WO2018010514A1 - 作为fgfr抑制剂的杂环化合物 - Google Patents

作为fgfr抑制剂的杂环化合物 Download PDF

Info

Publication number
WO2018010514A1
WO2018010514A1 PCT/CN2017/088038 CN2017088038W WO2018010514A1 WO 2018010514 A1 WO2018010514 A1 WO 2018010514A1 CN 2017088038 W CN2017088038 W CN 2017088038W WO 2018010514 A1 WO2018010514 A1 WO 2018010514A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
aryl
compound
cyclic
Prior art date
Application number
PCT/CN2017/088038
Other languages
English (en)
French (fr)
Inventor
孔祥龙
周超
郑之祥
Original Assignee
南京天印健华医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to SG11201900298RA priority Critical patent/SG11201900298RA/en
Priority to EP17826852.0A priority patent/EP3489223A4/en
Priority to AU2017295628A priority patent/AU2017295628B2/en
Priority to MX2019000451A priority patent/MX2019000451A/es
Priority to RU2019100164A priority patent/RU2742485C2/ru
Priority to KR1020197004058A priority patent/KR102386428B1/ko
Application filed by 南京天印健华医药科技有限公司 filed Critical 南京天印健华医药科技有限公司
Priority to JP2019523154A priority patent/JP6896852B2/ja
Priority to CA3030070A priority patent/CA3030070A1/en
Priority to CN201780002825.9A priority patent/CN108349896B/zh
Publication of WO2018010514A1 publication Critical patent/WO2018010514A1/zh
Priority to PH12019500036A priority patent/PH12019500036A1/en
Priority to US16/246,236 priority patent/US10590109B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/84Nitriles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the present invention relates to a process for the preparation of a class of heterocyclic compounds and pharmaceutical compositions containing the same, and to the use as a fibroblast growth factor receptor FGFR inhibitor.
  • the compounds of the invention are useful for treating or preventing a disease associated with FGFR, such as cancer.
  • Fibroblast growth factor is a family of polypeptides encoded by the FGF gene family with different biological activities and structural correlations. So far, the FGF family has found 22 members.
  • Fibroblast Growth Factors Receptors are a class of transmembrane tyrosine kinase receptors that mediate FGF signaling into the cytoplasm. FGFRs encoded by four independent genes, namely FGFR1, FGFR2, FGFR3 and FGFR4, have been identified. They are all single-stranded glycoprotein molecules composed of extracellular regions, transmembrane regions, and intracellular regions.
  • FGFR-FGF signaling system plays an important role in many biological processes such as cell proliferation, differentiation, migration, angiogenesis, and tissue repair.
  • FGFR4 is the major FGF receptor subtype in the liver. Ten of the more than 20 fibroblast growth factors (FGF) found to date have been able to bind to FGFR4, of which only FGF19 specifically binds to FGFR4. Recent studies have shown that changes in FGFR4, such as overexpression, mutation, translocation, and truncation, are associated with the progression of a variety of human cancers, including rhabdomyosarcoma, renal cell carcinoma, myeloma, breast cancer, stomach cancer, and colon cancer. , bladder cancer, pancreatic cancer and hepatocellular carcinoma.
  • FGF fibroblast growth factors
  • Rings A and R are each independently selected from substituted or unsubstituted aryl, heteroaryl, and when A or R is substituted, may be substituted by one or more substituents independently selected from hydrogen, halogen. , cyano, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered heterocyclic, aryl, heteroaryl, aldehyde, -C(O)R 1 , carboxyl, alkenyl, alkynyl, -OR 1 , -NR 2 R 3 , wherein the alkyl, cyclo, heterocyclyl, aryl or heteroaryl group is optionally selected from one or more selected from the group consisting of halogen, cyano, C1-C8 alkyl, C3 -C8 cyclic group, 3-8 membered heterocyclic group, -OR 4 , -OC(O)NR 5 R 6 , -C(O)OR 4 , -C(O)
  • X is selected from the group consisting of CR 7 R 8 , NR 7 , O, S;
  • R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are each independently selected from the group consisting of hydrogen, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered monocyclic heterocyclic, monocyclic heteroaryl Or a monocyclic aryl, alkenyl, alkynyl group, wherein said R 2 and R 3 , R 5 and R 6 may form a 3-7 membered heterocyclic group together with the N atom to which they are attached; R 7 and R 8 may form a 3-8 membered ring group or a 3-8 membered monocyclic heterocyclic group together with the C atom to which they are attached;
  • R 7 and R 8 are each independently selected from the group consisting of hydrogen, halogen, cyano, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered heterocyclic, aryl, heteroaryl, aldehyde, and -C ( O) R 1 , carboxy, alkenyl, alkynyl, -OR 1 , -NR 2 R 3 wherein the alkyl, cyclo, heterocyclyl, aryl or heteroaryl group is optionally selected by one or more From halogen, cyano, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered heterocyclyl, -OR 4 , -OC(O)NR 5 R 6 , -C(O)OR 4 , -C (O)NR 5 R 6 , -C(O)R 4 , -NR 5 R 6 , -NR 5 C(O)R 4 , -NR 4 C(O)NR 5 R 6
  • R 9 is independently selected from hydrogen, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered heterocyclyl, aryl, heteroaryl, aldehyde, -C(O)R 1 , alkenyl, alkyne a group wherein the alkyl, cyclo, heterocyclyl, aryl or heteroaryl group is optionally selected from one or more selected from the group consisting of halogen, cyano, C1-C8 alkyl, C3-C8 cyclo, 3-8 A heterocyclic group, -OR 4 , -OC(O)NR 5 R 6 , -C(O)OR 4 , -C(O)NR 5 R 6 , -C(O)R 4 , -NR 5 R 6 -NR 5 C(O)R 4 , -NR 4 C(O)NR 5 R 6 , -S(O)mR 4 , -NR 5 S(O)mR 4 , -SR
  • Z 1 , Z 2 , Z 3 are each independently selected from CR Z1 , CR Z2 , CR Z3 or N, and:
  • R Z1 , R Z2 , R Z3 are each independently selected from the group consisting of hydrogen, halogen, cyano, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered heterocyclic, aryl, heteroaryl, aldehyde, -C(O)R 1 , carboxy, alkenyl, alkynyl, -OR 1 , -NR 2 R 3 wherein the alkyl, cyclo, heterocyclyl, aryl or heteroaryl group is optionally a plurality selected from the group consisting of halogen, cyano, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered heterocyclic, -OR 4 , -OC(O)NR 5 R 6 , -C(O)OR 4 , -C(O)NR 5 R 6 , -C(O)R 4 , -NR 5 R 6 , -NR 5 C(O)R 4 ,
  • X is selected from the group consisting of CR 7 R 8 , NR 7 , O, S;
  • R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are each independently selected from the group consisting of hydrogen, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered monocyclic heterocyclic, monocyclic heteroaryl Or a monocyclic aryl, alkenyl, alkynyl group, wherein said R 2 and R 3 , R 5 and R 6 may form a 3-7 membered heterocyclic group together with the N atom to which they are attached; R 7 and R 8 may form a 3-8 membered ring group or a 3-8 membered monocyclic heterocyclic group together with the C atom to which they are attached;
  • R 7 and R 8 are each independently selected from the group consisting of hydrogen, halogen, cyano, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered heterocyclic, aryl, heteroaryl, aldehyde, and -C ( O) R 1 , carboxy, alkenyl, alkynyl, -OR 1 , -NR 2 R 3 wherein the alkyl, cyclo, heterocyclyl, aryl or heteroaryl group is optionally selected by one or more From halogen, cyano, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered heterocyclyl, -OR 4 , -OC(O)NR 5 R 6 , -C(O)OR 4 , -C (O)NR 5 R 6 , -C(O)R 4 , -NR 5 R 6 , -NR 5 C(O)R 4 , -NR 4 C(O)NR 5 R 6
  • R 9 is independently selected from hydrogen, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered heterocyclyl, aryl, heteroaryl, aldehyde, -C(O)R 1 , alkenyl, alkyne a group wherein the alkyl, cyclo, heterocyclyl, aryl or heteroaryl group is optionally selected from one or more selected from the group consisting of halogen, cyano, C1-C8 alkyl, C3-C8 cyclo, 3-8 A heterocyclic group, -OR 4 , -OC(O)NR 5 R 6 , -C(O)OR 4 , -C(O)NR 5 R 6 , -C(O)R 4 , -NR 5 R 6 -NR 5 C(O)R 4 , -NR 4 C(O)NR 5 R 6 , -S(O)mR 4 , -NR 5 S(O)mR 4 , -SR
  • a compound of the formula (I) is different A construct, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt thereof, characterized in that the compound of formula I is of formula IIIa, IIIb, IIIc, IIId, IIIe, IIIf:
  • R Z1 , R Z2 , R Z3 , R 10 , R 11 are each independently selected from the group consisting of hydrogen, halogen, cyano, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered heterocyclic, aryl, hetero An aryl group, an aldehyde group, -C(O)R 1 , a carboxyl group, an alkenyl group, an alkynyl group, -OR 1 , -NR 2 R 3 , wherein the alkyl group, the cyclic group, the heterocyclic group, the aryl group or the heteroaryl group The group is optionally selected from one or more selected from the group consisting of halogen, cyano, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered heterocyclic, -OR 4 , -OC(O)NR 5 R 6 ,- C(O)OR 4 , -C(O)NR 5 R 6
  • R 7 is independently selected from H, C 1 -C 8 alkyl, C 3 -C 8 cyclic, 3-8 membered monocyclic heterocyclyl, monocyclic heteroaryl or monocyclic aryl, wherein said alkyl, cyclo,
  • the heterocyclic group, aryl or heteroaryl group is optionally selected from one or more selected from the group consisting of halogen, cyano, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered heterocyclic, -OR 4 , -OC (O)NR 5 R 6 , -C(O)OR 4 , -C(O)NR 5 R 6 , -C(O)R 4 , -NR 5 R 6 , -NR 5 C(O)R 4 , -NR 4 C(O)NR 5 R 6 , -S(O)mR 4 , -NR 5 S(O)mR 4 , -SR 4 , -NR 4 S(O)
  • R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are each independently selected from the group consisting of hydrogen, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered monocyclic heterocyclic, monocyclic heteroaryl Or a monocyclic aryl, alkenyl, alkynyl group, wherein said R 2 and R 3 , R 5 and R 6 may form a 3-7 membered heterocyclic group together with the N atom to which they are attached; R 7 and R 8 may form a 3-8 membered ring group or a 3-8 membered monocyclic heterocyclic group together with the C atom to which they are attached;
  • the R 2 and R 3 , R 5 and R 6 together with the N atom to which they are attached may form a 3-8 membered heterocyclic group.
  • a compound of the formula (I), an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt thereof characterized in that The compound of formula I is of formula IV;
  • R Z1 and R Z2 are each independently selected from the group consisting of hydrogen, halogen, C1-C4 alkyl, C3-C7 cyclic, 4-6 membered monocyclic heterocyclic, 5-6 membered monocyclic heteroaryl or monocyclic aryl.
  • R 7 is selected from H, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered monocyclic heterocyclic, monocyclic heteroaryl or monocyclic aryl, wherein said alkyl, cyclic, heterocyclic
  • the aryl, aryl or heteroaryl group is optionally selected from one or more selected from the group consisting of halogen, cyano, C1-C8 alkyl, C3-C8 cyclo, 3-8 membered heterocyclyl, -OR 4 , -OC(O )NR 5 R 6 , -C(O)OR 4 , -C(O)NR 5 R 6 , -C(O)R 4 , -NR 5 R 6 , -NR 5 C(O)R 4 , -NR 4 C(O)NR 5 R 6 , -S(O)mR 4 , -NR 5 S(O)mR 4 , -SR 4 , -NR 4 S(O)
  • R 10 is independently selected from the group consisting of hydrogen, halogen, halogenated C1-C4 alkyl, cyano;
  • R 11 is independently selected from the group consisting of hydrogen, halogen, C1-C4 alkyl, halogenated C1-C4 alkoxy, C1-C6 alkoxy, HO-C1-C4 alkoxy, cyano, NR 2 R 3 , C1 -C4 alkoxy C1-C4 alkoxy, C1-C4 alkoxy halogenated C1-C4 alkoxy;
  • R 1 , R 2 and R 3 are each independently selected from hydrogen, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered monocyclic heterocyclic, monocyclic heteroaryl or monocyclic aryl, C1- a C3 alkylthio group, a haloalkoxy group substituted at any position by a hydroxyl group;
  • R 4 , R 5 and R 6 are each independently selected from hydrogen, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered monocyclic heterocyclic, monocyclic heteroaryl or monocyclic aryl, alkenyl And alkynyl, wherein said R 2 and R 3 , R 5 and R 6 may form a 3-7 membered heterocyclic group together with the N atom to which they are attached; said R 7 and R 8 may be bonded thereto The attached C atoms together form a 3-8 membered ring group or a 3-8 membered monocyclic heterocyclic group.
  • a compound of the formula (I) is different a compound, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt thereof, wherein the compound of formula I is of formula V;
  • R Z1 and R Z2 are each independently selected from the group consisting of hydrogen, halogen, C1-C4 alkyl, C3-C7 cyclic, 5-6 membered monocyclic heterocyclic, 5-6 membered monocyclic heteroaryl or monocyclic aryl.
  • R 7 is selected from the group consisting of hydrogen, C1-C4 alkyl, C3-6 cyclo, wherein the alkyl or cyclic group is optionally selected from one or more selected from the group consisting of C1-C3 alkyl, 4-6 membered monocyclic heterocyclic groups. Substituted by a substituent;
  • R 11 is selected from the group consisting of NR 2 R 3 , C1-C3 alkoxy, -O(CH 2 ) 0-1 -R 4 ; wherein R 4 is independently selected from hydrogen, C1-C8 alkyl, HO-C1-C8 An alkyl group, a C3-C8 cyclic group, a 3-8 membered monocyclic heterocyclic group, a monocyclic heteroaryl group or a monocyclic aryl group, wherein the alkyl group, the cyclic group, the heterocyclic group, the aryl group or the heteroaryl group is One or more selected from the group consisting of halogen, cyano, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered heterocyclic, -OR 5 , -OC(O)NR 5 R 6 , -C( O) OR 5 , -C(O)NR 5 R 6 , -C(O)R 5 , -NR 5
  • R 4 , R 5 and R 6 are each independently selected from hydrogen, C1-C8 alkyl, C3-C8 cyclic, 3-8 membered monocyclic heterocyclic, monocyclic heteroaryl or monocyclic aryl, alkenyl And alkynyl, wherein said R 2 and R 3 , R 5 and R 6 may form a 3-7 membered heterocyclic group together with the N atom to which they are attached; said R 7 and R 8 may be bonded thereto The attached C atoms together form a 3-8 membered ring group or a 3-8 membered monocyclic heterocyclic group.
  • Typical compounds of the invention include, but are not limited to:
  • the compounds of the invention are potent inhibitors of FGFR, especially effective inhibitors of FGFR4.
  • the compounds of the invention are therefore useful in the treatment or prevention of FGFR-mediated diseases, particularly FGFR4-mediated diseases including, but not limited to, cancer and inflammatory diseases.
  • FGFR-mediated diseases particularly FGFR4-mediated diseases including, but not limited to, cancer and inflammatory diseases.
  • Inventive The compounds are useful for treating or preventing cancer, such as rhabdomyosarcoma, renal cell carcinoma, myeloma, breast cancer, gastric cancer, colon cancer, bladder cancer, pancreatic cancer, and hepatocellular carcinoma.
  • the compounds of the invention are particularly useful for the treatment or prevention of liver cancer, especially hepatocellular carcinoma.
  • tumors in which an activating mutant of a receptor tyrosine kinase or an upregulation of a receptor tyrosine kinase is present will be particularly sensitive to such inhibitors.
  • the compounds of the invention which are selective inhibitors of FGFR4 have lower side effects.
  • the invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the compound of the formula (I) or an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable thereof Salts and pharmaceutically acceptable carriers, diluents, excipients.
  • the invention also includes a method of preparing the pharmaceutical composition, for example, by mixing a compound of the invention with a pharmaceutically acceptable carrier, diluent, excipient.
  • a pharmaceutically acceptable carrier for example, a pharmaceutically acceptable diluent, excipient.
  • the pharmaceutical compositions of the invention can be prepared by conventional methods in the art.
  • Another aspect of the invention relates to a compound of the formula (I) or an isomer, a prodrug, a stable isotopic derivative or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, Use of a granule for the preparation of a medicament for the treatment or prevention of a FGFR, in particular a FGFR4-mediated disease, such as a tumor or an inflammatory disease.
  • a FGFR4-mediated disease such as a tumor or an inflammatory disease.
  • Another aspect of the invention relates to a compound of the formula (I) or a tautomer, a meso group, a racemate, an enantiomer, a diastereomer, a mixture thereof, And a pharmaceutically acceptable salt thereof, or use of the pharmaceutical composition for the preparation of a medicament for treating and/or preventing diseases such as tumors and inflammation.
  • the medicament may be in any pharmaceutical dosage form including, but not limited to, tablets, capsules, solutions, lyophilized preparations, injections.
  • the pharmaceutical preparation of the present invention can be administered in the form of a dosage unit containing a predetermined amount of the active ingredient per dosage unit.
  • a dosage unit may comprise, for example, from 0.5 mg to 1 g, preferably from 1 mg to 700 mg, particularly preferably from 5 mg to 300 mg, of a compound of the invention, or a drug, depending on the condition being treated, the method of administration, and the age, weight and condition of the patient.
  • the formulations may be administered in the form of dosage units containing a predetermined amount of active ingredient per dosage unit.
  • Preferred dosage unit formulations are those containing the daily or divided doses indicated above or their corresponding fractions of the active ingredient.
  • pharmaceutical preparations of this type can be prepared using methods well known in the pharmaceutical art.
  • the pharmaceutical preparations of the invention may be adapted for administration by any suitable method desired, for example by oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral. (including subcutaneous, intramuscular, intravenous or intradermal) methods of administration.
  • Such formulations can be prepared by, for example, combining the active ingredient with one or more excipients or one or more adjuvants, using all methods known in the art of pharmacy.
  • a pharmaceutical preparation suitable for oral administration can be administered as a separate unit such as a capsule or tablet; a powder or granule; a solution or suspension in an aqueous or non-aqueous liquid; or an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. .
  • the invention also relates to a method of treating or preventing a FGFR, in particular a FGFR4-mediated disease, such as a tumor or an inflammatory disease, comprising administering to a patient in need thereof a therapeutically effective amount of the general formula (I) according to the invention.
  • a FGFR in particular a FGFR4-mediated disease, such as a tumor or an inflammatory disease
  • administering comprising administering to a patient in need thereof a therapeutically effective amount of the general formula (I) according to the invention.
  • Another aspect of the invention relates to a compound of the formula (I) or an isomer thereof, a prodrug, a stable isotopic derivative or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, or form Agent. It is used for the treatment or prevention of FGFR, in particular FGFR4-mediated diseases, such as tumors or inflammatory diseases.
  • Another aspect of the invention relates to a compound represented by the formula (I) or a tautomer, a mesogen, a racemate, an enantiomer thereof, as a disease for treating and/or preventing a tumor or the like, Diastereomers, mixtures thereof, and pharmaceutically acceptable salts thereof.
  • the invention also provides methods of making the compounds.
  • the first step in the structure of the compound (VI-1), R z2 and R 7 are identical to the structure of (VI), and the compound (VI-1) is catalyzed by palladium (palladium acetate) for the insertion of carbonyl, and 1,1- Bis(diphenylphosphino)ferrocene is used as a ligand, triethylamine is used as a base, and compound (VI-2) is synthesized.
  • R z2 and R 7 are identical to the structure of (VI).
  • the second step the ester in the compound (VI-2) is reduced by a reducing reagent (sodium borohydride) to synthesize the compound (VI-3), and the structures of the compound (VI-3) are R z2 and R 7 and (VI). It is the same.
  • a reducing reagent sodium borohydride
  • the third step the hydroxyl group in the structure of (VI-3) is substituted, and the compound (VI-4) is synthesized by using phosphorus tribromide as a reaction reagent, and R z2 and R 7 are in the structure of the compound (VI-4).
  • VI The structure is consistent.
  • Step 4 The bromine in the structure of (VI-4) is replaced by a nucleophile (morpholin-3-one, 4-methylpiperazin-2-one, etc.), using a base (sodium hydride) as a deproton.
  • Reagent, synthetic compound (VI-5), and R z2 and R 7 in the structure of compound (VI-5) are identical to those in (VI) structure.
  • R z2 and R 7 are identical to the structure of (VI), wherein the amino protecting group can be removed with an acid (trifluoroacetic acid) and the triethylamine is alkalized. Purification affords compound (VI).
  • R z1 , R z2 and R 7 are identical to the structure of (IV), and the compound (VI) is activated by an acylating agent (diphenyl carbonate or phenyl chloroformate) to synthesize Compound (VII), Compound (VI) is synthesized by acylation.
  • Compound (VII) is a base (hexamethyldisilazide lithium) as a deprotonating reagent.
  • R 10 and R 11 on the 2-aminopyridine ring are identical to the structure of (IV), and replace the phenol group in the compound (VII), using a base (hexamethyldisilazide lithium)
  • a base hexamethyldisilazide lithium
  • the compound (VIII) is synthesized, and in the compound (VIII), R z1 , R z2 and R 7 are identical to the structure of (IV).
  • R z1 , R z2 and R 7 are identical to the structure of (IV), wherein the acetal protecting group can be removed by acid, alkalized with sodium hydrogencarbonate, and purified to obtain a compound. (IV).
  • Cx-y denotes a range of the number of carbon atoms, wherein x and y are both integers, for example, a C3-8 ring group represents a ring group having 3-8 carbon atoms, -C0-2 alkane The group represents an alkyl group having 0 to 2 carbon atoms, wherein -C0 alkyl means a chemical single bond.
  • alkyl refers to a saturated aliphatic hydrocarbon group, including straight and branched chain groups of 1 to 20 carbon atoms, for example, may be from 1 to 18 carbon atoms, from 1 to 12 carbon atoms, Linear and branched groups of 1 to 8 carbon atoms, 1 to 6 carbon atoms or 1 to 4 carbon atoms.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, Tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 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-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, and various branched isomers thereof.
  • the alkyl group can be substituted or unsubstituted.
  • cyclic group refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon group comprising from 3 to 12 ring carbon atoms, for example from 3 to 12, from 3 to 10, from 3 to 3 8 or 3 to 6 ring carbon atoms, or may be 3, 4, 5, 6, 7, 8 membered rings.
  • monocyclic ring groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene Base, cyclooctyl and the like.
  • the cyclic group can be substituted or unsubstituted.
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic group comprising from 3 to 20 ring carbon atoms, for example from 3 to 16, from 3 to 12, 3 Up to 10, 3 to 8, or 3 to 6 ring atoms, wherein one or more ring atoms are selected from nitrogen, oxygen or S(O)m (where m is an integer from 0 to 2), but not included
  • ring portion of -OO-, -OS- or -SS-, the remaining ring atoms are carbon.
  • the heterocyclyl ring contains from 3 to 10 ring atoms, most preferably a 5- or 6-membered ring, wherein from 1 to 4 are heteroatoms More preferably, 1 to 3 are hetero atoms, and most preferably 1 to 2 are hetero atoms.
  • monocyclic heterocyclic groups include pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl and the like.
  • Polycyclic heterocyclic groups include spiro, fused, and bridged heterocyclic groups.
  • spiroheterocyclyl refers to a polycyclic heterocyclic group of 5 to 20 members, which shares an atom (referred to as a spiro atom) between the monocyclic rings, wherein one or more of the ring atoms are selected from nitrogen, oxygen or A hetero atom of S(O)m (where m is an integer of 0 to 2), and the remaining ring atoms are carbon. These may contain one or more double bonds, but none of the rings have a fully conjugated pi-electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • the spiro 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 monospirocyclic group and a bispirocyclic 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 yuan single spiro ring group.
  • a non-limiting embodiment of a spiro group includes
  • fused heterocyclyl refers to 5 to 20 members, and each ring in the system shares an adjacent pair of atomic polycyclic heterocyclic groups with other rings in the system, and one or more rings may contain One or more double bonds, but none of the rings have a fully conjugated ⁇ -electron system in which one or more ring atoms are selected from nitrogen, oxygen or S(O)m (where m is an integer from 0 to 2) heteroatoms The remaining ring atoms are carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic group preferably a bicyclic or tricyclic ring, more preferably a 5-membered/5-membered or 5-membered/6-membered bicyclic fused heterocyclic group.
  • fused heterocyclic groups include
  • the heterocyclyl ring may be fused to an aryl, heteroaryl or cyclic ring wherein the ring to which the parent structure is attached is a heterocyclic group, non-limiting examples comprising:
  • the heterocyclic group may be substituted or unsubstituted.
  • aryl refers to a 6 to 14 membered all-carbon monocyclic or fused polycyclic (ie, a ring that shares a pair of adjacent carbon atoms) groups, a polycyclic ring having a conjugated ⁇ -electron system (ie, The ring group with an adjacent pair of carbon atoms is preferably 6 to 10 members, such as phenyl and naphthyl, most preferably phenyl.
  • the aryl ring may be fused to a heteroaryl, heterocyclyl or cyclic ring wherein the ring to which the parent structure is attached is an aryl ring, non-limiting examples comprising:
  • the aryl group can be substituted or unsubstituted.
  • heteroaryl refers to a heteroaromatic system comprising from 1 to 4 heteroatoms, from 5 to 14 ring atoms, wherein the heteroatoms include oxygen, sulfur and nitrogen. It is preferably 5 to 10 yuan.
  • the heteroaryl group is 5- or 6-membered, such as furyl, thienyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, imidazolyl, tetrazolyl, oxazolyl, iso Oxazolyl or the like
  • the heteroaryl ring may be fused to an aryl, heterocyclic or cyclic ring wherein the ring to which the parent structure is attached is a heteroaryl ring, non-limiting examples comprising:
  • Heteroaryl groups can be substituted or unsubstituted.
  • halogen refers to fluoro, chloro, bromo or iodo.
  • cyano refers to -CN.
  • alkenyl refers to a straight, branched or cyclic non-aromatic hydrocarbon radical containing at least one carbon to carbon double bond. There may be from 1 to 3 carbon-carbon double bonds, preferably one carbon-carbon double bond. These include vinyl, propenyl, butenyl, 2-methylbutenyl and cyclohexenyl. The alkenyl group may be substituted.
  • C2-4 alkenyl refers to an alkenyl group having 2 to 4 carbon atoms.
  • alkynyl refers to a straight, branched or cyclic hydrocarbon radical containing at least one carbon to carbon triple bond. There may be 1-3 carbon-carbon triple bonds, preferably one carbon-carbon triple bond. These include ethynyl, propynyl, butynyl and 3-methylbutynyl.
  • C2-4 alkynyl refers to an alkynyl group having 2 to 4 carbon atoms.
  • alkoxy refers to a cyclic or acyclic alkyl group having the number of carbon atoms attached through an oxygen bridge, and includes an alkyloxy group, a cycloalkyloxy group, and a heterocycloalkyloxy group.
  • alkoxy as defined above for alkyl, heterocycloalkyl and cycloalkyl, "optionally” or “optionally” means that the subsequently described event or environment may, but need not, occur, the description includes The event or environment where the event occurs or does not occur.
  • heterocyclic group optionally substituted by an alkyl group means that an alkyl group may be, but 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 are independently substituted with each other 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.
  • the term "pharmaceutical composition” means a mixture containing one or more of the compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, as well as other components such as physiology/ Pharmaceutically acceptable carriers and excipients.
  • the purpose of the pharmaceutical composition is to promote the administration of the organism, which facilitates the absorption of the active ingredient and thereby exerts biological activity.
  • Root temperature as used herein means 15-30 °C.
  • stable isotope derivative includes an isotope-substituted derivative obtained by substituting any one of the hydrogen atoms of formula I with 1-5 deuterium atoms, and any carbon atom of formula I is 1-3 carbon 14
  • “Pharmaceutically acceptable salts” as described herein are discussed in Berge, et al., “Pharmaceutically acceptable salts", J. Pharm. Sci., 66, 1-19 (1977), and for pharmaceutical chemists It is apparent that the salts are substantially non-toxic and provide the desired pharmacokinetic properties, palatability, absorption, distribution, metabolism or excretion, and the like.
  • the pharmaceutically acceptable salts of the present invention can be synthesized by general chemical methods.
  • the preparation of the salt can be carried out by reacting the free base or acid with an equivalent stoichiometric or excess acid (inorganic or organic acid) or base in a suitable solvent or solvent composition.
  • prodrug as used in the present invention means that the compound is converted into the original active compound after being metabolized in the body. Typically, the prodrug is inactive or less active than the active parent compound, but can provide convenient handling, administration or improved metabolic properties.
  • isomers means that the compound of formula (I) of the present invention may have asymmetric centers and racemates, racemic mixtures and individual diastereomers, all of which include Stereoisomers, geometric isomers are all included in the present invention.
  • the geometric isomers include cis and trans isomers.
  • tumor includes both benign and malignant tumors, such as cancer.
  • cancer includes various malignancies, particularly malignant tumors in which FGFR, particularly FGFR4, is involved, including but not limited to rhabdomyosarcoma, renal cell carcinoma, myeloma, breast cancer, gastric cancer, colon cancer, Bladder cancer, pancreatic cancer and hepatocellular carcinoma.
  • inflammatory disease refers to any inflammatory disease in which FGFR, and in particular FGFR4, is involved in the onset of inflammation.
  • the structure of all compounds of the invention can be identified by nuclear magnetic resonance (1H NMR) and/or mass spectrometry (MS).
  • MS Low resolution mass spectrometry
  • the thin layer silica gel plate is Yantai Yellow Sea HSGF254 or Qingdao GF254 silica gel plate.
  • Column chromatography generally uses Yantai Yellow Sea 100-200 or 200-300 mesh silica gel as a carrier.
  • 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 Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc, Shanghai Bied Pharmaceutical, Shanghai A. Latin Chemical, Shanghai Miner Chemical, and Belling Chemical.
  • the solvent used in the reaction is an anhydrous solvent, wherein the anhydrous tetrahydrofuran is a commercially available tetrahydrofuran, the sodium block is used as a water removing agent, and the benzophenone is used as an indicator, and the solution is refluxed under nitrogen to the solution. Blue-violet, distilled and collected under nitrogen protection at room temperature.
  • Other anhydrous solvents were purchased from Aladdin Chemical and Lalene Chemical. All anhydrous solvents were transferred and used without special instructions under nitrogen protection.
  • 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 carbon monoxide atmosphere means that the reaction flask is connected to a carbon monoxide balloon having a volume of about 1 L;
  • the hydrogenation reaction is usually evacuated, charged with hydrogen, and operated three times.
  • reaction temperature is room temperature, and the temperature range is from 15 ° 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 The polarity is adjusted to adjust, and a small amount of triethylamine and an acidic or alkaline reagent may be added for adjustment.
  • Example 2 was synthesized with reference to the procedure of Example 1, but in the sixth step, 6-amino-4-chloroniguonitrile was substituted with 6-chloropyrimidin-4-amine.
  • Example 3 was synthesized with reference to the procedure of Example 1, but in the sixth step, 6-amino-4-chloroniguonitrile was replaced with 6-amino nicotine nitrile.
  • Example 4 was synthesized with reference to the procedure of Example 1. However, in the sixth step, 6-amino-4-chloronicotonitrile was replaced with 4-chloropyridin-2-amine.
  • Example 5 was synthesized by referring to the procedure of Example 1, except that in the seventh step, 2-methoxyethylamine was replaced with isopropylamine.
  • Example 6c was synthesized by reference to the procedure of the sixth step of Example 1, substituting 6-amino-4-isopropoxy nicotine nitrile for 6-amino-4-chloronicotonitrile. Obtain the target product 1-(6-(1,3-dioxin) Pentocyclo-2-yl)pyridin-2-yl)-3-(5-cyano-4-isopropoxypyridin-2-yl)-1-methylurea 6c (8 mg, white solid). Yield: 46%.
  • Example 6 was synthesized with reference to the procedure of the eighth step of Example 1.
  • the title product 3-(5-cyano-4-isopropoxypyridin-2-yl)-1-(6-formylpyridin-2-yl)-1-methylurea 7 (5 mg, white solid) . Yield: 71%.
  • Example 7 was synthesized by reference to the procedure of Example 6, except that in the first step, isopropanol was replaced with 2-methoxyethanolamine.
  • Di-tert-butyl(5-bromo-6-methylpyridin-2-yl)imide dicarbonate 9b (3.86 g, 10.00 mmol), 1-bromopyrrolidine-2,5-dione 4.45 g, 25.00 mmol), benzyl peroxy anhydride (0.24 g, 1.00 mmol) and carbon tetrachloride (100 mL) were mixed and stirred at 90 ° C for 16 hours. The mixture was cooled to room temperature, and then evaporated to dryness. 2-yl)imidodicarbonate 9c (4.00 g, yellow solid), yield: 74%.
  • ester 1.5:1 gives the desired product tert-butyl-(6-(2-methoxyethyl)-5-((3-carbonylmorpholine)methyl)pyridin-2-yl)(methyl)amino Carboxyl ester 9i (70 mg, white solid), yield: 95%.
  • Di-tert-butyl(5-bromo-6-methylpyridin-2-yl)imidodicarbonate 10b (80 g, 0.21 mol), 1-bromopyrrolidine-2,5-dione (110.00) g, 0.63 mol), benzyl peroxy anhydride (0.24 g, 0.06 mol) and carbon tetrachloride (600 mL) were mixed and stirred at 90 ° C for 16 hours. The mixture was cooled to room temperature, and then evaporated to dryness. 2-yl)imidodicarbonate 10c (90.00 g, yellow solid), yield: 80%.
  • Example 11 was synthesized by reference to the procedure of Example 9, but in the thirteenth step, 1-(4-chloro-5-cyanopyridin-2-yl)-3-(6-formyl-5-(( 3-carbonylmorpholine)methyl)pyridin-2-yl)urea substituted 3-(5-cyano-4-((2-methoxyethyl)amino)pyridin-2-yl)-1-(6 -(Dimethoxymethyl)-5-((3-carbonylmorpholino)methyl)pyridin-2-yl)-1-methylurea.
  • Example 12c was synthesized by reference to the procedure of the eleventh step of Example 9, substituting 6-amino-4-chloronicotonitrile with 6-amino-4-(2-methoxyethoxy) nicotine nitrile.
  • the desired product 3-(5-cyano-4-(2-methoxyethoxy)pyridin-2-yl)-1-(6-(dimethoxymethyl)-5-(3- Carbonylmorpholino)methyl)pyridin-2-yl)-1-methylurea 12c (8 mg, white solid), yield: 70%.
  • Example 12 was synthesized by referring to the procedure of the thirteenth step of Example 9. Using 3-(5-cyano-4-(2-methoxyethoxy)pyridin-2-yl)-1-(6-(dimethoxymethyl)-5-((3-carbonyl)? Phenyl)methyl)pyridin-2-yl)-1-methylurea substituted 3-(5-cyano-4-((2-methoxyethyl)amino)pyridin-2-yl)-1-( 6-(Dimethoxymethyl)-5-((3-carbonylmorpholine)methyl)pyridin-2-yl)-1-methylurea. The title product 3-(5-cyano-4-isopropoxypyridin-2-yl)-1-(6-formylpyridin-2-yl)-1-methylurea 12 (6 mg, white solid) . Yield: 82%.
  • Example 13 was synthesized with reference to the procedure of Example 12. However, in the first step, 2-methoxyethanol was replaced with isopropanol.
  • Example 14 was synthesized with reference to the procedure of Example 12. However, in the first step, 2-methoxyethanol was replaced with (R)-1-methoxypropan-2-ol.
  • Example 16 was synthesized by reference to the procedure of Example 15, but in the fifth step, 2-methoxyethylamine was replaced with isopropylamine.
  • Example 17 was synthesized by following the procedure of Example 15, but in the sixth step, 3-(4-chloro-5-cyanopyridin-2-yl)-1-(6-(dimethoxymethyl) was used. -5-((4-Methyl-2-carbonylpiperazin-1-yl)methyl)pyridin-2-yl)-1-methylurea substituted 3-(5-cyano-4-((2- Methoxyethyl)amino)pyridin-2-yl)-1-(6-(dimethoxymethyl)-5-((4-methyl-2-carbonylpiperazin-1-yl)methyl Pyridin-2-yl)-1-methylurea.
  • EtOAc EtOAc
  • EtOAc EtOAc
  • Example 19 was synthesized by following the procedure of Example 18 except that in the first step, (R)-1-methoxypropan-2-ol was replaced with isopropanol.
  • Example 20 was synthesized by following the procedure of Example 18, but in the first step, (R)-1-methoxypropan-2-ol was replaced with 2-methoxyethanol.
  • Example 22 was synthesized by referring to the procedure of Example 21, but in the fifth step, 1-(5-(((tert-butyldimethylsilyl)oxy)methyl)-6-(dimethoxy) was used.
  • Example 24 was synthesized by following the procedure of Example 18 except that (R)-1-methoxypropan-2-ol was replaced with (S)-1-methoxypropan-2-ol in the first step.
  • EtOAc EtOAc
  • EtOAc EtOAc
  • Example 26 was synthesized by following the procedure of Example 25, but using (S)-3-(5-cyano-4-((1-methoxypropan-2-yl)oxy)pyridine in the first step -2-yl)-1-(6-formyl-5-((4-methyl-2-carbonylpiperazin-1-yl)methyl)pyridin-2-yl)-1-methylurea substituted ( R)-3-(5-Cyano-4-((1-methoxypropan-2-yl)oxy)pyridin-2-yl)-1-(6-formyl-5-((4- Methyl-2-carbonylpiperazin-1-yl)methyl)pyridin-2-yl)-1-methylurea.
  • FGFR4 fibroblast growth factor receptor 4
  • the in vitro activity of FGFR4 was determined by measuring the phosphorylation level of the substrate in the kinase reaction using the HTRF kinase assay kit.
  • the reaction buffer contained the following components: 5-fold diluted Enzymatic buffer/kinase 5X (Cisbio, Cat. No.
  • 62EZBFDD main component 50 mM HEPES, pH 7.0), 5 mM MgCl 2 , 1 mM DTT; human recombinant FGFR4 catalytic domain protein ( Amino acids 460-802) were purchased from Tsinghua Protein Research Technology Center and diluted to 0.5 ng/uL of kinase solution in reaction buffer; substrate reaction solution including biotin-labeled tyrosine kinase substrate diluted to 500 nM with reaction buffer (Cisbio, catalog number 62TK0PEC) and 90 uM ATP, the assay solution consisted of a test buffer (Cisbio, Cat. No.
  • 62SDBRDF diluted to 0.125 ng/uL Eu 3+ labeled cage antibody (Cisbio, Cat. No. 61T66KLB) and 31.25 nM Streptomyces Avidin-labeled XL665 (Cisbio, Cat. No. 610SAXLB).
  • Percent inhibition 100-100* (signal compound - signal negative control ) / (signal positive control - signal negative control )
  • Compound IC 50 value (ID Business Solutions Ltd., UK) software is calculated by the following equation using a 10-point concentration XLfit:
  • the in vitro activity of FGFR1 was determined by detecting the phosphorylation level of the substrate in the kinase reaction using the HTRF kinase assay kit.
  • the reaction buffer contained the following components: 5-fold diluted Enzymatic buffer/kinase 5X (Cisbio, Cat. No.
  • 62EZBFDD main component 50 mM HEPES, pH 7.0), 5 mM MgCl 2 , 1 mM DTT; human recombinant FGFR1 catalytic domain protein ( Amino acid 308-731) was purified by the company itself and diluted with a reaction buffer to a 0.6 ng/uL kinase solution; the substrate reaction solution consisted of a 400 nM biotinylated tyrosine kinase substrate (Cisbio, diluted with reaction buffer). Catalog number 62TK0PEC) and 40uM ATP, the test solution consisted of a test buffer (Cisbio, Cat. No.
  • 62SDBRDF diluted to 0.125 ng / uL Eu 3 + labeled cage antibody (Cisbio, Cat. No. 61T66KLB) and 25nM streptavidin label XL665 (Cisbio, article number 610SAXLB).
  • Percent inhibition 100-100* (signal compound - signal negative control ) / (signal positive control - signal negative control )
  • Compound IC 50 value (ID Business Solutions Ltd., UK) software is calculated by the following equation using a 10-point concentration XLfit:
  • FGFR2 fibroblast growth factor receptor 2
  • the in vitro activity of FGFR2 was determined by measuring the phosphorylation level of the substrate in the kinase reaction using the HTRF kinase assay kit.
  • the reaction buffer contained the following components: 5-fold diluted Enzymatic buffer/kinase 5X (Cisbio, Cat. No. 62EZBFDD) (main component 50 mM HEPES, pH 7.0), 5 mM MgCl 2 , 1 mM DTT; human recombinant FGFR2 catalytic domain protein ( Amino acid 400-821) was purchased from Yishen Shenzhou Biotechnology Co., Ltd.
  • reaction buffer 0.045 ng/uL of kinase solution
  • substrate reaction solution including biotinylated tyrosine kinase diluted to 800 nM with reaction buffer Substrate (Cisbio, catalog number 62TK0PEC) and 50 uM ATP
  • the assay solution was diluted with assay buffer (Cisbio, Cat. No. 62SDBRDF) to 0.125 ng/uL Eu 3+ labeled cage antibody (Cisbio, Cat. No. 61T66KLB) and 50 nM strand Mold avidin-labeled XL665 (Cisbio, Cat. No. 610SAXLB).
  • Percent inhibition 100-100* (signal compound - signal negative control ) / (signal positive control - signal negative control )
  • Compound IC 50 value (ID Business Solutions Ltd., UK) software is calculated by the following equation using a 10-point concentration XLfit:
  • FGFR3 fibroblast growth factor receptor 3
  • the in vitro activity of FGFR3 was determined by measuring the phosphorylation level of the substrate in the kinase reaction using the HTRF kinase assay kit.
  • the reaction buffer contained the following components: 5-fold diluted Enzymatic buffer/kinase 5X (Cisbio, Cat. No. 62EZBFDD) (main component 50 mM HEPES, pH 7.0), 5 mM MgCl 2 , 1 mM DTT; human recombinant FGFR3 catalytic domain protein ( Amino acid 399-806) was purchased from Yishen Shenzhou Biotechnology Co., Ltd.
  • kinase solution with reaction buffer; substrate reaction solution including biotinylated tyrosine kinase diluted to 1000 nM with reaction buffer Substrate (Cisbio, catalog number 62TK0PEC) and 90 uM ATP, the assay solution was diluted with assay buffer (Cisbio, Cat. No. 62SDBRDF) to 0.125 ng/uL Eu 3+ labeled cage antibody (Cisbio, Cat. No. 61T66KLB) and 62.5 nM Streptavidin-labeled XL665 (Cisbio, Cat. No. 610SAXLB).
  • Percent inhibition 100-100* (signal compound - signal negative control ) / (signal positive control - signal negative control )
  • Compound IC 50 value (ID Business Solutions Ltd., UK) software is calculated by the following equation using a 10-point concentration XLfit:
  • the compounds of the invention have a selective inhibitory effect on FGFR4.
  • the effect of the compounds of the invention on Hep3B cell proliferation was assessed using a luminescent cell viability assay.
  • the luminescence signal generated in the assay is proportional to the number of viable cells in the medium by using a unique, stable luciferase assay for the indicator ATP of viable cellular metabolism. To detect the cell proliferation status of Hep3B.
  • CellTilter-Glo reagent (Promega, G7572) consists of CellTilter-Glo lyophilized powder and CellTilter-Glo buffer, which can be used to dissolve the lyophilized powder into the buffer.
  • Hep3B cells (ATCC, HB-8064) (cell sourced from Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences) were cultured in DMEM complete medium (Thermofisher, 11995073) containing 10% FBS (GBICO, 10099-141) and 100 units/ml of S.
  • DMEM complete medium Thermofisher, 11995073
  • FBS GBICO, 10099-141
  • the mixture (Thermofisher, 15140122), when the cells cover 80-90% in the culture vessel, digested with 0.25% trypsin (containing EDTA) (Thermofisher, 25200056) and then planted in a white 384-well plate (Thermofisher) , 164610), 1000 cells per well (27 ⁇ l DMEM complete medium), then 384-well plates were incubated overnight (18-20 hours) in a 37 ° C, 5% CO 2 incubator. Compounds were diluted in 100% DMSO to 5 mM, then serially diluted 4-fold with DMSO to a minimum concentration of 0.061 ⁇ M, and each concentration was diluted 50-fold with FBS-free DMEM medium.
  • the initial concentration of the compound can be lowered. After overnight, add 3 ⁇ l of DMEM-diluted compound to each well and mix gently by centrifugation. Add 10 ⁇ M BLU9931 as a negative pair. Photo (100% inhibition), plus 0.2% DMSO group as a positive control (0% inhibition). The 384-well plate was placed in a 37 ° C, 5% CO 2 incubator for further incubation. After 72 hours, it was taken out and allowed to stand at room temperature for 30 minutes. The CTG reagent was also taken out to room temperature. 15 ⁇ l of CTG reagent was added to each well and placed on a shaker. Lightly shake for 3 minutes to ensure sufficient cell lysis, leave for 10 minutes to stabilize the luminescence signal, then read the luminescence signal with EnVision (Perkin Elmer).
  • the percentage inhibition of Hep3B cell proliferation by the compound can be calculated by the following formula:
  • Percent inhibition 100-100* (signal compound - signal negative control ) / (signal positive control - signal negative control )
  • the compounds of the present invention have a good inhibitory effect on Hep3B cell proliferation.

