[go: up one dir, main page]

CN116801881A - Opioid receptor agonist, preparation method and application thereof - Google Patents

Opioid receptor agonist, preparation method and application thereof Download PDF

Info

Publication number
CN116801881A
CN116801881A CN202380008359.0A CN202380008359A CN116801881A CN 116801881 A CN116801881 A CN 116801881A CN 202380008359 A CN202380008359 A CN 202380008359A CN 116801881 A CN116801881 A CN 116801881A
Authority
CN
China
Prior art keywords
compound
group
membered
alkyl
pharmaceutically acceptable
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.)
Pending
Application number
CN202380008359.0A
Other languages
Chinese (zh)
Inventor
罗明
聂岳坤
李政
喻龙江
肖文喜
刘军华
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tiandi Hengyi Pharmaceutical Co ltd
Original Assignee
Tiandi Hengyi Pharmaceutical Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Tiandi Hengyi Pharmaceutical Co ltd filed Critical Tiandi Hengyi Pharmaceutical Co ltd
Publication of CN116801881A publication Critical patent/CN116801881A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/08Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4433Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic 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
    • C07D405/02Heterocyclic 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 two hetero rings
    • C07D405/04Heterocyclic 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 two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic 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
    • C07D405/14Heterocyclic 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/113Spiro-condensed systems with two or more oxygen atoms as ring hetero atoms in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/12Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D495/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Pain & Pain Management (AREA)
  • Psychiatry (AREA)
  • Rheumatology (AREA)
  • Urology & Nephrology (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pulmonology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides oxaspiro small molecule compounds, a pharmaceutical composition containing the compounds and application of the compounds as therapeutic agents, particularly as MOR receptor agonists and in preparing medicaments for treating and/or preventing related diseases such as pain. The MOR receptor agonist with novel structure provided by the invention has high activity, higher selectivity to MOR and obviously improved maximum efficiency Emax.

Description

阿片受体激动剂及其制备方法和用途Opioid receptor agonists and preparation methods and uses thereof

本申请要求于2022年1月19日提交中国专利局、申请号为202210057938.1发明名称为“阿片受体激动剂及其制备方法和用途”的中国专利申请的优先权,其全部内容通过引用结合在本申请中。This application claims priority to the Chinese patent application filed with the China Patent Office on January 19, 2022, with application number 202210057938.1 and the invention title is "Opioid Receptor Agonist and Preparation Method and Use thereof", the entire content of which is incorporated by reference. in this application.

技术领域Technical field

本发明属于药物化学领域,具体涉及一类氧杂螺环类小分子化合物、其制备方法及含有该化合物的药物组合物以及其作为治疗剂,特别是作为MOR受体激动剂和在制备治疗和预防疼痛等相关疾病的药物中的用途。The invention belongs to the field of medicinal chemistry, and specifically relates to a class of oxaspirocyclic small molecule compounds, their preparation methods and pharmaceutical compositions containing the compounds as well as their use as therapeutic agents, especially as MOR receptor agonists and in the preparation of therapeutic and Use in medicines to prevent pain and other related diseases.

背景技术Background technique

阿片受体是一种G蛋白偶联受体(G Protein-Coupled Receptor,GPCR),是内源性阿片肽及阿片类药物结合的靶点。人体内存在多种阿片受体,主要包括μ阿片受体(Mu opioid receptor,MOR)、δ阿片受体(Delta Opioid Receptor,DOR)和κ阿片受体(Kappa opoid receptor,KOR)三类,广泛分布在中枢神经系统、心脏、消化道、血管、肾脏等外周组织(Nature,2016,537(7619):185)。MOR与吗啡肽的结合能力最强,是吗啡、芬太尼等镇痛药主要作用的受体蛋白位点。Zadina等研究发现MOR受体与吗啡肽1的结合能力(360pM)是DOR受体、KOR受体与吗啡肽1结合力的4000倍和15000倍(Science 2001 Vol.293 No:311-315;Biochem Biophys Res Commun 235:567-570;Life Sci 61:409-415)。The opioid receptor is a G Protein-Coupled Receptor (GPCR), which is the binding target of endogenous opioid peptides and opioid drugs. There are many types of opioid receptors in the human body, including three types: mu opioid receptor (MOR), delta opioid receptor (DOR) and kappa opoid receptor (KOR). Distributed in the central nervous system, heart, digestive tract, blood vessels, kidneys and other peripheral tissues (Nature, 2016, 537(7619):185). MOR has the strongest binding ability to morphine peptides and is the main receptor protein site for analgesics such as morphine and fentanyl. Zadina et al. found that the binding ability of MOR receptor to morphinin 1 (360pM) is 4000 times and 15000 times that of DOR receptor and KOR receptor and morphinin 1 (Science 2001 Vol. 293 No: 311-315; Biochem Biophys Res Commun 235:567-570; Life Sci 61:409-415).

研究发现GPCR介导及调控生理功能主要经由激活G蛋白途径和β-arrestin途径。G蛋白信号途径主要包括钙离子等第二信使系统、腺苷酸环化酶、丝裂原活化蛋白激酶等。β-arrestin途径主要有三个方面:(1)作为负性调控因子与GPCR激酶作用使GPCRs发生受体脱敏反应从而中止G蛋白信号转导;(2)作为支架蛋白募集胞吞蛋白诱导GPCR内吞;(3)作为接头蛋白与GPCR下游信号分子形成复合物,以G蛋白非依赖的方式激活信号转导分子。早期研究显示,内源性脑啡肽和阿片类药物埃托啡可以激动G蛋白并引发受体内吞,而吗啡则不引发受体内吞,这是因为吗啡通过G蛋白信号通路而不是β-arrestin途径来发挥其生理功能(Zhang等,Proc Natl Acad Sci USA,1998,95(12):7157-7162)。研究发现给β-arrestin2基因敲除小鼠注射吗啡后,由G蛋白信号介导的镇痛效果更强且维持时间更长(Bohn等,Science,1999年)。由此可见,配体刺激G蛋白和/或β-arrestin信号的差异决定了GPCR的配体特异性细胞生物学效应,如果此类配体的负性β-arrestin偏爱性更强,甚至可以逃脱β-arrestin介导的受体脱敏,则G蛋白信号传递时间延长,镇痛作用更强。近年来研究发现,β-arrestin通路与MOR激动剂多个副作用相关,如便秘、呼吸抑制和镇痛耐受(Science 1999 Vol.286:2495-2498:J.Pharmacol.Exp.Ther.2005,314:1195-1201)。因此,研发一种可选择性激活G蛋白信号通路的“偏向性”MOR激动剂药物,即MOR的负性β-arrestin偏爱性配体设计药物,使β-arrestin介导的副作用降低,在镇痛领域具有显著的临床价值和社会意义。Studies have found that GPCR mediates and regulates physiological functions mainly through activating the G protein pathway and β-arrestin pathway. The G protein signaling pathway mainly includes second messenger systems such as calcium ions, adenylyl cyclase, mitogen-activated protein kinase, etc. The β-arrestin pathway has three main aspects: (1) As a negative regulatory factor, it interacts with GPCR kinases to cause receptor desensitization of GPCRs, thus terminating G protein signal transduction; (2) As a scaffold protein, it recruits endocytic proteins to induce GPCR internalization. Endocytosis; (3) As an adapter protein, it forms a complex with GPCR downstream signaling molecules and activates signal transduction molecules in a G protein-independent manner. Early studies have shown that endogenous enkephalins and the opioid etorphine can activate G proteins and trigger receptor endocytosis, while morphine does not trigger receptor endocytosis. This is because morphine activates G protein signaling pathways rather than β -arrestin pathway to exert its physiological functions (Zhang et al., Proc Natl Acad Sci USA, 1998, 95(12):7157-7162). Studies have found that after injecting morphine into β-arrestin2 knockout mice, the analgesic effect mediated by G protein signaling is stronger and lasts longer (Bohn et al., Science, 1999). It can be seen that the difference in ligand-stimulated G protein and/or β-arrestin signals determines the ligand-specific cell biological effects of GPCRs. If such ligands have a stronger negative β-arrestin preference, they can even escape Receptor desensitization mediated by β-arrestin prolongs the G protein signal transmission time and increases the analgesic effect. In recent years, studies have found that the β-arrestin pathway is related to multiple side effects of MOR agonists, such as constipation, respiratory depression and analgesic tolerance (Science 1999 Vol. 286: 2495-2498: J. Pharmacol. Exp. Ther. 2005, 314 :1195-1201). Therefore, it is necessary to develop a "biased" MOR agonist drug that can selectively activate the G protein signaling pathway, that is, a negative β-arrestin-biased ligand design drug for MOR, which can reduce the side effects mediated by β-arrestin. The field of pain has significant clinical value and social significance.

FDA于2020年8月批准了Trevena Inc公司药物Olinvyk(WO2012129495)的上市申请,目前关于G蛋白偏向性MOR激动剂的研发报道的专利有WO2017063509A1、WO2019205983A1、CN10920641A、WO2019072235A1、CN111662284A、WO2019052557A1等,虽然这些专利已经公开了一系列G蛋白偏向性MOR激动剂,但其分子结构与本发明提供的结构具有较大区别,且其药效、安全性暂未得到证实,临床上仍需要开发新的分子结构,以获得具有更好的药效、选择性、药用安全性、药物代谢结果的MOR激动剂。 The FDA approved the marketing application of Trevena Inc's drug Olinvyk (WO2012129495) in August 2020. The current patents reported on the research and development of G protein-biased MOR agonists include WO2017063509A1, WO2019205983A1, CN10920641A, WO2019072235A1, CN111662284A, WO 2019052557A1, etc., although these Patents have disclosed a series of G protein-biased MOR agonists, but their molecular structures are quite different from those provided by the present invention, and their efficacy and safety have not yet been confirmed, and new molecular structures still need to be developed clinically. , to obtain MOR agonists with better efficacy, selectivity, medicinal safety, and drug metabolism results.

发明内容Contents of the invention

针对现有技术的需求,本发明提供了一种结构新颖的可作为MOR受体激动剂的化合物,该类化合物表现出高活性,Emax也具有明显改善,以及对MOR较高的选择性。In response to the needs of the existing technology, the present invention provides a compound with a novel structure that can be used as a MOR receptor agonist. This type of compound exhibits high activity, a significant improvement in E max , and a high selectivity for MOR.

本发明第一方面提供了如式(Ⅳ)或(Ⅴ)或(Ⅵ)所示的化合物、其溶剂化物、立体异构体、氘代化合物、或其药学可接受的盐, The first aspect of the present invention provides compounds represented by formula (IV) or (V) or (VI), their solvates, stereoisomers, deuterated compounds, or pharmaceutically acceptable salts thereof,

其中,环B、环C各自独立的选自被取代的或未被取代的芳基、被取代的或未被取代的杂芳基;Wherein, Ring B and Ring C are each independently selected from substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl;

环D选自环烷基、杂环烷基;Ring D is selected from cycloalkyl and heterocycloalkyl;

R4、R5各自独立的选自H、氘原子、烷基、氧代、烷氧基、羟基、卤素、氰基、炔基、烯基、-(CH2)g-O-3至12元杂环基、-(CH2)g-O-3至12元环烷基、-(CH2)g-3至12元环烷基、-(CH2)g-3至12元杂环基、5至10元杂芳基、5至10元芳基、-S(=O)f-C1-6烷基、-O-C2-6炔基、-O-C2-6烯基;其中,所述杂环基、杂芳基、芳基、烷基、炔基、烯基、烷氧基可任选进一步被1至3个R6所取代;R 4 and R 5 are each independently selected from H, deuterium atom, alkyl group, oxo, alkoxy group, hydroxyl, halogen, cyano group, alkynyl group, alkenyl group, -(CH 2 ) g -O-3 to 12 Membered heterocyclyl, -(CH 2 ) g -O-3 to 12-membered cycloalkyl, -(CH 2 ) g -3 to 12-membered cycloalkyl, -(CH 2 ) g -3 to 12-membered heterocycle base, 5- to 10-membered heteroaryl, 5- to 10-membered aryl, -S(=O) f -C 1-6 alkyl, -OC 2-6 alkynyl, -OC 2-6 alkenyl; wherein, The heterocyclic group, heteroaryl group, aryl group, alkyl group, alkynyl group, alkenyl group, and alkoxy group may be optionally further substituted by 1 to 3 R 6 ;

其中,R6各自独立的选自氘原子、卤素、-OH、-C1-6烷基、-C1-6烷基-O-C1-6烷基、-O-C1-6烷基、3至6元环烷基、-O-C2-6炔基、-O-C2-6烯基、-C2-6炔基、-C2-6烯基、氨基、羧酸酯基、硝基、氰基、羟烷基、杂环基、芳基和杂芳基;Among them, R 6 is each independently selected from deuterium atom, halogen, -OH, -C 1-6 alkyl, -C 1-6 alkyl-OC 1-6 alkyl, -OC 1-6 alkyl, 3 to 6-membered ring alkyl, -OC 2-6 alkynyl, -OC 2-6 alkenyl, -C 2-6 alkynyl, -C 2-6 alkenyl, amino, carboxylate group, nitro, cyano group , hydroxyalkyl, heterocyclyl, aryl and heteroaryl;

所述g选自0、1、2、3、4、5、6;The g is selected from 0, 1, 2, 3, 4, 5, 6;

所述f选自0、1、2;The f is selected from 0, 1, 2;

p、q、L各自独立地为0、1、2、3或4;p, q, L are each independently 0, 1, 2, 3 or 4;

所述杂芳基、所述杂环烷基或所述杂环基上的杂原子各自独立地选自O、S或N。The heteroatom on the heteroaryl group, the heterocycloalkyl group or the heterocyclyl group is each independently selected from O, S or N.

