CN114591334A - Dihydropyrazolopyrimidinone derivatives - Google Patents

Dihydropyrazolopyrimidinone derivatives Download PDF

Info

Publication number
CN114591334A
CN114591334A CN202111465345.0A CN202111465345A CN114591334A CN 114591334 A CN114591334 A CN 114591334A CN 202111465345 A CN202111465345 A CN 202111465345A CN 114591334 A CN114591334 A CN 114591334A
Authority
CN
China
Prior art keywords
membered
cancer
alkyl
amino
hydroxy
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.)
Granted
Application number
CN202111465345.0A
Other languages
Chinese (zh)
Other versions
CN114591334B (en
Inventor
刘斌
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.)
Shandong Xuanzhu Pharma Co Ltd
Original Assignee
Xuanzhu Pharma Co Ltd
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 Xuanzhu Pharma Co Ltd filed Critical Xuanzhu Pharma Co Ltd
Publication of CN114591334A publication Critical patent/CN114591334A/en
Application granted granted Critical
Publication of CN114591334B publication Critical patent/CN114591334B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Abstract

The invention belongs to the technical field of medicines, and particularly relates to a dihydropyrazolopyrimidinone derivative compound serving as a Wee1 kinase inhibitor, a pharmaceutically acceptable salt or a stereoisomer thereof, a pharmaceutical composition and a preparation containing the compound, the pharmaceutically acceptable salt or the stereoisomer thereof, a method for preparing the compound, the pharmaceutically acceptable salt or the stereoisomer thereof, and application of the compound, the pharmaceutically acceptable salt or the stereoisomer thereof.

Description

Dihydropyrazolopyrimidinone derivatives
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a dihydropyrazolopyrimidinone derivative compound, pharmaceutically acceptable salt and a stereoisomer thereof, a pharmaceutical composition and a preparation containing the compound, the pharmaceutically acceptable salt and the stereoisomer thereof, a method for preparing the compound, the pharmaceutically acceptable salt and the stereoisomer thereof, and application of the compound, the pharmaceutically acceptable salt and the stereoisomer thereof.
Background
Cancer is a malignant disease which is difficult to treat all over the world, and has high treatment difficulty and high mortality rate. The recent global tumor statistics in 2018 show that, to date, 1819 ten thousand new cases of cancer and 960 ten thousand cases of cancer death are estimated globally. In 2019, month 1, the national cancer center released the latest national cancer statistics for the first phase. About 392.9 million people develop national malignancies each year, with an average of over 1 million diagnosed as cancer per day and 7.5 diagnosed as cancer per minute. Cancer has become a major disease affecting the health of residents in China, and the prevention and treatment of cancer also faces a severe form.
Currently, radiotherapy and chemotherapy are the most effective means of treating cancer in addition to surgical resection, while radiotherapy is the most effective non-surgical treatment for malignant tumors. Both radiation and a considerable number of anticancer drugs can cause DNA damage. After DNA damage, a series of cellular responses such as damaged DNA repair can be initiated to improve the survival of tumor cells, which is also one of the mechanisms of tumor cells against chemoradiotherapy. If the damaged DNA is not repaired in time and intact, the tumor cells die due to apoptosis or/and mitotic disorders. Therefore, by inhibiting the repair of such DNA damage, the sensitivity of cancer cells to radiotherapy and chemotherapy can be improved, and the proliferation of cells can be inhibited.
Wee1 protein kinase is a member of the serine/threonine protein kinase family, and was first isolated by Nurse et al in fission yeast cells (S.pombe). In humans, Wee1 contains 647 amino acids with a molecular weight of 96 kDa. In the DNA single-strand damage repair channel, Wee1 is positioned at the downstream of an ATR signal channel, after the ATR signal channel is activated, CHK1 is phosphorylated, activated CHK1 activates Wee1, CDC25 is inhibited (the phosphorylation of CDK1/Cyclin B complex is relieved, the activity of regulating and controlling the cell cycle is recovered), CDK1/Cyclin B is further phosphorylated, the CDK1/Cyclin B complex enters an inactive state, the cell cycle is blocked in a G2/M phase, and the time is gained for DNA damage repair; in addition, Weel can also regulate the repair of DNA double strand breaks that occur during DNA replication by phosphorylating CDK2, retarding the S phase of the cell cycle.
In the entire DDR (DNA damage repair) pathway, Wee1 plays a role primarily at the G2/M checkpoint. For p53 mutant tumor cells, the G2/M phase checkpoint is more dependent on the G1/S checkpoint defect itself to repair DNA damage, and the Wee1 inhibitor is more sensitive to p53 mutant tumor cells in terms of mechanism. Furthermore, Wee1 inhibitors can also be combined with other targets on the DDR pathway, such as ATM defects/mutations in tumor cells, combination with ATR inhibitors, etc., to more completely block DNA damage repair, achieving a "synthetic lethal" effect.
In conclusion, the Wee1 kinase inhibitor can not only synergistically enhance the effects of radiotherapy and chemotherapy, effectively inhibit tumor growth, but also reduce damage to normal cells and reduce side effects. At present, the research on the target medicine is still in the clinical experimental stage, and no medicine is on the market. Therefore, the development of a high-efficiency Wee1 kinase inhibitor has important clinical significance, and has wide market prospect in single use or combined use with other medicines.
Disclosure of Invention
The technical problem to be solved by the invention is to provide a dihydropyrazolopyrimidinone derivative compound which has a novel structure and has an inhibitory activity on Wee 1. Furthermore, the compounds can be used for inhibiting the activity of Wee1 kinase, thereby enhancing the immunity of organisms to tumors. Furthermore, the compounds can be used for treating one or more diseases mediated by Wee1, especially cancers. The compounds have good inhibition effect on various cancer cells, and have higher exposure and better in-vivo drug effect in organisms.
The technical scheme of the invention is as follows:
in one aspect, the present invention provides a compound represented by the following general formula (I), a pharmaceutically acceptable salt thereof, or a stereoisomer thereof,
Figure BDA0003391181930000021
wherein the content of the first and second substances,
X1、X2、X3are each independently selected from-C (R)2) -or-N-;
X4、X5each independently selected from-CH-or-N-;
each L is independently selected from-C (R)3)(R4)-、-O-、-N(R5) -or-S-;
ring A is selected from a 3-10 membered cycloalkyl, 3-10 membered heterocyclyl, 6-10 membered aryl or 5-10 membered heteroaryl optionally substituted with 1-4 of Q1;
ring B is selected from 3-10 membered cycloalkyl, 3-10 membered heterocyclyl, 6-10 membered aryl or 5-10 membered heteroaryl optionally substituted with 1-4Q 2;
ring C is selected from 5-15 membered bridged ring group, 5-10 membered spiro ring group, 5-15 membered bridged ring group or 5-10 membered spiroheterocyclic group optionally substituted with 1-4 of Q3, wherein ring carbon atoms in said 5-15 membered bridged ring group, 5-10 membered spiro ring group, 5-15 membered bridged ring group or 5-10 membered spiroheterocyclic group are optionally substituted with oxygen to form a carbonyl group;
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy or the following optionally substituted with 1-3 substituents: c1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, each of which substituents is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, - (CH)2)m-3-10 membered cycloalkyl, - (CH)2)m-3-10 membered heterocycloalkyl, - (CH)2)m-5-10 membered heteroaryl or- (CH)2)m-6-10 membered aryl;
each Q2, each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, halo C1-6Alkoxy, halo C1-6Alkylthio, hydroxy C1-6Alkoxy, hydroxy C1-6Alkylthio, amino C1-6Alkoxy radicalRadical, amino C1-6Alkylthio, - (CH)2)m-3-10 membered cycloalkyl, - (CH)2)m-3-10 membered heterocycloalkyl, - (CH)2)m-5-10 membered heteroaryl or- (CH)2)m-6-10 membered aryl;
R1selected from C optionally substituted by substituents1-6Alkyl radical, C2-6Alkenyl or C2-6Alkynyl, the substituents are respectively and independently selected from halogen, hydroxyl, amino, carboxyl, cyano, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl, halo C1-6Alkoxy or C1-6An alkylcarbonyl group;
R2、R3、R4、R5each independently selected from hydrogen, halogen, hydroxyl, amino, carboxyl and C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl, halo C1-6Alkoxy or C1-6An alkylcarbonyl group;
m and n are respectively and independently selected from 0, 1,2 or 3.
In certain embodiments, the compound, a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, wherein,
ring a is selected from 3-8 membered monocyclic cycloalkyl, 3-8 membered monocyclic heterocyclyl, phenyl or 5-8 membered monocyclic heteroaryl, optionally substituted with 1-4Q 1;
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy or the following optionally substituted with 1-3 substituents: c1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, each of which substituents is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, - (CH)2)m-3-10 membered cycloalkyl, - (CH)2)m-3-10 membered heterocycloalkyl, - (CH)2)m-5-10 membered heteroaryl or- (CH)2)m-6-10 membered aryl;
ring B is selected from phenyl or 5-8 membered mono heteroaryl optionally substituted with 1-4Q 2;
each Q2 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, halo C1-6Alkoxy, halo C1-6Alkylthio, hydroxy C1-6Alkoxy, hydroxy C1-6Alkylthio, amino C1-6Alkoxy, amino C1-6An alkylthio group;
ring C is selected from 5-15 membered bridged heterocyclic group or 5-10 membered spiroheterocyclic group optionally substituted with 1-4 of Q3, wherein ring-forming carbon atoms in said 5-15 membered bridged heterocyclic group or 5-10 membered spiroheterocyclic group are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, halo C1-6Alkoxy, halo C1-6Alkylthio, hydroxy C1-6Alkoxy, hydroxy C1-6Alkylthio, amino C1-6Alkoxy or amino C1-6Alkylthio, - (CH)2)m-3-10 membered cycloalkyl, - (CH)2)m-3-10 membered heterocycloalkyl, - (CH)2)m-5-10 membered heteroaryl or- (CH)2)m-6-10 membered aryl;
R1selected from C optionally substituted by substituents1-6Alkyl radical, C2-6Alkenyl or C2-6Alkynyl, the substituents are respectively and independently selected from halogen, hydroxyl, amino, carboxyl, cyano, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl, halo C1-6Alkoxy or C1-6An alkylcarbonyl group;
R2、R3、R4、R5each independently selected from hydrogen, halogen, hydroxyl, amino, carboxyl and C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl, halo C1-6Alkoxy or C1-6An alkylcarbonyl group;
m and n are respectively and independently selected from 0, 1 or 2.
In certain embodiments, ring a is selected from phenyl or 5-6 membered nitrogen containing monoheteroaryl optionally substituted with 1-3Q 1;
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy or the following optionally substituted with 1-3 substituents: c1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, each of which substituents is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, - (CH)2)m-3-10 membered cycloalkyl, - (CH)2)m-3-10 membered heterocycloalkyl, - (CH)2)m-5-10 membered heteroaryl or- (CH)2)m-6-10 membered aryl;
in certain embodiments, ring a is selected from phenyl or 5-6 membered nitrogen containing monoheteroaryl substituted with 1Q 1;
preferably, ring a is selected from phenyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl or pyridazinyl substituted by 1Q 1; q1 is selected from halogen, hydroxy, amino, nitro, cyano, carboxy or the following optionally substituted with 1-3 substituents: c1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, each of which substituents is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, - (CH)2)m-3-10 membered cycloalkyl, - (CH)2)m-3-10 membered heterocycloalkyl, - (CH)2)m-5-10 membered heteroaryl or- (CH)2)m-6-10 membered aryl.
In certain embodiments, ring a is selected from phenyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, or pyridazinyl substituted with 1Q 1; q1 is selected from halogen, hydroxy, amino, nitro, cyano, carboxy or the following optionally substituted with 1-3 substituents: c1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, each of which substituents is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, - (CH)2)m-3-6 membered cycloalkyl, - (CH)2)m-3-6 membered heterocycloalkyl, - (CH)2)m-5-6 membered heteroaryl or- (CH)2)m-6-8 membered aryl.
In certain embodiments, ring a is selected from pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl substituted with 1Q 1; q1 is selected from halogen, hydroxy, amino, nitro, cyano, carboxy or the following optionally substituted with 1-3 substituents: c1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, wherein the substituents are respectively and independently selected from halogen, hydroxyl, amino, nitro, cyano or carboxyl.
In certain embodiments, ring B is selected from phenyl or 5-6 membered monoheteroaryl optionally substituted with 1 or 2Q 2; each Q2 is independently selected from halogen, hydroxy, amino, nitro, cyano, C1-6Alkyl radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or amino C1-6An alkoxy group.
In certain embodiments, ring B is selected from phenyl, pyridyl, pyrazinyl, or pyridazinyl optionally substituted with 1 or 2Q 2; each Q2 is independently selected from halogen, hydroxy, amino, nitro, cyano, C1-6Alkyl radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxyC1-6Alkoxy or amino C1-6An alkoxy group.
In certain embodiments, ring B is selected from phenyl, pyridyl, pyrazinyl, or pyridazinyl optionally substituted with 1 or 2Q 2; each Q2 is independently selected from fluoro, chloro, bromo, iodo, cyano, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, methylamino, ethylamino, propylamino, isopropylamino, dimethylamino, diethylamino, monofluoromethyl, difluoromethyl, trifluoromethyl, methoxy, ethoxy, propoxy, isopropoxy, monofluoromethoxy, difluoromethoxy, or trifluoromethoxy.
In certain embodiments, ring a is selected from phenyl or 5-6 membered nitrogen containing monoheteroaryl optionally substituted with 1-3Q 1;
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy or the following optionally substituted with 1-3 substituents: c1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, each of which substituents is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, - (CH)2)m-3-10 membered cycloalkyl, - (CH)2)m-3-10 membered heterocycloalkyl, - (CH)2)m-5-10 membered heteroaryl or- (CH)2)m-6-10 membered aryl;
ring B is selected from phenyl or 5-6 membered monoheteroaryl optionally substituted with 1 or 2Q 2; each Q2 is independently selected from halogen, hydroxy, amino, nitro, cyano, C1-6Alkyl radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or amino C1-6An alkoxy group.
In certain embodiments, ring C is selected from a 5-15 membered nitrogen-containing bridged heterocyclic group or a 5-10 membered nitrogen-containing spiro heterocyclic group, optionally substituted with 1-4Q 3, wherein the ring carbon atoms in the 5-15 membered nitrogen-containing bridged heterocyclic group or the 5-10 membered nitrogen-containing spiro heterocyclic group are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, halo C1-6Alkoxy, halo C1-6Alkylthio, hydroxy C1-6Alkoxy, hydroxy C1-6Alkylthio, amino C1-6Alkoxy or amino C1-6An alkylthio group.
In certain embodiments, ring C is selected from 7-9 membered nitrogen-containing bridged heterocyclic group or 7-9 membered nitrogen-containing spiro heterocyclic group, optionally substituted with 1-2Q 3, wherein the ring carbon atoms in said 7-9 membered nitrogen-containing bridged heterocyclic group or 7-9 membered nitrogen-containing spiro heterocyclic group are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy or hydroxy C1-6An alkoxy group.
In certain embodiments, ring C is selected from 7-8 membered nitrogen-containing bridged heterocyclic group or 7-8 membered nitrogen-containing spiro heterocyclic group, optionally substituted with 1-2Q 3, wherein the ring carbon atoms in said 7-8 membered nitrogen-containing bridged heterocyclic group or 7-8 membered nitrogen-containing spiro heterocyclic group are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or 3-4 membered cycloalkyl.
In certain embodiments, ring C is selected from a 7-8 membered 1-2 nitrogen containing bridged heterocyclic group optionally substituted with 1-2Q 3 or a 7-8 membered 1-2 nitrogen containing spiroheterocyclic group, wherein the ring-forming carbon atoms in said bridged heterocyclic group or spiroheterocyclic group are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from halogen, hydroxyAmino, nitro, cyano, carboxyl, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or 3-4 membered cycloalkyl.
In certain embodiments, ring C is selected from a 7-8 membered 2 nitrogen-containing bridged heterocyclyl group optionally substituted with 1-2Q 3, or a 7-8 membered 2 nitrogen-containing spiroheterocyclyl group, wherein the ring-forming carbon atoms in said bridged heterocyclyl or spiroheterocyclyl group are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or 3-4 membered cycloalkyl.
In certain embodiments, ring C is selected from the following optionally substituted with 1-2Q 3:
Figure BDA0003391181930000041
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or 3-4 membered cycloalkyl.
In certain embodiments, ring C is selected from 7-9 membered nitrogen-containing bridged heterocyclic groups optionally substituted with 1-2 of Q3, wherein the ring-forming carbon atoms of said 7-9 membered nitrogen-containing bridged heterocyclic group are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or 3-4 membered cycloalkyl. In certain embodiments, ring C is selected from 7-8 membered 1-containing rings optionally substituted with 1-2Q 3-a bridged heterocyclic group of 2 nitrogens, wherein the ring-forming carbon atoms of said bridged heterocyclic group are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or 3-4 membered cycloalkyl.
In certain embodiments, ring C is selected from the following optionally substituted with 1-2Q 3: 2-azabicyclo [2.2.1] heptanyl, 7-azabicyclo [2.2.1] heptanyl, 3-azabicyclo [3.2.1] octanyl, 8-azabicyclo [3.2.1] octanyl, 2-azabicyclo [2.2.2] octanyl, 2, 5-diazabicyclo [2.2.1] heptanyl, 3, 6-diazabicyclo [3.1.1] heptanyl, 3, 8-diazabicyclo [3.2.1] octanyl, 2-oxa-5-azabicyclo [2.2.1] heptanyl, 8-oxa-3-azabicyclo [3.2.1] octanyl, 3, 8-diazabicyclo [3.2.1] octan-6-enyl or 3, 9-diazabicyclo [3.3.1] nonanyl, wherein the oxycarbonyl group forms a ring carbon atom;
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or 3-4 membered cycloalkyl.
In certain embodiments, ring C is selected from the following optionally substituted with 1-2Q 3:
Figure BDA0003391181930000051
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or 3-to 4-memberedA cycloalkyl group.
In certain embodiments, ring C is selected from the following optionally substituted with 1-2Q 3: 2-azabicyclo [2.2.1] heptanyl, 7-azabicyclo [2.2.1] heptanyl, 3-azabicyclo [3.2.1] octanyl, 8-azabicyclo [3.2.1] octanyl, 2-azabicyclo [2.2.2] octanyl, 2, 5-diazabicyclo [2.2.1] heptanyl, 3, 6-diazabicyclo [3.1.1] heptanyl or 3, 8-diazabicyclo [3.2.1] octanyl, wherein the groups are optionally substituted at a ring carbon atom by oxygen to form a carbonyl group;
each Q3 is independently selected from fluoro, chloro, hydroxy, amino, nitro, cyano, carboxy, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, hydroxymethyl, hydroxyethyl, hydroxyisopropyl, trifluoromethyl, trifluoromethoxy, or cyclopropyl.
In certain embodiments, ring C is selected from the following optionally substituted with 1-2Q 3: 2, 5-diazabicyclo [2.2.2] octane-yl, 2, 5-diazabicyclo [2.2.1] heptylalkyl, 3, 6-diazabicyclo [3.1.1] heptylalkyl or 3, 8-diazabicyclo [3.2.1] octane-yl, wherein the groups are optionally substituted with oxygen to form a carbonyl group at a ring carbon atom;
each Q3 is independently selected from fluoro, chloro, hydroxy, amino, nitro, cyano, carboxy, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, hydroxymethyl, hydroxyethyl, hydroxyisopropyl, trifluoromethyl, trifluoromethoxy, or cyclopropyl.
In certain embodiments, ring C is selected from 7-9 membered nitrogen containing spiroheterocyclic group optionally substituted with 1-2Q 3, wherein ring-forming carbon atoms in said 7-9 membered nitrogen containing spiroheterocyclic group are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or 3-4 membered cycloalkyl.
In certain embodiments, ring C is selected from the group consisting of a 7-8 membered 1-2 nitrogen containing spiroheterocyclic group optionally substituted with 1-2Q 3 wherein the ring-forming carbon atoms in said spiroheterocyclic group are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or 3-4 membered cycloalkyl.
In certain embodiments, ring C is selected from the following optionally substituted with 1-2Q 3: 5-azaspiro [2.4] heptyl, 2-azaspiro [3.3] heptyl, 2-azaspiro [3.5] nonanyl, 2, 6-diazaspiro [3.3] heptanyl, 2, 7-diazaspiro [3.5] nonanyl, 2, 5-diazaspiro [3.4] octanyl, 2-oxa-6-azaspiro [3.3] heptanyl, 6-oxa-2-azaspiro [3.4] octanyl, 6-azaspiro [3.4] octanyl, 2-azaspiro [4.4] nonanyl, 2-oxa-7-azaspiro [4.4] nonanyl, 6-azaspiro [3.4] oct-7-enyl, 2-oxa-6-azaspiro [3.4] oct-7-enyl or 2-azaspiro [4.4] non-7-enyl, wherein said group-forming carbon atoms are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or 3-4 membered cycloalkyl.
In certain embodiments, ring C is selected from the following optionally substituted with 1-2Q 3:
Figure BDA0003391181930000061
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, haloGeneration C1-6Alkoxy, hydroxy C1-6Alkoxy or 3-4 membered cycloalkyl.
In certain embodiments, ring C is selected from the following optionally substituted with 1-2Q 3: 5-azaspiro [2.4] heptylalkyl, 2-azaspiro [3.3] heptylalkyl, 2-azaspiro [3.5] nonanyl, 2, 6-diazaspiro [3.3] heptanyl, 2, 7-diazaspiro [3.5] nonanyl, 2, 5-diazaspiro [3.4] octanyl, 2-oxa-6-azaspiro [3.3] heptanyl, 6-oxa-2-azaspiro [3.4] octanyl, 6-azaspiro [3.4] octanyl, 2-azaspiro [4.4] nonanyl, wherein the groups may optionally be substituted at a ring carbon atom with oxygen to form a carbonyl group;
each Q3 is independently selected from fluoro, chloro, hydroxy, amino, nitro, cyano, carboxy, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, hydroxymethyl, hydroxyethyl, hydroxyisopropyl, trifluoromethyl, trifluoromethoxy, or cyclopropyl.
In certain embodiments, ring C is selected from the following optionally substituted with 1-2Q 3: 2, 6-diazaspiro [3.3] heptanyl, 2, 7-diazaspiro [3.5] nonanyl, 2, 5-diazaspiro [3.4] octanyl, wherein the ring carbon atoms are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from fluoro, chloro, hydroxy, amino, nitro, cyano, carboxy, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, hydroxymethyl, hydroxyethyl, hydroxyisopropyl, trifluoromethyl, trifluoromethoxy, or cyclopropyl. In certain embodiments, ring C is selected from the following optionally substituted with 1-2Q 3: 2-azabicyclo [2.2.1] heptanyl, 7-azabicyclo [2.2.1] heptanyl, 3-azabicyclo [3.2.1] octanyl, 8-azabicyclo [3.2.1] octanyl, 2-azabicyclo [2.2.2] octanyl, 2, 5-diazabicyclo [2.2.1] heptanyl, 3, 6-diazabicyclo [3.1.1] heptanyl, 3, 8-diazabicyclo [3.2.1] octanyl, 2-oxa-5-azabicyclo [2.2.1] heptanyl, 8-oxa-3-azabicyclo [3.2.1] octanyl, 3, 8-diazabicyclo [3.2.1] octan-6-enyl, 3, 9-diazabicyclo [3.3.1] nonanyl, 5-azabicyclo [4.1 ] heptanyl, 2-azaspiro [3.3] heptylalkyl, 2-azaspiro [3.5] nonanyl, 2, 6-diazaspiro [3.3] heptanyl, 2, 7-diazaspiro [3.5] nonanyl, 2, 5-diazaspiro [3.4] octanyl, 2-oxa-6-azaspiro [3.3] heptanyl, 6-oxa-2-azaspiro [3.4] octanyl, 6-azaspiro [3.4] octanyl, 2-azaspiro [4.4] nonanyl, 2-oxa-7-azaspiro [4.4] nonanyl, 6-azaspiro [3.4] oct-7-enyl, 2-oxa-6-azaspiro [3.4] oct-7-enyl or 2-azaspiro [4.4] non-7-enyl, wherein the ring-forming carbon atoms of the group are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy or hydroxy C1-6An alkoxy group.
In certain embodiments, R1Selected from C optionally substituted by substituents2-4Alkenyl, the substituents are respectively and independently selected from halogen, hydroxyl, amino, carboxyl, cyano and C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkyl or halo C1-6An alkoxy group.
In certain embodiments, R1Selected from-CH optionally substituted by a substituent2-CH=CH2The substituents are respectively and independently selected from halogen, hydroxyl, amino, carboxyl, cyano and C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkyl or halo C1-6An alkoxy group.
In certain embodiments, R1Is selected from-CH2-CH=CH2
In certain embodiments, the compounds of the present invention, pharmaceutically acceptable salts thereof, or stereoisomers thereof, wherein X is1、X2、X3Are each independently selected from-C (R)2) -or-N-;
X4、X5each independently selected from-CH-or-N-;
each L is independently selected from-C (R)3)(R4)-、-O-、-N(R5) -or-S-;
R2、R3、R4、R5each independently selected from hydrogen and C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl or halo C1-6An alkoxy group;
R1selected from C optionally substituted by substituents2-6Alkenyl, each of said substituents being independently selected from halogen, hydroxy, amino, carboxy, cyano, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl or halo C1-6An alkoxy group.
In certain embodiments, R2、R3、R4、R5Each independently selected from hydrogen, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, monofluoromethyl, difluoromethyl, trifluoromethyl or trifluoromethoxy.
In certain embodiments, X1、X2、X3Are each independently selected from-C (R)2) -or-N-;
X4、X5each independently selected from-CH-or-N-;
each L is independently selected from-C (R)3)(R4)-、-O-、-N(R5) -or-S-;
R2、R3、R4、R5each independently selected from hydrogen, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, monofluoromethyl, difluoromethyl, trifluoromethyl or trifluoromethoxy;
R1selected from C optionally substituted by substituents2-6Alkenyl, the substituents are respectively and independently selected from halogen, hydroxyl, amino, carboxyl, cyano and C1-6Alkyl radical, C1-6Alkoxy radicalBase, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl or halo C1-6An alkoxy group.
In certain embodiments, R1Selected from C optionally substituted by substituents2-4Alkenyl, preferably propen-3-yl, optionally substituted with substituents each independently selected from halogen, hydroxy, amino, carboxy, cyano, C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkyl or halo C1-6An alkoxy group.
In certain embodiments, X1、X2Each independently selected from-N-.
In certain embodiments, X3is-CH-.
In certain embodiments, X4、X5Are each independently-N-.
In certain embodiments, the compounds of the present invention, pharmaceutically acceptable salts thereof, or stereoisomers thereof, have the structure shown in formula (II) below:
Figure BDA0003391181930000071
wherein, ring A, ring B, ring C, Q1, Q2, Q3, L, R3、R4、R5、R1M, n are as defined above.
In certain embodiments, the compounds of the present invention, pharmaceutically acceptable salts thereof, or stereoisomers thereof, have the structure shown in formula (III) below:
Figure BDA0003391181930000072
wherein, ring A, ring B, ring C, Q1, Q2, Q3, L, R3、R4、R5M, n are as defined above.
In certain embodiments, the compounds of the present invention, pharmaceutically acceptable salts thereof, or stereoisomers thereof, have the structure shown in formula (III) below:
Figure BDA0003391181930000073
wherein ring a is selected from pyridyl, pyrimidinyl, pyrazinyl, or pyridazinyl substituted with 1Q 1; q1 is selected from fluoro, chloro, bromo, hydroxy, amino, nitro, cyano or the following optionally substituted with 1-3 substituents: methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, methoxy, ethoxy, propoxy and isopropoxy, wherein the substituents are respectively and independently selected from fluorine, chlorine, bromine, hydroxyl, amino, nitro, cyano, carboxyl, - (CH)2)m-cyclopropane, - (CH)2)m-cyclobutyl, - (CH)2)m-cyclopentyl or- (CH)2)m-a cyclohexane group;
ring B is phenyl or pyridyl;
each L is independently selected from-C (R)3)(R4) -or-N (R)5)-;R3、R4、R5Each independently selected from hydrogen, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, monofluoromethyl, difluoromethyl, trifluoromethyl, trifluoroethyl, monofluoromethoxy, difluoromethoxy or trifluoromethoxy;
m is selected from 0, 1 or 2; n is 1.
In certain embodiments, the compounds of the present invention, pharmaceutically acceptable salts thereof, or stereoisomers thereof, wherein,
ring a is selected from pyridyl, pyrimidinyl, pyrazinyl, or pyridazinyl substituted with 1Q 1; q1 is selected from fluoro, chloro, bromo, hydroxy, amino, nitro, cyano or the following optionally substituted with 1-3 substituents: methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, methoxy, ethoxy, propoxy and isopropoxy, wherein the substituents are respectively and independently selected from fluorine, chlorine, bromine and hydroxylRadical, amino, nitro, cyano, carboxyl, - (CH)2)m-cyclopropylalkyl, - (CH)2)m-cyclobutyl, - (CH)2)m-cyclopentyl or- (CH)2)m-a cyclohexane group;
ring B is selected from phenyl, pyridyl, pyrimidinyl, pyrazinyl or pyridazinyl.
In certain embodiments, ring B is phenyl or pyridyl.
In certain embodiments, each L is independently selected from-C (R)3)(R4) -or-N (R)5)-;R3、R4、R5Each independently selected from hydrogen, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, monofluoromethyl, difluoromethyl, trifluoromethyl, trifluoroethyl, monofluoromethoxy, difluoromethoxy or trifluoromethoxy.
In certain embodiments, each L is independently selected from-N (R)5)-;R5Each independently selected from hydrogen, methyl, ethyl, propyl, isopropyl, methoxy, trifluoromethyl or trifluoromethoxy; preferably, L is NH.
In certain embodiments, m is selected from 0, 1 or 2.
In certain embodiments, n is selected from 0, 1 or 2; preferably, n is 1.
The technical solutions of the present invention can be combined with each other to form a new technical solution, and the formed new technical solution is also included in the scope of the present invention.
In certain embodiments, the compound of formula (I), formula (II), or formula (III), a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, is selected from the group consisting of:
Figure BDA0003391181930000081
Figure BDA0003391181930000091
the invention also provides a pharmaceutical composition, which contains the compound shown in the general formula (I), the general formula (II) or the general formula (III), the pharmaceutically acceptable salt or the stereoisomer thereof, and one or more pharmaceutical carriers and/or diluents; the pharmaceutical composition can be prepared into any clinically or pharmaceutically acceptable dosage form, such as tablets, capsules, pills, granules, solutions, suspensions, syrups, injections (including injection, sterile powder for injection and concentrated solution for injection), suppositories, inhalants or sprays and the like.
In certain embodiments of the invention, the above-described pharmaceutical formulations may be administered to a patient or subject in need of such treatment by oral, parenteral, rectal, or pulmonary administration, and the like. For oral administration, the pharmaceutical composition can be prepared into oral preparations, for example, conventional oral solid preparations such as tablets, capsules, pills, granules and the like; it can also be made into oral liquid, such as oral solution, oral suspension, syrup, etc. When the composition is formulated into oral preparations, appropriate filler, binder, disintegrating agent, lubricant, etc. can be added. For parenteral administration, the pharmaceutical preparations can also be prepared into injections, including injections, sterile powders for injection, and concentrated solutions for injection. The injection can be prepared by conventional method in the existing pharmaceutical field, and can be prepared without adding additives or adding suitable additives according to the properties of the medicine. For rectal administration, the pharmaceutical composition may be formulated as a suppository or the like. For pulmonary administration, the pharmaceutical composition may be formulated as an inhalant or a spray.
The pharmaceutically acceptable carrier and/or diluent useful in the pharmaceutical composition or pharmaceutical formulation of the present invention may be any conventional carrier and/or diluent in the art of pharmaceutical formulation, and the selection of a particular carrier and/or diluent will depend on the mode of administration or the type and state of the disease used to treat a particular patient. The preparation of suitable pharmaceutical compositions for a particular mode of administration is well within the knowledge of those skilled in the pharmaceutical art.
In another aspect, the present invention also relates to the use of a compound of the aforementioned formula (I), formula (II) or formula (III), a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, for the preparation of a medicament for the prevention and/or treatment of diseases mediated by Wee1 and related diseases, which can be used in combination with one or more other medicaments for the prevention or treatment of diseases mediated by Wee1 and related conditions. The disease and related conditions are selected from cancer or benign tumors, including carcinoma in situ and metastatic carcinoma. Further, the cancer includes, but is not limited to, lung cancer, squamous cell carcinoma, bladder cancer, stomach cancer, ovarian cancer, peritoneal cancer, pancreatic cancer, breast cancer, head and neck cancer, cervical cancer, endometrial cancer, rectal cancer, liver cancer, kidney cancer, esophageal adenocarcinoma, esophageal squamous cell carcinoma, prostate cancer, thyroid cancer, cancer of the female genital tract, lymphoma, neurofibroma, bone cancer, skin cancer, brain cancer, colon cancer, testicular cancer, small cell lung cancer, non-small cell lung cancer, gastrointestinal stromal tumor, mast cell tumor, multiple myeloma, melanoma, leukemia, glioma, or sarcoma, and the like.
Further, the invention also relates to the application of a pharmaceutical preparation containing the compound shown in the general formula (I), the general formula (II) or the general formula (III), the pharmaceutically acceptable salt thereof or the stereoisomer thereof in preparing a medicament which can be combined with one or more medicaments for treating and/or preventing diseases mediated by Wee1 and related symptoms.
In another aspect, the invention is directed to medicaments containing a compound of formula (I), formula (II), or formula (III), pharmaceutically acceptable salts, or stereoisomers thereof, as described above, alone or in combination with one or more second therapeutically active agents that are useful in combination with the Wee1 inhibitor compounds of the present application for the treatment and/or prevention of diseases and related conditions mediated by Wee 1. Thus, in certain embodiments, the pharmaceutical composition further comprises one or more second therapeutically active agents. In certain embodiments, the second therapeutically active agent is selected from the group consisting of anti-cancer agents, including mitotic inhibitors, alkylating agents, anti-metabolites, antisense DNA or RNA, anti-tumor antibiotics, growth factor inhibitors, signaling inhibitors, cell cycle inhibitors, enzyme inhibitors, retinoid receptor modulators, proteasome inhibitors, topoisomerase inhibitors, biological response modifiers, hormonal agents, angiogenesis inhibitors, cell growth inhibitors, targeting antibodies, HMG-CoA reductase inhibitors, and prenyl protein transferase inhibitors.
In certain embodiments, the ingredients to be combined (e.g., the compound of the invention, a pharmaceutically acceptable salt thereof, a stereoisomer thereof, and the second therapeutically active agent) may be administered simultaneously or separately, sequentially and separately. For example, the second therapeutically active agent may be administered before, simultaneously with, or after the administration of the compound of the present invention, a pharmaceutically acceptable salt thereof, or a stereoisomer thereof. Furthermore, the components to be combined may also be administered in combination in the same formulation or in separate and distinct formulations.
In another aspect, the present invention also provides a method for treating diseases mediated by Wee1 and related disorders, comprising administering to a patient in need thereof an effective amount of a compound of formula (I) as described above, a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, a formulation or pharmaceutical composition as described above; the diseases and related conditions mediated by Wee1 are as defined above.
By "effective amount" is meant a dosage of a drug that reduces, delays, inhibits or cures a condition in a subject. The size of the administered dose is determined by the administration mode of the drug, the pharmacokinetics of the medicament, the severity of the disease, the individual physical signs (sex, weight, height, age) of the subject, and the like.
The medicine containing the compound shown in the general formula (I), the general formula (II) or the general formula (III), the pharmaceutically acceptable salt or the stereoisomer thereof can be combined with radiotherapy to improve the treatment effect of the radiotherapy, and can be used as a radiotherapy sensitizer.
In another aspect, the compounds of the invention may also be used as sensitizers for other anticancer agents in the field of cancer therapy. The anticancer agent is as described above. The sensitizer means a drug which enhances the therapeutic effect of radiotherapy and/or chemotherapy by being used in combination with radiotherapy and/or chemotherapy using an anticancer agent, additively or synergistically, in the field of cancer treatment.
[ detailed description of the invention ]
In the present invention, unless otherwise defined, scientific and technical terms used herein have meanings commonly understood by those skilled in the art, however, in order to better understand the present invention, definitions of some terms are provided below. To the extent that the definitions and explanations of terms provided herein do not conform to the meanings commonly understood by those skilled in the art, the definitions and explanations of terms provided herein shall control.
The "halogen" as referred to herein means a fluorine atom, a chlorine atom, a bromine atom or an iodine atom.
"C" according to the invention1-6Alkyl "denotes straight or branched alkyl having 1 to 6 carbon atoms, including for example" C1-4Alkyl group "," C1-3Alkyl group "," C1-2Alkyl group "," C2-6Alkyl group "," C2-5Alkyl group "," C2-4Alkyl group "," C2-3Alkyl group "," C3-6Alkyl group "," C3-5Alkyl group "," C3-4Alkyl "and the like, specific examples include, but are not limited to: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, isopentyl, 2-methylbutyl, neopentyl, 1-ethylpropyl, n-hexyl, isohexyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 3-dimethylbutyl, 2-dimethylbutyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 1, 2-dimethylpropyl, and the like. "C" according to the invention1-4Alkyl "means C1-6Specific examples of the alkyl group having 1 to 4 carbon atoms.
"C" according to the invention1-6Alkoxy "means" C1-6alkyl-O- ", said" C1-6Alkyl "is as defined above. "C" according to the invention1-4Alkoxy "means" C1-4alkyl-O- ", said" C1-4Alkyl "is as defined above.
"C" according to the invention1-6Alkyl sulfideThe radical "means" C1-6alkyl-S- ", said" C1-6Alkyl "is as defined above. "C" according to the invention1-4Alkylthio "means" C1-4alkyl-S- ", said" C1-4Alkyl "is as defined above.
The "hydroxyl group C" of the present invention1-6Alkyl, amino C1-6Alkyl, halo C1-6Alkyl "means C1-6One or more hydrogens of the alkyl group are each replaced by one or more hydroxyl, amino or halogen. C1-6Alkyl is as previously defined
The "hydroxyl group C" of the present invention1-6Alkoxy, amino C1-6Alkoxy, halo C1-6Alkoxy "means" C1-6One or more hydrogens of "alkoxy" are replaced with one or more hydroxy, amino, or halogen.
The "hydroxy group C" of the present invention1-6Alkylthio, amino C1-6Alkylthio, halo C1-6Alkylthio "means" C1-6Alkylthio "is one in which one or more hydrogens are replaced with one or more hydroxy, amino, or halogen.
"C" according to the invention1-6Alkylamino radical, di (C)1-6Alkyl) amino "means independently C1-6alkyl-NH-),
Figure BDA0003391181930000101
"C" according to the invention2-6Alkenyl "means a straight, branched or cyclic alkenyl group having 2 to 6 carbon atoms containing at least one double bond, and includes, for example," C2-4Alkenyl groups "and the like. Examples thereof include, but are not limited to: vinyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 1, 3-butadienyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 1, 3-pentadienyl, 1, 4-pentadienyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 1, 4-hexadienyl, cyclopentenyl, 1, 3-cyclopentadienyl, cyclohexenyl, 1, 4-cyclohexadienyl and the like.
"C" according to the invention2-6Alkynyl means containing at least one triple bond and having the number of carbon atomsStraight-chain or branched alkynyl radicals having 2 to 6 inclusive, e.g. "C2-4Alkynyl "and the like. Examples include, but are not limited to: ethynyl, propynyl, 2-butynyl, 2-pentynyl, 3-pentynyl, 4-methyl-2-pentynyl, 2-hexynyl, 3-hexynyl, 5-methyl-2-hexynyl and the like.
The "3-to 10-membered cycloalkyl" as referred to herein is meant to include "3-to 8-membered monocycloalkyl" and "8-to 10-membered fused ring alkyl".
