2、3或4;
p为1或2。
在另一方面,本发明提供了一种药物组合物,其含有本发明化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,及药学上可接受的赋形剂。在具体实施方案中,本发明化合物以有效量提供在所述药物组合物中。在具体实施方案中,本发明化合物以治疗有效量提供。在具体实施方案中,本发明化合物以预防有效量提供。在另一实施方案中,本发明的药物组合物还含有其它治疗剂。
在另一方面,本发明提供了一种试剂盒,其包括:第一容器,其中含有本发明化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物; 和任选地,第二容器,其中含有其它治疗剂;和任选地,第三容器,其中含有用于稀释或悬浮所述化合物和/或其它治疗剂的药学上可接受的赋形剂。
在另一方面,本发明还提供了本发明化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,或本发明的药物组合物在制备FGFR4酪氨酸激酶抑制剂中的用途。
在另一方面,本发明还提供了本发明化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,或本发明的药物组合物在制备用于治疗和/或预防FGFR4酪氨酸激酶介导的疾病的药物中的用途。
在另一方面,本发明还提供了本发明化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,或本发明药物组合物,其用于治疗和/或预防FGFR4酪氨酸激酶介导的疾病。
在另一方面,本发明还提供了一种在受试者中治疗和/或预防FGFR4酪氨酸激酶介导的疾病的方法,包括向所述受试者给药本发明的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,或本发明的药物组合物。
在上述方面的具体实施方案中,所述疾病为肿瘤,例如胃癌、甲状腺癌、前列腺癌、乳腺癌、肉瘤(例如横纹肌肉瘤)、皮肤癌(例如黑色素瘤)、肝癌(例如肝细胞癌和胆管上皮癌)、胰腺癌(例如胰腺上皮内瘤样病变和胰腺导管腺癌)、肺癌(例如非小细胞肺癌和肺腺癌)、肾癌(例如肾细胞癌)、结直肠癌和卵巢癌。
具体实施方式
定义
本发明使用的术语具有本领域技术人员所熟知的含义。当在本发明中进行定义时,优先采用本发明的定义。
当列出数值范围时,既定包括每个值和在所述范围内的子范围。例如“C 1-C 6烷基”包括C 1、C 2、C 3、C 4、C 5、C 6、C 1-C 6、C 1-C 5、C 1-C 4、C 1-C 3、C 1-C 2、C 2-C 6、C 2-C 5、C 2-C 4、C 2-C 3、C 3-C 6、C 3-C 5、C 3-C 4、C 4-C 6、C 4-C 5和C 5-C 6烷基。
“C 1-C 6烷基”是指具有1至6个碳原子的直链或支链饱和烃基团,也称为“低级烷基”。在一些实施方案中,C 1-C 4烷基是特别优选的。所述烯基的实例包括但不限于:甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、及其各种支链异构体等。烷基可以是任选取代的或未取代的。
“C 2-C 6烯基”是指具有2至6个碳原子和一个或多个碳-碳双键(例如,1、2或3个碳-碳双键)的直链或支链烃基团。一个或多个碳-碳双键可以在内部(例如,在2-丁烯基中)或端部(例如,在1-丁烯基中)。在一些实施方案中,C 2-C 4烯基是特别优选的。所述烯基的实例包括但不限于:乙烯基、1-丙烯 基、2-丙烯基、1-丁烯基、2-丁烯基、丁二烯基、戊烯基、戊二烯基、己烯基、及其各种支链异构体等。烯基可以是任选取代的或未取代的。
“C 2-C 6炔基”是指具有2至6个碳原子、一个或多个碳-碳叁键(例如,1、2或3个碳-碳叁键)以及任选一个或多个碳-碳双键(例如,1、2或3个碳-碳双键)的直链或支链烃基团。在一些实施方案中,C 2-C 4炔基是特别优选的。在一些实施方案中,炔基不含有任何双键。一个或多个碳叁键可以在内部(例如,在2-丁炔基中)或端部(例如,在1-丁炔基中)。所述炔基的实例包括但不限于:乙炔基、1-丙炔基、2-丙炔基、1-丁炔基、2-丁炔基、戊炔基、己炔基、及其各种支链异构体等。炔基可以是任选取代的或未取代的。
“C 1-C 6杂烷基”是指本文所定义的烷基,在母体链内,它进一步含有一或多个(例如,1、2、3或4个)杂原子(例如,氧、硫、氮、硼、硅、磷),其中,一个或多个杂原子在所述母体碳链内的相邻碳原子之间,和/或,一个或多个杂原子在碳原子和母体分子之间,即,在连接点之间。杂烷基可以是任选取代的或未取代的。在一些实施方案中,作为具体实例,C 1-C 6杂烷基包括C 1-C 6烷氧基、C 1-C 6烷硫基、C 1-C 6烷氨基等,其详细定义如下。
“C 1-C 6烷氧基”是指基团-OR,其中,R为取代或未取代的C 1-C 6烷基。在一些实施方案中,C 1-C 4烷氧基是特别优选的。具体的所述烷氧基包括但不限于:甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、叔丁氧基、仲丁氧基、正戊氧基、正己氧基和1,2-二甲基丁氧基、及其各种支链异构体等。烷氧基可以是任选取代的或未取代的。
“C 1-C 6烷硫基”是指基团-SR,其中,R为任选取代的C 1-C 6烷基。在一些实施方案中,C 1-C 4烷硫基是特别优选的。具体的所述C 1-C 6烷硫基包括但不限于:甲硫基、乙硫基、正丙硫基、异丙硫基、正丁硫基、叔丁硫基、仲丁硫基、正戊硫基、正己硫基和1,2-二甲基丁硫基、及其各种支链异构体等。烷硫基可以是任选取代的或未取代的。
“C 1-C 6烷氨基”是指基团-NHR或者-NR 2,其中,R为任选取代的C 1-C 6烷基。在一些实施方案中,C 1-C 4烷氨基是特别优选的。具体的所述C 1-C 6烷氨基包括但不限于:甲氨基、乙氨基、正丙氨基、异丙氨基、正丁氨基、叔丁氨基、二甲氨基、甲乙氨基和二乙氨基、及其各种支链异构体等。烷氨基可以是任选取代的或未取代的。
“卤素”是指氟(F)、氯(Cl)、溴(Br)和碘(I)。
“氰基”是指-CN。
“C 1-C 6卤代烷基”和“C 1-C 6卤代烷氧基”是指上述“C 1-C 6烷基”和“C 1-C 6烷氧基”,其被一个或多个卤素基团取代。在一些实施方案中,C 1-C 4卤代烷基是特别优选的,更优选C 1-C 2卤代烷基。在一些实施方案中,C 1-C 4卤代烷氧基是特别优选的,更优选C 1-C 2卤代烷氧基。示例性的所述卤代烷基包括但不限于:-CF 3、-CH 2F、-CHF 2、-CHFCH 2F、-CH 2CHF 2、-CF 2CF 3、-CCl 3、-CH 2Cl、-CHCl 2、2,2,2-三氟-1,1-二甲基-乙基等。示例性的所述卤代烷氧基包括但不限于:-OCH 2F、-OCHF 2、-OCF 3等。卤代烷基和卤代烷氧基可以是任选取代的或未取代的。
“C 3-C 8环烷基”是指具有3至8个环碳原子和零个杂原子的非芳香环烃基团。在一些实施方案中,C 3-C 6环烷基是特别优选的,更优选C 5-C 6环烷基。环烷基还包括其中上述环烷基与一个或多个环烷 基、杂环基、芳基或杂芳基稠合、桥接或成螺环的环体系,其中连接点在环烷基环上,且在这样的情况中,碳的数目继续表示环烷基体系中的碳的数目。示例性的所述环烷基包括但不限于:环丙基、环丙烯基、环丁基、环丁烯基、环戊基、环戊烯基、环己基、环己烯基、环已二烯基、环庚基、环庚烯基、环庚二烯基、环庚三烯基、环辛基、环辛烯基、二环[2.2.1]庚基、二环[2.2.2]辛基、及其各种支链异构体等。环烷基可以是任选取代的或未取代的。
“3-10元杂环基”或是指具有环碳原子和1至4个环杂原子的3至10元非芳香环系的基团,其中,每个杂原子独立地选自氮、氧、硫、硼、磷和硅。在包含一个或多个氮原子的杂环基中,只要化合价允许,连接点可为碳或氮原子。在一些实施方案中,3-8元杂环基是优选的,其为具有环碳原子和1至3个环杂原子的3至8元非芳香环系;在一些实施方案中,4-7元杂环基是特别优选的,其为具有环碳原子和1至3个环杂原子的4至7元非芳香环系;更优选5-6元杂环基,其为具有环碳原子和1至3个环杂原子的5至6元非芳香环系。示例性的包含一个杂原子的3元杂环基包括但不限于:氮杂环丙烷基、氧杂环丙烷基、硫杂环丙烷基。示例性的含有一个杂原子的4元杂环基包括但不限于:氮杂环丁烷基、氧杂环丁烷基和硫杂环丁烷基。示例性的含有一个杂原子的5元杂环基包括但不限于:四氢呋喃基、二氢呋喃基、四氢噻吩基、二氢噻吩基、吡咯烷基、二氢吡咯基和吡咯基-2,5-二酮。示例性的包含两个杂原子的5元杂环基包括但不限于:二氧杂环戊烷基、氧硫杂环戊烷基、二硫杂环戊烷基和噁唑烷-2-酮。示例性的包含三个杂原子的5元杂环基包括但不限于:三唑啉基、噁二唑啉基和噻二唑啉基。示例性的包含一个杂原子的6元杂环基包括但不限于:哌啶基、二氢吡喃基、四氢吡喃基、二氢吡啶基和硫杂环己烷基。示例性的包含两个杂原子的6元杂环基包括但不限于:哌嗪基、吗啉基、二硫杂环己烷基、二噁烷基。示例性的包含三个杂原子的6元杂环基包括但不限于:六氢三嗪基。示例性的含有一个杂原子的7元杂环基包括但不限于:氮杂环庚烷基、氧杂环庚烷基和硫杂环庚烷基。示例性的包含一个杂原子的8元杂环基包括但不限于:氮杂环辛烷基、氧杂环辛烷基和硫杂环辛烷基。示例性的与6元芳基环稠合的5元杂环基(在本文中也称作5,6-双环杂环基)包括但不限于:二氢吲哚基、异二氢吲哚基、二氢苯并呋喃基、二氢苯并噻吩基、苯并噁唑啉酮基,等等。示例性的与6元芳基环稠合的6元杂环基(本文还指的是6,6-双环杂环基)包括但不限于:四氢喹啉基、四氢异喹啉基,等等。杂环基包括螺环、稠环和桥环的杂环基。
“螺杂环基”指5至20元,单环之间共用一个原子(称螺原子)的多环杂环基团,其中一个或多个环原子选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。这些可以含有一个或多个双键,但没有一个环具有芳香性。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺环烷基分为单螺杂环基、双螺杂环基或多螺杂环基,优选为单螺环烷基和双螺环烷基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺环烷基。螺环烷基的非限制性实例包括:
Figure PCTCN2018104007-appb-000002
“稠杂环基”指5至20元,系统中的每个环与体系中的其他环共享毗邻的一对原子的多环杂环基 团,一个或多个环可以含有一个或多个双键,但没有一个环具有芳香性,其中一个或多个环原子选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠杂环烷基,优选为双环或三环,更优选为5元/5元或5元/6元双环稠杂环基。稠杂环基的非限制性实例包括:
Figure PCTCN2018104007-appb-000003
所述杂环基环可以稠合于芳基、杂芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基,非限制性实例包括:
Figure PCTCN2018104007-appb-000004
等。
杂环基可以是任选取代的或未取代的。
“6-14元芳基”是指具有6-14个环碳原子和零个杂原子的单环或多环的(例如,双环或三环)4n+2芳族环体系(例如,具有以环状排列共享的6、10或14个π电子)的基团。在一些实施方案中,芳基具有六个环碳原子(“6元芳基”;例如,苯基)。在一些实施方案中,芳基具有十个环碳原子(“10元芳基”;例如,萘基,例如,1-萘基和2-萘基)。在一些实施方案中,芳基具有十四个环碳原子(“14元芳基”;例如,蒽基)。在一些实施方案中,6-10元芳基是特别优选的,更优选6元芳基。所述芳基可以稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为芳基环,非限制性实例包括:
Figure PCTCN2018104007-appb-000005
芳基可以是取代的或未取代的。
“5-10元杂芳基”是指具有环碳原子和1-4个环杂原子的5-10元单环或双环的4n+2芳族环体系(例如,具有以环状排列共享的6或10个π电子)的基团,其中每个杂原子独立地选自氮、氧和硫。在含有一个或多个氮原子的杂芳基中,只要化合价允许,连接点可以是碳或氮原子。在一些实施方案中,5-6元杂芳基是特别优选的,其为具有环碳原子和1-4个环杂原子的5-6元单环的4n+2芳族环体系。示例性的含有一个杂原子的5元杂芳基包括但不限于:吡咯基、呋喃基和噻吩基。示例性的含有两个杂原子的5元杂芳基包括但不限于:咪唑基、吡唑基、噁唑基、异噁唑基、噻唑基和异噻唑基。示例性的含有三个杂原子的5元杂芳基包括但不限于:三唑基、噁二唑基和噻二唑基。示例性的含有四个杂原子的5元杂芳基包括但不限于:四唑基。示例性的含有一个杂原子的6元杂芳基包括但不限于:吡啶基。示例性的含有两个杂原子的6元杂芳基包括但不限于:哒嗪基、嘧啶基和吡嗪基。示例性的 含有三个或四个杂原子的6元杂芳基分别包括但不限于:三嗪基和四嗪基。示例性的含有一个杂原子的7元杂芳基包括但不限于:氮杂环庚三烯基、氧杂环庚三烯基和硫杂环庚三烯基。示例性的5,6-双环杂芳基包括但不限于:吲哚基、异吲哚基、吲唑基、苯并三唑基、苯并噻吩基、异苯并噻吩基、苯并呋喃基、苯并异呋喃基、苯并咪唑基、苯并噁唑基、苯并异噁唑基、苯并噁二唑基、苯并噻唑基、苯并异噻唑基、苯并噻二唑基、茚嗪基和嘌呤基。示例性的6,6-双环杂芳基包括但不限于:萘啶基、喋啶基、喹啉基、异喹啉基、噌琳基、喹喔啉基、酞嗪基和喹唑啉基。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,非限制性实例包括:
Figure PCTCN2018104007-appb-000006
杂芳基可以是任选取代的或未取代的。
本文使用的“亚烷基”、“亚烯基”、“亚炔基”、“亚芳基”、“亚杂芳基”、“亚环烷基”和“亚杂环基”分别指的是上述烷基、烯基、炔基、芳基、杂芳基、环烷基和杂环基的二价基团。所述基团可以是任选取代的或未取代的。
“C 1-C 6亚烷基”是指除去C 1-C 6烷基的一个氢而形成的二价的亚烷基,并且可以是取代或未取代的亚烷基。在一些实施方案中,C 1-C 4亚烷基是特别优选的。未取代的所述亚烷基包括但不限于:亚甲基(-CH 2-)、亚乙基(-CH 2CH 2-)、亚丙基(-CH 2CH 2CH 2-)、亚丁基(-CH 2CH 2CH 2CH 2-)、亚戊基(-CH 2CH 2CH 2CH 2CH 2-)、亚己基(-CH 2CH 2CH 2CH 2CH 2CH 2-),等等。示例性的取代的所述亚烷基,例如,被一个或多个烷基(甲基)取代的所述亚烷基,包括但不限于:取代的亚甲基(-CH(CH 3)-、-C(CH 3) 2-)、取代的亚乙基(-CH(CH 3)CH 2-、-CH 2CH(CH 3)-、-C(CH 3) 2CH 2-、-CH 2C(CH 3) 2-)、取代的亚丙基(-CH(CH 3)CH 2CH 2-、-CH 2CH(CH 3)CH 2-、-CH 2CH 2CH(CH 3)-、-C(CH 3) 2CH 2CH 2-、-CH 2C(CH 3) 2CH 2-、-CH 2CH 2C(CH 3) 2-),等等。
“C 2-C 6亚烯基”是指除去C 2-C 6烯基的一个氢而形成的二价的亚烯基,并且可以是取代或未取代的亚烯基。在一些实施方案中,C 2-C 4亚烯基是特别优选的。示例性的未取代的所述亚烯基包括但不限于:亚乙烯基(-CH=CH-)和亚丙烯基(例如,-CH=CHCH 2-、-CH 2-CH=CH-)。示例性的取代的所述亚烯基,例如,被一个或多个烷基(甲基)取代的亚烯基,包括但不限于:取代的亚乙基(-C(CH 3)=CH-、-CH=C(CH 3)-)、取代的亚丙烯基(-C(CH 3)=CHCH 2-、-CH=C(CH 3)CH 2-、-CH=CHCH(CH 3)-、-CH=CHC(CH 3) 2-、-CH(CH 3)-CH=CH-、-C(CH 3) 2-CH=CH-、-CH 2-C(CH 3)=CH-、-CH 2-CH=C(CH 3)-),等等。
“C 2-C 6亚炔基”是指除去C 2-C 6炔基的一个氢而形成的二价的亚炔基,并且可以是取代或未取代的亚炔基。在一些实施方案中,C 2-C 4亚炔基是特别优选的。示例性的所述亚炔基包括但不限于:亚乙炔基(-C≡C-)、取代或未取代的亚丙炔基(-C≡CCH 2-),等等。
“6-14元亚芳基”、“5-10元亚杂芳基”、“C 3-C 8亚环烷基”和“3-10元亚杂环基”分别指的是除去上述6-14元芳基、5-10元杂芳基、C 3-C 8环烷基和3-10元杂环基的一个氢而形成的二价基团。
本发明的所有基团均任选地被取代。“任选”或“任选地”意味着随后所描述地事件或情形可以但不必发生,该说明包括该事件或情形发生或不发生二者。例如,“任选被烷基取代的杂环基团”意味着烷基可以但不必须存在,该说明包括杂环基团被烷基取代的情形和杂环基团不被烷基取代的情形。
“取代的”指基团中的一个或多个氢原子,优选为最多5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验或理论)可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。
示例性的碳原子上的取代基包括但不局限于:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OR aa、-ON(R bb) 2、-N(R bb) 2、-N(R bb) 3 +X -、-N(OR cc)R bb、-SH、-SR aa、-SSR cc、-C(=O)R aa、-CO 2H、-CHO、-C(OR cc) 2、-CO 2R aa、-OC(=O)R aa、-OCO 2R aa、-C(=O)N(R bb) 2、-OC(=O)N(R bb) 2、-NR bbC(=O)R aa、-NR bbCO 2R aa、-NR bbC(=O)N(R bb) 2、-C(=NR bb)R aa、-C(=NR bb)OR aa、-OC(=NR bb)R aa、-OC(=NR bb)OR aa、-C(=NR bb)N(R bb) 2、-OC(=NR bb)N(R bb) 2、-NR bbC(=NR bb)N(R bb) 2、-C(=O)NR bbSO 2R aa、-NR bbSO 2R aa、-SO 2N(R bb) 2、-SO 2R aa、-SO 2OR aa、-OSO 2R aa、-S(=O)R aa、-OS(=O)R aa、-Si(R aa) 3、-OSi(R aa) 3、-C(=S)N(R bb) 2、-C(=O)SR aa、-C(=S)SR aa、-SC(=S)SR aa、-SC(=O)SR aa、-OC(=O)SR aa、-SC(=O)OR aa、-SC(=O)R aa、-P(=O) 2R aa、-OP(=O) 2R aa、-P(=O)(R aa) 2、-OP(=O)(R aa) 2、-OP(=O)(OR cc) 2、-P(=O) 2N(R bb) 2、-OP(=O) 2N(R bb) 2、-P(=O)(NR bb) 2、-OP(=O)(NR bb) 2、-NR bbP(=O)(OR cc) 2、-NR bbP(=O)(NR bb) 2、-P(R cc) 2、-P(R cc) 3、-OP(R cc) 2、-OP(R cc) 3、-B(R aa) 2、-B(OR cc) 2、-BR aa(OR cc)、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
或者在碳原子上的两个偕氢被基团=O、=S、=NN(R bb) 2、=NNR bbC(=O)R aa、=NNR bbC(=O)OR aa、=NNR bbS(=O) 2R aa、=NR bb或=NOR cc取代;
R aa的每个独立地选自烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个R aa基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R bb的每个独立地选自:氢、-OH、-OR aa、-N(R cc) 2、-CN、-C(=O)R aa、-C(=O)N(R cc) 2、-CO 2R aa、-SO 2R aa、-C(=NR cc)OR aa、-C(=NR cc)N(R cc) 2、-SO 2N(R cc) 2、-SO 2R cc、-SO 2OR cc、-SOR aa、-C(=S)N(R cc) 2、-C(=O)SR cc、-C(=S)SR cc、-P(=O) 2R aa、-P(=O)(R aa) 2、-P(=O) 2N(R cc) 2、-P(=O)(NR cc) 2、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个R bb基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R cc的每个独立地选自氢、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个R cc基团结合以形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代;
R dd的每个独立地选自:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OR ee、-ON(R ff) 2、-N(R ff) 2,、-N(R ff) 3 +X -、-N(OR ee)R ff、-SH、-SR ee、-SSR ee、-C(=O)R ee、-CO 2H、-CO 2R ee、-OC(=O)R ee、-OCO 2R ee、 -C(=O)N(R ff) 2、-OC(=O)N(R ff) 2、-NR ffC(=O)R ee、-NR ffCO 2R ee、-NR ffC(=O)N(R ff) 2、-C(=NR ff)OR ee、-OC(=NR ff)R ee、-OC(=NR ff)OR ee、-C(=NR ff)N(R ff) 2、-OC(=NR ff)N(R ff) 2、-NR ffC(=NR ff)N(R ff) 2、-NR ffSO 2R ee、-SO 2N(R ff) 2、-SO 2R ee、-SO 2OR ee、-OSO 2R ee、-S(=O)R ee、-Si(R ee) 3、-OSi(R ee) 3、-C(=S)N(R ff) 2、-C(=O)SR ee、-C(=S)SR ee、-SC(=S)SR ee、-P(=O) 2R ee、-P(=O)(R ee) 2、-OP(=O)(R ee) 2、-OP(=O)(OR ee) 2、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基、杂芳基,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代,或者两个偕R dd取代基可结合以形成=O或=S;
R ee的每个独立地选自烷基、卤代烷基、烯基、炔基、环烷基、芳基、杂环基和杂芳基,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代;
R ff的每个独立地选自氢、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者两个R ff基团结合形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R gg基团取代;
R gg的每个独立地是:卤素、-CN、-NO 2、-N 3、-SO 2H、-SO 3H、-OH、-OC 1-6烷基、-ON(C 1-6烷基) 2、-N(C 1-6烷基) 2、-N(C 1-6烷基) 3 +X -、-NH(C 1-6烷基) 2 +X -、-NH 2(C 1-6烷基) +X -、-NH 3 +X -、-N(OC 1-6烷基)(C 1-6烷基)、-N(OH)(C 1-6烷基)、-NH(OH)、-SH、-SC 1-6烷基、-SS(C 1-6烷基)、-C(=O)(C 1-6烷基)、-CO 2H、-CO 2(C 1-6烷基)、-OC(=O)(C 1-6烷基)、-OCO 2(C 1-6烷基)、-C(=O)NH 2、-C(=O)N(C 1-6烷基) 2、-OC(=O)NH(C 1-6烷基)、-NHC(=O)(C 1-6烷基)、-N(C 1-6烷基)C(=O)(C 1-6烷基)、-NHCO 2(C 1-6烷基)、-NHC(=O)N(C 1-6烷基) 2、-NHC(=O)NH(C 1-6烷基)、-NHC(=O)NH 2、-C(=NH)O(C 1-6烷基)、-OC(=NH)(C 1-6烷基)、-OC(=NH)OC 1-6烷基、-C(=NH)N(C 1-6烷基) 2、-C(=NH)NH(C 1-6烷基)、-C(=NH)NH 2、-OC(=NH)N(C 1-6烷基) 2、-OC(NH)NH(C 1-6烷基)、-OC(NH)NH 2、-NHC(NH)N(C 1-6烷基) 2、-NHC(=NH)NH 2、-NHSO 2(C 1-6烷基)、-SO 2N(C 1-6烷基) 2、-SO 2NH(C 1-6烷基)、-SO 2NH 2、-SO 2C 1-6烷基、-SO 2OC 1-6烷基、-OSO 2C 1-6烷基、-SOC 1-6烷基、-Si(C 1-6烷基) 3、-OSi(C 1-6烷基) 3、-C(=S)N(C 1-6烷基) 2、C(=S)NH(C 1-6烷基)、C(=S)NH 2、-C(=O)S(C 1-6烷基)、-C(=S)SC 1-6烷基、-SC(=S)SC 1-6烷基、-P(=O) 2(C 1-6烷基)、-P(=O)(C 1-6烷基) 2、-OP(=O)(C 1-6烷基) 2、-OP(=O)(OC 1-6烷基) 2、C 1-6烷基、C 1-6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 7环烷基、C 6-C 10芳基、C 3-C 7杂环基、C 5-C 10杂芳基;或者两个偕R gg取代基可结合形成=O或=S;其中,X -为反离子。
示例性的氮原子上取代基包括但不局限于:氢、-OH、-OR aa、-N(R cc) 2、-CN、-C(=O)R aa、-C(=O)N(R cc) 2、-CO 2R aa、-SO 2R aa、-C(=NR bb)R aa、-C(=NR cc)OR aa、-C(=NR cc)N(R cc) 2、-SO 2N(R cc) 2、-SO 2R cc、-SO 2OR cc、-SOR aa、-C(=S)N(R cc) 2、-C(=O)SR cc、-C(=S)SR cc、-P(=O) 2R aa、-P(=O)(R aa) 2、-P(=O) 2N(R cc) 2、-P(=O)(NR cc) 2、烷基、卤代烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基,或者连接至氮原子的两个R cc基团结合形成杂环基或杂芳基环,其中,每个烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基独立地被0、1、2、3、4或5个R dd基团取代,且其中R aa、R bb、R cc和R dd如上所述。
术语“可药用的盐”是指,在可靠的医学判断范围内,适合与人和低等动物的组织接触而没有过度毒性、刺激性、变态反应等等,并且与合理的益处/危险比例相称的那些盐。可药用的盐在本领域是 众所周知的。例如,Berge等人在J.Pharmaceutical Sciences(1977)66:1-19中详细描述的可药用的盐。本发明化合物的可药用的盐包括衍生自合适的无机和有机酸和无机和有机碱的盐。药学上可接受的无毒的酸加成盐的实例是与无机酸形成的盐,例如盐酸、氢溴酸、磷酸、硫酸和高氯酸,或与有机酸形成的盐,例如乙酸、草酸、马来酸、酒石酸、枸橼酸、琥珀酸或丙二酸。也包括使用本领域常规方法形成的盐,例如,离子交换方法。其它可药用的盐包括:已二酸盐、海藻酸盐、抗坏血酸盐、天冬氨酸盐、苯磺酸盐、苯甲酸盐、重硫酸盐、硼酸盐、丁酸盐、樟脑酸盐、樟脑磺酸盐、柠檬酸盐、环戊丙酸盐、二葡糖酸盐、十二烷基硫酸盐、乙磺酸盐、甲酸盐、富马酸盐、葡萄糖酸盐、甘油磷酸盐、葡糖酸盐、半硫酸盐、庚酸盐、己酸盐、氢碘酸盐、2-羟基-乙磺酸盐、乳糖酸盐、乳酸盐、月桂酸盐、月桂基硫酸盐、苹果酸盐、马来酸盐、丙二酸盐、甲磺酸盐、2-萘磺酸盐、烟酸盐、硝酸盐、油酸盐、草酸盐、棕榈酸盐、双羟萘酸盐、果胶酯酸盐、过硫酸盐、3-苯丙酸盐、磷酸盐、苦味酸盐、特戊酸盐、丙酸盐、硬脂酸盐、琥珀酸盐、硫酸盐、酒石酸盐、硫氰酸盐、对甲苯磺酸盐、十一烷酸盐、戊酸盐,等等。衍生自合适的碱的可药用的盐包括碱金属、碱土金属、铵和N +(C 1-4烷基) 4盐。代表性的碱金属或碱土金属盐包括钠、锂、钾、钙、镁盐,等等。如果合适的话,其它的可药用的盐包括与反离子形成的无毒的铵盐、季铵盐和胺阳离子,反离子例如卤离子、氢氧根、羧酸根、硫酸根、磷酸根、硝酸根、低级烷基磺酸根和芳基磺酸根。
给药的“受试者”包括但不限于:人(即,任何年龄组的男性或女性,例如,儿科受试者(例如,婴儿、儿童、青少年)或成人受试者(例如,年轻的成人、中年的成人或年长的成人))和/或非人的动物,例如,哺乳动物,例如,灵长类(例如,食蟹猴、恒河猴)、牛、猪、马、绵羊、山羊、啮齿动物、猫和/或狗。在一些实施方案中,受试者是人。在一些实施方案中,受试者是非人动物。
“疾病”、“障碍”和“病症”在本文中可互换地使用。
除非另作说明,否则,本文使用的术语“治疗”包括受试者患有具体疾病、障碍或病症时所发生的作用,它降低疾病、障碍或病症的严重程度,或延迟或减缓疾病、障碍或病症的发展(“治疗性治疗”),还包括在受试者开始患有具体疾病、障碍或病症之前发生的作用(“预防性治疗”)。
通常,化合物的“有效量”是指足以引起目标生物反应的数量。正如本领域普通技术人员所理解的那样,本发明化合物的有效量可以根据下列因素而改变:例如,生物学目标、化合物的药代动力学、所治疗的疾病、给药模式以及受试者的年龄健康情况和症状。有效量包括治疗有效量和预防有效量。
除非另作说明,否则,本文使用的化合物的“治疗有效量”是在治疗疾病、障碍或病症的过程中足以提供治疗益处的量,或使与疾病、障碍或病症有关的一或多种症状延迟或最小化的量。化合物的治疗有效量是指单独使用或与其它疗法联用时,治疗剂的量,它在治疗疾病、障碍或病症的过程中提供治疗益处。术语“治疗有效量”可以包括改善总体治疗、降低或避免疾病或病症的症状或病因、或增强其它治疗剂的治疗效果的量。
除非另作说明,否则,本文使用的化合物的“预防有效量”是足以预防疾病、障碍或病症的量,或足以预防与疾病、障碍或病症有关的一或多种症状的量,或防止疾病、障碍或病症复发的量。化合物的预防有效量是指单独使用或与其它药剂联用时,治疗剂的量,它在预防疾病、障碍或病症的过程中提供预防益处。术语“预防有效量”可以包括改善总体预防的量,或增强其它预防药剂的预防效果的量。
“组合”以及相关术语是指同时或依次给药本发明化合物和其它治疗剂。例如,本发明化合物可以与其它治疗剂以分开的单位剂型同时或依次给药,或与其它治疗剂一起在单一单位剂型中同时给药。
“前药”是指在体内通过例如在血液中水解转变成其具有医学效应的活性形式的化合物。药学上可接受的前药描述于T.Higuchi和V.Stella,Prodrugs as Novel Delivery Systems,A.C.S.Symposium Series,Vol.14,Edward B.Roche,ed.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987,以及D.Fleisher、S.Ramon和H.Barbra“Improved oral drug delivery:solubility limitations overcome by the use of prodrugs”,Advanced Drug Delivery Reviews(1996)19(2)115-130,将每篇引入本文作为参考。
前药为任何共价键合的载体,当将这种前药给予患者时,其在体内释放本发明化合物。通常通过修饰官能团来制备前药,该修饰使得前药在体内裂解产生母体化合物。前药包括,例如,其中羟基、氨基或巯基与任意基团键合的本发明化合物,当将其给予患者时,可以裂解形成羟基、氨基或巯基。因此,前药的代表性实例包括但不限于,本发明化合物通过其中的羟基、氨基或巯基官能团与乙酸、甲酸或苯甲酸形成的共价衍生物。另外,在羧酸(-COOH)的情况下,可以使用酯,例如甲酯、乙酯等。酯本身可以是有活性的和/或可以在人体体内条件下水解。合适的药学上可接受的体内可水解的酯包括容易在人体中分解而释放母体酸或其盐的那些。
本领域技术人员将理解,许多有机化合物可以与溶剂形成复合物,其在该溶剂中发生反应或从该溶剂中沉淀或结晶出来。这些复合物称为“溶剂合物”。当溶剂是水时,复合物称为“水合物”。本发明涵盖了本发明化合物的所有溶剂合物。
本发明化合物可包括一个或多个不对称中心,且因此可以存在多种立体异构体形式,例如,对映异构体、非对映异构体和外消旋体形式。例如,本发明化合物可为单独的对映异构体、非对映异构体或几何异构体(例如顺式和反式异构体),或者可为立体异构体的混合物的形式,包括外消旋混合物和富含一种或多种立体异构体的混合物。异构体可通过本领域技术人员已知的方法从混合物中分离,所述方法包括:手性高压液相色谱法(HPLC)以及手性盐的形成和结晶;或者优选的异构体可通过不对称合成来制备。
此外,本发明化合物还可以以“互变异构体”存在。“互变异构体”是指由于有机化合物的结构在两种官能团间产生平衡互相转换的现象而产生的异构体。
化合物
在一个实施方案中,本发明提供了一种通式(I)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物:
Figure PCTCN2018104007-appb-000007
其中:
环A不存在或者选自6-14元亚芳基、5-10元亚杂芳基、C 3-C 8亚环烷基或3-10元亚杂环基;
R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、-NR 11R 12、-C(O)R 13、-C(O)OR 13、-C(O)NR 11R 12、-NR 11C(O)R 13、-NR 11C(O)OR 13、-NR 11C(O)NR 11R 12、-OC(O)R 13、-OC(O)OR 13、-OC(O)NR 11R 12、-S(O) pR 13、-S(O) pOR 13、-S(O) pNR 11R 12、-OS(O) pR 13或-NR 11S(O) pR 13
R 6各自独立地选自H或-CH 2CH 2-,且当R 6为-CH 2CH 2-时,其另一端连接至X,任选地-CH 2CH 2-中的一个亚甲基被-O-或-NH-替代;
R 7各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;
R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;
R 11、R 12和R 13各自独立地选自H、C 1-C 6烷基、C 1-C 6杂烷基、C 3-C 8环烷基、3-10元杂环基、5-10元杂芳基或6-14元芳基;
X选自-O-、-NH-或-CH 2-;Y选自-O-、-NH-或-CH 2-;条件是X和Y中至少有一个是CH 2
W选自化学键、-NH-或-CH 2-;
m为0、1、2、3或4;
p为1或2。
在具体实施方案中,R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基、C 1-C 6烷氧基或C 1-C 6卤代烷氧基。
在具体实施方案中,R 1和R 5各自独立地选自卤素,优选为Cl或F。
在具体实施方案中,R 2和R 4各自独立地选自C 1-C 6烷氧基,优选为甲氧基。
在具体实施方案中,R 3为H。
在具体实施方案中,R 6选自H。
在具体实施方案中,R 7各自独立地选自H、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基,或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基 或C 1-C 6卤代烷基取代;优选R 7选自H或C 1-C 6烷基或-NR aR b,优选为甲基或-N(CH 3)CH 2CH 2N(CH 3) 2
在具体实施方案中,R 8选自C 2-C 6烯基或C 2-C 6炔基,优选选自乙烯基或丙炔基。
在具体实施方案中,环A选自6-14元亚芳基,优选为6元亚芳基。
在具体实施方案中,环A选自5-10元亚杂芳基,优选为5-6元亚杂芳基,优选为5元亚杂芳基。
在具体实施方案中,环A选自C 3-C 8亚环烷基。
在具体实施方案中,环A选自苯基、萘基、吡咯基、呋喃基、噻吩基、咪唑基、吡唑基、噁唑基、异噁唑基、噻唑基、异噻唑基、三唑基、噁二唑基、噻二唑基、四唑基、吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基、四嗪基或环己基的二价基团,优选选自亚苯基、亚吡唑基或亚环己基。
在具体实施方案中,W选自化学键。
在具体实施方案中,m为1。
在另一实施方案中,本发明提供了通式(II)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物:
Figure PCTCN2018104007-appb-000008
其中:
环A不存在或者选自6-14元亚芳基、5-10元亚杂芳基、C 3-C 8亚环烷基或3-10元亚杂环基;
R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、-NR 11R 12、-C(O)R 13、-C(O)OR 13、-C(O)NR 11R 12、-NR 11C(O)R 13、-NR 11C(O)OR 13、-NR 11C(O)NR 11R 12、-OC(O)R 13、-OC(O)OR 13、-OC(O)NR 11R 12、-S(O) pR 13、-S(O) pOR 13、-S(O) pNR 11R 12、-OS(O) pR 13或-NR 11S(O) pR 13
R 6各自独立地选自H或-CH 2CH 2-,且当R 6为-CH 2CH 2-时,其另一端连接至X,任选地-CH 2CH 2-中的一个亚甲基被-O-或-NH-替代;
R 7各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;
R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;
R 11、R 12和R 13各自独立地选自H、C 1-C 6烷基、C 1-C 6杂烷基、C 3-C 8环烷基、3-10元杂环基、5-10元杂芳基或6-14元芳基;
X选自-O-、-NH-或-CH 2-;Y选自-O-、-NH-或-CH 2-;条件是X和Y中至少有一个是CH 2
m为0、1、2、3或4;
p为1或2。
在具体实施方案中,环A选自6-14元亚芳基,优选为6元亚芳基。
在具体实施方案中,环A选自5-10元亚杂芳基,优选为5-6元亚杂芳基,优选为5元亚杂芳基。
在具体实施方案中,环A选自C 3-C 8亚环烷基。
在具体实施方案中,环A选自苯基、萘基、吡咯基、呋喃基、噻吩基、咪唑基、吡唑基、噁唑基、异噁唑基、噻唑基、异噻唑基、三唑基、噁二唑基、噻二唑基、四唑基、吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基、四嗪基或环己基的二价基团,优选选自亚苯基、亚吡唑基或亚环己基。
在具体实施方案中,R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基、C 1-C 6烷氧基或C 1-C 6卤代烷氧基。
在具体实施方案中,R 1和R 5各自独立地选自卤素,优选为Cl或F。
在具体实施方案中,R 2和R 4各自独立地选自C 1-C 6烷氧基,优选为甲氧基。
在具体实施方案中,R 3为H。
在具体实施方案中,R 6各自独立地选自H或-CH 2CH 2-,且当R 6为-CH 2CH 2-时,其另一端连接至X。在具体实施方案中,R 6为H。
在具体实施方案中,R 6选自H。
在具体实施方案中,R 7各自独立地选自H、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基,优选R 7选自H或C 1-C 6烷基或-NR aR b,优选为甲基或-N(CH 3)CH 2CH 2N(CH 3) 2,优选为H。
在具体实施方案中,R 8选自C 2-C 6烯基或C 2-C 6炔基,优选选自乙烯基或丙炔基。
在具体实施方案中,X选自-O-、-NH-或-CH 2-;Y选自-O-、-NH-或-CH 2-;条件是X和Y中至少有一个是CH 2
在具体实施方案中,X为-CH 2-;Y选自-O-、-NH-或-CH 2-;
在具体实施方案中,m为1。
在另一实施方案中,本发明提供了通式(III)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物:
Figure PCTCN2018104007-appb-000009
其中:
环A不存在或者选自6-14元亚芳基、5-10元亚杂芳基、C 3-C 8亚环烷基或3-10元亚杂环基;
R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6 烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、-NR 11R 12、-C(O)R 13、-C(O)OR 13、-C(O)NR 11R 12、-NR 11C(O)R 13、-NR 11C(O)OR 13、-NR 11C(O)NR 11R 12、-OC(O)R 13、-OC(O)OR 13、-OC(O)NR 11R 12、-S(O) pR 13、-S(O) pOR 13、-S(O) pNR 11R 12、-OS(O) pR 13或-NR 11S(O) pR 13
R 7各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;
R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;
R 11、R 12和R 13各自独立地选自H、C 1-C 6烷基、C 1-C 6杂烷基、C 3-C 8环烷基、3-10元杂环基、5-10元杂芳基或6-14元芳基;
m为0、1、2、3或4;
p为1或2。
在具体实施方案中,环A选自6-14元亚芳基,优选为6元亚芳基。
在具体实施方案中,环A选自5-10元亚杂芳基,优选为5-6元亚杂芳基,优选为5元亚杂芳基。
在具体实施方案中,环A选自C 3-C 8亚环烷基。
在具体实施方案中,环A选自苯基、萘基、吡咯基、呋喃基、噻吩基、咪唑基、吡唑基、噁唑基、异噁唑基、噻唑基、异噻唑基、三唑基、噁二唑基、噻二唑基、四唑基、吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基、四嗪基或环己基的二价基团,优选选自亚苯基、亚吡唑基或亚环己基。
在具体实施方案中,R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基、C 1-C 6烷氧基或C 1-C 6卤代烷氧基。
在具体实施方案中,R 1和R 5各自独立地选自H或卤素,优选为H、Cl或F。
在具体实施方案中,R 1和R 5各自独立地选自卤素,优选为Cl或F。
在具体实施方案中,R 2和R 4各自独立地选自C 1-C 6烷氧基,优选为甲氧基。
在具体实施方案中,R 3为H。
在具体实施方案中,R 7各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基,或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;优选R 7选自H、卤素、甲基、-N(CH 3)CH 2CH 2N(CH 3) 2、-N(CH 3) 2、4-甲基-哌嗪基或吗啉基,优选为H、卤素、甲基、-N(CH 3)CH 2CH 2N(CH 3) 2、-N(CH 3) 2或4-甲基-哌嗪基,优选为H、甲基、-N(CH 3)CH 2CH 2N(CH 3) 2或4-甲基-哌嗪基,优选为H或C 1-C 6烷基或-NR aR b,优选为甲基或-N(CH 3)CH 2CH 2N(CH 3) 2
在具体实施方案中,R 8选自C 2-C 6烯基或C 2-C 6炔基,优选选自乙烯基或丙炔基,优选为乙烯基。
在具体实施方案中,m为1。
在另一实施方案中,本发明提供了通式(IV)所示的化合物或其可药用的盐、互变异构体、外消旋 体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,其为通式(IV)所示的化合物:
Figure PCTCN2018104007-appb-000010
其中:
R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、-NR 11R 12、-C(O)R 13、-C(O)OR 13、-C(O)NR 11R 12、-NR 11C(O)R 13、-NR 11C(O)OR 13、-NR 11C(O)NR 11R 12、-OC(O)R 13、-OC(O)OR 13、-OC(O)NR 11R 12、-S(O) pR 13、-S(O) pOR 13、-S(O) pNR 11R 12、-OS(O) pR 13或-NR 11S(O) pR 13
R 7各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;
R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;
R 11、R 12和R 13各自独立地选自H、C 1-C 6烷基、C 1-C 6杂烷基、C 3-C 8环烷基、3-10元杂环基、5-10元杂芳基或6-14元芳基;
X选自-O-、-NH-或-CH 2-;Y选自-O-、-NH-或-CH 2-;条件是X和Y中至少有一个是CH 2
m为0、1、2、3或4;
p为1或2。
在具体实施方案中,R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基、C 1-C 6烷氧基或C 1-C 6卤代烷氧基。
在具体实施方案中,R 1和R 5各自独立地选自卤素,优选为Cl或F。
在具体实施方案中,R 2和R 4各自独立地选自C 1-C 6烷氧基,优选为甲氧基。
在具体实施方案中,R 3为H。
在具体实施方案中,R 7各自独立地选自H、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基,优选R 7选自H或C 1-C 6烷基或-NR aR b,优选为甲基或-N(CH 3)CH 2CH 2N(CH 3) 2
在具体实施方案中,R 8选自C 2-C 6烯基或C 2-C 6炔基,优选选自乙烯基或丙炔基。
在具体实施方案中,X选自-NH-,Y选自-CH 2-。
在具体实施方案中,X选自-CH 2-,Y选自-NH-。
在具体实施方案中,m为1。
在另一实施方案中,本发明提供了通式(V)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物:
Figure PCTCN2018104007-appb-000011
其中:
R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、-NR 11R 12、-C(O)R 13、-C(O)OR 13、-C(O)NR 11R 12、-NR 11C(O)R 13、-NR 11C(O)OR 13、-NR 11C(O)NR 11R 12、-OC(O)R 13、-OC(O)OR 13、-OC(O)NR 11R 12、-S(O) pR 13、-S(O) pOR 13、-S(O) pNR 11R 12、-OS(O) pR 13或-NR 11S(O) pR 13
R 7各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;
R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;
R 11、R 12和R 13各自独立地选自H、C 1-C 6烷基、C 1-C 6杂烷基、C 3-C 8环烷基、3-10元杂环基、5-10元杂芳基或6-14元芳基;
m为0、1或2;
p为1或2。
在具体实施方案中,R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基、C 1-C 6烷氧基或C 1-C 6卤代烷氧基。
在具体实施方案中,R 1和R 5各自独立地选自H或卤素,优选为H、Cl或F。
在具体实施方案中,R 1和R 5各自独立地选自卤素,优选为Cl或F。
在具体实施方案中,R 2和R 4各自独立地选自C 1-C 6烷氧基,优选为甲氧基。
在具体实施方案中,R 3为H。
在具体实施方案中,R 7各自独立地选自H、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基,优选R 7选自H或C 1-C 6烷基或-NR aR b,优选为甲基或-N(CH 3)CH 2CH 2N(CH 3) 2,优选为H或甲基。
在具体实施方案中,R 8选自C 2-C 6烯基或C 2-C 6炔基,优选选自乙烯基或丙炔基,优选为乙烯基。
在具体实施方案中,m为1。
在另一实施方案中,本发明提供了通式(VI)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,其为通式(VI)所示的化合 物:
Figure PCTCN2018104007-appb-000012
其中:
R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、-NR 11R 12、-C(O)R 13、-C(O)OR 13、-C(O)NR 11R 12、-NR 11C(O)R 13、-NR 11C(O)OR 13、-NR 11C(O)NR 11R 12、-OC(O)R 13、-OC(O)OR 13、-OC(O)NR 11R 12、-S(O) pR 13、-S(O) pOR 13、-S(O) pNR 11R 12、-OS(O) pR 13或-NR 11S(O) pR 13
R 7各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;