在本发明提供的一些实施例中,所述化合物选自式(Ⅶ)或(Ⅷ)或(Ⅸ)或(Ⅹ)或(Ⅺ)或(Ⅻ): In some embodiments provided by the present invention, the compound is selected from formula (VII) or (VIII) or (IX) or (X) or (XI) or (XII):

在本发明提供的一些实施例中,所述化合物选自下式结构: In some embodiments provided by the present invention, the compound is selected from the following formula:

在本发明提供的一些实施例中,所述化合物选自下式结构: In some embodiments provided by the present invention, the compound is selected from the following formula:

在本发明提供的一些实施例中,所述化合物选自下式结构: In some embodiments provided by the present invention, the compound is selected from the following formula:

优选的,所述的化合物、其溶剂化物、立体异构体、氘代化合物、或其药学可接受的盐,所述化合物选自下式结构: Preferably, the compound, its solvate, stereoisomer, deuterated compound, or pharmaceutically acceptable salt thereof, the compound is selected from the following formula:

其中,R4独立的选自氘原子、烷基、氧代、烷氧基、羟基、卤素、氰基、炔基、烯基、-(CH2)g-O-3至12元杂环基、-(CH2)g-O-3至12元环烷基、-(CH2)g-3至12元环烷基、-(CH2)g-3至12元杂环基、5至10元杂芳基、5至10元芳基、-S(=O)f-C1-6烷基、-O-C2-6炔基、-O-C2-6烯基;Among them, R 4 is independently selected from deuterium atom, alkyl group, oxo, alkoxy group, hydroxyl group, halogen, cyano group, alkynyl group, alkenyl group, -(CH 2 ) g -O-3 to 12-membered heterocyclic group , -(CH 2 ) g -O-3 to 12-membered cycloalkyl, -(CH 2 ) g -3 to 12-membered cycloalkyl, -(CH 2 ) g -3 to 12-membered heterocyclyl, 5 to 10-membered heteroaryl, 5 to 10-membered aryl, -S(=O) f -C 1-6 alkyl, -OC 2-6 alkynyl, -OC 2-6 alkenyl;

q选自1、2、3或4。q is selected from 1, 2, 3 or 4.

本发明提供的一些优选实施例中,化合物选自: In some preferred embodiments provided by the present invention, the compound is selected from:

本发明的第二方面涉及一种药物组合物,其包括本发明第一方面所示的化合物、其溶剂化物、立体异构 体、氘代化合物或其药学可接受的盐,以及药学上可接受的载体。The second aspect of the present invention relates to a pharmaceutical composition, which includes the compound shown in the first aspect of the present invention, its solvate, stereoisomer body, deuterated compound or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

本发明的第三方面提供了本发明第一方面所示的化合物、其溶剂化物、立体异构体、氘代化合物或其药学可接受的盐,或本发明第二方面所述的药物组合物在制备预防和/或治疗MOR受体激动剂介导的相关疾病的药物中的用途。The third aspect of the present invention provides the compounds shown in the first aspect of the present invention, their solvates, stereoisomers, deuterated compounds or pharmaceutically acceptable salts thereof, or the pharmaceutical compositions described in the second aspect of the present invention. Use in the preparation of drugs for preventing and/or treating MOR receptor agonist-mediated related diseases.

本发明提供的一些优选实施例中,MOR受体激动剂介导的相关疾病选自疼痛、免疫功能障碍、炎症、食管回流、神经和精神疾病、泌尿和生殖疾病、心血管疾病和呼吸疾病;优选地,所述疼痛选自术后疼痛、癌症引起的疼痛、神经性疼痛、创伤性疼痛和炎症引起的疼痛。In some preferred embodiments provided by the present invention, the related diseases mediated by MOR receptor agonists are selected from pain, immune dysfunction, inflammation, esophageal reflux, neurological and psychiatric diseases, urinary and reproductive diseases, cardiovascular diseases and respiratory diseases; Preferably, the pain is selected from the group consisting of postoperative pain, cancer-induced pain, neuropathic pain, traumatic pain and inflammation-induced pain.

术语解释Terminology explanation

本发明所用术语“C2-6炔基”指含有碳碳叁键的2~6个碳原子的炔烃部分去除一个氢原子衍生的直链或支链的炔烃基,如乙炔基、丙炔基、2-丁炔基、2-戊炔基、3-戊炔基、4-甲基-2-戊炔基、2-己炔基、3-己炔基等。The term "C 2-6 alkynyl" used in the present invention refers to a linear or branched alkynyl group derived from an alkyne moiety of 2 to 6 carbon atoms containing a carbon-carbon triple bond by removing one hydrogen atom, such as ethynyl, propyne base, 2-butynyl, 2-pentynyl, 3-pentynyl, 4-methyl-2-pentynyl, 2-hexynyl, 3-hexynyl, etc.

本发明上述的“环烷基”包括可能形成的所有单环、稠环(包括以并、螺、桥的形式稠合)的情形;例如:“3-12元环烷基”,可以是单环、双环、或者多环环烷基系统(也称为稠环系统)。在不特别指明的情况下,单环系统是含3-8个碳原子的环烃基基团,实例包括但不限于:环丙烷基、环丁烷基、环戊烷基、环己烷基、环庚烷基、环辛烷基等。The above-mentioned "cycloalkyl" in the present invention includes all possible single rings and fused rings (including fused in the form of parallel, spiro, and bridge); for example: "3-12-membered cycloalkyl" can be a single ring. cyclic, bicyclic, or polycyclic cycloalkyl systems (also called fused ring systems). Unless otherwise specified, a monocyclic system is a cyclic hydrocarbyl group containing 3 to 8 carbon atoms. Examples include but are not limited to: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, Cycloheptyl, cyclooctyl, etc.

稠环环烷基包括并环环烷基、桥环烷基、螺环烷基。Fused-ring cycloalkyl groups include pendant cycloalkyl groups, bridged cycloalkyl groups, and spirocycloalkyl groups.

并环环烷基可以为6-11元并环环烷基、7-10元并环环烷基,其代表性例子包括但不限于双环[3.1.1]庚烷、双环[2.2.1]庚烷、双环[2.2.2]辛烷、双环[3.2.2]壬烷、双环[3.3.1]壬烷和双环[4.2.1]壬烷。The cyclic cycloalkyl group can be a 6-11 membered cyclic cycloalkyl group or a 7-10 membered cyclic cycloalkyl group. Representative examples include but are not limited to bicyclo[3.1.1]heptane, bicyclo[2.2.1] Heptane, bicyclo[2.2.2]octane, bicyclo[3.2.2]nonane, bicyclo[3.3.1]nonane and bicyclo[4.2.1]nonane.

螺环烷基可以为7-12元螺环烷基、7-11元螺环烷基,其实例包括但不限于: 的基团。The spirocycloalkyl group can be a 7-12-membered spirocycloalkyl group or a 7-11-membered spirocycloalkyl group. Examples include but are not limited to: group.

上述的桥环烷基可以为6-11元桥环烷基、7-10元桥环烷基,其实例包括但不限于: 的基团。The above-mentioned bridged cycloalkyl group can be a 6-11-membered bridged cycloalkyl group or a 7-10-membered bridged cycloalkyl group. Examples thereof include but are not limited to: group.

本文所用术语“杂环烷基”(或“杂脂环”)是指一价的单环的非芳香族环系,其环原子由碳原子及选自氮、氧、硫和磷的杂原子构成,并且通过一个单键连接至母核或其他基团;常见的杂环烷基包括(但不限于)环氧乙烷基、氧杂环丁烷-3-基、氮杂环丁烷-3-基、四氢呋喃-2-基、吡咯烷-1-基、吡咯烷-2-基、四氢-2氢-吡喃-2-基、四氢-2氢-吡喃-4-基、哌啶-2-基、哌啶-4-基等。The term "heterocycloalkyl" (or "heteroalicyclic") as used herein refers to a monovalent monocyclic non-aromatic ring system in which the ring atoms are composed of carbon atoms and heteroatoms selected from nitrogen, oxygen, sulfur and phosphorus. composed of, and connected to the parent core or other groups through a single bond; common heterocycloalkyl groups include (but are not limited to) oxiranyl, oxetan-3-yl, azetidine- 3-yl, tetrahydrofuran-2-yl, pyrrolidin-1-yl, pyrrolidin-2-yl, tetrahydro-2hydro-pyran-2-yl, tetrahydro-2hydro-pyran-4-yl, Piperidin-2-yl, piperidin-4-yl, etc.

本文所用术语“杂环基”指3-12元的至少一个环碳原子被选自O、S、N的杂原子替代的非芳香性的环状基团,优选1-3个杂原子,同时包括碳原子、氮原子和硫原子可以被氧代。“3-12元杂环基”,是指单环杂环基、双环杂环基系统或多环杂环基系统(也称为稠环系统),包括饱和、部分饱和的杂环基,但不包括芳环。在不特别指明的情况下,包括可能形成的所有单环、稠环(包括以并、螺、桥的形式稠合)、饱和、部分饱和的情形。The term "heterocyclyl" as used herein refers to a 3-12 membered non-aromatic cyclic group in which at least one ring carbon atom is replaced by a heteroatom selected from O, S, and N, preferably 1-3 heteroatoms, and at the same time Including carbon atoms, nitrogen atoms and sulfur atoms can be oxygenated. "3-12 membered heterocyclyl" refers to a monocyclic heterocyclyl, a bicyclic heterocyclyl system or a polycyclic heterocyclyl system (also known as a fused ring system), including saturated and partially saturated heterocyclyl, but Excludes aromatic rings. Unless otherwise specified, it includes all single rings, fused rings (including fused in the form of union, spiro, and bridge), saturated, and partially saturated rings that may be formed.

单杂环基可以为3-8元单杂环基、3-6元单杂环基、4-7元单杂环基、5-7元单杂环基、5-6元单杂环基、5-6元含氧单杂环基、3-8元含氮单杂环基、5-6元含氮单杂环基、5-6元饱和单杂环基等,其实例包括但不限于: 的基团。The monoheterocyclyl group can be a 3-8-membered monoheterocyclyl group, a 3-6-membered monoheterocyclyl group, a 4-7-membered monoheterocyclyl group, a 5-7-membered monoheterocyclyl group, or a 5-6-membered monoheterocyclyl group. , 5-6-membered oxygen-containing monoheterocyclyl, 3-8-membered nitrogen-containing monoheterocyclyl, 5-6-membered nitrogen-containing monoheterocyclyl, 5-6-membered saturated monoheterocyclyl, etc. Examples include but are not Limited to: group.

稠杂环包括并杂环基、螺杂环基、桥杂环基,可以是饱和的、部分饱和的或不饱和的,但不是芳香性的。Condensed heterocycles include pendant heterocyclyl, spiroheterocyclyl and bridged heterocyclyl, which can be saturated, partially saturated or unsaturated, but are not aromatic.

并杂环基可以为6-12元并杂环基、7-10元并杂环基、6-10元并杂环基、6-12元饱和并杂环基,代表性实例包括但不限于:的基团。The heterocyclyl group can be a 6-12-membered heterocyclyl group, a 7-10-membered heterocyclyl group, a 6-10-membered heterocyclyl group, or a 6-12-membered saturated heterocyclyl group. Representative examples include but are not limited to : group.

螺杂环基可以为6-12元螺杂环基、7-11元螺杂环基、6-12元饱和螺环基,其实例包括但不限于: 的基团;The spiroheterocyclyl group can be a 6-12-membered spiroheterocyclyl group, a 7-11-membered spiroheterocyclyl group, or a 6-12-membered saturated spiroheterocyclyl group. Examples thereof include but are not limited to: group;

上述的桥杂环基可以为6-12元桥杂环基、7-11元桥杂环基、6-12元饱和桥环基,其实例包括但不限于:的基团。The above-mentioned bridged heterocyclyl group can be a 6-12-membered bridged heterocyclyl group, a 7-11-membered bridged heterocyclyl group, or a 6-12-membered saturated bridged heterocyclyl group. Examples thereof include but are not limited to: group.

本文所用术语“芳基”(或“芳环”)是指一价的单环或多环(包含稠合形式)的芳香族环系,其仅有碳原子和氢原子构成;常见的芳基包括(但不限于)苯基、萘基、蒽基、菲基、苊基、薁基、芴基、茚基、芘基等。上述芳基也包括杂环基并芳基、环烷基并芳基;The term "aryl" (or "aryl ring") used herein refers to a monovalent monocyclic or polycyclic (including fused form) aromatic ring system, which is composed only of carbon atoms and hydrogen atoms; common aryl groups Including (but not limited to) phenyl, naphthyl, anthracenyl, phenanthrenyl, acenaphthyl, azulenyl, fluorenyl, indenyl, pyrenyl, etc. The above-mentioned aryl groups also include heterocyclyl aryl groups and cycloalkyl aryl groups;

本文所用术语“杂芳基”(或“杂芳环”)是指一价的单环或多环(包含稠合形式)的芳香族环系,其环原子由碳原子及选自氮、氧、硫和磷的杂原子构成;常见的杂芳基包括(但不限于)苯并吡咯基、苯并呋喃基、苯并噻吩基、苯并咪唑基、苯并噁唑基、苯并噻唑基、氮杂环丁烷基、咔唑基、吡咯基、呋喃基、噻吩基、咪唑基、噁唑基、噻唑基、吡唑基、异噁唑基、异噻唑基、吲唑基、吲嗪基、吲哚基、喹啉基、异喹啉基、吩嗪基、吩噁嗪基、吩噻嗪基、蝶啶基、嘌呤基、吡嗪基、嘧啶基、哒嗪基、吡啶基、三唑基、四唑基等。上述杂芳基也包括杂环基并杂芳基、环烷基并杂芳基。The term "heteroaryl" (or "heteroaryl ring") as used herein refers to a monovalent monocyclic or polycyclic (including fused forms) aromatic ring system, the ring atoms of which are carbon atoms and selected from nitrogen, oxygen , composed of heteroatoms of sulfur and phosphorus; common heteroaryl groups include (but are not limited to) benzopyrrolyl, benzofuranyl, benzothienyl, benzimidazolyl, benzoxazolyl, benzothiazolyl , azetidinyl, carbazolyl, pyrrolyl, furyl, thienyl, imidazolyl, oxazolyl, thiazolyl, pyrazolyl, isoxazolyl, isothiazolyl, indazolyl, indazine base, indolyl, quinolyl, isoquinolinyl, phenazinyl, phenoxazinyl, phenothiazinyl, pteridinyl, purinyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyridyl, Triazolyl, tetrazolyl, etc. The above-mentioned heteroaryl group also includes heterocyclylheteroaryl and cycloalkylheteroaryl.

