WO2019037678A1 - 吡唑并[3,4-d]嘧啶-3-酮衍生物、其药物组合物及应用 - Google Patents

吡唑并[3,4-d]嘧啶-3-酮衍生物、其药物组合物及应用 Download PDF

Info

Publication number
WO2019037678A1
WO2019037678A1 PCT/CN2018/101290 CN2018101290W WO2019037678A1 WO 2019037678 A1 WO2019037678 A1 WO 2019037678A1 CN 2018101290 W CN2018101290 W CN 2018101290W WO 2019037678 A1 WO2019037678 A1 WO 2019037678A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
unsubstituted
compound
group
alkyl
Prior art date
Application number
PCT/CN2018/101290
Other languages
English (en)
French (fr)
Inventor
高大新
赵志明
陈寿军
武志恒
Original Assignee
上海迪诺医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 上海迪诺医药科技有限公司 filed Critical 上海迪诺医药科技有限公司
Publication of WO2019037678A1 publication Critical patent/WO2019037678A1/zh

Links

Images

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
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53831,4-Oxazines, e.g. morpholine ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-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 present invention relates to a pyrazolo[3,4-d]pyrimidin-3-one derivative capable of inhibiting Wee1 and its signaling pathway, and a pharmaceutically acceptable salt thereof, comprising the derivative and pharmaceutically acceptable thereof A pharmaceutical composition of a salt, and the use of the derivative and pharmaceutically acceptable salt thereof for the treatment of a disease mediated by Wee1 and for the preparation of a medicament for treating Wee1-mediated diseases.
  • Wee1 (Wee1G2 checkpoint kinase; gene number: 7465) is a member of the serine/threonine protein kinase family, which directly phosphorylates cyclin-dependent kinase 1 (CDK1) and phosphorylates the tyrosine of CDK1
  • CDK1 cyclin-dependent kinase 1
  • the acid 15 residue which is an inhibitory phosphorylation site, negatively regulates CDK1 activity.
  • Activation of the G2 checkpoint is primarily through the inhibition of mitosis, promoting the cyclin B-CDK1 complex. Normal cells repair damaged DNA during the G1 arrest period. However, cancer cell G1-S detection sites are often deleted, and it is necessary to rely on the function of G2-M detection sites for DNA repair.
  • P53-deficient tumor cells lack the function of the G1 checkpoint and thus rely on the G2 checkpoint as a response to DNA damage in cell cycle arrest. After DNA damage, the G2 checkpoint prevents damaged cells from entering mitosis, thereby protecting them from mitotic catastrophe and apoptosis.
  • Wee1 is an indispensable factor in the function of the G2 checkpoint. Abolishing the G2 detection site by Wee1 inhibitor may selectively sensitize P53-deficient cancer cells to DNA damage and avoid affecting surrounding normal tissues. Wee1 also regulates S-phase CDK activity, preventing the induction of DNA damage during normal S-phase progression. In addition, Wee1 plays an active mediating role in homologous recombination (HR) repair, and homologous recombination repair is an important pathway for DNA double-strand break repair.
  • HR homologous recombination
  • Upregulation of Wee1 can be seen in many different types of cancer, including hepatocellular carcinoma (Masaki, et al, 2003), breast cancer, cervical cancer, lung cancer (Iom, et al, 2009), squamous cell carcinoma (Magnussen, et Al, 2013), glioma DIPG (Mueller, et al, 2014), malignant glioma (Mir, et al, 2010; Music, et al, 2016), medulloblastoma (Harris, et al, 2014), leukemia (Tibas, et al, 2012; Porter, et al, 2012), melanoma (Magnussen, et al, 2012), and ovarian cancer (Slipicevic, et al, 2014).
  • Wee1 is associated with poor prognosis of many types of cancer. Inhibition of Wee1 caused apoptosis in some P53 inactivated tumor cells. Inhibition of Wee1 can be sensitive to cancer cells that are resistant to chemotherapy and radiation therapy.
  • the latest study (Pfister, et al, 2015) demonstrates the interaction between synthetic lethality and H3K36me3 deletion, epigenetic changes in some cancer cells, and Wee1 inhibition, thereby clearly defining Wee1 inhibition and more precise targeted gene changes. The relationship between cancer patients provides strong evidence.
  • Wee1 is currently a highly attractive therapeutic target in the field of cancer therapy.
  • Wee1 there are still many opportunities to expand and benefit from its application.
  • the compounds described herein, compositions and methods of use thereof will contribute to the development of Wee1 inhibitors to meet clinically unmet drug needs.
  • the technical problem to be solved by the present invention is to provide a novel pyrazolo[3,4-d]pyrimidin-3-one derivative, a preparation method thereof, a pharmaceutical composition and use thereof.
  • the pyrazolo[3,4-d]pyrimidin-3-one derivatives of the present invention have a good inhibitory effect on Wee1 and related signaling pathways, and can effectively treat and/or alleviate cancer.
  • the present invention provides a compound of the formula (I), an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt;
  • W is N or CR 7 ;
  • X is CR 8 or N;
  • U and V are respectively selected from N or CH, and U and V are not N at the same time; or, U and V are respectively selected from N or CR 8 , and U and V are not N at the same time;
  • R 1 is hydrogen, halogen, C 2-6 alkynyl, C 2-6 alkenyl, aryl, cycloalkyl, heterocycloalkyl, heteroaryl or substituted alkyl; when the alkyl group is substituted, Optionally substituted at one or more of the following positions: halogen, haloalkoxy, aminocycloalkyl, -SR a , -OR a , -OC(O)R a , -OC(O)NR a R b , -C(O)OR a , -C(O)R a , -C(O)NR a R b , -NR a R b , -NR a C(O)R b , -NR a S (O) 2 R b , -S(O) 1-2 R b , -S(O) 2 NR a R b and -NR a S(O) 2 NR a R b
  • R 2 is alkyl, haloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, arylalkyl, heteroarylalkyl, alkenyl or alkynyl;
  • R 4 , R 5 , R 6 and R 7 are each independently hydrogen, halogen, hydroxy, cyano, nitro, decyl, amino, alkyl, alkoxy, alkylthio, haloalkyl, haloalkoxy, C 2-6 alkynyl, C 2-6 alkenyl, aryl, cycloalkyl, heterocycloalkyl, heteroaryl, -OC(O)R c , -OC(O)OR c , -OC(O) N(R c ) 2 , -C(O)OR c , -C(O)R c , -C(O)N(R c ) 2 , -N(R c ) 2 , -NHC(O)R c , -NHC(O)OR c , -NHC(O)N(R c ) 2 , -NHS(O) 2 R c , -S(O)
  • R 3 and R 4 are independent substituents, or R 3 and R 4 together with the ring atom to which they are attached form an A ring which is a substituted or unsubstituted cycloalkyl group, a substituted or unsubstituted heterocycloalkane.
  • R 8 , R 9 or R 11 are each independently hydrogen, -C(O)OR c , -C(O)R c , -C(O)NR c R d , -S(O) 1-2 R c , -S(O) 2 NR c R d , substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or not Substituted heteroaryl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted heterocycloalkylalkyl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroarylalkyl, a substituted or unsubstituted alkenyl group, a substituted or unsubstituted alkynyl group
  • R 1 and R 9 are each independently substituted, or R 1 and R 9 are bonded to each other to form a 4-8 membered heterocycloalkyl group, which may further be 1 to 3 C 1-6 alkyl groups or C 3-6 cycloalkyl optionally substituted;
  • Each R a and each R b are independently selected from the group consisting of hydrogen, C 1-6 alkyl, C 1-6 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 ring Alkyl, 3-8 membered heterocycloalkyl, C 6-10 aryl, 5-6 membered heteroaryl, C 3-8 cycloalkyl C 1-6 alkyl, 3-8 membered heterocycloalkyl C a 1-6 alkyl group, a phenyl C 1-6 alkyl group, or a 5-6 membered heteroaryl C 1-6 alkyl group; the C 1-6 alkyl group, a C 1-6 alkoxy group, a C 2 ⁇ 6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 3-8 membered heterocycloalkyl, C 6-10 aryl, or 5-6 membered heteroaryl is unsubstituted or optionally 1 to 3 are selected from the group consist
  • R a and R b are independently substituted, or the R a and R b together with the N atom to which they are attached form a 3-12 membered heterocycloalkyl group, and the heterocycloalkyl group may further contain 1 to 3 selected a hetero atom or a group derived from N, O, S(O) 0-2 or C(O); the heterocycloalkyl group being unsubstituted or optionally 1 to 3 selected from the group consisting of halogen, hydroxy, amino, carboxy One or more substituents of a C 1-6 alkyl group, a halogenated C 1-6 alkyl group, a C 1-6 alkoxy group, a C 1-6 alkylamino group, and a halogenated C 1-6 alkoxy group Replace at any position;
  • Each R c and each R d are independently selected from the group consisting of hydrogen, C 1-6 alkyl, C 1-6 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 ring Alkyl, 3-8 membered heterocycloalkyl, C 6-10 aryl, 5-6 membered heteroaryl, C 3-8 cycloalkyl C 1-6 alkyl, 3-8 membered heterocycloalkyl C a 1-6 alkyl group, a phenyl C 1-6 alkyl group, or a 5-6 membered heteroaryl C 1-6 alkyl group; the C 1-6 alkyl group, a C 1-6 alkoxy group, a C 2 ⁇ 6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 3-8 membered heterocycloalkyl, C 6-10 aryl, or 5-6 membered heteroaryl is unsubstituted or optionally 1 to 3 are selected from the group consist
  • the present invention provides a compound of the formula (I'), an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt;
  • W is N or CR 7 ;
  • X is CR 8 or N;
  • U and V are respectively selected from N or CR 8 , and U and V are not N at the same time;
  • R 2 is alkyl, haloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, arylalkyl, heteroarylalkyl, alkenyl or alkynyl;
  • R 4 , R 5 , R 6 and R 7 are each independently hydrogen, halogen, hydroxy, cyano, nitro, decyl, amino, alkyl, alkoxy, alkylthio, haloalkyl, haloalkoxy, C 2-6 alkynyl, C 2-6 alkenyl, aryl, cycloalkyl, heterocycloalkyl, heteroaryl, -OC(O)R c , -OC(O)OR c , -OC(O) N(R c ) 2 , -C(O)OR c , -C(O)R c , -C(O)N(R c ) 2 , -N(R c ) 2 , -NHC(O)R c , -NHC(O)OR c , -NHC(O)N(R c ) 2 , -NHS(O) 2 R c , -S(O)
  • R 3 and R 4 are independent substituents, or R 3 and R 4 together with the ring atom to which they are attached form an A ring which is a substituted or unsubstituted cycloalkyl group, a substituted or unsubstituted heterocycloalkane.
  • R 8 , R 9 or R 11 are each independently hydrogen, -C(O)OR c , -C(O)R c , -C(O)NR c R d , -S(O) 1-2 R c , -S(O) 2 NR c R d , substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or not Substituted heteroaryl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted heterocycloalkylalkyl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroarylalkyl, a substituted or unsubstituted alkenyl group, a substituted or unsubstituted alkynyl group
  • Each R a and each R b are independently selected from the group consisting of hydrogen, C 1-6 alkyl, C 1-6 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 ring Alkyl, 3-8 membered heterocycloalkyl, C 6-10 aryl, 5-6 membered heteroaryl, C 3-8 cycloalkyl C 1-6 alkyl, 3-8 membered heterocycloalkyl C a 1-6 alkyl group, a phenyl C 1-6 alkyl group, or a 5-6 membered heteroaryl C 1-6 alkyl group; the C 1-6 alkyl group, a C 1-6 alkoxy group, a C 2 ⁇ 6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 3-8 membered heterocycloalkyl, C 6-10 aryl, or 5-6 membered heteroaryl is unsubstituted or optionally 1 to 3 are selected from the group consist
  • R a and R b are independently substituted, or the R a and R b together with the N atom to which they are attached form a 3-12 membered heterocycloalkyl group, and the heterocycloalkyl group may further contain 1 to 3 selected a hetero atom or a group derived from N, O, S(O) 0-2 or C(O); the heterocycloalkyl group being unsubstituted or optionally 1 to 3 selected from the group consisting of halogen, hydroxy, amino, carboxy One or more substituents of a C 1-6 alkyl group, a halogenated C 1-6 alkyl group, a C 1-6 alkoxy group, a C 1-6 alkylamino group, and a halogenated C 1-6 alkoxy group Replace at any position;
  • Each R c and each R d are independently selected from the group consisting of hydrogen, C 1-6 alkyl, C 1-6 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 ring Alkyl, 3-8 membered heterocycloalkyl, C 6-10 aryl, 5-6 membered heteroaryl, C 3-8 cycloalkyl C 1-6 alkyl, 3-8 membered heterocycloalkyl C a 1-6 alkyl group, a phenyl C 1-6 alkyl group, or a 5-6 membered heteroaryl C 1-6 alkyl group; the C 1-6 alkyl group, a C 1-6 alkoxy group, a C 2 ⁇ 6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 3-8 membered heterocycloalkyl, C 6-10 aryl, or 5-6 membered heteroaryl is unsubstituted or optionally 1 to 3 are selected from the group consist
  • the W is preferably N.
  • the U is preferably CH.
  • the V is preferably CR 8 ; wherein R 8 is as defined above.
  • the X and Y are preferably any combination of the following:
  • X is N and Y is NR 9 ; wherein R 9 is as defined above.
  • X is CH and Y is NR 9 ; wherein R 9 is as defined above.
  • X is CR 8 and Y is O; wherein R 8 is as defined above.
  • X is CR 8 and Y is O; wherein R 8 is as defined above.
  • R 8 and R 11 are as defined above.
  • X and Y are X to be CH and Y to be NR 9 ; wherein R 9 is as defined above.
  • the (1) position is connected to the mother core
  • a group Preferably, the (2) position is linked to the parent core.
  • the (1) position is connected to the mother core
  • a group Preferably, the (2) position is linked to the parent core.
  • R 1 is preferably H or a substituted C 1-6 alkyl group; and R 1 is more preferably H.
  • the R 2 is preferably:
  • the R 2 is more preferably:
  • R 3 when the alkyl group, cycloalkyl group, heterocycloalkyl group, cycloalkylalkyl group or heterocycloalkyl group is substituted, it is preferably substituted by 1 to 3 substituents at any position. .
  • the substituents are as defined above.
  • the R 3 is preferably a substituted or unsubstituted C 1-6 alkyl group, and when the alkyl group is substituted, it is preferably substituted with one hydroxy group at an arbitrary position.
  • the R 4 is preferably H.
  • R 3 and R 4 together with the ring atom to which they are attached form an A ring, which is preferably a substituted or unsubstituted monocyclic cycloalkyl group, a substituted or unsubstituted monocyclic heterocycloalkyl group. a substituted or unsubstituted phenyl group, or a substituted or unsubstituted 5-6 membered heteroaryl group;
  • the A ring is preferably unsubstituted.
  • the A ring when substituted, it is preferably substituted at any position by 1 to 3 substituents.
  • the substituents are as defined above.
  • the R 5 is preferably H.
  • the R 6 is preferably H.
  • the R 8 is preferably H
  • the R 8 is preferably -NR c R d , -NR c S(O) 1-2 R d , -NR c C(O)R d , -NR c S(O) 2 NR c R d , -C(O)OR c , -C(O)R c , -C(O)NR c R d , -S(O) 1-2 R c , -S(O) 2 NR c R d , Substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted ring Alkylalkyl, substituted or unsubstituted heterocycloalkylalkyl, substituted or unsubstituted ring Alky
  • the substituted or unsubstituted alkyl group is preferably a substituted or unsubstituted C 1-6 alkyl group; more preferably a substituted or unsubstituted C 1-4 alkyl group;
  • the substituted or unsubstituted cycloalkyl group is preferably a substituted or unsubstituted C 3-8 cycloalkyl group
  • the substituted or unsubstituted heterocycloalkyl group is preferably a substituted or unsubstituted 3-8 membered heterocycloalkyl group
  • the substituted or unsubstituted heterocycloalkyl group is preferably a substituted or unsubstituted one of the following groups: azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, high piperazine Azinyl, morpholinyl, (1S,4S)-2,5-diazabicyclo[2.2.1]heptyl, (1R,4R)-2,5-diazabicyclo[2.2.1]heptyl (1R,4R)-2-oxo-5-azabicyclo[2.2.1]heptyl, (1S,4S)-2-oxo-5-azabicyclo[2.2.1]heptyl, or 4, 7-diazaspiro[2.5]octyl;
  • the substituted or unsubstituted cycloalkylalkyl group is preferably a substituted or unsubstituted C 3-8 cycloalkyl C 1-3 alkyl group;
  • the substituted or unsubstituted heterocycloalkylalkyl group is preferably a substituted or unsubstituted 3-8 membered heterocycloalkyl C 1-3 alkyl group;
  • the substituted or unsubstituted aryl group is preferably a substituted or unsubstituted phenyl group.
  • the substituted or unsubstituted heteroaryl group is preferably a substituted or unsubstituted 5-6 membered heteroaryl group.
  • the substituted or unsubstituted alkyl group is preferably a substituted or unsubstituted C 1-6 alkyl group; more preferably a substituted or unsubstituted C 1-4 alkyl group;
  • the substituted or unsubstituted cycloalkyl group is preferably a substituted or unsubstituted C 3-8 cycloalkyl group
  • the substituted or unsubstituted heterocycloalkyl group is preferably a substituted or unsubstituted 3-8 membered heterocycloalkyl group
  • the substituted or unsubstituted heterocycloalkyl group is preferably a substituted or unsubstituted group such as pyrrolidinyl, piperidinyl, azepanyl, azetidinyl, Oxetanyl, 4-azaspiro[2.5]octyl, or 5-azaspiro[2.5]octyl;
  • the substituted or unsubstituted cycloalkylalkyl group is preferably a substituted or unsubstituted C 3-8 cycloalkyl C 1-3 alkyl group;
  • the substituted or unsubstituted heterocycloalkylalkyl group is preferably a substituted or unsubstituted 3-8 membered heterocycloalkyl C 1-3 alkyl group;
  • the substituted or unsubstituted aryl group is preferably a substituted or unsubstituted phenyl group.
  • the substituted or unsubstituted heteroaryl group is preferably a substituted or unsubstituted 5-6 membered heteroaryl group.
  • each R 10 is independently preferably H, F, Cl, C 1-6 alkyl, halogenated C 1-6 alkyl, halogenated C 1-6 alkoxy, hydroxy C 1-6 alkane Base, amino C 1-6 alkyl, amino C 3-8 cycloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, -CN, -NO 2 , -SR a , -OR a , -OC (O)R a , -OC(O)OR a , -OC(O)NR a R b , -C(O)OR a , -C(O)R a , -C(O)NR a R b , -NR a R b , -NR a C(O)R b , -N(R a )C(O)OR b , -N(R a )C(O)NR a R b , -NR
  • R a and R b are independently substituted, or the R a and R b together with the N atom to which they are attached form a 3-8 membered heterocycloalkyl group, and the heterocycloalkyl group may further contain 1 to 3 selected a hetero atom or a group derived from N, O, S(O) 0-2 or C(O); said heterocycloalkyl group being unsubstituted or optionally substituted by a substituent selected from a C 1-6 alkyl group Replace at any position;
  • each R 10 is independently more preferably H, D, F, Cl, amino, hydroxy, methyl, ethyl, propyl, butyl, isopropyl, isobutyl, t-butyl, or ring.
  • the compound of formula (I) and/or a pharmaceutically acceptable salt is a compound of formula (II) and/or a pharmaceutically acceptable salt thereof:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 9 , V, X and W are as defined above.
  • the compound of formula (I) and/or a pharmaceutically acceptable salt is a compound of formula (III) and/or a pharmaceutically acceptable salt thereof:
  • n 0, 1, 2, or 3;
  • R 1 , R 3 , R 4 , R 5 , X, R a , R b and W are as defined above.
  • n is one.
  • the compound of formula (I) and/or a pharmaceutically acceptable salt is a compound of formula (IV) and/or a pharmaceutically acceptable salt thereof:
  • Z is N or CH;
  • L is CH 2 , CH(CH 3 ), C(CH 3 ) 2 , or u is 0, 1, or 2;
  • v is 0, 1, or 2;
  • R 1 , R 3 , R 4 , R 5 , R 10 , X and W are as described above.
  • L is N
  • u is 1, and v is 1;
  • u is 1, and v is 2;
  • u is 0 and v is 1;
  • u is 0 and v is 2;
  • u is 1, and v is 0;
  • u is 2 and v is 0;
  • Z is N; and R 10 is hydrogen, C 1-6 alkyl, halo C 1-6 alkyl, hydroxy C 1-6 alkyl, amino C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, -OR a , -C(O)OR a , -C(O)R a , -C(O)NR a R b , -S(O) 1- 2 R b , or -S(O) 2 NR a R b ; R a and R b are each independently hydrogen or a C 1-6 alkyl group.
  • the compound of formula (I) and/or a pharmaceutically acceptable salt is a compound of formula (V) and/or a pharmaceutically acceptable salt thereof:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 8 , R 9 , U and W are as defined above.
  • U is CH
  • R 1 is H or methyl
  • R 8 is -NR c R d ;
  • R 8 is a substituted or unsubstituted 3-8 membered heterocycloalkyl; when the 3-8 membered heterocycloalkyl is substituted, the selectivity is 1 to 3 C 1-4 Alkylation, or C 3-6 cycloalkyl substitution at any position;
  • R 9 is C 1-4 alkyl, hydroxy C 1-4 alkyl, C 3-8 cycloalkyl, or 3-8 membered heterocycloalkyl;
  • R 2 is
  • the compound of formula (I') and/or a pharmaceutically acceptable salt is a compound of formula (VI) and/or a pharmaceutically acceptable salt thereof:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 8 , R 9 , U and W are as defined above.
  • U is CH
  • R 8 is -NR c R d ;
  • R 8 is a substituted or unsubstituted 3-8 membered heterocycloalkyl; when the 3-8 membered heterocycloalkyl is substituted, the selectivity is 1 to 3 C 1-4 Alkylation, or C 3-6 cycloalkyl substitution at any position;
  • R 9 is C 1-4 alkyl, hydroxy C 1-4 alkyl, C 3-8 cycloalkyl, or 3-8 membered heterocycloalkyl.
  • the compound of formula (I) and/or a pharmaceutically acceptable salt are optionally the following compounds:
  • the compound of formula (I) and/or a pharmaceutically acceptable salt are optionally the following compounds:
  • the compound of formula (I) and/or a pharmaceutically acceptable salt are optionally the following compounds:
  • the present invention also provides a method for producing the compound represented by the formula (I) or (I'), an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt, which is any of the following method:
  • step 1) 1a Compound 1c is obtained by buchwald coupling reaction with 1b; step 2) oxidation of the methylthio group in compound 1c with m-chloroperoxybenzoic acid (m-CPBA) to sulfoxide to obtain compound 1d; step 3) compound 1d and 1e Or 1e' is reacted under basic conditions to give a compound of formula I or I'.
  • step 1) Compound 1c is obtained by buchwald coupling reaction with 1b
  • step 2) oxidation of the methylthio group in compound 1c with m-chloroperoxybenzoic acid (m-CPBA) to sulfoxide to obtain compound 1d; step 3) compound 1d and 1e Or 1e' is reacted under basic conditions to give a compound of formula I or I'.
  • m-CPBA m-chloroperoxybenzoic acid
  • the conditions and steps may be the conditions and steps of the conventional reaction in the art, and the present invention particularly preferably the following reaction conditions: step 1) under nitrogen protection, in the 1,4-dioxane solvent,
  • step 1) under nitrogen protection, in the 1,4-dioxane solvent
  • the reaction is carried out by the action of a base (1,2-N,N-dimethylethylenediamine, potassium carbonate) and cuprous chloride, and the amount of the agent is preferably 1 to 50 mL/mmol of the compound 1a, and the reaction time is preferably 0-24 hours, the temperature is preferably room temperature to reflux of the solvent, more preferably 80 to 100 ° C, and the molar ratio of the compound 1a and 1b is preferably 1:0.9 to 1:1.5.
  • Step 2 The compound 1d is obtained by oxidizing the compound 1c with m-chloroperoxybenzoic acid in a dichloromethane solvent; the amount of the agent is preferably 1 to 50 mL/mmol of the compound 1c, and the reaction time is preferably 0-24 hours, and the temperature is preferably 0.
  • the molar ratio of compound 1c to m-CPBA is preferably 1:1 to 1:3; in step 3) in toluene, under basic conditions (N,N-diisopropylethylamine or triethylamine), 1d And 1e or Ie' react to obtain a compound of the formula I or I', the amount of the agent is preferably 1 to 50 mL / mmol of the compound 1d, the reaction time is preferably 0-24 hours, the temperature is preferably room temperature to reflux of the solvent, the compound 1d
  • the molar ratio of 1e to base is preferably 1:0.9:1 to 1:2.5:2.5; the molar ratio of compound 1d, 1e' and base is preferably 1:0.9:1 to 1:2.5:2.5.
  • an acidic system such as p-toluenesulfonic acid, hydrochloric acid, hydrogen chloride or trifluoroacetic acid is used, or in the purification process, for example, in the mobile phase of prep-HPLC, the above acidic system is present.
  • the compound of formula I or I' will be the corresponding p-toluenesulfonate, hydrochloride or trifluoroacetate salt and the like.
  • the amino group, the hydroxyl group or the carboxyl group is preferably protected by a protecting group.
  • a protecting group to avoid any side reactions. If the above amino protecting group or hydroxy protecting group is present, it is necessary to undergo a subsequent deprotection step to give a compound of formula I and I'.
  • Any suitable amino protecting group for example, a tert-butoxycarbonyl (Boc) group, can be used to protect the amino group.
  • Boc is used as a protecting group
  • subsequent deprotection reactions can be carried out under standard conditions, for example, p-toluenesulfonic acid/methanol system, dichloromethane/trifluoroacetic acid system, saturated hydrogen chloride ether solution, or trifluoromethanesulfonate
  • the deprotection reaction can be deprotected under standard conditions, for example, sodium hydroxide, potassium hydroxide, lithium hydroxide in tetrahydrofuran, water, and/or
  • the compound of the formula (I) or (I'), a pharmaceutically acceptable salt thereof, can be synthesized by a general chemical method.
  • the preparation of the salt can be carried out by reacting the free base or acid with an equivalent chemical equivalent or an excess of an acid (inorganic or organic acid) or a base (inorganic or organic base) in a suitable solvent or solvent composition.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of an active ingredient and a pharmaceutically acceptable excipient; the active ingredient comprising a compound of formula (I) or (I'), One or more of a conformation, a prodrug, a stable isotope derivative, and a pharmaceutically acceptable salt.
  • the active ingredient may also include other therapeutic agents for cancer, viral infection or autoimmune diseases.
  • the pharmaceutically acceptable excipient may include a pharmaceutically acceptable carrier, diluent, and/or excipient.
  • the pharmaceutical composition can be formulated into various types of dosage unit dosage forms, such as tablets, pills, powders, liquids, suspensions, emulsions, granules, capsules, suppositories, and injections (solutions and suspensions), etc., depending on the purpose of the treatment.
  • dosage unit dosage forms such as tablets, pills, powders, liquids, suspensions, emulsions, granules, capsules, suppositories, and injections (solutions and suspensions), etc.
  • any excipient known and widely used in the art can be used.
  • carriers such as lactose, white sugar, sodium chloride, glucose, urea, starch, calcium carbonate, kaolin, crystalline cellulose, and silicic acid; binders such as water, ethanol, propanol, ordinary syrup, dextrose solution, starch Solution, gelatin solution, carboxymethyl cellulose, shellac, methyl cellulose and potassium phosphate, polyvinylpyrrolidone, etc.
  • disintegrating agents such as dry starch, sodium alginate, agar powder and kelp powder, sodium bicarbonate, carbonic acid Fatty acid esters of calcium, polyethylene sorbitan, sodium lauryl sulfate, monoglyceride stearate, starch and lactose; disintegration inhibitors such as white sugar, glyceryl tristearate, coconut oil and hydrogenation Oil; adsorption promoters such as quatern
  • any excipient known and widely used in the art may be used, for example, a carrier such as lactose, starch, coconut oil, hardened vegetable oil, kaolin and talc, etc.; Such as gum arabic powder, gum tragacanth powder, gelatin and ethanol, etc.; disintegrating agents such as agar and kelp powder.
  • a carrier such as lactose, starch, coconut oil, hardened vegetable oil, kaolin and talc, etc.
  • disintegrating agents such as agar and kelp powder.
  • any excipient known and widely used in the art can be used, for example, polyethylene glycol, coconut oil, higher alcohols, esters of higher alcohols, gelatin and semi-synthetic glycerides, etc. .
  • the solution or suspension may be sterilized (preferably by adding an appropriate amount of sodium chloride, glucose or glycerin, etc.) to prepare an isotonic injection with blood.
  • Any of the commonly used carriers in the art can also be used in the preparation of the injection.
  • water, ethanol, propylene glycol, ethoxylated isostearyl alcohol, polyoxylated isostearyl alcohol, and fatty acid esters of polyethylene sorbitan can be added.
  • the content of the composition in the pharmaceutical composition is not particularly limited and can be selected within a wide range, and is usually from 5 to 95% by mass, preferably from 30 to 80% by mass. %.
  • the administration method of the pharmaceutical composition is not particularly limited.
  • Formulations of various dosage forms can be selected depending on the age, sex and other conditions and symptoms of the patient. For example, tablets, pills, solutions, suspensions, emulsions, granules or capsules are administered orally; injections can be administered alone or in combination with injectable solutions (eg, glucose solutions and amino acid solutions); suppositories are given Drug to the rectum.
  • injectable solutions eg, glucose solutions and amino acid solutions
  • suppositories are given Drug to the rectum.
  • the present invention also provides a compound represented by the formula (I) or (I'), an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt, or the pharmaceutical composition is prepared for Vee1 inhibition Application in the agent.
  • the present invention also provides a compound represented by the formula (I) or (I'), an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt, or the pharmaceutical composition for preparing a cancer Use in sensitizers for chemotherapy or radiotherapy.
  • the sensitizer of chemotherapeutic or radiotherapy refers to the combination of radiation therapy and/or chemotherapy using an anticancer agent, in the field of cancer therapy, additive or synergistically improving these radiotherapy and / or the therapeutic effects of chemotherapy.
  • the present invention also provides a compound represented by the formula (I) or (I'), an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt, or the pharmaceutical composition is prepared for treatment and / or use in a medicament for alleviating a disease associated with Wee1;
  • the invention preferably provides a compound of formula (I), an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt
  • the use of the pharmaceutical composition for the preparation of a medicament for treating and/or preventing a disease associated with Wee1; the disease comprising a tumor and a non-neoplastic disease.
  • the disease is preferably cancer.
  • the present invention preferably uses the compound of the formula (I) or (I'), an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt, or the pharmaceutical composition for the preparation of a treatment And/or ease the application of cancer drugs.
  • the present invention still further provides a compound represented by the formula (I) or (I'), an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt, or the pharmaceutical composition
  • a method of treating cancer comprising: administering a compound of the formula (I) or (I'), an isomer, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt thereof, in a dose required for treatment in a mammal, Or a pharmaceutical composition.
  • Said mammal preferably a human.
  • the present invention still further provides the compound represented by the formula (I) or (I'), an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt, or the pharmaceutical composition It may be used in combination with one or more other classes of therapeutic agents and/or methods of treatment for the treatment and/or alleviation of a related disease mediated by Wee1, preferably a cancer.
  • the present invention still further provides a combined preparation comprising a compound represented by the formula (I) or (I'), an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt, or the drug
  • a combined preparation comprising a compound represented by the formula (I) or (I'), an isomer thereof, a prodrug, a stable isotope derivative or a pharmaceutically acceptable salt, or the drug
  • the compositions are combined with other types of therapeutic agents and/or therapeutic methods for treating cancer.
  • the other kind of therapeutic agent for example, other kinds of therapeutic agents for treating cancer
  • the other kinds of therapeutic agents and/or treatment methods for treating cancer may include, but are not limited to, for cancer treatment: tubulin inhibitors, alkylating agents, topoisomerase I/II inhibitors, Platinum compounds, antibiotics, antimetabolites, hormones and hormone analogues, targeted therapies (eg special kinase inhibitors), immunotherapeutics, interferons, other anticancer agents for cancer therapy and radiotherapy One or more.
  • the tubulin inhibitor may be selected from, but not limited to, a vinblastine series (eg, vinblastine, vincristine, vinorelbine, vindesine), a taxane (docetaxel, One or more of paclitaxel) and eribulin mesylate.
  • a vinblastine series eg, vinblastine, vincristine, vinorelbine, vindesine
  • a taxane docetaxel, One or more of paclitaxel
  • eribulin mesylate eribulin mesylate.
  • the alkylating agent may be selected from one or more of nitrogen mustard, ethyleneimine derivative, methanesulfonate, nitrosourea, and triazene.
  • the topoisomerase I/II inhibitor may be selected from, but not limited to, camptothecin, 10-hydroxycamptothecin, irinotecan, irinotecan metabolite SN-38, topotecan, Sinon One or more of temcon, Exactecan, Karenitecin, 9-nitrocamptothecin, doxorubicin, and dexrazoxane.
  • the platinum compound may be selected from, but not limited to, cisplatin, carboplatin, nedaplatin, oxaliplatin, omalimin, tetraplatinum, isopropylplatinum, spiroplatinum, cis-diamine dihydrate.
  • the antimetabolite may be selected from, but not limited to, a folic acid antagonist, a pyrimidine analog, a purine analog, an adenosine deaminase inhibitor, for example, methotrexate, 5-fluorouracil, fluorourea One or more of glycosides, cytarabine, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, pentastatin and gemcitabine.
  • a folic acid antagonist a pyrimidine analog
  • a purine analog for example, methotrexate, 5-fluorouracil, fluorourea One or more of glycosides, cytarabine, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, pentastatin and gemcitabine.
  • the immunotherapeutic agent may be selected from, but not limited to, an anti-tumor vaccine (for example, a synthetic peptide, a DNA vaccine, and a recombinant virus), an oncolytic virus, a monoclonal antibody (for example, PD-1 monoclonal antibody, PD- L1 monoclonal antibody, CTLA-4 monoclonal antibody, etc.), novel adjuvants, cytokine therapy (eg, IL2 and GM-CSF), chimeric antigen receptor T cell therapy (CAR-T), small molecule immunomodulator, One or more of a tumor microenvironmental modulator and an anti-angiogenic factor.
  • an anti-tumor vaccine for example, a synthetic peptide, a DNA vaccine, and a recombinant virus
  • an oncolytic virus for example, a monoclonal antibody (for example, PD-1 monoclonal antibody, PD- L1 monoclonal antibody, CTLA-4 monoclonal antibody, etc.), novel adju
  • the antibiotic for cancer treatment may be selected from, but not limited to, actinomycin D, doxorubicin, daunorubicin, bleomycin, pilomycin, mitomycin C.
  • actinomycin D actinomycin D
  • doxorubicin daunorubicin
  • bleomycin bleomycin
  • pilomycin pilomycin
  • mitomycin C mitomycin C.
  • arubicin pirarubicin
  • epirubicin epirubicin
  • net statin idarubicin
  • sirolimus sirolimus
  • valrubicin valrubicin
  • the interferon for cancer treatment may be selected from, but not limited to, interferon alpha, interferon alpha-2a, interferon alpha-2b, interferon beta, interferon gamma-1a or interferon gamma- N1 and so on.
  • the cancer includes both metastatic and non-metastatic cancers, as well as family hereditary and sporadic cancers, and may also include solid tumors and non-solid tumors.
  • specific examples of the solid tumor may include, but are not limited to, eyes, bone, lung, stomach, pancreas, breast, prostate, brain (including glioblastoma and medulloblastoma), ovaries (including those Stromal cells, germ cells and mesenchymal cells produced from epithelial cells, bladder, testis, spinal cord, kidney (including adenocarcinoma, nephroblastoma), mouth, lips, throat, oral cavity (including squamous cell carcinoma), nasal cavity , small intestine, colon, rectum, parathyroid gland, gallbladder, bile duct, cervix, heart, inferior gland, bronchus, liver, ureter, vagina, anus, larynx, thyroid (including thyroid cancer and medullary carcinoma), esophagus, nose Pituitary gland, salivary gland, adrenal gland, head and neck intraepithelial neoplasia (including Bowen's disease and Paget's disease
  • the solid tumor is preferably human eye cancer, bone cancer, lung cancer, stomach cancer, pancreatic cancer, breast cancer, prostate cancer, brain cancer (including but not limited to malignant glioma, medulloblastoma), Ovarian cancer, bladder cancer, cervical cancer, testicular cancer, kidney cancer (including but not limited to adenocarcinoma, nephroblastoma), oral cancer (including squamous cell carcinoma), tongue cancer, laryngeal cancer, nasopharyngeal carcinoma, head and neck Cancer, colon cancer, small bowel cancer, rectal cancer, parathyroid cancer, thyroid cancer, esophageal cancer, gallbladder cancer, cholangiocarcinoma, cervical cancer, liver cancer, lung cancer (including but not limited to small cell lung cancer, non-small cell lung cancer), fluff One or more of epithelial cancer, osteosarcoma, Ewing's tumor, soft tissue sarcoma, and skin cancer.
  • brain cancer including but not limited to malignant gli
  • non-solid tumor may include, but are not limited to, lymphocytic leukemia (including acute lymphocytic leukemia, lymphoma, myeloma, chronic lymphocytic leukemia, Hodgkin's lymphoma). , non-Hodgkin's lymphoma, T-cell chronic lymphocytic leukemia, B-cell chronic lymphocytic leukemia), one of myeloid-associated leukemia (including acute myelogenous leukemia, chronic myelogenous leukemia) and AIDs-associated leukemia or A variety.
  • lymphocytic leukemia including acute lymphocytic leukemia, lymphoma, myeloma, chronic lymphocytic leukemia, Hodgkin's lymphoma
  • non-Hodgkin's lymphoma T-cell chronic lymphocytic leukemia
  • B-cell chronic lymphocytic leukemia B-cell chronic lymphocytic
  • the term "optionally substituted by one or more groups at any position" means that any one or more of the hydrogen atoms of the one or more atoms specified on the group are designated by The group is substituted, provided that it does not exceed the normal valence of the specified atom, which is a reasonable substitution that is common in the art at any position.
  • alkyl refers to a saturated straight or branched hydrocarbon group containing from 1 to 20 carbon atoms, preferably from 1 to 10 carbon atoms, more preferably from 1 to 8, from 1 to 4, or from 1 to 3
  • Representative examples of carbon atoms, alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, isobutyl, pentyl, hexyl, g. Base, octyl, decyl, decyl, 4,4-dimethylpentyl, 2,2,4-trimethylpentyl, undecyl, dodecyl, and various isomers thereof Body and so on.
  • cycloalkyl refers to a monocyclic or polycyclic group containing from 3 to 20 carbon atoms which is saturated or partially unsaturated (comprising 1 or 2 double bonds).
  • “monocyclic cycloalkyl” is preferably a 3-10 membered monocycloalkyl group, more preferably a 3-8 membered monocycloalkyl group, for example: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, ring Octyl, cyclodecyl, cyclododecyl, cyclohexenyl.
  • Polycyclic cycloalkyl includes “bridged ring”, “fused cycloalkyl” and “spirocycloalkyl”, and representative examples of “bridged ring” include, but are not limited to, borneol, bicyclo [2.2. 1] heptenyl, bicyclo [3.1.1] heptyl, bicyclo [2.2.1] heptyl, bicyclo [2.2.2] octyl, bicyclo [3.2.2] nonyl, bicyclo [3.3. 1] nonylalkyl, bicyclo[4.2.1]nonanyl and adamantyl, and the like.
  • Fused cycloalkyl embraces a cycloalkyl ring fused to a phenyl, cycloalkyl, or heteroaryl group, including but not limited to: benzocyclobutene, 2,3-di Hydrogen-1-H-indole, 2,3-cyclopentenopyridine, 5,6-dihydro-4H-cyclopentyl[B]thiophene, decalin and the like.
  • “Spirocycloalkyl” refers to a bicyclic group formed by the sharing of one carbon atom by two monocyclic cycloalkyl groups, including but not limited to: spiro[2,5]octyl, spiro[2,4]g Base, snail [4,5] thiol and the like.
  • the polycyclic cycloalkyl group preferably contains from 7 to 12 carbon atoms.
  • a monocyclic cycloalkyl or polycyclic cycloalkyl group can be attached to the parent molecule through any one or two carbon atoms of the ring.
  • heterocycloalkyl refers to a non-aromatic cyclic group consisting of a carbon atom and a heteroatom consisting of a hetero atom selected from nitrogen, oxygen or sulfur, including one or two double bonds, which is a cyclic group.
  • the group may be a monocyclic or polycyclic group, and in the present invention, the number of hetero atoms in the heterocycloalkyl group is preferably 1, 2, 3 or 4, and the nitrogen, carbon or sulfur atom in the heterocycloalkyl group may optionally be Oxidized.
  • the nitrogen atom can optionally be further substituted with other groups to form a tertiary or quaternary ammonium salt.
  • the "monocyclic heterocycloalkyl group” is preferably a 3-10 membered monocyclic heterocycloalkyl group, more preferably a 3-8 membered monocyclic heterocycloalkyl group.
  • aziridine Azacyclobutyl Azacycloheptyl Oxahydrobutyl Pyrrolidinyl Tetrahydrofuranyl Morpholinyl Thiomorpholyl Thiomorpholyl-S-oxide Piperidinyl Piperazinyl High piperazinyl Wait.
  • Polycycloheterocycloalkyl includes "fused heterocycloalkyl", "spiroheterocyclyl” and "bridge heterocycloalkyl”.
  • “Fused heterocycloalkyl” includes a monocyclic heterocycloalkyl ring fused to a phenyl, cycloalkyl, heterocycloalkyl or heteroaryl group, including but not limited to: 2,3 - dihydrobenzofuranyl, 1,3-dihydroisobenzofuranyl, indanyl, 2,3-dihydrobenzo[b]thienyl, dihydrobenzopyranyl, 1, 2,3,4-tetrahydroquinolyl and the like.
  • Spiroheterocyclyl means a bicyclic group formed by two heterocycloalkyl groups or a cycloalkyl group and a heterocycloalkyl group sharing one carbon atom, and spiroheterocyclyl groups include, but are not limited to, 5-aza [2.5 Xinji 4-aza[2.5]octyl 4-aza[2.4]heptyl 4,7-diazaspiro[2.5]octyl Wait.
  • Bridge heterocycloalkyl means a straight-chain group bridged by one or three additional carbon atoms or heteroatoms of any two unlinked ring atoms of a monocyclic heterocycloalkyl group (the linear chain) The group is selected from, but not limited to: -CH 2 -, -CH 2 CH 2 -, -CH 2 O-, -CH 2 NH-, -CH 2 CH 2 CH 2 -), bridged heterocycloalkyl including but not Limited to: (1R, 4R)-2,5-diazabicyclo[2.2.1]heptyl (1S,4S)-2,5-diazabicyclo[2.2.1]heptyl (1S,4S)-2-oxo-5-azabicyclo[2.2.1]heptyl (1R,4R)-2-oxo-5-azabicyclo[2.2.1]heptyl (1S,4R)-2-oxo-5-azabicyclo[2.2.1]heptyl (1S,4R)-2
  • Monocyclic heterocycloalkyl and polycyclic heterocycloalkyl groups can be attached to the parent molecule through any one or two ring atoms on the ring.
  • the above ring atoms specifically refer to carbon atoms and/or nitrogen atoms constituting the ring skeleton.
