WO2018214929A1 - 一种穿心莲内酯改构化合物的盐型、晶型及其制备方法 - Google Patents

一种穿心莲内酯改构化合物的盐型、晶型及其制备方法 Download PDF

Info

Publication number
WO2018214929A1
WO2018214929A1 PCT/CN2018/088168 CN2018088168W WO2018214929A1 WO 2018214929 A1 WO2018214929 A1 WO 2018214929A1 CN 2018088168 W CN2018088168 W CN 2018088168W WO 2018214929 A1 WO2018214929 A1 WO 2018214929A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
cell
crystal form
cells
Prior art date
Application number
PCT/CN2018/088168
Other languages
English (en)
French (fr)
Inventor
夏建华
江志赶
贺海鹰
王静
Original Assignee
南京明德新药研发股份有限公司
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 南京明德新药研发股份有限公司 filed Critical 南京明德新药研发股份有限公司
Priority to CN201880033723.8A priority Critical patent/CN110650953B/zh
Priority to ES18805893T priority patent/ES2955294T3/es
Priority to US16/616,300 priority patent/US11299482B2/en
Priority to EP18805893.7A priority patent/EP3632909B1/en
Publication of WO2018214929A1 publication Critical patent/WO2018214929A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory 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
    • C07D405/12Heterocyclic 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 linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • the invention relates to a salt form, a crystal form of a andrographolide modified compound, a preparation method thereof and a medical use thereof.
  • Idiopathic pulmonary fibrosis is a chronic, progressive, irreversible, and lethal fibrotic interstitial pneumonia. "Idiopathic" means that the exact cause of the disease is not known. Unexplained antigenic stimuli such as smoking and smog cause repeated damage to the lung tissue. In order to repair the damaged lung tissue, the increase in collagen and other substances increases the interstitial lung mass. It causes fibrosis of the lungs and irreversibly loses the function of supplying oxygen to other tissues and organs. Clinically, the disease is caused by hypoxia, which causes progressive dyspnea, dry cough, and gradual decline in lung function, which quickly leads to respiratory failure and death.
  • the invention provides a compound of formula (II).
  • the present invention provides a crystalline form A of a compound of formula (II), characterized in that its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 theta angles: 10.00 ⁇ 0.2°, 12.91 ⁇ 0.2°, 16.27 ⁇ 0.2°.
  • the A crystal form has an X-ray powder diffraction pattern having characteristic diffraction peaks at the following 2 ⁇ angles: 5.05 ⁇ 0.2°, 10.00 ⁇ 0.2°, 12.91 ⁇ 0.2°, 13.42 ⁇ 0.2°, 14.51 ⁇ 0.2°, 14.94 ⁇ 0.2°, 16.27 ⁇ 0.2°, 18.36 ⁇ 0.2°.
  • the above A crystal form has an XRPD pattern as shown in FIG.
  • the XRPD pattern analysis data of the above A crystal form is as shown in Table 1:
  • the A crystalline form has a differential scanning calorimetry curve having an endothermic peak at 119.27 ° C ⁇ 3 ° C and 144.42 ° C ⁇ 3 ° C.
  • the A crystal form described above has a DSC pattern as shown in FIG.
  • the above A crystal form has a thermogravimetric analysis curve having a weight loss of 0.1268% ⁇ 0.1% at 120.00 ° C ⁇ 3 ° C.
  • the A crystal form has a TGA pattern as shown in FIG.
  • the present invention provides a process for the preparation of the crystalline form of the compound A of the formula (II), which comprises adding a compound of the formula (II) in any form to an alcoholic organic solvent for heating or recrystallization or recrystallization.
  • the above alcohol solvent is selected from the group consisting of methanol, ethanol or isopropanol.
  • the beating temperature is selected from the group consisting of 35 ° C to 45 ° C.
  • the beating time is selected from the group consisting of 12 hours to 36 hours.
  • the weight ratio of the compound of formula (II) to the alcoholic organic solvent is selected from the group consisting of 1:1 to 3.
  • the present invention also provides the use of the above compound of the formula (II), or the above crystal form A, for the preparation of a medicament for the treatment of idiopathic pulmonary fibrosis, respiratory infection or pneumonia.
  • the invention also provides the use of compound h for the preparation of a medicament for the treatment of idiopathic pulmonary fibrosis, respiratory infection or pneumonia.
  • intermediate compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, combinations thereof with other chemical synthesis methods, and those skilled in the art.
  • Well-known equivalents, preferred embodiments include, but are not limited to, embodiments of the invention.
  • the present invention employs the following abbreviations: rt stands for room temperature; THF stands for tetrahydrofuran; NMP stands for N-methylpyrrolidone; MeSO 3 H stands for methanesulfonic acid; DME stands for ethylene glycol dimethyl ether; DCM stands for dichloromethane; Xphos stands for 2-bicyclohexylphosphine-2'4'6'-triisopropylbiphenyl; EtOAc stands for ethyl acetate; MeOH stands for methanol; acetone stands for acetone; 2-Me-THF stands for 2-methyltetrahydrofuran; IPA stands for isopropyl alcohol.
  • XRPD X-ray powder diffractometer
  • Test method Approximately 10-20 mg samples were used for XRPD detection.
  • DSC Differential Scanning Calorimeter
  • Test method Take the sample ( ⁇ 1mg) and put it into the DSC aluminum pot for testing. Heat the sample from 30 °C (room temperature) to 300 °C (or 350) at a heating rate of 10 °C/min under 50 mL/min N 2 . °C).
  • TGA Thermal Gravimetric Analyzer
  • Test method The sample (2-5 mg) was placed in a TGA platinum pot for testing, and the sample was heated from room temperature to 300 ° C or weight loss by 20% at a heating rate of 10 ° C / min under 25 mL / min N 2 .
  • the compound of the formula (II) mentioned in the present invention has good crystal form stability and is easy to be used for medicine; its inhibitory effect on cytokines related to the IPF-related pathway is obvious, and is found by the left unilateral pulmonary fibrosis model of SD rats, II) The compound has a significant inhibitory effect on IPF.
  • Figure 1 XRPD spectrum of Cu-K ⁇ radiation of the crystalline form of Compound A of formula (II);
  • Figure 4 A stereostructure ellipsoid of a single molecule of the compound of formula (III-1), comprising one molecule of a dichloromethane solvent;
  • Figure 5 A stereostructure ellipsoid of a bimolecular compound of formula (III-1) comprising one molecule of a dichloromethane solvent;
  • Figure 6 A cell stacking diagram of the compound of the formula (III-1) along the a-axis direction, which contains 1 molecule of a dichloromethane solvent.
  • the crude product (1091.8 g, 1.96 mol) was added to a 5 L reaction flask at a temperature of 40 ° C, then MeOH (3037 mL) was added. After the addition was completed, the reaction was stirred at 40 ° C for 24 h. Filtration, the filter cake was washed with MeOH (200 mL) and then filtered to give a crude material, which was dried in vacuo to give 869.34 g of fine white solid.
  • the compound h (1.0 g, 2.24 mmol) was dissolved in acetone (10 mL), and a solution of i (1.3 g, 3.36 mmol) in acetone (5 mL) was added at 15 ° C. After the addition was completed, the reaction was stirred at 15 ° C for 1 h. The reaction mixture was filtered, and the filter cake was washed with acetone (20mL) and filtered again, and the filter cake was dried in a vacuum oven to obtain 0.92 g of a white solid compound (III).
  • the single crystal data of the compound of the formula (III-1) are as follows.
  • Form A solid was weighed separately, placed on the bottom of the glass vial, sealed with a screw cap, and stored at -20 ° C for use as a control.
  • Biochemical test 1 THP-1 cell release IL-6 test
  • the inhibitory effect of the compound on LPS-induced release of IL-6 by THP-1 cells was evaluated by measuring the level of IL-6 in the cell culture supernatant.
  • THP-1 cell culture medium (RPMI 1640, Gibco #22400-089, 10% serum Gibco #10099-141)
  • THP-1 cells Dilute THP-1 cells with medium to 5 ⁇ 10 5 /mL and add the cells to a 96-well plate (100 ul / well, 5 ⁇ 10 5 cells / well);
  • the LPS 1 mg/ml solution was diluted to 800 ng/ml with the culture medium, and added to the cell culture well, 50 ul/well.
  • the cell culture plate was placed in a 37 ° C incubator, and the supernatant was collected after 24 hours of culture.
  • the results of the IL-6 level in the supernatant of the CBA assay are shown in Table 5:
  • Biochemical test 2 Test of Th1 pathway cytokine TNF- ⁇ and IL-6 release from mouse spleen cells
  • Th1 pathway cytokine TNF- ⁇ and IL-6 released by LPS-induced mouse spleen cells were evaluated by detecting the levels of Th1 pathway cytokines such as TNF- ⁇ and IL-6 in mouse spleen cell culture supernatants. Inhibition.
  • mice were sacrificed under aseptic conditions, the spleen was taken, and the spleen was ground in a filter having a pore size of 70 ⁇ m, and the obtained cell suspension was collected;
  • the LPS was prepared into a 4 ⁇ g/mL solution using a culture solution, and added to a cell culture well at 50 ⁇ L/well.
  • the compound was dissolved in DMSO to 100 mM, and diluted with DMSO into 3 gradients, respectively, 30 mM, 10 mM, 3 mM, respectively, 4 ⁇ L was added to 1 mL of the culture solution to prepare 120 ⁇ M, 40 ⁇ M, 12 ⁇ M; 50 ⁇ L was added to the corresponding wells.
  • the final concentrations of the compounds were 30 ⁇ M, 10 ⁇ M, and 3 ⁇ M, respectively.
  • Dexamethasone was added as a positive drug to individual cell wells at a final concentration of 100 nM.
  • Pirfenidone was added as a reference compound to individual cell wells at a final concentration of 500 ⁇ M.
  • the cell culture plate was placed in a 37 ° C incubator, and after 24 hours of culture, the supernatant was collected, and the levels of TNF- ⁇ and IL-6 in the supernatant were measured by CBA.
  • Biochemical test 3 Test of Th1 pathway cytokine IFN- ⁇ and IL-2 release from mouse spleen cells
  • the inhibitory effect of the compound on the release of Th1 pathway cytokine levels IFN- ⁇ and IL-2 from mouse spleen cells was evaluated by measuring the levels of Th1 pathway cytokines such as IFN- ⁇ and IL-2 in the culture supernatant of mouse spleen cells.
  • the anti-mouse CD3 antibody was diluted to 5 ⁇ g/mL with DPBS, added to a 96-well plate at 100 ⁇ L/well, and the plate was allowed to stand at 4 ° C overnight;
  • mice were sacrificed under aseptic conditions, the spleen was taken, and the spleen was ground in a filter having a pore size of 70 ⁇ m, and the obtained cell suspension was collected;
  • the compound was dissolved in DMSO to 100 mM, and diluted with DMSO into 3 gradients, respectively, 30 mM, 10 mM, 3 mM, respectively, 4 ⁇ L was added to 1 mL of the culture solution to prepare 120 ⁇ M, 40 ⁇ M, 12 ⁇ M; 50 ⁇ L was added to the corresponding wells.
  • the final concentrations of the compounds were 30 ⁇ M, 10 ⁇ M, and 3 ⁇ M, respectively.
  • Dexamethasone was added as a positive drug to individual cell wells at a final concentration of 100 nM.
  • Pirfenidone was added as a reference compound to individual cell wells at a final concentration of 500 ⁇ M.
  • the cell culture plate was placed in a 37 ° C incubator, and after 24 hours of culture, the supernatant was collected, and the levels of IFN- ⁇ and IL-2 in the supernatant were measured by CBA.
  • Biochemical test 4 Mouse spleen cells release Th2 pathway cytokine IL-13 test
  • the inhibitory effect of the compound on the release of Th2 pathway cytokine IL-13 from mouse spleen cells induced by LPS was evaluated by measuring the level of Th2 pathway cytokine IL-13 in the culture supernatant of mouse spleen cells.
  • CD4+ T cells isolated and sorted from the spleens of normal C57BL/6 mice
  • the anti-mouse CD3 antibody was diluted to 5 ⁇ g/mL with DPBS, added to a 96-well plate at 100 ⁇ L/well, and the plate was allowed to stand at 4 ° C overnight;
  • mice were sacrificed under aseptic conditions, the spleen was taken, and the spleen was ground in a filter having a pore size of 70 ⁇ m, and the obtained cell suspension was collected;
  • CD4+ T cells were sorted by mouse CD4+ T cell sorting kit, and the cell concentration was adjusted to 1 ⁇ 10 6 /mL;
  • Mouse anti-IFN ⁇ antibody and anti-IL-12 antibody were simultaneously added to the culture medium at a concentration of 40 ⁇ g/mL, and mouse IL-4 was added at a concentration of 40 ng/mL.
  • the prepared induction solution was added to the inoculation.
  • the cells were coated with 50 ⁇ L/well in the well of the anti-CD3 antibody; the three wells were left without the inducing solution as a control;
  • the compound was dissolved in DMSO to 100 mM, and diluted with DMSO into 3 gradients, respectively, 30 mM, 10 mM, 3 mM, respectively, 4 ⁇ L was added to 1 mL of the culture solution to prepare 120 ⁇ M, 40 ⁇ M, 12 ⁇ M; 50 ⁇ L was added to the corresponding wells.
  • the final concentrations of the compounds were 30 ⁇ M, 10 ⁇ M, and 3 ⁇ M, respectively.
  • Dexamethasone was added as a positive drug to individual cell wells at a final concentration of 100 nM.
  • Pirfenidone was added as a reference compound to individual cell wells at a final concentration of 500 ⁇ M.
  • the cell culture plate was placed in a 37 ° C incubator, and after 72 hours of culture, the supernatant was collected, and the level of IL-13 in the supernatant was measured by CBA.
  • Biochemical test 5 TGF- ⁇ test in mouse peritoneal macrophages
  • the inhibitory effect of the compound on the release of TGF- ⁇ from mouse peritoneal macrophages was evaluated by measuring the level of TGF- ⁇ in the culture supernatant of mouse peritoneal macrophages.
  • mice are sacrificed under aseptic conditions, and the abdominal skin is cut open to expose the abdominal wall;
  • the cells were resuspended in a culture medium containing 10% serum, and then plated in a Petri dish, and cultured at 37 ° C for 2 hours, and the suspension cells were washed away with DPBS;
  • the compound was dissolved in DMSO to 100 mM, and diluted with DMSO into 3 gradients, respectively, 30 mM, 10 mM, 3 mM, respectively, 4 ⁇ L was added to 1 mL of the culture solution to prepare 120 ⁇ M, 40 ⁇ M, 12 ⁇ M; 125 ⁇ L was added to the corresponding wells.
  • the final concentrations of the compounds were 30 ⁇ M, 10 ⁇ M, and 3 ⁇ M, respectively.
  • Dexamethasone was added as a positive drug to individual cell wells at a final concentration of 100 nM.
  • Pirfenidone was added as a reference compound to individual cell wells at a final concentration of 500 ⁇ M.
  • the cell culture plate was placed in a 37 ° C incubator, cultured for 24 hours, and the supernatant was collected, and the level of TGF- ⁇ in the supernatant was measured by ELISA.
  • the compound of formula (II) has obvious inhibitory effects on the cytokines of Th1, Th2 related pathway and the fibrosis factor TGF- ⁇ .
  • the inhibitory effect of the compound of formula (II) on IPF was found by the left unilateral pulmonary fibrosis model of SD rats.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pulmonology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)

Abstract

本发明公开了穿心莲内酯改构化合物的盐型、晶型、制备方法及其医药用途。

Description

一种穿心莲内酯改构化合物的盐型、晶型及其制备方法
相关申请的引用
本申请要求于2017年05月24日向中华人民共和国国家知识产权局提交的第201710373576.6号中国发明专利申请的权益,在此将其全部内容以援引的方式整体并入本文中。
技术领域
本发明涉及穿心莲内酯改构化合物的盐型、晶型、其制备方法及其医药用途。
背景技术
特发性肺纤维化(IPF)是一种慢性,进展性,不可逆,致死性的纤维化间质性肺炎。“特发性”指不知道确切发病原因,不明原因的抗原刺激如吸烟和霧霾导致肺组织反复受损,为了修复受损的肺组织,胶原蛋白等物质产生增加使肺间质增厚,导致肺的纤维化,不可逆转的丧失原本向其它组织器官供氧的功能。临床上该病以缺氧导致渐进呼吸困难,干咳,和肺功能逐渐下降为主要表现,很快导致呼吸衰竭而死亡。具有进展快、致残率和致死率极高的特点,患者的平均预期寿命为诊断后的3-5年,又被称为不是癌症的癌症。2008年全世界至少有500万人患病。2015年研究发现IPF的全球发病率达14-59人/10万,每年有3-3.5万新病例被确诊。我国有特发性肺纤维化患者60万左右,近年IPF发病率呈明显增长趋势。因缺乏统一的,完善的IPF诊断标准,很多患者还没有被确切诊断,所以实际患者远远大于60万。75岁以上老年人的发病率是35岁以下人群的100倍,70%的患者正在吸烟或有吸烟史。IPF中位生存期平均2~5年,5年死亡率高达70%。
发明内容
本发明提供了式(II)化合物。
Figure PCTCN2018088168-appb-000001
本发明提供了式(II)化合物的A晶型,其特征在于其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:10.00±0.2°,12.91±0.2°,16.27±0.2°。
本发明的一些方案中,上述A晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.05±0.2°、10.00±0.2°,12.91±0.2°,13.42±0.2°,14.51±0.2°,14.94±0.2°,16.27±0.2°,18.36±0.2°。
本发明的一些方案中,上述A晶型,其XRPD图谱如图1所示。
本发明的一些方案中,上述A晶型的XRPD图谱解析数据如表1所示:
表1 式(II)化合物A晶型的XRPD解析数据
Figure PCTCN2018088168-appb-000002
本发明的一些方案中,上述A晶型,其差示扫描量热曲线在119.27℃±3℃和144.42℃±3℃处具有吸热峰。
本发明的一些方案中,上述A晶型,其DSC图谱如图2所示。
本发明的一些方案中,上述A晶型,其热重分析曲线在120.00℃±3℃时失重达0.1268%±0.1%。
本发明的一些方案中,上述A晶型,其TGA图谱如图3所示。
本发明提供了式(II)化合物A晶型的制备方法,包括将任意一种形式的式(II)化合物加入到醇类有机溶剂中加热打浆或重结晶制得。
本发明的一些方案中,上述醇类溶剂选自:甲醇、乙醇或异丙醇。
本发明的一些方案中,上述打浆温度选自35℃~45℃。
本发明的一些方案中,上述打浆时间选自12小时~36小时。
本发明的一些方案中,上述式(II)化合物与醇类有机溶剂的重量比选自1∶1~3。
本发明还提供了上述式(II)化合物,或上述A晶型在制备治疗特发性肺纤维化、呼吸道感染或肺炎相关药物上的应用。
本发明还提供了化合物h在制备治疗特发性肺纤维化、呼吸道感染或肺炎相关药物上的应用。
Figure PCTCN2018088168-appb-000003
定义和说明
除非另有说明,本文所用的下列术语和短语旨在含有下列含义。一个特定的短语或术语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文出现商品名时,旨在指代其对应的商品或其活性成分。
本发明的中间体化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。
本发明具体实施方式的化学反应是在合适的溶剂中完成的,所述的溶剂须适合于本发明的化学变化及其所需的试剂和物料。为了获得本发明的化合物,有时需要本领域技术人员在已有实施方式的基础上对合成步骤或者反应流程进行修改或选择。
下面会通过实施例具体描述本发明,这些实施例并不意味着对本发明的任何限制。
本发明所使用的所有溶剂是市售的,无需进一步纯化即可使用。
本发明采用下述缩略词:r.t.代表室温;THF代表四氢呋喃;NMP代表N-甲基吡咯烷酮;MeSO 3H代表甲烷磺酸;DME代表乙二醇二甲醚;DCM代表二氯甲烷;Xphos代表2-双环己基膦-2’4’6’-三异丙基联苯;EtOAc代表乙酸乙酯;MeOH代表甲醇;acetone代表丙酮;2-Me-THF代表2-甲基四氢呋喃;IPA代表异丙醇。
化合物经手工或者
Figure PCTCN2018088168-appb-000004
软件命名,市售化合物采用供应商目录名称。
本发明粉末X-射线衍射(X-ray powder diffractometer,XRPD)方法
仪器型号:布鲁克D8 advance X-射线衍射仪
测试方法:大约10~20mg样品用于XRPD检测。
详细的XRPD参数如下:
光管:Cu,kα,
Figure PCTCN2018088168-appb-000005
光管电压:40kV,光管电流:40mA
发散狭缝:0.60mm
探测器狭缝:10.50mm
防散射狭缝:7.10mm
扫描范围:4-40deg
步径:0.02deg
步长:0.12秒
样品盘转速:15rpm
本发明差热分析(Differential Scanning Calorimeter,DSC)方法
仪器型号:TA Q2000差示扫描量热仪
测试方法:取样品(~1mg)置于DSC铝锅内进行测试,在50mL/min N 2条件下,以10℃/min的升温速率,加热样品从30℃(室温)到300℃(或350℃)。
本发明热重分析(Thermal Gravimetric Analyzer,TGA)方法
仪器型号:TA Q5000IR热重分析仪
测试方法:取样品(2~5mg)置于TGA铂金锅内进行测试,在25mL/min N 2条件下,以10℃/min的升温速率,加热样品从室温到300℃或失重20%。
技术效果
本发明提及的式(II)化合物晶型稳定性好,易于成药;其对与IPF相关通路的细胞因子的抑制作用明显,并通过SD大鼠左侧单侧肺纤维化模型发现,式(II)化合物对IPF的抑制 作用显著。
附图说明
图1:式(II)化合物A晶型的Cu-Kα辐射的XRPD谱图;
图2:式(II)化合物A晶型的DSC谱图;
图3:式(II)化合物A晶型的TGA谱图;
图4:式(III-1)化合物单分子的立体结构椭球图,其包含1分子的二氯甲烷溶剂;
图5:式(III-1)化合物双分子的立体结构椭球图,其包含1分子的二氯甲烷溶剂;
图6:式(III-1)化合物沿a轴方向的晶胞堆积图,其包含1分子的二氯甲烷溶剂。
具体实施方式
为了更好的理解本发明的内容,下面结合具体实施例来做进一步的说明,但具体的实施方式并不是对本发明的内容所做的限制。
实施例1:式(II)化合物的制备
Figure PCTCN2018088168-appb-000006
第一步:化合物b的制备
将化合物a(300.00g,856.04mmol)溶于二氯甲烷(3.00L),依次加入环戊基甲醛(84.85g,864.60mmol)和离子交换树脂-15(300.00g),然后20℃搅拌12小时。体系过滤浓缩得到300克白色固体化合物b,产率:81.39%。
1H NMR(400MHz,CDCl3)δ6.97(d,J=6.4Hz,1H),5.05(s,1H),4.92(s,1H),4.62(s,2H),4.46(d,J=6Hz,1H),4.29-4.26(m,1H),4.03(d,J=10.8Hz,1H),3.49-3.44(m,2H),2.59-2.46(m,4H),2.21(s,1H),2.08-1.87(m,2H),1.85-1.71(m,3H),1.69-1.56(m,9H),1.54(s,3H),1.52-1.26(m,3H),0.83(s,3H)。