本发明所示化合物的“立体异构体”是指当化合物存在不对称碳原子时,会产生对映异构体;当化合物存在碳碳双键或环状结构时,会产生顺反异构体;当化合物存在酮或肟时,会产生互变异构体,所有化合物的对映异构体、非对映异构体、消旋异构体、顺反异构体、互变异构体、几何异构体、差向异构体及其混合物,均包括在本发明范围中。The "stereoisomer" of the compound shown in the present invention means that when asymmetric carbon atoms exist in the compound, enantiomers will be produced; when a carbon-carbon double bond or cyclic structure exists in the compound, cis-trans isomers will occur body; when ketones or oximes are present in a compound, tautomers will be produced, including enantiomers, diastereomers, racemic isomers, cis-trans isomers, and tautomers of all compounds Isomers, geometric isomers, epimers and mixtures thereof are all included in the scope of the present invention.

具体实施方式Detailed ways

为使本发明的目的、技术方案、及优点更加清楚明白,以下举实施例,对本发明进一步详细说明。显然,所描述的实施例仅仅是本发明一部分实施例,而不是全部的实施例。基于本发明中的实施例,本领域普通技术人员所获得的所有其他实施例,都属于本发明保护的范围。In order to make the purpose, technical solutions, and advantages of the present invention more clear, the following examples are given to further describe the present invention in detail. Obviously, the described embodiments are only some of the embodiments of the present invention, but not all of the embodiments. Based on the embodiments of the present invention, all other embodiments obtained by those of ordinary skill in the art fall within the scope of protection of the present invention.

如本文所用,室温是指约20-30℃;“过夜”是指约10h~16h;1M、1N为1mmol/L,1μM为1μmol/L,1mM为1mmol/L,1nM为1nmol/L;eq:当量;As used herein, room temperature refers to about 20-30°C; "overnight" refers to about 10h to 16h; 1M, 1N is 1mmol/L, 1μM is 1μmol/L, 1mM is 1mmol/L, 1nM is 1nmol/L; eq :equivalent;

收率或产率=实际合成产物质量/理论合成产物质量×100%。 Yield or productivity = mass of actual synthesized product/mass of theoretical synthesized product × 100%.

实施例1Example 1

(1R,4R)-4-乙氧基-N-{2-[9-(吡啶-2-基)-6-氧杂螺[4.5]癸-2-烯-9-基]乙基}-1,2,3,4-四氢萘-1-胺(化合物141)的合成 (1R,4R)-4-ethoxy-N-{2-[9-(pyridin-2-yl)-6-oxaspiro[4.5]dec-2-en-9-yl]ethyl}- Synthesis of 1,2,3,4-tetralin-1-amine (compound 141)

步骤1:2-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-2-烯-9-基)乙酸(141-2)的合成Step 1: Synthesis of 2-9-(pyridin-2-yl)-6-oxaspiro[4.5]dec-2-en-9-yl)acetic acid (141-2)

室温下,将化合物141-1(4.66g,18.32mmol,1.0eq)溶解于1M HCl水溶液(23mL)中,升温至100℃后,在N2保护下搅拌反应16小时。待反应结束后,待其自然冷却至室温,水相用乙酸乙酯(100mL×3)洗涤,然后将水相用饱和NaOH水溶液调节pH到~13,用乙酸乙酯(100mL×3)洗涤,将水相用1M HCl水溶液调节pH到4~6,用乙酸乙酯(100mL×5)萃取。合并萃取后的有机相用盐水(100mL×2)洗涤,用无水硫酸钠干燥,过滤,滤液真空下浓缩得到淡黄色固体化合物141-2(4.52g,产率90.22%,[M+H]+:274.17。Compound 141-1 (4.66g, 18.32mmol, 1.0eq) was dissolved in 1M HCl aqueous solution (23mL) at room temperature. After the temperature was raised to 100°C, the reaction was stirred for 16 hours under N2 protection. After the reaction is completed, allow it to cool to room temperature naturally, wash the aqueous phase with ethyl acetate (100 mL The aqueous phase was adjusted to pH 4-6 with 1M HCl aqueous solution, and extracted with ethyl acetate (100 mL×5). The extracted organic phases were combined, washed with brine (100 mL × 2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under vacuum to obtain light yellow solid compound 141-2 (4.52 g, yield 90.22%, [M+H] + : 274.17.

步骤2:(R)-2-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-2-烯-9-基)乙酸和(S)苯乙胺盐(141-3)的合成Step 2: (R)-2-9-(pyridin-2-yl)-6-oxaspiro[4.5]dec-2-en-9-yl)acetic acid and (S) phenylethylamine salt (141-3 )Synthesis

室温下,向100mL的单口瓶中加入乙醇(EtOH,30mL)和化合物141-2(4.52g,16.46mmol),加热到50℃,在50℃下搅拌0.5小时,在50℃下缓慢滴加S-苯乙胺和乙醇(15mL)的溶液。加料完毕后,升温到80℃搅拌1小时。缓慢冷却到室温,并在室温下搅拌12小时。析出白色固体,过滤,固体用乙醇(10mL×2)洗涤2次,收集得到的白色固体加入到30mL的乙醇中,加热到80℃,待其溶清。在80℃下搅拌0.5小时,逐渐冷却到室温,待白色固体析出后,过滤,滤饼用乙醇(10mL×2)洗涤2次,收集固体,得到白色固体141-3(1.2g,e.e.%:99.6%)。At room temperature, add ethanol (EtOH, 30 mL) and compound 141-2 (4.52 g, 16.46 mmol) into a 100 mL single-neck bottle, heat to 50°C, stir at 50°C for 0.5 hours, and slowly add S dropwise at 50°C. - A solution of phenylethylamine and ethanol (15 mL). After the addition is completed, the temperature is raised to 80°C and stirred for 1 hour. Cool slowly to room temperature and stir at room temperature for 12 hours. A white solid precipitated, filtered, and the solid was washed twice with ethanol (10 mL × 2). The white solid obtained was collected and added to 30 mL of ethanol, heated to 80°C, and allowed to dissolve. Stir at 80°C for 0.5 hours, gradually cool to room temperature, filter after the white solid precipitates, wash the filter cake twice with ethanol (10mL×2), collect the solid, and obtain white solid 141-3 (1.2g, e.e.%: 99.6%).

步骤3:(R)-2-9-(吡啶-2-基)-6-氧杂螺[4.5]癸-2-烯-9-基)乙酸(141-4)的合成Step 3 Synthesis of (R)-2-9-(pyridin-2-yl)-6-oxaspiro[4.5]dec-2-en-9-yl)acetic acid (141-4)

将化合物141-3(1.2g)溶于20mL的水中,用1M氢氧化钠溶液调节pH>9,用二氯甲烷(50mL×3)洗涤,水相用1N盐酸溶液调节pH=4~6后用二氯甲烷(50mL×5)萃取,合并萃取有机相并用饱和食盐水(50mL×1)洗涤,无水硫酸钠干燥,过滤,旋干滤液,得到淡黄色油状化合物141-4(600mg)。 Dissolve compound 141-3 (1.2g) in 20mL of water, adjust the pH to >9 with 1M sodium hydroxide solution, wash with dichloromethane (50mL×3), and adjust the aqueous phase to pH=4~6 with 1N hydrochloric acid solution. Extract with dichloromethane (50 mL × 5), combine the extracted organic phases, wash with saturated brine (50 mL × 1), dry over anhydrous sodium sulfate, filter, and spin the filtrate to obtain light yellow oily compound 141-4 (600 mg).

步骤4:氮-甲基-氮-甲氧基-(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烯烃-9-基)乙酰胺(141-5)的合成Step 4: Nitrogen-methyl-nitrogen-methoxy-(R)-2-(9-(pyridin-2-yl)-6-oxaspiro[4.5]decene-9-yl)acetamide (141 -5) synthesis

室温下,将化合物141-4(600mg,2.2mmol,1.0eq)用二氯甲烷(30mL)溶解,然后依次加入甲氧基(甲基)胺盐酸盐(256.93mg,2.63mmol,1.2eq)、1-乙基-3(3-二甲基丙胺)碳二亚胺)(EDCI)(631.22mg,3.29mmol,1.5eq)和4-二甲氨基吡啶(DMAP)(26.82mg,219.51μmol,0.1eq),在N2保护下搅拌0.5h后,再加入N,N-二异丙基乙胺(DIPEA)(851.17mg,6.59mmol,3.0eq)搅拌反应过夜。待反应结束后,往反应液中加入NH4Cl饱和溶液(100mL)淬灭,再用二氯甲烷(100mL×5)萃取,合并有机相并用饱和食盐水(100mL×2)洗涤,无水Na2SO4干燥,过滤,滤液通过减压浓缩得到黄色稠状物141-5(592mg,产率:85.24%),[M+H]+:317.17。At room temperature, compound 141-4 (600 mg, 2.2 mmol, 1.0 eq) was dissolved in dichloromethane (30 mL), and then methoxy (methyl)amine hydrochloride (256.93 mg, 2.63 mmol, 1.2 eq) was added sequentially. , 1-ethyl-3(3-dimethylpropylamine)carbodiimide) (EDCI) (631.22mg, 3.29mmol, 1.5eq) and 4-dimethylaminopyridine (DMAP) (26.82mg, 219.51μmol, 0.1eq), stir for 0.5h under N2 protection, then add N,N-diisopropylethylamine (DIPEA) (851.17mg, 6.59mmol, 3.0eq) and stir the reaction overnight. After the reaction is completed, NH 4 Cl saturated solution (100 mL) was added to the reaction solution to quench, and then extracted with dichloromethane (100 mL × 5). The organic phases were combined and washed with saturated brine (100 mL × 2), and anhydrous Na Dried over 2 SO 4 and filtered, the filtrate was concentrated under reduced pressure to obtain yellow thick substance 141-5 (592 mg, yield: 85.24%), [M+H] + : 317.17.

步骤5:化合物氮-甲基-氮-甲氧基-(R)-2-(9-(吡啶-2-基)-6-氧杂螺[4.5]癸烯烃-9-基)乙醛(141-6)的合成Step 5: Compound nitrogen-methyl-nitrogen-methoxy-(R)-2-(9-(pyridin-2-yl)-6-oxaspiro[4.5]decene-9-yl)acetaldehyde ( 141-6) synthesis

室温下,将化合物141-5(438.6mg,1.39mmol,1.0eq)用甲苯(Tol,7mL)溶解,在-40℃、N2保护下,缓慢滴加红铝(Red-Al)(657.9mg,2.28mmol,1.05eq),加料完毕后搅拌反应4h。待反应结束后,往反应液中加入质量分数为10%的柠檬酸溶液(20mL)淬灭,再移至室温,向反应液中加入质量分数为10%柠檬酸水溶液(10mL),再移至室温,用1mol/L的HCl溶液调pH=2~3,用乙酸乙酯(30mL×1)萃取;水相再用5N NaOH溶液调pH=11~13,用二氯甲烷(50mL×3)萃取,合并有机相并用饱和食盐水(50mL×1)洗涤,用无水硫酸钠干燥,过滤,滤液通过减压浓缩得到橘红色稠状物141-6(287.2mg),收率:80.5%,[M+H]+:258.16。Compound 141-5 (438.6 mg, 1.39 mmol, 1.0 eq) was dissolved in toluene (Tol, 7 mL) at room temperature, and red aluminum (Red-Al) (657.9 mg) was slowly added dropwise at -40°C under N 2 protection. , 2.28mmol, 1.05eq), stir and react for 4 hours after the addition is completed. After the reaction is completed, add a 10% mass fraction of citric acid solution (20 mL) to the reaction solution to quench it, then move to room temperature, add a mass fraction of 10% citric acid aqueous solution (10 mL) to the reaction solution, and then move to At room temperature, use 1 mol/L HCl solution to adjust pH=2~3, and extract with ethyl acetate (30mL×1); use 5N NaOH solution to adjust the aqueous phase to pH=11~13, and use methylene chloride (50mL×3) Extract, combine the organic phases and wash with saturated brine (50 mL × 1), dry with anhydrous sodium sulfate, filter, and the filtrate is concentrated under reduced pressure to obtain orange-red thick substance 141-6 (287.2 mg), yield: 80.5%. [M+H] + :258.16.

步骤6:(1R,4R)-4-乙氧基-N-{2-[9-(吡啶-2-基)-6-氧杂螺[4.5]癸-2-烯-9-e]乙基}-1,2,3,4-四氢萘-1-胺(141)的合成Step 6: (1R, 4R)-4-ethoxy-N-{2-[9-(pyridin-2-yl)-6-oxaspiro[4.5]dec-2-ene-9-e]ethyl Synthesis of 1,2,3,4-tetralin-1-amine (141)

室温下,将化合物141-6(50mg,0.19mmol),化合物141-7(37.2mg,0.19mmol)溶于二氯甲烷(DCM)(3mL),再加入MgSO4(116.9mg,0.97mmol),在氮气保护下,室温搅拌12h,再加入硼氢化钠(22.1mg,0.58mmol),并搅拌2h,再加入甲醇(1mL),并搅拌0.5h。再用硅藻土过滤,滤液室温浓缩,大板(V 油醚(PE):V乙酸乙酯(EA)=0:1)纯化得到黄色稠状物141(35mg,产率41.7%),[M+H]+:433.40。Compound 141-6 (50 mg, 0.19 mmol) and compound 141-7 (37.2 mg, 0.19 mmol) were dissolved in dichloromethane (DCM) (3 mL) at room temperature, and then MgSO 4 (116.9 mg, 0.97 mmol) was added. Under nitrogen protection, stir at room temperature for 12 h, then add sodium borohydride (22.1 mg, 0.58 mmol), stir for 2 h, then add methanol (1 mL), and stir for 0.5 h. Then filter through diatomaceous earth, and the filtrate is concentrated at room temperature, and purified by large plate (V petroleum ether (PE) : V ethyl acetate (EA) = 0:1) to obtain yellow thick substance 141 (35 mg, yield 41.7%). [M+H] + :433.40.

