WO2021000934A1 - 一种苯并咪唑-2-酮类化合物的晶型、溶剂化物、溶剂化物的晶型及它们的制备方法 - Google Patents

一种苯并咪唑-2-酮类化合物的晶型、溶剂化物、溶剂化物的晶型及它们的制备方法 Download PDF

Info

Publication number
WO2021000934A1
WO2021000934A1 PCT/CN2020/100129 CN2020100129W WO2021000934A1 WO 2021000934 A1 WO2021000934 A1 WO 2021000934A1 CN 2020100129 W CN2020100129 W CN 2020100129W WO 2021000934 A1 WO2021000934 A1 WO 2021000934A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
crystal form
solvate
angles
ray powder
Prior art date
Application number
PCT/CN2020/100129
Other languages
English (en)
French (fr)
Inventor
雷茂义
罗云富
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 US17/624,462 priority Critical patent/US20220372001A1/en
Priority to EP20835633.7A priority patent/EP3995490A4/en
Priority to CN202080048994.8A priority patent/CN114072382B/zh
Priority to JP2022500106A priority patent/JP2022539257A/ja
Publication of WO2021000934A1 publication Critical patent/WO2021000934A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D235/26Oxygen atoms
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to the crystal form of compound 1, its solvate, the crystal form of its solvate and their preparation method, and also includes the application of the crystal form in the preparation of drugs for treating diseases related to TNF ⁇ .
  • Tumor necrosis factor is a cytokine that is mainly released by monocytes in response to immune stimuli. TNF ⁇ can promote most processes of cell differentiation, recruitment, proliferation and protein degradation. At low levels, TNF ⁇ has a protective effect against infectious agents, tumors and tissue damage. However, excessive release of TNF ⁇ can also cause diseases. For example, when TNF ⁇ is administered to mammals or humans, it can cause or aggravate inflammation, fever, cardiovascular effects, bleeding, blood coagulation, and acute reactions similar to acute infection and shock.
  • TNF ⁇ production in animals or humans often indicates the following diseases: endotoxemia and/or toxic shock syndrome, cachexia, adult respiratory stress syndrome, cancer (such as solid tumors and hematological tumors) ), heart disease (such as congestive heart failure), viral infection, genetic disease, inflammatory disease, allergic disease or autoimmune disease.
  • Cancer is a particularly devastating disease.
  • the increase in the level of TNF ⁇ in the blood indicates the risk of cancer or the spread of cancer.
  • cancer cells cannot survive in the circulatory system of healthy subjects.
  • One of the reasons is that the inner wall of blood vessels is a barrier to extravasation of tumor cells.
  • ELAM-1 on endothelial cells can mediate and promote colon cancer cells to adhere to endothelium treated with cytokines.
  • Cyclic adenylate plays a role in many diseases and disorders.
  • the increase of cAMP concentration in leukocytes during inflammation inhibits the activation of leukocytes, and subsequently releases inflammatory regulatory factors including TNF ⁇ and NF- ⁇ B.
  • Increased cAMP levels can also cause relaxation of the smooth muscles of the respiratory tract.
  • PDE cyclic nucleotide phosphodiesterase
  • PDE-4A, PDE-4B and PDE-4D in inflammatory cells is stronger than that of PDE-4C.
  • Inhibiting the PDE4 enzyme leads to an increase in cAMP levels, thereby regulating TNF ⁇ levels to achieve the purpose of treating diseases.
  • the present invention provides crystal form A of compound 1, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 11.91 ⁇ 0.20°, 19.36 ⁇ 0.20°, 23.17 ⁇ 0.2°.
  • the X-ray powder diffraction pattern of Form A of the above compound 1 has characteristic diffraction peaks at the following 2 ⁇ angles: 11.26 ⁇ 0.20°, 11.91 ⁇ 0.20°, 12.91 ⁇ 0.20°, 14.27 ⁇ 0.20 °, 19.36 ⁇ 0.20°, 22.26 ⁇ 0.20°, 23.17 ⁇ 0.20°, 24.97 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of Form A of the above compound 1 has characteristic diffraction peaks at the following 2 ⁇ angles: 11.26 ⁇ 0.20°, 11.91 ⁇ 0.20°, 12.91 ⁇ 0.20°, 14.27 ⁇ 0.20 °, 15.83 ⁇ 0.20°, 17.53 ⁇ 0.20°, 19.36 ⁇ 0.20°, 20.33 ⁇ 0.20°, 22.26 ⁇ 0.20°, 23.17 ⁇ 0.20°, 24.97 ⁇ 0.20°, 26.50 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of Form A of the above compound 1 has characteristic diffraction peaks at the following 2 ⁇ angles: 11.26°, 11.91°, 12.91°, 14.27°, 15.83°, 17.53°, 19.36°, 20.33°, 22.26°, 22.59°, 23.17°, 24.97°, 26.50°, 29.46°.
  • the XRPD pattern analysis data of the above compound 1 crystal form A is shown in Table 1:
  • the XRPD pattern of the above crystal form A of compound 1 is shown in FIG. 1.
  • the above-mentioned crystal form A of compound 1 has a differential scanning calorimetry curve with the starting point of the endothermic peak at 147.0 ⁇ 3.0°C.
  • the above-mentioned crystal form A of compound 1 has a DSC chart as shown in FIG. 2.
  • the above-mentioned crystal form A of compound 1 has a thermogravimetric analysis curve that has a weight loss of 0.70% at 140.0 ⁇ 3.0°C.
  • the above-mentioned crystal form A of compound 1 has a TGA pattern as shown in FIG. 3.
  • the present invention also provides a solvate represented by formula (I-1)
  • n is selected from 0.1 to 1.5.
  • n is selected from 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4 and 1.5.
  • the present invention also provides crystal form B of the solvate represented by formula (I-1-1), wherein n is 0.5, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ angles: 6.84 ⁇ 0.20°, 8.90 ⁇ 0.20°, 23.00 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the crystal form B of the above-mentioned solvate has characteristic diffraction peaks at the following 2 ⁇ angles: 6.84 ⁇ 0.20°, 8.90 ⁇ 0.20°, 11.27 ⁇ 0.20°, 12.75 ⁇ 0.20 °, 16.15 ⁇ 0.20°, 17.54 ⁇ 0.20°, 22.06 ⁇ 0.20°, 23.00 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the crystal form B of the above-mentioned solvate has characteristic diffraction peaks at the following 2 ⁇ angles: 6.84 ⁇ 0.20°, 8.90 ⁇ 0.20°, 11.27 ⁇ 0.20°, 12.75 ⁇ 0.20 °, 16.15 ⁇ 0.20°, 17.54 ⁇ 0.20°, 19.17 ⁇ 0.20°, 19.70 ⁇ 0.20°, 20.41 ⁇ 0.20°, 22.06 ⁇ 0.20°, 23.00 ⁇ 0.20°, 25.95 ⁇ 0.20°.
  • the X-ray powder diffraction pattern of the above-mentioned solvate crystal form B has characteristic diffraction peaks at the following 2 ⁇ angles: 6.84°, 8.90°, 11.27°, 12.75°, 13.29°, 14.94°, 16.15°, 17.54°, 17.93°, 19.17°, 19.70°, 20.41°, 20.79°, 22.06°, 22.72°, 23.00°, 23.86°, 25.95°, 27.82°, 28.45°, 30.14°, 32.87°.
  • the XRPD pattern of the crystalline form B of the above solvate is shown in FIG. 5.
  • the differential scanning calorimetry curve of the above-mentioned solvate of crystal form B has the starting point of the endothermic peak at 77.5 ⁇ 3.0°C.
  • the DSC spectrum of the crystalline form B of the above-mentioned solvate is shown in FIG. 6.
  • thermogravimetric analysis curve of the above-mentioned solvate crystal form B has a weight loss of 10.46% at 80.0 ⁇ 3.0°C.
  • the TGA pattern of the above-mentioned solvate crystal form B is shown in FIG. 7.
  • the present invention also provides the application of the above-mentioned crystal form A, the above-mentioned solvate or the above-mentioned solvate of the above-mentioned crystal form B of compound 1 in the preparation of drugs for treating TNF ⁇ -related diseases.
  • the compound 1 of the present invention has stable properties, low hygroscopicity, and good drug prospects; compound 1 of the present invention exhibits excellent in vitro activity of inhibiting phosphodiesterase 4B subtype (PDE4B); compound 1 of the present invention is shown in hPBMC It has excellent in vitro activity for inhibiting the production of TNF ⁇ ; the compound 1 of the present invention in the three dose groups of 0.1, 0.3 and 3 mg/ear has a significant improvement effect on the symptoms of PMA-induced ear edema in mice, and can significantly inhibit ear weight gain. And the three dose groups all showed a good dose-effect relationship.
  • PDE4B phosphodiesterase 4B subtype
  • 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 combining them with other chemical synthesis methods, and those skilled in the art.
  • Well-known equivalent alternatives, preferred implementations include but are not limited to the embodiments 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.
  • SXRD single crystal X-ray diffraction
  • the cultured single crystal is collected with a Bruker D8 venture diffractometer to collect diffraction intensity data
  • the light source is CuK ⁇ radiation
  • the scanning method After scanning and collecting relevant data, the direct method (Shelxs97) is further used to analyze the crystal structure to confirm the absolute configuration.
  • the compound is based on the conventional naming principles in the field or The software is named, and the commercially available compounds use the supplier catalog name.
  • Test method Approximately 10-20mg sample is used for XRPD detection.
  • Light tube voltage 40kV
  • light tube current 40mA
  • Fig. 1 is an XRPD spectrum of Cu-K ⁇ radiation of crystal form A of compound 1.
  • Figure 2 is a DSC spectrum of Compound 1 crystal form A.
  • Figure 3 is a TGA spectrum of Compound 1 crystal form A.
  • Figure 4 is the DVS spectrum of Compound 1 crystal form A.
  • Figure 5 is an XRPD spectrum of Cu-K ⁇ radiation of the B crystal form of Compound 1.
  • Fig. 6 is a DSC chart of Compound 1 crystal form B.