The "3-to 8-membered monocyclic alkyl" as used herein means a saturated or partially saturated monocyclic cyclic alkyl group having 3 to 8 carbon atoms and having no aromaticity, and includes "3-to 8-membered saturated monocyclic alkyl" and "3-to 8-membered partially saturated monocyclic alkyl"; preferred are "3-to 4-membered monocycloalkyl", "3-to 5-membered monocycloalkyl", "3-to 6-membered monocycloalkyl", "3-to 7-membered monocycloalkyl", "4-to 5-membered monocycloalkyl", "4-to 6-membered monocycloalkyl", "4-to 7-membered monocycloalkyl", "5-to 6-membered monocycloalkyl", "5-to 7-membered monocycloalkyl", "6-to 8-membered monocycloalkyl", "7-to 8-membered monocycloalkyl", "3-to 6-membered saturated monocycloalkyl", "5-to 8-membered saturated monocycloalkyl", "5-to 7-membered saturated monocycloalkyl", "5-to 6-membered saturated monocycloalkyl", and the like. Specific examples of said "3-to 8-membered saturated monocycloalkyl" include, but are not limited to: a cyclopropane group (cyclopropyl), a cyclobutane group (cyclobutyl), a cyclopentyl group (cyclopentyl), a cyclohexane group (cyclohexyl), a cycloheptyl group (cycloheptyl), a cyclooctyl group (cyclooctyl), etc.; specific examples of the "3-to 8-membered partially saturated monocycloalkyl" include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohex-1, 3-diene, cyclohex-1, 4-diene, cycloheptenyl, cyclohepta-1, 3-dienyl, cyclohepta-1, 4-dienyl, cyclohepta-1, 3, 5-trienyl, cyclooctenyl, cycloocta-1, 3-dienyl, cycloocta-1, 4-dienyl, cycloocta-1, 5-dienyl, cycloocta-1, 3, 5-trienyl, cyclooctatetraenyl and the like.
The 8-10-membered fused ring group is a saturated or partially saturated non-aromatic cyclic group containing 8-10 ring atoms, which is formed by two or more cyclic structures sharing two adjacent carbon atoms, wherein one ring in the fused ring can be an aromatic ring, but the fused ring does not have aromatic property as a whole; including "8-9-membered fused ring group", "9-10-membered fused ring group", etc., the fusion mode may be: 5-6 membered cycloalkyl and 5-6 membered cycloalkyl, benzo 5-6 membered saturated cycloalkyl and the like. Examples include, but are not limited to: bicyclo [3.1.0] hexanyl, bicyclo [4.1.0] heptanyl, bicyclo [2.2.0] hexanyl, bicyclo [3.2.0] heptanyl, bicyclo [4.2.0] octanyl, octahydropentanyl, octahydro-1H-indenyl, decahydronaphthyl, tetradecahydrophenanthryl, bicyclo [3.1.0] hex-2-enyl, bicyclo [4.1.0] hept-3-enyl, bicyclo [3.2.0] hept-3-enyl, bicyclo [4.2.0] oct-3-enyl, 1,2,3,3 a-tetrahydropentanyl, 2,3,3a,4,7,7 a-hexahydro-1H-indenyl, 1,2,3,4,4a,5,6,8 a-octahydronaphthyl, 1,2,4a,5,6,8 a-hexahydronaphthyl, 1,2,3,4,5, 8 a-hexahydronaphthyl, 10-decahydrophenanthryl, benzocyclopentyl, benzocyclohexyl, benzocyclohexenyl, benzocyclopentenyl, and the like.
The "3-to 10-membered heterocyclic group" described in the present invention includes "3-to 8-membered heteromonocyclic group" and "8-to 10-membered fused heterocyclic group".
The "3-to 8-membered heteromonocyclic group" according to the present invention means a saturated or partially saturated and non-aromatic monocyclic cyclic group containing at least one hetero atom (e.g., 1,2,3,4 or 5) which is a nitrogen atom, an oxygen atom and/or a sulfur atom and has 3 to 8 ring atoms, and optionally, a ring atom (e.g., a carbon atom, a nitrogen atom or a sulfur atom) in the cyclic structure may be oxo. The "3-to 8-membered heteromonocyclic group" described in the present invention includes "3-to 8-membered saturated heteromonocyclic group" and "3-to 8-membered partially saturated heteromonocyclic group". Preferably, the "3-8 membered heteromonocyclic group" described herein contains 1-3 heteroatoms; preferably, the "3-to 8-membered heteromonocyclic group" of the present invention contains 1 to 2 hetero atoms selected from nitrogen atom and/or oxygen atom; preferably, the "3-to 8-membered heteromonocyclic group" described herein contains 1 nitrogen atom. The "3-to 8-membered heteromonocyclic group" is preferably "3-to 7-membered heteromonocyclic group", "3-to 6-membered heteromonocyclic group", "4-to 7-membered heteromonocyclic group", "4-to 6-membered heteromonocyclic group", "6-to 8-membered heteromonocyclic group", "5-to 7-membered heteromonocyclic group", "5-to 6-membered heteromonocyclic group", "3-to 6-membered saturated nitrogen-containing heteromonocyclic group", "5-to 6-membered saturated nitrogen-containing heteromonocyclic group", and the like. For example, containing only 1 or 2 nitrogen atoms, or, alternatively, containing one nitrogen atom and 1 or 2 other heteroatoms (e.g., oxygen and/or sulfur atoms). Specific examples of "3-8 membered heteromonocyclic group" include, but are not limited to: aziridinyl, 2H-aziridinyl, diazacyclopropenyl, 3H-diazacyclopropenyl, azetidinyl, 1, 4-dioxanyl, 1, 3-dioxolanyl, 1, 4-dioxadienyl, tetrahydrofuryl, dihydropyrrolyl, pyrrolidinyl, imidazolidinyl, 4, 5-dihydroimidazolyl, pyrazolidinyl, 4, 5-dihydropyrazolyl, 2, 5-dihydrothienyl, tetrahydrothienyl, 4, 5-dihydrothiazolyl, thiazolidinyl, piperidinyl, tetrahydropyridinyl, piperidonyl, piperazinyl, morpholinyl, 4, 5-dihydrooxazolyl, 4, 5-dihydroisoxazolyl, 2, 3-dihydroisoxazolyl, 3H-diazacyclopropenyl, imidazolidinyl, 4, 5-dihydroimidazolyl, pyrazolidinyl, 4, 5-dihydrothiazolyl, thiazolidinyl, piperidyl, piperidonyl, tetrahydropyridinyl, piperidyl, piperazinyl, morpholinyl, 4, 5-dihydroisoxazolyl, 2, 3-dihydroisoxazolyl, and the like, Oxazolidinyl, 2H-1, 2-oxazinyl, 4H-1, 2-oxazinyl, 6H-1, 2-oxazinyl, 4H-1, 3-oxazinyl, 6H-1, 3-oxazinyl, 4H-1, 4-oxazinyl, 4H-1, 3-thiazinyl, 6H-1, 3-thiazinyl, 2H-pyranyl, 2H-pyran-2-onyl, 3, 4-dihydro-2H-pyranyl and the like.
The "8-to 10-membered fused heterocyclic group" as used herein refers to a saturated or partially saturated, nonaromatic cyclic group containing 8 to 10 ring atoms, wherein at least one ring atom of the fused ring may be an aromatic ring, but the fused ring as a whole does not have aromaticity, which is formed by two or more cyclic structures sharing two adjacent atoms with each other, and at least one ring atom of the fused ring is a heteroatom, which is a nitrogen atom, an oxygen atom and/or a sulfur atom, and optionally, a ring atom (e.g., a carbon atom, a nitrogen atom or a sulfur atom) of the cyclic structure may be oxo, and includes, but is not limited to, "8-to 9-membered fused heterocyclic group", "9-to 10-membered fused heterocyclic group" and the like, and may be fused in such a manner that the fused ring is a 5-to 6-membered heterocyclic group, a 5-to 6-membered cycloalkyl group, Benzo 5-6 membered heterocyclyl, benzo 5-6 membered saturated heterocyclyl, 5-6 membered heteroarylo 5-6 membered saturated heterocyclyl; 5-6 membered heteroaryl is as previously defined; specific examples of the "8-to 10-membered fused heterocyclic group" include, but are not limited to: pyrrolidinyl cyclopropyl, cyclopent aziridinyl, pyrrolidinyl cyclobutyl, pyrrolidinyl, pyrrolidinyl piperidinyl, pyrrolidinyl piperazinyl, pyrrolidinyl morpholinyl, piperidinyl morpholinyl, benzopyrrolidinyl, benzocyclopentyl, benzocyclohexyl, benzotetrahydrofuranyl, benzopyrrolidinyl, benzimidazolyl, benzoxazolinyl, benzothiazolinyl, benzisoxazolidinyl, benzisothiazolidinyl, benzopyriperidinyl, benzomorpholinyl, benzopyrazinyl, benzopyranyl, pyridocyclopentyl, pyridocyclohexyl, pyridotetrahydrofuranyl, pyridopyrrolidinyl, pyridoimidazolidinyl, pyridooxazolidinyl, pyridothiazolidinyl, pyridoisoxazolidinyl, pyridoisothiazolidinyl, pyridopiperidinyl, pyridomorpholinyl, and the like, Pyridopiperazinyl, pyridotetrahydropyranyl, pyrimidocyclopentyl, pyrimidocyclohexyl, pyrimidotetrahydrofuranyl, pyrimidopyrrolidinyl, pyrimidoimidazoimidazolidinyl, pyrimidooxazolidoalkyl, pyrimidoiizolidinyl, pyrimidoiisoxazolidinyl, pyrimidoiisothiazolidinyl, pyrimidoipiperidinyl, pyrimidoimorpholinyl, pyrimidoipiperazinyl, pyrimidoitetrahydropyranyl; tetrahydroimidazo [4,5-c ] pyridyl, 3, 4-dihydroquinazolinyl, 1, 2-dihydroquinoxalinyl, benzo [ d ] [1,3] dioxolyl, 2H-chromenyl, 2H-chromen-2-one, 4H-chromenyl, 4H-chromen-4-one, 4H-1, 3-benzoxazinyl, 4, 6-dihydro-1H-furo [3,4-d ] imidazolyl, 3a,4,6,6 a-tetrahydro-1H-furo [3,4-d ] imidazolyl, 4, 6-dihydro-1H-thieno [3,4-d ] imidazolyl, 4, 6-dihydro-1H-pyrrolo [3,4-d ] imidazolyl, octahydro-benzo [ d ] imidazolyl, decahydroquinolinyl, hexahydrothienoimidazolyl, hexahydrofuroimidazolyl, 4,5,6, 7-tetrahydro-1H-benzo [ d ] imidazolyl, octahydro-cyclopenta [ c ] pyrrolyl, 4H-1, 3-benzoxazinyl and the like.
The term "benzocyclopentyl", the structure of which refers to
Figure BDA0003391181930000121
(also referred to as 2, 3-dihydro-1H-indenyl); the term "benzo-pyrrolidine" structurally includes
Figure BDA0003391181930000122
Etc.; the term "pyridotetrahydrofuranyl" includes
Figure BDA0003391181930000123
Figure BDA0003391181930000124
Specific examples of the other "other condensed heterocyclic group as defined above" have a cyclic structure similar thereto.
The "6-to 10-membered aryl" as referred to in the present invention includes "6-to 8-membered monocyclic aryl" and "8-to 10-membered fused ring aryl".
The "6-to 8-membered monocyclic aryl" as referred to herein means a monocyclic aryl group containing 6 to 8 ring carbon atoms, examples of which include, but are not limited to: phenyl, cyclooctatetraenyl and the like; phenyl is preferred.
The "8-to 10-membered fused ring aryl" as referred to herein means an unsaturated aromatic cyclic group having 8 to 10 ring carbon atoms, formed by two or more cyclic structures sharing two adjacent atoms with each other, and is preferably a "9-to 10-membered fused ring aryl", and specific examples thereof are naphthyl and the like.
The "5-to 10-membered heteroaryl" as referred to herein includes "5-to 8-membered monoheteroaryl" and "8-to 10-membered fused heteroaryl".
The "5-to 8-membered monoheteroaryl group" according to the present invention means a monocyclic cyclic group having aromaticity, which contains 5 to 8 ring atoms, at least one of which is a heteroatom such as nitrogen atom, oxygen atom or sulfur atom. Optionally, a ring atom (e.g., a carbon atom, a nitrogen atom, or a sulfur atom) in the cyclic structure may be oxo. The "5-to 8-membered monoheteroaryl group" includes, for example, "5-to 7-membered monoheteroaryl group", "5-to 6-membered nitrogen-containing monoheteroaryl group", "6-membered nitrogen-containing monoheteroaryl group", and the like, in which the hetero atom contains at least one nitrogen atom, for example, contains only 1 or 2 nitrogen atoms, or contains one nitrogen atom and the other 1 or 2 hetero atoms (for example, oxygen atom and/or sulfur atom), or contains 2 nitrogen atoms and the other 1 or 2 hetero atoms (for example, oxygen atom and/or sulfur atom). Specific examples of "5-to 8-membered monocyclic heteroaryl" include, but are not limited to, furyl, thienyl, pyrrolyl, thiazolyl, isothiazolyl, thiadiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, imidazolyl, pyrazolyl, 1,2, 3-triazolyl, 1,2, 4-triazolyl, 1,2, 3-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,3, 4-oxadiazolyl, pyridyl, 2-pyridonyl, 4-pyridonyl, pyrimidinyl, pyridazinyl, pyrazinyl, 1,2, 3-triazinyl, 1,3, 5-triazinyl, 1,2,4, 5-tetrazinyl, azepinyl, 1, 3-diazacycloheptenyl, azepinyl, and the like. The "5-6 membered monoheteroaryl" refers to a specific example containing 5 to 6 ring atoms in the 5-8 membered heteroaryl.
The "8-to 10-membered fused heteroaryl group" as used herein refers to an unsaturated aromatic cyclic structure having 8 to 10 ring atoms (at least one of which is a heteroatom such as nitrogen atom, oxygen atom or sulfur atom) formed by two or more cyclic structures sharing two adjacent atoms with each other. Optionally, a ring atom (e.g., a carbon atom, a nitrogen atom, or a sulfur atom) in the cyclic structure may be oxo. Including "9-10 membered fused heteroaryl", "8-9 membered fused heteroaryl", etc., which can be fused in a benzo-5-6 membered heteroaryl, 5-6 membered heteroaryl and 5-6 membered heteroaryl, etc.; specific examples include, but are not limited to: pyrrolopyrrole, pyrrolofuran, pyrazolopyrrole, pyrazolothiophene, furothiophene, pyrazoloxazole, benzofuranyl, benzisofuranyl, benzothiophenyl, indolyl, isoindolyl, benzoxazolyl, benzimidazolyl, indazolyl, benzotriazolyl, quinolinyl, 2-quinolinonyl, 4-quinolinonyl, 1-isoquinolinyl, acridinyl, phenanthridinyl, pyridazinyl, phthalazinyl, quinazolinyl, quinoxalinyl, purinyl, naphthyridinyl, and the like.
The 5-10 membered spiro ring group is two or moreCyclic structures containing from 5 to 10 ring carbon atoms formed by the upper cyclic structures sharing a carbon atom with each other. Optionally, the carbon atoms in the cyclic structure may be oxidized. The "5-to 10-membered spiro ring group" includes, for example, "4-to 10-membered spiro ring group", "6-to 10-membered spiro ring group", "7-to 10-membered spiro ring group", "6-to 9-membered spiro ring group", "7-to 9-membered spiro ring group", "9-to 10-membered spiro ring group" and the like. Specific examples include, but are not limited to:
Figure BDA0003391181930000131
Figure BDA0003391181930000132
Figure BDA0003391181930000133
and the like. The "7-9 membered spirocyclic group" means a specific example containing 7 to 9 ring atoms in the 5-10 membered spirocyclic group.
The "5-15 membered bridged cyclic group" as used herein means a cyclic structure containing 5 to 15 ring carbon atoms, which is formed by two or more cyclic structures sharing two non-adjacent carbon atoms with each other. Optionally, the carbon atom in the cyclic structure may be oxo. "5-15-membered bridge ring group" includes, for example, "5-11-membered bridge ring group", "6-11-membered bridge ring group", "5-10-membered bridge ring group", "7-10-membered bridge ring group", "6-9-membered bridge ring group", "7-9-membered bridge ring group", "9-10-membered bridge ring group" and the like. Specific examples include, but are not limited to:
Figure BDA0003391181930000134
Figure BDA0003391181930000135
Figure BDA0003391181930000136
and the like. The "7-to 9-membered bridged ring group" means a specific example in which the 5-to 15-membered bridged ring group contains 7 to 9 ring atoms.
The 5-to 10-membered spiro complex of the inventionThe "cyclic group" means a cyclic structure containing 5 to 10 ring atoms (at least one of which is a hetero atom such as a nitrogen atom, an oxygen atom or a sulfur atom) formed by two or more cyclic structures sharing one ring atom with each other, and includes "5 to 10-membered saturated spiroheterocyclic group" and "5 to 10-membered partially saturated spiroheterocyclic group". Optionally, a ring atom (e.g., a carbon atom, a nitrogen atom, or a sulfur atom) in the cyclic structure may be oxidized. The "5-to 10-membered spiroheterocyclic group" includes, for example, "6-to 11-membered spiroheterocyclic group", "6-to 9-membered spiroheterocyclic group", "7-to 9-membered spiroheterocyclic group", "9-to 10-membered spiroheterocyclic group", "5-to 10-membered nitrogen-containing spiroheterocyclic group", "7-to 9-membered nitrogen-containing spiroheterocyclic group", "8-to 9-membered nitrogen-containing spiroheterocyclic group", "7-to 9-membered nitrogen-containing saturated spiroheterocyclic group", "8-to 9-membered nitrogen-containing saturated spiroheterocyclic group" and the like. Specific examples include, but are not limited to:
Figure BDA0003391181930000141
Figure BDA0003391181930000142
Figure BDA0003391181930000143
and the like.
The "5-to 15-membered bridged heterocyclic group" as used herein means a cyclic structure containing 5 to 15 ring atoms (at least one of which is a hetero atom such as a nitrogen atom, an oxygen atom or a sulfur atom) formed by two or more cyclic structures sharing two non-adjacent ring atoms with each other, and includes "5-to 15-membered saturated bridged heterocyclic group" and "5-to 15-membered partially saturated bridged heterocyclic group". Optionally, a ring atom (e.g., a carbon atom, a nitrogen atom, or a sulfur atom) in the cyclic structure may be oxidized. The "5-to 15-membered bridged heterocyclic group" includes, for example, "5-to 10-membered bridged heterocyclic group", "6-to 11-membered bridged heterocyclic group", "6-to 9-membered bridged heterocyclic group", "6-to 10-membered bridged heterocyclic group", "7-to 9-membered nitrogen-containing bridged heterocyclic group", "7-to 8-membered nitrogen-containing bridged heterocyclic group", "5-to 9-membered nitrogen-containing bridged heterocyclic group", "5-to 15-membered nitrogen-containing bridged heterocyclic group", "5-to 10-membered bridged heterocyclic group", "7-to 9-membered nitrogen-containing bridged heterocyclic groupNitrogen-saturated bridged heterocyclic group ", and the like. Specific examples include, but are not limited to:
Figure BDA0003391181930000144
Figure BDA0003391181930000145
Figure BDA0003391181930000146
and so on.
The expression "carbon atom, nitrogen atom or sulfur atom is oxo" as used herein means that C-O, N-O, S-O or SO is formed2The structure of (1).
The term "optionally substituted" as used herein means both the case where one or more hydrogen atoms on a substituent may be "substituted" or "unsubstituted" by one or more substituents.
"pharmaceutically acceptable salt" as used herein refers to an acidic functional group (e.g., -COOH, -OH, -SO) present in a compound3H, etc.) with a suitable inorganic or organic cation (base), including salts with alkali or alkaline earth metals, ammonium salts, and salts with nitrogen-containing organic bases; and salts of basic functional groups present in the compounds (e.g., -NH2, etc.) with suitable inorganic or organic anions (acids), including salts with inorganic or organic acids (e.g., carboxylic acids, etc.).
"isomers" as used herein refers to compounds of the present invention when they contain one or more asymmetric centers and thus may be present as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. The compounds of the present invention may have asymmetric centers that each independently produce two optical isomers. The scope of the present invention includes all possible optical isomers and mixtures thereof. The compounds of the present invention, if they contain an olefinic double bond, include cis-isomers and trans-isomers, unless otherwise specified. The compounds of the invention may exist in tautomeric (one of the functional group isomers) forms having different points of attachment of hydrogen through one or more double bond shifts, e.g., a ketone and its enol form are keto-enol tautomers. The compounds of the present invention contain a spiro ring structure, and substituents on the ring may be present on both sides of the ring to form the opposite cis (cis) and trans (trans) isomers, depending on the steric structure of the ring. Each tautomer and mixtures thereof are included within the scope of the present invention. All enantiomers, diastereomers, racemates, meso, cis-trans isomers, tautomers, geometric isomers, epimers, mixtures thereof and the like of the compounds are included within the scope of the present invention.
The compounds of the invention may be prepared by enantiospecific synthesis or by resolution from a mixture of enantiomers in such a way as to give the individual enantiomers. Conventional resolution techniques include the formation of salts of the free base of each of the enantiomers of an enantiomeric pair using optically active acids (followed by fractional crystallization and regeneration of the free base), the formation of salts of the acid form of each of the enantiomers of an enantiomeric pair using optically active amines (followed by fractional crystallization and regeneration of the free acid), the formation of esters or amides of each of the enantiomers of an enantiomeric pair using optically pure acids, amines or alcohols (followed by chromatographic separation and removal of the chiral auxiliary), or the resolution of mixtures of the enantiomers of the starting materials or final products using various well-known chromatographic methods.
When the stereochemistry of the disclosed compounds is named or depicted by structure, the named or depicted stereoisomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% pure by weight relative to the other stereoisomers. When a single isomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% pure by weight. The optical purity wt% is the ratio of the weight of an enantiomer to the weight of the enantiomer plus the weight of its optical isomer.
