CN111646995B - 4-amino-pyrimidoazenitrogen heterocycle-phenylurea derivative and preparation method and application thereof - Google Patents

4-amino-pyrimidoazenitrogen heterocycle-phenylurea derivative and preparation method and application thereof Download PDF

Info

Publication number
CN111646995B
CN111646995B CN202010123847.4A CN202010123847A CN111646995B CN 111646995 B CN111646995 B CN 111646995B CN 202010123847 A CN202010123847 A CN 202010123847A CN 111646995 B CN111646995 B CN 111646995B
Authority
CN
China
Prior art keywords
alkyl
amino
substituted
clj
cycloalkyl
Prior art date
Legal status (The legal status 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 status listed.)
Active
Application number
CN202010123847.4A
Other languages
Chinese (zh)
Other versions
CN111646995A (en
Inventor
陈俐娟
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sichuan University
Original Assignee
Sichuan University
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 Sichuan University filed Critical Sichuan University
Publication of CN111646995A publication Critical patent/CN111646995A/en
Application granted granted Critical
Publication of CN111646995B publication Critical patent/CN111646995B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention belongs to the field of chemical medicine, and particularly relates to a 4-amino-pyrimidoazepine-phenylurea derivative, and a preparation method and application thereof. The invention provides a 4-amino-pyrimidoazepine-phenylurea derivative, the structural formula of which is shown in formula I. In addition, the invention also provides a preparation method and application of the 4-amino-pyrimidoazepine-phenylurea derivative. The 4-amino-pyrimidoazepine-phenylurea derivative provided by the invention can be used as a FLT3 kinase inhibitor, has a good effect, and provides a new choice for preparing antitumor drugs.
Figure DDA0002393819110000011

Description

4-amino-pyrimidoazenitrogen heterocycle-phenylurea derivative and preparation method and application thereof
Technical Field
The invention belongs to the field of chemical medicine, and particularly relates to a 4-amino-pyrimidoazepine-phenylurea derivative, and a preparation method and application thereof.
Background
Leukemia is an abnormal clonal disease of hematopoietic stem cells, is arrested in different stages of cell development and malignant proliferation because of the loss of the ability of the leukemia cells to differentiate into mature functional blood cells, and the leukemia cells are proliferated and accumulated in large quantity in bone marrow and other hematopoietic tissues and infiltrate into other organs and tissues, so that normal hematopoiesis is inhibited, and symptoms such as anemia, hemorrhage, infection, infiltration of various organs and the like are clinically shown. Leukemia belongs to cell heterogeneous malignant tumor, has various types, complex etiology and different clinical manifestations, and some leukemias have the characteristics of quick onset, high mortality, short survival period, easy relapse, poor prognosis and extremely difficult cure.
Protein kinases, the family of receptor protein tyrosine kinases in particular, which catalyze the phosphorylation of hydroxyl groups on tyrosine, serine and threonine residues of proteins, play an important role as growth factor receptors in the control of many signal transduction pathways responsible for cellular functions, such as cell cycle, cell growth, cell differentiation and cell death. Dysregulation of receptor tyrosine kinases is often found in diseases such as proliferative disorders, inflammatory disorders, and immune system disorders.
FLT3 (Fms-like tyrosine kinase 3) is an Fms-like tyrosine kinase 3, which belongs to a member of the type III receptor tyrosine kinase (RTK III) family together with c-Kit, c-Fms and PDGFR, and the structure of FLT3 includes an extracellular domain consisting of 5 immunoglobulin-like molecules, a transmembrane domain and an intracellular tyrosine kinase domain. FLT3 is expressed predominantly on the cell surface of normal hematopoietic stem and progenitor cells, and its ligand is expressed predominantly in bone marrow stromal cells. When ligand is combined with the membrane outer structure domain of FLT3, the ligand promotes dimerization of FLT3 receptor, and simultaneously, the tyrosine kinase domain in the cell membrane generates autophosphorylation, and a series of downstream signal transduction pathways, such as Ras/MAPK, PI3K/Akt/mTOR and STAT5, are activated, so that the proliferation and differentiation of cells are regulated. FLT3 mutation usually leads to its abnormal activation, and in the absence of ligand binding, autophosphorylation activates downstream signaling pathways, leading to abnormal proliferation of hematopoietic cells and lymphocytes, leading to various hematological malignancies.
It has been demonstrated that there are two major activating mutations for FLT 3: point mutations in the activation loop in the internal tandem repeat (ITD) and kinase domain (TKD). FLT3-ITD mutations occur in about 25% of acute myeloid leukemia patients and are associated with some adverse prognosis, while FLT3-TKD mutations occur in about 5% of acute myeloid leukemia patients.
Numerous studies have shown that FLT3 mutation is one of the most common molecular genetic abnormalities and poor prognosis factors in AML, and its transduction involves multiple signaling pathways, making FLT3 an ideal drug target. In previous studies, more than 20 compounds have shown inhibition of FLT3 tyrosine kinase. Many have entered clinical trials. These small molecule tyrosinase inhibitors are mostly heterocyclic purine analogs, are ATP analogs, or intermediates with a structure similar to tyrosine covalently bound to ATP. FLT3 receptor is still expressed, but the ATP pathway is blocked, thus autophosphorylation and sustained phosphorylation of the substrate is terminated, blocking the signaling pathway of FLT3-ITD dependent cell lines, exerting a cytotoxic effect.
At present, research on FLT3-ITD and FLT3-TKD inhibitors belongs to a hotspot of drug development, a single-target FLT3 inhibitor AC220 is currently subjected to clinical three-stage tests and is expected to be on the market, but drug resistance in the later treatment stage becomes a problem to be solved urgently. Therefore, the development of inhibitors effective against both mutations is the direction of development.
Disclosure of Invention
In order to solve the problems, the invention provides a 4-amino-pyrimidoazepine-phenylurea derivative, the structural formula of which is shown as formula I:
Figure BDA0002393819090000021
wherein X is N or C;
R 1 is-H, -OH, halogen, C1-C10 alkoxy, C1-C10 haloalkoxy,
Figure BDA0002393819090000022
C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl,
Figure BDA0002393819090000023
Or substituted or unsubstituted C1-C10 alkyl; the substituent of the substituted C1-C10 alkyl is-H,
Figure BDA0002393819090000024
-CN, -OH, phenyl, halogen, C1-C8 alkoxy, C3-C8 cycloalkyl, C1-C8 carbonyl or substituted or unsubstituted 3-to 6-membered heterocycloalkyl; the heteroatom of the 3-6 membered heterocycloalkyl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted 3-to 6-membered heterocycloalkyl is-H, -OH, halogen, C1-C8 oxycarbonyl, C1-C8 alkyl, C1-C8 alkoxy or
Figure BDA0002393819090000025
R 4 is-H, -OH, halogen, C1-C8 alkyl, C1-C8 alkoxy or
Figure BDA0002393819090000026
R 5 ~R 10 Independently is-H or C1-C8 alkyl;
R 2 is-H, -OH, halogen, C1-C10 alkyl, C3-C10 cycloalkyl, C1-C10 haloalkyl, amino-substituted C1-C10 alkyl, benzyl, substituted or unsubstituted C5-C10 aryl or substituted or unsubstituted 5-to 10-membered heteroaryl; the heteroatom of the 5-to 10-membered heteroaryl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted C5-C10 aryl or 5-10 membered heteroaryl is-H, halogen or-NH 2 C1-C8 alkyl, C3-C8 cycloalkyl, C1-C8 haloalkyl, C1-C8 alkoxy, C1-C8 haloalkoxy, -OH or C1-C8 carbonyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C10 alkyl.
As a preferred embodiment of the present invention, X is N or C; r 1 is-H, -OH, halogen, C1-C8 alkoxy, C1-C8 haloalkoxy,
Figure BDA0002393819090000031
C2-C8 alkenyl, C2-C8 alkynylC3-C8 cycloalkyl,
Figure BDA0002393819090000032
Or substituted or unsubstituted C1-C8 alkyl; the substituent of the substituted C1-C8 alkyl is-H,
Figure BDA0002393819090000033
-CN, -OH, phenyl, halogen, C1-C6 alkoxy, C3-C6 cycloalkyl, C1-C6 carbonyl or substituted or unsubstituted 3-to 6-membered heterocycloalkyl; the heteroatom of the 3-6 membered heterocycloalkyl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted 3-to 6-membered heterocycloalkyl is-H, -OH, halogen, C1-C6 oxycarbonyl, C1-C6 alkyl, C1-C6 alkoxy or
Figure BDA0002393819090000034
R 4 is-H, -OH, halogen, C1-C6 alkyl, C1-C6 alkoxy or
Figure BDA0002393819090000035
R 5 ~R 10 Independently is-H or C1-C6 alkyl;
R 2 is-H, -OH, halogen, C1-C8 alkyl, C3-C8 cycloalkyl, C1-C8 haloalkyl, amino-substituted C1-C8 alkyl, benzyl, substituted or unsubstituted C5-C8 aryl, or substituted or unsubstituted 5-to 8-membered heteroaryl; the heteroatom of the 5-to 8-membered heteroaryl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted C5-C8 aryl or 5-8 membered heteroaryl is-H, halogen or-NH 2 C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, -OH or C1-C6 carbonyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C8 alkyl.
Further, as a preferable embodiment of the present invention, X is N or C; r 1 is-H, -OH, halogen, C1-C8 alkyl, C1-C6 alkoxy, C1-C6 haloalkoxy,
Figure BDA0002393819090000036
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl,
Figure BDA0002393819090000037
Or substituted C1-C6 alkyl; the substituent of the substituted C1-C6 alkyl is-H,
Figure BDA0002393819090000038
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 3-to 6-membered heterocycloalkyl; the heteroatom of the 3-6 membered heterocycloalkyl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted 3-to 6-membered heterocycloalkyl is-H, -OH, halogen, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000039
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA00023938190900000310
R 5 ~R 10 Independently is-H or C1-C4 alkyl;
R 2 is-H, -OH, halogen, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 haloalkyl, amino-substituted C1-C6 alkyl, benzyl, substituted or unsubstituted C5-C6 aryl or substituted or unsubstituted 5-to 6-membered heteroaryl; the heteroatom of the 5-6 membered heteroaryl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted C5-C6 aryl or 5-6 membered heteroaryl is-H, halogen or-NH 2 C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, -OH or C1-C4 carbonyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C6 alkyl.
Preferably, the 4-amino-pyrimidoazepine-phenylurea derivative has X being N or C; r 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000041
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000042
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 5-to 6-membered heterocycloalkyl; the heteroatom of the 5-6 membered heterocycloalkyl is N or O, and the number of the heteroatoms is 1-2; the substituent for substituting the 5-to 6-membered heterocycloalkyl is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000043
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000044
R 5 ~R 10 Independently is-H or C1-C4 alkyl;
R 2 is-H, -OH, halogen, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 haloalkyl, amino-substituted C1-C6 alkyl, benzyl, substituted or unsubstituted C5-C6 aryl or substituted or unsubstituted 5-to 6-membered heteroaryl; the heteroatom of the 5-6 membered heteroaryl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted C5-C6 aryl or 5-6 membered heteroaryl is-H, halogen or-NH 2 C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, -OH or C1-C4 carbonyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C6 alkyl.
Further, X is N or C; r 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000045
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000046
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl,
Figure BDA0002393819090000047
Or
Figure BDA0002393819090000048
R 4 is-H, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000049
R 5 ~R 10 Independently is-H or C1-C4 alkyl; r 11 is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA00023938190900000410
R 2 is-H, -OH, halogen, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 haloalkyl, amino-substituted C1-C6 alkyl, benzyl, substituted or unsubstituted C5-C6 aryl or substituted or unsubstituted 5-to 6-membered heteroaryl; the 5-6 membered heteroaryl has heteroatoms of N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted C5-C6 aryl or 5-6 membered heteroaryl is-H, halogen or-NH 2 C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, -OH or C1-C4 carbonyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C6 alkyl.
Further, X is N or C; r is 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000051
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000052
Figure BDA0002393819090000053
-CN, -OH, phenyl, halogen, methoxy, ethoxy,
Figure BDA0002393819090000054
Formyl, acetyl,
Figure BDA0002393819090000055
Figure BDA0002393819090000056
R 4 is-H, C1-C4 alkyl, tert-butyloxy or
Figure BDA0002393819090000057
R 5 ~R 10 independently-H, methyl or ethyl;
R 2 is-H, -OH, halogen, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 haloalkyl, amino-substituted C1-C6 alkyl, benzyl, substituted or unsubstituted C5-C6 aryl or substituted or unsubstituted 5-to 6-membered heteroaryl; the heteroatom of the 5-6 membered heteroaryl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted C5-C6 aryl or 5-6 membered heteroaryl is-H, halogen or-NH 2 C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, -OH or C1-C4 carbonyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C6 alkyl.
Most preferably, X is N or C; r is 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000058
T-butyloxycarbonyl group, C2-C6 alkenyl group, C2-C6 alkynyl group,
Figure BDA0002393819090000059
Figure BDA00023938190900000510
R 2 is-H, -OH, halogen, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 haloalkyl, amino-substituted C1-C6 alkyl, benzyl, substituted or unsubstituted C5-C6 aryl or substituted or unsubstituted 5-to 6-membered heteroaryl; the 5-6 membered heteroaryl has heteroatoms of N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted C5-C6 aryl or 5-6 membered heteroaryl is-H, halogen or-NH 2 C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, -OH or C1-C4 carbonyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C6 alkyl.
Preferably, the 4-amino-pyrimidoazepine-phenylurea derivative has X being N or C; r is 2 Is benzyl, C3-C6 cycloalkyl, substituted or unsubstituted C5-C6 aryl or substituted or unsubstituted 5-to 6-membered heteroaryl; the heteroatom of the 5-6 membered heteroaryl is N or O, and the number of the heteroatoms is 1-2; the substituent of the substituted C5-C6 aryl or 5-6 membered heteroaryl is-H, halogen or-NH 2 C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, -OH or C1-C4 carbonyl;
R 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000061
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000062
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 5-to 6-membered heterocycloalkyl; the heteroatom of the 5-6 membered heterocycloalkyl is N or O, and the number of the heteroatoms is 1-2; the substituent for substituting the 5-to 6-membered heterocycloalkyl is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000063
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000064
R 5 ~R 10 Independently is-H or C1-C4 alkyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C6 alkyl.
Further, X is N or C; r 2 Is benzyl, C3-C6 cycloalkyl,
Figure BDA0002393819090000065
Figure BDA0002393819090000066
R 12 ~R 19 Independently is-H, halogen, -NH 2 C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, -OH or C1-C4 carbonyl;
R 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000067
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000068
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 5-to 6-membered heterocycloalkyl; the heteroatom of the 5-6 membered heterocycloalkyl is N or O, and the number of the heteroatoms is 1-2; the substituent for substituting the 5-to 6-membered heterocycloalkyl is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000071
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000072
R 5 ~R 10 Independently is-H or C1-C4 alkyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C6 alkyl.
Further, X is N or C; r 2 Is benzyl, C3-C6 cycloalkyl,
Figure BDA0002393819090000073
Figure BDA0002393819090000074
R 16 ~R 19 Independently is-H, halogen, -NH 2 C1-C4 alkyl, C1-C4 haloalkyl, -OH or C1-C4 carbonyl;
R 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000075
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000076
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 5-to 6-membered heterocycloalkyl; the heteroatom of the 5-6 membered heterocycloalkyl is N or O, and the number of the heteroatoms is 1-2; the substituent for substituting the 5-to 6-membered heterocycloalkyl is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000077
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000078
R 5 ~R 10 Independently is-H or C1-C4 alkyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C6 alkyl.
Still further, X is N or C; r 2 Is benzyl, C3-C6 cycloalkyl,
Figure BDA0002393819090000079
Figure BDA00023938190900000710
R 16 ~R 19 Independently is-H, -F, -Cl, -Br, -CF 3 Methyl or acetyl;
R 1 is-H, C1-C8 alkyl,
Figure BDA00023938190900000711
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA00023938190900000712
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 5-to 6-membered heterocycloalkyl; the heteroatom of the 5-6 membered heterocycloalkyl is N or O, and the number of the heteroatoms is 1-2; the substituent for substituting the 5-to 6-membered heterocycloalkyl is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000081
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000082
R 5 ~R 10 Independently is-H or C1-C4 alkyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C6 alkyl.
Most preferably, X is N or C; r 2 Is composed of
Figure BDA0002393819090000083
Figure BDA0002393819090000084
R 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000085
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000086
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 5-to 6-membered heterocycloalkyl; the heteroatom of the 5-6 membered heterocycloalkyl is N or O, and the number of the heteroatoms is 1-2; the substituent for substituting the 5-to 6-membered heterocycloalkyl is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000087
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000088
R 5 ~R 10 Independently is-H or C1-C4 alkyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C6 alkyl.
Preferably, the 4-amino-pyrimidoazepine-phenylurea derivative has X being N or C; r 3 is-H, halogen, -OH or C1-C4 alkyl; r 2 Is benzyl, C3-C6 cycloalkyl, substituted or unsubstituted C5-C6 aryl or substituted or unsubstituted 5-to 6-membered heteroaryl; the heteroatom of the 5-6 membered heteroaryl is N or O, and the number of the heteroatoms is 1-2; the substituent of the substituted C5-C6 aryl or 5-6 membered heteroaryl is-H, halogen or-NH 2 C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, -OH or C1-C4 carbonyl;
R 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000089
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000091
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 5-to 6-membered heterocycloalkyl; the heteroatom of the 5-6 membered heterocycloalkyl is N or O, and the number of the heteroatoms is 1-2; the substituent for substituting the 5-to 6-membered heterocycloalkyl is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000092
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000093
R 5 ~R 10 Independently is-H or C1-C4 alkyl.
Further, X is N or C; r 3 is-H, halogen or C1-C4 alkyl; r 2 Is benzyl, C3-C6 cycloalkyl, substituted or unsubstituted C5-C6 aryl or substituted or unsubstituted 5-to 6-membered heteroaryl; the heteroatom of the 5-6 membered heteroaryl is N or O, and the number of the heteroatoms is 1-2; the substituent of the substituted C5-C6 aryl or 5-6 membered heteroaryl is-H, halogen or-NH 2 C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, -OH or C1-C4 carbonyl;
R 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000094
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000095
-CN, -OH, phenyl, halogenC1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 5-to 6-membered heterocycloalkyl; the heteroatom of the 5-6 membered heterocycloalkyl is N or O, and the number of the heteroatoms is 1-2; the substituent for substituting the 5-to 6-membered heterocycloalkyl is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000096
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000097
R 5 ~R 10 Independently is-H or C1-C4 alkyl.
Further, X is N or C; r is 3 is-H or halogen; r 2 Is benzyl, C3-C6 cycloalkyl, substituted or unsubstituted C5-C6 aryl or substituted or unsubstituted 5-to 6-membered heteroaryl; the heteroatom of the 5-6 membered heteroaryl is N or O, and the number of the heteroatoms is 1-2; the substituent of the substituted C5-C6 aryl or 5-6 membered heteroaryl is-H, halogen or-NH 2 C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, -OH or C1-C4 carbonyl;
R 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000098
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000099
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 5-to 6-membered heterocycloalkyl; the heteroatom of the 5-6 membered heterocycloalkyl is N or O, and the number of the heteroatoms is 1-2; the substituent for substituting the 5-to 6-membered heterocycloalkyl is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000101
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000102
R 5 ~R 10 Independently is-H or C1-C4 alkyl.
Most preferably, X is N or C; r 3 is-H, -F, -Cl or-Br; r 2 Is benzyl, C3-C6 cycloalkyl, substituted or unsubstituted C5-C6 aryl or substituted or unsubstituted 5-to 6-membered heteroaryl; the heteroatom of the 5-6 membered heteroaryl is N or O, and the number of the heteroatoms is 1-2; the substituent of the substituted C5-C6 aryl or 5-6 membered heteroaryl is-H, halogen or-NH 2 C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, -OH or C1-C4 carbonyl;
R 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000103
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000104
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 5-to 6-membered heterocycloalkyl; the heteroatom of the 5-6 membered heterocycloalkyl is N or O, and the number of the heteroatoms is 1-2; the substituent for substituting the 5-to 6-membered heterocycloalkyl is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000105
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000106
R 5 ~R 10 Independently is-H or C1-C4 alkyl.
AsIn the preferable technical scheme of the invention, X is N or C in the 4-amino-pyrimidoazepine-phenylurea derivative; r 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000107
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000108
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl,
Figure BDA0002393819090000109
R 4 is-H, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA00023938190900001010
R 5 ~R 10 Independently is-H or C1-C4 alkyl; r 11 is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA00023938190900001011
R 2 Is benzyl, C3-C6 cycloalkyl,
Figure BDA0002393819090000111
R 12 ~R 19 Independently is-H, halogen, -NH 2 C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, -OH or C1-C4 carbonyl;
R 3 is-H, halogen or C1-C4 alkyl.
Preferably, X is N or C; r 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000112
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000113
-CN, -OH, phenyl, halogen methoxy, ethoxy,
Figure BDA0002393819090000114
Formyl, acetyl,
Figure BDA0002393819090000115
Figure BDA0002393819090000116
R 4 is-H, C1-C4 alkyl, tert-butyloxy or
Figure BDA0002393819090000117
R 5 ~R 10 independently-H, methyl or ethyl;
R 2 is benzyl, C3-C6 cycloalkyl,
Figure BDA0002393819090000118
R 16 ~R 19 Independently is-H, halogen, -NH 2 C1-C4 alkyl, C1-C4 haloalkyl, -OH or C1-C4 carbonyl;
R 3 is-H or halogen.
Most preferably, X is N or C; r is 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000119
T-butyloxycarbonyl, C2-C6 alkenyl, C2-C6 alkynyl,
Figure BDA00023938190900001110
Figure BDA00023938190900001111
Figure BDA0002393819090000121
R 2 Is composed of
Figure BDA0002393819090000122
Figure BDA0002393819090000123
R 3 is-H, -F, -Cl or-Br.
As a preferred embodiment of the present invention, when R is the above-mentioned 4-amino-pyrimidoazepine-phenylurea derivative 2 Is composed of
Figure BDA0002393819090000124
And the structure is shown as formula II:
Figure BDA0002393819090000125
wherein X is N or C; r 1 is-H, -OH, halogen, C1-C10 alkoxy, C1-C10 haloalkoxy,
Figure BDA0002393819090000126
C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl,
Figure BDA0002393819090000127
Or substituted or unsubstituted C1-C10 alkyl; the substituent of the substituted C1-C10 alkyl is-H,
Figure BDA0002393819090000128
-CN, -OH, phenyl, halogen, C1-C8 alkoxy, C3-C8 cycloalkyl, C1-C8 carbonyl or substituted or unsubstituted 3-to 6-membered heterocycloalkyl; the heteroatom of the 3-6 membered heterocycloalkyl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted 3-to 6-membered heterocycloalkyl is-H, -OH, halogen, C1-C8 oxycarbonyl, C1-C8 alkyl, C1-C8 alkoxy or
Figure BDA0002393819090000131
R 4 is-H, -OH, halogen, C1-C8 alkyl, C1-C8 alkoxy or
Figure BDA0002393819090000132
R 5 ~R 10 Independently is-H or C1-C8 alkyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C10 alkyl.
Preferably, X is N or C; r 1 is-H, -OH, halogen, C1-C8 alkoxy, C1-C8 haloalkoxy,
Figure BDA0002393819090000133
C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 cycloalkyl,
Figure BDA0002393819090000134
Or substituted or unsubstituted C1-C8 alkyl; the substituent of the substituted C1-C8 alkyl is-H,
Figure BDA0002393819090000135
-CN, -OH, phenyl, halogen, C1-C6 alkoxy, C3-C6 cycloalkyl, C1-C6 carbonyl or substituted or unsubstituted 3-to 6-membered heterocycloalkyl; the heteroatom of the 3-6 membered heterocycloalkyl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted 3-to 6-membered heterocycloalkyl is-H, -OH, halogen, C1-C6 oxycarbonyl, C1-C6 alkyl, C1-C6 alkoxy or
Figure BDA0002393819090000136
R 4 is-H, -OH, halogen, C1-C6 alkyl, C1-C6 alkoxy or
Figure BDA0002393819090000137
R 5 ~R 10 Independently is-H or C1-C6 alkyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C8 alkyl.
Further preferably, X is N or C; r 1 is-H, -OH, halogen, C1-C8 alkyl, C1-C6 alkoxy, C1-C6 haloalkoxy,
Figure BDA0002393819090000138
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl,
Figure BDA0002393819090000139
Or substituted C1-C6 alkyl; the substituent of the substituted C1-C6 alkyl is-H,
Figure BDA00023938190900001310
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 3-to 6-membered heterocycloalkyl; the heteroatom of the 3-6 membered heterocycloalkyl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted 3-to 6-membered heterocycloalkyl is-H, -OH, halogen, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA00023938190900001311
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA00023938190900001312
R 5 ~R 10 Independently is-H or C1-C4 alkyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C6 alkyl.
Further, X is N or C; r 1 is-H, C1-C8 alkyl,
Figure BDA00023938190900001313
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000141
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 5-to 6-membered heterocycloalkyl; the heteroatom of the 5-6 membered heterocycloalkyl is N or O, and the number of the heteroatoms is 1-2; the substituent for substituting the 5-to 6-membered heterocycloalkyl is-H, C1-C4 oxycarbonyl, C1-C4 alkylC1-C4 alkoxy or
Figure BDA0002393819090000142
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000143
R 5 ~R 10 Independently is-H or C1-C4 alkyl;
R 3 is-H, -OH, halogen or C1-C4 alkyl.
Further, X is N or C; r 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000144
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000145
Figure BDA0002393819090000146
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl,
Figure BDA0002393819090000147
Figure BDA0002393819090000148
R 4 is-H, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000149
R 5 ~R 10 Independently is-H or C1-C4 alkyl; r 11 is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA00023938190900001410
R 3 is-H, -OH or halogen.
More preferably, X is N or C; r is 1 is-H, C1-C8 alkyl,
Figure BDA00023938190900001411
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA00023938190900001412
-CN, -OH, phenyl, halogen methoxy, ethoxy,
Figure BDA00023938190900001413
Formyl, acetyl,
Figure BDA00023938190900001414
Figure BDA00023938190900001415
R 4 is-H, C1-C4 alkyl, tert-butyloxy or
Figure BDA00023938190900001416
R 5 ~R 10 Independently is-H, methyl or ethyl;
R 3 is-H or halogen.
Most preferably, X is N or C; r 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000151
T-butyloxycarbonyl group, C2-C6 alkenyl group, C2-C6 alkynyl group,
Figure BDA0002393819090000152
Figure BDA0002393819090000153
R 3 is-H, -F, -Cl or-Br.
As a preferred embodiment of the present invention, the above 4-amino-pyrimidoazepine-phenylurea derivative has the structure wherein R is 2 Is composed of
Figure BDA0002393819090000154
R 3 When the structure is-H, the structure is shown as formula III:
Figure BDA0002393819090000155
wherein X is N or C; r 1 is-H, -OH, halogen, C1-C10 alkoxy, C1-C10 haloalkoxy,
Figure BDA0002393819090000156
C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl,
Figure BDA0002393819090000157
Or substituted or unsubstituted C1-C10 alkyl; the substituent of the substituted C1-C10 alkyl is-H,
Figure BDA0002393819090000158
-CN, -OH, phenyl, halogen, C1-C8 alkoxy, C3-C8 cycloalkyl, C1-C8 carbonyl or substituted or unsubstituted 3-to 6-membered heterocycloalkyl; the heteroatom of the 3-6 membered heterocycloalkyl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted 3-to 6-membered heterocycloalkyl is-H, -OH, halogen, C1-C8 oxycarbonyl, C1-C8 alkyl, C1-C8 alkoxy or
Figure BDA0002393819090000159
R 4 is-H, -OH, halogen, C1-C8 alkyl, C1-C8 alkoxy or
Figure BDA0002393819090000161
R 5 ~R 10 Independently is-H or C1-C8 alkyl.
Preferably, X is N or C; r 1 is-H, -OH, halogen, C1-C8 alkoxy, C1-C8 haloalkoxy,
Figure BDA0002393819090000162
C2-C8 alkenyl, C2-C8 alkynyl, C3-C8 ringAn alkyl group,
Figure BDA0002393819090000163
Or substituted or unsubstituted C1-C8 alkyl; the substituent of the substituted C1-C8 alkyl is-H,
Figure BDA0002393819090000164
-CN, -OH, phenyl, halogen, C1-C6 alkoxy, C3-C6 cycloalkyl, C1-C6 carbonyl or substituted or unsubstituted 3-to 6-membered heterocycloalkyl; the heteroatom of the 3-6 membered heterocycloalkyl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted 3-to 6-membered heterocycloalkyl is-H, -OH, halogen, C1-C6 oxycarbonyl, C1-C6 alkyl, C1-C6 alkoxy or
Figure BDA0002393819090000165
R 4 is-H, -OH, halogen, C1-C6 alkyl, C1-C6 alkoxy or
Figure BDA0002393819090000166
R 5 ~R 10 Independently is-H or C1-C6 alkyl.
Further preferably, X is N or C; r 1 is-H, -OH, halogen, C1-C8 alkyl, C1-C6 alkoxy, C1-C6 haloalkoxy,
Figure BDA0002393819090000167
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl,
Figure BDA0002393819090000168
Or substituted C1-C6 alkyl; the substituent of the substituted C1-C6 alkyl is-H,
Figure BDA0002393819090000169
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 3-to 6-membered heterocycloalkyl; the heteroatom of the 3-6 membered heterocycloalkyl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted 3-to 6-membered heterocycloalkyl is-H, -OH and halogenAn element, a C1-C4 oxycarbonyl group, a C1-C4 alkyl group, a C1-C4 alkoxy group or
Figure BDA00023938190900001610
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA00023938190900001611
R 5 ~R 10 Independently is-H or C1-C4 alkyl.
Further, X is N or C; r 1 is-H, C1-C8 alkyl,
Figure BDA00023938190900001612
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA00023938190900001613
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl or substituted or unsubstituted 5-to 6-membered heterocycloalkyl; the heteroatom of the 5-6 membered heterocycloalkyl is N or O, and the number of the heteroatoms is 1-2; the substituent for substituting the 5-to 6-membered heterocycloalkyl is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000171
R 4 is-H, -OH, halogen, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000172
R 5 ~R 10 Independently is-H or C1-C4 alkyl.
Further, X is N or C; r 1 is-H, C1-C8 alkyl,
Figure BDA0002393819090000173
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA0002393819090000174
Figure BDA0002393819090000175
-CN, -OH, phenyl, halogen, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl,
Figure BDA0002393819090000176
Figure BDA0002393819090000177
R 4 is-H, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000178
R 5 ~R 10 Independently is-H or C1-C4 alkyl; r 11 is-H, C1-C4 oxycarbonyl, C1-C4 alkyl, C1-C4 alkoxy or
Figure BDA0002393819090000179
More preferably, X is N or C; r is 1 is-H, C1-C8 alkyl,
Figure BDA00023938190900001710
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is-H,
Figure BDA00023938190900001711
Figure BDA00023938190900001712
-CN, -OH, phenyl, halogen methoxy, ethoxy,
Figure BDA00023938190900001713
Formyl, acetyl,
Figure BDA00023938190900001714
Figure BDA00023938190900001715
R 4 is-H, C1-C4 alkyl, tert-butyloxy or
Figure BDA00023938190900001716
R 5 ~R 10 independently-H, methyl or ethyl.
Most preferably, X is N or C; r 1 is-H, C1-C8 alkyl,
Figure BDA00023938190900001717
T-butyloxycarbonyl group, C2-C6 alkenyl group, C2-C6 alkynyl group,
Figure BDA00023938190900001718
Figure BDA00023938190900001719
Figure BDA0002393819090000181
As a preferred embodiment of the present invention, when R is the above-mentioned 4-amino-pyrimidoazepine-phenylurea derivative 2 Is composed of
Figure BDA0002393819090000182
R 3 is-H, X is C, R 1 Is composed of
Figure BDA0002393819090000183
Then, the structure is shown as formula IV:
Figure BDA0002393819090000184
wherein R is 16 ~R 19 Independently is-H, halogen, -NH 2 C1-C4 alkyl, C3-C6 cycloalkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, -OH or C1-C4 carbonyl.
Preferably, R 16 ~R 19 Independently is-H, halogen, -NH 2 C1-C4 alkyl, C1-C4 haloalkyl, -OH or C1-C4 carbonyl.
Most preferably, R 16 ~R 19 Independently is-H, -F, -Cl, -Br, -CF 3 Methyl or acetyl.
The 4-amino-pyrimidoazenitrogen heterocycle-phenylurea derivative has the following structural formula:
Figure BDA0002393819090000185
Figure BDA0002393819090000191
Figure BDA0002393819090000201
Figure BDA0002393819090000211
Figure BDA0002393819090000221
the invention also provides a synthesis method of the 4-amino-pyrimidoazenitrogen heterocycle-phenylurea derivative, which mainly adopts the following synthesis route:
Figure BDA0002393819090000222
the synthesis method of the compound comprises the following steps: the structure of the compound of the general formula (I) is divided into a part A and a part B, wherein the part A is an amine intermediate (formula V) compound, and the part B is an active urea intermediate (formula VI) compound or an isocyanate intermediate.
Figure BDA0002393819090000223
The first scheme is as follows: the synthesis method of the part A amine intermediate (formula V) compound is as follows:
Figure BDA0002393819090000231
the synthesis method of the pyrimido pyrrole intermediate is shown as the following scheme:
Figure BDA0002393819090000232
step a: ammoniation: taking 4-chloro-pyrrolopyrimidine (commercially available) as a starting material SM1, adding ammonia water (g/mL is approximately equal to 5-7 times) into a high-pressure kettle under the pressure of 10-15 atmospheric pressures, reacting for 4 hours, cooling, filtering to obtain an intermediate M1, and drying without further purification;
step b: iodination: dissolving the intermediate M1 in a solvent at room temperature, adding N-iodosuccinimide (NIS) (1.5 times of equivalent) to react for 1 hour, concentrating the solution, adding 30-40 times of water to pulp, adding 0.5 equivalent of sodium thiosulfate, filtering to obtain an intermediate M2, and drying without further purification; the solvent is tetrahydrofuran, DMF or acetonitrile, etc.;
step c: and (4) Boc protection: dissolving the intermediate M2 in a solvent, adding 2 equivalents of organic base, adding 1 equivalent of Boc anhydride (such as di-tert-butyl dicarbonate) and reacting at 50-60 ℃ for 2 hours, concentrating the reaction solution after the reaction is finished, pulping with 30-40 times of water, filtering to obtain an intermediate M3, and drying without further purification; the solvent is acetonitrile, DMF or dioxane, etc.; the organic base is triethylamine, DIEA or pyridine and the like;
step d: suzuki reaction: mixing the intermediate M3 with boric acid/boric acid pinacol ester, alkali, catalyst and the like, adding a solvent, reacting under the protection of nitrogen at the reaction temperature of 70-90 ℃ for 2-4 hours, treating, and simplifyingObtaining an intermediate M4 after single treatment, and further purifying; the alkali is inorganic alkali such as potassium carbonate, sodium carbonate, potassium bicarbonate or sodium bicarbonate; the catalyst is (dppf) PdCl 2 Or tetrakis (triphenylphosphine) palladium; the solvent is a mixed solvent of dioxane, ethanol and water or a mixed solvent of toluene, ethanol and water;
step e: removing Boc: dissolving the intermediate M4 in a solvent at room temperature, adding strong acid, adjusting the pH of the system to be about 1, adjusting the pH to be about 13 by using inorganic base after the reaction is finished, extracting by using dichloromethane, concentrating an organic phase, pulping by using diethyl ether, filtering to obtain an intermediate M5, and drying without further purification; the solvent is water or ethanol and the like; the strong acid is inorganic strong acid such as sulfuric acid, methanesulfonic acid or hydrochloric acid; the inorganic alkali is potassium hydroxide or sodium hydroxide and the like;
step f: and (3) substitution: dissolving the intermediate M5 in a solvent, adding 1.2 equivalents of halide, reacting at 70-80 ℃ for 1 hour, filtering the reaction solution to remove inorganic salts to obtain a solution of the intermediate M6 (the compound of the formula V), and carrying out no further purification; the solvent is acetonitrile, toluene, dioxane or DMF.
The synthesis method of the part B amine intermediate (formula VI) compound is as follows:
Figure BDA0002393819090000241
dissolving triphosgene in a solvent, adding 1 equivalent of amine raw material, adding organic base, reacting at 60-65 ℃ for 4-5 hours, filtering to remove solid residues after the reaction is finished, and concentrating the filtrate to obtain an intermediate (a compound shown in a formula VI); the solvent is tetrahydrofuran and the like; the organic base is triethylamine or DIEA and the like.
Figure BDA0002393819090000242
Then, dissolving the intermediate compound (formula V) in a solvent, adding 1 equivalent of the intermediate compound (formula VI), reacting at 70-80 ℃ for 1 hour, precipitating a large amount of solid, and filtering to obtain a filter cake, namely the final urea compound, namely the compound of the general formula (I); the solvent is acetonitrile, dioxane, toluene or DMF, etc.;
wherein X is N or C; r 1 is-H, -OH, halogen, C1-C10 alkoxy, C1-C10 haloalkoxy,
Figure BDA0002393819090000243
C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl,
Figure BDA0002393819090000244
Or substituted or unsubstituted C1-C10 alkyl; the substituent of the substituted C1-C10 alkyl is-H,
Figure BDA0002393819090000245
-CN, -OH, phenyl, halogen, C1-C8 alkoxy, C3-C8 cycloalkyl, C1-C8 carbonyl or substituted or unsubstituted 3-to 6-membered heterocycloalkyl; the heteroatom of the 3-6 membered heterocycloalkyl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted 3-to 6-membered heterocycloalkyl is-H, -OH, halogen, C1-C8 oxycarbonyl, C1-C8 alkyl, C1-C8 alkoxy or
Figure BDA0002393819090000246
R 4 is-H, -OH, halogen, C1-C8 alkyl, C1-C8 alkoxy or
Figure BDA0002393819090000247
R 5 ~R 10 Independently is-H or C1-C8 alkyl;
R 2 is-H, -OH, halogen, C1-C10 alkyl, C3-C10 cycloalkyl, C1-C10 haloalkyl, amino-substituted C1-C10 alkyl, benzyl, substituted or unsubstituted C5-C10 aryl or substituted or unsubstituted 5-to 10-membered heteroaryl; the heteroatom of the 5-to 10-membered heteroaryl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted C5-C10 aryl or 5-10 membered heteroaryl is-H, halogen or-NH 2 C1-C8 alkyl, C3-C8 cycloalkyl, C1-C8 haloalkylC1-C8 alkoxy, C1-C8 haloalkoxy, -OH or C1-C8 carbonyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C10 alkyl.
Scheme II:
Figure BDA0002393819090000251
the synthesis of the intermediate compound (formula V) is the same as the first scheme, the intermediate compound (formula V) is dissolved in a solvent, 1 equivalent of isocyanate intermediate is added, the reaction is carried out for 1 hour at 70-80 ℃, a large amount of solid is separated out, and a filter cake after filtration is the final urea compound, namely the compound of the general formula (I); the solvent is acetonitrile, dioxane, toluene or DMF, etc.;
wherein X is N or C; r 1 is-H, -OH, halogen, C1-C10 alkoxy, C1-C10 haloalkoxy,
Figure BDA0002393819090000252
C2-C10 alkenyl, C2-C10 alkynyl, C3-C10 cycloalkyl,
Figure BDA0002393819090000253
Or substituted or unsubstituted C1-C10 alkyl; the substituent of the substituted C1-C10 alkyl is-H,
Figure BDA0002393819090000254
-CN, -OH, phenyl, halogen, C1-C8 alkoxy, C3-C8 cycloalkyl, C1-C8 carbonyl or substituted or unsubstituted 3-to 6-membered heterocycloalkyl; the heteroatom of the 3-6 membered heterocycloalkyl is N, O or S, and the number of the heteroatoms is 1-3; the substituent of the substituted 3-to 6-membered heterocycloalkyl is-H, -OH, halogen, C1-C8 oxycarbonyl, C1-C8 alkyl, C1-C8 alkoxy or
Figure BDA0002393819090000255
R 4 is-H, -OH, halogen, C1-C8 alkyl, C1-C8 alkoxy or
Figure BDA0002393819090000256
R 5 ~R 10 Independently is-H or C1-C8 alkyl;
R 2 is-H, -OH, halogen, C1-C10 alkyl, C3-C10 cycloalkyl, C1-C10 haloalkyl, amino-substituted C1-C10 alkyl, benzyl, substituted or unsubstituted C5-C10 aryl or substituted or unsubstituted 5-to 10-membered heteroaryl; the heteroatom of the 5-to 10-membered heteroaryl is N, O or S, and the number of the heteroatoms is 1 to 3; the substituent of the substituted C5-C10 aryl or 5-10 membered heteroaryl is-H, halogen or-NH 2 C1-C8 alkyl, C3-C8 cycloalkyl, C1-C8 haloalkyl, C1-C8 alkoxy, C1-C8 haloalkoxy, -OH or C1-C8 carbonyl;
R 3 is-H, -OH, halogen, -NH 2 Or C1-C10 alkyl.
The invention also provides the 4-amino-pyrimidoazenitrogen heterocyclic-phenylurea derivative, including tautomers, stereoisomers, mixtures of all proportions thereof and isotopically substituted compounds thereof.
The invention also provides pharmaceutically acceptable salts of the 4-amino-pyrimidoazepine-phenylurea derivatives. Wherein the salt with an acid is obtained by reacting the free base of the parent compound with an inorganic or organic acid. The inorganic acid includes hydrochloric acid, hydrobromic acid, nitric acid, phosphoric acid, metaphosphoric acid, sulfuric acid, sulfurous acid, perchloric acid and the like. The organic acid includes acetic acid, propionic acid, acrylic acid, oxalic acid, (D) or (L) malic acid, fumaric acid, maleic acid, hydroxybenzoic acid, γ -hydroxybutyric acid, methoxybenzoic acid, phthalic acid, methanesulfonic acid, ethanesulfonic acid, naphthalene-1-sulfonic acid, naphthalene-2-sulfonic acid, p-toluenesulfonic acid, salicylic acid, tartaric acid, citric acid, lactic acid, mandelic acid, succinic acid, malonic acid, or the like.
The term "pharmaceutically acceptable" as used herein, means that which, within the scope of sound medical judgment, is suitable for use in contact with the tissues of human beings and other mammals without undue toxicity, irritation, allergic response and the like, and which, when administered to a recipient, provides, directly or indirectly, a compound of the invention or a prodrug of the compound.
The invention also provides pharmaceutically acceptable hydrates of the 4-amino-pyrimidoazenitrogen heterocyclic-phenylurea derivatives. The term "hydrate" refers to a compound that further binds stoichiometric or non-stoichiometric water by non-covalent intermolecular forces.
The invention also provides pharmaceutically acceptable polymorphic substances of the 4-amino-pyrimidoazepine-phenylurea derivatives. The term "polymorph" denotes a solid crystalline form of a compound or a complex thereof, which can be characterized by physical means, such as x-ray powder diffraction patterns or infrared spectroscopy.
The invention also provides a pharmaceutically acceptable pharmaceutical composition of the 4-amino-pyrimidoazepine-phenylurea derivative, which is prepared by adding pharmaceutically acceptable auxiliary components into the 4-amino-pyrimidoazepine-phenylurea derivative shown in formulas I to IV or salt or hydrate thereof. The auxiliary component is cyclodextrin, arginine or meglumine. The cyclodextrin is selected from alpha-cyclodextrin, beta-cyclodextrin, gamma-cyclodextrin and (C) 1-4 Alkyl) -alpha-cyclodextrin, (C) 1-4 Alkyl) -beta-cyclodextrin, (C) 1-4 Alkyl) -gamma-cyclodextrin, (hydroxy-C) 1-4 Alkyl) -alpha-cyclodextrin, (hydroxy-C) 1-4 Alkyl) -beta-cyclodextrin, (hydroxy-C) 1-4 Alkyl) -gamma-cyclodextrin, (carboxy-C) 1-4 Alkyl) -alpha-cyclodextrin, (carboxy-C) 1-4 Alkyl) -beta-cyclodextrin, (carboxy-C) 1-4 Alkyl) -gamma-cyclodextrin, saccharide ethers of alpha-cyclodextrin, saccharide ethers of beta-cyclodextrin, saccharide ethers of gamma-cyclodextrin, sulfobutyl ethers of alpha-cyclodextrin, sulfobutyl ethers of beta-cyclodextrin and sulfobutyl ethers of gamma-cyclodextrin. The auxiliary components also comprise a pharmaceutically acceptable carrier, adjuvant or vehicle. Can be used in pharmaceutically acceptable pharmaceutical composition, such as ion exchanger, aluminum oxide, aluminum stearate, and lecithin; buffer substances include phosphate, glycine, arginine, sorbic acid, and the like.
The pharmaceutical composition may be in liquid form or solid form. Wherein the liquid form may be an aqueous solution. The solid form may be in the form of a powder, granules, tablets or lyophilized powder. The pharmaceutical composition further comprises water for injection, saline solution, aqueous glucose solution, saline for injection/infusion, glucose for injection/infusion, grignard solution or Grignard solution containing lactate.
The invention also provides application of the 4-amino-pyrimidoazepine-phenylurea derivatives, salts, hydrates or pharmaceutical compositions shown in the formulas I to IV in preparation of FLT3 kinase inhibitors; in particular a mutant FLT3 kinase; especially FLT3/ITD mutant kinase.
The invention also provides application of the 4-amino-pyrimido nitrogen heterocyclic-phenylurea derivatives, salts, hydrates or pharmaceutical compositions shown in the formulas I to IV in preparation of medicines for treating cell proliferation disorder diseases.
The invention also provides application of the 4-amino-pyrimidoazepine-phenylurea derivatives, salts, hydrates or pharmaceutical compositions shown in the formulas I to IV in preparing medicaments for treating tumors.
Further, the tumor is a solid tumor and/or a hematological tumor.
Still further, the solid tumor includes lymphoma, B-cell lymphoma, diffuse large B-cell lymphoma, chronic lymphocytic lymphoma, lymphoplasmacytic lymphoma, ovarian cancer, breast cancer, prostate cancer, bladder cancer, kidney cancer, esophageal cancer, neck cancer, pancreatic cancer, colorectal cancer, gastric cancer, non-small cell lung cancer, thyroid cancer, brain cancer, lymphatic cancer, epidermal hyperproliferation, psoriasis, and/or prostatic hyperplasia.
Still further, the hematologic neoplasm comprises: acute myeloid leukemia, chronic myeloid leukemia, myeloma, acute lymphocytic leukemia, acute myelogenous leukemia, acute promyelocytic leukemia, chronic lymphocytic leukemia, chronic neutrophilic leukemia, acute undifferentiated cell leukemia, myelodysplastic syndrome, myelodysplasia, multiple myeloma, and/or myelosarcoma.
The invention also provides application of the 4-amino-pyrimidoazenitrogen heterocycle-phenylurea derivatives, salts, hydrates or pharmaceutical compositions shown in the formulas I to IV in preparation of oral or intravenous injection preparations. The oral or intravenous injection preparation at least comprises a 4-amino-pyrimidoazepine-phenylurea derivative, salt, hydrate or pharmaceutical composition shown in formulas I-IV and any excipient and/or adjuvant.
The 4-amino-pyrimidoazepine-phenylurea derivative provided by the invention can be used as a FLT3 kinase inhibitor, has a good effect, and provides a new choice for preparing antitumor drugs.
Drawings
FIG. 1MV4-11 verifies target access;
FIG. 2Molm-13 verifies the target pathway;
FIG. 3Molm-13 in vivo pharmacodynamic model;
FIG. 4MV4-11 in vivo pharmacodynamic model;
FIG. 5 survival study of NCG mice.
Detailed Description
The present invention will be described in further detail with reference to the following specific embodiments in the form of examples, but the present invention is not limited thereto.
In the examples, the reaction temperature is, without particular indication, room temperature, i.e.20-30 ℃.
Example 1:1- (4- (4-amino-7- (2-morpholinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (4-fluorophenyl) urea compound CLJ-1
Figure BDA0002393819090000271
Step a: preparation of M1 intermediate
Raw materials SM1 (20g, 130mmol) and ammonia water (150mL, 25% -28% industrial ammonia water) are weighed and added into a high-pressure reaction kettle, the system is sealed, a thermometer is inserted, the temperature is set to be 130 ℃, and after 4 hours of reaction, the temperature is cooled to room temperature. The reaction suspension is directly filtered, and the filter cake is washed by ether to obtain an intermediate M1. 1 H NMR(400MHz,DMSO)δ:11.45(s,1H),8.03(s,1H),7.06(d,J=3.2Hz,1H),6.88(s,2H),6.51(d,J=3.3Hz,1H).
Step b: preparation of M2 intermediate
Intermediate M1 (1lg, 112mmol) from the previous step was added to 200mL of tetrahydrofuran at room temperature, and NIS (37.8g, 168mmol) was added in portions and reacted for 1h. The reaction suspension was evaporated under reduced pressure to give a solid mixture, 300mL of water was added, sodium thiosulfate (8.8g, 56mmol) was added with stirring, the mixture turned from brown to yellow, stirring was continued for 0.5h, the mixture was filtered, and the filter cake was washed with ether to give intermediate M2. 1 H NMR(400MHz,DMSO)δ:11.97(s,1H),8.07(s,1H),7.36(s,1H),6.56(s,2H).
Step c: preparation of M3 intermediate
Intermediate M2 (28g, 107.7mmol), potassium carbonate (29.7g, 215.4mmol) and Boc anhydride (25.8g, 118.5mmol) in the previous step were added to 500mL of acetonitrile, and the resulting mixture was heated to 60 ℃ and reacted for 2 hours. And filtering the reaction liquid, discarding a filter cake, distilling the mother liquor under reduced pressure to obtain a mixture solid, adding 500mL of water for pulping, continuously stirring for 0.5h, filtering, draining the filter cake, transferring to a vacuum drying oven, and drying at 60 ℃ for 5h to obtain an intermediate M3. 1 H NMR(400MHz,DMSO)δ:8.28(s,1H),7.73(s,1H),6.87(s,2H),1.62(s,10H).
Step d: preparation of M4 intermediate
Intermediate M3 (9.56g, 26.6 mmol) in the previous step, p-aminobenzoate (5.5g, 31.8mmol), potassium carbonate (7.3g, 53.1mmol) and dppf (Pd) 2 Cl 2 ) The mixture was charged into a 250mL three-necked flask, and dioxane/ethanol/water =7 (120 mL total) was added as a solvent, and after replacing nitrogen three times, the mixture was transferred into an oil bath at 80 ℃ for reaction for 2h. After the reaction is finished, concentrating the reaction solution to be dry, mixing the sample, and separating the mixture by a column to obtain an intermediate M4. 1 H NMR(400MHz,DMSO)δ:9.57(s,1H),9.02(s,1H),8.28(s,1H),7.59(d,J=8.6Hz,2H),7.47(s,1H),7.44(d,J=8.5Hz,2H),6.52(s,1H),6.23(s,2H),1.61(s,9H),1.31(s,9H).
Step e: preparation of M5 intermediate
Intermediate M4 (5g, 15.4 mmol) in the previous step was added to 50mL of methylene chloride, and concentrated hydrochloric acid (10mL, 36-38% concentrated hydrochloric acid) was added to complete the reaction for 1 hour. 200mL of methylene chloride and 100mL of water were added to adjust the pH of the mixture to basic pH (about pH 10), and the organic phase was retained by extractionAfter spin-drying, the intermediate M5 is obtained. 1 H NMR(400MHz,DMSO)δ:11.57(s,1H),8.07(s,1H),7.12(d,J=8.3Hz,2H),7.03(s,1H),6.66(d,J=8.3Hz,2H),5.94(s,2H),5.16(s,2H).
Step f: preparation of intermediate M6
The intermediate M5 (225mg, 1mmol) obtained in the previous step, cesium carbonate (650mg, 2mmol) and N- (2-chloroethyl) morpholine hydrochloride (223mg, 1.2mmol) were added to 20mL of acetonitrile, and the mixture was heated to 80 ℃ for 1 hour. The reaction solution was spin dried and extracted with dichloromethane to obtain the purer intermediate M6. 1 H NMR(400MHz,CDCl 3 ) δ 8.68 (s, 1H), 7.25 (d, J =8.4hz, 4H), 7.13 (s, 1H), 6.80 (d, J =8.4hz, 2h), 4.44 (s, 2H), 3.72 (s, 4H), 2.87 (s, 2H), 2.59 (s, 4H). Step g: preparation of the end product CLJ-1
The intermediate M6 (338mg, 1mmol) in the previous step was added to 20mL of methylene chloride, 4-fluorophenylisocyanate (137mg, 1mmol) was added, and after 0.5 hour of reaction, a solid precipitated out, and the filter cake was rinsed with diethyl ether after filtration to obtain a high-purity final product CLJ-1. 1 H NMR(400MHz,DMSO)δ:9.56(s,1H),9.47(s,1H),8.54(s,1H),7.71(s,1H),7.62(d,J=8.6Hz,2H),7.52–7.45(m,2H),7.40(d,J=8.6Hz,2H),7.16–7.10(m,2H),4.75(t,J=6.5Hz,2H),3.89(s,4H),3.65(t,J=6.4Hz,4H).HRMS(ESI),m/z:476.2214[M+H] + .
EXAMPLE 2 CLJ-2- (4- (4-amino-7- (2-morpholinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (2-chlorophenyl) urea Compound
Figure BDA0002393819090000291
The phenyl 4-fluoroisocyanate was replaced with 2-chlorophenyl isocyanate in step g in the same manner as in the synthesis of example 1 to give CLJ-2 as a final product. 1 H NMR(400MHz,DMSO)δ:11.47(s,1H),10.10(s,1H),8.59(s,1H),8.55(s,1H),8.15(dd,J=8.3,1.5Hz,1H),7.72(s,1H),7.66(d,J=8.6Hz,2H),7.46(dd,J=8.0,1.5Hz,1H),7.42(d,J=8.6Hz,2H),7.34–7.28(m,1H),7.07–7.01(m,1H),4.75(t,J=6.5Hz,2H),3.89(s,4H),3.65(t,J=6.4Hz,3H).HRMS(ESI),m/z:492.1922[M+H] + .
EXAMPLE 3 1- (4- (4-amino-7- (2-morpholinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3-benzylurea Compound CLJ-3
Figure BDA0002393819090000292
The phenyl 4-fluoroisocyanate was replaced with benzyl isocyanate in step g in the same manner as in the synthesis of example 1 to give the final product CLJ-3. 1 H NMR(400MHz,DMSO)δ:11.58(s,1H),9.31(s,1H),8.55(s,1H),7.70(s,1H),7.59(d,J=8.6Hz,2H),7.37–7.30(m,6H),7.27–7.20(m,1H),7.05(s,1H),4.75(t,J=6.5Hz,2H),4.32(s,2H),3.87(m,9H),3.65(t,J=6.4Hz,3H).HRMS(ESI),m/z:472.2455[M+H] + .
EXAMPLE 4 CLJ-4-Compound of 1- (4- (4-amino-7- (2-morpholinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (4-chlorophenyl) urea
Figure BDA0002393819090000293
The phenyl 4-fluoroisocyanate was replaced with phenyl 4-chloroisocyanate in step g in the same manner as in the synthesis of example 1 to give CLJ-4 as a final product. 1 H NMR(400MHz,DMSO)δ:8.86(s,1H),8.82(s,1H),8.14(s,1H),7.56(d,J=8.5Hz,2H),7.53–7.49(m,2H),7.38(d,J=8.5Hz,2H),7.35–7.30(m,3H),6.05(s,2H),4.28(t,J=6.5Hz,2H),3.56–3.50(m,4H),2.71(t,J=6.4Hz,2H),2.46(s,4H).HRMS(ESI),m/z:492.1913[M+H] + .
EXAMPLE 5 CLJ-5-Compound of 1- (4- (4-amino-7- (2-morpholinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (3-chlorophenyl) urea
Figure BDA0002393819090000301
The phenyl 4-fluoroisocyanate was replaced with 3-chlorophenyl isocyanate in step g in the same manner as in the synthesis of example 1 to give CLJ-5 as a final product. 1 H NMR(400MHz,DMSO)δ:9.44(s,1H),9.33(s,1H),8.22(s,1H),7.73(s,1H),7.59(d,J=8.5Hz,2H),7.43–7.35(m,3H),7.32–7.27(m,2H),7.05–6.98(m,1H),6.39(s,2H),4.49(s,2H),3.73(s,4H),2.98(s,4H).HRMS(ESI),m/z:492.1915[M+H] + .
EXAMPLE 6 CLJ-6, a 1- (4- (4-amino-7- (2-morpholinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (2-fluorophenyl) urea compound
Figure BDA0002393819090000302
In the same manner as in the synthesis method of example 1, phenyl 4-fluoroisocyanate was replaced with phenyl 2-fluoroisocyanate in step g to obtain CLJ-6 as a final product. 1 H NMR(400MHz,DMSO)δ:9.23(s,1H),8.62(s,1H),8.16(dd,J=15.3,6.0Hz,2H),7.57(d,J=8.4Hz,2H),7.39(d,J=8.4Hz,2H),7.32(s,1H),7.29–7.20(m,1H),7.15(t,J=7.6Hz,1H),7.01(dd,J=12.7,6.5Hz,1H),6.04(s,2H),4.28(t,J=6.4Hz,2H),3.54(s,4H),2.70(t,J=6.5Hz,2H),2.45(s,4H).HRMS(ESI),m/z:476.2207[M+H] + .
EXAMPLE 7 CLJ-7-4- (4-amino-7- (2-morpholinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (4-chloro-3- (trifluoromethyl) phenyl) urea Compound
Figure BDA0002393819090000311
The phenyl 4-fluoroisocyanate was replaced in step g with 4-chloro-3-trifluoromethyl-phenyl isocyanate in the same manner as the synthesis of example 1 to give the final product CLJ-7. 1 H NMR(400MHz,DMSO)δ:11.11(s,1H),9.99(s,1H),9.66(s,1H),8.52(s,1H),8.14(d,J=1.8Hz,1H),7.70(s,1H),7.66–7.59(m,3H),7.41(d,J=8.6Hz,2H),4.73(t,J=6.3Hz,2H),3.87(s,4H),3.64(m,3H),3.41–3.13(m,4H).HRMS(ESI),m/z:560.1786[M+H] + .
EXAMPLE 8 CLJ-8, a 1- (4- (4-amino-7- (2-morpholinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (p-tolyl) urea compound
Figure BDA0002393819090000312
The phenyl 4-fluoroisocyanate was replaced with p-tolylisocyanate in step g in the same manner as in the synthesis of example 1 to give the final product CLJ-8. 1 H NMR(400MHz,DMSO)δ:9.46(s,1H),9.20(s,1H),8.52(s,1H),7.69(s,1H),7.62(d,J=8.6Hz,2H),7.39(d,J=8.6Hz,2H),7.36(d,J=8.5Hz,2H),7.09(d,J=8.3Hz,2H),4.73(t,J=6.5Hz,2H),3.88(s,4H),3.64(t,J=6.1Hz,3H),3.44–3.11(m,4H),2.25(s,3H).HRMS(ESI),m/z:472.2458[M+H] + .
EXAMPLE 9 1- (4- (4-amino-7- (2-morpholinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (3-chloro-4-methylphenyl) urea Compound CLJ-9
Figure BDA0002393819090000313
The phenyl 4-fluoroisocyanate was replaced with 3-chloro-4-methyl-phenyl isocyanate in step g in the same manner as in the synthesis of example 1 to give the final product CLJ-9. 1 H NMR(400MHz,DMSO)δ:11.40(s,1H),9.66(d,J=4.5Hz,2H),8.55(s,1H),7.72(s,1H),7.71(d,J=1.9Hz,1H),7.62(d,J=8.6Hz,2H),7.40(d,J=8.6Hz,2H),7.27–7.19(m,2H),4.75(t,J=6.5Hz,2H),3.89(s,4H),3.66(t,J=6.5Hz,3H),2.26(s,3H).HRMS(ESI),m/z:506.2066[M+H] + .
EXAMPLE 10 1- (4- (4-amino-7- (2-morpholinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (3-acetylphenyl) urea Compound CLJ-10
Figure BDA0002393819090000321
The phenyl 4-fluoroisocyanate was replaced with 3-acetylphenyl isocyanate in step g in the same manner as in the synthesis of example 1 to give CLJ-10 as a final product. 1 H NMR(400MHz,DMSO)δ:11.39(s,1H),9.70(d,J=8.8Hz,2H),8.54(s,1H),8.11(t,J=1.8Hz,1H),7.73–7.68(m,2H),7.65(d,J=8.6Hz,2H),7.61–7.57(m,1H),7.48–7.42(m,1H),7.41(d,J=8.6Hz,2H),4.75(t,J=6.5Hz,2H),3.89(s,4H),3.66(t,J=6.5Hz,2H),2.57(s,3H).HRMS(ESI),m/z:500.2406[M+H] + .
EXAMPLE 11 1- (4- (4-amino-7- (2-morpholinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3-cyclohexylurea Compound CLJ-11
Figure BDA0002393819090000322
The phenyl 4-fluoroisocyanate was replaced with cyclohexyl isocyanate in step g in the same manner as in the synthesis of example 1 to give the final product CLJ-11. 1 H NMR(400MHz,DMSO)δ:8.38(s,1H),8.12(s,1H),7.47(d,J=8.6Hz,2H),7.30(d,J=8.6Hz,2H),7.28(s,1H),6.09(d,J=7.9Hz,1H),4.27(t,J=6.6Hz,2H),3.56–3.51(m,4H),2.69(t,J=6.6Hz,2H),2.45(d,J=4.1Hz,4H),1.81(dd,J=8.5,3.9Hz,2H),1.67(dd,J=9.0,4.0Hz,2H),1.59–1.49(m,1H),1.38–1.25(m,2H),1.24–1.12(m,4H).HRMS(ESI),m/z:464.2773[M+H] + .
EXAMPLE 12 CLJ-12, a 1- (4- (4-amino-7- (2-morpholinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (4-bromophenyl) urea compound
Figure BDA0002393819090000331
The phenyl 4-fluoroisocyanate was replaced in step g with 4-bromobenzene isocyanate in the same manner as the synthesis of example 1 to give the final product CLJ-12. 1 H NMR(400MHz,DMSO)δ:8.86(s,1H),8.82(s,1H),8.14(s,1H),7.56(d,J=8.5Hz,2H),7.53–7.49(m,2H),7.38(d,J=8.5Hz,2H),7.35–7.30(m,3H),6.05(s,2H),4.28(t,J=6.5Hz,2H),3.56–3.50(m,4H),2.71(t,J=6.4Hz,2H),2.46(s,4H).HRMS(ESI),m/z:536.1408,538.1390[M+H] + .
EXAMPLE 13 CLJ-13- (4- (4-amino-7- (2-morpholinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound
Figure BDA0002393819090000332
Preparation of reactive urea intermediate 1, 3-bis (5- (tert-butyl) isoxazol-3-yl) urea
Triphosgene (47.07g, 156.9 mmol) was added to 300mL of tetrahydrofuran, and 3-amino-5-tert-butylisoxazole (20g, 142.65mmol) was dissolved in 100mL of tetrahydrofuran under ice bath, and then added dropwise to the triphosgene solution, and finally triethylamine (39.8mL, 285.3mmol) was added dropwise. The reaction was transferred to a 60 ℃ oil bath and reacted for 5h. After the reaction is completed, the reaction mixture is filtered, the filtrate is retained, and the filtrate is concentrated under reduced pressure to obtain a solid, namely the active urea intermediate. 1 H NMR(400MHz,DMSO)δ:6.71(s,1H),6.35(s,1H),4.90(s,2H),1.36(s,9H),1.35(s,9H).
Combining steps f and h into a one-pot reaction process
The intermediate M5 (225mg, 1mmol) obtained in the previous step, cesium carbonate (650mg, 2mmol) and N- (2-chloroethyl) morpholine hydrochloride (223mg, 1.2mmol) were added to 20mL of acetonitrile, and the mixture was heated to 80 ℃ to react for 1 hour. The reaction solution was filtered, and the mother liquor was retained. Transferring the mother liquor to 80 ℃, adding an active urea intermediate (306mg, 1mmol), reacting for 0.5h, separating out a large amount of solid, filtering, and leaching with diethyl ether to obtain the high-purity final product CLJ-13. 1 H NMR(400MHz,DMSO)δ:9.53(s,1H),8.91(s,1H),8.14(s,1H),7.56(d,J=8.3Hz,2H),7.40(d,J=8.2Hz,2H),7.33(s,1H),6.52(s,1H),6.06(s,2H),4.29(t,J=6.3Hz,2H),3.54(s,4H),2.71(t,J=6.4Hz,2H),2.46(s,4H),1.31(s,9H).HRMS(ESI),m/z:505.2672[M+H] + .
Example 14 1- (4- (4-amino-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-14
Figure BDA0002393819090000341
Adding the intermediate M5 (225mg, 1mmol) in the previous step into 20mL acetonitrile, heating to 80 ℃ for reaction, adding an active urea intermediate (306mg, 1mmol), reacting for 0.5h, precipitating a large amount of solid, filtering, and leaching with diethyl ether to obtain the high-purity intermediate M5Degree of final product CLJ-14. 1 H NMR(400MHz,DMSO)δ:11.74(s,1H),9.53(s,1H),8.93(s,1H),8.11(s,1H),7.55(d,J=8.5Hz,2H),7.40(d,J=8.5Hz,2H),7.19(d,J=2.3Hz,1H),6.52(s,1H),5.99(s,2H),1.31(s,9H).HRMS(ESI),m/z:392.1831[M+H] + .
EXAMPLE 15 CLJ-15-1- (4- (4-amino-7-tert-butoxycarbonyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound
Figure BDA0002393819090000342
And adding the intermediate M4 (325mg, 1mmol) in the last step into 20mL of acetonitrile, heating to 80 ℃ for reaction, adding an active urea intermediate (306mg, 1mmol), reacting for 0.5h, precipitating a large amount of solid, filtering, and leaching with diethyl ether to obtain the high-purity final product CLJ-15. 1 H NMR(400MHz,DMSO)δ:9.57(s,1H),9.02(s,1H),8.28(s,1H),7.59(d,J=8.6Hz,2H),7.47(s,1H),7.44(d,J=8.5Hz,2H),6.52(s,1H),6.23(s,2H),1.61(s,9H),1.31(s,9H).HRMS(ESI),m/z:592.2353[M+H] + .
EXAMPLE 16 tert-butyl 4- (2- (4-amino-5- (4- (3- (5- (tert-butyl) isoxazol-3-yl) ureido) phenyl) -7H-pyrrolo [2,3-d ] tert-butylpyrimidin-7-yl) ethyl) piperazine-1-carboxylate Compound CLJ-16
Figure BDA0002393819090000351
In the same manner as in example 13, N-Boc-2-ethylpiperazine was used in place of N- (2-chloroethyl) morpholine hydrochloride, to give CLJ-16 as a highly pure final product. 1 H NMR(400MHz,DMSO)δ:9.53(s,1H),8.91(s,1H),8.14(s,1H),7.56(d,J=8.5Hz,2H),7.39(d,J=8.5Hz,2H),7.33(s,1H),6.52(s,1H),6.06(s,2H),4.29(t,J=6.5Hz,2H),3.28(s,4H),2.73(t,J=6.5Hz,2H),2.46–2.37(m,4H),1.39(s,9H),1.31(s,9H).HRMS(ESI),m/z:604.3353[M+H] + .
EXAMPLE 17 CLJ-17- (4- (4-amino-7- (2-piperazinoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound
Figure BDA0002393819090000352
The compound CLJ-16 (252mg, 0.5mmol) was added to 20mL of methylene chloride, and concentrated hydrochloric acid (10mL, 36-38% concentrated hydrochloric acid) was added to complete the reaction after 1 hour. 200mL of methylene chloride and 100mL of water were added to adjust the pH of the mixture to basic pH (about 10), the organic phase was retained by extraction, and the final product, CLJ-17, was obtained after concentration. 1 H NMR(400MHz,DMSO)δ:9.58(s,1H),9.01(s,1H),8.14(s,1H),7.57(d,J=8.5Hz,2H),7.39(d,J=8.5Hz,2H),7.33(s,1H),6.52(s,1H),6.05(s,2H),4.27(t,J=6.6Hz,2H),2.75–2.63(m,6H),2.41(s,4H),1.31(s,9H).HRMS(ESI),m/z:504.2829[M+H] + .
EXAMPLE 18 CLJ-18- (4- (4-amino-7- (3-morpholinopropyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound
Figure BDA0002393819090000353
In the same manner as in example 13, high-purity CLJ-18 was obtained by substituting N- (2-chloroethyl) morpholine hydrochloride with N- (3-chloropropyl) morpholine. 1 H NMR(400MHz,DMSO)δ:9.53(s,1H),8.90(s,1H),8.14(s,1H),7.56(d,J=8.6Hz,2H),7.40(d,J=8.6Hz,2H),7.31(s,1H),6.52(s,1H),6.05(s,2H),4.20(t,J=7.0Hz,2H),3.59–3.51(m,4H),2.29(dd,J=14.3,7.0Hz,6H),2.02–1.91(m,2H),1.31(s,9H).HRMS(ESI),m/z:519.2825[M+H] + .
EXAMPLE 19 1- (4- (4-amino-7- (2- (dimethylamino) ethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-19
Figure BDA0002393819090000361
In the same manner as in the synthesis of example 13, N-dimethylThe N- (2-chloroethyl) morpholine hydrochloride is replaced by the-2-chloroethyl, and the high-purity final product CLJ-19 can be obtained. 1 H NMR(400MHz,DMSO)δ:9.54(s,1H),8.93(s,1H),8.14(s,1H),7.56(d,J=8.3Hz,2H),7.39(d,J=8.3Hz,2H),7.33(s,1H),6.52(s,1H),6.05(s,2H),4.26(t,J=6.6Hz,2H),2.67(t,J=6.5Hz,2H),2.20(s,6H),1.31(s,9H).HRMS(ESI),m/z:463.2563[M+H] + .
EXAMPLE 20 1- (4- (4-amino-7-allyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-20
Figure BDA0002393819090000362
The high purity final product CLJ-20 can be obtained by replacing N- (2-chloroethyl) morpholine hydrochloride with bromopropene in the same way as the synthesis method of the embodiment 13. 1 H NMR(400MHz,DMSO)δ:9.53(s,1H),8.91(s,1H),8.15(s,1H),7.56(d,J=8.5Hz,2H),7.40(d,J=8.4Hz,2H),7.25(s,1H),6.52(s,1H),6.06(m,3H),5.18(d,J=10.1Hz,1H),5.09(d,J=17.1Hz,1H),4.81(d,J=5.4Hz,2H),1.31(s,9H).HRMS(ESI),m/z:432.2149[M+H] + .
Example 21- (4- (4-amino-7- (cyanomethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-21
Figure BDA0002393819090000363
High purity final product CLJ-21 was obtained by substituting chloroacetonitrile for N- (2-chloroethyl) morpholine hydrochloride in the same manner as in example 13. 1 H NMR(400MHz,DMSO)δ:9.54(s,1H),8.94(s,1H),8.24(s,1H),7.58(d,J=7.4Hz,2H),7.42(d,J=7.6Hz,2H),7.38(s,1H),6.53(s,1H),6.26(s,2H),5.41(s,2H),1.31(s,9H).HRMS(ESI),m/z:431.1937[M+H] + .
EXAMPLE 22 1- (4- (4-amino-7-isopropyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-22
Figure BDA0002393819090000371
In the same manner as in example 13, high-purity CLJ-22 was obtained by substituting 2-iodopropane for N- (2-chloroethyl) morpholine hydrochloride. 1 H NMR(400MHz,DMSO)δ:9.55(s,1H),8.97(s,1H),8.33(s,1H),7.65(s,1H),7.59(d,J=8.6Hz,2H),7.43(d,J=8.5Hz,2H),6.52(s,1H),5.10–4.93(m,1H),1.49(d,J=6.8Hz,6H),1.31(s,9H).HRMS(ESI),m/z:434.2299[M+H] + .
EXAMPLE 23 1- (4- (4-amino-7- (4-hydroxybutyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-23
Figure BDA0002393819090000372
In the same manner as in example 13, 4-iodobutanol was used in place of N- (2-chloroethyl) morpholine hydrochloride to give high purity CLJ-23. 1 H NMR(400MHz,DMSO)δ:9.53(s,1H),8.91(s,1H),8.14(s,1H),7.56(d,J=8.4Hz,2H),7.40(d,J=8.4Hz,2H),7.32(s,1H),6.52(s,1H),6.05(s,2H),4.41(t,J=5.2Hz,1H),4.17(t,J=7.0Hz,2H),4.09(q,J=5.2Hz,1H),3.41(dd,J=11.8,6.2Hz,2H),3.18(d,J=5.2Hz,2H),1.88–1.76(m,2H),1.46–1.36(m,2H),1.31(s,9H).HRMS(ESI),m/z:464.2409[M+H] + .
EXAMPLE 24 CLJ-24-1- (4- (4-amino-7- (N, N-diethylformyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound
Figure BDA0002393819090000373
In the same manner as in example 13, N-diethylchloroformamide was used in place of N- (2-chloroethyl) morpholine hydrochloride to obtain high-purity CLJ-24. 1 H NMR(400MHz,DMSO)δ:9.55(s,1H),8.96(s,1H),8.20(s,1H),7.58(d,J=8.6Hz,2H),7.44(d,J=8.5Hz,2H),7.41(s,1H),6.53(s,1H),6.25(s,2H),3.39(m,4H),1.31(s,9H),1.21–1.06(m,6H).HRMS(ESI),m/z:491.2493[M+H] + .
EXAMPLE 25 CLJ-25, a 1- (4- (4-amino-7- (2- (methylsulfonyl) piperazin-1-yl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound
Figure BDA0002393819090000381
The compound CLJ-17 (252mg, 0.5 mmol) was dissolved in 20mL of dichloromethane, triethylamine (61mg, 0.6 mmol) was added, methanesulfonyl chloride (57mg, 0.5 mmol) diluted with dichloromethane was added under ice bath, after 1h of reaction, extraction was performed with dichloromethane and water, and the organic phase was spin-dried to give the purer final product CLJ-25. 1 H NMR(400MHz,DMSO)δ:9.53(s,1H),8.92(s,1H),8.15(s,1H),7.56(d,J=8.6Hz,2H),7.40(d,J=8.5Hz,2H),7.34(s,1H),6.52(s,1H),6.05(s,2H),4.30(t,J=6.4Hz,2H),3.12–3.03(m,4H),2.85(s,3H),2.79(t,J=6.5Hz,2H),2.62–2.54(m,4H),1.31(s,9H).HRMS(ESI),m/z:582.2610[M+H] + .
EXAMPLE 26 CLJ-26, a 1- (4- (4-amino-7- (2-methoxyethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound
Figure BDA0002393819090000382
In the same manner as the synthesis method in example 13, N- (2-chloroethyl) morpholine hydrochloride was replaced with chloroethyl methyl ether to obtain high-purity final product CLJ-26. 1 H NMR(400MHz,DMSO)δ:9.56(s,1H),8.95(s,1H),8.15(s,1H),7.56(d,J=8.4Hz,2H),7.39(d,J=8.4Hz,2H),7.29(s,1H),6.53(s,1H),6.09(s,2H),4.33(t,J=5.3Hz,2H),3.71(t,J=5.4Hz,2H),3.26(s,3H),1.31(s,9H).HRMS(ESI),m/z:450.2249[M+H] + .
EXAMPLE 27 CLJ-27 1- (4- (4-amino-7- (but-2-yn-1-yl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound
Figure BDA0002393819090000391
In the same manner as in example 13, 1-bromo-2-butyne was used in place of N- (2-chloroethyl) morpholine hydrochloride to give high purity final product CLJ-27. 1 H NMR(400MHz,DMSO)δ:9.55(s,1H),8.97(s,1H),8.17(s,1H),7.57(d,J=8.5Hz,2H),7.41(d,J=8.4Hz,2H),7.32(s,1H),6.53(s,1H),6.12(s,1H),4.97(d,J=2.3Hz,2H),1.81(dd,J=5.3,3.1Hz,3H),1.31(s,9H).HRMS(ESI),m/z:444.2147[M+H] + .
EXAMPLE 28 CLJ-28-1- (4- (4-amino-7- (pent-2-yn-1-yl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound
Figure BDA0002393819090000392
In the same manner as in example 13, 1-bromo-2-pentyne was substituted for N- (2-chloroethyl) morpholine hydrochloride to give high purity final product CLJ-28. 1 H NMR(400MHz,DMSO)δ:9.53(s,1H),8.93(s,1H),8.16(s,1H),7.56(d,J=8.6Hz,2H),7.40(d,J=8.5Hz,2H),7.31(s,1H),6.52(s,1H),6.12(s,2H),4.98(t,J=2.1Hz,2H),2.25–2.16(m,2H),1.30(s,9H),1.06(t,J=7.5Hz,3H).HRMS(ESI),m/z:458.2299[M+H] + .
EXAMPLE 29 CLJ-29-1- (4- (4-amino-7- (but-3-yn-1-yl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound
Figure BDA0002393819090000393
In the same manner as in example 13, 1-bromo-2-pentyne was substituted for N- (2-chloroethyl) morpholine hydrochloride to give high purity final product CLJ-29. 1 H NMR(400MHz,DMSO)δ:9.57(s,1H),8.99(s,1H),8.15(s,1H),7.56(d,J=8.4Hz,2H),7.39(d,J=8.3Hz,2H),7.32(s,1H),6.52(s,1H),6.05(s,2H),5.82(ddd,J=23.8,10.3,6.7Hz,1H),5.14–4.93(m,2H),4.24(t,J=7.0Hz,2H),2.56(dd,J=13.3,6.5Hz,2H),1.31(s,9H).HRMS(ESI),m/z:446.2302[M+H] + .
EXAMPLE 30 CLJ-30- (4- (4-amino-7- (2-ethoxyethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound
Figure BDA0002393819090000401
The same synthesis method as that in example 13 is adopted, and bromoethyl ether is used to replace N- (2-chloroethyl) morpholine hydrochloride to obtain the high-purity final product CLJ-30. 1 H NMR(400MHz,DMSO)δ:9.53(s,1H),8.92(s,1H),8.15(s,1H),7.56(d,J=8.4Hz,2H),7.39(d,J=8.4Hz,2H),7.29(s,1H),6.52(s,1H),6.06(s,2H),4.32(t,J=5.4Hz,2H),3.74(t,J=5.5Hz,2H),3.46(q,J=6.9Hz,2H),1.31(s,9H).HRMS(ESI),m/z:464.2406[M+H] + .
EXAMPLE 31 1- (4- (4-amino-7- (3-methylbut-2-en-1-yl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-31
Figure BDA0002393819090000402
In the same manner as in example 13, 3-dimethylallyl bromide was used in place of N- (2-chloroethyl) morpholine hydrochloride to give high-purity CLJ-31 as a final product. 1 H NMR(400MHz,DMSO)δ:9.53(s,1H),8.92(s,1H),8.15(s,1H),7.56(d,J=8.4Hz,2H),7.39(d,J=8.4Hz,2H),7.23(s,1H),6.52(s,1H),6.05(s,2H),5.41(t,J=6.8Hz,1H),4.76(d,J=7.0Hz,2H),1.81(s,3H),1.72(s,3H),1.31(s,9H).HRMS(ESI),m/z:460.2457[M+H] + .
EXAMPLE 32 1- (4- (4-amino-7- (2-methylallyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-32
Figure BDA0002393819090000403
In the same manner as in example 13, 2-methylallyl bromide was used in place of N- (2-chloroethyl) morpholine hydrochloride to give high-purity CLJ-32 as a final product. 1 H NMR(400MHz,DMSO)δ:9.53(s,1H),8.92(s,1H),8.15(s,1H),7.56(d,J=8.6Hz,2H),7.40(d,J=8.6Hz,2H),7.20(s,1H),6.52(s,1H),6.09(s,2H),4.87(s,1H),4.73(s,2H),4.61(s,1H),1.68(s,3H),1.31(s,9H).HRMS(ESI),m/z:446.2298[M+H] + .
EXAMPLE 33 CLJ-33-1- (4- (4-amino-7- (pent-4-en-1-yl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound
Figure BDA0002393819090000411
In the same manner as in example 13, 5-bromo-1-pentene was used in place of N- (2-chloroethyl) morpholine hydrochloride to give high-purity CLJ-33 as a final product. 1 H NMR(400MHz,DMSO)δ:9.77(s,1H),9.58(s,1H),8.48(s,1H),7.68(s,1H),7.61(d,J=8.6Hz,2H),7.42(d,J=8.6Hz,2H),6.53(s,1H),5.83(ddt,J=16.8,10.2,6.4Hz,1H),5.09–4.95(m,2H),4.27(t,J=7.0Hz,2H),2.05(dd,J=13.7,6.7Hz,2H),1.99–1.87(m,2H),1.31(s,9H).HRMS(ESI),m/z:460.2458[M+H] + .
EXAMPLE 34 CLJ-34-1- (4- (4-amino-7- (hex-5-en-1-yl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound
Figure BDA0002393819090000412
In the same manner as in example 13, high-purity CLJ-34 was obtained by substituting 5-bromo-1-pentene for N- (2-chloroethyl) morpholine hydrochloride. 1 H NMR(400MHz,DMSO)δ:9.78(s,1H),9.60(s,1H),8.48(s,1H),7.68(s,1H),7.61(d,J=8.6Hz,2H),7.41(d,J=8.6Hz,2H),6.53(s,1H),5.78(ddt,J=16.9,10.2,6.7Hz,1H),5.05–4.90(m,2H),4.27(t,J=7.0Hz,2H),2.06(q,J=7.2Hz,2H),1.83(dt,J=14.9,7.2Hz,2H),1.41–1.31(m,2H),1.31(s,9H).HRMS(ESI),m/z:474.2618[M+H] + .
EXAMPLE 35 1- (4- (4-amino-7-methyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-35
Figure BDA0002393819090000421
The same synthesis method as that in example 13 was used to replace N- (2-chloroethyl) morpholine hydrochloride with methyl iodide to obtain high purity final product CLJ-35. 1 H NMR(400MHz,DMSO)δ:9.53(s,1H),8.93(s,1H),8.11(s,1H),7.55(d,J=8.5Hz,2H),7.40(d,J=8.5Hz,2H),7.19(d,J=2.3Hz,1H),6.52(s,1H),5.99(s,2H),3.7(s,3H),1.31(s,9H).HRMS(ESI),m/z:406.1992[M+H] + .
Example 36 1- (4- (4-amino-7-ethyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-36
Figure BDA0002393819090000422
The same synthesis as in example 13 was carried out using iodoethane instead of N- (2-chloroethyl) morpholine hydrochloride to give high purity final product CLJ-36. 1 H NMR(400MHz,DMSO)δ:9.78(s,1H),9.61(s,1H),8.48(s,1H),7.69(s,1H),7.61(d,J=8.6Hz,2H),7.42(d,J=8.5Hz,2H),6.53(s,1H),4.30(q,J=7.2Hz,2H),1.43(t,J=7.2Hz,3H),1.31(s,9H).HRMS(ESI),m/z:420.2142[M+H] + .
Example 37 1- (4- (4-amino-7-propyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-37
Figure BDA0002393819090000423
High purity final product CLJ-37 was obtained by substituting iodopropane for N- (2-chloroethyl) morpholine hydrochloride in the same manner as in the synthesis of example 13. 1 H NMR(400MHz,DMSO)δ:9.80(s,1H),9.66(s,1H),8.48(s,1H),7.68(s,1H),7.61(d,J=8.5Hz,2H),7.41(d,J=8.5Hz,2H),6.53(s,1H),4.23(t,J=7.0Hz,2H),1.90–1.77(m,2H),1.31(s,9H),0.87(t,J=7.4Hz,3H).HRMS(ESI),m/z:434.2303[M+H] + .
Example 38 1- (4- (4-amino-7-butyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-38
Figure BDA0002393819090000431
High purity final product CLJ-38 was obtained by substituting N- (2-chloroethyl) morpholine hydrochloride with bromobutane in the same manner as in the synthesis of example 13. 1 H NMR(400MHz,DMSO)δ:9.78(s,1H),9.61(s,1H),8.48(s,1H),7.68(s,1H),7.61(d,J=8.6Hz,2H),7.41(d,J=8.5Hz,2H),6.53(s,1H),4.26(t,J=7.1Hz,2H),1.87–1.75(m,2H),1.31(s,9H),1.26(dd,J=14.9,7.5Hz,2H),0.91(t,J=7.4Hz,3H).HRMS(ESI),m/z:448.2460[M+H] + .
Example 39 1- (4- (4-amino-7-cyclopentyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-39
Figure BDA0002393819090000432
The same synthesis method as that of example 13 was followed, and N- (2-chloroethyl) morpholine hydrochloride was replaced with bromocyclopentane to obtain CLJ-39 as a high-purity final product. 1 H NMR(400MHz,DMSO)δ:9.78(s,1H),9.62(s,1H),8.47(s,1H),7.73(s,1H),7.61(d,J=8.6Hz,2H),7.44(d,J=8.5Hz,2H),6.53(s,1H),5.22–5.08(m,1H),2.16(d,J=7.9Hz,2H),2.02–1.83(m,4H),1.71(d,J=7.0Hz,2H),1.31(s,9H).HRMS(ESI),m/z:460.2457[M+H] + .
Example 40 1- (4- (4-amino-7-hexyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-40
Figure BDA0002393819090000433
The same synthesis method as that in example 13 was used to replace N- (2-chloroethyl) morpholine hydrochloride with N-bromo-hexane to obtain high-purity final product CLJ-40. 1 H NMR(400MHz,DMSO)δ:9.71(s,1H),9.40(s,1H),8.46(s,1H),7.67(s,1H),7.61(d,J=8.6Hz,2H),7.41(d,J=8.5Hz,2H),6.52(s,1H),4.25(t,J=7.1Hz,2H),1.88–1.77(m,2H),1.31(s,9H),1.28(m,6H),0.86(t,J=6.8Hz,3H).HRMS(ESI),m/z:476.2770[M+H] + .
EXAMPLE 41 1- (4- (4-amino-7-benzyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-41
Figure BDA0002393819090000441
High purity final product CLJ-41 was obtained by substituting benzyl bromide for N- (2-chloroethyl) morpholine hydrochloride in the same manner as in the synthesis of example 13. 1 H NMR(400MHz,DMSO)δ:9.71(s,1H),8.49(s,1H),7.74(s,1H),7.60(d,J=8.6Hz,2H),7.41(d,J=8.5Hz,2H),7.38–7.27(m,5H),6.52(s,1H),5.49(s,2H),1.31(s,9H).HRMS(ESI),m/z:482.2303[M+H] + .
Example 42 1- (4- (4-amino-7-propynyl-7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-42
Figure BDA0002393819090000442
The same synthesis method as that in example 13 is adopted, and N- (2-chloroethyl) morpholine hydrochloride is replaced by propargyl bromide to obtain the high-purity final product CLJ-42. 1 H NMR(400MHz,DMSO)δ:9.66(s,1H),9.28(s,1H),8.49(s,1H),7.66(s,1H),7.61(d,J=8.5Hz,2H),7.43(d,J=8.5Hz,2H),6.52(s,1H),5.15(d,J=2.3Hz,2H),3.52(t,J=2.4Hz,2H),1.31(s,9H).HRMS(ESI),m/z:430.1985[M+H] + .
EXAMPLE 43 1- (4- (4-amino-7- (2-cyanoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-43
Figure BDA0002393819090000443
The high purity final product CLJ-43 can be obtained by replacing N- (2-chloroethyl) morpholine hydrochloride with bromopropionitrile in the same way as the synthesis method of the embodiment 13. 1 H NMR(400MHz,DMSO)δ:9.71(s,1H),9.42(s,1H),8.50(s,1H),7.70(s,1H),7.63(d,J=8.6Hz,2H),7.42(d,J=8.5Hz,2H),6.53(s,1H),4.56(t,J=6.4Hz,2H),3.19(t,J=6.4Hz,2H),1.31(s,9H).HRMS(ESI),m/z:445.2097[M+H] + .
Example 44 CLJ-44, a 1- (4- (4-amino-7H-pyrrolo [2,3-d ] pyrimidin-5-) -2-fluorophenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound
Figure BDA0002393819090000451
Step i: preparation of M7 intermediate
Intermediate M3 (9g, 25mmol), 3-fluoro-4-aminophenylboronic acid pinacol ester (6.5g, 27.5 mmol), potassium carbonate (6.9g, 50mmol) and dppf (PdCl) 2 ) The mixture was charged into a 250mL three-necked flask, and dioxane/ethanol/water =7 (120 mL total) was added as a solvent, and after replacing nitrogen three times, the mixture was transferred into an oil bath at 80 ℃ for reaction for 2h. After the reaction is finished, concentrating the reaction solution, mixing the reaction solution with a sample, and separating the mixture by a column to obtain an intermediate M7.
Step j: preparation of M8 intermediate
The intermediate M7 (4 g,11.6 mmol) in the previous step was added to 50mL of methylene chloride, concentrated hydrochloric acid (10mL, 36-38% concentrated hydrochloric acid) was added, and the reaction was completed after 1 hour. 200mL of dichloromethane and 100mL of water were added to adjust the pH of the mixture to basic pH (about 10), the organic phase was retained by extraction and dried to give intermediate M8. 1 H NMR(400MHz,DMSO)δ:11.66(s,1H),8.08(s,1H),7.11(d,J=2.1Hz,1H),7.07(dd,J=12.5,1.8Hz,1H),6.98(dd,J=8.1,1.8Hz,1H),6.85(dd,J=9.5,8.2Hz,1H),5.99(s,2H),5.21(s,2H).
Step k: preparation of CLJ-44
And adding the intermediate M8 (243mg, 1mmol) in the previous step into 20mL of acetonitrile, adding an active urea intermediate (306mg, 1mmol), reacting for 0.5h, separating out a solid, filtering, and leaching a filter cake by using diethyl ether to obtain the high-purity final product CLJ-44. 1 H NMR(400MHz,DMSO)δ:13.04(s,1H),10.13(s,1H),9.20(s,1H),8.46(s,1H),8.23(t,J=8.5Hz,1H),7.61(d,J=2.3Hz,1H),7.37(dd,J=12.0,1.7Hz,1H),7.27(d,J=8.4Hz,1H),6.51(s,2H),1.31(s,9H).HRMS(ESI),m/z:410.1734[M+H] + .
EXAMPLE 45 1- (4- (4-amino-7-propynyl-7H-pyrrolo [2,3-d ] pyrimidin-5-) -2-fluorophenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-45
Figure BDA0002393819090000452
Figure BDA0002393819090000461
Combining steps l and m into a reaction process of a' one-pot method
The intermediate M8 (243mg, 1mmol) obtained in the previous step, cesium carbonate (650mg, 2mmol) and bromopropyne (143mg, 1.2mmol) were added to 20mL of acetonitrile, and the mixture was heated to 80 ℃ to react for 1 hour. The reaction solution was filtered, and the mother liquor was retained. Transferring the mother liquor to 80 ℃, adding an active urea intermediate (306mg, 1mmol), reacting for 0.5h, separating out a large amount of solid, filtering, and leaching with diethyl ether to obtain the high-purity final product CLJ-45. 1 H NMR(400MHz,DMSO)δ:10.01(s,1H),9.08(s,1H),8.52(s,1H),8.26(t,J=8.5Hz,1H),7.73(s,1H),7.39(dd,J=12.0,1.8Hz,1H),7.30–7.25(m,1H),6.51(s,1H),5.16(d,J=2.4Hz,2H),1.31(s,9H).HRMS(ESI),m/z:448.1891[M+H] + .
EXAMPLE 46 CLJ-46, a 1- (4- (4-amino-7-allyl-7H-pyrrolo [2,3-d ] pyrimidin-5) -2-fluorophenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound
Figure BDA0002393819090000462
The high purity final product, CLJ-46, was obtained by substituting bromopropyne for bromopropene in the same manner as in example 45. 1 H NMR(400MHz,DMSO)δ:9.86(s,1H),8.88(s,1H),8.20(t,J=8.5Hz,1H),8.16(s,1H),7.33(s,1H),7.33(dd,J=12.1,1.8Hz,2H),7.26(dd,J=8.5,1.3Hz,1H),6.51(s,1H),6.19(s,2H),6.05(ddd,J=22.5,10.6,5.5Hz,1H),5.18(dd,J=10.2,1.3Hz,1H),5.09(dd,J=17.1,1.4Hz,1H),4.80(d,J=5.5Hz,2H),1.31(s,9H).HRMS(ESI),m/z:450.2042[M+H] + .
EXAMPLE 47 1- (4- (4-amino-7- (but-3-en-1-yl) -7H-pyrrolo [2,3-d ] pyrimidin-5) -2-fluorophenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-47
Figure BDA0002393819090000463
In the same manner as in example 45, the bromopropyne was replaced with 4-bromo-1-butene, thereby obtaining high-purity final product CLJ-47. 1 H NMR(400MHz,DMSO)δ:10.05(s,1H),9.12(s,1H),8.49(s,1H),8.25(t,J=8.5Hz,1H),7.73(s,1H),7.36(dd,J=12.0,1.7Hz,1H),7.26(d,J=8.4Hz,1H),6.51(s,1H),5.80(ddt,J=17.0,10.3,6.7Hz,1H),5.10–4.99(m,2H),4.34(t,J=7.0Hz,2H),2.61(q,J=6.8Hz,2H),1.31(s,9H).HRMS(ESI),m/z:464.2212[M+H] + .
EXAMPLE 48 CLJ-48, a 1- (4- (4-amino-7-methoxyethyl-7H-pyrrolo [2,3-d ] pyrimidin-5) -2-fluorophenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound
Figure BDA0002393819090000471
In the same manner as the synthesis method in example 45, bromopropyne was replaced with chloroethyl methyl ether, thereby obtaining high-purity final product CLJ-48. 1 H NMR(400MHz,DMSO)δ:10.10(s,1H),9.16(s,1H),8.51(s,1H),8.25(t,J=8.5Hz,1H),7.70(s,1H),7.43–7.33(m,1H),7.26(m,1H),6.51(s,1H),4.43(t,J=5.1Hz,2H),3.76(t,J=5.3Hz,2H),1.31(s,9H).HRMS(ESI),m/z:468.2159[M+H] + .
Example 49 1- (4- (4-amino-7-ethoxyethyl-7H-pyrrolo [2,3-d ] pyrimidin-5) -2-fluorophenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-49
Figure BDA0002393819090000472
The synthesis method of example 45 was followed, and bromopropyne was replaced with bromoethyl ether to obtain high-purity final product CLJ-49. 1 H NMR(400MHz,DMSO)δ:10.02(s,1H),9.09(s,1H),8.48(s,1H),8.26(t,J=8.5Hz,1H),7.68(s,1H),7.37(dd,J=12.0,1.8Hz,1H),7.27(d,J=8.5Hz,1H),6.51(s,1H),4.42(t,J=5.3Hz,2H),3.78(t,J=5.3Hz,2H),3.46(dd,J=14.0,7.0Hz,2H),1.31(s,9H),1.06(t,J=7.0Hz,3H).HRMS(ESI),m/z:482.2313[M+H] + .
EXAMPLE 50 1- (4- (4-amino-7-cyanomethyl-7H-pyrrolo [2,3-d ] pyrimidin-5) -2-fluorophenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-50
Figure BDA0002393819090000481
In the same manner as in example 45, the bromopropyne was replaced with chloroacetonitrile, whereby CLJ-50, a high-purity final product, was obtained. 1 H NMR(400MHz,DMSO)δ:10.10(s,1H),9.16(s,1H),8.57(s,1H),8.26(t,J=8.5Hz,1H),7.72(s,1H),7.39(dd,J=11.9,1.8Hz,1H),7.28(d,J=8.4Hz,1H),6.51(s,1H),5.55(s,2H),1.31(s,9H).HRMS(ESI),m/z:449.1848[M+H] + .
EXAMPLE 51 1- (4- (4-amino-7- (2-cyanoethyl) -7H-pyrrolo [2,3-d ] pyrimidin-5) -2-fluorophenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-51
Figure BDA0002393819090000482
In the same manner as in the synthesis of example 45, bromopropionitrile was replacedPropyne bromide is adopted to obtain the high-purity final product CLJ-51. 1 H NMR(400MHz,DMSO)δ:10.05(s,1H),9.12(s,1H),8.54(s,1H),8.28(t,J=8.5Hz,1H),7.79(s,1H),7.65(s,1H),7.37(dd,J=11.9,1.8Hz,1H),7.28(d,J=8.5Hz,1H),6.51(s,1H),4.60–4.51(t,J=6.5Hz,2H),3.19(t,J=6.6Hz,2H),1.31(s,9H).HRMS(ESI),m/z:463.2004[M+H] + .
Example 52 1- (4- (4-amino-7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-52
Figure BDA0002393819090000483
The synthetic route is shown as follows;
Figure BDA0002393819090000484
the synthetic route corresponds essentially to example 13, except that the starting material is replaced by 4-chloropyrazolo [2,3-d ]]Pyrimidine is reacted in 6 steps to obtain the final product CLJ-52. 1 H NMR(400MHz,DMSO)δ:13.80(s,1H),9.35(s,1H),9.14(s,1H),8.50(s,1H),7.53(s,2H),7.24–7.18(m,2H),6.73–6.67(m,2H),6.46(s,1H),1.29(s,9H).HRMS(ESI),m/z:392.1907[M+H] + .
Example 53 CLJ-53-1- (4- (4-amino-7-allyl-7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound
Figure BDA0002393819090000491
In line with the route of example 52, except that bromopropene was added in g, and the reactive urea intermediate reacted with the next step was synthesized according to the "one-pot" synthesis to give final product CLJ-53. 1 H NMR(400MHz,DMSO)δ:9.56(s,1H),9.02(s,1H),8.26(s,1H),7.63(t,J=7.0Hz,4H),7.00(dd,J=114.0,63.4Hz,2H),6.53(s,1H),6.10–6.01(m,1H),5.19(d,J=10.3Hz,1H),5.11(d,J=17.1Hz,1H),4.97(d,J=5.5Hz,2H),1.31(s,9H).HRMS(ESI),m/z:433.2099[M+H] + .
EXAMPLE 54 1- (4- (4-amino-7- (2-morpholinoethyl) -7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound CLJ-54
Figure BDA0002393819090000492
This compound was synthesized in the same manner as in example 53 except that N- (2-chloroethylmorpholine) hydrochloride was used in place of bromopropene to give the desired end product CLJ-54. 1 H NMR(400MHz,DMSO)δ9.57(s,1H),9.04(s,1H),8.25(s,1H),7.66–7.63(m,2H),7.63–7.61(m,2H),6.75(s,2H),6.53(s,1H),4.48–4.46(m,2H),3.52–3.48(m,4H),2.83–2.81(m,2H),2.49–2.46(m,4H),1.31(s,9H).HRMS(ESI),m/z:506.2628[M+H] + .
EXAMPLE 55 CLJ-55- (4- (4-amino-7- (2- (piperidin-1-yl) ethyl) -7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound
Figure BDA0002393819090000493
This compound was synthesized in the same manner as in example 53 except that N- (2-chloroethylpiperidine) hydrochloride was used in place of bromopropene to obtain the objective end product CLJ-55. 1 H NMR(400MHz,DMSO)δ:9.58(s,1H),9.05(s,1H),8.25(s,1H),7.66–7.63(m,2H),7.63–7.61(m,2H),6.75(s,2H),6.53(s,1H),4.52–4.48(m,2H),4.44–4.40(m,2H),2.78–2.74(m,2H),2.44–2.38(m,4H),1.46–1.40(m 4H),1.31(s,9H).HRMS(ESI),m/z:504.2829[M+H] + .
EXAMPLE 56 1- (4- (4-amino-7- (cyclopropylmethyl) -7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound CLJ-56
Figure BDA0002393819090000501
The synthesis method of the compoundThe same procedure as in example 53, except for replacing bromopropene with 2-chlorocyclopropane, gave the desired final product CLJ-56. 1 H NMR(400MHz,DMSO)δ:9.56(s,1H),9.02(s,1H),8.25(s,1H),7.66–7.63(m,2H),7.63–7.61(m,2H),6.90(s,2H),6.53(s,1H),5.84–5.76(m,1H),5.05–4.97(m,2H),4.42–4.39(m,2H),2.66–2.61(m,2H),1.31(s,9H).HRMS(ESI),m/z:447.2257[M+H] + .
Example 57 1- (4- (4-amino-7-propargyl-7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-57
Figure BDA0002393819090000502
The synthesis of this compound was performed in the same manner as in example 53 except that bromopropyne was used in place of bromopropene to obtain the objective final product, CLJ-57. 1 H NMR(400MHz,DMSO)δ9.63(s,1H),9.10(s,1H),8.34(s,1H),7.72–7.68(m,4H),6.59(s,1H),5.51(s,2H),5.25(s,2H),3.43(s,1H),1.37(s,9H).HRMS(ESI),m/z:431.1944[M+H] + .
Example 58 1- (4- (4-amino-7-methyl-7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound CLJ-58
Figure BDA0002393819090000503
The synthesis of this compound was performed in the same manner as in example 53 except that methyl iodide was used instead of propylene bromide to obtain the desired final product, CLJ-58. 1 H NMR(400MHz,DMSO)δ:9.56(s,1H),9.05(s,1H),8.26(s,1H),7.66–7.61(m,4H),6.53(s,1H),5.41(s,2H),4.03–3.99(m,3H),1.31(s,9H).HRMS(ESI),m/z:407.1948[M+H] + .
Example 59 1- (4- (4-amino-7-ethyl-7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-59
Figure BDA0002393819090000511
The synthesis of this compound was performed in the same manner as in example 53 except that iodoethane was used instead of bromopropene to obtain CLJ-59, the objective final product. 1 H NMR(400MHz,DMSO)δ:9.58(s,1H),9.05(s,1H),8.26(s,1H),7.66–7.61(m,4H),6.53(s,1H),5.41(s,2H),3.97–3.93(m,2H),1.31(s,9H),1.26–1.22(m,3H).HRMS(ESI),m/z:421.2100[M+H] + .
EXAMPLE 60 1- (4- (4-amino-7- (2-cyanoethyl) -7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound CLJ-60
Figure BDA0002393819090000512
This compound was synthesized in the same manner as in example 53 except that chloropropionitrile was used in place of bromopropene to give the desired final product CLJ-60. 1 H NMR(400MHz,DMSO)δ9.62(s,1H),9.10(s,1H),8.34(s,1H),7.72–7.66(m,4H),6.59(s,1H),5.67(s,2H),4.66(s,2H),3.22–3.18(m,2H),1.36(s,9H).HRMS(ESI),m/z:446.2056[M+H] + .
Example 61 1- (4- (4-amino-7-hexyl-7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound CLJ-61
Figure BDA0002393819090000513
This compound was synthesized in the same manner as in example 53 except that 1-bromohexane was used in place of bromopropene to obtain the objective final product CLJ-61. 1 H NMR(400MHz,DMSO)δ9.56(s,1H),9.03(s,1H),8.25(s,1H),7.65–7.59(m,4H),6.54(s,1H),5.42(s,2H),4.34–4.31(m,2H),1.84–1.82(m,4H),1.31(s,9H),0.85–0.82(m,7H).HRMS(ESI),m/z:477.2396[M+H] + .
Example 62 1- (4- (4-amino-7-propyl-7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-62
Figure BDA0002393819090000521
The synthesis of this compound was performed in the same manner as in example 53 except that 1-bromopropyl was used instead of bromopropene to obtain the desired final product, CLJ-62. 1 H NMR(400MHz,DMSO)δ:9.56(s,1H),9.02(s,1H),8.25(s,1H),7.64–7.60(m,4H),6.53(s,1H),5.41(s,2H),4.32–4.28(m,2H),1.92–1.84(m,2H),1.31(s,9H),0.89–0.85(m,3H).HRMS(ESI),m/z:435.2256[M+H] + .
Example 63 1- (4- (4-amino-7-cyclopentyl-7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound CLJ-63
Figure BDA0002393819090000522
The synthesis of this compound was the same as in example 53 except that bromocyclopentane was substituted for bromopropene to give the desired end product, CLJ-63. 1 H NMR(400MHz,DMSO)δ9.55(s,1H),9.03(s,1H),8.23(s,1H),7.64–7.60(m,4H),6.72(s,2H),6.53(s,1H),5.28–5.19(m,1H),2.08–2.08(m,4H),1.92–1.87(m,2H),1.74–1.67(m,2H),1.31(s,9H).HRMS(ESI),m/z:461.2407[M+H] + .
Example 64 CLJ-64- (4- (4-amino-7- (2-ethoxyethyl) -7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound
Figure BDA0002393819090000523
The synthesis of this compound was the same as in example 53 except that bromoethyl ether was used instead of bromopropene to give the desired end product, CLJ-64. 1 H NMR(400MHz,DMSO)δ:9.57(s,1H),9.05(s,1H),8.25(s,1H),7.67–7.61(m,4H),6.88(s,2H),6.54(s,1H),4.49–4.42(m,4H),1.31(s,9H),1.04–1.00(m,5H).HRMS(ESI),m/z:465.2361[M+H] + .
EXAMPLE 65 CLJ-65-1- (4- (4-amino-7- (3-methylbut-2-en-1-yl) -7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea Compound
Figure BDA0002393819090000531
The synthesis of this compound was the same as in example 53 except that 3, 3-dimethylbromopropene was used instead of bromopropene to give the desired end product, CLJ-65. 1 H NMR(400MHz,DMSO)δ:9.57(s,1H),9.05(s,1H),8.25(s,1H),7.65–7.59(m,4H),6.53(s,1H),5.42(s,2H),4.94–4.88(m,3H),1.82–1.79(m,5H),1.70(s,3H),1.31(s,9H).HRMS(ESI),m/z:461.2407[M+H] + .
EXAMPLE 66 CLJ-66, a 1- (4- (4-amino-7- (2, 2-dimethoxyethyl) -7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound
Figure BDA0002393819090000532
The synthesis of this compound was the same as in example 53 except that 2, 2-dimethylbromoethyl was used instead of bromopropene to give the desired end product, CLJ-66. 1 H NMR(400MHz,DMSO)δ:9.57(s,1H),9.04(s,1H),8.27(s,1H),7.66–7.60(m,4H),6.54(s,1H),5.46(s,2H),4.96–4.93(m,1H),4.44–4.42(m,2H),3.33(s,6H),1.31(s,9H).HRMS(ESI),m/z:481.2307[M+H] + .
EXAMPLE 67 CLJ-67- (4- (4-amino-7- (4-oxopentyl) -7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (5-tert-butyl-isoxazol-3-yl) urea compound
Figure BDA0002393819090000533
The synthesis of this compound was the same as in example 53 except that 2, 2-dimethylbromoethyl was used instead of bromopropene to give the desired end product, CLJ-67. 1 H NMR(400MHz,DMSO)δ:9.57(s,1H),9.05(s,1H),8.21(s,1H),7.66–7.60(m,4H),6.54(s,1H),5.42(s,2H),4.30–4.25(m,2H),2.48–2.42(m,4H),2.05(s,3H),1.31(s,9H).HRMS(ESI),m/z:477.2396[M+H] + .
EXAMPLE 68 CLJ-68-1- (4- (4-amino-7H-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (3-tert-butyl-1H-pyrazol-5-yl) urea Compound
Figure BDA0002393819090000541
Preparation of active urea intermediate 1,3- (3-tert-butyl-1H-pyrazol-5-yl) urea
Figure BDA0002393819090000542
Triphosgene (4.71g, 15.7 mmol) was added to 50mL of tetrahydrofuran, and 3-amino-5-tert-butylpyrazole (1.99g, 14.3 mmol) was dissolved in 5mL of tetrahydrofuran under ice-cooling, and then added dropwise to the triphosgene solution, and finally triethylamine (4.0 mL,28.5 mmol) was added dropwise. The reaction was transferred to a 60 ℃ oil bath and reacted for 5h. And after the reaction is completed, filtering the reaction mixture, reserving filtrate, concentrating the filtrate under reduced pressure to obtain a solid, and separating the solid by a column to obtain the active urea intermediate. 1 H NMR(400MHz,DMSO)δ:12.66(s,2H),9.51(s,2H),6.30(s,2H),1.30(s,18H).
And adding the intermediate M5 (225mg, 1mmol) into 20mL of acetonitrile, heating to 80 ℃ for reaction, adding the active urea intermediate (390mg, 1mmol) in the previous step, reacting for 0.5h, precipitating a large amount of solid, filtering, and leaching with diethyl ether to obtain the high-purity final product CLJ-68. 1 H NMR(400MHz,DMSO)δ:12.64(s,1H),11.54(s,1H),9.52(s,1H),8.86(s,1H),8.11(s,1H),7.80(d,J=8.5Hz,2H),7.65(d,J=8.5Hz,2H),7.19(d,J=2.3Hz,1H),6.54(s,1H),6.01(s,2H),1.31(s,9H).HRMS(ESI),m/z:391.1924[M+H] + .
EXAMPLE 69 1- (4- (4-amino-7- (2-morpholinoethyl) -pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (3-tert-butyl-1H-pyrazol-5-yl) urea Compound CLJ-69
Figure BDA0002393819090000543
The synthesis and implementation of the compoundThe same procedure as in example 54, except that the isoxazole reactive intermediate was replaced with the reactive intermediate of pyrazolylurea, gave the desired final product, CLJ-69. 1 H NMR(400MHz,DMSO)δ12.00(s,1H),9.46(s,1H),9.05(s,1H),8.24(s,1H),7.61(dd,J=21.8,8.6Hz,4H),6.78(s,2H),6.02(s,1H),4.46(t,J=6.6Hz,2H),3.54–3.47(m,4H),2.80(t,J=6.7Hz,2H),2.46(d,J=3.8Hz,4H),1.27(s,9H).HRMS(ESI),m/z:505.2811[M+H] + .
EXAMPLE 70 1- (4- (4-amino-7-ethyl-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (3-tert-butyl-1H-pyrazol-5-yl) urea Compound CLJ-70
Figure BDA0002393819090000551
The synthesis of this compound is the same as in example 59 except that the isoxazole reactive intermediate is replaced by the reactive intermediate of pyrazolylurea to give the desired end product CLJ-70. 1 H NMR(400MHz,DMSO)δ:12.01(s,1H),9.42(s,1H),9.00(s,1H),8.25(s,1H),7.61(dd,J=18.2,8.6Hz,4H),6.69(s,2H),6.02(s,1H),4.37(dd,J=14.2,7.0Hz,2H),1.42(t,J=7.1Hz,3H),1.19(s,9H).HRMS(ESI),m/z:420.2279[M+H] + .
EXAMPLE 71 1- (4- (4-amino-7- (cyclopropylmethyl) -pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (3-tert-butyl-1H-pyrazol-5-yl) urea Compound CLJ-71
Figure BDA0002393819090000552
The synthesis of this compound is the same as in example 56 except that the active intermediate of pyrazolylurea is substituted for the active intermediate of isoxazole to give the desired end product CLJ-71. 1 H NMR(400MHz,DMSO)δ:12.00(s,1H),9.49(s,1H),9.04(s,1H),8.25(s,1H),7.61(dd,J=21.9,8.5Hz,4H),6.69(s,2H),6.03(s,1H),5.03(dd,J=35.8,13.0Hz,2H),4.40(d,J=7.1Hz,2H),2.63(dd,J=13.6,6.8Hz,2H),1.19(s,9H),0.84(dd,J=10.0,7.0Hz,1H).HRMS(ESI),m/z:446.2456[M+H] + .
Example 72 1- (4- (4-amino-7-propyl-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (3-tert-butyl-1H-pyrazol-5-yl) urea Compound CLJ-72
Figure BDA0002393819090000553
The synthesis of this compound is the same as in example 62 except that the active intermediate of pyrazolylurea is substituted for the active intermediate of isoxazole to give the desired end product CLJ-72. 1 H NMR(400MHz,DMSO)δ:12.00(s,1H),9.43(s,1H),8.98(s,1H),8.25(s,1H),7.61(dd,J=21.9,8.5Hz,4H),6.02(s,1H),4.29(t,J=6.2Hz,2H),1.90–1.84(m,2H),1.27(s,9H),0.86(m,3H).HRMS(ESI),m/z:434.2419[M+H] + .
Example 73 1- (4- (4-amino-7-cyclopentyl-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (3-tert-butyl-1H-pyrazol-5-yl) urea Compound CLJ-73
Figure BDA0002393819090000561
The synthesis of this compound is the same as in example 63 except that the active intermediate of pyrazolylurea is substituted for the active intermediate of isoxazole to give the desired end product CLJ-73. 1 H NMR(400MHz,DMSO)δ12.01(s,1H),9.40(s,1H),8.98(s,1H),8.23(s,1H),7.61(dd,J=19.4,8.4Hz,4H),6.72(s,2H),6.02(s,1H),5.23(dt,J=14.8,7.5Hz,1H),2.12–2.03(m,4H),1.90(d,J=8.5Hz,2H),1.73–1.65(m,2H),1.27(s,9H).HRMS(ESI),m/z:460.2624[M+H] + .
Example 74 CLJ-74- (4- (4-amino-7-allyl-pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (3-tert-butyl-1H-pyrazol-5-yl) urea Compound
Figure BDA0002393819090000562
The synthesis of this compound is the same as in example 53 except that the active intermediate of pyrazolylurea is substituted for the active intermediate of isoxazole to give the desired end product CLJ-74. 1 H NMR(400MHz,DMSO)δ:12.04(s,1H),9.41(s,1H),9.00(s,1H),8.25(s,1H),7.61(dd,J=16.8,7.2Hz,4H),,6.72(s,2H),6.11–6.04(m,1H),6.02(s,1H),5.19(d,J=10.4Hz,1H),5.11(d,J=17.5Hz,1H),4.97(s,2H),1.27(s,9H).HRMS(ESI),m/z:432.2307[M+H] + .
EXAMPLE 75 1- (4- (4-amino-7- (2-cyanoethyl) -pyrazolo [2,3-d ] pyrimidin-5-yl) phenyl) -3- (3-tert-butyl-1H-pyrazol-5-yl) urea Compound CLJ-75
Figure BDA0002393819090000563
The synthesis method of the compound is the same as that of example 60, except that the active intermediate of the pyrazole urea is used for replacing the active intermediate of the isoxazole to obtain the target final product CLJ-75. 1 H NMR(400MHz,DMSO)δ:12.00(s,1H),9.52(s,1H),9.05(s,1H),8.28(s,1H),7.63(dd,J=19.1,8.1Hz,4H),6.73(s,2H),6.03(s,1H),4.61(t,J=6.1Hz,2H),3.17(t,J=6.0Hz,2H),1.27(s,9H).HRMS(ESI),m/z:445.2244[M+H] + .
Pharmacodynamic test section
The following representative experiments, without limitation, were used to analyze the biological activity of the compounds of the present invention.
Measuring MV4-11, molm-13 and RS4 by an MTT method; 11 cell proliferation inhibition assay
The test compounds of the invention were tested for their effect on cancer cell viability on MV4-11 and Molm-13 cells, which are human leukemia cell lines, expressing the constitutive FLT3 receptor and containing the FLT3-ITD mutation. If the compound has strong growth inhibition activity on FLT3-ITD expression cells, the compound has obvious effect on FLT3-ITD mutant strains. If the growth inhibitory activity on cells expressing FLT3-WT is poor, the compound has poor effect on FLT3-WT wild type, and the selectivity is better. The FLT3-ITD high-specificity compound AC220 (CAS: 950769-58-1) is selected as a positive control, and is synthesized by the laboratory according to a preparation method of a literature (J.Med.chem.2009, 52, 7808-7816).
MV4-11 cells (from the American type culture center, culture and breed conservation by cell Bank of the national center for biotherapy, sichuan university) were plated in 96-well plates in a medium of 100. Mu.L IMDM, molm-13 cells (Lai-Mi)From the American type culture center, by national emphasis laboratory cell Bank of biotherapy of Sichuan university) was charged in 96-well culture dishes in a medium of 100. Mu.L RPMI1640 containing 10% fetal bovine serum with 10000-15000 cells per well, the test compound was prepared in 100% DMSO, added to the cells to obtain a concentration of 100nM to 0.032nM (6 concentration points at 5-fold dilution concentration) in culture dishes at 37 ℃ of 5 CO% 2 And culturing for 72h. RS4;11 cells (from the American type culture center, from national institute of biotherapy, national emphasis laboratory cell bank culture protection, sichuan university) were plated in 96-well plates in a medium of 100 μ L RPMI1640 containing 10% fetal bovine serum, 10000-15000 cells per well, the test compound was prepared in 100% DMSO, added to the cells to obtain a concentration of 1000nM, and the plates were incubated at 37 ℃ for 5% CO 2 And culturing for 72h. At the end point, 20 μ LMTT (5 mg/mL) was added to each well and the cells were incubated for an additional 1-4 hours. After overnight treatment with 20% SDS, an absorbance value at a wavelength of 570nM was obtained on a spectrophotometer (Molecular Devices, sunnyvale, USA). Calculation of IC Using percent growth compared to untreated control 50 The values and measurement results are shown in Table 1.
TABLE 1 IC inhibition of leukemia cell proliferation by test Compounds 50 Value of
Figure BDA0002393819090000571
Figure BDA0002393819090000581
Figure BDA0002393819090000591
Figure BDA0002393819090000601
******:0.01-0.1nM;*****:0.1-1nM;****:1-10nM;***:10-100nM;**:100-1000nM;*:>1000nM
The results show that the test compounds of the present invention have better inhibitory activity on the proliferation of MV4-11 and Molm-13 cells, some of which exhibit better antiproliferative activity than AC220, and on RS4;11 is poor in inhibitory activity and is a novel, potential and potential inhibitor for treating FLT3-ITD related diseases.
In vitro kinase inhibition assay
Buffer (8 mM) MOPS, pH 7.0,0.2mM EDTA, and 10mM MnCl were added to one reaction tube 2 ) A kinase to be tested, a substrate for the kinase to be tested, 10mM magnesium acetate and gamma 33P ATP solution, and different concentrations of compounds, then MgATP was added to the reaction to start the enzymatic reaction process, and incubated at room temperature for 40 min. Finally, stopping the reaction by using 5 microliter of 3% phosphate buffer solution, titrating 10 microliter of reaction solution onto a Filtermat A membrane, washing the Filtermat A membrane by using 75mM phosphate solution for three times, 5 minutes each time, washing the Filtermat A membrane by using methanol once again, finally drying the Filtermat A membrane and carrying out scintillation counting on the Filtermat A membrane, wherein the scintillation counting value reflects the phosphorylation degree of a substrate, so that the kinase activity inhibition condition can be characterized.
TABLE 2 partial compound FLT3 kinase inhibitor Activity of the present invention
Test compounds FLT3(IC 50 ,nM)
CLJ-13 20
CLJ-14 6
CLJ-20 7
CLJ-21 9
CLJ-22 20
CLJ-42 13
CLJ-44 4
The result shows that the compound has better in-vitro enzymology inhibitory activity on FLT 3.
TABLE 3 dissociation constants of the inventive compound CLJ-20 for FLT3 mutant kinase
Type of mutation Kd,nM
FLT3 WT 4.37
FLT3(D835V) 7.04
FLT3(ITD) 20.52
FLT3(ITD,D835V) 41.26
FLT3(ITD,F691L) 48.47
FLT3(N841I) 2.37
FLT3(R834Q) 10.15
FLT3(D835H) 5.14
FLT3(D835Y) 7.78
FLT3(K663Q) 2.48
The results show that CLJ-20 also has good combination effect on various mutations of FLT3 kinase.
Part of tested compounds verify the target effect on Western Blot
The test method comprises the following steps: MV4-11 cells, molm-13 cells, were treated with the indicated concentrations of the compounds. The cells were then harvested and total protein extracted with RIPA lysis buffer (beyond time Co. P0013B, fraction: 50mM Tris, pH 7.4,150mM NaCl,1% Triton X-100,1% sodium deoxycholate, 0.1% SDS, 1). 1mM sodium orthovanadate, sodium fluoride, EDTA and leupeptin). Protein concentration was measured by BCA protein assay (ThermoScientific, USA). An equivalent sample (30. Mu.g protein) was subjected to SDS-PAGE, and then the protein was transferred onto a PVDF membrane (Millipore, USA). After blocking with 5% skim milk for 1 hour at room temperature, the membranes were incubated with the indicated primary antibodies (FLT 3 (Cell signaling technology, 3462S), p-FLT3 (Cell signaling technology, 3464S), STAT5 (Cell signaling technology, 9363S), p-STAT5 (Abcam, AB 32364), ERK (Zen Bioscience, 220003), p-ERK (Zen Bioscience, 340767), β -Actin (Abways, AB 0035)) overnight at 4 ℃ and probed with the appropriate secondary antibody (Abways, F300409) coupled with horseradish peroxidase for 1 hour. The immunoreactive bands were visualized using enhanced chemiluminescence (Millipore, USA). The molecular size of the detected protein was determined by comparison with a relevant protein marker (ThermoScientific, USA).
The experimental results are shown in figures 1 and 2, and the results show that the tested compound CLJ-20 can significantly reduce p-FLT3, p-STAT5 and p-ERK in a dose-dependent manner, and AC220 also shows the same effect, which indicates that the tested compound really influences the phosphorylation of a downstream signal channel by inhibiting the activity of FLT 3.
In vivo pharmacodynamic experiment of compound CLJ-20 on NOD/SCID nude mice
The purpose of this experiment was to examine the in vivo anti-tumor effect of the compounds of the invention using a mouse subcutaneous tumor model using MV4-11 and Molm-13 cell lines as well as developing clinical drug AC220 as a positive control.
The test method comprises the following steps: 6-8 weeks NOD/SCID mice (purchased from Beijing Huafukang Biotech GmbH) were used, as 10 7 The cells with the tumor swelling to 200-500mm are inoculated in the subcutaneous posterior costal region of the mouse at the concentration of 0.1 mL/Molm-13 and MV4-11 cells 3 Thereafter, mice were grouped (6 per group) and dosed with 2% DMSO +2% PEG-400+96% normal saline, experimental groups were grouped as drug solvent control groups, and gavage was performed orally at 200. Mu.l per day, compound CLJ-20 was gavaged orally at a dose of 3mg/kg per day, compound CLJ-20 was gavaged orally at a dose of 10mg/kg per day, and positive drug AC220 was gavaged orally at 3mg/kg per day. The observation indexes are that the body weight of the mice and the long diameter and the short diameter of the tumor are measured once every two days, the volume of the tumor is calculated, and the physiological state of each group of mice is observed.
The experimental results are as follows: the Molm-13 model experiment results are shown in FIG. 3, and the MV4-11 model results are shown in FIG. 4. Experimental results show that the compound CLJ-20 has an obvious in-vivo anti-tumor effect on a Molm-13 model, can obviously inhibit tumor growth under the oral dosage of 3mg/kg, has the tumor inhibition rate of 94 percent, has the tumor inhibition rate of 96 percent at the time of 10mg/kg, has a better anti-tumor effect on AC220, has the tumor inhibition rate of 98 percent, and shows that the CLJ-20 and the AC220 have equivalent in-vivo effects. In MV4-11 model, the tumor-inhibiting rate of the test drug CLJ-20 was as high as 98% at 3mg/kg, and tumor regression was achieved in two mice, 10mg/kg on the eighth day of administration, and AC220 mg/kg group on the eighth day. No adverse reactions such as weight reduction, rash, diarrhea and the like of the mice are found in the administration process. While a slight decrease in body weight was observed in the AC220 group, indicating that CLJ-20 was less toxic over the range of doses administered at the doses tested.
In vivo survival study of compound CLJ-20 in NCG mouse model
The test method comprises the following steps: 7-8 weeks of NCG mice (purchased from Jiangsu Jiejicaokang Biotech, inc.) were used, as per 10 weeks 6 When the Molm-13 cells are inoculated in the tail vein of the mouse at a concentration, the cells can be massively proliferated in the blood circulation system of the mouse, so that the mouse has the symptoms of hind limb paralysis and even death. Mice were grouped (8 per group) and dosed with 2% DMSO +2% PEG-400+96% normal saline, experimental groups were drug solvent control groups, gavage was performed orally at 200 μ l per day, compound CLJ-20 was gavaged orally at a dose of 10mg/kg per day for 30 days continuously. The therapeutic effect of CLJ-20 was examined by observing and recording the survival time of mice after model establishment.
The results of the experiment show that the Median Survival Time (MST) of the mice in the solvent control group is 20 days, as shown in fig. 5. The median survival time of the 10mg/kg dose group reaches 46.5 days, is prolonged by 26.5 days compared with the control group, and is sufficiently prolonged by more than one time. This indicates that CLJ-20 has significant tumor growth inhibition effects in AML xenograft in situ models, greatly prolonging survival.
Although the present invention has been described in detail hereinabove by way of general description, specific examples and experiments, it will be apparent to those skilled in the art that modifications and improvements can be made thereto based on the present invention. Accordingly, it is intended that all such modifications and variations be included within the scope of the invention as claimed and not departing from the spirit thereof.

Claims (20)

1.4-amino-pyrimidoazepine-phenylurea derivatives having the structural formula shown in formula I:
Figure FDA0004053437130000011
wherein X is N or C;
R 1 is-H, C1-C8 alkyl,
Figure FDA0004053437130000012
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is
Figure FDA0004053437130000013
-CN, -OH, phenyl, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl,
Figure FDA0004053437130000014
R 4 Is C1-C4 alkoxy or
Figure FDA0004053437130000015
R 5 ~R 6 、R 9 ~R 10 Independently is C1-C4 alkyl; r 11 is-H, C1-C4 oxycarbonyl or
Figure FDA0004053437130000016
R 2 Is composed of
Figure FDA0004053437130000017
R 3 is-H or halogen.
2. 4-amino-pyrimidoazepine-phenylurea derivatives according to claim 1The method is characterized in that: x is N or C; r 1 is-H, C1-C8 alkyl,
Figure FDA0004053437130000018
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is
Figure FDA0004053437130000019
-CN, -OH, phenyl, methoxy, ethoxy,
Figure FDA00040534371300000110
Formyl, acetyl,
Figure FDA00040534371300000111
Figure FDA00040534371300000112
R 4 Is tert-butyloxy or
Figure FDA00040534371300000113
R 5 ~R 6 、R 9 ~R 10 Independently methyl or ethyl;
R 2 is composed of
Figure FDA00040534371300000114
R 3 is-H or halogen.
3. The 4-amino-pyrimidoazetidic-phenylurea derivative according to claim 2, characterized in that: x is N or C; r 1 is-H, C1-C8 alkyl,
Figure FDA0004053437130000021
T-butyloxycarbonyl group, C2-C6 alkenyl group, C2-C6 alkynyl group,
Figure FDA0004053437130000022
Figure FDA0004053437130000023
R 2 Is composed of
Figure FDA0004053437130000024
R 3 is-H, -F, -Cl or-Br.
4. The 4-amino-pyrimidoazetidic-phenylurea derivative according to claim 1, characterized in that: when R is 2 Is composed of
Figure FDA0004053437130000025
And the structure is shown as formula II:
Figure FDA0004053437130000026
wherein X is N or C; r is 1 is-H, C1-C8 alkyl,
Figure FDA0004053437130000027
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is
Figure FDA0004053437130000028
-CN, -OH, phenyl, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl,
Figure FDA0004053437130000029
R 4 Is C1-C4 alkoxy or
Figure FDA00040534371300000210
R 5 ~R 6 、R 9 ~R 10 Independently is C1-C4 alkyl;R 11 is-H, C1-C4 oxycarbonyl or
Figure FDA0004053437130000031
R 3 is-H or halogen.
5. The 4-amino-pyrimidoazetidic-phenylurea derivative according to claim 4, characterized in that: x is N or C; r 1 is-H, C1-C8 alkyl,
Figure FDA0004053437130000032
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is
Figure FDA0004053437130000033
-CN, -OH, phenyl, methoxy, ethoxy,
Figure FDA0004053437130000034
Formyl, acetyl,
Figure FDA0004053437130000035
Figure FDA0004053437130000036
R 4 Is tert-butyloxy or
Figure FDA0004053437130000037
R 5 ~R 6 、R 9 ~R 10 Independently methyl or ethyl;
R 3 is-H or halogen.
6. The 4-amino-pyrimidoazetidic-phenylurea derivative according to claim 5, characterized in that: x is N or C; r 1 is-H, C1-C8 alkyl,
Figure FDA0004053437130000038
T-butyloxycarbonyl group, C2-C6 alkenyl group, C2-C6 alkynyl group,
Figure FDA0004053437130000039
Figure FDA00040534371300000310
R 3 is-H, -F, -Cl or-Br.
7. The 4-amino-pyrimidoazetidic-phenylurea derivative according to claim 1, characterized in that: when R is 2 Is composed of
Figure FDA00040534371300000311
R 3 When the structure is-H, the structure is shown as formula III:
Figure FDA0004053437130000041
wherein X is N or C; r is 1 is-H, C1-C8 alkyl,
Figure FDA0004053437130000042
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is
Figure FDA0004053437130000043
-CN, -OH, phenyl, C1-C4 alkoxy, C3-C6 cycloalkyl, C1-C4 carbonyl,
Figure FDA0004053437130000044
R 4 Is C1-C4 alkoxy or
Figure FDA0004053437130000045
R 5 ~R 6 、R 9 ~R 10 Independently is C1-C4 alkyl; r 11 is-H, C1-C4 oxycarbonyl or
Figure FDA0004053437130000046
8. The 4-amino-pyrimidoazetidic-phenylurea derivative according to claim 7, characterized in that: x is N or C; r is 1 is-H, C1-C8 alkyl,
Figure FDA0004053437130000047
C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or substituted C1-C4 alkyl; the substituent of the substituted C1-C4 alkyl is
Figure FDA0004053437130000048
-CN, -OH, phenyl, methoxy, ethoxy,
Figure FDA0004053437130000049
Formyl, acetyl,
Figure FDA00040534371300000410
Figure FDA00040534371300000411
R 4 Is tert-butyloxy or
Figure FDA00040534371300000412
R 5 ~R 6 、R 9 ~R 10 Independently methyl or ethyl.
9. The 4-amino-pyrimidoazetidic-phenylurea derivative according to claim 8, characterized in that: x is N or C; r 1 is-H, C1-C8 alkyl,
Figure FDA00040534371300000413
Tert-butyloxycarbonyl radicalA C2-C6 alkenyl group, a C2-C6 alkynyl group,
Figure FDA00040534371300000414
Figure FDA00040534371300000415
Figure FDA0004053437130000051
4-amino-pyrimidoazetidine-phenylurea derivatives having the following structural formula:
Figure FDA0004053437130000052
Figure FDA0004053437130000061
Figure FDA0004053437130000071
Figure FDA0004053437130000081
11. a pharmaceutically acceptable salt of a 4-amino-pyrimidoazepine-phenylurea derivative according to any one of claims 1 to 10.
12. A pharmaceutically acceptable hydrate of a 4-amino-pyrimidoazetidi-n-acyclo-phenylurea derivative as claimed in any one of claims 1 to 10.
13. A pharmaceutical composition comprising the 4-amino-pyrimidoazepine-phenylurea derivative according to any one of claims 1 to 10, the salt according to claim 11 or the hydrate according to claim 12 as an active ingredient, together with a pharmaceutically acceptable carrier.
14. Use of a 4-amino-pyrimidoazepine-phenylurea derivative according to any one of claims 1 to 10, a salt according to claim 11, a hydrate according to claim 12 or a pharmaceutical composition according to claim 13 for the preparation of a FLT3 kinase inhibitor.
15. Use according to claim 14, characterized in that: the FLT3 kinase is a mutant FLT3 kinase.
16. Use according to claim 15, characterized in that: the mutant FLT3 kinase is FLT3/ITD mutant kinase.
17. Use of a 4-amino-pyrimidoazepine-phenylurea derivative according to any of claims 1 to 10, a salt according to claim 11, a hydrate according to claim 12 or a pharmaceutical composition according to claim 13 for the preparation of a medicament for the treatment of a tumor.
18. The use of claim 17, wherein the tumor comprises a solid tumor and/or a hematological tumor.
19. The use of claim 18, the solid tumor comprising: lymphoma, B-cell lymphoma, diffuse large B-cell lymphoma, chronic lymphocytic lymphoma, lymphoplasmacytic lymphoma, ovarian cancer, breast cancer, prostate cancer, bladder cancer, kidney cancer, esophageal cancer, neck cancer, pancreatic cancer, colorectal cancer, gastric cancer, non-small cell lung cancer, thyroid cancer, brain cancer, lymphatic cancer, epidermal hyperplasia, psoriasis and/or prostatic hyperplasia.
20. The use of claim 18, the hematological neoplasm comprising: acute myeloid leukemia, chronic myeloid leukemia, myeloma, acute lymphocytic leukemia, acute myelogenous leukemia, acute promyelocytic leukemia, chronic lymphocytic leukemia, chronic neutrophilic leukemia, acute undifferentiated cell leukemia, myelodysplastic syndrome, myelodysplasia, multiple myeloma, and/or myelosarcoma.
CN202010123847.4A 2019-03-04 2020-02-27 4-amino-pyrimidoazenitrogen heterocycle-phenylurea derivative and preparation method and application thereof Active CN111646995B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910160485 2019-03-04
CN2019101604853 2019-03-04

Publications (2)

Publication Number Publication Date
CN111646995A CN111646995A (en) 2020-09-11
CN111646995B true CN111646995B (en) 2023-03-21

Family

ID=72344703

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010123847.4A Active CN111646995B (en) 2019-03-04 2020-02-27 4-amino-pyrimidoazenitrogen heterocycle-phenylurea derivative and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN111646995B (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112552302B (en) * 2019-09-26 2022-05-13 深圳市塔吉瑞生物医药有限公司 Substituted aromatic fused ring derivatives, compositions and uses thereof
CN112961158B (en) * 2020-03-05 2022-07-01 四川大学华西医院 Aminopyrimidinopyrazole/pyrrole derivative and preparation method and application thereof
MX2022013984A (en) * 2020-05-08 2023-01-30 Halia Therapeutics Inc Inhibitors of nek7 kinase.
WO2021226547A2 (en) * 2020-05-08 2021-11-11 Halia Therapeutics, Inc. Targeted nek7 inhibition for modulation of the nlrp3 inflammasome
US20230203045A1 (en) * 2020-06-08 2023-06-29 Halia Therapeutics, Inc. Inhibitors of nek7 kinase
WO2022212326A1 (en) * 2021-03-29 2022-10-06 Halia Therapeutics, Inc. Nek7 inhibitors
CA3214042A1 (en) * 2021-04-05 2022-10-13 Halia Therapeutics, Inc. Nek7 inhibitors
WO2022226182A1 (en) * 2021-04-22 2022-10-27 Halia Therapeutics, Inc. Nek7 inhibitors
WO2023083373A1 (en) * 2021-11-15 2023-05-19 微境生物医药科技(上海)有限公司 Compound used as src inhibitor

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101675051A (en) * 2007-03-21 2010-03-17 惠氏公司 Pyrazolopyrimidine analogs and their use as mtor kinase and pi3 kinase inhibitors
CN101883774A (en) * 2007-10-16 2010-11-10 惠氏有限责任公司 Thienopyrimidine and Pyrazolopyrimidine compound and as the purposes of MTOR kinases and PI3 kinase inhibitor
CN101899057A (en) * 2010-07-21 2010-12-01 中国药科大学 Preparation method of pyrimido-oxazole derivative and use thereof in medicine
CN102266341A (en) * 2011-06-20 2011-12-07 广州市第十二人民医院 Application of pyrazolopyrimidine compounds in preparing medicines for treating lung cancer
WO2018052120A1 (en) * 2016-09-15 2018-03-22 Riken A hck inhibitor and a bcl-2 inhibitor for treating acute myeloid leukemia
CN107849045A (en) * 2015-04-21 2018-03-27 贵州百灵企业集团制药股份有限公司 Purine radicals N hydroxy pyrimidine carboxamides derivatives and its production and use

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2358720T3 (en) * 2008-10-16 2016-06-06 Univ California Heteroarylkinaseinhibitorer fused-ring
AU2012341028C1 (en) * 2011-09-02 2017-10-19 Mount Sinai School Of Medicine Substituted pyrazolo[3,4-D]pyrimidines and uses thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101675051A (en) * 2007-03-21 2010-03-17 惠氏公司 Pyrazolopyrimidine analogs and their use as mtor kinase and pi3 kinase inhibitors
CN101883774A (en) * 2007-10-16 2010-11-10 惠氏有限责任公司 Thienopyrimidine and Pyrazolopyrimidine compound and as the purposes of MTOR kinases and PI3 kinase inhibitor
CN101899057A (en) * 2010-07-21 2010-12-01 中国药科大学 Preparation method of pyrimido-oxazole derivative and use thereof in medicine
CN102266341A (en) * 2011-06-20 2011-12-07 广州市第十二人民医院 Application of pyrazolopyrimidine compounds in preparing medicines for treating lung cancer
CN107849045A (en) * 2015-04-21 2018-03-27 贵州百灵企业集团制药股份有限公司 Purine radicals N hydroxy pyrimidine carboxamides derivatives and its production and use
WO2018052120A1 (en) * 2016-09-15 2018-03-22 Riken A hck inhibitor and a bcl-2 inhibitor for treating acute myeloid leukemia

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Discovery of 7-Oxo-2,4,5,7-tetrahydro-6H-pyrazolo[3,4-c]pyridine Derivatives as Potent, Orally Available, and Brain-Penetrating Receptor Interacting Protein 1 (RIP1) Kinase Inhibitors: Analysis of Structure–Kinetic Relationships;Masato Yoshikawa,等;《Journal of Medicinal Chemistry》;20180227;第61卷(第6期);第2384-2409页 *
卟啉化合物的研究 Ⅴ。5,10,15-三对磺酰-5’-氟脲嘧啶苯基-20-对磺酸苯基卟啉的合成及抗肿瘤作用的研究;李平英; 林坤华; 黄素秋; 曾凡波;《武汉大学学报(自然科学版)》;19850702(第2期);第111-115页 *

Also Published As

Publication number Publication date
CN111646995A (en) 2020-09-11

Similar Documents

Publication Publication Date Title
CN111646995B (en) 4-amino-pyrimidoazenitrogen heterocycle-phenylurea derivative and preparation method and application thereof
EP2880035B1 (en) Novel pyrrolopyrimidine compounds as inhibitors of protein kinases
AU2008215384B2 (en) Crystalline forms of thiazolidinedione compound and its manufacturing method
JP7215687B2 (en) Compound having kinase inhibitory activity, production method and use thereof
EP3246327B1 (en) 3-acetylenyl-pyrazole-pyrimidine derivative, and preparation method therefor and uses thereof
CN112010839B (en) Crystalline forms of a targeted silk/threonine kinase inhibitor
WO2014082578A1 (en) Heteroaryl alkyne compound and application thereof
CA2586649A1 (en) Combination of a src kinase inhibitor and a bcr-abl inhibitor for the treatment of proliferative diseases
WO2012089106A1 (en) Aromatic alkyne derivative as protein kinase inhibitor and medical use thereof
TWI415613B (en) Anti-cancer agent resistance to overcome the agent
KR20230019110A (en) 4-amino-5-(6-(4-methylpiperazin-1-yl)-1H-benzo[D]imidazol-2-yl)thieno[2,3-b]pyridine-6(7H)- Salts and Crystalline Forms of One
JP6916562B2 (en) Compounds, pharmaceutically acceptable salts thereof, solvates, stereoisomers and tautomers, and drug compositions, hyperproliferative disorder therapeutic agents, hyperproliferative disorder prophylaxis agents, drugs, cancer therapeutic agents, cancer Prophylactic agents and kinase signaling regulators
CN104230952B (en) Compound containing pyrimidine skeleton, and preparation method and use of compound
JP2023548204A (en) CD73 inhibitors and their applications
KR20200084049A (en) 4-amino-N-(1-((3-chloro-2-fluorophenyl)amino)-6-methylisoquinolin-5-yl)thieno[3,2-d]pyrimidine-7-carboxa Salt of the mead, and its crystalline form
CN110903283B (en) Substituted quinazoline compound, pharmaceutical composition containing compound and application of compound
CN108299420B (en) Pentacyclic compounds as selective estrogen receptor down-regulators and uses thereof
CN107001317B (en) Highly selective substituted uracil PI3K inhibitor
CN113416181B (en) Quinazoline derivative and application thereof
CN115368378A (en) Substituted macrocyclic compounds, compositions containing the same and uses thereof
WO2021036814A1 (en) Pyrazole derivative and use thereof
CN105050602A (en) Pyridine compounds used as pi3 kinase inhibitors
CN109438279B (en) Small molecule compound for overcoming EGFR drug-resistant mutation and preparation method and application thereof
KR20210122192A (en) Compounds of Benzothiazole Derivatives
WO2020118753A1 (en) Pan-kit kinase inhibitor having quinoline structure and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant