CN118515653A - A kind of EZH2 specific inhibitor and its preparation method and use - Google Patents
A kind of EZH2 specific inhibitor and its preparation method and use Download PDFInfo
- Publication number
- CN118515653A CN118515653A CN202410986475.6A CN202410986475A CN118515653A CN 118515653 A CN118515653 A CN 118515653A CN 202410986475 A CN202410986475 A CN 202410986475A CN 118515653 A CN118515653 A CN 118515653A
- Authority
- CN
- China
- Prior art keywords
- compound
- reaction
- mmol
- mixture
- stereoisomer
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Granted
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D405/00—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
- C07D405/14—Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
- C07D471/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
- C07D471/04—Ortho-condensed systems
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Oncology (AREA)
- Hematology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Description
技术领域Technical Field
本发明涉及化学药物领域,具体涉及EZH2特异性抑制剂及其制备方法和用途。The present invention relates to the field of chemical medicines, and in particular to an EZH2 specific inhibitor and a preparation method and use thereof.
背景技术Background Art
果蝇zeste基因增强子的人类同源物2(enhancer of zeste homolog 2, EZH2),是表观遗传控制因子多梳抑制复合体2(polycomb repressive complex 2, PRC2)的核心组成部分,其通过催化S-腺苷-L-甲硫氨酸(SAM)对组蛋白H3赖氨酸K27中胺基的甲基化作用,实现对H3K27的单甲基化、双甲基化和三甲基化(H3K27me3),从而抑制抑癌基因的表达,参与调控细胞周期,细胞衰老、分化及癌症等生理或病例过程。多项研究表明,在许多实体瘤(包括乳腺癌、前列腺癌、膀胱癌、皮肤癌、肝癌、胰腺癌、肺癌、胃癌、卵巢癌)中存在EZH2的高表达,并且这种高表达与相关癌症的恶化、转移和不良预后有紧密的关系。另外, EZH2催化结构域SET中存在多个活性位点的功能获得性突变。例如,有20%以上的弥漫性大B细胞淋巴瘤和7%的滤泡性淋巴瘤中存在EZH2的酪氨酸641突变(Y641C、Y641F、Y641N、Y641S、Y641N和Y641H),这些突变使得H3K27的三甲基化水平升高,进而导致抑癌基因的沉默。除了这种依赖于EZH2的组蛋白甲基化造成抑癌基因沉默的机制外,EZH2也可以通过一种不依赖于PRC2的方式与非组蛋白底物进行甲基化来发挥功能,或者与其他因子一起构成转录复合体来激活下游靶基因的转录,提高靶基因的表达量。例如,在去势抵抗性前列腺癌细胞(CRPC)中,磷酸化的EZH2可以协助雄激素受体相关复合物激活雄激素的基因表达,从而导致去势抵抗性前列腺癌细胞的进一步发展。 这些证据表明,EZH2是一个非常有前途的治疗靶点。目前,已经有几种EZH2抑制剂,如Tazemetostat,已被FDA批准用于治疗上皮样肉瘤和滤泡性淋巴瘤;Valemetostat,已被MHLW批准用于成人T细胞淋巴瘤。然而,该类SAM竞争性抑制剂都存在各自的局限性,如对EZH1/2抑制活性不足,选择性不高、有耐药性突变,毒性大等问题。The human homolog of the Drosophila enhancer of zeste gene (EZH2) is a core component of the epigenetic control factor polycomb repressive complex 2 (PRC2). It catalyzes the methylation of the amino group in histone H3 lysine K27 by S-adenosyl-L-methionine (SAM) to achieve monomethylation, dimethylation and trimethylation of H3K27 (H3K27me3), thereby inhibiting the expression of tumor suppressor genes and participating in the regulation of cell cycle, cell senescence, differentiation and cancer and other physiological or case processes. Many studies have shown that EZH2 is highly expressed in many solid tumors (including breast cancer, prostate cancer, bladder cancer, skin cancer, liver cancer, pancreatic cancer, lung cancer, gastric cancer, and ovarian cancer), and this high expression is closely related to the deterioration, metastasis and poor prognosis of related cancers. In addition, there are multiple gain-of-function mutations in the active site of the EZH2 catalytic domain SET. For example, EZH2 tyrosine 641 mutations (Y641C, Y641F, Y641N, Y641S, Y641N, and Y641H) are present in more than 20% of diffuse large B-cell lymphomas and 7% of follicular lymphomas. These mutations increase the trimethylation level of H3K27, leading to the silencing of tumor suppressor genes. In addition to this mechanism of tumor suppressor gene silencing caused by EZH2-dependent histone methylation, EZH2 can also function by methylating non-histone substrates in a PRC2-independent manner, or form a transcription complex with other factors to activate the transcription of downstream target genes and increase the expression of target genes. For example, in castration-resistant prostate cancer cells (CRPC), phosphorylated EZH2 can assist the androgen receptor-associated complex to activate androgen gene expression, leading to the further development of castration-resistant prostate cancer cells. These evidences indicate that EZH2 is a very promising therapeutic target. Currently, there are several EZH2 inhibitors, such as Tazemetostat, which has been approved by the FDA for the treatment of epithelioid sarcoma and follicular lymphoma; Valemetostat, which has been approved by the MHLW for adult T-cell lymphoma. However, these SAM competitive inhibitors have their own limitations, such as insufficient inhibitory activity against EZH1/2, low selectivity, drug-resistant mutations, and high toxicity.
因此,需要开发一类新型的EZH2抑制剂。该抑制剂应具备更高的抑制EZH2的催化活性、更小的毒副作用,并在癌症治疗中发挥重要功能。Therefore, it is necessary to develop a new class of EZH2 inhibitors, which should have higher catalytic activity in inhibiting EZH2, less toxic side effects, and play an important role in cancer treatment.
发明内容Summary of the invention
本发明提供了抑制EZH2的化合物、其立体异构体、或式1化合物的不同立体异构体的混合物,上述化合物、立体异构体、混合物的药用盐,上述化合物、立体异构体、或式1化合物的不同立体异构体的混合物和上述化合物、立体异构体、混合物的药用盐的制备方法,以及用于治疗或预防与EZH1、EZH2和/或PRC2有关的疾病或症状的方法和药物组合物。The present invention provides compounds that inhibit EZH2, their stereoisomers, or mixtures of different stereoisomers of the compound of formula 1, pharmaceutically acceptable salts of the above compounds, stereoisomers, and mixtures, methods for preparing the above compounds, stereoisomers, or mixtures of different stereoisomers of the compound of formula 1 and pharmaceutically acceptable salts of the above compounds, stereoisomers, and mixtures, as well as methods and pharmaceutical compositions for treating or preventing diseases or symptoms associated with EZH1, EZH2 and/or PRC2.
在本发明的第一方面,本发明提供了具有式1的化合物或其立体异构体、或式1化合物的不同立体异构体的混合物、或上述化合物、立体异构体、混合物的药学上可接受的盐:In the first aspect of the present invention, the present invention provides a compound having formula 1 or a stereoisomer thereof, or a mixture of different stereoisomers of the compound of formula 1, or a pharmaceutically acceptable salt of the above compound, stereoisomer, or mixture:
其中,A1选自N或CR11;Wherein, A 1 is selected from N or CR 11 ;
A2选自N或CR12;A 2 is selected from N or CR 12 ;
A3选自N或CR13;A 3 is selected from N or CR 13 ;
R1选自H或卤素; R1 is selected from H or halogen;
R2选自H、卤素或C1-C3烷基;R 2 is selected from H, halogen or C 1 -C 3 alkyl;
R11选自H、卤素或C1-C3烷基;R 11 is selected from H, halogen or C 1 -C 3 alkyl;
R12选自H、卤素或C1-C3烷基;R 12 is selected from H, halogen or C 1 -C 3 alkyl;
R13选自H、卤素或C1-C3烷基;R 13 is selected from H, halogen or C 1 -C 3 alkyl;
L1选自-NH-或-CH2-:L 1 is selected from -NH- or -CH 2 -:
B1选自N或CH: B1 is selected from N or CH:
R3为C1-C3烷基;R 3 is C 1 -C 3 alkyl;
R4为H或C1-C3烷基; R4 is H or C1- C3 alkyl;
所述C3烷基为直链烷基、支链烷基或环烷基;The C 3 alkyl group is a straight chain alkyl group, a branched chain alkyl group or a cycloalkyl group;
C1选自N或CH; C1 is selected from N or CH;
L2选自-CHR5NH-、C(R5)2NH-、-CHR5C(=O)NH-、-C(R5)2C(=O)NH-、-CHR5C(=O)NHCH2-或- C(R5)2 C(=O)NHCH2-; L2 is selected from -CHR5NH- , C( R5 ) 2NH- , -CHR5C(=O)NH-, -C( R5 ) 2C (=O)NH- , -CHR5C(=O) NHCH2- or -C( R5 ) 2C (=O) NHCH2- ;
R5在每次出现时,各自独立地选自H、-OH、-CN、C1-C2烷基或卤素。R 5 , at each occurrence, is independently selected from H, -OH, -CN, C 1 -C 2 alkyl or halogen.
在本发明的一些实施方式中,化合物具有以下式2、式3、式4、式5、式6或式7的结构:In some embodiments of the present invention, the compound has the structure of Formula 2, Formula 3, Formula 4, Formula 5, Formula 6 or Formula 7:
其中R1为H或卤素;Wherein R 1 is H or halogen;
L1为-NH-, B1为CH;或L1为CH2, B1为N。L 1 is -NH-, B 1 is CH; or L 1 is CH 2 , B 1 is N.
在本发明的一些实施方式中,R1为卤素。In some embodiments of the present invention, R 1 is halogen.
在本发明的一些实施方式中,化合物具有以下式8或式9的结构:In some embodiments of the present invention, the compound has the structure of Formula 8 or Formula 9 below:
。 .
在本发明的一些实施方式中,式1-式9中的选自以下结构:In some embodiments of the present invention, Select from the following structures:
。 .
在本发明的一些实施方式中,R3为甲基或环丙基,R4为甲基或H。In some embodiments of the present invention, R 3 is methyl or cyclopropyl, and R 4 is methyl or H.
在本发明的一些实施方式中,R3和R4均为甲基。In some embodiments of the present invention, R 3 and R 4 are both methyl.
在本发明的一些实施方式中,R3为甲基,R4为H。In some embodiments of the present invention, R3 is methyl and R4 is H.
在本发明的一些实施方式中,R2选自H、卤素或甲基;In some embodiments of the present invention, R 2 is selected from H, halogen or methyl;
在本发明的一些实施方式中,R11选自H、卤素或甲基;In some embodiments of the present invention, R 11 is selected from H, halogen or methyl;
在本发明的一些实施方式中,R12选自H、卤素或甲基;In some embodiments of the present invention, R 12 is selected from H, halogen or methyl;
在本发明的一些实施方式中,R13选自H、卤素或甲基。In some embodiments of the present invention, R 13 is selected from H, halogen or methyl.
在本发明的一些实施方式中,L2选自-CH2NH-、-CH(OH)C(=O)NH-、-CHF2C(=O)NH-、-CH(CH3)C(=O)NH-、-CH(CN)C(=O)NH-、-CH2C(=O)NH-或- CH2C(=O)NHCH2-。In some embodiments of the invention, L2 is selected from -CH2NH- , -CH(OH)C(=O)NH-, -CHF2C(=O)NH-, -CH( CH3 )C(=O)NH-, -CH(CN ) C(=O)NH-, -CH2C(=O)NH- or -CH2C (=O) NHCH2- .
在本发明的一些实施方式中,化合物选自以下结构:In some embodiments of the invention, the compound is selected from the following structures:
。 .
本发明的第二方面,提供了一种药物组合物,包含式1 - 式9之一或化合物1-30之一的化合物、其立体异构体或可药用盐,以及至少一种可药用载剂。该药物组合物可进一步包含另外的活性药剂。In a second aspect of the present invention, a pharmaceutical composition is provided, comprising a compound of one of Formulas 1 to 9 or one of Compounds 1 to 30, a stereoisomer or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier. The pharmaceutical composition may further comprise another active agent.
在一些实施方式中,可药用载剂选自可药用媒介物和可药用辅助剂。在一些实施方案中,可药用载体选自稀释剂、溶剂、分散剂、抗氧化剂、防腐剂、缓冲剂、乳化剂、可药用填充剂、崩解剂、表面活性剂、粘合剂、调味剂、染料和润滑剂及它们的组合。In some embodiments, the pharmaceutically acceptable carrier is selected from a pharmaceutically acceptable vehicle and a pharmaceutically acceptable adjuvant. In some embodiments, the pharmaceutically acceptable carrier is selected from a diluent, a solvent, a dispersant, an antioxidant, a preservative, a buffer, an emulsifier, a pharmaceutically acceptable filler, a disintegrant, a surfactant, a binder, a flavoring agent, a dye and a lubricant and a combination thereof.
在一些实施方式中,另外的活性药剂可包括抗癌剂、生物制剂、放射类药物、激素类药物中的一种或多种。In some embodiments, the additional active pharmaceutical agents may include one or more of anticancer agents, biologics, radiopharmaceuticals, and hormonal drugs.
本发明的第三方面,提供了一种治疗由PRC2、EZH1与EZH2中至少之一介导的疾病或症状的方法,其特征在于,向需要所述治疗的受试者施用治疗有效量的式1 - 式9之一或化合物1-30之一的化合物、其立体异构体、化合物的不同立体异构体的混合物、或可药用盐,或包含式1 - 式9之一或化合物1-30之一的化合物、其立体异构体、化合物的不同立体异构体的混合物,或上述化合物、立体异构体或化合物的不同立体异构体的混合物的可药用盐的药物组合物。The third aspect of the present invention provides a method for treating a disease or symptom mediated by at least one of PRC2, EZH1 and EZH2, characterized in that a therapeutically effective amount of a compound of one of Formulas 1 to 9 or one of Compounds 1-30, a stereoisomer thereof, a mixture of different stereoisomers of the compound, or a pharmaceutically acceptable salt, or a pharmaceutical composition comprising a compound of one of Formulas 1 to 9 or one of Compounds 1-30, a stereoisomer thereof, a mixture of different stereoisomers of the compound, or a pharmaceutically acceptable salt of the above-mentioned compound, stereoisomer or mixture of different stereoisomers of the compound is administered to a subject in need of the treatment.
在一些实施方式中,由PRC2、EZH1与EZH2中至少之一介导的疾病为癌症,至少包括乳腺癌(例如,三阴性乳腺癌)、胶质母细胞瘤、前列腺癌、子宫癌、卵巢癌、胰腺癌、黑色素瘤、肾细胞癌、膀胱癌、结肠直肠癌、淋巴瘤、白血病、恶性横纹肌样瘤、或口咽癌等。In some embodiments, the disease mediated by at least one of PRC2, EZH1 and EZH2 is cancer, including at least breast cancer (e.g., triple-negative breast cancer), glioblastoma, prostate cancer, uterine cancer, ovarian cancer, pancreatic cancer, melanoma, renal cell carcinoma, bladder cancer, colorectal cancer, lymphoma, leukemia, malignant rhabdoid tumor, or oropharyngeal cancer, etc.
本发明的第四方面,提供了一种式1 - 式9之一或化合物1-30之一的化合物、其立体异构体、化合物的不同立体异构体的混合物或上述化合物、立体异构体或化合物的不同立体异构体的混合物的可药用盐,或含有其的药物组合物,用于制备治疗由PRC2、EZH1与EZH2中至少之一介导的疾病或症状的药物中的用途。In a fourth aspect of the present invention, a compound of one of Formulas 1 to 9 or one of Compounds 1 to 30, a stereoisomer thereof, a mixture of different stereoisomers of the compound, or a pharmaceutically acceptable salt of the above compound, stereoisomer or mixture of different stereoisomers of the compound, or a pharmaceutical composition containing the same is provided for use in the preparation of a medicament for treating a disease or symptom mediated by at least one of PRC2, EZH1 and EZH2.
本发明的第五方面,提供了一种式1 - 式9之一或化合物1-30之一的化合物及其立体异构体的制备方法。In a fifth aspect of the present invention, a method for preparing a compound of one of Formulas 1 to 9 or one of Compounds 1 to 30 and a stereoisomer thereof is provided.
本发明的化合物可以使用下述方法或通过有机合成领域的技术人员所理解的其他类似方法来制备。具有手性中心的式1 - 式9之一或化合物1-30之一的化合物能够通过使用基本上光学纯的起始原料或通过分离色谱法、重结晶法或本领域熟知的其他分离技术来制备成基本上光学纯的形式。The compounds of the present invention can be prepared using the following methods or other similar methods understood by those skilled in the art of organic synthesis. Compounds of one of Formulas 1 to 9 or one of Compounds 1-30 having a chiral center can be prepared in substantially optically pure form by using substantially optically pure starting materials or by separation chromatography, recrystallization, or other separation techniques well known in the art.
通过起始原料Boc保护氨基的对位取代酮和光学纯的叔丁基亚磺酰胺经过脱水缩合得到叔丁基亚磺酰亚胺中间体1(INT-1),INT-1再经过还原反应得到叔丁基亚磺酰胺中间体2(INT-2),INT-2 再选择性脱去叔丁基亚磺酰基得中间体3;或在R4需要进一步的烷基取代时,继续氨基单烷基化R4取代得中间体3(INT-3),INT3再通过脱去Boc保护剂后直接与杂环芳基卤代物经亲电芳基取代反应或者金属催化的偶联反应,得到对应中间体INT-4,INT-4与含氮杂双环芳基卤代物进行金属催化的偶联反应或亲电芳基取代反应,或者与氮杂双环芳基醛进行还原胺化反应获得中间体INT-5,INT-5水解M部分的酯基形成自由的羧酸后再与对应的氧杂环丁烷-3-胺或氧杂环丁烷-3-基甲胺进行酸胺缩合反应或M部分的酯基进行还原得到醛基后进行还原胺化反应得到式1化合物。The starting material Boc-protected amino para-substituted ketone and optically pure tert-butylsulfenamide are subjected to dehydration condensation to obtain tert-butylsulfenyl imide intermediate 1 (INT-1), INT-1 is then subjected to reduction reaction to obtain tert-butylsulfenamide intermediate 2 (INT-2), and INT-2 is then selectively de-tert-butylsulfenyl to obtain intermediate 3; or when R 4 needs further alkyl substitution, the amino group is further monoalkylated to R 4 is substituted to obtain intermediate 3 (INT-3), and INT3 is directly subjected to electrophilic aromatic substitution reaction or metal-catalyzed coupling reaction with heterocyclic aromatic halides after removing the Boc protecting agent to obtain the corresponding intermediate INT-4, INT-4 is subjected to metal-catalyzed coupling reaction or electrophilic aromatic substitution reaction with nitrogen-containing heterobicyclic aromatic halides, or is subjected to reductive amination reaction with nitrogen-containing heterobicyclic aromatic aldehydes to obtain intermediate INT-5, and INT-5 is subjected to acid-amine condensation reaction with corresponding oxetane-3-amine or oxetane-3-ylmethylamine after the ester group of the M portion is hydrolyzed to form a free carboxylic acid, or the ester group of the M portion is reduced to obtain an aldehyde group and then subjected to reductive amination reaction to obtain the compound of formula 1.
本发明的化合物作为有效的EZH2抑制剂,具有更强的抑制活性和合适的药物特性,可用于治疗由EZH1、EZH2和/或PRC2介导的疾病和/或疾病的症状。The compounds of the present invention are effective EZH2 inhibitors, have stronger inhibitory activity and suitable pharmaceutical properties, and can be used to treat diseases and/or symptoms of diseases mediated by EZH1, EZH2 and/or PRC2.
附图说明BRIEF DESCRIPTION OF THE DRAWINGS
图1 化合物8、对比例2(CN114746414A中的C36)与靶点蛋白分子对接结果图Figure 1 Docking results of compound 8, comparative example 2 (C36 in CN114746414A) and target protein molecules
具体实施方式DETAILED DESCRIPTION
术语定义Definition of terms
本发明中的术语“一种”或“一个”含义包括“至少一个”或“至少一种”,进而名词的含义包括单数和复数形式。例如,“另一种药剂”意为一种、两种或更多种的药剂。The term "a" or "an" in the present invention includes "at least one" or "at least one", and the meaning of the noun includes singular and plural forms. For example, "another agent" means one, two or more agents.
本发明中的术语“PRC2”为表观遗传控制因子多梳抑制复合体2(polycombrepressive complex 2)。The term "PRC2" in the present invention refers to the epigenetic control factor polycombrepressive complex 2.
本发明中的术语“EZH2”为果蝇zeste基因增强子的人类同源物2,PRC2的核心组成部分,通过对组蛋白H3赖氨酸27引入3甲基化(H3K27me3),来调控靶基因的转录,进而调节其表达、分化和发育。The term "EZH2" in the present invention is the human homolog 2 of the Drosophila enhancer of zeste gene, a core component of PRC2, which regulates the transcription of target genes by introducing 3 methylation (H3K27me3) on histone H3 lysine 27, thereby regulating their expression, differentiation and development.
本发明中的术语“受试者”指包括人的动物。The term "subject" in the present invention refers to animals including humans.
本发明中的术语“治疗有效量”指化合物产生其施用的预期效果(例如,由EZH1、EZH2和/或PRC2介导的疾病和/或疾病症状的改善,减轻相应疾病和/或疾病症状的严重性,和/或减慢相应疾病和/或症状的进展)的量。治疗有效量的精确量将取决于治疗目的,并且本领域技术人员可以使用已知技术确定(参见例如Lloyd (1999), The Art, Science andTechnology of Pharmaceutical Compounding)。The term "therapeutically effective amount" in the present invention refers to the amount of a compound that produces the desired effect of its administration (e.g., improvement of a disease and/or disease symptom mediated by EZH1, EZH2 and/or PRC2, reduction of severity of a corresponding disease and/or disease symptom, and/or slowing down the progression of a corresponding disease and/or symptom). The exact amount of a therapeutically effective amount will depend on the purpose of the treatment, and can be determined by a person skilled in the art using known techniques (see, e.g., Lloyd (1999), The Art, Science and Technology of Pharmaceutical Compounding).
如本文使用的术语“治疗”及其同源词指减缓或停止疾病进展。如本文使用的“治疗”及其同源词包括但不限于以下:完全或部分缓解,治愈由EZH1、EZH2和/或PRC2介导的疾病和/或症状,降低相应疾病和/或疾病的症状的风险。这些症状中的任何一种的改善或严重性的减轻可以根据本领域已知的方法和技术进行评价。As used herein, the term "treat" and its cognates refer to slowing or stopping the progression of a disease. As used herein, "treat" and its cognates include, but are not limited to, the following: complete or partial relief, cure of diseases and/or symptoms mediated by EZH1, EZH2 and/or PRC2, reduction in the risk of corresponding diseases and/or symptoms of diseases. The improvement of any of these symptoms or reduction in severity can be evaluated according to methods and techniques known in the art.
本发明中的术语“化合物”,指代本发明的化合物时,表示具有相同化学结构的分子的集合,除非另有指明为立体异构体的集合(例如,外消旋物的集合、顺式/反式立体异构体的集合、或(E)和(Z)立体异构体的集合),但不排除组成分子的原子所包含的同位素体。因此,本领域技术人员应当清楚,当表示化合物的特定化学结构中包含氘原子时,该化合物也包含少量的一个或多个氘原子位点实为氢原子的同位素体。本发明的化合物中,这类同位素体的相对量将取决于许多因素,包括,例如:用于制备化合物的试剂的同位素纯度、用于制备化合物的各种合成步骤中同位素掺入的效率。然而,如上文所述,这样的同位素体的相对量总计将小于化合物的49.9%。在其它实施方案中,这样的同位素体的相对量总计将小于化合物的47.5%、小于40%、小于32.5%、小于25%、小于17.5%、小于10%、小于5%、小于3%、小于1%或小于0.5%。The term "compound" in the present invention, when referring to the compounds of the present invention, means a collection of molecules having the same chemical structure, unless otherwise specified as a collection of stereoisomers (e.g., a collection of racemates, a collection of cis/trans stereoisomers, or a collection of (E) and (Z) stereoisomers), but does not exclude isotopologues contained in the atoms that make up the molecules. Therefore, it should be clear to those skilled in the art that when a deuterium atom is contained in a specific chemical structure of a compound, the compound also contains a small amount of isotopologues in which one or more deuterium atom sites are actually hydrogen atoms. In the compounds of the present invention, the relative amount of such isotopologues will depend on many factors, including, for example: the isotopic purity of the reagents used to prepare the compounds, the efficiency of isotope incorporation in the various synthetic steps used to prepare the compounds. However, as described above, the relative amount of such isotopologues will be less than 49.9% of the compound in total. In other embodiments, the relative amounts of such isotopologues will total less than 47.5%, less than 40%, less than 32.5%, less than 25%, less than 17.5%, less than 10%, less than 5%, less than 3%, less than 1%, or less than 0.5% of the compound.
本发明中的术语“立体异构体”,包括顺反异构体、对映异构体和非对映异构体。The term "stereoisomer" in the present invention includes cis-trans isomers, enantiomers and diastereomers.
本发明中的术语“可药用盐”或“药学上可接受的盐”是在酸与化合物的碱性基团如氨基官能团之间形成的,或者在碱与化合物的酸性基团如羧基官能团之间形成的。The term "pharmaceutically acceptable salt" or "pharmaceutically acceptable salt" in the present invention is formed between an acid and a basic group of a compound, such as an amino functional group, or between a base and an acidic group of a compound, such as a carboxyl functional group.
术语“可药用的”指在合理的医学判断范围之内,适合用于与人和其它哺乳动物的组织接触而没有过度的毒性、刺激、过敏反应等,并且与合理的效益/风险比相称的组分。The term "pharmaceutically acceptable" refers to a composition that is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and other mammals without excessive toxicity, irritation, allergic response, etc., and is commensurate with a reasonable benefit/risk ratio.
术语 “可药用盐” 或“药学上可接受的盐”指任何无毒的盐,其在施用于接受者后能够直接或间接地提供本发明内容的化合物。合适的可药用盐是例如在S.M.Berge等人在J.PharmaceuticalSciences、1977,66,第1至19页中公开的那些。The term "pharmaceutically acceptable salt" or "pharmaceutically acceptable salt" refers to any non-toxic salt that can directly or indirectly provide the compounds of the present invention after being administered to a recipient. Suitable pharmaceutically acceptable salts are those disclosed, for example, by S. M. Berge et al. in J. Pharmaceutical Sciences, 1977, 66, pp. 1 to 19.
通常用于形成可药用盐的酸包括无机酸,比如二硫化氢、盐酸、氢溴酸、氢碘酸、硫酸和磷酸,以及有机酸,比如对甲苯磺酸、水杨酸、酒石酸、酸式酒石酸(bitartaric acid)、抗坏血酸、马来酸、苯磺酸(besylic acid)、富马酸、葡糖酸、葡糖醛酸、甲酸、谷氨酸、甲磺酸、乙磺酸、苯磺酸、乳酸、草酸、对溴苯磺酸、碳酸、琥珀酸、柠檬酸、苯甲酸和乙酸,以及相关无机酸和有机酸。因此,这样的可药用盐包括硫酸盐、焦硫酸盐、硫酸氢盐、亚硫酸盐、亚硫酸氢盐、磷酸盐、磷酸一氢盐、磷酸二氢盐、偏磷酸盐、焦磷酸盐、氯化物、溴化物、碘化物、乙酸盐、丙酸盐、癸酸盐、辛酸盐、丙烯酸盐、甲酸盐、异丁酸盐、癸酸盐、庚酸盐、丙炔酸盐、草酸盐、丙二酸盐、琥珀酸盐、辛二酸盐、癸二酸盐、富马酸盐、马来酸盐、丁炔-1、4-二酸盐、己炔-l、6-二酸盐、苯甲酸盐、氯苯甲酸盐、甲基苯甲酸盐、二硝基苯甲酸盐、羟基苯甲酸盐、甲氧基苯甲酸盐、邻苯二甲酸盐、对苯二甲酸盐、磺酸盐、二甲苯磺酸盐、苯乙酸盐、苯丙酸盐、苯丁酸盐、柠檬酸盐、乳酸盐、β-羟基丁酸盐、羟乙酸盐、马来酸盐、酒石酸盐、甲磺酸盐、丙磺酸盐、萘-1-磺酸盐、萘-2-磺酸盐、扁桃酸盐和其它盐。在一些实施方案中,可药用酸加成盐包括与无机酸如盐酸和氢溴酸形成的盐,以及与有机酸如马来酸形成的盐。Acids commonly used to form pharmaceutically acceptable salts include inorganic acids such as hydrogen disulfide, hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, and phosphoric acid, and organic acids such as p-toluenesulfonic acid, salicylic acid, tartaric acid, bitartaric acid, ascorbic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucuronic acid, formic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, lactic acid, oxalic acid, p-bromobenzenesulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, and acetic acid, as well as related inorganic and organic acids. Thus, such pharmaceutically acceptable salts include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, octanoates, acrylates, formates, isobutyrates, decanoates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-1,4-dioic acid, In some embodiments, pharmaceutically acceptable acid addition salts include salts formed with inorganic acids such as hydrochloric acid and hydrobromic acid, and salts formed with organic acids such as maleic acid.
衍生自合适碱的可药用盐包括碱金属、碱土金属、铵和N+(C1-4烷基)4盐。本发明内容还预期了本发明所公开的化合物的任何碱性含氮基团的季铵化。碱金属和碱土金属盐的合适的非限制性实例包括钠盐、锂盐、钾盐、钙盐和镁盐。可药用盐的进一步非限制性实例包括使用抗衡离子比如卤离子、氢氧根、羧酸根、硫酸根、磷酸根、硝酸根、低级烷基磺酸根和芳基磺酸根形成的铵、季铵和胺阳离子。可药用盐的其它合适的非限制性实例包括苯磺酸盐和葡糖胺盐。Pharmaceutically acceptable salts derived from suitable bases include alkali metals, alkaline earth metals, ammonium and N + (C 1-4 alkyl) 4 salts. The present invention also contemplates the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Suitable non-limiting examples of alkali metal and alkaline earth metal salts include sodium salts, lithium salts, potassium salts, calcium salts and magnesium salts. Further non-limiting examples of pharmaceutically acceptable salts include ammonium, quaternary ammonium and amine cations formed using counterions such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, lower alkyl sulfonates and aryl sulfonates. Other suitable non-limiting examples of pharmaceutically acceptable salts include benzenesulfonates and glucosamine salts.
本发明中所述的活性药物,可包括抗癌剂、生物制剂、激素类药物及放射类药物的中的一种或多种:The active drugs described in the present invention may include one or more of anticancer agents, biological agents, hormone drugs and radioactive drugs:
抗癌剂,如阿扎胞昔(Azacitadine)、Doxil、全反式视黄酸、Blenoxane、Xeloda、5-FU、Ifex、Gleevec、Ellence、Targretin、CeeNU、Platinol、Valstar、Clolar、6-巯基嘌呤、Ara-C、Eloxatin、Leukeran、Panretin、Gemzar、Novantrone、Abraxane、2CdA、Emcyt、尼洛替尼(nilotinib)、L-PAM、DaunoXome、Paraplatin、六甲三聚氰胺、丝裂霉素C、Oncaspar、Hycamtin、硫酸醛基长春碱、Fludara、6-TG、盐酸柔红霉素、Nipent、Camptosar、Neutrexin、DIC、Accutane、Tespa、Vidaza、Temodar、Matulane、Cytoxan、Adriamycin、光辉霉素(mithracin)、Alimta、Lysodren、Ixempra、磷酸依托泊苷、Velban、Trisenox、伊沙匹隆(ixabepilone)、放线菌素D、Sandostatin、VM 26、Zanosar、Dacogen、氮芥、FUDR、LiposomalAra-C、Idamycin、Taxotere、左旋天门冬酰胺酶(L-asparaginase)、Hydrea、BCNU、有卡氯芥植入的prolifeprospan20、培唑帕尼(Pazopanib)、索拉非尼(Sorafenib)、厄洛替尼(Erlotinib)、奥希替尼(Osimertinib)。Anticancer agents such as Azacitadine, Doxil, all-trans retinoic acid, Blenoxane, Xeloda, 5-FU, Ifex, Gleevec, Ellence, Targretin, CeeNU, Platinol, Valstar, Clolar, 6-mercaptopurine, Ara-C, Eloxatin, Leukeran, Panretin, Gemzar, Novantrone, Abraxane, 2CdA, Emcyt, nilotinib, L-PAM, DaunoXome, Paraplatin, Hexamethasone Cyanamide, mitomycin C, Oncaspar, Hycamtin, vinblastine sulfate, Fludara, 6-TG, daunorubicin hydrochloride, Nipent, Camptosar, Neutrexin, DIC, Accutane, Tespa, Vidaza, Temodar, Matulane, Cytoxan, Adriamycin, mithracin, Alimta, Lysodren, Ixempra, etoposide phosphate, Velban, Trisenox, ixabepilone, actinomycin D, Sandostatin, VM 26. Zanosar, Dacogen, Nitrogen Mustard, FUDR, Liposomal Ara-C, Idamycin, Taxotere, L-asparaginase, Hydrea, BCNU, Prolife Prospan 20 with carbamazepine implant, Pazopanib, Sorafenib, Erlotinib, Osimertinib.
生物制剂,如干扰素、Herceptin、来那度胺(lenalidomide)、Tarceva、Velcade、卡介苗、Iressa、Revlimid、Zevalin、贝伐珠单抗(Bevacizumab)、派姆单抗(Pembrolizumab)、尼妥珠单抗(Nimotuzumab)、Bexxar、白细胞介素-2、Ontak、Campath、Rituxan、TrastuzumabDeruxtecan、厄洛替尼(Erlotinib)、Mylotarg、Abraxane、Ergamisol、Tykerb、Pegasys、Thalomid。Biologics, such as interferon, Herceptin, lenalidomide, Tarceva, Velcade, BCG, Iressa, Revlimid, Zevalin, Bevacizumab, Pembrolizumab, Nimotuzumab, Bexxar, Interleukin-2, Ontak, Campath, Rituxan, TrastuzumabDeruxtecan, Erlotinib, Mylotarg, Abraxane, Ergamisol, Tykerb, Pegasys, Thalomid.
激素类药物,如Arimidex、Cytadren、Evista、Delta-Cortef、Eligard、Faslodex、Femara、Halotestin、Megace、Nilandron、Nolvadex、Plenaxis、Zoladex、地塞美松磷酸钠、DeltaSone、甲羟孕酮、醋酸亮丙瑞林、氟维司群、依西美坦。Hormonal medicines such as Arimidex, Cytadren, Evista, Delta-Cortef, Eligard, Faslodex, Femara, Halotestin, Megace, Nilandron, Nolvadex, Plenaxis, Zoladex, dexamethasone sodium phosphate, DeltaSone, medroxyprogesterone, leuprolide acetate, fulvestrant, and exemestane.
