WO2023005281A1 - Preparation method for and application of novel cdk9 inhibitor having macrocyclic skeleton structure - Google Patents

Preparation method for and application of novel cdk9 inhibitor having macrocyclic skeleton structure Download PDF

Info

Publication number
WO2023005281A1
WO2023005281A1 PCT/CN2022/088343 CN2022088343W WO2023005281A1 WO 2023005281 A1 WO2023005281 A1 WO 2023005281A1 CN 2022088343 W CN2022088343 W CN 2022088343W WO 2023005281 A1 WO2023005281 A1 WO 2023005281A1
Authority
WO
WIPO (PCT)
Prior art keywords
acid
compound
pharmaceutically acceptable
acceptable salt
reaction
Prior art date
Application number
PCT/CN2022/088343
Other languages
French (fr)
Chinese (zh)
Inventor
卞金磊
李志裕
吴体智
喻彬
张立翱
杜泽坤
Original Assignee
中国药科大学
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 中国药科大学 filed Critical 中国药科大学
Publication of WO2023005281A1 publication Critical patent/WO2023005281A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/08Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • the invention relates to the field of medicinal chemistry, in particular to a macrocyclic CDK9 inhibitor and its preparation method and application.
  • CDKs Cyclin-dependent kinases
  • CDKs are key protein kinases in cell cycle regulation, which can effectively regulate DNA synthesis and mitosis. Combining with the corresponding chaperones to form a complex is a necessary condition for CDK protein function.
  • CDKs can be divided into periodic CDKs (including CDK1, CDK2, CDK3, CDK4, and CDK6) and transcriptional CDKs (including CDK7, CDK8, CDK9, CDK11, CDK12, CDK13, CDK19, etc.).
  • CDK9 protein is widely involved in the initiation, elongation and termination stages of transcription.
  • CDK9 can effectively promote RNA transcription elongation by phosphorylating the CTD end of RNA polymerase II, so CDK9 protein is the key to regulating RNA transcription Factors, and can effectively regulate the protein level of downstream proteins, including the anti-apoptotic protein MCL-1.
  • a number of studies have shown that CDK9 protein imbalance occurs in the occurrence and development of a variety of tumors, including leukemia and other blood tumors, prostate cancer, lung cancer and other solid tumors.
  • an article published in the journal Cell pointed out that the inhibition of CDK9 protein can activate genes that are suppressed by tumors, and promote the expression of tumor suppressor genes and cell differentiation.
  • the above studies show that finding small molecule drugs that target and inhibit CDK9 may be an effective strategy for the development of anti-tumor drugs.
  • CDK9 inhibitors are in the clinical research stage, but most of them are non-selective, which leads to many unpredictable toxic and side effects. Even the clinical trials of some drugs were terminated because of this. Due to the high homology of CDK family members, it is difficult to obtain highly selective CDK9 inhibitors. However, in view of the current clinical research situation and the need to further study the biological role of CDK9, the development of selective CDK9 inhibitors is very important. Currently, there are only three selective CDK inhibitors reported in Linchuan research, including Bayer's BAY-1143572 and BAY-1251152 and AstraZeneca's AZD4573. Clinical trials of BAY-1143572 have been terminated due to neutropenia, but the cause of this side effect has not been reported yet.
  • the main purpose of the present invention is to find a novel CDK9 small molecule inhibitor with high activity, high selectivity and good druggability.
  • Ar is selected from the following groups:
  • X is selected from hydrogen, deuterium, halogen, cyano, methyl, trifluoromethyl;
  • R 1 is selected from the following groups:
  • L is selected from the following structures:
  • the pharmaceutically acceptable salt form in the present invention refers to the salt formed by the compound represented by the general formula I and a pharmaceutically acceptable acid, including inorganic acid salts and organic acid salts.
  • Inorganic acid salts include: hydrochloric acid, sulfuric acid, phosphoric acid, carbonic acid, bicarbonate, nitric acid, monohydrogen phosphate, dihydrogen phosphate, hydrobromic acid or hydroiodic acid;
  • organic acids include: maleic acid, tartaric acid, citric acid , Methanesulfonic acid, succinic acid, acetic acid, p-toluenesulfonic acid, mandelic acid, isobutyric acid, malonic acid, etc.
  • Ar is selected from
  • X is halogen, such as F, Cl, Br, I; in a more specific example, X is F.
  • L is selected from:
  • the present invention provides any of the following compounds or pharmaceutically acceptable salts thereof:
  • the present invention also discloses a synthetic method, and most of the compounds described in the present invention can be obtained rapidly through the following synthetic methods:
  • Ar, X, R 1 and L are as defined above.
  • the present invention also provides a pharmaceutical composition, comprising the compound of the present invention, or a pharmaceutically acceptable salt or prodrug thereof.
  • the present invention also provides the application of the compound of the present invention or its pharmaceutically acceptable salt form in the preparation of CDK9 inhibitor drugs.
  • the inventors found that the compound of the present invention or its pharmaceutically acceptable salt form can effectively inhibit CDK9 protein Activity or application in the discovery of small molecule drugs targeting CDK9.
  • the present invention also provides the application of the compound of the present invention or a pharmaceutically acceptable salt form thereof in the preparation of antiviral drugs or antitumor drugs.
  • the viruses include: HIV virus, cytomegalovirus, Epstein-Barr virus, adenovirus, herpes, and human T-cell lymphoblastic virus.
  • the tumors include glioma, various types of leukemia, lymphoma, liver cancer, gastric cancer, prostate cancer, ovarian cancer, breast cancer, and lung cancer.
  • the compound described in the present invention is a CDK9 inhibitor with a new skeleton.
  • the inventors designed and synthesized a series of derivatives through a rational drug design method. Inhibitory activity and selectivity of CDK9.
  • Biological activity evaluation shows that the designed compound has significant CDK9 inhibitory activity, has good selectivity (more than 50 times) to CDK family members, and has good anti-proliferation effect on MV4-11, MCF7, HCT116, MOLM13 and other tumor cells active.
  • Embodiment 2 Compound T-2
  • Embodiment 3 Compound T-3
  • Embodiment 4 Compound T-4
  • Embodiment 5 Compound T-5
  • intermediate 12 takes intermediate 2 and methyl 2-methoxy-4-aminobenzoate (intermediate 11) as reaction raw materials, and the synthesis method is the same as that of intermediate 10, with a yield of 73%.
  • HRMS[ESI] + calcd for C 20 H 17 F 2 N 3 O 4 [M+H] + ,402.1187,found 402.1180.
  • Embodiment 6 Compound T-6
  • Embodiment 7 Compound T-7
  • Embodiment 8 Compound T-8
  • Embodiment 9 Compound T-9
  • the synthesis of intermediates 20, 21 and 22 can refer to the preparation method in the synthetic route of compound T1.
  • the synthesis of compound T-27 can refer to the synthesis method of compound T-26 described in Example 26.
  • the preparation of the compounds described in Examples 30 and 31 can refer to the synthesis method and route of compound T-5, and the starting material 4-fluoro-3 methoxyphenylboronic acid in this route is replaced by 4-fluoro-2 methoxy Phenylboronic acid, the linker chain and the amino side chain in the last step of the reaction can be replaced with specific groups.
  • the preparation of the compounds described in Examples 32-35 can refer to the synthesis method and route of compound T-5, and the starting material 4-fluoro-3 methoxyphenylboronic acid in this route is replaced by 5-methoxypyridine-3 -Boronic acid, the linker chain and the amino side chain in the last step of the reaction can be replaced with specific groups.
  • the preparation of the compound described in Example 36 can refer to the synthesis method and route of compound T-18, and the starting material 4-fluoro-3 methoxyphenylboronic acid in this route is replaced by 5-methoxypyridine-3-boronic acid , the linker connecting chain and the amino side chain in the penultimate step of the reaction can be replaced with specific groups.
  • the preparation of the compound described in Example 37 can refer to the synthesis method and route of compound T-18, and the starting material 4-fluoro-3 methoxyphenylboronic acid in this route is replaced by 5-methoxypyridine-3-boronic acid , the linker connecting chain and the amino side chain in the penultimate step of the reaction can be replaced with specific groups.
  • the synthesis of intermediate 43 can refer to the synthesis method of intermediate 18.
  • the synthesis of intermediate 46 can refer to the synthesis method of intermediate 19.
  • the preparation of the compounds described in Examples 39-40 can refer to the preparation method of compound T-5, wherein, 4-fluoro-3-methoxyphenylboronic acid is replaced by the corresponding boric acid or boric acid ester, and the intermediate 2 Synthetic Methods The required starting materials were synthesized.
  • the preparation of the compounds described in Examples 41-44 can refer to the preparation method of compound T-5, wherein the corresponding dichloropyrimidine derivatives can be used to synthesize the required starting materials according to the synthesis method of intermediate 2.
  • reaction solution was raised to room temperature, it was poured into an appropriate amount of ammonium chloride aqueous solution, extracted three times with dichloromethane, and the organic phases were combined and washed twice with saturated aqueous sodium chloride solution. Dry over sodium sulfate, filter with suction, concentrate, and finally use column chromatography to separate compound T-45 with a yield of 36%.
  • Example 46 The preparation of the compound described in Example 46 can refer to the synthesis method and route of compound T-45 in Example 45.
  • Example 47 The preparation of the compound described in Example 47 can refer to the synthesis method and route of compound T-5 in Example 5.
  • Example 50 For the preparation method of Example 50, refer to the synthesis method and route of compound T-25 described in Example 25.
  • the preparation of the compounds described in Examples 51-54 can refer to the synthesis method and route of compound T-5.
  • the experimental method is as follows:
  • the cytotoxicity of the compound to tumor cells with high expression of CDK9 was determined by MTT method: the cells were seeded in 96-well plate at 2000 cells/well, after the cells adhered to the wall, the culture medium was aspirated and added 200uL of the diluted drug was incubated at 37°C with 5% CO2 for 72h, and then 10uL/well of MTT was added. Incubate at 37°C for 4 hours, remove the supernatant, add 150uL of DMSO to each well, and detect the absorbance at 492nm with a multi-functional microplate reader. IC50 was calculated with GraphPad and the cell growth curve was plotted.
  • the experimental method is as follows:
  • the compound was prepared into a 10 mM stock solution with 100% DMSO and stored at low temperature in the dark.
  • the kinase reaction process is as follows:
  • test compound test concentration is 5nM, 20nM or 100nM, and repeated well detection.
  • Compounds were prepared at 100-fold final concentration in a 384-well plate. Then use Echo550 to transfer 250nl to 384 reaction plate for later use. Add 250 nl of 100% DMSO to negative control wells and positive control wells respectively.
  • Conversion%_sample is the conversion rate reading of the sample
  • Conversion%_min the average value of negative control wells, representing the conversion rate readings of wells without enzyme activity
  • Conversion%_max the average value of positive control wells, representing the conversion rate readings of wells without compound inhibition.
  • the concentration of the compound is indicated in the brackets, and the unit is nM, such as 90 (20) indicates that the inhibition rate of the compound to the enzyme is 90% at a concentration of 20 nM.
  • TG-02 is as follows:
  • the compounds of the present invention exhibit effective inhibitory activity on CDK9, and most of the compounds have an inhibitory rate of more than 90% on CDK9 at a concentration of 20 nM.
  • the compound of the present invention shows remarkable CDK9 selectivity.
  • it has significant anti-proliferation inhibitory activity against various tumor cells such as MV4-11, MCF-7, HCT-116 and MOLM13.
  • the dominant compounds T-9, T-10, T-52 and T-53 are all highly selective CDK9 inhibitors, with a selectivity of more than 50 times for CDK2, and they all have good anti-tumor cell proliferation activity.
  • Compound T-53 has an inhibitory IC 50 value of 7 nM for CDK9, and an IC 50 value of greater than 1000 nM for CDK2, with a selectivity greater than 100 times, which is significantly better than that of compound TG02.

Abstract

Disclosed are a preparation method for and an application of a novel CDK9 inhibitor having a macrocyclic skeleton structure (I). The compound can effectively inhibit the activity of CDK9 protein and has high selectivity for CDK9. Also disclosed are a preparation method for the compound and an application thereof in preventing and/or treating tumor-related diseases, comprising glioma, various types of leukemia, lymphoma, liver cancer, gastric cancer, prostate cancer, ovarian cancer, breast cancer, lung cancer, etc.