第二步:化合物c的制备
将化合物b(200.00g,464.49mmol)溶于2000mL二氯甲烷,0℃加入乙酸酐(490.50g,4.80mol)和吡啶(392.00g,4.96mol)。然后20℃搅拌17小时。反应液在35℃减压浓缩,加入6000mL水,有沉淀物析出,过滤所得残余物用石油醚(500mL*2)打浆得200克化合物c粗产品。
1H NMR(400MHz,CDCl 3)δ7.02(br.s.,1H),6.71(t,J=6.9Hz,1H),6.19(d,J=3.0Hz,1H),4.88(s,1H),4.62(d,J=5.5Hz,1H),4.45(s,1H),4.04(d,J=11.3Hz,1H),3.55-3.36(m,2H),2.63-2.52(m,1H),2.48-2.38(m,2H),2.35-2.23(m,1H),2.15-1.99(m,2H),1.94-1.82(m,3H),1.72(br.s.,3H),1.64-1.45(m,6H),1.39(s,3H),1.27(br.s.,3H),0.83(s,3H)。
第三步:化合物d的制备
将化合物c(200.00g,484.78mmol)溶于2000mL四氢呋喃,0℃加入溶于2000mL水的高锰酸钾(229.83g,1.45mol)溶液,然后20℃搅拌6小时。加入1000mL盐水,分液,有机相减压浓缩,所得残余物溶于1000mL乙酸乙酯,加入9000mL石油醚,过滤。滤液减压浓缩得78克化合物d粗产品。
1H NMR(400MHz,CDCl 3)δ9.65(d,J=2.0Hz,1H),4.85(s,1H),4.60(d,J=6.0Hz,1H),4.43(s,1H),4.03(d,J=11.0Hz,1H),3.53-3.42(m,2H),2.55-2.21(m,5H),2.14-2.06(m,2H),1.86-1.80(m,1H),1.72-1.67(m,3H),1.61-1.43(m,7H),1.38(s,3H),1.26-1.09(m,3H),0.83-0.66(m,3H)。
第四步:化合物e的制备
将化合物d(70.00g,202.02mmol)溶于1000mL四氢呋喃,0℃加入硼氢化钠(22.93g,606.06mmol),然后25℃搅拌4小时。加入500mL水淬灭,用乙酸乙酯萃取(200mL*5),合并有机相,用饱和氯化钠溶液洗涤(200mL*1),用无水硫酸钠干燥,过滤,滤液减压浓缩后,用硅胶柱色谱法以洗脱剂体系PE∶EA=10∶1至2∶1纯化所得残余物得40克化合物e,产率:56.8%。
1H NMR(400MHz,CDCl 3)δ4.87(s,1H),4.61(d,J=6Hz,2H),4.03(d,J=11.2Hz,1H),3.75(s,1H),3.52-3.43(m,3H),2.44-2.41(m,2H),2.08-1.75(m,3H),1.74-1.66(m,7H),1.57-1.54(m,6H),1.37(s,4H),1.26-1.25(m,3H),0.77(s,3H)。
第五步:化合物f的制备
将化合物e(35.00g,100.42mmol)溶于500mL二氯甲烷,25℃加入四溴化碳(36.63g,110.46mmol)和三苯基磷(28.97g,110.46mmol),然后25℃搅拌4小时。加入200mL水淬灭,用二氯甲烷(200mL*3)萃取,合并有机相,用饱和氯化钠溶液洗涤(200mL*1),用 无水硫酸钠干燥,过滤,滤液减压浓缩后,用硅胶柱色谱法以洗脱剂体系PE∶EtOAc=10∶1至5∶1纯化所得残余物得40克化合物f,产率:96.82%。
1H NMR(400MHz,CDCl 3)δ4.89(s,1H),4.61(d,J=6Hz,1H),4.53(s,1H),4.02(d,J=11.2Hz,1H),3.55-3.52(m,3H),3.46-3.29(m,1H),2.42(s,1H),2.08(s,1H),2.04-1.84(m,4H),1.84-1.81(m,3H),1.72-1.70(m,3H),1.59-1.54(m,6H),1.38(s,3H),1.26-1.22(m,3H),0.78(s,3H)。
第六步:化合物h的制备
将1-甲基哌啶-4-醇276g(84mg,0.73mmol)溶于N,N二甲基甲酰胺(10mL),在0℃加入钠氢(32mg,1.01mmol)并在0℃搅拌15分钟。将化合物f(300mg,0.73mmol)加入到反应液中,室温搅拌12小时。将反应液加入水(5mL),用乙酸乙酯(30mL)萃取,有机层用水(10mL x 3)洗涤,用无水硫酸钠干燥,过滤,滤液减压浓缩后经制备液相分离得到20mg化合物h,产率:6.1%。
MS m/z(ESI):446.6[M+1]
1H NMR(400MHz,CDCl 3)δ8.44-8.55(m,1H),4.54-4.66(m,4H),4.02-4.07(m,1H),3.40-3.53(m,3H),2.96-3.16(m,3H),2.77-2.92(m,2H),2.56-2.65(m,3H),2.25-2.40(m,2H),1.45-2.03(m,19H),1.30(s,3H),1.15-1.26(m,3H),0.76(s,3H)。
第七步:式(II)化合物的制备
将化合物h(1.55kg,3.28mol)溶于Acetone(15.5L)中,然后将该溶液加入到30L反应釜中,控制温度在22℃,加入含水柠檬酸(550.5g,2.62mol)的丙酮(7.8L)溶液,反应在22℃下继续搅拌17.7h,反应液中有大量固体析出,在氮气保护下过滤,滤饼加入丙酮(5.0L*4)洗涤,并重新过滤,在22℃下将滤饼加入到30L反应釜中,然后加入丙酮(15.5L),反应液在22℃下继续搅拌21.4h,在氮气保护下过滤,滤饼在真空干燥箱40℃干燥48h得到1.3067kg粗品灰白色固体式(II)化合物。
实施例2:A晶型的制备
控制温度在40℃,将粗品灰白色固体式(II)化合物(1091.8g,1.96mol)加入到5L反应瓶中,然后加入MeOH(3037mL),加料完毕,反应在40℃搅拌24h,反应液趁热过滤,滤饼用MeOH(200mL)洗涤,然后过滤得到粗品,粗品经真空干燥箱干燥得到869.34g精品白色固体即为A晶型。
实施例3:式(III)化合物的制备
Figure PCTCN2018088168-appb-000007
将化合物h(1.0g,2.24mmol)溶于丙酮(10mL)中,在15℃加入i(1.3g,3.36mmol)的丙酮(5mL)溶液,滴加完毕,反应在15℃下继续搅拌1h,反应液过滤,滤饼用丙酮(20mL)洗涤并重新过滤,滤饼在真空干燥箱干燥得到0.92g白色固体式(III)化合物。
在二氯甲烷和甲醇(体积比二氯甲烷∶甲醇=3∶7)的混合溶剂中培养式(III)化合物的单晶,并测试确认其绝对构型如式(III-1)所示。
Figure PCTCN2018088168-appb-000008
根据式(III-1)化合物构型,可推知式(II)化合物绝对构型如式(II-1)所示。
Figure PCTCN2018088168-appb-000009
式(III-1)化合物的单晶数据如下。
式(III-1)化合物晶体结构精修信息
Figure PCTCN2018088168-appb-000010
Figure PCTCN2018088168-appb-000011
式(III-1)化合物原子坐标参数及等价温度因子值
Figure PCTCN2018088168-appb-000012
Figure PCTCN2018088168-appb-000013
Figure PCTCN2018088168-appb-000014
式(III-1)化合物成键原子的键长和键角值
Figure PCTCN2018088168-appb-000015
Figure PCTCN2018088168-appb-000016
Figure PCTCN2018088168-appb-000017
Figure PCTCN2018088168-appb-000018
Figure PCTCN2018088168-appb-000019
Figure PCTCN2018088168-appb-000020
