WO2023040876A1 - 氮杂芳环类化合物及其药学上可接受的盐的多晶型物、药物组合物和应用 - Google Patents

氮杂芳环类化合物及其药学上可接受的盐的多晶型物、药物组合物和应用 Download PDF

Info

Publication number
WO2023040876A1
WO2023040876A1 PCT/CN2022/118654 CN2022118654W WO2023040876A1 WO 2023040876 A1 WO2023040876 A1 WO 2023040876A1 CN 2022118654 W CN2022118654 W CN 2022118654W WO 2023040876 A1 WO2023040876 A1 WO 2023040876A1
Authority
WO
WIPO (PCT)
Prior art keywords
crystal form
ray powder
compound
powder diffraction
free base
Prior art date
Application number
PCT/CN2022/118654
Other languages
English (en)
French (fr)
Inventor
赵双妮
王吉标
陶涛
Original Assignee
上海海雁医药科技有限公司
扬子江药业集团有限公司
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 上海海雁医药科技有限公司, 扬子江药业集团有限公司 filed Critical 上海海雁医药科技有限公司
Priority to CN202280060100.6A priority Critical patent/CN117915919A/zh
Publication of WO2023040876A1 publication Critical patent/WO2023040876A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders

Definitions

  • This application relates to the field of medical technology, in particular to an azaaromatic compound 3-(4-amino-6-(1-((1-ethyl-1H-pyrazol-3-yl)methyl)-1H -1,2,3-triazol-4-yl)-5-fluoropyrimidin-2-yl)-2-methylbenzonitrile polymorphs or pharmaceutically acceptable salts thereof, and polymorphs comprising the polymorph
  • the pharmaceutical composition of the crystal form, the preparation method and application of the polymorph and the pharmaceutical composition are examples of the crystal form, the preparation method and application of the polymorph and the pharmaceutical composition.
  • Adenosine is an endogenous nucleoside throughout human cells, composed of adenine and ribose, which is widely distributed inside and outside cells. Adenosine participates in a variety of physiological and biochemical functions in the body. For example, adenosine can directly enter the myocardium and be phosphorylated to generate adenosine triphosphate (ATP), which participates in the energy metabolism of the myocardium. In the central nervous system (Central Nervous System, CNS), adenosine controls the release of neurotransmitters and the response of postsynaptic neurons, and plays an important role in regulating movement, protecting neurons, affecting sleep and wakefulness, etc. . In pathological conditions, the extracellular adenosine concentration increases significantly under tumor or hypoxic conditions. Adenosine can play an important role in tumor immunosuppression by promoting tumor angiogenesis, proliferation, development and tumor migration.
  • ATP adenosine triphosphate
  • Adenosine receptor belongs to G protein-coupled receptor (Guanosine-binding Protein Coupled Receptor, GPCR) family, and its endogenous ligand is adenosine.
  • the currently known adenosine receptors are composed of four subtype receptors, A 1 , A 2A , A 2B and A 3 .
  • the combination of adenosine with A1 or A3 receptors can inhibit the production of cyclic adenosine monophosphate (cAMP); while the combination with A2A or A2B receptors can activate adenosine activating enzyme, and then up-regulate the level of cAMP, and play a further role.
  • cAMP cyclic adenosine monophosphate
  • a 1 and A 3 receptors are mainly expressed in the central nervous system, while A 2A and A 2B adenosine receptors are expressed in both the central nervous system and the peripheral system.
  • A2A and A2B are widely expressed in immune cells and have strong immunosuppressive functions.
  • the increase of extracellular adenosine concentration is one of the important mechanisms of immune escape of tumor cells, and its concentration level is jointly determined by the level of ATP and the expression levels of CD39 and CD73.
  • the increase in extracellular adenosine concentration is related to the release of large amounts of ATP by cell death or hypoxia in the tumor microenvironment, and its concentration can reach 10-20 times that of normal tissues.
  • Adenosine binds to adenosine receptors in the tumor microenvironment, which can inhibit anti-tumor responses, such as inhibiting the function of CD8+ T cells, enhancing the function of immunosuppressive regulatory T cells, and inhibiting the function of antigen-presenting cells through dendritic cells wait. Recent studies have shown that binding to A2A receptors can also inhibit the tumor-killing effect of natural killer cells.
  • A2A adenosine receptor antagonists can improve the activity and killing ability of dendritic antigen-presenting cells, T cells and natural killer cells, and inhibit regulatory T cells (T-regs), bone marrow-derived suppressive Cells (MDSCs) and tumor-associated macrophages (TAMs) eliminate tumor immune tolerance and promote the occurrence of tumor immune responses, which in turn lead to suppressed tumor growth and prolong the survival of mice.
  • T-regs regulatory T cells
  • MDSCs bone marrow-derived suppressive Cells
  • TAMs tumor-associated macrophages
  • A2B receptors have also been reported to promote tumor migration in mouse melanoma and triple-negative breast cancer models, so A2B receptor antagonists are also effective targets for cancer therapy.
  • blocking the activation of adenosine signaling pathway to reduce or relieve immunosuppression and enhance the anti-tumor function of immune cells—especially T cells is considered to be one of the effective methods for cancer treatment.
  • the A2A / A2B dual receptor antagonist is used to simultaneously block the activation of these two receptors. Mechanistically speaking, it is regulating different immune cell populations and comprehensively blocking the immune response brought about by adenosine in the microenvironment. Inhibitory effect has far-reaching clinical application value for tumor therapy.
  • the present application provides a polymorph of an azaaromatic compound represented by formula (X) or a pharmaceutically acceptable salt thereof, a pharmaceutical composition containing it, a preparation method or application thereof,
  • the pharmaceutically acceptable salt is selected from: hydrochloride, sulfate, citrate, fumarate or succinate.
  • the polymorphs of the compound of Formula X or the polymorphs of the pharmaceutically acceptable salt of the compound of Formula X are each independently in an anhydrous form, a hydrate form, or a solvate form.
  • Each of the X-ray powder diffraction patterns involved in the present application can be obtained independently using Cu-K ⁇ radiation.
  • the type I crystal form of the compound represented by formula (X) is provided, that is, the free base crystal form I, and its X-ray powder diffraction pattern has a characteristic diffraction peak at the following 2 ⁇ (°) angle: 7.57 ⁇ 0.2 , 13.41 ⁇ 0.2, 14.64 ⁇ 0.2, 19.81 ⁇ 0.2, and 24.43 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form I further includes one or two characteristic diffraction peaks selected from the following 2 ⁇ (°) angles: 24.87 ⁇ 0.2 and 27.46 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form I further includes 2 or more characteristic diffraction peaks selected from the following 2 ⁇ (°) angles: 8.56 ⁇ 0.2, 15.40 ⁇ 0.2, 18.55 ⁇ 0.2, 21.25 ⁇ 0.2, 23.05 ⁇ 0.2, 23.59 ⁇ 0.2, and 25.60 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form I has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 7.57 ⁇ 0.2, 8.56 ⁇ 0.2, 10.60 ⁇ 0.2, 11.80 ⁇ 0.2, 13.41 ⁇ 0.2 0.2, 14.16 ⁇ 0.2, 14.64 ⁇ 0.2, 15.40 ⁇ 0.2, 16.63 ⁇ 0.2, 17.23 ⁇ 0.2, 18.55 ⁇ 0.2, 19.81 ⁇ 0.2, 21.25 ⁇ 0.2, 22.18 ⁇ 0.2, 22.46 ⁇ 0.2, 23.05 ⁇ 0.2, 23.59 ⁇ 0.2, 24.43 ⁇ 0.2, 24.87 ⁇ 0.2, 25.60 ⁇ 0.2, 26.03 ⁇ 0.2, and 27.46 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form I has diffraction peaks at the 2 ⁇ (°) values shown in Table 1, and the relative intensities of each diffraction peak are shown in Table 1. Among them, the definition of each relative intensity symbol is shown in Table 12.
  • the X-ray powder diffraction pattern of the free base crystalline form I is substantially as shown in FIG. 1 .
  • the X-ray powder diffraction pattern of the free base crystalline form I is shown in FIG. 1 .
  • the differential scanning calorimetry curve of the free base crystalline form I has an endothermic peak at 179.10 ⁇ 3°C.
  • the differential scanning calorimetry curve of the free base crystalline form I has an endothermic peak at 179.10 ⁇ 2°C.
  • the differential scanning calorimetry curve of the free base crystalline form I has an endothermic peak at 179.10 ⁇ 0.5°C.
  • the differential scanning calorimetry curve of the free base Form I is substantially as shown in FIG. 2 .
  • the differential scanning calorimetry curve of the free base crystalline form I is shown in FIG. 2 .
  • the compound of formula (X) is mixed with the I-3 solvent, heated to dissolve at a temperature of 45°C to 85°C, cooled to 0°C to room temperature, separated to obtain the I-3 solid, and
  • said 1-3 solvent is methanol or ethanol.
  • the preparation method of free base crystal form I comprises the following steps:
  • the compound of formula (X) is mixed with the I-3 solvent, heated to dissolve at a temperature of 45°C to 55°C, cooled to 0°C to 4°C, separated to obtain the I-3 solid, and
  • said 1-3 solvent is methanol or ethanol.
  • the preparation method of free base crystal form I comprises the following steps:
  • said I-3A solvent is ethanol.
  • the preparation method of free base crystal form I comprises the following steps:
  • the compound of formula (X) is mixed with the I-3B solvent, heated to dissolve at a temperature of 60° C. to 70° C., cooled to room temperature, and separated to obtain the I-3 solid, and
  • said I-3B solvent is methanol.
  • the third aspect of the present application provides the type IV crystal form of the compound of formula (X), that is, the free base crystal form IV, whose X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 11.88 ⁇ 0.2, 14.17 ⁇ 0.2, 16.96 ⁇ 0.2, 22.63 ⁇ 0.2, 23.56 ⁇ 0.2, and 25.66 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form IV further includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 15.76 ⁇ 0.2 and 26.08 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form IV further includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 15.76 ⁇ 0.2, 26.08 ⁇ 0.2 and 37.48 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form IV further includes 2 or more characteristic diffraction peaks selected from the following 2 ⁇ (°) angles: 14.89 ⁇ 0.2, 21.73 ⁇ 0.2, 24.43 ⁇ 0.2, 26.65 ⁇ 0.2, 27.39 ⁇ 0.2, 28.42 ⁇ 0.2, and 30.24 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form IV further includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 8.71 ⁇ 0.2, 19.93 ⁇ 0.2 and 31.78 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form IV has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 8.71 ⁇ 0.2, 9.57 ⁇ 0.2, 11.88 ⁇ 0.2, 14.17 ⁇ 0.2, 14.89 ⁇ 0.2 0.2, 15.76 ⁇ 0.2, 16.96 ⁇ 0.2, 19.93 ⁇ 0.2, 21.73 ⁇ 0.2, 22.63 ⁇ 0.2, 23.56 ⁇ 0.2, 24.43 ⁇ 0.2, 25.66 ⁇ 0.2, 26.08 ⁇ 0.2, 26.65 ⁇ 0.2, 27.39 ⁇ 0.2, 28.42 ⁇ 0.2, 30.24 ⁇ 0.2, 31.78 ⁇ 0.2, and 33.15 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form IV has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 8.71 ⁇ 0.2, 9.57 ⁇ 0.2, 11.88 ⁇ 0.2, 14.17 ⁇ 0.2, 14.89 ⁇ 0.2 0.2, 15.76 ⁇ 0.2, 16.96 ⁇ 0.2, 19.93 ⁇ 0.2, 21.73 ⁇ 0.2, 22.63 ⁇ 0.2, 23.56 ⁇ 0.2, 24.43 ⁇ 0.2, 25.66 ⁇ 0.2, 26.08 ⁇ 0.2, 26.65 ⁇ 0.2, 27.39 ⁇ 0.2, 28.42 ⁇ 0.2, 30.24 ⁇ 0.2, 31.78 ⁇ 0.2, 33.15 ⁇ 0.2, and 37.48 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form IV has diffraction peaks at the 2 ⁇ (°) values shown in Table 2, and the relative intensity of each diffraction peak is shown in Table 2. Among them, the definition of each relative intensity symbol is shown in Table 12.
  • the X-ray powder diffraction pattern of the free base crystalline Form IV is substantially as shown in FIG. 4 .
  • the X-ray powder diffraction pattern of the free base crystalline form IV is shown in FIG. 4 .
  • the differential scanning calorimetry curve of the free base crystalline form IV has an endothermic peak at 170.43 ⁇ 3°C.
  • the differential scanning calorimetry curve of the free base crystalline form IV has an endothermic peak at 170.43 ⁇ 2°C.
  • the differential scanning calorimetry curve of the free base crystalline form IV has an endothermic peak at 170.43 ⁇ 0.5°C.
  • the differential scanning calorimetry curve of the free base crystalline Form IV is substantially as shown in FIG. 5 .
  • the differential scanning calorimetry curve of the free base crystalline Form IV is shown in FIG. 5 .
  • a method for preparing free base crystal form IV comprising the following steps:
  • the V-type crystal form of the compound of formula (X) is provided, that is, the free base crystal form V, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 11.88 ⁇ 0.2, 16.36 ⁇ 0.2, 16.99 ⁇ 0.2, 22.99 ⁇ 0.2, 23.47 ⁇ 0.2, and 26.41 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form V has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 11.88 ⁇ 0.2, 16.36 ⁇ 0.2, 16.99 ⁇ 0.2, 22.99 ⁇ 0.2, 23.47 ⁇ 0.2, 26.41 ⁇ 0.2 and 37.48 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form V further includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 7.90 ⁇ 0.2, 13.71 ⁇ 0.2, 14.14 ⁇ 0.2, 15.31 ⁇ 0.2, 17.80 ⁇ 0.2, 18.76 ⁇ 0.2, 21.34 ⁇ 0.2, 24.82 ⁇ 0.2, 25.51 ⁇ 0.2, 27.88 ⁇ 0.2, and 30.46 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form V further includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 7.90 ⁇ 0.2, 13.71 ⁇ 0.2, 14.14 ⁇ 0.2, 15.31 ⁇ 0.2, 17.80 ⁇ 0.2, 18.76 ⁇ 0.2, 21.34 ⁇ 0.2, 24.82 ⁇ 0.2, 25.51 ⁇ 0.2, 27.88 ⁇ 0.2, 30.46 ⁇ 0.2, and 43.60 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form V further includes 2 or more characteristic diffraction peaks selected from the following 2 ⁇ (°) angles: 15.77 ⁇ 0.2, 20.03 ⁇ 0.2, 28.40 ⁇ 0.2, 30.85 ⁇ 0.2, 32.71 ⁇ 0.2, 33.15 ⁇ 0.2, and 34.62 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form V has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 7.90 ⁇ 0.2, 8.62 ⁇ 0.2, 9.73 ⁇ 0.2, 11.88 ⁇ 0.2, 13.71 ⁇ 0.2 0.2, 14.14 ⁇ 0.2, 15.31 ⁇ 0.2, 15.77 ⁇ 0.2, 16.36 ⁇ 0.2, 16.99 ⁇ 0.2, 17.80 ⁇ 0.2, 18.76 ⁇ 0.2, 20.03 ⁇ 0.2, 21.34 ⁇ 0.2, 22.99 ⁇ 0.2, 23.47 ⁇ 0.2, 24.82 ⁇ 0.2, 25.51 ⁇ 0.2, 26.41 ⁇ 0.2, 27.88 ⁇ 0.2, 28.40 ⁇ 0.2, 30.46 ⁇ 0.2, 30.85 ⁇ 0.2, 32.71 ⁇ 0.2, 33.15 ⁇ 0.2, and 34.62 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form V has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 7.90 ⁇ 0.2, 8.62 ⁇ 0.2, 9.73 ⁇ 0.2, 11.88 ⁇ 0.2, 13.71 ⁇ 0.2 0.2, 14.14 ⁇ 0.2, 15.31 ⁇ 0.2, 15.77 ⁇ 0.2, 16.36 ⁇ 0.2, 16.99 ⁇ 0.2, 17.80 ⁇ 0.2, 18.76 ⁇ 0.2, 20.03 ⁇ 0.2, 21.34 ⁇ 0.2, 22.99 ⁇ 0.2, 23.47 ⁇ 0.2, 24.82 ⁇ 0.2, 25.51 ⁇ 0.2, 26.41 ⁇ 0.2, 27.88 ⁇ 0.2, 28.40 ⁇ 0.2, 30.46 ⁇ 0.2, 30.85 ⁇ 0.2, 32.71 ⁇ 0.2, 33.15 ⁇ 0.2, 34.62 ⁇ 0.2, 37.48 ⁇ 0.2, and 43.60 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form V has diffraction peaks at the 2 ⁇ (°) values shown in Table 3, and the relative intensity of each diffraction peak is shown in Table 3. Among them, the definition of each relative intensity symbol is shown in Table 12.
  • the X-ray powder diffraction pattern of the free base Form V is substantially as shown in FIG. 6 .
  • the X-ray powder diffraction pattern of the free base form V is shown in FIG. 6 .
  • the differential scanning calorimetry curve of the free base crystal form V has an endothermic peak at 179.02 ⁇ 3°C.
  • the differential scanning calorimetry curve of the free base crystalline form V has an endothermic peak at 179.02 ⁇ 2°C.
  • the differential scanning calorimetry curve of the free base form V has an endothermic peak at 179.02 ⁇ 0.5°C.
  • the differential scanning calorimetry curve of the free base Form V is substantially as shown in FIG. 7 .
  • the differential scanning calorimetry curve of the free base Form V is shown in FIG. 7 .
  • a method for preparing free base crystal form V comprising the following steps:
  • the seventh aspect of the present application provides the VIII crystal form of the compound of formula (X), that is, the free base crystal form VIII, whose X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 6.40 ⁇ 0.2, 13.12 ⁇ 0.2 and 15.91 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form VIII further includes one or two characteristic diffraction peaks selected from the following 2 ⁇ (°) angles: 22.69 ⁇ 0.2 and 26.95 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form VIII further includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 11.62 ⁇ 0.2, 12.36 ⁇ 0.2, 16.62 ⁇ 0.2, 18.34 ⁇ 0.2, 18.88 ⁇ 0.2, 24.94 ⁇ 0.2, and 29.65 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form VIII further includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 11.62 ⁇ 0.2, 12.36 ⁇ 0.2, 16.62 ⁇ 0.2, 18.34 ⁇ 0.2, 18.88 ⁇ 0.2, 24.94 ⁇ 0.2, 29.65 ⁇ 0.2, and 37.48 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form VIII has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 6.40 ⁇ 0.2, 11.62 ⁇ 0.2, 12.36 ⁇ 0.2, 13.12 ⁇ 0.2, 15.91 ⁇ 0.2 0.2, 16.62 ⁇ 0.2, 18.34 ⁇ 0.2, 18.88 ⁇ 0.2, 22.69 ⁇ 0.2, 24.94 ⁇ 0.2, 26.95 ⁇ 0.2, and 29.65 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form VIII has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 6.40 ⁇ 0.2, 11.62 ⁇ 0.2, 12.36 ⁇ 0.2, 13.12 ⁇ 0.2, 15.91 ⁇ 0.2 0.2, 16.62 ⁇ 0.2, 18.34 ⁇ 0.2, 18.88 ⁇ 0.2, 22.69 ⁇ 0.2, 24.94 ⁇ 0.2, 26.95 ⁇ 0.2, 29.65 ⁇ 0.2, and 37.48 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the free base crystal form VIII has diffraction peaks at the 2 ⁇ (°) values shown in Table 4, and the relative intensities of each diffraction peak are shown in Table 4. Among them, the definition of each relative intensity symbol is shown in Table 12.
  • the X-ray powder diffraction pattern of the free base Form VIII is substantially as shown in FIG. 8 .
  • the X-ray powder diffraction pattern of the free base form VIII is shown in FIG. 8 .
  • the differential scanning calorimetry curve of the free base crystalline form VIII has an endothermic peak at 162.93 ⁇ 3°C.
  • the differential scanning calorimetry curve of the free base crystalline form VIII has an endothermic peak at 162.93 ⁇ 2°C.
  • the differential scanning calorimetry curve of the free base crystalline form VIII has an endothermic peak at 162.93 ⁇ 0.5°C.
  • the differential scanning calorimetry curve of the free base Form VIII is substantially as shown in FIG. 9 .