Description

一种具有大环骨架结构的新型CDK9抑制剂的制备及其应用Preparation and application of a novel CDK9 inhibitor with macrocyclic skeleton structure 技术领域technical field
本发明涉及药物化学领域,具体为一种大环类CDK9抑制剂及其制备方法和应用。The invention relates to the field of medicinal chemistry, in particular to a macrocyclic CDK9 inhibitor and its preparation method and application.
背景技术Background technique
细胞周期依赖性蛋白激酶(Cyclin-dependent kinases,CDKs)是细胞周期调控的关键的蛋白激酶,可有效调节DNA合成和有丝分裂。与相应的伴侣蛋白结合形成复合物是发挥CDK蛋白功能的必要条件。根据其主要功能,可将CDK分为周期性CDK(包括CDK1,CDK2,CDK3,CDK4和CDK6)和转录性CDK(包括CDK7,CDK8,CDK9,CDK11,CDK12CDK13,CDK19等)。研究表明很多癌症存在“转录成瘾性”,故转录性CDK近来备受关注。其中,CDK9蛋白广泛参与了转录的起始、延伸和终止阶段,特别的是,CDK9可通过磷酸化RNA聚合酶II的CTD端进而有效地促使RNA转录延伸,故CDK9蛋白是调节RNA转录的关键因子,并可以有效调节下游蛋白,包括抗凋亡蛋白MCL-1在内的蛋白水平。多项研究表明,CDK9蛋白的失调出现在多种肿瘤的发生发展中,包括白血病等多种血液瘤,前列腺癌、肺癌等多种实体瘤。2018年,Cell期刊上发表文章指出,CDK9蛋白的抑制可激活被肿瘤抑制的基因,促进抑癌基因的表达和细胞分化。上述研究表明,寻找靶向抑制CDK9的小分子药物可能是一种有效的研发抗肿瘤药物的策略。Cyclin-dependent kinases (CDKs) are key protein kinases in cell cycle regulation, which can effectively regulate DNA synthesis and mitosis. Combining with the corresponding chaperones to form a complex is a necessary condition for CDK protein function. According to their main functions, CDKs can be divided into periodic CDKs (including CDK1, CDK2, CDK3, CDK4, and CDK6) and transcriptional CDKs (including CDK7, CDK8, CDK9, CDK11, CDK12, CDK13, CDK19, etc.). Studies have shown that many cancers have "transcriptional addiction", so transcriptional CDKs have recently attracted attention. Among them, CDK9 protein is widely involved in the initiation, elongation and termination stages of transcription. In particular, CDK9 can effectively promote RNA transcription elongation by phosphorylating the CTD end of RNA polymerase II, so CDK9 protein is the key to regulating RNA transcription Factors, and can effectively regulate the protein level of downstream proteins, including the anti-apoptotic protein MCL-1. A number of studies have shown that CDK9 protein imbalance occurs in the occurrence and development of a variety of tumors, including leukemia and other blood tumors, prostate cancer, lung cancer and other solid tumors. In 2018, an article published in the journal Cell pointed out that the inhibition of CDK9 protein can activate genes that are suppressed by tumors, and promote the expression of tumor suppressor genes and cell differentiation. The above studies show that finding small molecule drugs that target and inhibit CDK9 may be an effective strategy for the development of anti-tumor drugs.
目前已有十余个CDK9抑制剂处于临床研究阶段,但其中大部分为非选择性的,进而导致很多难以预料的毒副作用。甚至有部分药物的临床试验因此被终止。由于CDK家族成员的高度同源性,想获得高选择性的CDK9抑制剂有一定困难。但鉴于目前的临床研究情况和进一步深入研究CDK9生物学作用的需求,选择性CDK9抑制剂的开发至关重要。目前已报道的处于临川研究的选择性CDK抑制剂仅有三个,包括拜耳公司的BAY-1143572和BAY-1251152以及阿斯利康公司的AZD4573。由于嗜中性白血球减少症,BAY-1143572的临床试验已经被终止,但导致这一副作用的原因暂没有相关报道。At present, more than ten CDK9 inhibitors are in the clinical research stage, but most of them are non-selective, which leads to many unpredictable toxic and side effects. Even the clinical trials of some drugs were terminated because of this. Due to the high homology of CDK family members, it is difficult to obtain highly selective CDK9 inhibitors. However, in view of the current clinical research situation and the need to further study the biological role of CDK9, the development of selective CDK9 inhibitors is very important. Currently, there are only three selective CDK inhibitors reported in Linchuan research, including Bayer's BAY-1143572 and BAY-1251152 and AstraZeneca's AZD4573. Clinical trials of BAY-1143572 have been terminated due to neutropenia, but the cause of this side effect has not been reported yet.
发明内容Contents of the invention
本发明的主要目的是寻找具有高活性,高选择性,且兼具良好成药性质的新型CDK9小分子抑制剂。The main purpose of the present invention is to find a novel CDK9 small molecule inhibitor with high activity, high selectivity and good druggability.
一种如通式I所示结构的化合物,或其药学上可接受的盐型:A compound of the structure shown in general formula I, or a pharmaceutically acceptable salt thereof:
Figure PCTCN2022088343-appb-000001
Figure PCTCN2022088343-appb-000001
其中,Ar选自如下基团:Wherein, Ar is selected from the following groups:
Figure PCTCN2022088343-appb-000002
Figure PCTCN2022088343-appb-000002
X选自氢,氘,卤素,氰基,甲基,三氟甲基;X is selected from hydrogen, deuterium, halogen, cyano, methyl, trifluoromethyl;
R 1选自如下基团: R 1 is selected from the following groups:
Figure PCTCN2022088343-appb-000003
Figure PCTCN2022088343-appb-000003
L选自如下结构:L is selected from the following structures:
Figure PCTCN2022088343-appb-000004
Figure PCTCN2022088343-appb-000004
本发明所述的药学上可接受的盐型是指通式I所示的化合物和药学上可接受的酸形成的盐,包括无机酸盐和有机酸盐。其中无机酸盐包括:盐酸、硫酸、磷酸、碳酸、碳酸氢根、硝酸、磷酸一氢根、磷酸二氢根,氢溴酸或氢碘酸;有机酸包括:马来酸、酒石酸、柠檬酸、甲磺酸、琥珀酸、乙酸、对甲苯磺酸、扁桃酸、异丁酸、丙二酸等。The pharmaceutically acceptable salt form in the present invention refers to the salt formed by the compound represented by the general formula I and a pharmaceutically acceptable acid, including inorganic acid salts and organic acid salts. Inorganic acid salts include: hydrochloric acid, sulfuric acid, phosphoric acid, carbonic acid, bicarbonate, nitric acid, monohydrogen phosphate, dihydrogen phosphate, hydrobromic acid or hydroiodic acid; organic acids include: maleic acid, tartaric acid, citric acid , Methanesulfonic acid, succinic acid, acetic acid, p-toluenesulfonic acid, mandelic acid, isobutyric acid, malonic acid, etc.
在一些具体的实例中,Ar选自
Figure PCTCN2022088343-appb-000005
In some specific examples, Ar is selected from
Figure PCTCN2022088343-appb-000005
在一些具体的实例中,X为卤素,例如F、Cl、Br、I;在一种更为具体的实例中,X为F。In some specific examples, X is halogen, such as F, Cl, Br, I; in a more specific example, X is F.
在一些具体的实例中,L选自:In some specific examples, L is selected from:
Figure PCTCN2022088343-appb-000006
Figure PCTCN2022088343-appb-000006
在一些具体的实例中,本发明提供如下任一所述的化合物或其药学上可接受的盐:In some specific examples, the present invention provides any of the following compounds or pharmaceutically acceptable salts thereof:
Figure PCTCN2022088343-appb-000007
Figure PCTCN2022088343-appb-000007
Figure PCTCN2022088343-appb-000008
Figure PCTCN2022088343-appb-000008
Figure PCTCN2022088343-appb-000009
Figure PCTCN2022088343-appb-000009
Figure PCTCN2022088343-appb-000010
Figure PCTCN2022088343-appb-000010
Figure PCTCN2022088343-appb-000011
Figure PCTCN2022088343-appb-000011
Figure PCTCN2022088343-appb-000012
Figure PCTCN2022088343-appb-000012
Figure PCTCN2022088343-appb-000013
Figure PCTCN2022088343-appb-000013
Figure PCTCN2022088343-appb-000014
Figure PCTCN2022088343-appb-000014
Figure PCTCN2022088343-appb-000015
Figure PCTCN2022088343-appb-000015
本发明还公开一种合成方法,本发明所述的大部分化合物均可通过以下合成方法快速获得:The present invention also discloses a synthetic method, and most of the compounds described in the present invention can be obtained rapidly through the following synthetic methods:
Figure PCTCN2022088343-appb-000016
Figure PCTCN2022088343-appb-000016
其中,Ar、X、R 1、L定义如前所述。 Wherein, Ar, X, R 1 and L are as defined above.
本发明还提供一种药物组合物,包括本发明所述的化合物,或其药学上可接受的盐型或者前药。The present invention also provides a pharmaceutical composition, comprising the compound of the present invention, or a pharmaceutically acceptable salt or prodrug thereof.
本发明还提供本发明所述化合物或其药学上可接受的盐型在制备CDK9抑制剂药物中的应用,发明人发现本发明所述化合物或其药学上可接受的盐型可有效抑制CDK9蛋白活性或应用于以CDK9为靶点的小分子药物的发现。The present invention also provides the application of the compound of the present invention or its pharmaceutically acceptable salt form in the preparation of CDK9 inhibitor drugs. The inventors found that the compound of the present invention or its pharmaceutically acceptable salt form can effectively inhibit CDK9 protein Activity or application in the discovery of small molecule drugs targeting CDK9.
本发明还提供本发明所述化合物或其药学上可接受的盐型在制备抗病毒药物或者抗肿瘤药物中的应用。The present invention also provides the application of the compound of the present invention or a pharmaceutically acceptable salt form thereof in the preparation of antiviral drugs or antitumor drugs.
在本发明的一些实施例中,所述的病毒包括:HIV病毒、巨细胞病毒、EB病毒、腺病毒、疱疹、人T细胞淋巴细胞病毒。In some embodiments of the present invention, the viruses include: HIV virus, cytomegalovirus, Epstein-Barr virus, adenovirus, herpes, and human T-cell lymphoblastic virus.
本发明的一些实施例中,所述的肿瘤包括神经胶质瘤、各类白血病、淋巴癌、肝癌、胃癌、前列腺癌、卵巢癌、乳腺癌、肺癌。In some embodiments of the present invention, the tumors include glioma, various types of leukemia, lymphoma, liver cancer, gastric cancer, prostate cancer, ovarian cancer, breast cancer, and lung cancer.
本发明所述的化合物为新骨架的CDK9抑制剂,发明人通过合理的药物设计方法,设计合成了一系列衍生物,本发明所述的化合物通过大环固定优势化合物的构象,进一步提升其对CDK9的抑制活性和选择性。生物活性评价显示,所设计化合物具有显著的CDK9抑制活性,对CDK家族成员具有较好的选择性(大于50倍),同时对MV4-11,MCF7,HCT116,MOLM13等肿瘤细胞具有良好的抗增殖活性。The compound described in the present invention is a CDK9 inhibitor with a new skeleton. The inventors designed and synthesized a series of derivatives through a rational drug design method. Inhibitory activity and selectivity of CDK9. Biological activity evaluation shows that the designed compound has significant CDK9 inhibitory activity, has good selectivity (more than 50 times) to CDK family members, and has good anti-proliferation effect on MV4-11, MCF7, HCT116, MOLM13 and other tumor cells active.
具体实施方式Detailed ways
下面通过具体的实施例对本发明做进一步的详细说明,但并不将本发明限制在所举的实施例范围之中。The present invention will be further described in detail through specific examples below, but the present invention is not limited to the scope of the given examples.
实施例1:化合物T-1Example 1: Compound T-1
Figure PCTCN2022088343-appb-000017
Figure PCTCN2022088343-appb-000017
中间体2的合成:Synthesis of Intermediate 2:
将化合物1(10mmol),2,4-二氯-5-氟嘧啶(11mmol)溶解于20ml二氧六环中,然后加入Pd(PPh 3) 2Cl 2(3mmol),碳酸钠水溶液(20ml,2N),使用氮气置换空气三次后升温至100℃反应,约2小时后,TLC检测原料反应完全,停止反应,待反应液冷却后,加入适量水,使用乙酸乙酯萃取三次,合并有几层并用饱和氯化钠水溶液洗涤1次,无水硫酸钠干燥,抽滤,浓缩,最后使用柱层析分离纯化得白色固体(中间体2),收率约80%。HRMS[ESI] +,calcd for C 11H 7ClF 2N 2O[M+H] +,257.0288,found 257.0278. Compound 1 (10mmol), 2,4-dichloro-5-fluoropyrimidine (11mmol) was dissolved in 20ml dioxane, then Pd(PPh 3 ) 2 Cl 2 (3mmol), sodium carbonate aqueous solution (20ml, 2N), replace the air with nitrogen for three times and then raise the temperature to 100°C for reaction. After about 2 hours, TLC detects that the raw materials have reacted completely, and stop the reaction. After the reaction liquid is cooled, add an appropriate amount of water, extract three times with ethyl acetate, and combine several layers It was washed once with saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, filtered with suction, concentrated, and finally separated and purified by column chromatography to obtain a white solid (intermediate 2) with a yield of about 80%. HRMS[ESI] + ,calcd for C 11 H 7 ClF 2 N 2 O[M+H] + ,257.0288,found 257.0278.
中间体3的合成:Synthesis of Intermediate 3:
将中间体2(5mmol)溶解于30ml的DCM中,降温至-10℃以下,然后缓慢滴加三溴化硼(15mmol),控制温度在-10℃以下,待滴加完毕后将反应液移至室温搅拌反应,约2小时后,TLC检测原料反应完全,停止反应,将反应液降温至-10℃以下,加入适量甲醇淬灭,析出大量固体,抽滤,滤饼使用少量冷甲醇洗涤两次,干燥后得黄色固体,即中间体3,无需纯化直接进行下一步,收 率约85%。HRMS[ESI] +,calcd for C 10H 5ClF 2N 2O[M+H] +,243.0131,found 243.0127. Dissolve intermediate 2 (5mmol) in 30ml of DCM, lower the temperature to below -10°C, then slowly add boron tribromide (15mmol) dropwise, control the temperature below -10°C, and pipette the reaction solution after the addition is complete Stir the reaction at room temperature. After about 2 hours, TLC detects that the reaction of the raw materials is complete. Stop the reaction. Cool the reaction solution to below -10°C and add an appropriate amount of methanol to quench it. Once, a yellow solid was obtained after drying, that is, intermediate 3, which was directly carried out to the next step without purification, and the yield was about 85%. HRMS[ESI] + ,calcd for C 10 H 5 ClF 2 N 2 O[M+H] + ,243.0131,found 243.0127.
中间体4的合成:Synthesis of intermediate 4:
将中间体3(10mmol)溶解在30ml的DMF中,降温至0℃以下,加入碳酸钾(15mmol),然后将溴丙炔(11mmol)缓慢滴加到反应液中,控制温度在0℃以下,滴加完毕后将反应液移至室温搅拌反应,约6小时后,TLC检测原料反应完全,停止反应,反应液抽滤,使用适量DMF洗涤三次滤饼,滤液加入适量水并使用乙酸乙酯萃取三次,合并有机相,使用饱和氯化钠水溶液洗涤3次,无水硫酸钠干燥,抽滤,浓缩,使用柱层析分离纯化得中间体4,收率65%。HRMS[ESI] +,calcd for C 13H 9ClF 2N 2O[M+H] +,283.0444,found 283.0437. Intermediate 3 (10mmol) was dissolved in 30ml of DMF, cooled to below 0°C, potassium carbonate (15mmol) was added, and propyne bromide (11mmol) was slowly added dropwise to the reaction solution, the temperature was controlled below 0°C, After the dropwise addition, move the reaction solution to room temperature and stir for reaction. After about 6 hours, TLC detects that the raw materials have reacted completely, stop the reaction, filter the reaction solution with suction, wash the filter cake three times with an appropriate amount of DMF, add an appropriate amount of water to the filtrate and extract it with ethyl acetate Three times, the organic phases were combined, washed three times with saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, filtered with suction, concentrated, separated and purified by column chromatography to obtain intermediate 4 with a yield of 65%. HRMS[ESI] + ,calcd for C 13 H 9 ClF 2 N 2 O[M+H] + ,283.0444,found 283.0437.
中间体6的合成:Synthesis of Intermediate 6:
将4-硝基水杨酸(5,10mmol)溶解在30ml的DMF中,降温至0℃以下,然后分三次加入氢化钠(22mmol),再将溴丙炔(22mmol)缓慢滴加到反应液中,控制温度在0℃以下,滴加完毕后将反应液移至室温搅拌反应,约10小时后,TLC检测原料反应完全,停止反应,将反应液降温至0℃以下,加入适量水淬灭剩余的氢化钠,并使用乙酸乙酯萃取三次,饱和氯化钠水溶液洗涤3次,无水硫酸钠干燥,抽滤,浓缩,使用柱层析分离纯化得中间体6,收率80%。 1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.14(s,1H),7.84(s,1H),7.72(s,1H),6.11-6.02(m,2H),5.31-5.43(m,4H),4.72-4.74(m,4H). Dissolve 4-nitrosalicylic acid (5, 10mmol) in 30ml of DMF, lower the temperature to below 0°C, then add sodium hydride (22mmol) three times, then slowly add propyne bromide (22mmol) dropwise to the reaction solution In the process, the temperature is controlled below 0°C. After the dropwise addition, the reaction solution is moved to room temperature and stirred for reaction. After about 10 hours, TLC detects that the reaction of the raw materials is complete, stop the reaction, cool the reaction solution to below 0°C, and add an appropriate amount of water to quench The remaining sodium hydride was extracted three times with ethyl acetate, washed three times with saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, filtered with suction, concentrated, separated and purified by column chromatography to obtain intermediate 6 with a yield of 80%. 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.14 (s, 1H), 7.84 (s, 1H), 7.72 (s, 1H), 6.11-6.02 (m, 2H), 5.31-5.43 (m,4H),4.72-4.74(m,4H).
中间体7的合成:Synthesis of Intermediate 7:
将中间体6(2mmol)溶解在20ml四氢呋喃和水(4:1)的混合溶剂中,然后加入氢氧化钠(10mmol),室温搅拌反应约6小时,待TLC检测原料反应完全,停止反应,浓缩除去四氢呋喃,然后使用稀盐酸调节pH至酸性,析出大量固体,抽滤,用水洗涤滤饼三次,真空干燥后得中间体7,无需纯化直接进行下一步,收率85%。HRMS[ESI] +,calcd for C 10H 9NO 5[M-H] -,222.0841,found 222.0831. Dissolve intermediate 6 (2 mmol) in a mixed solvent of 20 ml tetrahydrofuran and water (4:1), then add sodium hydroxide (10 mmol), stir and react at room temperature for about 6 hours, and wait until TLC detects that the reaction of the raw materials is complete, stop the reaction, and concentrate Remove tetrahydrofuran, then use dilute hydrochloric acid to adjust the pH to acidic, precipitate a large amount of solid, filter with suction, wash the filter cake with water three times, and dry in vacuo to obtain intermediate 7, which can be directly carried out to the next step without purification, with a yield of 85%. HRMS[ESI] + ,calcd for C 10 H 9 NO 5 [MH] - ,222.0841,found 222.0831.
中间体8的合成:Synthesis of intermediate 8:
将中间体7(0.5mmol),HATU(1.75mmol)溶解在4ml的四氢呋喃中,然后加入DIPEA(1mmol),室温搅约15分钟,然后加入N,N-二甲基乙二胺(0.5mmol),室温搅拌反应约2小时,TLC检测原料反应完全,停止反应,柱层析分离纯化后可得中间体8,收率56%。HRMS[ESI] +,calcd for C 14H 19N 3O 4[M+H] +,294.1376,found 294.1362. Dissolve Intermediate 7 (0.5mmol), HATU (1.75mmol) in 4ml of tetrahydrofuran, then add DIPEA (1mmol), stir at room temperature for about 15 minutes, then add N,N-dimethylethylenediamine (0.5mmol) , stirred at room temperature for about 2 hours, TLC detected that the reaction of the raw materials was complete, and the reaction was stopped. After separation and purification by column chromatography, intermediate 8 was obtained, with a yield of 56%. HRMS[ESI] + ,calcd for C 14 H 19 N 3 O 4 [M+H] + ,294.1376,found 294.1362.
中间体9的合成:Synthesis of Intermediate 9:
将中间体8(3mmol)溶解在15ml的乙醇和水(4:1)的混合溶剂中,然后加入铁粉(15mmol)和5滴浓盐酸,升温至70℃反应约1小时,TLC检测原料反应完全,停止反应,冷却后,反应液抽滤,滤液使用碳酸钠水溶液调节pH至碱性,然后使用乙酸乙酯萃取,合并有机相,使用饱和氯化钠水溶液洗涤有机相3次,无水硫酸钠干燥,抽滤,浓缩得9,收率75%,无需纯化直接进行下一步反应。HRMS[ESI] +,calcd for C 14H 21N 3O 2[M+H] +,264.1634,found 264.1626. Dissolve intermediate 8 (3 mmol) in 15 ml of a mixed solvent of ethanol and water (4:1), then add iron powder (15 mmol) and 5 drops of concentrated hydrochloric acid, heat up to 70 ° C for about 1 hour, TLC detects the reaction of raw materials Completely, stop the reaction, after cooling, the reaction solution is suction filtered, the filtrate is adjusted to alkaline with sodium carbonate aqueous solution, then extracted with ethyl acetate, the organic phases are combined, and the organic phase is washed 3 times with saturated aqueous sodium chloride solution, anhydrous sulfuric acid It was dried over sodium, filtered with suction, and concentrated to obtain 9 with a yield of 75%, which was directly carried out to the next reaction without purification. HRMS[ESI] + ,calcd for C 14 H 21 N 3 O 2 [M+H] + ,264.1634,found 264.1626.