Figure PCTCN2018088168-appb-000021
Figure PCTCN2018088168-appb-000022
式(III-1)化合物扭转角值
Figure PCTCN2018088168-appb-000023
Figure PCTCN2018088168-appb-000024
Figure PCTCN2018088168-appb-000025
实验例1:A晶型在高温,高湿条件下的固体稳定性试验
平行称取2份A晶型样品,每份大约100mg,置于玻璃样品瓶的底部,摊成薄薄一层。样品用铝箔纸封瓶口,并在铝箔纸上扎些小孔,保证样品能与环境空气充分接触,置于40℃/75%湿度条件恒温恒湿箱。在上述条件下放置的样品于第5天,10天取样检测,检测结果与0天的初始检测结果进行比较,试验结果见下表-2所示:
表-2 A晶型的固体稳定性试验
时间点(天) 外观 晶型 纯度(%) 总杂质(%)
0 灰白色粉末 A晶型 98.51 1.49
5 灰白色粉末 A晶型 98.37 1.61
10 灰白色粉末 A晶型 98.45 1.55
实验结论:本发明晶型稳定性好,易于成药。
实验例2:A晶型在不同温度和湿度及光照条件下固体物理稳定性试验
平行称取2份式A晶型固体,每份大约100mg,放置于玻璃样品瓶的底部,摊成薄薄一层,铝箔纸封瓶口,并在铝箔纸上扎些小孔,保证样品能与环境空气充分接触。把制备的4份样品分别放置于25℃/92.5%的相对湿度,60℃,40℃/75%及光照条件下,考察样品第10天的物理稳定性。同时,单独称取一份大约100mg A晶型固体,放置于玻璃样品瓶的底部,用螺纹瓶盖密封后,保存于-20℃条件下,作为对照品使用。在第10天,取出所有样品,恢复至室温,观察样品外观变化,并用XRPD检测样品晶型。通过对加速样品与对照样品的比较,判断式(II)化合物A晶型的固体物理稳定性。下表-3为A晶型固体物理稳定性实验结果。
表-3 A晶型在不同温度和湿度条件下及光照下固体物理稳定性试验
Figure PCTCN2018088168-appb-000026
实验例3:式(II)化合物的多晶型研究
将式(II)化合物在相应溶剂中,在40℃下避光搅拌2天,将溶液离心取沉淀干燥后经XRPD检测,结果如下表4所示:
表4 式(II)化合物的多晶型研究
编号 溶剂 晶型
1 甲醇 A晶型
2 乙醇 A晶型
由上表可知,在上述溶剂条件下,式(II)化合物的A晶型稳定。
生化试验1:THP-1细胞释放IL-6测试
实验目的:
通过检测细胞培养上清内IL-6的水平,来评价化合物对LPS引起的THP-1细胞释放IL-6水平的抑制作用。
实验材料:
细胞系:THP-1细胞系
THP-1细胞培养基(RPMI 1640,Gibco#22400-089,10%血清Gibco#10099-141)
LPS,1mg/ml(Sigma#L5293)
DPBS(Hyclone,#SH30028.01B)
Human IL-6 CBA kit,BD#558276
CBA Human Soluble Protein Master Buffer Kit,BD#558265
地塞米松:J&K#308890
96孔细胞板,Corning#
CO 2培养箱,Thermo#371
离心机,Eppendorf#5810R
Vi-cell细胞计数仪,Beckman Coulter
FACSCalibur,BD#97500540
实验步骤和方法:
a)细胞接种
1)37℃水浴预热培养基。
2)将培养瓶中的悬液细胞吹匀,转移至离心管中,室温1200rpm离心5分钟,弃上清,加培养液重悬至10ml;
3)吸取1mL细胞重悬液,用Vi-cell计数;
4)用培养基稀释THP-1细胞到5×10 5/mL将细胞加入到96孔板(100ul/孔,5×10 5细胞/孔);
b)化合物加样:
1)用DMSO将化合物溶解成30mM,并用DMSO 3倍稀释成4个梯度,分别为30mM,10mM,3mM,1mM,分别取4ul溶液加至1ml培养液中,配成120uM,40uM,12uM,4uM,每孔取50ul加到加有细胞的孔中,终浓度分别为30uM,10uM,3uM,1uM。地塞米松作为阳性药加入单独的细胞孔中,终浓度为100nM。
c)细胞刺激
将LPS 1mg/ml溶液用培养液稀释至800ng/ml,加入细胞培养孔中,50ul/孔。
d)细胞孵育及检测
将细胞培养板置于37℃培养箱中,培养24小时后收上清,CBA检测上清中IL-6的水平实验结果见表5:
表5 CBA检测炎症因子IL-6测试结果-抑制率@10uM
供试样品 抑制率@10uM
化合物h A
注:A>60%;
结论:化合物h对炎症因子IL-6的抑制作用显著。
生化试验2:小鼠脾脏细胞释放Th1通路细胞因子TNF-α和IL-6的测试
实验目的:
通过检测小鼠脾脏细胞培养上清内TNF-α和IL-6等Th1通路细胞因子的水平,来评价化合物对LPS引起的小鼠脾脏细胞释放Th1通路细胞因子TNF-α和IL-6水平的抑制作用。
实验材料:
细胞:由正常C57BL/6小鼠脾脏分离培养的脾脏细胞
小鼠脾脏细胞培养基(RPMI 1640,Gibco#22400-089,10%血清Gibco#10099-141)
红细胞裂解液,Gibco A10492-01
LPS,Sigma#L2630
DPBS,Hyclone,#SH30028.01B
Mouse TNF-α CBA kit,BD#558299
Mouse IL-6 CBA kit,BD#558310
CBA Mouse Soluble Protein Master Buffer Kit,BD 558267
地塞米松:J&K#308890
吡非尼酮:TA00720266
96孔细胞板,Costar 3799
CO 2培养箱,Thermo#371
离心机,Eppendorf#5810R
Vi-cell细胞计数仪,Beckman Coulter
BD FACSCanto II Flow Cytometer,338962
实验步骤和方法:
a)小鼠脾脏细胞分离及接种
1)于无菌条件下处死小鼠,取脾脏,将脾脏置于孔径70μm的滤膜中研磨,收集所得细胞悬液;
2)将细胞悬液转移至离心管中,室温1200rpm离心5分钟,弃上清;
3)按照5mL/脾脏的比例加入红细胞裂解液,重悬细胞后静置5分钟;
4)向细胞悬液中加入3倍体积的DPBS,室温1200rpm离心5分钟,弃上清;
5)将细胞重悬,用Vi-cell仪器进行细胞计数,并将细胞浓度调整至1×10 6/mL;
6)将细胞加入到96孔板(100μL/孔,1×10 5细胞/孔);
b)细胞刺激
将LPS用培养液配制成4μg/mL溶液,加入细胞培养孔中,50μL/孔。
c)化合物加样:
用DMSO将化合物溶解成100mM,并用DMSO稀释成3个梯度,分别为30mM,10mM,3mM,分别取4μL加至1mL培养液中,配成120μM,40μM,12μM;分别取50μL加到相应孔中,化合物终浓度分别为30μM,10μM,3μM。地塞米松作为阳性药加入单独的细胞孔中,终浓度为100nM。吡非尼酮作为参照化合物加入单独的细胞孔中,终浓度为500μM。
d)细胞孵育及检测
将细胞培养板置于37℃培养箱中,培养24小时后收上清,CBA检测上清中TNF-α和IL-6的水平
生化试验3:小鼠脾脏细胞释放Th1通路细胞因子IFN-γ和IL-2的测试
实验目的:
通过检测小鼠脾脏细胞培养上清内IFN-γ和IL-2等Th1通路细胞因子的水平,来评价化合物对小鼠脾脏细胞释放Th1通路细胞因子水平IFN-γ和IL-2的抑制作用。
实验材料:
细胞:由正常C57BL/6小鼠脾脏分离培养的脾脏细胞
小鼠脾脏细胞培养基(RPMI 1640,Gibco#22400-089,10%血清Gibco#10099-141)
红细胞裂解液,Gibco A10492-01
Anti-mouse CD3抗体,BD 553057
DPBS,Hyclone,#SH30028.01B
Mouse IFN-γCBA kit,BD 558296
Mouse IL-2 CBA kit,BD 558297
CBA Mouse Soluble Protein Master Buffer Kit,BD 558267
地塞米松:J&K#308890
吡非尼酮:TA00720266
96孔细胞板,Costar 3599
CO 2培养箱,Thermo#371
离心机,Eppendorf#5810R
Vi-cell细胞计数仪,Beckman Coulter
BD FACSCanto II Flow Cytometer,338962
实验步骤和方法:
a)Anti-mouse CD3抗体包被
将anti-mouse CD3抗体用DPBS稀释至5μg/mL,按照100μL/孔加入96孔板中,将培养板于4℃静置过夜;
b)小鼠脾脏细胞分离及接种
1)于无菌条件下处死小鼠,取脾脏,将脾脏置于孔径70μm的滤膜中研磨,收集所得细胞悬液;
2)将细胞悬液转移至离心管中,室温1200rpm离心5分钟,弃上清;
3)按照5mL/脾脏的比例加入红细胞裂解液,重悬细胞后静置5分钟;
4)向细胞悬液中加入3倍体积的DPBS,室温1200rpm离心5分钟,弃上清;
5)将细胞重悬,用Vi-cell仪器进行细胞计数,并将细胞浓度调整至1×10 6/mL;
c)细胞刺激
1)将96孔板从4℃取出,用DPBS洗两遍;
2)将细胞加入已包被anti-mouse CD3抗体的孔中,50μL/孔,即1×10 5/孔;同时将细胞 以同样数量加入未包被的孔中,作为阴性对照;
d)化合物加样:
用DMSO将化合物溶解成100mM,并用DMSO稀释成3个梯度,分别为30mM,10mM,3mM,分别取4μL加至1mL培养液中,配成120μM,40μM,12μM;分别取50μL加到相应孔中,化合物终浓度分别为30μM,10μM,3μM。地塞米松作为阳性药加入单独的细胞孔中,终浓度为100nM。吡非尼酮作为参照化合物加入单独的细胞孔中,终浓度为500μM。
e)细胞孵育及检测
将细胞培养板置于37℃培养箱中,培养24小时后收上清,CBA检测上清中IFN-γ和IL-2的水平
生化试验4:小鼠脾脏细胞释放Th2通路细胞因子IL-13测试
实验目的:
通过检测小鼠脾脏细胞培养上清内Th2通路细胞因子IL-13的水平,来评价化合物对LPS引起的小鼠脾脏细胞释放Th2通路细胞因子IL-13水平的抑制作用。
实验材料:
细胞:由正常C57BL/6小鼠脾脏分离并分选的CD4+T细胞
细胞培养基(RPMI 1640,Gibco#22400-089,10%血清Gibco#10099-141)
红细胞裂解液
Anti-mouse CD3抗体,BD 553057
Anti-mouse IFNγ抗体,BD 554430
Anti-mouse IL-12抗体,BD 553375
小鼠IL-4,PeproTech 214-14
小鼠CD4+T细胞分选试剂盒,STEMCELL 19852A
DPBS,Hyclone,#SH30028.01B
Mouse IL-13 CBA kit,BD 558349
CBA Mouse Soluble Protein Master Buffer Kit,BD 558267
地塞米松:J&K#308890
吡非尼酮:TA00720266
96孔细胞板,Costar 3599
CO 2培养箱,Thermo#371
离心机,Eppendorf#5810R
Vi-cell细胞计数仪,Beckman Coulter
BD FACSCanto II Flow Cytometer,338962
实验步骤和方法:
a)Anti-mouse CD3抗体包被
将anti-mouse CD3抗体用DPBS稀释至5μg/mL,按照100μL/孔加入96孔板中,将培养板于4℃静置过夜;
b)小鼠脾脏CD4+T细胞分选及接种
1)于无菌条件下处死小鼠,取脾脏,将脾脏置于孔径70μm的滤膜中研磨,收集所得细胞悬液;
2)将细胞悬液转移至离心管中,室温1200rpm离心5分钟,弃上清;
3)按照5mL/脾脏的比例加入红细胞裂解液,重悬细胞后静置5分钟;
4)向细胞悬液中加入3倍体积的DPBS,室温1200rpm离心5分钟,弃上清;
5)将细胞重悬,用Vi-cell仪器进行细胞计数,并将细胞浓度调整至1×10 8/mL:
6)采用小鼠CD4+T细胞分选试剂盒分选出CD4+T细胞,并将细胞浓度调整至1×10 6/mL;
7)将包被过夜的96孔板从4℃取出,用DPBS洗两遍;
8)将分选所得的CD4+T细胞加入已包被anti-CD3抗体的孔中,50μL/孔,即1×10 5/孔;同时,将CD4+T细胞以同样数量加入未包被的孔中,作为阴性对照;
c)Th2细胞诱导
将小鼠anti-IFNγ抗体和anti-IL-12抗体同时加入培养液中,浓度均为40μg/mL,并加入小鼠IL-4,浓度为40ng/mL;将配制好的诱导液加入接种有细胞并包被anti-CD3抗体的孔中,50μL/孔;留三孔不加诱导液,作为对照;
d)化合物加样:
用DMSO将化合物溶解成100mM,并用DMSO稀释成3个梯度,分别为30mM,10mM,3mM,分别取4μL加至1mL培养液中,配成120μM,40μM,12μM;分别取50μL加到相应孔中,使化合物终浓度分别为30μM,10μM,3μM。地塞米松作为阳性药加入单独的细胞孔中,终浓度为100nM。吡非尼酮作为参照化合物加入单独的细胞孔中,终浓度为500μM。
e)细胞孵育及检测
将细胞培养板置于37℃培养箱中,培养72小时后收上清,CBA检测上清中IL-13的水平。
生化试验5:小鼠腹腔巨噬细胞释放TGF-β测试
实验目的:
通过检测小鼠腹腔巨噬细胞培养上清内TGF-β的水平,来评价化合物对小鼠腹腔巨噬细胞释放TGF-β的抑制作用。
实验材料:
细胞:由正常Balb/c小鼠腹腔灌洗而收集的巨噬细胞
细胞培养基(RPMI 1640,Gibco#22400-089,10%血清Gibco#10099-141)
小鼠TNF-α,PeproTech 315-01A
小鼠IL-4,PeproTech 214-14
DPBS,Hyclone,#SH30028.01B
Mouse TGF-βELISA kit,R&D MB100B
地塞米松:J&K#308890
吡非尼酮:TA00720266
48孔细胞板,Costar 3548
CO 2培养箱,Thermo#371
离心机,Eppendorf#5810R
Vi-cell细胞计数仪,Beckman Coulter
酶标仪,Molecular Devices SpectraMax i3
实验步骤和方法:
a)小鼠腹腔巨噬细胞收集及接种
1)于无菌条件下处死小鼠,将腹部皮肤剪开,暴露腹壁;
2)用5mL注射器吸取无血清的RPMI 1640培养液5mL;一手用镊子将腹壁轻轻提起,另一手持注射器将培养液缓缓注入腹腔;
3)待培养液充分冲洗腹腔后,持注射器将腹腔内培养液慢慢吸出,移至离心管中;
4)将所得细胞悬液于室温1200rpm离心5分钟,弃上清;
5)将细胞用含10%血清的培养液重悬后铺于培养皿中,于37℃培养2小时,用DPBS将悬浮细胞洗去;
6)用培养液将贴壁的巨噬细胞吹打下来,并用Vi-Cell仪器进行计数;将细胞浓度调整为1.6×10 6/mL;
7)将巨噬细胞加入48孔板的孔中,250μL/孔,即4×10 5/孔;
b)细胞刺激:
1)待细胞贴壁后,将培养液换成无血清的RPMI 1640培养液,250μL/孔;
2)用无血清的培养液配制含TNFα和IL-13的刺激液,浓度均为80ng/mL,加入接种细胞的孔中,125μL/孔;留三孔不加刺激液,作为空白对照;
c)化合物加样:
用DMSO将化合物溶解成100mM,并用DMSO稀释成3个梯度,分别为30mM,10mM,3mM,分别取4μL加至1mL培养液中,配成120μM,40μM,12μM;分别取125μL加到相应孔中,使化合物终浓度分别为30μM,10μM,3μM。地塞米松作为阳性药加入单独的细胞孔中,终浓度为100nM。吡非尼酮作为参照化合物加入单独的细胞孔中,终浓度为500μM。
e)细胞孵育及检测
将细胞培养板置于37℃培养箱中,培养24小时后收上清,ELISA检测上清中TGF-β的水平。
生化试验2~5体外细胞因子筛选结果如表6所示。
表6 式(II)化合物体外细胞因子筛选结果
Figure PCTCN2018088168-appb-000027
结论:式(II)化合物对Th1,Th2相关通路的细胞因子以及纤维化因子TGF-β的抑制作用明显。
体内药效实验结果:
通过SD大鼠左侧单侧肺纤维化模型发现,式(II)化合物对IPF的抑制作用显著。

Claims (15)

  1. 式(II)化合物。
    Figure PCTCN2018088168-appb-100001
  2. 式(II)化合物的A晶型,其特征在于其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:10.00±0.2°,12.91±0.2°,16.27±0.2°。
  3. 根据权利要求2所述的A晶型,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:5.05±0.2°、10.00±0.2°,12.91±0.2°,13.42±0.2°,14.51±0.2°,14.94±0.2°,16.27±0.2°,18.36±0.2°。
  4. 根据权利要求3所述的A晶型,其XRPD图谱如图1所示。
  5. 根据权利要求2~4所述的A晶型,其差示扫描量热曲线在119.27℃±3℃和144.42℃±3℃处具有吸热峰。
  6. 根据权利要求5所述的A晶型,其DSC图谱如图2所示。
  7. 根据权利要求2~4所述的A晶型,其热重分析曲线在120.00℃±3℃时失重达0.1268%±0.1%。
  8. 根据权利要求7所述的A晶型,其TGA图谱如图3所示。
  9. 式(II)化合物A晶型的制备方法,包括将任意一种形式的式(II)化合物加入到醇类有机溶剂中加热打浆或重结晶制得。
  10. 根据权利要求9所述的制备方法,其中,醇类溶剂选自:甲醇、乙醇或异丙醇。
  11. 根据权利要求9所述的制备方法,其中,打浆温度选自35℃~45℃。
  12. 根据权利要求9所述的制备方法,其中,打浆时间选自12小时~36小时。
  13. 根据权利要求9所述的制备方法,其中,式(II)化合物与醇类有机溶剂的重量比选自1∶1~3。
  14. 根据权利要求1所述的式(II)化合物,或根据权利要求2~4任意一项所述的A晶型在制备治疗特发性肺纤维化、呼吸道感染或肺炎相关药物上的应用。
  15. 化合物h在制备治疗特发性肺纤维化、呼吸道感染或肺炎相关药物上的应用。
    Figure PCTCN2018088168-appb-100002
PCT/CN2018/088168 2017-05-24 2018-05-24 一种穿心莲内酯改构化合物的盐型、晶型及其制备方法 WO2018214929A1 (zh)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CN201880033723.8A CN110650953B (zh) 2017-05-24 2018-05-24 一种穿心莲内酯改构化合物的盐型、晶型及其制备方法
ES18805893T ES2955294T3 (es) 2017-05-24 2018-05-24 Forma salina y cristalina de compuesto andrografólido modificado y método de preparación del mismo
US16/616,300 US11299482B2 (en) 2017-05-24 2018-05-24 Salt and crystal form of modified andrographolide compound and preparation method thereof
EP18805893.7A EP3632909B1 (en) 2017-05-24 2018-05-24 Salt and crystal form of modified andrographolide compound and preparation method thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201710373576.6 2017-05-24
CN201710373576 2017-05-24

Publications (1)

Publication Number Publication Date
WO2018214929A1 true WO2018214929A1 (zh) 2018-11-29

Family

ID=64396260

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/088168 WO2018214929A1 (zh) 2017-05-24 2018-05-24 一种穿心莲内酯改构化合物的盐型、晶型及其制备方法

Country Status (6)

Country Link
US (1) US11299482B2 (zh)
EP (1) EP3632909B1 (zh)
CN (1) CN110650953B (zh)
ES (1) ES2955294T3 (zh)
PT (1) PT3632909T (zh)
WO (1) WO2018214929A1 (zh)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105367558A (zh) * 2015-08-27 2016-03-02 沈阳药科大学 穿心莲内酯衍生物及其制备方法和用途
WO2016065264A1 (en) * 2014-10-24 2016-04-28 Biogen Ma Inc. Diterpenoid derivatives and methods of use thereof
CN106800551A (zh) * 2015-11-26 2017-06-06 南京明德新药研发股份有限公司 穿心莲内酯改构化合物

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017088738A1 (zh) 2015-11-26 2017-06-01 南京明德新药研发股份有限公司 穿心莲内酯改构化合物

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016065264A1 (en) * 2014-10-24 2016-04-28 Biogen Ma Inc. Diterpenoid derivatives and methods of use thereof
CN105367558A (zh) * 2015-08-27 2016-03-02 沈阳药科大学 穿心莲内酯衍生物及其制备方法和用途
CN106800551A (zh) * 2015-11-26 2017-06-06 南京明德新药研发股份有限公司 穿心莲内酯改构化合物

Also Published As

Publication number Publication date
ES2955294T3 (es) 2023-11-29
CN110650953B (zh) 2023-01-10
CN110650953A (zh) 2020-01-03
EP3632909A1 (en) 2020-04-08
EP3632909A4 (en) 2020-12-23
US11299482B2 (en) 2022-04-12
PT3632909T (pt) 2023-09-27
US20210284630A1 (en) 2021-09-16
EP3632909B1 (en) 2023-07-12

Similar Documents

Publication Publication Date Title
KR102225233B1 (ko) 바이러스 감염의 치료를 위한 2-아미노피리미딘 유도체
Hashimoto et al. Anti-AIDS agents—XXVII. Synthesis and anti-HIV activity of betulinic acid and dihydrobetulinic acid derivatives
US7112580B2 (en) 3-nitrogen-6, 7-dioxygen steroids and uses related thereto
CN104136437B (zh) N-(5s,6s,9r)-4-(2-氧代-2,3-二氢-1h-咪唑并[4,5-b]吡啶-1-基)哌啶-1-羧酸5-氨基-6-(2,3-二氟苯基)-6,7,8,9-四氢-5h-环庚三烯并[b]吡啶-9-基酯半硫酸盐
US20090124652A1 (en) Polymorphs of 1-(2-Methylpropyl)-1H-Imidazo[4,5-C][1,5]Naphthyridin-4-Amine Ethane-Sulfonate
JP2004500347A (ja) 色素標識イミダゾキノリン化合物
CN106029668B (zh) 吡咯并[3,2]嘧啶衍生物作为人类干扰素诱导剂
KR20130122612A (ko) 오에스아이 906의 다형체
CN114702439B (zh) 一类萘基脲-哌嗪类化合物及其制备方法和应用
EP3403651B1 (en) Isocorydine derivatives, preparation method and use thereof
WO2018214929A1 (zh) 一种穿心莲内酯改构化合物的盐型、晶型及其制备方法
CN108299330B (zh) 去氢枞酸噁唑烷酮衍生物及其制备方法和应用
WO2018214958A1 (zh) Ido1抑制剂的晶型及其制备方法
CN113354577B (zh) 一种单羰基类姜黄素类化合物及其制备方法与应用
CN113166106B (zh) 一种Syk抑制剂的盐及其结晶型
WO2011157743A1 (en) Alpha-crystalline form of carbabenzpyride
JP2022517396A (ja) Egfr阻害剤の塩、結晶形及びその製造方法
EP4365171A1 (en) Crystal form of triazolopyridine-substituted indazole compound and preparation method therefor
EP4122931A1 (en) Crystalline form of tlr8 agonist
WO2023143112A1 (zh) 氮杂苯并八元环化合物的盐型、晶型及其应用
US9145400B2 (en) Process for the preparation of form III of vilazodone hydrochloride
CN107556309B (zh) 多取代四氢萘啶类化合物的药物用途及其制备方法
JP2022530812A (ja) Wee1阻害剤化合物の結晶形及びその応用
CN104610412B (zh) 抗癌化合物、其制备方法以及制备抗癌药物的用途
WO2018014863A1 (zh) 一种硝基咪唑类化合物的晶型、盐型及其制备方法

Legal Events

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

Ref document number: 18805893

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2018805893

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2018805893

Country of ref document: EP

Effective date: 20200102