R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;
R 11、R 12和R 13各自独立地选自H、C 1-C 6烷基、C 1-C 6杂烷基、C 3-C 8环烷基、3-10元杂环基、5-10元杂芳基或6-14元芳基;
m为0、1、2、3或4;
p为1或2。
在具体实施方案中,R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基、C 1-C 6烷氧基或C 1-C 6卤代烷氧基。
在具体实施方案中,R 1和R 5各自独立地选自H或卤素,优选为H、Cl或F。
在具体实施方案中,R 1和R 5各自独立地选自卤素,优选为Cl或F。
在具体实施方案中,R 2和R 4各自独立地选自C 1-C 6烷氧基,优选为甲氧基。
在具体实施方案中,R 3为H。
在具体实施方案中,R 7各自独立地选自H、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基,或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;优选R 7选自H、卤素、甲基、-N(CH 3)CH 2CH 2N(CH 3) 2、-N(CH 3) 2、4-甲基-哌嗪基或吗啉基,优选为H、卤素、-N(CH 3)CH 2CH 2N(CH 3) 2、-N(CH 3) 2或4-甲基-哌嗪基,优选为H、-N(CH 3)CH 2CH 2N(CH 3) 2或4-甲基-哌嗪基,优选为H或C 1-C 6烷基或-NR aR b,优选为甲基或-N(CH 3)CH 2CH 2N(CH 3) 2
在具体实施方案中,R 8选自C 2-C 6烯基或C 2-C 6炔基,优选选自乙烯基或丙炔基,优选为乙烯基。
在具体实施方案中,m为1。
在另一实施方案中,本发明提供了通式(VII)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,其为通式(VI)所示的化合物:
Figure PCTCN2018104007-appb-000013
其中,
R 1和R 5各自独立地为卤素,优选地选自Cl或F;
R 2和R 4各自独立地为C 1-C 6烷氧基,优选地为-OCH 3
R 3为H;
R 7各自独立地选自卤素、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;优选地,R 7选自卤素、甲基、-N(CH 3) 2或吗啉基;
R 8为任选取代的C 2-C 6烯基;优选地,R 8为乙烯基;
m为0、1、2、3或4;优选地,m为0、1或2;优选地,m为1。
本发明的典型化合物包括,但并不限于:
Figure PCTCN2018104007-appb-000014
Figure PCTCN2018104007-appb-000015
Figure PCTCN2018104007-appb-000016
Figure PCTCN2018104007-appb-000017
Figure PCTCN2018104007-appb-000018
Figure PCTCN2018104007-appb-000019
Figure PCTCN2018104007-appb-000020
Figure PCTCN2018104007-appb-000021
Figure PCTCN2018104007-appb-000022
Figure PCTCN2018104007-appb-000023
Figure PCTCN2018104007-appb-000024
Figure PCTCN2018104007-appb-000025
基团
在本发明的通式(I)、(II)、(III)、(IV)、(V)、(VI)和(VII)中,各个通式中出现的基团R 1-R 8、R 11-R 13、X、Y、W和环A等如下定义,如果存在于所述通式中。同样,本发明还包括它们的定义之间互相组合的技术方案。
R 1
在本发明中,R 1选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、-NR 11R 12、-C(O)R 13、-C(O)OR 13、-C(O)NR 11R 12、-NR 11C(O)R 13、-NR 11C(O)OR 13、-NR 11C(O)NR 11R 12、-OC(O)R 13、-OC(O)OR 13、-OC(O)NR 11R 12、-S(O) pR 13、-S(O) pOR 13、-S(O) pNR 11R 12、-OS(O) pR 13或-NR 11S(O) pR 13,其中NR、R 12、R 13和p如说明书中所定义。优选地,R 1选自H、F、Cl、甲氧基或乙氧基。更优选地,R 1选自H、F或Cl。
R 2
在本发明中,R 2选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、-NR 11R 12、-C(O)R 13、-C(O)OR 13、-C(O)NR 11R 12、-NR 11C(O)R 13、-NR 11C(O)OR 13、 -NR 11C(O)NR 11R 12、-OC(O)R 13、-OC(O)OR 13、-OC(O)NR 11R 12、-S(O) pR 13、-S(O) pOR 13、-S(O) pNR 11R 12、-OS(O) pR 13或-NR 11S(O) pR 13,其中NR、R 12、R 13和p如说明书中所定义。优选地,R 2选自H、F、Cl、甲氧基或乙氧基。优选地,R 2选自H或甲氧基或乙氧基。
R 3
在本发明中,R 3选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、-NR 11R 12、-C(O)R 13、-C(O)OR 13、-C(O)NR 11R 12、-NR 11C(O)R 13、-NR 11C(O)OR 13、-NR 11C(O)NR 11R 12、-OC(O)R 13、-OC(O)OR 13、-OC(O)NR 11R 12、-S(O) pR 13、-S(O) pOR 13、-S(O) pNR 11R 12、-OS(O) pR 13或-NR 11S(O) pR 13,其中NR、R 12、R 13和p如说明书中所定义。优选地,R 3选自H、F、Cl、甲氧基或乙氧基。优选地,R 3选自H。
R 4
在本发明中,R 4选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、-NR 11R 12、-C(O)R 13、-C(O)OR 13、-C(O)NR 11R 12、-NR 11C(O)R 13、-NR 11C(O)OR 13、-NR 11C(O)NR 11R 12、-OC(O)R 13、-OC(O)OR 13、-OC(O)NR 11R 12、-S(O) pR 13、-S(O) pOR 13、-S(O) pNR 11R 12、-OS(O) pR 13或-NR 11S(O) pR 13,其中NR、R 12、R 13和p如说明书中所定义。优选地,R 4选自H、F、Cl、甲氧基或乙氧基。优选地,R 4选自H或甲氧基或乙氧基。
R 5
在本发明中,R 5选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、-NR 11R 12、-C(O)R 13、-C(O)OR 13、-C(O)NR 11R 12、-NR 11C(O)R 13、-NR 11C(O)OR 13、-NR 11C(O)NR 11R 12、-OC(O)R 13、-OC(O)OR 13、-OC(O)NR 11R 12、-S(O) pR 13、-S(O) pOR 13、-S(O) pNR 11R 12、-OS(O) pR 13或-NR 11S(O) pR 13,其中NR、R 12、R 13和p如说明书中所定义。优选地,R 5选自H、F、Cl、甲氧基或乙氧基。优选地,R 5选自H、F或Cl。
R 6
在本发明中,R 6选自H或-CH 2CH 2-。当R 6为-CH 2CH 2-时,其另一端连接至X,任选地-CH 2CH 2-中的一个亚甲基被-O-或-NH-替代。
R 7
在本发明中,R 7选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;R 13如说明书中所定义。优选地,R 7选自H或C 1-C 6烷基或-NR aR b。优选地,R 7为甲基或-N(CH 3)CH 2CH 2N(CH 3) 2
R 8
在本发明中,R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基,其中,任选的取代基如说明书中所定 义。优选地,R 8为乙烯基、乙炔基或丙炔基。
R 11
在本发明中,R 11选自H、C 1-C 6烷基、C 1-C 6杂烷基、C 3-C 8环烷基、3-10元杂环基、5-10元杂芳基或6-14元芳基。
R 12
在本发明中,R 12选自H、C 1-C 6烷基、C 1-C 6杂烷基、C 3-C 8环烷基、3-10元杂环基、5-10元杂芳基或6-14元芳基。
R 13
在本发明中,R 13选自H、C 1-C 6烷基、C 1-C 6杂烷基、C 3-C 8环烷基、3-10元杂环基、5-10元杂芳基或6-14元芳基。
环A
在本发明中,环A选自6-14元芳基、5-10元杂芳基、C 3-C 8环烷基或3-10元亚杂环基。优选地,环A为6-10元芳基、5-6元杂芳基。优选地,环A为6元芳基或5元杂芳基。优选地,环A选自苯基、萘基、吡咯基、呋喃基、噻吩基、咪唑基、吡唑基、噁唑基、异噁唑基、噻唑基、异噻唑基、三唑基、噁二唑基、噻二唑基、四唑基、吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基、四嗪基或环己基,优选选自苯基、吡唑基或环己基。
X和Y
在本发明中,X选自-O-、-NH-或-CH 2-,Y选自-O-、-NH-或-CH 2-;条件是X和Y中至少有一个是CH 2。优选地,X选自-O-,Y选自-CH 2-。优选地,X选自-NH-,Y选自-CH 2-。优选地,X选自-CH 2-,Y选自-CH 2-。优选地,Y选自-O-,X选自-CH 2-。优选地,Y选自-NH-,X选自-CH 2-。另外,当X与R 6连接成环时,此时X不可为-O-。
W
在本发明中,W选自化学键、-NH-或-CH 2-。
药物组合物、制剂和试剂盒
在另一方面,本发明提供了药物组合物,其包含本发明化合物(还称为“活性组分”)和药学上可接受的赋形剂。在一些实施方案中,所述药物组合物包含有效量的活性组分。在一些实施方案中,所述药物组合物包含治疗有效量的活性组分。在一些实施方案中,所述药物组合物包含预防有效量的活性组分。
用于本发明的药学上可接受的赋形剂是指不会破坏一起调配的化合物的药理学活性的无毒载剂、 佐剂或媒剂。可以用于本发明组合物中的药学上可接受的载剂、佐剂或媒剂包括(但不限于)离子交换剂、氧化铝、硬脂酸铝、卵磷脂、血清蛋白(如人类血清白蛋白)、缓冲物质(如磷酸盐)、甘氨酸、山梨酸、山梨酸钾、饱和植物脂肪酸的偏甘油酯混合物、水、盐或电解质(如硫酸鱼精蛋白)、磷酸氢二钠、磷酸氢钾、氯化钠、锌盐、硅胶、三硅酸镁、聚乙烯吡咯烷酮、基于纤维素的物质、聚乙二醇、羧甲基纤维素钠、聚丙烯酸酯、蜡、聚乙烯-聚氧丙烯-嵌段聚合物、聚乙二醇以及羊毛脂。
本发明还包括试剂盒(例如,药物包装)。所提供的试剂盒可以包括本发明化合物、其它治疗剂,以及含有本发明化合物、其它治疗剂的第一和第二容器(例如,小瓶、安瓿瓶、瓶、注射器和/或可分散包装或其它合适的容器)。在一些实施方案中,提供的试剂盒还可以任选包括第三容器,其含有用于稀释或悬浮本发明化合物和/或其它治疗剂的药用赋形剂。在一些实施方案中,提供在第一容器和第二容器中的本发明化合物和其它治疗剂组合形成一个单位剂型。
给药
本发明提供的药物组合物可以通过许多途径给药,包括但不限于:口服给药、肠胃外给药、吸入给药、局部给药、直肠给药、鼻腔给药、口腔给药、阴道给药、通过植入剂给药或其它给药方式。例如,本文使用的肠胃外给药包括皮下给药、皮内给药、静脉内给药、肌肉内给药、关节内给药、动脉内给药、滑膜腔内给药、胸骨内给药、脑脊髓膜内给药、病灶内给药、和颅内的注射或输液技术。
通常,给予有效量的本文所提供的化合物。按照有关情况,包括所治疗的病症、选择的给药途径、实际给予的化合物、个体患者的年龄、体重和响应、患者症状的严重程度,等等,可以由医生确定实际上给予的化合物的量。
当用于预防本发明所述病症时,给予处于形成所述病症危险之中的受试者本文所提供的化合物,典型地基于医生的建议并在医生监督下给药,剂量水平如上所述。处于形成具体病症的危险之中的受试者,通常包括具有所述病症的家族史的受试者,或通过遗传试验或筛选确定尤其对形成所述病症敏感的那些受试者。
还可以长期给予本文所提供的药物组合物(“长期给药”)。长期给药是指在长时间内给予化合物或其药物组合物,例如,3个月、6个月、1年、2年、3年、5年等等,或者可无限期地持续给药,例如,受试者的余生。在一些实施方案中,长期给药意欲在长时间内在血液中提供所述化合物的恒定水平,例如,在治疗窗内。
可以使用各种给药方法,进一步递送本发明的药物组合物。例如,在一些实施方案中,可以推注给药药物组合物,例如,为了使化合物在血液中的浓度提高至有效水平。推注剂量取决于通过身体的活性组分的目标全身性水平,例如,肌内或皮下的推注剂量使活性组分缓慢释放,而直接递送至静脉的推注(例如,通过IV静脉滴注)能够更加快速地递送,使得活性组分在血液中的浓度快速升高至有效水平。在其它实施方案中,可以以持续输液形式给予药物组合物,例如,通过IV静脉滴注,从而在受试者身体中提供稳态浓度的活性组分。此外,在其它实施方案中,可以首先给予推注剂量的药物组合物,而后持续输液。
口服组合物可以采用散装液体溶液或混悬剂或散装粉剂形式。然而,更通常,为了便于精确地剂 量给药,以单位剂量形式提供所述组合物。术语“单位剂型”是指适合作为人类患者及其它哺乳动物的单元剂量的物理离散单位,每个单位包含预定数量的、适于产生所需要的治疗效果的活性物质与合适药学赋形剂。典型的单位剂量形式包括液体组合物的预装填的、预先测量的安瓿或注射器,或者在固体组合物情况下的丸剂、片剂、胶囊剂等。在这种组合物中,所述化合物通常为较少的组分(约0.1至约50重量%,或优选约1至约40重量%),剩余部分为对于形成所需给药形式有用的各种载体或赋形剂以及加工助剂。
对于口服剂量,代表性的方案是,每天一个至五个口服剂量,尤其是两个至四个口服剂量,典型地是三个口服剂量。使用这些剂量给药模式,每个剂量提供大约0.01至大约20mg/kg的本发明化合物,优选的剂量各自提供大约0.1至大约10mg/kg,尤其是大约1至大约5mg/kg。
为了提供与使用注射剂量类似的血液水平,或比使用注射剂量更低的血液水平,通常选择透皮剂量,数量为大约0.01至大约20%重量,优选大约0.1至大约20%重量,优选大约0.1至大约10%重量,且更优选大约0.5至大约15%重量。
从大约1至大约120小时,尤其是24至96小时,注射剂量水平在大约0.1mg/kg/小时至至少10mg/kg/小时的范围。为了获得足够的稳定状态水平,还可以给予大约0.1mg/kg至大约10mg/kg或更多的预载推注。对于40至80kg的人类患者来说,最大总剂量不能超过大约2g/天。
适于口服给药的液体形式可包括合适的水性或非水载体以及缓冲剂、悬浮剂和分散剂、着色剂、调味剂,等等。固体形式可包括,例如,任何下列组份,或具有类似性质的化合物:粘合剂,例如,微晶纤维素、黄蓍胶或明胶;赋形剂,例如,淀粉或乳糖,崩解剂,例如,褐藻酸、Primogel或玉米淀粉;润滑剂,例如,硬脂酸镁;助流剂,例如,胶体二氧化硅;甜味剂,例如,蔗糖或糖精;或调味剂,例如,薄荷、水杨酸甲酯或橙味调味剂。
可注射的组合物典型地基于可注射用的无菌盐水或磷酸盐缓冲盐水,或本领域中已知的其它可注射的赋形剂。如前所述,在这种组合物中,活性化合物典型地为较少的组分,经常为约0.05至10%重量,剩余部分为可注射的赋形剂等。
典型地将透皮组合物配制为含有活性组分的局部软膏剂或乳膏剂。当配制为软膏剂时,活性组分典型地与石蜡或可与水混溶的软膏基质组合。或者,活性组分可与例如水包油型乳膏基质一起配制为乳膏剂。这种透皮制剂是本领域中公知的,且通常包括用于提升活性组分或制剂的稳定的皮肤渗透的其它组份。所有这种已知的透皮制剂和组份包括在本发明提供的范围内。
本发明化合物还可通过经皮装置给予。因此,经皮给药可使用贮存器(reservoir)或多孔膜类型、或者多种固体基质的贴剂实现。
用于口服给予、注射或局部给予的组合物的上述组份仅仅是代表性的。其它材料以及加工技术等阐述于Remington's Pharmaceutical Sciences,17th edition,1985,Mack Publishing Company,Easton,Pennsylvania的第8部分中,本文以引用的方式引入该文献。
本发明化合物还可以以持续释放形式给予,或从持续释放给药系统中给予。代表性的持续释放材料的描述可在Remington's Pharmaceutical Sciences中找到。
本发明还涉及本发明化合物的药学上可接受的制剂。在一个实施方案中,所述制剂包含水。在另 一个实施方案中,所述制剂包含环糊精衍生物。最常见的环糊精为分别由6、7和8个α-1,4-连接的葡萄糖单元组成的α-、β-和γ-环糊精,其在连接的糖部分上任选包括一个或多个取代基,其包括但不限于:甲基化的、羟基烷基化的、酰化的和磺烷基醚取代。在一些实施方案中,所述环糊精为磺烷基醚β-环糊精,例如,磺丁基醚β-环糊精,也称作Captisol。参见,例如,U.S.5,376,645。在一些实施方案中,所述制剂包括六丙基-β-环糊精(例如,在水中,10-50%)。
组合
本发明化合物或其组合物可以与其它治疗剂组合给药,以治疗所述疾病。已知治疗剂的实例包括但不限于阿德力霉素(Adriamycin)、地塞米松(dexamethasone)、长春新碱(vincristine)、环磷酰胺(cyclophosphamide)、氟尿嘧啶(fluorouracil)、拓朴替康(topotecan)、紫杉酚(taxol)、干扰素、铂衍生物、紫杉烷(taxane)(例如太平洋紫杉醇(paclitaxel))、长春花生物碱(例如长春碱(vinblastine))、蒽环霉素(anthracycline)(例如多柔比星(doxorubicin))、表鬼臼毒素(epipodophyllotoxin)(例如依托泊苷(etoposide))、顺铂(cisplatin)、mTOR抑制剂(例如雷帕霉素(rapamycin))、甲氨蝶呤(methotrexate)、放线菌素D(actinomycin D)、海兔毒素10(dolastatin 10)、秋水仙碱(colchicine)、吐根素(emetine)、三甲曲沙(trimetrexate)、氯苯氨啶(metoprine)、环孢霉素(cyclosporine)、道诺霉素(daunorubicin)、替尼泊苷(teniposide)、两性霉素(amphotericin)、烷化剂(例如苯丁酸氮芥(chlorambucil))、5-氟尿嘧啶、喜树碱(camptothecin)、顺铂、甲硝哒唑(metronidazole)以及Gleevec TM。在其它实施例中,本发明化合物与如阿瓦斯汀(Avastin)或维克替比(VECTIBIX)等生物剂组合给药。
在一些实施方案中,本发明化合物或其组合物可以与选自以下中的任一种或一种以上的抗增殖剂或化学治疗剂组合给药:阿巴瑞克(abarelix)、阿地白介素(aldesleukin)、阿仑单抗(alemtuzumab)、阿利维甲酸(alitretinoin)、别嘌呤醇(allopurinol)、六甲蜜胺(altretamine)、氨磷汀(amifostine)、阿那曲唑(anastrozole)、三氧化二砷、天冬酰胺酶、阿扎胞苷(azacitidine)、BCG Live、贝伐单抗(bevacuzimab)、氟尿嘧啶、贝瑟罗汀(bexarotene)、博莱霉素(bleomycin)、硼替佐米(bortezomib)、白消安(busulfan)、二甲睾酮(calusterone)、卡培他滨(capecitabine)、喜树碱、卡铂(carboplatin)、卡莫司汀(carmustine)、塞内昔布(celecoxib)、西妥昔单抗(cetuximab)、苯丁酸氮芥(chlorambucil)、克拉屈滨(cladribine)、氯法拉滨(clofarabine)、环磷酰胺、阿糖胞苷(cytarabine)、放线菌素D、达贝泊汀α(darbepoetin alfa)、道诺霉素、地尼白介素(denileukin)、右雷佐生(dexrazoxane)、多烯紫杉醇(docetaxel)、多柔比星、盐酸多柔比星、丙酸屈他雄酮(dromostanolone propionate)、表柔比星(epirubicin)、依伯汀α(epoetin alfa)、埃罗替尼(erlotinib)、雌莫司汀(estramustine)、磷酸依托泊苷、依托泊苷、依西美坦(exemestane)、非格司亭(filgrastim)、氟尿苷(floxuridine)、氟达拉宾(fludarabine)、氟维司群(fulvestrant)、吉非替尼、吉西他滨(gemcitabine)、吉妥珠单抗(gemtuzumab)、乙酸戈舍瑞林(goserelin acetate)、乙酸组氨瑞林(histrelin acetate)、羟基脲(hydroxyurea)、替伊莫单抗(ibritumomab)、艾达霉素(idarubicin)、异环磷酰胺(ifosfamide)、甲磺酸伊马替尼(imatinib mesylate)、干扰素α-2a、干扰素α-2b、伊立替康(irinotecan)、来那度胺(lenalidomide)、来曲唑(letrozole)、甲酰四氢叶酸(leucovorin)、乙酸亮丙立德(leuprolide acetate)、左旋咪唑(levamisole)、洛莫司汀(lomustine)、乙酸甲地孕酮(megestrol acetate)、美法仑 (melphalan)、巯嘌呤(mercaptopurine)、6-MP、巯乙磺酸钠(mesna)、甲氨蝶呤、补骨脂素(methoxsalen)、丝裂霉素C(mitomycin C)、米托坦(mitotane)、米托蒽醌(mitoxantrone)、诺龙(nandrolone)、奈拉滨(nelarabine)、诺非单抗(nofetumomab)、奥普瑞白介素(oprelvekin)、奥沙利铂(oxaliplatin)、太平洋紫杉醇、帕利夫明(palifermin)、帕米膦酸盐(pamidronate)、培加酶(pegademase)、培门冬酶(pegaspargase)、乙二醇化非格司亭(pegfilgrastim)、培美曲塞二钠(pemetrexed disodium)、喷司他汀(pentostatin)、哌泊溴烷(pipobroman)、普卡霉素(plicamycin)、卟吩姆钠(porfimer sodium)、丙卡巴肼(procarbazine)、奎纳克林(quinacrine)、拉布立酶(rasburicase)、利妥昔单抗(rituximab)、沙格司亭(sargramostim)、索拉非尼(sorafenib)、链脲霉素(streptozocin)、顺丁烯二酸舒尼替尼(sunitinib maleate)、滑石、他莫西芬(tamoxifen)、替莫唑胺(temozolomide)、替尼泊苷、VM-26、睾内酯(testolactone)、硫鸟嘌呤(thioguanine)、6-TG、噻替派(thiotepa)、拓朴替康、托瑞米芬(toremifene)、托西莫单抗(tositumomab)、曲妥珠单抗(trastuzumab)、维甲酸(tretinoin)、ATRA、尿嘧啶氮芥(uracil mustard)、伐柔比星(valrubicin)、长春碱、长春新碱(vincristine)、长春瑞滨(vinorelbine)、唑来膦酸盐(zoledronate)或唑来膦酸(zoledronic acid)。
本发明化合物还可以组合的治疗剂的其它实例包括但不限于:用于阿尔茨海默氏病(Alzheimer′s Disease)的治疗剂,如盐酸多奈哌齐(donepezil hydrochloride)和雷斯替明(rivastigmine);用于帕金森氏病(Parkinson′s Disease)的治疗剂,如L-DOPA/卡比多巴(carbidopa)、恩他卡朋(entacapone)、罗匹尼洛(ropinrole)、普拉克索(pramipexole)、溴麦角环肽(bromocriptine)、培高利特(pergolide)、三己芬迪(trihexephendyl)以及三环癸胺(amantadine);用于多发性硬化症(Multiple Sclerosis;MS)的治疗剂,如β干扰素、乙酸格拉默(glatiramer acetate)和米托蒽醌;用于哮喘的治疗剂,如舒喘宁(albuterol)和孟鲁司特;用于精神分裂症的治疗剂,如再普乐(zyprexa)、理斯必妥(risperdal)、思瑞康(seroquel)以及氟哌啶醇(haloperidol);消炎剂,如皮质类固醇、TNF阻断剂、IL-1RA、硫唑嘌呤(azathioprine)、环磷酰胺以及柳氮磺胺吡啶(sulfasalazine);免疫调节剂和免疫抑制剂,如环孢素(cyclosporin)、他克莫司(tacrolimus)、雷帕霉素、霉酚酸吗啉乙酯(mycophenolate mofetil)、干扰素、皮质类固醇、环磷酰胺、硫唑嘌呤以及柳氮磺胺吡啶;神经营养因子,如乙酰胆碱酯酶抑制剂、MAO抑制剂、干扰素、抗惊厥剂、离子通道阻断剂、利鲁唑(riluzole)以及抗帕金森氏病剂;用于心血管疾病的治疗剂,如β阻断剂、ACE抑制剂、利尿剂、硝酸盐、钙离子通道阻断剂以及他汀类(statin);用于肝病的治疗剂,如皮质类固醇、消胆胺(cholestyramine)、干扰素以及抗病毒剂;用于血液病症的治疗剂,如皮质类固醇、抗白血病剂以及生长因子;以及用于免疫缺陷病症的治疗剂,如γ球蛋白。
那些其它活性剂可以与含有本发明化合物的组合物分开给药,作为多次给药方案的一部分。或者,那些活性剂可以是单一剂型的一部分,与本发明化合物一起混合在单一组合物中。如果作为多次给药方案的一部分给药,那么两种活性剂可以同时、依次或彼此间隔一段时间(通常彼此间隔在5小时以内)提供。
适应症和疾病
FGFR-4在肝细胞癌(HCC)进展期间调控增殖、存活和α-胎蛋白分泌;FGFR-4抑制剂因此是用于此未满足医疗需要的有希望的潜在治疗剂(Ho等人,Journal of Hepatology,2009,50:118-27)。HCC 每年折磨全世界超过550,000人且是任何癌症类型中1年存活率最差者之一。
经由FGF19(纤维母细胞生长因子(FGF)家族成员,其由激素组成)参与调控血糖、血脂和能量稳态显示了FGFR-4与HCC间的联系的其它证据。已在FGF19基因转殖小鼠中观察到增加的肝细胞增殖和肝肿瘤形成。FGF19活化FGFR-4(其在肝中的主要受体),且认为FGFR-4活化是FGF19能够增加肝细胞增殖并诱导肝细胞癌形成的机制(Wu等人,J Biol Chem(2010)285(8):5165-5170)。FGF19还已被他人识别为HCC中的驱动基因(Sawey等人,Cancer Cell(2011)19:347-358)。因此,认为本文所公开化合物(其是FGFR-4的潜在和选择性抑制剂)可用于治疗HCC和其它肝癌。
肿瘤基因组筛查已在人类乳腺癌细胞系MDA-MB-453中鉴定出活化纤维母细胞生长因子受体4(FGFR-4)Y367C突变。因此,其已表明FGFR-4可能为乳腺癌中肿瘤生长的驱动物(Roidl等人,Oncogene(2010)29(10):1543-1552)。因此,认为本文所公开化合物(其为FGFR-4的强效选择性抑制剂)可用于治疗FGFR-4调节的乳腺癌。
FGFR-4上游基因的分子变化(例如,易位)会导致FGFR-4活化/过表达。例如,PAX3-FKHR易位/基因融合会导致FGFR-4过表达。因此机制所致的FGFR-4过表达与横纹肌肉瘤(RMS)相关联(Cao等人,Cancer Res(2010)70(16):6497-6508)。FGFR-4自身中的突变(例如,激酶结构域突变)会导致蛋白质过活化;此机制已与RMS亚族群相关(Taylor等人,J Clin Invest(2009)119:3395-3407)。因此,认为本文所公开化合物(其为FGFR-4的强效选择性抑制剂)可用于治疗FGFR-4调节的RMS和其它肉瘤。
其它疾病与FGFR-4上游基因的变化或与FGFR-4自身中的突变相关联。例如,FGFR-4的激酶结构域中的突变导致过活化,其与肺腺癌相关(Ding等人,Nature(2008)455(7216):1069-1075)。FGFR-4的扩增与诸如肾细胞癌的病状相关(TCGA暂行资料)。另外,使FGFR4沉默且抑制配体-受体结合显著减慢卵巢肿瘤生长,从而表明FGFR4的抑制剂可用于治疗卵巢癌(Zaid等人,Clin.Cancer Res.(2013)809)。
胆汁酸水平的致病性升高与FGF19水平变化相关(Vergnes等人,Cell Metabolism(2013)17,916-28)。因此,FGF19的水平降低在促进胆汁酸的合成中且因此在高脂血症的治疗中可具有益处。
因此,本发明化合物可以用于治疗多种与FGFR有关的疾病,其中所述疾病包括但不限于:胃癌、甲状腺癌、前列腺癌、乳腺癌、肉瘤(例如横纹肌肉瘤)、皮肤癌(例如黑色素瘤)、肝癌(例如肝细胞癌和胆管上皮癌)、胰腺癌(例如胰腺上皮内瘤样病变和胰腺导管腺癌)、肺癌(例如非小细胞肺癌和肺腺癌)、肾癌(例如肾细胞癌)、结直肠癌和卵巢癌。
实施例
本发明通式(I)所述的化合物或其可药用的盐的制备,可通过以下实施例中所述的示例性方法以及本领域技术人员所用的相关公开文献操作完成,但这些实施例并非限制着本发明的范围。
化合物的结构是通过核磁共振(NMR)或质谱(MS)来确定的。NMR的测定是用Bruker AVANCE-400或Varian Oxford-300核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6)、氘代氯仿(CDC1 3)、氘代甲醇(CD 3OD),内标为四甲基硅烷(TMS),化学位移是以10 -6(ppm)作为单位给出。
MS的测定用Agilent SQD(ESI)质谱仪(生产商:Agilent,型号:6110)或Shimadzu SQD(ESI)质谱仪(生产商:Shimadzu,型号:2020)。
HPLC的测定使用安捷伦1200DAD高压液相色谱仪(Sunfire C18,150×4.6mm,5μm,色谱柱)和Waters 2695-2996高压液相色谱仪(Gimini C18 150×4.6mm,5μm色谱柱)。
薄层层析硅胶板使用青岛海洋GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm~0.2mm,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm硅胶板。
柱层析一般使用青岛海洋200~300目硅胶为载体。
本发明的已知的起始原料可以采用或按照本领域已知的方法来合成,或可购买自ABCR GmbH&Co.KG,Acros Organics,Aldrich Chemical Company,韶远化学科技(Accela ChemBio Inc)、北京耦合化学品等公司。
实施例中如无特殊说明,反应均在氩气氛或氮气氛下进行。
氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
氢气氛是指反应瓶连接一个约1L容积的氢气气球。
加压氢化反应使用北京佳维科创科技有限公司GCD-500G高纯氢气发生器和BLT-2000中压氢化仪。
氢化反应通常抽真空,充入氢气,反复操作3次。
微波反应使用CEM Discover-SP型微波反应器。
实施例中如无特殊说明,反应的温度为室温,温度范围是20℃-30℃。
实施例中的反应进程的监测采用薄层色谱法(TLC),反应所使用的展开剂的体系有A:二氯甲烷和甲醇体系;B:石油醚和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节。
纯化化合物采用的柱层析的洗脱剂的体系和薄层色谱法的展开剂的体系包括A:二氯甲烷和甲醇体系;B:石油醚和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的酸性或碱性试剂(如三乙胺)进行调节。
实施例10
N-(2-((6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)氨基)-3-甲基苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000026
参照实例093b的操作步骤合成实例10a
第一步
2-(4-(3,5-二甲氧苯基)-2-羰基环己基)-2-羰基乙酸乙基酯10b
将二异丙基氨基锂于-78℃缓慢滴加到化合物3-(3,5-二甲氧苯基)环己烷-1-酮10a(9.5g,40.6mmol,1eq)的无水四氢呋喃溶液中,在-78℃搅拌半个小时。然后将草酸二乙酯(8.9g,60.9mmol,1.5eq)滴加到反应液中,升至室温,搅拌2小时。后处理:加10mL氯化铵水溶液淬灭反应,分层有机相减压脱溶,残余物经硅胶柱层析纯化(石油醚:乙酸乙酯=5:1),得到目标产物2-(4-(3,5-二甲氧苯基)-2-羰基环己基)-2-羰基乙酸乙基酯10b(7.7g,黄色液体),产率:57%。
MS m/z(ESI):335[M+1]。
第二步
6-(3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-羧酸乙基酯10c
将化合物2-(4-(3,5-二甲氧苯基)-2-羰基环己基)-2-羰基乙酸乙基酯10b(7.7g,23.1mmol,1eq),水合 肼(1.6g,1.1eq)溶于乙醇/醋酸10:1(90mL)中,在65℃下反应3小时。后处理:减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=2:1)纯化,得目标产物6-(3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-羧酸乙基酯10c(3.5g,黄色液体),产率:46%。
MS m/z(ESI):331[M+1]。
第三步
6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-羧酸乙基酯10d
将化合物6-(3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-羧酸乙基酯10c(800mg,2.4mmol,1eq),氯化钾(359mg,4.8mmol,2eq)溶于二氧六环/水3:1(40mL)中,在0℃条件下缓慢滴加oxane(3.0g,4.8mmol,2eq),继续搅拌3h。后处理:加50mL碳酸氢钠水溶液稀释,用乙酸乙酯萃取,有机相减压脱溶残余物经硅胶柱层析(石油醚:乙酸乙酯=2:1)纯化,得目标产物6-(3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-羧酸乙基酯10d(700mg,黄色液体),产率:72%。
MS m/z(ESI):399[M+1]。
第四步
6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-4,5,6,7-四氢-1H-吲唑-3-羧酸乙基酯10e
将化合物6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-羧酸乙基酯10d(2.5g,6.3mmol,1eq)溶于DMF(40mL)中,在0℃条件下缓慢滴加氢化钠(0.38g,9.5mmol,1.5eq),搅拌0.5h,加入对甲氧基氯苄(1.5g,9.5mmol,1.5eq),继续搅拌3h。后处理:加50mL水淬灭反应,用乙酸乙酯萃取,有机相减压脱溶残余物经硅胶柱层析(石油醚:乙酸乙酯=3:1)纯化,得目标产物6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-4,5,6,7-四氢-1H-吲唑-3-羧酸乙基酯10e(1.2g,白色固体)。
MS m/z(ESI):519[M+1]。
第五步
6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-4,5,6,7-四氢-1H-吲唑-3-羧酸10f
将化合物6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-4,5,6,7-四氢-1H-吲唑-3-羧酸乙基酯10e(2.0g,3.9mmol,1eq)溶于甲醇/四氢呋喃/水=10:10:3(23mL)中,在0℃条件下加氢氧化钠(0.46g,11.6mmol,3eq),升到50℃搅拌5h。后处理:有机相减压脱溶,加1N的稀盐酸50mL,用乙酸乙酯萃取,旋干残余物经硅胶柱层析(石油醚:乙酸乙酯=1:1)纯化,得目标产物6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-4,5,6,7-四氢-1H-吲唑-3-羧酸10f(1.1g,白色固体)。产率:58%。
MS m/z(ESI):491[M+1]。
第六步
(6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-4,5,6,7-四氢-1H-吲唑-3-基)氨基甲酸叔-丁基酯10g
将化合物6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-4,5,6,7-四氢-1H-吲唑-3-羧酸10f(1.1g,2.2mmol,1eq),叠氮磷酸二苯酯(935mg,3.4mmol,1.5eq),三乙胺(0.66g,6.6mmol,3eq)溶于叔丁醇 (20mL)中,氮气保护下回流反应4h。后处理:有机相减压脱溶,旋干残余物经硅胶柱层析(石油醚:乙酸乙酯=10:1)纯化,得目标产物(6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-4,5,6,7-四氢-1H-吲唑-3-基)氨基甲酸叔-丁基酯10g(0.6g,白色固体)。产率:50%。
MS m/z(ESI):562[M+1]。
第七步
6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-4,5,6,7-四氢-1氢-吲唑-3-胺10h
将化合物(6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-4,5,6,7-四氢-1H-吲唑-3-基)氨基甲酸叔-丁基酯10g(600mg,1.1mmol,1eq)溶于无水二氯甲烷(15mL)中,加入三氟乙酸(3mL),室温反应2h。后处理:有机相减压脱溶,加饱和碳酸氢钠水溶液(50mL),用乙酸乙酯萃取,旋干得目标产物6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-4,5,6,7-四氢-1H-吲唑-3-胺10h(0.3g,白色固体)。产率:60%。
MS m/z(ESI):342[M+1]。
第八步
6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-氮-(2-甲基-6-硝基苯基)-4,5,6,7-四氢-1氢-吲唑-3-胺10i
将化合物6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-4,5,6,7-四氢-1H-吲唑-3-胺10h(300mg,0.65mmol,1eq),2-溴-1-甲基-3-硝基苯(280mg,1.30mmol,2eq),三(二亚苄基丙酮)二钯(119mg,0.13mmol,0.2eq),2-二环己基磷-2,4,6-三异丙基联苯(124mg,0.26mmol,0.4eq),碳酸铯(424mg,1.32mmol,2eq)溶于无水DMF(10mL)中,在氮气保护条件下于90℃加热4h。后处理:有机相减压脱溶,加水溶液(20mL),用乙酸乙酯萃取,旋干残余物经硅胶柱层析(石油醚:乙酸乙酯=5:1)纯化,得目标产物6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-N-(2-甲基-6-硝基苯基)-4,5,6,7-四氢-1H-吲唑-3-胺10i(0.13g,黄色固体)。产率:34%。
MS m/z(ESI):597[M+1]。
第九步
6-(2,6-二氯-3,5-二甲氧苯基)-N-(2-甲基-6-硝基苯基)-4,5,6,7-四氢-1H-吲唑-3-胺10j
将化合物6-(2,6-二氯-3,5-二甲氧苯基)-1-(4-甲氧苄基)-N-(2-甲基-6-硝基苯基)-4,5,6,7-四氢-1H-吲唑-3-胺10i(130mg,0.22mmol,1eq)溶于无水二氯甲烷(30mL)中,加入三氟甲磺酸酐(0.5mL),室温反应30min。后处理:加饱和碳酸氢钠水溶液(20mL),用二氯甲烷萃取,旋干得目标产物6-(2,6-二氯-3,5-二甲氧苯基)-N-(2-甲基-6-硝基苯基)-4,5,6,7-四氢-1H-吲唑-3-胺10j(90mg,黄色固体)。产率:86%。
MS m/z(ESI):477[M+1]。
第十步
N1-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-6-甲基苯-1,2-二胺10k
将化合物6-(2,6-二氯-3,5-二甲氧苯基)-N-(2-甲基-6-硝基苯基)-4,5,6,7-四氢-1H-吲唑-3-胺10j(90mg, 0.19mmol,1eq)溶于乙醇(15mL)中,加入锌粉(123mg,1.9mmol,10eq)和氯化铵(218mg,1.9mmol,10eq),50℃反应3h。后处理:旋干溶剂,用乙酸乙酯稀释,水洗,旋干得目标产物N1-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-6-甲基苯-1,2-二胺10k(60mg,黄色固体)。产率:70%。
MS m/z(ESI):447[M+1]。
第十一步
N-(2-((6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)氨基)-3-甲基苯基)丙烯酰胺10
将化合物N1-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-6-甲基苯-1,2-二胺10k(40mg,0.09mmol,1eq)和N,N-二异丙基乙胺(23mg,0.18mmol,1eq)溶于无水二氯甲烷(10mL)中,降温至-40℃,缓慢加入丙烯酰氯(8mg,0.09mmol,1eq)的二氯甲烷溶液,反应0.5h。后处理:旋干溶剂,用制备液相分离,冻干得目标产物N-(2-((6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)氨基)-3-甲基苯基)丙烯酰胺10(5.8mg,白色固体)。产率:14%。
MS m/z(ESI):501[M+1]。
1H NMR(400MHz,DMSO)δ11.77(s,1H),9.67(s,1H),7.58(d,J=6.1Hz,1H),7.12–6.97(m,2H),6.88(d,J=10.6Hz,1H),6.46(dd,J=17.0,10.2Hz,1H),6.21(dd,J=17.0,1.7Hz,1H),5.73(dd,J=10.2,1.7Hz,1H),3.90(dd,J=18.3,3.2Hz,7H),3.32–3.20(m,2H),2.57(d,J=5.2Hz,1H),2.34–2.24(m,1H),2.16(s,3H),1.70(d,J=9.9Hz,1H)。
实施例15
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000027
参照实例093d的操作步骤合成实例15a。
第一步
6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑15b
将6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑15a(250mg,0.66mmol,1eq)加到5ml 乙酸中,加入NCS(184mg,1.39mmol,2.1eq),50℃反应3h。后处理:减压脱溶,残余物用石油醚/乙酸乙酯(2:1)体系过硅胶层析柱得到黄色固体产物6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑15b(200mg,黄色固体)。产率:68%。
MS m/z(ESI):448[M+1]。
第二步
2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺15c
将化合物6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑15b(200mg,0.45mmol,1eq)溶于乙醇(10mL)中,加入锌粉(290mg,4.5mmol,10eq)和氯化铵(517mg,4.5mmol,10eq),50℃反应3h。后处理:旋干溶剂,用乙酸乙酯稀释,水洗,有机相旋干得目标产物2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺15c(131mg,黄色固体)。产率:70%。
MS m/z(ESI):418[M+1]。
第三步
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺15
将化合物2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺15c(120mg,0.29mmol,1eq)和N,N-二异丙基乙胺(75mg,0.58mmol,1eq)溶于无水二氯甲烷(10mL)中,降温至-40℃,缓慢加入丙烯酰氯(26mg,0.29mmol,1eq)的二氯甲烷溶液,反应0.5h。后处理:旋干溶剂,用制备液相分离,冻干得目标产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺15(5mg,白色固体)。产率:4%。
MS m/z(ESI):501[M+1]。
1H NMR(400MHz,DMSO)δ12.98(s,1H),11.48(s,1H),8.49(d,J=6.9Hz,1H),7.59(d,J=7.0Hz,1H),7.33(t,J=7.6Hz,1H),7.18(t,J=7.1Hz,1H),6.90(s,1H),6.43–6.20(m,2H),5.82(d,J=9.2Hz,1H),4.03(s,1H),3.93(d,J=3.6Hz,6H),3.42(d,J=12.8Hz,1H),2.86–2.57(m,4H),1.84(d,J=11.6Hz,1H)。
实施例27
Figure PCTCN2018104007-appb-000028
第一步
6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-羧酸27b
将化合物6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-羧酸乙基酯27a(700mg,1.76mmol,1eq)溶于甲醇/四氢呋喃/水=5:5:2(12mL)中,在0℃条件下加氢氧化钠(0.21g,5.28mmol,3eq),升到50℃搅拌5h。后处理:有机相减压脱溶,加1N的稀盐酸15mL,固体过滤干燥,得目标产物6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-羧酸27b(0.52g,白色固体)。产率:70%。
MS m/z(ESI):371[M+1]。
第二步
6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-胺27c
将化合物6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-羧酸27b(2.5g,6.8mmol,1eq),叠氮磷酸二苯酯(2.3g,8.1mmol,1.2eq),三乙胺(1.4g,13.6mmol,2eq)溶于乙腈(40mL)中,氮气保护下回流反应4h。后处理:有机相减压脱溶,旋干残余物经硅胶柱层析(二氯甲烷:甲醇=10:1)纯化,得目标产物6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-胺27c(0.3g,淡黄色固体)。产率:13%。
MS m/z(ESI):342[M+1]。
第三步
N-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)丙烯酰胺27
将化合物6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-胺27c(10mg,0.03mmol,1eq)溶于5mL二氯甲烷,加入N,N-二异丙基乙胺(8mg,0.06mmol,2eq),降温至-40℃,滴加丙烯酰氯(8mg,0.06mmol,1eq)的二氯甲烷溶液,搅拌30min。后处理:减压脱溶,用制备液相分离,冻干得到目标产物N-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)丙烯酰胺27(3mg,白色固体)。产率:25%。
MS m/z(ESI):396[M+1]。
1H NMR(400MHz,MeOD)δ8.53(s,1H),7.42(dd,J=17.3,10.5Hz,1H),6.78(s,1H),6.60(dd,J=17.3,1.7Hz,1H),6.01(dd,J=10.5,1.7Hz,1H),4.12–4.00(m,1H),3.95(s,6H),3.44(dd,J=16.6,12.7Hz,1H),2.74(ddd,J=25.2,12.6,5.3Hz,1H),2.65–2.55(m,2H),2.37(ddd,J=15.3,12.4,5.5Hz,1H),1.86(d,J=9.2Hz,1H)。
实施例083
N-((6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)甲基)丙烯酰胺
Figure PCTCN2018104007-appb-000029
参照实例010d和027b步骤合成前四步。