  • cycloalkylalkyl refers to a linkage between a cycloalkyl group and a parent core structure through an alkyl group.
  • cycloalkylalkyl embraces the definitions of alkyl and cycloalkyl as described above.
  • heterocycloalkylalkyl refers to an alkyl linkage between a heterocycloalkyl group and a parent core structure.
  • heterocycloalkylalkyl embraces the definitions of alkyl and heterocycloalkyl as described above.
  • alkoxy refers to a cyclic or acyclic alkyl group having the number of carbon atoms attached through an oxygen bridge, and includes an alkyloxy group, a cycloalkyloxy group, and a heterocycloalkyloxy group.
  • alkoxy includes the definitions of alkyl, heterocycloalkyl and cycloalkyl as described above.
  • hydroxyalkyl means that any one or more hydrogen atoms on the alkyl group are replaced by a hydroxy group, including but not limited to: -CH 2 OH, -CH 2 CH 2 OH, -CH 2 CH 2 C(CH 3 ) 2 OH, -CH(CH 3 ) 2 OH.
  • alkylthio refers to a cyclic or acyclic alkyl group having the number of carbon atoms attached through a sulfur bridge, the alkylthio group comprising an alkylthio group, a cycloalkylthio group, and a heterocycloalkylsulfide group. base.
  • alkylthio embraces the definitions of alkyl, heterocycloalkyl and cycloalkyl as described above.
  • alkenyl refers to a straight, branched or cyclic non-aromatic hydrocarbon radical containing at least one carbon to carbon double bond. There may be from 1 to 3 carbon-carbon double bonds, preferably one carbon-carbon double bond.
  • C2-4 alkenyl refers to an alkenyl group having 2 to 4 carbon atoms
  • C2-6 alkenyl refers to an alkenyl group having 2 to 6 carbon atoms, including vinyl and propenyl. , butenyl, 2-methylbutenyl and cyclohexenyl.
  • the alkenyl group may be substituted.
  • alkynyl refers to a straight, branched or cyclic hydrocarbon radical containing at least one carbon to carbon triple bond. There may be 1-3 carbon-carbon triple bonds, preferably one carbon-carbon triple bond.
  • C 2-6 alkynyl refers to an alkynyl group having 2 to 6 carbon atoms, and includes ethynyl, propynyl, butynyl and 3-methylbutynyl.
  • aryl refers to any stable 6-20 membered monocyclic or polycyclic aromatic group such as phenyl, naphthyl, tetrahydronaphthyl, indanyl or biphenyl. Wait.
  • heteroaryl refers to an aromatic ring radical formed by the replacement of a carbon atom on at least one ring with a heteroatom selected from nitrogen, oxygen or sulfur, which may be a 5-7 membered monocyclic structure or 7-20.
  • a fused ring structure preferably a 5-6 membered heteroaryl group.
  • the number of heteroatoms is preferably 1, 2 or 3, including but not limited to: pyridyl, pyrimidinyl, piperazinyl, pyridazine-3(2H)-one, furyl, thienyl, thiazolyl , pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, 1,2,5-oxadiazolyl, 1,2,4-oxadiazolyl, 1,3,4-oxadi Azyl, 1,3,4-thiadiazole, 1,2,4-triazolyl, 1,2,3-triazolyl, tetrazolyl, oxazolyl, isoxazolyl, anthracene Sulfhydryl, isodecyl, benzofuranyl, benzothienyl, benzo[d][1,3]dioxolanyl, benzothiazolyl, benzoxazolyl,
  • arylalkyl refers to an alkyl linkage between the aryl group and the parent core structure.
  • arylalkyl embraces the definition of alkyl and aryl as defined above.
  • heteroarylalkyl refers to an alkyl linkage between a heterocycloalkyl group and a parent core structure.
  • heteroarylalkyl embraces the definitions of alkyl and heteroaryl as defined above.
  • halogen means fluoro, chloro, bromo or iodo.
  • haloalkyl refers to an alkyl group optionally substituted by halogen.
  • haloalkyl embraces the definitions of the above halo and alkyl.
  • haloalkoxy refers to an alkoxy group optionally substituted by halogen.
  • haloalkoxy includes the definitions of the above halo and alkoxy.
  • amino means -NH 2
  • alkylamino refers to at least one hydrogen atom is substituted with alkyl amino group, including but not limited to: -NHCH 3, -N (CH 3 ) 2, -NHCH 2 CH 3 , -N(CH 2 CH 3 ) 2 .
  • aminoalkyl means that any one of the hydrogen atoms on the alkyl group is replaced by an amino group, including but not limited to: -CH 2 NH 2 , -CH 2 CH 2 NH 2 .
  • aminoalkyl and alkylamino embrace the definition of alkyl and amino as defined above.
  • aminocycloalkyl means that any one of the hydrogen atoms on the cycloalkyl group is replaced by an amino group, including but not limited to:
  • aminocycloalkyl includes the definitions of the above cycloalkyl and amino groups.
  • nitro refers to -NO 2 .
  • cyano refers to -CN.
  • mercapto refers to -SH.
  • Root temperature as used herein means 15-30 °C.
  • the isotope-substituted derivative includes an isotope-substituted derivative obtained by substituting any hydrogen atom of the formula I with 1-5 deuterium atoms, and an isotope obtained by substituting any carbon atom of the formula I with 1-3 carbon atoms and 14 atoms.
  • prodrug is meant that the compound is converted to the original active compound after metabolism in the body. Typically, the prodrug is inactive or less active than the active parent compound, but can provide convenient handling, administration or improved metabolic properties.
  • “Pharmaceutically acceptable salts” as described herein are discussed in Berge, et al., “Pharmaceutically acceptable salts", J. Pharm. Sci., 66, 1-19 (1977), and for pharmaceutical chemists It is apparent that the salts are substantially non-toxic and provide the desired pharmacokinetic properties, palatability, absorption, distribution, metabolism or excretion, and the like.
  • the compounds of the present invention may have an acidic group, a basic group or an amphoteric group, and typical pharmaceutically acceptable salts include those prepared by reacting a compound of the present invention with an acid, for example, hydrochloride, hydrobromic acid Salt, sulfate, pyrosulfate, hydrogen sulfate, sulfite, bisulfite, phosphate, monohydrogen phosphate, dihydrogen phosphate, metaphosphate, pyrophosphate, nitrate, acetate, Propionate, citrate, octanoate, formate, acrylate, isobutyrate, hexanoate, heptanoate, oxalate, malonate, succinate, suberate, Benzoate, methyl benzoate, phthalate, maleate, methanesulfonate, p-toluenesulfonate, (D,L)-tartaric acid, citric acid, maleic acid, (D,
  • the pharmaceutically acceptable salt thereof may further include: an alkali metal salt such as a sodium or potassium salt; an alkaline earth metal salt such as a calcium or magnesium salt; an organic base salt such as ammonia and an alkane A salt formed from a base such as a hydroxyalkylamine, an amino acid (lysine, arginine) or N-methylglucamine.
  • an alkali metal salt such as a sodium or potassium salt
  • an alkaline earth metal salt such as a calcium or magnesium salt
  • an organic base salt such as ammonia and an alkane A salt formed from a base such as a hydroxyalkylamine, an amino acid (lysine, arginine) or N-methylglucamine.
  • “isomer” means that the compound of formula (I) of the present invention may have asymmetric centers and racemates, racemic mixtures and individual diastereomers, all of which include Stereoisomers, geometric isomers are all included in the present invention.
  • a compound of the formula I or a salt thereof, in stereoisomeric form is a single stereoisomer (enantiomer and diastereomer). Isomers) and mixtures thereof are included within the scope of the invention.
  • the invention also includes individual isomers of the compound or salt represented by Formula I, as well as mixtures with isomers in which one or more chiral centers are inverted.
  • the scope of the invention includes: mixtures of stereoisomers, as well as purified enantiomers or enantiomerically/diastereomerically enriched mixtures.
  • the invention includes mixtures of stereoisomers of all possible different combinations of all enantiomers and diastereomers.
  • the invention includes all combinations and subsets of stereoisomers of all the specific groups defined above.
  • the invention also includes geometric isomers of a compound of formula I or a salt thereof, including cis-isomers.
  • the reagents and starting materials used in the present invention are commercially available.
  • Figure 1 is a graph showing tumor volume changes of a compound 7-2-1 (15 mg/kg, 30 mg/kg, qd) and AZD1775 (60 mg/kg, bid) in a human lung cancer H1299 xenograft model.
  • the structures of all compounds of the invention can be identified by nuclear magnetic resonance ( 1 H NMR) and/or mass spectrometry (MS).
  • the 1 H NMR chemical shift ( ⁇ ) was recorded in PPM (10 -6 ).
  • NMR was performed on a Bruker AVANCE-400 spectrometer. Suitable solvents are deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD), deuterated dimethyl sulfoxide (DMSO-d 6 ), and tetramethylsilane as internal standard (TMS).
  • All of the compounds of the present invention can be separated by high performance liquid chromatography, silica gel column chromatography, thin layer chromatography, or flash column chromatography.
  • High performance liquid chromatography was prepared using Shimadzu LC-20 preparative liquid chromatography column: waters xbridge Pre C18, 10 um, 19 mm x 250 mm.
  • Separation condition 1 Mobile phase A: 0.05% aqueous trifluoroacetic acid, mobile phase B: acetonitrile; mobile phase B 40%, elution time: 20 minutes.
  • Separation condition 2 mobile phase A: 10 mmol/L aqueous ammonium hydrogencarbonate solution, mobile phase B: acetonitrile; gradient elution mobile phase B from 25% to 80%, elution time 30 minutes.
  • Separation condition 3 mobile phase A: 0.05% aqueous hydrochloric acid, mobile phase B: acetonitrile; gradient elution mobile phase B from 75% to 25%, elution time: 30 minutes.
  • Detection wavelength 214 nm & 254 nm; flow rate: 15.0 mL / minute.
  • Each of the compounds of the specific examples 37 to 75 of the present invention can be purified by prep-HPLC.
  • the compound obtained using the separation condition 1 is a trifluoroacetate salt
  • the compound obtained using the condition 2 is a free base
  • the compound obtained using the condition 3 is Hydrochloride.
  • Flash column chromatography Flash column chromatography (Flash column chromatography) (flash system / Cheetah TM) using Agela Technologies MP200, supporting the use of a separation column for Flash columm Silica-CS (80g) , Cat No.CS140080-0.
  • Thin layer chromatography is Yantai Xinnuo Chemical Co., Ltd. with a coating thickness of 0.2 ⁇ 0.03 mm and a size of 20 ⁇ 20 cm.
  • Silica gel column chromatography generally uses Yantai Yellow Sea 200-300 mesh silica gel as a carrier.
  • the eluent or developing agent used in silica gel column chromatography, thin layer chromatography or flash column chromatography is selected from one or more selected from the group consisting of ethyl acetate, methanol, dichloromethane, petroleum ether and n-hexane. .
  • the hydrogen atmosphere described in the examples of the present invention is provided by a hydrogen balloon.
  • Step 1 5-Nitroindole (1 g, 6.2 mmol), dimethylaminochloroethane hydrochloride (1.3 g, 9.3 mmol) and anhydrous potassium carbonate (3.4 g, 25 mmol) of N,N- A mixture of dimethylformamide (DMF) (15 mL) was stirred at 70 ° C for 3 hours. The obtained mixture was cooled to room temperature, then poured into ice water (50 mL), and the mixture was evaporated. Filtration and concentrating, and the residue was purified mjjjjjjjj
  • Step 2 Compound 1.2 (700 mg, 3.0 mmol) was added to a mixture of palladium carbon (100 mg, 10%) in methanol (20 mL), and the reaction system was replaced with hydrogen three times, and the mixture was stirred at room temperature overnight under hydrogen atmosphere. The reaction solution was filtered through Celite to remove the catalyst, and the filtrate was concentrated to give 1-(2-(dimethylamino)ethyl)-1H-indole-5-amine (comp. 1.3, 600 mg, yield: 98%) Oily. m/z: [M+H] + 204.0.
  • Step 1 To a solution of 5-nitroguanidine (4 g, 24.7 mmol) in DMF (100 mL) EtOAc (EtOAc) Stir at room temperature for 2 hours. The reaction solution was then cooled in an ice bath and then 2-bromoethanol (12.3 g, 98.4 mmol). The mixture was stirred at room temperature for 24 hours. The reaction was then diluted with EtOAc. EtOAc (EtOAc m. Compound 1.4 (2.1 g, yield: 41%) was obtained as a colorless oil.
  • Step 2 Dess-Martin reagent (6.2 g, 14.7 mmol) was added portionwise to a solution of compound 1.4 (1 g, 4.85 mmol) in dichloromethane (20 mL). The resulting mixture was allowed to warm to room temperature and stirred for 3 hours. The reaction mixture was washed with EtOAc EtOAc (EtOAc m.
  • Step 3 After a drop of acetic acid was added to a solution of compound 1.5 (400 mg, 1.96 mmol) and azetidine (168 mg, 2.94 mmol) in methanol (10 mL), the reaction mixture was stirred at room temperature for 30 min. Sodium cyanoborohydride (369 mg, 5.88 mmol) was added portionwise to the reaction mixture. The resulting mixture was further stirred at room temperature for 1 hour. Then, the reaction mixture was concentrated to dryness.
  • Step 4 To a mixed solution of compound 1.6 (270 mg, 1.10 mmol) in ethyl acetate (5 mL) and tetrahydrofuran (5 mL) was added palladium carbon (50 mg, 10%), and the mixture was replaced with hydrogen three times, then the mixture was hydrogen The mixture was stirred at room temperature for 3 hours under an atmosphere. The reaction system was filtered through celite, and the filtrate was evaporated to drynessieldielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielielie
  • Step 1 To a solution of 5-nitroindole (1.01 g, 6.2 mmol) in DMF (10.0 mL), NaCI (0.5 g, 60%, 12. The reaction system was stirred at room temperature for 30 minutes, and then 1-Boc-4-methanesulfonyloxypiperidine (1.72 g, 6.2 mmol) was added to the above reaction system, and the reaction mixture was stirred at 100 ° C for 12 hours. Then, the reaction was quenched with water and extracted with ethyl acetate. The organic phase was combined and washed with EtOAc EtOAc (EtOAc m. Rate: 75%) is yellow oil. m/z: [M+H] + 346.0.
  • Step 2 A mixed solution of the compound 2.2 (0.85 g, 2.4 mmol) of trifluoroacetic acid (2.0 mL) and dichloromethane (4.0 mL) was stirred at room temperature for 1 hour, and then the reaction system was concentrated under reduced pressure to give Compound 2.3 (0.55 g, crude) is a yellow oil. m/z: [M+H] + 246.0.
  • Step 3 Compound 2.3 (0.50 g, 2.0 mmol), 37% formaldehyde (0.33 g, 4.0 mmol), triethylamine (0.1 mL), acetic acid (2 drops) of 1,2-dichloroethane (10 mL) The solution was stirred at room temperature for 1 hour. Then, sodium cyanoborohydride (0.39 g, 6.0 mmol) was added to the above reaction system, and the obtained mixture was further stirred at room temperature for 2 hr. The reaction was quenched with aqueous sodium hydroxide (1.0M). The organic phase was combined and washed with EtOAc EtOAc (EtOAc m. : 66%) is a yellow solid. m/z: [M+H] + 260.0.
  • Step 4 To a solution of compound 2.4 (0.35 g, 1.3 mmol) in methanol (10.0 mL) was added palladium carbon (0.05 g, 10%), and the reaction system was replaced with hydrogen three times, then the reaction system was stirred at room temperature under a hydrogen atmosphere. hour. Filtration and concentration under reduced pressure gave 1-(1-methylpiperidin-4-yl)-1H-indole-5-amine (Compound 2.5, 0.10 g, crude). m/z: [M+H] + 230.2.
  • Step 1 5 -Nitroindole (1.62 g, 10.0 mmol) and 4-tetrahydropyranyl methanesulfonate (3.6 g, 20.0 mmol) were dissolved in DMF (20 mL) then EtOAc (EtOAc) g, 20.0 mmol), and the mixture was stirred with EtOAc EtOAc. The residue was purified to silicagel eluting elut elut elut elut elut elut elut elut elut m/z: [M+H] + 247.3.
  • Step 2 Palladium carbon (50 mg, 10%) was added to a solution of the compound 2.14 (950 mg, 3.8 mmol) in methanol (30 mL), and the reaction system was replaced with hydrogen three times, and then stirred under a hydrogen atmosphere for 30 minutes. The mixture was filtered through celite, and the filtrate was evaporated evaporated evaporated. It is a brown oil. m/z: [M+H] + 217.2.
  • Step 1 Acetic anhydride (49.9 mg, 0.49 mmol) was added to a solution of compound 2.3 (0.1 g, 0.41 mmol) and triethylamine (82.4 mg, 0.82 mmol) in dichloromethane (5 mL). The reaction system was stirred at room temperature for 2 hours. After diluting with dichloromethane, the mixture was washed with EtOAc EtOAc.
  • Step 2 Using the synthesis of compound 2.5, step 4, using compound 4.1 (100 mg, 0.35 mmol) to give 1-(4-(5-amino-1H-indol-1-yl)piperidin-1-yl) The ketone (Compound 4.2, 80 mg, Yield: 89%) was a pale yellow solid.
  • Step 1 2,2,2-Trifluoroethyl trifluoromethanesulfonate (284 mg, 1.22 mmol) was added to compound 2.3 (0.1 g, 0.41 mmol) and triethylamine (124 mg, 1.22 mmol) of tetrahydrofuran (In a 10 mL) solution, the reaction system was stirred at 70 ° C overnight. After the reaction mixture was cooled to room temperature, the residue was evaporated tolululululululululululululululululululululululululululululululululu m/z: [M+H] + 288.2.
  • Step 2 Using the synthesis of compound 2.5, step 4, using compound 5.1 (95 mg, 0.29 mmol) to give 1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)-1H- Indole-5-amine (Compound 5.2, 85 mg, Yield: 100%) was a pale yellow solid.
  • Step 2 A mixture of compound 6.1 (3.05 g, 8.1 mmol) in trifluoroacetic acid (4 mL) and dichloromethane (20 mL) was stirred at room temperature for 2 hr then quenched with saturated aqueous sodium hydrogen carbonate and diluted with water The resulting mixture was extracted with dichloromethane. The organic phase was combined and washed with EtOAc EtOAc (EtOAc m. , Yield: 49%) as a pale yellow solid. m/z: [M+H] + 278.2.
  • Step 3 Compound 6.2 (1.1 g, 4.0 mmol) and sodium methoxide (643 mg, 11.9 mmol) in ethanol (20 mL) was stirred at 55 ° C for 2 hr. Then, saturated brine was added to the reaction system, and the mixture was extracted with dichloromethane. The organic phase was combined, dried over anhydrous sodium sulfate m/z: [M+H] + 232.4.
  • Step 4 To a solution of the compound 6.3 (310 mg, 1.3 mmol) in DMF (20 mL) Methyl iodide (285 mg, 2.0 mmol) was stirred at room temperature for 30 min. It was then diluted with ethyl acetate (100 mL) and the organic phase was washed with brine. The organic phase was separated and dried over anhydrous sodium sulfate. m/z: [M+H] + 246.4.
  • Step 5 Palladium carbon (180 mg, 10%) was added to a solution of Compound 6.4 (358 mg) in methanol (15 mL). The reaction system was replaced with hydrogen three times and then stirred under a hydrogen atmosphere for 0.5 hour. The reaction mixture was filtered over EtOAc (EtOAc) m/z: [M+H] + 216.2.
  • Step 6 A solution of lithium tetrahydroaluminum in tetrahydrofuran (1.95 mL, 4.9 mmol, 2.5 M) was added dropwise to a solution of compound 6.5 (275 mg, 1.3 mmol) in methyl t-butyl ether (30 mL). The system was stirred at 60 ° C for 5 hours. The reaction was then quenched with EtOAc EtOAc (EtOAc)EtOAc. The organic phase was separated and dried over anhydrous sodium sulfate, filtered and evaporatedEtOAc. 248 mg, 3-step yield: 96%) as a red solid. m/z: [M+H] + 202.2.
  • Step 1 Isobutyl chloroformate (10.2 mL, 78 mmol) was added dropwise to monomethyl 1,1-cyclopropyldicarboxylate (10 g, 69.4 mmol) and triethylamine (10.8 mL) at -10 °C. A solution of 78 mmol) in tetrahydrofuran (200 mL) was stirred at -10 ° C for 1 hour. The reaction mixture was warmed to 0 ° C.
  • Step 2 Methanesulfonyl chloride (7.4 g, 64.5 mmol) was added dropwise to a solution of compound 7.1 (5.6 g, 43 mmol) and triethylamine (8.7 mL, 86 mmol) in dichloromethane (100 mL) The resulting mixture was stirred at room temperature for 3 hours. The reaction mixture was quenched with EtOAc (EtOAc)EtOAc. ) is a white solid.
  • Step 3 Compound 7.2 (10.7 g, 51.5 mmol) was added to a solution of 5-nitroindole (5.56 g, 34.3 mmol) and cesium carbonate (33.5 g, 103 mmol) in DMF (60 mL) The mixture was stirred at rt., and the mixture was evaporated to EtOAc EtOAc. Purification by silica gel column chromatography (EtOAc /EtOAcEtOAc m/z: [M+H] + 275.2.
  • Step 4 An aqueous solution (10 mL) of lithium hydroxide monohydrate (475 mg, 11.3 mmol) was added dropwise to a solution of Compound 7.3 (1.24 g, 4.5 mmol) in tetrahydrofuran (30 mL). The tetrahydrofuran was removed by concentration, and the aqueous phase was washed with a petroleum ether/ethyl acetate mixed solution (1/1), then adjusted to pH 3 with a saturated aqueous solution of citric acid, and the aqueous phase was extracted with ethyl acetate (10 mL ⁇ 3). The mixture was washed with EtOAc EtOAc m.
  • Step 6 Compound 7.5 (460 mg, 1.39 mmol) and palladium on carbon (100 mg, 10%). The reaction system was replaced with hydrogen three times, and then stirred under a hydrogen atmosphere for 2 hours. Filtration with celite, and the filtrate was concentrated under reduced pressure to give (1-((5-amino-1H-indol-1-yl)methyl)cyclopropyl)aminomethyl tert-butyl ester (Compound 7.6, 308 mg, Rate: 74%) as a brown solid. m/z: [M+H] + 302.2.
  • Step 1 Under ice-cooling conditions, sodium hydrogen (121 mg, 3.0 mmol, 60%) was added to a solution of compound 7.5 (500 mg, 1.5 mmol) in DMF (10 mL), and the reaction system was stirred for 0.5 hr. , the mixture was added to the above reaction system, and the mixture was stirred at room temperature for 2 hours. The reaction mixture was poured into ice water, and the aqueous phase was extracted with ethyl acetate (20 mL ⁇ 3). The organic phase was concentrated, and the residue was purified mjjjjjjjj m/z: [M+H] + 290.2.
  • Step 2 A mixture of compound 7.7 (362 mg, 1.0 mmol) and palladium carbon (100 mg, 10%) in methanol (15 mL) was replaced with hydrogen three times and then stirred at room temperature for 2 hours under hydrogen atmosphere. The reaction solution was filtered through Celite, and the filtrate was evaporated to dryness (diethyldiaminediethyldiethyldiphenyl) (((5-amino-1H-indol-1-yl)methyl)cyclopropyl)(methyl)aminomethyl-tert-butyl ester ( Compound 7.8, 315 mg, yield: 100%) was obtained as a brown solid. m/z: [M+H] + 316.2.
  • Step 2 Compound 8.1 (4 g, 19.4 mmol), dimethylamine hydrochloride (2.4 g, 29.1 mmol), HATU (11.0 g, 29.1 mmol) and N,N-diisopropylethylamine (9.6 mL, 58.2 mmol) of a solution of DMF (50 mL) was stirred at room temperature for 3 hr, and then water (150 mL) was added to the mixture, and the mixture was extracted with ethyl acetate (100 mL ⁇ 3). The mixture was dried over anhydrous sodium sulfate, filtered, evaporated, evaporated, evaporated, evaporated, evaporated. Yellow solid. m/z: [M+H] + 234.2.
  • 6-Bromo-2-aminonaphthalene 500 mg, 2.25 mmol
  • 1-methylpiperazine 270 mg, 2.7 mmol
  • cesium hydroxide hydrate 760 mg, 4.5 mmol
  • dimethyl sulfoxide 5.0 mL
  • the solution was stirred at 120 ° C for 20 hours, then the reaction system was cooled to room temperature, diluted with ice water (10 mL), and the aqueous phase was extracted with dichloromethane (20 mL ⁇ 2). Drying over sodium sulfate, filtration, EtOAc (EtOAc:EtOAc) 9.1, 70 mg, yield: 13%) was a brown solid.
  • EtOAc EtOAc
  • Step 1 2-Fluoro-4-nitrobenzonitrile (2 g, 12.0 mmol), ethyl decylacetate (1.37 g, 11.4 mmol) and triethylamine (5 mL, 36 mmol) were added to acetonitrile (30 mL). The system was stirred at room temperature for 16 hours. The reaction mixture was concentrated under reduced pressure. EtOAcjjjjjjjj
  • Step 2 A mixture of compound 10.1 (600 mg, 2.25 mmol), tert-butyl nitrite (349 mg, 3.38 mmol) and copper bromide (1 g, 4.51 mmol) in acetonitrile (20 mL) was stirred at 65 ° C for 16 h. The reaction solution was concentrated under reduced pressure and poured into water, filtered, and then filtered and evaporated to dryness to afford Compound 10.2 (600 mg, yield: 81%). m/z: [M+H] + 329.8.
  • Step 3 Compound 10.2 (600 mg, 1.82 mmol), 1-methylpiperazine (219 mg, 2.18 mmol), tris(dibenzylideneacetone) dipalladium (83 mg, 0.091 mmol), 1,1'-binaphthyl -2,2'-bisdiphenylphosphine (113 mg, 0.182 mmol) and cesium carbonate (1.18 g, 3.64 mmol) were added to toluene (15 mL), and the reaction was heated under reflux for 16 hours under nitrogen. The reaction mixture was filtered, and the filtrate was evaporated. mjjjjjjjj m/z: [M+H] + 349.8.
  • Step 4 Compound 10.3 (100 mg, 0.28 mmol) and palladium on carbon (50 mg, 10%) were added to ethyl acetate (15 mL). The reaction system was replaced with hydrogen three times, then stirred under a hydrogen atmosphere for 0.5 hour, using silicon. The reaction mixture was filtered through celite, and the filter cake was washed with ethyl acetate, and the filtrate was concentrated under reduced pressure to give ethyl 6-amino-3-(4-methylpiperidin-1-yl)benzo[b]thiophene-2-carboxylate. (Compound 10.4, 90 mg, crude) was obtained as a brown oil. m/z: [M+H] + 319.8.
  • Step 1 Compound 10.1 (500 mg, 1.88 mmol) and lithium chloride (995 mg, 23.5 mmol) were added to dimethyl sulfoxide (10 mL), and the reaction was stirred at 160 ° C for 5 hours. After the reaction mixture was cooled, EtOAc (EtOAc m. Compound 10.5 (200 mg, yield: 55%) was obtained as a brown solid. m/z: [M+H] + 194.8.
  • Step 2 Compound 10.5 (200 mg, 1.03 mmol), nitrogen mustard hydrochloride (297 mg, 1.54 mmol) and potassium carbonate (569 mg, 4.12 mmol) were added to tert-butanol (15 mL). Stir at °C for 3 hours. The reaction mixture was filtered, and then evaporated, evaporated, mjjjjjjj m/z: [M+H] + 277.8.
  • Step 3 Compound 10.6 (40 mg, 0.144 mmol), iron powder (40 mg, 0.72 mmol), ammonium chloride (39 mg, 0.72 mmol) and water (1 mL) were added to ethanol (15 mL) and the reaction was heated to reflux for 2 hours. . The reaction solution was then cooled to room temperature, filtered, and the filtrate was concentrated. The residue was washed with methylene chloride and methanol (10/1), and concentrated to give 3-(4-methylpiperazin-1-yl)benzene. And [b]thiophene-6-amine (compound 10.7, 34 mg, yield: 95%) was a dark brown liquid. m/z: [M+H] + 247.8.
  • Step 2 A solution of the compound 11.1 (82 mg, 0.28 mmol) and palladium carbon (40 mg, 10%) in methanol (10.0 mL) was replaced with hydrogen three times and then stirred at room temperature for 2 hours under a hydrogen atmosphere. The reaction solution was filtered through Celite, and then filtered and evaporated to ethyldiamine. %) is a brown solid.
  • Step 2 Palladium carbon (50 mg, 10%) was added to a solution of the compound 12.1 (760 mg, 2.9 mmol) in methanol (30 mL), and the reaction system was replaced with hydrogen three times, and then stirred under a hydrogen atmosphere for 30 minutes. The mixture was filtered through celite, and the filtrate was evaporated to dryness to give 3-(1-methylpiperidin-4-yl)-1H-indole-6-amine (Compound 12.2, 510 mg, yield: 77%) Brown oil. m/z: [M+H] + 230.2.
  • Step 2 To a suspension of compound 13.1 (1.76 g, 7.33 mmol) in acetic anhydride (20 mL) was added sodium acetate (2.39 g, 29.2 mmol), and the reaction was stirred at 130 ° C for 4 hours, then cooled to The reaction was quenched with EtOAc (EtOAc m. After the residue was cooled, a yellow solid was crystallised, filtered, and filtered, washed with petroleum ether and dried in vacuo to give compound 13.2 (1.05 g, yield: 55%) as a brown solid. m/z: [M+H] + 263.4.
  • Step 3 Compound 13.2 (200 mg, 0.76 mmol), 1-methylpiperazine (305 mg, 3.05 mmol) was dissolved in acetic acid (5 mL), and the reaction was subjected to microwave reaction at 150 ° C for 0.5 hour. The reaction mixture was concentrated under reduced vacuolululululululululululululululululululu m/z: [M+H] + 303.4.
  • Step 4 To a solution of the compound 13.3 (20 mg, 0.07 mmol) in methanol (5OmL), EtOAc (5 mg) was applied, and the system was replaced with hydrogen for 3 times and then stirred at room temperature for 1 hour under a hydrogen atmosphere. Filtration and concentration of the filtrate under reduced pressure gave 3-(4-methylpiperazin-1-yl)-1H-indole-6-amine (Compound 13.4, 16 mg, yield: 89%) as a brown solid. m/z: [M+H] + 231.2.
  • Step 1 To a solution of the compound 13.3 (110 mg, 0.36 mmol) in methanol (10.0 mL) was added triethylamine (120 mg, 1.10 mmol), and the reaction mixture was stirred at 80 ° C for 3 hours. The reaction system was concentrated under reduced pressure to give compound 13.5 (yield: yield: 100%). m/z: [M+H] + 261.4.
  • Step 2 To a solution of the compound 13.5 (94 mg, 0.36 mmol) in tetrahydrofuran (5.0 mL), sodium hydrogen (60%, 20 mg, 0.47 mmol) was added portionwise, and the reaction system was stirred at 0 ° C for 0.5 hour. Methyl iodide (80 mg, 0.54 mmol) was then stirred at 0 °C for 1 hour. The reaction system was concentrated under reduced pressure. EtOAc m. m/z: [M+H] + 275.4.
  • Step 3 Raney nickel (20 mg) was added to a solution of the compound 13.6 (45 mg, 0.16 mmol) in methanol (5.0 mL), and the reaction system was replaced with hydrogen three times, and then stirred for 1 hour under a hydrogen atmosphere. Filtration and concentration of the filtrate under reduced pressure gave 1-methyl-3-(4-methylpiperazin-1-yl)-1H-indole-6-amine (Compound 13.7, 40 mg, yield: 100%) as a brown solid. . m/z: [M+H] + 245.2.
  • Step 1 To a solution of 6-nitroguanidine (500 mg, 3.1 mmol) in toluene (10.0 mL) was added 2-bromopyridine (410 mg, 2.6 mmol) and cuprous iodide (25 mg, 0.13 mmol). Potassium phosphate (1.32 g, 6.2 mmol), N,N'-dimethylethylenediamine (55 mg, 0.63 mmol), then the reaction was stirred at 110 ° C for 16 hours. The reaction mixture was concentrated under reduced pressure. EtOAcjjjjjjj m/z: [M+H] + 240.2.
  • Step 2 Raney nickel (20 mg) was added to a solution of the compound 14.1 (130 mg, 0.54 mmol) in methanol (10.0 mL), and the reaction system was replaced with hydrogen for 3 times, and then stirred for 1 hour under a hydrogen atmosphere. Filtration and concentration of the filtrate under reduced pressure gave 1-(pyridin-2-yl)-1H-indole-5-amine (Compound 14.2, 113 mg, yield: 99%) as a brown solid. m/z: [M+H] + 210.3.
  • Step 3 A mixture of compound 15.2 (210 mg, 0.77 mmol) and palladium carbon (60 mg, 5%) in methanol (15 mL) was replaced with hydrogen three times. The reaction system was then stirred at room temperature for 1 hour under a hydrogen atmosphere. The solution was filtered, and the filtrate was concentrated under reduced pressure to give 1-methyl-4-(4-methylpiperazin-1-yl)-1H-indole-6-amine (Compound 15.3, 205 mg, yield: 100%) solid.
  • Step 1 N-Bromosuccinimide (2.12 g, 12 mmol) was added to a solution of 7-nitroquinoline (1.74 g, 10 mmol) in acetic acid (20 mL). The reaction system was then stirred at 80 ° C for 2 hours. After the reaction mixture was cooled to room temperature and stood still for 2 hr, a white precipitate was obtained, which was filtered and washed with petroleum ether and dried in vacuo to afford compound 16.1 (2.10 g, yield: 84%) as white solid.
  • Step 2 Compound 16.1 (2.1 g, 8.33 mmol) was dissolved in dimethyl sulfoxide (20 mL), followed by 1-methylpiperazine (0.9 g, 9 mmol), potassium carbonate (2.2 g, 16 mmol) and iodine Cuprous (152 mg, 0.8 mmol). After the reaction system was replaced with nitrogen for 3 times, it was stirred at 120 ° C for 3 hours under a nitrogen atmosphere, and then the reaction liquid was poured into ice water, and the aqueous phase was extracted with ethyl acetate (20 mL ⁇ 3), and the organic phase was combined, followed by water, The mixture was washed with EtOAc EtOAc EtOAc.
  • Step 3 Compound 16.2 (540 mg, 1.99 mmol) and Raney nickel (60 mg) were taken in methanol (15 mL). The reaction system was replaced with hydrogen three times. The reaction system was then stirred at room temperature for 1 hour under a hydrogen atmosphere. Filtration and concentration of the filtrate gave 3-(4-methylpiperazin-1-yl)quinolin-7-amine (Compound 16.3, 480 mg, yield: 100%) as a brown solid.
  • Step 1 2-Chloro-6-nitroquinoline (300 mg, 1.44 mmol) was added to a solution of 1-methylpiperazine (5 mL) in ethanol (20 mL). The reaction solution was cooled to room temperature, concentrated, evaporated, evaporated, evaporated.
  • Step 2 Compound 16.4 (100 mg, 0.367 mmol), palladium carbon (30 mg, 10%) was added to methanol (15 mL), and the reaction system was replaced with hydrogen three times, and then stirred at room temperature for 2 hours under a hydrogen atmosphere. The reaction liquid was filtered, and the filtrate was evaporated to dryness, mjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjj
  • Step 2 Raney nickel (20 mg) was added to a solution of the compound 17.1 (73 mg, 0.33 mmol) in methanol (5 mL), and the reaction system was replaced with hydrogen three times, and then stirred for 1 hour under a hydrogen atmosphere. Filtration and concentration of the filtrate under reduced pressure gave 1-(oxetane-3-yl)-1H-indole-6-amine (Compound 17.2, 60 mg, yield: 95%) as a yellow solid. m/z: [M+H] + 189.2.
  • step 1 The 3-iodooxetane in step 1 was replaced with methyl iodide using the synthesis of compound 17.2 to give 1-methyl-1H-indol-6-amine (compound 17.3). m/z: [M+H] + 147.2.
  • Step 1 4-Bromo-6-nitro-1H-indole (200 mg, 0.83 mmol), cyclopropylboronic acid (143 mg, 1.66 mmol), sodium carbonate (176 mg, 1.66 mmol), copper acetate (151 mg, 0.83)
  • a solution of 2,2-bipyridine (130 mg, 0.83 mmol) in dichloromethane (5 mL) was evaporated.
  • the reaction was then cooled to room temperature and diluted with dichloromethane and the organic phase was washed with water. The organic phase was separated, dried and evaporated tolululululululululululululululu
  • Steps 2 & 3 using the synthesis of Compound 15.3, Step 2 and Step 3, using compound 18.1 to give 1-cyclopropyl-4-(4-methylpiperazin-1-yl)-1H-indole-6-amine ( Compound 18.3) was a brown solid.
  • Step 1 A suspension of 4-bromo-6-nitro-1H-indole (300 mg, 1.24 mmol), bromoethanol (467 mg, 3.73 mmol) and potassium carbonate (176 mg, 1.66 mmol) in DMF (10 mL) Stir at 100 ° C overnight. Then, the reaction system was cooled to room temperature, poured into ice water, and the aqueous phase was extracted with ethyl acetate, and the organic phase was washed successively with water and brine. The organic phase was separated, dried and purified mjjjjjjjjjjj
  • Steps 2 & 3 Using the synthesis of Compound 15.3 Step 2 and Step 3, the compound 19.1 is reacted to give 2-(6-amino-4-(4-methylpiperazin-1-yl)-1H-indol-1-yl. Ethanol (Compound 19.3) is a brown solid.
  • Step 1 To a solution of 4-bromo-6-nitro-1H-indole (200 mg, 0.83 mmol) and di-di-di-butyl-di- butyl ester (271 mg, 1.24 mmol) in dichloromethane (10 mL) Aminopyridine (10 mg, 0.083 mmol), and the reaction was stirred at room temperature for 1 hour. Then, the reaction mixture was concentrated to dryness crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal crystal
  • Step 2 Compound 20.1 (140 mg, 0.41 mmol), palladium acetate (4.6 mg, 0.02 mmol), 1-methylpiperazine (41 mg, 0.41 mmol), cesium carbonate (200 mg, 0.62 mmol) and 2-dicyclohexyl
  • a mixture of phosphorus-2',6'-diisopropoxy-1,1'-biphenyl (19.3 mg, 0.04 mmol) in 1,4-dioxane (10 mL) was replaced with nitrogen three times and then reacted The system was heated to 110 ° C and stirred for 2 hours. The reaction system was cooled to room temperature and then filtered over Celite. The filter cake was washed with ethyl acetate. The filtrate was concentrated. (110 mg, yield: 74%) was obtained as a brown solid. m/z: [M+H] + 361.2.
  • Step 3 using the synthesis method of the compound 15.3 step 3, using the compound 20.2 to obtain 6-amino-4-(4-methylpiperazin-1-yl)-1H-indole-1-carboxylic acid tert-butyl ester (compound) 20.3) is a brown solid.
  • Step 1 Iron powder (2.2 g, 39.2 mmol) was added to a solution of compound 15.1 (2 g, 7.84 mmol) and ammonium chloride (629 mg, 11.8 mmol) in acetic acid (20 mL). The reaction mixture was filtered with EtOAc (EtOAc). Brown solid. m/z: [M+H] + 225.0.
  • Step 3 a solution of n-butyllithium in tetrahydrofuran (2.5 M, 2.5 mL, 6.28 mmol) was added dropwise to a solution of compound 21.2 (680 mg, 2.09 mmol) in anhydrous tetrahydrofuran (20 mL). After stirring at -78 ° C for 1 hour, a solution of 1-methyl-4-piperidone (284 mg, 2.50 mmol) in anhydrous tetrahydrofuran (1 mL) was added dropwise to the above reaction mixture, and stirring was continued at this temperature. After 2 hours, the reaction was quenched with EtOAc EtOAc EtOAc. (268 mg, yield: 36%) was a brown liquid. m/z: [M+H] + 360.0.
  • Step 4 Compound 21.3 (268 mg, 0.75 mmol) was dissolved in EtOAc (EtOAc) (EtOAc) It is a brown solid. m/z: [M+H] + 242.0.
  • Step 1 Compound 1-1 (222 mg, 1.0 mmol), Compound 1-2 (215 mg, 1.0 mmol), cuprous iodide (191 mg, 1.0 mmol), anhydrous potassium carbonate (276 mg, 2.0 mmol) and N N-Dimethylethylenediamine (88 mg, 1 mmol) was added to 1,4-dioxane (20 mL), and the mixture was warmed to 100 ° C and stirred overnight. The reaction system was then cooled to room temperature, and the reaction mixture was filtered. EtOAcjjjjjjjjjjj %) is a brown solid. m/z: [M+H] + 358.2.
  • Step 2 m-Chloroperoxybenzoic acid (68 mg, 0.34 mmol) was added to a solution of Compound 1-3 (120 mg, 0.34 mmol) in dichloromethane (10 mL). Dichloromethane was removed to give 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylsulfinyl)-1H-pyrazole[3 , 4-d]pyrimidin-3(2H)-one (Compound 1-4, 125 mg, Yield: 100%) was obtained as a white solid. m/z: [M+H] + 374.2.
  • Trifluoroacetic acid (2 mL) was added dropwise to a solution of Compound 2-1 (130 mg, 0.21 mmol) in dichloromethane (10 mL). The reaction mixture was stirred at room temperature for 2 hr. 1) Purification to give 2-allyl-6-((1-((1-aminocyclopropyl)methyl)-1H-indol-5-yl)amino)-1-(6-(2-hydroxyl) Prop-2-yl)pyridin-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one trifluoroacetate (Compound 1-1-2, 46.7 mg, Rate: 36%) is a yellow solid.
  • the compound 2-2 is reacted to obtain 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(( 1-((1-(Methylamino)cyclopropyl)methyl)-1H-indol-5-yl)amino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)- Ketone trifluoroacetate (Compound 1-1-3).
  • Step 1 Add ferrocene dichloride to a solution of compound 1-1-1 (210 mg, 0.41 mmol) and ammonium formate (52 mg, 0.82 mmol) in 1,4-dioxane (10 mL) at room temperature. Palladium (34 mg, 0.041 mmol) was replaced with nitrogen three times, then the reaction was slowly warmed to 100 ° C and stirred overnight. The reaction mixture was cooled to room temperature, filtered, and the filtered cake was washed with EtOAc. ) is a brown solid.
  • Step 2 To a solution of Compound 2-1 (110 mg, 0.233 mmol) in DMF (5 mL) H, propylpropyl bromide (94 mg, 0.7 mmol) was added and the mixture was stirred at room temperature overnight. Then, the reaction liquid was cooled to 0 ° C, and the reaction was quenched with ice water. The solution was extracted with dichloromethane (5 mL ⁇ 3).
  • the Wee1 enzyme catalysis test is carried out using the ATP-Glo Max luminescence detection kinase kit (Promega). Kinase activity was assessed by quantitative detection of the amount of ATP retained in the solution following the kinase reaction. The luminescent signal in the test is proportional to the amount of ATP and inversely proportional to the kinase activity.
  • the concentration of the compound in the test ranged from 0.5 nM to 30 ⁇ M. The compound was dissolved in 10% DMSO, and 5 ⁇ L of the solution was added to 50 ⁇ L of the reaction, and the concentration of DMSO in the final reaction was 1%. The reaction was carried out at 30 ° C for 50 minutes.
  • the 50 ⁇ L reaction mixture contained 40 mM Tris, pH 7.4, 10 mM MgCl 2 , 0.1 mg/ml BSA, 2 mM DTT, 0.1 mg/ml Poly(Lys, Tyr) substrate, 10 ⁇ M ATP and Wee1.
  • 50 ⁇ L of ATP-Glo Max luminescence assay kinase assay solution (Promega) was added and incubated for 15 minutes at room temperature. The luminescent signal was measured using a microplate reader.
  • a known Wee1 inhibitor was added as a positive control. Luminance data was analyzed using Graphpad software.
  • Non-linear regression analysis was used to plot the % activity value and the corresponding series compound concentration dose-effect curve.
  • IC 50 values determined by the concentration causing half-maximal percentage activity.
  • cellular assays are used to assess the biological activity of a compound. This trial was conducted using the human colon adenocarcinoma cell line WiDr. The activity of a specific Wee1 inhibitor was evaluated using the p-CDK1Y15 ELISA assay. The detailed test method is described as follows:
  • WiDr cells were cultured in Dulbecco's Modified Eagle's medium containing 10% FBS at 37 ° C and 5% CO 2 .
  • the compound concentration ranged from 0.5 nM to 30 ⁇ M.
  • the compound was diluted and added to a 24-well plate and incubated with the cells for 24 hours.
  • DMSO was used as a negative control.
  • a known Wee1 inhibitor was added as a positive control in some experiments.
  • the manufacturer's instructions in the p-CDK1Y15 assay, cells were lysed and subjected to a colorimetric ELISA kit test to determine the amount of p-CDK1Y15. The optical density is measured using a spectrophotometer. OD data analysis using Graphpad Software, curve fitting to obtain IC 50 values of the compounds.
  • the biological activity of the compound is evaluated using a cell test method.
  • MG63 ATCC CRL-1427
  • a human osteosarcoma cell line cultured in a 96-well plate of Dulbecco's Modified Eagle's medium, supplemented with 10% fetal bovine serum and 1% (v/v) L-glutamine, cultured at 37 °C and 5% CO 2 .
  • Compound concentrations ranged from 4.5 nM to 30 [mu]M.
  • the Wee1 inhibitor stock solution was dissolved in DMSO and added to the indicated concentration medium and incubated for 72 hours. Negative control cells were treated only with vehicle. In some experiments, a known Wee1 inhibitor was added as a positive control.
  • Cell viability was measured using Cell Counting Kit-8 (CCK-8, Sigma-Aldrich) under the direction of the product specification. Using Graphpad software for data analysis, and IC 50 values obtained compound and the fitted curve.
  • Example 4 In vivo pharmacodynamic experiment of human lung cancer H1299 xenograft tumor model
  • mice BALB/c nude mice, 24, 7-8 weeks, 16-21 g, purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd.
  • H1299 cell line (5 ⁇ 10 6 cells/cell) was inoculated subcutaneously into the right side of the experimental mice.
  • the inoculation amount of each mouse was 0.1 mL of PBS: Matrigel (volume ratio 1:1), and the tumor was observed regularly.
  • the growth condition was randomized according to tumor size and mouse body weight when the tumor grew to about 100 mm 3 , and was administered according to the administration schedule (Table 1). During the whole experiment, the body weight and tumor were measured 2 to 3 times per week. size.
  • the tumor volume change curves of the four experimental groups are shown in Figure 1.
  • the results show that compared to the positive control AZD1775, the present compound 7-2-1 is in the case of lower doses and longer dosing intervals in human lung cancer.
  • the H1299 xenograft model also showed better tumor inhibition.
  • the positive control in Biological Examples 1, 2, 3 and 4 of the present invention is AZD1775, chemical name: 2-allyl-1-(6-(2-hydroxyprop-2-yl)pyridin-2-yl) -6-((4-(4-Methylpiperazin-1-yl)phenyl)amino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (2-allyl- 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((4-(4-methylpiperazin-1-yl)phenyl)amino)-1H-pyrazolo[3,4-d ] pyrimidin-3 (2H)-one), the synthesis method can be referred to WO2007126122A1 Example 9.

Abstract

提供了一种如式(I)或(I')所示的吡唑并[3,4-d]嘧啶-3-酮衍生物和/或其药学上可接受的盐,以及含有如式(I)或(I')所示化合物的组合物和/或其药学上可接受的盐,制备方法和其作为Wee1抑制剂的用途及其作为癌症的化学疗法或放射性疗法的增敏剂中的用途;该化合物可以有效的抑制Wee1及相关信号通路,具有良好的治疗和/或缓解癌症的作用。

Description

吡唑并[3,4-d]嘧啶-3-酮衍生物、其药物组合物及应用
本申请要求申请日为2017年8月24日的中国专利申请CN201710737296.9、申请日为2017年9月20日的中国专利申请CN201710849306.8、申请日为2017年12月5日的中国专利申请CN201711263908.1、申请日为2018年3月15日的中国专利申请CN201810213347.2的优先权、申请日为2018年5月29日的中国专利申请CN201810531630.X。本申请引用上述中国专利申请的全文。
技术领域
本发明涉及一种可以抑制Wee1及其信号通路的吡唑并[3,4-d]嘧啶-3-酮衍生物及其药学上可接受的盐,包含所述衍生物及其药学上可接受盐的药物组合物,以及所述衍生物及其药学上可接受盐在治疗由Wee1介导的疾病及制备用于治疗Wee1介导的疾病的药物方面的应用。
背景技术
Wee1(Wee1G2检测点激酶;基因编号:7465)是丝氨酸/苏氨酸蛋白激酶家族中的一员,可直接磷酸化细胞周期蛋白依赖性激酶1(CDK1),磷酸化位点为CDK1的酪氨酸15残基,该位点是一个抑制性磷酸化位点,对CDK1活性起消极调节作用。G2检测点的激活主要是通过抑制有丝***,促进细胞周期蛋白B-CDK1复合物。正常细胞在G1逮捕期内对损伤的DNA进行修复,然而癌细胞G1-S检测点常常缺失,需要依靠G2-M检测点的功能来进行DNA修复。例如,P53缺陷肿瘤细胞缺乏G1检测点的功能,因而依赖G2检测点作为细胞周期停滞对DNA损伤进行应答反应。DNA受损后,G2检测点阻止受损细胞进入有丝***,进而保护其免受有丝***灾难及细胞凋亡。Wee1是G2检测点功能发挥中必不可少的因素。通过Wee1抑制剂来废除G2检测点可能选择性的使P53缺陷性癌细胞对DNA损伤敏感,避免对周围正常组织产生影响。Wee1也调节S期的CDK活性,阻止正常的S期进程中DNA损伤的诱导。此外,Wee1在同源重组(HR)修复中起积极调解作用,同源重组修复是DNA双链断裂修复的重要路径。
在多种不同类型的癌症中可见Wee1的上调,包括肝细胞癌(Masaki,et al,2003),乳腺癌,***,肺癌(Iom,et al,2009),鳞状细胞癌(Magnussen,et al,2013),神经胶质瘤DIPG(Mueller,et al,2014),恶性胶质瘤(Mir,et al,2010;Music,et al,2016),成神经管细胞瘤(Harris,et al,2014),白血病(Tibas,et al,2012;Porter,et al,2012),黑色素瘤(Magnussen,et al,2012),及卵巢癌(Slipicevic,et al,2014)。此外,Wee1的高度表达与多种类型癌症的不良预后有关。抑制Wee1可引起一些P53失活肿瘤细胞的凋亡。抑制Wee1可使对化疗及放疗产生抵抗的癌细胞变的敏感。最新的研究(Pfister,et al,2015)展示了合成致死与H3K36me3缺失、部分癌细胞表观遗传学改变及Wee1抑制之间的相互作用,从而对Wee1抑制和更精确的靶向基因改变明确的癌症患者之间的关系提供了强有力的证据。
因而,目前Wee1成为癌症治疗领域高度引人注目的治疗靶点。除了已有的对Wee1的研究外,仍然还有很多机遇去扩展它的应用并从中获益。目前尚无以Wee1作为治疗靶点的药物上市。本发明所描述的化合物、其组合物和应用方法将有助于Wee1抑制剂的发展,满足临床未满足的用药需求。
发明内容
本发明所要解决的技术问题在于,提供了一种新型吡唑并[3,4-d]嘧啶-3-酮衍生物、其制备方法、药物组合物及应用。本发明的吡唑并[3,4-d]嘧啶-3-酮衍生物对Wee1及相关信号通路具有良好的抑制作用,可以有效治疗和/或缓解癌症。
本发明提供了一种如式(I)所示化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐;
Figure PCTCN2018101290-appb-000001
其中,W为N或CR 7
X为CR 8、或N;Y为NR 9、CHR 8、-C(R 8)=C(R 11)-、O、或S;或者,Y为-N=C(R 11)-;
U和V分别选自N或CH,并且U和V不同时为N;或者,U和V分别选自N或CR 8,并且U和V不同时为N;
R 1为氢、卤素、C 2-6炔基、C 2-6烯基、芳基、环烷基、杂环烷基、杂芳基或取代的烷基;所述烷基被取代时,可选择性地被如下一个或多个基团取代在任意位置:卤素、卤代烷氧基、氨基环烷基、-SR a、-OR a、-OC(O)R a、-OC(O)NR aR b、-C(O)OR a、-C(O)R a、-C(O)NR aR b、-NR aR b、-NR aC(O)R b、-NR aS(O) 2R b、-S(O) 1-2R b、-S(O) 2NR aR b和-NR aS(O) 2NR aR b
R 2为烷基、卤代烷基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基;
R 3为氢、卤素、-CN、-NO 2、-SR c、-OR c、-OC(O)R c、-OC(O)OR c、-OC(O)NR cR d、-C(O)OR c、-C(O)R c、-C(O)NR cR d、-NR cR d、-NR cC(O)R d、-N(R c)C(O)OR d、-N(R c)C(O)NR cR d、-NR cS(O) 2R d、-NR cC(=NH)R d、-NR cC(=NH)NR cR d、-S(O) 1-2R c、-S(O) 2NR cR d、-NR cS(O) 2NR cR d、取代或未取代的烷基、取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的芳基、取代或未取代的杂芳基、取代或未取代的环烷基烷基、取代或未取代的杂环烷基烷基、取代或未取代的芳基烷基、取代或未取代的杂芳基烷基、取代或未取代的烯基、取代或未取代的炔基;所述烷基、环烷基、杂环烷基、芳基、杂芳基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基被取代时,可选择性地被如下一个或多个基团取代在任意位置:卤素、烷基、卤代烷基、卤代烷氧基、羟基烷基、氨基烷基、氨基环烷基、烯基、炔基、-CN、-NO 2、-SR a、-OR a、-OC(O)R a、 -OC(O)OR a、-OC(O)NR aR b、-C(O)OR a、-C(O)R a、-C(O)NR aR b、-NR aR b、-NR aC(O)R b、-N(R a)C(O)OR b、-N(R a)C(O)NR aR b、-NR aS(O) 2R b、-NR aC(=NH)R b、-NR aC(=NH)NR aR b、-S(O) 1-2R b、-S(O) 2NR aR b和-NR aS(O) 2NR aR b
R 4、R 5、R 6和R 7分别独立地为氢、卤素、羟基、氰基、硝基、巯基、氨基、烷基、烷氧基、烷硫基、卤代烷基、卤代烷氧基、C 2-6炔基、C 2-6烯基、芳基、环烷基、杂环烷基、杂芳基、-OC(O)R c、-OC(O)OR c、-OC(O)N(R c) 2、-C(O)OR c、-C(O)R c、-C(O)N(R c) 2、-N(R c) 2、-NHC(O)R c、-NHC(O)OR c、-NHC(O)N(R c) 2、-NHS(O) 2R c、-S(O) 0-2R c或-S(O) 2N(R c) 2
R 3和R 4为独立取代基,或者,R 3和R 4与其所连接的环原子一起形成A环,所述A环为取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的苯基、或取代或未取代的5-6元杂芳基;所述A环被取代时,可选择性地被如下一个或多个基团取代在任意位置:氧代基、硫代基、卤素、-CN、-NO 2、-SR c、-OR c、-OC(O)R c、-OC(O)OR c、-OC(O)NR cR d、-C(O)OR c、-C(O)R c、-C(O)NR cR d、-NR cR d、-NR cC(O)R d、-N(R c)C(O)OR d、-N(R c)C(O)NR cR d、-NR cS(O) 2R d、-NR cC(=NH)R d、-NR cC(=NH)NR cR d、-S(O) 1-2R c、-S(O) 2NR cR d、-NR cS(O) 2NR cR d、取代或未取代的烷基、取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的芳基、取代或未取代的杂芳基、取代或未取代的环烷基烷基、取代或未取代的杂环烷基烷基、取代或未取代的芳基烷基、取代或未取代的杂芳基烷基、取代或未取代的烯基、取代或未取代的炔基;所述烷基、环烷基、杂环烷基、芳基、杂芳基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基被取代时,可选择性地被如下一个或多个基团取代在任意位置:卤素、烷基、卤代烷基、卤代烷氧基、羟基烷基、氨基烷基、烯基、炔基、-CN、-NO 2、-SR c、-OR c、-OC(O)R c、-OC(O)OR c、-OC(O)N(R c) 2、-C(O)OR c、-C(O)R c、-C(O)N(R c) 2、-N(R c) 2、-NHC(O)R c、-NHC(O)OR c、-NHC(O)N(R c) 2、-NHS(O) 2R c、-NHC(=NH)R c、-NHC(=NH)N(R c) 2、-S(O) 1-2R c、-S(O) 2N(R c) 2和-NHS(O) 2N(R c) 2
R 8、R 9或R 11分别独立地为氢、-C(O)OR c、-C(O)R c、-C(O)NR cR d、-S(O) 1-2R c、-S(O) 2NR cR d、取代或未取代的烷基、取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的芳基、取代或未取代的杂芳基、取代或未取代的环烷基烷基、取代或未取代的杂环烷基烷基、取代或未取代的芳基烷基、取代或未取代的杂芳基烷基、取代或未取代的烯基、取代或未取代的炔基;所述烷基、环烷基、杂环烷基、芳基、杂芳基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基被取代时,可选择性地被如下一个或多个R 10取代在任意位置;或者,R 8或R 11分别独立地选自-NR cR d、-NR cS(O) 1-2R d、-NR cC(O)R d、或-NR cS(O) 2NR cR d
R 1和R 9为独立取代基,或者,R 1和R 9相互连接形成4-8元杂环烷基,所述杂环烷基还可进一步被1~3个C 1-6烷基或C 3-6环烷基任意取代;
每个R 10分别独立地为氢、卤素、烷基、卤代烷基、卤代烷氧基、羟基烷基、氨基烷基、氨基环烷基、烯基、炔基、-CN、-NO 2、-SR a、-OR a、-OC(O)R a、-OC(O)OR a、-OC(O)NR aR b、-C(O)OR a、-C(O)R a、-C(O)NR aR b、-NR aR b、-NR aC(O)R b、-N(R a)C(O)OR b、-N(R a)C(O)NR aR b、-NR aS(O) 2R b、-NR aC(=NH)R b、-NR aC(=NH)NR aR b、-S(O) 1-2R b、-S(O) 2NR aR b和 -NR aS(O) 2NR aR b;或每个R 10分别独立地为环烷基、或杂环烷基;或每个R 10分别独立地为R a
每个R a和每个R b分别独立地选自氢、C 1-6烷基、C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环烷基、C 6-10芳基、5-6元杂芳基、C 3-8环烷基C 1-6烷基、3-8元杂环烷基C 1-6烷基、苯基C 1-6烷基、或5-6元杂芳基C 1-6烷基;所述C 1-6烷基、C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环烷基、C 6-10芳基、或5-6元杂芳基为未取代或者选择性地被1~3个选自卤素、羟基、氨基、羧基、C 1-6烷基、卤代C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基和卤代C 1-6烷氧基中的一种或多种取代基取代在任意位置;
R a和R b为独立取代,或者,所述R a和R b与其共同连接的N原子一起形成3-12元杂环烷基,所述杂环烷基还可以进一步含有1~3个选自N、O、S(O) 0-2、C(O)的杂原子或基团;所述杂环烷基为未取代或者选择性被1~3个选自卤素、羟基、氨基、羧基、C 1-6烷基、卤代C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基和卤代C 1-6烷氧基中的一种或多种取代基取代在任意位置;
每个R c和每个R d分别独立地选自氢、C 1-6烷基、C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环烷基、C 6-10芳基、5-6元杂芳基、C 3-8环烷基C 1-6烷基、3-8元杂环烷基C 1-6烷基、苯基C 1-6烷基、或5-6元杂芳基C 1-6烷基;所述C 1-6烷基、C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环烷基、C 6-10芳基、或5-6元杂芳基为未取代或者选择性地被1~3个选自卤素、羟基、氨基、羧基、C 1-6烷基、卤代C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基和卤代C 1-6烷氧基中的一种或多种取代基取代在任意位置。
本发明提供了一种如式(I’)所示化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐;
Figure PCTCN2018101290-appb-000002
其中,W为N或CR 7
X为CR 8、或N;Y为NR 9、CHR 8、-C(R 8)=C(R 11)-、-N=C(R 11)-、O、或S;
U和V分别选自N或CR 8,并且U和V不同时为N;
R 2为烷基、卤代烷基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基;
R 3为氢、卤素、-CN、-NO 2、-SR c、-OR c、-OC(O)R c、-OC(O)OR c、-OC(O)NR cR d、-C(O)OR c、-C(O)R c、-C(O)NR cR d、-NR cR d、-NR cC(O)R d、-N(R c)C(O)OR d、-N(R c)C(O)NR cR d、-NR cS(O) 2R d、-NR cC(=NH)R d、-NR cC(=NH)NR cR d、-S(O) 1-2R c、-S(O) 2NR cR d、-NR cS(O) 2NR cR d、取代或未取代的烷基、取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的芳基、取代或未取代的杂芳基、取代或未取代的环烷基烷基、取代或未取代的杂环烷基烷基、取代或未取代的芳基烷基、取代或未取代的杂芳基烷基、 取代或未取代的烯基、取代或未取代的炔基;所述烷基、环烷基、杂环烷基、芳基、杂芳基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基被取代时,可选择性地被如下一个或多个基团取代在任意位置:卤素、烷基、卤代烷基、卤代烷氧基、羟基烷基、氨基烷基、氨基环烷基、烯基、炔基、-CN、-NO 2、-SR a、-OR a、-OC(O)R a、-OC(O)OR a、-OC(O)NR aR b、-C(O)OR a、-C(O)R a、-C(O)NR aR b、-NR aR b、-NR aC(O)R b、-N(R a)C(O)OR b、-N(R a)C(O)NR aR b、-NR aS(O) 2R b、-NR aC(=NH)R b、-NR aC(=NH)NR aR b、-S(O) 1-2R b、-S(O) 2NR aR b和-NR aS(O) 2NR aR b
R 4、R 5、R 6和R 7分别独立地为氢、卤素、羟基、氰基、硝基、巯基、氨基、烷基、烷氧基、烷硫基、卤代烷基、卤代烷氧基、C 2-6炔基、C 2-6烯基、芳基、环烷基、杂环烷基、杂芳基、-OC(O)R c、-OC(O)OR c、-OC(O)N(R c) 2、-C(O)OR c、-C(O)R c、-C(O)N(R c) 2、-N(R c) 2、-NHC(O)R c、-NHC(O)OR c、-NHC(O)N(R c) 2、-NHS(O) 2R c、-S(O) 0-2R c或-S(O) 2N(R c) 2
R 3和R 4为独立取代基,或者,R 3和R 4与其所连接的环原子一起形成A环,所述A环为取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的苯基、或取代或未取代的5-6元杂芳基;所述A环被取代时,可选择性地被如下一个或多个基团取代在任意位置:氧代基、硫代基、卤素、-CN、-NO 2、-SR c、-OR c、-OC(O)R c、-OC(O)OR c、-OC(O)NR cR d、-C(O)OR c、-C(O)R c、-C(O)NR cR d、-NR cR d、-NR cC(O)R d、-N(R c)C(O)OR d、-N(R c)C(O)NR cR d、-NR cS(O) 2R d、-NR cC(=NH)R d、-NR cC(=NH)NR cR d、-S(O) 1-2R c、-S(O) 2NR cR d、-NR cS(O) 2NR cR d、取代或未取代的烷基、取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的芳基、取代或未取代的杂芳基、取代或未取代的环烷基烷基、取代或未取代的杂环烷基烷基、取代或未取代的芳基烷基、取代或未取代的杂芳基烷基、取代或未取代的烯基、取代或未取代的炔基;所述烷基、环烷基、杂环烷基、芳基、杂芳基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基被取代时,可选择性地被如下一个或多个基团取代在任意位置:卤素、烷基、卤代烷基、卤代烷氧基、羟基烷基、氨基烷基、烯基、炔基、-CN、-NO 2、-SR c、-OR c、-OC(O)R c、-OC(O)OR c、-OC(O)N(R c) 2、-C(O)OR c、-C(O)R c、-C(O)N(R c) 2、-N(R c) 2、-NHC(O)R c、-NHC(O)OR c、-NHC(O)N(R c) 2、-NHS(O) 2R c、-NHC(=NH)R c、-NHC(=NH)N(R c) 2、-S(O) 1-2R c、-S(O) 2N(R c) 2和-NHS(O) 2N(R c) 2
R 8、R 9或R 11分别独立地为氢、-C(O)OR c、-C(O)R c、-C(O)NR cR d、-S(O) 1-2R c、-S(O) 2NR cR d、取代或未取代的烷基、取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的芳基、取代或未取代的杂芳基、取代或未取代的环烷基烷基、取代或未取代的杂环烷基烷基、取代或未取代的芳基烷基、取代或未取代的杂芳基烷基、取代或未取代的烯基、取代或未取代的炔基;所述烷基、环烷基、杂环烷基、芳基、杂芳基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基被取代时,可选择性地被如下一个或多个R 10取代在任意位置;或者,R 8或R 11分别独立地选自-NR cR d、-NR cS(O) 1-2R d、-NR cC(O)R d、或-NR cS(O) 2NR cR d
每个R 10分别独立地为氢、卤素、烷基、卤代烷基、卤代烷氧基、羟基烷基、氨基烷基、氨基环烷基、烯基、炔基、环烷基、杂环烷基、-CN、-NO 2、-SR a、-OR a、-OC(O)R a、 -OC(O)OR a、-OC(O)NR aR b、-C(O)OR a、-C(O)R a、-C(O)NR aR b、-NR aR b、-NR aC(O)R b、-N(R a)C(O)OR b、-N(R a)C(O)NR aR b、-NR aS(O) 2R b、-NR aC(=NH)R b、-NR aC(=NH)NR aR b、-S(O) 1-2R b、-S(O) 2NR aR b和-NR aS(O) 2NR aR b;或每个R 10分别独立地为R a
每个R a和每个R b分别独立地选自氢、C 1-6烷基、C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环烷基、C 6-10芳基、5-6元杂芳基、C 3-8环烷基C 1-6烷基、3-8元杂环烷基C 1-6烷基、苯基C 1-6烷基、或5-6元杂芳基C 1-6烷基;所述C 1-6烷基、C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环烷基、C 6-10芳基、或5-6元杂芳基为未取代或者选择性地被1~3个选自卤素、羟基、氨基、羧基、C 1-6烷基、卤代C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基和卤代C 1-6烷氧基中的一种或多种取代基取代在任意位置;
R a和R b为独立取代,或者,所述R a和R b与其共同连接的N原子一起形成3-12元杂环烷基,所述杂环烷基还可以进一步含有1~3个选自N、O、S(O) 0-2、C(O)的杂原子或基团;所述杂环烷基为未取代或者选择性被1~3个选自卤素、羟基、氨基、羧基、C 1-6烷基、卤代C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基和卤代C 1-6烷氧基中的一种或多种取代基取代在任意位置;
每个R c和每个R d分别独立地选自氢、C 1-6烷基、C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环烷基、C 6-10芳基、5-6元杂芳基、C 3-8环烷基C 1-6烷基、3-8元杂环烷基C 1-6烷基、苯基C 1-6烷基、或5-6元杂芳基C 1-6烷基;所述C 1-6烷基、C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环烷基、C 6-10芳基、或5-6元杂芳基为未取代或者选择性地被1~3个选自卤素、羟基、氨基、羧基、C 1-6烷基、卤代C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基和卤代C 1-6烷氧基中的一种或多种取代基取代在任意位置。
本发明中,所述W优选为N。
本发明中,所述U优选为CH。
本发明中,所述V优选为CR 8;其中,R 8的定义如前所述。
本发明中,所述X和Y优选为如下任一组合:
1)X为N、Y为NR 9;其中,R 9的定义如前所述。
2)X为CH、Y为NR 9;其中,R 9的定义如前所述。
3)X为CR 8、Y为O;其中,R 8的定义如前所述。
4)X为CR 8、Y为O;其中,R 8的定义如前所述。
5)X为CR 8、Y为-C(R 8)=C(R 11)-。其中,R 8和R 11的定义如前所述。
本发明中,所述X和Y更优选为X为CH、Y为NR 9;其中,R 9的定义如前所述。
本发明中,基团
Figure PCTCN2018101290-appb-000003
优选通过(1)位和母核连接;
本发明中,基团
Figure PCTCN2018101290-appb-000004
优选通过(2)位和母核连接。
本发明中,基团
Figure PCTCN2018101290-appb-000005
优选为以下结构:
Figure PCTCN2018101290-appb-000006
本发明中,基团
Figure PCTCN2018101290-appb-000007
优选为以下结构:
Figure PCTCN2018101290-appb-000008
Figure PCTCN2018101290-appb-000009
本发明中,基团
Figure PCTCN2018101290-appb-000010
优选通过(1)位和母核连接;
本发明中,基团
Figure PCTCN2018101290-appb-000011
优选通过(2)位和母核连接。
本发明中,基团
Figure PCTCN2018101290-appb-000012
优选为以下结构:
Figure PCTCN2018101290-appb-000013
Figure PCTCN2018101290-appb-000014
本发明中,所述R 1优选为H或取代的C 1-6烷基;所述R 1更优选为H。
本发明中,所述R 2优选为:
Figure PCTCN2018101290-appb-000015
本发明中,所述R 2更优选为:
Figure PCTCN2018101290-appb-000016
本发明中,所述R 3中,所述烷基、环烷基、杂环烷基、环烷基烷基或杂环烷基被取代时,优选被1~3个取代基取代在任意位置。所述取代基定义如上。
本发明中,所述R 3优选为取代或未取代的C 1-6烷基,当所述烷基被取代时,优选被1个羟基取代在任意位置。
本发明中,所述R 4优选为H。
本发明中,所述R 3和R 4与其所连接的环原子一起形成A环,所述A环优选为取代或未取代的单环环烷基、取代或未取代的单环杂环烷基、取代或未取代的苯基、或取代或未取代的5-6元杂芳基;
本发明中,所述A环优选未取代。
本发明中,所述A环被取代时,优选被1~3个取代基取代在任意位置。所述取代基定义如上。
本发明中,所述R 5优选为H。
本发明中,所述R 6优选为H。
本发明中,所述R 8优选为H;
本发明中,所述R 8优选为-NR cR d、-NR cS(O) 1-2R d、-NR cC(O)R d、-NR cS(O) 2NR cR d、-C(O)OR c、-C(O)R c、-C(O)NR cR d、-S(O) 1-2R c、-S(O) 2NR cR d、取代或未取代的烷基、取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的芳基、取代或未取代的杂芳基、取代或未取代的环烷基烷基、取代或未取代的杂环烷基烷基、取代或未取代的芳基烷基、取代或未取代的杂芳基烷基、取代或未取代的烯基、取代或未取代的炔基;所述烷基、环烷基、杂环烷基、芳基、杂芳基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基被取代时,可选择性地被如下一个或多个R 10取代在任意位置;优选被1~3个R 10取代在任意位置。
本发明中,所述R 8中,所述取代或未取代的烷基优选为取代或未取代的C 1-6烷基;更优选为取代或未取代的C 1-4烷基;
所述R 8中,所述取代或未取代的环烷基优选为取代或未取代的C 3-8环烷基;
所述R 8中,所述取代或未取代的杂环烷基优选为取代或未取代的3-8元杂环烷基;
所述R 8中,所述取代或未取代的杂环烷基优选为取代或未取代的如下任一基团:氮杂环丁基、吡咯烷基、哌啶基、哌嗪基、高哌嗪基、吗啉基、(1S,4S)-2,5-二氮杂双环[2.2.1]庚基、(1R,4R)-2,5-二氮杂双环[2.2.1]庚基、(1R,4R)-2-氧-5-氮杂双环[2.2.1]庚基、(1S,4S)-2-氧-5-氮杂双环[2.2.1]庚基、或4,7-二氮杂螺[2.5]辛基;
所述R 8中,所述取代或未取代的环烷基烷基优选为取代或未取代的C 3-8环烷基C 1-3烷基;
所述R 8中,所述取代或未取代的杂环烷基烷基优选为取代或未取代的3-8元杂环烷基C 1-3烷基;
所述R 8中,所述取代或未取代的芳基优选为取代或未取代的苯基。
所述R 8中,所述取代或未取代的杂芳基优选为取代或未取代的5-6元杂芳基。
本发明中,所述R 9中,所述取代或未取代的烷基优选为取代或未取代的C 1-6烷基;更优选为取代或未取代的C 1-4烷基;
所述R 9中,所述取代或未取代的环烷基优选为取代或未取代的C 3-8环烷基;
所述R 9中,所述取代或未取代的杂环烷基优选为取代或未取代的3-8元杂环烷基;
所述R 9中,所述取代或未取代的杂环烷基优选为取代或未取代的如下任一基团:吡咯烷基、哌啶基、氮杂环庚基、氮杂环丁基、氧杂环丁基、4-氮杂螺[2.5]辛基、或5-氮杂螺[2.5]辛基;
所述R 9中,所述取代或未取代的环烷基烷基优选为取代或未取代的C 3-8环烷基C 1-3烷基;
所述R 9中,所述取代或未取代的杂环烷基烷基优选为取代或未取代的3-8元杂环烷基C 1-3烷基;
所述R 9中,所述取代或未取代的芳基优选为取代或未取代的苯基。
所述R 9中,所述取代或未取代的杂芳基优选为取代或未取代的5-6元杂芳基。
本发明中,每个R 10独立地优选为H、F、Cl、C 1-6烷基、卤代C 1-6烷基、卤代C 1-6烷氧基、羟基C 1-6烷基、氨基C 1-6烷基、氨基C 3-8环烷基、C 2-6烯基、C 2-6炔基、-CN、-NO 2、-SR a、-OR a、-OC(O)R a、-OC(O)OR a、-OC(O)NR aR b、-C(O)OR a、-C(O)R a、-C(O)NR aR b、-NR aR b、-NR aC(O)R b、-N(R a)C(O)OR b、-N(R a)C(O)NR aR b、-NR aS(O) 2R b、-NR aC(=NH)R b、-NR aC(=NH)NR aR b、-S(O) 1-2R b、-S(O) 2NR aR b、或-NR aS(O) 2NR aR b;或者,每个R 10独立地优选为C 3-8环烷基、或3-8元杂环烷基;R a和R b分别独立地选自H、或C 1-6烷基。R a和R b为独立取代,或者,所述R a和R b与其共同连接的N原子一起形成3-8元杂环烷基,所述杂环烷基还可以进一步含有1~3个选自N、O、S(O) 0-2、C(O)的杂原子或基团;所述杂环烷基为未取代或者选择性被1个选自C 1-6烷基的取代基取代在任意位置;
本发明中,每个R 10独立地更优选为H、D、F、Cl、氨基、羟基、甲基、乙基、丙基、丁基、异丙基、异丁基、叔丁基、环丙烷、环丁烷、环戊烷、氮杂环丁基、氧杂环丁基、三氟甲基、1,1-二氟乙基、1,1,1-三氟乙基、甲氧基、乙氧基、三氟甲氧基、1,1-二氟乙氧基、氨基乙基、羟基乙基、-C(O)CH 3、-C(O)OCH 3、-NHCH 3、或-N(CH 3) 2
在一些实施方案中,如式(I)所示化合物和/或药学上可接受的盐为如式(II)所示化合物和/或其药学上可接受的盐:
Figure PCTCN2018101290-appb-000017
其中,R 1、R 2、R 3、R 4、R 5、R 9、V、X和W的定义如前所述。
在一些实施方案中,如式(I)所示化合物和/或药学上可接受的盐为如式(III)所示化合物和/或其药学上可接受的盐:
Figure PCTCN2018101290-appb-000018
其中,n为0、1、2、或3;
R 1、R 3、R 4、R 5、X、R a、R b和W的定义如前所述。
以下各优选实施方案均包括在式(III)的定义中:
在一些优选实施方案中,n为1。
在一些实施方案中,如式(I)所示化合物和/或药学上可接受的盐为如式(IV)所示化合物和/或其药学上可接受的盐:
Figure PCTCN2018101290-appb-000019
其中,Z为N或CH;L为CH 2、CH(CH 3)、C(CH 3) 2、或
Figure PCTCN2018101290-appb-000020
u为0、1、或2;v为0、1、或2;
R 1、R 3、R 4、R 5、R 10、X和W的定义如前所述。
以下各优选实施方案均包括在式(IV)的定义中:
在一些优选实施方案中,L为
Figure PCTCN2018101290-appb-000021
在一些优选实施方案中,u为1,v为1;
在一些优选实施方案中,u为1,v为2;
在一些优选实施方案中,u为0,v为1;
在一些优选实施方案中,u为0,v为2;
在一些优选实施方案中,u为1,v为0;
在一些优选实施方案中,u为2,v为0;
在一些优选实施方案中,Z为N;和R 10为氢、C 1-6烷基、卤代C 1-6烷基、羟基C 1-6烷基、氨基C 1-6烷基、C 2-6烯基、C 2-6炔基、-OR a、-C(O)OR a、-C(O)R a、-C(O)NR aR b、-S(O) 1-2R b、或-S(O) 2NR aR b;R a和R b分别独立地为氢、或C 1-6烷基。
在一些实施方案中,如式(I)所示化合物和/或药学上可接受的盐为如式(V)所示 化合物和/或其药学上可接受的盐:
Figure PCTCN2018101290-appb-000022
其中,R 1、R 2、R 3、R 4、R 5、R 8、R 9、U和W的定义如前所述。
以下各优选实施方案均包括在式(V)的定义中:
在一些优选实施方案中,U为CH;
在一些优选实施方案中,R 1为H或甲基;
在一些优选实施方案中,R 8为-NR cR d
在一些优选实施方案中,R 8为取代或未取代的3-8元杂环烷基;当所述3-8元杂环烷基被取代时,选择性被1~3个C 1-4烷基、或C 3-6环烷基取代在任意位置;
在一些优选实施方案中,R 9为C 1-4烷基、羟基C 1-4烷基、C 3-8环烷基、或3-8元杂环烷基;
在一些优选实施方案中,R 2
Figure PCTCN2018101290-appb-000023
在一些实施方案中,如式(I’)所示化合物和/或药学上可接受的盐为如式(VI)所示化合物和/或其药学上可接受的盐:
Figure PCTCN2018101290-appb-000024
其中,R 1、R 2、R 3、R 4、R 5、R 8、R 9、U和W的定义如前所述。
以下各优选实施方案均包括在式(VI)的定义中:
在一些优选实施方案中,U为CH;
在一些优选实施方案中,R 8为-NR cR d;;
在一些优选实施方案中,R 8为取代或未取代的3-8元杂环烷基;当所述3-8元杂环烷基被取代时,选择性被1~3个C 1-4烷基、或C 3-6环烷基取代在任意位置;
在一些优选实施方案中,R 9为C 1-4烷基、羟基C 1-4烷基、C 3-8环烷基、或3-8元杂环烷基。
在一些实施方案中,如式(I)所示的化合物和/或药学上可接受的盐任选为以下化合物:
Figure PCTCN2018101290-appb-000025
Figure PCTCN2018101290-appb-000026
Figure PCTCN2018101290-appb-000027
Figure PCTCN2018101290-appb-000028
Figure PCTCN2018101290-appb-000029
Figure PCTCN2018101290-appb-000030
在一些实施方案中,如式(I)所示的化合物和/或药学上可接受的盐任选为以下化合物:
Figure PCTCN2018101290-appb-000031
Figure PCTCN2018101290-appb-000032
在一些实施方案中,如式(I)所示的化合物和/或药学上可接受的盐任选为以下化合物:
Figure PCTCN2018101290-appb-000033
本发明还提供了所述如式(I)或(I’)所示化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐的制备方法,其为如下任一方法:
方法1:
Figure PCTCN2018101290-appb-000034
方法1中,所述R 1、R 2、R 3、R 4、R 5、R 6、U、V、Y和W的定义均同前所述;方法1包括如下步骤:步骤1)将1a与1b通过buchwald偶联反应得到化合物1c;步骤2)将化合物1c中的甲硫基用间氯过氧苯甲酸(m-CPBA)氧化为亚砜得到化合物1d;步骤3)将化合物1d和1e或1e’在碱性条件下反应得到通式I或I’所示化合物。
方法1中,所述的条件和步骤可为本领域常规的反应的条件和步骤,本发明特别优选以下反应条件:步骤1)氮气保护下,在1,4-二氧六环溶剂中,在碱(1,2-N,N-二甲基乙二胺、碳酸钾)和氯化亚铜的作用下反应得到,所述剂的用量优选1~50mL/mmol化 合物1a,所述反应时间优选0-24小时,温度优选室温到溶剂回流,更优选为80~100℃,化合物1a和1b的摩尔比优选1:0.9~1:1.5。步骤2)二氯甲烷溶剂中,用间氯过氧苯甲酸氧化化合物1c得到化合物1d;所述剂的用量优选1~50mL/mmol化合物1c,所述反应时间优选0-24小时,温度优选0℃~室温,化合物1c和m-CPBA的摩尔比优选1:1~1:3;步骤3)甲苯中,碱性条件下(N,N-二异丙基乙胺或三乙胺),1d和1e或Ie’反应得到通式I或I’所示化合物,所述剂的用量优选1~50mL/mmol化合物1d,所述反应时间优选0-24小时,温度优选室温到溶剂回流,化合物1d、1e和碱的摩尔比优选1:0.9:1~1:2.5:2.5;化合物1d、1e’和碱的摩尔比优选1:0.9:1~1:2.5:2.5。
在上述方法最后的合成步骤中使用对甲苯磺酸、盐酸、氯化氢、或三氟乙酸等酸性体系,或在纯化过程中,例如:prep-HPLC的流动相中存在上述酸性体系时,则所述的如式I或I’所示的化合物将会是相应的对甲苯磺酸盐、盐酸盐或三氟乙酸盐等。上述方法最后的合成步骤的纯化过程中,例如:使用硅胶柱层析或prep-TLC的洗脱条件为石油醚、乙酸乙酯、二氯甲烷、甲醇、正己烷中的一种或多种、或prep-HPLC的流动相使用碳酸氢铵等碱性体系时,则所述的如式I或I’所示的化合物将会是游离碱形式。
在上述方法中,在1a、1b、1e或1e’中存在不参与反应的氨基基团、羟基基团或羧基基团时,该氨基基团、羟基基团或羧基基团优选通过保护基保护,避免有任何副反应发生。如果存在上述氨基保护基团或羟基保护基团则需要经过后续的脱保护步骤后,得到如式I和I’所示化合物。任何合适的氨基保护基团,例如:叔丁氧羰基(Boc)基团,均可以用于保护氨基基团。如果使用Boc作为保护基,后续的脱保护反应可以在标准条件,例如,对甲苯磺酸/甲醇体系,二氯甲烷/三氟乙酸体系、饱和的氯化氢***溶液、或三氟甲磺酸三甲基硅酯/2,6-二甲基吡啶/二氯甲烷体系中进行;任何合适的羟基保护基团,例如:苄基,均可以用于保护氨基基团,后续的脱保护反应可以在标准条件,例如,钯碳/氢气;任何合适的羧基保护基团,例如:形成羧酸酯基团(例如,羧酸甲酯,羧酸乙酯),均可以用于保护羧基基团,后续的脱保护反应可以在标准条件,例如,氢氧化钠、氢氧化钾、氢氧化锂在四氢呋喃、水和/或甲醇溶剂中脱保护。上述脱保护反应优选在最后一步进行。
所述如式(I)或(I’)所示化合物,其药学上可接受的盐可通过一般的化学方法合成。
一般情况下,盐的制备可以通过游离碱或酸与等化学当量或者过量酸(无机酸或有机酸)或碱(无机碱或有机碱)在合适的溶剂或溶剂组合物中反应制得。
本发明还提供了一种药物组合物,其包括治疗有效量的活性组分以及药学上可接受的辅料;所述活性组分包括如式(I)或(I’)所示化合物、其异构体、前药、稳定的同位素衍生物和药学上可接受的盐中的一种或多种。
所述药物组合物中,所述活性组分还可包括癌症、病毒感染或自身免疫疾病的其它治疗剂。
所述药物组合物中,所述药学上可接受的辅料可包括药学上可接受的载体、稀释剂和/或赋形剂。
根据治疗目的,可将药物组合物制成各种类型的给药单位剂型,如片剂、丸剂、粉剂、液体、悬浮液、乳液、颗粒剂、胶囊、栓剂和针剂(溶液及悬浮液)等,优选液体、悬浮液、乳液、栓剂和针剂(溶液及悬浮液)等。
为了使片剂形式的药物组合物成形,可使用本领域任何已知并广泛使用的赋形剂。例如,载体,如乳糖、白糖、氯化钠、葡萄糖、尿素、淀粉、碳酸钙、高岭土、结晶纤维素和硅酸等;粘合剂,如水、乙醇、丙醇、普通糖浆、葡萄糖溶液、淀粉溶液、明胶溶液,羧甲基纤维素、紫胶、甲基纤维素和磷酸钾、聚乙烯吡咯烷酮等;崩解剂,如干淀粉、藻酸钠、琼脂粉和海带粉,碳酸氢钠、碳酸钙、聚乙烯脱水山梨醇的脂肪酸酯、十二烷基硫酸钠、硬脂酸单甘酯、淀粉和乳糖等;崩解抑制剂,如白糖、甘油三硬脂酸酯、椰子油和氢化油;吸附促进剂,如季胺碱和十二烷基硫酸钠等;润湿剂,如甘油、淀粉等;吸附剂,如淀粉、乳糖、高岭土、膨润土和胶体硅酸等;以及润滑剂,如纯净的滑石,硬脂酸盐、硼酸粉和聚乙二醇等。还可以根据需要选用通常的涂渍材料制成糖衣片剂、涂明胶膜片剂、肠衣片剂、涂膜片剂、双层膜片剂及多层片剂。
为了使丸剂形式的药物组合物成形,可使用本领域任何已知的并广泛使用的赋形剂,例如,载体,如乳糖,淀粉,椰子油,硬化植物油,高岭土和滑石粉等;粘合剂,如***树胶粉,黄蓍胶粉,明胶和乙醇等;崩解剂,如琼脂和海带粉等。
为了使栓剂形式的药物组合物成形,可使用本领域任何已知并广泛使用的赋性剂,例如,聚乙二醇,椰子油,高级醇,高级醇的酯,明胶和半合成的甘油酯等。
为了制备针剂形式的药物组合物,可将溶液或悬浮液消毒后(最好加入适量的氯化钠,葡萄糖或甘油等),制成与血液等渗压的针剂。在制备针剂时,也可使用本领域内任何常用的载体。例如,水,乙醇,丙二醇,乙氧基化的异硬脂醇,聚氧基化的异硬脂醇和聚乙烯脱水山梨醇的脂肪酸酯等。此外,还可加入通常的溶解剂、缓冲剂和止痛剂等。
本发明中,所述的组合物在药物组合物中的含量无特殊限制,可在很宽的范围内进行选择,通常可为质量百分比的5~95%,较佳的为质量百分比30~80%。
本发明中,所述药物组合物的给药方法没有特殊限制。可根据病人年龄、性别和其它条件及症状,选择各种剂型的制剂给药。例如,片剂、丸剂、溶液、悬浮液、乳液、颗粒剂或胶囊口服给药;针剂可以单独给药,或者和注射用输送液(如葡萄糖溶液及氨基酸溶液)混合进行静脉注射;栓剂为给药到直肠。
本发明还提供了如式(I)或(I’)所示化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,或所述药物组合物在制备Wee1抑制剂中的应用。
