WO2021219065A1 - 三氟甲基和氯双取代的磺酰胺类选择性bcl-2抑制剂的晶体 - Google Patents

三氟甲基和氯双取代的磺酰胺类选择性bcl-2抑制剂的晶体 Download PDF

Info

Publication number
WO2021219065A1
WO2021219065A1 PCT/CN2021/090885 CN2021090885W WO2021219065A1 WO 2021219065 A1 WO2021219065 A1 WO 2021219065A1 CN 2021090885 W CN2021090885 W CN 2021090885W WO 2021219065 A1 WO2021219065 A1 WO 2021219065A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
ray powder
diffraction pattern
formula
crystal
Prior art date
Application number
PCT/CN2021/090885
Other languages
English (en)
French (fr)
Inventor
丰巍伟
刘彦龙
张慧慧
刘飞
王斌
姚绎炎
陈智林
朱金蕾
陆成晖
Original Assignee
正大天晴药业集团股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 正大天晴药业集团股份有限公司 filed Critical 正大天晴药业集团股份有限公司
Priority to CN202180028439.3A priority Critical patent/CN115397825A/zh
Priority to EP21796153.1A priority patent/EP4129998A4/en
Priority to US17/922,223 priority patent/US20230174528A1/en
Publication of WO2021219065A1 publication Critical patent/WO2021219065A1/zh

Links

Images

Classifications

    • 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
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • the present application relates to crystals of trifluoromethyl and chloro-disubstituted sulfonamide selective BCL-2 inhibitors, preparation methods and their use in the prevention and treatment of anti-apoptotic protein BCL-2 related diseases such as cancer.
  • BCL-2 protein is divided into three families: BCL-2 family (its family members include BCL-2, BCL-XL, etc.), BAX family and BH3-only family, among which the BCL-2 family plays a role in anti-apoptosis , The members of the latter two families play a role in promoting apoptosis.
  • Anti-apoptotic BCL-2 family proteins are related to many diseases and are being studied as potential therapeutic drug targets. These targets for interventional therapy include, for example, BCL-2 family proteins BCL-2 and BCL-XL. Recently, inhibitors of BCL-2 family proteins have been reported in WO2012071374, WO2010138588, and WO2010065865. Although it teaches inhibitors that have high binding to the target protein, the binding affinity of the compound is only one of many parameters to be considered. One goal is to produce compounds that preferentially bind to one protein relative to another protein, that is, it is selective. In order to show this selectivity, it is well known that compounds show high binding affinity for a specific protein, and low binding affinity for another member.
  • the present application discloses that the compound of formula I has an effect on the anti-apoptotic BCL-2 protein and the anti-apoptotic BCL-XL protein.
  • the aspect also has better performance.
  • it also has better liver microsomal stability and optimized pharmacokinetic parameters, which has a better prospect for drug preparation.
  • drugs have excellent properties in the following aspects: drug activity, pharmacokinetics, bioavailability, melting point, stability, moisture absorption, solubility, etc.
  • the crystal of the compound of formula I developed in this application has low moisture absorption and good stability, including the stability of product purity and content during storage, the crystal form does not change, and it is easy to prepare. It can meet the needs of drugs in terms of production, storage and preparation.
  • this application provides a crystal of the compound of formula I,
  • the crystal of the compound of formula I of the present application is a type A crystal, which is characterized by a peak at 5.01, 6.61, 8.12, or 20.13 ⁇ 0.2° in 2 ⁇ in the X-ray powder diffraction pattern; in some embodiments , which is characterized by 2 ⁇ peaks at 5.01, 6.61, 8.12, 10.21, 14.89, 16.63 or 20.13 ⁇ 0.2° in the X-ray powder diffraction pattern; in some embodiments, it is characterized by 2 ⁇ in the X-ray powder diffraction pattern There are peaks at 5.01, 6.61, 8.12, 10.21, 12.88, 14.89, 16.63, 20.13, or 21.01 ⁇ 0.2°; in some embodiments, it is characterized by 2 ⁇ in the X-ray powder diffraction pattern at 5.01, 6.61, 8.12, 10.21. , 12.88, 13.74, 14.89, 16.63, 18.58, 20.13, 21.01, or 26.20 ⁇ 0.2°.
  • the crystal of the compound of formula I of the present application is a type A crystal, which is characterized by an X-ray powder diffraction pattern selected from 5.01, 6.61, 8.12, 10.21, 12.88, 14.89, 16.63, 20.13, or 21.01 ⁇ There are at least 7 or at least 8 diffraction peaks at 0.2° 2 ⁇ .
  • the crystal of the compound of formula I of the present application is a type A crystal, and its X-ray powder diffraction pattern is shown in FIG. 1.
  • the crystal of the compound of formula I of the present application is a type A crystal, and its DSC spectrum is shown in FIG. 2.
  • the crystal of the compound of formula I of the present application is a type A crystal, and its TG pattern is shown in FIG. 3.
  • the type A crystals of the compound of formula I can exist in the form of non-solvated crystals or in the form of solvated crystals.
  • the solvate here refers to the solvent formed by organic solvent and/or water and the corresponding compound. Compound.
  • this application provides a method for preparing the A crystal of the compound of formula I, which comprises mixing the compound of formula I with dichloromethane and methanol to clear, and concentrating and separating the solid.
  • the volume ratio of dichloromethane to methanol is selected from 20:1 to 100:1.
  • the volume-to-mass ratio of dichloromethane to the compound of formula I is 20-100 mL/g. In some embodiments, the volume-to-mass ratio of dichloromethane to the compound of formula I is 40-60 mL/g, preferably 50-60 mL/g; the volume-to-mass ratio of methanol to the compound of formula I is 0.1-10 mL/g, preferably 0.5 ⁇ 5mL/g.
  • concentration under reduced pressure is used.
  • the method for preparing the type A crystals of the compound of formula I according to the present invention includes: dissolving the compound of formula I in a mixed solution of dichloromethane and methanol, stirring and dissolving, and concentrating under reduced pressure to obtain the type A crystals.
  • the crystals of the compound of formula I of the present application are type B crystals, which are characterized by 2 ⁇ peaks at 5.31, 12.64, 19.08 or 24.21 ⁇ 0.2° in the X-ray powder diffraction pattern; in some embodiments , which is characterized by 2 ⁇ peaks at 5.31, 10.65, 12.64, 14.23, 19.08, 19.91, 22.71 or 24.21 ⁇ 0.2° in the X-ray powder diffraction pattern; in some embodiments, it is characterized by the X-ray powder diffraction pattern
  • the 2 ⁇ has a peak at 5.31, 9.80, 10.65, 12.12, 12.64, 14.23, 16.04, 18.13, 19.08, 19.91, 22.71, 24.21, or 25.93 ⁇ 0.2°; in some embodiments, it is characterized by an X-ray powder diffraction pattern Mid 2 ⁇ has peaks at 5.31, 9.80, 10.65, 12.12, 12.64, 13.57, 13.82, 14.23, 15.17, 16.04, 17.64,
  • the crystal of the compound of formula I of the present application is a type B crystal, which is characterized by an X-ray powder diffraction pattern selected from 5.31, 10.65, 12.64, 14.23, 19.08, 19.91, 22.71, or 24.21 ⁇ 0.2° There are at least 6 or at least 7 diffraction peaks at 2 ⁇ .
  • the crystal of the compound of formula I of the present application is a type B crystal, and its X-ray powder diffraction pattern is shown in FIG. 4.
  • the crystal of the compound of formula I of the present application is a type B crystal, and its DSC spectrum has the onset of an endothermic peak at 179.42 ⁇ 5°C.
  • the crystal of the compound of formula I of the present application is a type B crystal, and its DSC spectrum is shown in FIG. 5.
  • the crystal of the compound of formula I of the present application is a type B crystal, and its TG pattern is shown in FIG. 6.
  • the type B crystals of the compound of formula I can exist in the form of non-solvated crystals or in the form of solvated crystals.
  • the solvate here refers to the solvent formed by organic solvent and/or water and the corresponding compound. Compound.
  • this application provides a method for preparing the type B crystal of the compound of formula I, which includes mixing the compound of formula I with acetone to separate the solid.
  • the volume-to-mass ratio of acetone to the compound of formula I is 1-50 mL/g. In some embodiments, the above-mentioned volume-to-mass ratio is 5-20 mL/g, preferably 10 mL/g.
  • the method for preparing the type B crystal of the compound of formula I according to the present invention includes: mixing the compound of formula I with acetone, stirring at room temperature, collecting the precipitate by filtration, and vacuum drying to obtain the type B crystal.
  • the vacuum drying is performed at 40-60°C.
  • the crystal of the compound of formula I of the present application is a type C crystal, which is characterized by a peak at 5.52, 7.56, 9.22, 11.04, or 17.43 ⁇ 0.2° in 2 ⁇ in the X-ray powder diffraction pattern; in some implementations
  • the characteristic is that the 2 ⁇ in the X-ray powder diffraction pattern has peaks at 5.52, 7.56, 8.29, 9.22, 11.04, 15.81, 17.43, 18.51, or 22.59 ⁇ 0.2°; in some embodiments, the characteristic is X- The 2 ⁇ in the X-ray powder diffraction pattern has a peak at 5.52, 7.56, 8.29, 9.22, 11.04, 15.17, 15.81, 17.43, 18.51, or 20.40 ⁇ 0.2°; in some embodiments, it is characterized by 2 ⁇ in the X-ray powder diffraction pattern.
  • the crystal of the compound of formula I of the present application is a type C crystal, which is characterized by an X-ray powder diffraction pattern selected from 5.52, 7.56, 8.29, 9.22, 11.04, 15.81, 17.43, 18.51, or 22.59 ⁇ There are at least 7 or at least 8 diffraction peaks at 0.2° 2 ⁇ .
  • the crystal of the compound of formula I of the present application is a type C crystal, and its X-ray powder diffraction pattern is shown in FIG. 7.
  • the crystal of the compound of formula I of the present application is a type C crystal, and its DSC spectrum has the onset of an endothermic peak at 205.65 ⁇ 5°C.
  • the crystal of the compound of formula I of the present application is a type C crystal, and its DSC spectrum is shown in FIG. 8.
  • the crystal of the compound of formula I of the present application is a type C crystal, and its TG pattern is shown in FIG. 9.
  • the type C crystals of the compound of formula I can exist in the form of non-solvated crystals or in the form of solvated crystals.
  • the solvate here refers to the solvent formed by organic solvent and/or water and the corresponding compound. Compound.
  • the present application provides a method for preparing the type C crystal of the compound of formula I, which includes mixing the compound of formula I with methanol and methyl tert-butyl ether, and separating the solid.
  • the above preparation method wherein the volume-to-mass ratio of methanol to the compound of formula I is 5-100 mL/g, and the volume-to-mass ratio of methyl tert-butyl ether to the compound of formula I is 5-100 mL/g; in some In an embodiment, the volume-mass ratio of methanol to the compound of formula I is 10-50 mL/g, preferably 25 mL/g; the volume-mass ratio of methyl tert-butyl ether to the compound of formula I is 10-50 mL/g, preferably 25 mL/g g.
  • the method for preparing the type C crystal of the compound of formula I according to the present invention includes: mixing the compound of formula I with methanol and methyl tert-butyl ether, stirring at room temperature, collecting the precipitate by filtration, and drying to obtain The C-type crystal.
  • the vacuum drying is performed at 40-60°C.
  • the crystal of the compound of formula I of the present application is a type D crystal, which is characterized by a peak at 4.78, 12.83, 16.24, or 22.33 ⁇ 0.2° in 2 ⁇ in the X-ray powder diffraction pattern; in some embodiments , which is characterized by 2 ⁇ peaks at 4.78, 10.52, 12.83, 16.24, 18.44, 19.41, 22.33 or 23.20 ⁇ 0.2° in the X-ray powder diffraction pattern; in some embodiments, it is characterized by the X-ray powder diffraction pattern
  • the 2 ⁇ has a peak at 4.78, 9.63, 10.52, 12.83, 13.48, 15.79, 16.24, 17.89, 18.44, 19.41, 19.61, 22.33, or 23.20 ⁇ 0.2°; in some embodiments, it is characterized by an X-ray powder diffraction pattern
  • the middle 2 ⁇ has peaks at 4.78, 7.46, 9.63, 10.52, 11.17, 12.83, 13.48, 14.42, 15.79, 16.24,
  • the crystal of the compound of formula I of the present application is a type D crystal, which is characterized by an X-ray powder diffraction pattern selected from 4.78, 10.52, 12.83, 16.24, 18.44, 19.41, 22.33, or 23.20 ⁇ 0.2° There are at least 6 or at least 7 diffraction peaks at 2 ⁇ .
  • the crystal of the compound of formula I of the present application is a type D crystal, and its X-ray powder diffraction pattern is shown in FIG. 10.
  • the crystal of the compound of formula I of the present application is a type D crystal, and its DSC spectrum has the onset of an endothermic peak at 176.47 ⁇ 5°C.
  • the crystal of the compound of formula I of the present application is a type D crystal, and its DSC spectrum is shown in FIG. 11.
  • the crystal of the compound of formula I of the present application is a type D crystal, and its TG pattern is shown in FIG. 12.
  • the type D crystals of the compound of formula I can exist in the form of non-solvated crystals or in the form of solvated crystals.
  • the solvate here refers to the solvent formed by organic solvent and/or water and the corresponding compound. Compound.
  • the present application provides a method for preparing the D crystal of the compound of formula I, which includes mixing the compound of formula I with a solvent selected from tetrahydrofuran, a mixture of tetrahydrofuran and water, isopropanol or 1,4-dioxide Six rings, then separate the solids.
  • the volume mass ratio of the compound of formula I to the solvent is 5 to 200 mL/g; in some embodiments, the volume mass ratio is 20 to 100 mL/g.
  • the volume ratio of tetrahydrofuran to water is 20:1 to 0.1:1; preferably 10:1 to 0.5:1.
  • the method for preparing the D crystal of the compound of formula I according to the present invention includes: mixing the compound of formula I with a solvent, stirring at room temperature, collecting the precipitate by filtration, and drying with air to obtain the D crystal.
  • the blast drying is performed at 40-60°C.
  • the method for preparing the D crystal of the compound of formula I according to the present invention includes: mixing the compound of formula I with a solvent, stirring and dissolving at room temperature, filtering to obtain the supernatant, and transferring the supernatant to a clean In the container, slowly volatilize the solvent at room temperature to obtain the D-type crystal.
  • the crystals of the compound of formula I of the present application are E-type crystals, which are characterized by 2 ⁇ peaks at 4.22, 10.72, 15.17, or 15.65 ⁇ 0.2° in the X-ray powder diffraction pattern; in some embodiments , which is characterized by 2 ⁇ peaks at 4.22, 10.72, 14.62, 15.17, 15.65, 17.54, 19.55, 19.80 or 21.50 ⁇ 0.2° in the X-ray powder diffraction pattern; in some embodiments, it is characterized by X-ray powder In the diffraction pattern, 2 ⁇ has peaks at 4.22, 10.72, 13.82, 14.62, 15.17, 15.65, 16.92, 17.54, 19.55, 19.80, 21.50, 22.76, 23.35, or 26.06 ⁇ 0.2°; in some embodiments, the characteristic is X- The 2 ⁇ in the ray powder diffraction pattern is 4.22, 7.99, 8.75, 9.91, 10.72, 11.66, 12.75, 13.82, 14.62, 15.17
  • the crystal of the compound of formula I of the present application is an E-type crystal, which is characterized by an X-ray powder diffraction pattern selected from 4.22, 10.72, 14.62, 15.17, 15.65, 17.54, 19.55, 19.80 or 21.50 ⁇ There are at least 7 or at least 8 diffraction peaks at 0.2° 2 ⁇ .
  • the crystal of the compound of formula I of the present application is an E-type crystal, and its X-ray powder diffraction pattern is shown in FIG. 13.
  • the crystal of the compound of formula I of the present application is a type E crystal, and its DSC spectrum has the onset of an endothermic peak at 145.48 ⁇ 5°C.
  • the crystal of the compound of formula I of the present application is an E-type crystal, and its DSC spectrum is shown in FIG. 14.
  • the crystal of the compound of formula I of the present application is an E-type crystal, and its TG pattern is shown in FIG. 15.
  • the E-type crystals of the compound of formula I can exist in the form of non-solvated crystals or in the form of solvated crystals.
  • the solvate here refers to the solvent formed by organic solvent and/or water and the corresponding compound. Compound.
  • the present application provides a method for preparing the E crystal of the compound of formula I, which includes mixing the compound of formula I with p-xylene, separating and drying the solid.
  • the drying is vacuum drying at 50 to 120°C, preferably 80 to 100°C, for 5 to 10 hours.
  • the volume-mass ratio of p-xylene to the compound of formula I is 5-55 mL/g; in some embodiments, the volume-mass ratio is 10-40 mL/g, preferably 20 mL/g.
  • the method for preparing the E crystal of the compound of formula I according to the present invention includes: mixing the compound of formula I with a solvent, stirring at room temperature, collecting the solid precipitate by filtration, and vacuum drying the solid obtained by filtration, thereby The E-type crystal is obtained.
  • the solid obtained by filtration is vacuum dried at 50 to 120°C or at 80 to 100°C. In some embodiments, the solid obtained by filtration is vacuum dried at 50 to 120°C, preferably 80 to 100°C, for 5 to 10 hours.
  • the crystal of the compound of formula I of the present application is a type F crystal, which is characterized by a peak at 4.54, 9.08, or 19.24 ⁇ 0.2° in 2 ⁇ in the X-ray powder diffraction pattern; in some embodiments, it The characteristic is that the 2 ⁇ in the X-ray powder diffraction pattern has a peak at 4.54, 9.08, 14.90, 18.25, 19.24, 22.86, or 23.50 ⁇ 0.2°; in some embodiments, the characteristic is that the 2 ⁇ in the X-ray powder diffraction pattern is 4.54.
  • the crystals of the compound of formula I of the present application are type F crystals, which are characterized by an X-ray powder diffraction pattern selected from 4.54, 9.08, 13.66, 14.90, 17.46, 18.25, 19.24, 22.86, 23.50, There are at least 8 or at least 9 or at least 10 diffraction peaks at 2 ⁇ of 24.75 or 27.51 ⁇ 0.2°.
  • the crystal of the compound of formula I of the present application is a type F crystal, and its X-ray powder diffraction pattern is shown in FIG. 16.
  • the crystal of the compound of formula I of the present application is a type F crystal, and its DSC spectrum has the onset of an endothermic peak at 171.37 ⁇ 5°C.
  • the crystal of the compound of formula I of the present application is a type F crystal, and its DSC spectrum is shown in FIG. 17.
  • the crystal of the compound of formula I of the present application is a type F crystal, and its TG pattern is shown in FIG. 18.
  • the type F crystals of the compound of formula I can exist in the form of non-solvated crystals or in the form of solvated crystals.
  • the solvate here refers to the solvent formed by organic solvent and/or water and the corresponding compound. Compound.
  • this application provides a method for preparing the F crystal of the compound of formula I, which includes mixing the compound of formula I with acetonitrile or nitromethane, and separating the solids.
  • the volume mass ratio of acetonitrile or nitromethane to the compound of formula I is 5 to 200 mL/g; in some embodiments, the volume mass ratio is 20 to 100 mL/g, preferably 50 mL/g.
  • the method for preparing the F crystal of the compound of formula I of the present invention includes: mixing the compound of formula I with acetonitrile or nitromethane, stirring at room temperature, collecting the precipitate by filtration, and vacuum drying to obtain the F Type crystal.
  • the vacuum drying is performed at 40-60°C.
  • the crystals of the compound of formula I of the present application are type G crystals, which are characterized by 2 ⁇ peaks at 3.84, 10.39, 13.39, or 20.63 ⁇ 0.2° in the X-ray powder diffraction pattern; in some embodiments , which is characterized by 2 ⁇ peaks at 3.84, 10.39, 11.22, 13.39, 15.55, 16.78, 20.01 or 20.63 ⁇ 0.2° in the X-ray powder diffraction pattern; in some embodiments, it is characterized by the X-ray powder diffraction pattern
  • the 2 ⁇ has a peak at 3.84, 7.72, 9.56, 10.39, 11.22, 12.47, 13.39, 14.01, 15.55, 16.78, 19.02, 20.01, or 20.63 ⁇ 0.2°; in some embodiments, it is characterized by an X-ray powder diffraction pattern Mid 2 ⁇ is 3.84, 6.69, 7.72, 9.56, 10.39, 11.22, 12.47, 13.39, 14.01, 15.00, 15.55, 16.78,
  • the crystal of the compound of formula I of the present application is a type G crystal, which is characterized by an X-ray powder diffraction pattern selected from 3.84, 10.39, 11.22, 13.39, 15.55, 16.78, 20.01, or 20.63 ⁇ 0.2° There are at least 6 or at least 7 diffraction peaks at 2 ⁇ .
  • the crystal of the compound of formula I of the present application is a type G crystal, and its X-ray powder diffraction pattern is shown in FIG. 19.
  • the type G crystals of the compound of formula I can exist in the form of non-solvated crystals or in the form of solvated crystals.
  • the solvate here refers to the solvent formed by organic solvent and/or water and the corresponding compound. Compound.
  • the present application provides a method for preparing the G-type crystal of the compound of formula I, which includes mixing the compound of formula I with p-xylene, separating the solid, and the obtained solid without drying.
  • the volume-mass ratio of p-xylene to the compound of formula I is 60-200 mL/g; in some embodiments, the volume-mass ratio is 80-100 mL/g, preferably 94 mL/g.
  • the method for preparing the G-type crystal of the compound of formula I according to the present invention includes: mixing the compound of the formula I with p-xylene, stirring at room temperature, and collecting the precipitate by filtration to obtain the G-type crystal.
  • the crystals of the compound of formula I of the present application are H-type crystals, which are characterized by 2 ⁇ peaks at 4.65, 12.23, 14.09, or 22.04 ⁇ 0.2° in the X-ray powder diffraction pattern; in some embodiments , Which is characterized by 2 ⁇ peaks at 4.65, 9.36, 12.23, 13.33, 14.09, 17.27, 19.37, 22.04, or 22.95 ⁇ 0.2° in the X-ray powder diffraction pattern; in some embodiments, it is characterized by X-ray powder In the diffraction pattern, 2 ⁇ has peaks at 4.65, 9.36, 10.41, 12.23, 13.33, 14.09, 17.27, 18.88, 19.37, 20.58, 22.04, 22.49, 22.95, or 23.69 ⁇ 0.2°; in some embodiments, the characteristic is X- The 2 ⁇ in the ray powder diffraction pattern is 4.65, 7.20, 9.36, 10.41, 11.12, 12.23, 13.33, 14.09, 15.
  • the crystals of the compound of formula I of the present application are H-type crystals, which are characterized by an X-ray powder diffraction pattern selected from 4.65, 9.36, 12.23, 13.33, 14.09, 17.27, 19.37, 22.04, or 22.95 ⁇ There are at least 7 or at least 8 diffraction peaks at 0.2° 2 ⁇ .
  • the crystal of the compound of formula I of the present application is an H-type crystal, and its X-ray powder diffraction pattern is shown in FIG. 20.
  • the H-type crystals of the compound of formula I can exist in the form of non-solvated crystals or in the form of solvated crystals.
  • the solvate here refers to a solvent formed by an organic solvent and/or water and the corresponding compound. Compound.
  • this application provides a method for preparing the H-type crystal of the compound of formula I, which includes mixing the compound of formula I with 4-methyl-2-pentanone and separating the solid.
  • the volume-to-mass ratio of 4-methyl-2-pentanone to the compound of formula I is 5 to 200 mL/g; in some embodiments, the volume-to-mass ratio is 20 to 100 mL/g, preferably 50 mL/g.
  • the preparation method of the H-type crystal of the compound of formula I according to the present invention includes: mixing the compound of formula I with 4-methyl-2-pentanone, stirring at room temperature, collecting the precipitate by filtration, and drying under vacuum.
  • the H-type crystal is obtained.
  • the vacuum drying is performed at 50°C.
  • the present application provides a crystalline composition, wherein the crystals of the compound of formula I account for more than 50% of the weight of the crystalline composition, preferably more than 80%, more preferably more than 90%, and most preferably more than 95%.
  • the crystals of the compound of formula I are selected from the group consisting of the following crystals: type A crystals of the compound of formula I, type B crystals, type C crystals, D Type crystal, E type crystal, F type crystal, G type crystal or H type crystal.
  • the present application provides a crystalline composition, wherein the type A crystal, the type B crystal, the type C crystal, the type D crystal, the type E crystal, the type F crystal, the type G crystal, or the type H crystal of the compound of formula I account for
  • the weight of the crystalline composition is 50% or more, preferably 80% or more, more preferably 90% or more, and most preferably 95% or more.
  • the present application provides a pharmaceutical composition, which comprises a therapeutically effective amount of a crystal of a compound of formula I, or a crystal composition thereof.
  • the pharmaceutical composition of the present application further includes pharmaceutically acceptable excipients.
  • the present application describes a method for treating a disease related to the anti-apoptotic protein BCL-2 in a mammal, comprising administering a therapeutically effective amount of the crystal of the above-mentioned compound of formula I to a mammal (preferably a human) in need of the treatment , Its crystalline composition or its pharmaceutical composition.
  • this application describes the use of the crystal of the compound of formula I, its crystal composition or its pharmaceutical composition in the preparation of drugs for preventing or treating diseases related to the anti-apoptotic protein BCL-2.
  • this application describes the use of the crystal of the compound of formula I, its crystal composition or its pharmaceutical composition in the prevention or treatment of diseases related to the anti-apoptotic protein BCL-2.
  • the present application describes the crystal of the compound of the above formula I, its crystal form composition or its pharmaceutical composition for preventing or treating diseases related to the anti-apoptotic protein BCL-2.
  • the disease related to the anti-apoptotic protein BCL-2 is selected from cancer.
  • the cancer is selected from acute lymphoblastic leukemia.
  • the crystals of the compound of formula I above include type A crystals or type B crystals or type C crystals or type D crystals or type E crystals or type F crystals or type G crystals or type H crystals.
  • the crystals of the compound of formula I are selected from the group consisting of the following crystals: type A crystals, type B crystals, type C crystals, type D crystals, type E crystals, type F crystals, type G crystals of the compound of formula I And H-type crystal.
  • X-ray powder diffraction instrument model Bruker D2 Phaser; target tube-Cu.
  • TGA Thermal weight loss analysis
  • DSC Differential Scanning Calorimetry
  • the position of the peak or the relative intensity of the peak may be different due to factors such as measuring instrument, measuring method/condition and so on.
  • the measurement error of the 2 ⁇ value may be about ⁇ 0.2°. Therefore, when determining each crystal type, this error should be taken into account, and the error also belongs to the scope of this application.
  • the position of the endothermic peak of DSC may be different due to factors such as measuring instrument, measuring method/condition and so on.
  • the error may be about ⁇ 5°C, and may be about ⁇ 3°C. Therefore, when determining each crystal type, this error should be taken into account, and the error also belongs to the scope of this application.
  • “Pharmaceutically acceptable excipients” refer to inert substances that are administered together with the active ingredients to facilitate the administration of the active ingredients, including but not limited to those acceptable for use in humans or animals (such as those approved by the State Food and Drug Administration) Livestock) any glidant, sweetener, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersant, disintegrant, suspending agent, stabilizer, Isotonic agent, solvent or emulsifier.
  • auxiliary materials include calcium carbonate, calcium phosphate, various sugars and various starches, cellulose derivatives, gelatin, vegetable oils, and polyethylene glycols.
  • pharmaceutical composition refers to a mixture of one or more of the compounds of the application or their salts and pharmaceutically acceptable excipients.
  • the purpose of the pharmaceutical composition is to facilitate the administration of the compound of the present application to the organism.
  • the pharmaceutical composition of the present application can be prepared by combining the compound of the present application with suitable pharmaceutically acceptable excipients, for example, can be formulated into solid, semi-solid, liquid or gaseous preparations, such as tablets, pills, capsules, and powders. , Granules, ointments, emulsions, suspensions, suppositories, injections, inhalants, gels, microspheres and aerosols.
  • Typical routes for administering the crystalline, crystalline composition or pharmaceutical composition described herein include, but are not limited to, oral, rectal, topical, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, and intramuscular , Subcutaneous and intravenous administration.
  • the pharmaceutical composition of the present application can be manufactured by methods well known in the art, such as conventional mixing method, dissolution method, granulation method, sugar-coated pill method, grinding method, emulsification method, freeze-drying method, etc.
  • the pharmaceutical composition is in an oral form.
  • the pharmaceutical composition can be formulated by mixing the active compound with pharmaceutically acceptable excipients well known in the art. These auxiliary materials enable the compound of the present application to be formulated into tablets, pills, lozenges, sugar-coated agents, capsules, liquids, gels, slurries, suspensions, etc., for oral administration to patients.
  • the therapeutic dose of the compound of the present application may be determined according to, for example, the following: the specific use of the treatment, the way of administering the compound, the health and condition of the patient, and the judgment of the prescribing physician.
  • the ratio or concentration of the compound of the present application in the pharmaceutical composition may not be fixed, depending on various factors, including dosage, chemical properties (for example, hydrophobicity), and route of administration.
  • treatment means administering the compound or formulation described in this application to ameliorate or eliminate a disease or one or more symptoms related to the disease, and includes:
  • prevention means administering the compound, composition or formulation described in the present application to prevent a disease or one or more symptoms related to the disease, and includes: preventing the occurrence of a disease or disease state in a mammal, Especially when such mammals are susceptible to the disease state, but have not been diagnosed as having the disease state.
  • the term "therapeutically effective amount” refers to a sufficient amount of a drug or agent that is non-toxic but can achieve the desired effect.
  • the determination of the effective amount varies from person to person, and depends on the age and general conditions of the recipient, as well as the specific active substance. The appropriate effective amount in each case can be determined by those skilled in the art according to routine experiments.
  • the therapeutically effective amount of the crystals described in the present application is from about 0.0001 to 20 mg/Kg body weight/day, for example, from 0.001 to 10 mg/Kg body weight/day.
  • the dosage frequency of the crystals described in the present application is determined by the needs of the individual patient, for example, once or twice a day, or more times a day.
  • the administration may be intermittent, for example, where the patient receives a daily dose of crystals during a period of several days, and then during a period of several days or more, the patient does not receive the daily dose of crystals.
  • FIG. 1 A crystal XRPD pattern of the compound of formula I;
  • Boc stands for tert-butoxycarbonyl
  • THF stands for tetrahydrofuran
  • TBSCl stands for tert-butyldimethylchlorosilane.
  • a type B crystal of the compound of formula I is obtained.
  • the XRPD spectrum of the type B crystal is shown in Figure 4; the DSC spectrum is shown in Figure 5, and the TG spectrum is shown in Figure 6.
  • the XRPD spectrum of the D crystal is shown in Figure 10; the DSC spectrum is shown in Figure 11, and the TG spectrum is shown in Figure 12.
  • test sample Place the test sample in an open, suitable clean container, place it at 60°C for 10 days, and sample on the 5th and 10th days;
  • test sample Place the test sample in a constant humidity airtight container, and place it for 10 days at a relative humidity of 92.5% ⁇ 5% at 25°C, and take samples on the 5th and 10th days;
  • test sample is placed in a light box equipped with a fluorescent lamp or other suitable light device (open), placed under the condition of an illuminance of 4500lx ⁇ 500lx for 10 days, and samples are taken on the 5th and 10th days;
  • Injection volume 10 ⁇ L; detection wavelength: 280nm; flow rate: 1.0mL/min; column temperature: 30°C;
  • Phase A 0.1% formic acid aqueous solution
  • Phase B Acetonitrile, gradient elution
  • sample solution Take an appropriate amount of this product, accurately weigh it, dissolve it with methanol and dilute it quantitatively to make a solution containing about 0.5 mg per 1 mL as the test solution.
  • the hygroscopicity test method and conditions refer to the guidelines for the drug hygroscopicity test of the fourth general rule 9103 of the Chinese Pharmacopoeia 2015 edition: the hygroscopicity of a drug refers to the ability or degree of moisture absorption of the substance under certain temperature and humidity conditions.
  • the test product is a solid bulk drug that meets the drug quality standards, and the test results can be used as a reference for selecting appropriate drug packaging and storage conditions.
  • test product Take an appropriate amount of the test product and spread it flat in the above weighing bottle.
  • the thickness of the test product is generally about 1mm, and the weight is accurately weighed (m2).
  • dilution buffer in the kit (model: BCL-2/BAK(BH3) BINDING ASSAY KITS, from cisbio) to dilute the 500nM Tag1-BCL-2 protein stock solution to 5nM, and at the same time dilute the 20 ⁇ M Tag2-BAK protein stock solution Dilute to 120nM, first add 5 ⁇ L of Tag1-BCL-2 protein diluent to each well, then add the compound of formula I dissolved in DMSO to the well with a nanoliter sampler to make the final concentration of the compound 200nM-0.0488nM, 4 times Gradient, a total of 7 concentrations, blank control wells (without enzyme) and negative control wells (containing enzyme, with solvent DMSO), set up 2 multiple wells, and finally add 5 ⁇ L of Tag2-BAK protein diluent to each well.
  • kit model: BCL-2/BAK(BH3) BINDING ASSAY KITS, from cisbio
  • dilution buffer in the kit (model: BCL-XL/BAK(BH3) BINDING ASSAY KITS, from cisbio) to dilute 300nM Tag1-BCL-XL protein mother liquor to 2nM, and 10 ⁇ M Tag2-BAK protein mother liquor at the same time Dilute to 80nM, first add 5 ⁇ L of Tag1-BCL-XL protein diluent to each well, and then add the compound of formula I dissolved in DMSO to the well with a nanoliter sampler to make the final concentration of the compound 2000nM-0.488nM, 4 times Gradient, a total of 7 concentrations, blank control wells (without enzyme) and negative control wells (containing enzyme, with solvent DMSO), set up 2 multiple wells, and finally add 5 ⁇ L of Tag2-BAK protein diluent to each well.
  • kit model: BCL-XL/BAK(BH3) BINDING ASSAY KITS, from cisbio
  • Test Example 2 The compound's inhibitory effect on the proliferation of RS4; 11 cells
  • RS4;11 cells from Nanjing Kebai
  • RS4;11 cells from Nanjing Kebai
  • RPMI basal medium + 10wt% fetal bovine serum (FBS) for cell resuspension.
  • Count with a cell counter dilute with complete medium, adjust the cell density to 2 ⁇ 10 5 cells/mL, add the same amount of RPMI basal medium to adjust the serum concentration to 5%, and the cell density to 1 ⁇ 10 5 cells/mL mL seed plate.
  • the 300 ⁇ L final incubation system contains 30 ⁇ L liver microsomes (protein concentration: 5 mg/mL), 30 ⁇ L NADPH+MgCl 2 , 3 ⁇ L test compound (prepared in acetonitrile), and 237 ⁇ L PBS buffer (pH 7.4).
  • the ratio of organic solvent (acetonitrile) is 1% (volume ratio).
  • the reaction was terminated with 300 ⁇ L of ice acetonitrile containing internal standard.
  • SD rats weighing 180-220g, were adapted for 3 to 5 days, and then randomly divided into groups, 3 rats in each group, and intragastrically administered the formula I compound solution at a dose of 5 mg/kg.
  • test animals SD rats were fasted for 12 hours before the administration, and were given food 4 hours after the administration. They were free to drink water before and after the experiment and during the experiment.
  • PK parameters Formula I compound IG 5mg/kg T max (h) 4.00 ⁇ 0.00 C max (ng/mL) 739 ⁇ 226 AUC(0-24h)(ng*h/mL) 5973 ⁇ 2021 AUC(0- ⁇ )(ng*h/mL) 6558 ⁇ 1805 t 1/2 (h) 7.62 ⁇ 2.78 MRT(0-t)(h) 7.35 ⁇ 0.45
  • test animals male beagle dogs
  • Male beagle dogs were fasted for 12 hours before the administration, and were given food 4 hours after the administration. They were free to drink water before and after the experiment and during the experiment.
  • Test Example 5 Pharmacodynamic evaluation of the test substance in RS4; 11 human B-cell leukemia subcutaneous transplantation model
  • NOD/SCID mice female, 9-10 weeks (weeks of age at the time of tumor cell inoculation), weighing 16.3-22.0 g. Purchased from Ankai Yibo Biotechnology Co., Ltd., production license number: SCXK (Beijing) 2017-0006, animal certificate number: 11402400013155. Feeding environment: SPF level. The mice were inoculated subcutaneously with 1 ⁇ 10 7 RS4;11 cells on the right front back. The day of vaccination was defined as day 0. When the average volume of the tumor is 240mm 3 , random grouping is performed according to the size of the tumor. The administration was performed according to Table 16 below.
  • Table 16 The route of administration, dosage and schedule in human B-cell leukemia RS4; 11 subcutaneous animal model
  • n number of animals; administration volume is 10 ⁇ L/g.
  • StudyDirector TM version number 3.1.399.19, supplier Studylog System, Inc., S. San Francisco, CA, USA
  • T/C% Relative tumor proliferation rate
  • TGI% (1-T/C) ⁇ 100%.
  • T and C are the relative tumor volume (RTV) or tumor weight (TW) at a specific time point in the treatment group and the control group, respectively).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