  • the differential scanning calorimetry curve of the free base crystalline Form VIII is shown in FIG. 9 .
  • the eighth aspect of the present application provides a method for preparing free base crystal form VIII, comprising the following steps:
  • the free base crystal form I, free base crystal form IV or free base crystal form V described in the above aspects with the Z 3 -1 solvent, suspend and shake at room temperature for 70 to 80 hours, and separate to obtain the second Z 3 -1 solid, drying the Z 3 -1 solid to obtain the free base crystal form VIII; wherein the Z 3 -1 solvent is acetone or ethanol aqueous solution with a volume fraction of 50%.
  • the hydrochloride salt crystal form I of the compound of formula (X) is provided, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 11.95 ⁇ 0.2, 24.37 ⁇ 0.2, 25.21 ⁇ 0.2 and 26.35 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the hydrochloride salt form I also includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 8.83 ⁇ 0.2, 14.13 ⁇ 0.2, 15.33 ⁇ 0.2, 15.68 ⁇ 0.2, 18.86 ⁇ 0.2, 23.64 ⁇ 0.2, 27.67 ⁇ 0.2, and 28.57 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the hydrochloride salt form I has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 8.83 ⁇ 0.2, 11.95 ⁇ 0.2, 14.13 ⁇ 0.2, 15.33 ⁇ 0.2, 15.68 ⁇ 0.2, 17.25 ⁇ 0.2, 18.86 ⁇ 0.2, 23.64 ⁇ 0.2, 24.37 ⁇ 0.2, 25.21 ⁇ 0.2, 26.35 ⁇ 0.2, 27.67 ⁇ 0.2, and 28.57 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the hydrochloride salt form I has diffraction peaks at the 2 ⁇ (°) values shown in Table 5, and the relative intensity of each diffraction peak is shown in Table 5. Among them, the definition of each relative intensity symbol is shown in Table 12.
  • the X-ray powder diffraction pattern of the hydrochloride salt form I is substantially as shown in FIG. 10 .
  • the X-ray powder diffraction pattern of the hydrochloride salt form I is shown in FIG. 10 .
  • Its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 8.22 ⁇ 0.2, 11.29 ⁇ 0.2, 14.14 ⁇ 0.2, 16.96 ⁇ 0.2, 18.13 ⁇ 0.2, 22.27 ⁇ 0.2, 23.86 ⁇ 0.2 and 27.79 ⁇ 0.2 .
  • the X-ray powder diffraction pattern of the sulfate crystal form I further includes 2 or more characteristic diffraction peaks selected from the following 2 ⁇ (°) angles: 6.54 ⁇ 0.2 and 11.64 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the sulfate crystal form I further includes 2 or more characteristic diffraction peaks selected from the following 2 ⁇ (°) angles: 6.54 ⁇ 0.2, 11.64 ⁇ 0.2 and 37.48 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the sulfate crystal form I has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 6.54 ⁇ 0.2, 8.22 ⁇ 0.2, 11.29 ⁇ 0.2, 11.64 ⁇ 0.2, 14.14 ⁇ 0.2 0.2, 16.96 ⁇ 0.2, 18.13 ⁇ 0.2, 22.27 ⁇ 0.2, 23.86 ⁇ 0.2, and 27.79 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the sulfate crystal form I has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 6.54 ⁇ 0.2, 8.22 ⁇ 0.2, 11.29 ⁇ 0.2, 11.64 ⁇ 0.2, 14.14 ⁇ 0.2 0.2, 16.96 ⁇ 0.2, 18.13 ⁇ 0.2, 22.27 ⁇ 0.2, 23.86 ⁇ 0.2, 27.79 ⁇ 0.2, and 37.48 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the sulfate salt form I has diffraction peaks at the 2 ⁇ (°) values shown in Table 6, and the relative intensity of each diffraction peak is shown in Table 6. Among them, the definition of each relative intensity symbol is shown in Table 12.
  • the X-ray powder diffraction pattern of the sulfate salt form I is substantially as shown in FIG. 11 .
  • the X-ray powder diffraction pattern of the sulfate crystalline form I is shown in FIG. 11 .
  • the citrate crystal form I of the compound of formula (X) is provided, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 12.55 ⁇ 0.2, 14.50 ⁇ 0.2, 21.55 ⁇ 0.2 and 22.75 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the citrate crystal form I has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 12.55 ⁇ 0.2, 14.50 ⁇ 0.2, 21.55 ⁇ 0.2, 22.75 ⁇ 0.2, 37.51 ⁇ 0.2 and 43.63 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the citrate crystal form I further includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 5.38 ⁇ 0.2, 9.78 ⁇ 0.2 and 13.63 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the citrate crystal form I has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 5.38 ⁇ 0.2, 9.78 ⁇ 0.2, 12.55 ⁇ 0.2, 13.63 ⁇ 0.2, 14.50 ⁇ 0.2, 21.55 ⁇ 0.2, and 22.75 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the citrate crystal form I has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 5.38 ⁇ 0.2, 9.78 ⁇ 0.2, 12.55 ⁇ 0.2, 13.63 ⁇ 0.2, 14.50 ⁇ 0.2, 21.55 ⁇ 0.2, 22.75 ⁇ 0.2, 37.51 ⁇ 0.2, and 43.63 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the citrate crystal form I has diffraction peaks at the 2 ⁇ (°) values shown in Table 7, and the relative intensity of each diffraction peak is shown in Table 7. Among them, the definition of each relative intensity symbol is shown in Table 12.
  • the X-ray powder diffraction pattern of the citrate salt form I is substantially as shown in FIG. 12 .
  • the X-ray powder diffraction pattern of the citrate salt form I is shown in FIG. 12 .
  • the fumarate salt crystal form I of the compound of formula (X) is provided, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 7.12 ⁇ 0.2, 11.71 ⁇ 0.2 and 16.90 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the fumarate salt form I also includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 23.26 ⁇ 0.2, 23.65 ⁇ 0.2, 24.64 ⁇ 0.2 and 25.54 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the fumarate salt form I also includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 8.02 ⁇ 0.2, 15.69 ⁇ 0.2, 18.22 ⁇ 0.2 and 26.44 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the fumarate salt form I also includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 8.02 ⁇ 0.2, 15.69 ⁇ 0.2, 18.22 ⁇ 0.2 , 26.44 ⁇ 0.2 and 37.87 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the fumarate salt form I also includes 2 or more characteristic diffraction peaks selected from the following 2 ⁇ (°) angles: 10.48 ⁇ 0.2, 13.80 ⁇ 0.2 , 22.00 ⁇ 0.2 and 22.42 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the fumarate salt form I has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 7.12 ⁇ 0.2, 8.02 ⁇ 0.2, 10.48 ⁇ 0.2, 11.71 ⁇ 0.2 .
  • the X-ray powder diffraction pattern of the fumarate salt form I has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 7.12 ⁇ 0.2, 8.02 ⁇ 0.2, 10.48 ⁇ 0.2, 11.71 ⁇ 0.2 , 13.80 ⁇ 0.2, 15.69 ⁇ 0.2, 16.90 ⁇ 0.2, 18.22 ⁇ 0.2, 22.00 ⁇ 0.2, 22.42 ⁇ 0.2, 23.26 ⁇ 0.2, 23.65 ⁇ 0.2, 24.64 ⁇ 0.2, 25.54 ⁇ 0.2, 26.44 ⁇ 0.2, and 37.87 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the fumarate salt form I has diffraction peaks at the 2 ⁇ (°) values shown in Table 8, and the relative intensity of each diffraction peak is shown in Table 8. Among them, the definition of each relative intensity symbol is shown in Table 12.
  • the X-ray powder diffraction pattern of the fumarate salt form I is substantially as shown in FIG. 13 .
  • the X-ray powder diffraction pattern of the fumarate salt form I is shown in FIG. 13 .
  • the succinate salt crystal form I of the compound of formula (X) is provided, and its X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 13.78 ⁇ 0.2, 14.14 ⁇ 0.2 and 22.12 ⁇ 0.2.
  • the X-ray powder diffraction pattern of succinate salt form I has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 13.78 ⁇ 0.2, 14.14 ⁇ 0.2, 22.12 ⁇ 0.2 and 37.84 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the succinate salt form I further includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 4.87 ⁇ 0.2, 9.31 ⁇ 0.2, 18.16 ⁇ 0.2, 20.71 ⁇ 0.2 and 26.83 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the succinate salt form I has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 4.87 ⁇ 0.2, 9.31 ⁇ 0.2, 13.78 ⁇ 0.2, 14.14 ⁇ 0.2, 15.69 ⁇ 0.2, 18.16 ⁇ 0.2, 20.71 ⁇ 0.2, 22.12 ⁇ 0.2, and 26.83 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the succinate salt form I has characteristic diffraction peaks at the following 2 ⁇ (°) angles:
  • the X-ray powder diffraction pattern of the succinate salt form I has diffraction peaks at the 2 ⁇ (°) values shown in Table 9, and the relative intensity of each diffraction peak is shown in Table 9. Among them, the definition of each relative intensity symbol is shown in Table 12.
  • the X-ray powder diffraction pattern of the succinic salt form I is substantially as shown in FIG. 14 .
  • the X-ray powder diffraction pattern of the succinic salt crystal form I is shown in FIG. 14 .
  • the fourteenth aspect of the present application provides the succinate crystal form II of the compound of formula (X), whose X-ray powder diffraction pattern has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 12.49 ⁇ 0.2 and 14.50 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the succinate crystal form II also includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 11.86 ⁇ 0.2, 15.22 ⁇ 0.2, 16.90 ⁇ 0.2, 21.64 ⁇ 0.2, 22.87 ⁇ 0.2, and 25.75 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the succinate crystal form II also includes at least one characteristic diffraction peak selected from the following 2 ⁇ (°) angles: 11.86 ⁇ 0.2, 15.22 ⁇ 0.2, 16.90 ⁇ 0.2, 21.64 ⁇ 0.2, 22.87 ⁇ 0.2, 25.75 ⁇ 0.2, and 37.84 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the succinate crystal form II has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 5.35 ⁇ 0.2, 11.86 ⁇ 0.2, 12.49 ⁇ 0.2, 14.50 ⁇ 0.2, 15.22 ⁇ 0.2, 16.90 ⁇ 0.2, 21.64 ⁇ 0.2, 22.87 ⁇ 0.2, and 25.75 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the succinate crystal form II has characteristic diffraction peaks at the following 2 ⁇ (°) angles: 5.35 ⁇ 0.2, 11.86 ⁇ 0.2, 12.49 ⁇ 0.2, 14.50 ⁇ 0.2, 15.22 ⁇ 0.2, 16.90 ⁇ 0.2, 21.64 ⁇ 0.2, 22.87 ⁇ 0.2, 25.75 ⁇ 0.2, and 37.84 ⁇ 0.2.
  • the X-ray powder diffraction pattern of the succinate salt form II has diffraction peaks at the 2 ⁇ (°) values shown in Table 10, and the relative intensity of each diffraction peak is shown in Table 10. Among them, the definition of each relative intensity symbol is shown in Table 12.
  • the X-ray powder diffraction pattern of the succinic salt form II is substantially as shown in FIG. 15 .
  • the X-ray powder diffraction pattern of the succinate salt form II is shown in FIG. 15 .
  • the acid is hydrochloric acid, sulfuric acid, citric acid, fumaric acid, or succinic acid.
  • the solvent is ethanol, ethyl acetate, or acetone.
  • the anti-solvent is n-heptane.
  • composition comprising:
  • the seventeenth aspect of the present application provides free base crystal form I, free base crystal form IV, free base crystal form V, free base crystal form VIII, or hydrochloride crystal form of the compound of formula (X) as described above I, sulfate salt crystal form I, citrate crystal form I, fumarate salt crystal form I, succinate salt crystal form I, succinate crystal form II in the preparation of A2A receptor or A2B receptor inhibitors Applications.
  • polymorphs of the above-mentioned compound of formula (X) or a pharmaceutically acceptable salt thereof are provided, such as free base crystal form I, free base crystal form IV, free base crystal form V, Free base form VIII, or its hydrochloride form I, sulfate form I, citrate form I, fumarate form I, succinate form I, succinate form II, Or the use of the pharmaceutical composition described in the sixteenth aspect of the present application in the preparation of drugs for preventing or treating diseases mediated by adenosine A 2A receptors and/or adenosine A 2B receptors.
  • the present application provides a method for preventing or treating diseases mediated by adenosine A 2A receptors and/or adenosine A 2B receptors, comprising administering to patients in need thereof a therapeutically effective amount of A polymorphic form of the compound of formula (X) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as described in the above embodiments.
  • the application provides the polymorphic form of the compound of formula (X) or its pharmaceutically acceptable salt as described in the above embodiment, or the pharmaceutical composition as described in the above embodiment for the prevention or treatment of adenosine A 2A receptor and/or adenosine A 2B receptor-mediated diseases.
  • the disease is cancer or an immune-related disease.
  • the cancer is selected from the group consisting of prostate cancer, colon cancer, rectal cancer, pancreatic cancer, cervical cancer, stomach cancer, endometrial cancer, brain cancer, liver cancer, bladder cancer, ovarian cancer, testicular cancer, head cancer , neck cancer, melanoma, basal cancer, mesothelial lining cancer, white blood cell cancer, esophageal cancer, breast cancer, muscle cancer, connective tissue cancer, small cell lung cancer, non-small cell lung cancer, adrenal gland cancer, thyroid cancer, kidney cancer cancer and bone cancer; or malignant glioma, mesothelioma, renal cell carcinoma, gastric cancer, sarcoma, choriocarcinoma, basal cell carcinoma of the skin, and testicular seminoma.
  • the immune-related disease is selected from rheumatoid arthritis, renal failure, lupus, asthma, psoriasis, colitis, pancreatitis, allergies, fibrosis, anemic fibromyalgia, Alzheimer's Haimer's disease, congestive heart failure, stroke, aortic stenosis, arteriosclerosis, osteoporosis, Parkinson's disease, infection, Crohn's disease, ulcerative colitis, allergic contact dermatitis, and others Eczema, systemic sclerosis, and multiple sclerosis.
  • the compound of formula (X) of the present application the polymorphic form of the compound of formula X, the polymorphic form of the pharmaceutically acceptable salt of the compound of formula X, or the polymorphic form comprising the compound of formula (X) and the compound of formula (X)
  • the pharmaceutical composition of the polymorphic form of the pharmaceutically acceptable salt of the compound of formula X has good pharmacological activity of inhibiting A2A receptor and/or A2B receptor.
  • the polymorphic form of the compound of formula (X) and the polymorphic form of the pharmaceutically acceptable salt of the compound of formula X have good stability, so they have the potential to be developed into medicines.
  • Fig. 1 is the XPRD spectrum of the free base crystal form I of the compound of formula X prepared in Example 2 (using Cu-K ⁇ radiation, the abscissa is angle 2 ⁇ (°), and the ordinate is SQR (counts)), wherein SQR represents intensity square root of
  • Fig. 2 is the DSC spectrum of the free base crystal form I of the compound of formula X prepared in Example 2 (abscissa is temperature (°C), ordinate is heat flow rate (W/g));
  • Fig. 3 is the TGA spectrum of the free base crystal form I of the compound of formula X prepared in Example 2 (the abscissa is temperature (°C), and the ordinate is weight change (%));
  • Figure 4 is the XPRD spectrum of the free base crystal form IV of the compound of formula X prepared in Example 3 (using Cu-K ⁇ radiation, the abscissa is the angle 2 ⁇ (°), and the ordinate is SQR (counts)), wherein SQR represents the intensity square root of
  • Fig. 5 is the DSC spectrum of the free base crystal form IV of the compound of formula X prepared in Example 3 (the abscissa is temperature (°C), and the ordinate is heat flow rate (W/g));
  • Fig. 6 is the XPRD spectrum of the free base crystal form V of the compound of formula X prepared in Example 4 (using Cu-K ⁇ radiation, the abscissa is the angle 2 ⁇ (°), and the ordinate is the intensity (counts));
  • Figure 7 is the DSC spectrum of the free base crystal form V of the compound of formula X prepared in Example 4 (the abscissa is temperature (°C), and the ordinate is heat flow rate (W/g));
  • Fig. 8 is the XPRD spectrum of the free base crystal form VIII of the compound of formula X prepared in Example 5 (using Cu-K ⁇ radiation, the abscissa is the angle 2 ⁇ (°), and the ordinate is the intensity (counts));
  • Figure 9 is the DSC spectrum of the free base crystal form VIII of the compound of formula X prepared in Example 5 (the abscissa is temperature (°C), and the ordinate is heat flow rate (W/g));
  • Figure 10 is the XPRD spectrum of the hydrochloride salt form I of the compound of formula X prepared in Example 6 (using Cu-K ⁇ radiation, the abscissa is the angle 2 ⁇ (°), and the ordinate is the intensity (counts));
  • Figure 11 is the XPRD spectrum of the sulfate crystal form I of the compound of formula X prepared in Example 7 (using Cu-K ⁇ radiation, the abscissa is the angle 2 ⁇ (°), and the ordinate is the intensity (counts));
  • Figure 12 is the XPRD spectrum of the citrate crystal form I of the compound of formula X prepared in Example 8 (using Cu-K ⁇ radiation, the abscissa is the angle 2 ⁇ (°), and the ordinate is the intensity (counts));
  • Figure 13 is the XPRD spectrum of the fumarate salt form I of the compound of formula X prepared in Example 9 (using Cu-K ⁇ radiation, the abscissa is the angle 2 ⁇ (°), and the ordinate is the intensity (counts));
  • Figure 14 is the XPRD spectrum of the succinate salt form I of the compound of formula X prepared in Example 10 (using Cu-K ⁇ radiation, the abscissa is the angle 2 ⁇ (°), and the ordinate is the intensity (counts));
  • Fig. 15 is the XPRD spectrum of the succinate crystal form II of the compound of formula X prepared in Example 11 (using Cu-K ⁇ radiation, the abscissa is the angle 2 ⁇ (°), and the ordinate is the intensity (counts)).
  • first aspect”, “second aspect”, “third aspect”, “fourth aspect”, “fifth aspect” and so on are used for descriptive purposes only, and cannot be understood as indicating or implying relative importance or Quantity, nor should it be understood as implying the importance or quantity of the indicated technical features.
  • first”, “second”, “third”, “fourth”, “fifth” and so on are only for the purpose of non-exhaustive enumeration and description, and it should be understood that they do not constitute a closed limitation on the quantity.
  • the above numerical interval is considered continuous, and includes the minimum and maximum values of the range, and every value between such minimum and maximum values.
  • a range refers to an integer, every integer between the minimum and maximum of the range is included.