本发明还提供了如式(I)或(I’)所示化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,或所述药物组合物在制备癌症的化学疗法或放射性疗法的增敏剂中的应用。其中,所述化学疗法或放射线疗法的增敏剂是指在癌症治疗领域中个,通过与放射线疗法和/或使用抗癌剂的化学疗法组合使用,相加地或协同地提高这些放射线疗法和/或化学疗法的治疗效果的药物。
本发明还提供了如式(I)或(I’)所示化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,或所述药物组合物在制备治疗和/或缓解由Wee1介导的相关 疾病的药物中的应用;本发明优选提供了如式(I)所示化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,或所述药物组合物在制备治疗和/或预防由Wee1介导的相关疾病的药物中的应用;所述疾病包括肿瘤和非肿瘤性疾病。所述疾病优选为癌症。
本发明优选用所述如式(I)或(I’)所示化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,或所述药物组合物在制备治疗和/或缓解癌症药物中的应用。
本发明还进一步提供了用所述如式(I)或(I’)所示化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,或所述药物组合物治疗癌症的方法,包括:给予哺乳动物治疗所需剂量的如式(I)或(I’)所述化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,或药物组合物。
所述哺乳动物,优选人。
本发明还进一步提供了所述如式(I)或(I’)所示化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,或所述药物组合物还可和一种或多种其它种类的治疗剂和/或治疗方法联合用于治疗和/或缓解由Wee1介导的相关疾病,所述的疾病优选为癌症。
本发明还进一步提供一种联合制剂,包括如式(I)或(I’)所示化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,或所述药物组合物和其它种类的用于治疗癌症的治疗剂和/或治疗方法合用。
本发明中,所述其它种类的治疗剂(例如:用于治疗癌症的其它种类的治疗剂)可以和所述的如式(I)或(I’)所示化合物做成单一给药的治疗剂型,或者分别先后给药的治疗剂型。
本发明中,所述其它种类的用于治疗癌症的治疗剂和/或治疗方法可包括但不限于用于癌症治疗的:微管蛋白抑制剂、烷化剂、拓扑酶I/II抑制剂、铂类化合物、抗生素、抗代谢类药物、激素和激素类似物、靶向治疗(例如:特殊的激酶抑制剂)、免疫治疗剂、干扰素、其它用于癌症治疗的抗癌剂和放疗中的一种或多种。
本发明中,所述微管蛋白抑制剂可选自但不限于:长春碱系列(例如:长春碱、长春新碱、长春瑞滨、长春地辛)、紫杉烷类(多西他赛、紫杉醇)和甲磺酸艾日布林中的一种或多种。
本发明中,所述烷化剂可选自但不限于:氮芥、乙烯亚胺衍生物、甲烷磺酸酯类、亚硝脲类和三氮烯类中的一种或多种。
本发明中,所述拓扑酶I/II抑制剂可选自但不限于:喜树碱、10-羟基喜树碱、伊立替康、伊立替康代谢产物SN-38、拓扑替康、喜诺替康、Exactecan、Karenitecin、9-硝基喜树碱、阿霉素和右雷佐生中的一种或多种。
本发明中,所述铂类化合物可选自但不限于:顺铂、卡铂、奈达铂、奥沙利铂、奥马铂、四铂、异丙铂、螺铂、顺-二氨二水铂(II)-离子、或二氯(乙二胺)-铂(II)。
本发明中,所述抗代谢类药物可选自但不限于:叶酸拮抗剂、嘧啶类似物、嘌呤类似物、腺苷脱氨酶抑制剂,例如:甲氨蝶呤、5-氟尿嘧啶、氟脲苷、阿糖胞苷、6-巯基嘌 呤、6-硫鸟嘌呤、磷酸氟达拉滨、喷司他丁和吉西他滨中的一种或多种。
本发明中,所述免疫治疗剂可选自但不限于:抗肿瘤疫苗(例如:合成肽、DNA疫苗和重组病毒)、溶瘤病毒、单克隆抗体(例如:PD-1单抗、PD-L1单抗、CTLA-4单抗等)、新型佐剂、细胞因子治疗(例如:IL2和GM-CSF)、嵌合抗原受体T细胞治疗法(CAR-T)、小分子免疫调节剂、肿瘤微环境调节剂和抗血管生成因子中的一种或多种。
本发明中,所述用于癌症治疗的抗生素可选自但不限于:放线菌素D、多柔比星、柔红霉素、博来霉素、培洛霉素、丝裂霉素C、阿柔比星、吡柔比星、表柔比星、净司他丁斯酯、伊达比星、西罗莫司和戊柔比星中的一种或多种。
本发明中,所述用于癌症治疗的干扰素可选自但不限于:干扰素α、干扰素α-2a、干扰素α-2b、干扰素β、干扰素γ-1a或干扰素γ-n1等。
本发明中,所述的癌症包括转移性的和非转移性的癌症,也包括家族遗传性的和偶发性的癌症,还可包括固体肿瘤和非固体肿瘤。
本发明中,所述固体肿瘤的具体例子可包括但不限于:眼、骨、肺、胃、胰腺、乳腺、***、脑(包括胶质母细胞瘤和髓母细胞瘤)、卵巢(包括那些从上皮细胞产生的基质细胞,生殖细胞和***)、膀胱、睾丸、脊髓、肾脏(包括腺癌、肾母细胞瘤)、口、唇、咽喉、口腔(包括鳞状细胞癌)、鼻腔、小肠、结肠、直肠、甲状旁腺、胆囊、胆管、宫颈、心、咽下腺、支气管、肝、输尿管、***、***、喉腺、甲状腺(包括甲状腺癌和髓样癌),食道、鼻咽腺垂体、唾液腺、肾上腺、头颈部上皮内瘤样病变(包括Bowen病和Paget氏病),肉瘤(包括平滑肌肉瘤、横纹肌肉瘤、脂肪肉瘤、纤维肉瘤、骨肉瘤)、皮肤(包括黑色素瘤、卡波氏肉瘤、basocellular癌和鳞状细胞癌)等相关的肿瘤。
本发明中,所述固体肿瘤优选为人的眼癌、骨癌、肺癌、胃癌、胰腺癌、乳腺癌、***癌、脑癌(包括但不限于噁性胶质瘤、成神经管细胞瘤)、卵巢癌、膀胱癌、子***、睾丸癌、肾癌(包括但不限于腺癌、肾母细胞癌)、口腔癌(包括鳞状细胞癌)、舌癌、喉癌、鼻咽癌、头颈癌、结肠癌、小肠癌、直肠癌、甲状旁腺癌、甲状腺癌、食管癌、胆囊癌、胆管癌、***、肝癌、肺癌(包括但不限于小细胞肺癌、非小细胞肺癌)、绒毛上皮癌、骨肉瘤、尤文瘤、软组织肉瘤和皮肤癌中的一种或多种。
本发明中,所述非固体肿瘤(包括血液学肿瘤)的具体例子可包括但不限于:淋巴性白血病(包括急性淋巴细胞白血病、淋巴瘤、骨髓瘤、慢性淋巴细胞白血病、霍奇金淋巴瘤、非霍奇金淋巴瘤、T细胞慢性淋巴性白血病、B细胞慢性淋巴性白血病)、髓性相关的白血病(包括急性髓性白血病、慢性髓性白血病)和AIDs相关的白血病中的一种或多种。
本发明中,除非另有说明,术语“选择性地被一个或多个基团取代在任意位置”是指基团上所指定的一个或多个原子的任何一个或者多个氢原子用所指定的基团取代,条件是不超过指定原子的正常化合价,所述取代在任意位置均为本领域常见的合理取代。
本发明中,当与取代基的键合显示与连接环中两个原子的键合相交时,那么这样的取代基可键合在环上的任何可键合的环原子。
除非另有说明,在本发明说明书和权利要求书中出现的以下术语具有下述含义:
术语“烷基”是指包含1-20个碳原子的饱和直链或支链烃基,优选1-10个碳原子,更优选1-8,1-6,1-4,或1-3个碳原子,烷基的代表性例子包括但不限于:甲基、乙基、正丙基、异丙基、正丁基、仲丁基、叔丁基、异丁基、戊基、己基、庚基、辛基、壬基、癸基、4,4-二甲基戊基、2,2,4-三甲基戊基、十一烷基、十二烷基,及它们的各种异构体等。
术语“环烷基”是指包含3-20个碳原子的饱和或部分不饱和(包含1或2个双键)的单环或多环基团。“单环环烷基”优选3-10元单环烷基,更优选3-8元单环烷基,例如:环丙基、环丁基、环戊基、环己基、环庚基、环辛基、环癸基、环十二烷基、环己烯基。“多环环烷基”包括“桥环基”、“稠合环烷基”和“螺环烷基”,“桥环基”的代表性例子包括但不限于:冰片基、双环[2.2.1]庚烯基、双环[3.1.1]庚烷基、双环[2.2.1]庚烷基、双环[2.2.2]辛烷基、双环[3.2.2]壬烷基、双环[3.3.1]壬烷基、双环[4.2.1]壬烷基和金刚烷基等。“稠合环烷基”包含稠合到苯基、环烷基、或杂芳基上的环烷基环,稠合环烷基包括但不限于:苯并环丁烯、2,3-二氢-1-H-茚、2,3-环戊烯并吡啶、5,6-二氢-4H-环戊基[B]噻吩、十氢萘等。“螺环烷基”是指两个单环环烷基共用一个碳原子形成的双环基团,螺环烷基包括但不限于:螺[2,5]辛基、螺[2,4]庚基、螺[4,5]癸基等。所述多环环烷基优选包含7~12个碳原子。单环环烷基或多环环烷基可以通过环上任意的1个或2个碳原子链接到母体分子上。
术语“杂环烷基”指由碳原子以及选自氮、氧或硫等杂原子组成的饱和或部分不饱和(包含1或2个双键)的非芳香环状基团,此环状基团可为单环或多环基团,在本发明中,杂环烷基中杂原子个数优选1、2、3或4,杂环烷基中的氮、碳或硫原子可任选地被氧化。氮原子可任选进一步被其他基团取代而形成叔胺或季铵盐。“单环杂环烷基”优选3-10元单环杂环烷基,更优选3-8元单环杂环烷基。例如:氮丙啶基
Figure PCTCN2018101290-appb-000035
氮杂环丁基
Figure PCTCN2018101290-appb-000036
氮杂环庚基
Figure PCTCN2018101290-appb-000037
氧杂环丁基
Figure PCTCN2018101290-appb-000038
吡咯烷基
Figure PCTCN2018101290-appb-000039
四氢呋喃基
Figure PCTCN2018101290-appb-000040
吗啉基
Figure PCTCN2018101290-appb-000041
硫代吗啉基
Figure PCTCN2018101290-appb-000042
硫代吗啉基-S-氧化物
Figure PCTCN2018101290-appb-000043
哌啶基
Figure PCTCN2018101290-appb-000044
哌嗪基
Figure PCTCN2018101290-appb-000045
高哌嗪基
Figure PCTCN2018101290-appb-000046
等。“多环杂环烷基”包括“稠合杂环烷基”、“螺杂环基”和“桥杂环烷基”。“稠合杂环烷基”包含稠合到苯基、环烷基、杂环烷基或杂芳基的单环杂环烷基环,稠合杂环烷基包括但不限于: 2,3-二氢苯并呋喃基、1,3-二氢异苯并呋喃基、二氢吲哚基、2,3-二氢苯并[b]噻吩基、二氢苯并哌喃基、1,2,3,4-四氢喹啉基等。“螺杂环基”是指两个杂环烷基或一个环烷基和一个杂环烷基共用一个碳原子形成的双环基团,螺杂环基包括但不限于:5-氮杂[2.5]辛基
Figure PCTCN2018101290-appb-000047
4-氮杂[2.5]辛基
Figure PCTCN2018101290-appb-000048
4-氮杂[2.4]庚基
Figure PCTCN2018101290-appb-000049
4,7-二氮杂螺[2.5]辛基
Figure PCTCN2018101290-appb-000050
等。“桥杂环烷基”是指单环杂环烷基任意两个不相链接的环原子被1到3个额外的碳原子或杂原子形成的直链基团桥连接(所述的直链基团选自但不限于:-CH 2-、-CH 2CH 2-、-CH 2O-、-CH 2NH-、-CH 2CH 2CH 2-),桥杂环烷基包括但不限于:(1R,4R)-2,5-二氮杂双环[2.2.1]庚基
Figure PCTCN2018101290-appb-000051
(1S,4S)-2,5-二氮杂双环[2.2.1]庚基
Figure PCTCN2018101290-appb-000052
(1S,4S)-2-氧-5-氮杂双环[2.2.1]庚基
Figure PCTCN2018101290-appb-000053
(1R,4R)-2-氧-5-氮杂双环[2.2.1]庚基
Figure PCTCN2018101290-appb-000054
(1S,4R)-2-氮杂双环[2.2.1]庚基
Figure PCTCN2018101290-appb-000055
(1R,4S)-2-氮杂双环[2.2.1]庚基
Figure PCTCN2018101290-appb-000056
等。单环杂环烷基和多环杂环烷基可以通过环上任意的1个或2个环原子链接到母体分子上。上述环原子特指组成环骨架的碳原子和/或氮原子。
术语“环烷基烷基”是指环烷基与母核结构之间通过烷基连接。由此,“环烷基烷基”包含上述烷基和环烷基的定义。
术语“杂环烷基烷基”是指杂环烷基与母核结构之间通过烷基连接。由此,“杂环烷基烷基”包含上述烷基和杂环烷基的定义。
术语“烷氧基”指通过氧桥连接的具有所述碳原子数目的环状或者非环状烷基,包含烷基氧基、环烷基氧基和杂环烷基氧基。由此,“烷氧基”包含上述烷基、杂环烷基和环烷基的定义。
术语“羟基烷基”是指烷基上任意一个或多个氢原子被羟基所取代,包括但不限于:-CH 2OH、-CH 2CH 2OH、-CH 2CH 2C(CH 3) 2OH、-CH(CH 3) 2OH。
术语“烷硫基”指通过硫桥连接的具有所述碳原子数目的环状或者非环状烷基,所述烷硫基包含烷基硫基、环烷基硫基和杂环烷基硫基。由此,“烷硫基”包含上述烷基、 杂环烷基和环烷基的定义。
术语“烯基”指含有至少1个碳碳双键的直链、支链或者环状非芳香烃基。其中可以存在1-3个碳碳双键,优选存在1个碳碳双键。术语“C 2-4烯基”是指具有2-4个碳原子的烯基,术语“C 2-6烯基”是指具有2-6个碳原子的烯基,包括乙烯基、丙烯基、丁烯基、2-甲基丁烯基和环己烯基。所述的烯基可以被取代。
术语“炔基”是指含有至少1个碳碳三键的直链、支链或者环状烃基。其中可以存在1-3个碳碳三键,优选存在1个碳碳三键。术语“C 2-6炔基”是指具有2-6个碳原子的炔基,包括乙炔基、丙炔基、丁炔基和3-甲基丁炔基。
术语“芳基”是指任何稳定的6-20元单环或多环芳香族基团,例如:苯基、萘基、四氢萘基、2,3-二氢化茚基、或联苯基等。
术语“杂芳基”是指至少1个环上的碳原子被选自氮、氧或硫的杂原子置换所形成的芳香环基团,其可为5-7元单环结构或7-20稠合环结构,优选5-6元杂芳基。在本发明中,杂原子个数优选1、2或3,包括但不限于:吡啶基、嘧啶基、哌嗪基、哒嗪-3(2H)-酮基、呋喃基、噻吩基、噻唑基、吡咯基、咪唑基、吡唑基、噁唑基、异噁唑基、1,2,5-噁二唑基、1,2,4-噁二唑基、1,3,4-噁二唑基、1,3,4-噻二唑、1,2,4-三氮唑基、1,2,3-三氮唑基、四氮唑基、吲唑基、异吲唑基、吲哚基、异吲哚基、苯并呋喃基、苯并噻吩基、苯并[d][1,3]二氧戊环基、苯并噻唑基、苯并噁唑基、苯并咪唑基、1H-苯并[d][1,2,3]三氮唑基、喹啉基、异喹啉基、异喹啉酮基、喹唑啉基、1H-吡咯并[2,3-b]吡啶基、4-羟基噻吩并[3,2-c]吡啶基、4,5-二氢-4-氧代呋喃[3,2]吡啶基、4-羟基-5-氮杂吲哚基、呋喃[2,3-c]并吡啶-7(6H)-酮基、噻吩[2,3-c]并吡啶-7(6H)-酮基等。
术语“芳基烷基”是指芳基与母核结构之间通过烷基连接。由此,“芳基烷基”包含上述烷基和芳基的定义。
术语“杂芳基烷基”是指杂环烷基与母核结构之间通过烷基连接。由此,“杂芳基烷基”包含上述烷基和杂芳基的定义。
术语“卤素”表示氟、氯、溴或碘。
术语“卤代烷基”是指被卤素任意取代的烷基。由此,“卤代烷基”包含以上卤素和烷基的定义。
术语“卤代烷氧基”是指被卤素任意取代的烷氧基。由此,“卤代烷氧基”包含以上卤素和烷氧基的定义。
术语“氨基”是指-NH 2,术语“烷氨基”是指氨基上至少一个氢原子被烷基所取代,包括但不限于:-NHCH 3、-N(CH 3) 2、-NHCH 2CH 3、-N(CH 2CH 3) 2。术语“氨基烷基”是指烷基上任意一个氢原子被氨基所取代,包括但不限于:-CH 2NH 2、-CH 2CH 2NH 2。由此,“氨基烷基”和“烷氨基”包含上述烷基和氨基的定义。术语“氨基环烷基”是指环烷基上任意一个氢原子被氨基所取代,包括但不限于:
Figure PCTCN2018101290-appb-000057
Figure PCTCN2018101290-appb-000058
由此,“氨基环烷基”包含上述环烷基和氨基的定义。
术语“硝基”是指-NO 2
术语“氰基”是指-CN。
术语“巯基”是指-SH。
本发明所述“室温”是指15-30℃。
所述的同位素取代衍生物包括:式I中任意的氢原子被1-5个氘原子取代得到的同位素取代衍生物、式I中任意的碳原子被1-3个碳14原子取代得到的同位素取代衍生物或式I中任意的氧原子被1-3个氧18原子取代得到的同位素取代衍生物。
所述的“前药”是指化合物在体内代谢后转换成原始活性化合物。代表性地讲,前药为非活性物质,或者比活性母体化合物活性小,但可以提供方便的操作、给药或者改善代谢特性。
本发明所述的“药学上可接受的盐”在Berge,et al.,“Pharmaceutically acceptable salts”,J.Pharm.Sci.,66,1-19(1977)中有讨论,并对药物化学家来说是显而易见,所述的盐是基本上无毒性的,并能提供所需的药代动力学性质、适口性、吸收、分布、代谢或***等。本发明所述化合物可以具有酸性基团、碱性基团或两性基团,典型的药学上可接受的盐包括通过本发明化合物和酸反应制备得到的盐,例如:盐酸盐、氢溴酸盐、硫酸盐、焦硫酸盐、硫酸氢盐、亚硫酸盐、亚硫酸氢盐、磷酸盐、磷酸一氢盐、磷酸二氢盐、偏磷酸盐、焦磷酸盐、硝酸盐、乙酸盐、丙酸盐、癸酸盐、辛酸盐、甲酸盐、丙烯酸盐、异丁酸盐、己酸盐、庚酸盐、草酸盐、丙二酸盐、琥珀酸盐、辛二酸盐、苯甲酸盐、甲基苯甲酸盐、邻苯二甲酸盐、马来酸盐、甲磺酸盐、对甲苯磺酸盐、(D,L)-酒石酸,柠檬酸,马来酸,(D,L)-苹果酸,富马酸,丁二酸、琥珀酸盐、乳酸盐、三氟甲磺酸盐、萘-1-磺酸盐、扁桃酸盐、丙酮酸盐、硬脂酸盐、抗坏血酸盐、水杨酸盐。当本发明化合物含有酸性基团时,其药学上可接受的盐还可以包括:碱金属盐,例如钠或钾盐;碱土金属盐,例如钙或镁盐;有机碱盐,例如和氨、烷基氨类、羟基烷基氨类、氨基酸(赖氨酸、精氨酸)、N-甲基葡糖胺等形成的盐。
本发明所述“异构体”是指本发明的式(I)化合物可以有不对称中心和外消旋体、外消旋混合物和单个非对映异构体,所有这些异构体,包括立体异构体、几何异构体均包含在本发明中。在本发明中,式I化合物或其盐以立体异构的形式(例如,其含有一个或多个不对称碳原子)存在时,单独的立体异构体(对映异构体和非对映异构体)以及它们的混合物包括在本发明的范围内。本发明还包括式I表示的化合物或盐的单独异构体,以及与其中一个或多个手性中心反转的异构体的混合物。本发明的范围包括:立体异构体的混合物,以及纯化的对映异构体或对映异构体/非对映异构体富集的混合物。本发明包括所有对映异构体及非对应异构体所有可能的不同组合的立体异构体的混合物。本发明包括上文定义的所有具体基团的立体异构体的全部组合和子集。本发明还包括式I化合物或其盐的几何异构体,所述几何异构体包括顺反异构体。
在不违背本领域常识的基础上,上述各优选条件,可任意组合,即得本发明各较佳 实例。
本发明所用试剂和原料均市售可得。
附图说明
附图1是化合物7-2-1(15mg/kg,30mg/kg,qd)和AZD1775(60mg/kg,bid)在人肺癌H1299异种移植瘤模型中的肿瘤体积变化曲线。
具体实施方式
下面通过实施例的方式进一步说明本发明,但并不因此将本发明限制在所述的实施例范围之中。下列实施例中未注明具体条件的实验方法,按照常规方法和条件,或按照商品说明书选择。
本发明所有化合物的结构可通过核磁共振( 1H NMR)和/或质谱检测(MS)鉴定。 1H NMR化学位移(δ)以PPM记录(10 -6)。NMR通过Bruker AVANCE-400光谱仪进行。合适的溶剂是氘代氯仿(CDCl 3),氘代甲醇(CD 3OD),氘代二甲亚砜(DMSO-d 6),四甲基硅烷作为内标(TMS)。
低分辨率质谱(MS)由Agilent 1200HPLC/6120质谱仪测定,使用XBridge C18,4.6×50mm,3.5μm,梯度洗脱条件1:80-5%溶剂A 1和20-95%溶剂B 1(1.8分钟),然后95%溶剂B 1和5%溶剂A 1(3分钟以上),百分数为某一溶剂占总溶剂体积的体积百分数。溶剂A 1:0.01%三氟乙酸(TFA)的水溶液;溶剂B 1:0.01%三氟乙酸的乙腈溶液;百分数为溶质占溶液的体积百分数。梯度洗脱条件2:80-5%溶剂A 2和20-95%溶剂B 2(1.5分钟),然后95%溶剂B 2和5%溶剂A 2(2分钟以上),百分数为某一溶剂占总溶剂体积的体积百分数。溶剂A 2:10mM的碳酸氢铵的水溶液;溶剂B 2:乙腈。
本发明所有化合物可通过高效液相色谱仪、硅胶柱层析色谱、薄层层析色谱、或快速柱层析色谱进行分离。
高效液相色谱仪(prep-HPLC)使用岛津LC-20制备液相色谱,色谱柱为:waters xbridge Pre C18,10um,19mmx250mm。分离条件1:流动相A:0.05%三氟乙酸水溶液,流动相B:乙腈;流动相B为40%,洗脱时间:20分钟。分离条件2:流动相A:10mmol/L碳酸氢铵水溶液,流动相B:乙腈;梯度洗脱流动相B从25%到80%,洗脱时间30分钟。分离条件3:流动相A:0.05%盐酸水溶液,流动相B:乙腈;梯度洗脱流动相B从75%到25%,洗脱时间:30分钟。检测波长:214nm&254nm;流速:15.0mL/分钟。
本发明具体实施例37~75各化合物均可通过prep-HPLC纯化得到,使用分离条件1得到的化合物为三氟乙酸盐,使用条件2得到的化合物为游离碱,使用条件3得到的化合物为盐酸盐。
快速柱层析(Flash柱层析)(flash system/Cheetah TM)使用的是Agela Technologies MP200,配套使用的分离柱为Flash columm Silica-CS(80g),Cat No.CS140080-0。
薄层层析色谱(prep-TLC)是烟台新诺化工,涂层厚度0.2±0.03mm,规格20×20cm。硅胶柱层析一般使用烟台黄海200-300目硅胶作为载体。
硅胶柱层析色谱、薄层层析色谱、或快速柱层析色谱使用的洗脱剂或展开剂选自乙酸乙酯、甲醇、二氯甲烷、石油醚和正己烷中的一种或多种。
本发明实施例中所述氢气氛是用氢气球来提供。
实施例1:化合物1.3的合成
Figure PCTCN2018101290-appb-000059
步骤1:将5-硝基吲哚(1g,6.2mmol)、二甲胺基氯乙烷盐酸盐(1.3g,9.3mmol)和无水碳酸钾(3.4g,25mmol)的N,N-二甲基甲酰胺(DMF)(15mL)混合物在70℃下搅拌3小时。得到的混合物冷至室温,然后倒入冰水中(50mL)搅拌,混合物用乙酸乙酯萃取(20mL×3),合并有机相,依次用水、饱和食盐水洗涤,有机相用无水硫酸钠干燥、过滤、浓缩,残留物用硅胶柱层析(二氯甲烷/甲醇=20/1)纯化得化合物1.2(700mg,产率:32%)为无色油状液体。
步骤2:将化合物1.2(700mg,3.0mmol)加入到钯碳(100mg,10%)的甲醇(20mL)混合物中,用氢气将反应体系置换3次,得到的混合物在氢气氛下室温搅拌过夜,反应液用硅藻土过滤除去催化剂,滤液浓缩得到1-(2-(二甲基氨基)乙基)-1H-吲哚-5-胺(化合物1.3,600mg,产率:98%)为棕色油状物。m/z:[M+H] +204.0。
实施例2:化合物1.7的合成
Figure PCTCN2018101290-appb-000060
步骤1:冰浴条件下,向5-硝基吲哚(4g,24.7mmol)的DMF(100mL)溶液中分批加入钠氢(3.95g,60%,98.8mmol),得到的混合物缓慢升至室温并搅拌2小时。然后将反应液冰浴冷却,加入2-溴乙醇(12.3g,98.4mmol)。混合物继续在室温下搅拌24小时。然后将反应体系用乙酸乙酯稀释,依次用水、饱和食盐水洗涤,有机相用无水硫酸钠干燥、过滤、浓缩,残留物用硅胶柱层析(二氯甲烷/甲醇=20/1)纯化得化合物1.4(2.1g,产率:41%)为无色油状物。
步骤2:冰浴条件下,向化合物1.4(1g,4.85mmol)的二氯甲烷(20mL)溶液中分批加入戴斯-马丁试剂(6.2g,14.7mmol)。将得到的混合物升至室温搅拌3小时。然后将反应液用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤、浓缩得化合物1.5(810mg,产率:82%)为黄色固体。
步骤3:向化合物1.5(400mg,1.96mmol)和氮杂环丁烷(168mg,2.94mmol)的甲醇(10mL)溶液中加一滴醋酸后,将反应体系在室温下搅拌30分钟。向反应液中分批加入氰基硼氢化钠(369mg,5.88mmol)。将得到的混合物在室温下继续搅拌1小时。然后将反应液直接浓缩,残留物用硅胶柱层析(二氯甲烷/甲醇=10/1)纯化得化合物1.6(270mg,产率:57%)为黄色固体。
步骤4:向化合物1.6(270mg,1.10mmol)的乙酸乙酯(5mL)和四氢呋喃(5mL)混合溶液中加入钯炭(50mg,10%),得到的混合物用氢气置换3次,然后混合物在氢气氛下室温搅拌3小时。反应体系用硅藻土过滤,滤液减压浓缩得到1-(2-(氮杂环丁烷-1-基)乙基)-1H-吲哚-5-胺(化合物1.7,270mg,产率:100%)为黄色固体。
实施例3:化合物1.8~1.9的合成
利用化合物1.7的合成方法,将步骤3中的氮杂环丁烷替换为吡咯烷得到1-(2-(吡咯烷-1-基)乙基)-1H-吲哚-5-胺(化合物1.8),m/z:[M+H] +230.0。
利用化合物1.7的合成方法,将步骤3中的氮杂环丁烷替换为哌啶得到1-(2-(哌啶-1-基)乙基)-1H-吲哚-5-胺(化合物1.9),m/z:[M+H] +244.0。
实施例4:化合物2.5的合成
Figure PCTCN2018101290-appb-000061
步骤1:向5-硝基吲哚(1.01g,6.2mmol)的DMF(10.0mL)溶液中缓慢加入钠氢(0.5g,60%,12.4mmol)。反应体系在室温下搅拌30分钟,接着把1-Boc-4-甲磺酰氧基哌啶(1.72g,6.2mmol)加到上述反应体系中,将反应体系在100℃下搅拌12小时。然后,再加水淬灭反应,用乙酸乙酯萃取。合并有机相并用饱和食盐水洗涤,分离有机相并用无水硫酸钠干燥,过滤、浓缩,残留物用硅胶柱层析(35%的乙酸乙酯石油醚溶液)纯化得到化合物2.2(0.85g,产率:75%)为黄色油状。m/z:[M+H] +346.0。
步骤2:将化合物2.2(0.85g,2.4mmol)的三氟乙酸(2.0mL)和二氯甲烷(4.0mL)混合溶液在室温下搅拌1小时,然后将反应体系减压浓缩得到化合物2.3(0.55g,粗品)为黄色油状物。m/z:[M+H] +246.0。
步骤3:将化合物2.3(0.50g,2.0mmol)、37%甲醛(0.33g,4.0mmol)、三乙胺(0.1mL)、醋酸(2滴)的1,2-二氯乙烷(10mL)溶液在室温下搅拌1小时。然后把氰基硼氢化钠(0.39g,6.0mmol)加入到上述反应体系中,得到的混合物再在室温下搅拌2小时。用氢氧化钠水溶液(1.0M)淬灭反应,得到的混合物加水稀释后用乙酸乙酯萃取。合并有机相并用饱和食盐水洗涤,分离有机相并用无水硫酸钠干燥,过滤、浓缩,残留物用硅胶柱层析(5%的甲醇二氯甲烷溶液)纯化得到化合物2.4(0.35g,产率:66%)为黄色固体。m/z:[M+H] +260.0。
步骤4:向化合物2.4(0.35g,1.3mmol)的甲醇(10.0mL)溶液中加入钯碳(0.05g,10%),反应体系用氢气置换3次,然后反应体系在氢气氛下室温搅拌12小时。过滤、减压浓缩得到1-(1-甲基哌啶-4-基)-1H-吲哚-5-胺(化合物2.5,0.10g,粗品)为淡黄色固体。m/z:[M+H] +230.2。
实施例5:化合物2.6~2.9的合成
利用化合物2.5的合成方法,将步骤1中的1-Boc-4-甲磺酰氧基哌啶分别替换为1-Boc-3-甲磺酰氧基吡咯烷、1-Boc-3-溴环丁烷、1-Boc-3-甲磺酰氧基哌啶得到化合物2.6、2.7、2.9:
利用化合物2.5的合成方法,将步骤3中的甲醛替换为丙酮得到化合物2.8:
编号 名称 MS
2.6 1-(1-甲基吡咯烷-3-基)-1H-吲哚-5-胺 m/z:[M+H] +216.0
2.7 1-(1-甲基氮杂环丁烷-3-基)-1H-吲哚-5-胺 m/z:[M+H] +202.2
2.8 1-(1-异丙基哌啶-4-基)-1H-吲哚-5-胺 m/z:[M+H] +258.2
2.9 1-(1-甲基哌啶-3-基)-1H-吲哚-5-胺 m/z:[M+H] +230.2
实施例6:化合物2.10~2.11的合成
利用化合物2.5的合成方法,将步骤1中的5-硝基吲哚替换为5-硝基-1H-吡咯并[2,3-]吡啶、步骤3中的37%甲醛替换为丙酮得到1-(1-异丙基哌啶-4-基)-1H-吡咯并[2,3-b]吡啶-5-胺(化合物2.10)。
利用化合物2.5的合成方法,将步骤1中的5-硝基吲哚替换为5-硝基-1H-吡咯并[2,3-b]吡啶得到1-(1-甲基哌啶-4-基)-1H-吡咯并[2,3-b]吡啶-5-胺(化合物2.11)。
实施例7:化合物2.15的合成
Figure PCTCN2018101290-appb-000062
步骤1:将5-硝基吲哚(1.62g,10.0mmol)和4-四氢吡喃基甲磺酸酯(3.6g,20.0mmol)溶解于DMF(20mL)中,然后加入碳酸铯(6.5g,20.0mmol),得到的混合物在100℃下搅拌过夜,然后冷却至室温后用水(100mL)淬灭反应,用乙酸乙酯萃取,有机相分离后用无水硫酸钠干燥、减压浓缩,残留物用硅胶柱层析(石油醚/乙酸乙酯=2/1)纯化得到化合物2.14(950mg,产率:39%)为淡黄色固体。m/z:[M+H] +247.3。
步骤2:向化合物2.14(950mg,3.8mmol)的甲醇(30mL)溶液中加入钯碳(50mg,10%),反应体系用氢气置换3次,然后在氢气氛下搅拌30分钟。将混合物经硅藻土过滤,滤液减压浓缩后得到1-(四氢-2H-吡喃-4-基)-1H-吲哚-5-胺(化合物2.15,710mg,产率:85%)为棕色油状物。m/z:[M+H] +217.2。
实施例8:化合物3.1的合成
利用化合物2.5的合成方法,用5-硝基-1H-苯并咪唑和1-Boc-3-溴氮杂环丁烷为起始原料反应得到1-(1-甲基氮杂环丁烷-3-基)-1H-苯并[d]咪唑-5-胺(化合物3.1)。m/z:[M+H] +203.2。
实施例9:化合物4.2的合成
Figure PCTCN2018101290-appb-000063
步骤1:冰浴条件下,将醋酸酐(49.9mg,0.49mmol)加入到化合物2.3(0.1g,0.41mmol)和三乙胺(82.4mg,0.82mmol)的二氯甲烷(5mL)溶液中,反应体系在室温下搅拌2小时。然后用二氯甲烷稀释,混合物用水和饱和食盐水洗涤,分离有机相并用 无水硫酸钠干燥,过滤、浓缩得到化合物4.1(0.1g,产率:85%)为黄色固体。
步骤2:利用化合物2.5步骤4的合成方法,用化合物4.1(100mg,0.35mmol)反应得到1-(4-(5-氨基-1H-吲哚-1-基)哌啶-1-基)乙酮(化合物4.2,80mg,产率:89%)为淡黄色固体。
实施例10:化合物5.2的合成
Figure PCTCN2018101290-appb-000064
步骤1:把2,2,2-三氟乙基三氟甲烷磺酸酯(284mg,1.22mmol)加入到化合物2.3(0.1g,0.41mmol)和三乙胺(124mg,1.22mmol)的四氢呋喃(10mL)溶液中,反应体系在70℃下搅拌过夜。然后将反应液冷却至室温,直接减压浓缩,残留物用硅胶柱层析(10%的甲醇二氯甲烷溶液)纯化得到化合物5.1(95mg,产率:71%)为无色液体。m/z:[M+H] +288.2。
步骤2:利用化合物2.5步骤4的合成方法,用化合物5.1(95mg,0.29mmol)反应得到1-(1-(2,2,2-三氟乙基)哌啶-4-基)-1H-吲哚-5-胺(化合物5.2,85mg,产率:100%)为淡黄色固体。
实施例11:化合物6.6的合成
Figure PCTCN2018101290-appb-000065
步骤1:冰浴条件下,向5-硝基吲哚-2-甲酸乙酯(2.34g,10.0mmol)的DMF(50mL)溶液中加入钠氢(600mg,60%,15.0mmol),得到的混合物在室温下搅拌0.5小时,然后加入N-Boc-溴乙胺(2.92g,13.0mmol),反应体系在70℃下搅拌过夜。然后用乙酸乙酯(100mL)稀释,有机相用饱和食盐水洗涤。分离有机相并用无水硫酸钠干燥,过滤、减压浓缩。残留物用硅胶柱层析(石油醚/乙酸乙酯=4/1)纯化得到化合物6.1(3.05g,产率:81%)为淡黄色固体。m/z:[M-Boc] +278.1。
步骤2:将化合物6.1(3.05g,8.1mmol)的三氟乙酸(4mL)和二氯甲烷(20mL)混合溶液在室温下搅拌2小时,然后用饱和的碳酸氢钠水溶液淬灭反应,加水稀释,得到的混合物用二氯甲烷萃取。合并有机相并用饱和食盐水洗涤,分离有机相并用无水硫酸钠干燥,过滤、减压浓缩,残留物用硅胶柱层析(二氯甲烷/甲醇=10/1)纯化得到化合物6.2(1.1g,产率:49%)为淡黄色固体。m/z:[M+H] +278.2。
步骤3:化合物6.2(1.1g,4.0mmol)和甲醇钠(643mg,11.9mmol)的乙醇溶液(20mL)在55℃下搅拌2小时。然后向反应体系中加入饱和食盐水,用二氯甲烷萃取。 合并有机相,并用无水硫酸钠干燥,过滤、减压浓缩得化合物6.3(361mg,产率:39%)为黄色固体。m/z:[M+H] +232.4。
步骤4:冰浴条件下,向化合物6.3(310mg,1.3mmol)的DMF(20mL)溶液中,加入钠氢(81mg,2.0mmol,60%),得到的混合物在室温下搅拌15分钟,然后加入碘甲烷(285mg,2.0mmol),反应体系在室温下继续搅拌30分钟。然后用乙酸乙酯(100mL)稀释,有机相用饱和食盐水洗涤。分离有机相并用无水硫酸钠干燥,过滤、减压浓缩得化合物6.4(358mg,粗品)为黄色固体。m/z:[M+H] +246.4。
步骤5:向化合物6.4(358mg)的甲醇(15mL)溶液中加入钯碳(180mg,10%),反应体系用氢气置换3次后,在氢气氛下搅拌0.5小时。反应液用硅藻土过滤,滤液减压浓缩,得到化合物6.5(277mg,粗品)为红色固体。m/z:[M+H] +216.2。
步骤6:冰浴条件下,向化合物6.5(275mg,1.3mmol)的甲基叔丁基醚(30mL)溶液中滴加四氢铝锂的四氢呋喃溶液(1.95mL,4.9mmol,2.5M),反应体系在60℃搅拌5小时。然后用饱和的酒石酸钾钠水溶液淬灭反应,乙酸乙酯(100mL)稀释,有机相用饱和食盐水洗涤。分离有机相并用无水硫酸钠干燥,过滤、减压浓缩得2-甲基-1,2,3,4-四氢吡啶并[1,2-a]吲哚-8-胺(化合物6.6,248mg,3步产率:96%)为红色固体。m/z:[M+H] +202.2。
实施例12:化合物7.6的合成
Figure PCTCN2018101290-appb-000066
步骤1:在-10℃下,将氯甲酸异丁酯(10.2mL,78mmol)滴加到1,1-环丙基二甲酸单甲酯(10g,69.4mmol)和三乙胺(10.8mL,78mmol)的四氢呋喃(200mL)溶液中,并在-10℃下搅拌1小时,将反应液升至0℃,过滤掉反应液中固体,滤液待用。冰浴条件下,将硼氢化钠(7.87g,208mmol)溶入到四氢呋喃(100mL)和水(25mL)的混合溶液中,然后滴加到上述滤液中,滴加时间1小时。得到的混合物继续在冰浴条件下搅拌1小时。将反应液倒入到20%的柠檬酸冰水溶液中,搅拌5分钟,浓缩除掉有机溶液,水相用乙酸乙酯萃取,合并有机相,依次用水和饱和食盐用洗涤,无水硫酸钠干燥、过滤,减压浓缩得到化合物7.1(6.9g,产率:77%)为油状液体。
步骤2:冰浴条件下,将甲基磺酰氯(7.4g,64.5mmol)滴加到化合物7.1(5.6g,43mmol)和三乙胺(8.7mL,86mmol)的二氯甲烷(100mL)溶液中,得到的混合物在室温下搅拌3小时。反应液用二氯甲烷淬灭,依次用盐酸(1.0M)、水和饱和食盐水洗涤,有机相用无水硫酸钠干燥、过滤、减压浓缩得到化合物7.2(8.9g,产率:100%)为白色固体。
步骤3:将化合物7.2(10.7g,51.5mmol)加入到5-硝基吲哚(5.56g,34.3mmol) 和碳酸铯(33.5g,103mmol)的DMF(60mL)溶液中,得到的混合物在100℃下搅拌过夜,反应液冷却至室温并倒入到水中,水相用乙酸乙酯萃取(20mL×3),合并有机相,依次用水、饱和食盐水洗涤,有机相减压浓缩,残留物用硅胶柱层析纯化(石油醚/乙酸乙酯=5/1)得化合物7.3(8g,产率:72%)为黄色固体。m/z:[M+H] +275.2。
步骤4:将一水合氢氧化锂(475mg,11.3mmol)的水溶液(10mL)滴加到化合物7.3(1.24g,4.5mmol)的四氢呋喃(30mL)溶液中,反应体系在室温下搅拌过夜,减压浓缩除去四氢呋喃,水相先用石油醚/乙酸乙酯混合溶液(1/1)洗涤,然后用饱和的柠檬酸水溶液调pH=3,水相用乙酸乙酯萃取(10mL×3),合并有机相并用水和饱和食盐水洗涤,无水硫酸钠干燥、过滤、减压浓缩得到化合物7.4(920mg,产率:79%)为黄色固体。
步骤5:冰浴条件下,将叠氮磷酸二苯酯(605mg,2.2mmol)滴加到化合物7.4(520mg,2.0mmol)和三乙胺(607mg,6.0mmol)的甲苯(15mL)溶液中,反应体系在0℃下搅拌2小时,然后将反应体系升至室温继续搅拌4小时,将叔丁醇(10mL)加入到反应体系中升温至回流并搅拌过夜。然后将反应液冷却至室温,浓缩,将残留物溶于二氯甲烷(20mL),依次用饱和碳酸氢钠水溶液、水、饱和食盐水洗涤,浓缩有机相,残留物用硅胶柱层析纯化(石油醚/乙酸乙酯=5/1)得到化合物7.5(460mg,产率:69%)为黄色固体。m/z:[M-55] +276.2。
步骤6:将化合物7.5(460mg,1.39mmol)和钯碳(100mg,10%)混于甲醇(30mL)溶液中。反应体系用氢气置换3次,然后在氢气氛下搅拌2小时。用硅藻土过滤,所得滤液减压浓缩得到(1-((5-氨基-1H-吲哚-1-基)甲基)环丙基)氨甲基叔丁酯(化合物7.6,308mg,产率:74%)为棕色固体。m/z:[M+H] +302.2。
实施例13:化合物7.8的合成
Figure PCTCN2018101290-appb-000067
步骤1:冰浴条件下,将钠氢(121mg,3.0mmol,60%)加入到化合物7.5(500mg,1.5mmol)的DMF(10mL)溶液中,反应体系搅拌0.5小时后,将碘甲烷(639mg,4.5mmol)加入到上述反应体系中,室温搅拌2小时后将反应液倒入到冰水中,水相用乙酸乙酯萃取(20mL×3),合并有机相,依次用水、饱和食盐水洗涤,浓缩有机相,残留物用硅胶柱层析(石油醚/乙酸乙酯=5/1)纯化得到化合物7.7(362mg,产率:70%)为油状液体。m/z:[M+H] +290.2。
步骤2:将化合物7.7(362mg,1.0mmol)和钯碳(100mg,10%)的甲醇(15mL)混合物用氢气置换3次,然后在氢气氛下室温搅拌2小时。将反应液用硅藻土过滤,滤液减压浓缩得(1-((5-氨基-1H-吲哚-1-基)甲基)环丙基)(甲基)氨甲基叔丁酯(化合物7.8,315mg,产率:100%)为棕色固体。m/z:[M+H] +316.2。
实施例14:化合物8.3的合成
Figure PCTCN2018101290-appb-000068
步骤1:向5-硝基吲哚-2-羧酸乙酯(5g,21.3mmol)的甲醇(50mL)、四氢呋喃(20mL)和水(10mL)的混合物溶液中加入氢氧化钠(2.5g,64.0mmol),反应体系在室温下搅拌16小时。加水(100mL)稀释,水相用石油醚萃取(50mL×2),然后将水相用盐酸(2.0M)调节pH=6,用乙酸乙酯萃取(50mL×3),合并有机相后用饱和食盐水洗涤、无水硫酸钠干燥,过滤、减压浓缩得到化合物8.1(4g,产率:91%)为淡黄色固体。m/z:[M+H] +207.0。
步骤2:将化合物8.1(4g,19.4mmol)、二甲胺盐酸盐(2.4g,29.1mmol)、HATU(11.0g,29.1mmol)和N,N-二异丙基乙胺(9.6mL,58.2mmol)的DMF(50mL)溶液在室温下搅拌3小时,加入水(150mL)淬灭反应,得到的混合物用乙酸乙酯萃取(100mL×3),合并有机相后依次用水、饱和食盐水洗涤、无水硫酸钠干燥,过滤、减压浓缩,残留物用乙酸乙酯石油醚溶液(10%)打浆,过滤、滤饼真空干燥后得到化合物8.2(1.5g,产率:33%)为淡黄色固体。m/z:[M+H] +234.2。
步骤3:室温下,向化合物8.2(310mg,1.3mmol)的无水四氢呋喃(5mL)溶液中滴加氢化铝锂的四氢呋喃溶液(4.0mL,1.0M),然后将反应体系在70℃搅拌2小时,冷却至室温,并用氢氧化钠水溶液(2.0M,10mL)淬灭反应,然后用乙酸乙酯萃取(50mL×3),合并有机相后用饱和食盐水洗涤、无水硫酸钠干燥,过滤、减压浓缩,残留物用Flash柱层析(二氯甲烷/甲醇=10/1)纯化得到2-((二甲基氨基)甲基)-1H-吲哚-5-胺(化合物8.3,34mg,产率14%)为橘黄色油状物。m/z:[M+H] +190.2; 1H NMR(400MHz,DMSO-d 6):δ10.46(s,1H),6.99(d,J=8.4Hz,1H),6.60(s,1H),6.41(d,J=8.4Hz,1H),5.96(s,1H),4.38(br.s,2H),3.44(s,2H),2.16(s,6H)。
实施例15:化合物9.1的合成
Figure PCTCN2018101290-appb-000069
将6-溴-2-氨基萘(500mg,2.25mmol)、1-甲基哌嗪(270mg,2.7mmol)、氢氧化铯水合物(760mg,4.5mmol)的二甲基亚砜(5.0mL)溶液在120℃下搅拌20小时,然后将反应体系冷却至室温,加入冰水(10mL)稀释,水相用二氯甲烷(20mL×2)萃取,有机相依次用水、饱和食盐水洗涤,无水硫酸钠干燥,过滤、浓缩,残留物用Flash柱层析(石油醚/乙酸乙酯=1/3)纯化得到6-(4-甲基哌嗪-1-基)萘-2-胺(化合物9.1,70mg,产率:13%)为棕色固体。m/z:[M+H] +242.2。
实施例16:化合物10.4的合成