放射类药物,如Phosphocol、Iodotope、钐SM-153、Metastron。Radioactive drugs, such as Phosphocol, Iodotope, Samarium SM-153, Metastron.
本发明中的稀释剂、溶剂、分散剂、抗氧化剂、防腐剂、缓冲剂、乳化剂、可药用填充剂、崩解剂、表面活性剂、粘合剂、调味剂、染料和润滑剂具有它们在本领域公知的含义。The diluent, solvent, dispersant, antioxidant, preservative, buffer, emulsifier, pharmaceutically acceptable filler, disintegrant, surfactant, binder, flavoring, dye and lubricant in the present invention have their well-known meanings in the art.
本发明中的术语“卤素”,包括F、Cl、Br和I,即分别为氟、氯、溴和碘。在某些实施方式中,卤素优选为F、Cl、Br。The term "halogen" in the present invention includes F, Cl, Br and I, i.e., fluorine, chlorine, bromine and iodine, respectively. In certain embodiments, halogen is preferably F, Cl, Br.
本发明中的术语“烷基”指完全饱和的支链的、支链的或成环的,取代或未取代的烃链。例如,“C1-C3烷基”是包括1至3个碳原子的烷基,即C1、C2或C3,分别为甲基、乙基、丙基、异丙基或环丙基。在一些实施方案中,烷基基团是被取代的。The term "alkyl" in the present invention refers to a fully saturated branched, branched or cyclic, substituted or unsubstituted hydrocarbon chain. For example, "C 1 -C 3 alkyl" is an alkyl group comprising 1 to 3 carbon atoms, i.e., C 1 , C 2 or C 3 , methyl, ethyl, propyl, isopropyl or cyclopropyl, respectively. In some embodiments, the alkyl group is substituted.
本发明中的术语“直链烷基”指没有侧链或分支的烷基。The term "straight chain alkyl group" in the present invention refers to an alkyl group having no side chain or branch.
本发明中的术语“支链烷基”指烷基主链上的一个或多个碳原子连接侧链的烷基。The term "branched alkyl group" in the present invention refers to an alkyl group in which one or more carbon atoms on the main chain of the alkyl group are connected to a side chain.
本发明中的术语“环烷基”指碳原子通过单键形成闭合的环状的烷基。The term "cycloalkyl group" in the present invention refers to an alkyl group in which carbon atoms form a closed ring through single bonds.
本发明中的术语“被取代的”是指基团中的一个或多个H被其他原子或基团取代,例如“被1-3个卤素取代的C1-C3烷基”即C1-C3烷基中的1-3个H被卤素取代。The term "substituted" in the present invention means that one or more H in the group is replaced by other atoms or groups, for example, "C 1 -C 3 alkyl substituted by 1-3 halogens" means that 1-3 H in the C 1 -C 3 alkyl is replaced by halogens.
本发明中的术语“氰基”、“腈”意为“-CN”基团。The terms "cyano" and "nitrile" in the present invention mean a "-CN" group.
本发明中的术语“羟基”意为“-OH”基团。The term "hydroxyl" in the present invention means a "-OH" group.
本发明中的癌或肿瘤,包括弥漫性大B细胞淋巴瘤(DLBCL)、滤泡性淋巴瘤、白血病、多发性骨髓瘤、胃癌、恶性横纹肌样瘤、肝细胞癌、前列腺癌、乳腺癌、胆管和胆囊癌、膀胱癌、神经母细胞瘤、胶质瘤、胶质母细胞瘤和星形细胞瘤、宫颈癌、结肠癌、黑色素瘤、子宫内膜癌、食管癌、头颈癌、肺癌、鼻咽癌、卵巢癌、胰腺癌、肾细胞癌、直肠癌、甲状腺癌、甲状旁腺肿瘤、子宫肿瘤、横纹肌肉瘤、卡波西肉瘤、滑膜肉瘤、骨肉瘤和尤因氏肉瘤(Ewing’ssarcoma)。The cancer or tumor in the present invention includes diffuse large B-cell lymphoma (DLBCL), follicular lymphoma, leukemia, multiple myeloma, gastric cancer, malignant rhabdoid tumor, hepatocellular carcinoma, prostate cancer, breast cancer, bile duct and gallbladder cancer, bladder cancer, neuroblastoma, glioma, glioblastoma and astrocytoma, cervical cancer, colon cancer, melanoma, endometrial cancer, esophageal cancer, head and neck cancer, lung cancer, nasopharyngeal carcinoma, ovarian cancer, pancreatic cancer, renal cell carcinoma, rectal cancer, thyroid cancer, parathyroid tumors, uterine tumors, rhabdomyosarcoma, Kaposi's sarcoma, synovial sarcoma, osteosarcoma and Ewing's sarcoma.
乳腺肿瘤包括例如具有阳性激素受体状态的乳腺癌、具有负性激素受体状态的乳腺癌、Her-2阳性乳腺癌、激素受体和Her-2阴性乳腺癌、BRCA相关乳腺癌和炎性乳腺癌。呼吸道肿瘤包括例如非小细胞支气管癌和小细胞支气管癌、非小细胞肺癌和小细胞肺癌。Breast tumors include, for example, breast cancer with positive hormone receptor status, breast cancer with negative hormone receptor status, Her-2 positive breast cancer, hormone receptor and Her-2 negative breast cancer, BRCA-associated breast cancer, and inflammatory breast cancer. Respiratory tract tumors include, for example, non-small cell bronchial carcinoma and small cell bronchial carcinoma, non-small cell lung cancer, and small cell lung cancer.
脑肿瘤包括例如神经胶质瘤、成胶质细胞瘤、星形细胞瘤、脑膜瘤和成神经管细胞瘤。男性生殖器官肿瘤包括例如前列腺癌、恶性附睾肿瘤、恶性睾丸肿瘤和阴茎癌。Brain tumors include, for example, gliomas, glioblastomas, astrocytomas, meningiomas, and medulloblastomas. Male reproductive organ tumors include, for example, prostate cancer, malignant epididymal tumors, malignant testicular tumors, and penile cancer.
女性生殖器官的肿瘤包括例如子宫内膜癌、宫颈癌、卵巢癌、阴道癌和外阴癌。Tumors of the female reproductive organs include, for example, endometrial cancer, cervical cancer, ovarian cancer, vaginal cancer, and vulvar cancer.
胃肠道肿瘤包括例如结肠直肠癌、肛门癌、胃癌、胰腺癌、食道癌、胆囊癌、小肠癌、唾液腺癌、神经内分泌肿瘤和胃肠道间质瘤。Gastrointestinal tumors include, for example, colorectal cancer, anal cancer, gastric cancer, pancreatic cancer, esophageal cancer, gallbladder cancer, small intestine cancer, salivary gland cancer, neuroendocrine tumors, and gastrointestinal stromal tumors.
泌尿生殖道肿瘤包括例如膀胱癌、肾细胞癌以及肾盂和泌尿道癌。眼部肿瘤包括例如视网膜母细胞瘤和眼内黑素瘤。Genitourinary tract tumors include, for example, bladder cancer, renal cell carcinoma, and cancer of the renal pelvis and urinary tract. Ocular tumors include, for example, retinoblastoma and intraocular melanoma.
肝脏肿瘤包括例如肝细胞癌和胆管细胞癌。Liver tumors include, for example, hepatocellular carcinoma and cholangiocarcinoma.
皮肤肿瘤包括例如恶性黑素瘤、基底瘤、脊髓瘤、卡波西肉瘤和默克尔细胞癌。Skin tumors include, for example, malignant melanoma, basal carcinoma, myeloma, Kaposi's sarcoma, and Merkel cell carcinoma.
头部和颈部肿瘤包括例如喉癌以及咽癌和口腔癌。Head and neck tumors include, for example, laryngeal cancer as well as cancers of the pharynx and oral cavity.
肉瘤包括例如软组织肉瘤、滑膜肉瘤、横纹肌样肉瘤和骨肉瘤。淋巴瘤包括例如非霍奇金淋巴瘤、霍奇金淋巴瘤、皮肤淋巴瘤、中枢神经系统淋巴瘤和AIDS相关淋巴瘤。Sarcomas include, for example, soft tissue sarcomas, synovial sarcomas, rhabdoid sarcomas, and osteosarcomas. Lymphomas include, for example, non-Hodgkin's lymphoma, Hodgkin's lymphoma, cutaneous lymphoma, central nervous system lymphoma, and AIDS-related lymphoma.
膀胱癌、脑癌、乳腺癌、结肠直肠癌、慢性粒单核细胞白血病、MLL重排白血病、肺腺癌、淋巴瘤、髓母细胞瘤、黑素瘤、多发性癌症、 骨髓瘤、前列腺癌、恶性横纹肌样瘤、滑膜肉瘤、畸胎瘤/横纹肌样瘤或 T 细胞急性淋巴细胞白血病。Bladder cancer, brain cancer, breast cancer, colorectal cancer, chronic myelomonocytic leukemia, MLL-rearranged leukemia, lung adenocarcinoma, lymphoma, medulloblastoma, melanoma, multiple carcinomas, myeloma, prostate cancer, malignant rhabdoid tumor, synovial sarcoma, teratoma/rhabdoid tumor, or T-cell acute lymphoblastic leukemia.
本发明的缩略词具有本领域通常的含义或以下表1的定义:The abbreviations of the present invention have the common meanings in the art or the definitions in the following Table 1:
表1 缩略词表Table 1 Abbreviations
用于制备本发明的化合物的方法Process for preparing the compounds of the present invention
本发明的化合物可以使用实施例1-27的方法,或本领域技术人员理解的其他类似方法制备。The compounds of the present invention can be prepared using the methods of Examples 1-27, or other similar methods understood by those skilled in the art.
实施例1:(S)-2-(2-((-1-(-4-((8-氯-7-甲基喹啉-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺(化合物1)的合成Example 1: Synthesis of (S)-2-(2-((-1-(-4-((8-chloro-7-methylquinolin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 1)
1.关键起始物料(S)-(4-(1-氨基乙基)环己基)氨基甲酸叔丁酯(1-3)的制备:1. Preparation of key starting material (S)-tert-butyl (4-(1-aminoethyl)cyclohexyl)carbamate (1-3):
步骤A 中间体1-1的合成Step A Synthesis of Intermediate 1-1
向带搅拌子的反应瓶中依次加入(反式-4-乙酰环己基)氨基甲酸叔丁酯 (4.82g,20mmol, 1.0当量), (S)-叔丁基亚磺酰胺(2.91g, 24 mmol, 1.2当量),置换氮气三次,加入四氢呋喃(100mL),钛酸四乙酯(6.84g, 30mmol, 1.5当量),加热回流20h,点板监测。反应结束后,冷却至室温,在剧烈搅拌下,把反应液倒入饱和碳酸氢钠(100mL)的容器中,过滤,乙酸乙酯(100mL)洗涤滤饼, 合并有机相,减压旋蒸除去溶剂,获得化合物中间体1-1,直接进行下一步。To a reaction flask with a stirrer, tert-butyl (trans-4-acetylcyclohexyl)carbamate (4.82 g, 20 mmol, 1.0 equivalent), (S)-tert-butylsulfenamide (2.91 g, 24 mmol, 1.2 equivalent) were added in sequence, nitrogen was replaced three times, tetrahydrofuran (100 mL), tetraethyl titanate (6.84 g, 30 mmol, 1.5 equivalent) were added, and the mixture was heated to reflux for 20 h, and the plate was monitored. After the reaction was completed, the mixture was cooled to room temperature, and the reaction solution was poured into a container of saturated sodium bicarbonate (100 mL) under vigorous stirring, filtered, and the filter cake was washed with ethyl acetate (100 mL). The organic phases were combined, and the solvent was removed by vacuum rotary evaporation to obtain compound intermediate 1-1, which was directly carried out to the next step.
步骤B 中间体1-2的合成Step B Synthesis of Intermediate 1-2
将上述油状物1-1溶于四氢呋喃(50mL), 搅拌下加入硼氢化钠(1.51g, 40mmol,2.0当量), 室温下搅拌20h,点板监测。反应结束后,加水(40mL)淬灭。乙酸乙酯(10.0 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体1-2(4.29 g,12.4 mmol,产率62%),白色固体。1H NMR (500 MHz, CD3OD) δ 4.60 (brs, 1H), 3.25-3.12 (m, 1H), 1.96-1.78 (m,4H),1.43 (s, 9H), 1.23 (s, 9H), 1.22-1.13 (m, 7H). LCMS [M+H]+ m/z: 347.2。The above oily substance 1-1 was dissolved in tetrahydrofuran (50 mL), sodium borohydride (1.51 g, 40 mmol, 2.0 equivalents) was added under stirring, and stirred at room temperature for 20 h, and the reaction was monitored by spot plate. After the reaction was completed, water (40 mL) was added to quench. The mixed solution was extracted three times with ethyl acetate (10.0 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 1-2 (4.29 g, 12.4 mmol, yield 62%) as a white solid. 1 H NMR (500 MHz, CD 3 OD) δ 4.60 (brs, 1H), 3.25-3.12 (m, 1H), 1.96-1.78 (m,4H), 1.43 (s, 9H), 1.23 (s, 9H), 1.22-1.13 (m, 7H). LCMS [M+H] + m/ z: 347.2.
步骤C 关键起始物料1-3的合成Step C Synthesis of key starting materials 1-3
将上述固体1-2(4.29 g,12.4 mmol,1.0当量),溶于四氢呋喃(30 mL)和水(6mL),搅拌下加入碘(2.20g,8.68 mmol, 0.7当量),加热至70℃反应4h,点板监测。反应结束后,加入饱和硫代硫酸钠溶液(30.0 mL), 乙酸乙酯(10.0 mL)萃取上述混合溶液三次,合并有机相,饱和碳酸钠溶液(30 mL)洗涤两次,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到化合物1-3(2.57 g,10.6 mmol,产率86%),白色固体。1H NMR (500 MHz, CDCl3) δ 4.41 (brs, 1H), 3.35 (brs, 1H), 2.73-2.68 (m, 1H),2.04-2.02 (m, 2H), 1.85-1.76 (m, 4H), 1.45-1.42 (m, 9H), 1.09-1.02 (m, 7H). LCMS [M+H]+ m/z: 243.2。The solid 1-2 (4.29 g, 12.4 mmol, 1.0 equivalent) was dissolved in tetrahydrofuran (30 mL) and water (6 mL), and iodine (2.20 g, 8.68 mmol, 0.7 equivalent) was added under stirring, and the mixture was heated to 70 ° C for 4 h, and the plate was monitored. After the reaction was completed, saturated sodium thiosulfate solution (30.0 mL) and ethyl acetate (10.0 mL) were added to extract the mixed solution three times, and the organic phases were combined, washed twice with saturated sodium carbonate solution (30 mL), dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain compound 1-3 (2.57 g, 10.6 mmol, yield 86%) as a white solid. 1 H NMR (500 MHz, CDCl 3 ) δ 4.41 (brs, 1H), 3.35 (brs, 1H), 2.73-2.68 (m, 1H), 2.04-2.02 (m, 2H), 1.85-1.76 (m, 4H), 1.45-1.42 (m, 9H), 1.09 -1.02 (m, 7H). LCMS [M+H] + m/z: 243.2.
2.化合物1 的制备:2. Preparation of Compound 1:
步骤1 中间体1-4的合成Step 1 Synthesis of intermediate 1-4
将化合物5-溴-2-氟嘧啶(35.0 g,198 mmol,1.0当量),XPhos (9.43 g, 19.8mmol,0.1当量),Pd2(dba)3(9.06 g, 9.9 mmol,0.05 当量)加入装有搅拌子的两颈瓶中,在氮气氛围下,随后加入THF(200 mL),含有锌试剂的四氢呋喃溶液(0.5 M, 396 mL, 198mmol),将反应液在60℃下搅拌12h。反应结束后,冷却至室温,加入氯化铵饱和溶液(300mL)淬灭反应,用乙酸乙酯(200 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到化合物1-4(6.00 g,28.1 mmol,产率14%),淡黄色固体。1H NMR (500 MHz, CDCl3) δ 8.56 (d, J =1.2 Hz, 2H), 3.56 (s, 2H), 1.46 (s, 9H)。LCMS [M+H]+ m/z: 213.1。Compound 5-bromo-2-fluoropyrimidine (35.0 g, 198 mmol, 1.0 equivalent), XPhos (9.43 g, 19.8mmol, 0.1 equivalent), Pd 2 (dba) 3 (9.06 g, 9.9 mmol, 0.05 equivalent) were added to a two-necked bottle equipped with a stirrer, and then THF (200 mL) and a tetrahydrofuran solution containing a zinc reagent (0.5 M, 396 mL, 198 mmol) were added under a nitrogen atmosphere. The reaction solution was stirred at 60°C for 12 hours. After the reaction was completed, the mixture was cooled to room temperature, saturated ammonium chloride solution (300 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (200 mL), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation under reduced pressure. The obtained mixture was separated and purified by silica gel column chromatography to obtain compound 1-4 (6.00 g, 28.1 mmol, yield 14%) as a light yellow solid. 1 H NMR (500 MHz, CDCl 3 ) δ 8.56 (d, J =1.2 Hz, 2H), 3.56 (s, 2H), 1.46 (s, 9H). LCMS [M+H] + m/z: 213.1.
步骤2 中间体1-5的合成Step 2 Synthesis of intermediate 1-5
将化合物(S)-(4-(1-氨基乙基)环己基)氨基甲酸叔丁酯 1-3(0.46g, 2.0 mmol,1.0当量)溶于二甲基亚砜(10 mL)中,加入化合物1-4(0.42 g,2.0 mmol,1.0 当量),DIPEA(1.29 g,10.0 mmol,5.0 当量),得到的反应液于100℃下反应12h。反应结束后,冷却至室温,加入氯化铵饱和溶液(10 mL)淬灭反应,用乙酸乙酯(20 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体1-5为白色固体。LCMS [M+H]+ m/z: 435.3。Compound (S)-tert-butyl (4-(1-aminoethyl)cyclohexyl)carbamate 1-3 (0.46 g, 2.0 mmol, 1.0 equivalent) was dissolved in dimethyl sulfoxide (10 mL), and compound 1-4 (0.42 g, 2.0 mmol, 1.0 equivalent) and DIPEA (1.29 g, 10.0 mmol, 5.0 equivalent) were added. The resulting reaction solution was reacted at 100°C for 12 h. After the reaction was completed, the mixture was cooled to room temperature, saturated ammonium chloride solution (10 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (20 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The resulting mixture was separated and purified by silica gel column chromatography to obtain intermediate 1-5 as a white solid. LCMS [M+H] + m/z: 435.3.
步骤3 中间体1-6的合成Step 3 Synthesis of intermediate 1-6
将上述中间体1-5(0.43g,1.0 mmol,1.0当量)溶于甲醇(5.0 mL)中,向该反应液中滴加盐酸的乙酸乙酯溶液(4.0M,0.75 mL, 3.0当量),点板检测。反应结束后,减压旋蒸得到白色固体中间体。将白色固体、2,8-二氯-7-甲基喹啉(0.21 g,1.0 mmol,1.0 当量),BINAP(0.12 g,0.2 mmol,0.2当量),Pd2(dba)3(0.09 g,0.1 mmol,0.1 当量)以及叔丁醇钠(0.29 g,3.0 mmol,3.0当量)加入装有搅拌子的10 mL两颈瓶中,在氮气氛围下,抽换气三次,随后加入THF(5.0 mL),将反应液在65℃下搅拌4h。点板监测,反应结束后,冷却至室温,加入氯化铵饱和溶液(10 mL)淬灭反应,用乙酸乙酯(20 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体1-6(0.15 g,0.3 mmol,产率32%),淡黄色固体。LCMS [M+H]+ m/z:510.3。The intermediate 1-5 (0.43 g, 1.0 mmol, 1.0 equivalent) was dissolved in methanol (5.0 mL), and a hydrochloric acid ethyl acetate solution (4.0 M, 0.75 mL, 3.0 equivalent) was added dropwise to the reaction solution, and the plate was tested. After the reaction was completed, the white solid intermediate was obtained by rotary evaporation under reduced pressure. The white solid, 2,8-dichloro-7-methylquinoline (0.21 g, 1.0 mmol, 1.0 equivalent), BINAP (0.12 g, 0.2 mmol, 0.2 equivalent), Pd 2 (dba) 3 (0.09 g, 0.1 mmol, 0.1 equivalent) and sodium tert-butoxide (0.29 g, 3.0 mmol, 3.0 equivalent) were added to a 10 mL two-necked flask equipped with a stirrer, and the flask was vented three times under a nitrogen atmosphere, and then THF (5.0 mL) was added, and the reaction solution was stirred at 65 ° C for 4 h. The reaction was monitored by spot plate. After the reaction was completed, the mixture was cooled to room temperature, saturated ammonium chloride solution (10 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (20 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 1-6 (0.15 g, 0.3 mmol, yield 32%) as a light yellow solid. LCMS [M+H]+ m/z: 510.3.
步骤4 化合物1的合成Step 4 Synthesis of compound 1
将中间体1-6 (0.15 g,0.3 mmol,1.0 当量)溶于四氢呋喃(5.0 mL)和甲醇(5.0ml)中,随后加入2.0 M氢氧化钠(0.75mL,1.5 mmol,5.0 当量),常温反应2h,点板监测,原料反应完后,用1.0 M稀盐酸中和反应pH至7-8,二氯甲烷(10 mL)萃取三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂,得到的油状化合物直接进行下一步。The intermediate 1-6 (0.15 g, 0.3 mmol, 1.0 equivalent) was dissolved in tetrahydrofuran (5.0 mL) and methanol (5.0 ml), and then 2.0 M sodium hydroxide (0.75 mL, 1.5 mmol, 5.0 equivalent) was added. The reaction was carried out at room temperature for 2 h, and the plate was monitored. After the reaction of the raw materials, 1.0 M dilute hydrochloric acid was used to neutralize the reaction pH to 7-8, and dichloromethane (10 mL) was extracted three times. The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained oily compound was directly used for the next step.
将上述油状化合物溶于N, N-二甲基甲酰胺(5.0 mL)中,依次加入化合物DIPEA(0.39 g,3 mmol,10.0 当量),HATU(0.15 g,0.6 mmol,2.0 当量)和3-氧杂环丁胺(0.048g,0.6 mmol,2.0 当量),得到的反应液于常温下反应1h。反应结束后,加入氯化铵饱和溶液(5 mL)淬灭反应,用乙酸乙酯(5 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到目标化合物1为白色固体。1H NMR (500 MHz, CDCl3) δ 8.18 (s, 2H), 7.75 (d, J = 8.8 Hz, 1H),7.37 (d, J = 8.0 Hz, 1H), 7.05 (d, J = 8.0 Hz, 1H), 6.59 (d, J = 8.8 Hz, 1H),6.43 (d, J = 6.4 Hz, 1H), 5.22 (d, J = 8.7 Hz, 1H), 5.02 (dt, J = 13.2, 6.6Hz, 1H), 4.91 (t, J = 7.0 Hz, 1H), 4.47 (t, J = 6.3 Hz, 2H), 4.05-3.98 (m,1H), 3.78 (brs, 1H), 3.33 (s, 2H), 2.54 (s, 3H), 2.28-2.24 (m, 2H), 1.97-1.94(m, 1H), 1.88-1.84 (m, 1H), 1.50-1.48 (m, 1H), 1.36-1.23 (m, 4H), 1.20 (d, J= 6.5 Hz, 3H). LCMS [M+H]+ m/z: 509.3。The above oily compound was dissolved in N, N-dimethylformamide (5.0 mL), and compound DIPEA (0.39 g, 3 mmol, 10.0 equivalents), HATU (0.15 g, 0.6 mmol, 2.0 equivalents) and 3-oxetaneamine (0.048 g, 0.6 mmol, 2.0 equivalents) were added in sequence. The obtained reaction solution was reacted at room temperature for 1 h. After the reaction was completed, a saturated ammonium chloride solution (5 mL) was added to quench the reaction, and the above mixed solution was extracted three times with ethyl acetate (5 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain the target compound 1 as a white solid. 1 H NMR (500 MHz, CDCl 3 ) δ 8.18 (s, 2H), 7.75 (d, J = 8.8 Hz, 1H), 7.37 (d, J = 8.0 Hz, 1H), 7.05 (d, J = 8.0 Hz, 1H), 6.59 (d, J = 8.8 Hz, 1H), 6.43 (d, J = 6.4 Hz, 1H), 5.22 (d, J = 8.7 Hz, 1H), 5.02 (dt, J = 13.2, 6.6Hz, 1H), 4.91 (t, J = 7.0 Hz, 1H), 4.47 (t, J = 6.3 Hz, 2H), 4.05-3.98 (m, 1H), 3.78 (brs, 1H), 3.33 (s, 2H), 2.54 (s, 3H), 2.28-2.24 (m, 2H), 1.97-1.94(m, 1H), 1.88-1.84 (m, 1H), 1.50-1.48 (m, 1H), 1.36-1.23 (m, 4H ), 1.20 (d, J= 6.5 Hz, 3H). LCMS [M+H] + m/z: 509.3.
实施例2: 化合物(R)-2-(2-((1-(4-((8-氯-7-甲基喹啉-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物2)的合成Example 2: Synthesis of Compound (R)-2-(2-((1-(4-((8-chloro-7-methylquinolin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 2)
关键起始物料(R)-(4-(1-氨基乙基)环己基)氨基甲酸叔丁酯(2-2)的制备:Preparation of the key starting material (R)-tert-butyl (4-(1-aminoethyl)cyclohexyl)carbamate (2-2):
步骤A 中间体2-1的合成Step A Synthesis of Intermediate 2-1
参照实施例1中化合物1-3的制备方法合成向带搅拌子的反应瓶中加入中间体1-1(5.0g,14.5 mmol, 1.0当量), 置换氮气三次,加入四氢呋喃(40.0 mL),在-78℃下滴加三仲丁基硼氢化锂(1.0M in THF, 21.8mL, 21.8mmol,1.5当量),恢复至室温下搅拌3小时,点板监测。反应结束后,在0℃下缓慢用水淬灭,乙酸乙酯(20.0 mL)萃取上述混合溶液三次,合并有机相,饱和氯化钠溶液(40.0 mL)洗涤两次,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到化合物2-1,白色固体(2.9g,8.4mmol, 产率58%)。 1H NMR (500 MHz, CD3OD) δ 4.58 (brs, 1H), 3.32-3.06 (m,1H), 1.96-1.91 (m, 3H),1.77-1.74(m, 1H), 1.43 (s, 9H), 1.27-1.22 (m, 12H),1.17-1.06 (m, 4H). LCMS [M+H]+ m/z: 347.2。Synthesize by referring to the preparation method of compound 1-3 in Example 1. Add intermediate 1-1 (5.0 g, 14.5 mmol, 1.0 equivalent) to a reaction bottle with a stirrer, replace nitrogen three times, add tetrahydrofuran (40.0 mL), add tri-sec-butyl lithium borohydride (1.0 M in THF, 21.8 mL, 21.8 mmol, 1.5 equivalent) dropwise at -78 ° C, return to room temperature and stir for 3 hours, and monitor with a plate. After the reaction is completed, slowly quench with water at 0 ° C, extract the above mixed solution three times with ethyl acetate (20.0 mL), combine the organic phases, wash twice with saturated sodium chloride solution (40.0 mL), dry with anhydrous sodium sulfate, filter, and remove the solvent by vacuum rotary evaporation. The obtained mixture is separated and purified by silica gel column chromatography to obtain compound 2-1 as a white solid (2.9 g, 8.4 mmol, yield 58%). 1 H NMR (500 MHz, CD 3 OD) δ 4.58 (brs, 1H), 3.32-3.06 (m,1H), 1.96-1.91 (m, 3H), 1.77-1.74 (m, 1H), 1.43 (s, 9H), 1.27-1.22 (m, 12H), 1.17- 1.06 (m, 4H). LCMS [M+H] + m/z: 347.2.
步骤B 关键起始物料2-2的合成Step B Synthesis of key starting material 2-2
将上述固体2-1(2.9 g,8.4 mmol,1.0当量),溶于四氢呋喃(20 mL)和水(4 mL),搅拌下加入碘(1.49g,5.88mmol, 0.7当量),加热至70℃反应4h,点板监测。反应结束后,加入饱和硫代硫酸钠溶液(20 mL), 乙酸乙酯(10.0 mL)萃取上述混合溶液三次,合并有机相,饱和碳酸钠溶液(20.0 mL)洗涤两次,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到化合物2-2(1.93 g,7.14 mmol,产率85%),白色固体。1H NMR (500 MHz, CDCl3) δ 4.06 (brs, 1H), 3.28 (brs, 1H), 2.65-2.61 (m,1H), 1.98-1.96 (m, 2H), 1.79-1.54 (m, 4H), 1.45-1.42 (m, 9H), 1.04-0.96 (m,7H). LCMS [M+H]+ m/z: 243.2。The solid 2-1 (2.9 g, 8.4 mmol, 1.0 equivalent) was dissolved in tetrahydrofuran (20 mL) and water (4 mL), and iodine (1.49 g, 5.88 mmol, 0.7 equivalent) was added under stirring, and the mixture was heated to 70°C for 4 hours and monitored by spot plate. After the reaction, saturated sodium thiosulfate solution (20 mL) and ethyl acetate (10.0 mL) were added to extract the mixed solution three times, the organic phases were combined, washed twice with saturated sodium carbonate solution (20.0 mL), dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain compound 2-2 (1.93 g, 7.14 mmol, yield 85%) as a white solid. 1 H NMR (500 MHz, CDCl 3 ) δ 4.06 (brs, 1H), 3.28 (brs, 1H), 2.65-2.61 (m,1H), 1.98-1.96 (m, 2H), 1.79-1.54 (m, 4H), 1.45-1.42 (m, 9H), 1.04 -0.96 (m,7H). LCMS [M+H] + m/z: 243.2.
2.化合物2的制备:2. Preparation of Compound 2:
参照实施例1中化合物1的制备过程,在步骤2中用关键起始物料化合物(R)-(4-(1-氨基乙基)环己基)氨基甲酸叔丁酯 2-2替换关键起始物料化合物(S)-(4-(1-氨基乙基)环己基)氨基甲酸叔丁酯 1-3,其他操作步骤类似得到化合物2 为白色固体。1H NMR(500 MHz, CDCl3) δ 8.18 (s, 2H), 7.75 (d, J = 8.8 Hz, 1H), 7.37 (d, J = 8.0Hz, 1H), 7.05 (d, J = 8.0 Hz, 1H), 6.59 (d, J = 8.8 Hz, 1H), 6.43 (d, J = 6.4Hz, 1H), 5.22 (d, J = 8.7 Hz, 1H), 5.02 (dt, J = 13.2, 6.6 Hz, 1H), 4.91 (t,J = 7.0 Hz, 1H), 4.47 (t, J = 6.3 Hz, 2H), 4.05-3.98 (m, 1H), 3.78 (brs, 1H),3.33 (s, 2H), 2.54 (s, 3H), 2.28-2.24 (m, 2H), 1.97-1.94 (m, 1H), 1.88-1.84(m, 1H), 1.50-1.48 (m, 1H), 1.36-1.23 (m, 4H), 1.20 (d, J = 6.5 Hz, 3H). LCMS[M+H]+ m/z: 509.3。Referring to the preparation process of compound 1 in Example 1, in step 2, the key starting material compound (S)-tert-butyl (4-(1-aminoethyl)cyclohexyl)carbamate 1-3 was replaced with the key starting material compound (R)-tert-butyl (4-(1-aminoethyl)cyclohexyl)carbamate 2-2, and the other operation steps were similar to obtain compound 2 as a white solid. 1 H NMR (500 MHz, CDCl 3 ) δ 8.18 (s, 2H), 7.75 (d, J = 8.8 Hz, 1H), 7.37 (d, J = 8.0Hz, 1H), 7.05 (d, J = 8.0 Hz, 1H), 6.59 (d, J = 8.8 Hz, 1H), 6.4 3 (d, J = 6.4Hz, 1H), 5.22 (d, J = 8.7 Hz, 1H), 5.02 (dt, J = 13.2, 6.6 Hz, 1H), 4.91 (t,J = 7.0 Hz, 1H), 4.47 (t, J = 6.3 Hz, 2H), 4.05-3.98 (m , 1H), 3.78 (brs, 1H), 3.33 (s, 2H), 2.54 (s, 3H), 2.28-2.24 (m, 2H), 1.97-1.94 (m, 1H), 1.88-1.84(m, 1H), 1.50-1.48 (m, 1H), 1.36-1.23 (m , 4H), 1.20 (d, J = 6.5 Hz, 3H). LCMS[M+H] + m/z: 509.3.
实施例3: (S)-2-(2-((1-(4-((8-氯-7-甲基喹啉-2-基)氨基)环己基)乙基)(甲基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物3)的合成Example 3: Synthesis of (S)-2-(2-((1-(4-((8-chloro-7-methylquinolin-2-yl)amino)cyclohexyl)ethyl)(methyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 3)
1.关键起始物料(S)-(4-(1-(甲基氨基)乙基)环己基)氨基甲酸叔丁酯(3-2)的制备:1. Preparation of key starting material (S)-tert-butyl (4-(1-(methylamino)ethyl)cyclohexyl)carbamate (3-2):
步骤A 中间体3-1的合成Step A Synthesis of Intermediate 3-1
在反应瓶中先后加入化合物1-3(1.50 g,6.19 mmol),甲酸乙酯 (30.0 mL), 在50℃下反应12小时,LCMS检测反应。反应结束后,减压旋干溶剂,得到白色固体3-1,直接进行下一步。LCMS [M+Na]+ m/z: 293.2。Compound 1-3 (1.50 g, 6.19 mmol) and ethyl formate (30.0 mL) were added to the reaction flask, and the mixture was reacted at 50°C for 12 hours. The reaction was detected by LCMS. After the reaction, the solvent was dried under reduced pressure to obtain a white solid 3-1, which was directly used for the next step. LCMS [M+Na] + m/z: 293.2.
