WO2022063305A1 - Crystal form of glucoside compounds and use thereof - Google Patents

Crystal form of glucoside compounds and use thereof Download PDF

Info

Publication number
WO2022063305A1
WO2022063305A1 PCT/CN2021/121043 CN2021121043W WO2022063305A1 WO 2022063305 A1 WO2022063305 A1 WO 2022063305A1 CN 2021121043 W CN2021121043 W CN 2021121043W WO 2022063305 A1 WO2022063305 A1 WO 2022063305A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
solvent
formula
amorphous
heptane
Prior art date
Application number
PCT/CN2021/121043
Other languages
French (fr)
Chinese (zh)
Inventor
李翼
甘露
于涛
姚婷
吴成德
陈曙辉
Original Assignee
南京明德新药研发有限公司
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 南京明德新药研发有限公司 filed Critical 南京明德新药研发有限公司
Priority to CN202180063705.6A priority Critical patent/CN116322675A/en
Publication of WO2022063305A1 publication Critical patent/WO2022063305A1/en

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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

Definitions

  • the invention relates to a crystal form of a glucoside compound and its application, as well as its application in the preparation of medicines for treating related diseases.
  • Obesity, diabetes and related metabolic disorders have become important risk factors threatening human health.
  • SGLTs Sodium-glucose cotransporters
  • the members mainly include SGLT1 and SGLT2.
  • Transmembrane transport of glucose in the kidney Specifically, SGLT1 is mainly distributed in the intestinal mucosal cells of the small intestine, and is also expressed in a small amount in the myocardium and kidney. Its function is mainly to regulate the intestinal absorption process of glucose.
  • SGLT2 is expressed at high levels in the kidney and is mainly responsible for the regulation of the renal glucose reuptake process, that is, the glucose in the urine can be actively attached to the renal tubular epithelial cells during glomerular filtration and transported into the cells by the SGLT2 protein for reuse. Because the glucose transport process mediated by SGLTs does not intervene in the metabolism of sugar, thus avoiding the occurrence of adverse reactions of hypoglycemia and reducing the risk of cardiovascular diseases, SGLTs have gradually become one of the ideal targets for the treatment of diabetes. In view of this, some SGLTs inhibitors, especially highly selective SGLT2 inhibitors, have been developed successively.
  • SGLT1 inhibitors have good development prospects as novel diabetes and obesity treatment drugs. But so far, research on SGLT1 inhibitors is still in the clinical stage, and no drug has been approved for marketing.
  • LX2761 an SGLT1 inhibitor developed by Lexicon, which only acts on the gastrointestinal tract, is undergoing Phase I clinical research for diabetes treatment (WO2014081660).
  • the present invention provides an amorphous form of the compound of formula (I), the X-ray powder diffraction (XRPD) pattern of which is shown in FIG. 1 .
  • a glass transition signal is observed at 74.9 ⁇ 3°C for the above-described amorphous differential scanning calorimetry curve.
  • the DSC spectrum of the above-mentioned amorphous is shown in FIG. 2 .
  • the above-mentioned amorphous thermogravimetric analysis curve has a weight loss of 2.56% at 150.0 ⁇ 3°C.
  • the above-mentioned amorphous TGA spectrum is shown in FIG. 3 .
  • the present invention also provides an amorphous preparation method of the compound of formula (I), which includes an anti-solvent addition method, a slow cooling method, a suspension stirring method, a slow volatilization method, a gas-solid infiltration method and a temperature cycle method.
  • the above-mentioned preparation method wherein, the anti-solvent addition method comprises:
  • the solvent is ethanol, acetone, tetrahydrofuran, acetonitrile, n-propyl acetate, anisole or 2-methyltetrahydrofuran;
  • the anti-solvent is H2O or n-heptane.
  • the above-mentioned preparation method wherein, the slow cooling method comprises:
  • Solvents were tert-butanol, isopropyl acetate, acetonitrile:water (v/v, 1:3) or 2-methyltetrahydrofuran:cyclohexane (v/v, 1:3).
  • the suspension stirring method comprises:
  • the solvent is methyl tert-butyl ether, ethyl acetate: n-heptane (v/v, 1:3), 2-methyltetrahydrofuran: cyclohexane (v/v, 1:3), methyl tert-butyl Ether: n-heptane (v/v, 1:7), tetrahydrofuran: n-heptane (v/v, 1:7), ethyl acetate: n-heptane (v/v, 1:7), 2-butane Ketone: n-heptane (v/v, 1:7), acetonitrile: water (v/v, 1:9), acetone: water (v/v, 1:12), n-heptane, water, tetrahydrofuran: n- Pentane (v/v, 1:7), N-methylpyrrolidone:water (v/v, 1:5), dimethyl
  • the above-mentioned preparation method, wherein, the slow volatilization method comprises:
  • the solvent is ethyl acetate, chloroform, methanol, acetone, acetonitrile or tetrahydrofuran.
  • the above-mentioned preparation method wherein, the gas-solid permeation method comprises:
  • the temperature cycling method comprises:
  • the solvent was n-heptane, methyl tert-butyl ether, 2-methyltetrahydrofuran:n-heptane (v/v, 1:3) or N-methylpyrrolidone:water (v/v, 1:5).
  • the 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, the embodiments formed by their combination with other chemical synthesis methods, and those skilled in the art.
  • Well-known equivalents, preferred embodiments include, but are not limited to, the examples of the present invention.
  • the structure of the compound of the present invention can be confirmed by conventional methods well known to those skilled in the art. If the present invention relates to the absolute configuration of the compound, the absolute configuration can be confirmed by conventional technical means in the art. For example, single crystal X-ray diffraction method (SXRD), the cultured single crystal is collected by Bruker D8 venture diffractometer, the light source is CuK ⁇ radiation, and the scanning mode is: After scanning and collecting relevant data, the crystal structure was further analyzed by the direct method (Shelxs97), and the absolute configuration could be confirmed.
  • SXRD single crystal X-ray diffraction method
  • the cultured single crystal is collected by Bruker D8 venture diffractometer
  • the light source is CuK ⁇ radiation
  • the scanning mode is: After scanning and collecting relevant data, the crystal structure was further analyzed by the direct method (Shelxs97), and the absolute configuration could be confirmed.
  • the solvent used in the present invention is commercially available.
  • DCM stands for dichloromethane
  • DMF stands for N,N-dimethylformamide
  • DMSO stands for dimethyl sulfoxide
  • EtOH stands for ethanol
  • MeOH stands for methanol
  • TFA trifluoroacetic acid
  • ATP stands for Adenosine triphosphate
  • HEPES 4-hydroxyethylpiperazineethanesulfonic acid
  • MgCl2 for magnesium dichloride
  • T3P for 1 -propylphosphoric anhydride.
  • the crystal form of the compound of the present invention has good stability and is easy to be formulated into medicine; the amorphous form of the compound of formula (I) has a significant inhibitory effect on the activities of human SGLT1 and SGLT2 transporters; The concentration and oral bioavailability are very low, and it has the pharmacokinetic properties of directly acting on the intestinal SGLT1; after a single administration of the compound of formula (I) amorphous, it can reduce the blood glucose level of mice after oral glucose in a dose-dependent manner .
  • Test Method Approximately 10 mg of sample was used for XRPD detection.
  • Test method Take a sample (about 1-5mg) and place it in an mDSC aluminum pan for testing. Under the condition of 50mL/min N2 , heat the sample from 0°C to 250°C at a heating rate of 3°C/min.
  • Thermogravimetric Analysis (Thermal Gravimetric Analyzer, TGA) method of the present invention
  • Test method Take a sample (about 1-5mg) and place it in a TGA aluminum pan for testing. Under the condition of 10mL/min N2 , at a heating rate of 10°C/min, heat the sample from room temperature to 350°C.
  • Fig. 1 is the XRPD spectrum of the amorphous Cu-K ⁇ radiation of the compound of formula (I);
  • Fig. 2 is the amorphous DSC spectrogram of the compound of formula (I);
  • Figure 3 is an amorphous TGA spectrum of the compound of formula (I).
  • 2-Methyltetrahydrofuran (12L) was added to the reaction kettle, and compound A (1200.00g, 5.45mol, 1.0eq), compound B (1650.00g, 8.18mol, 1.5eq) and triphenylphosphine (2070.00g) were slowly added in batches , 7.91 mol, 1.45 equiv), start stirring.
  • diisopropyl azodicarboxylate 1430.00g, 7.09mol, 1.30 equiv, 1.38L was slowly added dropwise to the reaction kettle, and the dropwise addition was completed, and the reaction kettle was heated to an internal temperature of 10 ⁇ 20°C, stirring for 16 hours.
  • the crude product was dissolved in dichloromethane (14 L), transferred to a separator, and washed with saturated sodium bicarbonate and water once each. The organic phase was separated and concentrated to dryness under reduced pressure to give a white solid.
  • the white solid was transferred to a three-necked flask, ethyl acetate (2.8 L) was added, and the mixture was stirred for 12 hours. After suction filtration, the filter cake was washed once with ethyl acetate (1.4 L), and the filter cake was collected and dried in a vacuum drying oven for 16 hours to obtain compound D.
  • the product was confirmed by LCMS, LC-MS (m/z) 304.0 [M+H] + .
  • reaction solution was transferred to a separator, and washed with water (8L*1), 0.1M hydrochloric acid (8L*1), 0.5M potassium carbonate (8L*2) and water (8L*2) in sequence.
  • the organic phase was collected and concentrated to dryness under reduced pressure to give crude product.
  • the crude product was dissolved in a mixed solvent of dioxane and n-heptane (1:5, 11.76L) and then transferred to the reaction kettle.
  • the temperature in the reaction kettle was controlled to be 15-25°C, and the mixture was stirred for about 42 hours until the solid was precipitated. Suction filtration, and the filter cake was rinsed with a mixed solvent of dioxane and n-heptane (1:5, 1.56L).
  • the filter cake was collected and dried in a vacuum drying oven for 16 hours to obtain compound F.
  • the product was confirmed by LCMS, LC-MS (m/z) 619.5 [M+H] + .
  • the filter cake was collected, and the above operation was repeated once to obtain 1476.25 g of filter cake.
  • the filter cake was dissolved in dichloromethane (7.4L), methanol (7.4L) and thiol silica gel (585.00g) were added in sequence, heated to 35-45°C, and stirred for 16 hours. After filtration, the filter residue was washed once with a mixed solvent of dichloromethane and methanol (1:1, 7.4 L). The filtrate was collected, and SiliaMets Thiol (585.00 g) was added, heated to 35-45°C, and stirred for 16 hours. After filtration, the filter residue was washed once with a mixed solvent of dichloromethane and methanol (1:1, 7.4 L).
  • HCl/EtOAc solution (4M, 10.5L) and compound H (700.00 g, 0.77 mol, 1.0 equiv) were added to the reaction kettle in turn, and stirring was started. Control the inner temperature of the reaction kettle to 15-25°C and stir for 2 hours. After the reaction was completed, ethyl acetate (7 L) was added to the reaction kettle to dilute the reaction solution, and the reaction solution was transferred to a rotary evaporator and concentrated to dryness under reduced pressure. Subsequently, ethyl acetate (7L*2) was added and concentrated to dryness under reduced pressure to obtain a crude product.
  • Test number Solvent, v/v test results 1 t-BuOH Amorphous 2 IPAc Amorphous 3 ACN/ H2O , 1:3 Amorphous 4 # 2-MeTHF/Cyclohexane, 1:3 Amorphous
  • Test number Solvent, v/v test results 1 EtOAc Amorphous 2 CHCl 3 Amorphous 3 MeOH Amorphous 4 Acetone Amorphous 5 ACN Amorphous 6 THF Amorphous
  • indicates that a clear solution was obtained, which was evaporated at room temperature to obtain a solid.
  • Test number Solvent, v/v test results 1 n-Heptane Amorphous 2 MTBE Amorphous 3 2-MeTHF/n-Heptane, 1:3 Amorphous 4 NMP/ H2O , 1:5 Amorphous
  • Example 3 Stability test of the compound of formula (I) amorphous
  • Influencing factor experiment the samples under high temperature (60°C) were placed in an open disposable petri dish, and the samples under high humidity (25°C/75%RH) were placed in an open flat weighing bottle, and then placed in different The blast drying oven and desiccator were investigated.
  • the illuminated samples are placed in a clean disposable petri dish, spread into a thin layer, and covered with a quartz glass cover.
  • the packaging method of the shading control sample is the same as that of the illuminated sample, but the outside of the disposable petri dish is covered with aluminum film.
  • the requirements of Q1B were placed under illumination conditions (visible light 5000 ⁇ 500 lux; ultraviolet light 90 ⁇ w/cm 2 ) for investigation;
  • Table 12 Summary of 37°C solubility of compound of formula (I) amorphous in different media
  • the amorphous compound of formula (I) is morphologically stable under manual grinding and tableting conditions (350MPa).
  • Isotope preparation Dilute the isotope stock solution [ 14 C]Methyl ⁇ -D-glucopyranosid with assay buffer to a working concentration of 6 ⁇ M.
  • % inhibition rate (mean value of high signal wells-signal value of sample wells)/(mean value of high signal wells-mean value of low signal wells)*100.
  • the amorphous compound of formula (I) has a significant inhibitory effect on the activities of human SGLT1 and SGLT2 transporters.
  • the vehicle in the intravenous injection group is 5% (w/v) hydroxypropyl ⁇ -cyclodextrin (HP- ⁇ -CD) aqueous solution.
  • amorphous compound of formula (I) is mixed with an appropriate amount of the vehicle in the intravenous injection group, 0.25 mg/mL is prepared. Clear solution; Oral group solvent is 0.5% (w/v) methylcellulose/0.02% Tween 80 aqueous solution, the compound of formula (I) amorphous is mixed with an appropriate amount of oral group solvent to prepare 0.5mg/mL Homogeneous suspension.
  • Cmax is the maximum concentration
  • F% is oral bioavailability
  • AUC po is oral exposure
  • Vd ss is volume of distribution
  • Cl is clearance
  • T 1/2 is half-life.
  • the experimental animals were housed in the animal room to acclimate to the environment for 1 week after arriving at the facility.
  • the compound of formula (I) amorphous and vehicle were orally administered respectively, followed by oral administration of 2 g/kg, 5 mL/kg glucose solution.
  • the animals were collected blood from the tail tip for blood glucose detection, and the glucose tolerance curve was drawn according to the time, and the area under the curve (AUC 1-120 minutes ) was calculated.
  • * means p ⁇ 0.05 relative to the vehicle control group
  • ** means p ⁇ 0.01 relative to the vehicle control group
  • *** means p ⁇ 0.001 relative to the vehicle control group
  • **** means p ⁇ 0.001 relative to the vehicle control group 0.0001.
  • the amorphous compound of formula (I) can dose-dependently reduce the blood glucose level of mice after oral administration of glucose.