Abstract

本发明涉及一类杂环化合物、含有其的药物组合物、以及它们的制备方法,和其作为成纤维细胞生长因子受体FGFR抑制剂的用途。所述化合物为式I所示的杂环化合物,或其药学上可接受的盐、前药、溶剂化合物、多晶型体、异构体、稳定同位素衍生物。本发明还涉及所述化合物用于治疗或预防由FGFR介导的相关疾病例如癌症的用途以及应用其治疗所述疾病的方法。

Description

作为FGFR抑制剂的杂环化合物 技术领域
本发明涉及一类杂环化合物以及含有其的药物组合物的制备方法,和其作为成纤维细胞生长因子受体FGFR抑制剂的用途。本发明所述化合物可用于治疗或预防由FGFR介导的相关疾病,如癌症。
背景技术
成纤维细胞生长因子(fibroblast growth factor,FGF)是一类由FGF基因家族编码具有不同生物学活性、结构相关的多肽家族。到目前为止FGF家族已发现有22个成员。成纤维细胞生长因子受体(Fibroblast Growth Factors Receptors,FGFRs)是一类跨膜的酪氨酸激酶受体,它们介导FGF信号传递到细胞质中。目前已经确定了由4种独立基因编码的FGFRs,即FGFR1、FGFR2、FGFR3和FGFR4。它们都为单链的糖蛋白分子,由细胞外区、跨膜区和细胞内区组成。这些受体-配体相互作用导致受体二聚化、自磷酸化,然后与膜结合蛋白和胞质辅助蛋白质形成复合物,从而介导多种信号传导。FGFR-FGF信号传导***在细胞增殖、分化、迁移、血管生成和组织修复等许多生物学过程中发挥重要作用。
FGFR4是肝脏中主要的FGF受体亚型。迄今发现的20多种成纤维生长因子(FGF)中有10个能与FGFR4结合,其中仅有FGF19与FGFR4特异性结合。近年来研究表明,FGFR4的改变,如过表达、突变、易位、和截短等,与多种人癌症的进展有关,包括横纹肌肉瘤、肾细胞癌、骨髓瘤、乳腺癌、胃癌、结肠癌、膀胱癌、胰腺癌和肝细胞癌。
因此,可以预测,选择性抑制FGFR4可以用于治疗以上癌症,尤其是存在受体酪氨酸激酶的激活突变体或受体酪氨酸激酶的上调的肿瘤将对该类抑制剂特别敏感。
发明内容
本发明的目的在于提供一种如式I所示的化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐
Figure PCTCN2017088038-appb-000001
其中环A和R各自独立选自取代或未取代的芳基、杂芳基,当A或R被取代时,可被一个或多个取代基取代,所述的取代基独立选自氢、卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、芳基、杂芳基、醛基、-C(O)R1、羧基、烯基、炔基、-OR1、-NR2R3,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
X选自CR7R8、NR7、O、S;
Y选自-C(O)-、-C(=NR9)-、-S(O)m-;
R1、R2、R3、R4、R5、R6各自独立地选自氢、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基、烯基、炔基,其中所述的R2和R3、R5和R6可与它们所连接的N原子一起形成3-7元的杂环基;所述的R7和R8可与它们所连接的C原子一起形成3-8元的环基或3-8元单环杂环基;
R7、R8各自独立地选自氢、卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、芳基、杂芳基、醛基、-C(O)R1、羧基、烯基、炔基、-OR1、-NR2R3,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
R9独立地选自氢、C1-C8烷基、C3-C8环基、3-8元杂环基、芳基、杂芳基、醛基、-C(O)R1、烯基、炔基,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、 -C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
且m为1或2。
在本发明的一个实施方案中,一种通式(I)所述的化合物、其异构体、前药或其药学上可接受的盐,其特征在于所述的如式I所示的化合物为式II所示:
Figure PCTCN2017088038-appb-000002
Z1、Z2、Z3各自独立地选自CRZ1、CRZ2、CRZ3或N,并且:
Z1为N时,Z2、Z3不同时为N;
Z2为N时,Z1、Z3不同时为N;
Z3为N时,Z1、Z2不同时为N;
RZ1、RZ2、RZ3各自独立地选自氢、卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、芳基、杂芳基、醛基、-C(O)R1、羧基、烯基、炔基、-OR1、-NR2R3,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
X选自CR7R8、NR7、O、S;
Y选自-C(O)-、-C(=NR9)-、-S(O)m-;
R1、R2、R3、R4、R5、R6各自独立地选自氢、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基、烯基、炔基,其中所述的R2和R3、R5和R6可与它们所连接的N原子一起形成3-7元的杂环基;所述的R7和R8可与它们所连接的C原子一起形成3-8元的环基或3-8元单环杂环基;
R7、R8各自独立地选自氢、卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、芳基、杂芳基、醛基、-C(O)R1、羧基、烯基、炔 基、-OR1、-NR2R3,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
R9独立地选自氢、C1-C8烷基、C3-C8环基、3-8元杂环基、芳基、杂芳基、醛基、-C(O)R1、烯基、炔基,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
当Z1为CCH2OH、CCH2COOH、C-(4-哌啶)时,化合物(2-1)、(2-2)、(2-3)可能以异构体(2-1A)、(2-2A)、(2-3A)的形式存在:
Figure PCTCN2017088038-appb-000003
在本发明的另一个实施方案中,一种通式(I)所示的化合物、其异 构体、前药、稳定的同位素衍生物或其药学上可接受的盐,其特征在于所述的如式I所示的化合物为式IIIa、IIIb、IIIc、IIId、IIIe、IIIf:
Figure PCTCN2017088038-appb-000004
RZ1、RZ2、RZ3、R10、R11各自独立地选自氢、卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、芳基、杂芳基、醛基、-C(O)R1、羧基、烯基、炔基、-OR1、-NR2R3,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
R7独立地选自H、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
R1、R2、R3、R4、R5、R6各自独立地选自氢、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基、烯基、炔基,其中所述的R2和R3、R5和R6可与它们所连接的N原子一起形成3-7元的杂环基;所述的R7和R8可与它们所连接的C原子一起形成3-8元的环基或3-8元单环杂环基;
所述的R2和R3、R5和R6与它们所连接的N原子一起可形成3-8元的杂环基。
在本发明的另一个实施方案中,一种通式(I)所示的化合物、其异构体、前药、稳定的同位素衍生物或其药学上可接受的盐,其特征在于所述的如式I所示的化合物为式IV;
Figure PCTCN2017088038-appb-000005
RZ1、RZ2各自独立地选自氢、卤素、C1-C4烷基、C3-C7环基、4-6元单环杂环基、5-6元单环杂芳基或单环芳基、醛基、酮基、羧基、氰基、OR1、NR2R3,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、羟基、C1-C4烷基、C3-C7环基、4-6元杂环基、芳基或杂芳基的取代基所取代;
R7选自H、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
R10独立地选自氢、卤素、卤代C1-C4烷基、氰基;
R11独立地选自氢、卤素、C1-C4烷基、卤代C1-C4烷氧基、C1-C6烷氧基、HO-C1-C4烷氧基、氰基、NR2R3、C1-C4烷氧基C1-C4烷氧基、C1-C4烷氧基卤代C1-C4烷氧基;
R1、R2、R3各自独立地选自氢、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基、C1-C3烷硫基、被羟基在任意位置取代的卤代烷氧基;
R4、R5、R6各自独立地选自氢、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基、烯基、炔基,其中所述的R2和R3、R5和R6可与它们所连接的N原子一起形成3-7元的杂环基;所述的R7和R8可与它们所连接的C原子一起形成3-8元的环基或3-8元单环杂环基。
在本发明的另一个实施方案中,一种通式(I)所示的化合物、其异 构体、前药、稳定的同位素衍生物或其药学上可接受的盐,其特征在于所述的如式I所示的化合物为式V;
Figure PCTCN2017088038-appb-000006
RZ1、RZ2各自独立地选自氢、卤素、C1-C4烷基、C3-C7环基、5-6元单环杂环基、5-6元单环杂芳基或单环芳基、醛基、羧基,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、羟基、C1-C4烷基、5-6元杂环基、芳基或杂芳基的取代基所取代;
R7选自氢、C1-C4烷基、C3-6环基,其中所述烷基或环基任选被一个或多个选自C1-C3烷基、4-6元单环杂环基的取代基所取代;
R11选自NR2R3、C1-C3烷氧基、-O(CH2)0-1-R4;其中,R4独立地选自氢、C1-C8烷基、HO-C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR5、-OC(O)NR5R6、-C(O)OR5、-C(O)NR5R6、-C(O)R5、-NR5R6、-NR5C(O)R6、-NR7C(O)NR5R6、-S(O)mR5、-NR5S(O)mR6、-SR5、-NR7S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;R2、R3独立地选自氢、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基、C1-C3烷硫基、被羟基在任意位置取代的卤代烷氧基,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR5、-OC(O)NR5R6、-C(O)OR5、-C(O)NR5R6、-C(O)R5、-NR5R6、-NR5C(O)R6、-NR7C(O)NR5R6、-S(O)mR6、-NR5S(O)mR6、-SR5、-NR7S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
R4、R5、R6各自独立地选自氢、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基、烯基、炔基,其中所述的R2和R3、R5和R6可与它们所连接的N原子一起形成3-7元的杂环基;所述的R7和R8可与它们所连接的C原子一起形成3-8元的环基或3-8元单环杂环基。
本发明的典型化合物包括,但并不限于:
Figure PCTCN2017088038-appb-000007
Figure PCTCN2017088038-appb-000008
Figure PCTCN2017088038-appb-000009
Figure PCTCN2017088038-appb-000010
Figure PCTCN2017088038-appb-000011
Figure PCTCN2017088038-appb-000012
Figure PCTCN2017088038-appb-000013
Figure PCTCN2017088038-appb-000014
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、其混合物形式、及其可药用的盐。
本发明化合物为FGFR有效抑制剂,尤其是FGFR4有效选择性抑制剂。因此本发明化合物可用于治疗或者预防FGFR介导性疾病,尤其是FGFR4介导性疾病,包括但不限于癌症和炎症性疾病。本发明化 合物可用于治疗或者预防癌症,例如横纹肌肉瘤、肾细胞癌、骨髓瘤、乳腺癌、胃癌、结肠癌、膀胱癌、胰腺癌和肝细胞癌。本发明化合物特别可以治疗或者预防肝癌、尤其是肝细胞癌。尤其是存在受体酪氨酸激酶的激活突变体或受体酪氨酸激酶的上调的肿瘤将对该类抑制剂特别敏感。
作为FGFR4选择性抑制剂的本发明化合物具有更低的副作用。
本发明进一步涉及一种药物组合物,所述药物组合物含有所述的通式(I)所示的的化合物或其异构体、前药、稳定的同位素衍生物或其药学上可接受的盐及药学上可接受的载体、稀释剂、赋形剂。
本发明还包括制备所述药物组合物的方法,例如将本发明化合物与药学上可接受的载体、稀释剂、赋形剂混合在一起。本发明药物组合物可本领域常规方法制备。
本发明的另一方面涉及通式(I)所示的化合物或其异构体、前药、稳定的同位素衍生物或药学上可接受的盐、及药学上可接受的载体、稀释剂、赋形剂在制备药物中的用途,其中所述药物用于治疗或者预防FGFR、尤其是FGFR4介导的疾病,例如肿瘤或炎症性疾病。
本发明的另一方面涉及通式(I)所示的化合物或其互变异构体、内消旋、外消旋体、对映异构体、非对映异构体、其混合物形式、及其可药用的盐,或所述药物组合物在制备治疗和/或预防肿瘤与炎症等疾病的药物中的用途。
根据本发明,所述药物可以是任何药物剂型,包括但不限于片剂、胶囊剂、溶液剂、冻干制剂、注射剂。本发明的药物制剂可以以每剂量单位包含预定量的活性成分的剂量单位形式给药。这种单位可根据治疗的病症、给药方法和患者的年龄、体重和状况包含例如0.5毫克至1克,优选1毫克至700毫克,特别优选5毫克至300毫克的本发明的化合物,或药物制剂可以以每剂量单位包含预定量的活性成分的剂量单位形式给药。优选剂量单位制剂是包含如上指示的日剂量或分剂量或其相应分数的活性成分的那些。此外,可以使用制药领域中公知的方法制备这种类型的药物制剂。
本发明药物制剂可适于通过任何所需的合适方法给药,例如通过经口(包括口腔或舌下)、直肠、经鼻、局部(包括口腔、舌下或经皮)、***或肠道外(包括皮下、肌内、静脉内或皮内)方法给药。 可以使用制药领域中已知的所有方法通过例如将活性成分与一种或多种赋形剂或一种或多种辅助剂合并来制备这样的制剂。
适合口服给药的药物制剂可作为独立单位,例如胶囊或片剂;粉剂或颗粒剂;在水性或非水液体中的溶液或悬浮液;或水包油液体乳剂或油包水液体乳剂给药。
本发明还涉及一种治疗或者预防FGFR、尤其是FGFR4介导的疾病(例如肿瘤或炎症性疾病)的方法,其包括给予有需要的患者治疗有效量的本发明所述的通式(I)所示的化合物或其异构体、前药、稳定的同位素衍生物或药学上可接受的盐、及药学上可接受的载体、稀释剂、赋形剂。
本发明的另一方面涉及通式(I)所示的化合物或其异构体、前药、稳定的同位素衍生物或药学上可接受的盐及药学上可接受的载体、稀释剂、赋形剂。其用于治疗或者预防FGFR、尤其是FGFR4介导的疾病,例如肿瘤或炎症性疾病。
本发明的另一方面涉及作为治疗和/或预防肿瘤等疾病的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、其混合物形式、及其可药用的盐。
制备流程
本发明还提供制备所述化合物的方法。
流程1
Figure PCTCN2017088038-appb-000015
第一步:化合物(VI-1)结构中Rz2和R7与(VI)结构中是一致的,化 合物(VI-1)利用钯(醋酸钯)催化做插羰,且用1,1-双(二苯基膦)二茂铁做配体,三乙胺做碱,合成化合物(VI-2),化合物(VI-2)中Rz2和R7与(VI)结构中是一致的。
第二步:化合物(VI-2)中的酯被还原试剂(硼氢化钠)还原,合成化合物(VI-3),且化合物(VI-3)结构中Rz2和R7与(VI)结构中是一致的。
第三步:(VI-3)结构中的羟基被取代,利用三溴化磷做为反应试剂,合成化合物(VI-4),且化合物(VI-4)结构中Rz2和R7与(VI)结构中是一致的。
第四步:(VI-4)结构中的溴被亲核试剂(吗啉-3-酮,4-甲基哌嗪-2-酮,等)取代,利用碱(氢化钠)做为去质子试剂,合成化合物(VI-5),且化合物(VI-5)结构中Rz2和R7与(VI)结构中是一致的。
第五步:化合物(VI-5)结构中Rz2和R7与(VI)结构中是一致的,其中的氨基保护基团可以用酸(三氟乙酸)脱去,三乙胺碱化,纯化得到化合物(VI)。
流程2
Figure PCTCN2017088038-appb-000016
第一步:化合物(VI)结构中Rz1,Rz2和R7与(IV)结构中是一致的,化合物(VI)被酰化试剂(碳酸二苯酯或氯甲酸苯酯)活化,合成化合物(VII),化合物(VI)通过酰化合成化合物(VII)是用碱(六甲基二硅基胺基锂)做为去质子试剂。
第二步:2-氨基吡啶环上的R10和R11与(IV)结构中是一致的,并 取代化合物(VII)中的酚基,利用碱(六甲基二硅基胺基锂)做为去质子试剂,合成化合物(VIII),且化合物(VIII)中Rz1,Rz2和R7与(IV)结构中是一致的。
第三步:化合物(VIII)中Rz1,Rz2和R7与(IV)结构中是一致的,其中的缩醛保护基团可以用酸脱去,用碳酸氢钠碱化,纯化得到化合物(IV)。
流程3
Figure PCTCN2017088038-appb-000017
第一步:化合物(VI-1)结构中Rz2和R7与(IV)结构中是一致的,化合物(VI-1)通过钯催化与一些硼酸化合物做碳-碳偶联,合成化合物(VI-5),催化剂([1,1′-双(二苯基膦基)二茂铁]二氯化钯),碱(碳酸钾),且化合物(VI-5)结构中Rz1,Rz2和R10与(IV)结构中是一致的。第二步,第三步和第四步的操作在流程2中已阐述。
定义
除非有相反陈述,否则下列用在说明书和权利要求书中的术语具有下述含义。
在本文中使用的表示方式“Cx-y”表示碳原子数的范围,其中x和y均为整数,例如C3-8环基表示具有3-8个碳原子的环基,-C0-2烷基表示具有0-2个碳原子的烷基,其中-C0烷基是指化学单键。
在本文中,术语“烷基”指饱和的脂族烃基团,包括1至20个碳原子的直链和支链基团,例如可以是1至18个碳原子、1至12个碳原子、1至8个碳原子、1至6个碳原子或1至4个碳原子的直链和支链基团。非限制性实施例包括甲基、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。烷基可以是取代的或未取代的。
在本文中,术语“环基”指饱和或部分不饱和单环或多环环状烃基团,其包括3至12个环碳原子,例如可以是3至12个、3至10个、3至8个或3至6个环碳原子,或者可以是3、4、5、6、7、8元环。单环环基的非限制性实施例包含环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等。环基可以是取代的或未取代的。
在本文中,术语“杂环基”指饱和或部分不饱和单环或多环环状基团,其包括3至20个环碳原子,例如可以是3至16个、3至12个、3至10个、3至8个或3至6个环原子,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,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包括3至12个环原子,其中1~4个是杂原子,更优选杂环基环包含3至10个环原子,最优选5元环或6元环,其中1~4个是杂原子,更优选1~3个是杂原子,最优选1~2个是杂原子。单环杂环基的非限制性实施例包含吡咯烷基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基等。多环杂环基包括螺环、稠环和桥环的杂环基。
在本文中,术语“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,但没有一个环具有完全共扼的π电子***。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,优选为单螺环基和双螺环基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺环基。螺环基的非限制性实施例包含
Figure PCTCN2017088038-appb-000018
在本文中,术语“稠杂环基”指5至20元,***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,但没有一个环具有完全共扼的π电子***,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施例包含
Figure PCTCN2017088038-appb-000019
所述杂环基环可以稠合于芳基、杂芳基或环基环上,其中与母体结构连接在一起的环为杂环基,非限制性实施例包含:
Figure PCTCN2017088038-appb-000020
等。杂环基可以是取代的或未取代的。
在本文中,术语“芳基”指6至14元全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,具有共扼的π电子体系的多环(即其带有相邻对碳原子的环)基团,优选为6至10元,例如苯基和萘基,最优选苯基。所述芳基环可以稠合于杂芳基、杂环基或环基环上,其中与母体结构连接在一起的环为芳基环,非限制性实施例包含:
Figure PCTCN2017088038-appb-000021
芳基可以是取代的或未取代的。
在本文中,术语“杂芳基”指包含1至4个杂原子,5至14个环原子的杂芳族体系,其中杂原子包括氧、硫和氮。优选为5至10元。更 优选杂芳基是5元或6元,例如呋喃基、噻吩基、吡啶基、吡咯基、N-烷基吡咯基、嘧啶基、吡嗪基、咪唑基、四唑基、噁唑基、异噁唑基等,所述杂芳基环可以稠合于芳基、杂环基或环基环上,其中与母体结构连接在一起的环为杂芳基环,非限制性实施例包含:
Figure PCTCN2017088038-appb-000022
杂芳基可以是取代的或未取代的。
在本文中,术语“卤素”指氟、氯、溴或碘。
在本文中,术语“氰基”指-CN。
在本文中,术语“烯基”指含有至少1个碳碳双键的直链、支链或者环状非芳香烃基。其中可以存在1-3个碳碳双键,优选存在1个碳碳双键。包括乙烯基、丙烯基、丁烯基、2-甲基丁烯基和环己烯基。所述的烯基可以被取代。术语“C2-4烯基”是指具有2-4个碳原子的烯基。
在本文中,术语“炔基”是指含有至少1个碳碳三键的直链、支链或者环状烃基。其中可以存在1-3个碳碳三键,优选存在1个碳碳三键。包括乙炔基、丙炔基、丁炔基和3-甲基丁炔基。术语“C2-4炔基”是指具有2-4个碳原子的炔基。
在本文中,术语“烷氧基”指通过氧桥连接的具有所述碳原子数目的环状或者非环状烷基,包含烷基氧基、环烷基氧基和杂环烷基氧基。由此,“烷氧基”包含上述烷基、杂环烷基和环烷基的定义“任选”或“任选地”意味着随后所描述地事件或环境可以但不必发生,该说明包括该事件或环境发生或不发生地场合。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。
在本文中,术语“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。
在本文中,术语“药物组合物”表示含有一种或多种本文所述化合物或其生理学上/可药用的盐或前体药物与其他化学组分的混合物,以及其他组分例如生理学/可药用的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
本发明所述“室温”是指15-30℃。
在本文中,术语“稳定的同位素衍生物”包括:式I中任意的氢原子被1-5个氘原子取代得到的同位素取代衍生物、式I中任意的碳原子被1-3个碳14原子取代得到的同位素取代衍生物或式I中任意的氧原子被1-3个氧18原子取代得到的同位素取代衍生物。
本发明所述的“药学上可接受的盐”在Berge,et al.,“Pharmaceutically acceptable salts”,J.Pharm.Sci.,66,1-19(1977)中有讨论,并对药物化学家来说是显而易见,所述的盐是基本上无毒性的,并能提供所需的药代动力学性质、适口性、吸收、分布、代谢或***等。
本发明药学上可接受的盐可通过一般的化学方法合成。
一般情况下,盐的制备可以通过游离碱或酸与等化学当量或者过量酸(无机酸或有机酸)或碱在合适的溶剂或溶剂组合物中反应制得。
本发明所述的“前药”是指化合物在体内代谢后转换成原始活性化合物。代表性地讲,前药为非活性物质,或者比活性母体化合物活性小,但可以提供方便的操作、给药或者改善代谢特性。
本发明所述“异构体”是指本发明的式(I)化合物可以有不对称中心和外消旋体、外消旋混合物和单个非对映异构体,所有这些异构体,包括立体异构体、几何异构体均包含在本发明中。所述几何异构体包括顺反异构体。
在本文中,术语“肿瘤”包括良性肿瘤和恶性肿瘤,例如癌症。
在本文中,术语“癌症”包括各种恶性肿瘤,尤其是FGFR、尤其是FGFR4参与其发生的恶性肿瘤,包括但不限于横纹肌肉瘤、肾细胞癌、骨髓瘤、乳腺癌、胃癌、结肠癌、膀胱癌、胰腺癌和肝细胞癌。
在本文中,术语“炎症性疾病”是指FGFR、尤其是FGFR4参与其炎症发生的任何炎性疾病。
实施例
下面通过实施例的方式进一步说明本发明,但并不因此将本发明 限制在所述的实施例范围之中。下列实施例中未注明具体条件的实验方法,按照常规方法和条件,或按照商品说明书选择。
本发明所有化合物的结构可通过核磁共振(1H NMR)和/或质谱检测(MS)鉴定。
1H NMR化学位移(δ)以PPM记录(单位:10-6PPM)。NMR通过Bruker AVANCE-400光谱仪进行。合适的溶剂是氘代氯仿(CDCl3),氘代甲醇(CD3OD),氘代二甲亚砜(DMSO-d6),四甲基硅烷作为内标(TMS)。