第五步
6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-甲酰胺083f
将6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-羧酸083e(3.92g,10.57mmol,1eq)溶于40mL二氯甲烷中,控温0℃加入草酰氯(2.68g,21.13mmol,2eq),加入一滴无水N,N-二甲基甲酰胺催化,于室温搅拌反应2小时,然后向体系中通30分钟氨气。后处理:减压脱溶,剩余物通过硅胶柱层析纯化(二氯甲烷/甲醇=5:1)纯化,得到6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-甲酰胺083f(3.19g,黄色固体)产率82%。
MS m/z(ESI):370[M+1]。
第六步
(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)甲胺083g
将6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-甲酰胺083f(1.00g,2.70mmol,1eq)溶于60mL四氢呋喃中,加入硼烷四氢呋喃溶液(1M)(27mL,27.00mmol,10eq),回流搅拌过夜。后处理:控温0℃加水淬灭并稀释反应液,用盐酸溶液调节pH值到3,用乙酸乙酯萃取掉有机杂质后,再用氢氧化钠溶液将水相pH值调至9,用乙酸乙酯萃取,有机相减压脱溶,得到(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)甲胺083g(0.70g,黄色固体),产率:73%。
MS m/z(ESI):356[M+1]。
第七步
N-((6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)甲基)丙烯酰胺
将化合物(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)甲胺083g(300mg,0.85mmol,1eq)溶于15mL二氯甲烷,加入N,N-二异丙基乙胺(220mg,1.70mmol,2eq),降温至-40℃,逐滴加入丙烯酰氯(76mg,0.85mmol,1eq),搅拌30min。后处理:用水-乙酸乙酯体系萃取,减压脱溶, 残余物经高效液相色谱分离纯化,得到N-((6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)甲基)丙烯酰胺(15mg,黄色固体),产率:4.3%。
MS m/z(ESI):410[M+1]。
1H NMR(400MHz,DMSO)δ8.47(t,J=5.3Hz,1H),6.88(s,1H),6.28(dd,J=17.1,10.1Hz,1H),6.13(dd,J=17.1,2.2Hz,1H),5.61(dd,J=10.1,2.2Hz,1H),4.32(qd,J=15.1,5.4Hz,2H),3.90(dd,J=15.8,3.3Hz,7H),3.39(t,J=6.2Hz,1H),2.75–2.53(m,3H),2.40(t,J=12.6Hz,1H),1.77(d,J=9.4Hz,1H),1.31(dd,J=14.9,7.4Hz,1H)。
实施例91
N-(2-(5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000030
第一步
3-(2,6-二氯-3,5-二甲氧苯基)丙烯酰酸乙基酯091b
将2,6-二氯-3,5-二甲氧基苯甲醛(1g,4.27mmol),氢化钠(60%)(512mg,8.54mmol),四氢呋喃(15mL)混合,0℃下搅拌,膦酰基乙酸三乙酯(1.43g,6.41mmol)溶于四氢呋喃(3mL)缓慢滴加至反应体系中,升温至室温,继续搅拌2h。后处理:向此混合液加入饱和氯化铵溶液(20mL),有机相减压脱溶,得到目标产物3-(2,6-二氯-3,5-二甲氧苯基)丙烯酰酸乙基酯091b(1.1g,白色固体),产率84%。
MS m/z(ESI):305[M+1]。
第二步
5-(2,6-二氯-3,5-二甲氧苯基)-3-羰基戊-4-烯腈091c
氮气保护下将正丁基锂(3mL,7.2mmol)在零下78℃下混于无水四氢呋喃(20mL),乙腈(296mg,7.2mmol)缓慢滴加至反应体系中,零下78℃下反应1小时,3-(2,6-二氯-3,5-二甲氧苯基)丙烯酰酸乙基酯(1.1g,3.6mmol)溶于无水四氢呋喃(5mL)缓慢滴加至反应体系中,缓慢升温至室温,搅拌3小时。后处理:向此混合物加入饱和氯化氨水溶液(200mL)淬灭反应,有机相减压脱溶,得到目标产物5-(2,6-二氯-3,5-二甲氧苯基)-3-羰基戊-4-烯腈091c(800mg,白色固体),产率72%。
MS m/z(ESI):300[M+1]。
第三步
5-(2,6-二氯-3,5-二甲氧基苯乙烯基)-1H-吡唑-3-胺091d
将5-(2,6-二氯-3,5-二甲氧苯基)-3-羰基戊-4-烯腈(800mg,2.67mmol),80%水合肼(2mL),乙酸(1mL)和乙醇(10mL)混合,升温至60℃,搅拌3小时。后处理:冷却至室温,减压脱溶,残余物用硅胶柱层析纯化(石油醚:乙酸乙酯=1:1),得到目标产物5-(2,6-二氯-3,5-二甲氧基苯乙烯基)-1H-吡唑-3-胺091d(400mg,白色固体),产率38%。
MS m/z(ESI):314[M+1]。
第四步
5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-胺091e
氢气氛围下将5-(2,6-二氯-3,5-二甲氧基苯乙烯基)-1H-吡唑-3-胺(400mg,1.27mmol),钯碳催化剂(100mg),甲醇(10mL)混合,常温搅拌3小时。后处理:过滤,减压脱溶,得到目标产物5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-胺091e(380mg,白色固体),产率95%。
MS m/z(ESI):316[M+1]。
第五步
3-溴-5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑091f
将5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-胺(5g,15.8mmol),氢溴酸(20mL)混合,常温搅拌1小时。亚硝酸钠(1.2g,17.3mmol)溶于水(5mL),0℃下缓慢滴加至反应体系中,继续搅拌2小时。将次混合液加入至溴化亚铜(3.4g,23.4mmol)与氢溴酸(50mL)的混合液中,常温搅拌3小时。后处理:向此反应液中加入二氯甲烷(200mL×3)萃取,合并有机相减压脱溶,所得残余物经硅胶柱层析(石油醚:乙酸乙酯=1:1)纯化,得到目标产物3-溴-5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑091f(1.8g,白色固体),产率30%。
MS m/z(ESI):378[M+1]。
第六步
5-(2,6-二氯-3,5-二甲氧基苯乙基)-3-(2-硝基苯基)-1H-吡唑091g
将3-溴-5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑(500mg,1.32mmol),邻硝基苯硼酸(265mg, 1.59mmol),氢氧化钠(158mg,3.96mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(430mg,0.528mmol),乙二醇二甲醚(5mL),水(2.5mL)混合,氮气保护下升温至100℃,搅拌6小时。后处理:减压除去乙二醇二甲醚,残余物经二氯甲烷(50mL),水(50mL)萃取,有机相合并后减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=1:1)纯化,得到5-(2,6-二氯-3,5-二甲氧基苯乙基)-3-(2-硝基苯基)-1H-吡唑091g(300mg,白色固体),产率53%。
MS m/z(ESI):422[M+1]。
第七步
2-(5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-基)苯胺091h
将5-(2,6-二氯-3,5-二甲氧基苯乙基)-3-(2-硝基苯基)-1H-吡唑(300mg,0.71mmol),锌粉(463mg,7.1mmol),氯化铵(376mg,7.1mmol)和乙醇(5mL)混合,升温至50℃,搅拌2小时。后处理:冷却至室温,过滤,减压脱溶,得到目标产物2-(5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-基)苯胺091h(250mg,白色固体),产率85%。
MS m/z(ESI):392[M+1]。
第八步
N-(2-(5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-基)苯基)丙烯酰胺091
将2-(5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-基)苯胺(100mg,0.25mmol),N,N-二异丙基乙胺(90mg,0.75mmol)和二氯甲烷(3mL)混合,-40℃搅拌下缓慢加入丙烯酰氯(25mg,0.28mmol),继续搅拌30分钟。后处理:向此混合物加10mL水,再用二氯甲烷(10mL×3)萃取,有机相合并后减压脱溶,残余物经高效液相色谱制备分离纯化,得到目标产物N-(2-(5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-基)苯基)丙烯酰胺091(15mg,白色固体),产率13%。
MS m/z(ESI):446[M+1]。
1H NMR(400MHz,DMSO)δ10.20(s,1H),8.08(s,1H),7.70(ddd,J=12.8,9.1,4.7Hz,1H),7.62(d,J=8.0Hz,1H),7.47(d,J=7.7Hz,1H),7.35(t,J=7.8Hz,1H),6.86(s,1H),6.47(dd,J=17.7,9.3Hz,2H),6.28(dd,J=17.0,1.5Hz,1H),5.77(m,1H),3.93(s,6H),3.24(m,2H),2.80(m,2H)。
实例093
Figure PCTCN2018104007-appb-000031
参照实例098d合成实例093a。
第一步:
6-(2,6-二氟-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑093b
将6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑093d(2g,5mmol)和乙腈(100ml)混合,0℃下加入select F(3.5g,10mmol),缓慢升温至室温,搅拌过夜。后处理:减压脱溶,残余物加入二氯甲烷(100mL)、水(100mL)萃取,有机相减压脱溶,残余物用硅胶柱层析(石油醚:乙酸乙酯=5:4)纯化,得到6-(2,6-二氟-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑093b(640mg,1.5mmol,黄色固体),产率:15%。
MS m/z(ESI):416[M+1]。
第二步:
2-(6-(2,6-二氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺093c
将6-(2,6-二氟-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑(200mg,0.5mmol),加入锌粉(135mg,3mmol),氯化铵(260mg,5mmol),水(1mL)和乙醇(10ml)混合,升温至50℃,搅拌2小时。后处理:过滤,减压脱溶,加入二氯甲烷(10mL)、水(10mL)萃取,有机相减压脱溶,得到2-(6-(2,6-二氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺093f(120mg,0.3mmol,棕色固体),产率:60%。
MS m/z(ESI):386[M+1]。
第三步:
N-(2-(6-(2,6-二氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺093
将2-(6-(2,6-二氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺(120mg,0.3mmol),N,N-二异丙基乙胺(119mg,1mmol)和二氯甲烷(3mL)混合,-40℃搅拌下缓慢加入丙烯酰氯(30mg,0.3mmol),继续搅拌30分钟。后处理:向此混合物加10mL水,再用二氯甲烷(10mL×3)萃取,有机相合并后减压脱溶,残余物经高效液相色谱制备分离纯化,得到N-(2-(6-(2,6-二氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺093(40mg,0.1mmol,白色固体),产率:60%。
MS m/z(ESI):440[M+1]。
1H NMR(400MHz,DMSO)δ12.98(s,1H),11.51(s,1H),8.47(s,1H),7.57(d,J=6.6Hz,1H),7.33(t,J=7.6Hz,1H),7.18(t,J=7.5Hz,1H),6.94(t,J=8.3Hz,1H),6.42–6.21(m,2H),5.81(d,J=11.3Hz,1H),3.97–3.80(m,7H),3.41(d,J=7.3Hz,1H),2.94(t,J=5.7Hz,5H),2.11(d,J=8.5Hz,1H)。
通过手性柱拆分得到P1和P2。手性拆分条件:设备SFC,色谱柱:chiralpak-AD流动相:CO2-IPA(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000032
实施例096
N-(2-(5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-基)苯基)丁-2-炔酰胺
Figure PCTCN2018104007-appb-000033
参照实施例91操作步骤合成实施例96,但在第八步将2-(5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-基)苯胺(100mg,0.25mmol),2-丁炔酸(23mg,0.28mmol)和二氯甲烷(5mL)混合,0℃搅拌下缓慢滴加二环己基碳二亚胺(57mg,0.28mmol),4-二甲氨基吡啶(6mg,0.025mmol),继续搅拌30分钟。向此混合物加10mL水,再用二氯甲烷(10mL×3)萃取,有机相合并后减压脱溶,残余物经高效液相色谱制备分离纯化,得到目标产物N-(2-(5-(2,6-二氯-3,5-二甲氧苄基氨基)-1H-吡唑-3-基)苯基)丙烯酰胺096(10mg,白色固体),产率8%。
MS m/z(ESI):468[M+1]。
1H NMR(400MHz,DMSO)δ12.76(s,1H),10.63(s,1H),8.06(s,1H),7.47(t,J=8.3Hz,2H),7.32(s,1H),6.85(s,1H),6.45(s,1H),3.92(s,6H),3.23(m,2H),2.80(s,2H),2.06(s,3H)。
实例098
Figure PCTCN2018104007-appb-000034
第一步:
3-(3,5-二甲氧苯基)环己酮098b
将环己烯酮(4g,41mmol),2-5二甲氧基苯硼酸(8.5g,47mmol),碳酸铯(13g,41mmol),醋酸钯(1g,4mmol),三苯基膦(2.2g,8mmol),氯仿(0.5mL)和甲苯(100mL)混合,氮气保护下升温至85℃,搅拌24小时。后处理:减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=10:1)纯化,得到3-(3,5-二甲氧苯基)环己酮098b(4.5g,黄色液体),产率:45%。
MS m/z(ESI):235[M+1]。
第二步:
5-(3,5-二甲氧苯基)-2-(2-硝基苯甲酰)环己酮098c
将3-(3,5-二甲氧苯基)环己酮(4.2g,18mmol)和四氢呋喃(100ml)混合,氮气保护下降温至-78℃,缓慢滴加二异丙基氨基锂(8.3mL,20mmol),升温在-40℃,搅拌2小时,缓慢加入邻硝基苯甲酰氯(3.5g,18mmol),升至室温搅拌3小时。后处理:用饱和氯化铵水溶液淬灭,有机相减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=3:1)纯化,得到5-(3,5-二甲氧苯基)-2-(2-硝基苯甲酰)环己酮098c(3.6g,黄色固体),产率:40%。
MS m/z(ESI):384[M+1]。
第三步:
6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑098d
将5-(3,5-二甲氧苯基)-2-(2-硝基苯甲酰)环己酮(3.6g,10mmol)、水合肼(5mL),乙酸(5mL)和乙醇(50mL)混合,升温至65℃,搅拌3小时。后处理:减压脱溶,残余物经归家柱层析(石油醚:乙酸乙酯=3:1)纯化,得到6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑098d(2g,黄色固体),产 率:65%。
MS m/z(ESI):380[M+1]。
第四步:
6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑098e
将6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑(2g,5mmol),N-氯代丁二酰亚胺(,12mmol)和乙酸(100mL)中,升温至80℃反应2h。后处理:加入二氯甲烷和水分层,有机相减压脱溶,用石油醚/乙酸乙酯(1:1)体系过硅胶层析柱得到黄色固体产物6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑098e(2.2g,5mmol,黄色固体),产率:90%。
MS m/z(ESI):448[M+1]。
第五步:
2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺098f
将6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑098e(400mg,1mmol)置于10mL DMF中,加入锌粉(310mg,5mmol,5eq),氯化铵(530mg,10mmol,10eq),1ml水,50℃反应0.8h。后处理:减压脱溶,二氯甲烷和水相分层,旋干有机相得到固体粗品产物2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺098f(230mg,0.5mmol,白色固体),产率:65%。
MS m/z(ESI):418[M+1]。
第六步:
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丁-2-炔酰胺098
将2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺098f(250mg,0.6mmol)置于20mL DCM中,加入DIPEA(260mg,2mmol,3eq)、HATU(300mg,0.8mmol,1.2eq),后缓慢加入2-丁炔酸(45mg,0.6mmol,1eq),0℃升至室温反应1h。后处理:减压脱溶,加入二氯甲烷和水相分层,有机相旋蒸除去溶剂,用制备液相分离得到白色固体产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丁-2-炔酰胺098(80mg,0.16mmol,白色固体),产率:50%。
MS m/z(ESI):484[M+1]。
1H NMR(400MHz,DMSO)δ12.98(s,1H),11.60(s,1H),8.30(s,1H),7.57(s,1H),7.33(s,1H),7.18(s,1H),6.89(s,1H),4.02(s,1H),3.91(m,6H),3.31(m,1H),2.71(m,4H),2.01(m,3H),1.80(s,1H)。
实施例100
N-(2-(5-(2,6-二氯-3,5-二甲氧苄基氨基)-1H-吡唑-3-基)苯基)丁-2-炔酰胺
Figure PCTCN2018104007-appb-000035
第一步
3-(2-硝基苯基)-3-羰基丙腈100b
氮气保护下将正丁基锂(69mL,164mmol)在零下78℃下混于无水四氢呋喃(200mL),乙腈(6.7g,164mmol)缓慢滴加至反应体系中,零下78℃下反应1小时,2-硝基苯甲酸甲酯(15g,82mmol)溶于无水四氢呋喃(100mL)缓慢滴加至反应体系中,缓慢升温至室温,搅拌3小时。后处理:向此混合物加入饱和氯化氨水溶液(200mL)淬灭反应,有机相减压脱溶,得到目标产物3-(2-硝基苯基)-3-羰基丙腈100b(10g,黄色固体),产率59%。
MS m/z(ESI):191[M+1]。
第二步
5-(2-硝基苯基)-1H-吡唑-3-胺100c
将3-(2-硝基苯基)-3-羰基丙腈(10g,52mmol),80%水合肼(3.15g,78mmol),乙酸(10mL)和乙醇(100mL)混合,升温至60℃,搅拌3小时。后处理:冷却至室温,减压脱溶,残余物用硅胶柱层析纯化(石油醚:乙酸乙酯=1:1),得到目标产物5-(2-硝基苯基)-1H-吡唑-3-胺100c(8g,黄色固体),产率79%。
MS m/z(ESI):205[M+1]。
第三步
N-(2,6-二氯-3,5-二甲氧苄基)-5-(2-硝基苯基)-1H-吡唑-3-胺100d
将5-(2-硝基苯基)-1H-吡唑-3-胺(1g,4.9mmol),2,6-二氯-3,5-二甲氧基苯甲醛(1.1g,4.9mmol), 乙酸(2mL)和甲醇(10mL)混合,常温搅拌3小时。搅拌下缓慢加入氰基硼氢化钠(455mg,7.35mmol),继续搅拌30分钟。后处理:减压脱溶,残余物通过硅胶柱层析纯化(石油醚:乙酸乙酯=1:1),得到目标产物N-(2,6-二氯-3,5-二甲氧苄基)-5-(2-硝基苯基)-1H-吡唑-3-胺100d(1.8g,白色固体),产率87%。
MS m/z(ESI):423[M+1]。
第四步
5-(2-氨基苯基)-N-(2,6-二氯-3,5-二甲氧苄基)-1H-吡唑-3-胺100e
将N-(2,6-二氯-3,5-二甲氧苄基)-5-(2-硝基苯基)-1H-吡唑-3-胺(200mg,0.47mmol),锌粉(30mg,4.7mmol),氯化铵(249mg,4.7mmol)和乙醇(5mL)混合,升温至50℃,搅拌2小时。后处理:冷却至室温,过滤,减压脱溶,得到目标产物5-(2-氨基苯基)-N-(2,6-二氯-3,5-二甲氧苄基)-1H-吡唑-3-胺100e(150mg,白色固体),产率80%。
MS m/z(ESI):393[M+1]。
第五步
N-(2-(5-(2,6-二氯-3,5-二甲氧苄基氨基)-1H-吡唑-3-基)苯基)丁-2-炔酰胺100
将化合物5-(2-氨基苯基)-N-(2,6-二氯-3,5-二甲氧苄基)-1H-吡唑-3-胺(150mg,0.38mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(120mg,0.38mmol),N,N-二异丙基乙胺(147mg,1.14mmol)和二氯甲烷(5mL)混合,0℃搅拌下缓慢加入2-丁炔酸(40mg,0.40mmol),继续搅拌30分钟。后处理:向此混合物加10mL水,再用二氯甲烷(10mL×3)萃取,有机相合并后减压脱溶,残余物经高效液相色谱制备分离纯化,得到目标产物N-(2-(5-(2,6-二氯-3,5-二甲氧苄基氨基)-1H-吡唑-3-基)苯基)丁-2-炔酰胺100(15mg,白色固体),产率9%。
MS m/z(ESI):459[M+1]。
1H NMR(400MHz,DMSO)δ12.21(d,J=41.7Hz,1H),8.79(m,1H),8.42(s,1H),7.66(m,1H),7.23(dd,J=33.2,25.1Hz,1H),6.94(s,1H),6.56(s,1H),6.00(d,J=35.9Hz,2H),4.49(d,J=5.5Hz,2H),3.94(s,6H),2.06(s,3H)。
实施例101
N-(2-(3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-1H-吡唑-5-基)苯基)丁-2-炔酰胺
Figure PCTCN2018104007-appb-000036
第一步
4-(2-硝基苯基)-2,4-二羰基丁酸乙基酯
将化合物1-(2-硝基苯基)乙烷-1-酮101a(16.80g,101.82mmol,1eq),草酸二乙酯(29.73g,203.64mmol,2eq)溶于无水四氢呋喃(85mL)中,控温0℃分批加入新制的乙醇钠(13.85g,203.64mmol,2eq),0℃下反应2小时。后处理:加50mL水淬灭反应,用乙酸乙酯萃取,有机相减压脱溶得目标产物4-(2-硝基苯基)-2,4-二羰基丁酸乙基酯101b(21.05g,棕色液体),产率:78%。
MS m/z(ESI):265[M+1]。
第二步
5-(2-硝基苯基)-1H-吡唑-3-羧酸乙基酯
将化合物4-(2-硝基苯基)-2,4-二羰基丁酸乙基酯101b(21.05g,79.4mmol,1eq),水合肼(7.94g,158.8mmol,2eq)溶于乙醇/醋酸10:1(165mL)中,回流反应2小时。后处理:减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=5:1)纯化,得目标产物5-(2-硝基苯基)-1H-吡唑-3-羧酸乙基酯101c(16.7g,黄色固体),产率:81%。
MS m/z(ESI):261[M+1]。
第三步
(5-(2-硝基苯基)-1H-吡唑-3-基)甲醇
将化合物5-(2-硝基苯基)-1H-吡唑-3-羧酸乙基酯(7.48g,28.66mmol,1eq)溶于无水四氢呋喃(120mL)中,控温0℃分批加入四氢铝锂(1.31g,34.39mmol,1.2eq),室温下反应2小时。后处理:控温0℃依次加入1.3mL水,1.3mL氢氧化钠溶液(15%),充分搅拌后滤除固体,滤液减压脱溶的残余物经硅胶柱层析(石油醚:乙酸乙酯=1:1)纯化,得目标产物(5-(2-硝基苯基)-1H-吡唑-3-基)甲醇。
101d(4.52g,黄色固体),产率:72%。
MS m/z(ESI):219[M+1]。
第四步
3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-5-(2-硝基苯基)-1H-吡唑
将化合物(5-(2-硝基苯基)-1H-吡唑-3-基)甲醇101d(307mg,1.4mmol,1eq)和1,5-二氯-2,4-二甲氧基苯酚8b(312mg,1.4mmol,1eq)以及三苯基磷(440mg,1.68mmol,1.2eq)溶解到30mL无水四氢呋喃中,降温到0℃,滴加偶氮二甲酸二乙酯(292mg,1.68mmol,1.2eq),升到室温搅拌2小时。加入100mL饱和氯化钠,分层,水相用100mL乙酸乙酯萃取,合并有机相,干燥减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=1:1),得到目标产物3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-5-(2-硝基苯基)-1H-吡唑101e(550mg,白色固体),产率93%。
MS m/z(ESI):424[M+1]。
第五步
2-(3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-1H-吡唑-5-基)苯胺
将化合物3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-5-(2-硝基苯基)-1H-吡唑101e(515mg,1.21mmol,1eq)溶于乙醇(25mL)中,加入水(2.5mL),锌粉(395mg,6.07mmol,5eq)和氯化铵(262mg,4.86mmol,4eq),50℃反应2h。后处理:滤除固体,旋干滤液,用乙酸乙酯稀释,水洗,旋干得目标产物2-(3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-1H-吡唑-5-基)苯胺101f(239mg,黄色固体)。产率:50%。
MS m/z(ESI):394[M+1]。
第六步
N-(2-(3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-1H-吡唑-5-基)苯基)丁-2-炔酰胺
将化合物2-(3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-1H-吡唑-5-基)苯胺101f(120mg,0.31mmol,1eq),丁炔酸(26mg,0.31mmol,1eq),N,N-二异丙基乙胺(118mg,0.92mmol,3eq)溶于二氯甲烷(20mL)中,加入2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(139mg,0.37mmol,1.2eq),室温反应2h。后处理:加水稀释,用乙酸乙酯萃取,有机相旋干,通过制备液相分离冻干得目标产物N-(2-(3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-1H-吡唑-5-基)苯基)丁-2-炔酰胺101(2.6mg,淡黄色固体)。产率:1.9%。
MS m/z(ESI):460[M+1]。
1H NMR(400MHz,DMSO)δ13.73(s,1H),11.80(s,1H),8.29(s,1H),7.80(d,J=7.9Hz,1H),7.33(t,J=7.8Hz,1H),7.21(d,J=7.5Hz,1H),6.96(s,1H),6.80(s,1H),5.08(s,2H),3.93(s,6H),2.07(s,3H)。
实施例102
N-(2-(5-(((2,6-二氯-3,5-二甲氧苯基)氨基)甲基)-1H-吡唑-3-基)苯基)丁-2-炔酰胺
Figure PCTCN2018104007-appb-000037
参考实例101d合成102a
第一步
2,6-二氯-3,5-二甲氧基-N-((3-(2-硝基苯基)-1H-吡唑-5-基)甲基)苯胺102b
将化合物(3-(2-硝基苯基)-1H-吡唑-5-基)甲醇102a(100mg,0.46mmol,1eq)和N,N-二异丙基乙胺(119mg,0.92mmol,2eq)加到无水二氯甲烷(15mL)中,氮气保护下,于0℃滴加甲基磺酰氯(58mg,0.50mmol,1.1eq),升到室温反应30min。减压旋走二氯甲烷,加入碳酸钾(58mg,0.50mmol,1.1eq),2,6-二氯-3,5-二甲氧基苯胺(58mg,0.50mmol,1.1eq)以及5mL的DMF,加热到80℃反应2h。后处理:加入15mL水稀释,用乙酸乙酯萃取,有机相减压脱溶,残余物用石油醚/乙酸乙酯(1:2)体系过硅胶层析柱得到黄色固体产物2,6-二氯-3,5-二甲氧基-N-((3-(2-硝基苯基)-1H-吡唑-5-基)甲基)苯胺102b(51mg,黄色固体)。产率:26%。
MS m/z(ESI):423[M+1]。
第二步
N-((3-(2-氨基苯基)-1H-吡唑-5-基)甲基)-2,6-二氯-3,5-二甲氧基苯胺102c
将化合物2,6-二氯-3,5-二甲氧基-N-((3-(2-硝基苯基)-1H-吡唑-5-基)甲基)苯胺102b(520mg,1.2mmol,1eq)溶于乙醇(10mL)中,加入锌粉(390mg,6.0mmol,5eq)和氯化铵(690mg,6.0mmol,5eq),50℃反应3h。后处理:旋干溶剂,用乙酸乙酯稀释,水洗,有机相旋干得目标产物N-((3-(2-氨基苯基)-1H-吡唑-5-基)甲基)-2,6-二氯-3,5-二甲氧基苯胺102c(390mg,黄色固体)。产率:81%。
MS m/z(ESI):393[M+1]。
第四步
N-(2-(5-(((2,6-二氯-3,5-二甲氧苯基)氨基)甲基)-1H-吡唑-3-基)苯基)丁-2-炔酰胺102
将化合物N-((3-(2-氨基苯基)-1H-吡唑-5-基)甲基)-2,6-二氯-3,5-二甲氧基苯胺102c(80mg,0.2mmol,1eq),丁炔酸(19mg,0.2mmol,1eq),N,N-二异丙基乙胺(52mg,0.4mmol,1eq)溶于DMF(6mL)中,加入2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(114mg,0.3mmol,1.5eq),室温反应3h。后处理:加水稀释,用乙酸乙酯萃取,有机相旋干,通过制备液相分离冻干得目标产物N-(2-(5-(((2,6-二氯-3,5-二甲氧苯基)氨基)甲基)-1H-吡唑-3-基)苯基)丁-2-炔酰胺102(2mg,白色固体)。产率:7%。
MS m/z(ESI):459[M+1]。
1H NMR(400MHz,DMSO)δ11.86(s,1H),8.27(dd,J=51.5,7.8Hz,1H),7.87–7.62(m,2H),7.28(dd,J=13.9,8.2Hz,2H),7.20–7.12(m,1H),7.02(s,1H),6.65(s,1H),6.50(s,1H),4.78-4.59(m,2H),3.86(s,6H),2.07(d,J=6.5Hz,3H)。
实例103
Figure PCTCN2018104007-appb-000038
第一步:
参照实例098b合成3-(3,5-二甲氧苯基)环己酮103b。
MS m/z(ESI):235[M+1]。
第二步:
5-(3,5-二甲氧苯基)-2-(4-硝基-1H-吡唑-3-羰基)环己酮103c
将3-(3,5-二甲氧苯基)环己酮103b(1.2g,5mmol)置于100ml四氢呋喃中,氮气保护。于-78℃时加入LDA(6mmol),升温在-40℃,保持2h,加入1-氢-4-硝基-3-甲酰氯基吡唑(900mg,5mmol),升至室温反应3h。后处理:用饱和氯化铵水溶液淬灭,有机相减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=3:1)纯化,得到5-(3,5-二甲氧苯基)-2-(4-硝基-1H-吡唑-3-羰基)环己酮103c(630mg,2mmol,黄色固体),产率:40%。
MS m/z(ESI):374[M+1]。
第三步:
6-(3,5-二甲氧苯基)-3-(4-硝基-1H-吡唑-3-基)-4,5,6,7-四氢-1H-吲唑103d
将5-(3,5-二甲氧苯基)-2-(4-硝基-1H-吡唑-3-羰基)环己酮103c(630mg,1.7mmol)、水合肼(5mL),乙酸(5mL)和乙醇(50mL)混合,升温至65℃,搅拌3小时。后处理:减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=3:1)纯化,得到黄色固体产物6-(3,5-二甲氧苯基)-3-(4-硝基-1H-吡唑-3-基)-4,5,6,7-四氢-1H-吲唑103d(580mg,1.2mmol黄色固体),产率:60%。
MS m/z(ESI):370[M+1]。
第四步:
6-(2,6-二氯-3,5-二甲氧苯基)-3-(4-硝基-1H-吡唑-3-基)-4,5,6,7-四氢-1H-吲唑103e
将6-(3,5-二甲氧苯基)-3-(4-硝基-1H-吡唑-3-基)-4,5,6,7-四氢-1H-吲唑103d(295mg,0.8mmol)和20ml乙酸混合,加入NCS(220mg,1.8mmol),80℃反应2h。后处理:加入二氯甲烷和水分层,有机相减压脱溶,用石油醚/乙酸乙酯(1:1)体系过硅胶层析柱得到黄色固体产物6-(2,6-二氯-3,5-二甲氧苯基)-3-(4-硝基-1H-吡唑-3-基)-4,5,6,7-四氢-1H-吲唑103e(120mg,0.3mmol,黄色固体),产率:90%。
MS m/z(ESI):438[M+1]。
第五步:
3-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1H-吡唑-4-胺103f
将6-(2,6-二氯-3,5-二甲氧苯基)-3-(4-硝基-1H-吡唑-3-基)-4,5,6,7-四氢-1H-吲唑103e(120mg,0.3mmol)和N,N-二甲基甲酰胺(10mL)混合,加入锌粉(100mg,1.5mmol),氯化铵(160mg,3mmol),1ml水,50℃反应0.8小时。后处理:减压脱溶,加入二氯甲烷和水相分层,有机相减压脱溶得到固体粗品产物3-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1H-吡唑-4-胺103f(90mg,0.22mmol,淡黄色固体),产率:60%。
MS m/z(ESI):408[M+1]。
第六步:
N-(3-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1H-吡唑-4-基)丙烯酰胺103
将3-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1H-吡唑-4-胺103f(90mg,0.3mmol)和二氯甲烷(20mL)混合,加入二异丙基乙胺(129mg,1mmol),降至-40℃后缓慢加入丙烯酰氯(30mg, 0.3mmol),反应1h。后处理:加入二氯甲烷和水相分层,有机相减压脱溶,经制备液相分离得到白色固体产物N-(3-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1H-吡唑-4-基)丙烯酰胺103(40mg,0.1mmol,白色固体),30%产率:60%。
MS m/z(ESI):462[M+1]。
1H NMR(400MHz,DMSO)δ9.95(s,1H),8.18(s,1H),6.89(s,1H),6.44(m,1H),6.22(d,J=16.9Hz,1H),5.76(d,J=10.4Hz,1H),3.93(s,6H),3.43(m,2H),3.01(d,J=12.7Hz,1H),2.69(m,3H),1.82(d,J=8.6Hz,1H)。
实例107
N-(3-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1-甲基-1H-吡唑-4-基)丙烯酰胺
Figure PCTCN2018104007-appb-000039
参照实例103合成实例107,但在第二步将1-氢-4-硝基-3-甲酰氯基吡唑替换成1-甲基-4-硝基-3-甲酰氯基吡唑。
MS m/z(ESI):476[M+1]。
1H NMR(400MHz,DMSO)δ12.70(s,1H),10.00(s,1H),8.21(s,1H),6.89(s,1H),6.48–6.36(m,1H),6.22(d,J=16.6Hz,1H),5.77(d,J=10.7Hz,1H),3.97(d,J=7.9Hz,1H),3.93(s,6H),3.86(s,3H),3.47–3.37(m,1H),3.04(s,1H),2.78–2.56(m,3H),1.81(s,1H)。
实例109
N-(2-(6-(2,6-二氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丁-2-炔酰胺
Figure PCTCN2018104007-appb-000040
参照实例093合成实例109,但在第四步用2-丁炔酸代替丙烯酰氯:将098f(200mg,0.5mmol)和20mL二氯甲烷混合,加入N,N-二异丙基乙胺(190mg,1.5mmol)、HATU(260mg,0.7mmol),后缓慢加入2-丁炔酸(40mg,0.5mmol),0℃升至室温反应1h。后处理:加入二氯甲烷和水相分层,有机相减压脱溶,经制备液相分离得到白色固体产物N-(2-(6-(2,6-二氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丁-2-炔酰胺109(60mg,0.13mmol,白色固体)。
MS m/z(ESI):452[M+1]。
1H NMR(400MHz,DMSO)δ12.99(s,1H),11.59(s,1H),8.29(s,1H),7.58(s,1H),7.34(s,1H),7.20(s,1H),6.89(s,1H),4.01(s,1H),3.90(m,6H),3.30(m,1H),2.69(m,4H),1.99(m,3H),1.79(s,1H)。
实施例111
N-(2-(5-(2,6-二氯-3,5-二甲氧苄基氨基)-1H-吡唑-3-基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000041
参照实施例100操作步骤合成实施例111,但在第五步将5-(2-氨基苯基)-N-(2,6-二氯-3,5-二甲氧苄基)-1H-吡唑-3-胺(150mg,0.38mmol),N,N-二异丙基乙胺(147mg,1.14mmol)和二氯甲烷(5mL)混合,0℃搅拌下缓慢滴加丙烯酰氯(36mg,0.40mmol),继续搅拌30分钟。后处理:向此混合物加10mL水,再用二氯甲烷(10mL×3)萃取,有机相合并后减压脱溶,残余物经高效液相色谱制备分离纯化,得到目标产物N-(2-(5-(2,6-二氯-3,5-二甲氧苄基氨基)-1H-吡唑-3-基)苯基)丙烯酰胺111(10mg,白色固体),产率6%。
MS m/z(ESI):447[M+1]。
1H NMR(400MHz,DMSO)δ12.23(s,1H),11.98(s,1H),8.58(s,1H),7.66(d,J=7.6Hz,1H),7.28(t,J=7.7Hz,1H),7.14(t,J=7.3Hz,1H),6.94(s,1H),6.33(d,J=8.0Hz,2H),5.98(s,1H),5.83(m,1H),4.50(d,J=5.6Hz,2H),3.94(s,6H)。
实施例112
N-(2-(3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-1H-吡唑-5-基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000042
参照实例101步骤合成实例112,但在第六步用丙烯酰氯取代丁炔酸,合成制备N-(2-(3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-1H-吡唑-5-基)苯基)丙烯酰胺112。
第六步
N-(2-(3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-1H-吡唑-5-基)苯基)丙烯酰胺
将化合物化合物2-(3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-1H-吡唑-5-基)苯胺101f(100mg,0.25mmol,1eq)溶于15mL二氯甲烷,加入N,N-二异丙基乙胺(98mg,0.76mmol,3eq),降温至-40℃,逐滴加入丙烯酰氯(16mg,0.18mmol,0.7eq),搅拌30min。后处理:用水-乙酸乙酯体系萃取,减压脱溶得到目标产物N-(2-(3-((2,6-二氯-3,5-二甲氧基苯氧基)甲基)-1H-吡唑-5-基)苯基)丙烯酰胺粗品,经制备可得到(1.5mg,黄色固体),产率:1.3%。
MS m/z(ESI):448[M+1]。
1H NMR(400MHz,DMSO)δ13.57(s,1H),11.76(s,1H),8.53(s,1H),7.82(d,J=7.9Hz,1H),7.33(t,J=7.8Hz,1H),7.17(d,J=7.5Hz,1H),6.96(s,1H),6.79(s,1H),6.32(s,2H),5.86(s,1H),5.10(s,2H),3.89(d,J=26.7Hz,6H)。
实施例113
N-(2-(5-(((2,6-二氯-3,5-二甲氧苯基)氨基)甲基)-1H-吡唑-3-基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000043
参考实例102合成113a。
将化合物N-((3-(2-氨基苯基)-1H-吡唑-5-基)甲基)-2,6-二氯-3,5-二甲氧基苯胺113a(100mg,0.26mmol,1eq)溶于二氯甲烷(10mL),加入N,N-二异丙基乙胺(67mg,0.52mmol,2eq),降温至-40℃,滴加丙烯酰氯(23mg,0.26mmol,1eq)的二氯甲烷溶液,搅拌30min。后处理:减压脱溶,用制备液相 分离,冻干得到目标产物N-(2-(5-(((2,6-二氯-3,5-二甲氧苯基)氨基)甲基)-1H-吡唑-3-基)苯基)丙烯酰胺113(12mg,白色固体)。产率:10%。
MS m/z(ESI):447[M+1]。
1H NMR(400MHz,DMSO)δ13.15(s,1H),11.86(s,1H),8.54(d,J=7.3Hz,1H),7.69(d,J=7.5Hz,1H),7.30(t,J=7.5Hz,1H),7.15(d,J=7.0Hz,1H),6.66(s,1H),6.50(s,1H),6.32(d,J=9.6Hz,2H),5.85(d,J=7.1Hz,1H),5.25(s,1H),4.60(d,J=6.9Hz,2H),3.86(s,6H)。
实例114
Figure PCTCN2018104007-appb-000044
同实例093合成N-(2-(6-(2-氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺114。
MS m/z(ESI):422[M+1]。
1H NMR(400MHz,DMSO)δ12.98(s,1H),11.59(s,1H),8.48(s,1H),7.58(d,J=7.4Hz,1H),7.33(t,J=7.6Hz,1H),7.19(t,J=7.5Hz,1H),6.63(dd,J=6.9,2.8Hz,1H),6.55–6.45(m,1H),6.38–6.19(m,2H),5.82(d,J=11.4Hz,1H),3.79(d,J=35.1Hz,6H),3.28(s,1H),2.96–2.74(m,4H),1.97(s,2H)。
实施例117
N-(3-(5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-基氨基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000045
参照实施例091e合成117a。
第一步
3-(2,6-二氯-3,5-二甲氧基苯乙基)-N-(2-硝基苯基)-1H-吡唑-5-胺117b
将5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-胺(100mg,0.31mmol),邻氟硝基苯(53mg,0.37mmol),N,N-二甲基甲酰胺(3mL)混合,氮气保护下升温至120℃,搅拌3小时。后处理:减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=1:1)纯化,得到3-(2,6-二氯-3,5-二甲氧基苯乙基)-N-(2-硝基苯基)-1H-吡唑-5-胺117b(50mg,黄色固体),产率36%。
MS m/z(ESI):436[M+1]。
第二步
N1-(3-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-5-基)苯-1,2-二胺117c
将3-(2,6-二氯-3,5-二甲氧基苯乙基)-N-(2-硝基苯基)-1H-吡唑-5-胺(300mg,0.68mmol),锌粉(447mg,6.8mmol),氯化铵(360mg,6.8mmol)和乙醇(5mL)混合,升温至50℃,搅拌2小时。后处理:冷却至室温,过滤,减压脱溶,得到目标产物N1-(3-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-5-基)苯-1,2-二胺117c(240mg,白色固体),产率86%。
MS m/z(ESI):406[M+1]。
第三步
N-(2-(3-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-5-基氨基)苯基)丙烯酰胺
将N1-(3-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-5-基)苯-1,2-二胺(100mg,0.24mmol),N,N-二异丙基乙胺(92mg,0.72mmol)和二氯甲烷(5mL)混合,0℃搅拌下缓慢滴加丙烯酰氯(26mg,0.29mmol),继续搅拌30分钟。后处理:向此混合物加10mL水,再用二氯甲烷(10mL×3)萃取,有机相合并后减压脱溶,残余物经高效液相色谱制备分离纯化,得到目标产物N-(2-(3-(2,6-二氯-3,5-二甲氧基苯乙 基)-1H-吡唑-5-基氨基)苯基)丙烯酰胺(15mg,白色固体),产率13%
MS m/z(ESI):460[M+1]。
1H NMR(400MHz,DMSO)δ11.93(s,1H),9.63(s,1H),7.58(s,1H),7.37(d,J=12.9Hz,2H),7.07(t,J=7.8Hz,1H),6.85(s,1H),6.79(t,J=7.4Hz,1H),6.51(dd,J=16.8,10.4Hz,1H),6.25(d,J=17.1Hz,1H),5.74(m,2H),3.92(s,6H),3.18(m,2H),2.73(m,2H)。
实施例120
N-(3-(5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-基氨基)苯基)丁-2-炔酰胺
Figure PCTCN2018104007-appb-000046