  • the ranges may be combined. In other words, unless otherwise indicated, all ranges disclosed herein are to be understood to encompass any and all subranges subsumed therein.
  • the percentage content involved in this application refers to mass percentage for solid-liquid mixing and solid-solid phase mixing, and refers to volume percentage for liquid-liquid phase mixing.
  • the percentage concentration involved in this application refers to the final concentration.
  • the final concentration refers to the proportion of the added component in the system after the component is added.
  • the temperature parameters in this application are allowed to be treated at a constant temperature, and also allowed to be treated within a certain temperature range.
  • the isothermal treatment allows the temperature to fluctuate within the precision of the instrument control.
  • the compound of formula (X) is 3-(4-amino-6-(1-((1-ethyl-1H-pyrazol-3-yl)methyl)-1H-1,2,3 -triazol-4-yl)-5-fluoropyrimidin-2-yl)-2-methylbenzonitrile, which has high inhibitory activity on adenosine A 2A receptors and/or adenosine A 2B receptors.
  • the polymorphic form of the compound of formula (X) includes the polymorphic form of the free base of the compound of formula (X) and the polymorphic form of the pharmaceutically acceptable salt of the compound of formula (X).
  • the pharmaceutically acceptable salt is selected from hydrochloride, sulfate, citrate, fumarate or succinate.
  • Polymorphs of the compound of formula (X) and its pharmaceutically acceptable salts include but are not limited to free base crystal form I, free base crystal form IV, free base crystal form V, free base crystal form of the compound of formula (X) VIII, Hydrochloride Form I, Sulfate Form I, Citrate Form I, Fumarate Form I, Succinate Form I, and Succinate Form II.
  • therapeutically effective amount refers to the amount of the compound of the present application that will cause the individual's biological or medical response, such as reducing or inhibiting enzyme or protein activity or improving symptoms, relieving symptoms, slowing down or delaying disease progression or preventing diseases, etc. quantity.
  • pharmaceutically acceptable carrier should be non-toxic and inert in principle.
  • the form of the “pharmaceutically acceptable carrier” is not particularly limited, including but not limited to solid, semi-solid, liquid and the like.
  • the pharmaceutically acceptable carrier should be compatible with the patient, preferably a mammal, more particularly a human.
  • One of the roles of the pharmaceutically acceptable carrier is to be suitable for delivering the active agent to the target site of interest without terminating the activity of the agent.
  • pharmaceutically acceptable carrier includes buffers, sterile water for injection, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorbing agents compatible with pharmaceutical administration Retardants and the like.
  • Each carrier is “pharmaceutically acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • patient refers to an animal, preferably a mammal, more preferably a human.
  • mammal refers to warm-blooded vertebrate mammals including, for example, cats, dogs, rabbits, bears, foxes, wolves, monkeys, deer, mice, pigs and humans.
  • treating refers to alleviating, delaying progression, attenuating, preventing, or maintaining an existing disease or condition (eg, cancer). Treatment also includes curing, preventing its development, or alleviating to some extent one or more symptoms of a disease or disorder.
  • Solids exist either in amorphous or crystalline form. In the case of crystalline forms, the molecules are positioned within three-dimensional lattice sites. When a compound crystallizes from a solution or slurry, it can crystallize in a different spatial lattice arrangement (a property known as "polymorphism"), forming crystals with different crystalline forms, which are referred to as Known as "polymorphs". Different polymorphs of a given substance may differ from each other in one or more physical properties such as solubility and dissolution rate, true specific gravity, crystalline form, packing mode, flowability and/or solid state stability.
  • Production-scale crystallization can be accomplished by manipulating the solution such that the solubility limit of the compound is exceeded. This can be accomplished by various methods, for example, dissolving the compound at a relatively high temperature and then cooling the solution below the saturation limit; or reducing the volume of the liquid by boiling, evaporation at atmospheric pressure, vacuum drying, or by some other method; The solubility of a compound can be reduced by adding an anti-solvent or a solvent in which the compound has low solubility, or a mixture of such solvents. Another alternative is to adjust the pH to reduce solubility. For a detailed description of crystallization see Crystallization, Third Edition, J W Mullens, Butterworth-Heineman Ltd., 1993, ISBN 0750611294.
  • XRPD can detect information such as crystal form changes, crystallinity, and crystal structure state, and is a common method for identifying crystal forms.
  • the peak position of the XRPD pattern mainly depends on the structure of the crystal form, and the measurement of 2 ⁇ of the XRPD pattern may be slightly different between different instruments, so the value of 2 ⁇ cannot be regarded as absolute. According to the conditions of the instrument used in the test of this application, the diffraction peaks allow a certain error (such as ⁇ 0.2°). It can be understood that for different test instruments and test conditions, the error range is not absolute.
  • the crystal form of the compound of formula X in the present application has a specific crystal form, and has specific characteristic peaks in the XRPD pattern.
  • the X-ray powder diffraction pattern has a characteristic diffraction peak at a specific 2 ⁇ (°) angle", which means that the peak value of the peak is within the indicated numerical range, the indicated numerical point, and the vicinity of the indicated numerical range or near the indicated numerical point.
  • the X-ray powder diffraction pattern has a peak at a diffraction angle of 7.57 ⁇ 0.2
  • the peak of the peak can be located within the range of 7.57 ⁇ 0.2 or near this range, as long as it does not affect the identification of the crystal form as a whole.
  • ⁇ 0.2 here only means the error of the peak in the diffraction angle position, and has nothing to do with the peak shape and peak width of the peak.
  • the "substantially" in "the X-ray powder diffraction pattern is basically characterized by a specific figure" should also be understood similarly, as long as it can be recognized as a whole that a certain X-ray powder diffraction pattern is consistent with the X-ray powder diffraction pattern described in this application. Figures constitute substantially the same, it should be considered to fall within the scope of protection of the present application.
  • differential scanning calorimetry curve in the present application and the position of the endothermic peak shown therein should also be understood similarly, allowing slight discrepancies with the specific numerical values or specific numerical ranges or specific spectrograms disclosed in the present application. Inconsistent, but as long as part or all of the endothermic peak positions in the differential scanning calorimetry curve, or the entire curve is substantially consistent with the present application, it should be deemed to fall within the protection scope of the present application.
  • DSC scanning analysis Also known as “differential calorimetry scanning analysis", it is a technique to measure the relationship between the energy difference and temperature between the measured substance and the reference substance during the heating process.
  • the peak position, shape and number of peaks on the DSC spectrum are related to the properties of the substance, so it can be used to identify the substance qualitatively. This method is commonly used in the field to detect various parameters such as phase transition temperature, glass transition temperature, and heat of reaction of substances.
  • the peak position of the DSC spectrum may vary slightly between different instruments, so the value of the peak position of the DSC endothermic peak cannot be regarded as absolute.
  • the value of the experimental error or difference may be less than or equal to 5°C, or less than or equal to 4°C, or less than or equal to 3°C, or less than or equal to 2°C, or less than or equal to 1°C.
  • TGA is a technique for measuring the quality of substances with temperature under program control. It is suitable for checking the loss of solvent in crystals or the process of sublimation and decomposition of samples. It can be speculated that crystals contain crystal water or crystallization solvents.
  • the mass change shown by the TGA curve depends on many factors such as sample preparation and instrumentation; the mass change detected by TGA varies slightly between different instruments. According to the condition of the instrument used in the test of this application, the error of mass change is not absolute, and a certain error (such as ⁇ 0.1%) is allowed.
  • optimization of crystallization may include seeding the crystallization medium with crystals of the desired form. Additionally, many crystallization methods use combinations of the above strategies. One example is to dissolve the compound of interest in a solvent at elevated temperature, followed by the addition of an appropriate volume of antisolvent in a controlled manner to bring the system just below saturation levels. At this point, seeds of the desired form can be added (and the integrity of the seeds maintained) and the system cooled to complete crystallization.
  • room temperature generally refers to 4-30°C, preferably 20 ⁇ 5°C.
  • the polymorphic form of the compound of formula (X) in the present application has a specific crystal form, and has specific characteristic peaks in the X-ray powder diffraction pattern (XPRD).
  • XPRD X-ray powder diffraction pattern
  • the position of each peak is determined by 2 ⁇ (°). It is understood that different instruments and/or conditions may result in slightly different data, with variations in the position and relative intensity of peaks. Those skilled in the art should understand that when determining the crystal form based on the XRPD pattern, the peaks at low diffraction angles and their intensity, peak shape integrity and other factors are relatively more meaningful. In this application, after further research, the applicant found that when using the above-mentioned instrument for testing, strong background peaks of a blank metal disk appeared near the two 2 ⁇ (°) values of 37 and 44 at high diffraction angles.
  • the peak near 37° in Figure 4 is presumed to be the peak produced by a blank metal disk (such as the peak at 37.48° in Table 2), and the peak near 37° and 43° in Figure 6 is speculated to be a blank metal disk
  • the peaks produced (as the peaks at 37.48° and 43.60° in Table 3)
  • the peaks near 37° in Figure 8 are speculated to be the peaks produced by blank metal disks (as the peaks at 37.48° in Table 4)
  • Figure 11 The peak near 37 ° in the middle is presumed to be the peak produced by the blank metal disc (as the peak at 37.48 ° in Table 6)
  • the peak near 37 ° and 43 ° in Figure 12 is speculated to be the peak produced by the blank metal disc (
  • the peak near 37° in Figure 13 is speculated to be the peak produced by the blank metal disk (as the peak at 37.87° in Table 8), near 37° in Figure 14
  • each crystal form takes the diffraction peak with the highest peak height as the base peak, defines its relative intensity as 100%, and regards it as I 0 , and uses the ratio of the peak height of the other peaks to the base peak peak height as its relative intensity.
  • Intensity I/I 0 the division definition of the relative intensity of each peak is shown in Table 12 below.
  • the DSC spectrum was collected on a DISCOVERY DSC25 differential scanning calorimeter, and the test parameters are shown in Table 13 below.
  • the structure and purity of the compounds are determined by nuclear magnetic resonance ( 1 HNMR) and/or liquid chromatography-mass spectrometry (LC-MS).
  • 1 HNMR Bruker AVANCE-400 nuclear magnetic analyzer, the internal standard is tetramethylsilane (TMS).
  • LC-MS Agilent 1200 HPLC System/6140 MS liquid mass spectrometry (manufacturer: Agilent), column WatersX-Bridge, 150 ⁇ 4.6 mm, 3.5 ⁇ m.
  • the starting materials known in the application can be adopted or synthesized according to methods known in the art, or can be obtained from ABCR GmbH & Co. KG, Acros Organics, Aldrich Chemical Company, Shaoyuan Chemical Technology (Accela ChemBio Inc) and Darui Chemicals Wait for the company to buy.
  • DCM dichloromethane
  • DMF dimethylformamide
  • DMSO dimethylsulfoxide
  • THF tetrahydrofuran
  • EA ethyl acetate
  • PE petroleum ether
  • Pd(dppf)Cl2 [ 1 ,1'-bis(diphenylphosphine)ferrocene]palladium dichloride
  • Pd(PPh 3 ) 2 Cl 2 bis(triphenylphosphine)palladium(II) chloride
  • DPPA diphenylphosphoryl azide Esters
  • DBU 1,8-diazabicycloundec-7-ene.
  • the compound 1-ethylpyrazole-3-carboxylic acid ethyl ester (11.5g, 68.37mmol) was dissolved in THF (150mL), the temperature was lowered to -10°C under the protection of argon, and LiAlH 4 (3.89g, 102.56mmol) was added in batches ), and then stirred at about 0°C for 1 hour.
  • 5.0 g of sodium sulfate decahydrate was added in batches to the reaction solution at 0° C., stirred at 0° C. for 1 hour, then slowly raised to room temperature and stirred for 1 hour.
  • the obtained solid compound of formula X was sent to XRPD for detection, and its powder X-ray diffraction pattern had no characteristic peaks, and it was in an amorphous form.
  • Test Example 1 Inhibitory Activity of Formula X Compounds on A2A Receptors and A2B Receptors
  • CHO-K1/ADORA 2A /G ⁇ 15 (GenScript, M00246) and CHO-K1/ADORA 2B /G ⁇ 15 (GenScript, M00329) cells were cultured in Ham's F-12 (Gibco, 31765092) medium.
  • the culture medium conditions are 10% FBS, 200 ⁇ g/mL Zeocin and 100 ⁇ g/mL Hygromycin B or 10% FBS, 400 ⁇ g/mL G418 and 100 ⁇ g/mL Hygromycin B.
  • the screening steps are as follows:
  • mice healthy adult male ICR mice (weight 25g-40g, 12 mice, the mice in the intravenous injection group were free to drink water and food, and the intragastric administration group was fasted overnight, and free to drink water and food after 4 hours of administration), provided by Beijing Weiwei Provided by Tonglihua Laboratory Animal Technology Co., Ltd.;
  • ICR mice select animals that meet the experimental requirements before administration, and weigh the marks. ICR mice were administered via tail vein (2mg/kg, 5% DMSO, pH4.5 20% Captisol) and intragastrically (10mg/kg, 5% DMSO, pH4.5 20% Captisol).
  • Blood sample collection about 100 ⁇ L of blood was collected through the jugular vein at 0.083 hours, 0.25 hours, 0.5 hours, 1 hour, 2 hours, 4 hours, 6 hours, 7.5 hours and 24 hours after administration for both intravenous and intragastric administration.
  • the blood was transferred to a 1.5mLEP tube pre-added with EDTA2K, centrifuged for 4min (8000rpm, 4°C), and the plasma was separated. The whole process was completed within 15min after blood collection. All plasma samples need to be stored in a -20°C freezer until sample analysis.
  • Table 15 shows the pharmacokinetic properties parameters of the compound of formula X in the present application in mice in the form of intravenous administration.
  • Table 16 shows the parameters of pharmacokinetic properties of compound X of the present application in mice in the form of intragastric administration.
  • Test Example 3 In vivo drug efficacy experiment of the compound of formula X of the present application
  • This test example investigates the mouse melanoma cell line B16F10-OVA-hPD-L1 subcutaneously transplanted tumor mice, after administering the compound of formula X of this application through the oral administration route, test its effect on melanoma B16F10-OVA-hPD-L1 In vivo drug efficacy in L1 tumor-bearing mice.
  • mice female
  • mouse melanoma B16F10-OVA-hPD-L1 cells (Shanghai Jiaotong University cell bank), cultured in vitro as a monolayer, and the culture conditions are DMEM medium containing 10% fetal bovine serum , cultured at 37 °C in a 5% CO 2 incubator. Routine digestion with trypsin-EDTA was performed for passage. When the cells were in the exponential growth phase and the saturation was 80%-90%, the cells were harvested and counted.
  • Compound preparation Measure the compound of formula X and add it into the solvent (40% sulfobutyl- ⁇ -cyclodextrin (captisol) in acetic acid buffer, pH 4.0) to prepare a sample with the target concentration.
  • Experimental operation resuspend the cells in phosphate buffered saline at a density of 5 ⁇ 10 6 cells/mL.
  • the mice were randomly divided into groups according to their body weight, with 10 mice in each group.
  • the drug was administered twice a day for 19 days. Animals were weighed and their health monitored daily throughout the experimental period. Tumor diameters were measured twice a week with vernier calipers.
  • V 0.5 ⁇ a ⁇ b 2 , where a and b represent the long diameter and short diameter of the tumor, respectively.
  • the antitumor efficacy of compounds was evaluated by relative tumor proliferation rate T/C (%).
  • Relative tumor proliferation rate T/C (%) Vt/Vc ⁇ 100% (Vt: average tumor volume of treatment group; Vc: average tumor volume of negative control group). Vt and Vc take the data of the same day. The results at the end of the administration are shown in Table 17.
  • the DSC spectrum of the free base crystal form I is shown in Figure 2, in which the endothermic peak is 179.10°C, and there is an endothermic peak at 104.32°C, and the TGA spectrum is shown in Figure 3, and the weight loss before 150°C is about 8.8%.
  • Form I contains the solvent ethanol.