一种三氟甲基和氯双取代的磺酰胺类选择性BCL-2抑制剂的晶体,具体涉及式I化合物的晶体、制备方法及其在预防和治疗与抗凋亡蛋白BCL-2相关的疾病例如癌症中的用途。

Description

三氟甲基和氯双取代的磺酰胺类选择性BCL-2抑制剂的晶体
相关申请的引用
本申请要求于2020年04月29日向中华人民共和国国家知识产权局提交的第202010355842.4号中国专利申请的优先权和权益,在此将其全部内容以援引的方式整体并入文本中。
技术领域
本申请涉及三氟甲基和氯双取代的磺酰胺类选择性BCL-2抑制剂的晶体、制备方法及其在预防和治疗抗凋亡蛋白BCL-2相关疾病例如癌症中的用途。
背景技术
BCL-2蛋白分为三个家族:BCL-2家族(其家族成员包括BCL-2、BCL-XL等)、BAX家族和BH3-only家族,其中BCL-2家族起着抗细胞凋亡的作用,后两个家族的成员起着促细胞凋亡的作用。
抗细胞凋亡BCL-2族蛋白与许多疾病有关并且正研究作为潜在的治疗药物目标。用于介入疗法的这些目标包括,例如,BCL-2族蛋白BCL-2和BCL-XL等。最近,BCL-2族蛋白的抑制剂已经报道于WO2012071374、WO2010138588、WO2010065865。虽然其中教导了对靶蛋白具有高结合的抑制剂,但化合物结合亲合力仅仅是许多待考虑的参数之一。一个目标是产生这样的化合物:其相对于另一种蛋白,优先地结合到一种蛋白,即对其的选择性。为显示这种选择性,周知的是化合物显示对特定的蛋白的高结合亲合力,以及对另一成员的较低的结合亲合力。
本申请公开了式I化合物,对于抗细胞凋亡BCL-2蛋白及抗细胞凋亡BCL-XL蛋白的作用,其显示出较高的选择性,且在抑制抗细胞凋亡BCL-2蛋白活性方面也具有较好性能。同时,还具有较好的肝微粒体稳定性,以及优化的药代动力学参数,具有更好的成药前景。
一般希望药物在以下方面具有优良的性质:药物活性、药代动力学、生物利用度、熔点、稳定性、引湿性、溶解性等。本申请开发的式I化合物的结晶具备较低的引湿性、良好的稳定性,包括在存储过程产品纯度、含量的稳定,晶型不发生转变,易制备。可以满足药物在生产、储存和制剂等方面的需求。
Figure PCTCN2021090885-appb-000001
发明内容
一方面,本申请提供一种式I化合物的结晶,
Figure PCTCN2021090885-appb-000002
在一些实施方案中,本申请的式I化合物的结晶为A型结晶,其特征是X-射线粉末衍射图谱中2θ在5.01、6.61、8.12或20.13±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在5.01、 6.61、8.12、10.21、14.89、16.63或20.13±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在5.01、6.61、8.12、10.21、12.88、14.89、16.63、20.13或21.01±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在5.01、6.61、8.12、10.21、12.88、13.74、14.89、16.63、18.58、20.13、21.01或26.20±0.2°处有峰。
在另外一些实施方案中,本申请的式I化合物的结晶为A型结晶,其特征是X-射线粉末衍射图谱在选自5.01、6.61、8.12、10.21、12.88、14.89、16.63、20.13或21.01±0.2°的2θ处具有至少7个或至少8个衍射峰。
本申请的一些方案中,上述A型结晶的XRPD图谱中,衍射峰的峰位置及相对强度由下表1表示:
表1 A型结晶的XRPD图谱解析数据
Figure PCTCN2021090885-appb-000003
在一些实施方案中,本申请的式I化合物的结晶为A型结晶,其X-射线粉末衍射图谱如图1所示。
在一些实施方案中,本申请的式I化合物的结晶为A型结晶,其DSC图谱如图2所示。
在一些实施方案中,本申请的式I化合物的结晶为A型结晶,其TG图谱如图3所示。
式I化合物A型结晶可以是以非溶剂合物结晶的形式存在,也可以是以溶剂合物结晶的形式存在,此处的溶剂合物是指有机溶剂和/或水与相应化合物形成的溶剂合物。
在一些实施方案中,本申请提供了式I化合物A型结晶的制备方法,包括将式I化合物与二氯甲烷和甲醇混合溶清,浓缩分离固体。在一些实施方案中,上述二氯甲烷与甲醇的体积比选自20:1~100:1。
在一些实施方案中,在上述制备方法中,其中二氯甲烷与式I化合物的体积质量比为20~100mL/g。在一些实施方案中,其中二氯甲烷与式I化合物的体积质量比为40~60mL/g,优选50~60mL/g;甲醇与式I化合物的体积质量比为0.1~10mL/g,优选0.5~5mL/g。
在一些实施方式中,在上述制备方法中,采用减压浓缩。例如,本发明所述的式I化合物A型结晶的制备方法包括:将式I化合物溶于二氯甲烷和甲醇的混合溶液中,搅拌溶清,减压浓缩得到所述A型结晶。
在一些实施方案中,本申请的式I化合物的结晶为B型结晶,其特征是X-射线粉末衍射图谱中2θ在5.31、12.64、19.08或24.21±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在5.31、10.65、12.64、14.23、19.08、19.91、22.71或24.21±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在5.31、9.80、10.65、12.12、12.64、14.23、16.04、18.13、19.08、19.91、22.71、24.21或25.93±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在5.31、9.80、10.65、12.12、12.64、13.57、13.82、14.23、15.17、16.04、17.64、18.13、19.08、19.91、20.34、22.71、22.99、23.45、24.21、25.65或25.93±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在5.31、9.50、9.80、10.65、11.41、12.12、12.64、13.57、13.82、14.23、15.17、16.04、16.64、17.10、17.64、18.13、18.33、18.73、19.08、19.60、19.91、20.34、21.22、21.93、22.71、22.99、23.45、24.21、25.65或25.93±0.2°处有峰。
在另外一些实施方案中,本申请的式I化合物的结晶为B型结晶,其特征是X-射线粉末衍射图谱在选自5.31、10.65、12.64、14.23、19.08、19.91、22.71或24.21±0.2°的2θ处具有至少6或至少7个衍射峰。
本申请的一些方案中,上述B型结晶的XRPD图谱中,衍射峰的峰位置及相对强度由下表2表示:
表2 B型结晶的XRPD图谱解析数据
Figure PCTCN2021090885-appb-000004
在一些实施方案中,本申请的式I化合物的结晶为B型结晶,其X-射线粉末衍射图谱如图4所示。
在一些实施方案中,本申请的式I化合物的结晶为B型结晶,其DSC图谱在179.42±5℃处具有吸热峰的起始点。
在一些实施方案中,本申请的式I化合物的结晶为B型结晶,其DSC图谱如图5所示。
在一些实施方案中,本申请的式I化合物的结晶为B型结晶,其TG图谱如图6所示。
式I化合物B型结晶可以是以非溶剂合物结晶的形式存在,也可以是以溶剂合物结晶的形式存在,此处的溶剂合物是指有机溶剂和/或水与相应化合物形成的溶剂合物。
另一方面,本申请提供了式I化合物B型结晶的制备方法,包括将式I化合物与丙酮混合,分离固体。
在一些实施方案中,其中丙酮与式I化合物的体积质量比为1~50mL/g。在一些实施方案中,上述体积质量比为5~20mL/g,优选10mL/g。
在一些实施方案中,本发明所述的式I化合物B型结晶的制备方法包括:将式I化合物与丙酮混合,在室温下搅拌,过滤收集沉淀物,真空干燥得到所述B型结晶。在一些实施方式中,在上述制备方法中,所述真空干燥在40~60℃下进行。
在一些实施方案中,本申请的式I化合物的结晶为C型结晶,其特征是X-射线粉末衍射图谱中2θ在5.52、7.56、9.22、11.04或17.43±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在5.52、7.56、8.29、9.22、11.04、15.81、17.43、18.51或22.59±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在5.52、7.56、8.29、9.22、11.04、15.17、15.81、17.43、18.51或20.40±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在5.52、7.56、8.29、9.22、11.04、15.17、15.81、17.00、17.43、18.51、19.70、20.01、20.40、20.75或22.59±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在5.52、7.56、8.29、9.22、10.66、11.04、12.94、14.69、15.17、15.81、16.63、17.00、17.43、18.51、19.70、20.01、20.40、20.75、22.59、25.88、26.12±0.2°处有峰。
在另外一些实施方案中,本申请的式I化合物的结晶为C型结晶,其特征是X-射线粉末衍射图谱在选 自5.52、7.56、8.29、9.22、11.04、15.81、17.43、18.51或22.59±0.2°的2θ处具有至少7或至少8个衍射峰。
本申请的一些方案中,上述C型结晶的XRPD图谱中,衍射峰的峰位置及相对强度由下表3表示:
表3 C型结晶的XRPD图谱解析数据
Figure PCTCN2021090885-appb-000005
在一些实施方案中,本申请的式I化合物的结晶为C型结晶,其X-射线粉末衍射图谱如图7所示。
在一些实施方案中,本申请的式I化合物的结晶为C型结晶,其DSC图谱在205.65±5℃处具有吸热峰的起始点。
在一些实施方案中,本申请的式I化合物的结晶为C型结晶,其DSC图谱如图8所示。
在一些实施方案中,本申请的式I化合物的结晶为C型结晶,其TG图谱如图9所示。
式I化合物C型结晶可以是以非溶剂合物结晶的形式存在,也可以是以溶剂合物结晶的形式存在,此处的溶剂合物是指有机溶剂和/或水与相应化合物形成的溶剂合物。
在一些实施方案中,本申请提供了式I化合物C型结晶的制备方法,包括将式I化合物与甲醇和甲基叔丁基醚混合,分离固体。
在一些实施方案中,上述制备方法,其中甲醇与式I化合物的体积质量比为5~100mL/g,甲基叔丁基醚与式I化合物的体积质量比为5~100mL/g;在一些实施方案中,其中甲醇与式I化合物的体积质量比为10~50mL/g,优选25mL/g;甲基叔丁基醚与式I化合物的体积质量比为10~50mL/g,优选25mL/g。
在一些实施方案中,本发明所述的式I化合物C型结晶的制备方法包括:将式I化合物与甲醇和甲基叔丁基醚混合,在室温下搅拌,过滤收集沉淀物,真空干燥得到所述C型结晶。在一些实施方式中,在上述制备方法中,所述真空干燥在40~60℃下进行。
在一些实施方案中,本申请的式I化合物的结晶为D型结晶,其特征是X-射线粉末衍射图谱中2θ在4.78、12.83、16.24或22.33±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在4.78、10.52、12.83、16.24、18.44、19.41、22.33或23.20±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在4.78、9.63、10.52、12.83、13.48、15.79、16.24、17.89、18.44、19.41、19.61、22.33或23.20±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在4.78、7.46、9.63、10.52、11.17、12.83、13.48、14.42、15.79、16.24、17.89、18.44、19.41、19.61、20.44、22.33、23.20、26.48或27.05±0.2°处有峰。
在另外一些实施方案中,本申请的式I化合物的结晶为D型结晶,其特征是X-射线粉末衍射图谱在选自4.78、10.52、12.83、16.24、18.44、19.41、22.33或23.20±0.2°的2θ处具有至少6或至少7个衍射峰。
本申请的一些方案中,上述D型结晶的XRPD图谱中,衍射峰的峰位置及相对强度由下表4表示:
表4 D型结晶的XRPD解析数据
Figure PCTCN2021090885-appb-000006
在一些实施方案中,本申请的式I化合物的结晶为D型结晶,其X-射线粉末衍射图谱如图10所示。
在一些实施方案中,本申请的式I化合物的结晶为D型结晶,其DSC图谱在176.47±5℃处具有吸热峰的起始点。
在一些实施方案中,本申请的式I化合物的结晶为D型结晶,其DSC图谱如图11所示。
在一些实施方案中,本申请的式I化合物的结晶为D型结晶,其TG图谱如图12所示。
式I化合物D型结晶可以是以非溶剂合物结晶的形式存在,也可以是以溶剂合物结晶的形式存在,此处的溶剂合物是指有机溶剂和/或水与相应化合物形成的溶剂合物。
另一方面,本申请提供了式I化合物D型结晶的制备方法,包括将式I化合物与溶剂混合,所述溶剂选自四氢呋喃、四氢呋喃和水的混合、异丙醇或1,4-二氧六环,然后分离固体。
上述制备方法,其中式I化合物与溶剂的体积质量比为5~200mL/g;在一些实施方案中,上述体积质量比为20~100mL/g。
在一些实施方案中,在四氢呋喃和水的混合中,四氢呋喃与水的体积比为20:1~0.1:1;优选为10:1~0.5:1。
在一些实施方案中,本发明所述的式I化合物D型结晶的制备方法包括:将式I化合物与溶剂混合,在室温下搅拌,过滤收集沉淀物,鼓风干燥得到所述D型结晶。
在一些实施方式中,在上述制备方法中,所述鼓风干燥在40~60℃下进行。
在一些实施方案中,本发明所述的式I化合物D型结晶的制备方法包括:将式I化合物与溶剂混合,在室温下搅拌溶解,过滤得到上清液,将上清液转移至干净的容器中,在室温下使溶剂缓慢挥发,得到所述D型结晶。
在一些实施方案中,本申请的式I化合物的结晶为E型结晶,其特征是X-射线粉末衍射图谱中2θ在4.22、10.72、15.17或15.65±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在4.22、10.72、14.62、15.17、15.65、17.54、19.55、19.80或21.50±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在4.22、10.72、13.82、14.62、15.17、15.65、16.92、17.54、19.55、19.80、21.50、22.76、23.35或26.06±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在4.22、7.99、8.75、9.91、10.72、11.66、12.75、13.82、14.62、15.17、15.65、16.24、16.92、17.54、19.04、19.55、19.80、20.18、21.50、22.76、23.35、26.06、26.91、29.82或30.56±0.2°处有峰。
在另外一些实施方案中,本申请的式I化合物的结晶为E型结晶,其特征是X-射线粉末衍射图谱在选自4.22、10.72、14.62、15.17、15.65、17.54、19.55、19.80或21.50±0.2°的2θ处具有至少7或至少8个衍射峰。
本申请的一些方案中,上述E型结晶的XRPD图谱中,衍射峰的峰位置及相对强度由下表5表示:
表5 E型结晶的XRPD图谱解析数据
Figure PCTCN2021090885-appb-000007
在一些实施方案中,本申请的式I化合物的结晶为E型结晶,其X-射线粉末衍射图谱如图13所示。
在一些实施方案中,本申请的式I化合物的结晶为E型结晶,其DSC图谱在145.48±5℃处具有吸热峰的起始点。
在一些实施方案中,本申请的式I化合物的结晶为E型结晶,其DSC图谱如图14所示。
在一些实施方案中,本申请的式I化合物的结晶为E型结晶,其TG图谱如图15所示。
式I化合物E型结晶可以是以非溶剂合物结晶的形式存在,也可以是以溶剂合物结晶的形式存在,此处的溶剂合物是指有机溶剂和/或水与相应化合物形成的溶剂合物。
在一些实施方案中,本申请提供了式I化合物E型结晶的制备方法,包括将式I化合物与对二甲苯混合,分离并干燥固体。
在一些实施方式中,所述干燥为在50~120℃、优选80~100℃真空干燥5~10小时。
上述制备方法,对二甲苯与式I化合物的体积质量比为5~55mL/g;在一些实施方案中,上述体积质量比为10~40mL/g,优选20mL/g。
在一些实施方案中,本发明所述的式I化合物E型结晶的制备方法包括:将式I化合物与溶剂混合,在室温下搅拌,过滤收集固体沉淀物,将过滤得到的固体真空干燥,从而得到所述E型结晶。
在一些实施方案中,将过滤得到的固体在50~120℃真空干燥或者在80~100℃真空干燥。在一些实施方案中,将过滤得到的固体在50~120℃、优选80~100℃真空干燥5~10小时。
在一些实施方案中,本申请的式I化合物的结晶为F型结晶,其特征是X-射线粉末衍射图谱中2θ在4.54、9.08或19.24±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在4.54、9.08、14.90、18.25、19.24、22.86或23.50±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在4.54、9.08、13.66、14.90、17.46、18.25、19.24、22.86、23.50、24.75或27.51±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在4.54、8.20、9.08、13.66、13.97、14.51、14.90、15.51、16.63、17.46、18.25、19.24、19.82、21.11、22.37、22.86、23.50、24.75或27.51±0.2°处有峰。
在另外一些实施方案中,本申请的式I化合物的结晶为F型结晶,其特征是X-射线粉末衍射图谱在选自4.54、9.08、13.66、14.90、17.46、18.25、19.24、22.86、23.50、24.75或27.51±0.2°的2θ处具有至少8或至少9个或至少10个衍射峰。
本申请的一些方案中,上述F型结晶的XRPD图谱中,衍射峰的峰位置及相对强度由下表6表示:
表6 F型结晶的XRPD图谱解析数据
编号 2θ角 相对强度 编号 2θ角 相对强度 编号 2θ角 相对强度
  (±0.20°)     (±0.20°)     (±0.20°)  
1 4.54 100.0% 14 16.63 9.2% 27 23.50 38.4%
2 8.20 6.4% 15 17.09 3.3% 28 23.71 7.2%
3 8.98 42.5% 16 17.46 21.7% 29 24.75 16.2%
4 9.08 58.5% 17 18.25 25.1% 30 27.10 3.7%
5 11.90 4.5% 18 18.56 3.0% 31 27.51 13.6%
6 12.87 2.9% 19 19.24 98.9% 32 27.98 4.4%
7 13.66 20.8% 20 19.82 10.2% 33 28.60 3.8%
8 13.97 6.7% 21 20.03 6.2% 34 28.98 4.4%
9 14.51 6.0% 22 20.72 2.4% 35 31.56 2.7%
10 14.90 33.1% 23 21.11 6.1% 36 32.24 2.9%
11 15.20 3.8% 24 21.68 3.4% 37 32.43 3.5%
12 15.51 5.1% 25 22.37 11.4% 38 33.16 2.2%
13 16.03 3.1% 26 22.86 30.3%      
在一些实施方案中,本申请的式I化合物的结晶为F型结晶,其X-射线粉末衍射图谱如图16所示。
在一些实施方案中,本申请的式I化合物的结晶为F型结晶,其DSC图谱在171.37±5℃处具有吸热峰的起始点。
在一些实施方案中,本申请的式I化合物的结晶为F型结晶,其DSC图谱如图17所示。
在一些实施方案中,本申请的式I化合物的结晶为F型结晶,其TG图谱如图18所示。
式I化合物F型结晶可以是以非溶剂合物结晶的形式存在,也可以是以溶剂合物结晶的形式存在,此处的溶剂合物是指有机溶剂和/或水与相应化合物形成的溶剂合物。
另一方面,本申请提供了式I化合物F型结晶的制备方法,包括将式I化合物与乙腈或硝基甲烷混合,分离固体。
上述制备方法,乙腈或硝基甲烷与式I化合物的体积质量比为5~200mL/g;在一些实施方案中,上述体积质量比为20~100mL/g,优选50mL/g。
在一些实施方案中,本发明所述的式I化合物F型结晶的制备方法包括:将式I化合物与乙腈或硝基甲烷混合,在室温下搅拌,过滤收集沉淀物,真空干燥得到所述F型结晶。在一些实施方式中,在上述制备方法中,所述真空干燥在40~60℃下进行。
在一些实施方案中,本申请的式I化合物的结晶为G型结晶,其特征是X-射线粉末衍射图谱中2θ在3.84、10.39、13.39或20.63±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在3.84、10.39、11.22、13.39、15.55、16.78、20.01或20.63±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在3.84、7.72、9.56、10.39、11.22、12.47、13.39、14.01、15.55、16.78、19.02、20.01或20.63±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在3.84、6.69、7.72、9.56、10.39、11.22、12.47、13.39、14.01、15.00、15.55、16.78、18.59、19.02、19.47、20.01、20.63、22.50、23.44、23.69、24.11、25.48或26.47±0.2°处有峰。
在另外一些实施方案中,本申请的式I化合物的结晶为G型结晶,其特征是X-射线粉末衍射图谱在选自3.84、10.39、11.22、13.39、15.55、16.78、20.01或20.63±0.2°的2θ处具有至少6或至少7个衍射峰。
本申请的一些方案中,上述G型结晶的XRPD图谱中,衍射峰的峰位置及相对强度由下表7表示:
表7 G型结晶的XRPD图谱解析数据
Figure PCTCN2021090885-appb-000008
Figure PCTCN2021090885-appb-000009
在一些实施方案中,本申请的式I化合物的结晶为G型结晶,其X-射线粉末衍射图谱如图19所示。
式I化合物G型结晶可以是以非溶剂合物结晶的形式存在,也可以是以溶剂合物结晶的形式存在,此处的溶剂合物是指有机溶剂和/或水与相应化合物形成的溶剂合物。
在一些实施方案中,本申请提供了式I化合物G型结晶的制备方法,包括将式I化合物与对二甲苯混合,分离固体,所得固体不经干燥。
上述制备方法,对二甲苯与式I化合物的体积质量比为60~200mL/g;在一些实施方案中,上述体积质量比为80~100mL/g,优选94mL/g。
在一个实施方案中,本发明所述的式I化合物G型结晶的制备方法包括:将式I化合物与对二甲苯混合,在室温下搅拌,过滤收集沉淀物,得到所述G型结晶。
在一些实施方案中,本申请的式I化合物的结晶为H型结晶,其特征是X-射线粉末衍射图谱中2θ在4.65、12.23、14.09或22.04±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在4.65、9.36、12.23、13.33、14.09、17.27、19.37、22.04或22.95±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在4.65、9.36、10.41、12.23、13.33、14.09、17.27、18.88、19.37、20.58、22.04、22.49、22.95或23.69±0.2°处有峰;在一些实施方案中,其特征是X-射线粉末衍射图谱中2θ在4.65、7.20、9.36、10.41、11.12、12.23、13.33、14.09、15.44、16.41、16.68、17.27、18.31、18.88、19.37、20.58、22.04、22.49、22.95、23.69或25.12±0.2°处有峰。
在另外一些实施方案中,本申请的式I化合物的结晶为H型结晶,其特征是X-射线粉末衍射图谱在选自4.65、9.36、12.23、13.33、14.09、17.27、19.37、22.04或22.95±0.2°的2θ处具有至少7或至少8个衍射峰。
本申请的一些方案中,上述H型结晶的XRPD图谱中,衍射峰的峰位置及相对强度由下表8表示:
表8 H型结晶的XRPD图谱解析数据
Figure PCTCN2021090885-appb-000010
Figure PCTCN2021090885-appb-000011
在一些实施方案中,本申请的式I化合物的结晶为H型结晶,其X-射线粉末衍射图谱如图20所示。
式I化合物H型结晶可以是以非溶剂合物结晶的形式存在,也可以是以溶剂合物结晶的形式存在,此处的溶剂合物是指有机溶剂和/或水与相应化合物形成的溶剂合物。
在一些实施方案中,本申请提供了式I化合物H型结晶的制备方法,包括在将式I化合物与4-甲基-2-戊酮混合,分离固体。
上述制备方法,4-甲基-2-戊酮与式I化合物的体积质量比为5~200mL/g;在一些实施方案中,上述体积质量比为20~100mL/g,优选50mL/g。
在一些实施方案中,本发明所述的式I化合物H型结晶的制备方法包括:将式I化合物与4-甲基-2-戊酮混合,在室温下搅拌,过滤收集沉淀物,真空干燥得到所述H型结晶。在一些实施方式中,在上述制备方法中,所述真空干燥在50℃下进行。
另一方面,本申请提供一种结晶组合物,其中式I化合物的结晶占结晶组合物重量的50%以上,优选为80%以上,更优选为90%以上,最优选为95%以上。在优选的实施方案中,在所述结晶组合物中,所述式I化合物的结晶选自于由以下结晶所组成的组:式I化合物的A型结晶、B型结晶、C型结晶、D型结晶、E型结晶、F型结晶、G型结晶或H型结晶。
另一方面,本申请提供一种结晶组合物,其中式I化合物的A型结晶或B型结晶或C型结晶或D型结晶或E型结晶或F型结晶或G型结晶或H型结晶占结晶组合物重量的50%以上,优选为80%以上,更优选为90%以上,最优选为95%以上。
另一方面,本申请提供一种药物组合物,其包含治疗有效量的式I化合物的结晶、或其结晶组合物。在一些实施方案中,本申请的药物组合物还包含药学上可接受的辅料。
另一方面,本申请描述了治疗哺乳动物中的与抗凋亡蛋白BCL-2相关的疾病的方法,包括对需要该治疗的哺乳动物(优选人类)给予治疗有效量的上述式I化合物的结晶、其结晶组合物或者其药物组合物。
另一方面,本申请描述了上述式I化合物的结晶、其结晶组合物或者其药物组合物在制备预防或者治疗与抗凋亡蛋白BCL-2相关的疾病的药物中的用途。
另一方面,本申请描述了上述式I化合物的结晶、其结晶组合物或者其药物组合物在预防或者治疗与抗凋亡蛋白BCL-2相关的疾病中的用途。
另一方面,本申请描述了用于预防或者治疗与抗凋亡蛋白BCL-2相关的疾病的上述式I化合物的结晶、其晶型组合物或者其药物组合物。
其中,所述与抗凋亡蛋白BCL-2相关的疾病选自癌症。所述癌症选自急性淋巴细胞白血病。
其中,上述式I化合物的结晶包括A型结晶或B型结晶或C型结晶或D型结晶或E型结晶或F型结晶或G型结晶或H型结晶。例如,上述式I化合物的结晶选自于由以下结晶所组成的组:式I化合物的A型结晶、B型结晶、C型结晶、D型结晶、E型结晶、F型结晶、G型结晶和H型结晶。
X-射线粉末衍射(XRPD):仪器型号Bruker D2 Phaser;靶管-Cu。
热失重分析(TGA):仪器型号NETZSCH TG 209F3 TGA209F3A-0449-L;温度范围:30-300℃;扫描速率:10℃/min。
差示扫描量热分析(DSC):仪器型号,TA DSC25;温度范围:40-220℃;扫描速率:10℃/min。
定义和说明
除非另有说明,本文所用的下列术语和短语旨在含有下列含义。一个特定的短语或术语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文出现商品名时,旨在指代其对应的商品或其活性成分。
需要说明的是,在X-射线粉末衍射光谱中,峰的位置或峰的相对强度可能会因为测定仪器、测定方法/条件等因素而产生差异。对任何特定的晶型,峰的位置可能存在误差,2θ值的测定误差可以为约±0.2°。因此,在确定每种晶型时,应该将此误差考虑在内,在误差内也属于本申请的范围。
需要说明的是,对于同种晶型,DSC的吸热峰出现位置可能会因为测定仪器、测定方法/条件等因素而产生差异。对任何特定的晶型,吸热峰的位置可能存在误差,误差可以为约±5℃,可以为约±3℃。因此,在确定每种晶型时,应该将此误差考虑在内,在误差内也属于本申请的范围。
所述词语“包括(comprise)”或“包含(comprise)”及其英文变体例如comprises或comprising应理解为开放的、非排他性的意义,即“包括但不限于”。
“药学上可接受的辅料”是指与活性成份一同给药的、有利于活性成份给药的惰性物质,包括但不限于国家食品药品监督管理局许可的可接受的用于人或动物(例如家畜)的任何助流剂、增甜剂、稀释剂、防腐剂、染料/着色剂、矫味增强剂、表面活性剂、润湿剂、分散剂、崩解剂、助悬剂、稳定剂、等渗剂、溶剂或乳化剂。所述辅料的非限制性实例包括碳酸钙、磷酸钙、各种糖和各类淀粉、纤维素衍生物、明胶、植物油和聚乙二醇。
术语“药物组合物”是指一种或多种本申请的化合物或其盐与药学上可接受的辅料组成的混合物。药物组合物的目的是有利于对有机体给予本申请的化合物。
本申请的药物组合物可通过将本申请的化合物与适宜的药学上可接受的辅料组合而制备,例如可配制成固态、半固态、液态或气态制剂,如片剂、丸剂、胶囊剂、粉剂、颗粒剂、膏剂、乳剂、悬浮剂、栓剂、注射剂、吸入剂、凝胶剂、微球及气溶胶等。
给予本申请所述结晶、结晶组合物或其药物组合物的典型途径包括但不限于口服、直肠、局部、吸入、肠胃外、舌下、阴道内、鼻内、眼内、腹膜内、肌内、皮下、静脉内给药。
本申请的药物组合物可以采用本领域众所周知的方法制造,如常规的混合法、溶解法、制粒法、制糖衣药丸法、磨细法、乳化法、冷冻干燥法等。
在一些实施方案中,药物组合物是口服形式。对于口服给药,可以通过将活性化合物与本领域熟知的药学上可接受的辅料混合,来配制该药物组合物。这些辅料能使本申请的化合物被配制成片剂、丸剂、锭剂、糖衣剂、胶囊剂、液体、凝胶剂、浆剂、悬浮剂等,用于对患者的口服给药。
本申请化合物的治疗剂量可根据例如以下而定:治疗的具体用途、给予化合物的方式、患者的健康和状态,以及签处方医师的判断。本申请化合物在药用组合物中的比例或浓度可不固定,取决于多种因素,它们包括剂量、化学特性(例如疏水性)和给药途径。
术语“治疗”意为将本申请所述化合物或制剂进行给药以改善或消除疾病或与所述疾病相关的一个或多个症状,且包括:
(i)抑制疾病或疾病状态,即遏制其发展;
(ii)缓解疾病或疾病状态,即使该疾病或疾病状态消退。
术语“预防”意为将本申请所述化合物、组合物或制剂进行给药以预防疾病或与所述疾病相关的一个或多个症状,且包括:预防疾病或疾病状态在哺乳动物中出现,特别是当这类哺乳动物易患有该疾病状态,但尚未被诊断为已患有该疾病状态时。
针对药物或药理学活性剂而言,术语“治疗有效量”是指无毒的但能达到预期效果的药物或药剂的足够用量。有效量的确定因人而异,取决于受体的年龄和一般情况,也取决于具体的活性物质,个案中合适的有效量可以由本领域技术人员根据常规试验确定。
本申请所述结晶的治疗有效量为从约0.0001到20mg/Kg体重/天,例如从0.001到10mg/Kg体重/天。
本申请所述结晶的剂量频率由患者个体的需求决定,例如,每天1次或2次,或每天更多次。给药可以是间歇性的,例如,其中在若干天的期间内,患者接受结晶的每日剂量,接着在若干天或更多天的期间,患者不接受结晶的每日剂量。
为了描述和公开的目的,以引用的方式将所有的专利、专利申请和其它已确定的出版物在此明确地并入本文。这些出版物仅因为它们的公开早于本申请的申请日而提供。所有关于这些文件的日期的声明或这些文件的内容的表述是基于申请者可得的信息,并且不构成任何关于这些文件的日期或这些文件的内容的正确性的承认。而且,在任何国家,在本中对这些出版物的任何引用并不构成关于该出版物成为本领域的 公知常识的一部分的认可。
本申请所使用的所有溶剂是市售的,无需进一步纯化即可使用。
附图说明
图1式I化合物的A型结晶XRPD图谱;
图2式I化合物的A型结晶DSC图谱;
图3式I化合物的A型结晶TG图谱;
图4式I化合物的B型结晶XRPD图谱;
图5式I化合物的B型结晶DSC图谱;
图6式I化合物的B型结晶TG图谱;
图7式I化合物的C型结晶XRPD图谱;
图8式I化合物的C型结晶DSC图谱;
图9式I化合物的C型结晶TG图谱;
图10式I化合物的D型结晶XRPD图谱;
图11式I化合物的D型结晶DSC图谱;
图12式I化合物的D型结晶TG图谱;
图13式I化合物的E型结晶XRPD图谱;
图14式I化合物的E型结晶DSC图谱;
图15式I化合物的E型结晶TG图谱;
图16式I化合物的F型结晶XRPD图谱;
图17式I化合物的F型结晶DSC图谱;
图18式I化合物的F型结晶TG图谱;
图19式I化合物的G型结晶XRPD图谱;
图20式I化合物的H型结晶XRPD图谱。
具体实施方式
下面通过(但是不限于)以下的实施例、试验对本申请作更加详细的描述。
本申请采用下述缩略词:Boc代表叔丁氧羰基;THF代表四氢呋喃;TBSCl代表叔丁基二甲基氯硅烷。
实施例1:式I化合物的制备
Figure PCTCN2021090885-appb-000012
Figure PCTCN2021090885-appb-000013
1)化合物7-d的制备
将化合物1-c(198.6g)、1-Boc-哌嗪(175.5g)溶于乙腈(800mL)中,搅拌,降温至0℃,缓慢加入三乙酰氧基硼氢化钠(532.6g),室温搅拌5h。反应完全,加水(1L)和乙酸乙酯(300mL)萃取,收集有机相,无水Na 2SO 4干燥,过滤,浓缩,得化合物7-d(269.8g)。
2)化合物7-e的制备
将化合物7-d(269.8g)、异丙醇(800mL)、盐酸(浓度36~38wt%,169mL)混合,加热至65℃,反应3h。冷却析出固体,过滤,干燥,得化合物7-e(151.2g)。
化合物7-e: 1H NMR(500MHz,DMSO-d6),δ:7.82(s,1H),7.68(d,1H),7.36(d,1H),7.10(dd,1H),2.98(s,4H),2.63(d,2H),2.23(m,6H),1.89(m,2H),1.43(s,2H),0.94(s,6H).
13C NMR(125MHz,DMSO-d6),δ:133.1,132.6,131.9,130.9,129.3,128.6,126.5,125.7,124.9,124.6,122.7,60.2,49.4,44.7,35.2,29.4,28.4,27.1,25.2,21.4.ESI-MS:m/z=387.1[M+H] +.
3)化合物7-g的制备
将NaH(21.1g)溶于THF(100mL)中降温至-20℃搅拌10min,将2-[(1H-吡咯并[2,3-b]吡啶-5-基)氧基]-4-溴苯甲酸叔丁酯(化合物1-d,128.3g)溶于200mL THF中,缓慢滴加至反应液中,滴加过程中控制内温在0℃以下,滴加完毕后搅拌30min,向反应液中滴加TBSCl(64.7g)的THF(200mL)溶液,滴加过程中控制内温在-10℃左右,滴毕反应30min,反应完全后加入500mL饱和碳酸氢钠和乙酸乙酯萃取,收集有机相,无水Na 2SO 4干燥,过滤,浓缩,柱层析纯化得化合物7-f(150g)。ESI-MS:m/z=503.1[M+H] +
将化合物7-e(151.2g)、2-[(1-叔丁基二甲基硅基吡咯并[2,3-b]吡啶-5-基)氧基]-4-溴苯甲酸叔丁酯(化合物7-f,197.1g)、三(二亚苄基丙酮)二钯(2.7g)、[(4-(N,N-二甲氨基)苯基]二叔丁基膦(1.6g)、叔丁醇钠(187.4g)、甲苯(800mL)混合,搅拌,氮气保护,加热至100℃反应24h。反应结束,加水(1L)和乙酸乙酯(300mL)萃取,收集有机相,无水Na 2SO 4干燥,过滤,浓缩,得化合物7-g(181.9g)。
化合物7-g: 1H NMR(500MHz,DMSO-d6),δ:7.95(s,1H),7.82(s,1H),7.65(t,2H),7.37(m,4H),6.76(d,1H),6.47(s,1H),3.14(s,2H),2.64(d,1H),2.55(d,1H),2.19(m,5H),1.92(m,2H),1.42(t,2H),1.31(t,2H),1.22(m,9H),0.95(d,6H),0.84(s,10H),0.60(s,6H).
13C NMR(125MHz,DMSO-d6),δ:164.4 156.8,155.1,150.3,149.8,146.2,133.6,133.4,133.2,132.0, 131.9,131.5,129.5,129.0,128.8,126.4,126.1,124.4,122.8,114.4,113.8,110.2,106.8,103.3,80.1,60.6,52.6,47.1,44.7,35.2,29.4,27.9,27.2,26.7,25.4,19.0.ESI-MS:m/z=809.4[M+H] +
4)化合物7-h的制备
将化合物7-g(181.9g)、甲苯(1.8L)、三氟乙酸(107mL)的混合物加热至45℃,反应5h。浓缩反应液,加乙酸乙酯1.5L,饱和NaHCO 3水溶液、饱和氯化钠水溶液洗涤,无水硫酸钠干燥,过滤,浓缩,加1L甲苯和200mL乙酸乙酯,加热溶清后,降温析出固体,过滤,干燥,得化合物7-h(83.4g)。
化合物7-h: 1H NMR(500MHz,DMSO-d6),δ:7.98(s,1H),7.82(s,1H),7.73(d,1H),7.64(d,1H),7.46(s,1H),7.40(s,1H),7.32(d,1H),6.73(d,1H),6.36(d,1H),6.34(s,1H),3.09(s,4H),2.64(d,1H),2.55(d,1H),2.19(m,6H),1.88(m,2H),1.42(m,2H),1.25(m,2H),0.95(m,6H).
13C NMR(125MHz,DMSO-d6),δ:166.3,158.9,155.1,148.9,146.2,145.3,135.0,133.8,133.2,132.0,131.9,131.4,129.5,129.2,127.8,126.4,124.9,124.4,122.7,120.2,116.6,112.0,109.5,105.3,100.2,60.5,55.3,52.7,47.0,44.7,35.2,29.4,27.2,25.4.ESI-MS:m/z=639.2[M+H] +.
5)化合物8-k的制备
将3-硝基-4-氯苯磺酰胺(35.0g)、(S)-2-(氨甲基)-1,4-二氧六环盐酸盐(30.0g)和N,N-二异丙基乙胺(94.0g)溶于乙腈(400mL)中,加热至85℃,反应5h,室温冷却,静置过夜,抽滤,得化合物8-k(46.5g)ESI-MS:m/z=316.1[M-H]-。
6)式I化合物的制备
将化合物7-h(10g)和二氯甲烷(100mL)混合,室温搅拌,加入4-二甲氨基吡啶(2.8g)及1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐(4.4g),搅拌溶解,加入化合物8-k(5.0g)和三乙胺(4.5g),室温反应3h。依次用5wt%盐酸、饱和碳酸氢钠水溶液、饱和氯化钠水溶液洗涤,无水Na 2SO 4干燥,过滤,浓缩,经柱层析分离纯化得式I化合物的无定形(8.9g)。
式I化合物: 1H NMR(500MHz,DMSO-d6),δ:11.66(s,1H),11.37(s,1H),8.59(t,1H),8.57(d,1H),8.04(d,1H),7.89(d,1H),7.84(dd,1H),7.70(d,1H),7.54(d,1H),7.52(m,2H),7.40(m,1H),7.11(d,1H),6.75(dd,1H),6.40(dd,1H),6.29(d,1H),3.79(m,3H),3.65(m,2H),3.51(m,2H),3.42(m,2H),3.03(m,4H),2.67(d,1H),2.54(d,1H),2.17(m,6H),1.88(dd,2H),1.42(t,2H),0.96(s,6H).
13C NMR(125MHz,DMSO-d6),δ:164.0,158.2,154.0,147.9,146.9,145.9,144.3,135.6,134.3,132.9,132.6,131.8,130.2,130.0,128.3,128.2,127.0,125.2,125.0,122.8,120.3,118.3,115.7,114.1,109.7,103.6,100.4,73.4,68.5,66.4,66.2,58.6,45.1,44.4,43.9,34.6,29.3,29.1,27.2,24.8.ESI-MS:m/z=939.4[M+H] +.
实施例2:式I化合物A型结晶的制备
将实施例1制备的式I化合物5g溶于300mL二氯甲烷与甲醇的混合溶液中(二氯甲烷:甲醇=20:1或100:1,V:V),搅拌溶清,减压浓缩得A型结晶。A型结晶的XRPD图谱如图1所示;DSC图谱如图2所示,TG图谱如图3所示。
实施例3:式I化合物B型结晶的制备
称取约200mg的式I化合物A型结晶至4mL玻璃瓶中,加入2mL丙酮,将得到的悬浮液置于室温下磁力搅拌5小时,过滤收集沉淀物,最后于40~60℃真空干燥24小时。得到式I化合物B型结晶。B型结晶的XRPD图谱如图4所示;DSC图谱如图5所示,TG图谱如图6所示。
实施例4:式I化合物C型结晶的制备
称取约4g的式I化合物A型结晶至500mL反应瓶中,分别加入在100mL甲醇和100mL甲基叔丁基醚,将得到的悬浮液置于室温下磁力搅拌12小时,过滤收集沉淀物,于40~60℃真空干燥24小时。得到式I化合物C型结晶。C型结晶的XRPD图谱如图7所示;DSC图谱如图8所示,TG图谱如图9所示。
实施例5:式I化合物D型结晶的制备
1)称取约3g的式I化合物A型结晶至500mL反应瓶中,加入200mL四氢呋喃-水的混合溶剂(四氢呋喃:水=10:1~0.5:1),将得到的悬浮液置于室温下磁力搅拌12小时,过滤收集沉淀物,于40~60℃鼓风 干燥。得到式I化合物D型结晶。
2)称取约3g的式I化合物A型结晶至250mL反应瓶中,加入100mL异丙醇或1,4-二氧六环,将得到的悬浮液置于室温下磁力搅拌12小时,过滤收集沉淀物,于40~60℃鼓风干燥。得到式I化合物D型结晶。
3)称取约0.7g的式I化合物A型结晶至100mL反应瓶中,加入35mL四氢呋喃,室温下磁力搅拌使完全溶解,过滤得到上清液并转移到干净的40mL玻璃瓶中,用封口膜封住装有上清液的玻璃瓶并扎几个小孔,室温下放置使溶剂缓慢挥发,待溶剂挥发完全后收集固体。得到式I化合物D型结晶。
D型结晶的XRPD图谱如图10所示;DSC图谱如图11所示,TG图谱如图12所示。
实施例6:式I化合物E型结晶的制备
称取约20g的式I化合物A型结晶至1L反应瓶中,加入400mL对二甲苯,将得到的悬浮液置于室温下磁力搅拌5小时,过滤收集沉淀物,于80~100℃真空干燥5~10小时得到式I化合物E型结晶。E型结晶的XRPD图谱如图13所示;DSC图谱如图14所示,TG图谱如图15所示。
实施例7:式I化合物F型结晶的制备
称取约4g的式I化合物A型结晶至500mL反应瓶中,加入200mL乙腈或硝基甲烷中,将得到的悬浮液置于室温下磁力搅拌12小时,过滤收集沉淀物,于40~60℃真空干燥。得到式I化合物F型结晶。F型结晶的XRPD图谱如图16所示;DSC图谱如图17所示,TG图谱如图18所示。
实施例8:式I化合物G型结晶的制备
称取约4g的式I化合物A型结晶于500mL反应瓶中,加入375mL对二甲苯,将得到的悬浮液置于室温下磁力搅拌5小时,过滤收集沉淀物。得到式I化合物G型结晶。G型结晶的XRPD图谱如图19所示。
实施例9:式I化合物H型结晶的制备
称取约4g的式I化合物A型结晶于500mL反应瓶中,加入200mL 4-甲基-2-戊酮,将得到的悬浮液置于室温下磁力搅拌12小时,过滤收集沉淀物,于50℃真空干燥2小时。得到式I化合物H型结晶。H型结晶的XRPD图谱如图20所示。
实施例10:结晶稳定性试验
供试品置于敞口的、适宜的洁净容器中,60℃温度下放置10天,于第5天和第10天取样;
供试品置于恒湿密闭容器中,在25℃下于相对湿度92.5%±5%的条件下放置10天,于第5天和第10天取样;
供试品放在装有日光灯的光照箱或其他适宜的光照装置(敞口)内,于照度为4500lx±500lx的条件下放置10天,于第5天和第10天取样;
上述三种情况下,检测供试品在实验条件下放置后,产品的纯度及有关物质的含量。
仪器:Thermo U3000高效液相色谱仪;
检测器:Thermo VWD-3100紫外吸收检测器/DAD-3000二极管阵列检测器;
色谱柱:XSelect CSH C18(4.6×150mm,3.5μm);
进样体积:10μL;检测波长:280nm;流速:1.0mL/min;柱温:30℃;
流动相:A相:0.1%甲酸水溶液,B相:乙腈,梯度洗脱;
样品溶液的配制:取本品适量,精密称定,用甲醇溶解并定量稀释制成每1mL中约含0.5mg的溶液,作为供试品溶液。
结果见下表9。
表9
Figure PCTCN2021090885-appb-000014
实施例11:引湿性实验
引湿性试验方法和条件参照中国药典2015版第四部通则9103药物引湿性试验指导原则:药物的引湿性是指在一定温度及湿度条件下该物质吸收水分能力或程度的特性。供试品为符合药品质量标准的固体原料药,试验结果可作为选择适宜的药品包装和贮存条件的参考。
1).取干燥的具塞玻璃称量瓶(外径为50mm,高为15mm),于试验前一天置于适宜的25℃±1℃恒温干燥器(下部放置氯化铵或者硫酸铵饱和溶液)或人工气候箱(设定温度为25℃±1℃,相对湿度为80%±2%)内,精密称定重量(m1)。
2).取供试品适量,平铺于上述称量瓶中,供试品厚度一般约为1mm,精密称定重量(m2)。
3).将称量瓶敞口,并与瓶盖同置于上述恒温恒湿条件下24小时。
4).盖好称量瓶盖子,精密称定重量(m3)。
实验结果见表10。
Figure PCTCN2021090885-appb-000015
表10
晶型 引湿增重 晶型 引湿增重
A型结晶 1.38wt% E型结晶 0.11wt%
B 0.30wt% F型结晶 0.76wt%
C 0.11wt% H型结晶 0.25wt%
D 0.75wt%    
试验例1 体外蛋白结合抑制活性
1.1 BCL-2/BAK结合抑制活性筛选
用试剂盒(型号:BCL-2/BAK(BH3)BINDING ASSAY KITS,来源于cisbio)中的稀释缓冲液将500nM的Tag1-BCL-2蛋白母液稀释成5nM,同时将20μM的Tag2-BAK蛋白母液稀释成120nM,先每孔加入5μL的Tag1-BCL-2蛋白稀释液,然后用纳升加样仪将DMSO溶解的式I化合物加入到孔中,使化合物终浓度为200nM-0.0488nM,4倍梯度,共7个浓度,同时设空白对照孔(不含酶)与阴性对照孔(含酶,加溶媒DMSO),设2个复孔,最后再每孔加入5μL的Tag2-BAK蛋白稀释液,离心混匀,25℃孵育15min。用试剂盒中的检测缓冲液将100×的anti-Tag1-Eu 3+稀释成1×的使用浓度,同时将100×的anti-Tag2-XL665稀释成1×的使用浓度。将anti-Tag1-Eu 3+和anti-Tag2-XL665按1:1混匀,每孔加入5μL的混合液,25℃反应2h及以上。PE Envision多功能酶标仪进行读板(激发620nm,发射665nm),采用四参数拟合,计算IC 50(表11所示)。
1.2 BCL-XL/BAK结合抑制活性筛选
用试剂盒(型号:BCL-XL/BAK(BH3)BINDING ASSAY KITS,来源于cisbio)中的稀释缓冲液将300nM的Tag1-BCL-XL蛋白母液稀释成2nM,同时将10μM的Tag2-BAK蛋白母液稀释成80nM,先每孔加入5μL的Tag1-BCL-XL蛋白稀释液,然后用纳升加样仪将DMSO溶解的式I化合物加入到孔中,使化合物终浓度为2000nM-0.488nM,4倍梯度,共7个浓度,同时设空白对照孔(不含酶)与阴性对照孔(含酶,加溶媒DMSO),设2个复孔,最后再每孔加入5μL的Tag2-BAK蛋白稀释液,离心混匀,25℃孵育15min。用试剂盒中的检测缓冲液将100×的anti-Tag1-Eu 3+稀释成1×的使用浓度,同时将100×的anti-Tag2-XL665稀释成1×的使用浓度。将anti-Tag1-Eu 3+和anti-Tag2-XL665按1:1混匀,每孔加入5μL的混合液,25℃反应2h及以上。PE Envision多功能酶标仪进行读板(激发620nm,发射665nm),采用四参数拟合,计算IC 50(表11所示)。
表11 式I化合物抑制BCL-2/BAK和BCL-XL/BAK结合活性
Figure PCTCN2021090885-appb-000016
试验例2 化合物对RS4;11细胞的增殖抑制作用
取处于指数生长期状态良好的RS4;11细胞(来源于南京科佰),收集细胞至离心管,低速台式离心机,1500转/min,离心3min,弃上清,用移液器加入5mL完全培养基(RPMI基础培养基+10wt%胎牛血清(FBS))进行细胞重悬。使用细胞计数仪计数,用完全培养基进行稀释,调整细胞密度至2×10 5个/mL,在加入等量的RPMI基础培养基调整血清浓度为5%,细胞密度为1×10 5个/mL种板。使用排枪接种于96孔板上,100μL/孔,置于37℃、含5%CO 2饱和湿度的细胞培养箱中培养。培养24h后,使用纳升加样仪进行化合物加样,每一浓度设置2个复孔,以不加化合物的细胞作为阴性对照,72小时后加CCK-8试剂,10μL/孔,4小时后Envision酶标仪450nm处检测其吸光值,计算抑制率,抑制率(%)=(阴性对照组平均值—实验组平均值)/(阴性对照组平均值—空白组平均值)×100%,以化合物浓度对数为横坐标,抑制率为纵坐标,四参数分析,拟合量效曲线,计算IC 50(见表12)。
表12 化合物对RS4;11细胞的增殖抑制作用
Figure PCTCN2021090885-appb-000017
试验例3 体外肝微位体稳定性评价
300μL最终的温孵体系中,含30μL肝微粒体(蛋白浓度:5mg/mL),30μL NADPH+MgCl 2,3μL受试化合物(乙腈配制),237μL PBS缓冲液(PH7.4)。其中有机溶剂(乙腈)的比例为1%(体积比)。每个种属(小鼠、大鼠、人)做2份,每份0.3mL。每管先配好总体积为270μL的底物及酶的混匀液,和NADPH分别在37℃预温孵5min后,加入30μL NADPH+MgCl 2混合,分别于0、15、30、60min取出50μL用含内标的冰乙腈300μL终止反应。
50μL温孵样品,加入300μL含内标(地西泮20ng/mL)的冰乙腈沉淀,涡旋震荡5min后,离心(13000rpm,20℃)10min。吸取上清液70μL,加入70μL超纯水稀释混匀,1μL进样分析。化合物在人、大鼠和小鼠肝微粒体中消除参数见表13。
表13 化合物体外肝微粒体代谢稳定性(1μM)
Figure PCTCN2021090885-appb-000018
试验例4 体内药代动力学评价
4.1大鼠体内药代动力学评价
SD大鼠,体重180~220g,适应3~5天后,随机分组,每组3只,按5mg/kg剂量灌胃式I化合物溶液。
受试动物(SD大鼠)给药前禁食12h,给药后4h给食物,实验前后和实验过程中均自由饮水。
灌胃给药后,于0min、15min、30min、1h、2h、4h、6h、8h、10h、24h于眼眶取血0.2mL左右,EDTA-K2抗凝后,30min内转移到4℃,4000rpm,10min条件下离心分离血浆。收集全部血浆后立即于-20℃保存待测。
吸取50μL待测血浆样品,加入300μL含内标(地西泮20mg/mL)的乙腈溶液,振荡混匀5min,13000rpm离心10min,取上清75μL,加入75μL超纯水稀释,混匀,吸取2μL用于LC/MS/MS测定,记录色谱图。
通过大鼠体内药物动力学实验评估本申请化合物的口服暴露量。采用DAS3.2.5软件拟合化合物的药代参数。数据如下表14所示。
表14 化合物的药代参数
PK参数 式I化合物IG 5mg/kg
T max(h) 4.00±0.00
C max(ng/mL) 739±226
AUC(0-24h)(ng*h/mL) 5973±2021
AUC(0-∞)(ng*h/mL) 6558±1805
t 1/2(h) 7.62±2.78
MRT(0-t)(h) 7.35±0.45
4.2比格犬体内药代动力学评价
雄性比格犬3只,体重9~12kg,适应一段时间后,按2.5mg/kg剂量分别灌胃式I化合物溶液。
受试动物(雄性比格犬)给药前禁食12h,给药后4h给食物,实验前后和实验过程中均自由饮水。
灌胃给药后,于0.25h(15min)、0.5h(30min)、1h、1.5h、2h、4h、6h、8h、10h、24h、30h、48h、72h于前肢静脉取血约0.5mL置于EDTA-K2抗凝真空采血管中,血浆于30min内转移到4℃,4000rpm,10min条件下离心分离血浆。收集全部血浆后立即于-20℃保存待测。
吸取50μL待测血浆样品,加入300μL含内标(地西泮20ng/mL)的乙腈溶液,振荡混匀5min,13000rpm离心10min,取上清75μL,加入75μL超纯水稀释,混匀,吸取1μL用于LC/MS/MS测定,记录色谱图。
通过比格犬体内药物动力学实验评估本申请化合物的口服暴露量。采用DAS3.2.5软件拟合化合物的药代参数如下表15所示。
表15 化合物的比格犬体内药代参数
Figure PCTCN2021090885-appb-000019
试验例5 受试物在RS4;11人类B细胞白血病皮下移植模型中的药效学评价
NOD/SCID小鼠,雌性,9-10周(肿瘤细胞接种时的小鼠周龄),体重16.3-22.0g。购自安凯毅博生物技术有限公司,生产许可证号:SCXK(京)2017-0006,动物合格证编号:11402400013155。饲养环境:SPF级。小鼠右侧前背部皮下接种1×10 7RS4;11细胞。接种当天定义为第0天。待肿瘤平均体积240mm 3时,根据肿瘤大小随机分组。按下表16进行给药。
表16 人源B细胞白血病RS4;11皮下动物模型中的给药途径、剂量及方案
组别 n 给药组 剂量(mg/kg) 给药方式 给药时间
1 6 Vehicle   p.o. 单次
2 6 式I化合物(溶液) 25 p.o. 单次
注:n:动物只数;给药体积为10μL/g。
实验过程中观察到的临床症状均记录在原始数据中。肿瘤体积计算公式:肿瘤体积(mm 3)=1/2×(a×b 2)(其中a表示长径,b表示短径)。实验中使用StudyDirector TM(版本号3.1.399.19,供应商Studylog System,Inc.,S.San Francisco,CA,USA)软件收集数据,包括肿瘤的长短径的测量和动物体重的称量。原始数据由天平和游标卡尺测量后直接导入软件,数据的任何变动都将被记录在此软件中。相对肿瘤增殖率,T/C%,即在某一时间点,治疗组和对照组相对肿瘤体积或瘤重的百分比值。计算公式如下:
T/C%=T RTV/C RTV×100%(T RTV:治疗组平均RTV;C RTV:溶媒对照组平均RTV;RTV=V t/V 0,V 0为分组时该动物的瘤体积,V t为治疗后该动物的瘤体积)。
相对肿瘤抑制率,TGI(%),计算公式如下:TGI%=(1-T/C)×100%。(T和C分别为治疗组和对照组在某一特定时间点的相对肿瘤体积(RTV)或瘤重(TW))。
所有实验结果以平均瘤体积±SEM(平均标准误差)表示。用独立样本T检验方法比较治疗组相对肿瘤体积与对照组相比有无显著性差异。所有的数据均用SPSS 18.0进行分析。p<0.05为具有显著性差异。结果见表17。
表17 在人源B细胞白血病RS4;11皮下模型中各组药效分析表
Figure PCTCN2021090885-appb-000020
注:1.数据以“平均值±标准误差”表示;
2.T/C%=T RTV/C RTV×100%;TGI%=(1-T/C)×100%。
试验例6 人血小板毒性实验(Caspase3活性测定)
使用肝素钠抗凝管抽取10mL人全血,上下颠倒混匀,90g离心10min,收集上清,继续1950g离心10min。弃上清,用4mL PBS重悬混匀后,1190g离心5min,弃上清,再用4mL PBS重悬混匀,1190g离心5min,弃上清,用PBS重悬血小板并调整密度为2~3×10 8个/mL。按2~3×10 7个/mL的密度接种于96 孔板,100μL/孔,阴性对照孔加50μL对照缓冲液,化合物孔每孔加入50μL对应浓度的化合物,使化合物终浓度为2.5μM或1μM,37℃培养箱中孵育90min。将96孔板中的液体分别转移到1.5mL离心管中。4℃6000g离心5min,弃上清,置于冰上待用。用试剂盒中提供的水将5×的裂解液稀释成1×的裂解液,同时按1:200的比例加蛋白酶抑制剂cocktail,配制成待用裂解混合液。每个离心管中加入40μL裂解混合液,用移液器将底部的血小板重悬,冰上裂解15~20min,4℃14000g离心10min,样品分装待用。用试剂盒中提供的水将10×的检测液稀释成1×的检测液,同时按1:600的比例加底物Ac-DEVD-AMC,配置成反应混合液。空白对照孔加入5μL检测缓冲液,40μL反应混合液。样品阴性对照孔,加入5μL对照血小板裂解液,40μL反应混合液。化合物组,加入5μL血小板裂解液,40μL反应混合液。其中40μL的反应混合液最后加入,轻轻混合,PE Envision多功能酶标仪进行读板(激发360nm,发射460nm),每10min检测一次,检测6次。根据释放的AMC荧光强度的大小,可以测定Caspase-3的活性,即每个孔所对应拟合直线的斜率代表Caspase活性大小(所有数据做了归一化处理,基准为ABT-199)。结果见表18。
表18 化合物对人血小板Caspase3活性作用
Figure PCTCN2021090885-appb-000021
注:数据做了归一化处理。

Claims (17)

  1. 一种式I化合物的结晶,
    Figure PCTCN2021090885-appb-100001
  2. 如权利要求1所述的式I化合物的结晶,其特征在于,X-射线粉末衍射图谱中2θ在5.01、6.61、8.12或20.13±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在5.01、6.61、8.12、10.21、14.89、16.63或20.13±0.2°处有峰;
    或者,X-射线粉末衍射图谱在选自5.01、6.61、8.12、10.21、12.88、14.89、16.63、20.13或21.01±0.2°的2θ处具有至少7个或至少8个衍射峰;
    或者,X-射线粉末衍射图谱中2θ在5.01、6.61、8.12、10.21、12.88、14.89、16.63、20.13或21.01±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在5.01、6.61、8.12、10.21、12.88、13.74、14.89、16.63、18.58、20.13、21.01或26.20±0.2°处有峰;
    或者,X-射线粉末衍射图谱如图1所示。
  3. 如权利要求1所述的式I化合物的结晶,其特征在于,X-射线粉末衍射图谱中2θ在5.31、12.64、19.08或24.21±0.2°处有峰;
    或者,X-射线粉末衍射图谱在选自5.31、10.65、12.64、14.23、19.08、19.91、22.71或24.21±0.2°的2θ处具有至少6或至少7个衍射峰;
    或者,X-射线粉末衍射图谱中2θ在5.31、10.65、12.64、14.23、19.08、19.91、22.71或24.21±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在5.31、9.80、10.65、12.12、12.64、14.23、16.04、18.13、19.08、19.91、22.71、24.21或25.93±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在5.31、9.80、10.65、12.12、12.64、13.57、13.82、14.23、15.17、16.04、17.64、18.13、19.08、19.91、20.34、22.71、22.99、23.45、24.21、25.65或25.93±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在5.31、9.50、9.80、10.65、11.41、12.12、12.64、13.57、13.82、14.23、15.17、16.04、16.64、17.10、17.64、18.13、18.33、18.73、19.08、19.60、19.91、20.34、21.22、21.93、22.71、22.99、23.45、24.21、25.65或25.93±0.2°处有峰;
    或者,X-射线粉末衍射图谱如图4所示。
  4. 如权利要求3所述的式I化合物的结晶,其特征在于,DSC图谱在179.42±5℃处具有吸热峰的起始点;或者,DSC图谱如图5所示。
  5. 如权利要求1所述的式I化合物的结晶,其特征在于,X-射线粉末衍射图谱中2θ在5.52、7.56、9.22、11.04或17.43±0.2°处有峰;
    或者,X-射线粉末衍射图谱在选自5.52、7.56、8.29、9.22、11.04、15.81、17.43、18.51或22.59±0.2°的2θ处具有至少7或至少8个衍射峰;
    或者,X-射线粉末衍射图谱中2θ在5.52、7.56、8.29、9.22、11.04、15.81、17.43、18.51或22.59±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在5.52、7.56、8.29、9.22、11.04、15.17、15.81、17.43、18.51或20.40±0.2° 处有峰;
    或者,X-射线粉末衍射图谱中2θ在5.52、7.56、8.29、9.22、11.04、15.17、15.81、17.00、17.43、18.51、19.70、20.01、20.40、20.75或22.59±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在5.52、7.56、8.29、9.22、10.66、11.04、12.94、14.69、15.17、15.81、16.63、17.00、17.43、18.51、19.70、20.01、20.40、20.75、22.59、25.88、26.12±0.2°处有峰;
    或者,X-射线粉末衍射图谱如图7所示。
  6. 如权利要求5所述的式I化合物的结晶,其特征在于,DSC图谱在205.65±5℃处具有吸热峰的起始点;或者,DSC图谱如图8所示。
  7. 如权利要求1所述的式I化合物的结晶,其特征在于,X-射线粉末衍射图谱中2θ在4.78、12.83、16.24或22.33±0.2°处有峰;
    或者,X-射线粉末衍射图谱在选自4.78、10.52、12.83、16.24、18.44、19.41、22.33或23.20±0.2°的2θ处具有至少6或至少7个衍射峰;
    或者,X-射线粉末衍射图谱中2θ在4.78、10.52、12.83、16.24、18.44、19.41、22.33或23.20±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在4.78、9.63、10.52、12.83、13.48、15.79、16.24、17.89、18.44、19.41、19.61、22.33或23.20±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在4.78、7.46、9.63、10.52、11.17、12.83、13.48、14.42、15.79、16.24、17.89、18.44、19.41、19.61、20.44、22.33、23.20、26.48或27.05±0.2°处有峰;
    或者,X-射线粉末衍射图谱如图10所示。
  8. 如权利要求7所述的式I化合物的结晶,其特征在于,DSC图谱在176.47±5℃处具有吸热峰的起始点;或者,DSC图谱如图11所示。
  9. 如权利要求1所述的式I化合物的结晶,其特征在于,X-射线粉末衍射图谱中2θ在4.22、10.72、15.17或15.65±0.2°处有峰;
    或者,X-射线粉末衍射图谱在选自4.22、10.72、14.62、15.17、15.65、17.54、19.55、19.80或21.50±0.2°的2θ处具有至少7或至少8个衍射峰;
    或者,X-射线粉末衍射图谱中2θ在4.22、10.72、14.62、15.17、15.65、17.54、19.55、19.80或21.50±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在4.22、10.72、13.82、14.62、15.17、15.65、16.92、17.54、19.55、19.80、21.50、22.76、23.35或26.06±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在4.22、7.99、8.75、9.91、10.72、11.66、12.75、13.82、14.62、15.17、15.65、16.24、16.92、17.54、19.04、19.55、19.80、20.18、21.50、22.76、23.35、26.06、26.91、29.82或30.56±0.2°处有峰;
    或者,X-射线粉末衍射图谱如图13所示。
  10. 如权利要求9所述的式I化合物的结晶,其特征在于,DSC图谱在145.48±5℃处具有吸热峰的起始点;或者,DSC图谱如图14所示。
  11. 如权利要求1所述的式I化合物的结晶,其特征在于,X-射线粉末衍射图谱中2θ在4.54、9.08或19.24±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在4.54、9.08、14.90、18.25、19.24、22.86或23.50±0.2°处有峰;
    或者,X-射线粉末衍射图谱在选自4.54、9.08、13.66、14.90、17.46、18.25、19.24、22.86、23.50、24.75或27.51±0.2°的2θ处具有至少8或至少9个或至少10个衍射峰;
    或者,X-射线粉末衍射图谱中2θ在4.54、9.08、13.66、14.90、17.46、18.25、19.24、22.86、23.50、24.75或27.51±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在4.54、8.20、9.08、13.66、13.97、14.51、14.90、15.51、16.63、17.46、 18.25、19.24、19.82、21.11、22.37、22.86、23.50、24.75或27.51±0.2°处有峰;
    或者,X-射线粉末衍射图谱如图16所示。
  12. 如权利要求11所述的式I化合物的结晶,其特征在于,DSC图谱在171.37±5℃处具有吸热峰的起始点;或者,DSC图谱如图17所示。
  13. 如权利要求1所述的式I化合物的结晶,其特征在于,X-射线粉末衍射图谱中2θ在3.84、10.39、13.39或20.63±0.2°处有峰;
    或者,X-射线粉末衍射图谱在选自3.84、10.39、11.22、13.39、15.55、16.78、20.01或20.63±0.2°的2θ处具有至少6或至少7个衍射峰
    或者,X-射线粉末衍射图谱中2θ在3.84、10.39、11.22、13.39、15.55、16.78、20.01或20.63±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在3.84、7.72、9.56、10.39、11.22、12.47、13.39、14.01、15.55、16.78、19.02、20.01或20.63±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在3.84、6.69、7.72、9.56、10.39、11.22、12.47、13.39、14.01、15.00、15.55、16.78、18.59、19.02、19.47、20.01、20.63、22.50、23.44、23.69、24.11、25.48或26.47±0.2°处有峰;
    或者,X-射线粉末衍射图谱如图19所示。
  14. 如权利要求1所述的式I化合物的结晶,其特征在于,X-射线粉末衍射图谱中2θ在4.65、12.23、14.09或22.04±0.2°处有峰;
    或者,X-射线粉末衍射图谱在选自4.65、9.36、12.23、13.33、14.09、17.27、19.37、22.04或22.95±0.2°的2θ处具有至少7或至少8个衍射峰;
    或者,X-射线粉末衍射图谱中2θ在4.65、9.36、12.23、13.33、14.09、17.27、19.37、22.04或22.95±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在4.65、9.36、10.41、12.23、13.33、14.09、17.27、18.88、19.37、20.58、22.04、22.49、22.95或23.69±0.2°处有峰;
    或者,X-射线粉末衍射图谱中2θ在4.65、7.20、9.36、10.41、11.12、12.23、13.33、14.09、15.44、16.41、16.68、17.27、18.31、18.88、19.37、20.58、22.04、22.49、22.95、23.69或25.12±0.2°处有峰;
    或者,X-射线粉末衍射图谱如图20所示。
  15. 结晶组合物,所述结晶组合物包含权利要求1-14中任一项所述的式I化合物的结晶,其中权利要求1-14中任一项所述的式I化合物的结晶占所述结晶组合物重量的50%以上、优选80%以上、更优选90%以上、最优选95%以上。
  16. 药物组合物,其包含如权利要求1-14中任一项所述的式I化合物的结晶、或如权利要求15所述的结晶组合物。
  17. 如权利要求1-14中任一项所述的式I化合物的结晶、如权利要求15所述的结晶组合物或者如权利要求16所述的药物组合物在制备预防或者治疗与抗凋亡蛋白BCL-2相关的疾病的药物中的用途;
    任选地,所述与抗凋亡蛋白BCL-2相关的疾病为癌症;任选地,所述癌症为急性淋巴细胞白血病。
PCT/CN2021/090885 2020-04-29 2021-04-29 三氟甲基和氯双取代的磺酰胺类选择性bcl-2抑制剂的晶体 WO2021219065A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN202180028439.3A CN115397825A (zh) 2020-04-29 2021-04-29 三氟甲基和氯双取代的磺酰胺类选择性bcl-2抑制剂的晶体
EP21796153.1A EP4129998A4 (en) 2020-04-29 2021-04-29 CRYSTAL OF A TRIFLUOROMETHYL/CHLORODISUBSTITUTED SULFONAMIDE SELECTIVE BCL-2 INHIBITOR
US17/922,223 US20230174528A1 (en) 2020-04-29 2021-04-29 Crystal of trifluoromethyl/chloro disubstituted sulfonamide selective bcl-2 inhibitor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010355842 2020-04-29
CN202010355842.4 2020-04-29