The "dosage form" of the present invention refers to a form prepared from the drug suitable for clinical use, including, but not limited to, powders, tablets, granules, capsules, solutions, emulsions, suspensions, injections (including injections, sterile powders for injections and concentrated solutions for injections), sprays, aerosols, powders, lotions, liniments, ointments, plasters, pastes, patches, gargles or suppositories, more preferably powders, tablets, granules, capsules, solutions, injections, ointments, gargles or suppositories.
Advantageous effects of the invention
1. The compound, the pharmaceutically acceptable salt thereof or the stereoisomer thereof has excellent Wee1 activity inhibition effect, has good pharmacokinetic property in organisms, has lasting effect and high exposure and bioavailability, and can treat and/or prevent diseases mediated by Wee 1.
2. The compound, the pharmaceutically acceptable salt thereof or the stereoisomer thereof has a better therapeutic effect on Wee 1-mediated cancer.
3. The compound, the pharmaceutically acceptable salt thereof or the stereoisomer thereof can synergistically enhance the effects of radiotherapy and chemotherapy, effectively inhibit the growth of tumors, and can reduce the damage to normal cells and reduce side effects.
4. The compound of the invention has simple preparation process, high medicine purity, stable quality and easy large-scale industrial production.
Detailed description of the preferred embodiments
The technical solutions of the present invention will be described below in conjunction with the specific embodiments, and the above-mentioned contents of the present invention will be further described in detail, but it should not be understood that the scope of the above-mentioned subject matter of the present invention is limited to the following examples. All the technologies realized based on the above contents of the present invention belong to the scope of the present invention.
Example 1: preparation of 2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -6- ((4- (8-methyl-3, 8-diazabicyclo [3.2.1] oct-3-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (compound 1)
Preparation of ethyl 4- (2-allyl-2- (tert-butoxycarbonyl) hydrazino) -2- (methylthio) pyrimidine-5-carboxylate
Figure BDA0003391181930000161
Tert-butyl 1-allylhydrazine-1-carboxylate (0.37g,21.5mmol) and ethyl 4-chloro-2- (methylthio) pyrimidine-5-carboxylate (0.5g,21.5mmol) were dissolved in tetrahydrofuran (100.0mL), N-diisopropylethylamine (0.69g,53.8mmol) was added, reacted at 70 ℃ for 16h, the solvent was dried, and column chromatography (ethyl acetate/petroleum ether ═ 25%) gave the product (0.62g, 78.5%).
Preparation of ethyl 4- (2-allylhydrazino) -3- (methylthio) -1,2, 4-triazine-6-carboxylate
Figure BDA0003391181930000162
Ethyl 4- (2-allyl-2- (tert-butoxycarbonyl) hydrazino) -2- (methylthio) pyrimidine-5-carboxylate (0.62g,16.8mmol) was dissolved in trifluoroacetic acid (50.0mL), reacted at 70 ℃ for 1 hour, and spin-dried to give the crude product which was used directly in the next step.
Preparation of 2-allyl-6- (methylthio) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one
Figure BDA0003391181930000163
4- (2-allylhydrazino) -3- (methylthio) -1,2, 4-triazine-6-carboxylic acid ethyl ester (crude in the above step) was dissolved in 70.0mL of ethanol, an aqueous solution of sodium hydroxide (25.6mL,179mmol) was added, the reaction was carried out at 20 ℃ for 30 minutes, 3N diluted hydrochloric acid was added to adjust the pH to 4, dichloromethane and 5% methanol (50.0mL) were extracted, and the organic phase was dried by spinning to give 4.0g of crude product.
Preparation of 2- (6-bromopyridin-2-yl) propan-2-ol
Figure BDA0003391181930000164
Methyl 6-bromopicolinate (5.0g, 23.3mmol) was dissolved in 150mL tetrahydrofuran, and methyl magnesium bromide solution (16.0mL,100.0mmol) was added at 0 deg.C, reacted at 25 deg.C for 2h, quenched with saturated ammonium chloride solution, extracted with ethyl acetate, and concentrated by column chromatography to give 4.5g, 90% yield.
Preparation of 2-allyl-1- (6- (2- (hydroxyprop-2-yl) pyridin-2-yl) -6- (methylthio) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one
Figure BDA0003391181930000165
2-allyl-6- (methylthio) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (crude from the above step) was dissolved in 1, 4-dioxane (150mL), cuprous iodide (3.4g,18.0mmol), potassium carbonate (3.5g,25.2mmol), N' -dimethylethylenediamine (3.2g, 36.0mmol), 2- (6-bromopyridin-2-yl) propan-2-ol (5.1g,23.4mmol) were added, reacted at 95 ℃ for 16H, and column chromatography was concentrated (ethyl acetate/petroleum ether ═ 50%) to give 4.0g of product, three-step total yield: 66.61 percent.
Preparation of tert-butyl (1R,5S) -3- (4-nitrophenyl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylate
Figure BDA0003391181930000171
Tert-butyl 3, 8-diazabicyclo [3.2.1] octane-8-carboxylate (0.5g,2.4mmol) was dissolved in N, N-dimethylacetamide (10.0mL), 1-fluoro-4-nitrobenzene (0.33g,2.4mmol), N-diisopropylethylamine (0.9g,7.2mmol) was added, reacted at 90 ℃ for 16h, and column chromatography was concentrated (ethyl acetate/petroleum ether ═ 25%) to give the product (0.33g, 41.3%).
Preparation of (1R,5S) -3- (4-nitrophenyl) -3, 8-diazabicyclo [3.2.1] octane
Figure BDA0003391181930000172
Tert-butyl (1R,5S) -3- (4-nitrophenyl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylate (0.15g,0.45mmol), dissolved in 10.0mL of dichloromethane, added with 3.0mL of trifluoroacetic acid, reacted at room temperature for 1h, and spin-dried to give 190.0mg of crude product.
Preparation of (1R,5S) -8-methyl-3- (4-nitrophenyl) -3, 8-diazabicyclo [3.2.1] octane
Figure BDA0003391181930000173
(1R,5S) -3- (4-nitrophenyl) -3, 8-diazabicyclo [3.2.1] octane (crude product from the previous step) was dissolved in 12.0mL of N, N-dimethylformamide, sodium triacetoxyborohydride (0.3g,1.6mmol) and a formaldehyde solution (0.2g, 5.4mmol) were added, the reaction was carried out at 20 ℃ for 3 hours, 10.0mL of water was added to quench the reaction, and dichloromethane was extracted and concentrated to obtain 0.27 g.
Preparation of 9.4- ((1R,5S) -8-methyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) aniline
Figure BDA0003391181930000174
(1R,5S) -8-methyl-3- (4-nitrophenyl) -3, 8-diazabicyclo [3.2.1] octane (0.25g, 1.0mmol), dissolved in 6.0mL of methanol, 0.2g of palladium on carbon (10%), reacted for 1h, and concentrated to give 0.23g of crude product.
Preparation of 2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -6- ((4- (8-methyl-3, 8-diazabicyclo [3.2.1] oct-3-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one
Figure BDA0003391181930000175
2-allyl-1- (6- (2- (hydroxypropan-2-yl) pyridin-2-yl) -6- (methylthio) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (0.29g, 0.81mmol), dissolved in 3.0mL of toluene, 85% m-chloroperoxybenzoic acid (m-CPBA,0.21g,1.2mmol) was added and reacted at room temperature for 30 minutes, N-diisopropylethylamine (0.54g,4.2mmol), 4- ((1R,5S) -8-methyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) aniline (0.23g,1.1mmol) was added and reacted at room temperature for 16 hours, saturated sodium bicarbonate was reacted, extracted with ethyl acetate, dried, concentration and reverse phase column chromatography (water/methanol 60%) gave 23.0mg, 5.5% yield.
Molecular formula C29H34N8O2Molecular weight 526.0LC-MS (M/e):527.0(M + H)+)
1H-NMR(400MHz,CDCl3)δ:8.81(s,1H),7.84(t,J=7.8Hz,1H),7.75(d,J=8.0Hz,1H),7.51-7.39(m,2H),7.32(d,J=7.6Hz,1H),6.78(d,J=8.2Hz,2H),5.74-5.65(m,1H),5.03(d,J=10.1Hz,1H),4.93(d,J=17.2Hz,1H),4.73(d,J=6.0Hz,2H),3.96(s,1H),3.34(d,J=10.5Hz,2H),3.28(s,2H),3.02(d,J=10.5Hz,2H),2.36(s,3H),2.04-2.03(m,2H),1.79-1.77(m,2H),1.58(s,6H).
Example 2: preparation of 2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -6- ((4- (5-methyl-2, 5-diazabicyclo [2.2.2] oct-2-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (compound 2)
Preparation of 2-methyl-5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.2] octane
Figure BDA0003391181930000181
2-methyl-2, 5-diazabicyclo [2.2.2] octane (100mg,0.5mmol) was dissolved in 10ml of acetonitrile, and 1-fluoro-4-nitrobenzene (141mg,1.0mmol) and N, N-diisopropylethylamine (323mg,2.5mmol) were added and reacted at 60 ℃ for 16 hours. Concentration and silica gel column purification (methanol: dichloromethane ═ 1:7) gave 74mg of the objective compound in a yield of 60.0%.
Preparation of 4- (5-methyl-2, 5-diazabicyclo [2.2.2] oct-2-yl) aniline
Figure BDA0003391181930000182
2-methyl-5- (4-nitrophenyl) -2, 5-diazabicyclo [2.2.2] octane (70mg,0.28mmol) was dissolved in methanol (5mL), and palladium on carbon (40mg) was added to the solution to conduct a reaction at 16 ℃ for 1 hour under a hydrogen atmosphere. Concentration gave the crude product (72mg) which was used directly in the next reaction.
Preparation of 2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -6- ((4- (5-methyl-2, 5-diazabicyclo [2.2.2] oct-2-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one
Figure BDA0003391181930000183
2-allyl-1- (6- (2- (hydroxypropan-2-yl) pyridin-2-yl) -6- (methylthio) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (77mg,0.22mmol) was dissolved in toluene (10mL), 62.5% m-CPBA (77mg,0.28mmol) was added and reacted at 16 ℃ for 1 hour, N-diisopropylethylamine (148mg,1.14mmol) and 4- (5-methyl-2, 5-diazabicyclo [2.2.2] oct-2-yl) aniline (70mg,0.32mmol) were added and reacted at 16 ℃ for 5 hours, followed by concentration and purification with a C18 column (water: methanol ═ 3:1) to give 29mg of the title compound in 25.0% yield.
Molecular formula C29H34N8O2Molecular weight 526.6LC-MS (M/e):527.2(M + H)+)
1H-NMR(400MHz,DMSO-d6)δ:10.12(s,1H),8.79(s,1H),8.03(s,1H),7.76(d,J=7.6Hz,1H),7.60(d,J=8.0Hz,1H),7.59-7.52(m,2H),6.64(d,J=8.4Hz,2H),5.76-5.63(m,1H),5.32(s,1H),5.00(d,J=10.4Hz,1H),4.85-4.81(m,1H),4.69-4.67(m,2H),3.86(s,1H),3.63(d,J=9.6Hz,1H),3.23(d,J=9.2Hz,1H),2.90(d,J=8.8Hz,1H),2.33(s,3H),2.03-1.96(m,2H),1.83-1.56(m,3H),1.44(s,6H).
Example 3: preparation of 2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -6- (((4- (6-methyl-2, 6-diazaspiro [3.3] hept-2-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (compound 3)
Preparation of 2-methyl-6- (4-nitrophenyl) -2, 6-diazaspiro [3.3] heptane
Figure BDA0003391181930000191
2-methyl-2, 6-diazaspiro [3.3] heptane dihydrochloride (100mg,0.54mmol), p-fluoronitrobenzene (83mg,0.59mmol) and N, N-diisopropylethylamine (349mg,2.7mmol) were dissolved in acetonitrile (20mL) and reacted at 60 ℃ for 6h, the reaction was completed, a saturated sodium bicarbonate (3mL) solution was added, concentrated, and the crude product was purified by silica gel column chromatography (dichloromethane: methanol ═ 10:1) to give the product (100mg, yield 79.1%).
Preparation of 4- (6-methyl-2, 6-diazaspiro [3.3] hept-2-yl) aniline
Figure BDA0003391181930000192
2-methyl-6- (4-nitrophenyl) -2, 6-diazaspiro [3.3] heptane (30mg,0.13mmol) was dissolved in methanol (10mL), and palladium on carbon (15mg) was added to react at 25 ℃ for 0.5 hour under a hydrogen atmosphere. Filtration and concentration gave the crude product (28mg) which was used directly in the next reaction.
Preparation of 2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -6- (((4- (6-methyl-2, 6-diazaspiro [3.3] hept-2-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one
Figure BDA0003391181930000193
2-allyl-1- (6- (2- (hydroxypropan-2-yl) pyridin-2-yl) -6- (methylthio) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (32mg, 0.09mmol) was dissolved in toluene (7mL), m-chloroperoxybenzoic acid (20mg, 0.12mmol) was added and the reaction was completed at 25 ℃ for 0.5 hour, a solution of N, N-diisopropylethylamine (69mg, 0.53mmol) and 4- (6-methyl-2, 6-diazaspiro [3.3] hept-2-yl) aniline (28mg) in toluene (7mL) was added and the mixture was reacted at 25 ℃ for 18 hours, followed by concentration, purification with a C18 column (water: methanol ═ 1:4) to obtain the crude target compound (20 mg), purification by prep-TLC (dichloromethane: methanol: 12:1) gave 18mg of crude target compound, which was purified by preparative liquid phase purification under high pressure (water: methanol: 1:5) to give 2.3mg of product in 5.0% yield.
Molecular formula C28H32N8O2Molecular weight 512.3LC-MS (M/e):513.2(M + H)+)
1H-NMR(400MHz,CDCl3)δ:8.83(s,1H),7.86(t,J=8.0Hz,1H),7.76(d,J=8.0Hz,1H),7.54-7.26(m,4H),6.46(d,J=8.0Hz,2H),5.76-5.67(m,1H),5.07-4.94(m,2H),4.75(d,J=6.0Hz,2H),3.96(s,4H),3.42(s,4H),2.35(s,3H),1.60(s,6H).
Example 4: preparation of 6- (4- ((2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3,4-d ] pyrimidin-6-yl) amino) phenyl) -3-methyl-3, 6-diazabicyclo [3.1.1] hept-2-one (compound 4)
Preparation of tert-butyl 3-methyl-2-oxo-3, 6-diazabicyclo [3.1.1] heptane-6-carboxylate
Figure BDA0003391181930000194
Tert-butyl 2-oxo-3, 6-diazabicyclo [3.1.1] heptane-6-carboxylate (106mg,0.5mmol) was dissolved in tetrahydrofuran (10mL), 60% sodium hydride (40mg,1mmol) was added, reaction was carried out at 25 ℃ for 2h, iodomethane (142mg,1mmol) was added, reaction was carried out at 25 ℃ for 2h, quenching was carried out with saturated saline (20mL), extraction was carried out with ethyl acetate (30mL), the organic phase was dried over anhydrous sodium sulfate and spin-dried to give the crude product, which was used directly in the next step.
Preparation of 2.3-methyl-3, 6-diazabicyclo [3.1.1] hept-2-one hydrochloride
Figure BDA0003391181930000201
The crude product of 3-methyl-2-oxo-3, 6-diazabicyclo [3.1.1] heptane-6-carboxylic acid tert-butyl ester obtained in the previous step was dissolved in 4M dioxane hydrochloride (2mL), reacted at 20 ℃ for 16 hours, and spin-dried to obtain a crude product, which was used directly in the next step.
Preparation of 3-methyl-6- (4-nitrophenyl) -3, 6-diazabicyclo [3.1.1] heptan-2-one
Figure BDA0003391181930000202
The crude 3-methyl-3, 6-diazabicyclo [3.1.1] hept-2-one hydrochloride salt from the above step, 1-fluoro-4-nitrobenzene (141mg,1mmol) and diisopropylethylamine (194mg,1.5mmol) were dissolved in DMA (7mL), reacted at 90 ℃ for 16 hours, quenched with saturated brine (15mL), extracted with ethyl acetate (30mL), and organic phase spin-dried medium pressure reverse phase preparation (0-40% methanol/water) gave the product (40mg, 32.4%).
Preparation of 6- (4-aminophenyl) -3-methyl-3, 6-diazabicyclo [3.1.1] heptan-2-one
Figure BDA0003391181930000203
3-methyl-6- (4-nitrophenyl) -3, 6-diazabicyclo [3.1.1] heptan-2-one (50mg,0.2mmol) was reacted with 10% wet palladium on carbon (5mg) in methanol (7mL) under hydrogen (15Psi) at 15 ℃ for 1 hour, and the filtrate was filtered with suction and concentrated to give the product (40mg, 91.0%).
Preparation of 2-allyl-1- (6- (2- (hydroxyprop-2-yl) pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one
Figure BDA0003391181930000204
2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -6- (methylthio) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (100mg,0.28mmol) was reacted with 65% m-chloroperoxybenzoic acid (89mg,0.34mmol) in toluene (2mL) at 15 ℃ for 3 hours, concentrated, and purified by column chromatography (30% tetrahydrofuran/ethyl acetate) to give the product (80mg, 76.6%).
Preparation of 6- (4- ((2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3,4-d ] pyrimidin-6-yl) amino) phenyl) -3-methyl-3, 6-diazabicyclo [3.1.1] hept-2-one
Figure BDA0003391181930000211
2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (103mg,0.27mmol), 6- (4-aminophenyl) -3-methyl-3, 6-diazabicyclo [3.1.1] heptan-2-one (40mg,0.18mmol), and diisopropylethylamine (48mg,0.36mmol) were dissolved in THF (12mL) and reacted at 15 ℃ for 16 hours, spun dry, and purified by medium pressure reverse phase preparation (0-40% acetonitrile/water) to give the product (10mg, 10.3%).
The molecular formula is as follows: C28H30N8O3 molecular weight: 526.6LC-MS (M/e):527.2(M + H +)
1HNMR(400MHz,CDCl3):δ:10.11(s,1H),8.82(s,1H),8.10-7.95(m,1H),7.80-7.55(m,2H),7.55-7.45(m,2H),6.60-6.50(m,2H),5.70-5.60(m,1H),5.33(s,1H),5.05-4.95(m,1H),4.85-4.75(m,1H),4.75-4.65(m,2H),4.60-4.50(m,1H),4.10-4.05(m,1H),3.65-3.55(m,1H),3.30-3.20(m,1H),2.80-2.70(m,1H),2.57(m,3H),1.95-1.85(m,1H),1.45(s,6H).
Example 5: preparation of 3- (4- ((2-allyl-1- (6- (2-hydroxypropyl-2-yl) pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3,4-d ] pyrimidin-6-yl) amino) phenyl) -6-methyl-3, 6-diazabicyclo [3.1.1] heptan-2-one (Compound 5)
Preparation of tert-butyl 3- (4-nitrophenyl) -2-oxo-3, 6-diazabicyclo [3.1.1] heptane-6-carboxylate
Figure BDA0003391181930000212
2-oxo 3, 6-diazabicyclo [3.1.1] heptane-6-carboxylic acid tert-butyl ester (149mg,0.70mmol), p-nitroiodobenzene (262mg,1.05mmol), cuprous iodide (27mg,0.14mmol), potassium phosphate (149mg,3.5mmol) and 1, 10-phenanthroline (25mg,0.14mmol) are dissolved in DMF (15mL), reacted at 110 ℃ for 5 hours, quenched with saturated saline (20mL), extracted with ethyl acetate (30mL), dried with anhydrous sodium sulfate of organic phase, spin-dried, and purified by column chromatography (ethyl acetate/petroleum ether ═ 30%) to obtain the product (140mg, 59.8%).
Preparation of 3- (4-Nitrophenyl) -3, 6-diazabicyclo [3.1.1] heptan-2-one trifluoroacetate salt
Figure BDA0003391181930000213
Tert-butyl 3- (4-nitrophenyl) -2-oxo-3, 6-diazabicyclo [3.1.1] heptane-6-carboxylate (140mg,0.42mmol) was dissolved in dichloromethane (10mL) and trifluoroacetic acid (5mL), reacted at 20 ℃ for 1 hour, and spin-dried to give the crude product which was used directly in the next step.
Preparation of 3.6-methyl-3- (4-nitrophenyl) -3, 6-diazabicyclo [3.1.1] heptan-2-one
Figure BDA0003391181930000221
The crude 3- (4-nitrophenyl) -3, 6-diazabicyclo [3.1.1] heptan-2-one trifluoroacetate salt from the previous step, 37% aqueous formaldehyde (341mg,4.2mmol) and sodium borohydride acetate (267mg,1.26mmol) were dissolved in DMF (6mL) and reacted at 20 ℃ for 2 hours, quenched with saturated sodium bicarbonate (20mL), extracted with ethyl acetate (30mL), dried over anhydrous sodium sulfate for the organic phase, spun dry and purified by column chromatography (methanol/dichloromethane ═ 4%) to give the product (85mg, 81.9%).
Preparation of 3- (4-aminophenyl) -6-methyl-3, 6-diazabicyclo [3.1.1] heptan-2-one
Figure BDA0003391181930000222
6-methyl-3- (4-nitrophenyl) -3, 6-diazabicyclo [3.1.1] heptan-2-one (85mg,0.34mmol) was reacted with 10% wet palladium on carbon (8.5mg) in methanol (12mL) at 15 ℃ under hydrogen (15Psi) for 1 hour, filtered with suction, and the filtrate was concentrated to give the product (70mg, 93.7%).
Preparation of 3- (4- ((2-allyl-1- (6- (2-hydroxypropyl-2-yl) pyridin-2-yl) -3-oxo-2, 3-dihydro-1H-pyrazolo [3,4-d ] pyrimidin-6-yl) amino) phenyl) -6-methyl-3, 6-diazabicyclo [3.1.1] heptan-2-one
Figure BDA0003391181930000223
2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (103mg,0.27mmol) and 3- (4-aminophenyl) -6-methyl-3, 6-diazabicyclo [3.1.1] heptan-2-one (48mg,0.22mmol) were dissolved in toluene (8mL) and reacted at 40 ℃ for 16 hours, spun dry, medium pressure reverse phase preparation (0-40% acetonitrile/water) to give the product (20mg, 17.2%).
The molecular formula is as follows: c28H30N8O3Molecular weight: 526.6LC-MS (M/e):527.2(M + H)+)
1HNMR(400MHz,CDCl3):δ:10.38(s,1H),8.91(s,1H),8.15-8.05(m,1H),7.80-7.70(m,3H),7.65-7.55(m,1H),7.50-7.40(m,2H),5.70-5.60(m,1H),5.36(s,1H),5.05-4.95(m,1H),4.85-4.75(m,1H),4.75-4.65(m,2H),3.90-3.70(m,3H),3.55-3.45(m,1H),2.60-2.50(m,1H),2.25(m,3H),1.85-1.80(m,1H),1.45(s,6H).
Example 6: preparation of 2-allyl-6- ((4- (8-cyclopropyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) phenyl) amino) -1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (compound 6)
Preparation of 1.8-cyclopropyl-3- (4-nitrophenyl) -3, 8-diazabicyclo [3.2.1] octane
Figure BDA0003391181930000224
Dissolving 3- (4-nitrophenyl) -3, 8-diazabicyclo [3.2.1] octane trifluoroacetate (2.2g, crude product) in methanol (30mL), adding sodium cyanoborohydride (3.8g,60.5mmol), acetic acid (5.7mL,99.6mmol) and (1-ethoxycyclopropoxy) trimethylsilane (3.8g,21.8mmol) in sequence at 0 ℃, reacting for 4h at 60 ℃, detecting the completion of the reaction by LC-MS, diluting the reaction liquid with water, extracting a dichloromethane/methanol (10:1) mixed solvent, drying an organic phase, spin-drying, and separating the residue by column chromatography (dichloromethane: methanol ═ 10:1) to obtain the target compound (1.5 g).