步骤B 关键起始物料3-2的合成Step B Synthesis of key starting material 3-2
在氮气氛围下,将上述固体(1.60 g,5.92 mmol)溶于四氢呋喃溶液(160 mL),向体系中加入BH3 .THF (1.0 M,29.6 mL),在室温下反应12小时,点板监测。反应结束后,在0℃下滴加水(30 mL)淬灭反应。加入氢氧化钠(1.0 M,20 mL)和乙酸乙酯(100 mL),分离有机相,饱和氯化钠(30 mL)洗涤, 无水硫酸钠干燥。过滤,减压旋蒸除去溶剂溶剂,得到的混合物硅胶柱色谱层析分离,产物3-2 (460 mg,1.80 mmol,产率29%),白色固体。1H NMR (400MHz, CDCl3) δ 4.41 (br s, 1H), 3.81 (br s, 1H), 3.36 (br s, 1H), 2.57 (d, J =6.0 Hz, 2H), 2.36-2.47 (m, 1H), 2.00-2.11 (m, 2H), 1.70-1.95 (m, 3H), 1.44(s, 9H), 1.16-1.24 (m, 3H)。LCMS [M-56+H]+ m/z: 201.4。Under nitrogen atmosphere, the above solid (1.60 g, 5.92 mmol) was dissolved in tetrahydrofuran solution (160 mL), and BH 3 . THF (1.0 M, 29.6 mL) was added to the system. The reaction was allowed to react at room temperature for 12 hours and the plate was monitored. After the reaction was completed, water (30 mL) was added dropwise at 0°C to quench the reaction. Sodium hydroxide (1.0 M, 20 mL) and ethyl acetate (100 mL) were added, the organic phase was separated, washed with saturated sodium chloride (30 mL), and dried over anhydrous sodium sulfate. After filtering, the solvent was removed by rotary evaporation under reduced pressure, and the obtained mixture was separated by silica gel column chromatography to obtain product 3-2 (460 mg, 1.80 mmol, yield 29%) as a white solid. 1 H NMR (400MHz, CDCl 3 ) δ 4.41 (br s, 1H), 3.81 (br s, 1H), 3.36 (br s, 1H), 2.57 (d, J =6.0 Hz, 2H), 2.36-2.47 (m, 1H), 2.00-2.11 (m, 2H), 1. 70-1.95 (m, 3H), 1.44 (s, 9H), 1.16-1.24 (m, 3H). LCMS [M-56+H] + m/z: 201.4.
2.化合物3的制备:2. Preparation of compound 3:
步骤1 中间体3-3的合成Step 1 Synthesis of intermediate 3-3
将化合物3-2(460 mg,1.80 mmol,1.0当量)和化合物1-4(381 mg,1.80 mmol,1.0当量)溶于二甲基亚砜(5.0 mL)中,加入N, N-二异丙基乙胺(1.16 g,9.00 mmol,5.0当量)后于100 ℃搅拌过夜。点板监测,反应结束后,冷却至室温,加入饱和氯化铵溶液(10 mL)淬灭反应,用乙酸乙酯(5.0 mL)萃取上述混合溶液三次,合并有机相,用饱和氯化钠溶液洗涤三次后,经无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体3-3(420 mg,0.97 mmol,产率54%)。LC-MS [M+H]+ m/z: 449.3。Compound 3-2 (460 mg, 1.80 mmol, 1.0 equivalent) and compound 1-4 (381 mg, 1.80 mmol, 1.0 equivalent) were dissolved in dimethyl sulfoxide (5.0 mL), and N, N-diisopropylethylamine (1.16 g, 9.00 mmol, 5.0 equivalent) was added and stirred at 100 °C overnight. The reaction was monitored by spot plate. After the reaction was completed, it was cooled to room temperature, saturated ammonium chloride solution (10 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (5.0 mL). The organic phases were combined, washed three times with saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 3-3 (420 mg, 0.97 mmol, yield 54%). LC-MS [M+H] + m/z: 449.3.
步骤2 中间体3-4的合成Step 2 Synthesis of intermediate 3-4
将化合物3-3(100 mg,0.23 mmol,1.0当量)溶于甲醇(1.0 mL)中,加入4 M盐酸的1, 4-二氧六环溶液(1.0 mL),室温反应3小时,点板监测,反应结束,减压除去溶剂,得到的油状化合物直接进行下一步。将油状化合物、2,8-二氯-7-甲基喹啉(45 mg,0.23 mmol,1.0当量)、Pd2(dba)3(21 mg,0.023 mmol,0.1当量)、BINAP (29 mg,0.046 mmol,0.2当量)、叔丁醇钠(44 mg,0.46 mmol, 2.0当量)溶于四氢呋喃(5.0 mL)中,氮气保护下,40 ℃反应16小时后,点板监测,反应结束,冷却至室温,加入饱和氯化铵水溶液(15 mL),乙酸乙酯(15mL)萃取三次,合并有机相,无水硫酸钠干燥有机相,过滤,减压浓缩除去溶剂,通过硅胶柱色谱层析分离纯化得到黄色油状化合物3-4(37 mg,0.07 mmol,产率32%)。LCMS [M+H]+ m/z: 524.3Compound 3-3 (100 mg, 0.23 mmol, 1.0 equivalent) was dissolved in methanol (1.0 mL), and a 4 M hydrochloric acid solution in 1, 4-dioxane (1.0 mL) was added. The reaction was allowed to react at room temperature for 3 hours with a plate monitor. When the reaction was complete, the solvent was removed under reduced pressure, and the obtained oily compound was directly used for the next step. The oily compound, 2,8-dichloro-7-methylquinoline (45 mg, 0.23 mmol, 1.0 equivalent), Pd 2 (dba) 3 (21 mg, 0.023 mmol, 0.1 equivalent), BINAP (29 mg, 0.046 mmol, 0.2 equivalent), sodium tert-butoxide (44 mg, 0.46 mmol, 2.0 equivalent) were dissolved in tetrahydrofuran (5.0 mL), reacted at 40 °C for 16 hours under nitrogen protection, and the reaction was completed by spot plate monitoring. The reaction was cooled to room temperature, saturated aqueous ammonium chloride solution (15 mL) was added, and ethyl acetate (15 mL) was used for extraction three times. The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to remove the solvent. The yellow oily compound 3-4 (37 mg, 0.07 mmol, yield 32%) was obtained by separation and purification by silica gel column chromatography. LCMS [M+H] + m/z: 524.3
步骤3 化合物 3的合成Step 3 Synthesis of compound 3
将中间体3-4(37 mg,0.07 mmol,1.0当量)溶于四氢呋喃(1.0 mL)和甲醇(1.0mL)的混合溶剂中,随后加入2 M浓度的氢氧化钠水溶液(0.14 mL,0.28 mmol,4.0当量),室温反应。点板监测,原料反应完后,用1 M盐酸水溶液中和反应液至中性,二氯甲烷(15 mL)萃取三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸得黄色油状物,黄色油状物直接进行下一步反应。将油状物溶于N, N-二甲基甲酰胺(1.0 mL)中,依次向反应液中加入N, N-二异丙基乙胺(36 mg,0.28 mmol,4.0当量),HATU(53 mg,0.16 mmol,2.0当量)和3-氧杂环丁胺(11 mg,0.16 mmol,2.0当量),室温反应,反应结束后,向反应液中加入饱和氯化铵水溶液(10 mL),用乙酸乙酯(15 mL)萃取三次,合并有机相,饱和氯化钠水溶液洗涤有机层,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂,通过硅胶柱色谱层析分离纯化得白色固体合成物3(11.2 mg, 0.021 mmol, 产率30%)。1H NMR (500 MHz, CDCl3) δ 8.22 (s, 2H), 7.82(d, J = 8.7 Hz, 1H), 7.40 (d, J = 8.1 Hz, 1H), 7.10 (d, J = 8.0 Hz, 1H), 6.65(d, J = 8.2 Hz, 1H), 6.08 (d, J = 7.0 Hz, 1H), 5.10 -4.97 (m, 1H), 4.91 (t, J= 7.1 Hz, 2H), 4.75 -4.64 (m, 1H), 4.47 (t, J = 6.5 Hz, 2H), 3.75-3.67 (m,2H), 3.37 (s, 2H), 2.96 (s, 3H), 2.55 (s, 3H), 2.29-2.27 (m, 1H), 2.17-2.14(m, 1H), 2.03 -1.92 (m, 2H), 1.26-1.20 (m, 7H)。LCMS [M+H]+ m/z: 523.3。The intermediate 3-4 (37 mg, 0.07 mmol, 1.0 equivalent) was dissolved in a mixed solvent of tetrahydrofuran (1.0 mL) and methanol (1.0 mL), and then a 2 M aqueous sodium hydroxide solution (0.14 mL, 0.28 mmol, 4.0 equivalent) was added and reacted at room temperature. The reaction was monitored by spot plate. After the reaction of the raw material was completed, the reaction solution was neutralized with a 1 M aqueous hydrochloric acid solution to neutrality, extracted three times with dichloromethane (15 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and evaporated under reduced pressure to obtain a yellow oil, which was directly used for the next step. The oil was dissolved in N, N-dimethylformamide (1.0 mL), and N, N-diisopropylethylamine (36 mg, 0.28 mmol, 4.0 equivalents), HATU (53 mg, 0.16 mmol, 2.0 equivalents) and 3-oxetaneamine (11 mg, 0.16 mmol, 2.0 equivalents) were added to the reaction solution in sequence. The reaction was carried out at room temperature. After the reaction was completed, saturated aqueous ammonium chloride solution (10 mL) was added to the reaction solution, and the mixture was extracted three times with ethyl acetate (15 mL). The organic phases were combined, washed with saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The white solid synthetic compound 3 (11.2 mg, 0.021 mmol, yield 30%) was obtained by separation and purification by silica gel column chromatography. 1 H NMR (500 MHz, CDCl 3 ) δ 8.22 (s, 2H), 7.82(d, J = 8.7 Hz, 1H), 7.40 (d, J = 8.1 Hz, 1H), 7.10 (d, J = 8.0 Hz, 1H), 6.65(d, J = 8.2 Hz, 1H), 6. 08 (d, J = 7.0 Hz, 1H), 5.10 -4.97 (m, 1H), 4.91 (t, J= 7.1 Hz, 2H), 4.75 -4.64 (m, 1H), 4.47 (t, J = 6.5 Hz, 2H), 3.75-3.67 (m,2H), 3.37 (s, 2H), 2.96 (s, 3H), 2.55 (s, 3H), 2.29-2.27 (m, 1H), 2.17-2.14(m, 1H), 2.03 -1.92 (m, 2H), 1.26-1.20 (m, 7H). LCMS [M+H] + m/z: 523.3.
实施例4: (R)-2-(2-((1-(4-((8-氯-7-甲基喹啉-2-基)氨基)环己基)乙基)(甲基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物4)的合成Example 4: Synthesis of (R)-2-(2-((1-(4-((8-chloro-7-methylquinolin-2-yl)amino)cyclohexyl)ethyl)(methyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 4)
1关键起始物料(R)-(4-(1-(甲基氨基)乙基)环己基)氨基甲酸叔丁酯(4-2)的制备:1 Preparation of key starting material (R)-tert-butyl (4-(1-(methylamino)ethyl)cyclohexyl)carbamate (4-2):
步骤A 中间体4-1的合成Step A Synthesis of Intermediate 4-1
在反应瓶中先后加入化合物2-3 (1.50 g,6.19 mmol),甲酸乙酯(30 mL), 在50℃下反应12小时,LCMS检测反应。反应结束后,减压除去溶剂,得到白色固体即中间体4-1,直接进行下一步。LCMS [M+Na]+ m/z: 293.2。Compound 2-3 (1.50 g, 6.19 mmol) and ethyl formate (30 mL) were added to the reaction flask, and the mixture was reacted at 50°C for 12 hours. The reaction was detected by LCMS. After the reaction, the solvent was removed under reduced pressure to obtain a white solid, namely intermediate 4-1, which was directly subjected to the next step. LCMS [M+Na] + m/z: 293.2.
步骤B 关键起始物料4-2的合成Step B Synthesis of key starting material 4-2
在氮气氛围下,将4-1(1.60 g, 5.92 mmol)溶于四氢呋喃溶液(160 mL),向体系中加入BH3 .THF(1.0 M,29.6 mL),在室温下反应12小时,点板监测。反应结束后,在0 ℃下滴加水(30 mL)淬灭反应。加入氢氧化钠(1.0 M,20 mL)和乙酸乙酯(100 mL),分离有机相,饱和氯化钠(30 mL)洗涤, 无水硫酸钠干燥。过滤,减压旋蒸除去溶剂溶剂,得到的混合物硅胶柱色谱层析分离,产物4-2(394 mg,1.54 mmol,产率26%),白色固体。1H NMR (400 MHz,CDCl3) δ 4.33 (br s, 1H), 3.77 (br s, 1H), 3.30 (br d, J = 1.2 Hz, 1H), 2.68(br t, J = 6.0 Hz, 1H), 2.31 (d, J = 6.0 Hz, 3H), 2.00 (br d, J = 13.2 Hz,2H), 1.58-1.79 (m, 3H), 1.41-1.50 (m, 2H), 1.37 (s, 9H), 1.13 (br d, J = 6.8Hz, 3H)。LCMS [M-56+H]+ m/z: 201.4。Under nitrogen atmosphere, 4-1 (1.60 g, 5.92 mmol) was dissolved in tetrahydrofuran solution (160 mL), and BH 3 . THF (1.0 M, 29.6 mL) was added to the system. The reaction was allowed to react at room temperature for 12 hours and monitored by spot plate. After the reaction was completed, water (30 mL) was added dropwise at 0 °C to quench the reaction. Sodium hydroxide (1.0 M, 20 mL) and ethyl acetate (100 mL) were added, the organic phase was separated, washed with saturated sodium chloride (30 mL), and dried over anhydrous sodium sulfate. After filtration, the solvent was removed by rotary evaporation under reduced pressure, and the obtained mixture was separated by silica gel column chromatography to obtain the product 4-2 (394 mg, 1.54 mmol, yield 26%) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 4.33 (br s, 1H), 3.77 (br s, 1H), 3.30 (br d, J = 1.2 Hz, 1H), 2.68 (br t, J = 6.0 Hz, 1H), 2.31 (d, J = 6.0 Hz, 3H), 2.0 0 (br d, J = 13.2 Hz, 2H), 1.58-1.79 (m, 3H), 1.41-1.50 (m, 2H), 1.37 (s, 9H), 1.13 (br d, J = 6.8Hz, 3H). LCMS [M-56+H] + m/z: 201.4.
2化合物4的制备:2 Preparation of compound 4:
参照实施例3中化合物3的制备过程,在步骤1中用关键起始物料化合物4-2替换关键起始物料3-2,其他操作步骤类似得到化合物4 为白色固体。1H NMR (500 MHz, CDCl3) δ8.22 (s, 2H), 7.82 (d, J = 8.7 Hz, 1H), 7.40 (d, J = 8.1 Hz, 1H), 7.10 (d, J= 8.0 Hz, 1H), 6.65 (d, J = 8.2 Hz, 1H), 6.08 (d, J = 7.0 Hz, 1H), 5.10 -4.97(m, 1H), 4.91 (t, J = 7.1 Hz, 2H), 4.75 -4.64 (m, 1H), 4.47 (t, J = 6.5 Hz,2H), 3.75-3.67 (m, 2H), 3.37 (s, 2H), 2.96 (s, 3H), 2.55 (s, 3H), 2.29-2.27(m, 1H), 2.17-2.14 (m, 1H), 2.03 -1.92 (m, 2H), 1.26-1.20 (m, 7H). LCMS [M+H]+ m/z: 523.3。Referring to the preparation process of compound 3 in Example 3, the key starting material 3-2 was replaced with the key starting material compound 4-2 in step 1, and the other operation steps were similar to obtain compound 4 as a white solid. 1 H NMR (500 MHz, CDCl 3 ) δ8.22 (s, 2H), 7.82 (d, J = 8.7 Hz, 1H), 7.40 (d, J = 8.1 Hz, 1H), 7.10 (d, J= 8.0 Hz, 1H), 6.65 (d, J = 8.2 Hz, 1H), 6.0 8 (d, J = 7.0 Hz, 1H), 5.10 -4.97 (m, 1H), 4.91 (t, J = 7.1 Hz, 2H), 4.75 -4.64 (m, 1H), 4.47 (t, J = 6.5 Hz, 2H), 3.75-3.67 (m, 2H), 3.37 (s, 2H), 2.96 LCMS [M+H] + m/z: 523.3.
实施例5:(S)-2-(2-((1-(4-((8-氯喹啉-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺(化合物5)的合成Example 5: Synthesis of (S)-2-(2-((1-(4-((8-chloroquinolin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 5)
参照实施例1中化合物1的制备过程,在步骤3中用2,8-二氯喹啉替换2,8-二氯-7-甲基喹啉,其他操作步骤类似,得到化合物5为白色固体。1H NMR (500 MHz, DMSO-d6) δ8.82 (d, J = 6.5 Hz, 1H), 8.11 (s, 2H), 7.85 (d, J = 9.0 Hz, 1H), 7.60 (dd, J= 7.5, 1.5 Hz, 1H), 7.56 (dd, J = 8.0, 1.0 Hz, 1H), 7.12 (d, J = 7.0 Hz, 1H),7.07 (t, J = 7.5 Hz, 1H), 6.85 (d, J = 9.0 Hz, 1H), 6.78 (d, J = 9.0 Hz, 1H),4.81 – 4.74 (m, 1H), 4.70 (t, J = 6.5 Hz, 2H), 4.42 (t, J = 6.5 Hz, 2H), 3.95– 3.82 (m, 2H), 3.24 (s, 2H), 2.18 – 2.10 (m, 2H), 1.88 – 1.79 (m, 2H), 1.49– 1.41 (m, 1H), 1.26 – 1.20 (m, 2H), 1.18 – 1.15 (m, 2H), 1.09 (d, J = 6.5Hz, 3H).LC-MS [M+H]+ m/z: 495.3。Referring to the preparation process of compound 1 in Example 1, 2,8-dichloroquinoline was used to replace 2,8-dichloro-7-methylquinoline in step 3, and the other operation steps were similar to obtain compound 5 as a white solid. 1 H NMR (500 MHz, DMSO-d 6 ) δ8.82 (d, J = 6.5 Hz, 1H), 8.11 (s, 2H), 7.85 (d, J = 9.0 Hz, 1H), 7.60 (dd, J= 7.5, 1.5 Hz, 1H), 7.56 (dd, J = 8.0, 1.0 Hz, 1H), 7.12 (d, J = 7.0 Hz, 1H), 7.07 (t, J = 7.5 Hz, 1H), 6.85 (d, J = 9.0 Hz, 1H), 6.78 (d, J = 9.0 Hz, 1H), 4.81 – 4.74 (m, 1H), 4.70 (t, J = 6.5 Hz, 2H), 4.42 (t, J = 6.5 Hz, 2H), 3.95– 3.82 (m, 2H), 3.24 (s, 2H), 2.18 – 2.10 (m, 2H), 1.88 – 1.79 (m, 2H), 1.49– 1.41 (m, 1H), 1.26 – 1.20 (m, 2H), 1.18 – 1.15 (m, 2H), 1.09 (d, J = 6.5Hz, 3H). LC-MS [M+H] + m/z: 495.3.
实施例6:(S)-2-(2-((1-(4-((8-溴喹啉-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺(化合物6)的合成Example 6: Synthesis of (S)-2-(2-((1-(4-((8-bromoquinolin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 6)
参照实施例1中化合物1的制备过程,在步骤3中用8-溴-2-氯喹啉替换2,8-二氯-7-甲基喹啉,其他操作步骤类似,得到化合物6为白色固体。1H NMR (500 MHz, DMSO-d6) δ8.75 (d, J = 6.6 Hz, 1H), 8.04 (s, 2H), 7.77 (d, J = 8.9 Hz, 1H), 7.72 (dd, J= 7.6, 1.2 Hz, 1H), 7.54 (dd, J = 7.9, 1.1 Hz, 1H), 7.08 (d, J = 6.6 Hz, 1H),6.95 (t, J = 7.7 Hz, 1H), 6.78 (s, 1H), 6.69 (d, J = 8.9 Hz, 1H), 4.74-4.66(m, 1H), 4.64 (t, J = 6.7 Hz, 2H), 4.35 (t, J = 6.3 Hz, 2H), 3.84-3.76 (m,2H) 3.17 (s, 2H), 2.13-2.09 (m, 2H), 1.81-1.75 (m, 2H), 1.41-1.38 (m, 1H),1.38-1.05 (m, 4H), 1.03 (d, J = 6.5 Hz, 3H).. LCMS [M+H]+ m/z: 539.2。Referring to the preparation process of compound 1 in Example 1, 8-bromo-2-chloroquinoline was used to replace 2,8-dichloro-7-methylquinoline in step 3, and the other operation steps were similar to obtain compound 6 as a white solid. 1 H NMR (500 MHz, DMSO-d 6 ) δ8.75 (d, J = 6.6 Hz, 1H), 8.04 (s, 2H), 7.77 (d, J = 8.9 Hz, 1H), 7.72 (dd, J= 7.6, 1.2 Hz, 1H), 7.54 (dd, J = 7.9, 1.1 Hz, 1H), 7.08 (d, J = 6.6 Hz, 1H), 6.95 (t, J = 7.7 Hz, 1H), 6.78 (s, 1H), 6.69 (d, J = 8.9 Hz, 1H), 4.74-4.66(m, 1H), 4.64 (t, J = 6.7 Hz, 2H ), 4.35 (t, J = 6.3 Hz, 2H), 3.84-3.76 (m,2H) 3.17 (s, 2H), 2.13-2.09 (m, 2H), 1.81-1.75 (m, 2H), 1.41-1.38 (m, 1H),1.38-1.05 (m, 4H), 1.03 ( d, J = 6.5 Hz, 3H).. LCMS [M+H] + m/z: 539.2.
实施例7: (S)-2-(2-((1-(4-((8-溴喹唑啉-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺(化合物7)的合成Example 7: Synthesis of (S)-2-(2-((1-(4-((8-bromoquinazolin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 7)
参照实施例1中化合物1的制备过程,在步骤3中用8-溴-2-氯喹唑啉替换2,8-二氯-7-甲基喹啉,其他操作步骤类似,得到化合物7 为白色固体。1H NMR (500 MHz, DMSO-d6) δ 9.00 (s, 1H), 8.75 (d, J = 6.5 Hz, 1H), 8.04 (s, 2H), 7.94 (d, J = 7.5Hz, 1H), 7.73 (d, J = 8.0 Hz, 1H), 7.49 (d, J = 7.0 Hz, 1H), 7.05 (t, J = 7.5Hz, 1H), 6.79 (d, J = 9.0 Hz, 1H), 4.74 – 4.66 (m, 1H), 4.64 (t, J = 6.5 Hz,2H), 4.35 (t, J = 6.5 Hz, 2H), 3.86 – 3.70 (m, 2H), 3.17 (s, 2H), 2.07 – 1.99(m, 1H), 1.81 – 1.73 (m, 2H), 1.44 – 1.33 (m, 1H), 1.27 – 1.19 (m, 2H), 1.11– 1.03 (m, 2H), 1.02 (d, J = 6.5 Hz, 3H). LC-MS [M+H]+ m/z: 540.3。Referring to the preparation process of compound 1 in Example 1, 8-bromo-2-chloroquinazoline was used to replace 2,8-dichloro-7-methylquinoline in step 3, and the other operation steps were similar to obtain compound 7 as a white solid. 1 H NMR (500 MHz, DMSO-d 6 ) δ 9.00 (s, 1H), 8.75 (d, J = 6.5 Hz, 1H), 8.04 (s, 2H), 7.94 (d, J = 7.5Hz, 1H), 7.73 (d, J = 8.0 Hz, 1H), 7.49 (d, J = 7.0 Hz, 1H), 7.05 (t, J = 7.5Hz, 1H), 6.79 (d, J = 9.0 Hz, 1H), 4.74 – 4.66 (m, 1H), 4.64 (t, J = 6.5 Hz, 2H), 4.35 (t, J = 6.5 Hz, 2H), 3.86 – 3 .70 (m, 2H), 3.17 (s, 2H), 2.07 – 1.99 (m, 1H), 1.81 – 1.73 (m, 2H), 1.44 – 1.33 (m, 1H), 1.27 – 1.19 (m, 2H), 1.11 – 1.03 (m, 2H), 1.02 (d, J = 6.5 Hz, 3H). LC-MS [M+H] + m/z: 540.3.
实施例8:(S)-2-(2-((1-(4-((8-氯-1,7-萘啶-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺(化合物8)的合成Example 8: Synthesis of (S)-2-(2-((1-(4-((8-chloro-1,7-naphthyridin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 8)
化合物8的制备:Preparation of compound 8:
步骤1 中间体8-1的合成Step 1 Synthesis of intermediate 8-1
将化合物1-5(491 mg,1.1 mmol,1.0当量)溶于5.0 mL甲醇中,加入4 M盐酸的1,4-二氧六环溶液2.0 mL,室温反应3 h,点板监测,原料反应完后,旋干溶剂,得到的油状化合物直接进行下一步。然后,将上述油状化合物、2-氯-8-甲氧基-1,7-萘啶(240 mg,1.1mmol,1.0当量),BINAP(140 mg,0.23 mmol,0.2当量),Pd2(dba)3(110 mg,0.12 mmol,0.1当量)以及叔丁醇钠(434 mg,4.5 mmol,4.0当量)加入25 mL双颈瓶中,在氮气气氛下,抽换氮气三次,随后加入四氢呋喃(10 mL),将反应液在室温下搅拌过夜。反应结束后,加入饱和氯化铵水溶液(10 mL)淬灭反应,乙酸乙酯(10 mL)萃取上述混合溶液三次,将有机相收集,无水硫酸钠干燥,旋转蒸发除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体8-1 (47 mg,0.1 mmol,产率8%),[M+H]+ m/z: 496.3。Compound 1-5 (491 mg, 1.1 mmol, 1.0 equivalent) was dissolved in 5.0 mL methanol, and 2.0 mL of 4 M hydrochloric acid in 1,4-dioxane solution was added. The reaction was carried out at room temperature for 3 h, and the plate was monitored. After the reaction of the raw material was completed, the solvent was dried by spin drying, and the obtained oily compound was directly used for the next step. Then, the above oily compound, 2-chloro-8-methoxy-1,7-naphthyridine (240 mg, 1.1 mmol, 1.0 equivalent), BINAP (140 mg, 0.23 mmol, 0.2 equivalent), Pd 2 (dba) 3 (110 mg, 0.12 mmol, 0.1 equivalent) and sodium tert-butoxide (434 mg, 4.5 mmol, 4.0 equivalent) were added to a 25 mL double-necked flask. Under a nitrogen atmosphere, the nitrogen was replaced three times, and then tetrahydrofuran (10 mL) was added, and the reaction solution was stirred at room temperature overnight. After the reaction was completed, saturated aqueous ammonium chloride solution (10 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (10 mL), and the organic phase was collected, dried over anhydrous sodium sulfate, and the solvent was removed by rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 8-1 (47 mg, 0.1 mmol, yield 8%), [M+H] + m/z: 496.3.
步骤2 中间体8-2的合成Step 2 Synthesis of intermediate 8-2
首先,将中间体8-1(47 mg,0.1 mmol, 1.0当量)溶于5.0 mL乙酸乙酯中,随后加入4 M浓度的盐酸乙酸乙酯溶液(0.1 mL,0.4 mmol,4.0当量),40 ℃反应20 h,随后,往反应液中加入乙醇(2.0 mL),90 ℃回流20 h。点板监测,原料反应完后,减压除去溶剂,得到的油状化合物直接进行下一步。然后,将上述油状化合物溶于2.0 mL三氯氧磷中,110 ℃反应1.5 h。点板监测,反应结束后,减压除去三氯氧磷,用乙酸乙酯稀释反应液,然后在0℃下用NaHCO3饱和水溶液调节中性。用乙酸乙酯(4.0 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体8-2(35 mg,0.08 mmol,产率75%)。[M+H]+ m/z: 496.3。First, the intermediate 8-1 (47 mg, 0.1 mmol, 1.0 equivalent) was dissolved in 5.0 mL of ethyl acetate, and then a 4 M hydrochloric acid ethyl acetate solution (0.1 mL, 0.4 mmol, 4.0 equivalent) was added, and the reaction was carried out at 40 °C for 20 h. Then, ethanol (2.0 mL) was added to the reaction solution, and the reaction was refluxed at 90 °C for 20 h. After the reaction of the raw materials was completed, the solvent was removed under reduced pressure, and the obtained oily compound was directly used for the next step. Then, the above oily compound was dissolved in 2.0 mL of phosphorus oxychloride and reacted at 110 °C for 1.5 h. After the reaction was completed, the phosphorus oxychloride was removed under reduced pressure, and the reaction solution was diluted with ethyl acetate, and then adjusted to neutrality with a saturated aqueous solution of NaHCO 3 at 0 °C. The above mixed solution was extracted three times with ethyl acetate (4.0 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation under reduced pressure. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 8-2 (35 mg, 0.08 mmol, yield 75%). [M+H] + m/z: 496.3.
步骤3 化合物8的合成Step 3 Synthesis of compound 8
首先,将中间体8-2(35 mg,0.08 mmol, 1.0当量)溶于1.0 mL四氢呋喃和1.0 mL甲醇的混合溶剂中,随后加入2 M氢氧化钠水溶液(0.2 mL,0.4 mmol,5.0当量),常温反应2h,点板监测,原料反应完后,用1 M HCl水溶液中和反应至中性,用二氯甲烷(10 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压除去溶剂。得到的油状化合物直接进行下一步反应。 然后,将上述油状化合物溶于1.0 mL N, N-二甲基甲酰胺中,按顺序加入化合物N, N-二异丙基乙胺(39 mg,0.3 mmol,4.0当量),HATU(57 mg,0.15 mmol,2.0当量)和3-氧杂环丁胺(22 mg,0.3 mmol,2.0当量),得到的反应液于室温下反应1 h。反应结束后,加入饱和氯化铵水溶液(2.0 mL)淬灭反应,用乙酸乙酯(10 mL)萃取上述混合溶液三次,合并有机相,饱和氯化钠水溶液(10 mL)洗有机相,无水硫酸钠干燥有机相,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到8(6 mg,0.01 mmol,产率16%),为白色固体化合物。1H NMR (500 MHz, DMSO-d6) δ 8.76 (d, J = 6.5 Hz, 1H),8.05 (s, 2H), 7.89 (d, J = 5.0 Hz, 1H), 7.85 (d, J = 9.0 Hz, 1H), 7.49 (d, J= 5.0 Hz, 1H), 7.43 (d, J = 6.5 Hz, 1H), 6.93 (d, J = 9.0 Hz, 1H), 6.79 (d, J= 9.0 Hz, 1H), 4.75 – 4.67 (m, 1H), 4.64 (t, J = 6.5 Hz, 2H), 4.35 (t, J =6.5 Hz, 2H), 3.87 – 3.77 (m, 2H), 3.18 (s, 2H), 2.13 – 2.02 (m, 2H), 1.83 –1.72 (m, 2H), 1.44 – 1.34 (m, 1H), 1.14 – 1.09 (m, 4H), 1.03 (d, J = 6.5 Hz,3H). LC-MS [M+H]+ m/z: 496.3。First, the intermediate 8-2 (35 mg, 0.08 mmol, 1.0 equivalent) was dissolved in a mixed solvent of 1.0 mL tetrahydrofuran and 1.0 mL methanol, and then 2 M sodium hydroxide aqueous solution (0.2 mL, 0.4 mmol, 5.0 equivalent) was added, and the reaction was carried out at room temperature for 2 h, and the plate was monitored. After the reaction of the raw material was completed, it was neutralized with 1 M HCl aqueous solution until neutral, and the mixed solution was extracted three times with dichloromethane (10 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed under reduced pressure. The obtained oily compound was directly subjected to the next step of reaction. Then, the above oily compound was dissolved in 1.0 mL N, N-dimethylformamide, and compounds N, N-diisopropylethylamine (39 mg, 0.3 mmol, 4.0 equivalents), HATU (57 mg, 0.15 mmol, 2.0 equivalents) and 3-oxetaneamine (22 mg, 0.3 mmol, 2.0 equivalents) were added in sequence, and the resulting reaction solution was reacted at room temperature for 1 h. After the reaction was completed, saturated aqueous ammonium chloride solution (2.0 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (10 mL), the organic phases were combined, the organic phases were washed with saturated aqueous sodium chloride solution (10 mL), the organic phases were dried with anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain 8 (6 mg, 0.01 mmol, yield 16%) as a white solid compound. 1 H NMR (500 MHz, DMSO-d 6 ) δ 8.76 (d, J = 6.5 Hz, 1H), 8.05 (s, 2H), 7.89 (d, J = 5.0 Hz, 1H), 7.85 (d, J = 9.0 Hz, 1H), 7.49 (d, J= 5.0 Hz, 1H), 7 .43 (d, J = 6.5 Hz, 1H), 6.93 (d, J = 9.0 Hz, 1H), 6.79 (d, J= 9.0 Hz, 1H), 4.75 – 4.67 (m, 1H), 4.64 (t, J = 6.5 Hz, 2H), 4.35 (t, J =6.5 Hz, 2H ), 3.87 – 3.77 (m, 2H), 3.18 (s, 2H), 2.13 – 2.02 (m, 2H), 1.83 –1.72 (m, 2H), 1.44 – 1.34 (m, 1H), 1.14 – 1.09 (m, 4H), 1.03 (d, J = 6.5 Hz, 3H). LC-MS [M+H] + m/z: 496.3.
实施例9:2-(2-(((4-((8-氯-1,7-萘啶-2-基)氨基)环己基)(环丙基)甲基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺(化合物9)的合成Example 9: Synthesis of 2-(2-(((4-((8-chloro-1,7-naphthyridin-2-yl)amino)cyclohexyl)(cyclopropyl)methyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 9)
1.关键起始物料(4-(氨基(环丙基)甲基)环己基)氨基甲酸叔丁酯(9-5)的制备1. Preparation of the key starting material (tert-butyl 4-(amino(cyclopropyl)methyl)cyclohexyl)carbamate (9-5)
步骤A 中间体9-1的合成Step A Synthesis of Intermediate 9-1
将反式-4-((叔丁氧羰基)氨基)环己烷-1-羧酸(10 g,41.0 mmol,1.0当量),二甲羟胺盐酸盐(4.4 g,45.0 mmol,1.1当量),DMTMM(14.4 g,45.0 mmol,1.1当量),NMM(8.3g,82.0 mmol,2.0当量)溶于四氢呋喃(100 mL)和甲醇(100 mL)中,25 ℃反应4 h,点板监测反应结束后,加入水(200 mL)淬灭反应,用乙酸乙酯(200 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体9-1 (10 g,35.0 mmol,产率85%),LC-MS [M+H-56]+ m/z: 231.0。Trans-4-((tert-butyloxycarbonyl)amino)cyclohexane-1-carboxylic acid (10 g, 41.0 mmol, 1.0 equivalent), dimethylhydroxylamine hydrochloride (4.4 g, 45.0 mmol, 1.1 equivalent), DMTMM (14.4 g, 45.0 mmol, 1.1 equivalent), NMM (8.3 g, 82.0 mmol, 2.0 equivalent) were dissolved in tetrahydrofuran (100 mL) and methanol (100 mL), and the reaction was carried out at 25 °C for 4 h. After the reaction was completed by spot plate monitoring, water (200 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (200 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation under reduced pressure. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 9-1 (10 g, 35.0 mmol, yield 85%), LC-MS [M+H-56] + m/z: 231.0.