参照实施例117操作步骤合成实施例120,但在第三步将N1-(3-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-5-基)苯-1,2-二胺(100mg,0.24mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(137mg,0.36mmol),N,N-二异丙基乙胺(87mg,0.72mmol)和二氯甲烷(5mL)混合,0℃搅拌下缓慢加入2-丁炔酸(29mg,1.2mmol),继续搅拌30分钟。后处理:向此混合物加10mL水,再用二氯甲烷(10mL×3)萃取,有机相合并后减压脱溶,残余物经高效液相色谱制备分离纯化,得到目标产物N-(3-(5-(2,6-二氯-3,5-二甲氧基苯乙基)-1H-吡唑-3-基氨基)苯基)丁-2-炔酰胺120(15mg,白色固体),产率12%。
MS m/z(ESI):472[M+1]。
1H NMR(400MHz,DMSO)δ11.91(s,1H),9.95(s,1H),7.59(d,J=20.7Hz,1H),7.35(s,1H),7.28(d,J=7.6Hz,1H),7.08(t,J=7.6Hz,1H),6.84(d,J=6.5Hz,1H),6.77(t,J=7.5Hz,1H),5.72(s,1H),3.92(s,6H),3.18(m,2H),2.73(m,2H),2.05(s,3H)。
实施例122
N-((1R,2R)-2-(5-(2,6-二氯-3,5-二甲氧基苄基氨基)-1H-吡唑-3-基)环己基)丙烯酰胺
Figure PCTCN2018104007-appb-000047
第一步
(1R,2R)-2-(苄氧基羰基氨基)环己羧酸甲基酯122b
将(1R,2R)-2-(苄氧基羰基氨基)环己羧酸(1g,3.6mmol),二氯亚砜(2mL),甲醇(10mL)混合,常温搅拌过夜。后处理:减压脱溶,得到(1R,2R)-2-(苄氧基羰基氨基)环己羧酸甲基酯122b(1.1g,白色固体),产率95%。
MS m/z(ESI):292[M+1]。
第二步
(1R,2R)-2-(2-氰基乙酰基)环己基氨基甲酸苯甲基酯122c
氮气保护下将正丁基锂(3.08mL,7.4mmol)在零下78℃下混于无水四氢呋喃(20mL),乙腈(303mg,7.4mmol)缓慢滴加至反应体系中,零下78℃下反应1小时,(1R,2R)-2-(苄氧基羰基氨基)环己羧酸甲基酯(1.1g,3.7mmol)溶于无水四氢呋喃(5mL)缓慢滴加至反应体系中,缓慢升温至室温,搅拌3小时。后处理:向此混合物加入饱和氯化氨水溶液(200mL)淬灭反应,有机相减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=1:1)纯化,得到目标产物(1R,2R)-2-(2-氰基乙酰基)环己基氨基甲酸苯甲基酯122c(580mg,白色固体),产率51%。
MS m/z(ESI):301[M+1]。
第三步
(1R,2R)-2-(3-氨基-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯122d
将(1R,2R)-2-(2-氰基乙酰基)环己基氨基甲酸苯甲基酯(580mg,1.93mmol),80%水合肼(2mL),乙酸(2mL)和乙醇(20mL)混合,升温至60℃,搅拌3小时。后处理:冷却至室温,减压脱溶,残余物用硅胶柱层析纯化(石油醚:乙酸乙酯=1:1),得到目标产物(1R,2R)-2-(3-氨基-1H-吡唑-5-基)环己基氨基 甲酸苯甲基酯122d(320mg,黄色固体),产率60%。
MS m/z(ESI):315[M+1]。
第四步
(1R,2R)-2-(3-(2,2,2-三氟乙酰氨基)-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯122e
将(1R,2R)-2-(3-氨基-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯(320mg,1.01mmol),4-二甲氨基吡啶(49mg,0.4mmol),N,N-二异丙基乙胺(390mg,3.03mmol)和二氯甲烷(5mL)混合,0℃搅拌下缓慢加入三氟乙酸酐(428mg,2.02mmol),升温至常温,搅拌3小时。后处理:向此混合物加10mL水,再用二氯甲烷(10mL×3)萃取,有机相合并后减压脱溶,得到目标产物(1R,2R)-2-(3-(2,2,2-三氟乙酰氨基)-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯122e(300mg,白色固体),产率69%。
MS m/z(ESI):411[M+1]。
第五步
N-(5-((1R,2R)-2-氨基环己基)-1H-吡唑-3-基)-2,2,2-三氟乙酰胺122f
氢气氛围下将(1R,2R)-2-(3-(2,2,2-三氟乙酰氨基)-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯(300mg,0.73mmol),钯碳催化剂(100mg),甲醇(10mL)混合,常温搅拌3小时。后处理:过滤,减压脱溶,得到目标产物N-(5-((1R,2R)-2-氨基环己基)-1H-吡唑-3-基)-2,2,2-三氟乙酰胺122f(110mg,白色固体),产率95%。
MS m/z(ESI):277[M+1]。
第六步
(1R,2R)-2-(3-(2,2,2-三氟乙酰氨基)-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯122g
将N-(5-((1R,2R)-2-氨基环己基)-1H-吡唑-3-基)-2,2,2-三氟乙酰胺(110mg,0.39mmol),N,N-二异丙基乙胺(150mg,1.17mmol)和四氢呋喃(3mL)混合,常温搅拌下缓慢滴加二碳酸二叔丁酯(102mg,0.47mmol),继续搅拌3小时。后处理:减压脱溶,残余物经硅胶柱层析纯化,得到(1R,2R)-2-(3-(2,2,2-三氟乙酰氨基)-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯122g(90mg,白色固体),产率61%。
MS m/z(ESI):377[M+1]。
第七步
(1R,2R)-2-(3-氨基-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯122h
将(1R,2R)-2-(3-(2,2,2-三氟乙酰氨基)-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯(90mg,0.23mmol),氢氧化钾(26mg,0.46mmol)和甲醇(3mL)混合,升温至50℃,搅拌3小时。后处理:过滤,减压脱溶,得到(1R,2R)-2-(3-氨基-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯122h(60mg,白色固体),产率89%。
MS m/z(ESI):281[M+1]。
第八步
(1R,2R)-2-(3-(2,6-二氯-3,5-二甲氧苄基氨基)-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯122i
将(1R,2R)-2-(3-氨基-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯(100mg,0.35mmol),2,6-二氯-3,5-二甲氧基苯甲醛(99mg,0.42mmol),乙酸(0.5mL)和甲醇(3mL)混合,常温搅拌3小时。搅拌下缓慢加入氰基硼氢化钠(32mg,0.52mmol),继续搅拌30分钟。后处理:减压脱溶,残余物通过硅胶柱层析纯化(石油醚:乙酸乙酯=1:1),得到(1R,2R)-2-(3-(2,6-二氯-3,5-二甲氧苄基氨基)-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯122i(80mg,白色固体),产率67%。
MS m/z(ESI):499[M+1]。
第九步
5-((1R,2R)-2-氨基环己基)-N-(2,6-二氯-3,5-二甲氧苄基)-1H-吡唑-3-胺122j
将(1R,2R)-2-(3-(2,6-二氯-3,5-二甲氧苄基氨基)-1H-吡唑-5-基)环己基氨基甲酸苯甲基酯(150mg,0.30mmol)和4M盐酸二氧六环(3mL)混合,常温搅拌3小时。后处理:减压脱溶,得到5-((1R,2R)-2-氨基环己基)-N-(2,6-二氯-3,5-二甲氧苄基)-1H-吡唑-3-胺122j(100mg,白色固体),产率84%。
MS m/z(ESI):399[M+1]。
第十步
N-((1R,2R)-2-(3-(2,6-二氯-3,5-二甲氧苄基氨基)-1H-吡唑-5-基)环己基)丙烯酰胺122
将5-((1R,2R)-2-氨基环己基)-N-(2,6-二氯-3,5-二甲氧苄基)-1H-吡唑-3-胺(100mg,0.25mmol),N,N-二异丙基乙胺(96mg,0.75mmol)和二氯甲烷(5mL)混合,0℃搅拌下缓慢滴加丙烯酰氯(27mg,0.30mmol),继续搅拌30分钟。后处理:向此混合物加10mL水,再用二氯甲烷(10mL×3)萃取,有机相合并后减压脱溶,残余物经高效液相色谱制备分离纯化,得到目标产物N-((1R,2R)-2-(3-(2,6-二氯-3,5-二甲氧苄基氨基)-1H-吡唑-5-基)环己基)丙烯酰胺122(15mg,白色固体),产率13%。
MS m/z(ESI):453[M+1]。
1H NMR(400MHz,DMSO)δ11.13(s,1H),7.88(d,J=8.6Hz,1H),6.88(s,1H),6.04(m,2H),5.50(dd,J=9.9,2.4Hz,1H),5.29(s,1H),4.91(s,1H),4.36(d,J=5.8Hz,2H),3.91(s,6H),3.77(m,1H),1.88(d,J=10.2Hz,2H),1.68(d,J=11.6Hz,2H),1.34(m,4H),1.18(s,1H)。
实施例133
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-5-((2-(二甲氨基)乙基)(甲基)氨基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000048
第一步
4-氟-2-硝基苯甲酰氯133b
将化合物4-氟-2-硝基苯甲酸133a(5g,27.0mmol,1eq)加到二氯氩亚砜(60mL)中,滴加1mLDMF催化,回流反应3h。后处理:减压脱溶,旋干得目标产物4-氟-2-硝基苯甲酰氯133b(5.1g,淡黄色固体)。产率:93%。
第二步
5-(3,5-二甲氧苯基)-2-(4-氟-2-硝基苯甲酰)环己烷-1-酮133c
将二异丙基氨基锂(1.3mL,2.5mmol,1.1eq)于-78℃缓慢滴加到化合物3-(3,5-二甲氧苯基)环己烷-1-酮10a(540mg,2.3mmol,1eq)的无水四氢呋喃(10mL)溶液中,在-30℃到-40℃搅拌两个小时。然后将4-氟-2-硝基苯甲酰氯133b(515mg,2.5mmol,1.1eq)滴加到反应液中,升至室温,搅拌2小时。后处理:加10mL氯化铵水溶液淬灭反应,分层有机相减压脱溶,残余物经硅胶柱层析纯化(石油醚:乙酸乙酯=10:1),得到目标产物乙基5-(3,5-二甲氧苯基)-2-(4-氟-2-硝基苯甲酰)环己烷-1-酮133c(470mg,黄色液体),产率:51%。
MS m/z(ESI):402[M+1]。
第三步
6-(3,5-二甲氧苯基)-3-(4-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑133d
将化合物5-(3,5-二甲氧苯基)-2-(4-氟-2-硝基苯甲酰)环己烷-1-酮133c(460mg,1.15mmol,1eq),水合肼(143mg,2.3mmol,2eq)溶于乙醇/醋酸10:1(11mL)中,在65℃下反应3小时。后处理:减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=2:1)纯化,得目标产物6-(3,5-二甲氧苯基)-3-(4-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑133d(240mg,黄色固体),产率:53%。
MS m/z(ESI):398[M+1]。
第四步
6-(2,6-二氯-3,5-二甲氧苯基)-3-(4-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑113e
将6-(3,5-二甲氧苯基)-3-(4-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑133d(1g,2.5mmol,1eq)加到50ml乙酸中,加入NCS(1g,7.5mmol,3eq),65℃反应3h。后处理:减压脱溶,残余物用石油醚/乙酸乙酯(2:1)体系过硅胶层析柱得到黄色固体产物6-(2,6-二氯-3,5-二甲氧苯基)-3-(4-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑113e(1g,黄色固体)。产率:86%。
MS m/z(ESI):466[M+1]。
第五步
N1-(4-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-3-硝基苯基)-N1,N2,N2-三甲基乙烷-1,2-二胺113f
将6-(2,6-二氯-3,5-二甲氧苯基)-3-(4-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑113e(1.3g,2.8mmol,1eq)加到10ml二甲基亚砜中,加入N1,N1,N2-三甲基乙烷-1,2-二胺(1.4g,14.0mmol,5eq),150℃反应3h。后处理:减压脱溶,残余物用二氯甲烷/甲醇(8:1)体系过硅胶层析柱得到黄色固体产物N1-(4-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-3-硝基苯基)-N1,N2,N2-三甲基乙烷-1,2-二胺113f(387mg,黄色固体)。产率:25%。
MS m/z(ESI):548[M+1]。
第六步
4-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-N1-(2-(二甲氨基)乙基)-N1-甲基苯-1,3-二胺113g
将化合物N1-(4-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-3-硝基苯基)-N1,N2,N2-三甲基乙烷-1,2-二胺113f(380mg,0.69mmol,1eq)溶于乙醇(15mL)中,加入锌粉(226mg,3.47mmol,5eq)和氯化铵(184mg,3.47mmol,5eq),50℃反应3h。后处理:旋干溶剂,用乙酸乙酯稀释,水洗,有机相旋干得目标产物4-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-N1-(2-(二甲氨基)乙基)-N1-甲基苯-1,3-二胺113g(180mg,黄色固体)。产率:50%。
MS m/z(ESI):518[M+1]。
第七步
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-5-((2-(二甲氨基)乙基)(甲基)氨基)苯基)丙烯酰胺113
将化合物4-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-N1-(2-(二甲氨基)乙基)-N1-甲基苯-1,3-二胺113g(180mg,0.35mmol,1eq)溶于15mL二氯甲烷,加入N,N-二异丙基乙胺(90mg,0.70mmol,2eq),降温至-40℃,滴加丙烯酰氯(31mg,0.35mmol,1eq)的二氯甲烷溶液,搅拌30min。后处理:减压脱溶,用制备液相分离,冻干得到目标产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-5-((2-(二甲氨基)乙基)(甲基)氨基)苯基)丙烯酰胺113(11mg,白色固体)。产率:5.5%。
MS m/z(ESI):572[M+1]。
1H NMR(400MHz,DMSO)δ12.84(s,1H),10.87(s,1H),8.16(s,1H),6.89(s,1H),6.80(s,1H),6.71(dd,J=9.1,2.4Hz,1H),6.32(dd,J=16.9,10.0Hz,1H),6.18(d,J=16.7Hz,1H),5.72(d,J=10.9Hz,1H),4.08–3.98(m,1H),3.93(d,J=3.6Hz,6H),3.57–3.35(m,4H),2.93(s,3H),2.70(ddd,J=20.1,14.1,6.4Hz,4H),2.28(s,6H),1.80(d,J=10.2Hz,1H)。
通过手性柱拆分得到P1和P2。手性拆分条件:设备SFC,色谱柱:chiralpak-AD流动相:CO2-IPA(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000049
实例137
Figure PCTCN2018104007-appb-000050
第一步:
参照实例098b合成3-(3,5-二甲氧苯基)环己酮137b。
第二步:
5-(3,5-二甲氧苯基)-2-(5-氟-2-硝基苯甲酰)环己酮103c
将3-(3,5-二甲氧苯基)环己酮103b(4g,15mmol)和100mL四氢呋喃混合,氮气保护。于-78℃时加入二异丙基氨基锂(9mL,18mmol),升温至-40℃之间保持2h,加入2-硝基-5-氟-苯甲酰氯(3.5g,15mmol),升至室温反应3h。后处理:饱和氯化铵水溶液淬灭,加入二氯甲烷和水相分层,有机相减压脱溶后用石油醚/乙酸乙酯(3:1)体系过硅胶层析柱得到产物5-(3,5-二甲氧苯基)-2-(5-氟-2-硝基苯甲酰)环己酮137c(5g,1O mmol,黄色固体),产率:40%。
MS m/z(ESI):402[M+1]。
第三步:
6-(3,5-二甲氧苯基)-3-(5-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑137d
将5-(3,5-二甲氧苯基)-2-(5-氟-2-硝基苯甲酰)环己酮137c(4g,10mmol)、水合肼(2g,25mmol)、100ml乙酸/乙醇(1:10)混合,65℃搅拌3h。后处理:加入二氯甲烷和水相分层,有机相减压脱溶后用石油醚/乙酸乙酯(2:1)体系过硅胶层析柱得到黄色固体产物137d(3.1g,7mmol,黄色固体),产率:70%。
MS m/z(ESI):398[M+1]。
第四步:
6-(2,6-二氯-3,5-二甲氧苯基)-3-(5-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑137e
将6-(3,5-二甲氧苯基)-3-(5-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑137d(1g,2.5mmol)和50mL乙酸混合,加入N-氯代丁二酰亚胺(1g,7.5mmol),80℃反应2h。后处理:加入二氯甲烷和水相分层,有机相减压脱溶后用石油醚/乙酸乙酯(1:1)体系过硅胶层析柱得到黄色固体产物6-(2,6-二氯-3,5-二甲氧苯基)-3-(5-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑137e(1.1g,2.3mmol,黄色固体),产率:85%。
MS m/z(ESI):466[M+1]。
第五步:
N1-(3-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-硝基苯基)-N1,N2,N2-三甲基乙烷-1,2-二胺137f
将6-(2,6-二氯-3,5-二甲氧苯基)-3-(5-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑137e(1.1g,2.3mmol)、N-N’二甲基-N-甲基乙二胺(1.2g,12mmol)和5mL二甲基亚砜混合于封管中,110℃反应1h。后处理:减压脱溶得到固体粗品产物N1-(3-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-硝基苯基)-N1,N2,N2-三甲基乙烷-1,2-二胺137f(750mg,1.3mmol,黄色固体),产率:55%。
MS m/z(ESI):548[M+1]。
第六步:
3-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-N1-(2-(二甲氨基)乙基)-N1-甲基苯-1,4-二胺137g
将N1-(3-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-硝基苯基)-N1,N2,N2-三甲基乙烷-1,2-二胺137f(750mg,1.3mmol)和20ml乙醇混合,加入锌粉(500mg,8mmol),氯化铵(700mg,13mmol),3ml水,50℃反应2h。后处理:减压脱溶,加入二氯甲烷和水相分层,有机相用二氯甲烷/甲醇(10:1)体系过中性氧化铝层析柱得到固体产物3-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-N1-(2-(二甲氨基)乙基)-N1-甲基苯-1,4-二胺137g(400mg,0.77mmol,白色固体),产率:60%。
MS m/z(ESI):518[M+1]。
第七步:
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-((2-(二甲氨基)乙基)(甲基)氨基)苯基)丙烯酰胺137
将3-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-N1-(2-(二甲氨基)乙基)-N1-甲基苯-1,4-二胺137g(400mg,0.8mmol)和20mL DCM混合,加入N,N-二异丙基乙胺(300mg,2.4mmol),降至-40℃后缓慢加入丙烯酰氯(40mg,0.5mmol),反应1h。后处理:加入二氯甲烷和水相分层,有机相减压脱溶,经制备液相分离得到白色固体产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-((2-(二甲氨基)乙基)(甲基)氨基)苯基)丙烯酰胺137(100mg,0.2mmol,白色固体),产率:30%。
MS m/z(ESI):572[M+1]。
1H NMR(400MHz,DMSO)δ12.84(s,1H),10.87(s,1H),8.16(s,1H),6.89(s,1H),6.80(s,1H),6.71(dd,J=9.1,2.4Hz,1H),6.32(dd,J=16.9,10.0Hz,1H),6.18(d,J=16.7Hz,1H),5.72(d,J=10.9Hz,1H),4.08–3.98(m,1H),3.93(d,J=3.6Hz,6H),3.57–3.35(m,4H),2.93(s,3H),2.70(ddd,J=20.1,14.1,6.4Hz,4H),2.28(s,6H),1.80(d,J=10.2Hz,1H)。
通过手性柱拆分得到P1和P2。手性拆分条件:设备SFC,色谱柱:chiralpak-AD流动相:CO2-IPA(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000051
实施例140
Figure PCTCN2018104007-appb-000052
参照实施例103操作步骤合成实施例140,但在第一步用1-甲基-4-硝基-5-甲酰氯基吡唑替代1-氢-4-硝基-3-甲酰氯基吡唑。
MS m/z(ESI):476[M+1]。
1H NMR(400MHz,DMSO)δ12.91(s,1H),9.47(s,1H),7.85(s,1H),6.89(s,1H),6.52(dd,J=17.2,9.8Hz,1H),6.19(d,J=16.5Hz,1H),5.70(m,1H),3.93(d,J=2.5Hz,6H),3.77(s,3H),2.73(m,2H),2.34(s,1H),1.74(m,2H),1.24(s,2H)。
通过手性柱拆分得到P1和P2。手性拆分条件:设备SFC,色谱柱:chiralpak-AD流动相:CO2-IPA(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000053
实施例015-P1
(R)-N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000054
第一步:
(R)-3-(3,5-二甲氧苯基)环己烷-1-酮
氩气保护下,将3,5-二甲氧基苯硼酸(22.7g,123mmol),乙酰丙酮二(乙烯)铑(260mg,1mmol),(R)-BINAP(935mg,1.5mmol)加入到250mL二氧六环和25mL水的混合溶剂中后,再加入2-环 已烯-1-酮(4.8g,50mmol)。上述反应液于105℃油浴中反应6小时。后处理:降至室温,减压浓缩除去二氧六环后,加入150mL的乙酸乙酯,加入1.2M的稀盐酸150毫升洗有机相,弃去水相,有机相再用5%的氢氧化钠溶液150mL再洗一次,分出有机相浓缩至干,所得油状物使用中性氧化铝进行柱层析纯化(石油醚:乙酸乙酯=3:1)得(R)-3-(3,5-二甲氧苯基)环己烷-1-酮(4.2g,18mmol,无色油状物),收率:36%。
MS m/z(ESI):235[M+1] +
1H NMR(400MHz,CDCl 3):δ6.39(d,J=2.2Hz,2H),6.36(t,J=2.2Hz,1H),3.81(s,6H),3.01–2.90(m,1H),2.64–2.36(m,4H),2.19–2.07(m,2H),1.92–1.70(m,2H)。
第二步:
(R)-5-(3,5-二甲氧苯基)-2-(2-硝基苯甲酰基)环己酮
将(R)-3-(3,5-二甲氧苯基)环己酮(4.0g,18mmol)和四氢呋喃(100mL)混合,氮气保护下降温至-78℃,缓慢滴加二异丙基氨基锂(8.3mL,20mmol),升温在-40℃,搅拌2小时,缓慢加入邻硝基苯甲酰氯(3.5g,18mmol),升至室温搅拌3小时。后处理:用饱和氯化铵水溶液淬灭,有机相减压脱溶,得到(R)-5-(3,5-二甲氧苯基)-2-(2-硝基苯甲酰基)环己酮(8.7g,淡黄色油状物),下步直接使用,未进行进一步纯化。
MS m/z(ESI):384[M+1] +
第三步:
(R)-6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑
将(R)-5-(3,5-二甲氧苯基)-2-(2-硝基苯甲酰)环己酮粗品(8.0g)、水合肼(10mL,80%),乙酸(10mL)和乙醇(100mL)混合,升温至60℃,搅拌3小时。后处理:减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=3:1)纯化,得到(R)-6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑(3.2g,黄色固体),两步收率:50%。
MS m/z(ESI):380[M+1] +
1H NMR(400MHz,CDCl 3):δ7.80(d,J=7.9Hz,1H),7.62–7.57(m,2H),7.46–7.38(m,1H),6.43–6.36(m,3H),3.83(s,6H),3.02–2.92(m,1H),2.87-2.82(m,1H),2.66-2.60(m,1H),2.56-2.53(m,2H),2.13–2.04(m,1H),1.94–1.78(m,1H)。
第四步:
(R)-6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑
将(R)-6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑(2.5g,6.5mmol)溶于50mL乙腈中,降温至-45℃后滴加磺酰氯(2.6g,20mmol),滴加完毕后再于-45℃下反应3小时,加入5mL甲醇淬灭反应。后处理:自然升至室温后浓缩除去溶剂,加入50mL水和100mL乙酸乙酯,分液,有机相减压脱溶,用石油醚/乙酸乙酯(1:1)体系过硅胶层析柱得到(R)-6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑(2g,4.4mmol,类白色固体),产率:86%。
MS m/z(ESI):449[M+1] +
1H NMR(400MHz,CDCl 3):δ7.79-7.77(m,1H),7.63-7.57(m,2H),7.40–7.36(m,1H),6.55(s,1H),4.15-4.06(m,1H),3.95(s,6H),3.43-3.36(m,1H),2.78–2.72(m,1H),2.67–2.49(m,3H),1.85-1.81(m,1H)。
第五步:
(R)-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺
将(R)-6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑(1.5g,3.3mmol)溶于50mL乙酸乙酯中,加入钯碳(50mg,10%),在氢气氛围下室温搅拌10小时;后处理:过滤除去钯碳,滤液浓缩后得产物(R)-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺(1.5g,白色固体),收率:100%。
MS m/z(ESI):419[M+1] +
1H NMR(400MHz,CDCl 3):δ7.34-7.31(m,1H),7.04–7.02(m,1H),6.77–6.70(m,2H),6.55(s,1H),6.01(s,2H),4.17-4.11(m,2H),3.95(s,6H),3.51-3.44(m,1H),2.81-2.64(m,4H).
第六步:
(R)-N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺
将化合物(R)-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺(120mg,0.29mmol)和N,N-二异丙基乙胺(75mg,0.58mmol)溶于无水二氯甲烷(10mL)中,降温至-40℃,缓慢加入丙烯酰氯(26mg,0.29mmol),并于-40℃下反应0.5h。后处理:加入1mL甲醇淬灭反应后,旋干溶剂,用制备液相分离,冻干得目标产物(R)-N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺(85mg,白色固体),产率:62%。
MS m/z(ESI):473[M+1] +
1H NMR(400MHz,DMSO):δ12.96(s,1H),11.48(s,1H),8.49(d,J=7.9Hz,1H),7.59(d,J=7.3Hz,1H),7.32(t,J=7.7Hz,1H),7.17(t,J=7.3Hz,1H),6.89(s,1H),6.33-6.20(m,2H),5.81(d,J=10.8Hz,1H),4.03(br.s,1H),3.93(d,J=3.7Hz,6H),3.49-3.37(m,1H),2.82-2.58(m,4H),1.84(d,J=11.2Hz,1H)。
碱性高效液相纯度:99.59%(214nm),99.70%(254nm)。
ee:96%
实施例015-P2
(S)-N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000055
第一步:
(S)-3-(3,5-二甲氧苯基)环己烷-1-酮
氩气保护下,将3,5-二甲氧基苯硼酸(22.7g,123mmol),乙酰丙酮二(乙烯)铑(260mg,1mmol),(S)-BINAP(935mg,1.5mmol)加入到250mL二氧六环和25mL水的混合溶剂中后,再加入2-环已烯-1-酮(4.8g,50mmol),上述反应液于105℃油浴中反应6小时。后处理:降至室温,减压浓缩除去二氧六环后,加入150mL的乙酸乙酯,再加入1.2M的稀盐酸150mL洗有机相,弃去水相,有机相再用5%的氢氧化钠溶液150mL再洗一次,分出有机相浓缩至干,所得油状物使用中性氧化铝进行柱层析纯化(石油醚:乙酸乙酯=3:1)得(S)-3-(3,5-二甲氧苯基)环己烷-1-酮(3.5g,15mmol,无色油状物)收率:30%。
MS m/z(ESI):235[M+1] +
1H NMS(400MHz,CDCl 3):δ6.39(d,J=2.2Hz,2H),6.36(t,J=2.2Hz,1H),3.81(s,6H),3.01–2.91(m,1H),2.64–2.32(m,4H),2.17–2.07(m,2H),1.87–1.78(m,2H).
第二步:
(S)-5-(3,5-二甲氧苯基)-2-(2-硝基苯甲酰)环己酮
将(S)-3-(3,5-二甲氧苯基)环己酮(3.5g,15mmol)和四氢呋喃(100mL)混合,氮气保护下降温至-78℃,缓慢滴加二异丙基氨基锂(8.3mL,17mmol),升温至-40℃,搅拌2小时,缓慢加入邻硝基苯甲酰氯(3.5g,18mmol),升至室温搅拌3小时。后处理:用饱和氯化铵水溶液淬灭,有机相减压脱溶,得到(S)-5-(3,5-二甲氧苯基)-2-(2-硝基苯甲酰)环己酮(7.4g,淡黄色油状物),下步直接使用,未进行进一步纯化。
MS m/z(ESI):384[M+1] +
第三步:
(S)-6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑
将(S)-5-(3,5-二甲氧苯基)-2-(2-硝基苯甲酰)环己酮粗品(7.0g)、水合肼(10mL,80%),乙酸(10mL)和乙醇(100mL)混合,升温至60℃,搅拌3小时。后处理:减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=3:1)纯化,得到(S)-6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑(2.5g,黄色固体),两步收率:44%。
MS m/z(ESI):380[M+1] +
1H NMS(400MHz,CDCl 3):δ7.73-7.71(m,1H),7.60-7.54(m,2H),7.36–7.34(m,1H),6.41–6.34(m,3H),3.83(s,6H),2.97–2.82(m,1H),2.69–2.60(m,1H),2.52–2.40(m,3H),2.03(s,1H),1.84–1.78(m,1H)。
第四步:
(S)-6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑
将(S)-6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑(2.5g,6.5mmol)溶于50mL乙腈中,降温至-45℃后滴加磺酰氯(2.6g,20mmol),滴加完毕后再于-45℃下反应3小时,加入5mL甲醇淬灭反应。后处理:自然升至室温后浓缩除去溶剂,加入50mL水和100mL乙酸乙酯,分液,有机相减压脱溶,用石油醚/乙酸乙酯(1:1)体系过硅胶层析柱得到(S)-6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑(1.8g,4.0mmol,类白色固体),产率:61%。
MS m/z(ESI):449[M+1] +
1H NMS(400MHz,CDCl 3):δ7.86–7.80(m,1H),7.62–7.60(m,2H),7.46–7.41(m,1H),6.55(s,1H),4.12–4.10(m,1H),3.96(d,J=1.2Hz,6H),3.52–3.45(m,1H),2.82–2.64(m,2H),2.61–2.51(m,2H),1.87–1.83(m,1H)。
第五步:
(S)-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺
将(S)-6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吲唑(1.5g,3.3mmol)溶于50mL乙酸乙酯中,加入Pd/C(50mg),在氢气氛围下室温搅拌10小时;后处理:过滤除去钯碳,滤液浓缩后用纯乙酸乙酯过硅胶层析柱得到产物(S)-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺(0.8g,白色固体)。收率:57%
MS m/z(ESI):419[M+1] +
1H NMS(400MHz,CDCl 3):δ7.34–7.32(m,1H),7.28(s,1H),6.80–6.71(m,2H),6.55(s,1H),4.20–4.07(m,1H),3.95(s,6H),3.52–3.45(m,1H),2.82–2.66(m,4H),1.95–1.86(m,1H)。
第六步:
(S)-N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺
将化合物(S)-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺(120mg,0.29mmol) 和N,N-二异丙基乙胺(75mg,0.58mmol)溶于无水二氯甲烷(10mL)中,降温至-40℃,缓慢加入丙烯酰氯(26mg,0.29mmol),并于-40℃下反应0.5h。后处理:加入1ml甲醇淬灭反应后,旋干溶剂,用制备液相分离,冻干得目标产物(S)-N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺(90mg,白色固体),产率:65%。
MS m/z(ESI):473[M+1] +
1H NMS(400MHz,DMSO):δ12.96(s,1H),11.48(s,1H),8.49(d,J=8.1Hz,1H),7.59(d,J=7.4Hz,1H),7.32(t,J=7.6Hz,1H),7.17(t,J=7.4Hz,1H),6.89(s,1H),6.41–6.16(m,2H),5.81(d,J=9.6Hz,1H),4.03(br.s,1H),3.93(d,J=3.7Hz,6H),3.50-3.37(m,1H),2.86–2.59(m,4H),1.84(d,J=11.4Hz,1H)。
碱性液相纯度:99.59%(214nm),99.70%(254nm)。
ee:95%
实施例095
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶-3-基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000056
第一步:
4-(2-硝基苯甲酰)-3-羰基哌啶-1-羧酸叔丁基酯
将化合物3-羰基哌啶-1-羧酸叔-丁基酯095a(20g,100.5mmol,1eq),溶于400mL四氢呋喃中, 控温-78℃加入二异丙基氨基锂(60.3ml,120.6mmol,1.2eq),滴加完毕后升温到-40℃反应1小时,然后在-40℃向体系中加入2-硝基苯甲酰氯(18.6g,100.5mmol,1eq),升至室温反应三小时。后处理:加入饱和氯化铵的水溶液(400ml),乙酸乙酯(200ml)萃取三次,有机相减压脱溶得到目标粗产物4-(2-硝基苯甲酰)-3-羰基哌啶-1-羧酸叔丁基酯095b(42g,黄色油状物)产率90%。
第二步:
3-(2-硝基苯基)-1,4,5,7-四氢-6H-吡唑并[3,4-c]吡啶-6-羧酸叔丁基酯
将化合物4-(2-硝基苯甲酰)-3-羰基哌啶-1-羧酸叔丁基酯095b(40g,115mmol,1eq),水合肼(14.4g,287.5mmol,2.5eq)溶于乙醇/醋酸10:1(200mL)中,在65℃下反应3小时。后处理:减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=2:1)纯化,得目标产物3-(2-硝基苯基)-1,4,5,7-四氢-6H-吡唑并[3,4-c]吡啶-6-羧酸叔丁基酯095c(15.3g,黄色固体),产率:39%。
MS m/z(ESI):345[M+1] +
第三步:
1-(4-甲氧苄基)-3-(2-硝基苯基)-1,4,5,7-四氢-6H-吡唑并[3,4-c]吡啶-6-羧酸叔丁基酯
将化合物3-(2-硝基苯基)-1,4,5,7-四氢-6H-吡唑并[3,4-c]吡啶-6-羧酸叔丁基酯095c(4g,11.6mmol,1eq)溶于DMF(120mL)中,在0℃条件下缓慢滴加氢化钠(696mg,17.4mmol,1.5eq),搅拌0.5h,加入对甲氧基氯苄(2.7g,17.4mmol,1.5eq),继续搅拌3h。后处理:加100mL水淬灭反应,用乙酸乙酯萃取,有机相减压脱溶残余物经硅胶柱层析(石油醚:乙酸乙酯=3:1)纯化,得到目标产物1-(4-甲氧苄基)-3-(2-硝基苯基)-1,4,5,7-四氢-6H-吡唑并[3,4-c]吡啶-6-羧酸叔丁基酯095d(7g,棕色固体),产率90%。
MS m/z(ESI):445[M+1] +
第四步:
1-(4-甲氧苄基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶
将化合物1-(4-甲氧苄基)-3-(2-硝基苯基)-1,4,5,7-四氢-6H-吡唑并[3,4-c]吡啶-6-羧酸叔丁基酯095d(7g,15.7mmol,1eq)加入到二氯甲烷(150ml)中,然后0-10℃加入三氟乙酸溶液(50ml)在该温度下搅拌1小时。后处理:减压脱溶,残余物经硅胶柱层析(二氯甲烷:甲醇=10:1)纯化,得到目标化合物1-(4-甲氧苄基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶095e(3.2g,棕色固体),产率:59%。
MS m/z(ESI):345[M+1] +
第五步:
6-(3,5-二甲氧苯基)-1-(4-甲氧苄基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶
将化合物1-(4-甲氧苄基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶095e(3g,8.7mmol,1eq)加入到甲苯(100ml)中,加入1-溴-3,5-二甲氧基苯(2.8g,13.05mmol,1.5eq),三(二亚苄基 丙酮)二钯(1.6g,1.74mmol,0.2eq),联萘二苯磷(2.16g,3.48mmol,0.4eq),叔丁醇钠(3.46g,36.1mmol,3eq),氮气保护下反应6小时。后处理:减压脱溶,残余物经硅胶柱层析(二氯甲烷:甲醇=10:1)纯化,得到目标化合物6-(3,5-二甲氧苯基)-1-(4-甲氧苄基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶095f(1.38mg,黄色固体),产率31%。
MS m/z(ESI):501[M+1] +
第六步:
6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶
将化合物6-(3,5-二甲氧苯基)-1-(4-甲氧苄基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶095f(1.3g,2.6mmol,1eq)加入到三氟乙酸(50ml)中,然后升温80℃回流反应3小时。后处理:减压脱溶,残余物经硅胶柱层析(二氯甲烷:甲醇=10:1)纯化,得到目标化合物6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶095g(400mg,棕色固体),产率:40%。
MS m/z(ESI):401[M+1] +
第七步:
6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶
将化合物6-(3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶095g(250mg,0.66mmol,1eq)加入到乙腈(20ml)中,然后-40℃加入磺酰氯(240.6mg,1.78mmol,2.7eq)在该温度下搅拌1小时。后处理:加入30ml水淬灭,乙酸乙酯50ml萃取,有机相减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=1:1)纯化,得到目标化合物6-(2,6-二氯-3,5-二甲氧苯基)-3-(2-硝基苯基)-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶095h(150mg,浅黄色固体),产率:50%。
第八步和第九步参照实例289步骤合成。最终得到目标产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吡唑并[3,4-c]吡啶-3-基)苯基)丙烯酰胺。
MS m/z(ESI):474[M+1] +
1H NMR(400MHz,DMSO):δ12.80(s,1H),10.58(s,1H),8.29(s,1H),6.99(s,1H),6.86(s,1H),6.54(s,2H),6.18(s,3H),5.80(s,1H),3.74(s,6H),3.45(s,2H),2.84(s,2H),1.98(s,2H)。
实施例281
N-(5-(6-(2,6-二氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1-甲基-1H-吡唑-4-基)丙烯酰胺
Figure PCTCN2018104007-appb-000057
将6-(3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑(2g,5.2mmol)和乙腈(50ml)混合,0℃下加入select F(3.5g,10mmol),缓慢升温至室温,搅拌过夜。后处理:减压脱溶,残余物加入二氯甲烷(100mL)、水(100mL)萃取,有机相减压脱溶,残余物用硅胶柱层析(石油醚:乙酸乙酯=5:4)纯化,得到6-(2,6-二氟-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑(800mg,1.9mmol,黄色固体),产率:19%。
参照实例093的操作步骤合成实例281。得到最终产物N-(5-(6-(2,6-二氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1-甲基-1H-吡唑-4-基)丙烯酰胺。再通过手性柱拆分得到P1和P2。
手性拆分条件:设备SFC,色谱柱:chiralpak-AD流动相:CO2-IPA(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000058
MS m/z(ESI):443.8[M+1] +
P1: 1H NMR(400MHz,DMSO):δ12.92(s,1H),9.42(s,1H),7.84(s,1H),6.93(t,J=8.3Hz,1H),6.53(dd,J=17.0,10.2Hz,1H),6.19(dd,J=17.0,2.1Hz,1H),5.66(dd,J=10.2,2.1Hz,1H),3.86(s,6H),3.76(s,3H),3.43-3.36(m,1H),2.98–2.92(m,2H),2.47-2.38(m,1H),2.13–2.05(m,1H),1.90(d,J=12.4Hz,1H)。
e.e.99.0%
P2: 1H NMR(400MHz,DMSO):δ12.92(s,1H),9.42(s,1H),7.84(s,1H),6.93(t,J=8.3Hz,1H),6.53(dd,J=17.0,10.2Hz,1H),6.19(dd,J=17.0,2.1Hz,1H),5.66(dd,J=10.2,2.1Hz,1H),3.86(s,6H),3.76(s,3H),3.43-3.36(m,1H),2.98–2.92(m,2H),2.47-2.38(m,1H),2.13–2.05(m,1H),1.90(d,J=12.4Hz,1H)。
e.e.99.8%
实施例283
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-(4-甲基哌嗪-1-基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000059
参照实例137步骤合成实例283,但在第5步用1-甲基哌嗪取代N-N’二甲基-N-甲基乙二胺。最 终得到目标产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-(4-甲基哌嗪-1-基)苯基)丙烯酰胺。
最终产物(消旋体)分离条件:设备质谱引导的prep-HPLC;色谱柱:-Gemini-C18 150x 21.2mm,5um,流动相:ACN--H2O(0.05%NH3),梯度:60-80。
手性拆分条件:设备SFC,色谱柱:chiralpak-AS,流动相:CO2-ETOH(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000060
P1:MS m/z(ESI):569.9[M+1]
ee:96.995%
1H NMR(400MHz,MeOD):δ8.01(d,J=8.4Hz,1H),7.04(s,1H),6.94(d,J=8.4Hz,1H),6.67(s,1H),6.30–6.19(m,2H),5.67(d,J=9.3Hz,1H),4.53(s,2H),4.07(s,1H),3.83(d,J=3.4Hz,6H),3.25(s,5H),2.95(s,4H),2.65(dd,J=21.0,14.8Hz,3H),2.57(s,3H)。
P2:MS m/z(ESI):569.8[M+1]
ee:97.5503%