中间体10的合成:Synthesis of Intermediate 10:
将醋酸钯(0.5mmol)和XPhos(0.6mmol)溶解于60ml二氧六环中,氮气置换空气三次后,于室温搅拌约15分钟备用。将中间体4(10mmol),中间体9(10mmol),碳酸铯(30mmol)加入150ml三颈瓶中,然后加入上述新制的催化剂,氮气置换空气三次,然后升温至110-120℃回流反应,反应2-4小时后,TLC检测原料反应完全,停止反应,趁热抽滤,滤液浓缩后加入适量水,使用二氯甲烷萃取三次,合并有机相并使用饱和氯化钠洗涤2次,无水硫酸钠干燥,抽滤,浓缩,使用柱层析分离纯化得中间体10,收率86%。HRMS[ESI] +,calcd for C 27H 29F 2N 5O 3[M+H] +,510.2238,found 510.2231. Palladium acetate (0.5 mmol) and XPhos (0.6 mmol) were dissolved in 60 ml of dioxane, and the air was replaced with nitrogen three times, then stirred at room temperature for about 15 minutes and set aside. Add intermediate 4 (10mmol), intermediate 9 (10mmol), and cesium carbonate (30mmol) into a 150ml three-neck flask, then add the above-mentioned newly prepared catalyst, replace the air with nitrogen three times, then heat up to 110-120°C for reflux reaction, and react After 2-4 hours, TLC detects that the reaction of the raw materials is complete, stop the reaction, filter while hot, add an appropriate amount of water after the filtrate is concentrated, extract three times with dichloromethane, combine the organic phases and wash twice with saturated sodium chloride, anhydrous sulfuric acid Dry over sodium, filter with suction, concentrate, and use column chromatography to separate and purify to obtain intermediate 10 with a yield of 86%. HRMS[ESI] + ,calcd for C 27 H 29 F 2 N 5 O 3 [M+H] + ,510.2238,found 510.2231.
化合物T-1的合成:Synthesis of compound T-1:
将中间体10(0.5mmol)溶解在20ml二氯甲烷中,加入0.1ml稀盐酸(4N),氮气置换空气三次后,加入Grubbs二代催化剂(0.05mmol),再次使用氮气置换空气三次,然后升温至45℃反应,约2小时后,TLC检测原料反应完全,停止反应,冷却后抽滤,滤液浓缩后使用柱层析分离纯化,得化合物T-1,收率63%。Dissolve intermediate 10 (0.5 mmol) in 20 ml of dichloromethane, add 0.1 ml of dilute hydrochloric acid (4N), replace the air with nitrogen three times, add Grubbs second-generation catalyst (0.05 mmol), replace the air with nitrogen three times, and then raise the temperature Reaction at 45°C. After about 2 hours, TLC detected that the reaction of the raw materials was complete, and the reaction was stopped. After cooling, it was filtered with suction. After the filtrate was concentrated, it was separated and purified by column chromatography to obtain compound T-1 with a yield of 63%.
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.72(s,1H),8.19(s,2H),7.82(t,J=6.0Hz,2H),7.58(s,1H),7.47(s,J=9.0Hz,1H),6.90(d,J=9.0Hz,1H),4,30(s,2H),4.10(d,J=6Hz,2H),3.75(s,4H),3.62-3.60(m,3H),3.29-3.27(m,2H),3.20(s,4H),1.98(s,2H),1.90(s,2H),1.72(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.72 (s, 1H), 8.19 (s, 2H), 7.82 (t, J = 6.0Hz, 2H), 7.58 (s, 1H), 7.47(s, J=9.0Hz, 1H), 6.90(d, J=9.0Hz, 1H), 4, 30(s, 2H), 4.10(d, J=6Hz, 2H), 3.75(s, 4H) ,3.62-3.60(m,3H),3.29-3.27(m,2H),3.20(s,4H),1.98(s,2H),1.90(s,2H),1.72(s,4H).
实施例2-4所述化合物的制备方法可参考实施例1中的化合物T-1的合成方法和路线进行合成:The preparation method of the compound described in Examples 2-4 can be synthesized with reference to the synthesis method and route of compound T-1 in Example 1:
实施例2:化合物T-2Embodiment 2: Compound T-2
Figure PCTCN2022088343-appb-000018
Figure PCTCN2022088343-appb-000018
1H NMR(300MHz,CDCl 3):δ(ppm)=8.68(s,1H),8.38(s,1H),8.23(d,J=9.0Hz,1H),8.12(d,J=9.0Hz,1H),7.97(d,J=9.0Hz,1H),7.76-7.71(m,1H),7.49(s,1H),7.23(t,J=9.0Hz,1H),6.65(d,J=9.0Hz,1H),5.80(q,J=15.0Hz,1H),5.50(q,J=15.0Hz,1H),4.90(s,2H),4.80(s,2H),3.76(t,J=6.0Hz,4H),3.64(q,J=9.0Hz,2H),2.64(t,J=6.0Hz,2H),2.54(s,4H). 1 H NMR (300MHz, CDCl 3 ): δ (ppm) = 8.68 (s, 1H), 8.38 (s, 1H), 8.23 (d, J = 9.0Hz, 1H), 8.12 (d, J = 9.0Hz, 1H), 7.97(d, J=9.0Hz, 1H), 7.76-7.71(m, 1H), 7.49(s, 1H), 7.23(t, J=9.0Hz, 1H), 6.65(d, J=9.0 Hz,1H),5.80(q,J=15.0Hz,1H),5.50(q,J=15.0Hz,1H),4.90(s,2H),4.80(s,2H),3.76(t,J=6.0 Hz,4H),3.64(q,J=9.0Hz,2H),2.64(t,J=6.0Hz,2H),2.54(s,4H).
实施例3:化合物T-3Embodiment 3: Compound T-3
Figure PCTCN2022088343-appb-000019
Figure PCTCN2022088343-appb-000019
1H NMR(300MHz,CDCl 3):δ(ppm)=8.60(s,1H),8.30(s,1H),8.20(d,J=9.0Hz,1H),8.06(d,J=9.0Hz,1H),7.91(d,J=9.0Hz,1H),7.75-7.69(m,1H),7.45(s,1H),7.25(t,J=9.0Hz,1H),6.60(d,J=9.0Hz,1H),5.70(q,J=15.0Hz,1H),5.40(q,J=15.0Hz,1H),4.80(s,2H),4.44(t,J=6.0Hz,2H),3.75(t,J=6.0Hz,4H),3.70-3.59(m,2H),2.65(t,J=6.0Hz,2H),2.51-2.40(m,6H). 1 H NMR (300MHz, CDCl 3 ): δ (ppm) = 8.60 (s, 1H), 8.30 (s, 1H), 8.20 (d, J = 9.0Hz, 1H), 8.06 (d, J = 9.0Hz, 1H), 7.91(d, J=9.0Hz, 1H), 7.75-7.69(m, 1H), 7.45(s, 1H), 7.25(t, J=9.0Hz, 1H), 6.60(d, J=9.0 Hz,1H),5.70(q,J=15.0Hz,1H),5.40(q,J=15.0Hz,1H),4.80(s,2H),4.44(t,J=6.0Hz,2H),3.75( t,J=6.0Hz,4H),3.70-3.59(m,2H),2.65(t,J=6.0Hz,2H),2.51-2.40(m,6H).
实施例4:化合物T-4Embodiment 4: Compound T-4
Figure PCTCN2022088343-appb-000020
Figure PCTCN2022088343-appb-000020
1H NMR(300MHz,CDCl 3):δ(ppm)=8.65(s,1H),8.35(s,1H),8.21(d,J=9.0Hz,1H),8.12(d,J=9.0Hz,1H),7.95(d,J=9.0Hz,1H),7.79-7.70(m,1H),7.50(s,1H),7.30(t,J=9.0Hz,1H),6.65(d,J=9.0Hz,1H),5.50(q,J=15.0Hz,1H),5.35 (q,J=15.0Hz,1H),4.44(t,J=6.0Hz,2H),4.36(t,J=6.0Hz,2H),3.72(t,J=6.0Hz,4H),3.68-3.60(m,2H),2.65(t,J=6.0Hz,2H),2.55(s,4H),2.21(q,J=6.0Hz,2H),2.1(q,J=6.0Hz,2H). 1 H NMR (300MHz, CDCl 3 ): δ (ppm) = 8.65 (s, 1H), 8.35 (s, 1H), 8.21 (d, J = 9.0Hz, 1H), 8.12 (d, J = 9.0Hz, 1H), 7.95(d, J=9.0Hz, 1H), 7.79-7.70(m, 1H), 7.50(s, 1H), 7.30(t, J=9.0Hz, 1H), 6.65(d, J=9.0 Hz,1H),5.50(q,J=15.0Hz,1H),5.35(q,J=15.0Hz,1H),4.44(t,J=6.0Hz,2H),4.36(t,J=6.0Hz, 2H), 3.72(t, J=6.0Hz, 4H), 3.68-3.60(m, 2H), 2.65(t, J=6.0Hz, 2H), 2.55(s, 4H), 2.21(q, J=6.0 Hz,2H),2.1(q,J=6.0Hz,2H).
实施列5:化合物T-5Embodiment 5: Compound T-5
化合物T-5的合成路线如下:The synthetic route of compound T-5 is as follows:
Figure PCTCN2022088343-appb-000021
Figure PCTCN2022088343-appb-000021
中间体12的合成:Synthesis of intermediate 12:
中间体12的合成以中间体2和2-甲氧基-4-氨基苯甲酸甲酯(中间体11)为反应原料,合成方法同中间体10的合成,收率73%。HRMS[ESI] +,calcd for C 20H 17F 2N 3O 4[M+H] +,402.1187,found 402.1180. The synthesis of intermediate 12 takes intermediate 2 and methyl 2-methoxy-4-aminobenzoate (intermediate 11) as reaction raw materials, and the synthesis method is the same as that of intermediate 10, with a yield of 73%. HRMS[ESI] + ,calcd for C 20 H 17 F 2 N 3 O 4 [M+H] + ,402.1187,found 402.1180.
中间体13的合成:Synthesis of intermediate 13:
将中间体12(10mmol)溶解于60ml的DCM中,降温至-10℃以下,然后缓慢滴加三溴化硼(45mmol),控制温度在-10℃以下,待滴加完毕后将反应液移至室温搅拌反应,约1.5小时后,TLC检测原料反应完全,停止反应,将反应液降温至-10℃以下,加入适量甲醇淬灭,析出大量固体,抽滤,滤液使用冷甲醇洗涤两次,干燥后得黄色固体,即中间体13,收率65%,无需纯化直接进行下一步。HRMS[ESI] +,calcd for C 18H 13F 2N 3O 4[M+H] +,374.0874,found 374.0863. Dissolve intermediate 12 (10mmol) in 60ml of DCM, lower the temperature to below -10°C, then slowly add boron tribromide (45mmol) dropwise, control the temperature below -10°C, and pipette the reaction solution after the addition is complete Stir the reaction at room temperature. After about 1.5 hours, TLC detects that the reaction of the raw materials is complete, stop the reaction, cool the reaction solution to below -10°C, add an appropriate amount of methanol to quench, and precipitate a large amount of solid, filter with suction, and wash the filtrate twice with cold methanol. After drying, a yellow solid was obtained, ie intermediate 13, with a yield of 65%, which was directly carried out to the next step without purification. HRMS[ESI] + ,calcd for C 18 H 13 F 2 N 3 O 4 [M+H] + ,374.0874,found 374.0863.
中间体14的合成:Synthesis of Intermediate 14:
将中间体13(5mmol)溶解在20ml的DMF中,加入碳酸铯(12.5mmol)后搅拌约15分钟,然后加入1,4-二溴丁烷(5mmol),于室温下搅拌反应约6小时,TLC检测反应完全,停止反应,反应液抽滤,使用适量DMF洗涤滤饼,滤 液中加入适量水,然后使用稀盐酸(1N)调节pH至酸性,析出大量固体,抽滤,使用适量水洗涤滤饼三次,干燥后柱层析分离纯化得中间体14,收率45%。HRMS[ESI] +,calcd for C 22H 19F 2N 3O 4[M+H] +,428.1344,found 428.1338. Intermediate 13 (5 mmol) was dissolved in 20 ml of DMF, cesium carbonate (12.5 mmol) was added and stirred for about 15 minutes, then 1,4-dibromobutane (5 mmol) was added, and the reaction was stirred at room temperature for about 6 hours, TLC detects that the reaction is complete, stop the reaction, filter the reaction solution with suction, use an appropriate amount of DMF to wash the filter cake, add an appropriate amount of water to the filtrate, and then use dilute hydrochloric acid (1N) to adjust the pH to acidic, precipitate a large amount of solids, filter with suction, and use an appropriate amount of water to wash the filter cake. The cake was obtained three times, dried and purified by column chromatography to obtain intermediate 14 with a yield of 45%. HRMS[ESI] + ,calcd for C 22 H 19 F 2 N 3 O 4 [M+H] + ,428.1344,found 428.1338.
中间体15的合成:Synthesis of Intermediate 15:
将中间体14(2mmol)溶解在20ml的甲醇中,然后加入20%的NaOH水溶液(20ml),升温至75℃反应约5小时,TLC检测原料反应完全,停止反应,反应液冷却,使用稀盐酸调节pH至酸性,析出大量固体,抽滤,使用适量水洗涤滤饼,干燥后得中间体15,收率85%。HRMS[ESI] +,calcd for C 21H 17F 2N 3O 4[M+H] +,414.1187,found 414.1179. Dissolve intermediate 14 (2mmol) in 20ml of methanol, then add 20% NaOH aqueous solution (20ml), heat up to 75°C and react for about 5 hours, TLC detects that the raw materials have reacted completely, stop the reaction, cool the reaction solution, and use dilute hydrochloric acid After adjusting the pH to acidity, a large amount of solids were precipitated, filtered with suction, washed the filter cake with an appropriate amount of water, and dried to obtain intermediate 15 with a yield of 85%. HRMS[ESI] + ,calcd for C 21 H 17 F 2 N 3 O 4 [M+H] + ,414.1187,found 414.1179.
化合物T-5的合成:Synthesis of Compound T-5:
化合物T-5的合成使用中间体15和N,N-二甲基乙二胺为原料,合成方法同中间体8的合成,收率42%。 1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.27(s,1H),8.76(d,J=3.0Hz,1H),8.64(s,1H),8.38(d,J=6.0Hz,1H),8.15(d,J=9.0Hz,1H),7.86(d,J=9.0Hz,1H),7.69-7.65(m,1H),7.49(t,J=9.0Hz,1H),7.02(d,J=6.0Hz,1H),4.45(s,2H),4.25(s,2H),3.48(q,J=6.0Hz,4H),2.33(s,6H),1.97-1.93(m,4H). Compound T-5 was synthesized using intermediate 15 and N,N-dimethylethylenediamine as raw materials, the synthesis method was the same as that of intermediate 8, and the yield was 42%. 1 H NMR (300MHz, DMSO-d 6 ): δ(ppm)=10.27(s,1H),8.76(d,J=3.0Hz,1H),8.64(s,1H),8.38(d,J=6.0 Hz,1H),8.15(d,J=9.0Hz,1H),7.86(d,J=9.0Hz,1H),7.69-7.65(m,1H),7.49(t,J=9.0Hz,1H), 7.02(d, J=6.0Hz, 1H), 4.45(s, 2H), 4.25(s, 2H), 3.48(q, J=6.0Hz, 4H), 2.33(s, 6H), 1.97-1.93(m ,4H).
实施例6-17所述化合物的制备方法可参考化合物T-5的合成方法和路线进行合成:The preparation method of the compound described in Examples 6-17 can be synthesized with reference to the synthesis method and route of compound T-5:
实施例6:化合物T-6Embodiment 6: Compound T-6
Figure PCTCN2022088343-appb-000022
Figure PCTCN2022088343-appb-000022
1H NMR(300MHz,CDCl 3):δ(ppm)=8.63(d,J=3.0Hz,1H),8.38(d,J=3.0Hz,1H),8.29(s,1H),8.22(d,J=9.0Hz,1H),8.14(d,J=6.0Hz,1H),7.76-7.72(m,1H),7.48(s,1H),7.24(t,J=9.0Hz,1H),6.65(dd,J 1=9.0Hz,J 2=3.0Hz,1H),4.44(t,J=6.0Hz,2H),4.36(t,J=6.0Hz,2H),3.76(t,J=6.0Hz,4H),3.70-3.59(m,2H)2.64(t,J=6.0Hz,2H),2.54(s,4H),2.16(q,J=6.0Hz,2H),2.01(q,J=6.0Hz,2H). 1 H NMR (300MHz, CDCl 3 ): δ (ppm) = 8.63 (d, J = 3.0Hz, 1H), 8.38 (d, J = 3.0Hz, 1H), 8.29 (s, 1H), 8.22 (d, J=9.0Hz, 1H), 8.14(d, J=6.0Hz, 1H), 7.76-7.72(m, 1H), 7.48(s, 1H), 7.24(t, J=9.0Hz, 1H), 6.65( dd, J 1 =9.0Hz, J 2 =3.0Hz, 1H), 4.44(t, J=6.0Hz, 2H), 4.36(t, J=6.0Hz, 2H), 3.76(t, J=6.0Hz, 4H), 3.70-3.59(m, 2H), 2.64(t, J=6.0Hz, 2H), 2.54(s, 4H), 2.16(q, J=6.0Hz, 2H), 2.01(q, J=6.0Hz ,2H).
实施例7:化合物T-7Embodiment 7: Compound T-7
Figure PCTCN2022088343-appb-000023
Figure PCTCN2022088343-appb-000023
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.01(s,1H),8.73(d,J=9,1H),8.34(t,J=3Hz,1H),8.15(d,J=9.0Hz,1H),7.92(d,J=9Hz,1H),7.71-7.68(m,1H),7.49-7.43(m,1H),7.02(d,J=6Hz,1H),4.46(s,2H),4.25(s,2H),3.71-3.68(m,2H),1.86(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.01 (s, 1H), 8.73 (d, J = 9, 1H), 8.34 (t, J = 3Hz, 1H), 8.15 (d, J=9.0Hz, 1H), 7.92(d, J=9Hz, 1H), 7.71-7.68(m, 1H), 7.49-7.43(m, 1H), 7.02(d, J=6Hz, 1H), 4.46( s,2H),4.25(s,2H),3.71-3.68(m,2H),1.86(s,4H).
实施例8:化合物T-8Embodiment 8: Compound T-8
Figure PCTCN2022088343-appb-000024
Figure PCTCN2022088343-appb-000024
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.27(s,1H),8.57(d,J=3.0Hz,1H),8.36(d,J=3.0Hz,1H),8.22(dd,J 1=9.0Hz,J 2=3.0Hz,1H),7.82(d,J=9.0Hz,1H),7.68-7.64(m,1H),7.52(s,1H),7.21(t,J=9.0Hz,1H),6.54(d,J=9.0Hz,1H),4.44(t,J=6.0Hz,2H),4.36(t,J=6.0Hz,2H),3.96(t,J=6.0Hz,4H),3.75(t,J=6.0Hz,4H),2.65(t,J=6.0Hz,4H),2.21(q,J=6.0Hz,2H),2.11(q,J=6.0Hz,2H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.27 (s, 1H), 8.57 (d, J = 3.0Hz, 1H), 8.36 (d, J = 3.0Hz, 1H), 8.22 ( dd,J 1 =9.0Hz,J 2 =3.0Hz,1H),7.82(d,J=9.0Hz,1H),7.68-7.64(m,1H),7.52(s,1H),7.21(t,J =9.0Hz, 1H), 6.54(d, J=9.0Hz, 1H), 4.44(t, J=6.0Hz, 2H), 4.36(t, J=6.0Hz, 2H), 3.96(t, J=6.0 Hz, 4H), 3.75(t, J=6.0Hz, 4H), 2.65(t, J=6.0Hz, 4H), 2.21(q, J=6.0Hz, 2H), 2.11(q, J=6.0Hz, 2H).
实施例9:化合物T-9Embodiment 9: Compound T-9
Figure PCTCN2022088343-appb-000025
Figure PCTCN2022088343-appb-000025
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.30(s,1H),8.56(d,J=3.0Hz,1H),8.38(d,J=3.0Hz,1H),8.30(s,1H),8.22(d,J=9.0Hz,1H),7.80-7.72(m,1H),7.52(s,1H),7.21(t,J=9.0Hz,1H),6.60(d,J=9.0Hz,1H),4.44(t,J=6.0Hz,2H),4.36(t,J=6.0Hz,2H),3.72(t,J=6.0Hz,4H),2.55(s,4H),2.16(q,J=6.0Hz,2H),2.01(q,J=6.0Hz,2H),1.80-1.76(m,4H),1.33-1.29(m,1H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.30 (s, 1H), 8.56 (d, J = 3.0Hz, 1H), 8.38 (d, J = 3.0Hz, 1H), 8.30 ( s,1H),8.22(d,J=9.0Hz,1H),7.80-7.72(m,1H),7.52(s,1H),7.21(t,J=9.0Hz,1H),6.60(d,J =9.0Hz, 1H), 4.44(t, J=6.0Hz, 2H), 4.36(t, J=6.0Hz, 2H), 3.72(t, J=6.0Hz, 4H), 2.55(s, 4H), 2.16(q,J=6.0Hz,2H),2.01(q,J=6.0Hz,2H),1.80-1.76(m,4H),1.33-1.29(m,1H).
实施例10:化合物T-10Example 10: Compound T-10
Figure PCTCN2022088343-appb-000026
Figure PCTCN2022088343-appb-000026
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.72(s,1H),8.19(s,2H),7.82(t,J=6.0Hz,2H),7.58(s,1H),7.47(s,J=9.0Hz,1H),6.90(d,J=9.0Hz,1H),4.30(s,2H),4.10(d,J=6Hz,2H),3.75(s,4H),3.62-3.60(m,3H),3.29-3.27(m,2H),3.20(s,4H),1.98(s,2H),1.90(s,2H),1.72(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.72 (s, 1H), 8.19 (s, 2H), 7.82 (t, J = 6.0Hz, 2H), 7.58 (s, 1H), 7.47(s, J=9.0Hz, 1H), 6.90(d, J=9.0Hz, 1H), 4.30(s, 2H), 4.10(d, J=6Hz, 2H), 3.75(s, 4H), 3.62 -3.60(m,3H),3.29-3.27(m,2H),3.20(s,4H),1.98(s,2H),1.90(s,2H),1.72(s,4H).
实施例11:化合物T-11Example 11: Compound T-11
Figure PCTCN2022088343-appb-000027
Figure PCTCN2022088343-appb-000027
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.38(s,1H),8.68(d,J=3.0Hz,1H),8.38(d,J=3.0Hz,1H),8.23(dd,J 1=9.0Hz,J 2=3.0Hz,1H),8.12(d,J=9.0Hz,1H),7.97(d,J=9.0Hz,1H),7.76-7.71(m,1H),7.23(t,J=9.0Hz,1H),6.65(d,J=9.0Hz,1H),4.45(t,J=6.0Hz,2H),4.36(t,J=6.0Hz,2H),4.25(q,J=3.0Hz,1H),4.01(dt,J 1=12.0Hz,J 2=3.0Hz,2H),3.61(t,J=9.0Hz,2H),2.11-1.96(m,4H),1.86-1.79(m,4H),1.64-1.53(m,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.38 (s, 1H), 8.68 (d, J = 3.0Hz, 1H), 8.38 (d, J = 3.0Hz, 1H), 8.23 ( dd,J 1 =9.0Hz,J 2 =3.0Hz,1H), 8.12(d,J=9.0Hz,1H),7.97(d,J=9.0Hz,1H),7.76-7.71(m,1H), 7.23(t, J=9.0Hz, 1H), 6.65(d, J=9.0Hz, 1H), 4.45(t, J=6.0Hz, 2H), 4.36(t, J=6.0Hz, 2H), 4.25( q,J=3.0Hz,1H),4.01(dt,J1 = 12.0Hz,J2=3.0Hz,2H ) ,3.61(t,J=9.0Hz,2H),2.11-1.96(m,4H), 1.86-1.79(m,4H),1.64-1.53(m,4H).
实施例12:化合物T-12Example 12: Compound T-12
Figure PCTCN2022088343-appb-000028
Figure PCTCN2022088343-appb-000028
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.78(s,1H),8.15(s,2H),7.92(t,J=6.0Hz,2H),7.63(s,1H),7.47(s,J=9.0Hz,1H),6.92(d,J=9.0Hz,1H),4,23(s,2H),4.09(s,2H),3.59-3.57(m,3H),3.31-3.28(m,2H),2.61-2.57(m,4H),1.96(s,2H),1.92(s,2H),1.83(s,8H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.78 (s, 1H), 8.15 (s, 2H), 7.92 (t, J = 6.0Hz, 2H), 7.63 (s, 1H), 7.47(s, J=9.0Hz, 1H), 6.92(d, J=9.0Hz, 1H), 4, 23(s, 2H), 4.09(s, 2H), 3.59-3.57(m, 3H), 3.31 -3.28(m,2H),2.61-2.57(m,4H),1.96(s,2H),1.92(s,2H),1.83(s,8H).
实施例13:化合物T-13Example 13: Compound T-13
Figure PCTCN2022088343-appb-000029
Figure PCTCN2022088343-appb-000029
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.81(s,1H),8.21(s,2H),7.79(t,J=6.0Hz,2H),7.61(s,1H),7.50(s,J=9.0Hz,1H),6.91(d,J=9.0Hz,1H),4.31(s,2H),4.12(d,J=6Hz,2H),3.75(s,4H),3.64-3.62(m,1H),2.87-2.84(m,2H),2.63-2.61(m,2H),2.59(s,3H),2.03-1.98(m,4H),1.90-1.87(m,4H),1.78(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.81 (s, 1H), 8.21 (s, 2H), 7.79 (t, J = 6.0Hz, 2H), 7.61 (s, 1H), 7.50(s, J=9.0Hz, 1H), 6.91(d, J=9.0Hz, 1H), 4.31(s, 2H), 4.12(d, J=6Hz, 2H), 3.75(s, 4H), 3.64 -3.62(m,1H),2.87-2.84(m,2H),2.63-2.61(m,2H),2.59(s,3H),2.03-1.98(m,4H),1.90-1.87(m,4H) ,1.78(s,4H).
实施例14:化合物T-14Example 14: Compound T-14
Figure PCTCN2022088343-appb-000030
Figure PCTCN2022088343-appb-000030
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.16(s,1H),8.73(d,J=3.0Hz,1H),8.25-8.20(m,2H),7.86(d,J=9.0Hz,2H),7.59(s,1H),7.47(t,J=12.0Hz,1H),6.92(d,J=9.0Hz,1H),4.31(s,2H),4.0(s,2H),3.53(d,J=6.0Hz,2H),2.77(s,2H),2.53(s,3H),2.47(s,3H),1.91(s,2H),1.73(s,2H),1.57(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.16 (s, 1H), 8.73 (d, J = 3.0Hz, 1H), 8.25-8.20 (m, 2H), 7.86 (d, J =9.0Hz, 2H), 7.59(s, 1H), 7.47(t, J=12.0Hz, 1H), 6.92(d, J=9.0Hz, 1H), 4.31(s, 2H), 4.0(s, 2H ),3.53(d,J=6.0Hz,2H),2.77(s,2H),2.53(s,3H),2.47(s,3H),1.91(s,2H),1.73(s,2H),1.57 (s,4H).