Figure PCTCN2018101290-appb-000070
步骤1:将2-氟-4-硝基苯腈(2g,12.0mmol)、巯基乙酸乙酯(1.37g,11.4mmol)和三乙胺(5mL,36mmol)加入到乙腈(30mL)中,反应体系在室温下搅拌16小时。将反应液减压浓缩后,残留物用石油醚和乙酸乙酯(4/1)的混合溶液洗涤,过滤、滤饼真空干燥得化合物10.1(1.5g,产率:48%)为棕色固体。
步骤2:将化合物10.1(600mg,2.25mmol)、亚硝酸叔丁酯(349mg,3.38mmol)和溴化铜(1g,4.51mmol)的乙腈(20mL)混合物在65℃下搅拌16小时。将反应液减压浓缩后倒入水中,过滤、滤饼真空干燥得化合物10.2(600mg,产率:81%)为类白色固体。m/z:[M+H] +329.8。
步骤3:将化合物10.2(600mg,1.82mmol)、1-甲基哌嗪(219mg,2.18mmol)、三(二亚苄基丙酮)二钯(83mg,0.091mmol)、1,1'-联萘-2,2'-双二苯膦(113mg,0.182mmol)和碳酸铯(1.18g,3.64mmol)加入到甲苯(15mL)中,反应体系在氮气保护下加热回流16小时。将反应液过滤,滤液浓缩,残留物用硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化得化合物10.3(150mg,产率:24%)为黄色固体。m/z:[M+H] +349.8。
步骤4:将化合物10.3(100mg,0.28mmol)和钯碳(50mg,10%)加入到乙酸乙酯(15mL)中,反应体系用氢气置换3次,然后在氢气氛下搅拌0.5小时,用硅藻土过滤反应液,滤饼用乙酸乙酯洗涤,滤液减压浓缩得到6-氨基-3-(4-甲基哌啶-1-基)苯并[b]噻吩-2-羧酸乙酯(化合物10.4,90mg,粗品)为棕色油状物。m/z:[M+H] +319.8。
实施例17:化合物10.7的合成
Figure PCTCN2018101290-appb-000071
步骤1:将化合物10.1(500mg,1.88mmol)和氯化锂(995mg,23.5mmol)加入到二甲基亚砜(10mL)中,反应体系在160℃下搅拌5小时。将反应液冷却后,倒入水中,用乙酸乙酯萃取,有机相用无水硫酸钠干燥后,减压浓缩,残留物用硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化得化合物10.5(200mg,产率:55%)为类棕色固体。m/z:[M+H] +194.8。
步骤2:将化合物10.5(200mg,1.03mmol),氮芥盐酸盐(297mg,1.54mmol)和碳酸钾(569mg,4.12mmol)加入到叔丁醇(15mL)中,反应体系用微波加热到140℃搅拌3小时。将反应液过滤,减压浓缩后用prep-TLC(石油醚/乙酸乙酯=1/1)分离得到化合物10.6(40mg,产率:14%)为棕色液体。m/z:[M+H] +277.8。
步骤3:将化合物10.6(40mg,0.144mmol),铁粉(40mg,0.72mmol),氯化铵(39mg,0.72mmol)和水(1mL)加入到乙醇(15mL)中,反应体系加热回流2小时。然 后将反应液冷却至室温,过滤,滤液浓缩,残留物用二氯甲烷和甲醇(10/1)的混合溶剂洗涤,洗涤液浓缩得到3-(4-甲基哌嗪-1-基)苯并[b]噻吩-6-胺(化合物10.7,34mg,产率:95%)为深棕色液体。m/z:[M+H] +247.8。
实施例18:化合物11.2的合成
Figure PCTCN2018101290-appb-000072
步骤1:将化合物2.3(110mg,0.45mmol)、溴乙醇(168mg,1.35mmol)和碳酸钾(186mg,1.35mmol)的DMF(5mL)溶液在100℃下搅拌过夜。反应液冷却至室温,加水淬灭反应,水相用乙酸乙酯萃取(15mL×3),合并有机相,依次用水、饱和食盐水洗涤,有机相用无水硫酸钠干燥、过滤、减压浓缩,残留物用prep-TLC纯化(二氯甲烷/甲醇=10/1)得到化合物11.1(82mg,产率:60%)为棕色油状物。m/z:[M+H] +290.2。
步骤2:将化合物11.1(82mg,0.28mmol)和钯碳(40mg,10%)的甲醇(10.0mL)溶液用氢气置换3次,然后在氢气氛下室温搅拌2小时。反应液用硅藻土过滤,滤液减压浓缩得到2-(4-(5-氨基-1H-吲哚-1-基)哌啶-1-基)乙醇(化合物11.2,74mg,产率:100%)为棕色固体。
实施例19:化合物12.2的合成
Figure PCTCN2018101290-appb-000073
步骤1:向6-硝基吲哚(1.62g,10.0mmol)和1-甲基-4-哌啶酮(2.26g,20mmol)的甲醇(20mL)溶液中加入氢氧化钾(1.12g,20mmol)。反应体系加热至90℃搅拌过夜。然后将反应液冷却至室温后加水(100mL)淬灭反应,水相用乙酸乙酯萃取,分离有机相,用无水硫酸钠干燥、过滤、减压浓缩,残留物用硅胶柱层析(二氯甲烷/甲醇=10/1)纯化得到化合物12.1(760mg,产率:30%)为淡黄色固体。m/z:[M+H] +258.4。
步骤2:向化合物12.1(760mg,2.9mmol)的甲醇(30mL)溶液中加入钯碳(50mg,10%),反应体系用氢气置换3次,然后在氢气氛下搅拌30分钟。将混合物经硅藻土过滤,滤液减压浓缩后得到3-(1-甲基哌啶-4-基)-1H-吲哚-6-胺(化合物12.2,510mg,产率:77%)为棕色油状物。m/z:[M+H] +230.2。
实施例20:化合物12.3的合成
利用化合物12.2的合成方法,将步骤1中的6-硝基吲哚替换为1-甲基-6-硝基-1H-吲哚得到1-甲基-3-(1-甲基哌啶-4-基)-1H-吲哚-6-胺(化合物12.3)。
实施例21:化合物13.4的合成
Figure PCTCN2018101290-appb-000074
步骤1:向2-氨基-4-硝基苯甲酸(1.82g,10.0mmol)和碳酸钾(2.76g,20.0mmol)的水(30mL)溶液中加入氯乙酸(945mg,10.0mmol)。得到的混合物在90℃下搅拌过夜,冷却至室温后用盐酸(1M,100mL)淬灭反应并调节pH=4,将黄色沉淀物过滤,滤饼真空干燥后得到化合物13.1(1.76g,产率:73%)为淡黄色固体。
步骤2:室温下,向化合物13.1(1.76g,7.33mmol)的乙酸酐(20mL)悬浊液中加入醋酸钠(2.39g,29.2mmol),反应体系在130℃下搅拌4小时,然后冷却至室温,用水(100mL)淬灭反应后用二氯甲烷(20mL×2)萃取,分离有机相,用无水硫酸钠干燥、过滤、减压浓缩。残留物冷却后析出黄色固体,过滤,滤饼用石油醚润洗,真空干燥后得到化合物13.2(1.05g,产率:55%)为棕黄色固体。m/z:[M+H] +263.4。
步骤3:将化合物13.2(200mg,0.76mmol)、1-甲基哌嗪(305mg,3.05mmol)溶于醋酸(5mL)中,反应体系在150℃下微波反应0.5小时。反应液减压浓缩后经硅胶柱层析(二氯甲烷/甲醇=10/1)纯化得到化合物13.3(76.0mg,产率:33%)为棕色固体。m/z:[M+H] +303.4。
步骤4:向化合物13.3(20mg,0.07mmol)的甲醇(5.0mL)溶液中加入雷尼镍(5mg),体系用氢气置换3次,然后在氢气氛下室温搅拌1小时。过滤、滤液减压浓缩得到3-(4-甲基哌嗪-1-基)-1H-吲哚-6-胺(化合物13.4,16mg,产率:89%)为棕色固体。m/z:[M+H] +231.2。
实施例22:化合物13.7的合成
Figure PCTCN2018101290-appb-000075
步骤1:向化合物13.3(110mg,0.36mmol)的甲醇(10.0mL)溶液中加入三乙胺(120mg,1.10mmol),反应体系在80℃下搅拌3小时。反应体系减压浓缩得到化合物13.5(94mg,产率:100%)为黄色固体。m/z:[M+H] +261.4。
步骤2:冰浴条件下,向化合物13.5(94mg,0.36mmol)的四氢呋喃(5.0mL)溶液中分批加入钠氢(60%,20mg,0.47mmol),反应体系0℃下搅拌0.5小时,加入碘甲烷(80mg,0.54mmol),然后继续在0℃下搅拌1小时。将反应体系直接减压浓缩,残留物用prep-TLC(二氯甲烷/甲醇=10/1)纯化得到化合物13.6(45mg,产率:46%)为黄色固体。m/z:[M+H] +275.4。
步骤3:向化合物13.6(45mg,0.16mmol)的甲醇(5.0mL)溶液中加入雷尼镍(20mg),反应体系用氢气置换3次,然后在氢气氛下搅拌1小时。过滤、滤液减压浓缩得 到1-甲基-3-(4-甲基哌嗪-1-基)-1H-吲哚-6-胺(化合物13.7,40mg,产率:100%)为棕色固体。m/z:[M+H] +245.2。
实施例23:化合物14.2的合成
Figure PCTCN2018101290-appb-000076
步骤1:氮气保护下,向6-硝基吲哚(500mg,3.1mmol)的甲苯(10.0mL)溶液中依次加入2-溴吡啶(410mg,2.6mmol)、碘化亚铜(25mg,0.13mmol)、磷酸钾(1.32g,6.2mmol)、N,N’-二甲基乙二胺(55mg,0.63mmol),然后将反应体系在110℃下搅拌16小时。反应体系减压浓缩,残留物经Flash柱层析(二氯甲烷/甲醇=50/1)纯化得到化合物14.1(430mg,产率为58%)为黄色固体。m/z:[M+H] +240.2。
步骤2:向化合物14.1(130mg,0.54mmol)的甲醇(10.0mL)溶液中加入雷尼镍(20mg),反应体系用氢气置换3次,然后在氢气氛中搅拌1小时。过滤、滤液减压浓缩得到1-(吡啶-2-基)-1H-吲哚-5-胺(化合物14.2,113mg,产率:99%)为棕色固体。m/z:[M+H] +210.3。
实施例24:化合物14.3~14.4的合成
利用化合物14.2的合成方法,将步骤1中的2-溴吡啶替换为3-溴吡啶得到1-(吡啶-3-基)-1H-吲哚-5-胺(化合物14.3),m/z:[M+H] +210.2。
利用化合物14.2的合成方法,将步骤1中的2-溴吡啶替换为4-溴吡啶得到1-(吡啶-4-基)-1H-吲哚-5-胺(化合物14.4),m/z:[M+H] +210.2。
实施例25:化合物14.7~14.8的合成
利用化合物14.2的合成方法,用5-硝基-1H-吡咯并[2,3-]吡啶和3-溴吡啶为起始原料得到1-(吡啶-3-基)-1H-吡咯[2,3-b]吡啶-5-胺(化合物14.7),m/z:[M+H] +211.4。
利用化合物14.2的合成方法,用5-硝基-1H-吡咯并[2,3-]吡啶和2-溴吡啶为起始原料得到1-(吡啶-2-基)-1H-吡咯[2,3-b]吡啶-5-胺(化合物14.8),m/z:[M+H] +211.4。
实施例26:化合物15.3的合成
Figure PCTCN2018101290-appb-000077
步骤1:冰浴条件下,将钠氢(229mg,5.73mmol,60%)加入到4-溴-6-硝基-1H-吲哚(690mg,2.86mmol)的无水DMF(15mL)溶液中,得到的混合物在室温下搅拌0.5小时,然后将碘甲烷(1.22g,8.59mmol)加入到上述混合物中,将反应体系继续搅拌2小时。然后将反应液缓慢倒入搅拌的冰水中,水相用乙酸乙酯(10mL×2)萃取,合并有机相,依次用水、饱和食盐水洗涤,浓缩有机相,残留物用硅胶柱层析(石油醚/乙酸乙酯=5/1)纯化得到化合物15.1(529mg,产率:73%)为黄色固体。
步骤2:将三(二亚苄基丙酮)钯(7.2mg,7.8μmol)加入到化合物15.1(200mg,0.784mmol)、1-甲基哌嗪(78.4mg,0.784mmol)、碳酸铯(767.1mg,2.35mmol)和(±)-2,2'-双-(二苯膦基)-1,1'-联萘(14.7mg,0.024mmol)的1,4-二氧六环(15mL)溶液中,用氮气将反应体系置换3次,反应体系在氮气保护下在110℃下搅拌过夜,然后冷至室温后用硅藻土过滤,滤饼用乙酸乙酯洗涤,滤液减压浓缩,残留物用prep-TLC(二氯甲烷/甲醇=10/1)纯化得到化合物15.2(210mg,产率:98%)为黄色固体。
步骤3:将化合物15.2(210mg,0.77mmol)和钯碳(60mg,5%)的甲醇(15mL)混合物用氢气置换3次。然后将反应体系在氢气氛下室温搅拌1小时。溶液过滤、滤液减压浓缩得到1-甲基-4-(4-甲基哌嗪-1-基)-1H-吲哚-6-胺(化合物15.3,205mg,产率:100%)为棕色固体。
实施例27:化合物15.4~15.10的合成
利用化合物15.3的合成方法,将步骤2中的1-甲基哌嗪替换为吡咯烷得到1-甲基-4-(吡咯烷-1-基)-1H-吲哚-6-胺(化合物15.4)。
利用化合物15.3的合成方法,将步骤2中的1-甲基哌嗪替换为二甲胺盐酸盐得到N 4,N 4,1-三甲基-1H-吲哚-4,6-二胺(化合物15.5)。
利用化合物15.3的合成方法,将步骤2中的1-甲基哌嗪替换为吗啉得到1-甲基-4-吗啉基-1H-吲哚-6-胺(化合物15.6)。
利用化合物15.3的合成方法,将步骤2中的1-甲基哌嗪替换为1-Boc-哌嗪得到1-甲基-4-(1-N-Boc-哌嗪-4-基)-1H-吲哚-6-胺(化合物15.7)。m/z:[M+H] +331.0。
利用化合物15.3的合成方法,将步骤2中的1-甲基哌嗪替换为(S)-1-Boc-2-甲基哌嗪得到(S)-1-甲基-4-(1-N-Boc-2-甲基哌嗪-4-基)-1H-吲哚-6-胺(化合物15.8)。m/z:[M+H] +345.2。
利用化合物15.3的合成方法,将步骤2中的1-甲基哌嗪嗪替换为1-异丙基-哌嗪得到4-(4-异丙基哌嗪-1-基)-1-甲基-1H-吲哚-6-胺(化合物15.9)。m/z:[M+H] +273.0。
利用化合物15.3的合成方法,将步骤2中的1-甲基哌嗪替换为(1S,4S)-2-甲基-2,5-二氮杂双环[2.2.1]庚烷得到1-甲基-4-((1S,4S)-5-甲基-2,5-二氮杂双环[2.2.1]庚-2-基)-1H-吲哚-6-胺(化合物15.10)。m/z:[M+H] +257.2。
实施例28:化合物16.3的合成
Figure PCTCN2018101290-appb-000078
步骤1:室温下,将N-溴代丁二酰亚胺(2.12g,12mmol)加入到7-硝基喹啉(1.74g,10mmol)的醋酸(20mL)溶液中。然后反应体系在80℃下搅拌2小时。将反应液冷却至室温并静止2小时后出现白色沉淀,过滤,滤饼用石油醚洗涤,真空干燥得到化合物16.1(2.10g,产率:84%)为白色固体。
步骤2:将化合物16.1(2.1g,8.33mmol)溶于二甲基亚砜(20mL)中,依次加入1-甲基哌嗪(0.9g,9mmol),碳酸钾(2.2g,16mmol)和碘化亚铜(152mg,0.8mmol)。 反应体系用氮气置换3次后,在氮气保护下120℃下搅拌3小时,然后将反应液倒入到冰水中,水相用乙酸乙酯萃取(20mL×3),合并有机相,依次用水、饱和食盐水洗涤,减压浓缩,残留物经硅胶柱层析(二氯甲烷/甲醇=10/1)纯化得化合物16.2(540mg,产率:24%)为棕色固体。
步骤3:将化合物16.2(540mg,1.99mmol)和雷尼镍(60mg)加入于甲醇(15mL)中。反应体系用氢气置换3次。然后将反应体系在氢气氛下室温搅拌1小时。过滤,滤液浓缩得到3-(4-甲基哌嗪-1-基)喹啉-7-胺(化合物16.3,480mg,产率:100%)为棕色固体。
实施例29:化合物16.5的合成
Figure PCTCN2018101290-appb-000079
步骤1:将2-氯-6-硝基喹啉(300mg,1.44mmol)加入到1-甲基哌嗪(5mL)的乙醇(20mL)溶液中,反应体系加热回流搅拌16小时。将反应液冷却至室温,减压浓缩后倒入水中,过滤固体,固体真空干燥得化合物16.4(300mg,产率:77%)为黄色固体。
步骤2:将化合物16.4(100mg,0.367mmol),钯碳(30mg,10%)加入到甲醇(15mL)中,反应体系用氢气置换3次,然后在氢气氛下室温搅拌2小时。将反应液过滤,滤液减压浓缩得到2-(4-甲基哌嗪-1-基)喹啉-6-胺(化合物16.5,80mg,产率:91%)为棕色液体。
实施例30:化合物17.2的合成
Figure PCTCN2018101290-appb-000080
步骤1:向6-硝基吲哚(150mg,0.93mmol)的DMF(10.0mL)溶液中依次加入碳酸铯(910mg,2.79mmol)和3-碘氧杂环丁烷(205mg,1.11mmol),反应体系在70℃下搅拌3小时。然后冷却至室温并用乙酸乙酯(50mL)稀释,得到的混合物依次用水(20mL)、饱和食盐水(20mL)洗涤,有机相用无水硫酸钠干燥,过滤、减压浓缩,残留物用prep-TLC(石油醚/乙酸乙酯=2/1)纯化得到化合物17.1(73mg,产率:36%)为黄色固体。m/z:[M+H] +219.2。
步骤2:向化合物17.1(73mg,0.33mmol)的甲醇(5mL)溶液中加入雷尼镍(20mg),反应体系用氢气置换3次,然后在氢气氛中搅拌1小时。过滤、滤液减压浓缩得到1-(氧杂环丁烷-3-基)-1H-吲哚-6-胺(化合物17.2,60mg,产率:95%)为黄色固体。m/z:[M+H] +189.2。
实施例31:化合物17.3的合成
利用化合物17.2的合成方法,将步骤1中的3-碘氧杂环丁烷替换为碘甲烷得到1-甲 基-1H-吲哚-6-胺(化合物17.3)。m/z:[M+H] +147.2。
实施例32:化合物18.3的合成
Figure PCTCN2018101290-appb-000081
步骤1:将4-溴-6-硝基-1H-吲哚(200mg,0.83mmol)、环丙基硼酸(143mg,1.66mmol)、碳酸钠(176mg,1.66mmol)、醋酸铜(151mg,0.83mmol)和2,2-联吡啶(130mg,0.83mmol)的二氯甲烷(5mL)溶液在封管中,升温至100℃搅拌过夜。然后将反应体系冷却至室温,并用二氯甲烷稀释,有机相用水洗涤。分离有机相,浓缩,残留物用硅胶柱层析(石油醚/乙酸乙酯=5/1)纯化得到化合物18.1(215mg,产率:92%)为黄色固体。
步骤2&3:利用化合物15.3步骤2和步骤3的合成方法,用化合物18.1反应得到1-环丙基-4-(4-甲基哌嗪-1-基)-1H-吲哚-6-胺(化合物18.3)为棕色固体。
实施例33:化合物19.3的合成
Figure PCTCN2018101290-appb-000082
步骤1:将4-溴-6-硝基-1H-吲哚(300mg,1.24mmol)、溴乙醇(467mg,3.73mmol)和碳酸钾(176mg,1.66mmol)的DMF(10mL)混悬液在100℃下搅拌过夜。然后将反应体系冷却至室温,倒入冰水中,水相用乙酸乙酯萃取,有机相依次用水和饱和食盐水洗涤。分离有机相,浓缩,残留物用硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化得到化合物19.1(320mg,产率:90%)为黄色固体。
步骤2&3:利用化合物15.3步骤2和步骤3的合成方法,用化合物19.1反应得到2-(6-氨基-4-(4-甲基哌嗪-1-基)-1H-吲哚-1-基)乙醇(化合物19.3)为棕色固体。
实施例34:化合物20.3的合成
Figure PCTCN2018101290-appb-000083
步骤1:向4-溴-6-硝基-1H-吲哚(200mg,0.83mmol)和二羰基二叔丁酯(271mg,1.24mmol)的二氯甲烷(10mL)溶液中加入4-二甲氨基吡啶(10mg,0.083mmol),反应体系在室温下搅拌1小时。然后将反应液直接浓缩,残留物用硅胶柱层析(石油醚/乙酸乙酯=10/1)纯化得到化合物20.1(283mg,产率:100%)为黄色固体。
步骤2:将化合物20.1(140mg,0.41mmol)、醋酸钯(4.6mg,0.02mmol)、1-甲基哌嗪(41mg,0.41mmol)、碳酸铯(200mg,0.62mmol)和2-二环己基磷-2',6'-二异丙氧基-1,1'-联苯(19.3mg,0.04mmol)的1,4-二氧六环(10mL)混合物用氮气置换3次,然后将反应体系升温至110℃搅拌2小时,将反应体系冷却至室温后用硅藻土过滤,滤饼用乙酸乙酯淋洗,滤液浓缩,残留物用硅胶柱层析(乙酸乙酯)纯化得到化合物20.2(110mg,产率:74%)为棕色固体。m/z:[M+H] +361.2。
步骤3:利用化合物15.3步骤3的合成方法,用化合物20.2反应得到6-氨基-4-(4-甲基哌嗪-1-基)-1H-吲哚-1-羧酸叔丁酯(化合物20.3)为棕色固体。
实施例35:化合物21.5的合成
Figure PCTCN2018101290-appb-000084
步骤1:将铁粉(2.2g,39.2mmol)加入到化合物15.1(2g,7.84mmol)和氯化铵(629mg,11.8mmol)的醋酸(20mL)溶液中,得到的混合物回流搅拌1小时。将反应液热过滤,滤饼用醋酸洗涤,所得滤液浓缩,残留物用硅胶柱层析(石油醚/乙酸乙酯=5/1)纯化得到化合物21.1(1.1g,产率:62%)为棕色固体。m/z:[M+H] +225.0。
步骤2:将化合物21.1(1.05g,4.67mmol)加入到二碳酸二叔丁酯(10mL)中,得到的混合物升温至100℃搅拌1小时,减压浓缩除去溶剂,残留物用硅胶柱层析(石油醚/乙酸乙酯=10/1)纯化得到化合物21.2(1.3g,产率:86%)为白色固体。m/z:[M+H] +325.2。
步骤3:-78℃下,将正丁基锂的四氢呋喃溶液(2.5M,2.5mL,6.28mmol)滴加入到化合物21.2(680mg,2.09mmol)的无水四氢呋喃(20mL)溶液中,得到的混合物在-78℃的条件下搅拌1小时,然后将1-甲基-4-哌啶酮(284mg,2.50mmol)的无水四氢呋喃(1mL)溶液滴加入到上述反应液中,继续在此温度搅拌2小时,用饱和氯化铵的水溶液淬灭反应,得到的混合物用乙酸乙酯萃取,有机相浓缩,残留物用硅柱胶层析纯化(二氯甲烷/甲醇=10/1)得到化合物21.3(268mg,产率:36%)为棕色液体。m/z:[M+H] +360.0。
步骤4:将化合物21.3(268mg,0.75mmol)溶入到盐酸(6M,10mL)中,得到的混合物在100℃搅拌3小时,减压浓缩除去溶剂得到化合物21.4(207mg,产率:100%)为棕色固体。m/z:[M+H] +242.0。
步骤5:将化合物21.4(207mg,0.75mmol)和10%的钯/碳(100mg)的甲醇(15mL) 混合物用氢气置换3次。然后在氢气氛下室温搅拌1小时。溶液过滤,滤液减压浓缩,残留物溶于饱和的碳酸氢钠水溶液中,水相用二氯甲烷萃取,合并有机相并用无水硫酸钠干燥、减压浓缩,残留物用prep-TLC(二氯甲烷/甲醇=10/1)纯化所得1-甲基-4-(1-甲基哌啶-4-基)-1H-吲哚-6-胺(化合物21.5,35mg,产率:19%)为棕色固体。m/z:[M+H] +244.2。
实施例36:化合物1-4的合成
Figure PCTCN2018101290-appb-000085
步骤1:依次将化合物1-1(222mg,1.0mmol)、化合物1-2(215mg,1.0mmol)、碘化亚铜(191mg,1.0mmol)、无水碳酸钾(276mg,2.0mmol)和N,N-二甲基乙二胺(88mg,1mmol)加入到1,4-二氧六环(20mL)中,反应体系升温至100℃并搅拌过夜。然后将反应体系冷却至室温,将反应液过滤,滤液减压浓缩,残留物用硅胶柱层析纯化(石油醚/乙酸乙酯=1/1)得化合物1-3(120mg,产率:34%)为棕色固体。m/z:[M+H] +358.2。
步骤2:将间氯过氧苯甲酸(68mg,0.34mmol)加入到化合物1-3(120mg,0.34mmol)的二氯甲烷(10mL)溶液中,反应体系在室温下搅拌1小时,减压浓缩除去二氯甲烷得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-(甲基亚磺酰基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物1-4,125mg,产率:100%)为白色固体。m/z:[M+H] +374.2。
实施例37:化合物1-1-1的合成
Figure PCTCN2018101290-appb-000086
将化合物1-4(125mg,0.34mmol)、N,N-二异丙基乙胺(87mg,0.68mmol)和化合物1.3(68mg,0.34mmol)溶入到甲苯(10mL)中,反应体系升温至70℃搅拌过夜,然后减压浓缩除去溶剂,残留物用prep-HPLC(条件1)纯化得到2-烯丙基-6-((1-(2-(二甲基氨基)乙基)-1H-吲哚-5-基)氨基)-1-(6-(2-羟基丙-2-基)吡啶-2-基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物1-1-1,97.6mg,产率:46%)为白色固体。m/z:[M+H] +513.4; 1H NMR(400MHz,CD 3OD):δ8.81(s,1H),7.99(br.s,1H),7.96(t,J=8.0Hz,2H),7.85(d,J=8.0Hz,1H),7.64(dd,J=0.8,8.0Hz,1H),7.40(d,J=8.8Hz,1H),7.23(dd,J=2.4,8.8Hz,1H),7.29(d,J=3.2Hz,1H),6.44(d,J=2.8Hz,1H),5.69-5.76(m,1H),5.05(dd,J=1.2,10.4Hz,1H),4.93(dd,J=1.2,17.2Hz,1H),4.85(d,J=6.0Hz,2H),4.33(t,J=7.2Hz,2H), 2.78(t,J=7.2Hz,2H),2.33(s,6H),1.58(s,6H)。
实施例38:化合物1-1-2的合成
Figure PCTCN2018101290-appb-000087
利用化合物1-1-1的合成方法,将化合物1.3替换为7.6得到化合物2-1。
把三氟乙酸(2mL)滴加到化合物2-1(130mg,0.21mmol)的二氯甲烷(10mL)中,反应体系在室温下搅拌2小时,反应液浓缩,残留物用prep-HPLC(条件1)纯化得到2-烯丙基-6-((1-((1-氨基环丙基)甲基)-1H-吲哚-5-基)氨基)-1-(6-(2-羟基丙-2-基)吡啶-2-基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物1-1-2,46.7mg,产率:36%)为黄色固体。m/z:[M+H] +510.8; 1H NMR(400MHz,CD 3OD):δ8.71(s,1H),7.95(br.s,1H),7.85(t,J=8.0Hz,1H),7.72(d,J=7.6Hz,1H),7.54(dd,J=0.8,7.6Hz,1H),7.35(d,J=8.8Hz,1H),7.28(dd,J=6.8,8.8Hz,1H),7.22(d,J=3.2Hz,1H),6.47(d,J=3.2Hz,1H),5.65-5.57(m,1H),4.95(dd,J=1.2,10.6Hz,1H),4.79(dd,J=1.2,17.2Hz,1H),4.84-4.72(m,2H),4.38(s,2H),1.48(s,6H),1.07-0.95(m,4H)。
实施例39:化合物1-1-3的合成
Figure PCTCN2018101290-appb-000088
利用化合物1-1-1的合成方法,将化合物1.3替换为7.8得到化合物2-2。
利用化合物1-1-2的合成方法,用化合物2-2反应得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-((1-(甲基氨基)环丙基)甲基)-1H-吲哚-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物1-1-3)。m/z:[M+H] +525.4; 1H NMR(400MHz,DMSO-d 6):δ10.31(s,1H),8.86(s,1H),8.44(br.s,2H),8.14(s,1H),8.01(t,J=8.0Hz,1H),7.83(d,J=7.2Hz,1H),7.62(d,J=7.2Hz,1H),7.56(d,J=8.8Hz,1H),7.46(d,J=2.8Hz,1H),7.42(d,J=9.6Hz,1H),6.54(d,J=3.2Hz,1H),5.72-5.62(m,1H),5.34-5.31(m,1H),5.00(dd,J=1.2,10.8Hz,1H),4.83(dd,J=1.2,17.2Hz,1H),4.70(d,J=5.2Hz,2H),2.53(s,3H),1.46(s,3H),1.10-0.95(m,4H)。
实施例40:化合物1-2-1~1-2-3的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为1.7、1.8或1.9得到2-烯丙基-6-((1-(2-(氮杂环丁烷-1-基)乙基)-1H-吲哚-5-基)氨基)-1-(6-(2-羟基丙-2-基)吡啶-2-基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(1-2-1)、2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(2-(吡咯烷-1-基)乙基)-1H-吲哚-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(1-2-2)和2-烯丙基 -1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(2-(哌啶-1-基)乙基)-1H-吲哚-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(1-2-3):
Figure PCTCN2018101290-appb-000089
Figure PCTCN2018101290-appb-000090
实施例41:化合物1-3-1~1-3-4的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为2.5、2.6、2.7或2.9得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(1-甲基哌啶-4-基)-1H-吲哚-5-基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(1-3-1)、2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(1-甲基吡咯烷基-3-基)-1H-吲哚-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(1-3-2)、2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(1-甲基氮杂环丁烷-3-基)-1H-吲哚-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(1-3-3)和2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(1-甲基哌啶-3-基)-1H-吲哚-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(1-3-4):
Figure PCTCN2018101290-appb-000091
Figure PCTCN2018101290-appb-000092
实施例42:化合物1-4-1~1-4-4的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为2.8、4.2、5.2或11.2得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(1-异丙基哌啶-4-基)-1H-吲哚-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(1-4-1)、6-((1-(1-乙酰基哌啶-4-基)-1H-吲哚-5-基)氨基)-2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(1-4-2)、2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(1-(2,2,2-三氟乙基)哌啶-4-基)-1H-吲哚-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(1-4-3)和2-烯丙基-6-((1-(1-(2-羟基乙基)哌啶-4-基)-1H-吲哚-5-基)氨基)-1-(6-(2-羟基丙-2-基)吡啶-2-基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(1-4-4):
Figure PCTCN2018101290-appb-000093
Figure PCTCN2018101290-appb-000094
实施例43:化合物1-5-1的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为2.15得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(四氢-2H-吡喃-4-基)-1H-吲哚-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物1-5-1):
Figure PCTCN2018101290-appb-000095
m/z:[M+H] +526.4; 1H NMR(400MHz,DMSO-d 6):δ10.22(s,1H),8.84(s,1H),8.07(s,1H),8.02(t,J=8.0Hz,1H),7.83(d,J=8.0Hz,1H),7.62(d,J=7.6Hz,1H),7.53(d,J=8.8Hz,1H),7.52(d,J=2.8Hz,1H),7.38(d,J=8.0Hz,1H),6.45(d,J=3.2Hz,1H),5.72-5.62(m,1H),5.00(dd,J=1.2,10.0Hz,1H),4.83(dd,J=1.2,17.2Hz,1H),4.70(d,J=5.2Hz,2H),4.65-4.58(m,1H),4.01(dd,J=4.0,11.2Hz,2H),3.58(dd,J=9.6,12.0Hz,2H),2.06-1.96(m,4H),1.49(s,6H)。
实施例44:化合物1-6-1的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为8.3得到2-烯丙基-6-((2-((二甲基氨基)甲基)-1H-吲哚-5-基)氨基)-1-(6-(2-羟基丙-2-基)吡啶-2-基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物1-6-1):
Figure PCTCN2018101290-appb-000096
m/z:[M+H] +499.4; 1H NMR(400MHz,DMSO-d 6):δ11.23(s,1H),10.26(s,1H),9.72(s,1H),8.81(s,1H),8.10(br.s,1H),7.97(t,J=7.6Hz,1H),7.83(d,J=8.0Hz,1H),7.66(d,J=7.6Hz,1H),7.41(s,2H),6.65(d,J=2.0Hz,1H),5.72-5.62(m,1H),5.01(dd,J=1.2,10.0Hz,1H),4.83(dd,J=1.2,17.2Hz,1H),4.70(d,J=5.2Hz,2H),4.42(d,J=4.0Hz,2H),2.80(d,J=4.4Hz,6H),1.51(s,6H)。
实施例45:化合物1-7-1~1-7-3的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为14.2、14.3、或14.4得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(吡啶-2-基)-1H-吲哚-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(1-7-1)、2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(吡啶-3-基)-1H-吲哚-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(1-7-2)和2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(吡啶-4-基)-1H-吲哚-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(1-7-3):
Figure PCTCN2018101290-appb-000097
Figure PCTCN2018101290-appb-000098
Figure PCTCN2018101290-appb-000099
实施例46:化合物2-1-1的合成
Figure PCTCN2018101290-appb-000100
步骤1:室温条件下,向化合物1-1-1(210mg,0.41mmol)和甲酸氨(52mg,0.82mmol)的1,4-二氧六环(10mL)溶液中加入二茂铁二氯化钯(34mg,0.041mmol),将反应体系用氮气置换3次,然后将反应体系缓慢升至100℃并搅拌过夜。反应液冷至室温,过滤,滤饼用甲醇洗涤,所得滤液浓缩,残留物用硅胶柱层析纯化(二氯甲烷/甲醇=10/1)得化合物2-1(110mg,产率:57%)为棕色固体。
步骤2:冰浴条件下,向化合物2-1(110mg,0.233mmol)的DMF(5mL)溶液中加入钠氢(19mg,0.47mmol,60%),然后将反应体系缓慢升至室温并搅拌0.5小时,加入环丙基溴甲烷(94mg,0.7mmol),混合物室温条件下继续搅拌过夜。然后再将反应液冷至0℃,滴加冰水淬灭反应,溶液用二氯甲烷萃取(5mL×3),合并有机相,并依次用水、饱和食盐水洗涤,有机相浓缩,残留物用prep-HPLC(条件1)纯化得2-(环丙基甲基)-6-((1-(2-(二甲基氨基)乙基)-1H-吲哚-5-基)氨基)-1-(6-(2-羟基丙-2-基)吡啶-2-基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物2-1-1,24.3mg,产率:20%)为白色固体。m/z:[M+H] +526.9; 1H NMR(400MHz,CD 3OD):δ8.10-9.00(b,2H),7.59-7.66(m,2H),7.20-7.44(m,3H),6.66(s,1H),5.10-5.15(m,2H),4.65-4.69(m,2H),4.03-4.08(m,2H),3.69-3.72(t,J=6.8Hz,2H),3.01(s,6H),1.50-1.60(b,6H),0.50-0.54(m,2H), 0.25-0.27(m,2H)。
实施例47:化合物3-1-1的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为3.1得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(1-甲基氮杂环丁烷-3-基)-1H-苯并[d]咪唑-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物3-1-1):
Figure PCTCN2018101290-appb-000101
m/z:[M+H] +512.0; 1H NMR(400MHz,CD 3OD):δ9.89(s,1H),8.94(s,1H),8.52(s,1H),8.08-8.12(m,1H),7.91(s,1H),7.80-7.82(d,J=8.0Hz,1H),7.69-7.71(d,J=7.6Hz,1H),5.80-6.10(m,1H),5.71-5.78(m,1H),5.00-5.08(m,4H),4.78-4.84(m,4H),3.32(s,3H),1.60(s,6H)。
实施例48:化合物4-1-1的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为6.6得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((2-甲基-1,2,3,4-四氢吡嗪并[1,2-a]吲哚-8-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物4-1-1):
Figure PCTCN2018101290-appb-000102
m/z:[M+H] +511.1; 1H NMR(400MHz,CDCl 3):δ8.84(s,1H),7.97-7.80(m,3H),7.60(br.s,1H),7.36-7.30(m,1H),7.25-7.17(m,2H),6.18(s,1H),5.76-5.65(m,1H),5.07-5.02(m,1H),4.98-4.91(m,1H),4.76(d,J=6.4Hz,2H),4.12(t,J=5.6Hz,2H),3.97(br.s,1H),3.80(s,2H),2.95(t,J=5.6Hz,2H),2.53(s,3H),1.58(s,6H)。
实施例49:化合物5-1-1的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为9.1得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((6-(4-甲基哌嗪-1-基)萘-2-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物5-1-1):