Landscapes

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

Abstract

一种氮杂芳环类化合物及其药学上可接受的盐的多晶型物、包含其的药物组合物、其制备方法,和该多晶型物或包含其的药物组合物在制备预防或治疗由腺苷A2A受体和/或腺苷A2B受体介导的癌症或免疫相关疾病的药物中的应用。

Description

氮杂芳环类化合物及其药学上可接受的盐的多晶型物、药物组合物和应用
相关申请
本申请要求2021年9月15日申请的,申请号为No.202111080394.2,名称为“氮杂芳环类化合物及其药学上可接受的盐的多晶型物、药物组合物和应用”的中国专利申请的优先权,在此将其全文引入作为参考。
技术领域
本申请涉及医药技术领域,特别涉及一种氮杂芳环类化合物3-(4-氨基-6-(1-((1-乙基-1H-吡唑-3-基)甲基)-1H-1,2,3-***-4-基)-5-氟嘧啶-2-基)-2-甲基苯甲腈或其药学上可接受的盐的多晶型物,以及包含该多晶型物的药物组合物、该多晶型物及该药物组合物的制备方法和应用。
背景技术
腺苷(Adenosine)是一种遍布人体细胞的内源性核苷,由腺嘌呤和核糖组成,其广泛分布于细胞内和细胞外。腺苷参与体内多种生理生化功能,例如,腺苷能直接进入心肌,经磷酸化生成三磷酸腺苷酸(Adenosine triphosphate,ATP),参与心肌的能量代谢。在中枢神经***(Central Nervous System,CNS)中,腺苷控制着神经递质的释放和突触后神经元的反应,起着调节运动、保护神经元、影响睡眠以及觉醒等重要生命过程的作用。在病理状态下,细胞外的腺苷浓度在肿瘤或者缺氧条件下会显著增加。腺苷可以通过促进肿瘤血管生成、增殖、发展以及肿瘤迁移在肿瘤免疫抑制中发挥着重要作用。
腺苷受体(Adenosine Receptor,AR)属于G蛋白偶联受体(Guanosine-binding Protein Coupled Receptor,GPCR)家族,其内源配体为腺苷。目前已知的腺苷受体由A 1、A 2A、A 2B和A 3四种亚型受体组成。其中腺苷与A 1或A 3受体结合可以抑制环磷酸腺苷(cAMP)的产生;而与A 2A或A 2B受体结合可以活化腺苷活化酶,进而上调cAMP的水平,发挥进一步的生理调控作用。
A 1和A 3两种受体主要表达在中枢神经***中,而A 2A和A 2B两种腺苷受体在中枢神经***及外周***都有表达。在肿瘤微环境内,A 2A和A 2B两种腺苷受体广泛表达在免疫细胞中,具有很强的免疫抑制功能。细胞外腺苷浓度的增加是肿瘤细胞免疫逃逸的重要作用机制之一,其浓度水平是由ATP的水平以及CD39与CD73的表达水平共同决定的。细胞外腺苷浓度的增加与肿瘤微环境里细胞死亡或者缺氧释放大量的ATP相关,其浓度可以达到正常组织的10-20倍。腺苷与肿瘤微环境里的腺苷受体结合,可以抑制抗肿瘤反应,例如抑制CD8+T细胞的功能,增强免疫抑制调节T细胞的功能,通过树突状细胞抑制抗原递呈细胞的功能等。最近的研究表明,与A 2A受体结合还可以抑制自然杀伤细胞的肿瘤杀伤作用。进一步研究表明,A 2A腺苷受体拮抗剂能提高树突状抗原递呈细胞、T细胞以及自然杀伤细胞的活力和杀伤能力,抑制调节性T细胞(T-regs)、骨髓来源的抑制性细胞(MDSCs)和肿瘤相关巨噬细胞(TAM)的功能,消除肿瘤免疫耐受,促进肿瘤免疫应答的发生,进而导致肿瘤生长受到抑制,并延长小鼠的生存期。此外,A 2B受体还被报道在鼠黑色素瘤和三阴乳腺癌模型中可以促进肿瘤的迁移,因此A 2B受体拮抗剂也是有效的癌症治疗靶标。所以阻断腺苷信号通路活化减少或解除免疫抑制,增强免疫细胞-尤其是T细胞的抗肿瘤功能被认为是癌症治疗的有效手段之一。而A 2A/A 2B双重受体拮抗剂用来同时阻断这两种受体的活化,从机制上说,是在调控不同的免疫细胞群,综合阻断微环境内腺苷带来的免疫抑制作用,对肿瘤治疗有着深远的临床应用价值。
为了更好地满足市场需求,进一步开发具有A 2A/A 2B受体拮抗活性的化合物及其药学上可接受的盐和其多晶型物以有助于进一步的药物开发。
发明内容
本申请提供一种式(X)所示氮杂芳环类化合物或其药学上可接受的盐的多晶型物、包含其的药物组合物、其制备方法或应用,
Figure PCTCN2022118654-appb-000001
所述药学上可接受的盐选自:盐酸盐、硫酸盐、柠檬酸盐、富马酸盐或琥珀酸盐。
在一些实施方案中,所述式X化合物的多晶型物或式X化合物药学上可接受盐的多晶型物各自独立地为无水形式、水合物形式或溶剂合物形式。
本申请中涉及的各X射线粉末衍射图,各自可以独立地使用Cu-Kα辐射而获得。
本申请的第一方面,提供式(X)所示化合物的I型晶型,即游离碱晶型I,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:7.57±0.2、13.41±0.2、14.64±0.2、19.81±0.2和24.43±0.2。
在一些实施方案中,所述游离碱晶型I的X射线粉末衍射图谱还包括1个或2个选自下列2θ(°)角处的特征衍射峰:24.87±0.2和27.46±0.2。
在一些实施方案中,所述游离碱晶型I的X射线粉末衍射图谱还包括2个或更多个选自下列2θ(°)角处的特征衍射峰:8.56±0.2、15.40±0.2、18.55±0.2、21.25±0.2、23.05±0.2、23.59±0.2和25.60±0.2。
在一些实施方案中,所述游离碱晶型I的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:7.57±0.2、8.56±0.2、10.60±0.2、11.80±0.2、13.41±0.2、14.16±0.2、14.64±0.2、15.40±0.2、16.63±0.2、17.23±0.2、18.55±0.2、19.81±0.2、21.25±0.2、22.18±0.2、22.46±0.2、23.05±0.2、23.59±0.2、24.43±0.2、24.87±0.2、25.60±0.2、26.03±0.2和27.46±0.2。
在一些实施方案中,所述游离碱晶型I的X射线粉末衍射图在表1所示的2θ(°)值处具有衍射峰,各衍射峰相对强度如表1所示。其中,各相对强度符号的定义参见表12。
表1
2θ(°) I/I 0 2θ(°) I/I 0 2θ(°) I/I 0
7.57 VS 16.63 W 23.59 M
8.56 M 17.23 W 24.43 S
10.60 W 18.55 M 24.87 S
11.80 W 19.81 S 25.60 M
13.41 S 21.25 M 26.03 W
14.16 W 22.18 W 27.46 S
14.64 S 22.46 W - -
15.40 M 23.05 M - -
在一些实施方案中,所述游离碱晶型I的X射线粉末衍射图谱基本如图1所示。
在一些实施方案中,所述游离碱晶型I的X射线粉末衍射图谱如图1所示。
在一些实施方案中,所述游离碱晶型I的差示扫描量热曲线在179.10±3℃处具有吸热峰。
在一些实施方案中,所述游离碱晶型I的差示扫描量热曲线在179.10±2℃处具有吸热峰。
在一些实施方案中,所述游离碱晶型I的差示扫描量热曲线在179.10±0.5℃处具有吸热峰。
在一些实施方案中,所述游离碱晶型I的差示扫描量热曲线基本如图2所示。
在一些实施方案中,所述游离碱晶型I的差示扫描量热曲线如图2所示。
本申请的第二方面,提供游离碱晶型I的制备方法,包括如下步骤:
将式(X)化合物与第I-3溶剂混合,在45℃至85℃的温度下加热至溶解,冷却至0℃至室温,分离以获得第I-3固体,并且
将所述第I-3固体干燥,获得所述游离碱晶型I;
其中所述第I-3溶剂为甲醇或乙醇。
在一些具体的实施方案中,游离碱晶型I的制备方法包括如下步骤:
将式(X)化合物与第I-3溶剂混合,在45℃至55℃的温度下加热至溶解,冷却至0℃至4℃,分离以获得第I-3固体,并且
将所述第I-3固体干燥,获得所述游离碱晶型I;
其中所述第I-3溶剂为甲醇或乙醇。
在一些具体的实施方案中,游离碱晶型I的制备方法包括如下步骤:
将式(X)化合物与第I-3A溶剂混合,在70℃~80℃的温度下加热至溶解,冷却至室温,分离以获得第I-3固体,并且
将所述第I-3固体干燥,获得所述游离碱晶型I;
其中所述第I-3A溶剂为乙醇。
在一些具体的实施方案中,游离碱晶型I的制备方法包括如下步骤:
将式(X)化合物与第I-3B溶剂混合,在60℃~70℃的温度下加热至溶解,冷却至室温,分离以获得第I-3固体,并且
将所述第I-3固体干燥,获得所述游离碱晶型I;
其中所述第I-3B溶剂为甲醇。
本申请的第三方面,提供式(X)化合物的IV型晶型,即游离碱晶型IV,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:11.88±0.2、14.17±0.2、16.96±0.2、22.63±0.2、23.56±0.2和25.66±0.2。
在一些实施方案中,所述游离碱晶型IV的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:15.76±0.2和26.08±0.2。
在一些实施方案中,所述游离碱晶型IV的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:15.76±0.2、26.08±0.2和37.48±0.2。
在一些实施方案中,所述游离碱晶型IV的X射线粉末衍射图谱还包括2个或更多个选自下列2θ(°)角处的特征衍射峰:14.89±0.2、21.73±0.2、24.43±0.2、26.65±0.2、27.39±0.2、28.42±0.2和30.24±0.2。
在一些实施方案中,所述游离碱晶型IV的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:8.71±0.2、19.93±0.2和31.78±0.2。
在一些实施方案中,所述游离碱晶型IV的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:8.71±0.2、9.57±0.2、11.88±0.2、14.17±0.2、14.89±0.2、15.76±0.2、16.96±0.2、19.93±0.2、21.73±0.2、22.63±0.2、23.56±0.2、24.43±0.2、25.66±0.2、26.08±0.2、26.65±0.2、27.39±0.2、28.42±0.2、30.24±0.2、31.78±0.2和33.15±0.2。
在一些实施方案中,所述游离碱晶型IV的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:8.71±0.2、9.57±0.2、11.88±0.2、14.17±0.2、14.89±0.2、15.76±0.2、16.96±0.2、19.93±0.2、21.73±0.2、22.63±0.2、23.56±0.2、24.43±0.2、25.66±0.2、26.08±0.2、26.65±0.2、27.39±0.2、28.42±0.2、30.24±0.2、31.78±0.2、33.15±0.2和37.48±0.2。
在一些实施方案中,所述游离碱晶型IV的X射线粉末衍射图在表2所示的2θ(°)值处具有衍射峰,各衍射峰相对强度如表2所示。其中,各相对强度符号的定义参见表12。
表2
2θ(°) I/I 0 2θ(°) I/I 0 2θ(°) I/I 0
8.71 M 19.93 M 26.65 M
9.57 W 21.73 M 27.39 M
11.88 VS 22.63 S 28.42 M
14.17 VS 23.56 VS 30.24 M
14.89 M 24.43 M 31.78 M
15.76 S 25.66 VS 33.15 W
16.96 VS 26.08 S 37.48 S
在一些实施方案中,所述游离碱晶型IV的X射线粉末衍射图谱基本如图4所示。
在一些实施方案中,所述游离碱晶型IV的X射线粉末衍射图谱如图4所示。
在一些实施方案中,所述游离碱晶型IV的差示扫描量热曲线在170.43±3℃处具有吸热峰。
在一些实施方案中,所述游离碱晶型IV的差示扫描量热曲线在170.43±2℃处具有吸热峰。
在一些实施方案中,所述游离碱晶型IV的差示扫描量热曲线在170.43±0.5℃处具有吸热峰。
在一些实施方案中,所述游离碱晶型IV的差示扫描量热曲线基本如图5所示。
在一些实施方案中,所述游离碱晶型IV的差示扫描量热曲线如图5所示。
本申请的第四方面,提供游离碱晶型IV的制备方法,包括如下步骤:
将本申请的第一方面所述的游离碱晶型I与第IV-1溶剂混合,在室温下混悬振摇20至30小时,分离以获得第IV-1固体,将所述第IV-1固体干燥,获得所述游离碱晶型IV;其中所述第IV-1溶剂为异丙醇;或
将本申请的第一方面所述的游离碱晶型I与第IV-2溶剂混合,在室温下混悬振摇160至170小时,分离以获得第IV-2固体,将所述第IV-2固体干燥,获得所述游离碱晶型IV;其中所述第IV-2溶剂为异丙醇或水。
本申请的第五方面,提供式(X)化合物的V型晶型,即游离碱晶型V,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:11.88±0.2、16.36±0.2、16.99±0.2、22.99±0.2、23.47±0.2和26.41±0.2。
在一些实施方案中,所述游离碱晶型V的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:11.88±0.2、16.36±0.2、16.99±0.2、22.99±0.2、23.47±0.2、26.41±0.2和37.48±0.2。
在一些实施方案中,所述游离碱晶型V的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:7.90±0.2、13.71±0.2、14.14±0.2、15.31±0.2、17.80±0.2、18.76±0.2、21.34±0.2、24.82±0.2、25.51±0.2、27.88±0.2和30.46±0.2。
在一些实施方案中,所述游离碱晶型V的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:7.90±0.2、13.71±0.2、14.14±0.2、15.31±0.2、17.80±0.2、18.76±0.2、21.34±0.2、24.82±0.2、25.51±0.2、27.88±0.2、30.46±0.2和43.60±0.2。
在一些实施方案中,所述游离碱晶型V的X射线粉末衍射图谱还包括2个或更多个选自下列2θ(°)角处的特征衍射峰:15.77±0.2、20.03±0.2、28.40±0.2、30.85±0.2、32.71±0.2、33.15±0.2和34.62±0.2。
在一些实施方案中,所述游离碱晶型V的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:7.90±0.2、8.62±0.2、9.73±0.2、11.88±0.2、13.71±0.2、14.14±0.2、15.31±0.2、15.77±0.2、16.36±0.2、16.99±0.2、17.80±0.2、18.76±0.2、20.03±0.2、21.34±0.2、22.99±0.2、23.47±0.2、24.82±0.2、25.51±0.2、26.41±0.2、27.88±0.2、28.40±0.2、30.46±0.2、30.85±0.2、32.71±0.2、33.15±0.2和34.62±0.2。
在一些实施方案中,所述游离碱晶型V的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:7.90±0.2、8.62±0.2、9.73±0.2、11.88±0.2、13.71±0.2、14.14±0.2、15.31±0.2、15.77±0.2、16.36±0.2、16.99±0.2、17.80±0.2、18.76±0.2、20.03±0.2、21.34±0.2、22.99±0.2、23.47±0.2、24.82±0.2、25.51±0.2、26.41±0.2、27.88±0.2、28.40±0.2、30.46±0.2、30.85±0.2、32.71±0.2、33.15±0.2、34.62±0.2、37.48±0.2和43.60±0.2。
在一些实施方案中,所述游离碱晶型V的X射线粉末衍射图在表3所示的2θ(°)值处具有衍射峰,各衍射峰相对强度如表3所示。其中,各相对强度符号的定义参见表12。
表3
2θ(°) I/I 0 2θ(°) I/I 0 2θ(°) I/I 0
7.90 S 17.80 S 28.40 M
8.62 W 18.76 S 30.46 S
9.73 W 20.03 M 30.85 M
11.88 VS 21.34 S 32.71 M
13.71 S 22.99 VS 33.15 M
14.14 S 23.47 VS 34.62 M
15.31 S 24.82 S 37.48 VS
15.77 M 25.51 S 43.60 S
16.36 VS 26.41 VS - -
16.99 VS 27.88 S - -
在一些实施方案中,所述游离碱晶型V的X射线粉末衍射图谱基本如图6所示。
在一些实施方案中,所述游离碱晶型V的X射线粉末衍射图谱如图6所示。
在一些实施方案中,所述游离碱晶型V的差示扫描量热曲线在179.02±3℃处具有吸热峰。
在一些实施方案中,所述游离碱晶型V的差示扫描量热曲线在179.02±2℃处具有吸热峰。
在一些实施方案中,所述游离碱晶型V的差示扫描量热曲线在179.02±0.5℃处具有吸热峰。
在一些实施方案中,所述游离碱晶型V的差示扫描量热曲线基本如图7所示。
在一些实施方案中,所述游离碱晶型V的差示扫描量热曲线如图7所示。