  • Figure 7 is a TGA spectrum of the B crystal form of compound 1.
  • compound 4 (122.83g, 0.30mol) was dissolved in a mixed solvent of dichloromethane (500mL) and ethyl acetate (500mL), and then wet palladium carbon (7.50g, purity : 10%), hydrogen replacement three times, and the reaction mixture was stirred and reacted at 25°C-35°C and hydrogen (25-35psi) atmosphere for 16 hours (four batches were cast in parallel, and combined treatment). After the reaction was completed, the four batches of the combined reaction solution were filtered through Celite, and the filter cake was washed with dichloromethane (200 mL). The filtrate was spin-dried under reduced pressure to obtain a crude product.
  • the crude product was added to ethanol (3200mL), heated to 78°C and stirred at 78°C for 1 hour (until the reaction mixture is completely clear), turn off the heating, slowly lower the temperature to 20°C with stirring, and continue stirring at 20°C for 12 hours , A large amount of solids precipitated during stirring.
  • the reaction mixture was directly concentrated under reduced pressure, and the resulting residue was dissolved in ethyl acetate (1000 mL), washed with 1M dilute hydrochloric acid (1000 mL ⁇ 2), washed with water (1000 mL ⁇ 2), and washed with saturated brine (1000 mL) , The organic phase was dried with anhydrous sodium sulfate, filtered, and the filtrate was decompressed to remove the solvent.
  • the residue obtained was added to ethanol (360 mL), stirred for 30 minutes, filtered, the filter cake was washed with ethanol (100 mL), the filter cake was collected, and the solvent was removed under reduced pressure.
  • the product was added to ethanol (150 mL) and acetic acid In the ethyl ester (150 mL) mixed solvent, the reaction mixture was heated to 78°C, and stirring was continued at 78°C until the reaction solution was clear. The heating was turned off, the reaction solution was naturally cooled to 20°C under stirring, and stirring was continued for 12 hours, during which solids precipitated out, filtered, and the filter cake was washed with ethanol (50 mL ⁇ 2). The filter cake was collected, and the solvent was removed under reduced pressure. After the product was obtained, the product was added to a mixed solvent of ethanol (60 mL) and ethyl acetate (60 mL).
  • the reaction mixture was stirred at 20°C for 2 hours and filtered.
  • the filter cake was washed with ethanol ( 10 mL) was washed, the filter cake was collected, and the solvent was removed under reduced pressure, followed by vacuum drying for 6 hours (temperature 40-45° C., pressure: -0.08 MPa) to obtain crystal form A of target compound 1.
  • amorphous compound 1 Approximately 175 mg of amorphous compound 1 was added to 1.0 ml of ethanol, first dissolved by ultrasound, and then a large amount of white solid was precipitated by ultrasound. After stirring at room temperature for 3 hours, the solid obtained by centrifugation was the crystal form A of compound 1.
  • the moisture absorption and weight gain of compound 1 crystal form A at 25° C. and 80% RH is 0.05%, which is less than 0.2%, with no or almost no hygroscopicity.
  • the crystal form A of compound 1 has good stability in solvents such as methyl tert-butyl ether, toluene, water, and mixed solvents of alcohol and water.
  • the crystal form A of compound 1 has good stability under high temperature, high humidity or strong light conditions.
  • the crystal form A of compound 1 has good stability under accelerated conditions of 40°C/relative humidity 75%.
  • the crystal form A of compound 1 has good stability under long-term conditions of 25°C/relative humidity 60%.
  • Test Example 1 Inhibitory activity of compound 1 on phosphodiesterase 4B subtype (PDE4B enzyme)
  • the expression of AMP/GMP is determined by fluorescence polarization, that is, the binding of AMP/GMP antibody is traced to indicate the activity of the enzyme.
  • Experimental buffer solution 10mM Tris-HCl buffer solution (Tris-HCl) (pH 7.5), 5mM MgCl 2 , 0.01% polyoxyethylene lauryl ether (Brij 35), 1mM dithiothreitol (DTT) , And 1% DMSO.
  • the fluorescence polarization signal was calculated according to the AMP/GMP standard curve and the% enzyme activity relative to the DMSO control by Excel software, converted into nM. Curve fitting uses GraphPad Prism (drawing medical icons).
  • Compound 1 exhibited excellent in vitro activity for inhibiting phosphodiesterase 4B subtype (PDE4B).
  • Test Example 2 Evaluation of the inhibition of TNF ⁇ production in human peripheral blood mononuclear cells (hPBMC) in vitro
  • Compound 1 has inhibitory activity on lipopolysaccharide (LPS)-induced TNF ⁇ production in human peripheral blood mononuclear cells.
  • LPS lipopolysaccharide
  • PBMC cells were seeded into a 96-well plate of cell culture grade at a density of 100,000 cells/100 ⁇ L/well, and the cell culture medium was RPMI-1640 with 10% serum. Incubate for 2 hours at 37°C in a 5% CO 2 incubator. Add 16.8 ⁇ L/well of the test compound to the cells and incubate in a 37°C, 5% CO 2 incubator for 60 minutes, then add 16.8 ⁇ L/well of LPS to the cells in a 37°C, 5% CO 2 incubator Incubate for 18 hours, and the final DMSO concentration is 0.1%.
  • the compound dose is diluted gradually
  • compound 1 was diluted from the storage concentration with 100% DMSO to 1.5 mM.
  • the diluted compound was used as the first point and diluted 3 times with 100% DMSO for 9 points.
  • the third step is 125-fold dilution with serum-free medium. At this time, the concentration of DMSO is 0.8%. Then transfer 16.8 ⁇ L of the compound that has been diluted with medium to a 100 ⁇ L cell plate.
  • the cell plate After the compound is added, the cell plate is placed in a 37°C, 5% CO 2 incubator and incubated for 1 hour.
  • the first step is to dilute LPS with ultrapure water to a storage concentration of 1 mg/mL.
  • the storage concentration of LPS is diluted to 1 ⁇ g/mL with serum-free medium.
  • the third step is 1666.666 times dilution with serum-free medium.
  • the final concentration of DMSO is 0.1%. After adding LPS, place the cell plate in a 37°C, 5% CO 2 incubator and incubate 18 Hours.
  • Inhibition rate (1-(original value-HPE average)/(ZPE average-HPE average))*100
  • ZPE is: 0% inhibition (75pg/mL LPS, 0.1% DMSO), and HPE: 100% inhibition (without LPS, 0.1% DMSO).
  • IC50 calculation formula is: using a 4-parameter logistic dose-response equation, the concentration of the tested compound and the inhibition rate (%) are plotted, and the compound concentration required for 50% inhibition (IC 50 ) is determined.
  • Compound 1 exhibited excellent in vitro activity to inhibit TNF ⁇ production in hPBMC.
  • Test Example 3 PMA-induced CD-1 mouse ear edema in vivo model
  • Inflammatory edema also known as tissue edema
  • tissue edema is edema caused by exudate accumulated in the interstitial spaces caused by inflammation.
  • PMA protein kinase C
  • PKC Protein Kinase C
  • the purpose of this experiment is to investigate the therapeutic effect of compound 1 on PMA-induced CD-1 mouse ear edema model, so as to provide preclinical pharmacodynamic information for subsequent clinical studies.
  • CD-1 mice were sorted according to ear thickness and body weight. After removing 4 animals with large mean values, they were randomly divided into 6 normal control groups and treatment groups, with 10 mice in each treatment group. Take 10 ⁇ L of each with a concentration of 0.25mg/mL PMA and smear them on the front and back of the right ear of the mouse.
  • the first group is normal mice without any treatment; the second group is given a vehicle; the third, fourth, and fifth groups are given compound 1 at doses of 0.1 mg/ear, 0.3 mg/ear and 1 mg/ear, respectively , 30 minutes before PMA induction and 15 minutes after PMA induction, the drug was applied to the right ear of the mouse.
  • NA stands for “None”
  • BID stands for "Dosing twice a day”.
  • mice 10 hours after PMA induction Anesthetized the mice 10 hours after PMA induction to measure the thickness of the right ear. After measuring the thickness of the right ear, it was euthanized immediately, and the ear pieces were collected and weighed.
  • the experimental data are expressed by Mean ⁇ SEM, and the degree of ear swelling and ear weight are expressed by One-way ANOVA, and p ⁇ 0.05 is considered as a significant difference.
  • the ear thickness increased by 0.300-0.400mm, which is much higher than the normal swelling range -0.010 to 0.002mm, and the ear weight increased by 28.8mg on average, indicating that the establishment of the ear edema model was very successful.
  • Test compound 1 was significantly reduced at 0.1 mg/ear, 0.3 mg/ear and 1 mg/ear. At 10 hours, the degree of edema in mice with ear edema at the three doses and the ear edema inhibition rate were 22%, respectively. 39% and 88%, the ear weight gain inhibition rate was 39%, 46% and 85% respectively (compared with the vehicle control group, p values were all ⁇ 0.0001), and showed good dose-effect relationship.
  • Compound 1 in the three dose groups of 0.1, 0.3 and 3 mg/ear has a significant improvement effect on PMA-induced ear edema symptoms, and can significantly inhibit ear weight gain, and the three dose groups all show a good dose-effect relationship.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