低分辨率质谱(MS)由Agilent 1260HPLC/6120质谱仪测定,使用Agilent ZORBAX XDB-C18,4.6×50mm,3.5μm,梯度洗脱条件一:0∶95%溶剂A1和5%溶剂B1,1-2∶5%溶剂A1和95%溶剂B1;2.01-2.50∶95%溶剂A1和5%溶剂B1。百分数为某一溶剂占总溶剂体积的体积百分数。溶剂A1:0.01%甲酸水溶液;溶剂B1:0.01%甲酸的乙腈溶液;百分数为溶质占溶液的体积百分数。
薄层硅胶板是烟台黄海HSGF254或青岛GF254硅胶板。柱层析一般使用烟台黄海100-200或200-300目硅胶作为载体。
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Acros Organics,Aldrich Chemical Company,韶远化学科技(Accela ChemBio Inc)、上海毕得医药、上海阿拉丁化学、上海迈瑞尔化学、百灵威化学等公司。
实施例中如无特殊说明,反应所用溶剂均为无水溶剂,其中无水四氢呋喃使用市售四氢呋喃,以钠块为除水剂,以二苯甲酮作为指示剂,氮气保护下回流至溶液呈蓝紫色,蒸馏收集,氮气保护下室温储存,其他无水溶剂购自阿拉丁化学及百灵威化学,所有无水溶剂的转移和使用如无特殊说明,均需在氮气保护下进行。
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。
一氧化碳气氛是指反应瓶连接一个约1L容积的一氧化碳气球;
氢化反应通常抽真空,充入氢气,反复操作3次。
实施例中如无特殊说明,反应的温度为室温,温度范围是15℃-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,也可以加入少量的三乙胺和酸性或碱性试剂等进行调节。
实施例1
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000023
Figure PCTCN2017088038-appb-000024
第一步
6-氯-2-羟甲基吡啶
将化合物6-氯-2-吡啶甲酸甲酯1a(1.00g,5.85mmol)、硼氢化钠(0.38g,9.95mmol)和乙醇(15mL)混合,室温下搅拌6小时。此混合物用30mL水淬灭,用乙酸乙酯(50mL×2)萃取,有机相用饱和食盐水(50mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶得到目标产物6-氯-2-羟甲基吡啶1b(0.70g,黄色油)。产率:84%。产物不经纯化直接用于下一步反应。
1H NMR(400MHz,CDCl3)δ7.68(dd,J=8.0,7.6Hz,1H),7.27(d,J=8.0Hz,1H),7.26(d,J=7.6Hz,1H),4.76(d,J=5.2Hz,2H),3.07(t,J=5.6Hz,1H)。
第二步
(6-(甲基氨基)吡啶-2-基)甲醇
将化合物6-氯-2-羟甲基吡啶1b(1.50g,10.5mmol)和甲胺(15mL,30%的乙醇溶液)混合,100℃搅拌48小时。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10∶1-1∶2),得到目标产物(6-(甲基氨基)吡啶-2-基)甲醇1c(0.70g,黄色油),产率:48%。
MS m/z(ESI):139[M+1]。
第三步
6-(甲基氨基)甲基吡啶醛
将化合物(6-(甲基氨基)吡啶-2-基)甲醇1c(0.60g,4.35mmol),二氧化锰(3.78g,43.5mmol)和二氯甲烷(15mL)混合,40℃搅拌16小时。冷却至室温,过滤,滤液减压脱溶,得到目标产物6-(甲基氨基)甲基吡啶醛1d(0.50g,黄色固体),产率:72%。
MS m/z(ESI):137[M+1]。
第四步
6-(1,3-二噁戊环-2-基)-N-甲基吡啶-2-胺
将化合物6-(甲基氨基)甲基吡啶醛1d(0.80g,5.95mmol),乙二醇(1.80g,29.7mmol),对甲苯磺酸(0.10g,0.60mmol),4A分子筛(0.2g),和甲苯(15mL)混合,120℃搅拌5小时。冷却至室温,此混合物用30mL水稀释,用乙酸乙酯(50mL×2)萃取,有机相用饱和食 盐水(50mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯6∶1-2∶1),得到目标产物6-(1,3-二噁戊环-2-基)-N-甲基吡啶-2-胺(0.60g,黄色固体),产率:57%。
MS m/z(ESI):181[M+1]
1H NMR(400MHz,CDCl3)δ7.51(t,J=8.0Hz,1H),6.83(d,J=7.2Hz,1H),6.39(d,J=8.0Hz,1H),5.72(s,1H),4.66(brs,1H),4.20-4.14(m,2H),4.09-4.03(m,2H),2.93(d,J=4.8Hz,3H)。
第五步
苯基(6-(1,3-二噁戊环-2-基)吡啶-2-基)(甲基)氨基羧酸酯
将化合物6-(1,3-二噁戊环-2-基)-N-甲基吡啶-2-胺1e(54mg,0.30mmol),碳酸二苯酯(1.28g,0.60mmol),六甲基二硅基胺基锂(0.41mL,0.41mmol,1M四氢呋喃溶液)和四氢呋喃(3mL)混合,0℃搅拌2小时。此混合物用10mL饱和氯化铵溶液淬灭,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯4∶1),得到目标产物(6-(1,3-二噁戊环-2-基)吡啶-2-基)(甲基)氨基羧酸酯(60mg,白色固体),产率:67%。
MS m/z(ESI):301[M+1]
1H NMR(400MHz,CDCl3)δ7.96-7.94(m,1H),7.74-7.70(m,1H),7.45-7.32(m,2H),7.40-7.38(m,1H),7.28-7.25(m,1H),7.20-7.17(m,2H),5.87(s,1H),4.24-4.21(m,2H),4.13-4.09(m,2H),3.67(s,3H)。
第六步
1-(6-(1,3-二噁戊环-2-基)吡啶-2-基)-3-(4-氯-5-氰基吡啶-2-基)-1-甲基脲
将化合物苯基(6-(1,3-二噁戊环-2-基)吡啶-2-基)(甲基)氨基羧酸酯1f(60mg,0.20mmol),6-氨基-4-氯尼古丁腈(76mg,0.50mmol),六甲基二硅基胺基锂(0.4mL,0.4mmol,1M四氢呋喃溶液)和四氢呋喃(2mL)混合,室温搅拌2小时。此混合物用10mL饱和氯化铵溶液淬灭,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(二氯甲烷/甲醇30∶1),得到目标产物1-(6-(1,3-二噁戊环 -2-基)吡啶-2-基)-3-(4-氯-5-氰基吡啶-2-基)-1-甲基脲(22mg,白色固体),产率:31%。
MS m/z(ESI):360&362[M+1]
1H NMR(400MHz,DMSO-d6)δ13.44(s,1H),8.87(s,1H),8.33(s,1H),8.04-8.00(m,1H),7.42(d,J=8.4Hz,1H),7.36(d,J=7.6Hz,1H),5.82(s,1H),4.24-4.21(m,2H),4.04-4.01(m,2H),3.44(s,3H)。
第七步
1-(6-(1,3-二噁戊环-2-基)吡啶-2-基)-3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-甲基脲
将化合物1-(6-(1,3-二噁戊环-2-基)吡啶-2-基)-3-(4-氯-5-氰基吡啶-2-基)-1-甲基脲1g(22mg,0.06mmol),2-甲氧基乙胺(14mg,0.18mmol),二异丙基乙胺(24mg,0.18mmol)和N,N-二甲基乙酰胺(0.4mL)混合,70℃搅拌16小时。此混合物用10mL水稀释,用二氯甲烷(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯2∶1),得到目标产物1-(6-(1,3-二噁戊环-2-基)吡啶-2-基)-3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-甲基脲(10mg,白色固体),产率:41%。
MS m/z(ESI):399[M+1]。
第八步
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基吡啶-2-基)-1-甲基脲
将化合物1-(6-(1,3-二噁戊环-2-基)吡啶-2-基)-3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-甲基脲1h(10mg,0.025mmol),盐酸(0.5mL,37%),水(1mL)和四氢呋喃(2mL)混合,室温搅拌6小时。此混合物用饱和的碳酸钠溶液淬灭,用二氯甲烷(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(二氯甲烷/甲醇20∶1),得到目标产物3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基吡啶-2-基)-1-甲基脲(8mg,白色固体),产率:90%。
MS m/z(ESI):355[M+1]
1H NMR(400MHz,CDCl3)δ13.03(s,1H),10.18(s,1H),8.21(s, 1H),8.02-7.98(m,1H),7.75(d,J=7.6Hz,1H),7.59(s,1H),7.36(d,J=8.4Hz,1H),5.82(s,1H),3.66(t,J=4.8Hz,2H),3.56(s,3H),3.52-3.50(m,2H),3.44(s,3H)。
实施例2
3-(6-氯嘧啶-4-基)-1-(6-甲酰基吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000025
参照实施例1的操作步骤合成实施例2,但在第六步中用6-氯嘧啶-4-胺取代6-氨基-4-氯尼古丁腈。
MS m/z(ESI):292&294[M+1]
1H NMR(400MHz,CDCl3)δ13.51(s,1H),10.18(s,1H),8.67(s,1H),8.25(s,1H),8.02(dd,J=8.4,7.6Hz,1H),7.79(d,J=7.6Hz,1H),7.40(d,J=8.4Hz,1H),3.59(s,3H)。
实施例3
3-(5-氰基吡啶-2-基)-1-(6-甲酰基吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000026
参照实施例1的操作步骤合成实施例3,但在第六步中用6-氨基尼古丁腈取代6-氨基-4-氯尼古丁腈。
MS m/z(ESI):282[M+1]
1H NMR(400MHz,CDCl3)δ13.49(s,1H),10.19(s,1H),8.60(s,1H),8.34(d,J=7.6Hz,1H),8.03(dd,J=8.8,7.6Hz,1H),7.95-7.93(m,1H),7.77(d,J=7.6Hz,1H),7.39(d,J=8.8Hz,1H),3.59(s,3H)。
实施例4
3-(4-氯-5-氰基吡啶-2-基)-1-(6-甲酰基吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000027
参照实施例1的操作步骤合成实施例4。但在第六步中用4-氯吡啶-2-胺取代6-氨基-4-氯尼古丁腈。
MS m/z(ESI):291&293[M+1]
1H NMR(400MHz,CDCl3)δ13.09(s,1H),10.19(s,1H),8.29(d,J=2.0Hz,1H),8.22(d,J=5.2Hz,1H),8.00(dd,J=8.4,7.2Hz,1H),7.74(d,J=7.2Hz,1H),7.37(d,J=8.4Hz,1H),7.03(dd,J=5.2,2.0Hz,1H),3.58(s,3H)。
实施例5
3-(5-氰基-4-(异丙基氨基)吡啶-2-基)-1-(6-甲酰基吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000028
参照实施例1的操作步骤合成实施例5,但在第七步中用异丙胺取代2-甲氧基乙胺。
MS m/z(ESI):339[M+1]
1H NMR(400MHz,CDCl3)δ13.00(s,1H),10.18(s,1H),8.20(s,1H),8.02-7.97(m,1H),7.76-7.74(m,1H),7.60(s,1H),7.57-7.54(m,1H),7.36(d,J=8.4Hz,1H),3.92-3.89(m,1H),3.57(s,3H),1.34(d,J=6.4Hz,6H)。
实施例6
3-(5-氰基-4-异丙氧基吡啶-2-基)-1-(6-甲酰基吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000029
第一步
6-氨基-4-异丙氧基尼古丁腈
将化合物6-氨基-4-氯尼古丁腈6a(46mg,0.30mmol)、异丙醇(90mg,1.50mmol),氢化钠(72mg,1.80mmol,60%矿物油混合物)和N-甲基吡咯烷酮(1.5mL)混合,70℃下搅拌24小时。冷却至室温,此混合物用20mL水淬灭,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用制备硅胶板纯化(石油醚/乙酸乙酯1∶1),得到目标产物6-氨基-4-异丙氧基尼古丁腈6b(16mg,黄色固体)。产率:30%。
MS m/z(ESI):178[M+1]。
第二步
1-(6-(1,3-二噁戊环-2-基)吡啶-2-基)-3-(5-氰基-4-异丙氧基吡啶-2-基)-1-甲基脲
参照实施例1第六步的操作步骤合成实施例6c,用6-氨基-4-异丙氧基尼古丁腈取代6-氨基-4-氯尼古丁腈。得到目标产物1-(6-(1,3-二噁 戊环-2-基)吡啶-2-基)-3-(5-氰基-4-异丙氧基吡啶-2-基)-1-甲基脲6c(8mg,白色固体)。产率:46%。
MS m/z(ESI):384[M+1]。
第三步
3-(5-氰基-4-异丙氧基吡啶-2-基)-1-(6-甲酰基吡啶-2-基)-1-甲基脲
参照实施例1第八步的操作步骤合成实施例6。用1-(6-(1,3-二噁戊环-2-基)吡啶-2-基)-3-(5-氰基-4-异丙氧基吡啶-2-基)-1-甲基脲取代1-(6-(1,3-二噁戊环-2-基)吡啶-2-基)-3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-甲基脲。得到目标产物3-(5-氰基-4-异丙氧基吡啶-2-基)-1-(6-甲酰基吡啶-2-基)-1-甲基脲7(5mg,白色固体)。产率:71%。
MS m/z(ESI):340[M+1]
1H NMR(400MHz,CDCl3)δ13.33(s,1H),10.19(s,1H),8.38(s,1H),8.02(dd,J=8.0,7.6Hz,1H),7.96(s,1H),7.77(d,J=7.6Hz,1H),7.38(d,J=8.0Hz,1H),4.88-4.86(m,1H),3.58(s,3H),1.48(d,J=6.0Hz,6H)。
实施例7
3-(5-氰基-4-(2-甲氧基乙氧基)吡啶-2-基)-1-(6-甲酰基吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000030
参照实施例6的操作步骤合成实施例7,但在第一步中用2-甲氧基乙二醇胺取代异丙醇。
MS m/z(ESI):356[M+1]
1H NMR(400MHz,CDCl3)δ13.38(s,1H),10.18(s,1H),8.40(s,1H),8.02(dd,J=8.4,7.2Hz,1H),7.99(s,1H),7.77(d,J=7.2Hz,1H),7.38(d,J=8.4Hz,1H),4.38(t,J=4.8Hz,2H),3.86(t,J=4.8Hz,2H),3.58(s,3H),3.51(s,3H)。
实施例8
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(4-甲酰基嘧啶-2-基)-1-甲基脲、
Figure PCTCN2017088038-appb-000031
第一步
4-(二甲氧基甲基)嘧啶-2-胺
将化合物1,1-二甲氧基-N,N-二甲基甲胺8a(4.90g,41.36mmol)和1,1-二甲氧基丙烷-2-酮(4.90g,41.36mmol)混合,此混合物在100℃搅拌16小时,减压脱溶,残余物与盐酸胍(4.30g,45.00mmol),氢氧化钠(1.80g,45.00mmol)和水(15mL)混合,室温下搅拌48小时。过滤,得到目标产物4-(二甲氧基甲基)嘧啶-2-胺8b(2.00g,白色固体)。产率:27%。
MS m/z(ESI):170[M+1]
1H NMR(400MHz,CDCl3)δ8.36(d,J=4.8Hz,1H),6.87(d,J=5.2Hz,1H),5.16(s,1H),5.15(brs,2H),3.42(s,6H)。
第二步
4-(二甲氧基甲基)-N-甲基嘧啶-2-胺
将化合物4-(二甲氧基甲基)嘧啶-2-胺8b(1.00g,5.65mmol),碘甲烷(2.80g,19.77mmol)和丙酮(30mL)混合,此混合物在70℃搅拌16小时,冷却至室温,过滤,固体和10%的氢氧化钠(8mL)混合,80℃搅拌0.5小时,冷却至室温,此混合物用250mL冰水淬灭,用二氯甲烷(50mL×2)萃取,有机相用饱和食盐水(50mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,得到目标产物4-(二甲氧基甲基)-N-甲基嘧啶-2-胺8c(0.70g,黄色油)。产率:67%。
MS m/z(ESI):184[M+1]
1H NMR(400MHz,CDCl3)δ8.37(d,J=4.8Hz,1H),6.77(d,J=5.2Hz,1H),5.18(brs,1H),5.13(s,1H),3.42(s,6H),3.03(d,J=5.2Hz,3H)。
第三步
苯基(4-(二甲氧基甲基)嘧啶-2-基)(甲基)氨基羧酸酯
将化合物4-(二甲氧基甲基)-N-甲基嘧啶-2-胺8c(0.20g,1.09mmol),碳酸二苯酯(0.47g,2.19mmol),六甲基二硅基胺基锂(1.5mL,1.51mmol,1M四氢呋喃溶液)和四氢呋喃(5mL)混合,0℃搅拌2小时。此混合物用10mL饱和氯化铵溶液淬灭,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(二氯甲烷/甲醇50∶1),得到目标产物苯基(4-(二甲氧基甲基)嘧啶-2-yl)(甲基)氨基羧酸酯8d(40mg,白色固体)。产率:12%。
MS m/z(ESI):304[M+1]。
第四步
3-(4-氯-5-氰基吡啶-2-基)-1-(4-(二甲氧基甲基)嘧啶-2-基)-1-甲基脲
将化合物苯基(4-(二甲氧基甲基)嘧啶-2-yl)(甲基)氨基羧酸酯8d(40mg,0.13mmol),6-氨基-4-氯尼古丁腈(21mg,0.13mmol),六甲基二硅基胺基锂(0.26mL,0.26mmol,1M四氢呋喃溶液)和四氢呋喃(2mL)混合,室温搅拌2小时。此混合物用10mL饱和氯化铵溶液淬灭,用二氯甲烷(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将 有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(二氯甲烷/甲醇50∶1),得到目标产物3-(4-氯-5-氰基吡啶-2-基)-1-(4-(二甲氧基甲基)嘧啶-2-基)-1-甲基脲8e(25mg,白色固体)。产率:52%。
MS m/z(ESI):363&365[M+1]
1H NMR(400MHz,CDCl3)δ13.59(s,1H),8.74(d,J=5.2Hz,1H),8.56(s,1H),8.55(s,1H),7.30(d,J=5.2Hz,1H),5.34(s,1H),3.68(s,3H),3.51(s,6H)。
第五步
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(4-(二甲氧基甲基)嘧啶-2-基)-1-甲基脲
将化合物3-(4-氯-5-氰基吡啶-2-基)-1-(4-(二甲氧基甲基)嘧啶-2-基)-1-甲基脲8e(18mg,0.05mmol),2-甲氧基乙胺(15mg,0.20mmol),二异丙基乙胺(13mg,0.10mmol)和N,N-二甲基乙酰胺(0.4mL)混合,70℃搅拌16小时。此混合物用10mL水稀释,用二氯甲烷(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(二氯甲烷/甲醇50∶1),得到目标产物3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(4-(二甲氧基甲基)嘧啶-2-基)-1-甲基脲8f(15mg,黄色固体)。产率:75%。
MS m/z(ESI):402[M+1]
1H NMR(400MHz,CDCl3)δ13.11(s,1H),8.72(d,J=5.2Hz,1H),8.23(s,1H),7.65(s,1H),7.25(d,J=5.2Hz,1H),5.31(s,1H),5.30(brs,1H),3.67-3.65(m,2H),3.66(s,3H),3.54-3.52(m,2H),3.51(s,6H),3.44(s,3H)。
第六步
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(4-甲酰基嘧啶-2-基)-1-甲基脲
将化合物3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(4-(二甲氧基甲基)嘧啶-2-基)-1-甲基脲8f(15mg,0.04mmol),盐酸(0.8mL,37%),水(1mL)和四氢呋喃(2mL)混合,室温搅拌3小时。此混合物用饱和的碳酸钠溶液淬灭,用二氯甲烷(20mL×2)萃取,有机相用 饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(二氯甲烷/甲醇50∶1),得到目标产物3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(4-甲酰基嘧啶-2-基)-1-甲基脲8(6mg,白色固体)。产率:27%。
MS m/z(ESI):356[M+1]
1H NMR(400MHz,CDCl3)δ13.88(s,1H),10.06(s,1H),8.95(d,J=4.8Hz,1H),8.23(s,1H),7.65(s,1H),7.52(d,J=4.8Hz,1H),5.35(brs,1H),3.73(s,3H),3.69-3.65(m,2H),3.55-3.52(m,2H),3.45(s,3H)。
实施例9
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基-5-((3-羰基吗啉代)甲基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000032
第一步
二叔丁基(5-溴-6-甲基吡啶-2-基)酰亚胺基二碳酸酯
将化合物5-溴-6-甲基吡啶-2-胺9a(9.30g,50.00mmol)、2-碳酸-2-叔丁酯(27.00g,125mmol),N,N-二甲基吡啶-4-胺(0.31g,2.50mmol)和四氢呋喃(300mL)混合,室温下搅拌16小时。此混合物用300mL水淬灭,用乙酸乙酯(200mL×2)萃取,有机相用饱和食盐水(200mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯100∶0-95∶5),得到目标产物二叔丁基(5-溴-6-甲基吡啶-2-基)酰亚胺基二碳酸酯9b(15.00g,白色固体)。产率:78%。
1H NMR(400MHz,CDCl3)δ7.80(d,J=8.4Hz,1H),7.00(d,J=8.4Hz,1H),2.61(s,3H),1.46(s,18H)。
第二步
二叔丁基(5-溴-6-(二溴甲基)吡啶-2-基)酰亚胺基二碳酸酯
将化合二叔丁基(5-溴-6-甲基吡啶-2-基)酰亚胺基二碳酸酯9b(3.86g,10.00mmol),1-溴吡咯烷-2,5-二酮(4.45g,25.00mmol),苯甲过氧酸酐(0.24g,1.00mmol)和四氯化碳(100mL)混合,90℃搅拌16小时。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94∶6),得到目标产物二叔丁基(5-溴-6-(二溴甲基)吡啶-2-基)酰亚胺基二碳酸酯9c(4.00g,黄色固体),产率:74%。
1H NMR(400MHz,CDCl3)δ7.82(d,J=8.0Hz,1H),7.26(d,J=8.0Hz,1H),7.08(s,1H),1.50(s,18H)。
第三步
叔丁基(5-溴-6-(二甲氧基)吡啶-2-基)氨基羧酸酯
将化合物二叔丁基(5-溴-6-(二溴甲基)吡啶-2-基)酰亚胺基二碳酸酯9c(5.00g,96.00mmol),氢氧化钾(2.23g,0.38mol)和甲醇(30mL)混合,70℃搅拌16小时。冷却至室温,减压脱溶,残余物用50mL水溶解,用乙酸乙酯(50mL×3)萃取,有机相用饱和食盐水(50mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯100∶0-12∶1),得到目标产物叔丁基(5-溴-6-(二甲氧基)吡啶-2-基)氨基羧酸酯9d(0.50g,黄色固体),产率:16%。
MS m/z(ESI):347&349[M+1]。
第四步
叔丁基(5-溴-6-(二甲氧基)吡啶-2-基)(甲基)氨基羧酸酯
将化合物叔丁基(5-溴-6-(二甲氧基)吡啶-2-基)氨基羧酸酯9d(1.20g,3.47mmol),氢化钠(0.18g,4.51mmol,60%矿物油混合物),碘甲烷(0.59g,4.16mmol)和N,N-二甲基甲酰胺(8mL)混合,室温搅拌16小时。此混合物用30mL水淬灭,用乙酸乙酯(50mL×3)萃取,有机相用饱和食盐水(50mL×3)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯100∶0-96∶4),得到目标产物叔丁基(5-溴-6-(二甲氧基)吡啶-2-基)(甲基)氨基羧酸酯9e(0.40g,黄色油),产率:32%。
