WO2024027825A1 - Cdk inhibitor and polymorph of phosphate thereof - Google Patents

Cdk inhibitor and polymorph of phosphate thereof Download PDF

Info

Publication number
WO2024027825A1
WO2024027825A1 PCT/CN2023/111237 CN2023111237W WO2024027825A1 WO 2024027825 A1 WO2024027825 A1 WO 2024027825A1 CN 2023111237 W CN2023111237 W CN 2023111237W WO 2024027825 A1 WO2024027825 A1 WO 2024027825A1
Authority
WO
WIPO (PCT)
Prior art keywords
crystal form
ray powder
powder diffraction
diffraction pattern
compound
Prior art date
Application number
PCT/CN2023/111237
Other languages
French (fr)
Chinese (zh)
Inventor
姜辰
付强
Original Assignee
齐鲁制药有限公司
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 齐鲁制药有限公司 filed Critical 齐鲁制药有限公司
Publication of WO2024027825A1 publication Critical patent/WO2024027825A1/en

Links

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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings

Definitions

  • This application discloses a CDK inhibitor, multiple crystal forms of its phosphate, preparation methods thereof, and their application in treating cancer diseases.
  • Cyclin-dependent kinases are an important class of cellular enzymes that cooperate with cyclins and play an important role in the regulation of the cell cycle. Cyclin B/CDK1, cyclin A/CDK2, cyclin E/CDK2, cyclin D/CDK4, cyclin D/CDK6 and possibly other heterodimers are important regulators of different stages of the cell cycle Factor (Harper, J.W., Adams, P.D., Cyclin-Dependent Kinases, Chem. Rev. 2001, 101, 2511-2526).
  • CDK2 forms a kinase complex with cyclin E or A and plays a decisive role in driving the cell cycle from the G1 phase to the S phase and maintaining the S phase.
  • the main mechanism is that Cyclin E and CDK2 work together to phosphorylate the retinoblastoma susceptibility gene (Rb) protein.
  • Rb retinoblastoma susceptibility gene
  • the phosphorylation of the Rb protein leads to the release of E2F (transcription factor).
  • E2F transcription factor
  • the released E2F binds to the upstream of some genes ( Usually located in the promoter or enhancer region), it initiates the transcriptional expression of those genes related to the cell cycle, causing cells to enter the S phase from the end of G1 phase.
  • CDK2 abnormal expression of CDK2 is closely related to the occurrence of cancer, such as CCNE1 amplified ovarian cancer, KRAS mutant lung cancer, hormone-dependent breast cancer and prostate cancer (Tadesse S, Anshabo AT, Portman N, Lim E, Tilley W, Caldon CE, Wang S, Targeting CDK2 in cancer: challenges and opportunities for therapy, Drug Discovery Today, 2020, 25, 406-413).
  • CDK inhibitors have become a current research focus on anti-tumor drugs.
  • CDK4/6 target mainly for breast cancer, such as Pfizer's Palbociclib, Novartis' Ribociclib and Eli Lilly's abemaciclib.
  • Molecules including CDK2 multi-target inhibitors such as fadraciclib, Roscovitine and PF-06873600 are in different clinical stages.
  • no CDK2 inhibitors have been approved for marketing. Therefore, new CDK inhibitors continue to be developed, especially those that are effective against CDK2 targets. inhibitors, which are of great research significance.
  • Patent PCT/CN2022/074491 discloses a small molecule inhibitor targeting CDK2/4/6, especially the CDK2 target. Its structure is shown in formula (A), and its chemical name is N-(1-((4 -((3S,5S)-3,5-dimethylpiperazin-1-yl)phenyl)sulfonyl)piperidin-4-yl)-4-((S)-tetrahydrofuran-3-yl)oxy methyl)-5-(trifluoromethyl)pyrimidin-2-amine.
  • This small molecule inhibitor can selectively inhibit CDK 2/4/6 kinases compared to CDK 1/7/9 kinases. It is especially excellent in the inhibitory activity of CDK 2 kinases.
  • this small molecule inhibitor has good cell proliferation inhibitory activity, and has shown good tumor inhibitory activity and good tolerance in in vivo efficacy experiments, and is expected to be developed into clinical drugs.
  • the present application discloses a polymorphic form of a CDK inhibitor, a CDK inhibitor phosphate, a polymorphic form, a preparation method of the crystal form, and their application in the treatment of cancer diseases.
  • the first aspect of the application provides a compound of formula (A) (i.e., N-(1-((4-((3S,5S)-3,5-dimethylpiperazin-1-yl)phenyl)sulfonate) Crystalline Form I of acyl)piperidin-4-yl)-4-((S)-tetrahydrofuran-3-yl)oxy)-5-(trifluoromethyl)pyrimidin-2-amine), wherein,
  • the X-ray powder diffraction pattern of Form I has characteristic peaks at 2 ⁇ values of 10.49°, 12.10°, 17.74°, 19.88°, and 21.66°, and the 2 ⁇ error range is ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystalline form I has a 2 ⁇ value of 10.03°, 10.49°, 12.10°, There are characteristic peaks at 14.28°, 14.81°, 17.74°, 18.28°, 19.88°, 20.57°, 21.66°, 23.11°, 23.76°, and 26.29°, and the 2 ⁇ error range is ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystalline form I has a 2 ⁇ value of 10.03°, 10.49°, 11.48°, 12.10°, 13.50°, 14.28°, 14.81°, 16.16°, 16.87°
  • 2 ⁇ error range is ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned Form I is substantially as shown in Figure 1.
  • the DSC spectrum of the above-mentioned crystalline form I has an endothermic characteristic peak at about 200°C.
  • the TGA-DSC spectrum of the above crystalline Form I is substantially as shown in Figure 2.
  • the XRPD pattern diffraction peak analysis data of the crystal form I of the compound of formula (A) is basically as shown in Table 1.
  • the second aspect of the application also provides a compound of formula (A) (i.e. N-(1-((4-((3S,5S)-3,5-dimethylpiperazin-1-yl)phenyl)) Form II of sulfonyl)piperidin-4-yl)-4-((S)-tetrahydrofuran-3-yl)oxy)-5-(trifluoromethyl)pyrimidin-2-amine), the crystal
  • the X-ray powder diffraction pattern of Type II has characteristic peaks at 2 ⁇ values of 8.09°, 13.31°, 16.59°, 19.55°, 21.59°, and 25.01°, and the 2 ⁇ error range is ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystalline form II has a 2 ⁇ value of 6.13°, 8.09°, 11.60°, 13.31°, 14.44°, 16.59°, 17.13°, 18.27°, 19.55°
  • the X-ray powder diffraction pattern of the above-mentioned crystalline form II has a 2 ⁇ value of 6.13°, 8.09°, 9.94°, 11.60°, 13.31°, 14.44°, 16.59°, 17.13°, 17.71°
  • the X-ray powder diffraction pattern of the above-mentioned Form II is substantially as shown in Figure 3.
  • the TGA-DSC spectrum of the above-mentioned crystal form II is substantially as shown in Figure 4.
  • the XRPD pattern diffraction peak analysis data of the crystal form II of the compound of formula (A) is basically as shown in Table 2.
  • the third aspect of the application also provides a compound of formula (A) (i.e. N-(1-((4-((3S,5S)-3,5-dimethylpiperazin-1-yl)phenyl)) Sulfonyl)piperidin-4-yl)-4-((S)-tetrahydrofuran-3-yl)oxy)-5-(trifluoromethyl)pyrimidin-2-amine) phosphate, wherein formula (A ) compound and phosphoric acid molar ratio is 1:1,
  • the inventor of the present application has tried to form salts of the compound of formula (A) with various inorganic acids or organic acids, such as hydrochloric acid, sulfuric acid, phosphoric acid, maleic acid, fumaric acid, succinic acid and p-toluenesulfonic acid. etc., but only the phosphate of formula (A) was found to have better crystallinity, hygroscopicity, solubility and stability.
  • Some other acids cannot form salts with the compound of formula (A), some have poor crystallinity, or are mostly amorphous, and some can obtain crystal forms, but the crystal forms are prone to moisture or have poor stability.
  • the fourth aspect of the present application also provides a crystal form III of the phosphate salt of the compound of formula (A).
  • the X-ray powder diffraction pattern of the crystal form III has a 2 ⁇ value of 5.85°, 8.94°, 14.86°, and 16.00°. There is a characteristic peak at 2 ⁇ , and the 2 ⁇ error range is ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form III has a 2 ⁇ value of 5.85°, 8.94°, 10.29°, 13.21°, 14.86°, 16.00°, 17.01°, 17.65°, 19.25° There is a characteristic peak at 2 ⁇ , and the 2 ⁇ error range is ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above-mentioned crystal form III has a 2 ⁇ value of 3.43°, 5.85°, 8.94°, 10.29°, 11.69°, 12.30°, 13.21°, 14.86°, 16.00°
  • the X-ray powder diffraction pattern of Form III described above is substantially as shown in Figure 8.
  • the TGA-DSC spectrum of the above-mentioned Form III is substantially as shown in Figure 9.
  • the XRPD pattern diffraction peak analysis data of the above-mentioned crystal form III is basically as shown in Table 3.
  • the fifth aspect of the present application also provides a crystalline form IV of the phosphate of the compound of formula (A).
  • the X-ray powder diffraction pattern of the crystalline form IV has a 2 ⁇ value of 13.07°, 15.66°, 16.11°, and 16.84°. , there is a characteristic peak at 21.89°, and the 2 ⁇ error range is ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystalline form IV has a 2 ⁇ value of 7.81°, 13.07°, 15.20°, 15.66°, 16.11°, 16.84°, 19.61°, 21.89°, 22.16° , there is a characteristic peak at 23.57°, and the 2 ⁇ error range is ⁇ 0.2°.
  • the X-ray powder diffraction pattern of the above crystalline form IV has a 2 ⁇ value of 7.81°, 10.97°, 13.07°, 14.20°, 15.20°, 15.66°, 16.11°, 16.84°, 19.61°
  • the X-ray powder diffraction pattern of the above-described Form IV is substantially as shown in Figure 10.
  • the DSC spectrum of the above crystalline form IV has an endothermic characteristic peak at about 229°C.
  • the TGA-DSC spectrum of the above-mentioned Form IV is substantially as shown in Figure 11.
  • the XRPD pattern diffraction peak analysis data of the above-mentioned crystal form IV is basically as shown in Table 4.
  • the sixth aspect of the application also provides a pharmaceutical composition, which includes the crystal form I described in the first aspect of the application, the crystal form II described in the second aspect of the application, and the phosphate salt described in the third aspect of the application. , the crystal form III described in the fourth aspect of this application or the crystal form IV described in the fifth aspect of this application, and a pharmaceutically acceptable carrier.
  • the seventh aspect of the application also provides the crystal form I described in the first aspect of the application, the crystal form II described in the second aspect of the application, the phosphate described in the third aspect of the application, and the crystal form II described in the fourth aspect of the application.
  • the eighth aspect of the application also provides the crystal form I described in the first aspect of the application, the crystal form II described in the second aspect of the application, the phosphate described in the third aspect of the application, and the crystal form II described in the fourth aspect of the application.
  • the ninth aspect of the present application also provides the crystalline form I described in the first aspect of the present application, the crystalline form II described in the second aspect of the present application, and the third aspect of the present application for use as medicine or for treating CDK-mediated cancer.
  • the cancer includes ovarian cancer, breast cancer, acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), or small lymphocytic lymphoma (SLL).
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • the cancer is breast cancer.
  • the tenth aspect of this application also provides a preparation method for the crystal form I described in the first aspect of this application, including the following steps:
  • step (b) Add a second solvent to the solution in step (a) and stir for a certain period of time;
  • the first solvent in the above preparation method is selected from at least one of acetone, methanol, tetrahydrofuran, ethyl acetate, acetonitrile and dichloromethane.
  • the second solvent in the above preparation method is selected from at least one of water, n-heptane, and n-hexane.
  • the certain stirring time is 1-3 hours, preferably 2 hours.
  • the crystal form in this application has good chemical stability, physical stability and low hygroscopicity, is less affected by temperature, humidity and light, and is convenient for storage and formulation development.
  • Figure 1 is an XRPD (X-ray powder diffraction) spectrum of crystal form I of the compound of formula (A).
  • Figure 2 is a TGA-DSC (differential scanning calorimetry-thermogravimetric analysis) spectrum of crystal form I of the compound of formula (A).
  • Figure 3 is the XRPD spectrum of crystal form II of the compound of formula (A).
  • Figure 4 is a TGA-DSC spectrum of crystal form II of the compound of formula (A).
  • Figure 5 is a DVS (dynamic vapor phase adsorption) spectrum of crystal form I of the compound of formula (A).
  • Figure 6 is a comparative XRPD spectrum of crystal form I of the compound of formula (A) before and after the DVS experiment.
  • Figure 7 is a comparative XRPD spectrum before and after the stability experiment of the crystal form I of the compound of formula (A).
  • Figure 8 is an XRPD spectrum of Form III of the phosphate salt of the compound of formula (A).
  • Figure 9 is a TGA-DSC spectrum of Form III of the phosphate salt of the compound of formula (A).
  • Figure 10 is an XRPD spectrum of Form IV of the phosphate salt of the compound of formula (A).
  • Figure 11 is a TGA-DSC spectrum of Form IV of the phosphate salt of the compound of formula (A).
  • Figure 12 is a DVS spectrum of Form IV of the phosphate salt of the compound of formula (A).
  • Figure 13 is a comparative XRPD spectrum before and after the stability experiment of the phosphate form IV of the compound of formula (A).
  • pharmaceutically acceptable carrier refers to a medium generally accepted in the art for delivering biologically active agents to animals, especially mammals, including, for example, adjuvants, excipients, or Excipients such as diluents, preservatives, fillers, flow regulators, disintegrants, wetting agents, emulsifiers, suspending agents, sweeteners, flavorings, aromatics, antibacterial agents, antifungal agents, Lubricants and dispersants.
  • the formulation of pharmaceutically acceptable carriers depends on a number of factors within the purview of one of ordinary skill in the art.
  • compositions containing the agent include both aqueous and non-aqueous media and a variety of solid and semi-solid dosage forms.
  • Such carriers include many different ingredients and additives in addition to the active agent, and such additional ingredients are well known to those of ordinary skill in the art to be included in the formulation for a variety of reasons (e.g., to stabilize the active agent, binders, etc.) .
  • X-ray powder diffraction patterns have one or more measurement errors depending on slight changes in measurement conditions.
  • the structure of the crystals, crystals or crystal forms disclosed or claimed in this application may vary depending on test conditions, purity, equipment It exhibits similar but not identical analytical properties within a reasonable error range as other constant variables known to those skilled in the art.
  • the diffraction angle (2 ⁇ ) in powder X-ray powder diffraction usually produces an error within the range of ⁇ 0.20°. Therefore, this application not only includes crystals with completely consistent diffraction angles in powder X-ray powder diffraction, but also includes Crystals with consistent diffraction angles within an error range of ⁇ 0.20°.
  • the crystalline form of Compound A of the present application is not limited to crystals having the same X-ray powder diffraction pattern as shown in the accompanying drawings, and has substantially the same X-ray powder diffraction pattern as shown in the accompanying drawings. Any crystal with a diffraction pattern falls within the scope of this application.
  • DSC data can reflect changes in the form of a substance. Strong endothermic peaks can indicate that the substance has dehydrated or desolvated, or has undergone crystallization, or has melted. When reflecting the melting state, the corresponding temperature is usually understood as the substance. melting point. This value will be affected by compound purity, sample weight, heating rate, particle size, and calibration and maintenance of the test equipment. Those skilled in the art can understand that the temperature when a substance transforms from a solid state to a liquid state is usually a temperature range rather than a fixed point value.
  • the temperature corresponding to the endothermic peak can be characterized by the onset value or peak value or other reasonable values. or the melting point of a substance.
  • the maximum endothermic transition temperature of the crystal form may be within the range of the above-disclosed specific value ⁇ 5.0°C, preferably within the range of ⁇ 2.0°C.
  • thermogravimetric analysis TGA
  • evaporation loss of weight
  • the temperature at which the crystal form decomposes, sublimates, or evaporates can be within the range of ⁇ 3.0°C of the specific numerical value disclosed above, for example, within the range of ⁇ 2.0°C.
  • “Stability” of a crystalline form includes “chemical stability” and/or “physical stability”. “Chemical stability” refers to the degree of degradation reaction of the crystal form under certain temperature, humidity, and light conditions. “Chemical stability” reflects the stability of the crystal form under storage conditions. “Physical stability” refers to the degree to which the crystal form is converted into a solid form under certain specific conditions, such as high temperature, high humidity, grinding, tableting, desolvation, and adsorption of solvents, into another crystal form. Therefore, “physical stability” can reflect to a certain extent the stability of the crystal form during the use of preparations and other processes.
  • the crystalline structures of the present application can be prepared by a variety of methods, including crystallization or recrystallization from a suitable solvent, sublimation, growth from a melt, solid state conversion from another phase, crystallization from a supercritical fluid, and jet spray.
  • Techniques for crystallizing or recrystallizing a crystalline structure from a solvent mixture including solvent evaporation, lowering the temperature of the solvent mixture, seeding of a supersaturated solvent mixture of the molecule and/or salt, lyophilizing the solvent mixture, adding an antisolvent to the solvent mixture wait.
  • drying refers to the process of removing solvent from the obtained solid, including but not limited to natural drying at room temperature, high temperature drying, vacuum drying and other methods.
  • the intermediate compounds of the present application can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combining them with other chemical synthesis methods, and those skilled in the art. Well-known equivalents and preferred embodiments include but are not limited to the embodiments of the present application.
  • the naming of the title compound was converted from the compound structure with the help of Chemdraw. If there is any inconsistency between the compound name and the compound structure, it can be determined by comprehensively integrating relevant information and reaction routes; if it cannot be confirmed through other methods, the given compound structural formula shall prevail.
  • the preparation methods of some compounds in this application refer to the preparation methods of similar compounds mentioned above. Persons in the art should know that when using or referring to the preparation methods cited therein, the feed ratio of the reactants, the reaction solvent, the reaction temperature, etc. can be appropriately adjusted according to the different reactants.
  • the compounds of the present application can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combining them with other chemical synthesis methods, and methods well known to those skilled in the art. Equivalent alternatives and preferred implementations include but are not limited to the embodiments of this application.
  • the solid sample is analyzed with an X-ray powder diffractometer (X'Pert PRO). Take an appropriate amount of fine powder of the test sample, place it in the groove of the sample holder, and press it into a flat and dense plane with a glass piece.
  • the XRPD measurement parameters are shown in Table 5. .
  • Thermogravimetric analysis of solids was performed using TA Instrument's thermogravimetric analyzer. Approximately 1-5 mg of sample was placed in a peeled aluminum sample pan, the sample was heated according to the parameters listed in Table 6, and the data was analyzed using TRIOS.
  • Thermogravimetry-differential scanning calorimetry analysis of solids was performed using a simultaneous thermal analyzer from Mettler Toledo. Use a small spoon to take an appropriate amount of the test sample and place it in the crucible, spread it evenly, weigh it, heat the sample according to the parameters listed in Table 7, and use STARe to analyze the data.
  • the hygroscopicity of the samples was measured using the DVS Intrinsic dynamic moisture adsorption instrument. Place the sample into the tared sample basket, the instrument automatically weighs, and analyzes the sample according to the parameters in Table 8.
  • Chromatographic column Use an anion exchange chromatographic column [analytical column Ionpac TM AS11-HC (4mm ⁇ 250mm), guard column Ionpac TM AG11-HC (4mm ⁇ 50mm)];
  • AERS 500 4mm or equivalent suppressor AERS 500 4mm or equivalent suppressor
  • Running time approximately 20 minutes.
  • Step 1 Compound 1B-3 (508 mg, 1.81 mmol) was dissolved in dichloromethane (30 mL) at room temperature. Subsequently, N,N-diisopropylethylamine (702 mg, 5.43 mmol) and 4-bromo-benzenesulfonyl chloride (508 mg, 2.00 mmol) were added thereto under an ice-water bath. After the reaction solution was stirred at room temperature for 2 hours, water (60 ml) was added to quench the mixture. The mixture was extracted with dichloromethane (20 ml ⁇ 3 times), and the organic phases were combined.
  • Step 2 In a sealed jar, dissolve (S)-3-hydroxytetrahydrofuran (42 mg, 0.48 mmol) in tetrahydrofuran (2 mL). Subsequently, sodium hydride (21 mg, 0.88 mmol) was added thereto under an ice-water bath. After reacting for 15 minutes, compound 99-1 (200 mg, 0.40 mmol) was added, and the reaction solution was heated to 100°C. After reaction for 4 hours, water (60 ml) was added to the reaction solution to quench the reaction solution. The mixture was extracted with dichloromethane (20 ml ⁇ 3 times), and the organic phases were combined.
  • Step 1 Under nitrogen protection conditions, compound 99 (25g, 45.3 mmol), (2S, 6S)-2,6-dimethylpiperazine-1-carboxylic acid tert-butyl ester (11.6 g, 45.3 mmol) ), tris(dibenzylideneacetone)dipalladium (4.1 g, 4.53 mmol), 2-dicyclohexylphosphon-2,4,6-triisopropylbiphenyl (1.3 g, 9.02 mmol) and carbonic acid Cesium (29.4 g, 90.6 mmol) was dissolved in 1,4-dioxane (1.25 L). The reaction solution was heated to 100°C and stirred overnight.
  • Step 2 Compound 168-1 (18.1 g, 26.4 mmol) was dissolved in 1,4dioxane (100 mL). Subsequently, dioxane hydrochloride solution (100 ml) was added thereto. The reaction was stirred at room temperature overnight. The reaction solution was concentrated under reduced pressure. The obtained residue was washed with sodium bicarbonate aqueous solution (300 ml), the mixture was extracted with ethyl acetate (200 ml ⁇ 3 times), and the organic phases were combined. The organic phase Wash with saturated brine (100 ml), dry over anhydrous sodium sulfate, filter, and finally concentrate under reduced pressure.
  • This experiment uses the capillary migration ability change assay (MSA) method to test the inhibitory effect of the compound on CDK1/CDK2/CDK4/CDK6/CDK7/CDK9 kinase activity, and concludes that the compound’s inhibitory effect on CDK1/CDK2/CDK4/CDK6/CDK7/CDK9 kinase The half inhibitory concentration IC 50 of activity.
  • MSA capillary migration ability change assay
  • CDK1/CDK2/CDK4/CDK6/CDK7/CDK9 were purchased from Carna Company, Carliper substrate CTD3/substrate 18/substrate 8 were purchased from Gill Biochemical Company, Dinaciclib/Palbociclib were purchased from Selleckchem Company, DMSO (dimethyl sulfoxide) It was purchased from Sigma Company, and the 384-well plate was purchased from Corning Company.
  • test compound test concentration is 1 ⁇ M, 10 ⁇ M or 30 ⁇ M is the starting concentration, dilute 3 times to 10 concentrations, and detect in duplicate. Dilute to a 100x final concentration of 100% DMSO solution in a 384-well source plate. Use the Dispenser Echo 550 to transfer 250 nL of compound at 100x the final concentration to the destination 384-well plate. Add 250nL DMSO to the positive and negative control wells.
  • % inhibition rate (inhibition) (conversion%_max-conversion%_sample)/(conversion%_max-conversion%_min) ⁇ 100%
  • Conversion%_sample is the conversion rate reading of the sample
  • Conversion%_min the mean value of the negative control wells, representing the conversion rate reading of the wells without enzyme activity
  • Conversion%_max the mean value of the positive control wells, representing the conversion rate reading of the wells without compound inhibition.
  • the inhibitory IC 50 data of the compounds of the present application on CDK kinase activity are shown in Table 9. Among them: compounds with IC 50 ⁇ 0.5nM are represented by AA, compounds with 0.5nM ⁇ IC 50 ⁇ 2.5nM are represented by AB, compounds with 2.5nM ⁇ IC 50 ⁇ 10nM are represented by AC, and compounds with 10nM ⁇ IC 50 ⁇ 50nM are represented by B. To identify, compounds with 50nM ⁇ IC 50 ⁇ 100nM are identified with C, compounds with 100nM ⁇ IC 50 ⁇ 1000nM are identified with D, and compounds with IC 50 >1000nM are identified with E.
  • Compound A of this application has good CDK 2/4/6 kinase inhibitory activity, especially excellent in the inhibitory activity of CDK 2 kinase; Compound A of this application can be selected compared to CDK 1/7/9 kinase It specifically inhibits CDK 2/4/6 kinases, especially in the inhibitory activity of CDK 2 kinase. Its kinase selectivity can reach nearly 10 times, even dozens or hundreds of times.
  • This experiment uses the CellTiter-Glo method to test the inhibitory effect of compounds on HCC1806/NIH:OVCAR-3 cell proliferation, and Find the concentration IC 50 (nM) at which the compound inhibits cell growth by half.
  • HCC1806 was purchased from Tongpai (Shanghai) Biotechnology Co., Ltd.; NIH:OVCAR-3 was purchased from the ATCC Cell Bank of the United States.
  • FBS fetal bovine serum
  • Penicillin-Streptomycin Penicillin-Streptomycin
  • GlutaMAX-I Supplement purchased from GIBCO.
  • PF-06873600 was purchased from Selleck.
  • CellTiter-Glo reagent was purchased from Promega Company.
  • Envision microplate reader detects chemiluminescence signal.
  • the IC 50 of Compound A of the present application on the cell proliferation of the HCC1806/NIH:OVCAR-3 cell line can be less than 100nM, which has a better inhibitory effect than the reference compound PF-06873600.
  • HCC1806 was purchased from Tongpai (Shanghai) Biotechnology Co., Ltd.; NIH:OVCAR-3 was purchased from the ATCC Cell Bank of the United States.
  • fetal bovine serum (FBS), and Penicillin-Streptomycin were purchased from GIBCO.
  • PF-06873600 was purchased from MCE Company.
  • OVCAR-3 was purchased from ATCC cell bank in the United States. 1640 medium, fetal bovine serum (FBS), and Penicillin-Streptomycin were purchased from GIBCO. PF-06873600 was purchased from MCE Company.
  • the obtained solid was subjected to XRPD and TGA-DSC testing and characterization.
  • the solid was crystalline form I.
  • the TGA-DSC spectrum is shown in Figure 2, which has an obvious endothermic peak around 200°C.
  • the obtained solid was subjected to XRPD and TGA-DSC testing and characterization, and the solid was crystalline form II.
  • the TGA-DSC spectrum is shown in Figure 4.
  • Figure 5 is the DVS curve of Form I
  • Figure 6 is the XRPD spectrum of Form I before and after the DVS test.
  • the DVS results show that the crystalline form I absorbs moisture and gains weight by 0.14% at 80% RH and 0.16% at 90%RH, indicating that the crystalline form has almost no hygroscopicity.
  • the XRPD of the remaining solid after the DVS experiment was tested, and the results showed that The crystal form has not changed.
  • the PO 4 3- content measured by ion chromatography is 14.9%, which is close to the theoretical content of monophosphate PO 4 3- (13.9%). Combined with the above nuclear magnetic results, it is confirmed that it is in the form of monophosphate, that is, the compound of formula (A) and phosphoric acid The molar ratio is 1:1.
  • the obtained solid was subjected to XRPD and TGA-DSC testing and characterization, and the solid was crystalline form III.
  • the TGA-DSC spectrum is shown in Figure 9.
  • the obtained solid was characterized by XRPD and TGA-DSC tests and found that the solid was crystal form IV.
  • the TGA-DSC spectrum is shown in Figure 11. In the DSC spectrum, there is a characteristic endothermic peak at 229°C.
  • Figure 12 is the DVS curve of the crystal form IV.
  • the DVS results show that the hygroscopic weight gain of the crystal form IV is 0.398% at 80% RH and 0.491% at 90% RH, indicating that the crystal form is slightly hygroscopic.