Preparation of 4- (8-cyclopropyl-3, 8-diazabicyclo [3.2.1] octyl-3-yl) aniline
Figure BDA0003391181930000231
Dissolving 8-cyclopropyl-3- (4-nitrophenyl) -3, 8-diazabicyclo [3.2.1] octane (0.6g, 2.2mmol) in methanol (20mL), adding palladium/carbon (1.0g), replacing with hydrogen for three times, reacting at 25 ℃ for 1.5h, detecting by LC-MS (liquid chromatography-mass spectrometry) to finish the reaction, filtering the reaction solution, and spin-drying the filtrate to obtain a crude target compound (480mg), wherein the crude target compound is directly used in the next step.
Preparation of 2-allyl-6- ((4- (8-cyclopropyl-3, 8-diazabicyclo [3.2.1] oct-3-yl) phenyl) amino) -1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one
Figure BDA0003391181930000232
Dissolving 2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (580mg, crude) in toluene (20mL), adding 4- (8-cyclopropyl-3, 8-diazabicyclo [3.2.1] octyl-3-yl) aniline (400mg, crude) and DIEA (628mg, 4.86mmol), reacting at 25 ℃ for 14H, detecting by LC-MS, draining the reaction solution, separating by thin-layer chromatography (dichloromethane: methanol ═ 12:1, Rf ∼ 0.5) to obtain crude (500mg), separating by reverse phase column chromatography (C18 column, water: methanol ═ 90:10-40:60) to obtain target compound (98mg, yield 11%).
Molecular formula C31H36N8O2Molecular weight 552.7LC-MS (M/z):553.1(M + H)+)
1H-NMR(400MHz,CD3OD)δ:8.82(s,1H),7.88-7.82(m,3H),7.42-7.25(m,3H),6.80-6.73(m,2H),5.75-5.65(m,1H),5.04(d,1H,J=10.4),4.95(d,1H,J=17.2),4.76-4.74(m,2H),4.00(s,1H),3.50-3.46(m,3H),3.38-3.33(m,2H),3.31-2.90(m,2H),2.15-2.00(m,2H),1.93-1.80(m,2H),1.70-1.30(m,6H),0.55-0.45(m,4H).
Example 7: preparation of 2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -6- ((4- (7-methyl-4, 7-diazaspiro [2.5] octyl-4-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (compound 8)
Preparation of tert-butyl 4- (4-nitrophenyl) -4, 7-diazaspiro [2.5] octane-7-carboxylate
Figure BDA0003391181930000233
1-iodo-4-nitrobenzene (2.1g,8.3mmol) was dissolved in toluene (40mL), 4, 7-diazaspiro [2.5] octane-7-carboxylic acid tert-butyl ester (1.5g, 7.1mmol), bis (tri-tert-butylphosphine) palladium (562mg, 1.1mmol) and sodium tert-butoxide (1.3g, 13.8mmol) were added, reacted at 110 ℃ for 4h, LCMS detected that the reaction was complete, the reaction solution was poured into water, EA extracted, dried over anhydrous sodium sulfate, the organic phase was spin-dried, and chromatographed on silica gel column (EA: PE ═ 1: 3) to give the title compound (1.8g, yield 76.0%).
Preparation of 4- (4-Nitrophenyl) -4, 7-diazaspiro [2.5] octane trifluoroacetate
Figure BDA0003391181930000241
Tert-butyl 4- (4-nitrophenyl) -4, 7-diazaspiro [2.5] octane-7-carboxylate (900mg,2.7mmol) was dissolved in dichloromethane (8mL), trifluoroacetic acid (2mL) was added dropwise at 25 ℃ and reacted at 25 ℃ for 1h, the reaction was completed by LCMS detection, and the reaction solution was spin-dried to give a crude product (1.4 g).
Preparation of 3.7-methyl-4- (4-nitrophenyl) -4, 7-diazaspiro [2.5] octane
Figure BDA0003391181930000242
4- (4-Nitrophenyl) -4, 7-diazaspiro [2.5] octane trifluoroacetate (1.4g, crude product) was dissolved in N, N-dimethylformamide (20mL), an aqueous formaldehyde solution (37%) (2.2g, 27mmol) and sodium triacetoxyborohydride (1.7g,8.1mmol) were added, the mixture was left to react at 25 ℃ for 1 hour, water was added to the system to quench the reaction, the organic phase was extracted with dichloromethane, the organic phase was concentrated, and the product was purified with silica gel (methanol/dichloromethane ═ 1:10) to obtain the objective compound (650mg, yield in two steps 97.4%).
Preparation of 4- (7-methyl-4, 7-diazaspiro [2.5] octan-4-yl) aniline
Figure BDA0003391181930000243
7-methyl-4- (4-nitrophenyl) -4, 7-diazaspiro [2.5] octane (650mg, 2.63mmol) was dissolved in methanol (20mL), palladium on carbon (300mg) was added, the mixture was reacted at 25 ℃ for 1h, suction filtered, and the filtrate was concentrated to give a crude product (600mg) which was used directly in the next reaction.
Preparation of 2-allyl-1- (6- (2-hydroxyprop-2-yl) pyridin-2-yl) -6- ((4- (7-methyl-4, 7-diazaspiro [2.5] octyl-4-yl) phenyl) amino) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one
Figure BDA0003391181930000244
2-allyl-1- (6-2-hydroxypropan-2-yl) pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (440mg,1.18mmol) was dissolved in toluene (20mL), 4- (7-methyl-4, 7-diazaspiro [2.5] octan-4-yl) aniline (395mg, crude) and DIEA (458mg, 3.54mmol) were added, the reaction was allowed to react for 16H at 25 ℃, LCMS was checked for completion, the reaction was spun dry, silica gel column chromatography (MeOH: DCM ═ 1:10) to obtain a crude product, which was then slurried with ethyl acetate to obtain the desired compound (15mg, yield 2.6%).
Molecular formula C29H34N8O2Molecular weight 526.6LC-MS (M/e):527.1(M + H)+)
1H-NMR(400MHz,MeOD)δ:8.81(s,1H),7.95-8.05(m,1H),7.70-7.80(m,1H),7.62-7.68(m,1H),7.55-7.61(m,2H),7.06-7.09(m,2H),5.65-5.75(m,1H),5.05(d,J=10.4Hz,1H),4.90-4.96(m,1H),4.80-4.90(m,2H),4.60(s,1H),3.80-4.00(m,2H),2.95-3.10(m,3H),2.81(s,3H),1.59(s,6H),1.10-1.20(m,2H),0.90-1.10(m,2H).
Example 8: preparation of 2-allyl-6- ((4- (4-cyclopropyl-4, 7-diazaspiro [2.5] octan-7-yl) phenyl) amino) -1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (compound 9)
Preparation of tert-butyl 1.7- (4-nitrophenyl) -4, 7-diazaspiro [2.5] octane-4-carboxylate
Figure BDA0003391181930000251
To dimethyl sulfoxide (50.0mL) of 1-fluoro-4-nitrobenzene (1.0g,7.1mmol) were added tert-butyl 4, 7-diazaspiro [2.5] octane-4-carboxylate (1.7g, 8.0mmol) and potassium carbonate (2.0g, 14.2mmol) and reacted at 70 ℃ for 5 hours, the reaction mixture was poured into water, the solid was filtered by suction, the filter cake was washed with water, and the filter cake was spin-dried to give the desired compound (2.4g, yield: 99.7%).
Preparation of 7- (4-nitrophenyl) -4, 7-diazaspiro [2.5] octane
Figure BDA0003391181930000252
After trifluoroacetic acid (6.0mL) was added to 7- (4-nitrophenyl) -4, 7-diazaspiro [2.5] octane-4-carboxylic acid tert-butyl ester (1.8g,5.4mmol) in dichloromethane (12.0mL), the mixture was allowed to react at 25 ℃ for 1 hour, the reaction was concentrated and dried, diluted with water, adjusted to alkaline with saturated sodium bicarbonate solution, extracted with dichloromethane, the organic phase was dried over anhydrous sodium sulfate, and concentrated and dried directly for the next step.
Preparation of 4-cyclopropyl-7- (4-nitrophenyl) -4, 7-diazaspiro [2.5] octane
Figure BDA0003391181930000253
After adding acetic acid (1.3g,22.3mmol), sodium cyanoborohydride (841.5mg,13.4mmol) and (1-ethoxycyclopropoxy) trimethylsilane (836.6g,4.8mmol) to methanol (15.0mL) of 7- (4-nitrophenyl) -4, 7-diazaspiro [2.5] octane (crude) (500.0mg, 2.1mmol), the mixture was allowed to react at 65 ℃ for 5 hours, the system was quenched with water, extracted with dichloromethane, the organic phase was concentrated, and purified with a silica gel column (ethyl acetate/petroleum ether ═ 1:2) to obtain the objective compound (560.0mg, yield 95.6%).
Preparation of 4- (4-cyclopropyl-4, 7-diazaspiro [2.5] octyl-7-yl) aniline
Figure BDA0003391181930000254
To 4-cyclopropyl-7- (4-nitrophenyl) -4, 7-diazaspiro [2.5] octane (560.0mg, 2.0mmol) in methanol (6.0mL) was added palladium on charcoal (200mg), and the mixture was reacted at 25 ℃ for 1h, followed by suction filtration, and the filtrate was concentrated to give a crude product (500.0mg) which was used directly in the next reaction.
Preparation of 2-allyl-6- ((4- (4-cyclopropyl-4, 7-diazaspiro [2.5] octan-7-yl) phenyl) amino) -1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one
Figure BDA0003391181930000255
In 2-allyl-1- (6- (2-hydroxypropan-2-yl) pyridin-2-yl) -6- (methylsulfinyl) -1, 2-dihydro-3H-pyrazolo [3,4-d ] pyrimidin-3-one (crude, 500.0mg,1.3mmol) in toluene (20.0mL), n, N-diisopropylethylamine (840.5mg,6.5mmol) and 4- (4-cyclopropyl-4, 7-diazaspiro [2.5] octan-7-yl) aniline (486.8mg,2.0mmol) were added, and the mixture was reacted at 25 ℃ for 16 hours, quenched with saturated sodium bicarbonate, extracted with dichloromethane, the organic phase was concentrated, and purified with silica gel (methanol/dichloromethane ═ 1:20) to give the title compound (8.5 mg).
Molecular formula C31H36N8O2Molecular weight 552.7LC-MS (M/e):523.0(M + H)+)
1H-NMR(400MHz,CDCl3)δ:10.20-10.05(m,1H),8.81(s,1H),7.95-8.15(m,1H),7.85-7.70(m,1H),7.59-7.61(m,2H),6.90-6.88(m,2H),5.70-5.50(m,2H),5.31(s,1H),5.05-4.95(m,1H),4.80-4.90(m,1H),4.72-4.62(m,2H),3.20-3.10(m,2H),3.05-2.95(m,4H),2.00(s,1H),1.48(s,6H),0.80-0.70(m,2H),0.55-0.40(m,4H),0.32-0.20(m,2H).
Experimental protocol
An exemplary experimental scheme of a portion of the compounds of the invention is provided below to show the advantageous activity and advantageous technical effects of the compounds of the invention. It should be understood, however, that the following experimental protocols are only illustrative of the present disclosure and are not intended to limit the scope of the present disclosure.
Experimental example 1 inhibition of cell proliferation Activity by Compounds of the present invention
Test article: the structural formula and the preparation method of the partial compound are shown in the preparation examples of the invention.
Positive control drug: AZD1775, prepared according to the method disclosed in prior art WO2007126128, and having the structure shown below:
Figure BDA0003391181930000261
experimental methods
1. Test materials and reagents
Name (R) Source Goods number
DMEM high-sugar medium Gibco 11995-065
MEM medium Gibco 10370-021
FBS (fetal bovine serum) Gibco 10099-141C
Horse serum Solarbio S9050
Pyruvic acid sodium salt Gibco 11360-070
Glutamine Gibco 35050-061
Penicillin streptomycin double antibody Gibco 1507-063
0.25% Trypsin Gibco 25200-072
10 XDPBS (Du's phosphate buffer) Gibco 14200-075
96-hole bottom transparent white board Corning 3610
Gemcitabine (Guitar decitabine) MCE HY-B0003
CTG Promega G7571
2. Cell culture media mix and plating number
Figure BDA0003391181930000262
3. Procedure of experiment
3.1 preparation of Compounds
Test compounds and reference positive control in 10mM DMSO were diluted in DMSO 3-fold gradient at 8 concentrations. Test compounds and reference positive control 10mM DMSO solutions were prepared in DMSO at 1000-fold concentrations for combination, i.e., 300. mu.M, 100. mu.M, 30. mu.M.
Gemcitabine (Gemcitabine)10mM stock solution was prepared and diluted 1000-fold to the highest final concentration, i.e., 100. mu.M, and then diluted 5 concentrations with a 3-fold gradient of DMSO.
3.2 test procedure
Culturing cells according to the recommended conditions of the source prescription, removing the culture medium, washing with PBS once, digesting with 0.25% trypsin, collecting cells after the complete culture medium is terminated, resuspending to a proper concentration, inoculating to a 96-well plate with 90 μ L/well, 37 deg.C, 5% CO2The incubator was incubated for 24 hours.
The compound is used alone: the final concentration of 1000 times compound solution, medium dilution 100 times, each well of 10 u L,37 degrees C, 5% CO2And (5) detecting after the incubator is cultured for 72 hours.
Combination of compound and gemcitabine: gemcitabine solution 1000 times final concentration, medium diluted 100 times, each well was added with 10. mu.L of 5% CO at 37 ℃2The incubator continues to incubate for 24 hours. The final concentration of the compound solution was 1000 times, the medium was diluted 20 times, and 5. mu.L of the diluted medium was added to each well at 37 ℃ and 5% CO2The incubator is used for detection after 24 hours of continuous culture.
The culture plate is placed at room temperature in advance, 60 mu L of CTG detection solution is added into each hole, the plate is protected from light, the plate is shaken on an orbital shaker for 2 minutes to crack cells, and after the reaction lasts for 20 minutes, the cold light value is read by a multifunctional microplate reader.
4. Data processing
Data were analyzed using GraphPad Prism 8.0 software, fitted to the data using non-linear sigmoidal regression to derive a dose-effect curve, and IC was calculated therefrom50The value is obtained.
Results of the experiment
TABLE 1 in vitro cell inhibitory Activity of Compounds of the invention alone
Figure BDA0003391181930000271
TABLE 2 in vitro cell inhibitory Activity of the Compounds of the invention in combination with Gemcitabine
Figure BDA0003391181930000272
Figure BDA0003391181930000281
Conclusion of the experiment
The compound disclosed by the invention has a good inhibition effect on the cell proliferation activity of A431, SK-MES-1 when being used alone, and has a remarkable synergistic effect on the cell proliferation activity inhibition of MIAPaCa2 when being used together with gemcitabine.
Experimental example 2 Metabolic stability of liver microsomes in various species of the Compound of the present invention
And (3) testing the sample: the chemical name and the preparation method of the compound 3 are shown in the preparation examples of the compound.
Positive control drug: AZD1775, prepared according to methods disclosed in the prior art.
Experimental materials:
cyno monkey mixed liver microsomes were purchased from the reid liver disease research center (shanghai ltd) under the batch number: SHZS, liver microsome protein concentration of 20mg mL-1.
CD-1 mouse, SD rat, Beagle dog, and human mixed liver microsomes were purchased from corning, under the respective lot numbers 9025002 (mouse), 0112002(SD rat), 9259006(Beagle dog), and 38295 (human), and the concentrations of liver microsomes were 20 mg/mL-1.
The experimental initiation factor beta-NADPH is purchased from Solarbio company; phosphate Buffered Saline (PBS) pH 7.4 was self-prepared by the laboratory.
Preparing a test solution:
a proper amount of test powder is precisely weighed, a proper amount of dimethyl sulfoxide (DMSO) is added to dissolve the test powder to 1mM, and the test powder is diluted by 20 times to 50 mu M of working solution by using methanol.
The experimental method comprises the following steps:
TABLE 3 liver microsome metabolic stability experiment incubation system composition
Figure BDA0003391181930000282
The experimental operation steps are as follows:
(1) according to the ratio of "Experimental incubation System constitution" in Table 3 above, 5.85mL of 100mM PBS and 20mM MgCl were used for each compound2Solution 0.585mL and H2O3.57 mL, and a mixed solution 1 (not containing microsomes, a sample and. beta. -NADPH) for incubation was prepared. The positive pair drug verapamil of the experimental incubation system was also performed to demonstrate normal liver microsomal enzyme activity.
(2) Liver microsomes (20mg protein/mL) were removed from the-80 ℃ freezer and placed on a 37 ℃ water bath constant temperature shaker for pre-incubation for 3 min.
(3) For each compound, 1.9mL of mixed solution 1 of incubation system was taken for each species, and 56. mu.L of microsomes of different species was added to prepare mixed solution 2 of incubation system (containing no test substance and. beta. -NADPH).
(4) Sample set (microsome and β -NADPH containing): and adding 14 mu L of the test sample working solution with the concentration of 50 mu M into 616 mu L of the mixed solution 2 of the incubation system, and adding 70 mu L of 10mM beta-NADPH working solution. Mixing, and repeating the steps. The sampling time points are 0min, 5min, 10min, 20min, 30min and 60 min. This sample set was used to evaluate the metabolic stability of compounds mediated via β -NADPH.
(5) Control group (microsome-containing, no β -NADPH, water instead of β -NADPH): 264 mu L of the mixed solution 2 of the incubation system is taken, 6 mu L of the working solution of the test article with the concentration of 50 mu M is added, and 30 mu L of water is added. Mixing, and repeating the steps. Sampling time points were 0min and 60 min. This negative control group was used to evaluate whether compounds present non- β -NADPH mediated metabolism in the liver microsome incubation system.
(6) At each predetermined time point, 50. mu.L of the incubation sample was sampled from the incubation sample tube, added to a stop sample tube (containing 300. mu.L of cold stop reagent, containing 50ng/mL acetonitrile of the internal standard tolbutamide), vortexed, and the reaction was stopped.
(7) After vortexing for 10min, centrifuge for 5min (12000 rpm).
(8) Taking 100 mu L of supernatant, adding 100 mu L of water, mixing uniformly by vortex, and carrying out LC-MS/MS sample injection analysis.
And (3) data analysis:
the percent residual was converted by the ratio of the peak area of the test article to the internal standard in the following equation.
Figure BDA0003391181930000291
The experimental results are as follows:
TABLE 4 hepatic microsome stability results for the compounds of the invention
Figure BDA0003391181930000292
The experimental conclusion is that:
the compound has good stability in human, monkey, dog, rat and mouse liver microsomes, and the stability in the mouse liver microsomes is equivalent to that of a control; in the other species of liver microsomes, the stability of the compounds of the invention was significantly better than that of the control AZD 1775.
Experimental example 3 inhibition of in vitro kinase Activity by Compounds of the present invention
Test article: the structural formula and the preparation method of the partial compound are shown in the preparation examples of the invention. The abbreviations used in the following experiments have the following meanings:
DMSO, DMSO: dimethyl sulfoxide; HEPES (high efficiency particulate air): hydroxyethyl piperazine ethanethiosulfonic acid.
The experimental method comprises the following steps: compounds were evaluated on WEE1 Kinase in vitro using the ADP-Glo Kinase Assay.
The experimental steps are as follows:
1. compound dilution:
test compounds (10mM stock) were diluted 100-fold with 100% DMSO in 384 dilution plates (labcyte, PP-0200) at 1: 3, carrying out equal ratio dilution on the concentration of the compound to be tested: 100. mu.M, 33.33. mu.M, 11.11. mu.M, 3.7. mu.M, 1.23. mu.M, 0.41. mu.M, 0.137. mu.M, 0.046. mu.M, 0.015. mu.M, 0.005. mu.M, 0. mu.M.
2. Transfer 0.1. mu.L of test compound to 384 reaction plates (PE,6007290) with Echo, 1000rpm/min, centrifuge for 1 min.
3. Transfer 5. mu.L of kinase to 384 reaction plates, 1000rpm/min, centrifuge for 1min, and incubate for 15min at 25 ℃.
4. Transfer 5. mu.L of substrate mixture to 384 reaction plates, 1000rpm/min, centrifuge for 1min, and incubate at 25 ℃ for 60 min. In the reaction, the final concentration of the test compound was 1000nM, 333.33nM, 111.11nM, 37.04nM, 12.35nM, 4.12nM, 1.37nM, 0.46nM, 0.15nM, 0.05nM, 0 nM. The final concentration of DMSO was 1%.
5. Transfer 5. mu.L ADP-Glo to 384 reaction plates at 1000rpm/min, centrifuge for 1min, and incubate at 25 ℃ for 60 min.
6. Transfer 10. mu.L of Detection solution to 384 reaction plates at 1000rpm/min, centrifuge for 1min, and incubate for 60min at 25 ℃.
7. The RLU (relative luminescence unit) signal is read using an Envision multifunction board reader.
8. Data computation
1) The mean data and Standard Deviation (SD) of DMSO and the positive control compound AZD-1775 (100% inhibition at 1000 nM) were calculated for each sieve plate as high and low controls.
2) Percent inhibition (% inh) 100 × (max-compound signal)/(max-min)
"minimum" is the negative control well reading without compound added; "Max" is the reading of the positive control well plus positive control compound AZD-1775 (100% inhibition at 1000 nM).
3) Fitting Compound IC Using non-Linear regression equation50
The experimental results are as follows:
TABLE 5 in vitro enzymatic inhibitory Activity of the Compounds of the invention
Figure BDA0003391181930000301
And (4) experimental conclusion:
the compound of the invention has good inhibition activity on WEE 1.

Claims (10)

1. A compound shown as a formula (I), pharmaceutically acceptable salt thereof or stereoisomer thereof,
Figure FDA0003391181920000011
wherein the content of the first and second substances,
X1、X2、X3are each independently selected from-C (R)2) -or-N-;
X4、X5each independently selected from-CH-or-N-;
each L is independently selected from-C (R)3)(R4)-、-O-、-N(R5) -or-S-;
ring A is selected from a 3-10 membered cycloalkyl, 3-10 membered heterocyclyl, 6-10 membered aryl or 5-10 membered heteroaryl optionally substituted with 1-4 of Q1;
ring B is selected from 3-10 membered cycloalkyl, 3-10 membered heterocyclyl, 6-10 membered aryl or 5-10 membered heteroaryl optionally substituted with 1-4Q 2;
ring C is selected from 5-15 membered bridged ring group, 5-10 membered spiro ring group, 5-15 membered bridged ring group or 5-10 membered spiroheterocyclic group optionally substituted with 1-4 of Q3, wherein ring carbon atoms in said 5-15 membered bridged ring group, 5-10 membered spiro ring group, 5-15 membered bridged ring group or 5-10 membered spiroheterocyclic group are optionally substituted with oxygen to form a carbonyl group;
each Q1 is uniqueSelected from halogen, hydroxy, amino, nitro, cyano, carboxy or the following optionally substituted with 1-3 substituents: c1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, each of which substituents is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, - (CH)2)m-3-10 membered cycloalkyl, - (CH)2)m-3-10 membered heterocycloalkyl, - (CH)2)m-5-10 membered heteroaryl or- (CH)2)m-6-10 membered aryl;
each Q2, each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, halo C1-6Alkoxy, halo C1-6Alkylthio, hydroxy C1-6Alkoxy, hydroxy C1-6Alkylthio, amino C1-6Alkoxy, amino C1-6Alkylthio, - (CH)2)m-3-10 membered cycloalkyl, - (CH)2)m-3-10 membered heterocycloalkyl, - (CH)2)m-5-10 membered heteroaryl or- (CH)2)m-6-10 membered aryl;
R1selected from C optionally substituted by substituents1-6Alkyl radical, C2-6Alkenyl or C2-6Alkynyl, the substituent groups are respectively and independently selected from halogen, hydroxyl, amino, carboxyl, cyano, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl, halo C1-6Alkoxy or C1-6An alkylcarbonyl group;
R2、R3、R4、R5each independently selected from hydrogen, halogen, hydroxyl, amino, carboxyl and C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl, halo C1-6Alkoxy or C1-6An alkylcarbonyl group;
m and n are respectively and independently selected from 0, 1,2 or 3.
2. The compound of claim 1, a pharmaceutically acceptable salt thereof, or a stereoisomer thereof,
ring a is selected from phenyl or a 5-6 membered nitrogen containing monoheteroaryl optionally substituted with 1-3Q 1;
each Q1 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy or the following optionally substituted with 1-3 substituents: c1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylthio, each of which substituents is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, - (CH)2)m-3-10 membered cycloalkyl, - (CH)2)m-3-10 membered heterocycloalkyl, - (CH)2)m-5-10 membered heteroaryl or- (CH)2)m-6-10 membered aryl;
ring B is selected from phenyl or 5-6 membered mono heteroaryl optionally substituted with 1 or 2Q 2; each Q2 is independently selected from halogen, hydroxy, amino, nitro, cyano, C1-6Alkyl radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy, hydroxy C1-6Alkoxy or amino C1-6An alkoxy group.
3. The compound of any one of claims 1-2, a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, wherein:
ring C is selected from the following optionally substituted with 1-2Q 3: 2-azabicyclo [2.2.1] heptanyl, 7-azabicyclo [2.2.1] heptanyl, 3-azabicyclo [3.2.1] octanyl, 8-azabicyclo [3.2.1] octanyl, 2-azabicyclo [2.2.2] octanyl, 2, 5-diazabicyclo [2.2.1] heptanyl, 3, 6-diazabicyclo [3.1.1] heptanyl, 3, 8-diazabicyclo [3.2.1] octanyl, 2-oxa-5-azabicyclo [2.2.1] heptanyl, 8-oxa-3-azabicyclo [3.2.1] octanyl, 3, 8-diazabicyclo [3.2.1] octan-6-enyl, 3, 9-diazabicyclo [3.3.1] nonanyl, 5-azabicyclo [4.1 ] heptanyl, 2-azaspiro [3.3] heptylalkyl, 2-azaspiro [3.5] nonanyl, 2, 6-diazaspiro [3.3] heptanyl, 2, 7-diazaspiro [3.5] nonanyl, 2, 5-diazaspiro [3.4] octanyl, 2-oxa-6-azaspiro [3.3] heptanyl, 6-oxa-2-azaspiro [3.4] octanyl, 6-azaspiro [3.4] octanyl, 2-azaspiro [4.4] nonanyl, 2-oxa-7-azaspiro [4.4] nonanyl, 6-azaspiro [3.4] oct-7-enyl, 2-oxa-6-azaspiro [3.4] oct-7-enyl or 2-azaspiro [4.4] non-7-enyl, wherein the ring-forming carbon atoms of the group are optionally substituted with oxygen to form a carbonyl group;
each Q3 is independently selected from halogen, hydroxy, amino, nitro, cyano, carboxy, C1-6Alkyl, halo C1-6Alkyl, hydroxy C1-6Alkyl radical, C1-6Alkoxy, halo C1-6Alkoxy or hydroxy C1-6An alkoxy group.
4. The compound, pharmaceutically acceptable salt thereof, or stereoisomer thereof according to any one of claims 1-3,
X1、X2、X3are each independently selected from-C (R)2) -or-N-;
X4、X5each independently selected from-CH-or-N-;
each L is independently selected from-C (R)3)(R4)-、-O-、-N(R5) -or-S-;
R2、R3、R4、R5each independently selected from hydrogen, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, monofluoromethyl, difluoromethyl, trifluoromethyl or trifluoromethoxy;
R1selected from C optionally substituted by substituents2-6Alkenyl, the substituents are each independentlyIs selected from halogen, hydroxy, amino, carboxyl, cyano, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkylamino radical, di (C)1-6Alkyl) amino, halo C1-6Alkyl, hydroxy C1-6Alkyl, amino C1-6Alkyl or halo C1-6An alkoxy group.
5. The compound of any one of claims 1-4, a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, having a structure represented by the following general formula (II):
Figure FDA0003391181920000031
wherein, ring A, ring B, ring C, Q1, Q2, Q3, L, R3、R4、R5、R1M, n are as defined in any one of claims 1 to 4.
6. The compound, pharmaceutically acceptable salt thereof, or stereoisomer thereof according to any one of claims 1-5,
l is NH;
ring a is selected from pyridyl, pyrimidinyl, pyrazinyl, or pyridazinyl substituted with 1Q 1; q1 is selected from fluoro, chloro, bromo, hydroxy, amino, nitro, cyano or the following optionally substituted with 1-3 substituents: methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, methoxy, ethoxy, propoxy and isopropoxy, wherein the substituents are independently selected from fluoro, chloro, bromo, hydroxy, amino, nitro, cyano, carboxy, - (CH)2)m-cyclopropane, - (CH)2)m-cyclobutyl, - (CH)2)m-cyclopentyl or- (CH)2)m-a cyclohexane group;
ring B is selected from phenyl, pyridyl, pyrimidinyl, pyrazinyl, or pyridazinyl;
m is selected from 0, 1 or 2;
n is 1.
7. The compound of claim 1, a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, selected from the following compounds:
Figure FDA0003391181920000032
Figure FDA0003391181920000041
8. a pharmaceutical formulation comprising a compound according to any one of claims 1 to 7, a pharmaceutically acceptable salt thereof or a stereoisomer thereof, in a pharmaceutically acceptable dosage form, comprising one or more pharmaceutically acceptable excipients.
9. A pharmaceutical composition comprising a compound, pharmaceutically acceptable salt thereof, or stereoisomer thereof according to any one of claims 1-7, comprising one or more second therapeutically active agent selected from mitotic inhibitors, alkylating agents, antimetabolites, antisense DNA or RNA, antitumor antibiotics, growth factor inhibitors, signaling inhibitors, cell cycle inhibitors, enzyme inhibitors, retinoid receptor modulators, proteasome inhibitors, topoisomerase inhibitors, biological response modifiers, hormonal drugs, angiogenesis inhibitors, cell growth inhibitors, targeting antibodies, HMG-CoA reductase inhibitors, and prenyl protein transferase inhibitors.
10. Use of a compound of any one of claims 1-7, a pharmaceutically acceptable salt or stereoisomer thereof, a pharmaceutical preparation of claim 8, or a pharmaceutical composition of claim 9 for the manufacture of a medicament for the treatment and/or prevention of a disease and related disorder mediated by Wee1 selected from cancer or benign tumor selected from lung cancer, squamous cell carcinoma, bladder cancer, gastric cancer, ovarian cancer, peritoneal cancer, pancreatic cancer, breast cancer, head and neck cancer, cervical cancer, endometrial cancer, rectal cancer, liver cancer, kidney cancer, esophageal adenocarcinoma, esophageal squamous cell carcinoma, prostate cancer, thyroid cancer, female genital tract cancer, lymphoma, neurofibroma, bone cancer, skin cancer, brain cancer, colon cancer, testicular cancer, small cell lung cancer, non-small cell lung cancer, cancer of the stomach, cancer, squamous cell carcinoma of the esophagus, cancer of the prostate, thyroid cancer, cancer of the female genital tract, lymphoma, neurofibroma, bone cancer, skin cancer, brain cancer, colon cancer, testicular cancer, small cell lung cancer, non-cell lung cancer, cancer of the stomach cancer, cancer of the subject for example, cancer of the subject for example, cancer, Gastrointestinal stromal tumors, mast cell tumors, multiple myeloma, melanoma, leukemia, glioma, or sarcoma.
CN202111465345.0A 2020-12-04 2021-12-03 Dihydropyrazolopyrimidinone derivatives Active CN114591334B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202011407293 2020-12-04
CN2020114072937 2020-12-04

Publications (2)

Publication Number Publication Date
CN114591334A true CN114591334A (en) 2022-06-07
CN114591334B CN114591334B (en) 2023-10-20

Family

ID=81813674

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111465345.0A Active CN114591334B (en) 2020-12-04 2021-12-03 Dihydropyrazolopyrimidinone derivatives

Country Status (1)

Country Link
CN (1) CN114591334B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023179196A1 (en) * 2022-03-25 2023-09-28 药雅科技(上海)有限公司 Preparation and application of wee1 kinase inhibitor

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009054332A1 (en) * 2007-10-23 2009-04-30 Banyu Pharmaceutical Co., Ltd. Pyridone-substituted-dihydropyrazolopyrimidinone derivative
CN109422754A (en) * 2017-08-24 2019-03-05 上海迪诺医药科技有限公司 Pyrazolo [3,4-d] pyrimidine -3- ketone derivatives, its pharmaceutical composition and application
WO2019165204A1 (en) * 2018-02-23 2019-08-29 Newave Pharmaceutical Inc. 1,2-dihydro-3h-pyrazolo[3,4-d]pyrimidine-3-one compounds as inhibitors of wee-1 kinase
CN110198943A (en) * 2017-01-23 2019-09-03 石家庄智康弘仁新药开发有限公司 1,2- dihydro -3H- pyrazoles [3,4-d] pyrimidine -3- ketone derivatives as Wee1 inhibitor
CN110872296A (en) * 2018-08-31 2020-03-10 上海弘翊生物科技有限公司 isoindoline-1H-pyrazolo [3,4-d ] pyrimidone compound, and preparation method and application thereof
CN111315747A (en) * 2018-01-05 2020-06-19 四川科伦博泰生物医药股份有限公司 Dihydropyrazolone pyrimidine compound and preparation method and application thereof
CN112142748A (en) * 2019-06-28 2020-12-29 上海医药集团股份有限公司 Pyrazolone pyrimidine compound, preparation method and application thereof
CN112142747A (en) * 2019-06-28 2020-12-29 上海医药集团股份有限公司 Pyrazolone pyrimidine compound, preparation method and application thereof
CN113387962A (en) * 2020-03-12 2021-09-14 上海迪诺医药科技有限公司 Pyrazolo [3,4-d ] pyrimidine-3-one derivative, pharmaceutical composition and application thereof
CN115698006A (en) * 2020-06-17 2023-02-03 微境生物医药科技(上海)有限公司 Pyrazolo [3,4-d ] pyrimidin-3-one derivatives as Wee-1 inhibitors

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009054332A1 (en) * 2007-10-23 2009-04-30 Banyu Pharmaceutical Co., Ltd. Pyridone-substituted-dihydropyrazolopyrimidinone derivative
US20100221211A1 (en) * 2007-10-23 2010-09-02 Hidetomo Furuyama Pyridone-substituted-dihydropyrazolopyrimidinone derivative
CN110198943A (en) * 2017-01-23 2019-09-03 石家庄智康弘仁新药开发有限公司 1,2- dihydro -3H- pyrazoles [3,4-d] pyrimidine -3- ketone derivatives as Wee1 inhibitor
CN109422754A (en) * 2017-08-24 2019-03-05 上海迪诺医药科技有限公司 Pyrazolo [3,4-d] pyrimidine -3- ketone derivatives, its pharmaceutical composition and application
CN111315747A (en) * 2018-01-05 2020-06-19 四川科伦博泰生物医药股份有限公司 Dihydropyrazolone pyrimidine compound and preparation method and application thereof
WO2019165204A1 (en) * 2018-02-23 2019-08-29 Newave Pharmaceutical Inc. 1,2-dihydro-3h-pyrazolo[3,4-d]pyrimidine-3-one compounds as inhibitors of wee-1 kinase
CN110872296A (en) * 2018-08-31 2020-03-10 上海弘翊生物科技有限公司 isoindoline-1H-pyrazolo [3,4-d ] pyrimidone compound, and preparation method and application thereof
CN112142748A (en) * 2019-06-28 2020-12-29 上海医药集团股份有限公司 Pyrazolone pyrimidine compound, preparation method and application thereof
CN112142747A (en) * 2019-06-28 2020-12-29 上海医药集团股份有限公司 Pyrazolone pyrimidine compound, preparation method and application thereof
CN113387962A (en) * 2020-03-12 2021-09-14 上海迪诺医药科技有限公司 Pyrazolo [3,4-d ] pyrimidine-3-one derivative, pharmaceutical composition and application thereof
CN115698006A (en) * 2020-06-17 2023-02-03 微境生物医药科技(上海)有限公司 Pyrazolo [3,4-d ] pyrimidin-3-one derivatives as Wee-1 inhibitors

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023179196A1 (en) * 2022-03-25 2023-09-28 药雅科技(上海)有限公司 Preparation and application of wee1 kinase inhibitor

Also Published As

Publication number Publication date
CN114591334B (en) 2023-10-20

Similar Documents

Publication Publication Date Title
JP6726677B2 (en) Substituted 2-H-pyrazole derivatives as anticancer agents
TWI751163B (en) Fgfr4 inhibitor, its preparation method and use thereof
JP5752232B2 (en) Substituted pyrrolotriazine compounds as protein kinase inhibitors
CN110382499B (en) FGFR inhibitor and application thereof
TWI749126B (en) Cdk4/6 inhibitors
JP6667014B2 (en) Pyrimidine derivatives, their preparation and use in medicine
AU2012310168B2 (en) 6 - substituted 3 - (quinolin- 6 - ylthio) - [1,2,4] triazolo [4, 3 -a] pyradines as tyrosine kinase
CN111171049B (en) Tyrosine kinase inhibitors and uses thereof
EP3596073A1 (en) Tricyclic compounds for use in treatment of proliferative disorders
KR20050111781A (en) Substituted 8-perfluoroalkyl-6,7,8,9-tetrahydropyrimido[1,2-a]pyrimidin-4-one derivatives
TW202237585A (en) Cdk inhibitors
CA3092770A1 (en) Substituted imidazolidin-2-one derivatives as prmt5 inhibitors
CN116888108A (en) Novel EGFR degradation agent
CN113968856B (en) Compounds with kinase inhibitory activity
CN114591334B (en) Dihydropyrazolopyrimidinone derivatives
JP6977038B2 (en) GSK-3 inhibitor
CN115707708A (en) Wee1 kinase inhibitors
CA3216045A1 (en) Compounds as pd1/pd-l1 inhibitors and methods thereof
TWI749518B (en) Piperazine amide derivative, its preparation method and its use in medicine
CN115197221A (en) Dihydropyrazolopyrimidinone macrocyclic derivatives and application thereof
TW202214634A (en) Heterocyclic compound and derivative thereof
TW202222797A (en) Pyrazolo[1,5-a]pyridine compound, preparation method therefor and use thereof
KR20210108433A (en) Thienopyridinone compound
CN115843296B (en) CDK9 inhibitors and uses thereof
TW202310845A (en) Heteroaryl derivative compounds, and uses 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