MS m/z(ESI):361&363[M+1]。
第五步
甲基-6-((叔丁氧基羰基)(甲基)氨基)-2-(二甲氧基甲基)尼古丁酸酯
将化合物叔丁基(5-溴-6-(二甲氧基)吡啶-2-基)(甲基)氨基甲酸9e(0.45g,1.25mmol),醋酸钯(28mg,0.13mmol),1,1-双(二苯基膦)二茂铁(0.14g,0.25mmol),三乙胺(0.25g,2.50mmol),甲醇(3mL)和N,N-二甲基甲酰胺(20mL)混合,在一氧化碳气氛(1大气压)下100℃搅拌16小时。此混合物用100mL水淬灭,用乙酸乙酯(50mL×3)萃取,有机相用饱和食盐水(50mL×3)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯94∶6),得到目标产物甲基-6-((叔丁氧基羰基)(甲基)氨基)-2-(二甲氧基甲基)尼古丁酸酯9f(0.25g,黄色油),产率:59%。
MS m/z(ESI):341[M+1]
1H NMR(400MHz,CDCl3)δ8.07(d,J=8.8Hz,1H),7.84(d,J=8.8Hz,1H),6.08(s,1H),3.91(s,3H),3.52(s,6H),3.41(s,3H),1.53(s,9H)。
第六步
叔丁基(6-(二甲氧基甲基)-5-(羟甲基)吡啶-2-基)(甲基)氨基羧酸酯
将化合物甲基-6-((叔丁氧基羰基)(甲基)氨基)-2-(二甲氧基甲基)尼古丁酸酯9f(0.30g,0.88mmol),硼氢化钠(0.67g,17.65mmol),无水氯化钙(0.19g,1.77mmol)和甲醇(10mL)混合,65℃搅拌8小时。此混合物用10mL水淬灭,用乙酸乙酯(50mL×2)萃取,有机相用饱和食盐 水(50mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,得到目标产物叔丁基(6-(二甲氧基甲基)-5-(羟甲基)吡啶-2-基)(甲基)氨基羧酸酯9g(0.20g,白色固体),产率:73%。
MS m/z(ESI):313[M+1]。
第七步
叔丁基(5-(溴甲基)-6-(二甲氧基甲基吡啶-2-基)(甲基)氨基羧酸酯
将化合物叔丁基(6-(二甲氧基甲基)-5-(羟甲基)吡啶-2-基)(甲基)氨基羧酸酯9g(0.20g,0.64mmol),三溴化磷(0.21g,0.77mmol)和二氯甲烷(5mL)混合,0℃搅拌1小时。此混合物用10mL碳酸氢钠水溶液淬灭,用乙酸乙酯(50mL×2)萃取,有机相用饱和食盐水(50mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯100∶0-93∶7),得到目标产物叔丁基(5-(溴甲基)-6-(二甲氧基甲基吡啶-2-基)(甲基)氨基羧酸酯9h(0.15g,无色固体),产率:63%。
MS m/z(ESI):375&377[M+1]。
第八步
叔丁基-(6-(2-甲氧基乙基)-5-((3-羰基吗啉)甲基)吡啶-2-基)(甲基)氨基羧酸酯
将化合物叔丁基(5-(溴甲基)-6-(二甲氧基甲基吡啶-2-基)(甲基)氨基羧酸酯9h(70mg,0.19mmol),吗啉-3-酮(38mg,0.38mmol),氢化钠(19mg,0.47mmol,60%矿物油混合物)和N,N-二甲基甲酰胺(3mL)混合,室温搅拌1小时。此混合物用水淬灭,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯1.5∶1),得到目标产物叔丁基-(6-(2-甲氧基乙基)-5-((3-羰基吗啉)甲基)吡啶-2-基)(甲基)氨基羧酸酯9i(70mg,白色固体),产率:95%。
MS m/z(ESI):396[M+1]
1H NMR(400MHz,CDCl3)δ7.63-7.61(m,2H),5.22(s,1H),4.91(s,2H),4.26(s,2H),3.82-3.81(m,2H),3.45(s,6H),3.40(s,3H),3.27-3.26(m,2H),1.52(s,9H)。
第九步
4-((2-(二甲氧基甲基)-6-(甲基氨基)吡啶-3-基)甲基)吗啉-3-酮
将化合物叔丁基-(6-(2-甲氧基乙基)-5-((3-羰基吗啉)甲基)吡啶-2-基)(甲基)氨基羧酸酯9i(70mg,0.18mmol),三氟乙酸(1mL)和二氯甲烷(4mL)混合,室温搅拌6小时。用三乙胺碱化,减压脱溶,残余物用制备硅胶板纯化(石油醚/乙酸乙酯1∶1),得到目标产物4-((2-(二甲氧基甲基)-6-(甲基氨基)吡啶-3-基)甲基)吗啉-3-酮9j(46mg,无色固体),产率:86%。
MS m/z(ESI):296[M+1]。
第十步
苯基-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)(甲基)氨基羧酸酯
将化合物4-((2-(二甲氧基甲基)-6-(甲基氨基)吡啶-3-基)甲基)吗啉-3-酮9j(60mg,0.20mmol),碳酸二苯酯(87mg,0.40mmol),六甲基二硅基胺基锂(1.0mL,1.01mmol,1M四氢呋喃溶液)和四氢呋喃(5mL)混合,0℃搅拌0.5小时。此混合物用10mL饱和氯化铵溶液淬灭,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯2∶1),得到目标产物苯基(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)(甲基)氨基羧酸酯9k(45mg,无色油),产率:54%。
MS m/z(ESI):416[M+1]。
第十一步
3-(4-氯-5-氰基吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲
将化合物苯基(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)(甲基)氨基羧酸酯9k(45mg,0.11mmol),6-氨基-4-氯尼古丁腈(33mg,0.22mmol),六甲基二硅基胺基锂(0.3mL,0.33mmol,1M四氢呋喃溶液)和四氢呋喃(3mL)混合,室温搅拌1小时。此混合物用10mL饱和氯化铵溶液淬灭,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯1∶1),得到目标产物3-(4-氯-5-氰基吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲9l(40mg,白色固体),产率:78%。
MS m/z(ESI):475&477[M+1]。
第十二步
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲
将化合物3-(4-氯-5-氰基吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲91(20mg,0.04mmol),2-甲氧基乙胺(13mg,0.17mmol),二异丙基乙胺(11mg,0.08mmol)和N,N-二甲基乙酰胺(0.4mL)混合,50℃搅拌16小时。此混合物用10mL水稀释,用二氯甲烷(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯1∶1),得到目标产物3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲9m(15mg,白色固体),产率:69%。
MS m/z(ESI):514[M+1]。
第十三步
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲
将化合物3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲9m(15mg,0.03mmol),盐酸(0.8mL,37%),水(1mL)和四氢呋喃(2mL)混合,室温搅拌1小时。此混合物用饱和的碳酸钠溶液淬灭,用二氯甲烷(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用乙酸乙酯洗,得到目标产物3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲9(7mg,白色固体),产率:52%。
MS m/z(ESI):468[M+1]
1H NMR(400MHz,CDCl3)δ12.99(s,1H),10.26(s,1H),8.17(s,1H),7.98(d,J=8.4Hz,1H),7.56(s,1H),7.27(d,J=8.4Hz,1H),5.31(brs,1H),5.13(s,2H),4.26(s,2H),3.89(t,J=4.4Hz,2H),3.61(t,J=4.0Hz,2H),3.53(s,3H),3.51(t,J=4.4Hz,2H),3.42(s,3H),3.41(d,J=4.0Hz,2H)。
实施例10
3-(5-氰基-4-(异丙基氨基)吡啶-2-基)-1-(6-甲酰基-5-((3-羰基吗啉代)甲基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000033
第一步
二叔丁基(5-溴-6-甲基吡啶-2-基)酰亚胺基二碳酸酯
将化合物5-溴-6-甲基吡啶-2-胺10a(50g,0.27mol)、2-碳酸-2-叔丁酯(145.16g,0.67mol),N,N-二甲基吡啶-4-胺(3.20g,27.00mmol)和四氢呋喃(300mL)混合,室温下搅拌16小时。此混合物用300mL水淬灭,用乙酸乙酯(200mL×2)萃取,有机相用饱和食盐水(200mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用石油醚洗,得到目标产物二叔丁基(5-溴-6-甲基吡啶-2-基)酰亚胺基二碳酸酯10b(80.00g,白色固体),产率:77%。
1H NMR(400MHz,CDCl3)δ7.80(d,J=8.4,1H),7.00(d,J=8.4 Hz,1H),2.61(s,3H),1.46(s,18H)。
第二步
二叔丁基(5-溴-6-(二溴甲基)吡啶-2-基)酰亚胺基二碳酸酯
将化合二叔丁基(5-溴-6-甲基吡啶-2-基)酰亚胺基二碳酸酯10b(80g,0.21mol),1-溴吡咯烷-2,5-二酮(110.00g,0.63mol),苯甲过氧酸酐(0.24g,0.06mol)和四氯化碳(600mL)混合,90℃搅拌16小时。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94∶6),得到目标产物二叔丁基(5-溴-6-(二溴甲基)吡啶-2-基)酰亚胺基二碳酸酯10c(90.00g,黄色固体),产率:80%。
1H NMR(400MHz,CDCl3)δ7.82(d,J=8.0Hz,1H),7.26(d,J=8.0Hz,1H),7.08(s,1H),1.50(s,18H)。
第三步
5-溴-6-(二甲氧基甲基)吡啶-2-胺
将化合物二叔丁基(5-溴-6-(二溴甲基)吡啶-2-基)酰亚胺基二碳酸酯10c(90.00g,0.17mol),氢氧化钾(38.52g,0.66mol)和甲醇(300mL)混合,70℃搅拌72小时。冷却至室温,减压脱溶,残余物用500mL水溶解,用乙酸乙酯(500mL×3)萃取,有机相用饱和食盐水(500mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯100∶0-12∶1),得到目标产物5-溴-6-(二甲氧基甲基)吡啶-2-胺10d(22g,黄色固体),产率:54%。
MS m/z(ESI):247&249[M+1]
1H NMR(400MHz,CDCl3)δ7.54(d,J=8.4Hz,1H),6.38(d,J=8.4Hz,1H),5.61(s,1H),4.63(brs,2H),3.48(s,6H)。
第四步
5-溴-6-(二甲氧基甲基)-N-甲基吡啶-2-胺
将化合物5-溴-6-(二甲氧基甲基)吡啶-2-胺10d(22.00g,89.43mmol),甲醇钠(24.15g,447mmol),多聚甲醛(10.74g,358mmol)和甲醇(300mL)混合,80℃搅拌16小时。冷却,加入硼氢化钠(13.59g,358mmol),80℃搅拌1小时。此混合物用300mL水淬灭,用乙酸乙酯(500mL×3)萃取,有机相用饱和食盐水(500mL×3)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯100∶0-96∶4),得到目标产物5-溴-6-(二 甲氧基甲基)-N-甲基吡啶-2-胺10e(8.20g,黄色固体),产率:34%。
MS m/z(ESI):261&263[M+1]
1H NMR(400MHz,CDCl3)δ7.58(d,J=8.8Hz,1H),6.27(d,J=8.8Hz,1H),5.59(s,1H),4.87(brs,1H),3.48(s,6H),2.87(d,J=5.2Hz,3H)。
第五步
甲基-2-(二甲氧基甲基)-6-(甲基氨基)尼古丁酸酯
将化合物5-溴-6-(二甲氧基甲基)-N-甲基吡啶-2-胺10e(8.00g,30.77mmol),醋酸钯(0.69g,3.08mmol),1,1-双(二苯基膦)二茂铁(3.41g,6.16mmol),三乙胺(6.22g,61.54mmol),甲醇(30mL)和N,N-二甲基甲酰胺(400mL)混合,在一氧化碳气氛下100℃搅拌16小时。此混合物用700mL水淬灭,用乙酸乙酯(500mL×3)萃取,有机相用饱和食盐水(500mL×3)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯94∶6),得到目标产物甲基-2-(二甲氧基甲基)-6-(甲基氨基)尼古丁酸酯10f(2.30g,黄色固体),产率:30%。
MS m/z(ESI):241[M+1]
1H NMR(400MHz,CDCl3)δ8.05(d,J=8.8Hz,1H),6.32(d,J=8.8Hz,1H),6.24(s,1H),5.30(brs,1H),3.86(s,3H),3.51(s,6H),2.95(d,J=5.2Hz,3H)。
第六步
甲基-6-((叔丁氧基羰基)(甲基)氨基)-2-(二甲氧基甲基)尼古丁酸酯
将化合物甲基-2-(二甲氧基甲基)-6-(甲基氨基)尼古丁酸酯10f(2.00g,8.33mmol),2-碳酸-2-叔丁酯(3.60g,16.67mol),N,N-二甲基吡啶-4-胺(0.10g,0.83mmol)和四氢呋喃(50mL)混合,室温搅拌2小时。此混合物用100mL水淬灭,用乙酸乙酯(100mL×3)萃取,有机相用饱和食盐水(100mL×3)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯94∶6),得到目标产物甲基-6-((叔丁氧基羰基)(甲基)氨基)-2-(二甲氧基甲基)尼古丁酸酯10g(2.20g,黄色油),产率:78%。
MS m/z(ESI):341[M+1]
1H NMR(400MHz,CDCl3)δ8.07(d,J=8.8Hz,1H),7.84(d,J= 8.8Hz,1H),6.08(s,1H),3.91(s,3H),3.52(s,6H),3.49(s,3H),1.53(s,9H)。
第七步
叔丁基(6-(二甲氧基甲基)-5-(羟甲基)吡啶-2-基)(甲基)氨基羧酸酯
将化合物甲基-6-((叔丁氧基羰基)(甲基)氨基)-2-(二甲氧基甲基)尼古丁酸酯10g(2.20g,6.47mmol),硼氢化钠(2.46g,60.47mmol),无水氯化钙(1.42g,12.90mmol)和甲醇(20mL)混合,65℃搅拌2小时。此混合物用100mL水淬灭,用乙酸乙酯(100mL×2)萃取,有机相用饱和食盐水(100mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,得到目标产物叔丁基-(6-(二甲氧基甲基)-5-(羟甲基)吡啶-2-基)(甲基)氨基羧酸酯10h(1.70g,白色固体),产率:84%。
MS m/z(ESI):313[M+1]。
第八步
叔丁基(5-(溴甲基)-6-(二甲氧基甲基吡啶-2-基)(甲基)氨基羧酸酯
将化合物叔丁基(6-(二甲氧基甲基)-5-(羟甲基)吡啶-2-基)(甲基)氨基羧酸酯10g(1.70g,5.45mmol),三溴化磷(1.75g,6.54mmol)和二氯甲烷(50mL)混合,0℃搅拌0.5小时。此混合物用10mL碳酸氢钠水溶液淬灭,用乙酸乙酯(100mL×2)萃取,有机相用饱和食盐水(100mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯100∶0-94∶6),得到目标产物叔丁基-(5-(溴甲基)-6-(二甲氧基甲基吡啶-2-基)(甲基)氨基羧酸酯10i(0.80g,无色固体),产率:40%。
MS m/z(ESI):375&377[M+1]。
第九步
叔丁基-(6-(2-甲氧基乙基)-5-((3-羰基吗啉)甲基)吡啶-2-基)(甲基)氨基羧酸酯
将化合物叔丁基-(5-(溴甲基)-6-(二甲氧基甲基吡啶-2-基)(甲基)氨基羧酸酯10i(0.28g,0.76mmol),吗啉-3-酮(0.15g,1.52mmol),氢化钠(76mg,1.88mmol,60%矿物油混合物)和N,N-二甲基甲酰胺(10mL)混合,室温搅拌1小时。此混合物用水淬灭,用乙酸乙酯(50mL×3)萃取,有机相用饱和食盐水(50mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用硅胶柱层析纯化(石油醚/ 乙酸乙酯=100∶0-7∶3),得到目标产物叔丁基-(6-(2-甲氧基乙基)-5-((3-羰基吗啉)甲基)吡啶-2-基)(甲基)氨基羧酸酯10j(0.27g,白色固体),产率:91%。
MS m/z(ESI):396[M+1]。
第十步
4-((2-(二甲氧基甲基)-6-(甲基氨基)吡啶-3-基)甲基)吗啉-3-酮
将化合物叔丁基-(6-(2-甲氧基乙基)-5-((3-羰基吗啉)甲基)吡啶-2-基)(甲基)氨基羧酸酯10j(0.27g,0.18mmol),三氟乙酸(1mL)和二氯甲烷(4mL)混合,室温搅拌6小时。减压脱溶,得到目标产物4-((2-(二甲氧基甲基)-6-(甲基氨基)吡啶-3-基)甲基)吗啉-3-酮三氟乙酸盐10k(0.27g,黄色固体)。
MS m/z(ESI):296[M+1]。
第十一步
苯基-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)(甲基)氨基羧酸酯
将化合物4-((2-(二甲氧基甲基)-6-(甲基氨基)吡啶-3-基)甲基)吗啉-3-酮三氟乙酸盐10k(0.25g,0.60mmol),碳酸二苯酯(0.26g,1.20mmol),六甲基二硅基胺基锂(1.8mL,1.80mmol,1M四氢呋喃溶液)和四氢呋喃(8mL)混合,0℃搅拌0.5小时。此混合物用10mL饱和氯化铵溶液淬灭,用乙酸乙酯(50mL×2)萃取,有机相用饱和食盐水(50mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯2∶1),得到目标产物苯基-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)(甲基)氨基羧酸酯10l(0.13g,无色油),产率:51%。
MS m/z(ESI):416[M+1]。
第十二步
3-(4-氯-5-氰基吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲
将化合物苯基(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)(甲基)氨基羧酸酯10l(45mg,0.11mmol),6-氨基-4-氯尼古丁腈(33mg,0.22mmol),六甲基二硅基胺基锂(0.3mL,0.33mmol,1M四氢呋喃溶液)和四氢呋喃(3mL)混合,室温搅拌1小时。此混合物用10mL 饱和氯化铵溶液淬灭,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯1∶1),得到目标产物3-(4-氯-5-氰基吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲10m(38mg,白色固体),产率:74%。
MS m/z(ESI):475&477[M+1]。
第十三步
3-(5-氰基-4-(异丙基氨基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉代)甲基)吡啶-2-基)-1-甲基脲
将化合物3-(4-氯-5-氰基吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲10m(10mg,0.02mmol),异丙胺(5mg,0.08mmol),二异丙基乙胺(6mg,0.04mmol)和N,N-二甲基乙酰胺(0.4mL)混合,50℃搅拌16小时。此混合物用10mL水稀释,用二氯甲烷(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯1∶1),得到目标产物3-(5-氰基-4-(异丙基氨基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉代)甲基)吡啶-2-基)-1-甲基脲10n(5mg,白色固体),产率:48%。
MS m/z(ESI):498[M+1]。
第十四步
3-(5-氰基-4-(异丙基氨基)吡啶-2-基)-1-(6-甲酰基-5-((3-羰基吗啉代)甲基)吡啶-2-基)-1-甲基脲
将化合物3-(5-氰基-4-(异丙基氨基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉代)甲基)吡啶-2-基)-1-甲基脲10n(5mg,0.01mmol),盐酸(0.8mL,37%),水(1mL)和四氢呋喃(2mL)混合,室温搅拌1小时。此混合物用饱和的碳酸钠溶液淬灭,用二氯甲烷(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯1∶1),得到目标产物3-(5-氰基-4-(异丙基氨基)吡啶-2-基)-1-(6-甲酰基-5-((3-羰基吗啉代)甲基)吡啶-2-基)-1-甲基脲10(3mg,白色固体),产率:66%。
MS m/z(ESI):452[M+1]
1H NMR(400MHz,CDCl3)δ12.95(s,1H),10.26(s,1H),8.15(s,1H),7.98(d,J=8.8Hz,1H),7.56(s,1H),7.30(d,J=8.8Hz,1H),5.13(s,2H),4.78-4.76(m,1H),4.26(s,2H),3.89(t,J=4.4Hz,2H),3.88(brs,1H),3.53(s,3H),3.43(t,J=4.4Hz,2H),1.31(d,J=4.8Hz,6H)。
实施例11
1-(4-氯-5-氰基吡啶-2-基)-3-(6-甲酰基-5-((3-羰基吗啉)甲基)吡啶-2-基)脲
Figure PCTCN2017088038-appb-000034
参照实施例9的操作步骤合成实施例11,但在第十三步中用1-(4-氯-5-氰基吡啶-2-基)-3-(6-甲酰基-5-((3-羰基吗啉)甲基)吡啶-2-基)脲取代3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉代)甲基)吡啶-2-基)-1-甲基脲。
MS m/z(ESI):429&431[M+1]
1H NMR(400MHz,CDCl3)δ13.60(s,1H),10.26(s,1H),8.51(s,1H),8.46(s,1H),8.01(d,J=8.8Hz,1H),7.28(d,J=8.8Hz,1H),5.13(s,2H),4.26(s,2H),3.91(t,J=3.2Hz,2H),3.55(s,3H),3.44(t,J=3.2Hz,2H)。
实施例12
1-(5-氰基-4-(2-甲氧基乙氧基)吡啶-2-基)-3-(6-甲酰基-5-((3-羰基吗啉代)甲基)吡啶-2-基)脲
Figure PCTCN2017088038-appb-000035
Figure PCTCN2017088038-appb-000036
第一步
6-氨基-4-(2-甲氧基乙氧基)尼古丁腈
将化合物6-氨基-4-氯尼古丁腈12a(60mg,0.39mmol)、2-甲氧基乙醇(60mg,0.78mmol),氢化钠(34mg,0.86mmol,60%矿物油混合物)和N-甲基吡咯烷酮(1.5mL)混合,70℃下搅拌16小时。冷却至室温,此混合物用20mL水淬灭,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用制备硅胶板纯化(石油醚/乙酸乙酯=1∶1),得到目标产物6-氨基-4-(2-甲氧基乙氧基)尼古丁腈12b(30mg,白色固体),产率:40%。
MS m/z(ESI):194[M+1]。
第二步
3-(5-氰基-4-(2-甲氧基乙氧基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲
参照实施例9第十一步的操作步骤合成实施例12c,用6-氨基-4-(2-甲氧基乙氧基)尼古丁腈取代6-氨基-4-氯尼古丁腈。得到目标产物3-(5-氰基-4-(2-甲氧基乙氧基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉代)甲基)吡啶-2-基)-1-甲基脲12c(8mg,白色固体),产率:70%。
MS m/z(ESI):515[M+1]。
第三步
3-(5-氰基-4-(2-甲氧基乙氧基)吡啶-2-基)-1-(6-甲酰基-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲
参照实施例9第十三步的操作步骤合成实施例12。用3-(5-氰基-4-(2-甲氧基乙氧基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲取代3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((3-羰基吗啉)甲基)吡啶-2-基)-1-甲基脲。得到目标产物3-(5-氰基-4-异丙氧基吡啶-2-基)-1-(6-甲酰基吡啶-2-基)-1-甲基脲12(6mg,白色固体)。产率:82%。
MS m/z(ESI):469[M+1]
1H NMR(400MHz,CDCl3)δ13.34(s,1H),10.27(s,1H),8.36(s,1H),8.00(d,J=8.8Hz,1H),7.96(s,1H),7.18(d,J=7.6Hz,1H),5.13(s,2H),4.35(s,2H),4.26(d,J=3.2Hz,2H),3.90(t,J=3.2Hz,2H),3.83(t,J=4.0Hz,2H),3.54(s,3H),3.38(s,3H),3.43(t,J=4.0Hz,2H)。
实施例13
1-(5-氰基-4-异丙氧基吡啶-2-基)-3-(6-甲酰基-5-((3-羰基吗啉)甲基)吡啶-2-基)脲
Figure PCTCN2017088038-appb-000037
参照实施例12的操作步骤合成实施例13。但在第一步中用异丙醇取代2-甲氧基乙醇。
MS m/z(ESI):453[M+1]
1H NMR(400MHz,CDCl3)δ13.26(s,1H),10.27(s,1H),8.34(s,1H),8.00(d,J=8.8Hz,1H),7.93(s,1H),7.32(d,J=8.8Hz,1H),5.13(s,2H),4.87-4.85(m,1H),4.26(s,2H),3.54(s,3H),3.47(t,J=4.4Hz,2H),3.43(t,J=4.4Hz,2H),1.45(d,J=3.2Hz,6H)。
实施例14
(R)-3-(5-氰基-4-((1-甲氧基丙烷-2-基)氧代)吡啶-2-基)-1-(6-甲酰基-5-((3-羰基吗啉代)甲基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000038
参照实施例12的操作步骤合成实施例14。但在第一步中用(R)-1-甲氧基丙烷-2-醇取代2-甲氧基乙醇。
MS m/z(ESI):483[M+1]
1H NMR(400MHz,CDCl3)δ13.27(s,1H),10.26(s,1H),8.34(s,1H),7.99(s,1H),7.98(d,J=8.8Hz,1H),7.32(d,J=8.8Hz,1H),5.13(s,2H),4.87-4.86(m,1H),4.26(s,2H),3.90(t,J=4.4Hz,2H),3.64-3.60(m,1H),3.56-3.55(m,1H),3.54(s,3H),3.49(t,J=4.4Hz,2H),3.43(s,3H),1.43(d,J=3.2Hz,3H)。
实施例15
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲盐酸盐
Figure PCTCN2017088038-appb-000039
第一步
叔丁基-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2基)(甲基)氨基羧酸酯
将化合物叔丁基-(5-(溴甲基)-6-(二甲氧基甲基吡啶-2-基)(甲基)氨基羧酸酯15a(70mg,0.19mmol),4-甲基哌嗪-2-酮(43mg,0.38mmol),氢化钠(19mg,0.47mmol,60%矿物油混合物)和N,N-二甲基甲酰胺(3mL)混合,室温搅拌1小时。此混合物用水淬灭,用二氯甲烷(20mL×3)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯1.5∶1),得到目标产物叔丁基-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2基)(甲基)氨基羧酸酯15b(60mg,无色固体),产率:83%。
MS m/z(ESI):409[M+1]。
第二步
1-((2-(二甲氧基甲基)-6-(甲基氨基)吡啶-3-基)甲基)-4-甲基哌嗪-2-酮
将化合物叔丁基-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2基)(甲基)氨基羧酸酯15b(60mg,0.15mmol),三氟乙酸(1mL)和二氯甲烷(4mL)混合,室温搅拌6小时。减压脱溶,得到目标产物1-((2-(二甲氧基甲基)-6-(甲基氨基)吡啶-3-基)甲基)-4-甲基哌嗪-2-酮三氟乙酸盐15c(60mg,浅黄色固体),粗品。
MS m/z(ESI):309[M+1]。
第三步
苯基-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2基)(甲基)氨基羧酸酯
将化合物1-((2-(二甲氧基甲基)-6-(甲基氨基)吡啶-3-基)甲基)-4-甲基哌嗪-2-酮三氟乙酸盐15c(60mg,0.14mmol),碳酸二苯酯(60mg,0.28mmol),六甲基二硅基胺基锂(0.56mL,0.56mmol,1M四氢呋喃溶液)和四氢呋喃(5mL)混合,0℃搅拌0.5小时。此混合物用10mL饱和氯化铵溶液淬灭,用二氯甲烷(20mL×3)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(二氯甲烷/甲醇30∶1),得到目标产物苯基-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2基)(甲基) 氨基羧酸酯15d(30mg,无色固体),产率:36%。
MS m/z(ESI):429[M+1]。
第四步
3-(4-氯-5-氰基吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲
将化合物苯基-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2基)(甲基)氨基羧酸酯15d(30mg,0.07mmol),6-氨基-4-氯尼古丁腈(21mg,0.14mmol),六甲基二硅基胺基锂(0.21mL,0.21mmol,1M四氢呋喃溶液)和四氢呋喃(3mL)混合,室温搅拌1小时。此混合物用10mL饱和氯化铵溶液淬灭,用二氯甲烷(20mL×3)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(二氯甲烷/甲醇30∶1),得到目标产物3-(4-氯-5-氰基吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲15e(17mg,白色固体)。产率:50%。
MS m/z(ESI):488&490[M+1]
1H NMR(400MHz,CDCl3)δ13.82(s,1H),8.52(s,1H),8.46(s,1H),7.77(d,J=8.4Hz,1H),7.04(d,J=8.4Hz,1H),5.47(s,1H),4.86(s,2H),3.47(s,6H),3.46(s,3H),3.27(s,2H),3.20(t,J=4.4Hz,2H),2.62(t,J=4.4Hz,2H),2.35(s,3H)。
第五步
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲
将化合物3-(4-氯-5-氰基吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲15e(4mg,0.008mmol),2-甲氧基乙胺(2mg,0.024mmol),二异丙基乙胺(2mg,0.016mmol)和N,N-二甲基乙酰胺(0.4mL)混合,50℃搅拌16小时。此混合物用10mL水稀释,用二氯甲烷(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(二氯甲烷/甲醇25∶1),得到目标产物3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲15f(2mg,白色固体),产率: 46%。
MS m/z(ESI):527[M+1]。
第六步
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲盐酸盐
将化合物3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲15f(2mg,0.004mmol),盐酸(0.8mL,37%),水(1mL)和四氢呋喃(2mL)混合,室温搅拌1小时。减压去溶,得到目标产物3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲盐酸盐15(1.5mg,白色固体)。产率:67%。
MS m/z(ESI):481[M+1]
1H NMR(400MHz,CD3OD)δ8.40(s,1H),7.98(d,J=8.4Hz,1H),7.40(d,J=8.4Hz,1H),6.94(s,1H),6.92(s,1H),4.91(s,2H),3.75(t,J=6.8Hz,2H),3.69-3.60(m,2H),3.45-3.42(m,4H),3.37(s,3H),3.36(s,3H),3.26-3.21(m,2H),3.03(s,3H)。
实施例16
3-(5-氰基-4-(异丙基氨基)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000040
参照实施例15的操作步骤合成实施例16,但在第五步中用异丙胺取代2-甲氧基乙胺。
MS m/z(ESI):465[M+1]
1H NMR(400MHz,CDCl3)δ12.95(s,1H),10.25(s,1H),8.16(s,1H),7.95(d,J=8.0Hz,1H),7.56(s,1H),7.28(d,J=8.0Hz,1H),5.10(s,2H),4.76(brs,1H),3.89-3.87(m,1H),3.52(s,3H),3.36(s,2H),3.20(t,J=4.0Hz,2H),2.66(t,J=4.0Hz,2H),2.35(s,3H),1.32(d,J=5.2Hz,6H)。
实施例17
3-(4-氯-5-氰基吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000041
参照实施例15的操作步骤合成实施例17,但在第六步中用3-(4-氯-5-氰基吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲取代3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲。
MS m/z(ESI):442&444[M+1]
1H NMR(400MHz,CDCl3)δ13.59(s,1H),10.26(s,1H),8.51(s,1H),8.46(s,1H),7.95(d,J=8.4Hz,1H),7.31(d,J=8.4Hz,1H),5.10(s,2H),3.54(s,3H),3.38(s,2H),3.20(t,J=4.4Hz,2H),2.68(t,J=4.4Hz,2H),2.36(s,3H)。
实施例18
(R)-3-(5-氰基-4-((1-甲氧基丙烷-2-基)氧代)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000042
Figure PCTCN2017088038-appb-000043
第一步
(R)-6-氨基-4-((1-甲氧基丙烷-2-基)氧代)尼古丁腈
将化合物6-氨基-4-氯尼古丁腈18a(60mg,0.39mmol),(R)-1-甲氧基丙烷-2-醇(70mg,0.78mmol),氢化钠(34mg,0.86mmol,60%矿物油混合物)和N-甲基吡咯烷酮(1.5mL)混合,70℃下搅拌16小时。冷却至室温,此混合物用20mL水淬灭,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用制备硅胶板纯化(石油醚/乙酸乙酯1∶1),得到目标产物(R)-6-氨基-4-((1-甲氧基丙烷-2-基)氧代)尼古丁腈18b(17mg,白色固体),产率:21%。
MS m/z(ESI):208[M+1]
1H NMR(400MHz,CDCl3)δ8.17(s,1H),6.00(s,1H),4.94(brs,2H),4.63-4.60(m,1H),3.63-3.62(m,1H),3.54-3.51(m,1H),3.41(s,3H),1.38-1.36(m,3H)。
第二步
(R)-3-(5-氰基-4-((1-甲氧基丙烷-2-基)氧代)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲
将化合物苯基-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2基)(甲基)氨基羧酸酯(10mg,0.02mmol),(R)-6-氨基-4-((1-甲氧基丙烷-2-基)氧代)尼古丁腈18b(8mg,0.04mmol),六甲基二硅 基胺基锂(0.06mL,0.06mmol,1M四氢呋喃溶液)和四氢呋喃(2mL)混合,室温搅拌1小时。此混合物用10mL饱和氯化铵溶液淬灭,用二氯甲烷(20mL×3)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(二氯甲烷/甲醇30∶1),得到目标产物(R)-3-(5-氰基-4-((1-甲氧基丙烷-2-基)氧代)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲18c(6mg,白色固体)。产率:47%。
MS m/z(ESI):542[M+1]。
第三步
(R)-3-(5-氰基-4-((1-甲氧基丙烷-2-基)氧代)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲
将化合物(R)-3-(5-氰基-4-((1-甲氧基丙烷-2-基)氧代)吡啶-2-基)-1-(6-(二甲氧基甲基)-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲18c(6mg,0.01mmol),盐酸(0.8mL,37%),水(1mL)和四氢呋喃(2mL)混合,室温搅拌1小时。此混合物用饱和的碳酸钠溶液淬灭,用二氯甲烷(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(二氯甲烷/甲醇25∶1),得到目标产物(R)-3-(5-氰基-4-((1-甲氧基丙烷-2-基)氧代)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲18(4mg,白色固体),产率:73%。
MS m/z(ESI):496[M+1]
1H NMR(400MHz,CDCl3)δ13.28(s,1H),10.27(s,1H),8.35(s,1H),7.99(s,1H),7.94(d,J=7.6Hz,1H),7.27(d,J=7.6Hz,1H),5.11(s,2H),4.88-4.86(m,1H),3.66-3.59(m,2H),3.57(s,3H),3.49(s,3H),3.43(s,2H),3.20(t,J=4.4Hz,2H),2.67(t,J=4.4Hz,2H),2.36(s,3H),1.42-1.40(m,3H)。
实施例19
3-(5-氰基-4-异丙氧基吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000044
参照实施例18的操作步骤合成实施例19,但在第一步中用异丙醇取代(R)-1-甲氧基丙烷-2-醇。
MS m/z(ESI):466[M+1]
1H NMR(400MHz,CDCl3)δ13.28(s,1H),10.26(s,1H),8.34(s,1H),7.96(d,J=8.4Hz,1H),7.93(s,1H),7.26(d,J=8.4Hz,1H),5.10(s,2H),4.85-4.83(m,1H),3.49(s,3H),3.38(s,2H),3.21(t,J=4.4Hz,2H),2.69(t,J=4.4Hz,2H),2.36(s,3H),1.45(d,J=4.0Hz,6H)。
实施例20
3-(5-氰基-4-(2-甲氧基乙氧基)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000045
参照实施例18的操作步骤合成实施例20,但在第一步中用2-甲氧基乙醇取代(R)-1-甲氧基丙烷-2-醇。
MS m/z(ESI):482[M+1]
1H NMR(400MHz,CDCl3)δ13.34(s,1H),10.27(s,1H),8.36(s,1H),7.96(s,1H),7.95(d,J=8.4Hz,1H),7.31(d,J=8.4Hz,1H),5.11(s,2H),4.35(t,J=4.0Hz,2H),3.83(t,J=4.0Hz,2H),3.53(s,3H),3.48(s,3H),3.37(s,2H),3.20(t,J=4.4Hz,2H),2.67(t,J=4.4Hz,2H),2.36(s,3H)。
实施例21
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基-5-(羟甲基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000046
第一步
5-(((叔丁基二甲基甲硅烷基)氧代)甲基)-6-(二甲氧基甲基)-N-甲基吡啶-2-胺
将化合物(2-(二甲氧基甲基)-6-(甲基氨基)吡啶-3-基)甲醇21a(60mg,0.28mmol)、叔-丁基氯二甲基硅烷(64mg,0.42mmol),N-乙基-N-异丙基丙烷-2-胺(72mg,0.56mmol),N,N-二甲基吡啶-4-胺(7mg,0.06mmol)和二氯甲烷(4mL)混合,室温下搅拌6小时。此混合物用20mL水淬灭,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用制备硅胶板纯化(石油醚/乙酸乙酯=15∶1),得到目标产物5-(((叔-丁基二甲基甲硅烷基)氧代)甲基)-6-(二甲氧基甲基)-N-甲基吡啶-2-胺21b(60mg,无色油),产率:65%。
MS m/z(ESI):327[M+1]
1H NMR(400MHz,CDCl3)δ7.57(d,J=8.0Hz,1H),6.28(d,J=8.0Hz,1H),5.21(s,1H),4.69(s,2H),4.57(brs,1H),3.32(s,6H),2.80(s,3H),0.84(s,9H),0.08(s,6H)。
第二步
苯基-(5-(((叔丁基二甲基硅)氧)甲基)-6-(二甲氧基甲基)吡啶-2-基)(甲基)氨基羧酸酯
将化合物5-(((叔丁基二甲基甲硅烷基)氧代)甲基)-6-(二甲氧基甲基)-N-甲基吡啶-2-胺21b(22mg,0.07mmol),碳酸二苯酯(15mg,0.14mmol),六甲基二硅基胺基锂(0.21mL,0.21mmol,1M四氢呋喃溶液)和四氢呋喃(3mL)混合,0℃搅拌0.5小时。此混合物用10mL饱和氯化铵溶液淬灭,用乙酸乙酯(20mL×3)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯5∶1)。得到目标产物苯基-(5-(((叔丁基二甲基硅)氧)甲基)-6-(二甲氧基甲基)吡啶-2-基)(甲基)氨基羧酸酯21c(18mg,白色固体),产率:60%。
MS m/z(ESI):447[M+1]
1H NMR(400MHz,CDCl3)δ7.86(d,J=8.4Hz,1H),7.58(d,J=7.2Hz,1H),7.27-7.25(m,2H),7.15-7.04(m,3H),5.19(s,1H),4.83(s,2H),3.49(s,3H),3.34(s,6H),0.85(s,9H),0.08(s,6H)。
第三步
1-(5-(((叔丁基二甲基甲硅烷基)氧代)甲基)-6-(二甲氧基甲基)吡啶-2-基)-3-(4-氯-5-氰基吡啶-2-基)-1-甲基脲
将化合物苯基-(5-(((叔丁基二甲基硅)氧)甲基)-6-(二甲氧基甲基)吡啶-2-基)(甲基)氨基羧酸酯21c(18mg,0.04mmol),6-氨基-4-氯尼古丁腈(12mg,0.08mmol),六甲基二硅基胺基锂(0.12mL,0.12mmol,1M四氢呋喃溶液)和四氢呋喃(2mL)混合,室温搅拌1小时。此混合物用10mL饱和氯化铵溶液淬灭,用乙酸乙酯(20mL×3)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯3∶1),得到目标产物1-(5-(((叔-丁基二甲基甲硅烷基)氧代)甲基)-6-(二甲氧基甲基)吡啶-2-基)-3-(4-氯-5-氰基吡啶-2-基)-1-甲基脲21d(16mg,白色固体),产率:79%。
MS m/z(ESI):506&508[M+1]
1H NMR(400MHz,CDCl3)δ13.78(s,1H),8.40(s,1H),8.34(s,1H),7.95(d,J=8.8Hz,1H),6.97(d,J=8.8Hz,1H),5.35(s,1H),4.78(s,2H),3.37(s,3H),3.33(s,6H),0.83(s,9H),0.02(s,6H)。
第四步
1-(5-(((叔丁基二甲基甲硅烷基)氧代)甲基)-6-(二甲氧基甲基)吡啶-2-基)-3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-甲基脲
将化合物1-(5-(((叔-丁基二甲基甲硅烷基)氧代)甲基)-6-(二甲氧基甲基)吡啶-2-基)-3-(4-氯-5-氰基吡啶-2-基)-1-甲基脲21d(10mg,0.02mmol),2-甲氧基乙胺(3mg,0.06mmol),二异丙基乙胺(5mg,0.06mmol)和N,N-二甲基乙酰胺(0.4mL)混合,50℃搅拌16小时。此混合物用10mL水稀释,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯3∶1)。得到目标产物1-(5-(((叔丁基二甲基甲硅烷基)氧代)甲基)-6-(二甲氧基甲基)吡啶-2-基)-3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-甲基脲21e(9mg,白色固体),产率:76%。
MS m/z(ESI):545[M+1]。
第五步
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基-5-(羟甲基)吡啶-2-基)-1-甲基脲
将化合物1-(5-(((叔丁基二甲基甲硅烷基)氧代)甲基)-6-(二甲氧基甲基)吡啶-2-基)-3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-甲基脲21e(9mg,0.02mmol),盐酸(0.8mL,37%),水(1mL)和四氢呋喃(2mL)混合,室温搅拌1小时。此混合物用饱和的碳酸钠溶液淬灭,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯1∶1)。得到目标产物1-(5-(((叔丁基二甲基甲硅烷基)氧代)甲基)-6-(二甲氧基甲基)吡啶-2-基)-3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-甲基脲21(3mg,白色固体),产率:47%。
MS m/z(ESI):385[M+1]
1H NMR(400MHz,CDCl3)δmajor(21’):12.60(s,1H),8.08(s,1H),7.70(d,J=7.6Hz,1H),7.58(s,1H),7.07(d,J=7.6Hz,1H),6.53(s,1H),5.31(s,1H),5.25-5.23(m,1H),5.05-5.02(m,1H),3.63-3.61(m,2H),3.51-3.50(m,2H),3.49(s,3H),3.41(s,3H).Minor(21):12.94(s,1H),10.25(s,1H),8.17(s,1H),8.06(d,J=8.4Hz,1H),7.57(s,1H), 7.27(d,J=8.4Hz,1H),6.53(s,1H),5.31(s,1H),5.25-5.23(m,1H),5.05-5.02(m,1H),3.63-3.61(m,2H),3.51-3.50(m,2H),3.49(s,3H),3.41(s,3H)。
实施例22
3-(4-氯-5-氰基吡啶-2-基)-1-(6-甲酰基-5-(羟甲基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000047
参照实施例21的操作步骤合成实施例22,但在第五步中用1-(5-(((叔丁基二甲基甲硅烷基)氧代)甲基)-6-(二甲氧基甲基)吡啶-2-基)-3-(4-氯-5-氰基吡啶-2-基)-1-甲基脲取代3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基-5-(羟甲基)吡啶-2-基)-1-甲基脲。
MS m/z(ESI):346&348[M+1]
1H NMR(400MHz,CDCl3)δmajor(22’):13.10(s,1H),8.46(s,1H),8.45(s,1H),7.77(d,J=8.4Hz,1H),7.13(d,J=8.4Hz,1H),6.51(s,1H),5.29(s,1H),5.11-5.08(m,2H),3.53(s,3H).Minor(22):13.55(s,1H),10.25(s,1H),8.48(s,1H),8.47(s,1H),8.10(d,J=8.4Hz,1H),7.35(d,J=8.4Hz,1H),4.97(s,2H),4.08-4.06(m,1H),3.57(s,3H)。
实施例23
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基-5-(哌啶-4-基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000048
Figure PCTCN2017088038-appb-000049
第一步
叔丁基-6-((叔丁氧基羰基)(甲基)氨基)-2-(二甲氧基甲基)-5′,6′-二氢-[3,4′-联吡啶]-1′(2′H)-羧酸酯
将化合物叔丁基(5-溴-6-(二甲氧基甲基吡啶-2-基)(甲基)氨基羧酸酯23a(0.20g,0.56mmol),[1,1′-双(二苯基膦基)二茂铁]二氯化钯(90mg,0.11mmol),叔丁基-4-(4,4,5,5-四甲基-1,3,2-二噁硼戊环-2-基)-5,6-二氢吡啶-1-(2H)-羧酸酯(0.34g,1.11mmol),碳酸钾(0.23g,1.67mmol),水(1mL)和二氧六环(5mL)混合,氮气下,90℃搅拌16小时。此混合物用水稀释,用乙酸乙酯(50mL×3)萃取,有机相用饱和食盐水(50mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用硅胶层析纯化(石油醚/乙酸乙酯84∶16),得到目标叔丁基-6-((叔丁氧基羰基)(甲基)氨基)-2-(二甲氧基甲基)-5′,6′-二氢-[3,4′-联吡啶]-1′(2′H)-羧酸酯23b(0.13g,无色油),产率:51%。
MS m/z(ESI):464[M+1]
1H NMR(400MHz,CDCl3)δ7.64(d,J=8.4Hz,1H),7.41(d,J=8.4Hz,1H),5.67-5.65(m,1H),5.54(s,1H),4.07(s,2H),3.65(t,J=5.2Hz,2H),3.46(s,6H),3.45(s,3H),2.40-2.38(m,2H),1.53(s,9H),1.52(s,9H)。
第二步
叔丁基-4-(6-((叔丁氧基羰基)(甲基)氨基)-2-(二甲氧基甲基)吡啶-3-基)哌啶-1-羧酸酯
将化合物叔丁基-6-((叔-丁氧基羰基)(甲基)氨基)-2-(二甲氧基甲基)-5′,6′-二氢-[3,4′-联吡啶]-1′(2′H)-羧酸酯23b(0.13g,0.28mmol),钯碳(26mg,20%),甲醇(3mL)和乙酸乙酯(2mL)混合,室温搅拌16小时。过滤减压脱溶,得到目标产物叔丁基-4-(6-((叔丁氧基羰基)(甲基)氨基)-2-(二甲氧基甲基)吡啶-3-基)哌啶-1-羧酸酯23c(0.13g,无色油),粗品。
MS m/z(ESI):466[M+1]。
第三步
6-(二甲氧基甲基)-N-甲基-5-(哌啶-4-基)吡啶-2-胺
将化合物叔丁基-4-(6-((叔丁氧基羰基)(甲基)氨基)-2-(二甲氧基甲基)吡啶-3-基)哌啶-1-羧酸酯23c(0.13g,0.28mmol),三氟乙酸(1mL)和二氯甲烷(4mL)混合,室温搅拌6小时。减压脱溶,得到目标产物6-(二甲氧基甲基)-N-甲基-5-(哌啶-4-基)吡啶-2-胺三氟乙酸盐23d(95mg,浅黄色固体),粗品。
MS m/z(ESI):266[M+1]。
第四步
叔丁基-4-(2-(二甲氧基甲基)-6-(甲基氨基)吡啶-3-基)哌啶-1-羧酸酯
将化合物6-(二甲氧基甲基)-N-甲基-5-(哌啶-4-基)吡啶-2-胺三氟乙酸盐23d(95mg,0.25mmol),2-碳酸-2-叔丁酯(82mg,0.38mmol),三乙胺(76mg,0.75mmol)和二氯甲烷(4mL)混合,室温搅拌6小时。此混合物用20mL水淬灭,用乙酸乙酯(20mL×3)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯3∶1),得到目标产物叔丁基-4-(2-(二甲氧基甲基)-6-(甲基氨基)吡啶-3-基)哌啶-1-羧酸酯23e(80mg,白色固体),产率:65%。
MS m/z(ESI):366[M+1]。
第五步
叔丁基-4-(2-(二甲氧基甲基)-6-(甲基(苯氧基羰基)氨基)吡啶-3-基)哌啶-1-羧酸酯
将化合物叔丁基-4-(2-(二甲氧基甲基)-6-(甲基氨基)吡啶-3-基)哌啶-1-羧酸酯23e(40mg,0.11mmol),碳酸二苯酯(47mg,0.22mmol),六甲基二硅基胺基锂(0.33mL,0.33mmol,1M四氢呋喃溶液)和四氢呋喃(6mL)混合,0℃搅拌0.5小时。此混合物用10mL饱和氯化铵溶液淬灭,用乙酸乙酯(20mL×3)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯2∶1)。得到目标产物叔丁基-4-(2-(二甲氧基甲基)-6-(甲基(苯氧基羰基)氨基)吡啶-3-基)哌啶-1-羧酸酯23f(20mg,白色固体),产率:38%。
MS m/z(ESI):486[M+1]。
第六步
叔丁基-4-(6-(3-(4-氯-5-氰基吡啶-2-基)-1-甲基脲基)-2-(二甲氧基甲基)吡啶-3-基)哌啶-1-羧酸酯
将化合物叔丁基-4-(2-(二甲氧基甲基)-6-(甲基(苯氧基羰基)氨基)吡啶-3-基)哌啶-1-羧酸酯23f(20mg,0.04mmol),6-氨基-4-氯尼古丁腈(12mg,0.08mmol),六甲基二硅基胺基锂(0.12mL,0.12mmol,1M四氢呋喃溶液)和四氢呋喃(2mL)混合,室温搅拌1小时。此混合物用10mL饱和氯化铵溶液淬灭,用乙酸乙酯(20mL×3)萃取,有机相用饱和食盐水(20mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯1.5∶1),得到目标产物叔丁基-4-(6-(3-(4-氯-5-氰基吡啶-2-基)-1-甲基脲基)-2-(二甲氧基甲基)吡啶-3-基)哌啶-1-羧酸酯羧酸酯23g(10mg,白色固体),产率:45%。
MS m/z(ESI):545&547[M+1]。
第七步
叔丁基-4-(6-(3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-甲基脲基)-2-(二甲氧基甲基)吡啶-3-基)哌啶-1-羧酸酯
将化合物叔丁基-4-(6-(3-(4-氯-5-氰基吡啶-2-基)-1-甲基脲基)-2-(二甲氧基甲基)吡啶-3-基)哌啶-1-羧酸酯23g(10mg,0.02mmol),2-甲氧基乙胺(3mg,0.06mmol),二异丙基乙胺(5mg,0.06mmol)和N,N-二甲基乙酰胺(0.4mL)混合,50℃搅拌16小时。此混合物用10mL水稀释,用乙酸乙酯(20mL×2)萃取,有机相用饱和食盐水(20 mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,残余物用制备硅胶板纯化(石油醚/乙酸乙酯1.5∶1)。得到目标产物叔丁基-4-(6-(3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-甲基脲基)-2-(二甲氧基甲基)吡啶-3-基)哌啶-1-羧酸酯23h(6mg,白色固体),产率:56%。
MS m/z(ESI):584[M+1]。
第八步
3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基-5-(哌啶-4-基)吡啶-2-基)-1-甲基脲
将化合物叔丁基-4-(6-(3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-甲基脲基)-2-(二甲氧基甲基)吡啶-3-基)哌啶-1-羧酸酯23h(6mg,0.01mmol),盐酸(0.8mL,37%),水(1mL)和四氢呋喃(2mL)混合,室温搅拌1小时。减压去溶,得到目标产物3-(5-氰基-4-((2-甲氧基乙基)氨基)吡啶-2-基)-1-(6-甲酰基-5-(哌啶-4-基)吡啶-2-基)-1-甲基脲盐酸盐23(4mg,白色固体),产率:89%。
MS m/z(ESI):438[M+1]
1H NMR(400MHz,CD3OD)δ8.40(s,1H),8.01(d,J=8.4Hz,1H),7.37(d,J=7.6Hz,1H),6.92(s,1H),6.07(s,1H),3.71-3.58(m,6H),3.55(s,3H),3.43(s,3H),3.25-3.20(m,2H),3.19-3.15(m,1H),2.12-1.95(m,4H)。
实施例24
(S)-3-(5-氰基-4-((1-甲氧基丙烷-2-基)氧基)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000050
参照实施例18的操作步骤合成实施例24,但在第一步中用(S)-1-甲氧基丙烷-2-醇取代(R)-1-甲氧基丙烷-2-醇。
MS m/z(ESI):496[M+1]
1H NMR(400MHz,CDCl3)δ13.31(s,1H),10.26(s,1H),8.34(s,1H),7.99(s,1H),7.94(d,J=8.7Hz,1H),7.29(d,J=8.7Hz,1H),5.10(s,2H),4.99-4.74(m,1H),3.73-3.55(m,2H),3.52(s,3H),3.43(s,3H),3.37(t,J=5.3Hz,2H),3.20(s,2H),2.67(t,J=5.3Hz,2H),2.36(s,3H),1.41(d,J=6.3Hz,3H)。
实施例25
(R)-3-(5-氰基-4-((1-羟基丙烷-2-基)氧基)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000051
将化合物(R)-3-(5-氰基-4-((1-甲氧基丙烷-2-基)氧基)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲18(4.0g,8mmol)和二氯甲烷(80mL)混合,冰盐浴下滴加三溴化硼(20.2g,81mmol),滴加完毕,移除冰盐浴,室温下继续搅拌30分钟。此混合物缓慢倒入冰水(300mL)淬灭,饱和碳酸钠水溶液(200ml)调节pH至8-9,用二氯甲烷(150mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤除去干燥剂,减压脱溶得粗品,通过快速柱层析(二氯甲烷∶甲醇=20∶1)得到目标产物(R)-3-(5-氰基-4-((1-羟基丙烷-2-基)氧基)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲25(2.3g,4.8mmol,白色固体),产率:60%。
MS m/z(ESI):482[M+1]
1H NMR(400MHz,CDCl3)δ13.36(s,1H),10.26(s,1H),8.35(s,1H),7.99(s,1H),7.94(d,J=8.5Hz,1H),7.30(d,J=8.5Hz,1H),5.10 (s,2H),4.88-4.76(m,1H),3.91-3.78(m,2H),3.53(s,3H),3.42-3.38(s,2H),3.20(s,2H),2.75-2.63(m,2H),2.36(s,3H),1.41(d,J=6.0Hz,3H)。
实施例26
(S)-3-(5-氰基-4-((1-羟基丙烷-2-基)氧基)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲
Figure PCTCN2017088038-appb-000052
参照实施例25的操作步骤合成实施例26,但在第一步中用(S)-3-(5-氰基-4-((1-甲氧基丙烷-2-基)氧基)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲取代(R)-3-(5-氰基-4-((1-甲氧基丙烷-2-基)氧基)吡啶-2-基)-1-(6-甲酰基-5-((4-甲基-2-羰基哌嗪-1-基)甲基)吡啶-2-基)-1-甲基脲。
MS m/z(ESI):482[M+1]
1H NMR(400MHz,CDCl3)δ13.37(s,1H),10.26(s,1H),8.34(s,1H),7.97(s,1H),7.95(d,J=8.8Hz,1H),7.32(d,J=8.8Hz,1H),5.11(s,2H),4.91-4.74(m,1H),3.96-3.76(m,2H),3.54(s,3H),3.46-3.33(m,2H),3.22(s,2H),2.79-2.62(m,2H),2.38(s,3H),1.42(d,J=6.0Hz,3H)。
生物学实验
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成纤维细胞生长因子受体4(FGFR4)活性的影响。
实验方法概述如下:
使用HTRF激酶检测试剂盒,通过检测激酶反应中底物的磷酸化水平来测定FGFR4的体外活性。反应缓冲液包含以下组分:5倍稀释的Enzymatic buffer/kinase 5X(Cisbio,货号为62EZBFDD)(主要成分为50mM HEPES,pH 7.0)、5mM MgCl2、1mM DTT;人重组FGFR4 催化结构域蛋白(氨基酸460-802)购自清华蛋白质研究技术中心,用反应缓冲液稀释成0.5ng/uL的激酶溶液;底物反应溶液包括用反应缓冲液稀释成500nM的生物素标记的酪氨酸激酶底物(Cisbio,catalog number 62TK0PEC)和90uM ATP,检测液包括用检测缓冲液(Cisbio,货号为62SDBRDF)稀释成0.125ng/uL Eu3+标记的笼状抗体(Cisbio,货号61T66KLB)和31.25nM链霉亲和素标记的XL665(Cisbio,货号为610SAXLB)。
将化合物在100%DMSO中溶解稀释至100uM,然后用DMSO进行4倍的系列稀释至最低浓度为0.0061uM,每个浓度点再使用反应缓冲液稀释40倍。如果化合物IC50值非常低,可以降低化合物的起始浓度。
向384孔检测板(Thermo,货号264706)中添加4uL化合物溶液和2uL的FGFR4激酶溶液,混合均匀后室温孵育15分钟;随后加入4uL底物反应溶液,将反应混合物在室温孵育60分钟;随后加入与反应等体积的10uL检测液,混合均匀后室温放置。60分钟后,酶反应被检测液中的EDTA终止,磷酸化的产物同时被Eu3+标记的笼状抗体(供体)和链霉亲和素标记的XL665抗体(受体)识别,在激光激发后,靠近的供体和受体发生能量共振转移,其从供体(620nm)转移至受体(665nm)的能量可用Envision检测。665/620的比值与底物的磷酸化程度呈正相关,因此从而检测FGFR4激酶的活性。该实验中,未加蛋白组作为阴性对照(100%抑制),加蛋白但是未加化合物组作为阳性对照(0%抑制)。化合物对FGFR4活性抑制百分比可以用以下公式计算:
抑制百分比=100-100*(信号化合物-信号阴性对照)/(信号阳性对照-信号阴性对照)
化合物IC50值由10个浓度点用XLfit(ID Business Solutions Ltd.,UK)软件通过以下公式计算:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*slope factor))
其中Y为抑制百分比,Bottom为the bottom plateau of the curve(S型曲线的底部平台值),Top为the top plateau of the curve(S型曲线的顶部平台值),X为待测化合物浓度的对数值。
FGFR1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成纤维细胞生长因 子受体1(FGFR1)活性的影响。
实验方法概述如下:
使用HTRF激酶检测试剂盒,通过检测激酶反应中底物的磷酸化水平来测定FGFR1的体外活性。反应缓冲液包含以下组分:5倍稀释的Enzymatic buffer/kinase 5X(Cisbio,货号为62EZBFDD)(主要成分为50mM HEPES,pH 7.0)、5mM MgCl2、1mM DTT;人重组FGFR1催化结构域蛋白(氨基酸308-731)由公司自己纯化,用反应缓冲液稀释成0.6ng/uL的激酶溶液;底物反应溶液包括用反应缓冲液稀释成400nM的生物素标记的酪氨酸激酶底物(Cisbio,catalog number 62TK0PEC)和40uM ATP,检测液包括用检测缓冲液(Cisbio,货号为62SDBRDF)稀释成0.125ng/uL Eu3+标记的笼状抗体(Cisbio,货号61T66KLB)和25nM链霉亲和素标记的XL665(Cisbio,货号为610SAXLB)。
将化合物在100%DMSO中溶解稀释至1mM,然后用DMSO进行4倍的系列稀释至最低浓度为0.061uM,每个浓度点再使用反应缓冲液稀释40倍。如果化合物IC50值非常低,可以降低化合物的起始浓度。
向384孔检测板(Thermo,货号264706)中添加4uL化合物溶液和2uL的FGFR1激酶溶液,混合均匀后室温孵育15分钟;随后加入4uL底物反应溶液,将反应混合物在室温孵育60分钟;随后加入与反应等体积的10uL检测液,混合均匀后室温放置。60分钟后,酶反应被检测液中的EDTA终止,磷酸化的产物同时被Eu3+标记的笼状抗体(供体)和链霉亲和素标记的XL665抗体(受体)识别,在激光激发后,靠近的供体和受体发生能量共振转移,其从供体(620nm)转移至受体(665nm)的能量可用Envision检测。665/620的比值与底物的磷酸化程度呈正相关,因此从而检测FGFR1激酶的活性。该实验中,未加蛋白组作为阴性对照(100%抑制),加蛋白但是未加化合物组作为阳性对照(0%抑制)。化合物对FGFR1活性抑制百分比可以用以下公式计算:
抑制百分比=100-100*(信号化合物-信号阴性对照)/(信号阳性对照-信号阴性对照)
化合物IC50值由10个浓度点用XLfit(ID Business Solutions Ltd.,UK)软件通过以下公式计算:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*slope factor))
其中Y为抑制百分比,Bottom为the bottom plateau of the curve(S型曲线的底部平台值),Top为the top plateau of the curve(S型曲线的顶部平台值),X为待测化合物浓度的对数值。
FGFR2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成纤维细胞生长因子受体2(FGFR2)活性的影响。
实验方法概述如下:
使用HTRF激酶检测试剂盒,通过检测激酶反应中底物的磷酸化水平来测定FGFR2的体外活性。反应缓冲液包含以下组分:5倍稀释的Enzymatic buffer/kinase 5X(Cisbio,货号为62EZBFDD)(主要成分为50mM HEPES,pH 7.0)、5mM MgCl2、1mM DTT;人重组FGFR2催化结构域蛋白(氨基酸400-821)购自义翘神州生物技术有限公司,用反应缓冲液稀释成0.045ng/uL的激酶溶液;底物反应溶液包括用反应缓冲液稀释成800nM的生物素标记的酪氨酸激酶底物(Cisbio,catalog number 62TK0PEC)和50uM ATP,检测液包括用检测缓冲液(Cisbio,货号为62SDBRDF)稀释成0.125ng/uL Eu3+标记的笼状抗体(Cisbio,货号61T66KLB)和50nM链霉亲和素标记的XL665(Cisbio,货号为610SAXLB)。
将化合物在100%DMSO中溶解稀释至100uM,然后用DMSO进行4倍的系列稀释至最低浓度为0.0061uM,每个浓度点再使用反应缓冲液稀释40倍。如果化合物IC50值非常低,可以降低化合物的起始浓度。
向384孔检测板(Thermo,货号264706)中添加4uL化合物溶液和2uL的FGFR2激酶溶液,混合均匀后室温孵育15分钟;随后加入4uL底物反应溶液,将反应混合物在室温孵育60分钟;随后加入与反应等体积的10uL检测液,混合均匀后室温放置。60分钟后,酶反应被检测液中的EDTA终止,磷酸化的产物同时被Eu3+标记的笼状抗体(供体)和链霉亲和素标记的XL665抗体(受体)识别,在激光激发后,靠近的供体和受体发生能量共振转移,其从供体(620nm)转移至受体(665nm)的能量可用Envision检测。665/620的比值与底物的磷酸化程度呈正相关,因此从而检测FGFR2激酶的活性。该实验中,未加蛋白组作为阴性对照(100%抑制),加蛋白但是未加化合物组作为阳性 对照(0%抑制)。化合物对FGFR2活性抑制百分比可以用以下公式计算:
抑制百分比=100-100*(信号化合物-信号阴性对照)/(信号阳性对照-信号阴性对照)
化合物IC50值由10个浓度点用XLfit(ID Business Solutions Ltd.,UK)软件通过以下公式计算:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*slope factor))
其中Y为抑制百分比,Bottom为the bottom plateau of the curve(S型曲线的底部平台值),Top为the top plateau of the curve(S型曲线的顶部平台值),X为待测化合物浓度的对数值。
FGFR3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成纤维细胞生长因子受体3(FGFR3)活性的影响。
实验方法概述如下:
使用HTRF激酶检测试剂盒,通过检测激酶反应中底物的磷酸化水平来测定FGFR3的体外活性。反应缓冲液包含以下组分:5倍稀释的Enzymatic buffer/kinase 5X(Cisbio,货号为62EZBFDD)(主要成分为50mM HEPES,pH 7.0)、5mM MgCl2、1mM DTT;人重组FGFR3催化结构域蛋白(氨基酸399-806)购自义翘神州生物技术有限公司,用反应缓冲液稀释成0.3ng/uL的激酶溶液;底物反应溶液包括用反应缓冲液稀释成1000nM的生物素标记的酪氨酸激酶底物(Cisbio,catalog number 62TK0PEC)和90uM ATP,检测液包括用检测缓冲液(Cisbio,货号为62SDBRDF)稀释成0.125ng/uL Eu3+标记的笼状抗体(Cisbio,货号61T66KLB)和62.5nM链霉亲和素标记的XL665(Cisbio,货号为610SAXLB)。
将化合物在100%DMSO中溶解稀释至100uM,然后用DMSO进行4倍的系列稀释至最低浓度为0.0061uM,每个浓度点再使用反应缓冲液稀释40倍。如果化合物IC50值非常低,可以降低化合物的起始浓度。
向384孔检测板(Thermo,货号264706)中添加4uL化合物溶液和2uL的FGFR3激酶溶液,混合均匀后室温孵育15分钟;随后加入4uL底物反应溶液,将反应混合物在室温孵育60分钟;随后加入与反应等体积的10uL检测液,混合均匀后室温放置。60分钟后,酶反应被 检测液中的EDTA终止,磷酸化的产物同时被Eu3+标记的笼状抗体(供体)和链霉亲和素标记的XL665抗体(受体)识别,在激光激发后,靠近的供体和受体发生能量共振转移,其从供体(620nm)转移至受体(665nm)的能量可用Envision检测。665/620的比值与底物的磷酸化程度呈正相关,因此从而检测FGFR3激酶的活性。该实验中,未加蛋白组作为阴性对照(100%抑制),加蛋白但是未加化合物组作为阳性对照(0%抑制)。化合物对FGFR2活性抑制百分比可以用以下公式计算:
抑制百分比=100-100*(信号化合物-信号阴性对照)/(信号阳性对照-信号阴性对照)
化合物IC50值由10个浓度点用XLfit(ID Business Solutions Ltd.,UK)软件通过以下公式计算:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*slope factor))
其中Y为抑制百分比,Bottom为the bottom plateau of the curve(S型曲线的底部平台值),Top为the top plateau of the curve(S型曲线的顶部平台值),X为待测化合物浓度的对数值。
生物测试实施例:A:<10nM,B:10-100nM,C:100-1000nM,D:>1000nM,ND:未测
Figure PCTCN2017088038-appb-000053
Figure PCTCN2017088038-appb-000054
本发明的化合物对FGFR4具有选择性抑制作用。
Hep3B细胞增殖抑制的测定
使用发光细胞活力测试实验评估本发明的化合物对Hep3B细胞增殖的影响。
实验方法概述如下:
使用CellTilter-Glo(CTG)检测试剂盒,通过采用一种独特的、稳定性荧光素酶检测有活力细胞代谢的指示剂ATP,试验中产生的发光信号和培养基中的有活力细胞数呈正比,从而检测Hep3B的细胞增殖状况。
CellTilter-Glo试剂(Promega,G7572)由CellTilter-Glo冻干粉和CellTilter-Glo缓冲液组成,使用时将冻干粉溶解到缓冲液中即可。
Hep3B细胞(ATCC,HB-8064)(细胞来源中国科学院上海生命科学研究院)培养在DMEM完全培养基(Thermofisher,11995073)中含10%FBS(GBICO,10099-141)和100units/ml青链霉素混合液(Thermofisher,15140122),当细胞在培养容器中覆盖率达80-90%时,用0.25%胰酶(含EDTA)(Thermofisher,25200056)消化吹散后种植于白色384孔板(Thermofisher,164610),每孔1000细胞(27μ1DMEM完全培养基),然后384孔板置于37℃,5%CO2的培养箱中培养过夜(18-20小时)。将化合物在100%DMSO中溶解稀释至5mM,然后用DMSO进行4倍的系列稀释至最低浓度为0.061μM,每个浓度点再使用FBS-free的DMEM培养基稀释50倍。如果化合物IC50值非常低,可以降低化合物的起始浓度。过夜后每孔加入3μlDMEM稀释后的化合物,轻轻离心混匀,其中,加10μM BLU9931组作为阴性对 照(100%抑制),加0.2%DMSO组作为阳性对照(0%抑制)。该384孔板置于37℃,5%CO2的培养箱中继续培养,72小时后取出于室温放置30分钟,CTG试剂也取出平衡至室温,每孔加15μl CTG试剂,置于振荡器上轻轻震荡3分钟以确保细胞裂解充分,放置10分钟使冷光信号稳定,然后用EnVision(Perkin Elmer)读取冷光信号。
化合物对Hep3B细胞增殖抑制的百分比可以用以下公式计算:
抑制百分比=100-100*(信号化合物-信号阴性对照)/(信号阳性对照-信号阴性对照)
化合物IC50值由8个浓度点用XLfit(ID Business Solutions Ltd.,UK)软件通过以下公式计算:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*slope factor))
其中Y为抑制百分比,Bottom为the bottom plateau of the curve(S型曲线的底部平台值),Top为the top plateau of the curve(S型曲线的顶部平台值),X为待测化合物浓度的对数值。
生物测试实施例:A:<50nM,B:50-100nM,
Figure PCTCN2017088038-appb-000055
由上表可见,本发明化合物对Hep3B细胞增殖具有良好抑制作用。

Claims (10)

  1. 一种如式I所示的化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐
    Figure PCTCN2017088038-appb-100001
    其中环A和R各自独立选自取代或未取代的芳基、杂芳基,当A或R被取代时,可被一个或多个取代基在任意位置取代,所述的取代基独立选自氢、卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、芳基、杂芳基、醛基、-C(O)R1、羧基、烯基、炔基、-OR1、-NR2R3,其中所述烷基、烯基、炔基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
    X选自CR7R8、NR7、O、S的二价基团;
    Y选自-C(O)-、-C(=NR9)-、-S(O)m-;
    R1、R2、R3、R4、R5、R6各自独立地选自氢、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基、烯基、炔基,其中所述的R2和R3、R5和R6可与它们所连接的N原子一起形成3-7元的杂环基;所述的R7和R8可与它们所连接的C原子一起形成3-8元的环基或3-8元单环杂环基;
    R7、R8各自独立地选自氢、卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、芳基、杂芳基、醛基、-C(O)R1、羧基、烯基、炔基、-OR1、-NR2R3,其中所述烷基、烯基、炔基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
    R9独立地选自氢、C1-C8烷基、C3-C8环基、3-8元杂环基、芳基、杂芳基、醛基、-C(O)R1、烯基、炔基,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
    且m为1或2。
  2. 根据权利要求1所述的化合物、其异构体、前药、稳定的同位素衍生物或其药学上可接受的盐,其特征在于所述的如式I所示的化合物为式II所示:
    Figure PCTCN2017088038-appb-100002
    Z1、Z2、Z3各自独立地选自CRZ1、CRZ2、CRZ3或N,并且:
    Z1为N时,Z2、Z3不同时为N;
    Z2为N时,Z1、Z3不同时为N;
    Z3为N时,Z1、Z2不同时为N;
    RZ1、RZ2、RZ3各自独立地选自氢、卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、芳基、杂芳基、醛基、-C(O)R1、羧基、烯基、炔基、-OR1、-NR2R3,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
    X、Y、R、R1-9定义如权利要求1中所述;
    当Z1为CCH2OH、CCH2COOH、C-(4-哌啶)时,化合物(2-1)、(2-2)、(2-3)可能以异构体(2-1A)、(2-2A)、(2-3A)的形式存在:
    Figure PCTCN2017088038-appb-100003
  3. 根据权利要求1-2任一项所述的化合物、其异构体、前药、稳定的同位素衍生物或其药学上可接受的盐,其特征在于所述的如式I所示的化合物为式IIIa、IIIb、IIIc、IIId、IIIe、IIIf:
    Figure PCTCN2017088038-appb-100004
    RZ1、RZ2、RZ3、R10、R11各自独立地选自氢、卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、芳基、杂芳基、醛基、-C(O)R1、羧基、烯基、炔基、-OR1、-NR2R3,其中所述烷基、烯基、炔基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、 C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
    R7独立地选自H、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
    R1-6定义如权利要求1中所述;
    所述的R2和R3、R5和R6与它们所连接的N原子一起可形成3-8元的杂环基。
  4. 根据权利要求1-3任一项所述的化合物、其异构体、前药、稳定的同位素衍生物或其药学上可接受的盐,其特征在于所述的如式I所示的化合物为式IV;
    Figure PCTCN2017088038-appb-100005
    RZ1、RZ2各自独立地选自氢、卤素、C1-C4烷基、C3-C7环基、4-6元单环杂环基、5-6元单环杂芳基或单环芳基、醛基、酮基、羧基、氰基、OR1、NR2R3,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、羟基、C1-C4烷基、C3-C7环基、4-6元杂环基、芳基或杂芳基的取代基所取代;
    R7选自H、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR4、-OC(O)NR5R6、-C(O)OR4、-C(O)NR5R6、-C(O)R4、-NR5R6、-NR5C(O)R4、-NR4C(O)NR5R6、-S(O)mR4、-NR5S(O)mR4、-SR4、-NR4S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
    R10独立地选自氢、卤素、卤代C1-C4烷基、氰基;
    R11独立地选自氢、卤素、C1-C4烷基、卤代C1-C4烷氧基、C1-C6烷氧基、HO-C1-C4烷氧基、氰基、NR2R3、C1-C4烷氧基C1-C4烷氧基、C1-C4烷氧基卤代C1-C4烷氧基;
    R1、R2、R3各自独立地选自氢、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基、C1-C3烷硫基、被羟基在任意位置取代的卤代烷氧基;
    R4-6定义如权利要求1中所述。
  5. 根据权利要求1-4任一项所述的化合物、其异构体、前药、稳定的同位素衍生物或其药学上可接受的盐,其特征在于所述的如式I所示的化合物为式V;
    Figure PCTCN2017088038-appb-100006
    RZ1、RZ2各自独立地选自氢、卤素、C1-C4烷基、C3-C7环基、5-6元单环杂环基、5-6元单环杂芳基或单环芳基、醛基、羧基,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、羟基、C1-C4烷基、5-6元杂环基、芳基或杂芳基的取代基所取代;
    R7选自氢、C1-C4烷基、C3-6环基,其中所述烷基或环基任选被一个或多个选自C1-C3烷基、4-6元单环杂环基的取代基所取代;
    R11选自NR2R3、C1-C3烷氧基、-O(CH2)0-1-R4;其中,R4独立地选自氢、C1-C8烷基、HO-C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR5、-OC(O)NR5R6、-C(O)OR5、-C(O)NR5R6、-C(O)R5、-NR5R6、-NR5C(O)R6、-NR7C(O)NR5R6、-S(O)mR5、-NR5S(O)mR6、-SR5、-NR7S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
    R2、R3独立地选自氢、C1-C8烷基、C3-C8环基、3-8元单环杂环基、单环杂芳基或单环芳基、C1-C3烷硫基、被羟基在任意位置取代的卤代烷氧基,其中所述烷基、环基、杂环基、芳基或杂芳基任选被一 个或多个选自卤素、氰基、C1-C8烷基、C3-C8环基、3-8元杂环基、-OR5、-OC(O)NR5R6、-C(O)OR5、-C(O)NR5R6、-C(O)R5、-NR5R6、-NR5C(O)R6、-NR7C(O)NR5R6、-S(O)mR6、-NR5S(O)mR6、-SR5、-NR7S(O)mNR5R6、-S(O)mNR5R6的取代基所取代;
    R4-6定义如权利要求1中所述。
  6. 根据权利要求1所述的通式(I)所示的化合物、其异构体、前药、稳定的同位素衍生物或其药学上可接受的盐,其特征在于所述的如式I所示的化合物为
    Figure PCTCN2017088038-appb-100007
    Figure PCTCN2017088038-appb-100008
  7. 一种药物组合物,所述药物组合物根据权利要求1-6任意一项所述的化合物或其异构体、前药、稳定的同位素衍生物或其药学上可接受的盐及药学上可接受的载体、稀释剂、赋形剂。
  8. 根据权利要求1-6任意一项所述的化合物或其异构体、前药、稳定的同位素衍生物或药学上可接受的盐、或根据权利要求7所述的药物组合物在制备药物中的用途,其中所述药物用于治疗或者预防FGFR、尤其是FGFR4介导的疾病,例如癌症、尤其是肝细胞癌。
  9. 一种治疗或者预防FGFR、尤其是FGFR4介导的疾病(例如癌症、尤其是肝细胞癌)的方法,其包括给予有需要的患者治疗有效量的根据权利要求1-6任意一项所述的化合物或其异构体、前药、稳定的同位素衍生物或药学上可接受的盐、或根据权利要求7所述的药物组合物。
  10. 根据权利要求1-6任意一项所述的化合物或其异构体、前药、稳定的同位素衍生物或药学上可接受的盐、或根据权利要求7所述的药物组合物,其用于治疗或者预防FGFR、尤其是FGFR4介导的疾病,例如癌症、尤其是肝细胞癌。
PCT/CN2017/088038 2016-07-13 2017-06-13 作为fgfr抑制剂的杂环化合物 WO2018010514A1 (zh)

Priority Applications (11)

Application Number Priority Date Filing Date Title
EP17826852.0A EP3489223A4 (en) 2016-07-13 2017-06-13 HETEROCYCLIC COMPOUND USED AS AN FGFR INHIBITOR
AU2017295628A AU2017295628B2 (en) 2016-07-13 2017-06-13 Heterocyclic compound used as FGFR inhibitor
MX2019000451A MX2019000451A (es) 2016-07-13 2017-06-13 Compuesto heterociclico utilizado como inhibidor de fgfr.
RU2019100164A RU2742485C2 (ru) 2016-07-13 2017-06-13 Гетероциклическое соединение, используемое как ингибитор fgfr
KR1020197004058A KR102386428B1 (ko) 2016-07-13 2017-06-13 Fgfr 억제제로서 사용되는 헤테로시클릭 화합물
SG11201900298RA SG11201900298RA (en) 2016-07-13 2017-06-13 Heterocyclic compound used as fgfr inhibitor
JP2019523154A JP6896852B2 (ja) 2016-07-13 2017-06-13 Fgfr阻害剤として使用される複素環式化合物
CA3030070A CA3030070A1 (en) 2016-07-13 2017-06-13 Heterocyclic compound used as fgfr inhibitor
CN201780002825.9A CN108349896B (zh) 2016-07-13 2017-06-13 作为fgfr抑制剂的杂环化合物
PH12019500036A PH12019500036A1 (en) 2016-07-13 2019-01-04 Heterocyclic compound used as fgfr inhibitor
US16/246,236 US10590109B2 (en) 2016-07-13 2019-01-11 Heterocyclic compounds used as FGFR inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610550151.3A CN107619388A (zh) 2016-07-13 2016-07-13 作为fgfr抑制剂的杂环化合物
CN201610550151.3 2016-07-13

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/246,236 Continuation-In-Part US10590109B2 (en) 2016-07-13 2019-01-11 Heterocyclic compounds used as FGFR inhibitors

Publications (1)

Publication Number Publication Date
WO2018010514A1 true WO2018010514A1 (zh) 2018-01-18

Family

ID=60951887

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/088038 WO2018010514A1 (zh) 2016-07-13 2017-06-13 作为fgfr抑制剂的杂环化合物

Country Status (12)

Country Link
US (1) US10590109B2 (zh)
EP (1) EP3489223A4 (zh)
JP (1) JP6896852B2 (zh)
KR (1) KR102386428B1 (zh)
CN (2) CN107619388A (zh)
AU (1) AU2017295628B2 (zh)
CA (1) CA3030070A1 (zh)
MX (1) MX2019000451A (zh)
PH (1) PH12019500036A1 (zh)
RU (1) RU2742485C2 (zh)
SG (2) SG11201900298RA (zh)
WO (1) WO2018010514A1 (zh)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
EP3663285A4 (en) * 2017-08-04 2021-05-05 Abbisko Therapeutics Co., Ltd. FORMYL PYRIDINE DERIVATIVE WITH FGFR4-INHIBITING EFFECT, MANUFACTURING METHOD FOR IT AND USES THEREOF
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
US11247987B2 (en) 2017-10-06 2022-02-15 Forma Therapeutics, Inc. Inhibiting ubiquitin specific peptidase 30
US11535618B2 (en) 2018-10-05 2022-12-27 Forma Therapeutics, Inc. Fused pyrrolines which act as ubiquitin-specific protease 30 (USP30) inhibitors

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108129471A (zh) * 2018-03-20 2018-06-08 韩邦森 一种噻唑取代嘧啶类化合物的合成工艺
CN111039874B (zh) * 2019-12-13 2022-11-04 南开大学 一种2-胺基-4-甲基嘧啶化合物的零废水制备方法

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1508130A (zh) * 2002-12-18 2004-06-30 中国科学院大连化学物理研究所 N-苯基-n’-嘧啶基取代脲类衍生物的合成方法
CN1678311A (zh) * 2002-06-27 2005-10-05 诺沃挪第克公司 用作治疗剂的芳基羰基衍生物
WO2006009741A1 (en) * 2004-06-23 2006-01-26 Eli Lilly And Company Kinase inhibitors
WO2007059341A2 (en) * 2005-11-16 2007-05-24 Sgx Pharmaceuticals, Inc. Pyrazolothiazole protein kinase modulators
CN101304998A (zh) * 2005-08-04 2008-11-12 艾文蒂斯药品公司 取代的7-氮杂-吲唑类,含有其的组合物,制备方法及其用途
CN101553473A (zh) * 2006-11-10 2009-10-07 赛诺菲-安万特 在取代的吡唑、包含它们的组合物、生产方法和用途
CN102471280A (zh) * 2009-07-03 2012-05-23 赛诺菲 吡唑衍生物、其制备及其治疗用途
WO2013024427A1 (en) * 2011-08-16 2013-02-21 Glenmark Pharmaceuticals S.A. Novel urea derivatives as tec kinase inhibitors and uses thereof

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2850652B1 (fr) 2003-01-31 2008-05-30 Aventis Pharma Sa Nouveaux derives d'uree cyclique, leur preparation et leur utilisation pharmaceutique comme inhibiteurs de kinases
GB0512324D0 (en) * 2005-06-16 2005-07-27 Novartis Ag Organic compounds
EP1790342A1 (de) 2005-11-11 2007-05-30 Zentaris GmbH Pyridopyrazin-Derivate und deren Verwendung als Modulatoren der Signaltransduktionswege
BRPI0619146A2 (pt) 2005-12-02 2011-09-13 Bayer Pharmaceuticals Corp derivados de 4-amino-pirroltriazina substituìda úteis no tratamento de disordens e doenças hiperproliferativas associadas com angiogênesis
WO2015059668A1 (en) * 2013-10-25 2015-04-30 Novartis Ag Ring-fused bicyclic pyridyl derivatives as fgfr4 inhibitors
US9802917B2 (en) 2015-03-25 2017-10-31 Novartis Ag Particles of N-(5-cyano-4-((2-methoxyethyl)amino)pyridin-2-yl)-7-formyl-6-((4-methyl-2-oxopiperazin-1-yl)methyl)-3,4-dihydro-1,8-naphthyridine-1(2H)-carboxamide
US10189813B2 (en) * 2015-03-25 2019-01-29 Novartis Ag Formylated N-heterocyclic derivatives as FGFR4 inhibitors

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1678311A (zh) * 2002-06-27 2005-10-05 诺沃挪第克公司 用作治疗剂的芳基羰基衍生物
CN1508130A (zh) * 2002-12-18 2004-06-30 中国科学院大连化学物理研究所 N-苯基-n’-嘧啶基取代脲类衍生物的合成方法
WO2006009741A1 (en) * 2004-06-23 2006-01-26 Eli Lilly And Company Kinase inhibitors
CN101304998A (zh) * 2005-08-04 2008-11-12 艾文蒂斯药品公司 取代的7-氮杂-吲唑类,含有其的组合物,制备方法及其用途
WO2007059341A2 (en) * 2005-11-16 2007-05-24 Sgx Pharmaceuticals, Inc. Pyrazolothiazole protein kinase modulators
CN101553473A (zh) * 2006-11-10 2009-10-07 赛诺菲-安万特 在取代的吡唑、包含它们的组合物、生产方法和用途
CN102471280A (zh) * 2009-07-03 2012-05-23 赛诺菲 吡唑衍生物、其制备及其治疗用途
WO2013024427A1 (en) * 2011-08-16 2013-02-21 Glenmark Pharmaceuticals S.A. Novel urea derivatives as tec kinase inhibitors and uses thereof

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
AN, YING ET AL.: "Design and synthesis of novel benzoxazole analogs as Aurora B kinase inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 26, no. 13, 7 May 2016 (2016-05-07), pages 3067 - 3072, XP029568927 *
CLAYDEN, J. ET AL.: "N, N'-Diarylureas: ANew Family of Atropisomers Exhibiting Highly Diastereoselective Reactivity", J. ORG. CHEM., vol. 73, no. 12, 10 April 2008 (2008-04-10), pages 4415 - 4423, XP055457130 *
GAOQUAN LI ET AL.: "Synthesis and biological evaluation of 1-(2, 4, 5-trisubsti-tuted phenyl)-3-(5-cyanopyrazin-2-yl) ureas as potent Chk 1 kinase inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 16, no. 8, 30 January 2006 (2006-01-30), pages 2293 - 2298, XP027966327 *
KIM, K.H. ET AL.: "Conformational Switching on Platinum (II) Coordination Plane Triggered by Oxalate Anion", BULL. KOREAN CHEM. SOC., vol. 32, no. 9, 31 December 2011 (2011-12-31), pages 3497 - 3450, XP055457132 *
LU , T.W. ET AL.: "Molecular Switch Based on Very Weak Association between BPX26C6 and Two Recognition Units", ORGANIC LETTERS, vol. 15, no. 22, 30 October 2013 (2013-10-30), pages 5742 - 5745, XP055457134 *
NAGAMATSU, K. ET AL.: "Reactions of 2-Triphenylphosphoimino-L-Azaazulenes with Aryl Isocyanates and Aryl Isothiocyanates", HETEROCYCLES, vol. 67, no. 1, 31 December 2006 (2006-12-31), pages 337 - 351, XP055457126 *
See also references of EP3489223A4 *
YANG, ZHAO ET AL.: "Identification of inhibitors for vascular endothelial growth factor receptor by using dynamic combinatorial chemistry", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 26, no. 7, 22 January 2016 (2016-01-22), pages 1671 - 1674, XP029453962 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3663285A4 (en) * 2017-08-04 2021-05-05 Abbisko Therapeutics Co., Ltd. FORMYL PYRIDINE DERIVATIVE WITH FGFR4-INHIBITING EFFECT, MANUFACTURING METHOD FOR IT AND USES THEREOF
US11312701B2 (en) 2017-08-04 2022-04-26 Abbisko Therapeutics Co., Ltd Formylpyridine derivative having FGFR4 inhibitory activity, preparation method therefor and use thereof
US11247987B2 (en) 2017-10-06 2022-02-15 Forma Therapeutics, Inc. Inhibiting ubiquitin specific peptidase 30
US11535618B2 (en) 2018-10-05 2022-12-27 Forma Therapeutics, Inc. Fused pyrrolines which act as ubiquitin-specific protease 30 (USP30) inhibitors
US11814386B2 (en) 2018-10-05 2023-11-14 Forma Therapeutics, Inc. Fused pyrrolines which act as ubiquitin-specific protease 30 (USP30) inhibitors
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use

Also Published As

Publication number Publication date
EP3489223A4 (en) 2020-01-15
RU2742485C2 (ru) 2021-02-08
US10590109B2 (en) 2020-03-17
JP2019522055A (ja) 2019-08-08
US20190144427A1 (en) 2019-05-16
KR102386428B1 (ko) 2022-04-14
CN107619388A (zh) 2018-01-23
CN108349896A (zh) 2018-07-31
EP3489223A1 (en) 2019-05-29
CA3030070A1 (en) 2018-01-18
RU2019100164A3 (zh) 2020-08-27
SG11201900298RA (en) 2019-02-27
AU2017295628A1 (en) 2019-02-21
CN108349896B (zh) 2021-06-01
KR20190032420A (ko) 2019-03-27
MX2019000451A (es) 2019-09-27
PH12019500036A1 (en) 2019-10-28
JP6896852B2 (ja) 2021-06-30
AU2017295628B2 (en) 2021-05-13
SG10202013038VA (en) 2021-01-28
RU2019100164A (ru) 2020-08-13

Similar Documents

Publication Publication Date Title
CN108349896B (zh) 作为fgfr抑制剂的杂环化合物
TWI551595B (zh) 2,4-disubstituted benzene-1,5-diamine derivatives and their use, Its preparation of pharmaceutical compositions and pharmaceutical compositions
JP6035423B2 (ja) 新規な縮合ピリミジン化合物又はその塩
JP6457623B2 (ja) 2,4−二置換7H−ピロロ[2,3−d]ピリミジン誘導体、その製造方法および医薬における使用
WO2016026445A1 (zh) 作为fgfr激酶抑制剂的吲唑类化合物及其制备和应用
WO2021143701A1 (zh) 嘧啶-4(3h)-酮类杂环化合物、其制备方法及其在医药学上的应用
JP2024505732A (ja) ピリドピリミジノン系誘導体及びその製造方法と使用
JPWO2019208812A1 (ja) ベンゾイソオキサゾール化合物
KR20200013718A (ko) 바닌 억제제로서의 헤테로방향족 화합물
KR20200135827A (ko) Prmt5 억제제로서 치환된 이미다졸리딘-2-온 유도체
CN112313207B (zh) 一种氰基取代吡啶及氰基取代嘧啶类化合物、制备方法及其应用
WO2020215998A1 (zh) 嘧啶并五元杂环类化合物及其作为突变型idh2抑制剂的用途
WO2022017434A1 (zh) 一类具有激酶抑制活性的化合物
WO2018228275A1 (zh) 作为mnk抑制剂的杂环化合物
WO2021197250A1 (zh) 作为转染期间重排激酶抑制剂的新的化合物
WO2023036252A1 (zh) 吡咯并嘧啶类或吡咯并吡啶类衍生物及其医药用途
WO2023178928A1 (zh) 2-氨基-4-吲哚基嘧啶类化合物及其制备方法与应用
WO2020156319A1 (zh) N-甲酰胺衍生物、其制备方法及其在医药上的用途
WO2022148196A1 (zh) 多激酶抑制剂及其用途
JP7216105B2 (ja) Erkキナーゼ阻害活性を有する化合物及びその使用
CN114555597A (zh) 异柠檬酸脱氢酶(idh)抑制剂
WO2020207419A1 (zh) 哌嗪酰胺衍生物,其制备方法及其在医药上的用途
WO2019242587A1 (zh) 一种高选择性FGFR i抑制剂及其制备方法和应用
WO2022028556A1 (zh) Cdk9抑制剂及其用途
WO2022272106A1 (en) Cdk2 inhibitors and methods of using the same

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: 17826852

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3030070

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2019523154

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20197004058

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017295628

Country of ref document: AU

Date of ref document: 20170613

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017826852

Country of ref document: EP

Effective date: 20190213