Abstract

The present application provides a polymorph of a compound of formula (A), i.e., N-(1-((4-((3S,5S)-3,5-dimethylpiperazine-1-yl)phenyl)sulfonyl)piperidin-4-yl)-4-((S)-tetrahydrofuran-3-yl)oxy)-5(trifluoromethyl)pyrimidine-2-amine, and a polymorph of a phosphate of the compound of formula (A). A crystalline form provided by the present application has good chemical stability and physical stability, and low hygroscopicity, is less affected by temperature, humidity, and illumination, and facilitates storage and preparation development.

Description

一种CDK抑制剂及其磷酸盐的多晶型A polymorphic form of a CDK inhibitor and its phosphate
本申请要求于2022年08月05日提交中国专利局、申请号为CN202210947810.2、发明名称“一种CDK抑制剂的多晶型”的中国专利申请,2022年08月05日提交中国专利局、申请号为CN202210936709.7、发明名称“一种CDK抑制剂磷酸盐的多晶型”的中国专利申请,2023年07月19日提交中国专利局、申请号为CN202310889701.4、发明名称“一种CDK抑制剂及其磷酸盐的多晶型”的中国专利申请的优先权,其全部内容通过引用结合在本申请中。This application is required to be submitted to the China Patent Office on August 5, 2022, with the application number CN202210947810.2 and the invention title "A polymorphic form of a CDK inhibitor". It is submitted to the China Patent Office on August 5, 2022. , the Chinese patent application with the application number CN202210936709.7 and the invention name "A polymorphic form of a CDK inhibitor phosphate" was submitted to the China Patent Office on July 19, 2023. The application number is CN202310889701.4 and the invention name is "a polymorphic form of a CDK inhibitor phosphate". The priority of the Chinese patent application "A CDK inhibitor and polymorphic form of its phosphate", the entire content of which is incorporated by reference in this application.
技术领域Technical field
本申请公开了一种CDK抑制剂、其磷酸盐的多种晶型及其制备方法,以及它们在治疗癌症疾病中的应用。This application discloses a CDK inhibitor, multiple crystal forms of its phosphate, preparation methods thereof, and their application in treating cancer diseases.
背景技术Background technique
细胞周期是细胞生命活动的基本过程,控制着细胞的生长、增殖和分化。细胞周期蛋白-依赖性激酶(cyclin-dependent kinases,CDKs)是一类重要的细胞酶,与细胞周期蛋白(cyclins)协同,在细胞周期的调控中发挥着重要的作用。细胞周期蛋白B/CDK1、细胞周期蛋白A/CDK2、细胞周期蛋白E/CDK2、细胞周期蛋白D/CDK4、细胞周期蛋白D/CDK6和其他可能的杂二聚体是细胞周期不同阶段的重要调节因子(Harper,J.W.,Adams,P.D.,Cyclin-Dependent Kinases,Chem.Rev.2001,101,2511-2526)。The cell cycle is the basic process of cell life activities, controlling the growth, proliferation and differentiation of cells. Cyclin-dependent kinases (CDKs) are an important class of cellular enzymes that cooperate with cyclins and play an important role in the regulation of the cell cycle. Cyclin B/CDK1, cyclin A/CDK2, cyclin E/CDK2, cyclin D/CDK4, cyclin D/CDK6 and possibly other heterodimers are important regulators of different stages of the cell cycle Factor (Harper, J.W., Adams, P.D., Cyclin-Dependent Kinases, Chem. Rev. 2001, 101, 2511-2526).
CDK2作为细胞周期中重要的调控因子,与细胞周期蛋白E或A组成激酶复合物,在驱动细胞周期从G1期进入S期和维持S期的过程中起决定性作用。其机制主要是Cyclin E和CDK2共同作用使视网膜母细胞瘤易感基因(Rb)蛋白磷酸化,Rb蛋白的磷酸化则导致E2F(转录因子)的释放,释放的E2F结合在一些基因的上游(通常位于启动子或增强子区),启动那些与细胞周期相关基因的转录表达,使细胞从G1末期进入S期。已有众多研究表明,CDK2的异常表达与癌症的发生密切相关,例如CCNE1扩增的卵巢癌、KRAS突变型肺癌、激素依赖型的乳腺癌与前列腺癌等(Tadesse S,Anshabo AT,Portman N,Lim E,Tilley W,Caldon CE,Wang S,Targeting CDK2 in cancer:challenges and opportunities for therapy,Drug Discovery Today,2020,25,406-413)。As an important regulatory factor in the cell cycle, CDK2 forms a kinase complex with cyclin E or A and plays a decisive role in driving the cell cycle from the G1 phase to the S phase and maintaining the S phase. The main mechanism is that Cyclin E and CDK2 work together to phosphorylate the retinoblastoma susceptibility gene (Rb) protein. The phosphorylation of the Rb protein leads to the release of E2F (transcription factor). The released E2F binds to the upstream of some genes ( Usually located in the promoter or enhancer region), it initiates the transcriptional expression of those genes related to the cell cycle, causing cells to enter the S phase from the end of G1 phase. Numerous studies have shown that abnormal expression of CDK2 is closely related to the occurrence of cancer, such as CCNE1 amplified ovarian cancer, KRAS mutant lung cancer, hormone-dependent breast cancer and prostate cancer (Tadesse S, Anshabo AT, Portman N, Lim E, Tilley W, Caldon CE, Wang S, Targeting CDK2 in cancer: challenges and opportunities for therapy, Drug Discovery Today, 2020, 25, 406-413).
随着细胞周期蛋白依赖性激酶(CDKs)在细胞周期调控中的重要作用被确定,CDK抑制剂已经成为当前抗肿瘤药物的研究热点。目前全球已有多个CDK抑制剂批准上市,但多作用于CDK4/6靶点,主要以乳腺癌为适应症,例如辉瑞公司的Palbociclib,诺华公司的Ribociclib以及礼来公司的abemaciclib。包含CDK2的多靶点抑制剂fadraciclib、Roscovitine以及PF-06873600等分子处于不同的临床阶段,目前还没有CDK2抑制剂获批上市,因此,继续开发新型的CDK抑制剂,尤其是针对CDK2靶点有效的抑制剂,具有重大的研究意义。With the identification of the important role of cyclin-dependent kinases (CDKs) in cell cycle regulation, CDK inhibitors have become a current research focus on anti-tumor drugs. Currently, multiple CDK inhibitors have been approved for marketing around the world, but most of them act on the CDK4/6 target, mainly for breast cancer, such as Pfizer's Palbociclib, Novartis' Ribociclib and Eli Lilly's abemaciclib. Molecules including CDK2 multi-target inhibitors such as fadraciclib, Roscovitine and PF-06873600 are in different clinical stages. Currently, no CDK2 inhibitors have been approved for marketing. Therefore, new CDK inhibitors continue to be developed, especially those that are effective against CDK2 targets. inhibitors, which are of great research significance.
专利PCT/CN2022/074491中公开了一种针对CDK2/4/6,尤其是CDK2靶点的小分子抑制剂,其结构如式(A)所示,化学名称为N-(1-((4-((3S,5S)-3,5-二甲基哌嗪-1-基)苯基)磺酰基)哌啶-4-基)-4-((S)-四氢呋喃-3-基)氧基)-5-(三氟甲基)嘧啶-2-胺。该小分子抑制剂相对于CDK 1/7/9激酶,可选择性抑制CDK 2/4/6激酶,尤其在CDK 2激酶的抑制活性上表现出色,其激酶选择性可达近10倍,甚至几十倍、百倍以上,该小分子抑制剂有着较好的细胞增殖抑制活性,并且在体内药效实验中表现出了较好的肿瘤抑制活性与良好的耐受性,有望开发成临床药物。
Patent PCT/CN2022/074491 discloses a small molecule inhibitor targeting CDK2/4/6, especially the CDK2 target. Its structure is shown in formula (A), and its chemical name is N-(1-((4 -((3S,5S)-3,5-dimethylpiperazin-1-yl)phenyl)sulfonyl)piperidin-4-yl)-4-((S)-tetrahydrofuran-3-yl)oxy methyl)-5-(trifluoromethyl)pyrimidin-2-amine. This small molecule inhibitor can selectively inhibit CDK 2/4/6 kinases compared to CDK 1/7/9 kinases. It is especially excellent in the inhibitory activity of CDK 2 kinases. Its kinase selectivity can reach nearly 10 times, even Dozens or hundreds of times more, this small molecule inhibitor has good cell proliferation inhibitory activity, and has shown good tumor inhibitory activity and good tolerance in in vivo efficacy experiments, and is expected to be developed into clinical drugs.
发明内容Contents of the invention
本申请公开了一种CDK抑制剂的多晶型、CDK抑制剂磷酸盐及其多晶型、晶型的制备方法,以及它们在治疗癌症疾病中的应用。The present application discloses a polymorphic form of a CDK inhibitor, a CDK inhibitor phosphate, a polymorphic form, a preparation method of the crystal form, and their application in the treatment of cancer diseases.
具体的,specific,
本申请第一方面提供了一种式(A)化合物(即N-(1-((4-((3S,5S)-3,5-二甲基哌嗪-1-基)苯基)磺酰基)哌啶-4-基)-4-((S)-四氢呋喃-3-基)氧基)-5-(三氟甲基)嘧啶-2-胺)的晶型I,其中,所述晶型I的X-射线粉末衍射图谱在2θ值为10.49°、12.10°、17.74°、19.88°、21.66°处有特征峰,2θ误差范围为±0.2°。The first aspect of the application provides a compound of formula (A) (i.e., N-(1-((4-((3S,5S)-3,5-dimethylpiperazin-1-yl)phenyl)sulfonate) Crystalline Form I of acyl)piperidin-4-yl)-4-((S)-tetrahydrofuran-3-yl)oxy)-5-(trifluoromethyl)pyrimidin-2-amine), wherein, The X-ray powder diffraction pattern of Form I has characteristic peaks at 2θ values of 10.49°, 12.10°, 17.74°, 19.88°, and 21.66°, and the 2θ error range is ±0.2°.
在本申请的一些实施方案中,上述晶型I的X-射线粉末衍射图谱在2θ值为10.03°、10.49°、12.10°、 14.28°、14.81°、17.74°、18.28°、19.88°、20.57°、21.66°、23.11°、23.76°、26.29°处有特征峰,2θ误差范围为±0.2°。In some embodiments of the present application, the X-ray powder diffraction pattern of the above-mentioned crystalline form I has a 2θ value of 10.03°, 10.49°, 12.10°, There are characteristic peaks at 14.28°, 14.81°, 17.74°, 18.28°, 19.88°, 20.57°, 21.66°, 23.11°, 23.76°, and 26.29°, and the 2θ error range is ±0.2°.
在本申请的一些实施方案中,上述晶型I的X-射线粉末衍射图谱在2θ值为10.03°、10.49°、11.48°、12.10°、13.50°、14.28°、14.81°、16.16°、16.87°、17.74°、18.28°、19.88°、20.57°、21.10°、21.66°、22.13°、22.45°、23.11°、23.76°、24.67°、25.87°、26.29°、30.46°、32.57°处有特征峰,2θ误差范围为±0.2°。In some embodiments of the present application, the X-ray powder diffraction pattern of the above-mentioned crystalline form I has a 2θ value of 10.03°, 10.49°, 11.48°, 12.10°, 13.50°, 14.28°, 14.81°, 16.16°, 16.87° There are characteristic peaks at , 17.74°, 18.28°, 19.88°, 20.57°, 21.10°, 21.66°, 22.13°, 22.45°, 23.11°, 23.76°, 24.67°, 25.87°, 26.29°, 30.46°, 32.57°, 2θ error range is ±0.2°.
在本申请的一些实施方案中,上述晶型I的X-射线粉末衍射图谱基本上如图1所示。In some embodiments of the present application, the X-ray powder diffraction pattern of the above-mentioned Form I is substantially as shown in Figure 1.
在本申请的一些实施方案中,上述晶型I的DSC谱图在200℃左右处有一个吸热特征峰。In some embodiments of the present application, the DSC spectrum of the above-mentioned crystalline form I has an endothermic characteristic peak at about 200°C.
在本申请的一些实施方案中,上述晶型I的TGA-DSC谱图基本上如图2所示。In some embodiments of the present application, the TGA-DSC spectrum of the above crystalline Form I is substantially as shown in Figure 2.
本申请的一些实施方案中,上述式(A)化合物的晶型I的XRPD图谱衍射峰解析数据基本上如表1所示。In some embodiments of the present application, the XRPD pattern diffraction peak analysis data of the crystal form I of the compound of formula (A) is basically as shown in Table 1.
表1 式(A)化合物晶型I的XRPD衍射峰解析数据

Table 1 XRPD diffraction peak analysis data of crystal form I of the compound of formula (A)

本申请第二方面还提供了一种式(A)化合物(即N-(1-((4-((3S,5S)-3,5-二甲基哌嗪-1-基)苯基)磺酰基)哌啶-4-基)-4-((S)-四氢呋喃-3-基)氧基)-5-(三氟甲基)嘧啶-2-胺)的晶型II,所述晶型II的X-射线粉末衍射图谱在2θ值为8.09°、13.31°、16.59°、19.55°、21.59°、25.01°处有特征峰,2θ误差范围为±0.2°。The second aspect of the application also provides a compound of formula (A) (i.e. N-(1-((4-((3S,5S)-3,5-dimethylpiperazin-1-yl)phenyl)) Form II of sulfonyl)piperidin-4-yl)-4-((S)-tetrahydrofuran-3-yl)oxy)-5-(trifluoromethyl)pyrimidin-2-amine), the crystal The X-ray powder diffraction pattern of Type II has characteristic peaks at 2θ values of 8.09°, 13.31°, 16.59°, 19.55°, 21.59°, and 25.01°, and the 2θ error range is ±0.2°.
在本申请的一些实施方案中,上述晶型II的X-射线粉末衍射图谱在2θ值为6.13°、8.09°、11.60°、13.31°、14.44°、16.59°、17.13°、18.27°、19.55°、20.93°、21.59°、22.54°、25.01°、26.92°处有特征峰,2θ误差范围为±0.2°。In some embodiments of the present application, the X-ray powder diffraction pattern of the above-mentioned crystalline form II has a 2θ value of 6.13°, 8.09°, 11.60°, 13.31°, 14.44°, 16.59°, 17.13°, 18.27°, 19.55° There are characteristic peaks at , 20.93°, 21.59°, 22.54°, 25.01°, and 26.92°, and the 2θ error range is ±0.2°.
在本申请的一些实施方案中,上述晶型II的X-射线粉末衍射图谱在2θ值为6.13°、8.09°、9.94°、11.60°、13.31°、14.44°、16.59°、17.13°、17.71°、18.27°、19.55°、19.98°、20.93°、22.07°、21.59°、22.54°、25.01°、25.35°、25.60°、26.92°、29.34°处有特征峰,2θ误差范围为±0.2°。In some embodiments of the present application, the X-ray powder diffraction pattern of the above-mentioned crystalline form II has a 2θ value of 6.13°, 8.09°, 9.94°, 11.60°, 13.31°, 14.44°, 16.59°, 17.13°, 17.71° There are characteristic peaks at , 18.27°, 19.55°, 19.98°, 20.93°, 22.07°, 21.59°, 22.54°, 25.01°, 25.35°, 25.60°, 26.92°, and 29.34°, and the 2θ error range is ±0.2°.
在本申请的一些实施方案中,上述晶型II的X-射线粉末衍射图谱基本上如图3所示。In some embodiments of the present application, the X-ray powder diffraction pattern of the above-mentioned Form II is substantially as shown in Figure 3.
在本申请的一些实施方案中,上述晶型II的TGA-DSC谱图基本上如图4所示。In some embodiments of the present application, the TGA-DSC spectrum of the above-mentioned crystal form II is substantially as shown in Figure 4.
本申请的一些实施方案中,上述式(A)化合物的晶型II的XRPD图谱衍射峰解析数据基本上如表2所示。In some embodiments of the present application, the XRPD pattern diffraction peak analysis data of the crystal form II of the compound of formula (A) is basically as shown in Table 2.
表2 式(A)化合物晶型II的XRPD衍射峰解析数据

Table 2 XRPD diffraction peak analysis data of the crystal form II of the compound of formula (A)

本申请第三方面还提供了一种式(A)化合物(即N-(1-((4-((3S,5S)-3,5-二甲基哌嗪-1-基)苯基)磺酰基)哌啶-4-基)-4-((S)-四氢呋喃-3-基)氧基)-5-(三氟甲基)嘧啶-2-胺)的磷酸盐,其中式(A)化合物与磷酸的摩尔比例为1:1,
The third aspect of the application also provides a compound of formula (A) (i.e. N-(1-((4-((3S,5S)-3,5-dimethylpiperazin-1-yl)phenyl)) Sulfonyl)piperidin-4-yl)-4-((S)-tetrahydrofuran-3-yl)oxy)-5-(trifluoromethyl)pyrimidin-2-amine) phosphate, wherein formula (A ) compound and phosphoric acid molar ratio is 1:1,
本申请发明人在研究中尝试了式(A)化合物与多种无机酸或者有机酸成盐,例如盐酸、硫酸、磷酸、顺丁烯二酸、富马酸、丁二酸以及对甲苯磺酸等,但仅发现式(A)的磷酸盐具有较好的结晶度、引湿性、溶解度及稳定性。其他酸有的不能与式(A)化合物成盐,有的结晶度很差,或者多为无定型,有些可以得到晶型,但晶型易引湿或稳定性不佳。In the research, the inventor of the present application has tried to form salts of the compound of formula (A) with various inorganic acids or organic acids, such as hydrochloric acid, sulfuric acid, phosphoric acid, maleic acid, fumaric acid, succinic acid and p-toluenesulfonic acid. etc., but only the phosphate of formula (A) was found to have better crystallinity, hygroscopicity, solubility and stability. Some other acids cannot form salts with the compound of formula (A), some have poor crystallinity, or are mostly amorphous, and some can obtain crystal forms, but the crystal forms are prone to moisture or have poor stability.
本申请第四方面还提供了一种式(A)化合物的磷酸盐的晶型III,所述晶型III的X-射线粉末衍射图谱在2θ值为5.85°、8.94°、14.86°、16.00°处有特征峰,2θ误差范围为±0.2°。The fourth aspect of the present application also provides a crystal form III of the phosphate salt of the compound of formula (A). The X-ray powder diffraction pattern of the crystal form III has a 2θ value of 5.85°, 8.94°, 14.86°, and 16.00°. There is a characteristic peak at 2θ, and the 2θ error range is ±0.2°.
在本申请的一些实施方案中,上述晶型III的X-射线粉末衍射图谱在2θ值为5.85°、8.94°、10.29°、13.21°、14.86°、16.00°、17.01°、17.65°、19.25°处有特征峰,2θ误差范围为±0.2°。In some embodiments of the present application, the X-ray powder diffraction pattern of the above-mentioned crystal form III has a 2θ value of 5.85°, 8.94°, 10.29°, 13.21°, 14.86°, 16.00°, 17.01°, 17.65°, 19.25° There is a characteristic peak at 2θ, and the 2θ error range is ±0.2°.
在本申请的一些实施方案中,上述晶型III的X-射线粉末衍射图谱在2θ值为3.43°、5.85°、8.94°、10.29°、11.69°、12.30°、13.21°、14.86°、16.00°、17.01°、17.65°、19.25°、19.83°、20.71°、21.82°、22.28°、23.86°处有特征峰,2θ误差范围为±0.2°。In some embodiments of the present application, the X-ray powder diffraction pattern of the above-mentioned crystal form III has a 2θ value of 3.43°, 5.85°, 8.94°, 10.29°, 11.69°, 12.30°, 13.21°, 14.86°, 16.00° There are characteristic peaks at , 17.01°, 17.65°, 19.25°, 19.83°, 20.71°, 21.82°, 22.28°, and 23.86°, and the 2θ error range is ±0.2°.
在本申请的一些实施方案中,上述晶型III的X-射线粉末衍射图谱基本上如图8所示。In some embodiments of the present application, the X-ray powder diffraction pattern of Form III described above is substantially as shown in Figure 8.
在本申请的一些实施方案中,上述晶型III的TGA-DSC谱图基本上如图9所示。In some embodiments of the present application, the TGA-DSC spectrum of the above-mentioned Form III is substantially as shown in Figure 9.
在本申请的一些实施方案中,上述晶型III的XRPD图谱衍射峰解析数据基本上如表3所示。In some embodiments of the present application, the XRPD pattern diffraction peak analysis data of the above-mentioned crystal form III is basically as shown in Table 3.
表3 式(A)化合物磷酸盐晶型III的XRPD衍射峰解析数据

Table 3 XRPD diffraction peak analysis data of phosphate crystal form III of the compound of formula (A)

本申请第五方面还提供了一种式(A)化合物的磷酸盐的晶型IV,所述晶型IV的X-射线粉末衍射图谱在2θ值为13.07°、15.66°、16.11°、16.84°、21.89°处有特征峰,2θ误差范围为±0.2°。The fifth aspect of the present application also provides a crystalline form IV of the phosphate of the compound of formula (A). The X-ray powder diffraction pattern of the crystalline form IV has a 2θ value of 13.07°, 15.66°, 16.11°, and 16.84°. , there is a characteristic peak at 21.89°, and the 2θ error range is ±0.2°.
在本申请的一些实施方案中,上述晶型IV的X-射线粉末衍射图谱在2θ值为7.81°、13.07°、15.20°、15.66°、16.11°、16.84°、19.61°、21.89°、22.16°、23.57°有特征峰,2θ误差范围为±0.2°。In some embodiments of the present application, the X-ray powder diffraction pattern of the above crystalline form IV has a 2θ value of 7.81°, 13.07°, 15.20°, 15.66°, 16.11°, 16.84°, 19.61°, 21.89°, 22.16° , there is a characteristic peak at 23.57°, and the 2θ error range is ±0.2°.
在本申请的一些实施方案中,上述晶型IV的X-射线粉末衍射图谱在2θ值为7.81°、10.97°、13.07°、14.20°、15.20°、15.66°、16.11°、16.84°、19.61°、21.89°、22.16°、22.93°、23.57°、24.70°、25.92°、27.42°、28.61°处有特征峰,2θ误差范围为±0.2°。In some embodiments of the present application, the X-ray powder diffraction pattern of the above crystalline form IV has a 2θ value of 7.81°, 10.97°, 13.07°, 14.20°, 15.20°, 15.66°, 16.11°, 16.84°, 19.61° There are characteristic peaks at , 21.89°, 22.16°, 22.93°, 23.57°, 24.70°, 25.92°, 27.42°, and 28.61°, and the 2θ error range is ±0.2°.
在本申请的一些实施方案中,上述晶型IV的X-射线粉末衍射图谱基本上如图10所示。In some embodiments of the present application, the X-ray powder diffraction pattern of the above-described Form IV is substantially as shown in Figure 10.
在本申请的一些实施方案中,上述晶型IV的DSC谱图在229℃左右处有一个吸热特征峰。In some embodiments of the present application, the DSC spectrum of the above crystalline form IV has an endothermic characteristic peak at about 229°C.
在本申请的一些实施方案中,上述晶型IV的TGA-DSC谱图基本上如图11所示。In some embodiments of the present application, the TGA-DSC spectrum of the above-mentioned Form IV is substantially as shown in Figure 11.
在本申请的一些实施方案中,上述晶型IV的XRPD图谱衍射峰解析数据基本上如表4所示。In some embodiments of the present application, the XRPD pattern diffraction peak analysis data of the above-mentioned crystal form IV is basically as shown in Table 4.
表4 式(A)化合物磷酸盐晶型IV的XRPD衍射峰解析数据

Table 4 XRPD diffraction peak analysis data of phosphate crystal form IV of the compound of formula (A)

本申请第六方面还提供了一种药物组合物,其包含本申请第一方面所述的晶型I、本申请第二方面所述的晶型II、本申请第三方面所述的磷酸盐、本申请第四方面所述的晶型III或本申请第五方面所述的晶型IV,及可药用的载体。The sixth aspect of the application also provides a pharmaceutical composition, which includes the crystal form I described in the first aspect of the application, the crystal form II described in the second aspect of the application, and the phosphate salt described in the third aspect of the application. , the crystal form III described in the fourth aspect of this application or the crystal form IV described in the fifth aspect of this application, and a pharmaceutically acceptable carrier.
本申请第七方面还提供了本申请第一方面所述的晶型I、本申请第二方面所述的晶型II、本申请第三方面所述的磷酸盐、本申请第四方面所述的晶型III、本申请第五方面所述的晶型IV或本申请第六方面所述的药物组合物在制备用于治疗CDK介导的癌症药物中的用途。The seventh aspect of the application also provides the crystal form I described in the first aspect of the application, the crystal form II described in the second aspect of the application, the phosphate described in the third aspect of the application, and the crystal form II described in the fourth aspect of the application. The use of the crystal form III, the crystal form IV described in the fifth aspect of the present application, or the pharmaceutical composition described in the sixth aspect of the present application in the preparation of drugs for treating CDK-mediated cancer.
本申请第八方面还提供了本申请第一方面所述的晶型I、本申请第二方面所述的晶型II、本申请第三方面所述的磷酸盐、本申请第四方面所述的晶型III、本申请第五方面所述的晶型IV或本申请第六方面所述的药物组合物在治疗CDK介导的癌症中的用途。The eighth aspect of the application also provides the crystal form I described in the first aspect of the application, the crystal form II described in the second aspect of the application, the phosphate described in the third aspect of the application, and the crystal form II described in the fourth aspect of the application. The use of the crystal form III, the crystal form IV described in the fifth aspect of the application, or the pharmaceutical composition described in the sixth aspect of the application in the treatment of CDK-mediated cancer.
本申请第九方面还提供了用作药物或用于治疗CDK介导的癌症的本申请第一方面所述的晶型I、本申请第二方面所述的晶型II、本申请第三方面所述的磷酸盐、本申请第四方面所述的晶型III、本申请第五方面所述的晶型IV或本申请第六方面所述的药物组合物。The ninth aspect of the present application also provides the crystalline form I described in the first aspect of the present application, the crystalline form II described in the second aspect of the present application, and the third aspect of the present application for use as medicine or for treating CDK-mediated cancer. The phosphate, the crystal form III described in the fourth aspect of the application, the crystal form IV described in the fifth aspect of the application or the pharmaceutical composition described in the sixth aspect of the application.
在本申请的一些实施方案中,上述癌症包括卵巢癌、乳腺癌、急性髓细胞白血病(AML)、慢性淋巴细胞白血病(CLL)或小淋巴细胞淋巴瘤(SLL)。In some embodiments of the present application, the cancer includes ovarian cancer, breast cancer, acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), or small lymphocytic lymphoma (SLL).
在本申请的一些实施方案中,上述癌症为乳腺癌。In some embodiments of the present application, the cancer is breast cancer.
本申请第十方面还提供了本申请第一方面所述的晶型I的制备方法,包括以下步骤:The tenth aspect of this application also provides a preparation method for the crystal form I described in the first aspect of this application, including the following steps:
(a)将化合物A溶于第一溶剂中,得到溶液;(a) Dissolve compound A in the first solvent to obtain a solution;
(b)向步骤(a)的所述溶液中加入第二溶剂,搅拌一定时间;(b) Add a second solvent to the solution in step (a) and stir for a certain period of time;
(c)析出固体后,过滤,收集,干燥固体。(c) After the solid is precipitated, filter, collect, and dry the solid.
在本申请的一些实施方案中,上述制备方法中第一溶剂选自丙酮、甲醇、四氢呋喃、乙酸乙酯、乙腈以及二氯甲烷中的至少一种。In some embodiments of the present application, the first solvent in the above preparation method is selected from at least one of acetone, methanol, tetrahydrofuran, ethyl acetate, acetonitrile and dichloromethane.
在本申请的一些实施方案中,上述制备方法中第二溶剂选自水、正庚烷、正己烷中的至少一种。In some embodiments of the present application, the second solvent in the above preparation method is selected from at least one of water, n-heptane, and n-hexane.
在本申请的一些实施方案中,上述制备方法中,搅拌的一定时间为1-3小时,优选为2小时。In some embodiments of the present application, in the above preparation method, the certain stirring time is 1-3 hours, preferably 2 hours.
技术效果Technical effect
本申请中的晶型具有较好的化学稳定性、物理稳定性以及较低的吸湿性,受温度、湿度和光照影响较小,便于储存及制剂开发。The crystal form in this application has good chemical stability, physical stability and low hygroscopicity, is less affected by temperature, humidity and light, and is convenient for storage and formulation development.
附图说明Description of the drawings
图1为式(A)化合物的晶型I的XRPD(X-射线粉末衍射)谱图。Figure 1 is an XRPD (X-ray powder diffraction) spectrum of crystal form I of the compound of formula (A).
图2为式(A)化合物的晶型I的TGA-DSC(差示扫描量热-热重分析)谱图。Figure 2 is a TGA-DSC (differential scanning calorimetry-thermogravimetric analysis) spectrum of crystal form I of the compound of formula (A).
图3为式(A)化合物的晶型II的XRPD谱图。Figure 3 is the XRPD spectrum of crystal form II of the compound of formula (A).
图4为式(A)化合物的晶型II的TGA-DSC谱图。Figure 4 is a TGA-DSC spectrum of crystal form II of the compound of formula (A).
图5为式(A)化合物的晶型I的DVS(动态气相吸附)谱图。Figure 5 is a DVS (dynamic vapor phase adsorption) spectrum of crystal form I of the compound of formula (A).
图6为式(A)化合物的晶型I的DVS实验前后XRPD对比谱图。Figure 6 is a comparative XRPD spectrum of crystal form I of the compound of formula (A) before and after the DVS experiment.
图7为式(A)化合物的晶型I的稳定性实验前后XRPD对比谱图。Figure 7 is a comparative XRPD spectrum before and after the stability experiment of the crystal form I of the compound of formula (A).
图8为式(A)化合物的磷酸盐的晶型III的XRPD谱图。Figure 8 is an XRPD spectrum of Form III of the phosphate salt of the compound of formula (A).
图9为式(A)化合物的磷酸盐的晶型III的TGA-DSC谱图。Figure 9 is a TGA-DSC spectrum of Form III of the phosphate salt of the compound of formula (A).
图10为式(A)化合物的磷酸盐的晶型IV的XRPD谱图。Figure 10 is an XRPD spectrum of Form IV of the phosphate salt of the compound of formula (A).
图11为式(A)化合物的磷酸盐的晶型IV的TGA-DSC谱图。Figure 11 is a TGA-DSC spectrum of Form IV of the phosphate salt of the compound of formula (A).
图12为式(A)化合物的磷酸盐的晶型IV的DVS谱图。Figure 12 is a DVS spectrum of Form IV of the phosphate salt of the compound of formula (A).
图13为式(A)化合物的磷酸盐的晶型IV的稳定性实验前后XRPD对比谱图。Figure 13 is a comparative XRPD spectrum before and after the stability experiment of the phosphate form IV of the compound of formula (A).
具体实施方式Detailed ways
下面通过实施例对本申请进行详细描述,但并不意味着对本申请任何不利限制。本申请的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申请的实施例。对本领域的技术人员而言,在不脱离本申请精神和范围的情况下针对本申请具体实施方式进行各种变化和改进将是显而易见的。The present application is described in detail through examples below, but does not mean any adverse limitations to the present application. The compounds of the present application can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combining them with other chemical synthesis methods, and methods well known to those skilled in the art. Equivalent alternatives and preferred implementations include but are not limited to the embodiments of this application. It will be apparent to those skilled in the art that various changes and improvements can be made to the specific embodiments of the present application without departing from the spirit and scope of the present application.
定义和说明Definition and Description
除非另有说明,本文所用的下列术语和短语旨在具有下列含义。一个特定的术语或短语在没有特别 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。Unless otherwise stated, the following terms and phrases used herein are intended to have the following meanings. a specific term or phrase in no particular Defined situations should not be considered uncertain or unclear, but should be understood in their ordinary meaning.
术语“可药用的载体”是指本领域通常可接受的用于将生物活性药剂递送给动物、特别是哺乳动物的介质,根据给药方式和剂型的性质包括例如佐剂、赋形剂或赋形物,例如稀释剂、防腐剂、填充剂、流动调节剂、崩解剂、润湿剂、乳化剂、助悬剂、甜味剂、调味剂、芳香剂、抗菌剂、抗真菌剂、润滑剂和分散剂。药学上可接受的载体在本领域普通技术人员的眼界范围内根据大量因素配制。其包括但不限于:配制的活性药剂的类型和性质,要将含有该药剂的组合物给药的对象,组合物的预期给药途径,和目标治疗适应症。药学上可接受的载体包括含水介质和非水介质这两者以及多种固体和半固体剂型。除了活性药剂以外,这样的载体包括许多不同的成分和添加剂,因多种原因(例如稳定活性药剂、粘合剂等)在处方中包括的这样的另外的成分对于本领域普通技术人员是众所周知的。The term "pharmaceutically acceptable carrier" refers to a medium generally accepted in the art for delivering biologically active agents to animals, especially mammals, including, for example, adjuvants, excipients, or Excipients such as diluents, preservatives, fillers, flow regulators, disintegrants, wetting agents, emulsifiers, suspending agents, sweeteners, flavorings, aromatics, antibacterial agents, antifungal agents, Lubricants and dispersants. The formulation of pharmaceutically acceptable carriers depends on a number of factors within the purview of one of ordinary skill in the art. This includes, but is not limited to: the type and nature of the formulated active agent, the subjects to whom the composition containing the agent is to be administered, the intended route of administration of the composition, and the intended therapeutic indication. Pharmaceutically acceptable carriers include both aqueous and non-aqueous media and a variety of solid and semi-solid dosage forms. Such carriers include many different ingredients and additives in addition to the active agent, and such additional ingredients are well known to those of ordinary skill in the art to be included in the formulation for a variety of reasons (e.g., to stabilize the active agent, binders, etc.) .
本领域公知,X-射线粉末衍射图谱根据测量条件的微小变化,而具有一种或多种测量误差,本申请公开或要求保护的结晶、晶体或晶型的结构可能根据试验条件、纯度、设备和本领域技术人员已知的其它常几变量在合理误差范围内表现出类似但不完全相同的分析特性。例如,粉末X-射线粉末衍射中的衍射角(2θ)通常产生±0.20°的范围内的误差,所以,本申请不仅包括粉末X-射线粉末衍射中的衍射角完全一致的结晶,还包括在±0.20°的误差范围内衍射角一致的结晶。本申请的化合物A的结晶形式并不仅限于具有与附图中所示的X-射线粉末衍射图谱相同的X射线粉末衍射图谱的晶体,具有基本上与附图中所示相同的X-射线粉末衍射图谱的任何晶体均属于本申请范围内。It is known in the art that X-ray powder diffraction patterns have one or more measurement errors depending on slight changes in measurement conditions. The structure of the crystals, crystals or crystal forms disclosed or claimed in this application may vary depending on test conditions, purity, equipment It exhibits similar but not identical analytical properties within a reasonable error range as other constant variables known to those skilled in the art. For example, the diffraction angle (2θ) in powder X-ray powder diffraction usually produces an error within the range of ±0.20°. Therefore, this application not only includes crystals with completely consistent diffraction angles in powder X-ray powder diffraction, but also includes Crystals with consistent diffraction angles within an error range of ±0.20°. The crystalline form of Compound A of the present application is not limited to crystals having the same X-ray powder diffraction pattern as shown in the accompanying drawings, and has substantially the same X-ray powder diffraction pattern as shown in the accompanying drawings. Any crystal with a diffraction pattern falls within the scope of this application.
文中出现的“与附图中所示X-射线粉末衍射图谱基本上相同的X-射线粉末衍射图谱。应了解,在该上下文中使用的术语“基本上相同”亦意指示X-射线粉末衍射图谱的2θ角度值可因伴随这些测量的固有实验变化而具有轻微变化,两者为同一晶体形式。The occurrence of "substantially the same X-ray powder diffraction pattern as the X-ray powder diffraction pattern shown in the accompanying drawings." It will be understood that the term "substantially the same" used in this context is also intended to indicate X-ray powder diffraction The 2θ angle values of the spectrum may vary slightly due to the inherent experimental variations that accompany these measurements, both for the same crystal form.
应当理解用不同类型设备或用不同的测试条件可能给出稍微不同的DSC图谱和吸热转变温度读数。DSC数据可以反应物质形态的变化,强烈的吸热峰可以表示物质发生了脱水或脱溶剂,或者发生了转晶、或者发生了熔融等;当反映熔融状态时,对应的温度即通常理解为物质的熔点。该数值将受化合物纯度、样品重量、加热速度、粒径和测试设备的校验和维修的影响。本领域技术人员可以理解,物质由固态转化为液体状态时的温度通常为一个温度范围,而非固定点值,因此无论以onset值或peak值或其他合理数值均可以表征吸热峰对应的温度或物质的熔点。晶型的最大吸热转变温度可以在上述公开的具体数值±5.0℃的范围内,优选±2.0℃的范围内。It should be understood that using different types of equipment or using different test conditions may give slightly different DSC spectra and endothermic transition temperature readings. DSC data can reflect changes in the form of a substance. Strong endothermic peaks can indicate that the substance has dehydrated or desolvated, or has undergone crystallization, or has melted. When reflecting the melting state, the corresponding temperature is usually understood as the substance. melting point. This value will be affected by compound purity, sample weight, heating rate, particle size, and calibration and maintenance of the test equipment. Those skilled in the art can understand that the temperature when a substance transforms from a solid state to a liquid state is usually a temperature range rather than a fixed point value. Therefore, the temperature corresponding to the endothermic peak can be characterized by the onset value or peak value or other reasonable values. or the melting point of a substance. The maximum endothermic transition temperature of the crystal form may be within the range of the above-disclosed specific value ±5.0°C, preferably within the range of ±2.0°C.
本申请还采用热失重分析(TGA)对晶型发生分解或升华、蒸发的程度(失去重量)与温度的关系进行了分析。应当理解同种晶型受样品纯度、粒径、不同类型设备、不同的测试方法等的影响,所得到的数值存在一定误差。晶型发生分解或升华、蒸发时的温度可以在上述公开的具体数值±3.0℃的范围内,例如±2.0℃的范围内。This application also uses thermogravimetric analysis (TGA) to analyze the relationship between the degree of decomposition, sublimation, and evaporation (loss of weight) of the crystal form and temperature. It should be understood that the same crystal form is affected by sample purity, particle size, different types of equipment, different testing methods, etc., and there will be certain errors in the values obtained. The temperature at which the crystal form decomposes, sublimates, or evaporates can be within the range of ±3.0°C of the specific numerical value disclosed above, for example, within the range of ±2.0°C.
晶型的“稳定性”包括“化学稳定性”和/或“物理稳定性”。“化学稳定性”是指该晶型在一定温度、湿度、光照条件下发生降解反应的程度,“化学稳定性”反映了该晶型在储存条件下的稳定性。“物理稳定性”是指该晶型在某些特定条件下发生固态形式转化的程度,例如在高温、高湿、研磨、压片、脱溶剂、吸附溶剂的条件下,转化为另外一种晶型,因此“物理稳定性”可以在一定程度上反应晶型在制剂等使用过程中的稳定程度。"Stability" of a crystalline form includes "chemical stability" and/or "physical stability". "Chemical stability" refers to the degree of degradation reaction of the crystal form under certain temperature, humidity, and light conditions. "Chemical stability" reflects the stability of the crystal form under storage conditions. "Physical stability" refers to the degree to which the crystal form is converted into a solid form under certain specific conditions, such as high temperature, high humidity, grinding, tableting, desolvation, and adsorption of solvents, into another crystal form. Therefore, "physical stability" can reflect to a certain extent the stability of the crystal form during the use of preparations and other processes.
本申请的结晶结构可以通过各种方法制备,包括从合适的溶剂中结晶或重结晶、升华、从熔融体中生长、从另一相固态转化、从超临界流体中结晶和射流喷雾等。结晶结构从溶剂混合物中结晶或重结晶的技术,包括溶剂蒸发、降低溶剂混合物的温度、该分子和/或盐的过饱和溶剂混合物的引晶、冻干溶剂混合物、向溶剂混合物中加入反溶剂等。The crystalline structures of the present application can be prepared by a variety of methods, including crystallization or recrystallization from a suitable solvent, sublimation, growth from a melt, solid state conversion from another phase, crystallization from a supercritical fluid, and jet spray. Techniques for crystallizing or recrystallizing a crystalline structure from a solvent mixture, including solvent evaporation, lowering the temperature of the solvent mixture, seeding of a supersaturated solvent mixture of the molecule and/or salt, lyophilizing the solvent mixture, adding an antisolvent to the solvent mixture wait.
术语“干燥”是指将所得固体中的溶剂除去的过程,包括但不限于室温自然晾干,高温烘干,真空烘干等方式。The term "drying" refers to the process of removing solvent from the obtained solid, including but not limited to natural drying at room temperature, high temperature drying, vacuum drying and other methods.
本申请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申请的实施例。The intermediate compounds of the present application can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combining them with other chemical synthesis methods, and those skilled in the art. Well-known equivalents and preferred embodiments include but are not limited to the embodiments of the present application.
在本申请实施例中,标题化合物的命名是借助Chemdraw通过化合物结构转化过来的。若化合物名称与化合物结构存在不一致的情况,可通过综合相关信息和反应路线辅助确定;无法通过其他来确认的,以给出的化合物结构式为准。In the examples of this application, the naming of the title compound was converted from the compound structure with the help of Chemdraw. If there is any inconsistency between the compound name and the compound structure, it can be determined by comprehensively integrating relevant information and reaction routes; if it cannot be confirmed through other methods, the given compound structural formula shall prevail.
本申请中部分化合物的制备方法引用了前述类似化合物的制备方法。本领域人员应当知晓,在使用或参照使用其引用的制备方法时,反应物的投料比、反应溶剂、反应温度等可根据反应物的不同,进行适当的调整。The preparation methods of some compounds in this application refer to the preparation methods of similar compounds mentioned above. Persons in the art should know that when using or referring to the preparation methods cited therein, the feed ratio of the reactants, the reaction solvent, the reaction temperature, etc. can be appropriately adjusted according to the different reactants.
本申请的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申请的实施例。 The compounds of the present application can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combining them with other chemical synthesis methods, and methods well known to those skilled in the art. Equivalent alternatives and preferred implementations include but are not limited to the embodiments of this application.
仪器及分析方法Instruments and analytical methods
1、X-射线粉末衍射(XRPD)1. X-ray powder diffraction (XRPD)
固体样品用X-射线粉末衍射仪(X’Pert PRO)进行分析,取供试品细粉适量,置于样品架凹槽中,用玻璃片压制成平整致密的平面,XRPD测量参数见表5。The solid sample is analyzed with an X-ray powder diffractometer (X'Pert PRO). Take an appropriate amount of fine powder of the test sample, place it in the groove of the sample holder, and press it into a flat and dense plane with a glass piece. The XRPD measurement parameters are shown in Table 5. .
表5 XRPD测试参数
Table 5 XRPD test parameters
2、热重分析(TGA)2. Thermogravimetric analysis (TGA)
使用TA Instrument的热重分析仪对固体进行热重分析。约1-5mg样品置于已去皮的铝制样品盘中,按照表6中所列参数对样品进行加热,使用TRIOS对数据进行分析。Thermogravimetric analysis of solids was performed using TA Instrument's thermogravimetric analyzer. Approximately 1-5 mg of sample was placed in a peeled aluminum sample pan, the sample was heated according to the parameters listed in Table 6, and the data was analyzed using TRIOS.
表6 TGA分析方法参数
Table 6 TGA analysis method parameters
3、同步热分析(TGA-DSC)3. Simultaneous thermal analysis (TGA-DSC)
使用梅特勒托利多的同步热分析仪对固体进行热重-差示扫描量热连用分析。用小勺取供试品适量置于坩埚中,使铺布均匀,称重其重量,按照表7中所列参数对样品进行加热,使用STARe对数据进行分析。Thermogravimetry-differential scanning calorimetry analysis of solids was performed using a simultaneous thermal analyzer from Mettler Toledo. Use a small spoon to take an appropriate amount of the test sample and place it in the crucible, spread it evenly, weigh it, heat the sample according to the parameters listed in Table 7, and use STARe to analyze the data.
表7 TGA-DSC分析方法参数
Table 7 TGA-DSC analysis method parameters
4、动态水分吸脱附分析(DVS)4. Dynamic moisture adsorption and desorption analysis (DVS)
使用DVS Intrinsic动态水分吸附仪对样品的吸湿性进行测定。将样品置于已去皮的样品篮中,仪器自动称重,按照表8中的参数对样品进行分析。The hygroscopicity of the samples was measured using the DVS Intrinsic dynamic moisture adsorption instrument. Place the sample into the tared sample basket, the instrument automatically weighs, and analyzes the sample according to the parameters in Table 8.
表8 DVS分析方法参数
Table 8 DVS analysis method parameters
5、离子色谱分析5. Ion chromatography analysis
仪器:离子色谱仪-电导检测器;Instrument: ion chromatograph-conductivity detector;
色谱柱:用阴离子交换色谱柱[分析柱IonpacTM AS11-HC(4mm×250mm),保护柱IonpacTM AG11-HC (4mm×50mm)];Chromatographic column: Use an anion exchange chromatographic column [analytical column Ionpac TM AS11-HC (4mm × 250mm), guard column Ionpac TM AG11-HC (4mm×50mm)];
抑制器:AERS 500 4mm或效能相当的抑制器;Suppressor: AERS 500 4mm or equivalent suppressor;
柱温:30℃;Column temperature: 30℃;
流速:1.0mL/分钟;Flow rate: 1.0mL/min;
进样量:25μL;Injection volume: 25μL;
检测池温度:35℃;Detection pool temperature: 35℃;
流动相:30mmol/L氢氧化钾溶液;Mobile phase: 30mmol/L potassium hydroxide solution;
运行时间:约20分钟。Running time: approximately 20 minutes.
实施例1:式(A)化合物的制备Example 1: Preparation of compounds of formula (A)
中间体化合物99的制备
Preparation of intermediate compound 99
步骤1:在室温下,将化合物1B-3(508毫克,1.81毫摩尔)溶于二氯甲烷(30毫升)中。随后,在冰水浴下向其中加入N,N-二异丙基乙胺(702毫克,5.43毫摩尔)和4溴-苯磺酰氯(508毫克,2.00毫摩尔)。反应液在室温下搅拌2小时后,加入水(60毫升)淬灭。混合液用二氯甲烷(20毫升×3次)萃取,合并有机相,有机相先用饱和食盐水(30毫升)洗涤,然后用无水硫酸钠干燥,过滤,最后减压浓缩。所得残余物用硅胶柱层析纯化得到830毫克N-(1-(1-(4-溴苯基)磺酰基)哌啶-4-基)-4-氯-5-(三氟甲基)嘧啶-2-胺(99-1)。Step 1: Compound 1B-3 (508 mg, 1.81 mmol) was dissolved in dichloromethane (30 mL) at room temperature. Subsequently, N,N-diisopropylethylamine (702 mg, 5.43 mmol) and 4-bromo-benzenesulfonyl chloride (508 mg, 2.00 mmol) were added thereto under an ice-water bath. After the reaction solution was stirred at room temperature for 2 hours, water (60 ml) was added to quench the mixture. The mixture was extracted with dichloromethane (20 ml × 3 times), and the organic phases were combined. The organic phase was first washed with saturated brine (30 ml), then dried over anhydrous sodium sulfate, filtered, and finally concentrated under reduced pressure. The obtained residue was purified by silica gel column chromatography to obtain 830 mg of N-(1-(1-(4-bromophenyl)sulfonyl)piperidin-4-yl)-4-chloro-5-(trifluoromethyl) Pyrimidine-2-amine (99-1).
MS(ESI)M/Z:499.0[M+H]+MS(ESI)M/Z:499.0[M+H] + .
步骤2:在密封罐中,将(S)-3-羟基四氢呋喃(42毫克,0.48毫摩尔)溶于四氢呋喃(2毫升)中。随后,在冰水浴下向其中加入氢化钠(21毫克,0.88毫摩尔)。反应15分钟后,加入化合物99-1(200毫克,0.40毫摩尔),反应液加热至100℃反应4小时后,向反应液中加入水(60毫升)淬灭。混合液用二氯甲烷(20毫升×3次)萃取,合并有机相,有机相先用饱和食盐水(30毫升)洗涤,然后用无水硫酸钠干燥,过滤,最后减压浓缩。所得残余物用硅胶柱层析纯化得到150毫克(S)-N-(1-(((4-溴苯基)磺酰基)哌啶-4-基)-4-((四氢呋喃-3-基)氧基)-5-(三氟甲基)嘧啶-2-胺(化合物99)。Step 2: In a sealed jar, dissolve (S)-3-hydroxytetrahydrofuran (42 mg, 0.48 mmol) in tetrahydrofuran (2 mL). Subsequently, sodium hydride (21 mg, 0.88 mmol) was added thereto under an ice-water bath. After reacting for 15 minutes, compound 99-1 (200 mg, 0.40 mmol) was added, and the reaction solution was heated to 100°C. After reaction for 4 hours, water (60 ml) was added to the reaction solution to quench the reaction solution. The mixture was extracted with dichloromethane (20 ml × 3 times), and the organic phases were combined. The organic phase was first washed with saturated brine (30 ml), then dried over anhydrous sodium sulfate, filtered, and finally concentrated under reduced pressure. The obtained residue was purified by silica gel column chromatography to obtain 150 mg of (S)-N-(1-(((4-bromophenyl)sulfonyl)piperidin-4-yl)-4-((tetrahydrofuran-3-yl) )oxy)-5-(trifluoromethyl)pyrimidin-2-amine (compound 99).
MS(ESI)M/Z:551.0[M+H]+MS(ESI)M/Z:551.0[M+H] + .
1H NMR(400MHz,DMSO-d6)δ8.29(s,1H),8.02(d,J=7.4Hz,0.6H),7.92-7.83(m,2.4H),7.73-7.65(m,2H),5.62-5.51(m,1H),3.96-3.86(m,1H),3.85-3.68(m,4H),3.63-3.46(m,2H),2.65-2.55(m,2H),2.28-2.13(m,1H),2.03-1.84(m,3H),1.64-1.48(m,2H).1H NMR (400MHz, DMSO-d6) δ8.29 (s, 1H), 8.02 (d, J = 7.4Hz, 0.6H), 7.92-7.83 (m, 2.4H), 7.73-7.65 (m, 2H), 5.62-5.51(m,1H),3.96-3.86(m,1H),3.85-3.68(m,4H),3.63-3.46(m,2H),2.65-2.55(m,2H),2.28-2.13(m ,1H),2.03-1.84(m,3H),1.64-1.48(m,2H).
化合物A的制备
Preparation of Compound A
步骤1:在氮气保护条件下,将化合物99(25g,45.3毫摩尔),(2S,6S)-2,6-二甲基哌嗪-1-羧酸叔丁酯(11.6克,45.3毫摩尔),三(二亚苄基丙酮)二钯(4.1克,4.53毫摩尔),2-二环己基磷-2,4,6-三异丙基联苯(1.3克,9.02毫摩尔)和碳酸铯(29.4克,90.6毫摩尔)溶解于1,4-二氧六环(1.25升)中。反应液加热至100℃搅拌过夜。将反应液减压浓缩,并用乙酸乙酯(3×100毫升)萃取。合并有机相,有机相先用饱和食盐水(100毫升)洗涤,无水硫酸钠干燥,过滤,浓缩。所得残余物用硅胶柱层析纯化得到18.1克叔丁基(2S,6S)-2,6-二甲基-4-(4-((S)-四氢呋喃-3-基)氧基)-5-(三氟甲基)嘧啶-2-基)氨基)哌啶-1-基)磺酰基)苯基)哌嗪-1-羧酸叔丁酯(化合物168-1)。Step 1: Under nitrogen protection conditions, compound 99 (25g, 45.3 mmol), (2S, 6S)-2,6-dimethylpiperazine-1-carboxylic acid tert-butyl ester (11.6 g, 45.3 mmol) ), tris(dibenzylideneacetone)dipalladium (4.1 g, 4.53 mmol), 2-dicyclohexylphosphon-2,4,6-triisopropylbiphenyl (1.3 g, 9.02 mmol) and carbonic acid Cesium (29.4 g, 90.6 mmol) was dissolved in 1,4-dioxane (1.25 L). The reaction solution was heated to 100°C and stirred overnight. The reaction solution was concentrated under reduced pressure, and extracted with ethyl acetate (3×100 ml). The organic phases were combined, washed with saturated brine (100 ml), dried over anhydrous sodium sulfate, filtered and concentrated. The obtained residue was purified by silica gel column chromatography to obtain 18.1 g of tert-butyl (2S, 6S)-2,6-dimethyl-4-(4-((S)-tetrahydrofuran-3-yl)oxy)-5 -(Trifluoromethyl)pyrimidin-2-yl)amino)piperidin-1-yl)sulfonyl)phenyl)piperazine-1-carboxylic acid tert-butyl ester (compound 168-1).
MS(ESI)M/Z:685.2[M+H]+MS(ESI)M/Z:685.2[M+H] + .
步骤2:将化合物168-1(18.1克,26.4毫摩尔)溶解于1,4二氧六环(100毫升)。随后,向其中加入盐酸二氧六环溶液(100毫升)。将反应液在室温下搅拌过夜。将反应液减压浓缩。所得残余物中加入和碳酸氢钠水溶液(300毫升)洗涤,混合液用乙酸乙酯(200毫升×3次)萃取,合并有机相。有机相 用饱和食盐水(100毫升)洗涤,无水硫酸钠干燥,过滤,最后减压浓缩。所得残留物经硅胶柱层析纯化得到9.2克N-(1-((4-((3S,5S)-3,5-二甲基哌嗪-1-基)苯基)磺酰基)哌啶-4-基)-4-((S)-四氢呋喃-3-基)氧基)-5-(三氟甲基)嘧啶-2-胺(化合物A)。Step 2: Compound 168-1 (18.1 g, 26.4 mmol) was dissolved in 1,4dioxane (100 mL). Subsequently, dioxane hydrochloride solution (100 ml) was added thereto. The reaction was stirred at room temperature overnight. The reaction solution was concentrated under reduced pressure. The obtained residue was washed with sodium bicarbonate aqueous solution (300 ml), the mixture was extracted with ethyl acetate (200 ml × 3 times), and the organic phases were combined. The organic phase Wash with saturated brine (100 ml), dry over anhydrous sodium sulfate, filter, and finally concentrate under reduced pressure. The obtained residue was purified by silica gel column chromatography to obtain 9.2 g of N-(1-((4-((3S,5S)-3,5-dimethylpiperazin-1-yl)phenyl)sulfonyl)piperidine -4-yl)-4-((S)-tetrahydrofuran-3-yl)oxy)-5-(trifluoromethyl)pyrimidin-2-amine (Compound A).
MS(ESI)M/Z:585.2[M+H]+MS(ESI)M/Z:585.2[M+H] + .
1H NMR(400MHz,DMSO-d6)δ8.29(s,1H),8.00(d,J=7.1Hz,0.6H),7.85(d,J=6.9Hz,0.4H),7.49(d,J=8.8Hz,2H),7.02(d,J=9.0Hz,2H),5.60-5.51(m,1H),3.95-3.83(m,1H),3.81-3.65(m,4H),3.57-3.44(m,2H),3.37-3.33(m,2H),3.21-3.09(m,2H),3.02-2.91(m,2H),2.46-2.37(m,2H),2.28-2.03(m,2H),2.02-1.84(m,3H),1.66-1.47(m,2H),1.06(d,J=6.4Hz,6H). 1 H NMR (400MHz, DMSO-d 6 ) δ8.29 (s, 1H), 8.00 (d, J = 7.1Hz, 0.6H), 7.85 (d, J = 6.9Hz, 0.4H), 7.49 (d, J=8.8Hz,2H),7.02(d,J=9.0Hz,2H),5.60-5.51(m,1H),3.95-3.83(m,1H),3.81-3.65(m,4H),3.57-3.44 (m,2H),3.37-3.33(m,2H),3.21-3.09(m,2H),3.02-2.91(m,2H),2.46-2.37(m,2H),2.28-2.03(m,2H) ,2.02-1.84(m,3H),1.66-1.47(m,2H),1.06(d,J=6.4Hz,6H).
生物学测试评价Biological test evaluation
(一)CDK体外酶学实验(1) CDK in vitro enzymology experiment
本实验采用毛细管迁移能力变化实验(MSA)的方法测试化合物对CDK1/CDK2/CDK4/CDK6/CDK7/CDK9激酶活性的抑制作用,并得出化合物对CDK1/CDK2/CDK4/CDK6/CDK7/CDK9激酶活性的半数抑制浓度IC50This experiment uses the capillary migration ability change assay (MSA) method to test the inhibitory effect of the compound on CDK1/CDK2/CDK4/CDK6/CDK7/CDK9 kinase activity, and concludes that the compound’s inhibitory effect on CDK1/CDK2/CDK4/CDK6/CDK7/CDK9 kinase The half inhibitory concentration IC 50 of activity.
1.实验材料1. Experimental materials
CDK1/CDK2/CDK4/CDK6/CDK7/CDK9购自Carna公司,Carliper底物CTD3/底物18/底物8购自吉尔生化公司,Dinaciclib/Palbociclib购自Selleckchem公司,DMSO(二甲基亚砜)购自Sigma公司,384孔板购自Corning公司。CDK1/CDK2/CDK4/CDK6/CDK7/CDK9 were purchased from Carna Company, Carliper substrate CTD3/substrate 18/substrate 8 were purchased from Gill Biochemical Company, Dinaciclib/Palbociclib were purchased from Selleckchem Company, DMSO (dimethyl sulfoxide) It was purchased from Sigma Company, and the 384-well plate was purchased from Corning Company.
2.实验方法2. Experimental methods
(1)配制1×激酶缓冲液(1×Kinase buffer)。(1) Prepare 1×Kinase buffer.
(2)化合物浓度梯度的配制:受试化合物测试浓度1μM,10μM或30μM为起始浓度,3倍稀释10个浓度,复孔检测。在384孔板原始(source)板中稀释成100倍终浓度的100%DMSO溶液。使用分液器Echo 550向目的板384孔板转移250nL 100倍终浓度的化合物。阳性和阴性对照孔加入250nL DMSO。(2) Preparation of compound concentration gradient: test compound test concentration is 1 μM, 10 μM or 30 μM is the starting concentration, dilute 3 times to 10 concentrations, and detect in duplicate. Dilute to a 100x final concentration of 100% DMSO solution in a 384-well source plate. Use the Dispenser Echo 550 to transfer 250 nL of compound at 100x the final concentration to the destination 384-well plate. Add 250nL DMSO to the positive and negative control wells.
(3)用1×Kinase buffer配制2.5倍终浓度的激酶溶液。(3) Use 1×Kinase buffer to prepare a kinase solution with 2.5 times the final concentration.
(4)在化合物孔和阳性对照孔分别加10μL的2.5倍终浓度的激酶溶液;在阴性对照孔中加10μL的1×Kinase buffer。(4) Add 10 μL of 2.5 times the final concentration of kinase solution to the compound wells and positive control wells respectively; add 10 μL of 1×Kinase buffer to the negative control wells.
(5)1000rpm离心30秒,反应板振荡混匀后室温孵育10分钟。(5) Centrifuge at 1000 rpm for 30 seconds, shake and mix the reaction plate and incubate at room temperature for 10 minutes.
(6)用1×Kinase buffer配制25/15倍终浓度的ATP和激酶基质(Kinase substrate)混合溶液。(6) Use 1×Kinase buffer to prepare a mixed solution of ATP and Kinase substrate at 25/15 times the final concentration.
(7)加入15μL的25/15倍终浓度的ATP和底物的混合溶液,起始反应。(7) Add 15 μL of a mixed solution of ATP and substrate at 25/15 times the final concentration to start the reaction.
(8)将384孔板1000rpm离心30秒,振荡混匀后室温孵育相应的时间。(8) Centrifuge the 384-well plate at 1000 rpm for 30 seconds, shake and mix, and incubate at room temperature for the corresponding time.
(9)加入30μL终止检测液停止激酶反应,1000rpm离心30秒,振荡混匀。(9) Add 30 μL of stop detection solution to stop the kinase reaction, centrifuge at 1000 rpm for 30 seconds, and shake to mix.
(10)用Caliper EZ Reader读取转化率。(10) Use Caliper EZ Reader to read the conversion rate.
计算公式:Calculation formula:
%抑制率(inhibition)=(conversion%_max-conversion%_sample)/(conversion%_max-conversion%_min)×100%% inhibition rate (inhibition)=(conversion%_max-conversion%_sample)/(conversion%_max-conversion%_min)×100%
其中:Conversion%_sample是样品的转化率读数;Conversion%_min:阴性对照孔均值,代表没有酶活孔的转化率读数;Conversion%_max:阳性对照孔均值,代表没有化合物抑制孔的转化率读数。Among them: Conversion%_sample is the conversion rate reading of the sample; Conversion%_min: the mean value of the negative control wells, representing the conversion rate reading of the wells without enzyme activity; Conversion%_max: the mean value of the positive control wells, representing the conversion rate reading of the wells without compound inhibition.
拟合量效曲线Fitted dose-response curve
以浓度的log值作为X轴,百分比抑制率为Y轴,采用分析软件GraphPad Prism 5的log(inhibitor)vs.response-Variable slope拟合量效曲线,从而得出各个化合物对酶活性的IC50(半抑制浓度)值。Taking the log value of the concentration as the X-axis and the percentage inhibition rate as the Y-axis, use the log(inhibitor) vs. response-Variable slope of the analysis software GraphPad Prism 5 to fit the dose-effect curve to obtain the IC 50 of each compound on the enzyme activity. (half inhibitory concentration) value.
计算公式:Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))。Calculation formula: Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50 -X)*HillSlope)).
3.实验结果3.Experimental results
本申请化合物对CDK激酶活性的抑制IC50数据如表9中所展示。其中:IC50<0.5nM的化合物使用AA表示,0.5nM≤IC50<2.5nM的化合物使用AB表示,2.5nM≤IC50<10nM的化合物使用AC表示,10nM≤IC50<50nM的化合物用B来标识,50nM≤IC50<100nM的化合物用C来标识,100nM≤IC50<1000nM之间的化合物用D来标识,IC50>1000nM的化合物用E来标识。The inhibitory IC 50 data of the compounds of the present application on CDK kinase activity are shown in Table 9. Among them: compounds with IC 50 <0.5nM are represented by AA, compounds with 0.5nM≤IC 50 <2.5nM are represented by AB, compounds with 2.5nM≤IC 50 <10nM are represented by AC, and compounds with 10nM≤IC 50 <50nM are represented by B. To identify, compounds with 50nM≤IC 50 <100nM are identified with C, compounds with 100nM≤IC 50 <1000nM are identified with D, and compounds with IC 50 >1000nM are identified with E.
表9:CDK酶学抑制结果
Table 9: CDK enzymatic inhibition results
结论:本申请的化合物A具有较好的CDK 2/4/6激酶抑制活性,尤其在CDK 2激酶的抑制活性上表现出色;本申请的化合物A相对于CDK 1/7/9激酶,可选择性抑制CDK 2/4/6激酶,尤其在CDK 2激酶的抑制活性上表现出色,其激酶选择性可达近10倍,甚至几十倍、百倍。Conclusion: Compound A of this application has good CDK 2/4/6 kinase inhibitory activity, especially excellent in the inhibitory activity of CDK 2 kinase; Compound A of this application can be selected compared to CDK 1/7/9 kinase It specifically inhibits CDK 2/4/6 kinases, especially in the inhibitory activity of CDK 2 kinase. Its kinase selectivity can reach nearly 10 times, even dozens or hundreds of times.
(二)HCC1806/NIH:OVCAR-3细胞增殖抑制实验(2) HCC1806/NIH:OVCAR-3 cell proliferation inhibition experiment
本实验采用CellTiter-Glo的方法测试化合物对HCC1806/NIH:OVCAR-3细胞增殖的抑制作用,并 得出化合物抑制细胞生长半数的浓度IC50(nM)。This experiment uses the CellTiter-Glo method to test the inhibitory effect of compounds on HCC1806/NIH:OVCAR-3 cell proliferation, and Find the concentration IC 50 (nM) at which the compound inhibits cell growth by half.
1.实验材料1. Experimental materials
HCC1806购自通派(上海)生物科技有限公司;NIH:OVCAR-3购自美国ATCC细胞库。HCC1806 was purchased from Tongpai (Shanghai) Biotechnology Co., Ltd.; NIH:OVCAR-3 was purchased from the ATCC Cell Bank of the United States.
1640培养基、胎牛血清(FBS)、Penicillin-Streptomycin、GlutaMAX-I Supplement购自GIBCO。1640 medium, fetal bovine serum (FBS), Penicillin-Streptomycin, and GlutaMAX-I Supplement were purchased from GIBCO.
PF-06873600购自Selleck公司。PF-06873600 was purchased from Selleck.
CellTiter-Glo试剂购自Promega公司。CellTiter-Glo reagent was purchased from Promega Company.
2.实验方法2. Experimental methods
1)按照每孔600/1500个细胞的密度将HCC1806/NIH:OVCAR-3细胞接种于96孔培养板,每孔100μL。1) Seed HCC1806/NIH:OVCAR-3 cells into a 96-well culture plate at a density of 600/1500 cells per well, 100 μL per well.
2)第0天(Day 0):使用Echo向培养板细胞中加入100nL梯度稀释的待测化合物,DMSO终浓度为0.5%,将培养板置于细胞培养箱中孵育168小时(37℃,5vol%CO2)。空白对照加入每孔30nL的DMSO。2) Day 0: Use Echo to add 100nL gradient dilution of the test compound to the culture plate cells. The final concentration of DMSO is 0.5%. Place the culture plate in a cell culture incubator and incubate for 168 hours (37°C, 5vol %CO 2 ). For blank control, add 30 nL DMSO to each well.
3)第7天(Day 7):每孔加入30μL Cell Titer-Glo试剂,室温避光30分钟3) Day 7: Add 30μL Cell Titer-Glo reagent to each well and keep at room temperature for 30 minutes in the dark.
4)Envision酶标仪(PerkinElmer)检测化学发光信号。4) Envision microplate reader (PerkinElmer) detects chemiluminescence signal.
使用GraphPad Prism 6软件进行数据分析,得出化合物的IC50(nM)。Data analysis was performed using GraphPad Prism 6 software to obtain the IC 50 (nM) of the compound.
实验结果和结论:经测试,本申请化合物A对HCC1806/NIH:OVCAR-3细胞系的细胞增殖IC50可小于100nM,较参比化合物PF-06873600具有更好的抑制作用。Experimental results and conclusions: After testing, the IC 50 of Compound A of the present application on the cell proliferation of the HCC1806/NIH:OVCAR-3 cell line can be less than 100nM, which has a better inhibitory effect than the reference compound PF-06873600.
(三)HCC1806人乳腺癌模型体内药效实验(3) In vivo drug efficacy experiment of HCC1806 human breast cancer model
1.实验材料:1. Experimental materials:
HCC1806购自通派(上海)生物科技有限公司;NIH:OVCAR-3购自美国ATCC细胞库。HCC1806 was purchased from Tongpai (Shanghai) Biotechnology Co., Ltd.; NIH:OVCAR-3 was purchased from the ATCC Cell Bank of the United States.
1640培养基,胎牛血清(FBS),Penicillin-Streptomycin购自GIBCO。PF-06873600购自MCE公司。1640 medium, fetal bovine serum (FBS), and Penicillin-Streptomycin were purchased from GIBCO. PF-06873600 was purchased from MCE Company.
2.实验方法:2.Experimental method:
收集对数生长期的细胞,右侧皮下接种BALB/c裸小鼠建立肿瘤模型。接种当天命名为D0,接种后第四天(D4)当平均肿瘤体积达到150mm3左右时,挑选肿瘤体积适中的入组,每组6只。分组当天开始灌胃给药。每周2-3次统计体重数据和肿瘤体积数据,绘制体重和肿瘤生长曲线。肿瘤体积V=1/2×a×b2,其中a、b分别表示肿瘤长径和短径。Cells in the logarithmic growth phase were collected and inoculated subcutaneously on the right side of BALB/c nude mice to establish a tumor model. The day of inoculation was named D0. On the fourth day after inoculation (D4), when the average tumor volume reached about 150 mm 3 , those with moderate tumor volume were selected and included in each group, with 6 animals in each group. Administration by intragastric administration was started on the day of grouping. Collect weight data and tumor volume data 2-3 times a week, and draw weight and tumor growth curves. Tumor volume V=1/2×a×b 2 , where a and b represent the long and short diameters of the tumor respectively.
实验结果和结论:经测试,施用本申请化合物A的治疗组的肿瘤得到了有效的抑制,较参比化合物PF-06873600,本申请化合物具有更好的肿瘤抑制作用,且小鼠体重没有明显降低,显示了各治疗方案(10mg/kg BID、20mg/kg BID、30mg/kg QD)下均可良好耐受。Experimental results and conclusions: After testing, the tumors in the treatment group administered with the compound A of the present application were effectively inhibited. Compared with the reference compound PF-06873600, the compound of the present application had a better tumor inhibitory effect, and the weight of the mice was not significantly reduced. , showing that each treatment regimen (10mg/kg BID, 20mg/kg BID, 30mg/kg QD) can be well tolerated.
(四)OVCAR-3人卵巢癌模型体内药效实验(4) In vivo efficacy experiment of OVCAR-3 human ovarian cancer model
1.实验材料:1. Experimental materials:
OVCAR-3购自美国ATCC细胞库。1640培养基,胎牛血清(FBS),Penicillin-Streptomycin购自GIBCO。PF-06873600购自MCE公司。OVCAR-3 was purchased from ATCC cell bank in the United States. 1640 medium, fetal bovine serum (FBS), and Penicillin-Streptomycin were purchased from GIBCO. PF-06873600 was purchased from MCE Company.
2.实验方法:2.Experimental method:
收集对数生长期的细胞,右侧皮下接种BALB/c裸小鼠建立肿瘤模型。接种当天命名为D0,接种后27天(D27)当平均肿瘤体积达到180mm3左右时,挑选肿瘤体积适中的入组,每组6只。分组当天开始灌胃给药。连续给药21天。每周2-3次统计体重数据和肿瘤体积数据,绘制体重和肿瘤生长曲线。肿瘤体积V=1/2×a×b2,其中a、b分别表示肿瘤长径和短径。Cells in the logarithmic growth phase were collected and inoculated subcutaneously on the right side of BALB/c nude mice to establish a tumor model. The day of inoculation was named D0. On the 27th day after inoculation (D27), when the average tumor volume reached about 180 mm 3 , those with moderate tumor volume were selected and included in each group, with 6 animals in each group. Administration by intragastric administration was started on the day of grouping. Administration was continued for 21 days. Collect weight data and tumor volume data 2-3 times a week, and draw weight and tumor growth curves. Tumor volume V=1/2×a×b 2 , where a and b represent the long and short diameters of the tumor respectively.
实验结果和结论:经测试,施用本申请化合物A的治疗组的肿瘤得到了有效的抑制,较参比化合物PF-06873600,本申请化合物具有更好的肿瘤抑制作用,且小鼠体重没有明显降低,显示了各治疗方案(5mg/kg BID、7.5mg/kg BID、10mg/kg QD、20mg/kg QD)下均可良好耐受。Experimental results and conclusions: After testing, the tumors in the treatment group administered with the compound A of the present application were effectively inhibited. Compared with the reference compound PF-06873600, the compound of the present application had a better tumor inhibitory effect, and the weight of the mice was not significantly reduced. , showing that each treatment regimen (5mg/kg BID, 7.5mg/kg BID, 10mg/kg QD, 20mg/kg QD) can be well tolerated.
实施例2式(A)化合物的晶型I的制备Example 2 Preparation of Crystal Form I of Compound of Formula (A)
方法1:method 1:
将约10mg化合物A室温下溶于1.0mL丙酮中。然后,在室温下将4mL纯净水滴加到药液中。析出固体后,混悬液在室温条件下搅拌两小时,然后过滤,收集固体,并在室温晾干。Approximately 10 mg of Compound A was dissolved in 1.0 mL of acetone at room temperature. Then, 4 mL of purified water was added dropwise to the medicinal solution at room temperature. After the solid precipitated, the suspension was stirred at room temperature for two hours, then filtered, and the solid was collected and dried at room temperature.
将所得固体,进行XRPD、TGA-DSC测试表征,该固体为晶型I。The obtained solid was subjected to XRPD and TGA-DSC testing and characterization. The solid was crystalline form I.
XRPD谱图如图1所示。The XRPD spectrum is shown in Figure 1.
TGA-DSC谱图如图2所示,其在200℃左右有一明显的吸热特征峰。The TGA-DSC spectrum is shown in Figure 2, which has an obvious endothermic peak around 200°C.
方法2:Method 2:
将约10mg化合物A室温下溶于1.0mL丙酮中。然后,在室温下将4mL正庚烷滴加到药液中。析出固体后,混悬液在室温条件下搅拌两小时,然后过滤,收集固体,并在室温晾干。所得固体的XRPD谱图基本如图1所示。Approximately 10 mg of Compound A was dissolved in 1.0 mL of acetone at room temperature. Then, 4 mL of n-heptane was added dropwise to the medicinal solution at room temperature. After the solid precipitated, the suspension was stirred at room temperature for two hours, then filtered, and the solid was collected and dried at room temperature. The XRPD spectrum of the obtained solid is basically as shown in Figure 1.
方法3: Method 3:
将约10mg化合物A室温下溶于2.5mL乙酸乙酯中。然后,在室温下将10mL正庚烷滴加到药液中。析出固体后,混悬液在室温条件下搅拌两小时,然后过滤,收集固体,并在室温晾干。所得固体,的XRPD谱图基本如图1所示。Approximately 10 mg of Compound A was dissolved in 2.5 mL of ethyl acetate at room temperature. Then, 10 mL of n-heptane was added dropwise to the medicinal solution at room temperature. After the solid precipitated, the suspension was stirred at room temperature for two hours, then filtered, and the solid was collected and dried at room temperature. The XRPD spectrum of the obtained solid is basically as shown in Figure 1.
方法4:Method 4:
将约10mg化合物A室温下溶于2mL甲醇中。然后,在室温下将8mL纯净水滴加到药液中。析出固体后,混悬液在室温条件下搅拌两小时,然后过滤,收集固体,并在室温晾干。所得固体的XRPD谱图基本如图1所示。Approximately 10 mg of Compound A was dissolved in 2 mL of methanol at room temperature. Then, 8 mL of purified water was added dropwise to the medicinal solution at room temperature. After the solid precipitated, the suspension was stirred at room temperature for two hours, then filtered, and the solid was collected and dried at room temperature. The XRPD spectrum of the obtained solid is basically as shown in Figure 1.
方法5:Method 5:
将约10mg化合物A室温下溶于0.5mL四氢呋喃中。然后,在室温下将2.0mL正庚烷滴加到药液中。析出固体后,混悬液在室温条件下搅拌两小时,然后过滤,收集固体,并在室温晾干。所得固体的XRPD谱图基本如图1所示。About 10 mg of compound A was dissolved in 0.5 mL of tetrahydrofuran at room temperature. Then, 2.0 mL of n-heptane was added dropwise to the medicinal solution at room temperature. After the solid precipitated, the suspension was stirred at room temperature for two hours, then filtered, and the solid was collected and dried at room temperature. The XRPD spectrum of the obtained solid is basically as shown in Figure 1.
方法6:Method 6:
将约10mg化合物A室温下溶于3.0mL乙腈中。然后,在室温下将15mL纯净水滴加到药液中。析出固体后,混悬液在室温条件下搅拌两小时,然后过滤,收集固体,并在室温晾干。所得固体的XRPD谱图基本如图1所示。Approximately 10 mg of Compound A was dissolved in 3.0 mL of acetonitrile at room temperature. Then, 15 mL of purified water was added dropwise to the medicinal solution at room temperature. After the solid precipitated, the suspension was stirred at room temperature for two hours, then filtered, and the solid was collected and dried at room temperature. The XRPD spectrum of the obtained solid is basically as shown in Figure 1.
方法7:Method 7:
将约10mg化合物A室温下溶于0.5mL二氯甲烷中。然后,在室温下将2.0mL纯净水滴加到药液中。析出固体后,混悬液在室温条件下搅拌两小时,然后过滤,收集固体,并在室温晾干。所得固体的XRPD谱图基本如图1所示。Approximately 10 mg of Compound A was dissolved in 0.5 mL of methylene chloride at room temperature. Then, 2.0 mL of purified water was added dropwise to the medicinal solution at room temperature. After the solid precipitated, the suspension was stirred at room temperature for two hours, then filtered, and the solid was collected and dried at room temperature. The XRPD spectrum of the obtained solid is basically as shown in Figure 1.
实施例3式(A)化合物的晶型II的制备Example 3 Preparation of Crystalline Form II of Compound of Formula (A)
将约10mg化合物A室温下溶于1mL1,4-二氧六环中,过滤,将滤液置于室温下通风处挥发,待挥干后收集固体,并在室温晾干。Dissolve about 10 mg of compound A in 1 mL of 1,4-dioxane at room temperature, filter, and place the filtrate in a ventilated place at room temperature to evaporate. After evaporation, collect the solid and dry it at room temperature.
将所得固体,进行XRPD、TGA-DSC测试表征,该固体为晶型II。The obtained solid was subjected to XRPD and TGA-DSC testing and characterization, and the solid was crystalline form II.
XRPD谱图如图3所示。The XRPD spectrum is shown in Figure 3.
TGA-DSC谱图如图4所示。The TGA-DSC spectrum is shown in Figure 4.
引湿性实验Hygroscopicity test
参照中国药典中的《药物引湿性试验指导原则》,测试晶型I的水分吸附/脱附数据。Refer to the "Guiding Principles for Drug Hygroscopicity Testing" in the Chinese Pharmacopoeia to test the moisture adsorption/desorption data of crystalline form I.
图5为晶型I的DVS曲线图,图6为晶型I DVS测试前后的XRPD谱图。DVS结果表明,晶型I在80%RH时吸湿增重0.14%,在90%RH时吸湿增重0.16%,说明该晶型几乎无引湿性,测试DVS实验后剩余固体的XRPD,其结果表明晶型未发生改变。Figure 5 is the DVS curve of Form I, and Figure 6 is the XRPD spectrum of Form I before and after the DVS test. The DVS results show that the crystalline form I absorbs moisture and gains weight by 0.14% at 80% RH and 0.16% at 90%RH, indicating that the crystalline form has almost no hygroscopicity. The XRPD of the remaining solid after the DVS experiment was tested, and the results showed that The crystal form has not changed.
稳定性实验stability test
参照中国药典中的《原料药物与制剂稳定性试验指导原则》,考察晶型I在不同温度以及湿度下的稳定性。在第0、5天和10天(分别记为D0、D5和D10)使用HPLC测试纯度,XRPD测试晶型。晶型I的稳定性实验前后XRPD对比谱图如图7所示。Referring to the "Guiding Principles for Stability Testing of Raw Materials and Preparations" in the Chinese Pharmacopoeia, the stability of Form I at different temperatures and humidity was investigated. HPLC was used to test the purity on days 0, 5 and 10 (recorded as D0, D5 and D10 respectively), and XRPD was used to test the crystal form. The XRPD comparison spectra of Form I before and after the stability experiment are shown in Figure 7.
实验结果如表10所示,晶型I在高温、高湿条件下其物理性质稳定,其晶型未发生变化,化学纯度未见明显降低。The experimental results are shown in Table 10. The physical properties of the crystal form I are stable under high temperature and high humidity conditions, the crystal form does not change, and the chemical purity does not decrease significantly.
表10晶型I稳定性实验结果
Table 10 Form I stability experimental results
实施例4式(A)化合物的磷酸盐的晶型III的制备Example 4 Preparation of Form III of the Phosphate of the Compound of Formula (A)
室温下将1g式(A)化合物加入50ml乙醇中,再加入125μL浓磷酸,体系溶清后迅速析出固体,室温混悬打浆过夜,过滤,滤饼50℃下真空干燥,得到N-(1-((4-((3S,5S)-3,5-二甲基哌嗪-1-基)苯基)磺酰基)哌啶-4-基)-4-((S)-四氢呋喃-3-基)氧基)-5-(三氟甲基)嘧啶-2-胺磷酸盐。Add 1g of the compound of formula (A) to 50 ml of ethanol at room temperature, and then add 125 μL of concentrated phosphoric acid. After the system is dissolved, a solid precipitates out quickly. The mixture is suspended and beaten at room temperature overnight, filtered, and the filter cake is vacuum dried at 50°C to obtain N-(1- ((4-((3S,5S)-3,5-dimethylpiperazin-1-yl)phenyl)sulfonyl)piperidin-4-yl)-4-((S)-tetrahydrofuran-3- (yl)oxy)-5-(trifluoromethyl)pyrimidin-2-amine phosphate.
1H-NMR(400MHz,DMSO-d6):δ(ppm)8.29(s,1H),8.03-7.85(m,1H),7.54-7.52(m,2H),7.11-7.09(d,J=9.2Hz,2H),5.56-5.55(m,1H),3.94-3.86(m,1H),3.80-3.71(m,4H),3.57-3.45(m,6H),3.25-3.22(m,2H),2.50-2.33(m,2H),2.24-2.14(m,1H),1.98-1.91(m,3H),1.58-1.53(m,2H),1.24-1.23(d,J=6.0Hz,6H)。 1 H-NMR (400MHz, DMSO-d 6 ): δ (ppm) 8.29 (s, 1H), 8.03-7.85 (m, 1H), 7.54-7.52 (m, 2H), 7.11-7.09 (d, J= 9.2Hz,2H),5.56-5.55(m,1H),3.94-3.86(m,1H),3.80-3.71(m,4H),3.57-3.45(m,6H),3.25-3.22(m,2H) ,2.50-2.33(m,2H),2.24-2.14(m,1H),1.98-1.91(m,3H),1.58-1.53(m,2H),1.24-1.23(d,J=6.0Hz,6H) .
离子色谱实测的PO4 3-含量为14.9%,接近单磷酸盐PO4 3-理论含量(13.9%),结合上面的核磁结果确证为单磷酸盐的形式,即式(A)化合物与磷酸的摩尔比为1:1。The PO 4 3- content measured by ion chromatography is 14.9%, which is close to the theoretical content of monophosphate PO 4 3- (13.9%). Combined with the above nuclear magnetic results, it is confirmed that it is in the form of monophosphate, that is, the compound of formula (A) and phosphoric acid The molar ratio is 1:1.
将所得固体,进行XRPD、TGA-DSC测试表征,该固体为晶型III。The obtained solid was subjected to XRPD and TGA-DSC testing and characterization, and the solid was crystalline form III.
XRPD谱图如图8所示。The XRPD spectrum is shown in Figure 8.
TGA-DSC谱图如图9所示。The TGA-DSC spectrum is shown in Figure 9.
实施例5式(A)化合物的磷酸盐的晶型IV的制备Example 5 Preparation of Form IV of the Phosphate of the Compound of Formula (A)
将1g式(A)化合物磷酸盐晶型III加入高压反应釜中,氮气置换三次,在氮气氛围下升温至160℃,保温半小时后降至室温,将氮气排空,得到固体。Add 1 g of the phosphate crystal form III of the compound of formula (A) into a high-pressure reaction kettle, replace it with nitrogen three times, raise the temperature to 160°C in a nitrogen atmosphere, keep it for half an hour and then lower it to room temperature. Evacuate the nitrogen to obtain a solid.
将所得固体,进行XRPD、TGA-DSC测试表征,该固体为晶型IV。The obtained solid was characterized by XRPD and TGA-DSC tests and found that the solid was crystal form IV.
XRPD谱图如图10所示。The XRPD spectrum is shown in Figure 10.
TGA-DSC谱图如图11所示,其DSC谱图中,在229℃处有一个特征吸热峰。The TGA-DSC spectrum is shown in Figure 11. In the DSC spectrum, there is a characteristic endothermic peak at 229°C.
引湿性实验Hygroscopicity test
参照中国药典中的《药物引湿性试验指导原则》,测试晶型IV的水分吸附/脱附数据。Refer to the "Guiding Principles for Drug Hygroscopicity Testing" in the Chinese Pharmacopoeia to test the moisture adsorption/desorption data of crystalline form IV.
图12为晶型IV的DVS曲线图,DVS结果表明,晶型IV在80%RH时吸湿增重0.398%,在90%RH时吸湿增重0.491%,说明该晶型略有引湿性。Figure 12 is the DVS curve of the crystal form IV. The DVS results show that the hygroscopic weight gain of the crystal form IV is 0.398% at 80% RH and 0.491% at 90% RH, indicating that the crystal form is slightly hygroscopic.
稳定性实验stability test
参照中国药典中的《原料药物与制剂稳定性试验指导原则》,考察晶型IV在不同温度以及湿度下的稳定性。在第0、5天和10天使用HPLC测试纯度,XRPD测试晶型。晶型IV的稳定性实验前后XRPD对比谱图如图13所示。Refer to the "Guiding Principles for Stability Testing of Raw Materials and Preparations" in the Chinese Pharmacopoeia to examine the stability of Form IV at different temperatures and humidity. HPLC was used to test purity on days 0, 5 and 10, and XRPD was used to test crystal form. The XRPD comparison spectra of Form IV before and after the stability experiment are shown in Figure 13.
实验结果如表11所示,晶型IV在高温、高湿条件下其物理、化学性质稳定,其晶型未发生变化,化学纯度未见明显降低。The experimental results are shown in Table 11. The physical and chemical properties of crystal form IV are stable under high temperature and high humidity conditions, the crystal form does not change, and the chemical purity does not decrease significantly.
表11晶型IV稳定性实验结果
Table 11 Form IV stability experimental results
以上所述仅为本申请的较佳实施例,并非用于限定本申请的保护范围。凡在本申请的精神和原则之内所作的任何修改、等同替换、改进等,均包含在本申请的保护范围内。 The above descriptions are only preferred embodiments of the present application and are not intended to limit the protection scope of the present application. Any modifications, equivalent replacements, improvements, etc. made within the spirit and principles of this application are included in the protection scope of this application.

Claims (30)

  1. 一种式(A)化合物的晶型I,
    Form I of a compound of formula (A),
    其特征在于,所述晶型I的X-射线粉末衍射图谱在2θ值为10.49°、12.10°、17.74°、19.88°、21.66°处有特征峰,2θ误差范围为±0.2°。It is characterized in that the X-ray powder diffraction pattern of the crystal form I has characteristic peaks at 2θ values of 10.49°, 12.10°, 17.74°, 19.88°, and 21.66°, and the 2θ error range is ±0.2°.
  2. 如权利要求1所述的晶型I,其特征在于,所述晶型I的X-射线粉末衍射图谱在2θ值为10.03°、10.49°、12.10°、14.28°、14.81°、17.74°、18.28°、19.88°、20.57°、21.66°、23.11°、23.76°、26.29°处有特征峰,2θ误差范围为±0.2°。The crystal form I of claim 1, wherein the X-ray powder diffraction pattern of the crystal form I has a 2θ value of 10.03°, 10.49°, 12.10°, 14.28°, 14.81°, 17.74°, 18.28 There are characteristic peaks at 19.88°, 20.57°, 21.66°, 23.11°, 23.76°, and 26.29°, and the 2θ error range is ±0.2°.
  3. 如权利要求1所述的晶型I,其特征在于,所述晶型I的X-射线粉末衍射图谱在2θ值为10.03°、10.49°、11.48°、12.10°、13.50°、14.28°、14.81°、16.16°、16.87°、17.74°、18.28°、19.88°、20.57°、21.10°、21.66°、22.13°、22.45°、23.11°、23.76°、24.67°、25.87°、26.29°、30.46°、32.57°处有特征峰,2θ误差范围为±0.2°。The crystal form I of claim 1, wherein the X-ray powder diffraction pattern of the crystal form I has a 2θ value of 10.03°, 10.49°, 11.48°, 12.10°, 13.50°, 14.28°, 14.81 °, 16.16°, 16.87°, 17.74°, 18.28°, 19.88°, 20.57°, 21.10°, 21.66°, 22.13°, 22.45°, 23.11°, 23.76°, 24.67°, 25.87°, 26.29°, 30.46°, There is a characteristic peak at 32.57°, and the 2θ error range is ±0.2°.
  4. 如权利要求1-3中任意一项所述的晶型I,其特征在于,所述晶型I的X-射线粉末衍射图谱如图1所示。The crystalline form I according to any one of claims 1 to 3, characterized in that the X-ray powder diffraction pattern of the crystalline form I is as shown in Figure 1.
  5. 如权利要求1-4中任意一项所述的晶型I,其特征在于,所述晶型I的DSC谱图在200℃处有一个吸热特征峰。The crystal form I according to any one of claims 1 to 4, characterized in that the DSC spectrum of the crystal form I has an endothermic characteristic peak at 200°C.
  6. 如权利要求1-5任意一项所述的晶型I,其特征在于,所述晶型I的TGA-DSC谱图如图2所示。The crystal form I according to any one of claims 1 to 5, characterized in that the TGA-DSC spectrum of the crystal form I is as shown in Figure 2.
  7. 一种式(A)化合物的晶型II,
    Form II of a compound of formula (A),
    其特征在于,所述晶型II的X-射线粉末衍射图谱在2θ值为8.09°、13.31°、16.59°、19.55°、21.59°、25.01°处有特征峰,2θ误差范围为±0.2°。It is characterized in that the X-ray powder diffraction pattern of the crystal form II has characteristic peaks at 2θ values of 8.09°, 13.31°, 16.59°, 19.55°, 21.59°, and 25.01°, and the 2θ error range is ±0.2°.
  8. 如权利要求7所述的晶型II,其特征在于,所述晶型II的X-射线粉末衍射图谱在2θ值为6.13°、8.09°、11.60°、13.31°、14.44°、16.59°、17.13°、18.27°、19.55°、20.93°、21.59°、22.54°、25.01°、26.92°处有特征峰,2θ误差范围为±0.2°。The crystalline form II according to claim 7, wherein the X-ray powder diffraction pattern of the crystalline form II has a 2θ value of 6.13°, 8.09°, 11.60°, 13.31°, 14.44°, 16.59°, 17.13 There are characteristic peaks at 18.27°, 19.55°, 20.93°, 21.59°, 22.54°, 25.01°, and 26.92°, and the 2θ error range is ±0.2°.
  9. 如权利要求8所述的晶型II,其特征在于,所述晶型II的X-射线粉末衍射图谱在2θ值为6.13°、8.09°、9.94°、11.60°、13.31°、14.44°、16.59°、17.13°、17.71°、18.27°、19.55°、19.98°、20.93°、22.07°、21.59°、22.54°、25.01°、25.35°、25.60°、26.92°、29.34°处有特征峰,2θ误差范围为±0.2°。The crystal form II of claim 8, wherein the X-ray powder diffraction pattern of the crystal form II has a 2θ value of 6.13°, 8.09°, 9.94°, 11.60°, 13.31°, 14.44°, and 16.59 There are characteristic peaks at °, 17.13°, 17.71°, 18.27°, 19.55°, 19.98°, 20.93°, 22.07°, 21.59°, 22.54°, 25.01°, 25.35°, 25.60°, 26.92°, 29.34°, 2θ error The range is ±0.2°.
  10. 如权利要求7-9中任意一项所述的晶型II,其特征在于,所述晶型II的X-射线粉末衍射图谱如图3所示。The crystal form II according to any one of claims 7 to 9, characterized in that the X-ray powder diffraction pattern of the crystal form II is as shown in Figure 3.
  11. 如权利要求7-10中任意一项所述的晶型II,其特征在于,所述晶型II的TGA-DSC谱图如图4所示。The crystal form II according to any one of claims 7 to 10, characterized in that the TGA-DSC spectrum of the crystal form II is as shown in Figure 4.
  12. 一种式(A)化合物的磷酸盐,其特征在于,式(A)化合物与磷酸的摩尔比例为1:1,
    A phosphate salt of a compound of formula (A), characterized in that the molar ratio of the compound of formula (A) to phosphoric acid is 1:1,
  13. 一种权利要求12所述的式(A)化合物的磷酸盐的晶型III,
    A crystalline form III of the phosphate salt of the compound of formula (A) according to claim 12,
    其特征在于,所述晶型III的X-射线粉末衍射图谱在2θ值为5.85°、8.94°、14.86°、16.00°处有特征峰,2θ误差范围为±0.2°。It is characterized in that the X-ray powder diffraction pattern of the crystal form III has characteristic peaks at 2θ values of 5.85°, 8.94°, 14.86°, and 16.00°, and the 2θ error range is ±0.2°.
  14. 如权利要求13所述的晶型III,其特征在于,所述晶型III的X-射线粉末衍射图谱在2θ值为5.85°、8.94°、10.29°、13.21°、14.86°、16.00°、17.01°、17.65°、19.25°处有特征峰,2θ误差范围为±0.2°。The crystal form III of claim 13, wherein the X-ray powder diffraction pattern of the crystal form III has a 2θ value of 5.85°, 8.94°, 10.29°, 13.21°, 14.86°, 16.00°, and 17.01 There are characteristic peaks at 17.65° and 19.25°, and the 2θ error range is ±0.2°.
  15. 如权利要求13所述的晶型III,其特征在于,所述晶型III的X-射线粉末衍射图谱在2θ值为3.43°、5.85°、8.94°、10.29°、11.69°、12.30°、13.21°、14.86°、16.00°、17.01°、17.65°、19.25°、19.83°、20.71°、21.82°、22.28°、23.86°处有特征峰,2θ误差范围为±0.2°。The crystal form III of claim 13, wherein the X-ray powder diffraction pattern of the crystal form III has a 2θ value of 3.43°, 5.85°, 8.94°, 10.29°, 11.69°, 12.30°, and 13.21 There are characteristic peaks at 14.86°, 16.00°, 17.01°, 17.65°, 19.25°, 19.83°, 20.71°, 21.82°, 22.28°, and 23.86°, and the 2θ error range is ±0.2°.
  16. 如权利要求13-15中任意一项所述的晶型III,其特征在于,所述晶型III的X-射线粉末衍射图谱如图8所示。The crystal form III according to any one of claims 13 to 15, wherein the X-ray powder diffraction pattern of the crystal form III is as shown in Figure 8.
  17. 如权利要求13-16中任意一项所述的晶型III,其特征在于,所述晶型III的TGA-DSC谱图如图9所示。The crystal form III according to any one of claims 13 to 16, characterized in that the TGA-DSC spectrum of the crystal form III is as shown in Figure 9.
  18. 一种权利要求12所述的式(A)化合物的磷酸盐的晶型IV,
    A crystalline form IV of the phosphate salt of the compound of formula (A) according to claim 12,
    其特征在于,所述晶型IV的X-射线粉末衍射图谱在2θ值为13.07°、15.66°、16.11°、16.84°、21.89°处有特征峰,2θ误差范围为±0.2°。It is characterized in that the X-ray powder diffraction pattern of the crystal form IV has characteristic peaks at 2θ values of 13.07°, 15.66°, 16.11°, 16.84°, and 21.89°, and the 2θ error range is ±0.2°.
  19. 如权利要求18所述的晶型IV,其特征在于,所述晶型IV的X-射线粉末衍射图谱在2θ值为7.81°、13.07°、15.20°、15.66°、16.11°、16.84°、19.61°、21.89°、22.16°、23.57°有特征峰,2θ误差范围为±0.2°。The crystal form IV of claim 18, wherein the X-ray powder diffraction pattern of the crystal form IV has a 2θ value of 7.81°, 13.07°, 15.20°, 15.66°, 16.11°, 16.84°, and 19.61 There are characteristic peaks at 21.89°, 22.16°, and 23.57°, and the 2θ error range is ±0.2°.
  20. 如权利要求19所述的晶型IV,其特征在于,所述晶型IV的X-射线粉末衍射图谱在2θ值为7.81°、10.97°、13.07°、14.20°、15.20°、15.66°、16.11°、16.84°、19.61°、21.89°、22.16°、22.93°、23.57°、24.70°、25.92°、27.42°、28.61°处有特征峰,2θ误差范围为±0.2°。The crystal form IV of claim 19, wherein the X-ray powder diffraction pattern of the crystal form IV has a 2θ value of 7.81°, 10.97°, 13.07°, 14.20°, 15.20°, 15.66°, 16.11 There are characteristic peaks at °, 16.84°, 19.61°, 21.89°, 22.16°, 22.93°, 23.57°, 24.70°, 25.92°, 27.42°, and 28.61°, and the 2θ error range is ±0.2°.
  21. 如权利要求18-20中任意一项所述的晶型IV,其特征在于,所述晶型IV的X-射线粉末衍射图谱如图10所示。The crystal form IV according to any one of claims 18 to 20, wherein the X-ray powder diffraction pattern of the crystal form IV is as shown in Figure 10.
  22. 如权利要求18-21中任意一项所述的晶型IV,其特征在于,所述晶型IV的DSC谱图在229℃处有一个吸热特征峰。The crystal form IV according to any one of claims 18 to 21, wherein the DSC spectrum of the crystal form IV has an endothermic characteristic peak at 229°C.
  23. 如权利要求18-22中任意一项所述的晶型IV,其特征在于,所述晶型IV的TGA-DSC谱图如图11所示。The crystal form IV according to any one of claims 18 to 22, characterized in that the TGA-DSC spectrum of the crystal form IV is as shown in Figure 11.
  24. 一种药物组合物,其包含权利要求1-6中任意一项所述的晶型I、权利要求7-11中任意一项所述的晶型II、权利要求12所述的磷酸盐、权利要求13-17中任意一项所述的晶型III或权利要求18-23中任意一项所述的晶型IV,及可药用的载体。A pharmaceutical composition comprising the crystal form I described in any one of claims 1-6, the crystal form II described in any one of claims 7-11, the phosphate salt described in claim 12, The crystal form III according to any one of claims 13-17 or the crystal form IV according to any one of claims 18-23, and a pharmaceutically acceptable carrier.
  25. 权利要求1-6中任意一项所述的晶型I、权利要求7-11中任意一项所述的晶型II、权利要求12所述的磷酸盐、权利要求13-17中任意一项所述的晶型III、权利要求18-23中任意一项所述的晶型IV或权利要求24所述的药物组合物在制备用于治疗CDK介导的癌症药物中的用途。Crystal form I according to any one of claims 1 to 6, crystal form II according to any one of claims 7 to 11, phosphate according to claim 12, any one of claims 13 to 17 The use of the crystal form III, the crystal form IV according to any one of claims 18-23 or the pharmaceutical composition according to claim 24 in the preparation of drugs for the treatment of CDK-mediated cancer.
  26. 权利要求1-6中任意一项所述的晶型I、权利要求7-11中任意一项所述的晶型II、权利要求12所述的磷酸盐、权利要求13-17中任意一项所述的晶型III、权利要求18-23中任意一项所述的晶型IV或权利要求24所述的药物组合物在治疗CDK介导的癌症中的用途。Crystal form I according to any one of claims 1 to 6, crystal form II according to any one of claims 7 to 11, phosphate according to claim 12, any one of claims 13 to 17 The use of the crystal form III, the crystal form IV according to any one of claims 18-23 or the pharmaceutical composition according to claim 24 in the treatment of CDK-mediated cancer.
  27. 如权利要求25或26所述的用途,所述癌症包括卵巢癌、乳腺癌、急性髓细胞白血病、慢性淋巴细胞白血病或小淋巴细胞淋巴瘤。The use of claim 25 or 26, wherein the cancer includes ovarian cancer, breast cancer, acute myeloid leukemia, chronic lymphocytic leukemia or small lymphocytic lymphoma.
  28. 权利要求1-6中任意一项所述的晶型I的制备方法,其特征在于,包括以下步骤: The preparation method of crystal form I according to any one of claims 1-6, characterized in that it includes the following steps:
    (a)将化合物A溶于第一溶剂中,得到溶液;(a) Dissolve compound A in the first solvent to obtain a solution;
    (b)向步骤(a)的所述溶液中加入第二溶剂,搅拌;(b) Add the second solvent to the solution in step (a) and stir;
    (c)析出固体后,过滤,收集,干燥固体。(c) After the solid is precipitated, filter, collect, and dry the solid.
  29. 如权利要求28所述的制备方法,所述第一溶剂选自丙酮、甲醇、四氢呋喃、乙酸乙酯、乙腈以及二氯甲烷中的至少一种。The preparation method according to claim 28, wherein the first solvent is selected from at least one selected from the group consisting of acetone, methanol, tetrahydrofuran, ethyl acetate, acetonitrile and dichloromethane.
  30. 如权利要求28所述的制备方法,所述第二溶剂选自水、正庚烷、正己烷、甲基叔丁基醚中的至少一种。 The preparation method according to claim 28, wherein the second solvent is selected from at least one of water, n-heptane, n-hexane, and methyl tert-butyl ether.
PCT/CN2023/111237 2022-08-05 2023-08-04 Cdk inhibitor and polymorph of phosphate thereof WO2024027825A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN202210947810 2022-08-05
CN202210936709 2022-08-05
CN202210947810.2 2022-08-05
CN202210936709.7 2022-08-05
CN202310889701 2023-07-19
CN202310889701.4 2023-07-19

Publications (1)

Publication Number Publication Date
WO2024027825A1 true WO2024027825A1 (en) 2024-02-08

Family

ID=89848552

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/111237 WO2024027825A1 (en) 2022-08-05 2023-08-04 Cdk inhibitor and polymorph of phosphate thereof

Country Status (1)

Country Link
WO (1) WO2024027825A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101490041A (en) * 2006-05-26 2009-07-22 阿斯利康(瑞典)有限公司 2-heterocycloamino-4-imidazolylpyrimidines as agents for the inhibition of cell proliferation
WO2020180959A1 (en) * 2019-03-05 2020-09-10 Incyte Corporation Pyrazolyl pyrimidinylamine compounds as cdk2 inhibitors
US20210047294A1 (en) * 2019-08-14 2021-02-18 Incyte Corporation Imidazolyl pyrimidinylamine compounds as cdk2 inhibitors
WO2021170076A1 (en) * 2020-02-28 2021-09-02 Fochon Pharmaceuticals, Ltd. Compounds as cdk2/4/6 inhibitors
WO2022166793A1 (en) * 2021-02-05 2022-08-11 上海齐鲁制药研究中心有限公司 Cdk inhibitor

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101490041A (en) * 2006-05-26 2009-07-22 阿斯利康(瑞典)有限公司 2-heterocycloamino-4-imidazolylpyrimidines as agents for the inhibition of cell proliferation
WO2020180959A1 (en) * 2019-03-05 2020-09-10 Incyte Corporation Pyrazolyl pyrimidinylamine compounds as cdk2 inhibitors
US20210047294A1 (en) * 2019-08-14 2021-02-18 Incyte Corporation Imidazolyl pyrimidinylamine compounds as cdk2 inhibitors
WO2021170076A1 (en) * 2020-02-28 2021-09-02 Fochon Pharmaceuticals, Ltd. Compounds as cdk2/4/6 inhibitors
WO2022166793A1 (en) * 2021-02-05 2022-08-11 上海齐鲁制药研究中心有限公司 Cdk inhibitor

Similar Documents

Publication Publication Date Title
AU2018201897B2 (en) Solid Forms of an Epidermal Growth Factor Kinase Inhibitor
EP2571863B1 (en) Nilotinib salts and crystalline forms thereof
US8946249B2 (en) Compound, certain novel forms thereof, pharmaceutical compositions thereof and methods for preparation and use
KR20140138941A (en) Salts of an epidermal growth factor receptor kinase inhibitor
WO2018045993A1 (en) Crystal form, salt type of substituted 2-hydro-pyrazole derivative and preparation method therefor
EP1559715B1 (en) N-{2-chloro-4-[(6,7-dimethoxy-4-quinolyl)oxy]phenyl}-n&#39;-(5-methyl-3-isoxazolyl)urea salt in crystalline form
EP3812386A1 (en) Crystal form of compound for inhibiting the activity of cdk4/6 and use thereof
CN110551142A (en) salt and crystal form of fused ring pyrimidine compound, and preparation method and application thereof
WO2023174400A1 (en) Salt of substituted amino six-membered nitric heterocyclic compound, crystal form thereof, method for preparing same, and use thereof
WO2024027825A1 (en) Cdk inhibitor and polymorph of phosphate thereof
WO2023061433A1 (en) Polymorph of egfr inhibitor
TW202408510A (en) Polymorphic forms of CDK inhibitors and phosphates thereof, preparation methods thereof, pharmaceutical compositions containing the same and uses thereof
WO2020147838A1 (en) Salt of egfr inhibitor, crystal form, and preparation method therefor
WO2023093861A1 (en) Mono-p-toluenesulfonate of axl kinase inhibitor and crystal form thereof
US20190322646A1 (en) Crystalline forms of ap26113, and preparation method thereof
TWI826013B (en) Crystal forms of imidazolinone derivatives
WO2021073494A1 (en) The salts of a compound and the crystalline forms thereof
WO2023093859A1 (en) Salt of axl kinase inhibitor, preparation method therefor and use thereof
WO2021164789A1 (en) Crystal form of pyrazolopyrimidine compound and use thereof
WO2024032558A1 (en) Salt form and crystal form of 5,6-dihydrothieno[3,4-h]quinazoline compound and preparation method therefor
WO2023202706A1 (en) Salt form and crystal form of selenium heterocyclic compound and application thereof
CN108299419B (en) Novel crystal forms of novel EGFR kinase inhibitor and preparation method thereof
KR20220159457A (en) Salt Forms, Crystalline Forms and Uses of FGFR4 Inhibitors
TW202214635A (en) Salt form, crystal form, pharmaceutical composition and use of tyrosine kinase inhibitor
EA043251B1 (en) CRYSTAL FORM OF THE COMPOUND FOR INHIBITION OF CDK4/6 ACTIVITY AND ITS APPLICATION

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: 23849529

Country of ref document: EP

Kind code of ref document: A1