化合物(1)的晶型、其溶剂化物、其溶剂化物的晶型以及它们的制备方法,还包括所述晶型在制备治疗与TNFα相关疾病药物中的应用。

Description

一种苯并咪唑-2-酮类化合物的晶型、溶剂化物、溶剂化物的晶型及它们的制备方法
本申请主张如下优先权:
CN201910602489.2,2019.07.04;
CN202010426911.6,2020.05.19。
技术领域
本发明涉及化合物1的晶型、其溶剂化物、其溶剂化物的晶型以及它们的制备方法,还包括所述晶型在制备治疗与TNFα相关疾病药物中的应用。
背景技术
肿瘤坏死因子(TNFα)是一种主要由单核噬菌细胞应答免疫刺激物时释放的细胞因子。TNFα能够促进细胞的分化、募集、增殖和蛋白质降解等大多数过程。在低水平下,TNFα具有防止传染物、肿瘤和组织损伤的保护作用。但TNFα释放过多也会引起疾病,如给予哺乳动物或人TNFα时,会引起或加重炎症、发烧、心血管作用、出血、凝血以及与急性感染和休克状态相类似的急性反应。动物体或人体内产生过量的或不受控制的TNFα常提示患有如下疾病:内毒素血症和/或中毒休克综合症、恶病质、成人呼吸紧张综合症、癌症(如实体瘤和血液性肿瘤)、心脏病(如充血性心力衰竭)、病毒感染、遗传疾病、炎性疾病、变应性疾病或自身免疫疾病。
癌症是具有特别破坏性的疾病,血液中TNFα水平的提高预示存在患有癌症或癌症扩散的危险。通常,癌细胞不能在健康主体的循环系统中存活,其中一个原因在于血管内壁是瘤细胞外渗的屏障。研究表明,内皮细胞上的ELAM-1能介导促进结肠癌细胞黏附在用细胞因子处理的内皮上。
环腺苷酸(cAMP)在许多疾病和病症中起作用。发炎时白细胞中cAMP浓度的升高抑制了白细胞的激活,随后释放出包括TNFα和NF-κB等炎症调控因子。cAMP水平提高也会导致呼吸道平滑肌的松弛。
cAMP失活的主要细胞机制是由于被称为环核苷酸磷酸二酯酶(PDE)的一族同工酶破坏了cAMP。已知有11个PDE家族成员。迄今,已证实抑制PDE4酶对抑制炎症介质的释放及对松弛呼吸道平滑肌特别有效,因此PDE4酶已成为热门的药物靶点之一。依据不同的基因编码,PDE-4家族可以分为4个亚型(PDE-4A、B、C、D)。其中,PDE-4A、PDE-4B和PDE-4D在炎症细胞(如B细胞、T细胞和中性粒细胞等)中的表达强于PDE-4C。抑制PDE4酶,导致cAMP水平的升高,从而调节TNFα水平,达到治疗疾病目的。
发明内容
本发明提供了化合物1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:11.91±0.20°、19.36±0.20°、23.17±0.2°。
Figure PCTCN2020100129-appb-000001
在本发明的一些方案中,上述化合物1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:11.26±0.20°、11.91±0.20°、12.91±0.20°、14.27±0.20°、19.36±0.20°、22.26±0.20°、23.17±0.20°、24.97±0.20°。
在本发明的一些方案中,上述化合物1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:11.26±0.20°、11.91±0.20°、12.91±0.20°、14.27±0.20°、15.83±0.20°、17.53±0.20°、19.36±0.20°、20.33±0.20°、22.26±0.20°、23.17±0.20°、24.97±0.20°、26.50±0.20°。
在本发明的一些方案中,上述化合物1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:11.26°、11.91°、12.91°、14.27°、15.83°、17.53°、19.36°、20.33°、22.26°、22.59°、23.17°、24.97°、26.50°、29.46°。
本发明的一些方案中,上述化合物1的A晶型的XRPD图谱解析数据如表1所示:
表1
编号 2θ角(°) 相对强度(%) 编号 2θ角(°) 相对强度(%)
1 11.26 51.13 8 20.33 29.34
2 11.91 99.15 9 22.26 48.06
3 12.91 51.24 10 22.59 48.31
4 14.27 33.57 11 23.17 72.63
5 15.83 28.98 12 24.97 50.67
6 17.53 18.87 13 26.50 22.08
7 19.36 100.00 14 29.46 19.25
在本发明的一些方案中,上述化合物1的A晶型,其XRPD图谱如图1所示。
在本发明的一些方案中,上述的化合物1的A晶型,其差示扫描量热曲线在147.0±3.0℃处具有吸热峰的起始点。
在本发明的一些方案中,上述的化合物1的A晶型,其DSC图谱如图2所示。
在本发明的一些方案中,上述的化合物1的A晶型,其热重分析曲线在140.0±3.0℃时失重达0.70%。
在本发明的一些方案中,上述的化合物1的A晶型,其TGA图谱如图3所示。
本发明还提供了式(I-1)所示的溶剂化物
Figure PCTCN2020100129-appb-000002
n选自0.1~1.5。
在本发明的一些方案中,上述溶剂化物,其中,n选自0.1、0.2、0.3、0.4、0.5、0.6、0.7、0.8、0.9、1、1.1、1.2、1.3、1.4和1.5。
在本发明的一些方案中,上述溶剂化物,其中,n为0.5。
本发明还提供了式(I-1-1)所示溶剂化物的B晶型,其中,n为0.5,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:6.84±0.20°、8.90±0.20°、23.00±0.20°。
Figure PCTCN2020100129-appb-000003
在本发明的一些方案中,上述溶剂化物的B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:6.84±0.20°、8.90±0.20°、11.27±0.20°、12.75±0.20°、16.15±0.20°、17.54±0.20°、22.06±0.20°、23.00±0.20°。
在本发明的一些方案中,上述溶剂化物的B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:6.84±0.20°、8.90±0.20°、11.27±0.20°、12.75±0.20°、16.15±0.20°、17.54±0.20°、19.17±0.20°、19.70±0.20°、20.41±0.20°、22.06±0.20°、23.00±0.20°、25.95±0.20°。
在本发明的一些方案中,上述溶剂化物的B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:6.84°、8.90°、11.27°、12.75°、13.29°、14.94°、16.15°、17.54°、17.93°、19.17°、19.70°、20.41°、20.79°、22.06°、22.72°、23.00°、23.86°、25.95°、27.82°、28.45°、30.14°、32.87°。
本发明的一些方案中,上述溶剂化物的B晶型的XRPD图谱解析数据如表2所示:
表2
编号 2θ角(°) 相对强度(%) 编号 2θ角(°) 相对强度(%)
1 6.84 100.00 12 20.41 21.30
2 8.90 49.09 13 20.79 23.81
3 11.27 25.33 14 22.06 20.09
4 12.75 27.95 15 22.72 15.23
5 13.29 4.72 16 23.00 74.25
6 14.94 14.38 17 23.86 8.77
7 16.15 26.47 18 25.95 20.40
8 17.54 32.17 19 27.82 6.56
9 17.93 32.65 20 28.45 9.53
10 19.17 15.26 21 30.14 4.13
11 19.70 19.96 22 32.87 1.69
在本发明的一些方案中,上述溶剂化物的B晶型,其XRPD图谱如图5所示。
在本发明的一些方案中,上述溶剂化物的B晶型,其差示扫描量热曲线在77.5±3.0℃处具有吸热峰的起始点。
在本发明的一些方案中,上述溶剂化物的B晶型,其DSC图谱如图6所示。
在本发明的一些方案中,上述的溶剂化物的B晶型,其热重分析曲线在80.0±3.0℃时失重达10.46%。
在本发明的一些方案中,上述溶剂化物的B晶型,其TGA图谱如图7所示。
本发明还提供了上述化合物1的A晶型、上述溶剂化物或上述溶剂化物的B晶型在制备治疗与TNFα相关疾病药物中的应用。
技术效果
本发明化合物1的A晶型性质稳定,吸湿性小,成药前景良好;本发明化合物1展现出优异的抑制磷酸二酯酶4B亚型(PDE4B)的体外活性;本发明化合物1在hPBMC中展现出优异的抑制TNFα生成的体外活性;本发明化合物1在0.1,0.3和3mg/耳朵三个剂量组对PMA诱导的小鼠耳水肿症状均有显著的改善作用,均可显著抑制耳增重,并且三个剂量组均呈现良好的量效关系。
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 PCTCN2020100129-appb-000004
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。
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。
化合物依据本领域常规命名原则或者
Figure PCTCN2020100129-appb-000005
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 PCTCN2020100129-appb-000006
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
本发明含量测定方法
仪器型号:安捷伦1260高效液相色谱仪
色谱条件具体参数如下:
色谱柱:ACE Excel 3 super C18□4.6*150mm id□,P.N.:EXL-1111-1546U
柱温:35℃
流速:0.8mL/min
检测波长:230nm
进样体积:5μL
运行时间:15min
流动相A:0.04%三氟乙酸水溶液(V/V)
流动相B:100%乙腈
稀释剂:乙腈:纯水=50:50(V/V)
洗针液:乙腈:纯水=50:50(V/V)
梯度洗脱程序:
时间(min) 流动相A(%) 流动相B(%)
0.00 60 40
15.00 60 40
附图说明
图1为化合物1的A晶型的Cu-Kα辐射的XRPD谱图。
图2为化合物1的A晶型的DSC谱图。
图3为化合物1的A晶型的TGA谱图。
图4为化合物1的A晶型的DVS谱图。
图5为化合物1的B晶型的Cu-Kα辐射的XRPD谱图。
图6为化合物1的B晶型的DSC谱图。
图7为化合物1的B晶型的TGA谱图。
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:化合物1的无定型的制备
Figure PCTCN2020100129-appb-000007
步骤1:化合物4的合成
室温和氮气保护下,将化合物2(15.00g,94.29mmol)溶于N,N–二甲基甲酰胺(150mL)中,随后依 次加入化合物3(25.77g,94.29mmol)和碳酸钾(19.55g,141.43mmol),反应混合物加热至70℃并搅拌反应16小时。反应完毕后,冷却至室温,加入饱和食盐水(400mL),用乙酸乙酯(200mL×3)萃取。合并有机相,用饱和食盐水(200mL×3)洗涤,无水硫酸钠干燥,过滤,滤液减压除去溶剂,所得残余物经柱层析(洗脱剂:乙酸乙酯/石油醚=0:1至2:3,体积比)分离纯化得到目标化合物4。 1HNMR(400MHz,CDCl 3)δ:8.52(d,J=6.5Hz,1H),7.90(dd,J=8.9,3.0Hz,1H),7.20–7.15(m,1H),6.96–6.93(m,1H),6.90–6.85(m,2H),6.82–6.78(m,1H),5.20–5.15(m,1H),4.09–4.01(m,2H),3.86(s,3H),3.65(dd,J=14.7,8.1Hz,1H),3.48(dd,J=14.7,4.8Hz,1H),2.80(s,3H),1.45(t,J=6.9Hz,3H)。
步骤2:化合物5的合成
室温和氮气保护下,将化合物4(16.00g,38.79mmol)溶于乙醇(128mL)与乙酸乙酯(32mL)的混合溶剂中,随后加入湿钯碳(5.00g,纯度:10%),氢气置换三次,反应混合物在室温和氢气(30psi)保护下并搅拌反应16小时。反应完毕后,反应混合物过滤,用乙酸乙酯(100mL×3)洗涤滤饼,滤液减压除去溶剂,所得残余物经过柱层析(洗脱剂:乙酸乙酯/石油醚=0:1至3:2,体积比)分离纯化得到目标化合物5。 1H NMR(400MHz,DMSO-d 6)δ:7.07(d,J=1.2Hz,1H),6.91–6.84(m,2H),6.36(dd,J=10.8,2.9Hz,1H),6.30(dd,J=8.6,5.8Hz,1H),6.09(td,J=8.6,2.9,1H),4.99(s,2H),4.96(d,J=9.3Hz,1H),4.74(td,J=9.4,3.8Hz,1H),4.02–3.96(m,2H),3.73–3.67(m,4H),3.39–3.36(m,1H),3.01(s,3H),1.30(t,J=7.0Hz,3H)。
步骤3:化合物1的无定型的合成
室温下,将化合物5(12.2g,31.90mmol)溶于乙酸乙酯(120mL)中,随后加入羰基二咪唑(15.52g,95.70mmol),反应混合物在室温下搅拌反应16小时。反应完毕后,冷却至室温,加入1M稀盐酸(5mL)和水(50mL),用乙酸乙酯(80mL×3)萃取。合并有机相,用饱和食盐水(50mL×2)洗涤,无水硫酸钠干燥,过滤,滤液减压除去溶剂,所得残余物经过柱层析分离(洗脱剂:乙酸乙酯/石油醚=0:1至3:2,体积比),再经制备HPLC(流动相:乙腈/水,中性体系)纯化,真空冻干得到目标化合物1,即为无定型。MS–ESI m/z:409.0[M+H] +1H NMR(400MHz,CD 3OD)δ:7.11–7.08(m,2H),7.06–7.04(m,1H),6.93(d,J=8.4Hz,1H),6.83(dd,J=8.5,2.4Hz,1H),6.79–6.74(m,1H),6.00(dd,J=10.5,3.9Hz,1H),4.58(dd,J=14.8,10.8Hz,1H),4.08–3.96(m,3H),3.80(s,3H),2.94(s,3H),1.35(t,J=7.0Hz,3H)。
实施例2:化合物1的A晶型的制备
Figure PCTCN2020100129-appb-000008
步骤1:化合物4的合成
20℃–30℃下,将化合物2(200.05g,1.26mol)溶于N,N–二甲基乙酰胺(2000mL)中,随后加入化合物3(516.45g,1.89moL),再缓慢滴加二异丙基乙胺(325.00g,2.52mol)至上述溶液中(滴加时间约20分钟),滴加完毕后,反应混合物加热至110℃–120℃并在110℃–120℃下搅拌反应16小时。反应完毕后,冷却至15℃,反应液缓慢倒入到冰水中(10500mL)中,有大量固体析出,过滤,滤饼用乙醇(200mL)洗涤,收集滤饼,减压除去溶剂,然后将样品加入到乙醇(1500mL)中,在15℃下打浆搅拌16小时,过滤,滤饼用乙醇(200mL)洗涤,滤饼经减压除去溶剂,得到目标化合物4。 1HNMR(400MHz,CDCl 3)δ:8.52(d,J=6.5Hz,1H),7.90(dd,J=8.9,3.0Hz,1H),7.20–7.15(m,1H),6.96–6.93(m,1H),6.90–6.85(m,2H),6.82–6.78(m,1H),5.20–5.15(m,1H),4.09–4.01(m,2H),3.86(s,3H),3.65(dd,J=14.7,8.1Hz,1H),3.48(dd,J=14.7,4.8Hz,1H),2.80(s,3H),1.45(t,J=6.9Hz,3H)。
步骤2:化合物5的合成
20℃–25℃和氮气保护下,将化合物4(122.83g,0.30mol)溶于二氯甲烷(500mL)和乙酸乙酯(500mL)的混合溶剂中,随后加入湿钯碳(7.50g,纯度:10%),氢气置换三次,反应混合物在25℃–35℃和氢气(25–35psi)氛围下搅拌反应16小时(平行投了四批,合并处理)。反应完毕后,四批合并反应液经硅藻土过滤,滤饼用二氯甲烷(200mL)洗涤。滤液减压旋干,得到粗品。粗品加入到乙醇(3200mL)中,升温至78℃并在78℃下搅拌1小时(待反应混合液至完全澄清),关闭加热在搅拌下缓慢降温至20℃并在20℃下继续搅拌12小时,搅拌期间有大量固体析出。反应混合物过滤,滤饼用乙醇(200mL)洗涤,收集滤饼,减压旋干后得到406.15g产品,取329.50g产品用二氯甲烷(2000mL)溶解,经过柱层析分离(洗脱剂:二氯甲烷/甲醇=1:0,体积比),得到化合物5。 1H NMR(400MHz,DMSO-d 6)δ:7.07(d,J=1.2Hz,1H),6.91–6.84(m,2H),6.36(dd,J=10.8,2.9Hz,1H),6.30(dd,J=8.6,5.8Hz,1H),6.09(td,J=8.6,2.9,1H),4.99(s,2H),4.96(d,J=9.3Hz,1H),4.74(td,J=9.4,3.8Hz,1H),4.02–3.96(m,2H),3.73–3.67(m,4H),3.39–3.36(m,1H),3.01(s,3H),1.30(t,J=7.0Hz,3H)。
步骤3:化合物1的A晶型合成
20℃和氮气保护下,将化合物5(175.09g,0.46moL)溶于丙酮(1800mL)中,随后加入羰基二咪唑(163.32g,1.01mol),反应混合物在15℃–25℃下搅拌反应16小时。反应完毕后,反应混合物直接减压浓缩,所得残余物加入乙酸乙酯(1000mL)溶解,用1M稀盐酸(1000mL×2)洗涤,用水(1000mL×2)洗涤,用饱和食盐水(1000mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液减压除去溶剂。所得残余物加入到乙醇(360mL)中,搅拌30分钟,过滤,滤饼用乙醇(100mL)洗涤,收集滤饼,再减压除去溶剂,得到产品后,将产品加入到乙醇(150mL)和乙酸乙酯(150mL)混合溶剂中,反应混合物加热至78℃,并在78℃下继续搅拌直至反应液澄清。关闭加热,在搅拌下反应液自然冷却至20℃,并继续搅拌12小时,期间有固体析出,过滤,滤饼用乙醇(50mL×2)洗涤。收集滤饼,再减压除去溶剂,得到产品后,将产品加入到乙醇(60mL)和乙酸乙酯(60mL)混合溶剂中,反应混合物在20℃下搅拌2小时,过滤,滤饼用乙醇(10mL)洗涤,收集滤饼,再经减压除去溶剂后,真空干燥6小时(温度40–45℃,压力:–0.08MPa)得到目标化合物1的A晶型。MS–ESI m/z:409.0[M+H] +1H NMR(400MHz,CD 3OD)δ:7.11–7.08(m,2H),7.06–7.04(m,1H),6.93(d,J=8.4Hz,1H),6.83(dd,J=8.5,2.4Hz,1H),6.79–6.74(m,1H),6.00(dd,J=10.5,3.9Hz,1H),4.58(dd,J=14.8,10.8Hz,1H),4.08–3.96(m,3H),3.80(s,3H),2.94(s,3H),1.35(t,J=7.0Hz,3H)。
实施例3:化合物1的A晶型的制备
将约175mg无定型化合物1加入到1.0毫升乙醇中,先超声溶解,然后继续超声析出大量白色固体,室温悬浮搅拌3小时后,离心分离得到的固体,即为化合物1的A晶型。
实施例4:化合物1的溶剂化物的B晶型的制备
Figure PCTCN2020100129-appb-000009
将约24mg无定型化合物1加入到0.2毫升间二甲苯中,室温悬浮搅拌约2天,离心分离得到的固体,即为B晶型。 1H NMR(400MHz,CDCl 3)δ:8.21(s,1H),7.15(dd,J=7.4,7.4Hz,0.5H),7.03(dd,J=2.3Hz,1H),7.02(dd,J=8.3,2.3Hz,1H),7.00–6.97(m,2.5H),6.84–6.78(m,3H),5.76(dd,J=9.5,4.2Hz,1H),4.71(dd,J=14.8,9.5Hz,1H),4.06(q,J=7.0Hz,2H),3.84(dd,J=15.1,4.8Hz,1H),3.84(s,3H),2.78(s,3H),2.32(s,3H),1.44(t,J=7.0Hz,3H)。
实验例1:化合物1的A晶型的引湿性研究
实验材料:
SEM Advantage–1动态蒸汽吸附仪。
实验方法:
取化合物1的A晶型10~30mg置于DVS样品盘内进行测试。
实验结果:
化合物1的A晶型的DVS谱图如图4所示,△W=0.05%。
实验结论:
化合物1的A晶型在25℃和80%RH下的吸湿增重为0.05%,小于0.2%,无或几乎无有引湿性。
实验例2:化合物1的A晶型在不同溶剂中的稳定性实验
称取每份约15mg化合物1的A晶型17份,分别加入适量下表中的单一或混合溶剂,在室温或50℃条件下悬浮搅拌2周,离心收集固体,XRPD检测其晶型状态。结果见表3。
表3化合物1的A晶型在不同溶剂中的稳定性实验
编号 溶剂(体积比) 温度 状态(2周后) 晶型
1 甲基叔丁基醚 室温 悬浊液 A晶型
2 甲苯 室温 悬浊液 A晶型
3 室温 悬浊液 A晶型
4 丙酮:甲苯(2:1) 室温 悬浊液 A晶型
5 乙酸乙酯:间二甲苯(2:1) 室温 悬浊液 A晶型
6 乙腈:水(3:1) 室温 悬浊液 A晶型
7 二氯甲烷:间二甲苯(1:1) 室温 悬浊液 A晶型
8 异丙醇:水(98:2) 室温 悬浊液 A晶型
9 异丙醇:水(95:5) 室温 悬浊液 A晶型
10 异丙醇:水(92:8) 室温 悬浊液 A晶型
11 异丙醇:水(85:5) 室温 悬浊液 A晶型
12 2-甲基四氢呋喃:正辛烷(4:1) 50℃ 悬浊液 A晶型
13 乙醇:水(3:1) 50℃ 悬浊液 A晶型
14 1,4-二氧六环:正庚烷(1:1) 50℃ 悬浊液 A晶型
15 甲基异丁基酮:正己烷(2:1) 50℃ 悬浊液 A晶型
16 2-丁醇:间二甲苯(2:1) 50℃ 悬浊液 A晶型
17 二甲亚砜:水(2:1) 50℃ 悬浊液 A晶型
实验结论:化合物1的A晶型在甲基叔丁基醚、甲苯、水和醇类溶剂与水的混合溶剂等溶剂中均具有良好的稳定性。
实验例3:化合物1的A晶型在高温、高湿及强光条件下的固体稳定性实验
依据《原料药与制剂稳定性试验指导原则》(中国药典2015版四部通则9001),考察化合物1的A晶型在高温(60℃,敞口)、高湿(室温/相对湿度92.5%,敞口)及强光照(5000±500Lux,90μw/cm 2,封口)条件下的稳定性。
称取化合物1的A晶型1.5g,放入敞口的表面皿中,摊成薄薄一层。高温及高湿条件下放置的样品放入保干器中考察,于第5天、10天和30天取样检测,检测结果与0天的初始检测结果进行比较;强光照条件下放置的样品盖上石英玻璃盖,于第5天和10天取样检测,检测结果与0天的初始检测结果进行比较。实验结果见下表4所示。
表4化合物1的A晶型在高温、高湿及强光条件下的固体稳定性实验结果
Figure PCTCN2020100129-appb-000010
结论:化合物1的A晶型在高温,高湿或强光照条件下均具有良好的稳定性。
实验例4:化合物1的A晶型在加速条件下的固体稳定性实验
依据《原料药与制剂稳定性试验指导原则》(中国药典2015版四部通则9001),考察化合物1的A晶型在高温和高湿加速条件(40℃/相对湿度75%,密封)下的稳定性。
称取化合物1的A晶型约1.5g,装入双层低密度聚乙烯袋,每层低密度聚乙烯袋分别扎扣密封,再放入铝箔袋中并热封,于第1月、2月、3月和6月取样检测,检测结果与0天的初始检测结果进行比较。实验结果见下表5所示。
表5化合物1的A晶型在加速条件(40℃/相对湿度75%,密封)下的固体稳定性实验结果
Figure PCTCN2020100129-appb-000011
结论:化合物1的A晶型在40℃/相对湿度75%加速条件下具有良好的稳定性。
实验例5:化合物1的A晶型在长期条件下的固体稳定性实验
依据《原料药与制剂稳定性试验指导原则》(中国药典2015版四部通则9001),考察化合物1的A晶型在长期条件(25℃/相对湿度60%,密封)下的稳定性。
称取化合物1的A晶型约1.5g,装入双层低密度聚乙烯袋,每层低密度聚乙烯袋分别扎扣密封,再放入铝箔袋中并热封,于第3月、6月取样检测,检测结果与0天的初始检测结果进行比较。实验结果见下表6所示。
表6化合物1的A晶型在长期条件(25℃/相对湿度60%,密封)下的固体稳定性实验结果
Figure PCTCN2020100129-appb-000012
结论:化合物1的A晶型在25℃/相对湿度60%长期条件下具有良好的稳定性。
测试例1:化合物1对磷酸二酯酶4B亚型(PDE4B酶)的抑制活性
该生物实验是根据荧光偏振测定AMP/GMP表达,即示踪AMP/GMP抗体结合来表示酶的活性。
试剂:
实验缓冲溶液:10mM三羟甲基氨基甲烷-盐酸缓冲溶液(Tris-HCl)(pH 7.5),5mM MgCl 2,0.01%聚氧乙烯月桂醚(Brij 35),1mM二硫苏糖醇(DTT),和1%DMSO。
酶:重组人源PDE4B(基因登录号NM_002600;氨基酸305端)用N端GST标签在Sf9昆虫细胞中的杆状病毒来表达。MW=78kDa。
酶作用物:1μM cAMP
检测:
Figure PCTCN2020100129-appb-000013
抗体和AMP2/GMP2 AlexaFluor633示踪。
操作步骤:
1.将重组人源PDE4B酶和酶作用物(1μM cAMP)分别溶解到新鲜制备的实验缓冲液中;
2.将上述PDE4B酶缓冲溶液转移到反应孔中;
3.通过声学技术(回声550毫微升范围)将100%DMSO溶解的化合物1加到PDE4B酶缓冲溶液反应孔中,并在室温下孵育10分钟;
4.然后,将酶作用物缓冲溶液加到上述反应孔中以启动反应;
5.在室温下孵育1小时;
6.添加检测混合物(
Figure PCTCN2020100129-appb-000014
抗体和AMP2/GMP2 AlexaFluor633示踪)以终止反应,并在缓慢混合下孵育90分钟。荧光偏振测定范围是Ex/Em=620/688。
数据分析:
荧光偏振信号根据AMP/GMP标准曲线和通过Excel软件计算相对DMSO对照的%酶活性,换算成nM。曲线拟合使用GraphPad Prism(绘制医学图标)。
表7本发明化合物1体外筛选试验结果
化合物 IC 50(nM) *
化合物1 26.2
*三复孔,取平均值。
结论:
化合物1展现出优异的抑制磷酸二酯酶4B亚型(PDE4B)的体外活性。
测试例2:体外在人外周血单个核细胞(hPBMC)抑制TNFα生成作用的评价
实验目的:
化合物1对脂多糖(LPS)诱发人外周血单个核细胞的TNFα生成的抑制活性。
实验操作步骤:
1.PBMC实验
PBMC细胞以100000个/100μL/孔的密度种入细胞培养级别的96孔板中,细胞培养基是加10%血清的RPMI-1640。在37℃,5%CO 2培养箱中培养2个小时。在细胞里加入16.8μL/孔的待测化合物后在37℃,5%CO 2培养箱中培养60分钟,后在细胞里加入16.8μL/孔的LPS在37℃,5%CO 2培养箱中培养18小时,最终DMSO浓度为0.1%。
2.化合物剂量梯度稀释
第一步将化合物1从储藏浓度用100%的DMSO稀释到1.5mM。第二步将稀释过的化合物作为第一个点用100%DMSO 3倍稀释9个点。第三步用不含有血清的培养基125倍稀释,此时DMSO的浓度是0.8%。然后转16.8μL已经用培养基稀释好的化合物到100μL的细胞板里。
加好化合物后将细胞板放入37℃,5%CO 2培养箱中孵育1个小时。
3.LPS稀释
第一步将LPS用超纯水稀释到储藏浓度1mg/mL。第二步储藏浓度的LPS用不含血清的培养基稀释到1μg/mL。第三步用不含有血清的培养基1666.666倍稀释。然后转16.8μL已经用培养基稀释好的LPS到116.8μL的细胞板里,此时DMSO终浓度是0.1%,加好LPS后将细胞板放入37℃,5%CO 2培养箱中孵育18个小时。
4.ELISA实验
1)将TNF-α抗体在包被液中稀释至1倍体积,然后每孔100μL加到96孔高结合性能的板子中,板子用膜封住放到4℃冰箱中18个小时。
2)配制2000mL清洗缓冲液至1倍体积备用。
3)包被的板子过夜后,将包被液倒掉,用清洗缓冲液每孔300μL/孔清洗3遍。
4)板子清洗过后加每孔200μL的封闭缓冲液,板子用膜封住。放到25℃孵育箱中孵育一个小时。
5)将孵育18个小时的细胞板子放到离心机中离心,温度:25℃,转速:2000转,时间:10分钟,升速:9,降速:1。离心后取每孔100μL细胞上清到3599细胞板中,后放到4℃冰箱备用。
6)将细胞上清用封闭缓冲液稀释40倍放到4℃冰箱待用,后配制标准品也放置4℃冰箱备用。
7)封闭完成后,将封闭液倒掉,用清洗缓冲液每孔300μL清洗3遍。
8)将稀释好的细胞上清样品以及标准品加到ELISA板子中,板子用膜封住。后放到25℃孵育箱中孵育两个小时。
9)将板中液体倒掉,用清洗缓冲液每孔300μL清洗5遍。
10)配制抗体,并每孔加入100μL,用封板膜封板。后放到25℃孵育箱中孵育一个小时。
11)将板中液体倒掉,用清洗缓冲液每孔300μL清洗7遍。
12)配制显色液,每孔加100μL。后避光放到25℃孵育箱中孵育半个小时。
13)每孔加50μL终止液,离心,温度:25℃,转速:1000转,时间:1分钟,升速:9,降速:9。
14)离心后30分钟内在Envision上读数,设置为吸收光450减去吸收光570的值为最终的原始数据使用值。
5.数据处理
根据原始数据计算抑制率,抑制率计算公式为:
抑制率=(1-(原始值-HPE平均值)/(ZPE平均值-HPE平均值))*100
其中ZPE为:0%抑制(75pg/mL LPS,0.1%DMSO),HPE为:100%抑制(不含LPS,0.1%DMSO)。
用XLfit统计软件进行数据分析。IC50的计算公式为:用4参数logistic剂量响应方程,绘制了被测化合物的浓度和抑制率(%),并确定了50%抑制所需的化合物浓度(IC 50)。
表8本发明化合物1在hPBMC中对TNFα生成的抑制活性结果
化合物 IC 50(nM) *
化合物1 63.92
*三复孔,取平均值。
实验结论:
化合物1在hPBMC中展现出优异的抑制TNFα生成的体外活性。
测试例3:PMA诱导的CD-1小鼠耳水肿体内模型
实验目的:
炎性水肿,又称为组织水肿,是因为炎症而造成的渗出液聚集在组织间隙而产生的水肿。佛波醇12-十四酸酯13-乙酸酯(Phorbol 12-Myristate 13-Acetate,PMA)局部给药于小鼠耳朵时,可引起由蛋白激酶C(Protein Kinase C,PKC)介导明显的炎症反应,从而引发一系列类似于人特应性皮炎(Atopic Dermatitis,AD)的症状。在临床前评价治疗AD候选化合物的过程中,PMA诱导小鼠耳水肿动物模型通常被用来评价其有效性。
本实验目的是考察化合物1在PMA诱导CD-1小鼠耳水肿模型上的治疗效果,从而为之后的临床研究提供临床前药效学相关信息。
实验方法:
1.PMA配制
加入1mL丙酮完全溶解1mg PMA,再移取800μL母液并加入2400μL丙酮,配制成0.25mg/mL PMA。
2.PMA的诱导
CD-1小鼠,按照耳朵厚度和体重排序,剔除4只均值差异较大的动物后,随机分组成正常对照组6只和治疗组,治疗组每组10只小鼠。各取10μL浓度为0.25mg/mL PMA分别涂抹于小鼠右耳正反两面。
正常对照组的小鼠无需诱导。
3.给药和剂量设计
第一组为正常小鼠,不做任何处理;第二组给予溶媒;第三组、第四组、第五组给予化合物1,剂量分别为0.1mg/耳朵、0.3mg/耳朵和1mg/耳朵,在PMA诱导前30分钟和PMA诱导后15分钟分别在小鼠右耳进行药物涂抹。
表9实验分组及剂量设计
Figure PCTCN2020100129-appb-000015
Figure PCTCN2020100129-appb-000016
注:NA代表“无”;BID代表“一天给药两次”。
4.耳水肿发病指标测定
测量和取样:在PMA诱导10小时后麻醉小鼠测定右耳厚度。测定右耳厚度后,立即安乐死,收集耳片并称量重量。
5.统计学处理
实验数据应用平均数±标准误表示(Mean±SEM),耳朵肿胀程度和耳朵重量用单因素方差分析(One-way ANOVA),p<0.05认为有显著性差异。
实验结果:
PMA诱导后,通过10小时点的测量,耳厚度增长0.300-0.400mm,远高于正常肿胀幅度-0.010至0.002mm,耳重量平均增加28.8mg,提示此次耳水肿模型的建立非常成功。
受试化合物1在0.1mg/耳朵,0.3mg/耳朵和1mg/耳朵均可显著降低,10小时点时,三个剂量下的耳水肿小鼠的水肿程度,耳水肿抑制率分别为22%,39%和88%,耳增重抑制率分别为39%,46%和85%(与溶媒对照组相比,p值均<0.0001),并呈现良好的量效关系。
实验结论:
化合物1在0.1,0.3和3mg/耳朵三个剂量组对PMA诱导的耳水肿症状均有显著的改善作用,均可显著抑制耳增重,并且三个剂量组均呈现良好的量效关系。

Claims (22)

  1. 化合物1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:11.91±0.20°、19.36±0.20°、23.17±0.20°。
    Figure PCTCN2020100129-appb-100001
  2. 根据权利要求1所述化合物1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:11.26±0.20°、11.91±0.20°、12.91±0.20°、14.27±0.20°、19.36±0.20°、22.26±0.20°、23.17±0.20°、24.97±0.20°。
  3. 根据权利要求2所述化合物1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:11.26±0.20°、11.91±0.20°、12.91±0.20°、14.27±0.20°、15.83±0.20°、17.53±0.20°、19.36±0.20°、20.33±0.20°、22.26±0.20°、23.17±0.20°、24.97±0.20°、26.50±0.20°。
  4. 根据权利要求3所述化合物1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:11.26°、11.91°、12.91°、14.27°、15.83°、17.53°、19.36°、20.33°、22.26°、22.59°、23.17°、24.97°、26.50°、29.46°。
  5. 根据权利要求4所述化合物1的A晶型,其XRPD图谱如图1所示。
  6. 根据权利要求1~5任意一项所述的化合物1的A晶型,其差示扫描量热曲线在147.0±3.0℃处具有吸热峰的起始点。
  7. 根据权利要求6所述的化合物1的A晶型,其DSC图谱如图2所示。
  8. 根据权利要求1~5任意一项所述的化合物1的A晶型,其热重分析曲线在140.0±3.0℃时失重达0.70%。
  9. 根据权利要求8所述的化合物1的A晶型,其TGA图谱如图3所示。
  10. 式(I-1)所示的溶剂化物
    Figure PCTCN2020100129-appb-100002
    其中,n选自0.1~1.5。
  11. 根据权利要求10所述的溶剂化物,其中,n选自0.1、0.2、0.3、0.4、0.5、0.6、0.7、0.8、0.9、1、1.1、1.2、1.3、1.4和1.5。
  12. 根据权利要求11所述的溶剂化物,其中,n为0.5。
  13. 式(I-1-1)所示溶剂化物的B晶型,其中,n为0.5,其X射线粉末衍射图谱在下列2θ角处具有特征衍射峰:6.84±0.20°、8.90±0.20°、23.00±0.20°。
    Figure PCTCN2020100129-appb-100003
  14. 根据权利要求13所述的溶剂化物的B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:6.84±0.20°、8.90±0.20°、11.27±0.20°、12.75±0.20°、16.15±0.20°、17.54±0.20°、22.06±0.20°、23.00±0.20°。
  15. 根据权利要求14所述的溶剂化物的B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:6.84±0.20°、8.90±0.20°、11.27±0.20°、12.75±0.20°、16.15±0.20°、17.54±0.20°、19.17±0.20°、19.70±0.20°、20.41±0.20°、22.06±0.20°、23.00±0.20°、25.95±0.20°。
  16. 根据权利要求15所述的溶剂化物的B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:6.84°、8.90°、11.27°、12.75°、13.29°、14.94°、16.15°、17.54°、17.93°、19.17°、19.70°、20.41°、20.79°、22.06°、22.72°、23.00°、23.86°、25.95°、27.82°、28.45°、30.14°、32.87°。
  17. 根据权利要求16所述的溶剂化物的B晶型,其XRPD图谱如图5所示。
  18. 根据权利要求13~17任意一项所述的溶剂化物的B晶型,其差示扫描量热曲线在77.5±3.0℃处具有吸热峰的起始点。
  19. 根据权利要求18所述的溶剂化物的B晶型,其DSC图谱如图6所示。
  20. 根据权利要求13~17任意一项所述的溶剂化物的B晶型,其热重分析曲线在80.0±3.0℃时失重达10.46%。
  21. 根据权利要求20所述的溶剂化物的B晶型,其TGA图谱如图7所示。
  22. 根据权利要求1~9任意一项所述的化合物1的A晶型或根据权利要求10~12任意一项所述的溶剂化物或根据权利要求13~21任意一项所述的溶剂化物的B晶型在制备治疗与TNFα相关疾病药物中的应用。
PCT/CN2020/100129 2019-07-04 2020-07-03 一种苯并咪唑-2-酮类化合物的晶型、溶剂化物、溶剂化物的晶型及它们的制备方法 WO2021000934A1 (zh)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US17/624,462 US20220372001A1 (en) 2019-07-04 2020-07-03 Crystal form of benzimidazole-2-one compound, solvate thereof, crystal form of solvate thereof, and preparation method thereof
EP20835633.7A EP3995490A4 (en) 2019-07-04 2020-07-03 CRYSTAL FORM OF A BENZIMIDAZOLE-2-ONE COMPOUND, ITS SOLVATE, CRYSTAL FORM OR SOLVATE THEREOF AND PROCESS FOR THEIR PRODUCTION
CN202080048994.8A CN114072382B (zh) 2019-07-04 2020-07-03 一种苯并咪唑-2-酮类化合物的晶型、溶剂化物、溶剂化物的晶型及它们的制备方法
JP2022500106A JP2022539257A (ja) 2019-07-04 2020-07-03 ベンズイミダゾール-2-オン系化合物の結晶体、溶媒和物、溶媒和物の結晶体、およびそれらの調製方法

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201910602489 2019-07-04
CN201910602489.2 2019-07-04
CN202010426911.6 2020-05-19
CN202010426911 2020-05-19

Publications (1)

Publication Number Publication Date
WO2021000934A1 true WO2021000934A1 (zh) 2021-01-07

Family

ID=74100880

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/100129 WO2021000934A1 (zh) 2019-07-04 2020-07-03 一种苯并咪唑-2-酮类化合物的晶型、溶剂化物、溶剂化物的晶型及它们的制备方法

Country Status (5)

Country Link
US (1) US20220372001A1 (zh)
EP (1) EP3995490A4 (zh)
JP (1) JP2022539257A (zh)
CN (1) CN114072382B (zh)
WO (1) WO2021000934A1 (zh)

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002072576A1 (en) * 2001-03-09 2002-09-19 Pfizer Products Inc. Benzimidazole anti-inflammatory compounds
CN104761484A (zh) * 2014-11-24 2015-07-08 上海优拓医药科技有限公司 一种稳定的不含溶剂化物的阿普司特晶型ii及其制备方法
CN104892486A (zh) * 2015-06-25 2015-09-09 济南纽华医药科技有限公司 阿普斯特的新晶型及其制备方法
CN105407888A (zh) * 2013-06-21 2016-03-16 齐尼思表观遗传学公司 新双环溴结构域抑制剂
CN106008315A (zh) * 2016-06-16 2016-10-12 珠海联邦制药股份有限公司 一种阿普斯特晶型s及其制备方法
CN107686461A (zh) * 2016-08-04 2018-02-13 广东东阳光药业有限公司 阿普斯特的溶剂化物及其制备方法和用途
WO2018036470A1 (zh) * 2016-08-22 2018-03-01 南京明德新药研发股份有限公司 作为pde4抑制剂的并环类化合物
CN108440381A (zh) * 2018-03-15 2018-08-24 威海迪素制药有限公司 一种阿普斯特新晶型h及其制备方法

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002072576A1 (en) * 2001-03-09 2002-09-19 Pfizer Products Inc. Benzimidazole anti-inflammatory compounds
CN105407888A (zh) * 2013-06-21 2016-03-16 齐尼思表观遗传学公司 新双环溴结构域抑制剂
CN104761484A (zh) * 2014-11-24 2015-07-08 上海优拓医药科技有限公司 一种稳定的不含溶剂化物的阿普司特晶型ii及其制备方法
CN104892486A (zh) * 2015-06-25 2015-09-09 济南纽华医药科技有限公司 阿普斯特的新晶型及其制备方法
CN106008315A (zh) * 2016-06-16 2016-10-12 珠海联邦制药股份有限公司 一种阿普斯特晶型s及其制备方法
CN107686461A (zh) * 2016-08-04 2018-02-13 广东东阳光药业有限公司 阿普斯特的溶剂化物及其制备方法和用途
WO2018036470A1 (zh) * 2016-08-22 2018-03-01 南京明德新药研发股份有限公司 作为pde4抑制剂的并环类化合物
CN108440381A (zh) * 2018-03-15 2018-08-24 威海迪素制药有限公司 一种阿普斯特新晶型h及其制备方法

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3995490A4

Also Published As

Publication number Publication date
US20220372001A1 (en) 2022-11-24
EP3995490A4 (en) 2023-08-09
EP3995490A1 (en) 2022-05-11
JP2022539257A (ja) 2022-09-07
CN114072382A (zh) 2022-02-18
CN114072382B (zh) 2023-05-26

Similar Documents

Publication Publication Date Title
CN112168967A (zh) 治疗癌症的方法
WO2019242439A1 (zh) Arn-509的晶型及其制备方法和用途
CN107531745B (zh) 一种新的18α-甘草次酸衍生物及其医药用途
CN111902405B (zh) 靶向cdk4/6激酶抑制剂的晶型
WO2017219510A1 (zh) 一种具有抗肿瘤活性和抗炎活性的醌式查尔酮碳苷二聚体化合物及其制备方法
KR101406971B1 (ko) 플라본 계열 화합물을 유효성분으로 포함하는 면역 억제제 및 면역질환의 예방 또는 치료용 조성물
CA3091153A1 (en) Therapeutic agent for hepatocellular carcinoma
WO2021000934A1 (zh) 一种苯并咪唑-2-酮类化合物的晶型、溶剂化物、溶剂化物的晶型及它们的制备方法
WO2013138951A1 (zh) 喹唑啉衍生物及其作为细胞凋亡抑制剂的用途
WO2017219509A1 (zh) 一种具有抗肿瘤活性和抗炎活性的醌式查尔酮与黄酮醇结合物及其制备方法和应用
JP2016500096A (ja) ナトリウム−ヨウ素シンポータの新規阻害剤
JP2009286705A (ja) 新規抗腫瘍物質アルキルクマリン類とその用途
Wang et al. Hybrids of aurantiamide acetate and isopropylated genipin as potential anti‐inflammatory agents: The design, synthesis, and biological evaluation
RO129522A0 (ro) Agenţi antitumorali derivaţi ai n-()-1-metil-1h-pirazol-4-carboxamidei
CN115448874A (zh) 固体形式的周期蛋白依赖性激酶9抑制剂及其用途
WO2012078982A2 (en) Xzh-5 inhibits constitutive and interleukin-6-induced stat3 phosphorylation in human hepatocellular carcinoma cells
EP3747884B1 (en) Crystal form of 1h-imidazo[4,5-b]pyridine-2(3h)-one compound and preparation method therefor
CN110283138B (zh) 化合物、该化合物的制备方法及该化合物的应用和应用该化合物的产品
CN105524135B (zh) 毛酸浆内酯的制备方法及其在制备抗肿瘤药物中的应用
RU2784538C2 (ru) Кристаллическая форма соединения 1h-имидазо[4,5-b]пиридин-2(3h)-она и способ ее получения
CN113278025B (zh) 一类新骨架二萜二聚体化合物及其制备方法、药物组合物和应用
WO2022237682A1 (zh) 吡咯并三嗪类化合物的盐型、其晶型及其制备方法
WO2021077994A1 (zh) 一种低氧诱导因子脯氨酰羟化酶抑制剂晶型
TWI430800B (zh) 三萜化合物、其之製備方法及用途
CN110256394B (zh) 具有抗癌活性的化合物及其制备方法

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: 20835633

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2022500106

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2020835633

Country of ref document: EP

Effective date: 20220204