1H NMR(400MHz,CDCl3)δ8.60-8.56(m,1H),7.70-7.64(m,1H),7.36-7.32(m,2H),7.26-7.19(m,3H),7.18-7.13(m,1H),5.66-5.61(m,1H),5.49-5.45(m,1H),4.39-4.35(m,1H),3.95-3.88(m,1H),3.87-3.82(m,1H),3.71-3.64(m,2H),3.56-3.51(m,1H),2.59-2.49(m,3H),2.47-2.38(m,2H),2.29-2.22(m,1H),2.12-1.98(m,5H),1.96-1.90(m,1H),1.87-1.77(m,3H),1.30-1.20(m,5H). 1 H NMR (400MHz, CDCl 3 ) δ 8.60-8.56 (m, 1H), 7.70-7.64 (m, 1H), 7.36-7.32 (m, 2H), 7.26-7.19 (m, 3H), 7.18-7.13 (m, 1H), 5.66-5.61 (m, 1H), 5.49-5.45 (m, 1H), 4.39-4.35 (m, 1H), 3.95-3.88 (m, 1H), 3.87-3.82 (m, 1H) , 3.71-3.64(m, 2H), 3.56-3.51(m, 1H), 2.59-2.49(m, 3H), 2.47-2.38(m, 2H), 2.29-2.22(m, 1H), 2.12-1.98( m, 5H), 1.96-1.90 (m, 1H), 1.87-1.77 (m, 3H), 1.30-1.20 (m, 5H).

实施例2Example 2

(1R,4R)-4-乙氧基-N-{2-[4'-(吡啶-2-基)螺[双环[3.1.0]己烷-3,2'-氧烷]-4'-基]乙基}-1,2,3,4-四氢萘-1-胺(化合物143)的合成 (1R,4R)-4-ethoxy-N-{2-[4'-(pyridin-2-yl)spiro[bicyclo[3.1.0]hexane-3,2'-oxane]-4' Synthesis of -ethyl}-1,2,3,4-tetralin-1-amine (compound 143)

步骤1:氮-甲基-氮-甲氧基-(R)-2-(4'-(吡啶-2-基)四氢氧杂螺[双环[3.1.0]己烷-3,2'-吡喃]-4'-基乙酰胺(143-1)的合成Step 1: Nitrogen-methyl-nitrogen-methoxy-(R)-2-(4'-(pyridin-2-yl)tetrahydroxaspiro[bicyclo[3.1.0]hexane-3,2' -Synthesis of -pyran]-4'-acetamide (143-1)

冰水浴下,将化合物141-5(590mg,1.86mmol)溶解于DCM(15mL)中,在氮气保护下,加入2mol/L的二甲基锌(3.7mL,7.46mmol)、二碘甲烷(5.0g,18.6mmol),加料完毕,移至室温搅拌反应16h。反应结束后,向反应液中加入乙酸乙酯(50mL),再用饱和NaHCO3溶液(20mL×2)洗涤,有机相用无水硫酸钠干燥,过滤,滤液旋干,柱层析(VPE:VEA=2:1)纯化得到黄色稠状物143-1(223mg,产率36.26%),[M+H]+:331.19。Under an ice-water bath, compound 141-5 (590 mg, 1.86 mmol) was dissolved in DCM (15 mL). Under nitrogen protection, 2 mol/L dimethylzinc (3.7 mL, 7.46 mmol) and diiodomethane (5.0 g, 18.6 mmol), after the addition is completed, move to room temperature and stir for 16 hours. After the reaction, ethyl acetate (50 mL) was added to the reaction solution, and then washed with saturated NaHCO 3 solution (20 mL × 2). The organic phase was dried with anhydrous sodium sulfate, filtered, and the filtrate was spin-dried and subjected to column chromatography (V PE : V EA = 2:1) and purified to obtain yellow thick substance 143-1 (223 mg, yield 36.26%), [M+H] + : 331.19.

步骤2:(R)-2-(4'-(吡啶-2-基)四氢氧杂螺[双环[3.1.0]己烷-3,2'-吡喃]-4'-基乙醛(143-2)的合成Step 2: (R)-2-(4'-(pyridin-2-yl)tetrahydroxaspiro[bicyclo[3.1.0]hexane-3,2'-pyran]-4'-ylacetaldehyde Synthesis of (143-2)

室温下,将化合物143-1(223mg,0.68mmol)溶解于四氢呋喃(THF)(10mL)中,在-40℃、氮气保护下,缓慢滴加红铝(335mg,70%,1.16mmol),加料完毕,搅拌反应4h。反应结束后,向反应液中加入质量分数为10%的柠檬酸水溶液(10mL),再移至室温,用1mol/L的HCl溶液调pH=2~3,用乙酸乙酯(30mL×1)萃取;水相再用质量分数为10%的NaOH溶液调pH=11~13,用二氯甲烷(50mL×3)萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液浓缩得到棕色油状物143-2(160mg,产率87.37%),[M+H]+:272.23。Compound 143-1 (223 mg, 0.68 mmol) was dissolved in tetrahydrofuran (THF) (10 mL) at room temperature, and red aluminum (335 mg, 70%, 1.16 mmol) was slowly added dropwise at -40°C under nitrogen protection. After completion, stir the reaction for 4 hours. After the reaction is completed, add a 10% citric acid aqueous solution (10 mL) to the reaction solution, then move to room temperature, adjust pH = 2 to 3 with 1 mol/L HCl solution, and use ethyl acetate (30 mL × 1) Extraction; the aqueous phase is then adjusted to pH=11~13 with 10% NaOH solution, extracted with dichloromethane (50mL×3), the organic phases are combined, dried over anhydrous sodium sulfate, filtered, and the filtrate is concentrated to obtain a brown oil. Product 143-2 (160 mg, yield 87.37%), [M+H] + : 272.23.

步骤3:(1R,4R)-4-乙氧基-N-{2-[4'-(吡啶-2-基)螺[双环[3.1.0]己烷-3,2'-氧烷]-4'-基]乙基}-1,2,3,4-四氢萘-1-胺(143)的合成Step 3: (1R,4R)-4-ethoxy-N-{2-[4'-(pyridin-2-yl)spiro[bicyclo[3.1.0]hexane-3,2'-oxane] Synthesis of -4'-yl]ethyl}-1,2,3,4-tetralin-1-amine (143)

室温下,将化合物143-2(50mg,0.18mmol),化合物141-7(35mg,0.18mmol)溶于DCM(3mL),再加入MgSO4(111mg,0.92mmol),在氮气保护下,室温搅拌12h,再加入NaBH4(21mg,0.55mmol),并搅拌1h,再加入甲醇(1mL),并搅拌0.5h。再用硅藻土过滤,滤液室温浓缩,大板(VPE:VEA=0:1)纯化得到黄色稠状物143(32mg,产率39.0%),[M+H]+:447.43。Dissolve compound 143-2 (50 mg, 0.18 mmol) and compound 141-7 (35 mg, 0.18 mmol) in DCM (3 mL) at room temperature, then add MgSO 4 (111 mg, 0.92 mmol), and stir at room temperature under nitrogen protection. 12h, then add NaBH 4 (21 mg, 0.55 mmol), and stir for 1 h, then add methanol (1 mL), and stir for 0.5 h. It was then filtered through diatomaceous earth, and the filtrate was concentrated at room temperature and purified using a large plate (V PE : V EA = 0:1) to obtain yellow viscous substance 143 (32 mg, yield 39.0%), [M+H] + : 447.43.

1H NMR(400MHz,CDCl3)δ8.56-8.49(m,1H),7.69-7.63(m,1H),7.39-7.34(m,1H),7.33-7.30(m,1H),7.26-7.18(m,3H),7.17-7.12(m,1H),4.41-4.36(m,1H),3.84-3.62(m,5H),3.59-3.48(m,2H),2.39-2.28(m,2H),2.26-2.21(m,1H),2.17-2.04(m,3H),1.89-1.80(m,3H),1.37-1.22(m,7H),1.19-1.10(m,2H),1.09-0.99(m,2H),0.59-0.54(m,1H),0.30-0.24(m,1H). 1 H NMR (400MHz, CDCl 3 ) δ 8.56-8.49 (m, 1H), 7.69-7.63 (m, 1H), 7.39-7.34 (m, 1H), 7.33-7.30 (m, 1H), 7.26-7.18 (m, 3H), 7.17-7.12 (m, 1H), 4.41-4.36 (m, 1H), 3.84-3.62 (m, 5H), 3.59-3.48 (m, 2H), 2.39-2.28 (m, 2H) , 2.26-2.21(m, 1H), 2.17-2.04(m, 3H), 1.89-1.80(m, 3H), 1.37-1.22(m, 7H), 1.19-1.10(m, 2H), 1.09-0.99( m, 2H), 0.59-0.54 (m, 1H), 0.30-0.24 (m, 1H).

实施例3Example 3

(1R,4R)-4-乙氧基-氮-{2-[9-(吡啶-2-基)-2,6-二氧杂螺[4.5]癸烷-9]乙基}-1,2,3,4-四氢萘-1-胺(化合物156)的合成 (1R,4R)-4-ethoxy-nitrogen-{2-[9-(pyridin-2-yl)-2,6-dioxaspiro[4.5]decane-9]ethyl}-1, Synthesis of 2,3,4-tetralin-1-amine (compound 156)

室温下,将化合物156-1(50mg,0.19mmol),化合物141-7(37.2mg,0.19mmol)溶于DCM(3mL),再加入MgSO4(116.9mg,0.97mmol),在氮气保护下,室温搅拌12h,再加入硼氢化钠(22.1mg,0.58mmol),并搅拌2h,再加入甲醇(1mL),并搅拌0.5h。再用硅藻土过滤,滤液室温浓缩,大板(VPE:VEA=0:1)纯化 得到黄色稠状物156(35mg,产率41.7%),[M+H]+:437.27。Dissolve compound 156-1 (50 mg, 0.19 mmol) and compound 141-7 (37.2 mg, 0.19 mmol) in DCM (3 mL) at room temperature, then add MgSO 4 (116.9 mg, 0.97 mmol), and under nitrogen protection, Stir at room temperature for 12 h, then add sodium borohydride (22.1 mg, 0.58 mmol), stir for 2 h, then add methanol (1 mL), and stir for 0.5 h. It was then filtered through diatomaceous earth, and the filtrate was concentrated at room temperature and purified by large plate (V PE : V EA = 0:1) to obtain yellow thick substance 156 (35 mg, yield 41.7%), [M+H] + : 437.27.

1H NMR(400MHz,CDCl3)δ8.60-8.56(m,1H),7.70-7.64(m,1H),7.36-7.32(m,2H),7.26-7.19(m,3H),7.18-7.13(m,1H),4.39-4.35(m,1H),3.95-3.88(m,1H),3.87-3.82(m,1H),3.71-3.64(m,2H),3.56-3.51(m,1H),2.59-2.49(m,3H),2.47-2.38(m,2H),2.29-2.22(m,1H),2.12-1.98(m,5H),1.96-1.90(m,1H),1.87-1.77(m,3H),1.45-1.39(m,1H)1.30-1.20(m,5H)。 1 H NMR (400MHz, CDCl 3 ) δ 8.60-8.56 (m, 1H), 7.70-7.64 (m, 1H), 7.36-7.32 (m, 2H), 7.26-7.19 (m, 3H), 7.18-7.13 (m, 1H), 4.39-4.35 (m, 1H), 3.95-3.88 (m, 1H), 3.87-3.82 (m, 1H), 3.71-3.64 (m, 2H), 3.56-3.51 (m, 1H) , 2.59-2.49(m, 3H), 2.47-2.38(m, 2H), 2.29-2.22(m, 1H), 2.12-1.98(m, 5H), 1.96-1.90(m, 1H), 1.87-1.77( m, 3H), 1.45-1.39 (m, 1H) 1.30-1.20 (m, 5H).

实施例4Example 4

氮-(2-(9-(吡啶-2-基)-6-氧杂螺环[4.5]癸烷-2-烯-9-基)乙基)-2,3-二氢-1氢-茚-1-胺(化合物128)的合成 Nitrogen-(2-(9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-2-en-9-yl)ethyl)-2,3-dihydro-1hydro- Synthesis of inden-1-amine (compound 128)

氮气保护下,将化合物A(100mg,0.39mmol),化合物9(76mg,0.58mmol),二氯乙烷(DCE,6mL)依次加入单口瓶中,室温下加入钛酸四异丙酯(TIPT,1.5mL),60℃搅拌16h后,加入硼氢化钠(44mg,1.2mmol),60℃搅拌反应2小时。反应完后,加入1.5mL水淬灭,加10mL二氯甲烷,过滤,室温浓缩过柱(V二氯甲烷:V甲醇=10:1),得黄色稠状化合物128(20mg,产率14%),[M+H]+:375.3。Under nitrogen protection, compound A (100 mg, 0.39 mmol), compound 9 (76 mg, 0.58 mmol), and dichloroethane (DCE, 6 mL) were added to a one-neck bottle in sequence, and tetraisopropyl titanate (TIPT, TIPT, 1.5 mL), stirred at 60°C for 16 hours, then added sodium borohydride (44 mg, 1.2 mmol), and stirred at 60°C for 2 hours. After the reaction is completed, add 1.5 mL of water to quench, add 10 mL of dichloromethane, filter, and concentrate at room temperature through the column (V dichloromethane : V methanol = 10:1) to obtain yellow viscous compound 128 (20 mg, yield 14%) ), [M+H] + :375.3.

1H NMR(400MHz,CDCl3)δ8.58(d,J=8.0Hz,1H),7.675(t,J=8.0Hz,1H),7.32(d,J=8.0Hz,1H),7.20-7.10(m,5H),5.61(brs,1H),5.45(brs,1H),4.08-4.04(m,1H),3.94-3.89(m,1H),3.83-3.80(m,1H),2.95-2.88(m,1H),2.77-2.69(m,1H),2.63-2.38(m,5H),2.26-2.14(m,2H),2.04-1.94(m,3H),1.81-1.76(m,2H),1.70-1.56(m,2H). 1 H NMR (400MHz, CDCl 3 ) δ8.58 (d, J=8.0Hz, 1H), 7.675 (t, J=8.0Hz, 1H), 7.32 (d, J=8.0Hz, 1H), 7.20-7.10 (m, 5H), 5.61 (brs, 1H), 5.45 (brs, 1H), 4.08-4.04 (m, 1H), 3.94-3.89 (m, 1H), 3.83-3.80 (m, 1H), 2.95-2.88 (m, 1H), 2.77-2.69 (m, 1H), 2.63-2.38 (m, 5H), 2.26-2.14 (m, 2H), 2.04-1.94 (m, 3H), 1.81-1.76 (m, 2H) ,1.70-1.56(m,2H).

实施例5Example 5

氮-(2-(4'-(吡啶-2-基)四氢氧杂螺[双环[3.1.0]己烷-3,2'-吡喃]-4'-基)乙基)-2,3-二氢-1氢-茚-1-胺(化合物1)的合成 Nitrogen-(2-(4'-(pyridin-2-yl)tetrahydroxaspiro[bicyclo[3.1.0]hexane-3,2'-pyran]-4'-yl)ethyl)-2 , Synthesis of 3-dihydro-1hydro-indene-1-amine (compound 1)

制备方法参考实施例4,得黄色稠状化合物1(产率7%),[M+H]+:389.3。For the preparation method, refer to Example 4, and obtain yellow viscous compound 1 (yield 7%), [M+H] + : 389.3.

1H NMR(400MHz,CDCl3)δ8.59-8.50(m,1H),7.67-7.62(m,1H),7.38-7.27(m,2H),7.20-7.19(m,2H),7.15-7.10(m,2H),4.25(brs,1H),3.80-3.68(m,2H),3.04-2.98(m,1H),2.81-2.79(m,1H),2.77-2.66(m,1H),2.32-2.10(m,5H),1.90-1.84(m,5H),1.66-1.62(m,1H),1.30-1.25(m,2H),1.11-1.01(m,3H),0.55-0.52(m,1H),0.24-0.22(m,1H)。 1 H NMR (400MHz, CDCl 3 ) δ8.59-8.50 (m, 1H), 7.67-7.62 (m, 1H), 7.38-7.27 (m, 2H), 7.20-7.19 (m, 2H), 7.15-7.10 (m, 2H), 4.25 (brs, 1H), 3.80-3.68 (m, 2H), 3.04-2.98 (m, 1H), 2.81-2.79 (m, 1H), 2.77-2.66 (m, 1H), 2.32 -2.10(m, 5H), 1.90-1.84(m, 5H), 1.66-1.62(m, 1H), 1.30-1.25(m, 2H), 1.11-1.01(m, 3H), 0.55-0.52(m, 1H), 0.24-0.22(m, 1H).

对比例1Comparative example 1

氮-((3-甲氧基噻吩-2-基)甲基)-2-(4'-(吡啶-2-基)四氢氧杂螺[双环[3.1.0]己烷-3,2'-吡喃]-4'-基)乙胺(化合物91)的合成 Nitrogen-((3-methoxythiophen-2-yl)methyl)-2-(4'-(pyridin-2-yl)tetrahydroxaspiro[bicyclo[3.1.0]hexane-3,2 Synthesis of '-pyran]-4'-yl)ethylamine (compound 91)

氮气保护下,将中间体B(100mg,0.37mmol),中间体1(63mg,0.44mmol),硫酸镁(882mg,7.4mmol),二氯甲烷(DCM,6mL)依次加入单口瓶中,室温搅拌16h,加入硼氢化钠(42mg,1.1mmol),搅拌10分钟,加入甲醇(MeOH,0.5mL),搅拌反应2小时。反应完后,加入二氯甲烷(10mL),过滤,室温浓缩,粗品柱层析纯化(V二氯甲烷:V甲醇=10:1),得到黄色稠状化合物91(12.43mg,产率8.5%),[M+H]+:399.3。Under nitrogen protection, add intermediate B (100 mg, 0.37 mmol), intermediate 1 (63 mg, 0.44 mmol), magnesium sulfate (882 mg, 7.4 mmol), and dichloromethane (DCM, 6 mL) into a single-neck bottle in sequence, and stir at room temperature. At 16h, add sodium borohydride (42 mg, 1.1 mmol), stir for 10 minutes, add methanol (MeOH, 0.5 mL), and stir for 2 hours. After the reaction was completed, dichloromethane (10 mL) was added, filtered, concentrated at room temperature, and the crude product was purified by column chromatography (V dichloromethane : V methanol = 10:1) to obtain yellow viscous compound 91 (12.43 mg, yield 8.5%) ), [M+H] + :399.3.

1H NMR(400MHz,氘代氯仿(CDCl3))δ8.53-8.52(m,1H),7.62(dt,J1=2.0Hz,J2=7.6Hz,1H),7.27(d,J=8.0Hz 1H),7.13-7.07(m,2H),6.77(d,J=5.2Hz 1H),3.81-3.66(m,7H),2.60-2.53(m,1H),2.32-2.28(m,1H),2.23-2.19(m,1H),2.05-1.96(m,2H),1.89-1.80(m,4H),1.69-1.60(m,2H),1.31-1.26(m,1H),1.10-1.09(m,1H),1.04-1.01(m,1H),0.55-0.52(m,1H),0.25-0.22(m,1H). 1 H NMR (400MHz, deuterated chloroform (CDCl 3 )) δ8.53-8.52 (m, 1H), 7.62 (dt, J1=2.0Hz, J2=7.6Hz, 1H), 7.27 (d, J=8.0Hz 1H), 7.13-7.07(m, 2H), 6.77(d, J=5.2Hz 1H), 3.81-3.66(m, 7H), 2.60-2.53(m, 1H), 2.32-2.28(m, 1H), 2.23-2.19(m, 1H), 2.05-1.96(m, 2H), 1.89-1.80(m, 4H), 1.69-1.60(m, 2H), 1.31-1.26(m, 1H), 1.10-1.09(m , 1H), 1.04-1.01(m, 1H), 0.55-0.52(m, 1H), 0.25-0.22(m, 1H).

对比例2Comparative example 2

氮-((3-甲氧基噻吩-2-基)甲基)-2-(9-(吡啶-2-基)-6-氧杂螺环[4.5]癸烷-2-烯-9-基)乙胺(化合物83)的合成 Nitrogen-((3-methoxythiophen-2-yl)methyl)-2-(9-(pyridin-2-yl)-6-oxaspiro[4.5]decane-2-ene-9- Synthesis of ethylamine (compound 83)

氮气保护下,将中间体A(80mg,0.31mmol),中间体1(53mg,0.37mmol),硫酸镁(744mg,6.2mmol),二氯甲烷(6mL)依次加入单口瓶中,室温搅拌16h,加入硼氢化钠(35mg,0.93mmol),搅拌10分钟,加入甲醇(0.5mL),搅拌反应2小时。反应完后,加入二氯甲烷(10mL),过滤,室温浓缩,粗品柱层析纯化(V二氯甲烷:V甲醇=10:1),得黄色稠状化合物83(35mg,产率29%),[M+H]+:385.3。Under nitrogen protection, add Intermediate A (80 mg, 0.31 mmol), Intermediate 1 (53 mg, 0.37 mmol), magnesium sulfate (744 mg, 6.2 mmol), and dichloromethane (6 mL) into a single-neck bottle in sequence, and stir at room temperature for 16 hours. Add sodium borohydride (35 mg, 0.93 mmol), stir for 10 minutes, add methanol (0.5 mL), and stir for 2 hours. After the reaction was completed, dichloromethane (10 mL) was added, filtered, concentrated at room temperature, and the crude product was purified by column chromatography (V dichloromethane : V methanol = 10:1) to obtain yellow thick compound 83 (35 mg, yield 29%) , [M+H] + :385.3.

1H NMR(400MHz,CDCl3)δ8.57-8.55(m,1H),7.62(dt,J1=2.0Hz,J2=8.0Hz,1H),7.29(d,J=8.0Hz 1H),7.13-7.09(m,1H),7.04(d,J=5.6,Hz 1H),6.76(d,J=5.6Hz 1H),5.61(brs,1H),5.44(brs,1H),3.89-3.86(m,1H),3.83-3.79(m,4H),3.74-3.66(m,2H),2.54-2.37(m,5H),2.14-1.91(m,4H),1.81-1.72(m,2H),1.62-1.56(m,1H). 1 H NMR (400MHz, CDCl 3 ) δ8.57-8.55 (m, 1H), 7.62 (dt, J1=2.0Hz, J2=8.0Hz, 1H), 7.29 (d, J=8.0Hz 1H), 7.13- 7.09 (m, 1H), 7.04 (d, J=5.6, Hz 1H), 6.76 (d, J=5.6Hz 1H), 5.61 (brs, 1H), 5.44 (brs, 1H), 3.89-3.86 (m, 1H), 3.83-3.79(m, 4H), 3.74-3.66(m, 2H), 2.54-2.37(m, 5H), 2.14-1.91(m, 4H), 1.81-1.72(m, 2H), 1.62- 1.56(m,1H).

对比例3Comparative example 3

氮-((3-甲氧基噻吩-2-基)甲基)-2-(9-(吡啶-2-基)-2,6-二氧杂螺[4.5]癸烷-9-基)乙胺(化合物31) 的合成 Nitrogen-((3-methoxythiophen-2-yl)methyl)-2-(9-(pyridin-2-yl)-2,6-dioxaspiro[4.5]decan-9-yl) Synthesis of ethylamine (compound 31)

室温下,将中间体C(50mg,0.162mmol)用二氯甲烷(3mL)溶解,然后依次加入硫酸钠(136mg,0.96mmol)和中间体1(41mg,0.288mmol)。反应在氮气保护下室温过夜。反应16h后,加入硼氢化钠继续搅拌30分钟。反应完全后,用15mL水进行淬灭,加入乙酸乙酯(15×2mL)进行萃取然后用饱和碳酸钠溶液(10mL×2)洗涤,有机相用无水硫酸钠干燥,过滤,粗品用柱层析纯化(V石油醚:V乙酸乙酯=1:1)得到淡黄色油状液体31(20mg,产率22%),[M+H]+:389.2。At room temperature, Intermediate C (50 mg, 0.162 mmol) was dissolved in dichloromethane (3 mL), and then sodium sulfate (136 mg, 0.96 mmol) and Intermediate 1 (41 mg, 0.288 mmol) were added sequentially. The reaction was carried out overnight at room temperature under nitrogen protection. After reacting for 16 hours, add sodium borohydride and continue stirring for 30 minutes. After the reaction is complete, quench with 15 mL of water, add ethyl acetate (15 × 2 mL) for extraction, and then wash with saturated sodium carbonate solution (10 mL × 2). The organic phase is dried over anhydrous sodium sulfate, filtered, and the crude product is subjected to column layer After analytical purification (V petroleum ether : V ethyl acetate = 1:1), light yellow oily liquid 31 (20 mg, yield 22%) was obtained, [M+H] + : 389.2.

1H NMR(400MHz,氯仿-d(Chloroform-d))δ8.59(d,J=8.5,3.0Hz,1H),7.65(d,J=7.7,6.0,4.1,1.9Hz,1H),7.32(d,J=8.1Hz,1H),7.17–7.12(m,1H),7.05(d,J=5.5Hz,1H),6.79(d,J=5.4Hz,1H),4.87(s,1H),3.88–3.82(m,3H),3.79(s,3H),3.75(d,J=3.3Hz,1H),3.69(d,J=2.0Hz,2H),3.55(d,J=9.3Hz,1H),3.51(s,2H),3.18(d,J=10.0Hz,1H),2.86(d,J=10.0Hz,1H),2.47(dd,J=11.0,5.9Hz,2H),2.14(dd,J=10.3,4.8Hz,1H),2.03(d,J=13.7Hz,1H),1.92(d,J=9.0Hz,1H),1.77(d,J=4.8Hz,1H),1.45–1.39(m,1H),1.20–1.12(m,1H)。 1 H NMR (400MHz, chloroform-d (Chloroform-d)) δ8.59 (d, J=8.5, 3.0Hz, 1H), 7.65 (d, J=7.7, 6.0, 4.1, 1.9Hz, 1H), 7.32 (d, J=8.1Hz, 1H), 7.17–7.12 (m, 1H), 7.05 (d, J=5.5Hz, 1H), 6.79 (d, J=5.4Hz, 1H), 4.87 (s, 1H) , 3.88–3.82(m, 3H), 3.79(s, 3H), 3.75(d, J=3.3Hz, 1H), 3.69(d, J=2.0Hz, 2H), 3.55(d, J=9.3Hz, 1H), 3.51 (s, 2H), 3.18 (d, J = 10.0Hz, 1H), 2.86 (d, J = 10.0Hz, 1H), 2.47 (dd, J = 11.0, 5.9Hz, 2H), 2.14 ( dd, J=10.3, 4.8Hz, 1H), 2.03 (d, J=13.7Hz, 1H), 1.92 (d, J=9.0Hz, 1H), 1.77 (d, J=4.8Hz, 1H), 1.45– 1.39 (m, 1H), 1.20–1.12 (m, 1H).

生物学评价biological evaluation

Op-Mu激动剂cAMP测试实验Op-Mu agonist cAMP test experiment

本发明的化合物可以激活μ-阿片受体(MOR)。激活的MOR可以调节细胞内cAMP的水平,cAMP进入细胞核与报告基因荧光素酶(Luciferase)的cAMP应答元件(cAMP response element,CRE)区结合,启动报告基因的表达。荧光素酶与其底物反应可发出荧光,通过测定荧光信号反映化合物的激动活性。Compounds of the invention can activate μ-opioid receptors (MOR). Activated MOR can regulate the level of cAMP in cells. cAMP enters the nucleus and binds to the cAMP response element (CRE) region of the reporter gene luciferase (Luciferase), initiating the expression of the reporter gene. Luciferase reacts with its substrate to emit fluorescence, and the agonistic activity of the compound is reflected by measuring the fluorescence signal.

实验方法experimental method

实施例化合物激动MOR影响下游cAMP水平变化的活性通过以下的方法进行测试。The activity of the compounds of the Examples in agonizing MOR and affecting changes in downstream cAMP levels was tested by the following method.

1.材料与试剂 1.Materials and reagents

2.实验操作步骤2. Experimental steps

检测缓冲液:1×stimulation buffer,500μM 1-甲基-3-异丁基黄嘌呤(IBMX),ddH2O。Detection buffer: 1×stimulation buffer, 500 μM 1-methyl-3-isobutylxanthine (IBMX), ddH 2 O.

化合物配制:化合物用二甲基亚砜(DMSO)溶解后配制为终浓度为10mM的母液,稀释成0.08mM的工作浓度,用Echo移液器对化合物进行4倍梯度稀释,初始浓度为0.08mM,10个浓度梯度,并分别加50nL到384细胞板中,双复孔,终浓度为0.4μM,然后将细胞板1000rpm离心1min。用Echo移液器转移50nL毛喉素(Forskolin)(终浓度是1μM)至384细胞板中。Compound preparation: Dissolve the compound in dimethyl sulfoxide (DMSO) and prepare a stock solution with a final concentration of 10mM. Dilute it to a working concentration of 0.08mM. Use an Echo pipette to perform a 4-fold gradient dilution of the compound. The initial concentration is 0.08mM. , 10 concentration gradients, and add 50nL to the 384 cell plate respectively, in double wells, with a final concentration of 0.4μM, and then centrifuge the cell plate at 1000rpm for 1 minute. Use an Echo pipette to transfer 50 nL of Forskolin (final concentration is 1 μM) into the 384 cell plate.

细胞铺板:将冻存的细胞融化,1000转离心5min,弃掉上清液,用HBSS缓冲液清洗两次细胞后,用检测缓冲液重悬细胞,将细胞密度调整到5.0×105个/mL,加入至384孔板中,每孔10μL,5000个细胞。震荡20s,1000rpm离心1min,将细胞板放入23℃孵箱中培养60min。Cell plating: Thaw the frozen cells, centrifuge at 1000 rpm for 5 min, discard the supernatant, wash the cells twice with HBSS buffer, resuspend the cells with detection buffer, and adjust the cell density to 5.0× 105 cells/ mL, add to 384-well plate, 10 μL per well, 5000 cells. Shake for 20 s, centrifuge at 1000 rpm for 1 min, and place the cell plate in a 23°C incubator for 60 min.

标准曲线的配制:用检测缓冲液对标准品腺苷-3',5'-环磷酸(cAMP)进行4倍梯度稀释,共8个浓度点,最高浓度是800nM,按照微孔板布局图每孔加入10μL。Preparation of the standard curve: Use the detection buffer to perform a 4-fold gradient dilution of the standard adenosine-3',5'-cyclic phosphate (cAMP), with a total of 8 concentration points. The highest concentration is 800nM. Follow the microplate layout diagram for each step. Add 10 μL to the well.

检测试剂配制:用裂解缓冲液将Anti cAMP-Cryptate和AMP-d2稀释至1×,按照微孔板布局图,每孔加入10μL检测试剂,震荡20s,1000rpm离心1min,将细胞板放入23℃孵箱中培养60min;在Envision酶标仪上读板。Preparation of detection reagents: Dilute Anti cAMP-Cryptate and AMP-d2 to 1× with lysis buffer, add 10 μL detection reagent to each well according to the microplate layout, shake for 20 s, centrifuge at 1000 rpm for 1 min, and place the cell plate at 23°C. Incubate in the incubator for 60 minutes; read the plate on an Envision microplate reader.

3.结果分析3. Result analysis

运用Microsoft Excel软件计算活性百分比,对于激动剂使用公式%Effect=100×(Sample Raw Value-Low Control Average)/(High Control Average-Low Control Average),运用GraphPad Prism 5数据分析软件,对于激动剂选用Dose-response-Stimulation—log[agonist]vs.response--Variable slope模式进行拟合分析,得出各检测样品的EC50值。Use Microsoft Excel software to calculate the activity percentage. For agonists, use the formula %Effect=100×(Sample Raw Value-Low Control Average)/(High Control Average-Low Control Average). Use GraphPad Prism 5 data analysis software. For agonists, use Dose-response-Stimulation—log[agonist]vs.response—Variable slope mode was used for fitting analysis to obtain the EC 50 value of each test sample.

本发明的化合物激动MOR影响下游cAMP水平的变化通过以上的试验进行测定,实验结果表明此系列化合物表现出较强的Op-Mu激动效应,其中测得的具有典型代表的化合物的EC50值见表一。其中,设置6组对照,分别为化合物83、化合物91、化合物31、TRV130、TRV130(消旋)和SHR8554,其中TRV130、TRV130(消旋)和SHR8554分别具有以下结构式,TRV130制备方法参考专利CN103702561A;SHR8554制备方法参考专利CN107001347B;Emax为化合物引起cAMP水平变化的最大效能。The changes in downstream cAMP levels caused by the compounds of the present invention stimulating MOR were measured through the above experiments. The experimental results show that this series of compounds exhibit strong Op-Mu agonistic effects. The measured EC 50 values of typical representative compounds are shown in Table I. Among them, 6 groups of controls were set up, namely compound 83, compound 91, compound 31, TRV130, TRV130 (racemic) and SHR8554, among which TRV130, TRV130 (racemic) and SHR8554 respectively have the following structural formulas. The preparation method of TRV130 refers to patent CN103702561A; The preparation method of SHR8554 refers to patent CN107001347B; E max is the maximum potency of the compound to cause changes in cAMP levels.

表一:受试化合物对MOR受体影响cAMP水平的EC50和Emax Table 1: EC 50 and E max of test compounds affecting cAMP levels at MOR receptors

本发明实施例的优选化合物对Mu阿片受体具有明显的激动作用,部分化合物EC50值和Emax远优于对照组。The preferred compounds in the embodiments of the present invention have obvious agonistic effects on Mu opioid receptors, and the EC 50 values and E max of some compounds are much better than those of the control group.

Op-Kappa激动剂cAMP测试实验Op-Kappa agonist cAMP test experiment

ForsKolin(毛喉素)能够刺激人K阿片受体高表达细胞株-OPRKI细胞(DiscoveRx)cAMP的释放,而K阿片受体激动剂能够抑制forsKolin刺激的cAMP释放。通过检测受试化合物对forsKolin刺激的cAMP释放的抑制作用,能够测定化合物对人K片受体的激动活性。首先用一定浓度的forsKolin和不同浓度的受试化合物与人高表达K阿片受体细胞株一起孵育。使用基于时间分辨荧光共振能量转移(TR-FRET)的cAMP免疫测试法(LANCEPerKinElmer)来确定所激发的OPRK1细胞中的cAMP水平。具体方法如下:ForsKolin (forskolin) can stimulate the release of cAMP from human K opioid receptor high-expressing cell line-OPRKI cells (DiscoveRx), while K opioid receptor agonists can inhibit the cAMP release stimulated by forsKolin. By detecting the inhibitory effect of the test compound on forsKolin-stimulated cAMP release, the agonistic activity of the compound on the human K-sheet receptor can be determined. First, a certain concentration of forsKolin and different concentrations of test compounds are used to incubate with human high-expression K opioid receptor cell lines. A time-resolved fluorescence resonance energy transfer (TR-FRET)-based cAMP immunoassay (LANCEPerKinElmer) was used to determine cAMP levels in stimulated OPRK1 cells. The specific method is as follows:

检测缓冲液:1×stimulation buffer,500μM IBMX,ddH2O。化合物配制:化合物用DMSO溶解后配制为终浓度为10mM的母液,稀释成2mM的工作浓度,用Echo对化合物进行4倍梯度稀释,初始浓度为2mM,10个浓度梯度,并分别加50nL到384细胞板中,双复孔,终浓度为10μM,然后将细胞板1000rpm离心1min;用Echo转移50nL ForsKolin(终浓度是3μM)至384细胞板。Assay buffer: 1×stimulation buffer, 500μM IBMX, ddH 2 O. Compound preparation: Dissolve the compound in DMSO and prepare a stock solution with a final concentration of 10mM, dilute it to a working concentration of 2mM, use Echo to perform a 4-fold gradient dilution of the compound, with an initial concentration of 2mM, 10 concentration gradients, and add 50nL to 384. In the cell plate, double wells were prepared with a final concentration of 10 μM, and then the cell plate was centrifuged at 1000 rpm for 1 min; use Echo to transfer 50 nL ForsKolin (final concentration is 3 μM) to the 384 cell plate.

细胞铺板:将冻存的细胞融化,1000转离心5min,弃掉上清液,用HBSS缓冲液清洗两次细胞后,用检测缓冲液重悬细胞,将细胞密度调整到3.0×105个/mL,加入至384孔板中,每孔10μL,3000个细胞。震荡20s,1000rpm离心1min,将细胞板放入23℃孵箱中培养60min。标准曲线的配制:用检测缓冲液对标准品cAMP进行4倍梯度稀释,共8个浓度点,最高浓度是800nM,按照微孔板布局图每孔加入10μL。检测试剂配制:用裂解缓冲液将中间体Anti cAMP-Cryptate和AMP-d2稀释至1×,每孔加入10uL检测试剂,震荡20s,1000rpm离心1min,将细胞板放入23℃孵箱中培养60min。最后在Envision上读板。Cell plating: Thaw the frozen cells, centrifuge at 1000 rpm for 5 minutes, discard the supernatant, wash the cells twice with HBSS buffer, resuspend the cells with detection buffer, and adjust the cell density to 3.0× 105 cells/ mL, add to 384-well plate, 10 μL per well, 3000 cells. Shake for 20 s, centrifuge at 1000 rpm for 1 min, and place the cell plate in a 23°C incubator for 60 min. Preparation of the standard curve: Use the detection buffer to perform a 4-fold gradient dilution of the standard cAMP, with a total of 8 concentration points. The highest concentration is 800nM. Add 10 μL to each well according to the microwell plate layout. Preparation of detection reagent: Dilute the intermediates Anti cAMP-Cryptate and AMP-d2 to 1× with lysis buffer, add 10uL detection reagent to each well, shake for 20s, centrifuge at 1000rpm for 1min, and place the cell plate in a 23°C incubator for 60min. . Finally read the plate on Envision.

运用Microsoft Excel软件计算活性百分比,对于激动剂使用公式%Effect=100×(Sample Raw Value-Low Control Average)/(High Control Average-Low Control Average)。运用GraphPad Prism 5数据分析软件,对于激动剂选用Dose-response-Stimulation—log[agonist]vs.response--Variable slope模式进行拟合分析,得出各检测样品的EC50值。实验数据见表二。Use Microsoft Excel software to calculate the activity percentage, and for agonists use the formula %Effect=100×(Sample Raw Value-Low Control Average)/(High Control Average-Low Control Average). Using GraphPad Prism 5 data analysis software, the Dose-response-Stimulation—log[agonist]vs.response—Variable slope mode was used for fitting analysis for agonists, and the EC 50 value of each test sample was obtained. The experimental data are shown in Table 2.

表二:受试化合物对K阿片受体影响 Table 2: Effect of test compounds on K opioid receptors

本发明实施例化合物对激动K阿片受体的活性相比对照组明显较弱;表现出本发明化合物对MOR受体有高的选择性,推测本发明实施例化合物具有更低的副作用。The activity of the compounds of the present invention in stimulating K opioid receptors is significantly weaker than that of the control group; it shows that the compounds of the present invention have high selectivity for MOR receptors, and it is speculated that the compounds of the present invention have lower side effects.

Mμ阿片受体的β-arrestin信号通路的活性测试实验 Activity test experiment of β-arrestin signaling pathway of Mμ opioid receptor

本研究旨在通过CHO-K1/Arrestin/hMOR的EC50和EMAX测定来评估靶向μ-阿片受体MOR的激动剂的β-Arrestin招募效率。CHO-K1/Arrestin/hMOR细胞株表达与β-半乳糖苷酶供体片段融合的hMOR和与β-半乳糖苷酶受体片段融合的β-Arrestin。当β-arrestin与hMOR相互作用时,这些片段形成活性的β-半乳糖苷酶。准备384孔板,将60nL/孔系列稀释的化合物滴入384孔板,将20μLCHO-K1/Arrestin/hMOR细胞浮液注入测定板,细胞密度为7.5k细胞/孔。37℃,0.5%CO2(体积分数,其余为空气)孵育分析板120分钟,用蜻蜓法将10μL/孔检测试剂加入分析板中,培养皿在室温孵育60分钟,通过Envision检测化学发光信号,使用XLfit进行数据分析。实验数据见表三。This study aimed to evaluate the β-Arrestin recruitment efficiency of agonists targeting the μ-opioid receptor MOR through EC 50 and EMAX assays of CHO-K1/Arrestin/hMOR. The CHO-K1/Arrestin/hMOR cell line expresses hMOR fused to the β-galactosidase donor fragment and β-Arrestin fused to the β-galactosidase receptor fragment. When β-arrestin interacts with hMOR, these fragments form active β-galactosidase. Prepare a 384-well plate, drop 60 nL/well of serially diluted compounds into the 384-well plate, and inject 20 μL of CHO-K1/Arrestin/hMOR cell suspension into the assay plate. The cell density is 7.5k cells/well. Incubate the analysis plate at 37°C with 0.5% CO 2 (volume fraction, the rest is air) for 120 minutes. Add 10 μL/well detection reagent to the analysis plate using the dragonfly method. The culture dish is incubated at room temperature for 60 minutes. The chemiluminescence signal is detected by Envision. Use XLfit for data analysis. The experimental data are shown in Table 3.

表三:受试化合物对β-arrestin信号通路的影响 Table 3: Effect of test compounds on β-arrestin signaling pathway

本发明实施例化合物对β-arrestin信号通路几乎无激活作用,以及本发明化合物和对照组相比有更好的偏向性(cAMP与β-arrestin信号通路),推测本发明化合物相比对照组具有更低的副作用。The compounds of the present invention have almost no activating effect on the β-arrestin signaling pathway, and the compounds of the present invention have better bias (cAMP and β-arrestin signaling pathways) compared with the control group. It is speculated that the compounds of the present invention have better effects than the control group. Lower side effects.

测试本发明化合物对hERG钾电流的阻断作用Test the blocking effect of the compound of the present invention on hERG potassium current

试验系统Test system

细胞:中国仓鼠卵巢(CHO)细胞系,CHO-hERG细胞用于本试验。Cells: Chinese hamster ovary (CHO) cell line, CHO-hERG cells were used in this experiment.

细胞培养液及培养条件:完全培养基为F12培养基,补充加入10%胎牛血清,1%选择性抗生素(G418),89μg/mL潮霉素B(HB)。复苏培养基为F12培养基补充加入10vol%胎牛血清。CHO-hERG细胞生长在37℃(±2℃)、5%CO2(4%至8%)的高湿度培养箱中。细胞用复苏培养基复苏,完全培养基传代,用于膜片钳试验的细胞在最后一次传代时换成复苏培养基。Cell culture medium and culture conditions: The complete medium is F12 medium, supplemented with 10% fetal bovine serum, 1% Selective antibiotic (G418), 89μg/mL hygromycin B (HB). The recovery medium is F12 medium supplemented with 10vol% fetal bovine serum. CHO-hERG cells were grown in a high-humidity incubator at 37°C (±2°C), 5% CO 2 (4% to 8%). Cells were revived with recovery medium and passaged in complete medium. Cells used for patch-clamp experiments were replaced with recovery medium at the last passage.

细胞外液及内液成分: Extracellular and intracellular fluid components:

试验方法experiment method

(1)将处于指数生长期的CHO-hERG细胞收集并重悬在ECS中备用。(1) Collect CHO-hERG cells in the exponential growth phase and resuspend them in ECS for later use.

(2)手动膜片钳试验(2) Manual patch clamp test

全细胞膜片钳技术下记录hERG电流,记录温度为室温。膜片钳放大器输出信号通过数模转换以及2.9KHz低通滤波。数据记录用Patchmaster Pro软件采集。hERG currents were recorded under whole-cell patch-clamp technique, and the recording temperature was room temperature. The output signal of the patch clamp amplifier is digital-to-analog conversion and 2.9KHz low-pass filtered. Data recording was collected using Patchmaster Pro software.

细胞种在细胞记录槽中放置在倒置显微镜载物台上,随机选择记录槽中的一个细胞进行试验。灌流系统固定在倒置显微镜载物台上用ECS持续灌流细胞。Cells were seeded in a cell recording tank and placed on the stage of an inverted microscope, and a cell in the recording tank was randomly selected for experiment. The perfusion system was fixed on the stage of an inverted microscope and continuously perfused with ECS.

用毛细玻璃管制备手动膜片钳试验记录微电极,其中充灌细胞內液。在膜片钳试验当天,使用硼硅酸盐 玻璃管(BF150-117-10,SUTTER INSTRUMENT USA)制备电极。电极充灌ICS后电阻在2-5MΩ之间。Manual patch-clamp recording microelectrodes were prepared using capillary glass tubes filled with intracellular fluid. On the day of the patch clamp test, use borosilicate Glass tubes (BF150-117-10, SUTTER INSTRUMENT USA) were used to prepare electrodes. After the electrode is filled with ICS, the resistance is between 2-5MΩ.

钳制电压为-80mV,第一步去极化至+60mV并维持850ms开放hERG通道。然后,电压设置为-50mV并维持1275ms,产生反弹电流或者称为尾电流,尾电流的峰值将被测量并用于分析。最后,电压恢复到钳制电压(-80mV)。试验过程中,这个指令电压程序每间隔15s重复一次。The clamping voltage was -80mV, and the first step depolarized to +60mV and maintained open hERG channels for 850ms. Then, the voltage is set to -50mV and maintained for 1275ms, resulting in a rebound current or tail current, and the peak value of the tail current is measured and used for analysis. Finally, the voltage returns to the clamping voltage (-80mV). During the test, this command voltage program is repeated every 15 seconds.

在溶媒对照工作溶液灌流的记录开始阶段,监测尾电流峰值直至稳定3条以上扫描曲线后则可以灌流待测试的供试品/阳性对照工作溶液,直到供试品/阳性对照工作溶液对hERG电流峰值的抑制作用达到稳定状态。一般以最近的连续3个电流曲线峰值基本重合作为判断是否稳定状态的标准。达到稳定态势以后继续灌流下一浓度供试品。一个细胞上可以测试一个或多个供试品/阳性对照,或者同一种药物的多个浓度,不同供试品/阳性对照之间需用溶媒对照工作液冲洗直到hERG电流回复到加药物之前80%以上的大小。同一浓度下各记录细胞抑制率的标准差不超过15%。At the beginning of recording the perfusion of the vehicle control working solution, monitor the tail current peak until more than 3 scanning curves stabilize, then the test article/positive control working solution to be tested can be perfused until the test article/positive control working solution changes the hERG current The inhibitory effect of the peak reaches a steady state. Generally, the basic overlap of the three most recent consecutive current curve peaks is used as the criterion for judging whether the state is stable. After reaching a stable state, continue to perfuse the next concentration of test sample. One or more test articles/positive controls, or multiple concentrations of the same drug, can be tested on one cell. Different test articles/positive controls need to be rinsed with vehicle control working solution until the hERG current returns to 80% before adding the drug. % or more in size. The standard deviation of the inhibition rate of each recorded cell at the same concentration does not exceed 15%.

阳性对照西沙必利的测试浓度为0.1μM,重复测定两个细胞。根据科学文献报道,0.1μM的西沙必利抑制hERG电流超过50%。(Milnes,J.T.,et al.)。The test concentration of positive control cisapride was 0.1 μM, and the assay was repeated for two cells. According to reports in the scientific literature, cisapride at 0.1 μM inhibits hERG currents by more than 50%. (Milnes, J.T., et al.).

(3)手动膜片钳数据接受标准(3) Manual patch clamp data acceptance criteria

封接标准:全细胞模式形成后,施加钳制电压(-80mV),可以记录到细胞膜相关参数(Cm,Rm以及Ra)。一个好的的全细胞记录应该满足以下条件:路径电阻(Rs)小于10MΩ;膜电阻(Rm)大于500MΩ和膜电容(Cm)小于100pF。Sealing standard: After the whole cell mode is formed, a clamping voltage (-80mV) is applied, and the cell membrane related parameters (Cm, Rm and Ra) can be recorded. A good whole-cell recording should meet the following conditions: path resistance (Rs) less than 10MΩ; membrane resistance (Rm) greater than 500MΩ and membrane capacitance (Cm) less than 100pF.

电流大小:供试品/阳性对照品作用前峰电流幅度在400pA和5000pA之间。否则,放弃该细胞。Current size: The peak current amplitude before the test article/positive control substance acts is between 400pA and 5000pA. Otherwise, discard the cell.

漏电流:在-80mV的钳制电压下,漏电流绝对值应该小于200pA。电流幅度将会用-80mV下的漏电流校正。漏电流绝对值大于200pA的扫描曲线不能用于分析。Leakage current: Under the clamping voltage of -80mV, the absolute value of the leakage current should be less than 200pA. The current amplitude will be corrected using the leakage current at -80mV. Scanning curves with an absolute value of leakage current greater than 200pA cannot be used for analysis.

数据分析data analysis

对于每个细胞,每一个浓度的供试品及阳性对照的抑制百分比由记录到的电流反应用以下公式算出:(1–供试品/阳性对照灌流后记录到的尾峰值电流/溶媒对照灌流记录到的尾峰值电流(起始电流))×100%。For each cell, the percent inhibition of each concentration of test article and positive control was calculated from the recorded current response using the following formula: (1 – Tail peak current recorded after test article/positive control perfusion/vehicle control perfusion Recorded tail peak current (initial current)) × 100%.

对于每一个浓度记录到所有的细胞抑制百分比取均值,IC50值(半数抑制浓度)由Hill拟合的方法由浓度效应曲线中得出。The percent inhibition of all cells recorded for each concentration was averaged, and the IC 50 value (half inhibitory concentration) was derived from the concentration effect curve by Hill fitting.

试验结果test results

本发明部分化合物对hERG电流的抑制结果,具体见下表四;The specific inhibition results of some compounds of the present invention on hERG current are shown in Table 4 below;

表四:受试化合物对hERG电流的抑制结果 Table 4: Inhibition results of hERG current by test compounds

注:20μM>IC50>10μM为++,10μM>IC50>1μM为+。Note: 20μM>IC 50 >10μM is ++, 10μM>IC 50 >1μM is +.

本发明实施例化合物相比对照组具有较高的hERG IC50值,具有显著差异,表现出对hERG抑制作用更弱,说明本发明化合物的心脏毒性风险较低。 The compounds of the present invention have a higher hERG IC 50 value than the control group, with a significant difference, and show a weaker inhibitory effect on hERG, indicating that the risk of cardiotoxicity of the compounds of the present invention is low.

药代动力学实验Pharmacokinetic experiments

被研究化合物单次口服或者静脉给药(溶媒5vol%DMSO+10vol%Solutol(HS-15)+85vol%saline)于动物(例如小鼠、大鼠、犬或者猴子),在固定的时间点取血。血样采集后,立即温和地颠倒试管至少5次,保证混合充分后放置于冰上。血液用肝素抗凝,然后8000rpm离心5分钟,将血清与红细胞分离。用移液器吸岀血清转移至2mL的聚丙烯管,标明化合物的名称和时间点,在进行LC-MS分析前保存在-40℃冰箱,待测。髙浓度样品用空白血浆稀释测定时。样品处理后,用LCMS/MS对血浆中的物质进行定量分析。通过进行了验证的药动学计算机程序,用以这种方式获得的血浆浓度/时曲线来计算药动学参数。实验发现本发明化合物均具有较好的药代动力学性质。The compound to be studied is administered orally or intravenously in a single dose (vehicle 5vol% DMSO + 10vol% Solutol (HS-15) + 85vol% saline) to animals (such as mice, rats, dogs or monkeys), and is taken at fixed time points. Blood. Immediately after blood sample collection, gently invert the test tube at least 5 times to ensure thorough mixing and place on ice. The blood was anticoagulated with heparin and then centrifuged at 8000 rpm for 5 minutes to separate serum from red blood cells. Use a pipette to aspirate the serum and transfer it to a 2 mL polypropylene tube, mark the name of the compound and time point, and store it in a -40°C refrigerator until LC-MS analysis. When high concentration samples are diluted with blank plasma for measurement. After sample processing, the substances in the plasma were quantitatively analyzed using LCMS/MS. The plasma concentration/time curve obtained in this way was used to calculate pharmacokinetic parameters using a validated pharmacokinetic computer program. Experiments have found that the compounds of the present invention all have good pharmacokinetic properties.

SD雄性大鼠以表五组别剂量静脉给药后(各组为等摩尔剂量给药,溶媒为5vol%DMSO+10vol%Solutol(HS-15)+85vol%saline,每组3只),在固定的时间点取血检测。本发明的部分化合物的在大鼠血浆中的原型化合物药代动力学参数如下表五;表五中,IV为静脉给药,AUC为血浆浓度-时间曲线下面积,Cmax为最大血药浓度,T1/2为消除半衰期,Variable为变量,Mean为平均数,SD为标准差。After intravenous administration of doses in the five groups in Table 5 to SD male rats (each group was administered an equimolar dose, the vehicle was 5vol% DMSO + 10vol% Solutol (HS-15) + 85vol% saline, 3 rats in each group). Blood tests were taken at fixed time points. The pharmacokinetic parameters of prototype compounds of some compounds of the present invention in rat plasma are as follows in Table 5; in Table 5, IV refers to intravenous administration, AUC is the area under the plasma concentration-time curve, and C max is the maximum plasma concentration. , T 1/2 is the elimination half-life, Variable is the variable, Mean is the mean, and SD is the standard deviation.

表五:受试化合物药代动力学参数 Table 5: Pharmacokinetic parameters of test compounds

本发明实施例化合物在大鼠体内展现出良好的药代动力学性质;与对照组相比,本发明化合物在血浆中游离碱的AUC(h*ng/mL)均有显著提高。The compounds of the present invention exhibit good pharmacokinetic properties in rats; compared with the control group, the AUC (h*ng/mL) of the free base of the compounds of the present invention in plasma is significantly increased.

大鼠热板法镇痛药效实验Experiment on analgesic efficacy of hot plate method in rats

雌性SD大鼠,将大鼠开始训练日期记为D0。于D0,将测痛仪温度设定为52℃(52.0±0.5℃),大鼠置于热板同时计时,记录大鼠出现舔后足或跳跃时痛阈值所用的时间(s)。若大鼠于热板上超过30s没有出现热痛反应,立即取出大鼠,痛阈值记为30s。淘汰反应敏感和反应迟钝大鼠。于D1,取预筛后的大鼠,将测痛仪温度设定为52℃(52.0±0.5℃),大鼠置于热板并计时,记录大鼠出现舔后足或跳跃时痛阈值所用的时间(s),共测三次。以三次平均值作为大鼠baseline痛阈值。于D1,大鼠根据baseline痛阈值随机分组,每组8只动物。于D2,按照表中各剂量组,通过尾静脉注射方式给予溶媒或化合物(各组等摩尔剂量给药, 溶媒为5vol%DMSO+10vol%Solutol(HS-15)+85vol%saline)。于D2,各组大鼠给药后0.5min、0.5h、1h、3h,将测痛仪温度设定为52℃(52.0±0.5℃),大鼠置于热板并计时,记录大鼠出现舔后足或跳跃时痛阈值所用的时间(s),每个时间点测1次。若大鼠于热板上超过30s没有出现热痛反应,立即取出大鼠,痛阈值记为30s。计算各组各个时间点痛阈%MPE,评价各受试样品体内镇痛药效。实验数据以Mean±SEM表示,各组间数据采用GraphPad Prism进行方差分析(ANOVA)检验(Two Way ANOVA或One-Way ANOVA),P<0.05认为是有显著性差异。最大镇痛效应百分率(即痛阈%MPE)=(给药后痛阈值-基础痛阈值)/(30-基础痛阈值)×100%,实验结果下表六;表六中,Variable为变量,Average为平均数,SEM为标准误差。For female SD rats, the date the rat started training was recorded as D0. At D0, set the temperature of the pain meter to 52°C (52.0±0.5°C), place the rat on the hot plate and time it at the same time, and record the time (s) it takes for the rat to reach the pain threshold when licking its hind paws or jumping. If the rat does not show heat pain response on the hot plate for more than 30 s, the rat is taken out immediately, and the pain threshold is recorded as 30 s. Eliminate sensitive and unresponsive rats. On D1, take pre-screened rats, set the temperature of the pain meter to 52°C (52.0±0.5°C), place the rats on the hot plate and time, and record the pain threshold when the rats lick their hind paws or jump. time (s), measured three times in total. The average value of three times was used as the baseline pain threshold of rats. On D1, rats were randomly divided into groups according to the baseline pain threshold, with 8 animals in each group. On D2, the vehicle or compound was administered via tail vein injection according to each dose group in the table (equal molar doses were administered in each group, The solvent is 5vol% DMSO + 10vol% Solutol (HS-15) + 85vol% saline). On D2, 0.5min, 0.5h, 1h, and 3h after the administration of rats in each group, the temperature of the pain meter was set to 52°C (52.0±0.5°C). The rats were placed on the hot plate and timed, and the appearance of the rats was recorded. The time (s) it takes to reach the pain threshold when licking the hind paw or jumping, measured once at each time point. If the rat does not show heat pain response on the hot plate for more than 30 s, the rat is taken out immediately, and the pain threshold is recorded as 30 s. Calculate the %MPE of the pain threshold at each time point in each group, and evaluate the analgesic efficacy of each test sample in vivo. The experimental data are expressed as Mean ± SEM, and the data between each group were subjected to analysis of variance (ANOVA) test (Two Way ANOVA or One-Way ANOVA) using GraphPad Prism. P<0.05 was considered to be significantly different. Maximum analgesic effect percentage (ie, pain threshold %MPE) = (pain threshold after administration - basic pain threshold) / (30 - basic pain threshold) × 100%. The experimental results are in Table 6 below; in Table 6, Variable is a variable, Average is the mean and SEM is the standard error.

表六:受试化合物体内镇痛药效 Table 6: In vivo analgesic efficacy of test compounds

注:与对照组(溶媒,IV)相比,***p<0.001Note: ***p<0.001 compared with the control group (vehicle, IV)

本发明实施例的化合物相比对照组展现出更好的镇痛效果,在大鼠热板法镇痛药效研究中表现出痛阈更高,持续时间更长久的镇痛作用。The compounds of the embodiments of the present invention exhibit better analgesic effects than the control group. In the hot plate method analgesic efficacy study in rats, the compounds showed higher pain thresholds and longer lasting analgesic effects.

急性毒性实验acute toxicity test

化合物单次静脉给药(溶媒5vol%DMSO+10vol%Solutol(HS-15)+85vol%saline)于SD大鼠(每个化合物设置4-6剂量组,每剂量组10只,雌性各半),给药后后进行临床观察。临床观察第一天两次,第二天开始一天一次,连续14天。包括行为学观察、自主活动和神经系统行为和死亡情况等,得到化合物的最大耐受 量(MTD值)和半数致死量(LD50值)。实验结果显示本发明化合物在大鼠单次静脉给药的MTD值和LD50值相比对照组有显著提高,表明本发明化合物具有良好的安全性。A single intravenous administration of the compound (vehicle 5vol% DMSO + 10vol% Solutol (HS-15) + 85vol% saline) was administered to SD rats (4-6 dose groups were set for each compound, 10 rats per dose group, half female) , conduct clinical observation after administration. Clinical observation was conducted twice on the first day and once a day starting from the second day for 14 consecutive days. Including behavioral observations, autonomous activities, nervous system behavior and death conditions, etc., the maximum tolerated dose (MTD value) and the median lethal dose (LD 50 value) of the compound are obtained. The experimental results show that the MTD value and LD 50 value of the compound of the present invention after a single intravenous administration in rats are significantly improved compared with the control group, indicating that the compound of the present invention has good safety.

以上所述仅为本发明的较佳实施例,并不用以限制本发明,凡在本发明的精神和原则之内,所做的任何修改、等同替换、改进等,均应包含在本发明保护的范围之内。 The above are only preferred embodiments of the present invention and are not intended to limit the present invention. Any modifications, equivalent substitutions, improvements, etc. made within the spirit and principles of the present invention shall be included in the protection of the present invention. within the range.

Claims (10)

  1. A compound represented by the formula (IV) or (V) or (VI), a solvate, stereoisomer, deuterated compound, or a pharmaceutically acceptable salt thereof,
    wherein ring B, ring C are each independently selected from substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl;
    ring D is selected from cycloalkyl, heterocycloalkyl;
    R 4 、R 5 each independently selected from H, deuterium atom, alkyl, oxo, alkoxy, hydroxy, halogen, cyano, alkynyl, alkenyl, - (CH) 2 ) g -O-3 to 12 membered heterocyclyl, - (CH) 2 ) g -O-3 to 12 membered cycloalkyl, - (CH) 2 ) g -3 to 12 membered cycloalkyl, - (CH) 2 ) g -3 to 12 membered heterocyclyl, 5 to 10 membered heteroaryl, 5 to 10 membered aryl, -S (=o) f -C 1-6 Alkyl, -O-C 2-6 Alkynyl, -O-C 2-6 Alkenyl groups; wherein the heterocyclyl, heteroaryl, aryl, alkyl, alkynyl, alkenyl, alkoxy may optionally be further substituted with 1 to 3R 6 Substituted;
    the R is 6 Each independently selected from deuterium atoms, halogen, -OH, -C 1-6 Alkyl, -C 1-6 alkyl-O-C 1-6 Alkyl, -O-C 1-6 Alkyl, 3-to 6-membered cycloalkyl, -O-C 2-6 Alkynyl, -O-C 2-6 Alkenyl, -C 2-6 Alkynyl, -C 2-6 Alkenyl, amino, carboxylate, nitro, cyano, hydroxyalkyl, heterocyclyl, aryl, heteroaryl;
    the g is selected from 0, 1, 2, 3, 4, 5 and 6;
    Said f is selected from 0, 1, 2;
    p, q, L are each independently 0, 1, 2, 3 or 4;
    the heteroaryl, the heterocycloalkyl, or the heteroatoms on the heterocyclyl are each independently selected from O, S or N.
  2. The compound, solvate, stereoisomer, deuterated compound, or pharmaceutically acceptable salt thereof according to claim 1, wherein the compound is selected from the group consisting of formula (vii) or (viii) or (ix) or (x) or (xi) or (xii):
  3. the compound, solvate, stereoisomer, deuterated compound, or pharmaceutically acceptable salt thereof according to claim 1 wherein the compound is selected from the group consisting of the following structures:
  4. the compound, solvate, stereoisomer, deuterated compound, or pharmaceutically acceptable salt thereof according to claim 1 wherein the compound is selected from the group consisting of the following structures:
  5. the compound, solvate, stereoisomer, deuterated compound, or pharmaceutically acceptable salt thereof according to claim 1 wherein the compound is selected from the group consisting of the following structures:
    wherein R is 4 Independently selected from deuterium atom, alkyl group, oxo group, alkoxy group, hydroxy group, halogen, cyano group, alkynyl group, alkenyl group, - (CH) 2 ) g -O-3 to 12 membered heterocyclyl, - (CH) 2 ) g -O-3 to 12 membered cycloalkyl, - (CH) 2 ) g -3 to12 membered cycloalkyl, - (CH) 2 ) g -3 to 12 membered heterocyclyl, 5 to 10 membered heteroaryl, 5 to 10 membered aryl, -S (=o) f -C 1-6 Alkyl, -O-C 2-6 Alkynyl, -O-C 2-6 Alkenyl groups;
    q is selected from 1, 2, 3 or 4.
  6. The compound, solvate, stereoisomer, deuterated compound or a pharmaceutically acceptable salt thereof according to claim 1, wherein the compound is selected from the group consisting of:
  7. a pharmaceutical composition comprising a compound of any one of claims 1 to 5, a solvate, stereoisomer, deuterated compound or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  8. Use of a compound according to any one of claims 1 to 6, a solvate, stereoisomer, deuterated compound or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 7, for the manufacture of a medicament for the prevention and/or treatment of a disease associated with the modulation of the MOR receptor agonist.
  9. The use according to claim 8, wherein said MOR receptor agonist mediated related disorders are selected from pain, immune dysfunction, inflammation, esophageal reflux, neurological and psychiatric disorders, urinary and reproductive disorders, cardiovascular disorders and respiratory disorders.
  10. The use according to claim 9, wherein the pain is selected from postoperative pain, pain caused by cancer, neuropathic pain, traumatic pain and pain caused by inflammation.
CN202380008359.0A 2022-01-19 2023-01-18 Opioid receptor agonist, preparation method and application thereof Pending CN116801881A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN202210057938 2022-01-19
CN2022100579381 2022-01-19
PCT/CN2023/072810 WO2023138603A1 (en) 2022-01-19 2023-01-18 Opioid receptor agonist, and preparation method therefor and use thereof

Publications (1)

Publication Number Publication Date
CN116801881A true CN116801881A (en) 2023-09-22

Family

ID=87347858

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202310056408.XA Pending CN117263956A (en) 2022-01-19 2023-01-15 Opioid receptor agonist, preparation method and application thereof
CN202380008359.0A Pending CN116801881A (en) 2022-01-19 2023-01-18 Opioid receptor agonist, preparation method and application thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN202310056408.XA Pending CN117263956A (en) 2022-01-19 2023-01-15 Opioid receptor agonist, preparation method and application thereof

Country Status (2)

Country Link
CN (2) CN117263956A (en)
WO (1) WO2023138603A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN118994123B (en) * 2024-10-23 2025-02-14 江苏恒瑞医药股份有限公司 Method for preparing 9-(pyridin-2-yl)-6-oxaspiro[4.5]decane derivatives

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017063509A1 (en) * 2015-10-15 2017-04-20 江苏恒瑞医药股份有限公司 Oxa spiro derivative, preparation method therefor, and applications thereof in medicines
CN109206417A (en) * 2017-07-04 2019-01-15 四川海思科制药有限公司 Opioid receptor agonists and their applications
WO2019072235A1 (en) * 2017-10-13 2019-04-18 南京明德新药研发股份有限公司 2,6-dioxaspiro [4,5] decane derivatives and preparation method therefor and pharmaceutical applications thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106588899B (en) * 2015-10-15 2019-11-15 江苏恒瑞医药股份有限公司 6- oxaspiro [4.5] decane analog derivative, preparation method and its application in medicine that pyridyl group replaces
WO2019205983A1 (en) * 2018-04-28 2019-10-31 四川科伦博泰生物医药股份有限公司 Oxa-spiro compound and preparation method therefor and uses thereof
WO2020147848A1 (en) * 2019-01-17 2020-07-23 上海海雁医药科技有限公司 Tricyclic substituted oxaspiro derivative, preparation method therefor, and pharmaceutical use thereof
CN112789276B (en) * 2019-08-14 2024-07-30 上海海雁医药科技有限公司 Analgesic compounds, their preparation and medical use

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017063509A1 (en) * 2015-10-15 2017-04-20 江苏恒瑞医药股份有限公司 Oxa spiro derivative, preparation method therefor, and applications thereof in medicines
CN109206417A (en) * 2017-07-04 2019-01-15 四川海思科制药有限公司 Opioid receptor agonists and their applications
WO2019072235A1 (en) * 2017-10-13 2019-04-18 南京明德新药研发股份有限公司 2,6-dioxaspiro [4,5] decane derivatives and preparation method therefor and pharmaceutical applications thereof

Also Published As

Publication number Publication date
CN117263956A (en) 2023-12-22
WO2023138603A1 (en) 2023-07-27

Similar Documents

Publication Publication Date Title
JP6923683B2 (en) Opioid receptor ligands and their usage and manufacturing methods
TWI615393B (en) Acrylic derivative, preparation method thereof and use thereof in medicine
US20070254903A1 (en) Novel Spiroindoline or Spiroisoquinoline Compounds, Methods of Use and Compositions Thereof
CN104024251A (en) Benzenesulfonamide compounds and their use as therapeutic agents
TW201102375A (en) Hedgehog pathway antagonists and therapeutic applications thereof
TR201809633T4 (en) Pyrrolidine GPR40 modulators for the treatment of diseases such as diabetes.
CN116801881A (en) Opioid receptor agonist, preparation method and application thereof
CN112876463B (en) Intermediate for preparing PD-L1 antagonist and preparation method thereof
JP2020528922A (en) Thiazolopyridine derivative as an adenosine receptor antagonist
CN116239606A (en) A pyrrolesulfonyl derivative, its preparation method and application
HU228314B1 (en) Chemokine receptor antagonists, their use and pharmaceutical compositions containing them
CN107501279A (en) Furoquinoline diketones and their medicinal uses
TW202423910A (en) Compounds
JP6185650B2 (en) 5-Amino-quinoline-8-carboxamide derivatives as 5-HT4 receptor agonists
JP2024532558A (en) Thieno-fused ring compounds and their preparation and use
WO2022156449A1 (en) Novel benzoazepine compound, and composition and use thereof
CN116162093B (en) A kind of TYK2 inhibitor compound and its application
CN118184641A (en) Opioid receptor agonist, preparation method and application thereof
WO2023030319A1 (en) Opioid receptor agonist, and preparation method therefor and use thereof
CN117126146A (en) Opioid receptor agonist, preparation method and application thereof
TW202227056A (en) Vhl ligands and methods of use thereof
WO2024169788A1 (en) Fused ring compound and use thereof
TW202320769A (en) A kind of degradation agent and its application
CN119161287A (en) Acetylbenzamide derivatives, preparation method and application thereof
CN115850242A (en) Quinoline-2 (1H) -ketone derivative and preparation method and application thereof

Legal Events

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