Abstract

A crystal form (I) of glucoside compounds and the use thereof, and the use thereof in the preparation of a drug for treating related diseases.

Description

葡糖苷类化合物的晶型及其应用Crystal forms of glucosides and their applications
本申请主张如下优先权This application claims the following priority
CN202011032215.3,申请日:2020年09月27日。CN202011032215.3, application date: September 27, 2020.
技术领域technical field
本发明涉及一种葡糖苷类化合物的晶型及其应用,及其在制备治疗相关疾病的药物中的应用。The invention relates to a crystal form of a glucoside compound and its application, as well as its application in the preparation of medicines for treating related diseases.
背景技术Background technique
肥胖症、糖尿病及其引发的相关代谢紊乱疾病已成为威胁人类健康的重要危险因素。Obesity, diabetes and related metabolic disorders have become important risk factors threatening human health.
钠-葡萄糖共转运蛋白(sodium-glucose cotransporters,SGLTs)是一类在小肠黏膜和肾近曲小管中发现的葡萄糖转运蛋白家族,成员主要包括SGLT1和SGLT2两类,其功能是介导肠道和肾脏中葡萄糖的跨膜转运。具体而言,SGLT1主要分布于小肠的肠道粘膜细胞,在心肌和肾脏中也有少量表达,它的功能主要是调节葡萄糖的肠道吸收过程。SGLT2在肾脏中高水平表达,主要负责葡萄糖肾脏重摄取过程的调节,即尿液中的葡萄糖在经过肾小球过滤时可主动附着于肾小管上皮细胞并通过SGLT2蛋白转运进胞内被重新利用。由于SGLTs介导的葡萄糖转运过程不介入糖的代谢,从而避免了低血糖不良反应的发生,降低了引起心血管类疾病的风险,因此,SGLTs已逐渐成为治疗糖尿病的理想靶点之一。鉴于此,一些SGLTs抑制剂,尤其是高选择性的SGLT2抑制剂被相继开发。它们通过抑制SGLT2活性,特异性地抑制肾脏对葡萄糖的重吸收,从而增加葡萄糖在尿中的排泄,使糖尿病患者的血糖正常化。从2012年至今,已有多个SGLT2抑制剂先后被批准上市,成为治疗糖尿病的有效药物。Sodium-glucose cotransporters (SGLTs) are a family of glucose transporters found in the small intestinal mucosa and proximal renal tubules. The members mainly include SGLT1 and SGLT2. Transmembrane transport of glucose in the kidney. Specifically, SGLT1 is mainly distributed in the intestinal mucosal cells of the small intestine, and is also expressed in a small amount in the myocardium and kidney. Its function is mainly to regulate the intestinal absorption process of glucose. SGLT2 is expressed at high levels in the kidney and is mainly responsible for the regulation of the renal glucose reuptake process, that is, the glucose in the urine can be actively attached to the renal tubular epithelial cells during glomerular filtration and transported into the cells by the SGLT2 protein for reuse. Because the glucose transport process mediated by SGLTs does not intervene in the metabolism of sugar, thus avoiding the occurrence of adverse reactions of hypoglycemia and reducing the risk of cardiovascular diseases, SGLTs have gradually become one of the ideal targets for the treatment of diabetes. In view of this, some SGLTs inhibitors, especially highly selective SGLT2 inhibitors, have been developed successively. They specifically inhibit the renal reabsorption of glucose by inhibiting SGLT2 activity, thereby increasing the excretion of glucose in the urine and normalizing blood glucose in diabetic patients. Since 2012, a number of SGLT2 inhibitors have been approved for marketing and become effective drugs for the treatment of diabetes.
除了抑制SGLT2,近几年研究发现,适当抑制SGLT1能阻止肠道对葡萄糖的摄取,且不会导致明显的腹泻或者其他胃肠道反应。同时,通过抑制SGLT1能减少经肠道吸收入血的葡萄糖,进而增加肠道远端葡萄糖浓度,导致餐后GLP-1和PYY水平升高,从而发挥较良好的降糖作用,降低发生尿路感染和肾功能损伤等的风险。另外,通过控制肠道对葡萄糖吸收,还能降低食物中总能量的摄入,叠加GLP-1降低体重的作用,可以达到双重降低体重的目的。因此,开发SGLT1抑制剂已成为近年来糖尿病和肥胖症治疗的新方向。In addition to inhibiting SGLT2, studies in recent years have found that appropriate inhibition of SGLT1 can prevent intestinal uptake of glucose without causing significant diarrhea or other gastrointestinal reactions. At the same time, inhibiting SGLT1 can reduce the intestinal absorption of glucose into the blood, thereby increasing the glucose concentration in the distal part of the intestine, resulting in an increase in the postprandial GLP-1 and PYY levels, thus exerting a good hypoglycemic effect and reducing the occurrence of urinary tract. Risk of infection and kidney damage, etc. In addition, by controlling the intestinal absorption of glucose, it can also reduce the total energy intake in food, and superimpose the effect of GLP-1 to reduce body weight, which can achieve the purpose of double weight reduction. Therefore, the development of SGLT1 inhibitors has become a new direction for the treatment of diabetes and obesity in recent years.
综上所述,SGLT1抑制剂作为新型的糖尿病和肥胖症治疗药物有着良好的开发前景。但迄今为止,关于SGLT1抑制剂的研究仍处于临床阶段,还没有药物批准上市。目前,由Lexicon公司开发的只作用于胃肠道的SGLT1抑制剂LX2761正在针对糖尿病治疗开展临床I期研究(WO2014081660)。In conclusion, SGLT1 inhibitors have good development prospects as novel diabetes and obesity treatment drugs. But so far, research on SGLT1 inhibitors is still in the clinical stage, and no drug has been approved for marketing. Currently, LX2761, an SGLT1 inhibitor developed by Lexicon, which only acts on the gastrointestinal tract, is undergoing Phase I clinical research for diabetes treatment (WO2014081660).
Figure PCTCN2021121043-appb-000001
Figure PCTCN2021121043-appb-000001
发明内容SUMMARY OF THE INVENTION
本发明提供了式(I)化合物的无定形,其X射线粉末衍射(XRPD)图谱如图1所示。The present invention provides an amorphous form of the compound of formula (I), the X-ray powder diffraction (XRPD) pattern of which is shown in FIG. 1 .
Figure PCTCN2021121043-appb-000002
Figure PCTCN2021121043-appb-000002
在本发明的一些方案中,上述无定形的差示扫描量热曲线在74.9±3℃处可观察到一个玻璃态转化信号。In some aspects of the invention, a glass transition signal is observed at 74.9±3°C for the above-described amorphous differential scanning calorimetry curve.
在本发明的一些方案中,上述无定形的DSC图谱如图2所示。In some embodiments of the present invention, the DSC spectrum of the above-mentioned amorphous is shown in FIG. 2 .
在本发明的一些方案中,上述无定形的热重分析曲线在150.0±3℃时失重达2.56%。In some aspects of the present invention, the above-mentioned amorphous thermogravimetric analysis curve has a weight loss of 2.56% at 150.0±3°C.
在本发明的一些方案中,上述无定形的TGA图谱如图3所示。In some aspects of the present invention, the above-mentioned amorphous TGA spectrum is shown in FIG. 3 .
本发明还提供了式(Ⅰ)化合物的无定形的制备方法,包含反溶剂添加法、缓慢降温法、悬浮搅拌法、缓慢挥发法、气固渗透法和温度循环法。The present invention also provides an amorphous preparation method of the compound of formula (I), which includes an anti-solvent addition method, a slow cooling method, a suspension stirring method, a slow volatilization method, a gas-solid infiltration method and a temperature cycle method.
在本发明的一些方案中,上述的制备方法,其中,反溶剂添加法包含:In some aspects of the present invention, the above-mentioned preparation method, wherein, the anti-solvent addition method comprises:
1)将式(I)化合物加入到溶剂中形成澄清溶液;1) adding the compound of formula (I) to a solvent to form a clear solution;
2)向溶液中加入反溶剂;2) adding an anti-solvent to the solution;
其中,in,
溶剂为乙醇、丙酮、四氢呋喃、乙腈、乙酸正丙酯、苯甲醚或2-甲基四氢呋喃;The solvent is ethanol, acetone, tetrahydrofuran, acetonitrile, n-propyl acetate, anisole or 2-methyltetrahydrofuran;
反溶剂为H 2O或正庚烷。 The anti-solvent is H2O or n-heptane.
在本发明的一些方案中,上述的制备方法,其中,缓慢降温法包含:In some schemes of the present invention, the above-mentioned preparation method, wherein, the slow cooling method comprises:
1)50℃下,将式(I)化合物在溶剂中配制成澄清溶液;1) at 50 ° C, the compound of formula (I) is prepared into a clear solution in a solvent;
2)溶液以0.1℃/分钟从50℃降温至5℃;2) The solution is cooled from 50°C to 5°C at 0.1°C/min;
其中,in,
溶剂为叔丁醇、乙酸异丙酯、乙腈:水(v/v,1:3)或2-甲基四氢呋喃:环己烷(v/v,1:3)。Solvents were tert-butanol, isopropyl acetate, acetonitrile:water (v/v, 1:3) or 2-methyltetrahydrofuran:cyclohexane (v/v, 1:3).
在本发明的一些方案中,上述的制备方法,其中,悬浮搅拌法包含:In some aspects of the present invention, the above-mentioned preparation method, wherein, the suspension stirring method comprises:
1)将式(I)化合物加入到溶剂中形成悬浊液;1) adding the compound of formula (I) to a solvent to form a suspension;
2)悬浊液溶液用磁力搅拌转晶;2) The suspension solution is crystallized with magnetic stirring;
其中,in,
溶剂为甲基叔丁基醚、乙酸乙酯:正庚烷(v/v,1:3)、2-甲基四氢呋喃:环己烷(v/v,1:3)、甲基叔丁基醚:正庚烷(v/v,1:7)、四氢呋喃:正庚烷(v/v,1:7)、乙酸乙酯:正庚烷(v/v,1:7)、2-丁酮:正庚烷(v/v,1:7)、乙腈:水(v/v,1:9)、丙酮:水(v/v,1:12)、正庚烷、水、四氢呋喃:正戊烷(v/v,1:7)、N-甲基吡咯烷酮:水(v/v,1:5)、二甲基亚砜:水(v/v,1:5)或间二甲苯。The solvent is methyl tert-butyl ether, ethyl acetate: n-heptane (v/v, 1:3), 2-methyltetrahydrofuran: cyclohexane (v/v, 1:3), methyl tert-butyl Ether: n-heptane (v/v, 1:7), tetrahydrofuran: n-heptane (v/v, 1:7), ethyl acetate: n-heptane (v/v, 1:7), 2-butane Ketone: n-heptane (v/v, 1:7), acetonitrile: water (v/v, 1:9), acetone: water (v/v, 1:12), n-heptane, water, tetrahydrofuran: n- Pentane (v/v, 1:7), N-methylpyrrolidone:water (v/v, 1:5), dimethyl sulfoxide:water (v/v, 1:5) or m-xylene.
在本发明的一些方案中,上述的制备方法,其中,缓慢挥发法包含:In some aspects of the present invention, the above-mentioned preparation method, wherein, the slow volatilization method comprises:
1)将式(I)化合物溶解在溶剂中形成澄清溶液;1) dissolving the compound of formula (I) in a solvent to form a clear solution;
2)把澄清溶液用封口膜密封扎小孔后缓慢挥发析晶;2) Slowly volatilize and crystallize the clear solution after sealing the small holes with parafilm;
其中,in,
溶剂为乙酸乙酯、氯仿、甲醇、丙酮、乙腈或四氢呋喃。The solvent is ethyl acetate, chloroform, methanol, acetone, acetonitrile or tetrahydrofuran.
在本发明的一些方案中,上述的制备方法,其中,气固渗透法包含:In some aspects of the present invention, the above-mentioned preparation method, wherein, the gas-solid permeation method comprises:
1)将式(I)化合物置于小瓶中;1) placing the compound of formula (I) in a vial;
2)把上述小瓶敞口放置于水的氛围中,密封后室温下静置。2) The above-mentioned vial was opened and placed in an atmosphere of water, sealed and left to stand at room temperature.
在本发明的一些方案中,上述的制备方法,其中,温度循环法包含:In some aspects of the present invention, the above-mentioned preparation method, wherein, the temperature cycling method comprises:
1)50℃下,将式(I)化合物溶解在溶剂中形成悬浊液;1) at 50°C, the compound of formula (I) is dissolved in a solvent to form a suspension;
2)把悬浊液以0.1℃/分钟的速度按照50℃-5℃-50℃-5℃的程序进行升降温进行转晶;2) The temperature of the suspension is raised and lowered at a rate of 0.1°C/min according to the procedure of 50°C-5°C-50°C-5°C to transfer crystals;
其中,in,
溶剂为正庚烷、甲基叔丁基醚、2-甲基四氢呋喃:正庚烷(v/v,1:3)或N-甲基吡咯烷酮:水(v/v,1:5)。The solvent was n-heptane, methyl tert-butyl ether, 2-methyltetrahydrofuran:n-heptane (v/v, 1:3) or N-methylpyrrolidone:water (v/v, 1:5).
定义和说明Definition and Explanation
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。Unless otherwise specified, the following terms and phrases used herein are intended to have the following meanings. A particular phrase or term should not be considered indeterminate or unclear without a specific definition, but should be understood in its ordinary meaning. When a trade name appears herein, it is intended to refer to its corresponding commercial product or its active ingredient.
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。The 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, the embodiments formed by their combination with other chemical synthesis methods, and those skilled in the art. Well-known equivalents, preferred embodiments include, but are not limited to, the examples of the present invention.
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。The chemical reactions of specific embodiments of the present invention are carried out in suitable solvents suitable for the chemical changes of the present invention and their required reagents and materials. In order to obtain the compounds of the present invention, it is sometimes necessary for those skilled in the art to modify or select the synthetic steps or reaction schemes on the basis of the existing embodiments.
本发明的化合物可以通过本领域技术人员所熟知的常规方法来确认结构,如果本发明涉及化合物的绝对构型,则该绝对构型可以通过本领域常规技术手段予以确证。例如单晶X射线衍射法(SXRD),把培养出的单晶用Bruker D8 venture衍射仪收集衍射强度数据,光源为CuKα辐射,扫描方式:
Figure PCTCN2021121043-appb-000003
扫描,收集相关数据后,进一步采用直接法(Shelxs97)解析晶体结构,便可以确证绝对构型。
The structure of the compound of the present invention can be confirmed by conventional methods well known to those skilled in the art. If the present invention relates to the absolute configuration of the compound, the absolute configuration can be confirmed by conventional technical means in the art. For example, single crystal X-ray diffraction method (SXRD), the cultured single crystal is collected by Bruker D8 venture diffractometer, the light source is CuKα radiation, and the scanning mode is:
Figure PCTCN2021121043-appb-000003
After scanning and collecting relevant data, the crystal structure was further analyzed by the direct method (Shelxs97), and the absolute configuration could be confirmed.
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。The present invention will be specifically described below through examples, which do not imply any limitation to the present invention.
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。All solvents used in the present invention are commercially available and used without further purification.
本发明所使用的溶剂可经市售获得。本发明采用下述缩略词:DCM代表二氯甲烷;DMF代表N,N-二甲基甲酰胺;DMSO代表二甲亚砜;EtOH代表乙醇;MeOH代表甲醇;TFA代表三氟乙酸;ATP代表三磷酸腺苷;HEPES代表4-羟乙基哌嗪乙磺酸;MgCl 2代表二氯化镁;T 3P代表1-丙基磷酸酐。 The solvent used in the present invention is commercially available. The following abbreviations are used in the present invention: DCM stands for dichloromethane; DMF stands for N,N-dimethylformamide; DMSO stands for dimethyl sulfoxide; EtOH stands for ethanol; MeOH stands for methanol; TFA stands for trifluoroacetic acid; ATP stands for Adenosine triphosphate; HEPES for 4-hydroxyethylpiperazineethanesulfonic acid; MgCl2 for magnesium dichloride; T3P for 1 -propylphosphoric anhydride.
技术效果technical effect
本发明化合物的晶型稳定性好,易于成药;式(I)化合物无定形对人源SGLT1和SGLT2转运体活性有显著抑制作用;式(I)化合物无定形口服给药后,在体内血药浓度和口服生物利用度均很低,具有直接作用于肠道SGLT1的药代动力学性质;式(I)化合物无定形单次给药后可呈剂量依赖性降低小鼠葡萄糖口服后的血糖水平。The crystal form of the compound of the present invention has good stability and is easy to be formulated into medicine; the amorphous form of the compound of formula (I) has a significant inhibitory effect on the activities of human SGLT1 and SGLT2 transporters; The concentration and oral bioavailability are very low, and it has the pharmacokinetic properties of directly acting on the intestinal SGLT1; after a single administration of the compound of formula (I) amorphous, it can reduce the blood glucose level of mice after oral glucose in a dose-dependent manner .
本发明X-射线粉末衍射(X-ray powder diffractometer,XRPD)方法X-ray powder diffraction (X-ray powder diffractometer, XRPD) method of the present invention
仪器型号:PANalytical公司的X-射线衍射仪Instrument model: X-ray diffractometer from PANalytical
测试方法:大约10mg样品用于XRPD检测。Test Method: Approximately 10 mg of sample was used for XRPD detection.
详细的XRPD参数如表1所示:The detailed XRPD parameters are shown in Table 1:
表1Table 1
Figure PCTCN2021121043-appb-000004
Figure PCTCN2021121043-appb-000004
本发明调制差示扫描量热(Modulated Differential Scanning Calorimetry,mDSC)方法Modulated Differential Scanning Calorimetry (mDSC) method of the present invention
仪器型号:TA Discovery DSC 2500差示扫描量热仪Instrument model: TA Discovery DSC 2500 Differential Scanning Calorimeter
测试方法:取样品(约1-5mg)置于mDSC铝盘内进行测试,在50mL/min N 2条件下,以3℃/min的升温速率加热样品从0℃到250℃。 Test method: Take a sample (about 1-5mg) and place it in an mDSC aluminum pan for testing. Under the condition of 50mL/min N2 , heat the sample from 0°C to 250°C at a heating rate of 3°C/min.
详细的DSC参数如表2所示:The detailed DSC parameters are shown in Table 2:
表2Table 2
参数parameter 设定值set value
方法method 调制升温modulated warming
样品盘sample tray 铝盘,压盖Aluminium pan, gland
温度范围temperature range 0℃-250℃0℃-250℃
扫描速率(℃/分钟)Scan rate (℃/min) 33
保护气体Protective gas 氮气nitrogen
.
本发明热重分析(Thermal Gravimetric Analyzer,TGA)方法Thermogravimetric Analysis (Thermal Gravimetric Analyzer, TGA) method of the present invention
仪器型号:TA Q5000/Discovery TGA 5500热重分析仪Instrument Model: TA Q5000/Discovery TGA 5500 Thermogravimetric Analyzer
测试方法:取样品(约1-5mg)置于TGA铝盘内进行测试,在10mL/min N 2条件下,以10℃/min的升温速率,加热样品从室温到350℃。 Test method: Take a sample (about 1-5mg) and place it in a TGA aluminum pan for testing. Under the condition of 10mL/min N2 , at a heating rate of 10°C/min, heat the sample from room temperature to 350°C.
详细的TGA参数如表3所示:The detailed TGA parameters are shown in Table 3:
表3table 3
参数parameter 设定值set value
方法method 调制升温modulated warming
样品盘sample tray 铝盘,压盖Aluminium pan, gland
温度范围temperature range 0℃-250℃0℃-250℃
扫描速率(℃/分钟)Scan rate (℃/min) 33
保护气体Protective gas 氮气nitrogen
.
附图说明Description of drawings
图1为式(I)化合物无定形的Cu-Kα辐射的XRPD谱图;Fig. 1 is the XRPD spectrum of the amorphous Cu-Kα radiation of the compound of formula (I);
图2为式(I)化合物无定形的DSC谱图;Fig. 2 is the amorphous DSC spectrogram of the compound of formula (I);
图3为式(I)化合物无定形的TGA谱图。Figure 3 is an amorphous TGA spectrum of the compound of formula (I).
具体实施方式detailed description
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。In order to better understand the content of the present invention, further description will be given below in conjunction with specific embodiments, but the specific embodiments do not limit the content of the present invention.
实施例1:式(I)化合物的制备Example 1: Preparation of compounds of formula (I)
Figure PCTCN2021121043-appb-000005
Figure PCTCN2021121043-appb-000005
步骤1:化合物C的合成Step 1: Synthesis of Compound C
将2-甲基四氢呋喃(12L)加入反应釜,缓慢分批加入化合物A(1200.00g,5.45mol,1.0当量),化合物B(1650.00g,8.18mol,1.5当量)和三苯基膦(2070.00g,7.91mol,1.45当量),开启搅拌。在0-5℃下,将偶氮二甲酸二异丙酯(1430.00g,7.09mol,1.30当量,1.38L)缓慢滴加到反应釜中,滴加完毕,将反应釜升温至内温10~20℃,搅拌16小时。反应完毕后,缓慢分批加入氯化镁(4.14kg),搅拌2小时。再向反应釜中缓慢加入2-甲基四氢呋喃(12L),搅拌稀释0.5小时。过滤,收集滤液,向滤液中加入水(12L),搅拌5分钟后静置分层,分离有机相和水相,水相用乙酸乙酯(6L)萃取。合并有机相, 减压浓缩至干,得到粗品。向粗品中加入乙醇(12L),控制反应釜内温为15-25℃搅拌16小时。抽滤,滤饼用乙醇(1.2L)洗涤,收集滤饼,放入真空干燥箱中干燥16小时,得到化合物C。产物经LCMS确证,LC-MS(m/z)304.2[M+H-Boc] +2-Methyltetrahydrofuran (12L) was added to the reaction kettle, and compound A (1200.00g, 5.45mol, 1.0eq), compound B (1650.00g, 8.18mol, 1.5eq) and triphenylphosphine (2070.00g) were slowly added in batches , 7.91 mol, 1.45 equiv), start stirring. At 0-5°C, diisopropyl azodicarboxylate (1430.00g, 7.09mol, 1.30 equiv, 1.38L) was slowly added dropwise to the reaction kettle, and the dropwise addition was completed, and the reaction kettle was heated to an internal temperature of 10~ 20°C, stirring for 16 hours. After the completion of the reaction, magnesium chloride (4.14 kg) was slowly added in portions and stirred for 2 hours. Then, 2-methyltetrahydrofuran (12 L) was slowly added to the reaction kettle, and the mixture was stirred and diluted for 0.5 hour. Filtration, collecting the filtrate, adding water (12 L) to the filtrate, stirring for 5 minutes and then standing to separate the layers, separating the organic phase and the aqueous phase, and extracting the aqueous phase with ethyl acetate (6 L). The organic phases were combined and concentrated to dryness under reduced pressure to give crude product. Ethanol (12 L) was added to the crude product, and the inner temperature of the reaction kettle was controlled to be 15-25° C. and stirred for 16 hours. After suction filtration, the filter cake was washed with ethanol (1.2 L), the filter cake was collected, and dried in a vacuum drying oven for 16 hours to obtain compound C. The product was confirmed by LCMS, LC-MS (m/z) 304.2 [M+H-Boc] + .
步骤2:化合物D的合成Step 2: Synthesis of Compound D
将二氯甲烷(14L)加入反应釜,缓慢分批加入化合物C(1400.00g,3.47mol,1.0当量),开启搅拌。向反应釜中缓慢滴加三氟乙酸(2.1L,28.36mol,8.17当量),滴加完毕,保持反应釜内温为15~25℃,搅拌3小时。反应完毕后,将反应液转移至旋转蒸发仪中,减压浓缩至干,除去二氯甲烷和大部分三氟乙酸,得到粗品。粗品用二氯甲烷(14L)溶解后,转移至分液器中,分别用饱和碳酸氢钠和水各洗涤一次。分出有机相,减压浓缩至干,得到白色固体。将白色固体转移至三口瓶中,加入乙酸乙酯(2.8L),搅拌12小时。抽滤,滤饼用乙酸乙酯(1.4L)洗涤一次,收集滤饼,放入真空干燥箱中干燥16小时,得到化合物D。产物经LCMS确证,LC-MS(m/z)304.0[M+H] +Dichloromethane (14 L) was added to the reaction kettle, compound C (1400.00 g, 3.47 mol, 1.0 equiv) was slowly added in batches, and stirring was started. Trifluoroacetic acid (2.1 L, 28.36 mol, 8.17 equiv) was slowly added dropwise to the reaction kettle, the dropwise addition was completed, and the temperature in the reaction kettle was kept at 15-25° C. and stirred for 3 hours. After the completion of the reaction, the reaction solution was transferred to a rotary evaporator and concentrated to dryness under reduced pressure to remove dichloromethane and most of trifluoroacetic acid to obtain a crude product. The crude product was dissolved in dichloromethane (14 L), transferred to a separator, and washed with saturated sodium bicarbonate and water once each. The organic phase was separated and concentrated to dryness under reduced pressure to give a white solid. The white solid was transferred to a three-necked flask, ethyl acetate (2.8 L) was added, and the mixture was stirred for 12 hours. After suction filtration, the filter cake was washed once with ethyl acetate (1.4 L), and the filter cake was collected and dried in a vacuum drying oven for 16 hours to obtain compound D. The product was confirmed by LCMS, LC-MS (m/z) 304.0 [M+H] + .
步骤3:化合物F的合成Step 3: Synthesis of Compound F
将二氯甲烷(8L)加入反应釜中,缓慢分批加入化合物D(784.37g,2.59mol,1.0当量)和化合物E(863.00g,2.59mol,1.0当量),开启搅拌。再缓慢加入三乙胺(800.89g,7.91mol,3.06当量,1.10L),控制反应釜内温为10~20℃,搅拌15分钟。随后向反应釜中缓慢滴加T 3P(2.35L,3.96mol,1.53当量),滴加完毕,控制反应釜内温为10~20℃,搅拌16小时。反应结束后,将反应液转移至分液器中,依次用水(8L*1),0.1M盐酸(8L*1),0.5M碳酸钾(8L*2)和水(8L*2)充分洗涤。收集有机相,减压浓缩至干,得到粗品。粗品用二氧六环与正庚烷的混合溶剂(1:5,11.76L)溶解后转移至反应釜中,控制反应釜内温为15-25℃,搅拌约42小时至固体析出。抽滤,滤饼用二氧六环与正庚烷混合溶剂(1:5,1.56L)淋洗。收集滤饼,放入真空干燥箱中干燥16小时,得到化合物F。产物经LCMS确证,LC-MS(m/z)619.5[M+H] +Dichloromethane (8 L) was added to the reaction kettle, compound D (784.37 g, 2.59 mol, 1.0 equiv) and compound E (863.00 g, 2.59 mol, 1.0 equiv) were slowly added in batches, and stirring was started. Then slowly add triethylamine (800.89 g, 7.91 mol, 3.06 equiv, 1.10 L), control the inner temperature of the reaction kettle to be 10-20 °C, and stir for 15 minutes. Subsequently, T 3 P (2.35 L, 3.96 mol, 1.53 equiv.) was slowly added dropwise to the reaction kettle, and the dropwise addition was completed. After the reaction, the reaction solution was transferred to a separator, and washed with water (8L*1), 0.1M hydrochloric acid (8L*1), 0.5M potassium carbonate (8L*2) and water (8L*2) in sequence. The organic phase was collected and concentrated to dryness under reduced pressure to give crude product. The crude product was dissolved in a mixed solvent of dioxane and n-heptane (1:5, 11.76L) and then transferred to the reaction kettle. The temperature in the reaction kettle was controlled to be 15-25°C, and the mixture was stirred for about 42 hours until the solid was precipitated. Suction filtration, and the filter cake was rinsed with a mixed solvent of dioxane and n-heptane (1:5, 1.56L). The filter cake was collected and dried in a vacuum drying oven for 16 hours to obtain compound F. The product was confirmed by LCMS, LC-MS (m/z) 619.5 [M+H] + .
步骤4:化合物H的合成Step 4: Synthesis of Compound H
将二氧六环(6L),水(1.5L),化合物G(750.00g,1.49mol,1.0当量),化合物F(1105.75g,1.19mol,1.2当量),Pd(PPh 3) 4(86.07g,0.08mol,0.05当量)和碳酸钾(411.81g,2.98mol,2.0当量)依次加入反应釜中,开启搅拌,通入氮气流。将反应釜内温升至45~55℃,搅拌16小时。反应完毕后,将反应液转移至旋转蒸发仪中,减压浓缩至干,得到粗品。向粗品中加入乙酸乙酯(7.5L)溶解,转移至分液器中,加入水(7.5L),搅拌5分钟后静置分层。收集有机相,水相用乙酸乙酯(7.5L)萃取。合并有机相,然后用水(7.5L)洗涤一次。收集有机相,减压浓缩至干,得到粗品。向粗品中加入甲醇(15L),加热至55~65℃溶解澄清后降温至15~25℃搅拌16小时。过滤,滤饼用甲醇(15L)洗涤一次。收集滤饼,重复一次上述操作,得到滤饼1476.25g。滤饼用二氯甲烷(7.4L)溶解,再依次加入甲醇(7.4L)和硫醇硅胶(585.00g),加热至35~45℃,搅拌16小时。过滤,滤渣用二氯甲烷与甲醇的混合溶剂(1:1,7.4L)洗涤一次。收集滤液,再加入SiliaMets Thiol(585.00g),加热至35~45℃,搅拌16小时。过滤,滤渣用二氯甲烷与甲醇的混合溶剂(1:1,7.4L)洗涤一次。收集滤液,再加入SiliaMets Thiol(585.00g),加热至35~45℃,搅拌16小时。过滤,滤渣用二氯甲烷与甲醇的混合溶剂(1:1,7.4L)洗涤一次。将滤液转移至旋转蒸发仪中,减压浓缩至干,收集产品,放入真空干燥箱中干燥16小时,得到化合物H。产物经LCMS确证,LC-MS(m/z)915[M+H] +Dioxane (6L), water (1.5L), compound G (750.00 g, 1.49 mol, 1.0 equiv), compound F (1105.75 g, 1.19 mol, 1.2 equiv), Pd(PPh 3 ) 4 (86.07 g) , 0.08mol, 0.05 equiv) and potassium carbonate (411.81 g, 2.98 mol, 2.0 equiv) were sequentially added to the reaction kettle, the stirring was turned on, and nitrogen flow was introduced. The temperature in the reactor was raised to 45-55°C and stirred for 16 hours. After the completion of the reaction, the reaction solution was transferred to a rotary evaporator and concentrated to dryness under reduced pressure to obtain a crude product. Ethyl acetate (7.5 L) was added to the crude product to dissolve, transferred to a separator, water (7.5 L) was added, and the mixture was stirred for 5 minutes and then left to stand for separation. The organic phase was collected and the aqueous phase was extracted with ethyl acetate (7.5 L). The organic phases were combined and washed once with water (7.5 L). The organic phase was collected and concentrated to dryness under reduced pressure to give crude product. Methanol (15 L) was added to the crude product, heated to 55-65° C. to dissolve and clarify, then cooled to 15-25° C. and stirred for 16 hours. Filter and wash the filter cake once with methanol (15 L). The filter cake was collected, and the above operation was repeated once to obtain 1476.25 g of filter cake. The filter cake was dissolved in dichloromethane (7.4L), methanol (7.4L) and thiol silica gel (585.00g) were added in sequence, heated to 35-45°C, and stirred for 16 hours. After filtration, the filter residue was washed once with a mixed solvent of dichloromethane and methanol (1:1, 7.4 L). The filtrate was collected, and SiliaMets Thiol (585.00 g) was added, heated to 35-45°C, and stirred for 16 hours. After filtration, the filter residue was washed once with a mixed solvent of dichloromethane and methanol (1:1, 7.4 L). The filtrate was collected, and SiliaMets Thiol (585.00 g) was added, heated to 35-45°C, and stirred for 16 hours. After filtration, the filter residue was washed once with a mixed solvent of dichloromethane and methanol (1:1, 7.4L). The filtrate was transferred to a rotary evaporator, concentrated to dryness under reduced pressure, the product was collected, and dried in a vacuum drying oven for 16 hours to obtain compound H. The product was confirmed by LCMS, LC-MS (m/z) 915 [M+H] + .
步骤5:化合物J的合成Step 5: Synthesis of Compound J
将HCl/EtOAc溶液(4M,10.5L),化合物H(700.00g,0.77mol,1.0当量)依次加入反应釜中,开启搅拌。控制反应釜内温15~25℃搅拌2小时。反应完毕后,向反应釜中加入乙酸乙酯(7L)稀释反应液,并将反应液转移至旋转蒸发仪中,减压浓缩至干。随后再加入乙酸乙酯(7L*2)继续减压浓缩至干,得到粗品。向粗品中加入乙酸乙酯(10.5L),在15~25℃下搅拌16小时。过滤,滤饼用乙酸乙酯(1.05L)洗涤一次。收集滤饼,放入真空干燥箱中干燥16小时,得到化合物J。产物经LCMS确证,LC-MS(m/z)815[M+H] +HCl/EtOAc solution (4M, 10.5L) and compound H (700.00 g, 0.77 mol, 1.0 equiv) were added to the reaction kettle in turn, and stirring was started. Control the inner temperature of the reaction kettle to 15-25°C and stir for 2 hours. After the reaction was completed, ethyl acetate (7 L) was added to the reaction kettle to dilute the reaction solution, and the reaction solution was transferred to a rotary evaporator and concentrated to dryness under reduced pressure. Subsequently, ethyl acetate (7L*2) was added and concentrated to dryness under reduced pressure to obtain a crude product. Ethyl acetate (10.5 L) was added to the crude product, and the mixture was stirred at 15 to 25°C for 16 hours. Filter and wash the filter cake once with ethyl acetate (1.05 L). The filter cake was collected and dried in a vacuum oven for 16 hours to obtain compound J. The product was confirmed by LCMS, LC-MS (m/z) 815 [M+H] + .
步骤6:式(I)化合物的合成Step 6: Synthesis of Compound of Formula (I)
将甲醇(5.8L),化合物J(580.00g,0.71mol,1.0当量)依次加入反应釜,开启搅拌。随后加入25%NaOMe甲醇溶液(290mL),室温下搅拌3小时。反应完毕后,将反应液转移至旋转蒸发仪中,减压浓缩至干,得到粗品。向粗品中加入水(5.6L),在10~20℃下搅拌2小时。过滤,滤饼用水(1.12L)洗涤两次。收集滤饼,再向滤饼中加入水(5.6L),在10~20℃下搅拌16小时。过滤,滤饼用水(1.12L)洗涤两次。收集滤饼,放入真空干燥箱中干燥144小时,得到式(I)化合物。 1H NMR(400MHz,CD 3OD)δppm 1.09(t,J=7.50Hz,3H),1.73(br d,J=17.26Hz,2H),1.92(br d,J=12.51Hz,2H),2.14(s,3H),2.40-2.55(m,2H),2.61(q,J=7.42Hz,2H),2.68-2.85(m,2H),3.35-3.48(m,5H),3.53(br s,1H),3.65-3.88(m,2H),3.92-4.01(m,2H),4.13(d,J=9.13Hz,1H),4.39(d,J=9.51Hz,1H),4.56(br s,1H),6.85(d,J=8.38Hz,2H),7.05(d,J=8.38Hz,2H),7.10-7.31(m,5H)。 Methanol (5.8 L) and compound J (580.00 g, 0.71 mol, 1.0 equiv) were sequentially added to the reaction kettle, and stirring was started. 25% NaOMe in methanol (290 mL) was then added and stirred at room temperature for 3 hours. After the completion of the reaction, the reaction solution was transferred to a rotary evaporator and concentrated to dryness under reduced pressure to obtain a crude product. Water (5.6 L) was added to the crude product, and the mixture was stirred at 10 to 20°C for 2 hours. Filter and wash the filter cake twice with water (1.12 L). The filter cake was collected, water (5.6 L) was added to the filter cake, and the mixture was stirred at 10 to 20° C. for 16 hours. Filter and wash the filter cake twice with water (1.12 L). The filter cake was collected and dried in a vacuum drying oven for 144 hours to obtain the compound of formula (I). 1 H NMR (400 MHz, CD 3 OD) δppm 1.09 (t, J=7.50 Hz, 3H), 1.73 (br d, J=17.26 Hz, 2H), 1.92 (br d, J=12.51 Hz, 2H), 2.14 (s, 3H), 2.40-2.55(m, 2H), 2.61(q, J=7.42Hz, 2H), 2.68-2.85(m, 2H), 3.35-3.48(m, 5H), 3.53(br s, 1H), 3.65-3.88(m, 2H), 3.92-4.01(m, 2H), 4.13(d, J=9.13Hz, 1H), 4.39(d, J=9.51Hz, 1H), 4.56(br s, 1H), 6.85 (d, J=8.38Hz, 2H), 7.05 (d, J=8.38Hz, 2H), 7.10-7.31 (m, 5H).
实施例2:式(I)化合物无定形的制备Example 2: Preparation of Amorphous Compound of Formula (I)
表4:试验中选用溶剂的中英文名称对照表Table 4: Comparison table of Chinese and English names of solvents used in the test
英文English 中文Chinese 英文English 中文Chinese
MeOHMeOH 甲醇methanol MTBEMTBE 甲基叔丁基醚Methyl tert-butyl ether
EtOHEtOH 乙醇Ethanol AnisoleAnisole 苯甲醚anisole
MEKMEK 2-丁酮2-Butanone n-Heptanen-Heptane 正庚烷n-heptane
AcetoneAcetone 丙酮acetone TolueneToluene 甲苯Toluene
ACNACN 乙腈Acetonitrile DCMDCM 二氯甲烷Dichloromethane
IPAcIPAc 乙酸异丙酯isopropyl acetate DMSODMSO 二甲基亚砜dimethyl sulfoxide
THFTHF 四氢呋喃tetrahydrofuran H 2O H 2 O water
2-MeTHF2-MeTHF 2-甲基四氢呋喃2-Methyltetrahydrofuran NMPNMP N-甲基吡咯烷酮N-Methylpyrrolidone
CyclohexaneCyclohexane 环己烷Cyclohexane m-Xylenem-Xylene 间二甲苯m-xylene
n-Pentanen-Pentane 正戊烷n-pentane CHCl 3 CHCl 3 氯仿Chloroform
t-BuOHt-BuOH 叔丁醇tert-Butanol EtOAcEtOAc 乙酸乙酯Ethyl acetate
n-Propyl acetaten-Propyl acetate 乙酸丙酯Propyl acetate      
1.反溶剂添加法:1. Anti-solvent addition method:
称取约15~20毫克的式(I)化合物,25℃下溶解在正溶剂中配制澄清溶液,缓慢滴加相应反溶剂至上述溶液中直至固体出现,将得到的固体分离测试XRPD。结果如表5所示:Weigh about 15-20 mg of the compound of formula (I), dissolve it in a positive solvent at 25° C. to prepare a clear solution, slowly add the corresponding anti-solvent dropwise to the above solution until a solid appears, and separate the obtained solid for XRPD testing. The results are shown in Table 5:
表5:反溶剂添加试验Table 5: Antisolvent Addition Test
Figure PCTCN2021121043-appb-000006
Figure PCTCN2021121043-appb-000006
*:表示加入反溶剂后得到澄清溶液,转至-20℃搅拌直至固体出现。*: Indicates that a clear solution is obtained after adding anti-solvent, turn to -20°C and stir until solid appears.
2.缓慢降温法:2. Slow cooling method:
取约20毫克的式(I)化合物,在50℃条件下溶解在溶剂中配制澄清溶液,以0.1℃/分钟的速度按照50℃-5℃的程序进行降温,收集析出的固体并进行XRPD测试。结果如表6所示:Take about 20 mg of the compound of formula (I), dissolve it in a solvent at 50°C to prepare a clear solution, cool down at a rate of 0.1°C/min according to the procedure of 50°C-5°C, collect the precipitated solid and conduct XRPD test . The results are shown in Table 6:
表6:缓慢降温试验Table 6: Slow cooling test
试验编号Test number 溶剂,v/vSolvent, v/v 试验结果test results
11 t-BuOHt-BuOH 无定形Amorphous
22 IPAcIPAc 无定形Amorphous
33 ACN/H 2O,1:3 ACN/ H2O , 1:3 无定形Amorphous
4 # 4 # 2-MeTHF/Cyclohexane,1:32-MeTHF/Cyclohexane, 1:3 无定形Amorphous
#:表示降至5℃后得澄清溶液,转至-20℃放置,仍澄清,转至室温挥发析出固体。#: Indicates that a clear solution is obtained after the temperature drops to 5°C, and the solution is placed at -20°C, and the solution is still clear, and the solid is volatilized and precipitated at room temperature.
3.悬浮搅拌法:3. Suspension stirring method:
称量约15~20毫克的式(I)化合物,在不同溶剂中配制25℃或50℃条件下的悬浊液,磁力搅拌约6~8天后,离心收集固体并进行XRPD测试。结果如表7所示:Weigh about 15-20 mg of the compound of formula (I), prepare suspensions at 25°C or 50°C in different solvents, stir magnetically for about 6-8 days, collect the solid by centrifugation and conduct XRPD test. The results are shown in Table 7:
表7:悬浮搅拌试验Table 7: Suspension Stirring Test
Figure PCTCN2021121043-appb-000007
Figure PCTCN2021121043-appb-000007
Figure PCTCN2021121043-appb-000008
Figure PCTCN2021121043-appb-000008
4.缓慢挥发法:4. Slow volatilization method:
称取约15~20毫克的式(I)化合物,在25℃下溶解在溶剂中配制澄清溶液,用封口膜密封扎4-5个小孔后缓慢挥发析晶,收集所得固体并进行XRPD测试。结果如表8所示:Weigh about 15-20 mg of the compound of formula (I), dissolve it in a solvent at 25°C to prepare a clear solution, seal 4-5 small holes with a parafilm, slowly volatilize and crystallize, collect the obtained solid and conduct XRPD test . The results are shown in Table 8:
表8:缓慢挥发试验Table 8: Slow Evaporation Test
试验编号Test number 溶剂,v/vSolvent, v/v 试验结果test results
11 EtOAcEtOAc 无定形Amorphous
22 CHCl 3 CHCl 3 无定形Amorphous
33 MeOHMeOH 无定形Amorphous
44 AcetoneAcetone 无定形Amorphous
55 ACNACN 无定形Amorphous
66 THFTHF 无定形Amorphous
5.气固渗透法:5. Gas-solid infiltration method:
称量约20毫克的式(I)化合物置于3毫升小瓶中,另取20毫升小瓶并向其中加入相应溶剂,将3毫升小瓶敞口置于20毫升小瓶中,密封后室温下静置。收集固体并进行XRPD测试。结果如表9所示:About 20 mg of the compound of formula (I) was weighed into a 3 ml vial, another 20 ml vial was taken and the corresponding solvent was added to it, the 3 ml vial was opened and placed in a 20 ml vial, sealed and kept at room temperature. The solids were collected and tested by XRPD. The results are shown in Table 9:
表9:气固渗透试验Table 9: Gas-Solid Penetration Test
样品编号Sample serial number 溶剂solvent 试验结果test results
11 H 2O H 2 O 无定形Amorphous
2^2^ DCMDCM 无定形Amorphous
3^3^ EtOHEtOH 无定形Amorphous
4^4^ TolueneToluene 无定形Amorphous
5^5^ THFTHF 无定形Amorphous
^:表示得澄清溶液,转至室温挥发得到固体。^: indicates that a clear solution was obtained, which was evaporated at room temperature to obtain a solid.
6.温度循环法:6. Temperature cycle method:
称取约20毫克的式(I)化合物,在50℃条件下加至溶剂中配制悬浊液,以0.1℃/分钟的速度按照50℃-5℃-50℃-5℃的程序进行升降温,收集所得固体并进行XRPD测试。结果如表10所示:Weigh about 20 mg of the compound of formula (I), add it to the solvent at 50°C to prepare a suspension, and ramp up and down the temperature at a rate of 0.1°C/min according to the program of 50°C-5°C-50°C-5°C , the resulting solid was collected and tested by XRPD. The results are shown in Table 10:
表10:温度循环试验Table 10: Temperature Cycling Test
试验编号Test number 溶剂,v/vSolvent, v/v 试验结果test results
11 n-Heptanen-Heptane 无定形Amorphous
22 MTBEMTBE 无定形Amorphous
33 2-MeTHF/n-Heptane,1:32-MeTHF/n-Heptane, 1:3 无定形Amorphous
44 NMP/H 2O,1:5 NMP/ H2O , 1:5 无定形Amorphous
实施例3:式(I)化合物无定形的稳定性实验Example 3: Stability test of the compound of formula (I) amorphous
实验目的:Purpose:
对式(I)化合物无定形样品进行影响因素(高温、高湿及光照),长期条件(25℃/60%RH)和加速条件下(40℃/75%RH)稳定性的考察,评估样品的固体稳定性。The influence factors (high temperature, high humidity and light), long-term conditions (25°C/60%RH) and accelerated conditions (40°C/75%RH) stability of the amorphous samples of the compound of formula (I) were investigated, and the samples were evaluated solid stability.
实验方法:experimental method:
影响因素实验:高温条件(60℃)样品置于敞口的一次性培养皿中,高湿条件(25℃/75%RH)样品置于敞口的扁形称量瓶中,然后分别放入不同的鼓风干燥箱和保干器中考察。光照样品放入干净的一次性培养皿中,铺成薄层,盖上石英玻璃盖,遮光对照样品的包装方式与光照样品一致,但在一次性培养皿外面覆盖铝膜,按中国药典和ICH Q1B的要求,分别置于光照条件(可见光5000±500lux;紫外光90μw/cm 2)下考察; Influencing factor experiment: the samples under high temperature (60°C) were placed in an open disposable petri dish, and the samples under high humidity (25°C/75%RH) were placed in an open flat weighing bottle, and then placed in different The blast drying oven and desiccator were investigated. The illuminated samples are placed in a clean disposable petri dish, spread into a thin layer, and covered with a quartz glass cover. The packaging method of the shading control sample is the same as that of the illuminated sample, but the outside of the disposable petri dish is covered with aluminum film. According to the Chinese Pharmacopoeia and ICH The requirements of Q1B were placed under illumination conditions (visible light 5000±500 lux; ultraviolet light 90 μw/cm 2 ) for investigation;
长期和加速条件实验:将样品装入双层低密度聚乙烯袋,每层低密度聚乙烯袋分别用扎扣密封,一起放入铝箔袋中热封,然后分别放入长期条件(25℃/60%RH)和加速条件(40℃/75%RH)下考察。实验结果见表11:Long-term and accelerated condition experiments: The samples were put into double-layer low-density polyethylene bags, each layer of low-density polyethylene bags was sealed with a zipper, and then put into aluminum foil bags for heat sealing, and then put into long-term conditions (25℃/ 60%RH) and accelerated conditions (40°C/75%RH). The experimental results are shown in Table 11:
表11:式(I)化合物无定形固体稳定性实验结果Table 11: Amorphous solid stability test results of compounds of formula (I)
Figure PCTCN2021121043-appb-000009
Figure PCTCN2021121043-appb-000009
Figure PCTCN2021121043-appb-000010
Figure PCTCN2021121043-appb-000010
NA:未检测晶型。NA: No crystal form detected.
结论:式(I)化合物无定形具有良好的稳定性。Conclusion: The amorphous compound of formula (I) has good stability.
实施例4:式(I)化合物无定形溶解度实验Example 4: Amorphous solubility test of the compound of formula (I)
实验目的:Purpose:
测试37℃条件下,式(I)化合物无定形在不同pH介质(pH=1.0,2.0,3.8,4.5,5.5,6.0,6.8,7.4)、生物溶媒(SGF,FaSSIF,FeSSIF)及水中悬浮搅拌24小时后的溶解度。Under the condition of 37℃, the amorphous compound of formula (I) was suspended and stirred in different pH media (pH=1.0, 2.0, 3.8, 4.5, 5.5, 6.0, 6.8, 7.4), biological solvent (SGF, FaSSIF, FeSSIF) and water Solubility after 24 hours.
实验方法:experimental method:
1.配制不同pH介质:1. Prepare different pH media:
a)取11.4mL冰醋酸,加入100mL的容量瓶中,加超纯水定容,混匀即得2mol/L醋酸溶液;a) Take 11.4mL of glacial acetic acid, add it to a 100mL volumetric flask, add ultrapure water to make the volume, and mix well to obtain a 2mol/L acetic acid solution;
b)称取约2.722g磷酸二氢钾,加入100mL容量瓶中,加适量超纯水并超声溶解,再用超纯水定容,混匀即得0.2mol/L磷酸二氢钾溶液;b) Weigh about 2.722g of potassium dihydrogen phosphate, add it into a 100mL volumetric flask, add an appropriate amount of ultrapure water and dissolve by ultrasonic, then use ultrapure water to make up the volume, and mix to obtain a 0.2mol/L potassium dihydrogen phosphate solution;
c)取10mL的1.0mol/L氢氧化钠溶液,加入50mL容量瓶中,加超纯水定容,混匀即得0.2mol/L氢氧化钠溶液;c) Take 10mL of 1.0mol/L sodium hydroxide solution, add it to a 50mL volumetric flask, add ultrapure water to volume, and mix to obtain 0.2mol/L sodium hydroxide solution;
d)量取5mL的1.0mol/L盐酸溶液,加入到50mL的容量瓶中,加超纯水定容,混匀测得pH=0.99;d) Measure 5mL of 1.0mol/L hydrochloric acid solution, add it to a 50mL volumetric flask, add ultrapure water to volume, and mix to measure pH=0.99;
e)量取5mL的0.1mol/L盐酸溶液,加入到50mL的容量瓶中,加超纯水定容,混匀测得pH=1.98;e) Measure 5mL of 0.1mol/L hydrochloric acid solution, add it to a 50mL volumetric flask, add ultrapure water to volume, and mix to measure pH=1.98;
f)称取醋酸钠32.9mg,加入到50mL的容量瓶中,加入2mol/L醋酸溶液1.13mL,加适量超纯水并超声溶解,再用超纯水定容,混匀测得pH=3.81;f) Weigh 32.9 mg of sodium acetate, add it to a 50 mL volumetric flask, add 1.13 mL of 2mol/L acetic acid solution, add an appropriate amount of ultrapure water and dissolve it by ultrasonic, then use ultrapure water to dilute to volume, mix well to measure pH=3.81 ;
g)称取醋酸钠150.1mg,加入到50mL的容量瓶中,加入2mol/L醋酸溶液0.7mL,加适量超纯水并超声溶解,再用超纯水定容,混匀测得pH=4.49;g) Weigh 150.1 mg of sodium acetate, add it to a 50 mL volumetric flask, add 0.7 mL of 2 mol/L acetic acid solution, add an appropriate amount of ultrapure water and dissolve it by ultrasonic, then make up to volume with ultrapure water, mix well to measure pH=4.49 ;
h)称取醋酸钠0.325g,加入到50mL的容量瓶中,加入2mol/L醋酸溶液0.15mL,加适量超纯水并超声溶解,再用超纯水定容,混匀测得pH=5.47;h) Weigh 0.325g of sodium acetate, add it to a 50mL volumetric flask, add 0.15mL of 2mol/L acetic acid solution, add an appropriate amount of ultrapure water and dissolve it ultrasonically, then make up to volume with ultrapure water, mix well to measure pH=5.47 ;
i)取12.5mL 0.2mol/L磷酸二氢钾溶液,加入到50mL的容量瓶中,加入0.2mol/L氢氧化钠溶液1.4mL,加超纯水定容,混匀测得pH=6.03;i) Take 12.5mL of 0.2mol/L potassium dihydrogen phosphate solution, add it to a 50mL volumetric flask, add 1.4mL of 0.2mol/L sodium hydroxide solution, add ultrapure water to volume, mix well to measure pH=6.03;
j)取12.5mL 0.2mol/L磷酸二氢钾溶液,加入到50mL的容量瓶中,加入0.2mol/L氢氧化钠溶液5.6mL,加超纯水定容,混匀测得pH=6.78;j) Take 12.5mL of 0.2mol/L potassium dihydrogen phosphate solution, add it to a 50mL volumetric flask, add 5.6mL of 0.2mol/L sodium hydroxide solution, add ultrapure water to volume, mix well to measure pH=6.78;
k)取12.5mL 0.2mol/L磷酸二氢钾溶液,加入到50mL的容量瓶中,加入0.2mol/L氢氧化钠溶液9.775mL,加超纯水定容,混匀测得pH=7.42。k) Take 12.5mL of 0.2mol/L potassium dihydrogen phosphate solution, add it to a 50mL volumetric flask, add 9.775mL of 0.2mol/L sodium hydroxide solution, add ultrapure water to volume, mix well to measure pH=7.42.
2.称取约10mg式(I)化合物无定形样品至离心管中,加入1mL相应介质;2. Weigh about 10 mg of the amorphous sample of the compound of formula (I) into a centrifuge tube, and add 1 mL of the corresponding medium;
3.以约750rpm转速在37℃条件下磁力悬浮搅拌;3. Magnetic suspension stirring at about 750rpm at 37°C;
4.平衡24小时后,分离上清液用于pH及溶解度测试。4. After 24 hours of equilibration, the supernatant was separated for pH and solubility testing.
实验结果如表12所示:The experimental results are shown in Table 12:
表12:式(I)化合物无定形在不同介质中37℃溶解度汇总Table 12: Summary of 37°C solubility of compound of formula (I) amorphous in different media
实验编号Experiment number 介质medium 溶解度(mg/mL) # Solubility (mg/mL) # 24小时后pHpH after 24 hours
11 1.01.0 0.58770.5877 0.760.76
22 2.02.0 8.20378.2037 5.565.56
33 3.83.8 9.12909.1290 4.294.29
44 4.54.5 9.57989.5798 4.744.74
55 5.55.5 2.54892.5489 5.925.92
66 6.06.0 0.09050.0905 6.326.32
77 6.86.8 0.05590.0559 6.756.75
88 7.47.4 0.00520.0052 7.367.36
99 water 0.02790.0279 7.377.37
1010 SGFSGF 9.38219.3821 2.822.82
1111 FaSSIFFaSSIF 0.06310.0631 6.736.73
1212 FeSSIFFeSSIF 0.24460.2446 5.215.21
#:y=10336110.7x+5294.3,R2=0.9999,LOQ=0.12μg/mL,LOD=0.04μg/mL;SGF:模拟胃液;FaSSIF:禁食状态模拟肠液;FeSSIF:进食状态模拟肠液。#: y=10336110.7x+5294.3, R2=0.9999, LOQ=0.12 μg/mL, LOD=0.04 μg/mL; SGF: simulated gastric fluid; FaSSIF: simulated intestinal fluid in fasting state; FeSSIF: simulated intestinal fluid in fed state.
结论:式(I)化合物无定形在不同pH介质(pH=1.0,2.0,3.8,4.5,5.5,6.0,6.8,7.4)中的溶解度为0.005-9.6mg/mL;在生物溶媒(SGF,FaSSIF,FeSSIF)中的溶解度为0.06-9.4mg/mL;在水中的溶解度为0.03mg/mL。Conclusion: The solubility of the compound of formula (I) amorphous in different pH media (pH=1.0, 2.0, 3.8, 4.5, 5.5, 6.0, 6.8, 7.4) is 0.005-9.6 mg/mL; in biological solvents (SGF, FaSSIF) , FeSSIF) in the solubility of 0.06-9.4mg/mL; the solubility in water is 0.03mg/mL.
实施例5:式(I)化合物无定形的机械稳定性实验Example 5: Mechanical stability test of the amorphous compound of formula (I)
实验方法:experimental method:
1)在不加入水或溶剂条件下,手动研磨式(I)化合物无定形样品约5分钟后测试XRPD;1) Test XRPD after grinding the amorphous sample of the compound of formula (I) manually for about 5 minutes without adding water or solvent;
2)将式(I)化合物无定形样品加入圆形模具(直径6mm)中,加压直至样品所受压强达到350MPa左右,将压片后的样品直接测试XRPD。2) Add the amorphous sample of the compound of formula (I) into a circular mold (diameter 6mm), pressurize until the pressure on the sample reaches about 350MPa, and directly test the XRPD of the sample after tableting.
实验结果:Experimental results:
经过研磨和压片后,样品晶型未发生变化,仍为无定形。After grinding and tableting, the crystal form of the sample did not change and was still amorphous.
结论:式(I)化合物无定形在手动研磨及压片条件(350MPa)下形态稳定。Conclusion: The amorphous compound of formula (I) is morphologically stable under manual grinding and tableting conditions (350MPa).
实验例一、体外细胞活性测试Experimental example 1. In vitro cell activity test
1.实验目的1. Experimental purpose
通过测定进入高表达人源SGLT1和SGLT2(Human-SGLT1和Human-SGLT2)转运体细胞内的带[ 14C]标记的Methylα-D-glucopyranosid(甲基α-D-吡喃葡萄糖苷)的量,评估式(I)化合物无定形对人源 SGLT1和SGLT2转运体活性的抑制作用。 By measuring the amount of [ 14 C]-labeled Methylα-D-glucopyranosid (methyl α-D-glucopyranoside) entering the cells of transporters with high expression of human SGLT1 and SGLT2 (Human-SGLT1 and Human-SGLT2) , to evaluate the inhibitory effect of the compound of formula (I) amorphous on the activity of human SGLT1 and SGLT2 transporters.
2.实验方法2. Experimental method
1)实验设双复孔,实验重复3次;1) The experiment was set up with double holes, and the experiment was repeated 3 times;
2)试剂与细胞准备;2) Reagent and cell preparation;
a)配制实验缓冲液,各成分终浓度见表13:a) Preparation of experimental buffer, the final concentration of each component is shown in Table 13:
表13缓冲液组成Table 13 Buffer composition
试剂reagent 浓度(mM)Concentration (mM)
HEPESHEPES 1010
MgCl 2 MgCl 2 1.21.2
KClKCl 4.74.7
CaCl 2 CaCl 2 2.22.2
NaCl NaCl 120120
b)化合物准备:式(I)化合物无定形储存液为浓度为10mM,用DMSO稀释。b) Compound preparation: The amorphous stock solution of the compound of formula (I) was at a concentration of 10 mM, diluted with DMSO.
c)化合物稀释由Bravo自动移液工作站完成,3倍递减稀释10个浓度。在最终100μL的反应体系中,人源SGLT1和SGLT2靶点化合物的检测浓度从0.4μM稀释至0.0203nM,分别为:400.0000、133.3333、44.4444、14.8148、4.9383、1.6461、0.5487、0.1829、0.0610、0.0203nM。c) Compound dilution was done by Bravo automatic pipetting workstation, and 10 concentrations were decremented 3-fold. In the final 100 μL reaction system, the detection concentrations of human SGLT1 and SGLT2 target compounds were diluted from 0.4 μM to 0.0203 nM, respectively: 400.0000, 133.3333, 44.4444, 14.8148, 4.9383, 1.6461, 0.5487, 0.1829, 0.0610, 0.0203 nM .
d)同位素准备:用实验缓冲液将同位素贮存液[ 14C]Methylα-D-glucopyranosid稀释到6μM工作浓度。 d) Isotope preparation: Dilute the isotope stock solution [ 14 C]Methylα-D-glucopyranosid with assay buffer to a working concentration of 6 μM.
e)细胞准备:将人源SGLT1或人源SGLT2细胞以7×104个/孔种于96孔细胞板中,置于37℃,5%CO 2细胞培养箱中过夜培养。 e) Cell preparation: Human SGLT1 or human SGLT2 cells were seeded in a 96-well cell plate at 7×104 cells/well, and cultured overnight in a 37°C, 5% CO 2 cell incubator.
3)实验过程3) Experimental process
a)从培养箱中取出96孔板,甩掉培养基并用150μL/孔的实验缓冲液润洗细胞,再将缓冲液甩掉。加入49μL/孔新的缓冲液。a) Remove the 96-well plate from the incubator, shake off the medium and rinse the cells with 150 μL/well of experimental buffer, then shake off the buffer. Add 49 μL/well of new buffer.
b)根据96孔板布局,加入1μL梯度稀释的化合物溶液。加1μL DMSO至高信号孔(HC,DMSO终浓度1%);加1μL LX2761至低信号孔(LC,LX2761终浓度0.4μM)。b) Add 1 μL of serially diluted compound solutions according to the 96-well plate layout. Add 1 μL DMSO to high signal wells (HC, DMSO final concentration 1%); add 1 μL LX2761 to low signal wells (LC, LX2761 final concentration 0.4 μM).
c)每孔加入50μL 6μM[ 14C]Methylα-D-glucopyranosid同位素至96孔板。 c) Add 50 μL of 6 μM [ 14 C]Methylα-D-glucopyranosid isotope per well to a 96-well plate.
d)37℃孵育2小时。d) Incubate at 37°C for 2 hours.
e)取出96孔板,吸去反应体系。用预冷的实验缓冲液洗三次,加50μL/孔10%NaOH,置于摇床上震荡裂解5分钟。e) Take out the 96-well plate and aspirate the reaction system. Wash three times with pre-cooled experimental buffer, add 50 μL/well of 10% NaOH, and place on a shaker for lysing for 5 minutes.
f)将96孔板中的细胞裂解液分别转移至闪烁管中,每管加2mL闪烁液,用Tri-Carb液体闪烁计数仪读数。f) Transfer the cell lysate in the 96-well plate to scintillation vials, add 2 mL of scintillation fluid to each tube, and read with a Tri-Carb liquid scintillation counter.
g)分析实验数据,%抑制率=(高信号孔均值-样品孔信号值)/(高信号孔均值-低信号孔均值)*100。运用GraphPad Prism 5进行曲线拟合:Log(inhibitor)vs.response—Variable slope,得到IC 50g) Analysis of experimental data, % inhibition rate=(mean value of high signal wells-signal value of sample wells)/(mean value of high signal wells-mean value of low signal wells)*100. Curve fitting using GraphPad Prism 5: Log(inhibitor) vs. response—Variable slope yields IC50 .
3.实验结果见表14:3. The experimental results are shown in Table 14:
表14:体外细胞活性测试结果Table 14: In Vitro Cell Viability Test Results
化合物compound Human-SGLT1IC 50(nM) Human-SGLT1 IC50 (nM) Human-SGLT2IC 50(nM) Human-SGLT2IC 50 (nM)
式(I)化合物无定形Amorphous compound of formula (I) 1.8±0.41.8±0.4 1.6±1.01.6±1.0
结论:式(I)化合物无定形对人源SGLT1和SGLT2转运体活性有显著抑制作用。Conclusion: The amorphous compound of formula (I) has a significant inhibitory effect on the activities of human SGLT1 and SGLT2 transporters.
实验例二、大鼠体内DMPK研究Experimental example 2. DMPK study in rats
1.实验目的:1. Experimental purpose:
以雄性SD大鼠为受试动物,单次给药后测定式(I)化合物无定形血药浓度并评估药代动力学行为。Male SD rats were used as test animals, and the amorphous plasma concentration of the compound of formula (I) was determined after single administration and the pharmacokinetic behavior was evaluated.
2.实验操作:2. Experimental operation:
选择健康成年雄性SD大鼠6只,3只为静注组,3只为口服组。静注组溶媒为5%(w/v)羟丙基β环糊精(HP-β-CD)水溶液,式(I)化合物无定形与适量静注组溶媒混合后,制备得到0.25mg/mL澄清溶液;口服组溶媒为0.5%(w/v)甲基纤维素(methylcellulose)/0.02%吐温80水溶液,式(I)化合物无定形与适量口服组溶媒混合后,制备得到0.5mg/mL均一混悬液。大鼠0.5mg/kg静脉给药或5mg/kg口服给药后,采集不同时间点的血浆样品,应用LC-MS/MS方法分析式(I)化合物浓度,并用Phoenix WinNonlin软件(美国Pharsight公司)计算药代参数。Six healthy adult male SD rats were selected, 3 were in the intravenous group and 3 were in the oral group. The vehicle in the intravenous injection group is 5% (w/v) hydroxypropyl β-cyclodextrin (HP-β-CD) aqueous solution. After the amorphous compound of formula (I) is mixed with an appropriate amount of the vehicle in the intravenous injection group, 0.25 mg/mL is prepared. Clear solution; Oral group solvent is 0.5% (w/v) methylcellulose/0.02% Tween 80 aqueous solution, the compound of formula (I) amorphous is mixed with an appropriate amount of oral group solvent to prepare 0.5mg/mL Homogeneous suspension. After intravenous administration of 0.5 mg/kg or oral administration of 5 mg/kg in rats, plasma samples were collected at different time points, and the concentration of the compound of formula (I) was analyzed by LC-MS/MS method, and Phoenix WinNonlin software (Pharsight, USA) was used. Calculate pharmacokinetic parameters.
3.实验结果见表15:3. The experimental results are shown in Table 15:
表15:化合物PK测试结果Table 15: Compound PK test results
Figure PCTCN2021121043-appb-000011
Figure PCTCN2021121043-appb-000011
注:C max为最大浓度;F%为口服生物利用度;AUC po为口服暴露量;Vd ss为分布容积;Cl为清除率;T 1/2为半衰期。 Note: Cmax is the maximum concentration; F% is oral bioavailability; AUC po is oral exposure; Vd ss is volume of distribution; Cl is clearance; T 1/2 is half-life.
结论:式(I)化合物无定形口服给药后,在体内血药浓度和口服生物利用度均很低,具有直接作用于肠道SGLT1的药代动力学性质。Conclusion: After oral administration of the compound of formula (I) in amorphous form, the plasma concentration and oral bioavailability in vivo are very low, and it has the pharmacokinetic properties of directly acting on intestinal SGLT1.
实验例三、式(I)化合物无定形在小鼠OGTT模型药效研究Experimental Example 3. Pharmacodynamic study of amorphous compound of formula (I) in mouse OGTT model
1.实验动物:1. Experimental animals:
Figure PCTCN2021121043-appb-000012
Figure PCTCN2021121043-appb-000012
2.实验分组:2. Experimental grouping:
表16:实验分组信息Table 16: Experiment Grouping Information
Figure PCTCN2021121043-appb-000013
Figure PCTCN2021121043-appb-000013
3.实验流程:3. Experimental process:
1)动物适应及准备1) Animal adaptation and preparation
实验动物抵达设施后在动物房饲养适应环境1周。The experimental animals were housed in the animal room to acclimate to the environment for 1 week after arriving at the facility.
2)给药2) Administration
动物称重、禁食6h、测禁食血糖(0min血糖)并分组后,分别口服给予式(I)化合物无定形和溶媒,随即口服给予2g/kg,5mL/kg葡萄糖溶液。并于给糖后的15,30,60,90,120分钟分别对动物通过尾尖采血进行血糖检测,根据时间对血糖数据绘制糖耐量曲线,计算曲线下面积(AUC 1-120分钟)。 After the animals were weighed, fasted for 6 hours, measured fasting blood sugar (0min blood sugar) and grouped into groups, the compound of formula (I) amorphous and vehicle were orally administered respectively, followed by oral administration of 2 g/kg, 5 mL/kg glucose solution. At 15, 30, 60, 90, and 120 minutes after glucose administration, the animals were collected blood from the tail tip for blood glucose detection, and the glucose tolerance curve was drawn according to the time, and the area under the curve (AUC 1-120 minutes ) was calculated.
3)数据分析:3) Data analysis:
所有数值将表示为平均值。统计学分析使用Graphpad Prism 6单因素方差分析Tukey’s多重比较检验来评估。小于0.05的p值被认为具有统计学显着性。All values will be expressed as averages. Statistical analysis was assessed using Graphpad Prism 6 one-way ANOVA with Tukey's multiple comparison test. A p-value less than 0.05 was considered statistically significant.
4.实验结果:4. Experimental results:
表17:小鼠糖耐受量体内药效实验结果Table 17: In vivo efficacy test results of glucose tolerance in mice
Figure PCTCN2021121043-appb-000014
Figure PCTCN2021121043-appb-000014
注:*表示相对于溶媒对照组p<0.05,**表示相对于溶媒对照组p<0.01,***表示相对于溶媒对照组p<0.001and****表示相对于溶媒对照组p<0.0001。Note: * means p<0.05 relative to the vehicle control group, ** means p<0.01 relative to the vehicle control group, *** means p<0.001 relative to the vehicle control group and **** means p<0.001 relative to the vehicle control group 0.0001.
结论:相比溶媒对照组,式(I)化合物无定形单次给药后可呈剂量依赖性降低小鼠葡萄糖口服后的血糖水平。Conclusion: Compared with the vehicle control group, the amorphous compound of formula (I) can dose-dependently reduce the blood glucose level of mice after oral administration of glucose.

Claims (12)

  1. 式(I)化合物的无定形,其X射线粉末衍射(XRPD)图谱如图1所示。The amorphous form of the compound of formula (I), its X-ray powder diffraction (XRPD) pattern is shown in FIG. 1 .
    Figure PCTCN2021121043-appb-100001
    Figure PCTCN2021121043-appb-100001
  2. 根据权利要求1所述的无定形,其差示扫描量热曲线在74.9±3℃处可观察到一个玻璃态转化信号。The amorphous according to claim 1, the differential scanning calorimetry curve of which can observe a glass transition signal at 74.9±3°C.
  3. 根据权利要求2所述的无定形,其DSC图谱如图2所示。The amorphous according to claim 2, its DSC spectrum is shown in Figure 2.
  4. 根据权利要求1所述的无定形,其热重分析曲线在150.0±3℃时失重达2.56%。The amorphous according to claim 1, its thermogravimetric analysis curve is 2.56% weight loss at 150.0±3°C.
  5. 根据权利要求4所述的无定形,其TGA图谱如图3所示。The amorphous according to claim 4, its TGA spectrum is shown in Figure 3.
  6. 式(Ⅰ)化合物的无定形的制备方法,包含反溶剂添加法、缓慢降温法、悬浮搅拌法、缓慢挥发法、气固渗透法和温度循环法。The amorphous preparation method of the compound of formula (I) includes anti-solvent addition method, slow cooling method, suspension stirring method, slow volatilization method, gas-solid permeation method and temperature circulation method.
  7. 根据权利要求6所述的制备方法,其中,反溶剂添加法包含:The preparation method according to claim 6, wherein the anti-solvent addition method comprises:
    1)将式(I)化合物加入到溶剂中形成澄清溶液;1) adding the compound of formula (I) to a solvent to form a clear solution;
    2)向溶液中加入反溶剂;2) adding an anti-solvent to the solution;
    其中,in,
    溶剂为乙醇、丙酮、四氢呋喃、乙腈、乙酸正丙酯、苯甲醚或2-甲基四氢呋喃;The solvent is ethanol, acetone, tetrahydrofuran, acetonitrile, n-propyl acetate, anisole or 2-methyltetrahydrofuran;
    反溶剂为H 2O或正庚烷。 The anti-solvent is H2O or n-heptane.
  8. 根据权利要求6所述的制备方法,其中,缓慢降温法包含:preparation method according to claim 6, wherein, the slow cooling method comprises:
    1)50℃下,将式(I)化合物在溶剂中配制成澄清溶液;1) at 50 ° C, the compound of formula (I) is prepared into a clear solution in a solvent;
    2)溶液以0.1℃/分钟从50℃降温至5℃;2) The solution is cooled from 50°C to 5°C at 0.1°C/min;
    其中,in,
    溶剂为叔丁醇、乙酸异丙酯、乙腈:水(v/v,1:3)或2-甲基四氢呋喃:环己烷(v/v,1:3)。Solvents were tert-butanol, isopropyl acetate, acetonitrile:water (v/v, 1:3) or 2-methyltetrahydrofuran:cyclohexane (v/v, 1:3).
  9. 根据权利要求6所述的制备方法,其中,悬浮搅拌法包含:The preparation method according to claim 6, wherein the suspension stirring method comprises:
    1)将式(I)化合物加入到溶剂中形成悬浊液;1) adding the compound of formula (I) to a solvent to form a suspension;
    2)悬浊液溶液用磁力搅拌转晶;2) The suspension solution is crystallized with magnetic stirring;
    其中,in,
    溶剂为甲基叔丁基醚、乙酸乙酯:正庚烷(v/v,1:3)、2-甲基四氢呋喃:环己烷(v/v,1:3)、甲基叔丁基醚:正庚烷(v/v,1:7)、四氢呋喃:正庚烷(v/v,1:7)、乙酸乙酯:正庚烷(v/v,1:7)、2-丁酮:正庚烷(v/v,1:7)、乙腈:水(v/v,1:9)、丙酮:水(v/v,1:12)、正庚烷、水、四氢呋喃:正戊烷(v/v,1:7)、N-甲基吡咯烷酮:水(v/v,1:5)、二甲基亚砜:水(v/v,1:5)或间二甲苯。The solvent is methyl tert-butyl ether, ethyl acetate: n-heptane (v/v, 1:3), 2-methyltetrahydrofuran: cyclohexane (v/v, 1:3), methyl tert-butyl Ether: n-heptane (v/v, 1:7), tetrahydrofuran: n-heptane (v/v, 1:7), ethyl acetate: n-heptane (v/v, 1:7), 2-butane Ketone: n-heptane (v/v, 1:7), acetonitrile: water (v/v, 1:9), acetone: water (v/v, 1:12), n-heptane, water, tetrahydrofuran: n- Pentane (v/v, 1:7), N-methylpyrrolidone:water (v/v, 1:5), dimethyl sulfoxide:water (v/v, 1:5) or m-xylene.
  10. 根据权利要求6所述的制备方法,其中,缓慢挥发法包含:The preparation method according to claim 6, wherein the slow volatilization method comprises:
    1)将式(I)化合物溶解在溶剂中形成澄清溶液;1) dissolving the compound of formula (I) in a solvent to form a clear solution;
    2)把澄清溶液用封口膜密封扎小孔后缓慢挥发析晶;2) Slowly volatilize and crystallize the clear solution after sealing the small holes with parafilm;
    其中,in,
    溶剂为乙酸乙酯、氯仿、甲醇、丙酮、乙腈或四氢呋喃。The solvent is ethyl acetate, chloroform, methanol, acetone, acetonitrile or tetrahydrofuran.
  11. 根据权利要求6所述的制备方法,其中,气固渗透法包含:The preparation method according to claim 6, wherein the gas-solid permeation method comprises:
    1)将式(I)化合物置于小瓶中;1) placing the compound of formula (I) in a vial;
    2)把上述小瓶敞口放置于水的氛围中,密封后室温下静置。2) The above-mentioned vial was opened and placed in an atmosphere of water, sealed and left to stand at room temperature.
  12. 根据权利要求6所述的制备方法,其中,温度循环法包含:The preparation method according to claim 6, wherein the temperature cycling method comprises:
    1)50℃下,将式(I)化合物溶解在溶剂中形成悬浊液;1) at 50°C, the compound of formula (I) is dissolved in a solvent to form a suspension;
    2)把悬浊液以0.1℃/分钟的速度按照50℃-5℃-50℃-5℃的程序进行升降温进行转晶;2) The temperature of the suspension is raised and lowered at a rate of 0.1°C/min according to the procedure of 50°C-5°C-50°C-5°C to transfer crystals;
    其中,in,
    溶剂为正庚烷、甲基叔丁基醚、2-甲基四氢呋喃:正庚烷(v/v,1:3)或N-甲基吡咯烷酮:水(v/v,1:5)。The solvent was n-heptane, methyl tert-butyl ether, 2-methyltetrahydrofuran:n-heptane (v/v, 1:3) or N-methylpyrrolidone:water (v/v, 1:5).
PCT/CN2021/121043 2020-09-27 2021-09-27 Crystal form of glucoside compounds and use thereof WO2022063305A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202180063705.6A CN116322675A (en) 2020-09-27 2021-09-27 Crystal form of glucoside compound and application thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202011032215 2020-09-27
CN202011032215.3 2020-09-27

Publications (1)

Publication Number Publication Date
WO2022063305A1 true WO2022063305A1 (en) 2022-03-31

Family

ID=80844973

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/121043 WO2022063305A1 (en) 2020-09-27 2021-09-27 Crystal form of glucoside compounds and use thereof

Country Status (2)

Country Link
CN (1) CN116322675A (en)
WO (1) WO2022063305A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014081660A1 (en) * 2012-11-20 2014-05-30 Lexicon Pharmaceuticals, Inc. Inhibitors of sodium glucose cotransporter 1
CN106892948A (en) * 2015-12-17 2017-06-27 广东东阳光药业有限公司 Glucopyranosyl derivatives and its in application pharmaceutically
WO2019134667A1 (en) * 2018-01-05 2019-07-11 南京明德新药研发股份有限公司 Sglts inhibitor and application thereof
WO2019185026A1 (en) * 2018-03-30 2019-10-03 南京明德新药研发有限公司 Glucoside derivatives acting as inhibitors of sglts, and use thereof
WO2020200153A1 (en) * 2019-03-29 2020-10-08 南京明德新药研发有限公司 Glucoside derivative that acts as sglt1 inhibitor and application thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014081660A1 (en) * 2012-11-20 2014-05-30 Lexicon Pharmaceuticals, Inc. Inhibitors of sodium glucose cotransporter 1
CN106892948A (en) * 2015-12-17 2017-06-27 广东东阳光药业有限公司 Glucopyranosyl derivatives and its in application pharmaceutically
WO2019134667A1 (en) * 2018-01-05 2019-07-11 南京明德新药研发股份有限公司 Sglts inhibitor and application thereof
WO2019185026A1 (en) * 2018-03-30 2019-10-03 南京明德新药研发有限公司 Glucoside derivatives acting as inhibitors of sglts, and use thereof
WO2020200153A1 (en) * 2019-03-29 2020-10-08 南京明德新药研发有限公司 Glucoside derivative that acts as sglt1 inhibitor and application thereof

Also Published As

Publication number Publication date
CN116322675A (en) 2023-06-23

Similar Documents

Publication Publication Date Title
EP2870163B1 (en) Pharmaceutical compositions comprising rifaximin and amino acids, preparation method and use thereof
US9018225B1 (en) Rifaximin crystalline forms and methods of preparation thereof
CN105121409B (en) Crystalline form of dolutegravir sodium salt and preparation method therefor
WO2018049632A1 (en) Crystal form of ozanimod, and preparation method and pharmaceutical composition thereof
WO2023040513A1 (en) Crystal form of amg510 compound, and preparation method therefor and use thereof
WO2022063305A1 (en) Crystal form of glucoside compounds and use thereof
CN109963567A (en) The crystal form of bile acid derivative
WO2023193563A1 (en) Crystal form a of thienopyridine compound, and preparation method therefor and pharmaceutical composition thereof
WO2022258060A1 (en) Crystal form of lanifibranor and preparation method therefor
TWI758287B (en) An amine solvent complex of sodium-dependent glucose co-transporter protein, a preparation method for same and applications thereof
JP2021532165A (en) Addition salt of S1P1 receptor agonist and its crystalline form, and pharmaceutical composition
CN110869382B (en) Steroid derivative FXR agonist crystal or amorphous substance, preparation method and application thereof
WO2019101058A1 (en) Crystal form of urat1 inhibitor, and preparation method therefor
WO2021004388A1 (en) Crystal form of sglt inhibitor and application thereof
CN101223150A (en) Polymorphic forms of (s)-1-cyanobutan-2-yl (s)-1-(3-(3-(3-methoxy-4-(oxazol-5-yl) phenyl) ureido) phenyl) ethylcarbamate
CN113214163B (en) Albendazole-cholic acid derivative and preparation method and application thereof
WO2022179636A1 (en) Crystal forms of fxr receptor agonist
WO2023061437A1 (en) Salt of antitumor drug and crystal form thereof
CN113166121B (en) Solid form, crystal form and crystal form A of FXR agonist, and preparation method and application thereof
WO2020151672A1 (en) Dapagliflozin crystal form, preparation therefor, and use thereof
CN108299412A (en) The addition salts and its crystal form and pharmaceutical composition of a kind of S1P1 receptor stimulating agents
CN110117272A (en) The salt and its crystal form of cell cycle protein dependent kinase inhibitor

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: 21871674

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 11/09/2023)