步骤B 中间体9-2的合成Step B Synthesis of Intermediate 9-2
将中间体9-1(10 g,35.0 mmol)加入装有搅拌子的500 mL三颈瓶中,在氮气氛围下,抽换气三次,随后加入四氢呋喃(200 mL)。在低温浴中,将反应体系冷却至-10℃,用恒压滴液漏斗滴加3 M环丙烷溴化镁(14 mL),滴加完毕后,0 ℃反应4小时,点板监测反应完全。加入氯化铵饱和溶液(200 mL)淬灭反应,用乙酸乙酯(200 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的粗品用50 mL石油醚打浆纯化得到中间体9-2(6 g,22.5 mmol,产率64%),LC-MS [M+H]+ m/z: 268.3。The intermediate 9-1 (10 g, 35.0 mmol) was added to a 500 mL three-necked flask equipped with a stirrer. Under a nitrogen atmosphere, the flask was evacuated three times, and then tetrahydrofuran (200 mL) was added. The reaction system was cooled to -10 °C in a low-temperature bath, and 3 M cyclopropane magnesium bromide (14 mL) was added dropwise with a constant pressure dropping funnel. After the addition was complete, the reaction was carried out at 0 °C for 4 hours, and the reaction was monitored by a plate. A saturated ammonium chloride solution (200 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (200 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The crude product was purified by slurrying with 50 mL of petroleum ether to obtain the intermediate 9-2 (6 g, 22.5 mmol, yield 64%), LC-MS [M+H] + m/z: 268.3.
步骤C 中间体 9-3 的合成Step C Synthesis of Intermediate 9-3
将中间体9-2(4.0 g,15.0 mmol,1.0当量),钛酸四乙酯(2.2 g,18.0 mmol,1.2当量),S-叔丁基亚磺酰胺(5.1 g,22.5 mmol,1.5当量)加入装有搅拌子的100 mL三颈瓶中,在氮气氛围下,抽换气三次,随后加入四氢呋喃(60 mL)。80 ℃反应12小时,点板监测反应完全。加入氯化铵饱和溶液(100 mL)淬灭反应,用乙酸乙酯(100 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体9-3(1.0 g,2.7 mmol,产率18%),LC-MS [M+H]+ m/z :371.3。Intermediate 9-2 (4.0 g, 15.0 mmol, 1.0 equivalent), tetraethyl titanate (2.2 g, 18.0 mmol, 1.2 equivalent), S-tert-butylsulfenamide (5.1 g, 22.5 mmol, 1.5 equivalent) were added to a 100 mL three-necked flask equipped with a stirrer. The mixture was evacuated three times under a nitrogen atmosphere, and then tetrahydrofuran (60 mL) was added. The reaction was carried out at 80 °C for 12 hours, and the reaction was monitored to be complete by spot plate. A saturated ammonium chloride solution (100 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (100 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 9-3 (1.0 g, 2.7 mmol, yield 18%), LC-MS [M+H] + m/z: 371.3.
步骤D 中间体 9-4 的合成Step D Synthesis of Intermediate 9-4
将中间体9-3(500 mg,1.4 mmol,1.0当量)加入装有搅拌子的20 mL三颈瓶中,在氮气氛围下,抽换气三次,随后加入四氢呋喃(10 mL)和水(0.2 mL)。在低温浴中,将反应体系冷却至-50℃,分批加入硼氢化钠(158 mg,4.2 mmol,3.0当量),加完后,-50 ℃反应2小时,点板监测反应完全。加入氯化铵饱和溶液(10 mL)淬灭反应,用乙酸乙酯(20 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的粗品通过二氯甲烷(5.0 mL)和石油醚(15 mL)重结晶纯化得到中间体9-4(300 mg,0.81 mmol,产率58%),LC-MS [M+H]+ m/z: 373.2。Intermediate 9-3 (500 mg, 1.4 mmol, 1.0 equivalent) was added to a 20 mL three-necked flask equipped with a stirrer. Under a nitrogen atmosphere, the flask was evacuated three times, and then tetrahydrofuran (10 mL) and water (0.2 mL) were added. The reaction system was cooled to -50 °C in a low-temperature bath, and sodium borohydride (158 mg, 4.2 mmol, 3.0 equivalent) was added in batches. After the addition was complete, the reaction was carried out at -50 °C for 2 hours, and the reaction was monitored by a plate. A saturated ammonium chloride solution (10 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (20 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The crude product was purified by recrystallization from dichloromethane (5.0 mL) and petroleum ether (15 mL) to obtain intermediate 9-4 (300 mg, 0.81 mmol, yield 58%), LC-MS [M+H] + m/z: 373.2.
步骤E 中间体9-5的合成Step E Synthesis of Intermediate 9-5
将中间体9-4(300 mg,0.81 mmol,1.0当量),I2(1 M,1.6 mL,2.0当量)溶于四氢呋喃(5.0 mL)和甲醇(1.0 mL)中,50 ℃反应过夜,点板监测反应完全。加入1 M氢氧化钠水溶液(2.0 mL)和饱和硫代硫酸钠(2.0 mL)淬灭反应,用乙酸乙酯(10 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂,得到中间体9-5(200 mg,0.74 mmol, 产率18%)。LC-MS [M+H]+ m/z: 269.3。Intermediate 9-4 (300 mg, 0.81 mmol, 1.0 equivalent) and I 2 (1 M, 1.6 mL, 2.0 equivalent) were dissolved in tetrahydrofuran (5.0 mL) and methanol (1.0 mL), and the reaction was carried out at 50 °C overnight. The reaction was monitored by spot plate. 1 M sodium hydroxide aqueous solution (2.0 mL) and saturated sodium thiosulfate (2.0 mL) were added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (10 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation to obtain intermediate 9-5 (200 mg, 0.74 mmol, yield 18%). LC-MS [M+H] + m/z: 269.3.
2化合物9的制备过程2 Preparation process of compound 9
参照实施例8中化合物8的制备过程,用关键起始物料(4-(氨基(环丙基)甲基)环己基)氨基甲酸叔丁酯替换(S)-(4-(1-氨基乙基)环己基)氨基甲酸叔丁酯,其他操作步骤类似,得到化合物9为白色固体。1H NMR (500 MHz, CDCl3) δ 8.15 (s, 2H), 8.04 (d, J= 5.0 Hz, 1H), 7.76 (d, J = 9.0 Hz, 1H), 7.34 (d, J = 5.0 Hz, 1H), 6.80 (d, J= 9.0 Hz, 1H), 6.28 (d, J = 7.5 Hz, 1H), 5.25 (d, J = 9.0 Hz, 1H), 5.06-5.04(m Hz, 1H), 4.91 (t, J = 7.0 Hz, 2H), 4.47 (t, J = 6.5 Hz, 2H), 3.87 (brs,1H), 3.40 (m, 1H), 3.34 (s, 2H), 2.28 (d, J = 12.0 Hz, 2H), 2.10 – 1.93 (m,3H), 1.74 – 1.61 (m, 3H), 1.51 – 1.35 (m, 2H), 0.97 – 0.89 (m, 1H), 0.60 (m,1H), 0.43 (m, 1H), 0.32 (m, 2H). LC-MS [M+H]+ m/z: 522.3。Referring to the preparation process of compound 8 in Example 8, the key starting material (tert-butyl 4-(amino(cyclopropyl)methyl)cyclohexyl)carbamate was used to replace (S)-tert-butyl (4-(1-aminoethyl)cyclohexyl)carbamate, and the other operation steps were similar to obtain compound 9 as a white solid. 1 H NMR (500 MHz, CDCl 3 ) δ 8.15 (s, 2H), 8.04 (d, J= 5.0 Hz, 1H), 7.76 (d, J = 9.0 Hz, 1H), 7.34 (d, J = 5.0 Hz, 1H), 6.80 (d, J= 9.0 Hz, 1H), 6. 28 (d, J = 7.5 Hz, 1H), 5.25 (d, J = 9.0 Hz, 1H), 5.06-5.04(m Hz, 1H), 4.91 (t, J = 7.0 Hz, 2H), 4.47 (t, J = 6.5 Hz, 2H), 3.87 (brs,1H), 3.40 ( m, 1H), 3.34 (s, 2H), 2.28 (d, J = 12.0 Hz, 2H), 2.10 – 1.93 (m,3H), 1.74 – 1.61 (m, 3H), 1.51 – 1.35 (m, 2H), 0.97 – 0.89 (m, 1H), 0.60 (m,1H), 0.43 (m, 1H), 0.32 (m, 2H). LC-MS [M+H] + m/z: 522.3.
实施例10:(S)-2-(2-((1-(4-((8-氯-1,7-萘啶-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺(化合物10)的合成Example 10: Synthesis of (S)-2-(2-((1-(4-((8-chloro-1,7-naphthyridin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 10)
关键起始物料8-氯-2-(甲磺酰基)吡啶并[3,4-d]嘧啶(10-5)的制备:Preparation of the key starting material 8-chloro-2-(methylsulfonyl)pyrido[3,4-d]pyrimidine (10-5):
步骤A 中间体 10-1 的合成Step A Synthesis of Intermediate 10-1
将2-甲基巯基-5-溴嘧啶-4-甲酸甲酯(10.0 g, 38.00 mmol)溶于1,4-二氧六环(60 mL)和水(12 mL)的混合溶剂中,依次加入碳酸钾(15.8 g, 114.00 mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(1.7 g, 2.32 mmol)和(E)-1-乙氧乙烯基-2-硼酸频那醇酯(11.3 g, 57.00 mmol)。反应混合物在氮气体系下,在90 ℃搅拌2小时。LCMS监测反应完成后,待反应液冷却至室温,将反应液倒入水(100 mL)中,乙酸乙酯(80mL)萃取三次,合并有机相,并用饱和食盐水(30 mL)洗涤两次,无水硫酸钠干燥,过滤减压浓缩得到粗品。粗品通过柱层析(二氧化硅,石油醚:乙酸乙酯 = 20:1至10:1)纯化,得到中间体10-1(7.2 g,产率:75 %),为黄色油状物。LC-MS [M+H]+ m/z: 255.2。2-Methylmercapto-5-bromopyrimidine-4-carboxylic acid methyl ester (10.0 g, 38.00 mmol) was dissolved in a mixed solvent of 1,4-dioxane (60 mL) and water (12 mL), and potassium carbonate (15.8 g, 114.00 mmol), [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium dichloromethane complex (1.7 g, 2.32 mmol) and (E)-1-ethoxyvinyl-2-boronic acid pinacol ester (11.3 g, 57.00 mmol) were added in sequence. The reaction mixture was stirred at 90 °C for 2 hours under a nitrogen system. After the reaction was completed by LCMS monitoring, the reaction solution was cooled to room temperature, poured into water (100 mL), extracted three times with ethyl acetate (80 mL), and the organic phases were combined and washed twice with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to obtain a crude product. The crude product was purified by column chromatography (silica, petroleum ether:ethyl acetate = 20:1 to 10:1) to give intermediate 10-1 (7.2 g, yield: 75%) as a yellow oil. LC-MS [M+H] + m/z: 255.2.
步骤B 中间体 10-2 的合成Step B Synthesis of Intermediate 10-2
将10-1(6.5 g, 25.56 mmol)溶于胺的甲醇溶液(20 mL, 7 M)。反应混合物在85℃下搅拌2小时。TLC监测反应完成后,将反应减压浓缩得到中间体10-2(6.5 g,粗品),没有进一步纯化直接用于下一步。10-1 (6.5 g, 25.56 mmol) was dissolved in a methanol solution of amine (20 mL, 7 M). The reaction mixture was stirred at 85 °C for 2 hours. After the reaction was completed by TLC monitoring, the reaction was concentrated under reduced pressure to obtain intermediate 10-2 (6.5 g, crude product), which was used directly in the next step without further purification.
步骤C 中间体 10-3 的合成Step C Synthesis of Intermediate 10-3
将10-2(6.5 g, 27.20 mmol)溶于甲苯(100 mL)中,向反应液中加入对甲苯磺酸(468.0 mg, 2.72 mmol)。反应混合物在90 ℃下搅拌2小时。LCMS监测反应完成后,将反应液减压浓缩得到粗品。粗品通过柱层析(二氧化硅,二氯甲烷:甲醇 = 30:1至20:1)纯化,得到中间体10-3(4.2 g,产率:80 %),为黄色固体。LC-MS [M+H]+ m/z: 194.3。10-2 (6.5 g, 27.20 mmol) was dissolved in toluene (100 mL), and p-toluenesulfonic acid (468.0 mg, 2.72 mmol) was added to the reaction solution. The reaction mixture was stirred at 90 °C for 2 hours. After the reaction was completed as monitored by LCMS, the reaction solution was concentrated under reduced pressure to obtain a crude product. The crude product was purified by column chromatography (silica, dichloromethane: methanol = 30:1 to 20:1) to obtain intermediate 10-3 (4.2 g, yield: 80%) as a yellow solid. LC-MS [M+H] + m/z: 194.3.
步骤D 中间体 10-4 的合成Step D Synthesis of Intermediate 10-4
将10-3(4.1 g, 21.22 mmol)溶于乙腈(100 mL)中,向反应液中加入三氯氧磷(5.9 mL, 63.70 mmol)。反应混合物在70 ℃下搅拌2小时。LCMS监测反应完成后,待反应液冷却至室温,将反应液倒入冰水(130 mL)中,水相用饱和碳酸氢钠溶液调节pH至7~8,乙酸乙酯(50 mL)萃取三次,合并有机相,并用饱和食盐水(50 mL)洗涤两次,无水硫酸钠干燥,过滤减压浓缩得到中间体10-4(3.6 g,产率:80 %),为黄色固体。1H NMR (400 MHz, CDCl3)δ 9.22 (s, 1H), 8.40 (d, J = 5.4 Hz, 1H), 7.60 (d, J = 5.4 Hz, 1H), 2.75 (s,3H). LC-MS [M+H]+ m/z: 212.2。10-3 (4.1 g, 21.22 mmol) was dissolved in acetonitrile (100 mL), and phosphorus oxychloride (5.9 mL, 63.70 mmol) was added to the reaction solution. The reaction mixture was stirred at 70 °C for 2 hours. After the reaction was completed by LCMS monitoring, the reaction solution was cooled to room temperature and poured into ice water (130 mL). The aqueous phase was adjusted to pH 7-8 with saturated sodium bicarbonate solution, extracted three times with ethyl acetate (50 mL), and the organic phases were combined and washed twice with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to obtain intermediate 10-4 (3.6 g, yield: 80%) as a yellow solid. 1 H NMR (400 MHz, CDCl 3 )δ 9.22 (s, 1H), 8.40 (d, J = 5.4 Hz, 1H), 7.60 (d, J = 5.4 Hz, 1H), 2.75 (s,3H). LC-MS [M+H] + m/z: 212.2.
步骤 E 中间体 10-5 的合成Step E Synthesis of Intermediate 10-5
将10-4(2.0 g, 9.45 mmol)溶于二氯甲烷(20 mL)中,在冰浴条件下向反应液中加入间氯过氧苯甲酸(4.6 g, 22.68 mmol)。反应混合物在25 ℃下搅拌2小时。LCMS监测反应完成,将反应液倒入水(30 mL)中,乙酸乙酯(20 mL)萃取三次,合并有机相,并用饱和食盐水(30 mL)洗涤两次,无水硫酸钠干燥,过滤减压浓缩得到粗品。粗品通过柱层析(二氧化硅,石油醚:乙酸乙酯 = 10:1至1:5)纯化,得到化合物10-5(1.4 g,产率:61 %),为黄色固体。1H NMR (400 MHz, CDCl3) δ 9.74 (s, 1H), 8.74 (d, J = 5.5 Hz, 1H), 7.87 (d,J = 5.5 Hz, 1H), 3.57 (s, 3H). LC-MS [M+H]+ m/z: 244.0。10-4 (2.0 g, 9.45 mmol) was dissolved in dichloromethane (20 mL), and m-chloroperbenzoic acid (4.6 g, 22.68 mmol) was added to the reaction solution under ice bath conditions. The reaction mixture was stirred at 25 °C for 2 hours. LCMS monitored the completion of the reaction, and the reaction solution was poured into water (30 mL), extracted three times with ethyl acetate (20 mL), and the organic phases were combined and washed twice with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to obtain a crude product. The crude product was purified by column chromatography (silica, petroleum ether: ethyl acetate = 10:1 to 1:5) to obtain compound 10-5 (1.4 g, yield: 61%) as a yellow solid. 1 H NMR (400 MHz, CDCl 3 ) δ 9.74 (s, 1H), 8.74 (d, J = 5.5 Hz, 1H), 7.87 (d,J = 5.5 Hz, 1H), 3.57 (s, 3H). LC-MS [M+H] + m/z: 244.0.
2化合物10的制备过程2 Preparation process of compound 10
参照实施例1中化合物1的制备过程,在步骤3中用8-氯-2-(甲磺酰基)吡啶并[3,4-d]嘧啶替换2,8-二氯-7-甲基喹啉,其他后续操作步骤类似,得到化合物10为白色固体。1H NMR (500 MHz, CDCl3) δ 9.05-9.03 (m, 1H), 8.20 (s, 2H), 8.10 (d, J = 5.5Hz, 1H), 7.41 (d, J = 5.0 Hz, 1H), 6.27 (d, J = 6.5 Hz, 1H), 5.64-5.62 (m,1H), 5.17 (d, J = 9.0 Hz, 1H), 5.09 – 5.00 (m, 1H), 4.92 (t, J = 6.5 Hz, 2H),4.47 (t, J = 6.5 Hz, 2H), 4.08 – 4.01 (m, 1H), 4.00 – 3.92 (m, 1H), 3.36 (s,2H), 2.38 – 2.15 (m, 2H), 2.02 – 1.95 (m, 1H), 1.90 – 1.84 (m, 2H), 1.57 –1.48 (m, 1H), 1.31 – 1.27 (m, 2H), 1.21 (d, J = 6.5 Hz, 3H). LC-MS [M+H]+ m/z: 497.3。Referring to the preparation process of compound 1 in Example 1, 8-chloro-2-(methylsulfonyl)pyrido[3,4-d]pyrimidine was used to replace 2,8-dichloro-7-methylquinoline in step 3, and other subsequent operation steps were similar to obtain compound 10 as a white solid. 1 H NMR (500 MHz, CDCl 3 ) δ 9.05-9.03 (m, 1H), 8.20 (s, 2H), 8.10 (d, J = 5.5Hz, 1H), 7.41 (d, J = 5.0 Hz, 1H), 6.27 (d, J = 6.5 Hz, 1H), 5.64-5. ( m, 1H), 3.36 (s,2H), 2.38 – 2.15 (m, 2H), 2.02 – 1.95 (m, 1H), 1.90 – 1.84 (m, 2H), 1.57 –1.48 (m, 1H), 1.31 – 1.27 (m, 2H), 1.21 (d, J = 6.5 Hz, 3H). LC-MS [M+H] + m/z: 497.3.
实施例11: (S)-2-(2-((1-(4-((8-氯-1,7-萘啶-2-基)氨基)环己基)乙基)(甲基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物11)的合成Example 11: Synthesis of (S)-2-(2-((1-(4-((8-chloro-1,7-naphthyridin-2-yl)amino)cyclohexyl)ethyl)(methyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 11)
化合物11的制备:Preparation of compound 11:
步骤1 中间体11-1的合成Step 1 Synthesis of intermediate 11-1
将化合物3-3(100 mg,0.22 mmol,1.0 当量)溶于甲醇(1.0 mL)中,加入盐酸的1,4-二氧六环溶液(1.0 mL),室温反应3小时,点板监测,反应结束,旋干溶剂,得到的油状化合物直接进行下一步。将该油状化合物和2-氯-8-甲氧基-1,7-萘啶(43 mg,0.22 mmol,1.0当量),BINAP(14 mg,0.044 mmol,0.2 当量),Pd2(dba)3(20 mg,0.022 mmol,0.1当量)以及叔丁醇钠(42mg,0.44 mmol,2.0 当量)加入四氢呋喃(5.0 mL),N2保护下,40 ℃反应16小时后,点板监测,反应结束,冷却至室温,加入饱和氯化铵水溶液(15 mL),乙酸乙酯(15 mL)萃取三次,无水硫酸钠干燥有机相,过滤,减压旋蒸除去溶剂,通过硅胶柱色谱层析分离纯化得到黄色油状化合物11-1(47 mg,0.09 mmol,产率41%)。LCMS [M+H]+ m/z: 507.3。Compound 3-3 (100 mg, 0.22 mmol, 1.0 equivalent) was dissolved in methanol (1.0 mL), and a solution of hydrochloric acid in 1,4-dioxane (1.0 mL) was added. The reaction was allowed to react at room temperature for 3 hours with a spot plate for monitoring. When the reaction was complete, the solvent was dried and the obtained oily compound was directly used for the next step. The oily compound and 2-chloro-8-methoxy-1,7-naphthyridine (43 mg, 0.22 mmol, 1.0 equivalent), BINAP (14 mg, 0.044 mmol, 0.2 equivalent), Pd 2 (dba) 3 (20 mg, 0.022 mmol, 0.1 equivalent) and sodium tert-butoxide (42 mg, 0.44 mmol, 2.0 equivalent) were added to tetrahydrofuran (5.0 mL). Under N 2 protection, the reaction was carried out at 40 ° C for 16 hours. The reaction was monitored by spot plate. The reaction was cooled to room temperature, saturated aqueous ammonium chloride solution (15 mL) was added, and ethyl acetate (15 mL) was used for extraction three times. The organic phase was dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The yellow oily compound 11-1 (47 mg, 0.09 mmol, yield 41%) was obtained by separation and purification by silica gel column chromatography. LCMS [M+H] + m/z: 507.3.
步骤2 中间体 11-2 的合成Step 2 Synthesis of intermediate 11-2
将化合物11-1(47 mg,0.09 mmol,1.0 当量)溶于乙酸乙酯(5.0 mL)中,随后加入4 M浓度的盐酸的乙酸乙酯溶液(0.36 mL,0.36 mmol,4.0 当量),40 ℃反应20小时后,加入乙醇(5.0 mL),90 ℃回流2小时。点板监测,原料反应完,减压除去溶剂,得到的油状化合物直接进行下一步。将该油状化合物溶于三氯氧磷(2.0 mL)中,110 ℃反应1.5 小时。点板监测,反应结束,减压除去三氯氧磷,向反应液中加入冰的饱和碳酸氢钠水溶液,调节反应液至中性,乙酸乙酯(15 mL)萃取三次。合并有机相,无水硫酸钠干燥,过滤,减压除去溶剂,通过硅胶柱色谱层析分离纯化得到淡黄色油状化合物11-2(35 mg,0.07 mmol,产率78%),LCMS [M+H]+ m/z: 483.3。Compound 11-1 (47 mg, 0.09 mmol, 1.0 equivalent) was dissolved in ethyl acetate (5.0 mL), and then a 4 M hydrochloric acid solution in ethyl acetate (0.36 mL, 0.36 mmol, 4.0 equivalent) was added. After reacting at 40 °C for 20 hours, ethanol (5.0 mL) was added and refluxed at 90 °C for 2 hours. The raw material was reacted after monitoring by the spot plate, and the solvent was removed under reduced pressure. The obtained oily compound was directly used for the next step. The oily compound was dissolved in phosphorus oxychloride (2.0 mL) and reacted at 110 °C for 1.5 hours. The reaction was completed by monitoring by the spot plate, and phosphorus oxychloride was removed under reduced pressure. An ice-cold saturated sodium bicarbonate aqueous solution was added to the reaction solution, and the reaction solution was adjusted to neutral, and extracted with ethyl acetate (15 mL) three times. The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed under reduced pressure. The compound 11-2 was separated and purified by silica gel column chromatography to obtain a light yellow oily compound (35 mg, 0.07 mmol, yield 78%), LCMS [M+H] + m/z: 483.3.
步骤3 化合物 11 的合成Step 3 Synthesis of compound 11
将化合物11-2(35 mg,0.07 mmol,1.0当量)溶于四氢呋喃(1.0 mL)和甲醇(1.0mL)的混合溶剂中,加入2 M浓度的氢氧化钠水溶液(0.14 mL,0.28 mmol,4.0 当量),室温反应2小时,点板监测,原料反应完,用1 M盐酸水溶液调节反应液至中性,二氯甲烷(15 mL)萃取三次,合并有机相,无水硫酸钠干燥,过滤,减压除去溶剂,得到的油状化合物直接进行下一步。将该油状化合物溶于N, N-二甲基甲酰胺(1.0 mL)中,并向其中依次加入N, N-二异丙基乙胺(36 mg,0.28 mmol,4.0当量),HATU(27 mg,0.14 mmol,2.0当量)和3-氧杂环丁胺(21 mg,0.14 mmol,2.0当量),室温反应1小时后,点板监测,反应结束,向反应液中加入饱和氯化铵水溶液(10 mL),乙酸乙酯萃(15 mL)萃取三次,合并有机相,饱和氯化钠水溶液(10 mL)洗涤有机相,无水硫酸钠干燥,过滤,减压除去溶剂,通过硅胶柱色谱层析分离纯化得到白色固体11(7.8 mg,0.02 mmol,产率21%)。1H NMR (500 MHz, CDCl3) δ 8.22 (s,2H), 8.04 (d, J = 5.2 Hz, 1H), 7.77 (d, J = 8.9 Hz, 1H), 7.34 (d, J = 5.3 Hz,1H), 6.81 (d, J = 8.9 Hz, 1H), 6.19 (d, J = 7.1 Hz, 1H), 5.09 - 5.01 (m, 1H),4.91 (t, J = 7.1 Hz, 2H), 4.76 -4.69 (m, 1H), 4.46 (t, J = 6.4 Hz, 2H), 3.86(s, 1H), 3.36 (s, 2H), 2.97 (s, 3H), 2.36 - 2.28 (m, 1H), 2.23 - 2.16 (m,1H), 2.00 - 1.91 (m, 1H), 1.79 - 1.65 (m, 2H), 1.59 - 1.47 (m, 1H), 1.38 -1.23 (m, 3H), 1.22 (d, J = 6.8 Hz, 3H). LCMS [M+H]+ m/z: 510.3。Compound 11-2 (35 mg, 0.07 mmol, 1.0 equivalent) was dissolved in a mixed solvent of tetrahydrofuran (1.0 mL) and methanol (1.0 mL), and a 2 M aqueous sodium hydroxide solution (0.14 mL, 0.28 mmol, 4.0 equivalent) was added. The reaction was allowed to react at room temperature for 2 hours with a spot plate monitor. After the reaction of the raw materials was completed, the reaction solution was adjusted to neutrality with a 1 M aqueous hydrochloric acid solution, extracted three times with dichloromethane (15 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed under reduced pressure. The obtained oily compound was directly used for the next step. The oily compound was dissolved in N, N-dimethylformamide (1.0 mL), and N, N-diisopropylethylamine (36 mg, 0.28 mmol, 4.0 equivalents), HATU (27 mg, 0.14 mmol, 2.0 equivalents) and 3-oxetaneamine (21 mg, 0.14 mmol, 2.0 equivalents) were added thereto in sequence. After reacting at room temperature for 1 hour, the reaction was monitored by spot plate. When the reaction was completed, saturated aqueous ammonium chloride solution (10 mL) was added to the reaction solution, and the mixture was extracted with ethyl acetate (15 mL) three times. The organic phases were combined, washed with saturated aqueous sodium chloride solution (10 mL), dried over anhydrous sodium sulfate, filtered, and the solvent was removed under reduced pressure. The mixture was separated and purified by silica gel column chromatography to obtain a white solid 11 (7.8 mg, 0.02 mmol, yield 21%). 1 H NMR (500 MHz, CDCl 3 ) δ 8.22 (s,2H), 8.04 (d, J = 5.2 Hz, 1H), 7.77 (d, J = 8.9 Hz, 1H), 7.34 (d, J = 5.3 Hz,1H), 6.81 (d, J = 8.9 Hz, 1H), 6.19 (d, J = 7.1 Hz, 1H), 5.09 - 5.01 (m, 1H), 4.91 (t, J = 7.1 Hz, 2H), 4.76 -4.69 (m, 1H), 4.46 (t, J = 6.4 Hz, 2H), 3.86 (s, 1H), 3.36 (s, 2H), 2.97 (s, 3H), 2.36 - 2.28 (m, 1H), 2.23 - 2.16 (m,1H), 2.00 - 1.91 (m, 1H), 1.79 - 1.65 (m, 2H), 1.59 - 1.47 (m, 1H), 1.38 -1.23 (m, 3H), 1.22 ( d, J = 6.8 Hz, 3H). LCMS [M+H] + m/z: 510.3.
实施例12: (S)-2-(6-((1-(4-((8-氯-1,7-萘啶-2-基)氨基)环己基)乙基)氨基)吡啶-3-基)-N-(氧杂环丁烷-3-基)乙酰胺(化合物12)的合成Example 12: Synthesis of (S)-2-(6-((1-(4-((8-chloro-1,7-naphthyridin-2-yl)amino)cyclohexyl)ethyl)amino)pyridin-3-yl)-N-(oxetane-3-yl)acetamide (Compound 12)
化合物12的制备:Preparation of compound 12:
步骤1 中间体12-1的合成Step 1 Synthesis of intermediate 12-1
将化合物1-3(265 mg,1.1 mmol,1.2当量)、2-(6-溴吡啶-3-基)乙酸甲酯(210mg,0.91 mmol,1.0 当量),碘化亚铜(52 mg,0.27 mmol,0.3当量),L-脯氨酸(63 mg,0.55mmol,0.6当量)以及磷酸钾(386 mg,1.82 mmol,2.0当量)加入装有搅拌子的10 mL双颈瓶中,在氮气氛围下,抽换气三次,随后加入二甲基亚砜(3.0 mL),将反应液在100 ℃下搅拌过夜。点板监测,反应结束后,冷却至室温,加入氯化铵饱和溶液(10 mL)淬灭反应,用乙酸乙酯(20 mL)萃取上述混合溶液三次,合并有机相,饱和氯化钠水溶液洗有机相,无水硫酸钠干燥有机相,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体12-1(74 mg,0.19 mmol,产率17%)。LC-MS [M+H]+ m/z: 392.3。Compound 1-3 (265 mg, 1.1 mmol, 1.2 equivalents), methyl 2-(6-bromopyridin-3-yl)acetate (210 mg, 0.91 mmol, 1.0 equivalents), cuprous iodide (52 mg, 0.27 mmol, 0.3 equivalents), L-proline (63 mg, 0.55 mmol, 0.6 equivalents) and potassium phosphate (386 mg, 1.82 mmol, 2.0 equivalents) were added to a 10 mL double-necked flask equipped with a stirrer, and the mixture was evacuated three times under a nitrogen atmosphere, and then dimethyl sulfoxide (3.0 mL) was added, and the reaction solution was stirred at 100 °C overnight. The reaction was monitored by spot plate. After the reaction was completed, the mixture was cooled to room temperature, saturated ammonium chloride solution (10 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (20 mL), the organic phases were combined, the organic phases were washed with saturated sodium chloride aqueous solution, the organic phases were dried with anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 12-1 (74 mg, 0.19 mmol, yield 17%). LC-MS [M+H] + m/z: 392.3.
步骤2 中间体 12-2 的合成Step 2 Synthesis of intermediate 12-2
将中间体12-1(98 mg,0.25 mmol,1.0 当量)溶于甲醇(2.0 mL)中,加入4 M盐酸的乙酸乙酯溶液(0.25 mL),常温反应3 h,点板监测,原料反应完后,减压除去溶剂,得到的油状化合物直接进行下一步。The intermediate 12-1 (98 mg, 0.25 mmol, 1.0 equivalent) was dissolved in methanol (2.0 mL), and a 4 M hydrochloric acid solution in ethyl acetate (0.25 mL) was added. The reaction was carried out at room temperature for 3 h with spot plate monitoring. After the reaction of the raw material was completed, the solvent was removed under reduced pressure, and the obtained oily compound was directly used for the next step.
将上述油状化合物和2-氯-8-甲氧基-1,7-萘啶(46 mg,0.25 mmol,1.0当量)溶于二甲基亚砜(1.0 mL)中,加入碳酸钾(104 mg,0.75 mmol,3.0当量)后于100 ℃搅拌过夜。点板监测,反应结束后,冷却至室温,加入氯化铵饱和溶液(5.0 mL)淬灭反应,用乙酸乙酯(15 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体12-2(20 mg,0.044 mmol,产率18%)。LC-MS [M+H]+ m/z: 450.3。The above oily compound and 2-chloro-8-methoxy-1,7-naphthyridine (46 mg, 0.25 mmol, 1.0 equivalent) were dissolved in dimethyl sulfoxide (1.0 mL), potassium carbonate (104 mg, 0.75 mmol, 3.0 equivalent) was added and stirred at 100 °C overnight. The reaction was monitored by spot plate. After the reaction was completed, it was cooled to room temperature, saturated ammonium chloride solution (5.0 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (15 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 12-2 (20 mg, 0.044 mmol, yield 18%). LC-MS [M+H] + m/z: 450.3.
步骤3 中间体 12-3的合成Step 3 Synthesis of intermediate 12-3
首先,将中间体12-2(20 mg,0.044 mmol ,1.0当量)溶于乙酸乙酯(1.0 mL)中,随后加入4 M浓度的盐酸乙酸乙酯溶液(0.044 mL,0.18mmol,4.0当量),40 ℃反应20 h,随后,往反应液中加入乙醇(1.0 mL),90 ℃回流20 h。点板监测,原料反应完后,减压除去溶剂,得到的油状化合物直接进行下一步。First, the intermediate 12-2 (20 mg, 0.044 mmol, 1.0 equivalent) was dissolved in ethyl acetate (1.0 mL), and then a 4 M hydrochloric acid ethyl acetate solution (0.044 mL, 0.18 mmol, 4.0 equivalent) was added, and the mixture was reacted at 40 °C for 20 h. Subsequently, ethanol (1.0 mL) was added to the reaction solution, and the mixture was refluxed at 90 °C for 20 h. After the reaction of the raw material was completed, the solvent was removed under reduced pressure, and the obtained oily compound was directly used for the next step.
然后,将上述油状化合物溶于三氯氧磷(1.0 mL)中,110 ℃反应1.5 h。点板监测,反应结束后,减压除去三氯氧磷,用乙酸乙酯(5.0 mL)稀释反应液,然后在0 ℃下用NaHCO3饱和水溶液调节至中性。用乙酸乙酯萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体12-3(6 mg,0.013 mmol,产率30%)。LC-MS [M+H]+ m/z: 468.3。Then, the above oily compound was dissolved in phosphorus oxychloride (1.0 mL) and reacted at 110 °C for 1.5 h. The reaction was monitored by spot plate. After the reaction was completed, the phosphorus oxychloride was removed under reduced pressure, the reaction solution was diluted with ethyl acetate (5.0 mL), and then adjusted to neutral with a saturated aqueous solution of NaHCO 3 at 0 °C. The mixed solution was extracted three times with ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 12-3 (6 mg, 0.013 mmol, yield 30%). LC-MS [M+H] + m/z: 468.3.
步骤4 化合物 12 的合成Step 4 Synthesis of compound 12
首先,将中间体12-3(6 mg,0.013 mmol ,1.0当量)溶于四氢呋喃(0.5 mL)和甲醇(0.5 mL)的混合溶剂中,随后加入2M氢氧化钠水溶液(0.025 mL,0.05 mmol,5.0当量),常温反应2h,点板监测,原料反应完后,用1 M HCl水溶液中和反应至中性,用二氯甲烷(10mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压除去溶剂。得到的油状化合物直接进行下一步反应。First, the intermediate 12-3 (6 mg, 0.013 mmol, 1.0 equivalent) was dissolved in a mixed solvent of tetrahydrofuran (0.5 mL) and methanol (0.5 mL), and then a 2M sodium hydroxide aqueous solution (0.025 mL, 0.05 mmol, 5.0 equivalent) was added, and the reaction was carried out at room temperature for 2 h, and the plate was monitored. After the reaction of the raw material was completed, the reaction was neutralized with a 1 M HCl aqueous solution until it was neutral, and the mixed solution was extracted three times with dichloromethane (10 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed under reduced pressure. The obtained oily compound was directly subjected to the next step of reaction.
将上述油状化合物溶于N, N-二甲基甲酰胺(0.5 mL)中,按顺序加入化合物N, N-二异丙基乙胺(7 mg,0.052 mmol,4.0当量),HATU(10 mg,0.026 mmol,2.0当量)和3-氧杂环丁胺(2 mg,0.026 mmol,2.0当量),得到的反应液于常温下反应1 h。反应结束后,加入氯化铵饱和溶液(2.0 mL)淬灭反应,用乙酸乙酯(10 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到化合物12(2 mg,0.004 mmol,产率31%),为白色固体化合物。1H NMR (500 MHz,DMSO-d6) δ 8.82 (d, J = 6.5 Hz, 1H), 8.01 (d, J = 5.0 Hz, 1H), 7.97 (d, J =9.0 Hz, 1H), 7.84 (d, J = 2.0 Hz, 1H), 7.61 (d, J = 5.0 Hz, 1H), 7.55 (d, J =7.0 Hz, 1H), 7.30 (dd, J = 8.5, 2.0 Hz, 1H), 7.05 (d, J = 8.5 Hz, 1H), 6.49(d, J = 8.5 Hz, 1H), 6.34 – 6.17 (m, 1H), 4.85 – 4.78 (m, 1H), 4.75 (t, J =6.5 Hz, 2H), 4.46 (t, J = 6.0 Hz, 2H), 3.96 – 3.88 (m, 2H), 3.27 (s, 2H),2.27 – 2.14 (m, 2H), 1.95 – 1.83 (m, 2H), 1.54 – 1.47 (m, 1H), 1.28 – 1.21(m, 4H), 1.13 (d, J = 6.5 Hz, 3H).LC-MS [M+H]+ m/z: 495.3。The above oily compound was dissolved in N, N-dimethylformamide (0.5 mL), and compound N, N-diisopropylethylamine (7 mg, 0.052 mmol, 4.0 equivalents), HATU (10 mg, 0.026 mmol, 2.0 equivalents) and 3-oxetaneamine (2 mg, 0.026 mmol, 2.0 equivalents) were added in sequence, and the resulting reaction solution was reacted at room temperature for 1 h. After the reaction was completed, a saturated ammonium chloride solution (2.0 mL) was added to quench the reaction, and the above mixed solution was extracted three times with ethyl acetate (10 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The resulting mixture was separated and purified by silica gel column chromatography to obtain compound 12 (2 mg, 0.004 mmol, yield 31%) as a white solid compound. 1 H NMR (500 MHz, DMSO-d 6 ) δ 8.82 (d, J = 6.5 Hz, 1H), 8.01 (d, J = 5.0 Hz, 1H), 7.97 (d, J =9.0 Hz, 1H), 7.84 (d, J = 2.0 Hz, 1H), 7.61 (d, J = 5.0 Hz, 1H), 7.55 (d, J =7.0 Hz, 1H), 7.30 (dd, J = 8.5, 2.0 Hz, 1H), 7.05 (d, J = 8.5 Hz, 1H), 6.49 (d, J = 8.5 Hz, 1H), 6.34 – 6.17 (m, 1H), 4.85 – 4.78 (m, 1H), 4.75 (t, J =6.5 Hz, 2H), 4.46 (t, J = 6.0 Hz, 2H), 3.96 – 3.88 (m, 2H), 3.27 (s, 2H), 2.27 – 2.14 (m, 2H), 1.95 – 1.83 (m, 2H), 1.54 – 1.47 (m, 1H), 1.28 – 1.21 (m, 4H), 1.13 (d, J = 6.5 Hz, 3H). LC-MS [M+H] + m/z: 495.3.
实施例13:2-(2-(((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)丙酰胺(化合物13)的合成Example 13: Synthesis of 2-(2-(((S)-1-(4-((8-chloro-1,7-naphthyridin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)propanamide (Compound 13)
化合物13的制备:Preparation of compound 13:
步骤1 中间体13-1的合成Step 1 Synthesis of intermediate 13-1
将化合物1-4(300 mg,1.4 mmol,1.0当量)加入装有搅拌子的10 mL双颈瓶中,在氮气氛围下,抽换气三次,随后加入N, N-二甲基甲酰胺(3.0 mL)。在低温浴中,将反应体系冷却至-30℃,加入NaH(85 mg,2.1 mmol,1.5当量),加入碘甲烷(298 mg,2.1 mmol,1.5当量)反应1h,点板监测反应完全。加入氯化铵饱和溶液(10 mL)淬灭反应,用乙酸乙酯(20mL)萃取上述混合溶液三次,合并有机相,用饱和氯化钠水溶液(20 mL)水洗有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体13-1(60 mg,0.27 mmol,产率19%),LC-MS [M+H]+m/z: 227.2.Compound 1-4 (300 mg, 1.4 mmol, 1.0 equivalent) was added to a 10 mL double-necked flask equipped with a stirrer. The mixture was evacuated three times under a nitrogen atmosphere, and then N, N-dimethylformamide (3.0 mL) was added. The reaction system was cooled to -30°C in a low-temperature bath, and NaH (85 mg, 2.1 mmol, 1.5 equivalent) and iodomethane (298 mg, 2.1 mmol, 1.5 equivalent) were added to react for 1 h, and the reaction was monitored by spot plate. A saturated ammonium chloride solution (10 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (20 mL). The organic phases were combined, washed with a saturated sodium chloride aqueous solution (20 mL), dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 13-1 (60 mg, 0.27 mmol, yield 19%), LC-MS [M+H] + m/z: 227.2.
步骤2 中间体 13-2的合成Step 2 Synthesis of intermediate 13-2
将化合物13-1(225 mg,1.0 mmol,1.0当量)和化合物1-3 (222 mg,1.2 mmol,1.2当量)溶于DMSO(3.0 mL)中,加入N,N-二异丙基乙胺(645 mg,5.0 mmol,5.0当量),100℃过夜反应,点板监测反应完全。加入氯化铵饱和溶液(10 mL)淬灭反应,用乙酸乙酯(20 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体13-2(200 mg,0.45 mmol,产率45%),LC-MS [M+H]+m/z: 449.3.Compound 13-1 (225 mg, 1.0 mmol, 1.0 equivalent) and compound 1-3 (222 mg, 1.2 mmol, 1.2 equivalent) were dissolved in DMSO (3.0 mL), and N,N-diisopropylethylamine (645 mg, 5.0 mmol, 5.0 equivalent) was added. The reaction was carried out at 100°C overnight, and the reaction was monitored by spot plate. A saturated ammonium chloride solution (10 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (20 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 13-2 (200 mg, 0.45 mmol, yield 45%), LC-MS [M+H] + m/z: 449.3.
步骤3 中间体 13-3 的合成Step 3 Synthesis of intermediate 13-3
将中间体13-2(200 mg,0.45 mmol,1.0 当量)溶于甲醇 (3.0 mL) 中,加入4M盐酸的乙酸乙酯溶液(0.25 mL),常温反应3h,点板监测,原料反应完后,减压除去溶剂,得到的油状化合物直接进行下一步。Intermediate 13-2 (200 mg, 0.45 mmol, 1.0 equivalent) was dissolved in methanol (3.0 mL), and a 4M hydrochloric acid solution in ethyl acetate (0.25 mL) was added. The reaction was carried out at room temperature for 3 h with spot plate monitoring. After the reaction of the raw material was completed, the solvent was removed under reduced pressure, and the obtained oily compound was directly used for the next step.
将上述油状化合物和2-氯-8-甲氧基-1,7-萘啶(87 mg,0.45 mmol,1.0当量),BINAP(56 mg,0.09 mmol,0.2当量),Pd2(dba)3(46 mg,0.05 mmol,0.1当量)以及叔丁醇钠(172 mg,1.8 mmol,4.0当量)加入25 mL双颈瓶中,在氮气气氛下,抽换氮气三次,随后加入THF(4.0 mL),将反应液在40℃下搅拌过夜。点板监测,反应结束后,冷却至室温,加入氯化铵饱和溶液(5.0 mL)淬灭反应,用乙酸乙酯(15 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体13-3(23 mg,0.045 mmol,产率10%),LC-MS [M+H]+m/z: 507.3.The above oily compound and 2-chloro-8-methoxy-1,7-naphthyridine (87 mg, 0.45 mmol, 1.0 equivalent), BINAP (56 mg, 0.09 mmol, 0.2 equivalent), Pd 2 (dba) 3 (46 mg, 0.05 mmol, 0.1 equivalent) and sodium tert-butoxide (172 mg, 1.8 mmol, 4.0 equivalent) were added to a 25 mL double-necked flask, and nitrogen was replaced three times under a nitrogen atmosphere, and then THF (4.0 mL) was added, and the reaction solution was stirred at 40°C overnight. The reaction was monitored by spot plate. After the reaction was completed, it was cooled to room temperature, and a saturated ammonium chloride solution (5.0 mL) was added to quench the reaction. The above mixed solution was extracted three times with ethyl acetate (15 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 13-3 (23 mg, 0.045 mmol, yield 10%), LC-MS [M+H] + m/z: 507.3.
步骤4 中间体 13-4 的合成Step 4 Synthesis of intermediate 13-4
将中间体13-3(45 mg,0.09 mmol,1.0当量)溶于乙酸乙酯 (5.0 mL) 中,随后加入4M浓度的盐酸乙酸乙酯溶液(0.088 mL,0.36 mmol,4.0当量),40℃反应20h,随后,往反应液中加入乙醇 (5.0 mL) ,90℃回流20h。点板监测,原料反应完后,减压除去溶剂,得到的油状化合物直接进行下一步。Intermediate 13-3 (45 mg, 0.09 mmol, 1.0 equivalent) was dissolved in ethyl acetate (5.0 mL), and then a 4M hydrochloric acid ethyl acetate solution (0.088 mL, 0.36 mmol, 4.0 equivalent) was added, and the mixture was reacted at 40°C for 20 h. Then, ethanol (5.0 mL) was added to the reaction solution, and the mixture was refluxed at 90°C for 20 h. After the reaction of the raw material was completed, the solvent was removed under reduced pressure, and the obtained oily compound was directly used for the next step.
将上述油状化合物溶于三氯氧磷 (1.0 mL) 中,110℃反应1.5 h。点板监测,反应结束后,减压除去三氯氧磷,用EA稀释反应液,然后在0℃下用NaHCO3饱和水溶液调节至中性。用乙酸乙酯 (3.0 mL) 萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体13-4(40 mg,0.08 mmol,产率92%),LC-MS [M+H]+m/z: 483.3.The above oily compound was dissolved in phosphorus oxychloride (1.0 mL) and reacted at 110°C for 1.5 h. The reaction was monitored by spot plate. After the reaction was completed, the phosphorus oxychloride was removed under reduced pressure, the reaction solution was diluted with EA, and then adjusted to neutral with a saturated aqueous solution of NaHCO 3 at 0°C. The mixed solution was extracted three times with ethyl acetate (3.0 mL), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 13-4 (40 mg, 0.08 mmol, yield 92%), LC-MS [M+H] + m/z: 483.3.
步骤5 化合物 13 的合成Step 5 Synthesis of compound 13
将中间体13-4(40 mg,0.08 mmol,1.0当量)溶于四氢呋喃 (0.5 mL) 和甲醇(0.5 mL) 的混合溶剂中,随后加入2M氢氧化钠水溶液(0.20 mL,0.42 mmol,5.0当量),常温反应2h,点板监测,原料反应完后,用1M 盐酸水溶液中和反应,用二氯甲烷(10 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的油状化合物直接进行下一步反应。The intermediate 13-4 (40 mg, 0.08 mmol, 1.0 equivalent) was dissolved in a mixed solvent of tetrahydrofuran (0.5 mL) and methanol (0.5 mL), and then a 2M sodium hydroxide aqueous solution (0.20 mL, 0.42 mmol, 5.0 equivalent) was added. The reaction was carried out at room temperature for 2 h, and the plate was monitored. After the reaction of the raw material was completed, the reaction was neutralized with a 1M hydrochloric acid aqueous solution. The mixed solution was extracted three times with dichloromethane (10 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained oily compound was directly subjected to the next step of reaction.
将上述油状化合物溶于N, N-二甲基甲酰胺(1.0 mL)中, 按顺序加入化合物N,N-二异丙基乙胺(43 mg,0.33 mmol,4.0当量),HATU(63 mg,0.17 mmol,2.0当量)和3-氧杂环丁胺(12 mg,0.17 mmol,2.0当量),反应液在常温下反应1h。反应结束后,加入氯化铵饱和溶液(5.0 mL)淬灭反应,用乙酸乙酯(10 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到化合物13(24 mg,0.05 mmol,产率57%),为白色固体化合物. 1H NMR (500 MHz, CDCl3) δ 8.21(s, 2H), 8.01 (d, J = 5.0 Hz, 1H), 7.74 (d, J = 9.0 Hz, 1H), 7.33 (d, J = 5.0Hz, 1H), 6.79 (d, J = 9.0 Hz, 1H), 6.54-6.49 (m, 1H), 5.28 (d, J = 9.0 Hz,1H), 5.10 (brs, 1H), 5.04-4.95 (m, 1H), 4.92-4.84 (m, 2H), 4.46 (t, J = 6.5Hz, 1H), 4.42 (t, J = 6.5 Hz, 1H), 4.06-3.96 (m, 1H), 3.87-3.85 (m, 1H), 3.36(q, J = 7.0 Hz, 1H), 2.34 – 2.13 (m, 2H), 2.03 – 1.81 (m, 2H), 1.46 (d, J =7.0 Hz, 3H), 1.37 – 1.25 (m, 4H), 1.18 (d, J = 6.5 Hz, 3H).LC-MS [M+H]+m/z:510.3.The above oily compound was dissolved in N, N-dimethylformamide (1.0 mL), and N, N-diisopropylethylamine (43 mg, 0.33 mmol, 4.0 equivalents), HATU (63 mg, 0.17 mmol, 2.0 equivalents) and 3-oxetaneamine (12 mg, 0.17 mmol, 2.0 equivalents) were added in sequence, and the reaction solution was reacted at room temperature for 1 hour. After the reaction was completed, a saturated ammonium chloride solution (5.0 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (10 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain compound 13 (24 mg, 0.05 mmol, yield 57%) as a white solid compound. 1 H NMR (500 MHz, CDCl 3 ) δ 8.21 (s, 2H), 8.01 (d, J = 5.0 Hz, 1H), 7.74 (d, J = 9.0 Hz, 1H), 7.33 (d, J = 5.0Hz, 1H), 6.79 (d, J = 9.0 Hz, 1H), 6.54-6.49 (m, 1H), 5.28 (d, J = 9.0 Hz,1H), 5.10 (brs, 1H), 5.04-4.95 (m, 1H), 4.92-4.84 (m, 2H), 4.46 (t, J = 6.5Hz, 1H), 4.42 (t, J = 6.5 Hz, 1H), 4.06-3.96 (m, 1H), 3.87-3.85 (m, 1H), 3.36(q, J = 7.0 Hz, 1H), 2.34 – 2.13 (m, 2H), 2.03 – 1.81 ( m, 2H), 1.46 (d, J =7.0 Hz, 3H), 1.37 – 1.25 (m, 4H), 1.18 (d, J = 6.5 Hz, 3H).LC-MS [M+H] + m/z:510.3.
实施例14: (S)-8-氯-N-(4-(1-((5-(氧杂环丁烷-3-基氨基)甲基)嘧啶-2-基)氨基)乙基)环己基)-1,7-萘啶-2-胺 (化合物14)的合成Example 14: Synthesis of (S)-8-chloro-N-(4-(1-((5-(oxetane-3-ylamino)methyl)pyrimidin-2-yl)amino)ethyl)cyclohexyl)-1,7-naphthyridin-2-amine (Compound 14)
化合物14的制备:Preparation of compound 14:
步骤1 中间体 14-1 的合成Step 1 Synthesis of intermediate 14-1
将化合物2-氯嘧啶-5-羧酸甲酯(480 mg,2.0 mmol, 1.0当量)溶于二甲基亚砜(10 mL)中,向加入化合物1-3(340 mg, 2.0 mmol, 1.0当量),N, N-二异丙基乙胺(1290mg,10.0 mmol,5.0当量),得到的反应液于100 ℃反应12 h。反应结束后,冷却至室温,加入氯化铵饱和溶液(10 mL)淬灭反应,用乙酸乙酯(20 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体14-1(414 mg,1.1 mmol,产率55%)。LC-MS [M+H]+ m/z: 379.3。Compound 2-chloropyrimidine-5-carboxylic acid methyl ester (480 mg, 2.0 mmol, 1.0 equivalent) was dissolved in dimethyl sulfoxide (10 mL), and compound 1-3 (340 mg, 2.0 mmol, 1.0 equivalent) and N, N-diisopropylethylamine (1290 mg, 10.0 mmol, 5.0 equivalent) were added. The resulting reaction solution was reacted at 100 °C for 12 h. After the reaction was completed, the mixture was cooled to room temperature, and a saturated ammonium chloride solution (10 mL) was added to quench the reaction. The mixed solution was extracted three times with ethyl acetate (20 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The resulting mixture was separated and purified by silica gel column chromatography to obtain intermediate 14-1 (414 mg, 1.1 mmol, yield 55%). LC-MS [M+H]+ m/z: 379.3.
步骤2 中间体 14-2 的合成Step 2 Synthesis of intermediate 14-2
首先,将中间体14-1(414 mg,1.1 mmol,1.0当量)溶于5.0 mL甲醇中,加入4 M盐酸的1, 4-二氧六环溶液2.0 mL,室温反应3 h,点板监测,原料反应完后,旋干溶剂,得到的油状化合物直接进行下一步。First, the intermediate 14-1 (414 mg, 1.1 mmol, 1.0 equivalent) was dissolved in 5.0 mL of methanol, and 2.0 mL of 4 M hydrochloric acid in 1, 4-dioxane solution was added. The reaction was carried out at room temperature for 3 h and the plate was monitored. After the reaction of the raw material was completed, the solvent was dried and the obtained oily compound was directly used for the next step.
然后,将上述油状化合物和2-氯-8-甲氧基-1,7-萘啶(212 mg,1.09 mmol,1.0当量),BINAP(134 mg,0.22 mmol,0.2当量),Pd2(dba)3(100 mg,0.11 mmol,0.1当量)以及叔丁醇钠(415 mg,4.3 mmol,4.0当量)加入25 mL双颈瓶中,在氮气氛下,抽换气三次,随后加入四氢呋喃(10 mL),室温搅拌过夜。反应结束后,冷却至室温,加入饱和氯化铵水溶液(20mL)淬灭反应,用乙酸乙酯(15 mL)萃取三次,合并有机相,用无水硫酸钠干燥有机相,过滤,减压浓缩除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体14-2 (186mg,0.43 mmol,产率41%)。LC-MS [M+H]+ m/z: 437.3。Then, the above oily compound and 2-chloro-8-methoxy-1,7-naphthyridine (212 mg, 1.09 mmol, 1.0 equivalent), BINAP (134 mg, 0.22 mmol, 0.2 equivalent), Pd 2 (dba) 3 (100 mg, 0.11 mmol, 0.1 equivalent) and sodium tert-butoxide (415 mg, 4.3 mmol, 4.0 equivalent) were added to a 25 mL double-necked flask, evacuated three times under a nitrogen atmosphere, and then tetrahydrofuran (10 mL) was added and stirred at room temperature overnight. After the reaction was completed, it was cooled to room temperature, saturated aqueous ammonium chloride solution (20 mL) was added to quench the reaction, and it was extracted three times with ethyl acetate (15 mL), the organic phases were combined, the organic phases were dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to remove the solvent. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 14-2 (186 mg, 0.43 mmol, yield 41%). LC-MS [M+H]+ m/z: 437.3.
步骤3 中间体 14-3 的合成Step 3 Synthesis of intermediate 14-3
首先,将中间体14-2(186 mg,0.43 mmol,1.0当量)溶于5.0 mL乙酸乙酯中,随后加入4 M浓度的盐酸乙酸乙酯溶液(0.43 mL,1.72 mmol,4.0当量),40 ℃反应20 h,随后,向反应液中加入5.0 mL乙醇,90 ℃回流20 h。点板监测,原料反应完后,减压除去溶剂,得到的油状化合物直接进行下一步。First, the intermediate 14-2 (186 mg, 0.43 mmol, 1.0 equivalent) was dissolved in 5.0 mL of ethyl acetate, and then a 4 M hydrochloric acid ethyl acetate solution (0.43 mL, 1.72 mmol, 4.0 equivalent) was added, and the reaction was carried out at 40 °C for 20 h. Subsequently, 5.0 mL of ethanol was added to the reaction solution, and the reaction was refluxed at 90 °C for 20 h. After the reaction of the raw material was completed, the solvent was removed under reduced pressure, and the obtained oily compound was directly used for the next step.
然后,将上述油状化合物溶于2.0 mL三氯氧磷中,110 ℃反应1.5 h。点板监测,反应结束后,减压除去三氯氧磷,用乙酸乙酯(5.0 mL)稀释反应液,然后在0 ℃下用NaHCO3饱和水溶液调节至中性。用乙酸乙酯(5.0 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体14-3(47 mg,0.1 mmol,产率24%)。LC-MS [M+H]+ m/z: 455.3。Then, the above oily compound was dissolved in 2.0 mL of phosphorus oxychloride and reacted at 110 °C for 1.5 h. The reaction was monitored by spot plate. After the reaction was completed, the phosphorus oxychloride was removed under reduced pressure, the reaction solution was diluted with ethyl acetate (5.0 mL), and then adjusted to neutral with a saturated aqueous solution of NaHCO3 at 0 °C. The mixed solution was extracted three times with ethyl acetate (5.0 mL), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 14-3 (47 mg, 0.1 mmol, yield 24%). LC-MS [M+H]+ m/z: 455.3.
步骤4 化合物 14 的合成Step 4 Synthesis of compound 14
在氮气氛围下,将中间体14-3(47 mg,0.1 mmol,1.0当量)溶于1.0 mL四氢呋喃中,冷却至-78 ℃后,缓慢加入1.5 M的二异丁基氢化铝甲苯溶液(0.13 mL,0.2 mmol,2.0当量)。点板监测,反应结束后,加2 M氢氧化钠溶液(2.0 mL)淬灭反应,用乙酸乙酯(5.0mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的油状化合物直接进行下一步反应。在氮气保护下,将上述油状化合物(13 mg,0.03 mmol,1.0当量)和3-氧杂环丁胺(4.4 mg,0.06 mmol,2.0当量)溶于1.0 mL甲醇中,搅拌1 h。随后加入醋酸(1.8 mg,0.03 mmol,1.0当量)和氰基硼氢化钠(4 mg,0.06 mmol,2.0当量),室温反应过夜。点板监测反应,反应结束后,加水(5.0 mL)淬灭反应。用乙酸乙酯(5.0 mL)萃取上述混合溶液三次,将有机相收集,用无水硫酸钠干燥,随后通过旋转蒸发除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到14(1 mg,0.002 mmol,产率7%)。1H NMR (500MHz, CDCl3) δ 8.22 (s, 2H), 8.05 (d, J = 5.0Hz, 1H), 7.77 (d, J = 9.0 Hz,1H), 7.34 (d, J = 5.5 Hz, 1H), 6.80 (d, J = 9.0 Hz, 1H), 5.08 (d, J = 9.0 Hz,1H), 5.02 – 4.91 (m, 1H), 4.82 (t, J = 7.0 Hz, 2H), 4.45 (t, J = 6.5 Hz, 2H),4.08 – 3.97 (m, 2H), 3.57 (s, 2H), 2.33 – 2.26 (m, 2H), 2.00 – 1.95 (m, 1H),1.92 – 1.85 (m, 2H), 1.37 – 1.28 (m, 4H), 1.21 (d, J = 6.5 Hz, 3H). LC-MS [M+H]+ m/z: 468.3。Under nitrogen atmosphere, the intermediate 14-3 (47 mg, 0.1 mmol, 1.0 equivalent) was dissolved in 1.0 mL of tetrahydrofuran, cooled to -78 °C, and then 1.5 M diisobutylaluminum hydride toluene solution (0.13 mL, 0.2 mmol, 2.0 equivalent) was slowly added. The reaction was monitored by spot plate. After the reaction was completed, 2 M sodium hydroxide solution (2.0 mL) was added to quench the reaction. The mixed solution was extracted three times with ethyl acetate (5.0 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained oily compound was directly subjected to the next step of reaction. Under nitrogen protection, the above oily compound (13 mg, 0.03 mmol, 1.0 equivalent) and 3-oxetaneamine (4.4 mg, 0.06 mmol, 2.0 equivalent) were dissolved in 1.0 mL of methanol and stirred for 1 h. Then, acetic acid (1.8 mg, 0.03 mmol, 1.0 equivalent) and sodium cyanoborohydride (4 mg, 0.06 mmol, 2.0 equivalent) were added and reacted at room temperature overnight. The reaction was monitored by spot plate. After the reaction was completed, water (5.0 mL) was added to quench the reaction. The mixed solution was extracted three times with ethyl acetate (5.0 mL), and the organic phase was collected, dried over anhydrous sodium sulfate, and then the solvent was removed by rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain 14 (1 mg, 0.002 mmol, yield 7%). 1 H NMR (500MHz, CDCl 3 ) δ 8.22 (s, 2H), 8.05 (d, J = 5.0Hz, 1H), 7.77 (d, J = 9.0 Hz, 1H), 7.34 (d, J = 5.5 Hz, 1H), 6.80 (d, J = 9.0 Hz, 1H), 5.08 ( d, J = 9.0 Hz, 1H), 5.02 – 4.91 (m, 1H), 4.82 (t, J = 7.0 Hz, 2H), 4.45 (t, J = 6.5 Hz, 2H), 4.08 – 3.97 (m, 2H), 3.57 (s, 2H), 2.33 – 2.26 (m, 2H), 2.00 – 1.95 (m, 1H), 1.92 – 1.85 (m, 2H), 1.37 – 1.28 (m, 4H), 1.21 (d, J = 6.5 Hz, 3H). LC-MS [M+H]+ m/z: 468.3.
实施例15: (S)-2-(2-((1-(4-((8-氯-1,7-萘啶-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基甲基)乙酰胺(化合物15)的合成Example 15: Synthesis of (S)-2-(2-((1-(4-((8-chloro-1,7-naphthyridin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-N-(oxetan-3-ylmethyl)acetamide (Compound 15)
化合物15的制备:Preparation of compound 15:
将中间体8-2(6 mg,0.013 mmol,1.0当量)溶于四氢呋喃(0.5 mL)和甲醇(0.5mL)的混合溶剂中,随后加入2 M氢氧化钠水溶液(0.025 mL,0.05 mmol,4.0当量),室温反应2 h,点板监测,原料反应完后,用1 M 盐酸水溶液中和反应至中性,用二氯甲烷(10 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压除去溶剂。得到的油状化合物直接进行下一步反应。将上述油状化合物溶于N, N-二甲基甲酰胺(0.5 mL)中,按顺序加入化合物N, N-二异丙基乙胺(7 mg,0.052 mmol,4.0当量),HATU(10 mg,0.026 mmol,2.0当量)和3-氧杂环丁胺(2.3 mg,0.026 mmol,2.0当量),得到的反应液于室温下反应1h。反应结束后,加入氯化铵饱和溶液(1.0 mL)淬灭反应,用乙酸乙酯(5.0 mL)萃取上述混合溶液三次,合并有机相,饱和氯化钠溶液(5.0 mL)洗有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到15(3 mg,0.006 mmol,产率46%),为白色固体化合物。1H NMR (500 MHz, CD3OD) δ 8.08 (s, 2H), 7.81 (d, J =5.5 Hz, 1H), 7.72 (d, J = 9.0 Hz, 1H), 7.38 (d, J = 5.5 Hz, 1H), 6.85 (d, J =9.0 Hz, 1H), 4.80 – 4.75 (m, 1H), 4.67 (t, J = 6.0 Hz, 2H), 4.31 (t, J = 6.0Hz, 2H), 3.89 – 3.84 (m, 1H), 3.38 (d, J = 7.0 Hz, 2H), 3.24 (s, 2H), 3.10 –3.04 (m, 1H), 2.19 – 2.12 (m, 2H), 1.89 – 1.79 (m, 2H), 1.52 – 1.42 (m, 2H),1.26 – 1.22 (m, 3H), 1.10 (d, J = 6.5 Hz, 3H). LC-MS [M+H]+ m/z: 510.3。The intermediate 8-2 (6 mg, 0.013 mmol, 1.0 equivalent) was dissolved in a mixed solvent of tetrahydrofuran (0.5 mL) and methanol (0.5 mL), and then a 2 M sodium hydroxide aqueous solution (0.025 mL, 0.05 mmol, 4.0 equivalent) was added. The reaction was carried out at room temperature for 2 h, and the plate was monitored. After the reaction of the raw material was completed, the reaction was neutralized with a 1 M hydrochloric acid aqueous solution until it was neutral. The mixed solution was extracted three times with dichloromethane (10 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed under reduced pressure. The obtained oily compound was directly subjected to the next step of reaction. The above oily compound was dissolved in N, N-dimethylformamide (0.5 mL), and compounds N, N-diisopropylethylamine (7 mg, 0.052 mmol, 4.0 equivalents), HATU (10 mg, 0.026 mmol, 2.0 equivalents) and 3-oxetaneamine (2.3 mg, 0.026 mmol, 2.0 equivalents) were added in sequence, and the resulting reaction solution was reacted at room temperature for 1 h. After the reaction was completed, a saturated ammonium chloride solution (1.0 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (5.0 mL), the organic phases were combined, washed with a saturated sodium chloride solution (5.0 mL), dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The resulting mixture was separated and purified by silica gel column chromatography to obtain 15 (3 mg, 0.006 mmol, yield 46%) as a white solid compound. 1 H NMR (500 MHz, CD 3 OD) δ 8.08 (s, 2H), 7.81 (d, J =5.5 Hz, 1H), 7.72 (d, J = 9.0 Hz, 1H), 7.38 (d, J = 5.5 Hz, 1H), 6.85 (d, J =9.0 Hz, 1H), 4.8 0 – 4.75 (m, 1H), 4.67 (t, J = 6.0 Hz, 2H), 4.31 (t, J = 6.0Hz, 2H), 3.89 – 3.84 (m, 1H), 3.38 (d, J = 7.0 Hz, 2H), 3.24 (s, 2H), 3.10 –3.04 (m , 1H), 2.19 LC-MS [M+H] + m/z: 510.3.
实施例16: (S)-2-(2-((1-(4-((8-氯-1,6-萘啶-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物16)的合成Example 16: Synthesis of (S)-2-(2-((1-(4-((8-chloro-1,6-naphthyridin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 16)
参照实施例1中化合物1的制备过程,在步骤3中用2,8-二氯-[1,6]萘啶替换2,8-二氯-7-甲基喹啉,其他操作步骤类似,得到化合物16为白色固体。1H NMR (500 MHz,CDCl3) δ 8.71 (s, 1H), 8.56 (s, 1H), 8.19 (s, 2H), 7.94 – 7.78 (m, 1H), 6.77– 6.61 (m, 1H), 6.45 (d, J = 7.0 Hz, 1H), 5.15 (d, J = 9.0 Hz, 1H), 5.08 –5.01 (m, 1H), 4.92 (t, J = 7.0 Hz, 2H), 4.47 (t, J = 6.5 Hz, 2H), 4.08 – 3.98(m, 1H), 3.36 (s, 2H), 2.32 – 2.20 (m, 2H), 1.97 (d, J = 12.0 Hz, 1H), 1.88(d, J = 12.5 Hz, 1H), 1.54 – 1.46 (m, 1H), 1.36 – 1.24 (m, 4H), 1.20 (d, J =6.5 Hz, 3H).LC-MS [M+H]+ m/z: 496.3。Referring to the preparation process of compound 1 in Example 1, 2,8-dichloro-[1,6]naphthyridine was used to replace 2,8-dichloro-7-methylquinoline in step 3, and the other operation steps were similar to obtain compound 16 as a white solid. 1 H NMR (500 MHz, CDCl 3 ) δ 8.71 (s, 1H), 8.56 (s, 1H), 8.19 (s, 2H), 7.94 – 7.78 (m, 1H), 6.77– 6.61 (m, 1H), 6.45 (d, J = 7.0 Hz, 1H), 5.15 (d, J = 9.0 Hz, 1H), 5.08 –5.01 (m, 1H), 4.92 (t, J = 7.0 Hz, 2H), 4.47 (t, J = 6.5 Hz, 2H), 4.08 – 3.98(m, 1H), 3.36 (s, 2H), 2.32 – 2.20 (m , 2H), 1.97 (d, J = 12.0 Hz, 1H), 1.88 (d, J = 12.5 Hz, 1H), 1.54 – 1.46 (m, 1H), 1.36 – 1.24 (m, 4H), 1.20 (d, J =6.5 Hz, 3H). LC-MS [M+H] + m/z: 496.3.