本申请的第六方面,提供游离碱晶型V的制备方法,包括如下步骤:
将本申请的第一方面所述的游离碱晶型I与第V-1溶剂混合,在室温下混悬振摇160至170小时,分离以获得第V-1固体,将所述第V-1固体干燥,获得所述游离碱晶型V;其中所述第V-1溶剂为甲基叔丁基醚。
将本申请的第一方面所述的式(X)化合物与第V-2溶剂混合并溶解,在室温下放置,使所述第V-2溶剂挥发至干,获得所述游离碱晶型V;其中所述第V-2溶剂为丙酮。
本申请的第七方面,提供式(X)化合物的VIII型晶型,即游离碱晶型VIII,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:6.40±0.2、13.12±0.2和15.91±0.2。
在一些实施方案中,所述的游离碱晶型VIII的X射线粉末衍射图谱还包括1个或2个选自下列2θ(°)角处的特征衍射峰:22.69±0.2和26.95±0.2。
在一些实施方案中,所述的游离碱晶型VIII的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:11.62±0.2、12.36±0.2、16.62±0.2、18.34±0.2、18.88±0.2、24.94±0.2和29.65±0.2。
在一些实施方案中,所述的游离碱晶型VIII的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:11.62±0.2、12.36±0.2、16.62±0.2、18.34±0.2、18.88±0.2、24.94±0.2、29.65±0.2和37.48±0.2。
在一些实施方案中,所述游离碱晶型VIII的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:6.40±0.2、11.62±0.2、12.36±0.2、13.12±0.2、15.91±0.2、16.62±0.2、18.34±0.2、18.88±0.2、22.69±0.2、24.94±0.2、26.95±0.2和29.65±0.2。
在一些实施方案中,所述游离碱晶型VIII的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:6.40±0.2、11.62±0.2、12.36±0.2、13.12±0.2、15.91±0.2、16.62±0.2、18.34±0.2、18.88±0.2、22.69±0.2、24.94±0.2、26.95±0.2、29.65±0.2和37.48±0.2。
在一些实施方案中,所述游离碱晶型VIII的X射线粉末衍射图在表4所示的2θ(°)值处具有衍射峰,各衍射峰相对强度如表4所示。其中,各相对强度符号的定义参见表12。
表4
2θ(°) I/I 0 2θ(°) I/I 0 2θ(°) I/I 0
6.40 VS 16.62 M 26.95 M
11.62 M 18.34 M 29.65 M
12.36 M 18.88 M 37.48 M
13.12 VS 22.69 M - -
15.91 S 24.94 M - -
在一些实施方案中,所述游离碱晶型VIII的X射线粉末衍射图谱基本如图8所示。
在一些实施方案中,所述游离碱晶型VIII的X射线粉末衍射图谱如图8所示。
在一些实施方案中,所述游离碱晶型VIII的差示扫描量热曲线在162.93±3℃处具有吸热峰。
在一些实施方案中,所述游离碱晶型VIII的差示扫描量热曲线在162.93±2℃处具有吸热峰。
在一些实施方案中,所述游离碱晶型VIII的差示扫描量热曲线在162.93±0.5℃处具有吸热峰。
在一些实施方案中,所述游离碱晶型VIII的差示扫描量热曲线基本如图9所示。
在一些实施方案中,所述游离碱晶型VIII的差示扫描量热曲线如图9所示。
本申请的第八方面,提供游离碱晶型VIII的制备方法,包括如下步骤:
将以上各方面中所述的游离碱晶型I、游离碱晶型Ⅳ或游离碱晶型V与第Z 3-1溶剂混合,在室温下混悬振摇70至80小时,分离以获得第Z 3-1固体,将所述第Z 3-1固体干燥,获得所述游离碱晶型VIII;其中所述第Z 3-1溶剂为丙酮或体积分数为50%的乙醇水溶液。
本申请的第九方面,提供式(X)化合物的盐酸盐晶型I,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:11.95±0.2、24.37±0.2、25.21±0.2和26.35±0.2。
在一些实施方案中,所述盐酸盐晶型I的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:8.83±0.2、14.13±0.2、15.33±0.2、15.68±0.2、18.86±0.2、23.64±0.2、27.67±0.2和28.57±0.2。
在一些实施方案中,所述盐酸盐晶型I的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:8.83±0.2、11.95±0.2、14.13±0.2、15.33±0.2、15.68±0.2、17.25±0.2、18.86±0.2、23.64±0.2、24.37±0.2、25.21±0.2、26.35±0.2、27.67±0.2和28.57±0.2。
在一些实施方案中,所述盐酸盐晶型I的X射线粉末衍射图在表5所示的2θ(°)值处具有衍射峰,各衍射峰相对强度如表5所示。其中,各相对强度符号的定义参见表12。
表5
2θ(°) I/I 0 2θ(°) I/I 0 2θ(°) I/I 0
8.83 S 17.25 M 26.35 VS
11.95 VS 18.86 S 27.67 S
14.13 S 23.64 S 28.57 S
15.33 S 24.37 VS - -
15.68 S 25.21 VS - -
在一些实施方案中,所述盐酸盐晶型I的X射线粉末衍射图谱基本如图10所示。
在一些实施方案中,所述盐酸盐晶型I的X射线粉末衍射图谱如图10所示。
本申请的第十方面,提供式(X)化合物的硫酸盐晶型I,
其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:8.22±0.2、11.29±0.2、14.14±0.2、16.96±0.2、18.13±0.2、22.27±0.2、23.86±0.2和27.79±0.2。
在一些实施方案中,所述硫酸盐晶型I的X射线粉末衍射图谱还包括2个或更多个选自下列2θ(°)角处的特征衍射峰:6.54±0.2和11.64±0.2。
在一些实施方案中,所述硫酸盐晶型I的X射线粉末衍射图谱还包括2个或更多个选自下列2θ(°)角处的特征衍射峰:6.54±0.2、11.64±0.2和37.48±0.2。
在一些实施方案中,所述硫酸盐晶型I的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:6.54±0.2、8.22±0.2、11.29±0.2、11.64±0.2、14.14±0.2、16.96±0.2、18.13±0.2、22.27±0.2、23.86±0.2和27.79±0.2。
在一些实施方案中,所述硫酸盐晶型I的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:6.54±0.2、8.22±0.2、11.29±0.2、11.64±0.2、14.14±0.2、16.96±0.2、18.13±0.2、22.27±0.2、23.86±0.2、27.79±0.2和37.48±0.2。
在一些实施方案中,所述硫酸盐晶型I的X射线粉末衍射图在表6所示的2θ(°)值处具有衍射峰,各衍 射峰相对强度如表6所示。其中,各相对强度符号的定义参见表12。
表6
2θ(°) I/I 0 2θ(°) I/I 0 2θ(°) I/I 0
6.54 M 14.14 S 23.86 S
8.22 S 16.96 VS 27.79 S
11.29 S 18.13 S 37.48 M
11.64 M 22.27 S - -
在一些实施方案中,所述硫酸盐晶型I的X射线粉末衍射图谱基本如图11所示。
在一些实施方案中,所述硫酸盐晶型I的X射线粉末衍射图谱如图11所示。
本申请的第十一方面,提供式(X)化合物的柠檬酸盐晶型I,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:12.55±0.2、14.50±0.2、21.55±0.2和22.75±0.2。
在一些实施方案中,所述柠檬酸盐晶型I的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:12.55±0.2、14.50±0.2、21.55±0.2、22.75±0.2、37.51±0.2和43.63±0.2。
在一些实施方案中,所述柠檬酸盐晶型I的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:5.38±0.2、9.78±0.2和13.63±0.2。
在一些实施方案中,所述柠檬酸盐晶型I的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:5.38±0.2、9.78±0.2、12.55±0.2、13.63±0.2、14.50±0.2、21.55±0.2和22.75±0.2。
在一些实施方案中,所述柠檬酸盐晶型I的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:5.38±0.2、9.78±0.2、12.55±0.2、13.63±0.2、14.50±0.2、21.55±0.2、22.75±0.2、37.51±0.2和43.63±0.2。
在一些实施方案中,所述柠檬酸盐晶型I的X射线粉末衍射图在表7所示的2θ(°)值处具有衍射峰,各衍射峰相对强度如表7所示。其中,各相对强度符号的定义参见表12。
表7
2θ(°) I/I 0 2θ(°) I/I 0 2θ(°) I/I 0
5.38 S 13.63 S 22.75 VS
9.78 S 14.50 VS 37.51 VS
12.55 VS 21.55 VS 43.63 VS
在一些实施方案中,所述柠檬酸盐晶型I的X射线粉末衍射图谱基本如图12所示。
在一些实施方案中,所述柠檬酸盐晶型I的X射线粉末衍射图谱如图12所示。
本申请的第十二方面,提供式(X)化合物的富马酸盐晶型I,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:7.12±0.2、11.71±0.2和16.90±0.2。
在一些实施方案中,所述富马酸盐晶型I的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:23.26±0.2、23.65±0.2、24.64±0.2和25.54±0.2。
在一些实施方案中,所述富马酸盐晶型I的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:8.02±0.2、15.69±0.2、18.22±0.2和26.44±0.2。
在一些实施方案中,所述富马酸盐晶型I的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:8.02±0.2、15.69±0.2、18.22±0.2、26.44±0.2和37.87±0.2。
在一些实施方案中,所述富马酸盐晶型I的X射线粉末衍射图谱还包括2个或更多个选自下列2θ(°)角处的特征衍射峰:10.48±0.2、13.80±0.2、22.00±0.2和22.42±0.2。
在一些实施方案中,所述的富马酸盐晶型I的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:7.12±0.2、8.02±0.2、10.48±0.2、11.71±0.2、13.80±0.2、15.69±0.2、16.90±0.2、18.22±0.2、22.00±0.2、22.42±0.2、23.26±0.2、23.65±0.2、24.64±0.2、25.54±0.2和26.44±0.2。
在一些实施方案中,所述的富马酸盐晶型I的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:7.12±0.2、8.02±0.2、10.48±0.2、11.71±0.2、13.80±0.2、15.69±0.2、16.90±0.2、18.22±0.2、22.00±0.2、22.42±0.2、 23.26±0.2、23.65±0.2、24.64±0.2、25.54±0.2、26.44±0.2和37.87±0.2。
在一些实施方案中,所述富马酸盐晶型I的X射线粉末衍射图在表8所示的2θ(°)值处具有衍射峰,各衍射峰相对强度如表8所示。其中,各相对强度符号的定义参见表12。
表8
2θ(°) I/I 0 2θ(°) I/I 0 2θ(°) I/I 0
7.12 VS 16.90 VS 24.64 VS
8.02 S 18.22 S 25.54 VS
10.48 M 22.00 M 26.44 S
11.71 VS 22.42 M 37.87 S
13.80 M 23.26 VS - -
15.69 S 23.65 VS - -
在一些实施方案中,所述富马酸盐晶型I的X射线粉末衍射图谱基本如图13所示。
在一些实施方案中,所述富马酸盐晶型I的X射线粉末衍射图谱如图13所示。
本申请的第十三方面,提供式(X)化合物的琥珀酸盐晶型I,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:13.78±0.2、14.14±0.2和22.12±0.2。
在一些实施方案中,琥珀酸盐晶型I的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:13.78±0.2、14.14±0.2、22.12±0.2和37.84±0.2。
在一些实施方案中,所述琥珀酸盐晶型I的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:4.87±0.2、9.31±0.2、18.16±0.2、20.71±0.2和26.83±0.2。
在一些实施方案中,所述琥珀酸盐晶型I的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:4.87±0.2、9.31±0.2、13.78±0.2、14.14±0.2、15.69±0.2、18.16±0.2、20.71±0.2、22.12±0.2和26.83±0.2。
在一些实施方案中,所述琥珀酸盐晶型I的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:
4.87±0.2、9.31±0.2、13.78±0.2、14.14±0.2、15.69±0.2、18.16±0.2、20.71±0.2、22.12±0.2、26.83±0.2、37.84±0.2和44.07±0.2。
在一些实施方案中,所述琥珀酸盐晶型I的X射线粉末衍射图在表9所示的2θ(°)值处具有衍射峰,各衍射峰相对强度如表9所示。其中,各相对强度符号的定义参见表12。
表9
2θ(°) I/I 0 2θ(°) I/I 0 2θ(°) I/I 0
4.87 S 15.69 M 26.83 S
9.31 S 18.16 S 37.84 VS
13.78 VS 20.71 S 44.07 M
14.14 VS 22.12 S - -
在一些实施方案中,所述琥珀盐晶型I的X射线粉末衍射图谱基本如图14所示。
在一些实施方案中,所述琥珀盐晶型I的X射线粉末衍射图谱如图14所示。
本申请的第十四方面,提供式(X)化合物的琥珀酸盐晶型Ⅱ,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:12.49±0.2和14.50±0.2。
在一些实施方案中,所述琥珀酸盐晶型Ⅱ的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:11.86±0.2、15.22±0.2、16.90±0.2、21.64±0.2、22.87±0.2和25.75±0.2。
在一些实施方案中,所述琥珀酸盐晶型Ⅱ的X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:11.86±0.2、15.22±0.2、16.90±0.2、21.64±0.2、22.87±0.2、25.75±0.2和37.84±0.2。
在一些实施方案中,所述琥珀酸盐晶型Ⅱ的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:5.35±0.2、11.86±0.2、12.49±0.2、14.50±0.2、15.22±0.2、16.90±0.2、21.64±0.2、22.87±0.2和25.75±0.2。
在一些实施方案中,所述琥珀酸盐晶型Ⅱ的X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:5.35±0.2、11.86±0.2、12.49±0.2、14.50±0.2、15.22±0.2、16.90±0.2、21.64±0.2、22.87±0.2、25.75±0.2和37.84±0.2。
在一些实施方案中,所述琥珀酸盐晶型Ⅱ的X射线粉末衍射图在表10所示的2θ(°)值处具有衍射峰,各衍射峰相对强度如表10所示。其中,各相对强度符号的定义参见表12。
表10
2θ(°) I/I 0 2θ(°) I/I 0 2θ(°) I/I 0
5.35 M 15.22 S 25.75 S
11.86 S 16.90 S 37.84 S
12.49 VS 21.64 S - -
14.50 VS 22.87 S - -
在一些实施方案中,所述琥珀盐晶型Ⅱ的X射线粉末衍射图谱基本如图15所示。