实施例15:化合物T-15Example 15: Compound T-15
Figure PCTCN2022088343-appb-000031
Figure PCTCN2022088343-appb-000031
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.08(s,1H),8.75(s,1H),8.35(s,1H),8.18(s,1H),7.92(d,J=9Hz,1H),7.73-7.69(m,1H),7.49-7.46(m,1H),7.05(d,J=6Hz,1H),4.38(s,2H),4.21(s,2H),3.69-3.66(m,2H),1.93-1.81(m,4H),1.73(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.08 (s, 1H), 8.75 (s, 1H), 8.35 (s, 1H), 8.18 (s, 1H), 7.92 (d, J =9Hz,1H),7.73-7.69(m,1H),7.49-7.46(m,1H),7.05(d,J=6Hz,1H),4.38(s,2H),4.21(s,2H),3.69 -3.66(m,2H),1.93-1.81(m,4H),1.73(s,4H).
实施例16:化合物T-16Example 16: Compound T-16
Figure PCTCN2022088343-appb-000032
Figure PCTCN2022088343-appb-000032
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.81(s,1H),8.20(s,2H),7.83(t,J=6.0Hz,2H),7.57(s,1H),7.49(s,1H),6.93(d,J=9.0Hz,1H),4.4(s,1H),4.21(s,2H),4.12(s,2H),3.45-3.31(m,2H),1.89-1.73(m,6H),1.68-1.42(m,10H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.81 (s, 1H), 8.20 (s, 2H), 7.83 (t, J = 6.0Hz, 2H), 7.57 (s, 1H), 7.49(s,1H),6.93(d,J=9.0Hz,1H),4.4(s,1H),4.21(s,2H),4.12(s,2H),3.45-3.31(m,2H),1.89 -1.73(m,6H),1.68-1.42(m,10H).
实施例17:化合物T-17Example 17: Compound T-17
Figure PCTCN2022088343-appb-000033
Figure PCTCN2022088343-appb-000033
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.20(s,1H),8.75(d,J=3.0Hz,1H),8.30-8.26(m,2H),7.84(d,J=9.0Hz,2H),7.60(s,1H),7.45(t,J=12.0Hz,1H),6.90(d,J=9.0Hz,1H),4.31(s,2H),4.0(s,2H),3.55(d,J=6.0Hz,2H),2.75(s,2H),2.55(s,3H),2.41(s,3H),1.89(s,2H),1.74(s,2H),1.57(s,4H),1.04(t,J=9.0Hz,6H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.20 (s, 1H), 8.75 (d, J = 3.0Hz, 1H), 8.30-8.26 (m, 2H), 7.84 (d, J =9.0Hz, 2H), 7.60(s, 1H), 7.45(t, J=12.0Hz, 1H), 6.90(d, J=9.0Hz, 1H), 4.31(s, 2H), 4.0(s, 2H ),3.55(d,J=6.0Hz,2H),2.75(s,2H),2.55(s,3H),2.41(s,3H),1.89(s,2H),1.74(s,2H),1.57 (s,4H),1.04(t,J=9.0Hz,6H).
实施例18:化合物T-18Example 18: Compound T-18
化合物T-18的合成:Synthesis of compound T-18:
Figure PCTCN2022088343-appb-000034
Figure PCTCN2022088343-appb-000034
(1)T-18-1的合成:LW-012-1的合成可参考化合物T5的合成方法与路线。(1) Synthesis of T-18-1: For the synthesis of LW-012-1, please refer to the synthesis method and route of compound T5.
(2)将上述得到的T-18-1(0.35mmol)溶解在2ml二氯甲烷中,然后加入三氟乙酸(3ml),氮气保护,常温下反应约2小时,TLC检测反应完全,停止反应,使用2N的碳酸钠水溶液调节pH至碱性,并加入适量水和二氯甲烷搅拌约15分钟,分离有几层,无水硫酸钠干燥,抽滤,浓缩,使用柱层析分离纯化,得约105mg固体(T-19),收率63%。 1H NMR(300MHz,DMSO-d 6):δ(ppm)= 8.84(s,1H),8.51(s,1H),8.23(s,1H),7.74(s,2H),7.54(m,1H),7.49(s,1H),6.99(s,1H),4.18(s,2H),4.02(s,2H),3.64-3.34(m,2H),3.2(s,4H),2.5-2.34(m,6H),1.90-1.81(m,4H),1.68-1.51(m,4H). (2) Dissolve the T-18-1 (0.35mmol) obtained above in 2ml of dichloromethane, then add trifluoroacetic acid (3ml), under nitrogen protection, react at room temperature for about 2 hours, TLC detects that the reaction is complete, stop the reaction , use 2N sodium carbonate aqueous solution to adjust the pH to alkaline, add appropriate amount of water and dichloromethane and stir for about 15 minutes, separate several layers, dry over anhydrous sodium sulfate, filter with suction, concentrate, and use column chromatography to separate and purify to obtain About 105 mg solid (T-19), yield 63%. 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.84 (s, 1H), 8.51 (s, 1H), 8.23 (s, 1H), 7.74 (s, 2H), 7.54 (m, 1H ),7.49(s,1H),6.99(s,1H),4.18(s,2H),4.02(s,2H),3.64-3.34(m,2H),3.2(s,4H),2.5-2.34( m,6H),1.90-1.81(m,4H),1.68-1.51(m,4H).
实施例19-22所述的制备方法可参考化合物T-18的合成方法与路线进行合成:The preparation method described in Examples 19-22 can be synthesized with reference to the synthesis method and route of compound T-18:
实施例19:化合物T-19Example 19: Compound T-19
Figure PCTCN2022088343-appb-000035
Figure PCTCN2022088343-appb-000035
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.81(s,1H),8.59(s,1H),8.26(s,1H),7.79(s,2H),7.42(m,1H),7.41(s,1H),6.91(s,1H),4.31(s,2H),4.19(s,2H),3.61-3.55(m,1H),2.87-2.60(m,3H),2.13-1.98(m,2H),1.96-1.86(m,6H),1.75-1.51(m,6H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.81 (s, 1H), 8.59 (s, 1H), 8.26 (s, 1H), 7.79 (s, 2H), 7.42 (m, 1H ),7.41(s,1H),6.91(s,1H),4.31(s,2H),4.19(s,2H),3.61-3.55(m,1H),2.87-2.60(m,3H),2.13- 1.98(m,2H),1.96-1.86(m,6H),1.75-1.51(m,6H).
实施例20:化合物T-20Example 20: Compound T-20
Figure PCTCN2022088343-appb-000036
Figure PCTCN2022088343-appb-000036
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.78(s,1H),8.16(s,2H),7.83(t,J=6.0Hz,2H),7.65(s,1H),7.49(s,J=9.0Hz,1H),6.94(d,J=9.0Hz,1H),4.26(s,2H),4.08(s,2H),3.71(s,4H),3.63-3.60(m,1H),2.86-2.84(m,2H),2.65-2.61(m,3H),2.10-1.99(m,4H),1.93-1.86(m,4H),1.75(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.78 (s, 1H), 8.16 (s, 2H), 7.83 (t, J = 6.0Hz, 2H), 7.65 (s, 1H), 7.49(s, J=9.0Hz, 1H), 6.94(d, J=9.0Hz, 1H), 4.26(s, 2H), 4.08(s, 2H), 3.71(s, 4H), 3.63-3.60(m ,1H),2.86-2.84(m,2H),2.65-2.61(m,3H),2.10-1.99(m,4H),1.93-1.86(m,4H),1.75(s,4H).
实施例21:化合物T-21Example 21: Compound T-21
Figure PCTCN2022088343-appb-000037
Figure PCTCN2022088343-appb-000037
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.79(s,1H),8.50(s,1H),8.25(s,1H),7.76(s,2H),7.53(m,1H),7.46(s,1H),6.91(s,1H),4.19(s,2H),4.04(s,2H),3.66-3.36(m,2H),3.3(s,4H),2.54-2.38(m,6H),1.91-1.83(m,4H),1.66-1.51(m,2H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.79 (s, 1H), 8.50 (s, 1H), 8.25 (s, 1H), 7.76 (s, 2H), 7.53 (m, 1H ),7.46(s,1H),6.91(s,1H),4.19(s,2H),4.04(s,2H),3.66-3.36(m,2H),3.3(s,4H),2.54-2.38( m,6H),1.91-1.83(m,4H),1.66-1.51(m,2H).
实施例22:化合物T-22Example 22: Compound T-22
Figure PCTCN2022088343-appb-000038
Figure PCTCN2022088343-appb-000038
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.82(s,1H),8.53(s,1H),8.26(s,1H),7.78(s,2H),7.55(m,1H),7.48(s,1H),6.90(s,1H),4.19(s,2H),4.08(s,2H),3.63-3.34(m,2H),2.51-2.36(m,4H),1.98-1.85(m,4H),1.69(s,2H),1.61-1.50(m,2H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.82 (s, 1H), 8.53 (s, 1H), 8.26 (s, 1H), 7.78 (s, 2H), 7.55 (m, 1H ),7.48(s,1H),6.90(s,1H),4.19(s,2H),4.08(s,2H),3.63-3.34(m,2H),2.51-2.36(m,4H),1.98- 1.85(m,4H),1.69(s,2H),1.61-1.50(m,2H).
实施例23:化合物T-23Example 23: Compound T-23
化合物T-23的合成:Synthesis of Compound T-23:
Figure PCTCN2022088343-appb-000039
Figure PCTCN2022088343-appb-000039
(1)化合物T-23-1的合成可参考化合物T-5的合成方法与路线。(1) For the synthesis of compound T-23-1, please refer to the synthetic method and route of compound T-5.
(2)将上述获得的T-23-1(0.4mmol)溶解在5ml的甲醇中,然后加入50%的氢氧化钠水溶液(3ml),室温搅拌2小时,TLC检测原料反应完全,停止反应,使用稀盐酸调节pH至酸性,析出白色固体,抽滤后,滤饼干燥,使用柱层析分离得化合物T-23,收率54%。(2) Dissolve T-23-1 (0.4 mmol) obtained above in 5 ml of methanol, then add 50% aqueous sodium hydroxide solution (3 ml), stir at room temperature for 2 hours, TLC detects that the reaction of raw materials is complete, stop the reaction, Diluted hydrochloric acid was used to adjust the pH to acidic, and a white solid was precipitated. After suction filtration, the filter cake was dried and separated by column chromatography to obtain compound T-23 with a yield of 54%.
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.91(s,1H),8.88(s,1H),8.57(s,1H),8.29(s,1H),7.76(s,2H),7.58(m,1H),7.48(s,1H),6.99(s,1H),4.31(s,2H),4.09(s,2H),3.63-3.43(m,1H),2.18-2.03(m,1H),1.98-1.73(m,8H),1.61-1.43(m,8H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.91 (s, 1H), 8.88 (s, 1H), 8.57 (s, 1H), 8.29 (s, 1H), 7.76 (s, 2H ),7.58(m,1H),7.48(s,1H),6.99(s,1H),4.31(s,2H),4.09(s,2H),3.63-3.43(m,1H),2.18-2.03( m,1H),1.98-1.73(m,8H),1.61-1.43(m,8H).
实施例24:化合物T-24Example 24: Compound T-24
化合物T-24的合成路线:The synthetic route of compound T-24:
Figure PCTCN2022088343-appb-000040
Figure PCTCN2022088343-appb-000040
中间体16的合成:Synthesis of intermediate 16:
将中间体3(10mmol),6-氯-2-己酮(11mmol)溶解在30ml的DMF中,加入碳酸钾(15mmol),然后加入碘化钾(1mmol),升温至60℃反应,约3小时后,TLC检测原料反应完全,停止反应,反应液抽滤,使用适量乙酸乙酯洗涤滤饼,滤液加入适量水并使用乙酸乙酯萃取三次,合并有机相,使用饱和氯化钠水溶液洗涤3次,无水硫酸钠干燥,抽滤,浓缩,使用柱层析分离纯化得中间体16,收率53%。 1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.58(s,1H),7.54(t,J=6Hz,1H),7.35-7.29(m,2H),4.21(t,J=6Hz,2H),2.63(t,J=6Hz,2H),2.04(s,1H),1.71-1.54(m,4H). Dissolve Intermediate 3 (10mmol), 6-chloro-2-hexanone (11mmol) in 30ml of DMF, add potassium carbonate (15mmol), then add potassium iodide (1mmol), heat up to 60°C for reaction, after about 3 hours , TLC detects that the reaction of raw materials is complete, the reaction is stopped, the reaction solution is filtered with suction, the filter cake is washed with an appropriate amount of ethyl acetate, the filtrate is added with an appropriate amount of water and extracted three times with ethyl acetate, the organic phases are combined, and washed three times with saturated aqueous sodium chloride solution, Dry over anhydrous sodium sulfate, filter with suction, concentrate, and separate and purify by column chromatography to obtain intermediate 16 with a yield of 53%. 1 H NMR (300MHz, DMSO-d 6 ): δ(ppm)=8.58(s,1H),7.54(t,J=6Hz,1H),7.35-7.29(m,2H),4.21(t,J= 6Hz, 2H), 2.63(t, J=6Hz, 2H), 2.04(s, 1H), 1.71-1.54(m, 4H).
中间体17的合成:Synthesis of Intermediate 17:
将中间体16(10mmol)溶于40ml甲醇中,降温至0℃以下,然后分批次加入硼氢化钠(15mmol),反应液升温至室温反应约1h,TLC检测原料反应完全,停止反应,将反应液浓缩后降至0℃,加入适量冰水淬灭,然后使用乙酸乙酯萃取三次,再使用饱和氯化钠水溶液洗涤一次,无水硫酸钠干燥,抽滤,浓缩,使用异丙醚重结晶得到中间体17,收率83%。 1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.61(s,1H),7.51(t,J=6Hz,1H),7.35-7.30(m,2H),4.17(t,J=6Hz,2H),3.67-3.61(m,1H),1.76-1.58(m,6H),1.24(d,J=8Hz,3H). Dissolve intermediate 16 (10 mmol) in 40 ml of methanol, lower the temperature to below 0°C, then add sodium borohydride (15 mmol) in batches, the reaction solution is warmed up to room temperature and reacted for about 1 h, TLC detects that the reaction of the raw materials is complete, stop the reaction, and The reaction solution was concentrated and lowered to 0°C, quenched by adding an appropriate amount of ice water, extracted three times with ethyl acetate, washed once with saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, filtered with suction, concentrated, and reconstituted with isopropyl ether. Crystallization gave intermediate 17 in a yield of 83%. 1 H NMR (300MHz, DMSO-d 6 ): δ(ppm)=8.61(s,1H),7.51(t,J=6Hz,1H),7.35-7.30(m,2H),4.17(t,J= 6Hz, 2H), 3.67-3.61(m, 1H), 1.76-1.58(m, 6H), 1.24(d, J=8Hz, 3H).
中间体18的合成:Synthesis of intermediate 18:
将中间体17(10mmol)溶解在40ml的二氯甲烷中,然后加入三乙胺(20mmol),反应液降温至0℃以下,然后分批次加入对甲苯磺酰氯(20mmol),加完后升温至回流反应约3个小时,TLC检测原料反应完全,停止反应,冷却后加入适量冰水,搅拌15分钟后,分离出有机相并使用饱和氯化钠水溶液洗涤两次,无水硫酸钠干燥,抽滤,浓缩,使用柱层析分离纯化得中间体18,收率74%。 1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.21(s,1H),7.77-7.71(m,2H),7.50(t,J=8Hz,1H),7.40(m,2H),7.26-7.19(m,2H),4.56-4.49(m,1H),4.23(t,J=6Hz,2H),2.01(s,3H),1.74-1.61(m,4H),1.39-1.25(m,5H). Dissolve intermediate 17 (10mmol) in 40ml of dichloromethane, then add triethylamine (20mmol), cool the reaction solution below 0°C, then add p-toluenesulfonyl chloride (20mmol) in batches, and heat up after the addition After about 3 hours of reflux reaction, TLC detected that the reaction of the raw materials was complete, and the reaction was stopped. After cooling, an appropriate amount of ice water was added, and after stirring for 15 minutes, the organic phase was separated and washed twice with saturated aqueous sodium chloride solution, and dried over anhydrous sodium sulfate. Suction filtration, concentration, separation and purification by column chromatography gave intermediate 18 with a yield of 74%. 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.21 (s, 1H), 7.77-7.71 (m, 2H), 7.50 (t, J = 8Hz, 1H), 7.40 (m, 2H) ,7.26-7.19(m,2H),4.56-4.49(m,1H),4.23(t,J=6Hz,2H),2.01(s,3H),1.74-1.61(m,4H),1.39-1.25( m,5H).
中间体19的合成:Synthesis of Intermediate 19:
将中间体18(12mmol)和2-羟基-4-硝基苯甲酸甲酯(10mmol)溶解在80ml的DMF中,然后加入碳酸钾(15mmol),反应液升温至80℃反应过夜,待TLC检测原料反应完全,停止反应,反应液冷却后,抽滤,向滤液中加入适量水,并使用二氯甲烷萃取三次,合并有机相,并依次使用水、饱和氯化钠水溶液分别洗涤三次有机相,有机相使用无水硫酸钠干燥,抽滤,浓缩,使用乙酸乙酯和甲醇的混合溶剂重结晶得到中间体19,收率54%。 1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.51(s,1H),8.27(s,1H),7.79-7.70(m,2H),7.53(t,J=8Hz,1H),7.42(m,2H),7.28-7.21(m,2H),4.13(t,J=6Hz,2H),3.95(s,3H),3.83-3.75(m,1H),2.01(s,3H),1.69-1.63(m,4H),1.41-1.36(m,5H). Intermediate 18 (12mmol) and methyl 2-hydroxy-4-nitrobenzoate (10mmol) were dissolved in 80ml of DMF, then potassium carbonate (15mmol) was added, and the reaction solution was heated to 80°C for overnight reaction until TLC detection The raw material reaction is complete, stop the reaction, after the reaction solution is cooled, suction filter, add appropriate amount of water to the filtrate, and use dichloromethane to extract three times, combine the organic phase, and use water, saturated sodium chloride aqueous solution to wash the organic phase respectively three times successively, The organic phase was dried with anhydrous sodium sulfate, filtered with suction, concentrated, and recrystallized with a mixed solvent of ethyl acetate and methanol to obtain intermediate 19 with a yield of 54%. 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.51 (s, 1H), 8.27 (s, 1H), 7.79-7.70 (m, 2H), 7.53 (t, J = 8Hz, 1H) ,7.42(m,2H),7.28-7.21(m,2H),4.13(t,J=6Hz,2H),3.95(s,3H),3.83-3.75(m,1H),2.01(s,3H) ,1.69-1.63(m,4H),1.41-1.36(m,5H).
中间体20,21和22的合成可参考化合物T1合成路线中的制备方法。The synthesis of intermediates 20, 21 and 22 can refer to the preparation method in the synthetic route of compound T1.
化合物T-24的合成:Synthesis of Compound T-24:
化合物T-24的合成使用中间体22和N-(2-氨基乙基)吗啉为原料,合成方法同中间体8的合成,收率42%。 1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.79(s,1H),8.53(s,1H),8.25(s,1H),7.79(s,2H),7.51(m,1H),7.49(s,1H),6.98(s,1H),4.31(s,2H),4.20-4.04(m,1H),3.87(s,4H),3.66(s,2H),2.6(s,2H),2.49(s,4H),3.3(s,4H),1.90-1.82(m,4H),1.69-1.50(m,5H). The synthesis of compound T-24 uses intermediate 22 and N-(2-aminoethyl)morpholine as raw materials, the synthesis method is the same as that of intermediate 8, and the yield is 42%. 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.79 (s, 1H), 8.53 (s, 1H), 8.25 (s, 1H), 7.79 (s, 2H), 7.51 (m, 1H ),7.49(s,1H),6.98(s,1H),4.31(s,2H),4.20-4.04(m,1H),3.87(s,4H),3.66(s,2H),2.6(s, 2H),2.49(s,4H),3.3(s,4H),1.90-1.82(m,4H),1.69-1.50(m,5H).
实施例25:化合物T-25Example 25: Compound T-25
化合物T-25的合成路线:The synthetic route of compound T-25:
Figure PCTCN2022088343-appb-000041
Figure PCTCN2022088343-appb-000041
中间体24的合成:Synthesis of intermediate 24:
将中间体23(10mmol),1,5-二溴戊烷(12mmol)溶解在30ml的DMF中,加入碳酸钾(15mmol),然后加入碘化钾(1mmol),升温至100℃反应,约8小时后,TLC检测原料反应完全,停止反应,反应液抽滤,使用适量二氯甲烷洗涤滤饼,滤液加入适量水并使用二氯甲烷萃取三次,合并有机相,使用饱和氯化钠水溶液洗涤3次,无水硫酸钠干燥,抽滤,浓缩,使用柱层析分离纯化得中间体23,收率43%。HRMS[ESI] +,calcd for C 13H 17BrN 2O 4[M+H] +,345.0372,found 345.0371. Dissolve intermediate 23 (10mmol), 1,5-dibromopentane (12mmol) in 30ml of DMF, add potassium carbonate (15mmol), then add potassium iodide (1mmol), heat up to 100°C for reaction, after about 8 hours , TLC detects that the reaction of the raw material is complete, stop the reaction, the reaction solution is filtered with suction, the filter cake is washed with an appropriate amount of dichloromethane, the filtrate is added with an appropriate amount of water and extracted three times with dichloromethane, the organic phases are combined, and washed three times with saturated aqueous sodium chloride solution, Dry over anhydrous sodium sulfate, filter with suction, concentrate, and separate and purify by column chromatography to obtain intermediate 23 with a yield of 43%. HRMS[ESI] + ,calcd for C 13 H 17 BrN 2 O 4 [M+H] + ,345.0372,found 345.0371.
中间体25的合成:Synthesis of Intermediate 25:
将中间体24(10mmol),中间体3(10mmol)溶解在40ml DMF中,然后加入碳酸钾(15mmol),25℃反应约3小时,TLC检测原料反应完全,停止反应,反应液抽滤,使用适量二氯甲烷洗涤滤饼,滤液加入适量水并使用二氯甲烷萃取三次,合并有机相,使用饱和氯化钠水溶液洗涤3次,无水硫酸钠干燥,抽滤,浓缩,使用柱层析分离纯化得中间体25,收率75%。HRMS[ESI] +,calcd for C 23H 21ClF 2N 4O 5[M+H] +,508.1169,found 508.1173. Intermediate 24 (10mmol) and Intermediate 3 (10mmol) were dissolved in 40ml of DMF, then potassium carbonate (15mmol) was added, and reacted at 25°C for about 3 hours. TLC detected that the raw materials were completely reacted, and the reaction was stopped. The reaction solution was suction filtered and used Wash the filter cake with an appropriate amount of dichloromethane, add an appropriate amount of water to the filtrate and extract three times with dichloromethane, combine the organic phases, wash three times with saturated aqueous sodium chloride solution, dry over anhydrous sodium sulfate, filter with suction, concentrate, and separate by column chromatography Intermediate 25 was purified with a yield of 75%. HRMS[ESI] + ,calcd for C 23 H 21 ClF 2 N 4 O 5 [M+H] + ,508.1169,found 508.1173.
中间体26-28的合成可参照实施例1中所述的合成方法制备获得。The synthesis of intermediates 26-28 can be obtained by referring to the synthesis method described in Example 1.
化合物T-25的合成已中间体28和N-(2-氨基乙基)吗啉为原料,合成方法参照化合物T-1的合成。 1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.79(s,1H),8.21(s,2H),7.78(t,J=6.0Hz,2H),7.63(s,1H),7.45(s,J=9.0Hz,1H),6.92(d,J=9.0Hz,1H),4.32(s,2H),3.75(s,4H),3.62-3.60(m,2H),3.29-3.27(m,2H),3.04(s,2H),2.75(s,4H),1.97(s,2H),1.91(s,2H),1.78(s,2H). The synthesis of compound T-25 uses intermediate 28 and N-(2-aminoethyl)morpholine as raw materials, and the synthesis method refers to the synthesis of compound T-1. 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.79 (s, 1H), 8.21 (s, 2H), 7.78 (t, J = 6.0Hz, 2H), 7.63 (s, 1H), 7.45(s, J=9.0Hz, 1H), 6.92(d, J=9.0Hz, 1H), 4.32(s, 2H), 3.75(s, 4H), 3.62-3.60(m, 2H), 3.29-3.27 (m,2H),3.04(s,2H),2.75(s,4H),1.97(s,2H),1.91(s,2H),1.78(s,2H).
实施例26:化合物T-26Example 26: Compound T-26
Figure PCTCN2022088343-appb-000042
Figure PCTCN2022088343-appb-000042
中间体29的合成:Synthesis of Intermediate 29:
将中间体23(10mmol)溶解于20ml的二氯甲烷中,然后加入三乙胺(15mmol)并将反应液降温至0℃以下,将5-溴戊酰氯(20mmol)缓慢滴加进反应液中,控制温度在0℃以下,滴加完毕后将反应液升温回流反应,约2小时后,TLC检测原料反应完全,停止反应,加入适量冰水,搅拌约15分钟,静置分层,分离出有机相并使用饱和氯化钠水溶液洗涤2次,无水硫酸钠干燥,抽滤,浓缩,然后使用异丙醚重结晶即可获得中间体29,收率65%。HRMS[ESI] +,calcd for C 13H 15BrN 2O 5[M+H] +,359.0164,found 359.0154. Dissolve intermediate 23 (10mmol) in 20ml of dichloromethane, then add triethylamine (15mmol) and cool the reaction solution below 0°C, slowly add 5-bromovaleryl chloride (20mmol) dropwise into the reaction solution , control the temperature below 0°C, after the dropwise addition, raise the temperature of the reaction solution to reflux for reaction, after about 2 hours, TLC detects that the raw materials have reacted completely, stop the reaction, add an appropriate amount of ice water, stir for about 15 minutes, let stand to separate layers, and separate out The organic phase was washed twice with saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, filtered with suction, concentrated, and then recrystallized with isopropyl ether to obtain intermediate 29 with a yield of 65%. HRMS[ESI] + ,calcd for C 13 H 15 BrN 2 O 5 [M+H] + ,359.0164,found 359.0154.
后续的合成步骤可参照实施例25中所述的合成方法与路线,最终可得化合物T-26。 1H NMR(300MHz,DMSO-d 6):δ(ppm)=9.8(s,1H),8.61(s,1H),8.57(s,1H),8.21(s,1H),7.56(s,2H),7.45(m,2H),7.38(s,1H),6.90(s,1H),4.19(s,2H), 3.97(m,4H),3.62-3.59(m,2H),3.00-2.91(m,2H),2.75(s,4H),2.01(s,2H),1.71(s,4H). Subsequent synthetic steps can refer to the synthetic method and route described in Example 25 to finally obtain compound T-26. 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 9.8 (s, 1H), 8.61 (s, 1H), 8.57 (s, 1H), 8.21 (s, 1H), 7.56 (s, 2H ),7.45(m,2H),7.38(s,1H),6.90(s,1H),4.19(s,2H), 3.97(m,4H),3.62-3.59(m,2H),3.00-2.91( m,2H),2.75(s,4H),2.01(s,2H),1.71(s,4H).
实施例27:化合物T-27Example 27: Compound T-27
化合物T-27的合成路线:The synthetic route of compound T-27:
Figure PCTCN2022088343-appb-000043
Figure PCTCN2022088343-appb-000043
化合物T-27的合成可参照实施例26所述的化合物T-26的合成方法。The synthesis of compound T-27 can refer to the synthesis method of compound T-26 described in Example 26.
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.2(s,1H),8.69(s,1H),8.64(s,1H),8.23(s,1H),7.56(s,2H),7.47(m,2H),7.39(s,1H),6.92(s,1H),4.20(s,2H),3.98(m,4H),3.63-3.61(m,2H),3.03-2.91(m,2H),2.79(s,4H),2.08(s,2H),1.76(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.2 (s, 1H), 8.69 (s, 1H), 8.64 (s, 1H), 8.23 (s, 1H), 7.56 (s, 2H ),7.47(m,2H),7.39(s,1H),6.92(s,1H),4.20(s,2H),3.98(m,4H),3.63-3.61(m,2H),3.03-2.91( m,2H),2.79(s,4H),2.08(s,2H),1.76(s,4H).
实施例28:化合物T-28Example 28: Compound T-28
Figure PCTCN2022088343-appb-000044
Figure PCTCN2022088343-appb-000044
化合物T-28的合成可参考实施例26所述的化合物T-26的合成方法。For the synthesis of compound T-28, reference may be made to the synthesis method of compound T-26 described in Example 26.
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.1(s,1H),8.71(s,1H),8.56(s,1H),8.24(s,1H),7.56(s,2H),7.49(m,1H),7.42(s,2H),6.91(s,1H),4.21(s,2H),3.94(m,4H),3.63-3.60(m,2H),3.08-2.95(m,2H),2.83(s,4H),2.00(s,2H),1.93(s,2H),1.73(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.1 (s, 1H), 8.71 (s, 1H), 8.56 (s, 1H), 8.24 (s, 1H), 7.56 (s, 2H ),7.49(m,1H),7.42(s,2H),6.91(s,1H),4.21(s,2H),3.94(m,4H),3.63-3.60(m,2H),3.08-2.95( m,2H),2.83(s,4H),2.00(s,2H),1.93(s,2H),1.73(s,4H).
实施例29:化合物T-29Example 29: Compound T-29
Figure PCTCN2022088343-appb-000045
Figure PCTCN2022088343-appb-000045
化合物T-29的合成可参考实施例27所述的化合物T-27的合成方法。For the synthesis of compound T-29, reference may be made to the synthesis method of compound T-27 described in Example 27.
1H NMR(300MHz,DMSO-d 6):δ(ppm)=9.99(s,1H),8.79(s,1H),8.61(s,1H),8.29(s,1H),7.52(s,2H),7.41(m,1H),7.37(s,1H),6.99(s,2H),4.28(s,2H),3.91(s,4H),3.63-3.56(m,2H),3.09-2.92(m,2H),2.74(s,4H),2.12(s,2H),1.95(s,2H),1.81(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 9.99 (s, 1H), 8.79 (s, 1H), 8.61 (s, 1H), 8.29 (s, 1H), 7.52 (s, 2H ),7.41(m,1H),7.37(s,1H),6.99(s,2H),4.28(s,2H),3.91(s,4H),3.63-3.56(m,2H),3.09-2.92( m,2H),2.74(s,4H),2.12(s,2H),1.95(s,2H),1.81(s,4H).
实施例30和31所述化合物的制备可参考化合物T-5的合成方法和路线,将该路线中的起始原料4-氟-3甲氧基苯硼酸替换为4-氟-2甲氧基苯硼酸,linker连接链和最后一步反应中的氨基侧链替换为特定基团即可。The preparation of the compounds described in Examples 30 and 31 can refer to the synthesis method and route of compound T-5, and the starting material 4-fluoro-3 methoxyphenylboronic acid in this route is replaced by 4-fluoro-2 methoxy Phenylboronic acid, the linker chain and the amino side chain in the last step of the reaction can be replaced with specific groups.
实施例30:化合物T-30Example 30: Compound T-30
Figure PCTCN2022088343-appb-000046
Figure PCTCN2022088343-appb-000046
1H NMR(300MHz,DMSO-d 6):δ(ppm)=9.93(s,1H),8.77(s,1H),8.30(t,J=6.0Hz,1H),8.10(t,J=6.0Hz,1H),8.22(d,J=9.0Hz,1H),7.80-7.72(m,1H),7.53(s,1H),7.16(d,J=9.0Hz,1H),6.99(td,J 1=9.0Hz,J 2=3.0Hz,1H),4.45(t,J=6.0Hz,2H),4.38(t,J=6.0Hz,2H),3.75-3.71(m,4H),2.60-2.56(m,4H),2.20(q,J=6.0Hz,2H),2.11(q,J=6.0Hz,2H),1.80-1.76(m,4H),1.33-1.29(m,1H). 1 H NMR (300MHz, DMSO-d 6 ): δ(ppm)=9.93(s,1H),8.77(s,1H),8.30(t,J=6.0Hz,1H),8.10(t,J=6.0 Hz,1H),8.22(d,J=9.0Hz,1H),7.80-7.72(m,1H),7.53(s,1H),7.16(d,J=9.0Hz,1H),6.99(td,J 1 =9.0Hz, J 2 =3.0Hz, 1H), 4.45(t, J=6.0Hz, 2H), 4.38(t, J=6.0Hz, 2H), 3.75-3.71(m, 4H), 2.60-2.56 (m,4H),2.20(q,J=6.0Hz,2H),2.11(q,J=6.0Hz,2H),1.80-1.76(m,4H),1.33-1.29(m,1H).
实施例31:化合物T-31Example 31: Compound T-31
Figure PCTCN2022088343-appb-000047
Figure PCTCN2022088343-appb-000047
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.03(s,1H),8.71(s,1H),8.35(t,J=6.0Hz,1H),8.22(d,J=9.0Hz,1H),8.10(d,J=6.0Hz,1H),7.83-7.79(m,1H),7.45(s, 1H),7.20(t,J=9.0Hz,1H),6.89(d,J=9.0Hz,1H),4.55(t,J=6.0Hz,2H),4.40(t,J=6.0Hz,2H),3.80(t,J=6.0Hz,4H),3.72-3.65(m,2H)2.62(t,J=6.0Hz,2H),2.52(s,4H),2.11(q,J=6.0Hz,2H),2.01(q,J=6.0Hz,2H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.03 (s, 1H), 8.71 (s, 1H), 8.35 (t, J = 6.0Hz, 1H), 8.22 (d, J = 9.0 Hz,1H),8.10(d,J=6.0Hz,1H),7.83-7.79(m,1H),7.45(s,1H),7.20(t,J=9.0Hz,1H),6.89(d,J =9.0Hz, 1H), 4.55(t, J=6.0Hz, 2H), 4.40(t, J=6.0Hz, 2H), 3.80(t, J=6.0Hz, 4H), 3.72-3.65(m, 2H )2.62(t, J=6.0Hz, 2H), 2.52(s, 4H), 2.11(q, J=6.0Hz, 2H), 2.01(q, J=6.0Hz, 2H).
实施例32-35所述化合物的制备可参考化合物T-5的合成方法和路线,将该路线中的起始原料4-氟-3甲氧基苯硼酸替换为5-甲氧基吡啶-3-硼酸,linker连接链和最后一步反应中的氨基侧链替换为特定基团即可。The preparation of the compounds described in Examples 32-35 can refer to the synthesis method and route of compound T-5, and the starting material 4-fluoro-3 methoxyphenylboronic acid in this route is replaced by 5-methoxypyridine-3 -Boronic acid, the linker chain and the amino side chain in the last step of the reaction can be replaced with specific groups.
实施例32:化合物T-32Example 32: Compound T-32
Figure PCTCN2022088343-appb-000048
Figure PCTCN2022088343-appb-000048
1H NMR(300MHz,DMSO-d 6):δ(ppm)=9.21(s,1H),8.71(s,1H),8.28(s,1H),8.08(s,1H),7.80(s,1H),7.33(m,1H),7.24(s,1H),6.91(s,2H),4.31(s,2H),4.21(s,2H),3.97(s,4H),3.51-3.41(m,2H),2.94-2.81(m,2H),2.78(s,4H),1.95(s,4H),1.74(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 9.21 (s, 1H), 8.71 (s, 1H), 8.28 (s, 1H), 8.08 (s, 1H), 7.80 (s, 1H ),7.33(m,1H),7.24(s,1H),6.91(s,2H),4.31(s,2H),4.21(s,2H),3.97(s,4H),3.51-3.41(m, 2H),2.94-2.81(m,2H),2.78(s,4H),1.95(s,4H),1.74(s,4H).
实施例33:化合物T-33Example 33: Compound T-33
Figure PCTCN2022088343-appb-000049
Figure PCTCN2022088343-appb-000049
1H NMR(300MHz,DMSO-d 6):δ(ppm)=9.01(s,1H),8.81(s,1H),8.26(s,1H),8.03(s,1H),7.79(s,2H),7.39(m,1H),7.26(s,1H),6.99(s,1H),4.28(s,2H),4.13(s,2H),3.59-3.41(m,2H),2.91-2.73(m,6H),1.93-1.84(m,4H),1.69-1.58(m,8H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 9.01 (s, 1H), 8.81 (s, 1H), 8.26 (s, 1H), 8.03 (s, 1H), 7.79 (s, 2H ),7.39(m,1H),7.26(s,1H),6.99(s,1H),4.28(s,2H),4.13(s,2H),3.59-3.41(m,2H),2.91-2.73( m,6H),1.93-1.84(m,4H),1.69-1.58(m,8H).
实施例34:化合物T-34Example 34: Compound T-34
Figure PCTCN2022088343-appb-000050
Figure PCTCN2022088343-appb-000050
1H NMR(300MHz,DMSO-d 6):δ(ppm)=9.19(s,1H),8.89(s,1H),8.32(s,1H),8.05(s,1H),7.83(s,2H),7.35(m,1H),7.21(s,1H),6.92(s,1H),4.21(s,2H),4.11(s,2H),3.82-3.70(m,2H),1.95(s,4H),1.74(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 9.19 (s, 1H), 8.89 (s, 1H), 8.32 (s, 1H), 8.05 (s, 1H), 7.83 (s, 2H ),7.35(m,1H),7.21(s,1H),6.92(s,1H),4.21(s,2H),4.11(s,2H),3.82-3.70(m,2H),1.95(s, 4H), 1.74(s, 4H).
实施例35:化合物T-35Example 35: Compound T-35
Figure PCTCN2022088343-appb-000051
Figure PCTCN2022088343-appb-000051
1H NMR(300MHz,DMSO-d 6):δ(ppm)=9.08(s,1H),8.70(s,1H),8.28(s,1H),8.09(s,1H),7.80(s,1H),7.34(m,2H),7.21(s,1H),6.95(s,1H),4.29(s,2H),4.16(s,2H),3.92(s,4H),3.56-3.42(m,2H),2.93-2.81(m,2H),2.73(s,4H),1.90(s,4H),1.71(s,2H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 9.08 (s, 1H), 8.70 (s, 1H), 8.28 (s, 1H), 8.09 (s, 1H), 7.80 (s, 1H ),7.34(m,2H),7.21(s,1H),6.95(s,1H),4.29(s,2H),4.16(s,2H),3.92(s,4H),3.56-3.42(m, 2H),2.93-2.81(m,2H),2.73(s,4H),1.90(s,4H),1.71(s,2H).
实施例36:化合物T-36Example 36: Compound T-36
实施例36所述化合物的制备可参考化合物T-18的合成方法和路线,将该路线中的起始原料4-氟-3甲氧基苯硼酸替换为5-甲氧基吡啶-3-硼酸,linker连接链和倒数第二步反应中的氨基侧链替换为特定基团即可。The preparation of the compound described in Example 36 can refer to the synthesis method and route of compound T-18, and the starting material 4-fluoro-3 methoxyphenylboronic acid in this route is replaced by 5-methoxypyridine-3-boronic acid , the linker connecting chain and the amino side chain in the penultimate step of the reaction can be replaced with specific groups.
Figure PCTCN2022088343-appb-000052
Figure PCTCN2022088343-appb-000052
1H NMR(300MHz,DMSO-d 6):δ(ppm)=9.01(s,1H),8.73(s,1H),8.23(s,1H),8.04(s,1H),7.83(s,1H),7.35(m,2H),7.30(s,1H),6.91(s,1H),4.23(s,2H),4.15(s,2H),3.91-9.88(m,4H),3.58-3.44(m,3H),2.91-2.78(m,2H),2.71-2.68(m,4H),1.87(s,4H),1.69(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 9.01 (s, 1H), 8.73 (s, 1H), 8.23 (s, 1H), 8.04 (s, 1H), 7.83 (s, 1H ),7.35(m,2H),7.30(s,1H),6.91(s,1H),4.23(s,2H),4.15(s,2H),3.91-9.88(m,4H),3.58-3.44( m,3H),2.91-2.78(m,2H),2.71-2.68(m,4H),1.87(s,4H),1.69(s,4H).
实施例37:化合物T-37Example 37: Compound T-37
实施例37所述化合物的制备可参考化合物T-18的合成方法和路线,将该路线中的起始原料4-氟-3甲氧基苯硼酸替换为5-甲氧基吡啶-3-硼酸,linker连接链和倒数第二步反应中的氨基侧链替换为特定基团即可。The preparation of the compound described in Example 37 can refer to the synthesis method and route of compound T-18, and the starting material 4-fluoro-3 methoxyphenylboronic acid in this route is replaced by 5-methoxypyridine-3-boronic acid , the linker connecting chain and the amino side chain in the penultimate step of the reaction can be replaced with specific groups.
Figure PCTCN2022088343-appb-000053
Figure PCTCN2022088343-appb-000053
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.1(s,1H),9.07(s,1H),8.82(s,1H),8.75(s,1H),8.39(s,1H),8.08(s,1H),7.68-7.54(m,2H),7.38(s,1H),6.98(s,1H),4.20(s,2H),3.87(m,4H),3.61-3.57(m,2H),3.32-3.27(m,2H),2.75(s,4H),2.12(s,2H),1.71(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.1 (s, 1H), 9.07 (s, 1H), 8.82 (s, 1H), 8.75 (s, 1H), 8.39 (s, 1H ),8.08(s,1H),7.68-7.54(m,2H),7.38(s,1H),6.98(s,1H),4.20(s,2H),3.87(m,4H),3.61-3.57( m,2H),3.32-3.27(m,2H),2.75(s,4H),2.12(s,2H),1.71(s,4H).
实施例38:化合物T-38Example 38: Compound T-38
化合物T-38的合成路线:The synthetic route of compound T-38:
Figure PCTCN2022088343-appb-000054
Figure PCTCN2022088343-appb-000054
中间体43的合成可参照中间体18的合成方法。The synthesis of intermediate 43 can refer to the synthesis method of intermediate 18.
中间体45的合成:Synthesis of Intermediate 45:
将中间体44(30mmol),2,4-二氯-5-氟嘧啶(20mmol),碳酸钠(0.6mmol)和1,1'-双二苯基膦二茂铁二氯化钯(0.4mmol)加入到70ml的苯甲醚中,氮气置换空气三次。然后升温至130℃反应过夜,TLC检测原料反应完全,停止反应,冷却后抽滤,使用减压蒸馏(158℃)除去溶剂,然后使用柱层析分离纯化,得到中间体45,收率29%。HRMS[ESI] +,calcd for C 8H 6ClFN 4O[M+H] +,229.0214,found 229.0209. Intermediate 44 (30mmol), 2,4-dichloro-5-fluoropyrimidine (20mmol), sodium carbonate (0.6mmol) and 1,1'-bisdiphenylphosphinoferrocenepalladium dichloride (0.4mmol ) was added in the anisole of 70ml, and the air was replaced with nitrogen three times. Then the temperature was raised to 130°C and reacted overnight. TLC detected that the reaction of the raw materials was complete, and the reaction was stopped. After cooling, it was filtered with suction, and the solvent was removed by vacuum distillation (158°C), and then separated and purified by column chromatography to obtain intermediate 45 with a yield of 29%. . HRMS[ESI] + ,calcd for C 8 H 6 ClFN 4 O[M+H] + ,229.0214,found 229.0209.
中间体46的合成可参照中间体19的合成方法。The synthesis of intermediate 46 can refer to the synthesis method of intermediate 19.
后续中间体的合成方法可参考实施例25所述的化合物T-25的合成方法,最终获得化合物T-38。 1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.81(s,1H),8.74(s,1H),7.96-9.94(m,2H),7.34(m,2H),7.08(s,1H),4.21(s,2H),4.10(s,2H),3.87(s,4H),3.56-3.41(m,2H),3.35(s,3H),2.91-2.80(m,2H),2.71(s,4H),1.74(s,4H),1.35(s,4H). For the synthesis method of subsequent intermediates, refer to the synthesis method of compound T-25 described in Example 25 to finally obtain compound T-38. 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.81 (s, 1H), 8.74 (s, 1H), 7.96-9.94 (m, 2H), 7.34 (m, 2H), 7.08 (s ,1H),4.21(s,2H),4.10(s,2H),3.87(s,4H),3.56-3.41(m,2H),3.35(s,3H),2.91-2.80(m,2H), 2.71(s,4H),1.74(s,4H),1.35(s,4H).
实施例39-40所述化合物的制备可参照化合物T-5的制备方法,其中,将4-氟-3-甲氧基苯硼酸替换成相应的硼酸或硼酸酯,并参照中间体2的合成方法合成所需的起始原料。The preparation of the compounds described in Examples 39-40 can refer to the preparation method of compound T-5, wherein, 4-fluoro-3-methoxyphenylboronic acid is replaced by the corresponding boric acid or boric acid ester, and the intermediate 2 Synthetic Methods The required starting materials were synthesized.
实施例39:化合物T-39Example 39: Compound T-39
Figure PCTCN2022088343-appb-000055
Figure PCTCN2022088343-appb-000055
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.67(s,1H),9.26(s,1H),8.93(s,1H),8.01(d,J=3.0Hz,1H),7.91(d,J=9.0Hz,1H),7.79-7.72(m,1H),7.50(s,1H),7.19(t,J=9.0Hz,1H),6.85(d,J=9.0Hz,1H),4.44(t,J=6.0Hz,2H),4.36(t,J=6.0Hz,2H),3.75(s,4H),2.60(s,4H),2.16(t,J=6.0Hz,2H),2.01(t,J=6.0Hz,2H),1.92-1.88(m,4H),1.74-1.68(m,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.67 (s, 1H), 9.26 (s, 1H), 8.93 (s, 1H), 8.01 (d, J = 3.0Hz, 1H), 7.91(d, J=9.0Hz, 1H), 7.79-7.72(m, 1H), 7.50(s, 1H), 7.19(t, J=9.0Hz, 1H), 6.85(d, J=9.0Hz, 1H ),4.44(t,J=6.0Hz,2H),4.36(t,J=6.0Hz,2H),3.75(s,4H),2.60(s,4H),2.16(t,J=6.0Hz,2H ),2.01(t,J=6.0Hz,2H),1.92-1.88(m,4H),1.74-1.68(m,4H).
实施例40:化合物T-40Example 40: Compound T-40
Figure PCTCN2022088343-appb-000056
Figure PCTCN2022088343-appb-000056
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.47(s,1H),8.72(s,1H),8.26(d,J=3.0Hz,1H),8.19(s,1H),8.09(d,J=6.0Hz,1H),7.67-7.62(m,1H),7.28(s,1H),6.99(d,J=6.0Hz,1H),6.55(d,J=3.0Hz,1H),4.44(s,2H),4.36(s,2H),3.89(s,3H),3.77-3.73(m,4H),2.60-2.55(m,4H),2.11-.2.05(m,4H),1.90-1.85(m,4H),1.72-1.67(m,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.47 (s, 1H), 8.72 (s, 1H), 8.26 (d, J = 3.0Hz, 1H), 8.19 (s, 1H), 8.09(d, J=6.0Hz, 1H), 7.67-7.62(m, 1H), 7.28(s, 1H), 6.99(d, J=6.0Hz, 1H), 6.55(d, J=3.0Hz, 1H ),4.44(s,2H),4.36(s,2H),3.89(s,3H),3.77-3.73(m,4H),2.60-2.55(m,4H),2.11-.2.05(m,4H) ,1.90-1.85(m,4H),1.72-1.67(m,4H).
实施例41-44所述化合物的制备可参照化合物T-5的制备方法,其中,可使用相应的二氯嘧啶衍生物参照中间体2的合成方法合成所需的起始原料。The preparation of the compounds described in Examples 41-44 can refer to the preparation method of compound T-5, wherein the corresponding dichloropyrimidine derivatives can be used to synthesize the required starting materials according to the synthesis method of intermediate 2.
实施例41:化合物T-41Example 41: Compound T-41
Figure PCTCN2022088343-appb-000057
Figure PCTCN2022088343-appb-000057
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.36(s,1H),8.53(d,J=3.0Hz,1H),8.40(d,J=3.0Hz,1H),8.30(s,1H),8.16(d,J=9.0Hz,1H),7.80-7.76(m,1H),7.53(s,1H),7.26(t,J=9.0Hz,1H),6.75(d,J=9.0Hz,1H),4.44(t,J=6.0Hz,2H),4.36(t,J=6.0Hz,2H),3.74(t,J=6.0Hz,4H),2.60(t,J=6.0Hz,4H),2.15(q,J=6.0Hz,2H),2.06(q,J=6.0Hz,2H),1.90(t,J=6.0Hz,4H),1.73(t,J=6.0Hz,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 10.36 (s, 1H), 8.53 (d, J = 3.0Hz, 1H), 8.40 (d, J = 3.0Hz, 1H), 8.30 ( s,1H),8.16(d,J=9.0Hz,1H),7.80-7.76(m,1H),7.53(s,1H),7.26(t,J=9.0Hz,1H),6.75(d,J =9.0Hz, 1H), 4.44(t, J=6.0Hz, 2H), 4.36(t, J=6.0Hz, 2H), 3.74(t, J=6.0Hz, 4H), 2.60(t, J=6.0 Hz, 4H), 2.15(q, J=6.0Hz, 2H), 2.06(q, J=6.0Hz, 2H), 1.90(t, J=6.0Hz, 4H), 1.73(t, J=6.0Hz, 4H).
实施例42:化合物T-42Example 42: Compound T-42
Figure PCTCN2022088343-appb-000058
Figure PCTCN2022088343-appb-000058
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.46(s,1H),8.89(s,1H),8.45(d,J=3.0Hz,1H),8.30(s,1H),8.22(d,J=9.0Hz,1H),7.97-7.92(m,1H),7.48(s,1H),7.22(t,J=9.0Hz,1H),6.70(d,J=9.0Hz,1H),4.44(t,J=6.0Hz,2H),4.36(t,J=6.0Hz,2H),3.73(t,J=6.0Hz,4H),2.62-2.58(m,4H),2.15(q,J=6.0Hz,2H),2.08(q,J=6.0Hz,2H),1.91-1.86(m,4H),1.72-1.67(m,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ(ppm)=10.46(s,1H),8.89(s,1H),8.45(d,J=3.0Hz,1H),8.30(s,1H), 8.22(d, J=9.0Hz, 1H), 7.97-7.92(m, 1H), 7.48(s, 1H), 7.22(t, J=9.0Hz, 1H), 6.70(d, J=9.0Hz, 1H ), 4.44(t, J=6.0Hz, 2H), 4.36(t, J=6.0Hz, 2H), 3.73(t, J=6.0Hz, 4H), 2.62-2.58(m, 4H), 2.15(q ,J=6.0Hz,2H),2.08(q,J=6.0Hz,2H),1.91-1.86(m,4H),1.72-1.67(m,4H).