实施例17: (S)-2-(6-((1-(4-((8-氯-1,6-萘啶-2-基)氨基)环己基)乙基)氨基)吡啶-3-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物17)的合成Example 17: Synthesis of (S)-2-(6-((1-(4-((8-chloro-1,6-naphthyridin-2-yl)amino)cyclohexyl)ethyl)amino)pyridin-3-yl)-N-(oxetane-3-yl)acetamide (Compound 17)
参照实施例12中化合物12的制备过程,在步骤1中用2,8-二氯-[1,6]萘啶替换2-氯-8-甲氧基-1,7-萘啶,无需进行步骤2反应,其他操作步骤类似,得到化合物17为白色固体。1H NMR (500 MHz, CDCl3) δ 8.71 (s, 1H), 8.56 (s, 1H), 8.19 (s, 2H), 7.94 –7.78 (m, 1H), 6.77 – 6.61 (m, 1H), 6.45 (d, J = 7.0 Hz, 1H), 5.15 (d, J = 9.0Hz, 1H), 5.08 – 5.01 (m, 1H), 4.92 (t, J = 7.0 Hz, 2H), 4.47 (t, J = 6.5 Hz,2H), 4.08 – 3.98 (m, 1H), 3.36 (s, 2H), 2.32 – 2.20 (m, 2H), 1.97 (d, J =12.0 Hz, 1H), 1.88 (d, J = 12.5 Hz, 1H), 1.54 – 1.46 (m, 1H), 1.36 – 1.24 (m,4H), 1.20 (d, J = 6.5 Hz, 3H).LC-MS [M+H]+ m/z: 496.3。Referring to the preparation process of compound 12 in Example 12, 2,8-dichloro-[1,6]naphthyridine was used to replace 2-chloro-8-methoxy-1,7-naphthyridine in step 1, and there was no need to carry out the reaction in step 2. The other operation steps were similar to obtain compound 17 as a white solid. 1 H NMR (500 MHz, CDCl 3 ) δ 8.71 (s, 1H), 8.56 (s, 1H), 8.19 (s, 2H), 7.94 –7.78 (m, 1H), 6.77 – 6.61 (m, 1H), 6.45 (d, J = 7.0 Hz, 1H), 5.15 ( d, J = 9.0Hz, 1H), 5.08 – 5.01 (m, 1H), 4.92 (t, J = 7.0 Hz, 2H), 4.47 (t, J = 6.5 Hz, 2H), 4.08 – 3.98 (m, 1H), 3.36 (s, 2H), 2.32 – 2.20 (m, 2H), 1.97 (d, J =12.0 Hz, 1H), 1.88 (d, J = 12.5 Hz, 1H), 1.54 – 1.46 (m, 1H), 1.36 – 1.24 (m,4H), 1.20 (d, J = 6.5 Hz, 3H). LC-MS [M+H] + m/z: 496.3.
实施例18: (S)-2-(6-((1-(4-((8-溴-1,6-萘啶-2-基)氨基)环己基)乙基)氨基)吡啶-3-基)-N-(氧杂环丁烷-3-基)乙酰胺(化合物18)的合成Example 18: Synthesis of (S)-2-(6-((1-(4-((8-bromo-1,6-naphthyridin-2-yl)amino)cyclohexyl)ethyl)amino)pyridin-3-yl)-N-(oxetane-3-yl)acetamide (Compound 18)
1 关键起始物料8-溴-2-氯-[1,6]萘啶的制备:1 Preparation of key starting material 8-bromo-2-chloro-[1,6]naphthyridine:
步骤A 中间体 18-1的合成Step A Synthesis of Intermediate 18-1
将3,5-二溴吡啶-4-胺(5.0 g,5.35 mmol,1.0当量)溶于N,N-二甲基甲酰胺(20mL)中,依次加入丙烯酸乙酯(4.0 g,40.00 mmol,2.0当量),N,N-二异丙基乙胺(7.7 g,60.0 mmol,3.0当量),三(邻甲基苯基)磷(1.2 g,4.0 mmol,0.2当量)和醋酸钯(448 mg,2.0 mmol,0.1当量)。反应混合物在氮气体系下,100℃搅拌10小时。监测反应,反应完成后,待反应液冷却至室温,将反应液倒入水(20 mL)中,乙酸乙酯(10 mL)萃取三次,合并有机相,并用饱和氯化钠溶液(10 mL)洗涤三次,无水硫酸钠干燥,过滤,减压浓缩得到粗品。粗品通过柱层析纯化,得到中间体18-1(1.76 g,6.5 mmol,产率:32 %)。LCMS [M+H]+ m/z:271.0。3,5-Dibromopyridin-4-amine (5.0 g, 5.35 mmol, 1.0 equivalent) was dissolved in N,N-dimethylformamide (20 mL), and ethyl acrylate (4.0 g, 40.00 mmol, 2.0 equivalent), N,N-diisopropylethylamine (7.7 g, 60.0 mmol, 3.0 equivalent), tri(o-methylphenyl)phosphine (1.2 g, 4.0 mmol, 0.2 equivalent) and palladium acetate (448 mg, 2.0 mmol, 0.1 equivalent) were added in sequence. The reaction mixture was stirred at 100 °C for 10 hours under nitrogen system. The reaction was monitored. After the reaction was completed, the reaction solution was cooled to room temperature, poured into water (20 mL), extracted three times with ethyl acetate (10 mL), and the organic phases were combined and washed three times with saturated sodium chloride solution (10 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a crude product. The crude product was purified by column chromatography to obtain intermediate 18-1 (1.76 g, 6.5 mmol, yield: 32 %). LCMS [M+H] + m/z: 271.0.
步骤B 中间体 18-2的合成Step B Synthesis of Intermediate 18-2
将18-1(271 mg,1.00 mmol,1.0当量)溶于醋酸(2.0 mL)中,向反应液中加入三丁基膦(202 mg,1.00 mmol,1.0当量)。反应混合物在110 ℃下搅拌1小时。LCMS监测反应完成后,待反应液冷却至室温,将反应液倒入水(10 mL)中,通过硅藻土过滤,滤饼用乙酸乙酯(10 mL)洗涤,收集滤饼并减压浓缩得到中间体18-2(100 mg,0.44 mmol,44%),为白色固体。LCMS [M+H]+ m/z: 224.9。18-1 (271 mg, 1.00 mmol, 1.0 equivalent) was dissolved in acetic acid (2.0 mL), and tributylphosphine (202 mg, 1.00 mmol, 1.0 equivalent) was added to the reaction solution. The reaction mixture was stirred at 110 °C for 1 hour. After the reaction was completed as monitored by LCMS, the reaction solution was cooled to room temperature, poured into water (10 mL), filtered through diatomaceous earth, and the filter cake was washed with ethyl acetate (10 mL). The filter cake was collected and concentrated under reduced pressure to obtain intermediate 18-2 (100 mg, 0.44 mmol, 44%) as a white solid. LCMS [M+H] + m/z: 224.9.
步骤C 中间体18-3的合成Step C Synthesis of Intermediate 18-3
将上述得到的中间体18-2溶于三氯氧磷(1.0 mL)中。反应混合物在110 ℃搅拌2小时。LCMS监测反应完成后,待反应液冷却至室温,将反应液减压浓缩,得到的混合物通过硅胶柱色谱层析分离纯化得到化合物18-3(95 mg,0.39 mmol,产率:89%)。 1H NMR (500MHz, CDCl3) δ 9.18 (s, 1H), 9.02 (s, 1H), 8.26 (d, J = 8.5 Hz, 1H), 7.60 (d,J = 8.5 Hz, 1H), LCMS [M+H]+ m/z: 242.9。The intermediate 18-2 obtained above was dissolved in phosphorus oxychloride (1.0 mL). The reaction mixture was stirred at 110 °C for 2 hours. After the reaction was completed by LCMS monitoring, the reaction solution was cooled to room temperature, the reaction solution was concentrated under reduced pressure, and the obtained mixture was separated and purified by silica gel column chromatography to obtain compound 18-3 (95 mg, 0.39 mmol, yield: 89%). 1 H NMR (500MHz, CDCl 3 ) δ 9.18 (s, 1H), 9.02 (s, 1H), 8.26 (d, J = 8.5 Hz, 1H), 7.60 (d,J = 8.5 Hz, 1H), LCMS [M+H] + m/z: 242.9.
2 化合物18的制备:2 Preparation of compound 18:
参照实施例1中化合物1的制备过程,在步骤3中用8-溴-2-氯-[1,6]萘啶替换2,8-二氯-7-甲基喹啉,其他操作步骤类似,得到化合物18为白色固体。1H NMR (500 MHz,DMSO-d6) δ 8.88 (d, J = 6.5 Hz, 1H), 8.80 (s, 1H), 8.66 (s, 1H), 8.17 (s,2H), 7.99 (d, J = 9.0 Hz, 1H), 7.85 (d, J = 7.0 Hz, 1H), 6.95 – 6.84 (m, 2H),4.87 – 4.79 (m, 1H), 4.76 (t, J = 6.5 Hz, 2H), 4.47 (t, J = 6.5 Hz, 2H), 4.04– 3.97 (m, 1H), 3.97 – 3.90 (m, 1H), 3.30 (s, 2H), 2.25 – 2.15 (m, 2H), 1.95– 1.86 (m, 2H), 1.56 – 1.47 (m, 1H), 1.31 – 1.25 (m, 2H), 1.25 – 1.18 (m,2H), 1.15 (d, J = 6.5 Hz, 3H).LC-MS [M+H]+ m/z: 540.3。Referring to the preparation process of compound 1 in Example 1, 8-bromo-2-chloro-[1,6]naphthyridine was used to replace 2,8-dichloro-7-methylquinoline in step 3, and the other operation steps were similar to obtain compound 18 as a white solid. 1 H NMR (500 MHz, DMSO-d 6 ) δ 8.88 (d, J = 6.5 Hz, 1H), 8.80 (s, 1H), 8.66 (s, 1H), 8.17 (s,2H), 7.99 (d, J = 9.0 Hz, 1H), 7.85 (d, J = 7.0 Hz, 1H ), 6.95 – 6.84 (m, 2H), 4.87 – 4.79 (m, 1H), 4.76 (t, J = 6.5 Hz, 2H), 4.47 (t, J = 6.5 Hz, 2H), 4.04 – 3.97 (m, 1H), 3.97 – 3.90 (m, 1H), 3. 30 (s, 2H), 2.25 – 2.15 (m, 2H), 1.95 – 1.86 (m, 2H), 1.56 – 1.47 (m, 1H), 1.31 – 1.25 (m, 2H), 1.25 – 1.18 (m,2H), 1.15 (d, J = 6.5 Hz, 3H).LC-MS [M+H ] + m/z: 540.3.
实施例19: (S)-2-(2-((1-(4-((7-氯-1,6-萘啶-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物19)的合成Example 19: Synthesis of (S)-2-(2-((1-(4-((7-chloro-1,6-naphthyridin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 19)
参照实施例1中化合物1的制备过程,在步骤3中用2,7-二氯-1,6-萘啶替换2,8-二氯-7-甲基喹啉,其他操作步骤类似,得到化合物19为白色固体。1H NMR (500 MHz, CDCl3)δ 8.59 (s, 1H), 8.20 (s, 2H), 7.79 (d, J = 8.5 Hz, 1H), 7.46 (s, 1H), 6.60(d, J = 7.5 Hz, 1H), 6.38 (s, 1H), 5.12 (d, J = 9.0 Hz, 1H), 5.07 – 5.02 (m,1H), 4.91 (t, J = 7.0 Hz, 2H), 4.49 (t, J = 6.5 Hz, 2H), 4.06 – 3.99 (m, 1H),3.36 (s, 2H), 2.25 – 2.17 (m, 2H), 1.99 – 1.92 (m, 2H), 1.89 – 1.84 (m, 2H),1.46 – 1.42 (m, 4H), 1.20 (d, J = 6.5 Hz, 3H).LC-MS [M+H]+ m/z: 496.3。Referring to the preparation process of compound 1 in Example 1, 2,7-dichloro-1,6-naphthyridine was used to replace 2,8-dichloro-7-methylquinoline in step 3, and the other operation steps were similar to obtain compound 19 as a white solid. 1 H NMR (500 MHz, CDCl 3 )δ 8.59 (s, 1H), 8.20 (s, 2H), 7.79 (d, J = 8.5 Hz, 1H), 7.46 (s, 1H), 6.60 (d, J = 7.5 Hz, 1H), 6.38 (s, 1H), 5.12 (d, J = 9.0 Hz, 1H), 5.07 – 5.02 (m,1H), 4.91 (t, J = 7.0 Hz, 2H), 4.49 (t, J = 6.5 Hz, 2H), 4.06 – 3.99 (m, 1H), 3.36 (s, 2H), 2.25 – 2.17 (m, 2H) , 1.99 – 1.92 (m, 2H), 1.89 – 1.84 (m, 2H), 1.46 – 1.42 (m, 4H), 1.20 (d, J = 6.5 Hz, 3H). LC-MS [M+H] + m/z: 496.3.
实施例20: (S)-2-(2-((1-(4-((7-甲基-1,6-萘啶-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物20)的合成Example 20: Synthesis of (S)-2-(2-((1-(4-((7-methyl-1,6-naphthyridin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 20)
关键起始物料2-氯-7-甲基-1,6-萘啶(20-3)的制备:Preparation of key starting material 2-chloro-7-methyl-1,6-naphthyridine (20-3):
步骤A 中间体20-1的合成Step A Synthesis of Intermediate 20-1
将5-溴-2-甲基吡啶-4-胺(1.0 g,5.35 mmol)溶于N,N-二甲基甲酰胺(20 mL)中,依次加入丙烯酸乙酯(1.1 g,10.70 mmol,2.0当量),N,N-二异丙基乙胺(2.1 g,16.0mmol,3.0当量),三(邻甲基苯基)磷(325 mg,1.07 mmol,0.2当量)和醋酸钯(120 mg,0.54mmol)。反应混合物在氮气体系下,120℃搅拌10小时。监测反应,反应完成后,待反应液冷却至室温,将反应液倒入水(20 mL)中,乙酸乙酯(10 mL)萃取三次,合并有机相,并用饱和氯化钠溶液(10 mL)洗涤三次,无水硫酸钠干燥,过滤,减压浓缩得到粗品。粗品通过柱层析纯化,得到中间体20-1(850 mg,4.1 mmol,产率:77 %)。1H NMR (500 MHz, CDCl3) δ 8.34(s, 1H), 7.69 (d, J = 16.0 Hz, 1H), 6.44 (s, 1H), 6.36 (d, J = 16.0 Hz, 1H),4.49 (s, 2H), 4.27 (q, J = 7.0 Hz, 2H), 2.43 (s, 3H), 1.34 (t, J = 7.0 Hz,3H).5-Bromo-2-methylpyridin-4-amine (1.0 g, 5.35 mmol) was dissolved in N,N-dimethylformamide (20 mL), and ethyl acrylate (1.1 g, 10.70 mmol, 2.0 equivalents), N,N-diisopropylethylamine (2.1 g, 16.0 mmol, 3.0 equivalents), tri(o-methylphenyl)phosphine (325 mg, 1.07 mmol, 0.2 equivalents) and palladium acetate (120 mg, 0.54 mmol) were added in sequence. The reaction mixture was stirred at 120 °C for 10 hours under a nitrogen system. The reaction was monitored. After the reaction was completed, the reaction solution was cooled to room temperature, poured into water (20 mL), extracted three times with ethyl acetate (10 mL), and the organic phases were combined and washed three times with saturated sodium chloride solution (10 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a crude product. The crude product was purified by column chromatography to give intermediate 20-1 (850 mg, 4.1 mmol, yield: 77%). 1 H NMR (500 MHz, CDCl 3 ) δ 8.34 (s, 1H), 7.69 (d, J = 16.0 Hz, 1H), 6.44 (s, 1H), 6.36 (d, J = 16.0 Hz, 1H), 4.49 (s, 2H), 4.27 (q, J = 7.0 Hz, 2H), 2.43 (s, 3H), 1.34 (t, J = 7.0 Hz, 3H).
步骤B 中间体20-2的合成Step B Synthesis of Intermediate 20-2
将20-1(778 mg,3.76 mmol,1.0当量)溶于醋酸(15 mL)中,向反应液中加入三丁基膦(760 mg,3.76 mmol,1.0当量)。反应混合物在110 ℃下搅拌1小时。LCMS监测反应完成后,待反应液冷却至室温,将反应液倒入水(10 mL)中,通过硅藻土过滤,滤饼用乙酸乙酯(20 mL)洗涤,收集滤饼并减压浓缩得到中间体20-2,为白色固体。LCMS [M+H]+ m/z:161.0。20-1 (778 mg, 3.76 mmol, 1.0 equivalent) was dissolved in acetic acid (15 mL), and tributylphosphine (760 mg, 3.76 mmol, 1.0 equivalent) was added to the reaction solution. The reaction mixture was stirred at 110 °C for 1 hour. After the reaction was completed as monitored by LCMS, the reaction solution was cooled to room temperature, poured into water (10 mL), filtered through diatomaceous earth, and the filter cake was washed with ethyl acetate (20 mL). The filter cake was collected and concentrated under reduced pressure to obtain intermediate 20-2 as a white solid. LCMS [M+H] + m/z: 161.0.
步骤C 关键起始物料20-3的合成Step C Synthesis of key starting material 20-3
将上述得到的中间体20-2溶于三氯氧磷(4.0 mL)中。反应混合物在110 ℃搅拌2小时。LCMS监测反应完成后,待反应液冷却至室温,将反减压浓缩,得到的混合物通过硅胶柱色谱层析分离纯化得到化合物20-3(580 mg,3.24 mmol,产率86%)。1H NMR (500 MHz,CDCl3) δ 9.18 (s, 1H), 8.19 (d, J = 8.5 Hz, 1H), 7.66 (s, 1H), 7.44 (d, J =8.5 Hz, 1H).LCMS [M+H]+ m/z: 179.0。The intermediate 20-2 obtained above was dissolved in phosphorus oxychloride (4.0 mL). The reaction mixture was stirred at 110 °C for 2 hours. After the reaction was completed by LCMS monitoring, the reaction solution was cooled to room temperature, concentrated under reduced pressure, and the obtained mixture was separated and purified by silica gel column chromatography to obtain compound 20-3 (580 mg, 3.24 mmol, yield 86%). 1 H NMR (500 MHz,CDCl 3 ) δ 9.18 (s, 1H), 8.19 (d, J = 8.5 Hz, 1H), 7.66 (s, 1H), 7.44 (d, J =8.5 Hz, 1H).LCMS [M+H] + m/z: 179.0.
2 化合物20的制备:2 Preparation of compound 20:
参照实施例1中化合物1的制备过程,在步骤3中用2-氯-7-甲基-1,6-萘啶替换2,8-二氯-7-甲基喹啉,其他操作步骤类似,得到化合物20为白色固体。LC-MS [M+H]+ m/z:476.3。Referring to the preparation process of compound 1 in Example 1, 2-chloro-7-methyl-1,6-naphthyridine was used to replace 2,8-dichloro-7-methylquinoline in step 3, and the other operation steps were similar to obtain compound 20 as a white solid. LC-MS [M+H] + m/z: 476.3.
实施例21: (S)-2-(2-((1-(4-((5-氯吡啶并[4,3-d]嘧啶-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物21)的合成Example 21: Synthesis of (S)-2-(2-((1-(4-((5-chloropyrido[4,3-d]pyrimidin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 21)
1 关键起始物料5-氯-2-(甲磺酰基)吡啶并[4,3-d]嘧啶的制备1 Preparation of the key starting material 5-chloro-2-(methylsulfonyl)pyrido[4,3-d]pyrimidine
步骤A 中间体21-1的合成Step A Synthesis of Intermediate 21-1
将硫代氨基甲酰亚胺甲酯(10.0 g, 53.10 mmol)溶于N,N-二甲基甲酰胺(100mL)中,依次加入醋酸钠(8.7 g, 106.00 mmol)和(Z)-2-(乙氧基亚甲基)-3-氧代丁酸乙酯(19.8 g, 106.00 mmol)。反应混合物在80 ℃下搅拌4小时。LCMS监测反应完成后,待反应液冷却至室温,将反应液倒入水(50 mL)中,用滤纸过滤,水(30 mL)洗涤滤饼,收集滤饼减压浓缩得中间体21-1(11.2 g,粗品),没有进一步纯化直接用于下一步。LC-MS [M+H]+ m/z: 213.3。Methyl thiocarbamide (10.0 g, 53.10 mmol) was dissolved in N,N-dimethylformamide (100 mL), and sodium acetate (8.7 g, 106.00 mmol) and ethyl (Z)-2-(ethoxymethylene)-3-oxobutanoate (19.8 g, 106.00 mmol) were added in sequence. The reaction mixture was stirred at 80 °C for 4 hours. After the reaction was completed as monitored by LCMS, the reaction solution was cooled to room temperature, poured into water (50 mL), filtered with filter paper, and the filter cake was washed with water (30 mL). The filter cake was collected and concentrated under reduced pressure to obtain intermediate 21-1 (11.2 g, crude product), which was used directly in the next step without further purification. LC-MS [M+H] + m/z: 213.3.
步骤B 中间体 21-2 的合成Step B Synthesis of Intermediate 21-2
将21-1(11.2 g, 52.80 mmol)溶于N,N-二甲基甲酰胺(110 mL)中,向反应混合物中加入N,N-二甲基甲酰胺二甲基缩醛(14.1 mL, 106.00 mmol)。反应混合物在150 ℃下搅拌3小时。LCMS监测反应完成后,待反应液冷却至室温,将反应液倒入水(200 mL)中,乙酸乙酯(100 mL)萃取三次,合并有机相,并用饱和食盐水(50 mL)洗涤三次,无水硫酸钠干燥,过滤减压浓缩得到中间体21-2(14.0 g,粗品),没有进一步纯化直接用于下一步。LC-MS [M+H]+ m/z: 268.3 。21-1 (11.2 g, 52.80 mmol) was dissolved in N,N-dimethylformamide (110 mL), and N,N-dimethylformamide dimethyl acetal (14.1 mL, 106.00 mmol) was added to the reaction mixture. The reaction mixture was stirred at 150 °C for 3 hours. After the reaction was completed by LCMS monitoring, the reaction solution was cooled to room temperature, poured into water (200 mL), extracted three times with ethyl acetate (100 mL), and the organic phases were combined and washed three times with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to obtain intermediate 21-2 (14.0 g, crude product), which was used directly in the next step without further purification. LC-MS [M+H] + m/z: 268.3 .
步骤C 中间体 21-3 的合成Step C Synthesis of Intermediate 21-3
将21-2(14.0 g, 52.40 mmol)溶于乙醇(100 mL)中,依次加入乙酸铵(12.1 g,157.00 mmol)和氢氧化铵(14.0 mL, 360.00 mmol)。反应混合物在100 ℃下搅拌12小时。LCMS监测反应完成后,待反应液冷却至室温,将反应液通过滤纸过滤,滤饼用乙醇(100 mL)洗涤,收集固体并减压浓缩得到中间体21-3(7.5 g,粗品),没有进一步纯化直接用于下一步。LC-MS [M+H]+ m/z:194.2。Dissolve 21-2 (14.0 g, 52.40 mmol) in ethanol (100 mL), and add ammonium acetate (12.1 g, 157.00 mmol) and ammonium hydroxide (14.0 mL, 360.00 mmol) in turn. The reaction mixture was stirred at 100 °C for 12 hours. After the reaction was completed by LCMS monitoring, the reaction solution was cooled to room temperature, filtered through filter paper, and the filter cake was washed with ethanol (100 mL). The solid was collected and concentrated under reduced pressure to obtain intermediate 21-3 (7.5 g, crude product), which was used directly in the next step without further purification. LC-MS [M+H] + m/z: 194.2.
步骤D 中间体 21-4 的合成Step D Synthesis of Intermediate 21-4
将21-3(4.0 g, 20.70 mmol)溶于三氯氧磷(19.3 mL, 207.0 mmol)中。反应混合物在80 ℃下搅拌2小时。LCMS监测反应完成后,将反应液减压浓缩得到粗品。粗品通过柱层析(二氧化硅,石油醚:乙酸乙酯 = 10:1至2:1)纯化,得到中间体21-4(3.5 g,产率:80 %),为黄色固体。LC-MS [M+H]+ m/z: 212.1。21-3 (4.0 g, 20.70 mmol) was dissolved in phosphorus oxychloride (19.3 mL, 207.0 mmol). The reaction mixture was stirred at 80 °C for 2 hours. After the reaction was completed as monitored by LCMS, the reaction solution was concentrated under reduced pressure to obtain a crude product. The crude product was purified by column chromatography (silica, petroleum ether: ethyl acetate = 10:1 to 2:1) to obtain intermediate 21-4 (3.5 g, yield: 80 %) as a yellow solid. LC-MS [M+H] + m/z: 212.1.
步骤E 关键起始物料 21-5 的合成Step E Synthesis of key starting material 21-5
将21-4(2.5 g, 11.81 mmol)溶于二氯甲烷(250 mL)中,在冰浴条件下向反应混合物中间氯过氧苯甲酸(5.8 g, 28.30 mmol)。反应混合物在25 ℃下搅拌2小时。LCMS监测反应完成后,将反应液倒入水(200 mL)中,二氯甲烷(100 mL)萃取三次,合并有机相,并用饱和食盐水(50 mL)洗涤两次,无水硫酸钠干燥,过滤减压浓缩得到粗品。粗品通过柱层析(二氧化硅,石油醚:乙酸乙酯 = 10:1至1:1)纯化,得到化合物21-5(1.0 g,产率:35 %),为白色固体。1H NMR (400 MHz,CDCl3) δ 10.00 (s, 1H), 8.86 (d, J = 5.9 Hz, 1H),8.01 (dd, J = 5.9, 0.8 Hz, 1H), 3.49 (s, 3H). LC-MS [M+H]+ m/z: 244.0。21-4 (2.5 g, 11.81 mmol) was dissolved in dichloromethane (250 mL), and chloroperoxybenzoic acid (5.8 g, 28.30 mmol) was added to the reaction mixture under ice bath conditions. The reaction mixture was stirred at 25 °C for 2 hours. After the reaction was completed by LCMS monitoring, the reaction solution was poured into water (200 mL), extracted three times with dichloromethane (100 mL), the organic phases were combined, washed twice with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to obtain a crude product. The crude product was purified by column chromatography (silica, petroleum ether: ethyl acetate = 10:1 to 1:1) to obtain compound 21-5 (1.0 g, yield: 35%) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 10.00 (s, 1H), 8.86 (d, J = 5.9 Hz, 1H), 8.01 (dd, J = 5.9, 0.8 Hz, 1H), 3.49 (s, 3H). LC-MS [M+H] + m/z: 244.0.
2 化合物21的制备:2 Preparation of compound 21:
参照实施例1中化合物1的制备过程,在步骤3中用5-氯-2-(甲磺酰基)吡啶并[4,3-d]嘧啶替换2,8-二氯-7-甲基喹啉,其他操作步骤类似,得到化合物21为白色固体。1HNMR (500 MHz, CDCl3) δ 9.32 (brs, 1H), 8.30 (d, J = 6.0 Hz, 1H), 8.20 (s,2H), 7.28 (d, J = 6.0 Hz, 1H), 6.14 (d, J = 6.0 Hz, 1H), 5.61 (d, J = 8.0 Hz,1H), 5.19 – 5.11 (m, 1H), 5.09 – 5.01 (m, 1H), 4.92 (t, J = 6.5 Hz, 2H), 4.47(t, J = 6.5 Hz, 2H), 4.07 – 4.01 (m, 1H), 3.99 – 3.89 (m, 1H), 3.36 (s, 2H),2.28 – 2.19 (m, 2H), 1.99 – 1.94 (m, 1H), 1.91 – 1.86 (m, 1H), 1.55 – 1.49(m, 1H), 1.37 – 1.30 (m, 2H), 1.21 (d, J = 6.5 Hz, 3H).LC-MS [M+H]+ m/z:497.3。Referring to the preparation process of compound 1 in Example 1, 2,8-dichloro-7-methylquinoline was replaced with 5-chloro-2-(methylsulfonyl)pyrido[4,3-d]pyrimidine in step 3, and the other operation steps were similar to obtain compound 21 as a white solid. 1 HNMR (500 MHz, CDCl 3 ) δ 9.32 (brs, 1H), 8.30 (d, J = 6.0 Hz, 1H), 8.20 (s,2H), 7.28 (d, J = 6.0 Hz, 1H), 6.14 (d, J = 6.0 Hz, 1H), 5.61 (d, J = 8 .0 Hz,1H), 5.19 – 5.11 (m, 1H), 5.09 – 5.01 (m, 1H), 4.92 (t, J = 6.5 Hz, 2H), 4.47(t, J = 6.5 Hz, 2H), 4.07 – 4.01 (m, 1H), 3.99 – 3.89 (m, 1H), 3.36 (s, 2H), 2.28 – 2.19 (m, 2H), 1.99 – 1.94 (m, 1H), 1.91 – 1.86 (m, 1H), 1.55 – 1.49 (m, 1H), 1.37 – 1.30 (m, 2H), 1.21 (d, J = 6.5 Hz, 3H) .LC-MS [M+H] + m/z:497.3.
实施例22: (S)-2-(2-((1-(1-(1-(8-氯喹啉-2-基)甲基)哌啶-4-基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物22)的合成Example 22: Synthesis of (S)-2-(2-((1-(1-(1-(8-chloroquinolin-2-yl)methyl)piperidin-4-yl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 22)
化合物22的制备:Preparation of compound 22:
步骤1:中间体22-1的制备Step 1: Preparation of intermediate 22-1
将化合物(S)-4-(1-氨基乙基)哌啶-1-羧酸叔丁酯(0.46g, 2.0 mmol, 1.0当量)溶于二甲基亚砜(10 mL)中,加入化合物1-4(0.42 g,2.0 mmol,1.0 当量),DIPEA(1.29 g,10.0 mmol,5.0 当量),得到的反应液于100℃下反应12h。反应结束后,冷却至室温,加入氯化铵饱和溶液(10 mL)淬灭反应,用乙酸乙酯(20 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体22-1(0.67g,1.6 mmol,产率80%),淡黄色固体。LCMS [M+H]+ m/z: 421.3。Compound (S)-4-(1-aminoethyl)piperidine-1-carboxylic acid tert-butyl ester (0.46 g, 2.0 mmol, 1.0 equivalent) was dissolved in dimethyl sulfoxide (10 mL), and compound 1-4 (0.42 g, 2.0 mmol, 1.0 equivalent) and DIPEA (1.29 g, 10.0 mmol, 5.0 equivalent) were added. The resulting reaction solution was reacted at 100°C for 12 h. After the reaction was completed, the mixture was cooled to room temperature, saturated ammonium chloride solution (10 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (20 mL). The organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The resulting mixture was separated and purified by silica gel column chromatography to obtain intermediate 22-1 (0.67 g, 1.6 mmol, yield 80%) as a light yellow solid. LCMS [M+H] + m/z: 421.3.
步骤2:中间体22-2的制备Step 2: Preparation of intermediate 22-2
将上述中间体22-1(0.67g,1.6 mmol,1.0当量)溶于甲醇(5.0 mL)中,向该反应液中滴加盐酸的乙酸乙酯溶液(4.0M,1.2 mL, 3.0当量),点板检测。反应结束后,减压旋蒸得到白色固体。将该白色固体溶于1,2-二氯乙烷(5.0 mL)中,依次加入化合物8-氯喹啉-2-甲醛(0.31g,1.6 mmol,1.0当量),三乙酰氧基硼氢化钠 (0.68g,3.2 mmol,2.0当量),点板检测。反应结束后,加入氯化铵饱和溶液(5.0 mL)淬灭反应,用乙酸乙酯(10.0 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到中间体22-2(0.46 g,0.93 mmol,产率58%),淡黄色固体。LCMS [M+H]+ m/z: 496.3。The intermediate 22-1 (0.67 g, 1.6 mmol, 1.0 equivalent) was dissolved in methanol (5.0 mL), and a hydrochloric acid ethyl acetate solution (4.0 M, 1.2 mL, 3.0 equivalent) was added dropwise to the reaction solution, and the plate was tested. After the reaction was completed, a white solid was obtained by rotary evaporation under reduced pressure. The white solid was dissolved in 1,2-dichloroethane (5.0 mL), and compound 8-chloroquinoline-2-carboxaldehyde (0.31 g, 1.6 mmol, 1.0 equivalent) and sodium triacetoxyborohydride (0.68 g, 3.2 mmol, 2.0 equivalent) were added in sequence, and the plate was tested. After the reaction was completed, a saturated ammonium chloride solution (5.0 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (10.0 mL), and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by rotary evaporation under reduced pressure. The obtained mixture was separated and purified by silica gel column chromatography to obtain intermediate 22-2 (0.46 g, 0.93 mmol, yield 58%) as a light yellow solid. LCMS [M+H] + m/z: 496.3.
步骤3:目标化合物22的合成Step 3: Synthesis of target compound 22
将中间体22-2(40 mg,0.08 mmol ,1.0当量)溶于四氢呋喃(1.0 mL)和甲醇(1.0mL)的混合溶剂中,加入2 M氢氧化钠水溶液(0.16 mL,0.32 mmol,4.0当量),常温反应2h,点板监测,原料反应完后,1M 盐酸水溶液中和反应至中性,二氯甲烷(5.0 mL)萃取上述混合溶液三次,合并有机相,无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的油状化合物直接进行下一步反应。The intermediate 22-2 (40 mg, 0.08 mmol, 1.0 equivalent) was dissolved in a mixed solvent of tetrahydrofuran (1.0 mL) and methanol (1.0 mL), and a 2 M sodium hydroxide aqueous solution (0.16 mL, 0.32 mmol, 4.0 equivalent) was added. The reaction was carried out at room temperature for 2 h, and the plate was monitored. After the reaction of the raw material was completed, the reaction was neutralized with a 1 M hydrochloric acid aqueous solution until it was neutral. The mixed solution was extracted with dichloromethane (5.0 mL) three times, and the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained oily compound was directly subjected to the next step of reaction.
将上述油状化合物溶于N,N-二甲基甲酰胺(1.0 mL)中,按顺序加入化合物N,N-二异丙基乙胺(41 mg,0.32 mmol,4.0当量),HATU(61 mg,0.16 mmol,2.0当量)和3-氧杂环丁胺(12 mg,0.16 mmol,2.0当量),得到的反应液于室温下反应1h。反应结束后,加入饱和氯化铵水溶液(2.0 mL)淬灭反应,乙酸乙酯(10 mL)萃取上述混合溶液三次,合并有机相,饱和氯化钠溶液洗涤三次,有机相经无水硫酸钠干燥,过滤,减压旋蒸除去溶剂。得到的混合物通过硅胶柱色谱层析分离纯化得到化合物22(8 mg,0.016 mmol,产率20%)为白色固体。1H NMR (500 MHz, CD3OD) δ 8.33 (d, J = 8.4 Hz, 1H), 8.09 (s, 2H), 7.82 (d, J =7.9 Hz, 2H), 7.51-7.45 (m, 2H), 4.87-4.70 (m, 5H), 4.55 (s, 2H), 4.45 (t, J =6.2 Hz, 2H), 3.98-3.91 (m, 1H), 3.74 (d, J = 11.8 Hz, 2H), 3.11-3.02 (m, 2H),2.04-1.96 (m, 2H), 1.82-1.72 (m, 1H), 1.70-1.55 (m, 2H), 1.12 (d, J = 7.9 Hz,3H). LCMS [M+H]+ m/z: 495.3。The above oily compound was dissolved in N,N-dimethylformamide (1.0 mL), and compound N,N-diisopropylethylamine (41 mg, 0.32 mmol, 4.0 equivalents), HATU (61 mg, 0.16 mmol, 2.0 equivalents) and 3-oxetaneamine (12 mg, 0.16 mmol, 2.0 equivalents) were added in sequence, and the resulting reaction solution was reacted at room temperature for 1 h. After the reaction was completed, saturated aqueous ammonium chloride solution (2.0 mL) was added to quench the reaction, and the mixed solution was extracted three times with ethyl acetate (10 mL), the organic phases were combined, and the saturated sodium chloride solution was washed three times. The organic phase was dried over anhydrous sodium sulfate, filtered, and the solvent was removed by vacuum rotary evaporation. The obtained mixture was separated and purified by silica gel column chromatography to obtain compound 22 (8 mg, 0.016 mmol, yield 20%) as a white solid. 1 H NMR (500 MHz, CD 3 OD) δ 8.33 (d, J = 8.4 Hz, 1H), 8.09 (s, 2H), 7.82 (d, J =7.9 Hz, 2H), 7.51-7.45 (m, 2H), 4.87-4.70 (m, 5H), 4.55 (s, 2H ), 4.45 (t, J =6.2 Hz, 2H), 3.98-3.91 (m, 1H), 3.74 (d, J = 11.8 Hz, 2H), 3.11-3.02 (m, 2H), 2.04-1.96 (m, 2H), 1.82-1.72 (m, 1H), 1.70-1 .55 (m, 2H), 1.12 (d, J = 7.9 Hz, 3H). LCMS [M+H] + m/z: 495.3.
实施例23: (R)-2-(2-((1-(1-((8-氯喹啉-2-基)甲基)哌啶-4-基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物23)的合成Example 23: Synthesis of (R)-2-(2-((1-(1-((8-chloroquinolin-2-yl)methyl)piperidin-4-yl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 23)
利用化合物(R)-4-(1-氨基乙基)哌啶-1-羧酸叔丁酯代替对映异构体(S)-4-(1-氨基乙基)哌啶-1-羧酸叔丁酯,参照实施例22的操作进行,得到目标化合物23为白色固体。1H NMR (500 MHz, CD3OD) δ 8.33 (d, J = 8.4 Hz, 1H), 8.09 (s, 2H), 7.82 (d, J= 7.9 Hz, 2H), 7.51-7.45 (m, 2H), 4.87-4.70 (m, 5H), 4.55 (s, 2H), 4.45 (t, J= 6.2 Hz, 2H), 3.98-3.91 (m, 1H), 3.74 (d, J = 11.8 Hz, 2H), 3.11-3.02 (m,2H), 2.04-1.96 (m, 2H), 1.82-1.72 (m, 1H), 1.70-1.55 (m, 2H), 1.12 (d, J =6.5 Hz, 3H). LCMS [M+H]+ m/z: 495.3。Using compound (R)-4-(1-aminoethyl)piperidine-1-carboxylic acid tert-butyl ester instead of the enantiomer (S)-4-(1-aminoethyl)piperidine-1-carboxylic acid tert-butyl ester, refer to the procedures of Example 22 to obtain the target compound 23 as a white solid. 1 H NMR (500 MHz, CD 3 OD) δ 8.33 (d, J = 8.4 Hz, 1H), 8.09 (s, 2H), 7.82 (d, J= 7.9 Hz, 2H), 7.51-7.45 (m, 2H), 4.87-4.70 (m, 5H), 4.55 (s, 2 H), 4.45 (t, J= 6.2 Hz, 2H), 3.98-3.91 (m, 1H), 3.74 (d, J = 11.8 Hz, 2H), 3.11-3.02 (m,2H), 2.04-1.96 (m, 2H), 1.82-1.72 (m, 1H), 1.70 -1.55 (m, 2H), 1.12 (d, J =6.5 Hz, 3H). LCMS [M+H] + m/z: 495.3.
实施例24: (S)-2-(2-((1-(1-((8-溴喹啉-2-基)甲基)哌啶-4-基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物24)的合成Example 24: Synthesis of (S)-2-(2-((1-(1-((8-bromoquinolin-2-yl)methyl)piperidin-4-yl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 24)
利用化合物8-溴-2-喹啉甲醛替换化合物8-氯喹啉-2-甲醛,参照实施例22的操作进行,得到目标化合物24为白色固体。1H NMR (500 MHz, DMSO-d6) δ 8.75 (d, J = 6.5Hz, 1H), 8.37 (brs, 1H), 8.07 (dd, J = 7.5, 1.0 Hz, 1H),8.06 (s, 2H), 7.96(d, J = 8.9 Hz, 1H), 7.63 (d, J = 8.5 Hz, 1H), 7.45 (d, J = 8.9 Hz, 1H),6.85-6.83 (m, 1H), 4.72-4.64 (m, 1H), 4.63 (t, J = 7.1 Hz, 2H), 4.34 (t, J =6.5 Hz, 2H), 4.56 (s, 2H), 4.46 (t, J = 6.2 Hz, 2H), 3.97-3.90 (m, 1H), 3.73(d, J = 11.8 Hz, 2H), 3.10-3.00 (m, 2H), 2.05-1.94 (m, 2H), 1.83-1.73 (m,1H), 1.71-1.53 (m, 2H), 1.02 (d, J = 6.5 Hz, 3H). LCMS [M+H]+ m/z: 539.2The compound 8-bromo-2-quinolinecarboxaldehyde was used to replace the compound 8-chloroquinoline-2-carboxaldehyde, and the operation of Example 22 was referred to to obtain the target compound 24 as a white solid. 1 H NMR (500 MHz, DMSO-d 6 ) δ 8.75 (d, J = 6.5Hz, 1H), 8.37 (brs, 1H), 8.07 (dd, J = 7.5, 1.0 Hz, 1H), 8.06 (s, 2H), 7.96 (d, J = 8.9 Hz, 1H), 7.63 ( d, J = 8.5 Hz, 1H), 7.45 (d, J = 8.9 Hz, 1H), 6.85-6.83 (m, 1H), 4.72-4.64 (m, 1H), 4.63 (t, J = 7.1 Hz, 2H), 4.34 (t, J =6.5 Hz, 2H), 4.56 (s, 2H), 4.46 (t, J = 6.2 Hz, 2H), 3.97-3.90 (m, 1H), 3.73 (d, J = 11.8 Hz, 2H), 3.10-3.00 (m, 2H), 2.05-1.94 (m, 2H), 1.83-1.73 (m,1H), 1.71-1 .53 (m, 2H), 1.02 (d, J = 6.5 Hz, 3H). LCMS [M+H] + m/z: 539.2
实施例25: (S)-2-(2-((1-(1-((8-氯-1,7-萘啶-2-基)甲基)哌啶-4-基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物25)的合成Example 25: Synthesis of (S)-2-(2-((1-(1-((8-chloro-1,7-naphthyridin-2-yl)methyl)piperidin-4-yl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 25)
关键起始物料8-氯-1,7-萘啶-2-甲醛(25-3)的制备:Preparation of the key starting material 8-chloro-1,7-naphthyridine-2-carbaldehyde (25-3):
步骤A 中间体 25-1 的合成Step A Synthesis of Intermediate 25-1
将2-氯-8-甲氧基-1,7-萘啶(2.5 g, 12.85 mmol)溶于1,4-二氧六环(25 mL)和水(5.0 mL)的混合溶剂中,依次加入碳酸钠(4.1 g, 38.50 mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(1.0 g, 1.29 mmol)和乙烯三氟硼酸钾(3.4 g, 25.70mmol)。反应混合物在氮气体系下,在95 ℃搅拌2小时。LCMS监测反应完成后,待反应液冷却至室温,将反应液倒入水(100 mL)中,乙酸乙酯(50 mL)萃取三次,合并有机相,并用饱和氯化钠溶液(50 mL)洗涤两次,无水硫酸钠干燥,过滤减压浓缩得到粗品。粗品通过柱层析(二氧化硅,石油醚:乙酸乙酯 = 20:1至10:1)纯化,得到中间体25-1(2.1 g,产率:89%),为黄色油状物。LCMS [M+H]+ m/z: 187.3。2-Chloro-8-methoxy-1,7-naphthyridine (2.5 g, 12.85 mmol) was dissolved in a mixed solvent of 1,4-dioxane (25 mL) and water (5.0 mL), and sodium carbonate (4.1 g, 38.50 mmol), [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium dichloromethane complex (1.0 g, 1.29 mmol) and potassium ethylene trifluoroborate (3.4 g, 25.70 mmol) were added in sequence. The reaction mixture was stirred at 95 °C for 2 hours under a nitrogen system. After the reaction was completed by LCMS monitoring, the reaction solution was cooled to room temperature, poured into water (100 mL), extracted three times with ethyl acetate (50 mL), and the organic phases were combined and washed twice with a saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to obtain a crude product. The crude product was purified by column chromatography (silica, petroleum ether: ethyl acetate = 20:1 to 10:1) to give intermediate 25-1 (2.1 g, yield: 89%) as a yellow oil. LCMS [M+H] + m/z: 187.3.
步骤B 中间体 25-2 的合成Step B Synthesis of Intermediate 25-2
将25-1(2.1 g, 11.28 mmol)溶于四氢呋喃(20 mL)和水(20 mL)的混合溶剂中,依次加入二水合锇酸钾(0.4 g, 1.13 mmol)和高碘酸钠(7.3 g, 33.80 mmol)。反应混合物在25 ℃下搅拌3小时。LCMS监测反应完成后,将反应液倒入水(100 mL)中,乙酸乙酯(50mL)萃取三次,合并有机相,并用饱和氯化钠溶液(30 mL)洗涤两次,无水硫酸钠干燥,过滤减压浓缩得到粗品。粗品通过柱层析(二氧化硅,石油醚:乙酸乙酯 = 10:1至5:1)纯化,得到中间体25-2(1.6 g,产率:75 %),为白色固体。LCMS [M+H]+ m/z: 189.3。25-1 (2.1 g, 11.28 mmol) was dissolved in a mixed solvent of tetrahydrofuran (20 mL) and water (20 mL), and potassium osmate dihydrate (0.4 g, 1.13 mmol) and sodium periodate (7.3 g, 33.80 mmol) were added in sequence. The reaction mixture was stirred at 25 °C for 3 hours. After the reaction was completed by LCMS monitoring, the reaction solution was poured into water (100 mL), extracted three times with ethyl acetate (50 mL), the organic phases were combined, washed twice with saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to obtain a crude product. The crude product was purified by column chromatography (silica, petroleum ether: ethyl acetate = 10:1 to 5:1) to obtain intermediate 25-2 (1.6 g, yield: 75%) as a white solid. LCMS [M+H] + m/z: 189.3.
步骤C 关键起始物料 25-3 的合成Step C Synthesis of key starting material 25-3
将25-2(1.4 g, 7.44 mmol)溶于三氯氧磷(20 mL, 215.00 mmol)中。反应混合物在110 ℃下搅拌1小时。LCMS监测反应完成后,将反应液减压浓缩得到粗品。粗品通过柱层析(二氧化硅,石油醚:乙酸乙酯 = 5:1至3:1)纯化,得到关键起始物料25-3(525 mg,产率:37 %),为黄色固体。1H NMR (400 MHz,CDCl3) δ 10.30 (d, J = 0.8 Hz, 1H), 8.51 (d,J = 5.6 Hz, 1H), 8.39 – 8.36 (m, 1H), 8.29 (d, J = 8.5 Hz, 1H), 7.72 (d, J =5.6 Hz, 1H). LCMS [M+H]+ m/z: 193.2。25-2 (1.4 g, 7.44 mmol) was dissolved in phosphorus oxychloride (20 mL, 215.00 mmol). The reaction mixture was stirred at 110 °C for 1 hour. After the reaction was completed as monitored by LCMS, the reaction solution was concentrated under reduced pressure to obtain a crude product. The crude product was purified by column chromatography (silica, petroleum ether: ethyl acetate = 5:1 to 3:1) to obtain the key starting material 25-3 (525 mg, yield: 37%) as a yellow solid. 1 H NMR (400 MHz, CDCl 3 ) δ 10.30 (d, J = 0.8 Hz, 1H), 8.51 (d,J = 5.6 Hz, 1H), 8.39 – 8.36 (m, 1H), 8.29 (d, J = 8.5 Hz, 1H), 7.72 (d, J =5.6 Hz, 1 H). LCMS [M+H] + m/z: 193.2.
3化合物25的制备:3 Preparation of compound 25:
利用化合物8-氯-1,7-萘啶-2-甲醛替换化合物8-氯喹啉-2-甲醛,参照实施例22的操作进行,得到目标化合物25为白色固体。1H NMR (500 MHz, CD3OD) δ 8.34 (d, J =8.5 Hz, 1H), 8.27 (d, J = 5.5 Hz, 1H), 8.12 (s, 2H), 7.92 (d, J = 8.5 Hz,1H), 7.78 (d, J = 5.5 Hz, 1H), 4.89 – 4.84 (m, 1H), 4.82 (t, J = 6.5 Hz, 2H),4.51 (t, J = 6.5 Hz, 2H), 3.96 (s, 2H), 3.93 – 3.88 (m, 1H), 3.35 (s, 2H),3.11 – 3.04 (m, 2H), 2.33 – 2.24 (m, 2H), 1.85 – 1.73 (m, 2H), 1.54 – 1.48(m, 1H), 1.48 – 1.39 (m, 2H), 1.14 (d, J = 6.5 Hz, 3H). LCMS [M+H]+ m/z:496.3。The compound 8-chloro-1,7-naphthyridine-2-carbaldehyde was used to replace the compound 8-chloroquinoline-2-carbaldehyde, and the operation of Example 22 was referred to to obtain the target compound 25 as a white solid. 1 H NMR (500 MHz, CD 3 OD) δ 8.34 (d, J =8.5 Hz, 1H), 8.27 (d, J = 5.5 Hz, 1H), 8.12 (s, 2H), 7.92 (d, J = 8.5 Hz, 1H), 7.78 (d, J = 5.5 Hz, 1H), 4.8 9 – 4.84 (m, 1H), 4.82 (t, J = 6.5 Hz, 2H), 4.51 (t, J = 6.5 Hz, 2H), 3.96 (s, 2H), 3.93 – 3.88 (m, 1H), 3.35 (s, 2H), 3.11 – 3.04 (m, 2H), 2. 33 – 2.24 (m, 2H), 1.85 – 1.73 (m, 2H), 1.54 – 1.48 (m, 1H), 1.48 – 1.39 (m, 2H), 1.14 (d, J = 6.5 Hz, 3H). LCMS [M+H] + m/z:496.3.
实施例26: (S)-2-(2-((1-(1-((8-氯-1,6-萘啶-2-基)甲基)哌啶-4-基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺(化合物26)的合成Example 26: Synthesis of (S)-2-(2-((1-(1-((8-chloro-1,6-naphthyridin-2-yl)methyl)piperidin-4-yl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 26)
关键起始物料8-氯-1,6-萘啶-2-甲醛(26-5)的制备:Preparation of key starting material 8-chloro-1,6-naphthyridine-2-carbaldehyde (26-5):
步骤A 中间体26-1的合成Step A Synthesis of Intermediate 26-1
将3-溴-5-氯吡啶-4-胺(12.0 g, 57.8 mmol)溶于N, N-二甲基甲酰胺(120 mL)中,依次加入丙烯酸乙酯(11.6 g, 116.0 mmol),N,N-二异丙基乙胺(30.3 mL, 174.0mmol),三(邻甲基苯基)磷(3.5 g, 11.57 mmol)和醋酸钯(1.3 g, 5.78 mmol)。反应混合物在氮气体系下,100℃搅拌10小时。LCMS监测反应完成后,待反应液冷却至室温,将反应液倒入水(100 mL)中,乙酸乙酯(100 mL)萃取两次,合并有机相,并用饱和氯化钠溶液(100mL)洗涤两次,无水硫酸钠干燥,过滤,减压浓缩得到粗品。粗品通过柱层析(二氧化硅,石油醚:乙酸乙酯 = 10:1至3:1)纯化,得到中间体26-1(9.0 g,产率:69 %),为黄色固体。LCMS[M+H]+ m/z: 227.2。3-Bromo-5-chloropyridin-4-amine (12.0 g, 57.8 mmol) was dissolved in N, N-dimethylformamide (120 mL), and ethyl acrylate (11.6 g, 116.0 mmol), N, N-diisopropylethylamine (30.3 mL, 174.0 mmol), tri(o-methylphenyl)phosphine (3.5 g, 11.57 mmol) and palladium acetate (1.3 g, 5.78 mmol) were added in sequence. The reaction mixture was stirred at 100 °C for 10 hours under a nitrogen system. After the reaction was completed by LCMS monitoring, the reaction solution was cooled to room temperature, poured into water (100 mL), extracted twice with ethyl acetate (100 mL), and the organic phases were combined and washed twice with saturated sodium chloride solution (100 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a crude product. The crude product was purified by column chromatography (silica, petroleum ether:ethyl acetate = 10:1 to 3:1) to give Intermediate 26-1 (9.0 g, yield: 69%) as a yellow solid. LCMS [M+H] + m/z: 227.2.
步骤B 中间体 26-2 的合成Step B Synthesis of Intermediate 26-2
将26-1(6.0 g, 26.5 mmol)溶于醋酸(30 mL)中,向反应液中加入三丁基膦(6.5mL, 26.50 mmol)。反应混合物在110 ℃下搅拌1小时。LCMS监测反应完成后,待反应液冷却至室温,将反应液倒入水(100 mL)中,通过硅藻土过滤,滤饼用乙酸乙酯(20 mL)洗涤,收集滤饼并减压浓缩得到26-2(4.0 g,产率:84%),为白色固体。1H NMR (400 MHz, DMSO-d6) δ11.69 (s, 1H), 8.80 (s, 1H), 8.57 (s, 1H), 8.06 (d, J = 9.6 Hz, 1H), 6.68 (d,J = 9.6 Hz, 1H). LCMS [M+H]+ m/z: 181.2。26-1 (6.0 g, 26.5 mmol) was dissolved in acetic acid (30 mL), and tributylphosphine (6.5 mL, 26.50 mmol) was added to the reaction solution. The reaction mixture was stirred at 110 °C for 1 hour. After the reaction was completed by LCMS monitoring, the reaction solution was cooled to room temperature, poured into water (100 mL), filtered through diatomaceous earth, and the filter cake was washed with ethyl acetate (20 mL). The filter cake was collected and concentrated under reduced pressure to obtain 26-2 (4.0 g, yield: 84%) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ11.69 (s, 1H), 8.80 (s, 1H), 8.57 (s, 1H), 8.06 (d, J = 9.6 Hz, 1H), 6.68 (d,J = 9.6 Hz, 1H). LCMS [M+H] + m/z: 181.2 .
步骤C 中间体 26-3 的合成Step C Synthesis of Intermediate 26-3
将26-2(3.8 g, 21.04 mmol)溶于三氯氧磷(20 mL, 210.4 mmol)中。反应混合物在氮气体系下,在120 ℃搅拌2小时。LCMS监测反应完成后,待反应液冷却至室温,将反减压浓缩得到中间体26-3(4.0 g,粗品),没有进一步纯化直接用于下一步。LCMS [M+H]+ m/z:199.1。26-2 (3.8 g, 21.04 mmol) was dissolved in phosphorus oxychloride (20 mL, 210.4 mmol). The reaction mixture was stirred at 120 °C for 2 hours under nitrogen. After the reaction was completed, the reaction solution was cooled to room temperature and concentrated under reduced pressure to obtain intermediate 26-3 (4.0 g, crude product), which was used directly in the next step without further purification. LCMS [M+H] + m/z: 199.1.
步骤D 中间体 26-4 的合成Step D Synthesis of Intermediate 26-4
将26-3(4.0 g, 20.10 mmol)溶于1,4-二氧六环(40 mL)和水(8.0 mL)的混合溶剂中,依次加入碳酸钠(6.4 g, 60.30 mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(1.5 g, 2.01 mmol)和乙烯三氟硼酸钾(5.4 g, 40.2 mmol)。反应混合物在氮气体系下,在80 ℃搅拌8小时。LCMS监测反应完成后,待反应液冷却至室温,将反应液倒入水(100 mL)中,乙酸乙酯(100 mL)萃取三次,合并有机相,并用饱和氯化钠溶液(50 mL)洗涤两次,无水硫酸钠干燥,过滤减压浓缩得到粗品。粗品通过柱层析(二氧化硅,石油醚:乙酸乙酯 = 5:1至3:1)纯化,得到中间体26-4(2.0 g,产率:52 %),为白色固体。LCMS [M+H]+m/z: 191.1。26-3 (4.0 g, 20.10 mmol) was dissolved in a mixed solvent of 1,4-dioxane (40 mL) and water (8.0 mL), and sodium carbonate (6.4 g, 60.30 mmol), [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium dichloromethane complex (1.5 g, 2.01 mmol) and potassium ethylene trifluoroborate (5.4 g, 40.2 mmol) were added in sequence. The reaction mixture was stirred at 80 °C for 8 hours under a nitrogen system. After the reaction was completed as monitored by LCMS, the reaction solution was cooled to room temperature, poured into water (100 mL), extracted three times with ethyl acetate (100 mL), and the organic phases were combined and washed twice with a saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a crude product. The crude product was purified by column chromatography (silica, petroleum ether:ethyl acetate = 5:1 to 3:1) to give intermediate 26-4 (2.0 g, yield: 52%) as a white solid. LCMS [M+H] + m/z: 191.1.
步骤E 关键起始物料 26-5的合成Step E Synthesis of key starting material 26-5
将26-4(500 mg, 2.62 mmol)溶于四氢呋喃(10 mL)和水(10 mL)的混合溶剂中,依次加入二水合锇酸钾(80 mg, 0.26 mmol)和高碘酸钠(1.7 g, 7.87 mmol)。反应混合物在25℃下搅拌3小时。LCMS监测反应完成后,将反应液倒入水(20 mL)中,用乙酸乙酯(30mL)萃取三次,合并有机相,并用饱和氯化钠溶液(30 mL)洗涤两次,无水硫酸钠干燥,过滤减压浓缩得到粗品。粗品通过柱层析(二氧化硅,石油醚:乙酸乙酯 = 10:1至5:1)纯化,得到中间体26-5(300 mg,产率:59 %),为白色固体。1H NMR (400 MHz, CDCl3) δ 10.33 (d,J = 0.9 Hz, 1H), 9.29 (s, 1H), 8.96 (s, 1H), 8.53 (dd, J = 8.4, 0.9 Hz, 1H),8.22 (d, J = 8.4 Hz, 1H).LCMS [M+H]+ m/z: 193.2。26-4 (500 mg, 2.62 mmol) was dissolved in a mixed solvent of tetrahydrofuran (10 mL) and water (10 mL), and potassium osmate dihydrate (80 mg, 0.26 mmol) and sodium periodate (1.7 g, 7.87 mmol) were added in sequence. The reaction mixture was stirred at 25 °C for 3 hours. After the reaction was completed by LCMS monitoring, the reaction solution was poured into water (20 mL), extracted three times with ethyl acetate (30 mL), the organic phases were combined, washed twice with saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to obtain a crude product. The crude product was purified by column chromatography (silica, petroleum ether: ethyl acetate = 10:1 to 5:1) to obtain intermediate 26-5 (300 mg, yield: 59%) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 10.33 (d,J = 0.9 Hz, 1H), 9.29 (s, 1H), 8.96 (s, 1H), 8.53 (dd, J = 8.4, 0.9 Hz, 1H), 8.22 (d, J = 8.4 Hz, 1H).LCMS [M+H] + m/z: 193.2.
1化合物26的制备:1 Preparation of compound 26:
利用化合物8-氯-1,6-萘啶-2-甲醛替换化合物8-氯喹啉-2-甲醛,参照实施例22的操作进行,得到目标化合物26为白色固体。1H NMR (500 MHz, CD3OD) δ 9.24 (s, 1H),8.79 (s, 1H), 8.55 (d, J = 8.5 Hz, 1H), 8.20 (s, 2H), 7.96 (d, J = 8.5 Hz,1H), 4.95 – 4.91 (m, 1H), 4.89 (t, J = 6.5 Hz, 2H), 4.58 (t, J = 6.5 Hz, 2H),3.98 (s, 2H), 3.98 – 3.94 (m, 1H), 3.38 (s, 2H), 3.07 (t, J = 10.0 Hz, 2H),2.33 – 2.20 (m, 2H), 1.91 – 1.77 (m, 2H), 1.57 – 1.42 (m, 3H), 1.21 (d, J =6.5 Hz, 3H).LCMS [M+H]+ m/z: 496.3。The compound 8-chloro-1,6-naphthyridine-2-carbaldehyde was used to replace the compound 8-chloroquinoline-2-carbaldehyde, and the operation of Example 22 was referred to to obtain the target compound 26 as a white solid. 1 H NMR (500 MHz, CD 3 OD) δ 9.24 (s, 1H), 8.79 (s, 1H), 8.55 (d, J = 8.5 Hz, 1H), 8.20 (s, 2H), 7.96 (d, J = 8.5 Hz, 1H), 4.95 – 4.91 (m, 1H), 4. 89 (t, J = 6.5 Hz, 2H), 4.58 (t, J = 6.5 Hz, 2H), 3.98 (s, 2H), 3.98 – 3.94 (m, 1H), 3.38 (s, 2H), 3.07 (t, J = 10.0 Hz, 2H), 2.33 – 2.20 (m, 2H ), 1.91 – 1.77 (m, 2H), 1.57 – 1.42 (m, 3H), 1.21 (d, J =6.5 Hz, 3H).LCMS [M+H] + m/z: 496.3.
实施例27: (R)-2-(2-((1-(1-((8-氯-1,6-萘啶-2-基)甲基)哌啶-4-基)乙基)氨基)嘧啶-5-基)-N-(氧杂环丁烷-3-基)乙酰胺 (化合物27)的合成Example 27: Synthesis of (R)-2-(2-((1-(1-((8-chloro-1,6-naphthyridin-2-yl)methyl)piperidin-4-yl)ethyl)amino)pyrimidin-5-yl)-N-(oxetane-3-yl)acetamide (Compound 27)
利用化合物(R)-4-(1-氨基乙基)哌啶-1-羧酸叔丁酯替换(S)-4-(1-氨基乙基)哌啶-1-羧酸叔丁酯,参照实施例26的操作进行,得到目标化合物27为白色固体。1H NMR (500MHz, CD3OD) δ 9.20 (s, 1H), 8.75 (s, 1H), 8.51 (d, J = 8.5 Hz, 1H), 8.16 (s,2H), 7.91 (d, J = 8.5 Hz, 1H), 4.93 – 4.87 (m, 1H), 4.86 (t, J = 6.5 Hz, 2H),4.54 (t, J = 6.5 Hz, 2H), 3.95 (d, J = 4.5 Hz, 2H), 3.95 – 3.90 (m, 1H), 3.35(s, 2H), 3.04 (t, J = 10.0 Hz, 2H), 2.31 – 2.19 (m, 2H), 1.89 – 1.74 (m, 2H),1.54 – 1.41 (m, 3H), 1.17 (d, J = 6.5 Hz, 3H).LCMS [M+H]+ m/z: 496.3。Using compound (R)-4-(1-aminoethyl)piperidine-1-carboxylic acid tert-butyl ester to replace (S)-4-(1-aminoethyl)piperidine-1-carboxylic acid tert-butyl ester, refer to the procedures of Example 26 to obtain the target compound 27 as a white solid. 1 H NMR (500MHz, CD 3 OD) δ 9.20 (s, 1H), 8.75 (s, 1H), 8.51 (d, J = 8.5 Hz, 1H), 8.16 (s,2H), 7.91 (d, J = 8.5 Hz, 1H), 4.93 – 4.87 (m, 1H), 4. 86 (t, J = 6.5 Hz, 2H), 4.54 (t, J = 6.5 Hz, 2H), 3.95 (d, J = 4.5 Hz, 2H), 3.95 – 3.90 (m, 1H), 3.35 (s, 2H), 3.04 (t, J = 10.0 Hz, 2H), 2.31 – 2.19 (m, 2H), 1.89 – 1.74 (m, 2H),1.54 – 1.41 (m, 3H), 1.17 (d, J = 6.5 Hz, 3H).LCMS [M+H] + m/z: 496.3.
实施例28:2-(2-(S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-2-羟基-N-(氧杂环丁烷-3-基)乙酰胺 (化合物28)的合成Example 28: Synthesis of 2-(2-(S)-1-(4-((8-chloro-1,7-naphthyridin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-2-hydroxy-N-(oxetane-3-yl)acetamide (Compound 28)
参照实施例13的操作进行,得到目标化合物28为白色固体。 1H NMR (500 MHz,CDCl3) δ 8.58 (s, 2H), 8.01 (d, J = 5.0 Hz, 1H), 7.74 (d, J = 9.0 Hz, 1H),7.33 (d, J = 5.0 Hz, 1H), 6.79 (d, J = 9.0 Hz, 1H), 6.54-6.49 (m, 1H), 5.82(s, 1H), 5.28 (d, J = 9.0 Hz, 1H), 5.12 (br s, 1H), 5.10 (brs, 1H), 5.04-4.95(m, 1H), 4.92-4.84 (m, 2H), 4.46 (t, J = 6.5 Hz, 1H), 4.42 (t, J = 6.5 Hz,1H), 4.06-3.96 (m, 1H), 3.36 (q, J = 7.0 Hz, 1H), 2.34-2.13 (m, 2H), 2.03-1.81 (m, 2H), 1.37-1.25 (m, 4H), 1.18 (d, J = 6.5 Hz, 3H). LC-MS [M+H]+m/z:512.2.The reaction was carried out according to the procedure of Example 13 to obtain the target compound 28 as a white solid. 1 H NMR (500 MHz, CDCl 3 ) δ 8.58 (s, 2H), 8.01 (d, J = 5.0 Hz, 1H), 7.74 (d, J = 9.0 Hz, 1H), 7.33 (d, J = 5.0 Hz, 1H), 6.79 (d, J = 9.0 Hz, 1H), 6.5 4-6.49 (m, 1H), 5.82(s, 1H), 5.28 (d, J = 9.0 Hz, 1H), 5.12 (br s, 1H), 5.10 (brs, 1H), 5.04-4.95(m, 1H), 4.92-4.84 (m, 2H), 4.46 (t, J = 6.5 Hz, 1H), 4.42 (t, J = 6.5 Hz, 1H), 4.06-3.96 (m, 1H), 3.36 (q, J = 7.0 Hz, 1H), 2.34-2.13 (m, 2H), 2.03-1.81 (m, 2H), 1.37-1.25 (m, 4H), 1. 18 (d, J = 6.5 Hz, 3H). LC-MS [M+H] + m/z:512.2.
实施例29:(S)-2-(2-((1-(4-((8-氯-1,7-萘啶-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-2,2-二氟-N-(氧杂环丁烷-3-基)乙酰胺(化合物29)的合成Example 29: Synthesis of (S)-2-(2-((1-(4-((8-chloro-1,7-naphthyridin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-2,2-difluoro-N-(oxetane-3-yl)acetamide (Compound 29)
参照实施例13的操作进行,得到目标化合物29为白色固体。 1H NMR (500 MHz,CDCl3) δ 8.58 (s, 2H), 8.01 (d, J = 5.0 Hz, 1H), 7.74 (d, J = 9.0 Hz, 1H),7.33 (d, J = 5.0 Hz, 1H), 6.79 (d, J = 9.0 Hz, 1H), 6.54-6.49 (m, 1H), 5.28(d, J = 9.0 Hz, 1H), 5.10 (brs, 1H), 5.04-4.95 (m, 1H), 4.92-4.84 (m, 2H),4.46 (t, J = 6.5 Hz, 1H), 4.42 (t, J = 6.5 Hz, 1H), 4.06-3.96 (m, 1H), 3.36(q, J = 7.0 Hz, 1H), 2.34-2.13 (m, 2H), 2.03-1.81 (m, 2H), 1.37-1.25 (m, 4H),1.19 (d, J = 6.5 Hz, 3H). LC-MS [M+H]+m/z: 532.2.The reaction was carried out according to the procedure of Example 13 to obtain the target compound 29 as a white solid. 1 H NMR (500 MHz, CDCl 3 ) δ 8.58 (s, 2H), 8.01 (d, J = 5.0 Hz, 1H), 7.74 (d, J = 9.0 Hz, 1H), 7.33 (d, J = 5.0 Hz, 1H), 6.79 (d, J = 9.0 Hz, 1H), 6.5 4-6.49 (m, 1H), 5.28 (d, J = 9.0 Hz, 1H), 5.10 (brs, 1H), 5.04-4.95 (m, 1H), 4.92-4.84 (m, 2H), 4.46 (t, J = 6.5 Hz, 1H), 4.42 (t, J = 6.5 Hz, 1H), 4.06-3.96 (m, 1H), 3.36 (q, J = 7.0 Hz, 1H), 2.34-2.13 (m, 2H), 2.03-1.81 (m, 2H), 1.37-1.25 (m, 4H), 1.19 (d, J = 6.5 Hz, 3H). LC-MS [M+H ] + m/z: 532.2.
实施例30:2-(2-(((S)-1-(4-((8-氯-1,7-萘啶-2-基)氨基)环己基)乙基)氨基)嘧啶-5-基)-2-氰基-N-(氧杂环丁烷-3-基)乙酰胺 (化合物30) 的合成Example 30: Synthesis of 2-(2-(((S)-1-(4-((8-chloro-1,7-naphthyridin-2-yl)amino)cyclohexyl)ethyl)amino)pyrimidin-5-yl)-2-cyano-N-(oxetane-3-yl)acetamide (Compound 30)
参照实施例13的操作进行,得到目标化合物30为白色固体。 1H NMR (500 MHz,CDCl3) δ 8.58 (s, 2H), 8.20 (d, J = 5.0 Hz, 1H), 7.74 (d, J = 9.0 Hz, 1H),7.33 (d, J = 5.0 Hz, 1H), 6.79 (d, J = 9.0 Hz, 1H), 6.54-6.49 (m, 1H), 5.28(d, J = 9.0 Hz, 1H), 5.10 (brs, 1H), 5.04-4.95 (m, 1H), 4.92-4.84 (m, 2H),4.72 (t, J = 6.5 Hz, 1H), 4.42 (t, J = 6.5 Hz, 1H), 4.06-3.96 (m, 1H), 3.87-3.85 (m, 1H), 3.36 (q, J = 7.0 Hz, 1H), 2.34 – 2.13 (m, 2H), 2.03 – 1.81 (m,2H), 1.37 – 1.25 (m, 4H), 1.18 (d, J = 6.5 Hz, 3H). LC-MS [M+H]+m/z: 521.2.The reaction was carried out according to the procedure of Example 13 to obtain the target compound 30 as a white solid. 1 H NMR (500 MHz, CDCl 3 ) δ 8.58 (s, 2H), 8.20 (d, J = 5.0 Hz, 1H), 7.74 (d, J = 9.0 Hz, 1H), 7.33 (d, J = 5.0 Hz, 1H), 6.79 (d, J = 9.0 Hz, 1H), 6.5 4-6.49 (m, 1H), 5.28 (d, J = 9.0 Hz, 1H), 5.10 (brs, 1H), 5.04-4.95 (m, 1H), 4.92-4.84 (m, 2H), 4.72 (t, J = 6.5 Hz, 1H), 4.42 (t, J = 6.5 Hz, 1H), 4.06-3.96 (m, 1H), 3.87-3.85 (m, 1H), 3.36 (q, J = 7.0 Hz, 1H), 2.34 – 2.13 (m, 2H), 2.03 – 1.81 (m,2H), 1.37 – 1.25 (m, 4H), 1.18 (d, J = 6.5 Hz, 3H). LC-MS [M+H] + m/z: 521.2.
生物学测定Biological assays
使用如下所述的测定以及本领域已知的其他测定,可以评价本发明的化合物的抑制EZH2的能力。Compounds of the invention can be evaluated for their ability to inhibit EZH2 using the assays described below, as well as other assays known in the art.
实验主要仪器设备:Main experimental instruments and equipment:
离心机(Beckman Avanti J-15R);超低温冰箱(Thermo FDE60086FV);二氧化碳培养箱(ESCO CLM-170B-CN); 酶标仪(Spark tecan);排枪(20-200 µL);细胞计数仪(伯乐TC20)。Centrifuge (Beckman Avanti J-15R); ultra-low temperature refrigerator (Thermo FDE60086FV); carbon dioxide incubator (ESCO CLM-170B-CN); microplate reader (Spark tecan); slug gun (20-200 µL); cell counter (Bio-Rad TC20).
实施例31:EZH2 体外酶活性测定Example 31: EZH2 in vitro enzyme activity assay
步骤1Step 1
配制反应缓冲液(4X):80 mM Tris,pH 8 .0,0 .04% BSA,0 .04% Triton-100,2 mM DTT。调整PH至8.5,使用前加入DTT。Prepare reaction buffer (4X): 80 mM Tris, pH 8.0, 0.04% BSA, 0.04% Triton-100, 2 mM DTT. Adjust pH to 8.5 and add DTT before use.
步骤2Step 2
将本发明化合物在DMSO中每次稀释3倍,连续稀释9次以获得10种浓度。为了使反应体系中DMSO终浓度为1%。最后一步用水将化合物稀释10倍。将每种浓度的测试化合物(各1.92 μL)到8联排中。将含有1mg/mL野生型PRC2(6组分)复合体(0.1056 μL)、1 mM SAM(0.16 μL)、2 mM H3[21-44](0.08 μL)以及反应缓冲液 (2 μL)添加到8联排的孔中,使用排枪混匀后和测试化合物25℃孵育2 h。反应溶液中的最终组分包含40 nM 野生型PRC2、20 nM SAM、20 μM H3[21-44]与不同浓度的化合物(0 µM、0.00152 µM、0.00457 µM 、0.01372 µM 、0.04115 µM、0.12346 µM 、0.37037 µM 、1.11111 µM 、3.33333 µM 、10 µM)。空白对照由20 nM SAM、20 μM H3[21-44]组成。反应完成后向8联排的孔中添加4.8 μL淬灭溶液(1%TFA),在25℃下终止反应5 min。The compound of the present invention was diluted 3 times in DMSO each time, and diluted 9 times in succession to obtain 10 concentrations. In order to make the final DMSO concentration in the reaction system 1%. In the last step, the compound was diluted 10 times with water. Each concentration of the test compound (1.92 μL each) was added to 8 rows. A 1 mg/mL wild-type PRC2 (6 components) complex (0.1056 μL), 1 mM SAM (0.16 μL), 2 mM H3[21-44] (0.08 μL) and reaction buffer (2 μL) were added to the wells of the 8 rows, mixed with a spray gun, and incubated with the test compound at 25°C for 2 h. The final composition of the reaction solution contained 40 nM wild-type PRC2, 20 nM SAM, 20 μM H3[21-44] and different concentrations of compounds (0 μM, 0.00152 μM, 0.00457 μM, 0.01372 μM, 0.04115 μM, 0.12346 μM, 0.37037 μM, 1.11111 μM, 3.33333 μM, 10 μM). The blank control consisted of 20 nM SAM and 20 μM H3[21-44]. After the reaction was completed, 4.8 μL of quenching solution (1% TFA) was added to the wells of the 8-row wells and the reaction was terminated at 25°C for 5 min.
步骤3Step 3
添加6×MTase-GLoTM reagent(10×MTase-GLoTM reagent,3 mM MgCl2,50 mMNaCl、20 mM Tris,pH 8 .0,0 .01%BSA,0 .01%Triton-100,0.5 mM DTT)混合物4.8 μL,25℃反应30 min。反应完成后向管中加入Detection buffer 28.8 μL/孔, 混匀后将反应混合物20 μL/孔添加到384孔板中,以2000 rpm离心2 min,25℃反应30 min。在酶标仪上读取化学发光信号,检测时间为1000 ms。所有组别数据减去对照组相应数据,再根据标准曲线即可计算出相应的抑制率,并且计算IC50值。Add 4.8 μL of 6×MTase-GLo TM reagent (10×MTase-GLo TM reagent, 3 mM MgCl 2 , 50 mMNaCl, 20 mM Tris, pH 8.0, 0.01% BSA, 0.01% Triton-100, 0.5 mM DTT) mixture and react at 25°C for 30 min. After the reaction is completed, add 28.8 μL/well of detection buffer to the tube, mix well, add 20 μL/well of the reaction mixture to the 384-well plate, centrifuge at 2000 rpm for 2 min, and react at 25°C for 30 min. Read the chemiluminescent signal on the microplate reader, and the detection time is 1000 ms. Subtract the corresponding data of the control group from all group data, and then calculate the corresponding inhibition rate according to the standard curve, and calculate the IC 50 value.
实施例32:G401 ELISA(H3K27me3甲基化)测定Example 32: G401 ELISA (H3K27me3 methylation) assay
步骤1Step 1
将本发明的化合物在DMSO中每次稀释3倍、连续稀释9次,以获得总计10种浓度。然后将化合物添加到96孔板中培养的G401细胞中,以获得终浓度0 nM、0.152 nM、0.457 nM、1.372 nM、4.115 nM、12.346 nM、37.037 nM、111.111 nM、333.333 nM、1000 nM的化合物溶液。在ELISA程序前,将细胞培养在37℃、5%CO2的培养箱中培养48 h。The compounds of the present invention were diluted 3-fold each time in DMSO, serially diluted 9 times to obtain a total of 10 concentrations. The compounds were then added to G401 cells cultured in 96-well plates to obtain compound solutions with final concentrations of 0 nM, 0.152 nM, 0.457 nM, 1.372 nM, 4.115 nM, 12.346 nM, 37.037 nM, 111.111 nM, 333.333 nM, and 1000 nM. Cells were cultured in an incubator at 37°C and 5% CO2 for 48 h before the ELISA procedure.
步骤2Step 2
将96孔板中的细胞用1×PBS缓冲液洗涤并加入裂解缓冲液(0.4N HCl)100 μL/孔裂解,将板放在摇床上150 rpm、4℃孵育30 min。加入中和缓冲液(0 .5 M磷酸氢二钠,pH12 .5,1mM DTT;80 μL/孔)中和,并使用排枪吹打约20下以充分混合溶液,中和后将96孔板在2000 rpm下离心2min。The cells in the 96-well plate were washed with 1× PBS buffer and lysed by adding lysis buffer (0.4N HCl) at 100 μL/well, and the plate was incubated on a shaker at 150 rpm and 4°C for 30 min. Neutralization buffer (0.5 M disodium hydrogen phosphate, pH 12.5, 1 mM DTT; 80 μL/well) was added for neutralization, and the solution was mixed thoroughly by blowing for about 20 times with a spray gun. After neutralization, the 96-well plate was centrifuged at 2000 rpm for 2 min.
步骤3Step 3
取离心后96孔板中的细胞裂解液10-50 μL/孔到384孔板孔中,并取1×PBS 0~40μL将孔内体积调节至50 μL。将板密封,以2000 rpm离心2 min,并在4℃下孵育约16 h。Take 10-50 μL/well of the cell lysate in the centrifuged 96-well plate into the wells of the 384-well plate, and adjust the volume in the well to 50 μL with 0-40 μL of 1× PBS. Seal the plate, centrifuge at 2000 rpm for 2 min, and incubate at 4°C for about 16 h.
步骤4Step 4
孵育后,用TBST洗涤五次:洗涤操作为每孔添加60 μL TBST缓冲液,并放摇床上200 rpm、25℃洗涤5 min。After incubation, wash five times with TBST: add 60 μL TBST buffer to each well and wash on a shaker at 200 rpm and 25°C for 5 min.
步骤5Step 5
每孔添加50 μL封闭缓冲液(TBST,2%BSA),并将板以2000 rpm,离心2 min,放在摇床上200 rpm、25℃孵育1h。50 μL of blocking buffer (TBST, 2% BSA) was added to each well, and the plate was centrifuged at 2000 rpm for 2 min and incubated on a shaker at 200 rpm and 25°C for 1 h.
步骤6Step 6
去除封闭缓冲液,每孔添加30 μL一抗(2%BSA ,0.1 % H3K27me3;0.03 % H3)。将板以2000 rpm,离心2 min,并放在摇床上150 rpm、25℃孵育1h。The blocking buffer was removed and 30 μL of primary antibody (2% BSA, 0.1% H3K27me3; 0.03% H3) was added to each well. The plate was centrifuged at 2000 rpm for 2 min and incubated on a shaker at 150 rpm and 25°C for 1 h.
步骤7Step 7
用TBST洗涤五次去除一抗,每孔添加30 μL二抗(2%BSA ,0.04 % 抗兔抗体),384孔板以2000 rpm,离心2 min,并放在摇床上150 rpm、25℃孵育1h。The primary antibody was removed by washing five times with TBST, and 30 μL of secondary antibody (2% BSA, 0.04% anti-rabbit antibody) was added to each well. The 384-well plate was centrifuged at 2000 rpm for 2 min and incubated on a shaker at 150 rpm and 25°C for 1 h.
步骤8Step 8
用TBST洗涤五次去除二抗。每孔添加30 μL的 ECL底物,并将板以2000 rpm离心2min。The secondary antibody was removed by washing five times with TBST. 30 μL of ECL substrate was added to each well, and the plate was centrifuged at 2000 rpm for 2 min.
步骤9Step 9
室温孵育5分钟后,在酶标仪上读取信号。使用H3信号将H3K27me3甲基化读数归一化,然后对经DMSO处理的样品计算抑制百分率。使用GraphPad Prism程序将数据拟合至剂量反应曲线以获得测试化合物的IC50值。After 5 minutes of incubation at room temperature, the signal was read on a microplate reader. H3K27me3 methylation readings were normalized using the H3 signal, and the percentage inhibition was then calculated for DMSO-treated samples. The data were fitted to a dose-response curve using the GraphPad Prism program to obtain the IC 50 values of the test compounds.
本发明化合物依据上述实验方法进行测试,部分结果请见表2。化合物在靶点水平和G401细胞水平甲基化抑制剂活性基本都在1 uM以下,具有显著的靶点活性。本发明以靶向EZH2第一款上市药物Tazemetostat(EPZ-6438),中文名称:他泽司他为对比例1,CN114746414A专利中活性最好的化合物C36作为对比例2,部分发明中的化合物呈现出明显的优势。The compounds of the present invention were tested according to the above experimental method, and some of the results are shown in Table 2. The methylation inhibitor activity of the compounds at the target level and the G401 cell level is basically below 1 uM, and has significant target activity. The present invention uses Tazemetostat (EPZ-6438), the first marketed drug targeting EZH2, with the Chinese name: Tazemetostat, as comparative example 1, and the most active compound C36 in the CN114746414A patent as comparative example 2. The compounds in some inventions show obvious advantages.
表2化合物在蛋白水平和细胞水平的甲基化抑制活性Table 2 Methylation inhibitory activity of compounds at protein and cell levels
此发明中的化合物与对比例化合物相比较,普遍采用手性甲基的引入。以实施例中化合物8与对比例2的结构为例进行对比如下:Compared with the comparative example compounds, the compounds in this invention generally adopt the introduction of chiral methyl groups. The structures of compound 8 in the embodiment and comparative example 2 are compared as follows:
发明人发现,在化合物8中引入甲基结构后,令人惊讶地带来意料不到的活性效果。The inventors found that the introduction of a methyl structure into compound 8 surprisingly brought about an unexpected activity effect.
使用Glide软件在XP模式下做分子对接,并保留SAH分子以还原C36和SAH之间的相互作用, 提高对接计算的可靠性。C36和化合物8分子对接结果比较,图中红色分子为C36,绿色分子为化合物8,黄色分子是SAH。可以看到,化合物8和C36的整体构象一致(如图1所示,)化合物8中额外的甲基立体构型占据了由Y658的苯环以及C663和T678侧链形成的疏水空间,提高了分子与蛋白之间的结合能力,由此看出,甲基的引入对活性提高非常重要,与实验室测试结果高度吻合,如表3所示:Molecular docking was performed using Glide software in XP mode, and the SAH molecule was retained to restore the interaction between C36 and SAH, thereby improving the reliability of the docking calculation. Comparison of the molecular docking results of C36 and compound 8. The red molecule in the figure is C36, the green molecule is compound 8, and the yellow molecule is SAH. It can be seen that the overall conformation of compound 8 and C36 is consistent (as shown in Figure 1). The additional methyl stereo configuration in compound 8 occupies the hydrophobic space formed by the benzene ring of Y658 and the side chains of C663 and T678, which improves the binding ability between the molecule and the protein. It can be seen that the introduction of methyl groups is very important for improving activity, which is highly consistent with the laboratory test results, as shown in Table 3:
表3 化合物8、C36分别与靶点蛋白EZH2分子对接打分结果Table 3 Scoring results of molecular docking of compound 8 and C36 with target protein EZH2
细胞增殖的分析Analysis of cell proliferation
实施例33:Embodiment 33:
悬浮细胞:WSU-DLCL2/KARPAS 422细胞增殖的分析1) 培养基:Suspension cells: Analysis of WSU-DLCL2/KARPAS 422 cell proliferation 1) Culture medium:
10%FBS(Cat:A5669401;gibco)+ 89%RPMI-1640(Cat: PM150110;Pricella)+ 1%Pen Strep(Cat:15140;gibco)。10% FBS (Cat: A5669401; gibco) + 89% RPMI-1640 (Cat: PM150110; Pricella) + 1% Pen Strep (Cat: 15140; gibco).
2) 在铺96孔板之前,将WSU-DLCL2/KARPAS 422细胞使用50 µL/孔多聚赖氨酸处理一个小时以上,处理后使用100ul/孔PBS清洗,晾干。晾干后,将WSU-DLCL2/KARPAS 422细胞添加100 µL /孔培养基接种于96孔板中(细胞数约2000/孔,每个待测化合物进行两重复),第二天给药。2) Before plating in 96-well plates, WSU-DLCL2/KARPAS 422 cells were treated with 50 µL/well poly-lysine for more than one hour, washed with 100ul/well PBS, and dried. After drying, WSU-DLCL2/KARPAS 422 cells were added with 100 µL/well culture medium and seeded in 96-well plates (about 2000 cells/well, and two replicates were performed for each test compound), and drug administration was performed the next day.
3) 第二天,用DMSO配制不同浓度的待测化合物溶液。用DMSO进行等倍(3倍)稀释,配制9个不同浓度的相应待测化合物溶液。浓度分别为185.19、61.73、20.58、6.86、2.29、0.76、0.25、0.08、0.03 µM。3) On the second day, different concentrations of the test compound solutions were prepared with DMSO. 9 different concentrations of the corresponding test compound solutions were prepared by equal dilution (3 times) with DMSO. The concentrations were 185.19, 61.73, 20.58, 6.86, 2.29, 0.76, 0.25, 0.08, and 0.03 µM, respectively.
4) 稀释完成后分别取2 µL DMSO配制的不同浓度的待测化合物溶液加198µL培养基进行充分混匀(吹打20次以上)。随后向已铺细胞的96孔中分别加入25 µL/孔上述含有不同浓度待测化合物的培养基,吹打10次。最终得到终浓度为370.37、123.46、41.15、13.72、4.57、1.52、0.51、0.17、0.06 nM的含有待测化合物的培养基(含有0.2% DMSO),并配制DMSO对照溶液。4) After the dilution is completed, take 2 µL of the test compound solution prepared with DMSO at different concentrations and add 198 µL of culture medium to mix thoroughly (by pipetting more than 20 times). Then add 25 µL/well of the culture medium containing different concentrations of the test compound to the 96 wells where cells have been plated, and pipette 10 times. Finally, the final concentrations of the test compound-containing culture medium (containing 0.2% DMSO) of 370.37, 123.46, 41.15, 13.72, 4.57, 1.52, 0.51, 0.17, and 0.06 nM were obtained, and a DMSO control solution was prepared.
5) 随后将待测化合物或者DMSO处理的细胞放回37℃培养箱中进行培养,处理3-4days后,进行换药处理。5) The cells treated with the test compound or DMSO were then placed back into the 37°C incubator for culture. After 3-4 days of treatment, the treatment was changed.
6) 换药时:提前使用多聚赖氨酸处理过的96孔板中添加新的培养基80 µL。随后同第一次加药处理一样配制好不同浓度的含待测化合物培养基,分别向每个孔中加25 µL不同浓度的待测化合物培养基并使用排枪吹打4下混匀。步骤5) 中的WSU-DLCL2/KARPAS422细胞使用排枪吹打10下混匀细胞,随后取20 µL/孔的含细胞液移至上述加入了待测化合物培养基的96孔板中,然后吹打10次混匀。每隔3-4 days后,进行换药处理,共给药4次。6) When changing the medicine: add 80 µL of new culture medium to the 96-well plate treated with poly-lysine in advance. Then prepare different concentrations of culture medium containing the test compound as in the first drug addition treatment, add 25 µL of different concentrations of culture medium containing the test compound to each well and mix them by blowing 4 times with a dispenser. Use a dispenser to blow 10 times with a dispenser to mix the WSU-DLCL2/KARPAS422 cells in step 5), then take 20 µL/well of the cell solution and transfer it to the 96-well plate with the culture medium of the test compound added, and then blow 10 times to mix. Change the medicine every 3-4 days, and give the medicine 4 times in total.
7) 处理14 days后进行细胞活力检测。先使用排枪吹打培养基10-20下,充分吹散细胞,取20 µL/孔含有细胞的培养基,加入20 µL台盼蓝,充分混匀,进行细胞计数。7) Perform cell viability test 14 days after treatment. Use a spray gun to blow the culture medium 10-20 times to fully disperse the cells, take 20 µL/well of the culture medium containing cells, add 20 µL of trypan blue, mix thoroughly, and count the cells.
8) 细胞活力计算:计算公式 = 100*给药组/DMSO组8) Calculation of cell viability: Calculation formula = 100*drug administration group/DMSO group
所有组别数据减去对照组,再根据标准曲线即可计算出相应的抑制率,并且计算IC50值。The control group data was subtracted from all groups, and the corresponding inhibition rate was calculated based on the standard curve, and the IC 50 value was calculated.
实施例34:Embodiment 34:
贴壁细胞:G401细胞增殖的分析1) 培养基:Adherent cells: Analysis of G401 cell proliferation 1) Culture medium:
10%FBS(Cat:F8318;Sigma)+89%McCoy’s 5A(Cat:16600-082;gibco)+1%PenStrep(Cat:15140;gibco)。10% FBS (Cat: F8318; Sigma) + 89% McCoy’s 5A (Cat: 16600-082; gibco) + 1% PenStrep (Cat: 15140; gibco).
2) G401细胞500 µL /孔培养基接种于24孔板(细胞数约10000/孔,每个待测化合物进行单重复),第二天给药 。2) G401 cells were seeded in 24-well plates with 500 µL/well culture medium (about 10,000 cells/well, and each compound to be tested was replicated) and administered the next day.
3) 第二天,用DMSO配置不同浓度的待测化合物溶液。用DMSO进行等倍(3倍)稀释,配制9个不同浓度的相应待测化合物。浓度分别为5000、1666.67、555.56、185.19、61.73、20.58、6.86、2.29、0.76 µM。3) On the second day, prepare different concentrations of the test compound solution with DMSO. Make equal (3-fold) dilutions with DMSO to prepare 9 different concentrations of the corresponding test compound. The concentrations are 5000, 1666.67, 555.56, 185.19, 61.73, 20.58, 6.86, 2.29, and 0.76 µM.
4) 稀释完成后分别取2 µL DMSO配置的不同浓度的待测化合物溶液加198µL培养基进行充分混匀(吹打20次以上)。随后向已铺细胞的24孔中分别加入125 µL/孔上述含有不同浓度待测化合物的培养基。最终得到终浓度为10000、3333.33、1111.11、370.37、123.46、41.15、13.72、4.57、1.52 nM的含有待测化合物的培养基(含有0.2% DMSO),并配制DMSO对照溶液。4) After the dilution is completed, take 2 µL of the test compound solution of different concentrations prepared with DMSO and add 198 µL of culture medium to mix thoroughly (pipette more than 20 times). Then add 125 µL/well of the above culture medium containing different concentrations of the test compound to the 24 wells where cells have been plated. Finally, the final concentrations of the test compound culture medium (containing 0.2% DMSO) are obtained, and the DMSO control solution is prepared.
5) 随后将不同浓度待测化合物溶液或者DMSO处理的细胞放回37℃培养箱中进行培养,处理3-4 days后,进行换药处理。5) Then, the cells treated with different concentrations of the test compound solution or DMSO were placed back into the 37°C incubator for culture. After 3-4 days of treatment, the treatment was changed.
6) 换药时:吸掉G401细胞的培养基后用500 µL PBS洗涤一次,之后加入100 µL的胰酶,消化4min,再加入400 µL的培养基中和,以1000rpm,离心5min。离心后去掉上清后,取100 µL培养基重悬细胞,取10 µL重悬液铺于已添加490 µL新鲜培养基的24孔板中。向每个孔中加125 µL不同浓度的含待测化合物的培养基并摇晃混匀。每隔3-4 days后,进行换药处理。6) When changing the dressing: After removing the culture medium of G401 cells, wash once with 500 µL PBS, then add 100 µL trypsin, digest for 4 minutes, then add 400 µL culture medium to neutralize, centrifuge at 1000rpm for 5 minutes. After centrifugation, remove the supernatant, take 100 µL culture medium to resuspend the cells, take 10 µL of the resuspended liquid and spread it in a 24-well plate with 490 µL fresh culture medium added. Add 125 µL of culture medium containing the test compound of different concentrations to each well and shake to mix. Change the dressing every 3-4 days.
7) 第4次换药时,按照步骤6)的操作洗涤、消化和中和后,以500 µL培养基重悬细胞。然后取20 µL含细胞液添加于加有80 µL培养基 + 25 µL含有不同浓度待测化合物培养基的96孔黑板中,设置2个复孔,第14天进行Hoechst 染色。7) At the fourth dressing change, wash, digest and neutralize according to step 6), and resuspend the cells in 500 µL of culture medium. Then take 20 µL of cell solution and add it to a 96-well black plate with 80 µL of culture medium + 25 µL of culture medium containing different concentrations of the compound to be tested, set up 2 replicate wells, and perform Hoechst staining on the 14th day.
8) 14 days后收细胞,以倾覆法基本去掉所有的培养基,倒扣孔板于吸水纸上。8) After 14 days, harvest the cells, remove almost all the culture medium by inversion, and invert the well plate onto absorbent paper.
9) 配制固定染色液:将37%甲醛水按照10×使用,使用PBS配制3.7%的甲醛,每一个96孔板需要配置5ml 的3.7%的甲醛。将Hoechst 33342(1000 X)稀释于3.7%的甲醛溶液中,终浓度为10 ug/ml。9) Prepare the fixative staining solution: Use 37% formaldehyde water at 10×, and use PBS to prepare 3.7% formaldehyde. Each 96-well plate needs to be prepared with 5 ml of 3.7% formaldehyde. Dilute Hoechst 33342 (1000X) in 3.7% formaldehyde solution to a final concentration of 10 ug/ml.
10) 按照50 uL每孔,排枪加入固定染色液。水平摇床,30 min,RT,避光。10) Add 50 uL of fixative staining solution to each well by pipetting. Shake horizontally for 30 min at RT, away from light.
11) 倾覆法去掉基本所有的Hoechst 33342。排枪轻柔加入100 µL PBS进行清洗。水平摇床,5min,RT,避光。重复两次,最后使用倾覆法去掉所有的PBS,倒扣孔板于吸水纸上。11) Remove almost all Hoechst 33342 by tipping over. Gently add 100 µL PBS with a pipette to wash. Shake horizontally for 5 min at RT, away from light. Repeat twice, and finally remove all PBS by tipping over, and invert the plate onto absorbent paper.
12) 酶标仪激发光320(25),发射光460(20)。12) The excitation light of the microplate reader is 320 (25) and the emission light is 460 (20).
13) 已经读取的96孔板铝箔封闭存放于-20°。13) The read 96-well plate was sealed with aluminum foil and stored at -20°.
14) 贴壁细胞活力计算:14) Calculation of adherent cell viability:
细胞活力*(%)=[A(药物+)- A(空白)] / [A(药物-)- A(空白)] ×100%Cell viability* (%) = [A (drug+) - A (blank)] / [A (drug-) - A (blank)] × 100%
A(药物+):具有细胞和药物溶液的孔的吸光度;A (drug+): absorbance of wells with cells and drug solution;
A(药物-):具有细胞而没有药物溶液的孔的吸光度;A (drug-): absorbance of wells with cells but no drug solution;
A(空白):具有培养基而没有细胞的孔的吸光度。A (blank): absorbance of wells with culture medium but no cells.
*细胞活力:细胞增殖活力或细胞毒性活力*Cell viability: cell proliferation activity or cytotoxic activity
所有组别数据减去对照组,再根据标准曲线即可计算出相应的抑制率,并且计算IC50值。The control group data was subtracted from all groups, and the corresponding inhibition rate was calculated based on the standard curve, and the IC 50 value was calculated.
Karpas422, WSU-DLCL2和G401细胞是常见的EZH2突变或过表达而引起的肿瘤细胞,通常被作为细胞水平上的抗肿瘤活性评价模型。以Tazemetostat 和C36 作为对照化合物,本发明化合物依据上述实验方法进行测试显示出显著的抗肿瘤活性,部分结果请见表4。Karpas422, WSU-DLCL2 and G401 cells are common tumor cells caused by EZH2 mutation or overexpression, and are usually used as models for evaluating anti-tumor activity at the cellular level. Using Tazemetostat and C36 as control compounds, the compounds of the present invention were tested according to the above experimental method and showed significant anti-tumor activity. Some of the results are shown in Table 4.
表4化合物对肿瘤KARPAS422细胞的增殖抑制活性Table 4 Proliferation inhibition activity of compounds on tumor KARPAS422 cells
生物学测定Biological assays
实施例35:小鼠模型药代动力学性质测试Example 35: Pharmacokinetic properties testing in mouse models
口服给药:将化合物用10% DMSO + 10%Solutol + 80% Saline,给药剂量10 mg/kg, 于给药后0.25h,0.5h,1h,2h,4h,8h 和24h采血;Oral administration: The compound was mixed with 10% DMSO + 10% Solutol + 80% Saline at a dose of 10 mg/kg, and blood was collected at 0.25h, 0.5h, 1h, 2h, 4h, 8h and 24h after administration;
静脉给药:将化合物用10% DMSO + 10%Solutol + 80% Saline,给药剂量 5 mg/kg,于给药后0.083 h, 0.25 h, 0.5 h, 1 h, 2 h, 4 h, 8 h 和 24 h采血,采血方式为经颌下静脉或其他合适方式采血。Intravenous administration: The compound was mixed with 10% DMSO + 10% Solutol + 80% Saline at a dose of 5 mg/kg. Blood was collected via the submandibular vein or other appropriate methods at 0.083 h, 0.25 h, 0.5 h, 1 h, 2 h, 4 h, 8 h and 24 h after administration.
每个样品采集约30 μL/时间点,K2-EDTA抗凝,采集后放置冰上。并于1小时之内离心分离血浆(离心条件:6800g,6分钟,2-8ºC)。血浆样本在分析前存放时则放于-80 ºC冰箱内,待LC-MS/MS分析。About 30 μL of each sample was collected per time point, anticoagulated with K2-EDTA, and placed on ice after collection. The plasma was separated by centrifugation within 1 hour (centrifugation conditions: 6800g, 6 minutes, 2-8°C). The plasma samples were stored in a -80°C refrigerator before analysis and were ready for LC-MS/MS analysis.
根据测试所得的血药浓度-时间数据,采用WinNonlin软件求算药代动力学参数。化合物显示出良好的生物利用度,具有极佳的成药性前景。The pharmacokinetic parameters were calculated using WinNonlin software based on the blood drug concentration-time data obtained from the test. The compound showed good bioavailability and had excellent drugability prospects.
部分结果如表5所示:Some results are shown in Table 5:
表5 ICR小鼠静脉和口服给药的药代动力学参数Table 5 Pharmacokinetic parameters of intravenous and oral administration in ICR mice
Claims (10)
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202410986475.6A CN118515653B (en) | 2024-07-23 | 2024-07-23 | A kind of EZH2 specific inhibitor and its preparation method and use |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202410986475.6A CN118515653B (en) | 2024-07-23 | 2024-07-23 | A kind of EZH2 specific inhibitor and its preparation method and use |
Publications (2)
Publication Number | Publication Date |
---|---|
CN118515653A true CN118515653A (en) | 2024-08-20 |
CN118515653B CN118515653B (en) | 2024-12-10 |
Family
ID=92276938
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202410986475.6A Active CN118515653B (en) | 2024-07-23 | 2024-07-23 | A kind of EZH2 specific inhibitor and its preparation method and use |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN118515653B (en) |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN113302184A (en) * | 2018-11-15 | 2021-08-24 | 日本新药株式会社 | 1,3, 4-oxadiazolinone compounds and drugs |
CN114555589A (en) * | 2020-08-10 | 2022-05-27 | 诺华公司 | Compounds and compositions for inhibiting EZH2 |
CN114746414A (en) * | 2019-09-26 | 2022-07-12 | 诺华公司 | Aza-quinoline compounds and uses thereof |
CN117384153A (en) * | 2022-07-12 | 2024-01-12 | 上海赛岚生物科技有限公司 | Methyltransferase inhibitors and uses thereof |
-
2024
- 2024-07-23 CN CN202410986475.6A patent/CN118515653B/en active Active
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN113302184A (en) * | 2018-11-15 | 2021-08-24 | 日本新药株式会社 | 1,3, 4-oxadiazolinone compounds and drugs |
CN114746414A (en) * | 2019-09-26 | 2022-07-12 | 诺华公司 | Aza-quinoline compounds and uses thereof |
CN114555589A (en) * | 2020-08-10 | 2022-05-27 | 诺华公司 | Compounds and compositions for inhibiting EZH2 |
CN117384153A (en) * | 2022-07-12 | 2024-01-12 | 上海赛岚生物科技有限公司 | Methyltransferase inhibitors and uses thereof |
Non-Patent Citations (2)
Title |
---|
RAM W. SABNIS: "Novel Quinoline Compounds as EZH2 Inhibitors for Treating Cancer", 《ACS MED. CHEM. LETT.》, 13 April 2022 (2022-04-13), pages 755 - 756 * |
尤启冬主编: "《药物化学》", 31 January 2004, 尤启冬主编, pages: 33 * |
Also Published As
Publication number | Publication date |
---|---|
CN118515653B (en) | 2024-12-10 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
KR102041442B1 (en) | Deuterated diaminopyrimidine compounds and pharmaceutical compositions comprising such compounds | |
CN118359609A (en) | Heterocyclic compounds, preparation methods and uses thereof | |
CN118908978A (en) | Tetrahydro-imidazo [4,5-c ] pyridine derivatives as PD-L1 immunomodulators | |
CA3133753A1 (en) | Novel small molecule inhibitors of tead transcription factors | |
CN105121443B (en) | Certain protein kinase inhibitors | |
CN102015707A (en) | RAF inhibitor compounds and methods of use thereof | |
CN102858754A (en) | RAF inhibitor compounds and methods of use thereof | |
TW201509938A (en) | Inhibitors of e1 activating enzymes | |
TW201247610A (en) | Nuclear hormone receptor modulators | |
CN104603134A (en) | Amido-benzyl sulfone and sulfoxide derivatives | |
CN115710266A (en) | Pyridopyrimidine derivative, preparation method thereof and application thereof in medicines | |
KR20190033538A (en) | Imidazopyridine amine phenyl derivatives and uses thereof | |
CN112955142A (en) | Functionalized heterocyclic compounds as antiviral agents | |
JP6972384B2 (en) | Heterocyclic compound | |
CN110167941A (en) | Substituted fused heteroaryl compounds are as kinase inhibitor and its application | |
CA3150701A1 (en) | Alkynyl quinazoline compounds | |
WO2022160931A1 (en) | Pyridopyrimidine derivative, preparation method therefor and use thereof | |
TW202200575A (en) | An immunosuppressant, preparation method and applications thereof having the potential of being developed into a new generation of PD-1/PD-L1 suppressants | |
JP2024530956A (en) | Polycyclic compounds and uses thereof | |
EP3189052B1 (en) | Camkii inhibitors and uses thereof | |
KR20220070229A (en) | Aza-quinoline compounds and uses thereof | |
WO2019085895A1 (en) | Class of amino-substituted nitrogen-containing fused ring compounds, preparation method therefor, and use thereof | |
WO2019085894A1 (en) | Nitrogen-containing fused ring compound, preparation method therefor, and use thereof | |
TWI723480B (en) | Fused ring derivatives used as fgfr4 inhibitors | |
CN116283799A (en) | Quinazoline methionine adenosyltransferase 2A inhibitors |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination | ||
GR01 | Patent grant | ||
GR01 | Patent grant |