1H NMR(400MHz,MeOD):δ8.00(d,J=7.9Hz,1H),7.03(s,1H),6.94(d,J=9.1Hz,1H),6.67(s,1H),6.24(d,J=9.4Hz,2H),5.66(d,J=9.1Hz,1H),4.52(s,2H),4.07(s,1H),3.83(d,J=3.6Hz,6H),3.25(s,5H),2.92(d,J=8.9Hz,4H),2.68(dd,J=15.9,6.1Hz,2H),2.60(s,1H),2.53(s,3H)。
实施例284
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-5-(二甲氨基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000061
参照实例133合成实例284,但在第5步用二甲胺盐酸盐取代N-N’二甲基-N-甲基乙二胺。
手性拆分条件:设备SFC,色谱柱:chiralpak-IC,流动相:CO2-ETOH(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000062
MS m/z(ESI):516.4[M+1] +
P1: 1H NMR(400MHz,DMSO):δ12.73(s,1H),11.67(s,1H),8.08(s,1H),7.40(s,1H),7.23–7.15(m,1H),6.89(s,1H),6.54(d,J=7.4Hz,1H),6.26(s,1H),5.80(s,1H),3.93(d,J=3.7Hz,6H),2.94(s,6H),2.73(s,3H),2.00(d,J=7.5Hz,2H),1.82(s,2H).
P2: 1H NMR(400MHz,DMSO):δ12.74(s,1H),11.67(s,1H),8.09(s,1H),7.41(d,J=8.5Hz,1H),7.23–7.17(m,1H),6.89(s,1H),6.54(d,J=8.1Hz,1H),6.26(s,1H),5.81(s,1H),3.93(d,J=3.7Hz,6H),2.94(s,6H),2.73(s,3H),2.00(dd,J=14.4,6.8Hz,2H),1.82(s,2H).
实施例285
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-(二甲氨基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000063
参照实例137的操作步骤合成实例285。但在第5步用二甲胺盐酸盐取代N-N’二甲基-N-甲基乙二胺。再通过手性柱拆分得到P1,P2。
手性拆分条件:设备SFC,色谱柱:chiralpak-IC流动相:HEX-EtOH(DEA)
在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000064
MS m/z(ESI):514.7[M+1] +
P1: 1H NMR(400MHz,DMSO):δ12.90(s,1H),10.96(s,1H),8.22(s,1H),6.88(s,1H),6.83(s,1H),6.74(dd,J=9.1,2.8Hz,1H),6.41-6.25(m,1H),6.21-6.16(m,1H),5.71(dd,J=10.0,1.6Hz,1H),3.93(d,J=3.8Hz,6H),2.91(s,6H),2.84–2.58(m,5H),2.51(q,J=2Hz,2H)。
P2: 1H NMR(400MHz,DMSO):δ12.90(s,1H),10.96(s,1H),8.22(s,1H),6.88(s,1H),6.83(s,1H),6.74(dd,J=9.1,2.8Hz,1H),6.41-6.25(m,1H),6.21-6.16(m,1H),5.71(dd,J=10.0,1.6Hz,1H), 3.93(d,J=3.8Hz,6H),2.91(s,6H),2.84–2.58(m,5H),2.51(q,J=2Hz,2H)
ee:95.9%
实施例286
N-(2-(6-(2,6-二氯-3,5-二甲氧基苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-甲基苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000065
参照实例289合成实例286,但在第一步将4-氯-2-硝基苯甲酸取代成5-甲基-2-硝基苯甲酸。最终得到目标产物N-(2-(6-(2,6-二氯-3,5-二甲氧基苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-甲基苯基)丙烯酰胺。
MS m/z(ESI):486.1[M+1] +
1H NMR(400MHz,CDCl3):δ10.86(s,1H),8.52(d,J=8.3Hz,1H),7.31(s,1H),7.15(d,J=8.4Hz,1H),6.54(s,1H),6.32(m,2H),5.69(dd,J=10.0,1.4Hz,1H),4.20-4.14(m,1H),3.89-3.56(m,7H),3.66–3.54(m,1H),2.87–2.72(m,4H),2.35(s,3H)。
实施例287
N-(4-氯-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000066
参照实例289合成实例287,但在第一步将4-氯-2-硝基苯甲酸替换成5-氯-2-硝基苯甲酸。最终得到目标产物N-(4-氯-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺。
通过手性拆分得到两个异构体P1(27mg),P2(25mg).
手性拆分条件:设备SFC,色谱柱:chiralpak-OD,流动相:CO2-ETOH(DEA)。
在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000067
P1: 1H NMR(400MHz,CDCl3):δ11.26(s,1H),8.68(d,J=8.9Hz,1H),7.50(d,J=2.5Hz,1H),7.32–7.24(m,2H),6.54(s,1H),6.37-6.22(m,2H),5.73–5.70(m,1H),4.26–4.10(m,1H),3.94(s,3H),3.93(s,3H),3.63–3.56(m,1H),2.87–2.76(m,4H),1.98–1.95(m,1H).
P2: 1H NMR(400MHz,CDCl3):δ11.29(s,1H),8.67(d,J=8.9Hz,1H),7.50(d,J=2.5Hz,1H),7.29–7.24(m,1H),6.54(s,1H),6.37–6.21(m,2H),5.73–5.70(m,1H),4.22–4.12(m,1H),3.94(s,3H),3.93(s,3H),3.62–3.55(m,1H),2.91–2.72(m,4H),2.01–1.91(m,1H).
ee:99.1%
实施例288
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-5-甲基苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000068
参照实例289合成实例288,但在第一步用4-甲基-2-硝基苯甲酸取代4-氯-2-硝基苯甲酸。最终得到目标产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-5-甲基苯基)丙烯酰胺。
手性拆分条件:设备Chiral prepHPLC,色谱柱:chiralpak-IC,流动相:CO2-ETOH(DEA)。在机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000069
MS m/z(ESI):487.4[M+1] +
P1: 1H NMR(400MHz,DMSO):δ12.91(s,1H),11.48(s,1H),8.35(s,1H),7.48(d,J=7.8Hz,1H),6.99(d,J=8.2Hz,1H),6.89(s,1H),6.34–6.22(m,2H),5.81(d,J=9.4Hz,1H),4.02(s,1H),3.93(d,J=3.7Hz,6H),3.53–3.35(m,2H),2.74(t,J=17.6Hz,3H),2.34(s,3H),1.84(d,J=10.8Hz,1H)。
P2: 1H NMR(400MHz,DMSO):δ12.91(s,1H),11.48(s,1H),8.35(s,1H),7.48(d,J=8.0Hz,1H),7.00(d,J=7.6Hz,1H),6.89(s,1H),6.34–6.22(m,2H),5.81(d,J=9.2Hz,1H),4.02(s,1H),3.93(d,J=3.7Hz,6H),3.47–3.36(m,2H),2.76(d,J=15.3Hz,3H),2.34(s,3H),1.84(d,J=11.5Hz,1H)。
实施例289
N-(5-氯-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000070
第一步:
4-氯-2-硝基苯甲酰氯
将化合物4-氯-2-硝基苯甲酸289a(18g,89.3mmol,1eq)加入到二氯亚砜(150ml)溶液中,升温到80℃反应过夜。后处理:直接将反应体系中的残余的二氯亚砜溶液旋干得目标粗产物4-氯-2-硝基苯甲酰氯289b(20g,黄色油状物)直接用于下一步反应,产率90%。
MS m/z(ESI):221[M+1] +
第二步:
2-(4-氯-2-硝基苯甲酰)-5-(3,5-二甲氧苯基)环己烷-1-酮
将3-(3,5-二甲氧苯基)环己烷-1-酮(4.5g,19.2mmol,1eq)溶于50mL四氢呋喃中,控温-78℃加入二异丙基氨基锂(11.52ml,23.04mmol,1.2eq),滴加完毕后升温到-40℃反应1小时,然后在-40℃向体系中加入4-氯-2-硝基苯甲酰氯289b(4.2g,19.2mmol,1eq),升至室温反应三小时。后处理:加入饱和氯化铵的水溶液(50ml),乙酸乙酯(50ml)萃取三次,有机相减压脱溶得到目标粗产物2-(4-氯-2-硝基苯甲酰)-5-(3,5-二甲氧苯基)环己烷-1-酮289c(3.8g,黄色固体)产率47.5%。
第三步和第四步参照实例010步骤合成,得到目标产物3-(4-氯-2-硝基苯基)-6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑289e。
第五步:
5-氯-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺
将化合物3-(4-氯-2-硝基苯基)-6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑289e(200mg,0.41mmol,1eq)溶于乙酸乙酯(30mL)中,加入钯碳(150mg,75%)通入氢气,室温反应3h。后处理:抽滤除去钯碳,用乙酸乙酯洗3次,旋干得目标产物5-氯-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺289f(100mg,黄色固体),产率:54%。
MS m/z(ESI):453.8[M+1] +
第六步:
N-(5-氯-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺
将化合物5-氯-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯胺289f(100mg,0.22mmol,1eq)和N,N-二异丙基乙胺(85mg,0.66mmol,3eq)溶于无水二氯甲烷(30mL)中,降温至-40℃,缓慢加入丙烯酰氯(18mg,0.198mmol,0.9eq)的二氯甲烷溶液,反应0.5h。后处理:旋干溶剂,用制备液相分离,冻干得目标产物N-(5-氯-2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)苯基)丙烯酰胺(5.8mg,白色固体)。产率:14%。
手性拆分条件:设备Chiral prepHPLC,色谱柱:chiralpak-IC,流动相:CO2-ETOH(DEA)。在机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000071
MS m/z(ESI):507[M+1] +
P1: 1H NMR(400MHz,DMSO):δ13.06(s,1H),11.69(s,1H),8.62(s,1H),7.62(d,J=8.1Hz,1H),7.24(d,J=5.3Hz,1H),7.23(s,1H),6.90(s,1H),6.34–6.25(m,2H),5.87(d,J=11.0Hz,1H),4.03(s,1H),3.93(d,J=3.6Hz,6H),3.49–3.39(m,1H),2.78(d,J=15.8Hz,3H),2.65(d,J=12.6Hz,1H),1.85(d,J=10.8Hz,1H)。
P2: 1H NMR(400MHz,DMSO):δ13.07(s,1H),11.69(s,1H),8.62(s,1H),7.62(d,J=8.4Hz,1H),7.24(d,J=8.2Hz,1H),6.89(s,1H),6.31(t,J=5.9Hz,2H),5.88(s,1H),4.07–3.98(m,1H),3.93(d,J=3.6Hz,6H),3.48–3.39(m,1H),2.78(d,J=15.4Hz,4H),1.85(d,J=11.6Hz,1H)。
实施例291
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-5-氟苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000072
参照实例289步骤合成实例291,但在第一步用4-氟-2-硝基苯甲酸取代4-氯-2-硝基苯甲酸。最 终得到目标产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-5-氟苯基)丙烯酰胺。
手性拆分条件:设备Chiral prep-HPLC,色谱柱:chiralpak-AD,流动相:HEX-ETOH(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000073
P1:MS m/z(ESI):489.8[M+1] +
ee:100%
1H NMR(400MHz,DMSO):δ13.01(s,1H),11.74(s,1H),8.39(d,J=13.0Hz,1H),7.63(s,1H),7.02(s,1H),6.89(s,1H),6.30(d,J=2.6Hz,2H),5.87(d,J=7.7Hz,1H),4.02(s,1H),3.93(d,J=3.6Hz,6H),3.47–3.39(m,1H),2.82–2.61(m,4H),1.83(s,1H)。
P2:MS m/z(ESI):489.9[M+1] +
ee:100%
1H NMR(400MHz,MeOD)δ8.33(s,1H),7.60(s,1H),6.94(s,1H),6.77(s,1H),6.36(d,J=4.8Hz,2H),5.87–5.78(m,1H),4.16(s,1H),3.93(d,J=3.7Hz,6H),3.56(dd,J=15.8,12.2Hz,1H),2.78(dd,J=15.9,5.5Hz,4H),1.88(s,1H)。
实施例292
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-氟苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000074
参照实例291的操作步骤合成实例292。但在第1步用5-氟-2-硝基苯甲酸代替4-氟-2-硝基苯甲酸。最终得到目标产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-氟苯基)丙烯酰胺292。再通过手性柱拆分得到P1和P2。
手性拆分条件:设备SFC,色谱柱:chiralpak-IC流动相:HEX-EtOH(DEA)在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000075
MS m/z(ESI):490[M+1] +
P1: 1H NMR(400MHz,MeOD):δ8.83(s,1H),7.68(d,J=9.7Hz,1H),7.44(t,J=7.6Hz,1H),7.14(s,1H),6.75–6.61(m,2H),6.13(dd,J=7.6,2.8Hz,1H),4.47(s,1H),4.27(d,J=3.7Hz,6H),3.95–3.81(m,1H),3.13(s,4H),2.23(d,J=9.2Hz,1H)。
P2: 1H NMR(400MHz,MeOD):δ8.83(s,1H),7.68(d,J=9.7Hz,1H),7.44(t,J=7.6Hz,1H),7.14(s,1H),6.75–6.61(m,2H),6.13(dd,J=7.6,2.8Hz,1H),4.47(s,1H),4.27(d,J=3.7Hz,6H),3.95–3.81(m,1H),3.13(s,4H),2.23(d,J=9.2Hz,1H)。
实施例293
Figure PCTCN2018104007-appb-000076
第一步:
1-氰基环丙烷-1-甲酰氯
将化合物293a(2g,18mmol,1eq),溶于二氯亚砜(10ml)中,油浴加热到80℃,搅拌2h,降至室温。后处理:减压浓缩体系,得到粗品产物。1-氰基环丙烷-1-甲酰氯293b(1.9g,黄色油状物)产率83%。
第二步:
1-(4-(3,5-二甲氧苯基)-2-羰基环己烷-1-羰基)环丙烷-1-甲腈
将化合物3-(3,5-二甲氧苯基)环己烷-1-酮(3.4g,14mmol,1eq),溶于50mL四氢呋喃中,控温-78℃加入二异丙基氨基锂(8.4ml,16.8mmol,1.2eq),滴加完毕后升温到-40℃反应1小时,然后在-40℃向体系中加入1-氰基环丙烷-1-甲酰氯293b(1.8g,14mmol,1eq),升至室温反应三小时。后处理:加入饱和氯化铵的水溶液(30ml),乙酸乙酯(100ml)萃取三次,有机相减压脱溶得到目标粗产物1-(4-(3,5-二甲氧苯基)-2-羰基环己烷-1-羰基)环丙烷-1-甲腈293c(2.6g,黄色油状物)产率55%。
第三步:
1-(6-(3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)环丙烷-1-甲腈
将化合物1-(4-(3,5-二甲氧苯基)-2-羰基环己烷-1-羰基)环丙烷-1-甲腈293c(2.6g,8mmol,1eq),水合肼(1g,20mmol,2.5eq)溶于乙醇/醋酸10:1(20mL)中,在65℃下反应3小时。后处理:减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=2:1)纯化,得目标产物1-(6-(3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)环丙烷-1-甲腈293d(1.1g,黄色固体),产率:43%。
MS m/z(ESI):324.2[M+1] +
第四步:
1-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)环丙烷-1-甲腈
将化合物1-(6-(3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)环丙烷-1-甲腈293d(1.0g,3mmol,1eq),溶于ACN,-40℃搅拌,滴加二氯化砜。-40℃继续搅拌3h,反应结束,升至室温。后处理:减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=2:1)纯化,得目标产物1-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)环丙烷-1-甲腈293e(0.4g,黄色固体),产率:33%。
MS m/z(ESI):392.2[M+1] +
第五步:
(1-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)环丙基)甲胺
将化合物1-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)环丙烷-1-甲腈293e(0.4g,1mmol,1eq)和BH3(2ml,2mmol,2eq),溶于THF,封管60℃搅拌3h,后加入甲醇,60℃继续搅拌3h,反应结束。后处理:减压脱溶,残余物经硅胶柱层析(石油醚:乙酸乙酯=2:1)纯化,得目标产物(1-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)环丙基)甲胺293f(120mg,黄色固体),产率:30%。
MS m/z(ESI):396.1[M+1] +
第六步:
N-((1-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)环丙基)甲基)丙烯酰胺
将化合物(1-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)环丙基)甲胺293f(120mg,0.3mmol,1eq)和DIEA(116mg,0.9mmol,3eq),溶于DCM,-40℃搅拌,后滴加丙烯酰氯(28mg,0.3mmol,1eq),-40℃搅拌0.5h,反应结束。后处理:送制备。目标产物N-((1-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)环丙基)甲基)丙烯酰胺。
最终产物(消旋体)分离条件:设备质谱引导的prep-HPLC;色谱柱:-Gemini-C18 150x 21.2mm,5um,流动相:ACN--H2O(0.05%NH3),梯度:60-80。
手性拆分条件:设备SFC,色谱柱:chiralpak-AS,流动相:CO2-ETOH(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000077
P1: 1H NMR(400MHz,DMSO):δ12.23–12.18(m,1H),8.12–8.02(m,2H),6.86(s,1H),6.30–6.23(m,1H),6.06(dd,J=17.0,2.1Hz,1H),5.56(d,J=11.9Hz,1H),3.92(d,J=2.2Hz,7H),3.31(s,2H),2.68(s,1H),2.58(s,1H),1.75–1.70(m,1H),1.24(s,1H),0.91–0.65(m,6H)。
P2: 1H NMR(400MHz,DMSO):δ12.23–12.18(m,1H),8.12–8.02(m,2H),6.86(s,1H),6.30–6.23(m,1H),6.06(dd,J=17.0,2.1Hz,1H),5.56(d,J=11.9Hz,1H),3.92(d,J=2.2Hz,7H),3.31(s,2H),2.68(s,1H),2.58(s,1H),1.75–1.70(m,1H),1.24(s,1H),0.91–0.65(m,6H)。
MS m/z(ESI):450.1[M+1] +
实施例295
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-吗啉代苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000078
参照实例137步骤合成实例295,但在第5步用吗啉取代N1,N1,N2-三甲基乙烷-1,2-二胺。最终得到目标产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-吗啉代苯基)丙烯酰胺。
手性拆分条件:设备Chiral prep-HPLC,色谱柱:chiralpak-AD,流动相:HEX-ETOH(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000079
P1:MS m/z(ESI):557.0[M+1] +
ee:100%(214nm);100%(254nm)
1H NMR(400MHz,DMSO):δ12.93(s,1H),11.08(s,1H),8.30(s,1H),7.04(s,1H),6.95(d,J=8.3Hz,1H),6.89(s,1H),6.30(s,1H),6.22(s,1H),5.76(s,1H),4.03(s,1H),3.93(d,J=3.7Hz,6H),3.76(t,J=4.4Hz,4H),3.12(d,J=3.5Hz,4H),2.94–2.53(m,5H),1.80(s,1H)。
P2:MS m/z(ESI):557.0[M+1]
ee:98.128%(214nm);98.596%(254nm)
1H NMR(400MHz,DMSO):δ12.93(s,1H),11.09(s,1H),8.33(s,1H),7.01(d,J=41.8Hz,2H),6.89(s,1H),6.28(s,1H),6.22(s,1H),5.75(d,J=9.5Hz,1H),4.04(s,1H),3.93(d,J=3.2Hz,6H),3.75(s,4H),3.12(s,4H),2.76(d,J=20.8Hz,5H),1.82(s,1H)。
实施例296
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5-二氢-1H-吲唑-3-基)-5-氟苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000080
合成291过程中,分离得到的副产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5-二氢-1H-吲唑-3-基)-5-氟苯基)丙烯酰胺。
MS m/z(ESI):487.8[M+1] +
HPLC:91.273%(214nm);94.245%(254nm)
1H NMR(400MHz,DMSO):δ13.26(s,1H),12.80(s,1H),11.44(s,1H),9.71(s,1H),8.33(s,1H),7.55(s,2H),7.07(s,1H),6.92(s,1H),6.40(s,2H),6.28(d,J=16.9Hz,1H),5.84(s,1H),3.94(s,6H),2.95(s,1H),2.44–2.27(m,1H)。
实施例297
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5-二氢-1H-吲唑-3-基)-4-氟苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000081
合成292过程中,分离得到的副产物产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5-二氢-1H-吲唑-3-基)-4-氟苯基)丙烯酰胺。
MS m/z(ESI):488.0[M+1] +
1H NMR(400MHz,MeOD):δ8.36(s,1H),7.76(s,1H),7.28(s,1H),7.13(s,1H),6.81(s,1H),6.38(s,3H),5.80(d,J=8.4Hz,1H),3.96(s,6H),3.00(d,J=9.7Hz,1H),2.62(s,1H).
实施例298
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5-二氢-1H-吲唑-3-基)-5-甲基苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000082
合成288过程中,分离得到的副产物产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5-二氢-1H-吲唑-3-基)-5-甲基苯基)丙烯酰胺。
MS m/z(ESI):485.4[M+1] +
1H NMR(300MHz,DMSO)δ12.83(s,2H),11.08(s,1H),8.28(s,1H),7.42(d,J=35.7Hz,2H),7.18–6.81(m,4H),6.34(s,4H),6.18(d,J=16.7Hz,2H),5.70(d,J=8.6Hz,2H),3.92(s,12H),2.47(s,8H),2.34(s,6H)。
实施例299
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5-二氢-1H-吲唑-3-基)-4-(二甲氨基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000083
合成285过程中,分离得到的副产物产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5-二氢-1H-吲唑-3-基)-4-(二甲氨基)苯基)丙烯酰胺。
MS m/z(ESI):512.7[M+1] +
1H NMR(400MHz,DMSO):δ12.90(s,1H),10.96(s,1H),8.22(s,1H),6.88(s,1H),6.83(s,1H),6.74(dd,J=9.1,2.8Hz,1H),6.41–6.25(m,1H),6.18(dd,J=17.0,1.7Hz,1H),5.75–5.69(m,1H),3.93(d,J=3.8Hz,6H),2.91(s,6H),2.84–2.58(m,5H),2.51(dt,J=3.5,1.7Hz,2H)。
实施例300
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5-二氢-1H-吲唑-3-基)-4-吗啉代苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000084
合成295过程中,分离得到的副产物产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5-二氢-1H-吲唑-3-基)-4-吗啉代苯基)丙烯酰胺。
MS m/z(ESI):555.0[M+1] +
HPLC:82.029%(214nm);82.027%(254nm)
1H NMR(400MHz,DMSO):δ12.95(s,1H),7.95–7.68(m,1H),7.00(d,J=8.5Hz,2H),6.91(t,J=12.6Hz,2H),6.40(d,J=24.2Hz,2H),6.19(d,J=17.0Hz,1H),5.70(d,J=9.8Hz,1H),3.94(s,6H),3.75(s,4H),3.14(s,4H),2.71(d,J=24.4Hz,2H),2.41(s,2H)。
实施例301
N-(2-(6-(2,6-二氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-(4-甲基哌嗪-1-基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000085
6-(2,6-二氟-3,5-二甲氧苯基)-3-(5-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑
将6-(3,5-二甲氧苯基)-3-(5-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑(3.33g,8.13mmol)和乙腈(100ml)混合,0℃下加入select F(5.93g,16.26mmol),缓慢升温至室温,搅拌2h。后处理:减压脱溶,残余物加入二氯甲烷(100mL)、水(100mL)萃取,有机相减压脱溶,残余物用硅胶柱层析(石油醚:乙酸乙酯=5:1)纯化,得到6-(2,6-二氟-3,5-二甲氧苯基)-3-(5-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑(700mg,1.61mmol,黄色固体),产率:19%。
参照实例283的操作步骤合成实例301。但在第4步用6-(2,6-二氟-3,5-二甲氧苯基)-3-(5-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑代替6-(2,6-二氯-3,5-二甲氧苯基)-3-(5-氟-2-硝基苯基)-4,5,6,7-四氢-1H-吲唑。最终得到目标产物N-(2-(6-(2,6-二氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-4-(4-甲基哌嗪-1-基)苯基)丙烯酰胺301。再通过手性柱拆分得到P1和P2。手性拆分条件:设备SFC,色谱柱:chiralpak-AD流动相:CO2-IPA(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000086
P1:MS m/z(ESI):538.1[M+1] +
ee:100%
1H NMR(400MHz,DMSO):δ12.91(s,1H),11.08(s,1H),8.29(s,1H),7.02(s,1H),6.94(d,J=8.3Hz,2H),6.30(s,1H),6.19(d,J=17.0Hz,1H),5.73(d,J=10.1Hz,1H),3.86(s,6H),3.37(s,2H),3.14(s,4H),2.92(s,2H),2.67(s,1H),2.46(s,4H),2.22(s,3H),1.99(s,2H)。
P2:MS m/z(ESI):538.2[M+1] +
ee:97.8%
1H NMR(400MHz,DMSO):δ12.90(s,1H),11.08(s,1H),8.29(s,1H),7.02(s,1H),6.93(t,J=8.0Hz,2H),6.30(s,1H),6.19(d,J=16.9Hz,1H),5.73(d,J=10.1Hz,1H),3.86(s,6H),3.37(s,2H),3.14(s,4H),2.94(d,J=9.5Hz,2H),2.68(s,1H),2.46(s,4H),2.22(s,3H),2.08(s,1H),1.99(s,1H)。
实施例302
N-(5-(6-(2-氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1-甲基-1H-吡唑-4-基)丙烯酰胺
Figure PCTCN2018104007-appb-000087
6-(2-氟-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑
将6-(3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑(1.7g,4.4mmol)和乙腈(50ml)混合,0℃下加入select F(1.7g,4.4mmol),缓慢升温至室温,搅拌2h。后处理:减压脱溶,残余物加入二氯甲烷(50mL)、水(50mL)萃取,有机相减压脱溶,残余物用硅胶柱层析(石油醚:乙酸乙酯=5:4)纯化,得到6-(2-氟-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑(800mg,1.9mmol,黄色固体),产率:47%。
参照实例140的操作步骤合成实例302。但在第4步用6-(2-氟-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑代替6-(2,6-二氯-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑。最终得到目标产物N-(5-(6-(2-氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1-甲基-1H-吡唑-4-基)丙烯酰胺302。再通过手性柱拆分得到P1和P2。手性拆分条件:设备SFC,色谱柱:chiralpak-AD流动相:CO2-IPA(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000088
MS m/z(ESI):426.0[M+1] +
P1: 1H NMR(400MHz,DMSO)δ12.91(s,1H),9.41(s,1H),7.81(s,1H),6.61(dd,J=7.0,2.8Hz,1H),6.56–6.46(m,1H),6.18(dd,J=17.0,2.0Hz,1H),5.70–5.62(m,1H),3.83(s,3H),3.77(s,3H),3.75(s,3H),3.32–3.20(m,1H),2.96–2.72(m,1H),2.48–2.30(m,1H),1.89(s,1H)。
P2: 1H NMR(400MHz,DMSO)δ12.91(s,1H),9.41(s,1H),7.81(s,1H),6.61(dd,J=7.0,2.8Hz, 1H),6.56–6.46(m,1H),6.19(dd,J=17.0,2.0Hz,1H),5.66(dd,J=10.2,1.8Hz,1H),3.83(s,3H),3.77(s,3H),3.75(s,3H),3.25(d,J=9.5Hz,1H),2.90(dd,J=15.7,5.2Hz,1H),2.85–2.70(m,1H),2.49–2.30(m,1H),1.89(s,1H)。
e.e.99.6%
实施例303
N-(5-(6-(2-氯-6-氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1-甲基-1H-吡唑-4-基)丙烯酰胺
Figure PCTCN2018104007-appb-000089
6-(2-氯-6-氟-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑
将6-(2-氟-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑(556mg,1.38mmol),N-氯代丁二酰亚胺(184mg,1.38mmol)和乙酸(20mL)中,升温至80℃反应3h。后处理:加入二氯甲烷和水分层,有机相减压脱溶,用石油醚/乙酸乙酯(1:1)体系过硅胶层析柱得到黄色固体产物6-(2-氯-6-氟-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑(230mg,38%)。
参照实例302的操作步骤合成实例303。但在第5步用6-(2-氯-6-氟-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑代替6-(2-氟-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑。最终得到目标产物N-(5-(6-(2-氯-6-氟-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1-甲基-1H-吡唑-4-基)丙烯酰胺303。再通过手性柱拆分得到P1和P2。手性拆分条件:设备SFC,色谱柱:chiralpak-AD流动相:CO2-IPA(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000090
MS m/z(ESI):460.0[M+1] +
P1: 1H NMR(400MHz,DMSO):δ12.95(s,1H),9.48(s,1H),7.85(s,1H),6.91(d,J=7.8Hz,2H),6.53(dd,J=17.0,10.2Hz,1H),6.19(dd,J=17.0,2.1Hz,1H),5.66(dd,J=10.2,2.1Hz,1H),3.90(s,3H),3.89(s,3H),3.76(s,3H),3.65(dd,J=16.9,11.1Hz,2H),2.86(dd,J=15.6,5.2Hz,1H),2.58(dq,J=7.8,5.5Hz,1H),2.49–2.35(m,2H),2.21(s,1H)。
P2: 1H NMR(400MHz,DMSO):δ12.94(s,1H),9.47(s,1H),7.85(s,1H),6.91(d,J=7.8Hz,2H),6.52(dd,J=17.0,10.2Hz,1H),6.19(dd,J=17.0,2.1Hz,1H),5.66(dd,J=10.2,2.1Hz,1H),3.90(s,3H),3.89(s,3H),3.76(s,3H),3.71–3.58(m,2H),2.86(dd,J=15.7,5.3Hz,1H),2.38-2.46(m,2H),2.21(s,1H),1.87(d,J=11.9Hz,1H)。
实施例304
N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5-二氢-1H-吲唑-3-基)-5-(二甲氨基)苯基)丙烯酰胺
Figure PCTCN2018104007-appb-000091
合成284过程中,分离得到的副产物产物N-(2-(6-(2,6-二氯-3,5-二甲氧苯基)-4,5-二氢-1H-吲唑-3-基)-5-(二甲氨基)苯基)丙烯酰胺。
MS m/z(ESI):514.4[M+1] +
1H NMR(400MHz,DMSO):δ12.75(d,J=187.3Hz,1H),11.36(s,1H),9.41(s,1H),8.07(s,1H),7.27(s,3H),6.91(s,2H),6.60(s,3H),6.39–6.03(m,6H),5.75(s,2H),3.94(s,12H),2.95(s,12H),2.52(s,4H),2.34(s,4H)。
实施例310
N-(5-(6-(3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1-甲基-1H-吡唑-4-基)丙烯酰胺
Figure PCTCN2018104007-appb-000092
参照实例140的操作步骤合成实例310。但在第4步用6-(3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑代替6-(2,6-二氯-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑。最终得到目标产物N-(5-(6-(3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1-甲基-1H-吡唑-4-基)丙烯酰胺310。
手性拆分条件:设备SFC,色谱柱:chiralpak-AD流动相:CO2-IPA(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000093
MS m/z(ESI):407.8[M+1] +
P1: 1H NMR(400MHz,DMSO):δ12.89(s,1H),9.43(s,1H),7.80(s,1H),6.57–6.46(m,3H),6.37(t,J=2.2Hz,1H),6.19(dd,J=17.0,2.1Hz,1H),5.66(dd,J=10.2,2.0Hz,1H),3.77(s,3H),3.74(s,6H),3.00–2.86(m,2H),2.75(t,J=14.3Hz,1H),2.49–2.41(m,1H),2.36(d,J=16.9Hz,1H),1.97–1.75(m,2H).
P2: 1H NMR(400MHz,DMSO):δ12.89(s,1H),9.43(s,1H),7.80(s,1H),6.57–6.45(m,3H),6.37(t,J=2.2Hz,1H),6.18(dd,J=17.0,2.0Hz,1H),5.66(dd,J=10.2,1.9Hz,1H),3.77(s,3H),3.74(s,6H),2.92(d,J=11.9Hz,2H),2.75(t,J=14.1Hz,1H),2.49–2.41(m,1H),2.36(d,J=14.9Hz,1H),2.01–1.77(m,2H).
ee:100%
实施例311
N-(5-(6-(2-氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1-甲基-1H-吡唑-4-基)丙烯酰胺
Figure PCTCN2018104007-appb-000094
将6-(3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑(250mg,0.6mmol),N-氯代丁二酰亚胺(85mg,0.6mmol)和乙酸(10mL)中,升温至80℃反应3h。后处理:加入二氯甲烷和水分层,有机相减压脱溶,用石油醚/乙酸乙酯(1:1)体系过硅胶层析柱得到黄色固体产物6-(2-氯-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑(150mg,55%)。
参照实例302的操作步骤合成实例311。但在第5步用6-(2-氯-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑代替6-(2-氟-3,5-二甲氧苯基)-3-(1-甲基-4-硝基-1H-吡唑-5-基)-4,5,6,7-四氢-1H-吲唑。最终得到目标产物N-(5-(6-(2-氯-3,5-二甲氧苯基)-4,5,6,7-四氢-1H-吲唑-3-基)-1-甲基-1H-吡唑-4-基)丙烯酰胺311。
手性拆分条件:设备SFC,色谱柱:chiralpak-AD流动相:CO2-IPA(DEA)。在SFC机器上保留时间短的命名P1,保留时间长的命名P2。P1和P2中一个为R-异构体,另一个为S-异构体。
Figure PCTCN2018104007-appb-000095
MS m/z(ESI):442.0[M+1] +
P1: 1H NMR(400MHz,DMSO):δ12.96(s,1H),9.49(s,1H),7.83(s,1H),6.62(d,J=2.9Hz,2H),6.52(dd,J=17.0,10.2Hz,1H),6.19(d,J=17.0Hz,1H),5.66(d,J=10.2Hz,1H),3.86(s,3H),3.79(s,3H),3.76(s,3H),3.46(s,2H),2.94(dd,J=15.7,5.1Hz,1H),2.75–2.66(m,1H),2.38(d,J=14.5Hz,1H),1.90(s,1H).
P2: 1H NMR(400MHz,DMSO):δ12.99(s,1H),9.49(s,1H),7.83(s,1H),6.62(d,J=3.0Hz,2H), 6.52(dd,J=17.0,10.2Hz,1H),6.19(d,J=17.0Hz,1H),5.66(d,J=10.1Hz,1H),3.86(s,3H),3.79(s,3H),3.76(s,3H),3.53-3.42(m,2H),2.94(dd,J=15.5,4.7Hz,1H),2.76–2.65(m,1H),2.39(d,J=14.9Hz,1H),1.90(s,1H).
FGFR4的活性抑制测试
使用体外激酶检测实验评估本发明的化合物对酪氨酸激酶FGFR4活性的影响
实验方法概述如下:
使用CISBIO公司的
Figure PCTCN2018104007-appb-000096
KinEASE TM-TK 20000tests激酶检测试剂盒,试剂盒提供具有生物素标记的底物,Eu标记的磷酸化位点特异性抗体,及XL665标记的亲和素和相关缓冲液。FGFR4将底物磷酸化,Eu-Ab识别磷酸化底物,XL665-SA与底物上的生物素结合,导致Eu与XL665接近从而产生HTRF信号。通过读取HTRF信号的强弱来检测激酶的活性变化。在激酶检测实验中,主要包括两步反应,分别是激酶反应和检测反应。在激酶反应中,激酶消耗ATP将底物磷酸化,同时产成含有磷酸基团的底物。在检测反应中,加入检测试剂终止激酶反应,同时,检测试剂中的特异性抗体和XL665标记的亲和素分别与底物上的磷酸基团及生物素相结合产生HTRF信号,信号的强度与底物的磷酸化水平成正比,从而能够定量检测激酶FGFR4的活性。
检测过程中,化合物与酶的结合孵育在室温下进行,时间为60min。而激酶反应在37℃恒温进行,时间为40min。使用Corning 3674黑色384孔检测板,激酶人的FGFR4蛋白(460-802氨基酸)购自Carna公司(货号08-136),激酶底物为TK(购自Cisbio)和ATP(Sigma),使用酶标仪TECAN Spark 10M plate reader(TECAN)读取光信号。激酶反应缓冲液包括1X Enzymatic buffer(CISBIO)5mM MgCl 2(Sigma)、1mM DTT(Sigma);将激酶FGFR4使用缓冲液配制为0.25ug/mL浓度的激酶反应溶液;底物反应溶液包括0.75uM的底物和500μm的ATP。
化合物IC 50由10个浓度点通过以下公式计算。先用100%DMSO将化合物在96孔板中溶解稀释3个浓度:4mM,40uM,0.4uM,并转8ul化合物到384LDV Echo Source板,用Echo550转移化合物到Assay板,得到10个浓度点,每个浓度点设置两个复孔(起始点10uM,3倍稀释)。向384孔检测板中先添加6uL激酶溶液,混合均匀后室温孵育60分钟;随后加入4uL底物反应溶液,反应总体积为10uL,将反应混合物在37℃恒温反应40分钟;随后加入10uL激酶检测试剂并终止反应,然后在TECAN plate reader上读取数值。
抑制百分率基于以下公式计算:
抑制%=[1-(RLU 化合物-RLU min)/(RLU max-RLU min)]X100
其中RLU 化合物为给定化合物浓度下的冷光读数,RLU min为不加入激酶的情况下的冷光读数,RLU max为不加入化合物的情况下的冷光读数。通过使用Excel中XLfit程序来计算化合物的IC 50
化合物编号 IC50(nM)
015 80
093 29.22
096 348.25
98 7251.51
103 101.1
107 340.21
111 76.94
113 1881.25
114 158.48
133 4.56
137 27.82
140 24.62
Figure PCTCN2018104007-appb-000097
结论:本发明的化合物对酪氨酸激酶FGFR4的活性具有明显的抑制效应。
以上内容是结合具体的优选实施方式对本发明所作的进一步详细说明,不能认定本发明的具体实施只局限于这些说明。对于本发明所属技术领域的普通技术人员来说,在不脱离本发明构思的前提下, 还可以做出若干简单推演或替换,都应当视为属于本发明的保护范围。

Claims (13)

  1. 通式(I)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物:
    Figure PCTCN2018104007-appb-100001
    其中:
    环A不存在或者选自6-14元亚芳基、5-10元亚杂芳基、C 3-C 8亚环烷基或3-10元亚杂环基;
    R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、-NR 11R 12、-C(O)R 13、-C(O)OR 13、-C(O)NR 11R 12、-NR 11C(O)R 13、-NR 11C(O)OR 13、-NR 11C(O)NR 11R 12、-OC(O)R 13、-OC(O)OR 13、-OC(O)NR 11R 12、-S(O) pR 13、-S(O) pOR 13、-S(O) pNR 11R 12、-OS(O) pR 13或-NR 11S(O) pR 13
    R 6各自独立地选自H或-CH 2CH 2-,且当R 6为-CH 2CH 2-时,其另一端连接至X,任选地-CH 2CH 2-中的一个亚甲基被-O-或-NH-替代;
    R 7各自独立地选自H、卤素、氰基、NO 2、C 1-C 6烷基、C 1-C 6卤代烷基、C 2-C 6烯基、C 2-C 6炔基、C 3-C 8环烷基、-OR 13、或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;
    R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;
    R 11、R 12和R 13各自独立地选自H、C 1-C 6烷基、C 1-C 6杂烷基、C 3-C 8环烷基、3-10元杂环基、5-10元杂芳基或6-14元芳基;
    X选自-O-、-NH-或-CH 2-;Y选自-O-、-NH-或-CH 2-;条件是X和Y中至少有一个是CH 2
    W选自化学键、-NH-或-CH 2-;
    m为0、1、2、3或4;
    p为1或2。
  2. 根据权利要求1所述的通式(I)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,其为通式(II)所示的化合物:
    Figure PCTCN2018104007-appb-100002
    其中:
    环A选自6-10元亚芳基、5-6元亚杂芳基或C 3-C 8亚环烷基;
    R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基、C 1-C 6烷氧基或C 1-C 6卤代烷氧基;
    R 6各自独立地选自H或-CH 2CH 2-,且当R 6为-CH 2CH 2-时,其另一端连接至X;
    R 7各自独立地选自H、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;
    R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;
    X选自-O-、-NH-或-CH 2-;Y选自-O-、-NH-或-CH 2-;条件是X和Y中至少有一个是CH 2
    m为0、1、2、3或4;
    优选地,
    环A为亚苯基或5-6元亚杂芳基;优选地,环A为亚苯基;
    R 1和R 5各自独立地为卤素,优选地为Cl或F;
    R 2和R 4各自独立地为C 1-C 6烷氧基,优选地为-OCH 3
    R 3为H;
    R 6为H;
    R 7各自独立地选自H、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;优选地,R 7为H;
    R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;优选地,R 8为乙烯基或丙炔基;
    X为-CH 2-;Y选自-O-、-NH-或-CH 2-;
    m为0、1、2、3或4;优选地,m为0。
  3. 根据权利要求1所述的通式(I)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,其为通式(III)所示的化合物:
    Figure PCTCN2018104007-appb-100003
    其中:
    环A为6-10元亚芳基、5-6元亚杂芳基或C 3-C 8亚环烷基;优选地,环A选自苯基、萘基、吡咯基、呋喃基、噻吩基、咪唑基、吡唑基、噁唑基、异噁唑基、噻唑基、异噻唑基、三唑基、噁二唑基、噻二唑基、四唑基、吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基、四嗪基或环己基的二价基团,优选选自亚苯基、亚吡唑基或亚环己基;
    R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基、C 1-C 6烷氧基或C 1-C 6卤代烷氧基;
    R 7各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;
    R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;
    m为0、1、2、3或4;
    优选地,
    环A为亚苯基或5-6元亚杂芳基;优选地,环A为亚苯基或5元亚杂芳基;优选地,环A为亚苯基或亚吡唑基;
    R 1和R 5各自独立地选自H或卤素,优选地为H、Cl或F;
    R 2和R 4各自独立地为C 1-C 6烷氧基,优选地为-OCH 3
    R 3为H;
    R 7各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;优选地,R 7选自H、卤素、甲基、-N(CH 3)CH 2CH 2N(CH 3) 2、-N(CH 3) 2、4-甲基-哌嗪基或吗啉基;
    R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;优选地,R 8为乙烯基或丙炔基;
    m为0、1、2、3或4;优选地,m为0、1或2;优选地,m为1;
    优选地,
    环A为亚苯基或5-6元亚杂芳基;优选地,环A为亚苯基或5元亚杂芳基;优选地,环A为亚苯基或亚吡唑基;
    R 1和R 5各自独立地选自H或卤素,优选地为H、Cl或F;
    R 2和R 4各自独立地为C 1-C 6烷氧基,优选地为-OCH 3
    R 3为H;
    R 7各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;优选地,R 7选自H、卤素、甲基、-N(CH 3)CH 2CH 2N(CH 3) 2、-N(CH 3) 2或4-甲基-哌嗪基;
    R 8为任选取代的C 2-C 6烯基;优选地,R 8为乙烯基;
    m为0、1、2、3或4;优选地,m为0、1或2;优选地,m为1;
    优选地,
    环A为亚苯基或5-6元亚杂芳基;优选地,环A为亚苯基或5元亚杂芳基;优选地,环A为亚苯基或亚吡唑基;
    R 1和R 5各自独立地为卤素,优选地选自Cl或F;
    R 2和R 4各自独立地为C 1-C 6烷氧基,优选地为-OCH 3
    R 3为H;
    R 7各自独立地选自H、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;优选地,R 7选自H、甲基、-N(CH 3)CH 2CH 2N(CH 3) 2或4-甲基-哌嗪基;
    R 8为任选取代的C 2-C 6烯基;优选地,R 8为乙烯基;
    m为0、1、2、3或4;优选地,m为0、1或2;优选地,m为1。
  4. 根据权利要求1所述的通式(I)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,其为通式(IV)所示的化合物:
    Figure PCTCN2018104007-appb-100004
    其中:
    R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基、C 1-C 6烷氧基或C 1-C 6卤代烷氧基;
    R 7各自独立地选自H、C 1-C 6烷基或C 1-C 6卤代烷基;
    R 8选自C 2-C 6烯基或C 2-C 6炔基;
    X选自-NH-,Y选自-CH 2-;或
    X选自-CH 2-,Y选自-NH-;
    m为0、1、2、3或4;
    优选地,
    R 1和R 5各自独立地选自卤素,优选地为Cl或F;
    R 2和R 4各自独立地选自C 1-C 6烷氧基,优选地为-OCH 3
    R 3选自H;
    R 7选自H;
    R 8选自乙烯基或丙炔基;
    X为-NH-;
    Y为-CH 2-;
    m为1。
  5. 根据权利要求1所述的通式(I)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,其为通式(V)所示的化合物:
    Figure PCTCN2018104007-appb-100005
    其中:
    R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基、C 1-C 6烷氧基或C 1-C 6卤代烷氧基;
    R 7各自独立地选自H、C 1-C 6烷基或C 1-C 6卤代烷基;
    R 8选自C 2-C 6烯基或C 2-C 6炔基;
    m为0、1或2;
    优选地,
    R 1和R 5各自独立地为卤素,优选为Cl或F,更优选为Cl;
    R 2和R 4各自独立地为C 1-C 6烷氧基,优选为-OCH 3
    R 3选自H;
    R 7选自H或C 1-C 6烷基,优选为H或甲基;
    R 8选自乙烯基或丙炔基,优选为乙烯基;
    m为1;
    优选地,
    R 1和R 5各自独立地选自H或卤素,优选地为H、Cl或F;
    R 2和R 4各自独立地为C 1-C 6烷氧基,优选地为-OCH 3
    R 3为H;
    R 7各自独立地选自H、C 1-C 6烷基或C 1-C 6卤代烷基;优选地,R 7选自H或甲基;
    R 8为任选取代的C 2-C 6烯基;优选地,R 8为乙烯基;
    m为0、1或2;优选地,m为1。
  6. 根据权利要求1所述的通式(I)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,其为通式(VI)所示的化合物:
    Figure PCTCN2018104007-appb-100006
    其中:
    R 1、R 2、R 3、R 4和R 5各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基、C 1-C 6烷氧基或C 1-C 6卤代烷氧基;
    R 7各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基、C 1-C 6烷氧基、C 1-C 6卤代烷氧基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;
    R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;
    m为0、1、2、3或4;
    优选地,
    其中:
    R 1和R 5各自独立地选自卤素,优选为Cl或F;
    R 2和R 4各自独立地选自C 1-C 6烷氧基,优选为甲氧基;
    R 3为H;
    R 7选自H或C 1-C 6烷基或-NR aR b,其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;优选为H、甲基或-N(CH 3)CH 2CH 2N(CH 3) 2
    R 8选自C 2-C 6烯基或C 2-C 6炔基,优选选自乙烯基或丙炔基;
    m为1;
    优选地,
    R 1和R 5各自独立地选自H或卤素,优选地为H、Cl或F;
    R 2和R 4各自独立地为C 1-C 6烷氧基,优选地为-OCH 3
    R 3为H;
    R 7各自独立地选自H、卤素、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;优选地,R 7选自H、卤素、甲基、-N(CH 3)CH 2CH 2N(CH 3) 2、-N(CH 3) 2、4-甲基-哌嗪基或吗啉基;
    R 8选自任选取代的C 2-C 6烯基或C 2-C 6炔基;优选地,R 8为乙烯基或丙炔基;
    m为0、1、2、3或4;优选地,m为0、1或2;优选地,m为1;
    优选地,
    R 1和R 5各自独立地选自H或卤素,优选地为H、Cl或F;
    R 2和R 4各自独立地为C 1-C 6烷氧基,优选地为-OCH 3
    R 3为H;
    R 7各自独立地选自H、卤素或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;优选地,R 7选自H、卤素、-N(CH 3)CH 2CH 2N(CH 3) 2、-N(CH 3) 2或4-甲基-哌嗪基;
    R 8为任选取代的C 2-C 6烯基;优选地,R 8为乙烯基;
    m为0、1、2、3或4;优选地,m为0、1或2;优选地,m为1;
    优选地,
    R 1和R 5各自独立地为卤素,优选地选自Cl或F;
    R 2和R 4各自独立地为C 1-C 6烷氧基,优选地为-OCH 3
    R 3为H;
    R 7各自独立地选自H或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;优选地,R 7选自H、-N(CH 3)CH 2CH 2N(CH 3) 2或4-甲基-哌嗪基;
    R 8为任选取代的C 2-C 6烯基;优选地,R 8为乙烯基;
    m为0、1、2、3或4;优选地,m为0、1或2;优选地,m为1。
  7. 根据权利要求1所述的通式(I)所示的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,其为通式(VII)所示的化合物:
    Figure PCTCN2018104007-appb-100007
    其中,
    R 1和R 5各自独立地为卤素,优选地选自Cl或F;
    R 2和R 4各自独立地为C 1-C 6烷氧基,优选地为-OCH 3
    R 3为H;
    R 7各自独立地选自卤素、C 1-C 6烷基、C 1-C 6卤代烷基或-NR aR b;其中,R a和R b各自独立地选自H或任选被-NR cR d取代的C 1-C 6烷基;R c和R d各自独立地选自H或C 1-C 6烷基;或者,R a、R b和他们所连接的N原子一起形成3-10元杂环基(例如6元杂环基),其任选被C 1-C 6烷基或C 1-C 6卤代烷基取代;优选地,R 7选自卤素、甲基、-N(CH 3) 2或吗啉基;
    R 8为任选取代的C 2-C 6烯基;优选地,R 8为乙烯基;
    m为0、1、2、3或4;优选地,m为0、1或2;优选地,m为1。
  8. 化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,以及混合物,其中该化合物为:
    Figure PCTCN2018104007-appb-100008
    Figure PCTCN2018104007-appb-100009
    Figure PCTCN2018104007-appb-100010
    Figure PCTCN2018104007-appb-100011
    Figure PCTCN2018104007-appb-100012
    Figure PCTCN2018104007-appb-100013
    Figure PCTCN2018104007-appb-100014
    Figure PCTCN2018104007-appb-100015
  9. 药物组合物,其含有根据权利要求1-8中任一项所述的化合物或其可药用的盐、互变异构体、 外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,及药学上可接受的赋形剂。
  10. 根据权利要求1-8中任一项所述的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,或根据权利要求9所述的药物组合物在制备用于治疗和/或预防FGFR4酪氨酸激酶介导的疾病的药物中的用途。
  11. 一种在受试者中治疗和/或预防FGFR4酪氨酸激酶介导的疾病的方法,包括向所述受试者给药根据权利要求1-8中任一项所述的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,或根据权利要求9所述的药物组合物。
  12. 根据权利要求1-8中任一项所述的化合物或其可药用的盐、互变异构体、外消旋体、对映异构体、非对映异构体、前药、水合物或溶剂合物,及其混合物,或根据权利要求9所述的药物组合物,其用于治疗和/或预防FGFR4酪氨酸激酶介导的疾病。
  13. 根据权利要求10的用途或权利要求11的方法或权利要求12的化合物或组合物,其中所述疾病为肿瘤,例如胃癌、甲状腺癌、前列腺癌、乳腺癌、肉瘤(例如横纹肌肉瘤)、皮肤癌(例如黑色素瘤)、肝癌(例如肝细胞癌和胆管上皮癌)、胰腺癌(例如胰腺上皮内瘤样病变和胰腺导管腺癌)、肺癌(例如非小细胞肺癌和肺腺癌)、肾癌(例如肾细胞癌)、结直肠癌和卵巢癌。
PCT/CN2018/104007 2017-09-05 2018-09-04 芳香类衍生物、其制备方法及其在医药上的应用 WO2019047826A1 (zh)

Priority Applications (9)

Application Number Priority Date Filing Date Title
CN201880034411.9A CN110997637B (zh) 2017-09-05 2018-09-04 芳香类衍生物、其制备方法及其在医药上的应用
EP18854268.2A EP3680236A4 (en) 2017-09-05 2018-09-04 AROMATIC DERIVATIVE, MANUFACTURING PROCESS FOR IT AND MEDICAL APPLICATION OF IT
US16/644,919 US11279697B2 (en) 2017-09-05 2018-09-04 Aromatic derivative, preparation method for same, and medical applications thereof
KR1020207009736A KR102677016B1 (ko) 2017-09-05 2018-09-04 방향족 유도체, 그의 제조 방법 및 그의 의학적 응용
SG11202001979PA SG11202001979PA (en) 2017-09-05 2018-09-04 Aromatic derivative, preparation method for same, and medical applications thereof
CA3074885A CA3074885A1 (en) 2017-09-05 2018-09-04 Aromatic derivative, preparation method for same, and medical applications thereof
JP2020533340A JP7346420B2 (ja) 2017-09-05 2018-09-04 芳香族誘導体、その調製方法、およびその医学的適用
PH12020500436A PH12020500436A1 (en) 2017-09-05 2020-03-04 Aromatic derivative, preparation method for same, and medical applications thereof
JP2023144389A JP2023158113A (ja) 2017-09-05 2023-09-06 芳香族誘導体、その調製方法、およびその医学的適用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201710791233.1 2017-09-05
CN201710791233 2017-09-05

Publications (1)

Publication Number Publication Date
WO2019047826A1 true WO2019047826A1 (zh) 2019-03-14

Family

ID=65634601

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/104007 WO2019047826A1 (zh) 2017-09-05 2018-09-04 芳香类衍生物、其制备方法及其在医药上的应用

Country Status (10)

Country Link
US (1) US11279697B2 (zh)
EP (1) EP3680236A4 (zh)
JP (2) JP7346420B2 (zh)
KR (1) KR102677016B1 (zh)
CN (1) CN110997637B (zh)
CA (1) CA3074885A1 (zh)
PH (1) PH12020500436A1 (zh)
SG (1) SG11202001979PA (zh)
TW (1) TWI817955B (zh)
WO (1) WO2019047826A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020177534A1 (en) * 2019-03-05 2020-09-10 Bioardis Llc Aromatic derivatives, preparation methods, and medical uses thereof
WO2021060307A1 (ja) * 2019-09-25 2021-04-01 富士フイルム株式会社 イミダゾピリジン化合物またはその塩および医薬組成物

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
WO2012174476A2 (en) 2011-06-16 2012-12-20 Isis Pharmaceuticals, Inc. Antisense modulation of fibroblast growth factor receptor 4 expression
WO2014011900A2 (en) 2012-07-11 2014-01-16 Blueprint Medicines Inhibitors of the fibroblast growth factor receptor
WO2015008844A1 (ja) 2013-07-18 2015-01-22 大鵬薬品工業株式会社 Fgfr阻害剤耐性癌の治療薬
WO2015057938A1 (en) 2013-10-18 2015-04-23 Eisai R&D Management Co., Ltd. Pyrimidine fgfr4 inhibitors
WO2015059668A1 (en) 2013-10-25 2015-04-30 Novartis Ag Ring-fused bicyclic pyridyl derivatives as fgfr4 inhibitors
WO2015061572A1 (en) 2013-10-25 2015-04-30 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
WO2015197519A1 (de) 2014-06-24 2015-12-30 Covestro Deutschland Ag Verfahren zur herstellung von di- und polyaminen derdiphenylmethanreihe
WO2016064960A1 (en) 2014-10-22 2016-04-28 Incyte Corporation Bicyclic heterocycles as fgfr4 inhibitors
WO2016134320A1 (en) 2015-02-20 2016-08-25 Incyte Corporation Bicyclic heterocycles as fgfr inhibitors
WO2016134294A1 (en) 2015-02-20 2016-08-25 Incyte Corporation Bicyclic heterocycles as fgfr4 inhibitors
WO2016134314A1 (en) 2015-02-20 2016-08-25 Incyte Corporation Bicyclic heterocycles as fgfr4 inhibitors

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3029864B1 (en) 2014-12-01 2020-11-18 Harman Becker Automotive Systems GmbH Fast representation of station information in a fm receiver using a single tuner
CN105906630B (zh) 2015-04-06 2018-10-23 四川百利药业有限责任公司 用作fgfr抑制剂的n-(1h-吡唑-5-基)嘧啶并吡唑-4,6-二取代胺类化合物
WO2016164703A1 (en) 2015-04-09 2016-10-13 Eisai R & D Management Co., Ltd. Fgfr4 inhibitors
CN109224235B (zh) 2018-10-23 2020-05-19 孙喜家 一种双腔管
CN109331654A (zh) 2018-10-26 2019-02-15 高邮高和光电器材有限公司 一种有机废气光催化氧化装置

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
WO2012174476A2 (en) 2011-06-16 2012-12-20 Isis Pharmaceuticals, Inc. Antisense modulation of fibroblast growth factor receptor 4 expression
WO2014011900A2 (en) 2012-07-11 2014-01-16 Blueprint Medicines Inhibitors of the fibroblast growth factor receptor
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
WO2015008844A1 (ja) 2013-07-18 2015-01-22 大鵬薬品工業株式会社 Fgfr阻害剤耐性癌の治療薬
US20160136168A1 (en) * 2013-07-18 2016-05-19 Taiho Pharmaceutical Co., Ltd. Therapeutic agent for fgfr inhibitor-resistant cancer
WO2015057938A1 (en) 2013-10-18 2015-04-23 Eisai R&D Management Co., Ltd. Pyrimidine fgfr4 inhibitors
WO2015057963A1 (en) 2013-10-18 2015-04-23 Eisai R&D Management Co., Ltd. Fgfr4 inhibitors
WO2015061572A1 (en) 2013-10-25 2015-04-30 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
WO2015059668A1 (en) 2013-10-25 2015-04-30 Novartis Ag Ring-fused bicyclic pyridyl derivatives as fgfr4 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
WO2015197519A1 (de) 2014-06-24 2015-12-30 Covestro Deutschland Ag Verfahren zur herstellung von di- und polyaminen derdiphenylmethanreihe
WO2016064960A1 (en) 2014-10-22 2016-04-28 Incyte Corporation Bicyclic heterocycles as fgfr4 inhibitors
WO2016134320A1 (en) 2015-02-20 2016-08-25 Incyte Corporation Bicyclic heterocycles as fgfr inhibitors
WO2016134294A1 (en) 2015-02-20 2016-08-25 Incyte Corporation Bicyclic heterocycles as fgfr4 inhibitors
WO2016134314A1 (en) 2015-02-20 2016-08-25 Incyte Corporation Bicyclic heterocycles as fgfr4 inhibitors

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
BERGE ET AL., J. PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
BROWN, AP ET AL., TOXICOL. PATHOL., 2005, pages 449 - 455
CAO ET AL., CANCER RES, vol. 70, no. 16, 2010, pages 6497 - 6508
D. FLEISHERS. RAMONH. BARBRA: "Improved Oral Drug Delivery: Solubility Limitations Overcome by the Use of Prodrugs", ADVANCED DRUG DELIVERY REVIEWS, vol. 19, no. 2, 1996, pages 115 - 130
DING ET AL., NATURE, vol. 455, no. 7216, 2008, pages 1069 - 1075
ESWARAKUMAR, CYTOKINE & GROWTH FACTOR REVIEWS, 2005
HO ET AL., JOURNAL OF HEPATOLOGY, vol. 50, 2009, pages 118 - 27
ROIDL ET AL., ONCOGENE, vol. 29, no. 10, 2010, pages 1543 - 1552
SAWEY ET AL., CANCER CELL, vol. 19, 2011, pages 347 - 358
T. HIGUCHIV. STELLA: "Bioreversible Carriers in Drug Design", vol. 14, 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS, article "Prodrugs as Novel Delivery Systems, A.C.S. Symposium Series"
TAYLOR ET AL., J CLIN INVEST, vol. 119, 2009, pages 3395 - 3407
VERGNES ET AL., CELL METABOLISM, vol. 17, 2013, pages 916 - 28
WU ET AL., J BIOL CHEM, vol. 285, no. 8, 2010, pages 5165 - 5170
ZAID ET AL., CLIN. CANCER RES., 2013, pages 809

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020177534A1 (en) * 2019-03-05 2020-09-10 Bioardis Llc Aromatic derivatives, preparation methods, and medical uses thereof
WO2020177067A1 (en) * 2019-03-05 2020-09-10 Bioardis Llc Aromatic derivatives, preparation methods, and medical uses thereof
US11976058B2 (en) 2019-03-05 2024-05-07 Bioardis Llc Aromatic derivatives, preparation methods, and medical uses thereof
WO2021060307A1 (ja) * 2019-09-25 2021-04-01 富士フイルム株式会社 イミダゾピリジン化合物またはその塩および医薬組成物

Also Published As

Publication number Publication date
TWI817955B (zh) 2023-10-11
CN110997637B (zh) 2022-09-27
CA3074885A1 (en) 2019-03-14
EP3680236A1 (en) 2020-07-15
SG11202001979PA (en) 2020-04-29
PH12020500436A1 (en) 2021-02-08
US20200262827A1 (en) 2020-08-20
EP3680236A4 (en) 2021-04-21
KR20200083448A (ko) 2020-07-08
CN110997637A (zh) 2020-04-10
JP2020532581A (ja) 2020-11-12
TW201912631A (zh) 2019-04-01
KR102677016B1 (ko) 2024-06-19
JP7346420B2 (ja) 2023-09-19
US11279697B2 (en) 2022-03-22
JP2023158113A (ja) 2023-10-26

Similar Documents

Publication Publication Date Title
CN112601750B (zh) Ptpn11(shp2)抑制剂
CN109790169B (zh) 具有作为usp30抑制剂活性的氰基吡咯烷衍生物
TWI433845B (zh) 嗒酮衍生物
TWI542590B (zh) 1,2-雙取代雜環化合物
ES2316546T3 (es) 2-arilamino-pirimidinas para el tratamiento de trastornos asociados a gsk3.
JP5662143B2 (ja) 腫瘍の処置のためのMetキナーゼ阻害剤としての2−オキソ−3−ベンジルベンゾキサゾール−2−オン誘導体、および関連する化合物
WO2018019204A1 (zh) 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物
TW201833108A (zh) 醯胺類衍生物抑制劑及其製備方法和應用
JP5576358B2 (ja) ピリダジノン誘導体
RU2415853C2 (ru) Бензимидазолы, применимые в качестве ингибиторов протеинкиназ
BR112016013632B1 (pt) Carboxamidas, seus usos, seus intermediários e seu processo de preparação, e medicamento
US20160145238A1 (en) Heteroaryl substituted pyrazoles
KR20100031771A (ko) 피리미디닐 피리다지논 유도체
DE102008052943A1 (de) Azaindolderivate
CA2907730A1 (en) Diaminoheteroaryl substituted indazoles
DE102008028905A1 (de) 3-(3-Pyrimidin-2-yl-benzyl)-[1,2,4]triazolo[4,3-b]pyridazinderivate
JP2023158113A (ja) 芳香族誘導体、その調製方法、およびその医学的適用
WO2020143763A1 (zh) 卤代烯丙基胺类化合物及其应用
JP2021516674A (ja) 5−ヘテロアリール置換イミダゾール−3−カルボキサミドならびにその調製および使用の方法
WO2017190637A1 (zh) 用于抑制蛋白酪氨酸激酶活性的稠合嘧啶类化合物
WO2018072707A1 (zh) 芳香族醚类衍生物、其制备方法及其在医药上的应用
TW202128686A (zh) 稠合雜芳基類衍生物、其製備方法及其在醫藥上的應用
WO2017097113A1 (zh) 用于抑制蛋白酪氨酸激酶活性的氨基嘧啶类化合物
KR20110099742A (ko) 3-(3-피리미딘-2-일-벤질)-[1,2,4]트리아졸로[4,3-b]피리미딘 유도체
JP5643192B2 (ja) 腫瘍を処置するためのチロシンキナーゼモジュレーターとしてのジヒドロピラゾール誘導体

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: 18854268

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2020533340

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3074885

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018854268

Country of ref document: EP

Effective date: 20200406