实施例43:化合物T-43Example 43: Compound T-43
Figure PCTCN2022088343-appb-000059
Figure PCTCN2022088343-appb-000059
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.21(s,1H),8.44(s,1H),8.19(d,J=9.0Hz,2H),8.09(d,J=6.0Hz,1H),7.70-7.66(m,1H),7.25(s,1H),6.93(d,J=6.0 Hz,1H),6.54(d,J=3.0Hz,1H),4.40(s,2H),4.29(s,2H),3.70-3.64(m,4H),2.55-2.50(m,4H),2.16(t,J=6.0Hz,2H),2.12(s,3H),2.09-2.06(m,2H),1.86-1.82(m,4H),1.68-1.64(m,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ(ppm)=10.21(s,1H),8.44(s,1H),8.19(d,J=9.0Hz,2H),8.09(d,J=6.0 Hz, 1H), 7.70-7.66(m, 1H), 7.25(s, 1H), 6.93(d, J=6.0 Hz, 1H), 6.54(d, J=3.0Hz, 1H), 4.40(s, 2H ),4.29(s,2H),3.70-3.64(m,4H),2.55-2.50(m,4H),2.16(t,J=6.0Hz,2H),2.12(s,3H),2.09-2.06( m,2H),1.86-1.82(m,4H),1.68-1.64(m,4H).
实施例44:化合物T-44Example 44: Compound T-44
Figure PCTCN2022088343-appb-000060
Figure PCTCN2022088343-appb-000060
1H NMR(300MHz,DMSO-d 6):δ(ppm)=10.37(s,1H),8.30(s,1H),8.16(s,1H),8.00(d,J=9.0Hz,2H),7.73-7.70(m,1H),7.50(s,1H),7.19(t,J=9.0Hz,1H),6.70(d,J=9.0Hz,1H),4.42(t,J=6.0Hz,2H),4.33(t,J=6.0Hz,2H),3.74(t,J=6.0Hz,4H),2.58(t,J=6.0Hz,4H),2.11(t,J=6.0Hz,2H),2.03(t,J=6.0Hz,2H),1.86(t,J=6.0Hz,4H),1.70(t,J=6.0Hz,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ(ppm)=10.37(s,1H),8.30(s,1H),8.16(s,1H),8.00(d,J=9.0Hz,2H), 7.73-7.70(m,1H),7.50(s,1H),7.19(t,J=9.0Hz,1H),6.70(d,J=9.0Hz,1H),4.42(t,J=6.0Hz,2H ), 4.33(t, J=6.0Hz, 2H), 3.74(t, J=6.0Hz, 4H), 2.58(t, J=6.0Hz, 4H), 2.11(t, J=6.0Hz, 2H), 2.03(t, J=6.0Hz, 2H), 1.86(t, J=6.0Hz, 4H), 1.70(t, J=6.0Hz, 4H).
实施例45:化合物T-45Example 45: Compound T-45
Figure PCTCN2022088343-appb-000061
Figure PCTCN2022088343-appb-000061
化合物T-45的合成:Synthesis of Compound T-45:
(1)T-45-1的合成参考实施例5中所述的合成路线。(1) Synthesis of T-45-1 Refer to the synthetic route described in Example 5.
(2)a:将中间体T-45-1(1mmol)溶解在4ml的无水四氢呋喃中,降温至-10℃,氮气保护下滴加三乙胺(2mmol),然后再加入三甲基乙酰氯(1.2mmol),控制温度在-10℃,并在此温度下反应约1小时备用;b:将乙酰胺(1mmol)溶解在3ml无水四氢呋喃,降温至-78℃,氮气保护下缓慢加入n-Bu-Li(1.2mmol,2.7M in hexane),控制温度在-70℃以下,搅拌约1小时后,将a部分新制的反应液缓慢滴加进去,控制温度在-70℃以下,反应约2小时,TLC检测原料反应完全,反应液捉奸升至室温后倒入适量的氯化铵水溶液中,使用二氯甲烷萃取三次,合并有机相并使用饱和氯化钠水溶液洗涤2次,无水硫酸钠干燥,抽滤,浓缩,最后使用柱层析分离得化合物T-45,收率36%。 1H NMR(300MHz,DMSO-d 6): δ(ppm)=8.77(s,1H),8.18(s,2H),7.82(t,J=6.0Hz,2H),7.61(s,1H),7.44(s,J=9.0Hz,1H),6.90(d,J=9.0Hz,1H),4.22(s,2H),4.09(s,2H),2.76(s,3H)1.94(s,4H),1.75(s,4H). (2)a: Dissolve the intermediate T-45-1 (1mmol) in 4ml of anhydrous tetrahydrofuran, cool down to -10°C, add triethylamine (2mmol) dropwise under nitrogen protection, and then add trimethylethylamine Acyl chloride (1.2mmol), control the temperature at -10°C, and react at this temperature for about 1 hour for later use; b: Dissolve acetamide (1mmol) in 3ml of anhydrous tetrahydrofuran, cool to -78°C, and add slowly under nitrogen protection n-Bu-Li (1.2mmol, 2.7M in hexane), control the temperature below -70°C, stir for about 1 hour, slowly add the newly prepared reaction solution in part a, control the temperature below -70°C, and react After about 2 hours, TLC detected that the reaction of the raw materials was complete. After the reaction solution was raised to room temperature, it was poured into an appropriate amount of ammonium chloride aqueous solution, extracted three times with dichloromethane, and the organic phases were combined and washed twice with saturated aqueous sodium chloride solution. Dry over sodium sulfate, filter with suction, concentrate, and finally use column chromatography to separate compound T-45 with a yield of 36%. 1 H NMR (300MHz, DMSO-d 6 ): δ(ppm)=8.77(s,1H),8.18(s,2H),7.82(t,J=6.0Hz,2H),7.61(s,1H), 7.44(s,J=9.0Hz,1H),6.90(d,J=9.0Hz,1H),4.22(s,2H),4.09(s,2H),2.76(s,3H)1.94(s,4H) ,1.75(s,4H).
实施例46所述化合物的制备可参考实施例45中化合物T-45的合成方法与路线。The preparation of the compound described in Example 46 can refer to the synthesis method and route of compound T-45 in Example 45.
实施例46:化合物T-46Example 46: Compound T-46
Figure PCTCN2022088343-appb-000062
Figure PCTCN2022088343-appb-000062
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.79(s,1H),8.21(s,2H),7.85(t,J=6.0Hz,2H),7.63(s,1H),7.45(s,J=9.0Hz,1H),6.93(d,J=9.0Hz,1H),4.24(s,2H),4.11(s,2H),3.75(t,J=6Hz,4H),2.56-2.51(m,1H),2.12-1.94(m,8H),1.75-1.68(m,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.79 (s, 1H), 8.21 (s, 2H), 7.85 (t, J = 6.0Hz, 2H), 7.63 (s, 1H), 7.45(s, J=9.0Hz, 1H), 6.93(d, J=9.0Hz, 1H), 4.24(s, 2H), 4.11(s, 2H), 3.75(t, J=6Hz, 4H), 2.56 -2.51(m,1H),2.12-1.94(m,8H),1.75-1.68(m,4H).
实施例47所述化合物的制备可参考实施例5中化合物T-5的合成方法与路线。The preparation of the compound described in Example 47 can refer to the synthesis method and route of compound T-5 in Example 5.
实施例47Example 47
Figure PCTCN2022088343-appb-000063
Figure PCTCN2022088343-appb-000063
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.80(s,1H),8.23(s,2H),7.84(t,J=6.0Hz,2H),7.64(s,1H),7.46(s,J=9.0Hz,1H),6.95(d,J=9.0Hz,1H),5.28(s,1H),4.67(s,1H),4.25(s,2H),4.11(s,2H),3.75-3.68(m,3H),3.52-3.49(m,2H),1.97-1.93(m,4H),1.77-1.70(m,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.80 (s, 1H), 8.23 (s, 2H), 7.84 (t, J = 6.0Hz, 2H), 7.64 (s, 1H), 7.46(s, J=9.0Hz, 1H), 6.95(d, J=9.0Hz, 1H), 5.28(s, 1H), 4.67(s, 1H), 4.25(s, 2H), 4.11(s, 2H ),3.75-3.68(m,3H),3.52-3.49(m,2H),1.97-1.93(m,4H),1.77-1.70(m,4H).
实施例48-49所述化合物的制备可参照实施例18所述的合成方法和路线。The preparation of the compounds described in Examples 48-49 can refer to the synthesis method and route described in Example 18.
实施例48:化合物T-48Example 48: Compound T-48
Figure PCTCN2022088343-appb-000064
Figure PCTCN2022088343-appb-000064
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.82(s,1H),8.24(s,2H),7.87(t,J=6.0Hz,2H),7.65(s,1H),7.46(s,J=9.0Hz,1H),6.96(d,J=9.0Hz,1H),4.26(s,2H),4.12(s,2H),3.62-3.56(m,2H),3.01-2.89(m,2H),2.14-1.93(m,6H),1.76-1.69(m,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.82 (s, 1H), 8.24 (s, 2H), 7.87 (t, J = 6.0Hz, 2H), 7.65 (s, 1H), 7.46(s, J=9.0Hz, 1H), 6.96(d, J=9.0Hz, 1H), 4.26(s, 2H), 4.12(s, 2H), 3.62-3.56(m, 2H), 3.01-2.89 (m,2H),2.14-1.93(m,6H),1.76-1.69(m,4H).
实施例49:化合物T-49Example 49: Compound T-49
Figure PCTCN2022088343-appb-000065
Figure PCTCN2022088343-appb-000065
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.84(s,1H),8.51(s,1H),8.23(s,1H),7.74(s,2H),7.54(m,2H),7.49(s,1H),6.99(s,1H),4.18(s,2H),4.02(s,2H),3.64-3.34(m,2H),3.02-2.94(m,4H),2.5-2.34(m,6H),1.92-1.83(m,4H),1.69-1.47(m,6H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.84 (s, 1H), 8.51 (s, 1H), 8.23 (s, 1H), 7.74 (s, 2H), 7.54 (m, 2H ),7.49(s,1H),6.99(s,1H),4.18(s,2H),4.02(s,2H),3.64-3.34(m,2H),3.02-2.94(m,4H),2.5- 2.34(m,6H),1.92-1.83(m,4H),1.69-1.47(m,6H).
实施例50:化合物T-50Example 50: Compound T-50
实施例50的制备方法可参照实施例25所述的化合物T-25的合成方法与路线。For the preparation method of Example 50, refer to the synthesis method and route of compound T-25 described in Example 25.
Figure PCTCN2022088343-appb-000066
Figure PCTCN2022088343-appb-000066
1H NMR(300MHz,DMSO-d 6):δ(ppm)=8.81(s,1H),8.24(s,2H),7.79(t,J=6.0Hz,2H),7.65(s,1H),7.46(s,J=9.0Hz,1H),6.96(d,J=9.0Hz,1H),4.33(s,2H),3.77(s,4H),3.69-3.63(m,2H),3.25-3.21(m,2H),3.03(s,2H),2.75(s,4H),1.94(s,4H),1.77(s,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 8.81 (s, 1H), 8.24 (s, 2H), 7.79 (t, J = 6.0Hz, 2H), 7.65 (s, 1H), 7.46(s, J=9.0Hz, 1H), 6.96(d, J=9.0Hz, 1H), 4.33(s, 2H), 3.77(s, 4H), 3.69-3.63(m, 2H), 3.25-3.21 (m,2H),3.03(s,2H),2.75(s,4H),1.94(s,4H),1.77(s,4H).
实施例51-54所述化合物的制备可参考化合物T-5的合成方法和路线。The preparation of the compounds described in Examples 51-54 can refer to the synthesis method and route of compound T-5.
实施例51:化合物T-51Example 51: Compound T-51
Figure PCTCN2022088343-appb-000067
Figure PCTCN2022088343-appb-000067
1H NMR(300MHz,DMSO-d 6):δ(ppm)=9.03(s,1H),8.56(s,1H),8.49(s,1H),8.39(s,1H),8.34(s,1H),8.23-8.19(d,J=9Hz,1H),8.16(s,1H),7.58(s,1H),4.41(s,2H),4.25(s,2H),3.91(s,4H),3.75-3.63(m,1H),2.13-1.96(m,6H),1.85-1.74(m,6H),1.54-1.40(m,4H). 1 H NMR (300MHz, DMSO-d 6 ): δ (ppm) = 9.03 (s, 1H), 8.56 (s, 1H), 8.49 (s, 1H), 8.39 (s, 1H), 8.34 (s, 1H ),8.23-8.19(d,J=9Hz,1H),8.16(s,1H),7.58(s,1H),4.41(s,2H),4.25(s,2H),3.91(s,4H), 3.75-3.63(m,1H),2.13-1.96(m,6H),1.85-1.74(m,6H),1.54-1.40(m,4H).
实施例52:化合物T-52Example 52: Compound T-52
Figure PCTCN2022088343-appb-000068
Figure PCTCN2022088343-appb-000068
1H NMR(300MHz,CDCL3):δ(ppm)=8.91(s,1H),8.52(s,1H),8.49(s,1H),8.38(s,1H),8.31(s,1H),8.22-8.19(d,J=9Hz,1H),8.12(s,1H),7.56(s,1H),4.36(s,2H),4.19(s,2H),3.97(s,4H),3.79-3.68(m,1H),2.15-1.99(m,6H),1.82-1.70(m,6H),1.53-1.41(m,6H). 1 H NMR (300MHz, CDCL3): δ (ppm) = 8.91 (s, 1H), 8.52 (s, 1H), 8.49 (s, 1H), 8.38 (s, 1H), 8.31 (s, 1H), 8.22 -8.19(d,J=9Hz,1H),8.12(s,1H),7.56(s,1H),4.36(s,2H),4.19(s,2H),3.97(s,4H),3.79-3.68 (m,1H),2.15-1.99(m,6H),1.82-1.70(m,6H),1.53-1.41(m,6H).
实施例53:化合物T-53Example 53: Compound T-53
Figure PCTCN2022088343-appb-000069
Figure PCTCN2022088343-appb-000069
1H NMR(300MHz,CDCL3):δ(ppm)=8.37(s,1H),8.22(s,1H),8.14(d,J=6.0Hz,1H),8.04(s,1H),7.88(s,1H),7.67-7.62(m,1H),7.56(s,1H),7.23-7.16(m,1H),4.38(s,2H),4.19(s,2H),3.98(s,4H),3.77-3.67(m,1H),2.10-1.98(m,6H),1.80-1.1.71(m,6H),1.51-1.40(m,6H). 1 H NMR (300MHz, CDCL3): δ (ppm) = 8.37 (s, 1H), 8.22 (s, 1H), 8.14 (d, J = 6.0Hz, 1H), 8.04 (s, 1H), 7.88 (s ,1H),7.67-7.62(m,1H),7.56(s,1H),7.23-7.16(m,1H),4.38(s,2H),4.19(s,2H),3.98(s,4H), 3.77-3.67(m,1H),2.10-1.98(m,6H),1.80-1.1.71(m,6H),1.51-1.40(m,6H).
实施例54:化合物T-54Example 54: Compound T-54
Figure PCTCN2022088343-appb-000070
Figure PCTCN2022088343-appb-000070
1H NMR(300MHz,CDCL3):δ(ppm)=8.41(s,1H),8.25(s,1H),8.16(d,J=6.0Hz,1H),8.07(s,1H),7.91(s,1H),7.69-7.62(m,1H),7.55(s,1H),7.23-7.14(m,1H),4.32(s,2H),4.19(s,2H),3.97(s,4H),3.78-3.67(m,1H),2.05-1.93(m,6H),1.86-1.1.72(m,6H),1.50-1.41(m,4H). 1 H NMR (300MHz, CDCL3): δ (ppm) = 8.41 (s, 1H), 8.25 (s, 1H), 8.16 (d, J = 6.0Hz, 1H), 8.07 (s, 1H), 7.91 (s ,1H),7.69-7.62(m,1H),7.55(s,1H),7.23-7.14(m,1H),4.32(s,2H),4.19(s,2H),3.97(s,4H), 3.78-3.67(m,1H),2.05-1.93(m,6H),1.86-1.1.72(m,6H),1.50-1.41(m,4H).
以下为此发明部分化合物的生物活性评价,主要包括细胞水平与酶水平两个方面。The following is the biological activity evaluation of some compounds of this invention, which mainly includes two aspects of cell level and enzyme level.
一、本发明部分化合物对多种癌细胞的增殖抑制活性1. Proliferation inhibitory activity of some compounds of the present invention on various cancer cells
实验方法如下:The experimental method is as follows:
以MTT法测定化合物对CDK9高表达的肿瘤细胞(MV4-11,MCF7以及MOLM13)的细胞毒性:细胞以2000个/孔接种于96孔板中,待细胞贴壁后,吸除培养基,加入200uL稀释好的药物,37度,5%CO2培养72h,之后加入10uL/孔的MTT。37度孵育4个小时,吸去上清,每孔加入150uL的DMSO,多功能酶标仪492nm处检测吸光值。用GraphPad计算IC 50并绘制细胞生长曲线。 The cytotoxicity of the compound to tumor cells with high expression of CDK9 (MV4-11, MCF7 and MOLM13) was determined by MTT method: the cells were seeded in 96-well plate at 2000 cells/well, after the cells adhered to the wall, the culture medium was aspirated and added 200uL of the diluted drug was incubated at 37°C with 5% CO2 for 72h, and then 10uL/well of MTT was added. Incubate at 37°C for 4 hours, remove the supernatant, add 150uL of DMSO to each well, and detect the absorbance at 492nm with a multi-functional microplate reader. IC50 was calculated with GraphPad and the cell growth curve was plotted.
二、本发明部分化合物对CDK9和CDK2蛋白的抑制活性2. Inhibitory activity of some compounds of the present invention on CDK9 and CDK2 proteins
实验方法如下:The experimental method is as follows:
首先将化合物用100%DMSO配制成10mM储存液于低温下避光保存。Firstly, the compound was prepared into a 10 mM stock solution with 100% DMSO and stored at low temperature in the dark.
激酶反应过程如下:The kinase reaction process is as follows:
1)配制1×Kinase buffer。1) Prepare 1×Kinase buffer.
2)化合物浓度梯度的配制:受试化合物测试浓度为5nM,20nM或100nM,复孔检测。在384孔板中配置成100倍终浓度的化合物。然后用Echo550转移250nl到384反应板中备用。阴性对照孔和阳性对照孔中分别加250nl的100%DMSO。2) Preparation of compound concentration gradient: test compound test concentration is 5nM, 20nM or 100nM, and repeated well detection. Compounds were prepared at 100-fold final concentration in a 384-well plate. Then use Echo550 to transfer 250nl to 384 reaction plate for later use. Add 250 nl of 100% DMSO to negative control wells and positive control wells respectively.
3)用1×Kinase buffer配制2.5倍终浓度的激酶溶液。3) Use 1×Kinase buffer to prepare a kinase solution with a final concentration of 2.5 times.
4)在化合物孔和阳性对照孔分别加10μl的2.5倍终浓度的激酶溶液;在阴性对照孔中加10μl的1×Kinase buffer。4) Add 10 μl of 2.5-fold final concentration of kinase solution to compound wells and positive control wells; add 10 μl of 1×Kinase buffer to negative control wells.
5)1000rpm离心30秒,振荡混匀后室温孵育10分钟。5) Centrifuge at 1000 rpm for 30 seconds, shake and mix well, and incubate at room temperature for 10 minutes.
6)用1×Kinase buffer配制25/15倍终浓度的ATP和Kinase substrate的混合溶液。6) Use 1×Kinase buffer to prepare a mixed solution of ATP and Kinase substrate at 25/15 times the final concentration.
7)加入15μl的25/15倍终浓度的ATP和底物的混合溶液,起始反应。7) Add 15 μl of a mixed solution of ATP and substrate at 25/15 times the final concentration to initiate the reaction.
8)将384孔板1000rpm离心30秒,振荡混匀后室温孵育相应的时间。8) Centrifuge the 384-well plate at 1000 rpm for 30 seconds, shake and mix well, and incubate at room temperature for a corresponding period of time.
9)加入30μl终止检测液停止激酶反应,1000rpm离心30秒,振荡混匀。9) Add 30 μl of stop detection solution to stop the kinase reaction, centrifuge at 1000 rpm for 30 seconds, shake and mix well.
10)用Caliper EZ Reader读取转化率。10) Read the conversion rate with Caliper EZ Reader.
最后进行数据计算和分析:Finally, perform data calculation and analysis:
1)抑制率计算公式:1) Inhibition rate calculation formula:
Figure PCTCN2022088343-appb-000071
Figure PCTCN2022088343-appb-000071
其中:Conversion%_sample是样品的转化率读数;Conversion%_min:阴性对照孔均值,代表没有酶活孔的转化率读数;Conversion%_max:阳性对照孔均值,代表没有化合物抑制孔的转化率读数。Among them: Conversion%_sample is the conversion rate reading of the sample; Conversion%_min: the average value of negative control wells, representing the conversion rate readings of wells without enzyme activity; Conversion%_max: the average value of positive control wells, representing the conversion rate readings of wells without compound inhibition.
2)拟合量效曲线2) Fitting the dose-effect curve
以浓度的log值作为X轴,百分比抑制率为Y轴,采用分析软件GraphPad Prism 5拟合量效曲线,从而得出各个化合物对酶活性的IC 50值。 Taking the log value of the concentration as the X-axis and the percent inhibition rate as the Y-axis, the analysis software GraphPad Prism 5 was used to fit the dose-effect curve to obtain the IC 50 value of each compound on the enzyme activity.
公式如下:Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))The formula is as follows: Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))
本发明中部分化合物对CDK9和CDK2的抑制活性和其对MV4-11,MCF-7,HCT116以及MOLM13细胞的抗增殖活性如下表1所示:The inhibitory activity of some compounds of the present invention on CDK9 and CDK2 and their antiproliferative activity on MV4-11, MCF-7, HCT116 and MOLM13 cells are shown in Table 1 below:
表1.本发明中部分化合物的酶活性和细胞活性Table 1. Enzyme activity and cell activity of some compounds in the present invention
Figure PCTCN2022088343-appb-000072
Figure PCTCN2022088343-appb-000072
Figure PCTCN2022088343-appb-000073
Figure PCTCN2022088343-appb-000073
a:抑制率一栏中,括号内表示化合物浓度,单位为nM,如90(20)表示化合物在20nM浓度时对酶的抑制率为90%。 a : In the column of inhibition rate, the concentration of the compound is indicated in the brackets, and the unit is nM, such as 90 (20) indicates that the inhibition rate of the compound to the enzyme is 90% at a concentration of 20 nM.
其中,TG-02的结构如下:Among them, the structure of TG-02 is as follows:
Figure PCTCN2022088343-appb-000074
Figure PCTCN2022088343-appb-000074
根据表1所示数据,本发明所述的化合物对CDK9显示出有效的抑制活性,大部分化合物在20nM浓度下对CDK9的抑制率在90%以上。同时,与目前唯一报道的处于临床研究阶段的大环类广谱CDK抑制剂TG02相比,本发明所述化合物显示出显著的CDK9选择性。同时,其对MV4-11,MCF-7,HCT-116以及MOLM13等多种肿瘤细胞具有显著的抗增殖抑制活性。其中,优势化合物T-9,T-10,T-52和T-53均是高选择性的CDK9抑制剂,对CDK2的选择性在50倍以上,同时均具有不错的抗肿瘤细胞增殖活性。化合物T-53对CDK9抑制IC 50值为7nM,对CDK2抑制IC 50值大于1000nM,选择性大于100倍,显著优于化合物TG02。 According to the data shown in Table 1, the compounds of the present invention exhibit effective inhibitory activity on CDK9, and most of the compounds have an inhibitory rate of more than 90% on CDK9 at a concentration of 20 nM. At the same time, compared with TG02, the only reported macrocyclic broad-spectrum CDK inhibitor currently in clinical research stage, the compound of the present invention shows remarkable CDK9 selectivity. At the same time, it has significant anti-proliferation inhibitory activity against various tumor cells such as MV4-11, MCF-7, HCT-116 and MOLM13. Among them, the dominant compounds T-9, T-10, T-52 and T-53 are all highly selective CDK9 inhibitors, with a selectivity of more than 50 times for CDK2, and they all have good anti-tumor cell proliferation activity. Compound T-53 has an inhibitory IC 50 value of 7 nM for CDK9, and an IC 50 value of greater than 1000 nM for CDK2, with a selectivity greater than 100 times, which is significantly better than that of compound TG02.

Claims (10)

  1. 一种如通式I所示结构的化合物,或其药学上可接受的盐型,其特征在于:A compound of the structure shown in general formula I, or a pharmaceutically acceptable salt thereof, is characterized in that:
    Figure PCTCN2022088343-appb-100001
    Figure PCTCN2022088343-appb-100001
    其中,Ar选自如下基团:Wherein, Ar is selected from the following groups:
    Figure PCTCN2022088343-appb-100002
    Figure PCTCN2022088343-appb-100002
    X选自氢,氘,卤素,氰基,甲基,三氟甲基;X is selected from hydrogen, deuterium, halogen, cyano, methyl, trifluoromethyl;
    R 1选自如下基团: R 1 is selected from the following groups:
    Figure PCTCN2022088343-appb-100003
    L选自如下结构:
    Figure PCTCN2022088343-appb-100003
    L is selected from the following structures:
    Figure PCTCN2022088343-appb-100004
    Figure PCTCN2022088343-appb-100004
  2. 根据权利要求1所述化合物,或其药学上可接受的盐型,其特征在于药学上可接受的盐型是指通式I所示的化合物和药学上可接受的酸形成的盐,所述的药学上可接受的酸为无机酸盐或有机酸盐;优选的,无机酸盐包括:盐酸、硫酸、磷酸、碳酸、碳酸氢根、硝酸、磷酸一氢根、磷酸二氢根,氢溴酸或氢碘酸;优选的,有机酸盐包括:马来酸、酒石酸、柠檬酸、甲磺酸、琥珀酸、乙酸、对甲苯磺酸、扁桃酸、异丁酸或丙二酸。The compound according to claim 1, or its pharmaceutically acceptable salt form, is characterized in that the pharmaceutically acceptable salt form refers to the salt formed by the compound shown in general formula I and a pharmaceutically acceptable acid, said The pharmaceutically acceptable acid is an inorganic acid salt or an organic acid salt; preferably, the inorganic acid salt includes: hydrochloric acid, sulfuric acid, phosphoric acid, carbonic acid, bicarbonate, nitric acid, monohydrogen phosphate, dihydrogen phosphate, hydrogen bromide acid or hydroiodic acid; preferably, organic acid salts include: maleic acid, tartaric acid, citric acid, methanesulfonic acid, succinic acid, acetic acid, p-toluenesulfonic acid, mandelic acid, isobutyric acid or malonic acid.
  3. 根据权利要求1所述化合物,或其药学上可接受的盐型,其特征在于Ar选自
    Figure PCTCN2022088343-appb-100005
    The compound according to claim 1, or a pharmaceutically acceptable salt thereof, is characterized in that Ar is selected from
    Figure PCTCN2022088343-appb-100005
  4. 根据权利要求1所述化合物,或其药学上可接受的盐型,其特征在于X为卤素;优选X为F、Cl、Br、I;进一步优选X为F。The compound according to claim 1, or a pharmaceutically acceptable salt thereof, is characterized in that X is a halogen; preferably X is F, Cl, Br, I; more preferably X is F.
  5. 根据权利要求1所述化合物,或其药学上可接受的盐型,其特征在于L选自:The compound according to claim 1, or a pharmaceutically acceptable salt thereof, is characterized in that L is selected from:
    Figure PCTCN2022088343-appb-100006
    Figure PCTCN2022088343-appb-100006
  6. 如下任一所述的化合物或其药学上可接受的盐:A compound or a pharmaceutically acceptable salt thereof as described in any of the following:
    Figure PCTCN2022088343-appb-100007
    Figure PCTCN2022088343-appb-100007
    Figure PCTCN2022088343-appb-100008
    Figure PCTCN2022088343-appb-100008
    Figure PCTCN2022088343-appb-100009
    Figure PCTCN2022088343-appb-100009
    Figure PCTCN2022088343-appb-100010
    Figure PCTCN2022088343-appb-100010
  7. 一种制备权利要求1所述化合物的制备方法,A preparation method for preparing the compound described in claim 1,
    Figure PCTCN2022088343-appb-100011
    Figure PCTCN2022088343-appb-100011
    其中,Ar、X、R 1、L定义如前所述。 Wherein, Ar, X, R 1 and L are as defined above.
  8. 药物组合物,其特征在于,包括权利权利要求1-6任一项所述的化合物,或其药学上可接受的盐型。The pharmaceutical composition is characterized by comprising the compound according to any one of claims 1-6, or a pharmaceutically acceptable salt thereof.
  9. 权利要求1-6任一项所述的化合物,或其药学上可接受的盐型在制备CDK9抑制剂药物中的应用。Use of the compound according to any one of claims 1-6, or a pharmaceutically acceptable salt thereof, in the preparation of a CDK9 inhibitor drug.
  10. 权利要求1-6任一项所述的化合物,或其药学上可接受的盐型在制备抗病毒药物或者抗肿瘤药物中的应用;优选的,The compound described in any one of claims 1-6, or the application of its pharmaceutically acceptable salt form in the preparation of antiviral drugs or antitumor drugs; preferably,
    所述的病毒包括:HIV病毒、巨细胞病毒、EB病毒、腺病毒、疱疹、人T细胞淋巴细胞病毒;优选的,所述的肿瘤包括神经胶质瘤、各类白血病、淋巴癌、肝癌、胃癌、前列腺癌、卵巢癌、乳腺癌、肺癌。The viruses include: HIV virus, cytomegalovirus, Epstein-Barr virus, adenovirus, herpes, human T cell lymphocyte virus; preferably, the tumors include glioma, various types of leukemia, lymphoma, liver cancer, Stomach cancer, prostate cancer, ovarian cancer, breast cancer, lung cancer.
PCT/CN2022/088343 2021-07-27 2022-04-22 Preparation method for and application of novel cdk9 inhibitor having macrocyclic skeleton structure WO2023005281A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110850793.6 2021-07-27
CN202110850793.6A CN113603708B (en) 2021-07-27 2021-07-27 Preparation and application of CDK9 inhibitor with macrocyclic skeleton structure