Figure PCTCN2018101290-appb-000103
m/z:[M+H] +551.4; 1H NMR(400MHz,CDCl 3):δ8.86(s,1H),8.29(s,1H),8.03(t,J=8.0Hz,1H),7.86(d,J=8.0Hz,1H),7.69(dd,J=4.4,8.4Hz,2H),7.68(d,J=8.4Hz,1H),7.59(d,J=8.8Hz,1H),7.36(dd,J=2.4,9.2Hz,1H),7.25(d,J=2.0Hz,1H),5.76-5.69(m, 1H),5.03(d,J=10.4Hz,1H),4.92(dd,J=1.2,17.2Hz,1H),4.89-4.85(m,2H),3.97(d,J=12.8Hz,2H),3.67(d,J=11.2Hz,2H),3.38-3.29(m,2H),3.15-3.09(m,2H),3.01(s,3H),1.58(s,3H)。
实施例50:化合物6-1-1的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为10.4得到6-((2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-3-氧-2,3-二氢-1H-吡唑并[3,4-d]嘧啶-6-基)氨基)-3-(4-甲基哌嗪-1-基)苯并[b]噻吩-2-羧酸乙酯三氟乙酸盐(化合物6-1-1):
Figure PCTCN2018101290-appb-000104
m/z:[M+H] +628.9; 1H NMR(400MHz,CD 3OD):δ8.89(s,1H),8.53(s,1H),8.08(t,J=8.0Hz,1H),7.84(d,J=1.6Hz,1H),7.82(d,J=3.2Hz,1H),7.22(d,J=7.6Hz,1H),7.45(dd,J=3.2,9.2Hz,1H),5.77-5.70(m,1H),5.04(dd,J=1.2,10.0Hz,1H),4.92(dd,J=1.2,17.2Hz,1H),4.88-4.83(m,2H),4.35(q,J=7.2Hz,2H),3.48-3.45(m,4H),2.70(br.s,4H),2.41(s,3H),1.58(s,6H),1.40(t,J=7.2Hz,3H)。
实施例51:化合物6-1-2的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为10.7得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((3-(4-甲基哌嗪-1-基)苯并[b]噻吩-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物6-1-2):
Figure PCTCN2018101290-appb-000105
m/z:[M+H] +557.2; 1H NMR(400MHz,CD 3OD):δ8.87(s,1H),8.54(s,1H),8.06(t,J=8.0Hz,1H),7.83(d,J=8.0Hz,1H),7.68(d,J=5.2Hz,1H),7.67(d,J=6.4Hz,1H),7.44(d,J=8.0Hz,1H),6.75(s,1H),5.74-5.69(m,1H),5.04(dd,J=1.6,10.0Hz,1H),4.91(dd,J=1.6,17.6Hz,1H),4.58(s,2H),3.18(s,4H),2.73(s,4H),2.40(s,3H),1.58(s,6H)。
实施例52:化合物7-1-1的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为12.2得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((3-(1-甲基哌啶-4-基)-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物7-1-1):
Figure PCTCN2018101290-appb-000106
m/z:[M+H] +538.9; 1H NMR(400MHz,DMSO-d 6):δ10.85(s,1H),10.18(s,1H),8.87(s,1H),8.06-8.01(m,1H),7.91-7.82(m,3H),7.60(d,J=7.6Hz,1H),7.54(d,J=8.8Hz,1H),7.27(d,J=8.0Hz,1H),7.11(d,J=2.0Hz,1H),5.73-5.63(m,1H),5.01(d,J=10.4Hz,1H),4.84(d,J=17.2Hz,1H),4.70(d,J=6.0Hz,2H),3.21-3.12(m,2H),3.09-2.98(m,1H),2.84(d,J=4.8Hz,3H),2.21-2.12(m,2H),1.93-1.81(m,1H),1.48(s,6H)。
实施例53:化合物7-1-2的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为13.4得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((3-(4-甲基哌嗪-1-基)-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物7-1-2):
Figure PCTCN2018101290-appb-000107
m/z:[M+H] +540.2。
实施例54:化合物7-1-3的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为13.7得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-甲基-3-(4-甲基哌嗪-1-基)-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物7-1-3):
Figure PCTCN2018101290-appb-000108
m/z:[M+H] +554.4; 1H NMR(400MHz,CD 3OD):δ8.84(s,1H),7.94(t,J=8.0Hz,1H),7.82(s,1H),7.81(s,1H),7.67(d,J=8.0Hz,1H),7.51(d,J=8.4Hz,1H),7.17(d,J=1.6Hz,1H),6.79(s,1H),5.77-5.68(m,1H),5.04(d,J=10.0Hz,1H),4.84(d,J=16.0Hz,1H),4.80(d,J=6.0Hz,2H),3.66(s,3H),3.12(s,4H),2.71(s,4H),2.39(s,3H),1.58(s,6H)。
实施例55:化合物7-1-4的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为12.3得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-甲基-3-(1-甲基哌啶-4-基)-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物7-1-4):
Figure PCTCN2018101290-appb-000109
m/z:[M+H] +553.4; 1H NMR(400MHz,DMSO-d 6):δ10.30(br.s,1H),9.33(br.s,1H),8.88(s,1H),8.05-7.74(m,4H),7.69-7.50(m,2H),7.23(d,J=9.2Hz,1H),7.09(s,1H),5.73-5.60(m,2H),4.66(d,J=6.0Hz,2H),3.68(s,3H),3.20-2.94(m,5H),2.84(d,J=4.8Hz,3H),2.22-1.76(m,4H),1.46(s,6H)。
实施例56:化合物7-1-5的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为17.2得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(氧杂环丁烷-3-基)-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物7-1-5):
Figure PCTCN2018101290-appb-000110
m/z:[M+H] +498.2; 1H NMR(400MHz,CDCl 3):δ8.89(s,1H),7.91-7.85(m,2H),7.77-7.75(m,2H),7.59(d,J=8.4Hz,1H),7.45(d,J=3.2Hz,1H),7.39(d,J=7.6Hz,1H),6.60(d,J=3.2Hz,1H),5.73-5.68(m,1H),5.51-5.46(m,1H),5.13-5.08(m,2H),5.05-5.02(m,2H),4.93(dd,J=17.2,1.2Hz,2H),4.74(d,J=6.0Hz,2H),1.59(s,6H)。
实施例57:化合物7-1-6的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为17.3得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-甲基-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物7-1-6):
Figure PCTCN2018101290-appb-000111
m/z:[M+H] +456.2; 1H NMR(400MHz,CDCl 3):δ8.88(s,1H),7.82(d,J=3.6Hz,2H),7.73(s,1H),7.57(d,J=8.4Hz,1H),7.35-7.32(m,1H),7.14(d,J=7.6Hz,1H),7.05(d,J=3.2Hz,1H),6.48(d,J=2.8Hz,1H),5.70(m,1H),5.04(dd,J=10.4,0.8Hz,1H),4.93(dd,J=16.8,1.2Hz,1H),4.75(d,J=6.0Hz,2H),3.76(s,3H),1.58(s,6H)。
实施例58:化合物7-2-1的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为15.3得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-甲基-4-(4-甲基哌嗪-1-基)-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物7-2-1):
Figure PCTCN2018101290-appb-000112
m/z:[M+H] +554.4; 1H NMR(400MHz,CDCl 3):δ8.80(s,1H),8.25(d,J=2.0Hz,1H),8.22(s,1H),7.75(t,J=7.6Hz,1H),7.68(d,J=8.0Hz,1H),7.30(d,J=3.6Hz,1H),7.27(d,J=7.6Hz,1H),6.39(d,J=3.6Hz,1H),5.67-5.60(m,1H),4.97(d,J=10.4Hz,1H),4.87(d,J=18.0Hz,1H),4.75-4.71(m,1H),4.69(d,J=6.4Hz,1H),3.84(s,1H),2.96(d,J=11.6Hz,2H),2.31(s,3H),2.23-2.16(m,2H),2.04-2.01(m,4H),1.51(s,6H)。
化合物7-2-1prep-HPLC纯化过程使用纯化条件2或3,得到7-2-1相应的游离碱和盐酸盐。
实施例59:化合物7-2-2的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为15.4得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-甲基-4-(吡咯烷-1-基)-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物7-2-2):
Figure PCTCN2018101290-appb-000113
m/z:[M+H] +525.2; 1H NMR(400MHz,DMSO-d 6):δ10.07(br.s,1H),8.85(s,1H),8.00-7.92(m,1H),7.86-7.77(m,1H),7.63(d,J=7.6Hz,1H),7.33(s,1H),7.05(d,J=3.2Hz,1H),6.53(d,J=2.8Hz,1H),6.28(s,1H),5.72-5.59(m,1H),5.36(s,1H),5.02-4.96(m,1H),4.84-4.75(m,1H),4.64(d,J=7.2Hz,1H),3.64(s,3H),3.50-3.41(m,4H),2.02-1.89(m,4H),1.45(s,6H)。
实施例60:化合物7-2-3的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为15.5得到2-烯丙基-6-((4-(二甲基氨基)-1-甲基-1H-吲哚-6-基)氨基)-1-(6-(2-羟基丙-2-基)吡啶-2-基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物7-2-3):
Figure PCTCN2018101290-appb-000114
m/z:[M+H] +499.2; 1H NMR(400MHz,DMSO-d 6):δ10.29(br.s,1H),9.52(s,1H),7.99-7.95(m,1H),7.82-7.75(m,2H),7.63(d,J=7.8Hz,1H),7.30(br.s,1H),6.97-6.96(m,1H),6.57(s,1H),5.69-5.61(m,1H),5.05-4.98(m,1H),4.83-4.78(m,1H),4.66-4.62(m,2H),3.72(s,3H),3.04(s,6H),1.45(s,6H)。
实施例61:化合物7-2-4的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为18.3得到2-烯丙基-6-((1-环丙基-4-(4-甲基哌嗪-1-基)-1H-吲哚-6-基)氨基)-1-(6-(2-羟基丙-2-基)吡啶-2-基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物7-2-4):
Figure PCTCN2018101290-appb-000115
m/z:[M+H] +580.3; 1H NMR(400MHz,CDCl 3):δ8.89(s,1H),7.75-7.79(m,2H),7.48-7.54(m,2H),7.35(d,J=8.0Hz,1H),7.05-7.06(m,1H),6.68(s,1H),6.40-6.41(m,1H),5.69-5.75(m,1H),5.07(d,J=10.0Hz,1H),4.95(d,J=16.8Hz,1H),4.74(d,J=6.0Hz,1H),4.00(br.s,1H),3.22-3.30(m,4H),2.66(m,4H),2.41(s,3H),1.59(s,6H),0.98(s,4H)。
实施例62:化合物7-2-5的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为19.3得到2-烯丙基-6-((1-(2-羟基乙基)-4-(4-甲基哌嗪-1-基)-1H-吲哚-6-基)氨基)-1-(6-(2-羟基丙-2-基)吡啶-2-基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物7-2-5):
Figure PCTCN2018101290-appb-000116
m/z:[M+H] +584.3; 1H NMR(400MHz,CDCl 3):δ8.92(s,1H),8.40(br.s,1H),7.86(t,J=8.0Hz,1H),7.74(d,J=8.4Hz,1H),7.39(d,J=6.8Hz,1H),7.10(s,1H),6.59(s,1H),6.48(s,1H),5.66-5.74(m,1H),5.05(d,J=10.0Hz,1H),4.93(d,J=16.8Hz,1H),4.73(d,J=6.0Hz,1H),4.21-4.24(m,3H),3.95(t,J=4.8Hz,2H),3.17(s,4H),2.61-2.65(m,4H),2.40(s,3H),1.62(s,6H)。
实施例63:化合物7-2-6的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为15.6得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-甲基-4-吗啉基-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物7-2-6):
Figure PCTCN2018101290-appb-000117
m/z:[M+H] +541.2; 1H NMR(400MHz,CD 3OD):δ8.78(s,1H),7.89(t,J=8.0Hz,1H),7.73(d,J=7.6Hz,1H),7.63(d,J=7.7Hz,1H),7.52(s,1H),7.02(d,J=3.2Hz,1H),6.63(s, 1H),6.36(d,J=3.2Hz,1H),5.72-5.63(m,1H),5.00(d,J=10.0Hz,1H),4.84(s,1H),4.74(d,J=6.0Hz,2H),3.85-3.83(m,4H),3.65(s,3H),3.13-3.05(m,4H),1.55(s,6H)。
实施例64:化合物7-2-7和7-2-8的合成
Figure PCTCN2018101290-appb-000118
利用化合物1-1-1的合成方法,将化合物1.3替换为15.7得到4-(6-((2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-3-氧代-2,3-二氢-1H-吡唑并[3,4-d]嘧啶-6-基)氨基)-1-甲基-1H-吲哚-4-基)哌嗪-1-羧酸叔丁酯(化合物7-2-7)。
将化合物7-2-7(125mg,0.19mmol)溶解于二氯甲烷和三氟乙酸(4mL,1:1)中,反应体系在室温下搅拌1小时后,直接浓缩除去溶剂,残留物用prep-HPLC(方法1)纯化得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-甲基-4-(哌嗪-1-基)-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物7-2-8,30mg,产率:36%)为黄色固体。m/z:[M+H] +540.2; 1H NMR(400MHz,DMSO-d 6):δ10.20(s,1H),8.88(s,1H),8.79(s,1H),8.04-7.94(m,1H),7.85-7.60(m,1H),7.24(d,J=3.2Hz,1H),6.71(s,1H),6.42(d,J=2.8Hz,1H),5.72-5.60(m,1H),5.02-4.96(m,1H),4.85-4.77(m,1H),4.68-4.62(m,2H),3.70(s,3H),3.37-3.24(m,8H),1.45(s,6H)。
实施例65:化合物7-2-9和7-2-10的合成
Figure PCTCN2018101290-appb-000119
利用化合物1-1-1的合成方法,将化合物1.3替换为20.3得到6-((2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-3-氧代-2,3-二氢-1H-吡唑并[3,4-d]嘧啶-6-基)氨基)-4-(4-甲基哌嗪-1-基)-1H-吲哚-1-羧酸叔丁酯(化合物7-2-9)。m/z:[M+H] +640.4; 1H NMR(400MHz,CDCl 3):δ9.04(s,1H),8.28(br.s,1H),7.80-7.89(m,3H),7.51-7.52(m,1H),7.34(d,J=7.6Hz,1H),6.79(s,1H),6.56(s,1H),5.68-5.78(m,1H),5.07(d,J=10.8Hz,1H),4.96(d,J=16.8Hz,1H),4.75-4.80(m,2H),4.10-4.16(b,1H),3.15(s,4H),2.66(s,4H),2.43(s,3H),1.61(s,9H),1.57(s,6H)。
利用化合物7-2-8的合成方法,将化合物7-2-7替换为7-2-9得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((4-(4-甲基哌嗪-1-基)-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物7-2-10)。m/z:[M+H] +540.2; 1H NMR(400MHz,CDCl 3):δ8.83(s,1H),7.99(t,J=8.0Hz,1H),7.79-7.82(m,1H),7.64-7.74(m,2H),7.23-7.27(m,1H),6.68(br.s,1H),6.48(s,1H),5.68-5.76(m,1H),5.05(d,J=10.4Hz,1H),4.91-4.94(m, 1H),4.82-4.86(m,2H),3.83-3.86(m,2H),3.65(d,J=12.0Hz,2H),3.42-3.47(m,2H),3.03-3.15(m,5H),1.58(s,6H)。
实施例66:化合物7-2-11和7-2-12的合成
Figure PCTCN2018101290-appb-000120
利用化合物1-1-1的合成方法,将化合物1.3替换为15.8得到(S)-4-(6-((2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-3-氧代-2,3-二氢-1H-吡唑并[3,4-d]嘧啶-6-基)氨基)-1-甲基-1H-吲哚-4-基)-2-甲基哌嗪-1-羧酸叔丁酯(化合物7-2-11)。m/z:[M+H] +654.4。
利用化合物7-2-8的合成方法,将化合物7-2-7替换为7-2-11得到(S)-2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-甲基-4-(3-甲基哌嗪-1-基)-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物7-2-12)。m/z:[M+H] +554.2; 1H NMR(400MHz,CDCl 3):δ8.86(s,1H),7.79(d,J=2.2Hz,2H),7.44(s,1H),7.34(t,J=4.0Hz,1H),6.98(d,J=3.0Hz,1H),6.48(s,1H),6.45(d,J=2.8Hz,1H),5.73-5.64(m,1H),5.03(d,J=10.0Hz,1H),4.92(d,J=17.2Hz,1H),4.74(d,J=6.0Hz,2H),3.72(s,3H),3.55(d,J=11.2Hz,2H),3.20-3.09(m,3H),2.76-2.70(m,1H),2.42(t,J=10.8Hz,1H),1.57(s,6H),1.12(d,J=6.4Hz,3H)。
实施例67:化合物7-2-13的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为15.9得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((4-(4-异丙基哌嗪-1-基)-1-甲基-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物7-2-13):
Figure PCTCN2018101290-appb-000121
m/z:[M+H] +582.0; 1H NMR(400MHz,CD 3OD):δ8.90-8.73(m,1H),8.02-7.81(m,1H),7.80-7.69(m,1H),7.68-7.45(m,2H),7.08-6.96(m,1H),6.67-6.54(m,1H),6.43-6.30(m,1H),5.80-5.57(m,1H),5.11-4.99(m,1H),4.79-4.69(m,2H),3.74-3.56(m,3H),3.16(s,4H),2.88-2.60(m,5H),1.56(s,6H),1.20-1.03(m,6H)。
实施例68:化合物7-2-14的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为15.10得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-甲基-4-((1S,4S)-5-甲基-2,5-二氮杂双环[2.2.1]庚-2-基)-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物7-2-14):
Figure PCTCN2018101290-appb-000122
m/z:[M+H] +566.3; 1H NMR(400MHz,CDCl 3):δ8.86(s,1H),7.81(d,J=3.6Hz,2H),7.35(t,J=4.0Hz,1H),7.20(s,1H),6.90(d,J=3.2Hz,1H),6.51(d,J=2.8Hz,1H),6.08(s,1H),5.73-5.65(m,1H),5.03(d,J=10.0Hz,1H),4.93(d,J=16.8Hz,1H),4.78-4.68(m,2H),4.33(s,1H),3.77-3.72(m,2H),3.69(s,3H),3.59(s,1H),3.03(d,J=8.4Hz,1H),2.86(d,J=9.6Hz,1H),2.41(s,3H),2.06-1.98(m,2H),1.57(s,6H)。
实施例69:化合物7-3-1的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为21.5得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-甲基-4-(1-甲基哌啶-4-基)-1H-吲哚-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物7-3-1):
Figure PCTCN2018101290-appb-000123
m/z:[M+H] +553.0。
实施例70:化合物8-1-1的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为2.10得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(1-异丙基哌啶-4-基)-1H-吡咯并[2,3-b]吡啶-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物8-1-1):
Figure PCTCN2018101290-appb-000124
m/z:[M+H] +568.4; 1H NMR(400MHz,CDCl 3):δ8.86(s,1H),8.31(d,J=2.0Hz,1H),8.29(s,1H),7.83(t,J=8.0Hz,1H),7.55(d,J=8.0Hz,1H),7.41(d,J=3.6Hz,1H),7.34(d,J=7.6Hz,1H),6.46(d,J=3.2Hz,1H),5.74-5.67(m,1H),5.04(d,J=10.4Hz,1H),4.94(d,J=17.2Hz,1H),4.81-4.77(m,1H),4.76(d,J=6.4Hz,2H),3.91(s,1H),3.09(d,J=10.4Hz,2H),2.87(s,1H),2.49(s,2H),2.12(s,4H),1.59(d,J=10.4Hz,6H),1.13(d,J=6.4Hz,6H)。
实施例71:化合物8-1-2的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为2.11得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(1-甲基哌啶-4-基)-1H-吡咯并[2,3-b]吡啶-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物8-1-2):
Figure PCTCN2018101290-appb-000125
m/z:[M+H] +540.3; 1H NMR(400MHz,CDCl 3):δ8.80(s,1H),8.25(d,J=2.0Hz,1H),8.22(s,1H),7.75(t,J=7.6Hz,1H),7.68(d,J=8.0Hz,1H),7.30(d,J=3.6Hz,1H),7.27(d,J=7.6Hz,1H),6.39(d,J=3.6Hz,1H),5.67-5.60(m,1H),4.97(d,J=10.4Hz,1H),4.87(dd,J=0.8,17.2Hz,1H),4.75-4.70(m,1H),4.69(d,J=6.4Hz,2H),3.84(s,1H),2.96(d,J=10.6Hz,2H),2.31(s,3H),2.23-2.16(m,2H),2.04-2.01(m,4H),1.53(s,6H)。
实施例72:化合物8-2-1的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为14.7得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(吡啶-3-基)-1H-吡咯并[2,3-b]吡啶-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物8-2-1):
Figure PCTCN2018101290-appb-000126
m/z:[M+H] +520.2; 1H NMR(400MHz,CD 3OD):δ9.04(d,J=2.4Hz,1H),8.89(s,1H),8.60(dd,J=1.6,4.8Hz,1H),8.41(s,2H),8.29(dd,J=1.2,8.4Hz,1H),7.87(t,J=7.6Hz,1H),7.75(dd,J=4.0,8.4Hz,1H),7.59(d,J=4.0Hz,1H),7.51(dd,J=4.8,8.8Hz,1H),7.37(dd,J=4.0,7.6Hz,1H),6.68(d,J=3.6Hz,1H),5.76-5.66(m,1H),5.05(dd,J=1.2,10.0Hz,1H),4.95(dd,J=1.6,17.2Hz,1H),4.76(d,J=6.0Hz,2H),1.58(d,J=8.8Hz,6H)。
实施例73:化合物8-2-2的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为14.8得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((1-(吡啶-2-基)-1H-吡咯并[2,3-b]吡啶-5-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(化合物8-2-2):
Figure PCTCN2018101290-appb-000127
m/z:[M+H] +520.2; 1H NMR(400MHz,CD 3OD):δ8.87(s,1H),8.81(d,J=8.4Hz,1H), 8.53-8.46(m,3H),8.34(d,J=4.0Hz,1H),8.04-7.94(m,2H),7.78(d,J=7.6Hz,1H),7.66(d,J=8.0Hz,1H),7.30-7.26(m,1H),6.69(d,J=4.0Hz,1H),5.79-5.66(m,1H),5.07-5.02(m,2H),4.87-4.77(m,2H),1.57(s,6H)。
实施例74:化合物9-1-1的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为16.3得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((3-(4-甲基哌嗪-1-基)喹啉-7-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物9-1-1):
Figure PCTCN2018101290-appb-000128
m/z:[M+H] +552.3; 1H NMR(400MHz,DMSO-d 6):δ10.52(br.s,1H),8.95(s,1H),8.86(s,1H),8.48(s,1H),8.09-8.03(m,1H),7.89(d,J=8.4Hz,1H),7.82-7.70(m,2H),7.64(d,J=7.6Hz,1H),7.50(s,1H),5.77-5.62(m,1H),5.35(s,1H),5.04-4.80(m,2H),4.73(d,J=5.2Hz,2H),3.30-3.25(m,4H),2.58-2.51(m,4H),2.26(s,3H),1.47(s,6H)。
实施例75:化合物9-2-1的合成
利用化合物1-1-1的合成方法,将化合物1.3替换为16.5得到2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((2-(4-甲基哌嗪-1-基)喹啉-6-基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮三氟乙酸盐(化合物9-2-1):
Figure PCTCN2018101290-appb-000129
m/z:[M+H] +552.4; 1H NMR(400MHz,CD 3OD):δ10.50(br.s,1H),9.85(br.s,1H),8.93(s,1H),8.30(br.s,1H),8.00-8.10(m,2H),7.82-7.89(m,2H),7.63(t,J=7.2Hz,2H),7.36(d,J=9.2Hz,1H),5.62-5.74(m,1H),5.00(dd,J=10.4,1.6Hz,1H),4.83(dd,J=17.2,1.2Hz,1H),4.55-4.75(m,4H),3.55(s,3H),3.20-3.32(m,2H),3.07-3.19(m,2H),2.87(s,3H),1.46(s,6H)。
生物测试实施例
实施例1:Wee1的酶活性检测
本发明中,运用ATP-Glo Max发光检测激酶试剂盒(Promega)进行Wee1酶催化试验。通过定量检测激酶反应后溶液中保留的ATP数量来评价激酶活性。试验中发光信号与ATP数量呈正比,与激酶活性呈反比。试验中化合物浓度范围为0.5nM~30μM。将化合物用10%DMSO溶解,取5μL的溶液加入到50μL的反应中,最终反应中DMSO的浓度为1%。30℃下反应50分钟。50μL反应混合液中包含40mM三羟甲基氨基甲烷,pH7.4,10mM MgCl 2,0.1mg/ml BSA,2mM DTT,0.1mg/ml Poly(Lys,Tyr)底物,10μM  ATP及Wee1。酶促反应后,加入50μL ATP-Glo Max发光检测激酶试验溶液(Promega),并在室温下孵化15分钟。使用酶标仪测量发光信号。在部分试验中,加入已知的Wee1抑制剂作为阳性对照。运用Graphpad软件分析发光数据。将Wee1缺失情况下的发光强度和Wee1存在情况下的发光强度差定义为100%活性(Lut–Luc)。利用化合物存在情况下的发光强度信号(Lu),%活性按照以下方法计算:
%活性={(Lut–Lu)/(Lut–Luc)}×100%,其中Lu=化合物的发光强度:
运用非线性回归分析绘制%活性值与对应系列化合物浓度量效曲线,方程式为Y=B+(T-B)/1+10((LogEC 50-X)×Hill Slope),Y=活性百分比,B=最小活性百分比,T=最大活性百分比,X=化合物和Hill坡度的对数=坡度因子或Hill系数。IC 50值由引起半数最大活性百分比浓度决定。
化合物编号 IC 50(nM)
1-1-1 2.4
1-1-2 4.1
1-1-3 8.9
1-2-1 6.4
1-2-2 5.1
1-2-3 7.3
1-3-1 5.3
1-3-2 4.2
1-3-3 4.2
1-3-4 11.1
1-4-1 5.0
1-4-2 4.2
1-4-3 19.0
1-5-1 8.2
1-6-1 5.8
1-7-1 8.2
1-7-2 7.2
3-1-1 3.2
4-1-1 7.1
5-1-1 7.9
7-1-1 6.3
7-1-3 13.5
7-1-4 3.2
7-1-5 5.8
7-1-6 8.7
7-2-1 10.1
7-2-6 8.3
8-1-1 6.0
8-1-2 7.1
9-1-1 4.8
AZD1775 8.7
实施例2:以p-CDK1Y15ELISA测试为基础的Wee1细胞活性检测
在此项发明中,运用细胞试验来评价化合物的生物活性。这一试验运用人类结肠腺癌细胞系WiDr开展。运用p-CDK1Y15ELISA试验法评估特定Wee1抑制剂的活性。详细试验方法描述如下:
将WiDr细胞放在含有10%FBS的Dulbecco’s Modified Eagle’s介质中培养,培养环境37℃及5%CO 2。化合物浓度范围0.5nM~30μM。将化合物稀释后加入至24孔板中,与细胞一起孵化24个小时。将DMSO作为阴性对照。在部分试验中加入已知的Wee1抑制剂作为阳性对照。根据生产厂商的指示,在p-CDK1Y15试验中,细胞被溶解并经过比色ELISA试剂盒测试判断p-CDK1Y15的数量。运用分光光度计测量光密度。运用Graphpad软件分析OD数据,得到IC 50值和化合物的拟合曲线。
化合物编号 IC 50(nM)
1-1-1 99.7
1-3-1 55.5
1-3-2 65.3
1-4-1 59.4
7-1-1 41.1
7-2-1 65.7
AZD1775 254
实施例3:细胞增殖试验
本发明中,运用细胞试验法来评价化合物的生物活性。MG63(ATCC CRL-1427),人类骨肉瘤细胞系,将细胞种于Dulbecco’s Modified Eagle’s介质96孔板中培养,补给10%胎牛血清及1%(V/V)左旋谷酰胺,培养环境为37℃及5%CO 2。化合物浓度范围为4.5nM~30μM。将Wee1抑制剂储备液溶入DMSO并加入至指示浓度的介质中,孵化72小时。阴性对照细胞仅用vehicle进行处理。在部分试验中,加入已知的Wee1抑制剂作为阳性对照。在产品说明书的指示下运用细胞计数试剂盒-8(CCK-8,Sigma-Aldrich)来评价细胞活性。运用Graphpad软件对数据进行分析,并得到IC 50值及化合物拟合曲线。
化合物编号 IC 50(nM)
1-1-1 293
1-2-3 241
1-3-1 118
1-3-2 191
1-3-3 225
1-3-4 591
1-4-1 85.5
1-4-2 471
1-4-4 161
1-5-1 559
1-6-1 254
1-7-3 502
4-1-1 596
5-1-1 103
6-1-2 491
7-1-1 42.5
7-1-3 932
7-1-4 44.6
7-1-5 547
7-2-1 66.4
7-2-4 46.8
7-2-5 428
7-2-6 522
7-2-8 329
7-2-9 112
7-2-10 135
7-2-12 72.1
7-2-13 43.3
7-2-14 56.6
7-3-1 129
8-1-1 159
8-1-2 224
9-1-1 81.3
9-2-1 148
AZD1775 785
实施例4:人肺癌H1299异种移植瘤模型体内药效实验
细胞培养:人肺癌H1299细胞(购于ATCC)复苏后,常规培养于含有10%热灭活胎牛血清、100U/mL青霉素、100μg/mL链霉素和L-谷氨酰胺(2mM)的EMEM培养基 中,置于5%CO 2的恒温培养箱,37℃培养。用胰蛋白酶-EDTA消化后每周传代培养2次。收取指数生长期的细胞并计数,以供接种。
实验动物:BALB/c nude小鼠,24只,7-8周,16-21g,购于北京维通利华实验动物技术有限公司。
设置4个实验组,给药方案如表1所示:
表1
组别 小鼠数量 受试化合物 剂量 给药方式 给药计划
1 6 Vehicle -- p.o. bid×16天
2 6 AZD1775 60mg/kg p.o. bid×16天
3 6 化合物7-2-1 30mg/kg p.o. qd×16天
4 6 化合物7-2-1 15mg/kg p.o. qd×16天
注:p.o.:口服,bid:2次/天,qd:1次/天
实验方法:将H1299细胞株(5×10 6个/只)接种于实验小鼠右侧背部皮下,每只小鼠接种量是0.1mL的PBS:Matrigel(体积比1:1),定期观察肿瘤的生长情况,待肿瘤生长至100mm 3左右时根据肿瘤大小和小鼠体重随机分组,按照给药方案(表1)给药,整个实验过程中,每周测量2~3次小鼠体重和肿瘤大小。
肿瘤大小计算公式:肿瘤体积(mm 3)=0.5×(肿瘤长径×肿瘤短径 2)。
四个实验组肿瘤体积变化曲线见附图1,结果表明:相较于阳性对照AZD1775,本发明化合物7-2-1在更低给药剂量和更长给药间隔的情况下,在人肺癌H1299异种移植瘤模型上还能够显示出更好的抑瘤效果。
本发明中生物实施例1、2、3和4中的阳性对照为AZD1775,化学名称:2-烯丙基-1-(6-(2-羟基丙-2-基)吡啶-2-基)-6-((4-(4-甲基哌嗪-1-基)苯基)氨基)-1H-吡唑并[3,4-d]嘧啶-3(2H)-酮(2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((4-(4-methylpiperazin-1-yl)phenyl)amino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one),合成方法可参考WO2007126122A1实施例9。

Claims (13)

  1. 一种如式I或I’所示化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐;
    Figure PCTCN2018101290-appb-100001
    其中,W为N或CR 7
    X为CR 8或N;Y为NR 9、CHR 8、-C(R 8)=C(R 11)-、-N=C(R 11)-、O或S;
    U和V分别选自N或CR 8;并且U和V不同时为N;
    R 1为氢、卤素、C 2-6炔基、C 2-6烯基、芳基、环烷基、杂环烷基、杂芳基或取代的烷基;所述烷基被取代时,可选择性地被如下一个或多个基团取代在任意位置:卤素、卤代烷氧基、氨基环烷基、-SR a、-OR a、-OC(O)R a、-OC(O)NR aR b、-C(O)OR a、-C(O)R a、-C(O)NR aR b、-NR aR b、-NR aC(O)R b、-NR aS(O) 2R b、-S(O) 1-2R b、-S(O) 2NR aR b和-NR aS(O) 2NR aR b
    R 2为烷基、卤代烷基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基;
    R 3为氢、卤素、-CN、-NO 2、-SR c、-OR c、-OC(O)R c、-OC(O)OR c、-OC(O)NR cR d、-C(O)OR c、-C(O)R c、-C(O)NR cR d、-NR cR d、-NR cC(O)R d、-N(R c)C(O)OR d、-N(R c)C(O)NR cR d、-NR cS(O) 2R d、-NR cC(=NH)R d、-NR cC(=NH)NR cR d、-S(O) 1-2R c、-S(O) 2NR cR d、-NR cS(O) 2NR cR d、取代或未取代的烷基、取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的芳基、取代或未取代的杂芳基、取代或未取代的环烷基烷基、取代或未取代的杂环烷基烷基、取代或未取代的芳基烷基、取代或未取代的杂芳基烷基、取代或未取代的烯基、取代或未取代的炔基;所述烷基、环烷基、杂环烷基、芳基、杂芳基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基被取代时,可选择性地被如下一个或多个基团取代在任意位置:卤素、烷基、卤代烷基、卤代烷氧基、羟基烷基、氨基烷基、氨基环烷基、烯基、炔基、-CN、-NO 2、-SR a、-OR a、-OC(O)R a、-OC(O)OR a、-OC(O)NR aR b、-C(O)OR a、-C(O)R a、-C(O)NR aR b、-NR aR b、-NR aC(O)R b、-N(R a)C(O)OR b、-N(R a)C(O)NR aR b、-NR aS(O) 2R b、-NR aC(=NH)R b、-NR aC(=NH)NR aR b、-S(O) 1-2R b、-S(O) 2NR aR b和-NR aS(O) 2NR aR b
    R 4、R 5、R 6和R 7分别独立地为氢、卤素、羟基、氰基、硝基、巯基、氨基、烷基、烷氧基、烷硫基、卤代烷基、卤代烷氧基、C 2-6炔基、C 2-6烯基、芳基、环烷基、杂环烷基、杂芳基、-OC(O)R c、-OC(O)OR c、-OC(O)N(R c) 2、-C(O)OR c、-C(O)R c、-C(O)N(R c) 2、-N(R c) 2、-NHC(O)R c、-NHC(O)OR c、-NHC(O)N(R c) 2、-NHS(O) 2R c、-S(O) 0-2R c或-S(O) 2N(R c) 2
    R 3和R 4为独立取代基,或者,R 3和R 4与其所连接的环原子一起形成A环,所述A 环为取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的苯基、或取代或未取代的5-6元杂芳基;所述A环被取代时,可选择性地被如下一个或多个基团取代在任意位置:氧代基、硫代基、卤素、-CN、-NO 2、-SR c、-OR c、-OC(O)R c、-OC(O)OR c、-OC(O)NR cR d、-C(O)OR c、-C(O)R c、-C(O)NR cR d、-NR cR d、-NR cC(O)R d、-N(R c)C(O)OR d、-N(R c)C(O)NR cR d、-NR cS(O) 2R d、-NR cC(=NH)R d、-NR cC(=NH)NR cR d、-S(O) 1-2R c、-S(O) 2NR cR d、-NR cS(O) 2NR cR d、取代或未取代的烷基、取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的芳基、取代或未取代的杂芳基、取代或未取代的环烷基烷基、取代或未取代的杂环烷基烷基、取代或未取代的芳基烷基、取代或未取代的杂芳基烷基、取代或未取代的烯基、取代或未取代的炔基;所述烷基、环烷基、杂环烷基、芳基、杂芳基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基被取代时,可选择性地被如下一个或多个基团取代在任意位置:卤素、烷基、卤代烷基、卤代烷氧基、羟基烷基、氨基烷基、烯基、炔基、-CN、-NO 2、-SR c、-OR c、-OC(O)R c、-OC(O)OR c、-OC(O)N(R c) 2、-C(O)OR c、-C(O)R c、-C(O)N(R c) 2、-N(R c) 2、-NHC(O)R c、-NHC(O)OR c、-NHC(O)N(R c) 2、-NHS(O) 2R c、-NHC(=NH)R c、-NHC(=NH)N(R c) 2、-S(O) 1-2R c、-S(O) 2N(R c) 2和-NHS(O) 2N(R c) 2
    R 8和R 11分别独立地为氢、-C(O)OR c、-C(O)R c、-C(O)NR cR d、-S(O) 1-2R c、-S(O) 2NR cR d、-NR cR d、-NR cS(O) 1-2R d、-NR cC(O)R d、-NR cS(O) 2NR cR d、取代或未取代的烷基、取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的芳基、取代或未取代的杂芳基、取代或未取代的环烷基烷基、取代或未取代的杂环烷基烷基、取代或未取代的芳基烷基、取代或未取代的杂芳基烷基、取代或未取代的烯基、取代或未取代的炔基;所述烷基、环烷基、杂环烷基、芳基、杂芳基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基被取代时,可选择性地被如下一个或多个R 10取代在任意位置;
    R 9为氢、-C(O)OR c、-C(O)R c、-C(O)NR cR d、-S(O) 1-2R c、-S(O) 2NR cR d、取代或未取代的烷基、取代或未取代的环烷基、取代或未取代的杂环烷基、取代或未取代的芳基、取代或未取代的杂芳基、取代或未取代的环烷基烷基、取代或未取代的杂环烷基烷基、取代或未取代的芳基烷基、取代或未取代的杂芳基烷基、取代或未取代的烯基、取代或未取代的炔基;所述烷基、环烷基、杂环烷基、芳基、杂芳基、环烷基烷基、杂环烷基烷基、芳基烷基、杂芳基烷基、烯基或炔基被取代时,可选择性地被如下一个或多个R 10取代在任意位置;
    R 1和R 9为独立取代基,或者,R 1和R 9相互连接形成4-8元杂环烷基,所述杂环烷基还可进一步被1~3个C 1-6烷基或C 3-6环烷基任意取代;
    每个R 10分别独立地为氢、卤素、烷基、卤代烷基、卤代烷氧基、羟基烷基、氨基烷基、氨基环烷基、烯基、炔基、环烷基、杂环烷基、R a、-CN、-NO 2、-SR a、-OR a、-OC(O)R a、-OC(O)OR a、-OC(O)NR aR b、-C(O)OR a、-C(O)R a、-C(O)NR aR b、-NR aR b、-NR aC(O)R b、-N(R a)C(O)OR b、-N(R a)C(O)NR aR b、-NR aS(O) 2R b、-NR aC(=NH)R b、-NR aC(=NH)NR aR b、-S(O) 1-2R b、-S(O) 2NR aR b、或-NR aS(O) 2NR aR b
    R a和R b分别独立地选自氢、C 1-6烷基、C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷 基、3-8元杂环烷基、C 6-10芳基、5-6元杂芳基、C 3-8环烷基C 1-6烷基、3-8元杂环烷基C 1-6烷基、苯基C 1-6烷基、或5-6元杂芳基C 1-6烷基;所述C 1-6烷基、C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环烷基、C 6-10芳基、或5-6元杂芳基为未取代或者选择性地被1~3个选自卤素、羟基、氨基、羧基、C 1-6烷基、卤代C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基和卤代C 1-6烷氧基中的一种或多种取代基取代在任意位置;或者,所述R a和R b与其共同连接的N原子一起形成3-12元杂环烷基,所述杂环烷基还可以进一步含有1~3个选自N、O、S(O) 0-2、C(O)的杂原子或基团;所述杂环烷基为未取代或者选择性被1~3个选自卤素、羟基、氨基、羧基、C 1-6烷基、卤代C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基和卤代C 1-6烷氧基中的一种或多种取代基取代在任意位置;
    R c和R d分别独立地选自氢、C 1-6烷基、C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环烷基、C 6-10芳基、5-6元杂芳基、C 3-8环烷基C 1-6烷基、3-8元杂环烷基C 1-6烷基、苯基C 1-6烷基、或5-6元杂芳基C 1-6烷基;所述C 1-6烷基、C 1-6烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环烷基、C 6-10芳基、或5-6元杂芳基为未取代或者选择性地被1~3个选自卤素、羟基、氨基、羧基、C 1-6烷基、卤代C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基和卤代C 1-6烷氧基中的一种或多种取代基取代在任意位置。
  2. 如权利要求1所述的如式I或I’所示的化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,其特征在于:R 2
    Figure PCTCN2018101290-appb-100002
    Figure PCTCN2018101290-appb-100003
  3. 如权利要求1所述的如式I或I’所示的化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,其特征在于:基团
    Figure PCTCN2018101290-appb-100004
    为以下任一结构:
    Figure PCTCN2018101290-appb-100005
    基团
    Figure PCTCN2018101290-appb-100006
    为以下任一结构:
    Figure PCTCN2018101290-appb-100007
    R 1、R 8、R 9、R 11的定义如权利要求1所述。
  4. 如权利要求1所述的如式I所示的化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,其特征在于:所述的如式I所示的化合物为III、IV或V,
    Figure PCTCN2018101290-appb-100008
    III中,n为0、1、2、或3;R 1、R 3、R 4、R 5、X、R a、R b和W的定义如权利要求1所述;
    Figure PCTCN2018101290-appb-100009
    IV中,Z为N或CH;L为CH 2、CH(CH 3)、C(CH 3) 2、或
    Figure PCTCN2018101290-appb-100010
    u为0、1、或2;v为0、1、或2;R 1、R 3、R 4、R 5、R 10、X和W的定义如权利要求1所述;
    Figure PCTCN2018101290-appb-100011
    V中,R 1、R 2、R 3、R 4、R 5、R 8、R 9、U和W的定义如权利要求1所述。
  5. 如权利要求4所述的如式I所示的化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,其特征在于:W为N;
    和/或,R 3为取代或未取代的C 1-6烷基,当所述烷基被取代时,可被1个羟基取代在任意位置;
    和/或,R 4为H;
    和/或,R 5为H。
  6. 如权利要求4所述的如式I所示的化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,其特征在于:所述的如式I所示的化合物为V,
    Figure PCTCN2018101290-appb-100012
    其中,U为CH;
    和/或,R 1为H或甲基;
    和/或,R 2
    Figure PCTCN2018101290-appb-100013
    和/或,R 8为-NR cR d、或取代或未取代的3-8元杂环烷基;当所述3-8元杂环烷基被取代时,选择性被1~3个C 1-4烷基、或C 3-6环烷基取代在任意位置;
    和/或,R 9为C 1-4烷基、羟基C 1-4烷基、C 3-8环烷基、或3-8元杂环烷基。
  7. 如权利要求1所述的如式I或I’所示的化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,其特征在于:所述的如式I或I’所示的化合物为:
    Figure PCTCN2018101290-appb-100014
    Figure PCTCN2018101290-appb-100015
    Figure PCTCN2018101290-appb-100016
    Figure PCTCN2018101290-appb-100017
    Figure PCTCN2018101290-appb-100018
    Figure PCTCN2018101290-appb-100019
    Figure PCTCN2018101290-appb-100020
    Figure PCTCN2018101290-appb-100021
  8. 一种药物组合物,其包括治疗有效量的活性组分以及药学上可接受的辅料;所述活性组分包括如权利要求1~7任一项所述的如式I或I’所示的化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐;所述的药学上可接受的辅料为药学上可接受的载体、稀释剂和/或赋形剂。
  9. 如权利要求1~7任一项所述的如式I或I’所示的化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,或如权利要求8所述药物组合物在制备Wee1抑制剂中的应用。
  10. 如权利要求1~7任一项所述的如式I或I’所示的化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,或如权利要求8所述药物组合物在制备癌症的化学疗法或放射性疗法的增敏剂中的应用。
  11. 如权利要求1~7任一项所述的如式I或I’所示的化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,或如权利要求8所述药物组合物在制备治疗和/或缓解由Wee1介导的相关疾病的药物中的应用。
  12. 如权利要求11所述的应用,其特征在于:所述的Wee1介导的相关疾病为癌症。
  13. 一种联合制剂,包括如式I或I’所示化合物、其异构体、前药、稳定的同位素衍生物或药学上可接受的盐,或所述药物组合物和其它种类的用于治疗癌症的治疗剂和/或治疗方法合用;所述其它种类的用于治疗癌症的治疗剂和/或治疗方法可包括用于癌症治疗的:微管蛋白抑制剂、烷化剂、拓扑酶I/II抑制剂、铂类化合物、抗生素、抗代谢类药物、激素和激素类似物、靶向治疗(例如:特殊的激酶抑制剂)、免疫治疗剂、干扰素、其它用于癌症治疗的抗癌剂和放疗中的一种或多种。
PCT/CN2018/101290 2017-08-24 2018-08-20 吡唑并[3,4-d]嘧啶-3-酮衍生物、其药物组合物及应用 WO2019037678A1 (zh)

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
CN201710737296 2017-08-24
CN201710737296.9 2017-08-24
CN201710849306 2017-09-20
CN201710849306.8 2017-09-20
CN201711263908 2017-12-05
CN201711263908.1 2017-12-05
CN201810213347.2 2018-03-15
CN201810213347 2018-03-15
CN201810531630.X 2018-05-29
CN201810531630 2018-05-29

Publications (1)

Publication Number Publication Date
WO2019037678A1 true WO2019037678A1 (zh) 2019-02-28

Family

ID=65439380

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/101290 WO2019037678A1 (zh) 2017-08-24 2018-08-20 吡唑并[3,4-d]嘧啶-3-酮衍生物、其药物组合物及应用

Country Status (2)

Country Link
CN (1) CN109422754A (zh)
WO (1) WO2019037678A1 (zh)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019173082A1 (en) * 2018-03-09 2019-09-12 Zeno Royalties & Milestones, LLC Substituted l,2-dihydro-3h-pyrazolo[3,4-d]pyrimidin-3-ones
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
US10807994B2 (en) 2017-10-09 2020-10-20 Nuvation Bio Inc. Heterocyclic compounds and uses thereof
WO2020259703A1 (zh) 2019-06-28 2020-12-30 上海医药集团股份有限公司 一种吡唑酮并嘧啶类化合物、其制备方法及应用
WO2020259724A2 (zh) 2019-06-28 2020-12-30 上海医药集团股份有限公司 一种吡唑酮并嘧啶类化合物、其制备方法及应用
WO2021231653A1 (en) * 2020-05-15 2021-11-18 Recurium Ip Holdings, Llc Mono- and combination therapies
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
WO2022171126A1 (zh) * 2021-02-09 2022-08-18 微境生物医药科技(上海)有限公司 作为Wee-1抑制剂的稠环化合物
WO2022171088A1 (zh) * 2021-02-09 2022-08-18 微境生物医药科技(上海)有限公司 吡唑并[3,4-d]嘧啶-3-酮衍生物
WO2022171128A1 (zh) * 2021-02-09 2022-08-18 微境生物医药科技(上海)有限公司 作为Wee-1抑制剂的吡唑并[3,4-d]嘧啶-3-酮衍生物
WO2023016417A1 (zh) * 2021-08-11 2023-02-16 微境生物医药科技(上海)有限公司 作为Wee-1抑制剂的1,2-二氢-3H-吡唑[3,4-d]嘧啶-3-酮化合物
WO2023072301A1 (zh) * 2021-11-01 2023-05-04 正大天晴药业集团股份有限公司 吡唑[3,4-d]嘧啶-3-酮类化合物及其医药用途
EP4069240A4 (en) * 2019-12-20 2023-12-20 Recurium IP Holdings, LLC COMBINATIONS
EP4069235A4 (en) * 2019-12-20 2024-01-10 Recurium Ip Holdings Llc ASSOCIATIONS
US11884647B2 (en) 2019-10-18 2024-01-30 The Regents Of The University Of California Compounds and methods for targeting pathogenic blood vessels

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110872296B (zh) * 2018-08-31 2023-05-23 上海弘翊生物科技有限公司 一种二氢异吲哚-1H-吡唑并[3,4-d]嘧啶酮化合物、其制备方法和应用
AR119494A1 (es) * 2019-07-29 2021-12-22 Servier Lab DERIVADOS DE 6,7-DIHIDRO-5H-PIRIDO[2,3-c]PIRIDAZIN-8-ILO, COMPOSICIONES FARMACÉUTICAS QUE LOS CONTIENEN Y SUS USOS COMO AGENTES PROAPOPTÓTICOS
CN112442049A (zh) * 2019-09-03 2021-03-05 微境生物医药科技(上海)有限公司 作为Wee1抑制剂的嘧啶衍生物
EP4169919A1 (en) * 2020-06-17 2023-04-26 Wigen Biomedicine Technology (Shanghai) Co., Ltd. Pyrazolo[3,4-d]pyrimidine-3-ketone derivative as wee-1 inhibitor
CN114591334B (zh) * 2020-12-04 2023-10-20 山东轩竹医药科技有限公司 二氢吡唑并嘧啶酮衍生物
KR102549484B1 (ko) * 2020-12-08 2023-06-29 한국화학연구원 피라졸로피리미딘 설폰아마이드 유도체 및 이를 유효성분으로 포함하는 암 관련 질환의 예방 또는 치료용 약학적 조성물
CN115197221A (zh) * 2021-04-02 2022-10-18 轩竹生物科技股份有限公司 二氢吡唑并嘧啶酮类大环衍生物及其用途
JP2024515318A (ja) * 2021-04-30 2024-04-08 ウィゲン・バイオメディシン・テクノロジー・(シャンハイ)・カンパニー・リミテッド Wee-1阻害剤としての縮合環状化合物、その調製方法およびその使用
WO2024012549A1 (zh) * 2022-07-15 2024-01-18 映恩生物制药(苏州)有限公司 一种嘧啶并五元杂环化合物、其制备方法和用途

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101432284A (zh) * 2006-04-27 2009-05-13 万有制药株式会社 二氢吡唑并嘧啶酮衍生物
US20100221211A1 (en) * 2007-10-23 2010-09-02 Hidetomo Furuyama Pyridone-substituted-dihydropyrazolopyrimidinone derivative
WO2011034743A1 (en) * 2009-09-15 2011-03-24 Merck Sharp & Dohme Corp. Preparation of crystalline forms of dihydropyrazolopyrimidinone
CN104271576A (zh) * 2012-02-23 2015-01-07 艾伯维公司 激酶的吡啶并嘧啶酮抑制剂
CN105829315A (zh) * 2013-12-19 2016-08-03 阿尔麦克探索有限公司 可用作Wee-1激酶抑制剂的嘧啶并嘧啶酮类
WO2017075629A2 (en) * 2015-11-01 2017-05-04 The Regents Of The University Of Colorado, A Body Corporate Wee 1 kinase inhibitors and methods of making and using the same

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2745959A1 (en) * 2008-12-12 2010-06-17 Msd K.K. Dihydropyrimidopyrimidine derivatives
GB201612092D0 (en) * 2016-07-12 2016-08-24 Almac Discovery Ltd Pharmaceutical compounds

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101432284A (zh) * 2006-04-27 2009-05-13 万有制药株式会社 二氢吡唑并嘧啶酮衍生物
US20100221211A1 (en) * 2007-10-23 2010-09-02 Hidetomo Furuyama Pyridone-substituted-dihydropyrazolopyrimidinone derivative
WO2011034743A1 (en) * 2009-09-15 2011-03-24 Merck Sharp & Dohme Corp. Preparation of crystalline forms of dihydropyrazolopyrimidinone
CN104271576A (zh) * 2012-02-23 2015-01-07 艾伯维公司 激酶的吡啶并嘧啶酮抑制剂
CN105829315A (zh) * 2013-12-19 2016-08-03 阿尔麦克探索有限公司 可用作Wee-1激酶抑制剂的嘧啶并嘧啶酮类
WO2017075629A2 (en) * 2015-11-01 2017-05-04 The Regents Of The University Of Colorado, A Body Corporate Wee 1 kinase inhibitors and methods of making and using the same

Cited By (21)

* 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
US11261192B2 (en) * 2018-03-09 2022-03-01 Recurium Ip Holdings, Llc Substituted 1,2-dihydro-3H-pyrazolo[3,4-D]pyrimidin-3-ones
AU2019231551B2 (en) * 2018-03-09 2023-08-03 Recurium Ip Holdings, Llc Substituted 1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-ones
JP7300460B2 (ja) 2018-03-09 2023-06-29 リキュリウム アイピー ホールディングス リミテッド ライアビリティー カンパニー 置換1,2-ジヒドロ-3H-ピラゾロ[3,4-d]ピリミジン-3-オン
WO2019173082A1 (en) * 2018-03-09 2019-09-12 Zeno Royalties & Milestones, LLC Substituted l,2-dihydro-3h-pyrazolo[3,4-d]pyrimidin-3-ones
CN113614087B (zh) * 2019-03-22 2023-06-02 首药控股(北京)股份有限公司 Wee1抑制剂及其制备和用途
CN113614087A (zh) * 2019-03-22 2021-11-05 首药控股(北京)股份有限公司 Wee1抑制剂及其制备和用途
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
US11332473B2 (en) 2019-04-09 2022-05-17 Nuvation Bio Inc. Substituted pyrazolo[3,4-d]pyrimidines as Wee1 inhibitors
WO2020259703A1 (zh) 2019-06-28 2020-12-30 上海医药集团股份有限公司 一种吡唑酮并嘧啶类化合物、其制备方法及应用
WO2020259724A2 (zh) 2019-06-28 2020-12-30 上海医药集团股份有限公司 一种吡唑酮并嘧啶类化合物、其制备方法及应用
US11884647B2 (en) 2019-10-18 2024-01-30 The Regents Of The University Of California Compounds and methods for targeting pathogenic blood vessels
EP4069235A4 (en) * 2019-12-20 2024-01-10 Recurium Ip Holdings Llc ASSOCIATIONS
EP4069240A4 (en) * 2019-12-20 2023-12-20 Recurium IP Holdings, LLC COMBINATIONS
WO2021231653A1 (en) * 2020-05-15 2021-11-18 Recurium Ip Holdings, Llc Mono- and combination therapies
WO2022171126A1 (zh) * 2021-02-09 2022-08-18 微境生物医药科技(上海)有限公司 作为Wee-1抑制剂的稠环化合物
WO2022171128A1 (zh) * 2021-02-09 2022-08-18 微境生物医药科技(上海)有限公司 作为Wee-1抑制剂的吡唑并[3,4-d]嘧啶-3-酮衍生物
WO2022171088A1 (zh) * 2021-02-09 2022-08-18 微境生物医药科技(上海)有限公司 吡唑并[3,4-d]嘧啶-3-酮衍生物
WO2023016417A1 (zh) * 2021-08-11 2023-02-16 微境生物医药科技(上海)有限公司 作为Wee-1抑制剂的1,2-二氢-3H-吡唑[3,4-d]嘧啶-3-酮化合物
WO2023072301A1 (zh) * 2021-11-01 2023-05-04 正大天晴药业集团股份有限公司 吡唑[3,4-d]嘧啶-3-酮类化合物及其医药用途

Also Published As

Publication number Publication date
CN109422754A (zh) 2019-03-05

Similar Documents

Publication Publication Date Title
WO2019037678A1 (zh) 吡唑并[3,4-d]嘧啶-3-酮衍生物、其药物组合物及应用
TWI714567B (zh) 作為lsd1抑制劑之雜環化合物
JP6586104B2 (ja) 5−置換インダゾール−3−カルボキサミドならびにその調製および使用の方法
ES2904544T3 (es) Compuestos de indazol como inhibidores de la cinasa FGFR, preparación y uso de los mismos
CN104884458B (zh) 作为蛋白质激酶抑制剂的稠合杂环化合物
RU2537945C2 (ru) Триазиновые, пиримидиновые и пиридиновые аналоги и их применение в качестве терапевтических агентов и диагностических проб
CN108349896B (zh) 作为fgfr抑制剂的杂环化合物
JP2020511520A (ja) ピラゾロ[3,4−d]ピリミジン−3−オンの大環状誘導体、その医薬組成物及び応用
JP2014051516A (ja) タンパク質キナーゼ阻害剤として有用な5−シアノ−4−(ピロロ[2,3b]ピリジン−3−イル)−ピリミジン誘導体
KR20080052630A (ko) Raf 억제제 화합물 및 그의 사용 방법
WO2011114275A1 (en) Spirocyclic compounds and their use as therapeutic agents and diagnostic probes
CA2873975A1 (en) Thienopyrimidines
WO2017071516A1 (zh) 一种蛋白激酶抑制剂及其制备方法和医药用途
CN114981268A (zh) 嘧啶-4(3h)-酮类杂环化合物、其制备方法及其在医药学上的应用
EP3398947A1 (en) Nitrogen-containing fused heterocyclic compound, as well as preparation method, intermediate, composition and application thereof
TW202328124A (zh) 1,4-氧雜氮雜環庚烷衍生物及其用途
CN113387962A (zh) 吡唑并[3,4-d]嘧啶-3-酮衍生物、其药物组合物及应用
CA3221317A1 (en) Substituted fused azines as kras g12d inhibitors
WO2020103897A1 (zh) 杂环稠合嘧啶衍生物、其药物组合物及应用
JP2019038848A (ja) 白血病を予防および治療するためのマレイミド誘導体の使用
WO2018127195A1 (zh) 取代的稠合杂芳基化合物作为激酶抑制剂及其应用
TWI826535B (zh) 環狀二核苷酸類似物、其藥物組合物及應用
CN114249712A (zh) 嘧啶基衍生物、其制备方法及其用途
TW202400601A (zh) 作為parp抑製劑的取代的三環類化合物及其用途
CA3117319A1 (en) Fused bicyclic heterocycles as thereapeutic agents

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: 18849221

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC, EPO FORM 1205A DATED 28.09.2020

122 Ep: pct application non-entry in european phase

Ref document number: 18849221

Country of ref document: EP

Kind code of ref document: A1