Publications (1)

Publication Number Publication Date
WO2021219065A1 true WO2021219065A1 (zh) 2021-11-04

Family

ID=78374073

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/090885 WO2021219065A1 (zh) 2020-04-29 2021-04-29 三氟甲基和氯双取代的磺酰胺类选择性bcl-2抑制剂的晶体

Country Status (5)

Country Link
US (1) US20230174528A1 (zh)
EP (1) EP4129998A4 (zh)
CN (1) CN115397825A (zh)
TW (1) TW202144350A (zh)
WO (1) WO2021219065A1 (zh)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010065865A2 (en) 2008-12-05 2010-06-10 Abbott Laboratories Bcl-2-selective apoptosis-inducing agents for the treatment of cancer and immune diseases
WO2010138588A2 (en) 2009-05-26 2010-12-02 Abbott Laboratories Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
CN102307622A (zh) * 2008-12-04 2012-01-04 雅培制药有限公司 用于治疗癌症和免疫和自身免疫疾病的诱发细胞凋亡的药剂
WO2012071374A1 (en) 2010-11-23 2012-05-31 Abbott Laboratories Methods of treatment using selective bcl-2 inhibitors

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112888687B (zh) * 2018-10-29 2023-01-24 正大天晴药业集团股份有限公司 三氟甲基取代的磺酰胺类选择性bcl-2抑制剂

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102307622A (zh) * 2008-12-04 2012-01-04 雅培制药有限公司 用于治疗癌症和免疫和自身免疫疾病的诱发细胞凋亡的药剂
WO2010065865A2 (en) 2008-12-05 2010-06-10 Abbott Laboratories Bcl-2-selective apoptosis-inducing agents for the treatment of cancer and immune diseases
CN102307872A (zh) * 2008-12-05 2012-01-04 雅培制药有限公司 作为用于治疗癌症和免疫疾病的bcl-2-选择性诱发细胞凋亡药剂的磺酰胺衍生物
WO2010138588A2 (en) 2009-05-26 2010-12-02 Abbott Laboratories Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
CN104876927A (zh) * 2009-05-26 2015-09-02 艾伯维巴哈马有限公司 用于治疗癌症和免疫和自身免疫疾病的细胞程序死亡诱导药剂
WO2012071374A1 (en) 2010-11-23 2012-05-31 Abbott Laboratories Methods of treatment using selective bcl-2 inhibitors

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
"Chinese Pharmacopoeia", vol. IV, 2015, article "Guidelines for the Hygroscopicity Test of Drugs"
See also references of EP4129998A4

Also Published As

Publication number Publication date
CN115397825A (zh) 2022-11-25
EP4129998A4 (en) 2024-05-01
TW202144350A (zh) 2021-12-01
US20230174528A1 (en) 2023-06-08
EP4129998A1 (en) 2023-02-08

Similar Documents

Publication Publication Date Title
TWI791511B (zh) 細胞凋亡誘導劑
RU2506264C2 (ru) Пиримидин-замещенные пуриновые соединения в качестве ингибиторов киназы (или киназ)
CN109053595B (zh) 表皮生长因子受体激酶抑制剂的盐
US10150770B2 (en) Crystal form of bisulfate of JAK inhibitor and preparation method therefor
TWI820223B (zh) 嗎啉基喹唑啉類化合物的晶型、其製備方法及應用
US20180155335A1 (en) Salt and crystal forms of plk-4 inhibitor
BR112021008323A2 (pt) sulfonamida substituída com trifluorometil como inibidor seletivo de bcl-2
WO2018045993A1 (zh) 一种取代的2-氢-吡唑衍生物的晶型、盐型及其制备方法
WO2022170864A1 (zh) Belumosudil甲磺酸盐的晶型及其制备方法和用途
EP4036078A1 (en) Crystalline form of capsid protein assembly inhibitor containing n hetero five-membered ring, and application thereof
CN106957315A (zh) N-取代苯磺酰基-氮杂吲哚氧基苯甲酰胺类化合物及其制备药物的用途
US11230559B2 (en) Solid forms of [(1 S)-1 -[(2S,4R,5R)-5-(5-amino-2-oxo-thiazolo[4,5-D]pyrimidin-3-yl)-4-hydroxy-tetrahydrofuran-2-Yl]proptl] acetate
US9085607B2 (en) ACH-0142684 sodium salt polymorph, composition including the same, and method of manufacture thereof
US20220332684A1 (en) Crystal form of five-membered n heterocyclic compound, and application thereof
CN114728973A (zh) 一种核蛋白抑制剂的晶型及其应用
WO2021219065A1 (zh) 三氟甲基和氯双取代的磺酰胺类选择性bcl-2抑制剂的晶体
US20220009925A1 (en) Crystal of diarylthiohydantoin compound
WO2020119785A1 (zh) 一种Syk抑制剂的盐及其结晶型
WO2019205812A1 (zh) Acalabrutinib的新晶型及其制备方法和用途
US12012420B2 (en) Solid forms of [(1S)-1-[(2S,4R,5R)-5-(5-amino-2-oxo-thiazolo[4,5-d]pyrimidin-3-yl)-4-hydroxy-tetrahydrofuran-2-yl]propyl] acetate
US20220119415A1 (en) Solid forms of [(1s)-1-[(2s,4r,5r)-5-(5-amino-2-oxo-thiazolo[4,5-d]pyrimidin-3-yl)-4-hydroxy-tetrahydrofuran-2-yl]propyl] acetate
WO2018149309A1 (zh) 4-苯基噻唑衍生物的晶型及其制备方法
KR20220004102A (ko) Plk4 억제제 (1r,2s)-(e)-2-(3-(4-((시스-2,6-디메틸모르폴리노)메틸)스티릴)-1h-이미다졸-6-일)-5&#39;-메톡시스피로[시클로프로판-1,3&#39;-인돌린]-2&#39;-온 푸마레이트의 결정 형태 s4
WO2023006057A1 (zh) 氨基吡唑并嘧啶化合物的晶体
RU2810214C2 (ru) Кристалл диарилтиогидантоинового соединения

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: 21796153

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021796153

Country of ref document: EP

Effective date: 20221025

NENP Non-entry into the national phase

Ref country code: DE