Publications (1)

Publication Number Publication Date
WO2023005281A1 true WO2023005281A1 (en) 2023-02-02

Family

ID=78305557

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/088343 WO2023005281A1 (en) 2021-07-27 2022-04-22 Preparation method for and application of novel cdk9 inhibitor having macrocyclic skeleton structure

Country Status (2)

Country Link
CN (1) CN113603708B (en)
WO (1) WO2023005281A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113603708B (en) * 2021-07-27 2023-08-11 中国药科大学 Preparation and application of CDK9 inhibitor with macrocyclic skeleton structure
CN114409674B (en) * 2022-01-28 2023-07-25 山东大学 Synthesis method of JAK inhibitor Pacritinib
WO2023229430A1 (en) * 2022-05-27 2023-11-30 보로노이 주식회사 Heteroaryl derivative compound and uses thereof
WO2024046443A1 (en) * 2022-09-01 2024-03-07 Nutshell Biotech (Shanghai) Co., Ltd. Macrocyclic compounds as selective cdk inhibitors

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004078682A2 (en) * 2003-03-05 2004-09-16 Irm Llc Cyclic compounds and compositions as protein kinase inhibitors
CN101365703A (en) * 2005-11-16 2009-02-11 S*Bio私人有限公司 Heteroalkyl linked pyrimidine derivatives
CN108368129A (en) * 2015-10-08 2018-08-03 拜耳医药股份有限公司 New modification macrocyclic compound
WO2018177889A1 (en) * 2017-03-28 2018-10-04 Bayer Aktiengesellschaft Novel ptefb inhibiting macrocyclic compounds
CN110028475A (en) * 2019-05-13 2019-07-19 中国药科大学 Novel C DK9 inhibitor, preparation method and application based on benzofuran structure
CN111253371A (en) * 2019-11-29 2020-06-09 中国药科大学 Small molecule regulator targeting CDK9, and synthesis method and application thereof
CN113603708A (en) * 2021-07-27 2021-11-05 中国药科大学 Preparation method and application of novel CDK9 inhibitor with macrocyclic framework structure

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008060248A1 (en) * 2006-11-15 2008-05-22 S*Bio Pte Ltd. Indole sustituted pyrimidines and use thereof in the treatment of cancer
CN102143750B (en) * 2008-09-08 2014-04-02 默克专利有限公司 Macrocyclics pyrimidines as aurora kinase inhibitors
CA2999931A1 (en) * 2015-09-29 2017-04-06 Bayer Pharma Aktiengesellschaft Novel macrocyclic sulfondiimine compounds
WO2017060322A2 (en) * 2015-10-10 2017-04-13 Bayer Pharma Aktiengesellschaft Ptefb-inhibitor-adc
EP3865488A4 (en) * 2018-12-19 2021-12-22 Keythera (Suzhou) Pharmaceuticals Co. Ltd. Macrocyclic compound as cdk inhibitor, preparation method therefor, and use thereof in medicine

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004078682A2 (en) * 2003-03-05 2004-09-16 Irm Llc Cyclic compounds and compositions as protein kinase inhibitors
CN101365703A (en) * 2005-11-16 2009-02-11 S*Bio私人有限公司 Heteroalkyl linked pyrimidine derivatives
CN108368129A (en) * 2015-10-08 2018-08-03 拜耳医药股份有限公司 New modification macrocyclic compound
WO2018177889A1 (en) * 2017-03-28 2018-10-04 Bayer Aktiengesellschaft Novel ptefb inhibiting macrocyclic compounds
CN110028475A (en) * 2019-05-13 2019-07-19 中国药科大学 Novel C DK9 inhibitor, preparation method and application based on benzofuran structure
CN111253371A (en) * 2019-11-29 2020-06-09 中国药科大学 Small molecule regulator targeting CDK9, and synthesis method and application thereof
CN113603708A (en) * 2021-07-27 2021-11-05 中国药科大学 Preparation method and application of novel CDK9 inhibitor with macrocyclic framework structure

Also Published As

Publication number Publication date
CN113603708A (en) 2021-11-05
CN113603708B (en) 2023-08-11

Similar Documents

Publication Publication Date Title
WO2023005281A1 (en) Preparation method for and application of novel cdk9 inhibitor having macrocyclic skeleton structure
EP2144889B1 (en) Quinazolin-oxime derivatives as hsp90 inhibitors
CA2484209C (en) Protein kinase modulators and methods of use
US6750341B2 (en) Preparation of risperidone
JPH06220059A (en) Condensed pyrimidine derivative and its production
CN108350006B (en) Deuterium-modified Brigatinib derivative, pharmaceutical composition containing compound and application of deuterium-modified Brigatinib derivative
CN107383004B (en) 2-amino imidazopyridine derivative and preparation and application thereof
WO2023005280A1 (en) Preparation and application of aminopyrimidine derivative selectively targeting cdk9
CN108947911B (en) Benzimidazole compound with anti-hepatitis B virus activity and antibacterial activity and synthesis method and application thereof
CN103880822A (en) 2,4,6-trisubstituted pyrimidine compounds containing 1,2,3-triazole, preparation method and application thereof
CN106146512B (en) The preparation method of Buddhist nun is replaced according to Shandong
CN108017641B (en) Pyrazolopyrimidine compound as PI3K inhibitor and application thereof
CN109251196B (en) Aminobenzo [ d ] aza-quinazoline compound and preparation method and application thereof
CN108239069B (en) Inhibitor for fibroblast growth factor receptor and application thereof
CN111606888B (en) Pyrrole derivative and preparation method and application thereof
CN111875583A (en) Triazole derivative and preparation method and application thereof
JP6427695B2 (en) Salt form, crystal form of 1,2,5-thiadiazolidine-1,1-dioxide, process for its preparation and intermediates
CN103012410B (en) [1,2,4] triazole [4,3-b] sym-tetrazine derivation compound and preparation method thereof
CN108309984B (en) Application of propionyl aminoquinazoline compound in preparation of medicine for treating cervical cancer
CN112004809A (en) 5-chloro-2, 4-pyrimidine derivatives as antitumor agents
WO2017167251A1 (en) Manufacturing process for 1-(arylmethyl) quinazoline-2,4 (1h, 3h)-dione
CN114213396B (en) Indole-2-ketone compound and preparation method and application thereof
CN112079836B (en) Triazolopyrimidine compound, salt, composition and application thereof
CN112851671B (en) 4-substituted pyrido [2,3-d ] pyrimidine-7-ketone compound and preparation method and application thereof
US11691965B2 (en) Method for preparing pyrrolidinyl urea derivative

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22847900

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE