WO2018090939A1 - 8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物 - Google Patents

8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物 Download PDF

Info

Publication number
WO2018090939A1
WO2018090939A1 PCT/CN2017/111230 CN2017111230W WO2018090939A1 WO 2018090939 A1 WO2018090939 A1 WO 2018090939A1 CN 2017111230 W CN2017111230 W CN 2017111230W WO 2018090939 A1 WO2018090939 A1 WO 2018090939A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino
pyrimido
phenyl
dihydroimidazo
chloro
Prior art date
Application number
PCT/CN2017/111230
Other languages
English (en)
French (fr)
Inventor
蔡遂雄
田野
Original Assignee
上海瑛派药业有限公司
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 claimed from CN201710440456.3A external-priority patent/CN109020981A/zh
Application filed by 上海瑛派药业有限公司 filed Critical 上海瑛派药业有限公司
Priority to US16/461,512 priority Critical patent/US10703759B2/en
Priority to ES17872807T priority patent/ES2968252T3/es
Priority to CA3043945A priority patent/CA3043945A1/en
Priority to AU2017359844A priority patent/AU2017359844B2/en
Priority to KR1020197017242A priority patent/KR102534962B1/ko
Priority to CN201780068490.0A priority patent/CN109906227B/zh
Priority to EP17872807.7A priority patent/EP3543242B1/en
Priority to JP2019527384A priority patent/JP7240784B2/ja
Publication of WO2018090939A1 publication Critical patent/WO2018090939A1/zh
Priority to US16/883,397 priority patent/US11345711B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/14Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the invention belongs to the field of medicinal chemistry.
  • the present invention particularly relates to 8,9-dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-one compounds, and as therapeutically effective Wee1 kinase inhibitors, And the application of anticancer drugs.
  • the process of growth and proliferation of eukaryotic cells involves the mother cell producing two identical daughter cells through mitosis of the cell chromosome by accurately replicating its genome including genetic information.
  • the cell proliferation and division process is called the cell cycle, which includes the entire process from the completion of one division to the completion of the next division.
  • the cell cycle includes four growth stages, a protein after mitosis, a large number of synthetic G1 phases such as RNA, a S phase of DNA synthesis replication, a G2 phase of preparation before mitosis, and a M phase of mitosis.
  • the cells decide to divide and proliferate through the cell cycle depending on the condition of the cell and the need, or stop. Cell proliferation and division must maintain the integrity and correctness of their genetic information. Whether to enter the next stage of the cell cycle until the completion of the entire cell cycle is ensured and completed by multiple checkpoints during the cell cycle.
  • Each cell cycle checkpoint consists of a very complex system and consists of multiple factors.
  • the checkpoint during the G1 period determines whether or not the cell enters the cell cycle by examining the state inside and outside the cell, thereby determining whether the cell enters S phase DNA synthesis.
  • the G1 checkpoint is a complex system that includes the famous CDK4/CDK6.
  • Another important checkpoint is the completion of DNA replication (S phase) in the cell into the cell growth phase (G2 phase), the so-called G2-M checkpoint. This checkpoint examines whether the cell has DNA damage or defects after synthesizing the DNA, thereby determining whether the cell undergoes mitosis (M-phase) of the following chromosome segregation.
  • the cell cycle checkpoint at this stage includes the complex kinase Cdk1 complex including Cyclin-B-cdc2 (Nurse, P., 1990, Nature 344, 503-508).
  • Cdk1 Activation of Cdk1 leads to the initiation of mitosis, and its subsequent inactivation is accompanied by the completion of mitosis.
  • the activity of Cdk1 is regulated by cdc2 binding to Cyclin-A or Cyclin-B and its phosphorylation.
  • activation of the cyclin B-Cdk1 complex enables mitosis of cells (Lindqvist, A., et al, 2009, The Journal of cell biology 185, 193-202).
  • Cdc2 is maintained in an inactive state by phosphorylation prior to cell entry into mitosis. Its phosphorylation state is achieved by the tyrosine kinase Wee1 or the like. In addition, there are M phase cell cycle checkpoints.
  • Wee1 phosphorylates tyrosine 15 (Y15) on Cdk1 thereby inhibiting the activity of Cdk1 (McGowan, CH, et al, 1993, The EMBO journal 12, 75-85; Parker, LL, et al, 1992, Science 257, 1955 -1957). Therefore, Wee1 is a key inhibitory regulator of Cdk1 activity and plays an important role in the G2-M phase of detection, ensuring mitosis in the absence of DNA damage after DNA replication is completed (O'Connell, et al., 1997, The EMBO journal 16, 545-554).
  • inhibition of Wee1 activity can selectively cause cancer cells that are defective in cell cycle checkpoints to die; at the same time, it has little effect on normal cells with normal cell cycle checkpoints.
  • inhibitors of Wee1 are potentially targeted agents for the treatment of cancer and other cell proliferative disorders.
  • Wee1 inhibitors can be used in combination with anticancer drugs that cause DNA damage or inhibit DNA repair mechanisms, including the PARP inhibitor olaparib. Niraparib, Rucaparib, and Talazoparib; HDAC inhibitors vorinostat, romidepsin, pabisstat, and belistat, etc., are used to treat cancer or other cell proliferative disorders.
  • Wee1 inhibitors may also be combined with other anticancer drugs associated with cell division cell cycle checkpoints, including Chk1/2 inhibitors, CDK4/6 inhibitors such as Pabsini, ATM/ATR inhibitors, etc. Treat diseases such as cancer.
  • AZD1775 is the first Wee1 kinase inhibitor with single-agent anti-tumor activity in preclinical models.
  • Phase I clinical studies have demonstrated the efficacy of AZD1775 in single-agent patients with solid tumors carrying BRCA mutations, and confirmed the differentiation mechanism and DNA damage response by paired tumor biopsy to confirm its Wee1 kinase inhibition mechanism (JClin Oncol, 2015, 33) :3409-3415).
  • JClin Oncol, 2015, 33 :3409-3415
  • the efficacy of single drug in patients with advanced solid tumors and the combination with gemcitabine, cisplatin or carboplatin was studied, showing that it does not matter at a certain dose.
  • WO2012161812 discloses the following tricyclic compounds as Wee1 kinase inhibitors.
  • X is N or CR 1 ;
  • Y is N or CR 2 ;
  • Z is O, S or NH;
  • R 1 and R 2 are H or C 1-6 alkyl;
  • R 3 is C 1-8 alkyl, C 2-8 alkenyl, C 3-8 cycloalkyl, aryl, or heteroaryl, etc.;
  • R 4 is phenyl, naphthyl, tetrahydronaphthyl, anthracenyl or indanyl, or 5-16 yuan A cyclic, bicyclic or tricyclic heterocyclic group or the like.
  • WO2005021551 discloses the following tetracyclic pyrimidine or pyridine compounds as protein kinase inhibitors.
  • X is N or CH;
  • Y is NH, N(CN), O or S;
  • L is a 4-atom chain consisting of C and N atoms;
  • R a is H, C 1-8 alkyl, CN, benzene Or a benzyl group;
  • R 1 and R 2 are independently a saturated or unsaturated 5-, 6-, or 7-membered monocyclic ring which may be substituted, or 6-, 7-, 8-, 9-, 10- or 11-membered bicyclic ring (containing 0, 1, 2, 3 or 4 atoms selected from N, O and S, wherein O and S atoms are not present simultaneously, and C atoms in the ring are replaced by 0, 1 or 2 oxy groups )Wait.
  • the present invention provides novel 8,9-dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-one compounds as A kinase inhibitor, particularly a Wee1 kinase inhibitor.
  • the invention also provides pharmaceutical compositions comprising an effective amount of a compound of Formulas I, II and III for use in the treatment of cancer.
  • the pharmaceutical composition may also contain one or more pharmaceutically acceptable carriers or diluents for the treatment of cancer.
  • the pharmaceutical composition may further comprise at least one known anticancer drug or a pharmaceutically acceptable salt of the anticancer drug for treating cancer.
  • the invention also relates to a process for the preparation of the novel compounds of structural formulas I, II and III.
  • a compound useful in the present invention is a compound of formula I or a pharmaceutically acceptable salt or prodrug thereof:
  • A is N or CR 15 ;
  • R 1 is hydrogen, a C 1 -C 8 alkyl group which may be substituted, a C 2 -C 8 alkenyl group which may be substituted, a C 3 -C 8 cycloalkyl group which may be substituted, an aryl group which may be substituted, a substituted heterocyclic group or a heteroaryl group which may be substituted;
  • R 2 is a carbocyclic group which may be substituted, a heterocyclic group which may be substituted, an aryl group which may be substituted, or a heteroaryl group which may be substituted;
  • R 3 -R 7 and R 15 are independently hydrogen, halogen, amino group which may be substituted, alkoxy group which may be substituted, C 1 -C 10 alkyl group which may be substituted, haloalkyl group, alkenyl group, alkynyl group, Hydroxyalkyl, aminoalkyl, carboxyalkyl, nitro, cyano, amido, hydroxy, decyl, acyloxy, azide, carboxyl, ethylenedioxy, hydroxyamido or may be substituted Alkylthio group.
  • A is N.
  • R 1 and R 2 are aryl groups which may be substituted.
  • R 3 -R 7 are each independently hydrogen, halogen or C 1 -C 6 alkyl.
  • R 15 is hydrogen or C 1 -C 6 alkyl.
  • the substituent on R 1 is any one, any two or any three selected from the group consisting of halogen, C 1 -C 6 alkyl, C 1 -C 6 Alkoxy and halogenated C 1 -C 6 alkyl.
  • R 1 is selected from the group consisting of: hydrogen, C 1 -C 8 alkyl, C 2 -C 8 alkenyl, C 3 -C 8 cycloalkyl, heteroaryl, and optionally An aryl group substituted with 1 to 4 substituents selected from the group consisting of halogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxy and halogenated C 1 -C 6 alkyl.
  • R 1 is selected from the group consisting of 1-4, optionally selected from halogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, and halo C 1 -C 6 a phenyl group substituted with a substituent of an alkyl group; in certain embodiments, the number of substituents is two; in certain embodiments, at least one substituent is in the ortho position; in certain embodiments, at least one substitution
  • the base is a halogen; in certain embodiments, the substituent on the phenyl group is two, both of which are in the ortho position, and at least one of which is a halogen.
  • R 1 is selected from pyridyl, pyrimidinyl, thienyl, furanyl, pyrrolyl, and imidazolyl groups which may be substituted.
  • R 1 is selected from the group consisting of hydrogen, C 1 -C 8 alkyl, C 3 -C 8 cycloalkyl, and C 2 -C 8 alkenyl which may be substituted.
  • the substituent of R 2 is any one, any two, any three or any four selected from the group consisting of C 1 -C 6 alkyl groups which may be substituted, a C 1 -C 6 acyl group which may be substituted, a heterocyclic group which may be substituted, a halogen, an oxy group which may be substituted, a nitro group and a C 1 -C 6 alkylamino group which may be substituted; preferably, these may be
  • the substituent on the substituted group may be from 1 to 4 groups selected from C 1 -C 6 alkyl, C 1 -C 6 acyl, optionally 1-4 C 1 -C 6 alkane a substituted heterocyclic group, a halogen, -NR a R b and a hydroxy group, wherein R a and R b are each independently H and a C 1 -C 6 alkyl group; preferably, the heterocyclic group is selected from piperazinyl
  • the substituent of R 2 is any one, any two, any three or any four selected from the group consisting of: a piperazinyl group which may be substituted, which may be substituted Piperazinyl-C 1 -C 4 alkyl, piperidinyl which may be substituted, imidazolyl, 1,4-diazepanyl which may be substituted, C 1 -C 6 alkyl, C 1 - C 6 acyl group, morpholinyl group which may be substituted, morpholinyl-C 1 -C 4 alkyl group, halogen, halogenated C 1 -C 6 alkyl group, C 1 -C 6 alkoxy group which may be substituted, Substituted hydroxy C 1 -C 6 alkyl, substituted C 1 -C 6 alkyl, substituted piperidinylamino, C 1 -C 6 alkylamino which may be substituted, may be substituted Heterocycl
  • the piperazine group which may be substituted is a piperazinyl group which may be substituted with 1, 2 or 3 groups selected from C 1 -C 6 alkyl, hydroxy C. 1- C 6 alkyl and C 1- C 6 acyl.
  • the piperazinyl group has at least one substituent at the para position, optionally having one or two substituents at the meta position.
  • the substitutable piperidinyl group is a piperidinyl group which may be substituted with one group selected from a C 1 -C 6 alkyl group and a C 1 -C 6 alkylamino group. .
  • the morpholinyl group which may be substituted is a morpholinyl group which may be substituted with 1 or 2 groups selected from a C 1 -C 6 alkyl group.
  • R 2 is selected from the group consisting of an optionally substituted phenyl, pyridyl, pyrazinyl, tetrahydroisoquinolinyl, and 2',3'-dihydro-1'H-spiro ( Cyclopropane-1,4'-isoquinolinyl-7'-yl and 4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-2-yl.
  • R 2 is selected from the group consisting of phenyl substituted with an optionally substituted piperazinyl group, phenyl substituted with an optionally substituted piperidinyl group, and optionally 1-3 C 1 -C 6 alkyl or halogen substituted tetrahydroisoquinolinyl.
  • the piperazinyl is optionally substituted with from 1 to 3 C 1 -C 6 alkyl, hydroxy C 1 -C 6 alkyl and C 1 -C 6 acyl groups. Substituted.
  • the piperidinyl group is optionally substituted with one substituent selected from the group consisting of C 1 -C 6 alkyl and C 1 -C 6 alkylamino.
  • R 4 and R 5 are each independently selected from H, C 1 -C 6 alkyl and halogen, preferably all H.
  • R 6 and R 7 are each independently selected from the group consisting of H, C 1 -C 6 alkyl and halogen, preferably all H.
  • One group of preferred compounds of the invention is represented by a compound of formula II or a pharmaceutically acceptable salt or prodrug thereof:
  • R 3 - R 7 are as described in formula I;
  • Ar 1 is hydrogen, a C 1 -C 8 alkyl group which may be substituted, a C 2 -C 8 alkenyl group which may be substituted, a C 3 -C 8 cycloalkyl group which may be substituted, an aryl group which may be substituted, a substituted heterocyclic group or a heteroaryl group which may be substituted;
  • Ar 2 is a carbocyclic group which may be substituted, a heterocyclic group which may be substituted, an aryl group which may be substituted, or a heteroaryl group which may be substituted;
  • R 3 -R 7 are hydrogen.
  • Ar 1 and Ar 2 are aryl groups which may be substituted, and more preferred are phenyl groups which may be substituted.
  • the substituent on Ar 1 is any one, any two or any three selected from the group consisting of halogen, C 1 -C 6 alkyl, C 1 -C 6 Alkoxy and halo C 1 -C 6 alkyl; in certain embodiments, the number of substituents is 2; in certain embodiments, at least one substituent is in the ortho position; in certain embodiments At least one substituent is a halogen; in certain embodiments, the substituent on the phenyl group is two, two are in the ortho position, and at least one of them is a halogen.
  • Ar 1 is selected from: a heteroaryl group which may be substituted, and optionally 1-4 selected from halogen, C 1 -C 6 alkyl, C 1 -C 6 alkane An aryl group substituted with a substituent of an oxy group and a halogenated C 1 -C 6 alkyl group.
  • Ar 1 is selected from the group consisting of 1-4 selected from the group consisting of halogen, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, and halogenated C 1 -C 6 A phenyl group substituted with a substituent of an alkyl group.
  • the number of substituents is two; more preferably, at least one substituent is in the ortho position; more preferably, at least one substituent is a halogen.
  • Ar 1 is selected from pyridyl, pyrimidinyl, thienyl, furanyl, pyrrolyl and imidazolyl groups which may be substituted.
  • the substituent of Ar 2 is any one, any two, any three or any four selected from the group consisting of C 1 -C 6 alkyl groups which may be substituted, a C 1 -C 6 acyl group which may be substituted, a heterocyclic group which may be substituted, a halogen, an oxy group which may be substituted, a nitro group and a C 1 -C 6 alkylamino group which may be substituted; preferably, these may be
  • the substituent on the substituted group may be from 1 to 4 groups selected from C 1 -C 6 alkyl, C 1 -C 6 acyl, optionally 1-4 C 1 -C 6 alkane a substituted heterocyclic group, a halogen, -NR a R b and a hydroxy group, wherein R a and R b are each independently H and a C 1 -C 6 alkyl group; preferably, the heterocyclic group is selected from piperazinyl
  • the substituent of Ar 2 is any one, any two, any three or any four selected from the group consisting of: a piperazinyl group which may be substituted, which may be substituted Piperazinyl-C 1 -C 4 alkyl, piperidinyl which may be substituted, imidazolyl, 1,4-diazepanyl which may be substituted, C 1 -C 6 alkyl, C 1 - C 6 acyl group, morpholinyl group which may be substituted, morpholinyl-C 1 -C 4 alkyl group, halogen, halogenated C 1 -C 6 alkyl group, C 1 -C 6 alkoxy group which may be substituted, Substituted hydroxy C 1 -C 6 alkyl, substituted C 1 -C 6 alkyl, substituted piperidinylamino, C 1 -C 6 alkylamino which may be substituted, may be substituted Heterocycl
  • the piperazine group which may be substituted is a piperazinyl group which may be substituted with 1, 2 or 3 groups selected from C 1 -C 6 alkyl, hydroxy C. 1 -C 6 alkyl and C 1 -C 6 acyl.
  • the piperazinyl group has at least one substituent at the para position, optionally having one or two substituents at the meta position.
  • the substitutable piperidinyl group is a piperidinyl group which may be substituted with one group selected from a C 1 -C 6 alkyl group and a C 1 -C 6 alkylamino group. .
  • the morpholinyl group which may be substituted is a morpholinyl group which may be substituted with 1 or 2 groups selected from C 1 -C 6 alkyl groups.
  • Ar 2 is selected from the group consisting of an optionally substituted phenyl, pyridyl, pyrazinyl, tetrahydroisoquinolinyl, and 2',3'-dihydro-1'H-spiro ( Cyclopropane-1,4'-isoquinolinyl-7'-yl and 4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin-2-yl.
  • Ar 2 is selected from the group consisting of a phenyl group substituted with an optionally substituted piperazinyl group, a phenyl group substituted with an optionally substituted piperidinyl group, and optionally 1-3 C 1 -C 6 alkyl or halogen substituted tetrahydroisoquinolinyl.
  • the piperazinyl is optionally substituted with from 1 to 3 C 1 -C 6 alkyl, hydroxy C 1 -C 6 alkyl and C 1 -C 6 acyl groups. Substituted.
  • the piperidinyl group is optionally substituted with one substituent selected from the group consisting of C 1 -C 6 alkyl and C 1 -C 6 alkylamino.
  • R 4 and R 5 are each independently selected from the group consisting of H, C 1 -C 6 alkyl and halogen, preferably all H.
  • R 6 and R 7 are each independently selected from the group consisting of H, C 1 -C 6 alkyl and halogen, preferably all H.
  • R 1 or Ar 1 is selected from the group consisting of: H; C 1 -C 6 alkyl; C 2 -C 6 alkenyl; C 3 -C 8 cycloalkyl; thiazolyl; furanyl ; pyrrolyl; imidazolyl; pyrimidinyl; pyridyl; and optionally 1 or 2 selected from halogen, C 1 -C 4 alkyl, halogenated C 1 -C 4 alkyl and C 1 -C 4 alkoxy A substituted phenyl group.
  • R 1 is or Ar 1 is selected from:
  • R 2 or Ar 2 is selected from the group consisting of
  • R 8 is independently hydrogen, halogen, C 1 -C 4 alkyl and halogenated C 1 -C 4 alkyl, preferably hydrogen, fluorine, chlorine, bromine, methyl and trifluoromethyl
  • R 9 is independently Halogen, C 1 -C 4 alkyl and halo C 1 -C 4 alkyl, preferably fluoro, chloro, methyl and trifluoromethyl
  • R 10 is independently hydrogen, halogen, C 1 -C 4 alkyl and C 1 -C 4 alkoxy, preferably hydrogen, fluorine, chlorine, methyl and methoxy
  • R 11 is independently hydrogen, halogen and C 1 -C 4 alkyl, preferably hydrogen, fluorine, chlorine, bromine and Methyl
  • R 12 is independently halogen and C 1 -C 4 alkoxy, preferably chloro and methoxy
  • R 13 is independently hydrogen, halogen, C 1 -C 4 alkyl, C 1 -C 4 alkoxy And a
  • the compound of Formula II has the structure of Formula III:
  • Ar 1 is selected from the group consisting of: phenyl substituted by 1 or 2 substituents selected from the group consisting of halogen, halogenated C 1 -C 6 alkyl, C 1 -C 6 alkoxy and C 1 -C 6 alkyl;
  • Ar 2 is selected from:
  • a substituted phenyl group the substituent of which is selected from the group consisting of: halogen; nitro; C 1 -C 6 alkyl, optionally substituted with 1 piperazinyl or 1 morpholinyl group, optionally substituted with 1-3 hydroxy groups, Optionally substituted with 1-5 halo, or optionally substituted by -NR a R b , the piperazinyl is optionally substituted with 1-3 substituents selected from C 1 -C 4 alkyl; oxy, any Alternatively, it is substituted by a C 1 -C 6 alkyl group or a piperidinyl group, which is optionally substituted with from 1 to 3 substituents selected from a C 1 -C 4 alkyl group; an amino group having 1 hydrogen is piperidinyl Substituted, another hydrogen is unsubstituted or substituted by C 1 -C 4 alkyl, or 1 or 2 hydrogen thereof is substituted by C 1 -C 6 alkyl, which is optional
  • a tetrahydroisoquinolinyl group optionally substituted with from 1 to 3 substituents selected from C 1 -C 6 alkyl and halogen;
  • a pyridyl group optionally substituted with one piperazinyl group optionally substituted by 1 to 3 C 1 -C 6 alkyl groups;
  • a pyrazinyl group optionally substituted with one piperazinyl group optionally substituted by 1 to 3 C 1 -C 6 alkyl groups;
  • R a and R b are each independently H or a C 1 -C 6 alkyl group.
  • Ar 1 is selected from the group consisting of: a phenyl group substituted with two substituents selected from a halogen and a C 1 -C 6 alkyl group;
  • Ar 2 is selected from the group consisting of: substituted phenyl, the substituent of which is selected from the group consisting of: halogen, halogenated C 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, hydroxy C 1 -C 6 alkyl, NR a R b -C 1 -C 6 alkyl, optionally substituted with 1-3 C 1 -C 4 alkyl-substituted piperazinyl group substituted with a C 1 -C 4 alkyl, optionally substituted 1 - 3 C 1 -C 4 alkyl-substituted piperidinyl-O-, NR a R b -C 1 -C 6 alkoxy, NR a R b -C 1 -C 6 alkyl-NR a -, is 1-3 substituents selected from C 1 -C 6 alkyl, hydroxy-substituted C 1 -C 6 alky
  • Ar 1 is two meta positions selected from the group consisting of halogen, C 1 -C 3 alkyl, halogenated C 1 -C 3 alkyl, and C 1 -C 3 , respectively.
  • a disubstituted phenyl group substituted with a substituent of an alkoxy group preferably, at least one of the two substituents is a halogen
  • Ar 2 is a phenyl group substituted by 1, 2 or 3 substituents selected from the group consisting of : halogen, halogenated C 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, hydroxy C 1 -C 6 alkyl, NR a R b -C 1 -C 6 alkane group, optionally substituted with 1-3 C 1 -C 4 alkyl-substituted piperazinyl group substituted with a C 1 -C 4 alkyl, optionally substituted with 1-3 C 1 -C 4 alkyl-piperidinyl -O-, NR a R b -C 1 -C 6 alkoxy, NR a R b -C 1 -C 6 alkyl group
  • Ar 1 is preferably:
  • Ar 1 is more preferably:
  • Ar 2 is preferably:
  • R 8 is independently hydrogen, fluorine, chlorine, bromine, methyl and trifluoromethyl
  • R 9 is independently hydrogen, fluorine, chlorine, bromine, methyl and trifluoromethyl
  • R 10 is independently hydrogen, fluorine, Chlorine, bromine, methyl and methoxy
  • R 11 is independently hydrogen, fluorine, chlorine, bromine, methyl and trifluoromethyl
  • R 12 is independently chlorine, fluorine, bromine, methyl and methoxy
  • R 13 Independently hydrogen, chloro, methyl, methoxy and hydroxymethyl
  • R 14 is independently hydrogen, chloro, bromo and methyl
  • Ar 2 is more preferably:
  • R 8 is independently hydrogen, fluorine, chlorine, bromine and methyl
  • R 9 is independently hydrogen, chlorine and methyl
  • R 10 is independently hydrogen, chlorine, methyl and methoxy
  • R 11 is independently hydrogen, fluorine , chloro, bromo and methyl
  • R 12 is independently hydrogen, chloro, methyl and methoxy
  • R 14 is independently hydrogen, chloro and methyl.
  • the compound does not include the compound of Example 83; in certain embodiments, the compound of Formula I does not include R 1 in the ortho position as two methyl or two alkyl groups.
  • R 1 in the ortho position as two methyl or two alkyl groups.
  • preferred compounds of Formula I include, but are not limited to:
  • alkyl refers to an alkyl group itself or a group having up to ten carbon atoms in a straight or branched chain.
  • Useful alkyl groups include straight or branched C 1 -C 10 alkyl groups, preferably C 1 -C 6 alkyl groups.
  • the alkyl group is a C 1 -C 4 alkyl group.
  • Typical C 1 -C 10 alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl, 3-pentyl, hexyl and octyl groups which may be optionally substituted.
  • alkenyl means a straight or branched chain containing from 2 to 10 carbon atoms unless the carbon chain length is additionally limited, wherein at least one of the two carbon atoms in the chain contains a double bond; C 2 -C 6 alkenyl.
  • Typical alkenyl groups include ethenyl, 1-propenyl, 2-propenyl, 2-methyl-1-propenyl, 1-butenyl and 2-butenyl.
  • alkynyl means a straight or branched chain containing from 2 to 10 carbon atoms unless the carbon chain length is additionally limited, wherein at least one of the two carbon atoms in the chain contains a hydrazone linkage; C 2 -C 6 alkynyl.
  • Typical alkynyl groups include ethynyl, 1-propynyl, 1-methyl-2-propynyl, 2-propynyl, 1-butynyl and 2-butynyl.
  • Useful alkoxy groups include those substituted by the above C 1 -C 10 alkyl group, preferably C 1 -C 6 alkyl or C 1 -C 4 alkyl group, such as methoxy, ethoxy, and the like.
  • the alkyl group in the alkoxy group may be optionally substituted.
  • Substituents for alkoxy groups include, but are not limited to, halogen, morpholinyl, amino groups including alkylamino and dialkylamino groups and carboxyl groups (including ester groups thereof).
  • Useful alkylthio groups include a thio group substituted by the above C 1 -C 10 alkyl group, preferably a C 1 -C 6 alkyl group, and the alkyl group in the alkylthio group may be optionally substituted. Also included are such alkylthio sulfoxides and sulfones.
  • Useful amino groups and optionally substituted amino groups include -NH 2 , -NHR' and -NR'R", wherein R' and R" are C 1 -C 10 alkyl groups which may be optionally substituted, cycloalkyl, aryl Base, heteroaryl or amino.
  • R' and R" together with N form a 5-8 membered heterocycle such as piperidine, or R' and R" and N together with other N or O form a 5-8 membered heterocycle such as piperazine.
  • the alkyl group and the heterocyclic ring may be optionally substituted.
  • the arylalkenyl, arylalkynyl, heteroaryl and heteroarylalkyl groups may be substituted by one or more (e.g.
  • substituents selected from the group consisting of halogens, hydroxy groups , carboxy, amino, nitro, cyano, C 1 -C 6 acylamino, C 1 -C 6 acyloxy, C 1 -C 6 alkoxy, aryloxy, alkylthio, C 1 -C 6 Alkyl, C 1 -C 6 acyl, C 6 -C 10 aryl, C 3 -C 8 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 6 -C 10 aryl (C 2 -C 6 )alkenyl, C 6 -C 10 aryl(C 2 -C 6 )alkynyl, saturated and unsaturated heterocyclic or heteroaryl, methylenedioxy, C 1- C 6 haloalkyl, C 6 -C 10 aryl(C 1 -C 6 )alkyl, C 1 -C 6 hydroxy
  • an alkyl group, an alkoxy group, an alkylthio group, an alkenyl group, an alkynyl group, a cycloalkyl group, a carbonyl group, a carbocyclic ring, and a heterocyclic ring may be one or more (for example, 1 , 2, 3 or 4) substituents selected from the group consisting of halogen, hydroxy, carboxy, amino, nitro, cyano, C 1 -C 6 acylamino, C 1 -C 6 acyl, C 1 - C 6 acyloxy, C 1 -C 6 alkoxy, aryloxy, alkylthio, C 6 -C 10 aryl, C 3 -C 8 cycloalkyl, C 2 -C 6 alkenyl, C 2- C6 alkynyl, C 6 -C 10 aryl(C 2 -C 6 )alkenyl, C 6 -C 10 -C 10
  • aryl, arylalkyl, arylalkenyl, arylalkynyl, heteroaryl, and heteroarylalkyl can be one or more (eg, 1, 2, 3 or 4) substituents selected from the group consisting of halogen, methylene dioxy, C 1 -C 6 haloalkyl, C 6 -C 10 aryl, C 3 -C 8 cycloalkyl, C 1- C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 6 -C 10 aryl(C 1 -C 6 )alkyl, C 6 -C 10 aryl (C 2 -C 6 )alkenyl, C 6 -C 10 aryl(C 2 -C 6 )alkynyl, C 1 -C 6 hydroxyalkyl, nitro, amino, ureido, cyano, C 1 -C 6 acyla
  • the substituent is a heterocyclic group, an aryl group or a heteroaryl group
  • the number of the heterocyclic group, aryl or heteroaryl substituent is usually one.
  • aryl refers to a aryl group or as part of another group, and refers to a monocyclic, bicyclic or tricyclic aromatic group containing from 6 to 14 carbon atoms.
  • Useful aryl groups include C 6 -C 14 aryl groups, preferably C 6 -C 10 aryl groups.
  • Typical C 6 -C 14 aryl groups include phenyl, naphthyl, phenanthryl, anthryl, fluorenyl, fluorenyl, biphenyl, biphenylene and anthracenyl.
  • the "carbocyclic ring" referred to herein includes a cycloalkyl group and a partially saturated carbocyclic group.
  • Useful cycloalkyl groups are C 3 -C 8 cycloalkyl groups.
  • Typical cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • Useful saturated or partially saturated carbocyclic groups are the cycloalkyl and cycloalkenyl groups described above, such as cyclopentenyl, cycloheptenyl and cyclooctenyl.
  • Useful halogen or halogen groups include fluorine, chlorine, bromine and iodine.
  • Arylalkyl include any of the above C 6 -C 14 aryl-substituted C 1 -C 10 alkyl group used herein. Preferred arylalkyl groups are benzyl, phenethyl or naphthylmethyl.
  • arylalkenyl as used above includes any of a C 6 -C 14 aryl-substituted C 2 -C 10 alkenyl group.
  • arylalkynyl as used above includes any of a C 6 -C 14 aryl-substituted C 2 -C 10 alkynyl group.
  • aryloxy includes an oxy group substituted by any of the above C 6 -C 14 aryl groups, the aryl group of which may be optionally substituted.
  • Useful aryloxy groups include phenoxy and 4-methylphenoxy.
  • aryl alkoxy group includes C is any one of the above-described aryl group substituted with 1 -C 10 alkoxy group, an aryl group which may be optionally substituted.
  • Useful arylalkoxy groups include benzyloxy and phenylethoxy.
  • Useful haloalkyl groups include C 1 -C 10 alkyl groups substituted by one or more fluorine, chlorine, bromine or iodine atoms, preferably C 1 -C 6 alkyl groups, such as fluoromethyl, difluoromethyl, trifluoromethyl Base, pentafluoroethyl, 1,1-difluoroethyl, chloromethyl, chlorofluoromethyl and trichloromethyl.
  • Useful acylamino is any C 1 -C 6 acyl (alkanoyl) attached to an amino nitrogen, such as acetylamino, chloroacetamido, propionylamino, butyrylamino, pentanoylamino and acylamino And an aryl-substituted C 1 -C 6 acylamino group such as a benzoylamino group and a pentafluorobenzoylamino group.
  • Useful acyl groups include C 1 -C 6 acyl groups such as acetyl.
  • acyloxy groups are linked to the oxygen (-O-) any C 1- C 6 acyl (alkanoyl) such as formyloxy, acetoxy, propionyloxy, butyryloxy, valeryl Oxyl and hexanoyloxy.
  • heterocyclic ring refers to a saturated or partially saturated 3-7 membered monocyclic ring, or a 7-10 membered bicyclic ring system, which is selected from carbon atoms and from 1-, O, and S. It is composed of heteroatoms in which the hetero atom nitrogen and sulfur can be arbitrarily oxidized, and the nitrogen can also be arbitrarily quaternized, and includes the fusion of any heterocyclic ring defined above in the bicyclic ring system with the benzene ring. If the resulting compound is stable, then the carbon or nitrogen atom of the heterocycle can be substituted.
  • Useful saturated or partially saturated heterocyclic groups include tetrahydrofuranyl, pyranyl, piperidinyl, piperazinyl, 1,4-diazepanyl, pyrrolidinyl, imidazolidinyl, imidazolinyl, Dihydroindenyl, isoindoline, quinuclidinyl, morpholinyl, heterochroman, chromanyl, pyrazolidinyl, pyrazolinyl, tetrahydroisoquinolinyl, 2' , 3'-dihydro-1'H-spiro(cyclopropane-1,4'-isoquinoline)-7'-yl, 4,5,6,7-tetrahydropyrazolo[1,5-a Pyrazin-2-yl, tetronoyl and tetramoyl, these groups may be optionally substituted.
  • heteromatic ring means having 5 to 14 ring atoms and having 6, 10 or 14 ⁇ electrons are shared on the ring system. Further, the ring atom contained is a carbon atom and optionally 1-3 hetero atoms from oxygen, nitrogen, and sulfur.
  • Useful heteroaryl groups include thienyl (phenylthio), benzo[d]isothiazol-3-yl, benzo[b]thienyl, naphtho[2,3-b]thienyl, thiazide, Furanyl, pyranyl, isobenzofuranyl, chromenyl, anthranilyl, phenoxanthiinyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl (including but not limited to 2-pyridine , 3-pyridyl and 4-pyridyl), pyrazinyl, pyrimidinyl, pyridazinyl, pyridazinyl, isodecyl, 3H-indenyl, fluorenyl, oxazolyl, fluorenyl, 4H-quinazinyl, isoquinolyl, quinolyl, pyridazinyl, naphthyridin
  • such a nitrogen atom may be in the form of an N-oxide such as a pyridyl N-oxide, a pyrazinyl N-oxide, and a pyrimidinyl N-oxide.
  • heteroaryloxy includes oxy groups substituted by any of the above heteroaryl groups, wherein the heteroaryl group may have a substituent.
  • Useful heteroaryloxy groups include pyridyloxy, pyrazinyloxy, pyrrolyloxy, pyrazolyloxy, imidazolyloxy and phenylthiooxy.
  • heteroarylalkoxy refers to any of the above C1-10 alkoxy groups substituted by any of the above heteroaryl groups, wherein the heteroaryl group may have a substituent.
  • stereoisomers including optical isomers.
  • the present invention includes all stereoisomers and racemic mixtures of such stereoisomers, as well as the individual enantiomers which can be separated according to methods well known to those skilled in the art.
  • Examples of pharmaceutically acceptable salts include inorganic and organic acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, citrates, lactates, tartrates, maleates, fumarates, Mandelic acid salts and oxalates; and inorganic and organic base salts formed with bases such as sodium hydroxy, tris(hydroxymethyl)aminomethane (TRIS, tromethamine) and N-methylglucamine.
  • inorganic and organic acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, citrates, lactates, tartrates, maleates, fumarates, Mandelic acid salts and oxalates
  • inorganic and organic base salts formed with bases such as sodium hydroxy, tris(hydroxymethyl)aminomethane (TRIS, tromethamine) and N-methylglucamine.
  • prodrugs of the compounds of the invention include simple esters of carboxylic acid containing compounds (e.g., esters obtained by condensation with a C 1-4 alcohol according to methods known in the art); esters of hydroxy containing compounds (e.g., according to the present invention)
  • a method known in the art is an ester obtained by condensation with a C 1-4 carboxylic acid, a C 3-6 diacid or an anhydride thereof such as succinic anhydride and fumaric anhydride; an imine of an amino group-containing compound (for example, according to the art) Method of imine obtained by condensation with a C 1-4 aldehyde or ketone); carbamate of an amino group-containing compound, such as Leu et al. (J. Med.
  • the compounds of the invention can be prepared using methods known to those skilled in the art or by the novel methods of the invention.
  • the compounds of the invention having formula I can be prepared as shown in the reaction examples in Reaction Scheme 1.
  • N-tert-butoxycarbonyl-1,2-ethanediamine and substituted phenyl isocyanate, such as o-chlorophenyl isocyanate, are reacted in diethyl ether at room temperature to give the product (2-(3-(2-chlorophenyl) Ureido)ethyl)carbamic acid tert-butyl ester.
  • m-chloroperoxybenzoic acid in chloroform at room temperature to give the product 6-(2-chlorophenyl)-2-(methylsulfonyl)-8,9-dihydroimidazo[1,2-a]pyrimidine [5,4-e]pyrimidine-5(6H)-one.
  • the ketone and 4-(4-methylpiperazine)phenylamine are heated in isopropanol to give the title compound 6-(2-chlorophenyl)-2-((4-(4-methylpiperazine-1). -yl)phenyl)amino)-8,9-dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-one.
  • the target compound 6-(2-chloro-6-fluorophenyl)-2-((4-(4) can be obtained by substituting 1-chloro-3-fluoro-2-isocyanatophenyl ester for o-chlorophenyl isocyanate.
  • the title compound 6-(2-chloro-6-fluorophenyl)-2- can be obtained by substituting 4-(4-isopropylpiperazin-1-yl)aniline for 4-(4-methylpiperazine)aniline.
  • the target compound 6-(2-chloro-6-fluoro) can be obtained by substituting 2-methyl-1,2,3,4-tetrahydroisoquinolin-7-amine for 4-(4-methylpiperazine) aniline.
  • the target compound 6-(2-chloro-6- can be obtained by substituting 1-(4-aminophenyl)-N,N-dimethylpiperidin-4-amine for 4-(4-methylpiperazine)aniline.
  • Fluorophenyl -2-((4-(4-dimethylamino)piperidin-1-yl)phenyl)amino)-8,9-dihydroimidazo[1,2-a]pyrimido[5 , 4-e]pyrimidine-5(6H)-one.
  • the compound of the present invention can be produced as shown in the reaction examples in Reaction Scheme 2.
  • 1-(2-Aminoethyl)-3-(2-chlorophenyl)urea and methyl 4,6-dichloronicotinate, diisopropylethylamine are mixed and heated in acetonitrile to give the product 6- Chloro-4-((2-(3-(2-chlorophenyl)ureido)ethyl)amino)nicotinate methyl ester.
  • Methyl 6-chloro-4-((2-(3-(2-chlorophenyl)ureido)ethyl)amino)nicotinate is heated in phosphorus oxychloride to give the product 8-chloro-4-( 2-Chlorophenyl)-1,2-dihydroimidazo[1,2-a]pyrido[3,4-e]pyrimidin-5(4H)-one.
  • the compound of the present invention can be produced as shown in the reaction examples in Reaction Scheme 3.
  • 2-(Methanesulfonyl)-6-(2,6-dichlorophenyl)-8,9-dihydroimidazo[1,2-a]pyrimidine can be obtained by a method similar to that shown in Reaction Scheme 1. [5,4-e]pyrimidine-5(6H)-one.
  • the target compound 2- can be obtained by substituting 4-((3R,5S)-3,4,5-trimethylpiperazin-1-yl)aniline for 4-(4-methylpiperazin-1-yl)aniline ((4-((3R,5S)-3,4,5-Trimethylpiperazin-1-yl)phenyl)amino)-6-(2,6-dichlorophenyl)-8,9- Dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-one.
  • the target compound 6-(2) can be obtained by substituting 1-(4-aminophenyl)-N,N-dimethylpiperidin-4-amine for 4-(4-methylpiperazin-1-yl)aniline. 6-Dichlorophenyl)-2-((4-(4-(dimethylamino)piperidin-1-yl)phenyl)amino)-8,9-dihydroimidazo[1,2-a] Pyrimido[5,4-e]pyrimidine-5(6H)-one.
  • Formula I is a kinase inhibitor, particularly a Wee1 kinase inhibitor. Therefore, these compounds are useful for the treatment of Wee1-related diseases, such as cancer.
  • the invention also encompasses methods of treating an animal with an effective amount of a compound of Formula I, Formula II or Formula III, or a pharmaceutically acceptable salt or prodrug thereof.
  • the method of treatment is for the treatment of a kinase-related disease, in particular a Wee1 kinase-related disease, such as cancer.
  • Such diseases which can be treated or prevented by the methods or pharmaceutical compositions of the invention include, but are not limited to, liver cancer, melanoma, Hodgkin's disease, non-Hodgkin's lymphoma, acute lymphoblastic leukemia, chronic lymphoid leukemia, multiple myeloma , neuroblastoma, breast cancer, ovarian cancer, lung cancer, Wilms tumor, cervical cancer, testicular cancer, soft tissue sarcoma, primary macroglobulinemia, bladder cancer, chronic myeloid leukemia, primary brain Cancer, malignant melanoma, small cell lung cancer, gastric cancer, colon cancer, malignant pancreatic islet tumor, malignant carcinoid cancer, choriocarcinoma, mycosis fungoides, head and neck cancer, osteogenic sarcoma, pancreatic cancer, acute granulocytes Leukemia, hairy cell leukemia, rhabdomyosarcoma, Kaposi's sarcoma,
  • the invention also encompasses the use of other diseases, such as neurological or neuropsychiatric diseases or conditions, such as patients with depression, for the treatment or prevention of abnormalities in the activity of kinases, particularly Wee1.
  • an effective amount of the pharmaceutical formulation is administered to a patient having one or more of these symptoms.
  • the pharmaceutical preparations contain a therapeutically effective concentration of a compound of Formula I, Formula II or Formula III formulated for oral, intravenous, topical or topical administration for the treatment of cancer and other diseases.
  • the amount administered is an amount effective to ameliorate or eliminate one or more conditions.
  • an effective amount is an amount sufficient to ameliorate or in some way alleviate the symptoms associated with the disease.
  • Such doses can be administered as a single dose or can be administered according to an effective therapeutic regimen.
  • the amount administered may cure the disease, but administration is usually to improve the symptoms of the disease. Repeated administration is generally required to achieve the desired improvement in symptoms.
  • a pharmaceutical composition comprising a compound of Formula I, Formula II or Formula III comprising a kinase inhibitor, or a pharmaceutically acceptable salt thereof, in association with a pharmaceutically acceptable carrier.
  • Another embodiment of the invention relates to a pharmaceutical composition effective for treating cancer, wherein a compound of formula I, formula II or formula III comprising a kinase inhibitor, or a pharmaceutically acceptable salt or prodrug thereof, and at least one known An anticancer drug or a pharmaceutically acceptable salt of an anticancer drug is used in combination.
  • a compound of formula I, formula II or formula III comprising a kinase inhibitor, or a pharmaceutically acceptable salt or prodrug thereof, and at least one known An anticancer drug or a pharmaceutically acceptable salt of an anticancer drug is used in combination.
  • it is combined with other anticancer drugs related to DNA damage and repair mechanisms, including the PARP inhibitors Olapani, Niraparib, Rucaparib and Talazoparib; HDAC inhibitors vorinostat, romidepsin, Pabis He and Baileys; and so on.
  • anticancer drugs associated with cell division detection sites including Chk1/2 inhibitors, CDK4/6 inhibitors such as Pabsini, ATM/ATR inhibitors, and the like.
  • Other known anticancer drugs useful in combination therapy for cancer include, but are not limited to, alkylating agents such as busulfan, melphalan, chlorambucil, cyclophosphamide, ifosfamide, temozolomide, bendamustine, Cisplatin, mitomycin C, bleomycin and carboplatin; topoisomerase I inhibitors such as camptothecin, irinotecan and topotecan; topoisomerase II inhibitors such as doxorubicin, Epirubicin, aclarithromycin, mitoxantrone, methylhydroxy ellipticine and metoprolol; RNA/DNA antimetabolites such as 5-azacytidine, gemcitabine, 5-fluorouracil and methotrexate DNA; DNA antimetabolites such as 5-az
  • the compounds of the invention may be administered as a single pharmaceutical composition with at least one known anti-cancer drug.
  • the compounds of the invention may also be administered separately from at least one known anticancer drug.
  • the compound of the invention is administered at about the same time as at least one known anticancer drug, i.e., all drugs are administered simultaneously or sequentially, as long as the compound achieves a therapeutic concentration in the blood simultaneously.
  • the compound of the invention and at least one known anticancer drug are administered according to a respective dosage regimen as long as the compound reaches a therapeutic concentration in the blood.
  • Another embodiment of the present invention is a bioconjugate of the compound which is effective as a kinase inhibitor and which is effective for inhibiting tumors.
  • the bioconjugant capable of inhibiting tumors consists of the compound with at least one known medically active antibody, such as Herceptin or rituximab, or an auxin, such as DGF or NGF, or a cytokine, such as interleukin 2 Or 4, or any molecule that binds to the cell surface.
  • the antibody and other molecules are capable of delivering the compound to its target, making it an effective anticancer drug.
  • This bioconjugate can also increase the anticancer effect of medically active antibodies such as Herceptin or rituximab.
  • Another embodiment of the present invention relates to a pharmaceutical composition effective for inhibiting tumors, comprising a kinase inhibitor of Formula I, Formula II or Formula III, or a pharmaceutically acceptable salt or prodrug thereof, in combination with radiation therapy treatment.
  • the compounds of the invention may be administered at the same time or at different times with radiation therapy.
  • Another embodiment of the present invention relates to a pharmaceutical composition effective for post-operative treatment of cancer comprising a kinase inhibitor of Formula I, Formula II or Formula III, or a pharmaceutically acceptable salt or prodrug thereof.
  • the invention further relates to a method of treating a cancer in a mammal by surgically removing the tumor and then treating the mammal with a pharmaceutical composition of the invention.
  • the pharmaceutical composition of the present invention includes all of the pharmaceutical preparations of the present invention in an amount effective to achieve their intended purpose. While the needs of each individual are different, one skilled in the art can determine the optimal dosage for each portion of the pharmaceutical formulation.
  • the compound, or a pharmaceutically acceptable salt thereof is orally administered to a mammal daily for a dose of from about 0.0025 to 50 mg/kg body weight. Preferably, however, it is about 0.01 to 10 mg/kg administered orally per kilogram. If a known anticancer drug is also administered, the dose should be effective to achieve its intended purpose.
  • the optimal dosage of these known anticancer drugs is well known to those skilled in the art.
  • the unit oral dose can include from about 0.01 to 50 mg, preferably from about 0.1 to 10 mg, of the compound of the invention.
  • the unit dose may be administered one or more times per day in one or more tablets, each tablet containing from about 0.1 to 50 mg, conveniently from about 0.25 to 10 mg of the compound of the invention or a solvate thereof.
  • the concentration of the compound of the present invention may be about 0.01 to 100 mg per gram of the carrier.
  • the compounds of the invention can be administered as unprocessed pharmaceuticals.
  • the compounds of the invention may also be administered as part of a suitable pharmaceutical formulation containing a pharmaceutically acceptable carrier, including excipients and auxiliaries.
  • a pharmaceutically acceptable carrier including excipients and auxiliaries.
  • These pharmaceutically acceptable carriers facilitate the processing of the compounds into pharmaceutically acceptable pharmaceutical preparations.
  • Preferred pharmaceutical preparations especially those of the oral and preferred modes of administration, such as tablets, troches and capsules, and solutions suitable for injection or oral administration, comprise from about 0.01% to about 99%, preferably from about 0.25%. Up to 75% of active compound and excipients.
  • non-toxic pharmaceutically acceptable salts of the compounds of the invention are non-toxic pharmaceutically acceptable salts of the compounds of the invention.
  • the acid addition salt is formed by mixing a non-toxic pharmaceutically acceptable acid solution and a solution of the compound of the present invention.
  • the acid is, for example, hydrochloric acid, fumaric acid, maleic acid, succinic acid, acetic acid, citric acid, tartaric acid, carbonic acid, phosphoric acid, oxalic acid and the like.
  • the base addition salt is formed by mixing a non-toxic pharmaceutically acceptable base solution and a solution of the compound of the present invention.
  • the base is, for example, sodium hydroxide, potassium hydroxide, hydrogen choline, sodium carbonate, trishydroxymethylaminomethane, N-methyl-glucosamine or the like.
  • the pharmaceutical preparation of the present invention can be administered to any mammal as long as they can obtain the therapeutic effect of the compound of the present invention.
  • the most important of these mammals are human and veterinary animals, although the invention is not intended to be so limited.
  • the pharmaceutical preparation of the present invention can be administered by any route to achieve its intended purpose.
  • it can be administered by parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, buccal, intrathecal, intracranial, nasal or topical routes.
  • parenteral subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, buccal, intrathecal, intracranial, nasal or topical routes.
  • it can be administered orally.
  • the dosage of the drug will be determined by the age of the patient, the health and weight, the type of concurrent treatment, the frequency of treatment, and the desired therapeutic benefit.
  • the pharmaceutical preparations of the invention can be made in a known manner. For example, it is manufactured by a conventional mixing, granulating, tableting, dissolving, or freeze drying process. In the manufacture of oral formulations, the mixture can be selectively milled by combining the solid adjuvant with the active compound. If necessary or necessary, after adding an appropriate amount of auxiliary agent, the mixture of particles is processed to obtain a tablet or tablet core.
  • Suitable excipients are, in particular, fillers, such as sugars such as lactose or sucrose, mannitol or sorbitol; cellulose preparations and/or calcium phosphates, such as tricalcium phosphate or calcium hydrogen phosphate; and binders, such as starch pastes, including Corn starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methylcellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone.
  • fillers such as sugars such as lactose or sucrose, mannitol or sorbitol
  • cellulose preparations and/or calcium phosphates such as tricalcium phosphate or calcium hydrogen phosphate
  • binders such as starch pastes, including Corn starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methylcellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or poly
  • a disintegrating agent such as the above-mentioned starch, and carboxymethyl starch, crosslinked polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate may be added.
  • Adjuvants are especially flow regulators and lubricants, for example, silica, talc, stearic acid or a salt thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol.
  • the tablet core can be provided with a suitable coating that is resistant to gastric juice. For this purpose, a concentrated sugar solution can be applied.
  • This solution may contain gum arabic, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, a lacquer solution and a suitable organic solvent or solvent mixture.
  • a suitable cellulose solution such as cellulose acetate phthalic acid or hydroxypropyl methylcellulose phthalic acid can be used.
  • a dye or pigment can be added to the coating of the tablet or tablet core. For example, a combination for identifying or for characterizing the dosage of an active ingredient.
  • Other orally available pharmaceutical preparations include pressure-bonded capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol.
  • the pressure-bonded capsules may contain the active compound in the form of granules mixed with a filler such as lactose; a binder such as starch; and/or a lubricant such as talc or magnesium stearate, and a stabilizer.
  • the active compound is preferably dissolved or suspended in a suitable liquid such as a fat or liquid paraffin, to which a stabilizer may be added.
  • Formulations suitable for parenteral administration include aqueous solutions of the active compounds, such as solutions of aqueous salts and basic solutions.
  • an oily injection suspension of the appropriate active compound may be employed.
  • Suitable lipophilic solvents or vehicles include oils such as sesame oil, synthetic fatty acid esters such as ethyl oleate or triglycerides or polyethylene glycol 400, or hydrogenated castor oil, or cyclodextrin.
  • Aqueous injection suspensions may contain materials which increase the viscosity of the suspension, such as sodium carboxymethylcellulose, sorbitol, and/or dextran. Suspension stabilizers may also be included.
  • the compounds of the invention are formulated for topical and parenteral use and for the treatment of skin cancer.
  • the external preparation of the present invention can be prepared into an oil, a cream, an emulsion, an ointment or the like by a preferably suitable carrier.
  • suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular alcohols (greater than C 12).
  • Preferred carriers are those in which the active ingredient is soluble.
  • Emulsifiers, stabilizers, humectants and antioxidants, as well as agents which impart color or aroma if desired, may also be included.
  • these external preparations may contain a transdermal penetration enhancer. Examples of such enhancers can be found in U.S. Patent Nos. 3,989,816 and 4,444,762.
  • the cream is preferably formulated with a mineral oil, a mixture of self-emulsifying beeswax and water, and mixed with an active ingredient dissolved in a small amount of oil such as almond oil.
  • a typical example of a cream includes about 40 parts water, 20 parts beeswax, 40 parts mineral oil and 1 part almond oil.
  • the ointment can be formulated by mixing a vegetable oil containing an active ingredient such as almond oil and warm soft paraffin, and then cooling the mixture.
  • An example of a typical ointment includes about 30% by weight almond oil and 70% by weight white soft paraffin.
  • the invention also relates to the use of a compound of the invention for the manufacture of a medicament for the treatment of a clinical condition which is effective in inhibiting the activity of a kinase, in particular Wee1.
  • These drugs may include the above pharmaceutical compositions.
  • the starting material was 1-chloro-3-fluoro-2-isocyanatophenyl ester, N-tert-butoxycarbonyl-1,2-ethanediamine, Ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate and 4-(4-methylpiperazine)phenylamine, the title compound 6-(2-chloro-6-fluorophenyl)- 2-((4-(4-Methylpiperazin-1-yl)phenyl)amino)-8,9-dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidine- 5(6H)-one (58 mg, 38% yield, white solid).
  • Example 6-8 The following compounds of Examples 6-8 were obtained in analogy to the synthesis of Example 1 described, starting from 2-(methylsulfonyl)-6-(2-chloro-6-fluorophenyl)- 8,9-dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-one (prepared by a synthesis method similar to that of Examples 1a-1e, starting materials) N-tert-butoxycarbonyl-1,2-ethanediamine, ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate and phenyl 1-chloro-3-fluoro-2-isocyanate And the corresponding substituted aniline or substituted tetrahydroisoquinolinamine.
  • Example 5 The compounds of the following Examples 5, 9-26 were prepared in analogy to the synthesis of Example 4b described, starting from 2-(methylsulfonyl)-6-substituent-8,9. - Dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-one (prepared using a synthetic procedure analogous to Examples 1a-1e, starting material N- Tert-Butoxycarbonyl-1,2-ethanediamine, ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate and the corresponding substituted phenyl isocyanate) and the corresponding substituted amines.
  • Examples 27 and 28 were prepared in analogy to the synthesis of Examples 1a-e and 4b described, starting from 1-chloro-3-fluoro-2-isocyanatophenyl, (1-amino) Tert-butyl 2-methylpropan-2-yl)carbamate or tert-butyl 2-amino-2-methylpropylcarbamate, 4-chloro-2-(methylthio)pyrimidine-5-carboxylic acid Ester and 4-(4-methylpiperazine) aniline.
  • Example 32-33, 36, 54-56 and 85-87 were prepared in analogy to the synthesis of Example 1 described, starting from 2-(methylsulfonyl)-6-substituent.
  • -8,9-dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-one prepared by a synthetic method similar to that of Examples 1a-1e, starting The starting materials are N-tert-butoxycarbonyl-1,2-ethanediamine, ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate and the corresponding substituted phenyl isocyanate) and the corresponding substituted aniline .
  • Example 102, 103 and 105-115 were prepared in a similar manner to the synthesis of Example 1 described, starting from 2-(methylsulfonyl)-6-substituent-8.
  • 9-Dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-one prepared by a synthesis method similar to that of Examples 1a-1e, starting material N - tert-Butoxycarbonyl-1,2-ethanediamine, ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate and the corresponding substituted phenyl isocyanate) and the corresponding substituted anilines.
  • Example 93-101, 104 and 116-184 were prepared in a similar manner to the synthesis of Example 4b described, starting from 2-(methylsulfonyl)-6-substituent- 8,9-dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-one (prepared by a synthesis method similar to that of Examples 1a-1e, starting materials) It is N-tert-butoxycarbonyl-1,2-ethanediamine, ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate and the corresponding substituted phenyl isocyanate) and the corresponding substituted amine.
  • Example 93 is a by-product of the synthesis of the target compound of Example 19.
  • N-acetyl-3,5-dimethylaniline 3,5-dimethylaniline (8 g, 66 mmol) was dissolved in anhydrous dichloromethane (120 mL) at room temperature, and triethylamine was added. (18 mL, 132 mmol). Acetic anhydride (8.09 g, 79 mmol) was added to the reaction mixture at 0 ° C, and the reaction mixture was stirred at room temperature for 2 hr. The mixture was separated and the aqueous extracted with dichloromethane (50 mL). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate The title compound (10 g, 94% yield, white solid) was obtained.
  • N-acetyl-3,5-dimethyl-4-bromoaniline N-acetyl-3,5-dimethylaniline (10 g, 61.3 mmol) was dissolved in dichloromethane at room temperature ( In a mixed solvent of 400 mL) and methanol (160 mL), the reaction mixture was stirred at room temperature for 1 hour, and then tetrabutylammonium tribromide (32.5 g, 67.4 mmol) was added. The reaction mixture was stirred at room temperature. The organic solvent was concentrated under reduced vacuo.EtOAcEtOAc m.
  • N-acetyl-3,5-dimethyl-4-(4-methylpiperazin-1-yl)aniline N-acetyl-3,5-dimethyl-4 at room temperature -bromoaniline (400 mg, 1.65 mmol), N-methylpiperazine (199 mg, 1.98 mmol), tris(dibenzylideneacetone)dipalladium (76 mg, 0.08 mmol) and 2-(di-tert-butylphosphine) Benzene (49 mg, 0.16 mmol) was added to dry THF (10 mL). The reaction mixture was microwaved at 100 ° C for 1 h and then cooled to room temperature. Filtration and removal of the organic solvent under reduced pressure gave a crude product. The title compound (288 mg, 66% yield, yellow solid). LC-MS (ESI): m / z (M + H) + 262.41.
  • substituted amines can be obtained by a synthetic method similar to the above intermediates 1) to 5), or can be synthesized by a person skilled in the art according to known methods.
  • Wet1 kinase (human) was added to a reaction solution containing 20 mM Tris/HCl pH 8.5, 0.2 mM EDTA, 500 ⁇ M MLLYHQGKFLQTFCGSPLYRRR, 10 mM magnesium acetate and 10 ⁇ M [ ⁇ - 33 P]-ATP, and then 50-fold concentration was dissolved in 100.
  • the reaction was initiated by the addition of a Mg/ATP mixture and after incubation for 40 minutes at room temperature, the phosphoric acid solution was added to a final concentration of 0.5% to quench the reaction.
  • Example 1 Dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-one (Example 1) and its analogs have a good inhibitory effect on Wee1 kinase enzyme activity.
  • 10 ⁇ M of the compounds of Example 1 and Example 2 inhibited the enzymatic activity of Wee1 kinase by 96% and 95%, respectively, and the activity was similar to that of the reference compound AZD1775.
  • Example 4 inhibited Wee1 kinase enzyme activity by 51% at 0.01 ⁇ M, indicating that Example 4 has high inhibitory activity against Wee1 kinase.
  • K d affinity dissociation constant
  • Wee 1 kinase-tagged T7 phage was prepared using E. coli BL21 strain. During the growth phase of Escherichia coli (exponential growth phase), T7 phage were infected and incubated at 32 ° C until the cells were lysed. The lysate was centrifuged and filtered to remove cell debris. Partial sequence of human Wee1 (corresponding to amino acid M291 of reference sequence NP_003381.1) K575) is expressed on the T7 phage coat. A short DNA sequence was inserted into the T7 phage genome as an amplicon for amplification detection by qPCR.
  • Streptavidin-coated magnetic beads (Dynal M280) were reacted with biotin-labeled small molecule ligands for 30 minutes at room temperature as an affinity resin for the binding assay.
  • the ligand-bound resin was blocked with excess biotin and washed extensively with blocking buffer (SeaBlock (Pierce), 1% BSA, 0.05% Tween 20, 1 mM DTT) to remove unbound ligand to reduce non-specific binding.
  • the binding reaction was carried out by mixing 16 ⁇ l of phage lysate, 3.8 ⁇ l of ligand-binding resin, and 0.18 ⁇ l of the test compound (PBS/0.05% Tween 20/10 mM DTT/0.1% BSA/2 ⁇ g/ml ultrasonically degraded salmon semen DNA).
  • the test compound was formulated into a 111x stock solution in 100% DMSO. K d values calculated from the three 11-fold serial dilutions of the compound and three DMSO blank solution. The highest concentration of the tested compound was 100,000 nM and the final concentration of DMSO was 0.9%. All reactions were performed in a 384-well polypropylene assay plate with a final volume of 0.02 mL per well.
  • the qPCR reaction was performed by adding 2.5 ⁇ L of the kinase-containing eluate to 7.5 ⁇ L of a qPCR reaction solution containing a 0.15 ⁇ M amplicon primer and a 0.15 ⁇ M amplicon detection probe.
  • the qPCR protocol included a 10 minute hot start at 95 °C followed by 35 rounds of 15 ° 95 ° C and 1 minute 60 ° C for 35 rounds of amplification. Each concentration was repeated twice in the experiment.
  • LoVo cells were trypsinized, centrifuged at 800 rpm for 5 min, the supernatant was discarded, resuspended in fresh medium, counted, and seeded at a density of 6000 cells per well to a 96-well cell culture plate and placed in a 37 ° C 5% CO 2 incubator. Cultivate overnight.
  • test substance including the test compound and reference compound AZD1775 mother liquor was serially diluted to 8 concentrations in DMSO at a ratio of 1:3 and 1:10 (the last concentration was DMSO negative control): 10 ⁇ M, 3.3 ⁇ M, 1 ⁇ M , 0.33 ⁇ M, 0.1 ⁇ M, 0.033 ⁇ M, 0.01 ⁇ M, 0 ⁇ M (final concentration of DMSO 1 ⁇ ).
  • DMSO negative control 10 ⁇ M, 3.3 ⁇ M, 1 ⁇ M , 0.33 ⁇ M, 0.1 ⁇ M, 0.033 ⁇ M, 0.01 ⁇ M, 0 ⁇ M (final concentration of DMSO 1 ⁇ ).
  • Add 5 ⁇ L of each concentration to 120 ⁇ L of Petri (25-fold dilution) and mix by shaking.
  • the cells were cultured overnight, the medium was removed, 195 ⁇ L of fresh medium was added to each well, and 5 ⁇ L of the diluted medium containing the corresponding concentration of the test substance was added, respectively, and then the plate was placed in a 37 ° C 5% CO 2 incubator. 3d.
  • the stock solution was removed, and 100 ⁇ L of fresh serum-free DMEM containing MTT (0.5 mg/mL) was added to each well, and the culture was continued. After 4 h, the stock solution was removed, 100 ⁇ L of DMSO was added to each well, and shaken for 10 min in the dark, and placed in a multi-function reader to read the absorbance (OD value) at a wavelength of 552/630/690 nm.
  • the data was analyzed by the software Graph Pad Prism 5.0, and the inhibitory activity of the compound on cell proliferation was plotted in terms of cell viability and compound concentration.
  • Table 3 summarizes the data inhibition (IC 50) of compounds on the growth of LoVo cells.
  • Example 1 2 3 4 5 6 IC 50 ( ⁇ m) 3.575 0.4249 >100 0.1691 0.4307 0.3393
  • Example 7 8 9 10 11 12 IC 50 ( ⁇ m) 1.432 0.2018 0.1261 0.0887 0.1006 0.2186
  • Example 13 14 15 16 17 18 IC 50 ( ⁇ m) 0.07671 0.1491 1.728 4.755 0.2903 >1
  • Example 19 20 twenty one twenty two twenty three 27 IC 50 ( ⁇ m) >100 >100 2.133 >100 >3 >100
  • Example 28 32 33 34 35 36
  • NCI-H1299 cells were trypsinized, centrifuged at 800 rpm for 5 min, the supernatant was discarded, resuspended in fresh medium, counted, and seeded at a density of 1000 cells per well to a 96-well cell culture plate at 5% at 37 °C. The CO 2 incubator was cultured overnight.
  • test substance including the test compound and reference compound AZD1775 mother liquor was serially diluted to 8 concentrations in DMSO at a ratio of 1:3 and 1:10 (the last concentration was DMSO negative control): 10 ⁇ M, 3.3 ⁇ M, 1 ⁇ M , 0.33 ⁇ M, 0.1 ⁇ M, 0.033 ⁇ M, 0.01 ⁇ M, 0 ⁇ M (final concentration of DMSO 1 ⁇ ).
  • DMSO negative control 10 ⁇ M, 3.3 ⁇ M, 1 ⁇ M , 0.33 ⁇ M, 0.1 ⁇ M, 0.033 ⁇ M, 0.01 ⁇ M, 0 ⁇ M (final concentration of DMSO 1 ⁇ ).
  • Add 5 ⁇ L of each concentration to 120 ⁇ L of Petri (25-fold dilution) and mix by shaking.
  • the cells were cultured overnight, the medium was removed, 195 ⁇ L of fresh medium was added to each well, and 5 ⁇ L of the diluted medium containing the corresponding concentration of the test substance was added, respectively, and then the plate was placed in a 37 ° C 5% CO 2 incubator. 3d.
  • the stock solution was removed, and 100 ⁇ L of fresh serum-free DMEM containing MTT (0.5 mg/mL) was added to each well, and the culture was continued. After 4 h, the stock solution was removed, 100 ⁇ L of DMSO was added to each well, and shaken for 10 min in the dark, and placed in a multi-function reader to read the absorbance (OD value) at a wavelength of 552/630/690 nm.
  • the data was analyzed by the software Graph Pad Prism 5.0, and the inhibitory activity of the compound on cell proliferation was plotted in terms of cell viability and compound concentration.
  • Table 4 summarizes the data inhibition (IC 50) for Compound NCI-H1299 cell growth.
  • Example 1 2 3 4 5 6 IC 50 ( ⁇ m) 1.59 0.7817 >10 0.2036 0.9410 0.4611
  • Example 7 8 9 10
  • 11 12
  • Example 13 14 15 16 17 18
  • Example 19 20 twenty one twenty two twenty three 27 IC 50 ( ⁇ m) >10 >10 3.302 >10 >10 >10 Example 28
  • 32 33 34 35 36 IC 50 ( ⁇ m) >3 >10 >10 0.4679 0.2867 >10
  • Example 37 38
  • 40 41
  • Example 43 44
  • 46 47 48
  • Example 49 50 51 52 53

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

本发明提供8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物。具体而言,本发明提供式(I)的化合物,或其可药用盐或前药。式(I)中A和R1-R7为本文所定义。式(I)的化合物是Wee1激酶抑制剂。因此,本发明的化合物可用于治疗由Weel活性异常而导致的疾病。

Description

8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物 技术领域
本发明属于药物化学领域。本发明特别涉及8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物,及其作为治疗上有效的Wee1激酶抑制剂,和抗癌药物的应用。
背景技术
真核细胞的生长、增殖的过程包括母细胞通过准确复制其包括遗传信息的基因组,通过细胞染色体的有丝分裂产生两个相同的子细胞。这种细胞的增殖、分裂过程被称为细胞周期(cell cycle),这包括了细胞从一次分裂完成开始,到下一次分裂完成的整个过程。细胞周期包括四个生长阶段,有丝分裂后的蛋白质,RNA等大量合成的G1期,DNA合成复制的S期,有丝分裂前的准备阶段G2期和细胞进行有丝分裂的M期。细胞根据细胞状况和需要决定通过细胞周期进行分裂增殖,或停止。细胞增殖,分裂必须保持其遗传信息的完整和正确。是否进入细胞周期的下一阶段直至完成整个细胞周期是通过在细胞周期过程中的多个检验点(checkpoint)来保障并完成的。
在细胞周期的整个过程中有多个细胞周期检验点(cell cycle checkpoint)存在。每个细胞周期检验点都包括非常复杂的系统和由多个因子组成。在G1期内的检验点通过检验细胞内外的状态来决定是否进入细胞周期,从而决定细胞是否进入S期DNA合成。G1检验点是个复杂的系统,其中包括著名的CDK4/CDK6。另一个重要的检验点在细胞完成了DNA复制(S期)进入细胞生长期(G2期),即所谓的G2-M检验点。这个检验点检验细胞合成DNA后是否有DNA损伤或缺损,从而决定细胞是否进行下面染色体分离的有丝分裂(M-期)。这一阶段的细胞周期检验点包括了复杂的激酶Cdk1复合体包括Cyclin-B-cdc2(Nurse,P.,1990,Nature 344,503-508)。Cdk1的活化导致有丝分裂的起始,其随后的失活伴随着有丝分裂的完成。Cdk1的活性是通过cdc2结合细胞周期蛋白A(Cyclin-A)或细胞周期蛋白B(Cyclin-B)以及其磷酸化来调节的。比如,细胞周期蛋白B-Cdk1复合物的激活能使细胞有丝分裂(Lindqvist,A.,等人,2009,The Journal of cell biology 185,193-202)。Cdc2在细胞进入有丝分裂前通过磷酸化维持在无活性的状态。其磷酸化状态是通过络氨酸激酶Wee1等来实现的。另外,还有M期细胞周期检验点。
Wee1磷酸化Cdk1上的酪氨酸15(Y15)从而抑制Cdk1的活性(McGowan,C.H.,等人,1993,The EMBO journal 12,75-85;Parker,L.L.,等人,1992,Science 257,1955-1957)。因此,Wee1是Cdk1活性的关键抑制性调节剂,在G2-M期检测点起重要作用,保证当DNA复制完成后在DNA没有损伤的情况下进入有丝分裂(O’Connell,等人,1997,The EMBO journal 16,545-554)。Wee1的丧失或失活可以导致过早进入有丝分裂,引起有丝分裂的失败和细胞死亡(Stumpff,J.,等人,2004,Curr Biol 14,2143-2148)。一些肿瘤细胞的G1期细胞周期检验点有功能缺陷,依赖G2-M期检测点来保障细胞周期的进行(Sancar,A.,等人,2004,Annual review of biochemistry 73,39-85)。在这些癌细胞中由于p53蛋白功能的缺失,丧失Wee1表达或抑制Wee1的活性会导致G2-M期检验点的丧失,使肿瘤细胞对DNA损伤非常敏感,这个敏感化在丧失G1期检验点能力的肿瘤细胞中尤其突出(Wang,Y.,等人,2004,Cancer biology&therapy 3,305-313)。
综上所述,抑制Wee1的活性可以有选择性促使细胞周期检验点有缺陷的癌细胞死亡;同时,对细胞周期检验点正常的正常细胞则作用甚小。因此,Wee1的抑制剂有可能用于癌症及其它细胞增殖病症的治疗的靶向药物。
另外,由于抑制Wee1活性使细胞对DNA损伤的敏感度提高,Wee1抑制剂可以和造成DNA损伤或抑制DNA修复机制有关的抗癌药物联合使用,这包括和PARP抑制剂奥拉帕尼(olaparib)、Niraparib、Rucaparib和Talazoparib;HDAC抑制剂伏立诺他、罗咪地辛、帕比司他和贝利司他等等用于治疗癌症或其它细胞增殖病症。Wee1抑制剂还可能和其他与细胞分裂细胞周期检测点有关的抗癌药物的联合共用,包括Chk1/2抑制剂,CDK4/6抑制剂如帕博西尼,ATM/ATR抑制剂等等用于治疗癌症等病症。
Karnak等人(Clin Cancer Res,2014,20(9):5085-5096)的研究表明Wee1抑制剂AZD1775和PARP抑制剂olaparib联合使用可对放射治疗胰腺癌增敏。其结果证实了Wee1抑制剂和PARP抑制剂联合使用治疗胰腺癌可增敏放射疗效,支持了Wee1抑制使细胞对PARP抑制剂增敏的假设—通过抑制DNA修复和G2检验点功能对辐射治疗增敏,最终可导致未修复的损伤的DNA的积累直至细胞死亡。
另外,有报道(BMC Cancer,2015,15:462)将Wee1抑制剂MK1775和Chk1/2抑制剂AZD7762联合使用用于恶性黑素瘤细胞和异种移植模型中。结果显示,在Wee1和Chk1/2抑制剂的联合使用可协同单一药物的抑制效果,从而降低肿瘤细胞的增殖能力及激活了细胞凋亡机制;在异种移植模型中二者的联合使用可更好地抑制肿瘤生长。
AZD1775是第一个在临床前模型中具有单药抗肿瘤活性的Wee1激酶抑制剂。I期临床研究显示出AZD1775对携带BRCA突变的实体瘤患者的单药疗效,并通过配对肿瘤活检发现跟靶向有关的变化和DNA损伤应答证实了其Wee1激酶抑制机制(JClin Oncol,2015,33:3409-3415)。在AZD1775总共入组200多患者的一个临床I期中,研究了其在治疗晚期实体瘤患者的单药疗效和与吉西他滨、顺铂或卡铂联用的疗效,显示了其在一定的剂量下不管是单药还是与化药联用都是安全且可耐受的。在176例可评估疗效的患者中,94(53%)具有作为最佳应答的稳定疾病,以及17(10%)有部分应答。重要的是,AZD1775在TP53突变患者(n=19)的应答率为21%,而在TP53野生型患者(n=33)的应答率为12%,展现出其对TP53突变患者的巨大潜力(JClin Oncol,2016 Sep 6,pii:JCO675991)。
WO2012161812公开了以下三环化合物作为Wee1激酶抑制剂。其中,X是N或CR1;Y是N或CR2;Z是O、S或NH;R1和R2是H或C1-6烷基;R3是C1-8烷基,C2-8烯基,C3-8环烷基,芳基,或杂芳基等;R4是苯基,萘基,四氢萘基,茚基或茚满基,或5-16元单环、双环或三环杂环基等。
Figure PCTCN2017111230-appb-000001
WO2005021551公开了以下四环嘧啶或吡啶化合物作为蛋白激酶抑制剂。其中,X是N或CH;Y是NH,N(CN),O或S;L是由C和N原子组成的4-原子链;Ra是H,C1-8烷基,CN,苯基或苄基;R1和R2独立是可被取代的饱和或不饱和的5-,6-,或7-元单环,或6-,7-,8-,9-,10-或11-元双环(包含0,1,2,3或4个选自N,O和S的原子,其中O和S原子不同时存在,环中的C原子被0,1或2个氧基取代)等。
Figure PCTCN2017111230-appb-000002
发明内容
如结构式I、II和III所示,本发明提供了新颖的8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物作为激酶抑制剂,特别是Wee1激酶抑制剂。
本发明还提供了包含一个有效量的式I、II和III化合物的药用组合物,用来治疗癌症。
在一具体实施例中,所述药用组合物还可含有一种或多种可药用载体或稀释剂,用来治疗癌症。
在一具体实施例中,所述药用组合物还可含有至少一种已知的抗癌药物或所述抗癌药物的可药用盐,用来治疗癌症。
本发明也涉及到结构式I、II和III的新颖化合物的制备方法。
具体实施方式
如式I、II和III所示,本发明发现新颖的8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物作为激酶抑制剂,特别是Wee1激酶抑制剂。
应理解的是,本文所述的各实施方案的特征可任意组合,形成本文的技术方案;本文对各基团的定义适用于本文所述任一实施方案,例如,本文对烷基的取代基的定义适用于本文所述任一实施方案,除非该实施方案已清楚定义了烷基的取代基。
具体来说,可用于本发明的化合物是式I化合物或其可药用盐或前药:
Figure PCTCN2017111230-appb-000003
其中,A是N或CR15
R1为氢,可被取代的C1-C8烷基,可被取代的C2-C8烯基,可被取代的C3-C8环烷基,可被取代的芳基,可被取代的杂环基或可被取代的杂芳基;
R2为可被取代的碳环基,可被取代的杂环基,可被取代的芳基,或可被取代的杂芳基;
R3-R7和R15独立为氢、卤素、可被取代的氨基、可被取代的烷氧基、可被取代的C1-C10烷基、卤烷基、烯基、炔基、羟基烷基、氨基烷基、羧基烷基、硝基、氰基、酰氨基、羟基、巯基、酰氧基、叠氮基、羧基、亚乙基二氧基,羟基酰氨基或可被取代的烷硫基。
在一个或多个实施方案中,A是N。
在前述一个或多个实施方案中,R1和R2为可被取代的芳基。
在前述一个或多个实施方案中,R3-R7各自独立为氢、卤素或C1-C6烷基。
在前述一个或多个实施方案中,R15为氢或C1-C6烷基。
在前述一个或多个实施方案中,R1上的取代基为选自以下基团中的任意一个、任意两个或任意三个:卤素、C1-C6烷基、C1-C6烷氧基和卤代C1-C6烷基。
在前述一个或多个实施方案中,R1选自:氢,C1-C8烷基,C2-C8烯基,C3-C8环烷基,杂芳基,和任选地被1-4个选自卤素、C1-C6烷基、C1-C6烷氧基和卤代C1-C6烷基的取代基取代的芳基。
在前述一个或多个实施方案中,R1选自任选地被1-4个选自卤素、C1-C6烷基、C1-C6烷氧基和卤代C1-C6烷基的取代基取代的苯基;在某些实施方案中,取代基的数量为2个;在某些实施方案中,至少一个取代基位于邻位;在某些实施方案中,至少一个取代基为卤素;在某些实施方案中,该苯基上的取代基为2个,两个都位于邻位,且其中至少一个为卤素。
在前述一个或多个实施方案中,R1选自可被取代的吡啶基、嘧啶基、噻吩基、呋喃基、吡咯基和咪唑基。
在前述一个或多个实施方案中,R1选自氢,可被取代的C1-C8烷基、C3-C8环烷基和C2-C8烯基。
在前述一个或多个实施方案中,R2的取代基为选自以下基团中的任意一个、任意两个、任意三个或任意四个:可被取代的C1-C6烷基、可被取代的C1-C6酰基、可被取代的杂环基、卤素、可被取代的氧基、硝基和可被取代的C1-C6烷基氨基;优选地,这些可被取代的基团上的取代基可以是1-4个选自以下的基团:C1-C6烷基、C1-C6酰基、任选地被1-4个C1-C6烷基取代的杂环基、卤素、-NRaRb和羟基,其中,Ra和Rb各自独立为H和C1-C6烷基;优选地,所述杂环基选自哌嗪基、哌啶基、吗啉基和1,4-二氮杂环庚烷基。
在前述一个或多个实施方案中,R2的取代基为选自以下基团中的任意一个、任意两个、任意三个或任意四个:可被取代的哌嗪基、可被取代的哌嗪基-C1-C4烷基、可被取代的哌啶基、咪唑基、可被取代的1,4-二氮杂环庚烷基、C1-C6烷基、C1-C6酰基、可被取代的吗啉基、吗啉基-C1-C4烷基、卤素、卤代C1-C6烷基、可被取代的C1-C6烷氧基、可被取代的羟基C1-C6烷基、可被取代的氨基C1-C6烷基、可被取代的哌啶基氨基、可被取代的C1-C6烷基氨基、可被取代的杂环烷基-O-和硝基;优选地,所述可被取代的基团上的取代基可以是1-4个选自以下的基团:C1-C6烷基、C1-C6酰基、卤素、-NRaRb和羟基取代的C1-C6烷基,其中,Ra和Rb各自独立为H和C1-C6烷基。
在前述一个或多个实施方案中,所述可被取代的哌嗪基为可被1、2或3个选自以下的基团取代的哌嗪基:C1-C6烷基、羟基C1-C6烷基和C1-C6酰基。
在前述一个或多个实施方案中,所述哌嗪基至少在对位上具有1个取代基,任选地在间位上具有一个或两个取代基。
在前述一个或多个实施方案中,所述可被取代的哌啶基为可被1个选自C1-C6烷基和C1-C6烷基氨基的基团取代的哌啶基。
在前述一个或多个实施方案中,所述可被取代的吗啉基为可被1或2个选自C1-C6烷基的基团取代的吗啉基。
在前述一个或多个实施方案中,R2选自任选取代的苯基、吡啶基、吡嗪基、四氢异喹啉基和2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基和4,5,6,7-四氢吡唑并[1,5-a]吡嗪-2-基。
在前述一个或多个实施方案中,R2选自:被任选取代的哌嗪基取代的苯基、被任选取代的哌啶基取代的苯基和任选被1-3个C1-C6烷基或卤素取代的四氢异喹啉基。
在前述一个或多个实施方案中,所述哌嗪基任选地被1-3个选自C1-C6烷基、羟基C1-C6烷基和C1-C6酰基的取代基取代。
在前述一个或多个实施方案中,所述哌啶基任选地被1个选自C1-C6烷基和C1-C6烷基氨基的取代基取代。
在前述一个或多个实施方案中,R4和R5各自独立选自H,C1-C6烷基和卤素,优选全部为H。
在前述一个或多个实施方案中,R6和R7各自独立选自H,C1-C6烷基和卤素,优选全部为H。
本发明优选化合物的其中一组表示为式II化合物或其可药用盐或前药:
Figure PCTCN2017111230-appb-000004
其中,R3-R7如在式I中所述;
Ar1为氢,可被取代的C1-C8烷基,可被取代的C2-C8烯基,可被取代的C3-C8环烷基,可被取代的芳基,可被取代的杂环基或可被取代的杂芳基;和
Ar2为可被取代的碳环基,可被取代的杂环基,可被取代的芳基,或可被取代的杂芳基;
在前述一个或多个实施方案中,R3-R7为氢。
在前述一个或多个实施方案中,Ar1和Ar2为可被取代的芳基,更优选的是可被取代的苯基。
在前述一个或多个实施方案中,Ar1上的取代基为选自以下基团中的任意一个、任意两个或任意三个:卤素、C1-C6烷基、C1-C6烷氧基和卤代C1-C6烷基;在某些实施方案中,取代基的数量为2个;在某些实施方案中,至少一个取代基位于邻位;在某些实施方案中,至少一个取代基为卤素;在某些实施方案中,该苯基上的取代基为2个,两个都位于邻位,且其中至少一个为卤素。
在前述一个或多个实施方案中,Ar1选自:可被取代的杂芳基,和任选地被1-4个选自卤素、C1-C6烷基、C1-C6烷氧基和卤代C1-C6烷基的取代基取代的芳基。
在前述一个或多个实施方案中,Ar1选自任选地被1-4个选自卤素、C1-C6烷基、C1-C6烷氧基和卤代C1-C6烷基的取代基取代的苯基。优选地,在这些实施方案中,取代基的数量为2个;更优选地,至少一个取代基位于邻位;更优选地,至少一个取代基为卤素。
在前述一个或多个实施方案中,Ar1选自可被取代的吡啶基、嘧啶基、噻吩基、呋喃基、吡咯基和咪唑基。
在前述一个或多个实施方案中,Ar2的取代基为选自以下基团中的任意一个、任意两个、任意三个或任意四个:可被取代的C1-C6烷基、可被取代的C1-C6酰基、可被取代的杂环基、卤素、可被取代的氧基、硝基和可被取代的C1-C6烷基氨基;优选地,这些可被取代的基团上的取代基可以是1-4个选自以下的基团:C1-C6烷基、C1-C6酰基、任选地被1-4个C1-C6烷基取代的杂环基、卤素、-NRaRb和羟基,其中,Ra和Rb各自独立为H和C1-C6烷基;优选地,所述杂环基选自哌嗪基、哌啶基、吗啉基和1,4-二氮杂环庚烷基。
在前述一个或多个实施方案中,Ar2的取代基为选自以下基团中的任意一个、任意两个、任意三个或任意四个:可被取代的哌嗪基、可被取代的哌嗪基-C1-C4烷基、可被取代的哌啶基、咪唑基、可被取代的1,4-二氮杂环庚烷基、C1-C6烷基、C1-C6酰基、可被取代的吗啉基、吗啉基-C1-C4烷基、卤素、卤代C1-C6烷基、可被取代的C1-C6烷氧基、可被取代的羟基C1-C6烷基、可被取代的氨基C1-C6烷基、可被取代的哌啶基氨基、可被取代的C1-C6烷基氨基、可被取代的杂环烷基-O-和硝基;优选地,所述可被取代的基团上的取代基可以是1~4个选自以下的基团:C1-C6烷基、C1-C6酰基、卤素、-NRaRb和羟基取代的C1-C6烷基,其中,Ra和Rb各自独立为H和C1-C6烷基。
在前述一个或多个实施方案中,所述可被取代的哌嗪基为可被1、2或3个选自以下的基团取代的哌嗪基:C1-C6烷基、羟基C1-C6烷基和C1-C6酰基。
在前述一个或多个实施方案中,所述哌嗪基至少在对位上具有1个取代基,任选地在间位上具有一个或两个取代基。
在前述一个或多个实施方案中,所述可被取代的哌啶基为可被1个选自C1-C6烷基和C1-C6烷基氨基的基团取代的哌啶基。
在一个或多个实施方案中,可被取代的吗啉基为可被1或2个选自C1-C6烷基的基团取代的吗啉基。
在前述一个或多个实施方案中,Ar2选自任选取代的苯基、吡啶基、吡嗪基、四氢异喹啉基和2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基和4,5,6,7-四氢吡唑并[1,5-a]吡嗪-2-基。
在前述一个或多个实施方案中,Ar2选自:被任选取代的哌嗪基取代的苯基、被任选取代的哌啶基取代的苯基和任选被1-3个C1-C6烷基或卤素取代的四氢异喹啉基。
在前述一个或多个实施方案中,所述哌嗪基任选地被1-3个选自C1-C6烷基、羟基C1-C6烷基和C1-C6酰基的取代基取代。
在前述一个或多个实施方案中,所述哌啶基任选地被1个选自C1-C6烷基和C1-C6烷基氨基的取代基取代。
在前述一个或多个实施方案中,R4和R5各自独立选自H,C1-C6烷基和卤素,优选全部为H。
在前述一个或多个实施方案中,R6和R7各自独立选自H,C1-C6烷基和卤素,优选全部为H。
在前述一个或多个实施方案中,R1或Ar1选自:H;C1-C6烷基;C2-C6烯基;C3-C8环烷基;噻唑基;呋喃基;吡咯基;咪唑基;嘧啶基;吡啶基;和任选被1或2个选自卤素、C1-C4烷基、卤代C1-C4烷基和C1-C4烷氧基取代的苯基。
在前述一个或多个实施方案中,R1或Ar1选自:
Figure PCTCN2017111230-appb-000005
Figure PCTCN2017111230-appb-000006
在前述一个或多个实施方案中,R2或Ar2选自:
Figure PCTCN2017111230-appb-000007
Figure PCTCN2017111230-appb-000008
其中,R8独立为氢、卤素、C1-C4烷基和卤代C1-C4烷基,优选为氢、氟、氯、溴、甲基和三氟甲基;R9独立为卤素、C1-C4烷基和卤代C1-C4烷基,优选为氟、氯、甲基和三氟甲基;R10独立为氢、卤素、C1-C4烷基和C1-C4烷氧基,优选为氢、氟、氯、甲基和甲氧基;R11独立为氢、卤素和C1-C4烷基,优选为氢、氟、氯、溴和甲基;R12独立为卤素和C1-C4烷氧基,优选为氯和甲氧基;R13独立为氢、卤素、C1-C4烷基、C1-C4烷氧基和羟基取代的C1-C4烷基,优选为氢、氯、甲基、甲氧基和羟基甲基;R14独立为氢、卤素和C1-C4烷基,优选为氢、氯和甲基。
在前述一个或多个实施方案中,式II的化合物具有下式III所示的结构:
Figure PCTCN2017111230-appb-000009
式中,
Ar1选自:被1个或2个选自卤素、卤代C1-C6烷基、C1-C6烷氧基和C1-C6烷基的取代基取代的苯基;和
Ar2选自:
取代的苯基,其取代基选自:卤素;硝基;C1-C6烷基,任选被1个哌嗪基或1个吗啉基取代、任选被1-3个羟基取代、任选被1-5个卤素取代、或任选被-NRaRb取代,该哌嗪基任选被1-3个选自C1-C4烷基的取代基取代;氧基,任选被C1-C6烷基或哌啶基取代,该哌啶基任选被1-3个选自C1-C4烷基的取代基取代;氨基,其1个氢被哌啶基取代、另一个氢未被取代或被C1-C4烷基取代,或其1或2个氢被C1-C6烷基取代,该哌啶基任选被1-3个选自C1-C4烷基的取代基取代,该C1-C6烷基任选被-NRaRb取代;任选被1-3个选自C1-C6烷基、羟基C1-C6烷基和C1-C6酰基的取代基取代的哌嗪基;任选被1个选自-NRaRb的取代基取代的哌啶基;任选被1-3个C1-C6烷基取代的1,4-二氮杂环庚烷-1-基;咪唑基;和任选被1-3个C1-C6烷基取代的吗啉基;
任选被1-3个选自C1-C6烷基和卤素的取代基取代的四氢异喹啉基;
任选被1-3个选自C1-C6烷基、C1-C6酰基和卤素的取代基取代的2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基;
任选被1个任选被1-3个C1-C6烷基取代的哌嗪基取代的吡啶基;
任选被1个任选被1-3个C1-C6烷基取代的哌嗪基取代的吡嗪基;和
任选被1-3个C1-C6烷基取代的4,5,6,7-四氢吡唑并[1,5-a]吡嗪-2-基;
其中,Ra和Rb各自独立为H或C1-C6烷基。
在前述一个或多个实施方案中,式III中,
Ar1选自:被两个选自卤素和C1-C6烷基的取代基取代的苯基;和
Ar2选自:取代的苯基,其取代基选自:卤素,卤代C1-C6烷基,C1-C6烷基,C1-C6烷氧基,羟基C1-C6烷基,NRaRb-C1-C6烷基,任选被1-3个C1-C4烷基取代的哌嗪基取代的C1-C4烷基,任选被1-3个C1-C4烷基取代的哌啶基-O-,NRaRb-C1-C6烷氧基,NRaRb-C1-C6烷基-NRa-,被1-3个选自C1-C6烷基、羟基C1-C6烷基和C1-C6酰基的取代基取代的哌嗪基,被1个选自C1-C6烷基和-NRaRb的取代基取代的哌啶基,任选被1-3个C1-C6烷基取代的1,4-二氮杂环庚烷-1-基和任选被1-3个C1-C6烷基取代的吗啉基;被1-3个选自C1-C6烷基和卤素的取代基取代的四氢异喹啉基;被1-3个选自C1-C6烷基、C1-C4酰基和卤素的取代基取代的2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基;和被1-3个C1-C6烷基取代的4,5,6,7-四氢吡唑并[1,5-a]吡嗪-2-基;其中,Ra和Rb独自选自H和C1-C4烷基。
在前述一个或多个实施方案中,式III中,Ar1为两个间位分别被选自卤素、C1-C3烷基、卤代C1-C3烷基和C1-C3烷氧基的取代基取代的二取代苯基,优选地,两个取代基中至少一个为卤素;Ar2为:被1、2或3个取代基取代的苯基,所述取代基选自:卤素,卤代C1-C6烷基,C1-C6烷基,C1-C6烷氧基,羟基C1-C6烷基,NRaRb-C1-C6烷基,任选被1-3个C1-C4烷基取代的哌嗪基取代的C1-C4烷基,任选被1-3个C1-C4烷基取代的哌啶基-O-,NRaRb-C1-C6烷氧基,NRaRb-C1-C6烷基-NRa-,任选被1-3个选自C1-C6烷基和羟基C1-C6烷基的取代基取代的哌嗪基,被1个选自C1-C6烷基和-NRaRb的取代基取代的哌啶基,任选被1-3个C1-C6烷基取代的1,4-二氮杂环庚烷-1-基和任选被1-3个C1-C6烷基取代的吗啉基;被1-3个选自C1-C6烷基和卤素的取代基取代的四氢异喹啉基;和被1-3个选自C1-C6烷基、C1-C4酰基和卤素的取代基取代的2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基;其中,Ra和Rb独自选自H和C1-C4烷基。
在前述一个或多个实施方案中,式III中,
Ar1优选为:
Figure PCTCN2017111230-appb-000010
Ar1更为优选为:
Figure PCTCN2017111230-appb-000011
Ar2优选为:
Figure PCTCN2017111230-appb-000012
Figure PCTCN2017111230-appb-000013
其中,R8独立为氢、氟、氯、溴、甲基和三氟甲基;R9独立为氢、氟、氯、溴、甲基和三氟甲基;R10独立为氢、氟、氯、溴、甲基和甲氧基;R11独立为氢、氟、氯、溴、甲基和三氟甲基;R12独立为氯、氟、溴、甲基和甲氧基;R13独立为氢、氯、甲基、甲氧基和羟基甲基;R14独立为为氢、氯、溴和甲基;
Ar2更为优选为:
Figure PCTCN2017111230-appb-000014
其中,R8独立为氢、氟、氯、溴和甲基;R9独立为氢、氯和甲基;R10独立为氢、氯、甲基和甲氧基;R11独立为氢、氟、氯、溴和甲基;R12独立为氢、氯、甲基和甲氧基;R14独立为氢、氯和甲基。
在前述一个或多个实施方案中,所述化合物不包括实施例83的化合物;在某些实施方案中,所述式I化合物不包括R1为邻位为两个甲基或两个烷基取代的苯基、R2为甲基或烷基取代的哌嗪基单取代的苯基、R3-R7为H的化合物。
在前述一个或多个实施方案中,式I优选的化合物包括但不限于:
6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例1);
6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例2);
4-(2-氯苯基)-8-((4-(4-甲基哌嗪-1-基)苯基)氨基)-1,2-二氢咪唑并[1,2-a]吡啶并[3,4-e]嘧啶-5(4H)-酮(实施例3);
6-(2,6-二氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例4);
6-(2-氯-6-甲基苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例5);
6-(2-氯-6-氟苯基)-2-((4-(4-异丙基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例6);
6-(2-氯-6-氟苯基)-2-((4-(4-乙酰基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例7);
6-(2-氯-6-氟苯基)-2-((2-甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例8);
6-(2-氯-6-氟苯基)-2-((2,4,4-三甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例9);
6-(2-氯-6-氟苯基)-2-((2'-甲基-2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例10);
6-(2-氯-6-氟苯基)-2-((2'-乙酰基-2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例11);
6-(2,6-二氯苯基)-2-((2-甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例12);
6-(2,6-二氯苯基)-2-((2,4,4-三甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例13);
6-(2,6-二氯苯基)-2-((2'-甲基-2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例14);
6-(2,6-二甲基苯基)-2-((2-甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例15);
6-(2,6-二甲基苯基)-2-((2,4,4-三甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例16);
6-(2,6-二甲基苯基)-2-((2'-甲基-2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例17);
6-异丙基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例18);
6-叔丁基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例19);
6-环丙基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例20);
6-环己基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例21);
6-烯丙基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例22);
6-(噻吩-2-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例23);
6-(呋喃-2-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例24);
6-(1H-吡咯-2-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例25);
6-(1H-咪唑-5-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例26);
6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,8-二甲基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例27);
6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-9,9-二甲基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例28);
6-(2-氯-6-氟苯基)-2-((4-((2S,6R)-2,6-二甲基吗啉基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例32);
6-(2-氯-6-氟苯基)-2-((4-(吗啉基甲基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例33);
6-(2-氯-6-氟苯基)-2-((4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例34);
6-(2-氯-6-氟苯基)-2-((4-((3S,5R)-4-异丙基-3,5-二甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例35);
6-(2-氯-6-氟苯基)-2-((2-氟-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例36);
6-(2-氯-6-氟苯基)-2-((2-氟-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例37);
6-(2-氯-6-氟苯基)-2-((2-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例38);
6-(2-氯-6-氟苯基)-2-((2-氯-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例39);
6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)-2-(三氟甲基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例40);
6-(2-氯-6-氟苯基)-2-((2-三氟甲基-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例41);
6-(2-氯-6-氟苯基)-2-((2-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例42);
6-(2-氯-6-氟苯基)-2-((2-甲基-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例43);
6-(2-氯-6-氟苯基)-2-((3-氟-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例44);
6-(2-氯-6-氟苯基)-2-((3-氟-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例45);
6-(2-氯-6-氟苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例46);
6-(2-氯-6-氟苯基)-2-((3-氯-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例47);
6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)-3-(三氟甲基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例48);
6-(2-氯-6-氟苯基)-2-((3-三氟甲基-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例49);
6-(2-氯-6-氟苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例50);
6-(2-氯-6-氟苯基)-2-((3-甲基-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例51);
6-(2-氯-6-氟苯基)-2-((4-(4-异丙基哌嗪-1-基)-3-甲基苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例52);
6-(2-氯-6-氟苯基)-2-((4-((3S,5R)-4-异丙基-3,5-二甲基哌嗪-1-基)-3-甲基苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例53);
6-(2-氯-6-氟苯基)-2-((3-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例54);
6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)-3-硝基苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例55);
6-(2-氯-6-氟苯基)-2-((3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例56);
6-(2-氯-6-氟苯基)-2-((6-(4-甲基哌嗪-1-基)吡啶-3-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例57);
6-(2-氯-6-氟苯基)-2-((2'-异丙基-2',3'-二氢-1'H-螺[环丙烷-1,4'-异喹啉]-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例58);
6-(2-氯-6-氟苯基)-2-((5-甲基-4,5,6,7-四氢吡唑并[1,5-a]吡嗪-2-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例59);
6-(2,6-二氟苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例60);
6-(2-氟-6-三氟甲基苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例61);
6-(2-氟-6-甲基苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例62);
6-(2,6-二氯苯基)-2-((4-(4-异丙基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例63);
6-(2,6-二氯苯基)-2-((4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例64);
6-(2,6-二氯苯基)-2-((4-((3S,5R)-4-异丙基-3,5-二甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例65);
6-(2,6-二氯苯基)-2-((2-氟-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例66);
6-(2,6-二氯苯基)-2-((2-氯-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例67);
6-(2,6-二氯苯基)-2-((2-三氟甲基-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例68);
6-(2,6-二氯苯基)-2-((3-氟-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例69);
6-(2,6-二氯苯基)-2-((3-氟-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例70);
6-(2,6-二氯苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例71);
6-(2,6-二氯苯基)-2-((3-氯-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例72);
6-(2,6-二氯苯基)-2-((4-(4甲基哌嗪-1-基)-3-(三氟甲基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例73);
6-(2,6-二氯苯基)-2-((3-三氟甲基-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例74);
6-(2,6-二氯苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例75);
6-(2,6-二氯苯基)-2-((3-甲基-4-(4-异丙基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例76);
6-(2,6-二氯苯基)-2-((3-甲基-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例77);
6-(2,6-二氯苯基)-2-((3-甲基-4-((3S,5R)-4-异丙基-3,5-二甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例78);
6-(2,6-二氯苯基)-2-((5-甲基-4,5,6,7-四氢吡唑并[1,5-a]吡嗪-2-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例79);
6-(2-氯-6-三氟甲基苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例80);
6-(2-氯-6-甲基苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例81);
6-(2-氯-6-甲氧基苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例82);
6-(2,6-二甲基苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例83);
6-(2,6-二甲基苯基)-2-((4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例84);
6-(4-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例85);
6-(3-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例86);
6-(2,4-二氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例87);
6-(2-氯-4-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例88);
6-(2-氯-3-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例89);
6-(2,5-二氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例90);
6-(2,3-二氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例91);
6-(嘧啶-2-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例92);
2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例93);
6-环丁基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例94);
6-环戊基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例95);
6-苯基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例96);
6-(吡啶-2-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例97);
6-(吡啶-3-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例98);
6-(吡啶-4-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例99);
6-(2,6-二氟苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例100);
6-(2,6-二氟苯基)-2-((3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例101);
6-(2-氯-6-氟苯基)-2-((4-(1H-咪唑-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例102);
6-(2-氯-6-氟苯基)-2-((4-(4-甲基-1,4-二氮杂环庚烷-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例103);
6-(2-氯-6-氟苯基)-2-((4-((4-甲基哌嗪-1-基)甲基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例104);
6-(2-氯-6-氟苯基)-2-((4-(2-(二甲氨基)乙氧基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例105);
6-(2-氯-6-氟苯基)-2-((4-(3-(二甲氨基)丙氧基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例106);
6-(2-氯-6-氟苯基)-2-((4-((1-甲基哌啶-4-基)氧基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例107);
6-(2-氯-6-氟苯基)-2-((4-((2-(二甲氨基)乙基)氨基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例108);
6-(2-氯-6-氟苯基)-2-((4-((2-(二甲基氨基)乙基)(甲基)氨基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例109);
6-(2-氯-6-氟苯基)-2-((4-((3-(二甲氨基)丙基)氨基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例110);
6-(2-氯-6-氟苯基)-2-((4-((3-(二甲基氨基)丙基)(甲基)氨基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例111);
6-(2-氯-6-氟苯基)-2-((4-((1-甲基哌啶-4-基)氨基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例112);
6-(2-氯-6-氟苯基)-2-((4-(甲基(1-甲基哌啶-4-基)氨基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例113);
6-(2-氯-6-氟苯基)-2-((4-(1-甲基哌啶-4-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例114);
6-(2-氯-6-氟苯基)-2-((4-(4-二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例115);
6-(2-氯-6-氟苯基)-2-((3-氟-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例116);
6-(2-氯-6-氟苯基)-2-((3-氯-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例117);
6-(2-氯-6-氟苯基)-2-((3-溴-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例118);
6-(2-氯-6-氟苯基)-2-((3-甲基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮的(实施例119);
6-(2-氯-6-氟苯基)-2-((3-氟-5-甲基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例120);
6-(2-氯-6-氟苯基)-2-((5-(4-甲基哌嗪-1-基)吡嗪-2-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例121);
6-(2-氯-6-氟苯基)-2-((3,5-二氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例122);
6-(2-氯-6-氟苯基)-2-((3-氟-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例123);
6-(2-氯-6-氟苯基)-2-((3-氟-5-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例124);
6-(2-氯-6-氟苯基)-2-((3-氯-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例125);
6-(2-氯-6-氟苯基)-2-((3-氯-5-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例126);
6-(2-氯-6-氟苯基)-2-((3-溴-5-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例127);
6-(2-氯-6-氟苯基)-2-((3-甲基-5-三氟甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶[5,4-e]嘧啶-5(6H)-酮(实施例128);
6-(2-氯-6-氟苯基)-2-((3-甲氧基-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶[5,4-e]嘧啶-5(6H)-酮(实施例129);
6-(2,6-二氯苯基)-2-((5-氯-2-甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶[5,4-e]嘧啶-5(6H)-酮(实施例130);
6-(2,6-二氯苯基)-2-((2,5-二甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶[5,4-e]嘧啶-5(6H)-酮(实施例131);
6-(2,6-二氯苯基)-2-((5-氯-2,4,4-甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶[5,4-e]嘧啶-5(6H)-酮(实施例132);
6-(2,6-二氯苯基)-2-((2,4,4,5-四甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶[5,4-e]嘧啶-5(6H)-酮(实施例133);
6-(2,6-二氯苯基)-2-((5'-氯-2'-甲基-2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例134);
6-(2,6-二氯苯基)-2-((2',5'-二甲基-2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例135);
6-(2,6-二氯苯基)-2-((3,5-二氯-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例136);
6-(2,6-二氯苯基)-2-((3-氯-5-甲基-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例137);
6-(2,6-二氯苯基)-2-((4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例138);
6-(2,6-二氯苯基)-2-((3-氯-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例139);
6-(2,6-二氯苯基)-2-((3-溴-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例140);
6-(2,6-二氯苯基)-2-((3-甲基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例141);
6-(2,6-二氯苯基)-2-((3,5-二氯-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例142);
6-(2,6-二氯苯基)-2-((3-氟-5-甲基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例143);
6-(2,6-二氯苯基)-2-((3-氯-5-甲氧基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例144);
6-(2,6-二氯苯基)-2-((3-溴-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例145);
6-(2,6-二氯苯基)-2-((3-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例146);
6-(2,6-二氯苯基)-2-((3-氯-5-氟-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例147);
6-(2,6-二氯苯基)-2-((3-氟-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例148);
6-(2,6-二氯苯基)-2-((3-氟-5-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例149);
6-(2,6-二氯苯基)-2-((3,5-二氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例150);
6-(2,6-二氯苯基)-2-((3-氯-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例151);
6-(2,6-二氯苯基)-2-((3-氯-5-三氟甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例152);
6-(2,6-二氯苯基)-2-((3-氯-5-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例153);
6-(2,6-二氯苯基)-2-((3-溴-5-氟-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例154);
6-(2,6-二氯苯基)-2-((3-溴-5-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例155);
6-(2,6-二氯苯基)-2-((3-溴-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例156);
6-(2,6-二氯苯基)-2-((3-溴-5-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例157);
6-(2,6-二氯苯基)-2-((3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例158);
6-(2,6-二氯苯基)-2-((3-甲氧基-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例159);
6-(2-溴-6-氯苯基)-2-((3-氟-5-甲基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例160);
6-(2-溴-6-氯苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例161);
6-(2-溴-6-氯苯基)-2-((3-溴-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例162);
6-(2-溴-6-氯苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例163);
6-(2-溴-6-氯苯基)-2-((3,5-二氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例164);
6-(2-溴-6-氯苯基)-2-((3-氟-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例165);
6-(2-溴-6-氯苯基)-2-((3-氯-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例166);
6-(2-溴-6-氯苯基)-2-((3-溴-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例167);
6-(2-氟-6-甲基苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例168);
6-(2-氟-6-甲基苯基)-2-((3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例169);
6-(2-氯-6-甲基苯基)-2-((3-氟-5-甲基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例170);
6-(2-氯-6-甲基苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例171);
6-(2-氯-6-甲基苯基)-2-((3,5-二氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例172);
6-(2-氯-6-甲基苯基)-2-((3-氯-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例173);
6-(2-氯-6-甲基苯基)-2-((3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例174);
6-(2,6-二氯苯基)-2-((4-(哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例175);
6-(2,6-二氯苯基)-2-((3-氯-4-(哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例176);
6-(2,6-二氯苯基)-2-((3-甲基-4-(哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例177);
6-(2,6-二氯苯基)-2-((3-甲氧基-4-(哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例178);
6-(2-氯-6-氟苯基)-2-((3-(羟基甲基)-4-(哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例179);
6-(2,6-二氯苯基)-2-((3-(羟基甲基)-4-(哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例180);
6-(2,6-二氯苯基)-2-((3-(羟基甲基)-4-(甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例181);
6-(2,6-二氯苯基)-2-((4-(4-(2-羟基乙基)哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例182);
6-(2,6-二氯苯基)-2-((4-吗啉基苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例183);
6-(2,6-二氯苯基)-2-((3-((甲基氨基)甲基)-4-吗啉基苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例184);
6-(2-溴-6-氯苯基)-2-((3-氯-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例185);
6-(2,6-二氯苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)胺基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮二盐酸盐(实施例186)
或其可药用盐或前药。
本文所用“烷基”是指烷基本身或是直链或支链高达十个碳原子的基团。有用的烷基包括直链或支链C1-C10烷基,优选C1-C6烷基。在某些实施方案中,烷基为C1-C4烷基。典型的C1-C10烷基包括可被任意取代的甲基、乙基、丙基、异丙基、丁基、仲丁基、叔丁基、3-戊基、己基和辛基。
本文所用“烯基”是指直链或支链含有2-10个碳原子,除非碳链长度被另外限制,其中至少是链中的两个碳原子之间含有一个双键的基团;优选C2-C6烯基。典型的链烯基包括乙烯基、1-丙烯基、2-丙烯基、2-甲基-1-丙烯基、1-丁烯基和2-丁烯基。
本文所用“炔基”是指直链或支链含有2-10个碳原子,除非碳链长度被另外限制,其中至少是链中的两个碳原子之间含有一个叁键的基团;优选C2-C6炔基。典型的炔基包括乙炔基、1-丙炔基、1-甲基-2-丙炔基、2-丙炔基、1-丁炔基和2-丁炔基。
有用的烷氧基包括被上述C1-C10烷基、优选C1-C6烷基或C1-C4烷基取代的氧基,例如甲氧基、乙氧基等。烷氧基中的烷基可被任意取代。烷氧基的取代基包括但不限于卤素、吗啉基、氨基,所述氨基包括烷氨基和二烷氨基以及羧基(包括其酯基)。
有用的烷硫基包括被上述C1-C10烷基、优选C1-C6烷基取代的硫基,烷硫基中的烷基可被任意取代。同时还包括这类烷硫基的亚砜和砜。
有用的氨基和可被任意取代的氨基包括-NH2、-NHR’和-NR’R”,其中R’和R”是可被任意取代的C1-C10烷基,环烷基,芳基,杂芳基或氨基。或者R’和R”与N一起形成5-8员杂环例如哌啶,或者R’和R”与N以及与其它N或O一起形成5-8员杂环例如哌嗪。所述烷基和杂环可被任意取代。
本文中,除非另有说明,当被取代时,烷基、烷氧基、烷硫基、烯基、炔基、环烷基、羰基、碳环和杂环、芳基、芳基烷基、芳基链烯基、芳基炔基、杂芳基和杂芳基烷基可被一个或多个(例如1、2、3或4个)选自以下基团的取代基取代:卤素、羟基、羧基、氨基、硝基、氰基、C1-C6酰氨基、C1-C6酰氧基、C1-C6烷氧基、芳氧基、烷硫基、C1-C6烷基、C1-C6酰基、C6-C10芳基、C3-C8环烷基、C2-C6链烯基、C2-C6炔基、C6-C10芳基(C2-C6)链烯基、C6-C10芳基(C2-C6)炔基、饱和和不饱和的杂环基或杂芳基、亚甲基二氧基、C1-C6卤代烷基、C6-C10芳基(C1-C6)烷基、C1-C6羟烷基、脲基、巯基、叠氮基、羰基、二(C1-C10烷基)氨基、烷磺酰基、氨磺酰基、二烷基氨磺酰基和烷基亚磺酰基等。其中取代基本身也可被任意取代。
除非另有说明,当被取代时,优选地,烷基、烷氧基、烷硫基、烯基、炔基、环烷基、羰基、碳环和杂环可被一个或多个(例如1、2、3或4个)选自以下基团的取代基取代:卤素、羟基、羧基、氨基、硝基、氰基、C1-C6酰氨基、C1-C6酰基、C1-C6酰氧基、C1-C6烷氧基、芳氧基、烷硫基、C6-C10芳基、C3-C8环烷基、C2-C6链烯基、C2-C6炔基、C6-C10芳基(C2-C6)链烯基、C6-C10芳基(C2-C6)炔基、饱和和不饱和的杂环基或杂芳基。
除非另有说明,当被取代时,芳基、芳基烷基、芳基链烯基、芳基炔基、杂芳基和杂芳基烷基可被一个或多个(例如1、2、3或4个)选自以下基团的取代基取代:卤素、亚甲基二氧基、C1-C6卤代烷基、C6-C10芳基、C3-C8环烷基、C1-C6烷基、C2-C6链烯基、C2-C6炔基、C6-C10芳基(C1-C6)烷基、C6-C10芳基(C2-C6)链烯基、C6-C10芳基(C2-C6)炔基、C1-C6羟基烷基、硝基、氨基、脲基、氰基、C1-C6酰基氨基、羟基、巯基、C1-C6酰氧基、叠氮基、C1-C6烷氧基、羰基、羧基、二(C1-C10烷基)氨基、烷基磺酰基、氨基磺酰基、二烷基氨基磺酰基或烷基亚磺酰基。
应理解的是,本文各实施方案中,当取代基为杂环基、芳基或杂芳基时,该杂环基、芳基或杂芳基取代基的数量通常为1个。
本文所用“芳基”是指芳基本身或是作为其它基团的一部分,指含有6到14个碳原子的单环、双环或三环芳族基团。
有用的芳基包括C6-C14芳基,优选的是C6-C10芳基。典型的C6-C14芳基包括苯基、萘基、菲基、蒽基、茚基、薁基、联苯、亚联苯基和茀基。
这里所指“碳环”包括环烷基和部分饱和的碳环基团。有用的环烷基是C3-C8环烷基。典型的环烷基包括环丙级、环丁基、环戊基、环己基和环庚基。
有用的饱和的或是部分饱和的碳环基团是上面所描述的环烷基和环烯基,例如环戊烯基、环庚烯基和环辛烯基。
有用的卤素或卤素基团包括氟、氯、溴和碘。
本文所用“芳基烷基”包括被任一上述C6-C14芳基取代的C1-C10烷基。优选的芳基烷基是苄基、苯乙基或萘甲基。
本文所用“芳基烯基”包括被任一上述C6-C14芳基取代的C2-C10烯基。
本文所用“芳基炔基”包括被任一上述C6-C14芳基取代的C2-C10炔基。
本文所用“芳氧基”包括被任一上述C6-C14芳基取代的氧基,其芳基可被任意取代。有用的芳氧基包括苯氧基和4-甲基苯氧基。
本文所用“芳基烷氧基”包括被任一上述芳基取代的C1-C10烷氧基,其芳基可被任意取代。有用的芳基烷氧基包括苄氧基和苯基乙氧基。
有用的卤代烷基包括被一个或多个氟、氯、溴或碘原子取代的C1-C10烷基,优选C1-C6烷基,例如氟甲基、二氟甲基、三氟甲基、五氟乙基、1,1-二氟乙基、氯甲基、氯氟甲基和三氯甲基。
有用的酰基氨基(酰氨基)是连接在氨基氮上的任何C1-C6酰基(烷酰基),例如乙酰氨基、氯乙酰氨基、丙酰氨基、丁酰氨基、戊酰氨基和已酰氨基,以及芳基取代的C1-C6酰基氨基,例如苯甲酰氨基和五氟苯甲酰氨基。有用的酰基包括C1-C6酰基,如乙酰基。
有用的酰氧基是连接在氧(-O-)上的任何C1-C6酰基(烷酰基),例如甲酰氧基、乙酰氧基、丙酰氧基、丁酰氧基、戊酰氧基和己酰氧基。
本文所用杂环(杂环基)是指饱和或部分饱和的3-7员单环,或7-10员双环体系,它是由碳原子和从O、N、S中任选1--4个杂原子组成的,其中杂原子氮和硫都可以被任意氧化,氮也可以任意季铵化,并且包括双环体系中上述定义的任意杂环与苯环的融合。如果产生的化合物是稳定的话,那么杂环的碳原子或氮原子可被取代。
有用的饱和或部分饱和杂环基团包括四氢呋喃基、吡喃基、哌啶基、哌嗪基、1,4-二氮杂环庚烷基、吡咯烷基、咪唑烷基、咪唑啉基、二氢吲哚基、异二氢吲哚基、奎宁环基、吗啉基、异色满基、色满基、吡唑烷基、吡唑啉基、四氢异喹啉基、2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基、4,5,6,7-四氢吡唑并[1,5-a]吡嗪-2-基、tetronoyl和tetramoyl,这些基团可被任意取代。
本文所用“杂芳环”是指含有5-14个环原子,并且有6个,10个或14个π电子在环体系上共用。而且所含环原子是碳原子和从氧、氮、硫中任选的1-3个杂原子。
有用的杂芳基包括噻吩基(苯硫基)、苯并[d]异噻唑-3-基、苯并[b]噻吩基、萘并[2,3-b]噻吩基、噻蒽基、呋喃基、吡喃基、异苯并呋喃基、色烯基、夹氧蒽基、噻吩恶基(phenoxanthiinyl)、吡咯基、咪唑基、吡唑基、吡啶基(包括但不限制于2-吡啶基、3-吡啶基和4-吡啶基)、吡嗪基、嘧啶基、哒嗪基、吲嗪基、异吲哚基、3H-吲哚基、吲哚基、吲唑基、嘌呤基、4H-喹嗪基、异喹啉基、喹啉基、酞嗪基、萘啶基、喹唑啉基、噌啉基、蝶啶基、咔唑基、β-咔啉基、菲啶基、吖啶基、萘嵌间二氮(杂)苯基、菲咯啉基、吩嗪基、异噻唑基、吩噻嗪基、异恶唑基、呋咱基、吩恶嗪基、1,4-二氢喹喔啉-2,3-二酮、7-氨基异香豆素、吡啶并[1,2-a]嘧啶-4-酮、四氢化五员[c]吡唑-3-基、吡唑[1,5-a]嘧啶基、苯并异恶唑基如1,2-苯并异恶唑-3-基、苯并咪唑基、2-羟吲哚基、噻重氮基和2-氧代苯并咪唑基。当杂芳基在环中含有氮原子时,这样的氮原子可以呈N-氧化物形式,例如吡啶基N-氧化物、吡嗪基N-氧化物和嘧啶基N-氧化物。
本文所用“杂芳氧基”包括被任一上述杂芳基取代的氧基,其中杂芳基上可有取代基。有用的杂芳氧基包括吡啶氧基、吡嗪氧基、吡咯氧基、吡唑氧基、咪唑氧基和苯硫基氧基。
本文所用“杂芳基烷氧基”是指被任一上述杂芳基取代的任一上述C1-10烷氧基,其中杂芳基上可有取代基。
一些本发明化合物可能作为立体异构体,包括旋光异构体存在。本发明包括所有立体异构体和这样的立体异构体的外消旋混合物,以及可以根据本领域技术人员众所周知的方法分离出来的单独的对映体。
可药用盐的例子包括无机和有机酸盐,例如盐酸盐、氢溴酸盐、磷酸盐、硫酸盐、柠檬酸盐、乳酸盐、酒石酸盐、马来酸盐、富马酸盐、扁桃酸盐和草酸盐;以及与碱例如钠羟基、三(羟基甲基)氨基甲烷(TRIS,氨丁三醇)和N-甲基葡糖胺形成的无机和有机碱盐。
本发明化合物的前药的实施例包括含有羧酸的化合物的简单酯(例如依据本领域已知方法通过与C1-4醇缩合而获得的酯);含有羟基的化合物的酯(例如依据本领域已知方法通过与C1-4羧酸、C3-6二酸或其酸酐例如琥珀酸酐和富马酸酐缩合而获得的酯);含有氨基的化合物的亚胺(例如依据本领域已知方法通过与C1-4醛或酮缩合而获得的亚胺);含有氨基的化合物的氨基甲酸酯,例如Leu等人(J.Med.Chem.42:3623-3628(1999))和Greenwald等人(J.Med.Chem.42:3657-3667(1999))描述的那些酯;含有醇的化合物的醛缩醇或酮缩醇(例如依据本领域已知方法通过与氯甲基甲基醚或氯甲基乙基醚缩合而获得的那些缩醇)。
本发明化合物可使用本领域技术人员已知的方法或本发明新方法制得。具体来说,具有式Ⅰ的本发明化合物可如反应方案1中的反应实施例所示制得。N-叔丁氧羰基-1,2-乙二胺和取代异氰酸苯酯,如邻氯苯异氰酸酯,在乙醚中在室温下反应,得到产物(2-(3-(2-氯苯基)脲基)乙基)氨基甲酸叔丁酯。该氨基甲酸叔丁酯和盐酸二氧六环溶液在二氯甲烷中室温反应,得到产物1-(2-氨基乙基)-3-(2-氯苯基)脲(盐酸盐)。该脲(盐酸盐)和4-氯-2-(甲硫基)嘧啶-5-甲酸乙酯,二异丙基乙基胺在乙腈中加热反应,得到产物4-((2-(3-(2-氯苯基)脲基)乙基)氨基)-2-(甲硫基)嘧啶-5-甲酸乙酯。4-((2-(3-(2-氯苯基)脲基)乙基)氨基)-2-(甲硫基)嘧啶-5-甲酸乙酯在三氯氧磷中加热反应,得到产物6-(2-氯苯基)-2-(甲硫基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。6-(2-氯苯基)-2-(甲硫基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮和间氯过氧苯甲酸在氯仿中室温反应,得到产物6-(2-氯苯基)-2-(甲磺酰基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。该酮和4-(4-甲基哌嗪)苯胺在异丙醇中加热反应,得到目标化合物6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。
反应方案1
Figure PCTCN2017111230-appb-000015
其它相关化合物可用类似方法制得。例如,用1-氯-3-氟-2-异氰酸苯酯替代邻氯苯异氰酸酯可制得目标化合物6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。用4-(4-异丙基哌嗪-1-基)苯胺替代4-(4-甲基哌嗪)苯胺可制得目标化合物6-(2-氯-6-氟苯基)-2-((4-(4-异丙基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。用2-甲基-1,2,3,4-四氢异喹啉-7-胺替代4-(4-甲基哌嗪)苯胺可制得目标化合物6-(2-氯-6-氟苯基)-2-((2-甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。用1-(4-氨基苯基)-N,N-二甲基哌啶-4-胺替代4-(4-甲基哌嗪)苯胺可制得目标化合物6-(2-氯-6-氟苯基)-2-((4-(4-二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。
本发明化合物可如反应方案2中的反应实施例所示制得。1-(2-氨基乙基)-3-(2-氯苯基)脲和4,6-二氯烟酸甲酯,二异丙基乙基胺混合于乙腈中加热反应,得到产物6-氯-4-((2-(3-(2-氯苯基)脲基)乙基)氨基)烟酸甲酯。6-氯-4-((2-(3-(2-氯苯基)脲基)乙基)氨基)烟酸甲酯在三氯氧磷中加热反应,得到产物8-氯-4-(2-氯苯基)-1,2-二氢咪唑并[1,2-a]吡啶并[3,4-e]嘧啶-5(4H)-酮。8-氯-4-(2-氯苯基)-1,2-二氢咪唑并[1,2-a]吡啶并[3,4-e]嘧啶-5(4H)-酮,4-(4-甲基哌嗪)苯胺,三(二亚苄基丙酮)二钯,2-二环己基磷-2,4,6-三异丙基联苯和磷酸钾混合于叔丁醇和水中加热反应,得到目标化合物4-(2-氯苯基)-8-((4-(4-甲基哌嗪-1-基)苯基)氨基)-1,2-二氢咪唑并[1,2-a]吡啶并[3,4-e]嘧啶-5(4H)-酮。
反应方案2
Figure PCTCN2017111230-appb-000016
本发明化合物可如反应方案3中的反应实施例所示制得。应用类似于反应方案1所示方法可制得2-(甲磺酰基)-6-(2,6-二氯苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。2-(甲磺酰基)-6-(2,6-二氯苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮、4-(4-甲基哌嗪-1-基)苯胺和三氟乙酸在乙腈中室温反应,得到目标化合物6-(2,6-二氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。
反应方案3
Figure PCTCN2017111230-appb-000017
其它相关化合物可用类似方法制得。例如,用2,4,4-三甲基-1,2,3,4-四氢异喹啉-7-胺替代4-(4-甲基哌嗪-1-基)苯胺可制得目标化合物2-((2,4,4-三甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-6-(2,6-二氯苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。用4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯胺替代4-(4-甲基哌嗪-1-基)苯胺可制得目标化合物2-((4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-6-(2,6-二氯苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。用2-甲磺酰基-6-(2-氯-4-氟苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮替代2-(甲磺酰基)-6-(2,6-二氯苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮可制得目标化合物2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-6-(2-氯-4-氟苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。用2-甲磺酰基-6-异丙基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮替代2-(甲磺酰基)-6-(2,6-二氯苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮可制得目标化合物6-异丙基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。用2-甲磺酰基-6-(嘧啶-2-基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮替代2-(甲磺酰基)-6-(2,6-二氯苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮可制得目标化合物2-((4-(4-甲基哌嗪-1-基) 苯基)氨基)-6-(嘧啶-2-基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。用2-甲亚磺酰基-6-(2-氯-6-氟苯基)-8,8-二甲基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮替代2-(甲磺酰基)-6-(2,6-二氯苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮可制得目标化合物6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,8-二甲基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。用1-(4-氨基苯基)-N,N-二甲基哌啶-4-胺替代4-(4-甲基哌嗪-1-基)苯胺可制得目标化合物6-(2,6-二氯苯基)-2-((4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮。
本发明的一个重要方面是发现了式I(包括本文所述的式II和式III化合物)是激酶抑制剂,特别是Wee1激酶抑制剂。因此,这些化合物可用于治疗Wee1相关疾病,例如癌症。
本发明还包括给动物施用有效量的式I、式II或式III化合物或其可药用盐或前药的治疗方法。其中所述治疗方法用于治疗激酶相关疾病,特别是Wee1激酶相关疾病,例如癌症。可由本发明的方法或药物组合物治疗或预防的这类疾病包括但不限于肝癌、黑素瘤、霍奇金病、非霍奇金淋巴瘤、急性淋巴白血病、慢性淋巴白血病、多发性骨髓瘤、成神经细胞瘤、乳腺癌、卵巢癌、肺癌、维尔姆斯瘤、子宫颈癌、睾丸癌、软组织肉瘤、原发性巨球蛋白血症、膀胱癌、慢性粒细胞白血病、原发性脑癌、恶性黑素瘤、小细胞肺癌、胃癌、结肠癌、恶性胰腺胰岛瘤、恶性类癌性癌症、绒毛膜癌、蕈樣肉芽腫、头颈癌、骨原性肉瘤、胰腺癌、急性粒细胞白血病、毛细胞白血病、横纹肌肉瘤、卡波西肉瘤、泌尿生殖系统肿瘤病、甲状腺癌、食管癌、恶性高钙血症、子宫颈增生症、肾细胞癌、子宫内膜癌、真性红细胞增多症、特发性血小板增多症、肾上腺皮质癌、皮肤癌和前列腺癌。
本发明也包括用于治疗或预防因激酶(特别是Wee1)活性异常而引起的其他疾病,例如神经病学或神经精神疾病或病症,例如抑郁症患者。
在实施本发明治疗方法时,给有一种或多种这些症状的病人施用有效量的药物制剂。所述药物制剂含有有效治疗浓度的式I、式II或式III化合物,被配制成用于口服、静脉注射、局部或外用给药的形式,用于治疗癌症和其他疾病。给药量是有效地改善或消除一个或多个病症的药量。对于特定疾病的治疗,有效量是足以改善或以某些方式减轻与疾病有关的症状的药量。这样的药量可作为单一剂量施用,或者可依据有效的治疗方案给药。给药量也许可治愈疾病,但是给药通常是为了改善疾病的症状。一般需要反复给药来实现所需的症状改善。
在另一个实施方案中提供了一种药用组合物,其中含有激酶抑制剂的式I、式II或式III化合物或其可药用盐与可药用载体。
本发明另一个实施方案涉及能有效地治疗癌症的药用组合物,其中包含激酶抑制剂的式I、式II或式III化合物,或其可药用盐或前药,与至少一种已知的抗癌药物或抗癌药物的可药用盐联合共用。特别是和其他与DNA损伤和修复机理有关的抗癌药物的联合共用,包括PARP抑制剂奥拉帕尼、Niraparib、Rucaparib和Talazoparib;HDAC抑制剂伏立诺他、罗咪地辛、帕比司他和贝利司他;等等。以及和其他与细胞分裂检测点有关的抗癌药物的联合共用,包括Chk1/2抑制剂,CDK4/6抑制剂如帕博西尼,ATM/ATR抑制剂等等。其他可用于抗癌联合治疗的已知抗癌药物包括但不限于烷化剂例如白消安、马法兰、苯丁酸氮芥、环磷酰胺、异环磷酰胺、替莫唑胺、苯达莫司汀、顺铂、丝裂霉素C、博莱霉素和卡铂;拓扑异构酶Ⅰ抑制剂例如喜树碱、伊立替康和托泊替康;拓扑异构酶Ⅱ抑制剂例如阿霉素、表阿霉素、阿克拉霉素、米托蒽醌、甲基羟基玫瑰树碱和铭托泊普;RNA/DNA抗代谢物例如5-氮杂胞苷、吉西他滨、5-氟尿嘧啶和甲氨蝶呤;DNA抗代谢物例如5-氟-2′-去氧尿苷、氟达拉滨,奈拉滨、阿糖胞苷、普拉曲沙、培美曲塞、羟基脲和硫代鸟嘌呤;抗有丝分裂剂例如秋水仙碱、长春碱、长春新碱、长春瑞滨、紫杉醇,伊沙匹隆、卡巴他赛和多西他赛;抗体例如单抗,帕尼单抗、耐措妥珠单抗、纳武单抗、派姆单抗、雷莫芦单抗、贝伐珠单抗、帕妥珠单抗、曲妥珠单抗、西妥昔单抗、奥滨尤妥珠单抗、奥法木单抗、利妥昔单抗、阿仑单抗、替伊莫单抗、托西莫单抗、本妥昔单抗、达雷木单抗、埃罗妥珠单抗、T-DM1、Ofatumumab、Dinutuximab、Blinatumomab、易普利姆玛、阿瓦斯丁、赫赛汀和美罗华;激酶抑制剂例如伊马替尼、吉非替尼、厄洛替尼、奥斯替尼、阿法替尼、赛立替尼、艾乐替尼、克唑替尼、埃罗替尼、拉帕替尼、索拉非尼、瑞格非尼、维罗非尼、达拉非尼、阿柏西普、舒尼替尼、尼罗替尼、达沙替尼、博舒替尼、普拉替尼、依鲁替尼、卡博替尼、乐伐替尼、凡德他尼、曲美替尼、卡比替尼、阿昔替尼、替西罗莫司、Idelalisib、帕唑帕尼、特癌适和依维莫司。其他可用于抗癌组合治疗的已知抗癌药物包括他莫昔芬、来曲唑、氟维司群、米托胍腙、奥曲肽、视黄酸、砒霜、唑来膦酸、硼替佐米、卡非佐米、Ixazomib、维莫德吉、索尼德吉、狄诺塞麦、萨力多胺、来那度胺、Venetoclax、Aldesleukin(重组人白介素-2)和Sipueucel-T(前列腺癌治疗疫苗)。
在实施本发明的方法时,本发明化合物与至少一种已知的抗癌药物可作为单一的药用组合物一起给药。另外,本发明化合物也可与至少一种已知抗癌药分开给药。在一个实施方案,本发明化合物和至少一种已知的抗癌药差不多同时给药,即所有的药物同时施用或陆续施用,只要化合物在血液中同时达到治疗浓度即可。在另外一个实施方案,本发明的化合物和至少一种已知的抗癌药根据各自的剂量方案给药,只要化合物在血液中达到治疗浓度即可。
本发明的另一个实施方案,是一种由所述化合物组成的能有效的抑制肿瘤的,作为激酶抑制剂的生物耦合物。这个能抑制肿瘤的生物耦合物由所述化合物与至少一种已知的有医疗作用的抗体,如赫赛汀或美罗华,或生长素,如DGF或NGF,或细胞激素,如白细胞介素2或4,或任意能与细胞表面结合的分子组成。该抗体与其他分子能把所述化合物递送到其靶点,使之成为有效的抗癌药物。此生物耦合物也可以提高有医疗作用的抗体,如赫赛汀或美罗华的抗癌效果。
本发明的另一实施例涉及一种能有效的抑制肿瘤的药用组合物,包含式I、式II或式III所示的激酶抑制剂,或其可用药盐或前药,与放射疗法联合治疗。在此实施例,本发明化合物与放射治疗可在相同时间或不同时间给药。
本发明的另一实施例涉及一种能有效的用于癌症手术后治疗的药用组合物,包含式I、式II或式III所示的激酶抑制剂,或其可用药盐或前药。本发明还涉及用手术切除肿瘤,然后用本发明的药用组合物治疗该哺乳动物的癌症的治疗方法。
本发明的药用组合物包括所有本发明化合物的含有量能有效地实现其预期目标的药品制剂。虽然每个人的需求各不相同,本领域技术人员可确定药品制剂中每个部分的最佳剂量。一般情况下,所述化合物,或其可用药盐,对哺乳动物每天口服给药,药量按照约0.0025到50毫克/公斤体重。但最好是每公斤口服给药约0.01到10毫克/公斤。如果也施用一个已知的抗癌药物,其剂量应可有效地实现其预期的目的。这些已知的抗癌药物的最佳剂量是本领域技术人员所熟知的。
单位口服剂量可以包括约0.01到50毫克,最好是约0.1到10毫克的本发明化合物。单位剂量可给予一次或多次,每天为一片或多片,每片含有约0.1到50毫克,合宜地约0.25到10毫克的本发明化合物或其溶剂化物。
在外用制剂中,本发明化合物的浓度可以是每克载体约0.01到100毫克。
本发明化合物可作为未加工药品给药。本发明化合物也可以作为含有可药用载体(包括辅料和助剂)的一个合适的药物制剂的一部分给药。这些可药用载体有利于把化合物加工成可药用的药物制剂。优选的药物制剂,特别是那些口服的和优选的给药方式类型,如片剂,锭剂和胶囊,以及适合于注射或口服的溶液,包含约0.01%到99%,最好从约0.25%到75%的活性化合物以及辅料。
本发明的范围也包括本发明化合物的无毒性可药用盐。酸加成盐由混合一个无毒性可药用酸溶液和本发明的化合物溶液而形成。所述酸例如盐酸,富马酸,马来酸,琥珀酸,乙酸,柠檬酸,酒石酸,碳酸,磷酸,草酸等。碱加成盐由混合一个无毒性可药用碱溶液和本发明的化合物溶液而形成。所述碱例如氢氧化钠,氢氧化钾,氢胆碱,碳酸钠,三羟甲基氨基甲烷,N-甲基-葡萄糖胺等。
本发明的药物制剂可以给予任何哺乳动物,只要他们能获得本发明化合物的治疗效果。在这些哺乳动物中最为重要的是人类和兽医动物,虽然本发明不打算如此受限。
本发明的药物制剂可通过任何途径给药以达到其预期目的。例如,可以通过肠外,皮下,静脉,肌肉,腹腔内,透皮,口腔,鞘内,颅内,鼻腔或外用途径给药。作为替代或并行地,可以通过口服给药。药的剂量将根据病人的年龄,健康与体重,并行治疗的种类,治疗的频率,以及所需治疗效益来决定。
本发明的药物制剂可用已知的方式制造。例如,由传统的混合,制粒,制锭,溶解,或冷冻干燥过程制造。制造口服制剂时,可结合固体辅料和活性化合物,选择性研磨混合物。如果需要或必要时加入适量助剂后,加工颗粒混合物,获得片剂或锭剂芯。
合适的辅料特别是填料,例如糖类如乳糖或蔗糖,甘露醇或山梨醇;纤维素制剂和/或钙磷酸盐,例如磷酸三钙或磷酸氢钙;以及粘结剂,例如淀粉糊,包括玉米淀粉,小麦淀粉,大米淀粉,马铃薯淀粉,明胶,黄芪胶,甲基纤维素,羟丙基甲基纤维素,羧甲基纤维素钠,和/或聚乙烯吡咯烷酮。如果需要,可增加崩解剂,比如上面提到的淀粉,以及羧甲基淀粉,交联聚乙烯吡咯烷酮,琼脂,或褐藻酸或其盐,如海藻酸钠。辅助剂特别是流动调节剂和润滑剂,例如,硅石,滑石,硬脂酸或其盐,如硬脂酸镁或硬脂酸钙,和/或聚乙二醇。如果需要,可以给锭剂核芯提供可以抵抗胃液的合适包衣。为此,可以应用浓缩糖类溶液。这个溶液可以含有阿拉伯树胶,滑石,聚乙烯吡咯烷酮,聚乙二醇和/或二氧化钛,漆溶液和合适的有机溶剂或溶剂混合物。为了制备耐胃液的包衣,可使用适当的纤维素溶液,例如醋酸纤维素邻苯二甲酸或羟丙基甲基纤维素邻苯二甲酸。可向药片或锭剂核芯的包衣加入染料或色素。例如,用于识别或为了表征活性成分剂量的组合。
其他可口服的药物制剂包括明胶制成的压接式胶囊,以及用明胶和甘油或山梨醇等增塑剂制成的密封软胶囊。该压接式胶囊可含有颗粒形式的活性化合物,与填料例如乳糖;粘结剂例如淀粉;和/或润滑剂例如滑石粉或硬脂酸镁,以及稳定剂混合而成。在软胶囊,活性化合物最好是溶解或悬浮在适当的液体例如油脂或液体石蜡中,其中可加入稳定剂。
合适于肠外给药的制剂包括活性化合物的水溶液,如水溶性盐的溶液和碱性溶液。此外,可施用适当的活性化合物的油性注射悬浮液。合适的亲脂性溶剂或载体包括油脂例如香油,合成脂肪酸酯例如油酸乙酯或甘油三酯或聚乙二醇400,或氢化蓖麻油,或环糊精。水性注射悬浮液可含有增加悬浮液黏度的物质,例如羧甲基纤维素钠,山梨醇,和/或葡聚糖。也可以含有悬浮稳定剂。
按照本发明的一个方面,本发明的化合物采用外用和肠外配方,并用于治疗皮肤癌。
本发明的外用制剂可通过优选合适的载体来制成油剂,霜剂,乳液剂,药膏等。合适的载体包括植物或矿物油,白矿油(白软石蜡),支链脂肪或油脂,动物脂肪和高分子醇(大于C12)。优选的载体是活性成分能溶解在其中的那些载体。也可包括乳化剂,稳定剂,保湿剂和抗氧化剂,以及如果需要的话,给予颜色或香味的试剂。此外,这些外用制剂可包含透皮渗透增强剂。这种增强剂的例子可参见美国专利号3,989,816和4,444,762。
霜剂优选用矿物油,自乳化蜂蜡和水的混合物配制,与溶解于少量油例如杏仁油的活性成分混合而成。一个典型的霜剂例子包括约40份水,20份蜂蜡,40份矿物油和1份杏仁油。
药膏可以这样配制,将含有活性成分的植物油例如杏仁油和温热的软石蜡混合,然后使该混合物冷却。一个典型的药膏例子包括约30%重量的杏仁油和70%重量的白软石蜡。
本发明也涉及应用本发明的化合物制备治疗对抑制激酶(特别是Wee1)活性有效果的临床病症的药物。这些药物可包括上述药用组合物。
下列实施例是举例说明,而不是限制本发明的方法和制剂。其他对于本领域技术人员来说是显而易见的,和在临床治疗中通常会遇到的对各种条件和参数的适当修改和改进,都在本发明的精神和范围内。
实施例
一般性说明
所用试剂均是商品品质,溶剂均按照标准方法干燥纯化。使用电喷雾的单四级杆质谱仪(平台Ⅱ,安捷伦6110)分析质谱样品。使用Brücker Ascend 400核磁仪在400MHz记录1H NMR光谱,化学位移记录为以TMS作为内标(0.00ppm)从低场始以ppm为单位,耦合常数J值以Hz为单位。
实施例1
6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮
a)(2-(3-(2-氯苯基)脲基)乙基)氨基甲酸叔丁酯:在0℃下,将邻氯苯异氰酸酯(500mg,3.26mmol)缓慢加入到N-叔丁氧羰基-1,2-乙二胺(1.56g,9.76mmol)的乙醚 (20mL)溶液中。反应液在室温下搅拌3小时后,过滤,滤饼用少量乙醚洗涤,抽干后得到目标化合物(2-(3-(2-氯苯基)脲基)乙基)氨基甲酸叔丁酯(622mg,62%收率,白色固体)。LC-MS(ESI):m/z(M+1)314.37。
b)1-(2-氨基乙基)-3-(2-氯苯基)脲(盐酸盐):将(2-(3-(2-氯苯基)脲基)乙基)氨基甲酸叔丁酯(622mg,1.98mmol)溶于二氯甲烷(20mL)中,加入盐酸二氧六环溶液(4N,20mL)。反应液在室温下搅拌4小时后,减压除去溶剂,得到目标化合物1-(2-氨基乙基)-3-(2-氯苯基)脲(盐酸盐)(470mg,95%收率,白色固体)。LC-MS(ESI):m/z(M+1)214.42。
c)4-((2-(3-(2-氯苯基)脲基)乙基)氨基)-2-(甲硫基)嘧啶-5-甲酸乙酯:将1-(2-氨基乙基)-3-(2-氯苯基)脲(盐酸盐)(200mg,0.8mmol),4-氯-2-(甲硫基)嘧啶-5-甲酸乙酯(284mg,1.22mmol)和二异丙基乙基胺(485mg,3.75mmol)混合于乙腈(8mL)中。氮气保护,在80℃下搅拌反应3小时后冷却至室温,抽滤,滤饼用少量甲醇洗涤,固体抽干后得到目标化合物4-((2-(3-(2-氯苯基)脲基)乙基)氨基)-2-(甲硫基)嘧啶-5-甲酸乙酯(270mg,82%收率,白色固体)。LC-MS(ESI):m/z(M+1)410.33。
d)6-(2-氯苯基)-2-(甲硫基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮:将4-((2-(3-(2-氯苯基)脲基)乙基)氨基)-2-(甲硫基)嘧啶-5-甲酸乙酯(200mg,0.49mmol)溶解于三氯氧磷(8mL)中,加热至110℃下反应过夜。减压除去溶剂后加入乙酸乙酯稀释,用饱和碳酸氢钠水溶液洗,有机相用饱和食盐水洗,无水硫酸钠干燥,过滤,减压浓缩得到粗产物。柱层析分离纯化(硅胶,石油醚:乙酸乙酯=1:1为洗脱剂),得到目标化合物6-(2-氯苯基)-2-(甲硫基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(92mg,54%收率,黄色固体)。LC-MS(ESI):m/z(M+1)346.37。
e)6-(2-氯苯基)-2-(甲磺酰基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮:将6-(2-氯苯基)-2-(甲硫基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(92mg,0.27mmol)溶于氯仿(10mL)中,加入间氯过氧苯甲酸(80%,122mg,0.56mmol)。反应液在室温下搅拌4小时后,加入饱和硫代硫酸钠水溶液淬灭反应,乙酸乙酯萃取分液。有机相用饱和食盐水洗,无水硫酸钠干燥,过滤,减压浓缩得到粗产物。柱层析分离纯化(硅胶,二氯甲烷:甲醇=20:1为洗脱剂),得到目标化合物(54mg,53%收率,浅黄色固体)。LC-MS(ESI):m/z(M+1)378.31。
f)6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮:将6-(2-氯苯基)-2-(甲磺酰基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(54mg,0.14mmol)和4-(4-甲基哌嗪)苯胺(27mg,0.14mmol)混合于异丙醇(2mL)中。反应液在80℃下搅拌1小时后,减压除去溶剂得到粗产物。用制备液相色谱(C18柱,0-100%乙腈/水为流动相)分离纯化,得到目标化合物(12mg,18%收 率,白色固体)。LC-MS(ESI):m/z(M+1)489.39。1H NMR(400MHz,DMSO-d6):δ8.61(s,1H),8.24–8.13(m,2H),7.69–7.62(m,2H),7.53–7.45(m,3H),6.96–6.88(m,2H),4.17–4.07(m,2H),3.78(t,J=8.8Hz,2H),3.13–3.07(m,4H),2.48–2.42(m,4H),2.23(s,3H)。
实施例2
6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮
应用类似于所描述的实施例1的合成方法制得,起始原料为1-氯-3-氟-2-异氰酸苯酯、N-叔丁氧羰基-1,2-乙二胺、4-氯-2-(甲硫基)嘧啶-5-甲酸乙酯和4-(4-甲基哌嗪)苯胺,纯化后得到目标化合物6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(58mg,38%收率,白色固体)。LC-MS(ESI):m/z(M+1)507.36。1H NMR(400MHz,DMSO-d6):δ8.64(s,1H),8.17–8.16(m,1H),7.74–7.51(m,4H),7.48–7.43(m,1H),6.92(d,J=8.9Hz,2H),4.19–4.11(m,2H),3.84–3.77(m,2H),3.15–3.05(m,4H),2.49–2.45(m,4H),2.24(d,J=5.7Hz,3H)。
实施例3
4-(2-氯苯基)-8-((4-(4-甲基哌嗪-1-基)苯基)氨基)-1,2-二氢咪唑并[1,2-a]吡啶并[3,4-e]嘧啶-5(4H)-酮
a)8-氯-4-(2-氯苯基)-1,2-二氢咪唑并[1,2-a]吡啶并[3,4-e]嘧啶-5(4H)-酮:依次应用类似于所描述的实施例1c和1d的合成方法制得,起始原料为1-(2-氨基乙基)-3-(2-氯苯基)脲(盐酸盐)和4,6-二氯烟酸甲酯,纯化后得到目标化合物8-氯-4-(2-氯苯基)-1,2-二氢咪唑并[1,2-a]吡啶并[3,4-e]嘧啶-5(4H)-酮(173mg,66%收率,浅黄色固体)。LC-MS(ESI):m/z(M+1)333.26。
b)4-(2-氯苯基)-8-((4-(4-甲基哌嗪-1-基)苯基)氨基)-1,2-二氢咪唑并[1,2-a]吡啶并[3,4-e]嘧啶-5(4H)-酮:将8-氯-4-(2-氯苯基)-1,2-二氢咪唑并[1,2-a]吡啶并[3,4-e]嘧啶-5(4H)-酮(120mg,0.36mmol),4-(4-甲基哌嗪)苯胺(83mg,0.43mmol),三(二亚苄基丙酮)二钯(33mg,0.036mmol),2-二环己基磷-2,4,6-三异丙基联苯(34mg,0.072mmol)和磷酸钾(229mg,1.08mmol)混合于叔丁醇(4mL)和水(1mL)中。反应液在氮气保护下在80℃下搅拌过夜后,减压除去溶剂得到粗产物,用制备液相色谱(C18柱,0-100%乙腈/水为流动相)分离纯化,得到目标化合物4-(2-氯苯基)-8-((4-(4-甲基哌嗪-1-基)苯基)氨基)-1,2-二氢咪唑并[1,2-a]吡啶并[3,4-e]嘧啶-5(4H)-酮(55mg,31%收率,白色固体)。LC-MS(ESI):m/z(M+1)488.40。1H NMR(400MHz,DMSO-d6):δ9.37(s,1H),8.49 (s,1H),7.64–7.59(m,1H),7.51–7.37(m,5H),6.96–6.88(m,2H),5.95(s,1H),4.01–3.89(m,2H),3.80–3.72(m,2H),3.12–3.06(m,4H),2.48–2.43(m,4H),2.23(s,3H)。
实施例4
6-(2,6-二氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮
a)2-(甲磺酰基)-6-(2,6-二氯苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮:应用类似于所描述的实施例1a-1e的合成方法制得,起始原料为N-叔丁氧羰基-1,2-乙二胺、4-氯-2-(甲硫基)嘧啶-5-甲酸乙酯和1,3-二氯异氰酸苯酯。
b)6-(2,6-二氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮:室温下将2-(甲磺酰基)-6-(2,6-二氯苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(40mg,0.1mmol)和4-(4-甲基哌嗪-1-基)苯胺(30mg,0.15mmol)溶于乙腈(1mL)中,加入三氟乙酸(0.2mL)。反应液在室温下搅拌过夜后,减压浓缩得到粗产物,用制备液相色谱(C18柱,0-100%乙腈/水为流动相)分离纯化,得到目标化合物(18mg,36%收率,黄色固体)。LC-MS(ESI):m/z(M/2+1)263.16。1H NMR(400MHz,DMSO-d6):δ10.27(s,1H),8.65(s,1H),7.75–7.59(m,3H),7.61–7.41(m,2H),6.93(d,J=8.9Hz,2H),4.23–4.08(m,2H),3.87–3.75(m,2H),3.19–2.99(m,4H),2.49–2.45(m,4H),2.24(s,3H)。
应用类似于所描述的实施例1的合成方法制得下述实施例6-8的化合物,起始原料为2-(甲磺酰基)-6-(2-氯-6-氟苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(应用类似于实施例1a-1e的合成方法制得,起始原料为N-叔丁氧羰基-1,2-乙二胺、4-氯-2-(甲硫基)嘧啶-5-甲酸乙酯和1-氯-3-氟-2-异氰酸苯酯)和相应的取代苯胺或取代四氢异喹啉胺。
应用类似于所描述的实施例4b的合成方法可制得下述实施例5、9—26的化合物,起始原料为2-(甲(亚)磺酰基)-6-取代基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(应用类似于实施例1a-1e的合成方法制得,起始原料为N-叔丁氧羰基-1,2-乙二胺、4-氯-2-(甲硫基)嘧啶-5-甲酸乙酯和相应的取代异氰酸苯酯)和相应的取代胺。
Figure PCTCN2017111230-appb-000018
Figure PCTCN2017111230-appb-000019
Figure PCTCN2017111230-appb-000020
Figure PCTCN2017111230-appb-000021
Figure PCTCN2017111230-appb-000022
Figure PCTCN2017111230-appb-000023
应用类似于所描述的实施例1a-e和4b的合成方法制得实施例27和28的化合物,起始原料为1-氯-3-氟-2-异氰酸苯酯、(1-氨基-2-甲基丙-2-基)氨基甲酸叔丁酯或2-氨基-2-甲基丙基氨基甲酸叔丁酯、4-氯-2-(甲硫基)嘧啶-5-甲酸乙酯和4-(4-甲基哌嗪)苯胺。
实施例27
6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,8-二甲基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮
白色固体(60mg,63%收率)。LC-MS(ESI):m/z(M+1)535.28。1H NMR(400MHz,DMSO-d6)δ10.25(br s,1H),8.64(s,1H),7.76-7.59(m,2H),7.58–7.49(m,2H),7.49–7.42(m,1H),6.94(d,J=8.5Hz,2H),3.93–3.81(m,2H),3.10(t,J=4.6Hz,4H),2.45(t,J=4.8Hz,4H),2.22(s,3H),1.23(d,J=4.4Hz,6H)。
实施例28
6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-9,9-二甲基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮
黄色固体(10mg,19%收率)。LC-MS(ESI):(M+1)535.24。1H NMR(400MHz, DMSO-d6)δ10.17(br s,1H),8.66(s,1H),7.60–7.42(m,5H),6.94(d,J=8.9Hz,2H),3.55(s,2H),3.11(t,J=4.8Hz,4H),2.47(t,J=4.8Hz,4H),2.23(s,3H),1.76–1.57(m,6H)。
实施例29
2-甲磺酰基-6-(嘧啶-2-基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮
a)(4-硝基苯基-嘧啶-2-基)氨基甲酸酯:将2-氨基嘧啶(1g,10.5mmol)溶于无水二氯甲烷(30mL)中,加入吡啶(1.66mL,21mmol)。在0℃下,滴入对硝基苯基氯甲酸酯(2.1g,10.5mmol)的无水二氯甲烷(15mL)溶液。滴加完毕后,反应液在0℃下搅拌反应10min,抽滤,滤饼用二氯甲烷洗涤,干燥得到目标化合物(1.6g,60%收率,白色固体)。LC-MS(ESI):m/z[M+H]+261.08。
b)(2-(3-(嘧啶-2-基)脲基)乙基)氨基甲酸叔丁酯:将(4-硝基苯基-嘧啶-2-基)氨基甲酸酯(1g,3.8mmol)和(2-氨基乙基)氨基甲酸叔丁酯(1.85g,11.5mmol)加入到无水四氢呋喃(30mL)中,反应混合物在微波反应器中80℃反应1h。冷却至室温,减压浓缩得到粗产物。柱层析分离纯化(硅胶,二氯甲烷:甲醇=10:1为洗脱剂),得到目标化合物(760mg,70%收率,黄色固体)。LC-MS(ESI):m/z[M+H]+282.21。
c)2-甲磺酰基-6-(嘧啶-2-基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮:应用类似于实施例1b-1e的合成方法制得,起始原料为(2-(3-(嘧啶-2-基)脲基)乙基)氨基甲酸叔丁酯和4-氯-2-(甲硫基)嘧啶-5-甲酸乙酯,纯化后得到浅黄色固体。LC-MS(ESI):m/z[M+H]+346.11。
实施例30
2-氟-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯胺
a)(2S,6R)-1,2,6-三甲基-4-(3-氟-4-硝基苯基)哌嗪:室温下,将(2S,6R)-1,2,6-三甲基哌嗪(403mg,3.14mmol)溶于乙腈(10mL)中,加入N,N-二异丙基乙基胺(0.82mL,4.71mmol)和2,4-二氟硝基苯(500mg,3.14mmol)。反应液在80℃下搅拌反应3h后,冷却至室温,用乙酸乙酯(20mL)与水(20mL)萃取分液。有机相用饱和食盐水洗,无水硫酸钠干燥,过滤,减压浓缩得到粗产物。柱层析分离纯化(硅胶,二氯甲烷:甲醇=20:1为洗脱剂)得到目标化合物(268mg,32%收率,黄色固体)。LC-MS(ESI):m/z[M+H]+268.21。
b)2-氟-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯胺:室温下,向溶有(2S,6R)-1,2,6-三甲基-4-(3-氟-4-硝基苯基)哌嗪(268mg,1.0mmol)的乙酸乙酯(15mL)溶液中加入10%钯/碳(30mg)。反应液用氢气置换三次,室温氢化过夜后,抽滤除去钯/碳,滤液减压浓缩得到目标化合物(230mg,96%收率,黄色固体)。LC-MS(ESI):m/z[M+H]+ 238.32。
实施例31
2-氯-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯胺
室温下,向溶有(2S,6R)-1,2,6-三甲基-4-(3-氯-4-硝基苯基)哌嗪(应用类似于实施例30a的合成方法制得,起始原料为(2S,6R)-1,2,6-三甲基哌嗪和2-氯-4-氟-1-硝基苯。200mg,0.71mmol)的乙醇/水(10/2.5mL)溶液中加铁粉(185mg,3.31mmol)和氯化铵(188mg,3.52mmol)。反应液用氮气置换三次,50℃下搅拌反应4h后冷却至室温,抽滤除去铁粉,滤液减压浓缩。粗产物溶于乙酸乙酯(20mL)和水(10mL),分液,有机相用饱和食盐水洗,无水硫酸钠干燥,过滤,减压浓缩得到粗产物。柱层析分离纯化(硅胶,二氯甲烷:甲醇=15:1为洗脱剂)得到目标化合物(160mg,89%收率,黄色油状物)。LC-MS(ESI):m/z[M+H]+254.24。
其它的取代胺可应用类似于上述实施例30或实施例31的合成方法制得,或者由本领域技术人员按已知方法合成制得。
应用类似于所描述的实施例1的合成方法制得下述实施例32-33、36、54-56和85-87的化合物,起始原料为2-(甲磺酰基)-6-取代基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(应用类似于实施例1a-1e的合成方法制得,起始原料为N-叔丁氧羰基-1,2-乙二胺、4-氯-2-(甲硫基)嘧啶-5-甲酸乙酯和相应的取代异氰酸苯酯)和相应的取代苯胺。
应用类似于所描述的实施例4b的合成方法制得下述实施例34-35、37-53、57-84和88-92的化合物,起始原料为2-(甲(亚)磺酰基)-6-取代基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(应用类似于实施例1a-1e的合成方法制得,起始原料为N-叔丁氧羰基-1,2-乙二胺、4-氯-2-(甲硫基)嘧啶-5-甲酸乙酯和相应的取代异氰酸苯酯;实施例29)和相应的取代胺。
Figure PCTCN2017111230-appb-000024
Figure PCTCN2017111230-appb-000025
Figure PCTCN2017111230-appb-000026
Figure PCTCN2017111230-appb-000027
Figure PCTCN2017111230-appb-000028
Figure PCTCN2017111230-appb-000029
Figure PCTCN2017111230-appb-000030
Figure PCTCN2017111230-appb-000031
Figure PCTCN2017111230-appb-000032
Figure PCTCN2017111230-appb-000033
Figure PCTCN2017111230-appb-000035
Figure PCTCN2017111230-appb-000036
Figure PCTCN2017111230-appb-000037
应用类似于所描述的实施例1的合成方法制得下述实施例102、103和105—115的化合物,起始原料为2-(甲(亚)磺酰基)-6-取代基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(应用类似于实施例1a-1e的合成方法制得,起始原料为N-叔丁氧羰基-1,2-乙二胺、4-氯-2-(甲硫基)嘧啶-5-甲酸乙酯和相应的取代异氰酸苯酯)和相应的取代苯胺。
应用类似于所描述的实施例4b的合成方法制得下述实施例93—101、104和116—184的化合物,起始原料为2-(甲(亚)磺酰基)-6-取代基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(应用类似于实施例1a-1e的合成方法制得,起始原料为N-叔丁氧羰基-1,2-乙二胺、4-氯-2-(甲硫基)嘧啶-5-甲酸乙酯和相应的取代异氰酸苯酯)和相应的取代胺。
Figure PCTCN2017111230-appb-000038
Figure PCTCN2017111230-appb-000039
Figure PCTCN2017111230-appb-000040
Figure PCTCN2017111230-appb-000041
Figure PCTCN2017111230-appb-000042
Figure PCTCN2017111230-appb-000043
Figure PCTCN2017111230-appb-000044
Figure PCTCN2017111230-appb-000045
Figure PCTCN2017111230-appb-000046
Figure PCTCN2017111230-appb-000047
Figure PCTCN2017111230-appb-000048
Figure PCTCN2017111230-appb-000049
Figure PCTCN2017111230-appb-000050
Figure PCTCN2017111230-appb-000051
Figure PCTCN2017111230-appb-000052
Figure PCTCN2017111230-appb-000053
Figure PCTCN2017111230-appb-000054
Figure PCTCN2017111230-appb-000055
Figure PCTCN2017111230-appb-000056
Figure PCTCN2017111230-appb-000057
注:*实施例93化合物为合成实施例19目标化合物的副产物。
实施例186
6-(2,6-二氯苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)胺基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮二盐酸盐
将2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-6-(2,6-二氯苯基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例71,1.0克,1.8毫摩尔)溶于乙腈(5毫升)和纯净水(5毫升)中,加入盐酸(1N,3.6毫升)。将该混合物置于冻干机中冻干48小时后得到目标化合物(1.09克,96%收率,黄色固体)。LC-MS(ESI):m/z(M+1)557.16。1HNMR(400 MHz,DMSO-d6):δ10.64(brs,2.5H),8.82(s,1H),8.01–7.94(m,1H),7.82–7.68(m,3H),7.64–7.58(m,1H),7.24(d,J=8.8 Hz,1H),4.32–4.25(m,2H),3.91(t,J=8.6Hz,2H),3.54–3.47(m,2H),3.41–3.35(m,2H),3.23–3.15(m,2H),3.13–3.06(m,2H),2.84(d,J=4.7 Hz,3H)。
中间体:取代胺的合成
1)、4-(4-甲基-1,4-二氮杂环庚烷-1-基)苯胺
将N-甲基高哌嗪(414 mg,3.63 mmol)、对氟硝基苯(454 mg,3.22mmol)、碳酸钾(529 mg,3.83mmol)和DMSO(4 mL)加入50 mL单口瓶中,加热100℃反应4 h,停止反应。加入50 mL水搅拌,静置,抽滤得到1-甲基-4-(4-硝基苯基)-1,4-二氮杂环庚烷(黄色固体,500 mg,66%产率)。在50 mL反应瓶中加入1-甲基-4-(4-硝基苯基)-1,4-二氮杂环庚烷(500 mg,2.12 mmol)、铁粉(473 mg,8.48 mmol)、氯化铵(226 mg,4.24 mmol)、乙醇(10 mL)和水(10 mL)。然后加热回流1 h,停止反应。冷却,过滤,脱溶,然后用乙酸乙酯萃取(20 mL×3),有机相用无水硫酸钠干燥,过滤,脱溶得到粗产品,柱层析分离(甲醇:二氯甲烷=1:20)得到目标化合物(243 mg,56%收率)。
2)、4-(2-(二甲氨基)乙氧基)苯胺
将二甲氨基氯乙烷盐酸(1.44 g,10 mmol)和4-硝基苯酚(1.39 g,10 mmol)溶于20 mL DMF中,然后加入碳酸铯(5.29g,15mmol),加热100℃反应3 h。冷却,过滤, 脱溶,加入100mL水搅拌,用乙酸乙酯萃取,有机相用饱和碳酸钠洗涤,干燥,过滤,脱溶得到N,N-二甲基-2-(4-硝基酚氧基)乙胺的粗品(1.37g)。将得到的粗品(1.37g,6.5mmol)溶于乙醇(10mL)和水(10mL)中,然后加入铁粉(1.46g,26mmol)和氯化铵(696mg,13mmol),反应液加热回流2h。过滤,脱溶,用乙酸乙酯萃取,无水硫酸钠干燥,过滤,脱溶得粗产品。柱层析分离(甲醇:二氯甲烷=1:20)得到目标化合物(630mg,35%两步收率)。
3)、3,5-二甲基-4-(4-甲基哌嗪-1-基)苯胺
a)N-乙酰基-3,5-二甲基苯胺:在室温下,将3,5-二甲基苯胺(8g,66mmol)溶于无水二氯甲烷(120mL)中,加入三乙胺(18mL,132mmol)。在0℃下,将乙酸酐(8.09g,79mmol)加入到反应体系中,反应混合物在室温下搅拌反应2h后,加入饱和碳酸氢钠水溶液(50mL)。分液萃取,水相用二氯甲烷(50mL)萃取。合并有机相,用饱和食盐水洗,无水硫酸钠干燥,过滤,减压除去有机溶剂得到粗产品。柱层析分离纯化(硅胶,石油醚:乙酸乙酯=5:1为洗脱剂),得到目标化合物(10g,94%收率,白色固体)。LC-MS(ESI):m/z(M+H)+164.09。
b)N-乙酰基-3,5-二甲基-4-溴苯胺:在室温下,将N-乙酰基-3,5-二甲基苯胺(10g,61.3mmol)溶于二氯甲烷(400mL)和甲醇(160mL)的混合溶剂中,反应混合物在室温下搅拌反应1h后,加入四丁基三溴化铵(32.5g,67.4mmol)。反应混合物在室温下搅拌反应过液。减压浓缩除去有机溶剂,加入水(100mL)和乙酸乙酯(200mL)分液萃取,水相用乙酸乙酯(100mL)萃取。合并有机相,用饱和食盐水洗,无水硫酸钠干燥,过滤,减压除去有机溶剂得到粗产品。柱层析分离纯化(硅胶,石油醚:乙酸乙酯=5:1为洗脱剂),得到目标化合物(4.2g,28%收率,白色固体)。LC-MS(ESI):m/z(M+H)+242.09。
c)N-乙酰基-3,5-二甲基-4-(4-甲基哌嗪-1-基)苯胺:在室温下,将N-乙酰基-3,5-二甲基-4-溴苯胺(400mg,1.65mmol),N-甲基哌嗪(199mg,1.98mmol),三(二亚苄基丙酮)二钯(76mg,0.08mmol)和2-(二叔丁基膦)联苯(49mg,0.16mmol)加入到无水四氢呋喃(10mL)中,抽真空,置换氮气3次,加入双(三甲基硅基)氨基锂(1M,4.13mL)。反应混合物在100℃下微波反应1h后,冷却至室温。过滤,减压除去有机溶剂得到粗产品。柱层析分离纯化(硅胶,二氯甲烷:甲醇=15:1为洗脱剂),得到目标化合物(288mg,66%收率,黄色固体)。LC-MS(ESI):m/z(M+H)+262.41。
d)3,5-二甲基-4-(4-甲基哌嗪-1-基)苯胺:在室温下,将N-乙酰基-3,5-二甲基-4-(4-甲基哌嗪-1-基)苯胺(280mg,1.07mmol)溶于甲醇(10mL)中,加入浓盐酸(2.5mL)。反应混合物在90℃下搅拌反应1h后,冷却至室温,加入饱和碳酸氢钠水溶液调pH值至8-9。加入乙酸乙酯(15mL),分液萃取,水相用乙酸乙酯(15mL×2)萃取,合并有机相,用饱和食盐水洗,无水硫酸钠干燥,过滤,减压浓缩除去有机溶剂得到目标化合物(200mg,85%收率,黄色固体)。LC-MS(ESI):m/z(M+H)+220.21。
4)、3-氟-4-(4-甲基哌嗪-1-基)-5-甲基苯胺
a)1-(2-溴-4-硝基-6-氟苯基)-4-甲基哌嗪:在室温下,将3-氟-4-(4-甲基哌嗪-1-基)硝基苯(400mg,1.67mmol)加入到三氟乙酸(10mL)中,然后分别加入N-溴代丁二酰亚胺(NBS,595mg,3.34mmol)和浓硫酸(0.5mL)。反应混合物在50℃下搅拌反应过夜后,冷却至室温,减压除去三氟乙酸。加入饱和碳酸氢钠水溶液调pH至8-9,加入乙酸乙酯(30mL)。分液萃取,水相用乙酸乙酯(15mL×2)萃取,合并有机相,用饱和食盐水,无水硫酸钠干燥,过滤,减压除去有机溶剂得到粗产品。柱层析分离纯化(硅胶,二氯甲烷:甲醇=15:1为洗脱剂),得到目标化合物(480mg,90%收率,黄色固体)。LC-MS(ESI):m/z(M+H)+318.11。
b)1-(2-甲基-4-硝基-6-氟苯基)-4-甲基哌嗪:在室温下,将1-(2-溴-4-硝基-6-氟苯基)-4-甲基哌嗪(480mg,1.5mmol),甲基硼酸(906mg,15mmol),[1,1′-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(62mg,0.08mmol)和磷酸钾(964mg,4.5mmol)加入到二氧六环(10mL)和水(2mL)中。反应体系用氮气置换3次后,在100℃下搅拌反应过夜。冷却至室温,过滤,减压浓缩除去有机溶剂得到粗产品。柱层析分离纯化(硅胶,二氯甲烷:甲醇=15:1为洗脱剂),得到目标化合物(180mg,47%收率,黄色固体)。LC-MS(ESI):m/z(M+H)+254.21。
c)3-氟-4-(4-甲基哌嗪-1-基)-5-甲基苯胺:在室温下,将1-(2-甲基-4-硝基-6-氟苯基)-4-甲基哌嗪(180mg,0.71mmol)溶于乙酸乙酯(30mL)中,加入钯碳(20mg),反应体系用氢气置换3次。反应混合物在室温下搅拌反应3h后,过滤除去钯碳,减压除去有机溶剂得到目标化合物(149mg,94%收率,黄色固体)。LC-MS(ESI):m/z(M+H)+224.19。
5)、3-氯-4-(4-甲基哌嗪-1-基)-5-甲基苯胺
a)1-(2-氯-4-硝基-6-甲基苯基)-4-甲基哌嗪:将3-甲基-4-(4-甲基哌嗪-1-基)硝基苯(382mg,1.62mmol)加入到甲醇(5mL)中。将反应体系冷却至0℃,加入浓盐酸(2.5mL),缓慢滴加双氧水(2.5mL)。滴加完毕后,反应混合物在回流状态下搅拌反应过夜。将反应液冷却至室温,加入饱和亚硫酸钠水溶液(5mL),室温下搅拌反应10min后,加入饱和碳酸氢钠水溶液调pH至8-9。加入乙酸乙酯(20mL),分液萃取,水相用乙酸乙酯(15mL×2)萃取,合并有机相用饱和食盐水洗,无水硫酸钠干燥,过滤,减压浓缩除去有机溶剂得到粗产品。柱层析分离纯化(硅胶,二氯甲烷:甲醇=15:1为洗脱剂),得到目标化合物(406mg,93%收率,黄色固体)。LC-MS(ESI):m/z(M+H)+270.11。
b)3-氯-4-(4-甲基哌嗪-1-基)-5-甲基苯胺:将1-(2-氯-4-硝基-6-甲基苯基)-4-甲基哌嗪(120mg,0.44mmol)溶于乙醇(8mL)和水(2mL)的混合溶剂中,分别加入铁粉(125mg,2.22mmol)和氯化铵(238mg,4.4mmol)。反应混合物在80℃下搅拌反应4h后,冷却至室温。过滤,减压除去有机溶剂得到粗产品。柱层析分离纯化(硅胶,二氯甲烷:甲醇=15:1为洗脱剂),得到目标化合物(100mg,94%收率,黄色固体)。LC-MS(ESI):m/z(M+H)+240.11。
其它的取代胺可应用类似于上述中间体1)—5)的合成方法制得,或者由本领域技术人员按已知方法合成制得。
实施例187
应用Wee1激酶(人源)检测法测定6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮与其相似物对Wee1激酶的酶活性的抑制效应
在含20mM Tris/HCl pH 8.5,0.2mM EDTA,500μMLSNLYHQGKFLQTFCGSPLYRRR,10mM醋酸镁和10μM[γ-33P]-ATP的反应液中加入Wee1激酶(人源)孵育,而后加入50倍浓度的溶于100%DMSO中的待测化合物储液至终浓度10/1/0.1/0.01μM,混匀。加入Mg/ATP混合物启动反应,在室温下孵育40分钟后,加入磷酸溶液至终浓度0.5%淬灭反应。取10μL反应液滴到P30滤纸上用0.425%磷酸溶液洗4次之后用甲醇洗1次,干燥,液闪计数。每个化合物样品一式二份重复。实验阴性对照为缺少Wee1酶的所有组成部分,阳性为加入30%磷酸终止反应。在相同实验条件下对Wee1抑制剂AZD1775在10μM下进行了检测。表1汇总了化合物 的Wee1激酶抑制数据(计数和活性)。
表1.6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮与其相似物对Wee1激酶活性的抑制效应
Figure PCTCN2017111230-appb-000058
Figure PCTCN2017111230-appb-000059
Figure PCTCN2017111230-appb-000060
Figure PCTCN2017111230-appb-000061
Figure PCTCN2017111230-appb-000062
Figure PCTCN2017111230-appb-000063
因此,经Wee1激酶(人源)检测法测定,6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例1)与其相似物对Wee1激酶酶活性有好的抑制效应。其中10μM的实施例1和实施例2化合物对Wee1激酶的酶活性的抑制分别为96%和95%,活性与参考化合物AZD1775相似。实施例4在0.01μM时对Wee1激酶酶活性的抑制率达51%,显示实施例4对Wee1激酶有很高的抑制活性。
实施例188
对Wee1离解常数(Kd)的测定
测量化合物对Wee1亲和力的离解常数(Kd)是在DiscoveRx公司的KINOMEscanTM KdELECT平台上测定的。KINOMEscanTM实验是基于竞争性结合实验,通过qPCR精确测量通过和固定的配体竞争后结合在酶活性中心的化合物来计算其对Wee1的亲和性指标Kd的。
Wee 1激酶标记的T7噬菌体是用大肠杆菌BL21菌株制备的。生长期大肠杆菌(指数生长期)感染T7噬菌体后在32℃下振动孵育直到细胞溶解。将溶菌产物离心,过滤除去细胞碎片。部分序列的人Wee1(对应参考序列NP_003381.1的氨基酸M291到 K575)表达在T7噬菌体外壳上。一条短的DNA序列嵌入到T7噬菌体基因组内做为扩增子通过qPCR进行放大检测。
链霉亲和素包裹的磁珠(Dynal M280)与生物素标记的小分子配体在室温下反应30分钟作为用于结合试验的亲和树脂。结合配体的树脂用过量生物素封闭,用封闭缓冲液(SeaBlock(Pierce),1%BSA,0.05%Tween 20,1mM DTT)充分洗涤以去除未结合的配体来减少非特异性结合。
结合反应是将16μl噬菌体裂解物,3.8μl结合配体的树脂,和0.18μl待测化合物(PBS/0.05%Tween 20/10mM DTT/0.1%BSA/2μg/ml超声波降解的鲑鱼精液DNA)混合。待测化合物在100%DMSO中配制成111x的贮存溶液。Kd值由11个3倍系列稀释浓度的化合物和三个DMSO空白液来计算。所测化合物最高浓度为100,000nM,DMSO的终浓度为0.9%。所有反应在384孔的聚丙烯检测板中进行,每孔终体积为0.02mL。在室温下振动孵育1h,然后将树脂集中,用缓冲液(1x PBS,0.05%Tween 20)洗涤,除去被取代的激酶和待测化合物。洗涤后的树脂被重新悬浮在洗脱缓冲液(1x PBS,0.05%Tween 20,0.5μM无生物素标记的亲和配体)中,在室温下振动孵育30分钟。洗脱液中的激酶浓度通过qPCR检测。qPCR反应是将2.5μL含激酶的洗脱液加入到7.5μL含有0.15μM扩增子引物和0.15μM扩增子检测探针的qPCR反应液中。qPCR方案包含在95℃下的10分钟热启动,然后15秒95℃及1分钟60℃的35轮放大。实验中每个浓度重复两次。
每个化合物的Kd值用标准的剂量-响应曲线计算的Hill方程计算:响应=背景+(信号-背景)/(1+(Kd Hill斜率/剂量Hill斜率))。其中,Hill斜率设置为-1,曲线用非线性最小二乘方与Levenberg Marquardt算法拟合(Levenberg,K.,Q.Appl.Math.2,164-168(1944))。
计算出的Kd值显示了6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮与其相似物对Wee1激酶酶活性的抑制效应,汇总在表2中。
表2.6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮与其相似物对Wee1激酶酶活性的抑制效应(Kd值)
实施例 1 2
Kd(nM) 250 34
因此,经Kd的测定,6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例1)与其相似物对Wee1激酶酶活性有好的 抑制效应。
实施例189
应用MTT检测法测定6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮与其相似物对LoVo细胞增长的抑制作用
将新复苏的LoVo细胞培养传代至第三代后,且生长状态良好、融合度90%左右,开始用于实验。用胰酶消化LoVo细胞,800rpm离心5min,弃上清,用新鲜培基重悬,并计数,以6000个细胞每孔密度接种至96孔细胞培养板,置于37℃5%CO2培养箱培养过夜。受试物(包括待测化合物和参考化合物AZD1775)母液用DMSO按1:3和1:10比例分别进行连续系列稀释至8个浓度(最后一个浓度为DMSO阴性对照):10μM,3.3μM,1μM,0.33μM,0.1μM,0.033μM,0.01μM,0μM(DMSO终浓度为1‰)。每个浓度取5μL加入到120μL培基(25倍稀释),振荡混匀。取培养过夜的细胞,除去培养基,每孔加入195μL新鲜培基,再分别加入5μL稀释好的含相应浓度受试物的培基,随后将培养板置于37℃5%CO2培养箱培养3d。除去原液,每孔加100μL含MTT(0.5mg/mL)的新鲜无血清DMEM培基后,继续培养。4h后除去原液,每孔加入100μL DMSO,避光震荡10min,置于多功能读数仪读取552/630/690nm波长的吸光值(OD值)。用软件Graph Pad Prism 5.0分析数据,化合物对细胞增值的抑制活性以细胞存活率和化合物浓度为坐标绘图。IC50值以S形剂量反应曲线方程拟合,曲线方程为:Y=100/(1+10^(LogC-LogIC50)),其中C是化合物浓度。
表3汇总了化合物对LoVo细胞增长的抑制作用数据(IC50)。
表3.6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮与其相似物对LoVo细胞增长的抑制作用
实施例 1 2 3 4 5 6
IC50(μm) 3.575 0.4249 >100 0.1691 0.4307 0.3393
实施例 7 8 9 10 11 12
IC50(μm) 1.432 0.2018 0.1261 0.0887 0.1006 0.2186
实施例 13 14 15 16 17 18
IC50(μm) 0.07671 0.1491 1.728 4.755 0.2903 >1
实施例 19 20 21 22 23 27
IC50(μm) >100 >100 2.133 >100 >3 >100
实施例 28 32 33 34 35 36
IC50(μm) >1 0.8361 >100 0.3488 0.2615 >100
实施例 37 38 39 40 41 42
IC50(μm) >100 >100 >100 >100 >100 >100
实施例 43 44 45 46 47 48
IC50(μm) >100 0.0371 1.175 0.0261 0.0305 0.1174
实施例 49 50 51 52 53 54
IC50(μm) 0.0969 0.05893 0.1048 0.0874 0.0855 0.1547
实施例 55 56 57 58 59 60
IC50(μm) >100 0.0470 13.55 0.0900 >100 0.1585
实施例 61 62 63 64 65 66
IC50(μm) 0.7569 0.1935 0.0768 0.0965 0.1025 >100
实施例 67 68 69 70 71 72
IC50(μm) >100 >100 0.1048 0.1238 0.0443 0.0592
实施例 73 74 75 76 77 78
IC50(μm) 0.1938 0.1816 0.0402 0.0488 0.0524 0.0414
实施例 79 80 81 82 83 84
IC50(μm) >100 0.5393 0.1354 0.9464 2.899 3.058
实施例 85 86 87 88 89 90
IC50(μm) >100 >100 >100 >100 0.2200 1.308
实施例 91 92 93 94 95 96
IC50(μm) 0.1854 >100 >100 1.906 1.884 >100
实施例 97 98 99 100 101 102
IC50(μm) >100 >100 >100 0.1725 0.0954 >100
实施例 103 104 105 106 107 108
IC50(μm) 0.6776 0.4601 0.3727 0.3301 0.3605 1.502/
实施例 109 110 111 112 113 114
IC50(μm) 1.150 0.4715 0.8349 0.3926 0.4827 0.1049
实施例 115 116 117 118 119 120
IC50(μm) 0.2651 0.1232 0.0662 0.0811 0.0543 0.0359
实施例 121 122 123 124 125 126
IC50(μm) >100 0.0518 0.0391 0.0497 0.0264 0.0567
实施例 127 128 129 130 131 133
IC50(μm) 0.0423 0.0688 0.0703 0.6240 0.1873 0.1651
实施例 136 137 138 139 140 141
IC50(μm) 0.2139 0.1136 0.1986 0.0456 0.0582 0.03732
实施例 142 143 144 145 146 147
IC50(μm) 0.0514 0.0206 0.0615 0.0849 0.0833 0.0911
实施例 148 149 150 151 152 153
IC50(μm) 0.0398 0.0594 0.1158 0.0308 0.1387 0.0563
实施例 154 155 156 157 158 159
IC50(μm) 0.0633 0.0741 0.0627 0.0386 0.0585 0.05221
实施例 160 161 162 163 164 165
IC50(μm) 0.0273 0.0933 0.0652 0.0625 0.1010 0.0241
实施例 166 167 168 169 170 171
IC50(μm) 0.0555 0.0627 0.2956 0.1649 0.07690 0.2281
实施例 172 173 174 175 176 177
IC50(μm) 0.1170 0.0992 0.1357 0.1920 0.0736 0.05948
实施例 178 179 180 181 182 183
IC50(μm) 0.2806 2.862 2.383 0.1418 0.2426 0.7754
实施例 184 186 AZD1775      
IC50(μm) 1.489 0.04235 0.3889      
因此,经MTT检测法测定,6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例1)与其相似物对LoVo细胞增长有抑制作用。其中多个实施例,例如实施例75,对LoVo细胞增长的抑制作用比AZD1775强。
实施例190
应用MTT检测法测定6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮与其相似物对NCI-H1299细胞增长的抑制作用
将新复苏的NCI-H1299细胞培养传代至第三代后,且生长状态良好、融合度90%左右,开始用于实验。用胰酶消化NCI-H1299细胞,800 rpm离心5 min,弃上清,用新鲜培基重悬,并计数,以1000个细胞每孔密度接种至96孔细胞培养板,置于37℃5%CO2培养箱培养过夜。受试物(包括待测化合物和参考化合物AZD1775)母液用DMSO按 1:3和1:10比例分别进行连续系列稀释至8个浓度(最后一个浓度为DMSO阴性对照):10μM,3.3μM,1μM,0.33μM,0.1μM,0.033μM,0.01μM,0μM(DMSO终浓度为1‰)。每个浓度取5μL加入到120μL培基(25倍稀释),振荡混匀。取培养过夜的细胞,除去培养基,每孔加入195μL新鲜培基,再分别加入5μL稀释好的含相应浓度受试物的培基,随后将培养板置于37℃5%CO2培养箱培养3d。除去原液,每孔加100μL含MTT(0.5mg/mL)的新鲜无血清DMEM培基后,继续培养。4h后除去原液,每孔加入100μL DMSO,避光震荡10min,置于多功能读数仪读取552/630/690nm波长的吸光值(OD值)。用软件Graph Pad Prism 5.0分析数据,化合物对细胞增值的抑制活性以细胞存活率和化合物浓度为坐标绘图。IC50值以S形剂量反应曲线方程拟合,曲线方程为:Y=100/(1+10^(LogC-LogIC50)),其中C是化合物浓度。
表4汇总了化合物对NCI-H1299细胞增长的抑制作用数据(IC50)。
表4.6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮与其相似物对NCI-H1299细胞增长的抑制作用
实施例 1 2 3 4 5 6
IC50(μm) 1.59 0.7817 >10 0.2036 0.9410 0.4611
实施例 7 8 9 10 11 12
IC50(μm) 1.263 0.1593 0.1827 0.1718 0.7049 0.2318
实施例 13 14 15 16 17 18
IC50(μm) 0.1014 0.0987 0.9664 0.6170 0.4468 >10
实施例 19 20 21 22 23 27
IC50(μm) >10 >10 3.302 >10 >10 >10
实施例 28 32 33 34 35 36
IC50(μm) >3 >10 >10 0.4679 0.2867 >10
实施例 37 38 39 40 41 42
IC50(μm) >10 >10 >10 >10 >10 >10
实施例 43 44 45 46 47 48
IC50(μm) >10 0.5970 0.6158 0.5282 0.3300 1.122
实施例 49 50 51 52 53 54
IC50(μm) 0.5434 0.2292 0.1651 0.1273 0.1448 1.533
实施例 55 56 57 58 59 60
IC50(μm) >10 0.3177 >10 0.4265 >10 0.9918
实施例 61 62 63 64 65 66
IC50(μm) 1.229 0.9249 0.3596 0.1774 0.4045 >10
实施例 67 68 69 70 71 72
IC50(μm) >10 >10 0.4261 0.4477 0.6032 0.3282
实施例 73 74 75 76 77 78
IC50(μm) 1.433 0.7080 0.1614 0.1976 0.2168 0.1492
实施例 79 80 81 82 83 84
IC50(μm) >10 0.9778 0.3099 1.198 1.435 0.8364
实施例 85 86 87 88 89 90
IC50(μm) >10 >10 >10 >3 0.9571 >10
实施例 91 92 93 94 95 96
IC50(μm) 2.359 >10 >10 2.394 2.132 >10
实施例 97 98 99 100 101 102
IC50(μm) >10 >10 >10 0.5322 0.2142 >10
实施例 103 104 105 106 107 108
IC50(μm) 1.049 1.050 0.9111 0.8351 0.7944 0.8991
实施例 109 110 111 112 113 114
IC50(μm) 0.8844 0.5993 0.9132 0.4811 0.6715 0.2329
实施例 115 116 117 118 119 120
IC50(μm) 0.3813 0.2650 0.1026 0.0964 0.0923 0.0593
实施例 121 122 123 124 125 126
IC50(μm) >10 0.3345 0.2184 0.1384 0.0929 0.0670
实施例 127 128 129 130 131 133
IC50(μm) 0.0965 0.3060 0.0782 0.2429 0.0588 0.0460
实施例 136 137 138 139 140 141
IC50(μm) 0.3103 0.1221 0.1702 0.0859 0.0929 0.04285
实施例 142 143 144 145 146 147
IC50(μm) 0.1093 0.0583 0.1400 0.2193 0.2505 0.2093
实施例 148 149 150 151 152 153
IC50(μm) 0.1226 0.1127 0.3469 0.1746 1.947 0.2352
实施例 154 155 156 157 158 159
IC50(μm) 0.1740 0.1089 0.1500 0.2222 0.1054 0.05677
实施例 160 161 162 163 164 165
IC50(μm) 0.0628 0.2173 0.2068 0.0978 0.1590 0.0973
实施例 166 167 168 169 170 171
IC50(μm) 0.0993 0.1067 0.5994 0.2738 0.08986 0.4424
实施例 172 173 174 175 176 177
IC50(μm) 0.3624 0.2264 0.1518 0.2689 0.1260 0.08313
实施例 178 179 180 181 182 183
IC50(μm) 0.3597 70.57 2.047 0.1869 0.5029 1.042
实施例 184 186 AZD1775      
IC50(μm) 0.8786 0.1543 0.2873      
因此,经MTT检测法测定,6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮(实施例1)与其相似物对NCI H1299细胞增长有抑制作用。其中多个实施例,例如实施例75,对NCI H1299细胞增长的抑制作用比AZD1775强。
虽然已经充分地描述了本发明,但是本领域技术人员应当理解,可在不影响本发明范围或其任何实施方案的情况下,在广泛且等同的条件、制剂和其它参数范围内进行相同实施。本文所引用的所有专利、专利申请和出版物都全文引入本文以供参考。

Claims (10)

  1. 式I的化合物,或其可药用盐或前药:
    Figure PCTCN2017111230-appb-100001
    其中,A是N或CR15
    R1为氢,可被取代的C1-8烷基,可被取代的C2-8烯基,可被取代的C3-8环烷基,可被取代的芳基,或可被取代的杂芳基;
    R2为可被取代的杂环基,可被取代的芳基,或可被取代的杂芳基;
    R3-R7和R15独立为氢、卤素、可被取代的氨基、可被取代的烷氧基、可被取代的C1-10烷基、卤烷基、烯基、炔基、羟基烷基、氨基烷基、羧基烷基、硝基、氰基、酰氨基、羟基、巯基、酰氧基、叠氮基、羧基、亚乙基二氧基,羟基酰氨基或可被取代的烷硫基。
  2. 权利要求1的化合物,其中A为N,R1和R2为可被取代的芳基,R3-R7为氢。
  3. 权利要求1的化合物,其中为式II的化合物,或其可药用盐或前药:
    Figure PCTCN2017111230-appb-100002
    其中,R3-R7独立为氢、卤素、可被取代的氨基、可被取代的烷氧基、可被取代的C1-10烷基、卤烷基、烯基、炔基、羟基烷基、氨基烷基、羧基烷基、硝基、氰基、酰氨基、羟基、巯基、酰氧基、叠氮基、羧基、亚乙基二氧基,羟基酰氨基或可被取代的烷硫基。
    Ar1和Ar2为可被取代的芳基或可被取代的杂芳基;
  4. 权利要求3的化合物,其中R3-R7为氢,Ar1和Ar2为可被取代的苯基。
  5. 权利要求1的化合物,其中所述化合物选自:
    6-(2-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    4-(2-氯苯基)-8-((4-(4-甲基哌嗪-1-基)苯基)氨基)-1,2-二氢咪唑并[1,2-a]吡啶并[3,4-e]嘧啶-5(4H)-酮;
    6-(2,6-二氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-甲基苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(4-异丙基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(4-乙酰基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((2-甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((2,4,4-三甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((2'-甲基-2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((2'-乙酰基-2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((2-甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((2,4,4-三甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((2'-甲基-2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二甲基苯基)-2-((2-甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二甲基苯基)-2-((2,4,4-三甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二甲基苯基)-2-((2'-甲基-2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-异丙基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-叔丁基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-环丙基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-环己基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-烯丙基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(噻吩-2-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(呋喃-2-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(1H-吡咯-2-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(1H-咪唑-5-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,8-二甲基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-9,9-二甲基-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-((2S,6R)-2,6-二甲基吗啉基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(吗啉基甲基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-((3S,5R)-4-异丙基-3,5-二甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((2-氟-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((2-氟-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((2-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((2-氯-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)-2-(三氟甲基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((2-三氟甲基-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((2-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((2-甲基-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-氟-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-氟-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-氯-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)-3-(三氟甲基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-三氟甲基-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-甲基-4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(4-异丙基哌嗪-1-基)-3-甲基苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-((3S,5R)-4-异丙基-3,5-二甲基哌嗪-1-基)-3-甲基苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(4-甲基哌嗪-1-基)-3-硝基苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((6-(4-甲基哌嗪-1-基)吡啶-3-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((2'-异丙基-2',3'-二氢-1'H-螺[环丙烷-1,4'-异喹啉]-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((5-甲基-4,5,6,7-四氢吡唑并[1,5-a]吡嗪-2-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氟苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氟-6-三氟甲基苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氟-6-甲基苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((4-(4-异丙基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((4-((3S,5R)-4-异丙基-3,5-二甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((2-氟-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((2-氯-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((2-三氟甲基-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氟-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氟-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氯-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((4-(4甲基哌嗪-1-基)-3-(三氟甲基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-三氟甲基-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-甲基-4-(4-异丙基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-甲基-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-甲基-4-((3S,5R)-4-异丙基-3,5-二甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((5-甲基-4,5,6,7-四氢吡唑并[1,5-a]吡嗪-2-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-三氟甲基苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-甲基苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-甲氧基苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二甲基苯基)-2-((4-((3R,5S)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(4-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(3-氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,4-二氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-4-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-3-氟苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,5-二氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,3-二氯苯基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(嘧啶-2-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-环丁基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-环戊基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-苯基-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(吡啶-2-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(吡啶-3-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(吡啶-4-基)-2-((4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氟苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氟苯基)-2-((3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(1H-咪唑-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(4-甲基-1,4-二氮杂环庚烷-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-((4-甲基哌嗪-1-基)甲基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(2-(二甲氨基)乙氧基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(3-(二甲氨基)丙氧基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-((1-甲基哌啶-4-基)氧基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-((2-(二甲氨基)乙基)氨基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-((2-(二甲基氨基)乙基)(甲基)氨基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-((3-(二甲氨基)丙基)氨基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-((3-(二甲基氨基)丙基)(甲基)氨基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-((1-甲基哌啶-4-基)氨基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(甲基(1-甲基哌啶-4-基)氨基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(1-甲基哌啶-4-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((4-(4-二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-氟-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-氯-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-溴-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-甲基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮的;
    6-(2-氯-6-氟苯基)-2-((3-氟-5-甲基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((5-(4-甲基哌嗪-1-基)吡嗪-2-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3,5-二氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-氟-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-氟-5-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-氯-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-氯-5-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-溴-5-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-甲基-5-三氟甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-甲氧基-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((5-氯-2-甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((2,5-二甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((5-氯-2,4,4-甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((2,4,4,5-四甲基-1,2,3,4-四氢异喹啉-7-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((5'-氯-2'-甲基-2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((2',5'-二甲基-2',3'-二氢-1'H-螺(环丙烷-1,4'-异喹啉)-7'-基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3,5-二氯-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氯-5-甲基-4-((3S,5R)-3,4,5-三甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氯-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-溴-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-甲基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3,5-二氯-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氟-5-甲基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氯-5-甲氧基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-溴-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氯-5-氟-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氟-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氟-5-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3,5-二氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氯-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氯-5-三氟甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氯-5-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-溴-5-氟-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-溴-5-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-溴-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-溴-5-甲氧基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-甲氧基-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-溴-6-氯苯基)-2-((3-氟-5-甲基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-溴-6-氯苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-溴-6-氯苯基)-2-((3-溴-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-溴-6-氯苯基)-2-((3-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-溴-6-氯苯基)-2-((3,5-二氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-溴-6-氯苯基)-2-((3-氟-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-溴-6-氯苯基)-2-((3-氯-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-溴-6-氯苯基)-2-((3-溴-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氟-6-甲基苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氟-6-甲基苯基)-2-((3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-甲基苯基)-2-((3-氟-5-甲基-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-甲基苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-甲基苯基)-2-((3,5-二氯-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-甲基苯基)-2-((3-氯-5-甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-甲基苯基)-2-((3,5-二甲基-4-(4-甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((4-(哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氯-4-(哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-甲基-4-(哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-甲氧基-4-(哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-氯-6-氟苯基)-2-((3-(羟基甲基)-4-(哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-(羟基甲基)-4-(哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-(羟基甲基)-4-(甲基哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((4-(4-(2-羟基乙基)哌嗪-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((4-吗啉基苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-((甲基氨基)甲基)-4-吗啉基苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2-溴-6-氯苯基)-2-((3-氯-4-(4-(二甲氨基)哌啶-1-基)苯基)氨基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮;
    6-(2,6-二氯苯基)-2-((3-氯-4-(4-甲基哌嗪-1-基)苯基)胺基)-8,9-二氢咪唑并[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮二盐酸盐或其可药用盐或前药。
  6. 权利要求1~5中任一项所述的化合物在制备治疗或预防Wee1介导的疾病的药物中的用途。
  7. 权利要求6的用途,其中所述疾病是癌症;优选地,所述癌症选自肝癌、黑素瘤、霍奇金病、非霍奇金淋巴瘤、急性淋巴白血病、慢性淋巴白血病、多发性骨髓瘤、成神经细胞瘤、乳腺癌、卵巢癌、肺癌、维尔姆斯瘤、子宫颈癌、睾丸癌、软组织肉瘤、慢性淋巴细胞白血病、原发性巨球蛋白血症、膀胱癌、慢性粒细胞白血病、原发性脑癌、恶性黑素瘤、小细胞肺癌、胃癌、结肠癌、恶性胰腺胰岛瘤、恶性类癌性癌症、恶性黑素瘤、绒毛膜癌、蕈樣肉芽腫、头颈癌、骨原性肉瘤、胰腺癌、急性粒细胞白血病、毛细胞白血病、横纹肌肉瘤、卡波西肉瘤、泌尿生殖系统肿瘤、甲状腺癌、食管癌、恶性高钙血症、子宫颈增生症、肾细胞癌、子宫内膜癌、真性红细胞增多症、特发性血小板增多症、肾上腺皮质癌、皮肤癌和前列腺癌。
  8. 一种药用组合物,包括权利要求1~5中任一项所述的化合物与可药用载体。
  9. 权利要求8的药用组合物,其中所述组合物还含有至少一种已知的抗癌药物,或所述抗癌药物的可药用盐。
  10. 权利要求8或9的药用组合物,其中,所述组合物还含有至少一种选自下组的抗癌药物:白消安、马法兰、苯丁酸氮芥、环磷酰胺、异环磷酰胺、替莫唑胺、苯达莫司汀、顺铂、丝裂霉素C、博莱霉素、卡铂、喜树碱、伊立替康、托泊替康、阿霉素、表阿霉素、阿克拉霉素、米托蒽醌、甲基羟基玫瑰树碱、铭托泊普、5-氮杂胞苷、吉西他滨、5-氟尿嘧啶、甲氨蝶呤、5-氟-2'-去氧尿苷、氟达拉滨、奈拉滨、阿糖胞苷、普拉曲沙、培美曲塞、羟基脲、硫代鸟嘌呤、秋水仙碱、长春碱、长春新碱、长春瑞滨、紫杉醇、伊沙匹隆、卡巴他赛、多西他赛、单抗、帕尼单抗、耐措妥珠单抗、纳武单抗、派姆单抗、雷莫芦单抗、贝伐珠单抗、帕妥珠单抗、曲妥珠单抗、西妥昔单抗、奥滨尤妥珠单抗、奥法木单抗、利妥昔单抗、阿仑单抗、替伊莫单抗、托西莫单抗、本妥昔单抗、达雷木单抗、埃罗妥珠单抗、T-DM1、Ofatumumab、Dinutuximab、Blinatumomab、易普利姆玛、阿瓦斯丁、赫赛汀、美罗华、伊马替尼、吉非替尼、厄洛替尼、奥斯替尼、阿法替尼、赛立替尼、艾乐替尼、克唑替尼、埃罗替尼、拉帕替尼、索拉非尼、舒尼替尼、尼罗替尼、达沙替尼、帕唑帕尼、特癌适、依维莫司、伏立诺他、罗咪地辛、帕比司他、贝利司他、他莫昔芬、来曲唑、氟维司群、米托胍腙、奥曲肽、视黄酸、砒霜、唑来膦酸、硼替佐米、卡非佐米、Ixazomib、维莫德吉、索尼德吉、狄诺塞麦、萨力多胺、来那度胺、Venetoclax、Aldesleukin(重组人白介素-2)、 Sipueucel-T(前列腺癌治疗疫苗)、帕博西尼、奥拉帕尼、Niraparib,Rucaparib和Talazoparib。
PCT/CN2017/111230 2016-11-16 2017-11-16 8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物 WO2018090939A1 (zh)

Priority Applications (9)

Application Number Priority Date Filing Date Title
US16/461,512 US10703759B2 (en) 2016-11-16 2017-11-16 8,9-dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-ones
ES17872807T ES2968252T3 (es) 2016-11-16 2017-11-16 Compuesto 8,9-dihidroimidazol[1,2-a]pirimido[5,4-e]pirimidin-5(6H)-cetona
CA3043945A CA3043945A1 (en) 2016-11-16 2017-11-16 8,9-dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6h)-ones
AU2017359844A AU2017359844B2 (en) 2016-11-16 2017-11-16 8,9-dihydroimidazole[1,2-a]pyrimido[5,4-e]pyrimidine-5(6H)-ketone compound
KR1020197017242A KR102534962B1 (ko) 2016-11-16 2017-11-16 8,9-디하이드로이미다졸[1,2-a]피리미도[5,4-e]피리미딘-5(6H)-케톤계 화합물
CN201780068490.0A CN109906227B (zh) 2016-11-16 2017-11-16 8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物
EP17872807.7A EP3543242B1 (en) 2016-11-16 2017-11-16 8,9-dihydroimidazole[1,2-a]pyrimido[5,4-e]pyrimidine-5(6h)-ketone compound
JP2019527384A JP7240784B2 (ja) 2016-11-16 2017-11-16 8,9-ジヒドロイミダゾール[1,2-a]ピリミド[5,4-e]ピリミジン-5(6H)-ケトン類化合物
US16/883,397 US11345711B2 (en) 2016-11-16 2020-05-26 8,9-dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-ones

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201611009827 2016-11-16
CN201611009827.4 2016-11-16
CN201710440456.3A CN109020981A (zh) 2017-06-12 2017-06-12 8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物
CN201710440456.3 2017-06-12

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/461,512 A-371-Of-International US10703759B2 (en) 2016-11-16 2017-11-16 8,9-dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-ones
US16/883,397 Continuation US11345711B2 (en) 2016-11-16 2020-05-26 8,9-dihydroimidazo[1,2-a]pyrimido[5,4-e]pyrimidin-5(6H)-ones

Publications (1)

Publication Number Publication Date
WO2018090939A1 true WO2018090939A1 (zh) 2018-05-24

Family

ID=62145906

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/111230 WO2018090939A1 (zh) 2016-11-16 2017-11-16 8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物

Country Status (9)

Country Link
US (2) US10703759B2 (zh)
EP (1) EP3543242B1 (zh)
JP (1) JP7240784B2 (zh)
KR (1) KR102534962B1 (zh)
CN (1) CN109906227B (zh)
AU (1) AU2017359844B2 (zh)
CA (1) CA3043945A1 (zh)
ES (1) ES2968252T3 (zh)
WO (1) WO2018090939A1 (zh)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020192581A1 (zh) 2019-03-22 2020-10-01 首药控股(北京)有限公司 Wee1抑制剂及其制备和用途
WO2020210383A1 (en) * 2019-04-09 2020-10-15 Nuvation Bio Inc. Heterocyclic compounds and uses thereof
WO2020210377A1 (en) * 2019-04-09 2020-10-15 Nuvation Bio Inc. Heterocyclic compounds and uses thereof
US10807994B2 (en) 2017-10-09 2020-10-20 Nuvation Bio Inc. Heterocyclic compounds and uses thereof
WO2021073491A1 (zh) * 2019-10-16 2021-04-22 上海瑛派药业有限公司 二氢咪唑并嘧啶并嘧啶酮类化合物
US11299493B2 (en) 2017-10-09 2022-04-12 Nuvation Bio Inc. Heterocyclic compounds and uses thereof
US11332473B2 (en) 2019-04-09 2022-05-17 Nuvation Bio Inc. Substituted pyrazolo[3,4-d]pyrimidines as Wee1 inhibitors
WO2022188802A1 (en) * 2021-03-10 2022-09-15 Impact Therapeutics (Shanghai) , Inc Use of wee1 kinase inhibitors in the treatment of cancer
WO2022257581A1 (zh) * 2021-06-08 2022-12-15 四川大学 一类咪唑烷并嘧啶酮化合物及其在治疗HsClpP介导的疾病中的用途
WO2023083194A1 (zh) * 2021-11-09 2023-05-19 杭州格博生物医药有限公司 Wee1蛋白激酶降解剂及其用途
WO2023093840A1 (en) * 2021-11-26 2023-06-01 Impact Therapeutics (Shanghai) , Inc Use of wee1 kinase inhibitors in the treatment of cancer
WO2023138362A1 (zh) 2022-01-18 2023-07-27 江苏天士力帝益药业有限公司 Wee1抑制剂及其制备和用途
WO2023170065A1 (en) 2022-03-07 2023-09-14 Debiopharm International S.A. Methods of treating small cell lung cancer
JP7446287B2 (ja) 2018-09-27 2024-03-08 貝達薬業股▲ふん▼有限公司 Fgfr4阻害剤及びその使用

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115073460B (zh) * 2022-07-13 2023-07-25 苏州施安鼎泰生物医药技术有限公司 一种嘧啶并[5,4-c][2,6]萘啶衍生物及其制备方法以及药物组合物和应用

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3989816A (en) 1975-06-19 1976-11-02 Nelson Research & Development Company Vehicle composition containing 1-substituted azacycloheptan-2-ones
US4444762A (en) 1980-04-04 1984-04-24 Nelson Research & Development Company Vehicle composition containing 1-substituted azacyclopentan-2-ones
WO2005021551A1 (en) 2003-08-27 2005-03-10 Amgen Inc. Substituted heterocyclic compounds and methods of use
WO2012161812A1 (en) 2011-02-28 2012-11-29 Abbott Laboratories Tricyclic inhibitors of kinases
WO2015092431A1 (en) * 2013-12-19 2015-06-25 Almac Discovery Limited Pyrimidopyrimidinones useful as wee-1 kinase inhibitors

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2479644A1 (en) * 2002-04-03 2003-10-09 F. Hoffmann-La Roche Ag Imidazo fused compounds
CN103570731B (zh) * 2012-07-30 2016-05-18 中国科学院广州生物医药与健康研究院 嘧啶并三环或嘧啶并四环类化合物及其药用组合物和应用

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3989816A (en) 1975-06-19 1976-11-02 Nelson Research & Development Company Vehicle composition containing 1-substituted azacycloheptan-2-ones
US4444762A (en) 1980-04-04 1984-04-24 Nelson Research & Development Company Vehicle composition containing 1-substituted azacyclopentan-2-ones
WO2005021551A1 (en) 2003-08-27 2005-03-10 Amgen Inc. Substituted heterocyclic compounds and methods of use
WO2012161812A1 (en) 2011-02-28 2012-11-29 Abbott Laboratories Tricyclic inhibitors of kinases
WO2015092431A1 (en) * 2013-12-19 2015-06-25 Almac Discovery Limited Pyrimidopyrimidinones useful as wee-1 kinase inhibitors

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
BMC CANCER, vol. 15, 2015, pages 462
GREENWALD ET AL., J. MED. CHEM., vol. 42, 1999, pages 3657 - 3667
J CLIN ONCOL, 6 September 2016 (2016-09-06), pages JC0675991
J CLIN ONCOL, vol. 33, 2015, pages 3409 - 3415
KARNAK ET AL., CLIN CANCER RES, vol. 20, no. 9, 2014, pages 5085 - 5096
LEVENBERG, K., Q., APPL. MATH., vol. 2, 1944, pages 164 - 168
LINDQVIST, A. ET AL., THE JOURNAL OF CELL BIOLOGY, vol. 185, 2009, pages 193 - 202
MCGOWAN, C.H. ET AL., THE EMBO JOURNAL, vol. 12, 1993, pages 75 - 85
NURSE, P., NATURE, vol. 344, 1990, pages 503 - 508
O'CONNELL ET AL., THE EMBO JOURNAL, vol. 16, 1997, pages 545 - 554
PARKER, L.L. ET AL., SCIENCE, vol. 257, 1992, pages 1955 - 1957
SANCAR, A. ET AL., ANNUAL REVIEW OF BIOCHEMISTRY, vol. 73, 2004, pages 39 - 85
See also references of EP3543242A4
STUMPFF, J. ET AL., CURR BIOL, vol. 14, 2004, pages 2143 - 2148
WANG, Y. ET AL., CANCER BIOLOGY & THERAPY, vol. 3, 2004, pages 305 - 313

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10807994B2 (en) 2017-10-09 2020-10-20 Nuvation Bio Inc. Heterocyclic compounds and uses thereof
US11299493B2 (en) 2017-10-09 2022-04-12 Nuvation Bio Inc. Heterocyclic compounds and uses thereof
JP7446287B2 (ja) 2018-09-27 2024-03-08 貝達薬業股▲ふん▼有限公司 Fgfr4阻害剤及びその使用
WO2020192581A1 (zh) 2019-03-22 2020-10-01 首药控股(北京)有限公司 Wee1抑制剂及其制备和用途
WO2020210383A1 (en) * 2019-04-09 2020-10-15 Nuvation Bio Inc. Heterocyclic compounds and uses thereof
WO2020210377A1 (en) * 2019-04-09 2020-10-15 Nuvation Bio Inc. Heterocyclic compounds and uses thereof
US11332473B2 (en) 2019-04-09 2022-05-17 Nuvation Bio Inc. Substituted pyrazolo[3,4-d]pyrimidines as Wee1 inhibitors
CN114502559B (zh) * 2019-10-16 2024-02-02 上海瑛派药业有限公司 二氢咪唑并嘧啶并嘧啶酮类化合物
WO2021073491A1 (zh) * 2019-10-16 2021-04-22 上海瑛派药业有限公司 二氢咪唑并嘧啶并嘧啶酮类化合物
CN114502559A (zh) * 2019-10-16 2022-05-13 上海瑛派药业有限公司 二氢咪唑并嘧啶并嘧啶酮类化合物
WO2022188802A1 (en) * 2021-03-10 2022-09-15 Impact Therapeutics (Shanghai) , Inc Use of wee1 kinase inhibitors in the treatment of cancer
WO2022257581A1 (zh) * 2021-06-08 2022-12-15 四川大学 一类咪唑烷并嘧啶酮化合物及其在治疗HsClpP介导的疾病中的用途
WO2023083194A1 (zh) * 2021-11-09 2023-05-19 杭州格博生物医药有限公司 Wee1蛋白激酶降解剂及其用途
WO2023093840A1 (en) * 2021-11-26 2023-06-01 Impact Therapeutics (Shanghai) , Inc Use of wee1 kinase inhibitors in the treatment of cancer
WO2023138362A1 (zh) 2022-01-18 2023-07-27 江苏天士力帝益药业有限公司 Wee1抑制剂及其制备和用途
WO2023170065A1 (en) 2022-03-07 2023-09-14 Debiopharm International S.A. Methods of treating small cell lung cancer

Also Published As

Publication number Publication date
CA3043945A1 (en) 2018-05-24
EP3543242A4 (en) 2020-04-15
KR102534962B1 (ko) 2023-05-22
US20200385394A1 (en) 2020-12-10
US10703759B2 (en) 2020-07-07
CN109906227A (zh) 2019-06-18
JP2020502065A (ja) 2020-01-23
US20190308984A1 (en) 2019-10-10
EP3543242C0 (en) 2024-01-03
ES2968252T3 (es) 2024-05-08
AU2017359844B2 (en) 2021-09-30
EP3543242A1 (en) 2019-09-25
EP3543242B1 (en) 2024-01-03
CN109906227B (zh) 2022-03-11
AU2017359844A1 (en) 2019-06-27
KR20190099209A (ko) 2019-08-26
US11345711B2 (en) 2022-05-31
JP7240784B2 (ja) 2023-03-16

Similar Documents

Publication Publication Date Title
CN109906227B (zh) 8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物
RU2539568C2 (ru) Пиразолопиримидиновые соединения-ингибиторы jak и способы
CN110372720B (zh) 一类激酶抑制剂
US11345710B2 (en) Imidazo[1,2-b]pyrimido[4,5-d]pyridazin-5(6H)-ones and the use thereof
CN112142744A (zh) 取代的稠合杂芳双环化合物作为激酶抑制剂及其应用
JP2022526854A (ja) ホスファチジルイノシトール3-キナーゼ阻害剤
CA3230491A1 (en) Substituted tricyclic compounds as parp inhibitors and use thereof
WO2018127195A1 (zh) 取代的稠合杂芳基化合物作为激酶抑制剂及其应用
WO2023169226A1 (en) Substituted tricyclic compounds as parp inhibitors and the use thereof
CN111989332B (zh) 作为cdk抑制剂的大环化合物、其制备方法及其在医药上的应用
US20240208969A1 (en) Substituted fused bicyclic compounds as parp inhibitors and the use thereof
CN115260187A (zh) 吡啶酮化合物及其用途
CA3235663A1 (en) Substituted triazoloheteroaryl compounds as usp1 inhibitors and the use thereof
CN109020981A (zh) 8,9-二氢咪唑[1,2-a]嘧啶并[5,4-e]嘧啶-5(6H)-酮类化合物
WO2022116943A1 (zh) 取代的稠合双环化合物作为激酶抑制剂及其应用
TWI602818B (zh) 稠合雜環化合物作爲蛋白激酶抑制劑
JP2022548055A (ja) 置換イミダゾキノキサリン化合物およびその応用

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: 17872807

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3043945

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2019527384

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: 20197017242

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017359844

Country of ref document: AU

Date of ref document: 20171116

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017872807

Country of ref document: EP

Effective date: 20190617