在一些实施方案中,所述琥珀盐晶型Ⅱ的X射线粉末衍射图谱如图15所示。
本申请的第十五方面,提供式(X)化合物的药学上可接受的盐的多晶型物的制备方法,包括如下步骤:
将如上所述的式(X)化合物与酸按照1.1~1.5:1的酸(氢离子)与式(X)化合物摩尔比混合,添加1至3mL溶剂,超声加热使溶液澄清,在45℃至50℃下反应3至5小时,降低温度或加入反溶剂以析出固体形式的多晶型物。
在一些实施方案中,所述酸为盐酸、硫酸、柠檬酸、富马酸或琥珀酸。
在一些实施方案中,所述溶剂为乙醇、乙酸乙酯或丙酮。
在一些实施方案中,所述反溶剂为正庚烷。
本申请第十六方面提供了一种药物组合物,包括:
(a)如上所述的式(X)化合物或其药学上可接受盐的多晶型物;以及(b)药学上可接受的载体。
本申请的第十七方面,提供如上所述的式(X)化合物的游离碱晶型I、游离碱晶型IV、游离碱晶型V、游离碱晶型VIII,或其盐酸盐晶型I、硫酸盐晶型I、柠檬酸盐晶型I、富马酸盐晶型I、琥珀酸盐晶型I、琥珀酸盐晶型Ⅱ在制备A 2A受体或A 2B受体抑制剂中的应用。
本申请的第十八方面,提供如上所述的式(X)化合物或其药学上可接受盐的多晶型物,例如游离碱晶型I、游离碱晶型IV、游离碱晶型V、游离碱晶型VIII,或其盐酸盐晶型I、硫酸盐晶型I、柠檬酸盐晶型I、富马酸盐晶型I、琥珀酸盐晶型I、琥珀酸盐晶型Ⅱ,或本申请第十六方面所述的药物组合物在制备预防或治疗由腺苷A 2A受体和/或腺苷A 2B受体介导的疾病的药物中的用途。
本申请提供一种用于预防或治疗由腺苷A 2A受体和/或腺苷A 2B受体介导的疾病的方法,包括向有此需要的患者施用治疗有效量的如上实施方案中所述的式(X)化合物或其药学上可接受盐的多晶型物、或如上实施方案中所述的药物组合物。
本申请提供如上实施方案中所述的式(X)化合物或其药学上可接受盐的多晶型物、或如上实施方案中所述的药物组合物用于在预防或治疗由腺苷A 2A受体和/或腺苷A 2B受体介导的疾病中使用。
在一些实施方案中,所述疾病为癌症或免疫相关的疾病。
在一些实施方案中,所述癌症选自***癌、结肠癌、直肠癌、胰腺癌、子***、胃癌、子宫内膜癌、脑癌、肝癌、膀胱癌、卵巢癌、睾丸癌、头癌、颈癌、黑素瘤、基底癌、间皮内层癌、白血细胞癌、食道癌、乳腺癌、肌肉癌、***癌、小细胞肺癌、非小细胞肺癌、肾上腺癌、甲状腺癌、肾癌和骨癌;或是恶性胶质瘤、间皮瘤、肾细胞癌、胃癌、肉瘤、绒毛膜癌、皮肤基底细胞癌和睾丸***瘤。
在一些实施方案中,所述免疫相关的疾病选自类风湿性关节炎、肾衰竭、狼疮、哮喘、银屑病、结肠炎、胰腺炎、过敏、纤维化、贫血性纤维肌痛、阿尔茨海默氏病、充血性心力衰竭、中风、主动脉瓣狭窄、动脉硬化、骨质疏松症、帕金森氏病、感染、克罗恩氏病、溃疡性结肠炎、过敏性接触性皮炎和其他湿疹、***性硬化和多发性硬化症。
本申请的式(X)化合物、式X化合物的多晶型物、式X化合物药学上可接受盐的多晶型物,或包含式(X)化合物、式(X)化合物的多晶型物或式X化合物药学上可接受盐的多晶型物的药物组合物具有良好的抑制A 2A受体和/或A 2B受体的药理活性。另外,式(X)化合物的多晶型物及式X化合物药学上可接受的盐的多晶型物具有良好的稳定性,因此具有开发成为药物的潜力。
应理解,在本申请范围内,本申请的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的技术方案。限于篇幅,在此不再一一累述。
附图说明
为了更清楚地说明本申请实施例或传统技术中的技术方案,下面将对实施例或传统技术描述中所需要使用的附图作简单地介绍,显而易见地,下面描述中的附图仅仅是本申请的实施例,对于本领域普通技术人员来讲,在不付出创造性劳动的前提下,还可以根据公开的附图获得其他的附图。
图1为实施例2制备得到的式X化合物的游离碱晶型I的XPRD图谱(使用Cu-Kα辐射,横坐标为角度2θ(°),纵坐标为SQR(counts)),其中SQR表示强度的平方根;
图2为实施例2制备得到的式X化合物的游离碱晶型I的DSC图谱(横坐标为温度(℃),纵坐标为热流率(W/g));
图3为实施例2制备得到的式X化合物的游离碱晶型I的TGA图谱(横坐标为温度(℃),纵坐标为重量变化(%));
图4为实施例3制备得到的式X化合物的游离碱晶型Ⅳ的XPRD图谱(使用Cu-Kα辐射,横坐标为角度2θ(°),纵坐标为SQR(counts)),其中SQR表示强度的平方根;
图5为实施例3制备得到的式X化合物的游离碱晶型Ⅳ的DSC图谱(横坐标为温度(℃),纵坐标为热流率(W/g));
图6为实施例4制备得到的式X化合物的游离碱晶型V的XPRD图谱(使用Cu-Kα辐射,横坐标为角度2θ(°),纵坐标为强度(counts));
图7为实施例4制备得到的式X化合物的游离碱晶型V的DSC图谱(横坐标为温度(℃),纵坐标为热流率(W/g));
图8为实施例5制备得到的式X化合物的游离碱晶型Ⅷ的XPRD图谱(使用Cu-Kα辐射,横坐标为角度2θ(°),纵坐标为强度(counts));
图9为实施例5制备得到的式X化合物的游离碱晶型Ⅷ的DSC图谱(横坐标为温度(℃),纵坐标为热流率(W/g));
图10为实施例6制备得到的式X化合物的盐酸盐晶型I的XPRD图谱(使用Cu-Kα辐射,横坐标为角度2θ(°),纵坐标为强度(counts));
图11为实施例7制备得到的式X化合物的硫酸盐晶型I的XPRD图谱(使用Cu-Kα辐射,横坐标为角度2θ(°),纵坐标为强度(counts));
图12为实施例8制备得到的式X化合物的柠檬酸盐晶型I的XPRD图谱(使用Cu-Kα辐射,横坐标为角度2θ(°),纵坐标为强度(counts));
图13为实施例9制备得到的式X化合物的富马酸盐晶型I的XPRD图谱(使用Cu-Kα辐射,横坐标为角度2θ(°),纵坐标为强度(counts));
图14为实施例10制备得到的式X化合物的琥珀酸盐晶型I的XPRD图谱(使用Cu-Kα辐射,横坐标为角度2θ(°),纵坐标为强度(counts));
图15为实施例11制备得到的式X化合物的琥珀酸盐晶型Ⅱ的XPRD图谱(使用Cu-Kα辐射,横坐标为角度2θ(°),纵坐标为强度(counts))。
具体实施方式
以下结合具体实施例对本申请的式(X)化合物的多晶型物及式(X)化合物药学上可接受的盐的多晶型物、包含该多晶型物的药物组合物、该多晶型物及该药物组合物的制备方法、和其应用作进一步详细的说明。本申请可以以许多不同的形式来实现,并不限于本文所描述的实施方式。相反地,提供这些实施方式的目的是使对本申请公开内容理解更加透彻全面。
除非另有定义,本文所使用的所有的技术和科学术语与属于本申请的技术领域的技术人员通常理解的含义相同。本文中在本申请的说明书中所使用的术语只是为了描述具体的实施例的目的,不是旨在于限制本申请。
本申请中,“第一方面”、“第二方面”、“第三方面”、“第四方面”、“第五方面”等仅用于描述目的,不能理解为指示或暗示相对重要性或数量,也不能理解为隐含指明所指示的技术特征的重要性或数量。而且“第一”、“第二”、“第三”、“第四”、“第五”等仅起到非穷举式的列举描述目的,应当理解并不构成对数量的封闭式限定。
本申请中,以开放式描述的技术特征中,包括所列举特征组成的封闭式技术方案,也包括包含所列举特征的开放式技术方案。
本申请中,涉及到数值区间,如无特别说明,上述数值区间内视为连续,且包括该范围的最小值及最大值,以及这种最小值与最大值之间的每一个值。进一步地,当范围是指整数时,包括该范围的最小值与最大值之间的每一个整数。此外,当提供多个范围描述特征或特性时,可以合并该范围。换言之,除非另有指明,否则本文中所公开之所有范围应理解为包括其中所归入的任何及所有的子范围。
本申请中涉及的百分比含量,如无特别说明,对于固液混合和固相-固相混合均指质量百分比,对于液相-液相混合指体积百分比。
本申请中涉及的百分比浓度,如无特别说明,均指终浓度。所述终浓度,指添加成分在添加该成分后的体系中的占比。
本申请中的温度参数,如无特别限定,既允许为恒温处理,也允许在一定温度区间内进行处理。所述的恒温处理允许温度在仪器控制的精度范围内进行波动。
本申请的化合物
在本申请中,式(X)化合物为3-(4-氨基-6-(1-((1-乙基-1H-吡唑-3-基)甲基)-1H-1,2,3-***-4-基)-5-氟嘧啶-2-基)-2-甲基苯甲腈,其对腺苷A 2A受体和/或腺苷A 2B受体具有高抑制活性。
本申请中,式(X)化合物的多晶型物包括式(X)化合物游离碱的多晶型物,以及式(X)化合物药学上可接受盐的多晶型物。其中,药学上可接受的盐选自盐酸盐、硫酸盐、柠檬酸盐、富马酸盐或琥珀酸盐。式(X)化合物及其药学上可接受的盐的多晶型物包括但不限于式(X)化合物的游离碱晶型I、游离碱晶型Ⅳ、游离碱晶型Ⅴ、游离碱晶型VIII、盐酸盐晶型I、硫酸盐晶型I、柠檬酸盐晶型I、富马酸盐晶型I、琥珀酸盐晶型I和琥珀酸盐晶型Ⅱ。
如本文所用,“治疗有效量”是指将引起个体的生物学或医学响应,例如降低或抑制酶或蛋白质活性或改善症状、缓解病症、减缓或延迟疾病进程或预防疾病等的本申请化合物的量。
如本文所用,“药学可接受的载体”原则上应当是无毒、惰性的。“药学可接受的载体”的形态没有特别限定,包括但不限于固态、半固态、液态等。所述药学可接受的载体应与患者相兼容,所述患者以哺乳动物为佳,更具体地为人。所述药学可接受的载体的作用之一是适合将活性试剂输送到目标靶点而不终止试剂的活性。如本文所用,语言“药学上可接受的载体”包括与药物施用相容的缓冲剂、注射用无菌水、溶剂、分散介质、包衣、抗细菌剂及抗真菌剂、等渗剂及吸收延迟剂及诸如此类。在与配制物中其他成分兼容且对患者无害的意义上,每种载体为“药学上可接受的”。
如本文所用,“患者”是指一种动物,最好为哺乳动物,更好的为人。术语“哺乳动物”是指温血脊椎类哺乳动物,包括如猫、狗、兔、熊、狐狸、狼、猴子、鹿、鼠、猪和人类。
如本文所用,“治疗”是指减轻、延缓进展、衰减、预防,或维持现有疾病或病症(例如癌症)。治疗还包括将疾病或病症的一个或多个症状治愈、预防其发展或减轻到某种程度。
本申请中,“使用Cu-Kα辐射”指相应的谱图使用Cu靶的Kα射线检测获得,当采用其他方法检测时,各衍射峰可以存着本领域可接受范围内的偏差,不应理解为对本申请的限制。
多晶型物
固体不是以无定形的形式就是以结晶的形式存在。在结晶形式的情况下,分子定位于三维晶格格位内。当化合物从溶液或浆液中结晶出来时,它可以不同的空间点阵排列结晶(这种性质被称作“多晶型现象”),形成具有不同的结晶形式的晶体,这各种结晶形式被称作“多晶型物”。给定物质的不同多晶型物可在一个 或多个物理属性方面(如溶解度和溶解速率、真比重、晶形、堆积方式、流动性和/或固态稳定性)彼此不同。
结晶
可以通过操作溶液,使得化合物的溶解度极限被超过,从而完成生产规模的结晶。这可以通过多种方法来完成,例如,在相对高的温度下溶解化合物,然后冷却溶液至饱和极限以下;或者通过沸腾、常压蒸发、真空干燥或通过其它的一些方法来减小液体体积;可通过加入反溶剂或化合物在其中具有低的溶解度的溶剂或这样的溶剂的混合物,来降低化合物的溶解度。另一种可选方法是调节pH值以降低溶解度。有关结晶方面的详细描述请参见Crystallization,第三版,J W Mullens,Butterworth-Heineman Ltd.,1993,ISBN 0750611294。
晶型的鉴定和性质
本申请在制备式(X)化合物的多晶型物后,采用如下多种方式和仪器对其性质进行了研究。
X射线粉末衍射(XRPD)
测定晶型的X射线粉末衍射的方法在本领域中是已知的。XRPD可检测晶型的变化、结晶度、晶构状态等信息,是鉴别晶型的常用手段。XRPD图谱的峰位置主要取决于晶型的结构,不同仪器之间,XRPD图谱的2θ的量度可能会略有差别,因此所述2θ的数值不能视为绝对的。根据本申请试验所用仪器状况,衍射峰允许存在一定的误差(如±0.2°)。可以理解,对于不同的测试仪器及测试状况而言,误差的范围也不是绝对的。本申请的式X化合物的晶型,具有特定的晶型形态,在XRPD图中具有特定的特征峰。
本申请中,“X射线粉末衍射图在特定的2θ(°)角处具有特征衍射峰”,表示该峰的峰值在所指示的数值范围内,所指示的数值点,所指示数值范围的附近或所指示数值点的附近。由于测量仪器及测量状况等测量因素的不同,实际得到的X射线粉末衍射图中的某个峰或某些峰值的位置可能会略有偏移,也即与本申请指示的特征峰组合或者X射线粉末衍射图之间可能略有差别,但是,可以理解的是,对于本领域技术人员而言,能够从整体上鉴别略有差别的特征峰组合或者X射线粉末衍射图是否可以从实质上构成本申请所述的晶型。也因此,这些从实质上被认定为与发明的晶型一致的,应当认为在本申请的保护范围内。举例如,“X射线粉末衍射图在7.57±0.2衍射角处具有峰”表示,该峰的峰值可以位于7.57±0.2范围内及该范围的附近,只要不影响从整体上对晶型进行鉴别即可。此外,这里的“±0.2”仅表示该峰值在衍射角位置上的误差,与该峰的峰形、峰宽无关。
本申请中,“X射线粉末衍射图基本如特定图所表征”中的“基本”也应当作类似理解,只要能够从整体认定某个X射线粉末衍射图与本申请所述的X射线粉末衍射图构成实质上的一致,则应当认为落入了本申请的保护范围内。
可以理解的是,本申请中的差示扫描量热曲线及其中显示的吸热峰位置,也应当作类似理解,允许与本申请披露的具体数值或具体数值范围或具体谱图之间略有不符,但只要能够从整体上认定差示扫描量热曲线中的部分或全部吸热峰位置、或整个曲线与本申请实质上一致,则应当认为落入了本申请的保护范围内。
可以理解的是,对于其他表征结晶类型的谱图也可以作类似理解。
示差扫描量热分析(DSC)
又称“差示量热扫描分析”,是在加热过程中,测量被测物质与参比物之间的能量差与温度之间关系的一种技术。DSC图谱上的峰位置、形状和峰数目与物质的性质有关,故可以定性地用来鉴定物质。本领域常用该方法来检测物质的相变温度、玻璃化转变温度、反应热等多种参数。不同仪器之间,DSC图谱的峰位置可能会略有差别,因此所述DSC吸热峰的峰位置的数值不能视为绝对的。根据本申请试验所用仪器状况,实验误差或差别的数值可能小于等于5℃,或小于等于4℃,或小于等于3℃,或小于等于2℃,或小于等于1℃。
热重分析(TGA)
TGA是在程序控制下,测定物质的质量随温度变化的一种技术,适用于检查晶体中溶剂的丧失或样品升华、分解的过程,可推测晶体中含结晶水或结晶溶剂的情况。TGA曲线显示的质量变化取 决于样品制备和仪器等许多因素;不同仪器之间TGA检测的质量变化略有差别。根据本申请试验所用的仪器状况,质量变化的误差不是绝对的,允许存在一定的误差(如±0.1%)。
假如期望盐的形成与结晶同时发生,如果盐在反应介质中比原料溶解度小,那么加入适当的酸或碱可导致所需盐的直接结晶。同样,在最终想要的形式比反应物溶解度小的介质中,合成反应的完成可使最终产物直接结晶。
结晶的优化可包括用所需形式的晶体作为晶种接种于结晶介质中。另外,许多结晶方法使用上述策略的组合。一个实施例是在高温下将感兴趣的化合物溶解在溶剂中,随后通过受控方式加入适当体积的抗溶剂,以使体系正好在饱和水平之下。此时,可加入所需形式的晶种(并保持晶种的完整性),将体系冷却以完成结晶。
如本文所用,术语“室温”一般指4-30℃,较佳地指20±5℃。
多晶型物的鉴定和性质
X射线粉末衍射
本申请的式(X)化合物的多晶型物具有特定的晶型形态,在X射线粉末衍射图(XPRD)中具有特定的特征峰。XPRD图谱在Equinox3000S/N X射线粉末衍射分析仪上采集,XPRD参数如下表11所示。
表11
参数 XPRD
X射线源 Cu K(λ=1.54056 Angstrom)
光管设定 40千伏,30毫安
检测器 PSD
扫描范围(2θ°) 0°~120°
扫描步长(2θ°) 0.03
扫描速率 1秒/步
在X射线粉末衍射图中,各峰的位置由2θ(°)确定。可以理解,不同的仪器和/或条件可导致产生的数据会略有不同,各峰的位置和相对强度会有变化。本领域技术人员应理解,在依据XRPD图谱判定晶型时,低衍射角度的峰及其强度、峰形完整性等要素是相对更具有参考意义的。本申请中,申请人经过进一步的研究,发现在使用上述仪器测试时,在高衍射角度的2θ(°)值为37和44两处附近出现了空白金属盘的强背景峰。比如,图4中37°附近的峰推测为空白金属盘产生的峰(如表2中的37.48°处的峰),图6中37°附近的峰以及43°附近的峰推测为空白金属盘产生的峰(如表3中的37.48°、43.60°处的峰),图8中37°附近的峰推测为空白金属盘产生的峰(如表4中的37.48°处的峰),图11中37°附近的峰推测为空白金属盘产生的峰(如表6中的37.48°处的峰),图12中37°附近的峰以及43°附近的峰推测为空白金属盘产生的峰(如表7中的37.51°、43.63°处的峰),图13中37°附近的峰推测为空白金属盘产生的峰(如表8中的37.87°处的峰),图14中37°附近的峰以及43°附近的峰推测为空白金属盘产生的峰(如表9中的37.84°、44.07°处的峰),图15中37°附近的峰推测为空白金属盘产生的峰(如表10中的37.84°处的峰)。峰的强度划分仅仅反映了各位置上峰的近似大小。在本申请中,各晶型均以其峰高最高的衍射峰作为基峰,定义其相对强度为100%,作为I 0,其它各峰以其峰高与基峰峰高的比值作为其相对强度I/I 0,各峰相对强度的划分定义如下表12所示。
表12
相对强度I/I 0(%) 定义
50~100 VS(很强)
25~50 S(强)
10~25 M(中等)
1~10 W(弱)
差示扫描量热(DSC)
DSC图谱在DISCOVERY DSC25差示扫描量热仪上采集,测试参数如下表13所示。
表13
参数 DSC
方法 线性升温
样品盘 铝盘,压盖
温度范围 25℃–300℃
扫描速率(℃/分钟) 10
保护气体 氮气
可以理解的是,使用与上述仪器作用相同的其他类型的仪器或使用不同与本申请中使用的测试条件时,可能会得到另外的数值,因此,所引用的数值不应视为绝对的数值。
由于仪器的误差或操作人员的区别,本领域技术人员能理解,以上用于表征晶体的物理性质的参数可能有微小的差别,所以上述的参数仅用于辅助表征本申请提供的多晶型物,而不能视为是对本申请的多晶型物的限制。
试剂与仪器
本申请中,化合物的结构和纯度通过核磁共振( 1HNMR)和、或液质联用质谱(LC-MS)来确定。 1HNMR:BrukerAVANCE-400核磁仪,内标为四甲基硅烷(TMS)。LC-MS:Agilent 1200 HPLC System/6140 MS液质联用质谱仪(生产商:安捷伦),柱子WatersX-Bridge,150×4.6mm,3.5μm。
本申请已知的起始原料可以采用或按照本领域已知的方法来合成,或可以于ABCR GmbH&Co.KG,Acros Organics,Aldrich Chemical Company,韶远化学科技(Accela ChemBio Inc)和达瑞化学品等公司处购买。
如本文所用,DCM:二氯甲烷,DMF:二甲基甲酰胺,DMSO:二甲基亚砜,THF:四氢呋喃,EA:乙酸乙酯,PE:石油醚,Pd(dppf)Cl 2:[1,1'-双(二苯基磷)二茂铁]二氯化钯,Pd(PPh 3) 2Cl 2:双(三苯基膦)氯化钯(Ⅱ),DPPA:叠氮磷酸二苯酯,DBU:1,8-二氮杂二环十一碳-7-烯。
中间体V1的制备
Figure PCTCN2022118654-appb-000002
将2-甲基-3-溴苯腈(15g,76.51mmol)、双联频哪醇硼酸酯(23.32g,91.82mmol)、醋酸钾(15.02g,153.03mmol)和Pd(dppf)Cl 2(2.80g)溶解于DMSO(20mL)和二氧六环(100mL)的混合溶液中,氮气保护下,在100℃下搅拌3.5小时。反应完全,减压蒸干溶剂得到的固体产物,通过柱层析(EA:PE15%~40%)中分离得到化合物V1(21.05g,纯度:78.0%,收率:100%)。MS(ESI)244.1[M+H] +
中间体1-5的制备
Figure PCTCN2022118654-appb-000003
步骤一:
化合物V2-1(40g,198.60mmol)和三异丙基硅基乙炔(38.03g 208.53mmol)溶解在THF(400.00mL)中,氩气保护下,搅拌降温至0℃,依次加入CuI(3.78g,19.86mmol),Pd(PPh 3) 2Cl 2(6.97g,9.93mmol),然后缓慢滴加Et 3N(60.17g,595.79mmol)。滴加完毕,0℃搅拌2小时,升温至室温继续搅拌6小时,然后降温至0℃,滴加NH 3/H 2O(120.00g,2.61mol,120.00mL,37%纯度),然后室温搅拌过夜。LC-MS检测反应完全,得到化合物V2-2。MS(ESI)347[M+H]。
步骤二:
向步骤1中的反应液加入大量水中,DCM(200mL×3)萃取,无水硫酸钠干燥,减压浓缩剩余DCM 40mL左右,大量固体析出,搅拌加入石油醚60mL,过滤得到化合物V2-3(31g),剩下的母液经硅胶柱分离纯化(DCM:PE=30%-80%),得到化合物V2-3(43g)。MS(ESI)328[M+H] +
步骤三:
化合物V2-3(41g,125.04mmol)和化合物V1(42.56g,175.05mmol)溶解在二氧六环(400mL)和水(60mL)的混合溶剂中,室温搅拌氩气保护下,依次加入Na 2CO 3(26.51g,250.08mmol)和Pd(dppf) 2Cl(4.57g,6.25mmol),然后升温至100℃搅拌24小时。反应液倒入冰水中,DCM(300mL×2)萃取,无水硫酸钠干燥,减压浓缩干,硅胶柱分离纯化得到黄色固体V2-4(44g,107.69mmol,86.12%收率)。MS(ESI)409.2[M+H] +
步骤四:
化合物V2-4(44g,107.69mmol)加入到400mL的甲醇中,室温搅拌加入NH 4F(47.60g,1.40mol),然后氩气保护下升温至70℃搅拌8小时,反应液边搅拌边加入大量水中,大量固体洗出,过滤得到褐色产物V2-5(27g,107.04mmol,99.40%收率)。MS(ESI)253.1[M+H] +
实施例1
式X化合物的制备
Figure PCTCN2022118654-appb-000004
步骤一:
化合物1H-吡唑-3-羧酸乙酯(25g,178.39mmol)和碳酸铯(58.12g,178.39mmol)溶解在DMF(150mL)中,室温搅拌加入碘乙烷(30.61g,196.23mmol),然后升温至80℃搅拌过夜。反应液倒入水中,EA(150mL×2)萃取,无水硫酸钠干燥,减压浓缩干,硅胶柱分离纯化(EA:PE=0~70%)得到白色固体1-乙基吡唑-3-羧酸乙酯(11.5g,68.37mmol,38.33%收率)。MS(ESI)169[M+H] +
步骤二:
化合物1-乙基吡唑-3-羧酸乙酯(11.5g,68.37mmol)溶解在THF(150mL)中,氩气保护下降温至-10℃,分批加入LiAlH 4(3.89g,102.56mmol),然后0℃左右搅拌1小时。将5.0g的十水合硫酸钠0℃条件下分批加入反应液中,0℃搅拌1小时,慢慢升温至室温搅拌1小时。过滤,滤液减压浓缩干得到无色油状物(1-乙基吡唑-3-基)甲醇(7.8g,61.83mmol,90.43%收率)。MS(ESI)127[M+H] +
步骤三:
将化合物(1-乙基吡唑-3-基)甲醇(7.8g,61.83mmol)溶解在THF(60mL)中,氩气保护冰浴条件下搅拌加入DPPA(18.04g,74.19mmol),然后缓慢滴加DBU(14.12g,92.74mmol,13.84mL),控温15℃以下,滴加完毕升温至45℃搅拌过夜。反应液搅拌加入水200ml,用甲基叔丁基醚(100mL×2)萃取,合并有机相,用水(200ml)洗涤有机相,直接用于下一步反应。
步骤四:
向步骤三的有机相中加入化合物V2(15.52g,61.52mmol)的THF(100mL)和甲基叔丁基醚(200mL)溶液,氩气保护下室温搅拌,依次加入五水硫酸铜(153.60mg,615.21μmol)溶解在水(20mL)中,抗坏血酸钠(487.51mg,2.46mmol)溶解在水(20mL),然后升温至60℃搅拌16小时。反应液倒入水中,用二氯甲烷(1000ml×2)萃取,无水硫酸钠干燥,减压浓缩干,经过硅胶柱分离纯化(PE:DCM(含2%的三乙胺)=30%-100%),得到化合物X(19.00g)。
1H NMR(400MHz,DMSO-d 6)δ8.64(s,1H),7.91(d,J=7.0Hz,1H),7.82(d,J=7.8Hz,1H),7.67(d,J=2.2Hz,1H),7.51(s,2H),7.44(t,J=7.8Hz,1H),6.23(d,J=2.2Hz,1H),5.60(s,2H),4.06(q,J=7.3Hz,2H),2.61(s,3H),1.30(t,J=7.4Hz,3H)。
将所得固体式X化合物送XRPD检测,其粉末X射线衍射图无特征峰,为无定形形式。
测试例1:式X化合物对A 2A受体和A 2B受体的抑制活性
CHO-K1/ADORA 2A/Gα15(GenScript,M00246)和CHO-K1/ADORA 2B/Gα15(GenScript,M00329)细胞培养在Ham's F-12(Gibco,31765092)培养基内。培养基条件为含10%FBS、200μg/mL Zeocin及100μg/mL Hygromycin B或10%FBS、400μg/mL G418及100μg/mL Hygromycin B,具体培养条件详见对应说明书。筛选步骤如下:
(1)用不含血清的培养基将细胞密度调整为6×10 5个/ml。
(2)384孔板(Greiner Bio-One,784075)中每孔分别依次加入5μL细胞液、2.5μL的NECA(Sigma,119140-10MG)和2.5μL的化合物溶液,NECA终浓度为50nM(CHO-K1/ADORA2A)或10nM(CHO-K1/ADORA2B),化合物终浓度为3μM起始向下3倍稀释。
(3)置于37℃培养箱静置培养30mins。
(4)依次加入5μL的cAMP-d 2和5μL的cAMP-ab(Cisbio,62AM4PEB)。
(5)384孔板避光室温放置1小时。
(6)读板(Victor X5,PerkinElmer),XLfit非线性回归分析数据,算出化合物的IC 50。结果如下表14所示。
表14.式X化合物对A 2A受体和A 2B受体的抑制活性
化合物编号 A 2A受体(IC 50/μM) A 2B受体(IC 50/μM)
X <0.001 0.001
由表14可知,本申请实施例式X化合物对A 2A受体和A 2B受体均具有较高的抑制活性。
测试例2:体内药代试验
应用LC/MS/MS法测定了小鼠静脉给药和灌胃给药本申请式X化合物后不同时间点药物浓度,研究本申请式X化合物在小鼠体内的药代动力学行为,评价其药代动力学特征。
实验方案:
试验动物:健康成年雄性ICR小鼠(体重25g~40g,12只,静脉注射组小鼠自由饮水和饮食,灌胃给药组禁食过夜,给药4h后自由饮水和饮食),由北京维通利华实验动物技术有限公司提供;
给药方式与剂量:给药前挑选符合实验要求的动物,称重标记。ICR小鼠尾静脉给药(2mg/kg,5%DMSO,pH4.5 20%Captisol)和灌胃给药(10mg/kg,5%DMSO,pH4.5 20%Captisol)。
血样采集:采静脉及灌胃给药均分别于给药后的0.083小时、0.25小时、0.5小时、1小时、2小时、4小时、6小时、7.5小时和24小时经颈静脉采血约100μL。血液转移至预先加入EDTA2K的1.5mLEP管中,离心4min(8000rpm,4℃),分离出血浆,整个过程在采血后15min内完成。所有血浆样品均需要存放于-20℃冰箱直至样品分析。
本申请式X化合物在静脉给药方式下,小鼠体内的药代动力学性质参数如表15所示。
表15.静脉给药方式下药代动力学参数
化合物编号 清除率CLz(mL/min/kg) 曲线下面积AUC 0-t(hr·ng/mL)
式X化合物 17.2 1918
本申请化合物X在灌胃给药方式下,小鼠体内的药代动力学性质参数如表16所示。
表16.胃给药方式下药代动力学参数
化合物编号 C max(ng/mL) 曲线下面积AUClast(hr·ng/mL)
式X化合物 7060 8569
测试例3:本申请式X化合物的体内药效实验
实验方案:本测试例考察小鼠黑色素瘤细胞株B16F10-OVA-hPD-L1皮下移植瘤小鼠,通过口服给药途径施用本申请式X化合物后,测试其对黑色素瘤B16F10-OVA-hPD-L1荷瘤鼠的体内药效。
实验材料:C57BL/6小鼠(雌性);小鼠黑色素瘤B16F10-OVA-hPD-L1细胞(上海交通大学细胞库),体外单层培养,培养条件为采用含10%胎牛血清DMEM培养基,在37℃下5%CO 2培养箱中培养。用胰酶-EDTA进行常规消化处理传代。当细胞处于指数生长期,饱和度为80%~90%时,收取细胞,计数。
化合物配制:量取式X化合物,加入到溶媒(40%磺丁基-β-环糊精(captisol)的乙酸缓冲液,pH 4.0)中,制成目标浓度的样品。实验操作:将细胞重悬于磷酸盐缓冲液,密度为5×10 6个细胞/mL。将0.1mL的含5×10 5个B16F10-OVA-hPD-L1细胞的PBS溶液皮下接种于每只小鼠的右后背,接种当天按照小鼠体重进行随机分组,每组10只,开始给药,每天2次给药,持续19天。整个实验期间,每天称重并监测动物的健康。每周两次用游标卡尺测量肿瘤直径。
肿瘤体积V的计算公式为:V=0.5×a×b 2,a和b分别表示肿瘤的长径和短径。
化合物的抑瘤疗效用相对肿瘤增殖率T/C(%)评价。相对肿瘤增殖率T/C(%)=Vt/Vc×100%(Vt:治疗组平均瘤体积;Vc:阴性对照组平均瘤体积)。Vt与Vc取同一天数据。给药结束时的结果如表17所示。
表17.测试例3得到的相对肿瘤增殖率
Figure PCTCN2022118654-appb-000005
实施例2游离碱晶型I的制备
方法一
称取约20mg按照实施例1制备的式X化合物(无定形)于玻璃小瓶中,加入1mL的乙醇在50℃左右条件下加热至溶清,取出玻璃小瓶放在室温下降温,降到室温后再放置冰箱中降温至0~4℃,待固体析出,离心后弃去上清液,将固体放置烘箱中烘干,进行XPRD检测。所得固体产物的XPRD图如图1所示,在本申请中定义为游离碱晶型I。游离碱晶型I的DSC图谱如图2所示,其中吸热峰为179.10℃,在104.32℃处有一吸热峰,TGA图谱如图3所示,150℃之前失重约8.8%;说明游离碱晶型I中含有溶剂乙醇。
方法二
称取约20mg按照实施例1制备的式X化合物(无定形)于玻璃小瓶中,加入1mL的甲醇在50℃左右条件下加热至溶清,取出玻璃小瓶放在室温下缓慢降温,降到室温后再放置冰箱中降温至0~4℃,待固体析出,离心后弃去上清液,将固体放置烘箱中烘干,进行XPRD检测,证实得到游离碱晶型I。
方法三
称取约19g按照实施例1制备的式X化合物(无定形)于玻璃小瓶中,加入100mL乙醇,加热至回流(75℃左右)。降至室温,有固体析出,过滤得到固体产物。得到的固体进行XPRD检测,证实得到游离碱晶型I。
实施例3游离碱晶型Ⅳ的制备
方法一
称取约20mg式X化合物(游离碱晶型I)于玻璃小瓶中,加入1mL异丙醇,将玻璃小瓶在室温下混悬振摇24小时后,离心后弃去上清液,将固体放置烘箱中烘干,进行XPRD检测。所得固体产物的XPRD图如图4所示,在本申请中定义为游离碱晶型Ⅳ。游离碱晶型Ⅳ的DSC图谱如图5所示,其在170.43℃处有吸热峰。
方法二
称取约20mg式X化合物(游离碱晶型I)于玻璃小瓶中,加入1mL异丙醇或水,将玻璃小瓶在室 温下混悬振摇7天后,离心后弃去上清液,将固体放置烘箱中烘干,进行XPRD检测,证实得到游离碱晶型Ⅳ。
实施例4游离碱晶型Ⅴ的制备
方法一
称取约20mg式X化合物(无定形)于玻璃小瓶中,加入1mL的丙酮使化合物溶清,将溶液放置室温下进行缓慢挥发,取挥发得到的固体粉末进行XPRD检测,所得固体产物的XPRD图如图6所示,在本申请中定义为游离碱晶型V。游离碱晶型V的DSC图谱如图7所示,游离碱晶型Ⅴ在179.02℃处有一吸热峰。
方法二
称取约20mg式X化合物(游离碱晶型I)于玻璃小瓶中,加入1mL甲基叔丁基醚,将玻璃小瓶在室温下混悬振摇7天后,离心后弃去上清液,将固体放置烘箱中烘干,进行XPRD检测,证实得到游离碱晶型V。
实施例5游离碱晶型Ⅷ的制备
分别称取10mg游离碱晶型I、游离碱晶型Ⅳ、游离碱晶型V充分混合,取其中20mg于玻璃小瓶中,加入1mL丙酮或50%乙醇,在室温下混悬振摇3天后,离心取沉淀,干燥后测XPRD。所得固体产物的XPRD图如图8所示,在本申请中定义为游离碱晶型Ⅷ。游离碱晶型Ⅷ的DSC图谱如图9所示,游离碱晶型Ⅷ在162.93℃处有吸热峰。
实施例6盐酸盐晶型I的制备
称取20mg式X化合物(无定形),按照酸(氢离子)和游离碱摩尔比1.2:1的比例加入1M盐酸溶液,后加入2mL乙酸乙酯或丙酮,加热超声使澄清,50℃保温反应4小时,后缓慢降温使析出固体,离心收集固体。所得固体挥干溶剂后用于XRPD测试,其X射线粉末衍射图如图10所示,在本申请中定义为盐酸盐晶型I。
实施例7硫酸盐晶型I的制备
称取20mg式X化合物(无定形),按照酸(氢离子)和游离碱摩尔比1.2:1的比例加入0.5M硫酸溶液,后加入2mL乙酸乙酯或丙酮,加热超声使澄清,50℃保温反应4小时,后缓慢降温使析出固体,离心收集固体。所得固体挥干溶剂后用于XRPD测试,其X射线粉末衍射图如图11所示,在本申请中定义为硫酸盐晶型I。
实施例8柠檬酸盐晶型I的制备
称取20mg式X化合物(无定形),按照酸(氢离子)和游离碱摩尔比1.2:1的比例加入0.5mol/L柠檬酸溶液,后加入2mL丙酮,加热超声使澄清,50℃保温反应4小时,后缓慢降至室温,加入反溶剂正庚烷析出固体,离心收集固体。所得固体挥干溶剂后用于XRPD测试,其X射线粉末衍射图如图12所示,在本申请中定义为柠檬酸盐晶型I。
实施例9富马酸盐晶型I的制备
称取20mg式X化合物(无定形),按照酸(氢离子)和游离碱摩尔比1.2:1的比例加入0.25mol/L富马酸溶液,后加入2mL乙酸乙酯,加热超声使澄清,50℃保温反应4小时,后缓慢降至室温,加入反溶剂正庚烷析出固体,离心收集固体。所得固体挥干溶剂后用于XRPD测试,其X射线粉末衍射图如图13所示,在本申请中定义为富马酸盐晶型I。
实施例10琥珀酸盐晶型I的制备
称取20mg式X化合物(无定形),按照酸(氢离子)和游离碱摩尔比1.2:1的比例加入0.5mol/L琥珀酸溶液,后加入2mL乙酸乙酯,加热超声使澄清,50℃保温反应4小时,后缓慢降至室温,加入反溶剂正庚烷析出固体,离心收集固体。所得固体挥干溶剂后用于XRPD测试,其X射线粉末衍射图如图14所示,在本申请中定义为琥珀酸盐晶型I。
实施例11琥珀酸盐晶型Ⅱ的制备
称取20mg式X化合物(无定形),按照酸(氢离子)和游离碱摩尔比1.2:1的比例加入0.5mol/L琥珀酸溶液,后加入2mL丙酮,加热超声使澄清,50℃保温反应4小时,后缓慢降至室温,加入反溶剂正 庚烷析出固体,离心收集固体。所得固体挥干溶剂后用于XRPD测试,其X射线粉末衍射图如图15所示,在本申请中定义为琥珀酸盐晶型Ⅱ。
以上所述实施例的各技术特征可以进行任意的组合,为使描述简洁,未对上述实施例中的各个技术特征所有可能的组合都进行描述,然而,只要这些技术特征的组合不存在矛盾,都应当认为是本说明书记载的范围。
以上所述实施例仅表达了本申请的几种实施方式,便于具体和详细地理解本申请的技术方案,但并不能因此而理解为对发明专利保护范围的限制。应当指出的是,对于本领域的普通技术人员来说,在不脱离本申请构思的前提下,还可以做出若干变形和改进,这些都属于本申请的保护范围。应当理解,本领域技术人员在本申请提供的技术方案的基础上,通过合乎逻辑的分析、推理或者有限的试验得到的技术方案,均在本申请所附权利要求的保护范围内。因此,本申请专利的保护范围应以所附权利要求的内容为准,说明书及附图可以用于解释权利要求的内容。

Claims (11)

  1. 一种式X化合物或其药学上可接受的盐的多晶型物,
    Figure PCTCN2022118654-appb-100001
    所述药学上可接受的盐选自:盐酸盐、硫酸盐、柠檬酸盐、富马酸盐或琥珀酸盐。
  2. 根据权利要求1所述的多晶型物,其中所述式X化合物的多晶型物是选自以下的任一组:
    式X化合物的游离碱晶型I,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:
    7.57±0.2、13.41±0.2、14.64±0.2、19.81±0.2和24.43±0.2;
    式X化合物的游离碱晶型IV,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:
    11.88±0.2、14.17±0.2、16.96±0.2、22.63±0.2、23.56±0.2和25.66±0.2;
    式X化合物的游离碱晶型Ⅴ,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:
    11.88±0.2、16.36±0.2、16.99±0.2、22.99±0.2、23.47±0.2和26.41±0.2;和
    式X化合物的游离碱晶型VIII,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:
    6.40±0.2、13.12±0.2、15.91±0.2、22.69±0.2和26.95±0.2。
  3. 根据权利要求2所述的多晶型物,其中所述式X化合物的多晶型物是选自以下的任一组:
    式X化合物的游离碱晶型I,其X射线粉末衍射图谱还包括1个或2个选自下列2θ(°)角处的特征衍射峰:24.87±0.2和27.46±0.2;
    式X化合物的游离碱晶型IV,其X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:15.76±0.2和26.08±0.2;
    式X化合物的游离碱晶型Ⅴ,其X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:7.90±0.2、13.71±0.2、14.14±0.2、15.31±0.2、17.80±0.2、18.76±0.2、21.34±0.2、24.82±0.2、25.51±0.2、27.88±0.2和30.46±0.2;和
    式X化合物的游离碱晶型VIII,其X射线粉末衍射图谱还包括至少一个选自下列2θ(°)角处的特征衍射峰:11.62±0.2、12.36±0.2、16.62±0.2、18.34±0.2、18.88±0.2、24.94±0.2和29.65±0.2。
  4. 根据权利要求2所述的多晶型物,其中
    所述游离碱晶型I的X射线粉末衍射图谱基本如图1所示;
    所述游离碱晶型IV的X射线粉末衍射图谱基本如图4所示;
    所述游离碱晶型Ⅴ的X射线粉末衍射图谱基本如图6所示;或
    所述游离碱晶型VIII的X射线粉末衍射图谱基本如图8所示。
  5. 根据权利要求2所述的多晶型物,其中
    所述游离碱晶型I的差示扫描量热曲线在179.10±3℃处具有吸热峰;
    所述游离碱晶型IV的差示扫描量热曲线在170.43±3℃处具有吸热峰;
    所述游离碱晶型Ⅴ的差示扫描量热曲线在179.02±3℃处具有吸热峰;或
    所述游离碱晶型VIII的差示扫描量热曲线在162.93±3℃处具有吸热峰。
  6. 根据权利要求1所述的多晶型物,其中所述式X化合物药学上可接受的盐的多晶型物是选自以下的任一组:
    式X化合物的盐酸盐晶型I,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:
    11.95±0.2、24.37±0.2、25.21±0.2和26.35±0.2;
    式X化合物的硫酸盐晶型I,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:
    8.22±0.2、11.29±0.2、14.14±0.2、16.96±0.2、18.13±0.2、22.27±0.2、23.86±0.2和27.79±0.2;
    式X化合物的柠檬酸盐晶型I,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:
    12.55±0.2、14.50±0.2、21.55±0.2和22.75±0.2;
    式X化合物的富马酸盐晶型I,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:
    7.12±0.2、11.71±0.2、16.90±0.2、23.26±0.2、23.65±0.2、24.64±0.2和25.54±0.2;
    式X化合物的琥珀酸盐晶型I,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:
    4.87±0.2、9.31±0.2、13.78±0.2、14.14±0.2、18.16±0.2、20.71±0.2、22.12±0.2和26.83±0.2;和
    式X化合物的琥珀酸盐晶型Ⅱ,其X射线粉末衍射图谱在下列2θ(°)角处具有特征衍射峰:
    11.86±0.2、12.49±0.2、14.50±0.2、15.22±0.2、16.90±0.2、21.64±0.2、22.87±0.2和25.75±0.2。
  7. 根据权利要求6所述的多晶型物,其中
    所述盐酸盐晶型I的射线粉末衍射图谱基本如图10所示:
    所述硫酸盐晶型I的X射线粉末衍射图谱基本如图11所示;
    所述柠檬酸盐晶型I的X射线粉末衍射图谱基本如图12所示;
    所述富马酸盐晶型I的X射线粉末衍射图谱基本如图13所示;
    所述琥珀酸盐晶型I的X射线粉末衍射图谱基本如图14所示;或
    所述琥珀酸盐晶型Ⅱ的X射线粉末衍射图谱基本如图15所示。
  8. 一种药物组合物,包括:
    (a)权利要求1至7任一项所述的多晶型物;以及
    (b)药学上可接受的载体。
  9. 权利要求1-7任一项所述的多晶型物,或权利要求8所述的药物组合物在制备A 2A受体抑制剂或A 2B受体抑制剂中的应用。
  10. 权利要求1-7任一项所述的多晶型物、或权利要求8所述的药物组合物在制备预防或治疗由腺苷A 2A受体或腺苷A 2B受体介导的癌症或免疫相关疾病的药物中的用途。
  11. 一种用于预防或治疗由腺苷A 2A受体或腺苷A 2B受体介导的癌症或免疫相关疾病的方法,包括向有此需要的患者施用治疗有效量的根据权利要求1至7中任一项所述的多晶型物、或根据权利要求8所述的药物组合物。
PCT/CN2022/118654 2021-09-15 2022-09-14 氮杂芳环类化合物及其药学上可接受的盐的多晶型物、药物组合物和应用 WO2023040876A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280060100.6A CN117915919A (zh) 2021-09-15 2022-09-14 氮杂芳环类化合物及其药学上可接受的盐的多晶型物、药物组合物和应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111080394.2 2021-09-15
CN202111080394 2021-09-15

Publications (1)

Publication Number Publication Date
WO2023040876A1 true WO2023040876A1 (zh) 2023-03-23

Family

ID=85602437

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/118654 WO2023040876A1 (zh) 2021-09-15 2022-09-14 氮杂芳环类化合物及其药学上可接受的盐的多晶型物、药物组合物和应用

Country Status (2)

Country Link
CN (1) CN117915919A (zh)
WO (1) WO2023040876A1 (zh)

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017016463A1 (zh) * 2015-07-24 2017-02-02 上海海雁医药科技有限公司 Egfr抑制剂及其药学上可接受的盐和多晶型物及其应用
WO2017206904A1 (zh) * 2016-06-02 2017-12-07 上海海雁医药科技有限公司 Pi3k抑制剂及其药学上可接受的盐和多晶型物及其应用
CN110214012A (zh) * 2017-01-20 2019-09-06 艾库斯生物科学有限公司 用于治疗癌症相关疾病的唑嘧啶
WO2019206154A1 (zh) * 2018-04-24 2019-10-31 上海海雁医药科技有限公司 Cdk4/6抑制剂及其药学上可接受的盐和多晶型物及其应用
WO2019206155A1 (zh) * 2018-04-24 2019-10-31 上海海雁医药科技有限公司 Ezh2抑制剂及其药学上可接受的盐和多晶型物及其应用
WO2020187267A1 (zh) * 2019-03-18 2020-09-24 上海海雁医药科技有限公司 Btk抑制剂及其药学上可接受的盐和多晶型物及其应用
WO2021185256A1 (zh) * 2020-03-16 2021-09-23 上海海雁医药科技有限公司 取代的嘧啶或吡啶胺衍生物、其组合物及医药上的用途
CN113429410A (zh) * 2020-03-23 2021-09-24 上海海雁医药科技有限公司 多杂环取代的嘧啶或吡啶胺衍生物、其组合物及医药上的用途

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017016463A1 (zh) * 2015-07-24 2017-02-02 上海海雁医药科技有限公司 Egfr抑制剂及其药学上可接受的盐和多晶型物及其应用
WO2017206904A1 (zh) * 2016-06-02 2017-12-07 上海海雁医药科技有限公司 Pi3k抑制剂及其药学上可接受的盐和多晶型物及其应用
CN110214012A (zh) * 2017-01-20 2019-09-06 艾库斯生物科学有限公司 用于治疗癌症相关疾病的唑嘧啶
WO2019206154A1 (zh) * 2018-04-24 2019-10-31 上海海雁医药科技有限公司 Cdk4/6抑制剂及其药学上可接受的盐和多晶型物及其应用
WO2019206155A1 (zh) * 2018-04-24 2019-10-31 上海海雁医药科技有限公司 Ezh2抑制剂及其药学上可接受的盐和多晶型物及其应用
WO2020187267A1 (zh) * 2019-03-18 2020-09-24 上海海雁医药科技有限公司 Btk抑制剂及其药学上可接受的盐和多晶型物及其应用
WO2021185256A1 (zh) * 2020-03-16 2021-09-23 上海海雁医药科技有限公司 取代的嘧啶或吡啶胺衍生物、其组合物及医药上的用途
CN113429410A (zh) * 2020-03-23 2021-09-24 上海海雁医药科技有限公司 多杂环取代的嘧啶或吡啶胺衍生物、其组合物及医药上的用途

Also Published As

Publication number Publication date
CN117915919A (zh) 2024-04-19

Similar Documents

Publication Publication Date Title
IL212398A (en) 6-oxo-1,6-dihydropyridines and their use in drug preparation
UA79229C2 (en) 3-nitrogen-6,7-dioxygen steroids and uses related thereto
TW201236684A (en) Pharmaceutically acceptable salts of (E)-N-[4-[[3-chloro-4-(2-pyridylmethoxy)phenyl]amino]-3-cyano-7-ethoxy-6-quinolyl]-3-[(2R)-1-methylpyrrolidin-2-yl]prop-2-enamide, preparation process and pharmaceutical use there of
JP7406691B2 (ja) トリアゾロピリミジン系化合物の結晶形、塩のタイプおよびその調製方法
JP2018511634A (ja) B−rafキナーゼのマレイン酸塩、結晶形、調整方法、及びその使用
JP2020527166A (ja) Bcl−2タンパク質を阻害するためのN−ベンゼンスルホニルベンズアミド系化合物、その組成物および使用
WO2020228807A1 (zh) 一类嘧啶化合物的盐、多晶型物及其药物组合物、制备方法和应用
JPWO2004035572A1 (ja) N−{2−クロロ−4−[(6,7−ジメトキシ−4−キノリル)オキシ]フェニル}−n’−(5−メチル−3−イソキサゾリル)ウレアの塩の結晶形
JP2015528482A (ja) 結晶性化合物
US20210079021A1 (en) Crystal form and salt form of tgf-bri inhibitor and preparation method therefor
JP2022553833A (ja) エストロゲン受容体調節薬の塩及び形態
WO2023040876A1 (zh) 氮杂芳环类化合物及其药学上可接受的盐的多晶型物、药物组合物和应用
JP2022518605A (ja) ジアリールチオヒダントイン化合物結晶
KR102374933B1 (ko) c-Met 억제제의 결정형과 이의 염 형태 및 제조 방법
WO2023040863A1 (zh) 嘧啶衍生物及其药学上可接受的盐的多晶型物和应用
WO2020147838A1 (zh) 一种egfr抑制剂的盐、晶型及其制备方法
JP2019524857A (ja) キナゾリン誘導体の塩の結晶
JP2022528747A (ja) Fak阻害剤の塩及び結晶形態
WO2020140894A1 (zh) 一类含氟取代的苯并噻吩类化合物及其药物组合物及应用
WO2020192762A1 (zh) 一种a2a受体拮抗剂的盐型、晶型及其制备方法
TW201829398A (zh) 酪胺酸蛋白激酶調節劑、晶型及其用途
CN110903291B (zh) 一种杂芳基并[4,3-c]嘧啶-5-胺类衍生物的盐、盐的晶型及制备方法
CN112334473B (zh) 一种杂芳基并[4,3-c]嘧啶-5-胺类衍生物的晶型及制备方法
WO2022262699A1 (zh) 取代的苯并咪唑类化合物及包含该化合物的组合物及其用途
JP2023543055A (ja) テガビビントの結晶形態、調製方法、及びその使用

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: 22869245

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202280060100.6

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE